@article{WeberLassalleHaukeRamseretal.2018, author = {Weber-Lassalle, Nana and Hauke, Jan and Ramser, Juliane and Richters, Lisa and Groß, Eva and Bl{\"u}mcke, Britta and Gehrig, Andrea and Kahlert, Anne-Karin and M{\"u}ller, Clemens R. and Hackmann, Karl and Honisch, Ellen and Weber-Lassalle, Konstantin and Niederacher, Dieter and Borde, Julika and Thiele, Holger and Ernst, Corinna and Altm{\"u}ller, Janine and Neidhardt, Guido and N{\"u}rnberg, Peter and Klaschik, Kristina and Schroeder, Christopher and Platzer, Konrad and Volk, Alexander E. and Wang-Gohrke, Shan and Just, Walter and Auber, Bernd and Kubisch, Christian and Schmidt, Gunnar and Horvath, Judit and Wappenschmidt, Barbara and Engel, Christoph and Arnold, Norbert and Dworniczak, Bernd and Rhiem, Kerstin and Meindl, Alfons and Schmutzler, Rita K. and Hahnen, Eric}, title = {BRIP1 loss-of-function mutations confer high risk for familial ovarian cancer, but not familial breast cancer}, series = {Breast Cancer Research}, volume = {20}, journal = {Breast Cancer Research}, doi = {10.1186/s13058-018-0935-9}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-233433}, year = {2018}, abstract = {Background Germline mutations in the BRIP1 gene have been described as conferring a moderate risk for ovarian cancer (OC), while the role of BRIP1 in breast cancer (BC) pathogenesis remains controversial. Methods To assess the role of deleterious BRIP1 germline mutations in BC/OC predisposition, 6341 well-characterized index patients with BC, 706 index patients with OC, and 2189 geographically matched female controls were screened for loss-of-function (LoF) mutations and potentially damaging missense variants. All index patients met the inclusion criteria of the German Consortium for Hereditary Breast and Ovarian Cancer for germline testing and tested negative for pathogenic BRCA1/2 variants. Results BRIP1 LoF mutations confer a high OC risk in familial index patients (odds ratio (OR) = 20.97, 95\% confidence interval (CI) = 12.02-36.57, P < 0.0001) and in the subgroup of index patients with late-onset OC (OR = 29.91, 95\% CI = 14.99-59.66, P < 0.0001). No significant association of BRIP1 LoF mutations with familial BC was observed (OR = 1.81 95\% CI = 1.00-3.30, P = 0.0623). In the subgroup of familial BC index patients without a family history of OC there was also no apparent association (OR = 1.42, 95\% CI = 0.70-2.90, P = 0.3030). In 1027 familial BC index patients with a family history of OC, the BRIP1 mutation prevalence was significantly higher than that observed in controls (OR = 3.59, 95\% CI = 1.43-9.01; P = 0.0168). Based on the negative association between BRIP1 LoF mutations and familial BC in the absence of an OC family history, we conclude that the elevated mutation prevalence in the latter cohort was driven by the occurrence of OC in these families. Compared with controls, predicted damaging rare missense variants were significantly more prevalent in OC (P = 0.0014) but not in BC (P = 0.0693) patients. Conclusions To avoid ambiguous results, studies aimed at assessing the impact of candidate predisposition gene mutations on BC risk might differentiate between BC index patients with an OC family history and those without. In familial cases, we suggest that BRIP1 is a high-risk gene for late-onset OC but not a BC predisposition gene, though minor effects cannot be excluded.}, language = {en} } @article{VigoritoKuchenbaeckerBeesleyetal.2016, author = {Vigorito, Elena and Kuchenbaecker, Karoline B. and Beesley, Jonathan and Adlard, Julian and Agnarsson, Bjarni A. and Andrulis, Irene L. and Arun, Banu K. and Barjhoux, Laure and Belotti, Muriel and Benitez, Javier and Berger, Andreas and Bojesen, Anders and Bonanni, Bernardo and Brewer, Carole and Caldes, Trinidad and Caligo, Maria A. and Campbell, Ian and Chan, Salina B. and Claes, Kathleen B. M. and Cohn, David E. and Cook, Jackie and Daly, Mary B. and Damiola, Francesca and Davidson, Rosemarie and de Pauw, Antoine and Delnatte, Capucine and Diez, Orland and Domchek, Susan M. and Dumont, Martine and Durda, Katarzyna and Dworniczak, Bernd and Easton, Douglas F. and Eccles, Diana and Ardnor, Christina Edwinsdotter and Eeles, Ros and Ejlertsen, Bent and Ellis, Steve and Evans, D. Gareth and Feliubadalo, Lidia and Fostira, Florentia and Foulkes, William D. and Friedman, Eitan and Frost, Debra and Gaddam, Pragna and Ganz, Patricia A. and Garber, Judy and Garcia-Barberan, Vanesa and Gauthier-Villars, Marion and Gehrig, Andrea and Gerdes, Anne-Marie and Giraud, Sophie and Godwin, Andrew K. and Goldgar, David E. and Hake, Christopher R. and Hansen, Thomas V. O. and Healey, Sue and Hodgson, Shirley and Hogervorst, Frans B. L. and Houdayer, Claude and Hulick, Peter J. and Imyanitov, Evgeny N. and Isaacs, Claudine and Izatt, Louise and Izquierdo, Angel and Jacobs, Lauren and Jakubowska, Anna and Janavicius, Ramunas and Jaworska-Bieniek, Katarzyna and Jensen, Uffe Birk and John, Esther M. and Vijai, Joseph and Karlan, Beth Y. and Kast, Karin and Khan, Sofia and Kwong, Ava and Laitman, Yael and Lester, Jenny and Lesueur, Fabienne and Liljegren, Annelie and Lubinski, Jan and Mai, Phuong L. and Manoukian, Siranoush and Mazoyer, Sylvie and Meindl, Alfons and Mensenkamp, Arjen R. and Montagna, Marco and Nathanson, Katherine L. and Neuhausen, Susan L. and Nevanlinna, Heli and Niederacher, Dieter and Olah, Edith and Olopade, Olufunmilayo I. and Ong, Kai-ren and Osorio, Ana and Park, Sue Kyung and Paulsson-Karlsson, Ylva and Pedersen, Inge Sokilde and Peissel, Bernard and Peterlongo, Paolo and Pfeiler, Georg and Phelan, Catherine M. and Piedmonte, Marion and Poppe, Bruce and Pujana, Miquel Angel and Radice, Paolo and Rennert, Gad and Rodriguez, Gustavo C. and Rookus, Matti A. and Ross, Eric A. and Schmutzler, Rita Katharina and Simard, Jacques and Singer, Christian F. and Slavin, Thomas P. and Soucy, Penny and Southey, Melissa and Steinemann, Doris and Stoppa-Lyonnet, Dominique and Sukiennicki, Grzegorz and Sutter, Christian and Szabo, Csilla I. and Tea, Muy-Kheng and Teixeira, Manuel R. and Teo, Soo-Hwang and Terry, Mary Beth and Thomassen, Mads and Tibiletti, Maria Grazia and Tihomirova, Laima and Tognazzo, Silvia and van Rensburg, Elizabeth J. and Varesco, Liliana and Varon-Mateeva, Raymonda and Vratimos, Athanassios and Weitzel, Jeffrey N. and McGuffog, Lesley and Kirk, Judy and Toland, Amanda Ewart and Hamann, Ute and Lindor, Noralane and Ramus, Susan J. and Greene, Mark H. and Couch, Fergus J. and Offit, Kenneth and Pharoah, Paul D. P. and Chenevix-Trench, Georgia and Antoniou, Antonis C.}, title = {Fine-Scale Mapping at 9p22.2 Identifies Candidate Causal Variants That Modify Ovarian Cancer Risk in BRCA1 and BRCA2 Mutation Carriers}, series = {PLoS ONE}, volume = {11}, journal = {PLoS ONE}, number = {7}, doi = {10.1371/journal.pone.0158801}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-166869}, pages = {e0158801}, year = {2016}, abstract = {Population-based genome wide association studies have identified a locus at 9p22.2 associated with ovarian cancer risk, which also modifies ovarian cancer risk in BRCA1 and BRCA2 mutation carriers. We conducted fine-scale mapping at 9p22.2 to identify potential causal variants in BRCA1 and BRCA2 mutation carriers. Genotype data were available for 15,252 (2,462 ovarian cancer cases) BRCA1 and 8,211 (631 ovarian cancer cases) BRCA2 mutation carriers. Following genotype imputation, ovarian cancer associations were assessed for 4,873 and 5,020 SNPs in BRCA1 and BRCA 2 mutation carriers respectively, within a retrospective cohort analytical framework. In BRCA1 mutation carriers one set of eight correlated candidate causal variants for ovarian cancer risk modification was identified (top SNP rs10124837, HR: 0.73, 95\%CI: 0.68 to 0.79, p-value 2× 10-16). These variants were located up to 20 kb upstream of BNC2. In BRCA2 mutation carriers one region, up to 45 kb upstream of BNC2, and containing 100 correlated SNPs was identified as candidate causal (top SNP rs62543585, HR: 0.69, 95\%CI: 0.59 to 0.80, p-value 1.0 × 10-6). The candidate causal in BRCA1 mutation carriers did not include the strongest associated variant at this locus in the general population. In sum, we identified a set of candidate causal variants in a region that encompasses the BNC2 transcription start site. The ovarian cancer association at 9p22.2 may be mediated by different variants in BRCA1 mutation carriers and in the general population. Thus, potentially different mechanisms may underlie ovarian cancer risk for mutation carriers and the general population.}, language = {en} } @article{VergoteMacarullaHirschetal.2023, author = {Vergote, Ignace and Macarulla, Teresa and Hirsch, Fred R. and Hagemann, Carsten and Miller, David Scott}, title = {Tumor Treating Fields (TTFields) therapy concomitant with taxanes for cancer treatment}, series = {Cancers}, volume = {15}, journal = {Cancers}, number = {3}, issn = {2072-6694}, doi = {10.3390/cancers15030636}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-305007}, year = {2023}, abstract = {Non-small cell lung cancer, ovarian cancer, and pancreatic cancer all present with high morbidity and mortality. Systemic chemotherapies have historically been the cornerstone of standard of care (SOC) regimens for many cancers, but are associated with systemic toxicity. Multimodal treatment combinations can help improve patient outcomes; however, implementation is limited by additive toxicities and potential drug-drug interactions. As such, there is a high unmet need to develop additional therapies to enhance the efficacy of SOC treatments without increasing toxicity. Tumor Treating Fields (TTFields) are electric fields that exert physical forces to disrupt cellular processes critical for cancer cell viability and tumor progression. The therapy is locoregional and is delivered noninvasively to the tumor site via a portable medical device that consists of field generator and arrays that are placed on the patient's skin. As a noninvasive treatment modality, TTFields therapy-related adverse events mainly consist of localized skin reactions, which are manageable with effective acute and prophylactic treatments. TTFields selectively target cancer cells through a multi-mechanistic approach without affecting healthy cells and tissues. Therefore, the application of TTFields therapy concomitant with other cancer treatments may lead to enhanced efficacy, with low risk of further systemic toxicity. In this review, we explore TTFields therapy concomitant with taxanes in both preclinical and clinical settings. The summarized data suggest that TTFields therapy concomitant with taxanes may be beneficial in the treatment of certain cancers.}, language = {en} } @article{OsorioMilneKuchenbaeckeretal.2014, author = {Osorio, Ana and Milne, Roger L. and Kuchenbaecker, Karoline and Vaclov{\´a}, Tereza and Pita, Guillermo and Alonso, Rosario and Peterlongo, Paolo and Blanco, Ignacio and de la Hoya, Miguel and Duran, Mercedes and Diez, Orland and Ram{\´o}n y Cajal, Teresa and Konstantopoulou, Irene and Mart{\´i}nez-Bouzas, Christina and Conejero, Raquel Andr{\´e}s and Soucy, Penny and McGuffog, Lesley and Barrowdale, Daniel and Lee, Andrew and Arver, Brita and Rantala, Johanna and Loman, Niklas and Ehrencrona, Hans and Olopade, Olufunmilayo I. and Beattie, Mary S. and Domchek, Susan M. and Nathanson, Katherine and Rebbeck, Timothy R. and Arun, Banu K. and Karlan, Beth Y. and Walsh, Christine and Lester, Jenny and John, Esther M. and Whittemore, Alice S. and Daly, Mary B. and Southey, Melissa and Hopper, John and Terry, Mary B. and Buys, Saundra S. and Janavicius, Ramunas and Dorfling, Cecilia M. and van Rensburg, Elizabeth J. and Steele, Linda and Neuhausen, Susan L. and Ding, Yuan Chun and Hansen, Thomas V. O. and J{\o}nson, Lars and Ejlertsen, Bent and Gerdes, Anne-Marie and Infante, Mar and Herr{\´a}ez, Bel{\´e}n and Moreno, Leticia Thais and Weitzel, Jeffrey N. and Herzog, Josef and Weeman, Kisa and Manoukian, Siranoush and Peissel, Bernard and Zaffaroni, Daniela and Scuvera, Guilietta and Bonanni, Bernardo and Mariette, Frederique and Volorio, Sara and Viel, Alessandra and Varesco, Liliana and Papi, Laura and Ottini, Laura and Tibiletti, Maria Grazia and Radice, Paolo and Yannoukakos, Drakoulis and Garber, Judy and Ellis, Steve and Frost, Debra and Platte, Radka and Fineberg, Elena and Evans, Gareth and Lalloo, Fiona and Izatt, Louise and Eeles, Ros and Adlard, Julian and Davidson, Rosemarie and Cole, Trevor and Eccles, Diana and Cook, Jackie and Hodgson, Shirley and Brewer, Carole and Tischkowitz, Marc and Douglas, Fiona and Porteous, Mary and Side, Lucy and Walker, Lisa and Morrison, Patrick and Donaldson, Alan and Kennedy, John and Foo, Claire and Godwin, Andrew K. and Schmutzler, Rita Katharina and Wappenschmidt, Barbara and Rhiem, Kerstin and Engel, Christoph and Meindl, Alftons and Ditsch, Nina and Arnold, Norbert and Plendl, Hans J{\"o}rg and Niederacher, Dieter and Sutter, Christian and Wang-Gohrke, Shan and Steinemann, Doris and Preisler-Adams, Sabine and Kast, Karin and Varon-Mateeva, Raymonda and Gehrig, Andrea and Stoppa-Lyonnet, Dominique and Sinilnikova, Olga M. and Mazoyer, Sylvie and Damiola, Francesca and Poppe, Bruce and Claes, Kathleen and Piedmonte, Marion and Tucker, Kathy and Backes, Floor and Rodr{\´i}guez, Gustavo and Brewster, Wendy and Wakeley, Katie and Rutherford, Thomas and Cald{\´e}s, Trinidad and Nevanlinna, Heli and Aittom{\"a}ki, Kristiina and Rookus, Matti A. and van Os, Theo A. M. and van der Kolk, Lizet and de Lange, J. L. and Meijers-Heijboer, Hanne E. J. and van der Hout, A. H. and van Asperen, Christi J. and Gom{\´e}z Garcia, Encarna B. and Encarna, B. and Hoogerbrugge, Nicoline and Coll{\´e}e, J. Margriet and van Deurzen, Carolien H. M. and van der Luijt, Rob B. and Devilee, Peter and Olah, Edith and L{\´a}zaro, Conxi and Teul{\´e}, Alex and Men{\´e}ndez, Mireia and Jakubowska, Anna and Cybulski, Cezary and Gronwald, Jecek and Lubinski, Jan and Durda, Katarzyna and Jaworska-Bieniek, Katarzyna and Johannsson, Oskar Th. and Maugard, Christine and Montagna, Marco and Tognazzo, Silvia and Teixeira, Manuel R. and Healey, Sue and Olswold, Curtis and Guidugli, Lucia and Lindor, Noralane and Slager, Susan and Szabo, Csilla I. and Vijai, Joseph and Robson, Mark and Kauff, Noah and Zhang, Liying and Rau-Murthy, Rohini and Fink-Retter, Anneliese and Singer, Christine F. and Rappaport, Christine and Kaulich, Daphne Geschwantler and Pfeiler, Georg and Tea, Muy-Kheng and Berger, Andreas and Phelan, Catherine M. and Greene, Mark H. and Mai, Phuong L. and Lejbkowicz, Flavio and Andrulis, Irene and Mulligan, Anna Marie and Glendon, Gord and Toland, Amanda Ewart and Bojesen, Anders and Pedersen, Inge Sokilde and Sunde, Lone and Thomassen, Mads and Kruse, Torben A. and Jensen, Uffe Birk and Friedman, Eitan and Laitman, Yeal and Shimon, Shanie Paluch and Simard, Jaques and Easton, Douglas F. and Offit, Kenneth and Couch, Fergus J. and Chenevix-Trench, Georgia and Antoniou, Antonis C. and Benitez, Javier}, title = {DNA Glycosylases Involved in Base Excision Repair May Be Associated with Cancer Risk in BRCA1 and BRCA2 Mutation Carriers}, series = {PLOS Genetics}, volume = {4}, journal = {PLOS Genetics}, number = {e1004256}, issn = {1553-7404}, doi = {10.1371/journal.pgen.1004256}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-116820}, year = {2014}, abstract = {Single Nucleotide Polymorphisms (SNPs) in genes involved in the DNA Base Excision Repair (BER) pathway could be associated with cancer risk in carriers of mutations in the high-penetrance susceptibility genes BRCA1 and BRCA2, given the relation of synthetic lethality that exists between one of the components of the BER pathway, PARP1 (poly ADP ribose polymerase), and both BRCA1 and BRCA2. In the present study, we have performed a comprehensive analysis of 18 genes involved in BER using a tagging SNP approach in a large series of BRCA1 and BRCA2 mutation carriers. 144 SNPs were analyzed in a two stage study involving 23,463 carriers from the CIMBA consortium (the Consortium of Investigators of Modifiers of BRCA1 and BRCA2). Eleven SNPs showed evidence of association with breast and/or ovarian cancer at p<0.05 in the combined analysis. Four of the five genes for which strongest evidence of association was observed were DNA glycosylases. The strongest evidence was for rs1466785 in the NEIL2 (endonuclease VIII-like 2) gene (HR: 1.09, 95\% CI (1.03-1.16), p = 2.7x10(-3)) for association with breast cancer risk in BRCA2 mutation carriers, and rs2304277 in the OGG1 (8-guanine DNA glycosylase) gene, with ovarian cancer risk in BRCA1 mutation carriers (HR: 1.12 95\% CI: 1.03-1.21, p = 4.8x10(-3)). DNA glycosylases involved in the first steps of the BER pathway may be associated with cancer risk in BRCA1/2 mutation carriers and should be more comprehensively studied.}, language = {en} } @article{OliveiraFerrerSchmalfeldtDietletal.2022, author = {Oliveira-Ferrer, Leticia and Schmalfeldt, Barbara and Dietl, Johannes and Bartmann, Catharina and Schumacher, Udo and St{\"u}rken, Christine}, title = {Ovarian cancer-cell pericellular hyaluronan deposition negatively impacts prognosis of ovarian cancer patients}, series = {Biomedicines}, volume = {10}, journal = {Biomedicines}, number = {11}, issn = {2227-9059}, doi = {10.3390/biomedicines10112944}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-297539}, year = {2022}, abstract = {Background: Hyaluronan (HA), a component of the extracellular matrix, is frequently increased under pathological conditions including cancer. Not only stroma cells but also cancer cells themselves synthesize HA, and the interaction of HA with its cognate receptors promotes malignant progression and metastasis. Methods: In the present study, HA deposition in tissue sections was analyzed by hyaluronan-binding protein (HABP) ligand histochemistry in 17 borderline tumors and 102 primary and 20 recurrent ovarian cancer samples. The intensity and, particularly, localization of the HA deposition were recorded: for the localization, the pericellular deposition around the ovarian cancer cells was distinguished from the deposition within the stromal compartment. These histochemical data were correlated with clinical and pathological parameters. Additionally, within a reduced subgroup of ovarian cancer samples (n = 70), the RNA levels of several HA-associated genes were correlated with the HA localization and intensity. Results: Both stroma-localized and pericellular tumor-cell-associated HA deposition were observed. Cancer-cell pericellular HA deposition, irrespective of its staining intensity, was significantly associated with malignancy, and in the primary ovarian cancer cohort, it represents an independent unfavorable prognostic marker for overall survival. Furthermore, a significant association between high CD44, HAS2 and HAS3 mRNA levels and a cancer-cell pericellular HA-deposition pattern was noted. In contrast, stromal hyaluronan deposition had no impact on ovarian cancer prognosis. Conclusions: In conclusion, the site of HA deposition is of prognostic value, but the amount deposited is not. The significant association of only peritumoral cancer-cell HA deposition with high CD44 mRNA expression levels suggests a pivotal role of the CD44-HA signaling axis for malignant progression in ovarian cancer.}, language = {en} } @article{MontalbandelBarrioPenskiSchlahsaetal.2016, author = {Montalb{\´a}n del Barrio, Itsaso and Penski, Cornelia and Schlahsa, Laura and Stein, Roland G. and Diessner, Joachim and W{\"o}ckel, Achim and Dietl, Johannes and Lutz, Manfred B. and Mittelbronn, Michel and Wischhusen, J{\"o}rg and H{\"a}usler, Sebastian F. M.}, title = {Adenosine-generating ovarian cancer cells attract myeloid cells which differentiate into adenosine-generating tumor associated macrophages - a self-amplifying, CD39- and CD73-dependent mechanism for tumor immune escape}, series = {Journal for ImmunoTherapy of Cancer}, volume = {4}, journal = {Journal for ImmunoTherapy of Cancer}, number = {49}, doi = {10.1186/s40425-016-0154-9}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-146624}, year = {2016}, abstract = {Background Ovarian cancer (OvCA) tissues show abundant expression of the ectonucleotidases CD39 and CD73 which generate immunomodulatory adenosine, thereby inhibiting cytotoxic lymphocytes. Little, however, is known about the effect of adenosine on myeloid cells. Considering that tumor associated macrophages (TAM) and myeloid-derived suppressor cells (MDSC) constitute up to 20 \% of OvCA tissue, we investigated the effect of adenosine on myeloid cells and explored a possible contribution of myeloid cells to adenosine generation in vitro and ex vivo. Methods Monocytes were used as human blood-derived myeloid cells. After co-incubation with SK-OV-3 or OAW-42 OvCA cells, monocyte migration was determined in transwell assays. For conversion into M2-polarized "TAM-like" macrophages, monocytes were co-incubated with OAW-42 cells. Ex vivo TAMs were obtained from OvCA ascites. Macrophage phenotypes were investigated by intracellular staining for IL-10 and IL-12. CD39 and CD73 expression were assessed by FACS analysis both on in vitro-induced TAM-like macrophages and on ascites-derived ex situ-TAMs. Myeloid cells in solid tumor tissue were analyzed by immunohistochemistry. Generation of biologically active adenosine by TAM-like macrophages was measured in luciferase-based reporter assays. Functional effects of adenosine were investigated in proliferation-experiments with CD4+ T cells and specific inhibitors. Results When CD39 or CD73 activity on OvCA cells were blocked, the migration of monocytes towards OvCA cells was significantly decreased. In vivo, myeloid cells in solid ovarian cancer tissue were found to express CD39 whereas CD73 was mainly detected on stromal fibroblasts. Ex situ-TAMs and in vitro differentiated TAM-like cells, however, upregulated the expression of CD39 and CD73 compared to monocytes or M1 macrophages. Expression of ectonucleotidases also translated into increased levels of biologically active adenosine. Accordingly, co-incubation with these TAMs suppressed CD4+ T cell proliferation which could be rescued via blockade of CD39 or CD73. Conclusion Adenosine generated by OvCA cells likely contributes to the recruitment of TAMs which further amplify adenosine-dependent immunosuppression via additional ectonucleotidase activity. In solid ovarian cancer tissue, TAMs express CD39 while CD73 is found on stromal fibroblasts. Accordingly, small molecule inhibitors of CD39 or CD73 could improve immune responses in ovarian cancer.}, language = {en} } @article{HaeuslerdelBarrioDiessneretal.2014, author = {H{\"a}usler, Sebastian F. M. and del Barrio, Itsaso Montalb{\´a}n and Diessner, Joachim and Stein, Roland G. and Strohschein, Jenny and H{\"o}nig, Arnd and Dietl, Johannes and Wischhusen, J{\"o}rg}, title = {Anti-CD39 and anti-CD73 antibodies A1 and 7G2 improve targeted therapy in ovarian cancer by blocking adenosine-dependent immune evasion}, series = {American Journal of Translational Research}, volume = {6}, journal = {American Journal of Translational Research}, number = {2}, issn = {1943-8141}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-120016}, pages = {129-139}, year = {2014}, abstract = {The ectonucleotidases CD39 and CD73 degrade ATP to adenosine which inhibits immune responses via the \(A_{2A}\) adenosine receptor (ADORA2A) on T and NK cells. The current study investigates the potential therapeutic use of the specific anti CD39- and anti CD73-antibodies A1 (CD39) and 7G2 (CD73) as these two ectonucleotidases are overexpressed in ovarian cancer (OvCA). As expected, NK cell cytotoxicity against the human ovarian cancer cell lines OAW-42 or SK-OV-3 was significantly increased in the presence of A1 or 7G2 antibody. While this might partly be due to antibody-dependent cell-mediated cytotoxicity, a luciferase-dependent assay for quantifying biologically active adenosine further showed that A1 and 7G2 can inhibit CD39 and CD73-dependent adenosine-generation. In turn, the reduction in adenosine levels achieved by addition of A1 and 7G2 to OAW-42 or SK-OV-3 cells was found to de-inhibit the proliferation of \(CD4^+\) T cells in coculture with OvCA cells. Likewise, blocking of CD39 and CD73 on OvCA cells via A1 and 7G2 led to an increased cytotoxicity of alloreactive primed T cells. Thus, antibodies like A1 and 7G2 could improve targeted therapy in ovarian cancer not only by specifically labeling overexpressed antigens but also by blocking adenosine-dependent immune evasion in this immunogenic malignancy.}, language = {en} } @article{HerbertFickHeydarianetal.2022, author = {Herbert, Saskia-Laureen and Fick, Andrea and Heydarian, Motaharehsadat and Metzger, Marco and W{\"o}ckel, Achim and Rudel, Thomas and Kozjak-Pavlovic, Vera and Wulff, Christine}, title = {Establishment of the SIS scaffold-based 3D model of human peritoneum for studying the dissemination of ovarian cancer}, series = {Journal of Tissue Engineering}, volume = {13}, journal = {Journal of Tissue Engineering}, issn = {2041-7314}, doi = {10.1177/20417314221088514}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-301311}, pages = {1}, year = {2022}, abstract = {Ovarian cancer is the second most common gynecological malignancy in women. More than 70\% of the cases are diagnosed at the advanced stage, presenting as primary peritoneal metastasis, which results in a poor 5-year survival rate of around 40\%. Mechanisms of peritoneal metastasis, including adhesion, migration, and invasion, are still not completely understood and therapeutic options are extremely limited. Therefore, there is a strong requirement for a 3D model mimicking the in vivo situation. In this study, we describe the establishment of a 3D tissue model of the human peritoneum based on decellularized porcine small intestinal submucosa (SIS) scaffold. The SIS scaffold was populated with human dermal fibroblasts, with LP-9 cells on the apical side representing the peritoneal mesothelium, while HUVEC cells on the basal side of the scaffold served to mimic the endothelial cell layer. Functional analyses of the transepithelial electrical resistance (TEER) and the FITC-dextran assay indicated the high barrier integrity of our model. The histological, immunohistochemical, and ultrastructural analyses showed the main characteristics of the site of adhesion. Initial experiments using the SKOV-3 cell line as representative for ovarian carcinoma demonstrated the usefulness of our models for studying tumor cell adhesion, as well as the effect of tumor cells on endothelial cell-to-cell contacts. Taken together, our data show that the novel peritoneal 3D tissue model is a promising tool for studying the peritoneal dissemination of ovarian cancer.}, language = {en} } @phdthesis{Heinlein2013, author = {Heinlein, U-Ju}, title = {Adenosinrezeptoren auf Ovarialkarzinomzellen}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-81920}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2013}, abstract = {Seit der Entdeckung, dass Adenosin auch als Botenstoff dient, besch{\"a}ftigen sich Forschungsgruppen mit Adenosinrezeptoren und ihrer m{\"o}glichen therapeutischen Modulation, insbesondere in Zusammenhang mit Krebserkrankungen. Bislang sind die Rezeptoren auf diversen Krebszellen nachgewiesen worden. So konnten beispielsweise in einer Brustkrebszelllinie A2B Adenosinrezeptoren nachgewiesen werden, deren Stimulation zu einer Hemmung der wachstumsf{\"o}rdernden MAP Kinase f{\"u}hrt. Pharmaka zur weitgehend selektiven Aktivierung oder Hemmung einzelner Adenosinrezeptor-Subtypen stehen ebenfalls zur Verf{\"u}gung. Beim Ovarialkarzinom mit seiner leider meist erst sp{\"a}t auftretenden Symptomatik besteht derzeit noch keine M{\"o}glichkeit zur fr{\"u}hen Diagnosestellung, sodass die Prognose ausgesprochen ung{\"u}nstig ausf{\"a}llt und die Erkrankung bei Frauen eine der h{\"a}ufigsten krebsbedingten Todesursachen darstellt. Daher war es ein Ziel dieser Arbeit herauszufinden, ob Adenosinrezeptoren auf diesen Zellen einen m{\"o}glichen therapeutischen Angriffspunkt bieten. Dazu untersuchten wir die vier Ovarialkarzinomzelllinien OVCAR-3, SK-OV-3, PA-1 und OAW-42 auf eine m{\"o}gliche Expression von allen vier Adenosinrezeptorsubtypen. Zun{\"a}chst wurden mit radioaktiv markierten Liganden ([3H]CCPA, [3H]NECA und [3H]HEMADO) Bindungsstudien f{\"u}r den A1-, A2A- und A3-Subtyp durchgef{\"u}hrt. Die Expression des A2B-Rezeptors wurde mithilfe eines funktionellen Nachweises, der Stimulation der Adenylylcyclase mithilfe von NECA (einem unspezifischen Adenosinrezeptoragonisten) analysiert. Im Anschluss daran untersuchten wir an OAW-42 und SK-OV-3 Zellen, ob sich ihr Proliferationsverhalten durch eine Stimulation mit NECA ver{\"a}ndern ließe und ob sich das Ansprechen auf g{\"a}ngige Chemotherapeutika bzw. einen Todesliganden {\"a}ndern w{\"u}rde. Trotz des erfolgreichen Nachweises von Adenosinrezeptoren auf allen Zelllinien waren die Ergebnisse der Proliferationsstudien aber nicht eindeutig. OAW-42 und SK-OV-3 Zellen reagierten zwar auf eine NECA-Stimulation mit sinkendem BrdU-Einbau, OAW-42 Zellen zeigten aber nach Behandlung mit NECA eine leicht erh{\"o}hte Resistenz gegen{\"u}ber Cisplatin. NECA-behandelte SK-OV-3 Zellen reagierten hingegen etwas sensitiver auf Doxorubicin und Fas-Ligand. Die Unterschiede waren aber insgesamt sehr gering und wurden daher von uns als nicht entscheidender Effekt gewertet. Auch Untersuchungen zur Expression der Adenosin-generierenden Enzyme CD39 und CD73 vor und nach NECA-Stimulation blieben ohne erkennbare Ver{\"a}nderung. Insofern ergaben unsere Untersuchungen keine Hinweise darauf, dass Adenosinrezeptoren eine m{\"o}gliche therapeutische Zielstruktur darstellen k{\"o}nnten. Zuk{\"u}nftige Studien k{\"o}nnen aber die gewonnenen Daten als Grundlage und Ausgangspunkt n{\"u}tzen, um auch andere Tumorzellarten zu untersuchen und im Kampf gegen den Krebs nach neuen potenziellen pharmakologischen Angriffspunkten zu suchen.}, subject = {Eierstockkrebs}, language = {de} } @article{HarterHaukeHeitzetal.2017, author = {Harter, Philipp and Hauke, Jan and Heitz, Florian and Reuss, Alexander and Kommoss, Stefan and Marm{\´e}, Frederik and Heimbach, Andr{\´e} and Prieske, Katharina and Richters, Lisa and Burges, Alexander and Neidhardt, Guido and de Gregorio, Nikolaus and El-Balat, Ahmed and Hilpert, Felix and Meier, Werner and Kimmig, Rainer and Kast, Karin and Sehouli, Jalid and Baumann, Klaus and Jackisch, Christian and Park-Simon, Tjoung-Won and Hanker, Lars and Kr{\"o}ber, Sandra and Pfisterer, Jacobus and Gevensleben, Heidrun and Schnelzer, Andreas and Dietrich, Dimo and Neunh{\"o}ffer, Tanja and Krockenberger, Mathias and Brucker, Sara Y. and N{\"u}rnberg, Peter and Thiele, Holger and Altm{\"u}ller, Janine and Lamla, Josefin and Elser, Gabriele and du Bois, Andreas and Hahnen, Eric and Schmutzler, Rita}, title = {Prevalence of deleterious germline variants in risk genes including \(BRCA1/2\) in consecutive ovarian cancer patients (AGO-TR-1)}, series = {PLoS ONE}, volume = {12}, journal = {PLoS ONE}, number = {10}, doi = {10.1371/journal.pone.0186043}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-173553}, year = {2017}, abstract = {Background Identification of families at risk for ovarian cancer offers the opportunity to consider prophylactic surgery thus reducing ovarian cancer mortality. So far, identification of potentially affected families in Germany was solely performed via family history and numbers of affected family members with breast or ovarian cancer. However, neither the prevalence of deleterious variants in \(BRCA1/2\) in ovarian cancer in Germany nor the reliability of family history as trigger for genetic counselling has ever been evaluated. Methods Prospective counseling and germline testing of consecutive patients with primary diagnosis or with platinum-sensitive relapse of an invasive epithelial ovarian cancer. Testing included 25 candidate and established risk genes. Among these 25 genes, 16 genes (\(ATM\), \(BRCA1\), \(BRCA2\), \(CDH1\), \(CHEK2\), \(MLH1\), \(MSH2\), \(MSH6\), \(NBN\), \(PMS2\), \(PTEN\), \(PALB2\), \(RAD51C\), \(RAD51D\), \(STK11\), \(TP53\)) were defined as established cancer risk genes. A positive family history was defined as at least one relative with breast cancer or ovarian cancer or breast cancer in personal history. Results In total, we analyzed 523 patients: 281 patients with primary diagnosis of ovarian cancer and 242 patients with relapsed disease. Median age at primary diagnosis was 58 years (range 16-93) and 406 patients (77.6\%) had a high-grade serous ovarian cancer. In total, 27.9\% of the patients showed at least one deleterious variant in all 25 investigated genes and 26.4\% in the defined 16 risk genes. Deleterious variants were most prevalent in the \(BRCA1\) (15.5\%), \(BRCA2\) (5.5\%), \(RAD51C\) (2.5\%) and \(PALB2\) (1.1\%) genes. The prevalence of deleterious variants did not differ significantly between patients at primary diagnosis and relapse. The prevalence of deleterious variants in \(BRCA1/2\) (and in all 16 risk genes) in patients <60 years was 30.2\% (33.2\%) versus 10.6\% (18.9\%) in patients \(\geq\)60 years. Family history was positive in 43\% of all patients. Patients with a positive family history had a prevalence of deleterious variants of 31.6\% (36.0\%) versus 11.4\% (17.6\%) and histologic subtype of high grade serous ovarian cancer versus other showed a prevalence of deleterious variants of 23.2\% (29.1\%) and 10.2\% (14.8\%), respectively. Testing only for \(BRCA1/2\) would miss in our series more than 5\% of the patients with a deleterious variant in established risk genes. Conclusions 26.4\% of all patients harbor at least one deleterious variant in established risk genes. The threshold of 10\% mutation rate which is accepted for reimbursement by health care providers in Germany was observed in all subgroups analyzed and neither age at primary diagnosis nor histo-type or family history sufficiently enough could identify a subgroup not eligible for genetic counselling and testing. Genetic testing should therefore be offered to every patient with invasive epithelial ovarian cancer and limiting testing to \(BRCA1/2\) seems to be not sufficient.}, language = {en} } @article{FernandezRodriguezQuilesBlancoetal.2012, author = {Fern{\´a}ndez-Rodr{\´i}guez, Juana and Quiles, Francisco and Blanco, Ignacio and Teul{\´e}, Alex and Feliubadal{\´o}, L{\´i}dia and del Valle, Jes{\´u}s and Salinas, M{\´o}nica and Izquierdo, {\´A}ngel and Darder, Esther and Schindler, Detlev and Capell{\´a}, Gabriel and Brunet, Joan and L{\´a}zaro, Conxi and Angel Pujana, Miguel}, title = {Analysis of SLX4/FANCP in non-BRCA1/2-mutated breast cancer families}, series = {BMC Cancer}, volume = {12}, journal = {BMC Cancer}, number = {84}, doi = {10.1186/1471-2407-12-84}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-131772}, year = {2012}, abstract = {Background: Genes that, when mutated, cause Fanconi anemia or greatly increase breast cancer risk encode for proteins that converge on a homology-directed DNA damage repair process. Mutations in the SLX4 gene, which encodes for a scaffold protein involved in the repair of interstrand cross-links, have recently been identified in unclassified Fanconi anemia patients. A mutation analysis of SLX4 in German or Byelorussian familial cases of breast cancer without detected mutations in BRCA1 or BRCA2 has been completed, with globally negative results. Methods: The genomic region of SLX4, comprising all exons and exon-intron boundaries, was sequenced in 94 Spanish familial breast cancer cases that match a criterion indicating the potential presence of a highly-penetrant germline mutation, following exclusion of BRCA1 or BRCA2 mutations. Results: This mutational analysis revealed extensive genetic variation of SLX4, with 21 novel single nucleotide variants; however, none could be linked to a clear alteration of the protein function. Nonetheless, genotyping 10 variants (nine novel, all missense amino acid changes) in a set of controls (138 women and 146 men) did not detect seven of them. Conclusions: Overall, while the results of this study do not identify clearly pathogenic mutations of SLX4 contributing to breast cancer risk, further genetic analysis, combined with functional assays of the identified rare variants, may be warranted to conclusively assess the potential link with the disease.}, language = {en} } @phdthesis{Dombrowski2010, author = {Dombrowski, Yvonne}, title = {Charakterisierung von GDF-15 als Immunmodulator im Ovarialkarzinom}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-48885}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2010}, abstract = {GDF-15 ist ein atypisches Mitglied der TGF-b-Superfamilie. Unter physiologischen Bedingungen kommt es nur in der Plazenta in gr{\"o}ßeren Mengen vor, w{\"a}hrend es in zahlreichen Tumoren {\"u}berexprimiert gefunden wurde. Die genaue Funktion von GDF-15 im Tumorkontext ist nicht genau gekl{\"a}rt. Aufgrund der h{\"a}ufigen und hohen Expression in Tumoren scheint GDF-15 eine wesentliche Funktion im Tumorprogress auszu{\"u}ben. Das Ovarialkarzinom (OvCA) nimmt die Stellung als t{\"o}dlichste gyn{\"a}kologische Erkrankung ein. Da der Tumor meist erst in fortgeschrittenen Stadien diagnostiziert wird, sind bis heute die Heilungschancen schlecht. H{\"a}ufig kommt es zum Rezidiv nach zun{\"a}chst erfolgreicher Chemotherapie und mit 30\% ist die 5-Jahres-{\"U}berlebenschance gering. F{\"u}r die chemoresistenten F{\"a}lle gibt es bis zum heutigen Zeitpunkt keine effektive Therapie. Dies verdeutlicht die Notwendigkeit, neue innovative Therapiestrategien zu entwickeln. G{\"u}nstige immunologische Parameter korrelieren mit der {\"U}berlebensdauer von OvCA-Patientinnen, was die Immuntherapie beim OvCA in den Fokus der experimentellen klinischen Therapie r{\"u}ckt. Doch um neue immuntherapeutische Strategien entwickeln zu k{\"o}nnen, m{\"u}ssen zun{\"a}chst immunologisch relevante Angriffspunkte identifiziert werden. Das in vielen Tumoren exprimierte GDF-15 ist mit einem der st{\"a}rksten immunsuppressiven Faktoren verwandt, was die Vermutung nahe legt, dass auch GDF-15 eine immunologisch relevante Funktion im Tumorkontext aus{\"u}ben k{\"o}nnte. Daher wurden die Expression und die m{\"o}gliche Funktion von GDF-15 als Immunmodulator im Ovarialkarzinom untersucht. Expressionsanalysen von OvCA-Gewebe und prim{\"a}ren OvCA-Zellen zeigten, dass GDF-15 das am st{\"a}rksten {\"u}berexprimierte Gen der untersuchten TGF-b-Familienmitglieder im OvCA ist. Auch als sezerniertes Protein wird GDF-15 in vivo und in vitro im OvCA detektiert, was auf eine funktionale Rolle von GDF-15 im OvCA hindeutet. Normalerweise eliminiert das Immunsystem entartete k{\"o}rpereigene Zellen. Manchmal gelingt es Tumorzellen jedoch, sich dieser Immun{\"u}berwachung zu entziehen und dem Immunsystem zu „entwischen". Inwieweit GDF-15 bei der Koordination des „immune escape" des OvCA eine Rolle spielt, sollte im Fokus dieser Arbeit stehen. Das Hauptaugenmerk lag dabei auf der Wirkung von GDF-15 auf NK-Zellen, da diese als fr{\"u}he Effektoren und wichtige Mediatoren zwischen angeborenem und adaptivem Immunsystem nicht nur eine Schl{\"u}sselrolle bei der immunologische {\"U}berwachung spielen, sondern sich dadurch auch als ideale Werkzeuge f{\"u}r die Tumorimmuntherapie auszeichnen. Exogenes GDF-15 hemmt in vitro die Lyseaktivit{\"a}t von NK-Zellen gegen{\"u}ber OvCA-Zellen. Endogene GDF-15-Defizienz der OvCA-Zellen sensitiviert diese f{\"u}r NK-Zell-Lyse und endogene GDF-15-{\"U}berexpression mindert die NK-Lyseaktivit{\"a}t. Die Hemmung der NK-Lyseaktivit{\"a}t kann durch verschiedene synergistisch wirkende Mechanismen erfolgen: durch Rezeptormodulation, durch direkte Modulation des Lysemechanismus und durch Apoptoseregulation. Wie TGF-b1 reguliert GDF-15 die Expression des aktivierenden NK-Rezeptors NKG2D von der Zelloberfl{\"a}che herunter und induziert zus{\"a}tzlich die Expression des inhibierenden Rezeptors CD305 und die des mit NKG2A- und NKG2C-assoziierten Rezeptors CD94. Daneben greift GDF-15 direkt in den Lysemechanismus der NK-Zellen ein, indem es die Granzym B-Expression beeinflusst. Dar{\"u}ber hinaus sensitiviert GDF-15 Immunzellen f{\"u}r die Apoptose durch die Induktion von Fas/CD95. Signaltransduktionsanalysen zeigen, dass GDF-15 in Immunzellen die SMAD-Proteine zeitverz{\"o}gert zu TGF-b aktiviert, was auf eine indirekte Wirkung schließen l{\"a}sst. Zus{\"a}tzlich kann GDF-15 auch die die p38/MAPK in Immunzellen aktivieren. Die Genregulation von GDF-15 und TGF-b1 in NK-Zellen ist sehr verschieden. Beide Zytokine regulieren {\"u}berwiegend Gene aus gleichen Funktionalit{\"a}tsclustern, allerdings sind die einzelnen von TGF-b1 und GDF-15 regulierten Gene verschieden. Nur drei Gene (CD55, Caspase-8 und Apolipoprotein 6) sind durch GDF-15 und TGF-b1 gleich reguliert. Zusammengefasst zeigt sich eine funktionale Analogie von GDF-15 und TGF-b1 in NK-Zellen. TGF-b1 scheint eine st{\"a}rkere Wirkung zu induzieren, daf{\"u}r zeigt GDF-15 hier ein breiteres Funktionalit{\"a}tsspektrum. Durch die Charakterisierung der funktionalen Rolle von GDF-15 als Immunmodulator in Tumoren ist hier ein neuer potentieller Angriffspunkt identifiziert worden, welcher Grundlage f{\"u}r neue Tumortherapiestrategien, nicht nur f{\"u}r das OvCA, sondern auch f{\"u}r andere GDF-15-exprimierende Tumore sein kann.}, subject = {Immunmodulator}, language = {de} } @phdthesis{Bruttel2015, author = {Bruttel, Valentin Stefan}, title = {Soluble HLA-G binds to dendritic cells which likely suppresses anti-tumour immune responses in regional lymph nodes in ovarian carcinoma}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-127252}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2015}, abstract = {Zusammenfassung Einleitung HLA-G, ein nicht-klassisches HLA bzw. MHC Klasse Ib Molek{\"u}l, kann sowohl als membrangebundenes als auch als l{\"o}sliches Molek{\"u}l verschiedenste Immunzellpopulationen effektiv inhibieren. Unter physiologischen Bedingungen wird HLA-G vor allem in der Plazenta exprimiert, wo es dazu beitr{\"a}gt den semiallogenen Embryo vor einer Abstoßung durch das m{\"u}tterliche Immunsystem zu besch{\"u}tzen. Außerdem wird HLA-G in einer Vielzahl von Tumoren wie zum Beispiel in Ovarialkarzinomen {\"u}berexprimiert. Ziel dieser Arbeit war es besonders die Rolle von l{\"o}slichem HLA-G im Ovarialkarzinom und die Expression von HLA-G in verschiedenen Subtypen des Ovarialkarzinoms genauer zu untersuchen. Ergebnisse Anhand eines Tissue Microarrays wurde best{\"a}tigt dass HLA-G unter physiologischen Bedingungen nur in sehr wenigen Geweben wie Plazenta oder Testes exprimiert wird. Außerdem wurden erstmals auch im Nebennierenmark hohe Expressionslevel detektiert. Im Gegensatz zur physiologischen Expression wurde HLA-G in ser{\"o}sen, muzin{\"o}sen, endometrioiden und Klarzellkarzinomen und somit in Tumoren aller untersuchten Subtypen des Ovarialkarzinoms detektiert. Am h{\"a}ufigsten war HLA-G in hochgradigen ser{\"o}sen Karzinomen {\"u}berexprimiert. Hier konnte gezeigt werden dass auf Genexpressionslevel in Ovarialkarzinomen die Expression des immunsuppressiven HLA-G mit der Expression von klassischen MHC Molek{\"u}len wie HLA-A, -B oder -C hochsignifikant korreliert. Außerdem konnte in Aszitesproben von Patientinnen mit Ovarialkarzinomen hohe Konzentrationen von l{\"o}slichem HLA-G nachgewiesen werden. Auch auf metastasierten Tumorzellen in regionalen Lymphknoten war HLA-G nachweisbar. {\"U}berraschenderweise wurde aber besonders viel HLA-G auf Dendritischen Zellen in Lymphknoten detektiert. Da in Monozyten und Dendritischen Zellen von gesunden Spendern durch IL-4 oder IL-10 im Gegensatz zu Literatur keine Expression von HLA-G induzierbar war, untersuchten wir ob Dendritische Zellen l{\"o}sliches HLA-G binden. Es konnte gezeigt werden, dass besonders Dendritische Zellen die in Gegenwart von IL-4, IL-10 und GM-CSF aus Monozyten generiert wurden (DC-10) effektiv l{\"o}sliches HLA-G {\"u}ber ILT Rezeptoren binden. In Abh{\"a}ngigkeit von ihrer Beladung mit HLA-G hemmen auch fixierte DC-10 Zellen noch die Proliferation von zytotoxischen CD8+ T Zellen. Zudem wurden regulatorische T Zellen induziert. Schlussfolgerungen Besonders in den am h{\"a}ufigsten diagnostizierten hochgradigen ser{\"o}sen Ovarialkarzinomen ist HLA-G in den meisten F{\"a}llen {\"u}berexprimiert. Durch die Expression immunsuppressiver MHC Klasse Ib Molek{\"u}le wie HLA-G k{\"o}nnen wahrscheinlich auch Tumore wachsen, die noch klassische MHC Molek{\"u}le exprimieren und aufgrund ihrer Mutationslast eigentlich vom Immunsystem erkannt und eliminiert werden m{\"u}ssten. L{\"o}sliches HLA-G k{\"o}nnte zudem lokal Immunantworten gegen Tumorantigene unterdr{\"u}cken indem es an Dendritische Zellen in regionalen Lymphknoten bindet. Diese Zellen pr{\"a}sentieren nomalerweise zytotoxischen T Zellen Tumorantigene und spielen daher eine entscheidende Rolle in der Entstehung von protektiven Immunantworten. Mit l{\"o}slichem HLA-G beladene Dendritische Zellen hemmen jedoch die Proliferation von CD8+ T Zellen und induzieren regulatorische T Zellen. Dadurch k{\"o}nnten Ovarialkarzinome "aus der Ferne" auch in metastasenfreien Lymphknoten die Entstehung von gegen den Tumor gerichteten Immunantworten unterdr{\"u}cken. Dieser erstmals beschriebene Mechanismus k{\"o}nnte auch in anderen malignen Erkrankungen eine Rolle spielen, da l{\"o}sliches HLA-G in einer Vielzahl von Tumorindikationen nachgewiesen wurde.}, subject = {HLA-G}, language = {en} } @article{BekesLoebHolzheuetal.2019, author = {Bekes, Inga and L{\"o}b, Sanja and Holzheu, Iris and Janni, Wolfgang and Baumann, Lisa and W{\"o}ckel, Achim and Wulff, Christine}, title = {Nectin-2 in ovarian cancer: how is it expressed and what might be its functional role?}, series = {Cancer Science}, volume = {110}, journal = {Cancer Science}, number = {6}, doi = {10.1111/cas.13992}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-202748}, pages = {1872- 1882}, year = {2019}, abstract = {Nectin-2 is an adhesion molecule that has been reported to play a role in tumor growth, metastasis and tumor angiogenesis. Herein, we investigated Nectin-2 in ovarian cancer patients and in cell culture. Tumor as well as peritoneal biopsies of 60 ovarian cancer patients and 22 controls were dual stained for Nectin-2 and CD31 using immunohistochemistry. Gene expression of Nectin-2 was quantified by real-time PCR and differences analyzed in relation to various tumor characteristics. In the serum of patients, vascular endothelial growth factor (VEGF) was quantified by ELISA. Effect of VEGF on Nectin-2 expression as well as permeability was investigated in HUVEC. In tumor biopsies, Nectin-2 protein was mainly localized in tumor cells, whereas in peritoneal biopsies, clear colocalization was found in the vasculature. T3 patients had a significantly higher percentage of positive lymph nodes and this correlated with survival. Nectin-2 was significantly upregulated in tumor biopsies in patients with lymph node metastasis and with residual tumor >1 cm after surgery. Nectin-2 expression was significantly suppressed in the peritoneal endothelium of patients associated with significantly increased VEGF serum levels. In cell culture, VEGF stimulation led to a significant downregulation of Nectin-2 which was reversed by VEGF-inhibition. In addition, Nectin-2 knockdown in endothelial cells was associated with significantly increased endothelial permeability. Nectin-2 expression in ovarian cancer may support tumor cell adhesion, leading to growth and lymph node metastasis. In addition, VEGF-induced Nectin-2 suppression in peritoneal endothelium may support an increase in vascular permeability leading to ascites production.}, language = {en} } @article{BekesFriedlKoehleretal.2016, author = {Bekes, Inga and Friedl, Thomas W. P. and K{\"o}hler, Tanja and M{\"o}bus, Volker and Janni, Wolfgang and W{\"o}ckel, Achim and Wulff, Christine}, title = {Does VEGF facilitate local tumor growth and spread into the abdominal cavity by suppressing endothelial cell adhesion, thus increasing vascular peritoneal permeability followed by ascites production in ovarian cancer?}, series = {Molecular Cancer}, volume = {15}, journal = {Molecular Cancer}, number = {13}, doi = {10.1186/s12943-016-0497-3}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-169298}, year = {2016}, abstract = {Background Ovarian cancer is mostly associated with pathologically regulated permeability of peritoneal vessels, leading to ascites. Here, we investigated the molecular regulation of endothelial permeability by the vascular endothelial growth factor (VEGF) and both tight and adherens junction proteins (VE-cadherin and claudin 5) with regards to the tumor biology of different ovarian cancer types. Methods Serum and ascites samples before and after surgery, as well as peritoneal biopsies of 68 ovarian cancer patients and 20 healthy controls were collected. In serum and ascites VEGF protein was measured by ELISA. In peritoneal biopsies co-localization of VE-cadherin and claudin 5 was investigated using immunohistochemical dual staining. In addition, the gene expression of VE-cadherin and claudin 5 was quantified by Real-time PCR. Differences in VEGF levels, VE-cadherin and claudin 5 gene expression were analyzed in relation to various tumor characteristics (tumor stage, grading, histological subtypes, resection status after surgery) and then compared to controls. Furthermore, human primary ovarian cancer cells were co-cultured with human umbilical vein endothelial cells (HUVEC) and changes in VE-cadherin and claudin 5 were investigated after VEGF inhibition. Results VEGF was significantly increased in tumor patients in comparison to controls and accumulates in ascites. The highest VEGF levels were found in patients diagnosed with advanced tumor stages, with tumors of poor differentiation, or in the group of solid / cystic-solid tumors. Patients with residual tumor after operation showed significantly higher levels of VEGF both before and after surgery as compared to tumor-free resected patients. Results of an immunohistochemical double-staining experiment indicated co-localization of VE-cadherin and claudin 5 in the peritoneal vasculature. Compared to controls, expression of VE-cadherin and claudin 5 was significantly suppressed in peritoneal vessels of tumor patients, but there were no significant differences regarding VE-cadherin and claudin 5 expression in relation to different tumor characteristics. A significant positive correlation was found between VE-cadherin and claudin 5 expression. VEGF inhibition in vitro was associated with significant increase in VE-cadherin and claudin 5. Conclusions Our results indicate that increased peritoneal permeability in ovarian cancer is due to down-regulation of adhesion proteins via tumor derived VEGF. Advanced ovarian cancer with aggressive tumor biology may be associated with early dysregulation of vascular permeability leading to ascites. These patients may benefit from therapeutic VEGF inhibition.}, language = {en} } @article{AntoniouKuchenbaeckerSoucyetal.2012, author = {Antoniou, Antonis C. and Kuchenbaecker, Karoline B. and Soucy, Penny and Beesley, Jonathan and Chen, Xiaoqing and McGuffog, Lesley and Lee, Andrew and Barrowdale, Daniel and Healey, Sue and Sinilnikova, Olga M. and Caligo, Maria A. and Loman, Niklas and Harbst, Katja and Lindblom, Annika and Arver, Brita and Rosenquist, Richard and Karlsson, Per and Nathanson, Kate and Domchek, Susan and Rebbeck, Tim and Jakubowska, Anna and Lubinski, Jan and Jaworska, Katarzyna and Durda, Katarzyna and Zlowowcka-Perłowska, Elżbieta and Osorio, Ana and Dur{\´a}n, Mercedes and Andr{\´e}s, Raquel and Ben{\´i}tez, Javier and Hamann, Ute and Hogervorst, Frans B. and van Os, Theo A. and Verhoef, Senno and Meijers-Heijboer, Hanne E. J. and Wijnen, Juul and Garcia, Encarna B. G{\´o}mez and Ligtenberg, Marjolijn J. and Kriege, Mieke and Coll{\´e}e, Margriet and Ausems, Margreet G. E. M. and Oosterwijk, Jan C. and Peock, Susan and Frost, Debra and Ellis, Steve D. and Platte, Radka and Fineberg, Elena and Evans, D. Gareth and Lalloo, Fiona and Jacobs, Chris and Eeles, Ros and Adlard, Julian and Davidson, Rosemarie and Cole, Trevor and Cook, Jackie and Paterson, Joan and Douglas, Fiona and Brewer, Carole and Hodgson, Shirley and Morrison, Patrick J. and Walker, Lisa and Rogers, Mark T. and Donaldson, Alan and Dorkins, Huw and Godwin, Andrew K. and Bove, Betsy and Stoppa-Lyonnet, Dominique and Houdayer, Claude and Buecher, Bruno and de Pauw, Antoine and Mazoyer, Sylvie and Calender, Alain and L{\´e}on{\´e}, M{\´e}lanie and Bressac-de Paillerets, Brigitte and Caron, Olivier and Sobol, Hagay and Frenay, Marc and Prieur, Fabienne and Ferrer, Sandra Fert and Mortemousque, Isabelle and Buys, Saundra and Daly, Mary and Miron, Alexander and Terry, Mary Beth and Hopper, John L. and John, Esther M. and Southey, Melissa and Goldgar, David and Singer, Christian F. and Fink-Retter, Anneliese and Muy-Kheng, Tea and Geschwantler Kaulich, Daphne and Hansen, Thomas V. O. and Nielsen, Finn C. and Barkardottir, Rosa B. and Gaudet, Mia and Kirchhoff, Tomas and Joseph, Vijai and Dutra-Clarke, Ana and Offit, Kenneth and Piedmonte, Marion and Kirk, Judy and Cohn, David and Hurteau, Jean and Byron, John and Fiorica, James and Toland, Amanda E. and Montagna, Marco and Oliani, Cristina and Imyanitov, Evgeny and Isaacs, Claudine and Tihomirova, Laima and Blanco, Ignacio and Lazaro, Conxi and Teul{\´e}, Alex and Del Valle, J. and Gayther, Simon A. and Odunsi, Kunle and Gross, Jenny and Karlan, Beth Y. and Olah, Edith and Teo, Soo-Hwang and Ganz, Patricia A. and Beattie, Mary S. and Dorfling, Cecelia M. and Jansen van Rensburg, Elizabeth and Diez, Orland and Kwong, Ava and Schmutzler, Rita K. and Wappenschmidt, Barbara and Engel, Christoph and Meindl, Alfons and Ditsch, Nina and Arnold, Norbert and Heidemann, Simone and Niederacher, Dieter and Preisler-Adams, Sabine and Gadzicki, Dorothea and Varon-Mateeva, Raymonda and Deissler, Helmut and Gehrig, Andrea and Sutter, Christian and Kast, Karin and Fiebig, Britta and Sch{\"a}fer, Dieter and Caldes, Trinidad and de la Hoya, Miguel and Nevanlinna, Heli and Muranen, Taru A. and Lesp{\´e}rance, Bernard and Spurdle, Amanda B. and Neuhausen, Susan L. and Ding, Yuan C. and Wang, Xianshu and Fredericksen, Zachary and Pankratz, Vernon S. and Lindor, Noralane M. and Peterlongo, Paulo and Manoukian, Siranoush and Peissel, Bernard and Zaffaroni, Daniela and Bonanni, Bernardo and Bernard, Loris and Dolcetti, Riccardo and Papi, Laura and Ottini, Laura and Radice, Paolo and Greene, Mark H. and Loud, Jennifer T. and Andrulis, Irene L. and Ozcelik, Hilmi and Mulligan, Anna Marie and Glendon, Gord and Thomassen, Mads and Gerdes, Anne-Marie and Jensen, Uffe B. and Skytte, Anne-Bine and Kruse, Torben A. and Chenevix-Trench, Georgia and Couch, Fergus J. and Simard, Jacques and Easton, Douglas F.}, title = {Common variants at 12p11, 12q24, 9p21, 9q31.2 and in ZNF365 are associated with breast cancer risk for BRCA1 and/or BRCA2 mutation carriers}, series = {Breast Cancer Research}, volume = {14}, journal = {Breast Cancer Research}, number = {R33}, organization = {CIMBA; SWE-BRCA; HEBON; EMBRACE; GEMO Study Collaborators; kConFab Investigators}, doi = {10.1186/bcr3121}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-130449}, year = {2012}, abstract = {Introduction: Several common alleles have been shown to be associated with breast and/or ovarian cancer risk for BRCA1 and BRCA2 mutation carriers. Recent genome-wide association studies of breast cancer have identified eight additional breast cancer susceptibility loci: rs1011970 (9p21, CDKN2A/B), rs10995190 (ZNF365), rs704010 (ZMIZ1), rs2380205 (10p15), rs614367 (11q13), rs1292011 (12q24), rs10771399 (12p11 near PTHLH) and rs865686 (9q31.2). Methods: To evaluate whether these single nucleotide polymorphisms (SNPs) are associated with breast cancer risk for BRCA1 and BRCA2 carriers, we genotyped these SNPs in 12,599 BRCA1 and 7,132 BRCA2 mutation carriers and analysed the associations with breast cancer risk within a retrospective likelihood framework. Results: Only SNP rs10771399 near PTHLH was associated with breast cancer risk for BRCA1 mutation carriers (per-allele hazard ratio (HR) = 0.87, 95\% CI: 0.81 to 0.94, P-trend = 3 x 10\(^{-4}\)). The association was restricted to mutations proven or predicted to lead to absence of protein expression (HR = 0.82, 95\% CI: 0.74 to 0.90, P-trend = 3.1 x 10\(^{-5}\), P-difference = 0.03). Four SNPs were associated with the risk of breast cancer for BRCA2 mutation carriers: rs10995190, P-trend = 0.015; rs1011970, P-trend = 0.048; rs865686, 2df P = 0.007; rs1292011 2df P = 0.03. rs10771399 (PTHLH) was predominantly associated with estrogen receptor (ER)-negative breast cancer for BRCA1 mutation carriers (HR = 0.81, 95\% CI: 0.74 to 0.90, P-trend = 4 x 10\(^{-5}\)) and there was marginal evidence of association with ER- negative breast cancer for BRCA2 mutation carriers (HR = 0.78, 95\% CI: 0.62 to 1.00, P-trend = 0.049). Conclusions: The present findings, in combination with previously identified modifiers of risk, will ultimately lead to more accurate risk prediction and an improved understanding of the disease etiology in BRCA1 and BRCA2 mutation carriers.}, language = {en} }