Prevention of colorectal cancer and dietary management.
Journal: 2015/April - Chinese Clinical Oncology
ISSN: 2304-3873
Abstract:
OBJECTIVE
This systematic review focuses on dietary and lifestyle risk factors for colorectal cancer (CRC) prevention and chemoprevention among high-risk populations.
METHODS
We searched PubMed for English-language articles about dietary components, lifestyle risk factors, and chemoprevention agents in relation to colorectal cancer and their references published from 1980 through 2013. We reviewed articles jointly for the most clinically important information, emphasizing randomized trials and meta-analyses where available.
RESULTS
There is convincing evidence that intake of garlic, vitamin B6 and magnesium, active living, maintaining a healthy weight and waist, avoiding or reducing red meat, alcohol, and smoking, as well as hormone replacement therapy among women may significantly protect against developing colorectal cancer. There is less consistent evidence for fruit and vegetable intake (fiber and folate), fish and Omega-3 fatty acids, selenium, dairy, calcium and vitamin D. For high-risk populations, aspirin have been shown to protect against the development of colonic adenomas and CRC, while a minimal effective dose remains unclear.
CONCLUSIONS
Colorectal cancer can be prevented in general population through dietary and lifestyle interventions, and aspirin may be a good choice of chemoprevention agent among high risk individuals.
Relations:
Content
Citations
(7)
References
(151)
Drugs
(11)
Affiliates
(1)
Similar articles
Articles by the same authors
Discussion board
Chin Clin Oncol 2(2): 13

Prevention of colorectal cancer and dietary management

Objectives

This systematic review focuses on dietary and lifestyle risk factors for colorectal cancer (CRC) prevention and chemoprevention among high-risk populations.

Methods and materials

We searched PubMed for English-language articles about dietary components, lifestyle risk factors, and chemoprevention agents in relation to colorectal cancer and their references published from 1980 through 2013. We reviewed articles jointly for the most clinically important information, emphasizing randomized trials and meta-analyses where available.

Results

There is convincing evidence that intake of garlic, vitamin B6 and magnesium, active living, maintaining a healthy weight and waist, avoiding or reducing red meat, alcohol, and smoking, as well as hormone replacement therapy among women may significantly protect against developing colorectal cancer. There is less consistent evidence for fruit and vegetable intake (fiber and folate), fish and Omega-3 fatty acids, selenium, dairy, calcium and vitamin D. For high-risk populations, aspirin have been shown to protect against the development of colonic adenomas and CRC, while a minimal effective dose remains unclear.

Conclusions

Colorectal cancer can be prevented in general population through dietary and lifestyle interventions, and aspirin may be a good choice of chemoprevention agent among high risk individuals.

Introduction

Colorectal cancer (CRC) is among the most commonly diagnosed cancers, with about 1 million new cases and 600,000 deaths worldwide each year (1). Incidence rates vary markedly around the world, with the highest incidence in Australia and New Zealand, Europe and North America, and the lowest in Africa and South-Central Asia (Figure 1) (2). Because of the high incidence of colorectal cancer in Western countries, it is commonly regarded as a Western life-style disease. However, the incidence rates have been increasing in economically transitioning countries, including Eastern European countries, most parts of Asia, and select countries of South America (3).

An external file that holds a picture, illustration, etc.
Object name is nihms-670898-f0001.jpg

Age-standardized colorectal cancer incidence rates by sex and region of the world. Source: Jemal et al. Global cancer statistics. CA Cancer J Clin 2011;61:69-90

The majority of CRCs are sporadic rather than familial, despite the striking increase in incidence that results from the form of the disease associated with inherited susceptibility (e.g., hereditary non-polyposis colon cancer, HNPCC, and familial adenomatous polyposis, FAP). The role of diet and lifestyle factors has long been suspected and investigated in CRC development, with specific dietary constituents, in addition to excessive caloric intake, weight gain, physical inactivity, smoking, and heavy alcohol intake all thought to result in elevated risk (4,5). The differences in rates by country, and elevated risk among immigrants from a low- to high-risk country (6), support that environmental factors are important in CRC risk. In one study, it was estimated that dietary factors contributed to nearly 50% of all CRC cases diagnosed, while the attributable risk was only about 10% for family history (7). Therefore, a healthy diet and lifestyle are seen as essential in primary prevention of CRC (8), and this review aims to summarize the most up-to-date evidence for these modifiable risk factors. With respect to additional risk modifying interventions, chemoprevention has been a major goal in cancer, and colorectal cancer is seen as amenable to this approach through safe and cost effective agents. We also summarize evidence to date on chemoprevention of CRC.

This review evaluated and summarized scientific evidence on dietary, lifestyle, and chemo-prevention of colorectal cancer. We searched the PubMed database for studies published in English, for key epidemiological studies and animal studies, large case-control and cohort studies, randomized controlled trials (RCTs), and meta-analyses of studies of these different types in humans, with particular focus on studies from the past decade. We summarized the findings in the text and listed key points in Table 1 by level and source of evidence.

Table 1

Summary of evidence on diet and lifestyle risk factors for colorectal cancer

Source of evidenceSummary points
Convincing/rather consistentRed meatMeta-analysis of observational studies (mostly prospective studies)Red and processed meat intake significantly increase CRC risk by ~20%, contrasting highest vs. lowest intake, with a dose-response relationship until 140 g/day; association is strong in men; frequency of consumption may matter more than total amount consumed; avoid burned meat
Vitamin B6Meta-analysis of prospective studiesVitamin B6 intake (statistically nonsignificant) and blood PLP levels (statistically significant) were inversely associated with CRC risk
GarlicSmall randomized trialSignificant suppression of adenoma in CRC patients with 12-mo high-dose of aged garlic extract; 29% reduction in developing new adenoma
Meta-analysis of observational studiesApproximately 30% reduction in CRC risk, contrasting high vs. low (16 g/wk average difference across studies) consumption of garlic
MagnesiumMeta-analysis of large cohort studies10% risk reduction contrasting the highest vs. lowest category of magnesium intake, with dose response
AlcoholMeta-analysis of 60 observational studiesSignificant increase in CRC risk with dose-response; 21% risk increase for moderate drinkers (2-3 drink/d) and 52% risk increase for heavy drinkers (4+ drink/d), compared to never drinkers
ObesityMeta-analysis of 31 observational studiesCRC risk increases by 7% (4-10%) with a 2 kg/m increase in BMI; CRC increases by 4% (2-5%) with a 2-cm increase in waist circumference
Physical activityMeta-analyses of observational studiesApproximately 20% risk reduction, contrasting physically active individuals vs. inactive
SmokingMeta-analyses of 100+ observational studies18% risk elevation contrasting smokers vs. nonsmokers, with dose-response; Risk may reduce immediately following cessation of smoking
Hormone replacement therapyMeta-analysis of observational studiesA 33% risk reduction for recent users, compared to nonusers; Protective effect may be short lived and disappear following cessation of use
Randomized trialA 44% reduced CRC risk (95% CI, 19-62%), comparing the E+P arm vs. placebo arm
Probable/less consistentFruit and vegetables (Dietary fiber)Observational studiesMany reported an inverse association but a few large cohort studies reported null association
Meta-analysis of large cohort studiesInconsistent. The inverse association is weak, about ~10% risk reduction
Randomized trialNo association
Fruit and vegetables (folate)Case-control studyMinimal essential serum folate should be above 8.0 ng/mL for reduced risk of developing colorectal adenoma
Cohort studiesApproximately 40% risk reduction contrasting highest vs. lowest quartile of intake; both long- and short-term intake may reduce risk
Meta-analysis of randomized or pseudo-randomized trialsEvidence inconsistent; most meta-analyses reported null results
Fish and Omega-3 fatty acids (FA)Longitudinal cohort study(Omega-3 FA) Significant risk reduction over 22-year follow-up
Observational studies(Fish) Evidence inconsistent
Meta-analysis of observational studies(Fish) A significant 12% reduction in CRC risk contrasting highest vs. lowest fish consumption
Randomized trial(Fish) No marked change of apoptosis and mitosis within the colonic crypt with 6-mo intervention with oil-rich or lean fish
Probable/less consistentSeleniumRandomized trialSelenium supplementation reduced CRC risk but results were attenuated and insignificant after additional years of follow-up
Meta-analysis of observational and clinical trialsNo association in women but significant inverse association in men
Dairy/calciumMeta-analysis of observational studiesInverse association, reduced risk with greater intake
Meta-analysis of randomized trialsInverse association among individuals with a history of adenomas, but no association otherwise
Vitamin DMeta-analysis of longitudinal studiesInverse association yet studies were limited to small sample sizes

Dietary and nutritional components

Red meat

Whether red meat is a culprit in causing CRC has been the subject of scientific debate. Extensive evidence suggests that long-term consumption of red meat or processed meats may increase CRC risk (9-11). A recent meta-analysis of 21 prospective studies showed that consumption of red and processed meats was associated with elevated CRC risk [relative risk, RR=1.22, 95% confidence interval (CI), 1.11-1.34, contrasting the highest versus lowest quartiles of intake], with a linear increase of CRC (RR=1.14, 95% CI, 1.04-1.24) associated with every 100 g/day increase of meat intake until a plateau was encountered at 140 g/day (12). Another meta-analysis of 25 prospective studies that same year found the association was similar by tumor site (colon cancer: RR=1.11, 95% CI, 1.03-1.19, and rectal cancer: RR=1.19, 95% CI, 0.97-1.46) but differed between men (RR=1.21, 95% CI, 1.04-1.42) and women (RR=1.01, 95% CI, 0.87-1.17) (13). Another meta-analysis of both cohort and case-control studies (22 in total) reported that the frequency of red meat consumption rather than total amount of consumed meat is associated with a higher CRC carcinogenesis (14). In addition, the processing method may matter. Very well-cooked meat or meats cooked in direct contact with flames were reported to raise the CRC risk, which may be explained by the carcinogenic heterocyclic amines produced during cooking (15,16). Other plausible biological mechanisms include endogenous formation of nitroso compounds in the gastrointestinal tract by red meat intake, but not by white meat intake (17).

Fruit and vegetables (fiber and folic acid)

Many observational studies have reported an inverse association between dietary fiber and CRC risk, with a relative reduction of up to 40% (18-22), although a few large cohort studies reported small, statistically null associations (23-25). A large pooled analysis of thirteen prospective cohorts suggested that dietary fiber intake was inversely associated with CRC risk in age-adjusted analyses, but no association remained after accounting for other dietary risk factors, including red meat, alcohol, folate, and total milk (26). A recent meta-analysis reported a dose-response analysis indicating that for each 10 g/day total dietary fiber, the relative risk of developing CRC was 0.90 (95% CI, 0.86-0.94) (27). However, two randomized trials found that high dietary fiber did not affect the recurrence of colorectal adenoma (28,29).

Dietary folate or folic acid (from dietary supplements and food fortification) is necessary to synthesize, repair and methylate DNA. It is especially important during periods of rapid cell division and growth such as pregnancy and infancy. Folate is also thought to help prevent changes to DNA that may lead to cancer, and its rule in CRC carcinogenesis has been extensively studied. Humans cannot synthesize folate de novo, and therefore folate has to be supplied through diet to meet their daily requirements, with fresh fruits and vegetables being the major sources. There are growing data and a continuing controversy over the effect of folic acid supplementation on cancer risk. Both the Nurse's Health Study and secondary analysis from the Wheat Bran Fiber randomized trial reported that high dietary folate was associated with 40% reduced risk of colorectal adenoma (highest vs. lowest quartile) (30,31). The Nurses’ Health Study and Health Professionals Follow-Up Study further suggested that both long- and short-term intakes of total folate were associated with a lower risk of colorectal adenoma (32). With respect to dose, one case-control study based on suspected patients undergoing screening reported that the minimal essential serum folate concentrations should be above 8.0 ng/mL for reducing risk of developing colorectal adenoma (33). One meta-analysis summarized 3 randomized or pseudo-randomized trials and concluded that folate status was inversely related to the risk of developing CRC (34); however, a recently published larger meta-analysis of 15 randomized trials reported that folic acid supplementation has no significant effect on CRC risk (35). A meta-analysis of 3 large randomized trials of folic acid supplementation among patients with an adenoma history also reported no association (36).

Garlic

Garlic is characterized by a high content in organo-sulfur compounds and flavonoids. The allyl sulfur constituents in garlic comprise about 1% of its dry weight and are responsible for its health benefits (37,38). Garlic also contains other constituents such as flavonoids and selenium that are considered to have antioxidant properties and anticarcinogenic activity. Preclinical investigations provide convincing evidence that garlic and related sulfur-containing compounds inhibit carcinogen-induced tumors in various organs (39). One RCT conducted within a small sample of 37 patients with CRC found (I) a significant suppression in both the total size and number of adenomas in CRC patients after 12-mo of high-dose aged garlic extract (40), and (II) a 29% reduction in developing at least one new adenoma (41). A meta-analysis of 4 case control and 3 cohort studies confirmed the inverse association between garlic intake and CRC risk, with an approximate 30% relative reduction in incidence contrasting high vs. low (16 g/wk average difference across studies) consumption of garlic (42).

Fish and Omega-3 fatty acids

Long-chain n-3 fatty acids have been suggested to play a protective role in colorectal cancer development in laboratory and animal studies, with the mechanism of action conjectured to be inhibition of the cyclooxygenase-2 (COX-2) enzyme and the production of arachidonic acid (n-6) derived eicosanoids (40-42). A long-term prospective study of U.S. men reported a significantly reduced CRC risk contrasting the highest versus lowest quartiles of n-3 fatty acids (odds ratio, OR=0.74, 95% CI, 0.57-0.95) (43). Fish is the main dietary source of the long-chain n-3 fatty acids. Some observational studies found an inverse association between fish consumption and CRC risk (10,43-45), while others did not (46-48). A recent meta-analysis pooled data from 22 cohort and 19 case-control studies, and reported a 12% (OR=0.88, 95% CI, 0.80-0.95) reduction in CRC risk contrasting highest vs. lowest fish consumption (49). However, a multi-center, randomized controlled trial investigated the effect of 6-month intervention with oil-rich or lean fish on apoptosis and mitosis within the colonic crypt, and found no marked change in these endpoints (50).

Vitamin B6

Vitamin B6 is widely distributed in foods, with good sources including meats, whole grain products, vegetables, nuts and bananas. Vitamin B6 is involved in almost 100 enzymatic reactions, among which one function involves transferring 1-carbon groups for DNA synthesis and methylation (51). Therefore, vitamin B6 deficiency may increase CRC risk through aberrations in DNA synthesis, repair, and methylation. Vitamin B may also suppress colorectal carcinogenesis by reducing cell proliferation, angiogenesis, oxidative stress, inflammation, and nitric oxide synthesis (52,53). A meta-analysis summarized evidence from prospective studies, with 9 studies on vitamin B6 intake and 4 studies on blood pyridoxal 5’-phosphate (PLP, the principal active coenzyme form of vitamin B6) levels, in relation to CRC risk. The pooled RRs of CRC contrasting highest vs. lowest category of vitamin B6 intake and blood PLP levels were 0.90 (95% CI, 0.75-1.07) and 0.52 (95% CI, 0.38-0.71), respectively (54).

Dairy, calcium and vitamin D

In vitro and in vivo studies have suggested that high dietary intake of calcium and vitamin D may reduce CRC risk by a variety of mechanisms, such as reducing epithelial cell exposure to toxicity, inhibiting proliferation of intestinal mucosa and epithelial cells via the intracellular action of calcium (55); Vitamin D's protective effect against colorectal neoplasia is by reducing epithelial cell proliferation (56). There have been numerous epidemiological studies on calcium and CRC risk, as well as studies on vitamin D and CRC risk. However, there remains a lack of clarity of the effects on CRC of both these dietary components, due to inconsistent findings among numerous epidemiological studies and multiple pooled analyses. Overall, results from observational studies found calcium intake to be not associated with a substantially lower CRC risk (57,58) particularly for the more reliable large prospective studies (57). A meta-analysis of summarizing a large number of observational studies mostly supported an inverse association for both calcium and milk/dairy products (a rich source of both calcium and vitamin D) (59,60). The evidence from randomized trials only shows a protective effect of calcium on adenoma recurrence among individuals with a history of adenomas (RR=0.80, 95% CI, 0.69-0.94) for those receiving calcium 1,200 to 2,000 mg/d, but no effect in general populations (RR=0.62, 95% CI, 0.11-3.40) (59). It should be noted that the statistical power in the two clinical trials conducted in general populations is limited (59).

In 1980, Garland hypothesized that lower levels of vitamin D resulting from weaker UV-B radiation at higher latitudes may account for the striking geographical pattern of CRC mortality (60). The evidence from epidemiologic studies are limited to small sample sizes, yet in general found vitamin D inversely associated with CRC risk (57,61-64). Meta-analyses of longitudinal studies reported a 50% lower CRC risk associated with a serum 25(OH)D level ≥33 ng/mL, compared to ≤12 ng/mL (65), or an OR of 0.57 (95% CI, 0.43-0.76) for an increase of 25(OH)D by 20 ng/mL (66).

Selenium

As an essential trace element involved in different physiological functions in the human body, selenium has received increasing attention as a possible cancer prevention substance, through several possible mechanisms for the potential anticarcinogenic effects including apoptosis (67), protection from oxidative DNA damage (68), and increased immune function (69). However, the epidemiologic evidence on associations between selenium and CRC risk has been mixed, inconclusive and limited to small studies. Many observational studies support a protective effect of selenium on CRC risk (70-72) but not consistently (73,74). A meta-analysis of 12 observational studies and 2 clinical trials reported no association between selenium and CRC risk in women, but an inverse association in men (OR=0.68, 95% CI, 0.57-0.82) (75). There was a large randomized trial on selenium supplementation for skin cancer prevention, and the secondary analysis showed a 58% reduction (95% CI, 0.18-0.95) in CRC incidence among participants randomly assigned to take selenium (200 μg daily) (76), although the results were attenuated and no longer statistically significant after additional years of follow- up (77). A pooled analysis of 3 randomized trials on various nutritional interventions for CRC prevention reported a pooled OR of 0.66 (95% CI, 0.50-0.87), contrasting participants of highest vs. lowest blood selenium values (78). Yet, none of these trials were designed to test selenium as an intervention, and all participants were at high adenoma risk after recent colonoscopic adenoma resection (79).

Magnesium

Magnesium is abundant in many foods, and particularly rich in spices, nuts, cereals, coffee, cocoa, tea and green-leafy vegetables. Magnesium is involved in a wide variety of biochemical reactions that modulate key cell functions, and has a crucial role in genomic stability and DNA synthesis (80). Epidemiologic studies suggested that magnesium may be associated with a decreased CRC risk but the findings are inconsistent. A meta-analysis of 8 prospective studies containing 338,979 participants and 8,000 CRC cases reported a summary RR of 0.89 (95% CI, 0.79-1.00) for the highest vs. lowest category of magnesium intake, with evidence of a dose-response (81).

Dietary pattern, obesity and related mechanisms

Dietary pattern summarizes the total diet or the key dietary components including food items, food groups, and nutrients (82), and may provide additional insights with the combined effects of many food components. Summarized epidemiologic evidence suggest that healthier pattern consisting of greater intakes of fruits and vegetables, and lower intakes of red and processed meat, appeared protective against colorectal adenoma and cancer incidence, while a less healthy pattern characterized by higher intakes of red and processed meat, as well as potatoes and refined carbohydrates, may increase risk (83). There has also been great interest in total dietary consumption and its consequences, specifically obesity, in relation to colorectal and other cancers (84). Because these risks are modifiable, these pathways are seen as key in cancer control efforts (85). Colon cancer is among those specifically identified as linked to obesity; for example, two large prospective cohort studies have demonstrated that being obese [body mass index (BMI) >30 kg/m] confers a 1.5-fold greater risk of developing colon cancer relative to individuals of normal weight (BMI 18.5-24.9 kg/m) (86,87). With respect to mortality, the Cancer Prevention Study II, which prospectively followed more than 900,000 US adults, similarly found a 1.5- to 1.8-fold increased risk of colon cancer related death for obese individuals, reflecting the increased incidence of and perhaps the increased mortality from colon cancer once diagnosed (88). A meta-analysis of 31 observational studies reported that for a 2 kg/m increase in BMI, the CRC risk increased by 7% (95% CI, 4-10%), and for a 2-cm increase in waist circumference, the risk increased by 4% (95% CI, 2-5%) (89). Another meta-analysis showed that the inverse association between BMI and CRC risk was stronger for colon than rectal cancer, and for men than women (90). Some recent research probing obesity pathways concern the issue of ‘secondary prevention’ after diagnosis and treatment of early stage disease. Following earlier studies by Meyerhardt et al. (91) and Dignam et al. (92), which found excess risk of both cancer recurrence and overall mortality among obese patients utilizing large participant cohorts from multicenter clinical trials for stage II and III colon cancer, subsequent studies turned to elucidating the role of dietary constituents among colon cancer patients. The first of these studies examined the ‘Western’ diet pattern characterized by relatively high red and processed meats, refined grain, and sugar content, and indicated that those with higher consumption of these components experienced significantly elevated recurrence and mortality risk (93). A more recent study more closely examined the Western diet and the role of dietary glycemic measures (94). This study found that high carbohydrate intake and glycemic load significantly increased recurrence and mortality. These effects were present among all body types but strongest among those who were overweight or obese. In relation to CRC risk, the Western dietary pattern has previously been reported to be associated with an elevated CRC incidence (95). However, the Women's Health Initiative Dietary Modification Trial, a large randomized trial conducted in postmenopausal women, suggested that a low-fat dietary pattern intervention did not reduce CRC risk during 8 years of follow-up (96).

This work relates to a large body of research focused on potential mechanisms by which obesity leads to higher colon cancer incidence. One prominent theme involves metabolic and insulin related pathways (97-104). Insulin, insulin-like growth factors (IGF), and IGF binding proteins have all been causatively implicated for colon cancer via mitogenic effects on the colonic mucosa and other mechanisms. In a prospective nested case-control study within the Physicians’ Health Study, Ma et al. (101) found that men in the highest quintile for IGF-1 had a 2.5 increased risk of colorectal cancer compared to the lowest quintile. Among women, an analysis of the Nurse's Health Study cohort found a comparable >2-fold for those in the highest quartile of IGF-1 compared with those in the lowest (102). That study, as well as investigations by Kaaks et al. (103) and a more recent study by Ma et al. (104) also observed that colorectal cancer risk increased with increasing levels of C-peptide (a marker of insulin production). Interestingly, reduction in IGF exposure via increased presence of its binding protein was found to significantly reduce colon cancer mortality among affected individuals in a large cohort study (105). This particular pathway represents but one of numerous putative links linking cancer and obesity (84).

Obviously, a critical component of obesity is physical activity. The association between physical activity and CRC risk is well established, with the majority of relevant studies finding physically active individuals benefit from substantial risk reduction (approximately 20%) comparing to the sedentary ones, confirmed by two meta-analyses (106,107). The evidence is strong and consistent for both proximal colon and distal colon cancers with a similar magnitude (107). The main challenge is disentangling the direct effects of physical activity from strongly related factors such as obesity and comorbidities that may influence outcomes. Randomized intervention trials have been completed with positive findings, and others are underway; these studies have been mostly focused on colorectal cancer survivors (clinical event reduction) or biomarkers (prevention studies) for practical reasons (108-110).

Other common lifestyle exposures

Alcohol

Alcohol consumption is one of the most important known causes of human cancer, possibly through genotoxic effect of acetaldehyde (111) and interference of folate absorption by alcohol (112). The most recent meta-analysis pooled data from 27 cohort and 34 case-control studies, and reported significantly increased CRC risk with a dose-response relationship (113): comparing to never drinkers, moderate drinkers who consume 2-3 drinks per day have 21% increased CRC risk and heavy drinkers who have 4 or more drinks per day have 52% increased CRC risk. These results are consistent with other pooled analyses, with increased risk observed for both colon and rectal cancers (114-116).

Smoking

Cigarette smoking has been shown to cause many nonpulmonary cancers with no direct tobacco-related carcinogens, yet the association between smoking and CRC remains controversial. A meta-analysis of 106 observational studies estimated that cigarette smokers are 1.18 times (95% CI, 1.11-1.25 times) more likely to develop CRC compared to those who never smoke, with evidence of a dose-response (117). Another meta-analysis of 36 studies reported that duration and age of initiation were also significantly associated with CRC incidence (118). A recent study reported that the excess CRC risk due to smoking decreased immediately after quitting for proximal colon and rectal cancer, but not until about 20 years post-quitting for distal colon cancer (119).

Hormone replacement therapy (HRT)

Estrogen/progestin replacement therapy is prescribed to control postmenopausal symptom or to prevent hormone deficiency-related diseases such as osteoporosis. Existing evidence shows that HRT is associated with lower CRC risk, yet the mechanism remained unclear. Case control studies have shown significant reduction in CRC risk among ever users (120,121). Large cohort studies suggested a modest risk reduction among ever users (122), but there was a lack of dose-response relationship (123). Also, the association was found stronger among women aged 65+, with a body mass index <30 and who regularly use aspirin or ibuprofen (124). A meta-analysis concluded that recent HRT users had a 33% reduction in CRC risk (pooled RR=0.67, 95% CI, 0.59-0.77) but no association was observed with ever use of HRT and duration of use did not matter (125). Other studies also supported that the protective effect of HRT is short-lived and disappears following cessation of use (124). The Women's Health Initiative trial found that women who took estrogen plus progestin had 44% reduced risk (95% CI, 19-62%) of colorectal cancer compared to women who took placebo but colorectal cancers in the estrogen plus progestin group were at a more advanced stage (126). The Women's Health Initiative trial later reported that there was no significant difference in colorectal cancer incidence between the estrogen-only and placebo arms (127).

Chemoprevention of colorectal cancer

Prevention of colorectal cancer and its precursor conditions via some substance or compound, or chemoprevention, offers great potential if the intervention is safe and cost-effective. The main focus of these has not been nutritional, but rather medicinal agents with reasonably safe adverse event profiles. For more than two decades, accumulating evidence from observational studies (128-133) and large randomized trials (134-137) suggests that aspirin and other non-steroidal anti-inflammatory drugs (NSAIDs) protect against the development of colonic adenomas and CRC, and reduces disease recurrence (136-139). Meta-analysis of 17 case-control studies showed an inverse association between regular use of aspirin and reduced CRC risk (pooled OR=0.62, 95% CI, 0.58-0.67), while the association was weaker in cohort studies presumably due to substantial variation between studies in measuring aspirin exposure (140). Evidence from randomized trials has not been consistent. Two trials conducted among patients with a history of CRC or adenomas showed that low-dose aspirin has a chemopreventive effect on new adenomas (136,137). Yet among healthy individuals, two earlier large trials of low-dose aspirin (the Physicians’ Health Study and the Women's Health Study) showed no effect on CRC incidence during 10-year follow-up, possibly due to an insufficient dose (134,141,142). Recent long-term follow-up of four aspirin trials (Primary: Thrombosis Prevention Trial, British Doctors Aspirin Trial; Secondary: Swedish Aspirin Low Dose Trial, UK-TIA Aspirin Trial) showed a reduction of 50% in CRC risk after five or more years of regular consumption with a dosage of at least 75 mg daily (75-300 mg) (143). Aspirin has emerged as the most likely NSAID for use in chemoprevention because of its known cardiovascular benefit and available safety and efficacy data (144). Other traditional NSAIDs, particularly selective COX-2 inhibitors such as celecoxib, have been shown to cause regression of adenomas in familial adenomatous polyposis, and are now given to patients at high risk of CRC (138,139), and yet cannot be routinely recommended concerning potential cardiovascular events. The Colorectal Adenoma/carcinoma Prevention Programme (CAPP) was launched in 1990, with CAPP1 investigating familial adenomatous polyposis in 200 young adults and CAPP2 being the first large-scale genetically targeted chemoprevention trial in 1,000 adults with HNPCC (also known as Lynch syndrome). The CAPP2 randomized trial reported that 600 mg aspirin per day for a mean of 25 months substantially reduced CRC incidence among carriers of hereditary CRC (145). However, the minimum dose of aspirin to achieve the protective effect is still uncertain and will be the objective of a new CAPP trial in Lynch Syndrome. The underlying mechanism of NSAIDs inhibiting carcinogenesis remains inconclusive, with proposed explanations as increased apoptosis and impairment of tumor cell growth by inhibition of cyclooxygenase-2 (COX-2) (146).

The side-effects of NSAIDs are well documented and mainly attributed to inhibition of COX activity. The most frequently reported serious adverse events associated with aspirin use are related to gastrointestinal bleeding, even with low-dose aspirin (147). Comparing to other NSAIDs, aspirin has lower risk of occlusive cardiovascular events, yet the dose-dependent risk of bleeding complications with aspirin intake may limit its potential for primary prevention of CRC (148). In additional to optimal dose, optimum treatment duration and age of initiation also remained uncertain (144). Concerning risk and benefit, the United States Preventive Services Task Force recommended against the routine use of aspirin for CRC prevention in 2007 (149). However, the 2011 pooled analysis with 8 randomized controlled trials showed that daily aspirin for 5-10 years reduced 5-year cancer mortality by 34% (P=0.03) and a 20-year cancer mortality by 20% (P<0.001) (150). Considering the bleeding complications are not life-threatening, the risk-benefit of aspirin use in CRC prevention requires a formal re-evaluation. As toxicity of aspirin largely depends on dosage, the minimal effective dose required for CRC prevention is critical (151).

Dietary and nutritional components

Red meat

Whether red meat is a culprit in causing CRC has been the subject of scientific debate. Extensive evidence suggests that long-term consumption of red meat or processed meats may increase CRC risk (9-11). A recent meta-analysis of 21 prospective studies showed that consumption of red and processed meats was associated with elevated CRC risk [relative risk, RR=1.22, 95% confidence interval (CI), 1.11-1.34, contrasting the highest versus lowest quartiles of intake], with a linear increase of CRC (RR=1.14, 95% CI, 1.04-1.24) associated with every 100 g/day increase of meat intake until a plateau was encountered at 140 g/day (12). Another meta-analysis of 25 prospective studies that same year found the association was similar by tumor site (colon cancer: RR=1.11, 95% CI, 1.03-1.19, and rectal cancer: RR=1.19, 95% CI, 0.97-1.46) but differed between men (RR=1.21, 95% CI, 1.04-1.42) and women (RR=1.01, 95% CI, 0.87-1.17) (13). Another meta-analysis of both cohort and case-control studies (22 in total) reported that the frequency of red meat consumption rather than total amount of consumed meat is associated with a higher CRC carcinogenesis (14). In addition, the processing method may matter. Very well-cooked meat or meats cooked in direct contact with flames were reported to raise the CRC risk, which may be explained by the carcinogenic heterocyclic amines produced during cooking (15,16). Other plausible biological mechanisms include endogenous formation of nitroso compounds in the gastrointestinal tract by red meat intake, but not by white meat intake (17).

Fruit and vegetables (fiber and folic acid)

Many observational studies have reported an inverse association between dietary fiber and CRC risk, with a relative reduction of up to 40% (18-22), although a few large cohort studies reported small, statistically null associations (23-25). A large pooled analysis of thirteen prospective cohorts suggested that dietary fiber intake was inversely associated with CRC risk in age-adjusted analyses, but no association remained after accounting for other dietary risk factors, including red meat, alcohol, folate, and total milk (26). A recent meta-analysis reported a dose-response analysis indicating that for each 10 g/day total dietary fiber, the relative risk of developing CRC was 0.90 (95% CI, 0.86-0.94) (27). However, two randomized trials found that high dietary fiber did not affect the recurrence of colorectal adenoma (28,29).

Dietary folate or folic acid (from dietary supplements and food fortification) is necessary to synthesize, repair and methylate DNA. It is especially important during periods of rapid cell division and growth such as pregnancy and infancy. Folate is also thought to help prevent changes to DNA that may lead to cancer, and its rule in CRC carcinogenesis has been extensively studied. Humans cannot synthesize folate de novo, and therefore folate has to be supplied through diet to meet their daily requirements, with fresh fruits and vegetables being the major sources. There are growing data and a continuing controversy over the effect of folic acid supplementation on cancer risk. Both the Nurse's Health Study and secondary analysis from the Wheat Bran Fiber randomized trial reported that high dietary folate was associated with 40% reduced risk of colorectal adenoma (highest vs. lowest quartile) (30,31). The Nurses’ Health Study and Health Professionals Follow-Up Study further suggested that both long- and short-term intakes of total folate were associated with a lower risk of colorectal adenoma (32). With respect to dose, one case-control study based on suspected patients undergoing screening reported that the minimal essential serum folate concentrations should be above 8.0 ng/mL for reducing risk of developing colorectal adenoma (33). One meta-analysis summarized 3 randomized or pseudo-randomized trials and concluded that folate status was inversely related to the risk of developing CRC (34); however, a recently published larger meta-analysis of 15 randomized trials reported that folic acid supplementation has no significant effect on CRC risk (35). A meta-analysis of 3 large randomized trials of folic acid supplementation among patients with an adenoma history also reported no association (36).

Garlic

Garlic is characterized by a high content in organo-sulfur compounds and flavonoids. The allyl sulfur constituents in garlic comprise about 1% of its dry weight and are responsible for its health benefits (37,38). Garlic also contains other constituents such as flavonoids and selenium that are considered to have antioxidant properties and anticarcinogenic activity. Preclinical investigations provide convincing evidence that garlic and related sulfur-containing compounds inhibit carcinogen-induced tumors in various organs (39). One RCT conducted within a small sample of 37 patients with CRC found (I) a significant suppression in both the total size and number of adenomas in CRC patients after 12-mo of high-dose aged garlic extract (40), and (II) a 29% reduction in developing at least one new adenoma (41). A meta-analysis of 4 case control and 3 cohort studies confirmed the inverse association between garlic intake and CRC risk, with an approximate 30% relative reduction in incidence contrasting high vs. low (16 g/wk average difference across studies) consumption of garlic (42).

Fish and Omega-3 fatty acids

Long-chain n-3 fatty acids have been suggested to play a protective role in colorectal cancer development in laboratory and animal studies, with the mechanism of action conjectured to be inhibition of the cyclooxygenase-2 (COX-2) enzyme and the production of arachidonic acid (n-6) derived eicosanoids (40-42). A long-term prospective study of U.S. men reported a significantly reduced CRC risk contrasting the highest versus lowest quartiles of n-3 fatty acids (odds ratio, OR=0.74, 95% CI, 0.57-0.95) (43). Fish is the main dietary source of the long-chain n-3 fatty acids. Some observational studies found an inverse association between fish consumption and CRC risk (10,43-45), while others did not (46-48). A recent meta-analysis pooled data from 22 cohort and 19 case-control studies, and reported a 12% (OR=0.88, 95% CI, 0.80-0.95) reduction in CRC risk contrasting highest vs. lowest fish consumption (49). However, a multi-center, randomized controlled trial investigated the effect of 6-month intervention with oil-rich or lean fish on apoptosis and mitosis within the colonic crypt, and found no marked change in these endpoints (50).

Vitamin B6

Vitamin B6 is widely distributed in foods, with good sources including meats, whole grain products, vegetables, nuts and bananas. Vitamin B6 is involved in almost 100 enzymatic reactions, among which one function involves transferring 1-carbon groups for DNA synthesis and methylation (51). Therefore, vitamin B6 deficiency may increase CRC risk through aberrations in DNA synthesis, repair, and methylation. Vitamin B may also suppress colorectal carcinogenesis by reducing cell proliferation, angiogenesis, oxidative stress, inflammation, and nitric oxide synthesis (52,53). A meta-analysis summarized evidence from prospective studies, with 9 studies on vitamin B6 intake and 4 studies on blood pyridoxal 5’-phosphate (PLP, the principal active coenzyme form of vitamin B6) levels, in relation to CRC risk. The pooled RRs of CRC contrasting highest vs. lowest category of vitamin B6 intake and blood PLP levels were 0.90 (95% CI, 0.75-1.07) and 0.52 (95% CI, 0.38-0.71), respectively (54).

Dairy, calcium and vitamin D

In vitro and in vivo studies have suggested that high dietary intake of calcium and vitamin D may reduce CRC risk by a variety of mechanisms, such as reducing epithelial cell exposure to toxicity, inhibiting proliferation of intestinal mucosa and epithelial cells via the intracellular action of calcium (55); Vitamin D's protective effect against colorectal neoplasia is by reducing epithelial cell proliferation (56). There have been numerous epidemiological studies on calcium and CRC risk, as well as studies on vitamin D and CRC risk. However, there remains a lack of clarity of the effects on CRC of both these dietary components, due to inconsistent findings among numerous epidemiological studies and multiple pooled analyses. Overall, results from observational studies found calcium intake to be not associated with a substantially lower CRC risk (57,58) particularly for the more reliable large prospective studies (57). A meta-analysis of summarizing a large number of observational studies mostly supported an inverse association for both calcium and milk/dairy products (a rich source of both calcium and vitamin D) (59,60). The evidence from randomized trials only shows a protective effect of calcium on adenoma recurrence among individuals with a history of adenomas (RR=0.80, 95% CI, 0.69-0.94) for those receiving calcium 1,200 to 2,000 mg/d, but no effect in general populations (RR=0.62, 95% CI, 0.11-3.40) (59). It should be noted that the statistical power in the two clinical trials conducted in general populations is limited (59).

In 1980, Garland hypothesized that lower levels of vitamin D resulting from weaker UV-B radiation at higher latitudes may account for the striking geographical pattern of CRC mortality (60). The evidence from epidemiologic studies are limited to small sample sizes, yet in general found vitamin D inversely associated with CRC risk (57,61-64). Meta-analyses of longitudinal studies reported a 50% lower CRC risk associated with a serum 25(OH)D level ≥33 ng/mL, compared to ≤12 ng/mL (65), or an OR of 0.57 (95% CI, 0.43-0.76) for an increase of 25(OH)D by 20 ng/mL (66).

Selenium

As an essential trace element involved in different physiological functions in the human body, selenium has received increasing attention as a possible cancer prevention substance, through several possible mechanisms for the potential anticarcinogenic effects including apoptosis (67), protection from oxidative DNA damage (68), and increased immune function (69). However, the epidemiologic evidence on associations between selenium and CRC risk has been mixed, inconclusive and limited to small studies. Many observational studies support a protective effect of selenium on CRC risk (70-72) but not consistently (73,74). A meta-analysis of 12 observational studies and 2 clinical trials reported no association between selenium and CRC risk in women, but an inverse association in men (OR=0.68, 95% CI, 0.57-0.82) (75). There was a large randomized trial on selenium supplementation for skin cancer prevention, and the secondary analysis showed a 58% reduction (95% CI, 0.18-0.95) in CRC incidence among participants randomly assigned to take selenium (200 μg daily) (76), although the results were attenuated and no longer statistically significant after additional years of follow- up (77). A pooled analysis of 3 randomized trials on various nutritional interventions for CRC prevention reported a pooled OR of 0.66 (95% CI, 0.50-0.87), contrasting participants of highest vs. lowest blood selenium values (78). Yet, none of these trials were designed to test selenium as an intervention, and all participants were at high adenoma risk after recent colonoscopic adenoma resection (79).

Magnesium

Magnesium is abundant in many foods, and particularly rich in spices, nuts, cereals, coffee, cocoa, tea and green-leafy vegetables. Magnesium is involved in a wide variety of biochemical reactions that modulate key cell functions, and has a crucial role in genomic stability and DNA synthesis (80). Epidemiologic studies suggested that magnesium may be associated with a decreased CRC risk but the findings are inconsistent. A meta-analysis of 8 prospective studies containing 338,979 participants and 8,000 CRC cases reported a summary RR of 0.89 (95% CI, 0.79-1.00) for the highest vs. lowest category of magnesium intake, with evidence of a dose-response (81).

Dietary pattern, obesity and related mechanisms

Dietary pattern summarizes the total diet or the key dietary components including food items, food groups, and nutrients (82), and may provide additional insights with the combined effects of many food components. Summarized epidemiologic evidence suggest that healthier pattern consisting of greater intakes of fruits and vegetables, and lower intakes of red and processed meat, appeared protective against colorectal adenoma and cancer incidence, while a less healthy pattern characterized by higher intakes of red and processed meat, as well as potatoes and refined carbohydrates, may increase risk (83). There has also been great interest in total dietary consumption and its consequences, specifically obesity, in relation to colorectal and other cancers (84). Because these risks are modifiable, these pathways are seen as key in cancer control efforts (85). Colon cancer is among those specifically identified as linked to obesity; for example, two large prospective cohort studies have demonstrated that being obese [body mass index (BMI) >30 kg/m] confers a 1.5-fold greater risk of developing colon cancer relative to individuals of normal weight (BMI 18.5-24.9 kg/m) (86,87). With respect to mortality, the Cancer Prevention Study II, which prospectively followed more than 900,000 US adults, similarly found a 1.5- to 1.8-fold increased risk of colon cancer related death for obese individuals, reflecting the increased incidence of and perhaps the increased mortality from colon cancer once diagnosed (88). A meta-analysis of 31 observational studies reported that for a 2 kg/m increase in BMI, the CRC risk increased by 7% (95% CI, 4-10%), and for a 2-cm increase in waist circumference, the risk increased by 4% (95% CI, 2-5%) (89). Another meta-analysis showed that the inverse association between BMI and CRC risk was stronger for colon than rectal cancer, and for men than women (90). Some recent research probing obesity pathways concern the issue of ‘secondary prevention’ after diagnosis and treatment of early stage disease. Following earlier studies by Meyerhardt et al. (91) and Dignam et al. (92), which found excess risk of both cancer recurrence and overall mortality among obese patients utilizing large participant cohorts from multicenter clinical trials for stage II and III colon cancer, subsequent studies turned to elucidating the role of dietary constituents among colon cancer patients. The first of these studies examined the ‘Western’ diet pattern characterized by relatively high red and processed meats, refined grain, and sugar content, and indicated that those with higher consumption of these components experienced significantly elevated recurrence and mortality risk (93). A more recent study more closely examined the Western diet and the role of dietary glycemic measures (94). This study found that high carbohydrate intake and glycemic load significantly increased recurrence and mortality. These effects were present among all body types but strongest among those who were overweight or obese. In relation to CRC risk, the Western dietary pattern has previously been reported to be associated with an elevated CRC incidence (95). However, the Women's Health Initiative Dietary Modification Trial, a large randomized trial conducted in postmenopausal women, suggested that a low-fat dietary pattern intervention did not reduce CRC risk during 8 years of follow-up (96).

This work relates to a large body of research focused on potential mechanisms by which obesity leads to higher colon cancer incidence. One prominent theme involves metabolic and insulin related pathways (97-104). Insulin, insulin-like growth factors (IGF), and IGF binding proteins have all been causatively implicated for colon cancer via mitogenic effects on the colonic mucosa and other mechanisms. In a prospective nested case-control study within the Physicians’ Health Study, Ma et al. (101) found that men in the highest quintile for IGF-1 had a 2.5 increased risk of colorectal cancer compared to the lowest quintile. Among women, an analysis of the Nurse's Health Study cohort found a comparable >2-fold for those in the highest quartile of IGF-1 compared with those in the lowest (102). That study, as well as investigations by Kaaks et al. (103) and a more recent study by Ma et al. (104) also observed that colorectal cancer risk increased with increasing levels of C-peptide (a marker of insulin production). Interestingly, reduction in IGF exposure via increased presence of its binding protein was found to significantly reduce colon cancer mortality among affected individuals in a large cohort study (105). This particular pathway represents but one of numerous putative links linking cancer and obesity (84).

Obviously, a critical component of obesity is physical activity. The association between physical activity and CRC risk is well established, with the majority of relevant studies finding physically active individuals benefit from substantial risk reduction (approximately 20%) comparing to the sedentary ones, confirmed by two meta-analyses (106,107). The evidence is strong and consistent for both proximal colon and distal colon cancers with a similar magnitude (107). The main challenge is disentangling the direct effects of physical activity from strongly related factors such as obesity and comorbidities that may influence outcomes. Randomized intervention trials have been completed with positive findings, and others are underway; these studies have been mostly focused on colorectal cancer survivors (clinical event reduction) or biomarkers (prevention studies) for practical reasons (108-110).

Red meat

Whether red meat is a culprit in causing CRC has been the subject of scientific debate. Extensive evidence suggests that long-term consumption of red meat or processed meats may increase CRC risk (9-11). A recent meta-analysis of 21 prospective studies showed that consumption of red and processed meats was associated with elevated CRC risk [relative risk, RR=1.22, 95% confidence interval (CI), 1.11-1.34, contrasting the highest versus lowest quartiles of intake], with a linear increase of CRC (RR=1.14, 95% CI, 1.04-1.24) associated with every 100 g/day increase of meat intake until a plateau was encountered at 140 g/day (12). Another meta-analysis of 25 prospective studies that same year found the association was similar by tumor site (colon cancer: RR=1.11, 95% CI, 1.03-1.19, and rectal cancer: RR=1.19, 95% CI, 0.97-1.46) but differed between men (RR=1.21, 95% CI, 1.04-1.42) and women (RR=1.01, 95% CI, 0.87-1.17) (13). Another meta-analysis of both cohort and case-control studies (22 in total) reported that the frequency of red meat consumption rather than total amount of consumed meat is associated with a higher CRC carcinogenesis (14). In addition, the processing method may matter. Very well-cooked meat or meats cooked in direct contact with flames were reported to raise the CRC risk, which may be explained by the carcinogenic heterocyclic amines produced during cooking (15,16). Other plausible biological mechanisms include endogenous formation of nitroso compounds in the gastrointestinal tract by red meat intake, but not by white meat intake (17).

Fruit and vegetables (fiber and folic acid)

Many observational studies have reported an inverse association between dietary fiber and CRC risk, with a relative reduction of up to 40% (18-22), although a few large cohort studies reported small, statistically null associations (23-25). A large pooled analysis of thirteen prospective cohorts suggested that dietary fiber intake was inversely associated with CRC risk in age-adjusted analyses, but no association remained after accounting for other dietary risk factors, including red meat, alcohol, folate, and total milk (26). A recent meta-analysis reported a dose-response analysis indicating that for each 10 g/day total dietary fiber, the relative risk of developing CRC was 0.90 (95% CI, 0.86-0.94) (27). However, two randomized trials found that high dietary fiber did not affect the recurrence of colorectal adenoma (28,29).

Dietary folate or folic acid (from dietary supplements and food fortification) is necessary to synthesize, repair and methylate DNA. It is especially important during periods of rapid cell division and growth such as pregnancy and infancy. Folate is also thought to help prevent changes to DNA that may lead to cancer, and its rule in CRC carcinogenesis has been extensively studied. Humans cannot synthesize folate de novo, and therefore folate has to be supplied through diet to meet their daily requirements, with fresh fruits and vegetables being the major sources. There are growing data and a continuing controversy over the effect of folic acid supplementation on cancer risk. Both the Nurse's Health Study and secondary analysis from the Wheat Bran Fiber randomized trial reported that high dietary folate was associated with 40% reduced risk of colorectal adenoma (highest vs. lowest quartile) (30,31). The Nurses’ Health Study and Health Professionals Follow-Up Study further suggested that both long- and short-term intakes of total folate were associated with a lower risk of colorectal adenoma (32). With respect to dose, one case-control study based on suspected patients undergoing screening reported that the minimal essential serum folate concentrations should be above 8.0 ng/mL for reducing risk of developing colorectal adenoma (33). One meta-analysis summarized 3 randomized or pseudo-randomized trials and concluded that folate status was inversely related to the risk of developing CRC (34); however, a recently published larger meta-analysis of 15 randomized trials reported that folic acid supplementation has no significant effect on CRC risk (35). A meta-analysis of 3 large randomized trials of folic acid supplementation among patients with an adenoma history also reported no association (36).

Garlic

Garlic is characterized by a high content in organo-sulfur compounds and flavonoids. The allyl sulfur constituents in garlic comprise about 1% of its dry weight and are responsible for its health benefits (37,38). Garlic also contains other constituents such as flavonoids and selenium that are considered to have antioxidant properties and anticarcinogenic activity. Preclinical investigations provide convincing evidence that garlic and related sulfur-containing compounds inhibit carcinogen-induced tumors in various organs (39). One RCT conducted within a small sample of 37 patients with CRC found (I) a significant suppression in both the total size and number of adenomas in CRC patients after 12-mo of high-dose aged garlic extract (40), and (II) a 29% reduction in developing at least one new adenoma (41). A meta-analysis of 4 case control and 3 cohort studies confirmed the inverse association between garlic intake and CRC risk, with an approximate 30% relative reduction in incidence contrasting high vs. low (16 g/wk average difference across studies) consumption of garlic (42).

Fish and Omega-3 fatty acids

Long-chain n-3 fatty acids have been suggested to play a protective role in colorectal cancer development in laboratory and animal studies, with the mechanism of action conjectured to be inhibition of the cyclooxygenase-2 (COX-2) enzyme and the production of arachidonic acid (n-6) derived eicosanoids (40-42). A long-term prospective study of U.S. men reported a significantly reduced CRC risk contrasting the highest versus lowest quartiles of n-3 fatty acids (odds ratio, OR=0.74, 95% CI, 0.57-0.95) (43). Fish is the main dietary source of the long-chain n-3 fatty acids. Some observational studies found an inverse association between fish consumption and CRC risk (10,43-45), while others did not (46-48). A recent meta-analysis pooled data from 22 cohort and 19 case-control studies, and reported a 12% (OR=0.88, 95% CI, 0.80-0.95) reduction in CRC risk contrasting highest vs. lowest fish consumption (49). However, a multi-center, randomized controlled trial investigated the effect of 6-month intervention with oil-rich or lean fish on apoptosis and mitosis within the colonic crypt, and found no marked change in these endpoints (50).

Vitamin B6

Vitamin B6 is widely distributed in foods, with good sources including meats, whole grain products, vegetables, nuts and bananas. Vitamin B6 is involved in almost 100 enzymatic reactions, among which one function involves transferring 1-carbon groups for DNA synthesis and methylation (51). Therefore, vitamin B6 deficiency may increase CRC risk through aberrations in DNA synthesis, repair, and methylation. Vitamin B may also suppress colorectal carcinogenesis by reducing cell proliferation, angiogenesis, oxidative stress, inflammation, and nitric oxide synthesis (52,53). A meta-analysis summarized evidence from prospective studies, with 9 studies on vitamin B6 intake and 4 studies on blood pyridoxal 5’-phosphate (PLP, the principal active coenzyme form of vitamin B6) levels, in relation to CRC risk. The pooled RRs of CRC contrasting highest vs. lowest category of vitamin B6 intake and blood PLP levels were 0.90 (95% CI, 0.75-1.07) and 0.52 (95% CI, 0.38-0.71), respectively (54).

Dairy, calcium and vitamin D

In vitro and in vivo studies have suggested that high dietary intake of calcium and vitamin D may reduce CRC risk by a variety of mechanisms, such as reducing epithelial cell exposure to toxicity, inhibiting proliferation of intestinal mucosa and epithelial cells via the intracellular action of calcium (55); Vitamin D's protective effect against colorectal neoplasia is by reducing epithelial cell proliferation (56). There have been numerous epidemiological studies on calcium and CRC risk, as well as studies on vitamin D and CRC risk. However, there remains a lack of clarity of the effects on CRC of both these dietary components, due to inconsistent findings among numerous epidemiological studies and multiple pooled analyses. Overall, results from observational studies found calcium intake to be not associated with a substantially lower CRC risk (57,58) particularly for the more reliable large prospective studies (57). A meta-analysis of summarizing a large number of observational studies mostly supported an inverse association for both calcium and milk/dairy products (a rich source of both calcium and vitamin D) (59,60). The evidence from randomized trials only shows a protective effect of calcium on adenoma recurrence among individuals with a history of adenomas (RR=0.80, 95% CI, 0.69-0.94) for those receiving calcium 1,200 to 2,000 mg/d, but no effect in general populations (RR=0.62, 95% CI, 0.11-3.40) (59). It should be noted that the statistical power in the two clinical trials conducted in general populations is limited (59).

In 1980, Garland hypothesized that lower levels of vitamin D resulting from weaker UV-B radiation at higher latitudes may account for the striking geographical pattern of CRC mortality (60). The evidence from epidemiologic studies are limited to small sample sizes, yet in general found vitamin D inversely associated with CRC risk (57,61-64). Meta-analyses of longitudinal studies reported a 50% lower CRC risk associated with a serum 25(OH)D level ≥33 ng/mL, compared to ≤12 ng/mL (65), or an OR of 0.57 (95% CI, 0.43-0.76) for an increase of 25(OH)D by 20 ng/mL (66).

Selenium

As an essential trace element involved in different physiological functions in the human body, selenium has received increasing attention as a possible cancer prevention substance, through several possible mechanisms for the potential anticarcinogenic effects including apoptosis (67), protection from oxidative DNA damage (68), and increased immune function (69). However, the epidemiologic evidence on associations between selenium and CRC risk has been mixed, inconclusive and limited to small studies. Many observational studies support a protective effect of selenium on CRC risk (70-72) but not consistently (73,74). A meta-analysis of 12 observational studies and 2 clinical trials reported no association between selenium and CRC risk in women, but an inverse association in men (OR=0.68, 95% CI, 0.57-0.82) (75). There was a large randomized trial on selenium supplementation for skin cancer prevention, and the secondary analysis showed a 58% reduction (95% CI, 0.18-0.95) in CRC incidence among participants randomly assigned to take selenium (200 μg daily) (76), although the results were attenuated and no longer statistically significant after additional years of follow- up (77). A pooled analysis of 3 randomized trials on various nutritional interventions for CRC prevention reported a pooled OR of 0.66 (95% CI, 0.50-0.87), contrasting participants of highest vs. lowest blood selenium values (78). Yet, none of these trials were designed to test selenium as an intervention, and all participants were at high adenoma risk after recent colonoscopic adenoma resection (79).

Magnesium

Magnesium is abundant in many foods, and particularly rich in spices, nuts, cereals, coffee, cocoa, tea and green-leafy vegetables. Magnesium is involved in a wide variety of biochemical reactions that modulate key cell functions, and has a crucial role in genomic stability and DNA synthesis (80). Epidemiologic studies suggested that magnesium may be associated with a decreased CRC risk but the findings are inconsistent. A meta-analysis of 8 prospective studies containing 338,979 participants and 8,000 CRC cases reported a summary RR of 0.89 (95% CI, 0.79-1.00) for the highest vs. lowest category of magnesium intake, with evidence of a dose-response (81).

Dietary pattern, obesity and related mechanisms

Dietary pattern summarizes the total diet or the key dietary components including food items, food groups, and nutrients (82), and may provide additional insights with the combined effects of many food components. Summarized epidemiologic evidence suggest that healthier pattern consisting of greater intakes of fruits and vegetables, and lower intakes of red and processed meat, appeared protective against colorectal adenoma and cancer incidence, while a less healthy pattern characterized by higher intakes of red and processed meat, as well as potatoes and refined carbohydrates, may increase risk (83). There has also been great interest in total dietary consumption and its consequences, specifically obesity, in relation to colorectal and other cancers (84). Because these risks are modifiable, these pathways are seen as key in cancer control efforts (85). Colon cancer is among those specifically identified as linked to obesity; for example, two large prospective cohort studies have demonstrated that being obese [body mass index (BMI) >30 kg/m] confers a 1.5-fold greater risk of developing colon cancer relative to individuals of normal weight (BMI 18.5-24.9 kg/m) (86,87). With respect to mortality, the Cancer Prevention Study II, which prospectively followed more than 900,000 US adults, similarly found a 1.5- to 1.8-fold increased risk of colon cancer related death for obese individuals, reflecting the increased incidence of and perhaps the increased mortality from colon cancer once diagnosed (88). A meta-analysis of 31 observational studies reported that for a 2 kg/m increase in BMI, the CRC risk increased by 7% (95% CI, 4-10%), and for a 2-cm increase in waist circumference, the risk increased by 4% (95% CI, 2-5%) (89). Another meta-analysis showed that the inverse association between BMI and CRC risk was stronger for colon than rectal cancer, and for men than women (90). Some recent research probing obesity pathways concern the issue of ‘secondary prevention’ after diagnosis and treatment of early stage disease. Following earlier studies by Meyerhardt et al. (91) and Dignam et al. (92), which found excess risk of both cancer recurrence and overall mortality among obese patients utilizing large participant cohorts from multicenter clinical trials for stage II and III colon cancer, subsequent studies turned to elucidating the role of dietary constituents among colon cancer patients. The first of these studies examined the ‘Western’ diet pattern characterized by relatively high red and processed meats, refined grain, and sugar content, and indicated that those with higher consumption of these components experienced significantly elevated recurrence and mortality risk (93). A more recent study more closely examined the Western diet and the role of dietary glycemic measures (94). This study found that high carbohydrate intake and glycemic load significantly increased recurrence and mortality. These effects were present among all body types but strongest among those who were overweight or obese. In relation to CRC risk, the Western dietary pattern has previously been reported to be associated with an elevated CRC incidence (95). However, the Women's Health Initiative Dietary Modification Trial, a large randomized trial conducted in postmenopausal women, suggested that a low-fat dietary pattern intervention did not reduce CRC risk during 8 years of follow-up (96).

This work relates to a large body of research focused on potential mechanisms by which obesity leads to higher colon cancer incidence. One prominent theme involves metabolic and insulin related pathways (97-104). Insulin, insulin-like growth factors (IGF), and IGF binding proteins have all been causatively implicated for colon cancer via mitogenic effects on the colonic mucosa and other mechanisms. In a prospective nested case-control study within the Physicians’ Health Study, Ma et al. (101) found that men in the highest quintile for IGF-1 had a 2.5 increased risk of colorectal cancer compared to the lowest quintile. Among women, an analysis of the Nurse's Health Study cohort found a comparable >2-fold for those in the highest quartile of IGF-1 compared with those in the lowest (102). That study, as well as investigations by Kaaks et al. (103) and a more recent study by Ma et al. (104) also observed that colorectal cancer risk increased with increasing levels of C-peptide (a marker of insulin production). Interestingly, reduction in IGF exposure via increased presence of its binding protein was found to significantly reduce colon cancer mortality among affected individuals in a large cohort study (105). This particular pathway represents but one of numerous putative links linking cancer and obesity (84).

Obviously, a critical component of obesity is physical activity. The association between physical activity and CRC risk is well established, with the majority of relevant studies finding physically active individuals benefit from substantial risk reduction (approximately 20%) comparing to the sedentary ones, confirmed by two meta-analyses (106,107). The evidence is strong and consistent for both proximal colon and distal colon cancers with a similar magnitude (107). The main challenge is disentangling the direct effects of physical activity from strongly related factors such as obesity and comorbidities that may influence outcomes. Randomized intervention trials have been completed with positive findings, and others are underway; these studies have been mostly focused on colorectal cancer survivors (clinical event reduction) or biomarkers (prevention studies) for practical reasons (108-110).

Other common lifestyle exposures

Alcohol

Alcohol consumption is one of the most important known causes of human cancer, possibly through genotoxic effect of acetaldehyde (111) and interference of folate absorption by alcohol (112). The most recent meta-analysis pooled data from 27 cohort and 34 case-control studies, and reported significantly increased CRC risk with a dose-response relationship (113): comparing to never drinkers, moderate drinkers who consume 2-3 drinks per day have 21% increased CRC risk and heavy drinkers who have 4 or more drinks per day have 52% increased CRC risk. These results are consistent with other pooled analyses, with increased risk observed for both colon and rectal cancers (114-116).

Smoking

Cigarette smoking has been shown to cause many nonpulmonary cancers with no direct tobacco-related carcinogens, yet the association between smoking and CRC remains controversial. A meta-analysis of 106 observational studies estimated that cigarette smokers are 1.18 times (95% CI, 1.11-1.25 times) more likely to develop CRC compared to those who never smoke, with evidence of a dose-response (117). Another meta-analysis of 36 studies reported that duration and age of initiation were also significantly associated with CRC incidence (118). A recent study reported that the excess CRC risk due to smoking decreased immediately after quitting for proximal colon and rectal cancer, but not until about 20 years post-quitting for distal colon cancer (119).

Hormone replacement therapy (HRT)

Estrogen/progestin replacement therapy is prescribed to control postmenopausal symptom or to prevent hormone deficiency-related diseases such as osteoporosis. Existing evidence shows that HRT is associated with lower CRC risk, yet the mechanism remained unclear. Case control studies have shown significant reduction in CRC risk among ever users (120,121). Large cohort studies suggested a modest risk reduction among ever users (122), but there was a lack of dose-response relationship (123). Also, the association was found stronger among women aged 65+, with a body mass index <30 and who regularly use aspirin or ibuprofen (124). A meta-analysis concluded that recent HRT users had a 33% reduction in CRC risk (pooled RR=0.67, 95% CI, 0.59-0.77) but no association was observed with ever use of HRT and duration of use did not matter (125). Other studies also supported that the protective effect of HRT is short-lived and disappears following cessation of use (124). The Women's Health Initiative trial found that women who took estrogen plus progestin had 44% reduced risk (95% CI, 19-62%) of colorectal cancer compared to women who took placebo but colorectal cancers in the estrogen plus progestin group were at a more advanced stage (126). The Women's Health Initiative trial later reported that there was no significant difference in colorectal cancer incidence between the estrogen-only and placebo arms (127).

Alcohol

Alcohol consumption is one of the most important known causes of human cancer, possibly through genotoxic effect of acetaldehyde (111) and interference of folate absorption by alcohol (112). The most recent meta-analysis pooled data from 27 cohort and 34 case-control studies, and reported significantly increased CRC risk with a dose-response relationship (113): comparing to never drinkers, moderate drinkers who consume 2-3 drinks per day have 21% increased CRC risk and heavy drinkers who have 4 or more drinks per day have 52% increased CRC risk. These results are consistent with other pooled analyses, with increased risk observed for both colon and rectal cancers (114-116).

Smoking

Cigarette smoking has been shown to cause many nonpulmonary cancers with no direct tobacco-related carcinogens, yet the association between smoking and CRC remains controversial. A meta-analysis of 106 observational studies estimated that cigarette smokers are 1.18 times (95% CI, 1.11-1.25 times) more likely to develop CRC compared to those who never smoke, with evidence of a dose-response (117). Another meta-analysis of 36 studies reported that duration and age of initiation were also significantly associated with CRC incidence (118). A recent study reported that the excess CRC risk due to smoking decreased immediately after quitting for proximal colon and rectal cancer, but not until about 20 years post-quitting for distal colon cancer (119).

Hormone replacement therapy (HRT)

Estrogen/progestin replacement therapy is prescribed to control postmenopausal symptom or to prevent hormone deficiency-related diseases such as osteoporosis. Existing evidence shows that HRT is associated with lower CRC risk, yet the mechanism remained unclear. Case control studies have shown significant reduction in CRC risk among ever users (120,121). Large cohort studies suggested a modest risk reduction among ever users (122), but there was a lack of dose-response relationship (123). Also, the association was found stronger among women aged 65+, with a body mass index <30 and who regularly use aspirin or ibuprofen (124). A meta-analysis concluded that recent HRT users had a 33% reduction in CRC risk (pooled RR=0.67, 95% CI, 0.59-0.77) but no association was observed with ever use of HRT and duration of use did not matter (125). Other studies also supported that the protective effect of HRT is short-lived and disappears following cessation of use (124). The Women's Health Initiative trial found that women who took estrogen plus progestin had 44% reduced risk (95% CI, 19-62%) of colorectal cancer compared to women who took placebo but colorectal cancers in the estrogen plus progestin group were at a more advanced stage (126). The Women's Health Initiative trial later reported that there was no significant difference in colorectal cancer incidence between the estrogen-only and placebo arms (127).

Chemoprevention of colorectal cancer

Prevention of colorectal cancer and its precursor conditions via some substance or compound, or chemoprevention, offers great potential if the intervention is safe and cost-effective. The main focus of these has not been nutritional, but rather medicinal agents with reasonably safe adverse event profiles. For more than two decades, accumulating evidence from observational studies (128-133) and large randomized trials (134-137) suggests that aspirin and other non-steroidal anti-inflammatory drugs (NSAIDs) protect against the development of colonic adenomas and CRC, and reduces disease recurrence (136-139). Meta-analysis of 17 case-control studies showed an inverse association between regular use of aspirin and reduced CRC risk (pooled OR=0.62, 95% CI, 0.58-0.67), while the association was weaker in cohort studies presumably due to substantial variation between studies in measuring aspirin exposure (140). Evidence from randomized trials has not been consistent. Two trials conducted among patients with a history of CRC or adenomas showed that low-dose aspirin has a chemopreventive effect on new adenomas (136,137). Yet among healthy individuals, two earlier large trials of low-dose aspirin (the Physicians’ Health Study and the Women's Health Study) showed no effect on CRC incidence during 10-year follow-up, possibly due to an insufficient dose (134,141,142). Recent long-term follow-up of four aspirin trials (Primary: Thrombosis Prevention Trial, British Doctors Aspirin Trial; Secondary: Swedish Aspirin Low Dose Trial, UK-TIA Aspirin Trial) showed a reduction of 50% in CRC risk after five or more years of regular consumption with a dosage of at least 75 mg daily (75-300 mg) (143). Aspirin has emerged as the most likely NSAID for use in chemoprevention because of its known cardiovascular benefit and available safety and efficacy data (144). Other traditional NSAIDs, particularly selective COX-2 inhibitors such as celecoxib, have been shown to cause regression of adenomas in familial adenomatous polyposis, and are now given to patients at high risk of CRC (138,139), and yet cannot be routinely recommended concerning potential cardiovascular events. The Colorectal Adenoma/carcinoma Prevention Programme (CAPP) was launched in 1990, with CAPP1 investigating familial adenomatous polyposis in 200 young adults and CAPP2 being the first large-scale genetically targeted chemoprevention trial in 1,000 adults with HNPCC (also known as Lynch syndrome). The CAPP2 randomized trial reported that 600 mg aspirin per day for a mean of 25 months substantially reduced CRC incidence among carriers of hereditary CRC (145). However, the minimum dose of aspirin to achieve the protective effect is still uncertain and will be the objective of a new CAPP trial in Lynch Syndrome. The underlying mechanism of NSAIDs inhibiting carcinogenesis remains inconclusive, with proposed explanations as increased apoptosis and impairment of tumor cell growth by inhibition of cyclooxygenase-2 (COX-2) (146).

The side-effects of NSAIDs are well documented and mainly attributed to inhibition of COX activity. The most frequently reported serious adverse events associated with aspirin use are related to gastrointestinal bleeding, even with low-dose aspirin (147). Comparing to other NSAIDs, aspirin has lower risk of occlusive cardiovascular events, yet the dose-dependent risk of bleeding complications with aspirin intake may limit its potential for primary prevention of CRC (148). In additional to optimal dose, optimum treatment duration and age of initiation also remained uncertain (144). Concerning risk and benefit, the United States Preventive Services Task Force recommended against the routine use of aspirin for CRC prevention in 2007 (149). However, the 2011 pooled analysis with 8 randomized controlled trials showed that daily aspirin for 5-10 years reduced 5-year cancer mortality by 34% (P=0.03) and a 20-year cancer mortality by 20% (P<0.001) (150). Considering the bleeding complications are not life-threatening, the risk-benefit of aspirin use in CRC prevention requires a formal re-evaluation. As toxicity of aspirin largely depends on dosage, the minimal effective dose required for CRC prevention is critical (151).

Conclusions

It has been estimated that dietary factors account for nearly half of all colorectal cancer cases, and therefore diet and lifestyle are key intervention points in primary prevention. This review summarized and updated the relevant epidemiologic evidence for dietary constituents and other modifiable factors, some of which are likely correlated with diet behavior. There is convincing, or moderately convincing and rather consistent, evidence that intakes of garlic, vitamin B6 and magnesium, maintaining a healthy weight and waist via both diet and exercise, and avoiding or reducing red meat, alcohol, and smoking, may significantly protect against developing colorectal cancer. There is high quality yet less consistent evidence for the following dietary components: fruit and vegetable intake (fiber and folate), fish and Omega-3 fatty acids, selenium, dairy, calcium and vitamin D. For high risk populations for whom dietary and lifestyle interventions may not be sufficient for primary prevention, aspirin and other chemoprevention agents may be considered, although the minimal effective dose remains unclear. Ongoing studies may shed light on additional safe interventions that can reduce colorectal cancer and will likely have other health benefits.

Acknowledgements

This work was supported by Public Health Service grant NCI P30-CA-14599 from the National Cancer Institute, United States National Institutes of Health.

Department of Health Studies, University of Chicago, Chicago, IL, USA
Corresponding to: James J. Dignam. Department of Health Studies, University of Chicago, Chicago, IL, USA. ude.ogacihcu.dsb.htlaeh@mangidj.

Abstract

Objectives

This systematic review focuses on dietary and lifestyle risk factors for colorectal cancer (CRC) prevention and chemoprevention among high-risk populations.

Methods and materials

We searched PubMed for English-language articles about dietary components, lifestyle risk factors, and chemoprevention agents in relation to colorectal cancer and their references published from 1980 through 2013. We reviewed articles jointly for the most clinically important information, emphasizing randomized trials and meta-analyses where available.

Results

There is convincing evidence that intake of garlic, vitamin B6 and magnesium, active living, maintaining a healthy weight and waist, avoiding or reducing red meat, alcohol, and smoking, as well as hormone replacement therapy among women may significantly protect against developing colorectal cancer. There is less consistent evidence for fruit and vegetable intake (fiber and folate), fish and Omega-3 fatty acids, selenium, dairy, calcium and vitamin D. For high-risk populations, aspirin have been shown to protect against the development of colonic adenomas and CRC, while a minimal effective dose remains unclear.

Conclusions

Colorectal cancer can be prevented in general population through dietary and lifestyle interventions, and aspirin may be a good choice of chemoprevention agent among high risk individuals.

Keywords: Colorectal cancer (CRC), dietary, lifestyle, prevention, chemoprevention, aspirin, high risk individuals, meta-analyses
Abstract

Footnotes

Cite this article as: Hou N, Huo D, Dignam JJ. Prevention of colorectal cancer and dietary management. Chin Clin Oncol 2013;2(2):13. doi: 10.3978/j.issn.2304-3865.2013.04.03

Disclosure: The authors declare no conflict of interest.

Footnotes

References

  • 1. Weitz J, Koch M, Debus J, et al Colorectal cancer. Lancet. 2005;365:153–65.[PubMed][Google Scholar]
  • 2. Jemal A, Bray F, Center MM, et al Global cancer statistics. CA Cancer J Clin. 2011;61:69–90.[PubMed][Google Scholar]
  • 3. Center MM, Jemal A, Ward EInternational trends in colorectal cancer incidence rates. Cancer Epidemiol Biomarkers Prev. 2009;18:1688–94.[PubMed][Google Scholar]
  • 4. Doll R, Peto RThe causes of cancer: quantitative estimates of avoidable risks of cancer in the United States today. J Natl Cancer Inst. 1981;66:1191–308.[PubMed][Google Scholar]
  • 5. Giovannucci E, Stampfer MJ, Colditz G, et al Relationship of diet to risk of colorectal adenoma in men. J Natl Cancer Inst. 1992;84:91–8.[PubMed][Google Scholar]
  • 6. Flood DM, Weiss NS, Cook LS, et al Colorectal cancer incidence in Asian migrants to the United States and their descendants. Cancer Causes Control. 2000;11:403–11.[PubMed][Google Scholar]
  • 7. Kune GA, Bannerman S, Watson LFAttributable risk for diet, alcohol, and family history in the Melbourne Colorectal Cancer Study. Nutr Cancer. 1992;18:231–5.[PubMed][Google Scholar]
  • 8. Shike MDiet and lifestyle in the prevention of colorectal cancer: an overview. Am J Med. 1999;106:11S–15S. discussion 50S-51S. [[PubMed][Google Scholar]
  • 9. Chao A, Thun MJ, Connell CJ, et al Meat consumption and risk of colorectal cancer. JAMA. 2005;293:172–82.[PubMed][Google Scholar]
  • 10. Norat T, Bingham S, Ferrari P, et al Meat, fish, and colorectal cancer risk: the European Prospective Investigation into cancer and nutrition. J Natl Cancer Inst. 2005;97:906–16.[Google Scholar]
  • 11. Cross AJ, Ferrucci LM, Risch A, et al A large prospective study of meat consumption and colorectal cancer risk: an investigation of potential mechanisms underlying this association. Cancer Res. 2010;70:2406–14.[Google Scholar]
  • 12. Chan DS, Lau R, Aune D, et al Red and processed meat and colorectal cancer incidence: meta-analysis of prospective studies. PLoS One. 2011;6:e20456.[Google Scholar]
  • 13. Alexander DD, Weed DL, Cushing CA, et al Meta-analysis of prospective studies of red meat consumption and colorectal cancer. Eur J Cancer Prev. 2011;20:293–307.[PubMed][Google Scholar]
  • 14. Smolińska K, Paluszkiewicz P. Risk of colorectal cancer in relation to frequency and total amount of red meat consumption. Systematic review and meta-analysis. Arch Med Sci. 2010;6:605–10.
  • 15. Sinha R, Chow WH, Kulldorff M, et al Well-done, grilled red meat increases the risk of colorectal adenomas. Cancer Res. 1999;59:4320–4.[PubMed][Google Scholar]
  • 16. Ferrucci LM, Sinha R, Huang WY, et al Meat consumption and the risk of incident distal colon and rectal adenoma. Br J Cancer. 2012;106:608–16.[Google Scholar]
  • 17. Bingham SA, Hughes R, Cross AJEffect of white versus red meat on endogenous N-nitrosation in the human colon and further evidence of a dose response. J Nutr. 2002;132:3522S–3525S.[PubMed][Google Scholar]
  • 18. Levi F, Pasche C, Lucchini F, et al Dietary fibre and the risk of colorectal cancer. Eur J Cancer. 2001;37:2091–6.[PubMed][Google Scholar]
  • 19. Bingham SA, Day NE, Luben R, et al. European Prospective Investigation into Cancer and Nutrition. Dietary fibre in food and protection against colorectal cancer in the European Prospective Investigation into Cancer and Nutrition (EPIC): an observational study. Lancet. 2003;361:1496–501.[PubMed]
  • 20. Terry P, Giovannucci E, Michels KB, et al Fruit, vegetables, dietary fiber, and risk of colorectal cancer. J Natl Cancer Inst. 2001;93:525–33.[PubMed][Google Scholar]
  • 21. Peters U, Sinha R, Chatterjee N, et al. Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial Project Team. Dietary fibre and colorectal adenoma in a colorectal cancer early detection programme. Lancet. 2003;361:1491–5.[PubMed]
  • 22. Dahm CC, Keogh RH, Spencer EA, et al Dietary fiber and colorectal cancer risk: a nested case-control study using food diaries. J Natl Cancer Inst. 2010;102:614–26.[PubMed][Google Scholar]
  • 23. Mai V, Flood A, Peters U, et al Dietary fibre and risk of colorectal cancer in the Breast Cancer Detection Demonstration Project (BCDDP) follow-up cohort. Int J Epidemiol. 2003;32:234–9.[PubMed][Google Scholar]
  • 24. Schatzkin A, Mouw T, Park Y, et al Dietary fiber and whole-grain consumption in relation to colorectal cancer in the NIH-AARP Diet and Health Study. Am J Clin Nutr. 2007;85:1353–60.[PubMed][Google Scholar]
  • 25. Fuchs CS, Giovannucci EL, Colditz GA, et al Dietary fiber and the risk of colorectal cancer and adenoma in women. N Engl J Med. 1999;340:169–76.[PubMed][Google Scholar]
  • 26. Park Y, Hunter DJ, Spiegelman D, et al Dietary fiber intake and risk of colorectal cancer: a pooled analysis of prospective cohort studies. JAMA. 2005;294:2849–57.[PubMed][Google Scholar]
  • 27. Aune D, Chan DS, Lau R, et al Dietary fibre, whole grains, and risk of colorectal cancer: systematic review and dose-response meta-analysis of prospective studies. BMJ. 2011;343:d6617.[Google Scholar]
  • 28. 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. 2000;342:1156–62.[PubMed]
  • 29. 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. 2000;342:1149–55.[PubMed]
  • 30. Giovannucci E, Stampfer MJ, Colditz GA, et al Folate, methionine, and alcohol intake and risk of colorectal adenoma. J Natl Cancer Inst. 1993;85:875–84.[PubMed][Google Scholar]
  • 31. Martínez ME, Henning SM, Alberts DSFolate and colorectal neoplasia: relation between plasma and dietary markers of folate and adenoma recurrence. Am J Clin Nutr. 2004;79:691–7.[PubMed][Google Scholar]
  • 32. Lee JE, Willett WC, Fuchs CS, et al Folate intake and risk of colorectal cancer and adenoma: modification by time. Am J Clin Nutr. 2011;93:817–25.[Google Scholar]
  • 33. Fujimori S, Gudis K, Takahashi Y, et al Determination of the minimal essential serum folate concentration for reduced risk of colorectal adenoma. Clin Nutr. 2011;30:653–8.[PubMed][Google Scholar]
  • 34. Fife J, Raniga S, Hider PN, et al Folic acid supplementation and colorectal cancer risk: a meta-analysis. Colorectal Dis. 2011;13:132–7.[PubMed][Google Scholar]
  • 35. Qin X, Cui Y, Shen L, et al Folic acid supplementation and cancer risk: A meta-analysis of randomized controlled trials. Int J Cancer. 2013 [Epub ahead of print] [[PubMed][Google Scholar]
  • 36. Figueiredo JC, Mott LA, Giovannucci E, et al Folic acid and prevention of colorectal adenomas: a combined analysis of randomized clinical trials. Int J Cancer. 2011;129:192–203.[Google Scholar]
  • 37. Ngo SN, Williams DB, Cobiac L, et al Does garlic reduce risk of colorectal cancer? A systematic review. J Nutr. 2007;137:2264–9.[PubMed][Google Scholar]
  • 38. El-Bayoumy K, Sinha R, Pinto JT, et al Cancer chemoprevention by garlic and garlic-containing sulfur and selenium compounds. J Nutr. 2006;136:864S–869S.[PubMed][Google Scholar]
  • 39. Knowles LM, Milner JAPossible mechanism by which allyl sulfides suppress neoplastic cell proliferation. J Nutr. 2001;131:1061S–6S.[PubMed][Google Scholar]
  • 40. Tanaka S, Haruma K, Yoshihara M, et al Aged garlic extract has potential suppressive effect on colorectal adenomas in humans. J Nutr. 2006;136:821S–826S.[PubMed][Google Scholar]
  • 41. Tanaka S, Haruma K, Kunihiro M, et al Effects of aged garlic extract (AGE) on colorectal adenomas: a double-blinded study. Hiroshima J Med Sci. 2004;53:39–45.[PubMed][Google Scholar]
  • 42. Fleischauer AT, Poole C, Arab LGarlic consumption and cancer prevention: meta-analyses of colorectal and stomach cancers. Am J Clin Nutr. 2000;72:1047–52.[PubMed][Google Scholar]
  • 43. Hall MN, Chavarro JE, Lee IM, et al A 22-year prospective study of fish, n-3 fatty acid intake, and colorectal cancer risk in men. Cancer Epidemiol Biomarkers Prev. 2008;17:1136–43.[Google Scholar]
  • 44. Jedrychowski W, Maugeri U, Pac A, et al. Protective effect of fish consumption on colorectal cancer risk. Hospital-based case-control study in Eastern Europe. Ann Nutr Metab. 2008;53:295–302.[PubMed]
  • 45. Kato I, Akhmedkhanov A, Koenig K, et al Prospective study of diet and female colorectal cancer: the New York University Women's Health Study. Nutr Cancer. 1997;28:276–81.[PubMed][Google Scholar]
  • 46. Sugawara Y, Kuriyama S, Kakizaki M, et al Fish consumption and the risk of colorectal cancer: the Ohsaki Cohort Study. Br J Cancer. 2009;101:849–54.[Google Scholar]
  • 47. Kobayashi M, Tsubono Y, Otani T, et al Fish, long-chain n-3 polyunsaturated fatty acids, and risk of colorectal cancer in middle-aged Japanese: the JPHC study. Nutr Cancer. 2004;49:32–40.[PubMed][Google Scholar]
  • 48. English DR, MacInnis RJ, Hodge AM, et al Red meat, chicken, and fish consumption and risk of colorectal cancer. Cancer Epidemiol Biomarkers Prev. 2004;13:1509–14.[PubMed][Google Scholar]
  • 49. Wu S, Feng B, Li K, et al Fish consumption and colorectal cancer risk in humans: a systematic review and meta-analysis. Am J Med. 2012;125:551–9.e5.[PubMed][Google Scholar]
  • 50. Pot GK, Majsak-Newman G, Geelen A, et al Fish consumption and markers of colorectal cancer risk: a multicenter randomized controlled trial. Am J Clin Nutr. 2009;90:354–61.[PubMed][Google Scholar]
  • 51. Selhub JFolate, vitamin B12 and vitamin B6 and one carbon metabolism. J Nutr Health Aging. 2002;6:39–42.[PubMed][Google Scholar]
  • 52. Matsubara K, Komatsu S, Oka T, et al Vitamin B6-mediated suppression of colon tumorigenesis, cell proliferation, and angiogenesis (review). J Nutr Biochem. 2003;14:246–50.[PubMed][Google Scholar]
  • 53. Shen J, Lai CQ, Mattei J, et al Association of vitamin B-6 status with inflammation, oxidative stress, and chronic inflammatory conditions: the Boston Puerto Rican Health Study. Am J Clin Nutr. 2010;91:337–42.[Google Scholar]
  • 54. Larsson SC, Orsini N, Wolk AVitamin B6 and risk of colorectal cancer: a meta-analysis of prospective studies. JAMA. 2010;303:1077–83.[PubMed][Google Scholar]
  • 55. Lipkin M, Newmark HEffect of added dietary calcium on colonic epithelial-cell proliferation in subjects at high risk for familial colonic cancer. N Engl J Med. 1985;313:1381–4.[PubMed][Google Scholar]
  • 56. Shabahang M, Buras RR, Davoodi F, et al Growth inhibition of HT-29 human colon cancer cells by analogues of 1,25-dihydroxyvitamin D3. Cancer Res. 1994;54:4057–64.[PubMed][Google Scholar]
  • 57. Martínez ME, Willett WCCalcium, vitamin D, and colorectal cancer: a review of the epidemiologic evidence. Cancer Epidemiol Biomarkers Prev. 1998;7:163–8.[PubMed][Google Scholar]
  • 58. Bergsma-Kadijk JA, van't Veer P, Kampman E, et al Calcium does not protect against colorectal neoplasia. Epidemiology. 1996;7:590–7.[PubMed][Google Scholar]
  • 59. Carroll C, Cooper K, Papaioannou D, et al Supplemental calcium in the chemoprevention of colorectal cancer: a systematic review and meta-analysis. Clin Ther. 2010;32:789–803.[PubMed][Google Scholar]
  • 60. Garland CF, Garland FCDo sunlight and vitamin D reduce the likelihood of colon cancer? Int J Epidemiol. 1980;9:227–31.[PubMed][Google Scholar]
  • 61. Wactawski-Wende J, Kotchen JM, Anderson GL, et al Calcium plus vitamin D supplementation and the risk of colorectal cancer. N Engl J Med. 2006;354:684–96.[PubMed][Google Scholar]
  • 62. Garland CF, Comstock GW, Garland FC, et al Serum 25-hydroxyvitamin D and colon cancer: eight-year prospective study. Lancet. 1989;2:1176–8.[PubMed][Google Scholar]
  • 63. Braun MM, Helzlsouer KJ, Hollis BW, et al Colon cancer and serum vitamin D metabolite levels 10-17 years prior to diagnosis. Am J Epidemiol. 1995;142:608–11.[PubMed][Google Scholar]
  • 64. Feskanich D, Ma J, Fuchs CS, et al Plasma vitamin D metabolites and risk of colorectal cancer in women. Cancer Epidemiol Biomarkers Prev. 2004;13:1502–8.[PubMed][Google Scholar]
  • 65. Gorham ED, Garland CF, Garland FC, et al Optimal vitamin D status for colorectal cancer prevention: a quantitative meta analysis. Am J Prev Med. 2007;32:210–6.[PubMed][Google Scholar]
  • 66. Yin L, Grandi N, Raum E, et al Meta-analysis: longitudinal studies of serum vitamin D and colorectal cancer risk. Aliment Pharmacol Ther. 2009;30:113–25.[PubMed][Google Scholar]
  • 67. Sanmartín C, Plano D, Sharma AK, et al Selenium compounds, apoptosis and other types of cell death: an overview for cancer therapy. Int J Mol Sci. 2012;13:9649–72.[Google Scholar]
  • 68. Ramoutar RR, Brumaghim JLEffects of inorganic selenium compounds on oxidative DNA damage. J Inorg Biochem. 2007;101:1028–35.[PubMed][Google Scholar]
  • 69. Broome CS, McArdle F, Kyle JA, et al An increase in selenium intake improves immune function and poliovirus handling in adults with marginal selenium status. Am J Clin Nutr. 2004;80:154–62.[PubMed][Google Scholar]
  • 70. Connelly-Frost A, Poole C, Satia JA, et al Selenium, folate, and colon cancer. Nutr Cancer. 2009;61:165–78.[Google Scholar]
  • 71. Fernández-Bañares F, Cabré E, Esteve M, et al Serum selenium and risk of large size colorectal adenomas in a geographical area with a low selenium status. Am J Gastroenterol. 2002;97:2103–8.[PubMed][Google Scholar]
  • 72. Clark LC, Hixson LJ, Combs GF, Jr, et al Plasma selenium concentration predicts the prevalence of colorectal adenomatous polyps. Cancer Epidemiol Biomarkers Prev. 1993;2:41–6.[PubMed][Google Scholar]
  • 73. Garland M, Morris JS, Stampfer MJ, et al Prospective study of toenail selenium levels and cancer among women. J Natl Cancer Inst. 1995;87:497–505.[PubMed][Google Scholar]
  • 74. van den Brandt PA, Goldbohm RA, van't Veer P, et al A prospective cohort study on toenail selenium levels and risk of gastrointestinal cancer. J Natl Cancer Inst. 1993;85:224–9.[PubMed][Google Scholar]
  • 75. Takata Y, Kristal AR, King IB, et al Serum selenium, genetic variation in selenoenzymes, and risk of colorectal cancer: primary analysis from the Women's Health Initiative Observational Study and meta-analysis. Cancer Epidemiol Biomarkers Prev. 2011;20:1822–30.[Google Scholar]
  • 76. Clark LC, Combs GF, Jr, Turnbull BW, et al. Effects of selenium supplementation for cancer prevention in patients with carcinoma of the skin. A randomized controlled trial. Nutritional Prevention of Cancer Study Group. JAMA. 1996;276:1957–63.[PubMed]
  • 77. Duffield-Lillico AJ, Reid ME, Turnbull BW, et al Baseline characteristics and the effect of selenium supplementation on cancer incidence in a randomized clinical trial: a summary report of the Nutritional Prevention of Cancer Trial. Cancer Epidemiol Biomarkers Prev. 2002;11:630–9.[PubMed][Google Scholar]
  • 78. Jacobs ET, Jiang R, Alberts DS, et al Selenium and colorectal adenoma: results of a pooled analysis. J Natl Cancer Inst. 2004;96:1669–75.[PubMed][Google Scholar]
  • 79. Duffield-Lillico AJ, Shureiqi I, Lippman SMCan selenium prevent colorectal cancer? A signpost from epidemiology. J Natl Cancer Inst. 2004;96:1645–7.[PubMed][Google Scholar]
  • 80. Hartwig ARole of magnesium in genomic stability. Mutat Res. 2001;475:113–21.[PubMed][Google Scholar]
  • 81. Chen GC, Pang Z, Liu QFMagnesium intake and risk of colorectal cancer: a meta-analysis of prospective studies. Eur J Clin Nutr. 2012;66:1182–6.[PubMed][Google Scholar]
  • 82. Randi G, Edefonti V, Ferraroni M, et al Dietary patterns and the risk of colorectal cancer and adenomas. Nutr Rev. 2010;68:389–408.[PubMed][Google Scholar]
  • 83. Miller PE, Lesko SM, Muscat JE, et al Dietary patterns and colorectal adenoma and cancer risk: a review of the epidemiological evidence. Nutr Cancer. 2010;62:413–24.[PubMed][Google Scholar]
  • 84. Calle EE, Kaaks ROverweight, obesity and cancer: epidemiological evidence and proposed mechanisms. Nat Rev Cancer. 2004;4:579–91.[PubMed][Google Scholar]
  • 85. Wiseman M. The second World Cancer Research Fund/American Institute for Cancer Research expert report. Food, nutrition, physical activity, and the prevention of cancer: a global perspective. Proc Nutr Soc. 2008;67:253–6.[PubMed]
  • 86. 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. 1997;89:948–55.[PubMed]
  • 87. Giovannucci E, Ascherio A, Rimm EB, et al Physical activity, obesity, and risk for colon cancer and adenoma in men. Ann Intern Med. 1995;122:327–34.[PubMed][Google Scholar]
  • 88. 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. 2003;348:1625–38.[PubMed]
  • 89. Moghaddam AA, Woodward M, Huxley RObesity and risk of colorectal cancer: a meta-analysis of 31 studies with 70,000 events. Cancer Epidemiol Biomarkers Prev. 2007;16:2533–47.[PubMed][Google Scholar]
  • 90. Ning Y, Wang L, Giovannucci ELA quantitative analysis of body mass index and colorectal cancer: findings from 56 observational studies. Obes Rev. 2010;11:19–30.[PubMed][Google Scholar]
  • 91. Meyerhardt JA, Catalano PJ, Haller DG, et al Influence of body mass index on outcomes and treatment-related toxicity in patients with colon carcinoma. Cancer. 2003;98:484–95.[PubMed][Google Scholar]
  • 92. Dignam JJ, Polite BN, Yothers G, et al Body mass index and outcomes in patients who receive adjuvant chemotherapy for colon cancer. J Natl Cancer Inst. 2006;98:1647–54.[PubMed][Google Scholar]
  • 93. Meyerhardt JA, Sato K, Niedzwiecki D, et al Dietary glycemic load and cancer recurrence and survival in patients with stage III colon cancer: findings from CALGB 89803. J Natl Cancer Inst. 2012;104:1702–11.[Google Scholar]
  • 94. Meyerhardt JA, Niedzwiecki D, Hollis D, et al Association of dietary patterns with cancer recurrence and survival in patients with stage III colon cancer. JAMA. 2007;298:754–64.[PubMed][Google Scholar]
  • 95. Fung T, Hu FB, Fuchs C, et al Major dietary patterns and the risk of colorectal cancer in women. Arch Intern Med. 2003;163:309–14.[PubMed][Google Scholar]
  • 96. 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. 2006;295:643–54.[PubMed][Google Scholar]
  • 97. Kaaks R, Lukanova AEnergy balance and cancer: the role of insulin and insulin-like growth factor-I. Proc Nutr Soc. 2001;60:91–106.[PubMed][Google Scholar]
  • 98. Giovannucci E, Pollak MN, Platz EA, et al A prospective study of plasma insulin-like growth factor-1 and binding protein-3 and risk of colorectal neoplasia in women. Cancer Epidemiol Biomarkers Prev. 2000;9:345–9.[PubMed][Google Scholar]
  • 99. Manousos O, Souglakos J, Bosetti C, et al IGF-I and IGF-II in relation to colorectal cancer. Int J Cancer. 1999;83:15–7.[PubMed][Google Scholar]
  • 100. Schoen RE, Tangen CM, Kuller LH, et al Increased blood glucose and insulin, body size, and incident colorectal cancer. J Natl Cancer Inst. 1999;91:1147–54.[PubMed][Google Scholar]
  • 101. Ma J, Pollak MN, Giovannucci E, et al Prospective study of colorectal cancer risk in men and plasma levels of insulin-like growth factor (IGF)-I and IGF-binding protein-3. J Natl Cancer Inst. 1999;91:620–5.[PubMed][Google Scholar]
  • 102. Wei EK, Ma J, Pollak MN, et al A prospective study of C-peptide, insulin-like growth factor-I, insulin-like growth factor binding protein-1, and the risk of colorectal cancer in women. Cancer Epidemiol Biomarkers Prev. 2005;14:850–5.[PubMed][Google Scholar]
  • 103. Kaaks R, Toniolo P, Akhmedkhanov A, et al Serum C-peptide, insulin-like growth factor (IGF)-I, IGF-binding proteins, and colorectal cancer risk in women. J Natl Cancer Inst. 2000;92:1592–600.[PubMed][Google Scholar]
  • 104. Ma J, Giovannucci E, Pollak M, et al A prospective study of plasma C-peptide and colorectal cancer risk in men. J Natl Cancer Inst. 2004;96:546–53.[PubMed][Google Scholar]
  • 105. Haydon AM, Macinnis RJ, English DR, et al Effect of physical activity and body size on survival after diagnosis with colorectal cancer. Gut. 2006;55:62–7.[Google Scholar]
  • 106. Wolin KY, Yan Y, Colditz GA, et al Physical activity and colon cancer prevention: a meta-analysis. Br J Cancer. 2009;100:611–6.[Google Scholar]
  • 107. Boyle T, Keegel T, Bull F, et al Physical activity and risks of proximal and distal colon cancers: a systematic review and meta-analysis. J Natl Cancer Inst. 2012;104:1548–61.[PubMed][Google Scholar]
  • 108. Courneya KS, Booth CM, Gill S, et al The Colon Health and Life-Long Exercise Change trial: a randomized trial of the National Cancer Institute of Canada Clinical Trials Group. Curr Oncol. 2008;15:279–85.[Google Scholar]
  • 109. Meyerhardt JA, Heseltine D, Niedzwiecki D, et al Impact of physical activity on cancer recurrence and survival in patients with stage III colon cancer: findings from CALGB 89803. J Clin Oncol. 2006;24:3535–41.[PubMed][Google Scholar]
  • 110. Winzer BM, Whiteman DC, Reeves MM, et al Physical activity and cancer prevention: a systematic review of clinical trials. Cancer Causes Control. 2011;22:811–26.[PubMed][Google Scholar]
  • 111. Boffetta P, Hashibe MAlcohol and cancer. Lancet Oncol. 2006;7:149–56.[PubMed][Google Scholar]
  • 112. Giovannucci E, Rimm EB, Ascherio A, et al Alcohol, low-methionine--low-folate diets, and risk of colon cancer in men. J Natl Cancer Inst. 1995;87:265–73.[PubMed][Google Scholar]
  • 113. Fedirko V, Tramacere I, Bagnardi V, et al Alcohol drinking and colorectal cancer risk: an overall and dose-response meta-analysis of published studies. Ann Oncol. 2011;22:1958–72.[PubMed][Google Scholar]
  • 114. Mizoue T, Inoue M, Wakai K, et al. Research Group for Development and Evaluation of Cancer Prevention Strategies in Japan. Alcohol drinking and colorectal cancer in Japanese: a pooled analysis of results from five cohort studies. Am J Epidemiol. 2008;167:1397–406.[PubMed]
  • 115. Moskal A, Norat T, Ferrari P, et al Alcohol intake and colorectal cancer risk: a dose-response meta-analysis of published cohort studies. Int J Cancer. 2007;120:664–71.[PubMed][Google Scholar]
  • 116. Cho E, Smith-Warner SA, Ritz J, et al Alcohol intake and colorectal cancer: a pooled analysis of 8 cohort studies. Ann Intern Med. 2004;140:603–13.[PubMed][Google Scholar]
  • 117. Botteri E, Iodice S, Bagnardi V, et al Smoking and colorectal cancer: a meta-analysis. JAMA. 2008;300:2765–78.[PubMed][Google Scholar]
  • 118. Liang PS, Chen TY, Giovannucci ECigarette smoking and colorectal cancer incidence and mortality: systematic review and meta-analysis. Int J Cancer. 2009;124:2406–15.[PubMed][Google Scholar]
  • 119. Gong J, Hutter C, Baron JA, et al A pooled analysis of smoking and colorectal cancer: timing of exposure and interactions with environmental factors. Cancer Epidemiol Biomarkers Prev. 2012;21:1974–85.[Google Scholar]
  • 120. Rennert G, Rennert HS, Pinchev M, et al Use of hormone replacement therapy and the risk of colorectal cancer. J Clin Oncol. 2009;27:4542–7.[Google Scholar]
  • 121. Hoffmeister M, Raum E, Winter J, et al Hormone replacement therapy, body mass, and the risk of colorectal cancer among postmenopausal women from Germany. Br J Cancer. 2007;97:1486–92.[Google Scholar]
  • 122. Franceschi S, La Vecchia CColorectal cancer and hormone replacement therapy: an unexpected finding. Eur J Cancer Prev. 1998;7:427–38.[PubMed][Google Scholar]
  • 123. Kabat GC, Miller AB, Rohan TEOral contraceptive use, hormone replacement therapy, reproductive history and risk of colorectal cancer in women. Int J Cancer. 2008;122:643–6.[PubMed][Google Scholar]
  • 124. Purdue MP, Mink PJ, Hartge P, et al Hormone replacement therapy, reproductive history, and colorectal adenomas: data from the Prostate, Lung, Colorectal and Ovarian (PLCO) Cancer Screening Trial (United States). Cancer Causes Control. 2005;16:965–73.[PubMed][Google Scholar]
  • 125. Nanda K, Bastian LA, Hasselblad V, et al Hormone replacement therapy and the risk of colorectal cancer: a meta-analysis. Obstet Gynecol. 1999;93:880–8.[PubMed][Google Scholar]
  • 126. Chlebowski RT, Wactawski-Wende J, Ritenbaugh C, et al Estrogen plus progestin and colorectal cancer in postmenopausal women. N Engl J Med. 2004;350:991–1004.[PubMed][Google Scholar]
  • 127. Ritenbaugh C, Stanford JL, Wu L, et al. Women's Health Initiative Investigators. Conjugated equine estrogens and colorectal cancer incidence and survival: the Women's Health Initiative randomized clinical trial. Cancer Epidemiol Biomarkers Prev. 2008;17:2609–18.
  • 128. Chan AT, Giovannucci EL, Meyerhardt JA, et al Long-term use of aspirin and nonsteroidal anti-inflammatory drugs and risk of colorectal cancer. JAMA. 2005;294:914–23.[Google Scholar]
  • 129. Chan AT, Giovannucci EL, Schernhammer ES, et al A prospective study of aspirin use and the risk for colorectal adenoma. Ann Intern Med. 2004;140:157–66.[PubMed][Google Scholar]
  • 130. García-Rodríguez LA, Huerta-Alvarez CReduced risk of colorectal cancer among long-term users of aspirin and nonaspirin nonsteroidal antiinflammatory drugs. Epidemiology. 2001;12:88–93.[PubMed][Google Scholar]
  • 131. Greenberg ER, Baron JA, Freeman DH, Jr, et al. Reduced risk of large-bowel adenomas among aspirin users. The Polyp Prevention Study Group. J Natl Cancer Inst. 1993;85:912–6.[PubMed]
  • 132. Logan RF, Little J, Hawtin PG, et al Effect of aspirin and non-steroidal anti-inflammatory drugs on colorectal adenomas: case-control study of subjects participating in the Nottingham faecal occult blood screening programme. BMJ. 1993;307:285–9.[Google Scholar]
  • 133. Tangrea JA, Albert PS, Lanza E, et al Non-steroidal anti-inflammatory drug use is associated with reduction in recurrence of advanced and non-advanced colorectal adenomas (United States). Cancer Causes Control. 2003;14:403–11.[PubMed][Google Scholar]
  • 134. Gann PH, Manson JE, Glynn RJ, et al Low-dose aspirin and incidence of colorectal tumors in a randomized trial. J Natl Cancer Inst. 1993;85:1220–4.[PubMed][Google Scholar]
  • 135. 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. 2007;369:1603–13.[PubMed]
  • 136. Baron JA, Cole BF, Sandler RS, et al A randomized trial of aspirin to prevent colorectal adenomas. N Engl J Med. 2003;348:891–9.[PubMed][Google Scholar]
  • 137. Sandler RS, Halabi S, Baron JA, et al A randomized trial of aspirin to prevent colorectal adenomas in patients with previous colorectal cancer. N Engl J Med. 2003;348:883–90.[PubMed][Google Scholar]
  • 138. Bertagnolli MM, Eagle CJ, Zauber AG, et al Celecoxib for the prevention of sporadic colorectal adenomas. N Engl J Med. 2006;355:873–84.[PubMed][Google Scholar]
  • 139. Arber N, Eagle CJ, Spicak J, et al Celecoxib for the prevention of colorectal adenomatous polyps. N Engl J Med. 2006;355:885–95.[PubMed][Google Scholar]
  • 140. Algra AM, Rothwell PMEffects of regular aspirin on long-term cancer incidence and metastasis: a systematic comparison of evidence from observational studies versus randomised trials. Lancet Oncol. 2012;13:518–27.[PubMed][Google Scholar]
  • 141. Cook NR, Lee IM, Gaziano JM, et al Low-dose aspirin in the primary prevention of cancer: the Women's Health Study: a randomized controlled trial. JAMA. 2005;294:47–55.[PubMed][Google Scholar]
  • 142. Stürmer T, Glynn RJ, Lee IM, et al Aspirin use and colorectal cancer: post-trial follow-up data from the Physicians’ Health Study. Ann Intern Med. 1998;128:713–20.[PubMed][Google Scholar]
  • 143. Rothwell PM, Wilson M, Elwin CE, et al Long-term effect of aspirin on colorectal cancer incidence and mortality: 20-year follow-up of five randomised trials. Lancet. 2010;376:1741–50.[PubMed][Google Scholar]
  • 144. Cuzick J, Otto F, Baron JA, et al Aspirin and non-steroidal anti-inflammatory drugs for cancer prevention: an international consensus statement. Lancet Oncol. 2009;10:501–7.[PubMed][Google Scholar]
  • 145. Burn J, Gerdes AM, Macrae F, et al Long-term effect of aspirin on cancer risk in carriers of hereditary colorectal cancer: an analysis from the CAPP2 randomised controlled trial. Lancet. 2011;378:2081–7.[Google Scholar]
  • 146. Brown JR, DuBois RNCOX-2: a molecular target for colorectal cancer prevention. J Clin Oncol. 2005;23:2840–55.[PubMed][Google Scholar]
  • 147. McQuaid KR, Laine LSystematic review and meta-analysis of adverse events of low-dose aspirin and clopidogrel in randomized controlled trials. Am J Med. 2006;119:624–38.[PubMed][Google Scholar]
  • 148. Antithrombotic Trialists’ Collaboration Collaborative meta-analysis of randomised trials of antiplatelet therapy for prevention of death, myocardial infarction, and stroke in high risk patients. BMJ. 2002;324:71–86.
  • 149. U.S. Preventive Services Task Force Routine aspirin or nonsteroidal anti-inflammatory drugs for the primary prevention of colorectal cancer: U.S. Preventive Services Task Force recommendation statement. Ann Intern Med. 2007;146:361–4.[PubMed]
  • 150. 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. 2011;377:31–41.[PubMed][Google Scholar]
  • 151. Chan AT, Arber N, Burn J, et al Aspirin in the chemoprevention of colorectal neoplasia: an overview. Cancer Prev Res (Phila) 2012;5:164–78.[Google Scholar]
Collaboration tool especially designed for Life Science professionals.Drag-and-drop any entity to your messages.