Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2009 Feb 1.
Published in final edited form as: Pharmacogenomics J. 2008 Jan 15;8(4):237–247. doi: 10.1038/sj.tpj.6500487

A review of gene-drug interactions for non-steroidal anti-inflammatory drug (NSAID) use in preventing colorectal neoplasia

James T Cross 1,2, Elizabeth M Poole 1,3, Cornelia M Ulrich 1,3
PMCID: PMC2603576  NIHMSID: NIHMS71855  PMID: 18195728

Abstract

Non-steroidal anti-inflammatory drugs (NSAIDs) have been shown to be effective chemopreventive agents for colorectal neoplasia. Polymorphisms in NSAID targets or metabolizing enzymes may affect NSAID efficacy or toxicity.

We conducted a literature review to summarize current evidence of gene-drug interactions between NSAID use and polymorphisms in COX1, COX2, ODC, UGT1A6, and CYP2C9 on risk of colorectal neoplasia by searching the OVID and PubMed.

Of 134 relevant search results, thirteen investigated an interaction. One study reported a significant interaction between NSAID use and the COX1 Pro17Leu polymorphism (p = 0.03) whereby the risk reduction associated with NSAID use among homozygous wild-type genotypes was not observed among NSAID users with variant alleles. Recent pharmacodynamic data support the potential for gene-drug interactions for COX1 Pro17Leu. Statistically significant interactions have also been reported for ODC (315G>A), UGT1A6 (Thr181Ala + Arg184Ser or Arg184Ser alone), and CYP2C9 (*2/*3). No statistically significant interactions have been reported for polymorphisms in COX2; however an interaction with COX2 -765G>C approached significance (p = 0.07) in one study. Among seven remaining studies, reported interactions were not statistically significant for COX1, COX2, and ODC gene polymorphisms. Most studies were of limited sample size. Definitions of NSAID use differed substantially between studies.

The literature on NSAID-gene interactions to date is limited. Reliable detection of gene-NSAID interactions will require greater sample sizes, consistent definitions of NSAID use, and evaluation of clinical trial subjects of chemoprevention studies.

Keywords: non-steroidal anti-inflammatory drugs, cyclooxygenase, prostaglandin H synthase, colorectal cancer, colorectal adenoma, pharmacogenetics

INTRODUCTION

The National Cancer Institute estimates that over 150,000 new cases of colorectal cancer and 52,000 deaths will be reported in the United States in 2007, making it second to lung cancer in total deaths.1 Americans possess a one in eighteen lifetime risk of developing colorectal cancer.2 Five-year relative survival rates range from 9% for distally diagnosed CRC to 90% for localized CRC. This disparity drives public health efforts to increase early detection and to slow or prevent altogether the progression of colorectal carcinogenesis.

Inflammation is a known risk factor for colorectal cancer. Several inflammatory conditions predispose to colorectal cancer, such as ulcerative colitis3 and Crohn’s disease.4 Non-steroidal anti-inflammatory drugs (NSAIDs), including aspirin, represent a potential means of decreasing inflammation in the colonic epithelium.5 There are two main subtypes of NSAIDs: nonselective and selective COX2 inhibitors. The COX2-selective drugs (coxibs) exhibit higher affinity for and therefore target the COX2 enzyme.5 NSAIDs have been successful in preventing colorectal neoplasia in high-risk populations, such as subjects with a prior diagnosis of CRC or colorectal adenoma. Recently, two randomized placebo-controlled trials (RCTs) showed aspirin to significantly reduce the risk of recurrent adenomatous polyps by 19% to 35%.6, 7 Two other RCTs showed a 33%-36% risk reduction for celecoxib and even greater reduction in the risk of advanced adenoma.8, 9 The magnitude of risk reduction from rofecoxib versus placebo was recently shown to be comparable to that of aspirin.10 However, rofecoxib is no longer commercially available due to concerns of cardiovascular toxicity.11-13 Gastrointestinal and cardiovascular toxicity from aspirin/NSAID and coxibs, respectively, have spurred research to identify genetic variations which might alter the risk-benefit trade-off of these drugs in clinically meaningful ways and allow tailoring of chemoprevention.5

NSAIDs inhibit the cyclooxygenase (COX) activity of COX enzymes (i.e., prostaglandin H synthases), which in turn decreases prostaglandin production.14 COX1 is constitutively expressed in many tissues and is linked to homeostatic functions, whereas COX2 is an inducible form involved in inflammatory and proliferative responses.15-17 Genetic variability in downstream enzymes in the prostaglandin or lipoxygenase pathway (which competes with the COX enzymes to metabolize arachidonic acid), may also play a role in colorectal cancer, because these may affect the overall availability and balance of inflammatory mediators in the body.

Recent interest has emerged in the role of ornithine decarboxylase (ODC) and NSAIDs in colorectal cancer. NSAIDs, including celecoxib, inhibit this enzyme.18 Whereas COX1 and COX2 mediate prostaglandin synthesis, ODC catalyzes the synthesis of polyamines, which are associated with carcinogenesis (increased cell division, up-regulation of genes involved in metastasis and tumor invasion, and down-regulation of apoptosis).19, 20 Increased intracellular polyamine concentrations are positively associated with risk of cancer19, 21, including sporadic colorectal cancer22, and are negatively associated with apoptotic activity and cell death.23 ODC is overexpressed in cancerous versus normal colon epithelium.19, 24-27 Thus, the chemopreventive properties of NSAIDs in colorectal cancer may stem in part from their activity on ODC-mediated polyamine synthesis.28-30

NSAIDs are primarily metabolized by two major classes of enzymes: the cytochrome P450 2C enzymes (CYP2C) and the UDP-glucuronosyltransferases (UGTs). The major metabolizers of NSAIDs are CYP2C931 and UGT1A632, although other UGTs and CYPs may play minor roles. Both of these enzymes have common polymorphisms that are associated with less efficient drug metabolism. In CYP2C9, two polymorphisms, Arg144Cys (also referred to as *2) and Ile359Leu (also referred to as *3) show markedly decreased warfarin metabolism compared to wild-type.33, 34 Similarly, there are two known variant alleles in UGT1A6 that have been associated with decreased enzyme activity; the first is characterized by amino-acid changes at amino acids 181 and 184 (Thr181Ala + Arg184Ser) and the second by Arg184Ser alone.35, 36 These known functional genetic polymorphisms may interact with NSAID use to affect risk of colorectal neoplasia.

Polymorphisms in COX1, COX2, and ODC appear to alter the risk of colorectal neoplasia.5, 37-43 Since genetic polymorphisms and NSAID use can each modify the risk for colorectal neoplasia, pharmacogenetic studies may help to identify the population for whom NSAIDs have the most favorable risk-benefit profile for colorectal adenoma prevention. Here, we review the potential interactions between NSAIDs and genetic polymorphisms in defining risk for colorectal neoplasia and discuss future considerations for research.

RESULTS

We identified 135,360 articles about aspirin or other NSAIDs, 475,640 about polymorphisms or mutations, 223,165 that concerned the colon or rectum, and 1,610,007 that concerned neoplasia (cancer, polyp, adenom-, or neoplas-). One hundred thirty-four publications contained keywords from all four search sets. Of these, thirteen studies reported on NSAID-drug interactions where colorectal neoplasia was the clinical outcome (Table 1).

Table 1. Characteristics of studies evaluating gene-NSAID interactions*.

Study Genes Study
design
# Cases/controls NSAID
use
Study
region
Colorectal study
endpoint
Population
description
Covariates
Lin 200241 COX2 Case-control 161/219 Not defined California/North Carolina Adenomatous polyp; Cancer African-American; 30-74 years old; spoke English Matched: age, sex, sigmoidoscopy date, center
Bigler 200157 CYP2C9
UGT1A6
Case-controls 441/488 ASA/NSAID users: >once daily for >1 year Minnesota Adenomatous polyp 30-74 years old Adjusted: age, sex, smoking, hormone replacement therapy
Martinez 200330 ODC Case-control 341/347 ASA use collected, not defined Arizona Adenomatous polyp 40-80 years old Adjusted: age, sex, # colonoscopies after baseline
Cox 200442 COX2 Case-control 292/274 Regular use for at least 6 consecutive months Spain Cancer Presented only as ORs Matched: age, sex
Ulrich 200437 COX1 Case-control 715/621 Regular ASA/NSAID use: > once daily Minnesota Adenomatous, hyperplastic polyp Caucasian; English speaking; 30-74 years old Adjusted: age, sex
Ali 200573 COX2 Case-control 726/729 Use of Aspirin, Ibuprofen, none or both (not clearly defined) USA Advanced adenomatous polyp Caucasian; 55-74 years old Matched: age, sex Adjusted: age, sex, smoking, NSAID
Chan 200558 UGT1A6 case-control 530/532 ASA users: twice weekly USA Adenomatous polyp Not reported Adjusted: age, smoking hx, BMI, physical activity, family hx of CRC, meat intake, alcohol intake, multivitamin use, folate intake, calcium intake
Ulrich 200538 COX2 Case-control 690/584 Regular ASA/NSAID users: > once daily Minnesota Adenomatous, hyperplastic polyp Caucasian; English-speaking; 30-70 years old. Adjusted: age, sex, BMI, calories, alcohol, fiber, hormone use, & smoking

In ten of these thirteen studies, investigators reported on gene-NSAID interactions and the risk for development of colorectal adenomatous polyps; the other three reported on interactions and the risk for colorectal cancer.42, 44, 45 All studies included age and sex as matching or adjustment variables. Other covariates included in some of the studies were smoking status, time since colonoscopy, fiber intake, alcohol consumption, BMI, and family history of colorectal cancer. The studies were conducted in primarily Caucasian populations, however two studies were conducted in African-American subjects.41, 44 NSAID use was not consistently defined in these studies and varied by dose, duration, or frequency. Such inconsistency has the potential to lead to exposure misclassification across studies, and consequently the comparability of results. The studies ranged in size from 161 cases and 219 controls41 to 2295 cases and 2903 controls.45

COX1

Four COX1 polymorphisms (Arg8Trp, Leu15-Leu16del, Pro17Leu, and Leu237Met) have been evaluated in the literature for an interaction with NSAID exposure on the risk for colorectal neoplasia (Table 2). Although much attention has focused on the role of COX2 in NSAID pharmacodynamics, recent findings suggest a role for COX1 in colorectal carcinogenesis46-48 and in the safety of NSAIDs, particularly coxibs.49 Pro17Leu is a single nucleotide polymorphism that results in an amino acid change in exon 2 of the COX1 gene.50 In one study, NSAID use was associated with an adenoma risk reduction only among Pro17Leu wild-type NSAID users compared to wild-type nonusers (OR: 0.6; 95% CI, 0.5-0.8; p = 0.03).50 The Pro17Leu polymorphism is located in the signal peptide of COX1 and is cleaved to form the mature protein; therefore it is unclear what functional effects this polymorphism would have. However, this polymorphism has been reported to be in complete linkage disequilibrium with a promoter polymorphism, -842A>G, which may affect binding of transcription factors.51 The Pro17Leu variant has been associated with altered prostaglandin production52 and coxib selectivity.49 No statistically significant interactions have been reported for the Arg8Trp, Leu15-16del, or Leu237Met polymorphisms. Few studies have investigated potential functional effects of these COX1 polymorphisms.49, 51, 53with little evidence for changes in enzyme function. However, a haplotype containing -842A>G, Arg8Trp, and Leu237Met was associated with differential aspirin response in one study.53 The impact of COX1 polymorphisms on peroxidase activity or peroxide regulation has not yet been studied. This is an important aspect of COX1 regulation, because peroxides are required for the initation of COX1 activity.54 However, as is often the case with rare variant alleles, these polymorphisms may require larger studies to detect interactions.

Table 2. Interactions of NSAID use reported in the literature for COX1, COX2, or ODC and risk of colorectal neoplasia.

Mutation Primary Outcome Main effect OR [95%CI] OR comparison Interactions Interaction Comparison 1st Author Yr
COX1
Leu15-Leu16del Adenomatous polyps 3.6 [1.2-11.2] het vs. wt p = 0.12 Leu15-16del potentially associated with stronger risk of adenoma among nonusers of ASA/NSAID. Ulrich 200437
Hyperplastic polyps 2.1 [0.5-9.1] het vs. wt Ulrich 200437
Leu237Met Adenomatous polyps 0.8 [0.5-1.4] het vs. wt p = 0.22 No significant interaction, but ASA/NSAID use ↓ risk for Leu237 wt (OR: 0.6; 95% CI 0.5-0.9) but not for Leu237 het/hzv. Ulrich 200437
Adenomatous polyps 0.86 [0.28-2.67] het/hzv vs. wt No significant interaction. Siezen 200653
Hyperplastic polyps 1.0 [0.5-2.0] het vs. wt Ulrich 200437
Pro17Leu Adenomatous polyps 0.9 [0.6-1.2] het/hzv vs. wt p = 0.03 ASA/NSAID use ↓ risk for P17L wt (OR: 0.6; 95% CI 0.5-0.8) but not P17L het/hzv. Ulrich 200437
Hyperplastic polyps 0.7 [0.4-1.1] het/hzv vs. wt Ulrich 200437
Arg8Trp Adenomatous polyps 1.1 [0.8-1.6] het/hzv vs. wt p = 0.31 No significant Arg8Trp*NSAID interaction. Ulrich 200437
Hyperplastic polyps 1.2 [0.8-1.9] het/hzv vs. wt Ulrich 200437
Trp8Arg Adenomatous polyps 0.90 [0.49-1.65] het/hzv vs. wt No significant interaction. Siezen 200653
COX2
-1329A>G Adenomatous polyps 1.20 [0.78-1.84]
0.95 [0.34-2.65]
het vs. wt
hzv vs. wt
No significant interaction. Siezen 200653
T5229G Advanced adenomatous polyps 0.94 [0.74-1.20]
0.73 [0.42-1.27]
het vs. wt
hzv vs. wt
Ali 200573
-663 GTdel Advanced adenomatous polyps 0.65 [0.40-1.04] het del vs. wt Interaction studied in haplotype analysis only. Ali 200573
-765G>C Adenomatous polyps 1.12 [0.70-1.78] het/hzv vs. wt No significant interaction. Siezen 200653
Adenomatous polyps (n=494) 1.00 [0.74,1.35] het vs. wt 0.66 [0.48-0.92]
1.02 [0.69-1.51]
0.64 [0.40-1.02]
0.26 [0.07-0.89]
0.82 [0.25-2.73]
p = 0.07
Wt users vs. wt nonusers
Het nonusers vs wt nonusers
Het users vs wt nonusers
Hzv nonusers vs wt nonusers
Hzv users vs wt nonusers
Hzv genotype
Marginally nonsignificant interaction for NSAID use & genotype (P = 0.07, het/hzv vs wt).
↓ risk for hzv non
Ulrich 200538
Adenomatous polyps (n=494) 0.53 [0.22-1.28] hzv vs. wt Ulrich 200538
Hyperplastic polyps(n=186) 0.97 [0.65-1.46] het vs. wt Ulrich 200538
Hyperplastic polyps(n=186) 0.24 [0.05-1.11] hzv vs. wt Ulrich 200538
-798A>G Advanced adenomatous polyps 1.02 [0.81-1.27]
0.80 [0.43-1.50]
het vs. wt
hzv vs. wt
Ali 200573
T8494C Advanced adenomatous polyps 1.17 [0.94-1.46]
1.14 [0.82-1.59]
het vs. wt
hzv vs. wt
Ali 200573
G10335A Cancer 2.17 [0.99-4.78] het/hzv vs. wt Cox 200442
C1629G Cancer 1.59 [0.56-4.52] het vs. wt
hzv vs. wt
Cox 200442
T2242C Adenomatous polyps 1.30 [0.86-1.98]
1.15 [0.55-2.41]
het vs. wt
hzv vs. wt
Non significant interaction. Siezen 200653
G3050C Cancer 1.30 [0.90-1.87]
1.50 [0.63-3.57]
het vs. wt
hzv vs. wt
Cox 200442
A401G Cancer 0.92 [0.62-1.38]
0.78 [0.33-2.05]
het vs. wt
hzv vs. wt
Cox 200442
5209T>G Cancer 1.05 [0.73-1.52]
0.99 [0.42-2.33]
het vs. wt
hzv vs. wt
Cox 200442
T8473C Cancer 1.01 [0.71-1.45]
1.05 [0.58-1.91]
het vs. wt
hzv vs. wt
Cox 200442
G926C Cancer 0.92 [0.61-1.39]
1.13 [0.46-2.80]
het vs. wt
hzv vs. wt
Cox 200442
A9850G Cancer 2.49 [1.17-5.32] het/hzv vs. wt NSAID*9850A>G nonsignificant (p-value = 0.19).
↓ risk for wt homozygous (AA=0.55 [0.36-0.84]).
Het/hzv users ↑ risk vs. nonusers (AG/GG=1.08, 95% CI 0.17-6.77).
NSAID*9850A>G wt homozygous het/hzv Cox 200442
Val102Val (G>C) Adenomatous polyps 0.65 [0.42-1.01] het/hzv vs. wt No significant interaction. Siezen 200653
Val511Ala Cancer 0.67 [0.28-1.56] het/hzv vs. wt Lin 200241
Cancer 1.19 [0.39-3.61] het/hzv vs. wt excluding NSAID users Lin 200241
Cancer 0.62 [0.33-1.16] het/hzv vs. wt p = 0.59 Interaction no statistically significant.
Significant ↓ risk among wt users (0.66, 0.45-0.95) was nonsignificant among het/hzv users.
Sansbury 200645
Adenomatous polyps 0.56 [0.25-1.27] het/hzv vs. wt Lin 200241
Adenomatous polyps 0.29 [0.08-1.08] het/hzv vs. wt excluding NSAID users Lin 200241
ODC
G315A Adenomatous polyps 1.03 [0.89-1.20]
0.98 [0.73-1.32]
1.02 [0.88-1.17]
het vs. wt hzv vs. wt
het/hzv vs. wt
p = 0.04 Significant interaction: ASA use ↓ adenoma risk for ODC 315G>A het/hzv but not for wt. Barry 200643
Advanced lesions 0.90 [0.61-1.34]
0.70 [0.29-1.69]
0.89 [0.61-1.30]
het vs. wt
hzv vs. wt
het/hzv vs. wt
p = 0.02 Significant interaction: ASA use ↓ advanced lesion risk for ODC 315G>A het/hzv but not for wt. Barry 200643
Adenomatous polyps 0.96 [0.68-1.34]
0.48 [0.24-0.99]
het vs wt
hzv vs wt
p = 0.13 No interaction between ODC*ASA use and adenoma risk. Martinez 200330
Adenomatous polyps 1.05 [0.70-1.58]
0.68 [0.30-1.51]
het nonuser vs wt nonuser
hzv nonuser vs wt nonuser
Martinez 200330
Adenomatous polyps 0.83 [0.51-1.34]
0.64 [0.37-1.09]
0.10 [0.02-0.66]
wt user vs wt nonuser
het user vs wt nonuser
hzv user vs wt nonuser
Martinez 200330
UGT1A6
Thr181Ala + Arg184Ser Adenomatous polyps 0.97 [0.74-1.26] het/hzv vs. wt Risk reduction with ASA use stronger among those with any variant allele. Bigler 200157
Adenomatous polyps 0.74 [0.39-1.41]
0.41 [0.24-0.71]
wt user (>7 pills/week) vs.wt nonuser variant allele user (>7 pills/week) vs. variant allele nonuser p = 0.02 Risk reduction with ASA use stronger among those with variant allele. Chan 200558
Adenomatous polyp recurrence 0.68 [0.52-0.89] het/hzv vs. wt p = 0.70 No interaction between ASA use and UGT1A6 variant alleles for polyp recurrence Hubner 200659
Cancer 1.08 [0.94-1.24]
0.94 [0.76-1.15]
het/hzv vs. wt (colon cancer)
het/hzv vs. wt (rectal cancer)
p = 0.39 (ibuprofen)
p = 0.40 (ASA)
No interaction between UGT1A6*ASA/ibuprofen use and adenoma risk. Samowitz 200646
CYP2C9
Adenomatous polyps 1.10 [0.83-1.46] het
hzv vs. wt
Risk reduction with ASA use stronger among those wt. Bigler 200157
Adenomatous polyp recurrence 1.09 [0.82-1.44] het
hzv vs. wt
p = 0.98 No interaction between ASA use and CYP2C9 variant alleles for polyp recurrence Hubner 200659
Cancer 1.04 [0.90-1.21]
0.93 [0.76-1.14]
het
hzv vs. wt (colon cancer)
het
hzv vs. wt (rectal cancer)
p = 0.41 (ibuprofen)
p = 0.02 (ASA)
Risk reduction with ASA use stronger among those with variant allele; no interaction with ibuprofen use. Samowitz 200646

COX2

Seventeen COX2 polymorphisms have been tested for interaction with NSAID exposure (see Table 2 for the full list). The most commonly evaluated polymorphisms were those occurring at -765G>C and Val511Ala, the latter of which only occurs in non-Caucasian populations. One study (494 cases and 584 controls) has reported large risk reductions in colorectal adenoma for - 765 homozygous variant (CC) nonusers compared to wild-type (GG) nonusers (OR: 0.26, 95%CI: 0.07-0.89). When stratified on NSAID use, homozygous variant non-users were at decreased risk of adenoma (OR: 0.26, 95% CI: 0.07-0.89) compared to wild-type non-users, whereas there was no decrease in risk among homozygous variant NSAID users (OR: 0.82, 95% CI: 0.25-2.73). This interaction approached statistical significance (p = 0.07).38 However, a smaller study of 337 adenoma cases and 368 controls found no evidence of interaction between this polymorphism and NSAID use.55 The -765G>C polymorphism is relatively frequent (minor allele frequency ∼17%)38 and has been shown to suppress COX2 promoter activity56, although not consistently so.57 In atherosclerosis, patients with the -765CC genotype possess significantly lower levels of C-reactive protein and interleukin-6, biomarkers of inflammatory disease.

Two studies have reported on the COX-2 Val511Ala polymorphism (which is not found in Caucasians) and NSAID use in African-Americans. One examined this interaction in regards to colorectal cancer (240 cases and 326 controls)44, while the other studied the interaction for distal adenoma (240 of 380 subjects were evaluated by sigmoidoscopy alone).41 The former study noted that the significantly decreased risk of cancer among wild-type NSAID users (OR: 0.66; 95% CI, 0.45-0.95) was even greater among those carrying at least one variant allele (OR: 0.29, 95% CI, 0.08-1.06), indicating that those with a variant allele may benefit more from NSAID use. However, the interaction was statistically non-significant (p = 0.59). The latter study (161 cases, 219 controls) found significant reductions in risk among those who were either NSAID users or carried the A allele (or both) compared to those with neither exposure41, but did not evaluate multiplicative interaction. Earlier functional analyses that showed that the Val511Ala variant did not modify the inhibitory effects of several NSAIDs, including celecoxib and indomethacin, thus an NSAID interaction with this polymorphism may be less likely.58

In a hospital-based case-control study conducted in Spain, subjects carrying at least one variant allele of the 9850A>G polymorphism in the COX2 gene showed a significantly increased risk of colorectal cancer (OR: 2.49; 95% CI, 1.17-5.32).42 The interaction with NSAID use, however, was not statistically significant (p = 0.19). This was the only study identified in the literature that reported on this polymorphism with respect to colorectal neoplasia. However, this study was fairly small (N = 292/274 controls/cases), so statistical power with respect to interactions was limited. Additionally, the use of hospital controls may bias results, because underlying comorbidities that may be associated with NSAID use can attenuate the true association between exposure (NSAID use) and outcome (cancer). This polymorphism has not been associated with functional effects, so it may be unlikely that a true association between this polymorphism and colorectal neoplasia risk exists.

ODC

Two studies have tested for interaction between the ODC 315G>A polymorphism and NSAID exposure and the risk for colorectal neoplasia (Table 2). This polymorphism is in a regulatory region of the gene near transcription factor binding sites and has been associated with differential RNA expression.59 Martinez and colleagues reported that the homozygous variant (AA) genotype was associated with a significant reduction in the risk of adenoma (OR: 0.48; 95% CI, 0.24-0.99).30 Although the NSAID interaction was not statistically significant, the risk among homozygous variant (AA) NSAID users was greatly reduced compared to wild-type (GG) nonusers (OR: 0.10; 95% CI, 0.02-0.66); whereas a risk reduction was not observed among homozygous variant nonusers versus wild-type nonusers (OR: 0.68; 95% CI, 0.30-1.51; p-interaction = 0.13). Barry et al. examined specimens from an RCT and did not observe a main association with this ODC polymorphism on risk of adenoma, but did report a statistically significant interaction of 315G>A genotype and aspirin use on adenoma risk.43 Aspirin users with at least one variant allele had a significant reduction in adenoma risk (RR: 0.77; 95%CI, 0.63-0.95; p-interaction = 0.04) and advanced adenoma risk (RR: 0.51; 95%CI, 0.29-0.90; p-interaction = 0.02) compared to those on placebo with at least on variant allele. No risk reduction associated with aspirin use was observed among those with the wild-type genotype. This suggests that the combination of NSAID use and ODC variants cumulatively reduces risk.

UGT1A6

Four studies have investigated potential interactions between the known functional polymorphisms in UGT1A6 (Thr181Ala + Arg184Ser or Arg184Ser alone) and colorectal neoplasia risk. In a study of 441 adenoma cases and 451 controls, the risk reduction for regular aspirin users was seen only among those with at least one variant allele (OR: 0.53, 95% CI, 0.33-0.86, p-interaction not reported).60 Similarly, in a case-control study of 313 women with adenoma and 303 control women, the risk reduction associated with regular NSAID use was stronger among women with any variant UGT1A6 genotype compared to those with the wild-type alleles (p-interaction = 0.02).61 Two other studies reported no interaction between UGT1A6 genotype and NSAID use.45, 62

CYP2C9

Three of the studies listed above also investigated interactions between the *2 and *3 polymorphisms in CYP2C9 and NSAID use on risk of colorectal neoplasia.45, 60, 62 In the study by Bigler et al, the colorectal adenoma risk reduction associated with aspirin use was only seen among those with the wild-type CYP2C9 genotype (OR: 0.50, 95% CI 0.32-0.78, p-interaction not reported). No risk reduction was seen among non-aspirin NSAID users.60 A subsequent study found a significant interaction between the *2 and *3 genotypes and ibuprofen use, in which those with the variant alleles had a greater decrease in risk with regular ibuprofen use than those with the wild-type alleles (p-interaction = 0.02).45 Hubner et al reported no interaction between CYP2C9 genotypes and aspirin treatment in an RCT of aspirin for prevention of adenoma recurrence; however, the study was small, with 266 patients on aspirin and 280 on placebo.62 The discrepancy among these three studies indicates that the interaction between CYP2C9 polymorphisms and NSAID use requires confirmation in additional studies.

OTHER REPORTED INTERACTIONS

Although not within the scope of this review, two studies have investigated interactions between NSAID use and polymorphisms in other prostaglandin-related genes, such as PPARγ, PPARδ, ALOX5, ALOX15, and PGIS.55, 63 Most of these have only been examined in one or two studies and results require confirmation.

DISCUSSION

We reviewed the literature for COX1, COX2, ODC, UGT1A6, and CYP2C9 pharmacogenetic interactions and the risk of colorectal adenoma or cancer. To date, research has overwhelmingly focused on COX2 polymorphisms. However, all COX2 and NSAID pharmacogenetic interactions we identified were not statistically significant, probably in large part attributable to limited sample sizes for detecting true interactions. On the other hand, statistically significant interactions were reported for the COX1 signal peptide polymorphism Pro17Leu, ODC 315G>A, UGT1A6 Thr181Ala/Arg184Ser, and CYP2C9 *2 and *3. The interactions for ODC, UGT1A6, and CYP2C9 have been observed in two studies, suggesting important pharmacogenetic relationships. To date, only interactions between COX2 polymorphisms and COX2-nonselective NSAIDs have been evaluated. All polymorphisms reviewed here have yet to be tested for interactions with coxibs (e.g., celecoxib and lumiracoxib). This information will be critical to tailor cancer chemoprevention with these highly potent agents. It will also be important to evaluate these polymorphisms in conjunction with each other.

Recent randomized trials confirm the chemopreventive properties of NSAIDs in colorectal neoplasia. In one randomized trial, aspirin led to a statistically significant reduction of adenoma at three years at the 81 mg daily dose (but not at the 325 mg daily dose).6 In another, the 325 mg dose resulted in a significant risk reduction at the 325 mg once-daily dose7, yet the heterogeneity in response for the 325 mg dose is not well understood. Before adopting aspirin as a chemopreventive agent, understanding the sources of such variability in efficacy is warranted. The same applies to the gastrointestinal toxicity of nonselective NSAIDs, which have been estimated to cause 25% of all reported drug-related adverse events.64 The Hypertension Optimal Treatment randomized trial demonstrated an increased risk for non-fatal major gastrointestinal bleeding among aspirin users versus placebo (RR, 1.8, p < 0.001)65, specifically among female aspirin users. Although the Physicians’ Health Study did not show an increased risk of gastrointestinal bleeding for aspirin users, the trial had an aspirin tolerability run-in period that would have eliminated many persons susceptible to such toxicity. Identifying genetic predictors of gastrointestinal toxicity may ultimately help to define the optimal risk-benefit for specific subpopulations.66

Interestingly, results from a recent cyclooxygenase inhibition study suggest that the COX1 Pro17Leu polymorphism may play a role in the cardiotoxicity of coxibs.49 Inhibition of COX1 by coxibs decreased in a statistically significant manner among Pro17Leu variants.49 Decreased COX1 inhibition corresponds with increased levels of thromboxane A2 (TXA2), which is involved in platelet function. This increase would further offset an existing imbalance between COX1-derived TXA2 and COX2-derived prostacyclin resulting from the selective inhibition of COX2 by coxibs.67, 68 If this imbalance indeed contributes to the cardiovascular risk associated with use coxibs, as has been suggested69, our efforts to describe future tests for pharmacogenetic interactions should consider cardiovascular risk as an outcome of interest, in addition to that of colorectal neoplasia.

Some have questioned altogether whether coxibs should continue to be evaluated for their potential as colorectal chemopreventive agents70, due to the known cardiovascular risks associated with this class of drugs71-74 that are not observed with other NSAIDs such as aspirin.66 The value of coxibs for chemoprevention will therefore lie in our ability to define the population of individuals at most likely to benefit and to be less likely to experience drug-related serious adverse events. When deciding on how to minimize their risk of colorectal cancer, certain populations at increased risk of cancer, such as those with familial adenomatous polyposis, may place greater value on the cancer-preventive properties of coxibs than on their potential for cardiovascular adverse events.75 We ought to consider the efficacy and adverse effects of all available NSAIDs to tailor chemoprevention based on genetic and other factors in favor of the greatest benefit:risk ratio.

In summary, inflammation is an established risk factor for colorectal cancer and polymorphisms in genes regulating inflammatory processes appear to alter the risk for neoplasia and the efficacy of NSAIDs in colorectal cancer chemoprevention. Studies investigating potential interactions between NSAID use and genetic polymorphisms in inflammation have been of limited power due to inadequate sample size; studies with fewer than 400 cases and 400 controls are most likely underpowered for detecting most gene-NSAID interactions. Our understanding of pharmacogenetic interactions between anti-inflammatory drug use and genetic polymorphisms and these health outcomes may pave the road to chemoprevention of colorectal cancer by allowing us to optimize the risk-benefit balance associated with NSAIDs.

METHODS

Search terms were used to identify publications that assessed interactions between NSAID use and polymorphisms in NSAID-related genes (i.e. CYP2C9, UGT1A6, and prostaglandin synthase) on the risk of colorectal neoplasia from the Ovid MEDLINE® database.76 We queried the titles, abstracts, and keywords of indexed and in-process publications through April 26, 2007. We searched for articles containing main effects of NSAID/aspirin drug exposure and gene polymorphisms and colorectal neoplasia using the following four sets of search terms: (1) NSAID, antiinflammatory, anti-inflammatory, nonsteroidal, non-steroidal, aspirin, or acetylsalicylic, and (2) polymorphism, variant, or mutation, and (3) cancer, adenom-, polyp, or neoplas-, and (4) colon, rectum, rectal, colorectal, or colonic. We considered relevant articles to be any that contained at least one term from each set of keywords and reviewed content for analyses of potential gene-NSAID interactions.

We abstracted information on the study design (including sample size, covariates, endpoint measures, and inclusion criteria), the risk estimates for the gene and drug main associations, and the results from interaction tests.

Acknowledgements

Work was supported by grants from the National Institutes of Health R01 CA114467; R03 CA123577; R25 CA094880, and R25 CA092408-06 We would like to thank Rachel Galbraith for her technical assistance with the manuscript.

Abbreviations used

CI

confidence interval

COX

cyclooxygenase

NSAIDs

non-steroidal anti-inflammatory drugs

ODC

ornithine decarboxylase

OR

odds ratio

PTGS1

prostaglandin H synthase1

PTGS2

prostaglandin H synthase 2

hzv

heterozygous variant

References

  • 1.Estimated new cancer cases and deaths for 2006. National Cancer Institute; Bethesda, MD: 2006. [Google Scholar]
  • 2.Cancer fast facts sheet: Cancer of the colon and rectum. National Cancer Institute; Bethesda, MD: 2006. [Google Scholar]
  • 3.Ekbom A, Helmick C, Zack M, Adami HO. Ulcerative colitis and colorectal cancer. A population-based study. N Engl J Med. 1990;323:1228–33. doi: 10.1056/NEJM199011013231802. [DOI] [PubMed] [Google Scholar]
  • 4.Jess T, Gamborg M, Matzen P, Munkholm P, Sorensen TI. Increased risk of intestinal cancer in crohn’s disease: A meta-analysis of population-based cohort studies. Am J Gastroenterol. 2005;100:2724–9. doi: 10.1111/j.1572-0241.2005.00287.x. [DOI] [PubMed] [Google Scholar]
  • 5.Ulrich CM, Bigler J, Potter JD. Non-steroidal anti-inflammatory drugs for cancer prevention: Promise, perils, and pharmacogenetics. Nat Rev Cancer. 2006;6:130–40. doi: 10.1038/nrc1801. [DOI] [PubMed] [Google Scholar]
  • 6.Baron JA, Cole BF, Sandler RS, Haile RW, Ahnen D, Bresalier R, et al. A randomized trial of aspirin to prevent colorectal adenomas. New Engl J Med. 2003;348:891–9. doi: 10.1056/NEJMoa021735. [DOI] [PubMed] [Google Scholar]
  • 7.Sandler RS, Halabi S, Baron JA, Budinger S, Paskett E, Keresztes R, et al. A randomized trial of aspirin to prevent colorectal adenomas in patients with previous colorectal cancer. New Engl J Med. 2003;348:883–90. doi: 10.1056/NEJMoa021633. [DOI] [PubMed] [Google Scholar]
  • 8.Bertagnolli MM, Eagle CJ, Zauber AG, Redston M, Solomon DH, Kim K, et al. Celecoxib for the prevention of sporadic colorectal adenomas. New Engl J Med. 2006;355:873–874. doi: 10.1056/NEJMoa061355. [DOI] [PubMed] [Google Scholar]
  • 9.Arber N, Eagle CJ, Spicak J, Racz I, Dite P, Hajer J, et al. Celecoxib for the prevention of colorectal adenomatous polyps. New Engl J Med. 2006;355:885–895. doi: 10.1056/NEJMoa061652. [DOI] [PubMed] [Google Scholar]
  • 10.Baron JA, Sandler RS, Bresalier RS, Quan H, Riddell R, Lanas A, et al. A randomized trial of rofecoxib for the chemoprevention of colorectal adenomas. Gastroenterol. 2006;131:1674–82. doi: 10.1053/j.gastro.2006.08.079. [DOI] [PubMed] [Google Scholar]
  • 11.Topol EJ. Failing the public health--rofecoxib, merck, and the fda. N Engl J Med. 2004;351:1707–9. doi: 10.1056/NEJMp048286. [DOI] [PubMed] [Google Scholar]
  • 12.Bresalier RS, Sandler RS, Quan H, Bolognese JA, Oxenius B, Horgan K, et al. Cardiovascular events associated with rofecoxib in a colorectal adenoma chemoprevention trial. N Engl J Med. 2005;352:1092–102. doi: 10.1056/NEJMoa050493. [DOI] [PubMed] [Google Scholar]
  • 13.Solomon DH, Schneeweiss S, Glynn RJ, Kiyota Y, Levin R, Mogun H, et al. Relationship between selective cyclooxygenase-2 inhibitors and acute myocardial infarction in older adults. Circulation. 2004;109:2068–73. doi: 10.1161/01.CIR.0000127578.21885.3E. [DOI] [PubMed] [Google Scholar]
  • 14.Vane JR. Inhibition of prostaglandin synthesis as a mechanism of action for aspirin-like drugs. Nature New Biol. 1971;231:232–5. doi: 10.1038/newbio231232a0. [DOI] [PubMed] [Google Scholar]
  • 15.Taketo MM. Cyclooxygenase-2 inhibitors in tumorigenesis (part i) J Natl Cancer Inst. 1998;90:1529–36. doi: 10.1093/jnci/90.20.1529. [DOI] [PubMed] [Google Scholar]
  • 16.Smith WL, DeWitt DL, Garavito RM. Cyclooxygenases: Structural, cellular, and molecular biology. Annu Rev Biochem. 2000;69:145–82. doi: 10.1146/annurev.biochem.69.1.145. [DOI] [PubMed] [Google Scholar]
  • 17.Gupta RA, Dubois RN. Colorectal cancer prevention and treatment by inhibition of cyclooxygenase-2. Nat Rev Cancer. 2001;1:11–21. doi: 10.1038/35094017. [DOI] [PubMed] [Google Scholar]
  • 18.Ostrowski J, Wocial T, Skurzak H, W B. Do altering in ornithine decarboxylase activity and gene expression contribute to antiproliferative properties of cox inhibitors? Br J Cancer. 2003;88:1143–1151. doi: 10.1038/sj.bjc.6600815. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Gerner EW, Meyskens FL., Jr. Polyamines and cancer: Old molecules, new understanding. Nat Rev Cancer. 2004;4:781–92. doi: 10.1038/nrc1454. [DOI] [PubMed] [Google Scholar]
  • 20.Babbar N, Ignatenko NA, Casero RA, Gerner EW. Cyclooxygenase-independent induction of apoptosis by sulindac sulfone is mediated by polyamines in colon cancer. J Biol Chem. 2003;278:47762–47775. doi: 10.1074/jbc.M307265200. [DOI] [PubMed] [Google Scholar]
  • 21.Janne J, Poso H, A R. Polyamines in rapid growth and cancer. Biochim Biophys Acta. 1978;473:241–93. doi: 10.1016/0304-419x(78)90015-x. [DOI] [PubMed] [Google Scholar]
  • 22.Kingsnorth AN, Lumsden AB, W HM. Polyamines in colorectal cancer. Br J Surg. 1984;71:791–794. doi: 10.1002/bjs.1800711019. [DOI] [PubMed] [Google Scholar]
  • 23.Scornioni Manipulation of the expression of regulatory genes of polyamine metabolism results in specific alterations of the cell-cycle progression. Biochem J. 2001;354:217–223. doi: 10.1042/0264-6021:3540217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Porter CW, Herrera-Ornelas L, Pera P, Petrelli NF, Mittelman A. Polyamine biosynthetic activity in normal and neoplastic human colorectal tissue. Cancer. 1987;60:1275–1281. doi: 10.1002/1097-0142(19870915)60:6<1275::aid-cncr2820600619>3.0.co;2-i. [DOI] [PubMed] [Google Scholar]
  • 25.LaMuraglia GM, Lacaine F, Malt RA. High ornithine decarboxylase activity and polyamine levels in human colorectal neoplasia. Ann Surg. 1986;204:89–93. doi: 10.1097/00000658-198607000-00013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Koo HB, Sigurdson ER, Daly JM, Berenson M, Groshen S, Decosse JJ. Ornithine decarboxylase levels in the rectal mucosa of patients with colonic neoplasia. J Surg Oncol. 1988;38:240–243. doi: 10.1002/jso.2930380407. [DOI] [PubMed] [Google Scholar]
  • 27.Wolter F, Ulrich S, Stein J. Molecular mechanisms of the chemopreventive effects of resveratrol and its analogs in colorectal cancer: Key role of polyamines? J Nutr. 2004;134:3219–3222. doi: 10.1093/jn/134.12.3219. [DOI] [PubMed] [Google Scholar]
  • 28.Turchanowa L, Dauletbaev N, Milovic V, Stein J. Nonsteroidal anti-inflammatory drugs stimulate spermidine/spermine acetyltransferase and deplete polyamine content in colon cancer cells. Eur J Clin Invest. 2001;31:887–893. doi: 10.1046/j.1365-2362.2001.00901.x. [DOI] [PubMed] [Google Scholar]
  • 29.Carbone PP, Douglas JA, Larson PO, Verma AK, Blair IA, Pomplun M, et al. Phase i chemoprevention study of piroxicam and alpha-difluoromethylornithine. Cancer Epidemiol Biomarkers Prev. 1998;7:907–912. [PubMed] [Google Scholar]
  • 30.Martinez ME, O’Brien TG, Fultz KE, Babbar N, Yerushalmi H, Qu N, et al. Pronounced reduction in adenoma recurrence associated with aspirin use and a polymorphism in the ornithine decarboxylase gene. Proc Natl Acad Sci U S A. 2003;100:7859–64. doi: 10.1073/pnas.1332465100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Miners JO, Birkett DJ. Cytochrome p4502c9: An enzyme of major importance in human drug metabolism. Br J Clin Pharmacol. 1998;45:525–38. doi: 10.1046/j.1365-2125.1998.00721.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Kuehl GE, Lampe JW, Potter JD, Bigler J. Glucuronidation of nonsteroidal anti-inflammatory drugs (nsaids): Identifying the enzymes responsible in human liver microsomes. Drug Metabol Dispos. 2005;33:1027–35. doi: 10.1124/dmd.104.002527. [DOI] [PubMed] [Google Scholar]
  • 33.Takahashi H, Kashima T, Nomoto S, Iwade K, Tainaka H, Shimizu T, et al. Comparisons between in-vitro and in-vivo metabolism of (s)-warfarin: Catalytic activities of cdna-expressed cyp2c9, its leu359 variant and their mixture versus unbound clearance in patients with the corresponding cyp2c9 genotypes. Pharmacogene. 1998;8:365–73. doi: 10.1097/00008571-199810000-00001. [DOI] [PubMed] [Google Scholar]
  • 34.Rettie AE, Wienkers LC, Gonzalez FJ, Trager WF, Korzekwa KR. Impaired (s)-warfarin metabolism catalysed by the r144c allelic variant of cyp2c9. Pharmacogenet. 1994;4:39–42. doi: 10.1097/00008571-199402000-00005. [DOI] [PubMed] [Google Scholar]
  • 35.Ciotti M, Marrone A, Potter C, Owens IS. Genetic polymorphism in the human ugt1a6 (planar phenol) udp-glucuronosyltransferase: Pharmacological implications. Pharmacogenet. 1997;7:485–95. doi: 10.1097/00008571-199712000-00007. [DOI] [PubMed] [Google Scholar]
  • 36.Lampe JW, Bigler J, Horner NK, Potter JD. Udp-glucuronosyltransferase (ugt1a1*28 and ugt1a6*2) polymorphisms in caucasians and asians: Relationships to serum bilirubin concentrations. Pharmacogenet. 1999;9:341–9. doi: 10.1097/00008571-199906000-00009. [DOI] [PubMed] [Google Scholar]
  • 37.Ulrich CM, Bigler J, Sparks R, Whitton J, Sibert JG, Goode EL, et al. Polymorphisms in ptgs1 (=cox-1) and risk of colorectal polyps. Cancer Epidemiol Biomarkers Prev. 2004;13:889–893. [PubMed] [Google Scholar]
  • 38.Ulrich CM, Whitton J, Yu JH, Sibert J, Sparks R, Potter JD, et al. Ptgs2 (cox-2) -765g > c promoter variant reduces risk of colorectal adenoma among nonusers of nonsteroidal anti-inflammatory drugs. Cancer Epidemiol Biomarkers Prev. 2005;14:616–9. doi: 10.1158/1055-9965.EPI-04-0510. [DOI] [PubMed] [Google Scholar]
  • 39.Goodman JE, Bowman ED, Chanock SJ, Alberg AJ, Harris CC. Arachidonate lipoxygenase (alox) and cyclooxygenase (cox) polymorphisms and colon cancer risk. Carcinogenesis. 2004;25:2467–72. doi: 10.1093/carcin/bgh260. [DOI] [PubMed] [Google Scholar]
  • 40.Koh WP, Yuan JM, Van Den Berg D, Lee HP, Yu MC. Interaction between cyclooxygenase-2 gene polymorphism and dietary n-6 polyunsaturated fatty acids on colon cancer risk: The singapore chinese health study. Br J Cancer. 2004;90:1760–1764. doi: 10.1038/sj.bjc.6601797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Lin HJ, Lakkides KM, Keku TO, Reddy ST, Louie AD, Kau IH, et al. Prostaglandin h synthase 2 variant (val511ala) in african americans may reduce the risk for colorectal neoplasia. Cancer Epidemio Biomarkers Prev. 2002;11:1305–15. [PubMed] [Google Scholar]
  • 42.Cox DG, Pontes C, Guino E, Navarro M, Osorio A, Canzian F, et al. Polymorphisms in prostaglandin synthase 2/cyclooxygenase 2 (ptgs2/cox2) and risk of colorectal cancer. Br J Cancer. 2004;91:339–43. doi: 10.1038/sj.bjc.6601906. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Barry E. Ornithine decarboxylase polymorphism modification of response to aspirin treatment for colorectal adenoma prevention. J Natl Cancer Inst. 2006;98:1494–1500. doi: 10.1093/jnci/djj398. [DOI] [PubMed] [Google Scholar]
  • 44.Sansbury LB, Millikan RC, Schroeder JC, North KE, Moorman PG, Keku TO, et al. Cox-2 polymorphism, use of nonsteroidal anti-inflammatory drugs, and risk of colon cancer in african americans (united states) Cancer Causes Control. 2006;17:257–66. doi: 10.1007/s10552-005-0417-0. [DOI] [PubMed] [Google Scholar]
  • 45.Samowitz WS, Wolff RK, Curtin K, Sweeney C, Ma KN, Andersen K, et al. Interactions between cyp2c9 and ugt1a6 polymorphisms and nonsteroidal anti-inflammatory drugs in colorectal cancer prevention. Clin Gastroenterol Hepatol. 2006;4:894–901. doi: 10.1016/j.cgh.2006.04.021. [DOI] [PubMed] [Google Scholar]
  • 46.Oshima M, Dinchuk JE, Kargman SL, Oshima H, Hancock B, Kwong E, et al. Suppression of intestinal polyposis in apc delta716 knockout mice by inhibition of cyclooxygenase 2 (cox-2) Cell. 1996;87:803–9. doi: 10.1016/s0092-8674(00)81988-1. [DOI] [PubMed] [Google Scholar]
  • 47.Chulada PC, Thompson MB, Mahler JF, Doyle CM, Gaul BW, Lee C, et al. Genetic disruption of ptgs-1, as well as ptgs-2, reduces intestinal tumorigenesis in min mice. Cancer Res. 2000;60:4705–8. [PubMed] [Google Scholar]
  • 48.Hansen-Petrik MB, McEntee MF, Jull B, Shi H, Zemel MB, Whelan J. Prostaglandin e(2) protects intestinal tumors from nonsteroidal anti-inflammatory drug-induced regression in apc(min/+) mice. Cancer Res. 2002;62:403–8. [PubMed] [Google Scholar]
  • 49.Fries S, Grosser T, Price TS, Lawson JA, Kapoor S, DeMarco S, et al. Marked interindividual variability in the response to selective inhibitors of cyclooxygenase-2. Gastroenterol. 2006;130:55–64. doi: 10.1053/j.gastro.2005.10.002. [DOI] [PubMed] [Google Scholar]
  • 50.Ulrich CM, Bigler J, Sibert J, Greene EA, Sparks R, Carlson CS, et al. Cyclooxygenase 1 (cox1) polymorphisms in african-american and caucasian populations. Hum Mutat. 2002;20:409–10. doi: 10.1002/humu.9080. [DOI] [PubMed] [Google Scholar]
  • 51.Halushka MK, Walker LP, Halushka PV. Genetic variation in cyclooxygenase 1: Effects on response to aspirin. Clin Pharmacol Ther. 2003;73:122–30. doi: 10.1067/mcp.2003.1. [DOI] [PubMed] [Google Scholar]
  • 52.Scott BT, Hasstedt SJ, Bovill EG, Callas PW, Valliere JE, Wang L, et al. Characterization of the human prostaglandin h synthase 1 gene (ptgs1): Exclusion by genetic linkage analysis as a second modifier gene in familial thrombosis. Blood Coagul Fibrinolysis. 2002;13:519–31. doi: 10.1097/00001721-200209000-00007. [DOI] [PubMed] [Google Scholar]
  • 53.Maree AO, Curtin RJ, Chubb A, Dolan C, Cox D, O’Brien J, et al. Cyclooxygenase-1 haplotype modulates platelet response to aspirin. J Thromb Haemost. 2005;3:2340–5. doi: 10.1111/j.1538-7836.2005.01555.x. [DOI] [PubMed] [Google Scholar]
  • 54.Kulmacz RJ, Wang LH. Comparison of hydroperoxide initiator requirements for the cyclooxygenase activities of prostaglandin h synthase-1 and -2. J Biol Chem. 1995;270:24019–23. doi: 10.1074/jbc.270.41.24019. [DOI] [PubMed] [Google Scholar]
  • 55.Siezen CL, Tijhuis MJ, Kram NR, van Soest EM, de Jong DJ, Fodde R, et al. Protective effect of nonsteroidal anti-inflammatory drugs on colorectal adenomas is modified by a polymorphism in peroxisome proliferator-activated receptor delta. Pharmacogenet Genomics. 2006;16:43–50. doi: 10.1097/01.fpc.0000182778.03180.f3. [DOI] [PubMed] [Google Scholar]
  • 56.Papafili A, Hill MR, Brull DJ, McAnulty RJ, Marshall RP, Humphries SE, et al. Common promoter variant in cyclooxygenase-2 represses gene expression: Evidence of role in acute-phase inflammatory response. Arterioscler, Thromb Vasc Biol. 2002;22:1631–6. doi: 10.1161/01.atv.0000030340.80207.c5. comment. [DOI] [PubMed] [Google Scholar]
  • 57.Orbe J, Beloqui O, Rodriguez JA, Belzunce MS, Roncal C, JA P. Protective effect of the g-765c cox-2 polymorphism on subclinical atherosclerosis and inflammatory markers in asymptomatic subjects with cardiovascular risk factors. Clin Chim Acta. 2006;368:138–143. doi: 10.1016/j.cca.2005.12.019. [DOI] [PubMed] [Google Scholar]
  • 58.Fritsche E, Baek SJ, King LM, Zeldin DC, Eling TE, Bell DA. Functional characterization of cyclooxygenase-2 polymorphisms. J Pharmacol Exp Ther. 2001;299:468–76. [PubMed] [Google Scholar]
  • 59.Guo Y, Harris RB, Rosson D, Boorman D, O’Brien TG. Functional analysis of human ornithine decarboxylase alleles. Cancer Res. 2000;60:6314–7. [PubMed] [Google Scholar]
  • 60.Bigler J, Whitton J, Lampe JW, Fosdick L, Bostick RM, Potter JD. Cyp2c9 and ugt1a6 genotypes modulate the protective effect of aspirin on colon adenoma risk. Cancer Res. 2001;61:3566–9. [PubMed] [Google Scholar]
  • 61.Chan AT, Tranah GJ, Giovannucci EL, Hunter DJ, Fuchs CS. Genetic variants in the ugt1a6 enzyme, aspirin use, and the risk of colorectal adenoma. J Natl Cancer Inst. 2005;97:457–60. doi: 10.1093/jnci/dji066. [DOI] [PubMed] [Google Scholar]
  • 62.Hubner RA, Muir KR, Liu JF, Logan RF, Grainge M, Armitage N, et al. Genetic variants of ugt1a6 influence risk of colorectal adenoma recurrence. Clin Cancer Res. 2006;12:6585–9. doi: 10.1158/1078-0432.CCR-06-0903. [DOI] [PubMed] [Google Scholar]
  • 63.Poole E, Bigler J, Whitton J, Potter J, Sibert J, Ulrich C. Prostacyclin synthase and arachidonate 5-lipoxygenase polymorphisms and risk of colorectal polyps. Cancer Epidemiol Biomarkers Prev. 2006;15:502–508. doi: 10.1158/1055-9965.EPI-05-0804. [DOI] [PubMed] [Google Scholar]
  • 64.Singh G. Gastrointestinal complications of prescription and over-the-counter nonsteroidal anti-inflammatory drugs: A view from the aramis database. Arthritis, rheumatism, and aging medical information system. Am J Ther. 2000;7:115–21. doi: 10.1097/00045391-200007020-00008. [DOI] [PubMed] [Google Scholar]
  • 65.Hansson L, Zanchetti A, Carruthers S, Dahlof B, Elmfeldt D, Julius S, et al. Effects of intensive blood pressure lowering and low dose aspirin in patients with hypertension: Principal results of the hypertension optimal treatment (hot) randomised trial. Lancet. 1998;351:1755–1762. doi: 10.1016/s0140-6736(98)04311-6. [DOI] [PubMed] [Google Scholar]
  • 66.Steering committee of the physicians’ health study research group Final report on the aspirin component of the ongoing physicians’ health study. N Engl J Med. 1989;321:129–135. doi: 10.1056/NEJM198907203210301. [DOI] [PubMed] [Google Scholar]
  • 67.McAdam BF, Catella-Lawson F, Mardini IA, Kapoor S, Lawson JA, FitzGerald GA. Systemic biosynthesis of prostacyclin by cyclooxygenase (cox)-2: The human pharmacology of a selective inhibitor of cox-2. Proc Natl Acad Sci U S A. 1999;96:272–7. doi: 10.1073/pnas.96.1.272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Catella-Lawson F, McAdam B, Morrison BW, Kapoor S, Kujubu D, Antes L, et al. Effects of specific inhibition of cyclooxygenase-2 on sodium balance, hemodynamics, and vasoactive eicosanoids. J Pharmacol Exp Ther. 1999;289:735–741. [PubMed] [Google Scholar]
  • 69.Cheng Y, Austin SC, Rocca B, Koller BH, Coffman TM, Grosser T, et al. Role of prostacyclin in the cardiovascular response to thromboxane a2. Science. 2002;296:539–541. doi: 10.1126/science.1068711. [DOI] [PubMed] [Google Scholar]
  • 70.Psaty BM, Potter JD. Risks and benefits of celecoxib to prevent recurrent adenomas. N Engl J Med. 2006;355:950–952. doi: 10.1056/NEJMe068158. [DOI] [PubMed] [Google Scholar]
  • 71.Silverstein FE, Faich G, Goldstein JL, Simon LS, Pincus T, Whelton A, et al. Gastrointestinal toxicity with celecoxib vs nonsteroidal anti-inflammatory drugs for osteoarthritis and rheumatoid arthritis: The class study: A randomized controlled trial. Celecoxib long-term arthritis safety study. JAMA. 2000;284:1247–55. doi: 10.1001/jama.284.10.1247. [DOI] [PubMed] [Google Scholar]
  • 72.Solomon SD, McMurray JJ, Pfeffer MA, Wittes J, Fowler R, Finn P, et al. Cardiovascular risk associated with celecoxib in a clinical trial for colorectal adenoma prevention. N Engl J Med. 2005;352:1071–80. doi: 10.1056/NEJMoa050405. [DOI] [PubMed] [Google Scholar]
  • 73.Solomon SD, Pfeffer MA, McMurray JJ, Fowler R, Finn P, Levin B, et al. Effect of celecoxib on cardiovascular events and blood pressure in two trials for the prevention of colorectal adenomas. Circulation. 2006;114:1028–1035. doi: 10.1161/CIRCULATIONAHA.106.636746. [DOI] [PubMed] [Google Scholar]
  • 74.Motsko SP, Rascati KL, Busti AJ, Wilson JP, Barner JC, Lawson KA, et al. Temporal relationship between use of nsaids, including selective cox-2 inhibitors, and cardiovascular risk. Drug Safety. 2006;29:621–632. doi: 10.2165/00002018-200629070-00007. [DOI] [PubMed] [Google Scholar]
  • 75.Peterson SK, Watts BG, McGivern B, Burke S, Latchford A, Phillips R, et al. Collaborative Group of the Americas on Inherited Colon Cancer . Fap-affected adults’ responses to reported cardiovascular risks associated with celecoxib. Utah; Salt Lake City: 2005. [Google Scholar]
  • 76.Ovid, medline®. Ovid Technologies Inc., Copyright 2000-2006.
  • 77.Ali IU, Luke BT, Dean M, Greenwald P. Allelic variants in regulatory regions of cyclooxygenase-2: association with advanced colorectal adenoma. Br J Cancer. 2005;93:953–9. doi: 10.1038/sj.bjc.6602806. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES