Skip to main content

Effects of NSAIDs on the risk factors of colorectal cancer: a mini review

Abstract

Evidence from epidemiological and experimental studies has shown that non-steroidal anti-inflammatory drugs (NSAIDs) reduce the risk of colorectal cancer (CRC). The function of NSAIDs and the molecular targets for chemopreventive effects on CRC have been extensively studied and their data were reported. However, the relation between NSAIDs and the risk factors of CRC have not been fully elucidated yet. Thus, relations between NSAIDs and the risk factors of CRC, such as overweight and obesity, alcohol, aging, hypertriglyceridemia and smoking, are summarized with our data and with recent reported data in this review.

Background

Non-steroidal anti-inflammatory drugs (NSAIDs) relieve pain, reduce inflammation, lower fevers and prevent blood from clotting. Thus, NSAIDs are used to treat inflammatory conditions such as arthritis. One of the traditional NSAIDs, aspirin, was used to protect against heart disease. Conversely, NSAIDs can increase the risk of gastrointestinal (GI) bleedings/ulcer and interfere with kidney function. The severity of side effects increases by taking NSAIDs longer.

A large number of epidemiological and experimental studies have shown that NSAIDs reduce the risk of colorectal cancer (CRC). Meta-analysis of randomized trials revealed that use of aspirin for approximately 5 years reduces incidence and mortality due to CRC by 30–40 % after 20 years of follow-up [1].

The effectiveness of NSAIDS may be attributed to their potent inhibition of cyclooxygenase (COX) enzymes because COX-2 expression and prostaglandin (PG) E2 synthesis are elevated in CRC. The COXs/PGH synthases have two enzymes, COX-1 and COX-2, and COX is the limiting enzyme of the PG synthesis pathway. The constitutive enzyme COX-1 has low expression in normal human colorectal tissue, whereas the inducible enzyme COX-2 is elevated in tissue involved in inflammation and in cancer. Traditional NSAIDs block the actions of both COX-1 and COX-2, and selective COX-2 inhibitors are a special category of NSAIDs. In addition, aspirin can inhibit proliferation and induce apoptosis of colon cancer cells [2]. The inhibition and induction by aspirin include the following: (i) the interruption of nuclear factor kappa B (NF-κB), (ii) the interruption of extracellular signal-regulated kinases, (iii) the induction of caspase 8 and 9, and (iv) the inhibition of β-catenin signaling.

As shown above, the function and molecular targets of NSAIDs have been well studied, and several pieces of evidence have shown their chemopreventive effects on CRC. However, the relation between NSAIDs and the risk factors of CRC have not been well examined. In this review, such relationships are summarized with our data and recent reported data in the text, Table 1 and 2.

Table 1 Effects of NSAIDs on risk factors of CRC
Table 2 Effects of NSAIDs plus each risk factors on CRC risk

Risk factors of colorectal cancer

A risk factor is any attribute, characteristic or exposure of an individual that increases the likelihood of developing a disease or injury [3]. In the case of cancer, it is defined as factors that increase the chance of developing cancer. Some people with several risk factors never develop cancer. There is an intensity of correlation between risk factors and cancer. Thus, there might be some weak risk factors of which we are not aware.

Several risk factors of CRC have been demonstrated to date. The World Health Organization (WHO) lists factors that are convincing evidence for the risk of CRC in daily life as follows: (i) consuming red meat, (ii) consuming processed meat, (iii) alcoholic drinks (men), (iv) body fatness / abdominal fatness, and (v) adult attained height increase [4]. Other risk factors are reported as aging [5] and a family history of CRC. Several medical histories of diseases are known to increase the risk of CRC, such as (i) genetic predisposition including familial adenomatous polyposis and hereditary nonpolyposis CRC, (ii) high-risk adenoma, (iii) inflammatory bowel disease, (iv) type-2 diabetes, and (v) hypertriglyceridemia [6]. Moreover, smoking is a strong factor in the development of colorectal adenoma [7].

From recent published literature, we took particular note of overweight and obesity, alcohol, aging, hypertriglyceridemia and smoking. The relation between each risk factor and NSAIDs is described below.

Obesity and NSAIDs

Obesity is an important risk factor for CRC [8], and there is a significant positive correlation between body mass index (BMI) and CRC risk [9, 10]. Visceral abdominal fat area measured by CT scan is more significantly associated with colorectal adenoma [11]. Visceral fat accumulation causes increases in adipokines such as leptin, inflammatory cytokines such as tumor necrosis factor (TNF) α and interleukin (IL)-6, and growth factors such as insulin-like growth factor (IGF)-I and vascular endothelial growth factor (VEGF) [12, 13]. These factors cause chronic low-grade inflammatory states, increased proliferation and angiogenesis, and promote colon carcinogenesis [12, 13].

The use of NSAIDs is associated with reduced risk of inflammation-associated cancers, and obesity-related cancers are included in inflammation-related cancers [14]. Compared with non-users of NSAIDs, lower risk of obesity-related cancers in NSAID users (hazard ratio (HR) = 0.88; 95 % confidence interval (CI), 0.85–0.92) has been reported [14]. Although the preventive effects of aspirin and NSAIDs on obesity-related cancers are inconclusive, there are several reports showing that aspirin reduced risks of obesity-related cancers including CRC, particularly among obese people. In the Aspirin/Folate Polyp Prevention Study, a randomized controlled trial of aspirin and folic acid to prevent colorectal adenoma, a daily dose of 325 mg aspirin reduced the risk ratio for advanced adenomas compared with placebo among obese subjects (relative risk (RR) = 0.44; 95 % CI, 0.17–1.10) but not among those with normal weight (RR = 1.23; 95 % CI, 0.55–2.77) [15]. In the CAPP2 study that recruited participants with Lynch syndrome, CRC risk was 2.41x greater for obese participants (95 % CI, 1.06–2.96; P = 0.03) than for underweight and normal-weight participants, and the obesity-related excess risk of CRC was confined to those randomly assigned to the aspirin placebo group (adjusted HR = 2.75; 95 % CI, 1.12–6.79, P = 0.03), but the risk was abrogated in those taking aspirin [16].

PGE2 levels in rectal mucosa have been reported to be positively associated with BMI [17]. PGE2 has been shown to promote colorectal carcinogenesis [18] and metastasis [19] in mouse models, and inhibition of PGE2 synthesis is a potential target for prevention of CRC [20]. Therefore, decrease of obesity-related CRC by NSAID use could be due to inhibition of PGE2 production.

Obesity causes insulin resistance with hyperglycemia and hyperinsulinemia, and these contribute to tumor development [21]. Metformin, an insulin resistance-improving, anti-diabetic agent, has been reported to reduce risks of various cancers including colon cancer in diabetic and non-diabetic populations [22, 23]. Metformin inhibits cell proliferation via activation of AMP-activated protein kinase (AMPK) [24]. Serum adiponectin is decreased in obese people, and also activates AMPK and inhibits cell proliferation of colon cancer cells [24]. Recently, it has been reported that aspirin activates AMPK and inhibits mTOR signaling in colon cancer cells [25]. Not all, but several NSAIDs, such as salicylic acid, ibuprofen or diclofenac, which have acidic structures, also induce AMPK activation [26, 27]. This effect is considered to be a COX-independent anti-inflammatory property of aspirin and a subgroup of NSAIDs and may contribute to decrease obesity-related cancer risks.

Alcohol and NSAIDs

In a meta-analysis of cohort and case–control studies, there are reports describing moderately increased risks of CRC with a dose–response relation for rising alcohol consumption. A polled analysis of eight cohort studies also recorded a dose–response relation between the risk of CRC and the amount of alcohol consumption [28].

The mechanisms by which alcohol intake exerts its carcinogenic effect are not fully understood yet. Acetaldehyde, a metabolite of alcohol, is implicated in esophageal carcinogenesis but is not strongly implicated in colorectal carcinogenesis. Recently, single nucleotide polymorphisms (SNPs) in the alcohol dehydrogenase, ADH1B, and aldehyde dehydrogenase, ALDH2, of moderate/heavy drinkers were shown to be contributing factors for CRC [29]. Aspirin and salicylate could inhibit both human ADH (metabolize ethanol to acetaldehyde) and ALDH (degradation of acetaldehyde) activities [30]. To date, the effect of aspirin on acetaldehyde production is not fully understood yet.

Ethanol is known as an irritant for the digestive tract. Ethanol intake is known to be an independent risk factor for GI bleeding. It is posited that the GI bleeding risk from aspirin is high in individuals who consume three or more alcoholic drinks every day [31].

Several reports suggest that ethanol exposure alters the cytokine levels and inflammatory status in a variety of tissues in vitro and in vivo, including the colon [32, 33]. Furthermore, chronic alcohol intake promoted intestinal tumorigenesis and tumor invasion in Apc Min/+ mice [34]. In a report, Wimberly et al. suggested that mast cell-mediated inflammation could be one of the mechanisms by which alcohol promotes intestinal carcinogenesis.

Recently, Landi and colleagues [35] studied the association between SNPs in the IL-6, IL-8, TNFα and PPARG genes and the risk of CRC by a hospital-based case control study. These genes are known to play important roles in inflammation of the colorectum, and common allelic variants are related to changes in biological functions in the inflammation pathway. In their study, Landi and colleagues observed an association between increased CRC risk and the C-allele of a SNP −174 G > C in the IL-6 gene (Odds ratio (OR) = 1.65, 95 % CI, 0.99–2.74). Several reports showed the association of C-allele carriers with inflammatory-related conditions, such as increased plasma levels of C-reactive protein [36], higher serum levels of IL-6 after coronary artery bypass surgery [37] and asymptomatic carotid artery atherosclerosis [38]. In terms of CRC risk, the effect of alcohol drinking was evident only in the subgroup of IL-6 C-allele carriers (OR = 2.19, 95 % CI, 1.3–3.7), and the use of NSAIDs halved the risk from 2.02 (95 % CI, 1.38–2.95) to 1.02 (95 % CI, 0.65–1.61) in the carriers of the C-allele.

These reports suggest that the carcinogenic effect of alcohol may be partly through the induction of acetaldehyde and inflammation, and NSAID use may effectively protect CRC development in a social drinker.

Aging and NSAIDs

Disruption of normal tissue function dramatically accelerates in old age. Aging is the greatest risk factor for numerous pathologies, including cancer, stroke, neurodegenerative disorders, heart disease and type-2 diabetes [5]. Chronic inflammation is one of the main processes that contributes to age-related disease and causes disruption of normal functioning of tissues. Notably, there is a robust increase of mRNA and secretion of numerous cytokines, chemokines, growth factors and proteases in the senescent cells, and these cells may cause a low level of chronic inflammation systemically during aging [39].

Yeast, nematodes and flies have been recognized as excellent model systems for studying the underlying mechanism of aging and identifying chemicals altering longevity, mainly because of their short lifespans. A growing number of reports showed the effects of NSAIDs on the lifespan extension in yeast [40], nematodes [40, 41], flies [40, 42] and mice [43]. He et al. [40] reported that ibuprofen extended the replicative life span of Saccharomyces cerevisiae cells by destabilizing the high-affinity tryptophan transporter. He et al. also found that ibuprofen caused small size at birth and moderate delay in initiation of cell division, which was observed in most long-lived yeast mutants. Meanwhile, celecoxib extended both mean and maximum lifespans in C. elegans [41]. The physical health, as indicated by the age-associated decay rate of motor activity, was also significantly improved in celecoxib-treated nematodes without affecting the nutritional value. However, no homologs of mammalian COXs have been identified in unicellular organisms, including C. elegans. Furthermore, as analogs of celecoxib that lacked COX-2 inhibitory activity also exhibited a similar effect on nematode lifespan, the anti-aging effect of celecoxib might be independent of its COX-2 inhibitory activity. Indeed, celecoxib was shown to inhibit the activity of 3’-phosphoinositide-dependent kinase-1 (PDK-1), a key component of the insulin/IGF-1 signaling cascade that is involved in lifespan regulation in C. elegans. Other studies have demonstrated that NSAIDs have antioxidative effects through anti-radical activity and membrane-stabilizing action [44, 45].

Thus, NSAIDs might be effective for slowing aging and prevention of age-related diseases through not only their anti-inflammatory effects via COX-2 inhibitory action but through potential secondary targets including PDK-1 inhibition and the anti-oxidative effect.

Triglycerides and NSAIDs

The triglyceride values (TG, Neutral fat value) are one of the three indices characterizing metabolic syndrome. Metabolic syndrome is evoked by an accumulation of abdominal fat, and, as previously mentioned, several obesity-associated cancers [46] may occur.

The serum TG value is correlated with a high-fat diet and is higher in patients in an obese state. When the serum TG value increases, colorectal adenomas (often in such as colon polyps) are more likely to occur. This occurrence has been noted in several epidemiological studies. For example, high TGs in hypertriglyceridemia were associated with colorectal adenoma (OR = 1.5, 95 % CI, 1.1–2.0 for the highest versus the lowest quartile, P trend = 0.03). A stronger association was observed between three or more adenoma cases and study controls (OR = 2.3, 95 % CI, 1.3–4.2, P trend < 0.001) [7]. In Japanese men, risk of CRC is increased under high-TG levels [6]. Animal experiments supported the relation between high-TG and carcinogenesis by explaining the underlying mechanisms. Thus, high TG might be understood as a risk factor for CRC [47, 48].

On the other hand, there are a variety of discussions about whether NSAIDs can adjust the TG value. NSAIDs can inhibit the enzymatic activity of COX and attenuate the expression level of PGE2. PGE2 is activated through an EP3 receptor at the thermoregulatory center in the hypothalamus and functions to raise the set point of body temperature by increasing the cellular metabolism. This linkage means NSAIDs decrease TG levels. Furthermore, PGE2 induces the expression of TNFα, and the TG value also increases because of the inhibition of lipoprotein lipase (LPL) [49]. LPL catalyzes the hydrolysis of plasma TG.

The lowering of the TG value by NSAIDs is also found in several studies in the literature. For example, when NSAIDs were used in animal experimental models, they significantly reduced the total cholesterol, TG and low-density lipoprotein (LDL) concentrations in the plasma of hyperlipidemic rats [49, 50].

Niho et al. investigated the influence of the general COX inhibitor, indomethacin, in Min mice and found that treatment with 10 ppm indomethacin in the diet for 14 weeks caused 90 % reduction in serum TG values, along with a reduction in the number of intestinal polyps to 25 % of the untreated control value [51]. In this experiment, LPL mRNA levels in the liver were slightly increased by indomethacin treatment. In humans, indomethacin does not affect serum lipids.

In another model animal experiment, aspirin could improve serum high-TG. Human apolipoprotein CI (apoCI)-expressing mice (APOC1 mice), an animal model with elevated plasma TG levels, as well as normolipidemic wild-type mice, were fed a high-fat diet and treated with aspirin. Aspirin treatment reduced hepatic NF-κB activity in high-fat diet-fed APOC1 and wild-type mice, and in addition, aspirin decreased plasma TG levels (−32 %, P < 0.05) in hypertriglyceridemic APOC1 mice. This TG-lowering effect could not be explained by enhanced VLDL-TG clearance, but aspirin selectively reduced hepatic production of VLDL-TG in both APOC1 (−28 %, P < 0.05) and wild-type mice (−33 %, P < 0.05) without affecting VLDL-apoB production [52]. In humans, higher proportions of patients in the salsalate (one of the NSAIDS) treatment groups experienced decreasing circulating TG values and increasing adiponectin concentrations [53]. For preventing colorectal carcinogenesis, significant consideration should be given to the use of NSAIDs to decrease plasma TG levels.

Smoking and NSAIDs

Smoking is a strong risk factor for the incidence of colorectal adenomas [7] but a weak risk factor for the colorectal adenocarcinomas [54]. Conversely, NSAIDs including aspirin are known to reduce the adenoma recurrence rate. Evidence has accumulated that shows current smoking abrogates or inversely affects the use of aspirin.

We recently performed two trials using enteric-coated aspirin and found that aspirin similarly increased the risk of colorectal adenomas in current smokers [55]. One of these trials is the J-CAPP Study [56]. It is a randomized controlled trial involving 311 patients from Asia with colorectal adenomas and/or early-stage adenocarcinomas (adenocarcinomas with invasions confined to the mucosa) that evaluated the effects of 100 mg/day enteric-coated aspirin for two years. In subgroup analyses, we found a reduced adenoma recurrence rate with an OR of 0.37 (95 % CI, 0.21–0.68) in non-smokers (never-smokers/ex-smokers) and 3.45 (95 % CI, 1.12–10.64) in current smokers. The other trial is the J-FAPP Study II [57]. It is also a randomized controlled trial. It involved 34 subjects with Asian familial adenomatous polyposis that evaluated the effects of 100 mg/day enteric-coated aspirin for 6 to 10 months. In the trial, the OR for a reduction in the diameter of polyps was 0.10 (95 % CI, 0.01–0.98) in non-smokers and 3.00 (95 % CI, 0.15–59.89) in current smokers.

After our findings, two papers confirmed the effects of aspirin use on the status of smoking for colorectal adenoma recurrence. A randomized, double-blind, placebo-controlled trial was performed at centers in Europe, Russia, or the US using 75 mg aspirin for 3 years for patients with 1 or more sporadic adenomas removed from the colon or rectum [58]. Its subgroup analyses revealed OR values of 0.65 (95 % CI, 0.26 − 1.22) and 1.70 (95 % CI, 0.70–4.09) in non-smokers and current smokers, respectively. The other paper was a cross-sectional study including 2,918 consecutive colonoscopy patients over a 30-month period at a university hospital in the US [59]. The incidental rate ratio (IRR) of polyps was 1.72 (95 % CI, 1.46–2.02) in active smokers and 0.73 (95 % CI, 0.61–0.86) in daily aspirin users compared to those without aspirin. Current smoking interacts significantly with aspirin, resulting in an IRR of 1.69 (95 % CI, 1.28–2.24) that shows loss of aspirin protection.

The mechanism by which smoking influences the effect of aspirin is unknown. One clue is that smoking may be associated with resistance to aspirin, possibly through excessive thromboxane production [60, 61]. Further studies are warranted because this issue is very important for the clinical use of aspirin in the future.

Conclusion with recent recommendation of low-dose aspirin

On September 15, 2015, the U.S. Preventive Services Task Force (USPSTF) posted a draft recommendation statement with several limitations about taking low-dose aspirin every day to help prevent cardiovascular disease (CVD), such as heart attack and stroke, and CRC [62]. They stated that patients aged 50–59 who have a 10 % or greater 10-year CVD risk [63] but who are not at increased risk for bleeding and have at least a 10-year life expectancy fall within the USPSTF B recommendation to take low-dose aspirin daily for at least 10 years. The definition of the B recommendation is that there is high certainty that the net benefit of low-dose aspirin is moderate, or there is moderate certainty that the net benefit is moderate to substantial [62].

The USPSTF also posted that it is insufficient to assess the benefit and harm of aspirin use for those older than 80, and it recommended the use of aspirin for prevention of stroke in women < 55 and for prevention of myocardial infarction in men < 45. For several NSAIDs, such as diclofenac, the NSAIDs will increase the risk of death and recurrent myocardial infraction in patients with prior myocardial infraction [64]. From our data, smoking status should be included in the USPSTF recommendation statement. Use of NSAIDs for pain relief may increase physical activation in a convincing way to reduce the risk of CRC [4]. It will be worthwhile to know the effects of NSAIDs on the risk factors of CRC and utilize them for a person’s physical health as well as to prevent CRC.

Abbreviations

ADH:

Alcohol dehydrogenases

ALDH:

Aldehyde dehydrogenase

AMPK:

AMP-activated protein kinase

BMI:

Body mass index

CI:

Confidence interval

COX:

Cyclooxygenase

CRC:

Colorectal cancer

CVD:

Cardiovascular disease

GI:

Gastrointestinal

HR:

Hazard ratio

IGF:

Insulin-like growth factor

IL:

Interleukin

IRR:

Incidental rate ratio

LDL:

Low-density lipoprotein

LPL:

Lipoprotein lipase

NSAIDs:

Non-steroidal anti-inflammatory drugs

NF-κB:

Nuclear factor-kappaB

PDK-1:

3’-phosphoinositide-dependent kinase-1

PG:

Prostaglandin

RR:

Relative risk

OR:

Odds ratio

SNPs:

Single nucleotide polymorphisms

TG:

Triglyceride

TNF:

Tumor necrosis factor

USPSTF:

U.S. Preventive Services Task Force

WHO:

World Health Organization

References

  1. Rothwell PM, Wilson M, Elwin CE, Norrving B, Algra A, Warlow CP, et al. Long-term effect of aspirin on colorectal cancer incidence and mortality: 20-year follow-up of five randomised trials. Lancet. 2010;376:1741–50.

    Article  CAS  PubMed  Google Scholar 

  2. Komiya M, Fujii G, Takahashi M, Iigo M, Mutoh M. Prevention and intervention trials for colorectal cancer. Japanese J Clin Oncol. 2013;43:685–94.

    Article  Google Scholar 

  3. World Health Organization. http://www.who.int/topics/risk_factors/en/ (2015). Accessed 6 Nov 2015.

  4. World Cancer Research Fund/American Institute for Cancer Research. Food, Nutrition and the Prevention of Cancer: a Global Perspective, World Cancer Research Fund/ American Institute for Cancer Research, Washington DC, 2007.

  5. Niccoli T, Partridge L. Ageing as a risk factor for disease. Curr Biol. 2012;22:R741–52.

    Article  CAS  PubMed  Google Scholar 

  6. Inoue M, Noda M, Kurahashi N, Iwasaki M, Sasazuki S, Iso H, et al. Impact of metabolic factors on subsequent cancer risk: results from a large-scale population-based cohort study in Japan. Eur J Cancer Prev. 2009;18:240–7.

    Article  PubMed  Google Scholar 

  7. Otani T, Iwasaki M, Ikeda S, Kozu T, Saito H, Mutoh M, et al. Serum triglycerides and colorectal adenoma in a case–control study among cancer screening examinees. Cancer Causes and Control. 2006;17:1245–52.

    Article  PubMed  Google Scholar 

  8. Yee YK, Tan VPY, Chan P, Hung IFN, Pang R, Wong BCY. Epidemiology of colorectal cancer in Asia. J Gastroenterol Hepatol. 2009;24:1810–6.

    Article  PubMed  Google Scholar 

  9. Matsuo K, Mizoue T, Tanaka K, Tsuji I, Sugawara Y, Sasazuki S, et al. Association between body mass index and the colorectal cancer risk in Japan: pooled analysis of population-based cohort studies in Japan. Ann Oncol. 2012;23:479–90.

    Article  CAS  PubMed  Google Scholar 

  10. Goh LY, Goh KL. Obesity: an epidemiological perspective from Asia and its relationship to gastrointestinal and liver cancers. J Gastroenterol Hepatol. 2013;28 Suppl 4:54–8.

    Article  PubMed  Google Scholar 

  11. Nagata N, Sakamoto K, Arai T, Niikura R, Shimbo T, Shinozaki M, et al. Visceral abdominal fat measured by computed tomography is associated with an increased risk of colorectal adenoma. Int J Cancer. 2014;135:2273–81.

    Article  CAS  PubMed  Google Scholar 

  12. Riondino S, Roselli M, Palmirotta R, Della-Morte D, Ferroni P, Guadagni F. Obesity and colorectal cancer: role of adipokines in tumor initiation and progression. World J Gastroenterol. 2014;20:5177–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Huang XF, Chen JZ. Obesity, the PI3K/Akt signal pathway and colon cancer. Obes Rev. 2009;10:610–6.

    Article  CAS  PubMed  Google Scholar 

  14. Shebl FM, Hsing AW, Park Y, Hollenbeck AR, Chu LW, Meyer TE, et al. Non-steroidal anti-inflammatory drugs use is associated with reduced risk of inflammation-associated cancers: NIH-AARP study. PLoS One. 2014;9, e114633.

    Article  PubMed  PubMed Central  Google Scholar 

  15. Kim S, Baron JA, Mott LA, Burke CA, Church TR, McKeown-Eyssen GE, et al. Aspirin may be more effective in preventing colorectal adenomas in patients with higher BMI (United States). Cancer Causes Control. 2006;17:1299–304.

    Article  PubMed  Google Scholar 

  16. Movahedi M, Bishoo DT, Macrae F, Mecklin JP, Moeslein G, Oischwang S, et al. Obesity, aspirin, and risk of colorectal cancer in carriers of hereditary colorectal cancer: Prospective investigationin the CAPP2 study. J Clin Oncol. 2015;33:3591–7.

    Article  PubMed  Google Scholar 

  17. Martinez ME, Heddens D, Earnest DL, Bogert CL, Roe D, Einspahr J, et al. Physical activity, body mass index, and prostaglandin E2 levels in rectal mucosa. J Natl Cancer Inst. 1999;91:950–3.

    Article  CAS  PubMed  Google Scholar 

  18. Kawamori T, Uchiya N, Sugimura T, Wakabayashi K. Enhancement of colon carcinogenesis by prostaglandin E2 administration. Carcinogenesis. 2003;24:985–90.

    Article  CAS  PubMed  Google Scholar 

  19. Wang D, Fu L, Sun H, Guo L, DuBois RN. Prostaglandin E2 promotes colorectal cancer stem cell expansion and metastasis in mice. Gastroenterology. 2015;149:1884–95.

    Article  CAS  PubMed  Google Scholar 

  20. Mutoh M, Takahashi M, Wakabayashi K. Roles of prostanoids in colon carcinogenesis and their potential targeting for cancer chemoprevention. Curr Pharm Des. 2006;12:2375–82.

    Article  CAS  PubMed  Google Scholar 

  21. Becker S, Dossus L, Kaaks R. Obesity related hyperinsulinaemia and hyperglycaemia and cancer development. Arch Physiol Biochem. 2009;115:86–96.

    Article  CAS  PubMed  Google Scholar 

  22. Decensi A, Puntoni M, Goodwin P, Cazzaniga M, Gennari A, Bonanni B, et al. Metformin and cancer risk in diabetic patients: a systematic review and meta-analysis. Cancer Prev Res (Phila). 2010;3:1451–61.

    Article  CAS  Google Scholar 

  23. Kasznicki J, Sliwinska A, Drzewoski J. Metformin in cancer prevention and therapy. Ann Transl Med. 2014. doi:10.3978/j.issn.2305-5839.2014.06.01.

    PubMed  PubMed Central  Google Scholar 

  24. Zakikhani M, Dowling RJ, Sonenberg N, Pollak MN. The effects of adiponectin and metformin on prostate and colon neoplasia involve activation of AMP-activated protein kinase. Cancer Prev Res (Phila). 2008;1:369–75.

    Article  CAS  Google Scholar 

  25. Din FV, Valanciute A, Houde VP, Zibrova D, Green KA, Sakamoto K, et al. Aspirin inhibits mTOR signaling, activates AMP-activated protein kinase, and induces autophagy in colorectal cancer cells. Gastroenterology. 2012;142:1504–15.e3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. King TS, Russe OQ, Möser CV, Ferreirós N, Kynast KL, Knothe C, et al. AMP-activated protein kinase is activated by non-steroidal anti-inflammatory drugs. Eur J Pharmacol. 2015;762:299–305.

    Article  CAS  PubMed  Google Scholar 

  27. O'Brien AJ, Villani LA, Broadfield LA, Houde VP, Galic S, Blandino G, et al. Salicylate activates AMPK and synergizes with metformin to reduce the survival of prostate and lung cancer cells ex vivo through inhibition of de novo lipogenesis. Biochem J. 2015;469:177–87.

    Article  PubMed  Google Scholar 

  28. Boffetta P, Hashibe M. Alcohol and cancer. Lancet Oncol. 2006;7:149–56.

    Article  CAS  PubMed  Google Scholar 

  29. Shiotani A, Ishikawa H, Mutoh M, Takeshita T, Nakamura T, Morimoto K, et al. Genetic polymorphisms in ADH1B and ALDH2 are associated with colorectal tumors in Japan. J Cancer Therapy. In press

  30. Lee SL, Lee TP, Wu ML, Chi YC, Liu CM, Lai CL, et al. Inhibition of human alcohol and aldehyde dehydrogenases by aspirin and salicylate: assessment of the effects on first-pass metabolism of ethanol. Biochem Pharmacol. 2015;95:71–9.

    Article  CAS  PubMed  Google Scholar 

  31. Food and Drug Administration, HHS. Organ-specific warnings; internal analgesic, antipyretic, and antirheumatic drug products for over-the-counter human use; final monograph. Final rule Fed Regist. 2009;74:19385–409.

    Google Scholar 

  32. Crews FT, Bechara R, Brown LA, Guidot DM, Mandrekar P, Oak S, et al. Cytokines and alcohol. Alcohol Clin Exp Res. 2006;30:720–30.

    Article  CAS  PubMed  Google Scholar 

  33. Saeed RW, Varma S, Peng T, Tracey KJ, Sherry B, Metz CN. Ethanol blocks leukocyte recruitment and endothelial cell activation in vivo and in vitro. J Immunol. 2004;173:6376–83.

    Article  CAS  PubMed  Google Scholar 

  34. Wimberly AL, Forsyth CB, Khan MW, Pemberton A, Khazaie K, Keshavar-zian A. Ethanol-induced mast cell-mediated inflammation leads to increased susceptibility of intestinal tumorigenesis in the APC Delta468 min mouse model of colon cancer. Alcohol Clin Exp Res. 2013;37:E199–208.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Landi S, Moreno V, Gioia-Patricola L, Guino E, Navarro M, de Oca J, et al. Association of common polymorphisms in inflammatory genes interleukin (IL)6, IL8, tumor necrosis factor alpha, NFKB1, and peroxisome proliferator-activated receptor gamma with colorectal cancer. Cancer Res. 2003;63:3560–6.

    CAS  PubMed  Google Scholar 

  36. Vickers MA, Green FR, Terry C, Mayosi BM, Julier C, Lathrop M, et al. Genotype at a promoter polymor- phism of the interleukin-6 gene is associated with baseline levels of plasma C-reactive protein. Cardiovasc Res. 2002;53:1029–34.

    Article  CAS  PubMed  Google Scholar 

  37. Brull DJ, Montgomery HE, Sanders J, Dhamrait S, Luong L, Rumley A, et al. Interleukin-6 gene -174 g > c and -572 g > c promoter polymorphisms are strong predictors of plasma interleukin-6 levels after coronary artery bypass surgery. Arterioscler Thromb Vasc Biol. 2001;21:1458–63.

    Article  CAS  PubMed  Google Scholar 

  38. Haukim N, Bidwell JL, Smith AJ, Keen LJ, Gallagher G, Kimberly R, et al. Cytokine gene polymorphism in human disease: on-line databases, supplement 2. Genes Immun. 2002;3:313–30.

    Article  CAS  PubMed  Google Scholar 

  39. Campisi J, Andersen JK, Kapahi P, Melov S. Cellular senescence: a link between cancer and age-related degenerative disease? Semin Cancer Biol. 2011;21:354–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  40. He C, Tsuchiyama SK, Nguyen QT, Plyusnina EN, Terrill SR, Sahibzada S, et al. Enhanced longevity by ibuprofen, conserved in multiple species, occurs in yeast through inhibition of tryptophan import. PLoS Genet. 2014;10, e1004860.

    Article  PubMed  PubMed Central  Google Scholar 

  41. Ching TT, Chiang WC, Chen CS, Hsu AL. Celecoxib extends C. elegans lifespan via inhibition of insulin-like signaling but not cyclooxygenase-2 activity. Aging Cell. 2011;10:506–19.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Danilov A, Shaposhnikov M, Shevchenko O, Zemskaya N, Zhavoronkov A, Moskalev A. Influence of non-steroidal anti-inflammatory drugs on Drosophila melanogaster longevity. Oncotarget. 2015;6:19428–44.

    PubMed  PubMed Central  Google Scholar 

  43. Strong R, Miller RA, Astle CM, Floyd RA, Flurkey K, Hensley KL, et al. Nordihydroguaiaretic acid and aspirin increase lifespan of genetically heterogeneous male mice. Aging Cell. 2008;7:641–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Orhan H, Dogruer DS, Cakir B, Sahin G, Sahin MF. The in vitro effects of new non-steroidal antiinflammatory compounds on antioxidant system of human erythrocytes. Exp Toxicol Pathol. 1999;51:397–402.

    Article  CAS  PubMed  Google Scholar 

  45. Orhan H, Sahin G. In vitro effects of NSAIDS and paracetamol on oxidative stress-related parameters of human erythrocytes. Exp Toxicol Pathol. 2001;53:133–40.

    Article  CAS  PubMed  Google Scholar 

  46. Ishino K, Mutoh M, Totsuka Y, Nakagama H. Metabolic syndrome: A novel high-risk state for colorectal cancer. Cancer letts (SpI; Technologies in Carcinogenesis). 2013;334:56–61.

    Article  CAS  Google Scholar 

  47. Ikeda K, Mutoh M, Teraoka N, Nakanishi H, Wakabayashi K, Taguchi R. Increase of oxidant-related triglycerides and phosphatidylcholines in serum and small intestinal mucosa during development of intestinal polyp formation in Min mice. Cancer Sci. 2011;102:79–87.

    Article  CAS  PubMed  Google Scholar 

  48. Mutoh M, Niho N, Komiya M, Takahashi M, Ohtsubo R, Nakatogawa K, et al. Plasminogen activator inhibitor-1 (Pai-1) blockers suppress intestinal polyp formation in Min mice. Carcinogenesis. 2008;29:824–9.

    Article  CAS  PubMed  Google Scholar 

  49. Takasu S, Mutoh M, Takahashi M, Nakagama H. Lipoprotein lipase as a candidate target for cancer prevention/therapy. Biochem Res Int. 2012;2012:398697.

    Article  PubMed  PubMed Central  Google Scholar 

  50. Kourounakis AP, Victoratos P, Peroulis N, Stefanou N, Yiangou M, Hadjipetrou L, et al. Experimental hyperlipidemia and the effect of NSAIDs. Exp Mol Pathol. 2002;73:135–8.

    Article  CAS  PubMed  Google Scholar 

  51. Niho N, Mutoh M, Komiya M, Ohta T, Sugimura T, Wakabayashi K. Improvement of hyperlipidemia by indomethacin in Min mice. Int J Cancer. 2007;121:1665–9.

    Article  CAS  PubMed  Google Scholar 

  52. van Diepen JA, Vroegrijk IO, Berbée JF, Shoelson SE, Romijn JA, Havekes LM, et al. Aspirin reduces hypertriglyceridemia by lowering VLDL-triglyceride production in mice fed a high-fat diet. Am J Physiol Endocrinol Metab. 2011;301:E1099–107.

    Article  PubMed  PubMed Central  Google Scholar 

  53. Goldfine AB, Fonseca V, Jablonski KA, Pyle L, Staten MA, Shoelson SE, et al. The effects of salsalate on glycemic control in patients with Type 2 Diabetes: A randomized trial. Ann Intern Med. 2010;152:346–57.

    Article  PubMed  PubMed Central  Google Scholar 

  54. Ma E, Sasazuki S, Iwasaki M, Sawada N, Inoue M, Tsugane S, et al. 10-Year risk of colorectal cancer: development and validation of a prediction model in middle-aged Japanese men. Cancer Epidemiol. 2010;34:534–41.

    Article  PubMed  Google Scholar 

  55. Mutoh M, Ishikawa H. Metformin use and lung cancer risk--Letter. Cancer Prev Res (Phila). 2015;8:760.

    Article  Google Scholar 

  56. Ishikawa H, Wakabayashi K, Suzuki S, Mutoh M, Hirata K, Nakamura T, et al. Preventive effects of low-dose aspirin on colorectal adenoma growth in patients with familial adenomatous polyposis: Double-blind, randomized clinical study. Cancer Med. 2013;2:50–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Ishikawa H, Mutoh M, Suzuki S, Tokudome S, Saida Y, Abe T, et al. The preventive effects of low-dose enteric-coated aspirin tablets on the development of colorectal tumours in Asian patients: a randomized trial. Gut. 2014;63:1755–9.

    Article  CAS  PubMed  Google Scholar 

  58. Pommergaard HC, Burcharth J, Rosenberg J, Raskov H. Aspirin, calcitriol, and calcium do not prevent adenoma recurrence in a randomized controlled trial. Gastroenterology. 2015 doi: 10.1053/j.gastro.2015.09.010. [Epub ahead of print].

  59. Drew DA, Goh G, Mo A, Grady JJ, Forouhar F, Egan G, et al. Colorectal polyp prevention by daily aspirin use is abrogated among active smokers. Cancer Causes Control. 2015. [Epub ahead of print].

  60. Hankey GJ, Eikelboom JW. Aspirin resistance. Lancet. 2006;367:606–17.

    Article  CAS  PubMed  Google Scholar 

  61. Mirkhel A, Peyster E, Sundeen J, Greene L, Michelson AD, Hasan A, et al. Frequency of aspirin resistance in a community hospital. Am J Cardiol. 2006;98:577–9.

    Article  CAS  PubMed  Google Scholar 

  62. U.S. Preventive Services TASK FORCE. http://www.uspreventiveservicestaskforce.org (2015). Accessed 6 Nov 2015.

  63. American Diabetes Association Standards of Medical Care in Diabetes (2015). http://care.diabetesjournals.org/content/suppl/2014/12/99.pdf. Accessed 6 Nov 2015.

  64. Schjerning Olsen AM, Fosbøl EL, Lindhardsen J, Folke F, Charlot M, Selmer C, et al. Duration of treatment with nonsteroidal anti-inflammatory drugs and impact on risk of death and recurrent myocardial infarction in patients with prior myocardial infarction: a nationwide cohort study. Circulation. 2011;123:2226–35.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported by a Grant-in-Aid for Scientific Research from the Japan Society for the Promotion of Science (JSPS KAKENHI Grant number 25290049), and also supported by Grants-in-Aid for National Cancer Center Research and Development Fund, for the U.S. – Japan Cooperative Medical Science Program, from the Ministry of Health, Labor and Welfare of Japan.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Michihiro Mutoh.

Additional information

Competing interest

The authors declare that they have no competing interests.

Authors’ contributions

TH, GF, SM, MT and MM wrote the manuscript. YT conducted the literature review. JT and KW was involved in the critical revision or supervision of the manuscript. All authors read and approved the final manuscript.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hamoya, T., Fujii, G., Miyamoto, S. et al. Effects of NSAIDs on the risk factors of colorectal cancer: a mini review. Genes and Environ 38, 6 (2016). https://doi.org/10.1186/s41021-016-0033-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s41021-016-0033-0

Keywords