Abstract
Background
Conventional observational studies have identified several potential therapeutic agents that may lower risk of colorectal cancer development. However, these studies are susceptible to unmeasured and residual confounding and reverse causation, undermining robust causal inference.
Methods
We used Mendelian randomization (MR), a genetic epidemiological method that can strengthen causal inference, to evaluate the effect of previously reported therapeutic agents on colorectal cancer risk, including medications, dietary micronutrients, and exogenous hormones. Genetic instruments were constructed using genome-wide association studies (GWASs) of molecular traits (e.g. circulating levels of protein drug targets, blood-based biomarkers of micronutrients, circulating levels of endogenous hormones). Using summary statistics from these GWASs and a colorectal cancer risk GWAS (cases=78,473, controls=107,143), we employed Wald ratios and inverse-variance weighted models to estimate causal effects.
Results
We found evidence for associations of genetically-proxied elevated omega-3 fatty acids (OR 1.10; 95% CI 1.03, 1.18; p=6.20x10-3) and reduced plasma ACE levels (OR 1.08; 95% CI 1.03, 1.13; p=9.36x10-4) with colorectal cancer risk. Findings for ACE inhibition were consistent across sensitivity analyses.
Conclusions
Reduced plasma ACE levels were robustly linked to increased colorectal cancer risk. Further work is required to better understand the mechanism behind this finding and whether this translates to adverse effects via medication use (i.e. ACE inhibitors).
Impact
These findings provide updated evidence on the role of previously reported therapeutic agents in colorectal cancer risk, helping to prioritise further evaluation of those agents with potential aetiological roles in cancer development.
Introduction
Colorectal cancer presents an increasing public health burden, with over 1.9 million new cases globally in 2022, predicted to rise to 3.2 million annually by 2040 (1, 2). Incidence has been increasing in low-and middle-income countries in particular, correlated with economic growth and adoption of a western lifestyle, and amongst a younger demographic (i.e. under the age of 50) in high-income countries (3–6). An individual’s risk of developing colorectal cancer is influenced by several factors, including hereditary genetic mutations and family history, which account for 5-10% of colorectal cancer cases, and a number of environmental and lifestyle factors (7–9). Whilst many of these risk factors are modifiable, such as elevated body mass index (BMI), physical inactivity, and alcohol consumption; lifestyle and behavioural changes can be difficult to implement. Screening and removal of polyps during colonoscopy in high-income countries has been an effective strategy for reducing colorectal cancer incidence and mortality in average-risk groups; however, given incomplete uptake (10–12), screening alone is insufficient to tackle overall disease burden, and additional prevention strategies are needed (13).
Therapeutic prevention is one strategy that could be used to reduce an individual’s risk of developing colorectal cancer. Therapeutic prevention involves administering a synthetic, natural, or biological agent that can prevent, reverse or delay the onset of disease (14, 15). These agents could be used for primary prevention in a healthy population (i.e. showing no signs of disease) to reduce their overall risk, or in individuals who have had polyps (a precursor to colorectal cancer) removed during colonoscopy screening, known as secondary prevention (15, 16). Examples of successful preventive therapy agents for colorectal cancer include aspirin, for which a daily dose has been shown to reduce cancer incidence (17). Given the potential adverse effects of aspirin, including gastrointestinal bleeding, aspirin is currently only recommended for groups at high-risk of developing colorectal cancer (e.g. individuals with Lynch syndrome), and so effective therapeutic agents with more favourable safety profiles for cancer prevention are needed (18). There has been great success of preventive therapy in other disease areas including cardiovascular disease; however, there are fewer examples of successful preventive therapy for cancer, in part due to the longer timeframe of cancer development and limited availability of short-term biomarkers of drug target efficacy (14, 15). For clinical trials to be conducted, significant epidemiological evidence is required to select preventive agents for testing (15). There have been many preventive agents linked to reduced risk of colorectal cancer in the observational epidemiological literature that are pharmacologically actionable (i.e. given as a supplement or drug), including dietary micronutrients, medications and exogenous hormones. However, findings from conventional observational studies can be susceptible to biases such as confounding, due to either unmeasured or imprecisely measured confounders, and reverse causation, where associations are driven by the outcome influencing the presumed exposure. It can therefore be difficult to distinguish true causal effects from spurious correlations, and the suitability of these agents as intervention targets is unclear.
Mendelian randomization uses germline genetic variants to instrument exposures of interest, here dietary micronutrients, medications and hormones. As germline genetic variants are randomly allocated at meiosis and fixed from conception, conventional sources of bias such as confounding and reverse causation should be minimised, strengthening causal inference (19–21). We used Mendelian randomization to reassess these previously identified observational relationships and provide evidence to support their causal nature (19–21).
Materials and Methods
We used a two-sample MR framework to evaluate the causal relevance of preventive agents that have been reported to be associated with reduced colorectal cancer risk in observational studies. We have previously published a protocol detailing our methods and proposed analyses for this study (Figure 1) (22). In brief, we first conducted a literature search of reviews of therapeutic prevention and colorectal cancer risk to identify potential preventive agents. The search strategy is available in our protocol (22) and articles were included if they reviewed observational studies conducted in humans. All reported preventive agents were extracted from each review. For each agent that we identified, we attempted to generate a genetic instrument for a corresponding molecular trait (e.g. for drugs, this was circulating levels of protein drug targets; for dietary micronutrients, this was blood-based biomarkers of micronutrients; and for exogenous hormones, this was circulating levels of endogenous hormones). For instrument selection, we searched for genome-wide association studies (GWASs) using the GWAS catalogue (23), the IEU Open GWAS (24), PubMed and the preprint servers medRxiv/bioRxiv, to identify the largest GWAS conducted in individuals of European ancestry and with complete summary data available. Study specific covariates and reported units for each study are available in Supplementary Table 1. For any exposures not reported in standardised units, we transformed these prior to analyses. We then conducted a two-sample MR analysis of these potential preventive agents and colorectal cancer risk, using a GWAS meta-analysis of colorectal cancer risk available in individuals of European ancestry (cases=78,473, controls=107,143) (adjusted for age, sex and 10 genetic principal components of ancestry (PCs)) (25). Between study heterogeneity was calculated using the I2 statistic and variants with I2 >65% were excluded. Effect estimates for genetically proxied micronutrients and hormones were scaled to reflect increasing levels, whilst for the protein drug targets, estimates were scaled to reflect decreasing levels, to mimic the hypothetical intervention (i.e. micronutrient supplementation, hormone replacement therapy, or inhibition of proteins by drugs). For all molecular traits found to have evidence for an association with colorectal cancer risk (defined as p<0.05), and for which we had >10 single-nucleotide polymorphisms (SNPs) in the instrument, we conducted ‘pleiotropy-robust’ methods to examine sensitivity of results to horizontal pleiotropy bias (i.e. when a genetic variant influences the outcome, either directly or indirectly, independently of the exposure). For any protein drug targets found to have an effect on colorectal cancer risk, we performed genetic colocalisation analyses using the ‘coloc’ package in R (26), to determine if there is a shared causal variant, which is necessary, although not sufficient, to infer a causal relationship between these traits. The posterior probabilities for a number of configurations were calculated: H0 = neither the protein drug target or colorectal cancer has a genetic association in the region, H1 = only the protein drug target has a genetic association in the region, H2 = only colorectal cancer has a genetic association in the region, H3 = both the protein drug target and colorectal cancer are associated, but with different causal variants, H4 = both the protein drug target and colorectal cancer are associated and share a single causal variant. A posterior probability of ≥0.5 was used to indicate support for a configuration.
Figure 1. Outline of methods used in these analyses.
Overview of methods and sensitivity analyses conducted in this study. Abbreviations: GWAS = genome-wide association study; MR = Mendelian randomization; colorectal cancer = colorectal cancer; FDR = false discovery rate; PRESSO = Pleiotropy RESidual Sum and Outlier; CAUSE = Causal Analysis Using Summary Effect estimates; Horse after the horseshoe prior.
We detail here where we have deviated from, or made additions to, our published protocol. Firstly, our protocol specified that to instrument protein drug targets and biomarkers we would allow weakly correlated SNPs (r2<0.1) in our genetic instrument and account for this by using a generalised IVW method (27), using a correlation matrix generated from a reference panel of 10,000 individuals from UK Biobank. To add to this, when instrumenting drug target biomarkers, we used a ±250kb region around the cognate gene for the protein drug target to generate these instruments. We deviated from this approach when constructing instruments for ACE inhibition, where we instrumented plasma levels of ACE using a region ±1MB either side of the ACE gene (28) and an LD (linkage disequilibrium) r2<0.001 to select independent SNPs. This approach is consistent with previously published work, investigating the effect of genetically proxied ACE inhibition using serum ACE levels, on cancer risk (29). For all other exposures (e.g. micronutrients and hormones), we used a genome-wide significance (p<5x10-8) threshold and an r2<0.001 to select instruments, as stated in the protocol. Secondly, to account for potential sex-specific genetic influences on hormone levels, we conducted MR analyses of hormone levels using colorectal cancer data stratified by sex (female: cases=26,843, controls=32,820; male: cases=31,288, controls=34,527) (30), which was not previously stated in the protocol. Thirdly, our protocol specified that a false-discovery rate (FDR) correction of 5% was used to define strong evidence. Given that our hypotheses were guided by observational evidence, we deviated from the protocol and interpreted a p-value threshold of <0.05 as indicative of some evidence of an association, carrying forward all findings reaching this threshold for sensitivity analyses. The FDR corrected finding, along with the results of sensitivity analyses, were used to inform our interpretation of findings in the discussion. We also conducted additional sensitivity analyses for correlated horizontal pleiotropy that was not previously specified in the protocol, including MR-Horse (31) and CAUSE (32). Correlated pleiotropy occurs when the genetic variants used to instrument an exposure, affect a confounder that acts on both the exposure and outcome. CAUSE attempts to determine if genetic associations for two traits are consistent with a causal effect by comparing whether a sharing model, allowing for horizontal pleiotropic effects, or a causal model fit the data better (32). Regarding the colocalisation analyses, we used the default parameters (e.g. the prior probabilities of the SNP being associated with the exposure, the outcome or both traits is specified as 1x10-4, 1x10-4 and 1x10-5, respectively), as specified in the protocol. We did not specify in the protocol that ±100kb either side of the lead SNP was used to define the region and a more stringent threshold of 5x10-6 was used to test p12 (the SNP being associated with both traits), as a sensitivity analysis. For any findings with evidence of an effect on colorectal cancer risk in the main analysis, we conducted MR analyses using colorectal cancer data stratified by anatomical subsite, including colon (cases=32,002, controls=64,159), rectal (cases=16,212, controls=64,159), proximal (cases=15,706, controls=64,159), and distal (cases=14,376, controls=64,159) (30), as specified in the protocol. Additionally, we conducted analyses restricted to early age at onset (<50 years at diagnosis) (cases=6,176, controls=65,829) (33). In addition, a z-test was used to assess differences in findings between anatomical subsites (i.e. colon and rectal, distal and proximal) and age at onset (i.e. early onset and overall), as well as differences between sexes in the hormone analysis. When performing z-tests we accounted for potential sample overlap in control groups by calculating decoupled standard errors for effect estimates (34–36). There was overlap between cases of early onset and overall colorectal cancer, however we did not account for this in the z-test.
The Strengthening the Reporting of Observational Studies in Epidemiology using Mendelian Randomization (STROBE-MR) (37) guidelines were used to structure the reporting of this study.
Results
Literature review
We identified over 40 potential preventive agents in our literature review (Supplementary Table 2). Of these, 18 had a corresponding molecular trait (e.g. levels of protein drug targets, circulating dietary biomarkers and circulating levels of endogenous hormones) that could be instrumented and for which GWAS summary statistics of direct measures of these traits were available in individuals of European ancestry (Table 1). Characteristics of genetic variants used to instrument each trait and estimates of r2 and F-statistic are presented in Supplementary Table 3.
Table 1. Potential preventive agents identified from reviews of observational studies of colorectal cancer risk and the corresponding molecular trait instrumented in Mendelian randomization analyses.
Preventive agents identified in the literature that had a corresponding molecular trait that was deemed instrumentable. These agents included dietary micronutrients, drugs and endogenous hormones. For each drug, the biological target of the drug was identified as it’s molecular trait, (e.g. the levels of the protein that the drug targets). For dietary micronutrients and endogenous hormones, molecular traits were identified that correspond to the agent (e.g. blood-based biomarkers for vitamin levels).
Abbreviations: GWAS = genome-wide association study; PMID = PubMed identifier; LDL = low-density lipoprotein; HMG-CoA = 3-hydroxy-3-methylglutaryl coenzyme A; NPC1L1 = NPC1 like intracellular cholesterol transporter 1; PCSK9 = proprotein convertase subtilisin/kexin type 9; IL23R = interleukin 23 receptor; IL12B = interleukin 12B; ACE = angiotensin-converting enzyme; SBP = systolic blood pressure; NCC = sodium chloride cotransporter; ADRB1 = β1 adrenoceptor; SHBG = sex hormone binding globulin.
| Preventive agent | Molecular trait | GWAS PMID | Sample size |
|---|---|---|---|
| Dietary long-chain omega-3 polyunsaturated fatty acids |
Circulating long-chain omega-3 polyunsaturated fatty acids |
35692035 (70) | 114999 |
| Dietary long-chain omega-6 polyunsaturated fatty acids |
Circulating long-chain omega-6 polyunsaturated fatty acids |
35692035 (70) | 114999 |
| Dietary calcium | Serum calcium levels | 33462484 (71) | 313387 |
| Dietary vitamin D | Serum 25 hydroxyvitamin D | 32059762 (72) | 443734 |
| Dietary folate levels | Serum folate levels | 30339177 (73) | 2232 |
| Dietary selenium | Circulating selenium levels | 23720494 (74) | 5477 |
| Dietary vitamin A | Serum retinol levels | 38374065 (75) | 22274 |
| Dietary vitamin C | Plasma vitamin C levels | 33203707 (76) | 52018 |
| Dietary vitamin E | Circulating alpha-tocopherol levels |
36635386 (77) | 8192 |
| Dietary β carotene | Circulating β carotene levels | 19185284 (78) | 1190 |
| Dietary magnesium | Serum magnesium levels | 20700443 (79) | 15366 |
| Statins | LDL cholesterol levels (due to HMGCR inhibition) |
24097068 (80) | 1320016 |
| LDL cholesterol levels (due to NPC1L1 inhibition) |
24097068 (80) | 1320016 | |
| LDL cholesterol levels (due to PCSK9 inhibition) |
24097068 (80) | 1320016 | |
| Guselkumab | Plasma IL23 protein levels (i.e. IL23R inhibition, IL12B inhibition) |
29875488 (81) | 3301 |
| Antihypertensives | Plasma ACE levels | 37794186 (82) | 54219 |
| SBP (due to NCC inhibition) | 30224653 (83) | 757601 | |
| SBP (due to ADRB1 inhibition) | 30224653 (83) | 757601 | |
| Testosterone | Bioavailable testosterone levels |
32042192 (84) | 188507 (women) 178782 (men) |
| Total testosterone levels | 32042192 (84) | 230454 (women) 194453 (men) |
|
| Sex hormone binding globulin | Serum SHBG levels | 32042192 (84) | 189473 (women) 180726 (men) |
| Progesterone/17-hydroxyprogesterone | Circulating progesterone levels | 34822396 (85) | 1877 (women) 2220 (men) |
| 17-hydroxyprogesterone levels | 34822396 (85) | 1329 (women) 2220 (men) |
Two-sample MR analyses
We took all circulating micronutrients and protein drug targets for which we could construct genetic instruments and, using MR, tested their effect on colorectal cancer risk (Figure 2) (Supplementary Table 4). Effect estimates are presented as follows: odds ratios (ORs) per standard deviation (SD) unit increase (for micronutrients and hormones) or decrease (for protein drug targets) in genetically proxied exposure; 95% confidence intervals; p-value.
Figure 2. Effect estimates from initial Mendelian randomization analyses of circulating micronutrients and protein drug targets on colorectal cancer risk.
Effect estimates are presented as odds ratios (ORs) per standard deviation (SD) unit increase (for micronutrients) or decrease (for protein drug targets) in genetically proxied exposure (95% confidence intervals). The odds ratio was calculated using the multiplicative random effects inverse variance weighted (IVW) method, except where the number of SNPs was limited (e.g. ≤3) in which case the fixed effects IVW was used. For those exposures instrumented by one SNP, the Wald ratio was used. Abbreviations: CI = confidence interval; SD = standard deviation; IL23R = interleukin 23 receptor; IL12B = interleukin 12B; ADRB1 = β1 adrenoceptor; NCC = ACE = angiotensin I converting enzyme; HMGCR = 3-hydroxy-3-methylglutaryl-CoA reductase; NPC1L = NPC1 like intracellular cholesterol transporter 1; PCSK9 = proprotein convertase subtilisin/kexin type 9; MR = Mendelian randomization; colorectal cancer = colorectal cancer.
We initially found evidence that genetically proxied omega-3 fatty acids increased colorectal cancer risk (1.10; 1.03, 1.18; 6.20x10-3). This finding was generally consistent across ‘pleiotropy-robust’ sensitivity analyses (i.e. confidence intervals for all estimates overlapped) (Supplementary Table 5) but just exceeded the FDR corrected p-value threshold (Supplementary Table 4). Leave-one-out analyses indicated that a single SNP, rs174564, may be driving the effect estimate for omega-3 fatty acids on colorectal cancer risk, as removal of this SNP attenuated the effect to the null (1.02; 0.92, 1.13; 0.72), although confidence intervals did overlap with the overall effect estimate (Supplementary Table 6). We found strong evidence that a decrease in plasma ACE levels, the mechanism of action of ACE inhibitors, increased colorectal cancer risk (OR per unit decrease in inverse-rank normalized protein expression: 1.08; 1.03, 1.13; 9.36x10-4) (Figure 2). There was an insufficient number of independent SNPs in the genetic instrument to conduct pleiotropy-robust analyses on ACE inhibition, but this result met the FDR corrected p-value threshold. We found some evidence that the effect of ACE inhibition was stronger in colon cancer than rectum, (1.12, 1.05, 1.19; 2x10-4 and 1.04; 0.98, 1.10; 0.213, respectively) (pdiff=0.071)) (Figure 3, Supplementary Table 7-8). We found some evidence that circulating calcium levels had a protective effect on colorectal cancer risk (0.86; 0.74, 1.00; 0.045), which was consistent across anatomical subsites (Figure 3). Although this finding did not meet the FDR corrected p-value threshold, it was consistent across most of the pleiotropy-robust methods (Supplementary Table 5), except for CAUSE which did not find evidence for a causal relationship (Supplementary Table 9).
Figure 3. Mendelian randomization effect estimates stratified by anatomical subsite and restricted to early age at onset for micronutrients and drug targets found to have an effect on overall colorectal cancer.
Effect estimates are presented as odds ratios (ORs) per standard deviation (SD) unit increase (for micronutrients) or decrease (for protein drug targets) in genetically proxied exposure (95% confidence intervals). The odds ratio was calculated using the multiplicative random effects inverse variance weighted (IVW) method, except where the number of SNPs was limited (e.g. ≤3) in which case the fixed effects IVW was used. For those exposures instrumented by one SNP, the Wald ratio was used. Effect estimates are scaled to reflect increasing micronutrient levels and decreasing protein levels. Abbreviations: CI = confidence interval; SD = standard deviation; ACE = angiotensin converting enzyme.
In analyses examining endogenous hormones we found evidence that genetically-proxied circulating progesterone had a protective effect on colorectal cancer risk in men (0.68; 0.49, 0.94; 0.019) (Figure 4) (Supplementary Table 10). However, this finding did not meet the FDR-corrected p-value threshold. We found little evidence of an effect of any other hormones on colorectal risk in either men or women. Progesterone in men was instrumented using 1 SNP, so we therefore could not conduct pleiotropy-robust sensitivity analyses.
Figure 4. Effect estimates from Mendelian randomization analyses of circulating hormones on colorectal cancer risk in men and women.
Effect estimates are presented as odds ratios (ORs) per standard deviation (SD) unit increase in genetically proxied hormone levels (95% confidence intervals). The odds ratio was calculated using the multiplicative random effects inverse variance weighted (IVW) method, except where the number of SNPs was limited (e.g. ≤3) in which case the fixed effects IVW was used. For those exposures instrumented by one SNP, the Wald ratio was used. Effect estimates are scaled to reflect increasing levels of circulating hormones. Abbreviations: SD = standard deviation; SHBG = sex hormone-binding globulin.
Multivariable Mendelian Randomization
The univariable IVW estimated effect for omega 3 fatty acids on colorectal cancer risk was OR=1.10, 95% CIs 1.03, 1.18, p=6.20x10-3. Given the genetic correlation between omega-3 and omega-6 fatty acids, estimated previously using LD score regression to be over 60% (38), and that the FADS SNP, rs174564, is strongly correlated with metabolism of both omega-3 and omega-6 fatty acids, it can be difficult to disentangle their independent effects on an outcome. We performed multivariable MR (MVMR) analyses to estimate the effect of omega-3 fatty acids whilst adjusting for omega-6 fatty acids (Supplementary Table 11). Given that the FADS SNP, rs174564, may have been driving the effect estimate for omega-3 fatty acids on colorectal cancer in the main analysis, we repeated this analysis removing this SNP from the omega-3 instrument. We also recalculated the univariable IVW estimate for omega-3 on colorectal cancer risk with this same SNP removed (Supplementary Table 11). The MVMR effect estimate for omega-3 with rs174564 included (OR=1.07, 95% CIs=0.96, 1.18, p=0.24) and excluded (OR=1.00, 95% CIs=0.86, 1.17, p=1.00) attenuated towards the null. The univariable IVW estimate for omega-3 excluding rs174564 (OR=1.04, 95% CIs=0.95, 1.14, p=0.19) also attenuated to the null.
Steiger filtering
Steiger filtering can test the causal direction of SNP effects to evaluate if effect estimates are being driven by reverse causation (i.e. where the causal effect of a SNP on the exposure is mediated by the outcome). If the variance explained by a SNP was larger for the molecular trait than for colorectal cancer, this was consistent with a scenario where the molecular trait causally influences colorectal cancer risk, rather than the reverse. For ACE, omega-3 fatty acids and calcium, all SNPs were found to explain more of the variance in the molecular trait, consistent with the molecular trait causally influencing colorectal cancer.
Colocalisation
The colocalisation analyses of ACE inhibition and colorectal cancer risk found strong evidence that there is a shared causal variant in this region (posterior probability H4=0.94) (Figure 5) (Supplementary Table 12). This finding was consistent when using a more stringent threshold for the prior probability that the SNP was associated with both traits (e.g. p12=5x10-6) (posterior probability H4=0.89).
Figure 5. Regional association plots showing association of genetic variants with ACE inhibition and colorectal cancer risk ±100kb from the lead SNP (rs4343) used to instrument ACE.
Regional association plots showing the genomic location of variants and their association (-log10(P)) with levels of ACE and colorectal cancer risk, generated using the LocusCompareR package. Posterior probability for a shared causal variant associated with both plasma ACE levels and colorectal cancer risk was 0.94, the regional association plots do not appear to support the presence of multiple independent variants driving the associations with either trait. Abbreviations: ACE = angiotensin-converting enzyme; chr = chromosome; Mb = megabase; colorectal cancer = colorectal cancer.
We attempted to perform SUSIE, which allows for multiple causal SNPs in the region, but no credible sets were found for colorectal cancer risk in the ACE locus as no SNPs in the colorectal cancer dataset reached the default minimum p-value threshold (1x10-6). SUSIE does not assess for evidence of an association in datasets where no SNPs fall below the minimum p-value threshold.
Discussion
This Mendelian randomization analysis investigated the effects of genetically proxied circulating micronutrients, hormone levels, and inhibition of protein drug targets on colorectal cancer risk. To our knowledge, this is the largest and most comprehensive appraisal of previously reported therapeutic agents for colorectal cancer prevention, conducted in individuals of European ancestry using Mendelian randomization. We found strong evidence that genetically proxied inhibition of ACE increased colorectal cancer risk. We initially found evidence that genetically proxied elevated circulating omega-3 fatty acids increased colorectal cancer risk, however further analysis suggested other factors may have been driving this effect. We also found weak evidence for a protective effect of genetically proxied elevated circulating calcium levels.
We found strong evidence that genetically proxied ACE inhibition had an adverse effect on colorectal cancer risk, supported by genetic colocalisation of these traits around the ACE gene region. There was suggestive evidence that this effect was stronger in colon cancer, compared to rectal, however these analyses had limited statistical power. These findings are consistent with a previously published MR study investigating the effect of genetically proxied inhibition of antihypertensive drugs targets and a number of cancers, including colorectal cancer (29). The latter study differed in the method employed for instrument generation, permitting correlated SNPs (r2 <0.1) located within 100kb from the ACE gene (resulting in 14 SNPs) from a smaller GWAS of serum ACE levels (n=4,174) than used in the current study. The previous study also used summary data from a smaller GWAS for colorectal cancer risk (cases=58,221, controls=67,694) that made up a subset of the larger GWAS used in the current study (cases=78,473, controls=107,143), which enabled more precise effect estimation in the current study. Observational studies have found conflicting evidence on the association of ACE inhibitor use and colorectal cancer risk, for example; a large population-based case-control study (cases=15,560, controls=62,525) reported an increased risk of colorectal cancer (OR: 1.30, 95% CI: 1.22, 1.39) among long-term ACE inhibitor users (defined as 1000 daily doses within the past 5 years) as compared to age and gender matched controls (39). Whilst a large, retrospective, cohort study of 1,693,297 people (cases=28,460) found no evidence for an association of cumulative duration of ACE inhibitor use with colorectal cancer risk (HR: 1.03, 95% CI: 0.99, 1.07) (40). A dose response meta-analysis of 7 observational studies, including case-control and cohort studies (total cases=15,220, total controls=1,565,018) found an association of ACE inhibitor use with reduced colorectal cancer risk (RR: 0.81, 95% CI: 0.70, 0.92), however there was substantial heterogeneity between studies (I2=71.1%) (41). Given that cancer is not the indication for ACE inhibitors, there have not been any RCTs of ACE inhibitor use and colorectal cancer risk specifically. A recent unpublished study investigating the effects of several drugs and cancer risk using a target trial emulation framework, found that Captopril, the generic name of an ACE inhibitor, was associated with an increased colorectal cancer risk (HR = 2.15, 95% CI = [1.81, 2.57]), as compared to control drugs (medRxiv 2024.05.29.24308170). As this finding was generated using an alternative methodology and was consistent with the current study, it provides “triangulation” of the link between genetically proxied ACE inhibition and colorectal cancer risk.
The potential carcinogenic mechanisms of ACE inhibition are difficult to pinpoint given that ACE has a number of functions and effects in the body. ACE is primarily responsible for converting angiotensin I into angiotensin II, however it is capable of cleaving several other substrates (42). Bradykinin is one of these substrates and is involved in tissue injury and inflammation, as well as potentially influencing the tumour microenvironment, so increased levels, due to inhibition of ACE, may therefore play a role in tumorigenesis (43). Substance P is another substrate cleaved by ACE, which is involved in stimulating pro-inflammatory cytokine production and binds the NK-1 receptor, found on colon adenocarcinoma cells, to facilitate cell proliferation and migration of tumour cells (43). Angiotensin II can regulate levels of transferrin receptors which have been found to promote colon tumorigenesis and progression (44, 45), which could suggest a potential mechanism for increased risk of colon cancer over rectal. It would be valuable in future work to further investigate these potential mechanistic pathways in relation to ACE inhibition and increasing cancer risk.
Genetically-proxied higher levels of circulating omega-3 fatty acids were found to have an adverse effect on colorectal cancer risk which is inconsistent with previous meta-analyses of observational studies that have found an inverse association of higher omega-3 fatty acids on colorectal cancer risk (46–49). However, it can be difficult to disentangle the effects of omega-3 and omega-6 fatty acids in MR analyses given the large genetic overlap between the fatty acids. This was demonstrated by the MVMR analyses finding attenuated effect estimates for omega-3 fatty acids when adjusting for omega-6. In addition to this, in the leave-one-out analysis, a single SNP, rs174564, was identified as driving the MR effect estimate. After removing rs174564 from the analysis, little effect of omega-3 on colorectal cancer risk was found. This SNP is located in the fatty acid desaturase (FADS) gene region, suggesting that activity of this enzyme accounts for the effect on colorectal cancer risk. As the desaturase enzymes encoded by the FADS genes are involved in rate-limiting steps in the biosynthesis of both omega-3 and omega-6 fatty acids, it is not clear if the effect of FADS variants on colorectal cancer risk is independent of omega-6. Previous work has looked at individual omega-3 fatty acids and colorectal cancer risk and also found little evidence for an effect after removing SNPs from the FADS region, including for overall increased omega-3 fatty acids (OR: 1.11, 95% CIs: 0.97-1.28, p=0.13) (36). An RCT found little evidence for an effect of supplementation with omega-3 fatty acid, eicosapentaenoic acid (EPA), on risk of having any colorectal adenomas (RR: 0.98, 95% CIs: 0.87-1.12), although secondary analyses did identify an effect on reduction of number of colorectal adenomas (incidence rate ratio (IRR): 0.86, 95% CIs: 0.74-0.99) (50).
We found weak evidence that genetically proxied circulating calcium levels had a protective effect on colorectal cancer risk and that the direction of effect remained consistent across sensitivity analyses. The modest effect we observed in this study was consistent across anatomical subsites, so it appears that this effect was not driven by an effect at a particular site. There has been some evidence from several systematic reviews and meta-analyses of randomized controlled trials (RCTs) that calcium supplementation has a modest preventive effect on recurrent colorectal adenomas. One systematic review and meta-analysis looked at four randomised, double-blind, placebo-controlled trials of calcium supplementation (ranging from 1200-2000 mg/d) with the number of participants ranging from 194 to 1523 (total participants in calcium group=1487, total participants in placebo group=1497) (51). This study found an overall modest protective effect of calcium supplementation on recurrence of colorectal adenomas (random-effects risk ratio (RR) = 0.87, 95% CI: 0.77-0.98, I2=38.7%) (51). Another systematic review and meta-analysis included two trials comparing supplemental calcium (ranging from 1200-2000 mg/d) versus placebo for recurrence of adenomas, in 354-832 participants (total participants in calcium group=585, total participants in placebo group=601) (52). This analysis also found a risk reduction in developing an adenoma in the calcium supplementation arm compared to placebo (random-effects RR:0.82, 95%CI: 0.69–0.98, I2=0%) (52). Another systematic review of RCTs found contradictory results, with some studies finding increased risk or no effect, however, they were unable to perform a meta-analysis due to high heterogeneity between studies (53). A meta-analysis of cohort studies, including six studies with a total of 920,837 participants (including 8,839 colorectal cancer cases), also found a protective association of calcium supplementation with colorectal cancer risk (random effects RR per 300 mg/d = 0.91, 95% CI = 0.86-0.98, I2 = 67%) (54). A recent large prospective study of diet and colorectal cancer risk in 542,778 participants from the Million Women Study (12,251 incident cases over 16.6 years) found a strong inverse association for dietary calcium (RR per 300 mg/day = 0.83, 95% CI 0.77–0.89, p<1x10-6) (55). Serum calcium levels are tightly regulated and therefore may not be reflective of dietary intake (56, 57), which could explain why the magnitude of effect in the current study, where we used instruments generated from serum levels, was much smaller than studies measuring the effect of dietary or supplemental calcium. As previous prevention trials have focussed on recurrent adenomas and, given the latency period from colorectal adenoma to carcinoma (i.e. up to 10 years) (58, 59), large, longer-term primary prevention trials are required to confirm the potential protective effects of calcium supplementation on colorectal cancer development.
There are some considerations to acknowledge when conducting MR studies of nutritional factors. Firstly, sample sizes of GWASs are often small and so there are limited genetic variants available to instrument traits. For example, when constructing an instrument for beta-carotene, only one SNP was available. Subsequent MR studies may also be underpowered to detect effects given the small effect sizes of genetic variants on micronutrients, often due to a small heritable component of dietary micronutrient levels. There is often a lack of biological understanding of how genetic instruments influence micronutrient levels, limiting our ability to understand if variants are likely to be valid instruments, and subsequently our interpretation of MR findings. Circulating levels of biomarkers may also not reflect cellular levels of these biomarkers and often do not correlate with dietary intake (60), therefore limiting the application of findings to dietary interventions.
We found little evidence for an effect of genetically-proxied inhibition of the protein targets of lipid-lowering medication (e.g. statins), on colorectal cancer risk, and effect estimates were not consistent in direction. This is in disagreement with previous conventional observational studies that have found that lipid-lowering medications are associated with reduced colorectal cancer risk (61, 62). This relationship is also supported by a recent MVMR study that found an adverse effect of LDL cholesterol on colorectal cancer, adjusted for high-density lipoprotein (HDL) cholesterol and triglycerides (63). However, in the current analysis, we may have been underpowered to detect an effect of lipid-lowering drug targets on colorectal cancer given the far fewer number of genetic variants available to instrument these traits as compared to LDL cholesterol levels.
One strength of this analysis was that when proxying the effect of drug targets, genetic variants were selected that were located within genes that encode the drug’s protein target, which should minimise risk of horizontal pleiotropy bias and reverse causation. For most of the protein drug targets, with the exception of ACE, we instrumented a downstream biomarker of that particular drug (e.g. for lipid-lowering drugs this was reduced LDL cholesterol levels, and for antihypertensives this was reduced systolic blood pressure (SBP)). An advantage of using these variants instead of genetic variants associated with circulating levels of proteins (i.e. protein quantitative trait loci (pQTL)) is that these biomarkers are further along the causal pathway to the outcome, so these variants are more likely to mimic the drug effect. For ACE, we used variants associated with circulating plasma levels of this protein (pQTLs). This approach is consistent with previously published work, investigating the effect of genetically proxied ACE inhibition on cancer risk (29). The top variant (rs4343) in the ACE instrument explains a significant proportion of the variance in levels of this protein (9%) and is also associated with systolic blood pressure. A limitation of this finding is that the magnitude of the effect of ACE inhibition on colorectal cancer may be underestimated, since the effect of ACE inhibitors on lowering ACE levels is likely more potent than the genetic effect from the variants used here as proxies for ACE levels. Given that the genetic instrument used for ACE levels was associated with lower systolic blood pressure, people with these variants may be less likely to develop hypertension and subsequently less likely to be prescribed and treated with pharmacological ACE inhibitors.
There are lower rates of colorectal cancer in women as compared to men, and it has been suggested that sex hormones may play a role in this disparity (6, 64). However, we found little evidence for an effect of most of the hormones tested on colorectal cancer risk. We found evidence that progesterone has a protective effect on colorectal cancer risk in men, however the confidence intervals for this effect were wide, indicating uncertainty around the true estimate. This finding also does not explain the sex disparity observed in colorectal cancer rates. A RCT examining the effect of taking a daily combined oestrogen and progestin tablet on colorectal cancer risk in women initially found evidence for a protective effect on colorectal cancer risk (hazard ratio (HR): 0.63, 95% CIs: 0.43-0.92) (65). However, this study later found this finding was due to diagnostic delays and those in the treatment group were more likely to be diagnosed with advanced disease (66). This is in agreement with our findings, that showed little evidence for an effect of progesterone on colorectal cancer risk in women. This analysis was limited as we were unable to conduct further sensitivity analyses for these findings given the small number of SNPs available to instrument progesterone.
These analyses were restricted to individuals of European ancestry and therefore findings may not be generalisable to other ancestry groups. This is particularly relevant in the setting of therapeutic agents, given that they may have different pharmacokinetic/pharmacodynamic (PKPD) properties depending on germline pharmacogenomic variants that vary across populations (67, 68). In future, when GWAS data is available in different populations for the molecular traits examined here, there would be value in repeating these analyses using a more diverse cohort.
Conclusion
These analyses found genetic evidence for a causal effect of several potential preventive agents on colorectal cancer risk. Reduced plasma ACE levels were robustly linked to increased colorectal cancer risk, and there was some evidence to support that elevated calcium and progesterone (in men) were inversely associated with colorectal cancer. However, given the limitations outlined here, further research is required to understand if our findings reflect the effects of the corresponding medication, supplement or exogenous hormone on colorectal cancer risk, and whether they can be translated to clinical interventions. Specifically, additional epidemiological evidence with non-overlapping sources of bias is needed to further evaluate the association between ACE inhibition and colorectal cancer risk, ideally with evidence from RCTs. Additionally, it would be useful to investigate whether different classes of ACE inhibitors have differing adverse outcomes in the long-term. This may be aided by further work investigating the potential mechanisms underlying the association found here. With new large-scale genetic and proteomic datasets becoming available, there is likely great potential for identifying novel targets for cancer prevention in future (69).
Supplementary Material
Acknowledgments
ASTERISK: We are very grateful to those without whom this project would not have existed. We also thank all those who agreed to participate in this study, including the patients and the healthy control persons, as well as all the physicians, technicians and students.
CCFR: The Colon CFR graciously thanks the generous contributions of their study participants, dedication of study staff, and the financial support from the U.S. 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).
CLUE II: We thank the participants of Clue I and 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 Cohort Studies.
Maryland Cancer Registry (MCR) 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 NU58DP007114, 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.
COLON and NQplus: the authors would like to thank the COLON and NQplus investigators at Wageningen University & Research and the involved clinicians in the participating hospitals.
CORSA: We kindly thank all individuals who agreed to participate in the CORSA study. Furthermore, we thank all cooperating physicians and students and the Biobank Graz of the Medical University of Graz.
CPS-II: The authors express sincere appreciation to all Cancer Prevention Study-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 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. Czech Republic CCS: We are thankful to all clinicians in major hospitals in the Czech Republic, without whom the study would not be practicable. We are also sincerely grateful to all patients participating in this study.
DACHS: We thank all participants and cooperating clinicians, and everyone who provided excellent technical assistance.
EDRN: We acknowledge all contributors to the development of the resource at University of Pittsburgh School of Medicine, Department of Gastroenterology, Department of Pathology, Hepatology and Nutrition and Biomedical Informatics.
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.
EPICOLON: We are sincerely grateful to all patients participating in this study who were recruited as part of the EPICOLON project. We acknowledge the Spanish National DNA Bank, Biobank of Hospital Clínic–IDIBAPS and Biobanco Vasco for the availability of the samples. The work was carried out (in part) at the Esther Koplowitz Centre, Barcelona.
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 (NPCR) and/or the National Cancer Institute’s Surveillance, Epidemiology, and End Results (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, Wyoming. The authors assume full responsibility for analyses and interpretation of these data.
Kentucky: We would like to acknowledge the staff at the Kentucky Cancer Registry. LCCS: We acknowledge the contributions of all who conducted this study which was originally reported as 10.1093/carcin/24.2.275.
NCCCS I & II: We would like to thank the study participants, and the NC Colorectal Cancer Study staff.
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.
PLCO: The authors thank the PLCO Cancer Screening Trial screening center investigators and the staff from Information Management Services Inc and Westat Inc. Most importantly, we thank the study participants for their contributions that made this study possible.
Cancer incidence data have been provided by the District of Columbia Cancer Registry, Georgia Cancer Registry, Hawaii Cancer Registry, Minnesota Cancer Surveillance System, Missouri Cancer Registry, Nevada Central Cancer Registry, Pennsylvania Cancer Registry, Texas Cancer Registry, Virginia Cancer Registry, and Wisconsin Cancer Reporting System. All are supported in part by funds from the Center for Disease Control and Prevention, National Program for Central Registries, local states or by the National Cancer Institute, Surveillance, Epidemiology, and End Results program. The results reported here and the conclusions derived are the sole responsibility of the authors.
SEARCH: We thank the SEARCH team
SELECT: We thank the research and clinical staff at the sites that participated on SELECT study, without whom the trial would not have been successful. We are also grateful to the 35,533 dedicated men who participated in SELECT.
WHI: The authors thank the WHI investigators and staff for their dedication, and the study participants for making the program possible. A full listing of WHI investigators can be found at: https://s3-us-west-2.amazonaws.com/www-whi-org/wp-content/uploads/WHI-Investigator-Long-List.pdf
This manuscript is the result of funding in whole or in part by the National Institutes of Health (NIH). It is subject to the NIH Public Access Policy. Through acceptance of this federal funding, NIH has been given a right to make this manuscript publicly available in PubMed Central upon the Official Date of Publication, as defined by NIH.
GECCO funding: Genetics and Epidemiology of Colorectal Cancer Consortium (GECCO): National Cancer Institute, National Institutes of Health, U.S. Department of Health and Human Services (U01 CA164930, R01 CA273198, R01 CA059045, R21 CA191312, R01201407). Genotyping/Sequencing services were provided by the Center for Inherited Disease Research (CIDR) contract number 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
ASTERISK: a Hospital Clinical Research Program (PHRC-BRD09/C) from the University Hospital Center of Nantes (CHU de Nantes) and supported by the Regional Council of Pays de la Loire, the Groupement des Entreprises Françaises dans la Lutte contre le Cancer (GEFLUC), the Association Anne de Bretagne Génétique and the Ligue Régionale Contre le Cancer (LRCC).
The ATBC Study is supported by the Intramural Research Program of the U.S. National Cancer Institute, National Institutes of Health, Department of Health and Human Services.
CLUE II funding was from the National Cancer Institute (U01 CA086308, Early Detection Research Network; P30 CA006973), National Institute on Aging (U01 AG018033), and the American Institute for Cancer Research.
ColoCare: This work was supported by the National Institutes of Health (grant numbers R01 CA189184 (Li/Ulrich), U01 CA206110 (Ulrich/Li/Siegel/Figueiredo/Colditz, 2P30CA015704- 40 (Gilliland), R01 CA207371 (Ulrich/Li)), the Matthias Lackas-Foundation, the German Consortium for Translational Cancer Research, and the EU TRANSCAN initiative.
The Colon Cancer Family Registry (CCFR, www.coloncfr.org) is supported in part by funding from the National Cancer Institute (NCI), National Institutes of Health (NIH) (award U01 CA167551). Support for case ascertainment was provided in part from the Surveillance, Epidemiology, and End Results (SEER) Program and the following U.S. state cancer registries: AZ, CO, MN, NC, NH; 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 Center for Inherited Disease Research (CIDR), which is funded by the NIH to the Johns Hopkins University, contract number HHSN268201200008I. Additional funding for the 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 Colon Cancer Family Registry (CCFR), nor does mention of trade names, commercial products, or organizations imply endorsement by the US Government, any cancer registry, or the CCFR.
COLON: The COLON study is sponsored by Wereld Kanker Onderzoek Fonds, including funds from grant 2014/1179 as part of the World Cancer Research Fund International Regular Grant Programme, by Alpe d’Huzes and the Dutch Cancer Society (UM 2012–5653, UW 2013-5927, UW2015-7946), and by TRANSCAN (JTC2012-MetaboCCC, JTC2013-FOCUS). The Nqplus study is sponsored by a ZonMW investment grant (98-10030); by PREVIEW, the project PREVention of diabetes through lifestyle intervention and population studies in Europe and around the World (PREVIEW) project which received funding from the European Union Seventh Framework Programme (FP7/2007–2013) under grant no. 312057; by funds from TI Food and Nutrition (cardiovascular health theme), a public–private partnership on precompetitive research in food and nutrition; and by FOODBALL, the Food Biomarker Alliance, a project from JPI Healthy Diet for a Healthy Life.
Colorectal Cancer Transdisciplinary (CORECT) Study: The CORECT Study was supported by the National Cancer Institute, National Institutes of Health (NCI/NIH), U.S. Department of Health and Human Services (grant numbers U19 CA148107, R01 CA081488, P30 CA014089, R01 CA197350; P01 CA196569; R01 CA201407; R01 CA242218), National Institutes of Environmental Health Sciences, National Institutes of Health (grant number T32 ES013678) and a generous gift from Daniel and Maryann Fong.
CORSA: The CORSA study was funded by Austrian Research Funding Agency (FFG) BRIDGE (grant 829675, to Andrea Gsur), the “Herzfelder’sche Familienstiftung” (grant to Andrea Gsur) and was supported by COST Action BM1206.
CPS-II: The American Cancer Society funds the creation, maintenance, and updating of the Cancer Prevention Study-II (CPS-II) cohort. The study protocol was approved by the institutional review boards of Emory University, and those of participating registries as required.
CRCGEN: Colorectal Cancer Genetics & Genomics, Spanish study was supported by Instituto de Salud Carlos III, co-funded by FEDER funds –a way to build Europe–(grants PI14-613 and PI09-1286), Agency for Management of University and Research Grants (AGAUR) of the Catalan Government (grant 2017SGR723), Junta de Castilla y León (grant LE22A10-2), the Spanish Association Against Cancer (AECC) Scientific Foundation grant GCTRA18022MORE and the Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), action Genrisk. Sample collection of this work was supported by the Xarxa de Bancs de Tumors de Catalunya sponsored by Pla Director d’Oncología de Catalunya (XBTC), Plataforma Biobancos PT13/0010/0013 and ICOBIOBANC, sponsored by the Catalan Institute of Oncology. We thank CERCA Programme, Generalitat de Catalunya for institutional support.
Czech Republic CCS: This work was supported by the Czech Science Foundation (21-04607X, 21-27902S), by the Grant Agency of the Ministry of Health of the Czech Republic (grants AZV NU21-07-00247 and AZV NU21-03-00145), and Charles University Research Fund (Cooperation 43-Surgical disciplines).
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 (NCT), 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 (IARC) and also by the Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London which has additional infrastructure support provided by the NIHR Imperial Biomedical Research Centre (BRC). The national cohorts are supported by: 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 (DKFZ), 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 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).
EPIC: The coordination of EPIC is financially supported by International Agency for Research on Cancer (IARC) and also by the Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London which has additional infrastructure support provided by the NIHR Imperial Biomedical Research Centre (BRC). The national cohorts are supported by: 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 (DKFZ), 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 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).
EPICOLON: This work was supported by grants from Fondo de Investigación Sanitaria/FEDER (PI08/0024, PI08/1276, PS09/02368, P111/00219, PI11/00681, PI14/00173, PI14/00230, PI17/00509, 17/00878, PI20/00113, PI20/00226, Acción Transversal de Cáncer), Xunta de Galicia (PGIDIT07PXIB9101209PR), Ministerio de Economia y Competitividad (SAF07-64873, SAF 2010-19273, SAF2014-54453R), Fundación Científica de la Asociación Española contra el Cáncer (GCB13131592CAST), Beca Grupo de Trabajo “Oncología” AEG (Asociación Española de Gastroenterología), Fundación Privada Olga Torres, FP7 CHIBCHA Consortium, Agència de Gestió d’Ajuts Universitaris i de Recerca (AGAUR, Generalitat de Catalunya, 2014SGR135, 2014SGR255, 2017SGR21, 2017SGR653), Catalan Tumour Bank Network (Pla Director d’Oncologia, Generalitat de Catalunya), PERIS (SLT002/16/00398, Generalitat de Catalunya), CERCA Programme (Generalitat de Catalunya) and COST Action BM1206 and CA17118. CIBERehd is funded by the Instituto de Salud Carlos III.
ESTHER/VERDI. This work was supported by grants from the Baden-Württemberg Ministry of Science, Research and Arts and the German Cancer Aid.
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 National Institutes of Health (P01 CA087969, UM1 CA186107, R01 CA137178, R01 CA151993, and R35 CA197735), and PHS by the National Institutes of Health (R01 CA042182).
Hawaii Adenoma Study: NCI grants R01 CA072520.
HCES-CRC: the Hwasun Cancer Epidemiology Study–Colon and Rectum Cancer (HCES-CRC; grants from Chonnam National University Hwasun Hospital, HCRI15011-1).
Kentucky: This work was supported by the following grant support: Clinical Investigator Award from Damon Runyon Cancer Research Foundation (CI-8); NCI R01CA136726. LCCS: The Leeds Colorectal Cancer Study was funded by the Food Standards Agency and Cancer Research UK Programme Award (C588/A19167).
MCCS cohort recruitment was funded by VicHealth and Cancer Council Victoria. The MCCS was further supported by Australian NHMRC grants 509348, 209057, 251553 and 504711 and by infrastructure provided by Cancer Council Victoria. Cases and their vital status were ascertained through the Victorian Cancer Registry (VCR) and the Australian Institute of Health and Welfare (AIHW), including the National Death Index and the Australian Cancer Database. BMLynch was supported by MCRF18005 from the Victorian Cancer Agency.
MEC: National Institutes of Health (R37 CA054281, P01 CA033619, and R01 CA063464).
MECC: This work was supported by the National Institutes of Health, U.S. Department of Health and Human Services (R01 CA081488, R01 CA197350, U19 CA148107, R01 CA242218, and a generous gift from Daniel and Maryann Fong.
MSKCC: The work at Sloan Kettering in New York was supported by the Robert and Kate Niehaus Center for Inherited Cancer Genomics and the Romeo Milio Foundation. Moffitt: This work was supported by funding from the National Institutes of Health (grant numbers R01 CA189184, P30 CA076292), Florida Department of Health Bankhead-Coley Grant 09BN-13, and the University of South Florida Oehler Foundation. Moffitt contributions were supported in part by the Total Cancer Care Initiative, Collaborative Data Services Core, and Tissue Core at the H. Lee Moffitt Cancer Center & Research Institute, a National Cancer Institute-designated Comprehensive Cancer Center (grant number P30 CA076292).
NCCCS I & II: We acknowledge funding support for this project from the National Institutes of Health, R01 CA066635 and P30 DK034987.
NFCCR: This work was supported by an Interdisciplinary Health Research Team award from the Canadian Institutes of Health Research (CRT 43821); the National Institutes of Health, U.S. Department of Health and Human Serivces (U01 CA074783); and National Cancer Institute of 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.
OSUMC: OCCPI funding was provided by Pelotonia and HNPCC funding was provided by the NCI (CA016058 and CA067941).
PLCO: Intramural Research Program of the Division of Cancer Epidemiology and Genetics and supported by contracts from the Division of Cancer Prevention, National Cancer Institute, NIH, DHHS. Funding was provided by National Institutes of Health (NIH), Genes, Environment and Health Initiative (GEI) Z01 CP 010200, NIH U01 HG004446, and NIH GEI U01 HG 004438.
SEARCH: The University of Cambridge has received salary support in respect of PDPP from the NHS in the East of England through the Clinical Academic Reserve. Cancer Research UK (C490/A16561); the UK National Institute for Health Research Biomedical Research Centres at the University of Cambridge.
SELECT: Research reported in this publication was supported in part by the National Cancer Institute of the National Institutes of Health under Award Numbers U10 CA037429 (CD Blanke), and UM1 CA182883 (CM Tangen/IM Thompson). The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.
SMS and REACH: This work was supported by the National Cancer Institute (grant P01 CA074184 to J.D.P. and P.A.N., grants R01 CA097325, R03 CA153323, and K05 CA152715 to P.A.N., and the National Center for Advancing Translational Sciences at the National Institutes of Health (grant KL2 TR000421 to A.N.B.-H.)
The Swedish Low-risk Colorectal Cancer Study: The study was supported by grants from the Swedish research council; K2015-55X-22674-01-4, K2008-55X-20157-03-3, K2006-72X-20157-01-2 and the Stockholm County Council (ALF project).
Swedish Mammography Cohort and Cohort of Swedish Men: This work is supported by the Swedish Research Council /Infrastructure grant, the Swedish Cancer Foundation, and the Karolinska Institute´s Distinguished Professor Award to Alicja Wolk.
UK Biobank: This research has been conducted using the UK Biobank Resource under Application Number 8614
VITAL: National Institutes of Health (K05 CA154337).
WHI: The WHI program is funded by the National Heart, Lung, and Blood Institute, National Institutes of Health, U.S. Department of Health and Human Services through contracts 75N92021D00001, 75N92021D00002, 75N92021D00003, 75N92021D00004, 75N92021D00005
Funding
EF is supported by a Wellcome Trust PhD studentship (grant number: 228277/Z/23/Z) on the Molecular, Genetic, Lifecourse Epidemiology programme (218495/Z/19/Z). RMM is a National Institute for Health Research Senior Investigator (NIHR202411). PCH and RMM are supported by a Cancer Research UK 25 (C18281/A29019) programme grant (the Integrative Cancer Epidemiology Programme). RMM is also supported by the NIHR Bristol Biomedical Research Centre which is funded by the NIHR (BRC-1215-20011) and is a partnership between University Hospitals Bristol and Weston NHS Foundation Trust and the University of Bristol. Department of Health and Social Care disclaimer: The views expressed are those of the author(s) and not necessarily those of the NHS, the NIHR or the Department of Health and Social Care.
Footnotes
Conflict of interest
RCG has a graduate scholarship from Pfizer. RCG has research funding from TD Bank. RCG has paid consulting or advisory roles for Astrazeneca, Eisai, Incyte, Knight Therapeutics, Guardant Health, and Ipsen. RCG is an unpaid consultant with Tempus. The other authors declare no potential conflicts of interest.
Data Availability
We obtained publicly available summary genetic association data for colorectal cancer from GECCO (https://www.ebi.ac.uk/gwas/publications/36539618). Approval was received to use restricted summary genetic association data (including for sex, anatomical subsite and early age at onset specific colorectal cancer) from GECCO consortia after submitting a proposal to GECCO (kafdem@fredhutch.org). We obtained publicly available summary genetic association data for all exposures, the GWAS PMID for each study used is presented in Table 1 and details for accessing summary data can be found in each paper.
References
- 1.International WCRF. Colorectal cancer statistics. [cited 2025 Oct 8]. Available from: https://www.wcrf.org/cancer-trends/colorectal-cancer-statistics/
- 2.Morgan E, Arnold M, Gini A, Lorenzoni V, Cabasag CJ, Laversanne M, et al. Global burden of colorectal cancer in 2020 and 2040: incidence and mortality estimates from GLOBOCAN. Gut. 2023;72(2):338–44. doi: 10.1136/gutjnl-2022-327736. [DOI] [PubMed] [Google Scholar]
- 3.Rawla P, Sunkara T, Barsouk A. Epidemiology of colorectal cancer: incidence, mortality, survival, and risk factors. Prz Gastroenterol. 2019;14(2):89–103. doi: 10.5114/pg.2018.81072. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Siegel RL, Torre LA, Soerjomataram I, Hayes RB, Bray F, Weber TK, et al. Global patterns and trends in colorectal cancer incidence in young adults. Gut. 2019;68(12):2179–85. doi: 10.1136/gutjnl-2019-319511. [DOI] [PubMed] [Google Scholar]
- 5.Vuik FE, Nieuwenburg SA, Bardou M, Lansdorp-Vogelaar I, Dinis-Ribeiro M, Bento MJ, et al. Increasing incidence of colorectal cancer in young adults in Europe over the last 25 years. Gut. 2019;68(10):1820–6. doi: 10.1136/gutjnl-2018-317592. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Arnold M, Sierra MS, Laversanne M, Soerjomataram I, Jemal A, Bray F. Global patterns and trends in colorectal cancer incidence and mortality. Gut. 2017;66(4):683–91. doi: 10.1136/gutjnl-2015-310912. [DOI] [PubMed] [Google Scholar]
- 7.Kaur RJ, Pichurin PN, Hines JM, Singh RJ, Grebe SK, Bancos I. Adrenal Cortical Carcinoma Associated With Lynch Syndrome: A Case Report and Review of Literature. J Endocr Soc. 2019;3(4):784–90. doi: 10.1210/js.2019-00050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Stoffel E, Mukherjee B, Raymond VM, Tayob N, Kastrinos F, Sparr J, et al. Calculation of risk of colorectal and endometrial cancer among patients with Lynch syndrome. Gastroenterology. 2009;137(5):1621–7. doi: 10.1053/j.gastro.2009.07.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Lynch HT, Chapelle Adl. Hereditary Colorectal Cancer. New England Journal of Medicine. 2003;348(10):919–32. doi: 10.1056/NEJMra012242. [DOI] [PubMed] [Google Scholar]
- 10.Cancer Research UK. Cancer in the UK 2020: Socio-economic deprivation. 2020;9 [Google Scholar]
- 11.Wales PH. Screening Division Inequalities Report 2020-21. 2022:8–10.
- 12.White A, Ironmonger L, Steele RJC, Ormiston-Smith N, Crawford C, Seims A. A review of sex-related differences in colorectal cancer incidence, screening uptake, routes to diagnosis, cancer stage and survival in the UK. BMC Cancer. 2018;18(1):906. doi: 10.1186/s12885-018-4786-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Levin B, Lieberman DA, McFarland B, Smith RA, Brooks D, Andrews KS, et al. Screening and surveillance for the early detection of colorectal cancer and adenomatous polyps, 2008: a joint guideline from the American Cancer Society, the US Multi-Society Task Force on Colorectal Cancer, and the American College of Radiology. CA Cancer J Clin. 2008;58(3):130–60. doi: 10.3322/CA.2007.0018. [DOI] [PubMed] [Google Scholar]
- 14.Steward WP, Brown K. Cancer chemoprevention: A rapidly evolving field. British Journal of Cancer. 2013;109(1):1–7. doi: 10.1038/bjc.2013.280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Serrano D, Bonanni B, Brown K. Therapeutic cancer prevention: achievements and ongoing challenges - a focus on breast and colorectal cancer. Mol Oncol. 2019;13(3):579–90. doi: 10.1002/1878-0261.12461. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Adami HO, Day NE, Trichopoulos D, Willett WC. Primary and secondary prevention in the reduction of cancer morbidity and mortality. European Journal of Cancer. 2001;37:118–27. doi: 10.1016/s0959-8049(01)00262-3. [DOI] [PubMed] [Google Scholar]
- 17.Rothwell PM, Wilson M, Elwin CE, Norrving B, Algra A, Warlow CP, et al. Long-term effect of aspirin on colorectal cancer incidence and mortality: 20-year follow-up of five randomised trials. Lancet. 2010;376(9754):1741–50. doi: 10.1016/S0140-6736(10)61543-7. [DOI] [PubMed] [Google Scholar]
- 18.NICE. Effectiveness of aspirin in the prevention of colorectal cancer in people with Lynch syndrome. National Guideline Alliance (UK); 2020. pp. 7–16. [PubMed] [Google Scholar]
- 19.Lawlor DA, Harbord RM, Sterne JA, Timpson N, Davey Smith G. Mendelian randomization: using genes as instruments for making causal inferences in epidemiology. Stat Med. 2008;27(8):1133–63. doi: 10.1002/sim.3034. [DOI] [PubMed] [Google Scholar]
- 20.Ebrahim S, Davey Smith G. Mendelian randomization: can genetic epidemiology help redress the failures of observational epidemiology? Hum Genet. 2008;123(1):15–33. doi: 10.1007/s00439-007-0448-6. [DOI] [PubMed] [Google Scholar]
- 21.Sanderson E, Glymour MM, Holmes MV, Kang H, Morrison J, Munafò MR, et al. Mendelian randomization. Nat Rev Methods Primers. 2022;2 doi: 10.1038/s43586-021-00092-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Fryer E, Martin R, Haycock P, Yarmolinsky J. Investigating the causal effect of previously reported therapeutic agents for colorectal cancer prevention: protocol for a Mendelian randomization analysis. Wellcome Open Research. 2024;9(30) doi: 10.12688/wellcomeopenres.20861.2. [version 2; peer review: 2 approved, 1 approved with reservations] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Sollis E, Mosaku A, Abid A, Buniello A, Cerezo M, Gil L, et al. The NHGRI-EBI GWAS Catalog: knowledgebase and deposition resource. Nucleic Acids Res. 2023;51(D1):D977–d85. doi: 10.1093/nar/gkac1010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Lyon MS, Andrews SJ, Elsworth B, Gaunt TR, Hemani G, Marcora E. The variant call format provides efficient and robust storage of GWAS summary statistics. Genome Biology. 2021;22(1):32. doi: 10.1186/s13059-020-02248-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Fernandez-Rozadilla C, Timofeeva M, Chen Z, Law P, Thomas M, Schmit S, et al. Deciphering colorectal cancer genetics through multi-omic analysis of 100,204 cases and 154,587 controls of European and east Asian ancestries. Nat Genet. 2023;55(1):89–99. doi: 10.1038/s41588-022-01222-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Giambartolomei C, Vukcevic D, Schadt EE, Franke L, Hingorani AD, Wallace C, et al. Bayesian test for colocalisation between pairs of genetic association studies using summary statistics. PLoS Genet. 2014;10(5):e1004383. doi: 10.1371/journal.pgen.1004383. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Yavorska OO, Burgess S. MendelianRandomization: an R package for performing Mendelian randomization analyses using summarized data. Int J Epidemiol. 2017;46(6):1734–9. doi: 10.1093/ije/dyx034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Fauman EB, Hyde C. An optimal variant to gene distance window derived from an empirical definition of cis and trans protein QTLs. BMC Bioinformatics. 2022;23(1):169. doi: 10.1186/s12859-022-04706-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Yarmolinsky J, Díez-Obrero V, Richardson TG, Pigeyre M, Sjaarda J, Paré G, et al. Genetically proxied therapeutic inhibition of antihypertensive drug targets and risk of common cancers: A mendelian randomization analysis. PLOS Medicine. 2022;19(2):e1003897. doi: 10.1371/journal.pmed.1003897. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Huyghe JR, Harrison TA, Bien SA, Hampel H, Figueiredo JC, Schmit SL, et al. Genetic architectures of proximal and distal colorectal cancer are partly distinct. Gut. 2021;70(7):1325–34. doi: 10.1136/gutjnl-2020-321534. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Grant AJ, Burgess S. A Bayesian approach to Mendelian randomization using summary statistics in the univariable and multivariable settings with correlated pleiotropy. The American Journal of Human Genetics. 2024;111(1):165–80. doi: 10.1016/j.ajhg.2023.12.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Morrison J, Knoblauch N, Marcus JH, Stephens M, He X. Mendelian randomization accounting for correlated and uncorrelated pleiotropic effects using genome-wide summary statistics. Nature Genetics. 2020;52(7):740–7. doi: 10.1038/s41588-020-0631-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Laskar RS, Qu C, Huyghe JR, Harrison T, Hayes RB, Cao Y, et al. Genome-wide association studies and Mendelian randomization analyses provide insights into the causes of early-onset colorectal cancer. Ann Oncol. 2024;35(6):523–36. doi: 10.1016/j.annonc.2024.02.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Han B, Duong D, Sul JH, de Bakker PI, Eskin E, Raychaudhuri S. A general framework for meta-analyzing dependent studies with overlapping subjects in association mapping. Hum Mol Genet. 2016;25(9):1857–66. doi: 10.1093/hmg/ddw049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Lin DY, Sullivan PF. Meta-analysis of genome-wide association studies with overlapping subjects. Am J Hum Genet. 2009;85(6):862–72. doi: 10.1016/j.ajhg.2009.11.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Haycock PC, Borges MC, Burrows K, Lemaitre RN, Burgess S, Khankari NK, et al. The association between genetically elevated polyunsaturated fatty acids and risk of cancer. eBioMedicine. 2023;91 doi: 10.1016/j.ebiom.2023.104510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Skrivankova VW, Richmond RC, Woolf BAR, Yarmolinsky J, Davies NM, Swanson SA, et al. Strengthening the Reporting of Observational Studies in Epidemiology Using Mendelian Randomization: The STROBE-MR Statement. Jama. 2021;326(16):1614–21. doi: 10.1001/jama.2021.18236. [DOI] [PubMed] [Google Scholar]
- 38.Zagkos L, Dib MJ, Pinto R, Gill D, Koskeridis F, Drenos F, et al. Associations of genetically predicted fatty acid levels across the phenome: A mendelian randomisation study. PLoS Med. 2022;19(12):e1004141. doi: 10.1371/journal.pmed.1004141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Hallas J, Christensen R, Andersen M, Friis S, Bjerrum L. Long term use of drugs affecting the renin-angiotensin system and the risk of cancer: a population-based case-control study. Br J Clin Pharmacol. 2012;74(1):180–8. doi: 10.1111/j.1365-2125.2012.04170.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Qi J, Bhatti P, Spinelli JJ, Murphy RA. Antihypertensive medications and risk of colorectal cancer in British Columbia. Front Pharmacol. 2023;14:1301423. doi: 10.3389/fphar.2023.1301423. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Chen X, Yi CH, Ya KG. Renin-angiotensin system inhibitor use and colorectal cancer risk and mortality: A dose-response meta analysis. J Renin Angiotensin Aldosterone Syst. 2020;21(3):1470320319895646. doi: 10.1177/1470320319895646. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Bernstein KE, Ong FS, Blackwell WL, Shah KH, Giani JF, Gonzalez-Villalobos RA, et al. A modern understanding of the traditional and nontraditional biological functions of angiotensin-converting enzyme. Pharmacol Rev. 2013;65(1):1–46. doi: 10.1124/pr.112.006809. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Okwan-Duodu D, Landry J, Shen XZ, Diaz R. Angiotensin-converting enzyme and the tumor microenvironment: mechanisms beyond angiogenesis. American Journal of Physiology-Regulatory, Integrative and Comparative Physiology. 2013;305(3):R205–R15. doi: 10.1152/ajpregu.00544.2012. [DOI] [PubMed] [Google Scholar]
- 44.Kim H, Villareal LB, Liu Z, Haneef M, Falcon DM, Martin DR, et al. Transferrin Receptor-Mediated Iron Uptake Promotes Colon Tumorigenesis. Adv Sci (Weinh) 2023;10(10):e2207693. doi: 10.1002/advs.202207693. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Chen YJ, Qian ZM, Sheng Y, Zheng J, Liu Y. Angiotensin II down-regulates transferrin receptor 1 and ferroportin 1 expression in Neuro-2a cells via activation of type-1 receptor. Neurosci Lett. 2020;716:134684. doi: 10.1016/j.neulet.2019.134684. [DOI] [PubMed] [Google Scholar]
- 46.Wang J, Zhang Y, Zhao L. Omega-3 PUFA intake and the risk of digestive system cancers: A meta-analysis of observational studies. Medicine (Baltimore) 2020;99(19):e20119. doi: 10.1097/MD.0000000000020119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Aglago EK, Huybrechts I, Murphy N, Casagrande C, Nicolas G, Pischon T, et al. Consumption of Fish and Long-chain n-3 Polyunsaturated Fatty Acids Is Associated With Reduced Risk of Colorectal Cancer in a Large European Cohort. Clin Gastroenterol Hepatol. 2020;18(3):654–66.:e6. doi: 10.1016/j.cgh.2019.06.031. [DOI] [PubMed] [Google Scholar]
- 48.Nguyen S, Li H, Yu D, Cai H, Gao J, Gao Y, et al. Dietary fatty acids and colorectal cancer risk in men: A report from the Shanghai Men’s Health Study and a meta-analysis. Int J Cancer. 2021;148(1):77–89. doi: 10.1002/ijc.33196. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Kim Y, Kim J. Intake or Blood Levels of n-3 Polyunsaturated Fatty Acids and Risk of Colorectal Cancer: A Systematic Review and Meta-analysis of Prospective Studies. Cancer Epidemiol Biomarkers Prev. 2020;29(2):288–99. doi: 10.1158/1055-9965.EPI-19-0931. [DOI] [PubMed] [Google Scholar]
- 50.Hull MA, Sprange K, Hepburn T, Tan W, Shafayat A, Rees CJ, et al. Eicosapentaenoic acid and aspirin, alone and in combination, for the prevention of colorectal adenomas (seAFOod Polyp Prevention trial): a multicentre, randomised, double-blind, placebo-controlled, 2 × 2 factorial trial. The Lancet. 2018;392(10164):2583–94. doi: 10.1016/S0140-6736(18)31775-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Bonovas S, Fiorino G, Lytras T, Malesci A, Danese S. Calcium supplementation for the prevention of colorectal adenomas: A systematic review and meta-analysis of randomized controlled trials. World J Gastroenterol. 2016;22(18):4594–603. doi: 10.3748/wjg.v22.i18.4594. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Carroll C, Cooper K, Papaioannou D, Hind D, Pilgrim H, Tappenden P. Supplemental calcium in the chemoprevention of colorectal cancer: a systematic review and meta-analysis. Clin Ther. 2010;32(5):789–803. doi: 10.1016/j.clinthera.2010.04.024. [DOI] [PubMed] [Google Scholar]
- 53.Cruz-Pierard SM, Nestares T, Amaro-Gahete FJ. Vitamin D and Calcium as Key Potential Factors Related to Colorectal Cancer Prevention and Treatment: A Systematic Review. Nutrients. 2022;14(22) doi: 10.3390/nu14224934. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Keum N, Aune D, Greenwood DC, Ju W, Giovannucci EL. Calcium intake and colorectal cancer risk: Dose–response meta-analysis of prospective observational studies. International Journal of Cancer. 2014;135(8):1940–8. doi: 10.1002/ijc.28840. [DOI] [PubMed] [Google Scholar]
- 55.Papier K, Bradbury KE, Balkwill A, Barnes I, Smith-Byrne K, Gunter MJ, et al. Diet-wide analyses for risk of colorectal cancer: prospective study of 12,251 incident cases among 542,778 women in the UK. Nature Communications. 2025;16(1):375. doi: 10.1038/s41467-024-55219-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Mallette LE. Regulation of Blood Calcium in Humans. Endocrinology and Metabolism Clinics of North America. 1989;18(3):601–10. [PubMed] [Google Scholar]
- 57.Sharma EYS. Physiology, Calcium. StatPearls Publishing; Treasure Island (FL): 2025. pp. 1–6. [PubMed] [Google Scholar]
- 58.Davies RJ, Miller R, Coleman N. Colorectal cancer screening: prospects for molecular stool analysis. Nature Reviews Cancer. 2005;5(3):199–209. doi: 10.1038/nrc1569. [DOI] [PubMed] [Google Scholar]
- 59.Haggar FA, Boushey RP. Colorectal cancer epidemiology: incidence, mortality, survival, and risk factors. Clin Colon Rectal Surg. 2009;22(4):191–7. doi: 10.1055/s-0029-1242458. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Landberg R, Karra P, Hoobler R, Loftfield E, Huybrechts I, Rattner JI, et al. Dietary biomarkers—an update on their validity and applicability in epidemiological studies. Nutrition Reviews. 2023;82(9):1260–80. doi: 10.1093/nutrit/nuad119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Katona BW, Weiss JM. Chemoprevention of Colorectal Cancer. Gastroenterology. 2020;158(2):368–88. doi: 10.1053/j.gastro.2019.06.047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Lepore Signorile M, Grossi V, Fasano C, Simone C. Colorectal Cancer Chemoprevention: A Dream Coming True? International Journal of Molecular Sciences. 2023;24(8):7597. doi: 10.3390/ijms24087597. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Chan WC, Liu L, Bouras E, Zuber V, Wen W, Long J, et al. Associations of blood lipids and LDL cholesterol lowering drug-targets with colorectal cancer risk: a Mendelian randomisation study. British Journal of Cancer. 2025;132(1):103–10. doi: 10.1038/s41416-024-02900-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.McMichael AJ, Potter JD. Reproduction, endogenous and exogenous sex hormones, and colon cancer: a review and hypothesis. J Natl Cancer Inst. 1980;65(6):1201–7. [PubMed] [Google Scholar]
- 65.Rossouw JE, Anderson GL, Prentice RL, LaCroix AZ, Kooperberg C, Stefanick ML, et al. Risks and benefits of estrogen plus progestin in healthy postmenopausal women: principal results From the Women’s Health Initiative randomized controlled trial. Jama. 2002;288(3):321–33. doi: 10.1001/jama.288.3.321. [DOI] [PubMed] [Google Scholar]
- 66.Simon MS, Chlebowski RT, Wactawski-Wende J, Johnson KC, Muskovitz A, Kato I, et al. Estrogen plus progestin and colorectal cancer incidence and mortality. J Clin Oncol. 2012;30(32):3983–90. doi: 10.1200/JCO.2012.42.7732. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Hernandez S, Hindorff LA, Morales J, Ramos EM, Manolio TA. Patterns of pharmacogenetic variation in nine biogeographic groups. Clin Transl Sci. 2024;17(9):e70017. doi: 10.1111/cts.70017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Sun G, Li YN, Davies JR, Block RC, Kothapalli KS, Brenna JT, et al. Fatty acid desaturase insertion-deletion polymorphism rs66698963 predicts colorectal polyp prevention by the n-3 fatty acid eicosapentaenoic acid: a secondary analysis of the seAFOod polyp prevention trial. Am J Clin Nutr. 2024;120(2):360–8. doi: 10.1016/j.ajcnut.2024.06.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Yarmolinsky J, Lee MA, Lau E, Moratalla-Navarro F, Vincent EE, Li-Gao R, et al. Proteogenomic and observational evidence implicate ANGPTL4 as a potential therapeutic target for colorectal cancer prevention. J Natl Cancer Inst. 2025;117(9):1836–47. doi: 10.1093/jnci/djaf137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Borges MC, Haycock PC, Zheng J, Hemani G, Holmes MV, Smith Davey, et al. Role of circulating polyunsaturated fatty acids on cardiovascular diseases risk: analysis using Mendelian randomization and fatty acid genetic association data from over 114,000 UK Biobank participants. BMC Med. 2022;20(1):210. doi: 10.1186/s12916-022-02399-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Sinnott-Armstrong N, Tanigawa Y, Amar D, Mars N, Benner C, Aguirre M, et al. Genetics of 35 blood and urine biomarkers in the UK Biobank. Nat Genet. 2021;53(2):185–94. doi: 10.1038/s41588-020-00757-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Manousaki D, Mitchell R, Dudding T, Haworth S, Harroud A, Forgetta V, et al. Genome-wide Association Study for Vitamin D Levels Reveals 69 Independent Loci. Am J Hum Genet. 2020;106(3):327–37. doi: 10.1016/j.ajhg.2020.01.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Shane B, Pangilinan F, Mills JL, Fan R, Gong T, Cropp CD, et al. The 677C→T variant of MTHFR is the major genetic modifier of biomarkers of folate status in a young, healthy Irish population. Am J Clin Nutr. 2018;108(6):1334–41. doi: 10.1093/ajcn/nqy209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Evans DM, Zhu G, Dy V, Heath AC, Madden PA, Kemp JP, et al. Genome-wide association study identifies loci affecting blood copper, selenium and zinc. Hum Mol Genet. 2013;22(19):3998–4006. doi: 10.1093/hmg/ddt239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Reay WR, Kiltschewskij DJ, Di Biase MA, Gerring ZF, Kundu K, Surendran P, et al. Genetic influences on circulating retinol and its relationship to human health. Nat Commun. 2024;15(1):1490. doi: 10.1038/s41467-024-45779-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Zheng JS, Luan J, Sofianopoulou E, Imamura F, Stewart ID, Day FR, et al. Plasma Vitamin C and Type 2 Diabetes: Genome-Wide Association Study and Mendelian Randomization Analysis in European Populations. Diabetes Care. 2021;44(1):98–106. doi: 10.2337/dc20-1328. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Chen Y, Lu T, Pettersson-Kymmer U, Stewart ID, Butler-Laporte G, Nakanishi T, et al. Genomic atlas of the plasma metabolome prioritizes metabolites implicated in human diseases. Nat Genet. 2023;55(1):44–53. doi: 10.1038/s41588-022-01270-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Ferrucci L, Perry JR, Matteini A, Perola M, Tanaka T, Silander K, et al. Common variation in the beta-carotene 15,15’-monooxygenase 1 gene affects circulating levels of carotenoids: a genome-wide association study. Am J Hum Genet. 2009;84(2):123–33. doi: 10.1016/j.ajhg.2008.12.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Meyer TE, Verwoert GC, Hwang SJ, Glazer NL, Smith AV, van Rooij FJ, et al. Genome-wide association studies of serum magnesium, potassium, and sodium concentrations identify six Loci influencing serum magnesium levels. PLoS Genet. 2010;6(8) doi: 10.1371/journal.pgen.1001045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Willer CJ, Schmidt EM, Sengupta S, Peloso GM, Gustafsson S, Kanoni S, et al. Discovery and refinement of loci associated with lipid levels. Nat Genet. 2013;45(11):1274–83. doi: 10.1038/ng.2797. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Sun BB, Maranville JC, Peters JE, Stacey D, Staley JR, Blackshaw J, et al. Genomic atlas of the human plasma proteome. Nature. 2018;558(7708):73–9. doi: 10.1038/s41586-018-0175-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Sun BB, Chiou J, Traylor M, Benner C, Hsu YH, Richardson TG, et al. Plasma proteomic associations with genetics and health in the UK Biobank. Nature. 2023;622(7982):329–38. doi: 10.1038/s41586-023-06592-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Evangelou E, Warren HR, Mosen-Ansorena D, Mifsud B, Pazoki R, Gao H, et al. Genetic analysis of over 1 million people identifies 535 new loci associated with blood pressure traits. Nat Genet. 2018;50(10):1412–25. doi: 10.1038/s41588-018-0205-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Ruth KS, Day FR, Tyrrell J, Thompson DJ, Wood AR, Mahajan A, et al. Using human genetics to understand the disease impacts of testosterone in men and women. Nat Med. 2020;26(2):252–8. doi: 10.1038/s41591-020-0751-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Pott J, Horn K, Zeidler R, Kirsten H, Ahnert P, Kratzsch J, et al. Sex-Specific Causal Relations between Steroid Hormones and Obesity-A Mendelian Randomization Study. Metabolites. 2021;11(11) doi: 10.3390/metabo11110738. [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
We obtained publicly available summary genetic association data for colorectal cancer from GECCO (https://www.ebi.ac.uk/gwas/publications/36539618). Approval was received to use restricted summary genetic association data (including for sex, anatomical subsite and early age at onset specific colorectal cancer) from GECCO consortia after submitting a proposal to GECCO (kafdem@fredhutch.org). We obtained publicly available summary genetic association data for all exposures, the GWAS PMID for each study used is presented in Table 1 and details for accessing summary data can be found in each paper.





