Esophageal Cancer Open Access
Copyright ©2005 Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. May 7, 2005; 11(17): 2531-2538
Published online May 7, 2005. doi: 10.3748/wjg.v11.i17.2531
Phase I/II enzyme gene polymorphisms and esophageal cancer risk: A meta-analysis of the literature
Chun-Xia Yang, Department of Epidemiology, Huaxi Public Health School, Sichuan University, Chengdu 610041, Sichuan Province, China
Chun-Xia Yang, Keitaro Matsuo, Kazuo Tajima, Division of Epidemiology and Prevention, Aichi Cancer Center Research Institute, Nagoya 464-8681, Japan
Zhi-Ming Wang, Huaxi Public Health School, Sichuan University, Chengdu 610041, Sichuan Province, China
Author contributions: All authors contributed equally to the work.
Correspondence to: Keitaro Matsuo, Division of Epidemiology and Prevention, Aichi Cancer Center Research Institute, 1-1 Kanokoden, Chikusa-ku, Nagoya 464-8681, Japan. kmastuo@aichi-cc.jp
Telephone: +81-52-762-6111 Fax: +81-52-763-5233
Received: September 24, 2004
Revised: September 25, 2004
Accepted: November 19, 2004
Published online: May 7, 2005

Abstract

AIM: Phase I/II enzymes metabolize environmental carcin-ogens and several functional polymorphisms have been reported in their encoding genes. Although their significance with regard to esophageal carcinogenicity has been examined epidemiologically, it remains controversial. The present systematic review of the literature was performed to clarify associations.

METHODS: Eligible studies were case-control or cohort studies published until September 2004 that were written in any language. From PubMed and a manual review of refe-rence lists in relevant review articles, we obtained 16 studies related to the CYP1A1 Ile-Val substitution in exon 7, CYP1A1 MspI polymorphisms, CYP2E1 RsaI polymorphisms, GSTM1 null type, GSTT1 null type and GSTP1 Ile104Val. All were of case-control design. Summary statistics were odds ratios (ORs) comparing heterozygous-, homozygous-non-wild type or these two in combination with the homozygous wild type, or the null type with the non-null type for GSTM1 and GSTT1. A random effect model was used to estimate the summary ORs. A meta-regression analysis was applied to explore sources of heterogeneity.

RESULTS: Individuals with the Ile-Val substitution in CYP1A1 exon 7 had increased esophageal cancer risk, with ORs (95%CI) compared with Ile/Ile of 1.37 (1.09-1.71), 2.52 (1.62-3.91) and 1.44 (1.17-1.78) for Ile-Val, Val/Val genotype and the combined group. No significant association was found between esophageal cancer risk and the other genetic parameters.

CONCLUSION: A significant association exists between the CYP1A1 Ile-Val polymorphism and risk of esophageal cancer. Polymorphisms that increase the internal exposure to activated carcinogens may increase the risk of esophageal cancer.

Key Words: CYPs, GSTs, Gene polymorphisms, Esophageal cancer, Meta-analysis



INTRODUCTION

Most environmental chemical carcinogens undergo activation by phase I enzymes, often in an oxidation reaction, and detoxication by phase II enzymes. The cytochrome P450 enzyme superfamily constitutes the majority of phase I enzymes, while the glutathione-S-transferases (GSTs) and N-acetyltransferase are primarily responsible for the detoxication of xenobiotics. The drug-metabolizing enzymes often display genetic polymorphisms, which may alter the enzyme activity and thus impact on the risk of cancer.

The enzyme CYP1A1 is involved in the activation of major classes of tobacco procarcinogens, like polyaromatic hydrocarbons and aromatic amines, and is present in many epithelial tissues[1]. CYP1A1 Ile-Val substitution in the heme-binding region results in a two-fold increase in microsomal enzyme activity and is in complete linkage disequilibrium in Caucasians with the CYP1A1 MspI polymorphism, which has also been associated experimentally with increased catalytic activity[2]. CYP2E1 is primarily responsible for the metabolic activation of many low molecular weight carcinogens[3], including certain nitrosamines, which may be involved in carcinogenesis of the esophagus. This enzyme is also believed to participate in the oxidation of other compounds, such as ethanol, to produce reactive free radicals that may initiate lipid peroxidation and consequently influence carcinogenesis[4]. The variant c2 allele recognized by RsaI digestion in the 5’-flanking region of the gene appears to be associated with decreased enzyme activity[5].

GSTs are a family of multifunctional enzymes which metabolize a variety of xenobiotics with a large overlap in the substrate specificity. Individuals who are homozygous for the null GSTM1 or null GSTT1 alleles lack the respective enzyme functions[6,7]. GSTP1 is a major GST isoform expressed in human esophagus[8], which can eliminate DNA oxidative products of thymidine or uracil propenal[9]. After induction by cytochrome P450, some cigarette-related carcinogens, such as benzo[a]pyrene diol epoxide and acrolein, can also be eliminated by GSTP1[10]. The Ile-Val substitution at residue 104 may be associated with a higher level of DNA adducts[11], thus increasing the susceptibility to cancer induction.

Therefore, the CYP1A1 Val allele, the CYP1A1 MspI non-wild allele, the null type of GSTM1 and GSTT1 as well as the GSTP1 Val allele may increase the risk of esophageal cancer, while the CYP2E1 c2 allele (recognized by RsaI digestion) may decrease the risk. Based on the possible biological significance of CYP1A1, CYP2E1, GSTM1, GSTT1 and GSTP1 polymorphisms on cancer susceptibility, several epidemiologic studies have been conducted to assess their association with esophageal cancer. However, most studies featured only small samples and the results were not always consistent. To obtain a better understanding of the significance of gene polymorphisms with regard to esophageal cancer risk, we performed a systematic review of all the relevant studies published in the literature.

MATERIALS AND METHODS
Selection of studies

Before the study, we defined inclusion criteria as follows: (1) any study design giving relative risk (an OR or a risk ratio) for candidate gene (CYP1A1, CYP2E1, GSTM1, GSTT1, and GSTP1) polymorphisms regarding the risk of esophageal cancer (including both squamous cell carcinomas and adenocarcinomas); (2) inclusion of non-cancer or disease-free subjects as a control group; (3) already published in any language but cited in PubMed.

All the studies were obtained via PubMed using key words “CYP1A1”, “CYP2E1”, “GSTM1”, “GESTT1” and “GSTP1” in combination with “esophageal cancer” to identify potentially relevant articles. A total of 45 articles were captured, and 21, 28, 20, 12, and 12 were related to CYP1A1, CYP2E1, GSTM1, GSTT1, and GSTP1, respectively. We selected all the studies, which provided a relative risk with the candidate gene.

We examined abstracts of all the candidate articles to decide whether to include/exclude in the further detailed review. Thereby, we excluded a total of 22 studies due to inappropriate study design; among them, 8, 18, 5, 4, and 4 were related to CYP1A1, CYP2E1, GSTM1, GSTT1, and GSTP1, respectively. Among the 22 excluded articles, five were reviews[1, 12-15], three concerned the expression of cytochrome P450 (CYPs) in esophageal mucosa[16-18], four covered animal experiments[19-22], three compared gene polymorphism frequencies among different populations[23-25], one focused on the metabolism of N-nitrosobenzylmethylamine by human cytochrome P450 enzyme[26], one was related to gastric cancer, not esophageal cancer[27], and other five were incompatible with the inclusion criteria[28-32].

Other studies were further excluded based upon detailed review because in three cases[33-35] they were the same study as in two other papers[36]. The newest two studies were retained for the analysis. Three more studies were excluded because they did not provide relevant information required for our analysis. Most of them did not apply subjects with other diseases as control groups and did not provide relative risk of esophageal cancer for candidate gene polymorphisms[37-39]. One study was excluded since it was the only example which examined associations between a tandem repeat polymorphism of CYP2E1 and the risk of cancer[40].

Finally, a total of 16 case-control studies were included in the meta-analysis (Table 1), 9, 5, 12, 6 and 7 concerning CYP1A1, CYP2E1, GSTM1, GSTT1 and GSTP1, respectively. All the potentially relevant articles were reviewed by two independent investigators (Y.CX. and M.K.).

Table 1 Summary of studies included in the analysis of CYP1A1, CYP2E1, GASTM1, GSTT1 and GSTP1.
StudyYrStudy designCases Mean ageControls Mean ageCases Man (%)Controls Man (%)PolymorphismsResultAdjusted factorsReference
Hori H19972NANA78 (83)NACYP1A1, MspINS42
CYP1A1, IIe-ValNS
GSTM1NS
CYP2E1, RsalNS
Nimura Y19971NANA67 (75)76 (55)CYP1A1, IIe-ValS43
GSTM1NS
Morita S1997262.250.545 (85)112 (85)CYP1A1, IIe-ValNS44
CYP2E1, RsalNS
GSTM1NS
Morita S1998262.149.856 (85)102 (62)GSTP1S45
Lin DX1998NA55.553.327 (60)23 (50)CYP2E1, RsalSAdjusted by age and sex36
CYP2E1,DralNS
GSTM1NS
GSTT1NS
GSTP1NS
Van Lieshout EM19992NA5227 (79)98 (40)CYP1A1, MspIS46
CYP1A1, IIe-ValNS
GSTM1NS
GSTT1NS
GSTP1S
Shao G20002555374 (69)80 (72)CYP1A1, IIe-ValNS47
GSTM1S
Lee JM20002NANA82 (91)228 (89)GSTP1NSAdjusted by potential factors48
Tan W2000254.553.699 (66)99 (66)CYP2E1, RsalSAdjusted by age, sex and smoking49
GSTM1S
GSTT1NS
GSTP1NS
Wu M-T2002160.661.2133 (91)298 (92)CYP1A1, MspINSAdjusted by age, sex, smoking and alcohol, etc.50
CYP1A1, IIe-ValS
Gao CM20022NANA55 (59)131 (66)CYP2E1, RsalNSAdjusted by age, sex and potential factors51
Yokoyama A2002261.758.8234 (100)634 (100)GSTM1NS52
Gao CM20022NANA78 (55)149 (67)GSTM1SAdjusted by age and sex53
GSTT1NS
Wang LD20032NANA32 (53)NACYP1A1, MspINS54
CYP1A1, IIe-ValNS
GSTM1NS
GSTT1NS
GSTP1NS
Casson AG20032NANA38 (84)38 (84)CYP1A1, MspINSAdjusted by age, sex and smoking55
CYP1A1, IIe-ValNS
GSTM1NS
GSTT1NS
GSTP1S
Wang AH20041NANA97 (76)78 (77)CYP1A1, IIe-ValS37
GSTM1S

We also tried to use “esophageal” combined with candidate genes as keywords to search for much more relevant articles as well as check the reference lists in the reviews and selected original investigations and found no additional eligible articles.

Data abstraction

Two investigators using a standard information extraction form independently abstracted data. Characteristics abstracted from the articles included the name of the first author, year of publication, location of the study, study design, mean age for all cases and controls, the percentage of males in the case and control groups, matched factors as well as adjusted factors; number of cases, number of controls, number of cases and controls with each genotype of candidate polymorphisms, and overall crude or adjusted odds ratios (ORs) with their 95%CI. For one study[41], which supplied the result for both present controls and total controls (including historic control and the present control), total control data were selected for our meta-analysis.

Statistical analysis

The STATA statistical package (version 8, stata, College Station, TX) was used for the meta-analysis. The homozygous wild type was used as the reference group for CYP1A1, CYP2E1 and GSTP1, and the non-null type for GSTM1 and GSTT1. With four papers[36,44,47,48] whose reference groups were defined in the opposite way, the ORs were inverted for our analysis. Adjusted ORs were employed for the present meta-analysis if available in the reports, otherwise, crude ORs were used. Since some of the original studies did not provide the ORs but the genotype frequencies were available, crude ORs were then calculated and employed for our meta-analysis. A random-effect model was applied to obtain summary ORs and their 95%CIs since the results with fixed-effect models are the same as with random-effect models if there is no heterogeneity across the studies. A random-effect model should be applied if heterogeneity exists. Publication bias was graphically assessed by funnel plots and statistically assessed by Egger’s test. Meta-regression analysis was applied to explore potential sources of heterogeneity. The factors, study design, Chinese population (yes/no), Asian population (yes/no), publication year (after 2000 or not), number of cases and controls (both greater than 100 or not) and matching (matched for sex and age or not) were examined. Statistical significance was defined as a P-value less than 0.05 except for meta-regression analyses, which used a P-value 0.10 because of the relatively weak statistical power.

RESULTS

In the final analysis, we had a total of 16 case-control studies consisting of 3 hospital-based (controls selected from non-cancer patients), 12 population-based (controls selected from the healthy population) and 1 without a clear type. Among them, 9 were studies of the CYP1A1 exon 7 Ile-Val substitution, 1[53] without any Ile-Val substitution in either cases and controls, 5 concerned the CYP1A1 MspI polymorphism, 5 the CYP2E1 Rsal polymorphism, 12 the GSTM1 null type, 6 the GSTT1 null type and 7 the GSTP1 Ile-Val substitution (Table 1).

For CYP1A1 exon 7 Ile-Val substitution, all ORs for the Ile/Val genotype and the combined group were larger than 1 when compared with the Ile/Ile genotype, although only one study demonstrated a significantly increased risk. In three of eight cases, the Val/Val genotype was associated with significantly increased ORs (Table 2). The meta-analysis with a total of 754 cases and 1563 controls showed significantly increased ORs of 1.37 (1.09-1.71), 2.52 (1.62-3.91) and 1.44 (1.17-1.78) for Ile-Val and Val/Val genotypes and the combined group, respectively. There was no heterogeneity across the studies, so that the results for the fixed-effect model were the same as for the random-effect model for CYP1A1 exon 7 Ile-Val substitution. In contrast, no significantly increased risk of esophageal cancer was observed for the CYP1A1 MspI polymorphism.

Table 2 Summary of the meta-analysis of CYP1A1, CYP2E1, GSTP1 and esophageal cancer risk.
StudyCountryCasesControlCases wt/wtCases hetero1Cases homo1Control wt/wt1Control hetero1Control homo1OR1 (95%CI)23OR2 (95%CI)23OR3 (95%CI)23
CYP1A1 Ile-Val:
Hori HJapan9142852372275133201.47 (0.89-2.41)0.53 (0.06-2.29)1.35 (0.83-2.19)
Nimura YChina89137502613923871.26 (0.65-2.41)3.42 (1.17-10.72)1.59 (0.89-2.87)
Morita SJapan5313232201804931.02 (0.49-2.08)0.83 (0.02-10.84)1.0 (0.5-1.9)
van Lieshout EMNetherlands3424726802073731.72 (0.62-4.30)0 (0-10.62)1.59 (0.58-3.95)
Shao GChina10711143568555151.40 (0.78-2.53)2.05 (0.54-8.49)1.46 (0.83-2.59)
Wu M-TTaiwan146324686216179127181.34 (0.86-2.07)42.48 (1.15-5.34)41.42 (0.94-2.14)
Wang LDChina623830284201621.17 (0.47-2.93)1.33 (0.17-15.97)1.19 (0.49-2.88)
Casson AGCanada454545004500NA6NA6NA6
Wang AHChina1271012156503148221.72 (0.83-3.58)3.35 (1.49-7.61)2.24 (1.14-4.43)
Meta-analysis results754156336729394984499801.37 (1.09-1.71)2.52 (1.62-3.91)1.44 (1.17-1.78)
CYP1A1 MspI:
Hori HJapan9424233501110697391.66 (0.96-2.88)0.91 (0.38-2.06)1.44 (0.86-2.44)
van Lieshout EMNetherlands34247221202073733.05 (1.26-7.08)0 (0-12.63)2.82 (1.17-6.51)
Wu M-TTaiwan146324606521136146420.98 (0.63-1.53)41.24 (0.65-2.36)41.04 (0.68-1.57)
Casson AGCanada454538NANA33NANANANA0.6 (0.2-1.8)4
Wang LDChina623833254122240.41 (0.16-1.08)0.36 (0.06-2.33)0.41 (0.16-1.02)
Meta-analysis results38189618615236494302881.21 (0.64-2.32)1.02 (0.62-1.68)1.07 (0.64-1.80)
CYP2E1:
Hori HJapan7963349246412202191.00 (0.57-1.72)2.66 (0.83-7.33)1.14 (0.68-1.89)
Morita SJapan5313234181854251.07 (0.51-2.22)0.50 (0.01-4.72)1.0 (0.5-2.0)
Lin DXChina45453663202230.15 (0.04-0.48)0.56 (0.07-4.59)0.21 (0.08-0.56)4,5
Tan WChina1501501073112667770.25 (0.14-0.43)1.06 (0.36-3.34)0.31 (0.24-0.40)4,5
Gao CMChina931965531712162131.13 (0.60-2.13)41.23 (0.40-3.77)41.15 (0.64-2.07)4
Meta-analysis results420115628111029704405470.59 (0.28-1.23)1.33 (0.72-2.44)0.63 (0.30-1.30)
GSTP1:
Morita SJapan6616461501134830.19 (0.07-0.52)0 (0-4.6)0.13 (0.04-0.45)5
Lin DXChina423629121221130.83 (0.28-2.51)0.25 (0.005-3.48)0.7 (0.3-1.8)
van Lieshout EMNetherlands342471021314689123.44 (1.47-8.55)3.65 (0.56-16.82)3.47 (1.51-8.46)
Lee JMTaiwan9025465NANA160NANANA6NA60.65 (0.39-1.11)4,5
Tan WChina15015093489915360.89 (0.53-1.48)0.95 (0.58-1.55)1.0 (0.8-1.3)
Wang LDChina623829303241311.91 (0.76-4.89)2.48 (0.18-135.66)1.95 (0.79-4.87)4
Casson AGCanada454519224261272.5 (1.0-6.3)0.8 (0.2-3.1)1.8 (0.8-4.3)4
Meta-analysis results48993430613820582226321.17 (0.55-2.49)1.02 (0.65-1.58)1.01 (0.60-1.70)

For CYP2E1, two out of five investigations suggested that the c2 allele may significantly decrease the risk with adjusted ORs (95%CI) of 0.31 (0.24-0.40) and 0.21 (0.08-0.56) for the homozygous and combined group, respectively. The meta-analysis showed non-significantly decreased ORs for the c1/c2 and combined group (Table 2). For GSTPP1, one of seven showed significantly increased risk with ORs (95%CI) of 3.44 (1.47-8.55), 3.65 (0.56-16.82) and 3.47 (1.51-8.46) for the hetero, homo and combined group, respectively, while one indicated an opposite association. Another study showed a marginally increased OR for the hetero of 2.5 (1.0-6.3) but the meta-analysis generated a null result (Table 2). For GSTM1, 3 of 12 studies showed the null type to significantly increase the risk but the meta-analysis failed to confirm this result (Table 3). For GSTT1, all the studies were homogenous and both the fixed-effect and random-effect models generated the same result. All the studies and the meta-analysis found no relationship between this gene polymorphism and risk of esophageal cancer (Table 3).

Table 3 Summary of the meta-analysis of GSTM1, GSTTI and esophageal cancer risk.
StudyCountryCasesControlsCaseCaseControlControlOR (95%CI)
GSTM1:Non-nullNullNon-nullNull
Hori HJapan9442853412321960.92 (0.57-1.47)
Nimura YChina89137424774631.31 (0.74-2.32)
Morita SJapan53132302377551.1 (0.6-2.0)
Lin DXChina4545252024211.0 (0.4-2.3)3
van Lieshout EMNetherlands3424717171191280.93 (0.42-2.04)
Shao GChina107111683956551.76 (1.03-2.74)
Tan WChina1501501044674760.43 (0.33-0.56)34
Yokoyama AJapan2346341311033133210.77 (0.56-1.05)
Gao CMChina14122335106901332.17 (1.35-3.50)3
Wang LDChina6238352719190.77 (0.32-1.88)
Casson AGCanada4545192620251.1 (0.5-2.7)3
Wang AHChina127101537457441.81 (1.03-3.18)
Meta-analysisresults11812291612569115511361.07 (0.76-1.51)
GSTT1:
Lin DXChina4545261922230.7 (0.3-1.5)3
van Lieshout EMNetherlands34247286198490.87 (0.28-2.29)
Tan WChina150150906091591.11 (0.83-1.43)34
Gao CMChina14122367741041190.90 (0.59-1.39)3
Wang LDChina6238283418201.09 (0.45-2.65)
Casson AGCanada454537833120.6 (0.2-1.7)3
Meta-analysis results4777482762014662820.99 (0.80-1.22)

We also examined publication bias for each polymorphism and only the GSTM1 polymorphism showed a significant existence. Regarding CYP1A1 Ile-Val, the test was far from statistically significant. In addition, the source of heterogeneity was examined by meta-regression analysis for potential factors such as Asian and Chinese population, publication year, study design, and matching. No obvious source of heterogeneity was identified except studies in Asian populations for the GSTP1 polymorphism (Table 4).

Table 4 Results of meta-regression analysis and Egger’s test for publication bias.
Number of studiesEgger’s test Forpublicationbias1 PResults of meta-regression test
Asian Yes/no Coefficient3Chinese Yes/no Coefficient3Publication Year Coefficient3Design 1 or 22 Coefficient3Matching Yes/no Coefficient3
CYP1A1 Ile-Val80.96-0.10.190.11-0.2-0.22
CYP1A1 MspI50.96-0.42-0.67-0.90.01-0.01
CYP2E RsaI50.321-0.890.761.3-0.13
GSTM1120.040.060.210.25-0.34-0.16
GSTT160.110.340.34-0.160.370.16
GSTP170.99-1.254-0.070.880.40.02
DISCUSSION

In this systematic review, we found a significant association between the CYP1A1 Ile-Val polymorphism and the risk of esophageal cancer, while failing to detect links with other gene polymorphisms examined.

The CYPs superfamily, which plays a central part in the metabolism of carcinogens through activating oxidation reactions, may be expressed in esophageal mucosa[16-18]. The CYP1A1 Ile-Val substitution in exon 7 results in a two-fold increase in microsomal enzyme activity[2] and therefore the Val allele would be expected to increase the susceptibility to esophageal cancer. In fact, our results are in line with eight of the studies previously published, although five of them failed to find a significant association, possibly because of small sample sizes (Table 2). One meta-(OR for Val/Val genotype, 1.62 (0.93-2.82)) and one pooled analysis (OR for Val/Val genotype, 1.54 (0.97-1.46)) of lung cancers and another of head and neck cancer (OR for Val/Val type, 1.35 (0.95-1.82)) also showed that the CYP1A1 Val allele may increase cancer risk, although this was not significant[54-56]. MspI polymorphisms in the 3’-flanking region of the CYP1A1 are completely linked with the Ile-Val substitution in exon 7 in Caucasians, which has also been associated experimentally with increased catalytic activity[2]. However, this complete linkage between MspI and Ile-Val substitution could not been found in Asian population[41,49]. The previous five studies on this polymorphism and esophageal cancer risk showed different results. Only one study in Caucasians showed the MspI non-wild allele, which was completely linked with the Val allele in control group to significantly increase the risk of esophageal cancer (Table 2). The meta-analysis showed no significance with ORs around unity (Table 2). This may be because the MspI polymorphism itself does not alter activity of the CYP1A1 enzyme. Increased enzyme activity[2] and susceptibility to esophageal cancer[45] in Caucasians may be because of the high association between the MspI polymorphism and the Ile-Val substitution in exon 7. This should be clarified in further studies.

In contrast to CYP1A1, no association was found in the present meta-analysis with the CYP2E1 c2 allele. CYP2E1 is primarily responsible for metabolic activation of many low molecular weight carcinogens[3], including certain nitrosamines, which may be involved in carcinogenesis of the esophagus. The variant c2 allele appears to be associated with decreased enzyme activity[5]. Possible explanations for the lack of any association found here include (1) a small number of studies, (2) greater influence of other polymorphisms in CYP2E1 such as Dral and tandem repeat polymorphisms and (3) difference in exposure level to xenobiotics across the study populations. These issues must be considered in future investigations.

We also failed to find any association with GST gene polymorphisms. GSTM1 and GSTT1 null type cannot encode functional enzymes and therefore affected individuals would be expected to be more vulnerable to carcinogens. The GSTP1 Ile104Val substitution may also change the enzyme activity of GSTP1 and modulate susceptibility. A meta- and pooled analyses on head and neck cancer showed GSTM1 (OR = 1.32, 95%CI, 1.07-1.62) and GSTT1 (OR = 1.25, 95%CI, 1.00-1.57) to modestly increase susceptibility[54], but most previous studies on esophageal cancer and our meta-analysis failed to find any relationship. Possible explanations include (1) significance of these enzymes may vary with the cancer site; (2) GSTs metabolize a variety of xenobiotics with a large overlap in the substrate specificity and individuals lacking only one functional enzyme also can metabolize the carcinogens by other GST enzymes; and (3) publication bias may exist together with heterogeneity across studies, which may decrease the statistical power.

As is often the case with meta-analyses, there were several limitations to the present study. Possible sources of heterogeneity, such as differences in study design, publication year and countries/ethnicities, must be considered although meta-regression did not demonstrate the existence of any significant variation except in ethnicity for GSTP1. Possible publication bias is another threat for our summary ORs, although it was detected only for GSTM1. In addition, as adjusted ORs are much more accurate than crude ORs but not available for certain studies, and adjusted and matching factors differed across the studies, residual confounding might have influenced our analysis. Finally, literature-based meta-analysis rather than individual data-based meta-analysis could be a potential source of bias.

In conclusion, we found here a significant association between the CYP1A1 Ile-Val polymorphism and the risk of esophageal cancer by systematic review. Harboring the Val allele, expected to increase the internal exposure to activated carcinogens, thus appears to elevate the risk of esophageal cancer.

ACKNOWLEDGMENTS

The first author, Chun-Xia Yang, was the recipient of a “Special Japan-China Sasakawa Medical Fellowship” during the period of research for and compilation of this paper.

Footnotes

Science Editor Guo SY Language Editor Elsevier HK

References
1.  Bartsch H, Nair U, Risch A, Rojas M, Wikman H, Alexandrov K. Genetic polymorphism of CYP genes, alone or in combination, as a risk modifier of tobacco-related cancers. Cancer Epidemiol Biomarkers Prev. 2000;9:3-28.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Landi MT, Bertazzi PA, Shields PG, Clark G, Lucier GW, Garte SJ, Cosma G, Caporaso NE. Association between CYP1A1 genotype, mRNA expression and enzymatic activity in humans. Pharmacogenetics. 1994;4:242-246.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 141]  [Cited by in F6Publishing: 154]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
3.  Yang CS, Yoo JS, Ishizaki H, Hong JY. Cytochrome P450IIE1: roles in nitrosamine metabolism and mechanisms of regulation. Drug Metab Rev. 1990;22:147-159.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 271]  [Cited by in F6Publishing: 280]  [Article Influence: 8.2]  [Reference Citation Analysis (0)]
4.  Albano E, Tomasi A, Persson JO, Terelius Y, Goria-Gatti L, Ingelman-Sundberg M, Dianzani MU. Role of ethanol-inducible cytochrome P450 (P450IIE1) in catalysing the free radical activation of aliphatic alcohols. Biochem Pharmacol. 1991;41:1895-1902.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 113]  [Cited by in F6Publishing: 109]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
5.  Marchand LL, Wilkinson GR, Wilkens LR. Genetic and dietary predictors of CYP2E1 activity: a phenotyping study in Hawaii Japanese using chlorzoxazone. Cancer Epidemiol Biomarkers Prev. 1999;8:495-500.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Pemble S, Schroeder KR, Spencer SR, Meyer DJ, Hallier E, Bolt HM, Ketterer B, Taylor JB. Human glutathione S-transferase theta (GSTT1): cDNA cloning and the characterization of a genetic polymorphism. Biochem J. 1994;300:271-276.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Strange RC, Fryer AA. The glutathione S-transferases: influence of polymorphism on cancer susceptibility. IARC Sci Publ. 1999;148:231-249.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Nakajima T, Wang RS, Nimura Y, Pin YM, He M, Vainio H, Murayama N, Aoyama T, Iida F. Expression of cytochrome P450s and glutathione S-transferases in human esophagus with squamous-cell carcinomas. Carcinogenesis. 1996;17:1477-1481.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 45]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
9.  Berhane K, Widersten M, Engström A, Kozarich JW, Mannervik B. Detoxication of base propenals and other alpha, beta-unsaturated aldehyde products of radical reactions and lipid peroxidation by human glutathione transferases. Proc Natl Acad Sci USA. 1994;91:1480-1484.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 293]  [Cited by in F6Publishing: 311]  [Article Influence: 10.4]  [Reference Citation Analysis (0)]
10.  Harries LW, Stubbins MJ, Forman D, Howard GC, Wolf CR. Identification of genetic polymorphisms at the glutathione S-transferase Pi locus and association with susceptibility to bladder, testicular and prostate cancer. Carcinogenesis. 1997;18:641-644.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 500]  [Cited by in F6Publishing: 520]  [Article Influence: 19.3]  [Reference Citation Analysis (0)]
11.  Ryberg D, Skaug V, Hewer A, Phillips DH, Harries LW, Wolf CR, Ogreid D, Ulvik A, Vu P, Haugen A. Genotypes of glutathione transferase M1 and P1 and their significance for lung DNA adduct levels and cancer risk. Carcinogenesis. 1997;18:1285-1289.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 265]  [Cited by in F6Publishing: 287]  [Article Influence: 10.6]  [Reference Citation Analysis (0)]
12.  Xing D, Tan W, Lin D. Genetic polymorphisms and susceptibility to esophageal cancer among Chinese population (review). Oncol Rep. 2003;10:1615-1623.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Ribeiro Pinto LF, Teixeira Rossini AM, Albano RM, Felzenszwalb I, de Moura Gallo CV, Nunes RA, Andreollo NA. Mechanisms of esophageal cancer development in Brazilians. Mutat Res. 2003;544:365-373.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 24]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
14.  Zhang Z, Bian J. Progress in researches on the relationship between genetic polymorphisms of alcohol-metabolizing enzymes and cancers. Zhonghua YiXue YiChuanXue ZaZhi. 2001;18:62-65.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Wargovich MJ. Experimental evidence for cancer preventive elements in foods. Cancer Lett. 1997;114:11-17.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 62]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
16.  Godoy W, Albano RM, Moraes EG, Pinho PR, Nunes RA, Saito EH, Higa C, Filho IM, Kruel CD, Schirmer CC. CYP2A6/2A7 and CYP2E1 expression in human oesophageal mucosa: regional and inter-individual variation in expression and relevance to nitrosamine metabolism. Carcinogenesis. 2002;23:611-616.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 44]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
17.  Murray GI, Shaw D, Weaver RJ, McKay JA, Ewen SW, Melvin WT, Burke MD. Cytochrome P450 expression in oesophageal cancer. Gut. 1994;35:599-603.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 50]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
18.  Lechevrel M, Casson AG, Wolf CR, Hardie LJ, Flinterman MB, Montesano R, Wild CP. Characterization of cytochrome P450 expression in human oesophageal mucosa. Carcinogenesis. 1999;20:243-248.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 70]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
19.  Maliakal PP, Coville PF, Wanwimolruk S. Decreased hepatic drug metabolising enzyme activity in rats with nitrosamine-induced tumours. Drug Metabol Drug Interact. 2002;19:13-27.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 10]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
20.  Mori Y, Koide A, Kobayashi Y, Morimura K, Kaneko M, Fukushima S. Effect of ethanol treatment on metabolic activation and detoxification of esophagus carcinogenic N-nitrosamines in rat liver. Mutagenesis. 2002;17:251-256.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 25]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
21.  Chen SC, Wang X, Xu G, Zhou L, Vennerstrom JL, Gonzalez F, Gelboin HV, Mirvish SS. Depentylation of [3H-pentyl]methyl-n-amylnitrosamine by rat esophageal and liver microsomes and by rat and human cytochrome P450 isoforms. Cancer Res. 1999;59:91-98.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Lechevrel M, Wild CP. Absence of a differential induction of cytochrome P450 2E1 by different alcoholic beverages in rats: implications for the aetiology of human oesophageal cancer. Arch Toxicol. 1997;71:690-695.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 6]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
23.  Kato S, Shields PG, Caporaso NE, Hoover RN, Trump BF, Sugimura H, Weston A, Harris CC. Cytochrome P450IIE1 genetic polymorphisms, racial variation, and lung cancer risk. Cancer Res. 1992;52:6712-6715.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Adams CH, Werely CJ, Victor TC, Hoal EG, Rossouw G, van Helden PD. Allele frequencies for glutathione S-transferase and N-acetyltransferase 2 differ in African population groups and may be associated with oesophageal cancer or tuberculosis incidence. Clin Chem Lab Med. 2003;41:600-605.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 31]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
25.  Sepehr A, Kamangar F, Abnet CC, Fahimi S, Pourshams A, Poustchi H, Zeinali S, Sotoudeh M, Islami F, Nasrollahzadeh D. Genetic polymorphisms in three Iranian populations with different risks of esophageal cancer, an ecologic comparison. Cancer Lett. 2004;213:195-202.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 21]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
26.  Morse MA, Lu J, Stoner GD, Murphy SE, Peterson LA. Metabolism of N-nitrosobenzylmethylamine by human cytochrome P-450 enzymes. J Toxicol Environ Health A. 1999;58:397-411.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 12]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
27.  Nishimoto IN, Hanaoka T, Sugimura H, Nagura K, Ihara M, Li XJ, Arai T, Hamada GS, Kowalski LP, Tsugane S. Cytochrome P450 2E1 polymorphism in gastric cancer in Brazil: case-control studies of Japanese Brazilians and non-Japanese Brazilians. Cancer Epidemiol Biomarkers Prev. 2000;9:675-680.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Carstensen U, Alexandrie AK, Högstedt B, Rannug A, Bratt I, Hagmar L. B- and T-lymphocyte micronuclei in chimney sweeps with respect to genetic polymorphism for CYP1A1 and GST1 (class Mu). Mutat Res. 1993;289:187-195.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 34]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
29.  Nomura F, Itoga S, Uchimoto T, Tomonaga T, Nezu M, Shimada H, Ochiai T. Transcriptional activity of the tandem repeat polymorphism in the 5'-flanking region of the human CYP2E1 gene. Alcohol Clin Exp Res. 2003;27:42S-46S.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 21]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
30.  Xing D, Song N, Tan W. Detection of malondialdehyde-DNA adduct level by 32P-postlabeling assay in normal human esophageal epithelium and esophageal squamous cell carcinoma. Zhonghua ZhongLiu ZaZhi. 2001;23:473-476.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Eads CA, Lord RV, Wickramasinghe K, Long TI, Kurumboor SK, Bernstein L, Peters JH, DeMeester SR, DeMeester TR, Skinner KA. Epigenetic patterns in the progression of esophageal adenocarcinoma. Cancer Res. 2001;61:3410-3418.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Tanabe H, Ohhira M, Ohtsubo T, Watari J, Yokota K, Kohgo Y. Genetic polymorphism of aldehyde dehydrogenase 2 in patients with upper aerodigestive tract cancer. Alcohol Clin Exp Res. 1999;23:17S-20S.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 23]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
33.  Lin D, Tang Y, Peng Q. Genetic polymorphisms of cytochrome P450 2E1 and glutathione S-transferase P1 and susceptibility to esophageal cancer. Zhonghua ZhongLiu ZaZhi. 1998;20:94-97.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Lin D, Tang Y, Lu S. Glutathione S-transferase M1, T1 genotypes and the risk of esophageal cancer: a case-control study. Zhonghua LiuXingBingXue ZaZhi. 1998;19:195-199.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Wang AH, Sun CS, Li LS, Huang JY, Chen QS. Relationship of tobacco smoking CYP1A1 GSTM1 gene polymorphism and esophageal cancer in Xi'an. World J Gastroenterol. 2002;8:49-53.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Lin DX, Tang YM, Peng Q, Lu SX, Ambrosone CB, Kadlubar FF. Susceptibility to esophageal cancer and genetic polymorphisms in glutathione S-transferases T1, P1, and M1 and cytochrome P450 2E1. Cancer Epidemiol Biomarkers Prev. 1998;7:1013-1018.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Roth MJ, Dawsey SM, Wang G, Tangrea JA, Zhou B, Ratnasinghe D, Woodson KG, Olivero OA, Poirier MC, Frye BL. Association between GSTM1*0 and squamous dysplasia of the esophagus in the high risk region of Linxian, China. Cancer Lett. 2000;156:73-81.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 25]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
38.  Chao YC, Wang LS, Hsieh TY, Chu CW, Chang FY, Chu HC. Chinese alcoholic patients with esophageal cancer are genetically different from alcoholics with acute pancreatitis and liver cirrhosis. Am J Gastroenterol. 2000;95:2958-2964.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 73]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
39.  Lucas D, Ménez C, Floch F, Gourlaouen Y, Sparfel O, Joannet I, Bodénez P, Jezequel J, Gouérou H, Berthou F. Cytochromes P4502E1 and P4501A1 genotypes and susceptibility to cirrhosis or upper aerodigestive tract cancer in alcoholic caucasians. Alcohol Clin Exp Res. 1996;20:1033-1037.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 52]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
40.  Itoga S, Nomura F, Makino Y, Tomonaga T, Shimada H, Ochiai T, Iizasa T, Baba M, Fujisawa T, Harada S. Tandem repeat polymorphism of the CYP2E1 gene: an association study with esophageal cancer and lung cancer. Alcohol Clin Exp Res. 2002;26:15S-19S.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Hori H, Kawano T, Endo M, Yuasa Y. Genetic polymorphisms of tobacco- and alcohol-related metabolizing enzymes and human esophageal squamous cell carcinoma susceptibility. J Clin Gastroenterol. 1997;25:568-575.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 81]  [Cited by in F6Publishing: 88]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
42.  Nimura Y, Yokoyama S, Fujimori M, Aoki T, Adachi W, Nasu T, He M, Ping YM, Iida F. Genotyping of the CYP1A1 and GSTM1 genes in esophageal carcinoma patients with special reference to smoking. Cancer. 1997;80:852-857.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
43.  Morita S, Yano M, Shiozaki H, Tsujinaka T, Ebisui C, Morimoto T, Kishibuti M, Fujita J, Ogawa A, Taniguchi M. CYP1A1, CYP2E1 and GSTM1 polymorphisms are not associated with susceptibility to squamous-cell carcinoma of the esophagus. Int J Cancer. 1997;71:192-195.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 3]  [Reference Citation Analysis (0)]
44.  Morita S, Yano M, Tsujinaka T, Ogawa A, Taniguchi M, Kaneko K, Shiozaki H, Doki Y, Inoue M, Monden M. Association between genetic polymorphisms of glutathione S-transferase P1 and N-acetyltransferase 2 and susceptibility to squamous-cell carcinoma of the esophagus. Int J Cancer. 1998;79:517-520.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
45.  van Lieshout EM, Roelofs HM, Dekker S, Mulder CJ, Wobbes T, Jansen JB, Peters WH. Polymorphic expression of the glutathione S-transferase P1 gene and its susceptibility to Barrett's esophagus and esophageal carcinoma. Cancer Res. 1999;59:586-589.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Shao G, Su Y, Huang G, Wen B. Relationship between CYP1A1, GSTM1 genetic polymorphisms and susceptibility to esophageal squamous cell carcinoma. Zhonghua LiuXingBingXue ZaZhi. 2000;21:420-423.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Lee JM, Lee YC, Yang SY, Shi WL, Lee CJ, Luh SP, Chen CJ, Hsieh CY, Wu MT. Genetic polymorphisms of p53 and GSTP1,but not NAT2,are associated with susceptibility to squamous-cell carcinoma of the esophagus. Int J Cancer. 2000;89:458-464.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 4]  [Reference Citation Analysis (0)]
48.  Tan W, Song N, Wang GQ, Liu Q, Tang HJ, Kadlubar FF, Lin DX. Impact of genetic polymorphisms in cytochrome P450 2E1 and glutathione S-transferases M1, T1, and P1 on susceptibility to esophageal cancer among high-risk individuals in China. Cancer Epidemiol Biomarkers Prev. 2000;9:551-556.  [PubMed]  [DOI]  [Cited in This Article: ]
49.  Wu MT, Lee JM, Wu DC, Ho CK, Wang YT, Lee YC, Hsu HK, Kao EL. Genetic polymorphisms of cytochrome P4501A1 and oesophageal squamous-cell carcinoma in Taiwan. Br J Cancer. 2002;87:529-532.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 27]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
50.  Gao C, Takezaki T, Wu J, Li Z, Wang J, Ding J, Liu Y, Hu X, Xu T, Tajima K. Interaction between cytochrome P-450 2E1 polymorphisms and environmental factors with risk of esophageal and stomach cancers in Chinese. Cancer Epidemiol Biomarkers Prev. 2002;11:29-34.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 35]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
51.  Yokoyama A, Kato H, Yokoyama T, Tsujinaka T, Muto M, Omori T, Haneda T, Kumagai Y, Igaki H, Yokoyama M. Genetic polymorphisms of alcohol and aldehyde dehydrogenases and glutathione S-transferase M1 and drinking, smoking, and diet in Japanese men with esophageal squamous cell carcinoma. Carcinogenesis. 2002;23:1851-1859.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 168]  [Cited by in F6Publishing: 176]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
52.  Gao CM, Takezaki T, Wu JZ, Li ZY, Liu YT, Li SP, Ding JH, Su P, Hu X, Xu TL. Glutathione-S-transferases M1 (GSTM1) and GSTT1 genotype, smoking, consumption of alcohol and tea and risk of esophageal and stomach cancers: a case-control study of a high-incidence area in Jiangsu Province, China. Cancer Lett. 2002;188:95-102.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 88]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
53.  Casson AG, Zheng Z, Chiasson D, MacDonald K, Riddell DC, Guernsey JR, Guernsey DL, McLaughlin J. Associations between genetic polymorphisms of Phase I and II metabolizing enzymes, p53 and susceptibility to esophageal adenocarcinoma. Cancer Detect Prev. 2003;27:139-146.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 53]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
54.  Hashibe M, Brennan P, Strange RC, Bhisey R, Cascorbi I, Lazarus P, Oude Ophuis MB, Benhamou S, Foulkes WD, Katoh T. Meta- and pooled analyses of GSTM1, GSTT1, GSTP1, and CYP1A1 genotypes and risk of head and neck cancer. Cancer Epidemiol Biomarkers Prev. 2003;12:1509-1517.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Houlston RS. CYP1A1 polymorphisms and lung cancer risk: a meta-analysis. Pharmacogenetics. 2000;10:105-114.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 82]  [Cited by in F6Publishing: 87]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
56.  Le Marchand L, Guo C, Benhamou S, Bouchardy C, Cascorbi I, Clapper ML, Garte S, Haugen A, Ingelman-Sundberg M, Kihara M. Pooled analysis of the CYP1A1 exon 7 polymorphism and lung cancer (United States). Cancer Causes Control. 2003;14:339-346.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 85]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]