Your browser doesn't support javascript.
loading
Germline variation in the insulin-like growth factor pathway and risk of Barrett's esophagus and esophageal adenocarcinoma.
Dighe, Shruti G; Chen, Jianhong; Yan, Li; He, Qianchuan; Gharahkhani, Puya; Onstad, Lynn; Levine, David M; Palles, Claire; Ye, Weimin; Gammon, Marilie D; Iyer, Prasad G; Anderson, Lesley A; Liu, Geoffrey; Wu, Anna H; Dai, James Y; Chow, Wong-Ho; Risch, Harvey A; Lagergren, Jesper; Shaheen, Nicholas J; Bernstein, Leslie; Corley, Douglas A; Prenen, Hans; deCaestecker, John; MacDonald, David; Moayyedi, Paul; Barr, Hugh; Love, Sharon B; Chegwidden, Laura; Attwood, Stephen; Watson, Peter; Harrison, Rebecca; Ott, Katja; Moebus, Susanne; Venerito, Marino; Lang, Hauke; Mayershofer, Rupert; Knapp, Michael; Veits, Lothar; Gerges, Christian; Weismüller, Josef; Gockel, Ines; Vashist, Yogesh; Nöthen, Markus M; Izbicki, Jakob R; Manner, Hendrik; Neuhaus, Horst; Rösch, Thomas; Böhmer, Anne C; Hölscher, Arnulf H; Anders, Mario.
Afiliación
  • Dighe SG; Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA.
  • Chen J; Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA.
  • Yan L; Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA.
  • He Q; Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.
  • Gharahkhani P; Statistical Genetics, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia.
  • Onstad L; Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.
  • Levine DM; Department of Biostatistics, University of Washington, School of Public Health, Seattle, WA, USA.
  • Palles C; Gastrointestinal Cancer Genetics Laboratory, Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK.
  • Ye W; Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.
  • Gammon MD; Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA.
  • Iyer PG; Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA.
  • Anderson LA; Department of Epidemiology and Public Health, Queen's University of Belfast, Royal Group of Hospitals, Belfast, UK.
  • Liu G; Department of Pharmacogenomic Epidemiology, Ontario Cancer Institute, Toronto, Ontario, Canada.
  • Wu AH; Department of Preventive Medicine, University of Southern California/Norris Comprehensive Cancer Center, Los Angeles, CA, USA.
  • Dai JY; Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.
  • Chow WH; Department of Epidemiology, MD Anderson Cancer Center, Houston, TX, USA.
  • Risch HA; Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, CT, USA.
  • Lagergren J; Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.
  • Shaheen NJ; Department of Surgery, School of Cancer and Pharmaceutical Sciences, King's College London.
  • Bernstein L; Division of Gastroenterology and Hepatology, University of North Carolina School of Medicine, University of North Carolina, Chapel Hill, NC, USA.
  • Corley DA; Department of Population Sciences, Beckman Research Institute and City of Hope Comprehensive Cancer Center, Duarte, CA, USA.
  • Prenen H; Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA.
  • deCaestecker J; Gastroenterology, San Francisco Medical Center, Kaiser Permanente Northern California, San Francisco, California, USA.
  • MacDonald D; Oncology Department, University Hospital Antwerp, Edegem, Belgium.
  • Moayyedi P; Digestive Diseases Centre, University Hospitals of Leicester, Leicester, UK.
  • Barr H; Department of Oral Biological and Medical Sciences, University of British Columbia, Vancouver, British Columbia, Canada.
  • Love SB; Farncombe Family Digestive Health Research Institute, Department of Medicine, McMaster University, Hamilton, Ontario, Canada.
  • Chegwidden L; Department of Upper GI Surgery, Gloucestershire Royal Hospital, Gloucester, UK.
  • Attwood S; Centre for Statistics in Medicine, University of Oxford, Oxford, UK; MRC Clinical Trials Unit at University College London, London, UK.
  • Watson P; University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK.
  • Harrison R; Department of General Surgery, North Tyneside General Hospital, North Shields, UK.
  • Ott K; Department of Medicine, Institute of Clinical Science, Royal Victoria Hospital, Belfast, UK.
  • Moebus S; Department of Pathology, Leicester Royal Infirmary, Leicester, UK.
  • Venerito M; Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany.
  • Lang H; Department of General, Visceral and Thorax Surgery, RoMed Klinikum Rosenheim, Rosenheim, Germany.
  • Mayershofer R; Biometry and Epidemiology, Institute for Urban Public Health, University Hospitals, University of Duisburg-Essen, Essen, Germany.
  • Knapp M; Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Hospital, Magdeburg, Germany.
  • Veits L; Department of General, Visceral and Transplant Surgery, University Medical Center, University of Mainz, Mainz, Germany.
  • Gerges C; Gastroenterologie am Burgweiher, Bonn, Germany.
  • Weismüller J; Institute for Medical Biometry, Informatics, and Epidemiology, University of Bonn, Bonn, Germany.
  • Gockel I; Institute of Pathology, Klinikum Bayreuth, Bayreuth, Germany.
  • Vashist Y; Department of Internal Medicine, Evangelisches Krankenhaus, Düsseldorf, Germany.
  • Nöthen MM; Gastroenterologische Gemeinschaftspraxis, Koblenz, Germany.
  • Izbicki JR; Department of Visceral, Transplant, Thoracic and Vascular Surgery, University Hospital of Leipzig, Leipzig, Germany.
  • Manner H; Department of Surgery, Asklepios Harzklinik Goslar, Goslar, Germany.
  • Neuhaus H; Institute of Human Genetics, Medical Faculty, University of Bonn, Bonn, Germany.
  • Rösch T; General, Visceral and Thoracic Surgery Department and Clinic, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
  • Böhmer AC; Department of Internal Medicine II, Frankfurt Hoechst Hospital, Frankfurt, Germany.
  • Hölscher AH; Department of Internal Medicine, Evangelisches Krankenhaus, Düsseldorf, Germany.
  • Anders M; Department of Interdisciplinary Endoscopy, University Hospital Hamburg-Eppendorf, Hamburg, Germany.
Carcinogenesis ; 42(3): 369-377, 2021 04 17.
Article en En | MEDLINE | ID: mdl-33300568
ABSTRACT
Genome-wide association studies (GWAS) of esophageal adenocarcinoma (EAC) and its precursor, Barrett's esophagus (BE), have uncovered significant genetic components of risk, but most heritability remains unexplained. Targeted assessment of genetic variation in biologically relevant pathways using novel analytical approaches may identify missed susceptibility signals. Central obesity, a key BE/EAC risk factor, is linked to systemic inflammation, altered hormonal signaling and insulin-like growth factor (IGF) axis dysfunction. Here, we assessed IGF-related genetic variation and risk of BE and EAC. Principal component analysis was employed to evaluate pathway-level and gene-level associations with BE/EAC, using genotypes for 270 single-nucleotide polymorphisms (SNPs) in or near 12 IGF-related genes, ascertained from 3295 BE cases, 2515 EAC cases and 3207 controls in the Barrett's and Esophageal Adenocarcinoma Consortium (BEACON) GWAS. Gene-level signals were assessed using Multi-marker Analysis of GenoMic Annotation (MAGMA) and SNP summary statistics from BEACON and an expanded GWAS meta-analysis (6167 BE cases, 4112 EAC cases, 17 159 controls). Global variation in the IGF pathway was associated with risk of BE (P = 0.0015). Gene-level associations with BE were observed for GHR (growth hormone receptor; P = 0.00046, false discovery rate q = 0.0056) and IGF1R (IGF1 receptor; P = 0.0090, q = 0.0542). These gene-level signals remained significant at q < 0.1 when assessed using data from the largest available BE/EAC GWAS meta-analysis. No significant associations were observed for EAC. This study represents the most comprehensive evaluation to date of inherited genetic variation in the IGF pathway and BE/EAC risk, providing novel evidence that variation in two genes encoding cell-surface receptors, GHR and IGF1R, may influence risk of BE.
Asunto(s)

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: Esófago de Barrett / Somatomedinas / Neoplasias Esofágicas / Adenocarcinoma / Biomarcadores de Tumor Tipo de estudio: Etiology_studies / Observational_studies / Prognostic_studies / Risk_factors_studies / Systematic_reviews Límite: Aged / Female / Humans / Male / Middle aged Idioma: En Revista: Carcinogenesis Año: 2021 Tipo del documento: Article País de afiliación: Estados Unidos

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: Esófago de Barrett / Somatomedinas / Neoplasias Esofágicas / Adenocarcinoma / Biomarcadores de Tumor Tipo de estudio: Etiology_studies / Observational_studies / Prognostic_studies / Risk_factors_studies / Systematic_reviews Límite: Aged / Female / Humans / Male / Middle aged Idioma: En Revista: Carcinogenesis Año: 2021 Tipo del documento: Article País de afiliación: Estados Unidos