Your browser doesn't support javascript.
loading
Genetically proxied glucose-lowering drug target perturbation and risk of cancer: a Mendelian randomisation analysis.
Yarmolinsky, James; Bouras, Emmanouil; Constantinescu, Andrei; Burrows, Kimberley; Bull, Caroline J; Vincent, Emma E; Martin, Richard M; Dimopoulou, Olympia; Lewis, Sarah J; Moreno, Victor; Vujkovic, Marijana; Chang, Kyong-Mi; Voight, Benjamin F; Tsao, Philip S; Gunter, Marc J; Hampe, Jochen; Pellatt, Andrew J; Pharoah, Paul D P; Schoen, Robert E; Gallinger, Steven; Jenkins, Mark A; Pai, Rish K; Gill, Dipender; Tsilidis, Kostas K.
Afiliação
  • Yarmolinsky J; MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK. james.yarmolinsky@bristol.ac.uk.
  • Bouras E; Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK. james.yarmolinsky@bristol.ac.uk.
  • Constantinescu A; Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece.
  • Burrows K; MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK.
  • Bull CJ; Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK.
  • Vincent EE; MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK.
  • Martin RM; Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK.
  • Dimopoulou O; MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK.
  • Lewis SJ; Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK.
  • Moreno V; School of Translational Health Sciences, University of Bristol, Bristol, UK.
  • Vujkovic M; MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK.
  • Chang KM; Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK.
  • Voight BF; School of Translational Health Sciences, University of Bristol, Bristol, UK.
  • Tsao PS; MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK.
  • Gunter MJ; Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK.
  • Hampe J; NIHR Bristol Biomedical Research Centre, University Hospitals Bristol and Weston NHS Foundation Trust and the University of Bristol, Bristol, UK.
  • Pellatt AJ; MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK.
  • Pharoah PDP; Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK.
  • Schoen RE; MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK.
  • Gallinger S; Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK.
  • Jenkins MA; Biomarkers and Susceptibility Unit, Oncology Data Analytics Program, Catalan Institute of Oncology (ICO), L'Hospitalet de Llobregat, Barcelona, Spain.
  • Pai RK; Colorectal Cancer Group, ONCOBELL Program, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain.
  • Gill D; Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA.
  • Tsilidis KK; Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
Diabetologia ; 66(8): 1481-1500, 2023 08.
Article em En | MEDLINE | ID: mdl-37171501
ABSTRACT
AIMS/

HYPOTHESIS:

Epidemiological studies have generated conflicting findings on the relationship between glucose-lowering medication use and cancer risk. Naturally occurring variation in genes encoding glucose-lowering drug targets can be used to investigate the effect of their pharmacological perturbation on cancer risk.

METHODS:

We developed genetic instruments for three glucose-lowering drug targets (peroxisome proliferator activated receptor γ [PPARG]; sulfonylurea receptor 1 [ATP binding cassette subfamily C member 8 (ABCC8)]; glucagon-like peptide 1 receptor [GLP1R]) using summary genetic association data from a genome-wide association study of type 2 diabetes in 148,726 cases and 965,732 controls in the Million Veteran Program. Genetic instruments were constructed using cis-acting genome-wide significant (p<5×10-8) SNPs permitted to be in weak linkage disequilibrium (r2<0.20). Summary genetic association estimates for these SNPs were obtained from genome-wide association study (GWAS) consortia for the following cancers breast (122,977 cases, 105,974 controls); colorectal (58,221 cases, 67,694 controls); prostate (79,148 cases, 61,106 controls); and overall (i.e. site-combined) cancer (27,483 cases, 372,016 controls). Inverse-variance weighted random-effects models adjusting for linkage disequilibrium were employed to estimate causal associations between genetically proxied drug target perturbation and cancer risk. Co-localisation analysis was employed to examine robustness of findings to violations of Mendelian randomisation (MR) assumptions. A Bonferroni correction was employed as a heuristic to define associations from MR analyses as 'strong' and 'weak' evidence.

RESULTS:

In MR analysis, genetically proxied PPARG perturbation was weakly associated with higher risk of prostate cancer (for PPARG perturbation equivalent to a 1 unit decrease in inverse rank normal transformed HbA1c OR 1.75 [95% CI 1.07, 2.85], p=0.02). In histological subtype-stratified analyses, genetically proxied PPARG perturbation was weakly associated with lower risk of oestrogen receptor-positive breast cancer (OR 0.57 [95% CI 0.38, 0.85], p=6.45×10-3). In co-localisation analysis, however, there was little evidence of shared causal variants for type 2 diabetes liability and cancer endpoints in the PPARG locus, although these analyses were likely underpowered. There was little evidence to support associations between genetically proxied PPARG perturbation and colorectal or overall cancer risk or between genetically proxied ABCC8 or GLP1R perturbation with risk across cancer endpoints. CONCLUSIONS/

INTERPRETATION:

Our drug target MR analyses did not find consistent evidence to support an association of genetically proxied PPARG, ABCC8 or GLP1R perturbation with breast, colorectal, prostate or overall cancer risk. Further evaluation of these drug targets using alternative molecular epidemiological approaches may help to further corroborate the findings presented in this analysis. DATA

AVAILABILITY:

Summary genetic association data for select cancer endpoints were obtained from the public domain breast cancer ( https//bcac.ccge.medschl.cam.ac.uk/bcacdata/ ); and overall prostate cancer ( http//practical.icr.ac.uk/blog/ ). Summary genetic association data for colorectal cancer can be accessed by contacting GECCO (kafdem at fredhutch.org). Summary genetic association data on advanced prostate cancer can be accessed by contacting PRACTICAL (practical at icr.ac.uk). Summary genetic association data on type 2 diabetes from Vujkovic et al (Nat Genet, 2020) can be accessed through dbGAP under accession number phs001672.v3.p1 (pha004945.1 refers to the European-specific summary statistics). UK Biobank data can be accessed by registering with UK Biobank and completing the registration form in the Access Management System (AMS) ( https//www.ukbiobank.ac.uk/enable-your-research/apply-for-access ).
Assuntos
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Neoplasias da Próstata / Neoplasias da Mama / Neoplasias Colorretais / Diabetes Mellitus Tipo 2 Idioma: En Ano de publicação: 2023 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Neoplasias da Próstata / Neoplasias da Mama / Neoplasias Colorretais / Diabetes Mellitus Tipo 2 Idioma: En Ano de publicação: 2023 Tipo de documento: Article