1
|
Osler TS, Schoeman M, Pretorius WJS, Mathew CG, Edge J, Urban MF. Application of genetic testing criteria for hereditary breast cancer in South Africa. Breast Cancer Res Treat 2025:10.1007/s10549-024-07585-3. [PMID: 39776011 DOI: 10.1007/s10549-024-07585-3] [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: 09/22/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025]
Abstract
PURPOSE Breast cancer (BC) is the commonest cancer in South African women. A proportion are associated with a pathogenic or likely pathogenic (P/LP) variant in a BC susceptibility gene. Clinical guidelines for genetic testing are used to optimise variant detection while containing costs. We assessed the detection rate in women of diverse ancestries who met the South African National Department of Health (NDOH) testing guidelines, and analysed relationships between testing criteria, participant characteristics and presence of a BRCA1/2 P/LP variant. METHODS Records from 376 women with BC who met NDOH criteria and had genetic testing were included. Demographic, clinical and test result data were collated to describe detection rates according to criteria met, and a multivariate analysis conducted to find variables most frequently associated with a P/LP variant. RESULTS P/LP variant prevalence in women meeting NDOH testing criteria was 19.9% (75/376). Women meeting ≥ 2 guideline criteria were over twice as likely to have a P/LP variant (OR 2.27, 95%CI 1.27-4.07, p = 0.006), highlighting the guidelines' capacity to stratify risk. Family history (OR 1.97; 95%CI 1.05-3.70, p = 0.03) and Black African ancestry (OR 2.58; 95%CI 1.28-5.18, p < 0.01) were independently associated with having a BRCA1/2 P/LP variant when controlling for other variables. Notably, although Black African participants were less likely to report a family history, those that did had higher odds of a P/LP variant in BRCA1/2. CONCLUSION These results demonstrate the usefulness of the NDOH guidelines in women of diverse ancestries and provide insight into the factors associated with P/LP variants in understudied African populations.
Collapse
Affiliation(s)
- T S Osler
- Sydney Brenner Institute for Molecular Bioscience and Division of Human Genetics, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.
- Division of Human Genetics, Faculty of Health Sciences, National Health Laboratory Service and University of the Witwatersrand, Johannesburg, South Africa.
| | - M Schoeman
- Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, University of Stellenbosch and Tygerberg Hospital, Parow, South Africa
| | - W J S Pretorius
- Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, University of Stellenbosch and Tygerberg Hospital, Parow, South Africa
| | - C G Mathew
- Sydney Brenner Institute for Molecular Bioscience and Division of Human Genetics, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Division of Human Genetics, Faculty of Health Sciences, National Health Laboratory Service and University of the Witwatersrand, Johannesburg, South Africa
| | - J Edge
- Department of Surgery, Charlotte Maxeke Johannesburg Academic Hospital and University of the Witwatersrand, Johannesburg, South Africa
| | - M F Urban
- Division of Human Genetics, Faculty of Health Sciences, National Health Laboratory Service and University of the Witwatersrand, Johannesburg, South Africa
- Department of Obstetrics and Gynaecology, Faculty of Medicine and Health Sciences, University of Stellenbosch and Tygerberg Hospital, Parow, South Africa
| |
Collapse
|
2
|
Landeros N, Vargas-Roig L, Denita S, Mampel A, Hasbún R, Araya H, Castillo I, Valdes C, Flores M, Salter JS, Vasquez K, Romero J, Pérez-Castro R. Regional Hereditary Cancer Program in Chile: A scalable model of genetic counseling and molecular diagnosis to improve clinical outcomes for patients with hereditary cancer across Latin America. Biol Res 2024; 57:99. [PMID: 39710803 DOI: 10.1186/s40659-024-00579-x] [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: 08/12/2024] [Accepted: 12/09/2024] [Indexed: 12/24/2024] Open
Abstract
BACKGROUND Breast cancer is a leading cause of cancer-related mortality worldwide, with hereditary forms accounting for approximately 10% of cases. In Chile, significant gaps exist in genetic counseling and testing, particularly within the public health system. This study presents the implementation and outcomes of the first regional hereditary cancer program in the Maule region of Chile, aimed at improving detection and management of hereditary breast cancer. METHODS A cohort of 48 high-risk breast cancer patients from the Hospital Regional de Talca received genetic counseling and underwent Next-Generation Sequencing multigene panel testing. The program was established through collaboration between multiple institutions, leveraging telemedicine and outsourcing sequencing analysis to address regional gaps. RESULTS Pathogenic or likely pathogenic variants were identified in 12% of patients, including in BRCA1, BRCA2, TP53, and PALB2. Notably, novel pathogenic variants in BRCA1 (rs80357505) and TP53 (rs1131691022) were discovered, highlighting the unique genetic landscape of the Chilean population. Additionally, 70 variants of uncertain significance were found across 42 genes, particularly in FAN1, MSH6, and FANCI, underscoring the need for further research. The program's collaborative approach effectively bridged critical gaps in genetic services, providing high-quality care within the public health system despite limited resources. CONCLUSIONS The Regional Hereditary Cancer Program addresses significant gaps in genetic counseling and testing in Chile's public health system. This scalable model enhances early detection and personalized treatment for hereditary cancer patients and could be adapted to other regions across Latin America.
Collapse
Affiliation(s)
- Natalia Landeros
- Unidad de Innovación en Prevención y Oncología de Precisión Centro Oncológico, Facultad de Medicina, Unidad de Innovación en Prevención y Oncología de Precisión Universidad Católica del Maule, Talca, 3480094, Chile
- In Vivo Tumor Biology Research Facility, Centro Oncológico, Facultad de Medicina, Universidad Católica del Maule, Talca, 3480094, Chile
- Biomedical Research Labs, Facultad de Medicina, Universidad Católica del Maule, Talca, 3480094, Chile
| | - Laura Vargas-Roig
- Tumor Biology Laboratory, Institute of Medicine and Experimental Biology of Cuyo, National Research Council of Argentine, Mendoza, Argentina
- Medical School, National University of Cuyo, Mendoza, Argentina
| | | | - Alejandra Mampel
- Tumor Biology Laboratory, Institute of Medicine and Experimental Biology of Cuyo, National Research Council of Argentine, Mendoza, Argentina
- Medical School, National University of Cuyo, Mendoza, Argentina
- University Hospital, Mendoza, Argentina
| | - Rafael Hasbún
- Hospital Regional de Talca (HRT), Talca, 3480094, Chile
| | - Hernán Araya
- Hospital Regional de Talca (HRT), Talca, 3480094, Chile
| | - Iván Castillo
- Unidad de Innovación en Prevención y Oncología de Precisión Centro Oncológico, Facultad de Medicina, Unidad de Innovación en Prevención y Oncología de Precisión Universidad Católica del Maule, Talca, 3480094, Chile
- Hospital Regional de Talca (HRT), Talca, 3480094, Chile
| | - Camila Valdes
- Hospital Regional de Talca (HRT), Talca, 3480094, Chile
| | | | | | - Katherin Vasquez
- Biomedical Research Labs, Facultad de Medicina, Universidad Católica del Maule, Talca, 3480094, Chile
| | - Jacqueline Romero
- Biomedical Research Labs, Facultad de Medicina, Universidad Católica del Maule, Talca, 3480094, Chile
| | - Ramón Pérez-Castro
- Unidad de Innovación en Prevención y Oncología de Precisión Centro Oncológico, Facultad de Medicina, Unidad de Innovación en Prevención y Oncología de Precisión Universidad Católica del Maule, Talca, 3480094, Chile.
- In Vivo Tumor Biology Research Facility, Centro Oncológico, Facultad de Medicina, Universidad Católica del Maule, Talca, 3480094, Chile.
- Biomedical Research Labs, Facultad de Medicina, Universidad Católica del Maule, Talca, 3480094, Chile.
| |
Collapse
|
3
|
Jahan N, Taraba J, Boddicker NJ, Giridhar KV, Leon-Ferre RA, Tevaarwerk AJ, Cathcart-Rake E, O'Sullivan CC, Peethambaram PP, Hobday TJ, Mina LA, Batalini F, Advani P, Sideras K, Haddad TC, Ruddy KJ, Goetz MP, Couch FJ, Yadav S. Real-World Evidence on Prescribing Patterns and Clinical Outcomes of Metastatic Breast Cancer Patients Treated with PARP Inhibitors: The Mayo Clinic Experience. Clin Breast Cancer 2024:S1526-8209(24)00284-2. [PMID: 39516069 DOI: 10.1016/j.clbc.2024.10.006] [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: 06/22/2024] [Revised: 09/22/2024] [Accepted: 10/10/2024] [Indexed: 11/16/2024]
Abstract
PURPOSE This study evaluates real-world outcomes, toxicities, and prescribing patterns of PARP inhibitors (PARPis) for the treatment of metastatic breast cancer (MBC). PATIENTS AND METHODS Electronic health records of 62 MBC patients treated with olaparib (n = 48) or talazoparib (n = 14) at Mayo Clinic System between 2017 and 2022 were analyzed. Time-to-treatment-failure (TTF) was assessed utilizing the Kaplan-Meier method. Predictors of TTF were identified in a multivariate Cox-proportional hazard regression model adjusting for relevant tumor and demographic characteristics. RESULTS Among 62 patients who received PARPis for MBC, 55 had germline (g) pathogenic variants (PVs) (gBRCA1 = 24, gBRCA2 = 26, and gPALB2 = 4) and 8 patients had somatic (s) PVs (sBRCA1 = 4, sBRCA2 = 2, sATM = 1, sCDKN2A = 1). Median TTF in the gBRCA1, gBRCA2, and gPALB2 PV carriers were 7, 8, and 9 months, respectively (P = .37). Complete or partial responses were observed among 51.8% of patients with gBRCA or gPALB2 PVs. In multivariate analysis, HER2 positivity (hazard ratio, HR: 4.9, P = .007) and somatic PVs in homologous recombination repair (HRR) genes other than BRCA (sATM or sCDKN2A) (HR: 11.7, P = .01) were associated with a shorter TTF. No significant difference in TTF was observed by the type of PARPi, estrogen and progesterone receptor status, age, or number of prior therapies. Eight (16.7%) patients receiving olaparib and seven (50%) receiving talazoparib required dose reductions due to toxicities. CONCLUSIONS In real-world practice, PARPis are well-tolerated with promising TTF in gBRCA1/2 and gPALB2 carriers. Further studies will delineate the clinical efficacy of PARPis in other MBC subsets, such as sBRCA mutations, HER2-positive disease, and CNS metastasis.
Collapse
Affiliation(s)
- Nusrat Jahan
- Division of Hematology and Oncology, University of Alabama at Birmingham, Birmingham, AL; Department of Oncology, Mayo Clinic, Rochester, MN.
| | - Jodi Taraba
- Department of Oncology, Mayo Clinic, Rochester, MN
| | | | | | | | | | | | | | | | | | - Lida A Mina
- Department of Hematology and Oncology, Mayo Clinic, Scottsdale, AZ
| | - Felipe Batalini
- Department of Hematology and Oncology, Mayo Clinic, Scottsdale, AZ
| | - Pooja Advani
- Department of Hematology and Oncology, Mayo Clinic, Jacksonville, FL
| | | | | | | | | | - Fergus J Couch
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | | |
Collapse
|
4
|
Yadav S, Couch FJ, Domchek SM. Germline Genetic Testing for Hereditary Breast and Ovarian Cancer: Current Concepts in Risk Evaluation. Cold Spring Harb Perspect Med 2024; 14:a041318. [PMID: 38151326 PMCID: PMC11293548 DOI: 10.1101/cshperspect.a041318] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
Our understanding of hereditary breast and ovarian cancer has significantly improved over the past two decades. In addition to BRCA1/2, pathogenic variants in several other DNA-repair genes have been shown to increase the risks of breast and ovarian cancer. The magnitude of cancer risk is impacted not only by the gene involved, but also by family history of cancer, polygenic risk scores, and, in certain genes, pathogenic variant type or location. While estimates of breast and ovarian cancer risk associated with pathogenic variants are available, these are predominantly based on studies of high-risk populations with young age at diagnosis of cancer, multiple primary cancers, or family history of cancer. More recently, breast cancer risk for germline pathogenic variant carriers has been estimated from population-based studies. Here, we provide a review of the field of germline genetic testing and risk evaluation for hereditary breast and ovarian cancers in high-risk and population-based settings.
Collapse
Affiliation(s)
- Siddhartha Yadav
- Department of Oncology, Mayo Clinic, Rochester, Minnesota 55905, USA
| | - Fergus J Couch
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota 55901, USA
| | - Susan M Domchek
- Basser Center for BRCA, Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
5
|
Petkov VI, Byun JS, Ward KC, Schussler NC, Archer NP, Bentler S, Doherty JA, Durbin EB, Gershman ST, Cheng I, Insaf T, Gonsalves L, Hernandez BY, Koch L, Liu L, Monnereau A, Morawski BM, Schwartz SM, Stroup A, Wiggins C, Wu XC, Bonds S, Negoita S, Penberthy L. Reporting tumor genomic test results to SEER registries via linkages. J Natl Cancer Inst Monogr 2024; 2024:168-179. [PMID: 39102888 DOI: 10.1093/jncimonographs/lgae013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 02/28/2024] [Accepted: 02/29/2024] [Indexed: 08/07/2024] Open
Abstract
BACKGROUND Precision medicine has become a mainstay of cancer care in recent years. The National Cancer Institute (NCI) Surveillance, Epidemiology, and End Results (SEER) Program has been an authoritative source of cancer statistics and data since 1973. However, tumor genomic information has not been adequately captured in the cancer surveillance data, which impedes population-based research on molecular subtypes. To address this, the SEER Program has developed and implemented a centralized process to link SEER registries' tumor cases with genomic test results that are provided by molecular laboratories to the registries. METHODS Data linkages were carried out following operating procedures for centralized linkages established by the SEER Program. The linkages used Match*Pro, a probabilistic linkage software, and were facilitated by the registries' trusted third party (an honest broker). The SEER registries provide to NCI limited datasets that undergo preliminary evaluation prior to their release to the research community. RESULTS Recently conducted genomic linkages included OncotypeDX Breast Recurrence Score, OncotypeDX Breast Ductal Carcinoma in Situ, OncotypeDX Genomic Prostate Score, Decipher Prostate Genomic Classifier, DecisionDX Uveal Melanoma, DecisionDX Preferentially Expressed Antigen in Melanoma, DecisionDX Melanoma, and germline tests results in Georgia and California SEER registries. CONCLUSIONS The linkages of cancer cases from SEER registries with genomic test results obtained from molecular laboratories offer an effective approach for data collection in cancer surveillance. By providing de-identified data to the research community, the NCI's SEER Program enables scientists to investigate numerous research inquiries.
Collapse
Affiliation(s)
- Valentina I Petkov
- Surveillance Research Program, Division of Cancer Control and Population Sciences, National Cancer Institute, Bethesda, MD, USA
| | - Jung S Byun
- Surveillance Research Program, Division of Cancer Control and Population Sciences, National Cancer Institute, Bethesda, MD, USA
| | | | | | - Natalie P Archer
- Cancer Epidemiology and Surveillance Branch, Texas Department of State Health Services, Austin, TX, USA
| | - Suzanne Bentler
- Iowa Cancer Registry, The University of Iowa, Iowa City, IA, USA
| | - Jennifer A Doherty
- Hunstman Cancer Institute, University of Utah, Salt Lake City, UT, USA
- Department of Population Health Sciences, University of Utah, Salt Lake City, UT, USA
| | - Eric B Durbin
- Cancer Research Informatics Shared Resource Facility, Markey Cancer Center, Kentucky Cancer Registry, University of Kentucky, Lexington, KY, USA
| | | | - Iona Cheng
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA
| | - Tabassum Insaf
- New York State Department of Health, New York State Cancer Registry, Albany, NY, USA
| | - Lou Gonsalves
- Connecticut Department of Public Health, Connecticut Tumor Registry, Hartford, CT, USA
| | | | - Lori Koch
- Illinois State Cancer Registry, Springfield, IL, USA
| | - Lihua Liu
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Alain Monnereau
- Public Health Institute, Cancer Registry of Greater California, Sacramento, CA, USA
| | | | - Stephen M Schwartz
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | | | - Charles Wiggins
- Department of Internal Medicine, University of New Mexico, Albuquerque, NM, USA
| | - Xiao-Cheng Wu
- School of Medicine, Louisiana State University, New Orleans, LA, USA
| | - Sarah Bonds
- Surveillance Research Program, Division of Cancer Control and Population Sciences, National Cancer Institute, Bethesda, MD, USA
| | - Serban Negoita
- Surveillance Research Program, Division of Cancer Control and Population Sciences, National Cancer Institute, Bethesda, MD, USA
| | - Lynne Penberthy
- Surveillance Research Program, Division of Cancer Control and Population Sciences, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
6
|
Cheo SW, Ong PY, Ow SGW, Chan GHJ, Tan DSP, Lim YW, Kong HL, Wong ALA, Lim SE, Walsh RJ, Wong ASC, Low JJH, Ngoi NYL, Lim JSL, Lee SC. Therapeutic applications of germline testing for cancer predisposition genes in Asia in the real world. ESMO Open 2024; 9:103482. [PMID: 38833967 PMCID: PMC11179091 DOI: 10.1016/j.esmoop.2024.103482] [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: 11/04/2023] [Revised: 04/28/2024] [Accepted: 04/30/2024] [Indexed: 06/06/2024] Open
Abstract
BACKGROUND Germline genetic testing is traditionally carried out in patients suspected with hereditary cancer syndrome for enhanced cancer surveillance and/or preventive strategies, but is increasingly carried out for therapeutic indications. MATERIALS AND METHODS We conducted a retrospective review of patients who underwent germline genetic testing at our centre to determine the prevalence of actionable pathogenic germline variants (PGV) and their clinical utility. RESULTS From 2000 to 2022, 1154 cancer patients underwent germline testing, with the majority (945/1154) tested with multi-gene panels. Four hundred and eleven (35.6%) patients harboured a PGV and 334 (81%) were clinically actionable. BRCA1/2 accounted for 62.3% of actionable mutations, followed by mismatch repair (18%), and other homologous recombination repair (HRR) genes (19.7%). One hundred and fifty-two germline-positive patients have advanced cancers, and 79 received germline-directed therapies (poly ADP ribose polymerase inhibitors = 75; immunotherapy = 4). Median duration of immunotherapy and poly ADP ribose polymerase were 20.5 months (range 5-40 months) and 8 months (range 1-76 months), respectively. Among BRCA/HRR mutation carriers who received platinum-based chemotherapy, pathological complete response rate in the neoadjuvant setting was 53% (n = 17 breast cancers) and objective response rate was >80% in the advanced setting (n = 71). CONCLUSIONS One-third of cancer patients tested carried a PGV and ∼80% were clinically actionable. Three-quarters of germline-positive advanced cancer patients received germline-directed therapies in the real world, underscoring the practical utility of germline testing to guide cancer therapeutics.
Collapse
Affiliation(s)
- S W Cheo
- Department of Haematology-Oncology, National University Cancer Institute, Singapore
| | - P Y Ong
- Department of Haematology-Oncology, National University Cancer Institute, Singapore
| | - S G W Ow
- Department of Haematology-Oncology, National University Cancer Institute, Singapore
| | - G H J Chan
- Department of Haematology-Oncology, National University Cancer Institute, Singapore
| | - D S P Tan
- Department of Haematology-Oncology, National University Cancer Institute, Singapore; Cancer Science Institute, National University of Singapore, Singapore; Yong Loo Lin School of Medicine, National University of Singapore, Singapore; NUS Centre for Cancer Research (N2CR), National University of Singapore, Singapore
| | - Y W Lim
- Department of Haematology-Oncology, National University Cancer Institute, Singapore
| | | | - A L A Wong
- Department of Haematology-Oncology, National University Cancer Institute, Singapore
| | - S E Lim
- Department of Haematology-Oncology, National University Cancer Institute, Singapore
| | - R J Walsh
- Department of Haematology-Oncology, National University Cancer Institute, Singapore
| | - A S C Wong
- Department of Haematology-Oncology, National University Cancer Institute, Singapore
| | - J J H Low
- Department of Obstetrics and Gynaecology, National University Hospital, Singapore
| | - N Y L Ngoi
- Department of Haematology-Oncology, National University Cancer Institute, Singapore
| | - J S L Lim
- Department of Haematology-Oncology, National University Cancer Institute, Singapore; Cancer Science Institute, National University of Singapore, Singapore; Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - S C Lee
- Department of Haematology-Oncology, National University Cancer Institute, Singapore; Cancer Science Institute, National University of Singapore, Singapore; Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| |
Collapse
|
7
|
Freire MV, Martin M, Segers K, Sepulchre E, Leroi N, Coupier J, Kalantari HR, Wolter P, Collignon J, Polus M, Plomteux O, Josse C, Bours V. Digenic Inheritance of Mutations in Homologous Recombination Genes in Cancer Patients. J Pers Med 2024; 14:584. [PMID: 38929805 PMCID: PMC11204488 DOI: 10.3390/jpm14060584] [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: 04/07/2024] [Revised: 05/21/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES BRCA1, BRCA2, ATM, and CHEK2 are known cancer predisposition genes (CPGs), but tumor risk in patients with simultaneous pathogenic variants (PVs) in CPGs remains largely unknown. In this study, we describe six patients from five families with multiple cancers who coinherited a combination of PVs in these genes. METHODS PVs were identified using NGS DNA sequencing and were confirmed by Sanger. RESULTS Families 1, 2, and 3 presented PVs in BRCA2 and ATM, family 4 in BRCA2 and BRCA1, and family 5 in BRCA2 and CHEK2. PVs were identified using NGS DNA sequencing and were confirmed by Sanger. The first family included patients with kidney, prostate, and breast cancer, in addition to pancreatic adenocarcinomas. In the second family, a female had breast cancer, while a male from the third family had prostate, gastric, and pancreatic cancer. The fourth family included a male with pancreatic cancer, and the fifth family a female with breast cancer. CONCLUSIONS The early age of diagnosis and the development of multiple cancers in the reported patients indicate a very high risk of cancer in double-heterozygous patients associated with PVs in HR-related CPGs. Therefore, in families with patients who differ from other family members in terms of phenotype, age of diagnosis, or type of cancer, the cascade testing needs to include the study of other CPGs.
Collapse
Affiliation(s)
- Maria Valeria Freire
- Department of Human Genetics, GIGA Research Center, University of Liège and CHU Liège, Av. Hippocrate 1/11, 4000 Liège, Belgium;
| | - Marie Martin
- Department of Human Genetics, CHU Liège, Domaine Universitaire, 4000 Liège, Belgium; (M.M.); (K.S.); (E.S.); (N.L.)
| | - Karin Segers
- Department of Human Genetics, CHU Liège, Domaine Universitaire, 4000 Liège, Belgium; (M.M.); (K.S.); (E.S.); (N.L.)
| | - Edith Sepulchre
- Department of Human Genetics, CHU Liège, Domaine Universitaire, 4000 Liège, Belgium; (M.M.); (K.S.); (E.S.); (N.L.)
| | - Natacha Leroi
- Department of Human Genetics, CHU Liège, Domaine Universitaire, 4000 Liège, Belgium; (M.M.); (K.S.); (E.S.); (N.L.)
| | - Jérôme Coupier
- Department of Human Genetics, CHU Liège, Domaine Universitaire, 4000 Liège, Belgium; (M.M.); (K.S.); (E.S.); (N.L.)
| | | | - Pascal Wolter
- Onco-Hematology Department, St Nikolaus Hospital, Hufengasse 4/8, 4700 Eupen, Belgium;
| | - Joëlle Collignon
- Department of Medical Oncology, GIGA Research Center, University of Liège and CHU Liège, Domaine Universitaire, 4000 Liège, Belgium; (J.C.); (C.J.)
| | - Marc Polus
- Department of Gastroenterology, CHU Liège, Av. Hippocrate 1/11, 4000 Liège, Belgium;
| | - Olivier Plomteux
- Gastro-Enterology Department, CHC, Boulevard Patience et Beaujonc 2, 4000 Liège, Belgium;
| | - Claire Josse
- Department of Medical Oncology, GIGA Research Center, University of Liège and CHU Liège, Domaine Universitaire, 4000 Liège, Belgium; (J.C.); (C.J.)
| | - Vincent Bours
- Department of Human Genetics, GIGA Research Center, University of Liège and CHU Liège, Av. Hippocrate 1/11, 4000 Liège, Belgium;
| |
Collapse
|
8
|
Agaoglu NB, Unal B, Hayes CP, Walker M, Ng OH, Doganay L, Can ND, Rana HQ, Ghazani AA. Genomic disparity impacts variant classification of cancer susceptibility genes in Turkish breast cancer patients. Cancer Med 2024; 13:e6852. [PMID: 38308423 PMCID: PMC10905328 DOI: 10.1002/cam4.6852] [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: 05/20/2023] [Revised: 11/23/2023] [Accepted: 12/10/2023] [Indexed: 02/04/2024] Open
Abstract
OBJECTIVE Turkish genome is underrepresented in large genomic databases. This study aims to evaluate the effect of allele frequency in the Turkish population in determining the clinical utility of germline findings in breast cancer, including invasive lobular carcinoma (ILC), mixed invasive ductal and lobular carcinoma (IDC-L), and ductal carcinoma (DC). METHODS Two clinic-based cohorts from the Umraniye Research and Training Hospital (URTH) were used in this study: a cohort consisting of 132 women with breast cancer and a non-cancer cohort consisting of 492 participants. The evaluation of the germline landscape was performed by analysis of 27 cancer genes. The frequency and type of variants in the breast cancer cohort were compared to those in the non-cancer cohort to investigate the effect of population genetics. The variant allele frequencies in Turkish Variome and gnomAD were statistically evaluated. RESULTS The genetic analysis identified 121 variants in the breast cancer cohort (actionable = 32, VUS = 89) and 223 variants in the non-cancer cohort (actionable = 25, VUS = 188). The occurrence of 21 variants in both suggested a possible genetic population effect. Evaluation of allele frequency of 121 variants from the breast cancer cohort showed 22% had a significantly higher value in Turkish Variome compared to gnomAD (p < 0.0001, 95% CI) with a mean difference of 60 times (ranging from 1.37-354.4). After adjusting for variant allele frequency using the ancestry-appropriate database, 6.7% (5/75) of VUS was reclassified to likely benign. CONCLUSION To our knowledge, this is the first study of population genetic effects in breast cancer subtypes in Turkish women. Our findings underscore the need for a large genomic database representing Turkish population-specific variants. It further highlights the significance of the ancestry-appropriate population database for accurate variant assessment in clinical settings.
Collapse
Affiliation(s)
- Nihat B. Agaoglu
- Department of Medical Genetics, Division of Cancer GeneticsUmraniye Training and Research HospitalIstanbulTurkey
| | - Busra Unal
- Department of Medical Genetics, Division of Cancer GeneticsUmraniye Training and Research HospitalIstanbulTurkey
- Division of GeneticsBrigham and Women's HospitalBostonMassachusettsUSA
| | - Connor P. Hayes
- Division of GeneticsBrigham and Women's HospitalBostonMassachusettsUSA
| | - McKenzie Walker
- Division of GeneticsBrigham and Women's HospitalBostonMassachusettsUSA
| | - Ozden Hatirnaz Ng
- Department of Medical Biology, School of MedicineAcibadem UniversityIstanbulTurkey
| | - Levent Doganay
- Department of Medical Genetics, Division of Cancer GeneticsUmraniye Training and Research HospitalIstanbulTurkey
| | - Nisan D. Can
- Department of Molecular Biology Genetics and BiotechnologyIstanbul Technical UniversityIstanbulTurkey
| | - Huma Q. Rana
- Division of Cancer Genetics and PreventionDana‐Farber Cancer InstituteBostonMassachusettsUSA
- Department of MedicineBrigham and Women's HospitalBostonMassachusettsUSA
- Harvard Medical SchoolBostonMassachusettsUSA
| | - Arezou A. Ghazani
- Division of GeneticsBrigham and Women's HospitalBostonMassachusettsUSA
- Department of MedicineBrigham and Women's HospitalBostonMassachusettsUSA
- Harvard Medical SchoolBostonMassachusettsUSA
| |
Collapse
|
9
|
Boujemaa M, Nouira F, Jandoubi N, Mejri N, Bouaziz H, Charfeddine C, Ben Nasr S, Labidi S, El Benna H, Berrazega Y, Rachdi H, Daoud N, Benna F, Haddaoui A, Abdelhak S, Samir Boubaker M, Boussen H, Hamdi Y. Uncovering the clinical relevance of unclassified variants in DNA repair genes: a focus on BRCA negative Tunisian cancer families. Front Genet 2024; 15:1327894. [PMID: 38313678 PMCID: PMC10834681 DOI: 10.3389/fgene.2024.1327894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 01/02/2024] [Indexed: 02/06/2024] Open
Abstract
Introduction: Recent advances in sequencing technologies have significantly increased our capability to acquire large amounts of genetic data. However, the clinical relevance of the generated data continues to be challenging particularly with the identification of Variants of Uncertain Significance (VUSs) whose pathogenicity remains unclear. In the current report, we aim to evaluate the clinical relevance and the pathogenicity of VUSs in DNA repair genes among Tunisian breast cancer families. Methods: A total of 67 unsolved breast cancer cases have been investigated. The pathogenicity of VUSs identified within 26 DNA repair genes was assessed using different in silico prediction tools including SIFT, PolyPhen2, Align-GVGD and VarSEAK. Effects on the 3D structure were evaluated using the stability predictor DynaMut and molecular dynamics simulation with NAMD. Family segregation analysis was also performed. Results: Among a total of 37 VUSs identified, 11 variants are likely deleterious affecting ATM, BLM, CHEK2, ERCC3, FANCC, FANCG, MSH2, PMS2 and RAD50 genes. The BLM variant, c.3254dupT, is novel and seems to be associated with increased risk of breast, endometrial and colon cancer. Moreover, c.6115G>A in ATM and c.592+3A>T in CHEK2 were of keen interest identified in families with multiple breast cancer cases and their familial cosegregation with disease has been also confirmed. In addition, functional in silico analyses revealed that the ATM variant may lead to protein immobilization and rigidification thus decreasing its activity. We have also shown that FANCC and FANCG variants may lead to protein destabilization and alteration of the structure compactness which may affect FANCC and FANCG protein activity. Conclusion: Our findings revealed that VUSs in DNA repair genes might be associated with increased cancer risk and highlight the need for variant reclassification for better disease management. This will help to improve the genetic diagnosis and therapeutic strategies of cancer patients not only in Tunisia but also in neighboring countries.
Collapse
Affiliation(s)
- Maroua Boujemaa
- Laboratory of Biomedical Genomics and Oncogenetics, LR20IPT05, Institut Pasteur de Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Fatma Nouira
- Laboratory of Bioactive Substances, Center of Biotechnology of Borj Cedria, University of Tunis El Manar, Hamam, Tunisia
| | - Nouha Jandoubi
- Laboratory of Biomedical Genomics and Oncogenetics, LR20IPT05, Institut Pasteur de Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Nesrine Mejri
- Laboratory of Biomedical Genomics and Oncogenetics, LR20IPT05, Institut Pasteur de Tunis, University of Tunis El Manar, Tunis, Tunisia
- Medical Oncology Department, Abderrahman Mami Hospital, Faculty of Medicine Tunis, University Tunis El Manar, Tunis, Tunisia
| | - Hanen Bouaziz
- Laboratory of Biomedical Genomics and Oncogenetics, LR20IPT05, Institut Pasteur de Tunis, University of Tunis El Manar, Tunis, Tunisia
- Surgical Oncology Department, Salah Azaiez Institute of Cancer, Tunis, Tunisia
| | - Cherine Charfeddine
- Laboratory of Biomedical Genomics and Oncogenetics, LR20IPT05, Institut Pasteur de Tunis, University of Tunis El Manar, Tunis, Tunisia
- High Institute of Biotechnology of Sidi Thabet, Biotechpole of Sidi Thabet, University of Manouba, Ariana, Tunisia
| | - Sonia Ben Nasr
- Laboratory of Biomedical Genomics and Oncogenetics, LR20IPT05, Institut Pasteur de Tunis, University of Tunis El Manar, Tunis, Tunisia
- Department of Medical Oncology, Military Hospital of Tunis, Tunis, Tunisia
| | - Soumaya Labidi
- Laboratory of Biomedical Genomics and Oncogenetics, LR20IPT05, Institut Pasteur de Tunis, University of Tunis El Manar, Tunis, Tunisia
- Medical Oncology Department, Abderrahman Mami Hospital, Faculty of Medicine Tunis, University Tunis El Manar, Tunis, Tunisia
| | - Houda El Benna
- Laboratory of Biomedical Genomics and Oncogenetics, LR20IPT05, Institut Pasteur de Tunis, University of Tunis El Manar, Tunis, Tunisia
- Medical Oncology Department, Abderrahman Mami Hospital, Faculty of Medicine Tunis, University Tunis El Manar, Tunis, Tunisia
| | - Yosra Berrazega
- Medical Oncology Department, Abderrahman Mami Hospital, Faculty of Medicine Tunis, University Tunis El Manar, Tunis, Tunisia
| | - Haifa Rachdi
- Medical Oncology Department, Abderrahman Mami Hospital, Faculty of Medicine Tunis, University Tunis El Manar, Tunis, Tunisia
| | - Nouha Daoud
- Medical Oncology Department, Abderrahman Mami Hospital, Faculty of Medicine Tunis, University Tunis El Manar, Tunis, Tunisia
| | - Farouk Benna
- Radiation Oncology Department, Salah Azaiez Institute, Tunis, Tunisia
| | | | - Sonia Abdelhak
- Laboratory of Biomedical Genomics and Oncogenetics, LR20IPT05, Institut Pasteur de Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Mohamed Samir Boubaker
- Laboratory of Biomedical Genomics and Oncogenetics, LR20IPT05, Institut Pasteur de Tunis, University of Tunis El Manar, Tunis, Tunisia
- Laboratory of Human and Experimental Pathology, Institut Pasteur de Tunis, Tunis, Tunisia
| | - Hamouda Boussen
- Laboratory of Biomedical Genomics and Oncogenetics, LR20IPT05, Institut Pasteur de Tunis, University of Tunis El Manar, Tunis, Tunisia
- Medical Oncology Department, Abderrahman Mami Hospital, Faculty of Medicine Tunis, University Tunis El Manar, Tunis, Tunisia
| | - Yosr Hamdi
- Laboratory of Biomedical Genomics and Oncogenetics, LR20IPT05, Institut Pasteur de Tunis, University of Tunis El Manar, Tunis, Tunisia
- Laboratory of Human and Experimental Pathology, Institut Pasteur de Tunis, Tunis, Tunisia
| |
Collapse
|
10
|
Yang X, Smirnov A, Buonomo OC, Mauriello A, Shi Y, Bischof J, Woodsmith J, Melino G, Candi E, Bernassola F. A primary luminal/HER2 negative breast cancer patient with mismatch repair deficiency. Cell Death Discov 2023; 9:365. [PMID: 37783677 PMCID: PMC10545677 DOI: 10.1038/s41420-023-01650-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 08/23/2023] [Accepted: 09/13/2023] [Indexed: 10/04/2023] Open
Abstract
Here, we present the case of a 47-year-old woman diagnosed with luminal B breast cancer subtype and provide an in-depth analysis of her gene mutations, chromosomal alterations, mRNA and protein expression changes. We found a point mutation in the FGFR2 gene, which is potentially hyper-activating the receptor function, along with over-expression of its ligand FGF20 due to genomic amplification. The patient also harbors somatic and germline mutations in some mismatch repair (MMR) genes, with a strong MMR mutational signature. The patient displays high microsatellite instability (MSI) and tumor mutational burden (TMB) status and increased levels of CTLA-4 and PD-1 expression. Altogether, these data strongly implicate that aberrant FGFR signaling, and defective MMR system might be involved in the development of this breast tumor. In addition, high MSI and TMB in the context of CTLA-4 and PD-L1 positivity, suggest the potential benefit of immune checkpoint inhibitors. Accurate characterization of molecular subtypes, based on gene mutational and expression profiling analyses, will be certainly helpful for individualized treatment and targeted therapy of breast cancer patients, especially for those subtypes with adverse outcome.
Collapse
Affiliation(s)
- Xue Yang
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133, Rome, Italy
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, Soochow University, Suzhou, 215000, China
| | - Artem Smirnov
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133, Rome, Italy
- Istituto Dermopatico Immacolata (IDI-IRCCS), 00100, Rome, Italy
| | - Oreste Claudio Buonomo
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Alessandro Mauriello
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Yufang Shi
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, Soochow University, Suzhou, 215000, China
| | - Julia Bischof
- Indivumed GmbH, Falkenried, Germany Biochemistry Laboratory, 88 Building D, 20251, Hamburg, Germany
| | - Jonathan Woodsmith
- Indivumed GmbH, Falkenried, Germany Biochemistry Laboratory, 88 Building D, 20251, Hamburg, Germany
| | - Gerry Melino
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133, Rome, Italy.
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany.
| | - Eleonora Candi
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133, Rome, Italy.
- Istituto Dermopatico Immacolata (IDI-IRCCS), 00100, Rome, Italy.
| | - Francesca Bernassola
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133, Rome, Italy.
| |
Collapse
|
11
|
Chami A, de Souza Zózimo TR, Alves TM, Matosinho CGR, Santos C, Simões MM, Cabral WLR, de Paula Ricardo BF, da Silva Filho AL, Carvalho MRS, da Conceição Braga L. In deep bioinformatic characterization of a novel fumarate hydratase variant FH c.199T > G; (p.Tyr67Asp) in hereditary leiomyomatosis and renal cell carcinoma. Fam Cancer 2023; 22:481-486. [PMID: 37316640 DOI: 10.1007/s10689-023-00335-2] [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: 02/10/2022] [Accepted: 04/25/2023] [Indexed: 06/16/2023]
Abstract
Hereditary leiomyomatosis and renal cell carcinoma (HLRCC) is a rare, autosomal dominant tumor predisposition syndrome characterized by variable development of multiple skin and uterus leiomyomas and aggressive forms of renal cell carcinoma (RCC). Mutations in fumarate hydratase (FH), one of the proteins in homologous recombination repair, precede the development of HLRCC with high penetrance. Considering the risk of early metastasis of RCC, FH has been included in mutation screening panels. The identification of a pathogenic FH variant guides the screening for tumors in the carriers. However, variants of uncertain significance (VUS) are frequent findings, limiting the clinical value of the mutation screening. Here, we describe the associated phenotype and an in-depth, multi-step Bioinformatic evaluation of the germline FH c.199T > G (p.Tyr67 > Asp) variant segregated in an HLRCC family. Evidence for FH c.199T > G; (p.Tyr67Asp) pathogenicity includes the variant segregation with the disease in three affected family members, its absence in populational databases, and the deep evolutionary conservation of the Tyr67 residue. At the protein level, this residue substitution causes the loss of molecular bonds and ionic interactions, affecting molecular dynamics and protein stability. Considering ACMG/AMP criteria, we propose the reclassification of the FH c.199T > G; (p.Tyr67Asp) variant to "likely pathogenic". In addition, the in-depth, in silico approach used here allowed us to understand how and why FH c.199T > G; (p.Tyr67Asp) could cause HLRCC. This could help in clinical management decisions concerning the monitoring of unaffected family members having this variant.
Collapse
Affiliation(s)
- Anisse Chami
- Programa de Pós-Graduação em Tocoginecologia da Universidade Estadual de São Paulo - UNESP, Botucatu, SP, Brazil
- Rede Mater Dei de Saúde, Belo Horizonte, MG, Brazil
| | - Thalía Rodrigues de Souza Zózimo
- Programa de Pós-Graduação em Genética, Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais - UFMG, Campus Pampulha, Belo Horizonte, MG, Brazil
| | - Thamiris Matias Alves
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas - Bloco E3 - Sala 175, Universidade Federal de Minas Gerais - UFMG, Av. Antônio Carlos, 6627 - Campus Pampulha, Belo Horizonte, 31270-901, Brazil
| | - Carolina Guimarães Ramos Matosinho
- Programa de Pós-Graduação em Genética, Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais - UFMG, Campus Pampulha, Belo Horizonte, MG, Brazil
| | | | | | | | | | - Agnaldo Lopes da Silva Filho
- Programa de Pós-Graduação em Tocoginecologia da Universidade Estadual de São Paulo - UNESP, Botucatu, SP, Brazil
- Departamento de Ginecologia e Obstetrícia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Maria Raquel Santos Carvalho
- Programa de Pós-Graduação em Genética, Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais - UFMG, Campus Pampulha, Belo Horizonte, MG, Brazil.
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas - Bloco E3 - Sala 175, Universidade Federal de Minas Gerais - UFMG, Av. Antônio Carlos, 6627 - Campus Pampulha, Belo Horizonte, 31270-901, Brazil.
| | - Letícia da Conceição Braga
- Núcleo de Pesquisa Básica e Translacional, Instituto Mário Penna, Belo Horizonte, MG, Brazil
- OncoTag Desenvolvimento de Produto e Serviços Para Saúde Humana, Belo Horizonte, MG, Brazil
| |
Collapse
|
12
|
Joshi U, Budhathoki P, Gaire S, Yadav SK, Shah A, Adhikari A, Choong G, Couzi R, Giridhar KV, Leon-Ferre RA, Boughey JC, Hieken TJ, Mutter R, Ruddy KJ, Haddad TC, Goetz MP, Couch FJ, Yadav S. Clinical outcomes and prognostic factors in triple-negative invasive lobular carcinoma of the breast. Breast Cancer Res Treat 2023; 200:217-224. [PMID: 37210429 PMCID: PMC10782581 DOI: 10.1007/s10549-023-06959-3] [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: 03/06/2023] [Accepted: 04/21/2023] [Indexed: 05/22/2023]
Abstract
PURPOSE Triple-negative invasive lobular carcinoma (TN-ILC) of breast cancer is a rare disease and the clinical outcomes and prognostic factors are not well-defined. METHODS Women with stage I-III TN-ILC or triple-negative invasive ductal carcinoma (TN-IDC) of the breast undergoing mastectomy or breast-conserving surgery between 2010 and 2018 in the National Cancer Database were included. Kaplan-Meier curves and multivariate Cox proportional hazard regression were used to compare overall survival (OS) and evaluate prognostic factors. Multivariate logistic regression was performed to analyze the factors associated with pathological response to neoadjuvant chemotherapy. RESULTS The median age at diagnosis for women with TN-ILC was 67 years compared to 58 years in TN-IDC (p < 0.001). There was no significant difference in the OS between TN-ILC and TN-IDC in multivariate analysis (HR 0.96, p = 0.44). Black race and higher TNM stage were associated with worse OS, whereas receipt of chemotherapy or radiation was associated with better OS in TN-ILC. Among women with TN-ILC receiving neoadjuvant chemotherapy, the 5-year OS was 77.3% in women with a complete pathological response (pCR) compared to 39.8% in women without any response. The odds of achieving pCR following neoadjuvant chemotherapy were significantly lower in women with TN-ILC compared to TN-IDC (OR 0.53, p < 0.001). CONCLUSION Women with TN-ILC are older at diagnosis but have similar OS compared to TN-IDC after adjusting for tumor and demographic characteristics. Administration of chemotherapy was associated with improved OS in TN-ILC, but women with TN-ILC were less likely to achieve complete response to neoadjuvant therapy compared to TN-IDC.
Collapse
Affiliation(s)
- Utsav Joshi
- Department of Internal Medicine, Rochester General Hospital, Rochester, NY, 14621, USA
| | - Pravash Budhathoki
- Department of Internal Medicine, Bronxcare Health System, Bronx, NY, 10457, USA
| | - Suman Gaire
- Department of Internal Medicine, Mount Sinai Hospital Chicago, Chicago, IL, 60608, USA
| | - Sumeet K Yadav
- Department of Hospital Internal Medicine, Mayo Clinic, Mankato, MN, 56001, USA
| | - Anish Shah
- Department of Internal Medicine, Bronxcare Health System, Bronx, NY, 10457, USA
| | - Anurag Adhikari
- Department of Internal Medicine, Jacobi Medical Center, New York, NY, 10461, USA
| | - Grace Choong
- Department of Oncology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Rima Couzi
- Department of Oncology, Johns Hopkins Hospital, Baltimore, MD, 21231, USA
| | | | | | - Judy C Boughey
- Division of Breast and Melanoma Surgical Oncology, Department of Surgery, Mayo Clinic, Rochester, MN, 55905, USA
| | - Tina J Hieken
- Division of Breast and Melanoma Surgical Oncology, Department of Surgery, Mayo Clinic, Rochester, MN, 55905, USA
| | - Robert Mutter
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Kathryn J Ruddy
- Department of Oncology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Tufia C Haddad
- Department of Oncology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Matthew P Goetz
- Department of Oncology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Fergus J Couch
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA
| | | |
Collapse
|
13
|
Yadav S, Boddicker NJ, Na J, Polley EC, Hu C, Hart SN, Gnanaolivu RD, Larson N, Holtegaard S, Huang H, Dunn CA, Teras LR, Patel AV, Lacey JV, Neuhausen SL, Martinez E, Haiman C, Chen F, Ruddy KJ, Olson JE, John EM, Kurian AW, Sandler DP, O'Brien KM, Taylor JA, Weinberg CR, Anton-Culver H, Ziogas A, Zirpoli G, Goldgar DE, Palmer JR, Domchek SM, Weitzel JN, Nathanson KL, Kraft P, Couch FJ. Contralateral Breast Cancer Risk Among Carriers of Germline Pathogenic Variants in ATM, BRCA1, BRCA2, CHEK2, and PALB2. J Clin Oncol 2023; 41:1703-1713. [PMID: 36623243 PMCID: PMC10022863 DOI: 10.1200/jco.22.01239] [Citation(s) in RCA: 66] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 10/03/2022] [Accepted: 11/21/2022] [Indexed: 01/11/2023] Open
Abstract
PURPOSE To estimate the risk of contralateral breast cancer (CBC) among women with germline pathogenic variants (PVs) in ATM, BRCA1, BRCA2, CHEK2, and PALB2. METHODS The study population included 15,104 prospectively followed women within the CARRIERS study treated with ipsilateral surgery for invasive breast cancer. The risk of CBC was estimated for PV carriers in each gene compared with women without PVs in a multivariate proportional hazard regression analysis accounting for the competing risk of death and adjusting for patient and tumor characteristics. The primary analyses focused on the overall cohort and on women from the general population. Secondary analyses examined associations by race/ethnicity, age at primary breast cancer diagnosis, menopausal status, and tumor estrogen receptor (ER) status. RESULTS Germline BRCA1, BRCA2, and CHEK2 PV carriers with breast cancer were at significantly elevated risk (hazard ratio > 1.9) of CBC, whereas only the PALB2 PV carriers with ER-negative breast cancer had elevated risks (hazard ratio, 2.9). By contrast, ATM PV carriers did not have significantly increased CBC risks. African American PV carriers had similarly elevated risks of CBC as non-Hispanic White PV carriers. Among premenopausal women, the 10-year cumulative incidence of CBC was estimated to be 33% for BRCA1, 27% for BRCA2, and 13% for CHEK2 PV carriers with breast cancer and 35% for PALB2 PV carriers with ER-negative breast cancer. The 10-year cumulative incidence of CBC among postmenopausal PV carriers was 12% for BRCA1, 9% for BRCA2, and 4% for CHEK2. CONCLUSION Women diagnosed with breast cancer and known to carry germline PVs in BRCA1, BRCA2, CHEK2, or PALB2 are at substantially increased risk of CBC and may benefit from enhanced surveillance and risk reduction strategies.
Collapse
Affiliation(s)
| | | | - Jie Na
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN
| | - Eric C. Polley
- Department of Public Health Sciences, University of Chicago, Chicago, IL
| | - Chunling Hu
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Steven N. Hart
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN
| | | | - Nicole Larson
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN
| | - Susan Holtegaard
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Huaizhi Huang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN
| | - Carolyn A. Dunn
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Lauren R. Teras
- Department of Population Science, American Cancer Society, Atlanta, GA
| | - Alpa V. Patel
- Department of Population Science, American Cancer Society, Atlanta, GA
| | | | | | - Elena Martinez
- Department of Family Medicine and Public Health, University of California, San Diego, CA
| | - Christopher Haiman
- Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Fei Chen
- Keck School of Medicine, University of Southern California, Los Angeles, CA
| | | | - Janet E. Olson
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN
| | - Esther M. John
- Department of Epidemiology and Population Health, Stanford University School of Medicine, Stanford, CA
- Division of Oncology, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
| | - Allison W. Kurian
- Department of Epidemiology and Population Health, Stanford University School of Medicine, Stanford, CA
- Division of Oncology, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
| | - Dale P. Sandler
- National Institute of Environmental Health Sciences, Durham, NC
| | | | - Jack A. Taylor
- National Institute of Environmental Health Sciences, Durham, NC
| | | | | | | | - Gary Zirpoli
- Slone Epidemiology Center at Boston University, Boston, MA
| | | | | | - Susan M. Domchek
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
- Basser Center for BRCA, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | | | - Katherine L. Nathanson
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
- Basser Center for BRCA, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Peter Kraft
- Harvard University T.H. Chan School of Public Health, Boston, MA
| | - Fergus J. Couch
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| |
Collapse
|
14
|
Abstract
IMPORTANCE The recent successes of poly-ADP ribose polymerase (PARP) inhibitors and belzutifan support germline genetic data as an exciting, accessible source for biomarkers in cancer treatment. This study hypothesizes, however, that most oncology clinical trials using germline data largely prioritize BRCA1/2 as biomarkers and PARP inhibitors as therapy. OBJECTIVE To characterize past and ongoing oncology trials that use germline data. DESIGN, SETTING, AND PARTICIPANTS This retrospective cross-sectional study of oncology trials used the Informa Trialtrove database to evaluate trial attributes. Trials using germline information (including the terms germline, hereditary, or inherited in the title, treatment plan, interventions, end points, objectives, results, or notes) and conducted globally between December 1, 1990, and April 4, 2022 (data freeze date), were included. MAIN OUTCOMES AND MEASURES Trials by cancer type, phase, participants, sponsor type, end points, outcomes, and locations were described. Associated biomarkers and mechanisms of action for studied therapeutic interventions were counted. How germline data in trial inclusion and exclusion criteria are associated with end points, outcomes, and enrollment were also examined. RESULTS A total of 887 of 84 297 (1.1%) oncology clinical trials in the Trialtrove database that use germline data were identified. Most trials were conducted in cancer types where PARP inhibitors are already approved. A total of 74.8% (672) of trials were performed in the phase 2 setting or above. Trials were primarily sponsored by industry (523 trials [59.0%]), academia (382 trials [43.1%]), and the government (274 trials [30.9%]), where trials may have multiple sponsor types. Among 343 trials using germline data with outcomes in Trialtrove, 180 (52.5%) reported meeting primary end points. Although BRCA1/2 are the most frequent biomarkers seen (BRCA1, 224 trials [25.3%]; BRCA2, 228 trials [25.7%]), trials also examine pharmacogenomic variants and germline mediators of somatic biomarkers. PARP inhibitors or immunotherapy were tested in 69.9% of trials; PARP inhibition was the most frequently studied mechanism (367 trials [41.4%]). An overwhelming number of trials using germline data were conducted in the US, Canada, and Europe vs other countries, mirroring disparities in cancer genetics data. Germline data in inclusion and exclusion criteria are associated with altered end point, outcomes, and enrollment compared with oncology trials with no germline data use. Examples of inclusion and exclusion criteria regarding germline data that may unintentionally exclude patients were identified. CONCLUSIONS AND RELEVANCE These findings suggest that for germline biomarkers to gain clinical relevance, trials must expand biomarkers, therapies, and populations under study.
Collapse
Affiliation(s)
- Ashwin V Kammula
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
- University of Maryland, College Park
| | - Alejandro A Schäffer
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Padma Sheila Rajagopal
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| |
Collapse
|
15
|
Effects of Apatinib Mesylate Monotherapy on the Incidence of Adverse Reactions and Immune Function in Patients with Breast Cancer after Radical Mastectomy. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:4022282. [PMID: 35990841 PMCID: PMC9385297 DOI: 10.1155/2022/4022282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 06/26/2022] [Accepted: 07/15/2022] [Indexed: 11/23/2022]
Abstract
Objective To assess the effects of monotherapy with apatinib mesylate on the incidence of adverse events and immune function in breast cancer patients after a radical mastectomy. Methods Between December 2018 and August 2020, 90 patients with breast cancer scheduled for a radical mastectomy in People's Liberation Army Navy 971 Hospital were randomly recruited and assigned at a ratio of 1 : 1 to receive either conventional treatment (conventional group) or apatinib mesylate after radical mastectomy (study group). The primary endpoint was disease control rate (DCR), and the secondary endpoints were adverse events and the immune function of the patients. Results Monotherapy with apatinib mesylate was associated with a higher DCR (86.67%) versus conventional postoperative treatment (42.23%). All patients in the study group had documented adverse events, including 2 (4.45%) cases of headache, 3 (6.67%) cases of dizziness, 9 (20.00%) cases of hypertension, 6 (13.34%) cases of hand-foot syndrome, 3 (6.67%) cases of thrombocytopenia, 1 (2.23%) case of tinnitus, 7 (15.56%) cases of fatigue, 2 (4.45%) cases of anemia, 2 (4.45%) cases of oral pain, and 10 (22.23%) cases of leukopenia. There were 23 cases of intermittent discontinuation due to adverse events during treatment, 15 cases of dose reduction, and 3 cases of discontinuation due to adverse events. The difference in preoperative and postoperative T-cell subsets and natural killer (NK) cells between the two groups did not come up to the statistical standard (P > 0.05). Monotherapy with apatinib mesylate resulted in significantly lower levels of CD4+, CD4+/CD8+, and NK cells and higher CD8+ levels versus conventional treatment at 1 week and 4 weeks postoperatively (P < 0.05). Conclusion Apatinib mesylate monotherapy after radical mastectomy yields a high DCR, a lower incidence of adverse events, and improved immune recovery. Clinical trials are, however, required prior to clinical promotion.
Collapse
|
16
|
Vargason AB, Turner CE, Shriver CD, Ellsworth RE. Influence of germline test results on surgical decision making in women with invasive breast cancer. Cancer Genet 2022; 266-267:81-85. [PMID: 35868102 DOI: 10.1016/j.cancergen.2022.07.003] [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: 04/05/2022] [Revised: 06/10/2022] [Accepted: 07/10/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND While therapeutic mastectomy with contralateral prophylactic mastectomy (TM+CPM) and/or bilateral salpingo-oophorectomy (BSO) are recommended for women with pathogenic variants (PV) in some cancer predisposition genes, evidence for the utility of these surgeries for women with PV in other genes currently is insufficient. In conjunction, current guidelines recommend that clinical management should not be influenced by a return of a variant of uncertain significance (VUS). Return of germline test results may, however, influence surgical decision making regardless of current guidelines. We thus evaluated surgical choices amongst a cohort of women with invasive breast cancer who underwent clinical genetic testing. METHODS Germline test results and all surgical procedures were extracted for women who had unilateral invasive breast cancer and had clinical testing before definitive surgery (n = 591). Results were classified as pathogenic/likely pathogenic (PV, 17.1%), VUS (19.5%) or benign/likely benign (63.4%). Data were analyzed using chi-square tests with p<0.05 defining significance. RESULTS Rates of TM+CPM and BSO were not significantly different for women with VUS compared to those with benign findings. Rates of TM+CPM were significantly higher for women with PV in BRCA1 and BRCA2, PALB2, PTEN and TP53, as well in genes with insufficient data to recommend risk-reducing mastectomy. Rates of BSO were significantly higher in women with PV in BRCA1 and BRCA2, PALB2, PTEN and TP53 and BRIP1, RAD51C and RAD51D compared to those with benign findings. CONCLUSION Overall, surgical choices for women with a VUS were more similar to those from women with benign variants than to those with PV, however, in the group with PV in genes for which insufficient evidence exists for the benefit of risk-reducing mastectomy, rates of TM+CPM were high. Thus, while the management of women with VUS is in agreement with ACMG guidelines, patients with mutations in other cancer genes demonstrate a preference for more aggressive breast surgeries.
Collapse
Affiliation(s)
- Ashlee B Vargason
- Breast Care Clinic, Department of Surgery, Walter Reed National Military Medical Center, 8901 Rockville Pike, Bethesda, MD 20889, USA.
| | - Clesson E Turner
- National Human Genome Research Institute, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA.
| | - Craig D Shriver
- Murtha Cancer Center/Research Program, Uniformed Services University of the Health Sciences and Walter Reed National Military Medical Center, 8901 Rockville Pike, Bethesda, MD 20889, USA; Department of Surgery, Uniformed Services University of the Health Sciences, 8901 Rockville Pike, Bethesda, MD 20889, USA.
| | - Rachel E Ellsworth
- Murtha Cancer Center/Research Program, Uniformed Services University of the Health Sciences and Walter Reed National Military Medical Center, 8901 Rockville Pike, Bethesda, MD 20889, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Dr., Bethesda, MD 20817, USA.
| |
Collapse
|
17
|
Kashyap D, Pal D, Sharma R, Garg VK, Goel N, Koundal D, Zaguia A, Koundal S, Belay A. Global Increase in Breast Cancer Incidence: Risk Factors and Preventive Measures. BIOMED RESEARCH INTERNATIONAL 2022; 2022:9605439. [PMID: 35480139 PMCID: PMC9038417 DOI: 10.1155/2022/9605439] [Citation(s) in RCA: 223] [Impact Index Per Article: 74.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/25/2022] [Accepted: 03/21/2022] [Indexed: 02/07/2023]
Abstract
Breast cancer is a global cause for concern owing to its high incidence around the world. The alarming increase in breast cancer cases emphasizes the management of disease at multiple levels. The management should start from the beginning that includes stringent cancer screening or cancer registry to effective diagnostic and treatment strategies. Breast cancer is highly heterogeneous at morphology as well as molecular levels and needs different therapeutic regimens based on the molecular subtype. Breast cancer patients with respective subtype have different clinical outcome prognoses. Breast cancer heterogeneity emphasizes the advanced molecular testing that will help on-time diagnosis and improved survival. Emerging fields such as liquid biopsy and artificial intelligence would help to under the complexity of breast cancer disease and decide the therapeutic regimen that helps in breast cancer management. In this review, we have discussed various risk factors and advanced technology available for breast cancer diagnosis to combat the worst breast cancer status and areas that need to be focused for the better management of breast cancer.
Collapse
Affiliation(s)
- Dharambir Kashyap
- Department of Histopathology, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Deeksha Pal
- Department of Translational and Regenerative Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Riya Sharma
- Department of Pulmonary Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Vivek Kumar Garg
- Department of Medical Laboratory Technology, University Institute of Applied Health Sciences, Chandigarh University (Gharuan), Mohali 140313, India
| | - Neelam Goel
- Department of Information Technology, University Institute of Engineering & Technology, Panjab University, Chandigarh 160014, India
| | - Deepika Koundal
- Department of Systemics, School of Computer Science, University of Petroleum & Energy Studies, Dehradun, India
| | - Atef Zaguia
- Department of computer science, College of Computers and Information Technology, Taif University, P.O. BOX 11099, Taif 21944, Saudi Arabia
| | - Shubham Koundal
- Department of Medical Laboratory Technology, University Institute of Applied Health Sciences, Chandigarh University (Gharuan), Mohali 140313, India
| | - Assaye Belay
- Department of Statistics, Mizan-Tepi University, Ethiopia
| |
Collapse
|
18
|
Jakuboski SH, McDonald JA, Terry MB. Do current family history-based genetic testing guidelines contribute to breast cancer health inequities? NPJ Breast Cancer 2022; 8:36. [PMID: 35319016 PMCID: PMC8941019 DOI: 10.1038/s41523-022-00391-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 01/11/2022] [Indexed: 02/03/2023] Open
Abstract
Prior to the recommended age for population-based breast cancer screening by mammography, which ranges from 40-50 years depending on guidelines, the main way to identify higher risk women for earlier breast cancer (BC) screening to improve outcomes and discuss targeted chemoprevention is through specific clinical guidelines which are largely based on family history of breast cancer and known mutations in breast cancer susceptibility genes. The annual percent change (APC) in early-onset BC continues to rise, with the higher early-onset cancer burden and mortality continuing to be seen in non-Hispanic black (NHB) women compared to non-Hispanic white (NHW) women. Coupled with the increasing incidence overall as well as the lower percent of BC family history reported in NHB women compared with that of NHW women means that continued reliance on guidelines to identify women for genetic screening and initiation of early BC screening based largely on family history could lead to even greater BC health inequities. The similarity in the prevalence of mutations in key BC susceptibility genes between NHB and NHW women contrasts sharply to the differences in age-specific incidence rates between NHB and NHW women, supporting that there must be environmental modifiers that are contributing to the increased incidence in NHB women. This reality further argues for identifying NHB women early in adulthood through genetic testing who may benefit from tailored BC risk-reduction programs and early BC screening.
Collapse
Affiliation(s)
| | - Jasmine A McDonald
- Mailman School of Public Health, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Mary Beth Terry
- Mailman School of Public Health, Columbia University Irving Medical Center, New York, NY, 10032, USA.
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, 10032, USA.
| |
Collapse
|
19
|
What happens after referral? Completion rates of genetic counseling evaluations in breast cancer patients. J Natl Med Assoc 2022; 114:90-93. [PMID: 35039176 DOI: 10.1016/j.jnma.2021.12.006] [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: 08/18/2021] [Revised: 10/25/2021] [Accepted: 12/17/2021] [Indexed: 11/21/2022]
Abstract
Breast cancer (BC) is a common and often life-altering diagnosis for affected women and their families. Studies have indicated approximately 10% of breast cancer cases are inheritable. When patients are aware of their genetic status early, they are better equipped to make therapy decisions related to their cancer. Additionally, if patients are aware of pathogenic mutations, they can evaluate options such as chemoprevention with endocrine agents, prophylactic surgery, and have the ability to inform family members of their potential risk. Unfortunately, the shortage of genetic counselors has led to a large clinical demand delaying consultation. Although our institution employs genetic counselors on staff, the national shortage of counselors with this expertise has led to a disproportionate availability of providers to meet the clinical volume. This can lead to genetic counseling consultation often occurring beyond the patient's cancer treatment phase. Therefore, we sought to evaluate our referral patterns in an effort to determine whether qualifying patients were scheduled, evaluate delays in consultation, examine completion rates for genetic testing, and assess whether genetic counseling affected their subsequent care.
Collapse
|
20
|
Yadav S, Hu C, Nathanson KL, Weitzel JN, Goldgar DE, Kraft P, Gnanaolivu RD, Na J, Huang H, Boddicker NJ, Larson N, Gao C, Yao S, Weinberg C, Vachon CM, Trentham-Dietz A, Taylor JA, Sandler DR, Patel A, Palmer JR, Olson JE, Neuhausen S, Martinez E, Lindstrom S, Lacey JV, Kurian AW, John EM, Haiman C, Bernstein L, Auer PW, Anton-Culver H, Ambrosone CB, Karam R, Chao E, Yussuf A, Pesaran T, Dolinsky JS, Hart SN, LaDuca H, Polley EC, Domchek SM, Couch FJ. Germline Pathogenic Variants in Cancer Predisposition Genes Among Women With Invasive Lobular Carcinoma of the Breast. J Clin Oncol 2021; 39:3918-3926. [PMID: 34672684 PMCID: PMC8660003 DOI: 10.1200/jco.21.00640] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 06/16/2021] [Accepted: 09/16/2021] [Indexed: 12/14/2022] Open
Abstract
PURPOSE To determine the contribution of germline pathogenic variants (PVs) in hereditary cancer testing panel genes to invasive lobular carcinoma (ILC) of the breast. MATERIALS AND METHODS The study included 2,999 women with ILC from a population-based cohort and 3,796 women with ILC undergoing clinical multigene panel testing (clinical cohort). Frequencies of germline PVs in breast cancer predisposition genes (ATM, BARD1, BRCA1, BRCA2, BRIP1, CDH1, CHEK2, PALB2, PTEN, RAD51C, RAD51D, and TP53) were compared between women with ILC and unaffected female controls and between women with ILC and infiltrating ductal carcinoma (IDC). RESULTS The frequency of PVs in breast cancer predisposition genes among women with ILC was 6.5% in the clinical cohort and 5.2% in the population-based cohort. In case-control analysis, CDH1 and BRCA2 PVs were associated with high risks of ILC (odds ratio [OR] > 4) and CHEK2, ATM, and PALB2 PVs were associated with moderate (OR = 2-4) risks. BRCA1 PVs and CHEK2 p.Ile157Thr were not associated with clinically relevant risks (OR < 2) of ILC. Compared with IDC, CDH1 PVs were > 10-fold enriched, whereas PVs in BRCA1 were substantially reduced in ILC. CONCLUSION The study establishes that PVs in ATM, BRCA2, CDH1, CHEK2, and PALB2 are associated with an increased risk of ILC, whereas BRCA1 PVs are not. The similar overall PV frequencies for ILC and IDC suggest that cancer histology should not influence the decision to proceed with genetic testing. Similar to IDC, multigene panel testing may be appropriate for women with ILC, but CDH1 should be specifically discussed because of low prevalence and gastric cancer risk.
Collapse
Affiliation(s)
| | | | - Katherine L. Nathanson
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
- Basser Center for BRCA, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | | | | | - Peter Kraft
- Harvard University T.H. Chan School of Public Health, Boston, MA
| | | | - Jie Na
- Mayo Clinic, Rochester, MN
| | - Hongyan Huang
- Harvard University T.H. Chan School of Public Health, Boston, MA
| | | | | | - Chi Gao
- Harvard University T.H. Chan School of Public Health, Boston, MA
| | - Song Yao
- Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | | | | | | | | | | | - Alpa Patel
- Behavioral and Epidemiology Research Group, American Cancer Society, Atlanta, GA
| | | | | | | | | | | | | | | | | | - Christopher Haiman
- Keck School of Medicine, University of Southern California, Los Angeles, CA
| | | | - Paul W. Auer
- UWM Joseph J. Zilber School of Public Health, Milwaukee, WI
| | | | | | | | | | | | | | | | | | | | | | - Susan M. Domchek
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
- Basser Center for BRCA, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | | |
Collapse
|
21
|
McReynolds LJ, Giri N, Leathwood L, Risch MO, Carr AG, Alter BP. Risk of cancer in heterozygous relatives of patients with Fanconi anemia. Genet Med 2021; 24:245-250. [PMID: 34906449 DOI: 10.1016/j.gim.2021.08.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 06/10/2021] [Accepted: 08/19/2021] [Indexed: 10/19/2022] Open
Abstract
PURPOSE Fanconi anemia (FA) is a cancer-prone inherited bone marrow failure syndrome caused by biallelic pathogenic variants in one of >22 genes in the FA/BRCA DNA repair pathway. A major concern is whether the risk of cancer is increased in individuals with a single pathogenic FA gene variant. METHODS We evaluated the risk of cancer in the relatives of patients with FA in the National Cancer Institute Inherited Bone Marrow Failure Syndrome cohort. We genotyped all available relatives and determined the rates, types of cancer and the age of patients at cancer diagnosis. We calculated the observed-to-expected (O/E) cancer ratios using data from the Surveillance, Epidemiology, and End Results Program adjusted for age, sex, and birth cohort. RESULTS The risk of cancer was not increased among all FA relatives and FA heterozygotes (O/E ratios of 0.78 and 0.79, respectively). In particular, the risk of cancer was not increased among FANCA or FANCC heterozygotes (O/E ratios of 0.92 and 0.71, respectively). Relatives did not have typical FA cancers, and age at cancer diagnosis was not younger than expected. CONCLUSION Understanding the risk of cancer in individuals with single pathogenic FA variants is critical for counseling and management. We did not find increased risk of cancer in these individuals. These findings do not extend to the known cancer predisposition autosomal dominant FA genes, namely BRCA1, BRCA2, PALB2, BRIP1, and RAD51C.
Collapse
Affiliation(s)
- Lisa J McReynolds
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD.
| | - Neelam Giri
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD
| | | | | | | | - Blanche P Alter
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD
| |
Collapse
|
22
|
Dillon J, Ademuyiwa FO, Barrett M, Moss HA, Wignall E, Menendez C, Hughes KS, Plichta JK. Disparities in Genetic Testing for Heritable Solid-Tumor Malignancies. Surg Oncol Clin N Am 2021; 31:109-126. [PMID: 34776060 DOI: 10.1016/j.soc.2021.08.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Genetic testing offers providers a potentially life saving tool for identifying and intervening in high-risk individuals. However, disparities in receipt of genetic testing have been consistently demonstrated and undoubtedly have significant implications for the populations not receiving the standard of care. If correctly used, there is the potential for genetic testing to play a role in decreasing health disparities among individuals of different races and ethnicities. However, if genetic testing continues to revolutionize cancer care while being disproportionately distributed, it also has the potential to widen the existing mortality gap between various racial and ethnic populations.
Collapse
Affiliation(s)
- Jacquelyn Dillon
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Foluso O Ademuyiwa
- Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Megan Barrett
- Department of Obstetrics & Gynecology, Duke University Medical Center, Durham, NC, USA
| | - Haley A Moss
- Department of Obstetrics & Gynecology, Duke University Medical Center, Durham, NC, USA; Duke Cancer Institute, Durham, NC, USA. https://twitter.com/haleyarden1
| | | | - Carolyn Menendez
- Department of Surgery, Duke University Medical Center, Durham, NC, USA; Clinical Cancer Genetics, Duke Cancer Institute, Durham, NC, USA. https://twitter.com/@CSMenendez
| | - Kevin S Hughes
- Surgical Oncology, Massachusetts General Hospital, Boston, MA, USA
| | - Jennifer K Plichta
- Department of Surgery, Duke University Medical Center, Durham, NC, USA; Department of Population Health Sciences, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
23
|
Hayat M, Chen WC, Brandenburg JT, Babb de Villiers C, Ramsay M, Mathew CG. Genetic Susceptibility to Breast Cancer in Sub-Saharan African Populations. JCO Glob Oncol 2021; 7:1462-1471. [PMID: 34623906 PMCID: PMC8509920 DOI: 10.1200/go.21.00089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 07/28/2021] [Accepted: 09/01/2021] [Indexed: 12/24/2022] Open
Affiliation(s)
- Mahtaab Hayat
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Division of Human Genetics, National Health Laboratory Service and School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Wenlong Carl Chen
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- National Cancer Registry, National Health Laboratory Service, Johannesburg, South Africa
- Non-communicable Diseases Research Division, Wits Health Consortium (PTY) Ltd, Johannesburg, South Africa
| | - Jean-Tristan Brandenburg
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Chantal Babb de Villiers
- Division of Human Genetics, National Health Laboratory Service and School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Michèle Ramsay
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Christopher G. Mathew
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Division of Human Genetics, National Health Laboratory Service and School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Medical and Molecular Genetics, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| |
Collapse
|
24
|
Yadav S, Giridhar KV, Leone JP, Leon-Ferre RA, Ruddy KJ. A practical guide to endocrine therapy in the management of estrogen receptor-positive male breast cancer. BREAST CANCER MANAGEMENT 2021. [DOI: 10.2217/bmt-2021-0001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The majority (more than 90%) of male breast cancers (MaBCs) are estrogen receptor-positive, such that endocrine therapy is the mainstay of MaBC treatment. Endocrine therapy has been associated with improved overall survival in observational studies on MaBC, though large randomized clinical trials have never been completed to confirm this benefit in this population. Tamoxifen is currently the preferred drug for both metastatic and adjuvant treatment of MaBC. Known differences in treatment patterns and hormonal milieu between men and women may warrant a unique approach to the management of toxicities in men. This review provides a detailed discussion of endocrine therapy for MaBC.
Collapse
Affiliation(s)
| | | | - Jose Pablo Leone
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | | | - Kathryn J Ruddy
- Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
25
|
Gao C, Polley EC, Hart SN, Huang H, Hu C, Gnanaolivu R, Lilyquist J, Boddicker NJ, Na J, Ambrosone CB, Auer PL, Bernstein L, Burnside ES, Eliassen AH, Gaudet MM, Haiman C, Hunter DJ, Jacobs EJ, John EM, Lindström S, Ma H, Neuhausen SL, Newcomb PA, O'Brien KM, Olson JE, Ong IM, Patel AV, Palmer JR, Sandler DP, Tamimi R, Taylor JA, Teras LR, Trentham-Dietz A, Vachon CM, Weinberg CR, Yao S, Weitzel JN, Goldgar DE, Domchek SM, Nathanson KL, Couch FJ, Kraft P. Risk of Breast Cancer Among Carriers of Pathogenic Variants in Breast Cancer Predisposition Genes Varies by Polygenic Risk Score. J Clin Oncol 2021; 39:2564-2573. [PMID: 34101481 PMCID: PMC8330969 DOI: 10.1200/jco.20.01992] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 02/19/2021] [Accepted: 04/20/2021] [Indexed: 12/11/2022] Open
Abstract
PURPOSE This study assessed the joint association of pathogenic variants (PVs) in breast cancer (BC) predisposition genes and polygenic risk scores (PRS) with BC in the general population. METHODS A total of 26,798 non-Hispanic white BC cases and 26,127 controls from predominately population-based studies in the Cancer Risk Estimates Related to Susceptibility consortium were evaluated for PVs in BRCA1, BRCA2, ATM, CHEK2, PALB2, BARD1, BRIP1, CDH1, and NF1. PRS based on 105 common variants were created using effect estimates from BC genome-wide association studies; the performance of an overall BC PRS and estrogen receptor-specific PRS were evaluated. The odds of BC based on the PVs and PRS were estimated using penalized logistic regression. The results were combined with age-specific incidence rates to estimate 5-year and lifetime absolute risks of BC across percentiles of PRS by PV status and first-degree family history of BC. RESULTS The estimated lifetime risks of BC among general-population noncarriers, based on 10th and 90th percentiles of PRS, were 9.1%-23.9% and 6.7%-18.2% for women with or without first-degree relatives with BC, respectively. Taking PRS into account, more than 95% of BRCA1, BRCA2, and PALB2 carriers had > 20% lifetime risks of BC, whereas, respectively, 52.5% and 69.7% of ATM and CHEK2 carriers without first-degree relatives with BC, and 78.8% and 89.9% of those with a first-degree relative with BC had > 20% risk. CONCLUSION PRS facilitates personalization of BC risk among carriers of PVs in predisposition genes. Incorporating PRS into BC risk estimation may help identify > 30% of CHEK2 and nearly half of ATM carriers below the 20% lifetime risk threshold, suggesting the addition of PRS may prevent overscreening and enable more personalized risk management approaches.
Collapse
Affiliation(s)
- Chi Gao
- Harvard T.H. Chan School of Public Health, Boston, MA
| | | | | | - Hongyan Huang
- Harvard T.H. Chan School of Public Health, Boston, MA
| | | | | | | | | | - Jie Na
- Mayo Clinic, Rochester, MN
| | | | - Paul L. Auer
- UWM Joseph J. Zilber School of Public Health, Milwaukee, WI
| | | | | | - A. Heather Eliassen
- Harvard T.H. Chan School of Public Health, Boston, MA
- Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Mia M. Gaudet
- Department of Population Science, American Cancer Society, Atlanta, GA
| | - Christopher Haiman
- Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - David J. Hunter
- Harvard T.H. Chan School of Public Health, Boston, MA
- University of Oxford, Oxford, United Kingdom
| | - Eric J. Jacobs
- Department of Population Science, American Cancer Society, Atlanta, GA
| | | | - Sara Lindström
- Department of Epidemiology, University of Washington, Seattle, WA
- Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Huiyan Ma
- Beckman Research Institute of City of Hope, Duarte, CA
| | | | - Polly A. Newcomb
- Department of Epidemiology, University of Washington, Seattle, WA
- Fred Hutchinson Cancer Research Center, Seattle, WA
| | | | | | | | - Alpa V. Patel
- Department of Population Science, American Cancer Society, Atlanta, GA
| | - Julie R. Palmer
- Boston University School of Medicine and Slone Epidemiology Center, Boston, MA
| | - Dale P. Sandler
- National Institute of Environmental Health Sciences, Durham, NC
| | - Rulla Tamimi
- Population Health Sciences Department, Weill Cornell Medicine, New York, NY
| | - Jack A. Taylor
- National Institute of Environmental Health Sciences, Durham, NC
| | - Lauren R. Teras
- Department of Population Science, American Cancer Society, Atlanta, GA
| | | | | | | | - Song Yao
- Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | | | | | - Susan M. Domchek
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | | | | | - Peter Kraft
- Harvard T.H. Chan School of Public Health, Boston, MA
| |
Collapse
|
26
|
Yadav SK, Silwal S, Yadav S, Krishnamoorthy G, Chisti MM. A Systematic Comparison of Overall Survival Between Men and Women With Triple Negative Breast Cancer. Clin Breast Cancer 2021; 22:161-169. [PMID: 34419351 DOI: 10.1016/j.clbc.2021.07.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 01/03/2021] [Accepted: 07/02/2021] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Triple-negative breast cancer (TNBC) in men is very rare. The clinical characteristics, prognostic factors, and overall survival of men with TNBC have not been characterized. METHODS The study population consisted of men and women with a diagnosis of stage I-III TNBC between 2010 and 2016 in the National Cancer Database. Baseline demographic and tumor characteristics between men and women were compared using Pearson's Chi-Square test for categorical variables and Mann-Whitney U test for continuous variables. Kaplan-Meier and multivariate Cox proportional hazards regression model was used to compare survival and identify prognostic factors. RESULTS A total of 311 men and 95,406 women with TNBC were included in the final analysis. The 3-year and 5-year overall survival was 74.8% and 68.8% in men, while it was 83.2% and 74.8% in women, respectively. In multivariate analysis, men were found to have a significantly worse overall survival compared to women (HR, 1.49, 95% CI, 1.19-1.86, P= .01). Older age at diagnosis, higher TNM stage, undergoing mastectomy and not undergoing chemotherapy or radiation were identified as independent negative prognostic factors in men with TNBC. CONCLUSION In one of the largest studies of men with TNBC, men were noted to have a poorer overall survival compared to women, despite adjusting for usual prognostic factors. Further research into differences in tumor biology, treatment patterns and compliance with therapy between men and women are needed to understand the underlying etiologies for the survival difference in TNBC.
Collapse
Affiliation(s)
| | - Swechchha Silwal
- Department of Internal Medicine/Transitional Year, Detroit Medical Center Sinai Grace Hospital, Detroit, MI
| | | | | | | |
Collapse
|
27
|
Feng Q, Nickels E, Muskens IS, de Smith AJ, Gauderman WJ, Yee AC, Ricker C, Mack T, Leavitt AD, Godley LA, Wiemels JL. Increased burden of familial-associated early-onset cancer risk among minority Americans compared to non-Latino Whites. eLife 2021; 10:e64793. [PMID: 34155975 PMCID: PMC8219377 DOI: 10.7554/elife.64793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 06/04/2021] [Indexed: 01/11/2023] Open
Abstract
Background The role of race/ethnicity in genetic predisposition of early-onset cancers can be estimated by comparing family-based cancer concordance rates among ethnic groups. Methods We used linked California health registries to evaluate the relative cancer risks for first-degree relatives of patients diagnosed between ages 0 and 26, and the relative risks of developing distinct second primary malignancies (SPMs). From 1989 to 2015, we identified 29,631 cancer patients and 62,863 healthy family members. We calculated the standardized incident ratios (SIRs) of early-onset primary cancers diagnosed in proband siblings and mothers, as well as SPMs detected among early-onset patients. Analyses were stratified by self-identified race/ethnicity. Results Given probands with cancer, there were increased relative risks of any cancer for siblings and mothers (SIR = 3.32; 95% confidence interval [CI]: 2.85-3.85) and of SPMs (SIR = 7.27; 95% CI: 6.56-8.03). Given a proband with solid cancer, both Latinos (SIR = 4.98; 95% CI: 3.82-6.39) and non-Latino Blacks (SIR = 7.35; 95% CI: 3.36-13.95) exhibited significantly higher relative risk of any cancer in siblings and mothers when compared to non-Latino White subjects (SIR = 3.02; 95% CI: 2.12-4.16). For hematologic cancers, higher familial risk was evident for Asian/Pacific Islanders (SIR = 7.56; 95% CI: 3.26-14.90) compared to non-Latino whites (SIR = 2.69; 95% CI: 1.62-4.20). Conclusions The data support a need for increased attention to the genetics of early-onset cancer predisposition and environmental factors in race/ethnic minority families in the United States. Funding This work was supported by the V Foundation for funding this work (Grant FP067172).
Collapse
Affiliation(s)
- Qianxi Feng
- Department of Preventive Medicine, USC Keck School of MedicineLos AngelesUnited States
| | - Eric Nickels
- Department of Preventive Medicine, USC Keck School of MedicineLos AngelesUnited States
- Children's Hospital Los AngelesLos AngelesUnited States
| | - Ivo S Muskens
- Department of Preventive Medicine, USC Keck School of MedicineLos AngelesUnited States
| | - Adam J de Smith
- Department of Preventive Medicine, USC Keck School of MedicineLos AngelesUnited States
| | - W James Gauderman
- Department of Preventive Medicine, USC Keck School of MedicineLos AngelesUnited States
| | - Amy C Yee
- Department of Preventive Medicine, USC Keck School of MedicineLos AngelesUnited States
| | - Charite Ricker
- Norris Comprehensive Cancer Center, USC Keck School of MedicineLos AngelesUnited States
| | - Thomas Mack
- Department of Preventive Medicine, USC Keck School of MedicineLos AngelesUnited States
| | - Andrew D Leavitt
- Departments of Medicine and Laboratory Medicine, University of California, San FranciscoSan FranciscoUnited States
| | - Lucy A Godley
- Departments of Medicine and Human Genetics, The University of ChicagoChicagoUnited States
| | - Joseph L Wiemels
- Department of Preventive Medicine, USC Keck School of MedicineLos AngelesUnited States
| |
Collapse
|
28
|
Affiliation(s)
- Ismail Jatoi
- Division of Surgical Oncology and Endocrine Surgery, University of Texas Health Science Center, San Antonio
| | - Zoe Kemp
- Department of Cancer Genetics, Royal Marsden Hospital, NHS Trust, London, United Kingdom
- Department of Breast Medical Oncology, Royal Marsden Hospital, NHS Trust, London, United Kingdom
| |
Collapse
|
29
|
Abdel-Razeq H. Expanding the search for germline pathogenic variants for breast cancer. How far should we go and how high should we jump? The missed opportunity! Oncol Rev 2021; 15:544. [PMID: 34267891 PMCID: PMC8256373 DOI: 10.4081/oncol.2021.544] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 06/08/2021] [Indexed: 12/09/2022] Open
Abstract
Since the identification of BRCA1 and BRCA2 genes 3 decades ago, genetic testing and genetic counseling have become an integral part of routine clinical practice. The risk of breast cancer among carriers of germline pathogenic variants, like BRCA1 and BRCA2, is well established. Risk-reducing interventions, including bilateral mastectomies and salpingo-oophorectomies are both effective and have become more acceptable. Many researchers and professional societies view current guidelines as restrictive and may miss many at-risk women, and are calling to expand testing to include all patients with breast cancer, regardless of their personal or family history of cancer, while others are calling for wider adoption to even include all healthy women at age 30 or older. This review will address expanding testing in two directions; horizontally to include more patients, and even healthy women, and vertically to include more genes using next-generation sequencing-based multi-gene panel testing.
Collapse
Affiliation(s)
- Hikmat Abdel-Razeq
- Department of Internal Medicine, King Hussein Cancer Center, School of Medicine, University of Jordan, Amman, Jordan
| |
Collapse
|
30
|
Molecular Biomarkers for Contemporary Therapies in Hormone Receptor-Positive Breast Cancer. Genes (Basel) 2021; 12:genes12020285. [PMID: 33671468 PMCID: PMC7922594 DOI: 10.3390/genes12020285] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/08/2021] [Accepted: 02/12/2021] [Indexed: 02/06/2023] Open
Abstract
Systemic treatment of hormone receptor-positive (HR+) breast cancer is undergoing a renaissance, with a number of targeted therapies including CDK4/6, mTOR, and PI3K inhibitors now approved for use in combination with endocrine therapies. The increased use of targeted therapies has changed the natural history of HR+ breast cancers, with the emergence of new escape mechanisms leading to the inevitable progression of disease in patients with advanced cancers. The identification of new predictive and pharmacodynamic biomarkers to current standard-of-care therapies and discovery of new therapies is an evolving and urgent clinical challenge in this setting. While traditional, routinely measured biomarkers such as estrogen receptors (ERs), progesterone receptors (PRs), and human epidermal growth factor receptor 2 (HER2) still represent the best prognostic and predictive biomarkers for HR+ breast cancer, a significant proportion of patients either do not respond to endocrine therapy or develop endocrine resistant disease. Genomic tests have emerged as a useful adjunct prognostication tool and guide the addition of chemotherapy to endocrine therapy. In the treatment-resistant setting, mutational profiling has been used to identify ESR1, PIK3CA, and AKT mutations as predictive molecular biomarkers to newer therapies. Additionally, pharmacodynamic biomarkers are being increasingly used and considered in the metastatic setting. In this review, we summarise the current state-of-the-art therapies; prognostic, predictive, and pharmacodynamic molecular biomarkers; and how these are impacted by emerging therapies for HR+ breast cancer.
Collapse
|
31
|
Incorvaia L, Fanale D, Bono M, Calò V, Fiorino A, Brando C, Corsini LR, Cutaia S, Cancelliere D, Pivetti A, Filorizzo C, La Mantia M, Barraco N, Cusenza S, Badalamenti G, Russo A, Bazan V. BRCA1/2 pathogenic variants in triple-negative versus luminal-like breast cancers: genotype-phenotype correlation in a cohort of 531 patients. Ther Adv Med Oncol 2020; 12:1758835920975326. [PMID: 33403015 PMCID: PMC7747114 DOI: 10.1177/1758835920975326] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 10/29/2020] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Several available data suggest the association between specific molecular subtypes and BRCA1/2 mutational status. Previous investigations showed the association between BRCA1/2 pathogenic variants (PVs) in specific genomic regions and phenotypic variations of cancer relative risk, while the role of PV type and location in determining the breast cancer (BC) phenotypic features remains still unclear. The aim of this research was to describe the germline BRCA1/2 PVs in triple-negative breast cancer (TNBC) versus luminal-like BC and their potential leverage on BC phenotype. PATIENTS & METHODS We retrospectively collected and analyzed all clinical information of 531 patients with BC genetically tested for germline BRCA1/2 PVs by Next-Generation Sequencing analysis at University Hospital Policlinico "P. Giaccone" of Palermo (Sicily) from January 2016 to February 2020. RESULTS Our results corroborate the evidence that BRCA1-related tumors often have a profile which resembles the TNBC subtype, whereas BRCA2-associated tumors have a profile that resembles luminal-like BC, especially the Luminal B subtype. Interestingly, our findings suggest that the PVs identified in TNBC were not largely overlapping with those in luminal-like tumors. Differences in the frequency of two PVs potentially associated with different molecular tumor subtypes were observed. BRCA1-633delC was detected with relatively higher prevalence in patients with TNBC, whereas BRCA2-1466delT was found mainly in Luminal B tumors, but in no TNBC patient. CONCLUSION Future studies examining the type and location of BRCA1/2 PVs within different molecular subtypes are required to verify our hypothesis and could provide an interesting insight into the complex topic of genotype-phenotype correlations. Additionally, a more in-depth understanding of the potential correlations between BRCA PVs and clinical and phenotypic features of hereditary BC syndrome patients could be the key to develop better strategies of prevention and surveillance in BRCA-positive carriers without disease.
Collapse
Affiliation(s)
- Lorena Incorvaia
- Section of Medical Oncology, Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), University of Palermo, Palermo, Italy
| | - Daniele Fanale
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Marco Bono
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Valentina Calò
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Alessia Fiorino
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Chiara Brando
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Lidia Rita Corsini
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Sofia Cutaia
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Daniela Cancelliere
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Alessia Pivetti
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Clarissa Filorizzo
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Maria La Mantia
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Nadia Barraco
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Stefania Cusenza
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Giuseppe Badalamenti
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Antonio Russo
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Via del Vespro 129, Palermo, 90127, Italy
| | - Viviana Bazan
- Section of Medical Oncology, Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), University of Palermo, Palermo, Italy
| |
Collapse
|
32
|
Amirkhani Namagerdi A, d'Angelo D, Ciani F, Iannuzzi CA, Napolitano F, Avallone L, De Laurentiis M, Giordano A. Triple-Negative Breast Cancer Comparison With Canine Mammary Tumors From Light Microscopy to Molecular Pathology. Front Oncol 2020; 10:563779. [PMID: 33282730 PMCID: PMC7689249 DOI: 10.3389/fonc.2020.563779] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 10/19/2020] [Indexed: 12/12/2022] Open
Abstract
Many similar characteristics in human and dog cancers including, spontaneous development, clinical presentation, tumor heterogeneity, disease progression, and response to standard therapies have promoted the approval of this comparative model as an alternative to mice. Breast cancer represents the second most frequent neoplasm in humans after lung cancer. Triple-negative breast cancers (TNBC) constitute around 15% of all cases of breast cancer and do not express estrogen receptor (ER), progesterone receptor (PR), and do not overexpress human epidermal growth factor receptor 2 (HER2). As a result, they do not benefit from hormonal or trastuzumab-based therapy. Patients with TNBC have worse overall survival than patients with non-TNBC. Lehmann and collaborators described six different molecular subtypes of TNBC which further demonstrated its transcriptional heterogeneity. This six TNBC subtype classification has therapeutic implications. Breast cancer is the second most frequent neoplasm in sexually intact female dogs after skin cancer. Canine mammary tumors are a naturally occurring heterogeneous group of cancers that have several features in common with human breast cancer (HBC). These similarities include etiology, signaling pathway activation, and histological classification. Molecularly CMTs are more like TNBCs, and therefore dogs are powerful spontaneous models of cancer to test new therapeutic approaches, particularly for human TNBCs. More malignant tumors of the breast are more often ER and PR negative in both humans and dogs. Promising breast cancer biomarkers in both humans and canines are cancer-associated stroma (CAS), circulating tumor cells and tumor DNA (ctDNA), exosomes and miRNAs, and metabolites.
Collapse
Affiliation(s)
| | - Danila d'Angelo
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Naples, Italy
| | - Francesca Ciani
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Naples, Italy
| | | | - Francesco Napolitano
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Naples, Italy.,CCEINGE, Biotecnologie Avanzate, Naples, Italy
| | - Luigi Avallone
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Naples, Italy
| | - Michelino De Laurentiis
- Breast Oncology Division, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Naples, Italy
| | - Antonio Giordano
- Center for Biotechnology, College of Science and Technology, Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, PA, United States.,Department of Medical Biotechnologies, University of Siena, Siena, Italy
| |
Collapse
|
33
|
Reid S, Cadiz S, Pal T. Disparities in Genetic Testing and Care among Black women with Hereditary Breast Cancer. CURRENT BREAST CANCER REPORTS 2020; 12:125-131. [PMID: 33603954 PMCID: PMC7885902 DOI: 10.1007/s12609-020-00364-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
PURPOSE OF REVIEW Despite a steady improvement in breast cancer survival rates over the past several decades, mortality disparities remain among Black women, who have a 42% higher death rate compared to non-Hispanic white (NHW) women. Hereditary breast cancer (HBC) accounts for 5-10% of all breast cancer cases, the majority of which are due to the BRCA1 and BRCA2 (BRCA) genes. Despite the availability of BRCA testing for over 25 years, there remain disproportionately lower rates of genetic testing among Blacks compared to NHW due to a multitude of factors. The intent of this review is to discuss racial disparities focused on HBC across diverse populations and review the existing gaps to be addressed when delivering gene-based care. RECENT FINDINGS The factors contributing to the racial survival disparity are undoubtedly complex and likely an interplay between tumor biology, genomics, patterns of care and socioeconomic factors. Advances in genomic technologies that now allow for full characterization of germline DNA sequencing are integral in defining the complex and multifactorial cause of breast cancer and may help to explain the existing racial survival disparities. SUMMARY Identification of inherited cancer risk may lead to cancer prevention, early cancer detection, treatment guidance, and ultimately has great potential to improve outcomes. Consequently, advances in HBC diagnosis and treatment without widespread implementation have the potential to further widen the existing breast cancer mortality gap between Black and NHW women.
Collapse
Affiliation(s)
- Sonya Reid
- Vanderbilt University Medical Center, Nashville, TN
| | | | - Tuya Pal
- Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
34
|
Walsh EM, Smith KL, Stearns V. Management of hormone receptor-positive, HER2-negative early breast cancer. Semin Oncol 2020; 47:187-200. [PMID: 32546323 PMCID: PMC7374796 DOI: 10.1053/j.seminoncol.2020.05.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 05/07/2020] [Accepted: 05/07/2020] [Indexed: 12/24/2022]
Abstract
The majority of breast cancers are diagnosed at an early stage and are hormone receptor (HR)-positive and human epidermal growth factor receptor 2 (HER2)-negative. Significant advances have been made in the management of early stage HR-positive, HER2-negative breast cancer, resulting in improved survival outcomes. In this review, we discuss important factors to consider in the management of this disease. In particular, we discuss the role of adjuvant endocrine therapy, specific endocrine therapy agents, the duration of adjuvant endocrine therapy, treatment-related side effects, and the role of genomic assays and other biomarkers when considering treatment recommendations for individuals with HR-positive, HER2-negative early breast cancer. Finally, we address emerging data to individualize therapeutic decision-making and provide future considerations.
Collapse
Affiliation(s)
- Elaine M Walsh
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD
| | - Karen L Smith
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD
| | - Vered Stearns
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD.
| |
Collapse
|
35
|
Reid S, Pal T. Update on multi-gene panel testing and communication of genetic test results. Breast J 2020; 26:1513-1519. [PMID: 32639074 PMCID: PMC7484453 DOI: 10.1111/tbj.13971] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 11/14/2019] [Indexed: 12/20/2022]
Abstract
With technological advances, multi-gene panel testing has become increasingly used to identify patients at risk for hereditary breast cancer (HBC). There are currently evidence-based interventions and breast cancer screening strategies that exist for cancer prevention and early detection among patients with HBC. Moreover, in addition to the personal impact of identifying HBC, this information may be shared with at-risk family members to amplify the benefits of testing and subsequent care among those at high risk. Opportunities and challenges with the utilization of updated multi-gene panel testing for HBC, including: (a) tumor sequencing with germline consequences; (b) genetic counseling implications; and (c) strategies to improve the communication of genetic test results to family members will be reviewed. With the advances and expansion of genetic testing, all health care providers need to be updated on both the importance and complexities of HBC counseling and testing, in order to optimize patient care.
Collapse
Affiliation(s)
- Sonya Reid
- Vanderbilt University Medical Center, Nashville, Tennessee
| | - Tuya Pal
- Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
36
|
Murphy BL, Yi M, Arun BK, Gutierrez Barrera AM, Bedrosian I. Contralateral Risk-Reducing Mastectomy in Breast Cancer Patients Who Undergo Multigene Panel Testing. Ann Surg Oncol 2020; 27:4613-4621. [PMID: 32720048 DOI: 10.1245/s10434-020-08889-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 06/15/2020] [Indexed: 01/06/2023]
Abstract
BACKGROUND An increasing number of breast cancer patients are undergoing expanded genetic testing and are being identified as germline mutation carriers. We sought to determine rates of contralateral risk-reducing mastectomy (CRRM) in patients with various germline mutations. PATIENTS AND METHODS All women ≥ 18 years of age with unilateral breast cancer who underwent multigene panel testing between January 1, 2014 and August 1, 2019 at our academic institution were identified. Demographic, tumor, and treatment variables were identified from the medical record. Multivariable analyses were performed to compare factors associated with performance of CRRM. RESULTS We identified 1613 patients, of whom 28.1% had a pathogenic variant and 40.1% had variants of uncertain significance (VUS). Overall, 420 patients (26.0%) underwent a CRRM. On multivariable analysis, factors associated with CRRM included age < 50 years (OR 3.8, 95% CI 3.0, 5.0), race (OR 0.5, 95% CI 0.3, 0.7 and OR 0.4, 95% CI 0.2, 0.7 for Black and Asian women, respectively, versus White women), and the presence of any germline mutation or VUS (OR 13.2, 95% CI 8.7, 20.2 for BRCA1/2; OR 3.9, 95% CI 2.7, 5.8 for non-BRCA germline mutation; and OR 1.8, 95% CI 1.3, 2.6 for VUS). CONCLUSIONS In breast cancer patients who undergo multigene panel testing, a sizeable number of women with pathogenic non-BRCA germline findings are opting for CRRM. Given that the risk of contralateral breast cancer in women with most pathogenic mutations other than BRCA1/2 remains poorly characterized, these data have implications for risk counseling and for ascertaining the true risks of contralateral breast cancer in this population.
Collapse
Affiliation(s)
- Brittany L Murphy
- Department of Breast Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Min Yi
- Department of Breast Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Banu K Arun
- Department of Breast Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Isabelle Bedrosian
- Department of Breast Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
37
|
Sciaraffa T, Guido B, Khan SA, Kulkarni S. Breast cancer risk assessment and management programs: A practical guide. Breast J 2020; 26:1556-1564. [PMID: 32662170 DOI: 10.1111/tbj.13967] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 01/22/2020] [Indexed: 11/28/2022]
Abstract
Breast cancer risk assessment continues to evolve as emerging knowledge of breast cancer risk drivers and modifiers enables better identification of high-risk women who may benefit from increased screening or targeted risk-reduction protocols. The ongoing development of breast cancer Risk Assessment and Management Programs (RAMPs) presents an opportunity to decrease breast cancer disease incidence with evidence-based interventions. The goal of this review was to provide a practical guide for providers seeking to establish or update a breast cancer risk assessment and management program. We outline genetic/familial, personal, reproductive, and lifestyle-related factors while discussing the incorporation of risk modeling for precise risk estimate personalization. We further describe the process for determining a risk management plan: information gathering, generation of a risk profile, and articulation and implementation of risk reduction. We also include an overview of clinical workflows in breast cancer management programs and underlines the logistics of establishing a program as well as general principles for guiding the formulation of an individualized risk management plan. We discuss practical considerations, such as clinic structure and operation, allocation of resources, and patient education. Other critical aspects of program design, including identification of the target population, delineation of the core components of the clinical experience, definition of provider roles, description of referral mechanisms, and the launching of a marketing plan are also addressed. The process of risk assessment is both anxiety-provoking and empowering for women at increased risk. New knowledge has enabled strategies to both understand the risk and control it through evidence-based risk management. These benefits can now be realized by an increasing number of unaffected, high-risk patients collaborating with risk management practitioners. Continuation of these efforts will lead to further progress in both risk stratification and risk management of women at elevated breast cancer risk in the near future.
Collapse
Affiliation(s)
- Theresa Sciaraffa
- Department of Obstetrics and Gynecology, Northwestern Memorial Hospital, Chicago, Illinois, USA
| | - Barbara Guido
- Department of Surgery, Northwestern Memorial Hospital, Chicago, Illinois, USA
| | - Seema A Khan
- Department of Surgery, Northwestern Memorial Hospital, Chicago, Illinois, USA
| | - Swati Kulkarni
- Department of Surgery, Northwestern Memorial Hospital, Chicago, Illinois, USA
| |
Collapse
|
38
|
Kaname T. A commentary on germline mutations of multiple breast cancer-related genes are differentially associated with triple-negative breast cancers and prognostic factors. J Hum Genet 2020; 65:589-590. [PMID: 32393812 DOI: 10.1038/s10038-020-0767-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 04/19/2020] [Indexed: 11/09/2022]
Affiliation(s)
- Tadashi Kaname
- Department of Genome Medicine, National Center for Child Health and Development, Tokyo, Japan.
| |
Collapse
|
39
|
Incorvaia L, Fanale D, Badalamenti G, Bono M, Calò V, Cancelliere D, Castiglia M, Fiorino A, Pivetti A, Barraco N, Cutaia S, Russo A, Bazan V. Hereditary Breast and Ovarian Cancer in Families from Southern Italy (Sicily)-Prevalence and Geographic Distribution of Pathogenic Variants in BRCA1/2 Genes. Cancers (Basel) 2020; 12:E1158. [PMID: 32380732 PMCID: PMC7280980 DOI: 10.3390/cancers12051158] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 05/01/2020] [Accepted: 05/03/2020] [Indexed: 12/20/2022] Open
Abstract
Recent advances in the detection of germline pathogenic variants (PVs) in BRCA1/2 genes have allowed a deeper understanding of the BRCA-related cancer risk. Several studies showed a significant heterogeneity in the prevalence of PVs across different populations. Because little is known about this in the Sicilian population, our study was aimed at investigating the prevalence and geographic distribution of inherited BRCA1/2 PVs in families from this specific geographical area of Southern Italy. We retrospectively collected and analyzed all clinical information of 1346 hereditary breast and/or ovarian cancer patients genetically tested for germline BRCA1/2 PVs at University Hospital Policlinico "P. Giaccone" of Palermo from January 1999 to October 2019. Thirty PVs were more frequently observed in the Sicilian population but only some of these showed a specific territorial prevalence, unlike other Italian and European regions. This difference could be attributed to the genetic heterogeneity of the Sicilian people and its historical background. Therefore hereditary breast and ovarian cancers could be predominantly due to BRCA1/2 PVs different from those usually detected in other geographical areas of Italy and Europe. Our investigation led us to hypothesize that a higher prevalence of some germline BRCA PVs in Sicily could be a population-specific genetic feature of BRCA-positive carriers.
Collapse
Affiliation(s)
- Lorena Incorvaia
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), 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; (D.F.); (G.B.); (M.B.); (V.C.); (D.C.); (M.C.); (A.F.); (A.P.); (N.B.); (S.C.); (V.B.)
| | - Giuseppe Badalamenti
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy; (D.F.); (G.B.); (M.B.); (V.C.); (D.C.); (M.C.); (A.F.); (A.P.); (N.B.); (S.C.); (V.B.)
| | - Marco Bono
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy; (D.F.); (G.B.); (M.B.); (V.C.); (D.C.); (M.C.); (A.F.); (A.P.); (N.B.); (S.C.); (V.B.)
| | - Valentina Calò
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy; (D.F.); (G.B.); (M.B.); (V.C.); (D.C.); (M.C.); (A.F.); (A.P.); (N.B.); (S.C.); (V.B.)
| | - Daniela Cancelliere
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy; (D.F.); (G.B.); (M.B.); (V.C.); (D.C.); (M.C.); (A.F.); (A.P.); (N.B.); (S.C.); (V.B.)
| | - Marta Castiglia
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy; (D.F.); (G.B.); (M.B.); (V.C.); (D.C.); (M.C.); (A.F.); (A.P.); (N.B.); (S.C.); (V.B.)
| | - Alessia Fiorino
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy; (D.F.); (G.B.); (M.B.); (V.C.); (D.C.); (M.C.); (A.F.); (A.P.); (N.B.); (S.C.); (V.B.)
| | - Alessia Pivetti
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy; (D.F.); (G.B.); (M.B.); (V.C.); (D.C.); (M.C.); (A.F.); (A.P.); (N.B.); (S.C.); (V.B.)
| | - Nadia Barraco
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy; (D.F.); (G.B.); (M.B.); (V.C.); (D.C.); (M.C.); (A.F.); (A.P.); (N.B.); (S.C.); (V.B.)
| | - Sofia Cutaia
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy; (D.F.); (G.B.); (M.B.); (V.C.); (D.C.); (M.C.); (A.F.); (A.P.); (N.B.); (S.C.); (V.B.)
| | - Antonio Russo
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy; (D.F.); (G.B.); (M.B.); (V.C.); (D.C.); (M.C.); (A.F.); (A.P.); (N.B.); (S.C.); (V.B.)
| | - Viviana Bazan
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy; (D.F.); (G.B.); (M.B.); (V.C.); (D.C.); (M.C.); (A.F.); (A.P.); (N.B.); (S.C.); (V.B.)
| |
Collapse
|
40
|
Yadav S, Hu C, Hart SN, Boddicker N, Polley EC, Na J, Gnanaolivu R, Lee KY, Lindstrom T, Armasu S, Fitz-Gibbon P, Ghosh K, Stan DL, Pruthi S, Neal L, Sandhu N, Rhodes DJ, Klassen C, Peethambaram PP, Haddad TC, Olson JE, Hoskin TL, Goetz MP, Domchek SM, Boughey JC, Ruddy KJ, Couch FJ. Evaluation of Germline Genetic Testing Criteria in a Hospital-Based Series of Women With Breast Cancer. J Clin Oncol 2020; 38:1409-1418. [PMID: 32125938 PMCID: PMC7193748 DOI: 10.1200/jco.19.02190] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/17/2020] [Indexed: 12/19/2022] Open
Abstract
PURPOSE To determine the sensitivity and specificity of genetic testing criteria for the detection of germline pathogenic variants in women with breast cancer. MATERIALS AND METHODS Women with breast cancer enrolled in a breast cancer registry at a tertiary cancer center between 2000 and 2016 were evaluated for germline pathogenic variants in 9 breast cancer predisposition genes (ATM, BRCA1, BRCA2, CDH1, CHEK2, NF1, PALB2, PTEN, and TP53). The performance of the National Comprehensive Cancer Network (NCCN) hereditary cancer testing criteria was evaluated relative to testing of all women as recommended by the American Society of Breast Surgeons. RESULTS Of 3,907 women, 1,872 (47.9%) meeting NCCN criteria were more likely to carry a pathogenic variant in 9 predisposition genes compared with women not meeting criteria (9.0% v 3.5%; P < .001). Of those not meeting criteria (n = 2,035), 14 (0.7%) had pathogenic variants in BRCA1 or BRCA2. The sensitivity of NCCN criteria was 70% for 9 predisposition genes and 87% for BRCA1 and BRCA2, with a specificity of 53%. Expansion of the NCCN criteria to include all women diagnosed with breast cancer at ≤ 65 years of age achieved > 90% sensitivity for the 9 predisposition genes and > 98% sensitivity for BRCA1 and BRCA2. CONCLUSION A substantial proportion of women with breast cancer carrying germline pathogenic variants in predisposition genes do not qualify for testing by NCCN criteria. Expansion of NCCN criteria to include all women diagnosed at ≤ 65 years of age improves the sensitivity of the selection criteria without requiring testing of all women with breast cancer.
Collapse
Affiliation(s)
| | - Chunling Hu
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Steven N. Hart
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | | | - Eric C. Polley
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | - Jie Na
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | - Rohan Gnanaolivu
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | - Kun Y. Lee
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Tricia Lindstrom
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | - Sebastian Armasu
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | | | | | | | | | | | | | | | | | | | | | - Janet E. Olson
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | - Tanya L. Hoskin
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | | | - Susan M. Domchek
- Perelman School of Medicine, University of Pennsylvania, and Basser Center for BRCA, Philadelphia, PA
| | | | | | - Fergus J. Couch
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| |
Collapse
|