In English | En español
Questions About Cancer? 1-800-4-CANCER

Colorectal Cancer Prevention (PDQ®)

  • Last Modified: 02/27/2014

Page Options

  • Print This Document
  • Email This Document

Who is at Risk?

For the great majority of people, the major factor that increases a person’s risk for colorectal cancer (CRC) is increasing age. Risk increases dramatically after age 50 years; 90% of all CRCs are diagnosed after this age. The history of CRC in a first-degree relative, especially if before the age of 55 years, roughly doubles the risk. Other risk factors are weaker than age and family history. People with inflammatory bowel disease have a much higher risk of CRC. A small percentage (<5%) of CRCs occur in people with a genetic predisposition, including familial adenomatous polyposis and hereditary nonpolyposis coli.

Overview

Note: Separate PDQ summaries on Colorectal Cancer Screening; Colon Cancer Treatment; and Rectal Cancer Treatment are also available.

Factors Associated With Increased Risk of Colorectal Cancer

Excessive alcohol use

Based on solid evidence from observational studies, excessive alcohol use is associated with an increased risk of colorectal cancer (CRC).[1-3]

Magnitude of Effect: A pooled analysis of eight cohort studies estimated an adjusted relative risk (RR) of 1.41 (95% confidence interval [CI], 1.16–1.72) for consumption exceeding 45 g/day.[3]

Study Design: Cohort studies.
Internal Validity: Fair.
Consistency: Fair.
External Validity: Fair.
Cigarette smoking

Based on solid evidence, cigarette smoking is associated with increased incidence and mortality from CRC.

Magnitude of Effect: A pooled analysis of 106 observational studies estimated an adjusted RR (current smokers vs. never smokers) for developing CRC of 1.18 (95% CI, 1.11–1.25).[4]

Study Design: One-hundred six observational studies.
Internal Validity: Fair.
Consistency: Good.
External Validity: Good.
Obesity

Based on solid evidence, obesity is associated with increased incidence and mortality from CRC.

Magnitude of Effect: In one large cohort study, the adjusted RR for developing colon cancer for women with a body mass index of more than 29 was 1.45 (95% CI, 1.02–2.07).[5,6] A similar increase in CRC mortality was found in another large cohort study.[7]

Study Design: Large cohort studies.
Internal Validity: Fair.
Consistency: Good.
External Validity: Good.
Factors Associated With a Decreased Risk of Colorectal Cancer

Physical activity

Based on solid evidence, regular physical activity is associated with a decreased incidence of CRC.

Magnitude of Effect: A meta-analysis of 52 observational studies found a statistically significant 24% reduction in CRC incidence (RR = 0.76; 95% CI, 0.72–0.81).[8]

Study Design: Cohort studies and meta-analysis.
Internal Validity: Fair.
Consistency: Good.
External Validity: Good.
Interventions Associated With Decreased Risk of Colorectal Cancer

Nonsteroidal anti-inflammatory drugs

Benefits

There is inadequate evidence that the use of nonsteroidal anti-inflammatory drugs (NSAIDs) reduces the risk of CRC. In people without genetic predisposition but with a prior history of a colonic adenoma that had been removed, three randomized controlled trials (RCT) found that celecoxib [9,10] and rofecoxib [11] decrease the incidence of recurrent adenoma, although follow-up was too short to determine whether the incidence of CRC would have been affected.

Based on solid evidence, NSAIDs reduce the risk of adenomas, but the extent to which this translates into a reduction of CRC is uncertain.

Study Design: No adequate studies with CRC outcome.
Internal Validity: Not applicable (N/A).
Consistency: N/A.
External Validity: N/A.
Harms

Based on solid evidence, harms of NSAID use are relatively common and potentially serious, and include upper gastrointestinal bleeding, chronic kidney disease, and serious cardiovascular events such as myocardial infarction, heart failure, and hemorrhagic stroke.[12]

Magnitude of Effect: The estimated average excess risk of upper gastrointestinal complications in average-risk people attributable to NSAIDs is 4/1,000 to 5/1,000 people per year.[13,14] The excess risk varies with the underlying gastrointestinal risk, however, it likely exceeds ten extra cases per 1,000 people per year in more than 10% of users.[15] Serious cardiovascular events are increased by 50% to 60%.[14]

Study Design: Evidence obtained from RCTs and high quality systematic reviews and meta-analyses.[13,14]
Internal Validity: Good.
Consistency: Good.
External Validity: Good.
Aspirin

Benefits

Based on solid evidence, daily aspirin (acetylsalicylic acid [ASA]) for at least 5 years reduces CRC incidence and mortality. This is based on two reports of extended follow-up of two RCTs [16,17] and meta-analysis of observational studies.[16] A third report that adds extended follow-up of an additional two RCTs (with a meta-analysis of all four RCTs) adds certainty to this conclusion.[18]

Magnitude of Effect: After 23 years of follow-up, incidence of CRC in the placebo group was 3.8% and in the ASA group 2.5% (hazard ratio [HR] = 0.63; 95% CI, 0.47–0.85). In the report from all four RCTs, the 20-year risk of death due to CRC in trials that allocated ASA for at least 5 years was reduced by about 40% (HR = 0.60; 95% CI, 0.45–0.81), absolute risk reduction was from about 3.1% to 1.9%. The primary effect was on mortality from proximal colon cancer.

Study Design: Extended follow-up of four RCTs.
Internal Validity: Fair, some data from registries and death certificates, some loss to follow-up; variation in ASA dose; adherence to ASA unknown after end of trials (5–9 years).
Consistency: Consistent.
External Validity: Fair, most data from British men; fewer data from women.
Harms

Based on solid evidence, harms of ASA use include excessive bleeding, including gastrointestinal bleeds and hemorrhagic stroke.

Magnitude of Effect: The estimated average excess risk of upper gastrointestinal complications is 10 to 30 per 1,000 people for a period of 10 years, on the higher end for men and on the lower end for women. Risk increases with age.[19]

Study Design: Evidence obtained from large databases.[15,20]
Internal Validity: Good.
Consistency: Good.
External Validity: Good, data from national databases.
Hormone therapy (estrogen plus progestin)

Based on solid evidence, combined hormone therapy (conjugated equine estrogen and progestin) decreases the incidence of invasive CRC.[21]

Based on fair evidence, combination conjugated equine estrogen and progestin has little or no benefit in reducing mortality from CRC. Data from the Women’s Health Initiative (WHI), a randomized, placebo-controlled trial evaluating estrogen plus progestin, with a mean intervention of 5.6 years and a follow-up of 11.6 years showed that women taking combined hormone therapy had a statistically significant higher stage of cancer (regional and distant) at diagnosis but not a statistically significant number of deaths from CRC compared with women taking the placebo.[21]

Magnitude of Effect: There were fewer CRCs in the combined hormone therapy group than in the placebo group (0.12% vs. 0.16%; HR = 0.72; 95% CI, 0.56–0.94). A meta-analysis of cohort studies observed a RR of 0.86 (95% CI, 0.76–0.97) for incidence of CRC associated with combined hormone therapy.

There were 37 CRC deaths in the combined hormone therapy arm compared with 27 deaths in the placebo arm (0.04% vs. 0.03%; HR 1.29; 95% CI, 0.78–2.11).

  • Study Design: Randomized controlled trial and cohort studies
  • Internal Validity: Good.
  • Consistency: Good for effect on incidence; N/A for effect on mortality; results were based on one trial.
  • External Validity: Good.
Estrogen-only therapy

Benefits

Based on fair evidence, conjugated equine estrogens do not affect the incidence of, or survival from, invasive CRC.[22]

Magnitude of Effect: N/A.

Study Design: One randomized controlled trial.
Internal Validity: Good.
Consistency: N/A.
External Validity: Good.
Harms

Based on solid evidence, harms of postmenopausal combined estrogen plus progestin hormone use include increased risk of breast cancer, coronary heart disease, and thromboembolic events.

Magnitude of Effect: The WHI showed a 26% increase in invasive breast cancer in the combined hormone group, a 29% increase in coronary heart disease events, a 41% increase in stroke rates, and a twofold higher rate of thromboembolic events.[23]

Study Design: Evidence from RCTs.
Internal Validity: Good.
Consistency: Good.
External Validity: Fair.
Polyp removal

Benefits

Based on fair evidence, removal of adenomatous polyps reduces the risk of CRC. Much of this reduction likely comes from removal of large (i.e., >1.0 cm) polyps, while the benefit of removing smaller polyps—which are much more common—is unknown. Some but not all observational evidence indicates that this reduction may be greater for left-sided CRC than for right-sided CRC.[24-26]

Magnitude of Effect: Unknown, probably greater for larger polyps (i.e., >1.0 cm) than smaller ones.[27]

Study Design: Evidence obtained from cohort studies and one RCT of sigmoidoscopy.[25]
Internal Validity: Good.
Consistency: Consistent.
External Validity: Good.
Harms

Based on solid evidence, the major harms of polyp removal include perforation of the colon and bleeding.

Magnitude of Effect: Seven to nine events per 1,000 procedures.[28-30]

Study Design: Evidence from retrospective cohort studies.[29,30]
Internal Validity: Good.
Consistency: Good.
External Validity: Good.
Diet modification

A diet low in fat and meat and high in fiber, fruits, and vegetables

Benefits

Based on fair evidence, a diet low in fat and meat and high in fiber, fruits, and vegetables started as an adult does not reduce the risk of CRC by a clinically important degree.

Study Design: Evidence obtained from pooled analyses of multiple cohort studies and RCTs.
Internal Validity: Fair, but measurement error is a potential problem.
Consistency: Some inconsistency, with one large pooled study [31] finding a reduced risk of distal colon cancer associated with fruit and vegetable intake. RCTs have been consistent.
External Validity: Good, studies have included large population-based studies.
Harms

There are no known harms from dietary modification, including reduction of fatty acids or meats and an increase in the intake of fiber, fruits, and vegetables.

Study Design: Cohort and RCTs.
Internal Validity: Good.
Consistency: Good.
External Validity: Good.
Calcium supplementation

Benefits

The evidence is inadequate to determine whether calcium supplementation reduces the risk of CRC.

Study Design: Pooled and individual prospective cohort studies, meta-analysis of three RCTs with adenoma recurrence as an outcome, and one large individual RCT in women with CRC as an outcome.
Internal Validity: Good.
Consistency: Poor; the RCT with CRC as an outcome [32] found no reduction in CRC incidence, while prospective cohort studies found a reduction in CRC incidence between high and low calcium groups; three RCTs found a reduction in adenoma recurrence with calcium supplementation.[33]
External Validity: N/A.
Harms

The evidence is fair that elemental calcium without vitamin D as a supplement in the level of 1,000 to 1,200 mg/day increases the risk of myocardial infarction. Based on fair evidence, calcium supplementation with vitamin D at doses less than 1,000 has few harms.

Magnitude of Effect: In a meta-analysis of RCTs of calcium alone, the risk of myocardial infarction was increased from 4.8% to 5.8% (HR = 1.31; 95% CI, 1.02–1.67).[34]

Study Design: Pooled and individual prospective cohort studies plus RCTs.
Internal Validity: Good.
Consistency: Poor (one meta-analysis of 11 RCTs for calcium alone found increased myocardial infarction; no other individual studies found this).[34]
External Validity: Good.
Statins

Benefits

Based on solid evidence, statins do not reduce the incidence or mortality from CRC.

Study Design: Meta-analyses of RCTs.[35-37]
Internal Validity: Good.
Consistency: Good.
External Validity: N/A.
Harms

Based on solid evidence, the harms of statins are small.

Study Design: Observational studies,[38] multiple RCTs, and a review.[39]
Internal Validity: Good.
Consistency: Good.
External Validity: Good.
References
  1. Longnecker MP, Orza MJ, Adams ME, et al.: A meta-analysis of alcoholic beverage consumption in relation to risk of colorectal cancer. Cancer Causes Control 1 (1): 59-68, 1990.  [PUBMED Abstract]

  2. Gapstur SM, Potter JD, Folsom AR: Alcohol consumption and colon and rectal cancer in postmenopausal women. Int J Epidemiol 23 (1): 50-7, 1994.  [PUBMED Abstract]

  3. Cho E, Smith-Warner SA, Ritz J, et al.: Alcohol intake and colorectal cancer: a pooled analysis of 8 cohort studies. Ann Intern Med 140 (8): 603-13, 2004.  [PUBMED Abstract]

  4. Botteri E, Iodice S, Bagnardi V, et al.: Smoking and colorectal cancer: a meta-analysis. JAMA 300 (23): 2765-78, 2008.  [PUBMED Abstract]

  5. Martínez ME, Giovannucci E, Spiegelman D, et al.: Leisure-time physical activity, body size, and colon cancer in women. Nurses' Health Study Research Group. J Natl Cancer Inst 89 (13): 948-55, 1997.  [PUBMED Abstract]

  6. Giovannucci E, Ascherio A, Rimm EB, et al.: Physical activity, obesity, and risk for colon cancer and adenoma in men. Ann Intern Med 122 (5): 327-34, 1995.  [PUBMED Abstract]

  7. Calle EE, Rodriguez C, Walker-Thurmond K, et al.: Overweight, obesity, and mortality from cancer in a prospectively studied cohort of U.S. adults. N Engl J Med 348 (17): 1625-38, 2003.  [PUBMED Abstract]

  8. Wolin KY, Yan Y, Colditz GA, et al.: Physical activity and colon cancer prevention: a meta-analysis. Br J Cancer 100 (4): 611-6, 2009.  [PUBMED Abstract]

  9. Bertagnolli MM, Eagle CJ, Zauber AG, et al.: Celecoxib for the prevention of sporadic colorectal adenomas. N Engl J Med 355 (9): 873-84, 2006.  [PUBMED Abstract]

  10. Arber N, Eagle CJ, Spicak J, et al.: Celecoxib for the prevention of colorectal adenomatous polyps. N Engl J Med 355 (9): 885-95, 2006.  [PUBMED Abstract]

  11. Lanas A, Baron JA, Sandler RS, et al.: Peptic ulcer and bleeding events associated with rofecoxib in a 3-year colorectal adenoma chemoprevention trial. Gastroenterology 132 (2): 490-7, 2007.  [PUBMED Abstract]

  12. Bresalier RS, Sandler RS, Quan H, et al.: Cardiovascular events associated with rofecoxib in a colorectal adenoma chemoprevention trial. N Engl J Med 352 (11): 1092-102, 2005.  [PUBMED Abstract]

  13. Rostom A, Dubé C, Lewin G, et al.: Nonsteroidal anti-inflammatory drugs and cyclooxygenase-2 inhibitors for primary prevention of colorectal cancer: a systematic review prepared for the U.S. Preventive Services Task Force. Ann Intern Med 146 (5): 376-89, 2007.  [PUBMED Abstract]

  14. Kearney PM, Baigent C, Godwin J, et al.: Do selective cyclo-oxygenase-2 inhibitors and traditional non-steroidal anti-inflammatory drugs increase the risk of atherothrombosis? Meta-analysis of randomised trials. BMJ 332 (7553): 1302-8, 2006.  [PUBMED Abstract]

  15. Hernández-Díaz S, García Rodríguez LA: Cardioprotective aspirin users and their excess risk of upper gastrointestinal complications. BMC Med 4: 22, 2006.  [PUBMED Abstract]

  16. Flossmann E, Rothwell PM; British 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 369 (9573): 1603-13, 2007.  [PUBMED Abstract]

  17. 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 376 (9754): 1741-50, 2010.  [PUBMED Abstract]

  18. Rothwell PM, Fowkes FG, Belch JF, et al.: Effect of daily aspirin on long-term risk of death due to cancer: analysis of individual patient data from randomised trials. Lancet 377 (9759): 31-41, 2011.  [PUBMED Abstract]

  19. US Preventive Services Task Force: Aspirin for the prevention of cardiovascular disease: U.S. Preventive Services Task Force recommendation statement. Ann Intern Med 150 (6): 396-404, 2009.  [PUBMED Abstract]

  20. Hernández-Díaz S, Rodríguez LA: Incidence of serious upper gastrointestinal bleeding/perforation in the general population: review of epidemiologic studies. J Clin Epidemiol 55 (2): 157-63, 2002.  [PUBMED Abstract]

  21. Simon MS, Chlebowski RT, Wactawski-Wende J, et al.: Estrogen plus progestin and colorectal cancer incidence and mortality. J Clin Oncol 30 (32): 3983-90, 2012.  [PUBMED Abstract]

  22. Ritenbaugh C, Stanford JL, Wu L, et al.: Conjugated equine estrogens and colorectal cancer incidence and survival: the Women's Health Initiative randomized clinical trial. Cancer Epidemiol Biomarkers Prev 17 (10): 2609-18, 2008.  [PUBMED Abstract]

  23. Writing Group for the Women's Health Initiative Investigators: Risks and benefits of estrogen plus progestin in healthy postmenopausal women: principal results From the Women's Health Initiative randomized controlled trial. JAMA 288 (3): 321-33, 2002.  [PUBMED Abstract]

  24. Brenner H, Hoffmeister M, Arndt V, et al.: Protection from right- and left-sided colorectal neoplasms after colonoscopy: population-based study. J Natl Cancer Inst 102 (2): 89-95, 2010.  [PUBMED Abstract]

  25. Atkin WS, Edwards R, Kralj-Hans I, et al.: Once-only flexible sigmoidoscopy screening in prevention of colorectal cancer: a multicentre randomised controlled trial. Lancet 375 (9726): 1624-33, 2010.  [PUBMED Abstract]

  26. Brenner H, Chang-Claude J, Seiler CM, et al.: Protection from colorectal cancer after colonoscopy: a population-based, case-control study. Ann Intern Med 154 (1): 22-30, 2011.  [PUBMED Abstract]

  27. Robertson DJ, Greenberg ER, Beach M, et al.: Colorectal cancer in patients under close colonoscopic surveillance. Gastroenterology 129 (1): 34-41, 2005.  [PUBMED Abstract]

  28. Nelson DB, McQuaid KR, Bond JH, et al.: Procedural success and complications of large-scale screening colonoscopy. Gastrointest Endosc 55 (3): 307-14, 2002.  [PUBMED Abstract]

  29. Levin TR, Zhao W, Conell C, et al.: Complications of colonoscopy in an integrated health care delivery system. Ann Intern Med 145 (12): 880-6, 2006.  [PUBMED Abstract]

  30. Warren JL, Klabunde CN, Mariotto AB, et al.: Adverse events after outpatient colonoscopy in the Medicare population. Ann Intern Med 150 (12): 849-57, W152, 2009.  [PUBMED Abstract]

  31. Koushik A, Hunter DJ, Spiegelman D, et al.: Fruits, vegetables, and colon cancer risk in a pooled analysis of 14 cohort studies. J Natl Cancer Inst 99 (19): 1471-83, 2007.  [PUBMED Abstract]

  32. Wactawski-Wende J, Kotchen JM, Anderson GL, et al.: Calcium plus vitamin D supplementation and the risk of colorectal cancer. N Engl J Med 354 (7): 684-96, 2006.  [PUBMED Abstract]

  33. Shaukat A, Scouras N, Schünemann HJ: Role of supplemental calcium in the recurrence of colorectal adenomas: a metaanalysis of randomized controlled trials. Am J Gastroenterol 100 (2): 390-4, 2005.  [PUBMED Abstract]

  34. Bolland MJ, Avenell A, Baron JA, et al.: Effect of calcium supplements on risk of myocardial infarction and cardiovascular events: meta-analysis. BMJ 341: c3691, 2010.  [PUBMED Abstract]

  35. Baigent C, Keech A, Kearney PM, et al.: Efficacy and safety of cholesterol-lowering treatment: prospective meta-analysis of data from 90,056 participants in 14 randomised trials of statins. Lancet 366 (9493): 1267-78, 2005.  [PUBMED Abstract]

  36. Dale KM, Coleman CI, Henyan NN, et al.: Statins and cancer risk: a meta-analysis. JAMA 295 (1): 74-80, 2006.  [PUBMED Abstract]

  37. Browning DR, Martin RM: Statins and risk of cancer: a systematic review and metaanalysis. Int J Cancer 120 (4): 833-43, 2007.  [PUBMED Abstract]

  38. Hippisley-Cox J, Coupland C: Unintended effects of statins in men and women in England and Wales: population based cohort study using the QResearch database. BMJ 340: c2197, 2010.  [PUBMED Abstract]

  39. Joy TR, Hegele RA: Narrative review: statin-related myopathy. Ann Intern Med 150 (12): 858-68, 2009.  [PUBMED Abstract]

Description of the Evidence



Background

Incidence and mortality

Colorectal cancer (CRC) is the third most common malignant neoplasm worldwide [1] and the second leading cause of cancer deaths (irrespective of gender) in the United States.[2] It is estimated that there will be 136,830 new cases diagnosed in the United States in 2014 and 50,310 deaths due to this disease.[2] Between 2006 and 2010, CRC incidence rates in the United States declined by 3.7% per year among adults aged 50 years and older.[2] For the past 20 years, the mortality rate has been declining in both men and women. Between 2006 and 2010, the mortality rate declined by 2.5% per year in men and by 3.0% per year in women. In adults younger than 50 years, CRC incidence rates increased by about 1.8% per year.[2] The overall 5-year survival rate is 65%. About 5% of Americans are expected to develop the disease within their lifetimes.[2,3] The risk of CRC begins to increase after the age of 40 years and rises sharply at ages 50 to 55 years; the risk doubles with each succeeding decade, and continues to rise exponentially. Despite advances in surgical techniques and adjuvant therapy, there has been only a modest improvement in survival for patients who present with advanced neoplasms.[4,5] Hence, effective primary and secondary preventive approaches must be developed to reduce the morbidity and mortality from CRC.

Definition of prevention

Primary prevention involves the use of medications or other interventions before the clinical appearance of CRC with the intent of preventing clinical CRC and CRC mortality.

Etiology and pathogenesis of colorectal cancer

Genetics,[6,7] experimental,[8,9] and epidemiologic [10-12] studies suggest that CRC results from complex interactions between inherited susceptibility and environmental factors. The exact nature and contribution of these factors to CRC incidence and mortality is the subject of ongoing research.

Factors associated with increased risk of colorectal cancer

Excessive alcohol use

There is evidence of an association of CRC with alcoholic beverage consumption. In a meta-analysis of eight cohort studies, the relative risk (RR) for consumption of 45 g/day (i.e., about three standard drinks/day) compared with nondrinkers was 1.41 (95% confidence interval [CI], 1.16–1.72).[13] Case-control studies suggest a modest-to-strong positive relationship between alcohol consumption and large bowel cancers.[14,15] A meta-analysis found that the association did not vary by sex or location within the large bowel.[16]

Five studies have reported a positive association between alcohol intake and colorectal adenomas.[17] A case-control study of diet, genetic factors, and the adenoma-carcinoma sequence was conducted in Burgundy.[18] It separated adenomas smaller than 10.0 mm in diameter from larger adenomas. A positive association between current alcohol intake and adenomas was found to be limited to the larger adenomas, suggesting that alcohol intake could act at the promotional phase of the adenoma-carcinoma sequence.[18]

A large cohort study found a dose-response relationship between alcohol intake and death from CRC, with a RR of 1.2 (95% CI, 1.0–1.5) for four or more drinks per day compared with nondrinkers.[19]

Cigarette smoking

Most case-control studies of cigarette exposure and adenomas have found an elevated risk for smokers.[20] In addition, a significantly increased risk of adenoma recurrence following polypectomy has been associated with smoking in both men and women.[20] In the Nurses’ Health Study, the minimum induction period for cancer appears to be at least 35 years.[21] Similarly, in the Health Professionals Follow-up Study, a history of smoking was associated with both small and large adenomas and with a long induction period of at least 35 years for CRC.[22] In the Cancer Prevention Study II (CPS II), a large nationwide cohort study, multivariate-adjusted CRC mortality rates were highest among current smokers, intermediate among former smokers, and lowest in nonsmokers, with increased risk observed after 20 or more years of smoking in men and women combined.[23] On the basis of CPS II data, it was estimated that 12% of CRC deaths in the U.S. population in 1997 were attributable to smoking. A large population-based cohort study of Swedish twins found that heavy smoking of 35 or more years' duration was associated with a nearly threefold increased risk of developing colon cancer, though subsite analysis found a statistically significant effect only for rectal but not colon cancer.[24] Another large population-based case-control study supports the view that current tobacco use and tobacco use within the last 10 years is associated with colon cancer. A 50% increase in risk was associated with smoking more than a pack a day relative to never smoking.[25] However, a 28-year follow-up of 57,000 Finns showed no association between the development of CRC and baseline smoking status, though there was a 57% to 71% increased risk in persistent smokers.[26] No relationship was found between cigarette smoking, even smoking of long duration, and recurrence of adenomas in a population followed for 4 years after initial colonoscopy.[27]

A meta-analysis of 106 observational studies found a RR (ever smokers compared with nonsmokers) for CRC incidence of 1.18 (95% CI, 1.11–1.25), with an absolute risk increase of 10.8 cases per 100,000 person-years (95% CI, 7.9–13.6). There was a statistically significant dose-response effect. In 17 studies with data on CRC mortality, cigarette smoking was associated with CRC death, with a RR (ever smokers vs. never smokers) of 1.25 (95% CI, 1.14–1.37), and an absolute increase in the death rate of 6.0 deaths per 100,000 person-years. For both incidence and mortality, the association was stronger for rectal cancer than for colon cancer.[28]

Obesity

At least three large cohort studies have found an association between obesity and CRC incidence or mortality.[29-31] The Nurses’ Health Study found that women with a body mass index (BMI) of more than 29, compared with women with a BMI of less than 21, had an adjusted RR for CRC incidence of 1.45 (95% CI ,1.02–2.07).[29] In the CPS II [31], men and women with a BMI of 30 to 34.9 had an adjusted RR for CRC mortality (compared with people with a BMI of 18.5–24.9) of 1.47 (95% CI, 1.30–1.66), with a statistically significant dose-response effect.[31] The effects were similar in men and women.

Factors associated with a decreased risk of colorectal cancer

Physical activity

A sedentary lifestyle has been associated in some [32,33] but not all [34] studies with an increased risk of CRC. Numerous observational studies that have examined the relationship between physical activity and colon cancer risk.[35] Most of these studies have shown an inverse relationship between level of physical activity and colon cancer incidence. The average RR reduction is reportedly 40% to 50%. Large U.S. cohort studies have found statistically significant adjusted RR of 0.54 (95% CI, 0.33–0.90) [29] and 0.53 (95% CI, 0.32–0.88) [30] when comparing people with high versus low average energy expenditure. A meta-analysis of 52 observational studies found an overall adjusted RR of 0.76 (95% CI, 0.72–0.81), with similar results for men and women.[36]

Interventions associated with a decreased risk of colorectal cancer: Benefits and harms

Nonsteroidal anti-inflammatory drugs

One large cohort study (301,240 people with 3,894 colorectal cancer cases) found an association between daily or weekly nonaspirin (non–acetylsalicylic acid [non-ASA]) nonsteroidal anti-inflammatory drug (NSAID) use and reduced 10-year incidence of proximal and distal colon cancer, but not rectal cancer, with a hazard ratio (HR) of 0.67 (95% CI, 0.58–0.77) for daily use for colon cancer. Because exposure to non-ASA NSAIDs was assessed only once, assessment was by self-report, and there is no information on dose or duration of use, the certainty of this single study must be rated low. Further research is needed before this finding can be accepted.[37]

Although evidence is currently inadequate to determine whether NSAIDs reduce CRC incidence, proponents suggest that any effect of these drugs results from their ability to inhibit the activity of cyclooxygenase (COX). COX is important in the transformation of arachidonic acid into prostanoids, prostaglandins, and thromboxane A2. NSAIDs include not only aspirin (ASA, which is considered separately here) and other, first-generation nonselective inhibitors of the two functional isoforms of COX, termed COX-1 and COX-2, but also newer second-generation drugs that inhibit primarily COX-2. Normally, COX-1 is expressed in most tissues and primarily plays a housekeeping role (e.g., gastrointestinal mucosal protection and platelet aggregation). COX-2 activity is crucial in stress responses and in mediating and propagating the pain and inflammation that are characteristic of arthritis.[38]

Nonselective COX inhibitors include indomethacin (Indocin); sulindac (Clinoril); piroxicam (Feldene); diflunisal (Dolobid); ibuprofen (Advil, Motrin); ketoprofen (Orudis); naproxen (Naprosyn); and naproxen sodium (Aleve, Anaprox). Selective COX-2 inhibitors include celecoxib (Celebrex), rofecoxib (Vioxx), and valdecoxib (Bextra). Rofecoxib and valdecoxib are no longer marketed because of an associated increased risk of serious cardiovascular events.

Both celecoxib and rofecoxib have been associated with serious cardiovascular events including dose-related death from cardiovascular causes, myocardial infarction, stroke, or heart failure.[39-42] Four trials that demonstrated this increased risk are summarized in the Table. In addition, a network meta-analysis of all large scale randomized, controlled trials (RCTs) comparing any NSAID to any other NSAID or placebo found that there is little evidence to suggest that any of the investigated drugs are safe in terms of cardiovascular effects. Naproxen seemed least harmful.[43]

Cardiovascular Risks Associated With Celecoxib and Rofecoxib Dose/Drugs
Authors Dose/Trial Drug Risk Indication 
bid = twice a day; qd = every day; CI = confidence interval; HR = hazard ratio; OR = odds ratio; RR = relative risk.
[40]Rofecoxib <25 mg/qd; rofecoxib >25 mg/qdOR = 1.47 (0.99–2.17) 3 vs. 58 (1.27–10.17)Nested case-control study all users
[42]Celecoxib 200 mg/qd vs. 400 mg bid3.4%; HR = 3.4 (95% CI, 1.4–7.8)Sporadic adenoma prevention trial (N = 2,035)
[41]Rofecoxib 25 mg/qdRR = 1.92 (95% CI, 1.19–3.11; P = .008)Chemoprevention sporadic adenoma
[39]Rofecoxib 25 mg/qdRR (estimated) = 2.66 (95% CI, 1.03–6.86; P = .04)Chemoprevention sporadic adenoma median study Rx 7.4 months

Other major harms from all NSAIDs are gastrointestinal bleeding and renal impairment. The incidence of reported major gastrointestinal bleeding events appears to be dose-related.[44]

Celecoxib reduces the incidence of adenomas; however, celecoxib does not have a clinical role in reducing the risk of sporadic CRC. Its long-term efficacy in preventing CRC has not been shown due to increased risk of cardiovascular events, and because there are other effective ways, such as screening to reduce CRC mortality.[45] A population-based retrospective cohort study of nonaspirin NSAID use among individuals aged 65 years and older was associated with lower risk of CRC, particularly with longer durations of use.[46]

Several rigorous studies have demonstrated the effectiveness of sulindac in reducing the size and number of adenomas in familial polyposis.[47,48] In a randomized, double-blind, placebo-controlled study of 77 patients with familial adenomatous polyposis, patients receiving 400 mg of celecoxib twice a day had a 28.0% reduction in the mean number of colorectal adenomas (P = .003 for the comparison with placebo) and a 30.7% reduction in the polyp burden (sum of polyp diameters; P = .001) as compared with reductions of 4.5% and 4.9%, respectively, in the placebo group. The reductions in the group receiving 100 mg of celecoxib twice a day were 11.9% (P = .33 for the comparison with placebo) and 14.6% (P = .09), respectively. The incidence of adverse events was similar among the groups.[49]

The NSAID piroxicam, at a dose of 20 mg/day, reduced mean rectal prostaglandin concentration by 50% in individuals with a history of adenomas.[50] Several studies assessing the effect of ASA or other nonsteroidals on polyp recurrence following polypectomy are in progress.[51] In several of these studies, mucosal prostaglandin concentration is being measured.

The potential for the use of NSAIDs as a primary prevention measure is being studied. There are, however, several unresolved issues that mitigate against making general recommendations for their use. These include a paucity of knowledge about the proper dose and duration for these agents, and concern about whether the potential preventive benefits such as a reduction in the frequency or intensity of screening or surveillance could counterbalance long-term risks such as gastrointestinal ulceration and hemorrhagic stroke for the average-risk individual.[52]

Aspirin

The preponderance of evidence from both observational studies and long-term follow-up of RCTs indicates that daily ASA for at least 5 years reduces the incidence of CRC. Among a group of more than 600,000 adults enrolled in an American Cancer Society study, mortality in regular users of ASA was about 40% lower for cancers of the colon and rectum.[53,54] In a report from the Health Professionals Follow-up Study of 47,000 males, regular use of ASA (at least 2 times per week) was associated with a 30% overall reduction in CRC, including a 50% reduction in advanced cases.[55] In the Women's Health Study (WHS), a randomized 2 x 2 factorial trial of 100 mg of ASA every other day for an average of 10 years, similar rates of breast, colorectal, or other site-specific cancers were observed in both the ASA and placebo arms.[56] In a report from the Nurses’ Health Study involving 82,911 women followed for 20 years, the multivariate RR for colon cancer was 0.77 (95% CI, 0.67–0.88) among women who regularly used ASA (≥2 standard 325-mg tablets per week) compared with nonregular use. Significant RR was not observed, however, until more than 10 years of use. The benefit appeared to be dose-related (e.g., women who used more than 14 ASA per week for longer than 10 years had a multivariate RR for cancer of 0.47 [95% CI, 0.31–0.71]).

A systematic review of 46 observational studies of ASA and CRC in 2007 found a reduction in CRC (odds ratio [OR] for any use 0.80 [0.73–0.87]).[57] A large cohort study (301,240 people with 3,894 colorectal cancer cases) published after this systematic review found an association between weekly or daily ASA use and reduced 10-year incidence of distal and rectal (but not proximal) colorectal cancer, with an HR of 0.76 (95% CI, 0.64–0.90) for rectal cancer for daily use. However, use was assessed at only one time, and there is no information about dose or duration of use.[37]

In the Physicians’ Health Study, 22,000 men aged 40 to 84 years were randomly assigned to receive placebo or ASA (325 mg every other day) for 5 years. There was no reduction in invasive cancers or adenomas at a median follow-up of 4.5 years.[58] In a subsequent analysis of more than 12 years, both randomized and observational analyses indicated that there was no association between the use of ASA and the incidence of CRC. The low dose of ASA and the short treatment period may account for the null findings.[59]

In a randomized study of 635 patients with prior CRC (T1–T2 N0 M0) who had undergone curative resection, ASA intake at 325 mg/day was associated with a decrease in the adjusted RR of any recurrent adenoma as compared with the placebo group (0.65; 95% CI, 0.46–0.91) after a median duration of treatment of 31 months. The time to detection of a first adenoma was longer in the ASA group than in the placebo group (HR for the detection of a new polyp, 0.54; 95% CI, 0.43–0.94, P = .022). Harms of treatment included upper gastrointestinal hemorrhage and hemorrhagic stroke.[60] In a study of 1,121 patients with a recent history of colorectal adenomas, after a mean duration of treatment of 33 months, the unadjusted RRs of any adenoma (as compared with the placebo group) were 0.81 in the 81-mg ASA group (95% CI, 0.69–0.96) and 0.96 in the 325-mg ASA group (95% CI, 0.81–1.13). For advanced neoplasms (adenomas measuring at least 10.0 mm in diameter or with tubulovillous or villous features, severe dysplasia, or invasive cancer), the RRs were 0.59 (95% CI, 0.38–0.92) in the 81-mg ASA group, and 0.83 (95% CI, 0.55–1.23) in the 325-mg ASA group.[61] Harms of treatment were similar in the two groups and included upper gastrointestinal bleeding and hemorrhagic stroke.

Four reports in 2007, 2010, 2011, and 2012 [57,62-64] have analyzed long-term follow-up of RCTs of daily ASA versus the control. The 2007 analysis reported on two RCTs with reliable follow-up of more than 20 years. This report found that the use of 300 mg or more of ASA per day for at least 5 years reduced the incidence of CRC after a latency of 10 years (RR at 10–19 years [0.60; 95% CI, 0.42-0.87]). The 2010 analysis analyzed long-term follow-up data from four RCTs, finding that allocation to ASA for 5 or more years reduced the 20-year incidence and mortality of proximal colon cancer (adjusted incidence HR = 0.35; 95% CI, 0.20–0.63; adjusted mortality HR = 0.24; 95% CI, 0.11–0.52) and also reduced the 20-year risk of rectal cancer (RR = 0.58; 95% CI, 0.36–0.92) but not distal colon cancer. There was no increase in benefit at doses more than 75 mg/day. The absolute 20-year risk reduction in fatal CRC was 1.76% (95% CI, 0.61–2.91).

The 2011 analysis examined data from eight RCTs, seven of which provided individual patient data and three of which provided 20-year follow-up data. In trials with allocation to ASA of at least 5 years, the 20-year HR for CRC mortality was 0.60 (95% CI, 0.45–0.81). Six RCTs, including five from the United Kingdom, were included in a meta-analysis in which patients were randomly assigned to receive either aspirin or placebo and mean scheduled duration of trial treatment was 4 years or more. Individual patient data for all in-trial cancer deaths were obtained. In the three United Kingdom trials, cancer deaths after completion of the trials were obtained via death certification and cancer registration, taking the follow-up to 20 years after randomization. Based on meta-analysis of ORs from each trial rather than on more sensitive actuarial analysis of the individual patient data, allocation to aspirin in the RCTs reduced the 20-year risk of death due to colorectal (and esophageal) cancer. ORs for maximum aspirin use were 0.55 for colorectal cancer risk (95% CI, 0.41–0.76) and 0.47 for esophageal cancer risk (95% CI, 0.27–0.81) and for any aspirin use were 0.58 for colorectal cancer risk (95% CI, 0.44–0.78) and 0.51 for esophageal cancer (95% CI, 0.31–0.83).

In a large cohort study, an association between recent daily aspirin use and lower-cancer mortality in the gastrointestinal tract (RR = 0.61; 95% CI, 0.47–0.78), liver (RR = 0.52; 95% CI, 0.30–0.93), and bladder (RR = 0.52; 95% CI, 0.28–0.97) were observed among the 100,139 analysis-eligible participants from the CPS II Nutrition Cohort established by the American Cancer Society in 1982. The analysis excluded participants who had a history of cancer in the baseline year or whose records contained incomplete information on aspirin use or smoking, and was based on follow-up questionnaires mailed to participants in 1997 (the baseline year for the analysis), 1999, 2001, and 2003. Mortality follow-up continued through Dec 31, 2008 via automated linkage to the National Death Index vital status and cause of death codes (ICD-10); death certificates were obtained for 99.3% of known deaths.[65]

The WHS, the largest randomized trial of aspirin to date (N = 39,876), found no reduction in the incidence of colon or other cancers during the 10-year active intervention. However, among women who voluntarily participated in extended follow-up (N = 33,682; 16,913 from the intervention group and 16,769 from the placebo group), there was a significant reduction in CRC incidence (HR = 0.58; 95% CI, 0.42–0.8, P < .001) with a median follow-up of 8 years. Calculated from the beginning of the intervention period through the extended follow-up (median, 18 years) an overall reduction in CRC incidence was observed for the WHS (HR = 0.80; 95% CI, 0.67–0.97; P = .021; P = .012 intervention period versus extended follow-up). During the intervention phase, women were randomly assigned to receive either an annual supply of aspirin (100 mg) or placebo, taken every other day. During the extended follow-up, the intervention was discontinued. Protocol compliance and medical incidences were tracked via identical annual questionnaires throughout the 18-year period. Medical record reviews by a panel of experts blinded to random assignment confirmed the endpoints reported. Whereas previously reported meta-analyses of randomized trials of daily aspirin use demonstrated a reduction in colon cancer incidence with extended follow-up, these findings from the WHS demonstrate a similar effect from aspirin taken every other day.[66]

Postmenopausal female hormone supplements

Several observational studies have suggested a decreased risk of colon cancer among users of postmenopausal female hormone supplements.[67-70] For rectal cancer, most studies have observed no association or a slightly elevated risk.[71-73]

The Women’s Health Initiative (WHI) trial examined, as a secondary endpoint, the effect of combined estrogen and progestin therapy and estrogen-only therapy on CRC incidence and mortality. Among women in the combined estrogen plus progestin group of the WHI, an extended follow-up (mean, 11.6 years) confirmed that fewer CRC were diagnosed in the combined hormone therapy group than in the placebo group (HR = 0.72; 95% CI, 0.56–0.94); the CRCs in women in the combined group were more likely to have lymph node involvement than the CRCs in women in the placebo group (50.5% vs. 28.6%; P < .001) and were classified at higher stages (regional and distant) (68.8% vs. 51.4%; P = .003). The number of CRC deaths in the combined group was higher than in the placebo group (37 vs. 27 deaths), but the difference was not statistically significant (HR = 1 .29; 95% CI, 0.78–2.11).[74]

The estrogen-only intervention component of the WHI was conducted among women who had a hysterectomy, with CRC incidence included as a secondary trial endpoint. CRC incidence was not decreased among women who had taken estrogens; after a median of 7.1 years of follow-up, 58 invasive cancers occurred in the estrogen arm compared with 53 invasive cancers in the placebo arm (HR = 1.12; 95% CI, 0.77–1.63). Tumor stage and grade were similar in the two groups; deaths after CRC were 34% in the hormone group compared with 30% in the placebo group (HR = 1.34; 95% CI, 0.58–3.19).[75]

Polyp removal

An analysis of data from the National Polyp Study (NPS), with external, historical controls, has commonly been cited to show a reduction of 76% to 90% in the subsequent incidence of CRC after colonoscopic polypectomy compared with three nonconcurrent, historical control groups.[76] This study may be biased in several ways that inflate the apparent efficacy of polyp removal; the main problem is that potential enrollees in the NPS were excluded if they had CRC at their baseline examination. Because no such exclusions (or baseline colonoscopy examinations) were done in the three comparison groups, persons who had CRC at baseline would be counted as having incident CRC in subsequent follow-up. Although adjustments were attempted, it is not possible to know the magnitude of the impact of this problem on the result because it is not known how long CRC may be present without causing symptoms.

A long-term follow-up study (median follow-up, 15.8 years; maximum, 23 years) of the NPS cohort suggested an approximately 53% reduction in CRC mortality due to polypectomy (not just exclusion of persons with CRC at initial exam). However, the degree of reduction must be viewed with caution because this study did not have a direct comparison group, relying mainly on comparison to expected data from the Surveillance, Epidemiology and End Results Program. Further, details are not clear about exactly what the program of colonoscopy was that may have led to decreased mortality. Patients in the NPS were assigned to colonoscopy at years 1 and 3; colonoscopy was also offered to one of the two comparison groups at year 1; all participants were offered colonoscopy at year 6. However, following year 6, the exact surveillance that patients may have undergone and how that surveillance might have been associated with decreased CRC mortality were not clear. [77]

It is expected that further follow-up in the United Kingdom Flexible Sigmoidoscopy Screening Trial will be able to provide more detail about the long-term effect of polypectomy, at least on the left side of the colon.[77]

Other evidence about the benefit of sigmoidoscopy screening (at which time both polyps and early cancer would be removed) suggests that the impact of endoscopic screening, at least on the left side of the colon, is substantial and prolonged. In an RCT, 170,000 persons were randomly assigned to one-time sigmoidoscopy versus usual care. At sigmoidoscopy, polyps were removed and cancer was detected and referred for treatment. Based on sigmoidoscopy findings, persons were considered to have low risk if they had normal exams or only one or two small (<1 cm) tubular adenomas; such persons were not referred either for colonoscopy workup, or for colonoscopic surveillance. In a follow-up of 10 years, the left-sided CRC incidence in the low-risk group (about 95% of attendees were low risk) was 0.02% to 0.04% per year—a very low risk of CRC compared with average risk. The cause of reduced risk—whether due to detection and removal of large polyps or small ones, or selection of individuals at lower risk—is yet unclear.[78] The natural history of large polyps is not well known, but some evidence suggests that such lesions become clinical CRC at a rate of approximately 1% per year.[79] As a result of the strong data about the impact of endoscopy on the left colon, evidence from multiple studies has raised questions about the ability of endoscopy to reduce CRC mortality in the right colon.[80-82] Thus, it is unclear what the overall impact of endoscopy (e.g., colonoscopy screening) is, and whether there may be a large difference in impact on the left side of the colon compared with the right side.[80]

Other studies suggest that the polyps with the greatest potential to progress to CRC are larger polyps (i.e., >1.0 cm), which include most of those with villous or high-grade histologic features. Retrospective cohort studies also show the harms associated with polypectomy, including bleeding.[83,84]

Diet modification

Dietary fat and meat intake

Colon cancer rates are high in populations with high total fat intakes and are lower in those consuming less fat.[85] On average, fat comprises 40% to 45% of total caloric intake in high-incidence Western countries; in low-risk populations, fat accounts for only 10% of dietary calories.[86] In laboratory studies, a high-fat intake increases the incidence of induced colon tumors in experimental animals.[87,88] Several case-control studies have explored the association of colon cancer risk with meat or fat consumption as well as protein and energy intake.[10,89] Although positive associations with meat consumption or with fat intake have been found frequently, the results have not always achieved statistical significance.[90] A number of prospective cohort studies have been conducted in the United States and abroad. In Japan, an increased risk of colon cancer with increased frequency of meat consumption was observed in the group with infrequent vegetable consumption among a group of 265,000 men and women.[91] In Norway, an increased risk for processed meat only was found,[92] a finding that was confirmed in the Netherlands.[93] A clearly defined gradient in the risk for frequency of meat and poultry consumption was not observed in a population of Seventh Day Adventists that included a large proportion of vegetarians.[94] A prospective study among female nurses showed an increased risk of colon cancer associated with red meat consumption (beef, pork, lamb, and processed meat) and also with the intake of saturated and monounsaturated fat, predominantly derived from animals.[95] In two other large prospective studies, the CPS II and the Iowa Women’s Health Study (IWHS), no increase in the risk of colon cancer was seen with meat or fat consumption.[96,97] In a prospective cohort study of a low-risk population of non-Hispanic white members of the Adventist Health Study, a positive association between meat (both red and white) intake and colon cancer was observed (RR for ≥1 time per week vs. no meat intake = 1.85, 95% CI, 1.19–2.87, P for trend = .01).[98] It has been hypothesized that the heterocyclic amines (HCAs) formed when meat and fish are cooked at high temperatures may contribute to the increased risk of CRCs associated with meat consumption that has been observed in epidemiologic studies. A population-based case-control study in Sweden, however, found no evidence of increased risk associated with total HCA intake; for colon cancer the RR was 0.6 (95% CI, 0.4–1.0), and for rectal cancer it was 0.7 (95% CI, 0.4–1.1).[99,100]

A randomized controlled dietary modification study was undertaken among 48,835 postmenopausal women aged 50 to 79 years who were also enrolled in the WHI. The intervention promoted a goal of reducing total fat intake by 20%, while increasing daily intake of vegetables, fruits, and grains. The intervention group accomplished a reduction of fat intake of approximately 10% more than did the comparison group during the 8.1 years of follow-up. There was no evidence of reduction in invasive CRCs between the intervention and comparison groups with a HR of 1.08 (95% CI, 0.90–1.29).[101] Likewise, there was no benefit of the low-fat diet on all-cancer mortality, overall mortality, or cardiovascular disease.[102]

Explanations for the conflicting results regarding whether dietary fat or meat intake affects the risk of CRC [93] include:

  • Validity of dietary questionnaires used.
  • Differences in the average age of the population studied.
  • Variations in methods of meat preparation (in some instances, mutagenic and carcinogenic HCAs could have been released at high temperatures).[103]
  • Variability in the consumption of other foods such as vegetables.[104]

In addition, some epidemiological studies have reported lower incidence rates of colon cancer in populations with high intakes of both fat and fiber, compared with populations with high levels of fat but low levels of fiber consumption.[105] Although far from clear-cut, the available evidence suggests CRC risk is possibly associated with some interaction of dietary fat, protein, and caloric intake.

Six case-control studies and two cohort studies have explored potential dietary risk factors for colorectal adenomas.[20,106] Three of the eight studies found that higher fat consumption was associated with increased risk. High fat intake has been found to increase the risk of adenoma recurrence following polypectomy.[107] In a multicenter RCT, a diet low in fat (20% of total calories) and high in fiber, fruits, and vegetables did not reduce the risk of recurrence of colorectal adenomas.[108]

Thus, the evidence is inadequate to determine whether reducing dietary fat and meat would reduce CRC incidence.

Bile acids

A central effect of bile acids in the etiology and pathogenesis of CRC has been claimed.[109] An increased bile acid concentration in the intestinal tract accompanies a high-fat diet because bile acids are released from the gallbladder after fat ingestion. The concentration of bile acids in the colon is heavily influenced by the amount and type of fat in the diet.[110] The potential mechanism of action of bile salts in colorectal carcinogenesis is unknown, although it has been suggested that it is mediated by diacylglycerol.[111] The conversion of dietary phospholipids to diacylglycerol by intestinal bacteria is enhanced by a high-fat diet. It is proposed that diacylglycerol enters the cell directly, stimulating protein kinase C, which is involved in intracellular signal transduction. There is no high-quality evidence from either observational studies or RCTs to substantiate this claim.

Dietary fiber, vegetables, and fruit

The evidence on whether dietary fiber exerts a protective role in reducing the incidence of CRC is mixed. Most animal and epidemiologic studies show a protective effect of dietary fiber on colon carcinogenesis.[51] The term fiber is used to describe a complex mixture of compounds, including insoluble fiber (typified by wheat bran and cellulose) and soluble fiber (usually dried beans). Ingestion of fiber could modify carcinogenesis in the large bowel by a number of potential mechanisms.[112-114] These mechanisms include binding to bile acids, increasing fecal water and possibly diluting carcinogens, and decreasing transit time (not an obvious factor). Fiber may act as a substrate for bacterial fermentation with a resultant increase in bacterial mass and the production of short-chain fatty acids, typified by butyrate.[114] Butyrate has been shown to have anticarcinogenic effects in vitro and is regarded as an important fuel for the colonic epithelium.[115,116] A meta-analysis of 13 case-control studies from nine countries concluded that intake of fiber-rich foods is inversely related to cancers of both the colon and rectum.[117] The analysis did not include fiber supplements. The inverse association with fiber was observed in 12 of the 13 studies and was similar in magnitude for left-sided and right-sided colon and rectal cancers, in men and women, and in different age groups. It has been suggested that the inverse association with fiber may be reflective of some other closely associated dietary constituents, such as the anticarcinogens found in vegetables, fruits, legumes, nuts, and grains.[10,117] These substances include phenolic compounds, sulfur-containing compounds, and flavones.[118,119] In a prospective cohort study of a low-risk population, an inverse association was found with legume intake and the risk of CRC (RR for >2 times/week vs. 1 time/week = 0.53 [95% CI, 0.33–0.86; P for trend = .03]).[98]

Other studies have corroborated the effects of dietary fiber. One study used a supplement of 10 g/day of wheat bran, cellulose, and oat bran and found a decreased mutagenic activity of fecal contents in those receiving wheat bran and cellulose supplementation, although no measurable inhibition was observed during oat bran supplementation.[8] Fecal-total and secondary bile acid excretion increased during oat fiber supplementation.

Despite the evidence from case-control studies of a protective effect, results from the large prospective Nurses’ Health Study (NHS) found no difference in the risk of CRC between women in the highest quintile group compared with the lowest quintile group with respect to dietary fiber, after adjusting for age, known risk factors, and total energy intake (RR = 0.95; 95% CI, 0.73–1.25).[120]

Many epidemiologic studies have examined the relationship between fruit and vegetable intake and the incidence of colon and/or rectal cancer,[121] with considerable variation in findings. Perhaps the most definitive analysis to date is a prospective study that examined dietary intake data based on food frequency questionnaires from 88,764 women in the NHS and 47,325 men in the Health Professionals Follow-up Study.[122] The study included a total of 1,743,645 person-years of follow-up, 937 cases of colon cancer, and 244 cases of rectal cancer. On the basis of analyses adjusted for numerous covariates, the authors found no association in women or men between overall fruit and vegetable consumption and the risk of colon or rectal cancer. Associations were not observed when the data were examined for subgroups of fruits or vegetables (with the exception of legumes, which were associated with an increased risk of colon cancer in women) or individual fruits or vegetables (with the exception of prunes, which were associated with an increased risk of colon cancer in men). Results did not change when data were examined by vitamin use status, smoking status, or family history of CRC, nor were elevated risks seen when individuals with very low levels of fruit and vegetable consumption were compared with those having the highest levels. For women and men combined, the covariate-adjusted RR of colon cancer associated with one additional serving of fruits and vegetables per day was 1.02 (95% CI, 0.98–1.05); the comparable RR for rectal cancer was 1.02 (95% CI, 0.95–1.09).

In a population-based prospective cohort study of 61,463 women in Sweden, individuals who consumed very low amounts of fruits and vegetables (<1.5 servings of fruit and vegetables/day) had a RR for developing CRC of 1.65 (95% CI, 1.23–2.2; P trend = .001) as compared with those individuals who consumed more than 2.5 servings. There was little evidence, however, of a benefit for higher as compared with moderate consumption (more than vs. fewer than 3.5 servings). Limitations of this study are that dietary intake during the study period was not reassessed over time, and the influence of physical activity could not be accurately determined. In addition, the conclusion about very low amounts of intake of fruits and vegetables is based on a retrospective subdivision of the lowest quartile of consumption, and its strength has not been adjusted for other potential confounding factors.[123]

Six case-control studies and three cohort studies have explored potential dietary risk factors for colorectal adenomas.[20,106,120] Four of the nine found an association of fiber, carbohydrates, and/or vegetables with reduced risk. In one study, cases with moderate or severe dysplasia had a significantly lower intake of cruciferous vegetables than those with mild dysplasia. No significant effect of dietary fiber on colorectal adenoma was found in the large cohort study of U.S. nurses.[120]

High-fiber cereal supplements during a 3-year period did not result in a decrease in adenoma recurrence in a RCT of 1,303 individuals.[124] In a multicenter RCT, a diet low in fat (20% of total calories) and high in fiber (18 g of dietary fiber/1,000 kcal) and fruits and vegetables (3.5 servings per 1,000 kcal) was not associated with a reduction in the risk of recurrence of colorectal adenomas.[108]

Vitamins

In a prospective cohort study of 35,215 women in the IWHS, an inverse association between the risk of colon cancer and vitamin E intake was found; the RR for the highest compared with the lowest quartile was 0.3 (95% CI, 0.19–0.54).[125] The WHS, however, showed no relationship between CRC in women and the use of 600 IU of vitamin E every other day.[126] In a meta-analysis of 14 randomized trials of supplemental antioxidant vitamins encompassing 170,025 individuals, no evidence of prevention of colorectal adenomas or cancer or other gastrointestinal tumors was found.[127] A systematic review of published observational studies that provide sufficient data to calculate the dose-response relationship of serum 25-hydroxyvitamin D or oral intake of vitamin D with the risk of CRC was conducted. The results suggested that a daily intake of 1,000 IU of vitamin D—half the safe upper limit for intake established by the National Academy of Sciences—and a concentration of serum 25-hydroxyvitamin D of 33 ng/mL were each associated with a 50% lower risk of CRC.[128] In a population-based case-control study, an inverse relationship between vitamin D intake and risk of CRC was found.[129]

A prospective cohort study observed that higher energy-adjusted folate intake in the form of multivitamins containing folic acid was related to a lower risk for colon cancer (RR = 0.69; 95% CI, 0.52–0.93) for intake of more than 400 µg/day compared with intake of 200 µg/day or less after controlling for age, family history of CRC, ASA use, smoking, body mass, physical activity, and intakes of red meat, alcohol, methionine, and fiber.[130] In a double-blind, placebo-controlled, two-factor, phase III RCT (ASA/Folate Polyp Prevention Study) involving 1,021 men and women with a recent history of colorectal adenoma, folic acid (1 mg/day) was associated with higher risks of developing at least one advanced adenoma (11.6% for folic acid [n = 35]); 6.9% for placebo [n = 21]; unadjusted RR = 1.67; 95% CI, 1.00–2.80; P = .05). Folic acid ingestion was associated with higher risks of having three or more adenomas and of non-CRCs. There was no effect modification by sex, age, smoking, alcohol use, BMI, baseline plasma folate, or ASA use. There was no apparent effect on overall adenoma incidence (44.1% for folic acid [n = 221]); 42.4% for placebo [n = 206]; unadjusted use ratio 1.04; 95% CI, 0.9–1.20; P = .58).[131]

Calcium

It has been hypothesized that orally ingested calcium lowers colon cancer risk by binding bile acids and fatty acids, thereby reducing exposure to toxic intraluminal compounds.[132] Indirect effects on bile acid metabolism and a direct effect on colonic epithelial cells are also possible.

Several [133-136] but not all [106,137] epidemiologic studies have observed an inverse relationship between calcium intake and cancer risk. Interpretation of these studies can be quite complex. In Utah, an inverse relationship between colon cancer and calcium was observed in a study that compared members of the Church of Jesus Christ of Latter-Day Saints (Mormons) and Seventh Day Adventists with a group from the U.S. population at large. Both study groups have higher calcium intakes, mainly milk and dairy products, than the national average. Unlike the Seventh Day Adventists, however, the Mormon group had a consumption of meats and fat similar to that of the general population.

Experimental studies in rodents [138] and some but not all human studies [139-142] have described a decrease in colonic epithelial cell proliferation after the administration of calcium citrate. Human studies using the labeling index are dependent on a complex methodology.[143] A randomized placebo-controlled trial tested the effect of calcium supplementation (3 g calcium carbonate daily [1,200 mg elemental calcium]) on the risk of recurrent adenoma.[144] The primary endpoint was the proportion of patients (72% of whom were male) in whom at least one adenoma was detected following a first and/or second follow-up endoscopy. A modest decrease in risk was found for both developing at least one recurrent adenoma (adjusted risk ratio [ARR] = 0.81; 95% CI, 0.67–0.99) and in the average number of adenomas (ARR = 0.76; 95% CI, 0.60–0.96). The investigators found the effect of calcium was similar across age, sex, and baseline dietary intake categories of calcium, fat, or fiber. The study was limited to individuals with a recent history of colorectal adenomas and could not determine the effect of calcium on risk of the first adenoma, nor was it large enough or of sufficient duration to examine the risk of invasive CRC. After calcium supplementation is stopped, the lower risk may persist up to 5 years.[145] The results of other ongoing adenoma recurrence studies are awaited with interest. It is important to note that the dose of calcium salt administered may be important; the usual daily doses in trials have ranged from 1,250 to 2,000 mg of calcium.

In a randomized, double-blind, placebo-controlled trial involving 36,282 postmenopausal women, the administration of 500 mg of elemental calcium and 200 IU of vitamin D3 twice daily for an average of 7.0 years was not associated with a reduction in invasive CRC (HR = 1.08; 95% CI, 0.86–1.34; P = .051).[146] The relatively short duration of follow-up, considering the latency period of CRC of 10 to 15 years and suboptimal doses of calcium and vitamin D, may account for the negative effects of this trial, though other factors may also be responsible.[147]

Use of statins

Overall, evidence indicates that statin use neither increases nor decreases the incidence or mortality of CRC. Although some case-control studies have shown a reduction in risk, neither a large cohort study [148] nor a meta-analysis of four RCTs [149] found any effect of statin use.

References
  1. Shike M, Winawer SJ, Greenwald PH, et al.: Primary prevention of colorectal cancer. The WHO Collaborating Centre for the Prevention of Colorectal Cancer. Bull World Health Organ 68 (3): 377-85, 1990.  [PUBMED Abstract]

  2. American Cancer Society: Cancer Facts and Figures 2014. Atlanta, Ga: American Cancer Society, 2014. Available online. Last accessed May 21, 2014. 

  3. Howlader N, Noone AM, Krapcho M, et al., eds.: SEER Cancer Statistics Review, 1975-2008. Bethesda, Md: National Cancer Institute, 2011. Also available online. Last accessed July 2, 2014. 

  4. Moertel CG, Fleming TR, Macdonald JS, et al.: Levamisole and fluorouracil for adjuvant therapy of resected colon carcinoma. N Engl J Med 322 (6): 352-8, 1990.  [PUBMED Abstract]

  5. Krook JE, Moertel CG, Gunderson LL, et al.: Effective surgical adjuvant therapy for high-risk rectal carcinoma. N Engl J Med 324 (11): 709-15, 1991.  [PUBMED Abstract]

  6. Willett W: The search for the causes of breast and colon cancer. Nature 338 (6214): 389-94, 1989.  [PUBMED Abstract]

  7. Fearon ER, Vogelstein B: A genetic model for colorectal tumorigenesis. Cell 61 (5): 759-67, 1990.  [PUBMED Abstract]

  8. Reddy B, Engle A, Katsifis S, et al.: Biochemical epidemiology of colon cancer: effect of types of dietary fiber on fecal mutagens, acid, and neutral sterols in healthy subjects. Cancer Res 49 (16): 4629-35, 1989.  [PUBMED Abstract]

  9. Reddy BS, Tanaka T, Simi B: Effect of different levels of dietary trans fat or corn oil on azoxymethane-induced colon carcinogenesis in F344 rats. J Natl Cancer Inst 75 (4): 791-8, 1985.  [PUBMED Abstract]

  10. Potter JD: Reconciling the epidemiology, physiology, and molecular biology of colon cancer. JAMA 268 (12): 1573-7, 1992 Sep 23-30.  [PUBMED Abstract]

  11. Wynder EL, Reddy BS: Dietary fat and fiber and colon cancer. Semin Oncol 10 (3): 264-72, 1983.  [PUBMED Abstract]

  12. Chen CD, Yen MF, Wang WM, et al.: A case-cohort study for the disease natural history of adenoma-carcinoma and de novo carcinoma and surveillance of colon and rectum after polypectomy: implication for efficacy of colonoscopy. Br J Cancer 88 (12): 1866-73, 2003.  [PUBMED Abstract]

  13. Cho E, Smith-Warner SA, Ritz J, et al.: Alcohol intake and colorectal cancer: a pooled analysis of 8 cohort studies. Ann Intern Med 140 (8): 603-13, 2004.  [PUBMED Abstract]

  14. Newcomb PA, Storer BE, Marcus PM: Cancer of the large bowel in women in relation to alcohol consumption: a case-control study in Wisconsin (United States). Cancer Causes Control 4 (5): 405-11, 1993.  [PUBMED Abstract]

  15. Meyer F, White E: Alcohol and nutrients in relation to colon cancer in middle-aged adults. Am J Epidemiol 138 (4): 225-36, 1993.  [PUBMED Abstract]

  16. Longnecker MP, Orza MJ, Adams ME, et al.: A meta-analysis of alcoholic beverage consumption in relation to risk of colorectal cancer. Cancer Causes Control 1 (1): 59-68, 1990.  [PUBMED Abstract]

  17. Boutron MC, Faivre J: Diet and the adenoma-carcinoma sequence. Eur J Cancer Prev 2 (Suppl 2): 95-8, 1993.  [PUBMED Abstract]

  18. Boutron MC, Faivre J: Alcohol, tobacco and the adenoma-carcinoma sequence: a case-control study in Burgundy, France. [Abstract] Gastroenterology 104 (Suppl 4): A-390, 1993. 

  19. Thun MJ, Peto R, Lopez AD, et al.: Alcohol consumption and mortality among middle-aged and elderly U.S. adults. N Engl J Med 337 (24): 1705-14, 1997.  [PUBMED Abstract]

  20. Neugut AI, Jacobson JS, DeVivo I: Epidemiology of colorectal adenomatous polyps. Cancer Epidemiol Biomarkers Prev 2 (2): 159-76, 1993 Mar-Apr.  [PUBMED Abstract]

  21. Giovannucci E, Colditz GA, Stampfer MJ, et al.: A prospective study of cigarette smoking and risk of colorectal adenoma and colorectal cancer in U.S. women. J Natl Cancer Inst 86 (3): 192-9, 1994.  [PUBMED Abstract]

  22. Giovannucci E, Rimm EB, Stampfer MJ, et al.: A prospective study of cigarette smoking and risk of colorectal adenoma and colorectal cancer in U.S. men. J Natl Cancer Inst 86 (3): 183-91, 1994.  [PUBMED Abstract]

  23. Chao A, Thun MJ, Jacobs EJ, et al.: Cigarette smoking and colorectal cancer mortality in the cancer prevention study II. J Natl Cancer Inst 92 (23): 1888-96, 2000.  [PUBMED Abstract]

  24. Terry P, Ekbom A, Lichtenstein P, et al.: Long-term tobacco smoking and colorectal cancer in a prospective cohort study. Int J Cancer 91 (4): 585-7, 2001.  [PUBMED Abstract]

  25. Slattery ML, Potter JD, Friedman GD, et al.: Tobacco use and colon cancer. Int J Cancer 70 (3): 259-64, 1997.  [PUBMED Abstract]

  26. Knekt P, Hakama M, Järvinen R, et al.: Smoking and risk of colorectal cancer. Br J Cancer 78 (1): 136-9, 1998.  [PUBMED Abstract]

  27. Baron JA, Sandler RS, Haile RW, et al.: Folate intake, alcohol consumption, cigarette smoking, and risk of colorectal adenomas. J Natl Cancer Inst 90 (1): 57-62, 1998.  [PUBMED Abstract]

  28. Botteri E, Iodice S, Bagnardi V, et al.: Smoking and colorectal cancer: a meta-analysis. JAMA 300 (23): 2765-78, 2008.  [PUBMED Abstract]

  29. Martínez ME, Giovannucci E, Spiegelman D, et al.: Leisure-time physical activity, body size, and colon cancer in women. Nurses' Health Study Research Group. J Natl Cancer Inst 89 (13): 948-55, 1997.  [PUBMED Abstract]

  30. Giovannucci E, Ascherio A, Rimm EB, et al.: Physical activity, obesity, and risk for colon cancer and adenoma in men. Ann Intern Med 122 (5): 327-34, 1995.  [PUBMED Abstract]

  31. Calle EE, Rodriguez C, Walker-Thurmond K, et al.: Overweight, obesity, and mortality from cancer in a prospectively studied cohort of U.S. adults. N Engl J Med 348 (17): 1625-38, 2003.  [PUBMED Abstract]

  32. White E, Jacobs EJ, Daling JR: Physical activity in relation to colon cancer in middle-aged men and women. Am J Epidemiol 144 (1): 42-50, 1996.  [PUBMED Abstract]

  33. Slattery ML, Schumacher MC, Smith KR, et al.: Physical activity, diet, and risk of colon cancer in Utah. Am J Epidemiol 128 (5): 989-99, 1988.  [PUBMED Abstract]

  34. Kune GA, Kune S, Watson LF: Body weight and physical activity as predictors of colorectal cancer risk. Nutr Cancer 13 (1-2): 9-17, 1990.  [PUBMED Abstract]

  35. Friedenreich CM: Physical activity and cancer prevention: from observational to intervention research. Cancer Epidemiol Biomarkers Prev 10 (4): 287-301, 2001.  [PUBMED Abstract]

  36. Wolin KY, Yan Y, Colditz GA, et al.: Physical activity and colon cancer prevention: a meta-analysis. Br J Cancer 100 (4): 611-6, 2009.  [PUBMED Abstract]

  37. Ruder EH, Laiyemo AO, Graubard BI, et al.: Non-steroidal anti-inflammatory drugs and colorectal cancer risk in a large, prospective cohort. Am J Gastroenterol 106 (7): 1340-50, 2011.  [PUBMED Abstract]

  38. Hinz B, Brune K: Cyclooxygenase-2--10 years later. J Pharmacol Exp Ther 300 (2): 367-75, 2002.  [PUBMED Abstract]

  39. Kerr DJ, Dunn JA, Langman MJ, et al.: Rofecoxib and cardiovascular adverse events in adjuvant treatment of colorectal cancer. N Engl J Med 357 (4): 360-9, 2007.  [PUBMED Abstract]

  40. Graham DJ, Campen D, Hui R, et al.: Risk of acute myocardial infarction and sudden cardiac death in patients treated with cyclo-oxygenase 2 selective and non-selective non-steroidal anti-inflammatory drugs: nested case-control study. Lancet 365 (9458): 475-81, 2005.  [PUBMED Abstract]

  41. Bresalier RS, Sandler RS, Quan H, et al.: Cardiovascular events associated with rofecoxib in a colorectal adenoma chemoprevention trial. N Engl J Med 352 (11): 1092-102, 2005.  [PUBMED Abstract]

  42. Solomon SD, McMurray JJ, Pfeffer MA, et al.: Cardiovascular risk associated with celecoxib in a clinical trial for colorectal adenoma prevention. N Engl J Med 352 (11): 1071-80, 2005.  [PUBMED Abstract]

  43. Trelle S, Reichenbach S, Wandel S, et al.: Cardiovascular safety of non-steroidal anti-inflammatory drugs: network meta-analysis. BMJ 342: c7086, 2011.  [PUBMED Abstract]

  44. Chan AT, Giovannucci EL, Meyerhardt JA, et al.: Long-term use of aspirin and nonsteroidal anti-inflammatory drugs and risk of colorectal cancer. JAMA 294 (8): 914-23, 2005.  [PUBMED Abstract]

  45. Arber N, Eagle CJ, Spicak J, et al.: Celecoxib for the prevention of colorectal adenomatous polyps. N Engl J Med 355 (9): 885-95, 2006.  [PUBMED Abstract]

  46. Smalley W, Ray WA, Daugherty J, et al.: Use of nonsteroidal anti-inflammatory drugs and incidence of colorectal cancer: a population-based study. Arch Intern Med 159 (2): 161-6, 1999.  [PUBMED Abstract]

  47. Labayle D, Fischer D, Vielh P, et al.: Sulindac causes regression of rectal polyps in familial adenomatous polyposis. Gastroenterology 101 (3): 635-9, 1991.  [PUBMED Abstract]

  48. Giardiello FM, Hamilton SR, Krush AJ, et al.: Treatment of colonic and rectal adenomas with sulindac in familial adenomatous polyposis. N Engl J Med 328 (18): 1313-6, 1993.  [PUBMED Abstract]

  49. Steinbach G, Lynch PM, Phillips RK, et al.: The effect of celecoxib, a cyclooxygenase-2 inhibitor, in familial adenomatous polyposis. N Engl J Med 342 (26): 1946-52, 2000.  [PUBMED Abstract]

  50. Earnest DL, Hixson LJ, Fennerty MB, et al.: Inhibition of prostaglandin synthesis: potential for chemoprevention of human colon cancer. Cancer Bull 43(6): 561-568, 1991. 

  51. Vargas PA, Alberts DS: Colon cancer: the quest for prevention. Oncology (Huntingt) 7 (11 Suppl): 33-40, 1993. 

  52. Imperiale TF: Aspirin and the prevention of colorectal cancer. N Engl J Med 348 (10): 879-80, 2003.  [PUBMED Abstract]

  53. Thun MJ, Namboodiri MM, Heath CW Jr: Aspirin use and reduced risk of fatal colon cancer. N Engl J Med 325 (23): 1593-6, 1991.  [PUBMED Abstract]

  54. Thun MJ, Namboodiri MM, Calle EE, et al.: Aspirin use and risk of fatal cancer. Cancer Res 53 (6): 1322-7, 1993.  [PUBMED Abstract]

  55. Giovannucci E, Rimm EB, Stampfer MJ, et al.: Aspirin use and the risk for colorectal cancer and adenoma in male health professionals. Ann Intern Med 121 (4): 241-6, 1994.  [PUBMED Abstract]

  56. 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 294 (1): 47-55, 2005.  [PUBMED Abstract]

  57. Flossmann E, Rothwell PM; British 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 369 (9573): 1603-13, 2007.  [PUBMED Abstract]

  58. Gann PH, Manson JE, Glynn RJ, et al.: Low-dose aspirin and incidence of colorectal tumors in a randomized trial. J Natl Cancer Inst 85 (15): 1220-4, 1993.  [PUBMED Abstract]

  59. 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 128 (9): 713-20, 1998.  [PUBMED Abstract]

  60. 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 348 (10): 883-90, 2003.  [PUBMED Abstract]

  61. Baron JA, Cole BF, Sandler RS, et al.: A randomized trial of aspirin to prevent colorectal adenomas. N Engl J Med 348 (10): 891-9, 2003.  [PUBMED Abstract]

  62. 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 376 (9754): 1741-50, 2010.  [PUBMED Abstract]

  63. Rothwell PM, Fowkes FG, Belch JF, et al.: Effect of daily aspirin on long-term risk of death due to cancer: analysis of individual patient data from randomised trials. Lancet 377 (9759): 31-41, 2011.  [PUBMED Abstract]

  64. Algra AM, Rothwell PM: Effects of regular aspirin on long-term cancer incidence and metastasis: a systematic comparison of evidence from observational studies versus randomised trials. Lancet Oncol 13 (5): 518-27, 2012.  [PUBMED Abstract]

  65. Jacobs EJ, Newton CC, Gapstur SM, et al.: Daily aspirin use and cancer mortality in a large US cohort. J Natl Cancer Inst 104 (16): 1208-17, 2012.  [PUBMED Abstract]

  66. 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 159 (2): 77-85, 2013.  [PUBMED Abstract]

  67. Calle EE, Miracle-McMahill HL, Thun MJ, et al.: Estrogen replacement therapy and risk of fatal colon cancer in a prospective cohort of postmenopausal women. J Natl Cancer Inst 87 (7): 517-23, 1995.  [PUBMED Abstract]

  68. Newcomb PA, Storer BE: Postmenopausal hormone use and risk of large-bowel cancer. J Natl Cancer Inst 87 (14): 1067-71, 1995.  [PUBMED Abstract]

  69. Grodstein F, Newcomb PA, Stampfer MJ: Postmenopausal hormone therapy and the risk of colorectal cancer: a review and meta-analysis. Am J Med 106 (5): 574-82, 1999.  [PUBMED Abstract]

  70. Terry MB, Neugut AI, Bostick RM, et al.: Risk factors for advanced colorectal adenomas: a pooled analysis. Cancer Epidemiol Biomarkers Prev 11 (7): 622-9, 2002.  [PUBMED Abstract]

  71. Risch HA, Howe GR: Menopausal hormone use and colorectal cancer in Saskatchewan: a record linkage cohort study. Cancer Epidemiol Biomarkers Prev 4 (1): 21-8, 1995 Jan-Feb.  [PUBMED Abstract]

  72. Gerhardsson de Verdier M, London S: Reproductive factors, exogenous female hormones, and colorectal cancer by subsite. Cancer Causes Control 3 (4): 355-60, 1992.  [PUBMED Abstract]

  73. Prihartono N, Palmer JR, Louik C, et al.: A case-control study of use of postmenopausal female hormone supplements in relation to the risk of large bowel cancer. Cancer Epidemiol Biomarkers Prev 9 (4): 443-7, 2000.  [PUBMED Abstract]

  74. Simon MS, Chlebowski RT, Wactawski-Wende J, et al.: Estrogen plus progestin and colorectal cancer incidence and mortality. J Clin Oncol 30 (32): 3983-90, 2012.  [PUBMED Abstract]

  75. Ritenbaugh C, Stanford JL, Wu L, et al.: Conjugated equine estrogens and colorectal cancer incidence and survival: the Women's Health Initiative randomized clinical trial. Cancer Epidemiol Biomarkers Prev 17 (10): 2609-18, 2008.  [PUBMED Abstract]

  76. Winawer SJ, Zauber AG, Ho MN, et al.: Prevention of colorectal cancer by colonoscopic polypectomy. The National Polyp Study Workgroup. N Engl J Med 329 (27): 1977-81, 1993.  [PUBMED Abstract]

  77. Zauber AG, Winawer SJ, O'Brien MJ, et al.: Colonoscopic polypectomy and long-term prevention of colorectal-cancer deaths. N Engl J Med 366 (8): 687-96, 2012.  [PUBMED Abstract]

  78. Atkin WS, Edwards R, Kralj-Hans I, et al.: Once-only flexible sigmoidoscopy screening in prevention of colorectal cancer: a multicentre randomised controlled trial. Lancet 375 (9726): 1624-33, 2010.  [PUBMED Abstract]

  79. Stryker SJ, Wolff BG, Culp CE, et al.: Natural history of untreated colonic polyps. Gastroenterology 93 (5): 1009-13, 1987.  [PUBMED Abstract]

  80. Brenner H, Chang-Claude J, Seiler CM, et al.: Protection from colorectal cancer after colonoscopy: a population-based, case-control study. Ann Intern Med 154 (1): 22-30, 2011.  [PUBMED Abstract]

  81. Baxter NN, Goldwasser MA, Paszat LF, et al.: Association of colonoscopy and death from colorectal cancer. Ann Intern Med 150 (1): 1-8, 2009.  [PUBMED Abstract]

  82. Brenner H, Hoffmeister M, Arndt V, et al.: Protection from right- and left-sided colorectal neoplasms after colonoscopy: population-based study. J Natl Cancer Inst 102 (2): 89-95, 2010.  [PUBMED Abstract]

  83. Levin TR, Zhao W, Conell C, et al.: Complications of colonoscopy in an integrated health care delivery system. Ann Intern Med 145 (12): 880-6, 2006.  [PUBMED Abstract]

  84. Warren JL, Klabunde CN, Mariotto AB, et al.: Adverse events after outpatient colonoscopy in the Medicare population. Ann Intern Med 150 (12): 849-57, W152, 2009.  [PUBMED Abstract]

  85. Rose DP, Boyar AP, Wynder EL: International comparisons of mortality rates for cancer of the breast, ovary, prostate, and colon, and per capita food consumption. Cancer 58 (11): 2363-71, 1986.  [PUBMED Abstract]

  86. Reddy BS: Dietary fat and its relationship to large bowel cancer. Cancer Res 41 (9 Pt 2): 3700-5, 1981.  [PUBMED Abstract]

  87. Reddy BS, Narisawa T, Vukusich D, et al.: Effect of quality and quantity of dietary fat and dimethylhydrazine in colon carcinogenesis in rats. Proc Soc Exp Biol Med 151 (2): 237-9, 1976.  [PUBMED Abstract]

  88. Nauss KM, Locniskar M, Newberne PM: Effect of alterations in the quality and quantity of dietary fat on 1,2-dimethylhydrazine-induced colon tumorigenesis in rats. Cancer Res 43 (9): 4083-90, 1983.  [PUBMED Abstract]

  89. Potter JD, McMichael AJ: Diet and cancer of the colon and rectum: a case-control study. J Natl Cancer Inst 76 (4): 557-69, 1986.  [PUBMED Abstract]

  90. Bingham SA: Diet and large bowel cancer. J R Soc Med 83 (7): 420-2, 1990.  [PUBMED Abstract]

  91. Hirayama T, Tannenbaum SR, Reddy BS, et al.: A large-scale cohort study on the relationship between diet and selected cancers of the digestive organs. In: Bruce WR, Correa P, Lipkin M, et al., eds.: Gastrointestinal cancer: endogenous factors. [Cold Spring Harbor, NY]: Cold Spring Harbor Laboratory, 1981, Branbury Report 7, 409-429. 

  92. Bjelke E: Epidemiology of colorectal cancer, with emphasis on diet. Int Congr Ser 484: 158-174, 1980. 

  93. Goldbohm RA, van den Brandt PA, van 't Veer P, et al.: A prospective cohort study on the relation between meat consumption and the risk of colon cancer. Cancer Res 54 (3): 718-23, 1994.  [PUBMED Abstract]

  94. Phillips RL, Snowdon DA: Dietary relationships with fatal colorectal cancer among Seventh-Day Adventists. J Natl Cancer Inst 74 (2): 307-17, 1985.  [PUBMED Abstract]

  95. Willett WC, Stampfer MJ, Colditz GA, et al.: Relation of meat, fat, and fiber intake to the risk of colon cancer in a prospective study among women. N Engl J Med 323 (24): 1664-72, 1990.  [PUBMED Abstract]

  96. Thun MJ, Calle EE, Namboodiri MM, et al.: Risk factors for fatal colon cancer in a large prospective study. J Natl Cancer Inst 84 (19): 1491-500, 1992.  [PUBMED Abstract]

  97. Bostick RM, Potter JD, Sellers TA, et al.: Relation of calcium, vitamin D, and dairy food intake to incidence of colon cancer among older women. The Iowa Women's Health Study. Am J Epidemiol 137 (12): 1302-17, 1993.  [PUBMED Abstract]

  98. Singh PN, Fraser GE: Dietary risk factors for colon cancer in a low-risk population. Am J Epidemiol 148 (8): 761-74, 1998.  [PUBMED Abstract]

  99. Augustsson K, Skog K, Jägerstad M, et al.: Dietary heterocyclic amines and cancer of the colon, rectum, bladder, and kidney: a population-based study. Lancet 353 (9154): 703-7, 1999.  [PUBMED Abstract]

  100. Forman D: Meat and cancer: a relation in search of a mechanism. Lancet 353 (9154): 686-7, 1999.  [PUBMED Abstract]

  101. Beresford SA, Johnson KC, Ritenbaugh C, et al.: Low-fat dietary pattern and risk of colorectal cancer: the Women's Health Initiative Randomized Controlled Dietary Modification Trial. JAMA 295 (6): 643-54, 2006.  [PUBMED Abstract]

  102. Howard BV, Van Horn L, Hsia J, et al.: Low-fat dietary pattern and risk of cardiovascular disease: the Women's Health Initiative Randomized Controlled Dietary Modification Trial. JAMA 295 (6): 655-66, 2006.  [PUBMED Abstract]

  103. Sugimura T: Carcinogenicity of mutagenic heterocyclic amines formed during the cooking process. Mutat Res 150 (1-2): 33-41, 1985 Jun-Jul.  [PUBMED Abstract]

  104. Lee HP, Gourley L, Duffy SW, et al.: Colorectal cancer and diet in an Asian population--a case-control study among Singapore Chinese. Int J Cancer 43 (6): 1007-16, 1989.  [PUBMED Abstract]

  105. Reddy BS, Hedges AR, Laakso K, et al.: Metabolic epidemiology of large bowel cancer: fecal bulk and constituents of high-risk North American and low-risk Finnish population. Cancer 42 (6): 2832-8, 1978.  [PUBMED Abstract]

  106. Kampman E, Giovannucci E, van 't Veer P, et al.: Calcium, vitamin D, dairy foods, and the occurrence of colorectal adenomas among men and women in two prospective studies. Am J Epidemiol 139 (1): 16-29, 1994.  [PUBMED Abstract]

  107. Neugut AI, Garbowski GC, Lee WC, et al.: Dietary risk factors for the incidence and recurrence of colorectal adenomatous polyps. A case-control study. Ann Intern Med 118 (2): 91-5, 1993.  [PUBMED Abstract]

  108. Schatzkin A, Lanza E, Corle D, et al.: Lack of effect of a low-fat, high-fiber diet on the recurrence of colorectal adenomas. Polyp Prevention Trial Study Group. N Engl J Med 342 (16): 1149-55, 2000.  [PUBMED Abstract]

  109. Cheah PY: Hypotheses for the etiology of colorectal cancer--an overview. Nutr Cancer 14 (1): 5-13, 1990.  [PUBMED Abstract]

  110. Reddy BS, Engle A, Simi B, et al.: Effect of dietary fiber on colonic bacterial enzymes and bile acids in relation to colon cancer. Gastroenterology 102 (5): 1475-82, 1992.  [PUBMED Abstract]

  111. Morotomi M, Guillem JG, LoGerfo P, et al.: Production of diacylglycerol, an activator of protein kinase C, by human intestinal microflora. Cancer Res 50 (12): 3595-9, 1990.  [PUBMED Abstract]

  112. Steinmetz KA, Potter JD: Vegetables, fruit, and cancer. I. Epidemiology. Cancer Causes Control 2 (5): 325-57, 1991.  [PUBMED Abstract]

  113. Steinmetz KA, Potter JD: Vegetables, fruit, and cancer. II. Mechanisms. Cancer Causes Control 2 (6): 427-42, 1991.  [PUBMED Abstract]

  114. Jacobs LR: Fiber and colon cancer. Gastroenterol Clin North Am 17 (4): 747-60, 1988.  [PUBMED Abstract]

  115. Roediger WE: The effect of bacterial metabolites on nutrition and function of the colonic mucosa: symbiosis between man and bacteria. In: Kasper H, Goebell H, eds.: Colon and Nutrition. Lancaster, Pa: Lancaster Press. Falk Symposium 32, 1981, pp 11-25. 

  116. Jacobs LR: Relationship between dietary fiber and cancer: metabolic, physiologic, and cellular mechanisms. Proc Soc Exp Biol Med 183 (3): 299-310, 1986.  [PUBMED Abstract]

  117. Howe GR, Benito E, Castelleto R, et al.: Dietary intake of fiber and decreased risk of cancers of the colon and rectum: evidence from the combined analysis of 13 case-control studies. J Natl Cancer Inst 84 (24): 1887-96, 1992.  [PUBMED Abstract]

  118. Potter JD: Epidemiology of diet and cancer: evidence of human maladaptation. In: Micozzi MS, Moon TE, eds.: Macronutrients: Investigating their Role in Cancer. New York: Marcel Dekker, 1992, pp 55-84. 

  119. Kritchevsky D: Dietary guidelines. The rationale for intervention. Cancer 72 (3 Suppl): 1011-4, 1993.  [PUBMED Abstract]

  120. Fuchs CS, Giovannucci EL, Colditz GA, et al.: Dietary fiber and the risk of colorectal cancer and adenoma in women. N Engl J Med 340 (3): 169-76, 1999.  [PUBMED Abstract]

  121. World Cancer Research Fund, American Institute for Cancer Research: Food, Nutrition and the Prevention of Cancer: A Global Perspective. Washington, DC: The Institute, 1997. 

  122. Michels KB, Edward Giovannucci, Joshipura KJ, et al.: Prospective study of fruit and vegetable consumption and incidence of colon and rectal cancers. J Natl Cancer Inst 92 (21): 1740-52, 2000.  [PUBMED Abstract]

  123. Terry P, Giovannucci E, Michels KB, et al.: Fruit, vegetables, dietary fiber, and risk of colorectal cancer. J Natl Cancer Inst 93 (7): 525-33, 2001.  [PUBMED Abstract]

  124. Alberts DS, Martínez ME, Roe DJ, et al.: Lack of effect of a high-fiber cereal supplement on the recurrence of colorectal adenomas. Phoenix Colon Cancer Prevention Physicians' Network. N Engl J Med 342 (16): 1156-62, 2000.  [PUBMED Abstract]

  125. Bostick RM, Potter JD, McKenzie DR, et al.: Reduced risk of colon cancer with high intake of vitamin E: the Iowa Women's Health Study. Cancer Res 53 (18): 4230-7, 1993.  [PUBMED Abstract]

  126. Lee IM, Cook NR, Gaziano JM, et al.: Vitamin E in the primary prevention of cardiovascular disease and cancer: the Women's Health Study: a randomized controlled trial. JAMA 294 (1): 56-65, 2005.  [PUBMED Abstract]

  127. Bjelakovic G, Nikolova D, Simonetti RG, et al.: Antioxidant supplements for prevention of gastrointestinal cancers: a systematic review and meta-analysis. Lancet 364 (9441): 1219-28, 2004.  [PUBMED Abstract]

  128. Gorham ED, Garland CF, Garland FC, et al.: Vitamin D and prevention of colorectal cancer. J Steroid Biochem Mol Biol 97 (1-2): 179-94, 2005.  [PUBMED Abstract]

  129. Pritchard RS, Baron JA, Gerhardsson de Verdier M: Dietary calcium, vitamin D, and the risk of colorectal cancer in Stockholm, Sweden. Cancer Epidemiol Biomarkers Prev 5 (11): 897-900, 1996.  [PUBMED Abstract]

  130. Giovannucci E, Stampfer MJ, Colditz GA, et al.: Multivitamin use, folate, and colon cancer in women in the Nurses' Health Study. Ann Intern Med 129 (7): 517-24, 1998.  [PUBMED Abstract]

  131. Cole BF, Baron JA, Sandler RS, et al.: Folic acid for the prevention of colorectal adenomas: a randomized clinical trial. JAMA 297 (21): 2351-9, 2007.  [PUBMED Abstract]

  132. Wargovich MJ, Eng VW, Newmark HL, et al.: Calcium ameliorates the toxic effect of deoxycholic acid on colonic epithelium. Carcinogenesis 4 (9): 1205-7, 1983.  [PUBMED Abstract]

  133. Slattery ML, Sorenson AW, Ford MH: Dietary calcium intake as a mitigating factor in colon cancer. Am J Epidemiol 128 (3): 504-14, 1988.  [PUBMED Abstract]

  134. Kune S, Kune GA, Watson LF: Case-control study of dietary etiological factors: the Melbourne Colorectal Cancer Study. Nutr Cancer 9 (1): 21-42, 1987.  [PUBMED Abstract]

  135. Yang CY, Chiu HF: Calcium and magnesium in drinking water and risk of death from rectal cancer. Int J Cancer 77 (4): 528-32, 1998.  [PUBMED Abstract]

  136. Zheng W, Anderson KE, Kushi LH, et al.: A prospective cohort study of intake of calcium, vitamin D, and other micronutrients in relation to incidence of rectal cancer among postmenopausal women. Cancer Epidemiol Biomarkers Prev 7 (3): 221-5, 1998.  [PUBMED Abstract]

  137. Manousos O, Day NE, Trichopoulos D, et al.: Diet and colorectal cancer: a case-control study in Greece. Int J Cancer 32 (1): 1-5, 1983.  [PUBMED Abstract]

  138. Wargovich MJ, Baer AR: Basic and clinical investigations of dietary calcium in the prevention of colorectal cancer. Prev Med 18 (5): 672-9, 1989.  [PUBMED Abstract]

  139. Lipkin M, Newmark H: Effect of added dietary calcium on colonic epithelial-cell proliferation in subjects at high risk for familial colonic cancer. N Engl J Med 313 (22): 1381-4, 1985.  [PUBMED Abstract]

  140. Buset M, Lipkin M, Winawer S, et al.: Inhibition of human colonic epithelial cell proliferation in vivo and in vitro by calcium. Cancer Res 46 (10): 5426-30, 1986.  [PUBMED Abstract]

  141. Wargovich MJ, Isbell G, Shabot M, et al.: Calcium supplementation decreases rectal epithelial cell proliferation in subjects with sporadic adenoma. Gastroenterology 103 (1): 92-7, 1992.  [PUBMED Abstract]

  142. Bostick RM, Potter JD, Fosdick L, et al.: Calcium and colorectal epithelial cell proliferation: a preliminary randomized, double-blinded, placebo-controlled clinical trial. J Natl Cancer Inst 85 (2): 132-41, 1993.  [PUBMED Abstract]

  143. Konishi H, Steinbach G, Hittelman WN, et al.: Cell kinetic analysis of intact rat colonic crypts by confocal microscopy and immunofluorescence. Gastroenterology 111 (6): 1493-500, 1996.  [PUBMED Abstract]

  144. 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 340 (2): 101-7, 1999.  [PUBMED Abstract]

  145. Grau MV, Baron JA, Sandler RS, et al.: Prolonged effect of calcium supplementation on risk of colorectal adenomas in a randomized trial. J Natl Cancer Inst 99 (2): 129-36, 2007.  [PUBMED Abstract]

  146. Wactawski-Wende J, Kotchen JM, Anderson GL, et al.: Calcium plus vitamin D supplementation and the risk of colorectal cancer. N Engl J Med 354 (7): 684-96, 2006.  [PUBMED Abstract]

  147. Forman MR, Levin B: Calcium plus vitamin D3 supplementation and colorectal cancer in women. N Engl J Med 354 (7): 752-4, 2006.  [PUBMED Abstract]

  148. Jacobs EJ, Rodriguez C, Brady KA, et al.: Cholesterol-lowering drugs and colorectal cancer incidence in a large United States cohort. J Natl Cancer Inst 98 (1): 69-72, 2006.  [PUBMED Abstract]

  149. Dale KM, Coleman CI, Henyan NN, et al.: Statins and cancer risk: a meta-analysis. JAMA 295 (1): 74-80, 2006.  [PUBMED Abstract]

Changes to This Summary (02/27/2014)

The PDQ cancer information summaries are reviewed regularly and updated as new information becomes available. This section describes the latest changes made to this summary as of the date above.

Description of the Evidence

Updated statistics with estimated new cases and deaths for 2014 (cited American Cancer Society as reference 1).

This summary is written and maintained by the PDQ Screening and Prevention Editorial Board, which is editorially independent of NCI. The summary reflects an independent review of the literature and does not represent a policy statement of NCI or NIH. More information about summary policies and the role of the PDQ Editorial Boards in maintaining the PDQ summaries can be found on the About This PDQ Summary and PDQ NCI's Comprehensive Cancer Database pages.

Questions or Comments About This Summary

If you have questions or comments about this summary, please send them to Cancer.gov through the Web site’s Contact Form. We can respond only to email messages written in English.

About This PDQ Summary



Purpose of This Summary

This PDQ cancer information summary for health professionals provides comprehensive, peer-reviewed, evidence-based information about colorectal cancer prevention. It is intended as a resource to inform and assist clinicians who care for cancer patients. It does not provide formal guidelines or recommendations for making health care decisions.

Reviewers and Updates

This summary is reviewed regularly and updated as necessary by the PDQ Screening and Prevention Editorial Board, which is editorially independent of the National Cancer Institute (NCI). The summary reflects an independent review of the literature and does not represent a policy statement of NCI or the National Institutes of Health (NIH).

Board members review recently published articles each month to determine whether an article should:

  • be discussed at a meeting,
  • be cited with text, or
  • replace or update an existing article that is already cited.

Changes to the summaries are made through a consensus process in which Board members evaluate the strength of the evidence in the published articles and determine how the article should be included in the summary.

Any comments or questions about the summary content should be submitted to Cancer.gov through the Web site's Contact Form. Do not contact the individual Board Members with questions or comments about the summaries. Board members will not respond to individual inquiries.

Levels of Evidence

Some of the reference citations in this summary are accompanied by a level-of-evidence designation. These designations are intended to help readers assess the strength of the evidence supporting the use of specific interventions or approaches. The PDQ Screening and Prevention Editorial Board uses a formal evidence ranking system in developing its level-of-evidence designations.

Permission to Use This Summary

PDQ is a registered trademark. Although the content of PDQ documents can be used freely as text, it cannot be identified as an NCI PDQ cancer information summary unless it is presented in its entirety and is regularly updated. However, an author would be permitted to write a sentence such as “NCI’s PDQ cancer information summary about breast cancer prevention states the risks succinctly: [include excerpt from the summary].”

The preferred citation for this PDQ summary is:

National Cancer Institute: PDQ® Colorectal Cancer Prevention. Bethesda, MD: National Cancer Institute. Date last modified <MM/DD/YYYY>. Available at: http://cancer.gov/cancertopics/pdq/prevention/colorectal/HealthProfessional. Accessed <MM/DD/YYYY>.

Images in this summary are used with permission of the author(s), artist, and/or publisher for use within the PDQ summaries only. Permission to use images outside the context of PDQ information must be obtained from the owner(s) and cannot be granted by the National Cancer Institute. Information about using the illustrations in this summary, along with many other cancer-related images, is available in Visuals Online, a collection of over 2,000 scientific images.

Disclaimer

The information in these summaries should not be used as a basis for insurance reimbursement determinations. More information on insurance coverage is available on Cancer.gov on the Coping with Cancer: Financial, Insurance, and Legal Information page.

Contact Us

More information about contacting us or receiving help with the Cancer.gov Web site can be found on our Contact Us for Help page. Questions can also be submitted to Cancer.gov through the Web site’s Contact Form.

Get More Information From NCI

Call 1-800-4-CANCER

For more information, U.S. residents may call the National Cancer Institute's (NCI's) Cancer Information Service toll-free at 1-800-4-CANCER (1-800-422-6237) Monday through Friday from 8:00 a.m. to 8:00 p.m., Eastern Time. A trained Cancer Information Specialist is available to answer your questions.

Chat online

The NCI's LiveHelp® online chat service provides Internet users with the ability to chat online with an Information Specialist. The service is available from 8:00 a.m. to 11:00 p.m. Eastern time, Monday through Friday. Information Specialists can help Internet users find information on NCI Web sites and answer questions about cancer.

Write to us

For more information from the NCI, please write to this address:

NCI Public Inquiries Office
9609 Medical Center Dr.
Room 2E532 MSC 9760
Bethesda, MD 20892-9760

Search the NCI Web site

The NCI Web site provides online access to information on cancer, clinical trials, and other Web sites and organizations that offer support and resources for cancer patients and their families. For a quick search, use the search box in the upper right corner of each Web page. The results for a wide range of search terms will include a list of "Best Bets," editorially chosen Web pages that are most closely related to the search term entered.

There are also many other places to get materials and information about cancer treatment and services. Hospitals in your area may have information about local and regional agencies that have information on finances, getting to and from treatment, receiving care at home, and dealing with problems related to cancer treatment.

Find Publications

The NCI has booklets and other materials for patients, health professionals, and the public. These publications discuss types of cancer, methods of cancer treatment, coping with cancer, and clinical trials. Some publications provide information on tests for cancer, cancer causes and prevention, cancer statistics, and NCI research activities. NCI materials on these and other topics may be ordered online or printed directly from the NCI Publications Locator. These materials can also be ordered by telephone from the Cancer Information Service toll-free at 1-800-4-CANCER (1-800-422-6237).