1
|
Tsuchimochi S, Yamamoto Y, Taguchi A, Kawazu M, Sone K, Ikemura M, Tamai K, Kitamura S, Yoshimoto D, Fukaya S, Ishizaka A, Quynh AD, Nishijima A, Miyamoto Y, Mori M, Hiraike O, Hasegawa K, Ushiku T, Oda K, Hirota Y, Osuga Y. BRCA1 Promoter Methylation in Ovarian Cancer: Clinical Relevance and a Novel Diagnostic Approach Using Fragment Analysis. Cancer Sci 2025. [PMID: 40202107 DOI: 10.1111/cas.70078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 03/26/2025] [Accepted: 03/28/2025] [Indexed: 04/10/2025] Open
Abstract
Homologous recombination deficiency (HRD) tests, including MyChoice CDx, are companion diagnostics for poly (ADP-ribose) polymerase (PARP) inhibitors. BRCA1 promoter hypermethylation, a major HRD cause, may correlate with poorer prognosis. This study aimed to develop a simple, accurate method for detecting BRCA1 promoter hypermethylation and elucidate the characteristics of such cases. BRCA1 promoter methylation was analyzed using bisulfite sequencing (BIS-seq) in high-grade serous ovarian carcinoma specimens. We developed a newly developed BRCA1 methylation assay, BRCA1-Fragment Analysis of Methylation (BRCA1-FAM), which combines restriction enzyme digestion with fragment analysis. The accuracy of this assay was compared to the results of BIS-seq. We evaluated the relationship between BRCA1 promoter hypermethylation and prognosis and examined its association with BRCA1 expression and loss of heterozygosity. BRCA1 mutations and promoter methylation were mutually exclusive in the analyzed cases, with methylation observed in 28.9% (22/76) of primary debulking surgery cases. The BRCA1-FAM showed high sensitivity (91.3%) and specificity (100%) for detecting BRCA1 promoter hypermethylation, comparable to BIS-seq. Cases with BRCA1 promoter hypermethylation had significantly poorer progression-free survival (log-rank test, p = 0.048). Among these cases, 86.4% displayed abnormal BRCA1 immunostaining, with lower frequencies of BRCA1 loss of heterozygosity compared to those of other groups. BRCA1 promoter hypermethylation is associated with poor prognosis, underscoring the importance of its identification for HRD stratification. BRCA1-FAM is a simple and highly accurate method for evaluating BRCA1 promoter methylation. This approach may potentially enhance the precision of personalized therapies for ovarian cancer.
Collapse
Affiliation(s)
- Saki Tsuchimochi
- Department of Obstetrics and Gynecology, The University of Tokyo, Tokyo, Japan
| | - Yoko Yamamoto
- Department of Obstetrics and Gynecology, The University of Tokyo, Tokyo, Japan
| | - Ayumi Taguchi
- Department of Obstetrics and Gynecology, The University of Tokyo, Tokyo, Japan
| | - Masahito Kawazu
- Division of Cell Therapy, Chiba Cancer Center, Research Institute, Chiba, Japan
| | - Kenbun Sone
- Department of Obstetrics and Gynecology, The University of Tokyo, Tokyo, Japan
| | - Masako Ikemura
- Department of Pathology, The University of Tokyo, Tokyo, Japan
| | - Kana Tamai
- Department of Obstetrics and Gynecology, The University of Tokyo, Tokyo, Japan
| | - Shuhei Kitamura
- Department of Obstetrics and Gynecology, The University of Tokyo, Tokyo, Japan
| | - Daisuke Yoshimoto
- Department of Obstetrics and Gynecology, The University of Tokyo, Tokyo, Japan
| | - Sayuri Fukaya
- Department of Obstetrics and Gynecology, The University of Tokyo, Tokyo, Japan
| | - Aya Ishizaka
- Department of Obstetrics and Gynecology, The University of Tokyo, Tokyo, Japan
| | - Anh Duong Quynh
- Department of Obstetrics and Gynecology, The University of Tokyo, Tokyo, Japan
| | - Akira Nishijima
- Department of Obstetrics and Gynecology, The University of Tokyo, Tokyo, Japan
| | - Yuichiro Miyamoto
- Department of Obstetrics and Gynecology, The University of Tokyo, Tokyo, Japan
| | - Mayuyo Mori
- Department of Obstetrics and Gynecology, The University of Tokyo, Tokyo, Japan
| | - Osamu Hiraike
- Department of Obstetrics and Gynecology, The University of Tokyo, Tokyo, Japan
| | - Kosei Hasegawa
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Saitama, Japan
| | - Tetsuo Ushiku
- Department of Pathology, The University of Tokyo, Tokyo, Japan
| | - Katsutoshi Oda
- Department of Clinical Genomics, The University of Tokyo, Tokyo, Japan
| | - Yasushi Hirota
- Department of Obstetrics and Gynecology, The University of Tokyo, Tokyo, Japan
| | - Yutaka Osuga
- Department of Obstetrics and Gynecology, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
2
|
Lønning PE, Nikolaienko O, Knappskog S. Constitutional Epimutations: From Rare Events Toward Major Cancer Risk Factors? JCO Precis Oncol 2025; 9:e2400746. [PMID: 40179326 PMCID: PMC11995855 DOI: 10.1200/po-24-00746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 01/09/2025] [Accepted: 01/14/2025] [Indexed: 04/05/2025] Open
Abstract
Constitutional epimutations are epigenetic aberrations that arise in normal cells prenatally. Two major forms exist: secondary constitutional epimutations (SCEs), associated with cis-acting genetic aberrations, and primary constitutional epimutations (PCEs), for which no associated genetic aberrations were identified. Some SCEs have been associated with risk of cancer (MLH1 and MSH2 with colon or endometrial cancers, BRCA1 with familial breast and ovarian cancers), although such epimutations are rare, with a total of <100 cases reported. This contrasts recent findings for PCE, where low-level mosaic BRCA1 epimutations are recorded in 5%-10% of healthy females across all age groups, including newborns. BRCA1 PCEs predict an elevated risk of high-grade serous ovarian cancer and triple-negative breast cancer (TNBC) and are estimated to account for about 20% of all TNBCs. A similarly high population frequency is observed for mosaic constitutional epimutations in MGMT, occurring as PCE or SCE, but not in MLH1. Contrasting BRCA1 and MLH1, a potential association with cancer risk for MGMT epimutations is yet unclear. In this review, we provide a summary of findings linking constitutional epimutations to cancer risk with emphasis on PCE. We also highlight challenges in detection of PCE exemplified by low-level mosaic epimutations in BRCA1 and indicate the need for further studies, hypothesizing that improved knowledge about PCE may add significantly to our understanding of cancer risk, carcinogenesis, and potentially development of other diseases as well.
Collapse
Affiliation(s)
| | - Oleksii Nikolaienko
- Department of Oncology, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Stian Knappskog
- Department of Oncology, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Science, University of Bergen, Bergen, Norway
| |
Collapse
|
3
|
Karimnia N, Wilson AL, Doran BR, Do J, Matthews A, Ho GY, Plebanski M, Jobling TW, Stephens AN, Bilandzic M. A Novel 3D High-Throughput Phenotypic Drug Screening Pipeline to Identify Drugs with Repurposing Potential for the Treatment of Ovarian Cancer. Adv Healthc Mater 2025; 14:e2404117. [PMID: 40109101 PMCID: PMC12023816 DOI: 10.1002/adhm.202404117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 02/24/2025] [Indexed: 03/22/2025]
Abstract
Ovarian cancer (OC) poses a significant clinical challenge due to its high recurrence rates and resistance to standard therapies, particularly in advanced stages where recurrence is common, and treatment is predominantly palliative. Personalized treatments, while effective in other cancers, remain underutilized in OC due to a lack of reliable biomarkers predicting clinical outcomes. Accordingly, precision medicine approaches are limited, with PARP inhibitors showing efficacy only in specific genetic contexts. Drug repurposing offers a promising, rapidly translatable strategy by leveraging existing pharmacological data to identify new treatments for OC. Patient-derived polyclonal spheroids, isolated from ascites fluid closely mimic the clinical behavior of OC, providing a valuable model for drug testing. Using these spheroids, a high-throughput drug screening pipeline capable of evaluating both cytotoxicity and anti-migratory properties of a diverse drug library, including FDA-approved, investigational, and newly approved compounds is developed. The findings highlight the importance of 3D culture systems, revealing a poor correlation between drug efficacy in traditional 2D models and more clinically relevant 3D spheroids. This approach has expedited the identification of promising candidates, such as rapamycin, which demonstrated limited activity as a monotherapy but synergized effectively with standard treatments like cisplatin and paclitaxel in vitro. In combination with platinum-based therapy, Rapamycin led to significant in vitro cytotoxicity and a marked reduction in tumor burden in a syngeneic in vivo model. This proof-of-concept study underscores the potential of drug repurposing to rapidly advance new treatments into clinical trials for OC, offering renewed hope for patients with advanced disease.
Collapse
Affiliation(s)
- Nazanin Karimnia
- Hudson Institute of Medical Research27–31 Wright StClaytonVIC3168Australia
- Department of Molecular and Translational SciencesMonash University45 Kanooka GroveClaytonVIC3168Australia
| | - Amy L. Wilson
- Hudson Institute of Medical Research27–31 Wright StClaytonVIC3168Australia
- Department of Molecular and Translational SciencesMonash University45 Kanooka GroveClaytonVIC3168Australia
| | - Brittany R. Doran
- Hudson Institute of Medical Research27–31 Wright StClaytonVIC3168Australia
- Department of Molecular and Translational SciencesMonash University45 Kanooka GroveClaytonVIC3168Australia
| | - Jennie Do
- Hudson Institute of Medical Research27–31 Wright StClaytonVIC3168Australia
- Department of Molecular and Translational SciencesMonash University45 Kanooka GroveClaytonVIC3168Australia
| | - Amelia Matthews
- Hudson Institute of Medical Research27–31 Wright StClaytonVIC3168Australia
- Department of Molecular and Translational SciencesMonash University45 Kanooka GroveClaytonVIC3168Australia
| | - Gwo Y. Ho
- School of Clinical SciencesMonash University27 Rainforest WalkClaytonVIC3168Australia
- Department of OncologyMonash Health823–865 Centre RdBentleighVIC3165Australia
| | - Magdalena Plebanski
- School of Health and Biomedical SciencesRMIT UniversityBundoora campus West203, 1/2 Plenty RdBundooraVIC3083Australia
| | - Thomas W. Jobling
- Monash Medical CentreDepartment of Gynecology OncologyMonash Health823–865 Centre RdBentleigh EastVIC3165Australia
| | - Andrew N. Stephens
- Hudson Institute of Medical Research27–31 Wright StClaytonVIC3168Australia
- Department of Molecular and Translational SciencesMonash University45 Kanooka GroveClaytonVIC3168Australia
| | - Maree Bilandzic
- Hudson Institute of Medical Research27–31 Wright StClaytonVIC3168Australia
- Department of Molecular and Translational SciencesMonash University45 Kanooka GroveClaytonVIC3168Australia
| |
Collapse
|
4
|
Cui HT, Zhu QY, Zhao HW, Liu HL, Wang N. Pregnancy is associated with the prognosis of ovarian cancer patients with abdominal metastasis. Am J Cancer Res 2025; 15:168-181. [PMID: 39949936 PMCID: PMC11815367 DOI: 10.62347/jujq9225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 12/06/2024] [Indexed: 02/16/2025] Open
Abstract
This study aims to explore a new approach to reduce the recurrence risk and improve the prognosis of ovarian cancer (OC) patients with abdominal metastasis by analyzing the clinical characteristics and prognostic factors. A total of 292 OC patients with abdominal metastasis, treated at Henan Provincial People's Hospital between 2021 and 2023 were included in this retrospective study. Follow-up was conducted for one year to observe the recurrence, with 285 patients completing the observation. The patients were then categorized into relapsing and non-relapsing groups based on whether they experienced a relapse within one-year follow-up. Independent sample t-tests and χ 2 tests were used for inter-group comparison. Both univariate and multivariate logistic regression analyses were utilized to screen factors affecting recurrence. The variance inflation factor (VIF) was used to analyze whether the variables in the model had multicollinearity. Receiver Operating Characteristic (ROC) curves and nomographs were used to construct models for predicting one-year recurrence in OC patients with abdominal metastasis. Area under curve (AUC) of ROC and Hosmer-Lemeshow goodness of fit test were used to evaluate the accuracy of the model. The prediction model was verified by internal verification and external verification. The number of pregnancies, the number of births, diabetes mellitus, tumor diameter, tumor reduction combined with intraperitoneal chemotherapy, CA-125, HE-4, NLR, PLR, MLR showed association with patient recurrence. Logistic regression analysis revealed that lower pregnancy frequency and elevated levels of CA-125, HE-4, PLR and MLR were independent risk factors for increased risk of recurrence. In addition, the nomogram-based model demonstrated strong predictive accuracy for one-year recurrence. OC patients with abdominal metastasis present diverse clinical manifestations, among which fewer pregnancies and elevated levels of CA-125, HE-4, PLR, and MLR may be independent risk factors for increased risk of recurrence. Individualized interventions based on these prognostic factors are essential to reduce risk and enhance patient quality of life.
Collapse
Affiliation(s)
- Hai-Tao Cui
- Department of Gynecology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University Zhengzhou 450000, Henan, China
| | - Qian-Yong Zhu
- Department of Gynecology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University Zhengzhou 450000, Henan, China
| | - Hong-Wei Zhao
- Department of Gynecology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University Zhengzhou 450000, Henan, China
| | - Hui-Li Liu
- Department of Gynecology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University Zhengzhou 450000, Henan, China
| | - Na Wang
- Department of Gynecology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University Zhengzhou 450000, Henan, China
| |
Collapse
|
5
|
Barnicle A, Ray-Coquard I, Rouleau E, Cadoo K, Simpkins F, Aghajanian C, Leary A, Poveda A, Lheureux S, Pujade-Lauraine E, You B, Ledermann J, Matulonis U, Gourley C, Timms KM, Lai Z, Hodgson DR, Elks CE, Dearden S, Egile C, Lao-Sirieix P, Harrington EA, Brown JS. Patterns of genomic instability in > 2000 patients with ovarian cancer across six clinical trials evaluating olaparib. Genome Med 2024; 16:145. [PMID: 39695768 PMCID: PMC11657106 DOI: 10.1186/s13073-024-01413-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 11/12/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND The introduction of poly(ADP-ribose) polymerase (PARP) inhibitors represented a paradigm shift in the treatment of ovarian cancer. Genomic data from patients with high-grade ovarian cancer in six phase II/III trials involving the PARP inhibitor olaparib were analyzed to better understand patterns and potential causes of genomic instability. PATIENTS AND METHODS Homologous recombination deficiency (HRD) was assessed in 2147 tumor samples from SOLO1, PAOLA-1, Study 19, SOLO2, OPINION, and LIGHT using next-generation sequencing technology. Genomic instability scores (GIS) were assessed in BRCA1 and/or BRCA2 (BRCA)-mutated (BRCAm), non-BRCA homologous recombination repair-mutated (non-BRCA HRRm), and non-HRRm tumors. RESULTS BRCAm was identified in 1021/2147 (47.6%) tumors. BRCAm tumors had significantly higher GIS than non-BRCAm tumors (P < 0.001) and high biallelic loss (815/838; 97.3%) regardless of germline (658/672; 97.9%) or somatic (101/108; 93.5%) BRCAm status. In non-BRCA HRRm tumors (n = 121) a similar proportion were HRD-positive (GIS ≥ 42: 55/121; 45.5%) relative to HRD-negative (GIS < 42: 52/121; 43.0%). GIS was highly variable in non-BRCA HRRm (median 42 [interquartile range (IQR) 29-58]) and non-HRRm (n = 1005; median 32 [IQR 20-55]) tumors. Gene mutations with high GIS included HRR genes BRIP1 (median 46 [IQR 41-58]), RAD51C (median 58 [IQR 48-66]), RAD51D (median 62 [IQR 54-69]), and PALB2 (median 64 [IQR 58-74]), and non-HRR genes NF1 (median 49 [IQR 25-60]) and RB1 (median 55 [IQR 30-71]). CCNE1-amplified and PIK3CA-mutated tumors had low GIS (CCNE1-amplified: median 24 [IQR 18-29]; PIK3CA-mutated: median 32 [IQR 14-52]) and were predominantly non-BRCAm. CONCLUSIONS These analyses provide valuable insight into patterns of genomic instability and potential drivers of HRD, besides BRCAm, in ovarian cancer and will help guide future research into the potential clinical effectiveness of anti-cancer treatments in ovarian cancer, including PARP inhibitors as well as other precision oncology agents. TRIAL REGISTRATION The SOLO1 trial was registered at ClinicalTrials.gov (NCT01844986) on April 30, 2013; the PAOLA-1 trial was registered at ClinicalTrials.gov (NCT02477644) on June 18, 2015 (retrospectively registered); Study 19 was registered at ClinicalTrials.gov (NCT00753545) on September 12, 2008 (retrospectively registered); the SOLO2 trial was registered at ClinicalTrials.gov (NCT01874353) on June 7, 2013; the OPINION trial was registered at ClinicalTrials.gov (NCT03402841) on January 3, 2018; the LIGHT trial was registered at ClinicalTrials.gov (NCT02983799) on November 4, 2016.
Collapse
Affiliation(s)
- Alan Barnicle
- Translational Medicine, Oncology R&D, AstraZeneca, Cambridge Biomedical Campus, 1 Francis Crick Avenue, Cambridge, CB2 0AA, UK.
| | - Isabelle Ray-Coquard
- Medical Oncology Department, Centre Léon Bérard and University Claude Bernard Lyon, and Groupe d'Investigateurs Nationaux pour l'Etude des Cancers Ovariens (GINECO), Lyon, France
| | - Etienne Rouleau
- Department of Medical Biology and Pathology, Gustave Roussy, Cancer Genetics Laboratory, Gustave Roussy, Villejuif, France
| | - Karen Cadoo
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Fiona Simpkins
- Department of Obstetrics and Gynecology, Jordan Center for Gynecologic Oncology at the Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | | | | | - Andrés Poveda
- Initia Oncology, Hospital Quironsalud, Valencia, Spain
| | - Stephanie Lheureux
- Department of Medical Oncology, Princess Margaret Hospital, Toronto, ONT, Canada
| | - Eric Pujade-Lauraine
- Association de Recherche Cancers Gynécologiques (ARCAGY)-GINECO, and GINECO, Paris, France
| | - Benoit You
- Medical Oncology, IC-HCL, EPSILYON, Univ Lyon 1, and GINECO, Lyon, France
| | - Jonathan Ledermann
- UCL Cancer Institute, University College London and UCL Hospitals, London, UK
| | | | - Charlie Gourley
- Cancer Research UK Scotland Centre, University of Edinburgh, Edinburgh, UK
| | | | - Zhongwu Lai
- Translational Medicine, Oncology R&D, Research and Early Development, AstraZeneca, Boston, MA, USA
| | - Darren R Hodgson
- Translational Medicine, Oncology R&D, Research and Early Development, AstraZeneca, Boston, MA, USA
| | - Cathy E Elks
- Precision Medicine and Biosamples, Oncology R&D, AstraZeneca, Cambridge, UK
| | - Simon Dearden
- Precision Medicine and Biosamples, Oncology R&D, AstraZeneca, Cambridge, UK
| | - Coumaran Egile
- Precision Medicine and Biosamples, Oncology R&D, AstraZeneca, Cambridge, UK
| | - Pierre Lao-Sirieix
- Precision Medicine and Biosamples, Oncology R&D, AstraZeneca, Cambridge, UK
| | - Elizabeth A Harrington
- Translational Medicine, Oncology R&D, AstraZeneca, Cambridge Biomedical Campus, 1 Francis Crick Avenue, Cambridge, CB2 0AA, UK
| | - Jessica S Brown
- Translational Medicine, Oncology R&D, AstraZeneca, Cambridge Biomedical Campus, 1 Francis Crick Avenue, Cambridge, CB2 0AA, UK
| |
Collapse
|
6
|
Lønning PE, Nikolaienko O, Knappskog S. Constitutional BRCA1 Epimutations: A Key for Understanding Basal-Like Breast and High-Grade Serous Ovarian Cancer. Hum Mutat 2024; 2024:7353984. [PMID: 40225940 PMCID: PMC11919088 DOI: 10.1155/2024/7353984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 08/08/2024] [Indexed: 04/15/2025]
Abstract
Germline pathogenic genetic variants in the BRCA1 and BRCA2 genes are the most frequent causes of familial breast and ovarian cancer. Contrasting BRCA2, epimutations in the BRCA1 gene are frequently detected in tissue from triple-negative breast (TNBC) and high-grade serous ovarian cancers (HGSOC). While studies over the last decade have reported BRCA1 epimutations in white blood cells (WBC) from breast and ovarian cancer patients, the potential hazard ratio for incident TNBC and HGSOC was not formally assessed until recently. Conducting a prospective nested case-control study on women participating in the American Women's Health Initiative Study, we provided firm evidence that mosaic WBC BRCA1 epimutations, even at allele frequencies < 0.1%, are associated with a significantly increased risk of both incident HGSOC and TNBC > 5 years after WBC collection. In a second study assessing BRCA1 epimutations in WBC and matched tumor samples from TNBC, our results indicated such epimutations to be the underlying cause of around 20% of TNBC, far exceeding the percentage of cases carrying BRCA1 germline pathogenic genetic variants. We detected primary constitutional BRCA1 epimutations in tissues derived from all three germ layers. They occur independently of BRCA1 promoter haplotypes but are present on the same allele in all WBC within affected individuals. Moreover, epimutations are consistently found on the same allele in normal and tumor breast tissue as well as in WBC. This finding, together with BRCA1 epimutations detected in WBC from newborns, strongly indicates an early embryonic event with clonal expansion affecting all germ layers. Future work in the field must lead to an understanding of exactly when and how the BRCA1 epimutations occur and, most importantly, whether primary constitutional epimutations in genes other than BRCA1 may cause an elevated risk of other cancer types.
Collapse
Affiliation(s)
- Per E. Lønning
- K.G. Jebsen Center for Genome-Directed Cancer TherapyDepartment of OncologyHaukeland University Hospital, Bergen, Norway
| | - Oleksii Nikolaienko
- K.G. Jebsen Center for Genome-Directed Cancer TherapyDepartment of OncologyHaukeland University Hospital, Bergen, Norway
- Department of Clinical ScienceUniversity of Bergen, Bergen, Norway
| | - Stian Knappskog
- K.G. Jebsen Center for Genome-Directed Cancer TherapyDepartment of OncologyHaukeland University Hospital, Bergen, Norway
- Department of Clinical ScienceUniversity of Bergen, Bergen, Norway
| |
Collapse
|
7
|
Zhao Y, Yuan H, Chen Y, Yao H, Li N, Wu L, Yuan G. Outcomes of secondary cytoreductive surgery in patients with platinum-sensitive recurrent ovarian cancer progressed after prior poly (adenosine diphosphate-ribose) polymerase inhibitors: A retrospective cohort study. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2024; 50:108383. [PMID: 38704898 DOI: 10.1016/j.ejso.2024.108383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 04/22/2024] [Accepted: 04/30/2024] [Indexed: 05/07/2024]
Abstract
OBJECTIVE To evaluate the impact of previous poly (adenosine diphosphate-ribose) polymerase (PARP) inhibitor therapy on the effectiveness of secondary cytoreductive surgery (SCS) in patients with platinum-sensitive recurrent ovarian cancer (PSROC). METHODS We identified patients with PSROC who underwent SCS at the Cancer Hospital, Chinese Academy of Medical Science, between January 2010 and December 2022. Postoperative complications within 30 days were categorized using the Accordion Severity Grading System. The Kaplan‒Meier method was used to estimate both overall survival (OS) and progression-free survival (PFS), and multivariate analysis was used to identify independent prognostic factors. RESULTS Of the 265 patients included, 39 received prior PARP inhibitor therapy (Group A), and 226 did not (Group B). The rates of complete resection after SCS did not significantly differ between the two groups (79.5 % for Group A vs. 81.0 % for Group B; p = 0.766). As of December 2023, Group A exhibited a significantly shorter median PFS (14.2 months) than Group B (22.5 months; p = 0.002). Furthermore, the 3-year OS rate was lower in Group A (72.5 %) than in Group B (82.7 %; p = 0.015). The incidence of severe postoperative complications was comparable between Groups A and B (7.7 % vs. 1.8 %; p = 0.061). Multivariate analysis revealed that prior PARP inhibitor therapy significantly reduced the median PFS (hazard ratio (HR) = 4.434; p = 0.021) and OS (HR = 2.076; p = 0.010). CONCLUSIONS SCS for PSROC demonstrated reduced efficacy in patients previously treated with PARP inhibitors compared to those without prior PARP inhibitor treatment.
Collapse
Affiliation(s)
- Yuxi Zhao
- Department of Gynecologic Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Hua Yuan
- Department of Gynecologic Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yiran Chen
- Department of Gynecologic Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Hongwen Yao
- Department of Gynecologic Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Ning Li
- Department of Gynecologic Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Lingying Wu
- Department of Gynecologic Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Guangwen Yuan
- Department of Gynecologic Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
8
|
He W, Zhu H, Zhang S, Shu G, Lei H, Yin G, Ni X, Wang M, Wu Q. Promoter Methylation Changes in DNA Damage-Response Genes in Ovarian Cancer and Their Correlation with Prognosis. CLIN EXP OBSTET GYN 2024; 51. [DOI: 10.31083/j.ceog5105109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2024]
Abstract
Background: Ovarian cancer has a poor prognosis, and DNA damage-response (DDR) genes are associated with both its occurrence and prognosis. However, previous studies have mostly focused on genetic mutations, with no clear conclusions on epigenetic factors such as DNA methylation. Methods: In this study, we comprehensively investigated the relationship between promoter methylation of DDR genes and ovarian cancer prognosis. We performed combined multidata analysis of the promoter methylation, expression, homologous recombination defieiency (HRD) score, and drug sensitivity of 377 DDR genes in ovarian cancer by utilizing The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) datasets. We then validated abnormal promoter methylation and its relationship with overall survival in clinical samples. Results: Our analysis identified 52 methylation-driven DDR genes that exhibited abnormal expression due to abnormal promoter methylation. These genes are mostly related to BRCA1-related DNA damage repair and cell cycle regulatory pathways. Further studies revealed six of these genes, BRCA1, PTTG1, TTK, AURKA, CDC6, and E2F1, to be significantly associated with HRD scores. Among them, E2F1, PTTG1, and CDC6 are associated with drug sensitivity. Finally, we verified in 81 ovarian cancer samples that methylation of the promoter of these three genes was significantly associated with patient survival. Conclusions: Our study identified a large number of methylation-driven aberrantly expressed DDR genes in ovarian cancer, some of which affect disease prognosis. Levels of methylation of these gene promoters may serve as potential prognostic markers.
Collapse
Affiliation(s)
- Wanhong He
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, 200237 Shanghai, China
| | - Haijun Zhu
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, 200237 Shanghai, China
| | - Sufen Zhang
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, 200237 Shanghai, China
| | - Guang Shu
- Department of Pathology, Xiangya Hospital, School of Basic Medical Sciences, Central South University, 410013 Changsha, Hunan, China
| | - Han Lei
- Department of Pathology, Xiangya Hospital, School of Basic Medical Sciences, Central South University, 410013 Changsha, Hunan, China
| | - Gang Yin
- Department of Pathology, Xiangya Hospital, School of Basic Medical Sciences, Central South University, 410013 Changsha, Hunan, China
| | - Xiaohua Ni
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, 200237 Shanghai, China
| | - Maonan Wang
- Department of Pathology, Xiangya Hospital, School of Basic Medical Sciences, Central South University, 410013 Changsha, Hunan, China
| | - Qihan Wu
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, 200237 Shanghai, China
| |
Collapse
|
9
|
Chen C, Zhu Y, Zhang H, Xiao L. Prognostic Effects of RASSF1A, BRCA1, APC, and p16 Promoter Methylation in Ovarian Cancer: A Meta-Analysis. Gynecol Obstet Invest 2024; 89:363-375. [PMID: 38615670 DOI: 10.1159/000538673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 03/25/2024] [Indexed: 04/16/2024]
Abstract
INTRODUCTION DNA methylation plays an important role in the carcinogenesis, progression, and prognosis of various human cancers. RASSF1A, BRCA1, APC, and p16 are the frequently methylated genes among patients with ovarian cancer. Therefore, our study aimed to better determine the prognostic and cancer characteristics effects of RASSF1A, BRCA1, APC, and p16 promoter methylation in ovarian cancer patients. METHODS Databases such as PubMed, Web of Science, EMBASE, CNKI, and WanFang were searched for published studies up to March 4, 2024. The outcomes are shown as OR and HR with their 95% CIs. Then, the random or fixed-effect model was performed to evaluate the effect sizes. RESULTS Finally, 27 articles were included in this meta-analysis. No significant relationships were observed between RASSF1A, BRCA1, and APC promoter methylation and the clinical prognostic (including overall survival and progression-free survival) and cancer characteristics (including ascites, lymph node metastasis, and pelvic peritoneal metastasis) in ovarian cancer. p16 promoter methylation was significantly related to poor progression-free survival (PFS) (HR = 1.52, 95% CI = 1.14-2.04) and overall survival (OS) (HR = 1.39, 95% CI = 1.06, to 1.83) in univariate and poor PFS in multivariate Cox regression models (HR = 1.42, 95% CI = 1.05-1.92). Besides, our results indicated that the clinical stage was associated with inferior OS while there was no significant association between tumor grade and OS. CONCLUSION RASSF1A, BRCA1, and APC promoter methylation were not significantly associated with clinical prognostic and cancer characteristics. p16 may be a useful biomarker for predicting PFS in ovarian cancer. Furthermore, the clinical stage was significantly associated with OS. In further research, more prospective and multicenter validation studies remain needed.
Collapse
Affiliation(s)
- Cheng Chen
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ying Zhu
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Haibo Zhang
- Central Laboratory, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Lan Xiao
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
10
|
Geissler F, Nesic K, Kondrashova O, Dobrovic A, Swisher EM, Scott CL, J. Wakefield M. The role of aberrant DNA methylation in cancer initiation and clinical impacts. Ther Adv Med Oncol 2024; 16:17588359231220511. [PMID: 38293277 PMCID: PMC10826407 DOI: 10.1177/17588359231220511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 11/21/2023] [Indexed: 02/01/2024] Open
Abstract
Epigenetic alterations, including aberrant DNA methylation, are now recognized as bone fide hallmarks of cancer, which can contribute to cancer initiation, progression, therapy responses and therapy resistance. Methylation of gene promoters can have a range of impacts on cancer risk, clinical stratification and therapeutic outcomes. We provide several important examples of genes, which can be silenced or activated by promoter methylation and highlight their clinical implications. These include the mismatch DNA repair genes MLH1 and MSH2, homologous recombination DNA repair genes BRCA1 and RAD51C, the TERT oncogene and genes within the P15/P16/RB1/E2F tumour suppressor axis. We also discuss how these methylation changes might occur in the first place - whether in the context of the CpG island methylator phenotype or constitutional DNA methylation. The choice of assay used to measure methylation can have a significant impact on interpretation of methylation states, and some examples where this can influence clinical decision-making are presented. Aberrant DNA methylation patterns in circulating tumour DNA (ctDNA) are also showing great promise in the context of non-invasive cancer detection and monitoring using liquid biopsies; however, caution must be taken in interpreting these results in cases where constitutional methylation may be present. Thus, this review aims to provide researchers and clinicians with a comprehensive summary of this broad, but important subject, illustrating the potentials and pitfalls of assessing aberrant DNA methylation in cancer.
Collapse
Affiliation(s)
- Franziska Geissler
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Ksenija Nesic
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Olga Kondrashova
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Alexander Dobrovic
- University of Melbourne Department of Surgery, Austin Health, Heidelberg, VIC, Australia
| | | | - Clare L. Scott
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, VIC, Australia
- Peter MacCallum Cancer Centre and Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
- Royal Women’s Hospital, Parkville, VIC, Australia
- Royal Melbourne Hospital, Parkville, VIC, Australia
| | - Matthew J. Wakefield
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
11
|
Wilczyński J, Paradowska E, Wilczyński M. High-Grade Serous Ovarian Cancer-A Risk Factor Puzzle and Screening Fugitive. Biomedicines 2024; 12:229. [PMID: 38275400 PMCID: PMC10813374 DOI: 10.3390/biomedicines12010229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 01/14/2024] [Accepted: 01/17/2024] [Indexed: 01/27/2024] Open
Abstract
High-grade serous ovarian cancer (HGSOC) is the most lethal tumor of the female genital tract. Despite extensive studies and the identification of some precursor lesions like serous tubal intraepithelial cancer (STIC) or the deviated mutational status of the patients (BRCA germinal mutation), the pathophysiology of HGSOC and the existence of particular risk factors is still a puzzle. Moreover, a lack of screening programs results in delayed diagnosis, which is accompanied by a secondary chemo-resistance of the tumor and usually results in a high recurrence rate after the primary therapy. Therefore, there is an urgent need to identify the substantial risk factors for both predisposed and low-risk populations of women, as well as to create an economically and clinically justified screening program. This paper reviews the classic and novel risk factors for HGSOC and methods of diagnosis and prediction, including serum biomarkers, the liquid biopsy of circulating tumor cells or circulating tumor DNA, epigenetic markers, exosomes, and genomic and proteomic biomarkers. The novel future complex approach to ovarian cancer diagnosis should be devised based on these findings, and the general outcome of such an approach is proposed and discussed in the paper.
Collapse
Affiliation(s)
- Jacek Wilczyński
- Department of Gynecological Surgery and Gynecological Oncology, Medical University of Lodz, 4 Kosciuszki Str., 90-419 Lodz, Poland
| | - Edyta Paradowska
- Laboratory of Virology, Institute of Medical Biology of the Polish Academy of Sciences, 106 Lodowa Str., 93-232 Lodz, Poland;
| | - Miłosz Wilczyński
- Department of Surgical, Endoscopic and Gynecological Oncology, Polish Mother’s Health Center—Research Institute, 281/289 Rzgowska Str., 93-338 Lodz, Poland;
- Department of Surgical and Endoscopic Gynecology, Medical University of Lodz, 4 Kosciuszki Str., 90-419 Lodz, Poland
| |
Collapse
|
12
|
Incorvaia L, Perez A, Marchetti C, Brando C, Gristina V, Cancelliere D, Pivetti A, Contino S, Di Giovanni E, Barraco N, Bono M, Giurintano A, Bazan Russo TD, Gottardo A, Cutaia S, Pedone E, Peri M, Corsini LR, Fanale D, Galvano A, Scambia G, Badalamenti G, Russo A, Bazan V. Theranostic biomarkers and PARP-inhibitors effectiveness in patients with non-BRCA associated homologous recombination deficient tumors: Still looking through a dirty glass window? Cancer Treat Rev 2023; 121:102650. [PMID: 37939446 DOI: 10.1016/j.ctrv.2023.102650] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/16/2023] [Accepted: 10/30/2023] [Indexed: 11/10/2023]
Abstract
Breast cancer susceptibility gene 1 (BRCA1) and breast cancer susceptibility gene 2 (BRCA2) deleterious variants were the first and, still today, the main biomarkers of poly(ADP)ribose polymerase (PARP)-inhibitors (PARPis) benefit. The recent, increased, numbers of individuals referred for counseling and multigene panel testing, and the remarkable expansion of approved PARPis, not restricted to BRCA1/BRCA2-Pathogenic Variants (PVs), produced a strong clinical need for non-BRCA biomarkers. Significant limitations of the current testing and assays exist. The different approaches that identify the causes of Homologous Recombination Deficiency (HRD), such as the germline and somatic Homologous Recombination Repair (HRR) gene PVs, the testing showing its consequences, such as the genomic scars, or the novel functional assays such as the RAD51 foci testing, are not interchangeable, and should not be considered as substitutes for each other in clinical practice for guiding use of PARPi in non-BRCA, HRD-associated tumors. Today, the deeper knowledge on the significant relationship among all proteins involved in the HRR, not limited to BRCA, expands the possibility of a successful non-BRCA, HRD-PARPi synthetic lethality and, at the same time, reinforces the need for enhanced definition of HRD biomarkers predicting the magnitude of PARPi benefit.
Collapse
Affiliation(s)
- Lorena Incorvaia
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Alessandro Perez
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Claudia Marchetti
- Department of Woman's and Child Health and Public Health Sciences, Gynecologic Oncology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy; Catholic University of the Sacred Heart, Rome, Italy
| | - Chiara Brando
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Valerio Gristina
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Daniela Cancelliere
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Alessia Pivetti
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Silvia Contino
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Emilia Di Giovanni
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Nadia Barraco
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Marco Bono
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Ambra Giurintano
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Tancredi Didier Bazan Russo
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Andrea Gottardo
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Sofia Cutaia
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Erika Pedone
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Marta Peri
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Lidia Rita Corsini
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Daniele Fanale
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Antonio Galvano
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Giovanni Scambia
- Department of Woman's and Child Health and Public Health Sciences, Gynecologic Oncology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy; Catholic University of the Sacred Heart, Rome, Italy
| | - Giuseppe Badalamenti
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Antonio Russo
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy.
| | - Viviana Bazan
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| |
Collapse
|
13
|
Zhunussova G, Omarbayeva N, Kaidarova D, Abdikerim S, Mit N, Kisselev I, Yergali K, Zhunussova A, Goncharova T, Abdrakhmanova A, Djansugurova L. Determination of genetic predisposition to early breast cancer in women of Kazakh ethnicity. Oncotarget 2023; 14:860-877. [PMID: 37791908 PMCID: PMC10549772 DOI: 10.18632/oncotarget.28518] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 09/21/2023] [Indexed: 10/05/2023] Open
Abstract
Breast cancer (BC) is the most common type of cancer among women in Kazakhstan. To date, little data are available on the spectrum of genetic variation in Kazakh women with BC. We aimed to identify population-specific genetic markers associated with the risk of developing early-onset BC and test their association with clinical and prognostic factors. The study included 224 Kazakh women diagnosed with BC (≤40 age). Entire coding regions (>1700 exons) and the flanking noncoding regions of 94 cancer-associated genes were sequenced from blood DNA using MiSeq platform. We identified 38 unique pathogenic variants (PVs) in 13 different cancer-predisposing genes among 57 patients (25.4%), of which 6 variants were novel. In total, 12 of the 38 distinct PVs were detected recurrently, including BRCA1 c.5266dup, c.5278-2del, and c.2T>C, and BRCA2 c.9409dup and c.9253del that may be founder in this population. BRCA1 carriers were significantly more likely to develop triple-negative BC (OR = 6.61, 95% CI 2.44-17.91, p = 0.0002) and have family history of BC (OR = 3.17, 95% CI 1.14-8.76, p = 0.03) compared to non-carriers. This study allowed the identification of PVs specific to early-onset BC, which may be used as a foundation to develop regional expertise and diagnostic tools for early detection of BC in young Kazakh women.
Collapse
Affiliation(s)
- Gulnur Zhunussova
- Laboratory of Molecular Genetics, Institute of Genetics and Physiology, Almaty 050060, Kazakhstan
- Al-Farabi Kazakh National University, Almaty 050060, Kazakhstan
| | - Nazgul Omarbayeva
- Kazakh Institute of Oncology and Radiology, Almaty 050060, Kazakhstan
- Asfendiyarov Kazakh National Medical University, Almaty 050060, Kazakhstan
| | - Dilyara Kaidarova
- Kazakh Institute of Oncology and Radiology, Almaty 050060, Kazakhstan
- Asfendiyarov Kazakh National Medical University, Almaty 050060, Kazakhstan
| | - Saltanat Abdikerim
- Laboratory of Molecular Genetics, Institute of Genetics and Physiology, Almaty 050060, Kazakhstan
- Al-Farabi Kazakh National University, Almaty 050060, Kazakhstan
| | - Natalya Mit
- Laboratory of Molecular Genetics, Institute of Genetics and Physiology, Almaty 050060, Kazakhstan
| | - Ilya Kisselev
- Laboratory of Molecular Genetics, Institute of Genetics and Physiology, Almaty 050060, Kazakhstan
| | - Kanagat Yergali
- Laboratory of Molecular Genetics, Institute of Genetics and Physiology, Almaty 050060, Kazakhstan
| | - Aigul Zhunussova
- Laboratory of Molecular Genetics, Institute of Genetics and Physiology, Almaty 050060, Kazakhstan
| | | | - Aliya Abdrakhmanova
- Kazakh Institute of Oncology and Radiology, Almaty 050060, Kazakhstan
- Asfendiyarov Kazakh National Medical University, Almaty 050060, Kazakhstan
| | - Leyla Djansugurova
- Laboratory of Molecular Genetics, Institute of Genetics and Physiology, Almaty 050060, Kazakhstan
- Al-Farabi Kazakh National University, Almaty 050060, Kazakhstan
| |
Collapse
|
14
|
Blanc-Durand F, Tang R, Pommier M, Nashvi M, Cotteret S, Genestie C, Le Formal A, Pautier P, Michels J, Kfoury M, Hervé R, Mengue S, Wafo E, Elies A, Miailhe G, Uzan J, Rouleau E, Leary A. Clinical Relevance of BRCA1 Promoter Methylation Testing in Patients with Ovarian Cancer. Clin Cancer Res 2023; 29:3124-3129. [PMID: 37067532 DOI: 10.1158/1078-0432.ccr-22-3328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/12/2022] [Accepted: 04/13/2023] [Indexed: 04/18/2023]
Abstract
PURPOSE Homologous recombination deficiency (HRD) is closely related to PARP inhibitor (PARPi) benefit in ovarian cancer. The capacity of BRCA1 promoter methylation to predict prognosis and HRD status remains unclear. We aimed to correlate BRCA1 promoter methylation levels in patients with high-grade ovarian cancer to HRD status and clinical behavior to assess its clinical relevance. EXPERIMENTAL DESIGN This is a retrospective monocentric analysis of patients centrally tested for genomic instability score (GIS) by MyChoice CDx (Myriad Genetics). The detection of BRCA1 promoter methylation and quantification of methylation levels were performed by quantitative droplet digital PCR methodology. High BRCA1 methylation was defined as ≥70% and deemed to be associated with homozygous silencing. RESULTS Of 100 patients, 11% harbored a deleterious BRCA1/2 mutation. GIS was considered positive (score ≥ 42) for 52 patients and negative for 48 patients. Using a 70% cutoff, 19% (15/79) of BRCA wild-type ovarian cancer had high BRCA1 methylation levels. All of the highly methylated tumors were classified as HRD, achieving a positive predictive value of 100%. We detected 14% (11/79) low-methylated tumors (1%-69%), and all of them were also classified as HRD. Mean GIS was 61.5 for BRCAmut, 66.4 for high-BRCAmeth, 58.9 for low-BRCAmeth, and 33.3 for BRCAwt unmethylated (P < 0.001). Low methylation levels detected in samples previously exposed to chemotherapy appeared to be associated with poor outcome post-platinum. CONCLUSIONS Patients with ovarian cancer with high levels of BRCA1 hypermethylation are very likely to have high GIS and therefore represent good candidates for PARPi treatment. These results may be highly relevant to other tumor types for HRD prediction. See related commentary by Garg and Oza, p. 2957.
Collapse
Affiliation(s)
- Félix Blanc-Durand
- Medical Oncology Department, Gynecology Unit, Institut Gustave Roussy, Villejuif, France
| | - Roseline Tang
- Cancer Genetics Unit, Department of Biology and Pathology, Institut Gustave Roussy, Villejuif, France
| | - Margaux Pommier
- Cancer Genetics Unit, Department of Biology and Pathology, Institut Gustave Roussy, Villejuif, France
| | - Marzieh Nashvi
- Cancer Genetics Unit, Department of Biology and Pathology, Institut Gustave Roussy, Villejuif, France
| | - Sophie Cotteret
- Cancer Genetics Unit, Department of Biology and Pathology, Institut Gustave Roussy, Villejuif, France
| | - Catherine Genestie
- Pathology Unit, Department of Biology and Pathology, Institut Gustave Roussy, Villejuif, France
| | | | - Patricia Pautier
- Medical Oncology Department, Gynecology Unit, Institut Gustave Roussy, Villejuif, France
| | - Judith Michels
- Medical Oncology Department, Gynecology Unit, Institut Gustave Roussy, Villejuif, France
| | - Maria Kfoury
- Medical Oncology Department, Gynecology Unit, Institut Gustave Roussy, Villejuif, France
| | - Robert Hervé
- Oncology Unit, Centre Hospitalier Polynesie Francaise, Papeete, French Polynesia
| | - Sylvie Mengue
- Oncology Unit, Centre Hospitalier Polynesie Francaise, Papeete, French Polynesia
| | - Estelle Wafo
- Gynecology Unit, Centre Hospitalier Intercommunal Creteil, Créteil, France
| | - Antoine Elies
- Gynecology Unit, Centre Hospitalier Intercommunal Creteil, Créteil, France
| | - Gregoire Miailhe
- Gynecology Unit, Groupe Hospitalier Est Francilien, Jossigny, France
| | - Jennifer Uzan
- Gynecology Unit, Groupe Hospitalier Est Francilien, Jossigny, France
| | - Etienne Rouleau
- Cancer Genetics Unit, Department of Biology and Pathology, Institut Gustave Roussy, Villejuif, France
- INSERM U981, Institut Gustave Roussy, Villejuif, France
| | - Alexandra Leary
- Medical Oncology Department, Gynecology Unit, Institut Gustave Roussy, Villejuif, France
- INSERM U981, Institut Gustave Roussy, Villejuif, France
| |
Collapse
|
15
|
Cordani N, Bianchi T, Ammoni LC, Cortinovis DL, Cazzaniga ME, Lissoni AA, Landoni F, Canova S. An Overview of PARP Resistance in Ovarian Cancer from a Molecular and Clinical Perspective. Int J Mol Sci 2023; 24:11890. [PMID: 37569269 PMCID: PMC10418869 DOI: 10.3390/ijms241511890] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 07/18/2023] [Accepted: 07/23/2023] [Indexed: 08/13/2023] Open
Abstract
Epithelial ovarian cancer (EOC), a primarily high-grade serous carcinoma (HGSOC), is one of the major causes of high death-to-incidence ratios of all gynecological cancers. Cytoreductive surgery and platinum-based chemotherapy represent the main treatments for this aggressive disease. Molecular characterization of HGSOC has revealed that up to 50% of cases have a deficiency in the homologous recombination repair (HRR) system, which makes these tumors sensitive to poly ADP-ribose inhibitors (PARP-is). However, drug resistance often occurs and overcoming it represents a big challenge. A number of strategies are under investigation, with the most promising being combinations of PARP-is with antiangiogenetic agents and immune checkpoint inhibitors. Moreover, new drugs targeting different pathways, including the ATR-CHK1-WEE1, the PI3K-AKT and the RAS/RAF/MEK, are under development both in phase I and II-III clinical trials. Nevertheless, there is still a long way to go, and the next few years promise to be exciting.
Collapse
Affiliation(s)
- Nicoletta Cordani
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (T.B.); (L.C.A.); (M.E.C.); (A.A.L.); (F.L.)
| | - Tommaso Bianchi
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (T.B.); (L.C.A.); (M.E.C.); (A.A.L.); (F.L.)
- Clinic of Obstetrics and Gynecology, Fondazione IRCCS San Gerardo dei Tintori, University of Milano-Bicocca, 20900 Monza, Italy
| | - Luca Carlofrancesco Ammoni
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (T.B.); (L.C.A.); (M.E.C.); (A.A.L.); (F.L.)
| | | | - Marina Elena Cazzaniga
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (T.B.); (L.C.A.); (M.E.C.); (A.A.L.); (F.L.)
- Phase 1 Research Centre, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| | - Andrea Alberto Lissoni
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (T.B.); (L.C.A.); (M.E.C.); (A.A.L.); (F.L.)
- Clinic of Obstetrics and Gynecology, Fondazione IRCCS San Gerardo dei Tintori, University of Milano-Bicocca, 20900 Monza, Italy
| | - Fabio Landoni
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (T.B.); (L.C.A.); (M.E.C.); (A.A.L.); (F.L.)
- Clinic of Obstetrics and Gynecology, Fondazione IRCCS San Gerardo dei Tintori, University of Milano-Bicocca, 20900 Monza, Italy
| | - Stefania Canova
- Medical Oncology Unit, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy;
| |
Collapse
|
16
|
Wang C, Block MS, Cunningham JM, Sherman ME, McCauley BM, Armasu SM, Vierkant RA, Traficante N, Talhouk A, Ramus SJ, Pejovic N, Köbel M, Jorgensen BD, Garsed DW, Fereday S, Doherty JA, Ariyaratne D, Anglesio MS, Widschwendter M, Pejovic T, Bosquet JG, Bowtell DD, Winham SJ, Goode EL. Methylation Signature Implicated in Immuno-Suppressive Activities in Tubo-Ovarian High-Grade Serous Carcinoma. Cancer Epidemiol Biomarkers Prev 2023; 32:542-549. [PMID: 36790339 PMCID: PMC10073286 DOI: 10.1158/1055-9965.epi-22-0941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/07/2022] [Accepted: 01/23/2023] [Indexed: 02/16/2023] Open
Abstract
BACKGROUND Better understanding of prognostic factors in tubo-ovarian high-grade serous carcinoma (HGSC) is critical, as diagnosis confers an aggressive disease course. Variation in tumor DNA methylation shows promise predicting outcome, yet prior studies were largely platform-specific and unable to evaluate multiple molecular features. METHODS We analyzed genome-wide DNA methylation in 1,040 frozen HGSC, including 325 previously reported upon, seeking a multi-platform quantitative methylation signature that we evaluated in relation to clinical features, tumor characteristics, time to recurrence/death, extent of CD8+ tumor-infiltrating lymphocytes (TIL), gene expression molecular subtypes, and gene expression of the ATP-binding cassette transporter TAP1. RESULTS Methylation signature was associated with shorter time to recurrence, independent of clinical factors (N = 715 new set, hazard ratio (HR), 1.65; 95% confidence interval (CI), 1.10-2.46; P = 0.015; N = 325 published set HR, 2.87; 95% CI, 2.17-3.81; P = 2.2 × 10-13) and remained prognostic after adjustment for gene expression molecular subtype and TAP1 expression (N = 599; HR, 2.22; 95% CI, 1.66-2.95; P = 4.1 × 10-8). Methylation signature was inversely related to CD8+ TIL levels (P = 2.4 × 10-7) and TAP1 expression (P = 0.0011) and was associated with gene expression molecular subtype (P = 5.9 × 10-4) in covariate-adjusted analysis. CONCLUSIONS Multi-center analysis identified a novel quantitative tumor methylation signature of HGSC applicable to numerous commercially available platforms indicative of shorter time to recurrence/death, adjusting for other factors. Along with immune cell composition analysis, these results suggest a role for DNA methylation in the immunosuppressive microenvironment. IMPACT This work aids in identification of targetable epigenome processes and stratification of patients for whom tailored treatment may be most beneficial.
Collapse
Affiliation(s)
- Chen Wang
- Department of Quantitative Health Sciences, Division of Computational Biology, Mayo Clinic, Rochester, MN, USA
| | | | - Julie M. Cunningham
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Mark E. Sherman
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL, USA
| | - Bryan M. McCauley
- Department of Quantitative Health Sciences, Division of Clinical Trials and Biostatistics, Mayo Clinic, Rochester, MN, USA
| | - Sebastian M. Armasu
- Department of Quantitative Health Sciences, Division of Clinical Trials and Biostatistics, Mayo Clinic, Rochester, MN, USA
| | - Robert A. Vierkant
- Department of Quantitative Health Sciences, Division of Computational Biology, Mayo Clinic, Rochester, MN, USA
| | - Nadia Traficante
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Australian Ovarian Cancer Study Group
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Centre for Cancer Research, The Westmead Institute for Medical Research and Department of Gynaecological Oncology, Westmead Hospital, The University of Sydney, Sydney, New South Wales, Australia
| | - Aline Talhouk
- British Columbia’s Ovarian Cancer Research (OVCARE) Program, BC Cancer, Vancouver General Hospital, and University of British Columbia, Vancouver, BC, Canada
- Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, BC, Canada
| | - Susan J. Ramus
- School of Clinical Medicine, Faculty of Medicine, University of NSW Sydney, Sydney, New South Wales, Australia
- Adult Cancer Program, Lowy Cancer Research Centre, University of NSW Sydney, Sydney, New South Wales, Australia
| | | | - Martin Köbel
- Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, AB, Canada
| | - Brooke D. Jorgensen
- Department of Quantitative Health Sciences, Division of Epidemiology, Mayo Clinic, Rochester, MN, USA
| | - Dale W. Garsed
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Sian Fereday
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Jennifer A. Doherty
- Huntsman Cancer Institute, Department of Population Health Sciences, University of Utah, Salt Lake City, UT, USA
| | | | - Michael S. Anglesio
- British Columbia’s Ovarian Cancer Research (OVCARE) Program, BC Cancer, Vancouver General Hospital, and University of British Columbia, Vancouver, BC, Canada
- Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Martin Widschwendter
- European Translational Oncology Prevention and Screening (EUTOPS) Institute, Universität Innsbruck, Hall in Tirol, Austria
| | - Tanja Pejovic
- Department of Obstetrics and Gynecology, Oregon Health & Science University, Portland, OR, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Jesus Gonzalez Bosquet
- Department of Obstetrics and Gynecologic, Division of Gynecologic Oncology, University of Iowa, Iowa City, IA, USA
| | - David D. Bowtell
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Stacey J. Winham
- Department of Quantitative Health Sciences, Division of Computational Biology, Mayo Clinic, Rochester, MN, USA
| | - Ellen L. Goode
- Department of Quantitative Health Sciences, Division of Epidemiology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
17
|
Sahnane N, Rivera D, Libera L, Carnevali I, Banelli B, Facchi S, Gismondi V, Paudice M, Cirmena G, Vellone VG, Sessa F, Varesco L, Tibiletti MG. Pyrosequencing Assay for BRCA1 Methylation Analysis: Results from a Cross-Validation Study. J Mol Diagn 2023; 25:217-226. [PMID: 36739964 DOI: 10.1016/j.jmoldx.2023.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 12/14/2022] [Accepted: 01/10/2023] [Indexed: 02/05/2023] Open
Abstract
Epithelial ovarian cancers (EOCs) harboring germline or somatic pathogenic variants in BRCA1 and BRCA2 genes show sensitivity to poly(ADP-ribose) polymerase inhibition. It has been suggested that BRCA1 promoter methylation is perhaps a better determinant of therapy response, because of its intrinsic dynamic feature, with respect to genomic scars or gene mutation. Conflicting evidence was reported so far, and the lack of a validated assay to measure promoter methylation was considered a main confounding factor in data interpretation. To contribute to the validation process of a pyrosequencing assay for BRCA1 promoter methylation, 109 EOCs from two Italian centers were reciprocally blindly investigated. By comparing two different pyrosequencing assays, addressing a partially overlapping region of BRCA1 promoter, an almost complete concordance of results was obtained. Moreover, the clinical relevance of this approach was also supported by the finding of BRCA1 transcript down-regulation in BRCA1-methylated EOCs. These findings could lead to the development of a simple and cheap pyrosequencing assay for diagnostics, easily applicable to formalin-fixed, paraffin-embedded tissues. This technique may be implemented in routine clinical practice in the near future to identify EOCs sensitive to poly(ADP-ribose) polymerase inhibitor therapy, thus increasing the subset of women affected by EOCs who could benefit from such treatment.
Collapse
Affiliation(s)
- Nora Sahnane
- Unit of Pathology, Ospedale di Circolo, Azienda Socio Sanitaria Territoriale (ASST) Sette Laghi Hospital, Varese, Italy; Research Center for Familial and Hereditary Tumors, Department of Medicine and Surgery, University of Insubria, Varese, Italy.
| | - Daniela Rivera
- Hereditary Cancer Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale Policlinico San Martino, Genoa, Italy
| | - Laura Libera
- Research Center for Familial and Hereditary Tumors, Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Ileana Carnevali
- Unit of Pathology, Ospedale di Circolo, Azienda Socio Sanitaria Territoriale (ASST) Sette Laghi Hospital, Varese, Italy; Research Center for Familial and Hereditary Tumors, Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Barbara Banelli
- Tumor Epigenetics Laboratory, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale Policlinico San Martino, Genoa, Italy
| | - Sofia Facchi
- Research Center for Familial and Hereditary Tumors, Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Viviana Gismondi
- Hereditary Cancer Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale Policlinico San Martino, Genoa, Italy
| | - Michele Paudice
- Department of Surgical Sciences and Integrated Diagnostics, University of Genoa, Genoa, Italy
| | - Gabriella Cirmena
- Hereditary Cancer Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale Policlinico San Martino, Genoa, Italy
| | - Valerio G Vellone
- Department of Surgical Sciences and Integrated Diagnostics, University of Genoa, Genoa, Italy; Anatomic Pathology University Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale Policlinico San Martino, Genoa, Italy
| | - Fausto Sessa
- Unit of Pathology, Ospedale di Circolo, Azienda Socio Sanitaria Territoriale (ASST) Sette Laghi Hospital, Varese, Italy; Research Center for Familial and Hereditary Tumors, Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Liliana Varesco
- Hereditary Cancer Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale Policlinico San Martino, Genoa, Italy
| | - Maria G Tibiletti
- Unit of Pathology, Ospedale di Circolo, Azienda Socio Sanitaria Territoriale (ASST) Sette Laghi Hospital, Varese, Italy; Research Center for Familial and Hereditary Tumors, Department of Medicine and Surgery, University of Insubria, Varese, Italy
| |
Collapse
|
18
|
Hollis RL. Molecular characteristics and clinical behaviour of epithelial ovarian cancers. Cancer Lett 2023; 555:216057. [PMID: 36627048 DOI: 10.1016/j.canlet.2023.216057] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/05/2023] [Accepted: 01/05/2023] [Indexed: 01/09/2023]
Abstract
Ovarian carcinoma (OC) is an umbrella term for multiple distinct diseases (histotypes), each with their own developmental origins, clinical behaviour and molecular profile. Accordingly, OC management is progressing away from a one-size-fits all approach, toward more molecularly-driven, histotype-specific management strategies. Our knowledge of driver events in high grade serous OC, the most common histotype, has led to major advances in treatments, including PARP inhibitor use. However, these agents are not suitable for all patients, most notably for many of those with rare OC histotypes. Identification of additional targeted therapeutic strategies will require a detailed understanding of the molecular landscape in each OC histotype. Until recently, tumour profiling studies in rare histotypes were sparse; however, significant advances have been made over the last decade. In particular, reports of genomic characterisation in endometrioid, clear cell, mucinous and low grade serous OC have significantly expanded our understanding of mutational events in these tumour types. Nonetheless, substantial knowledge gaps remain. This review summarises our current understanding of each histotype, highlighting recent advances in these unique diseases and outlining immediate research priorities for accelerating progress toward improving patient outcomes.
Collapse
Affiliation(s)
- Robert L Hollis
- Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, UK.
| |
Collapse
|
19
|
Quesada S, Solassol J, Ray-Coquard I, Fabbro M. [Homologous recombination deficiency in epithelial ovarian cancers: from molecular characterization to patient journey]. Bull Cancer 2023; 110:371-381. [PMID: 36863920 DOI: 10.1016/j.bulcan.2023.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 02/03/2023] [Accepted: 02/06/2023] [Indexed: 03/04/2023]
Abstract
High-grade serous ovarian carcinoma (HGSOC), the most frequent and aggressive form of epithelial ovarian cancer is characterized in half of cases by homologous recombination deficiency (HRD). This molecular alteration is defined by distinct causes and consequences. The main and most characterized cause is the presence of an alteration affecting BRCA1 and BRCA2 genes. Regarding consequences, a specific genomic instability leads to increased sensitivity to platinum salts and poly (ADP-ribose) polymerase (PARPi) inhibitors. This latter point enabled the advent of PARPi in first and second line maintenance. As such, the initial and rapid evaluation of HRD status with molecular tests is a key step in the management of HGSOC. Until recently, the range of tests offered proved to be very limited and suffered from technical and medical limitations. This has recently led to the development and validation of alternatives, including academic ones. This "state of the art" review will bring a synthesis concerning the assessment of HRD status in HGSOCs. After a brief introduction of HRD (including main causes and consequences) and of its predictive value regarding PARPi, we will discuss the limitations of current molecular tests and the existing alternatives. Finally, we will contextualize this to the situation in France, with special consideration concerning the positioning and the financial coverage of these tests, with the perspective of optimizing patient management .
Collapse
Affiliation(s)
- Stanislas Quesada
- Institut régional du cancer de Montpellier (ICM/UNICANCER), département d'oncologie médicale, Montpellier, France.
| | - Jérôme Solassol
- Laboratoire de biologie des tumeurs solides, centre hospitalier universitaire (CHU) de Montpellier, Montpellier, France
| | - Isabelle Ray-Coquard
- Centre Léon-Bérard (CLB-UNICANCER), département d'oncologie médicale, université Claude-Bernard (UCB Lyon 1), Lyon, France
| | - Michel Fabbro
- Institut régional du cancer de Montpellier (ICM/UNICANCER), département d'oncologie médicale, Montpellier, France
| |
Collapse
|
20
|
Pujade-Lauraine E, Brown J, Barnicle A, Wessen J, Lao-Sirieix P, Criscione SW, du Bois A, Lorusso D, Romero I, Petru E, Yoshida H, Vergote I, Colombo N, Hietanen S, Provansal M, Schmalfeldt B, Pignata S, Martín Lorente C, Berton D, Runnebaum IB, Ray-Coquard I. Homologous Recombination Repair Gene Mutations to Predict Olaparib Plus Bevacizumab Efficacy in the First-Line Ovarian Cancer PAOLA-1/ENGOT-ov25 Trial. JCO Precis Oncol 2023; 7:e2200258. [PMID: 36716415 PMCID: PMC9928987 DOI: 10.1200/po.22.00258] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
PURPOSE The PAOLA-1/ENGOT-ov25 trial of maintenance olaparib plus bevacizumab for newly diagnosed advanced high-grade ovarian cancer demonstrated a significant progression-free survival (PFS) benefit over placebo plus bevacizumab, particularly in patients with homologous recombination deficiency (HRD)-positive tumors. We explored whether mutations in non-BRCA1 or BRCA2 homologous recombination repair (non-BRCA HRRm) genes predicted benefit from olaparib plus bevacizumab in PAOLA-1. METHODS Eight hundred and six patients were randomly assigned (2:1). Tumors were analyzed using the Myriad MyChoice HRD Plus assay to assess non-BRCA HRRm and HRD status; HRD was based on a genomic instability score (GIS) of ≥ 42. In this exploratory analysis, PFS was assessed in patients harboring deleterious mutations using six non-BRCA HRR gene panels, three devised for this analysis and three previously published. RESULTS The non-BRCA HRRm prevalence ranged from 30 of 806 (3.7%) to 79 of 806 (9.8%) depending on the gene panel used, whereas 152 of 806 (18.9%) had non-BRCA1 or BRCA2 mutation HRD-positive tumors. The majority of tumors harboring non-BRCA HRRm had a low median GIS; however, a GIS of > 42 was observed for tumors with mutations in five HRR genes (BLM, BRIP1, RAD51C, PALB2, and RAD51D). Rates of gene-specific biallelic loss were variable (0% to 100%) in non-BRCA HRRm tumors relative to BRCA1-mutated (99%) or BRCA2-mutated (86%) tumors. Across all gene panels tested, hazard ratios for PFS (95% CI) ranged from 0.92 (0.51 to 1.73) to 1.83 (0.76 to 5.43). CONCLUSION Acknowledging limitations of small subgroup sizes, non-BRCA HRRm gene panels were not predictive of PFS benefit with maintenance olaparib plus bevacizumab versus placebo plus bevacizumab in PAOLA-1, irrespective of the gene panel tested. Current gene panels exploring HRRm should not be considered a substitute for HRD determined by BRCA mutation status and genomic instability testing in first-line high-grade ovarian cancer.
Collapse
Affiliation(s)
- Eric Pujade-Lauraine
- ARCAGY-GINECO, Paris, France,Eric Pujade-Lauraine, MD, 8 rue Lamennais, 75008 Paris, France; e-mail:
| | | | | | | | | | | | | | - Domenica Lorusso
- Fondazione IRCCS Istituto Nazionale Tumori, Milan and MITO, Italy
| | - Ignacio Romero
- Instituto Valenciano de Oncología, Valencia and GEICO, Spain
| | - Edgar Petru
- Universitätsklinik für Frauenheilkunde und Geburtshilfe der Med, Universität Graz, Graz and AGO, Austria
| | - Hiroyuki Yoshida
- Saitama Medical University International Medical Center, Saitama and GOTIC, Japan
| | - Ignace Vergote
- University Hospital Leuven, Leuven Cancer Institute, Leuven and BGOG, Belgium
| | - Nicoletta Colombo
- University of Milan-Bicocca and Istituto Europeo di Oncologia IRCCS, Milan and MANGO, Italy
| | | | | | | | - Sandro Pignata
- Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, Naples and MITO, Italy
| | | | - Dominique Berton
- L'Institut de Cancérologie de l'Ouest (ICO), Center René Gauducheau, Saint Herblain and GINECO, France
| | - Ingo B. Runnebaum
- Jena University Hospital, Universitaets-Frauenklinik Jena and AGO, Germany
| | - Isabelle Ray-Coquard
- Center Léon Bérard and University Claude Bernard Lyon 1, Lyon and GINECO, France
| |
Collapse
|
21
|
Oswald AJ, Gourley C. Development of Homologous Recombination Functional Assays for Targeting the DDR. Cancer Treat Res 2023; 186:43-70. [PMID: 37978130 DOI: 10.1007/978-3-031-30065-3_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2023]
Abstract
Identification of tumours that have homologous recombination deficiency (HRD) has become of increasing interest following the licensing of PARP inhibitors. Potential methods to assess HRD status include; clinical selection for platinum sensitive disease, mutational/methylation status, genomic scars/signature and functional RAD51 assays. Homologous recombination (HR) is a dynamic process with the potential to evolve over a disease course, particularly in relation to previous treatment. This is one of the major drawbacks of genomic scars/signatures, as they only demonstrate historic HR status. Functional HR assays have the benefit of giving a real time HR status readout and therefore have the potential for clearer identification of patients who may benefit from PARP inhibitors at that specific time point. However, the development of RAD51 foci assays ready for clinical practice has been challenging. Pre-clinical considerations have included; controlling for variation in tumour proliferation, tissue type and whether DNA damage induction is required. Furthermore, the assays require correlation with clinical outcomes, an understanding of how they complement current testing modalities and validation of test performance in large cohorts. Despite these challenges, given the profound benefit from PARP inhibitors seen in those with an HRD phenotype to date, the ongoing development and validation of these functional HR assays remains of high clinical importance.
Collapse
Affiliation(s)
- Ailsa J Oswald
- Cancer Research UK Scotland Centre, University of Edinburgh, Edinburgh, UK.
| | - Charlie Gourley
- Cancer Research UK Scotland Centre, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
22
|
Jang J, Kim Y, Kim JH, Cho SM, Lee KA. Cost-Effectiveness Analysis of Germline and Somatic BRCA Testing in Patients With Advanced Ovarian Cancer. Ann Lab Med 2023; 43:73-81. [PMID: 36045059 PMCID: PMC9467835 DOI: 10.3343/alm.2023.43.1.73] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 04/14/2022] [Accepted: 08/02/2022] [Indexed: 12/27/2022] Open
Abstract
Background BRCA testing is necessary for establishing a management strategy for ovarian cancer. Several BRCA testing strategies, including germline and somatic testing, are implemented in clinical practice in Korea. We aimed to comparatively evaluate their cost-effectiveness from patients' perspective. Methods We developed a decision model comprising five BRCA testing strategies implemented in Korea: (1) germline testing first, followed by somatic tumor testing for patients without a germline variant; (2) somatic testing first, followed by germline testing for patients with a variant detected by somatic testing; (3) both germline and somatic testing; (4) germline testing alone; and (5) somatic testing alone, with no testing as the comparator. One-way sensitivity analysis was conducted to test the uncertainty of key parameters. Results Assuming a willingness-to-pay of $20,000 per progression-free life-year gain (PF-LYG), all five strategies were considered cost-effective. Strategy 4 was the most cost-effective option, with an incremental cost-effectiveness ratio (ICER) of $2,547.7 per PF-LYG, followed by strategy 1, with an ICER of $3,978.4 per PF-LYG. Even when the parameter values were varied within the possible range, the ICERs of all strategies did not exceed the willingness-to-pay threshold. Conclusions Considering the importance of knowing a patient's BRCA gene status, germline testing first, followed by somatic testing, may be a reasonable option.
Collapse
Affiliation(s)
- Jaehyeok Jang
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Yoonjung Kim
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Jae-Hoon Kim
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Seoul, Korea
| | - Sun-Mi Cho
- Department of Laboratory Medicine, CHA Bundang Medical Center, CHA University, Seongnam, Korea
| | - Kyung-A Lee
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
23
|
DNA methylation of the immediate upstream region of BRCA1 major transcription start sites is an independent favorable prognostic factor in patients with high-grade serous ovarian cancer. Gynecol Oncol 2022; 167:513-518. [PMID: 36253303 DOI: 10.1016/j.ygyno.2022.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 10/05/2022] [Accepted: 10/07/2022] [Indexed: 11/04/2022]
Abstract
OBJECTIVE To establish a quantitative method to evaluate the DNA methylation level of an immediate upstream region of major BRCA1 transcriptional start sites (TSSs), and to investigate whether methylation of the region is a prognostic factor in high-grade serous ovarian cancer patients after neoadjuvant chemotherapy. METHODS Ninety-two FFPE samples of advanced high-grade serous ovarian cancers after neoadjuvant chemotherapy between 2011 and 2018 were used for mutation and methylation analysis. DNA methylation levels were assessed by pyrosequencing and DNA methylation microarray. An association between methylation level (or a mutation) and progression-free survival was assessed by Kaplan-Meier analysis. RESULT Major BRCA1 transcripts and CpG sites immediately upstream of their TSSs were identified, and a pyrosequencing method was developed. BRCA1 methylation, BRCA1/2 mutations, and a RAD51C mutation were detected in 17/79 (21.5%), 17/92 (18.5%), and 1/92 (1.1%) high-grade serious ovarian cancer samples. In univariate analysis, BRCA1 methylation and no residual tumor were associated with progression-free survival (BRCA1 methylation: P = 0.025, no residual tumor: P = 0.0026). Multivariate analysis showed that both BRCA1 methylation (P = 0.038, HR = 0.47, 95% CI: 0.21-0.96) and no residual tumor (P = 0.012, HR = 0.49, 95% CI: 0.28-0.85) were significant favorable prognostic factors. CONCLUSION A quantitative method to estimate the methylation level of the immediate upstream region of major BRCA1 TSSs was established. Methylation of the region of was an independent favorable prognostic factor in high-grade serous ovarian cancer patients.
Collapse
|
24
|
Oda K, Aoki D, Tsuda H, Nishihara H, Aoyama H, Inomata H, Shimada M, Enomoto T. Japanese nationwide observational multicenter study of tumor BRCA1/2 variant testing in advanced ovarian cancer. Cancer Sci 2022; 114:271-280. [PMID: 36254756 PMCID: PMC9807512 DOI: 10.1111/cas.15518] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 07/13/2022] [Accepted: 07/26/2022] [Indexed: 01/07/2023] Open
Abstract
The association between germline BRCA1 and BRCA2 pathogenic variants (mutations: gBRCAm) and ovarian cancer risk is well established. Germline testing alone cannot detect somatic BRCA1/2 pathogenic variants (sBRCAm), which is calculated based on the proportion of tumor BRCAm (tBRCAm) from tumor samples and gBRCAm. Homologous recombination deficiency (HRD) results mainly from genetic/epigenetic alterations in homologous recombination repair-related genes and can be evaluated by genomic instability status. In Japan, the prevalence of tBRCAm, sBRCAm, and HRD remains unclear. This multicenter, cross-sectional, observational study, CHaRacterIzing the croSs-secTional approach to invEstigate the prevaLence of tissue BRCA1/2 mutations in newLy diagnosEd advanced ovarian cancer patients (CHRISTELLE), evaluated the prevalence of tBRCAm, sBRCAm, and HRD in tumor specimens from newly diagnosed patients with ovarian cancer who underwent gBRCA testing. Of the 205 patients analyzed, 26.8% had a tBRCAm, including tBRCA1m (17.6%) and tBRCA2m (9.3%). The overall prevalence of tBRCAm, gBRCAm, sBRCAm, and HRD-positive status was 26.8%, 21.5%, 6.3%, and 60.0%, respectively. The calculated sBRCAm/tBRCAm ratio was 23.6% (13/55), and the prevalence of gBRCA variant of uncertain significance was 3.9%. These results suggest gBRCA testing alone cannot clearly identify the best course of treatment, highlighting the importance of sBRCA testing in Japan. The present results also suggest that testing for tBRCA and HRD should be encouraged in advanced ovarian cancer patients to drive precision medicine.
Collapse
Affiliation(s)
- Katsutoshi Oda
- Division of Integrative Genomics, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Daisuke Aoki
- Department of Obstetrics and GynecologyKeio University School of MedicineTokyoJapan
| | - Hitoshi Tsuda
- Department of Basic PathologyNational Defense Medical CollegeSaitamaJapan
| | | | | | | | - Muneaki Shimada
- Department of Obstetrics and GynecologyTohoku University Graduate School of MedicineMiyagiJapan
| | - Takayuki Enomoto
- Department of Obstetrics and GynecologyNiigata University Medical SchoolNiigataJapan
| |
Collapse
|
25
|
Miller RE, Elyashiv O, El-Shakankery KH, Ledermann JA. Ovarian Cancer Therapy: Homologous Recombination Deficiency as a Predictive Biomarker of Response to PARP Inhibitors. Onco Targets Ther 2022; 15:1105-1117. [PMID: 36217436 PMCID: PMC9547601 DOI: 10.2147/ott.s272199] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 09/13/2022] [Indexed: 11/05/2022] Open
Abstract
Poly(adenosine diphosphate-ribose) polymerase (PARP) inhibitors have revolutionised the management of patients with high-grade serous and endometrioid ovarian cancer demonstrating significant improvements in progression-free survival. Whilst the greatest benefit is seen with BRCA1/2 mutant cancers, it is clear that the benefit extends beyond this group. This sensitivity is thought to be due to homologous recombination deficiency (HRD), which is present in up to 50% of the high-grade serous cancers. Several different HRD assays exist, which fall into one of three main categories: homologous recombination repair (HRR)-related gene analysis, genomic "scars" and/or mutational signatures, and real-time HRD functional assessment. We review the emerging data on HRD as a predictive biomarker for PARP inhibitors and discuss the merits and disadvantages of different HRD assays.
Collapse
Affiliation(s)
- Rowan E Miller
- Department of Medical Oncology, University College London Hospital, London, UK
- Department of Medical Oncology, St Bartholomew’s Hospital, London, UK
| | - Osnat Elyashiv
- Department of Medical Oncology, University College London Hospital, London, UK
| | | | - Jonathan A Ledermann
- Department of Medical Oncology, University College London Hospital, London, UK
- UCL Cancer Institute, University College London, London, UK
| |
Collapse
|
26
|
Kloeber JA, Lou Z. Critical DNA damaging pathways in tumorigenesis. Semin Cancer Biol 2022; 85:164-184. [PMID: 33905873 PMCID: PMC8542061 DOI: 10.1016/j.semcancer.2021.04.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 04/13/2021] [Accepted: 04/15/2021] [Indexed: 12/22/2022]
Abstract
The acquisition of DNA damage is an early driving event in tumorigenesis. Premalignant lesions show activated DNA damage responses and inactivation of DNA damage checkpoints promotes malignant transformation. However, DNA damage is also a targetable vulnerability in cancer cells. This requires a detailed understanding of the cellular and molecular mechanisms governing DNA integrity. Here, we review current work on DNA damage in tumorigenesis. We discuss DNA double strand break repair, how repair pathways contribute to tumorigenesis, and how double strand breaks are linked to the tumor microenvironment. Next, we discuss the role of oncogenes in promoting DNA damage through replication stress. Finally, we discuss our current understanding on DNA damage in micronuclei and discuss therapies targeting these DNA damage pathways.
Collapse
Affiliation(s)
- Jake A Kloeber
- Department of Oncology, Mayo Clinic, Rochester, MN, 55905, USA; Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, 55905, USA; Mayo Clinic Medical Scientist Training Program, Mayo Clinic, Rochester, MN, 55905, USA
| | - Zhenkun Lou
- Department of Oncology, Mayo Clinic, Rochester, MN, 55905, USA.
| |
Collapse
|
27
|
Bound NT, Vandenberg CJ, Kartikasari AER, Plebanski M, Scott CL. Improving PARP inhibitor efficacy in high-grade serous ovarian carcinoma: A focus on the immune system. Front Genet 2022; 13:886170. [PMID: 36159999 PMCID: PMC9505691 DOI: 10.3389/fgene.2022.886170] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 08/05/2022] [Indexed: 12/03/2022] Open
Abstract
High-grade serous ovarian carcinoma (HGSOC) is a genomically unstable malignancy responsible for over 70% of all deaths due to ovarian cancer. With roughly 50% of all HGSOC harboring defects in the homologous recombination (HR) DNA repair pathway (e.g., BRCA1/2 mutations), the introduction of poly ADP-ribose polymerase inhibitors (PARPi) has dramatically improved outcomes for women with HR defective HGSOC. By blocking the repair of single-stranded DNA damage in cancer cells already lacking high-fidelity HR pathways, PARPi causes the accumulation of double-stranded DNA breaks, leading to cell death. Thus, this synthetic lethality results in PARPi selectively targeting cancer cells, resulting in impressive efficacy. Despite this, resistance to PARPi commonly develops through diverse mechanisms, such as the acquisition of secondary BRCA1/2 mutations. Perhaps less well documented is that PARPi can impact both the tumour microenvironment and the immune response, through upregulation of the stimulator of interferon genes (STING) pathway, upregulation of immune checkpoints such as PD-L1, and by stimulating the production of pro-inflammatory cytokines. Whilst targeted immunotherapies have not yet found their place in the clinic for HGSOC, the evidence above, as well as ongoing studies exploring the synergistic effects of PARPi with immune agents, including immune checkpoint inhibitors, suggests potential for targeting the immune response in HGSOC. Additionally, combining PARPi with epigenetic-modulating drugs may improve PARPi efficacy, by inducing a BRCA-defective phenotype to sensitise resistant cancer cells to PARPi. Finally, invigorating an immune response during PARPi therapy may engage anti-cancer immune responses that potentiate efficacy and mitigate the development of PARPi resistance. Here, we will review the emerging PARPi literature with a focus on PARPi effects on the immune response in HGSOC, as well as the potential of epigenetic combination therapies. We highlight the potential of transforming HGSOC from a lethal to a chronic disease and increasing the likelihood of cure.
Collapse
Affiliation(s)
- Nirashaa T. Bound
- Cancer Biology and Stem Cells, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Cancer Ageing and Vaccines (CAVA), Translational Immunology & Nanotechnology Research Program, School of Health & Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - Cassandra J. Vandenberg
- Cancer Biology and Stem Cells, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Apriliana E. R. Kartikasari
- Cancer Ageing and Vaccines (CAVA), Translational Immunology & Nanotechnology Research Program, School of Health & Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - Magdalena Plebanski
- Cancer Ageing and Vaccines (CAVA), Translational Immunology & Nanotechnology Research Program, School of Health & Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - Clare L. Scott
- Cancer Biology and Stem Cells, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
- Peter MacCallum Cancer Centre, Parkville, VIC, Australia
- Royal Women’s Hospital, Parkville, VIC, Australia
| |
Collapse
|
28
|
Hollis RL, Meynert AM, Michie CO, Rye T, Churchman M, Hallas-Potts A, Croy I, McCluggage WG, Williams AR, Bartos C, Iida Y, Okamoto A, Dougherty B, Barrett JC, March R, Matakidou A, Roxburgh P, Semple CA, Harkin DP, Kennedy R, Herrington CS, Gourley C. Multiomic Characterization of High-Grade Serous Ovarian Carcinoma Enables High-Resolution Patient Stratification. Clin Cancer Res 2022; 28:3546-3556. [PMID: 35696721 PMCID: PMC9662902 DOI: 10.1158/1078-0432.ccr-22-0368] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 04/13/2022] [Accepted: 06/09/2022] [Indexed: 01/07/2023]
Abstract
PURPOSE High-grade serous ovarian carcinoma (HGSOC) is the most common ovarian cancer type; most patients experience disease recurrence that accumulates chemoresistance, leading to treatment failure. Genomic and transcriptomic features have been associated with differential outcome and treatment response. However, the relationship between events at the gene sequence, copy number, and gene-expression levels remains poorly defined. EXPERIMENTAL DESIGN We perform multiomic characterization of a large HGSOC cohort (n = 362) with detailed clinical annotation to interrogate the relationship between patient subgroups defined by specific molecular events. RESULTS BRCA2-mutant (BRCA2m) and EMSY-overexpressing cases demonstrated prolonged survival [multivariable hazard ratios (HR) 0.40 and 0.51] and significantly higher first- and second-line chemotherapy response rate. CCNE1-gained (CCNE1g) cases demonstrated underrepresentation of FIGO stage IV cases, with shorter survival but no significant difference in treatment response. We demonstrate marked overlap between the TCGA- and Tothill-derived subtypes. IMR/C2 cases displayed higher BRCA1/2m frequency (25.5%, 32.5%) and significantly greater immune cell infiltration, whereas PRO/C5 cases had the highest CCNE1g rate (23.9%, 22.2%) and were uniformly low in immune cell infiltration. The survival benefit for cases with aberrations in homologous recombination repair (HRR) genes was apparent across all transcriptomic subtypes (HR range, 0.48-0.68). There was significant co-occurrence of RB loss and HRR gene aberrations; RB loss was further associated with favorable survival within HRR-aberrant cases (multivariable HR, 0.50). CONCLUSIONS These data paint a high-resolution picture of the molecular landscape in HGSOC, better defining patients who may benefit most from specific molecular therapeutics and highlighting those for whom novel treatment strategies are needed to improve outcomes.
Collapse
Affiliation(s)
- Robert L. Hollis
- Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
- Corresponding Author: Robb L. Hollis, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XU, Scotland, UK. E-mail:
| | - Alison M. Meynert
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Caroline O. Michie
- Edinburgh Cancer Centre, Western General Hospital, NHS Lothian, Edinburgh, UK
| | - Tzyvia Rye
- Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Michael Churchman
- Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Amelia Hallas-Potts
- Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Ian Croy
- Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | | | | | - Clare Bartos
- Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Yasushi Iida
- Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
- The Jikei University School of Medicine, Tokyo, Japan
| | - Aikou Okamoto
- The Jikei University School of Medicine, Tokyo, Japan
| | - Brian Dougherty
- Translational Medicine, Oncology R&D, AstraZeneca, Waltham, Massachusetts
| | - J. Carl Barrett
- Translational Medicine, Oncology R&D, AstraZeneca, Waltham, Massachusetts
| | - Ruth March
- Precision Medicine and Biosamples, Oncology R&D, AstraZeneca, Cambridge, UK
| | - Athena Matakidou
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Patricia Roxburgh
- Institute of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Belfast, UK
- Beatson West of Scotland Cancer Centre, Glasgow, UK
| | - Colin A. Semple
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - D. Paul Harkin
- Almac Diagnostics, Craigavon, UK
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK
| | - Richard Kennedy
- Almac Diagnostics, Craigavon, UK
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK
| | - C. Simon Herrington
- Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Charlie Gourley
- Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
29
|
Kiendrebeogo IT, Zoure AA, Zongo FI, Ouedraogo SY, Sawadogo AY, Amegnona J, Sombie HK, Bazie JTVE, Sorgho PA, Yonli AT, Ouedraogo MNL, Obiri-Yeboah D, Zongo N, Bambara HA, Simpore J. Screening of BRCA1 (c.5177_5180delGAAA rs80357867 and c.4986+6T>C rs80358086) and the BRCA2 (c.6445_6446delAT rs80359592) Genes for Breast Cancer Prevention in Burkina Faso. Ethiop J Health Sci 2022; 32:699-708. [PMID: 35950060 PMCID: PMC9341024 DOI: 10.4314/ejhs.v32i4.5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 04/12/2022] [Indexed: 11/17/2022] Open
Abstract
Background The objective of this study is to search for mutations in the BRCA1 (c.5177_5180delGAAA and c.4986+6T>C) and BRCA2 genes (c.6445_6446delAT) in a population of women diagnosed with breast cancer. Methods This is a case-control study that involved 140 participants, including 70 patients with histologically diagnosed breast cancer and 70 healthy women without breast cancer. Mutations in the BRCA1 (rs80357867, rs80358086) and BRCA2 (rs80359592) genes were tested by real-time PCR. The 95% confidence interval Odds Ratio (OR) was used to estimate the associations between specific genotypes and breast cancer. Results The study revealed that no mutations were detected for rs80359592. Similarly, no reference allele (TTTC/TTTC) of rs80357867 was found in this study. However, the homozygous double mutant (-/) genotype of this rs80357867 was observed in 11.43% and 1.43% of patients and controls respectively, while 88.57% of patients and 98.57% of controls had a heterozygous deletion (TTTC/-). Concerning rs80358086, 8.57% of the patients had a heterozygous mutation (A/G) with no significantly risk association with occurrence of breast cancer (OR = 6.46; 95% CI: 0.75-55.21; p = 0.11). In addition, this heterozygous mutation was significantly associated with a family history of breast cancer (OR=128; 95% CI: 9.46-1730.93) and breast cancer risk in nonmultiparous women (OR=6; 95% CI: 1-35.90; p= 0.05) but no association with overweight/obesity (OR=1.66; 95% CI: 0.18-15.35; p=1). Conclusion This study shows high frequencies of heterozygous mutation of rs80357867 and rs80358086 from patients. In Burkina Faso, these results could help with early diagnosis of breast cancer in patients.
Collapse
Affiliation(s)
- Isabelle T Kiendrebeogo
- Laboratory of Molecular Biology and Genetics, UFR/SVT, University Joseph KI-ZERBO, Burkina Faso
- Pietro Annigoni Biomolecular Research Center, Burkina Faso
| | - Abdou A Zoure
- Laboratory of Molecular Biology and Genetics, UFR/SVT, University Joseph KI-ZERBO, Burkina Faso
- Institute of Health Sciences Research, Department of Biomedical and Public Health, Burkina Faso
| | - Fabienne I Zongo
- Laboratory of Molecular Biology and Genetics, UFR/SVT, University Joseph KI-ZERBO, Burkina Faso
| | - Serge Y Ouedraogo
- Laboratory of Molecular Biology and Genetics, UFR/SVT, University Joseph KI-ZERBO, Burkina Faso
- Pietro Annigoni Biomolecular Research Center, Burkina Faso
| | - Alexis Y Sawadogo
- Service of Gynecology, University Hospital Center Bogodogo, University Joseph KI-ZERBO, UFR/SDS, Burkina Faso
| | - Jospin Amegnona
- Laboratory of Molecular Biology and Genetics, UFR/SVT, University Joseph KI-ZERBO, Burkina Faso
| | - Herman K Sombie
- Laboratory of Molecular Biology and Genetics, UFR/SVT, University Joseph KI-ZERBO, Burkina Faso
- Pietro Annigoni Biomolecular Research Center, Burkina Faso
| | - Jean T Valérie Elvira Bazie
- Laboratory of Molecular Biology and Genetics, UFR/SVT, University Joseph KI-ZERBO, Burkina Faso
- Pietro Annigoni Biomolecular Research Center, Burkina Faso
| | - Pegdwendé A Sorgho
- Laboratory of Molecular Biology and Genetics, UFR/SVT, University Joseph KI-ZERBO, Burkina Faso
- Pietro Annigoni Biomolecular Research Center, Burkina Faso
| | - Albert T Yonli
- Laboratory of Molecular Biology and Genetics, UFR/SVT, University Joseph KI-ZERBO, Burkina Faso
- Pietro Annigoni Biomolecular Research Center, Burkina Faso
| | - Marie N Lamoussa Ouedraogo
- Laboratory of Molecular Biology and Genetics, UFR/SVT, University Joseph KI-ZERBO, Burkina Faso
- Faculty of Health Sciences, University Saint Thomas d'Aquin
| | - Dorcas Obiri-Yeboah
- Department of Microbiology and Immunology, School of Medical Sciences, University of Cape Coast
| | - Nayi Zongo
- Department of General and Digestive Surgery, University Hospital Center Yalgado Ouedraogo, University Joseph KI-ZERBO, UFR/SDS, Burkina Faso
| | - Hierrhum A Bambara
- Service of oncology, University Hospital Center Bogodogo, University Joseph KI-ZERBO, UFR/SDS, Burkina Faso
| | - Jacques Simpore
- Laboratory of Molecular Biology and Genetics, UFR/SVT, University Joseph KI-ZERBO, Burkina Faso
- Pietro Annigoni Biomolecular Research Center, Burkina Faso
- Faculty of Health Sciences, University Saint Thomas d'Aquin
| |
Collapse
|
30
|
Morgan RD, Burghel GJ, Flaum N, Bulman M, Smith P, Clamp AR, Hasan J, Mitchell CL, Salih Z, Woodward ER, Lalloo F, Crosbie EJ, Edmondson RJ, Wallace AJ, Jayson GC, Evans DGR. BRCA1/2 in non-mucinous epithelial ovarian cancer: tumour with or without germline testing? Br J Cancer 2022; 127:163-167. [PMID: 35260807 PMCID: PMC9276796 DOI: 10.1038/s41416-022-01773-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 02/04/2022] [Accepted: 02/22/2022] [Indexed: 11/09/2022] Open
Abstract
National guidelines recommend testing all cases of non-mucinous epithelial ovarian cancer (NMEOC) for germline (blood) and somatic (tumour) BRCA1/2 pathogenic variants (PVs). We performed paired germline and somatic BRCA1/2 testing in consecutive cases of NMEOC (n = 388) to validate guidelines. Thirty-four somatic BRCA1/2 (sBRCA) PVs (9.7%) were detected in 350 cases with germline BRCA1/2 (gBRCA) wild-type. All sBRCA PVs were detected in non-familial cases. By analysing our regional germline BRCA1/2 database there were 92/1114 (8.3%) gBRCA PVs detected in non-familial cases (only 3% ≥70 years old) and 245/641 (38.2%) in familial cases. Germline non-familial cases were dominated by BRCA2 in older women (8/271 ≥ 70 years old, all BRCA2). The ratio of sBRCA-to-gBRCA was ≤1.0 in women aged <70 years old, compared to 5.2 in women aged ≥70 years old (P = 0.005). The likelihood of missed germline BRCA1/2 PVs (copy-number variants missed on most somatic assays) by testing only tumour DNA was 0.4% in women aged ≥70 years old. We recommend reflex tumour BRCA1/2 testing in all NMEOC cases, and that gBRCA testing is not required for women aged ≥70 years old with no identifiable tumour BRCA1/2 PV and/or family history of breast, ovarian, prostate and/or pancreatic cancer.
Collapse
Affiliation(s)
- Robert D Morgan
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK. .,Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
| | - George J Burghel
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Manchester, UK
| | - Nicola Flaum
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK.,Division of Evolution & Genomic Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Michael Bulman
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Manchester, UK
| | - Philip Smith
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Manchester, UK
| | - Andrew R Clamp
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK.,Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Jurjees Hasan
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | - Claire L Mitchell
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | - Zena Salih
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | - Emma R Woodward
- Division of Evolution & Genomic Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,Department of Clinical Genetics, Manchester University NHS Foundation Trust, Manchester, UK
| | - Fiona Lalloo
- Department of Clinical Genetics, Manchester University NHS Foundation Trust, Manchester, UK
| | - Emma J Crosbie
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,Department of Gynaecological Surgery, Manchester University NHS Foundation Trust, Manchester, UK
| | - Richard J Edmondson
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,Department of Gynaecological Surgery, Manchester University NHS Foundation Trust, Manchester, UK
| | - Andrew J Wallace
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Manchester, UK
| | - Gordon C Jayson
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK.,Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - D Gareth R Evans
- Division of Evolution & Genomic Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,Department of Clinical Genetics, Manchester University NHS Foundation Trust, Manchester, UK
| |
Collapse
|
31
|
Mekonnen N, Yang H, Shin YK. Homologous Recombination Deficiency in Ovarian, Breast, Colorectal, Pancreatic, Non-Small Cell Lung and Prostate Cancers, and the Mechanisms of Resistance to PARP Inhibitors. Front Oncol 2022; 12:880643. [PMID: 35785170 PMCID: PMC9247200 DOI: 10.3389/fonc.2022.880643] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 05/18/2022] [Indexed: 11/30/2022] Open
Abstract
Homologous recombination (HR) is a highly conserved DNA repair mechanism that protects cells from exogenous and endogenous DNA damage. Breast cancer 1 (BRCA1) and breast cancer 2 (BRCA2) play an important role in the HR repair pathway by interacting with other DNA repair proteins such as Fanconi anemia (FA) proteins, ATM, RAD51, PALB2, MRE11A, RAD50, and NBN. These pathways are frequently aberrant in cancer, leading to the accumulation of DNA damage and genomic instability known as homologous recombination deficiency (HRD). HRD can be caused by chromosomal and subchromosomal aberrations, as well as by epigenetic inactivation of tumor suppressor gene promoters. Deficiency in one or more HR genes increases the risk of many malignancies. Another conserved mechanism involved in the repair of DNA single-strand breaks (SSBs) is base excision repair, in which poly (ADP-ribose) polymerase (PARP) enzymes play an important role. PARP inhibitors (PARPIs) convert SSBs to more cytotoxic double-strand breaks, which are repaired in HR-proficient cells, but remain unrepaired in HRD. The blockade of both HR and base excision repair pathways is the basis of PARPI therapy. The use of PARPIs can be expanded to sporadic cancers displaying the “BRCAness” phenotype. Although PARPIs are effective in many cancers, their efficacy is limited by the development of resistance. In this review, we summarize the prevalence of HRD due to mutation, loss of heterozygosity, and promoter hypermethylation of 35 DNA repair genes in ovarian, breast, colorectal, pancreatic, non-small cell lung cancer, and prostate cancer. The underlying mechanisms and strategies to overcome PARPI resistance are also discussed.
Collapse
Affiliation(s)
- Negesse Mekonnen
- Department of Pharmacy, Research Institute of Pharmaceutical Science, Seoul National University College of Pharmacy, Seoul, South Korea
- Department of Veterinary Science, School of Animal Science and Veterinary Medicine, Bahir Dar University, Bahir Dar, Ethiopia
| | - Hobin Yang
- Department of Pharmacy, Research Institute of Pharmaceutical Science, Seoul National University College of Pharmacy, Seoul, South Korea
| | - Young Kee Shin
- Department of Pharmacy, Research Institute of Pharmaceutical Science, Seoul National University College of Pharmacy, Seoul, South Korea
- Bio-MAX/N-Bio, Seoul National University, Seoul, South Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Seoul National University Graduate School of Convergence Science and Technology, Seoul, South Korea
- LOGONE Bio Convergence Research Foundation, Center for Companion Diagnostics, Seoul, South Korea
- *Correspondence: Young Kee Shin,
| |
Collapse
|
32
|
Gallardo-Rincón D, Montes-Servín E, Alamilla-García G, Montes-Servín E, Bahena-González A, Cetina-Pérez L, Morales Vásquez F, Cano-Blanco C, Coronel-Martínez J, González-Ibarra E, Espinosa-Romero R, María Alvarez-Gómez R, Pedroza-Torres A, Castro-Eguiluz D. Clinical Benefits of Olaparib in Mexican Ovarian Cancer Patients With Founder Mutation BRCA1-Del ex9-12. Front Genet 2022; 13:863956. [PMID: 35734436 PMCID: PMC9207274 DOI: 10.3389/fgene.2022.863956] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 05/02/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Ovarian cancer (OC) is gynecologic cancer with the highest mortality rate. It is estimated that 13–17% of ovarian cancers are due to heritable mutations in BRCA1 and BRCA2. The BRCA1 (BRCA1-Del ex9-12) Mexican founder mutation is responsible for 28–35% of the cases with ovarian cancer. The aim was to describe the PFS of OC patients treated with olaparib, emphasizing patients carrying the Mexican founder mutation (BRCA1-Del ex9-12). Methods: In this observational study, of 107 patients with BRCAm, 35 patients were treated with olaparib from November 2016 to May 2021 at the Ovarian Cancer Program (COE) of Mexico; patient information was extracted from electronic medical records. Results: Of 311 patients, 107 (34.4%) were with BRCAm; 71.9% (77/107) were with BRCA1, of which 27.3% (21/77) were with BRCA1-Del ex9-12, and 28.1% (30/107) were with BRCA2 mutations. Only 35 patients received olaparib treatment, and the median follow-up was 12.87 months. The PFS of BRCA1-Del ex9-12 was NR (non-reach); however, 73% of the patients received the treatment at 36 vs. 11.59 months (95% CI; 10.43–12.75) in patients with other BRCAm (p = 0.008). Almost 50% of patients required dose reduction due to toxicity; the most frequent adverse events were hematological in 76.5% and gastrointestinal in 4%. Conclusion: Mexican OC BRCA1-Del ex9-12 patients treated with olaparib had a significant increase in PFS regardless of the line of treatment compared to other mutations in BRCA.
Collapse
Affiliation(s)
- Dolores Gallardo-Rincón
- Ovarian and Endometrial Cancer Program (COE), Instituto Nacional de Cancerología (INCan), Mexico City, Mexico
- Department of Medical Oncology, Instituto Nacional de Cancerología (INCan), Mexico City, Mexico
- *Correspondence: Dolores Gallardo-Rincón,
| | - Edgar Montes-Servín
- Ovarian and Endometrial Cancer Program (COE), Instituto Nacional de Cancerología (INCan), Mexico City, Mexico
| | - Gabriela Alamilla-García
- Ovarian and Endometrial Cancer Program (COE), Instituto Nacional de Cancerología (INCan), Mexico City, Mexico
- Department of Medical Oncology, Instituto Nacional de Cancerología (INCan), Mexico City, Mexico
| | - Elizabeth Montes-Servín
- Ovarian and Endometrial Cancer Program (COE), Instituto Nacional de Cancerología (INCan), Mexico City, Mexico
| | - Antonio Bahena-González
- Ovarian and Endometrial Cancer Program (COE), Instituto Nacional de Cancerología (INCan), Mexico City, Mexico
- Department of Medical Oncology, Instituto Nacional de Cancerología (INCan), Mexico City, Mexico
| | - Lucely Cetina-Pérez
- Department of Clinical Research and Medical Oncology, Instituto Nacional de Cancerología (INCan), Mexico City, Mexico
- Cervical Cancer Program (Micaela), Instituto Nacional de Cancerología (INCan), Mexico City, Mexico
| | - Flavia Morales Vásquez
- Department of Medical Oncology, Instituto Nacional de Cancerología (INCan), Mexico City, Mexico
| | - Claudia Cano-Blanco
- Department of Medical Oncology, Instituto Nacional de Cancerología (INCan), Mexico City, Mexico
| | - Jaime Coronel-Martínez
- Department of Medical Oncology, Instituto Nacional de Cancerología (INCan), Mexico City, Mexico
| | - Ernesto González-Ibarra
- Ovarian and Endometrial Cancer Program (COE), Instituto Nacional de Cancerología (INCan), Mexico City, Mexico
| | - Raquel Espinosa-Romero
- Ovarian and Endometrial Cancer Program (COE), Instituto Nacional de Cancerología (INCan), Mexico City, Mexico
- Department of Medical Oncology, Instituto Nacional de Cancerología (INCan), Mexico City, Mexico
| | - Rosa María Alvarez-Gómez
- Department of Clinical Research and Medical Oncology, Instituto Nacional de Cancerología (INCan), Mexico City, Mexico
- Hereditary Cancer Clinic, Instituto Nacional de Cancerología (INCan), Mexico City, Mexico
| | - Abraham Pedroza-Torres
- Hereditary Cancer Clinic, Instituto Nacional de Cancerología (INCan), Mexico City, Mexico
- Catedrático CONACYT, Instituto Nacional de Cancerología, Mexico City, Mexico
| | - Denisse Castro-Eguiluz
- Cervical Cancer Program (Micaela), Instituto Nacional de Cancerología (INCan), Mexico City, Mexico
- Catedrático CONACYT, Instituto Nacional de Cancerología, Mexico City, Mexico
| |
Collapse
|
33
|
Jimenez-Sainz J, Krysztofiak A, Garbarino J, Rogers F, Jensen RB. The Pathogenic R3052W BRCA2 Variant Disrupts Homology-Directed Repair by Failing to Localize to the Nucleus. Front Genet 2022; 13:884210. [PMID: 35711920 PMCID: PMC9197106 DOI: 10.3389/fgene.2022.884210] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 04/22/2022] [Indexed: 12/04/2022] Open
Abstract
The BRCA2 germline missense variant, R3052W, resides in the DNA binding domain and has been previously classified as a pathogenic allele. In this study, we sought to determine how R3052W alters the cellular functions of BRCA2 in the DNA damage response. The BRCA2 R3052W mutated protein exacerbates genome instability, is unable to rescue homology-directed repair, and fails to complement cell survival following exposure to PARP inhibitors and crosslinking drugs. Surprisingly, despite anticipated defects in DNA binding or RAD51-mediated DNA strand exchange, the BRCA2 R3052W protein mislocalizes to the cytoplasm precluding its ability to perform any DNA repair functions. Rather than acting as a simple loss-of-function mutation, R3052W behaves as a dominant negative allele, likely by sequestering RAD51 in the cytoplasm.
Collapse
Affiliation(s)
| | | | | | | | - Ryan B. Jensen
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
34
|
Azzollini J, Vingiani A, Agnelli L, Tamborini E, Perrone F, Conca E, Capone I, Busico A, Peissel B, Rosina E, Ducceschi M, Mantiero M, Lopez S, Raspagliesi F, Niger M, Duca M, Damian S, Proto C, de Braud F, Pruneri G, Manoukian S. Management of BRCA Tumour Testing in an Integrated Molecular Tumour Board Multidisciplinary Model. Front Oncol 2022; 12:857515. [PMID: 35463374 PMCID: PMC9026437 DOI: 10.3389/fonc.2022.857515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 03/17/2022] [Indexed: 11/13/2022] Open
Abstract
Tumour testing of the BRCA1/2 genes is routinely performed in patients with different cancer histological subtypes. To accurately identify patients with tumour-detected germline pathogenic variants (PVs) is a relevant issue currently under investigation. This study aims at evaluating the performance of the tumour-to-germline diagnostic flowchart model defined at our Institutional Molecular Tumour Board (MTB). Results from tumour BRCA sequencing of 641 consecutive unselected cancer patients were discussed during weekly MTB meetings with the early involvement of clinical geneticists for appropriate referral to genetic counselling. The overall tumour detection rate of BRCA1/2 PVs was 8.7% (56/641), ranging from 24.4% (31/127) in high-grade ovarian cancer to 3.9% (12/304) in tumours not associated with germline BRCA1/2 PVs. Thirty-seven patients with PVs (66%) were evaluated by a clinical geneticist, and in 24 of them (64.9%), germline testing confirmed the presence of the PV in blood. Nine of these patients (37.5%) were not eligible for germline testing according to the criteria in use at our institution. Cascade testing was subsequently performed on 18 relatives. The tumour-to-germline diagnostic pipeline, developed in the framework of our institutional MTB, compared with guideline-based germline testing following genetic counselling, proved to be effective in identifying a higher number of germline BRCA PVs carriers.
Collapse
Affiliation(s)
- Jacopo Azzollini
- Unit of Medical Genetics, Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Andrea Vingiani
- Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.,Oncology and Hemato-oncology Department, University of Milan, Milan, Italy
| | - Luca Agnelli
- Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.,Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Elena Tamborini
- Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Federica Perrone
- Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Elena Conca
- Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Iolanda Capone
- Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Adele Busico
- Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Bernard Peissel
- Unit of Medical Genetics, Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Erica Rosina
- Unit of Medical Genetics, Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Monika Ducceschi
- Department of Gynecologic Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Mara Mantiero
- Department of Gynecologic Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Salvatore Lopez
- Department of Gynecologic Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Francesco Raspagliesi
- Department of Gynecologic Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Monica Niger
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Matteo Duca
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Silvia Damian
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Claudia Proto
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Filippo de Braud
- Oncology and Hemato-oncology Department, University of Milan, Milan, Italy.,Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Giancarlo Pruneri
- Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.,Oncology and Hemato-oncology Department, University of Milan, Milan, Italy
| | - Siranoush Manoukian
- Unit of Medical Genetics, Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
35
|
Klyukina LA, Sosnova EA, Ishchenko AA. Carcinogenic Potential Of Ovulation Stimulation In BRCA1/2 Gene Mutation Carriers In Assisted Reproductive Technology Programs. RUSSIAN OPEN MEDICAL JOURNAL 2022. [DOI: 10.15275/rusomj.2022.0116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Mutations of the BRCA1/2 genes constitute a fundamental and independent risk factor in the genesis of both breast cancer and ovarian cancer. The specifics of the infertility treatment effect on the risk of developing cancer in carriers of mutations in the BRCA1/2 genes remain unclear and require a comprehensive investigation. In this review, we analyzed published sources on the possible relationship between the infertility treatment and the risk of ovarian and breast cancer in BRCA1/2 mutation carriers.
Collapse
Affiliation(s)
| | - Elena A. Sosnova
- I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Anton A. Ishchenko
- Federal Budgetary Institution Medical and Rehabilitation Center, Moscow, Russia
| |
Collapse
|
36
|
Toward More Comprehensive Homologous Recombination Deficiency Assays in Ovarian Cancer, Part 1: Technical Considerations. Cancers (Basel) 2022; 14:cancers14051132. [PMID: 35267439 PMCID: PMC8909526 DOI: 10.3390/cancers14051132] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/19/2022] [Accepted: 02/22/2022] [Indexed: 02/05/2023] Open
Abstract
Simple Summary High-grade serous ovarian cancer (HGSOC) is the most frequent and lethal form of ovarian cancer and is associated with homologous recombination deficiency (HRD) in 50% of cases. This specific alteration is associated with sensitivity to PARP inhibitors (PARPis). Despite vast prognostic improvements due to PARPis, current molecular assays assessing HRD status suffer from several limitations, and there is an urgent need for a more accurate evaluation. In these companion reviews (Part 1: Technical considerations; Part 2: Medical perspectives), we develop an integrative review to provide physicians and researchers involved in HGSOC management with a holistic perspective, from translational research to clinical applications. Abstract High-grade serous ovarian cancer (HGSOC), the most frequent and lethal form of ovarian cancer, exhibits homologous recombination deficiency (HRD) in 50% of cases. In addition to mutations in BRCA1 and BRCA2, which are the best known thus far, defects can also be caused by diverse alterations to homologous recombination-related genes or epigenetic patterns. HRD leads to genomic instability (genomic scars) and is associated with PARP inhibitor (PARPi) sensitivity. HRD is currently assessed through BRCA1/2 analysis, which produces a genomic instability score (GIS). However, despite substantial clinical achievements, FDA-approved companion diagnostics (CDx) based on GISs have important limitations. Indeed, despite the use of GIS in clinical practice, the relevance of such assays remains controversial. Although international guidelines include companion diagnostics as part of HGSOC frontline management, they also underscore the need for more powerful and alternative approaches for assessing patient eligibility to PARP inhibitors. In these companion reviews, we review and present evidence to date regarding HRD definitions, achievements and limitations in HGSOC. Part 1 is dedicated to technical considerations and proposed perspectives that could lead to a more comprehensive and dynamic assessment of HR, while Part 2 provides a more integrated approach for clinicians.
Collapse
|
37
|
Quesada S, Fabbro M, Solassol J. Toward More Comprehensive Homologous Recombination Deficiency Assays in Ovarian Cancer Part 2: Medical Perspectives. Cancers (Basel) 2022; 14:cancers14041098. [PMID: 35205846 PMCID: PMC8870335 DOI: 10.3390/cancers14041098] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/19/2022] [Accepted: 02/20/2022] [Indexed: 02/07/2023] Open
Abstract
Simple Summary High-grade serous ovarian cancer (HGSOC—the most frequent and aggressive form of ovarian cancer) represents an important challenge for clinicians. Half of HGSOC cases exhibit homologous recombination deficiency (HRD), mainly through alterations in BRCA1 and BRCA2. This leads to sensitivity to PARP inhibitors, a novel class of breakthrough molecules that improved HGSOC prognoses. To date, three companion diagnostic assays have received FDA approval for the evaluation of HRD status, but their use remains controversial. In this companion review (Part 1: Technical considerations; Part 2: Medical perspectives), we develop an integrative perspective, from translational research to clinical application, that could help physicians and researchers manage HGSOC. Abstract High-grade serous ovarian cancer (HGSOC) is the most frequent and aggressive form of ovarian cancer, representing an important challenge for clinicians. Half of HGSOC cases have homologous recombination deficiency (HRD), which has specific causes (mainly alterations in BRCA1/2, but also other alterations encompassed by the BRCAness concept) and consequences, both at molecular (e.g., genomic instability) and clinical (e.g., sensitivity to PARP inhibitor) levels. Based on its prevalence and clinical impact, HRD status merits investigation. To date, three PARP inhibitors have received FDA/EMA approval. For some approvals, the presence of specific molecular alterations is required. Three companion diagnostic (CDx) assays based on distinct technical and medical considerations have received FDA approval to date. However, their use remains controversial due to their technical and medical limitations. In this companion and integrated review, we take a “bench-to-bedside” perspective on HRD definition and evaluation in the context of HGSOC. Part 1 of the review adopts a molecular perspective regarding technical considerations and the development of CDx. Part 2 focuses on the clinical impact of HRD evaluation, primarily through currently validated CDx and prescription of PARP inhibitors, outlining achievements, limitations and medical perspectives.
Collapse
Affiliation(s)
- Stanislas Quesada
- Medical Oncology Department, Institut Régional du Cancer de Montpellier (ICM), 34298 Montpellier, France;
- Faculty of Medicine, University of Montpellier, 34090 Montpellier, France;
- Correspondence:
| | - Michel Fabbro
- Medical Oncology Department, Institut Régional du Cancer de Montpellier (ICM), 34298 Montpellier, France;
- Montpellier Research Cancer Institute (IRCM), Institut National de la Santé et de la Recherche Médicale (INSERM) U1194, University of Montpellier, 34298 Montpellier, France
| | - Jérôme Solassol
- Faculty of Medicine, University of Montpellier, 34090 Montpellier, France;
- Montpellier Research Cancer Institute (IRCM), Institut National de la Santé et de la Recherche Médicale (INSERM) U1194, University of Montpellier, 34298 Montpellier, France
- Department of Pathology and Onco-Biology, Centre Hospitalier Universitaire (CHU) Montpellier, 34295 Montpellier, France
| |
Collapse
|
38
|
Liu H, Zhang Z, Chen L, Pang J, Wu H, Liang Z. Next-Generation Sequencing Reveals a Very Low Prevalence of Deleterious Mutations of Homologous Recombination Repair Genes and Homologous Recombination Deficiency in Ovarian Clear Cell Carcinoma. Front Oncol 2022; 11:798173. [PMID: 35096598 PMCID: PMC8791260 DOI: 10.3389/fonc.2021.798173] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 12/14/2021] [Indexed: 12/31/2022] Open
Abstract
Ovarian clear cell carcinoma (OCCC) is aggressive and drug-resistant. The prevalence of homologous recombination repair (HRR) gene mutations and homologous recombination deficiency (HRD) remains largely unknown. It is also not clear whether the commonly used molecular-based classification for endometrial carcinoma (EC) is potentially applicable in OCCC. In this study, surgically resected samples were collected from 44 patients with OCCC. Genomic alterations were determined using next-generation sequencing. HRD was estimated by genomic instability. Of 44 patients with OCCC, two (4.5%) harbored likely pathogenic mutations in HRR genes. Notably, no pathogenic or likely pathogenic mutations were found in BRCA1/2. A total of 24 variants of uncertain significance (VUS) in HRR-related genes occurred in 18 (40.9%) patients. HRD was observed in only one case (2.3%). In addition, TP53 mutation and microsatellite instability-high (MSI-H) were identified in three patients (6.8%) and in one patient (2.3%), respectively. TP53 mutation was significantly associated with disease-free survival and overall survival. No POLE mutations were found. In conclusion, our results revealed a very low prevalence of HRR gene mutations and HRD in OCCC. Moreover, TP53 mutations and MSI-H are uncommon, while POLE mutations are extremely rare in OCCC. Our findings indicate that the evaluation of HRR gene mutations, HRD status, POLE mutations, and MSI-H may have limited clinical significance for OCCC treatment and prognostic stratification.
Collapse
Affiliation(s)
- Hangqi Liu
- Department of Pathology, State Key Laboratory of Complex Severe and Rare Disease, Molecular Pathology Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhiwen Zhang
- Department of Pathology, State Key Laboratory of Complex Severe and Rare Disease, Molecular Pathology Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Longyun Chen
- Department of Pathology, State Key Laboratory of Complex Severe and Rare Disease, Molecular Pathology Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Junyi Pang
- Department of Pathology, State Key Laboratory of Complex Severe and Rare Disease, Molecular Pathology Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Huanwen Wu
- Department of Pathology, State Key Laboratory of Complex Severe and Rare Disease, Molecular Pathology Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhiyong Liang
- Department of Pathology, State Key Laboratory of Complex Severe and Rare Disease, Molecular Pathology Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
39
|
Wu N, Wei L, Li L, Li F, Yu J, Liu J. Perspectives on the role of breast cancer susceptibility gene in breast cancer. Int J Clin Oncol 2022; 27:495-511. [PMID: 35064849 DOI: 10.1007/s10147-021-02098-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 11/26/2021] [Indexed: 11/27/2022]
Abstract
PURPOSE Breast cancer susceptibility gene 1/2 can repair damaged DNA through homologous recombination. Besides, the local immune microenvironment of breast cancer is closely linked to the prognosis of patients. But the relationship of breast cancer susceptibility gene 1/2 expression and local immunosuppressive microenvironment in breast cancer is not clear. The aim of this study was to discuss the correlation between them. METHODS The fresh primary breast tumors and paired normal tissues of 156 cases of breast cancer patients as well as peripheral blood of 156 cases among them in Tianjin Medical University Cancer Institute and Hospital from January 2014 to October 2018 were collected. The association between breast cancer susceptibility gene 1/2 germline mutation and immune status of microenvironment in situ was analyzed. RESULTS The results indicated that the germline mutation of breast cancer susceptibility gene 1/2 was inconsistent with the breast cancer susceptibility gene 1/2 protein expression, and the proportion of immune cells in patients with negative expression of breast cancer susceptibility gene 1/2 protein was higher than patients with positive expression of breast cancer susceptibility gene 1/2 protein (p < 0.05). And the expression of programmed cell death protein 1, cytotoxic T-Lymphocyte Antigen 4, programmed death ligand-1 of CD3+ T cells in patients with negative expression of breast cancer susceptibility gene 1/2 protein was higher than patients with positive expression of breast cancer susceptibility gene 1/2 protein (p < 0.05). The breast cancer susceptibility gene 1 protein expression was significantly correlated with family history of breast cancer patients (p = 0.006), local lymph node metastases (p = 0.001), and TNM staging (p ≤ 0.001). The breast cancer susceptibility gene 2 protein expression was significantly related to local lymph node metastases (p ≤ 0.001), III stage rate(p = 0.003) and molecular subtyping (p ≤ 0.001). Besides, the 5 years disease free survival was worse for G1 group and pathological III stage patients than other groups and other TNM stage patients. CONCLUSION In short, the immune therapy may be a potential therapy method for breast cancer patients with negative expression of breast cancer susceptibility gene 1/2 protein.
Collapse
Affiliation(s)
- Nan Wu
- Cancer Prevention Center, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tiyuanbei, Huanhuxi Rode, Hexi District, Tianjin, 300060, China
| | - Lijuan Wei
- Cancer Prevention Center, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tiyuanbei, Huanhuxi Rode, Hexi District, Tianjin, 300060, China
| | - Lijuan Li
- Cancer Prevention Center, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tiyuanbei, Huanhuxi Rode, Hexi District, Tianjin, 300060, China
| | - Fangxuan Li
- Cancer Prevention Center, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tiyuanbei, Huanhuxi Rode, Hexi District, Tianjin, 300060, China
| | - Jinpu Yu
- The Molecular Diagnostics, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tiyuanbei, Huanhuxi Rode, Hexi District, Tianjin, 300060, China.
| | - Juntian Liu
- Cancer Prevention Center, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tiyuanbei, Huanhuxi Rode, Hexi District, Tianjin, 300060, China.
- The Second Department of Breast Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China.
| |
Collapse
|
40
|
Chiang YC, Lin PH, Cheng WF. Homologous Recombination Deficiency Assays in Epithelial Ovarian Cancer: Current Status and Future Direction. Front Oncol 2021; 11:675972. [PMID: 34722237 PMCID: PMC8551835 DOI: 10.3389/fonc.2021.675972] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 09/17/2021] [Indexed: 01/02/2023] Open
Abstract
Epithelial ovarian cancer (EOC) patients are generally diagnosed at an advanced stage, usually relapse after initial treatments, which include debulking surgery and adjuvant platinum-based chemotherapy, and eventually have poor 5-year survival of less than 50%. In recent years, promising survival benefits from maintenance therapy with poly(ADP-ribose) polymerase (PARP) inhibitor (PARPi) has changed the management of EOC in newly diagnosed and recurrent disease. Identification of BRCA mutations and/or homologous recombination deficiency (HRD) is critical for selecting patients for PARPi treatment. However, the currently available HRD assays are not perfect predictors of the clinical response to PARPis in EOC patients. In this review, we introduce the concept of synthetic lethality, the rationale of using PARPi when HRD is present in tumor cells, the clinical trials of PARPi incorporating the HRD assays for EOC, the current HRD assays, and other HRD assays in development.
Collapse
Affiliation(s)
- Ying-Cheng Chiang
- Department of Obstetrics and Gynecology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Po-Han Lin
- Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan.,Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Wen-Fang Cheng
- Department of Obstetrics and Gynecology, College of Medicine, National Taiwan University, Taipei, Taiwan.,Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.,Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
41
|
Paik ES, Heo EJ, Choi CH, Kim JH, Kim JW, Kim YM, Park SY, Lee JW, Kim JW, Kim BG. Prevalence and clinical characterization of BRCA1 and BRCA2 mutations in Korean patients with epithelial ovarian cancer. Cancer Sci 2021; 112:5055-5067. [PMID: 34657357 PMCID: PMC8645710 DOI: 10.1111/cas.15166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 10/11/2021] [Accepted: 10/12/2021] [Indexed: 12/24/2022] Open
Abstract
This study was performed to investigate the prevalence, clinical characteristics, and treatment response according to BRCA1 and BRCA2 (BRCA) mutations in Korean patients with epithelial ovarian cancer (EOC). Two‐hundred and ninety‐eight Korean women diagnosed with high‐grade serous and/or endometrioid EOC from 2010 to 2015 were tested for germline and 86 specimens for somatic BRCA mutations, regardless of the family history. Clinical characteristics including survival outcomes were compared in patients with and without BRCA mutations (NCT02963688). A total of 43 different germline BRCA mutations were identified in 78 patients among 298 patients (26.2%). Somatic BRCA mutations were identified in 11 (12.8%) patients among patients without germline BRCA mutations. Haplotype analysis demonstrated no founder mutations in our Korean patient cohort. Insignificant differences in age at diagnosis, primary site, and residual disease after surgery were observed between patients with and without BRCA mutations. In multivariate analysis for overall survival (OS), the presence of BRCA mutation was significantly associated with OS (P = .049) in addition to platinum sensitivity (P < .001), indicating it is an independent prognostic factor for survival regardless of platinum sensitivity to first‐line chemotherapy. In addition, a higher response rate to subsequent chemotherapy after recurrence was observed in EOC patients with BRCA mutations resulting in better OS. In the current study, the prevalence of BRCA mutations in Korean patients with EOC was higher than previously reported in other ethnic groups. We demonstrated characteristics and treatment response in Korean EOC patients with BRCA mutations. These findings may provide valuable information to be considered in future clinical trials including Asian patients.
Collapse
Affiliation(s)
- E Sun Paik
- Department of Obstetrics and Gynecology, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Eun Jin Heo
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Chel Hun Choi
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jae-Hoon Kim
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University School of Medicine, Seoul, Korea
| | - Jae-Weon Kim
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul National University School of Medicine, Seoul, Korea
| | - Yong-Man Kim
- Department of Obstetrics and Gynecology, Asan Medical Center, Ulsan University School of Medicine, Seoul, Korea
| | - Sang-Yoon Park
- Center for Uterine Cancer, Research Institute and Hospital, National Cancer Center, Goyang, Korea
| | - Jeong-Won Lee
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jong-Won Kim
- Department of Clinical Laboratory Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Byoung-Gie Kim
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
42
|
Nesic K, Kondrashova O, Hurley RM, McGehee CD, Vandenberg CJ, Ho GY, Lieschke E, Dall G, Bound N, Shield-Artin K, Radke M, Musafer A, Chai ZQ, Eftekhariyan Ghamsari MR, Harrell MI, Kee D, Olesen I, McNally O, Traficante N, Cancer Study AO, DeFazio A, Bowtell DDL, Swisher EM, Weroha SJ, Nones K, Waddell N, Kaufmann SH, Dobrovic A, Wakefield MJ, Scott CL. Acquired RAD51C promoter methylation loss causes PARP inhibitor resistance in high grade serous ovarian carcinoma. Cancer Res 2021; 81:4709-4722. [PMID: 34321239 DOI: 10.1158/0008-5472.can-21-0774] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 05/10/2021] [Accepted: 07/07/2021] [Indexed: 11/16/2022]
Abstract
In high-grade serous ovarian carcinoma (HGSC), deleterious mutations in DNA repair gene RAD51C are established drivers of defective homologous recombination and are emerging biomarkers of PARP inhibitor (PARPi) sensitivity. RAD51C promoter methylation (meRAD51C) is detected at similar frequencies to mutations, yet its effects on PARPi responses remain unresolved. In this study, three HGSC patient-derived xenograft (PDX) models with methylation at most or all examined CpG sites in the RAD51C promoter show responses to PARPi. Both complete and heterogeneous methylation patterns were associated with RAD51C gene silencing and homologous recombination deficiency (HRD). PDX models lost meRAD51C following treatment with PARPi rucaparib or niraparib, where a single unmethylated copy of RAD51C was sufficient to drive PARPi resistance. Genomic copy number profiling of one of the PDX models using SNP arrays revealed that this resistance was acquired independently in two genetically distinct lineages. In a cohort of 11 patients with RAD51C-methylated HGSC, various patterns of meRAD51C were associated with genomic 'scarring', indicative of HRD history, but exhibited no clear correlations with clinical outcome. Differences in methylation stability under treatment pressure were also observed between patients, where one HGSC was found to maintain meRAD51C after 6 lines of therapy (4 platinum-based), whilst another HGSC sample was found to have heterozygous meRAD51C and elevated RAD51C gene expression (relative to homozygous meRAD51C controls) after only neo-adjuvant chemotherapy. As meRAD51C loss in a single gene copy was sufficient to cause PARPi resistance in PDX, methylation zygosity should be carefully assessed in previously treated patients when considering PARPi therapy.
Collapse
Affiliation(s)
- Ksenija Nesic
- Cancer Biology and Stem Cells, Walter and Eliza Hall Institute of Medical Research
| | - Olga Kondrashova
- Genetics and Computational Biology, QIMR Berghofer Medical Research Institute
| | | | | | | | - Gwo-Yaw Ho
- Stem Cells and Cancer Division, Walter and Eliza Hall Institute of Medical Research
| | - Elizabeth Lieschke
- Stem Cells and Cancer Division, Walter and Eliza Hall Institute of Medical Research
| | | | | | - Kristy Shield-Artin
- Stem Cells and Cancer Division, Walter and Eliza Hall Institute of Medical Research
| | - Marc Radke
- Obstetrics and Gynecology, University of Washington Medical Center
| | - Ashan Musafer
- Translational Genomics and Epigenomics Group, Olivia Newton-John Cancer Wellness & Research Centre
| | - Zi Qing Chai
- Olivia Newton-John Cancer Wellness & Research Centre
| | | | - Maria I Harrell
- Obstetrics and Gynecology, University of Washington Medical Center
| | | | | | - Orla McNally
- Department of Obstetrics and Gynaecology, Royal Women's Hospital
| | - Nadia Traficante
- Cancer Genetics and Genomics Laboratory and Australian Ovarian Cancer Study, Peter MacCallum Cancer Centre
| | | | - Anna DeFazio
- Centre for Cancer Research, University of Sydney, Westmead Institute for Medical Research
| | - David D L Bowtell
- Cancer Genetics and Genomics Laboratory and Austrialian Ovarian Cancer Study, Peter MacCallum Cancer Centre
| | | | | | - Katia Nones
- Cell and Molecular Biology, QIMR Berghofer Medical Research Institute
| | - Nicola Waddell
- Medical Genomics Laboratory, QIMR Berghofer Medical Research Institute
| | | | - Alexander Dobrovic
- Translational Genomics and Epigenomics Laboratory, University of Melbourne
| | - Matthew J Wakefield
- Stem Cells and Cancer Division, Walter and Eliza Hall Institute of Medical Research
| | - Clare L Scott
- Cancer Biology and Stem Cells Division Division, Walter and Eliza Hall Institute of Medical Research
| |
Collapse
|
43
|
Hurley RM, McGehee CD, Nesic K, Correia C, Weiskittel TM, Kelly RL, Venkatachalam A, Hou X, Pathoulas NM, Meng XW, Kondrashova O, Radke MR, Schneider PA, Flatten KS, Peterson KL, Becker MA, Wong EM, Southey MS, Dobrovic A, Lin KK, Harding TC, McNeish I, Ross CA, Wagner JM, Wakefield MJ, Scott CL, Haluska P, Wahner Hendrickson AE, Karnitz LM, Swisher EM, Li H, Weroha SJ, Kaufmann SH. Characterization of a RAD51C-silenced high-grade serous ovarian cancer model during development of PARP inhibitor resistance. NAR Cancer 2021; 3:zcab028. [PMID: 34316715 PMCID: PMC8271218 DOI: 10.1093/narcan/zcab028] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 05/28/2021] [Accepted: 06/22/2021] [Indexed: 12/13/2022] Open
Abstract
Acquired PARP inhibitor (PARPi) resistance in BRCA1- or BRCA2-mutant ovarian cancer often results from secondary mutations that restore expression of functional protein. RAD51C is a less commonly studied ovarian cancer susceptibility gene whose promoter is sometimes methylated, leading to homologous recombination (HR) deficiency and PARPi sensitivity. For this study, the PARPi-sensitive patient-derived ovarian cancer xenograft PH039, which lacks HR gene mutations but harbors RAD51C promoter methylation, was selected for PARPi resistance by cyclical niraparib treatment in vivo. PH039 acquired PARPi resistance by the third treatment cycle and grew through subsequent treatment with either niraparib or rucaparib. Transcriptional profiling throughout the course of resistance development showed widespread pathway level changes along with a marked increase in RAD51C mRNA, which reflected loss of RAD51C promoter methylation. Analysis of ovarian cancer samples from the ARIEL2 Part 1 clinical trial of rucaparib monotherapy likewise indicated an association between loss of RAD51C methylation prior to on-study biopsy and limited response. Interestingly, the PARPi resistant PH039 model remained platinum sensitive. Collectively, these results not only indicate that PARPi treatment pressure can reverse RAD51C methylation and restore RAD51C expression, but also provide a model for studying the clinical observation that PARPi and platinum sensitivity are sometimes dissociated.
Collapse
Affiliation(s)
- Rachel M Hurley
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905 USA
| | - Cordelia D McGehee
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905 USA
| | - Ksenija Nesic
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
| | - Cristina Correia
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905 USA
| | - Taylor M Weiskittel
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905 USA
| | - Rebecca L Kelly
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905 USA
| | - Annapoorna Venkatachalam
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905 USA
| | - Xiaonan Hou
- Division of Medical Oncology, Mayo Clinic, Rochester, MN 55905 USA
| | | | - X Wei Meng
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905 USA
| | - Olga Kondrashova
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
| | - Marc R Radke
- Department of Obstetrics & Gynecology, University of Washington, Seattle, WA 98195, USA
| | | | - Karen S Flatten
- Division of Oncology Research, Mayo Clinic, Rochester, MN 55905 USA
| | - Kevin L Peterson
- Division of Oncology Research, Mayo Clinic, Rochester, MN 55905 USA
| | - Marc A Becker
- Division of Medical Oncology, Mayo Clinic, Rochester, MN 55905 USA
| | - Ee Ming Wong
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Victoria 3800, Australia
| | - Melissa S Southey
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Victoria 3800, Australia
| | - Alexander Dobrovic
- University of Melbourne Department of Surgery, Austin Hospital, Heidelberg, Victoria 3084, Australia
| | - Kevin K Lin
- Clovis Oncology, San Francisco, CA 94158, USA
| | | | - Iain McNeish
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, Hammersmith Campus, London, W12 0NN United Kingdom
| | - Christian A Ross
- Division of Information Technology, Mayo Clinic, Rochester, MN 55905, USA
| | - Jill M Wagner
- Division of Medical Oncology, Mayo Clinic, Rochester, MN 55905 USA
| | - Matthew J Wakefield
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
| | - Clare L Scott
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
| | - Paul Haluska
- Division of Medical Oncology, Mayo Clinic, Rochester, MN 55905 USA
| | | | - Larry M Karnitz
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905 USA
| | - Elizabeth M Swisher
- Department of Obstetrics & Gynecology, University of Washington, Seattle, WA 98195, USA
| | - Hu Li
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905 USA
| | - S John Weroha
- Division of Medical Oncology, Mayo Clinic, Rochester, MN 55905 USA
| | - Scott H Kaufmann
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905 USA
| |
Collapse
|
44
|
Kalachand RD, Stordal B, Madden S, Chandler B, Cunningham J, Goode EL, Ruscito I, Braicu EI, Sehouli J, Ignatov A, Yu H, Katsaros D, Mills GB, Lu KH, Carey MS, Timms KM, Kupryjanczyk J, Rzepecka IK, Podgorska A, McAlpine JN, Swisher EM, Bernards SS, O'Riain C, O'Toole S, O'Leary JJ, Bowtell DD, Thomas DM, Prieske K, Joosse SA, Woelber L, Chaudhry P, Häfner N, Runnebaum IB, Hennessy BT. BRCA1 Promoter Methylation and Clinical Outcomes in Ovarian Cancer: An Individual Patient Data Meta-Analysis. J Natl Cancer Inst 2021; 112:1190-1203. [PMID: 32413141 DOI: 10.1093/jnci/djaa070] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 04/23/2020] [Accepted: 05/11/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND BRCA1 methylation has been associated with homologous recombination deficiency, a biomarker of platinum sensitivity. Studies evaluating BRCA1-methylated tubal and ovarian cancer (OC) do not consistently support improved survival following platinum chemotherapy. We examine the characteristics of BRCA1-methylated OC in a meta-analysis of individual participant data. METHODS Data of 2636 participants across 15 studies were analyzed. BRCA1-methylated tumors were defined according to their original study. Associations between BRCA1 methylation and clinicopathological characteristics were evaluated. The effects of methylation on overall survival (OS) and progression-free survival (PFS) were examined using mixed-effects models. All statistical tests were 2-sided. RESULTS 430 (16.3%) tumors were BRCA1-methylated. BRCA1 methylation was associated with younger age and advanced-stage, high-grade serous OC. There were no survival differences between BRCA1-methylated and non-BRCA1-methylated OC (median PFS = 20.0 vs 18.5 months, hazard ratio [HR] = 1.01, 95% CI = 0.87 to 1.16; P = .98; median OS = 46.6 vs 48.0 months, HR = 1.02, 95% CI = 0.87 to 1.18; P = .96). Where BRCA1/2 mutations were evaluated (n = 1248), BRCA1 methylation displayed no survival advantage over BRCA1/2-intact (BRCA1/2 wild-type non-BRCA1-methylated) OC. Studies used different methods to define BRCA1 methylation. Where BRCA1 methylation was determined using methylation-specific polymerase chain reaction and gel electrophoresis (n = 834), it was associated with improved survival (PFS: HR = 0.80, 95% CI = 0.66 to 0.97; P = .02; OS: HR = 0.80, 95% CI = 0.63 to 1.00; P = .05) on mixed-effects modeling. CONCLUSION BRCA1-methylated OC displays similar clinicopathological features to BRCA1-mutated OC but is not associated with survival. Heterogeneity within BRCA1 methylation assays influences associations. Refining these assays may better identify cases with silenced BRCA1 function and improved patient outcomes.
Collapse
Affiliation(s)
- Roshni D Kalachand
- Medical Oncology Group, Department of Molecular Medicine, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | - Britta Stordal
- Department of Natural Sciences, Middlesex University, Hendon, London NW4 4BT, UK
| | - Stephen Madden
- Data Science Centre, Royal College of Surgeons in Ireland, Beaux Lane House, Dublin, Ireland
| | - Benjamin Chandler
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Julie Cunningham
- Division of Experimental Pathology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Ellen L Goode
- Division of Epidemiology, Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Ilary Ruscito
- Department of Gynecology, European Competence Center for Ovarian Cancer, Campus Virchow Klinikum, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany.,Cell Therapy Unit and Laboratory of Tumor Immunology, Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Elena I Braicu
- Department of Gynecology, European Competence Center for Ovarian Cancer, Campus Virchow Klinikum, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Jalid Sehouli
- Department of Gynecology, European Competence Center for Ovarian Cancer, Campus Virchow Klinikum, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Atanas Ignatov
- Department of Gynecology and Obstetrics, University Medical Center, Regensburg, Germany
| | - Herbert Yu
- University of Hawaii Cancer Centre, Honolulu, HI, USA
| | - Dionyssios Katsaros
- AOU Citta della Salute and Department of Surgical Sciences, Gynecologic Oncology, University of Torino, Italy
| | - Gordon B Mills
- Department of Cell, Development and Cancer Biology Knight Cancer Institute, Oregon Health and Sciences University, Portland, OR, USA
| | - Karen H Lu
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mark S Carey
- Division of Gynecologic Oncology, Department of Obstetrics and Gynaecology, University of British Columbia, Vancouver, British Columbia, Canada
| | | | - Jolanta Kupryjanczyk
- Department of Pathology and Laboratory Diagnostics, Maria Sklodowska-Curie Institute-Oncology Center, Warsaw, Poland
| | - Iwona K Rzepecka
- Department of Pathology and Laboratory Diagnostics, Maria Sklodowska-Curie Institute-Oncology Center, Warsaw, Poland
| | - Agnieszka Podgorska
- Department of Pathology and Laboratory Diagnostics, Maria Sklodowska-Curie Institute-Oncology Center, Warsaw, Poland
| | - Jessica N McAlpine
- Division of Gynecologic Oncology, Department of Obstetrics and Gynaecology, University of British Columbia, Vancouver, British Columbia, Canada
| | | | | | - Ciaran O'Riain
- Department of Histopathology, Trinity College Dublin, Central Pathology Laboratory, St. James's Hospital, Dublin, Ireland
| | - Sharon O'Toole
- Department of Obstetrics and Gynaecology/Histopathology, Trinity College Dublin, Trinity Centre for Health Sciences, St. James's Hospital, Dublin, Ireland.,Emer Casey Research Laboratory, Molecular Pathology Laboratory, The Coombe Women and Infants University Hospital, Dublin, Ireland
| | - John J O'Leary
- Department of Histopathology, Trinity College Dublin, Central Pathology Laboratory, St. James's Hospital, Dublin, Ireland.,Emer Casey Research Laboratory, Molecular Pathology Laboratory, The Coombe Women and Infants University Hospital, Dublin, Ireland
| | | | - David M Thomas
- Genomic Cancer Medicine, Cancer Division, Garvan Institute of Medical Research, The Kinghorn Cancer Centre, Darlinghurst, Australia
| | - Katharina Prieske
- Department of Gynecology and Gynecologic Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Simon A Joosse
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Linn Woelber
- Department of Gynecology and Gynecologic Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Parvesh Chaudhry
- Department of Radiotherapy, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Norman Häfner
- Department for Gynaecology and Reproductive Medicine, Jena University Hospital-Friedrich Schiller University Jena, Jena, Germany
| | - Ingo B Runnebaum
- Department for Gynaecology and Reproductive Medicine, Jena University Hospital-Friedrich Schiller University Jena, Jena, Germany
| | - Bryan T Hennessy
- Medical Oncology Group, Department of Molecular Medicine, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland.,Department of Medical Oncology, Beaumont Hospital, Dublin, Ireland.,Our Lady of Lourdes Hospital, Drogheda, Ireland
| |
Collapse
|
45
|
Ramarao-Milne P, Kondrashova O, Barry S, Hooper JD, Lee JS, Waddell N. Histone Modifying Enzymes in Gynaecological Cancers. Cancers (Basel) 2021; 13:cancers13040816. [PMID: 33669182 PMCID: PMC7919659 DOI: 10.3390/cancers13040816] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 02/10/2021] [Accepted: 02/11/2021] [Indexed: 12/16/2022] Open
Abstract
Simple Summary Epigenetics is a process that allows genetic control, without the involvement of sequence changes to DNA or genes. In cancer, epigenetics is a key event in tumour development that can alter the expression of cancer driver genes and result in genomic instability. Due to the critical role of epigenetics in malignant transformation, therapies that target these processes have been developed to treat cancer. Here, we provide a summary of the epigenetic changes that have been described in a variety of gynaecological cancers. We then highlight how these changes are being targeted in preclinical models and clinical trials for gynaecological cancers. Abstract Genetic and epigenetic factors contribute to the development of cancer. Epigenetic dysregulation is common in gynaecological cancers and includes altered methylation at CpG islands in gene promoter regions, global demethylation that leads to genome instability and histone modifications. Histones are a major determinant of chromosomal conformation and stability, and unlike DNA methylation, which is generally associated with gene silencing, are amenable to post-translational modifications that induce facultative chromatin regions, or condensed transcriptionally silent regions that decondense resulting in global alteration of gene expression. In comparison, other components, crucial to the manipulation of chromatin dynamics, such as histone modifying enzymes, are not as well-studied. Inhibitors targeting DNA modifying enzymes, particularly histone modifying enzymes represent a potential cancer treatment. Due to the ability of epigenetic therapies to target multiple pathways simultaneously, tumours with complex mutational landscapes affected by multiple driver mutations may be most amenable to this type of inhibitor. Interrogation of the actionable landscape of different gynaecological cancer types has revealed that some patients have biomarkers which indicate potential sensitivity to epigenetic inhibitors. In this review we describe the role of epigenetics in gynaecological cancers and highlight how it may exploited for treatment.
Collapse
Affiliation(s)
- Priya Ramarao-Milne
- Medical Genomics Group, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia; (P.R.-M.); (O.K.); (N.W.)
- Faculty of Medicine, The University of Queensland, Brisbane, QLD 4006, Australia
| | - Olga Kondrashova
- Medical Genomics Group, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia; (P.R.-M.); (O.K.); (N.W.)
| | - Sinead Barry
- Department of Gynaecological Oncology, Mater Hospital Brisbane, Brisbane, QLD 4101, Australia;
- Mater Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, QLD 4102, Australia;
| | - John D. Hooper
- Mater Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, QLD 4102, Australia;
| | - Jason S. Lee
- Faculty of Medicine, The University of Queensland, Brisbane, QLD 4006, Australia
- Epigenetics and Disease Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD 4000, Australia
- Correspondence: ; Tel.: +61-7-38453951
| | - Nicola Waddell
- Medical Genomics Group, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia; (P.R.-M.); (O.K.); (N.W.)
- Faculty of Medicine, The University of Queensland, Brisbane, QLD 4006, Australia
| |
Collapse
|
46
|
Arakelyan A, Melkonyan A, Hakobyan S, Boyarskih U, Simonyan A, Nersisyan L, Nikoghosyan M, Filipenko M, Binder H. Transcriptome Patterns of BRCA1- and BRCA2- Mutated Breast and Ovarian Cancers. Int J Mol Sci 2021; 22:1266. [PMID: 33525353 PMCID: PMC7865215 DOI: 10.3390/ijms22031266] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/06/2021] [Accepted: 01/07/2021] [Indexed: 02/06/2023] Open
Abstract
Mutations in the BRCA1 and BRCA2 genes are known risk factors and drivers of breast and ovarian cancers. So far, few studies have been focused on understanding the differences in transcriptome and functional landscapes associated with the disease (breast vs. ovarian cancers), gene (BRCA1 vs. BRCA2), and mutation type (germline vs. somatic). In this study, we were aimed at systemic evaluation of the association of BRCA1 and BRCA2 germline and somatic mutations with gene expression, disease clinical features, outcome, and treatment. We performed BRCA1/2 mutation centered RNA-seq data analysis of breast and ovarian cancers from the TCGA repository using transcriptome and phenotype "portrayal" with multi-layer self-organizing maps and functional annotation. The results revealed considerable differences in BRCA1- and BRCA2-dependent transcriptome landscapes in the studied cancers. Furthermore, our data indicated that somatic and germline mutations for both genes are characterized by deregulation of different biological functions and differential associations with phenotype characteristics and poly(ADP-ribose) polymerase (PARP)-inhibitor gene signatures. Overall, this study demonstrates considerable variation in transcriptomic landscapes of breast and ovarian cancers associated with the affected gene (BRCA1 vs. BRCA2), as well as the mutation type (somatic vs. germline). These results warrant further investigations with larger groups of mutation carriers aimed at refining the understanding of molecular mechanisms of breast and ovarian cancers.
Collapse
Affiliation(s)
- Arsen Arakelyan
- Group of Bioinformatics, Institute of Molecular Biology National Academy of Sciences of Armenia, 0014 Yerevan, Armenia; (S.H.); (A.S.); (L.N.); (M.N.)
- Institute of Biomedicine and Pharmacy, Russian-Armenian University, 0051 Yerevan, Armenia
| | - Ani Melkonyan
- Laboratory of Human Genomics and Immunomics, Institute of Molecular Biology National Academy of Sciences of Armenia, 0014 Yerevan, Armenia;
| | - Siras Hakobyan
- Group of Bioinformatics, Institute of Molecular Biology National Academy of Sciences of Armenia, 0014 Yerevan, Armenia; (S.H.); (A.S.); (L.N.); (M.N.)
| | - Uljana Boyarskih
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences (SB RAS), 630090 Novosibirsk, Russia; (U.B.); (M.F.)
| | - Arman Simonyan
- Group of Bioinformatics, Institute of Molecular Biology National Academy of Sciences of Armenia, 0014 Yerevan, Armenia; (S.H.); (A.S.); (L.N.); (M.N.)
| | - Lilit Nersisyan
- Group of Bioinformatics, Institute of Molecular Biology National Academy of Sciences of Armenia, 0014 Yerevan, Armenia; (S.H.); (A.S.); (L.N.); (M.N.)
| | - Maria Nikoghosyan
- Group of Bioinformatics, Institute of Molecular Biology National Academy of Sciences of Armenia, 0014 Yerevan, Armenia; (S.H.); (A.S.); (L.N.); (M.N.)
- Institute of Biomedicine and Pharmacy, Russian-Armenian University, 0051 Yerevan, Armenia
| | - Maxim Filipenko
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences (SB RAS), 630090 Novosibirsk, Russia; (U.B.); (M.F.)
| | - Hans Binder
- Interdisciplinary Centre for Bioinformatics, University of Leipzig, D-04107 Leipzig, Germany;
| |
Collapse
|
47
|
Sahnane N, Carnevali I, Formenti G, Casarin J, Facchi S, Bombelli R, Di Lauro E, Memoli D, Salvati A, Rizzo F, Sessa F, Tibiletti MG. BRCA Methylation Testing Identifies a Subset of Ovarian Carcinomas without Germline Variants That Can Benefit from PARP Inhibitor. Int J Mol Sci 2020; 21:E9708. [PMID: 33352687 PMCID: PMC7767143 DOI: 10.3390/ijms21249708] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 12/16/2020] [Accepted: 12/17/2020] [Indexed: 12/15/2022] Open
Abstract
Homologous Recombination Deficiency (HRD) is a frequent feature of high-grade epithelial ovarian carcinoma (EOC), associated with sensitivity to PARP-inhibitors (PARPi). The best characterized causes of HRD in EOCs are germline or somatic mutations in BRCA1 and BRCA2 genes. Although promoter methylation is a well-known mechanism of gene transcriptional repression, few data have been published about BRCA gene methylation in EOCs. In this retrospective study, we quantitatively analyzed by pyrosequencing a selected series of 90 formalin-fixed (FFPE) primary EOCs without BRCA germline mutations. We identified 20/88 (22.7%) EOCs showing BRCA promoter methylation, including 17/88 (19.3%) in BRCA1 and 4/86 (4.6%) in BRCA2 promoters, one of which showing concomitant BRCA1 methylation. Mean methylation levels were 49.6% and 45.8% for BRCA1 and BRCA2, respectively, with methylation levels ≥50% in 10/20 methylated EOCs. Constitutive BRCA methylation was excluded by testing blood-derived DNA. In conclusion, pyrosequencing methylation analysis of BRCA genes is a robust, quantitative and sensitive assay applicable to FFPE samples. Remarkably, a considerable subset of germline BRCA-negative EOCs showed somatic methylation and, likely, HRD. A subpopulation of women with BRCA methylation, even without BRCA mutations, could potentially benefit from PARP-inhibitors; further clinical studies are needed to clarify the predictive role of somatic BRCA methylation of PARP-therapy response.
Collapse
Affiliation(s)
- Nora Sahnane
- SC Anatomia Patologica, Ospedale di Circolo, ASST Settelaghi, 21100 Varese, Italy; (N.S.); (I.C.); (E.D.L.); (F.S.)
- Centro di Ricerca dei Tumori Eredo-Familiari, Dipartimento di Medicina e Chirurgia, University of Insubria, 21100 Varese, Italy
| | - Ileana Carnevali
- SC Anatomia Patologica, Ospedale di Circolo, ASST Settelaghi, 21100 Varese, Italy; (N.S.); (I.C.); (E.D.L.); (F.S.)
- Centro di Ricerca dei Tumori Eredo-Familiari, Dipartimento di Medicina e Chirurgia, University of Insubria, 21100 Varese, Italy
| | - Giorgio Formenti
- Dipartimento di Ostetricia e Ginecologia, ASST Settelaghi, University of Insubria, 21100 Varese, Italy; (G.F.); (J.C.)
| | - Jvan Casarin
- Dipartimento di Ostetricia e Ginecologia, ASST Settelaghi, University of Insubria, 21100 Varese, Italy; (G.F.); (J.C.)
| | - Sofia Facchi
- Dipartimento di Medicina e Chirurgia, University of Insubria, 21100 Varese, Italy; (S.F.); (R.B.)
| | - Raffaella Bombelli
- Dipartimento di Medicina e Chirurgia, University of Insubria, 21100 Varese, Italy; (S.F.); (R.B.)
| | - Eleonora Di Lauro
- SC Anatomia Patologica, Ospedale di Circolo, ASST Settelaghi, 21100 Varese, Italy; (N.S.); (I.C.); (E.D.L.); (F.S.)
| | - Domenico Memoli
- Programma di Genomica Medica, AOU SS Giovanni di Dio e Ruggi d’Aragona Università di Salerno, 84131 Salerno, Italy; (D.M.); (A.S.); (F.R.)
| | - Annamaria Salvati
- Programma di Genomica Medica, AOU SS Giovanni di Dio e Ruggi d’Aragona Università di Salerno, 84131 Salerno, Italy; (D.M.); (A.S.); (F.R.)
| | - Francesca Rizzo
- Programma di Genomica Medica, AOU SS Giovanni di Dio e Ruggi d’Aragona Università di Salerno, 84131 Salerno, Italy; (D.M.); (A.S.); (F.R.)
- Laboratorio di Medicina Molecolare e Genomica Dipartimento di Medicina, Chirurgia e Odontoiatria “Scuola Medica Salernitana” Università di Salerno, Baronissi, 84084 Salerno, Italy
| | - Fausto Sessa
- SC Anatomia Patologica, Ospedale di Circolo, ASST Settelaghi, 21100 Varese, Italy; (N.S.); (I.C.); (E.D.L.); (F.S.)
- Centro di Ricerca dei Tumori Eredo-Familiari, Dipartimento di Medicina e Chirurgia, University of Insubria, 21100 Varese, Italy
- Dipartimento di Medicina e Chirurgia, University of Insubria, 21100 Varese, Italy; (S.F.); (R.B.)
| | - Maria Grazia Tibiletti
- SC Anatomia Patologica, Ospedale di Circolo, ASST Settelaghi, 21100 Varese, Italy; (N.S.); (I.C.); (E.D.L.); (F.S.)
- Centro di Ricerca dei Tumori Eredo-Familiari, Dipartimento di Medicina e Chirurgia, University of Insubria, 21100 Varese, Italy
| |
Collapse
|
48
|
Miller RE, Leary A, Scott CL, Serra V, Lord CJ, Bowtell D, Chang DK, Garsed DW, Jonkers J, Ledermann JA, Nik-Zainal S, Ray-Coquard I, Shah SP, Matias-Guiu X, Swisher EM, Yates LR. ESMO recommendations on predictive biomarker testing for homologous recombination deficiency and PARP inhibitor benefit in ovarian cancer. Ann Oncol 2020; 31:1606-1622. [PMID: 33004253 DOI: 10.1016/j.annonc.2020.08.2102] [Citation(s) in RCA: 273] [Impact Index Per Article: 54.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 08/14/2020] [Accepted: 08/17/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Homologous recombination repair deficiency (HRD) is a frequent feature of high-grade serous ovarian, fallopian tube and peritoneal carcinoma (HGSC) and is associated with sensitivity to PARP inhibitor (PARPi) therapy. HRD testing provides an opportunity to optimise PARPi use in HGSC but methodologies are diverse and clinical application remains controversial. MATERIALS AND METHODS To define best practice for HRD testing in HGSC the ESMO Translational Research and Precision Medicine Working Group launched a collaborative project that incorporated a systematic review approach. The main aims were to (i) define the term 'HRD test'; (ii) provide an overview of the biological rationale and the level of evidence supporting currently available HRD tests; (iii) provide recommendations on the clinical utility of HRD tests in clinical management of HGSC. RESULTS A broad range of repair genes, genomic scars, mutational signatures and functional assays are associated with a history of HRD. Currently, the clinical validity of HRD tests in ovarian cancer is best assessed, not in terms of biological HRD status per se, but in terms of PARPi benefit. Clinical trials evidence supports the use of BRCA mutation testing and two commercially available assays that also incorporate genomic instability for identifying subgroups of HGSCs that derive different magnitudes of benefit from PARPi therapy, albeit with some variation by clinical scenario. These tests can be used to inform treatment selection and scheduling but their use is limited by a failure to consistently identify a subgroup of patients who derive no benefit from PARPis in most studies. Existing tests lack negative predictive value and inadequately address the complex and dynamic nature of the HRD phenotype. CONCLUSIONS Currently available HRD tests are useful for predicting likely magnitude of benefit from PARPis but better biomarkers are urgently needed to better identify current homologous recombination proficiency status and stratify HGSC management.
Collapse
Affiliation(s)
- R E Miller
- Department of Medical Oncology, University College London, London, UK; Department of Medical Oncology, St Bartholomew's Hospital, London, UK
| | - A Leary
- Department of Medicine and INSERM U981, Gustave Roussy Cancer Center, Université Paris-Saclay, Paris, France
| | - C L Scott
- Peter MacCallum Cancer Centre, Melbourne, Australia; The University of Melbourne, Melbourne, Australia
| | - V Serra
- Experimental Therapeutics Group Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - C J Lord
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK; CRUK Gene Function Laboratory, The Institute of Cancer Research, London, UK
| | - D Bowtell
- Peter MacCallum Cancer Centre, Melbourne, Australia; The University of Melbourne, Melbourne, Australia
| | - D K Chang
- Glasgow Precision Oncology Laboratory, Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, UK; West of Scotland Pancreatic Unit, Glasgow Royal Infirmary, Glasgow, UK
| | - D W Garsed
- Peter MacCallum Cancer Centre, Melbourne, Australia; The University of Melbourne, Melbourne, Australia
| | - J Jonkers
- Division of Molecular Pathology, Oncode Institute, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - J A Ledermann
- UCL Cancer Institute, University College London, London, UK
| | - S Nik-Zainal
- Department of Medical Genetics, School of Clinical Medicine, University of Cambridge, Cambridge, UK; MRC Cancer Unit, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - I Ray-Coquard
- Centre Leon Berard, Lyon, France; University Claude Bernard Groupe University of Lyon, France
| | - S P Shah
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, USA
| | - X Matias-Guiu
- Departments of Pathology, Hospital U Arnau de Vilanova and Hospital U de Bellvitge, Universities of Lleida and Barcelona, Irblleida, Idibell, Ciberonc, Barcelona, Spain
| | - E M Swisher
- Department of Obstetrics and Gynecology, University of Washington, Seattle, USA
| | - L R Yates
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge; Guy's Cancer Centre, Guys and St Thomas' NHS Foundation Trust, London, UK.
| |
Collapse
|
49
|
Hurley LC, Levin NK, Chatterjee M, Coles J, Muszkat S, Howarth Z, Dyson G, Tainsky MA. Evaluation of paraneoplastic antigens reveals TRIM21 autoantibodies as biomarker for early detection of ovarian cancer in combination with autoantibodies to NY-ESO-1 and TP53. Cancer Biomark 2020; 27:407-421. [PMID: 32083570 DOI: 10.3233/cbm-190988] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND The majority of ovarian cancer cases are diagnosed at an advanced stage with poor prognosis. This study evaluates autoantibodies against tumor antigens to identify candidate biomarkers for early detection of ovarian cancer in women at increased risk. OBJECTIVE To assess the immunoreactivity of paraneoplastic antigens and tumor associated antigens with high-grade serous ovarian cancer (HGSOC) samples. METHODS Five paraneoplastic antigens along with three tumor-associated antigens were evaluated with HGSOC patient serum samples. Validation screening was performed with n= 164 serum samples consisting of: 50 late stage HGSOC, 14 early stage HGSOC, 50 benign ovarian cyst, and 50 healthy control samples on ELISA and western blot. The four markers TRIM21, NY-ESO-1, TP53, and PAX8 were evaluated on a second validation serum set, n= 150. RESULTS TRIM21 achieved the highest sensitivity in the first validation screening of 33% with 100% specificity. Combining TRIM21 with NY-ESO-1, TP53, and PAX8 provided 67% sensitivity with 94% specificity, and 56% sensitivity at 98% specificity. These four markers resulted in 46% sensitivity with 98% specificity in the second validation cohort; TRIM21 achieved the highest individual sensitivity of 36%. CONCLUSIONS Autoantibodies to TRIM21, NY-ESO-1, and TP53 may complement CA125 in screening of women at genetic risk for ovarian cancer.
Collapse
Affiliation(s)
- Laura C Hurley
- Department of Oncology, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Nancy K Levin
- Department of Oncology, School of Medicine, Wayne State University, Detroit, MI, USA.,Molecular Therapeutics Program, Karmanos Cancer Institute, Detroit, MI, USA
| | - Madhumita Chatterjee
- Department of Oncology, School of Medicine, Wayne State University, Detroit, MI, USA.,Molecular Therapeutics Program, Karmanos Cancer Institute, Detroit, MI, USA
| | - Jasmine Coles
- Department of Oncology, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Shlomo Muszkat
- Department of Oncology, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Zachary Howarth
- Department of Oncology, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Gregory Dyson
- Department of Oncology, School of Medicine, Wayne State University, Detroit, MI, USA.,Molecular Therapeutics Program, Karmanos Cancer Institute, Detroit, MI, USA
| | - Michael A Tainsky
- Department of Oncology, School of Medicine, Wayne State University, Detroit, MI, USA.,Center for Molecular Medicine and Genetics, School of Medicine, Wayne State University, Detroit, MI, USA.,Molecular Therapeutics Program, Karmanos Cancer Institute, Detroit, MI, USA
| |
Collapse
|
50
|
Sheta R, Bachvarova M, Plante M, Renaud MC, Sebastianelli A, Gregoire J, Navarro JM, Perez RB, Masson JY, Bachvarov D. Development of a 3D functional assay and identification of biomarkers, predictive for response of high-grade serous ovarian cancer (HGSOC) patients to poly-ADP ribose polymerase inhibitors (PARPis): targeted therapy. J Transl Med 2020; 18:439. [PMID: 33213473 PMCID: PMC7678187 DOI: 10.1186/s12967-020-02613-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 11/06/2020] [Indexed: 11/13/2022] Open
Abstract
Background Poly(ADP-ribose) polymerase inhibitors (PARPis) specifically target homologous recombination deficiency (HRD) cells and display good therapeutic effect in women with advanced-stage BRCA1/2-mutated breast and epithelial ovarian cancer (EOC). However, about 50% of high grade serous ovarian cancers (HGSOC) present with HRD due to epigenetic BRCA1 inactivation, as well as genetic/epigenetic inactivation(s) of other HR genes, a feature known as “BRCAness”. Therefore, there is a potential for extending the use of PARPis to these patients if HR status can be identified. Methods We have developed a 3D (spheroid) functional assay to assess the sensitivity of two PARPis (niraparib and olaparib) in ascites-derived primary cell cultures (AsPCs) from HGSOC patients. A method for AsPCs preparation was established based on a matrix (agarose), allowing for easy isolation and successive propagation of monolayer and 3D AsPCs. Based on this method, we performed cytotoxicity assays on 42 AsPCs grown both as monolayers and spheroids. Results The response to PARPis treatment in monolayer AsPCs, was significantly higher, compared to 3D AsPCs, as 88% and 52% of the monolayer AsPCs displayed sensitivity to niraparib and olaparib respectively, while 66% of the 3D AsPCs were sensitive to niraparib and 38% to olaparib, the latter being more consistent with previous estimates of HRD (40%–60%) in EOC. Moreover, niraparib displayed a significantly stronger cytotoxic effect in both in 3D and monolayer AsPCs, which was confirmed by consecutive analyses of the HR pathway activity (γH2AX foci formation) in PARPis-sensitive and resistant AsPCs. Global gene expression comparison of 6 PARPi-resistant and 6 PARPi-sensitive 3D AsPCs was indicative for the predominant downregulation of numerous genes and networks with previously demonstrated roles in EOC chemoresistance, suggesting that the PARPis-sensitive AsPCs could display enhanced sensitivity to other chemotherapeutic drugs, commonly applied in cancer management. Microarray data validation identified 24 potential gene biomarkers associated with PARPis sensitivity. The differential expression of 7 selected biomarkers was consecutively confirmed by immunohistochemistry in matched EOC tumor samples. Conclusion The application of this assay and the potential biomarkers with possible predictive significance to PARPis therapy of EOC patients now need testing in the setting of a clinical trial.
Collapse
Affiliation(s)
- Razan Sheta
- Department of Molecular Medicine, Université Laval, Québec, QC, G1V 0A6, Canada.,Centre de recherche du CHU de Québec, Oncology division, L'Hôtel-Dieu de Québec, 9 rue McMahon, Québec, QC, G1R 3S3, Canada
| | - Magdalena Bachvarova
- Centre de recherche du CHU de Québec, Oncology division, L'Hôtel-Dieu de Québec, 9 rue McMahon, Québec, QC, G1R 3S3, Canada
| | - Marie Plante
- Centre de recherche du CHU de Québec, Oncology division, L'Hôtel-Dieu de Québec, 9 rue McMahon, Québec, QC, G1R 3S3, Canada.,Department of Obstetrics and Gynecology, Université Laval, Québec, QC, G1V 0A6, Canada
| | - Marie-Claude Renaud
- Centre de recherche du CHU de Québec, Oncology division, L'Hôtel-Dieu de Québec, 9 rue McMahon, Québec, QC, G1R 3S3, Canada.,Department of Obstetrics and Gynecology, Université Laval, Québec, QC, G1V 0A6, Canada
| | - Alexandra Sebastianelli
- Centre de recherche du CHU de Québec, Oncology division, L'Hôtel-Dieu de Québec, 9 rue McMahon, Québec, QC, G1R 3S3, Canada.,Department of Obstetrics and Gynecology, Université Laval, Québec, QC, G1V 0A6, Canada
| | - Jean Gregoire
- Centre de recherche du CHU de Québec, Oncology division, L'Hôtel-Dieu de Québec, 9 rue McMahon, Québec, QC, G1R 3S3, Canada.,Department of Obstetrics and Gynecology, Université Laval, Québec, QC, G1V 0A6, Canada
| | - Jamilet Miranda Navarro
- Bioinformatics Department, Center for Genetic Engineering and Biotechnology, 10600, Havana, CP, Cuba
| | - Ricardo Bringas Perez
- Bioinformatics Department, Center for Genetic Engineering and Biotechnology, 10600, Havana, CP, Cuba
| | - Jean-Yves Masson
- Centre de recherche du CHU de Québec, Oncology division, L'Hôtel-Dieu de Québec, 9 rue McMahon, Québec, QC, G1R 3S3, Canada.,Department of Molecular Biology, Medical Biochemistry, and Pathology, Laval University Cancer Research Center, Québec, QC, G1V 0A6, Canada
| | - Dimcho Bachvarov
- Department of Molecular Medicine, Université Laval, Québec, QC, G1V 0A6, Canada. .,Centre de recherche du CHU de Québec, Oncology division, L'Hôtel-Dieu de Québec, 9 rue McMahon, Québec, QC, G1R 3S3, Canada.
| |
Collapse
|