Your browser doesn't support javascript.
loading
Gene-environment interactions involving functional variants: Results from the Breast Cancer Association Consortium.
Barrdahl, Myrto; Rudolph, Anja; Hopper, John L; Southey, Melissa C; Broeks, Annegien; Fasching, Peter A; Beckmann, Matthias W; Gago-Dominguez, Manuela; Castelao, J Esteban; Guénel, Pascal; Truong, Thérèse; Bojesen, Stig E; Gapstur, Susan M; Gaudet, Mia M; Brenner, Hermann; Arndt, Volker; Brauch, Hiltrud; Hamann, Ute; Mannermaa, Arto; Lambrechts, Diether; Jongen, Lynn; Flesch-Janys, Dieter; Thoene, Kathrin; Couch, Fergus J; Giles, Graham G; Simard, Jacques; Goldberg, Mark S; Figueroa, Jonine; Michailidou, Kyriaki; Bolla, Manjeet K; Dennis, Joe; Wang, Qin; Eilber, Ursula; Behrens, Sabine; Czene, Kamila; Hall, Per; Cox, Angela; Cross, Simon; Swerdlow, Anthony; Schoemaker, Minouk J; Dunning, Alison M; Kaaks, Rudolf; Pharoah, Paul D P; Schmidt, Marjanka; Garcia-Closas, Montserrat; Easton, Douglas F; Milne, Roger L; Chang-Claude, Jenny.
Afiliação
  • Barrdahl M; Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany.
  • Rudolph A; Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany.
  • Hopper JL; Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, VIC, Australia.
  • Southey MC; Department of Pathology, The University of Melbourne, Melbourne, VIC, Australia.
  • Broeks A; Division of Molecular Pathology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands.
  • Fasching PA; Department of Gynaecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany.
  • Beckmann MW; David Geffen School of Medicine, Department of Medicine Division of Hematology and Oncology, University of California at Los Angeles, Los Angeles, CA.
  • Gago-Dominguez M; Department of Gynaecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany.
  • Castelao JE; Genomic Medicine Group, Galician Foundation of Genomic Medicine, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complejo Hospitalario Universitario de Santiago, Servicio Galego de Saúde, SERGAS, Santiago De Compostela, Spain.
  • Guénel P; Moores Cancer Center, University of California San Diego, La Jolla, CA.
  • Truong T; Oncology and Genetics Unit, Instituto de Investigación Sanitaria Galicia Sur (IISGS), Xerencia de Xestion Integrada de Vigo-SERGAS, Vigo, Spain.
  • Bojesen SE; CESP-Cancer and Environment team, INSERM U1018, Université Paris-Sud, Université Paris-Saclay, Villejuif, France.
  • Gapstur SM; CESP-Cancer and Environment team, INSERM U1018, Université Paris-Sud, Université Paris-Saclay, Villejuif, France.
  • Gaudet MM; Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark.
  • Brenner H; Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark.
  • Arndt V; Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
  • Brauch H; Epidemiology Research Program, American Cancer Society, Atlanta, GA.
  • Hamann U; Epidemiology Research Program, American Cancer Society, Atlanta, GA.
  • Mannermaa A; Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany.
  • Lambrechts D; Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany.
  • Jongen L; German Cancer Consortium (DKTK), Heidelberg, Germany.
  • Flesch-Janys D; Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany.
  • Thoene K; German Cancer Consortium (DKTK), Heidelberg, Germany.
  • Couch FJ; Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany.
  • Giles GG; University of Tübingen, Tübingen, Germany.
  • Simard J; Molecular Epidemiology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany.
  • Goldberg MS; Translational Cancer Research Area, University of Eastern Finland, Kuopio, Finland.
  • Figueroa J; Pathology and Forensic Medicine, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland.
  • Michailidou K; Imaging Center, Department of Clinical Pathology, Kuopio University Hospital, Kuopio, Finland.
  • Bolla MK; Vesalius Research Center, VIB, Leuven, Belgium.
  • Dennis J; Laboratory for Translational Genetics, Department of Human Genetics, University of Leuven, Leuven, Belgium.
  • Wang Q; Leuven Multidisciplinary Breast Center, Department of Oncology, KU Leuven and Leuven Cancer Institute, University Hospitals Leuven, Leuven, Belgium.
  • Eilber U; Institute for Medical Biometrics and Epidemiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
  • Behrens S; Department of Cancer Epidemiology, University Cancer Center Hamburg (UCCH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
  • Czene K; Department of Cancer Epidemiology, University Cancer Center Hamburg (UCCH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
  • Hall P; Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN.
  • Cox A; Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, VIC, Australia.
  • Cross S; Cancer Epidemiology and Intelligence Division, Cancer Council Victoria, Melbourne, VIC, Australia.
  • Swerdlow A; Genomics Center, Centre Hospitalier Universitaire de Québec Research Center, Laval University, Québec City, QC, Canada.
  • Schoemaker MJ; Department of Medicine, McGill University, Montréal, QC, Canada.
  • Dunning AM; Division of Clinical Epidemiology, Royal Victoria Hospital, McGill University, Montréal, QC, Canada.
  • Kaaks R; Usher Institute of Population Health Sciences and Informatics, The University of Edinburgh Medical School, Teviot Place Edinburgh, Edinburgh, United Kingdom.
  • Pharoah PDP; Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD.
  • Schmidt M; Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Worts Causeway, Cambridge, United Kingdom.
  • Garcia-Closas M; Department of Electron Microscopy/Molecular Pathology, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus, Nicosia.
  • Easton DF; Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Worts Causeway, Cambridge, United Kingdom.
  • Milne RL; Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Worts Causeway, Cambridge, United Kingdom.
  • Chang-Claude J; Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Worts Causeway, Cambridge, United Kingdom.
Int J Cancer ; 141(9): 1830-1840, 2017 11 01.
Article em En | MEDLINE | ID: mdl-28670784
ABSTRACT
Investigating the most likely causal variants identified by fine-mapping analyses may improve the power to detect gene-environment interactions. We assessed the interplay between 70 single nucleotide polymorphisms identified by genetic fine-scale mapping of susceptibility loci and 11 epidemiological breast cancer risk factors in relation to breast cancer. Analyses were conducted on up to 58,573 subjects (26,968 cases and 31,605 controls) from the Breast Cancer Association Consortium, in one of the largest studies of its kind. Analyses were carried out separately for estrogen receptor (ER) positive (ER+) and ER negative (ER-) disease. The Bayesian False Discovery Probability (BFDP) was computed to assess the noteworthiness of the results. Four potential gene-environment interactions were identified as noteworthy (BFDP < 0.80) when assuming a true prior interaction probability of 0.01. The strongest interaction result in relation to overall breast cancer risk was found between CFLAR-rs7558475 and current smoking (ORint = 0.77, 95% CI 0.67-0.88, pint = 1.8 × 10-4 ). The interaction with the strongest statistical evidence was found between 5q14-rs7707921 and alcohol consumption (ORint =1.36, 95% CI 1.16-1.59, pint = 1.9 × 10-5 ) in relation to ER- disease risk. The remaining two gene-environment interactions were also identified in relation to ER- breast cancer risk and were found between 3p21-rs6796502 and age at menarche (ORint = 1.26, 95% CI 1.12-1.43, pint =1.8 × 10-4 ) and between 8q23-rs13267382 and age at first full-term pregnancy (ORint = 0.89, 95% CI 0.83-0.95, pint = 5.2 × 10-4 ). While these results do not suggest any strong gene-environment interactions, our results may still be useful to inform experimental studies. These may in turn, shed light on the potential interactions observed.
Assuntos
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Neoplasias da Mama / Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD / Estudos de Associação Genética / Interação Gene-Ambiente Idioma: En Ano de publicação: 2017 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Neoplasias da Mama / Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD / Estudos de Associação Genética / Interação Gene-Ambiente Idioma: En Ano de publicação: 2017 Tipo de documento: Article