Next Article in Journal
Demethylating Agents in the Treatment of Cancer
Next Article in Special Issue
In Silico Screening of Nonsteroidal Anti-Inflammatory Drugs and Their Combined Action on Prostaglandin H Synthase-1
Previous Article in Journal
Linezolid Resistance in Staphylococci
Previous Article in Special Issue
NSAIDs in the Acute Treatment of Migraine: A Review of Clinical and Experimental Data
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

NSAIDs and Cell Proliferation in Colorectal Cancer

School of Medicine & Medical Science, University College Dublin, C206 Health Sciences Building, Belfield, Dublin 4, Ireland
*
Author to whom correspondence should be addressed.
Pharmaceuticals 2010, 3(7), 2007-2021; https://0-doi-org.brum.beds.ac.uk/10.3390/ph3072007
Submission received: 8 April 2010 / Revised: 17 May 2010 / Accepted: 22 June 2010 / Published: 24 June 2010
(This article belongs to the Special Issue Non-Steroidal Anti-Inflammatory Drugs)

Abstract

:
Colon cancer is common worldwide and accounts for significant morbidity and mortality in patients. Fortunately, epidemiological studies have demonstrated that continuous therapy with NSAIDs offers real promise of chemoprevention and adjunct therapy for colon cancer patients. Tumour growth is the result of complex regulation that determines the balance between cell proliferation and cell death. How NSAIDs affect this balance is important for understanding and improving treatment strategies and drug effectiveness. NSAIDs inhibit proliferation and impair the growth of colon cancer cell lines when tested in culture in vitro and many NSAIDs also prevent tumorigenesis and reduce tumour growth in animal models and in patients, but the relationship to inhibition of tumour cell proliferation is less convincing, principally due to gaps in the available data. High concentrations of NSAIDs are required in vitro to achieve cancer cell inhibition and growth retardation at varying time-points following treatment. However, the results from studies with colon cancer cell xenografts are promising and, together with better comparative data on anti-proliferative NSAID concentrations and doses (for in vitro and in vivo administration), could provide more information to improve our understanding of the relationships between these agents, dose and dosing regimen, and cellular environment.

1. Introduction

Colon cancer is common worldwide. Nearly a million people develop the disease every year and in the United States alone, there are 50,000 cancer-related deaths annually [1]. The mechanisms that lead to and sustain carcinogenesis have been investigated extensively and are multifactorial. Irrespective of the initiating or sustaining causes, however, the primary manifestation of cancer is excessive growth of cells and tissue. Growth of normal and abnormal tissue is achieved by a balance between cell production and cell death i.e. proliferation that is counterbalanced by apoptosis and necrosis [2].
There is a wealth of experimental evidence demonstrating that colon cancer is associated with dysregulation and overexpression of the prostaglandin-synthesizing enzyme cyclooxygenase (COX) as well as accompanying overproduction of prostaglandin, abnormal cell and tissue adjustments in relation to vascularization, cell adhesion, apoptosis and proliferation. One important theme in the body of evidence is the fact that non-steroidal anti-inflammatory drugs (NSAIDs) are able to alter intestinal tumour growth rates and modulate carcinogenesis by a variety of reported methods including inhibition of COX activity and disruption of prostaglandin homeostasis, interruption of nuclear factor kappa B (NF-kB) signaling [3,4,5,6], and of extracellular signal-regulated kinases (ERK/MAPK) [7], induction of various apoptotic pathways [8,9,10], as well as effects on cell cycling [11,12,13,14]. All of these mechanisms either contribute and enhance, or antagonise and counterbalance, the proliferative behaviour that is observed in tumour cells.
This review will focus on assessment and evaluation of the data that is currently available on the effects on cell proliferation in the enteric epithelium from pharmacological intervention with NSAIDs.

2. Wnt and Colon Cancer

A majority of NSAIDs inhibit the cyclooxygenase enzymes (COX-1 and COX-2) and in the process interrupt the biosynthetic conversion of arachidonic acid to eicosanoids [15]. COX-1 is expressed widely in tissues, including the gastric mucosa and platelets, while the normally low level of COX-2 is rapidly increased via induction by pro-inflammatory cytokines [15,16]. Increased levels of expression of COX-2 are associated with colorectal tumours: up to 85% of colorectal adenomas and carcinomas express COX-2 [17,18,19,20].
The Wnt pathway plays an important role in the regulation of cell proliferation and differentiation. Disruption of this carefully balanced regulation leads to disordered proliferation. Wnt signalling helps to control the levels of cellular beta catenin, between pools bound to adenomatous polyposis coli (APC) and to the cell adhesion molecule E-cadherin. Wnt ligand signalling leads to interaction with membrane receptor proteins (frizzled proteins) that are receptors for Wnt ligands. The interaction triggers a signalling cascade that alters the relationship between the scaffold protein Axin and glycogen synthase kinase-3beta (GSK3beta). Axin binds adenomatous polyposis coli APC to the multidocking protein beta-catenin. GSK3beta phosphorylates beta catenin for ubiquitin-mediated proteosomal degradation [21,22].
Wnt-induced alteration of Axin interaction leads to release of beta catenin from the APC-GSK complex so that unphosphorylated beta catenin is stabilised in the cytoplasm and able to translocate to the nucleus [23,24,25,26,27,28] where it associates with transcription factors of the TCF/Lef family and other co-factors to form complexes that activate downstream target genes that regulate proliferation, differentiation and apoptosis: c-myc, cyclin D1 and COX-2 [29,30,31,32].
As with many cancers, colorectal cancers develop from accumulated mutations, deletions or truncations in oncogenes and tumour suppressor genes such as APC, ras and p53 [33]. The loss of functional APC is associated with colon cancer and this is evident in mice with mutant APC: these APCmin mice develop multiple intestinal tumours [34]. Similar mutations in APC in humans results in the hereditary condition familial adenomatous polyposis (FAP) in which multiple colonic polyps develop in patients, with a potential for these polyps to progress and become adenomatous or carcinomatous [35]. These polyps show significant expression of COX-2 and therefore represent a target for therapeutic control with NSAIDs [36,37].

3. NSAIDs Inhibit Colon Cancer Cell Proliferation in vitro

A variety of colorectal tumour cell lines have been used in in vitro investigations of the effects of NSAIDs on several aspects of tumour initiation and progression (Table 1).
When HT29 colorectal adenoma cell lines are treated with different NSAIDs, proliferation is reduced as early as 48 hours after treatment with naproxen and piroxicam. Similar anti-proliferative effects have been reported with indomethacin after 72 hours [38], aspirin after 96 hours [38], NS398 after 72 hours [39], 5-aminosalicylic acid after 48 hours [40], and with sulindac sulfide after 24 hours [41]. However, Piazza and colleagues [42] were unable to inhibit growth in these same cell lines with aspirin, naproxen or salicylic acid [42], but found that ibuprofen and sulindac sulfoxide inhibited growth with a low potency in contrast to higher potency shown with indomethacin, sulindac sulfide and diclofenac. They also suggested that their data (for these compounds) showed no relationship between potencies for inhibition of COX, and effects on growth or apoptosis.
With HCA-7 colorectal carcinoma cells, reduced proliferation was evident following treatment with celecoxib within 12 hours [43]. In RKO rectal cancer cell lines proliferation was inhibited after 72 hours following exposure to NS398 and sulindac [39] and with HT115 colon cancer cell lines, anti-proliferative effects were reported within 24 hours following treatment with 5-aminosalicylic acid. Using SW620 cell lines, the inhibition of proliferation by indomethacin and sulindac is apparent between 24–72 hours after administration of these agents to culture [44,45]. All of these colon cancer cell lines express COX.
When cell lines that do not express COX are treated with NSAIDs, proliferation is also inhibited. Celecoxib reduces proliferation in HCT116 colon cancer cells after 72 hours [46] and the anti-proliferative effect seen in these cells with indomethacin and nimesulide occurs earlier [47]. Similarly in HT15 colon cancer cell lines, treatment with celecoxib, sulindac and piroxicam inhibits proliferation at various timepoint between 24–72 hours and in DLD-1 colon cancer cell lines, proliferation is reduced after 24 hours when treatment with 5-aminosalicylic acid is applied [48]. These reports suggest that the anti-proliferative effect of these NSAIDs may not be related to their ability to inhibit cyclooxygenase and that non-COX mechanisms might be involved. This is reflected in the extensive data that has been generated from studies that have investigated other pathways and mechanisms: NF-kB, ERK/MAPK, peroxisome proliferator-activated receptor gamma (PPARγ), caspases, and cyclins.
Most of the evidence allows the conclusion that NSAIDs inhibit proliferation of colon cancer cells in vitro and that the mechanism of action for the anti-proliferative activity shown by NSAIDs appears to be unrelated to cellular COX expression and activity.
Table 1. Colon cancer cell lines and the concentration of NSAIDs used in studies that show growth inhibition. The time-point at which inhibition of cell growth is recorded is also presented. Non-COX-expressing cell lines are presented in the lower part of the table. Studies are identified according to the numerical listing given in the bibliography. 1 number of viable/attached cells, 2 MTT assay, 3 CFSE labeling, flow-cytometry, 4 growth ratio: number of treated/untreated cells. HT29, HCA7, HT115 and SW620 express COX-2; RKO express COX-1 and COX-2. 5-ASA = 5-aminosalicylic acid, CFSE = carboxyfluorescein diacetate succinimidyl ester, MTT = 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide.
Table 1. Colon cancer cell lines and the concentration of NSAIDs used in studies that show growth inhibition. The time-point at which inhibition of cell growth is recorded is also presented. Non-COX-expressing cell lines are presented in the lower part of the table. Studies are identified according to the numerical listing given in the bibliography. 1 number of viable/attached cells, 2 MTT assay, 3 CFSE labeling, flow-cytometry, 4 growth ratio: number of treated/untreated cells. HT29, HCA7, HT115 and SW620 express COX-2; RKO express COX-1 and COX-2. 5-ASA = 5-aminosalicylic acid, CFSE = carboxyfluorescein diacetate succinimidyl ester, MTT = 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide.
Cell LineNSAIDDose uMEffect ProliferationTime-PointRef.
COX-expressing
HT-29Aspirin400down196 h[26]
HT-29Indomethacin100down172 h[26]
HT-29Naproxen200down148 h[26]
HT-29Piroxicam300down148 h[26]
HT-29sulindac sulfide185down124 h[29]
HT29sulindac sulfide120down124 h[30]
HT29sulindac sulfone150down172 h[27]
HT29NS398120down172 h[27]
HT295-ASA 36000down248 h[28]
HCA-7celecoxib50down212 h[31]
RKONS398120down172 h[27]
RKOsulindac sulfone150down172 h[27]
HT1155-ASA5000down324 h[36]
SW620sulindac1600down224–72 h[32]
SW620indomethacin400down296 h[33]
Non-COX-expressing
HCT116indomethacin600down124–72 h[35]
HCT116nimesulide100down124–72 h[35]
HCT116celecoxib12.5down472 h[34]
HT-15celecoxib50down212 h[31]
HT-15sulindac sulfide200down124 h[29]
HT-15piroxicam900down172 h[29]
DLD-15-ASA5000down324 h[36]

4. Anti-Proliferative Dose Range of NSAIDs in vitro

Much of the available evidence indicates that most NSAIDs inhibit tumour cell proliferation in vitro. However, these effects are influenced by several factors including micromolar drug concentrations, duration of treatment, and timepoint at which anti-proliferative effects are assayed. Sulindac sulfone, a sulindac metabolite that does not have anti-inflammatory properties, is anti-proliferative in HT29 COX-expressing colon cancer cell lines within 72 hours when delivered at a concentration of 150 μM, whereas the active anti-inflammatory sulfide metabolite of sulindac inhibits proliferation within 24 hours when administered at a concentration of 120–185 μM [39,41,42]. Other studies in SW620 colon cancer cell lines have employed concentrations of sulindac of up to 1,600 μM to inhibit proliferation in 24–72 hours [44]. Concentrations of between 100–400 μM are required to inhibit proliferation in COX-expressing colon cancer cell lines using aspirin, indomethacin, naproxen and piroxicam [38,45].
In tumour cell lines that do not express COX, a variety of concentrations of NSAIDs have been shown to inhibit proliferation—ranging from 100 μM of nimesulide to 600 μM of indomethacin in HCT116 colon cancer cell lines [47]. Interestingly, concentrations as low as 12.5–50 μM of celecoxib have been employed to reduce proliferation in these cells [43,46]. Many of these studies have reported the effects of various concentrations of NSAIDs on proliferation in tumour cell lines but few have investigated these effects simultaneously in COX-expressing and non-COX-expressing cell lines while controlling for drug concentration and time. For instance, 300 μM of piroxicam inhibits proliferation in HT29 COX-expressing colon cancer cells after 48 hours, but 900 μM of the drug is anti-proliferative in non-COX-expressing HT-15 colon cancer cells after 72 hours [38,41]. Two separate studies using indomethacin in HT29 and non-COX-expressing HCT116 colon cancer cell lines demonstrated anti-proliferative effects with concentrations of 100 μM and 600 μM of the drug, respectively, after 72 hours [38,47]. It is difficult to draw meaningful conclusions about the relative contribution of the individual influences of COX, concentration and time from the data in these studies. Stolfi and co-workers [48] have recently shown that a high concentration (5,000 μM) of the drug mesalazine (5-aminosalicylic acid) inhibits proliferation after 24 hours in COX-expressing HT115 and DLD-1 non-COX-expressing colon cancer cell lines. Further carefully designed investigations are required to provide data on the individual and collective influences of COX, in vitro drug concentrations and duration of treatment for various NSAIDs.
Given the wide range of doses of the different NSAIDs that have been used and shown to inhibit proliferation, it is reasonable to conclude that for any given NSAID, it is possible to find an appropriate concentration that will inhibit proliferation if a wide range of concentrations is tested.

5. NSAIDs Inhibit Colon Cancer Cell Proliferation in vivo

Most of the available data on the effect of NSAIDs on colonic tumour proliferation in vivo relate to studies carried out using animal models of colonic tumours (Table 2). Colonic tumours have been induced chemically using N-methyl-N-nitrosourea, 1,2-dimethylhydrazine (DMH) or azoxymethane (AOM). One common genetic model that has been employed is the APC min mouse because it is a closely mimics human familial adenomatous polyposis (FAP), a condition in which multiple polyps develop in the colon. These APC min mice also develop numerous polyps in the small intestine and this allows for investigations that enumerate and compare polyps between treatment regimes. The administration of various NSAIDs in these animal models has been shown to be associated with inhibition of initiation and progression of polyps as well as a reduction in the tumour load. Mahmoud and colleagues [49] showed that longterm administration of sulindac sulfide in drinking water (11 weeks) reduced the development of small intestinal polyps in APC min mice, an effect that was absent in the colon [49,50]. However in DMH-treated mice, although development of colonic tumours was prevented by treatment with sulindac for 24 weeks, colonic cell proliferation increased after 18 weeks of sulindac treatment [51,52]. Similar NSAID-induced chemoprevention in animal models has been reported with indomethacin [53], aspirin [54], piroxicam [55], celecoxib [56,57], and nimesulide [58]. All of these studies are characterized by the administration of NSAIDs over a lengthy treatment period ranging from 6–46 weeks.
Table 2. The doses of NSAIDs administered in vivo in tumour model experiments and their effect on initiation and progression of carcinogenesis. The duration of treatment is presented and the effect on proliferation is indicated. NMNU = n-methyl-N-nitrosourea, AOM = azoxymethane, DMH = 1,2-dimethylhydrazine, APC = adenomatous polyposis coli, NA = not measured.
Table 2. The doses of NSAIDs administered in vivo in tumour model experiments and their effect on initiation and progression of carcinogenesis. The duration of treatment is presented and the effect on proliferation is indicated. NMNU = n-methyl-N-nitrosourea, AOM = azoxymethane, DMH = 1,2-dimethylhydrazine, APC = adenomatous polyposis coli, NA = not measured.
NSAIDDose ppmDurationProliferationInhibition EffectModelRodentRef.
Small Intestine
celecoxib1,500 ppm6 wksNAtumor numberAPCC57BL[44]
piroxicam50–200 ppm6 wksNAtumor numberAPCC57BL[43]
sulindac sulfide20 mg/kg11 wksNAtumorigenesisAPCC57BL[37]
Colon
indomethacin10 ppm1–30 wksNAtumorigenesisNMNUFisher rats[41]
aspirin200–400 ppm52 wksNAtumorigenesisAOMF344 rats[42]
nimesulide0.04% w/w14 wksdownNAAOMCD-1 mice[46]
celecoxib300 ppm46 wksdownNAAOMF344 rats[45]
celecoxib1,500 ppm6 wksNAtumor numberAPCC57BL[44]
sulindac5 mg/kg18 wksupNADMHBALB/C[40]
sulindac5 mg/kg24 wksNAtumorigenesisDMHBALB/C[39]
sulindac160 ppm10 wksNAno changeAPCC57BL[38]
sulindac sulfide20 mg/kg11wksNAno changeAPCC57BL[37]
Prevention of tumorigenesis or reduction in tumor load can be a reflection of decreased cell proliferation or of increased cell death. For many of these studies, the data on cell proliferation or cell death was either not sought or else not presented. Whereas the data for NSAID effects on cell proliferation from in vitro studies is unequivocal in their demonstration of anti-proliferation, the limited data on cell proliferation from in vivo studies using animal models of colon cancer shows more conflicting findings. Indomethacin, aspirin and piroxicam have beneficial effects in tumorigenesis and reduction in colonic tumour load in vivo but whether these effects are related to anti-proliferative action by the NSAIDs is not certain [53,54,55]. Celecoxib treatment reduces tumor numbers and inhibits cell proliferation [56,57]. The data on the various forms of sulindac suggests that the effect of this drug on tumorigenesis is variable and may be dependent on the animal model employed [49,50,51,52].
Some of this inconsistency in the findings from studies in animal models is reflected in the results of investigations in patients. In a study of patients with FAP, treatment with sulindac (150 mg twice daily) for nine months reduced the number and size of colorectal adenomas [59]. A second study found that treatment with standard doses of sulindac (25–150 mg twice daily) for 48 months did not prevent adenomas in patients [60]. However several clinical trials have shown beneficial prevention of colorectal cancer in patients following treatments with aspirin and celecoxib [61,62,63].

6. Anti-Proliferative Dose Range of NSAIDs in vivo

Many of the studies in animal models have used doses of NSAIDs that would not ordinarily be employed clinically in patients. There are however very few studies that have used clinically relevant doses of NSAIDs to investigate colon cancer in animal models. Moorghen and co-workers [52] examined cell proliferation in the colonic mucosal epithelium in mice following treatment with low-dose sulindac for 18 weeks. Their finding of sulindac-mediated increase in cell proliferation led them to question the rationale behind the therapeutic manipulation of crypt cell proliferation in order to reduce the risk of colon cancer.
While the main focus has been on investigating the treatment of colon cancer with NSAIDs, very little attention has been given to the effect on or the response of the normal bowel following NSAID treatment (Table 3). For example, what happens to proliferation in the unaffected (non-adenomatous) parts of intestine in colon cancer models following treatment with NSAIDs? The distinction between affected and unaffected parts of the bowel is important because COX-2 is upregulated in 90% of human colon cancer and 40% of premalignant adenomas of the colon but is absent in the normal colon [64,65]. This provides some of the rationale for in vitro studies that employ non-COX-expressing colon cancer cell lines. Hollingshead and colleagues [47] found that proliferation was inhibited in HCT116 non-COX-expressing colon cancer cells and in normal colonic epithelium in mice when treated with different in vitro and in vivo doses of nimesulide for 72 hours and five days respectively. With a large dose of celecoxib administered over 45 days, Williams and co-workers [43] found that mitosis remained normal in the small and large bowel. In the same study, they found that growth of HCA-7 colon cancer cell xenografts reduced with an identical treatment regime with celecoxib. Similar growth inhibition of xenografted colon cancer cells has been observed with the sulindac derivative SS amide [66].
When CD-1 mice were treated twice daily with clinically relevant doses of nimesulide or indomethacin and examined after six and 24 hours, changes in small intestinal cell proliferation were found to be dependent on enteric region and duration of treatment [67,68]. Indomethacin down-regulated cell production in proximal intestinal crypts after six hours while nimesulide reduced cell proliferation in proximal and distal intestinal crypts. After 24 hours of treatment with either drug, the numbers of proliferating cells in the crypts showed regional variation with increases in some regions and reductions in others. The number of cells produced by these crypts however was downregulated in indomethacin-treated mice but remained, in nimesulide-treated mice, similar to the numbers in untreated mice.
Table 3. NSAIDs and enteric cell proliferation in vivo. The duration of treatment is indicated.
Table 3. NSAIDs and enteric cell proliferation in vivo. The duration of treatment is indicated.
NSAIDDoseDurationProliferationModelRef.
Small Intestine
celecoxib1,250 mg/kg chow 45 daysNo changeC57BL[31]
indomethacin10 mg/kg body weight6 hvariableCD-1[55]
indomethacin10 mg/kg body weight24 hvariableCD-1[56]
nimesulide10 mg/kg body weight6 hdownCD-1[55]
nimesulide10 mg/kg body weight24 hvariableCD-1[56]
Colon
celecoxib1,250 mg/kg chow45 daysNo changeC57BL[31]
nimesulide400 mg/kg5 daysdownC57BL[35]
Duration of treatment with NSAIDs is also important. While it is recognized that there will be different regimes (dose, duration) for each NSAID, drawing conclusions from the available evidence is impaired by the variety in application of treatment regimes in these studies. There may be a need to begin to differentiate between short term (acute) and longterm (adaptive) treatment regimes with respect to the anti-proliferative effects of NSAIDs in the colon. Most of the in vitro studies have reported effects for NSAIDs over a time period of between 24–96 hours. There is a need for correspondingly short in vivo investigations with NSAIDs in order to allow comparisons of the data and a better understanding of the relationships between drug, cell environment and measured endpoint. There is also a need to explore ways of extending the duration of in vitro studies with colon cancer cell lines in order to narrow the gap between the in vitro study design and in vivo treatment regimes.

7. NSAIDs and Other Cancers

While the data reviewed so far has been obtained from studies examining influences on colorectal cancer, there is extensive literature on the relationship between various NSAIDs and other cancers in the skin, stomach, breast, prostate, pancreas, ovary and the urinary bladder (extensively reviewed by Baron) [69]. While the findings from these have contributed to our understanding of the complexity of the relationship between NSAIDs and tumours, data evaluations from population-based studies continue to provide conflicting conclusions.
Some epidemiological studies suggest that the use of aspirin do not reduce the risk of PSA-detected prostate cancer while other investigations have found that aspirin may be useful in populations with a high risk of developing prostate cancer [70,71]. The relationship between NSAIDs and prostate cancer also appears to depend on prior, regular or current usage of these drugs [72,73]. Conflicting outcomes have also been reported from studies of breast cancer risk and usage of various NSAIDs [74,75] and led Cuzick et al. [76] to conclude that, despite the wealth of published reports, there is still insufficient data on risk-benefit profiling of NSAIDs for use in cancer chemoprevention. They suggest that more controlled trials with new study designs are necessary.
With these non-colorectal cancers, the mechanisms by which NSAIDs affect tumour behaviour are also variable and include disruptions of signalling pathways as well as interruptions of metabolism of other therapeutic agents (if used in combination treatments) [77].

8. Conclusions

The available evidence suggest the followings:
  • At an appropriate concentration in vitro, most NSAIDs inhibit proliferation in colon cancer cell lines.
  • When administered in vitro to colon cancer cell lines in culture, the limited evidence available indicates that clinically realistic doses of NSAIDs do not inhibit cell proliferation. There is evidence demonstrating that clinical doses of NSAIDs inhibit proliferation and reduce tumour growth in xenografted tumours derived from colon cancer cell lines.
  • Acute effects on proliferation seen in vitro with NSAIDs treatment cannot be tested/reproduced following treatment in vivo because the concentrations used in vitro are toxic and lethal to cells, tissue and organs in vivo.
  • Many NSAIDs will prevent carcinogenesis and slow tumour growth in animal models but the limited data relating to the ability of NSAIDs to inhibit tumour proliferation is mixed.
  • The concentrations of NSAIDs required to inhibit colorectal cancer cell proliferation in vitro are much higher than the doses required to inhibit cell proliferation in colonic tumours in vivo.
  • In studies with colon cancer cell lines, it has proven difficult to reproduce the adaptive effects on proliferation seen in vivo following NSAIDs administration, due largely to methodological and technical limitations imposed by a cell culture approach.
  • A distinction between acute effects (within the first 96 hours) and adaptive effects (long term treatment) that are observed when treatment is given over weeks and months may be necessary in order to allow more useful and appropriate comparisons between data sets.
  • Better comparative data is required from in vitro and in vivo studies using low doses of NSAIDs over short and long durations of treatment, and from studies of NSAID effects in normal colonic tissue.
The published evidence from clinical studies and patient therapy indicates that aspirin, sulindac and celecoxib offer promise for effective use in colon cancer. Further studies are needed to establish useful in vitro and in vivo rodent models that reproduce the clinical data in terms of dosages, duration of treatment and effect on tumour tissue and cells. These models might then provide the basis for targeted experiments and investigations into mechanisms of actions of the NSAIDs used.

References

  1. Jemal, A.; Murray, T.; Ward, E.; Samuels, A.; Tiwari, R.C.; Ghafoor, A.; Feuer, E.J.; Thun, M.J. Cancer statistics, 2005. CA Cancer J. Clin. 2005, 55, 10–30. [Google Scholar]
  2. Raz, A. Is inhibition of cyclooxygenase required for the anti-tumorigenic effects of nonsteroidal, anti-inflammatory drugs (NSAIDs)? In vitro versus in vivo results and the relevance for the prevention and treatment of cancer. Biochem. Pharmacol. 2002, 63, 343–347. [Google Scholar] [PubMed]
  3. Kopp, E.; Ghosh, S. Inhibition of NF-κB by sodium salicylate and aspirin. Science 1994, 265, 956–959. [Google Scholar]
  4. Yin, M.J.; Yamamoto, Y.; Gaynor, R.B. The anti-inflammatory agents aspirin and salicylate inhibit the activity of IκB kinase-β. Nature 1998, 396, 77–80. [Google Scholar]
  5. Stark, L.A.; Din, F.V.; Zwacka, R.M.; Dunlop, M.G. Aspirin-induced activation of the NF-kappaB signaling pathway: A novel mechanism for aspirin-mediated apoptosis in colon cancer cells. FASEB J. 2001, 15, 1273–1275. [Google Scholar]
  6. Loveridge, C.J.; MacDonald, A.D.; Thoms, H.C.; Dunlop, M.G.; Stark, L.A. The proapoptotic effects of sulindac, sulindac sulfone and indomethacin are mediated by nucleolar translocation of the RelA(p65) subunit of NF-κB. Oncogene 2008, 27, 2648–2655. [Google Scholar]
  7. Pan, M.R.; Chang, H.C.; Hung, W.C. Non-steroidal anti-inflammatory drugs suppress the ERK signaling pathway via block of Ras/c-Raf interaction and activation of MAP kinase phosphatases. Cell Signal. 2008, 20, 1134–1141. [Google Scholar]
  8. Gu, Q.; Wang, J.D.; Xia, H.H.; Lin, M.C.; He, H.; Zou, B.; Tu, S.P.; Yang, Y.; Liu, X.G.; Lam, S.K.; Wong, W.M.; Chan, A.O.; Yuen, M.F.; Kung, H.F.; Wong, B.C. Activation of the caspase-8/Bid and Bax pathways in aspirin-induced apoptosis in gastric cancer. Carcinogenesis 2005, 26, 541–546. [Google Scholar]
  9. Jana, N.R. NSAIDs and apoptosis. Cell. Mol. Life Sci. 2008, 65, 1295–1301. [Google Scholar]
  10. Vaish, V.; Tanwar, L.; Sanyal, S.N. The role of NF-κB and PPARgamma in experimentally induced colorectal cancer and chemoprevention by cyclooxygenase-2 inhibitors. Tumour Biol. 2010. [Epub ahead of print]. [Google Scholar]
  11. Huang, Y.C.; Chuang, L.Y.; Hung, W.C. Mechanisms underlying nonsteroidal anti-inflammatory drug-induced p27(Kip1) expression. Mol. Pharmacol. 2002, 62, 1515–1521. [Google Scholar]
  12. Bock, J.M.; Menon, S.G.; Goswami, P.C.; Sinclair, L.L.; Bedford, N.S.; Domann, F.E.; Trask, D.K. Relative non-steroidal anti-inflammatory drug (NSAID) antiproliferative activity is mediated through p21-induced G1 arrest and E2F inhibition. Mol. Carcinog. 2007, 46, 857–864. [Google Scholar]
  13. Luciani, M.G.; Campregher, C.; Gasche, C. Aspirin blocks proliferation in colon cells by inducing a G1 arrest and apoptosis through activation of the checkpoint kinase ATM. Carcinogenesis 2007, 28, 2207–2217. [Google Scholar]
  14. Chen, X.J.; Xiao, W.; Qu, X.; Zhou, S.Y. NS-398 enhances the efficacy of gemcitabine against lung adenocarcinoma through up-regulation of p21WAF1 and p27KIP1 protein. Neoplasma 2008, 55, 200–204. [Google Scholar]
  15. Botting, R.M. Inhibitors of cyclooxygenases: mechanisms, selectivity and uses. J. Physiol. Pharmacol. 2006, 57 Suppl. 5, 113–124. [Google Scholar]
  16. Jones, D.A.; Carlton, D.P.; McIntyre, T.M.; Zimmerman, G.A.; Prescott, S.M. Molecular cloning of human prostaglandin endoperoxide synthase type II and demonstration of expression in response to cytokines. J. Biol. Chem. 1993, 268, 9049–9054. [Google Scholar]
  17. Rigas, B.; Goldman, I.S.; Levine, L. Altered eicosanoid levels in human colon cancer. J. Lab. Clin. Med. 1993, 122, 518–523. [Google Scholar]
  18. Eberhart, C.E.; Coffey, R.J.; Radhika, A.; Giardiello, F.M.; Ferrenbach, S.; DuBois, R.N. Up-regulation of cyclooxygenase 2 gene expression in human colorectal adenomas and adenocarcinomas. Gastroenterology 1994, 107, 1183–1188. [Google Scholar]
  19. Sheng, H.; Shao, J.; Kirkland, S.C.; Isakson, P.; Coffey, R.J.; Morrow, J.; Beauchamp, R.D.; DuBois, R.N. Inhibition of human colon cancer cell growth by selective inhibition of cyclooxygenase-2. J. Clin. Invest. 1997, 99, 2254–2259. [Google Scholar]
  20. Fujita, T.; Matsui, M.; Takaku, K.; Uetake, H.; Ichikawa, W.; Taketo, M.M.; Sugihara, K. Size- and invasion-dependent increase in cyclooxygenase 2 levels in human colorectal carcinomas. Cancer Res. 1998, 58, 4823–4826. [Google Scholar]
  21. Kikuchi, A. Tumor formation by genetic mutations in the components of the Wnt signaling pathway. Cancer Sci. 2003, 94, 225–229. [Google Scholar]
  22. Pinto, D.; Clevers, H. Wnt control of stem cells and differentiation in the intestinal epithelium. Exp. Cell Res. 2005, 306, 357–363. [Google Scholar]
  23. Yanagawa, S.; van Leeuwen, F.; Wodarz, A.; Klingensmith, J.; Nusse, R. The dishevelled protein is modified by wingless signaling in Drosophila. Gene.Develop. 1995, 9, 1087–1097. [Google Scholar] [CrossRef]
  24. Yost, C.; Torres, M.; Miller, J.R.; Huang, E.; Kimelman, D.; Moon, R.T. The axis-inducing activity, stability, and subcellular distribution of beta-catenin is regulated in Xenopus embryos by glycogen synthase kinase 3. Gene.Develop. 1996, 10, 1443–1454. [Google Scholar] [CrossRef]
  25. Fagotto, F.; Gluck, U.; Gumbiner, B.M. Nuclear localization signal-independent and importin/karyopherin-independent nuclear import of β-catenin. Curr. Biol. 1998, 8, 181–190. [Google Scholar]
  26. Kishida, S.; Yamamoto, H.; Hino, S.; Ikeda, S.; Kishida, M.; Kikuchi, A. DIX domains of Dvl and axin are necessary for protein interactions and their ability to regulate beta-catenin stability. Mol. Cell. Biol. 1999, 19, 4414–4422. [Google Scholar]
  27. Yamamoto, H.; Kishida, S.; Kishida, M.; Ikeda, S.; Takada, S.; Kikuchi, A. Phosphorylation of axin, a Wnt signal negative regulator, by glycogen synthase kinase-3β regulates its stability. J. Biol. Chem. 1999, 274, 10681–10684. [Google Scholar] [PubMed]
  28. Coghlan, M.P.; Culbert, A.A.; Cross, D.A.; Corcoran, S.L.; Yates, J.W.; Pearce, N.J.; Rausch, O.L.; Murphy, G.J.; Carter, P.S.; Roxbee Cox, L.; Mills, D.; Brown, M.J.; Haigh, D.; Ward, R.W.; Smith, D.G.; Murray, K.J.; Reith, A.D.; Holder, J.C. Selective small molecule inhibitors of glycogen synthase kinase-3 modulate glycogen metabolism and gene transcription. Chem. Biol. 2000, 7, 793–803. [Google Scholar]
  29. He, T.C.; Sparks, A.B.; Rago, C.; Hermeking, H.; Zawel, L.; da Costa, L.T.; Morin, P.J.; Vogelstein, B.; Kinzler, K.W. Identification of c-MYC as a target of the APC pathway. Science 1998, 281, 1509–1512. [Google Scholar]
  30. Shtutman, M.; Zhurinsky, J.; Simcha, I.; Albanese, C.; D'Amico, M.; Pestell, R.; Ben-Ze'ev, A. The cyclin D1 gene is a target of the β-catenin/LEF-1 pathway. Proc. Nat. Acad. Sci. USA 1999, 96, 5522–5527. [Google Scholar]
  31. Tetsu, O.; McCormick, F. β-catenin regulates expression of cyclin D1 in colon carcinoma cells. Nature 1999, 398, 422–426. [Google Scholar] [CrossRef] [PubMed]
  32. Araki, Y.; Okamura, S.; Hussain, S.P.; Nagashima, M.; He, P.; Shiseki, M.; Miura, K.; Harris, C.C. Regulation of cyclooxygenase-2 expression by the Wnt and ras pathways. Cancer Res. 2003, 63, 728–734. [Google Scholar]
  33. Vogelstein, B.; Kinzler, K.W. Cancer genes and the pathways they control. Nature Med. 2004, 10, 789–799. [Google Scholar]
  34. Oshima, M.; Oshima, H.; Kitagawa, K.; Kobayashi, M.; Itakura, C.; Taketo, M. Loss of Apc heterozygosity and abnormal tissue building in nascent intestinal polyps in mice carrying a truncated Apc gene. Proc. Nat. Acad. Sci. USA 1995, 92, 4482–4486. [Google Scholar]
  35. Levy, D.B.; Smith, K.J.; Beazer-Barclay, Y.; Hamilton, S.R.; Vogelstein, B.; Kinzler, K.W. Inactivation of both APC alleles in human and mouse tumors. Cancer Res. 1994, 54, 5953–5958. [Google Scholar]
  36. Takeda, H.; Sonoshita, M.; Oshima, H.; Sugihara, K.; Chulada, P.C.; Langenbach, R.; Oshima, M.; Taketo, M.M. Cooperation of cyclooxygenase 1 and cyclooxygenase 2 in intestinal polyposis. Cancer Res. 2003, 63, 4872–4877. [Google Scholar]
  37. Swamy, M.V.; Patlolla, J.M.; Steele, V.E.; Kopelovich, L.; Reddy, B.S.; Rao, C.V. Chemoprevention of familial adenomatous polyposis by low doses of atorvastatin and celecoxib given individually and in combination to APCMin mice. Cancer Res. 2006, 66, 7370–7377. [Google Scholar]
  38. Shiff, S.J.; Koutsos, M.I.; Qiao, L.; Rigas, B. Nonsteroidal antiinflammatory drugs inhibit the proliferation of colon adenocarcinoma cells: Effects on cell cycle and apoptosis. Exp. Cell Res. 1996, 222, 179–188. [Google Scholar]
  39. Shureiqi, I.; Chen, D.; Lotan, R.; Yang, P.; Newman, R.A.; Fischer, S.M.; Lippman, S.M. 15-Lipoxygenase-1 mediates nonsteroidal anti-inflammatory drug-induced apoptosis independently of cyclooxygenase-2 in colon cancer cells. Cancer Res. 2000, 60, 6846–6850. [Google Scholar]
  40. Kim, Y.H.; Kim, M.H.; Kim, B.J.; Kim, J.J.; Chang, D.K.; Son, H.J.; Rhee, P.L.; Rhee, J.C. Inhibition of cell proliferation and invasion in a human colon cancer cell line by 5-aminosalicylic acid. Dig. Liver Dis. 2009, 41, 328–337. [Google Scholar]
  41. Hanif, R.; Pittas, A.; Feng, Y.; Koutsos, M.I.; Qiao, L.; Staiano-Coico, L.; Shiff, S.I.; Rigas, B. Effects of nonsteroidal anti-inflammatory drugs on proliferation and on induction of apoptosis in colon cancer cells by a prostaglandin-independent pathway. Biochem. Pharmacol. 1996, 52, 237–245. [Google Scholar]
  42. Piazza, G.A.; Rahm, A.K.; Finn, T.S.; Fryer, B.H.; Li, H.; Stoumen, A.L.; Pamukcu, R.; Ahnen, D.J. Apoptosis primarily accounts for the growth-inhibitory properties of sulindac metabolites and involves a mechanism that is independent of cyclooxygenase inhibition, cell cycle arrest, and p53 induction. Cancer Res. 1997, 57, 2452–2459. [Google Scholar]
  43. Williams, C.S.; Watson, A.J.; Sheng, H.; Helou, R.; Shao, J.; DuBois, R.N. Celecoxib prevents tumor growth in vivo without toxicity to normal gut: Lack of correlation between in vitro and in vivo models. Cancer Res. 2000, 60, 6045–6051. [Google Scholar]
  44. Han, A.; Song, Z.; Tong, C.; Hu, D.; Bi, X.; Augenlicht, L.H.; Yang, W. Sulindac suppresses β-catenin expression in human cancer cells. Eur. J. Pharmacol. 2008, 583, 26–31. [Google Scholar]
  45. Guo, Q.; Wu, M.; Lian, P.; Liao, M.; Xiao, Z.; Wang, X.; Shen, S. Synergistic effect of indomethacin and NGX6 on proliferation and invasion by human colorectal cancer cells through modulation of the Wnt/β-catenin signaling pathway. Mol. Cell. Biochem. 2009, 330, 71–81. [Google Scholar]
  46. Shureiqi, I.; Jiang, W.; Zuo, X.; Wu, Y.; Stimmel, J.B.; Leesnitzer, L.M.; Morris, J.S.; Fan, H.Z.; Fischer, S.M.; Lippman, S.M. The 15-lipoxygenase-1 product 13-S-hydroxyoctadecadienoic acid down-regulates PPAR-δ to induce apoptosis in colorectal cancer cells. Proc. Nat. Acad. Sci. USA 2003, 100, 9968–9973. [Google Scholar]
  47. Hollingshead, H.E.; Borland, M.G.; Billin, A.N.; Willson, T.M.; Gonzalez, F.J.; Peters, J.M. Ligand activation of peroxisome proliferator-activated receptor-β/δ (PPARβ/δ) and inhibition of cyclooxygenase 2 (COX2) attenuate colon carcinogenesis through independent signaling mechanisms. Carcinogenesis 2008, 29, 169–176. [Google Scholar]
  48. Stolfi, C.; Fina, D.; Caruso, R.; Caprioli, F.; Sarra, M.; Fantini, M.C.; Rizzo, A.; Pallone, F.; Monteleone, G. Cyclooxygenase-2-dependent and -independent inhibition of proliferation of colon cancer cells by 5-aminosalicylic acid. Biochem. Pharmacol. 2008, 75, 668–676. [Google Scholar]
  49. Mahmoud, N.N.; Boolbol, S.K.; Dannenberg, A.J.; Mestre, J.R.; Bilinski, R.T.; Martucci, C.; Newmark, H.L.; Chadburn, A.; Bertagnolli, M.M. The sulfide metabolite of sulindac prevents tumors and restores enterocyte apoptosis in a murine model of familial adenomatous polyposis. Carcinogenesis 1998, 19, 87–91. [Google Scholar]
  50. Rao, C.V.; Rivenson, A.; Simi, B.; Zang, E.; Kelloff, G.; Steele, V.; Reddy, B.S. Chemoprevention of colon carcinogenesis by sulindac, a nonsteroidal anti-inflammatory agent. Cancer Res. 1995, 55, 1464–1472. [Google Scholar]
  51. Moorghen, M.; Ince, P.; Finney, K.J.; Sunter, J.P.; Appleton, D.R.; Watson, A.J. A protective effect of sulindac against chemically-induced primary colonic tumours in mice. J. Pathol. 1988, 156, 341–347. [Google Scholar]
  52. Moorghen, M.; Orde, M.; Finney, K.J.; Appleton, D.R.; Watson, A.J. Sulindac enhances cell proliferation in DMH-treated mouse colonic mucosa. Cell Prolif. 1998, 31, 59–70. [Google Scholar]
  53. Narisawa, T.; Satoh, M.; Sano, M.; Takahashi, T. Inhibition of initiation and promotion by N-methylnitrosourea-induced colon carcinogenesis in rats by non-steroid anti-inflammatory agent indomethacin. Carcinogenesis 1983, 4, 1225–1227. [Google Scholar]
  54. Reddy, B.S.; Rao, C.V.; Rivenson, A.; Kelloff, G. Inhibitory effect of aspirin on azoxymethane-induced colon carcinogenesis in F344 rats. Carcinogenesis 1993, 14, 1493–1497. [Google Scholar]
  55. Jacoby, R.F.; Marshall, D.J.; Newton, M.A.; Novakovic, K.; Tutsch, K.; Cole, C.E.; Lubet, R.A.; Kelloff, G.J.; Verma, A.; Moser, A.R.; Dove, W.F. Chemoprevention of spontaneous intestinal adenomas in the Apc Min mouse model by the nonsteroidal anti-inflammatory drug piroxicam. Cancer Res. 1996, 56, 710–714. [Google Scholar]
  56. Jacoby, R.F.; Seibert, K.; Cole, C.E.; Kelloff, G.; Lubet, R.A. The cyclooxygenase-2 inhibitor celecoxib is a potent preventive and therapeutic agent in the min mouse model of adenomatous polyposis. Cancer Res. 2000, 60, 5040–5044. [Google Scholar]
  57. Rao, C.V.; Steele, V.E.; Swamy, M.V.; Patlolla, J.M.; Guruswamy, S.; Kopelovich, L. Inhibition of azoxymethane-induced colorectal cancer by CP-31398, a TP53 modulator, alone or in combination with low doses of celecoxib in male F344 rats. Cancer Res. 2009, 69, 8175–8182. [Google Scholar] [PubMed]
  58. Kohno, H.; Suzuki, R.; Sugie, S.; Tanaka, T. Suppression of colitis-related mouse colon carcinogenesis by a COX-2 inhibitor and PPAR ligands. BMC cancer 2005, 5, 46. [Google Scholar]
  59. Giardiello, F.M.; Hamilton, S.R.; Krush, A.J.; Piantadosi, S.; Hylind, L.M.; Celano, P.; Booker, S.V.; Robinson, C.R.; Offerhaus, G.J. Treatment of colonic and rectal adenomas with sulindac in familial adenomatous polyposis. N. Engl. J. Med. 1993, 328, 1313–1316. [Google Scholar]
  60. Giardiello, F.M.; Yang, V.W.; Hylind, L.M.; Krush, A.J.; Petersen, G.M.; Trimbath, J.D.; Piantadosi, S.; Garrett, E.; Geiman, D.E.; Hubbard, W.; Offerhaus, G.J.; Hamilton, S.R. Primary chemoprevention of familial adenomatous polyposis with sulindac. N. Engl. J. Med. 2002, 346, 1054–1059. [Google Scholar]
  61. Thun, M.J.; Namboodiri, M.M.; Heath, C.W., Jr. Aspirin use and reduced risk of fatal colon cancer. N. Engl. J. Med. 1991, 325, 1593–1596. [Google Scholar]
  62. Giovannucci, E.; Rimm, E.B.; Stampfer, M.J.; Colditz, G.A.; Ascherio, A.; Willett, W.C. Aspirin use and the risk for colorectal cancer and adenoma in male health professionals. Ann. Intern. Med. 1994, 121, 241–246. [Google Scholar]
  63. Lanas, A.; Ferrandez, A. NSAIDs and the colon. Curr. Opin. Gastroenterol. 2009, 25, 44–49. [Google Scholar]
  64. Sano, H.; Kawahito, Y.; Wilder, R.L.; Hashiramoto, A.; Mukai, S.; Asai, K.; Kimura, S.; Kato, H.; Kondo, M.; Hla, T. Expression of cyclooxygenase-1 and -2 in human colorectal cancer. Cancer Res. 1995, 55, 3785–3789. [Google Scholar]
  65. Kargman, S.L.; O'Neill, G.P.; Vickers, P.J.; Evans, J.F.; Mancini, J.A.; Jothy, S. Expression of prostaglandin G/H synthase-1 and -2 protein in human colon cancer. Cancer Res. 1995, 55, 2556–2559. [Google Scholar]
  66. Piazza, G.A.; Keeton, A.B.; Tinsley, H.N.; Gary, B.D.; Whitt, J.D.; Mathew, B.; Thaiparambil, J.; Coward, L.; Gorman, G.; Li, Y.; Sani, B.; Hobrath, J.V.; Maxuitenko, Y.Y.; Reynolds, R.C. A novel sulindac derivative that does not inhibit cyclooxygenases but potently inhibits colon tumor cell growth and induces apoptosis with antitumor activity. Cancer Prev. Res. 2009, 2, 572–580. [Google Scholar]
  67. McGarvey, M.A.; O'Kelly, F.; Ettarh, R.R. Nimesulide inhibits crypt epithelial cell proliferation at 6 hours in the small intestine in CD-1 mice. Dig. Dis. Sci. 2007, 52, 2087–2094. [Google Scholar]
  68. McGarvey, M.A.; Bass, G.; Ettarh, R.R. Nimesulide alters cell recruitment into mitosis in murine intestinal crypts without influencing the cell production rate. Dig. Dis. Sci. 2007, 52, 1471–1478. [Google Scholar]
  69. Baron, J.A. Epidemiology of non-steroidal anti-inflammatory drugs and cancer. Prog. Exp. Tumor Res. 2003, 37, 1–24. [Google Scholar]
  70. Mahmud, S.M.; Tanguay, S.; Begin, L.R.; Franco, E.L.; Aprikian, A.G. Non-steroidal anti-inflammatory drug use and prostate cancer in a high-risk population. Eur. J. Cancer Prev. 2006, 15, 158–164. [Google Scholar]
  71. Murad, A.S.; Down, L.; Smith, G.D.; Donovan, J.L.; Lane, J.A.; Hamdy, F.C.; Neal, D.E.; Martin, R.M. Associations of aspirin, non-steroidal anti-inflammatory drug and paracetamol use with PSA-detected prostate cancer: Findings from a large, population-based, case-control study (the ProtecT study). Int. J. Cancer 2010. [Epub ahead of print]. [Google Scholar]
  72. Jacobs, E.J.; Rodriguez, C.; Mondul, A.M.; Connell, C.J.; Henley, S.J.; Calle, E.E.; Thun, M.J. A large cohort study of aspirin and other nonsteroidal anti-inflammatory drugs and prostate cancer incidence. J. Natl. Cancer Inst. 2005, 97, 975–980. [Google Scholar]
  73. Platz, E.A.; Rohrmann, S.; Pearson, J.D.; Corrada, M.M.; Watson, D.J.; De Marzo, A.M.; Landis, P.K.; Metter, E.J.; Carter, H.B. Nonsteroidal anti-inflammatory drugs and risk of prostate cancer in the Baltimore Longitudinal Study of Aging. Cancer Epidemiol. Biomarkers Prev. 2005, 14, 390–396. [Google Scholar]
  74. Brasky, T.M.; Bonner, M.R.; Moysich, K.B.; Ambrosone, C.B.; Nie, J.; Tao, M.H.; Edge, S.B.; Kallakury, B.V.; Marian, C.; Trevisan, M.; Shields, P.G.; Freudenheim, J.L. Non-steroidal anti-inflammatory drug (NSAID) use and breast cancer risk in the Western New York Exposures and Breast Cancer (WEB) Study. Cancer Causes Control 2010. [Epub ahead of print]. [Google Scholar]
  75. Cronin-Fenton, D.P.; Pedersen, L.; Lash, T.L.; Friis, S.; Baron, J.A.; Sorensen, H.T. Prescriptions for selective cyclooxygenase-2 inhibitors, non-selective non-steroidal anti-inflammatory drugs, and risk of breast cancer in a population-based case-control study. Breast Cancer Res. 2010, 12, R15. [Google Scholar] [CrossRef] [PubMed]
  76. Cuzick, J.; Otto, F.; Baron, J.A.; Brown, P.H.; Burn, J.; Greenwald, P.; Jankowski, J.; La Vecchia, C.; Meyskens, F.; Senn, H.J.; Thun, M. Aspirin and non-steroidal anti-inflammatory drugs for cancer prevention: An international consensus statement. Lancet Oncol. 2009, 10, 501–507. [Google Scholar]
  77. Rayburn, E.R.; Ezell, S.J.; Zhang, R. Anti-inflammatory agents for cancer therapy. Mol. Cell. Pharmacol. 2009, 1, 29–43. [Google Scholar]

Share and Cite

MDPI and ACS Style

Ettarh, R.; Cullen, A.; Calamai, A. NSAIDs and Cell Proliferation in Colorectal Cancer. Pharmaceuticals 2010, 3, 2007-2021. https://0-doi-org.brum.beds.ac.uk/10.3390/ph3072007

AMA Style

Ettarh R, Cullen A, Calamai A. NSAIDs and Cell Proliferation in Colorectal Cancer. Pharmaceuticals. 2010; 3(7):2007-2021. https://0-doi-org.brum.beds.ac.uk/10.3390/ph3072007

Chicago/Turabian Style

Ettarh, Raj, Anthony Cullen, and Alvise Calamai. 2010. "NSAIDs and Cell Proliferation in Colorectal Cancer" Pharmaceuticals 3, no. 7: 2007-2021. https://0-doi-org.brum.beds.ac.uk/10.3390/ph3072007

Article Metrics

Back to TopTop