Clinical Pathology - Research Publications

Permanent URI for this collection

Search Results

Now showing 1 - 10 of 16
  • 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
    Thumbnail Image
    Interactions between folate intake and genetic predictors of gene expression levels associated with colorectal cancer risk
    Haas, CB ; Su, Y-R ; Petersen, P ; Wang, X ; Bien, SA ; Lin, Y ; Albanes, D ; Weinstein, SJ ; Jenkins, MA ; Figueiredo, JC ; Newcomb, PA ; Casey, G ; Le Marchand, L ; Campbell, PT ; Moreno, V ; Potter, JD ; Sakoda, LC ; Slattery, ML ; Chan, AT ; Li, L ; Giles, GG ; Milne, RL ; Gruber, SB ; Rennert, G ; Woods, MO ; Gallinger, SJ ; Berndt, S ; Hayes, RB ; Huang, W-Y ; Wolk, A ; White, E ; Nan, H ; Nassir, R ; Lindor, NM ; Lewinger, JP ; Kim, AE ; Conti, D ; Gauderman, WJ ; Buchanan, DD ; Peters, U ; Hsu, L (NATURE PORTFOLIO, 2022-11-07)
    Observational studies have shown higher folate consumption to be associated with lower risk of colorectal cancer (CRC). Understanding whether and how genetic risk factors interact with folate could further elucidate the underlying mechanism. Aggregating functionally relevant genetic variants in set-based variant testing has higher power to detect gene-environment (G × E) interactions and may provide information on the underlying biological pathway. We investigated interactions between folate consumption and predicted gene expression on colorectal cancer risk across the genome. We used variant weights from the PrediXcan models of colon tissue-specific gene expression as a priori variant information for a set-based G × E approach. We harmonized total folate intake (mcg/day) based on dietary intake and supplemental use across cohort and case-control studies and calculated sex and study specific quantiles. Analyses were performed using a mixed effects score tests for interactions between folate and genetically predicted expression of 4839 genes with available genetically predicted expression. We pooled results across 23 studies for a total of 13,498 cases with colorectal tumors and 13,918 controls of European ancestry. We used a false discovery rate of 0.2 to identify genes with suggestive evidence of an interaction. We found suggestive evidence of interaction with folate intake on CRC risk for genes including glutathione S-Transferase Alpha 1 (GSTA1; p = 4.3E-4), Tonsuko Like, DNA Repair Protein (TONSL; p = 4.3E-4), and Aspartylglucosaminidase (AGA: p = 4.5E-4). We identified three genes involved in preventing or repairing DNA damage that may interact with folate consumption to alter CRC risk. Glutathione is an antioxidant, preventing cellular damage and is a downstream metabolite of homocysteine and metabolized by GSTA1. TONSL is part of a complex that functions in the recovery of double strand breaks and AGA plays a role in lysosomal breakdown of glycoprotein.
  • Item
    No Preview Available
    Genetically Predicted Circulating C-Reactive Protein Concentration and Colorectal Cancer Survival: A Mendelian Randomization Consortium Study
    Hua, X ; Dai, JY ; Lindstrom, S ; Harrison, TA ; Lin, Y ; Alberts, SR ; Alwers, E ; Berndt, S ; Brenner, H ; Buchanan, DD ; Campbell, PT ; Casey, G ; Chang-Claude, J ; Gallinger, S ; Giles, GG ; Goldberg, RM ; Gunter, MJ ; Hoffmeister, M ; Jenkins, MA ; Joshi, AD ; Ma, W ; Milne, RL ; Murphy, N ; Pai, RK ; Sakoda, LC ; Schoen, RE ; Shi, Q ; Slattery, ML ; Song, M ; White, E ; Le Marchand, L ; Chan, AT ; Peters, U ; Newcomb, PA (AMER ASSOC CANCER RESEARCH, 2021-07)
    BACKGROUND: A positive association between circulating C-reactive protein (CRP) and colorectal cancer survival was reported in observational studies, which are susceptible to unmeasured confounding and reverse causality. We used a Mendelian randomization approach to evaluate the association between genetically predicted CRP concentrations and colorectal cancer-specific survival. METHODS: We used individual-level data for 16,918 eligible colorectal cancer cases of European ancestry from 15 studies within the International Survival Analysis of Colorectal Cancer Consortium. We calculated a genetic-risk score based on 52 CRP-associated genetic variants identified from genome-wide association studies. Because of the non-collapsibility of hazard ratios from Cox proportional hazards models, we used the additive hazards model to calculate hazard differences (HD) and 95% confidence intervals (CI) for the association between genetically predicted CRP concentrations and colorectal cancer-specific survival, overall and by stage at diagnosis and tumor location. Analyses were adjusted for age at diagnosis, sex, body mass index, genotyping platform, study, and principal components. RESULTS: Of the 5,395 (32%) deaths accrued over up to 10 years of follow-up, 3,808 (23%) were due to colorectal cancer. Genetically predicted CRP concentration was not associated with colorectal cancer-specific survival (HD, -1.15; 95% CI, -2.76 to 0.47 per 100,000 person-years; P = 0.16). Similarly, no associations were observed in subgroup analyses by stage at diagnosis or tumor location. CONCLUSIONS: Despite adequate power to detect moderate associations, our results did not support a causal effect of circulating CRP concentrations on colorectal cancer-specific survival. IMPACT: Future research evaluating genetically determined levels of other circulating inflammatory biomarkers (i.e., IL6) with colorectal cancer survival outcomes is needed.
  • Item
    Thumbnail Image
    DNA Methylation Signatures and the Contribution of Age-Associated Methylomic Drift to Carcinogenesis in Early-Onset Colorectal Cancer
    Joo, JE ; Clendenning, M ; Wong, EM ; Rosty, C ; Mahmood, K ; Georgeson, P ; Winship, IM ; Preston, SG ; Win, AK ; Dugue, P-A ; Jayasekara, H ; English, D ; Macrae, FA ; Hopper, JL ; Jenkins, MA ; Milne, RL ; Giles, GG ; Southey, MC ; Buchanan, DD (MDPI, 2021-06)
    We investigated aberrant DNA methylation (DNAm) changes and the contribution of ageing-associated methylomic drift and age acceleration to early-onset colorectal cancer (EOCRC) carcinogenesis. Genome-wide DNAm profiling using the Infinium HM450K on 97 EOCRC tumour and 54 normal colonic mucosa samples was compared with: (1) intermediate-onset CRC (IOCRC; diagnosed between 50-70 years; 343 tumour and 35 normal); and (2) late-onset CRC (LOCRC; >70 years; 318 tumour and 40 normal). CpGs associated with age-related methylation drift were identified using a public dataset of 231 normal mucosa samples from people without CRC. DNAm-age was estimated using epiTOC2. Common to all three age-of-onset groups, 88,385 (20% of all CpGs) CpGs were differentially methylated between tumour and normal mucosa. We identified 234 differentially methylated genes that were unique to the EOCRC group; 13 of these DMRs/genes were replicated in EOCRC compared with LOCRCs from TCGA. In normal mucosa from people without CRC, we identified 28,154 CpGs that undergo ageing-related DNAm drift, and of those, 65% were aberrantly methylated in EOCRC tumours. Based on the mitotic-based DNAm clock epiTOC2, we identified age acceleration in normal mucosa of people with EOCRC compared with normal mucosa from the IOCRC, LOCRC groups (p = 3.7 × 10-16) and young people without CRC (p = 5.8 × 10-6). EOCRC acquires unique DNAm alterations at 234 loci. CpGs associated with ageing-associated drift were widely affected in EOCRC without needing the decades-long accrual of DNAm drift as commonly seen in intermediate- and late-onset CRCs. Accelerated ageing in normal mucosa from people with EOCRC potentially underlies the earlier age of diagnosis in CRC carcinogenesis.
  • Item
    Thumbnail Image
    The MLH1 polymorphism rs1800734 and risk of endometrial cancer with microsatellite instability (vol 12, 102, 2020)
    Russell, H ; Kedzierska, K ; Buchanan, DD ; Thomas, R ; Tham, E ; Mints, M ; Keranen, A ; Giles, GG ; Southey, MC ; Milne, RL ; Tomlinson, I ; Church, D ; Spurdle, AB ; O'Mara, TA ; Lewis, A (BMC, 2021-04-01)
    An amendment to this paper has been published and can be accessed via the original article.
  • Item
    Thumbnail Image
    Genetic architectures of proximal and distal colorectal cancer are partly distinct
    Huyghe, JR ; Harrison, TA ; Bien, SA ; Hampel, H ; Figueiredo, JC ; Schmit, SL ; Conti, D ; Chen, S ; Qu, C ; Lin, Y ; Barfield, R ; Baron, JA ; Cross, AJ ; Diergaarde, B ; Duggan, D ; Harlid, S ; Imaz, L ; Kang, HM ; Levine, DM ; Perduca, V ; Perez-Cornago, A ; Sakoda, LC ; Schumacher, FR ; Slattery, ML ; Toland, AE ; van Duijnhoven, FJB ; Van Guelpen, B ; Agudo, A ; Albanes, D ; Alonso, MH ; Anderson, K ; Arnau-Collell, C ; Arndt, V ; Banbury, BL ; Bassik, MC ; Berndt, S ; Bezieau, S ; Bishop, DT ; Boehm, J ; Boeing, H ; Boutron-Ruault, M-C ; Brenner, H ; Brezina, S ; Buch, S ; Buchanan, DD ; Burnett-Hartman, A ; Caan, BJ ; Campbell, PT ; Carr, PR ; Castells, A ; Castellvi-Bel, S ; Chan, AT ; Chang-Claude, J ; Chanock, SJ ; Curtis, KR ; de la Chapelle, A ; Easton, DF ; English, DR ; Feskens, EJM ; Gala, M ; Gallinger, SJ ; Gauderman, WJ ; Giles, GG ; Goodman, PJ ; Grady, WM ; Grove, JS ; Gsur, A ; Gunter, MJ ; Haile, RW ; Hampe, J ; Hoffmeister, M ; Hopper, JL ; Hsu, W-L ; Huang, W-Y ; Hudson, TJ ; Jenab, M ; Jenkins, MA ; Joshi, AD ; Keku, TO ; Kooperberg, C ; Kuhn, T ; Kury, S ; Le Marchand, L ; Lejbkowicz, F ; Li, C ; Li, L ; Lieb, W ; Lindblom, A ; Lindor, NM ; Mannisto, S ; Markowitz, SD ; Milne, RL ; Moreno, L ; Murphy, N ; Nassir, R ; Offit, K ; Ogino, S ; Panico, S ; Parfrey, PS ; Pearlman, R ; Pharoah, PDP ; Phipps, A ; Platz, EA ; Potter, JD ; Prentice, RL ; Qi, L ; Raskin, L ; Rennert, G ; Rennert, HS ; Riboli, E ; Schafmayer, C ; Schoen, RE ; Seminara, D ; Song, M ; Su, Y-R ; Tangen, CM ; Thibodeau, SN ; Thomas, DC ; Trichopoulou, A ; Ulrich, CM ; Visvanathan, K ; Vodicka, P ; Vodickova, L ; Vymetalkova, V ; Weigl, K ; Weinstein, SJ ; White, E ; Wolk, A ; Woods, MO ; Wu, AH ; Abecasis, GR ; Nickerson, DA ; Scacheri, PC ; Kundaje, A ; Casey, G ; Gruber, SB ; Hsu, L ; Moreno, V ; Hayes, RB ; Newcomb, PA ; Peters, U (BMJ PUBLISHING GROUP, 2021-07)
    OBJECTIVE: An understanding of the etiologic heterogeneity of colorectal cancer (CRC) is critical for improving precision prevention, including individualized screening recommendations and the discovery of novel drug targets and repurposable drug candidates for chemoprevention. Known differences in molecular characteristics and environmental risk factors among tumors arising in different locations of the colorectum suggest partly distinct mechanisms of carcinogenesis. The extent to which the contribution of inherited genetic risk factors for CRC differs by anatomical subsite of the primary tumor has not been examined. DESIGN: To identify new anatomical subsite-specific risk loci, we performed genome-wide association study (GWAS) meta-analyses including data of 48 214 CRC cases and 64 159 controls of European ancestry. We characterised effect heterogeneity at CRC risk loci using multinomial modelling. RESULTS: We identified 13 loci that reached genome-wide significance (p<5×10-8) and that were not reported by previous GWASs for overall CRC risk. Multiple lines of evidence support candidate genes at several of these loci. We detected substantial heterogeneity between anatomical subsites. Just over half (61) of 109 known and new risk variants showed no evidence for heterogeneity. In contrast, 22 variants showed association with distal CRC (including rectal cancer), but no evidence for association or an attenuated association with proximal CRC. For two loci, there was strong evidence for effects confined to proximal colon cancer. CONCLUSION: Genetic architectures of proximal and distal CRC are partly distinct. Studies of risk factors and mechanisms of carcinogenesis, and precision prevention strategies should take into consideration the anatomical subsite of the tumour.
  • Item
    No Preview Available
    Mendelian randomization analyses suggest a role for cholesterol in the development of endometrial cancer
    Kho, P-F ; Amant, F ; Annibali, D ; Ashton, K ; Attia, J ; Auer, PL ; Beckmann, MW ; Black, A ; Brinton, L ; Buchanan, DD ; Chanock, SJ ; Chen, C ; Chen, MM ; Cheng, THT ; Cook, LS ; Crous-Bous, M ; Czene, K ; De Vivo, I ; Dennis, J ; Doerk, T ; Dowdy, SC ; Dunning, AM ; Duerst, M ; Easton, DF ; Ekici, AB ; Fasching, PA ; Fridley, BL ; Friedenreich, CM ; Garcia-Closas, M ; Gaudet, MM ; Giles, GG ; Goode, EL ; Gorman, M ; Haiman, CA ; Hall, P ; Hankinson, SE ; Hein, A ; Hillemanns, P ; Hodgson, S ; Hoivik, EA ; Holliday, EG ; Hunter, DJ ; Jones, A ; Kraft, P ; Krakstad, C ; Lambrechts, D ; Le Marchand, L ; Liang, X ; Lindblom, A ; Lissowska, J ; Long, J ; Lu, L ; Magliocco, AM ; Martin, L ; McEvoy, M ; Milne, RL ; Mints, M ; Nassir, R ; Otton, G ; Palles, C ; Pooler, L ; Proietto, T ; Rebbeck, TR ; Renner, SP ; Risch, HA ; Ruebner, M ; Runnebaum, I ; Sacerdote, C ; Sarto, GE ; Schumacher, F ; Scott, RJ ; Setiawan, VW ; Shah, M ; Sheng, X ; Shu, X-O ; Southey, MC ; Tham, E ; Tomlinson, I ; Trovik, J ; Turman, C ; Tyrer, JP ; van den Berg, D ; Wang, Z ; Wentzensen, N ; Xia, L ; Xiang, Y-B ; Yang, HP ; Yu, H ; Zheng, W ; Webb, PM ; Thompson, DJ ; Spurdle, AB ; Glubb, DM ; O'Mara, TA (WILEY, 2021-01-15)
    Blood lipids have been associated with the development of a range of cancers, including breast, lung and colorectal cancer. For endometrial cancer, observational studies have reported inconsistent associations between blood lipids and cancer risk. To reduce biases from unmeasured confounding, we performed a bidirectional, two-sample Mendelian randomization analysis to investigate the relationship between levels of three blood lipids (low-density lipoprotein [LDL] and high-density lipoprotein [HDL] cholesterol, and triglycerides) and endometrial cancer risk. Genetic variants associated with each of these blood lipid levels (P < 5 × 10-8 ) were identified as instrumental variables, and assessed using genome-wide association study data from the Endometrial Cancer Association Consortium (12 906 cases and 108 979 controls) and the Global Lipids Genetic Consortium (n = 188 578). Mendelian randomization analyses found genetically raised LDL cholesterol levels to be associated with lower risks of endometrial cancer of all histologies combined, and of endometrioid and non-endometrioid subtypes. Conversely, higher genetically predicted HDL cholesterol levels were associated with increased risk of non-endometrioid endometrial cancer. After accounting for the potential confounding role of obesity (as measured by genetic variants associated with body mass index), the association between genetically predicted increased LDL cholesterol levels and lower endometrial cancer risk remained significant, especially for non-endometrioid endometrial cancer. There was no evidence to support a role for triglycerides in endometrial cancer development. Our study supports a role for LDL and HDL cholesterol in the development of non-endometrioid endometrial cancer. Further studies are required to understand the mechanisms underlying these findings.
  • Item
    Thumbnail Image
    Biological Aging Measures Based on Blood DNA Methylation and Risk of Cancer: A Prospective Study
    Dugue, P-A ; Bassett, JK ; Wong, EM ; Joo, JE ; Li, S ; Yu, C ; Schmidt, DF ; Makalic, E ; Doo, NW ; Buchanan, DD ; Hodge, AM ; English, DR ; Hopper, JL ; Giles, GG ; Southey, MC ; Milne, RL (OXFORD UNIV PRESS, 2021-02)
    BACKGROUND: We previously investigated the association between 5 "first-generation" measures of epigenetic aging and cancer risk in the Melbourne Collaborative Cohort Study. This study assessed cancer risk associations for 3 recently developed methylation-based biomarkers of aging: PhenoAge, GrimAge, and predicted telomere length. METHODS: We estimated rate ratios (RRs) for the association between these 3 age-adjusted measures and risk of colorectal (N = 813), gastric (N = 165), kidney (N = 139), lung (N = 327), mature B-cell (N = 423), prostate (N = 846), and urothelial (N = 404) cancer using conditional logistic regression models. We also assessed associations by time since blood draw and by cancer subtype, and we investigated potential nonlinearity. RESULTS: We observed relatively strong associations of age-adjusted PhenoAge with risk of colorectal, kidney, lung, mature B-cell, and urothelial cancers (RR per SD was approximately 1.2-1.3). Similar findings were obtained for age-adjusted GrimAge, but the association with lung cancer risk was much larger (RR per SD = 1.82, 95% confidence interval [CI] = 1.44 to 2.30), after adjustment for smoking status, pack-years, starting age, time since quitting, and other cancer risk factors. Most associations appeared linear, larger than for the first-generation measures, and were virtually unchanged after adjustment for a large set of sociodemographic, lifestyle, and anthropometric variables. For cancer overall, the comprehensively adjusted rate ratio per SD was 1.13 (95% CI = 1.07 to 1.19) for PhenoAge and 1.12 (95% CI = 1.05 to 1.20) for GrimAge and appeared larger within 5 years of blood draw (RR = 1.29, 95% CI = 1.15 to 1.44 and 1.19, 95% CI = 1.06 to 1.33, respectively). CONCLUSIONS: The methylation-based measures PhenoAge and GrimAge may provide insights into the relationship between biological aging and cancer and be useful to predict cancer risk, particularly for lung cancer.