Abstract
Background
Physical activity is associated with lower colorectal cancer (CRC) risk, but its association with molecular subtypes defined by genetic and epigenetic alterations of the disease is unclear. Such information may enhance the understanding of the mechanisms related to the benefits of physical activity.
Methods
Pooled observational (cases: n = 5386; controls: n = 6798; studies n = 5) and genome-wide association data (cases: n = 8178; controls: n = 10 472; studies n = 5) were used. We used multivariable logistic regression models and Mendelian randomization to assess the association between physical activity and the risk of CRC subtypes defined by individual tumor markers (and marker combinations), namely microsatellite instability status, CpG island methylator phenotype status, and BRAF and KRAS mutations. We used case-only analysis to test for differences between molecular subtypes. We applied Bonferroni correction to account for multiple tests.
Results
In the pooled observational analysis, higher levels of physical activity were associated with lower CRC risk (Obs-per 1SD, odds ratio [OR] = 0.94, 95% confidence interval [CI] = 0.90 to 0.97), with an association that was stronger in males (Obs-per 1SD, OR = 0.91, 95% CI = 0.87 to 0.96) than in females (Obs-per 1SD, OR = 0.97, 95% CI = 0.91 to 1.03; Pinteraction = .04). Higher physical activity was associated with a lower risk of CRC across all molecular subtypes, especially in males. There was no difference in the associations by subtypes by pooled observational or Mendelian randomization analyses. The findings did not differ by study design, anatomical site, and early or late age onset of CRC.
Conclusions
Our findings suggest that physical activity is not differentially associated with the 4 major molecular subtypes involved in colorectal carcinogenesis, indicating that its benefits extend broadly across colorectal cancer pathogenesis.
Introduction
An important element of colorectal cancer (CRC) primary prevention involves identifying modifiable lifestyle factors that influence the risk of developing the disease. Such factors associated with CRC development include smoking,1 anthropometric measures (eg, body mass index [BMI],2,3 waist circumference2) alcohol intake,4 diet (eg, wholegrains, fiber, dairy products, processed and red meat),5 and physical activity.6-8 Physical activity may reduce CRC risk through lower circulating insulin9,10 and pro-inflammatory factors,11,12 enhanced immune response,11,13 and beneficial changes to the gut microbiome.14,15
CRC is one of the most common cancers and is responsible for a large burden of cancer death.16 It is characterized by substantial genetic and epigenetic diversity.17 Detailed molecular characterization of CRC using clinically relevant genetic and epigenetic markers shows great potential for improving prognosis18 and guiding treatment decisions.19 Mutations in the KRAS gene play a crucial role in developing colorectal adenomas, occurring in approximately 30%-40% of sporadic cases.20 Additionally, microsatellite instability, identified by frequent changes in repetitive DNA sequences, is present in approximately 15% of CRC cases and is associated with a more favorable prognosis.21 Some microsatellite instability–high CRC cases also feature the CpG island methylator phenotype and BRAF c.1799T>A (p.V600E) mutations.7 BRAF p. V600E is associated with a worse prognosis,22 whereas BRAF mutations predict response to encorafenib plus cetuximab,23,24 KRAS mutations indicate poor response to estimated glomerular filtration rate inhibitors,25 and microsatellite instability–high status predicts response to immunotherapy.26,27 Currently, there is no evidence of whether the association of physical activity with CRC varies by tumor molecular markers; however, such information might enhance the understanding of the mechanisms related to the benefits of physical activity.
This study aimed to illuminate the association between physical activity and the risk of CRC by tumor molecular subtypes (microsatellite instability, CpG island methylator phenotype, BRAF, and KRAS mutation status). Thus, we performed an observational and a 2-sample Mendelian randomization analysis using data from the Genetics and Epidemiology of Colorectal Cancer Consortium (GECCO) and the Colon Cancer Family Registry (CCFR).
Methods
Observational analysis
Study population
This study sample included CRC cases and controls nested in 3 cohorts (Cancer Prevention Study-II [CPS-II],28,29 Nurses’ Health Study [NHS],30 and Health Professionals Follow-up Study [HPFS]) and 2 case-control studies (Darmkrebs: Chancen der Verhütung durch Screening Study [DACHS]31,32 and Diet, Activity and Lifestyle Study [DALS])33,34 within GECCO with available tumor marker and physical activity data (Table S1). All CRC cases were defined as colorectal adenocarcinoma and confirmed by pathological records, medical records, and/or death certificate information. Additional information on the contributing studies is included in the Supplements (Table S1 and Description of included studies). All participants provided written informed consent. A research ethics committee or institutional review board approved each study.
Tumor molecular subtypes of CRC
Testing for microsatellite instability, CpG island methylator phenotype, and mutations in the BRAF and KRAS genes was conducted previously by each study and according to individual study protocols. Briefly, microsatellite instability testing was primarily conducted using polymerase chain reaction following accepted guidelines (CPS-II, HPFS, NHS)35 with 4 or more interpretable markers typically required to classify tumors (Table S2). DALS and DACHS used a mononucleotide panel of 2 and 3 markers, respectively.
The studies used polymerase chain reaction, sequencing, and immunohistochemistry techniques to assess BRAF and KRAS mutations, as detailed in the Supplements (harmonization of colorectal tumor marker data). CpG island methylator phenotype status was determined using methylation analyses (Table S3). The CPS-II, HPFS, and NHS used MethyLight to determine CpG island methylator phenotype status. CPS-II, HPFS, and NHS used an 8-gene panel.36 CpG island methylator phenotype categories for our analyses were CpG island methylator phenotype high and CpG island methylator phenotype low/negative. DACHS determined CpG island methylator phenotype status using a different 5-gene panel,36 and DALS determined CpG island methylator phenotype status using a classic panel of CpG islands.37,38 Additionally, we combined markers to create subtype classifications: subtypes 1-5 were created according to the Jass classification,39 and types 6-16 were numbered consecutively by the status of microsatellite instability, CpG island methylator phenotype, BRAF, and KRAS.
Physical activity definition
In cohort studies, data on physical activity were collected at baseline through in-person interviews or structured self-administered questionnaires. In case-control studies, this information was obtained from cases and controls, referencing the period 1-2 years before enrollment. Dietary variables were ascertained using food frequency questionnaires. The exposure of interest was physical activity, defined as the sum of leisure time and undifferentiated activity, expressed as the metabolic equivalent of task (MET) hours per week. As the range of MET hours per week was different among the studies, and to make the effect estimates comparable, MET hours per week were standardized by subtracting the mean (CPS-II = 16, HPFS = 33, NHS = 17, DACHS = 52, DALS = 38) and dividing it by the SD (CPS-II = 11, HPFS = 28, NHS = 16, DACHS = 33, DALS = 18) (Figure S1).
Confounders
In cohort studies (CPS-II, HPFS, and NHS), sociodemographic and lifestyle information was collected at baseline through in-person interviews or structured self-administered questionnaires. In case-control studies (DACHS and DALS), this information was gathered from cases and controls with reference to the period 1-2 years before enrollment. Dietary variables were ascertained using food frequency questionnaires. A multistep iterative data-harmonization procedure was applied, reconciling each study’s unique protocols and data collection instruments. Multiple quality-control checks were performed, and outlying values of variables were truncated to the minimum or maximum value of an established range for each variable. Variables were combined into a single dataset with standard definitions, standardized coding, and standardized permissible values.
Statistical analysis
Physical activity was modeled continuously (per 1-SD) and categorically in tertiles defined among controls only. Multinomial logistic regression models were used to estimate the relative risk ratio for the association between physical activity and CRC subtypes (compared with control participants) defined by tumor markers (microsatellite instability high vs microsatellite stable or microsatellite instability low, CpG island methylator phenotype high vs low/negative, and BRAF or KRAS mutated vs wild type). Logistic regression models were performed to assess if the differences in the associations of physical activity with CRC across molecularly defined subtypes were statistically significant among cases only. In the case-only analysis, we also conducted a logistic regression analysis defining Jass type 4 (microsatellite stable or microsatellite instability low, CpG island methylator phenotype low/negative, BRAF wild type, KRAS mutated) as the reference group to evaluate significance of differences among Jass types of tumor marker combinations. Jass types with at least 50 cases were assessed in relation to physical activity. The multivariable models included study population, age (continuous, years), sex (male, female), smoking status (never, former, current smokers), alcohol consumption (continuous, grams per day), education (less than high school graduate, high school graduate, some college, college graduate), and red meat intake (continuous, servings per day).
The analysis included only participants with complete information. Separate analyses were conducted by study design (cohort and case-control studies), sex, cancer anatomical site (proximal colon, distal colon, colon, rectum), and by early (aged 50 years and younger) and later (older than 50 years) age onset CRC. A sensitivity analysis was conducted after imputing missing values for physical activity and all covariates used in the adjustment model (see Table 1) using multiple imputations by chained equations40 with 5 imputed datasets and 5 iterations using the random forest method. Diagnostic plots suggested a good performance of this method (Figure S2-S4). As body size and adiposity is potentially on the causal pathway linking physical activity with colorectal cancer, we did not include BMI in the main analyses. However, we additionally adjusted our main model for BMI, processed meat, fiber, fruits, and vegetables without observing any difference in the results. Finally, we pooled study-specific odds ratios (ORs) for the associations between physical activity and overall CRC using random effects meta-analysis models to evaluate for heterogeneity between studies.
Table 1.
Baseline characteristics of cases and controls used in observational analysis
| Characteristics | Cases, No. (%) (n = 5386) | Controls, No. (%) (n = 6798) |
|---|---|---|
| Study | ||
| CPS-II | 860 (16) | 1003 (14.8) |
| DACHS | 2009 (37.3) | 2789 (41) |
| DALS | 1095 (20.3) | 1162 (17.1) |
| HPFS | 629 (11.7) | 602 (8.9) |
| NHS | 793 (14.7) | 1242 (18.3) |
| Age,a mean (SD), y | 69 (10) | 69 (10) |
| Unknown | 5 (<0.1) | 0 |
| Sex | ||
| Male | 2834 (52.6) | 3430 (50.5) |
| Female | 2552 (47.4) | 3368 (49.5) |
| Smoking status | ||
| Never smoker | 2290 (42.5) | 3217 (47.3) |
| Former smoker | 2389 (44.4) | 2869 (42.2) |
| Current smoker | 634 (11.8) | 661 (9.7) |
| Unknown | 73 (1.4) | 51 (0.8) |
| Body mass index, kg/m2 | ||
| Mean (SD) | 27 (4.6) | 26.1 (4.2) |
| Underweight, <18.5 | 48 (0.9) | 74 (1.1) |
| Normal, 18.5 to <24.9 | 1824 (33.9) | 2795 (41.1) |
| Overweight, 25- 30 | 2276 (42.3) | 2813 (41.4) |
| Obese, >30 | 1088 (20.2) | 1001 (14.7) |
| Unknown | 228 (3.8) | 115 (1.7) |
| Dietary intake, mean (SD) | ||
| Red meat, servings per day | 0.85 (0.61) | 0.77 (0.55) |
| Unknown | 141 (2.6) | 167 (2.5) |
| Processed meat, servings per day | 0.48 (0.46) | 0.45 (0.43) |
| Unknown | 279 (5.2) | 253 (3.7) |
| Fruits, servings per day | 1.64 (1.44) | 1.67 (1.36) |
| Unknown | 230 (4.3) | 316 (4.6) |
| Vegetables, servings per day | 2.47 (1.91) | 2.41 (1.85) |
| Unknown | 225 (4.2) | 312 (4.6) |
| Fiber, gm per day | 21 (9) | 21 (9) |
| Unknown | 2260 (42) | 2987 (44) |
| Education level | ||
| Less than high school graduate | 627 (11.6) | 598 (8.8) |
| High school graduate or completed General Education Development test | 1592 (29.6) | 1984 (29.2) |
| Some college or technical school | 1162 (21.6) | 1194 (17.6) |
| College graduate | 1882 (34.9) | 2980 (43.8) |
| Unknown | 123 (2.3) | 42 (0.6) |
| First-degree relative with CRC | 565 (10.3) | 483 (7.1) |
| Unknown | 390 (7.2) | 368 (5.4) |
| Location of CRC | ||
| Distal colon | 1722 (32) | — |
| Proximal colon | 2373 (44.1) | — |
| Rectum, including rectosigmoid junction | 1230 (22.8) | — |
| Unknown | 61 (1.1) | — |
| CRC stage | ||
| Stage 1 or local | 1438 (26.7) | |
| Stage 2-3 or regional | 3146 (58.4) | |
| Stage 4 or distant | 601 (11.2) | |
| Unknown | 201 (3.7) | |
| BRAF | ||
| Wild type | 4262 (79.2) | — |
| Mutated | 543 (10.1) | — |
| Unknown | 577 (10.7) | — |
| KRAS | ||
| Wild type | 3162 (58.7) | — |
| Mutated | 1630 (30.3) | — |
| Unknown | 594 (11) | — |
| MSI | ||
| MSS/MSI low | 4221 (78.4) | — |
| MSI high | 705 (13.1) | — |
| Unknown | 460 (8.5) | — |
| CIMP | ||
| Low/negative | 4015 (74.5) | — |
| High | 926 (17.2) | — |
| Unknown | 445 (8.3) | — |
| Jass type (subtypes combinations) | ||
| [1] MSI-high, CIMP+, BRAF mut, KRAS wt | 260 (4.8) | — |
| [2] MSS or MSI-low, CIMP+, BRAF mut, KRAS wt | 103 (1.9) | — |
| [3] MSS or MSI-low, CIMP-, BRAF wt, KRAS mut | 1161 (21.6) | — |
| [4] MSS or MSI-low, CIMP-, BRAF wt, KRAS wt | 1868 (34.7) | — |
| [5] MSI-high, CIMP-, BRAF wt, KRAS wt | 98 (1.8) | — |
| [6] MSS or MSI-low, CIMP-, BRAF mut, KRAS wt | 77 (1.4) | — |
| [7] MSS or MSI-low, CIMP-, BRAF mut, KRAS mut | 5 (0.1) | — |
| [8] MSS or MSI-low, CIMP+, BRAF wt, KRAS wt | 115 (2.1) | — |
| [9] MSS or MSI-low, CIMP+, BRAF wt, KRAS mut | 147 (2.7) | — |
| [10] MSS or MSI-low, CIMP+, BRAF mut, KRAS mut | — | — |
| [11] MSI-high, CIMP-, BRAF wt, KRAS mut | 59 (1.1) | — |
| [12] MSI-high, CIMP-, BRAF mut, KRAS wt | 13 (0.2) | — |
| [13] MSI-high, CIMP-, BRAF mut, KRAS mut | 2 (0) | — |
| [14] MSI-high, CIMP+, BRAF wt, KRAS wt | 108 (2) | — |
| [15] MSI-high, CIMP+, BRAF wt, KRAS mut | 21 (0.4) | — |
| [16] MSI-high, CIMP+, BRAF mut, KRAS mut | 9 (0.2) | — |
| Unknown | 1913 (32.1) | — |
Abbreviations: CIMP = CpG island methylator phenotype; CIMP- = CIMP low/negative; CIMP+ = CIMP high; CPSII = Cancer Prevention Study II; CRC = colorectal cancer; DACHS = Darmkrebs: Chancen der Verhctung durch Screening Study; DALS = Activity and Lifestyle Study; HPFS = Health Professionals Follow-up Study; MSI = microsatellite instability; MSS = microsatellite stable; mut = mutated; NHS = Nurses’ Health Study; wt = wild type.
Figures are expressed as No (%) unless otherwise specified. The em dashes mean that there are not available data as they are relevant only to colorectal cancer cases and not for those without (controls).
Age at diagnosis (cases) and selection (controls).
We used a Bonferroni-corrected threshold of 0.004 (0.05/12; 4 subtype classifications based on 4 individual tumor markers being tested × 3 analyses: men only, women only, sex combined) to assess statistical significance for the case-control and case-only analyses of the primary subtypes (microsatellite instability, CpG island methylator phenotype–status BRAF, and KRAS). For secondary analyses (marginal analysis for overall CRC and subsequent subgroup analyses), we considered a 2-sided P value less than .05 as statistically significant. Associations with P values between the Bonferroni-corrected and nominal threshold (.05) were characterized as suggestive. Analyses were performed using R v4.3.1 (R Foundation for Statistical Computing, Vienna, Austria).
Mendelian randomization
Study population
For overall CRC, summary statistics were drawn from a genome-wide association study (GWAS) meta-analysis of 17 studies, encompassing 78 473 CRC cancer cases and 107 143 controls of European descent.41 Summary data for CRC molecular subtypes were drawn from a GWAS meta-analysis of 10 studies (CCFR,42 CPSII,28,29 NHS,30 HPFS, DACHS,31,32 DALS,33,34 Early Detection Research Network,43 European Prospective Investigation into Cancer Sweden,44 Melbourne Collaborative Cohort Study,45 and Northern Sweden Health and Disease Study46) within the CCFR and the GECCO consortia (Table S1). The current study includes 8178 CRC cases and 10 472 controls of European ancestry, with available information on the 4 molecular markers (Tables S4 and S5). Additional information on contributing studies is included in Table S3 and the Supplements (Table S1 and description of included studies).
Physical activity definition
Summary statistics for accelerometer-based average physical activity were obtained from a GWAS of 91 105 UK Biobank participants that explained approximately 0.2% of the variance in physical activity.47 Physical activity was objectively measured using a wrist-worn accelerometer worn continuously for 7 days. Activity levels were recorded in milligravity units (mg), with a mean of 29.0 (8.14 mg). One standard deviation corresponds to approximately 50 minutes of moderate-intensity activity (eg, brisk walking) per week.
Statistical analysis
Mendelian randomization leverages genetic variants, typically single-nucleotide polymorphisms, as instrumental variables to estimate the causal effect of an exposure on an outcome.48,49 Because genetic variants are randomly assorted at meiosis and fixed at conception, Mendelian randomization is less susceptible to confounding and reverse causation than observational studies. For Mendelian randomization estimates to be valid, 3 core assumptions must hold: (1) the genetic variants should be associated with the exposure, (2) they should not be associated with confounders of the exposure-outcome association, and (3) they should influence the outcome solely through the exposure and not via alternative biological pathways. A violation of the third assumption—known as horizontal pleiotropy—can bias results.
To create the genetic instruments, all relevant genetic variants were selected if they demonstrated genome-wide significant association (P < 5 × to represent genetic susceptibility to physical activity and were uncorrelated ( < 0.001) within a 10-Mb window based on the European 1000 genomes reference panel. F-statistics of at least 10 were considered of adequate instrument strength.
To estimate study power, we considered that the genetic instruments explained 0.2% of the variance in physical activity; given the sample size for overall CRC, the minimum detectable odds ratio with 80% power at a 5% significance level was 0.74. The primary method was random-effects inverse variance–weighted Mendelian randomization.50 To account for potential horizontal pleiotropy, several Mendelian randomization sensitivity analyses (Mendelian randomization–Egger,51 weighted median,52 weighted mode53) were performed, each providing a valid Mendelian randomization estimate under different combinations of assumptions. We additionally implemented the Mendelian randomization pleiotropy residual sum and outlier test (Mendelian randomization–Pleiotropy RESidual Sum and Outlie [PRESSO]) to detect and exclude potential outlying genetic variants.54 Mendelian randomization analysis was performed with R v4.1.2 using the TwoSampleMR package.
We applied a Bonferroni-corrected significance threshold of a P value less than .012 (0.05/4), accounting for the 4 primary tumor markers evaluated (Mendelian randomization status, CpG island methylator phenotype status, BRAF, and KRAS mutations), in the case-control and case-only analyses. For the secondary analysis assessing overall CRC risk, a 2-sided P value less than .05 was considered statistically significant. Associations with P values between the Bonferroni-corrected and nominal threshold (0.05) were characterized as suggestive.
Results
Observational analysis
Study population characteristics
The study sample comprised 5386 CRC cases and 6798 controls from 5 observational studies (Table 1). Compared with controls, individuals with a CRC diagnosis were more likely to be male than females (52.6% vs 50.5%), former or current smokers (56.2% vs 51.9%), and obese (20.2% vs 14.7%) and to have a first-degree relative with CRC (10.3% vs 7.1%). Among cases, 13.1% were microsatellite instability high (n = 705), 17.2% were CpG island methylator phenotype high (n = 926), 10.1% were BRAF-mutated (n = 543), and 30.3% were KRAS mutated (n = 1630), with the type 4 (34.7%) being the most frequent Jass type.
Physical activity and overall CRC risk
Higher MET hours per week of physical activity per 1 standard deviation (across all studies approximately 30.1 MET hours per week) were associated with lower odds of developing CRC (Obs-per 1SD, OR = 0.94, 95% confidence interval [CI] = 0.90 to 0.97), with a stronger association observed in males (Obs-per 1SD, OR = 0.91, 95% CI = 0.87 to 0.96), and no association observed in females (Obs-per 1SD, OR = 0.97, 95% CI = 0.92 to 1.03) (Pinteraction = .04) (Table 2). A similar association between physical activity and overall CRC was found when individual study odds ratios were pooled in a meta-analysis with a low between-study heterogeneity (I2 = 27.4%, PCochran Q = .24, τ2 < 0.01) (Figure S5). In the subgroup analysis by study design, the association between physical activity and overall CRC risk was present only in the cohort (Obs-per 1SD, OR = 0.89, 95% CI = 0.84 to 0.95) and not in case-control studies (Obs-per 1SD, OR = 0.97, 95% CI = 0.92 to 1.02; Pheterogeneity = .0768) (Table S6). When analyzed by anatomical site, physical activity was associated with lower odds of rectal (cohorts: Obs-per 1SD, OR = 0.80, 95% CI = 0.67 to 0.95) and colon cancer (cohorts: Obs-per 1SD, OR = 0.84, 95% CI = 0.76 to 0.93) in males (Tables S7-S14).
Table 2.
Association between physical activity, colorectal cancer, and its molecular subtypes according to the observational analysisa,b,c
| Standardized MET h/wk | Overall CRC OR (95% CI) | MSI |
CIMP |
BRAF
|
KRAS
|
||||
|---|---|---|---|---|---|---|---|---|---|
| MSS or MSI-low RR ratio (95% CI) | MSI-high RR ratio (95% CI) | CIMP-low/negative RR ratio (95% CI) | CIMP-high RR ratio (95% CI) | BRAF-wild type RR ratio (95% CI) | BRAF-mutated RR ratio (95% CI) | KRAS-wild type RR ratio (95% CI) | KRAS-mutated RR ratio (95% CI) | ||
| Sex combined | |||||||||
| No. cases | 4989 | 3915 | 651 | 3720 | 849 | 3947 | 493 | 2930 | 1501 |
| −1.58 to −0.591 | Referent | Referent | Referent | Referent | Referent | Referent | Referent | Referent | Referent |
| −0.590 to 0.2 | 0.85 (0.78 to 0.93) | 0.84 (0.77 to 0.93) | 0.83 (0.68 to 1.01) | 0.85 (0.77 to 0.94) | 0.90 (0.75 to 1.06) | 0.86 (0.78 to 0.95) | 0.89 (0.71 to 1.11) | 0.83 (0.75 to 0.93) | 0.85 (0.74 to 0.98) |
| 0.21 to 3.03 | 0.86 (0.78 to 0.94) | 0.86 (0.78 to 0.95) | 0.80 (0.66 to 0.98) | 0.88 (0.80 to 0.98) | 0.79 (0.66 to 0.95) | 0.86 (0.78 to 0.95) | 0.80 (0.64 to 1.01) | 0.82 (0.74 to 0.92) | 0.85 (0.74 to 0.97) |
| P trend | .0012 | .0033 | .0314 | .0121 | .0111 | .0027 | .0670 | .0004 | .0176 |
| Per 1-SD | 0.94 (0.90 to 0.97) | 0.94 (0.90 to 0.98) | 0.91 (0.83 to 0.99) | 0.95 (0.91 to 0.99) | 0.90 (0.83 to 0.97) | 0.94 (0.90 to 0.98) | 0.90 (0.82 to 1.00) | 0.92 (0.88 to 0.97) | 0.93 (0.88 to 0.99) |
| P | .0011 | .0043 | .0302 | .0105 | .0081 | .0029 | .0461 | .0007 | .0144 |
| P difference | NA | .36 | .17 | .40 | .75 | ||||
| Male | |||||||||
| No. cases | 2718 | 2206 | 268 | 2123 | 360 | 2242 | 180 | 1583 | 836 |
| −1.58 to −0.591 | Referent | Referent | Referent | Referent | Referent | Referent | Referent | Referent | Referent |
| −0.590 to 0.2 | 0.82 (0.73 to 0.93) | 0.80 (0.70 to 0.91) | 0.84 (0.63 to 1.14) | 0.81 (0.71 to 0.93) | 0.79 (0.61 to 1.02) | 0.80 (0.70 to 0.91) | 0.92 (0.65 to 1.32) | 0.80 (0.69 to 0.93) | 0.81 (0.67 to 0.98) |
| 0.21 to 3.03 | 0.77 (0.68 to 0.88) | 0.79 (0.69 to 0.91) | 0.65 (0.47 to 0.89) | 0.81 (0.71 to 0.93) | 0.62 (0.47 to 0.82) | 0.78 (0.68 to 0.89) | 0.68 (0.46 to 1.00) | 0.72 (0.62 to 0.84) | 0.83 (0.69 to 1.01) |
| P trend | .0001 | .0007 | .0079 | .0021 | .0008 | .0003 | .0561 | .0001 | .0567 |
| Per 1-SD | 0.91 (0.87 to 0.96) | 0.92 (0.87 to 0.98) | 0.86 (0.75 to 0.98) | 0.93 (0.88 to 0.98) | 0.82 (0.72 to 0.92) | 0.92 (0.87 to 0.97) | 0.87 (0.74 to 1.02) | 0.89 (0.84 to 0.95) | 0.93 (0.86 to 1.00) |
| P | .0004 | .0042 | .0247 | .0122 | .0008 | .0015 | .0896 | .0002 | .0618 |
| P difference | NA | .28 | .02 | .52 | .30 | ||||
| Female | |||||||||
| No. cases | 2271 | 1709 | 383 | 1597 | 489 | 1705 | 313 | 1347 | 665 |
| −1.58 to −0.591 | Referent | Referent | Referent | Referent | Referent | Referent | Referent | Referent | Referent |
| −0.590 to 0.2 | 0.89 (0.78 to 1.01) | 0.90 (0.78 to 1.04) | 0.82 (0.64 to 1.07) | 0.89 (0.77 to 1.03) | 0.99 (0.79 to 1.25) | 0.93 (0.81 to 1.08) | 0.87 (0.66 to 1.16) | 0.88 (0.75 to 1.03) | 0.89 (0.73 to 1.09) |
| 0.21 to 3.03 | 0.98 (0.85 to 1.12) | 0.96 (0.82 to 1.11) | 0.96 (0.74 to 1.26) | 0.98 (0.84 to 1.14) | 0.96 (0.75 to 1.22) | 0.97 (0.84 to 1.13) | 0.90 (0.67 to 1.21) | 0.96 (0.82 to 1.14) | 0.86 (0.69 to 1.06) |
| P trend | .6109 | .5526 | .6951 | .7197 | .7551 | .6945 | .4617 | .5840 | .1528 |
| Per 1-SD | 0.97 (0.92 to 1.03) | 0.97 (0.91 to 1.03) | 0.96 (0.85 to 1.05) | 0.97 (0.92 to 1.03) | 0.98 (0.88 to 1.09) | 0.97 (0.91 to 1.04) | 0.93 (0.81 to 1.06) | 0.97 (0.91 to 1.04) | 0.93 (0.84 to 1.02) |
| P | .3847 | .3097 | .4661 | .3046 | .6885 | .4148 | .2645 | .4312 | .1141 |
| P difference | NA | .97 | .69 | .58 | .38 | ||||
Abbreviations: CI = confidence interval; CIMP = CpG island methylator phenotype; CRC = colorectal cancer; MET = metabolic equivalent of task; MSI = microsatellite instability; MSS = microsatellite stable; OR = odds ratio; RR = relative risk.
Controls are used as the referents for all effect estimates.
Models are adjusted for the study population, age, sex (when not stratified), smoking status, alcohol consumption, education, and red meat intake.
Case-only analysis used to calculate Pdifference.
Physical activity and molecular subtypes
Higher MET hours per week were associated with a lower risk of CRC across all molecular subtypes, especially in males. The case-only analysis showed no statistically significant difference in the association of MET hours per week with different molecular subtypes, after multiple testing correction (Table 2). The associations across all molecular subtypes did not differ by study design (Table S6), anatomical site (Tables S7-S14), and early- and later-age onset of CRC (Table S15). A reanalysis of the data with imputed missing values for physical activity and confounders yielded identical findings (Table S16).
Of 16 possible combined CRC subtypes, defined by microsatellite instability, CpG island methylator phenotype, BRAF and KRAS, 10 had 50 or more cases and were included in the combined sex analysis. There was no evidence of differences by Jass type in case-only analyses in sex-combined or sex-specific analyses (Figure 1).
Figure 1.
Association between physical activity (per 1-SD increase in MET hours per week) and Jass classified types of colorectal cancer, overall and stratified by sex, according to the observational analysis. Controls were used as the referent for all relative risk ratios. The models were adjusted for age, sex, study population, alcohol consumption, smoking status, education, and red meat intake. Jass types with more than 50 cases were included. P values were calculated using multinomial logistic regression comparing CRC cases with cancer-free controls separately for each defined Jass type. Pdifference was calculated using multinomial logistic regression comparing cases of each Jass type with all additional cases not belonging to that type. Abbreviations: CI = confidence interval; CIMP = CpG island methylator phenotype; CIMP- = CIMP low/negative; CIMP+ = CIMP high; CRC = colorectal cancer; MET = metabolic equivalent of task; MSI = microsatellite instability; MSS = microsatellite stable; mut = mutated; RR = relative risk; wt = wild type.
Mendelian randomization analysis
Physical activity and molecular subtypes
Six genetic variants were used as genetic instruments for physical activity, with the F-statistics ranging from 29.5 to 46.4 (Table S17). Higher genetically predicted levels (per 1-SD increase = 8.14 mg, corresponding to approximately 50 minutes of moderate activity per week) of accelerometer-measured physical activity were inversely associated with overall CRC (inverse variance–weighted OR = 0.63, 95% CI = 0.45 to 0.89) and its tumor subtypes defined by individual molecular markers, with suggestive (after correcting for multiple testing) associations (microsatellite stable or microsatellite instability low [inverse variance–weighted OR = 0.46, 95% CI = 0.23 to 0.95], BRAF wild type [inverse variance–weighted OR = 0.43, 95% CI = 0.21 to 0.87], and KRAS wild type [inverse variance–weighted OR = 0.41, 95% CI = 0.19 to 0.90]), consistent with the observational analysis (Figure 2; Table S18). The sensitivity analyses generated similar and consistent associations (Table S18). There was no heterogeneity in the associations between genetically predicted levels of accelerometer-measured physical activity and molecular subtypes.
Figure 2.
The association of genetically predicted levels of accelerometer-measured physical activity (per 1-SD [8.14 milligravities] increase) with CRC risk based on the individual tumor molecular markers, according to the Mendelian randomization analysis. P was calculated using the inverse variance–weighted method comparing CRC cases with cancer-free controls. Pdifference was calculated using the inverse variance–weighted method comparing cases of each molecular subtype. Abbreviations: CI = confidence interval; CIMP = CpG island methylator phenotype; CRC = colorectal cancer; MET = metabolic equivalent of task; MSI = microsatellite instability; MSS = microsatellite stable; OR = odds ratio.
Discussion
We performed an observational pooled analysis using individual-level and a 2-sample Mendelian randomization analysis using GWAS summary data to illuminate the association between physical activity and the risk of CRC by tumor molecular subtypes. Higher levels of physical activity were associated with a lower risk of CRC in males but not in females. Higher physical activity was associated with a lower risk of CRC across all molecular subtypes, especially in males, but there was no difference in the associations by subtypes, and the Mendelian randomization findings agreed. The associations across all molecular subtypes did not differ by study design, anatomical site, and early and later age onset CRC.
Our findings align with previous meta-analyses and individual cohort studies. Specifically, Johnson et al.55 conducted a meta-analysis across 12 cohort studies and found that each 2-unit increase in standardized physical activity score was associated with an 8% (95% CI = 0.88 to 0.96) lower CRC risk. In addition, 2 prior Mendelian randomization studies on genetically predicted higher self-reported and accelerometer-measured physical activity showed lower CRC risk.6,56 Another meta-analysis combining cohort and case-control studies demonstrated that individuals with the highest levels of recreational physical activity had a 20% lower risk of colon cancer (95% CI = 0.71 to 0.89) and a 13% lower risk of rectal cancer (95% CI = 0.75 to 1.01) compared with those with the lowest levels of activity.57 We observed that the protective association of physical activity with overall CRC risk was stronger in males than in females, consistent with previous evidence.58,59 This sex difference could be partly attributed to hormonal variations and differences13 and potential biological differences in how males and females respond to exercise.60 In addition, sex-specific differences in the type and intensity of physical activity may also contribute to the variation in CRC risk between males and females.59
CRC is a heterogeneous disease with several distinct molecular subtypes, suggesting different pathways of tumorigenesis and progression.17 Therefore, considering molecular markers that differentiate these pathways is crucial for a deeper understanding of how lifestyle factors, like physical activity, influence CRC risk. Previous research has identified variations in the relationship between smoking and CRC risk based on these molecular markers,61 however, evidence regarding physical activity remains limited. Our findings indicate that the inverse association of physical activity with CRC is not specific to any specific molecular phenotype of CRC. This aligns with a smaller pooled analysis from 3 CRC patient cohorts, namely the Assessment of Targeted Therapies Against Colorectal Cancer, the biomarker-based protocol (Integromics), and The Cancer Genome Atlas, which found no differentiation in the association of physical activity between CpG island methylator phenotype–low/negative and CpG island methylator phenotype–high tumors.62 Similarly, long-term vigorous physical activity was associated with a lower risk of CpG island methylator phenotype–low and CpG island methylator phenotype–high tumors, using data from a large population-based case-control study of incident colon cancer.63 These results suggest that the benefits of physical activity may apply broadly across various CRC molecular subtypes, reinforcing its importance in CRC prevention strategies.
This is the largest study investigating the association between physical activity and the risk of CRC by tumor molecular subtypes. A major strength of our study is the ability to pool individual-level data from 5 observational studies with available information on physical activity measurements and tumor-marker status, providing insights into how physical activity is associated with different pathways of colorectal tumorigenesis. Another key strength is that we triangulated the results, employing a Mendelian randomization analysis. Triangulation integrates evidence from different approaches, each with different and unrelated sources of bias, to potentially strengthen causal inference.64 Some limitations should be considered when interpreting the findings. Our analysis used well-known molecular subtypes (CpG island methylator phenotype, microsatellite instability, KRAS, BRAF). Nevertheless, comprehensive tumor characterization, including next-generation sequencing of somatic mutations to include genome-wide DNA methylation analysis, histone modification profiling, gene expression studies, immune profiling through protein-based assays, and spatial transcriptomics could allow a deeper classification of tumor subtypes. The included studies used different tools to assess physical activity, introducing heterogeneity, and some studies measured recreational and others undifferentiated physical activity. To deal with this heterogeneity, we used z scores of physical activity in the analysis. In the observational pooled analysis, physical activity data were obtained through self-reports, which can be subject to social desirability and approval biases,65 but the results were not different when we performed Mendelian randomization based on accelerometer-derived activity. Investigation by Jass subtypes resulted in small sample sizes for some types because of their rarity, which may have limited statistical power to detect modest associations. Despite the adjustment for several potential confounders, we cannot rule out the possibility of unmeasured and residual confounding, although further adjustments in sensitivity analyses did not alter the results. Finally, this study included predominantly White populations (United States, Europe, Canada, and Australia); therefore, the findings may not be generalizable to other racial and ethnic groups.
In conclusion, this study confirms the benefits of physical activity on CRC risk, especially in males. The evidence suggests that physical activity does not differentially influence the major molecular subtypes, defined by microsatellite instability, CpG island methylator phenotype, BRAF, and KRAS involved in colorectal carcinogenesis. Maintaining regular physical activity plays a vital role in preventing colorectal cancer.
Supplementary Material
Acknowledgments
CCFR: The CCFR graciously thanks the generous contributions of their study participants, dedication of study staff, and the financial support from the US National Cancer Institute without which this important registry would not exist. The authors would like to thank the study participants and staff of the Seattle Colon Cancer Family Registry and the Hormones and Colon Cancer study (CORE studies).
Campaign Against Cancer and Heart Disease (CLUE) II: We thank the participants of CLUE II and appreciate the continued efforts of the staff at the Johns Hopkins George W. Comstock Center for Public Health Research and Prevention in the conduct of the CLUE II Cohort Study. Cancer data was provided by the Maryland Cancer Registry, Center for Cancer Prevention and Control, Maryland Department of Health, with funding from the State of Maryland and the Maryland Cigarette Restitution Fund. The collection and availability of cancer registry data is also supported by the cooperative agreement NU58DP006333, funded by the Centers for Disease Control and Prevention. Its contents are solely the responsibility of the authors and do not necessarily represent the official views of the Centers for Disease Control and Prevention or the Department of Health and Human Services.
CPS-II: The authors express sincere appreciation to all CPS-II participants and to each member of the study and biospecimen management group. The authors would like to acknowledge the contribution to this study from central cancer registries supported through the Centers for Disease Control and Prevention’s National Program of Cancer Registries and cancer registries supported by the National Cancer Institute’s Surveillance Epidemiology and End Results (SEER) Program. The authors assume full responsibility for all analyses and interpretation of results. The views expressed here are those of the authors and do not necessarily represent the American Cancer Society or the American Cancer Society—Cancer Action Network.
DACHS: We thank all participants and cooperating clinicians, and everyone who provided excellent technical assistance.
Early Detection Research Network (EDRN): We acknowledge all contributors to the development of the resource at the University of Pittsburgh School of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Department of Pathology, and Biomedical Informatics.
European Prospective Investigation into Cancer (EPIC): Where authors are identified as personnel of the International Agency for Research on Cancer/World Health Organization, the authors alone are responsible for the views expressed in this article, and they do not necessarily represent the decisions, policy, or views of the International Agency for Research on Cancer/World Health Organization.
Harvard cohorts: The study protocol was approved by the institutional review boards of the Brigham and Women’s Hospital and Harvard T.H. Chan School of Public Health and those of participating registries as required. We acknowledge Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital as home of the NHS. The authors would like to acknowledge the contribution to this study from central cancer registries supported through the Centers for Disease Control and Prevention’s National Program of Cancer Registries and/or the National Cancer Institute’s SEER Program. Central registries may also be supported by state agencies, universities, and cancer centers. Participating central cancer registries include the following: Alabama, Alaska, Arizona, Arkansas, California, Colorado, Connecticut, Delaware, Florida, Georgia, Hawaii, Idaho, Indiana, Iowa, Kentucky, Louisiana, Massachusetts, Maine, Maryland, Michigan, Mississippi, Montana, Nebraska, Nevada, New Hampshire, New Jersey, New Mexico, New York, North Carolina, North Dakota, Ohio, Oklahoma, Oregon, Pennsylvania, Puerto Rico, Rhode Island, Seattle SEER Registry, South Carolina, Tennessee, Texas, Utah, Virginia, West Virginia, and Wyoming. The authors assume full responsibility for analyses and interpretation of these data.
ISACC: The authors would like to thank all those at the ISACC Coordinating Center for helping bring together the data and people that made this project possible.
Northern Sweden Health and Disease Study (NSHDS) investigators thank the Västerbotten Intervention Programme, the Northern Sweden MONICA study, the Biobank Research Unit at Umeå University and Biobanken Norr at Region Västerbotten for providing data and samples and acknowledge the contribution from Biobank Sweden, supported by the Swedish Research Council.
The funders had no role in the design of the study; the collection, analysis, and interpretation of the data; the writing of the manuscript; or the decision to submit the manuscript for publication.
Contributor Information
Christos V Chalitsios, Department of Hygiene and Epidemiology, University of Ioannina, Ioannina, Greece.
Georgios Markozannes, Department of Hygiene and Epidemiology, University of Ioannina, Ioannina, Greece; Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom.
Elom K Aglago, Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom.
Sonja I Berndt, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States.
Daniel D Buchanan, Colorectal Oncogenomics Group, Department of Clinical Pathology, Melbourne Medical School, The University of Melbourne, Parkville, Australia; University of Melbourne Centre for Cancer Research, The University of Melbourne, Parkville, Australia; Genomic Medicine and Family Cancer Clinic, The Royal Melbourne Hospital, Parkville, Victoria, Australia.
Peter T Campbell, Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, United States.
Yin Cao, Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, St Louis, MO, United States; Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, St Louis, MO, United States; Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St Louis, MO, United States.
Andrew T Chan, Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States; Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States; Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States; Broad Institute of Harvard and MIT, Cambridge, MA, United States; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA, United States; Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA, United States.
Niki Dimou, Nutrition and Metabolism Branch, International Agency for Research on Cancer, Lyon, France.
David A Drew, Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States; Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States.
Amy J French, Division of Laboratory Genetics, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States.
Peter Georgeson, Colorectal Oncogenomics Group, Department of Clinical Pathology, Melbourne Medical School, The University of Melbourne, Parkville, Australia; University of Melbourne Centre for Cancer Research, The University of Melbourne, Parkville, Australia.
Marios Giannakis, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States; Broad Institute of MIT and Harvard, Cambridge, MA, United States; Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States.
Stephen B Gruber, Department of Medical Oncology and Therapeutics Research and Center for Precision Medicine, City of Hope National Medical Center, Duarte, CA, United States.
Marc J Gunter, Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom; Nutrition and Metabolism Branch, International Agency for Research on Cancer, Lyon, France.
Tabitha A Harrison, Public Health Sciences Division, Seattle, WA, United States.
Michael Hoffmeister, Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (Deutsches Krebsforschungszentrum, DKFZ), Heidelberg, Germany.
Li Hsu, Public Health Sciences Division, Seattle, WA, United States; Department of Biostatistics, University of Washington, Seattle, WA, United States.
Wen-Yi Huang, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States.
Meredith A J Hullar, Public Health Sciences Division, Seattle, WA, United States.
Jeroen R Huyghe, Public Health Sciences Division, Seattle, WA, United States.
Brigid M Lynch, Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Victoria, Australia; Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Victoria, Australia.
Victor Moreno, Unit of Biomarkers and Susceptibility, Oncology Data Analytics Program, Catalan Institute of Oncology, L’Hospitalet del Llobregat, Barcelona, Spain; ONCOBELL Program, Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, Barcelona, Spain; Consortium for Biomedical Research in Epidemiology and Public Health, Madrid, Spain; Department of Clinical Sciences, Faculty of Medicine and health Sciences and Universitat de Barcelona Institute of Complex Systems, University of Barcelona, L’Hospitalet de Llobregat, Barcelona, Spain.
Neil Murphy, Nutrition and Metabolism Branch, International Agency for Research on Cancer, Lyon, France.
Christina C Newton, Department of Population Science, American Cancer Society, Atlanta, GA, United States.
Jonathan A Nowak, Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States.
Mireia Obón-Santacana, Unit of Biomarkers and Susceptibility, Oncology Data Analytics Program, Catalan Institute of Oncology, L’Hospitalet del Llobregat, Barcelona, Spain; ONCOBELL Program, Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, Barcelona, Spain; Consortium for Biomedical Research in Epidemiology and Public Health, Madrid, Spain; Department of Clinical Sciences, Faculty of Medicine and health Sciences and Universitat de Barcelona Institute of Complex Systems, University of Barcelona, L’Hospitalet de Llobregat, Barcelona, Spain.
Shuji Ogino, Department of Epidemiology, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA, United States; Broad Institute of MIT and Harvard, Cambridge, MA, United States; Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States; Tokyo Medical and Dental University (Institute of Science Tokyo), Tokyo, Japan.
Conghui Qu, Public Health Sciences Division, Seattle, WA, United States.
Stephanie L Schmit, Genomic Medicine Institute, Cleveland Clinic, Cleveland, OH, United States; Population and Cancer Prevention Program, Case Comprehensive Cancer Center, Cleveland, OH, United States.
Robert S Steinfelder, Public Health Sciences Division, Seattle, WA, United States.
Wei Sun, Public Health Sciences Division, Seattle, WA, United States.
Claire E Thomas, Public Health Sciences Division, Seattle, WA, United States.
Amanda E Toland, Departments of Cancer Biology and Genetics, The Ohio State University, Comprehensive Cancer Center, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States.
Quang M Trinh, Ontario Institute for Cancer Research, Toronto, Ontario, Canada.
Tomotaka Ugai, Department of Epidemiology, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA, United States.
Caroline Y Um, Department of Population Science, American Cancer Society, Atlanta, GA, United States.
Bethany Van Guelpen, Department of Diagnostics and Intervention, Oncology Unit, Umeå University, Umeå, Sweden; Wallenberg Centre for Molecular Medicine, Umeå University, Umeå, Sweden.
Syed H Zaidi, Ontario Institute for Cancer Research, Toronto, Ontario, Canada.
Robert E Schoen, Department of Medicine and Epidemiology, University of Pittsburgh Medical Center, Pittsburgh, PA, United States.
Michael O Woods, Discipline of Genetics, Memorial University of Newfoundland, St John’s, Canada.
Hermann Brenner, Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (Deutsches Krebsforschungszentrum, DKFZ), Heidelberg, Germany; German Cancer Consortium (Deutsches Konsortium für Translationale Krebsforschung, DKTK), German Cancer Research Center (Deutsches Krebsforschungszentrum, DKFZ), Heidelberg, Germany.
Laura Andreson, Health Research Methods, Evidence and Impact, McMaster University, Hamilton, Ontario, Canada.
Andrew J Pellatt, Intermountain Health, Salt Lake City, UT, United States.
Ulrike Peters, Public Health Sciences Division, Seattle, WA, United States; Department of Epidemiology, University of Washington School of Public Health, Seattle, WA, United States.
Amanda I Phipps, Public Health Sciences Division, Seattle, WA, United States; Department of Epidemiology, University of Washington School of Public Health, Seattle, WA, United States.
Konstantinos K Tsilidis, Department of Hygiene and Epidemiology, University of Ioannina, Ioannina, Greece; Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom.
Author contributions
Christos V. Chalitsios (Conceptualization, Data curation, Formal analysis, Investigation, Methodology, Software, Visualization, Writing—original draft, Writing—review & editing), Georgios Markozannes (Methodology, Writing—review & editing), Elom K. Aglago (Writing—review & editing), Sonja I. Berndt (Writing—review & editing), Daniel D. Buchanan (Writing—review & editing), Peter T. Campbell (Writing—review & editing), Yin Cao (Writing—review & editing), Andrew T. Chan (Writing—review & editing), Niki Dimou (Writing—review & editing), David A. Drew (Writing—review & editing), Amy J. French (Writing—review & editing), Peter Georgeson (Writing—review & editing), Marios Giannakis (Writing—review & editing), Stephen B. Gruber (Writing—review & editing), Marc J. Gunter (Writing—review & editing), Tabitha A. Harrison (Writing—review & editing), Michael Hoffmeister (Writing—review & editing), Li Hsu (Writing—review & editing), Wen-Yi Huang (Writing—review & editing), Meredith A.J. Hullar (Writing—review & editing), Jeroen R. Huyghe (Writing—review & editing), Brigid M. Lynch (Writing—review & editing), Victor Moreno (Writing—review & editing), Neil Murphy (Writing—review & editing), Christina C. Newton (Writing—review & editing), Jonathan A. Nowak (Writing—review & editing), Mireia Obón-Santacana (Writing—review & editing), Shuji Ogino (Writing—review & editing), Conghui Qu (Writing—review & editing), Stephanie L. Schmit (Writing—review & editing), Robert S. Steinfelder (Writing—review & editing), Wei Sun (Writing—review & editing), Claire E. Thomas (Writing—review & editing), Amanda E. Toland (Writing—review & editing), Quang M. Trinh (Writing—review & editing), Tomotaka Ugai (Writing—review & editing), Caroline Y. Um (Writing—review & editing), Bethany van Guelpen (Writing—review & editing), Syed H. Zaidi (Writing—review & editing), Robert E. Schoen (Writing—review & editing), Michael O. Woods (Writing—review & editing), Hermann Brenner (Writing—review & editing), Laura Andreson (Writing—review & editing), Andrew J. Pellatt (Writing—review & editing), Ulrike Peters (Conceptualization, Data curation, Supervision, Writing—review & editing), Amanda I. Phipps (Conceptualization, Data curation, Supervision, Writing—review & editing), and Konstantinos K. Tsilidis (Conceptualization, Methodology, Supervision, Writing—review & editing)
Supplementary material
Supplementary material is available at JNCI Cancer Spectrum online.
Funding
GECCO: National Cancer Institute (NCI), National Institutes of Health (NIH), US Department of Health and Human Services (U01 CA137088, R01 CA176272). Genotyping/Sequencing services were provided by the Center for Inherited Disease Research (CIDR) contract number HHSN268201700006I and HHSN268201200008I. This research was funded in part through the NIH/NCI Cancer Center Support Grant P30 CA015704. Scientific Computing Infrastructure at Fred Hutch funded by ORIP grant S10OD028685.
CCFR: The CCFR (www.coloncfr.org) is supported in part by funding from the NCI, NIH (award U01 CA167551). Support for case ascertainment was provided in part from the Surveillance, Epidemiology, and End Results (SEER) Program and the following US state cancer registries: Arizona, Colorado, Minnesota, North Carolina, New Hampshire; and by the Victoria Cancer Registry (Australia) and Ontario Cancer Registry (Canada). The CCFR Set-1 (Illumina 1M/1M-Duo) and Set-2 (Illumina Omni1-Quad) scans were supported by NIH awards U01 CA122839 and R01 CA143237 (to GC). The CCFR Set-3 (Affymetrix Axiom CORECT Set array) was supported by NIH award U19 CA148107 and R01 CA81488 (to SBG). The CCFR Set-4 (Illumina OncoArray 600K SNP array) was supported by NIH award U19 CA148107 (to SBG) and by the CIDR, which is funded by the NIH to the Johns Hopkins University, contract number HHSN268201200008I. Additional funding for the Ontario Colorectal Cancer Family Registry (OFCCR)/ARCTIC was through award GL201-043 from the Ontario Research Fund (to BWZ), award 112746 from the Canadian Institutes of Health Research (to TJH), through a Cancer Risk Evaluation (CaRE) Program grant from the Canadian Cancer Society (to SG), and through generous support from the Ontario Ministry of Research and Innovation. The SFCCR Illumina HumanCytoSNP array was supported in part through NCI/NIH awards U01/U24 CA074794 and R01 CA076366 (to PAN). The content of this manuscript does not necessarily reflect the views or policies of the NCI, NIH, or any of the collaborating centers in the CCFR, nor does mention of trade names, commercial products, or organizations imply endorsement by the US government, any cancer registry, or the CCFR.
CPS-II: The American Cancer Society funds the creation, maintenance, and updating of the CPS-II cohort. The study protocol was approved by the institutional review boards of Emory University and those of participating registries as required.
DACHS: This work was supported by the German Research Council (BR 1704/6-1, BR 1704/6-3, BR 1704/6-4, CH 117/1-1, HO 5117/2-1, HE 5998/2-1, KL 2354/3-1, RO 2270/8-1 and BR 1704/17-1), the Interdisciplinary Research Program of the National Center for Tumor Diseases, Germany, and the German Federal Ministry of Education and Research (01KH0404, 01ER0814, 01ER0815, 01ER1505A and 01ER1505B).
DALS: National Institutes of Health (R01 CA048998 to M. L. Slattery).
EDRN: This work is funded and supported by the NCI, EDRN Grant (U01-CA152753).
EPIC: The coordination of EPIC is financially supported by International Agency for Research on Cancer and by the Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, which has additional infrastructure support provided by the National Institute for Health and Care Research (NIHR) Imperial Biomedical Research Centre. The national cohorts are supported by the Danish Cancer Society (Denmark); Ligue Contre le Cancer, Institut Gustave Roussy, Mutuelle Générale de l’Education Nationale, Institut National de la Santé et de la Recherche Médicale (INSERM) (France); German Cancer Aid, German Cancer Research Center, German Institute of Human Nutrition Potsdam- Rehbruecke (DIfE), Federal Ministry of Education and Research (BMBF) (Germany); Associazione Italiana per la Ricerca sul Cancro-AIRC-Italy, Compagnia di SanPaolo and National Research Council (Italy); Dutch Ministry of Public Health, Welfare and Sports (VWS), Netherlands Cancer Registry (NKR), LK Research Funds, Dutch Prevention Funds, Dutch ZON (Zorg Onderzoek Nederland), World Cancer Research Fund (WCRF), Statistics Netherlands (The Netherlands); Health Research Fund (FIS) - Instituto de Salud Carlos III (ISCIII), regional governments of Andalucía, Asturias, Basque Country, Murcia and Navarra, and the Catalan Institute of Oncology—ICO (Spain); Swedish Cancer Society, Swedish Research Council and Region Skåne and Region Västerbotten (Sweden); Cancer Research UK (14136 to EPIC-Norfolk; C8221/A29017 to EPIC-Oxford), Medical Research Council (1000143 to EPIC-Norfolk; MR/M012190/1 to EPIC-Oxford) (United Kingdom).
Harvard cohorts: HPFS is supported by the National Institutes of Health (P01 CA055075, UM1 CA167552, U01 CA167552, R01 CA137178, R01 CA151993, and R35 CA197735), NHS by the NIH (P01 CA087969, UM1 CA186107, R01 CA137178, R01 CA151993, and R35 CA197735).
Melbourne Collaborative Cohort Study (MCCS): The MCCS cohort recruitment was funded by VicHealth and Cancer Council Victoria. The MCCS was further augmented by Australian National Health and Medical Research Council grants 209057, 396414, and 1074383 and by infrastructure provided by Cancer Council Victoria. Cases and their vital status were ascertained through the Victorian Cancer Registry.
Newfoundland Case-Control Study (NFCCR): This work was supported by an Interdisciplinary Health Research Team award from the Canadian Institutes of Health Research (CRT 43821); the NIH, US Department of Health and Human Services (U01 CA074783); and NCI Canada grants (18223 and 18226). The authors wish to acknowledge the contribution of Alexandre Belisle and the genotyping team of the McGill University and Génome Québec Innovation Centre, Montréal, Canada, for genotyping the Sequenom panel in the NFCCR samples. Funding was provided to Michael O. Woods by the Canadian Cancer Society Research Institute.
NSHDS: The research was supported by Biobank Sweden through funding from the Swedish Research Council (VR 2017-00650, VR 2017-01737), the Swedish Cancer Society (CAN 2017/581), Region Västerbotten (VLL-841671, VLL-833291), Knut and Alice Wallenberg Foundation (VLL-765961), and the Lion’s Cancer Research Foundation (several grants) and Insamlingsstiftelsen, both at Umeå University.
Conflicts of interest
U.P. was a consultant with AbbVie, and her husband holds individual stocks in the following companies: BioNTech SE—ADR, Amazon, CureVac BV, NanoString Technologies, Google/Alphabet Inc Class C, NVIDIA Corp, and Microsoft Corp. B.L., who is a JNCI Cancer Spectrum editorial board member and co-author on this paper, was not involved in the editorial review or decision to publish the manuscript.
Data availability
Tumor markers and epidemiologic data are available on request and permission. Please contact gecco@fredhutch.org to request the standardized proposal form. The principal investigators of each contributing study will evaluate and approve the proposal, and data access will be managed centrally.
References
- 1. Tsoi KKF, Pau CYY, Wu WKK, Chan FKL, Griffiths S, Sung JJY. Cigarette smoking and the risk of colorectal cancer: a meta-analysis of prospective cohort studies. Clin Gastroenterol Hepatol. 2009;7:682-688.e5. [DOI] [PubMed] [Google Scholar]
- 2. Papadimitriou N, Qu C, Harrison TA, et al. Body size and risk of colorectal cancer molecular defined subtypes and pathways: Mendelian randomization analyses. eBioMedicine. 2024;101:105010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Murphy N, Newton CC, Song M, et al. Body mass index and molecular subtypes of colorectal cancer. JNCI J Natl Cancer Inst. 2023;115:165-173. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Fedirko V, Tramacere I, Bagnardi V, et al. Alcohol drinking and colorectal cancer risk: an overall and dose–response meta-analysis of published studies. Ann Oncol. 2011;22:1958-1972. [DOI] [PubMed] [Google Scholar]
- 5. Stern MC, Sanchez Mendez J, Kim AE, et al. Genome-wide gene–environment interaction analyses to understand the relationship between red meat and processed meat intake and colorectal cancer risk. Cancer Epidemiol Biomarkers Prev. 2024;33:400-410. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Papadimitriou N, Dimou N, Tsilidis KK, et al. Physical activity and risks of breast and colorectal cancer: a Mendelian randomisation analysis. Nat Commun. 2020;11:597. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Moore SC, Lee I-M, Weiderpass E, et al. Association of leisure-time physical activity with risk of 26 types of cancer in 1.44 million adults. JAMA Intern Med. 2016;176:816-825. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Kyu HH, Bachman VF, Alexander LT, et al. Physical activity and risk of breast cancer, colon cancer, diabetes, ischemic heart disease, and ischemic stroke events: systematic review and dose-response meta-analysis for the Global Burden of Disease Study 2013. BMJ. 2016;354:i3857. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Ulrich CM, Himbert C, Holowatyj AN, Hursting SD. Energy balance and gastrointestinal cancer: risk, interventions, outcomes and mechanisms. Nat Rev Gastroenterol Hepatol. 2018;15:683-698. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Bowers LW, Rossi EL, Flanagan CH, deGraffenried LA, Hursting SD. The role of the Insulin/IGF system in cancer: lessons learned from clinical trials and the energy balance-cancer link. Front Endocrinol. 2015;6:1-10. [Google Scholar]
- 11. Friedenreich CM, Neilson HK, Lynch BM. State of the epidemiological evidence on physical activity and cancer prevention. Eur J Cancer. 2010;46:2593-2604. [DOI] [PubMed] [Google Scholar]
- 12. McTiernan A. Mechanisms linking physical activity with cancer. Nat Rev Cancer. 2008;8:205-211. [DOI] [PubMed] [Google Scholar]
- 13. Zhang X, Ashcraft KA, Betof Warner A, Nair SK, Dewhirst MW. Can exercise-induced modulation of the tumor physiologic microenvironment improve antitumor immunity? Cancer Res. 2019;79:2447-2456. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Fernandez DM, Clemente JC, Giannarelli C. Physical activity, immune system, and the microbiome in cardiovascular disease. Front Physiol. 2018;9:763. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Allen JM, Mailing LJ, Niemiro GM, et al. Exercise alters gut microbiota composition and function in lean and obese humans. Med Sci Sports Exerc. 2018;50:747-757. [DOI] [PubMed] [Google Scholar]
- 16. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68:394-424. [DOI] [PubMed] [Google Scholar]
- 17. Nosho K, Kure S, Irahara N, et al. A prospective cohort study shows unique epigenetic, genetic, and prognostic features of synchronous colorectal cancers. Gastroenterology. 2009;137:1609-1620.e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Phipps AI, Limburg PJ, Baron JA, et al. Association between molecular subtypes of colorectal cancer and patient survival. Gastroenterology. 2015;148:77-87.e2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Kocarnik JM, Shiovitz S, Phipps AI. Molecular phenotypes of colorectal cancer and potential clinical applications. Gastroenterol Rep (Oxf). 2015;3:269-276. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Slattery ML, Anderson K, Curtin K, et al. Lifestyle factors and Ki-ras mutations in colon cancer tumors. Mutat Res 2001;483:73-81. [DOI] [PubMed] [Google Scholar]
- 21. Jass JR. HNPCC and Sporadic MSI-H colorectal cancer: a review of the morphological similarities and differences. Fam Cancer 2004;3:93-100. [DOI] [PubMed] [Google Scholar]
- 22. Aasebø K, Bruun J, Bergsland CH, et al. Prognostic role of tumour-infiltrating lymphocytes and macrophages in relation to MSI, CDX2 and BRAF status: a population-based study of metastatic colorectal cancer patients. Br J Cancer. 2022;126:48-56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Kopetz S, Yoshino T, Van Cutsem E, et al. Encorafenib, cetuximab and chemotherapy in BRAF-mutant colorectal cancer: a randomized phase 3 trial. Nat Med. 2025. [cited 2025 Feb 7]; Available from: https://www.nature.com/articles/s41591-024-03443-3 [Google Scholar]
- 24. Tabernero J, Grothey A, Van Cutsem E, et al. Encorafenib plus cetuximab as a new standard of care for previously treated BRAF V600E–Mutant Metastatic Colorectal Cancer: Updated Survival Results and Subgroup Analyses from the BEACON Study. J Clin Oncol. 2021;39:273-284. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Allegra CJ, Jessup JM, Somerfield MR, et al. American society of clinical oncology provisional clinical opinion: testing for KRAS gene mutations in patients with metastatic colorectal carcinoma to predict response to anti–epidermal growth factor receptor monoclonal antibody therapy. J Clin Oncol. 2009;27:2091-2096. [DOI] [PubMed] [Google Scholar]
- 26. Andre T, Elez E, Van Cutsem E, et al. ; CheckMate 8HW Investigators. Nivolumab plus Ipilimumab in Microsatellite-Instability–High Metastatic Colorectal Cancer. N Engl J Med. 2024;391:2014-2026. [DOI] [PubMed] [Google Scholar]
- 27. Diaz LA, Shiu K-K, Kim T-W, et al. ; KEYNOTE-177 Investigators. Pembrolizumab versus chemotherapy for microsatellite instability-high or mismatch repair-deficient metastatic colorectal cancer (KEYNOTE-177): final analysis of a randomised, open-label, phase 3 study. Lancet Oncol. 2022;23:659-670. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Calle EE, Rodriguez C, Jacobs EJ, et al. The American Cancer Society Cancer Prevention Study II Nutrition Cohort: Rationale, study design, and baseline characteristics. Cancer. 2002;94:500-511. [DOI] [PubMed] [Google Scholar]
- 29. Campbell PT, Deka A, Briggs P, et al. Establishment of the Cancer Prevention Study II Nutrition Cohort Colorectal Tissue Repository. Cancer Epidemiol Biomarkers Prev. 2014;23:2694-2702. [DOI] [PubMed] [Google Scholar]
- 30. Belanger CF, Hennekens CH, Rosner B, Speizer FE. The Nurses’ Health Study. Am J Nurs. 1978;78:1039-1040. [PubMed] [Google Scholar]
- 31. Brenner H, Chang–Claude J, Jansen L, Knebel P, Stock C, Hoffmeister M. Reduced risk of colorectal cancer up to 10 years after screening, surveillance, or diagnostic colonoscopy. Gastroenterology. 2014;146:709-717. [DOI] [PubMed] [Google Scholar]
- 32. Jia M, Jansen L, Walter V, et al. No association of CpG island methylator phenotype and colorectal cancer survival: population-based study. Br J Cancer. 2016;115:1359-1366. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Slattery ML, Friedman GD, Potter JD, Edwards S, Caan BJ, Samowitz W. A description of age, sex, and site distributions of colon carcinoma in three geographic areas. Cancer. 1996;78:1666-1670. [DOI] [PubMed] [Google Scholar]
- 34. Slattery ML, Berry TD, Potter J, Cann B. Diet diversity, diet composition, and risk of colon cancer (United States). Cancer Causes Control 1997;8:872-882. [DOI] [PubMed] [Google Scholar]
- 35. Boland CR, Thibodeau SN, Hamilton SR, et al. A national cancer institute workshop on microsatellite instability for cancer detection and familial predisposition: development of international criteria for the determination of microsatellite instability in colorectal cancer. Cancer Res. 1998;15:5248-5257. [Google Scholar]
- 36. Warth A, Kloor M, Schirmacher P, Bläker H. Genetics and epigenetics of small bowel adenocarcinoma: the interactions of CIN, MSI, and CIMP. Mod Pathol. 2011;24:564-570. [DOI] [PubMed] [Google Scholar]
- 37. Park SJ, Rashid A, Lee JH, Kim SG, Hamilton SR, Wu TT. Frequent CpG Island methylation in serrated adenomas of the colorectum. Am J Pathol. 2003;162:815-822. [Google Scholar]
- 38. Rashid A, Shen L, Morris JS, Issa JPJ, Hamilton SR. CpG Island methylation in colorectal adenomas. Am J Pathol. 2001;159:1129-1135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Jass JR. Classification of colorectal cancer based on correlation of clinical, morphological and molecular features. Histopathology. 2007;50:113-130. [DOI] [PubMed] [Google Scholar]
- 40. White IR, Royston P, Wood AM. Multiple imputation using chained equations: Issues and guidance for practice. Stat Med. 2011;30:377-399. [DOI] [PubMed] [Google Scholar]
- 41. Fernandez-Rozadilla C, Timofeeva M, Chen Z. 55:89–99.
- 42. Newcomb PA, Baron J, Cotterchio M, et al. ; Colon Cancer Family Registry. Colon cancer family registry: an international resource for studies of the genetic epidemiology of colon cancer. Cancer Epidemiol Biomarkers Prev. 2007;16:2331-2343. [DOI] [PubMed] [Google Scholar]
- 43. Amin W, Singh H, Dzubinski LA, Schoen RE, Parwani AV. Design and utilization of the colorectal and pancreatic neoplasm virtual biorepository: an early detection research network initiative. J Pathol Inform. 2010;1:22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Riboli E, Kaaks R. The EPIC project: rationale and study design. Int J Epidemiol. 1997;26 Suppl 1:S6-14. [DOI] [PubMed] [Google Scholar]
- 45. Milne RL, Fletcher AS, MacInnis RJ, et al. Cohort profile: The Melbourne Collaborative Cohort Study (Health 2020). Int J Epidemiol. 2017;46:1757-1757i. [DOI] [PubMed] [Google Scholar]
- 46. Dahlin AM, Palmqvist R, Henriksson ML, et al. The role of the CpG Island methylator phenotype in colorectal cancer prognosis depends on microsatellite instability screening status. Clin Cancer Res. 2010;16:1845-1855. [DOI] [PubMed] [Google Scholar]
- 47. Doherty A, Smith-Byrne K, Ferreira T, et al. GWAS identifies 14 loci for device-measured physical activity and sleep duration. Nat Commun. 2018;9:5257. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Davey Smith G, Ebrahim S. ‘Mendelian randomization’: can genetic epidemiology contribute to understanding environmental determinants of disease? *Int J Epidemiol. 2003;32:1-22. [DOI] [PubMed] [Google Scholar]
- 49. Davey Smith G, Hemani G. Mendelian randomization: genetic anchors for causal inference in epidemiological studies. Hum Mol Genet. 2014;23:R89-98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Bowden J, Del Greco M F, Minelli C, Davey Smith G, Sheehan N, Thompson J. A framework for the investigation of pleiotropy in two‐sample summary data Mendelian randomization. Stat Med. 2017;36:1783-1802. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Bowden J, Davey Smith G, Burgess S. Mendelian randomization with invalid instruments: effect estimation and bias detection through Egger regression. Int J Epidemiol. 2015;44:512-525. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Bowden J, Davey Smith G, Haycock PC, Burgess S. Consistent Estimation in Mendelian Randomization with Some Invalid Instruments Using a Weighted Median Estimator. Genet Epidemiol. 2016;40:304-314. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Hartwig FP, Davey Smith G, Bowden J. Robust inference in summary data Mendelian randomization via the zero modal pleiotropy assumption. Int J Epidemiol. 2017;46:1985-1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Verbanck M, Chen CY, Neale B, Do R. Detection of widespread horizontal pleiotropy in causal relationships inferred from Mendelian randomization between complex traits and diseases. Nat Genet. 2018;50:693-698. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Johnson CM, Wei C, Ensor JE, et al. Meta-analyses of colorectal cancer risk factors. Cancer Causes Control. 2013;24:1207-1222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Zhang X, Theodoratou E, Li X, et al. Genetically predicted physical activity levels are associated with lower colorectal cancer risk: a Mendelian randomisation study. Br J Cancer. 2021;124:1330-1338. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Mahmood S, MacInnis RJ, English DR, Karahalios A, Lynch BM. Domain-specific physical activity and sedentary behaviour in relation to colon and rectal cancer risk: a systematic review and meta-analysis. Int J Epidemiol. 2017;46:1797-1813. [DOI] [PubMed] [Google Scholar]
- 58. Park SY, Wilkens LR, Haiman CA, Le Marchand L. Physical activity and colorectal cancer risk by sex, race/ethnicity, and subsite: the multiethnic cohort study. Cancer Prev Res (Phila Pa). 2019;12:315-326. [Google Scholar]
- 59. Howard RA, Freedman DM, Park Y, Hollenbeck A, Schatzkin A, Leitzmann MF. Physical activity, sedentary behavior, and the risk of colon and rectal cancer in the NIH-AARP Diet and Health Study. Cancer Causes Control. 2008;19:939-953. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Shephard RJ, Shek PN. Associations between physical activity and susceptibility to cancer. Sports Med. 1998;26:293-315. [DOI] [PubMed] [Google Scholar]
- 61. Wang X, Amitay E, Harrison TA, et al. Association between smoking and molecular subtypes of colorectal cancer. JNCI Cancer Spectr. 2021;5:pkab056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Advani SM, Swartz MD, Loree J, et al. Epidemiology and molecular-pathologic characteristics of CpG Island Methylator Phenotype (CIMP) in colorectal cancer. Clin Colorectal Cancer. 2021;20:137-147.e1. [DOI] [PubMed] [Google Scholar]
- 63. Slattery ML, Curtin K, Sweeney C, et al. Diet and lifestyle factor associations with CpG island methylator phenotype and BRAF mutations in colon cancer. Int J Cancer. 2007;120:656-663. [DOI] [PubMed] [Google Scholar]
- 64. Lawlor DA, Tilling K, Davey Smith G. Triangulation in aetiological epidemiology. Int J Epidemiol 2016;45:1866-1886. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Adams SA, Matthews CE, Ebbeling CB, et al. The effect of social desirability and social approval on self-reports of physical activity. Am J Epidemiol 2005;161:389-398. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
Tumor markers and epidemiologic data are available on request and permission. Please contact gecco@fredhutch.org to request the standardized proposal form. The principal investigators of each contributing study will evaluate and approve the proposal, and data access will be managed centrally.


