Chemopreventive strategies for sporadic colorectal cancer: a narrative review
Review Article

Chemopreventive strategies for sporadic colorectal cancer: a narrative review

Savanna Monson1, Pin-Jung Chen1, Alexandra Gangi2, Kevin Waters3, Sandrine Billet1, Andrew Hendifar1, Shelly Lu1,4, Jason A. Zell5, Jun Gong1

1Department of Medicine, Division of Medical Oncology, Cedars-Sinai Medical Center, Los Angeles, CA, USA; 2Department of Surgery, Division of Surgical Oncology, Cedars-Sinai Medical Center, Los Angeles, CA, USA; 3Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA; 4Department of Medicine, Division of Digestive and Liver Diseases, Cedars-Sinai Medical Center, Los Angeles, CA, USA; 5Department of Medicine, Division of Hematology/Oncology, University of California, Irvine, Orange, CA, USA

Contributions: (I) Conception and design: J Gong; (II) Administrative support: None; (III) Provision of study materials or patients: None; (IV) Collection and assembly of data: S Monson, PJ Chen, JA Zell, J Gong; (V) Data analysis and interpretation: S Monson, PJ Chen, S Lu, JA Zell, J Gong; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

Correspondence to: Jun Gong, MD. Department of Medicine, Division of Medical Oncology, Cedars-Sinai Medical Center, 8700 Beverly Blvd, AC 1042B, Los Angeles, CA 90048, USA. Email: jun.gong@cshs.org.

Background and Objective: Globally, colorectal cancer (CRC) is the third most commonly diagnosed cancer. CRC is known to arise from precancerous polyps known as adenomas. Although there are genetic syndromes (i.e., familial adenomatous polyposis syndrome) that carry a high lifetime risk of CRC, the majority of CRC cases are sporadic. Sporadic CRC develops via molecular events that occur early and frequently in the transformation of the normal colon epithelium to invasive cancer. Prevention of sporadic CRC (i.e., CRC chemoprevention) has been a topic of interest in the past decades due to its large public health burden. The objective of this study is to review various chemopreventive agents studied in randomized controlled trials (RCTs) to evaluate their effectiveness and safety in preventing sporadic CRC.

Methods: This review focuses on recent RCTs using potential chemopreventive agents to prevent sporadic CRC formation, either directly or through reduction of its known precursors such as adenomas or aberrant crypt foci (ACF) through interventions including nonsteroidal anti-inflammatory drugs (NSAIDs), vitamins and minerals, and metabolic agents.

Key Content and Findings: Multiple RCTs have been conducted in sporadic CRC chemoprevention. NSAIDs have proven promising in sporadic CRC chemoprevention but have been limited due to increased cardiovascular risk, particularly for celecoxib and rofecoxib.

Conclusions: There is active investigation into establishing the first sporadic CRC chemoprevention strategy. Building from previous trials, the choice of study population, selection of endpoints, safety and tolerability, availability of surrogate biomarkers, and novel mechanisms of action targeting the adenoma-carcinoma sequence remain important points to consider for all future trials of sporadic CRC chemoprevention.

Keywords: Colorectal cancer (CRC); chemoprevention; adenoma; primary prevention


Received: 27 July 2024; Accepted: 12 November 2024; Published online: 07 January 2025.

doi: 10.21037/tgh-24-97


Introduction

Colorectal cancer (CRC) now represents the first- and second-leading cause of death in U.S. men and women, respectively, under 50 years of age (1). Globally, CRC represents the third most diagnosed cancer with an estimated 1.9 million new cases and the second-leading cause of cancer death with an estimated 930,000 deaths in 2020 (2). Even though CRC constitutes a large public health burden, there exists no established method of primary prevention. Screening colonoscopies and preventive colectomy represent the only methods of secondary prevention for CRC (3,4).

CRCs arise from precancerous lesions known as adenomas where evolution from the normal colorectal epithelium to adenomas and ultimately to invasive cancer can take up to 10 years to develop (5). Although certain hereditary CRC syndromes are characterized by germline mutations [e.g., Adenoma Prevention with Celecoxib (APC)] resulting in the formation of thousands of polyps and a high lifetime risk of CRC [i.e., familial adenomatous polyposis (FAP) syndrome], most CRCs (approximately 75%) are sporadic, arising from well-described early and frequent molecular alterations in key genes implicated in colorectal tumorigenesis (6,7). The progression from normal epithelium to CRC in sporadic cases typically follows the chromosomal instability (CIN) pathway, which is similar to the molecular etiology seen in FAP (8,9). The process often begins with mutations in the APC gene, occurring in approximately 70–80% of cases (10,11). These mutations disrupt the Wnt signaling pathway, promoting cell proliferation and inhibiting differentiation, leading to early adenoma formation (12-14). As the adenoma grows, additional mutations accumulate, frequently including activating mutations in KRAS (40–50% of cases), which further promote cell proliferation and survival (15,16). The transition to carcinoma is marked by the loss of function in tumor suppressor genes like TP53 (50–70% of cases) and SMAD4, allowing cells to evade apoptosis and promote invasive growth. Finally, additional genetic and epigenetic alterations accumulate, leading to metastatic potential (17,18).

The U.S. Preventive Services Task Force (USPSTF) currently recommends screening colonoscopies for standard-risk individuals at age 45 years (3). Colonoscopy is unique among cancer screening tools, as it is both diagnostic and therapeutic since removal of precancerous polyps can be achieved. Although CRC chemoprevention, which is the use of supplements, synthetic or natural as a means of primary prevention for CRC, has undergone active investigation, there exists no approved chemopreventive agent for CRC (19).

In this review, we highlight CRC chemoprevention trials that have investigated chemopreventive agents aiming to prevent CRC development through reduction of CRC itself or through reduction of known precursor lesions such as adenomas and aberrant crypt foci (ACF). We specifically aimed to identify randomized controlled trials (RCTs) that have examined potential chemopreventive agents, including nonsteroidal anti-inflammatory drugs (NSAIDs), minerals, vitamins, and metabolic agents with published results for the prevention of sporadic CRC. We present this article in accordance with the Narrative Review reporting checklist (available at https://tgh.amegroups.com/article/view/10.21037/tgh-24-97/rc).


Methods

Search engines including PubMed, Science Direct, and Google Scholar were used to identify articles (Table 1). For articles discussing RCTs, there was no year specification, and we selected for all other articles discussing CRC chemoprevention we felt to be of relevance to highlight in this review. The inclusion criteria for this review include RCTs with or without a placebo, chemopreventive trials in sporadic CRC, and trials with available results. Exclusion criteria include study populations with a confirmed genetic predisposition to CRC, including Lynch and FAP syndromes. Observational, prospective or retrospective cohort, case-controlled, animal, in vitro studies, and trials without published results were excluded. The purpose of this review is to focus solely on potential chemopreventive agents that have been clinically studied in humans for sporadic CRC prevention.

Table 1

Search strategy summary

Items Specification
Date of search 11/21/2023–3/25/2024
Databases and other sources searched PubMed, Science Direct, Google Scholar
Search terms used Chemoprevention, prevention, colorectal cancer, adenoma, polyp
Timeframe No year specification, all dates searched
Inclusion and exclusion criteria Inclusion criteria: randomized controlled trials with or without a placebo, must have published results in English
Exclusion criteria: study populations with a confirmed genetic predisposition to colorectal cancer, including Lynch and familial adenomatous polyposis syndromes, observational, prospective or retrospective cohort, case-controlled, animal, in vitro studies, and trials without published results were excluded
Selection process First and senior authors conducted the search with verification conducted by both first and senior authors

NSAIDs

Aspirin

Aspirin has been the most studied CRC chemopreventive agent. Classified as an NSAID, aspirin’s chemopreventive potential against CRC appears rooted through its effects on prostaglandin synthesis and catabolism via cyclooxygenase (COX) 1/2 enzymes in colorectal epithelial cells, inhibition of WNT-β-catenin signaling, platelet inactivation, and immunosurveillance (20,21). Early evidence supportive of aspirin’s ability to reduce CRC cases in patients with cardiovascular disease (CVD) led to the USPSTF draft statement in 2015 recommending the use of aspirin for the primary prevention of CRC and CVD (20). Specifically, aspirin was recommended for adults aged 50–59 years (grade B recommendation) or aged 60–69 years (grade C recommendation) with a ≥10% 10-year CVD risk with a life expectancy of ≥10 years willing to take aspirin for ≥10 years without an increased risk of bleeding (20). This recommendation was noticeably withdrawn in an update of the USPSTF 2016 recommendation, following a systematic review of RCTs of aspirin in CRC chemoprevention wherein the USPSTF concluded that there was inadequate evidence to support the daily use of low-dose aspirin in reducing CRC incidence and mortality (22).

In the years preceding and postdating the USPSTF 2016 recommendation, there have been over a dozen RCTs involving aspirin in CRC chemoprevention (Table 2). The British Doctors Aspirin Trial, UK Transient Ischemic Attack Trial (UKTIA), Physician’s Health Study (PHS), Thrombosis Prevention Trial (TPT), Swedish Aspirin Low-dose Trial (SALT), The Women’s Health Study (WHS), and Aspirin in Reducing Events in the Elderly (ASPREE) were randomized trials that looked at the outcome of CRC incidence using at least 75 mg of aspirin daily, except for the WHS which used alternate day dosing of 100 mg aspirin (23-29). Among these early RCTs, the British Doctors Aspirin Trial, TPT, and WHS demonstrated reductions in the incidence of CRC with up to 500 mg of daily aspirin, while the UKTIA, PHS, and SALT studies did not demonstrate significant reductions in CRC incidence with up to 300 mg of daily aspirin (23-28). In a pooled analysis of The British Doctors Aspirin Trial, SALT, UKTIA, and Dutch TIA Aspirin trials, there appeared not to be a reduction of CRC incidence for doses greater than 75 mg daily but the benefit increased with 5 years or longer of aspirin with differential benefit based on tumor subsite location: 70% reduction for proximal colon cancer [hazard ratio (HR) 0.35, 95% confidence interval (CI): 0.20–0.63, P<0.0001], and 42% reduction for rectal cancer (HR 0.58, 95% CI: 0.36–0.92, P=0.02) (27).

Table 2

Aspirin-based randomized controlled trials of colorectal cancer chemoprevention (23-36)

Agent Study, years active Study design (sample size) Eligibility Dose/duration Key findings Ref.
Aspirin British Doctors Aspirin Trial, 1978–1984 RCT (n=5,139) Male physicians 500 mg/day vs. no aspirin, median treatment 6 years Reduction in CRC incidence (HR 0.70) (25)
Aspirin UK Transient Ischemic Attack Trial (UKTIA), 1979–1984 RCT (n=2,449) Recent transient ischemic attack or minor ischemic stroke 300 mg/day, 1,200 mg/day, or placebo, median treatment 4.4 years No significant reduction in CRC incidence (HR 0.82) (25)
Aspirin & beta carotene Physician’s Health Study (PHS), 1982–1988 RCT (n=22,071) Male physicians 325 mg/alternate day vs. placebo, mean follow-up 5 years No significant reduction in CRC risk (HR 1.03) (26,28)
Aspirin Swedish Aspirin Low-dose Trial (SALT), 1984–1990 RCT (n=1,360) Recent transient ischemic attack, minor stroke, or retinal artery occlusion 75 mg/day vs. placebo, median treatment 2.7 years No significant reduction in CRC incidence (OR 0.71) (27)
Aspirin & warfarin Thrombosis Prevention Trial (TPT), 1989–1997 RCT (n=5,085) Men, increased risk of vascular events 75 mg/day vs. placebo & warfarin vs. placebo, median treatment 6.9 years Reduction in CRC incidence (OR 0.61) (27)
Aspirin & vitamin E Women’s Health Study (WHS), 1992–2004 RCT (n=39,876) Women health care professionals 100 mg/alternate day vs. placebo, median 10-year follow-up Reduction in CRC incidence (HR 0.80) (23,24)
Aspirin Aspirin in Reducing Events in the Elderly (ASPREE), 2010–2017 RCT (n=19,114) >70 years of age (≥65 years if Black or Hispanic) 100 mg/day vs. placebo, median follow-up 4.7 years Increased CRC incidence (HR 1.77) (29)
Aspirin Cancer & Leukemia Group B (CALGB), 1993–2000 RCT (n=635) Prior CRC or polyps 325 mg/day vs. placebo, median time to colonoscopy 12. 8 months Reduced adenoma risk (RR 0.65) (36)
Aspirin & folate Aspirin/Folate Polyp Prevention Study (AFPPS), 1994–1998 RCT (n=1,121) Prior removed adenoma 81 mg/day, 325 mg/day or placebo, or folate 1 mg/day vs. placebo (3×2 factorial design), colonoscopy after 3 years (approximately) 81 mg reduced adenoma risk (RR 0.81) and advanced adenoma risk (RR 0.59) (30)
Aspirin Association Pour la Prevention par l’Aspirine du Cancer Colorectal (APACC), 1996–2000 RCT (n= 272) Prior removed adenoma 160 mg/day, 300 mg/day, or placebo, colonoscopy after 1 and 4 years Reduced adenoma risk at 1 year (RR 0.73) but no significant reduction of adenoma risk at 4 years (31,32)
Aspirin & folate United Kingdom Colorectal Adenoma Prevention Trial (UKCAP), 1997–2001 RCT (n=945) Prior removed adenoma 300 mg/day or folate 0.5 mg/day or aspirin with folate or placebo (2×2 factorial design), colonoscopy after 3 years (approximately) Reduced adenoma risk (RR 0.79) and advanced adenoma risk (RR 0.63) (35)
Aspirin Japan Colorectal Aspirin Polyp Prevention (JCAPP), 2007–2009 RCT (n=311) Prior resected CRC or removed adenoma 100 mg/day vs. placebo, colonoscopy after 2 years Reduced CRC and adenoma risk (OR 0.60) (34)
Aspirin & EPA Systematic Evaluation of Aspirin and Fish Oil Polyp Prevention Trial (SeAFOod), 2011–2016 RCT (n=707) Prior removed adenomas 2 g/day EPA, 300 mg/day aspirin, EPA with aspirin, or placebo (2×2 factorial design), colonoscopy after 1 year No significant reduction in adenoma risk with either (RR 0.98 EPA, RR 0.99 aspirin) (33)

RCT, randomized controlled trial; CRC, colorectal cancer; HR, hazard ratio; OR, odds ratio; RR, relative risk; EPA, eicosapentaenoic acid.

The ASPREE trial was a recent RCT where elderly subjects >70 years of age (≥65 years if Black or Hispanic) without CVD, dementia, or disability were randomly assigned to take 100 mg of aspirin or a placebo once a day for a median of 4.7 years (29). There was an increased incidence of CRC with daily aspirin use compared to placebo (HR 1.77, 95% CI: 1.02–3.06) and, surprisingly, a higher incidence in deaths due to gastrointestinal (GI) cancers in the aspirin group (risk of cancer-related death of 6.7 events/1,000 person-years) compared to the placebo group (5.1 events/1,000 person-years, HR 1.31, 95% CI: 1.10–1.56).

The Cancer & Leukemia Group B (CALGB), Aspirin/Folate Polyp Prevention Study (AFPPS), United Kingdom Colorectal Adenoma Prevention Trial (UKCAP), Association Pour la Prevention par l’Aspirine du Cancer Colorectal (APACC), Japan Colorectal Aspirin Polyp Prevention (JCAPP) trial, and Systematic Evaluation of Aspirin and Fish Oil Polyp Prevention Trial (SeAFOod) were all RCTs that looked at polyp occurrence as the outcome (30-36). These studies enrolled subjects with either prior CRC or adenomas that have been resected to receive daily aspirin of 81–325 mg showed a significant reduction in adenoma risk in all but one study, when compared to placebo (Table 2). A meta-analysis including the AFPPS, CALGB, UKCAP, and APACC trials found adenomas in 37% of the participants assigned to the placebo group and 33% assigned to the intervention group, which translated to a statistically significant 17% relative reduction in adenoma risk with aspirin (any dose) vs. placebo (pooled risk ratio of 0.83, 95% CI: 0.72–0.96, absolute risk reduction of 6.7%) (37). However, when looking at the incidence of CRC in the AFPPS, UKCAP and APACC trials (excluding the CALGB trial as some of its participants had been previously diagnosed with CRC), there were no significant difference in CRC rates between the placebo and aspirin (intervention groups, P=0.81) (37).

Although these RCTs presented aspirin as one of the earliest potential chemopreventive agents for sporadic CRC, major bleeding events have been a key side effect (37,38). Meta-analyses of over 54,000 subjects randomized to aspirin and over 52,000 subjects randomized to placebo across 11 RCTs have identified an excess risk of major GI bleeding in those receiving aspirin [relative risk (RR) 1.55, 95% CI: 1.32–1.83], equivalent to approximately 1 per bleed per 1,000 persons per year (38). In a pooled analysis of RCTs of aspirin for adenoma prevention, there was no significant differences in rates of major bleeding across doses of aspirin (2.5%) compared to placebo (2.79%) (37).


Non-aspirin (NA)-NSAIDs

Celecoxib, rofecoxib, sulindac & etodolac

Other NSAIDs besides aspirin include celecoxib, etodolac, ibuprofen, nabumetone, naproxen/naproxen sodium, rofecoxib, sulindac, valdecoxib that are routinely available. We will focus NA-NSAIDs, celecoxib, rofecoxib, sulindac, and etodolac, based on published results from 5 RCTs using these NA-NSAIDs for sporadic CRC chemoprevention (39-43). NA-NSAIDs competitively inhibit COX-1 and COX-2 as their mechanism of action, but due to increased GI bleeding risk potentially associated with COX-1 inhibition, NA-NSAIDs that selectively inhibit COX-2 have been tested in RCTs for adenoma prevention (19).

In the APC Trial, 2,035 participants aged 31–88 years of age who had previously undergone colonoscopic removal of adenomas were randomized to take 200 or 400 mg of celecoxib twice daily, or placebo with repeat colonoscopies after 1 and 3 years of intervention (41). The trial found adenomas in 60.7% in those who were allocated to the placebo group, 43.2% in those who were allocated to the 200 mg celecoxib group, and 37.5% in those assigned to the 400 mg celecoxib group at 3 years, corresponding to a RR of 0.67 (95% CI: 0.59–0.77) for the 200 mg celecoxib group and a RR of 0.55 (95% CI: 0.48–0.64) for the 400 mg celecoxib group (Table 3). In a subsequent celecoxib trial, the Prevention of Colorectal Sporadic Adenomatous Polyps (PreSAP) trial, 1,738 participants who had all polyps removed by colonoscopy within 3 months of enrollment were randomized to 400 mg of celecoxib daily or placebo for 3 years (39). The 3-year estimated cumulative adenoma rate was 33.6% with celecoxib vs. 49.3% with placebo (RR 0.64, 95% CI: 0.56–0.75, P<0.001). The Adenomatous Polyp Prevention on Vioxx (APPROVe) trial studied the effect of another COX-2 inhibitor, rofecoxib, on adenomas diagnosed during 3 years of intervention (40). In APPROVe, 2,587 participants of ≥40 years of age with at least 1 histologically confirmed large-bowel adenoma within 12 weeks of study entry were randomized to 25 mg of rofecoxib daily or placebo with colonoscopy at 1 and 3 years. Rofecoxib conferred a significant reduction at 1 year (RR 0.65, 95% CI: 0.57–0.73) than the subsequent 2 years (RR 0.81, 95% CI: 0.71–0.93) (40).

Table 3

Non-aspirin, NSAID-based randomized controlled trials of colorectal cancer chemoprevention (39-43)

Agent Study, years active Study design (sample size) Eligibility Dose/duration Key findings Ref.
DFMO & sulindac Randomized Placebo-Controlled, Double-Blind Trial of DFMO plus Sulindac for Prevention of Sporadic Colorectal Adenomas, 1998–2008 RCT (n=305) Prior adenoma removed 150 mg/day sulindac and 500 mg/day DFMO vs. placebo for 36 months Reduced adenoma risk (risk ratio 0.30) and advanced adenoma risk (risk ratio 0.085) (42)
Celecoxib Adenoma Prevention with Celecoxib (APC), 1999–2002 RCT (n=2,035) Prior adenoma removed 200 mg BID, 400 mg/BID, or placebo, colonoscopy after 1 and 3 years Reduced adenoma risk at 3 years (RR 0.67 low dose, RR 0.55 high dose) but increased cardiovascular risk (41)
Rofecoxib Adenomatous Polyp Prevention on Vioxx (APPROVe), 2000–2004 RCT (n=2,586) Prior adenoma removed 25 mg/day vs. placebo, colonoscopy after 1 and 3 years Reduced adenoma risk (RR 0.65 at 1 year, RR 0.81 at 3 years) but with increased significant upper GI events (i.e., perforations, ulcers, obstructions, and bleeds) and thrombotic cardiovascular events (40)
Celecoxib Prevention of Colorectal Sporadic Adenomatous Polyps (PreSAP) Trial, 2001–2005 RCT (n=1,738) Prior adenoma removed 400 mg/day vs. placebo, colonoscopy after 1 and 3 years Reduced adenoma risk (RR 0.64) and advanced adenoma risk at 3 years (RR 0.49) (39)
Sulindac or etodolac Randomized double-blind trial of sulindac and etodolac to eradicate ACF and sporadic colorectal polyps, 2002–2007 RCT (n=189) Prior ACF or prior adenoma removed 150 mg BID sulindac, 200 mg BID etodolac, or placebo for 2 months Reduction of ACF number (P=0.0075) and total polyp risk (risk ratio 0.44) with sulindac but not etodolac (43)

NSAID, nonsteroidal anti-inflammatory drug; DFMO, difluoromethylornithine or eflornithine; RCT, randomized controlled trial; BID, twice daily; RR, relative risk; GI, gastrointestinal; ACF, aberrant crypt foci.

Notably, despite the promise of these COX-2 inhibitors in adenoma prevention, rofecoxib was associated with increased thrombotic cardiovascular events (RR 1.89, 95% CI: 1.18–3.04) and upper GI perforation, obstruction, symptomatic ulcers, or bleeding (RR 4.91, 95% CI: 1.98–14.5), relative to placebo (40), resulting in the halting of the APPROVe trial and the voluntary withdrawal of new drug applications (NDAs) for rofecoxib submitted to the U.S. Food and Drug Administration (FDA). Although the PreSAP trial did not identify a significant increase in cardiovascular risk with celecoxib, the concurrent APC Trial showed a significantly increased risk of serious cardiovascular events (i.e., death from cardiovascular events, nonfatal myocardial infarction, stroke, or heart failure) with both low-dose (risk ratio 2.6, 95% CI: 1.1–6.1) and high-dose celecoxib (risk ratio 3.4, 95% CI: 1.5–7.9), when compared to placebo, leading to suspension of celecoxib administration in both the PreSAP and APC trials on December 17, 2004 (39,41).

In a recent RCT, sulindac (COX-1/2 inhibitor) and etodolac (more selective for COX-2 than COX-1 inhibition) were investigated on the prevention of ACF and adenomas after 2 months (43). One hundred and eighty-nine subjects who had prior history of ACF or polyps removed were randomized to 300 mg of sulindac once a day, 400 mg of etodolac once a day, or placebo (Table 3). The frequency of ACF at 2 months was significantly reduced in the sulindac group relative to the placebo group (P=0.0075), but the ACF number in the etodolac group was not significantly different relative to placebo (P=0.67). There were no cardiovascular events observed and no significant difference in adverse events across groups, the majority of which were grade 1 in severity (43). Polyamine inhibition has been shown to prevent colon carcinogenesis in mouse models as well in human clinical trials (44). In another RCT, 305 subjects with a prior adenoma resected within 5 years of study entry were randomized to placebo or DFMO (500 mg daily)/sulindac (150 mg daily). The safety and chemopreventive effects of sulindac (which, in addition to COX-1/2 inhibitory effects, also exports polyamines from cells via a PPAR-gamma mechanism) were highlighted in combination with difluoromethylornithine or eflornithine (DFMO), an irreversible inhibitor of ornithine decarboxylase (ODC) (42). At 3-year colonoscopy, there was a significant reduction in colorectal adenomas with the combination compared to placebo (RR 0.30, 95% CI: 0.18–0.49, P<0.001) with a more marked reduction with advanced adenomas (RR 0.085, 95% CI: 0.011–0.65, P<0.001). Of note, there was a non-significant but numeric increase in serious cardiovascular events in the combination (8.4%) vs. placebo (4.9%) groups, which in subset analysis appeared to be limited to those in the trial having elevated CVD risk at baseline (42,45). There were no significant differences in grade ≥3 adverse events or hearing changes from baseline (side effect unique to DFMO) between groups (42).


Metabolic agents

Agents commonly used to treat diabetes and hyperlipidemia, henceforth referred to as metabolic agents, have also been investigated as potential chemopreventive strategies for sporadic CRC (Table 4). These RCTs often occurred in patients with obesity or preexisting risk factors for CVD with the intent to decrease risks and complications from metabolic syndrome (46-57).

Table 4

Metabolic agent-based randomized controlled trials of colorectal cancer chemoprevention (46-57)

Agent Study, years active Study design (sample size) Eligibility Dose/duration Key findings Ref.
Pravastatin West of Scotland Coronary Prevention Study Group, 1989–1995 RCT (n=6,595) No prior MI history 40 mg/day vs. placebo, mean follow-up 13.2 years for incident cancers No significant reduction in CRC incidence (HR 0.82) (50)
Pravastatin Cholesterol and Recurrent Events (CARE) trial, 1989–1996 RCT (n=4,159) Prior MI 40 mg/day vs. placebo, median follow-up 5 years No significant reduction in CRC incidence (RR 0.57, 95% CI: 0.28–1.16) (48,55)
Simvastatin Scandinavian Simvastatin Survival Study (4S), 1988–1994 RCT (n=4,444) Prior MI or angina pectoris 20 mg/day vs. placebo, 10.4-year follow-up No significant reduction in CRC incidence (RR 0.78, 95% CI: 0.46–1.32) and all cancer risk (RR 0.88) (48,56)
Lovastatin Air Force/Texas Coronary Atherosclerosis Prevention Study (AFCAPS/TEXCAPS), 1990–1997 RCT (n=6,605) Average total cholesterol and LDL-C levels with below-average HDL-C levels 20-40 mg/day vs. placebo, mean follow-up 5.2 years Nonsignificant increase in CRC incidence (RR 1.25, 95% CI: 0.70–2.24) (48,49)
Pravastatin Long-term Intervention with Pravastatin in Ischemic Disease (LIPID), 1990–1997 RCT (n=9,014) Prior MI or unstable angina pectoris 40 mg/day vs. placebo for 6 years No significant reduction in CRC incidence (RR 0.89) (54)
Pravastatin GISSI Prevenzione (GISSI-P) trial, 1993–1996 RCT (n=4,271) Prior MI 20 mg/day vs. no treatment, mean follow-up 2 years No significant change in cancer incidence at any particular site (46)
Simvastatin Heart Protection Study (HPS), 1994–2001 RCT (n=20,536) Substantial 5-year risk of death from CHD 40 mg/day vs. placebo, mean follow-up 5 years No significant reduction in CRC incidence (RR 0.87, 95% CI: 0.68–1.12) (48,51)
Pravastatin Antihypertensive and Lipid-Lowering Treatment to Prevent Heart Attack Trial (ALLHAT-LLT), 1994–2002 RCT (n=10,355) Stage 1–2 hypertension with ≥1 additional CHD risk factor 40 mg/day vs. usual care, mean follow-up 4.8 years Nonsignificant increased CRC risk (RR 1.21, 95% CI: 0.79–1.86) (47,48)
Metformin Hosono K et al., 2008–2010 RCT (n=26) Nondiabetic with prior ACF 250 mg/day vs. control for 1 month Decreased mean number of ACF (53)
Metformin Zell J et al., 2011–2013 RCT (n=32) Obesity with prior adenoma removed 500 mg/day escalated up to 2,000 mg/day for 12 weeks No significant difference in tissue S6Ser235 (P=0.77) and Ki-67 (P=0.43) (57)
Metformin Higurashi T et al., 2011–2014 RCT (n=151) Nondiabetic with prior adenoma removed 250 mg/day vs. placebo, colonoscopy after 1 year Reduction in adenoma risk (RR 0.60) (52)

RCT, randomized controlled trial; MI, myocardial infarction; CRC, colorectal cancer; HR, hazard ratio; RR, relative risk; CI, confidence interval; LDL-C, low-density lipoprotein cholesterol; HDL-C, high-density lipoprotein cholesterol; CHD, coronary heart disease; ACF, aberrant crypt foci.

Statins

Lipid-lowering agents of the statin class are competitive hydroxymethylglutaryl-CoA reductase (HMG-CoA) inhibitors and have been shown to inhibit the growth and development of colorectal tumors in preclinical models of chemically or genetically-induced colorectal tumorigenesis (19,58).

The West of Scotland Coronary Prevention Study was an RCT comparing pravastatin (40 mg oral daily) with placebo in 6,595 men with hypercholesterolemia who did not have a history of myocardial infarction, with a mean follow-up of 13.2 years for incident cancers (50). Of 122 incident CRC detected during the total follow-up period, there was no significant reduction in CRC incidence between pravastatin (56/3,291 or 1.7%) and placebo (66/3,286 or 2.0%) groups (HR 0.82, 95% CI: 0.58–1.17, P=0.28). The CARE trial randomized 4,159 men and postmenopausal women with prior myocardial infarction to 40 mg oral daily of pravastatin or placebo and similarly found no significant reduction in CRC incidence (RR 0.57, 95% CI: 0.28–1.16) with a median follow-up of 5 years (48,55).

In the Scandinavian simvastatin survival study, 4,444 patients who have been previously diagnosed with a myocardial infarction or angina were randomized to receive 20 mg of simvastatin once a day or placebo with a median follow-up of 10.4 years (48,56). Out of 57/4,444 incident CRCs during this follow-up period, there was no significant reduction in CRC incidence (RR 0.78, 95% CI: 0.46–1.32).

The AFCAPS/TEXCAPS study randomized 6,605 men and women with average cholesterol risk to 20–40 mg of daily lovastatin or placebo and identified a nonsignificant increase in CRC incidence (RR 1.25, 95% CI: 0.70–2.24) with a mean follow-up of 5.2 years (48,49). A nonsignificant increased risk of CRC was also observed in the ALLHAT-LLT trial that randomized 10,355 patients with ≥1 CVD risk factor and stage 1–2 hypertension to pravastatin 40 mg daily or placebo (RR 1.21, 95% CI: 0.79–1.86) (47,48).

Several subsequent pravastatin trials such as the LIPID and GISSI Prevenzione trials randomized patients with prior myocardial infarction or unstable angina pectoris to pravastatin 20–40 mg oral daily or control have similarly shown no significant reduction in CRC incidence or incidence of other cancer sites (Table 4) (46,54).

Metformin

Metformin is a biguanide used to treat individuals with type 2 diabetes and having been shown to elicit potential antitumor properties and reduce ACF and polyp formation in genetic mouse models of FAP and azoxymethane-treated animals, a mouse model of chemically-induced CRC (19,58). To date, 2 RCTs and 1 single-arm phase IIa trial have been published on metformin’s chemopreventive potential in sporadic CRC (Table 4).

Zell et al. conducted a phase IIa trial of 32 subjects with obesity and a prior history of adenomas to receive metformin beginning at 500 mg oral daily escalated up to 2,000 mg daily for 12 weeks and did not identify a significant difference in rectal tissue pS6Ser235/236 or Ki67 immunostaining levels as surrogate biomarkers of metformin’s chemopreventive potential in sporadic CRC (57). Of note, in contrast to RCTs, biomarker testing in this trial was done on the target tissue of origin (normal rectal mucosa) rather than in adenomas.

In an RCT by Higurashi et al., nondiabetic participants with a prior history of resected adenomas were assigned to one of two groups: 250 mg of metformin (n=79) once a day or matching placebo (n=72) once a day with a colonoscopy at 1 year (52). The trial found that the metformin group showed a decrease in the number of polyps compared to the placebo group [total polyps 27/71 subjects with metformin (38.0%, 95% CI: 26.7–49.3%) vs. 35/62 with placebo (56.5%, 95% CI: 44.1–68.8%)] for a RR of 0.67 (95% CI: 0.47–0.97, P=0.034). The rate of adenomas with metformin was 22/71 (30.6%, 95% CI: 19.9–41.2%) vs. 32/62 with placebo (51.6%, 95% CI: 39.2–64.1%) for a RR of 0.60, 95% CI: 0.39–0.92, P=0.016).

Hosono et al. conducted a randomized study to investigate the use of metformin on ACF prevention (53). Twenty-six nondiabetic patients with prior history of ACF were randomized to 250 mg of metformin (n=12) once a day or matching placebo (n=14) for 1 month. The metformin group had a significantly lower mean number of ACF at 1 month (8.78±6.45 pretreatment vs. 5.11±4.99 at 1 month, P=0.007) than the control group (7.23±6.65 baseline vs. 7.56±6.75 at 1 month, P=0.609).


Minerals and vitamins

There has been interest in exploring the potential of minerals or vitamins including folate, calcium, and vitamin D to prevent sporadic CRC with results published from multiple RCTs (Table 5).

Table 5

Vitamin and mineral-based randomized controlled trials of colorectal cancer chemoprevention (59-68)

Agent Study, years active Study design (sample size) Eligibility Dose/duration Key findings Ref.
Aspirin & folate Aspirin/Folate Polyp Prevention Study (AFPPS), 1994–2004 RCT (n=1,021) Prior adenoma 1 mg/day folic acid vs. placebo, separately randomized to receive aspirin 81 or 325 mg/day or placebo, colonoscopy after 3 and 6 or 8 years No significant reduction in adenoma risk at first (RR 1.04) and second (RR 1.13) follow-up (59,60)
Folate Jaszewski R et al., 1998–2005 RCT (n=94) Prior adenoma removed 5 mg/day vs. placebo for 3 years Reduction in adenoma risk (OR 2.77) (61)
Folate, B6, and B12 Women’s Antioxidant and Folic Acid Cardiovascular Study (WAFACS), 1998–2005 RCT (n=5,442) Women health professional with preexisting cardiovascular disease or ≥3 coronary risk factors 2.5 mg/day folic acid, 50 mg/day vitamin B6, and 1 mg/day vitamin B12 vs. placebo for up to 7.3 years No significant reduction in CRC incidence (HR 0.81) (62)
Folate Vitamins to Prevent Stroke Trial (VITATOPS), 1998–2008 RCT (n=8,164) Precent stroke or transient ischemic attack 2 mg folic acid, 25 mg vitamin B6, 500 μg vitamin B12 vs. placebo, median duration 3.4 years No significant reduction in CRC incidence (HR 0.98) (63)
Folate Gao Q et al., 2008–2012 RCT (n=800) Age >50 years with no prior adenoma on colonoscopy 1 mg/day vs. no intervention for 3 years Reduction in adenoma risk (RR 0.49) but no significant reduction in CRC incidence (RR 1.00) (64)
Calcium Calcium Polyp Prevention Study, 1988–1996 RCT (n=930) Prior adenoma removed 3 g/day of calcium carbonate vs. placebo, colonoscopy after 1 and 3 years Reduction in adenoma risk (RR 0.81) in those who completed both endoscopies (65)
Calcium or fiber European Cancer Prevention Organization (ECP) Intervention Study, 1991–1994 RCT (n=665) Prior adenoma removed 2 g/day calcium, 3–5 g/day fiber or placebo, colonoscopy after 3 years Nonsignificant reduction in adenoma risk with calcium (OR 0.66, 95% CI: 0.38–1.17) but increased adenoma risk with fiber (OR 1.67, 95% CI: 1.01–2.76) (66)
Calcium & vitamin D Women’s Health Initiative (WHI), 1993–2005 RCT (n=36,282) Postmenopausal women aged 50–79 years 500 mg calcium and 200 IU of vitamin D3 BID vs. placebo, mean treatment 7 years No significant reduction in CRC incidence (HR 1.08) (67)
Vitamin D & omega 3 Vitamin D and OmegA-3 Trial (VITAL), 2011–2017 RCT (n=25,871) No cancer or CVD at baseline 2,000 IU/day vitamin D3, 1 g/day omega-3 fatty acids, or placebo (2×2 factorial design), mean treatment 5 years No significant reduction in CRC incidence (HR 1.09) for vitamin D (68)

RCT, randomized controlled trial; RR, relative risk; OR, odds ratio; CRC, colorectal cancer; HR, hazards ratio; CI, confidence interval; IU, international units; BID, twice daily; CVD, cardiovascular disease.

Folate

Folate or folic acid is a B vitamin (vitamin B9) that is critical to DNA synthesis and cell division, and its chemopreventive effects in CRC have been purported to involve its regulation of normal DNA synthesis and repair (19,58). There have been 5 RCTs investigating folate in sporadic CRC chemoprevention (Table 5).

The AFPPS was a phase III RCT that sought to investigate the impact of folic acid supplementation and aspirin in the development of colorectal adenomas (59,60). Of 1,021 participants with a history of a previous adenoma, 516 participants were randomized to 1 mg of folic acid oral daily and the remainder to placebo. They were then randomized again and assigned to take 81 or 325 mg of aspirin a day. The trial ultimately did not show any decrease in colorectal adenomas with folic acid (44.1% adenomas) relative to placebo (42.4% adenomas, unadjusted RR 1.04, 95% CI: 0.90–1.20, P=0.58). Following a second colonoscopy, the risk of ≥1 adenoma was 41.9% with folic acid vs. 37.2% with placebo (unadjusted RR 1.13, 95% CI: 0.93–1.37, P=0.23).

Jaszewski et al. conducted an RCT where 49 participants were randomized to 5 mg of folic acid daily and 45 participants were assigned to the placebo group for 3 years (61). A significantly reduced number of adenomas was detected in the folic acid group vs. placebo [64% lower risk ratio, odds ratio (OR) of 2.77, P=0.02514].

The Women’s Antioxidant and Folic Acid Cardiovascular Study (WAFACS) studied the use of folic acid, vitamin B6, and vitamin B12 in 5,442 women health professionals diagnosed with pre-existing CVD or ≥3 CVD risk factors (62). Participants were randomized to 2.5 mg of folic acid, 50 mg of vitamin B6, and 1 mg of vitamin B12 or a placebo once daily for up to 7.3 years. The study found no difference in the development of CRC between the intervention and placebo groups (HR 0.81, 95% CI: 0.43–1.50, P=0.50) and no significant difference in any cancer death across groups (HR 0.82, 95% CI: 0.56–1.21, P=0.32).

The Vitamins to Prevent Stroke Trial (VITATOPS) randomized 8,164 patients who had experienced a stroke or transient ischemic attack to the combination of 2 mg folic acid, 25 mg vitamin B6, and 500 µg vitamin B12 or placebo once a day for a median of 3.4 years. Hankey et al. did not find a difference in the development of any cancers between the intervention and placebo groups (4.04% B vitamins group vs. 4.59% placebo group, RR 0.86, 95% CI: 0.70–1.07) and there was no significant reduction in CRC incidence (HR 0.98) (63).

In a clinical trial by Gao et al., participants >50 years with no prior adenoma on colonoscopy were randomized to 1 mg of folic acid daily or no intervention for 3 years (64). The trial found colorectal adenomas in 14.88% of patients in the folic acid group vs. 30.70% in the control group (unadjusted RR 0.49, 95% CI: 0.37–0.63, P<0.01). This potential chemopreventive effect was seen across left-sided adenomas (unadjusted RR 0.54, 95% CI: 0.38–0.76, P=0.001) and advanced adenomas (unadjusted RR 0.36, 95% CI: 0.16–0.81, P=0.01), but there was no significance difference between the groups for ≥3 more adenomas, right-sided adenoma, or CRC incidence.

Calcium and vitamin D

Calcium is the most abundant mineral in the body, while vitamin D is essential for calcium homeostasis; both have been implicated in colorectal adenoma and carcinoma formation through their regulation of growth factor signaling, APC/β-catenin pathway signaling, and bile acid-binding that intersect on proliferation and angiogenesis of colonocytes (19,58,69-71). Unsurprisingly, there have been investigations at the RCT level on the ability of vitamin D and calcium to prevent sporadic CRC (Table 5).

Of the earliest, the Calcium Polyp Prevention Study enrolled 930 patients with a previous diagnosis of adenoma and randomized them to 3 g of calcium carbonate (n=409) oral daily or placebo (n=423) with colonoscopies at 1 and 3 years (65). The intervention group showed a decrease in the number of adenomas found during colonoscopy compared to the placebo group (mean number per patient 0.55 vs. 0.73 with placebo, P=0.03) with an unadjusted RR for having ≥1 adenoma in the calcium group of 0.83 (95% CI: 0.68–1.00, P=0.05) vs. placebo (adjusted RR 0.81, 95% CI: 0.67–0.99, P=0.04). There was a significant reduction in adenoma risk (RR 0.81) in patients that completed both colonoscopies with calcium over placebo.

The European Cancer Prevention Organization (ECP) Intervention Study randomized 665 patients who previously had been diagnosed with colorectal adenomas to 2 g of elemental calcium, 3–5 g of fiber (ispaghula husk), or placebo daily with a colonoscopy at 3 years (66). The trial found a decrease in adenomas in the calcium group, but the fiber group showed an increase in adenoma risk, when compared to placebo (Table 5). When adjusted for recurrence, the odds ratio was 0.66 (95% CI: 0.38–1.17, P=0.16) in the calcium group vs. 1.67 (95% CI: 1.01–2.76, P=0.042) in the fiber group. Notably, the significantly higher odds ratio in the fiber group was associated with a higher baseline dietary calcium intake than those with intake below the median (interaction test, P=0.028).

In a Women’s Health Initiative (WHI) trial involving postmenopausal women aged 50–79 years, 18,176 patients were randomly assigned to 500 mg of calcium carbonate combined with 200 international units (IU) of vitamin D3 twice daily and 18,106 patients were assigned to a matching placebo twice daily for a mean treatment duration of 7 years (67). The trial found no significant difference in CRC incidence between the intervention and placebo groups (HR 1.08, 95% CI: 0.86–1.34, P=0.51).

In the recently conducted Vitamin D and OmegA-3 TriaL (VITAL), 25,871 patients with no cancer or CVD history at baseline were randomized to receive vitamin D3 2,000 IU daily and omega-3 fatty acids at 1 g daily or placebo for a mean treatment duration of 5 years (68). Of 98 CRC diagnosed, there was no significant reduction in CRC incidence (HR 1.09, 95% CI: 0.73–1.62) with the combination compared to placebo.


Discussion

The complexities of designing chemoprevention clinical trials in CRC have long been well known and have been described extensively elsewhere (19,58). The basic tenets of an ideal chemopreventive agent in CRC have historically included the need for the drug to demonstrate efficacy, safety, tolerability, low cost, widespread availability, and ease of administration (19,58). Over the past 4 decades, numerous RCTs have been performed with hopes of establishing the first chemopreventive drug for sporadic CRC, a disease by in which colonoscopy remains the only consensus guidelines recommended method for prevention (secondary). Where several classes of drugs have failed (48,72), others have shown promise (73-75). Promising agents have shown potential in reducing adenomas but face challenges in balancing efficacy with toxicity. Understanding the latent effects and determining the optimal dosage and targeted population are necessary to maximize benefits and minimize risks. We end this review with a discussion of key concepts that we can learn from previously conducted RCTs and build upon for the next steps in the successful clinical development of chemopreventive strategies for sporadic CRC.

Study population

The choice of patient population to investigate the chemopreventive effects of a drug remains fundamental to the design of a clinical trial and, ultimately, impactful to the population in which the findings can be translated. At one end of the spectrum lies the highest risk patients for colorectal adenomas and CRC. FAP syndrome, for example, is characterized by a lifetime risk of CRC of up to 100% but only about 1% of all CRC cases arise in FAP patients with a prevalence of around 1 in 10,000 live births (76). Inclusion of the highest risk populations would enrich for events of interest in a CRC chemoprevention trial, i.e., adenoma or CRC formation, which could reduce planned sample size (improving feasibility). However, rarer patient subsets may require more patients or longer study durations to meaningfully accrue. Primary prevention clinical trials aiming to prevent the initial occurrence of CRC or precancerous lesions in average risk individuals would, theoretically, yield the highest quality results with practice-changing implications, however such trials are infeasible given the extraordinary sample sizes needed to detect small effects and the duration of follow-up required (decades) (77,78). As sporadic CRC represents the overwhelming majority of CRCs diagnosed globally, striking a balance between the high-risk and low-risk patients for sporadic CRC would be important to account for patient characteristics that could impact how long a study takes, the sample size requirements, and funding needed to support a chemoprevention trial.

Of the positive RCTs published and in recent meta-analyses, it is noteworthy to point out that the majority of these trials were conducted in study populations with a previous history of precursor lesions to CRC, such as ACF or adenoma. These trials focus on secondary prevention, aiming to prevent the recurrence of these lesions. Patients with established lesions are ideal as they are at higher risk, allowing researchers to assess the effectiveness of chemopreventive agents more efficiently than in primary prevention trials, which require larger populations and longer follow-up periods. Additionally, some trials have focused on tertiary prevention, targeting patients with a history of resected CRC. In this group, the goal is to prevent recurrence or progression to metastatic disease, both of which pose significant risks. However, in such patients, the high baseline risk of recurrence and metastasis can complicate efforts to demonstrate the benefits of chemoprevention, as these risks may outweigh the preventive effects on new adenomas or CRC (Tables 2-5).

Overall, the distinction between secondary/tertiary prevention and primary prevention is significant because patients who have already had adenomas/CRC removed are inherently at a higher risk of developing further adenomas or CRC compared to individuals who have never developed adenomas or CRC. Patients with a history of adenomas have a 30–60% recurrence rate within 3–5 years (41,78-80), while those with a history of resected CRC have a 1.5–3% risk of developing metachronous CRC within 5 years after resection (81). Due to the differences in baseline risk, secondary/tertiary prevention trials are often more feasible with shorter follow-up periods and may show effects more quickly. Despite the challenges, they are crucial for advancing chemoprevention strategies.

Another high-risk group to consider targeting includes first-degree relatives of CRC patients especially those diagnosed with early-onset CRC (before age 50 years). First-degree relatives of CRC patients have approximately double the risk of developing CRC compared to the general population. The risk is even higher for first-degree relatives of patients diagnosed with early-onset CRC (82,83). Our search results did not find specific chemopreventive trials targeting this population. This gap in the literature suggests an opportunity for future research to focus on chemoprevention strategies.

Choice of endpoint

Beyond the need to select an ideal study population, the choice of primary endpoint is an important consideration for assessment of chemopreventive effects in sporadic CRC. Building on the promise of NSAIDs such as aspirin, NA-NSAIDs showed chemopreventive potential in sporadic CRC in randomized phase III trials (Table 3). Much can be learned from the developmental pathway for celecoxib, in particular, and the FDA’s initial approval of celecoxib for CRC chemoprevention. The celecoxib experience set a regulatory precedent for using surrogate endpoints like polyp reduction but underscored the importance of long-term follow-up and managing the risk of adverse events, especially in high-risk populations.

Polyp reduction has been recognized by the FDA as a valid endpoint when celecoxib became the first drug treatment approved for FAP in 1999 due to a mean reduction in polyps in subjects treated for 6 months compared to placebo (84). Of note, the FDA label was withdrawn in 2012 due to lack of completion of the post-marketing celecoxib study and subsequently when 2 phase III clinical trials showed that although celecoxib significantly reduced the occurrence of sporadic colorectal adenomas, this agent could not be routinely recommended for chemoprevention due to risk of cardiovascular events (39,41). Nevertheless, this approval sets an important precedent and milestone for CRC chemoprevention trials. Accelerated approval was based on a study of 83 patients with FAP syndrome, of whom 25 had an intact colon (84). A mean reduction in the number of polyps was 28% for 400 mg celecoxib twice daily vs. 12% for 100 mg twice daily vs. 5% for placebo from baseline to after 6 months of intervention.

Aside from endpoints using rates of adenoma formation or frank CRC, several CRC chemoprevention trials have employed biomarker endpoints. Unfortunately, multiple clinical trials investigating biomarker endpoints as surrogate markers of a drug’s chemopreventive effects for sporadic CRC have been negative to date (57,69-71). Notably, these trials have measured the experimental treatment’s effects on biomarkers in normal colorectal mucosa. Whether chemopreventive effects are better evaluated via biomarkers assessed in neoplastic colorectal tissues (e.g., ACF or adenomas) remains an important unanswered question. Surrogate biomarker endpoints could circumvent the need for substantially larger sample sizes and longer study durations that can prove prohibitive, while serving as a marker for the agent’s potential to inhibit a key molecular pathway in colorectal tumorigenesis. If positive, these studies would then provide support for larger, prospective RCTs where the potential for CRC chemoprevention could be validated.

Chemopreventive latency and dosage

Whether the chemopreventive effects of a drug is dependent on its dosage remains an active area of investigation in the primary prevention of CRC. Early signals that an increasing dose may have more chemopreventive potential was demonstrated in the APC Trial when celecoxib at 200 mg twice daily showed a RR for adenomas of 0.67, while a higher dose of 400 mg twice daily showed reduced adenoma risk at 3-year colonoscopy (RR 0.55), relative to placebo (41). However, higher doses of celecoxib were significantly associated with increased cardiovascular risk, which were essentially prohibitive to continued use of high-dose celecoxib (80). The body of evidence for aspirin in sporadic CRC chemoprevention has also afforded some insight into the dosage question. Specifically, meta-analyses have identified that low-dose aspirin (OR 0.71, 95% CI: 0.41–1.24) to be effective in preventing advanced colorectal neoplastic lesions, comparable to high-dose aspirin (OR 0.81, 95% CI: 0.50–1.28) while having a more favorable safety profile than high-dose aspirin (73). Therefore, although studies have pointed to potential dose-dependent chemopreventive effects for certain drugs, the applicability of higher doses are often limited by otherwise prohibitive toxicities.

The concept of latency for chemopreventive effects in sporadic CRC has been studied based on follow-up data of more than 20 years from 2 large RCTs of aspirin vs. no aspirin in CRC chemoprevention. The British Doctors Aspirin Trial and UKTIA trials treated patients with aspirin for 5–6 years or 1–7 years, respectively, and firstly demonstrated that patients allocated to aspirin for at least 5 years had a lower incidence of CRC (HR 0.63, 95% CI: 0.47–0.85, P=0.002) than all patients enrolled (HR 0.74, 95% CI: 0.56–0.97, P=0.02), relative to placebo (25). However, when stratified by 10-year follow-up periods, a lower CRC incidence was observed in years 10–19 of follow-up (pooled HR 0.60, 95% CI: 0.42–0.87, P=0.007) than years 0–9 of follow-up (pooled HR 0.92, 95% CI: 0.56–1.49, P=0.73). It has been postulated that ≥300 mg of daily aspirin for 5 years can be effective in CRC chemoprevention, with a 10-year latency of effect that is consistent with observational studies and knowledge of the adenoma-carcinoma sequence (25). This understanding is informative in the design and follow-up considerations for sporadic CRC chemoprevention trials given that the best predicted effect of longer-term aspirin, for example, is estimated to occur 10–14 years after randomization with at least 5 years of continuous dosing of aspirin (25). Even if latent effects of a chemopreventive agent is unknown or nonexistent, the longer follow-up of the APC Trial has provided insight on what happens after withdrawal of an effective chemopreventive agent for sporadic CRC (80). At the time of the year 5 colonoscopy, >90% of participants were off study for 1 year or more (median duration of celecoxib treatment 3.1 years) and the APC Trial reassuringly showed similar rates of adenoma detection across treatment groups and placebo. This addresses a well-known concern that a chemopreventive drug may be active only by inhibition of low-grade lesions rather than preventing disease progression, allowing lesions with higher malignant potential to progress upon withdrawal of the drug (80). One example of this phenomenon would manifest in a drug-associated reduction in adenoma size but not severity that limits colonoscopic detection and removal of more advanced lesions (80).

Toxicity

Safety remains paramount in assessments of all chemopreventive strategies for CRC. Although the NA-NSAIDs, particularly celecoxib and rofecoxib, have piqued interest in sporadic CRC chemoprevention given their positive phase III trials building, further evaluation has uncovered safety concerns limiting their use. Notably, rofecoxib was associated with increased thrombotic cardiovascular events (RR 1.89, 95% CI: 1.18–3.04) and upper GI perforation, obstruction, symptomatic ulcers, or bleeding (RR 4.91, 95% CI: 1.98–14.5), relative to placebo (40), resulting in stoppage of the APPROVe trial and the voluntary withdrawal of NDAs submitted to the FDA. Cardiovascular risk with celecoxib was higher in aspirin users and those with CVD risk factors at baseline (e.g., history of atherosclerotic heart disease, age >65 years, smoking, hypertension, or hyperlipidemia), while the rates were lower in those with no baseline risk factors (80).

Indeed, risk stratification by CVD risk may be warranted in future clinical trials for the NSAID class of drugs being evaluated for sporadic CRC chemoprevention (45). Recent meta-analyses of all RCTs of chemopreventive agents for sporadic CRC have corroborated findings that after balancing benefits and harms, routine use of NA-NSAIDs, although most effective for the prevention of colorectal adenomas, cannot be recommended based on current evidence (73,75). Low-dose aspirin, however, appears to have the most favorable risk/benefit profile of all classes of chemopreventive drugs for sporadic CRC (73). The margin of benefit for aspirin use in CRC chemoprevention has not been enough to impact the recommendations of consensus guidelines, however. Ultimately, precedents such as these have reinforced the importance of drug tolerability and the need to weigh the safety profile of an agent against its chemopreventive benefits, as the implementation of chemopreventive agents as a standards-of-care for sporadic CRC chemoprevention have been limited when safety concerns outweigh the benefits.

Future directions

Personalized chemopreventive strategies spanning from vaccines to targeted therapies and biomarkers of chemopreventive effects such as PIK3CA mutations are in active investigation and have bene reviewed elsewhere (19,58). Notably, the NSAID sulindac has shown efficacy in prevention of sporadic colorectal adenomas with a favorable cardiovascular risk profile (42,43). Additionally, there is evidence to support that individuals carrying 2 copies of the G allele of the ornithine decarboxylase-1 (ODC1) gene may have reduced risk of adenoma recurrence following treatment with eflornithine and sulindac providing rationale of a potential population of patients that could be molecularly selected for sporadic CRC prevention (85). The promise of eflornithine and sulindac is currently being investigated in an ongoing randomized, double-blind, placebo-controlled phase III clinical trial (SWOG S0820/PACES) in reducing the risk of adenomas or second primary CRC in survivors of stage 0–III CRC (86). The SWOG S0820/PACES trial is randomized double-blind placebo-controlled trial of combination eflornithine 500 mg daily and sulindac 150 mg oral daily vs. combination matching placebos. Participant eligibility includes those 18 and older who have been diagnosed with stage 0–3 colon or rectosigmoid adenocarcinoma. The trial completed accrual of 354 patients in June 2023, and passed its single planned analysis (for futility) in May of 2023. As such, the intervention continues for enrolled patients and results will be available after June 2026. The primary hypotheses of this study will be that the combination of eflornithine and sulindac will produce a 60% reduction in the rate of high-risk adenomas and second primary CRC vs. placebos, among stage 0–III colon cancer patients (86). The results of the SWOG S0820/PACES trial along with active trials of novel chemopreventive agents are eagerly anticipated as the search for the first established chemopreventive agent in sporadic CRC continues.


Conclusions

Sporadic CRC constitutes a large public health burden. Despite knowledge of the adenoma-carcinoma sequence, there has yet to be a defined method of primary prevention for CRC. Colonoscopy remains the only recommended and standard approach for CRC prevention (secondary). Multiple classes of agents including NSAIDs, metabolic agents, vitamins, and minerals have been explored for their chemopreventive potential in sporadic CRC. Although NSAIDs and NA-NSAIDs have demonstrated positive results for sporadic CRC chemoprevention in RCTs, they have been limited by cardiovascular risk or a lacking of uniform and conclusive results supporting their efficacy. Future trials in this arena are investigating novel NSAIDs and other classes of drugs as the search continues to establish the first sporadic CRC chemoprevention strategy that can be implemented into routine practice.


Acknowledgments

Funding: This manuscript was supported by the 2024 Kure It Cancer Research Grant from Kure It Cancer Research, awarded to J.G.


Footnote

Reporting Checklist: The authors have completed the Narrative Review reporting checklist. Available at https://tgh.amegroups.com/article/view/10.21037/tgh-24-97/rc

Peer Review File: Available at https://tgh.amegroups.com/article/view/10.21037/tgh-24-97/prf

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://tgh.amegroups.com/article/view/10.21037/tgh-24-97/coif). J.G. serves as an unpaid editorial board member of Translational Gastroenterology and Hepatology from August 2024 to December 2026. A.H. reports existing or prior grants from Ipsen and NGM Biopharmaceuticals (Inst); consulting or advisory role at Amgen, Faraday Pharmaceuticals, Ipsen, Merus, Norvartis, Pancreatic Cancer Action Network, Regeneron, Valar Labs, Varian Medical Systems; support for travel from Halozyme; other relationship with Rayzebio. J.A.Z reports existing or prior grants from NIH and consulting fees from Exact Sciences and Tempus. J.G. reports consulting fees and honoraria from EMD Serono, Exelixis, Natera, Eisai, Janssen, Pfizer, Bayer, Taiho, Agenus, Seagen, and serves as the Chair of the Data Safety Monitoring Board at Cedars-Sinai Medical Center. The other authors have no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Siegel RL, Giaquinto AN, Jemal A. Cancer statistics, 2024. CA Cancer J Clin 2024;74:12-49. [Crossref] [PubMed]
  2. Morgan E, Arnold M, Gini A, et al. Global burden of colorectal cancer in 2020 and 2040: incidence and mortality estimates from GLOBOCAN. Gut 2023;72:338-44. [Crossref] [PubMed]
  3. US Preventive Services Task Force. Screening for Colorectal Cancer: US Preventive Services Task Force Recommendation Statement. JAMA 2021;325:1965-77. [Crossref] [PubMed]
  4. Rex DK, Boland CR, Dominitz JA, et al. Colorectal Cancer Screening: Recommendations for Physicians and Patients from the U.S. Multi-Society Task Force on Colorectal Cancer. Am J Gastroenterol 2017;112:1016-30. [Crossref] [PubMed]
  5. Brenner H, Hoffmeister M, Stegmaier C, et al. Risk of progression of advanced adenomas to colorectal cancer by age and sex: estimates based on 840,149 screening colonoscopies. Gut 2007;56:1585-9. [Crossref] [PubMed]
  6. Cheng X, Xu X, Chen D, et al. Therapeutic potential of targeting the Wnt/β-catenin signaling pathway in colorectal cancer. Biomed Pharmacother 2019;110:473-81. [Crossref] [PubMed]
  7. Goss KH, Groden J. Biology of the adenomatous polyposis coli tumor suppressor. J Clin Oncol 2000;18:1967-79. [Crossref] [PubMed]
  8. Fearon ER, Vogelstein B. A genetic model for colorectal tumorigenesis. Cell 1990;61:759-67. [Crossref] [PubMed]
  9. Kinzler KW, Vogelstein B. Lessons from hereditary colorectal cancer. Cell 1996;87:159-70. [Crossref] [PubMed]
  10. Powell SM, Zilz N, Beazer-Barclay Y, et al. APC mutations occur early during colorectal tumorigenesis. Nature 1992;359:235-7. [Crossref] [PubMed]
  11. Miyoshi Y, Nagase H, Ando H, et al. Somatic mutations of the APC gene in colorectal tumors: mutation cluster region in the APC gene. Hum Mol Genet 1992;1:229-33. [Crossref] [PubMed]
  12. Korinek V, Barker N, Morin PJ, et al. Constitutive transcriptional activation by a beta-catenin-Tcf complex in APC-/- colon carcinoma. Science 1997;275:1784-7. [Crossref] [PubMed]
  13. Anastas JN, Moon RT. WNT signalling pathways as therapeutic targets in cancer. Nat Rev Cancer 2013;13:11-26. [Crossref] [PubMed]
  14. Schneikert J, Behrens J. The canonical Wnt signalling pathway and its APC partner in colon cancer development. Gut 2007;56:417-25. [Crossref] [PubMed]
  15. Vogelstein B, Fearon ER, Kern SE, et al. Allelotype of colorectal carcinomas. Science 1989;244:207-11. [Crossref] [PubMed]
  16. Bos JL, Fearon ER, Hamilton SR, et al. Prevalence of ras gene mutations in human colorectal cancers. Nature 1987;327:293-7. [Crossref] [PubMed]
  17. Takaku K, Oshima M, Miyoshi H, et al. Intestinal tumorigenesis in compound mutant mice of both Dpc4 (Smad4) and Apc genes. Cell 1998;92:645-56. [Crossref] [PubMed]
  18. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell 2011;144:646-74. [Crossref] [PubMed]
  19. Lepore Signorile M, Grossi V, Fasano C, et al. Colorectal Cancer Chemoprevention: A Dream Coming True? Int J Mol Sci 2023;24:7597. [Crossref] [PubMed]
  20. Drew DA, Cao Y, Chan AT. Aspirin and colorectal cancer: the promise of precision chemoprevention. Nat Rev Cancer 2016;16:173-86. [Crossref] [PubMed]
  21. Simoni O, Scarpa M, Castagliuolo I, et al. IMMUNOREACT 7: Regular aspirin use is associated with immune surveillance activation in colorectal cancer. Cancer 2024;130:2272-86. [Crossref] [PubMed]
  22. US Preventive Services Task Force. Aspirin Use to Prevent Cardiovascular Disease: US Preventive Services Task Force Recommendation Statement. JAMA 2022;327:1577-84. [Crossref] [PubMed]
  23. Cook NR, Lee IM, Gaziano JM, et al. Low-dose aspirin in the primary prevention of cancer: the Women's Health Study: a randomized controlled trial. JAMA 2005;294:47-55. [Crossref] [PubMed]
  24. Cook NR, Lee IM, Zhang SM, et al. Alternate-day, low-dose aspirin and cancer risk: long-term observational follow-up of a randomized trial. Ann Intern Med 2013;159:77-85. [Crossref] [PubMed]
  25. Flossmann E, Rothwell PMBritish Doctors Aspirin Trial and the UK-TIA Aspirin Trial. Effect of aspirin on long-term risk of colorectal cancer: consistent evidence from randomised and observational studies. Lancet 2007;369:1603-13. [Crossref] [PubMed]
  26. Gann PH, Manson JE, Glynn RJ, et al. Low-dose aspirin and incidence of colorectal tumors in a randomized trial. J Natl Cancer Inst 1993;85:1220-4. [Crossref] [PubMed]
  27. Rothwell PM, Wilson M, Elwin CE, et al. Long-term effect of aspirin on colorectal cancer incidence and mortality: 20-year follow-up of five randomised trials. Lancet 2010;376:1741-50. [Crossref] [PubMed]
  28. Stürmer T, Glynn RJ, Lee IM, et al. Aspirin use and colorectal cancer: post-trial follow-up data from the Physicians' Health Study. Ann Intern Med 1998;128:713-20. [Crossref] [PubMed]
  29. McNeil JJ, Nelson MR, Woods RL, et al. Effect of Aspirin on All-Cause Mortality in the Healthy Elderly. N Engl J Med 2018;379:1519-28. [Crossref] [PubMed]
  30. Baron JA, Cole BF, Sandler RS, et al. A randomized trial of aspirin to prevent colorectal adenomas. N Engl J Med 2003;348:891-9. [Crossref] [PubMed]
  31. Benamouzig R, Deyra J, Martin A, et al. Daily soluble aspirin and prevention of colorectal adenoma recurrence: one-year results of the APACC trial. Gastroenterology 2003;125:328-36. [Crossref] [PubMed]
  32. Benamouzig R, Uzzan B, Deyra J, et al. Prevention by daily soluble aspirin of colorectal adenoma recurrence: 4-year results of the APACC randomised trial. Gut 2012;61:255-61. [Crossref] [PubMed]
  33. Hull MA, Sprange K, Hepburn T, 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. Lancet 2018;392:2583-94. [Crossref] [PubMed]
  34. Ishikawa H, Mutoh M, Suzuki S, et al. The preventive effects of low-dose enteric-coated aspirin tablets on the development of colorectal tumours in Asian patients: a randomised trial. Gut 2014;63:1755-9. [Crossref] [PubMed]
  35. Logan RF, Grainge MJ, Shepherd VC, et al. Aspirin and folic acid for the prevention of recurrent colorectal adenomas. Gastroenterology 2008;134:29-38. [Crossref] [PubMed]
  36. Sandler RS, Halabi S, Baron JA, et al. A randomized trial of aspirin to prevent colorectal adenomas in patients with previous colorectal cancer. N Engl J Med 2003;348:883-90. [Crossref] [PubMed]
  37. Cole BF, Logan RF, Halabi S, et al. Aspirin for the chemoprevention of colorectal adenomas: meta-analysis of the randomized trials. J Natl Cancer Inst 2009;101:256-66. [Crossref] [PubMed]
  38. Elwood P, Morgan G, Watkins J, et al. Aspirin and cancer treatment: systematic reviews and meta-analyses of evidence: for and against. Br J Cancer 2024;130:3-8. [Crossref] [PubMed]
  39. Arber N, Eagle CJ, Spicak J, et al. Celecoxib for the prevention of colorectal adenomatous polyps. N Engl J Med 2006;355:885-95. [Crossref] [PubMed]
  40. Baron JA, Sandler RS, Bresalier RS, et al. A randomized trial of rofecoxib for the chemoprevention of colorectal adenomas. Gastroenterology 2006;131:1674-82. [Crossref] [PubMed]
  41. Bertagnolli MM, Eagle CJ, Zauber AG, et al. Celecoxib for the prevention of sporadic colorectal adenomas. N Engl J Med 2006;355:873-84. [Crossref] [PubMed]
  42. Meyskens FL Jr, McLaren CE, Pelot D, et al. Difluoromethylornithine plus sulindac for the prevention of sporadic colorectal adenomas: a randomized placebo-controlled, double-blind trial. Cancer Prev Res (Phila) 2008;1:32-8. [Crossref] [PubMed]
  43. Takayama T, Nagashima H, Maeda M, et al. Randomized double-blind trial of sulindac and etodolac to eradicate aberrant crypt foci and to prevent sporadic colorectal polyps. Clin Cancer Res 2011;17:3803-11. [Crossref] [PubMed]
  44. Coni S, Bordone R, Ivy DM, et al. Combined inhibition of polyamine metabolism and eIF5A hypusination suppresses colorectal cancer growth through a converging effect on MYC translation. Cancer Lett 2023;559:216120. [Crossref] [PubMed]
  45. Zell JA, Pelot D, Chen WP, et al. Risk of cardiovascular events in a randomized placebo-controlled, double-blind trial of difluoromethylornithine plus sulindac for the prevention of sporadic colorectal adenomas. Cancer Prev Res (Phila) 2009;2:209-12. [Crossref] [PubMed]
  46. Results of the low-dose (20 mg) pravastatin GISSI Prevenzione trial in 4271 patients with recent myocardial infarction: do stopped trials contribute to overall knowledge? GISSI Prevenzione Investigators (Gruppo Italiano per lo Studio della Sopravvivenza nell'Infarto Miocardico). Ital Heart J 2000;1:810-20. [PubMed]
  47. ALLHAT Officers and Coordinators for the ALLHAT Collaborative Research Group. Major outcomes in moderately hypercholesterolemic, hypertensive patients randomized to pravastatin vs usual care: The Antihypertensive and Lipid-Lowering Treatment to Prevent Heart Attack Trial (ALLHAT-LLT). JAMA 2002;288:2998-3007. [Crossref] [PubMed]
  48. Bonovas S, Filioussi K, Flordellis CS, et al. Statins and the risk of colorectal cancer: a meta-analysis of 18 studies involving more than 1.5 million patients. J Clin Oncol 2007;25:3462-8. [Crossref] [PubMed]
  49. Downs JR, Clearfield M, Weis S, et al. Primary prevention of acute coronary events with lovastatin in men and women with average cholesterol levels: results of AFCAPS/TexCAPS. Air Force/Texas Coronary Atherosclerosis Prevention Study. JAMA 1998;279:1615-22. [Crossref] [PubMed]
  50. Ford I, Murray H, Packard CJ, et al. Long-term follow-up of the West of Scotland Coronary Prevention Study. N Engl J Med 2007;357:1477-86. [Crossref] [PubMed]
  51. MRC/BHF Heart Protection Study of cholesterol lowering with simvastatin in 20,536 high-risk individuals: a randomised placebo-controlled trial. Lancet 2002;360:7-22. [Crossref] [PubMed]
  52. Higurashi T, Hosono K, Takahashi H, et al. Metformin for chemoprevention of metachronous colorectal adenoma or polyps in post-polypectomy patients without diabetes: a multicentre double-blind, placebo-controlled, randomised phase 3 trial. Lancet Oncol 2016;17:475-83. [Crossref] [PubMed]
  53. Hosono K, Endo H, Takahashi H, et al. Metformin suppresses colorectal aberrant crypt foci in a short-term clinical trial. Cancer Prev Res (Phila) 2010;3:1077-83. [Crossref] [PubMed]
  54. Long-term effectiveness and safety of pravastatin in 9014 patients with coronary heart disease and average cholesterol concentrations: the LIPID trial follow-up. Lancet 2002;359:1379-87. [Crossref] [PubMed]
  55. Sacks FM, Pfeffer MA, Moye LA, et al. The effect of pravastatin on coronary events after myocardial infarction in patients with average cholesterol levels. Cholesterol and Recurrent Events Trial investigators. N Engl J Med 1996;335:1001-9. [Crossref] [PubMed]
  56. Strandberg TE, Pyörälä K, Cook TJ, et al. Mortality and incidence of cancer during 10-year follow-up of the Scandinavian Simvastatin Survival Study (4S). Lancet 2004;364:771-7. [Crossref] [PubMed]
  57. Zell JA, McLaren CE, Morgan TR, et al. A Phase IIa Trial of Metformin for Colorectal Cancer Risk Reduction among Individuals with History of Colorectal Adenomas and Elevated Body Mass Index. Cancer Prev Res (Phila) 2020;13:203-12. [Crossref] [PubMed]
  58. Katona BW, Weiss JM. Chemoprevention of Colorectal Cancer. Gastroenterology 2020;158:368-88. [Crossref] [PubMed]
  59. Cole BF, Baron JA, Sandler RS, et al. Folic acid for the prevention of colorectal adenomas: a randomized clinical trial. JAMA 2007;297:2351-9. [Crossref] [PubMed]
  60. Figueiredo JC, Levine AJ, Grau MV, et al. Colorectal adenomas in a randomized folate trial: the role of baseline dietary and circulating folate levels. Cancer Epidemiol Biomarkers Prev 2008;17:2625-31. [Crossref] [PubMed]
  61. Jaszewski R, Misra S, Tobi M, et al. Folic acid supplementation inhibits recurrence of colorectal adenomas: a randomized chemoprevention trial. World J Gastroenterol 2008;14:4492-8. [Crossref] [PubMed]
  62. Zhang SM, Cook NR, Albert CM, et al. Effect of combined folic acid, vitamin B6, and vitamin B12 on cancer risk in women: a randomized trial. JAMA 2008;300:2012-21. [Crossref] [PubMed]
  63. Hankey GJ, Eikelboom JW, Yi Q, et al. Treatment with B vitamins and incidence of cancer in patients with previous stroke or transient ischemic attack: results of a randomized placebo-controlled trial. Stroke 2012;43:1572-7. [Crossref] [PubMed]
  64. Gao QY, Chen HM, Chen YX, et al. Folic acid prevents the initial occurrence of sporadic colorectal adenoma in Chinese older than 50 years of age: a randomized clinical trial. Cancer Prev Res (Phila) 2013;6:744-52. [Crossref] [PubMed]
  65. Baron JA, Beach M, Mandel JS, et al. Calcium supplements for the prevention of colorectal adenomas. Calcium Polyp Prevention Study Group. N Engl J Med 1999;340:101-7. [Crossref] [PubMed]
  66. Bonithon-Kopp C, Kronborg O, Giacosa A, et al. Calcium and fibre supplementation in prevention of colorectal adenoma recurrence: a randomised intervention trial. European Cancer Prevention Organisation Study Group. Lancet 2000;356:1300-6. [Crossref] [PubMed]
  67. Wactawski-Wende J, Kotchen JM, Anderson GL, et al. Calcium plus vitamin D supplementation and the risk of colorectal cancer. N Engl J Med 2006;354:684-96. [Crossref] [PubMed]
  68. Manson JE, Cook NR, Lee IM, et al. Vitamin D Supplements and Prevention of Cancer and Cardiovascular Disease. N Engl J Med 2019;380:33-44. [Crossref] [PubMed]
  69. Ahearn TU, McCullough ML, Flanders WD, et al. A randomized clinical trial of the effects of supplemental calcium and vitamin D3 on markers of their metabolism in normal mucosa of colorectal adenoma patients. Cancer Res 2011;71:413-23. [Crossref] [PubMed]
  70. Ahearn TU, Shaukat A, Flanders WD, et al. A randomized clinical trial of the effects of supplemental calcium and vitamin D3 on the APC/β-catenin pathway in the normal mucosa of colorectal adenoma patients. Cancer Prev Res (Phila) 2012;5:1247-56. [Crossref] [PubMed]
  71. Tu H, Flanders WD, Ahearn TU, et al. Effects of calcium and vitamin D3 on transforming growth factors in rectal mucosa of sporadic colorectal adenoma patients: a randomized controlled trial. Mol Carcinog 2015;54:270-80. [Crossref] [PubMed]
  72. Qin T, Du M, Du H, et al. Folic acid supplements and colorectal cancer risk: meta-analysis of randomized controlled trials. Sci Rep 2015;5:12044. [Crossref] [PubMed]
  73. Dulai PS, Singh S, Marquez E, et al. Chemoprevention of colorectal cancer in individuals with previous colorectal neoplasia: systematic review and network meta-analysis. BMJ 2016;355:i6188. [Crossref] [PubMed]
  74. Jung YS, Park CH, Eun CS, et al. Metformin use and the risk of colorectal adenoma: A systematic review and meta-analysis. J Gastroenterol Hepatol 2017;32:957-65. [Crossref] [PubMed]
  75. Ma Y, You W, Cao Y, et al. Chemoprevention of colorectal cancer in general population and high-risk population: a systematic review and network meta-analysis. Chin Med J (Engl) 2023;136:788-98. [PubMed]
  76. Kanth P, Grimmett J, Champine M, et al. Hereditary Colorectal Polyposis and Cancer Syndromes: A Primer on Diagnosis and Management. Am J Gastroenterol 2017;112:1509-25. [Crossref] [PubMed]
  77. Ferlitsch M, Reinhart K, Pramhas S, et al. Sex-specific prevalence of adenomas, advanced adenomas, and colorectal cancer in individuals undergoing screening colonoscopy. JAMA 2011;306:1352-8. [Crossref] [PubMed]
  78. Yamaji Y, Mitsushima T, Ikuma H, et al. Incidence and recurrence rates of colorectal adenomas estimated by annually repeated colonoscopies on asymptomatic Japanese. Gut 2004;53:568-72. [Crossref] [PubMed]
  79. Shi X, Yang Z, Wu Q, et al. Colorectal adenoma recurrence rates among post-polypectomy patients in the placebo-controlled groups of randomized clinical trials: a meta-analysis. Oncotarget 2017;8:62371-81. [Crossref] [PubMed]
  80. Bertagnolli MM, Eagle CJ, Zauber AG, et al. Five-year efficacy and safety analysis of the Adenoma Prevention with Celecoxib Trial. Cancer Prev Res (Phila) 2009;2:310-21. [Crossref] [PubMed]
  81. Ng SC, Kong J, Stupart D, et al. Is Colonoscopic Surveillance Necessary after Curative Resection of Colorectal Cancer in Elderly Patients? World Journal of Colorectal Surgery 2019;8:65-8. [Crossref]
  82. Ochs-Balcom HM, Kanth P, Cannon-Albright LA. Early-onset colorectal cancer risk extends to second- and third-degree relatives. Cancer Epidemiol 2021;73:101973. [Crossref] [PubMed]
  83. Song M, Emilsson L, Roelstraete B, et al. Risk of colorectal cancer in first degree relatives of patients with colorectal polyps: nationwide case-control study in Sweden. BMJ 2021;373:n877. [Crossref] [PubMed]
  84. Henney JE. Celecoxib indicated for FAP. JAMA 2000;283:1131. [Crossref] [PubMed]
  85. Zell JA, McLaren CE, Chen WP, et al. Ornithine decarboxylase-1 polymorphism, chemoprevention with eflornithine and sulindac, and outcomes among colorectal adenoma patients. J Natl Cancer Inst 2010;102:1513-6. [Crossref] [PubMed]
  86. Zell J, You YN, Boughey JC. PACES trial: Evaluating the effectiveness of eflornithine and sulindac in preventing colon adenomas. Bull Am Coll Surg 2015;100:70-1. [PubMed]
doi: 10.21037/tgh-24-97
Cite this article as: Monson S, Chen PJ, Gangi A, Waters K, Billet S, Hendifar A, Lu S, Zell JA, Gong J. Chemopreventive strategies for sporadic colorectal cancer: a narrative review. Transl Gastroenterol Hepatol 2025;10:11.

Download Citation