51
|
Park EJ, Kim HD, Choi EK, Hoe KL, Kim DU. Co-treatment of birinapant with TRAIL synergistically induces apoptosis by downregulating cFLIP(L) in MDA-MB-453 cell lines. Biochem Biophys Res Commun 2020; 533:289-295. [PMID: 32958259 DOI: 10.1016/j.bbrc.2020.09.031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 09/11/2020] [Indexed: 12/24/2022]
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has received much attention owing to its ability to specifically induce cell death in cancer. However, several types of cancer, including some forms of breast cancer, are resistant to TRAIL. Various chemotherapeutic agents, phytochemicals, and TRAIL combination therapies have been proposed to resolve TRAIL resistance. Here, we explored the sensitization effect of birinapant on TRAIL-induced apoptosis in the MDA-MB-453 cell line. Although neither birinapant nor TRAIL showed any cytotoxic effect when used alone, apoptosis was induced when birinapant and TRAIL were used together. Our data suggest that the combination of birinapant and TRAIL induces downregulation of FLICE-like inhibitory protein (cFLIP) (L) protein expression. Interestingly, cFLIP(L) overexpression reversed apoptosis caused by co-treatment with TRAIL. Taken together, our results indicate that a combination of birinapant and TRAIL may be a promising treatment for TRAIL-resistant breast cancer.
Collapse
Affiliation(s)
- Eun Jung Park
- Rare Disease Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Hae Dong Kim
- Department of New Drug Discovery and Development, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Eun Kyoung Choi
- Rare Disease Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea.
| | - Kwang-Lae Hoe
- Department of New Drug Discovery and Development, Chungnam National University, Daejeon, 34134, Republic of Korea.
| | - Dong-Uk Kim
- Rare Disease Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea.
| |
Collapse
|
52
|
Canada follows the US in the rise of bilateral mastectomies for unilateral breast cancer: a 23-year population cohort study. Breast Cancer Res Treat 2020; 185:517-525. [PMID: 33128192 DOI: 10.1007/s10549-020-05965-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 10/05/2020] [Indexed: 12/11/2022]
Abstract
INTRODUCTION The use of contralateral prophylactic mastectomy (CPM) continues to grow despite the absence of evidence supporting a survival benefit. This study's objectives were to (1) describe the trends in the rates of unilateral and bilateral mastectomy (BM) in women diagnosed with unilateral breast cancer (UBC) in Ontario, Canada from 1991 to 2013, and (2) identify factors associated with BM to treat UBC. METHODS This retrospective cohort analysis included all women aged 18 and older diagnosed with UBC between January 1991 and December 2013. Health administrative data from the Institute for Clinical Evaluative Sciences, the Ontario Cancer Registry, and the Discharge Abstract Database were used to identify all breast cancer and mastectomy cases. Age-adjusted mastectomy rates were plotted over time. Univariable and multivariable analyses included clinically significant covariates. RESULTS From 1991 to 2013 there were 172,165 cases of UBC and 64,886 mastectomies (37.7%) performed in Ontario. 13.6% of the mastectomies were bilateral. BM rates increased over sixfold (from 4 to 25%) across all age groups under age 70 over a 23-year period. On multivariable analysis, younger age, higher income, rural community, earlier breast cancer stage, lobular histology, availability of reconstruction and teaching hospitals were associated with increased odds of BM. CONCLUSIONS This is the largest population study of breast cancer patients in Canada and shows an increasing rate of BM for UBC. The results are similar to those already described in the US and highlight the importance of continued efforts to promote efficient communication and evidence-based decision-making prior to breast surgery.
Collapse
|
53
|
Tuazon AMDA, Lott P, Bohórquez M, Benavides J, Ramirez C, Criollo A, Estrada-Florez A, Mateus G, Velez A, Carmona J, Olaya J, Garcia E, Polanco-Echeverry G, Stultz J, Alvarez C, Tapia T, Ashton-Prolla P, Vega A, Lazaro C, Tornero E, Martinez-Bouzas C, Infante M, De La Hoya M, Diez O, Browning BL, Rannala B, Teixeira MR, Carvallo P, Echeverry M, Carvajal-Carmona LG. Haplotype analysis of the internationally distributed BRCA1 c.3331_3334delCAAG founder mutation reveals a common ancestral origin in Iberia. Breast Cancer Res 2020; 22:108. [PMID: 33087180 PMCID: PMC7579869 DOI: 10.1186/s13058-020-01341-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 09/16/2020] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND The BRCA1 c.3331_3334delCAAG founder mutation has been reported in hereditary breast and ovarian cancer families from multiple Hispanic groups. We aimed to evaluate BRCA1 c.3331_3334delCAAG haplotype diversity in cases of European, African, and Latin American ancestry. METHODS BC mutation carrier cases from Colombia (n = 32), Spain (n = 13), Portugal (n = 2), Chile (n = 10), Africa (n = 1), and Brazil (n = 2) were genotyped with the genome-wide single nucleotide polymorphism (SNP) arrays to evaluate haplotype diversity around BRCA1 c.3331_3334delCAAG. Additional Portuguese (n = 13) and Brazilian (n = 18) BC mutation carriers were genotyped for 15 informative SNPs surrounding BRCA1. Data were phased using SHAPEIT2, and identical by descent regions were determined using BEAGLE and GERMLINE. DMLE+ was used to date the mutation in Colombia and Iberia. RESULTS The haplotype reconstruction revealed a shared 264.4-kb region among carriers from all six countries. The estimated mutation age was ~ 100 generations in Iberia and that it was introduced to South America early during the European colonization period. CONCLUSIONS Our results suggest that this mutation originated in Iberia and later introduced to Colombia and South America at the time of Spanish colonization during the early 1500s. We also found that the Colombian mutation carriers had higher European ancestry, at the BRCA1 gene harboring chromosome 17, than controls, which further supported the European origin of the mutation. Understanding founder mutations in diverse populations has implications in implementing cost-effective, ancestry-informed screening.
Collapse
Affiliation(s)
| | - Paul Lott
- Genome Center, University of California Davis, Davis, CA USA
| | | | | | | | | | | | | | - Alejandro Velez
- Hospital Pablo Tobon Uribe, Medellín, Colombia
- Dinamica IPS, Medellín, Colombia
| | | | - Justo Olaya
- Hospital Universitario Hernando Moncaleano Perdomo, Neiva, Colombia
| | - Elisha Garcia
- Genome Center, University of California Davis, Davis, CA USA
| | | | - Jacob Stultz
- Genome Center, University of California Davis, Davis, CA USA
| | | | - Teresa Tapia
- Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Patricia Ashton-Prolla
- Department of Genetics, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- Post-graduate Course in Genetics and Molecular Biology, UFRGS, Porto Alegre, Brazil
- Medical Genetics Service, Hospital de Clinicas de Porto Alegre (HCPA), Porto Alegre, Brazil
| | - Ana Vega
- Fundación Pública Galega de Medicina Xenómica, Grupo de Medicina Xenómica-USC, CIBERER, IDIS, Santiago de Compostela, Spain
| | - Conxi Lazaro
- Hereditary Cancer Program, Catalan Institute of Oncology, Oncobell Program-IDIBELL, Hospitalet de Llobregat, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Eva Tornero
- Hereditary Cancer Program, Catalan Institute of Oncology, Oncobell Program-IDIBELL, Hospitalet de Llobregat, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | | | - Mar Infante
- Cancer Genetics Group, Institute of Genetics and Molecular Biology (UVa-CSIC), Valladolid, Spain
| | - Miguel De La Hoya
- Laboratorio de Oncología Molecular, Hospital Clínico San Carlos. IdISSC (Instituto de Investigación Sanitaria San Carlos), Madrid, Spain
| | - Orland Diez
- Grupo de Cáncer Hereditario, Instituto Oncológico Vall d’Hebron (VHIO), Madrid, Spain
| | - Brian L. Browning
- Department of Medicine, Division of Medical Genetics, University of Washington, Seattle, WA USA
| | - Bruce Rannala
- Department of Evolution and Ecology, University of California Davis, Davis, CA USA
| | - Manuel R. Teixeira
- Portuguese Oncology Institute of Porto (IPO Porto) and Biomedical Sciences Institute (ICBAS), University of Porto, Porto, Portugal
| | - Pilar Carvallo
- Pontificia Universidad Católica de Chile, Santiago, Chile
| | | | - Luis G. Carvajal-Carmona
- Genome Center, University of California Davis, Davis, CA USA
- Division de Investigaciones, Fundacion de Genética y Genómica, Ibague, Colombia
- University of California Davis Comprehensive Cancer Center, Sacramento, CA, USA
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, USA
| |
Collapse
|
54
|
Dhar S, Datta A, Brosh RM. DNA helicases and their roles in cancer. DNA Repair (Amst) 2020; 96:102994. [PMID: 33137625 DOI: 10.1016/j.dnarep.2020.102994] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 09/28/2020] [Indexed: 12/15/2022]
Abstract
DNA helicases, known for their fundamentally important roles in genomic stability, are high profile players in cancer. Not only are there monogenic helicase disorders with a strong disposition to cancer, it is well appreciated that helicase variants are associated with specific cancers (e.g., breast cancer). Flipping the coin, DNA helicases are frequently overexpressed in cancerous tissues and reduction in helicase gene expression results in reduced proliferation and growth capacity, as well as DNA damage induction and apoptosis of cancer cells. The seminal roles of helicases in the DNA damage and replication stress responses, as well as DNA repair pathways, validate their vital importance in cancer biology and suggest their potential values as targets in anti-cancer therapy. In recent years, many laboratories have characterized the specialized roles of helicase to resolve transcription-replication conflicts, maintain telomeres, mediate cell cycle checkpoints, remodel stalled replication forks, and regulate transcription. In vivo models, particularly mice, have been used to interrogate helicase function and serve as a bridge for preclinical studies that may lead to novel therapeutic approaches. In this review, we will summarize our current knowledge of DNA helicases and their roles in cancer, emphasizing the latest developments.
Collapse
Affiliation(s)
- Srijita Dhar
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Arindam Datta
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Robert M Brosh
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA.
| |
Collapse
|
55
|
Robbe J, Moretta J, Vicier C, Sabatier R, Noguès C, Gonçalves A. Inhibiteurs de PARP dans les cancers du sein : développement clinique actuel et perspectives. Bull Cancer 2020; 107:1024-1041. [DOI: 10.1016/j.bulcan.2020.07.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 06/25/2020] [Accepted: 07/05/2020] [Indexed: 12/16/2022]
|
56
|
Diéras V, Han HS, Kaufman B, Wildiers H, Friedlander M, Ayoub JP, Puhalla SL, Bondarenko I, Campone M, Jakobsen EH, Jalving M, Oprean C, Palácová M, Park YH, Shparyk Y, Yañez E, Khandelwal N, Kundu MG, Dudley M, Ratajczak CK, Maag D, Arun BK. Veliparib with carboplatin and paclitaxel in BRCA-mutated advanced breast cancer (BROCADE3): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol 2020; 21:1269-1282. [PMID: 32861273 DOI: 10.1016/s1470-2045(20)30447-2] [Citation(s) in RCA: 209] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/17/2020] [Accepted: 06/19/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND BRCA1 or BRCA2-mutated breast cancers are sensitive to poly(ADP-ribose) polymerase (PARP) inhibitors and platinum agents owing to deficiency in homologous recombination repair of DNA damage. In this trial, we compared veliparib versus placebo in combination with carboplatin and paclitaxel, and continued as monotherapy if carboplatin and paclitaxel were discontinued before progression, in patients with HER2-negative advanced breast cancer and a germline BRCA1 or BRCA2 mutation. METHODS BROCADE3 was a randomised, double-blind, placebo-controlled, phase 3 trial done at 147 hospitals in 36 countries. Eligible patients (aged ≥18 years) had deleterious germline BRCA1 or BRCA2 mutation-associated, histologically or cytologically confirmed advanced HER2-negative breast cancer, an Eastern Cooperative Oncology Group performance status of 0-2, and had received up to two previous lines of chemotherapy for metastatic disease. Patients were randomly assigned (2:1) by interactive response technology by means of permuted blocks within strata (block size of 3 or 6) to carboplatin (area under the concentration curve 6 mg/mL per min intravenously) on day 1 and paclitaxel (80 mg/m2 intravenously) on days 1, 8, and 15 of 21-day cycles combined with either veliparib (120 mg orally twice daily, on days -2 to 5) or matching placebo. If patients discontinued carboplatin and paclitaxel before progression, they could continue veliparib or placebo at an intensified dose (300 mg twice daily continuously, escalating to 400 mg twice daily if tolerated) until disease progression. Patients in the control group could receive open-label veliparib monotherapy after disease progression. Randomisation was stratified by previous platinum use, history of CNS metastases, and oestrogen and progesterone receptor status. The primary endpoint was investigator-assessed progression-free survival per Response Evaluation Criteria in Solid Tumors version 1.1. Efficacy analyses were done by intention to treat, which included all randomly assigned patients with a centrally confirmed BRCA mutation, and safety analyses included all patients who received at least one dose of velilparib or placebo. This study is ongoing and is registered with ClinicalTrials.gov, NCT02163694. FINDINGS Between July 30, 2014, and Jan 17, 2018, 2202 patients were screened, of whom 513 eligible patients were enrolled and randomly assigned. In the intention-to-treat population (n=509), 337 patients were assigned to receive veliparib plus carboplatin-paclitaxel (veliparib group) and 172 were assigned to receive placebo plus carboplatin-paclitaxel (control group). Median follow-up at data cutoff (April 5, 2019) was 35·7 months (IQR 24·9-43·6) in the veliparib group and 35·5 months (23·1-45·9) in the control group. Median progression-free survival was 14·5 months (95% CI 12·5-17·7) in the veliparib group versus 12·6 months (10·6-14·4) in the control group (hazard ratio 0·71 [95% CI 0·57-0·88], p=0·0016). The most common grade 3 or worse adverse events were neutropenia (272 [81%] of 336 patients in the veliparib group vs 143 [84%] of 171 patients in the control group), anaemia (142 [42%] vs 68 [40%]), and thrombocytopenia (134 [40%] vs 48 [28%]). Serious adverse events occurred in 115 (34%) patients in the veliparib group versus 49 (29%) patients in the control group. There were no study drug-related deaths. INTERPRETATION The addition of veliparib to a highly active platinum doublet, with continuation as monotherapy if the doublet were discontinued, resulted in significant and durable improvement in progression-free survival in patients with germline BRCA mutation-associated advanced breast cancer. These data indicate the utility of combining platinum and PARP inhibitors in this patient population. FUNDING AbbVie.
Collapse
Affiliation(s)
- Véronique Diéras
- Institut Curie, Paris, France; Breast Oncology, Centre Eugène Marquis, Rennes, France.
| | - Hyo S Han
- Moffitt Cancer Center, Tampa, FL, USA
| | - Bella Kaufman
- Tel-Aviv University, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Hans Wildiers
- Department of General Medical Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Michael Friedlander
- Prince of Wales Clinical School UNSW and Prince of Wales Hospital, Sydney, NSW, Australia
| | - Jean-Pierre Ayoub
- Centre Hospitalier de l'Université de Montréal, Montreal, QC, Canada
| | | | - Igor Bondarenko
- Dnipropetrovsk Medical Academy, City Clinical Hospital Number 4, Dnipro, Ukraine
| | - Mario Campone
- Institut de Cancérologie de l'Ouest Saint-Herblain, France
| | | | - Mathilde Jalving
- University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Cristina Oprean
- University of Medicine and Pharmacy Timisoara and Oncomed SRL, Timisoara, Romania
| | | | | | - Yaroslav Shparyk
- Lviv State Regional Treatment and Diagnostic Oncology Center, Lviv, Ukraine
| | | | | | | | | | | | | | - Banu K Arun
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
57
|
Ataseven B, Tripon D, Rhiem K, Harter P, Schneider S, Heitz F, Baert T, Traut A, Pauly N, Ehmann S, Plett H, Schmutzler RK, du Bois A. Prevalence of BRCA1 and BRCA2 Mutations in Patients with Primary Ovarian Cancer - Does the German Checklist for Detecting the Risk of Hereditary Breast and Ovarian Cancer Adequately Depict the Need for Consultation? Geburtshilfe Frauenheilkd 2020; 80:932-940. [PMID: 32905297 PMCID: PMC7467803 DOI: 10.1055/a-1222-0042] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 07/14/2020] [Indexed: 01/09/2023] Open
Abstract
BackgroundBRCA1/2
mutations are the leading cause of hereditary epithelial ovarian cancer (EOC). The German Consortium for Hereditary Breast and Ovarian Cancer has defined inclusion criteria, which are retrievable as a checklist and facilitate genetic counselling/testing for affected persons with a mutation probability of ≥ 10%. Our objective was to evaluate the prevalence of the
BRCA1/2
mutation(s) based on the checklist score (CLS).
Methods
A retrospective data analysis was performed on EOC patients with a primary diagnosis treated between 1/2011 – 5/2019 at the Central Essen Clinics, where a
BRCA1/2
genetic analysis result and a CLS was available. Out of 545 cases with a
BRCA1/2
result (cohort A), 453 cases additionally had an extended gene panel result (cohort B).
Results
A
BRCA1/2
mutation was identified in 23.3% (127/545) in cohort A, pathogenic mutations in non-
BRCA1/2
genes were revealed in a further 6.2% in cohort B. In cohort A, 23.3% (127/545) of patients had a
BRCA1
(n = 92) or
BRCA2
(n = 35) mutation. Singular EOC (CLS 2) was present in 40.9%. The prevalence for a
BRCA1/2
mutation in cohort A was 10.8%, 17.2%, 25.0%, 35.1%, 51.4% and 66.7% for patients with CLS 2, 3, 4, 5, 6 and ≥ 7 respectively. The mutation prevalence in cohort B was 15.9%, 16.4%, 28.2%, 40.4%, 44.8% and 62.5% for patients with CLS 2, 3, 4, 5, 6 and ≥ 7 respectively.
Conclusions
The
BRCA1/2
mutation prevalence in EOC patients positively correlates with a rising checklist score. Already with singular EOC, the prevalence of a
BRCA1/2
mutation exceeds the required 10% threshold. Our data support the recommendation of the S3 guidelines Ovarian Cancer of offering genetic testing to all patients with EOC. Optimisation of the checklist with clear identification of the testing indication in this population should therefore be aimed for.
Collapse
Affiliation(s)
- Beyhan Ataseven
- Abteilung für Gynäkologie und Gynäkologische Onkologie, Evang. Kliniken Essen-Mitte, Essen.,Klinik und Poliklinik für Frauenheilkunde und Geburtshilfe der LMU München, München
| | - Denise Tripon
- Abteilung für Gynäkologie und Gynäkologische Onkologie, Evang. Kliniken Essen-Mitte, Essen
| | - Kerstin Rhiem
- Zentrum Familiärer Brust- und Eierstockkrebs, Universitätsklinik Köln, Köln
| | - Philipp Harter
- Abteilung für Gynäkologie und Gynäkologische Onkologie, Evang. Kliniken Essen-Mitte, Essen
| | - Stephanie Schneider
- Abteilung für Gynäkologie und Gynäkologische Onkologie, Evang. Kliniken Essen-Mitte, Essen
| | - Florian Heitz
- Abteilung für Gynäkologie und Gynäkologische Onkologie, Evang. Kliniken Essen-Mitte, Essen.,Klinik für Gynäkologie mit Zentrum für onkologische Chirurgie (CVK) und Klinik für Gynäkologie (CBF), Charité - Universitätsmedizin Berlin, Berlin
| | - Thais Baert
- Abteilung für Gynäkologie und Gynäkologische Onkologie, Evang. Kliniken Essen-Mitte, Essen.,Abteilung für Onkologie und Tumorimmunologie, KU Leuven, Leuven, Belgien
| | - Alexander Traut
- Abteilung für Gynäkologie und Gynäkologische Onkologie, Evang. Kliniken Essen-Mitte, Essen
| | - Nina Pauly
- Abteilung für Gynäkologie und Gynäkologische Onkologie, Evang. Kliniken Essen-Mitte, Essen
| | - Sarah Ehmann
- Abteilung für Gynäkologie und Gynäkologische Onkologie, Evang. Kliniken Essen-Mitte, Essen
| | - Helmut Plett
- Abteilung für Gynäkologie und Gynäkologische Onkologie, Evang. Kliniken Essen-Mitte, Essen.,Klinik für Gynäkologie mit Zentrum für onkologische Chirurgie (CVK) und Klinik für Gynäkologie (CBF), Charité - Universitätsmedizin Berlin, Berlin
| | - Rita K Schmutzler
- Zentrum Familiärer Brust- und Eierstockkrebs, Universitätsklinik Köln, Köln
| | - Andreas du Bois
- Abteilung für Gynäkologie und Gynäkologische Onkologie, Evang. Kliniken Essen-Mitte, Essen
| |
Collapse
|
58
|
Bakkach J, Mansouri M, Derkaoui T, Loudiyi A, El Fahime E, Barakat A, Ghailani Nourouti N, Martinez De Villarreal J, Cortijo Bringas C, Bennani Mechita M. Contribution of BRCA1 and BRCA2 germline mutations to early onset breast cancer: a series from north of Morocco. BMC Cancer 2020; 20:859. [PMID: 32894085 PMCID: PMC7487731 DOI: 10.1186/s12885-020-07352-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 08/27/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND To date, the contribution of BRCA1/2 mutations in Moroccan early onset breast cancer patients remains unknown. Here we assess these genetic alterations for the first time in a cohort from North of Morocco. METHODS Thirty-three patients diagnosed with breast cancer at the age of ≤40 years were recruited irrespective of breast and/or ovarian cancer family history. Coding regions and intron-exon boundaries of BRCA1 and BRCA2 genes were sequenced from peripheral blood DNA using Ion Proton (Thermo Fisher Scientific) next generation sequencing platform. RESULTS Overall, five BRCA germline mutations were identified (15.1%). The frequency of mutations among patients with family history of breast cancer was 16.7%. Three mutations were found in BRCA1 (9%) and two within the BRCA2 gene (6%). These are three frameshift mutations (c.798_799del, c.2125_2126insA, c.5116_5119delAATA), one missense (c.116G > A) and one nonsense mutation (c.289G > T). The mutation c.5116_5119delAATA has a founder effect in North Africa. Moreover, one variant of unknown significance was identified in BRCA2 (c.4090A > G). Most BRCA mutations carriers (80%) had no family history of breast cancer. CONCLUSION Our data do not support the hypothesis that BRCA mutations alone explain the higher frequency of breast cancer in Moroccan young women. The young age (≤40 years) for breast cancer diagnosis seems to be strongly predictive of BRCA mutation status in Moroccan patients. These results will help in decision making with regard to genetic counseling and testing in the national scale.
Collapse
Affiliation(s)
- Joaira Bakkach
- Biomedical Genomics and Oncogenetics Research Laboratory, Faculty of Sciences and Techniques of Tangier, University Abdelmalek Essaâdi, P.A: 416-Tangier, Tangier, Morocco
| | - Mohamed Mansouri
- Biomedical Genomics and Oncogenetics Research Laboratory, Faculty of Sciences and Techniques of Tangier, University Abdelmalek Essaâdi, P.A: 416-Tangier, Tangier, Morocco
- Oncology Clinic Al AMAL of Tangier, Tangier, Morocco
| | - Touria Derkaoui
- Biomedical Genomics and Oncogenetics Research Laboratory, Faculty of Sciences and Techniques of Tangier, University Abdelmalek Essaâdi, P.A: 416-Tangier, Tangier, Morocco
| | - Ali Loudiyi
- Oncology Clinic Al AMAL of Tangier, Tangier, Morocco
| | - ElMostafa El Fahime
- Functional Genomic Plateform, Units of Technical Support to Scientific Research, National Center of Scientific and Technical Research, Rabat, Morocco
| | - Amina Barakat
- Biomedical Genomics and Oncogenetics Research Laboratory, Faculty of Sciences and Techniques of Tangier, University Abdelmalek Essaâdi, P.A: 416-Tangier, Tangier, Morocco
| | - Naima Ghailani Nourouti
- Biomedical Genomics and Oncogenetics Research Laboratory, Faculty of Sciences and Techniques of Tangier, University Abdelmalek Essaâdi, P.A: 416-Tangier, Tangier, Morocco
| | | | | | - Mohcine Bennani Mechita
- Biomedical Genomics and Oncogenetics Research Laboratory, Faculty of Sciences and Techniques of Tangier, University Abdelmalek Essaâdi, P.A: 416-Tangier, Tangier, Morocco
| |
Collapse
|
59
|
Nomograms for prediction of overall and cancer-specific survival in young breast cancer. Breast Cancer Res Treat 2020; 184:597-613. [PMID: 32886273 DOI: 10.1007/s10549-020-05870-5] [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: 02/29/2020] [Accepted: 08/08/2020] [Indexed: 12/24/2022]
Abstract
PURPOSE To assess the prognostic risk factors and establish prognostic nomograms based on lymph node ratio (LNR) to predict the survival of young patients with breast cancer (BC). METHODS Patients aged < 40 years and diagnosed with BC between 2010 and 2016 from the Surveillance, Epidemiology and End Results database were assessed. Nomograms incorporating LNR were constructed to predict overall survival (OS) and breast cancer-specific survival (BCSS) based on Cox proportional hazards model. The performance of the nomograms was assessed by C-index, calibration curves, receiver operating characteristic (ROC) curves, decision curve analysis (DCA), and risk group stratification and compared with the TNM staging system. RESULTS Based on the univariate and multivariate Cox regression analysis, significant prognostic factors were identified and integrated to create the nomograms for OS and BCSS. The calibration curves indicated optimal agreement between model predictions and actual observations. The nomograms showed favorable sensitivity with a C-index of 0.8351 (95% CI 0.8234-0.8469) for OS and 0.8474 (95% CI 0.8355-0.8594) for BCSS. The ROC curves of the nomograms showed better predictive ability than those of the TNM staging system for OS (AUC: 0.8503 vs. 0.7819) and BSCC (AUC: 0.8607 vs. 0.8081). Significant differences in Kaplan-Meier curves were observed in patients stratified into different risk groups (p < 0.001). CONCLUSIONS These nomograms provided more accurate individualized risk prediction of OS and BCSS and may assist clinicians in making decisions for young patients with BC.
Collapse
|
60
|
Malhotra H, Kowtal P, Mehra N, Pramank R, Sarin R, Rajkumar T, Gupta S, Bapna A, Bhattacharyya GS, Gupta S, Maheshwari A, Mannan AU, Reddy Kundur R, Sekhon R, Singhal M, Smruti B, SP S, Suryavanshi M, Verma A. Genetic Counseling, Testing, and Management of HBOC in India: An Expert Consensus Document from Indian Society of Medical and Pediatric Oncology. JCO Glob Oncol 2020; 6:991-1008. [PMID: 32628584 PMCID: PMC7392772 DOI: 10.1200/jgo.19.00381] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/21/2020] [Indexed: 01/08/2023] Open
Abstract
PURPOSE Hereditary breast and ovarian cancer (HBOC) syndrome is primarily characterized by mutations in the BRCA1/2 genes. There are several barriers to the implementation of genetic testing and counseling in India that may affect clinical decisions. These consensus recommendations were therefore convened as a collaborative effort to improve testing and management of HBOC in India. DESIGN Recommendations were developed by a multidisciplinary group of experts from the Indian Society of Medical and Pediatric Oncology and some invited experts on the basis of graded evidence from the literature and using a formal Delphi process to help reach consensus. PubMed and Google Scholar databases were searched to source relevant articles. RESULTS This consensus statement provides practical insight into identifying patients who should undergo genetic counseling and testing on the basis of assessments of family and ancestry and personal history of HBOC. It discusses the need and significance of genetic counselors and medical professionals who have the necessary expertise in genetic counseling and testing. Recommendations elucidate requirements of pretest counseling, including discussions on genetic variants of uncertain significance and risk reduction options. The group of experts recommended single-site mutation testing in families with a known mutation and next-generation sequencing coupled with multiplex ligation probe amplification for the detection of large genomic rearrangements for unknown mutations. Recommendations for surgical and lifestyle-related risk reduction approaches and management using poly (ADP-ribose) polymerase inhibitors are also detailed. CONCLUSION With rapid strides being made in the field of genetic testing/counseling in India, more oncologists are expected to include genetic testing/counseling as part of their clinical practice. These consensus recommendations are anticipated to help homogenize genetic testing and management of HBOC in India for improved patient care.
Collapse
Affiliation(s)
- Hemant Malhotra
- Department of Medical Oncology, Sri Ram Cancer Center, Mahatma Gandhi Medical College Hospital, Jaipur, India
| | - Pradnya Kowtal
- Sarin Laboratory and OIC Sanger Sequencing Facility, Advanced Centre for Treatment Research, and Education in Cancer, Navi Mumbai, India
| | - Nikita Mehra
- Department of Medical Oncology, Cancer Institute (WIA), Chennai, India
| | - Raja Pramank
- Department of Medical Oncology, All India Institute of Medical Sciences, New Delhi, India
| | - Rajiv Sarin
- Radiation Oncology, Cancer Genetics Unit, Tata Memorial Centre and PI Sarin Laboratory, Advanced Centre for Treatment Research and Education in Cancer, Navi Mumbai, India
| | | | - Sudeep Gupta
- Tata Memorial Centre Advanced Centre for Treatment, Research, and Education in Cancer, Navi Mumbai, India
| | - Ajay Bapna
- Department of Medical Oncology, Bhagwan Mahavir Cancer Hospital Research Center, Jaipur, India
| | | | - Sabhyata Gupta
- Department of Gynae Oncology, Medanta-The Medicity, Gurgaon, India
| | - Amita Maheshwari
- Department of Gynecologic Oncology, Tata Memorial Centre, Mumbai, India
| | - Ashraf U. Mannan
- Clinical Diagnostics, Strand Center for Genomics and Personalized Medicine, Strand Life Sciences, Bangalore, India
| | | | - Rupinder Sekhon
- Gynae Oncology, Rajiv Gandhi Cancer Institute and Research Centre, New Delhi, India
| | | | - B.K. Smruti
- Bombay Hospital and Medical Research Centre, Mumbai, India
| | - Somashekhar SP
- Manipal Comprehensive Cancer Center, Manipal Hospital, Bengaluru, India
| | - Moushumi Suryavanshi
- Molecular Diagnostics, Rajiv Gandhi Cancer Institute and Research Centre, New Delhi, India
| | - Amit Verma
- Molecular Oncology and Cancer Genetics, Max Hospital, New Delhi, India
| |
Collapse
|
61
|
Hughes E, Tshiaba P, Gallagher S, Wagner S, Judkins T, Roa B, Rosenthal E, Domchek S, Garber J, Lancaster J, Weitzel J, Kurian AW, Lanchbury JS, Gutin A, Robson M. Development and Validation of a Clinical Polygenic Risk Score to Predict Breast Cancer Risk. JCO Precis Oncol 2020; 4:PO.19.00360. [PMID: 32923876 PMCID: PMC7446363 DOI: 10.1200/po.19.00360] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/07/2020] [Indexed: 01/14/2023] Open
Abstract
PURPOSE Women with a family history of breast cancer are frequently referred for hereditary cancer genetic testing, yet < 10% are found to have pathogenic variants in known breast cancer susceptibility genes. Large-scale genotyping studies have identified common variants (primarily single-nucleotide polymorphisms [SNPs]) with individually modest breast cancer risk that, in aggregate, account for considerable breast cancer susceptibility. Here, we describe the development and empirical validation of an SNP-based polygenic breast cancer risk score. METHODS A panel of 94 SNPs was examined for association with breast cancer in women of European ancestry undergoing hereditary cancer genetic testing and negative for pathogenic variants in breast cancer susceptibility genes. Candidate polygenic risk scores (PRSs) as predictors of personal breast cancer history were developed through multivariable logistic regression models adjusted for age, cancer history, and ancestry. An optimized PRS was validated in 2 independent cohorts (n = 13,174; n = 141,160). RESULTS Within the training cohort (n = 24,259), 4,291 women (18%) had a personal history of breast cancer and 8,725 women (36%) reported breast cancer in a first-degree relative. The optimized PRS included 86 variants and was highly predictive of breast cancer status in both validation cohorts (P = 6.4 × 10-66; P < 10-325). The odds ratio (OR) per unit standard deviation was consistent between validations (OR, 1.45 [95% CI, 1.39 to 1.52]; OR 1.47 [95% CI, 1.45 to 1.49]). In a direct comparison, the 86-SNP PRS outperformed a previously described PRS of 77 SNPs. CONCLUSION The validation and implementation of a PRS for women without pathogenic variants in known breast cancer susceptibility genes offers potential for risk stratification to guide surveillance recommendations.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Susan Domchek
- University of Pennsylvania School of Medicine, Philadelphia, PA
| | | | | | | | | | | | | | - Mark Robson
- Memorial Sloan Kettering Cancer Center, New York City, NY
| |
Collapse
|
62
|
Gonçalves A, Bertucci A, Bertucci F. PARP Inhibitors in the Treatment of Early Breast Cancer: The Step Beyond? Cancers (Basel) 2020; 12:cancers12061378. [PMID: 32471249 PMCID: PMC7352970 DOI: 10.3390/cancers12061378] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/18/2020] [Accepted: 05/20/2020] [Indexed: 02/06/2023] Open
Abstract
Exquisitely exploiting defects in homologous recombination process, poly(ADP-ribose) polymerase (PARP) inhibitors have recently emerged as a promising class of therapeutics in human epidermal growth factor receptor 2 (HER2)-negative advanced breast cancer with germline breast cancer 1 (BRCA1) or breast cancer 2 (BRCA2) mutations (gBRCA1/2m). In this setting, PARP inhibitors, either as single agent or in combination with platinum-based chemotherapy, significantly increased progression-free survival, as compared to conventional chemotherapy. Accordingly, further therapeutic advances are expected at an earlier stage of the disease. In the neoadjuvant setting, veliparib failed to increase the pathological complete response rate when added to a carboplatin-based regimen, in unselected triple-negative breast cancer patients. Similarly, when administered before anthracycline-cyclophosphamide, the neoadjuvant olaparib-paclitaxel combination was not superior to carboplatin–paclitaxel, in patients with HER2-negative breast cancer and BRCA1/2 mutation, or homologous recombination defect. Yet, neoadjuvant talazoparib, administered as a single-agent in patients with HER2-negative breast cancer and germline BRCA1/2 mutation, achieved an impressive pathological complete response rate of nearly 50%. In the adjuvant setting, the results from the OlympiA phase III study, evaluating adjuvant olaparib in HER2-negative early breast cancer and germline BRCA1/2 mutations, are eagerly awaited. Ongoing trials should clarify whether PARP inhibitors might improve outcome when administered in the adjuvant or neoadjuvant setting in early breast cancer patients with BRCA1/2 mutation or homologous recombination defect.
Collapse
Affiliation(s)
- Anthony Gonçalves
- Medical Oncology Department, Cancer Research Center of Marseille (CRCM), INSERM U1068, CNRS UMR7258, Institut Paoli-Calmettes, Aix-Marseille University, 13009 Marseille, France; (A.B.); (F.B.)
- Predictive Oncology Laboratory, Cancer Research Center of Marseille (CRCM), Inserm U1068, CNRS UMR7258, Aix-Marseille University, 13009 Marseille, France
- Correspondence: ; Tel.: +33-4-91-22-37-89; Fax: +33-4-91-22-36-70
| | - Alexandre Bertucci
- Medical Oncology Department, Cancer Research Center of Marseille (CRCM), INSERM U1068, CNRS UMR7258, Institut Paoli-Calmettes, Aix-Marseille University, 13009 Marseille, France; (A.B.); (F.B.)
| | - François Bertucci
- Medical Oncology Department, Cancer Research Center of Marseille (CRCM), INSERM U1068, CNRS UMR7258, Institut Paoli-Calmettes, Aix-Marseille University, 13009 Marseille, France; (A.B.); (F.B.)
- Predictive Oncology Laboratory, Cancer Research Center of Marseille (CRCM), Inserm U1068, CNRS UMR7258, Aix-Marseille University, 13009 Marseille, France
| |
Collapse
|
63
|
Kim EK, Park SY, Kim SW. Clinicopathological characteristics of BRCA-associated breast cancer in Asian patients. J Pathol Transl Med 2020; 54:265-275. [PMID: 32397691 PMCID: PMC7385261 DOI: 10.4132/jptm.2020.04.07] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 04/07/2020] [Indexed: 12/20/2022] Open
Abstract
BRCA1/2 germline mutations account for the majority of hereditary breast cancers. Since the identification of the BRCA genes, several attempts have been made to define the clinicopathological characteristics of BRCA-associated breast cancer in comparison with sporadic breast cancer. Asians constitute 60% of the world population, and although the incidence of breast cancer in Asia remains low compared to the West, breast cancer is the most prevalent female cancer in the region. The epidemiological aspects of breast cancer are different between Asians and Caucasians. Asian patients present with breast cancer at a younger age than Western patients. The contributions of BRCA1/2 mutations to breast cancer incidence are expected to differ between Asians and Caucasians, and the different genetic backgrounds among races are likely to influence the breast cancer phenotypes. However, most large-scale studies on the clinicopathological characteristics of BRCA-associated breast cancer have been on Western patients, while studies on Asian populations were small and sporadic. In this review, we provide an overview of the clinical and pathological characteristics of BRCA-associated breast cancer, incorporating findings on Asian patients.
Collapse
Affiliation(s)
- Eun-Kyu Kim
- Department of Surgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - So Yeon Park
- Department of Pathology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Sung-Won Kim
- Department of Surgery, Daerim St. Mary's Hospital, Seoul, Korea
| |
Collapse
|
64
|
Genetic Testing for Cancer Predisposition Syndromes in Adolescents and Young Adults (AYAs). CURRENT GENETIC MEDICINE REPORTS 2020. [DOI: 10.1007/s40142-020-00187-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
65
|
Chen L, Fu F, Huang M, Lv J, Zhang W, Wang C. The spectrum of BRCA1 and BRCA2 mutations and clinicopathological characteristics in Chinese women with early-onset breast cancer. Breast Cancer Res Treat 2020; 180:759-766. [PMID: 32072338 DOI: 10.1007/s10549-020-05573-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 02/07/2020] [Indexed: 12/19/2022]
Abstract
PURPOSE BRCA1/2 mutations represent a high risk of breast cancer and are related to early-onset breast cancer. However, few studies have reported the relationship between BRCA1/2 mutations and their clinical characteristics in early-onset breast cancers. This study is the first article that characterizes the risk factor profiles in Chinese patients selected by the age of onset (≤ 40 years old). We found some differences in the prevalence of germline BRCA1/2 mutations between Asian and Western countries. METHODS A total of 1371 consecutive unselected Chinese early-onset breast cancer patients were enrolled from the Fujian Medical University Union Hospital, China, and screened for germline BRCA1/2 mutations. Full-exome sequencing in next-generation sequencing technology was performed in all patients to examine BRCA1/2 mutations. RESULTS In our study, 25 (1.8%) and 61 (4.4%) patients were identified with BRCA1 and BRCA2 mutations, respectively, among the unselected early-onset breast cancer patients. BRCA1 mutations were associated with pregnancies (p = 0.026), and BRCA1 carriers had a higher likelihood of being HR positive (p < 0.001), HER2 negative (p < 0.001), or high grade (p = 0.002) than noncarriers. Among BRCA2 mutations, the age of onset was younger in carriers than in noncarriers (p = 0.017), and BRCA2 carriers were more likely to have lymph node metastasis (p = 0.004). HR-positive or HER2-negative patients were likely to be positive for BRCA2 mutations (p < 0.001). Overall, 14 BRCA1 mutations and 8 BRCA2 mutations were first reported in our study CONCLUSION: This study provided some information about the spectrum of BRCA1/2 mutations and characterized the risk factors for early-onset breast cancer in China.
Collapse
Affiliation(s)
- Lili Chen
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Fangmeng Fu
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Meng Huang
- Fujian Center for Disease Control and Prevention, Fuzhou, China
| | - Jinxing Lv
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Wenzhe Zhang
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Chuan Wang
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China.
| |
Collapse
|
66
|
Terkelsen T, Christensen LL, Fenton DC, Jensen UB, Sunde L, Thomassen M, Skytte AB. Population frequencies of pathogenic alleles of BRCA1 and BRCA2: analysis of 173 Danish breast cancer pedigrees using the BOADICEA model. Fam Cancer 2020; 18:381-388. [PMID: 31435815 DOI: 10.1007/s10689-019-00141-9] [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] [Indexed: 01/11/2023]
Abstract
The Breast and Ovarian Analysis of Disease Incidence and Carrier Estimation Algorithm (BOADICEA) calculates the probability that a woman carries a pathogenic variant in BRCA1 or BRCA2 based on her pedigree and the population frequencies of pathogenic alleles of BRCA1 (0.0006394) and BRCA2 (0.00102) in the United Kingdom (UK). BOADICEA allows the clinician to define the population frequencies of pathogenic alleles of BRCA1 and BRCA2 for other populations but only includes preset values for the Ashkenazy Jewish and Icelandic populations. Among 173 early-onset breast cancer pedigrees in Denmark, BOADICEA discriminated well between carriers and non-carriers of pathogenic variants (area under the receiver operating characteristics curve: 0.81; 95% CI 0.74-0.86) but underestimated the frequency of carriers of pathogenic variants in BRCA1 or BRCA2 as measured by the observed-to-expected ratio (O/E 1.83; 95% CI 1.18-2.84). This reflects findings from older studies of BOADICEA in UK, German, Italian, and Chinese populations, all accounting for the different calibration for different carrier probabilities. To improve the performance of BOADICEA for non-UK populations, we developed a method to derive population frequencies of pathogenic alleles of BRCA1 and BRCA2. Compared to the UK population frequencies, we estimated the Danish population frequencies of pathogenic alleles to be higher for BRCA1 (0.0015; 95% CI 0.00064-0.0034) and lower for BRCA2 (0.00052; 95% CI 0.00018-0.0017) after adjusting for the different calibration of BOADICEA for different carrier probabilities. Incorporating additional population frequencies into BOADICEA could improve its performance for non-UK populations.
Collapse
Affiliation(s)
- Thorkild Terkelsen
- Department of Clinical Genetics, Aarhus University Hospital, Brendstrupgaardsvej 21C, 8200, Aarhus N, Denmark.
| | | | | | - Uffe Birk Jensen
- Department of Clinical Genetics, Aarhus University Hospital, Brendstrupgaardsvej 21C, 8200, Aarhus N, Denmark
| | - Lone Sunde
- Department of Clinical Genetics, Aarhus University Hospital, Brendstrupgaardsvej 21C, 8200, Aarhus N, Denmark
| | - Mads Thomassen
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
| | - Anne-Bine Skytte
- Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
67
|
Abstract
Breast cancer is the most common malignancy in Jordan and the third leading cause of cancer death after lung and colorectal cancers. Although the incidence of breast cancer in Jordan is lower than that in industrialized nations, the number of new cases has been significantly increasing, and women present with breast cancer at a younger age and with more advanced disease than women in Western countries. Jordan is a medium-income country with limited resources and a young population structure. Therefore, breast cancer poses a particularly challenging burden on the country's health care system. Despite ongoing endeavors to improve breast cancer care at both public and private levels, more work is needed to achieve downstaging of the disease and improve access, awareness, and participation in early detection. Multimodality treatment facilities and supportive care are available; however, the quality of care varies widely according to where the patient is treated, and most treatment facilities remain located centrally, thus, creating access difficulties. The King Hussein Cancer Center, the only comprehensive cancer center in Jordan, has changed the practice of oncology in the country via implementation of a multidisciplinary approach to treatment, monitoring of treatment outcomes, and investments in ongoing cancer research. However, there remains no national system for ensuring provision of high-quality cancer care nationwide. Here, we review the epidemiology of breast cancer and the current status of breast cancer care in Jordan, we compare our treatment outcomes with international ones, and we highlight challenges and improvement opportunities.
Collapse
Affiliation(s)
- Hikmat Abdel-Razeq
- Department of Internal Medicine, Section of Hematology and Medical Oncology, King Hussein Cancer Center, Amman, Jordan
- School of Medicine, University of Jordan, Amman, Jordan
| | - Asem Mansour
- Department of Radiology, King Hussein Cancer Center, Amman, Jordan
| | - Dima Jaddan
- Department of Internal Medicine, Section of Hematology and Medical Oncology, King Hussein Cancer Center, Amman, Jordan
| |
Collapse
|
68
|
Adedokun B, Zheng Y, Ndom P, Gakwaya A, Makumbi T, Zhou AY, Yoshimatsu TF, Rodriguez A, Madduri RK, Foster IT, Sallam A, Olopade OI, Huo D. Prevalence of Inherited Mutations in Breast Cancer Predisposition Genes among Women in Uganda and Cameroon. Cancer Epidemiol Biomarkers Prev 2019; 29:359-367. [PMID: 31871109 DOI: 10.1158/1055-9965.epi-19-0506] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 07/23/2019] [Accepted: 12/09/2019] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Sub-Saharan Africa (SSA) has a high proportion of premenopausal hormone receptor negative breast cancer. Previous studies reported a strikingly high prevalence of germline mutations in BRCA1 and BRCA2 among Nigerian patients with breast cancer. It is unknown if this exists in other SSA countries. METHODS Breast cancer cases, unselected for age at diagnosis and family history, were recruited from tertiary hospitals in Kampala, Uganda and Yaoundé, Cameroon. Controls were women without breast cancer recruited from the same hospitals and age-matched to cases. A multigene sequencing panel was used to test for germline mutations. RESULTS There were 196 cases and 185 controls with a mean age of 46.2 and 46.6 years for cases and controls, respectively. Among cases, 15.8% carried a pathogenic or likely pathogenic mutation in a breast cancer susceptibility gene: 5.6% in BRCA1, 5.6% in BRCA2, 1.5% in ATM, 1% in PALB2, 0.5% in BARD1, 0.5% in CDH1, and 0.5% in TP53. Among controls, 1.6% carried a mutation in one of these genes. Cases were 11-fold more likely to carry a mutation compared with controls (OR = 11.34; 95% confidence interval, 3.44-59.06; P < 0.001). The mean age of cases with BRCA1 mutations was 38.3 years compared with 46.7 years among other cases without such mutations (P = 0.03). CONCLUSIONS Our findings replicate the earlier report of a high proportion of mutations in BRCA1/2 among patients with symptomatic breast cancer in SSA. IMPACT Given the high burden of inherited breast cancer in SSA countries, genetic risk assessment could be integrated into national cancer control plans.
Collapse
Affiliation(s)
- Babatunde Adedokun
- Center for Clinical Cancer Genetics and Global Health, Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Yonglan Zheng
- Center for Clinical Cancer Genetics and Global Health, Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Paul Ndom
- Hôpital Général Yaoundé, Yaoundé, Cameroon
| | | | | | | | - Toshio F Yoshimatsu
- Center for Clinical Cancer Genetics and Global Health, Department of Medicine, The University of Chicago, Chicago, Illinois
| | | | - Ravi K Madduri
- Globus, The University of Chicago, Chicago, Illinois.,Data Science and Learning Division, Argonne National Laboratory, Lemont, Illinois
| | - Ian T Foster
- Globus, The University of Chicago, Chicago, Illinois.,Data Science and Learning Division, Argonne National Laboratory, Lemont, Illinois
| | - Aminah Sallam
- Center for Clinical Cancer Genetics and Global Health, Department of Medicine, The University of Chicago, Chicago, Illinois.,Yale School of Medicine, New Haven, Connecticut
| | - Olufunmilayo I Olopade
- Center for Clinical Cancer Genetics and Global Health, Department of Medicine, The University of Chicago, Chicago, Illinois.
| | - Dezheng Huo
- Center for Clinical Cancer Genetics and Global Health, Department of Medicine, The University of Chicago, Chicago, Illinois. .,Department of Public Health Sciences, The University of Chicago, Chicago, Illinois
| |
Collapse
|
69
|
Hsu CY, Hsu WF, Yen AMF, Chen HH. Sampling-based Markov regression model for multistate disease progression: Applications to population-based cancer screening program. Stat Methods Med Res 2019; 29:2198-2216. [PMID: 31744392 DOI: 10.1177/0962280219885400] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
To develop personalized screening and surveillance strategies, the information required to superimpose state-specific covariates into the multi-step progression of disease natural history often relies on the entire population-based screening data, which are costly and infeasible particularly when a new biomarker is proposed. Following Prentice's case-cohort concept, a non-standard case-cohort design from a previous study has been adapted for constructing multistate disease natural history with two-stage sampling. Nonetheless, the use of data only from first screens may invoke length-bias and fail to consider the test sensitivity. Therefore, a new sampling-based Markov regression model and its variants are proposed to accommodate additional subsequent follow-up data on various detection modes to construct state-specific covariate-based multistate disease natural history with accuracy and efficiency. Computer simulation algorithms for determining the required sample size and the sampling fraction of each detection mode were developed either through power function or the capacity of screening program. The former is illustrated with breast cancer screening data from which the effect size and the required sample size regarding the effect of BRCA on multistate outcome of breast cancer were estimated. The latter is applied to population-based colorectal cancer screening data to identify the optimal sampling fraction of each detection mode.
Collapse
Affiliation(s)
- Chen-Yang Hsu
- Graduate Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei.,School of Nursing, National Taipei University of Nursing and Health Sciences, Taipei
| | - Wen-Feng Hsu
- Graduate Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei.,Department of Internal Medicine, National Taiwan University Hospital, Taipei
| | - Amy Ming-Fang Yen
- Department of Internal Medicine, National Taiwan University Hospital, Taipei
| | - Hsiu-Hsi Chen
- Graduate Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei.,Innovation and Policy Center for Population Health and Sustainable Environment, College of Public Health, National Taiwan University, Taipei
| |
Collapse
|
70
|
Abstract
Ageing appears to be a nearly universal feature of life, ranging from unicellular microorganisms to humans. Longevity depends on the maintenance of cellular functionality, and an organism's ability to respond to stress has been linked to functional maintenance and longevity. Stress response pathways might indeed become therapeutic targets of therapies aimed at extending the healthy lifespan. Various progeroid syndromes have been linked to genome instability, indicating an important causal role of DNA damage accumulation in the ageing process and the development of age-related pathologies. Recently, non-cell-autonomous mechanisms including the systemic consequences of cellular senescence have been implicated in regulating organismal ageing. We discuss here the role of cellular and systemic mechanisms of ageing and their role in ageing-associated diseases.
Collapse
Affiliation(s)
- Paulo F L da Silva
- Institute for Genome Stability in Ageing and Disease, Medical Faculty, University of Cologne, Joseph-Stelzmann-Strasse 26, 50931 Cologne, Germany.,Cologne Excellence Cluster for Cellular Stress Responses in Ageing-Associated Diseases (CECAD), Center for Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Strasse 26, 50931 Cologne, Germany
| | - Björn Schumacher
- Institute for Genome Stability in Ageing and Disease, Medical Faculty, University of Cologne, Joseph-Stelzmann-Strasse 26, 50931 Cologne, Germany.,Cologne Excellence Cluster for Cellular Stress Responses in Ageing-Associated Diseases (CECAD), Center for Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Strasse 26, 50931 Cologne, Germany
| |
Collapse
|
71
|
Schrauder MG, Brunel-Geuder L, Häberle L, Wunderle M, Hoyer J, Csorba R, Reis A, Schulz-Wendtland R, Beckmann MW, Lux MP. Cost effectiveness of bilateral risk-reducing mastectomy and salpingo-oophorectomy. Eur J Med Res 2019; 24:32. [PMID: 31521205 PMCID: PMC6744699 DOI: 10.1186/s40001-019-0391-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Accepted: 09/03/2019] [Indexed: 11/23/2022] Open
Abstract
Background Growing demand for risk-reducing surgery in individuals with inherited susceptibility to cancer leads to the question whether these procedures are cost effective for the executing hospitals. This study compared the clinical costs for bilateral risk-reducing mastectomy (BRRM) with and without different types of reconstruction, risk-reducing salpingo-oophorectomy (RRSO), and their combinations with corresponding reimbursements in the statutory health-care system in Germany. Patients and methods Real total costs of care for BRRM with and without reconstruction, RRSO, and their combinations were calculated as the sum of all personnel and technical costs. These costs calculated in a German University hospital were compared with the sum of all reimbursements in the German DRG-based health-care system. Results While sole RRSO, BRRM without reconstruction, and BRRM with secondary DIEP (deep inferior epigastric perforator)—reconstruction still result in a small benefit, we even found shortfalls for the hospital with all other prophylactic operations under consideration. The calculated deficits were especially high for BRRM with implant-based breast reconstruction and for combined operations when the risk reduction is achieved with a minimum of separate operations. Conclusions Risk-reducing surgery in BRCA-mutation carriers is frequently not cost-covering for the executing hospitals in the German health-care system. Thus, appropriate concepts are required to ensure a nationwide care.
Collapse
Affiliation(s)
- Michael G Schrauder
- Department of Gynecology and Obstetrics, Erlangen University Hospital, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Universitätsstraße 21-23, 91054, Erlangen, Germany. .,Department of Obstetrics and Gynecology, Hospital of Aschaffenburg-Alzenau, Aschaffenburg, Germany.
| | - Lisa Brunel-Geuder
- Department of Gynecology and Obstetrics, Erlangen University Hospital, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Universitätsstraße 21-23, 91054, Erlangen, Germany
| | - Lothar Häberle
- Department of Gynecology and Obstetrics, Erlangen University Hospital, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Universitätsstraße 21-23, 91054, Erlangen, Germany
| | - Marius Wunderle
- Department of Gynecology and Obstetrics, Erlangen University Hospital, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Universitätsstraße 21-23, 91054, Erlangen, Germany
| | - Juliane Hoyer
- Institute of Human Genetics, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Roland Csorba
- Department of Obstetrics and Gynecology, Hospital of Aschaffenburg-Alzenau, Aschaffenburg, Germany.,Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - André Reis
- Institute of Human Genetics, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Rüdiger Schulz-Wendtland
- Institute of Radiology, Erlangen University Hospital, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Matthias W Beckmann
- Department of Gynecology and Obstetrics, Erlangen University Hospital, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Universitätsstraße 21-23, 91054, Erlangen, Germany
| | - Michael P Lux
- Department of Gynecology and Obstetrics, Erlangen University Hospital, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Universitätsstraße 21-23, 91054, Erlangen, Germany
| |
Collapse
|
72
|
Owens DK, Davidson KW, Krist AH, Barry MJ, Cabana M, Caughey AB, Doubeni CA, Epling JW, Kubik M, Landefeld CS, Mangione CM, Pbert L, Silverstein M, Simon MA, Tseng CW, Wong JB. Risk Assessment, Genetic Counseling, and Genetic Testing for BRCA-Related Cancer: US Preventive Services Task Force Recommendation Statement. JAMA 2019; 322:652-665. [PMID: 31429903 DOI: 10.1001/jama.2019.10987] [Citation(s) in RCA: 322] [Impact Index Per Article: 53.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
IMPORTANCE Potentially harmful mutations of the breast cancer susceptibility 1 and 2 genes (BRCA1/2) are associated with increased risk for breast, ovarian, fallopian tube, and peritoneal cancer. For women in the United States, breast cancer is the most common cancer after nonmelanoma skin cancer and the second leading cause of cancer death. In the general population, BRCA1/2 mutations occur in an estimated 1 in 300 to 500 women and account for 5% to 10% of breast cancer cases and 15% of ovarian cancer cases. OBJECTIVE To update the 2013 US Preventive Services Task Force (USPSTF) recommendation on risk assessment, genetic counseling, and genetic testing for BRCA-related cancer. EVIDENCE REVIEW The USPSTF reviewed the evidence on risk assessment, genetic counseling, and genetic testing for potentially harmful BRCA1/2 mutations in asymptomatic women who have never been diagnosed with BRCA-related cancer, as well as those with a previous diagnosis of breast, ovarian, tubal, or peritoneal cancer who have completed treatment and are considered cancer free. In addition, the USPSTF reviewed interventions to reduce the risk for breast, ovarian, tubal, or peritoneal cancer in women with potentially harmful BRCA1/2 mutations, including intensive cancer screening, medications, and risk-reducing surgery. FINDINGS For women whose family or personal history is associated with an increased risk for harmful mutations in the BRCA1/2 genes, or who have an ancestry associated with BRCA1/2 gene mutations, there is adequate evidence that the benefits of risk assessment, genetic counseling, genetic testing, and interventions are moderate. For women whose personal or family history or ancestry is not associated with an increased risk for harmful mutations in the BRCA1/2 genes, there is adequate evidence that the benefits of risk assessment, genetic counseling, genetic testing, and interventions are small to none. Regardless of family or personal history, the USPSTF found adequate evidence that the overall harms of risk assessment, genetic counseling, genetic testing, and interventions are small to moderate. CONCLUSIONS AND RECOMMENDATION The USPSTF recommends that primary care clinicians assess women with a personal or family history of breast, ovarian, tubal, or peritoneal cancer or who have an ancestry associated with BRCA1/2 gene mutations with an appropriate brief familial risk assessment tool. Women with a positive result on the risk assessment tool should receive genetic counseling and, if indicated after counseling, genetic testing. (B recommendation) The USPSTF recommends against routine risk assessment, genetic counseling, or genetic testing for women whose personal or family history or ancestry is not associated with potentially harmful BRCA1/2 gene mutations. (D recommendation).
Collapse
Affiliation(s)
| | - Douglas K Owens
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California
- Stanford University, Stanford, California
| | - Karina W Davidson
- Feinstein Institute for Medical Research at Northwell Health, Manhasset, New York
| | - Alex H Krist
- Fairfax Family Practice Residency, Fairfax, Virginia
- Virginia Commonwealth University, Richmond
| | | | | | | | | | | | | | | | | | - Lori Pbert
- University of Massachusetts Medical School, Worcester
| | | | | | - Chien-Wen Tseng
- University of Hawaii, Honolulu
- Pacific Health Research and Education Institute, Honolulu, Hawaii
| | - John B Wong
- Tufts University School of Medicine, Boston, Massachusetts
| |
Collapse
|
73
|
Nelson HD, Pappas M, Cantor A, Haney E, Holmes R. Risk Assessment, Genetic Counseling, and Genetic Testing for BRCA-Related Cancer in Women: Updated Evidence Report and Systematic Review for the US Preventive Services Task Force. JAMA 2019; 322:666-685. [PMID: 31429902 DOI: 10.1001/jama.2019.8430] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
IMPORTANCE Pathogenic mutations in breast cancer susceptibility genes BRCA1 and BRCA2 increase risks for breast, ovarian, fallopian tube, and peritoneal cancer in women; interventions reduce risk in mutation carriers. OBJECTIVE To update the 2013 US Preventive Services Task Force review on benefits and harms of risk assessment, genetic counseling, and genetic testing for BRCA1/2-related cancer in women. DATA SOURCES Cochrane libraries; MEDLINE, PsycINFO, EMBASE (January 1, 2013, to March 6, 2019, for updates; January 1, 1994, to March 6, 2019, for new key questions and populations); reference lists. STUDY SELECTION Discriminatory accuracy studies, randomized clinical trials (RCTs), and observational studies of women without recently diagnosed BRCA1/2-related cancer. DATA EXTRACTION AND SYNTHESIS Data on study methods, setting, population characteristics, eligibility criteria, interventions, numbers enrolled and lost to follow-up, outcome ascertainment, and results were abstracted. Two reviewers independently assessed study quality. MAIN OUTCOMES AND MEASURES Cancer incidence and mortality; discriminatory accuracy of risk assessment tools for BRCA1/2 mutations; benefits and harms of risk assessment, genetic counseling, genetic testing, and risk-reducing interventions. RESULTS For this review, 103 studies (110 articles; N = 92 712) were included. No studies evaluated the effectiveness of risk assessment, genetic counseling, and genetic testing in reducing incidence and mortality of BRCA1/2-related cancer. Fourteen studies (n = 43 813) of 8 risk assessment tools to guide referrals to genetic counseling demonstrated moderate to high accuracy (area under the receiver operating characteristic curve, 0.68-0.96). Twenty-eight studies (n = 8060) indicated that genetic counseling was associated with reduced breast cancer worry, anxiety, and depression; increased understanding of risk; and decreased intention for testing. Twenty studies (n = 4322) showed that breast cancer worry and anxiety were higher after testing for women with positive results and lower for others; understanding of risk was higher after testing. In 8 RCTs (n = 54 651), tamoxifen (relative risk [RR], 0.69 [95% CI, 0.59-0.84]; 4 trials), raloxifene (RR, 0.44 [95% CI, 0.24-0.80]; 2 trials), and aromatase inhibitors (RR, 0.45 [95% CI, 0.26-0.70]; 2 trials) were associated with lower risks of invasive breast cancer compared with placebo; results were not specific to mutation carriers. Mastectomy was associated with 90% to 100% reduction in breast cancer incidence (6 studies; n = 2546) and 81% to 100% reduction in breast cancer mortality (1 study; n = 639); oophorectomy was associated with 69% to 100% reduction in ovarian cancer (2 studies; n = 2108); complications were common with mastectomy. CONCLUSIONS AND RELEVANCE Among women without recently diagnosed BRCA1/2-related cancer, the benefits and harms of risk assessment, genetic counseling, and genetic testing to reduce cancer incidence and mortality have not been directly evaluated by current research.
Collapse
Affiliation(s)
- Heidi D Nelson
- Pacific Northwest Evidence-based Practice Center, Oregon Health & Science University, Portland
| | - Miranda Pappas
- Pacific Northwest Evidence-based Practice Center, Oregon Health & Science University, Portland
| | - Amy Cantor
- Pacific Northwest Evidence-based Practice Center, Oregon Health & Science University, Portland
| | - Elizabeth Haney
- Pacific Northwest Evidence-based Practice Center, Oregon Health & Science University, Portland
| | - Rebecca Holmes
- Pacific Northwest Evidence-based Practice Center, Oregon Health & Science University, Portland
| |
Collapse
|
74
|
Polansky H, Schwab H. How latent viruses cause breast cancer: An explanation based on the microcompetition model. Bosn J Basic Med Sci 2019; 19:221-226. [PMID: 30579323 DOI: 10.17305/bjbms.2018.3950] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 12/11/2018] [Indexed: 12/23/2022] Open
Abstract
Most breast cancer cases show a decrease in the concentration of the breast cancer type 1 susceptibility protein (BRCA1). However, only a small portion of these cases have a mutated BRCA1 gene. Although many attempts have been made to identify the reason for the decrease in BRCA1 concentration in sporadic, non-heritable breast cancer cases, the cause is still unknown. In this review, we use the Microcompetition Model to explain how certain latent viruses, which are frequently detected in breast cancer tumors, can decrease the expression of the BRCA1 gene and cause the development of breast tumors.
Collapse
Affiliation(s)
- Hanan Polansky
- The Center for the Biology of Chronic Disease (CBCD), New York, NY, USA.
| | | |
Collapse
|
75
|
Two-Staged Implant-Based Breast Reconstruction: A Long-Term Outcome Study in a Young Population. ACTA ACUST UNITED AC 2019; 55:medicina55080481. [PMID: 31416221 PMCID: PMC6723805 DOI: 10.3390/medicina55080481] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 07/18/2019] [Accepted: 08/12/2019] [Indexed: 11/17/2022]
Abstract
Background and objectives: Differences in patient anatomy and physiology exist between young and older patients undergoing breast reconstruction after mastectomy. Breast cancer has been described as being more aggressive, more likely to receive radiation, contralateral mastectomy, as well as bilateral reconstruction in young patients. Our purpose is to report long-term experience on two-staged implant-based breast reconstruction (IBR) in young females, with complication sub-analysis based on obesity and adjuvant radiation. Materials and Methods: Retrospective chart review of all consecutive young patients who underwent two-staged IBR at our institution, between 2000 and 2016, was performed. Patients between 15 and 40 years old with least 1-year follow-up were included. Univariate logistic regression models and receiver operating characteristic (ROC) curves were created. Results: Overall 594 breasts met our inclusion criteria. The mean age was 34 years, and the median follow-up was 29.6 months. Final IBR was achieved in 98% of breasts. Overall, 12% of breasts had complications, leading to explantations of 5% of the devices. Adjuvant radiation was followed by higher rates of total device explantations (p = 0.003), while obese patients had higher rates of total complications (p < 0.001). For each point increase in BMI, the odds of developing complications increased 8.1% (p < 0.001); the cutoff BMI to predict higher complications was 24.81 kg/m2. Conclusions: This population demonstrates high successful IBR completion and low explantation rates. These data suggest that obese women and those with planned adjuvant radiation deserve special counseling about their higher risk of complications.
Collapse
|
76
|
Krassuski L, Vennedey V, Stock S, Kautz-Freimuth S. Effectiveness of decision aids for female BRCA1 and BRCA2 mutation carriers: a systematic review. BMC Med Inform Decis Mak 2019; 19:154. [PMID: 31370837 PMCID: PMC6670224 DOI: 10.1186/s12911-019-0872-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 07/09/2019] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Female BRCA1 and BRCA2 mutation carriers have an increased lifetime risk of developing breast and/or ovarian cancer. Hence, they face the difficult decision of choosing a preventive strategy such as risk-reducing surgeries or intensified breast screening. To help these women during their decision process, several patient decision aids (DA) were developed and evaluated in the last 15 years. Until now, there is no conclusive evidence on the effectiveness of these DA. This study aims 1) to provide the first systematic literature review about DA addressing preventive strategy decisions for female BRCA1 and BRCA2 mutation carriers, 2) to analyze the quality of the existing evidence, 3) to evaluate the effects of DA on decision and information related outcomes, on the actual choice for preventive measure and on health outcomes. METHODS A systematic literature review was conducted using six electronic databases (inclusion criteria: DA addressing preventive strategies, female BRCA1 and BRCA2 mutation carriers, 18 to 75 years, knowledge of test result). The quality of the included randomized controlled trials (RCT) was evaluated with the Cochrane Collaboration's risk of bias tool. The quality of included one-group pretest-posttest design studies was evaluated with the ROBINS-I tool. Outcomes of included studies were extracted and qualitatively summarized. RESULTS A total of 2093 records were identified. Six studies were included for further evaluation (5 RCT, 1 one-group pretest-posttest design study). One RCT was formally included, but data presentation did not allow for further analyses. The risk of bias was high in three RCT and unclear in one RCT. The risk of bias in the one-group pretest-posttest study was serious. The outcome assessment showed that the main advantages of DA are linked to the actual decision process: Female BRCA1 and BRCA2 mutation carriers using a DA had less decisional conflict, were more likely to reach a decision and were more satisfied with their decision. CONCLUSIONS Decision aids can support female BRCA1 and BRCA2 mutation carriers during their decision process by significantly improving decision related outcomes. More high-quality evidence is needed to evaluate possible effects on information related outcomes, health outcomes and the actual choice for preventive measures.
Collapse
Affiliation(s)
- Lisa Krassuski
- Institute for Health Economics and Clinical Epidemiology, The University Hospital of Cologne (AöR), Gleueler Straße 176-178, 50935, Cologne, Germany.
| | - Vera Vennedey
- Institute for Health Economics and Clinical Epidemiology, The University Hospital of Cologne (AöR), Gleueler Straße 176-178, 50935, Cologne, Germany
| | - Stephanie Stock
- Institute for Health Economics and Clinical Epidemiology, The University Hospital of Cologne (AöR), Gleueler Straße 176-178, 50935, Cologne, Germany
| | - Sibylle Kautz-Freimuth
- Institute for Health Economics and Clinical Epidemiology, The University Hospital of Cologne (AöR), Gleueler Straße 176-178, 50935, Cologne, Germany.
| |
Collapse
|
77
|
Sa-Nguanraksa D, Sasanakietkul T, O-Charoenrat C, Kulprom A, O-Charoenrat P. Gail Model Underestimates Breast Cancer Risk in Thai Population. Asian Pac J Cancer Prev 2019; 20:2385-2389. [PMID: 31450910 PMCID: PMC6852814 DOI: 10.31557/apjcp.2019.20.8.2385] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Indexed: 11/25/2022] Open
Abstract
Background: The Gail model is the most widely used method for breast cancer risk estimation. This model has
been studied and verified for its validity in many groups but there has yet to be a study to validate the Gail model in a
Thai population. This study aims to evaluate whether the Gail model can accurately calculate the risk of breast cancer
among Thai women. Methods: The subjects were recruited from the Division of Head, Neck, and Breast Surgery,
Department of Surgery, Siriraj Hospital. The patients attending the division were asked to enroll in the study and
complete questionnaires. Gail model scores were then calculated. Relationships between parameters were examined
using the Pearson’s chi-square test, Fisher’s exact test, and independent-samples t-test. Results: There were 514
women recruited. Age, parity, age at first-live birth, and history of atypical ductal hyperplasia (ADH) were significant
risk factors for breast cancer. The 5-year and lifetime risk score for breast cancer calculated by the Gail model were
not significantly different between the patient and the control subjects. The proportions of the subjects with lifetime
risk ≥20% were significantly higher in breast cancer patients (p=0.049). Conclusion: The Gail model underestimated
the risk of breast cancer in Thai women. Calibration of the model is still required before adoption in Thai population.
Collapse
Affiliation(s)
- Doonyapat Sa-Nguanraksa
- Division of Head Neck and Breast Surgery, Department of Surgery, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand.
| | - Thanyawat Sasanakietkul
- Division of Head Neck and Breast Surgery, Department of Surgery, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand. ,Minimally Invasive and Endocrine Surgery Division, Department of Surgery, Police General Hospital, Bangkok, Thailand
| | - Chayanuch O-Charoenrat
- Division of Head Neck and Breast Surgery, Department of Surgery, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand.
| | - Anchalee Kulprom
- Division of Head Neck and Breast Surgery, Department of Surgery, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand.
| | - Pornchai O-Charoenrat
- Division of Head Neck and Breast Surgery, Department of Surgery, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
78
|
Hamdan D, Nguyen TT, Leboeuf C, Meles S, Janin A, Bousquet G. Genomics applied to the treatment of breast cancer. Oncotarget 2019; 10:4786-4801. [PMID: 31413819 PMCID: PMC6677666 DOI: 10.18632/oncotarget.27102] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 07/05/2019] [Indexed: 12/20/2022] Open
Abstract
Breast cancer remains a major health issue in the world with 1.7 million new cases in 2012 worldwide. It is the second cause of death from cancer in western countries. Genomics have started to modify the treatment of breast cancer, and the developments should become more and more significant, especially in the present era of treatment personalization and with the implementation of new technologies. With molecular signatures, genomics enabled a de-escalation of chemotherapy and personalized treatments of localized forms of estrogen-dependent breast cancers. Genomics can also make a real contribution to constitutional genetics, so as to identify mutations in a panel of candidate genes. In this review, we will discuss the contributions of genomics applied to the treatment of breast cancer, whether already validated contributions or possible future applications linked to research data.
Collapse
Affiliation(s)
- Diaddin Hamdan
- Hôpital La Porte Verte, Versailles F-78004, France.,U942, Université Paris-Diderot, INSERM, Paris F-75010, France
| | - Thi Thuy Nguyen
- U942, Université Paris-Diderot, INSERM, Paris F-75010, France.,National Cancer Hospital, Medical Oncology Department 2, Ha Noi 110000, Viet Nam.,Ha Noi Medical University, Oncology Department, Ha Noi 116001, Viet Nam
| | - Christophe Leboeuf
- U942, Université Paris-Diderot, INSERM, Paris F-75010, France.,AP-HP-Hôpital Saint-Louis, Laboratoire de Pathologie, Paris F-75010, France
| | - Solveig Meles
- U942, Université Paris-Diderot, INSERM, Paris F-75010, France
| | - Anne Janin
- U942, Université Paris-Diderot, INSERM, Paris F-75010, France.,AP-HP-Hôpital Saint-Louis, Laboratoire de Pathologie, Paris F-75010, France
| | - Guilhem Bousquet
- U942, Université Paris-Diderot, INSERM, Paris F-75010, France.,AP-HP-Hôpital Avicenne, Service d'Oncologie Médicale, Bobigny F-93000, France.,Université Paris 13, Leonard de Vinci, Villetaneuse F-93430, France
| |
Collapse
|
79
|
Reisländer T, Lombardi EP, Groelly FJ, Miar A, Porru M, Di Vito S, Wright B, Lockstone H, Biroccio A, Harris A, Londoño-Vallejo A, Tarsounas M. BRCA2 abrogation triggers innate immune responses potentiated by treatment with PARP inhibitors. Nat Commun 2019; 10:3143. [PMID: 31316060 PMCID: PMC6637138 DOI: 10.1038/s41467-019-11048-5] [Citation(s) in RCA: 137] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 06/18/2019] [Indexed: 01/21/2023] Open
Abstract
Heterozygous germline mutations in BRCA2 predispose to breast and ovarian cancer. Contrary to non-cancerous cells, where BRCA2 deletion causes cell cycle arrest or cell death, tumors carrying BRCA2 inactivation continue to proliferate. Here we set out to investigate adaptation to loss of BRCA2 focusing on genome-wide transcriptome alterations. Human cells in which BRCA2 expression is inhibited for 4 or 28 days are subjected to RNA-seq analyses revealing a biphasic response to BRCA2 abrogation. The early, acute response consists of downregulation of genes involved in cell cycle progression, DNA replication and repair and is associated with cell cycle arrest in G1. Surprisingly, the late, chronic response consists predominantly of upregulation of interferon-stimulated genes (ISGs). Activation of the cGAS-STING-STAT pathway detected in these cells further substantiates the concept that BRCA2 abrogation triggers cell-intrinsic immune signaling. Importantly, we find that treatment with PARP inhibitors stimulates the interferon response in cells and tumors lacking BRCA2.
Collapse
Affiliation(s)
- Timo Reisländer
- Genome Stability and Tumourigenesis Group, The CR-UK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Emilia Puig Lombardi
- Institut Curie, PSL Research University, CNRS, UMR3244, F-75005, Paris, France
- Sorbonne Universités, UPMC Univ Paris 06, CNRS, UMR3244, F-75005, Paris, France
| | - Florian J Groelly
- Genome Stability and Tumourigenesis Group, The CR-UK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Ana Miar
- Hypoxia and Angiogenesis Group, Weatherall Institute of Molecular Medicine, Department of Oncology, University of Oxford, Oxford, OX3 9DS, UK
| | - Manuela Porru
- Area of Translational Research, IRCCS Regina Elena National Cancer Institute, 00144, Rome, Italy
| | - Serena Di Vito
- Area of Translational Research, IRCCS Regina Elena National Cancer Institute, 00144, Rome, Italy
| | - Benjamin Wright
- Bioinformatics and Statistical Genetics Core, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - Helen Lockstone
- Bioinformatics and Statistical Genetics Core, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - Annamaria Biroccio
- Area of Translational Research, IRCCS Regina Elena National Cancer Institute, 00144, Rome, Italy
| | - Adrian Harris
- Hypoxia and Angiogenesis Group, Weatherall Institute of Molecular Medicine, Department of Oncology, University of Oxford, Oxford, OX3 9DS, UK
| | - Arturo Londoño-Vallejo
- Institut Curie, PSL Research University, CNRS, UMR3244, F-75005, Paris, France
- Sorbonne Universités, UPMC Univ Paris 06, CNRS, UMR3244, F-75005, Paris, France
| | - Madalena Tarsounas
- Genome Stability and Tumourigenesis Group, The CR-UK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK.
| |
Collapse
|
80
|
Catana A, Apostu AP, Antemie RG. Multi gene panel testing for hereditary breast cancer - is it ready to be used? Med Pharm Rep 2019; 92:220-225. [PMID: 31460501 PMCID: PMC6709965 DOI: 10.15386/mpr-1083] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 02/28/2019] [Accepted: 03/27/2019] [Indexed: 02/06/2023] Open
Abstract
Breast cancer is one of the most common malignancies and the leading cause of death among women worldwide. About 20% of breast cancers are hereditary. Approximately 30% of the mutations have remained negative after testing BRCA1/2 even in families with a Mendelian inheritance pattern for breast cancer. Additional non-BRCA genes have been identified as predisposing for breast cancer. Multi gene panel testing tries to cover and explain the BRCA negative inherited breast cancer, improving efficiency, speed and costs of the breast cancer screening. We identified 23 studies reporting results from individuals who have undergone multi gene panel testing for hereditary breast cancer and noticed a prevalence of 1-12% of non-BRCA genes, but also a high level of variants of uncertain significance. A result with a high level of variants of uncertain significance is likely to be more costly than bring benefits, as well as increase the anxiety for patients. Regarding further development of multi gene panel testing, more research is required to establish both the optimal care of patients with cancer (specific treatments like PARP inhibitors) and the management of unaffected individuals (chemoprevention and/or prophylactic surgeries). Early detection in these patients as well as prophylactic measures will significantly increase the chance of survival. Therefore, multi gene panel testing is not yet ready to be used outside clear guidelines. In conclusion, studies on additional cohorts will be needed to better define the real prevalence, penetrance and the variants of these genes, as well as to describe clear evidence-based guidelines for these patients.
Collapse
Affiliation(s)
- Andreea Catana
- Genetics Department, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | | | - Razvan-Geo Antemie
- Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| |
Collapse
|
81
|
Heikkinen SMM, Madanat-Harjuoja LM, Seppä KJM, Rantanen ME, Hirvonen EM, Malila NK, Pitkäniemi JM. Familial aggregation of early-onset cancers. Int J Cancer 2019; 146:1791-1799. [PMID: 31199509 PMCID: PMC7027840 DOI: 10.1002/ijc.32512] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 05/22/2019] [Accepted: 05/28/2019] [Indexed: 02/06/2023]
Abstract
This registry‐linkage study evaluates familial aggregation of cancer among relatives of a population‐based series of early‐onset (≤40 years) cancer patients in Finland. A cohort of 376,762 relatives of early‐onset cancer patients diagnosed between 1970 and 2012 in 40,538 families was identified. Familial aggregation of early‐onset breast, colorectal, brain and other central nervous system (CNS) cancer and melanoma was explored by standardized incidence ratios (SIR), stratified by relatedness. Gender‐, age‐ and period‐specific population cancer incidences were used as reference. Cumulative risks for siblings and offspring of the proband up to age ≤40 years were also estimated. Almost all early‐onset cancers were sporadic (98% or more). Among first‐degree relatives, SIR was largest in colorectal cancer (14, 95% confidence interval 9.72–18), and lowest in melanoma (1.93, 1.05–3.23). Highest relative‐specific SIRs were observed for siblings in families, where also parent had concordant cancer, 90 (43–165) for colorectal cancer and 29 (11–64) for CNS cancer. In spouses, all SIRs were at population level. Cumulative risk of colorectal cancer by age 41 was 0.98% in siblings and 0.10% in population, while in breast cancer the corresponding risks were 2.05% and 0.56%. In conclusion, early‐onset cancers are mainly sporadic. Findings support high familial aggregation in early‐onset colorectal and CNS cancers. Familial aggregation in multiplex families with CNS cancers was mainly attributed to neurofibromatosis and in colorectal cancer to FAP‐ and HNPCC‐syndromes. The pattern of familial aggregation of early‐onset breast cancer could be seen to support very early exposure to environmental factors and/or rare genetic factors. What's new? The tendency for certain cancer types to cluster in families generally is explained by shared environmental exposures or inherited mutations. In particular, early‐onset cancer, diagnosed between ages 0 and 40, is considered indicative of familial factors. Here, investigation of cancer risk among more than 376,760 relatives of probands, or individuals with early‐onset cancer, shows that the likelihood of early‐onset cancer affecting even just one other relative in addition to the proband is exceedingly rare. Nearly all early‐onset cancers in the study population were sporadic. Estimated cumulative risks observed for specific cancers may prove useful in the context of genetic counseling.
Collapse
Affiliation(s)
- Sanna M M Heikkinen
- Finnish Cancer Registry, Institute for Statistical and Epidemiological Cancer Research, Helsinki, Finland
| | - Laura-Maria Madanat-Harjuoja
- Finnish Cancer Registry, Institute for Statistical and Epidemiological Cancer Research, Helsinki, Finland.,Dana Farber Cancer Institute, Boston, MA
| | - Karri J M Seppä
- Finnish Cancer Registry, Institute for Statistical and Epidemiological Cancer Research, Helsinki, Finland
| | - Matti E Rantanen
- Finnish Cancer Registry, Institute for Statistical and Epidemiological Cancer Research, Helsinki, Finland
| | - Elli M Hirvonen
- Finnish Cancer Registry, Institute for Statistical and Epidemiological Cancer Research, Helsinki, Finland
| | - Nea K Malila
- Finnish Cancer Registry, Institute for Statistical and Epidemiological Cancer Research, Helsinki, Finland
| | - Janne M Pitkäniemi
- Finnish Cancer Registry, Institute for Statistical and Epidemiological Cancer Research, Helsinki, Finland.,Department of Public Health, School of Medicine, University of Helsinki, Helsinki, Finland.,Faculty of Social Sciences, University of Tampere, Tampere, Finland
| |
Collapse
|
82
|
Overexpression of Kynurenine 3-Monooxygenase Correlates with Cancer Malignancy and Predicts Poor Prognosis in Canine Mammary Gland Tumors. JOURNAL OF ONCOLOGY 2019; 2019:6201764. [PMID: 31186637 PMCID: PMC6521384 DOI: 10.1155/2019/6201764] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 04/08/2019] [Indexed: 12/01/2022]
Abstract
Tumor biomarkers are developed to indicate tumor status, clinical outcome, or prognosis. Since currently there are no effective biomarkers for canine mammary tumor (CMT), this study intended to verify whether kynurenine 3-monooxygenase (KMO), one of the key enzymes involved in tryptophan catabolism, is competent for predicting prognosis in patients with CMT. By investigating a series of 86 CMT clinical cases, we found that both gene and protein expression of KMO discriminated malignant from benign CMTs and was significantly higher in stage IV and V tumors than in lower-stage CMTs. About 73.7% of malignant CMTs showed strong expression of KMO which correlated with lower overall survival rates in patients. Further, downregulation of KMO activity significantly inhibited cell proliferation of CMT cells. Taken together, the findings indicated that KMO is a potential biomarker for tumor diagnosis, and this might open up new perspectives for clinical applications of CMT.
Collapse
|
83
|
Identifying a BRCA2 c.5722_5723del mutation in a Han-Chinese family with breast cancer. Biosci Rep 2019; 39:BSR20182471. [PMID: 30940775 PMCID: PMC6488854 DOI: 10.1042/bsr20182471] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 03/19/2019] [Accepted: 04/01/2019] [Indexed: 12/17/2022] Open
Abstract
Breast cancer (BC) is the most common female cancer found worldwide. It is responsible for 25% of all cancer patients in females. Hereditary BC accounts for about 5–10% of all BC cases. The breast cancer 1 gene (BRCA1) and the breast cancer 2 gene (BRCA2) are the two most-studied BC susceptibility genes. Genetic testing for disease-causing mutations in BRCA1, BRCA2, and other BC susceptibility genes is strongly recommended for members of families having a BC family history. The present study found a heterozygous c.5722_5723del mutation in the BRCA2 exon 11 of a large Han-Chinese BC family using whole exome sequencing and Sanger sequencing. It may cause DNA double-strand breaks repair dysfunction by disturbing homologous recombination, further resulting in BC. The study findings may help supplement and further improve genetic testing strategies and BC risk estimation methodologies in China.
Collapse
|
84
|
Germline Missense Variants in BRCA1: New Trends and Challenges for Clinical Annotation. Cancers (Basel) 2019; 11:cancers11040522. [PMID: 31013702 PMCID: PMC6520942 DOI: 10.3390/cancers11040522] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 03/13/2019] [Accepted: 03/30/2019] [Indexed: 12/24/2022] Open
Abstract
Genetic testing allows for the identification of germline DNA variations, which are associated with a significant increase in the risk of developing breast cancer (BC) and ovarian cancer (OC). Detection of a BRCA1 or BRCA2 pathogenic variant triggers several clinical management actions, which may include increased surveillance and prophylactic surgery for healthy carriers or treatment with the PARP inhibitor therapy for carriers diagnosed with cancer. Thus, standardized validated criteria for the annotation of BRCA1 and BRCA2 variants according to their pathogenicity are necessary to support clinical decision-making and ensure improved outcomes. Upon detection, variants whose pathogenicity can be inferred by the genetic code are typically classified as pathogenic, likely pathogenic, likely benign, or benign. Variants whose impact on function cannot be directly inferred by the genetic code are labeled as variants of uncertain clinical significance (VUS) and are evaluated by multifactorial likelihood models that use personal and family history of cancer, segregation data, prediction tools, and co-occurrence with a pathogenic BRCA variant. Missense variants, coding alterations that replace a single amino acid residue with another, are a class of variants for which determination of clinical relevance is particularly challenging. Here, we discuss current issues in the missense variant classification by following a typical life cycle of a BRCA1 missense variant through detection, annotation and information dissemination. Advances in massively parallel sequencing have led to a substantial increase in VUS findings. Although the comprehensive assessment and classification of missense variants according to their pathogenicity remains the bottleneck, new developments in functional analysis, high throughput assays, data sharing, and statistical models are rapidly changing this scenario.
Collapse
|
85
|
Zavala VA, Serrano-Gomez SJ, Dutil J, Fejerman L. Genetic Epidemiology of Breast Cancer in Latin America. Genes (Basel) 2019; 10:E153. [PMID: 30781715 PMCID: PMC6410045 DOI: 10.3390/genes10020153] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 02/13/2019] [Accepted: 02/14/2019] [Indexed: 12/20/2022] Open
Abstract
The last 10 years witnessed an acceleration of our understanding of what genetic factors underpin the risk of breast cancer. Rare high- and moderate-penetrance variants such as those in the BRCA genes account for a small proportion of the familial risk of breast cancer. Low-penetrance alleles are expected to underlie the remaining heritability. By now, there are about 180 genetic polymorphisms that are associated with risk, most of them of modest effect. In combination, they can be used to identify women at the lowest or highest ends of the risk spectrum, which might lead to more efficient cancer prevention strategies. Most of these variants were discovered in populations of European descent. As a result, we might be failing to discover additional polymorphisms that could explain risk in other groups. This review highlights breast cancer genetic epidemiology studies conducted in Latin America, and summarizes the information that they provide, with special attention to similarities and differences with studies in other populations. It includes studies of common variants, as well as moderate- and high-penetrance variants. In addition, it addresses the gaps that need to be bridged in order to better understand breast cancer genetic risk in Latin America.
Collapse
Affiliation(s)
- Valentina A Zavala
- Department of Medicine, Division of General Internal Medicine, University of California San Francisco, San Francisco, CA 94143-1793, USA.
| | - Silvia J Serrano-Gomez
- Grupo de investigación en biología del cáncer, Instituto Nacional de Cancerología, Bogotá 11001000, Colombia.
| | - Julie Dutil
- Cancer Biology Division, Ponce Research Institute, Ponce Health Sciences University, Ponce, PR 00732, USA.
| | - Laura Fejerman
- Department of Medicine, Division of General Internal Medicine, University of California San Francisco, San Francisco, CA 94143-1793, USA.
| |
Collapse
|
86
|
Murthy P, Muggia F. Women's cancers: how the discovery of BRCA genes is driving current concepts of cancer biology and therapeutics. Ecancermedicalscience 2019; 13:904. [PMID: 30915162 PMCID: PMC6411414 DOI: 10.3332/ecancer.2019.904] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Indexed: 12/15/2022] Open
Abstract
Over the last two decades, discoveries related to the breast cancer susceptibility genes 1 and 2 (BRCA1 and BRCA2) have profoundly changed our understanding and management of hereditary breast and ovarian cancers. The concept of synthetic lethality, which arises when cells become vulnerable to a combination of deficiencies in DNA repair, has driven the expanding roles of poly (adenosine diphosphate (ADP)-ribose) polymerase inhibitors in breast and ovarian cancers, and prevention strategies are taking into account the tissue specificity, natural history (fallopian tube origin of some high-grade serous ovarian cancers) and hormone sensitivity of BRCA-associated cancers. Current research has focussed on further elucidating the roles of BRCA proteins in DNA repair, investigating other key DNA repair processes and proteins and linking aberrant DNA repair with carcinogenesis. The ultimate goal is to translate this evolving knowledge into improving the clinical care and treatment of patients with pathogenic BRCA variants or other deficiencies in homologous recombination (HR). In this review, we will discuss 1) the role of BRCA proteins in DNA repair; 2) emerging concepts in the biology of HR deficiency and 3) implications for prevention and treatment.
Collapse
Affiliation(s)
- Pooja Murthy
- New York University School of Medicine, New York, NY 10016, USA
- Maimonides Cancer Center, Brooklyn, NY 11220, USA
| | - Franco Muggia
- New York University School of Medicine, New York, NY 10016, USA
| |
Collapse
|
87
|
Wendt C, Margolin S. Identifying breast cancer susceptibility genes - a review of the genetic background in familial breast cancer. Acta Oncol 2019; 58:135-146. [PMID: 30606073 DOI: 10.1080/0284186x.2018.1529428] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Heritage is the most important risk factor for breast cancer. About 15-20% of breast cancer is familial, referring to affected women who have one or more first- or second-degree relatives with the disease. The heritable component in these families is substantial, especially in families with aggregation of breast cancer with low age at onset. Identifying breast cancer susceptibility genes: Since the discovery of the highly penetrant autosomal dominant susceptibility genes BRCA1 and BRCA2 in the 1990s, several more breast cancer genes that confer a moderate to high risk of breast cancer have been identified. Furthermore, during the last decade, advances in genomic technologies have led to large scale genotyping in genome-wide association studies that have identified a considerable amount of common low penetrance loci. In total, the high risk genes, BRCA1, BRCA2, TP53, STK11, CD1 and PTEN account for approximately 20% of the familial risk. Moderate risk variants account for up to 5% of the inherited familial risk. The more than 180 identified low-risk loci explain 18% of the familial risk. Altogether more than half of the genetic background in familial breast cancer remains unclear. Other genes and low risk loci that explain a part the remaining fraction will probably be identified. Clinical aspects and future perspectives: Definitive clinical recommendations can be drawn only for carriers of germline variants in a limited number of high and moderate risk genes for which an association with breast cancer has been established. Future progress in evaluating previously identified breast cancer candidate variants and low risk loci as well as exploring new ones can play an important role in improving individual risk prediction in familial breast cancer.
Collapse
Affiliation(s)
- Camilla Wendt
- Department of Oncology, Södersjukhuset, Stockholm, Sweden
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Sara Margolin
- Department of Oncology, Södersjukhuset, Stockholm, Sweden
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
88
|
Central body fatness is a stronger predictor of cancer risk than overall body size. Nat Commun 2019; 10:383. [PMID: 30670692 PMCID: PMC6342989 DOI: 10.1038/s41467-018-08159-w] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 12/19/2018] [Indexed: 01/06/2023] Open
Abstract
The importance of body size versus weight distribution for cancer risk is unclear. We investigated associations between measures of body size and shape and the risk of developing cancer. The study population consisted of 26,607 participants from the Alberta’s Tomorrow Project cohort. Two main measures of body shape and size were examined: i) body mass index (BMI) and ii) waist circumference (WC). Incident cancers were identified via linkage to the Alberta Cancer Registry. Cox proportional hazards models were used. Males and females classified as obese (BMI ≥ 30 kg /m−2) have a 33% and 22% increased risk of all-cancer, respectively, than their normal weight counterparts. Similar all-cancer risk increases are observed for those above sex-specific WC guidelines. Mutual adjustment for WC attenuates the association between BMI and all-cancer risk, especially among females. Central adiposity appears to be a stronger predictor of all-cancer risk than body size. Obesity is linked to increased cancer risk but the impact of body size versus weight distribution in determining the increased risk is unclear. Here the authors examined body mass index, waist circumference, and waist to hip ratio in relation to all-cancer incidence and incidence of seven individual cancers in a population of approximately 26,000 individual and conclude that central adiposity appears to be a stronger predictor of all-cancer risk than body size.
Collapse
|
89
|
De Silva S, Tennekoon KH, Karunanayake EH. Overview of the genetic basis toward early detection of breast cancer. BREAST CANCER-TARGETS AND THERAPY 2019; 11:71-80. [PMID: 30718964 PMCID: PMC6345186 DOI: 10.2147/bctt.s185870] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Cancer is a socioeconomical burden in any nation. Out of that, breast cancer is identified as the most common malignancy worldwide among women irrespective of age. As women are an important segment in a community, the weakening of their strength toward the development of a nation is a critical problem in each nation. In this review, it was aimed to discuss the characteristics of cancer genome, cancer genetics, and cancer epigenetics in general and then focus on discussing both genetic and nongenetic factors responsible for the predisposition of breast cancer in humans. More emphasis was placed on genes responsible for the early onset of the disease and which can be used as genetic tools in the identification of the disease at an early stage. Then the context of genetic involvement toward the breast cancer occurrence before age of 40 years was highlighted accordingly. In addition to genetic testing, the review paid adequate attention to mention novel liquid biopsy techniques and other clinical, laboratory, and radiologic assessments. These techniques can be used in early detection and recurrence as well as the surveillance of the patients after primary therapies.
Collapse
Affiliation(s)
- Sumadee De Silva
- Institute of Biochemistry, Molecular Biology and Biotechnology, University of Colombo, Colombo, Sri Lanka,
| | - Kamani Hemamala Tennekoon
- Institute of Biochemistry, Molecular Biology and Biotechnology, University of Colombo, Colombo, Sri Lanka,
| | - Eric Hamilton Karunanayake
- Institute of Biochemistry, Molecular Biology and Biotechnology, University of Colombo, Colombo, Sri Lanka,
| |
Collapse
|
90
|
Dominguez-Valentin M, Nakken S, Tubeuf H, Vodak D, Ekstrøm PO, Nissen AM, Morak M, Holinski-Feder E, Martins A, Møller P, Hovig E. Potentially pathogenic germline CHEK2 c.319+2T>A among multiple early-onset cancer families. Fam Cancer 2019; 17:141-153. [PMID: 28608266 DOI: 10.1007/s10689-017-0011-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
To study the potential contribution of genes other than BRCA1/2, PTEN, and TP53 to the biological and clinical characteristics of multiple early-onset cancers in Norwegian families, including early-onset breast cancer, Cowden-like and Li-Fraumeni-like syndromes (BC, CSL and LFL, respectively). The Hereditary Cancer Biobank from the Norwegian Radium Hospital was used to identify early-onset BC, CSL or LFL for whom no pathogenic variants in BRCA1/2, PTEN, or TP53 had been found in routine diagnostic DNA sequencing. Forty-four cancer susceptibility genes were selected and analyzed by our in-house designed TruSeq amplicon-based assay for targeted sequencing. Protein- and RNA splicing-dedicated in silico analyses were performed for all variants of unknown significance (VUS). Variants predicted as the more likely to affect splicing were experimentally analyzed by minigene assay. We identified a CSL individual carrying a variant in CHEK2 (c.319+2T>A, IVS2), here considered as likely pathogenic. Out of the five VUS (BRCA2, CDH1, CHEK2, MAP3K1, NOTCH3) tested in the minigene splicing assay, only NOTCH3 c.14090C>T (p.Ser497Leu) showed a significant effect on RNA splicing, notably by inducing partial skipping of exon 9. Among 13 early-onset BC, CSL and LFL patients, gene panel sequencing identified a potentially pathogenic variant in CHEK2 that affects a canonical RNA splicing signal. Our study provides new information on genetic loci that may affect the risk of developing cancer in these patients and their families, demonstrating that genes presently not routinely tested in molecular diagnostic settings may be important for capturing cancer predisposition in these families.
Collapse
Affiliation(s)
- Mev Dominguez-Valentin
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.
| | - Sigve Nakken
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Hélène Tubeuf
- Normandy Centre for Genomic and Personalized Medicine, Inserm-U1245, UNIROUEN, Normandie Univ, Rouen, France.,Interactive Biosoftware, Rouen, France
| | - Daniel Vodak
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Per Olaf Ekstrøm
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Anke M Nissen
- Medizinische Klinik und Poliklinik IV, Campus Innenstadt, Klinikum der Universität München, Ziemssenstr. 1, Munich, Germany.,MGZ-Medizinisch Genetisches Zentrum, Munich, Germany
| | - Monika Morak
- Medizinische Klinik und Poliklinik IV, Campus Innenstadt, Klinikum der Universität München, Ziemssenstr. 1, Munich, Germany.,MGZ-Medizinisch Genetisches Zentrum, Munich, Germany
| | - Elke Holinski-Feder
- Medizinische Klinik und Poliklinik IV, Campus Innenstadt, Klinikum der Universität München, Ziemssenstr. 1, Munich, Germany.,MGZ-Medizinisch Genetisches Zentrum, Munich, Germany
| | - Alexandra Martins
- Normandy Centre for Genomic and Personalized Medicine, Inserm-U1245, UNIROUEN, Normandie Univ, Rouen, France
| | - Pål Møller
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.,Department of Human Medicine, Universität Witten/Herdecke, Witten, Germany.,Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Eivind Hovig
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.,Department of Informatics, University of Oslo, Oslo, Norway.,Instituteof Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
91
|
Germline Mutations in BRCA1 and BRCA2 in Breast Cancer Patients with High Genetic Risk in Turkish Population. Int J Breast Cancer 2019; 2019:9645147. [PMID: 30713775 PMCID: PMC6332947 DOI: 10.1155/2019/9645147] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 11/25/2018] [Indexed: 12/23/2022] Open
Abstract
Background The guidelines recommend considering the BRCA1 and BRCA2 germline mutations in female patients with breast carcinomas. In this retrospective study, the BRCA1/2 mutation prevalence in high-risk breast carcinoma patients in a Turkish population was investigated. Materials and Methods In high genetic risk breast carcinoma patients, the BRCA1 and BRCA2 germline mutations were identified by applying next-generation sequencing. Results The results showed BRCA1/2 mutations in 19% of the total patients. In those with first-degree relatives with breast carcinoma histories, the BRCA1/2 mutation prevalence was also 19%. In the patients younger than 40 years old, the BRCA1/2 mutation prevalence was 19.5%. In the triple-negative breast carcinoma patients younger than 60 years old, the BRCA1/2 mutation prevalence was 24.2%. In the patients younger than 40 years old with triple-negative breast carcinomas, BRCA1/2 mutation positivity was found in 37.5% of the patients. Overall, in the Turkish population, the BRCA1/2 mutation prevalence ranges from 19% to 37% in patients with high-risk breast carcinomas. Conclusion It is recommended to check for BRCA1/2 mutations in all high-risk breast carcinoma patients in the Turkish population.
Collapse
|
92
|
Nicolas E, Bertucci F, Sabatier R, Gonçalves A. Targeting BRCA Deficiency in Breast Cancer: What are the Clinical Evidences and the Next Perspectives? Cancers (Basel) 2018; 10:cancers10120506. [PMID: 30544963 PMCID: PMC6316565 DOI: 10.3390/cancers10120506] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 11/27/2018] [Accepted: 12/09/2018] [Indexed: 12/24/2022] Open
Abstract
Breast cancers (BC) associated with germline mutations of BRCA1/2 represent 3–5% of cases. BRCA1/2-associated BC have biological features leading to genomic instability and potential sensitivity to DNA damaging agents, including poly(ADP-ribose) polymerase (PARP) and platinum agents. In this review, we will summarize clinical trials of chemotherapy and PARP inhibitors (PARPi), alone or in combination, at the early or late stage of BRCA1/2-associated BC. We will also present the mechanisms of resistance to PARPi as well as the new therapeutic strategies of association with PARPi. Finally, we will discuss under which conditions the use of DNA damaging agents can be extended to the BRCA1/2-wild type population, the BRCAness concept.
Collapse
Affiliation(s)
- Emanuel Nicolas
- Department of Medical Oncology, Institut Paoli-Calmettes, 13009 Marseille, France.
| | - François Bertucci
- Department of Medical Oncology, Institut Paoli-Calmettes, 13009 Marseille, France.
- CRCM-Predictive Oncology laboratory, Institut Paoli-Calmettes, Inserm U1068, CNRS UMR7258, Aix-Marseille Univ, 13009 Marseille, France.
| | - Renaud Sabatier
- Department of Medical Oncology, Institut Paoli-Calmettes, 13009 Marseille, France.
- CRCM-Predictive Oncology laboratory, Institut Paoli-Calmettes, Inserm U1068, CNRS UMR7258, Aix-Marseille Univ, 13009 Marseille, France.
| | - Anthony Gonçalves
- Department of Medical Oncology, Institut Paoli-Calmettes, 13009 Marseille, France.
- CRCM-Predictive Oncology laboratory, Institut Paoli-Calmettes, Inserm U1068, CNRS UMR7258, Aix-Marseille Univ, 13009 Marseille, France.
| |
Collapse
|
93
|
Li JY, Jing R, Wei H, Wang M, Xiaowei Q, Liu H, Jian L, Ou JH, Jiang WH, Tian FG, Sheng Y, Li HY, Xu H, Zhang RS, Guan AH, Liu K, Jiang HC, Ren Y, He JJ, Huang W, Liao N, Cai X, Ming J, Ling R, Xu Y, Hu CY, Zhang J, Guo B, Ouyang L, Shuai P, Liu Z, Zhong L, Zeng Z, Zhang T, Xuan Z, Tan X, Liang J, Pan Q, Chen L, Zhang F, Fan LJ, Zhang Y, Yang X, BoLi J, Chen C, Jiang J. Germline mutations in 40 cancer susceptibility genes among Chinese patients with high hereditary risk breast cancer. Int J Cancer 2018; 144:281-289. [PMID: 29752822 DOI: 10.1002/ijc.31601] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 04/13/2018] [Accepted: 04/25/2018] [Indexed: 01/08/2023]
Abstract
Multigene panel testing of breast cancer predisposition genes have been extensively conducted in Europe and America, which is relatively rare in Asia however. In this study, we assessed the frequency of germline mutations in 40 cancer predisposition genes, including BRCA1 and BRCA2, among a large cohort of Chinese patients with high hereditary risk of BC. From 2015 to 2016, consecutive BC patients from 26 centers of China with high hereditary risk were recruited (n = 937). Clinical information was collected and next-generation sequencing (NGS) was performed using blood samples of participants to identify germline mutations. In total, we acquired 223 patients with putative germline mutations, including 159 in BRCA1/2, 61 in 15 other BC susceptibility genes and 3 in both BRCA1/2 and non-BRCA1/2 gene. Major mutant non-BRCA1/2 genes were TP53 (n = 18), PALB2 (n = 11), CHEK2 (n = 6), ATM (n = 6) and BARD1 (n = 5). No factors predicted pathologic mutations in non-BRCA1/2 genes when treated as a whole. TP53 mutations were associated with HER-2 positive BC and younger age at diagnosis; and CHEK2 and PALB2 mutations were enriched in patients with luminal BC. Among high hereditary risk Chinese BC patients, 23.8% contained germline mutations, including 6.8% in non-BRCA1/2 genes. TP53 and PALB2 had a relatively high mutation rate (1.9 and 1.2%). Although no factors predicted for detrimental mutations in non-BRCA1/2 genes, some clinical features were associated with mutations of several particular genes.
Collapse
Affiliation(s)
- Jun-Yan Li
- Department of Breast Surgery, Southwest Hospital, the Third Military Medical University, Chongqing, 400038, China
| | - Ruilin Jing
- Annoroad Gene Technology (Beijing) Co. Ltd, Beijing, 100176, China
| | - Hongyi Wei
- Department of Breast Surgery, Southwest Hospital, the Third Military Medical University, Chongqing, 400038, China
| | - Minghao Wang
- Department of Breast Surgery, Southwest Hospital, the Third Military Medical University, Chongqing, 400038, China
| | - Qi Xiaowei
- Department of Breast Surgery, Southwest Hospital, the Third Military Medical University, Chongqing, 400038, China
| | - Haoxi Liu
- Department of Breast Surgery, Southwest Hospital, the Third Military Medical University, Chongqing, 400038, China
| | - Liu Jian
- Department of Medical Oncology, Fujian Provincial Cancer Hospital, Fuzhou, Fujian, 350014, China
| | - Jiang-Hua Ou
- Department of Breast Surgery, The Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830011, China
| | - Wei-Hua Jiang
- Department of Breast Surgery, The Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830011, China
| | - Fu-Guo Tian
- Department of Breast Surgery, Shanxi Cancer Hospital, Taiyuan, Shanxi, 030013, China
| | - Yuan Sheng
- Department of Thyroid and Breast Surgery, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Heng-Yu Li
- Department of Thyroid and Breast Surgery, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Hong Xu
- Department of Breast Surgery, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, 110042, China
| | - Rui-Shan Zhang
- Department of Breast Surgery, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, 110042, China
| | - Ai-Hua Guan
- Department of Breast Surgery, Jilin Cancer Hospital & Institute, Changchun, Jilin, 130000, China
| | - Ke Liu
- Department of Breast Surgery, Jilin Cancer Hospital & Institute, Changchun, Jilin, 130000, China
| | - Hong-Chuan Jiang
- Department of General Surgery, Beijing Chaoyang Hospital Affiliated to Capital Medical University, Beijing, 100020, China
| | - Yu Ren
- Department of Breast Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Jian-Jun He
- Department of Breast Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Weiwei Huang
- Department of Medical Oncology, Fujian Provincial Cancer Hospital, Fuzhou, Fujian, 350014, China
| | - Ning Liao
- Department of Breast, Cancer Center, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510080, China
| | - Xiangjun Cai
- Department of Hepatobiliary and General Surgery, PLA No.202 Hospital, Shenyang Liaoning, 110812, China
| | - Jia Ming
- Department of Breast, Thyroid, and Pancreas Surgery, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Rui Ling
- Depatement of Thyroid, Breast and Vascular Surgery, Xijing hospital, Forth Military University, Xi'an, Shaanxi, 710032, China
| | - Yan Xu
- Department of Breast surgery, Daping Hospital, Research Institute of Surgery, Third Military Medical University, Chongqing, 400038, China
| | - Chun-Yan Hu
- Department of Breast surgery, Daping Hospital, Research Institute of Surgery, Third Military Medical University, Chongqing, 400038, China
| | - Jianguo Zhang
- Department of Breast Surgery, the Second Affiliated Hospital of Harbin Medical College, Harbin, Heilongjiang, 150086, China
| | - Baoliang Guo
- Department of Breast Surgery, the Second Affiliated Hospital of Harbin Medical College, Harbin, Heilongjiang, 150086, China
| | - Lizhi Ouyang
- Hunan Cancer Hospital, Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 4100013, China
| | - Ping Shuai
- Health Management Center, Sichuan Provincial People's Hospital, Chengdu, Sichuan, 610072, China
| | - Zhenzhen Liu
- Department of Breast Surgery, Henan Cancer Hospital, Zhengzhou, Henan, 450008, China
| | - Ling Zhong
- Department of Breast Surgery, Southwest Hospital, the Third Military Medical University, Chongqing, 400038, China
| | - Zhen Zeng
- Department of Breast Surgery, Southwest Hospital, the Third Military Medical University, Chongqing, 400038, China
| | - Ting Zhang
- Department of Breast Surgery, Southwest Hospital, the Third Military Medical University, Chongqing, 400038, China
| | - Zhaoling Xuan
- Annoroad Gene Technology (Beijing) Co. Ltd, Beijing, 100176, China
| | - Xuanni Tan
- Department of Breast Surgery, Southwest Hospital, the Third Military Medical University, Chongqing, 400038, China
| | - Junbin Liang
- Annoroad Gene Technology (Beijing) Co. Ltd, Beijing, 100176, China
| | - Qinwen Pan
- Department of Breast Surgery, Southwest Hospital, the Third Military Medical University, Chongqing, 400038, China
| | - Li Chen
- Department of Breast Surgery, Southwest Hospital, the Third Military Medical University, Chongqing, 400038, China
| | - Fan Zhang
- Department of Breast Surgery, Southwest Hospital, the Third Military Medical University, Chongqing, 400038, China
| | - Lin-Jun Fan
- Department of Breast Surgery, Southwest Hospital, the Third Military Medical University, Chongqing, 400038, China
| | - Yi Zhang
- Department of Breast Surgery, Southwest Hospital, the Third Military Medical University, Chongqing, 400038, China
| | - Xinhua Yang
- Department of Breast Surgery, Southwest Hospital, the Third Military Medical University, Chongqing, 400038, China
| | - Jing BoLi
- Department of Breast Surgery, Southwest Hospital, the Third Military Medical University, Chongqing, 400038, China
| | - Chongjian Chen
- Annoroad Gene Technology (Beijing) Co. Ltd, Beijing, 100176, China
| | - Jun Jiang
- Department of Breast Surgery, Southwest Hospital, the Third Military Medical University, Chongqing, 400038, China
| |
Collapse
|
94
|
Vande Perre P, Toledano D, Corsini C, Escriba E, Laporte M, Bertet H, Yauy K, Toledano A, Galibert V, Baudry K, Clotet L, Million E, Picot M, Geneviève D, Pujol P. Role of the general practitioner in the care of BRCA1 and BRCA2 mutation carriers: General practitioner and patient perspectives. Mol Genet Genomic Med 2018; 6:957-965. [PMID: 30308700 PMCID: PMC6305637 DOI: 10.1002/mgg3.464] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 07/12/2018] [Accepted: 07/20/2018] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND General practitioners (GPs) have an increasing role in referring patients with putative mutation in BRCA1/2 genes for genetics consultation and for long-term follow-up of mutation carriers. METHODS We compared the expectations of the GPs' role according to BRCA1/2 mutation carriers and to GPs themselves. RESULTS Overall, 38% (58/152) of eligible GPs and 70% (176/252) of eligible patients were surveyed. Although 81% of GPs collected the family history, only 24% considered that they know criteria indicating genetics consultation and 39% sufficient knowledge of BRCA1/2 guidelines to answer patients' questions. Twelve% of GPs were aware of the French national guidelines. Among unsatisfied patients, 40% felt that their GP was able to answer (moderately, sufficiently, or completely) specific questions about BRCA1/2 care as compared with 81% in satisfied patients. Only 33% of GPs reported being informed directly by the geneticist about the patients' results. GPs' main expectations for their role in BRCA1/2 carrier care were psychological support and informing relatives about screening (72% and 71%, respectively), which contrasts with the perceptions of patients, who mainly requested medical advice for BRCA1/2-related care (51%). CONCLUSION There is an important need for GP training and enhancing interactions between GPs and geneticists to improve the GP's role in BRCA1/2 screening and management.
Collapse
Affiliation(s)
- Pierre Vande Perre
- Department of Cancer GeneticsMontpellier University Hospital (CHRU)MontpellierFrance
- Université Toulouse III Paul SabatierToulouseFrance
| | - Daniel Toledano
- Department of Cancer GeneticsBreast Disease CenterSaint Louis HospitalParisFrance
| | - Carole Corsini
- Department of Cancer GeneticsMontpellier University Hospital (CHRU)MontpellierFrance
| | - Elsa Escriba
- Department of Cancer GeneticsMontpellier University Hospital (CHRU)MontpellierFrance
| | - Marine Laporte
- Department of Cancer GeneticsMontpellier University Hospital (CHRU)MontpellierFrance
| | - Helena Bertet
- Epidemiology and Clinical Research DepartmentINSERM U1411Clinical Investigation CentreMontpellier University HospitalMontpellierFrance
| | - Kevin Yauy
- Department of GeneticsMontpellier University Hospital (CHRU)MontpellierFrance
- University of MontpellierMontpellierFrance
| | - Alain Toledano
- Department of RadiotherapyHartmann Radiotherapy CenterAmerican Hospital of ParisNeuillyFrance
| | - Virginie Galibert
- Department of Cancer GeneticsMontpellier University Hospital (CHRU)MontpellierFrance
| | - Karen Baudry
- Department of Cancer GeneticsMontpellier University Hospital (CHRU)MontpellierFrance
| | - Lucie Clotet
- University Department of General MedicineUniversity of MontpellierMontpellierFrance
| | - Elodie Million
- University Department of General MedicineUniversity of MontpellierMontpellierFrance
| | - Marie‐Christine Picot
- Department of Medical StatisticsINSERM U1046CNRS UMR 9214University of MontpellierMontpellierFrance
| | - David Geneviève
- Department of GeneticsMontpellier University Hospital (CHRU)MontpellierFrance
- University of MontpellierMontpellierFrance
| | - Pascal Pujol
- Department of Cancer GeneticsMontpellier University Hospital (CHRU)MontpellierFrance
- University of MontpellierMontpellierFrance
| |
Collapse
|
95
|
Loi M, Desideri I, Olmetto E, Francolini G, Greto D, Bonomo P, Simontacchi G, Di Brina L, Meattini I, Livi L. BRCA mutation in breast cancer patients: Prognostic impact and implications on clinical management. Breast J 2018; 24:1019-1023. [PMID: 30066348 DOI: 10.1111/tbj.13100] [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: 02/15/2017] [Revised: 06/08/2017] [Accepted: 06/08/2017] [Indexed: 11/29/2022]
Abstract
BRCA1/2 mutations are involved in breast cancer (BC) susceptibility but their influence on outcome is unclear. We reviewed BC patients tested for BRCA to determine biological features and influence on outcome. BRCA-1 was correlated to younger age (P = 0.035), nodal involvement (P = 0.030), higher tumor grade (P = 0.0022) and Ki-67 (P = 0.014), ER/PgR negative status (P = 0.00042 and 0.0091, respectively), and use of adjuvant chemotherapy (P = 0.000038); BRCA was neither predictive for chemotherapy administration nor resulted in impaired outcome or occurrence of secondary BC. BRCA status did not influence outcome despite higher biological aggressiveness and younger age at presentation.
Collapse
Affiliation(s)
- Mauro Loi
- Radiotherapy Department, University of Florence, Florence, Italy
| | - Isacco Desideri
- Radiotherapy Department, University of Florence, Florence, Italy
| | - Emanuela Olmetto
- Radiotherapy Department, University of Florence, Florence, Italy
| | | | - Daniela Greto
- Radiotherapy Department, University of Florence, Florence, Italy
| | - Pierluigi Bonomo
- Radiotherapy Department, University of Florence, Florence, Italy
| | | | - Lucia Di Brina
- Radiotherapy Department, University of Florence, Florence, Italy
| | - Icro Meattini
- Radiotherapy Department, University of Florence, Florence, Italy
| | - Lorenzo Livi
- Radiotherapy Department, University of Florence, Florence, Italy
| |
Collapse
|
96
|
Scannell Bryan M, Argos M, Andrulis IL, Hopper JL, Chang-Claude J, Malone KE, John EM, Gammon MD, Daly MB, Terry MB, Buys SS, Huo D, Olopade OI, Genkinger JM, Whittemore AS, Jasmine F, Kibriya MG, Chen LS, Ahsan H. Germline Variation and Breast Cancer Incidence: A Gene-Based Association Study and Whole-Genome Prediction of Early-Onset Breast Cancer. Cancer Epidemiol Biomarkers Prev 2018; 27:1057-1064. [PMID: 29898891 DOI: 10.1158/1055-9965.epi-17-1185] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 04/03/2018] [Accepted: 06/08/2018] [Indexed: 01/15/2023] Open
Abstract
Background: Although germline genetics influences breast cancer incidence, published research only explains approximately half of the expected association. Moreover, the accuracy of prediction models remains low. For women who develop breast cancer early, the genetic architecture is less established.Methods: To identify loci associated with early-onset breast cancer, gene-based tests were carried out using exome array data from 3,479 women with breast cancer diagnosed before age 50 and 973 age-matched controls. Replication was undertaken in a population that developed breast cancer at all ages of onset.Results: Three gene regions were associated with breast cancer incidence: FGFR2 (P = 1.23 × 10-5; replication P < 1.00 × 10-6), NEK10 (P = 3.57 × 10-4; replication P < 1.00 × 10-6), and SIVA1 (P = 5.49 × 10-4; replication P < 1.00 × 10-6). Of the 151 gene regions reported in previous literature, 19 (12.5%) showed evidence of association (P < 0.05) with the risk of early-onset breast cancer in the early-onset population. To predict incidence, whole-genome prediction was implemented on a subset of 3,076 participants who were additionally genotyped on a genome wide array. The whole-genome prediction outperformed a polygenic risk score [AUC, 0.636; 95% confidence interval (CI), 0.614-0.659 compared with 0.601; 95% CI, 0.578-0.623], and when combined with known epidemiologic risk factors, the AUC rose to 0.662 (95% CI, 0.640-0.684).Conclusions: This research supports a role for variation within FGFR2 and NEK10 in breast cancer incidence, and suggests SIVA1 as a novel risk locus.Impact: This analysis supports a shared genetic etiology between women with early- and late-onset breast cancer, and suggests whole-genome data can improve risk assessment. Cancer Epidemiol Biomarkers Prev; 27(9); 1057-64. ©2018 AACR.
Collapse
Affiliation(s)
- Molly Scannell Bryan
- Department of Public Health Sciences, University of Chicago, Chicago, Illinois. .,University of Illinois at Chicago, Chicago, Illinois
| | - Maria Argos
- University of Illinois at Chicago, Chicago, Illinois
| | - Irene L Andrulis
- Lunefeld-Tanenbaum Research Institute, Sinai Health System and Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - John L Hopper
- University of Melbourne, Parkville, Victoria, Australia
| | - Jenny Chang-Claude
- Deutsches Krebsforschungszentrum in der Helmholtz-Gemeinshaft, Heidelberg, Germany.,University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Esther M John
- Cancer Prevention Institute of California, Fremont, California.,Stanford Cancer Institute, Stanford, California
| | - Marilie D Gammon
- University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Mary B Daly
- Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | | | | | - Dezheng Huo
- Department of Public Health Sciences, University of Chicago, Chicago, Illinois
| | | | | | | | - Farzana Jasmine
- Department of Public Health Sciences, University of Chicago, Chicago, Illinois
| | - Muhammad G Kibriya
- Department of Public Health Sciences, University of Chicago, Chicago, Illinois
| | - Lin S Chen
- Department of Public Health Sciences, University of Chicago, Chicago, Illinois
| | - Habibul Ahsan
- Department of Public Health Sciences, University of Chicago, Chicago, Illinois
| |
Collapse
|
97
|
|
98
|
Rebbeck TR, Friebel TM, Friedman E, Hamann U, Huo D, Kwong A, Olah E, Olopade OI, Solano AR, Teo SH, Thomassen M, Weitzel JN, Chan TL, Couch FJ, Goldgar DE, Kruse TA, Palmero EI, Park SK, Torres D, van Rensburg EJ, McGuffog L, Parsons MT, Leslie G, Aalfs CM, Abugattas J, Adlard J, Agata S, Aittomäki K, Andrews L, Andrulis IL, Arason A, Arnold N, Arun BK, Asseryanis E, Auerbach L, Azzollini J, Balmaña J, Barile M, Barkardottir RB, Barrowdale D, Benitez J, Berger A, Berger R, Blanco AM, Blazer KR, Blok MJ, Bonadona V, Bonanni B, Bradbury AR, Brewer C, Buecher B, Buys SS, Caldes T, Caliebe A, Caligo MA, Campbell I, Caputo S, Chiquette J, Chung WK, Claes KB, Collée JM, Cook J, Davidson R, de la Hoya M, De Leeneer K, de Pauw A, Delnatte C, Diez O, Ding YC, Ditsch N, Domchek SM, Dorfling CM, Velazquez C, Dworniczak B, Eason J, Easton DF, Eeles R, Ehrencrona H, Ejlertsen B, EMBRACE, Engel C, Engert S, Evans DG, Faivre L, Feliubadaló L, Ferrer SF, Foretova L, Fowler J, Frost D, Galvão HCR, Ganz PA, Garber J, Gauthier-Villars M, Gehrig A, GEMO Study Collaborators, Gerdes AM, Gesta P, Giannini G, Giraud S, Glendon G, et alRebbeck TR, Friebel TM, Friedman E, Hamann U, Huo D, Kwong A, Olah E, Olopade OI, Solano AR, Teo SH, Thomassen M, Weitzel JN, Chan TL, Couch FJ, Goldgar DE, Kruse TA, Palmero EI, Park SK, Torres D, van Rensburg EJ, McGuffog L, Parsons MT, Leslie G, Aalfs CM, Abugattas J, Adlard J, Agata S, Aittomäki K, Andrews L, Andrulis IL, Arason A, Arnold N, Arun BK, Asseryanis E, Auerbach L, Azzollini J, Balmaña J, Barile M, Barkardottir RB, Barrowdale D, Benitez J, Berger A, Berger R, Blanco AM, Blazer KR, Blok MJ, Bonadona V, Bonanni B, Bradbury AR, Brewer C, Buecher B, Buys SS, Caldes T, Caliebe A, Caligo MA, Campbell I, Caputo S, Chiquette J, Chung WK, Claes KB, Collée JM, Cook J, Davidson R, de la Hoya M, De Leeneer K, de Pauw A, Delnatte C, Diez O, Ding YC, Ditsch N, Domchek SM, Dorfling CM, Velazquez C, Dworniczak B, Eason J, Easton DF, Eeles R, Ehrencrona H, Ejlertsen B, EMBRACE, Engel C, Engert S, Evans DG, Faivre L, Feliubadaló L, Ferrer SF, Foretova L, Fowler J, Frost D, Galvão HCR, Ganz PA, Garber J, Gauthier-Villars M, Gehrig A, GEMO Study Collaborators, Gerdes AM, Gesta P, Giannini G, Giraud S, Glendon G, Godwin AK, Greene MH, Gronwald J, Gutierrez-Barrera A, Hahnen E, Hauke J, HEBON, Henderson A, Hentschel J, Hogervorst FB, Honisch E, Imyanitov EN, Isaacs C, Izatt L, Izquierdo A, Jakubowska A, James P, Janavicius R, Jensen UB, John EM, Joseph V, Kaczmarek K, Karlan BY, Kast K, KConFab Investigators, Kim SW, Konstantopoulou I, Korach J, Laitman Y, Lasa A, Lasset C, Lázaro C, Lee A, Lee MH, Lester J, Lesueur F, Liljegren A, Lindor NM, Longy M, Loud JT, Lu KH, Lubinski J, Machackova E, Manoukian S, Mari V, Martínez-Bouzas C, Matrai Z, Mebirouk N, Meijers-Heijboer HE, Meindl A, Mensenkamp AR, Mickys U, Miller A, Montagna M, Moysich KB, Mulligan AM, Musinsky J, Neuhausen SL, Nevanlinna H, Ngeow J, Nguyen HP, Niederacher D, Nielsen HR, Nielsen FC, Nussbaum RL, Offit K, Öfverholm A, Ong KR, Osorio A, Papi L, Papp J, Pasini B, Pedersen IS, Peixoto A, Peruga N, Peterlongo P, Pohl E, Pradhan N, Prajzendanc K, Prieur F, Pujol P, Radice P, Ramus SJ, Rantala J, Rashid MU, Rhiem K, Robson M, Rodriguez GC, Rogers MT, Rudaitis V, Schmidt AY, Schmutzler RK, Senter L, Shah PD, Sharma P, Side LE, Simard J, Singer CF, Skytte AB, Slavin TP, Snape K, Sobol H, Southey M, Steele L, Steinemann D, Sukiennicki G, Sutter C, Szabo CI, Tan YY, Teixeira MR, Terry MB, Teulé A, Thomas A, Thull DL, Tischkowitz M, Tognazzo S, Toland AE, Topka S, Trainer AH, Tung N, van Asperen CJ, van der Hout AH, van der Kolk LE, van der Luijt RB, Van Heetvelde M, Varesco L, Varon-Mateeva R, Vega A, Villarreal-Garza C, von Wachenfeldt A, Walker L, Wang-Gohrke S, Wappenschmidt B, Weber BHF, Yannoukakos D, Yoon SY, Zanzottera C, Zidan J, Zorn KK, Hutten Selkirk CG, Hulick PJ, Chenevix-Trench G, Spurdle AB, Antoniou AC, Nathanson KL, for the CIMBA Consortium. Mutational spectrum in a worldwide study of 29,700 families with BRCA1 or BRCA2 mutations. Hum Mutat 2018; 39:593-620. [PMID: 29446198 PMCID: PMC5903938 DOI: 10.1002/humu.23406] [Show More Authors] [Citation(s) in RCA: 218] [Impact Index Per Article: 31.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 01/09/2018] [Accepted: 01/19/2018] [Indexed: 01/19/2023]
Abstract
The prevalence and spectrum of germline mutations in BRCA1 and BRCA2 have been reported in single populations, with the majority of reports focused on White in Europe and North America. The Consortium of Investigators of Modifiers of BRCA1/2 (CIMBA) has assembled data on 18,435 families with BRCA1 mutations and 11,351 families with BRCA2 mutations ascertained from 69 centers in 49 countries on six continents. This study comprehensively describes the characteristics of the 1,650 unique BRCA1 and 1,731 unique BRCA2 deleterious (disease-associated) mutations identified in the CIMBA database. We observed substantial variation in mutation type and frequency by geographical region and race/ethnicity. In addition to known founder mutations, mutations of relatively high frequency were identified in specific racial/ethnic or geographic groups that may reflect founder mutations and which could be used in targeted (panel) first pass genotyping for specific populations. Knowledge of the population-specific mutational spectrum in BRCA1 and BRCA2 could inform efficient strategies for genetic testing and may justify a more broad-based oncogenetic testing in some populations.
Collapse
Affiliation(s)
- Timothy R. Rebbeck
- Harvard TH Chan School of Public Health and Dana Farber Cancer Institute, 1101 Dana Building, 450 Brookline Ave, Boston, MA 02215, USA
| | - Tara M. Friebel
- Harvard TH Chan School of Public Health and Dana Farber Cancer Institute, 1101 Dana Building, 450 Brookline Ave, Boston, MA 02215, USA
| | - Eitan Friedman
- The Susanne Levy Gertner Oncogenetics Unit, Institute of Human Genetics, Chaim Sheba Medical Center, Ramat Gan 52621, and the Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Ute Hamann
- Molecular Genetics of Breast Cancer, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, 69120 Heidelberg, Germany
| | - Dezheng Huo
- 5841 South Maryland Avenue, MC 2115 Chicago, IL, USA
| | - Ava Kwong
- The Hong Kong Hereditary Breast Cancer Family Registry, Cancer Genetics Center, Hong Kong Sanatorium and Hospital, Hong Kong
| | - Edith Olah
- Department of Molecular Genetics, National Institute of Oncology, Budapest, Hungary
| | | | - Angela R. Solano
- INBIOMED, Faculty of Medicine, University of Buenos Aires/CONICET and CEMIC, Department of Clinical Chemistry, Medical Direction, Buenos Aires, Paraguay 2155, C1121ABG, Argentina
| | - Soo-Hwang Teo
- Cancer Research Initiatives Foundation, Sime Darby Medical Centre, 1 Jalan SS12/1A, Subang Jaya, 47500, Malaysia
| | - Mads Thomassen
- Department of Clinical Genetics, Odense University Hospital, Sonder Boulevard 29, Odense C, Denmark
| | - Jeffrey N. Weitzel
- Clinical Cancer Genetics, City of Hope, 1500 East Duarte Road, Duarte, California 91010 USA
| | - TL Chan
- Division of Molecular Pathology, Department of Pathology, Hong Kong Sanatorium & Hospital, 1/F Li Shu Fan Block, 2 Village Road, Happy Valley, Hong Kong
| | - Fergus J. Couch
- Department of Laboratory Medicine and Pathology, and Health Sciences Research, Mayo Clinic, 200 First Street SW, Rochester, Minnesota, USA
| | - David E. Goldgar
- Department of Dermatology, University of Utah School of Medicine, 30 North 1900 East, SOM 4B454, Salt Lake City, UT 84132, USA
| | - Torben A. Kruse
- Department of Clinical Genetics, Odense University Hospital, Sonder Boulevard 29, Odense C, Denmark
| | - Edenir Inêz Palmero
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo, Brazil
| | - Sue Kyung Park
- 1) Department of Preventive Medicine, Seoul National University College of Medicine; 2) Department of Biomedical Science, Seoul National University Graduate School; 3) Cancer Research Center, Seoul National University, 103 Daehak-ro, Jongno-gu, Seoul, Korea
| | - Diana Torres
- Molecular Genetics of Breast Cancer, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, 69120 Heidelberg, Germany
- Institute of Human Genetics, Pontificia Universidad Javeriana, Carrera 7, Bogota, 11001000, Colombia
| | - Elizabeth J. van Rensburg
- Cancer Genetics Laboratory, Department of Genetics, University of Pretoria, Private Bag X323, Arcadia 0007, South Africa
| | - Lesley McGuffog
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Strangeways Research Laboratory, Worts Causeway, Cambridge, UK
| | - Michael T. Parsons
- Genetics and Computational Biology Department, QIMR Berghofer Medical Research Institute, Herston Road, Brisbane, QLD 4006, Australia
| | - Goska Leslie
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Strangeways Research Laboratory, Worts Causeway, Cambridge, UK
| | - Cora M. Aalfs
- Department of Clinical Genetics, Academic Medical Center, P.O. Box 22700, 1100 DE Amsterdam, The Netherlands
| | - Julio Abugattas
- City of Hope Clinical Cancer Genomics Community Research Network, 1500 East Duarte Road, Duarte, CA 91010, USA
| | - Julian Adlard
- Yorkshire Regional Genetics Service, Chapel Allerton Hospital, Leeds, UK
| | - Simona Agata
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, Via Gattamelata 64, Padua, Italy
| | - Kristiina Aittomäki
- Department of Clinical Genetics, Helsinki University Hospital, P.O. BOX 160 (Meilahdentie 2), 00029 HUS, Finland
| | - Lesley Andrews
- Hereditary Cancer Clinic, Prince of Wales Hospital, High Street, Randwick, NSW 2031 Australia
| | - Irene L. Andrulis
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario M5G 1X5, Canada; Department of Molecular Genetics, University of Toronto, Toronto, Ontario
| | - Adalgeir Arason
- Department of Pathology, hus 9, Landspitali-LSH v/Hringbraut, 101 Reykjavik, Iceland
| | - Norbert Arnold
- Department of Gynaecology and Obstetrics, University Hospital of Schleswig-Holstein, Campus Kiel, Christian-Albrechts University Kiel, Germany
| | - Banu K. Arun
- Department of Breast Medical Oncology and Clinical Cancer Genetics Program, University Of Texas MD Anderson Cancer Center, 1515 Pressler Street, CBP 5, Houston, TX, USA
| | - Ella Asseryanis
- Dept of OB/GYN and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria, Waehringer Guertel 18-20, A 1090 Vienna, Austria
| | - Leo Auerbach
- Dept of OB/GYN and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria, Waehringer Guertel 18-20, A 1090 Vienna, Austria
| | - Jacopo Azzollini
- Unit of Medical Genetics, Department of Medical Oncology and Hematology, Fondazione IRCCS (Istituto Di Ricovero e Cura a Carattere Scientifico) Instituto Nazionale Tumori (INT), Via Giacomo Venezian 1, 20133 Milan, Italy
| | - Judith Balmaña
- Department of Medical Oncology. University Hospital, Vall d'Hebron, Barcelona, Spain
| | - Monica Barile
- Division of Cancer Prevention and Genetics, Istituto Europeo di Oncologia (IEO), via Ripamonti 435, 20141 Milan, Italy
| | - Rosa B. Barkardottir
- Laboratory of Cell Biology, Department of Pathology, hus 9, Landspitali-LSH v/Hringbraut, 101 Reykjavik, Iceland and BMC (Biomedical Centre), Faculty of Medicine, University of Iceland, Vatnsmyrarvegi 16, 101 Reykjavik, Iceland
| | - Daniel Barrowdale
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Strangeways Research Laboratory, Worts Causeway, Cambridge, UK
| | - Javier Benitez
- Human Genetics Group and Genotyping Unit (CEGEN), Human Cancer Genetics Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain. Biomedical Network on Rare Diseases (CIBERER), Madrid, Spain
| | - Andreas Berger
- Dept of OB/GYN, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Raanan Berger
- The Institute of Oncology, Chaim Sheba Medical Center, Ramat Gan 52621, Israel
| | - Amie M. Blanco
- UCSF Cancer Genetics and Prevention Program, San Francisco, CA 94143-1714
| | - Kathleen R. Blazer
- Clinical Cancer Genetics, City of Hope, 1500 East Duarte Road, Duarte, California 91010 USA
| | - Marinus J. Blok
- Department of Clinical Genetics, Maastricht University Medical Center, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
| | - Valérie Bonadona
- Unité de Prévention et d’Epidémiologie Génétique, Centre Léon Bérard, 28 rue Laënnec, Lyon, France
| | - Bernardo Bonanni
- Division of Cancer Prevention and Genetics, Istituto Europeo di Oncologia (IEO), via Ripamonti 435, 20141 Milan, Italy
| | - Angela R. Bradbury
- Department of Medicine, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - Carole Brewer
- Department of Clinical Genetics, Royal Devon & Exeter Hospital, Exeter, UK
| | - Bruno Buecher
- Service de Génétique, Institut Curie, 26, rue d’Ulm, Paris Cedex 05, France
| | - Saundra S. Buys
- Department of Medicine, Huntsman Cancer Institute, 2000 Circle of Hope, Salt Lake City, UT 84112, USA
| | - Trinidad Caldes
- Molecular Oncology Laboratory, Hospital Clinico San Carlos, IdISSC, CIBERONC. Martin Lagos s/n, Madrid, Spain
| | - Almuth Caliebe
- Institute of Human Genetics, University Hospital of Schleswig-Holstein, Campus Kiel, Christian-Albrechts University Kiel, Germany
| | - Maria A. Caligo
- Section of Genetic Oncology, Dept. of Laboratory Medicine, University and University Hospital of Pisa, Pisa, Italy
| | - Ian Campbell
- Research Division, Peter MacCallum Cancer Centre, 305 Gratten Street, Melbourne, VIC 3000, Australia
| | - Sandrine Caputo
- Service de Génétique, Institut Curie, 26, rue d’Ulm, Paris Cedex 05, France
| | - Jocelyne Chiquette
- CRCHU de Quebec-oncologie, Centre des maladies du sein Deschênes-Fabia, Hôpital du Saint-Sacrement,1050, chemin Sainte-Foy, Québec Canada
| | - Wendy K. Chung
- Departments of Pediatrics and Medicine, 1150 St. Nicholas Avenue, Columbia University, New York, NY, 10032 USA
| | - Kathleen B.M. Claes
- Center for Medical Genetics, Ghent University, De Pintelaan 185, 9000 Gent, Belgium
| | - J. Margriet Collée
- Department of Clinical Genetics, Family Cancer Clinic, Erasmus University Medical Center, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Jackie Cook
- Sheffield Clinical Genetics Service, Sheffield Children’s Hospital, Sheffield, UK
| | - Rosemarie Davidson
- Department of Clinical Genetics, South Glasgow University Hospitals, Glasgow, UK
| | - Miguel de la Hoya
- Molecular Oncology Laboratory, Hospital Clinico San Carlos, IdISSC, CIBERONC. Martin Lagos s/n, Madrid, Spain
| | - Kim De Leeneer
- Center for Medical Genetics, Ghent University, De Pintelaan 185, 9000 Gent, Belgium
| | - Antoine de Pauw
- Service de Génétique, Institut Curie, 26, rue d’Ulm, Paris Cedex 05, France
| | - Capucine Delnatte
- Unité d'oncogénétique, ICO-Centre René Gauducheau, Boulevard Jacques Monod, 44805 Nantes Saint Herblain Cedex, France
| | - Orland Diez
- Oncogenetics Group, Vall d’Hebron Institute of Oncology (VHIO), Clinical and Molecular Genetics Area, Vall d’Hebron University Hospital, Passeig Vall d'Hebron 119-129, Barcelona, Spain
| | - Yuan Chun Ding
- Department of Population Sciences, Beckman Research Institute of City of Hope, Duarte, CA USA
| | - Nina Ditsch
- Department of Gynaecology and Obstetrics, Ludwig-Maximilian University Munich, Germany
| | - Susan M. Domchek
- Department of Medicine, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - Cecilia M. Dorfling
- Cancer Genetics Laboratory, Department of Genetics, University of Pretoria, Private Bag X323, Arcadia 0007, South Africa
| | - Carolina Velazquez
- Cáncer Hereditario, Instituto de Biología y Genética Molecular, IBGM, Universidad de Valladolid, Centro Superior de Investigaciones Científicas, UVA-CSIC. Valladolid, Spain
| | - Bernd Dworniczak
- Institute of Human Genetics, University of Münster, Münster, Germany
| | - Jacqueline Eason
- Nottingham Clinical Genetics Service, Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Douglas F. Easton
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Strangeways Research Laboratory, Worts Causeway, Cambridge, UK
| | - Ros Eeles
- Oncogenetics Team, The Institute of Cancer Research and Royal Marsden NHS Foundation Trust, Sutton, UK
| | - Hans Ehrencrona
- Department of Clinical Genetics, Lund University Hospital, Lund, Sweden
| | - Bent Ejlertsen
- Department of Oncology, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, DK-2100 Copenhagen, Denmark
| | - EMBRACE
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Strangeways Research Laboratory, Worts Causeway, Cambridge, UK
| | - Christoph Engel
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Germany
| | - Stefanie Engert
- Department of Gynaecology and Obstetrics, Division of Tumor Genetics, Klinikum rechts der Isar, Technical University Munich, Germany
| | - D. Gareth Evans
- Genomic Medicine, Manchester Academic Health Sciences Centre, Division of Evolution and Genomic Sciences, University of Manchester, Central Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| | - Laurence Faivre
- Centre de Lutte Contre le Cancer Georges François Leclerc, 1 rue Professeur Marion, BP 77 980, Dijon Cedex, France and Genomic and Immunotherapy Medical Institute, Dijon University Hospital, Dijon, France
| | - Lidia Feliubadaló
- Molecular Diagnostic Unit, Hereditary Cancer Program, ICO-IDIBELL (Catalan Institute of Oncology-Bellvitge Biomedical Research Institute), CIBERONC, Gran Via de l'Hospitalet, 199-203. 08908 L'Hospitalet. Barcelona, Spain
| | - Sandra Fert Ferrer
- Laboratoire de Génétique Chromosomique, Hôtel Dieu Centre Hospitalier, BP 1125 Chambéry, France
| | - Lenka Foretova
- Department of Cancer Epidemiology and Genetics, Masaryk Memorial Cancer Institute, Zluty kopec 7, Brno, 65653, Czech Republic
| | - Jeffrey Fowler
- Ohio State University /Columbus Cancer Council, Columbus, OH 43221, USA
| | - Debra Frost
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Strangeways Research Laboratory, Worts Causeway, Cambridge, UK
| | | | - Patricia A. Ganz
- UCLA Schools of Medicine and Public Health, Division of Cancer Prevention & Control Research, Jonsson Comprehensive Cancer Center, 650 Charles Young Drive South, Room A2-125 HS, Los Angeles, CA 90095-6900, USA
| | - Judy Garber
- Cancer Risk and Prevention Clinic, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, USA
| | | | - Andrea Gehrig
- Centre of Familial Breast and Ovarian Cancer, Department of Medical Genetics, Institute of Human Genetics, University Würzburg, Germany
| | - GEMO Study Collaborators
- Institut Curie, Department of Tumour Biology, Paris, France; Institut Curie, INSERM U830, Paris, France
| | - Anne-Marie Gerdes
- Department of Clinical Genetics, Rigshospitalet 4062, Blegdamsvej 9, København Ø, Denmark
| | - Paul Gesta
- Service Régional Oncogénétique Poitou-Charentes, Centre Hospitalier, 79021 Niort
| | - Giuseppe Giannini
- Department of Molecular Medicine, University La Sapienza, and Istituto Pasteur - Fondazione Cenci-Bolognetti, viale Regina Elena 291, 00161 Rome, Italy
| | - Sophie Giraud
- Bâtiment Cheney D, Centre Léon Bérard, 28 rue Laënnec, Lyon, France
| | - Gord Glendon
- Ontario Cancer Genetics Network: Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario M5G 1X5, Canada
| | - Andrew K. Godwin
- Department of Pathology and Laboratory Medicine, 3901 Rainbow Boulevard,4019 Wahl Hall East, MS 3040, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Mark H. Greene
- Clinical Genetics Branch, DCEG, NCI, NIH, 9609 Medical Center Drive, Room 6E-454, Bethesda, MD, USA
| | - Jacek Gronwald
- Department of Genetics and Pathology, Pomeranian Medical University, Unii Lubelskiej 1, Szczecin, Poland
| | - Angelica Gutierrez-Barrera
- Department of Breast Medical Oncology and Clinical Cancer Genetics Program, University Of Texas MD Anderson Cancer Center, 1515 Pressler Street, CBP 5, Houston, TX, USA
| | - Eric Hahnen
- Center for Familial Breast and Ovarian Cancer, Center for Integrated Oncology (CIO), Medical Faculty, University Hospital Cologne, Cologne, Germany
| | - Jan Hauke
- Center for Familial Breast and Ovarian Cancer, Center for Integrated Oncology (CIO), Medical Faculty, University Hospital Cologne, Cologne, Germany
| | - HEBON
- The Hereditary Breast and Ovarian Cancer Research Group Netherlands (HEBON), Coordinating center: Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Alex Henderson
- Institute of Genetic Medicine, Centre for Life, Newcastle Upon Tyne Hospitals NHS Trust, Newcastle upon Tyne, UK
| | - Julia Hentschel
- Institute of Human Genetics, University Leipzig, 04107 Leipzig, Germany
| | - Frans B.L. Hogervorst
- Family Cancer Clinic, Netherlands Cancer Institute, P.O. Box 90203, 1006 BE Amsterdam, The Netherlands
| | - Ellen Honisch
- Department of Gynaecology and Obstetrics, University Hospital Düsseldorf, Heinrich-Heine University Düsseldorf, Germany
| | | | - Claudine Isaacs
- Lombardi Comprehensive Cancer Center, Georgetown University, 3800 Reservoir Road NW, Washington, DC, USA
| | - Louise Izatt
- Clinical Genetics, Guy’s and St. Thomas’ NHS Foundation Trust, London, UK
| | - Angel Izquierdo
- Genetic Counseling Unit, Hereditary Cancer Program, IDIBGI (Institut d'Investigació Biomèdica de Girona), Catalan Institute of Oncology, CIBERONC, Av. França s/n. 1707 Girona, Spain
| | - Anna Jakubowska
- Department of Genetics and Pathology, Pomeranian Medical University, Unii Lubelskiej 1, Szczecin, Poland
| | - Paul James
- Parkville Familial Cancer Centre, Peter MacCallum Cancer Centre, 305 Gratten Street, Melbourne, VIC 3000, Australia
| | - Ramunas Janavicius
- Vilnius University Hospital Santariskiu Clinics, Hereditary Cancer Competence Center Hematology, Oncology and Transfusion Medicine Center Room P519 Santariskiu st. 2, LT-08661 Vilnius, Lithuania
| | - Uffe Birk Jensen
- Department of Clinical Genetics, Aarhus University Hospital, Brendstrupgaardsvej 21C, Aarhus N, Denmark
| | - Esther M. John
- Department of Epidemiology, Cancer Prevention Institute of California, 2201 Walnut Avenue, Suite 300, Fremont, CA 94538, USA and Department of Health Research and Policy (Epidemiology) and Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Vijai Joseph
- Clinical Genetics Research Laboratory, Dept. of Medicine, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10044, USA
| | - Katarzyna Kaczmarek
- Department of Genetics and Pathology, Pomeranian Medical University, Unii Lubelskiej 1, Szczecin, Poland
| | - Beth Y. Karlan
- Women's Cancer Program at the Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Suite 290W, Los Angeles, CA, USA
| | - Karin Kast
- Department of Gynecology and Obstetrics, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany
| | - KConFab Investigators
- Research Department, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia and The Sir Peter MacCallum Department of Oncology University of Melbourne, Parkville, Australia
| | - Sung-Won Kim
- Department of Surgery, Daerim St. Mary's Hospital, 657 Siheung-daero, Yeongdeungpo-gu, Seoul, Korea
| | - Irene Konstantopoulou
- Molecular Diagnostics Laboratory, INRASTES (Institute of Nuclear and Radiological Sciences and Technology), National Centre for Scientific Research "Demokritos", Patriarchou Gregoriou & Neapoleos str., Aghia Paraskevi Attikis, Athens, Greece
| | - Jacob Korach
- The Gyneco-Oncology Department, Chaim Sheba Medical Center, Ramat Gan 52621, Israel
| | - Yael Laitman
- The Susanne Levy Gertner Oncogenetics Unit, Institute of Human Genetics, Chaim Sheba Medical Center, Ramat Gan 52621, and the Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Adriana Lasa
- Servicio de Genética-CIBERER U705, Hospital de la Santa Creu i Sant Pau, Barcelona
| | - Christine Lasset
- Unité de Prévention et d’Epidémiologie Génétique, Centre Léon Bérard, 28 rue Laënnec, Lyon, France
| | - Conxi Lázaro
- Molecular Diagnostic Unit, Hereditary Cancer Program, ICO-IDIBELL (Catalan Institute of Oncology-Bellvitge Biomedical Research Institute), CIBERONC, Gran Via de l'Hospitalet, 199-203. 08908 L'Hospitalet. Barcelona, Spain
| | - Annette Lee
- The Feinstein Institute for Medical Research 350 Community Drive Manhasset NY
| | - Min Hyuk Lee
- Department of Surgery, Soonchunhyang University and Seoul Hospital, 59 Daesagwan-Ro, Yongsan-Gu, Seoul, Korea
| | - Jenny Lester
- Women's Cancer Program at the Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Suite 290W, Los Angeles, CA, USA
| | - Fabienne Lesueur
- Institut Curie, PSL Research University, Mines ParisTech, Inserm U900, 26 rue d'Ulm, F-75005 Paris, France
| | - Annelie Liljegren
- Department of Oncology Radiumhemmet and Institution of Oncology and Patology, Karolinska University Hospital and Karolinska Institutet
| | - Noralane M. Lindor
- Department of Health Sciences Research, Mayo Clinic, 13400 E. Scottsdale Blvd., Scottsdale, AZ, USA
| | - Michel Longy
- Oncogénétique, Institut Bergonié, 229 cours de l'Argonne, 33076 Bordeaux, France
| | - Jennifer T. Loud
- Clinical Genetics Branch, DCEG, NCI, NIH, 9609 Medical Center Drive, Room 6E-536, Bethesda, MD, USA
| | - Karen H. Lu
- Department of Gynecological Oncology and Clinical Cancer Genetics Program, University Of Texas MD Anderson Cancer Center, 1515 Pressler Street, CPB 6, Houston, TX, USA
| | - Jan Lubinski
- Department of Genetics and Pathology, Pomeranian Medical University, Unii Lubelskiej 1, Szczecin, Poland
| | - Eva Machackova
- Department of Cancer Epidemiology and Genetics, Masaryk Memorial Cancer Institute, Zluty kopec 7, Brno, 65653, Czech Republic
| | - Siranoush Manoukian
- Unit of Medical Genetics, Department of Medical Oncology and Hematology, Fondazione IRCCS (Istituto Di Ricovero e Cura a Carattere Scientifico) Instituto Nazionale Tumori (INT), Via Giacomo Venezian 1, 20133 Milan, Italy
| | - Véronique Mari
- Centre Antoine Lacassagne, 33 Avenue de Valombrose, Nice, France
| | - Cristina Martínez-Bouzas
- Laboratorio de Genética Molecular, Servicio de Genética, Hospital Universitario Cruces, BioCruces Health Research Institute, Spain
| | - Zoltan Matrai
- Department of Surgery, National Institute of Oncology, Budapest, Hungary
| | - Noura Mebirouk
- Institut Curie, PSL Research University, Mines ParisTech, Inserm U900, 26 rue d'Ulm, F-75005 Paris, France
| | - Hanne E.J. Meijers-Heijboer
- Department of Clinical Genetics, VU University Medical Center, P.O. Box 7057, 1007 MB Amsterdam, The Netherlands
| | - Alfons Meindl
- Department of Gynaecology and Obstetrics, Division of Tumor Genetics, Klinikum rechts der Isar, Technical University Munich, Germany
| | - Arjen R. Mensenkamp
- Department of Human Genetics, Radboud University Medical Center, P.O. Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Ugnius Mickys
- Vilnius university Santariskiu hospital, National Center of Pathology, Baublio st. 5, Vilnius, Lithuania
| | - Austin Miller
- NRG Oncology, Statistics and Data Management Center, Roswell Park Cancer Institute, Elm St & Carlton St, Buffalo, NY 14263, USA
| | - Marco Montagna
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, Via Gattamelata 64, Padua, Italy
| | - Kirsten B. Moysich
- Department of Cancer Prevention and Control, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Anna Marie Mulligan
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Jacob Musinsky
- Clinical Genetics Research Laboratory, Dept. of Medicine, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10044, USA
| | - Susan L. Neuhausen
- Department of Population Sciences, Beckman Research Institute of City of Hope, Duarte, CA USA
| | - Heli Nevanlinna
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Biomedicum Helsinki, P.O. BOX 700 (Haartmaninkatu 8), 00029 HUS, Finland
| | - Joanne Ngeow
- Cancer Genetics Service, Division of Medical Oncology, National Cancer Centre Singapore, 11 Hospital Drive, Singapore 169610
| | - Huu Phuc Nguyen
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, Germany
| | - Dieter Niederacher
- Department of Gynaecology and Obstetrics, University Hospital Düsseldorf, Heinrich-Heine University Düsseldorf, Germany
| | - Henriette Roed Nielsen
- Department of Clinical Genetics, Odense University Hospital, Sonder Boulevard 29, Odense C, Denmark
| | - Finn Cilius Nielsen
- Center for Genomic Medicine, Rigshospitalet, University of Copenhagen, Denmark
| | | | - Kenneth Offit
- Clinical Genetics Research Laboratory, Dept. of Medicine, Cancer Biology and Genetics, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10044, USA
| | - Anna Öfverholm
- Department of Clinical Genetics, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Kai-ren Ong
- West Midlands Regional Genetics Service, Birmingham Women’s Hospital Healthcare NHS Trust, Edgbaston, Birmingham, UK
| | - Ana Osorio
- Human Genetics Group, Human Cancer Genetics Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain. Biomedical Network on Rare Diseases (CIBERER), Madrid, Spain
| | - Laura Papi
- Unit of Medical Genetics, Department of Biomedical, Experimental and Clinical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| | - Janos Papp
- Department of Molecular Genetics, National Institute of Oncology, Budapest, Hungary
| | - Barbara Pasini
- Department of Medical Sciences, University of Turin, Via Santena 19, 10126 Turin, Italy
| | - Inge Sokilde Pedersen
- Section of Molecular Diagnostics, Department of Biochemistry, Aalborg University Hospital, Reberbansgade 15, Aalborg, Denmark
| | - Ana Peixoto
- Department of Genetics, Portuguese Oncology Institute of Porto (IPO Porto), Porto, Portugal, and Biomedical Sciences Institute (ICBAS), University of Porto, Porto, Portugal
| | - Nina Peruga
- Department of Genetics and Pathology, Pomeranian Medical University, Unii Lubelskiej 1, Szczecin, Poland
| | - Paolo Peterlongo
- IFOM, The FIRC (Italian Foundation for Cancer Research) Institute of Molecular Oncology, via Adamello 16, 20139 Milan, Italy
| | - Esther Pohl
- Center for Familial Breast and Ovarian Cancer, Center for Integrated Oncology (CIO), Medical Faculty, University Hospital Cologne, Cologne, Germany
| | - Nisha Pradhan
- Clinical Genetics Research Laboratory, Dept. of Medicine, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10044, USA
| | - Karolina Prajzendanc
- Department of Genetics and Pathology, Pomeranian Medical University, Unii Lubelskiej 1, Szczecin, Poland
| | - Fabienne Prieur
- Service de Génétique Clinique Chromosomique et Moléculaire, Hôpital Nord, CHU Saint Etienne, St Etienne cedex 2, France
| | - Pascal Pujol
- Unité d'Oncogénétique, CHU Arnaud de Villeneuve, 34295 Montpellier Cedex 5, France
| | - Paolo Radice
- Unit of Molecular Bases of Genetic Risk and Genetic Testing, Department of Research, Fondazione IRCCS (Istituto Di Ricovero e Cura a Carattere Scientifico) Istituto Nazionale Tumori (INT), c/o Amaedeolab, via GA Amadeo 42, 20133 Milan, Italy
| | - Susan J. Ramus
- School of Women's and Children's Health, UNSW Sydney, Australia
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Australia
| | - Johanna Rantala
- Department of Clinical Genetics, Karolinska University Hospital L5:03, Stockholm S-171 76, Sweden
| | - Muhammad Usman Rashid
- Molecular Genetics of Breast Cancer, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, 69120 Heidelberg, Germany
- Department of Basic Sciences, Shaukat Khanum Memorial Cancer Hospital and Research Centre (SKMCH & RC) 7A, Block R3, Johar Town, Lahore, Punjab 54000, Pakistan
| | - Kerstin Rhiem
- Center for Familial Breast and Ovarian Cancer, Center for Integrated Oncology (CIO), Medical Faculty, University Hospital Cologne, Cologne, Germany
| | - Mark Robson
- Clinical Genetics Services, Dept. of Medicine, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY, USA
| | - Gustavo C. Rodriguez
- Division of Gynecologic Oncology, North Shore University Health System, Clinical Professor, University of Chicago, 2650 Ridge Avenue, Suite 1507 Walgreens, Evanston, IL 60201, USA
| | - Mark T. Rogers
- All Wales Medical Genetics Services, University Hospital of Wales, Cardiff, UK
| | - Vilius Rudaitis
- Vilnius University Hospital Santariskiu Clinics, Centre of Woman's Health and pathology, Department of Gynecology, Santariskiu st. 2, Vilnius, Lithuania
| | - Ane Y. Schmidt
- Center for Genomic Medicine, Rigshospitalet, University of Copenhagen, Denmark
| | - Rita Katharina Schmutzler
- Center for Familial Breast and Ovarian Cancer, Center for Integrated Oncology (CIO), Medical Faculty, University Hospital Cologne, Cologne, Germany
| | - Leigha Senter
- Clinical Cancer Genetics Program, Division of Human Genetics, Department of Internal Medicine, The Comprehensive Cancer Center, The Ohio State University, Columbus, USA
| | - Payal D. Shah
- Department of Medicine, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - Priyanka Sharma
- Department of Hematology and Oncology, University of Kansas Medical Center, Suite 210, 2330 Shawnee Mission Parkway, Westwood, KS, USA
| | - Lucy E. Side
- North East Thames Regional Genetics Service, Great Ormond Street Hospital for Children NHS Trust, London, UK
| | - Jacques Simard
- Genomics Center, Centre Hospitalier Universitaire de Québec Research Center and Laval University, 2705 Laurier Boulevard, Quebec City (Quebec), Canada
| | - Christian F. Singer
- Dept of OB/GYN and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria, Waehringer Guertel 18-20, A 1090 Vienna, Austria
| | - Anne-Bine Skytte
- Department of Clinical Genetics, Aarhus University Hospital, Brendstrupgaardsvej 21C, Aarhus N, Denmark
| | - Thomas P. Slavin
- Clinical Cancer Genetics, City of Hope, 1500 East Duarte Road, Duarte, California 91010 USA
| | - Katie Snape
- Medical Genetics Unit, St George's, University of London, UK
| | - Hagay Sobol
- Département Oncologie Génétique, Prévention et Dépistage, Institut Paoli-Calmettes, 232 boulevard Sainte-Margueritte, Marseille, France
| | - Melissa Southey
- Département Oncologie Génétique, Prévention et Dépistage, Institut Paoli-Calmettes, 232 boulevard Sainte-Margueritte, Marseille, France
| | - Linda Steele
- Department of Population Sciences, Beckman Research Institute of City of Hope, Duarte, CA USA
| | - Doris Steinemann
- Institute of Cell and Molecular Pathology, Hannover Medical School, Hannover, Germany
| | - Grzegorz Sukiennicki
- Department of Genetics and Pathology, Pomeranian Medical University, Unii Lubelskiej 1, Szczecin, Poland
| | - Christian Sutter
- Department of Human Genetics, University Hospital Heidelberg, Germany
| | - Csilla I. Szabo
- National Human Genome Research Institute, National Institutes of Health Building 50, Room 5312, 50 South Drive, MSC 004, Bethesda, MD, USA
| | - Yen Y. Tan
- Dept of OB/GYN, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Manuel R. Teixeira
- Department of Genetics, Portuguese Oncology Institute of Porto (IPO Porto), Porto, Portugal, and Biomedical Sciences Institute (ICBAS), University of Porto, Porto, Portugal
| | - Mary Beth Terry
- Department of Epidemiology, Columbia University, New York, NY, USA
| | - Alex Teulé
- Genetic Counseling Unit, Hereditary Cancer Program, IDIBELL (Bellvitge Biomedical Research Institute), Catalan Institute of Oncology, CIBERONC, Gran Via de l'Hospitalet, 199-203. 08908 L'Hospitalet, Barcelona, Spain
| | - Abigail Thomas
- Department of Health Sciences Research, Mayo Clinic, 200 First Street SW, Rochester, Minnesota, USA
| | - Darcy L. Thull
- Department of Medicine, Magee-Womens Hospital, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Marc Tischkowitz
- Program in Cancer Genetics, Departments of Human Genetics and Oncology, McGill University, Montreal, Quebec, Canada
| | - Silvia Tognazzo
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, Via Gattamelata 64, Padua, Italy
| | - Amanda Ewart Toland
- Division of Human Genetics, Departments of Internal Medicine and Cancer Biology and Genetics, Comprehensive Cancer Center, The Ohio State University, 460 W. 12 Avenue, Columbus, OH, USA
| | - Sabine Topka
- Clinical Genetics Research Laboratory, Dept. of Medicine, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10044, USA
| | - Alison H Trainer
- Parkville Familial Cancer Centre, Royal Melbourne Hospital, Melbourne, Australia
| | - Nadine Tung
- Department of Medical Oncology, Beth Israel Deaconess Medical Center, 330 Brookline Avenue Boston, Massachusetts 02215, USA
| | - Christi J. van Asperen
- Department of Clinical Genetics, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, The Netherlands
| | | | | | - Rob B. van der Luijt
- Department of Medical Genetics, University Medical Center Utrecht, The Netherlands
| | | | - Liliana Varesco
- Unit of Hereditary Cancer, Department of Epidemiology, Prevention and Special Functions, IRCCS (Istituto Di Ricovero e Cura a Carattere Scientifico) AOU San Martino - IST Istituto Nazionale per la Ricerca sul Cancro, largo Rosanna Benzi 10, 16132 Genoa, Italy
| | | | - Ana Vega
- Fundación Pública Galega Medicina Xenómica, calle Choupana s/n, Edificio de Consultas, Planta menos dos Santiago de Compostal, A Coruña, Spain
| | - Cynthia Villarreal-Garza
- Departamento de Investigacion y de Tumores Mamarios del Instituto Nacional de Cancerologia, Mexico City; and Centro de Cancer de Mama del Hospital Zambrano Hellion, Tecnologico de Monterrey, San Pedro Garza Garcia, Nuevo Leon
| | | | - Lisa Walker
- Oxford Regional Genetics Service, Churchill Hospital, Oxford, UK
| | - Shan Wang-Gohrke
- Department of Gynaecology and Obstetrics, University Hospital Ulm, Germany
| | - Barbara Wappenschmidt
- Department of Genetics and Pathology, Pomeranian Medical University, Unii Lubelskiej 1, Szczecin, Poland
| | | | - Drakoulis Yannoukakos
- Molecular Diagnostics Laboratory, INRASTES (Institute of Nuclear and Radiological Sciences and Technology), National Centre for Scientific Research "Demokritos", Patriarchou Gregoriou & Neapoleos str., Aghia Paraskevi Attikis, Athens, Greece
| | - Sook-Yee Yoon
- Cancer Research Initiatives Foundation, Sime Darby Medical Centre, 1 Jalan SS12/1A, Subang Jaya, 47500, Malaysia
| | - Cristina Zanzottera
- Unit of Medical Genetics, Department of Medical Oncology and Hematology, Fondazione IRCCS (Istituto Di Ricovero e Cura a Carattere Scientifico) Instituto Nazionale Tumori (INT), Via Giacomo Venezian 1, 20133 Milan, Italy
| | - Jamal Zidan
- Institute of Oncology, Rivka Ziv Medical Center, 13000 Zefat, Israel
| | - Kristin K. Zorn
- Magee-Womens Hospital, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Christina G. Hutten Selkirk
- Center for Medical Genetics, NorthShore University HealthSystem,1000 Central St, Suite 620, Evanston, IL, USA
| | - Peter J. Hulick
- Medical Director, Center for Medical Genetics, North Shore University Health System, Clinical Assistant Professor of Medicine, University of Chicago Pritzker School of Medicine, 1000 Central Street, Suite 620, Evanston, IL 60201, USA
| | - Georgia Chenevix-Trench
- Genetics and Computational Biology Department, QIMR Berghofer Medical Research Institute, Herston Road, Brisbane, QLD 4006, Australia
| | - Amanda B. Spurdle
- Genetics and Computational Biology Department, QIMR Berghofer Medical Research Institute, Herston Road, Brisbane, QLD 4006, Australia
| | - Antonis C. Antoniou
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Strangeways Research Laboratory, Worts Causeway, Cambridge, UK
| | - Katherine L. Nathanson
- Department of Medicine, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA
| | | |
Collapse
|
99
|
Wei H, Wang M, Ou J, Jiang W, Tian F, Sheng Y, Li H, Xu H, Zhang R, Guan A, Wang C, Jiang H, Ren Y, He J, Liu J, Huang W, Liao N, Cai X, Ming J, Ling R, Xu Y, Hu C, Zhang J, Guo B, Ouyang L, Shuai P, Liu Z, Zhong L, Jing R, Zeng Z, Zhang M, Zhang T, Xuan Z, Tan X, Liang J, Pan Q, Chen L, Zhang F, Fan L, Zhang Y, Yang X, Li J, Chen C, Jiang J. Multicenter cross-sectional screening of the BRCA gene for Chinese high hereditary risk breast cancer populations. Oncol Lett 2018; 15:9420-9428. [PMID: 29805665 DOI: 10.3892/ol.2018.8538] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 10/25/2017] [Indexed: 11/06/2022] Open
Abstract
Due to lack of systematic reviews, BRCA, DNA Repair Associated (BRCA) mutations in the Chinese population are not completely understood. The following study investigates the prevalence and type of BRCA mutations in Chinese patients with high hereditary risk of breast cancer (BC). Patients Drwere recruited from 14 cities between October 2015 and February 2016, and were selected based on family and personal medical history. BRCA mutations were analyzed by collecting blood samples from all participants. 437 BC patients were included. A total of seventy-six (17.4%) mutation carriers were identified with no geographic difference. The mutation rate in the early-onset BC patients was lower compared to family history of breast/ovarian cancer (OC), bilateral BC, male BC, BC&OC or meeting ≥2 criteria (9.2 vs. 21.7, 24.0, 22.2, 16.7 and 24.3%, respectively, P=0.007). A total of 61 mutation sites were identified (BRCA1 32, BRCA2 29) including 47.5% novel sites and extra 10 variants of uncertain significance. A total of five sites were repeated in more than one unrelated patient. A total of 11 sites were associated with hereditary breast and ovarian cancer syndrome, two of which were confirmed by family pedigrees. Compared with BRCA- patients, patients with BRCA1 mutation tended to be triple-negative BC (P<0.001), whereas patients with BRCA2 mutation were more likely to be hormone receptor positive BC (P=0.02). The present study provides a general BRCA mutation profile in the Chinese population. The prevalence of BRCA mutation in BC patients with high hereditary risk is lower compared with Western populations. Chinese mutation type is different with Western people, without obvious founder mutation.
Collapse
Affiliation(s)
- Hongyi Wei
- Department of Breast Surgery, Southwest Hospital, The Third Military Medical University, Chongqing 400038, P.R. China
| | - Minghao Wang
- Department of Breast Surgery, Southwest Hospital, The Third Military Medical University, Chongqing 400038, P.R. China
| | - Jianghua Ou
- Department of Breast Surgery, The Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830011, P.R. China
| | - Weihua Jiang
- Department of Breast Surgery, The Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830011, P.R. China
| | - Fuguo Tian
- Breast Surgery, Shanxi Cancer Hospital, Taiyuan, Shanxi 030013, P.R. China
| | - Yuan Sheng
- Department of Thyroid and Breast Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, P.R. China
| | - Hengyu Li
- Department of Thyroid and Breast Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, P.R. China
| | - Hong Xu
- Department of Breast Surgery, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110042, P.R. China
| | - Ruishan Zhang
- Department of Breast Surgery, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110042, P.R. China
| | - Aihua Guan
- Department of Breast Surgery, Jilin Cancer Hospital and Institute, Changchun, Jilin 130000, P.R. China
| | - Changqing Wang
- Department of Breast Surgery, Jilin Cancer Hospital and Institute, Changchun, Jilin 130000, P.R. China
| | - Hongchuan Jiang
- Department of General Surgery, Beijing Chaoyang Hospital Affiliated to Capital Medical University, Beijing 100020, P.R. China
| | - Yu Ren
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Jianjun He
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Jian Liu
- Department of Medical Oncology, Fujian Provincial Cancer Hospital, Fuzhou, Fujian 350014, P.R. China
| | - Weiwei Huang
- Department of Medical Oncology, Fujian Provincial Cancer Hospital, Fuzhou, Fujian 350014, P.R. China
| | - Ning Liao
- Breast Department, Cancer Center, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Xiangjun Cai
- Department of Hepatobiliary and General Surgery, PLA No. 202 Hospital, Shenyang, Liaoning 110812, P.R. China
| | - Jia Ming
- Department of Breast, Thyroid, and Pancreas Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Rui Ling
- Depatement of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Forth Military University, Xi'an, Shaanxi 710032, P.R. China
| | - Yan Xu
- Department of Breast Surgery, Daping Hospital, Research Institute of Surgery, Third Military Medical University, Chongqing 400038, P.R. China
| | - Chunyan Hu
- Department of Breast Surgery, Daping Hospital, Research Institute of Surgery, Third Military Medical University, Chongqing 400038, P.R. China
| | - Jianguo Zhang
- Department of Breast Surgery, The Second Affiliated Hospital of Harbin Medical College, Harbin, Heilongjiang 150086, P.R. China
| | - Baoliang Guo
- Department of Breast Surgery, The Second Affiliated Hospital of Harbin Medical College, Harbin, Heilongjiang 150086, P.R. China
| | - Lizhi Ouyang
- Department of Breast Surgery, Hunan Cancer Hospital, Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 4100013, P.R. China
| | - Ping Shuai
- Health Management Center, Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
| | - Zhenzhen Liu
- Department of Breast Surgery, Henan Cancer Hospital, Zhengzhou, Henan 450008, P.R. China
| | - Ling Zhong
- Department of Breast Surgery, Southwest Hospital, The Third Military Medical University, Chongqing 400038, P.R. China
| | - Ruilin Jing
- Annoroad Gene Technology (Beijing) Co., Ltd., Beijing 100176, P.R. China
| | - Zhen Zeng
- Department of Breast Surgery, Southwest Hospital, The Third Military Medical University, Chongqing 400038, P.R. China
| | - Meng Zhang
- Annoroad Gene Technology (Beijing) Co., Ltd., Beijing 100176, P.R. China
| | - Ting Zhang
- Department of Breast Surgery, Southwest Hospital, The Third Military Medical University, Chongqing 400038, P.R. China
| | - Zhaoling Xuan
- Annoroad Gene Technology (Beijing) Co., Ltd., Beijing 100176, P.R. China
| | - Xuanni Tan
- Department of Breast Surgery, Southwest Hospital, The Third Military Medical University, Chongqing 400038, P.R. China
| | - Junbin Liang
- Annoroad Gene Technology (Beijing) Co., Ltd., Beijing 100176, P.R. China
| | - Qinwen Pan
- Department of Breast Surgery, Southwest Hospital, The Third Military Medical University, Chongqing 400038, P.R. China
| | - Li Chen
- Department of Breast Surgery, Southwest Hospital, The Third Military Medical University, Chongqing 400038, P.R. China
| | - Fan Zhang
- Department of Breast Surgery, Southwest Hospital, The Third Military Medical University, Chongqing 400038, P.R. China
| | - Linjun Fan
- Department of Breast Surgery, Southwest Hospital, The Third Military Medical University, Chongqing 400038, P.R. China
| | - Yi Zhang
- Department of Breast Surgery, Southwest Hospital, The Third Military Medical University, Chongqing 400038, P.R. China
| | - Xinhua Yang
- Department of Breast Surgery, Southwest Hospital, The Third Military Medical University, Chongqing 400038, P.R. China
| | - Jingbo Li
- Department of Breast Surgery, Southwest Hospital, The Third Military Medical University, Chongqing 400038, P.R. China
| | - Chongjian Chen
- Annoroad Gene Technology (Beijing) Co., Ltd., Beijing 100176, P.R. China
| | - Jun Jiang
- Department of Breast Surgery, Southwest Hospital, The Third Military Medical University, Chongqing 400038, P.R. China
| |
Collapse
|
100
|
Norum J, Grindedal EM, Heramb C, Karsrud I, Ariansen SL, Undlien DE, Schlichting E, Mæhle L. BRCA mutation carrier detection. A model-based cost-effectiveness analysis comparing the traditional family history approach and the testing of all patients with breast cancer. ESMO Open 2018; 3:e000328. [PMID: 29682331 PMCID: PMC5905828 DOI: 10.1136/esmoopen-2018-000328] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 02/20/2018] [Accepted: 02/21/2018] [Indexed: 12/29/2022] Open
Abstract
Background Identification of BRCA mutation carriers among patients with breast cancer (BC) involves costs and gains. Testing has been performed according to international guidelines, focusing on family history (FH) of breast and/or ovarian cancer. An alternative is testing all patients with BC employing sequencing of the BRCA genes and Multiplex Ligation Probe Amplification (MLPA). Patients and methods A model-based cost-effectiveness analysis, employing data from Oslo University Hospital, Ullevål (OUH-U) and a decision tree, was done. The societal and the healthcare perspectives were focused and a lifetime perspective employed. The comparators were the traditional FH approach used as standard of care at OUH-U in 2013 and the intervention (testing all patients with BC) performed in 2014 and 2015 at the same hospital. During the latter period, 535 patients with BC were offered BRCA testing with sequencing and MLPA. National 2014 data on mortality rates and costs were implemented, a 3% discount rate used and the costing year was 2015. The incremental cost-effectiveness ratio was calculated in euros (€) per life-year gained (LYG). Results The net healthcare cost (healthcare perspective) was €40 503/LYG. Including all resource use (societal perspective), the cost was €5669/LYG. The univariate sensitivity analysis documented the unit cost of the BRCA test and the number of LYGs the prominent parameters affecting the result. Diagnostic BRCA testing of all patients with BC was superior to the FH approach and cost-effective within the frequently used thresholds (healthcare perspective) in Norway (€60 000–€80 000/LYG).
Collapse
Affiliation(s)
- Jan Norum
- Department of Surgery, Finnmark Hospital, Hammerfest, Norway
| | | | - Cecilie Heramb
- Department of Medical Genetics, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Inga Karsrud
- Section for Breast and Endocrine Surgery, Department of Cancer, Oslo University Hospital, Oslo, Norway
| | - Sarah Louise Ariansen
- Department of Medical Genetics, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Dag Erik Undlien
- Section for Breast and Endocrine Surgery, Department of Cancer, Oslo University Hospital, Oslo, Norway
| | - Ellen Schlichting
- Section for Breast and Endocrine Surgery, Department of Cancer, Oslo University Hospital, Oslo, Norway
| | - Lovise Mæhle
- Department of Medical Genetics, Oslo University Hospital and University of Oslo, Oslo, Norway
| |
Collapse
|