Clinical Pathology - Research Publications

Permanent URI for this collection

Search Results

Now showing 1 - 10 of 47
  • Item
    Thumbnail Image
    Association between circulating inflammatory markers and adult cancer risk: a Mendelian randomization analysis.
    Yarmolinsky, J ; Robinson, JW ; Mariosa, D ; Karhunen, V ; Huang, J ; Dimou, N ; Murphy, N ; Burrows, K ; Bouras, E ; Smith-Byrne, K ; Lewis, SJ ; Galesloot, TE ; Kiemeney, LA ; Vermeulen, S ; Martin, P ; Albanes, D ; Hou, L ; Newcomb, PA ; White, E ; Wolk, A ; Wu, AH ; Le Marchand, L ; Phipps, AI ; Buchanan, DD ; International Lung Cancer Consortium, ; PRACTICAL Consortium, ; Zhao, SS ; Gill, D ; Chanock, SJ ; Purdue, MP ; Davey Smith, G ; Brennan, P ; Herzig, K-H ; Järvelin, M-R ; Amos, CI ; Hung, RJ ; Dehghan, A ; Johansson, M ; Gunter, MJ ; Tsilidis, KK ; Martin, RM (Elsevier BV, 2024-02)
    BACKGROUND: Tumour-promoting inflammation is a "hallmark" of cancer and conventional epidemiological studies have reported links between various inflammatory markers and cancer risk. The causal nature of these relationships and, thus, the suitability of these markers as intervention targets for cancer prevention is unclear. METHODS: We meta-analysed 6 genome-wide association studies of circulating inflammatory markers comprising 59,969 participants of European ancestry. We then used combined cis-Mendelian randomization and colocalisation analysis to evaluate the causal role of 66 circulating inflammatory markers in risk of 30 adult cancers in 338,294 cancer cases and up to 1,238,345 controls. Genetic instruments for inflammatory markers were constructed using genome-wide significant (P < 5.0 Ã— 10-8) cis-acting SNPs (i.e., in or ±250 kb from the gene encoding the relevant protein) in weak linkage disequilibrium (LD, r2 < 0.10). Effect estimates were generated using inverse-variance weighted random-effects models and standard errors were inflated to account for weak LD between variants with reference to the 1000 Genomes Phase 3 CEU panel. A false discovery rate (FDR)-corrected P-value ("q-value") <0.05 was used as a threshold to define "strong evidence" to support associations and 0.05 ≤ q-value < 0.20 to define "suggestive evidence". A colocalisation posterior probability (PPH4) >70% was employed to indicate support for shared causal variants across inflammatory markers and cancer outcomes. Findings were replicated in the FinnGen study and then pooled using meta-analysis. FINDINGS: We found strong evidence to support an association of genetically-proxied circulating pro-adrenomedullin concentrations with increased breast cancer risk (OR: 1.19, 95% CI: 1.10-1.29, q-value = 0.033, PPH4 = 84.3%) and suggestive evidence to support associations of interleukin-23 receptor concentrations with increased pancreatic cancer risk (OR: 1.42, 95% CI: 1.20-1.69, q-value = 0.055, PPH4 = 73.9%), prothrombin concentrations with decreased basal cell carcinoma risk (OR: 0.66, 95% CI: 0.53-0.81, q-value = 0.067, PPH4 = 81.8%), and interleukin-1 receptor-like 1 concentrations with decreased triple-negative breast cancer risk (OR: 0.92, 95% CI: 0.88-0.97, q-value = 0.15, PPH4 = 85.6%). These findings were replicated in pooled analyses with the FinnGen study. Though suggestive evidence was found to support an association of macrophage migration inhibitory factor concentrations with increased bladder cancer risk (OR: 2.46, 95% CI: 1.48-4.10, q-value = 0.072, PPH4 = 76.1%), this finding was not replicated when pooled with the FinnGen study. For 22 of 30 cancer outcomes examined, there was little evidence (q-value ≥0.20) that any of the 66 circulating inflammatory markers examined were associated with cancer risk. INTERPRETATION: Our comprehensive joint Mendelian randomization and colocalisation analysis of the role of circulating inflammatory markers in cancer risk identified potential roles for 4 circulating inflammatory markers in risk of 4 site-specific cancers. Contrary to reports from some prior conventional epidemiological studies, we found little evidence of association of circulating inflammatory markers with the majority of site-specific cancers evaluated. FUNDING: Cancer Research UK (C68933/A28534, C18281/A29019, PPRCPJT∖100005), World Cancer Research Fund (IIG_FULL_2020_022), National Institute for Health Research (NIHR202411, BRC-1215-20011), Medical Research Council (MC_UU_00011/1, MC_UU_00011/3, MC_UU_00011/6, and MC_UU_00011/4), Academy of Finland Project 326291, European Union's Horizon 2020 grant agreement no. 848158 (EarlyCause), French National Cancer Institute (INCa SHSESP20, 2020-076), Versus Arthritis (21173, 21754, 21755), National Institutes of Health (U19 CA203654), National Cancer Institute (U19CA203654).
  • Item
    No Preview Available
    Deciphering colorectal cancer genetics through multi-omic analysis of 100,204 cases and 154,587 controls of European and east Asian ancestries
    Fernandez-Rozadilla, C ; Timofeeva, M ; Chen, Z ; Law, P ; Thomas, M ; Bien, S ; Diez-Obrero, V ; Li, L ; Fernandez-Tajes, J ; Palles, C ; Sherwood, K ; Harris, S ; Svinti, V ; McDonnell, K ; Farrington, S ; Studd, J ; Vaughan-Shaw, P ; Shu, X-O ; Long, J ; Cai, Q ; Guo, X ; Lu, Y ; Scacheri, P ; Studd, J ; Huyghe, J ; Harrison, T ; Shibata, D ; Haiman, C ; Devall, M ; Schumacher, F ; Melas, M ; Rennert, G ; Obon-Santacana, M ; Martin-Sanchez, V ; Moratalla-Navarro, F ; Oh, JH ; Kim, J ; Jee, SH ; Jung, KJ ; Kweon, S-S ; Shin, M-H ; Shin, A ; Ahn, Y-O ; Kim, D-H ; Oze, I ; Wen, W ; Matsuo, K ; Matsuda, K ; Tanikawa, C ; Ren, Z ; Gao, Y-T ; Jia, W-H ; Potter, J ; Jenkins, M ; Win, AK ; Pai, R ; Figueiredo, J ; Haile, R ; Gallinger, S ; Woods, M ; Newcomb, P ; Shibata, D ; Cheadle, J ; Kaplan, R ; Maughan, T ; Kerr, R ; Kerr, D ; Kirac, I ; Boehm, J ; Mecklin, L-P ; Jousilahti, P ; Knekt, P ; Aaltonen, L ; Rissanen, H ; Pukkala, E ; Eriksson, J ; Cajuso, T ; Hanninen, U ; Kondelin, J ; Palin, K ; Tanskanen, T ; Renkonen-Sinisalo, L ; Zanke, B ; Mannisto, S ; Albanes, D ; Weinstein, S ; Ruiz-Narvaez, E ; Palmer, J ; Buchanan, D ; Platz, E ; Visvanathan, K ; Ulrich, C ; Siegel, E ; Brezina, S ; Gsur, A ; Campbell, P ; Chang-Claude, J ; Hoffmeister, M ; Brenner, H ; Slattery, M ; Potter, J ; Tsilidis, K ; Schulze, M ; Gunter, M ; Murphy, N ; Castells, A ; Castellvi-Bel, S ; Moreira, L ; Arndt, V ; Shcherbina, A ; Stern, M ; Pardamean, B ; Bishop, T ; Giles, G ; Southey, M ; Idos, G ; McDonnell, K ; Abu-Ful, Z ; Greenson, J ; Shulman, K ; Lejbkowicz, F ; Offit, K ; Su, Y-R ; Steinfelder, R ; Keku, T ; van Guelpen, B ; Hudson, T ; Hampel, H ; Pearlman, R ; Berndt, S ; Hayes, R ; Martinez, ME ; Thomas, S ; Corley, D ; Pharoah, P ; Larsson, S ; Yen, Y ; Lenz, H-J ; White, E ; Li, L ; Doheny, K ; Pugh, E ; Shelford, T ; Chan, A ; Cruz-Correa, M ; Lindblom, A ; Shibata, D ; Joshi, A ; Schafmayer, C ; Scacheri, P ; Kundaje, A ; Nickerson, D ; Schoen, R ; Hampe, J ; Stadler, Z ; Vodicka, P ; Vodickova, L ; Vymetalkova, V ; Papadopoulos, N ; Edlund, C ; Gauderman, W ; Thomas, D ; Shibata, D ; Toland, A ; Markowitz, S ; Kim, A ; Gruber, S ; van Duijnhoven, F ; Feskens, E ; Sakoda, L ; Gago-Dominguez, M ; Wolk, A ; Naccarati, A ; Pardini, B ; FitzGerald, L ; Lee, SC ; Ogino, S ; Bien, S ; Kooperberg, C ; Li, C ; Lin, Y ; Prentice, R ; Qu, C ; Bezieau, S ; Tangen, C ; Mardis, E ; Yamaji, T ; Sawada, N ; Iwasaki, M ; Haiman, C ; Le Marchand, L ; Wu, A ; Qu, C ; McNeil, C ; Coetzee, G ; Hayward, C ; Deary, I ; Harris, S ; Theodoratou, E ; Reid, S ; Walker, M ; Ooi, LY ; Moreno, V ; Casey, G ; Gruber, S ; Tomlinson, I ; Zheng, W ; Dunlop, M ; Houlston, R ; Peters, U (NATURE PORTFOLIO, 2023-01)
    Colorectal cancer (CRC) is a leading cause of mortality worldwide. We conducted a genome-wide association study meta-analysis of 100,204 CRC cases and 154,587 controls of European and east Asian ancestry, identifying 205 independent risk associations, of which 50 were unreported. We performed integrative genomic, transcriptomic and methylomic analyses across large bowel mucosa and other tissues. Transcriptome- and methylome-wide association studies revealed an additional 53 risk associations. We identified 155 high-confidence effector genes functionally linked to CRC risk, many of which had no previously established role in CRC. These have multiple different functions and specifically indicate that variation in normal colorectal homeostasis, proliferation, cell adhesion, migration, immunity and microbial interactions determines CRC risk. Crosstissue analyses indicated that over a third of effector genes most probably act outside the colonic mucosa. Our findings provide insights into colorectal oncogenesis and highlight potential targets across tissues for new CRC treatment and chemoprevention strategies.
  • Item
    No Preview Available
    A Genetic Locus within the FMN1/GREM1 Gene Region Interacts with Body Mass Index in Colorectal Cancer Risk
    Aglago, EK ; Kim, A ; Lin, Y ; Qu, C ; Evangelou, M ; Ren, Y ; Morrison, J ; Albanes, D ; Arndt, V ; Barry, EL ; Baurley, JW ; Berndt, S ; Bien, SA ; Bishop, DT ; Bouras, E ; Brenner, H ; Buchanan, DD ; Budiarto, A ; Carreras-Torres, R ; Casey, G ; Cenggoro, TW ; Chen, AT ; Chang-Claude, J ; Chen, X ; Conti, D ; Devall, M ; Diez-Obrero, V ; Dimou, N ; Drew, D ; Figueiredo, JC ; Gallinger, S ; Giles, GG ; Gruber, SB ; Gsur, A ; Gunter, MJ ; Hampel, H ; Harlid, S ; Hidaka, A ; Harrison, TA ; Hoffmeister, M ; Huyghe, JR ; Jenkins, MA ; Jordahl, K ; Joshi, AD ; Kawaguchi, ES ; Keku, TO ; Kundaje, A ; Larsson, SC ; Le Marchand, L ; Lewinger, JP ; Li, L ; Lynch, BM ; Mahesworo, B ; Mandic, M ; Obon-Santacana, M ; Morento, V ; Murphy, N ; Men, H ; Nassir, R ; Newcomb, PA ; Ogino, S ; Ose, J ; Pai, RK ; Palmer, JR ; Papadimitriou, N ; Pardamean, B ; Peoples, AR ; Platz, EA ; Potter, JD ; Prentice, RL ; Rennert, G ; Ruiz-Narvaez, E ; Sakoda, LC ; Scacheri, PC ; Schmit, SL ; Schoen, RE ; Shcherbina, A ; Slattery, ML ; Stern, MC ; Su, Y-R ; Tangen, CM ; Thibodeau, SN ; Thomas, DC ; Tian, Y ; Ulrich, CM ; van Duijnhoven, FJB ; Van Guelpen, B ; Visvanathan, K ; Vodicka, P ; Wang, J ; White, E ; Wolk, A ; Woods, MO ; Wu, AH ; Zemlianskaia, N ; Hsu, L ; Gauderman, WJ ; Peters, U ; Tsilidis, KK ; Campbell, PT (AMER ASSOC CANCER RESEARCH, 2023-08-01)
    UNLABELLED: Colorectal cancer risk can be impacted by genetic, environmental, and lifestyle factors, including diet and obesity. Gene-environment interactions (G × E) can provide biological insights into the effects of obesity on colorectal cancer risk. Here, we assessed potential genome-wide G × E interactions between body mass index (BMI) and common SNPs for colorectal cancer risk using data from 36,415 colorectal cancer cases and 48,451 controls from three international colorectal cancer consortia (CCFR, CORECT, and GECCO). The G × E tests included the conventional logistic regression using multiplicative terms (one degree of freedom, 1DF test), the two-step EDGE method, and the joint 3DF test, each of which is powerful for detecting G × E interactions under specific conditions. BMI was associated with higher colorectal cancer risk. The two-step approach revealed a statistically significant G×BMI interaction located within the Formin 1/Gremlin 1 (FMN1/GREM1) gene region (rs58349661). This SNP was also identified by the 3DF test, with a suggestive statistical significance in the 1DF test. Among participants with the CC genotype of rs58349661, overweight and obesity categories were associated with higher colorectal cancer risk, whereas null associations were observed across BMI categories in those with the TT genotype. Using data from three large international consortia, this study discovered a locus in the FMN1/GREM1 gene region that interacts with BMI on the association with colorectal cancer risk. Further studies should examine the potential mechanisms through which this locus modifies the etiologic link between obesity and colorectal cancer. SIGNIFICANCE: This gene-environment interaction analysis revealed a genetic locus in FMN1/GREM1 that interacts with body mass index in colorectal cancer risk, suggesting potential implications for precision prevention strategies.
  • Item
    No Preview Available
    Elucidating the Risk of Colorectal Cancer for Variants in Hereditary Colorectal Cancer Genes
    Mahmood, K ; Thomas, M ; Qu, C ; Hsu, L ; Buchanan, DD ; Peters, U (W B SAUNDERS CO-ELSEVIER INC, 2023-10)
  • Item
    No Preview Available
    Validation of a Genetic-Enhanced Risk Prediction Model for Colorectal Cancer in a Large Community-Based Cohort.
    Su, Y-R ; Sakoda, LC ; Jeon, J ; Thomas, M ; Lin, Y ; Schneider, JL ; Udaltsova, N ; Lee, JK ; Lansdorp-Vogelaar, I ; Peterse, EFP ; Zauber, AG ; Zheng, J ; Zheng, Y ; Hauser, E ; Baron, JA ; Barry, EL ; Bishop, DT ; Brenner, H ; Buchanan, DD ; Burnett-Hartman, A ; Campbell, PT ; Casey, G ; Castellví-Bel, S ; Chan, AT ; Chang-Claude, J ; Figueiredo, JC ; Gallinger, SJ ; Giles, GG ; Gruber, SB ; Gsur, A ; Gunter, MJ ; Hampe, J ; Hampel, H ; Harrison, TA ; Hoffmeister, M ; Hua, X ; Huyghe, JR ; Jenkins, MA ; Keku, TO ; Marchand, LL ; Li, L ; Lindblom, A ; Moreno, V ; Newcomb, PA ; Pharoah, PDP ; Platz, EA ; Potter, JD ; Qu, C ; Rennert, G ; Schoen, RE ; Slattery, ML ; Song, M ; van Duijnhoven, FJB ; Van Guelpen, B ; Vodicka, P ; Wolk, A ; Woods, MO ; Wu, AH ; Hayes, RB ; Peters, U ; Corley, DA ; Hsu, L (American Association for Cancer Research (AACR), 2023-03-06)
    BACKGROUND: Polygenic risk scores (PRS) which summarize individuals' genetic risk profile may enhance targeted colorectal cancer screening. A critical step towards clinical implementation is rigorous external validations in large community-based cohorts. This study externally validated a PRS-enhanced colorectal cancer risk model comprising 140 known colorectal cancer loci to provide a comprehensive assessment on prediction performance. METHODS: The model was developed using 20,338 individuals and externally validated in a community-based cohort (n = 85,221). We validated predicted 5-year absolute colorectal cancer risk, including calibration using expected-to-observed case ratios (E/O) and calibration plots, and discriminatory accuracy using time-dependent AUC. The PRS-related improvement in AUC, sensitivity and specificity were assessed in individuals of age 45 to 74 years (screening-eligible age group) and 40 to 49 years with no endoscopy history (younger-age group). RESULTS: In European-ancestral individuals, the predicted 5-year risk calibrated well [E/O = 1.01; 95% confidence interval (CI), 0.91-1.13] and had high discriminatory accuracy (AUC = 0.73; 95% CI, 0.71-0.76). Adding the PRS to a model with age, sex, family and endoscopy history improved the 5-year AUC by 0.06 (P < 0.001) and 0.14 (P = 0.05) in the screening-eligible age and younger-age groups, respectively. Using a risk-threshold of 5-year SEER colorectal cancer incidence rate at age 50 years, adding the PRS had a similar sensitivity but improved the specificity by 11% (P < 0.001) in the screening-eligible age group. In the younger-age group it improved the sensitivity by 27% (P = 0.04) with similar specificity. CONCLUSIONS: The proposed PRS-enhanced model provides a well-calibrated 5-year colorectal cancer risk prediction and improves discriminatory accuracy in the external cohort. IMPACT: The proposed model has potential utility in risk-stratified colorectal cancer prevention.
  • Item
    No Preview Available
    Genome-Wide Interaction Analysis of Genetic Variants With Menopausal Hormone Therapy for Colorectal Cancer Risk
    Tian, Y ; Kim, AE ; Bien, SA ; Lin, Y ; Qu, C ; Harrison, TA ; Carreras-Torres, R ; Diez-Obrero, V ; Dimou, N ; Drew, DA ; Hidaka, A ; Huyghe, JR ; Jordahl, KM ; Morrison, J ; Murphy, N ; Obon-Santacana, M ; Ulrich, CM ; Ose, J ; Peoples, AR ; Ruiz-Narvaez, EA ; Shcherbina, A ; Stern, MC ; Su, Y-R ; van Duijnhoven, FJB ; Arndt, V ; Baurley, JW ; Berndt, S ; Bishop, DT ; Brenner, H ; Buchanan, DD ; Chan, AT ; Figueiredo, JC ; Gallinger, S ; Gruber, SB ; Harlid, S ; Hoffmeister, M ; Jenkins, MA ; Joshi, AD ; Keku, TO ; Larsson, SC ; Le Marchand, L ; Li, L ; Giles, GG ; Milne, RL ; Nan, H ; Nassir, R ; Ogino, S ; Budiarto, A ; Platz, EA ; Potter, JD ; Prentice, RL ; Rennert, G ; Sakoda, LC ; Schoen, RE ; Slattery, ML ; Thibodeau, SN ; Van Guelpen, B ; Visvanathan, K ; White, E ; Wolk, A ; Woods, MO ; Wu, AH ; Campbell, PT ; Casey, G ; Conti, D ; Gunter, MJ ; Kundaje, A ; Lewinger, JP ; Moreno, V ; Newcomb, PA ; Pardamean, B ; Thomas, DC ; Tsilidis, KK ; Peters, U ; Gauderman, WJ ; Hsu, L ; Chang-Claude, J (OXFORD UNIV PRESS INC, 2022-08-08)
    BACKGROUND: The use of menopausal hormone therapy (MHT) may interact with genetic variants to influence colorectal cancer (CRC) risk. METHODS: We conducted a genome-wide, gene-environment interaction between single nucleotide polymorphisms and the use of any MHT, estrogen only, and combined estrogen-progestogen therapy with CRC risk, among 28 486 postmenopausal women (11 519 CRC patients and 16 967 participants without CRC) from 38 studies, using logistic regression, 2-step method, and 2- or 3-degree-of-freedom joint test. A set-based score test was applied for rare genetic variants. RESULTS: The use of any MHT, estrogen only and estrogen-progestogen were associated with a reduced CRC risk (odds ratio [OR] = 0.71, 95% confidence interval [CI] = 0.64 to 0.78; OR = 0.65, 95% CI = 0.53 to 0.79; and OR = 0.73, 95% CI = 0.59 to 0.90, respectively). The 2-step method identified a statistically significant interaction between a GRIN2B variant rs117868593 and MHT use, whereby MHT-associated CRC risk was statistically significantly reduced in women with the GG genotype (OR = 0.68, 95% CI = 0.64 to 0.72) but not within strata of GC or CC genotypes. A statistically significant interaction between a DCBLD1 intronic variant at 6q22.1 (rs10782186) and MHT use was identified by the 2-degree-of-freedom joint test. The MHT-associated CRC risk was reduced with increasing number of rs10782186-C alleles, showing odds ratios of 0.78 (95% CI = 0.70 to 0.87) for TT, 0.68 (95% CI = 0.63 to 0.73) for TC, and 0.66 (95% CI = 0.60 to 0.74) for CC genotypes. In addition, 5 genes in rare variant analysis showed suggestive interactions with MHT (2-sided P < 1.2 × 10-4). CONCLUSION: Genetic variants that modify the association between MHT and CRC risk were identified, offering new insights into pathways of CRC carcinogenesis and potential mechanisms involved.
  • Item
    No Preview Available
    Does genetic predisposition modify the effect of lifestyle-related factors on DNA methylation?
    Yu, C ; Hodge, AM ; Wong, EM ; Joo, JE ; Makalic, E ; Schmidt, DF ; Buchanan, DD ; Severi, G ; Hopper, JL ; English, DR ; Giles, GG ; Milne, RL ; Southey, MC ; Dugue, P-A (TAYLOR & FRANCIS INC, 2022-12-02)
    Lifestyle-related phenotypes have been shown to be heritable and associated with DNA methylation. We aimed to investigate whether genetic predisposition to tobacco smoking, alcohol consumption, and higher body mass index (BMI) moderates the effect of these phenotypes on blood DNA methylation. We calculated polygenic scores (PGS) to quantify genetic predisposition to these phenotypes using training (N = 7,431) and validation (N = 4,307) samples. Using paired genetic-methylation data (N = 4,307), gene-environment interactions (i.e., PGS × lifestyle) were assessed using linear mixed-effects models with outcomes: 1) methylation at sites found to be strongly associated with smoking (1,061 CpGs), alcohol consumption (459 CpGs), and BMI (85 CpGs) and 2) two epigenetic ageing measures, PhenoAge and GrimAge. In the validation sample, PGS explained ~1.4% (P = 1 × 10-14), ~0.6% (P = 2 × 10-7), and ~8.7% (P = 7 × 10-87) of variance in smoking initiation, alcohol consumption, and BMI, respectively. Nominally significant interaction effects (P < 0.05) were found at 61, 14, and 7 CpGs for smoking, alcohol consumption, and BMI, respectively. There was strong evidence that all lifestyle-related phenotypes were positively associated with PhenoAge and GrimAge, except for alcohol consumption with PhenoAge. There was weak evidence that the association of smoking with GrimAge was attenuated in participants genetically predisposed to smoking (interaction term: -0.022, standard error [SE] = 0.012, P = 0.058) and that the association of alcohol consumption with PhenoAge was attenuated in those genetically predisposed to drink alcohol (interaction term: -0.030, SE = 0.015, P = 0.041). In conclusion, genetic susceptibility to unhealthy lifestyles did not strongly modify the association between observed lifestyle behaviour and blood DNA methylation. Potential associations were observed for epigenetic ageing measures, which should be replicated in additional studies.
  • Item
    Thumbnail Image
    Genetic variants associated with circulating C-reactive protein levels and colorectal cancer survival: Sex-specific and lifestyle factors specific associations
    Huang, Y ; Hua, X ; Labadie, JD ; Harrison, TA ; Dai, JY ; Lindstrom, S ; Lin, Y ; Berndt, S ; Buchanan, DD ; Campbell, PT ; Casey, G ; Gallinger, SJ ; Gunter, MJ ; Hoffmeister, M ; Jenkins, MA ; Sakoda, LC ; Schoen, RE ; Diergaarde, B ; Slattery, ML ; White, E ; Giles, G ; Brenner, H ; Chang-Claude, J ; Joshi, A ; Ma, W ; Pai, RK ; Chan, AT ; Peters, U ; Newcomb, PA (WILEY, 2022-05-01)
    Elevated blood levels of C-reactive protein (CRP) have been linked to colorectal cancer (CRC) survival. We evaluated genetic variants associated with CRP levels and their interactions with sex and lifestyle factors in association with CRC-specific mortality. Our study included 16 142 CRC cases from the International Survival Analysis in Colorectal Cancer Consortium. We identified 618 common single nucleotide polymorphisms (SNPs) associated with CRP levels from the NHGRI-EBI GWAS Catalog. Cox proportional hazards regression was used to estimate hazard ratios (HRs) and 95% confidence intervals (CIs) for associations between SNPs and CRC-specific mortality adjusting for age, sex, genotyping platform/study and principal components. We investigated their interactions with sex and lifestyle factors using likelihood ratio tests. Of 5472 (33.9%) deaths accrued over up to 10 years of follow-up, 3547 (64.8%) were due to CRC. No variants were associated with CRC-specific mortality after multiple comparison correction. We observed strong evidence of interaction between variant rs1933736 at FRK gene and sex in relation to CRC-specific mortality (corrected Pinteraction  = .0004); women had higher CRC-specific mortality associated with the minor allele (HR = 1.11, 95% CI = 1.04-1.19) whereas an inverse association was observed for men (HR = 0.88, 95% CI = 0.82-0.94). There was no evidence of interactions between CRP-associated SNPs and alcohol, obesity or smoking. Our study observed a significant interaction between sex and a CRP-associated variant in relation to CRC-specific mortality. Future replication of this association and functional annotation of the variant are needed.
  • Item
    No Preview Available
    Methylation scores for smoking, alcohol consumption and body mass index and risk of seven types of cancer
    Dugue, P-A ; Yu, C ; Hodge, AMM ; Wong, EM ; Joo, JEE ; Jung, C-H ; Schmidt, D ; Makalic, E ; Buchanan, DDD ; Severi, G ; English, DRR ; Hopper, JLL ; Milne, RLL ; Giles, GGG ; Southey, MCC (WILEY, 2023-08-01)
    Methylation marks of exposure to health risk factors may be useful markers of cancer risk as they might better capture current and past exposures than questionnaires, and reflect different individual responses to exposure. We used data from seven case-control studies nested within the Melbourne Collaborative Cohort Study of blood DNA methylation and risk of colorectal, gastric, kidney, lung, prostate and urothelial cancer, and B-cell lymphoma (N cases = 3123). Methylation scores (MS) for smoking, body mass index (BMI), and alcohol consumption were calculated based on published data as weighted averages of methylation values. Rate ratios (RR) and 95% confidence intervals for association with cancer risk were estimated using conditional logistic regression and expressed per SD increase of the MS, with and without adjustment for health-related confounders. The contribution of MS to discriminate cases from controls was evaluated using the area under the curve (AUC). After confounder adjustment, we observed: large associations (RR = 1.5-1.7) with lung cancer risk for smoking MS; moderate associations (RR = 1.2-1.3) with urothelial cancer risk for smoking MS and with mature B-cell neoplasm risk for BMI and alcohol MS; moderate to small associations (RR = 1.1-1.2) for BMI and alcohol MS with several cancer types and cancer overall. Generally small AUC increases were observed after inclusion of several MS in the same model (colorectal, gastric, kidney, urothelial cancers: +3%; lung cancer: +7%; B-cell neoplasms: +8%). Methylation scores for smoking, BMI and alcohol consumption show independent associations with cancer risk, and may provide some improvements in risk prediction.
  • Item
    Thumbnail Image
    Body mass index and molecular subtypes of colorectal cancer
    Murphy, N ; Newton, CC ; Song, M ; Papadimitriou, N ; Hoffmeister, M ; Phipps, A ; Harrison, TA ; Newcomb, PA ; Aglago, EK ; Berndt, S ; Brenner, H ; Buchanan, DD ; Cao, Y ; Chan, AT ; Chen, X ; Cheng, I ; Chang-Claude, J ; Dimou, N ; Drew, D ; Farris, AB ; French, AJ ; Gallinger, S ; Georgeson, P ; Giannakis, M ; Giles, GG ; Gruber, SB ; Harlid, S ; Hsu, L ; Huang, W-Y ; Jenkins, MA ; Laskar, RS ; Le Marchand, L ; Limburg, P ; Lin, Y ; Mandic, M ; Nowak, JA ; Obon-Santacana, M ; Ogino, S ; Qu, C ; Sakoda, LC ; Schoen, RE ; Southey, MC ; Stadler, ZK ; Steinfelder, RS ; Sun, W ; Thibodeau, SN ; Toland, AE ; Trinh, QM ; Tsilidis, KK ; Ugai, T ; Van Guelpen, B ; Wang, X ; Woods, MO ; Zaidi, SH ; Gunter, MJ ; Peters, U ; Campbell, PT (OXFORD UNIV PRESS INC, 2023-02)
    BACKGROUND: Obesity is an established risk factor for colorectal cancer (CRC), but the evidence for the association is inconsistent across molecular subtypes of the disease. METHODS: We pooled data on body mass index (BMI), tumor microsatellite instability status, CpG island methylator phenotype status, BRAF and KRAS mutations, and Jass classification types for 11 872 CRC cases and 11 013 controls from 11 observational studies. We used multinomial logistic regression to estimate odds ratios (OR) and 95% confidence intervals (CI) adjusted for covariables. RESULTS: Higher BMI was associated with increased CRC risk (OR per 5 kg/m2 = 1.18, 95% CI = 1.15 to 1.22). The positive association was stronger for men than women but similar across tumor subtypes defined by individual molecular markers. In analyses by Jass type, higher BMI was associated with elevated CRC risk for types 1-4 cases but not for type 5 CRC cases (considered familial-like/Lynch syndrome microsatellite instability-H, CpG island methylator phenotype-low or negative, BRAF-wild type, KRAS-wild type, OR = 1.04, 95% CI = 0.90 to 1.20). This pattern of associations for BMI and Jass types was consistent by sex and design of contributing studies (cohort or case-control). CONCLUSIONS: In contrast to previous reports with fewer study participants, we found limited evidence of heterogeneity for the association between BMI and CRC risk according to molecular subtype, suggesting that obesity influences nearly all major pathways involved in colorectal carcinogenesis. The null association observed for the Jass type 5 suggests that BMI is not a risk factor for the development of CRC for individuals with Lynch syndrome.