Clinical Pathology - Research Publications

Permanent URI for this collection

Search Results

Now showing 1 - 10 of 16
  • Item
    Thumbnail Image
    DNA methylation-based biological aging and cancer risk and survival: Pooled analysis of seven prospective studies
    Dugue, P-A ; Bassett, JK ; Joo, JE ; Jung, C-H ; Wong, EM ; Moreno-Betancur, M ; Schmidt, D ; Makalic, E ; Li, S ; Severi, G ; Hodge, AM ; Buchanan, DD ; English, DR ; Hopper, JL ; Southey, MC ; Giles, GG ; Milne, RL (WILEY, 2018-04-15)
    The association between aging and cancer is complex. Recent studies have developed measures of biological aging based on DNA methylation and called them "age acceleration." We aimed to assess the associations of age acceleration with risk of and survival from seven common cancers. Seven case-control studies of DNA methylation and colorectal, gastric, kidney, lung, prostate and urothelial cancer and B-cell lymphoma nested in the Melbourne Collaborative Cohort Study were conducted. Cancer cases, vital status and cause of death were ascertained through linkage with cancer and death registries. Conditional logistic regression and Cox models were used to estimate odds ratios (OR) and hazard ratios (HR) and 95% confidence intervals (CI) for associations of five age acceleration measures derived from the Human Methylation 450 K Beadchip assay with cancer risk (N = 3,216 cases) and survival (N = 1,726 deaths), respectively. Epigenetic aging was associated with increased cancer risk, ranging from 4% to 9% per five-year age acceleration for the 5 measures considered. Heterogeneity by study was observed, with stronger associations for risk of kidney cancer and B-cell lymphoma. An associated increased risk of death following cancer diagnosis ranged from 2% to 6% per five-year age acceleration, with no evidence of heterogeneity by cancer site. Cancer risk and mortality were increased by 15-30% for the fourth versus first quartile of age acceleration. DNA methylation-based measures of biological aging are associated with increased cancer risk and shorter cancer survival, independently of major health risk factors.
  • Item
    Thumbnail Image
    Associations of alcohol intake, smoking, physical activity and obesity with survival following colorectal cancer diagnosis by stage, anatomic site and tumor molecular subtype
    Jayasekara, H ; English, DR ; Haydon, A ; Hodge, AM ; Lynch, BM ; Rosty, C ; Williamson, EJ ; Clendenning, M ; Southey, MC ; Jenkins, MA ; Room, R ; Hopper, JL ; Milne, RL ; Buchanan, DD ; Giles, GG ; MacInnis, RJ (WILEY, 2018-01-15)
    The influence of lifestyle factors on survival following a diagnosis of colorectal cancer (CRC) is not well established. We examined associations between lifestyle factors measured before diagnosis and CRC survival. The Melbourne Collaborative Cohort Study collected data on alcohol intake, cigarette smoking and physical activity, and body measurements at baseline (1990-1994) and wave 2 (2003-2007). We included participants diagnosed to 31 August 2015 with incident stages I-III CRC within 10-years post exposure assessment. Information on tumor characteristics and vital status was obtained. Tumor DNA was tested for microsatellite instability (MSI) and somatic mutations in oncogenes BRAF (V600E) and KRAS. We estimated hazard ratios (HRs) for associations between lifestyle factors and overall and CRC-specific mortality using Cox regression. Of 724 eligible CRC cases, 339 died (170 from CRC) during follow-up (average 9.0 years). Exercise (non-occupational/leisure-time) was associated with higher CRC-specific survival for stage II (HR = 0.25, 95% CI: 0.10-0.60) but not stages I/III disease (p for interaction = 0.01), and possibly for colon and KRAS wild-type tumors. Waist circumference was inversely associated with CRC-specific survival (HR = 1.25 per 10 cm increment, 95% CI: 1.08-1.44), independent of stage, anatomic site and tumor molecular status. Cigarette smoking was associated with lower overall survival, with suggestive evidence of worse survival for BRAF mutated CRC, but not with CRC-specific survival. Alcohol intake was not associated with survival. Survival did not differ by MSI status. We have identified pre-diagnostic predictors of survival following CRC that may have clinical and public health relevance.
  • Item
    Thumbnail Image
    Lifetime alcohol intake is associated with an increased risk of KRAS plus and BRAF-/KRAS- but not BRAF plus colorectal cancer
    Jayasekara, H ; MacInnis, RJ ; Williamson, EJ ; Hodge, AM ; Clendenning, M ; Rosty, C ; Walters, R ; Room, R ; Southey, MC ; Jenkins, MA ; Milne, RL ; Hopper, JL ; Giles, GG ; Buchanan, DD ; English, DR (WILEY, 2017-04)
    Ethanol in alcoholic beverages is a causative agent for colorectal cancer. Colorectal cancer is a biologically heterogeneous disease, and molecular subtypes defined by the presence of somatic mutations in BRAF and KRAS are known to exist. We examined associations between lifetime alcohol intake and molecular and anatomic subtypes of colorectal cancer. We calculated usual alcohol intake for 10-year periods from age 20 using recalled frequency and quantity of beverage-specific consumption for 38,149 participants aged 40-69 years from the Melbourne Collaborative Cohort Study. Cox regression was performed to derive hazard ratios (HRs) and 95% confidence intervals (CIs) for the association between lifetime alcohol intake and colorectal cancer risk. Heterogeneity in the HRs across subtypes of colorectal cancer was assessed. A positive dose-dependent association between lifetime alcohol intake and overall colorectal cancer risk (mean follow-up = 14.6 years; n = 596 colon and n = 326 rectal cancer) was observed (HR = 1.08, 95% CI: 1.04-1.12 per 10 g/day increment). The risk was greater for rectal than colon cancer (phomogeneity  = 0.02). Alcohol intake was associated with increased risks of KRAS+ (HR = 1.07, 95% CI: 1.00-1.15) and BRAF-/KRAS- (HR = 1.05, 95% CI: 1.00-1.11) but not BRAF+ tumors (HR = 0.89, 95% CI: 0.78-1.01; phomogeneity  = 0.01). Alcohol intake is associated with an increased risk of KRAS+ and BRAF-/KRAS- tumors originating via specific molecular pathways including the traditional adenoma-carcinoma pathway but not with BRAF+ tumors originating via the serrated pathway. Therefore, limiting alcohol intake from a young age might reduce colorectal cancer originating via the traditional adenoma-carcinoma pathway.
  • Item
    Thumbnail Image
    Identification of nine new susceptibility loci for endometrial cancer
    O'Mara, TA ; Glubb, DM ; Amant, F ; Annibali, D ; Ashton, K ; Attia, J ; Auer, PL ; Beckmann, MW ; Black, A ; Bolla, MK ; Brauch, H ; Brenner, H ; Brinton, L ; Buchanan, DD ; Burwinkel, B ; Chang-Claude, J ; Chanock, SJ ; Chen, C ; Chen, MM ; Cheng, THT ; Clarke, CL ; Clendenning, M ; Cook, LS ; Couch, FJ ; Cox, A ; Crous-Bous, M ; Czene, K ; Day, F ; Dennis, J ; Depreeuw, J ; Doherty, JA ; Dork, T ; Dowdy, SC ; Duerst, M ; Ekici, AB ; Fasching, PA ; Fridley, BL ; Friedenreich, CM ; Fritschi, L ; Fung, J ; Garcia-Closas, M ; Gaudet, MM ; Giles, GG ; Goode, EL ; Gorman, M ; Haiman, CA ; Hall, P ; Hankison, SE ; Healey, CS ; Hein, A ; Hillemanns, P ; Hodgson, S ; Hoivik, EA ; Holliday, EG ; Hopper, JL ; Hunter, DJ ; Jones, A ; Krakstad, C ; Kristensen, VN ; Lambrechts, D ; Le Marchand, L ; Liang, X ; Lindblom, A ; Lissowska, J ; Long, J ; Lu, L ; Magliocco, AM ; Martin, L ; McEvoy, M ; Meindl, A ; Michailidou, K ; Milne, RL ; Mints, M ; Montgomery, GW ; Nassir, R ; Olsson, H ; Orlow, I ; Otton, G ; Palles, C ; Perry, JRB ; Peto, J ; Pooler, L ; Prescott, J ; Proietto, T ; Rebbeck, TR ; Risch, HA ; Rogers, PAW ; Ruebner, M ; Runnebaum, I ; Sacerdote, C ; Sarto, GE ; Schumacher, F ; Scott, RJ ; Setiawan, VW ; Shah, M ; Sheng, X ; Shu, X-O ; Southey, MC ; Swerdlow, AJ ; Tham, E ; Trovik, J ; Turman, C ; Tyrer, JP ; Vachon, C ; Vanden Berg, D ; Vanderstichele, A ; Wang, Z ; Webb, PM ; Wentzensen, N ; Werner, HMJ ; Winham, SJ ; Wolk, A ; Xia, L ; Xiang, Y-B ; Yang, HP ; Yu, H ; Zheng, W ; Pharoah, PDP ; Dunning, AM ; Kraft, P ; De Vivo, I ; Tomlinson, I ; Easton, DF ; Spurdle, AB ; Thompson, DJ (NATURE PUBLISHING GROUP, 2018-08-09)
    Endometrial cancer is the most commonly diagnosed cancer of the female reproductive tract in developed countries. Through genome-wide association studies (GWAS), we have previously identified eight risk loci for endometrial cancer. Here, we present an expanded meta-analysis of 12,906 endometrial cancer cases and 108,979 controls (including new genotype data for 5624 cases) and identify nine novel genome-wide significant loci, including a locus on 12q24.12 previously identified by meta-GWAS of endometrial and colorectal cancer. At five loci, expression quantitative trait locus (eQTL) analyses identify candidate causal genes; risk alleles at two of these loci associate with decreased expression of genes, which encode negative regulators of oncogenic signal transduction proteins (SH2B3 (12q24.12) and NF1 (17q11.2)). In summary, this study has doubled the number of known endometrial cancer risk loci and revealed candidate causal genes for future study.
  • Item
    Thumbnail Image
    Methylation of Breast Cancer Predisposition Genes in Early-Onset Breast Cancer: Australian Breast Cancer Family Registry
    Scott, CM ; Joo, JE ; O'Callaghan, N ; Buchanan, DD ; Clendenning, M ; Giles, GG ; Hopper, JL ; Wong, EM ; Southey, MC ; Toland, AE (PUBLIC LIBRARY SCIENCE, 2016-11-30)
    DNA methylation can mimic the effects of both germline and somatic mutations for cancer predisposition genes such as BRCA1 and p16INK4a. Constitutional DNA methylation of the BRCA1 promoter has been well described and is associated with an increased risk of early-onset breast cancers that have BRCA1-mutation associated histological features. The role of methylation in the context of other breast cancer predisposition genes has been less well studied and often with conflicting or ambiguous outcomes. We examined the role of methylation in known breast cancer susceptibility genes in breast cancer predisposition and tumor development. We applied the Infinium HumanMethylation450 Beadchip (HM450K) array to blood and tumor-derived DNA from 43 women diagnosed with breast cancer before the age of 40 years and measured the methylation profiles across promoter regions of BRCA1, BRCA2, ATM, PALB2, CDH1, TP53, FANCM, CHEK2, MLH1, MSH2, MSH6 and PMS2. Prior genetic testing had demonstrated that these women did not carry a germline mutation in BRCA1, ATM, CHEK2, PALB2, TP53, BRCA2, CDH1 or FANCM. In addition to the BRCA1 promoter region, this work identified regions with variable methylation at multiple breast cancer susceptibility genes including PALB2 and MLH1. Methylation at the region of MLH1 in these breast cancers was not associated with microsatellite instability. This work informs future studies of the role of methylation in breast cancer susceptibility gene silencing.
  • Item
    No Preview Available
    Mendelian randomization analysis of C-reactive protein on colorectal cancer risk
    Wang, X ; Dai, JY ; Albanes, D ; Arndt, V ; Berndt, SI ; Bezieau, S ; Brenner, H ; Buchanan, DD ; Butterbach, K ; Caan, B ; Casey, G ; Campbell, PT ; Chan, AT ; Chen, Z ; Chang-Claude, J ; Cotterchio, M ; Easton, DF ; Giles, GG ; Giovannucci, E ; Grady, WM ; Hoffmeister, M ; Hopper, JL ; Hsu, L ; Jenkins, MA ; Joshi, AD ; Lampe, JW ; Larsson, SC ; Lejbkowicz, F ; Li, L ; Lindblom, A ; Le Marchand, L ; Martin, V ; Milne, RL ; Moreno, V ; Newcomb, PA ; Offitt, K ; Ogino, S ; Pharoah, PDP ; Pinchev, M ; Potter, JD ; Rennert, HS ; Rennert, G ; Saliba, W ; Schafmayer, C ; Schoen, RE ; Schrotz-King, P ; Slattery, ML ; Song, M ; Stegmaier, C ; Weinstein, SJ ; Wolk, A ; Woods, MO ; Wu, AH ; Gruber, SB ; Peters, U ; White, E (OXFORD UNIV PRESS, 2019-06)
    BACKGROUND: Chronic inflammation is a risk factor for colorectal cancer (CRC). Circulating C-reactive protein (CRP) is also moderately associated with CRC risk. However, observational studies are susceptible to unmeasured confounding or reverse causality. Using genetic risk variants as instrumental variables, we investigated the causal relationship between genetically elevated CRP concentration and CRC risk, using a Mendelian randomization approach. METHODS: Individual-level data from 30 480 CRC cases and 22 844 controls from 33 participating studies in three international consortia were used: the Genetics and Epidemiology of Colorectal Cancer Consortium (GECCO), the Colorectal Transdisciplinary Study (CORECT) and the Colon Cancer Family Registry (CCFR). As instrumental variables, we included 19 single nucleotide polymorphisms (SNPs) previously associated with CRP concentration. The SNP-CRC associations were estimated using a logistic regression model adjusted for age, sex, principal components and genotyping phases. An inverse-variance weighted method was applied to estimate the causal effect of CRP on CRC risk. RESULTS: Among the 19 CRP-associated SNPs, rs1260326 and rs6734238 were significantly associated with CRC risk (P = 7.5 × 10-4, and P = 0.003, respectively). A genetically predicted one-unit increase in the log-transformed CRP concentrations (mg/l) was not associated with increased risk of CRC [odds ratio (OR) = 1.04; 95% confidence interval (CI): 0.97, 1.12; P = 0.256). No evidence of association was observed in subgroup analyses stratified by other risk factors. CONCLUSIONS: In spite of adequate statistical power to detect moderate association, we found genetically elevated CRP concentration was not associated with increased risk of CRC among individuals of European ancestry. Our findings suggested that circulating CRP is unlikely to be a causal factor in CRC development.
  • Item
    Thumbnail Image
    Type 2 diabetes mellitus, blood cholesterol, triglyceride and colorectal cancer risk in Lynch syndrome
    Dashti, SG ; Li, WY ; Buchanan, DD ; Clendenning, M ; Rosty, C ; Winship, IM ; Macrae, FA ; Giles, GG ; Hardikar, S ; Hua, X ; Thibodeau, SN ; Figueiredo, JC ; Casey, G ; Haile, RW ; Gallinger, S ; Le Marchand, L ; Newcomb, PA ; Potter, JD ; Lindor, NM ; Hopper, JL ; Jenkins, MA ; Win, AK (NATURE PUBLISHING GROUP, 2019-11-12)
    BACKGROUND: Type 2 diabetes mellitus and high total cholesterol and triglycerides are known to be associated with increased colorectal cancer risk for the general population. These associations are unknown for people with a germline DNA mismatch repair gene mutation (Lynch syndrome), who are at high risk of colorectal cancer. METHODS: This study included 2023 (56.4% female) carriers with a mismatch repair gene mutation (737 in MLH1, 928 in MSH2, 230 in MSH6, 106 in PMS2, 22 in EPCAM) recruited by the Colon Cancer Family Registry between 1998 and 2012. Weighted Cox regression was used to estimate the hazard ratios (HR) and 95% confidence intervals (CI) for the associations between self-reported type 2 diabetes, high cholesterol, triglyceride and colorectal cancer risk. RESULTS: Overall, 802 carriers were diagnosed with colorectal cancer at a median age of 42 years. A higher risk of colorectal cancer was observed in those with self-reported type-2 diabetes (HR 1.92; 95% CI, 1.03-3.58) and high cholesterol (HR 1.76; CI 1.23-2.52) compared with those without these conditions. There was no evidence of high triglyceride being associated with colorectal cancer risk. CONCLUSION: For people with Lynch syndrome, self-reported type-2 diabetes mellitus and high cholesterol were associated with increased colorectal cancer risk.
  • Item
    Thumbnail Image
    Genetic variant predictors of gene expression provide new insight into risk of colorectal cancer
    Bien, SA ; Su, Y-R ; Conti, DV ; Harrison, TA ; Qu, C ; Guo, X ; Lu, Y ; Albanes, D ; Auer, PL ; Banbury, BL ; Berndt, SI ; Bezieau, S ; Brenner, H ; Buchanan, DD ; Caan, BJ ; Campbell, PT ; Carlson, CS ; Chan, AT ; Chang-Claude, J ; Chen, S ; Connolly, CM ; Easton, DF ; Feskens, EJM ; Gallinger, S ; Giles, GG ; Gunter, MJ ; Hampe, J ; Huyghe, JR ; Hoffmeister, M ; Hudson, TJ ; Jacobs, EJ ; Jenkins, MA ; Kampman, E ; Kang, HM ; Kuehn, T ; Kury, S ; Lejbkowicz, F ; Le Marchand, L ; Milne, RL ; Li, L ; Li, CI ; Lindblom, A ; Lindor, NM ; Martin, V ; McNeil, CE ; Melas, M ; Moreno, V ; Newcomb, PA ; Offit, K ; Pharaoh, PDP ; Potter, JD ; Qu, C ; Riboli, E ; Rennert, G ; Sala, N ; Schafmayer, C ; Scacheri, PC ; Schmit, SL ; Severi, G ; Slattery, ML ; Smith, JD ; Trichopoulou, A ; Tumino, R ; Ulrich, CM ; van Duijnhoven, FJB ; Van Guelpen, B ; Weinstein, SJ ; White, E ; Wolk, A ; Woods, MO ; Wu, AH ; Abecasis, GR ; Casey, G ; Nickerson, DA ; Gruber, SB ; Hsu, L ; Zheng, W ; Peters, U (SPRINGER, 2019-04)
    Genome-wide association studies have reported 56 independently associated colorectal cancer (CRC) risk variants, most of which are non-coding and believed to exert their effects by modulating gene expression. The computational method PrediXcan uses cis-regulatory variant predictors to impute expression and perform gene-level association tests in GWAS without directly measured transcriptomes. In this study, we used reference datasets from colon (n = 169) and whole blood (n = 922) transcriptomes to test CRC association with genetically determined expression levels in a genome-wide analysis of 12,186 cases and 14,718 controls. Three novel associations were discovered from colon transverse models at FDR ≤ 0.2 and further evaluated in an independent replication including 32,825 cases and 39,933 controls. After adjusting for multiple comparisons, we found statistically significant associations using colon transcriptome models with TRIM4 (discovery P = 2.2 × 10- 4, replication P = 0.01), and PYGL (discovery P = 2.3 × 10- 4, replication P = 6.7 × 10- 4). Interestingly, both genes encode proteins that influence redox homeostasis and are related to cellular metabolic reprogramming in tumors, implicating a novel CRC pathway linked to cell growth and proliferation. Defining CRC risk regions as one megabase up- and downstream of one of the 56 independent risk variants, we defined 44 non-overlapping CRC-risk regions. Among these risk regions, we identified genes associated with CRC (P < 0.05) in 34/44 CRC-risk regions. Importantly, CRC association was found for two genes in the previously reported 2q25 locus, CXCR1 and CXCR2, which are potential cancer therapeutic targets. These findings provide strong candidate genes to prioritize for subsequent laboratory follow-up of GWAS loci. This study is the first to implement PrediXcan in a large colorectal cancer study and findings highlight the utility of integrating transcriptome data in GWAS for discovery of, and biological insight into, risk loci.
  • Item
    No Preview Available
    Novel Common Genetic Susceptibility Loci for Colorectal Cancer
    Schmit, SL ; Edlund, CK ; Schumacher, FR ; Gong, J ; Harrison, TA ; Huyghe, JR ; Qu, C ; Melas, M ; Van den Berg, DJ ; Wang, H ; Tring, S ; Plummer, SJ ; Albanes, D ; Alonso, MH ; Amos, CI ; Anton, K ; Aragaki, AK ; Arndt, V ; Barry, EL ; Berndt, SI ; Bezieau, S ; Bien, S ; Bloomer, A ; Boehm, J ; Boutron-Ruault, M-C ; Brenner, H ; Brezina, S ; Buchanan, DD ; Butterbach, K ; Caan, BJ ; Campbell, PT ; Carlson, CS ; Castelao, JE ; Chan, AT ; Chang-Claude, J ; Chanock, SJ ; Cheng, I ; Cheng, Y-W ; Chin, LS ; Church, JM ; Church, T ; Coetzee, GA ; Cotterchio, M ; Correa, MC ; Curtis, KR ; Duggan, D ; Easton, DF ; English, D ; Feskens, EJM ; Fischer, R ; FitzGerald, LM ; Fortini, BK ; Fritsche, LG ; Fuchs, CS ; Gago-Dominguez, M ; Gala, M ; Gallinger, SJ ; Gauderman, WJ ; Giles, GG ; Giovannucci, EL ; Gogarten, SM ; Gonzalez-Villalpando, C ; Gonzalez-Villalpando, EM ; Grady, WM ; Greenson, JK ; Gsur, A ; Gunter, M ; Haiman, CA ; Hampe, J ; Harlid, S ; Harju, JF ; Hayes, RB ; Hofer, P ; Hoffmeister, M ; Hopper, JL ; Huang, S-C ; Huerta, JM ; Hudson, TJ ; Hunter, DJ ; Idos, GE ; Iwasaki, M ; Jackson, RD ; Jacobs, EJ ; Jee, SH ; Jenkins, MA ; Jia, W-H ; Jiao, S ; Joshi, AD ; Kolonel, LN ; Kono, S ; Kooperberg, C ; Krogh, V ; Kuehn, T ; Kury, S ; LaCroix, A ; Laurie, CA ; Lejbkowicz, F ; Lemire, M ; Lenz, H-J ; Levine, D ; Li, CI ; Li, L ; Lieb, W ; Lin, Y ; Lindor, NM ; Liu, Y-R ; Loupakis, F ; Lu, Y ; Luh, F ; Ma, J ; Mancao, C ; Manion, FJ ; Markowitz, SD ; Martin, V ; Matsuda, K ; Matsuo, K ; McDonnell, KJ ; McNeil, CE ; Milne, R ; Molina, AJ ; Mukherjee, B ; Murphy, N ; Newcomb, PA ; Offit, K ; Omichessan, H ; Palli, D ; Cotore, JPP ; Perez-Mayoral, J ; Pharoah, PD ; Potter, JD ; Qu, C ; Raskin, L ; Rennert, G ; Rennert, HS ; Riggs, BM ; Schafmayer, C ; Schoen, RE ; Sellers, TA ; Seminara, D ; Severi, G ; Shi, W ; Shibata, D ; Shu, X-O ; Siegel, EM ; Slattery, ML ; Southey, M ; Stadler, ZK ; Stern, MC ; Stintzing, S ; Taverna, D ; Thibodeau, SN ; Thomas, DC ; Trichopoulou, A ; Tsugane, S ; Ulrich, CM ; van Duijnhoven, FJB ; van Guelpan, B ; Vijai, J ; Virtamo, J ; Weinstein, SJ ; White, E ; Win, AK ; Wolk, A ; Woods, M ; Wu, AH ; Wu, K ; Xiang, Y-B ; Yen, Y ; Zanke, BW ; Zeng, Y-X ; Zhang, B ; Zubair, N ; Kweon, S-S ; Figueiredo, JC ; Zheng, W ; Le Marchand, L ; Lindblom, A ; Moreno, V ; Peters, U ; Casey, G ; Hsu, L ; Conti, DV ; Gruber, SB (OXFORD UNIV PRESS INC, 2019-02)
    BACKGROUND: Previous genome-wide association studies (GWAS) have identified 42 loci (P < 5 × 10-8) associated with risk of colorectal cancer (CRC). Expanded consortium efforts facilitating the discovery of additional susceptibility loci may capture unexplained familial risk. METHODS: We conducted a GWAS in European descent CRC cases and control subjects using a discovery-replication design, followed by examination of novel findings in a multiethnic sample (cumulative n = 163 315). In the discovery stage (36 948 case subjects/30 864 control subjects), we identified genetic variants with a minor allele frequency of 1% or greater associated with risk of CRC using logistic regression followed by a fixed-effects inverse variance weighted meta-analysis. All novel independent variants reaching genome-wide statistical significance (two-sided P < 5 × 10-8) were tested for replication in separate European ancestry samples (12 952 case subjects/48 383 control subjects). Next, we examined the generalizability of discovered variants in East Asians, African Americans, and Hispanics (12 085 case subjects/22 083 control subjects). Finally, we examined the contributions of novel risk variants to familial relative risk and examined the prediction capabilities of a polygenic risk score. All statistical tests were two-sided. RESULTS: The discovery GWAS identified 11 variants associated with CRC at P < 5 × 10-8, of which nine (at 4q22.2/5p15.33/5p13.1/6p21.31/6p12.1/10q11.23/12q24.21/16q24.1/20q13.13) independently replicated at a P value of less than .05. Multiethnic follow-up supported the generalizability of discovery findings. These results demonstrated a 14.7% increase in familial relative risk explained by common risk alleles from 10.3% (95% confidence interval [CI] = 7.9% to 13.7%; known variants) to 11.9% (95% CI = 9.2% to 15.5%; known and novel variants). A polygenic risk score identified 4.3% of the population at an odds ratio for developing CRC of at least 2.0. CONCLUSIONS: This study provides insight into the architecture of common genetic variation contributing to CRC etiology and improves risk prediction for individualized screening.
  • Item
    No Preview Available
    Genetic variant predictors of gene expression provide new insight into risk of colorectal cancer (vol 138, pg 307, 2019)
    Bien, SA ; Su, Y-R ; Conti, DV ; Harrison, TA ; Qu, C ; Guo, X ; Lu, Y ; Albanes, D ; Auer, PL ; Banbury, BL ; Berndt, SI ; Bezieau, S ; Brenner, H ; Buchanan, DD ; Caan, BJ ; Campbell, PT ; Carlson, CS ; Chan, AT ; Chang-Claude, J ; Chen, S ; Connolly, CM ; Easton, DF ; Feskens, EJM ; Gallinger, S ; Giles, GG ; Gunter, MJ ; Hampe, J ; Huyghe, JR ; Hoffmeister, M ; Hudson, TJ ; Jacobs, EJ ; Jenkins, MA ; Kampman, E ; Kang, HM ; Kuehn, T ; Kury, S ; Lejbkowicz, F ; Le Marchand, L ; Milne, RL ; Li, L ; Li, CI ; Lindblom, A ; Lindor, NM ; Martin, V ; McNeil, CE ; Melas, M ; Moreno, V ; Newcomb, PA ; Offit, K ; Pharaoh, PDP ; Potter, JD ; Qu, C ; Riboli, E ; Rennert, G ; Sala, N ; Schafmayer, C ; Scacheri, PC ; Schmit, SL ; Severi, G ; Slattery, ML ; Smith, JD ; Trichopoulou, A ; Tumino, R ; Ulrich, CM ; van Duijnhoven, FJB ; Van Guelpen, B ; Weinstein, SJ ; White, E ; Wolk, A ; Woods, MO ; Wu, AH ; Abecasis, GR ; Casey, G ; Nickerson, DA ; Gruber, SB ; Hsu, L ; Zheng, W ; Peters, U (SPRINGER, 2019-07)
    Every author has erroneously been assigned to the affiliation "62". The affiliation 62 belongs to the author Graham Casey.