1
|
Yang X, Mooij TM, Leslie G, Ficorella L, Andrieu N, Kast K, Singer CF, Jakubowska A, van Gils CH, Tan YY, Engel C, Adank MA, van Asperen CJ, Ausems MGEM, Berthet P, Collee MJ, Cook JA, Eason J, Spaendonck-Zwarts KYV, Evans DG, Gómez García EB, Hanson H, Izatt L, Kemp Z, Lalloo F, Lasset C, Lesueur F, Musgrave H, Nambot S, Noguès C, Oosterwijk JC, Stoppa-Lyonnet D, Tischkowitz M, Tripathi V, Wevers MR, Zhao E, van Leeuwen FE, Schmidt MK, Easton DF, Rookus MA, Antoniou AC. Validation of the BOADICEA model in a prospective cohort of BRCA1/2 pathogenic variant carriers. J Med Genet 2024; 61:803-809. [PMID: 38834293 PMCID: PMC11287562 DOI: 10.1136/jmg-2024-109943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 05/12/2024] [Indexed: 06/06/2024]
Abstract
BACKGROUND No validation has been conducted for the BOADICEA multifactorial breast cancer risk prediction model specifically in BRCA1/2 pathogenic variant (PV) carriers to date. Here, we evaluated the performance of BOADICEA in predicting 5-year breast cancer risks in a prospective cohort of BRCA1/2 PV carriers ascertained through clinical genetic centres. METHODS We evaluated the model calibration and discriminatory ability in the prospective TRANsIBCCS cohort study comprising 1614 BRCA1 and 1365 BRCA2 PV carriers (209 incident cases). Study participants had lifestyle, reproductive, hormonal, anthropometric risk factor information, a polygenic risk score based on 313 SNPs and family history information. RESULTS The full multifactorial model considering family history together with all other risk factors was well calibrated overall (E/O=1.07, 95% CI: 0.92 to 1.24) and in quintiles of predicted risk. Discrimination was maximised when all risk factors were considered (Harrell's C-index=0.70, 95% CI: 0.67 to 0.74; area under the curve=0.79, 95% CI: 0.76 to 0.82). The model performance was similar when evaluated separately in BRCA1 or BRCA2 PV carriers. The full model identified 5.8%, 12.9% and 24.0% of BRCA1/2 PV carriers with 5-year breast cancer risks of <1.65%, <3% and <5%, respectively, risk thresholds commonly used for different management and risk-reduction options. CONCLUSION BOADICEA may be used to aid personalised cancer risk management and decision-making for BRCA1 and BRCA2 PV carriers. It is implemented in the free-access CanRisk tool (https://www.canrisk.org/).
Collapse
Affiliation(s)
- Xin Yang
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Strangeways Research Laboratory, Cambridge, UK
| | - Thea M Mooij
- Department of Epidemiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Goska Leslie
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Strangeways Research Laboratory, Cambridge, UK
| | - Lorenzo Ficorella
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Strangeways Research Laboratory, Cambridge, UK
| | - Nadine Andrieu
- INSERM U900, Paris, France
- Institut Curie, Paris, France
- Mines ParisTech, Fontainebleau, France
- PSL Research University, Paris, France
| | - Karin Kast
- Center for Hereditary Breast and Ovarian Cancer, Center for Integrated Oncology (CIO), Medical Faculty, University Hospital Cologne, Cologne, Germany
| | - Christian F Singer
- Department of OB/GYN and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Anna Jakubowska
- Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
- Independent Laboratory of Molecular Biology and Genetic Diagnostics, Pomeranian Medical University, Szczecin, Poland
| | - Carla H van Gils
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Yen Y Tan
- Department of OB/GYN and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Christoph Engel
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
| | - Muriel A Adank
- Department of Clinical Genetics, The Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Christi J van Asperen
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Margreet G E M Ausems
- Devision Laboratories, Pharmacy and Biomedical Genetics, Department of Genetics, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Pascaline Berthet
- Oncogénétique Département de Biopathologie, Centre François Baclesse, Caen, France
| | - Margriet J Collee
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Jackie A Cook
- Sheffield Clinical Genetics Service, Scheffield Children's Hospital, Sheffield, UK
| | - Jacqueline Eason
- Nottingham Clinical Genetics Service, Nottingham University Hospitals NHS Trust, Nottingham, UK
| | | | - D Gareth Evans
- The Prevent Breast Cancer Research Unit, The Nightingale Centre, Manchester University NHS Foundation Trust, Manchester, UK
- Genomic Medicine, Division of Evolution and Genomic Sciences, The University of Manchester, St Mary's Hospital, Manchester University NHS Foundation Trust, Manchester, UK
- Manchester Breast Centre, Oglesby Cancer Research Centre, The Christie, University of Manchester, Manchester, UK
| | | | - Helen Hanson
- South West Thames Regional Genetics Service, St George's University Hospitals NHS Foundation Trust, London, UK
| | - Louise Izatt
- Department of Clinical Genetics, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Zoe Kemp
- Department of Cancer Genetics, Royal Marsden Hospital, NHS Trust, London, UK
| | - Fiona Lalloo
- Clinical Genetics Service, Manchester Centre for Genomic Medicine, Manchester University Hospitals Foundation Trust, Manchester, UK
| | - Christine Lasset
- Université Claude Bernard Lyon 1, Villeurbanne, France
- CNRS UMR 5558, Lyon, France
- Centre Léon Bérard, Unité de Prévention et Epidémiologie Génétique, Lyon, France
| | - Fabienne Lesueur
- INSERM U900, Paris, France
- Institut Curie, Paris, France
- Mines ParisTech, Fontainebleau, France
- PSL Research University, Paris, France
| | - Hannah Musgrave
- Yorkshire Regional Genetics Service, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Sophie Nambot
- Centre de Génétique et Centre de Référence Anomalies du Développement et Syndromes Malformatifs, CHU de Dijon, Hôpital d'Enfants, Dijon, France
- Centre de Lutte contre le Cancer Georges François Leclerc, Dijon, France
| | - Catherine Noguès
- Département d'Anticipation et de Suivi des Cancers, Oncogénétique Clinique, Institut Paoli Calmettes, Marseille, France
- Aix Marseille Univ, INSERM, IRD, SESSTIM, Marseille, France
| | - Jan C Oosterwijk
- Department of Genetics, University of Groningen, University Medical Center, Groningen, The Netherlands
| | - Dominique Stoppa-Lyonnet
- Institut Curie, Service de Génétique, Paris, France
- Université Paris CIté, Paris, France
- INSERM U830, Paris, France
| | - Marc Tischkowitz
- Department of Medical Genetics, National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | - Vishakha Tripathi
- Clinical Genetics Service, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Marijke R Wevers
- Department of Clinical Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Emily Zhao
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Strangeways Research Laboratory, Cambridge, UK
| | - Flora E van Leeuwen
- Department of Epidemiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Marjanka K Schmidt
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands
- Division of Molecular Pathology and Division of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Douglas F Easton
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Strangeways Research Laboratory, Cambridge, UK
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Matti A Rookus
- Department of Epidemiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Antonis C Antoniou
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Strangeways Research Laboratory, Cambridge, UK
| |
Collapse
|
2
|
Mohammadi S, Ghaderi S, Mohammadi M, Ghaznavi H, Mohammadian K. Breast percent density changes in digital mammography pre- and post-radiotherapy. J Med Radiat Sci 2024. [PMID: 38571377 DOI: 10.1002/jmrs.788] [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: 10/07/2023] [Accepted: 03/20/2024] [Indexed: 04/05/2024] Open
Abstract
INTRODUCTION Breast cancer (BC), the most frequently diagnosed malignancy among women worldwide, presents a public health challenge and affects mortality rates. Breast-conserving therapy (BCT) is a common treatment, but the risk from residual disease necessitates radiotherapy. Digital mammography monitors treatment response by identifying post-operative and radiotherapy tissue alterations, but accurate assessment of mammographic density remains a challenge. This study used OpenBreast to measure percent density (PD), offering insights into changes in mammographic density before and after BCT with radiation therapy. METHODS This retrospective analysis included 92 female patients with BC who underwent BCT, chemotherapy, and radiotherapy, excluding those who received hormonal therapy or bilateral BCT. Percent/percentage density measurements were extracted using OpenBreast, an automated software that applies computational techniques to density analyses. Data were analysed at baseline, 3 months, and 15 months post-treatment using standardised mean difference (SMD) with Cohen's d, chi-square, and paired sample t-tests. The predictive power of PD changes for BC was measured based on the receiver operating characteristic (ROC) curve analysis. RESULTS The mean age was 53.2 years. There were no significant differences in PD between the periods. Standardised mean difference analysis revealed no significant changes in the SMD for PD before treatment compared with 3- and 15-months post-treatment. Although PD increased numerically after radiotherapy, ROC analysis revealed optimal sensitivity at 15 months post-treatment for detecting changes in breast density. CONCLUSIONS This study utilised an automated breast density segmentation tool to assess the changes in mammographic density before and after BC treatment. No significant differences in the density were observed during the short-term follow-up period. However, the results suggest that quantitative density assessment could be valuable for long-term monitoring of treatment effects. The study underscores the necessity for larger and longitudinal studies to accurately measure and validate the effectiveness of quantitative methods in clinical BC management.
Collapse
Affiliation(s)
- Sana Mohammadi
- Department of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Sadegh Ghaderi
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahdi Mohammadi
- Department of Medical Physics and Biomedical Engineering, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamid Ghaznavi
- Department of Radiology, Faculty of Paramedical Sciences, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Kamal Mohammadian
- Department of Radiation Oncology, Hamadan University of Medical Sciences, Mahdieh Center, Hamadan, Iran
| |
Collapse
|
3
|
Majithia J, Mahajan A, Vaish R, Prakash G, Patwardhan S, Sarin R. Imaging Recommendations for Diagnosis, Staging, and Management of Hereditary Malignancies. Indian J Med Paediatr Oncol 2023. [DOI: 10.1055/s-0042-1760325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2023] Open
Abstract
AbstractHereditary cancer syndromes, characterized by genetically distinct neoplasms developing in specific organs in more than one family members, predispose an individual to early onset of distinct site-specific tumors. Early age of onset, multiorgan involvement, multiple and bilateral tumors, advanced disease at presentation, and aggressive tumor histology are few characteristic features of hereditary cancer syndromes. A multidisciplinary approach to hereditary cancers has led to a paradigm shift in the field of preventive oncology and precision medicine. Imaging plays a pivotal role in the screening, testing, and follow-up of individuals and their first- and second-degree relatives with hereditary cancers. In fact, a radiologist is often the first to apprise the clinician about the possibility of an underlying hereditary cancer syndrome based on pathognomonic imaging findings. This article focuses on the imaging spectrum of few common hereditary cancer syndromes with specific mention of the imaging features of associated common and uncommon tumors in each syndrome. The screening and surveillance recommendations for each condition with specific management approaches, in contrast to sporadic cases, have also been described.
Collapse
Affiliation(s)
- Jinita Majithia
- Department of Radiodiagnosis, Tata Memorial Hospital, Mumbai, Maharashtra, India
| | - Abhishek Mahajan
- Department of Radiology, The Clatterbridge Cancer Centre NHS Foundation Trust, Liverpool, United Kingdom
| | - Richa Vaish
- Department of Head and Neck Oncology, Tata Memorial Hospital, Mumbai, Maharashtra, India
| | - Gagan Prakash
- Department of Uro-Oncology, Tata Memorial Hospital, Mumbai, Maharashtra, India
| | - Saket Patwardhan
- Department of Radiodiagnosis, Tata Memorial Hospital, Mumbai, Maharashtra, India
| | - Rajiv Sarin
- Department of Radiation Oncology and In-Charge Cancer Genetics, Tata Memorial Hospital and Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Mumbai, Maharashtra, India
| |
Collapse
|
4
|
Kim J, Haffty BG. Genetic Factors in the Screening and Imaging for Breast Cancer. Korean J Radiol 2023; 24:378-383. [PMID: 37056158 PMCID: PMC10157325 DOI: 10.3348/kjr.2023.0012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 02/20/2023] [Accepted: 03/01/2023] [Indexed: 04/05/2023] Open
Affiliation(s)
- Jongmyung Kim
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers Robert Wood Johnson Medical School and Rutgers New Jersey Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Bruce George Haffty
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers Robert Wood Johnson Medical School and Rutgers New Jersey Medical School, Rutgers University, New Brunswick, NJ, USA
| |
Collapse
|
5
|
Li S, Nguyen TL, Nguyen-Dumont T, Dowty JG, Dite GS, Ye Z, Trinh HN, Evans CF, Tan M, Sung J, Jenkins MA, Giles GG, Hopper JL, Southey MC. Genetic Aspects of Mammographic Density Measures Associated with Breast Cancer Risk. Cancers (Basel) 2022; 14:2767. [PMID: 35681745 PMCID: PMC9179294 DOI: 10.3390/cancers14112767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 04/27/2022] [Accepted: 05/30/2022] [Indexed: 11/26/2022] Open
Abstract
Cumulus, Altocumulus, and Cirrocumulus are measures of mammographic density defined at increasing pixel brightness thresholds, which, when converted to mammogram risk scores (MRSs), predict breast cancer risk. Twin and family studies suggest substantial variance in the MRSs could be explained by genetic factors. For 2559 women aged 30 to 80 years (mean 54 years), we measured the MRSs from digitized film mammograms and estimated the associations of the MRSs with a 313-SNP breast cancer polygenic risk score (PRS) and 202 individual SNPs associated with breast cancer risk. The PRS was weakly positively correlated (correlation coefficients ranged 0.05−0.08; all p < 0.04) with all the MRSs except the Cumulus-white MRS based on the “white but not bright area” (correlation coefficient = 0.04; p = 0.06). After adjusting for its association with the Altocumulus MRS, the PRS was not associated with the Cumulus MRS. There were MRS associations (Bonferroni-adjusted p < 0.04) with one SNP in the ATXN1 gene and nominally with some ESR1 SNPs. Less than 1% of the variance of the MRSs is explained by the genetic markers currently known to be associated with breast cancer risk. Discovering the genetic determinants of the bright, not white, regions of the mammogram could reveal substantial new genetic causes of breast cancer.
Collapse
Affiliation(s)
- Shuai Li
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, VIC 3051, Australia; (S.L.); (T.L.N.); (J.G.D.); (G.S.D.); (Z.Y.); (H.N.T.); (C.F.E.); (M.A.J.); (G.G.G.)
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge CB1 8RN, UK
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, VIC 3168, Australia; (T.N.-D.); (M.C.S.)
| | - Tuong L. Nguyen
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, VIC 3051, Australia; (S.L.); (T.L.N.); (J.G.D.); (G.S.D.); (Z.Y.); (H.N.T.); (C.F.E.); (M.A.J.); (G.G.G.)
| | - Tu Nguyen-Dumont
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, VIC 3168, Australia; (T.N.-D.); (M.C.S.)
- Department of Clinical Pathology, The University of Melbourne, Parkville, VIC 3051, Australia
| | - James G. Dowty
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, VIC 3051, Australia; (S.L.); (T.L.N.); (J.G.D.); (G.S.D.); (Z.Y.); (H.N.T.); (C.F.E.); (M.A.J.); (G.G.G.)
| | - Gillian S. Dite
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, VIC 3051, Australia; (S.L.); (T.L.N.); (J.G.D.); (G.S.D.); (Z.Y.); (H.N.T.); (C.F.E.); (M.A.J.); (G.G.G.)
- Genetic Technologies Limited, Fitzroy, VIC 3065, Australia
| | - Zhoufeng Ye
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, VIC 3051, Australia; (S.L.); (T.L.N.); (J.G.D.); (G.S.D.); (Z.Y.); (H.N.T.); (C.F.E.); (M.A.J.); (G.G.G.)
| | - Ho N. Trinh
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, VIC 3051, Australia; (S.L.); (T.L.N.); (J.G.D.); (G.S.D.); (Z.Y.); (H.N.T.); (C.F.E.); (M.A.J.); (G.G.G.)
| | - Christopher F. Evans
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, VIC 3051, Australia; (S.L.); (T.L.N.); (J.G.D.); (G.S.D.); (Z.Y.); (H.N.T.); (C.F.E.); (M.A.J.); (G.G.G.)
| | - Maxine Tan
- Electrical and Computer Systems Engineering Discipline, School of Engineering, Monash University Malaysia, Bandar Sunway 47500, Malaysia;
- School of Electrical and Computer Engineering, The University of Oklahoma, Norman, OK 73019, USA
| | - Joohon Sung
- Department of Public Health Sciences, Division of Genome and Health Big Data, Graduate School of Public Health, Seoul National University, Seoul 08826, Korea;
| | - Mark A. Jenkins
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, VIC 3051, Australia; (S.L.); (T.L.N.); (J.G.D.); (G.S.D.); (Z.Y.); (H.N.T.); (C.F.E.); (M.A.J.); (G.G.G.)
| | - Graham G. Giles
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, VIC 3051, Australia; (S.L.); (T.L.N.); (J.G.D.); (G.S.D.); (Z.Y.); (H.N.T.); (C.F.E.); (M.A.J.); (G.G.G.)
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, VIC 3168, Australia; (T.N.-D.); (M.C.S.)
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, VIC 3004, Australia
| | - John L. Hopper
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, VIC 3051, Australia; (S.L.); (T.L.N.); (J.G.D.); (G.S.D.); (Z.Y.); (H.N.T.); (C.F.E.); (M.A.J.); (G.G.G.)
| | - Melissa C. Southey
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, VIC 3168, Australia; (T.N.-D.); (M.C.S.)
- Department of Clinical Pathology, The University of Melbourne, Parkville, VIC 3051, Australia
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, VIC 3004, Australia
| |
Collapse
|
6
|
Reye G, Huang X, Britt KL, Meinert C, Blick T, Xu Y, Momot KI, Lloyd T, Northey JJ, Thompson EW, Hugo HJ. RASSF1A Suppression as a Potential Regulator of Mechano-Pathobiology Associated with Mammographic Density in BRCA Mutation Carriers. Cancers (Basel) 2021; 13:cancers13133251. [PMID: 34209669 PMCID: PMC8269117 DOI: 10.3390/cancers13133251] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/23/2021] [Accepted: 06/26/2021] [Indexed: 01/26/2023] Open
Abstract
High mammographic density (MD) increases breast cancer (BC) risk and creates a stiff tissue environment. BC risk is also increased in BRCA1/2 gene mutation carriers, which may be in part due to genetic disruption of the tumour suppressor gene Ras association domain family member 1 (RASSF1A), a gene that is also directly regulated by tissue stiffness. High MD combined with BRCA1/2 mutations further increase breast cancer risk, yet BRCA1/2 mutations alone or in combination do not increase MD. The molecular basis for this additive effect therefore remains unclear. We studied the interplay between MD, stiffness, and BRCA1/2 mutation status in human mammary tissue obtained after prophylactic mastectomy from women at risk of developing BC. Our results demonstrate that RASSF1A expression increased in MCF10DCIS.com cell cultures with matrix stiffness up until ranges corresponding with BiRADs 4 stiffnesses (~16 kPa), but decreased in higher stiffnesses approaching malignancy levels (>50 kPa). Similarly, higher RASSF1A protein was seen in these cells when co-cultivated with high MD tissue in murine biochambers. Conversely, local stiffness, as measured by collagen I versus III abundance, repressed RASSF1A protein expression in BRCA1, but not BRCA2 gene mutated tissues; regional density as measured radiographically repressed RASSF1A in both BRCA1/2 mutated tissues. The combinatory effect of high MD and BRCA mutations on breast cancer risk may be due to RASSF1A gene repression in regions of increased tissue stiffness.
Collapse
Affiliation(s)
- Gina Reye
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD 4006, Australia; (G.R.); (X.H.); (T.B.); (E.W.T.)
- Translational Research Institute, Woolloongabba, QLD 4102, Australia
| | - Xuan Huang
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD 4006, Australia; (G.R.); (X.H.); (T.B.); (E.W.T.)
- Translational Research Institute, Woolloongabba, QLD 4102, Australia
| | - Kara L. Britt
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia;
| | - Christoph Meinert
- Herston Biofabrication Institute, Metro North Hospital and Health Service, Brisbane, QLD 4029, Australia;
- Gelomics Pty. Ltd., Brisbane, QLD 4059, Australia
| | - Tony Blick
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD 4006, Australia; (G.R.); (X.H.); (T.B.); (E.W.T.)
- Translational Research Institute, Woolloongabba, QLD 4102, Australia
| | - Yannan Xu
- Central Analytical Research Facility, Queensland University of Technology, Brisbane, QLD 4000, Australia;
| | - Konstantin I. Momot
- Faculty of Science, School of Chemistry and Physics, Queensland University of Technology, Brisbane, QLD 4000, Australia;
| | - Thomas Lloyd
- Department of Radiology, The Princess Alexandra Hospital, Woollongabba, QLD 4102, Australia;
| | - Jason J. Northey
- Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA;
| | - Erik W. Thompson
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD 4006, Australia; (G.R.); (X.H.); (T.B.); (E.W.T.)
- Translational Research Institute, Woolloongabba, QLD 4102, Australia
| | - Honor J. Hugo
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD 4006, Australia; (G.R.); (X.H.); (T.B.); (E.W.T.)
- Translational Research Institute, Woolloongabba, QLD 4102, Australia
- School of Health and Behavioural Sciences, University of the Sunshine Coast, Sippy Downs, QLD 4556, Australia
- School of Medicine and Dentistry, Griffith University, Birtinya, QLD 4575, Australia
- Correspondence:
| |
Collapse
|
7
|
Evans DG, van Veen EM, Howell A, Astley S. Heritability of mammographic breast density. Quant Imaging Med Surg 2020; 10:2387-2391. [PMID: 33269237 DOI: 10.21037/qims-2020-20] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- D Gareth Evans
- Clinical Genetics Service, Manchester Centre for Genomic Medicine, Manchester University Hospitals NHS Foundation Trust, Manchester, UK.,NW Genomic Laboratory Hub, Manchester Centre for Genomic Medicine, Manchester University Hospitals NHS Foundation Trust, Manchester, UK.,Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK.,Prevent Breast Cancer Centre, Wythenshawe Hospital Manchester Universities Foundation Trust, Wythenshawe, Manchester, UK.,Manchester Breast Centre, The Christie Hospital, Manchester, UK
| | - Elke M van Veen
- NW Genomic Laboratory Hub, Manchester Centre for Genomic Medicine, Manchester University Hospitals NHS Foundation Trust, Manchester, UK.,Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Anthony Howell
- Prevent Breast Cancer Centre, Wythenshawe Hospital Manchester Universities Foundation Trust, Wythenshawe, Manchester, UK.,Manchester Breast Centre, The Christie Hospital, Manchester, UK
| | - Susan Astley
- Prevent Breast Cancer Centre, Wythenshawe Hospital Manchester Universities Foundation Trust, Wythenshawe, Manchester, UK.,Manchester Breast Centre, The Christie Hospital, Manchester, UK.,Division of Informatics, Imaging and Data Science, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| |
Collapse
|
8
|
Cheasley D, Devereux L, Hughes S, Nickson C, Procopio P, Lee G, Li N, Pridmore V, Elder K, Bruce Mann G, Kader T, Rowley SM, Fox SB, Byrne D, Saunders H, Fujihara KM, Lim B, Gorringe KL, Campbell IG. The TP53 mutation rate differs in breast cancers that arise in women with high or low mammographic density. NPJ Breast Cancer 2020; 6:34. [PMID: 32802943 PMCID: PMC7414106 DOI: 10.1038/s41523-020-00176-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Accepted: 07/13/2020] [Indexed: 01/01/2023] Open
Abstract
Mammographic density (MD) influences breast cancer risk, but how this is mediated is unknown. Molecular differences between breast cancers arising in the context of the lowest and highest quintiles of mammographic density may identify the mechanism through which MD drives breast cancer development. Women diagnosed with invasive or in situ breast cancer where MD measurement was also available (n = 842) were identified from the Lifepool cohort of >54,000 women participating in population-based mammographic screening. This group included 142 carcinomas in the lowest quintile of MD and 119 carcinomas in the highest quintile. Clinico-pathological and family history information were recorded. Tumor DNA was collected where available (n = 56) and sequenced for breast cancer predisposition and driver gene mutations, including copy number alterations. Compared to carcinomas from low-MD breasts, those from high-MD breasts were significantly associated with a younger age at diagnosis and features associated with poor prognosis. Low- and high-MD carcinomas matched for grade, histological subtype, and hormone receptor status were compared for somatic genetic features. Low-MD carcinomas had a significantly increased frequency of TP53 mutations, higher homologous recombination deficiency, higher fraction of the genome altered, and more copy number gains on chromosome 1q and losses on 17p. While high-MD carcinomas showed enrichment of tumor-infiltrating lymphocytes in the stroma. The data demonstrate that when tumors were matched for confounding clinico-pathological features, a proportion in the lowest quintile of MD appear biologically distinct, reflective of microenvironment differences between the lowest and highest quintiles of MD.
Collapse
Affiliation(s)
- Dane Cheasley
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC Australia
| | - Lisa Devereux
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC Australia
- Lifepool, Peter MacCallum Cancer Centre, Melbourne, VIC Australia
| | - Siobhan Hughes
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC Australia
| | - Carolyn Nickson
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Melbourne, VIC Australia
- Cancer Research Division, Cancer Council NSW, Sydney, NSW Australia
- Sydney School of Public Health, University of Sydney, Sydney, NSW Australia
| | - Pietro Procopio
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Melbourne, VIC Australia
- Cancer Research Division, Cancer Council NSW, Sydney, NSW Australia
- Sydney School of Public Health, University of Sydney, Sydney, NSW Australia
| | - Grant Lee
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Melbourne, VIC Australia
| | - Na Li
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC Australia
| | | | - Kenneth Elder
- Department of Surgery, University of Melbourne, Melbourne, VIC Australia
- The Royal Melbourne and Royal Women’s Hospitals, Parkville, VIC Australia
- The Edinburgh Breast Unit, Western General Hospital, Edinburgh, UK
| | - G. Bruce Mann
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC Australia
- Department of Surgery, University of Melbourne, Melbourne, VIC Australia
- The Royal Melbourne and Royal Women’s Hospitals, Parkville, VIC Australia
| | - Tanjina Kader
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC Australia
| | - Simone M. Rowley
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC Australia
| | - Stephen B. Fox
- Department of Pathology, Peter MacCallum Cancer Centre, and University of Melbourne, Melbourne, VIC Australia
| | - David Byrne
- Department of Pathology, Peter MacCallum Cancer Centre, and University of Melbourne, Melbourne, VIC Australia
| | - Hugo Saunders
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC Australia
| | - Kenji M. Fujihara
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC Australia
| | - Belle Lim
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC Australia
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Kylie L. Gorringe
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC Australia
- Cancer Genetics and Genomics Program, Peter MacCallum Cancer Centre, Melbourne, VIC Australia
| | - Ian G. Campbell
- Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC Australia
| |
Collapse
|
9
|
Porembka JH, Ma J, Le-Petross HT. Breast density, MR imaging biomarkers, and breast cancer risk. Breast J 2020; 26:1535-1542. [PMID: 32654416 DOI: 10.1111/tbj.13965] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 01/03/2020] [Indexed: 11/29/2022]
Abstract
Mammographic breast density and various breast MRI features are imaging biomarkers that can predict a woman's future risk of breast cancer. While mammographic density (MD) has been established as an independent risk factor for the development of breast cancer, MD assessment methods need to be accurate and reproducible for widespread clinical use in stratifying patients based on their risk. In addition, a number of breast MRI biomarkers using contrast-enhanced and noncontrast-enhanced techniques are also being investigated as risk predictors. The validation and standardization of these breast MRI biomarkers will be necessary for population-based clinical implementation of patient risk stratification, as well. This review provides an update on MD assessment methods, breast MRI biomarkers, and their ability to predict breast cancer risk.
Collapse
Affiliation(s)
- Jessica H Porembka
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jingfei Ma
- Department of Imaging Physics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Huong T Le-Petross
- Diagnostic Imaging Division, Department of Breast Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
10
|
Skarping I, Förnvik D, Sartor H, Heide-Jørgensen U, Zackrisson S, Borgquist S. Mammographic density is a potential predictive marker of pathological response after neoadjuvant chemotherapy in breast cancer. BMC Cancer 2019; 19:1272. [PMID: 31888552 PMCID: PMC6937786 DOI: 10.1186/s12885-019-6485-4] [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: 06/27/2019] [Accepted: 12/20/2019] [Indexed: 12/12/2022] Open
Abstract
Background Our aim is to study if mammographic density (MD) prior to neoadjuvant chemotherapy is a predictive factor in accomplishing a pathological complete response (pCR) in neoadjuvant-treated breast cancer patients. Methods Data on all neoadjuvant treated breast cancer patients in Southern Sweden (2005–2016) were retrospectively identified, with patient and tumor characteristics retrieved from their medical charts. Diagnostic mammograms were used to evaluate and score MD as categorized by breast composition with the Breast Imaging-Reporting and Data System (BI-RADS) 5th edition. Logistic regression was used in complete cases to assess the odds ratios (OR) for pCR compared to BI-RADS categories (a vs b-d), adjusting for patient and pre-treatment tumor characteristics. Results A total of 302 patients were included in the study population, of which 57 (18.9%) patients accomplished pCR following neoadjuvant chemotherapy. The number of patients in the BI-RADS category a, b, c, and d were separately 16, 120, 140, and 26, respectively. In comparison to patients with BI-RADS breast composition a, patients with denser breasts had a lower OR of accomplishing pCR: BI-RADS b 0.32 (95%CI 0.07–0.1.5), BI-RADS c 0.30 (95%CI 0.06–1.45), and BI-RADS d 0.06 (95%CI 0.01–0.56). These associations were measured with lower point estimates, but wider confidence interval, in premenopausal patients; OR of accomplishing pCR for BI-RADS d in comparison to BI-RADS a: 0.03 (95%CI 0.00–0.76). Conclusions The likelihood of accomplishing pCR is indicated to be lower in breast cancer patients with higher MD, which need to be analysed in future studies for improved clinical decision-making regarding neoadjuvant treatment.
Collapse
Affiliation(s)
- Ida Skarping
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Skåne University Hospital, Lund, Sweden.
| | - Daniel Förnvik
- Medical Radiation Physics, Department of Translational Medicine, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Hanna Sartor
- Diagnostic Radiology, Department of Translational Medicine, Lund University, Skåne University Hospital, Lund and Malmö, Sweden
| | - Uffe Heide-Jørgensen
- Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus, Denmark
| | - Sophia Zackrisson
- Diagnostic Radiology, Department of Translational Medicine, Lund University, Skåne University Hospital, Lund and Malmö, Sweden
| | - Signe Borgquist
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Skåne University Hospital, Lund, Sweden.,Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
11
|
Davaadelger B, Choi MR, Singhal H, Clare SE, Khan SA, Kim JJ. BRCA1 mutation influences progesterone response in human benign mammary organoids. Breast Cancer Res 2019; 21:124. [PMID: 31771627 PMCID: PMC6878650 DOI: 10.1186/s13058-019-1214-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Accepted: 10/15/2019] [Indexed: 01/12/2023] Open
Abstract
Background Women, who carry a germline BRCA1 gene mutation, have a markedly increased risk of developing breast cancer during their lifetime. While BRCA1 carriers frequently develop triple-negative, basal-like, aggressive breast tumors, hormone signaling is important in the genesis of BRCA1 mutant breast cancers. We investigated the hormone response in BRCA1-mutated benign breast tissue using an in vitro organoid system. Methods Scaffold-free, multicellular human breast organoids generated from benign breast tissues from non-carrier or BRCA1 mutation carriers were treated in vitro with a stepwise menstrual cycle hormone regimen of estradiol (E2) and progesterone (P4) over the course of 28 days. Results Breast organoids exhibited characteristics of the native breast tissue, including expression of hormone receptors, collagen production, and markers of luminal and basal epithelium, and stromal fibroblasts. RNA sequencing analysis revealed distinct gene expression in response to hormone treatment in the non-carrier and BRCA1-mutated organoids. The selective progesterone receptor modulator, telapristone acetate (TPA), was used to identify specifically PR regulated genes. Specifically, extracellular matrix organization genes were regulated by E2+P4+TPA in the BRCA1-mutated organoids but not in the non-carrier organoids. In contrast, in the non-carrier organoids, known PR target genes such as the cell cycle genes were inhibited by TPA. Conclusions These data show that BRCA1 mutation influences hormone response and in particular PR activity which differs from that of non-carrier organoids. Our organoid model system revealed important insights into the role of PR in BRCA1-mutated benign breast cells and the critical paracrine actions that modify hormone receptor (HR)-negative cells. Further analysis of the molecular mechanism of BRCA1 and PR crosstalk is warranted using this model system.
Collapse
Affiliation(s)
- Batzaya Davaadelger
- Division of Reproductive Science in Medicine, Department of Obstetrics and Gynecology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, 4-117, Chicago, IL, 60611, USA
| | - Mi-Ran Choi
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Hari Singhal
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Susan E Clare
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Seema A Khan
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - J Julie Kim
- Division of Reproductive Science in Medicine, Department of Obstetrics and Gynecology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, 4-117, Chicago, IL, 60611, USA.
| |
Collapse
|
12
|
A review of the influence of mammographic density on breast cancer clinical and pathological phenotype. Breast Cancer Res Treat 2019; 177:251-276. [PMID: 31177342 DOI: 10.1007/s10549-019-05300-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 05/27/2019] [Indexed: 10/26/2022]
Abstract
PURPOSE It is well established that high mammographic density (MD), when adjusted for age and body mass index, is one of the strongest known risk factors for breast cancer (BC), and also associates with higher incidence of interval cancers in screening due to the masking of early mammographic abnormalities. Increasing research is being undertaken to determine the underlying histological and biochemical determinants of MD and their consequences for BC pathogenesis, anticipating that improved mechanistic insights may lead to novel preventative or treatment interventions. At the same time, technological advances in digital and contrast mammography are such that the validity of well-established relationships needs to be re-examined in this context. METHODS With attention to old versus new technologies, we conducted a literature review to summarise the relationships between clinicopathologic features of BC and the density of the surrounding breast tissue on mammography, including the associations with BC biological features inclusive of subtype, and implications for the clinical disease course encompassing relapse, progression, treatment response and survival. RESULTS AND CONCLUSIONS There is reasonable evidence to support positive relationships between high MD (HMD) and tumour size, lymph node positivity and local relapse in the absence of radiotherapy, but not between HMD and LVI, regional relapse or distant metastasis. Conflicting data exist for associations of HMD with tumour location, grade, intrinsic subtype, receptor status, second primary incidence and survival, which need further confirmatory studies. We did not identify any relationships that did not hold up when data involving newer imaging techniques were employed in analysis.
Collapse
|
13
|
Vreemann S, Dalmis MU, Bult P, Karssemeijer N, Broeders MJM, Gubern-Mérida A, Mann RM. Amount of fibroglandular tissue FGT and background parenchymal enhancement BPE in relation to breast cancer risk and false positives in a breast MRI screening program : A retrospective cohort study. Eur Radiol 2019; 29:4678-4690. [PMID: 30796568 PMCID: PMC6682856 DOI: 10.1007/s00330-019-06020-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 12/18/2018] [Accepted: 01/18/2019] [Indexed: 12/17/2022]
Abstract
Objectives The purpose of this study is to evaluate the predictive value of the amount of fibroglandular tissue (FGT) and background parenchymal enhancement (BPE), measured at baseline on breast MRI, for breast cancer development and risk of false-positive findings in women at increased risk for breast cancer. Methods Negative baseline MRI scans of 1533 women participating in a screening program for women at increased risk for breast cancer between January 1, 2003, and January 1, 2014, were selected. Automated tools based on deep learning were used to obtain quantitative measures of FGT and BPE. Logistic regression using forward selection was used to assess relationships between FGT, BPE, cancer detection, false-positive recall, and false-positive biopsy. Results Sixty cancers were detected in follow-up. FGT was only associated to short-term cancer risk; BPE was not associated with cancer risk. High FGT and BPE did lead to more false-positive recalls at baseline (OR 1.259, p = 0.050, and OR 1.475, p = 0.003) and to more frequent false-positive biopsies at baseline (OR 1.315, p = 0.049, and OR 1.807, p = 0.002), but were not predictive for false-positive findings in subsequent screening rounds. Conclusions FGT and BPE, measured on baseline MRI, are not predictive for overall breast cancer development in women at increased risk. High FGT and BPE lead to more false-positive findings at baseline. Key Points • Amount of fibroglandular tissue is only predictive for short-term breast cancer risk in women at increased risk. • Background parenchymal enhancement measured on baseline MRI is not predictive for breast cancer development in women at increased risk. • High amount of fibroglandular tissue and background parenchymal enhancement lead to more false-positive findings at baseline MRI. Electronic supplementary material The online version of this article (10.1007/s00330-019-06020-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Suzan Vreemann
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Geert Grooteplein 10, route 766, 6525 GA, Nijmegen, the Netherlands
| | - Mehmet U Dalmis
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Geert Grooteplein 10, route 766, 6525 GA, Nijmegen, the Netherlands
| | - Peter Bult
- Department of Pathology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Nico Karssemeijer
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Geert Grooteplein 10, route 766, 6525 GA, Nijmegen, the Netherlands
| | - Mireille J M Broeders
- Department for Health Evidence, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Albert Gubern-Mérida
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Geert Grooteplein 10, route 766, 6525 GA, Nijmegen, the Netherlands
| | - Ritse M Mann
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Geert Grooteplein 10, route 766, 6525 GA, Nijmegen, the Netherlands.
| |
Collapse
|
14
|
Grubstein A, Rapson Y, Benzaquen O, Rozenblatt S, Gadiel I, Atar E, Yerushalmi R, Cohen MJ. Comparison of background parenchymal enhancement and fibroglandular density at breast magnetic resonance imaging between BRCA gene mutation carriers and non-carriers. Clin Imaging 2018; 51:347-351. [PMID: 29982132 DOI: 10.1016/j.clinimag.2018.06.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 05/24/2018] [Accepted: 06/11/2018] [Indexed: 01/01/2023]
Abstract
OBJECTIVE High background parenchymal enhancement and amount of fibroglandular tissue on breast magnetic resonance imaging are related to increased breast cancer risk. This study sought to compare these parameters between BRCA mutation carriers and non-carriers and to evaluate the potential implications of the findings for short term follow-up. MATERIALS AND METHODS Magnetic resonance imaging studies of known BRCA mutation carriers, were compared to age-matched non-carrier studies performed in the same center during the same period. The groups were compared for qualitative background parenchymal enhancement and amount of fibroglandular tissue using the Breast Imaging Reporting and Data System (BI-RADS). RESULTS Breast parenchymal enhancement was high in up to one-third of the cohort: 22% of carriers and 33% of controls (p = 0.013). These results were sustained on separate analysis of menstrual-cycle-timed examinations. Amount of fibroglandular tissue was high in most cases: 62% of carriers and 75% of controls (p = 0.004). A BI-RADS final assessment score of 3 was more common in patients with high parenchymal enhancement, especially controls. CONCLUSION BRCA mutation carriers demonstrated lower levels of breast parenchymal enhancement and amount of fibroglandular tissue than age-matched non-carriers. These differences are probably influenced by hormonal status, as well as highlight different risks in distinctive subgroups of breast cancer (hormone-enriched, mutation-associated defective DNA damage repair), affecting considerations of preventive medical treatment. Differences in the indications for imaging between the carrier and non-carrier groups (screening for mutations and breast cancer evaluation, respectively) probably accounted for the higher rate of BI-RADS 3 in the control group.
Collapse
Affiliation(s)
- Ahuva Grubstein
- Department of Imaging, Rabin Medical Center - Beilinson Hospital, Petach Tikva 4941492, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel.
| | - Yael Rapson
- Department of Imaging, Rabin Medical Center - Beilinson Hospital, Petach Tikva 4941492, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel.
| | - Oshra Benzaquen
- Department of Imaging, Rabin Medical Center - Beilinson Hospital, Petach Tikva 4941492, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel.
| | - Shira Rozenblatt
- Department of Imaging, Rabin Medical Center - Beilinson Hospital, Petach Tikva 4941492, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel.
| | - Itay Gadiel
- Department of Imaging, Rabin Medical Center - Beilinson Hospital, Petach Tikva 4941492, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel.
| | - Eli Atar
- Department of Imaging, Rabin Medical Center - Beilinson Hospital, Petach Tikva 4941492, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel.
| | - Rinat Yerushalmi
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel; Institute of Oncology, Davidoff Cancer Center, Rabin Medical Center, Petach Tikva 4941492, Israel.
| | | |
Collapse
|
15
|
Evans DG, Howell SJ, Howell A. Personalized prevention in high risk individuals: Managing hormones and beyond. Breast 2018; 39:139-147. [PMID: 29610032 DOI: 10.1016/j.breast.2018.03.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 03/17/2018] [Accepted: 03/24/2018] [Indexed: 12/01/2022] Open
Abstract
Increasing numbers of women are being identified at 'high-risk' of breast cancer, defined by The National Institute of Health and Care Excellence (NICE) as a 10-year risk of ≥8%. Classically women have been so identified through family history based risk algorithms or genetic testing of high-risk genes. Recent research has shown that assessment of mammographic density and single nucleotide polymorphisms (SNPs), when combined with established risk factors, trebles the number of women reaching the high risk threshold. The options for risk reduction in such women include endocrine chemoprevention with the selective estrogen receptor modulators tamoxifen and raloxifene or the aromatase inhibitors anastrozole or exemestane. NICE recommends offering anastrozole to postmenopausal women at high-risk of breast cancer as cost effectiveness analysis showed this to be cost saving to the National Health Service. Overall uptake to chemoprevention has been disappointingly low but this may improve with the improved efficacy of aromatase inhibitors, particularly the lack of toxicity to the endometrium and thrombogenic risks. Novel approaches to chemoprevention under investigation include lower dose and topical tamoxifen, denosumab, anti-progestins and metformin. Although oophorectomy is usually only recommended to women at increased risk of ovarian cancer it has been shown in numerous studies to reduce breast cancer risks in the general population and in those with mutations in BRCA1/2. However, recent evidence from studies that have confined analysis to true prospective follow up have cast doubt on the efficacy of oophorectomy to reduce breast cancer risk in BRCA1 mutation carriers, at least in the short-term.
Collapse
Affiliation(s)
- D Gareth Evans
- Manchester Centre for Genomic Medicine, Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK; Prevent Breast Cancer and Nightingale Breast Screening Centre, Wythenshawe Hospital Manchester Universities Foundation Trust, Manchester, UK; Manchester Academic Health Science Centre, University of Manchester, Manchester, UK; The Christie NHS Foundation Trust, Manchester, UK; Manchester Centre for Genomic Medicine, Central Manchester University Hospitals NHS Foundation Trust, Manchester, UK; Manchester Breast Centre, Manchester Cancer Research Centre, University of Manchester, Manchester, UK.
| | - Sacha J Howell
- Prevent Breast Cancer and Nightingale Breast Screening Centre, Wythenshawe Hospital Manchester Universities Foundation Trust, Manchester, UK; Manchester Academic Health Science Centre, University of Manchester, Manchester, UK; The Christie NHS Foundation Trust, Manchester, UK; Manchester Breast Centre, Manchester Cancer Research Centre, University of Manchester, Manchester, UK
| | - Anthony Howell
- Prevent Breast Cancer and Nightingale Breast Screening Centre, Wythenshawe Hospital Manchester Universities Foundation Trust, Manchester, UK; Manchester Academic Health Science Centre, University of Manchester, Manchester, UK; The Christie NHS Foundation Trust, Manchester, UK; Manchester Breast Centre, Manchester Cancer Research Centre, University of Manchester, Manchester, UK
| |
Collapse
|
16
|
McLean KE, Stone J. Role of breast density measurement in screening for breast cancer. Climacteric 2018; 21:214-220. [DOI: 10.1080/13697137.2018.1424816] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- K. E. McLean
- Centre for Genetic Origins of Health and Disease, Curtin University and The University of Western Australia, Perth, WA, Australia
| | - J. Stone
- Centre for Genetic Origins of Health and Disease, Curtin University and The University of Western Australia, Perth, WA, Australia
| |
Collapse
|
17
|
Lecler A, Dunant A, Delaloge S, Wehrer D, Moussa T, Caron O, Balleyguier C. Breast tissue density change after oophorectomy in BRCA mutation carrier patients using visual and volumetric analysis. Br J Radiol 2018; 91:20170163. [PMID: 29182397 DOI: 10.1259/bjr.20170163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVE BRCA1/2 mutations account for 30-50% of hereditary breast cancers and bilateral oophorectomy is associated with a reduced risk of breast cancer in these patients. Breast density is a well-established breast cancer risk factor and is also associated with increased risk in BRCA carriers. The aim of the study was to evaluate the impact of oophorectomy on mammographic breast density and to assess which method of breast density assessment is more sensitive to change over time. METHODS Retrospective study of 50 BRCA1/2 patients who underwent bilateral oophorectomy and had at least a baseline and post-surgery mammogram. Mammographic breast density was determined by Volpara and consensus visual assessment by two radiologists. The primary endpoint was change in density between baseline and the first mammogram post-surgery. RESULTS At baseline, there was a non-significant trend for decreased density with increasing age. Volumetric breast density (VBD) significantly decreased after oophorectomy from a median VBD of 12.5% at baseline to 10.2% post-surgery which was driven by a reduction in fibroglandular volume. There was a higher absolute decrease in VBD in patients aged between 40-50 (p < 0.01). Using Volpara Density Grades (analogous to BI-RADS 4th edition density categories), 84% of females displayed a decrease in density category over the study period compared to only 76% using the radiologists' visual classification (p < 0.001) Conclusion: Oophorectomy is associated with a decrease in breast density and younger patients exhibit a larger absolute decrease. Volpara is more sensitive to identify change over time compared to visual assessment. Advances in knowledge: Oophorectomy is associated with a significant decrease in VBD in patients with BRCA mutations and Volpara Density Grades were more sensitive to identify decreases in density compared to visually assessed BI-RADS categories. Decreases in breast density following oophorectomy surgery in BRCA patients may be one of the mechanisms contributing to the observed decreased breast cancer risk after surgery. However, further studies are needed to investigate the relationship between breast density, oophorectomy and breast cancer risk in BRCA patients.
Collapse
Affiliation(s)
- Augustin Lecler
- 1 Department of Radiology, Fondation Ophtalmologique Rothschild , Fondation Ophtalmologique Rothschild , Paris , France.,2 Department of Radiology, Gustave Roussy , Gustave Roussy , Villejuif , France
| | - Ariane Dunant
- 3 Department of Biostatistic and Epidemiology, Gustave Roussy , Gustave Roussy , Villejuif , France
| | - Suzette Delaloge
- 4 Department of Medical Oncology, Gustave Roussy , Gustave Roussy , Villejuif , France
| | - Delphine Wehrer
- 5 Department of Genetics, Gustave Roussy , Gustave Roussy , Villejuif , France
| | - Tania Moussa
- 2 Department of Radiology, Gustave Roussy , Gustave Roussy , Villejuif , France
| | - Olivier Caron
- 5 Department of Genetics, Gustave Roussy , Gustave Roussy , Villejuif , France
| | - Corinne Balleyguier
- 2 Department of Radiology, Gustave Roussy , Gustave Roussy , Villejuif , France.,6 Department of Radiology, University Paris-Sud , University Paris-Sud , Orsay , France
| |
Collapse
|
18
|
Ironside AJ, Jones JL. Stromal characteristics may hold the key to mammographic density: the evidence to date. Oncotarget 2017; 7:31550-62. [PMID: 26784251 PMCID: PMC5058777 DOI: 10.18632/oncotarget.6912] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 01/02/2016] [Indexed: 12/11/2022] Open
Abstract
There is strong epidemiological data indicating a role for increased mammographic density (MD) in predisposing to breast cancer, however, the biological mechanisms underlying this phenomenon are less well understood. Recently, studies of human breast tissues have started to characterise the features of mammographically dense breasts, and a number of in-vitro and in-vivo studies have explored the potential mechanisms through which dense breast tissue may exert this tumourigenic risk. This article aims to review both the pathological and biological evidence implicating a key role for the breast stromal compartment in MD, how this may be modified and the clinical significance of these findings. The epidemiological context will be briefly discussed but will not be covered in detail.
Collapse
Affiliation(s)
- Alastair J Ironside
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - J Louise Jones
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
| |
Collapse
|
19
|
Toriola AT, Dang HX, Hagemann IS, Appleton CM, Colditz GA, Luo J, Maher CA. Increased breast tissue receptor activator of nuclear factor-κB ligand (RANKL) gene expression is associated with higher mammographic density in premenopausal women. Oncotarget 2017; 8:73787-73792. [PMID: 29088745 PMCID: PMC5650300 DOI: 10.18632/oncotarget.17909] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 05/01/2017] [Indexed: 11/25/2022] Open
Abstract
Increased mammographic breast density is associated with a 4-6-fold increased risk of breast cancer, yet lifestyle factors that can reduce dense breasts are yet to be identified, and viable prevention strategies to reduce breast density-associated breast cancer development are yet to be developed. We investigated the associations of breast tissue receptor activator of nuclear factor-κB (RANK) pathway gene expression with mammographic density in 48 premenopausal women, with no previous history of cancer. Gene expression levels were measured in total RNA isolated from formalin-fixed paraffin-embedded breast tissue samples, using the NanoString nCounter platform. Mammographic density was classified based on the American College of Radiology Breast Imaging Reporting and Data (BI-RADS). Linear regression was used to evaluate associations between gene expression and mammographic density. The mean age of participants was 44.4 years. Women with higher breast tissue RANKL (TNFSF11) (p-value = 0.0076), and TNF (p-value = 0.007) gene expression had higher mammographic density. Our finding provides mechanistic support for a breast cancer chemoprevention trial with a RANKL inhibitor among high-risk premenopausal women with dense breasts.
Collapse
Affiliation(s)
- Adetunji T. Toriola
- Department of Surgery, Division of Public Health Sciences, and Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Ha X. Dang
- The McDonnell Genome Institute, and Department of Medicine, Washington University, St. Louis, MO, USA
| | - Ian S. Hagemann
- Genomics and Pathology Services, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Catherine M. Appleton
- Division of Diagnostic Radiology, Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Graham A. Colditz
- Department of Surgery, Division of Public Health Sciences, and Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Jingqin Luo
- Department of Surgery, Division of Public Health Sciences, and Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Christopher A. Maher
- The McDonnell Genome Institute, and Department of Medicine, Washington University, St. Louis, MO, USA
| |
Collapse
|
20
|
Milne RL, Antoniou AC. Modifiers of breast and ovarian cancer risks for BRCA1 and BRCA2 mutation carriers. Endocr Relat Cancer 2016; 23:T69-84. [PMID: 27528622 DOI: 10.1530/erc-16-0277] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 08/15/2016] [Indexed: 12/20/2022]
Abstract
Pathogenic mutations in BRCA1 and BRCA2 are associated with high risks of breast and ovarian cancer. However, penetrance estimates for mutation carriers have been found to vary substantially between studies, and the observed differences in risk are consistent with the hypothesis that genetic and environmental factors modify cancer risks for women with these mutations. Direct evidence that this is the case has emerged in the past decade, through large-scale international collaborative efforts. Here, we describe the methodological challenges in the identification and characterisation of these risk-modifying factors, review the latest evidence on genetic and lifestyle/hormonal risk factors that modify breast and ovarian cancer risks for women with BRCA1 and BRCA2 mutations and outline the implications of these findings for cancer risk prediction. We also review the unresolved issues in this area of research and identify strategies of clinical implementation so that women with BRCA1 and BRCA2 mutations are no longer counselled on the basis of 'average' risk estimates.
Collapse
Affiliation(s)
- Roger L Milne
- Cancer Epidemiology CentreCancer Council Victoria, Melbourne, Australia Centre for Epidemiology and BiostatisticsMelbourne School of Population and Global Health, University of Melbourne, Parkville, Victoria, Australia
| | - Antonis C Antoniou
- Centre for Cancer Genetic EpidemiologyDepartment of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| |
Collapse
|
21
|
Gastounioti A, Conant EF, Kontos D. Beyond breast density: a review on the advancing role of parenchymal texture analysis in breast cancer risk assessment. Breast Cancer Res 2016; 18:91. [PMID: 27645219 PMCID: PMC5029019 DOI: 10.1186/s13058-016-0755-8] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND The assessment of a woman's risk for developing breast cancer has become increasingly important for establishing personalized screening recommendations and forming preventive strategies. Studies have consistently shown a strong relationship between breast cancer risk and mammographic parenchymal patterns, typically assessed by percent mammographic density. This paper will review the advancing role of mammographic texture analysis as a potential novel approach to characterize the breast parenchymal tissue to augment conventional density assessment in breast cancer risk estimation. MAIN TEXT The analysis of mammographic texture provides refined, localized descriptors of parenchymal tissue complexity. Currently, there is growing evidence in support of textural features having the potential to augment the typically dichotomized descriptors (dense or not dense) of area or volumetric measures of breast density in breast cancer risk assessment. Therefore, a substantial research effort has been devoted to automate mammographic texture analysis, with the aim of ultimately incorporating such quantitative measures into breast cancer risk assessment models. In this paper, we review current and emerging approaches in this field, summarizing key methodological details and related studies using novel computerized approaches. We also discuss research challenges for advancing the role of parenchymal texture analysis in breast cancer risk stratification and accelerating its clinical translation. CONCLUSIONS The objective is to provide a comprehensive reference for researchers in the field of parenchymal pattern analysis in breast cancer risk assessment, while indicating key directions for future research.
Collapse
Affiliation(s)
- Aimilia Gastounioti
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Emily F Conant
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Despina Kontos
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
22
|
Evans DG, Astley S, Stavrinos P, Harkness E, Donnelly LS, Dawe S, Jacob I, Harvie M, Cuzick J, Brentnall A, Wilson M, Harrison F, Payne K, Howell A. Improvement in risk prediction, early detection and prevention of breast cancer in the NHS Breast Screening Programme and family history clinics: a dual cohort study. PROGRAMME GRANTS FOR APPLIED RESEARCH 2016. [DOI: 10.3310/pgfar04110] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BackgroundIn the UK, women are invited for 3-yearly mammography screening, through the NHS Breast Screening Programme (NHSBSP), from the ages of 47–50 years to the ages of 69–73 years. Women with family histories of breast cancer can, from the age of 40 years, obtain enhanced surveillance and, in exceptionally high-risk cases, magnetic resonance imaging. However, no NHSBSP risk assessment is undertaken. Risk prediction models are able to categorise women by risk using known risk factors, although accurate individual risk prediction remains elusive. The identification of mammographic breast density (MD) and common genetic risk variants [single nucleotide polymorphisms (SNPs)] has presaged the improved precision of risk models.ObjectivesTo (1) identify the best performing model to assess breast cancer risk in family history clinic (FHC) and population settings; (2) use information from MD/SNPs to improve risk prediction; (3) assess the acceptability and feasibility of offering risk assessment in the NHSBSP; and (4) identify the incremental costs and benefits of risk stratified screening in a preliminary cost-effectiveness analysis.DesignTwo cohort studies assessing breast cancer incidence.SettingHigh-risk FHC and the NHSBSP Greater Manchester, UK.ParticipantsA total of 10,000 women aged 20–79 years [Family History Risk Study (FH-Risk); UK Clinical Research Network identification number (UKCRN-ID) 8611] and 53,000 women from the NHSBSP [aged 46–73 years; Predicting the Risk of Cancer At Screening (PROCAS) study; UKCRN-ID 8080].InterventionsQuestionnaires collected standard risk information, and mammograms were assessed for breast density by a number of techniques. All FH-Risk and 10,000 PROCAS participants participated in deoxyribonucleic acid (DNA) studies. The risk prediction models Manual method, Tyrer–Cuzick (TC), BOADICEA (Breast and Ovarian Analysis of Disease Incidence and Carrier Estimation Algorithm) and Gail were used to assess risk, with modelling based on MD and SNPs. A preliminary model-based cost-effectiveness analysis of risk stratified screening was conducted.Main outcome measuresBreast cancer incidence.Data sourcesThe NHSBSP; cancer registration.ResultsA total of 446 women developed incident breast cancers in FH-Risk in 97,958 years of follow-up. All risk models accurately stratified women into risk categories. TC had better risk precision than Gail, and BOADICEA accurately predicted risk in the 6268 single probands. The Manual model was also accurate in the whole cohort. In PROCAS, TC had better risk precision than Gail [area under the curve (AUC) 0.58 vs. 0.54], identifying 547 prospective breast cancers. The addition of SNPs in the FH-Risk case–control study improved risk precision but was not useful inBRCA1(breast cancer 1 gene) families. Risk modelling of SNPs in PROCAS showed an incremental improvement from using SNP18 used in PROCAS to SNP67. MD measured by visual assessment score provided better risk stratification than automatic measures, despite wide intra- and inter-reader variability. Using a MD-adjusted TC model in PROCAS improved risk stratification (AUC = 0.6) and identified significantly higher rates (4.7 per 10,000 vs. 1.3 per 10,000;p < 0.001) of high-stage cancers in women with above-average breast cancer risks. It is not possible to provide estimates of the incremental costs and benefits of risk stratified screening because of lack of data inputs for key parameters in the model-based cost-effectiveness analysis.ConclusionsRisk precision can be improved by using DNA and MD, and can potentially be used to stratify NHSBSP screening. It may also identify those at greater risk of high-stage cancers for enhanced screening. The cost-effectiveness of risk stratified screening is currently associated with extensive uncertainty. Additional research is needed to identify data needed for key inputs into model-based cost-effectiveness analyses to identify the impact on health-care resource use and patient benefits.Future workA pilot of real-time NHSBSP risk prediction to identify women for chemoprevention and enhanced screening is required.FundingThe National Institute for Health Research Programme Grants for Applied Research programme. The DNA saliva collection for SNP analysis for PROCAS was funded by the Genesis Breast Cancer Prevention Appeal.
Collapse
Affiliation(s)
- D Gareth Evans
- Department of Genomic Medicine, Institute of Human Development, Manchester Academic Health Science Centre (MAHSC), Central Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| | - Susan Astley
- Institute of Population Health, Centre for Imaging Sciences, University of Manchester, Manchester, UK
| | - Paula Stavrinos
- The Nightingale Centre and Genesis Prevention Centre, University Hospital of South Manchester, Manchester, UK
| | - Elaine Harkness
- Institute of Population Health, Centre for Imaging Sciences, University of Manchester, Manchester, UK
| | - Louise S Donnelly
- The Nightingale Centre and Genesis Prevention Centre, University Hospital of South Manchester, Manchester, UK
| | - Sarah Dawe
- The Nightingale Centre and Genesis Prevention Centre, University Hospital of South Manchester, Manchester, UK
| | - Ian Jacob
- Department of Health Economics, University of Manchester, Manchester, UK
| | - Michelle Harvie
- The Nightingale Centre and Genesis Prevention Centre, University Hospital of South Manchester, Manchester, UK
| | - Jack Cuzick
- Wolfson Institute of Preventive Medicine, Queen Mary University of London, London, UK
| | - Adam Brentnall
- Wolfson Institute of Preventive Medicine, Queen Mary University of London, London, UK
| | - Mary Wilson
- The Nightingale Centre and Genesis Prevention Centre, University Hospital of South Manchester, Manchester, UK
| | | | - Katherine Payne
- Department of Health Economics, University of Manchester, Manchester, UK
| | - Anthony Howell
- Institute of Population Health, Centre for Imaging Sciences, University of Manchester, Manchester, UK
- The Nightingale Centre and Genesis Prevention Centre, University Hospital of South Manchester, Manchester, UK
| |
Collapse
|
23
|
James PA, Sawyer S, Boyle S, Young MA, Kovalenko S, Doherty R, McKinley J, Alsop K, Beshay V, Harris M, Fox S, Lindeman GJ, Mitchell G. Large genomic rearrangements in the familial breast and ovarian cancer gene BRCA1 are associated with an increased frequency of high risk features. Fam Cancer 2016; 14:287-95. [PMID: 25678442 DOI: 10.1007/s10689-015-9785-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Large genomic rearrangements (LGRs) account for at least 10% of the mutations in BRCA1 and 5% of BRCA2 mutations in outbred hereditary breast and ovarian cancer (HBOC) families. Data from some series suggest LGRs represent particularly penetrant mutations. 1,034 index cases from HBOC families underwent comprehensive BRCA1 and BRCA2 mutation testing, including screening for LGRs. The personal and family history of 254 identified mutation carriers were compared based on mutation type. Thirty-six LGRs were detected; 32/122 (26%) BRCA1 and 4/132 (3%) BRCA2 mutations. High risk features (bilateral breast cancer, diagnosis <40 years, ovarian cancer, male breast cancer) were more commonly associated with an LGR than a non-LGR mutation (p = 0.008), In families with a BRCA1 LGR the mean age of breast cancer diagnosis was younger than in families with a non-LGR BRCA1 mutation (42.5 vs. 46.1 years, p = 0.007). Across the entire group of mutation positive families the number of relatives affected by breast or ovarian cancer was increased [LGR 3.7 vs. non- LGR 2.8 per family, p value (adjusted for genotype) = 0.047]. Excluding index cases, the odds ratio for breast cancer in BRCA1 families with an LGR was 1.42 (95% CI 1.24-1.63) and for ovarian cancer 1.66 (95% CI 1.10-2.49). The increased cancer risk was reflected in significantly higher risk assessments by mutation prediction tools. LGRs are associated with higher cancer risks. If validated, LGRs could be included in cancer risk prediction tools to improve personalised cancer risk prediction estimates and may guide cost-minimising mutation screening strategies in some healthcare settings.
Collapse
Affiliation(s)
- Paul A James
- Familial Cancer Centre, Peter MacCallum Cancer Centre, St Andrews Place, East Melbourne, VIC, 3002, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Ramón Y Cajal T, Chirivella I, Miranda J, Teule A, Izquierdo Á, Balmaña J, Sánchez-Heras AB, Llort G, Fisas D, Lope V, Hernández-Agudo E, Juan-Fita MJ, Tena I, Robles L, Guillén-Ponce C, Pérez-Segura P, Luque-Molina MS, Hernando-Polo S, Salinas M, Brunet J, Salas-Trejo MD, Barnadas A, Pollán M. Mammographic density and breast cancer in women from high risk families. Breast Cancer Res 2015; 17:93. [PMID: 26163143 PMCID: PMC4499171 DOI: 10.1186/s13058-015-0604-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 06/24/2015] [Indexed: 11/15/2022] Open
Abstract
Introduction Mammographic density (MD) is one of the strongest determinants of sporadic breast cancer (BC). In this study, we compared MD in BRCA1/2 mutation carriers and non-carriers from BRCA1/2 mutation-positive families and investigated the association between MD and BC among BRCA1/2 mutation carriers per type of mutation and tumor subtype. Methods The study was carried out in 1039 female members of BRCA1 and BRCA2 mutation-positive families followed at 16 Spanish Genetic Counseling Units. Participants’ density was scored retrospectively from available mammograms by a single blinded radiologist using a 5-category scale (<10 %, 10-25 %, 25-50 %, 50-75 %, >75 %). In BC cases, we selected mammograms taken prior to diagnosis or from the contralateral breast, whereas, in non-cases, the last screening mammogram was evaluated. MD distribution in carriers and non-carriers was compared using ordinal logistic models, and the association between MD and BC in BRCA1/2 mutation carriers was studied using logistic regression. Huber-White robust estimators of variance were used to take into account correlations between family members. A similar multinomial model was used to explore this association by BC subtype. Results We identified and scored mammograms from 341 BRCA1, 350 BRCA2 mutation carriers and 229 non-carriers. Compared to non-carriers, MD was significantly lower among BRCA2 mutation carriers (odds ratio (OR) =0.71; P-value=0.04), but not among BRCA1 carriers (OR=0.84; P-value=0.33). MD was associated with subsequent development BC (OR per category of MD=1.45; 95 % confidence interval=1.18-1.78, P-value<0.001), with no significant differences between BRCA1 and BRCA2 mutation carriers (P-value=0.48). Finally, no statistically significant differences were observed in the association of MD with specific BC subtypes. Conclusions Our study, the largest to date on this issue, confirms that MD is an independent risk factor for all BC subtypes in either BRCA1 and BRCA2 mutation carriers, and should be considered a phenotype risk marker in this context.
Collapse
Affiliation(s)
| | - Isabel Chirivella
- Medical Oncology Department, Hospital Clinico Universitario de Valencia, Valencia, Spain.
| | - Josefa Miranda
- Foundation General Directorate Public Health and Foundation for the Promotion of Health and Biomedical Research in the Valencian Region, FISABIO - Public Health, Valencia, Spain.
| | - Alexandre Teule
- Hereditary Cancer Program, Catalan Institue of Oncology-IDIBELL, Barcelona, Spain.
| | - Ángel Izquierdo
- Hereditary Cancer Program, Catalan Institute of Oncology-IDIBGI, Girona, Spain.
| | - Judith Balmaña
- Medical Oncology Deartment, Hospital Vall Hebron/Vall Hebron Institute of Oncology, Barcelona, Spain.
| | | | - Gemma Llort
- Genetic Counseling Unit, Corporació Sanitaria Parc tauli, Consorci Sanitari de Terrassa, Terrasa, Spain.
| | - David Fisas
- Medical Oncology Department, Hospital Santa Creu I Sant Pau, Barcelona, Spain.
| | - Virginia Lope
- National Center for Epidemiology, Carlos III Institute of Health, Monforte de Lemos 5, 28029, Madrid, Spain. .,Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Carlos III Institute of Health, Madrid, Spain. .,Consortium Cancer Epidemiology Research Group, Oncology and Hematology Area, IIS Puerta de Hierro (IDIPHIM), Madrid, Spain.
| | - Elena Hernández-Agudo
- Breast Cancer Unit, Clinical Research Programme, Spanish National Cancer Center (CNIO), Madrid, Spain.
| | - María José Juan-Fita
- Medical Oncology Department, Foundation of the Valencian Oncologic Institute, Valencia, Spain.
| | - Isabel Tena
- Medical Oncology Department, Hospital Provincial de Castellón, Castellón, Spain.
| | - Luis Robles
- Medical Oncology Department, Hospital 12 de Octubre, Madrid, Spain.
| | - Carmen Guillén-Ponce
- Medical Oncology Department, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, Spain.
| | - Pedro Pérez-Segura
- Medical Oncology Department, Hospital Clínico San Carlos, Madrid, Spain.
| | | | | | - Mónica Salinas
- Hereditary Cancer Program, Catalan Institue of Oncology-IDIBELL, Barcelona, Spain.
| | - Joan Brunet
- Hereditary Cancer Program, Catalan Institute of Oncology-IDIBGI, Girona, Spain. .,Medical Sciences Department, School of Medicine, University of Girona, Girona, Spain.
| | - María Dolores Salas-Trejo
- Foundation General Directorate Public Health and Foundation for the Promotion of Health and Biomedical Research in the Valencian Region, FISABIO - Public Health, Valencia, Spain.
| | - Agustí Barnadas
- Medical Oncology Department, Hospital Santa Creu I Sant Pau, Barcelona, Spain.
| | - Marina Pollán
- National Center for Epidemiology, Carlos III Institute of Health, Monforte de Lemos 5, 28029, Madrid, Spain. .,Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Carlos III Institute of Health, Madrid, Spain. .,Consortium Cancer Epidemiology Research Group, Oncology and Hematology Area, IIS Puerta de Hierro (IDIPHIM), Madrid, Spain.
| |
Collapse
|
25
|
Cho GY, Moy L, Kim SG, Klautau Leite AP, Baete SH, Babb JS, Sodickson DK, Sigmund EE. Comparison of contrast enhancement and diffusion-weighted magnetic resonance imaging in healthy and cancerous breast tissue. Eur J Radiol 2015. [PMID: 26220915 DOI: 10.1016/j.ejrad.2015.06.023] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
OBJECTIVE To measure background parenchymal enhancement (BPE) and compare with other contrast enhancement values and diffusion-weighted MRI parameters in healthy and cancerous breast tissue at the clinical level. MATERIALS AND METHODS This HIPAA-compliant, IRB approved retrospective study enrolled 77 patients (38 patients with breast cancer - mean age 51.8 ± 10.0 years; 39 high-risk patients for screening evaluation - mean age 46.3 ± 11.7 years), who underwent contrast-enhanced 3T breast MRI. Contrast enhanced MRI and diffusion-weighted imaging were performed to quantify BPE, lesion contrast enhancement, and apparent diffusion coefficient (ADC) metrics in fibroglandular tissue (FGT) and lesions. RESULTS BPE did not correlate with ADC values. Mean BPE for the lesion-bearing patients was higher (43.9%) compared to that of the high-risk screening patients (28.3%, p=0.004). Significant correlation (r=0.37, p<0.05) was found between BPE and lesion contrast enhancement. CONCLUSION No significant association was observed between parenchymal or lesion enhancement with conventional apparent diffusion metrics, suggesting that proliferative processes are not co-regulated in cancerous and parenchymal tissue.
Collapse
Affiliation(s)
- Gene Young Cho
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, New York, NY 10016, USA; Sackler Institute of Graduate Biomedical Sciences, New York University School of Medicine, New York, NY 10016, USA; Center for Advanced Imaging Innovation and Research (CAI(2)R), Department of Radiology, New York University School of Medicine, New York, NY 10016, USA.
| | - Linda Moy
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, New York, NY 10016, USA; Center for Advanced Imaging Innovation and Research (CAI(2)R), Department of Radiology, New York University School of Medicine, New York, NY 10016, USA; New York University Langone Medical Center - Cancer Institute, New York, NY 10016, USA
| | - Sungheon G Kim
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, New York, NY 10016, USA; Center for Advanced Imaging Innovation and Research (CAI(2)R), Department of Radiology, New York University School of Medicine, New York, NY 10016, USA
| | | | - Steven H Baete
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, New York, NY 10016, USA; Center for Advanced Imaging Innovation and Research (CAI(2)R), Department of Radiology, New York University School of Medicine, New York, NY 10016, USA
| | - James S Babb
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, New York, NY 10016, USA; Center for Advanced Imaging Innovation and Research (CAI(2)R), Department of Radiology, New York University School of Medicine, New York, NY 10016, USA
| | - Daniel K Sodickson
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, New York, NY 10016, USA; Center for Advanced Imaging Innovation and Research (CAI(2)R), Department of Radiology, New York University School of Medicine, New York, NY 10016, USA
| | - Eric E Sigmund
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, New York, NY 10016, USA; Center for Advanced Imaging Innovation and Research (CAI(2)R), Department of Radiology, New York University School of Medicine, New York, NY 10016, USA
| |
Collapse
|
26
|
Huo CW, Chew G, Hill P, Huang D, Ingman W, Hodson L, Brown KA, Magenau A, Allam AH, McGhee E, Timpson P, Henderson MA, Thompson EW, Britt K. High mammographic density is associated with an increase in stromal collagen and immune cells within the mammary epithelium. Breast Cancer Res 2015; 17:79. [PMID: 26040322 PMCID: PMC4485361 DOI: 10.1186/s13058-015-0592-1] [Citation(s) in RCA: 125] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 05/20/2015] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION Mammographic density (MD), after adjustment for a women's age and body mass index, is a strong and independent risk factor for breast cancer (BC). Although the BC risk attributable to increased MD is significant in healthy women, the biological basis of high mammographic density (HMD) causation and how it raises BC risk remain elusive. We assessed the histological and immunohistochemical differences between matched HMD and low mammographic density (LMD) breast tissues from healthy women to define which cell features may mediate the increased MD and MD-associated BC risk. METHODS Tissues were obtained between 2008 and 2013 from 41 women undergoing prophylactic mastectomy because of their high BC risk profile. Tissue slices resected from the mastectomy specimens were X-rayed, then HMD and LMD regions were dissected based on radiological appearance. The histological composition, aromatase immunoreactivity, hormone receptor status and proliferation status were assessed, as were collagen amount and orientation, epithelial subsets and immune cell status. RESULTS HMD tissue had a significantly greater proportion of stroma, collagen and epithelium, as well as less fat, than LMD tissue did. Second harmonic generation imaging demonstrated more organised stromal collagen in HMD tissues than in LMD tissues. There was significantly more aromatase immunoreactivity in both the stromal and glandular regions of HMD tissues than in those regions of LMD tissues, although no significant differences in levels of oestrogen receptor, progesterone receptor or Ki-67 expression were detected. The number of macrophages within the epithelium or stroma did not change; however, HMD stroma exhibited less CD206(+) alternatively activated macrophages. Epithelial cell maturation was not altered in HMD samples, and no evidence of epithelial-mesenchymal transition was seen; however, there was a significant increase in vimentin(+)/CD45(+) immune cells within the epithelial layer in HMD tissues. CONCLUSIONS We confirmed increased proportions of stroma and epithelium, increased aromatase activity and no changes in hormone receptor or Ki-67 marker status in HMD tissue. The HMD region showed increased collagen deposition and organisation as well as decreased alternatively activated macrophages in the stroma. The HMD epithelium may be a site for local inflammation, as we observed a significant increase in CD45(+)/vimentin(+) immune cells in this area.
Collapse
Affiliation(s)
- Cecilia W Huo
- University of Melbourne Department of Surgery, St. Vincent's Hospital, Level 2, Clinical Sciences Building, 29 Regent Street, Fitzroy, VIC, 3065, Australia.
| | - Grace Chew
- University of Melbourne Department of Surgery, St. Vincent's Hospital, Level 2, Clinical Sciences Building, 29 Regent Street, Fitzroy, VIC, 3065, Australia.
| | - Prue Hill
- Department of Pathology, St. Vincent's Hospital, 41 Victoria Parade, Fitzroy, VIC, 3065, Australia.
| | - Dexing Huang
- St. Vincent's Institute, 9 Princes Street, Fitzroy, VIC, 3065, Australia.
| | - Wendy Ingman
- Discipline of Surgery, Faculty of Health Sciences, School of Medicine, The Queen Elizabeth Hospital, University of Adelaide, Adelaide, Australia. .,Robinson Research Institute, University of Adelaide, Ground Floor, Norwich Centre, 55 King William Road, North Adelaide, SA, 5006, Australia.
| | - Leigh Hodson
- Discipline of Surgery, Faculty of Health Sciences, School of Medicine, The Queen Elizabeth Hospital, University of Adelaide, Adelaide, Australia. .,Robinson Research Institute, University of Adelaide, Ground Floor, Norwich Centre, 55 King William Road, North Adelaide, SA, 5006, Australia.
| | - Kristy A Brown
- Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, VIC, 3168, Australia.
| | - Astrid Magenau
- Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Australia. .,St Vincent's Clinical School, Faculty of Medicine, University of NSW, Sydney, Australia. .,St Vincent's Clinical School, Faculty of Medicine, University of NSW, Clayton, Australia.
| | - Amr H Allam
- Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Australia. .,St Vincent's Clinical School, Faculty of Medicine, University of NSW, Sydney, Australia. .,St Vincent's Clinical School, Faculty of Medicine, University of NSW, Clayton, Australia.
| | - Ewan McGhee
- St Vincent's Clinical School, Faculty of Medicine, University of NSW, Sydney, Australia.
| | - Paul Timpson
- Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Australia. .,St Vincent's Clinical School, Faculty of Medicine, University of NSW, Sydney, Australia. .,St Vincent's Clinical School, Faculty of Medicine, University of NSW, Clayton, Australia.
| | - Michael A Henderson
- University of Melbourne Department of Surgery, St. Vincent's Hospital, Level 2, Clinical Sciences Building, 29 Regent Street, Fitzroy, VIC, 3065, Australia. .,Peter MacCallum Cancer Centre, 2 St. Andrews Place, East Melbourne, VIC, 3002, Australia.
| | - Erik W Thompson
- University of Melbourne Department of Surgery, St. Vincent's Hospital, Level 2, Clinical Sciences Building, 29 Regent Street, Fitzroy, VIC, 3065, Australia. .,St. Vincent's Institute, 9 Princes Street, Fitzroy, VIC, 3065, Australia. .,Institute of Health and Biomedical Innovation and School of Biomedical Sciences, Queensland University of Technology, 60 Musk Avenue, Kelvin Grove, QLD, 4059, Australia.
| | - Kara Britt
- The Beatson Institute for Cancer Research, Switchback Road, Bearsden Glasgow, G61 1BD, UK. .,The Sir Peter MacCallum Department of Oncology, University of Melbourne, St. Andrews Place, East Melbourne, VIC, 3002, Australia. .,Department of Anatomy and Developmental Biology, Monash University, 19 Innovation Walk, Clayton, VIC, s, Australia.
| |
Collapse
|
27
|
Gur D, Klym AH, King JL, Bandos AI, Sumkin JH. Impact of the new density reporting laws: radiologist perceptions and actual behavior. Acad Radiol 2015; 22:679-83. [PMID: 25837723 DOI: 10.1016/j.acra.2015.02.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 01/09/2015] [Accepted: 02/03/2015] [Indexed: 11/17/2022]
Abstract
RATIONALE AND OBJECTIVES To assess radiologists' perceptions of how the new Breast Density Notification Act (BDNA) of Pennsylvania would affect their breast density reporting and their actual reporting patterns after implementation. MATERIALS AND METHODS Under an institutional review board-approved protocol, we surveyed 21 radiologists about how they believe the new law affected their breast density reporting patterns and analyzed actual changes for 16 respondents before and after the law took effect. Three hundred consecutive reports were assessed for each radiologist before and after the effective date. The distributions of reported density Breast Imaging Reporting and Data System (BI-RADS) (1-4) were compared using a type III test in the context of an ordinal mixed model accounting for between-reader variability and adjusting for age (PROC GLIMMIX, SAS, version 9.3) using a two-sided .05 significance level. RESULTS Seventeen radiologists responded to the survey; however, one retired shortly after responding. Of the 16 respondents, 56% (nine of 16) did not favor the law, 13% (two of 16) were in favor, and 31% (five of 16) were neutral. The fraction perceived that after implementation, they rated more, equally, or less frequently breasts as scattered fibroglandular densities (BI-RADS 2) versus heterogeneously dense rating (BI-RADS 3) was 50% (eight of 16), 44% (seven of 16), and 6% (one of 16), respectively. In practice, 44% (seven of 16) performed differently than their survey answers. Fourteen of 16 radiologists increased the frequency of reported BI-RADS 2 scores after BDNA implementation with seven having statistically significant (P < .05) increases after adjusting for age differences. CONCLUSIONS Radiologists' reporting patterns changed, at least for a short duration, after the new density reporting law and for some of the radiologists in an unexpected way.
Collapse
Affiliation(s)
- David Gur
- Department of Radiology, Imaging Research, University of Pittsburgh, 3362 Fifth Avenue, Pittsburgh, PA 15213.
| | - Amy H Klym
- Department of Radiology, Imaging Research, University of Pittsburgh, 3362 Fifth Avenue, Pittsburgh, PA 15213
| | - Jill L King
- Department of Radiology, Imaging Research, University of Pittsburgh, 3362 Fifth Avenue, Pittsburgh, PA 15213
| | - Andriy I Bandos
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jules H Sumkin
- Department of Radiology, Breast Imaging, Magee-Womens Hospital of UPMC, Pittsburgh, Pennsylvania
| |
Collapse
|
28
|
Mammographic density and breast cancer risk by family history in women of white and Asian ancestry. Cancer Causes Control 2015; 26:621-6. [PMID: 25761408 DOI: 10.1007/s10552-015-0551-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 03/04/2015] [Indexed: 10/23/2022]
Abstract
PURPOSE Mammographic density, i.e., the radiographic appearance of the breast, is a strong predictor of breast cancer risk. To determine whether the association of breast density with breast cancer is modified by a first-degree family history of breast cancer (FHBC) in women of white and Asian ancestry, we analyzed data from four case-control studies conducted in the USA and Japan. METHODS The study population included 1,699 breast cancer cases and 2,422 controls, of whom 45% reported white (N = 1,849) and 40% Asian (N = 1,633) ancestry. To standardize mammographic density assessment, a single observer re-read all mammograms using one type of interactive thresholding software. Logistic regression was applied to estimate odds ratios (OR) while adjusting for confounders. RESULTS Overall, 496 (12%) of participants reported a FHBC, which was significantly associated with breast cancer risk in the adjusted model (OR 1.51; 95% CI 1.23-1.84). There was a statistically significant interaction on a multiplicative scale between FHBC and continuous percent density (per 10 % density: p = 0.03). The OR per 10% increase in percent density was higher among women with a FHBC (OR 1.30; 95% CI 1.13-1.49) than among those without a FHBC (OR 1.14; 1.09-1.20). This pattern was apparent in whites and Asians. The respective ORs were 1.45 (95% CI 1.17-1.80) versus 1.22 (95% CI 1.14-1.32) in whites, whereas the values in Asians were only 1.24 (95% CI 0.97-1.58) versus 1.09 (95% CI 1.00-1.19). CONCLUSIONS These findings support the hypothesis that women with a FHBC appear to have a higher risk of breast cancer associated with percent mammographic density than women without a FHBC.
Collapse
|
29
|
Leclerc M, Antoniou AC, Simard J, Lakhal-Chaieb L. Analysis of multivariate failure times in the presence of selection bias with application to breast cancer. J R Stat Soc Ser C Appl Stat 2014. [DOI: 10.1111/rssc.12091] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
| | | | - Jacques Simard
- Centre Hospitalier Universitaire de Québec Research Center and Laval University; Québec Canada
| | | | | | | | | |
Collapse
|
30
|
Assi V, Massat NJ, Thomas S, MacKay J, Warwick J, Kataoka M, Warsi I, Brentnall A, Warren R, Duffy SW. A case-control study to assess the impact of mammographic density on breast cancer risk in women aged 40-49 at intermediate familial risk. Int J Cancer 2014; 136:2378-87. [PMID: 25333209 DOI: 10.1002/ijc.29275] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Accepted: 09/12/2014] [Indexed: 11/08/2022]
Abstract
Mammographic density is a strong risk factor for breast cancer, but its potential application in risk management is not clear, partly due to uncertainties about its interaction with other breast cancer risk factors. We aimed to quantify the impact of mammographic density on breast cancer risk in women aged 40-49 at intermediate familial risk of breast cancer (average lifetime risk of 23%), in particular in premenopausal women, and to investigate its relationship with other breast cancer risk factors in this population. We present the results from a case-control study nested with the FH01 cohort study of 6,710 women mostly aged 40-49 at intermediate familial risk of breast cancer. One hundred and three cases of breast cancer were age-matched to one or two controls. Density was measured by semiautomated interactive thresholding. Absolute density, but not percent density, was a significant risk factor for breast cancer in this population after adjusting for area of nondense tissue (OR per 10 cm(2) = 1.07, 95% CI 1.00-1.15, p = 0.04). The effect was stronger in premenopausal women, who made up the majority of the study population. Absolute density remained a significant predictor of breast cancer risk after adjusting for age at menarche, age at first live birth, parity, past or present hormone replacement therapy, and the Tyrer-Cuzick 10-year relative risk estimate of breast cancer. Absolute density can improve breast cancer risk stratification and delineation of high-risk groups alongside the Tyrer-Cuzick 10-year relative risk estimate.
Collapse
Affiliation(s)
- Valentina Assi
- Centre for Cancer Prevention, Wolfson Institute of Preventive Medicine, Queen Mary University of London, London, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Prosperi MCF, Ingham SL, Howell A, Lalloo F, Buchan IE, Evans DG. Can multiple SNP testing in BRCA2 and BRCA1 female carriers be used to improve risk prediction models in conjunction with clinical assessment? BMC Med Inform Decis Mak 2014; 14:87. [PMID: 25274085 PMCID: PMC4197237 DOI: 10.1186/1472-6947-14-87] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Accepted: 09/25/2014] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND Several single nucleotide polymorphisms (SNPs) at different loci have been associated with breast cancer susceptibility, accounting for around 10% of the familial component. Recent studies have found direct associations between specific SNPs and breast cancer in BRCA1/2 mutation carriers. Our aim was to determine whether validated susceptibility SNP scores improve the predictive ability of risk models in comparison/conjunction to other clinical/demographic information. METHODS Female BRCA1/2 carriers were identified from the Manchester genetic database, and included in the study regardless of breast cancer status or age. DNA was extracted from blood samples provided by these women and used for gene and SNP profiling. Estimates of survival were examined with Kaplan-Meier curves. Multivariable Cox proportional hazards models were fit in the separate BRCA datasets and in menopausal stages screening different combinations of clinical/demographic/genetic variables. Nonlinear random survival forests were also fit to identify relevant interactions. Models were compared using Harrell's concordance index (1 - c-index). RESULTS 548 female BRCA1 mutation carriers and 523 BRCA2 carriers were identified from the database. Median Kaplan-Meier estimate of survival was 46.0 years (44.9-48.1) for BRCA1 carriers and 48.9 (47.3-50.4) for BRCA2. By fitting Cox models and random survival forests, including both a genetic SNP score and clinical/demographic variables, average 1 - c-index values were 0.221 (st.dev. 0.019) for BRCA1 carriers and 0.215 (st.dev. 0.018) for BRCA2 carriers. CONCLUSIONS Random survival forests did not yield higher performance compared to Cox proportional hazards. We found improvement in prediction performance when coupling the genetic SNP score with clinical/demographic markers, which warrants further investigation.
Collapse
Affiliation(s)
- Mattia CF Prosperi
- />Institute of Population Health, Centre for Health Informatics, University of Manchester, Manchester, UK
| | - Sarah L Ingham
- />Institute of Population Health, Centre for Health Informatics, University of Manchester, Manchester, UK
| | - Anthony Howell
- />Genesis Prevention Centre, University Hospital of South Manchester, Manchester, UK
| | - Fiona Lalloo
- />Department of Genetic Medicine, Manchester Academic Health Science Centre, St. Mary’s Hospital, University of Manchester, Manchester, UK
| | - Iain E Buchan
- />Institute of Population Health, Centre for Health Informatics, University of Manchester, Manchester, UK
| | - Dafydd Gareth Evans
- />Genesis Prevention Centre, University Hospital of South Manchester, Manchester, UK
- />Department of Genetic Medicine, Manchester Academic Health Science Centre, St. Mary’s Hospital, University of Manchester, Manchester, UK
| |
Collapse
|
32
|
Gierach GL, Li H, Loud JT, Greene MH, Chow CK, Lan L, Prindiville SA, Eng-Wong J, Soballe PW, Giambartolomei C, Mai PL, Galbo CE, Nichols K, Calzone KA, Olopade OI, Gail MH, Giger ML. Relationships between computer-extracted mammographic texture pattern features and BRCA1/2 mutation status: a cross-sectional study. Breast Cancer Res 2014. [PMID: 25159706 DOI: 10.1186/preaccept-1744229618121391] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
INTRODUCTION Mammographic density is similar among women at risk of either sporadic or BRCA1/2-related breast cancer. It has been suggested that digitized mammographic images contain computer-extractable information within the parenchymal pattern, which may contribute to distinguishing between BRCA1/2 mutation carriers and non-carriers. METHODS We compared mammographic texture pattern features in digitized mammograms from women with deleterious BRCA1/2 mutations (n = 137) versus non-carriers (n = 100). Subjects were stratified into training (107 carriers, 70 non-carriers) and testing (30 carriers, 30 non-carriers) datasets. Masked to mutation status, texture features were extracted from a retro-areolar region-of-interest in each subject's digitized mammogram. Stepwise linear regression analysis of the training dataset identified variables to be included in a radiographic texture analysis (RTA) classifier model aimed at distinguishing BRCA1/2 carriers from non-carriers. The selected features were combined using a Bayesian Artificial Neural Network (BANN) algorithm, which produced a probability score rating the likelihood of each subject's belonging to the mutation-positive group. These probability scores were evaluated in the independent testing dataset to determine whether their distribution differed between BRCA1/2 mutation carriers and non-carriers. A receiver operating characteristic analysis was performed to estimate the model's discriminatory capacity. RESULTS In the testing dataset, a one standard deviation (SD) increase in the probability score from the BANN-trained classifier was associated with a two-fold increase in the odds of predicting BRCA1/2 mutation status: unadjusted odds ratio (OR) = 2.00, 95% confidence interval (CI): 1.59, 2.51, P = 0.02; age-adjusted OR = 1.93, 95% CI: 1.53, 2.42, P = 0.03. Additional adjustment for percent mammographic density did little to change the OR. The area under the curve for the BANN-trained classifier to distinguish between BRCA1/2 mutation carriers and non-carriers was 0.68 for features alone and 0.72 for the features plus percent mammographic density. CONCLUSIONS Our findings suggest that, unlike percent mammographic density, computer-extracted mammographic texture pattern features are associated with carrying BRCA1/2 mutations. Although still at an early stage, our novel RTA classifier has potential for improving mammographic image interpretation by permitting real-time risk stratification among women undergoing screening mammography.
Collapse
Affiliation(s)
- Gretchen L Gierach
- Hormonal and Reproductive Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, 9609 Medical Center Drive, Rm, 7-E108, Bethesda 20892-9774, MD, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Gierach GL, Li H, Loud JT, Greene MH, Chow CK, Lan L, Prindiville SA, Eng-Wong J, Soballe PW, Giambartolomei C, Mai PL, Galbo CE, Nichols K, Calzone KA, Olopade OI, Gail MH, Giger ML. Relationships between computer-extracted mammographic texture pattern features and BRCA1/2 mutation status: a cross-sectional study. Breast Cancer Res 2014; 16:424. [PMID: 25159706 PMCID: PMC4268674 DOI: 10.1186/s13058-014-0424-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 07/31/2014] [Indexed: 12/24/2022] Open
Abstract
Introduction Mammographic density is similar among women at risk of either sporadic or BRCA1/2-related breast cancer. It has been suggested that digitized mammographic images contain computer-extractable information within the parenchymal pattern, which may contribute to distinguishing between BRCA1/2 mutation carriers and non-carriers. Methods We compared mammographic texture pattern features in digitized mammograms from women with deleterious BRCA1/2 mutations (n = 137) versus non-carriers (n = 100). Subjects were stratified into training (107 carriers, 70 non-carriers) and testing (30 carriers, 30 non-carriers) datasets. Masked to mutation status, texture features were extracted from a retro-areolar region-of-interest in each subject’s digitized mammogram. Stepwise linear regression analysis of the training dataset identified variables to be included in a radiographic texture analysis (RTA) classifier model aimed at distinguishing BRCA1/2 carriers from non-carriers. The selected features were combined using a Bayesian Artificial Neural Network (BANN) algorithm, which produced a probability score rating the likelihood of each subject’s belonging to the mutation-positive group. These probability scores were evaluated in the independent testing dataset to determine whether their distribution differed between BRCA1/2 mutation carriers and non-carriers. A receiver operating characteristic analysis was performed to estimate the model’s discriminatory capacity. Results In the testing dataset, a one standard deviation (SD) increase in the probability score from the BANN-trained classifier was associated with a two-fold increase in the odds of predicting BRCA1/2 mutation status: unadjusted odds ratio (OR) = 2.00, 95% confidence interval (CI): 1.59, 2.51, P = 0.02; age-adjusted OR = 1.93, 95% CI: 1.53, 2.42, P = 0.03. Additional adjustment for percent mammographic density did little to change the OR. The area under the curve for the BANN-trained classifier to distinguish between BRCA1/2 mutation carriers and non-carriers was 0.68 for features alone and 0.72 for the features plus percent mammographic density. Conclusions Our findings suggest that, unlike percent mammographic density, computer-extracted mammographic texture pattern features are associated with carrying BRCA1/2 mutations. Although still at an early stage, our novel RTA classifier has potential for improving mammographic image interpretation by permitting real-time risk stratification among women undergoing screening mammography. Electronic supplementary material The online version of this article (doi:10.1186/s13058-014-0424-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Gretchen L Gierach
- Hormonal and Reproductive Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, 9609 Medical Center Drive, Rm, 7-E108, Bethesda 20892-9774, MD, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Ekpo EU, McEntee MF. Measurement of breast density with digital breast tomosynthesis--a systematic review. Br J Radiol 2014; 87:20140460. [PMID: 25146640 DOI: 10.1259/bjr.20140460] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Digital breast tomosynthesis (DBT) has gained acceptance as an adjunct to digital mammography in screening. Now that breast density reporting is mandated in several states in the USA, it is increasingly important that the methods of breast density measurement be robust, reliable and consistent. Breast density assessment with DBT needs some consideration since quantitative methods are modelled for two-dimensional (2D) mammography. A review of methods used for breast density assessment with DBT was performed. Existing evidence shows Cumulus has better reproducibility than that of the breast imaging reporting and data system (BI-RADS®) but still suffers from subjective variability; MedDensity is limited by image noise, whilst Volpara and Quantra are robust and consistent. The reported BI-RADs inter-reader breast density agreement (k) ranged from 0.65 to 0.91, with inter-reader correlation (r) ranging from 0.70 to 0.93. The correlation (r) between BI-RADS and Cumulus ranged from 0.54-0.94, whilst that of BI-RADs and MedDensity ranged from 0.48-0.78. The reported agreement (k) between BI-RADs and Volpara is 0.953. Breast density correlation between DBT and 2D mammography ranged from 0.73 to 0.97, with agreement (k) ranging from 0.56 to 0.96. To avoid variability and provide more reliable breast density information for clinicians, automated volumetric methods are preferred.
Collapse
Affiliation(s)
- E U Ekpo
- 1 Discipline of Medical Radiation Science, Faculty of Health Science, University of Sydney, Sydney, NSW, Australia
| | | |
Collapse
|
35
|
Ethnic background and genetic variation in the evaluation of cancer risk: a systematic review. PLoS One 2014; 9:e97522. [PMID: 24901479 PMCID: PMC4046957 DOI: 10.1371/journal.pone.0097522] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Accepted: 04/21/2014] [Indexed: 11/19/2022] Open
Abstract
The clinical use of genetic variation in the evaluation of cancer risk is expanding, and thus understanding how determinants of cancer susceptibility identified in one population can be applied to another is of growing importance. However there is considerable debate on the relevance of ethnic background in clinical genetics, reflecting both the significance and complexity of genetic heritage. We address this via a systematic review of reported associations with cancer risk for 82 markers in 68 studies across six different cancer types, comparing association results between ethnic groups and examining linkage disequilibrium between risk alleles and nearby genetic loci. We find that the relevance of ethnic background depends on the question. If asked whether the association of variants with disease risk is conserved across ethnic boundaries, we find that the answer is yes, the majority of markers show insignificant variability in association with cancer risk across ethnic groups. However if the question is whether a significant association between a variant and cancer risk is likely to reproduce, the answer is no, most markers do not validate in an ethnic group other than the discovery cohort's ancestry. This lack of reproducibility is not attributable to studies being inadequately populated due to low allele frequency in other ethnic groups. Instead, differences in local genomic structure between ethnic groups are associated with the strength of association with cancer risk and therefore confound interpretation of the implied physiologic association tracked by the disease allele. This suggest that a biological association for cancer risk alleles may be broadly consistent across ethnic boundaries, but reproduction of a clinical study in another ethnic group is uncommon, in part due to confounding genomic architecture. As clinical studies are increasingly performed globally this has important implications for how cancer risk stratifiers should be studied and employed.
Collapse
|
36
|
O'Flynn EAM, Wilson RM, Allen SD, Locke I, Scurr E, deSouza NM. Diffusion-weighted imaging of the high-risk breast: Apparent diffusion coefficient values and their relationship to breast density. J Magn Reson Imaging 2014; 39:805-11. [PMID: 24038529 DOI: 10.1002/jmri.24243] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Accepted: 05/03/2013] [Indexed: 12/13/2022] Open
Abstract
PURPOSE To document the apparent diffusion coefficient (ADC) of fibroglandular breast tissue in women at high-risk of developing breast cancer and investigate the relationship between ADC and breast density. MATERIALS AND METHODS Local research ethics approval was obtained. A total of 33 high-risk women including 17 BRCA1/2 mutation carriers (mean age, 43 years) and 16 women postmantle irradiation (mean age 40 years) underwent diffusion-weighted MRI between days 6 and 16 of their menstrual cycle. ADC histograms from a region of interest in fibroglandular tissue and mammographic breast density measurements were obtained. Mean, percentile ADC values (10th, 25th, 50th, 75th, 90th) and skew were compared for the two groups; ADC and mammographic breast density were correlated. RESULTS Mean ADC values (×10(-6) mm(2) /s) were 2017 ± 197 in postmantle irradiated women and 1827 ± 289 in BRCA1/2 mutation carriers (P = 0.035) with significant differences at all percentiles (P < 0.0001) but not skew (P = 0.44). ADC values showed weak positive correlation with mammographic breast density in BRCA1/2 mutation carriers (r = 0.51, P = 0.043) but not in postmantle radiotherapy patients (r = 0.49, P = 0.13). CONCLUSION Higher ADC values seen in fibroglandular tissue postmantle irradiation compared with BRCA1/2 mutation carriers has potential to improve tumor detection in these patients. Lack of correlation between ADC and breast density postmantle irradiation may be a result of microstructural changes.
Collapse
Affiliation(s)
- Elizabeth A M O'Flynn
- Clinical Magnetic Resonance Group, Cancer Research UK and EPSRC Cancer Imaging Centre, Royal Marsden NHS Foundation Trust, Downs Road, Sutton, Surrey, United Kingdom
| | | | | | | | | | | |
Collapse
|
37
|
Ahmed M, Lalloo F, Evans DG. Update on genetic predisposition to breast cancer. Expert Rev Anticancer Ther 2014; 9:1103-13. [DOI: 10.1586/era.09.38] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
38
|
High mammographic density in women of Ashkenazi Jewish descent. Breast Cancer Res 2013; 15:R40. [PMID: 23668689 PMCID: PMC4053164 DOI: 10.1186/bcr3424] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Accepted: 05/13/2013] [Indexed: 11/21/2022] Open
Abstract
Introduction Percent mammographic density (PMD) adjusted for age and body mass index is one of the strongest risk factors for breast cancer and is known to be approximately 60% heritable. Here we report a finding of an association between genetic ancestry and adjusted PMD. Methods We selected self-identified Caucasian women in the California Pacific Medical Center Research Institute Cohort whose screening mammograms placed them in the top or bottom quintiles of age-adjusted and body mass index-adjusted PMD. Our final dataset included 474 women with the highest adjusted PMD and 469 with the lowest genotyped on the Illumina 1 M platform. Principal component analysis (PCA) and identity-by-descent analyses allowed us to infer the women's genetic ancestry and correlate it with adjusted PMD. Results Women of Ashkenazi Jewish ancestry, as defined by the first principal component of PCA and identity-by-descent analyses, represented approximately 15% of the sample. Ashkenazi Jewish ancestry, defined by the first principal component of PCA, was associated with higher adjusted PMD (P = 0.004). Using multivariate regression to adjust for epidemiologic factors associated with PMD, including age at parity and use of postmenopausal hormone therapy, did not attenuate the association. Conclusions Women of Ashkenazi Jewish ancestry, based on genetic analysis, are more likely to have high age-adjusted and body mass index-adjusted PMD. Ashkenazi Jews may have a unique set of genetic variants or environmental risk factors that increase mammographic density.
Collapse
|
39
|
de Bruin MA, Kwong A, Goldstein BA, Lipson JA, Ikeda DM, McPherson L, Sharma B, Kardashian A, Schackmann E, Kingham KE, Mills MA, West DW, Ford JM, Kurian AW. Breast cancer risk factors differ between Asian and white women with BRCA1/2 mutations. Fam Cancer 2013; 11:429-39. [PMID: 22638769 DOI: 10.1007/s10689-012-9531-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The prevalence and penetrance of BRCA1 and BRCA2 (BRCA1/2) mutations may differ between Asians and whites. We investigated BRCA1/2 mutations and cancer risk factors in a clinic-based sample. BRCA1/2 mutation carriers were enrolled from cancer genetics clinics in Hong Kong and California according to standardized entry criteria. We compared BRCA mutation position, cancer history, hormonal and reproductive exposures. We analyzed DNA samples for single-nucleotide polymorphisms reported to modify breast cancer risk. We performed logistic regression to identify independent predictors of breast cancer. Fifty Asian women and forty-nine white American women were enrolled. BRCA1 mutations were more common among whites (67 vs. 42 %, p = 0.02), and BRCA2 mutations among Asians (58 vs. 37 %, p = 0.04). More Asians had breast cancer (76 vs. 53 %, p = 0.03); more whites had relatives with breast cancer (86 vs. 50 %, p = 0.0003). More whites than Asians had breastfed (71 vs. 42 %, p = 0.005), had high BMI (median 24.3 vs. 21.2, p = 0.04), consumed alcohol (2 drinks/week vs. 0, p < 0.001), and had oophorectomy (61 vs. 34 %, p = 0.01). Asians had a higher frequency of risk-associated alleles in MAP3K1 (88 vs. 59 %, p = 0.005) and TOX3/TNRC9 (88 vs. 55 %, p = 0.0002). On logistic regression, MAP3K1 was associated with increased breast cancer risk for BRCA2, but not BRCA1 mutation carriers; breast density was associated with increased risk among Asians but not whites. We found significant differences in breast cancer risk factors between Asian and white BRCA1/2 mutation carriers. Further investigation of racial differences in BRCA1/2 mutation epidemiology could inform targeted cancer risk-reduction strategies.
Collapse
Affiliation(s)
- Monique A de Bruin
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Gierach GL, Yang XR, Figueroa JD, Sherman ME. Emerging Concepts in Breast Cancer Risk Prediction. CURRENT OBSTETRICS AND GYNECOLOGY REPORTS 2012; 2:43-52. [DOI: 10.1007/s13669-012-0034-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
41
|
Saadatmand S, Rutgers EJT, Tollenaar RAEM, Zonderland HM, Ausems MGEM, Keymeulen KBMI, Schlooz-Vries MS, Koppert LB, Heijnsdijk EAM, Seynaeve C, Verhoef C, Oosterwijk JC, Obdeijn IM, de Koning HJ, Tilanus-Linthorst MMA. Breast density as indicator for the use of mammography or MRI to screen women with familial risk for breast cancer (FaMRIsc): a multicentre randomized controlled trial. BMC Cancer 2012; 12:440. [PMID: 23031619 PMCID: PMC3488502 DOI: 10.1186/1471-2407-12-440] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Accepted: 09/20/2012] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND To reduce mortality, women with a family history of breast cancer often start mammography screening at a younger age than the general population. Breast density is high in over 50% of women younger than 50 years. With high breast density, breast cancer incidence increases, but sensitivity of mammography decreases. Therefore, mammography might not be the optimal method for breast cancer screening in young women. Adding MRI increases sensitivity, but also the risk of false-positive results. The limitation of all previous MRI screening studies is that they do not contain a comparison group; all participants received both MRI and mammography. Therefore, we cannot empirically assess in which stage tumours would have been detected by either test.The aim of the Familial MRI Screening Study (FaMRIsc) is to compare the efficacy of MRI screening to mammography for women with a familial risk. Furthermore, we will assess the influence of breast density. METHODS/DESIGN This Dutch multicentre, randomized controlled trial, with balanced randomisation (1:1) has a parallel grouped design. Women with a cumulative lifetime risk for breast cancer due to their family history of ≥20%, aged 30-55 years are eligible. Identified BRCA1/2 mutation carriers or women with 50% risk of carrying a mutation are excluded. Group 1 receives yearly mammography and clinical breast examination (n = 1000), and group 2 yearly MRI and clinical breast examination, and mammography biennially (n = 1000).Primary endpoints are the number and stage of the detected breast cancers in each arm. Secondary endpoints are the number of false-positive results in both screening arms. Furthermore, sensitivity and positive predictive value of both screening strategies will be assessed. Cost-effectiveness of both strategies will be assessed. Analyses will also be performed with mammographic density as stratification factor. DISCUSSION Personalized breast cancer screening might optimize mortality reduction with less over diagnosis. Breast density may be a key discriminator for selecting the optimal screening strategy for women < 55 years with familial breast cancer risk; mammography or MRI. These issues are addressed in the FaMRIsc study including high risk women due to a familial predisposition. TRIAL REGISTRATION Netherland Trial Register NTR2789.
Collapse
Affiliation(s)
- Sepideh Saadatmand
- Department of Surgery, Erasmus University Medical Centre, Rotterdam, Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Killick E, Bancroft E, Kote-Jarai Z, Eeles R. Beyond prostate-specific antigen - future biomarkers for the early detection and management of prostate cancer. Clin Oncol (R Coll Radiol) 2012; 24:545-55. [PMID: 22682955 DOI: 10.1016/j.clon.2012.05.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2011] [Revised: 03/02/2012] [Accepted: 05/08/2012] [Indexed: 12/31/2022]
Abstract
Prostate-specific antigen is currently commonly used as a screening biomarker for prostate cancer, but it has limitations in both sensitivity and specificity. The development of novel biomarkers for early cancer detection has the potential to improve survival, reduce unnecessary investigations and benefit the health economy. Here we review the use and limitations of prostate-specific antigen and its subtypes, urinary biomarkers including PCA3, alpha-methylacyl-CoA racemase, the TMPRSS2-ERG fusion gene and microseminoprotein-beta, and other novel markers in both serum and urine. Many of these biomarkers are at early stages of development and require evaluation in prospective trials to determine their potential usefulness in clinical practice. Genetic profiling may allow for the targeting of high-risk populations for screening and may offer the opportunity to combine biomarker results with genotype to aid risk assessment.
Collapse
Affiliation(s)
- E Killick
- Institute of Cancer Research, Sutton, Surrey, UK.
| | | | | | | |
Collapse
|
43
|
Vachon CM, Scott CG, Fasching PA, Hall P, Tamimi RM, Li J, Stone J, Apicella C, Odefrey F, Gierach GL, Jud SM, Heusinger K, Beckmann MW, Pollan M, Fernández-Navarro P, Gonzalez-Neira A, Benitez J, van Gils CH, Lokate M, Onland-Moret NC, Peeters PHM, Brown J, Leyland J, Varghese JS, Easton DF, Thompson DJ, Luben RN, Warren RML, Wareham NJ, Loos RJF, Khaw KT, Ursin G, Lee E, Gayther SA, Ramus SJ, Eeles RA, Leach MO, Kwan-Lim G, Couch FJ, Giles GG, Baglietto L, Krishnan K, Southey MC, Le Marchand L, Kolonel LN, Woolcott C, Maskarinec G, Haiman CA, Walker K, Johnson N, McCormack VA, Biong M, Alnaes GIG, Gram IT, Kristensen VN, Børresen-Dale AL, Lindström S, Hankinson SE, Hunter DJ, Andrulis IL, Knight JA, Boyd NF, Figuero JD, Lissowska J, Wesolowska E, Peplonska B, Bukowska A, Reszka E, Liu J, Eriksson L, Czene K, Audley T, Wu AH, Pankratz VS, Hopper JL, dos-Santos-Silva I. Common breast cancer susceptibility variants in LSP1 and RAD51L1 are associated with mammographic density measures that predict breast cancer risk. Cancer Epidemiol Biomarkers Prev 2012; 21:1156-66. [PMID: 22454379 PMCID: PMC3569092 DOI: 10.1158/1055-9965.epi-12-0066] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Mammographic density adjusted for age and body mass index (BMI) is a heritable marker of breast cancer susceptibility. Little is known about the biologic mechanisms underlying the association between mammographic density and breast cancer risk. We examined whether common low-penetrance breast cancer susceptibility variants contribute to interindividual differences in mammographic density measures. METHODS We established an international consortium (DENSNP) of 19 studies from 10 countries, comprising 16,895 Caucasian women, to conduct a pooled cross-sectional analysis of common breast cancer susceptibility variants in 14 independent loci and mammographic density measures. Dense and nondense areas, and percent density, were measured using interactive-thresholding techniques. Mixed linear models were used to assess the association between genetic variants and the square roots of mammographic density measures adjusted for study, age, case status, BMI, and menopausal status. RESULTS Consistent with their breast cancer associations, the C-allele of rs3817198 in LSP1 was positively associated with both adjusted dense area (P = 0.00005) and adjusted percent density (P = 0.001), whereas the A-allele of rs10483813 in RAD51L1 was inversely associated with adjusted percent density (P = 0.003), but not with adjusted dense area (P = 0.07). CONCLUSION We identified two common breast cancer susceptibility variants associated with mammographic measures of radiodense tissue in the breast gland. IMPACT We examined the association of 14 established breast cancer susceptibility loci with mammographic density phenotypes within a large genetic consortium and identified two breast cancer susceptibility variants, LSP1-rs3817198 and RAD51L1-rs10483813, associated with mammographic measures and in the same direction as the breast cancer association.
Collapse
Affiliation(s)
- Celine M Vachon
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55905, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
High and low mammographic density human breast tissues maintain histological differential in murine tissue engineering chambers. Breast Cancer Res Treat 2012; 135:177-87. [DOI: 10.1007/s10549-012-2128-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2012] [Accepted: 06/04/2012] [Indexed: 01/11/2023]
|
45
|
Barnes DR, Antoniou AC. Unravelling modifiers of breast and ovarian cancer risk for BRCA1 and BRCA2 mutation carriers: update on genetic modifiers. J Intern Med 2012; 271:331-43. [PMID: 22443199 DOI: 10.1111/j.1365-2796.2011.02502.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Pathogenic mutations in the tumour suppressor genes BRCA1 and BRCA2 confer increased risks for breast and ovarian cancer and account for approximately 15% of the excess familial risk of breast cancer amongst first-degree relatives of patients with breast cancer. There is considerable evidence indicating that these risks vary by other genetic and environmental factors clustering in families. In the past few years, based on the availability of genome-wide association data and samples from large collaborative studies, several common alleles have been found to modify breast or ovarian cancer risk for BRCA1 and BRCA2 mutation carriers. These common alleles explain a small proportion of the genetic variability in breast or ovarian cancer risk for mutation carriers, suggesting more modifiers remain to be identified. We review the so far identified genetic modifiers of breast and ovarian cancer risk and consider the implications for risk prediction. BRCA1 and BRCA2 mutation carriers could be some of the first to benefit from clinical applications of common variants identified through genome-wide association studies. However, to be able to provide more individualized risk estimates, it will be important to understand how the associations vary with different tumour characteristics and their interactions with other genetic and environmental modifiers.
Collapse
Affiliation(s)
- D R Barnes
- Department of Public Health and Primary Care, Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge, UK
| | | |
Collapse
|
46
|
Varghese JS, Thompson DJ, Michailidou K, Lindström S, Turnbull C, Brown J, Leyland J, Warren RML, Luben RN, Loos RJ, Wareham NJ, Rommens J, Paterson AD, Martin LJ, Vachon CM, Scott CG, Atkinson EJ, Couch FJ, Apicella C, Southey MC, Stone J, Li J, Eriksson L, Czene K, Boyd NF, Hall P, Hopper JL, Tamimi RM, Rahman N, Easton DF. Mammographic breast density and breast cancer: evidence of a shared genetic basis. Cancer Res 2012; 72:1478-84. [PMID: 22266113 PMCID: PMC3378688 DOI: 10.1158/0008-5472.can-11-3295] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Percent mammographic breast density (PMD) is a strong heritable risk factor for breast cancer. However, the pathways through which this risk is mediated are still unclear. To explore whether PMD and breast cancer have a shared genetic basis, we identified genetic variants most strongly associated with PMD in a published meta-analysis of five genome-wide association studies (GWAS) and used these to construct risk scores for 3,628 breast cancer cases and 5,190 controls from the UK2 GWAS of breast cancer. The signed per-allele effect estimates of single-nucleotide polymorphisms (SNP) were multiplied with the respective allele counts in the individual and summed over all SNPs to derive the risk score for an individual. These scores were included as the exposure variable in a logistic regression model with breast cancer case-control status as the outcome. This analysis was repeated using 10 different cutoff points for the most significant density SNPs (1%-10% representing 5,222-50,899 SNPs). Permutation analysis was also conducted across all 10 cutoff points. The association between risk score and breast cancer was significant for all cutoff points from 3% to 10% of top density SNPs, being most significant for the 6% (2-sided P = 0.002) to 10% (P = 0.001) cutoff points (overall permutation P = 0.003). Women in the top 10% of the risk score distribution had a 31% increased risk of breast cancer [OR = 1.31; 95% confidence interval (CI), 1.08-1.59] compared with women in the bottom 10%. Together, our results show that PMD and breast cancer have a shared genetic basis that is mediated through a large number of common variants.
Collapse
Affiliation(s)
- Jajini S Varghese
- Centre for Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Deborah J Thompson
- Centre for Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Kyriaki Michailidou
- Centre for Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Sara Lindström
- Department of Epidemiology, Harvard School of Public Health, Boston, MA, USA
| | - Clare Turnbull
- Section of Cancer Genetics, The Institute of Cancer Research, Sutton, Surrey, UK
| | - Judith Brown
- Centre for Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Jean Leyland
- Centre for Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Ruth ML Warren
- Department of Radiology, University of Cambridge, Addenbrooke’s NHS Foundation Trust Cambridge, UK
| | - Robert N Luben
- Centre for Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Ruth J Loos
- MRC Epidemiology Unit, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, UK
| | - Nicholas J Wareham
- MRC Epidemiology Unit, Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, UK
| | - Johanna Rommens
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Andrew D Paterson
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
- Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Lisa J Martin
- Campbell Family Institute for Breast Cancer Research, Ontario Cancer Institute, Toronto, Ontario, Canada
| | - Celine M Vachon
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | | | | | - Fergus J Couch
- Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Carmel Apicella
- Centre for M.E.G.A. Epidemiology, Melbourne School of Population Health, The University of Melbourne, Melbourne, Australia
| | - Melissa C Southey
- Genetic Epidemiology Laboratory, Department of Pathology, The University of Melbourne, Melbourne, Australia
| | - Jennifer Stone
- Centre for M.E.G.A. Epidemiology, Melbourne School of Population Health, The University of Melbourne, Melbourne, Australia
| | - Jingmei Li
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Human Genetics, Genome Institute of Singapore, Singapore
| | - Louise Eriksson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Kamila Czene
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Norman F Boyd
- Campbell Family Institute for Breast Cancer Research, Ontario Cancer Institute, Toronto, Ontario, Canada
| | - Per Hall
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - John L Hopper
- Centre for M.E.G.A. Epidemiology, Melbourne School of Population Health, The University of Melbourne, Melbourne, Australia
| | - Rulla M Tamimi
- Department of Epidemiology, Harvard School of Public Health, Boston, MA, USA
- Channing Laboratory, Brigham and Women’s Hospital, Boston, MA, USA
| | - Nazneen Rahman
- Section of Cancer Genetics, The Institute of Cancer Research, Sutton, Surrey, UK
| | - Douglas F Easton
- Centre for Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| |
Collapse
|
47
|
Antoniou AC, Kuchenbaecker KB, Soucy P, Beesley J, Chen X, McGuffog L, Lee A, Barrowdale D, Healey S, Sinilnikova OM, Caligo MA, Loman N, Harbst K, Lindblom A, Arver B, Rosenquist R, Karlsson P, Nathanson K, Domchek S, Rebbeck T, Jakubowska A, Lubinski J, Jaworska K, Durda K, Złowowcka-Perłowska E, Osorio A, Durán M, Andrés R, Benítez J, Hamann U, Hogervorst FB, van Os TA, Verhoef S, Meijers-Heijboer HEJ, Wijnen J, Gómez Garcia EB, Ligtenberg MJ, Kriege M, Collée JM, Ausems MGEM, Oosterwijk JC, Peock S, Frost D, Ellis SD, Platte R, Fineberg E, Evans DG, Lalloo F, Jacobs C, Eeles R, Adlard J, Davidson R, Cole T, Cook J, Paterson J, Douglas F, Brewer C, Hodgson S, Morrison PJ, Walker L, Rogers MT, Donaldson A, Dorkins H, Godwin AK, Bove B, Stoppa-Lyonnet D, Houdayer C, Buecher B, de Pauw A, Mazoyer S, Calender A, Léoné M, Bressac- de Paillerets B, Caron O, Sobol H, Frenay M, Prieur F, Ferrer SF, Mortemousque I, Buys S, Daly M, Miron A, Terry MB, Hopper JL, John EM, Southey M, Goldgar D, Singer CF, Fink-Retter A, Tea MK, Kaulich DG, Hansen TVO, Nielsen FC, Barkardottir RB, Gaudet M, Kirchhoff T, Joseph V, Dutra-Clarke A, Offit K, Piedmonte M, Kirk J, Cohn D, Hurteau J, Byron J, Fiorica J, Toland AE, Montagna M, Oliani C, Imyanitov E, Isaacs C, Tihomirova L, Blanco I, Lazaro C, Teulé A, Valle JD, Gayther SA, Odunsi K, Gross J, Karlan BY, Olah E, Teo SH, Ganz PA, Beattie MS, Dorfling CM, van Rensburg EJ, Diez O, Kwong A, Schmutzler RK, Wappenschmidt B, Engel C, Meindl A, Ditsch N, Arnold N, Heidemann S, Niederacher D, Preisler-Adams S, Gadzicki D, Varon-Mateeva R, Deissler H, Gehrig A, Sutter C, Kast K, Fiebig B, Schäfer D, Caldes T, de la Hoya M, Nevanlinna H, Muranen TA, Lespérance B, Spurdle AB, Neuhausen SL, Ding YC, Wang X, Fredericksen Z, Pankratz VS, Lindor NM, Peterlongo P, Manoukian S, Peissel B, Zaffaroni D, Bonanni B, Bernard L, Dolcetti R, Papi L, Ottini L, Radice P, Greene MH, Loud JT, Andrulis IL, Ozcelik H, Mulligan AM, Glendon G, Thomassen M, Gerdes AM, Jensen UB, Skytte AB, Kruse TA, Chenevix-Trench G, Couch FJ, Simard J, Easton DF. Common variants at 12p11, 12q24, 9p21, 9q31.2 and in ZNF365 are associated with breast cancer risk for BRCA1 and/or BRCA2 mutation carriers. Breast Cancer Res 2012; 14:R33. [PMID: 22348646 PMCID: PMC3496151 DOI: 10.1186/bcr3121] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2011] [Revised: 11/15/2011] [Accepted: 02/20/2012] [Indexed: 01/15/2023] Open
Abstract
INTRODUCTION Several common alleles have been shown to be associated with breast and/or ovarian cancer risk for BRCA1 and BRCA2 mutation carriers. Recent genome-wide association studies of breast cancer have identified eight additional breast cancer susceptibility loci: rs1011970 (9p21, CDKN2A/B), rs10995190 (ZNF365), rs704010 (ZMIZ1), rs2380205 (10p15), rs614367 (11q13), rs1292011 (12q24), rs10771399 (12p11 near PTHLH) and rs865686 (9q31.2). METHODS To evaluate whether these single nucleotide polymorphisms (SNPs) are associated with breast cancer risk for BRCA1 and BRCA2 carriers, we genotyped these SNPs in 12,599 BRCA1 and 7,132 BRCA2 mutation carriers and analysed the associations with breast cancer risk within a retrospective likelihood framework. RESULTS Only SNP rs10771399 near PTHLH was associated with breast cancer risk for BRCA1 mutation carriers (per-allele hazard ratio (HR) = 0.87, 95% CI: 0.81 to 0.94, P-trend = 3 × 10-4). The association was restricted to mutations proven or predicted to lead to absence of protein expression (HR = 0.82, 95% CI: 0.74 to 0.90, P-trend = 3.1 × 10-5, P-difference = 0.03). Four SNPs were associated with the risk of breast cancer for BRCA2 mutation carriers: rs10995190, P-trend = 0.015; rs1011970, P-trend = 0.048; rs865686, 2df-P = 0.007; rs1292011 2df-P = 0.03. rs10771399 (PTHLH) was predominantly associated with estrogen receptor (ER)-negative breast cancer for BRCA1 mutation carriers (HR = 0.81, 95% CI: 0.74 to 0.90, P-trend = 4 × 10-5) and there was marginal evidence of association with ER-negative breast cancer for BRCA2 mutation carriers (HR = 0.78, 95% CI: 0.62 to 1.00, P-trend = 0.049). CONCLUSIONS The present findings, in combination with previously identified modifiers of risk, will ultimately lead to more accurate risk prediction and an improved understanding of the disease etiology in BRCA1 and BRCA2 mutation carriers.
Collapse
Affiliation(s)
- Antonis C Antoniou
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Worts Causeway, Cambridge CB1 8RN, UK
| | - Karoline B Kuchenbaecker
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Worts Causeway, Cambridge CB1 8RN, UK
| | - Penny Soucy
- Cancer Genomics Laboratory, Centre Hospitalier Universitaire de Québec, 2705 Laurier Boulevard, T3-57, Quebec City, QC Canada
| | - Jonathan Beesley
- Genetics and Population Health Division, Queensland Institute of Medical Research, 300 Herston Rd, Herston, Brisbane, QLD 4006, Australia
| | - Xiaoqing Chen
- Genetics and Population Health Division, Queensland Institute of Medical Research, 300 Herston Rd, Herston, Brisbane, QLD 4006, Australia
| | - Lesley McGuffog
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Worts Causeway, Cambridge CB1 8RN, UK
| | - Andrew Lee
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Worts Causeway, Cambridge CB1 8RN, UK
| | - Daniel Barrowdale
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Worts Causeway, Cambridge CB1 8RN, UK
| | - Sue Healey
- Genetics and Population Health Division, Queensland Institute of Medical Research, 300 Herston Rd, Herston, Brisbane, QLD 4006, Australia
| | - Olga M Sinilnikova
- Unité Mixte de Génétique Constitutionnelle des Cancers Fréquents, Centre Hospitalier Universitaire de Lyon/Centre Léon Bérard, 28 rue Laënnec, Lyon 69373, France and INSERM U1052, CNRS UMR5286, Université Lyon 1, Cancer Research Center of Lyon, 28 rue Laënnec, Lyon 69373, France
| | - Maria A Caligo
- Section of Genetic Oncology, Dept. of Laboratory Medicine, University and University Hospital of Pisa, Via Roma 57, 56125 Pisa, Italy
| | - Niklas Loman
- Department of Oncology, Lund University Hospital, Lund, Sweden
| | - Katja Harbst
- Department of Oncology, Lund University Hospital, Lund, Sweden
| | - Annika Lindblom
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden
| | - Brita Arver
- Department of Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Richard Rosenquist
- Department of Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Per Karlsson
- Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Kate Nathanson
- Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Susan Domchek
- Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Tim Rebbeck
- Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Anna Jakubowska
- Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Jan Lubinski
- Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Katarzyna Jaworska
- Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Katarzyna Durda
- Department of Genetics and Pathology, Pomeranian Medical University, Szczecin and Postgraduate School of Molecular Medicine, Warsaw Medical University, Warsaw, Poland
| | | | - Ana Osorio
- Human Genetics Group, Human Cancer Genetics Programme, Spanish National Cancer Research Centre, Madrid, Spain and Spanish Network on Rare Diseases (CIBERER)
| | - Mercedes Durán
- Institute of Biology and Molecular Genetics. Universidad de Valladolid (IBGM-UVA), Valladolid, Spain
| | - Raquel Andrés
- Oncology unit. Hospital clinico Universitario "Lozano Blesa", Zaragoza, Spain
| | - Javier Benítez
- Human Genetics Group and Genotyping Unit, Human Cancer Genetics Programme, Spanish National Cancer Research Centre, Madrid, Spain and Spanish Network on Rare Diseases (CIBERER)
| | - Ute Hamann
- Molecular Genetics of Breast Cancer, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | - Frans B Hogervorst
- Family Cancer Clinic, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Theo A van Os
- Department of Clinical Genetics, Academic Meical Center, Amsterdam, The Netherlands
| | - Senno Verhoef
- Department of Clinical Genetics, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | - Juul Wijnen
- Department of Clinical Genetics and GROM, School for Oncology and Developmental Biology, MUMC, Maastricht, The Netherlands
| | - Encarna B Gómez Garcia
- Department of Clinical Genetics and GROM, School for Oncology and Developmental Biology, MUMC, Maastricht, The Netherlands
| | - Marjolijn J Ligtenberg
- Department of Human Genetics, Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands
| | - Mieke Kriege
- Department of Clinical Genetics, Family Cancer Clinic, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - J Margriet Collée
- Department of Clinical Genetics, Family Cancer Clinic, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Margreet GEM Ausems
- Department of Medical Genetics, University Medical Center Utrecht, PO Box 85090, 3508 AB Utrecht, The Netherlands
| | - Jan C Oosterwijk
- Department of Genetics, University Medical Center, Groningen University, Groningen, The Netherlands
| | - Susan Peock
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Worts Causeway, Cambridge CB1 8RN, UK
| | - Debra Frost
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Worts Causeway, Cambridge CB1 8RN, UK
| | - Steve D Ellis
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Worts Causeway, Cambridge CB1 8RN, UK
| | - Radka Platte
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Worts Causeway, Cambridge CB1 8RN, UK
| | - Elena Fineberg
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Worts Causeway, Cambridge CB1 8RN, UK
| | - D Gareth Evans
- Genetic Medicine, Manchester Academic Health Sciences Centre, Central Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| | - Fiona Lalloo
- Genetic Medicine, Manchester Academic Health Sciences Centre, Central Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| | - Chris Jacobs
- Clinical Genetics, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Ros Eeles
- Oncogenetics Team, The Institute of Cancer Research and Royal Marsden NHS Foundation Trust, UK
| | | | - Rosemarie Davidson
- Ferguson-Smith Centre for Clinical Genetics, Yorkhill Hospitals, Glasgow, UK
| | - Trevor Cole
- West Midlands Regional Genetics Service, Birmingham Women's Hospital Healthcare NHS Trust, Edgbaston, Birmingham, UK
| | - Jackie Cook
- Sheffield Clinical Genetics Service, Sheffield Children's Hospital, Sheffield, UK
| | - Joan Paterson
- Department of Clinical Genetics, East Anglian Regional Genetics Service, Addenbrookes Hospital, Cambridge, UK
| | - Fiona Douglas
- Institute of Genetic Medicine, Centre for Life, Newcastle Upon Tyne Hospitals NHS Trust, Newcastle upon Tyne, UK
| | - Carole Brewer
- Department of Clinical Genetics, Royal Devon & Exeter Hospital, Exeter, UK
| | - Shirley Hodgson
- Medical Genetics Unit, St George's, University of London, UK
| | - Patrick J Morrison
- Northern Ireland Regional Genetics Centre, Belfast Health and Social Care Trust, and Department of Medical Genetics, Queens University Belfast, Belfast UK
| | - Lisa Walker
- Oxford Regional Genetics Service, Churchill Hospital, Oxford, UK
| | - Mark T Rogers
- All Wales Medical Genetics Services, University Hospital of Wales, Cardiff, UK
| | - Alan Donaldson
- Clinical Genetics Department, St Michael's Hospital, Bristol, UK
| | - Huw Dorkins
- North West Thames Regional Genetics Service, Kennedy-Galton Centre, Harrow, UK
| | - Andrew K Godwin
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Betsy Bove
- Clinical Molecular Genetics Laboratory, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Dominique Stoppa-Lyonnet
- Service de Génétique Oncologique, Institut Curie, Paris, France, Unité INSERM U830, Institut Curie, Paris, France, Université Paris Descartes, Faculté de Médecine, Paris, France
| | - Claude Houdayer
- Service de Génétique Oncologique, Institut Curie, Paris, France and Université Paris Descartes, Faculté de Pharmacie, Paris, France
| | - Bruno Buecher
- Service de Génétique Oncologique, Institut Curie, 26 rue d'Ulm, Paris, France
| | - Antoine de Pauw
- Service de Génétique Oncologique, Institut Curie, Paris, France
| | - Sylvie Mazoyer
- INSERM U1052, CNRS UMR5286, Université Lyon 1, Centre de Recherche en Cancérologie de Lyon, Lyon, France
| | - Alain Calender
- Unité Mixte de Génétique Constitutionnelle des Cancers Fréquents, Hospices Civils de Lyon/Centre Léon Bérard, Lyon, France
| | - Mélanie Léoné
- Unité Mixte de Génétique Constitutionnelle des Cancers Fréquents, Hospices Civils de Lyon/Centre Léon Bérard, Lyon, France
| | - Brigitte Bressac- de Paillerets
- Service de Génétique, Institut de Cancérologie Gustave Roussy, Villejuif, France and INSERM U946, Fondation Jean Dausset, Paris, France
| | - Olivier Caron
- Consultation de Génétique, Département de Médecine, Institut de Cancérologie Gustave Roussy, Villejuif, France
| | - Hagay Sobol
- Département Oncologie génétique, Prévention et Dépistage, INSERM CIC-P9502, Institut Paoli-Calmettes/Université d'Aix-Marseille II, Marseille, France
| | | | - Fabienne Prieur
- Service de Génétique Clinique Chromosomique et Moléculaire, Centre Hospitalier Universitaire de St Etienne, St Etienne, France
| | - Sandra Fert Ferrer
- Laboratoire de Génétique Chromosomique, Hôtel Dieu Centre Hospitalier, BP 1125 Chambéry, France
| | | | - Saundra Buys
- Huntsman Cancer Institute, 2000 Circle of Hope, Salt Lake City, UT 84112, USA
| | - Mary Daly
- Division of Population Science, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111, USA
| | - Alexander Miron
- Department of Cancer Biology, Dana-Farber Cancer Institute, and Department of Surgery, Harvard Medical School, 27 Drydock Avenue, Boston, MA 02210, USA
| | - Mary Beth Terry
- Department of Epidemiology, Columbia University, New York, NY, USA
| | - John L Hopper
- Centre for Molecular, Environmental, Genetic and Analytic (MEGA) Epidemiology, Melbourne School of Population Health, Level 1, 723 Swanston Street, The University of Melbourne, VIC 3010, Australia
| | - Esther M John
- Department of Epidemiology, Cancer Prevention Institute of California, 2201 Walnut Avenue, Suite 300, Fremont, CA 94538, USA
| | - Melissa Southey
- Genetic Epidemiology Laboratory, Department of Pathology, University of Melbourne, Australia
| | - David Goldgar
- Department of Dermatology, University of Utah School of Medicine, 30 North 1900 East, SOM 4B454, Salt Lake City, UT 84132, USA
| | - Christian F Singer
- Dept of OB/GYN and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Anneliese Fink-Retter
- Dept of OB/GYN and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Muy-Kheng Tea
- Dept of OB/GYN and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | | | - Thomas VO Hansen
- Center for Genomic Medicine, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Finn C Nielsen
- Center for Genomic Medicine, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Rosa B Barkardottir
- Department of Pathology, Landspitali - University Hospital, Reykjavik Iceland and Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Mia Gaudet
- Epidemiology Research Program, American Cancer Society, Atlanta, GA, USA
| | - Tomas Kirchhoff
- Department of Environmental Medicine, NYU Cancer Institute, New York University School of Medicine, New York, NY, USA
| | - Vijai Joseph
- Clinical Cancer Genetics Laboratory, Memorial Sloane Kettering Cancer Center, New York, NY, USA
| | - Ana Dutra-Clarke
- Clinical Cancer Genetics Laboratory, Memorial Sloane Kettering Cancer Center, New York, NY, USA
| | - Kenneth Offit
- Clinical Cancer Genetics Laboratory, Memorial Sloane Kettering Cancer Center, New York, NY, USA
| | - Marion Piedmonte
- Statistical and Data Center, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Judy Kirk
- Australia New Zealand (ANZGOG), Westmead Hospital, Sydney, Australia
| | - David Cohn
- Ohio State University, Columbus Cancer Council, Columbus, OH, USA
| | - Jean Hurteau
- Evanston CCOP - NorthShore University Health System; University of Chicago, Chicago, IL, USA
| | - John Byron
- Southern Pines Women's Health Center, P.C., University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - James Fiorica
- Sarasota Memorial Healthcare, Tufts Medical Center, Sarasota, Florida, USA
| | - Amanda E Toland
- Department of Molecular Virology, Immunology and Medical Genetics and Internal Medicine, Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Marco Montagna
- Immunology and Molecular Oncology Unit, Istituto Oncologico Veneto IOV - IRCCS, Padua, Italy
| | | | - Evgeny Imyanitov
- Laboratory of Molecular Oncology, N.N. Petrov Institute of Oncology, St.-Petersburg, Russia
| | - Claudine Isaacs
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington DC, USA
| | | | - Ignacio Blanco
- Genetic Counselling Unit, Hereditary Cancer Program, IDIBELL-Catalan Institute of Oncology, Barcelona, Spain
| | - Conxi Lazaro
- Molecular Diagnostic Unit, Hereditary Cancer Program, IDIBELL-Catalan Institute of Oncology, Barcelona, Spain
| | - Alex Teulé
- Genetic Counselling Unit, Hereditary Cancer Program, IDIBELL-Catalan Institute of Oncology, Barcelona, Spain
| | - J Del Valle
- Molecular Diagnostic Unit, Hereditary Cancer Program, IDIBELL-Catalan Institute of Oncology, Barcelona, Spain
| | - Simon A Gayther
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Kunle Odunsi
- Department of Gynecologic Oncology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Jenny Gross
- Women's Cancer Program at the Samuel Oschin Comprehensive Cancer Institute at Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Beth Y Karlan
- Women's Cancer Program at the Samuel Oschin Comprehensive Cancer Institute at Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Edith Olah
- Department of Molecular Genetics, National Institute of Oncology, Budapest, Hungary
| | - Soo-Hwang Teo
- Cancer Research Initiatives Foundation, Sime Darby Medical Centre, Malaysia and University Malaya Cancer Research Institute, University Malaya Medical Centre, Kuala Lumpur, Malaysia
| | - Patricia A Ganz
- Jonsson Comprehensive Cancer Center at UCLA, Los Angeles, CA, USA
| | - Mary S Beattie
- UCSF Cancer Risk Program, University of California, San Francisco, CA; UCSF Departments of Medicine, Epidemiology, and Biostatistics, Sand Francisco, CA, USA
| | - Cecelia M Dorfling
- Cancer Genetics Laboratory, Department of Genetics, University of Pretoria, South Africa
| | | | - Orland Diez
- Oncogenetics Laboratory. Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron University Hospital. Barcelona, Spain
| | - Ava Kwong
- The Hong Kong Hereditary Breast Cancer Family Registry; The Universtiy of Hong Kong; Cancer Genetics Center, Hong Kong Sanatorium and Hospital, Hong Kong
| | - Rita K Schmutzler
- Centre of Familial Breast and Ovarian Cancer, Department of Gynaecology and Obstetrics and Centre for Integrated Oncology (CIO), University hospital of Cologne, Cologne, Germany
| | - Barbara Wappenschmidt
- Centre of Familial Breast and Ovarian Cancer, Department of Gynaecology and Obstetrics and Centre for Integrated Oncology (CIO), University hospital of Cologne, Cologne, Germany
| | - Christoph Engel
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
| | - Alfons Meindl
- Department of Gynaecology and Obstetrics, Division of Tumour Genetics, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Nina Ditsch
- Department of Gynaecology and Obstetrics, Ludwig-Maximilian University Munich, Munich, Germany
| | - Norbert Arnold
- Department of Gynaecology and Obstetrics, University Hospital of Schleswig-Holstein, Campus Kiel, Christian-Albrechts University Kiel, Kiel, Germany
| | - Simone Heidemann
- Institute of Human Genetics, University Hospital of Schleswig-Holstein, Campus Kiel, Christian-Albrechts University Kiel, Kiel, Germany
| | - Dieter Niederacher
- Department of Gynaecology and Obstetrics, University Hospital Düsseldorf, Heinrich-Heine University, Düsseldorf, Germany
| | | | - Dorothea Gadzicki
- Institute of Cell and Molecular Pathology, Hannover Medical School, Hannover, Germany
| | | | - Helmut Deissler
- Department of Gynaecology and Obstetrics, University Hospital Ulm, Germany
| | - Andrea Gehrig
- Centre of Familial Breast and Ovarian Cancer, Department of Medical Genetics, Institute of Human Genetics, University Würzburg, Würzburg, Germany
| | - Christian Sutter
- Institute of Human Genetics, Department of Human Genetics, University Hospital Heidelberg, Germany
| | - Karin Kast
- Department of Gynaecology and Obstetrics, University Hospital Carl Gustav Carus, Technical University. Dresden, Germany
| | - Britta Fiebig
- Institute of Human Genetics, University Regensburg, Regensbirg. Germany
| | - Dieter Schäfer
- Institute of Human Genetics, University Hospital Frankfurt a.M., Germany Molecular Oncology Laboratory, Hospital Clinico San Carlos, Madrid, Spain
| | - Trinidad Caldes
- Molecular Oncology Laboratory, Hospital Clinico San Carlos, Martin Lagos s/n, Madrid, Spain
| | - Miguel de la Hoya
- Molecular Oncology Laboratory, Hospital Clinico San Carlos, Martin Lagos s/n, Madrid, Spain
| | - Heli Nevanlinna
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Central Hospital, Biomedicum Helsinki, P.O. BOX 700, 00029 HUS, Helsinki, Finland
| | - Taru A Muranen
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Central Hospital, Biomedicum Helsinki, P.O. BOX 700, 00029 HUS, Helsinki, Finland
| | - Bernard Lespérance
- Faculty of Medicine - Medicine and Medical Specialties, Université de Montréal Hemato-oncology service, Hôpital du Sacré-Coeur de Montréal, 5400 Gouin Blvd West Montreal, QC, Canada
| | - Amanda B Spurdle
- Genetics and Population Health Division, Queensland Institute of Medical Research, 300 Herston Rd, Herston, Brisbane, QLD 4006, Australia
| | - Susan L Neuhausen
- Department of Population Sciences, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Yuan C Ding
- Department of Population Sciences, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Xianshu Wang
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | | | | | - Noralane M Lindor
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Paolo Peterlongo
- Unit of Molecular Bases of Genetic Risk and Genetic Testing, Department of Preventive and Predicted Medicine, Fondazione IRCCS Istituto Nazionale Tumouri (INT), Milan, Italy and IFOM, Fondazione Istituto FIRC di Oncologia Molecolare, Milan, Italy
| | - Siranoush Manoukian
- Unit of Medical Genetics, Department of Preventive and Predictive Medicine, Fondazione IRCCS Istituto Nazionale Tumouri (INT), Milan, Italy
| | - Bernard Peissel
- Unit of Medical Genetics, Department of Preventive and Predictive Medicine, Fondazione IRCCS Istituto Nazionale Tumouri (INT), Milan, Italy
| | - Daniela Zaffaroni
- Unit of Medical Genetics, Department of Preventive and Predictive Medicine, Fondazione IRCCS Istituto Nazionale Tumouri (INT), Milan, Italy
| | - Bernardo Bonanni
- Division of Cancer Prevention and Genetics, Istituto Europeo di Oncologia (IEO), Milan Italy
| | - Loris Bernard
- Department of Experimental Oncology, Istituto Europeo di Oncologia, Milan, Italy and Consortium for Genomics Technology (Cogentech), Milan, Italy
| | - Riccardo Dolcetti
- Cancer Bioimmunotherapy Unit, Centro di Riferimento Oncologico, IRCCS, Aviano (PN), Italy
| | - Laura Papi
- Medical Genetics Unit, Department of Clinical Physiopathology, University of Florence, Firenze, Italy
| | - Laura Ottini
- Department of Molecular Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Paolo Radice
- Unit of Molecular Bases of Genetic Risk and Genetic Testing, Department of Preventive and Predicted Medicine, Fondazione IRCCS Istituto Nazionale Tumouri (INT), Milan, Italy and IFOM, Fondazione Istituto FIRC di Oncologia Molecolare, Milan, Italy
| | - Mark H Greene
- Clinical Genetics Branch, DCEG, NCI; Room EPS 7032, Rockville, MD 20852, USA
| | - Jennifer T Loud
- Clinical Genetics Branch, DCEG, NCI; Room EPS 7032, Rockville, MD 20852, USA
| | - Irene L Andrulis
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, ON; Cancer Care Ontario, Departments of Molecular Genetics and Laboratory Medicine and Pathobiology, University of Toronto, ON, Canada
| | - Hilmi Ozcelik
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, ON; Department of Laboratory Medicine and Pathobiology, University of Toronto, ON, Canada
| | - Anna Marie Mulligan
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada; Department of Laboratory Medicine, and the Keenan Research Centre of the Li Ka Shing Knowledge Institute, St Michael's Hospital, Toronto, ON, Canada
| | - Gord Glendon
- Ontario Cancer Genetics Network: Cancer Care Ontario, Toronto, ON, Canada
| | - Mads Thomassen
- Department of Clinical Genetics, Odense University Hospital, Denmark
| | - Anne-Marie Gerdes
- Department of Clincial Genetics, Rigshospital and Copenhagen University, Denmark
| | - Uffe B Jensen
- Department of Clinical Genetics, Skejby Hospital, Aarhus, Denmark
| | | | - Torben A Kruse
- Department of Clinical Genetics, Odense University Hospital, Denmark
| | - Georgia Chenevix-Trench
- Genetics and Population Health Division, Queensland Institute of Medical Research, 300 Herston Rd, Herston, Brisbane, QLD 4006, Australia
| | - Fergus J Couch
- Department of Laboratory Medicine and Pathology, and Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Jacques Simard
- Cancer Genomics Laboratory, Centre Hospitalier Universitaire de Québec, 2705 Laurier Boulevard, T3-57, Quebec City and Canada Research Chair in Oncogenetics, Department of Molecular Medicine, Faculty of Medicine, Laval University, QC, Canada
| | - Douglas F Easton
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Worts Causeway, Cambridge CB1 8RN, UK
| |
Collapse
|
48
|
Evans DG, Howell A. Are We Ready for Online Tools in Decision Making for BRCA1/2 Mutation Carriers? J Clin Oncol 2012; 30:471-3. [DOI: 10.1200/jco.2011.40.1562] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Affiliation(s)
- D. Gareth Evans
- The University of Manchester, Manchester Academic Health Science Centre, Central Manchester University Hospitals Foundation Trust, St. Mary's Hospital; and Genesis Prevention Centre, University Hospital of South Manchester, Wythenshawe, Manchester, United Kingdom
| | - Anthony Howell
- Genesis Prevention Centre, University Hospital of South Manchester, Wythenshawe, Manchester, United Kingdom
| |
Collapse
|
49
|
|
50
|
|