Ovarian cancer predisposition beyond BRCA1 and BRCA2 genes

2020 ◽  
Vol 30 (11) ◽  
pp. 1803-1810
Author(s):  
Antonella Pietragalla ◽  
Martina Arcieri ◽  
Claudia Marchetti ◽  
Giovanni Scambia ◽  
Anna Fagotti

Several genes associated with hereditary ovarian cancer have been discovered as a result of the work done with next generation sequencing. It is estimated that approximately 23% of ovarian carcinomas have a hereditary predisposition. The most common hereditary condition is represented by germline mutations in BRCA1 or BRCA2 genes that account for 20–25% of high grade serous ovarian cancer. A number of other hereditary ovarian cancers are associated with different genes, with a crucial role in the DNA damage response pathway, such as the mismatch repair genes in Lynch syndrome, TP53 in Li-Fraumeni syndrome, STK11 in Peutz-Jeghers syndrome, CHEK2, RAD51, BRIP1, and PALB2. The goal of this manuscript is to summarize the published data regarding the molecular pathways involved in the pathogenesis of non-BRCA related hereditary ovarian cancer and to provide a tool that might be useful in discussing risk assessment, genetic testing, prevention strategies, as well as clinical and therapeutic implications for patients with ovarian cancer.

2011 ◽  
Vol 152 (40) ◽  
pp. 1596-1608 ◽  
Author(s):  
József Gábor Joó ◽  
Szabolcs Ládi ◽  
B. Zsolt Nagy ◽  
Zoltán Langmár

Mutations in BRCA1 and BRCA2 genes account for the majority of hereditary breast and ovarian cancers. Approximately 10% of cases of ovarian cancer are due to germline mutations in BRCA1 and BRCA2. Ovarian cancer associated with BRCA1 and BRCA2 mutations has a distinct histological phenotype. This type of cancer is predominantly of serous or endometrioid histology and is high grade. Patients with BRCA1 or BRCA2 mutations should be offered risk-reducing salpingo-oophorectomy by age 40 years, or when childbearing is complete. Nowadays there are no differences between the treatments provided for sporadic and hereditary ovarian cancer, although there are indications that targeted therapy is effective in women with BRCA1/BRCA2-associated tumors. Retrospective studies reveal a high level of sensitivity to platinum agents in BRCA-associated tumors and initial trials show good efficacy and tolerability for polyADP-ribose polymerase inhibitors in mutation carriers with advanced ovarian cancers. These agents might also potentially be used in chemoprevention. Authors review the current management of hereditary ovarian cancer. Orv. Hetil., 2011, 152, 1596–1608.


Cancers ◽  
2020 ◽  
Vol 12 (10) ◽  
pp. 2834
Author(s):  
Ana Barbosa ◽  
Pedro Pinto ◽  
Ana Peixoto ◽  
Joana Guerra ◽  
Carla Pinto ◽  
...  

Since the approval of PARP inhibitors for the treatment of high-grade serous ovarian cancer, in addition to cancer risk assessment, BRCA1 and BRCA2 genetic testing also has therapeutic implications (germline and somatic variants) and should be offered to these patients at diagnosis, irrespective of family history. However, variants in other genes besides BRCA1 and BRCA2 are associated with ovarian cancer predisposition, which would be missed by a genetic testing aimed only at indication for PARP inhibitor treatment. In this study, we aimed to evaluate the yield of clinically actionable germline variants using next-generation sequencing of a customized panel of 10 genes for the analysis of formalin-fixed paraffin-embedded samples from 96 ovarian carcinomas, a strategy that allows the detection of both somatic and germline variants in a single test. In addition to 13.7% of deleterious germline BRCA1/BRCA2 carriers, we identified 7.4% additional patients with pathogenic germline variants in other genes predisposing for ovarian cancer, namely RAD51C, RAD51D, and MSH6, representing 35% of all pathogenic germline variants. We conclude that the strategy of reflex gene-panel tumor testing enables the identification of clinically actionable germline variants in a significantly higher proportion of ovarian cancer patients, which may be valuable information in patients with advanced disease that have run out of approved therapeutic options. Furthermore, this approach increases the chance to make available genetic counseling, presymptomatic genetic testing, and gynecological cancer prophylaxis to female relatives who turn out to be healthy carriers of deleterious germline variants.


Cancers ◽  
2020 ◽  
Vol 12 (8) ◽  
pp. 2151 ◽  
Author(s):  
Carolina Velázquez ◽  
De Leeneer K. ◽  
Eva M. Esteban-Cardeñosa ◽  
Francisco Avila Cobos ◽  
Enrique Lastra ◽  
...  

In this study, we aim to gain insight in the germline mutation spectrum of ATM, BARD1, BRIP1, ERCC4, PALB2, RAD51C and RAD51D in breast and ovarian cancer families from Spain. We have selected 180 index cases in whom a germline mutation in BRCA1 and BRCA2 was previously ruled out. The importance of disease-causing variants in these genes lies in the fact that they may have possible therapeutic implications according to clinical guidelines. All variants were assessed by combined annotation dependent depletion (CADD) for scoring their deleteriousness. In addition, we used the cancer genome interpreter to explore the implications of some variants in drug response. Finally, we compiled and evaluated the family history to assess whether carrying a pathogenic mutation was associated with age at diagnosis, tumour diversity of the pedigree and total number of cancer cases in the family. Eight unequivocal pathogenic mutations were found and another fourteen were prioritized as possible causal variants. Some of these molecular results could contribute to cancer diagnosis, treatment selection and prevention. We found a statistically significant association between tumour diversity in the family and carrying a variant with a high score predicting pathogenicity (p = 0.0003).


Author(s):  
Р.Р. Фаисханова ◽  
Д.С. Прокофьева ◽  
Э.К. Хуснутдинова ◽  
Д.Д. Сакаева ◽  
М.Г. Гордиев

Значительное количество злокачественных новообразований яичников являются наследственными (до 30% всех новообразований возникают в результате высокой генетической предрасположенности). Известно, по меньшей мере, 16 генов-кандидатов наследственного рака яичников (РЯ), а с внедрением и широким применением полногеномного секвенирования растет число генов и генетических вариантов, потенциально вовлеченных в патогенез семейных форм заболевания, хотя вклад этих генов в развитие наследственного РЯ еще предстоит доказать. Обнаружение специфических мутаций в ряде генов-кандидатов РЯ у здоровых женщин может оправдать не только более интенсивные и персонализированные программы наблюдения пациенток из групп риска, химиопрофилактики и/или профилактических операций, но и может дать фундаментальные знания о патогенезе развития опухолей. В статье описаны особенности клинического течения наследственного РЯ, обусловленного мутациями в ключевых генах кандидатах (BRCA1, BRCA2, TP53, BARD1, CHEK2, RAD51, PALB2). Описаны основные типы мутаций при BRCA-ассоциированном РЯ, возрастные особенности и показатели выживаемости. Показано, что большая часть (примерно 65-85%) наследственных опухолей яичников приходится на герминальные мутации в генах BRCA1 и BRCA2. Считается, что мутации в данных генах приводят прежде всего к онкогинекологическим заболеваниям в эстрогенчувствительных органах-мишенях, однако по некоторым данным имеется также вероятность возникновения рака желудка, толстой кишки, эндометрия, поджелудочной железы, меланомы кожи, опухолей мочевого пузыря, головы и шеи. Приблизительно 6% наследственного РЯ приходится на герминальные мутации в генах BARD1, BRIP1, CHEK, MRE11A, MSH6, NBN, PALB2, RAD50, RAD51C и TP53, белковые продукты которых участвуют в восстановлении гомологичной рекомбинации. Мутации в средне- и низкопенетрантных генах в отдельности обуславливают минимальный риск, но благодаря мультипликативным и/или кумулятивным эффектам могут приводить к относительно высокому риску для носителей. В статье подробно описан онкогенез РЯ, обусловленного мутациями в генах MMR (синдром Линча), которые являются второй по значимости причиной наследственного РЯ и составляют от 2% до 15% всех случаев заболевания. Описаны также другие белки, кодируемые генами RAD51, RAD50, ATM, MRE11 и PALB2, которые взаимодействуют с продуктами генов BRCA1/2 в механизме гомологичной рекомбинационной репарации. A significant number of ovarian malignancies are hereditary (up to 30% of all neoplasms result from a high genetic predisposition). At least 16 candidate genes for hereditary ovarian cancer are known, and with the introduction and widespread use of genome-wide sequencing, an increasing number of genes and genetic variants are potentially involved in the pathogenesis of familial forms of the disease, although the contribution of these genes to the development of hereditary ovarian cancer . The detection of specific mutations in a number of ovarian cancer candidate genes in healthy women can justify not only more intensive and personalized surveillance programs, chemoprophylactic approaches and / or preventive operations, but also can provide fundamental knowledge about the pathogenesis of tumor development. This article describes the clinical features of hereditary ovarian cancer due to mutations in the key candidate genes (BRCA1, BRCA2, TP53, BARD1, CHEK2, RAD51, PALB2). The main types of mutations in BRCA - associated ovarian cancer, age-related features and survival rates are described. It was shown that the majority (approximately 65-85%) of hereditary ovarian tumors account for germline mutations in the BRCA1 and BRCA2 genes. It is believed that mutations in these genes lead primarily to gynecological oncological diseases in estrogen-sensitive target organs, however, according to some reports, there is also the likelihood of cancer of the stomach, colon, endometrium, pancreas, skin melanoma, bladder, head and neck tumors. Approximately 6% of hereditary OC is accounted for by germline mutations of the BARD1, BRIP1, CHEK, MRE11A, MSH6, NBN, PALB2, RAD50, RAD51C and TP53 genes, the protein products of which are involved in the restoration of homologous recombination. Medium and low penetrant genes individually carry only minimal risk, but due to the multiplicative and / or cumulative effects, they can cause a relatively high risk for carriers. The article describes in detail the oncogenesis of ovarian cancer due to a mutation in the genes for the restoration of the mismatch MMR - Lynch syndrome, which is the second leading cause of hereditary ovarian cancer and makes up from 2% to 15% of all cases of the disease. Other proteins encoded by the RAD51, RAD50, ATM, MRE11 and PALB2 genes that interact with the products of the BRCA1 / 2 genes in the mechanism of homologous recombination repair are also described in detail


2007 ◽  
Vol 25 (18_suppl) ◽  
pp. 5513-5513
Author(s):  
R. Boustany ◽  
P. Pautier ◽  
A. Rey ◽  
S. Delaloge ◽  
A. Chompret ◽  
...  

5513 Background: Germline mutations of the BRCA1 and BRCA2 genes account for the majority of hereditary breast-ovarian carcinomas. Nevertheless, in some patients with family history of ovarian cancers, neither point mutations nor genomic alterations are identified. Recently, IRIS gene, an open reading frame that extended from codon 1 of BRCA1 to a termination point in intron 11, has been identified. The encoded protein has been reported to play a role in controlling the replication origins firing (ROF) pathway. The study presented here aims at characterizing whether ROF-related proteins are differentially expressed among familial ovarian cancers associated with a germline BRCA1 or 2 mutation, familial ovarian cancers with wild-type BRCA1/2 genes, and sporadic ovarian cancers. Methods: Tumor samples from 72 patients with ovarian cancer and screened for BRCA1 and BRCA2 mutation because of family history of breast/ovarian cancer were collected. These cases were matched with 134 sporadic ovarian cancers (controls) according to age, year of diagnosis, tumor stage, histological subtype, and grade. The cases distributed among 26 BRCA1-linked (BRCA1*) tumors, 9 BRCA2-linked (BRCA2*) tumors and 37 with no identified mutation (BRCA1wt/BRCA2wt). Tissue micro-arrays were prepared from the paraffin blocks. P53, MCM3, MCM4, Geminin, PTTG and BRCA1-IRIS immuno-expression were scored with no information on the sample group as follows: the final score was the product of the positive cells percentage by the staining intensity, the final result being used as a continuous variable. Differences between cases and controls were tested by a Wilcoxon test for paired samples. Results: IRIS expression was significantly higher in familial cancers than in controls (P=0.002). When BRCA1/2 genes status was taken into account, differences remained significant when BRCA1wt/BRCA2wt tumors (P=0.04), but not when BRCA1* tumors were compared with controls. However, the latter showed significant higher expression of Geminin than controls (P=0.04). Conclusions: BRCA-1 IRIS protein is overaccumulated in ovarian cancers developed by patients with family history. Our results suggest IRIS may play a role in the development of ovarian cancers and could be related with an ovarian susceptibility. No significant financial relationships to disclose.


2012 ◽  
Vol 22 (3) ◽  
pp. 367-371 ◽  
Author(s):  
Jeffrey D. Seidman ◽  
Anna Yemelyanova ◽  
Jonathan A. Cosin ◽  
Anthony Smith ◽  
Robert J. Kurman

ObjectivePublished data are conflicting on the influence of cell type on prognosis in ovarian cancer. The recent separation of low-grade serous carcinoma as a distinctive cell type of ovarian cancer with an indolent behavior, in retrospect, suggests that survival in studies that have not separated this group may be inaccurate.MethodsAn unselected series of 262 International Federation of Gynecology and Obstetrics stage III ovarian carcinomas was studied. Diagnostic classification of each tumor was made with particular attention to recent refinements in cell-type classification. Survival curves were constructed according to Kaplan-Meier and compared with the log-rank test.ResultsThe 5-year survival for 207 high-grade serous carcinomas was 40%, as compared with 71% for 18 patients with low-grade serous carcinoma (P = 0.0113). Low-grade serous carcinoma was significantly more likely to be optimally debulked (P = 0.0039) and significantly less likely to be substage IIIC (P < 0.0001). The survival for carcinosarcoma was significantly inferior to all serous carcinomas (P = 0.0322). The significance of this latter comparison was lost when carcinosarcomas were compared with only high-grade serous carcinoma (P > 0.05).ConclusionsLow-grade serous carcinoma has a significantly better prognosis than high-grade serous carcinoma and also differs with regard to substage distribution and proportion of patients optimally debulked. Because of its excellent prognosis, failure to separate low-grade serous carcinomas, notwithstanding its infrequent occurrence, can change the results of survival analyses that do not make this separation.


2019 ◽  
Vol 65 (1) ◽  
pp. 56-62
Author(s):  
Alisa Villert ◽  
Larisa Kolomiets ◽  
Natalya Yunusova ◽  
Yevgeniya Fesik

High-grade ovarian carcinoma is a histopathological diagnosis, however, at the molecular level, ovarian cancer represents a heterogeneous group of diseases. Studies aimed at identifying molecular genetic subtypes of ovarian cancer are conducted in order to find the answer to the question: can different molecular subgroups influence the choice of treatment? One of the achievements in this trend is the recognition of the dualistic model that categorizes various types of ovarian cancer into two groups designated high-grade (HG) and low-grade (LG) tumors. However, the tumor genome sequencing data suggest the existence of 6 ovarian carcinoma subtypes, including two LG and four HG subtypes. Subtype C1 exhibits a high stromal response and the lowest survival. Subtypes C2 and C4 demonstrate higher number of intratumoral CD3 + cells, lower stromal gene expression and better survival than sybtype C1. Subtype C5 (mesenchymal) is characterized by mesenchymal cells, over-expression of N-cadherin and P-cadherin, low expression of differentiation markers, and lower survival rates than C2 and C4. The use of a consensus algorithm to determine the subtype allows identification of only a minority of ovarian carcinomas (approximately 25%) therefore, the practical importance of this classification requires additional research. There is evidence that it makes sense to randomize tumors into groups with altered expression of angiogenic genes and groups with overexpression of the immune response genes, as in the angiogenic group there is a comparative superiority in terms of survival. The administration of bevacizumab in the angiogenic group improves survival, while the administration of bevacizumab in the immune group even worsens the outcome. Molecular subtypes with worse survival rates (proliferative and mesenchymal) also benefit most from bevacizumab treatment. This review focuses on some of the advances in understanding molecular, cellular, and genetic changes in ovarian carcinomas with the results achieved so far regarding the formulation of molecular subtypes of ovarian cancer, however further studies are needed.


Sign in / Sign up

Export Citation Format

Share Document