Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Genetic Polymorphisms of Glutathione S-Transferase Genes GSTM1, GSTT1 and Risk of Hepatocellular Carcinoma

  • Kang Song ,

    Contributed equally to this work with: Kang Song, Jiayong Yi

    Affiliation Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, People’s Republic of China

  • Jiayong Yi ,

    Contributed equally to this work with: Kang Song, Jiayong Yi

    Affiliation Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai, People’s Republic of China

  • Xizhong Shen ,

    supercai_yu@126.com (YC); xizhongshen001@163.com (XZS)

    Affiliation Department of Gastroenterology, Zhongshan Hospital, Fudan University, Shanghai, People’s Republic of China

  • Yu Cai

    supercai_yu@126.com (YC); xizhongshen001@163.com (XZS)

    Affiliation Department of Gastroenterology, Zhongshan Hospital, Fudan University, Shanghai, People’s Republic of China

Abstract

Background

A number of case-control studies were conducted to investigate the association of glutathione S-transferase (GST) genetic polymorphisms and hepatocellular carcinoma (HCC) risk. However, these studies have yielded contradictory results. We therefore performed a meta-analysis to derive a more precise estimation of the association between polymorphisms on GSTM1, GSTT1 and HCC.

Methodology/Prinicpal Findings

PubMed, EMBASE, ISI web of science and the CNKI databases were systematically searched to identify relevant studies. Data were abstracted independently by two reviewers. Odds ratios (ORs) and 95% confidence intervals (95% CIs) were used to assess the strength of association. Potential sources of heterogeneity were also assessed by subgroup analysis and meta-regression. Funnel plots and Egger’s linear regression were used to test publication bias among the articles. A total of 34 studies including 4,463 cases and 6,857 controls were included in this meta-analysis. In a combined analysis, significantly increased HCC risks were found for null genotype of GSTM1 (OR = 1.29, 95% CI: 1.06–1.58; P = 0.01) and GSTT1 (OR = 1.43, 95% CI: 1.22–1.68; P<10−5). Potential sources of heterogeneity were explored by subgroup analysis and meta-regression. Significant results were found in East Asians and Indians when stratified by ethnicity; whereas no significant associations were found among Caucasians and African populations. By pooling data from 12 studies that considered combinations of GSTT1 and GSTM1 null genotypes, a statistically significant increased risk for HCC (OR = 1.88, 95% CI: 1.41–2.50; P<10−4) was detected for individuals with combined deletion mutations in both genes compared with positive genotypes.

Conclusions/Significance

This meta-analysis suggests that the GSTM1 and GSTT1 null genotype may slightly increase the risk of HCC and that interaction between unfavourable GSTs genotypes may exist.

Introduction

Hepatocellular carcinoma (HCC) is the fifth most common cancer and the third cause of cancer-related death worldwide [1]. The etiologic importance of chronic infection with hepatitis B virus (HBV) and hepatitis C virus (HCV) in HCC has been well established [2], [3]. Not only is HCC an inevitable consequence of chronic HBV or HCV infection, but also other HCC risk factors, such as tobacco smoking, alcohol drinking and aflatoxin exposure, are related to susceptibility to HCC [4], [5]. However, only a minority of patients at risk develops HCC and it is likely that other risk factors such as environmental carcinogenic compounds may contribute to HCC development. This has maintained interest in other biochemical and genetic factors that might contribute to the underlying pathophysiology of HCC.

The glutathione S-transferases (GSTs) are a gene superfamily of phase II metabolic enzymes that detoxify free radicals, particularly in tobacco smoke, products of oxidative stress, and carcinogens such as benzopyrene and other polycyclic aromatic hydrocarbons. The most potent mutagenic and carcinogenic of the aflatoxins is aflatoxins B1 (AFB1) which is mainly metabolized by cytochrome P450 3A4 into the genotoxic metabolite AFB1–8,9-exo-epoxide. This metabolite can bind to DNA, causing G-to-T transversions [6] that may ultimately lead to cancer. Detoxification prevents formation of DNA adducts; the metabolite may be conjugated to glutathione by GSTs or may be hydrolyzed. In addition to their role in phase II detoxification, GSTs also play an important role in modulating the induction of other enzymes and proteins for cellular functions, such as DNA repair [7]. GSTM1 and GSTT1 are the most extensively studied genes in the GST gene superfamily. Polymorphic deletion variants in the GSTM1 and GSTT1 genes produce either a functional enzyme (non-deletion alleles or heterozygous deletion, GSTM1-1 and GSTT1-1) or result in the complete absence of the enzyme (homozygous deletion alleles, GSTM1-null and GSTT1-null) [8]. Therefore, these enzymes may be related to the risk for HCC.

Over the past few years, considerable efforts have been devoted to exploring the relationships between the GSTT1 and GSTM1 null polymorphisms and HCC risk among various populations. However, existing studies have yielded inconsistent results. These disparate findings may be due partly to insufficient power, false-positive results and publication biases. The interpretation of these studies has been further complicated by the use of different control source. In addition, with the increased studies in recent years among Asian, Caucasian, and other populations, there is a need to reconcile these data. We therefore performed a meta-analysis of the published studies to clarify this inconsistency and to establish a comprehensive picture of the relationship between GSTM1, GSTT1 and HCC.

Materials and Methods

Literature Search Strategy and Selection Criteria

Genetic association studies published before the end of June 2012 on HCC and polymorphisms in the GST gene were identified through a search of PubMed, Web of Science, EMBASE and CNKI (Chinese National Knowledge Infrastructure). Search term were keywords relating to the relevant gene (e.g. ‘glutathione S-transferase’, ‘GST’, GSTM1’, GSTT1’) in combination with words related to liver cancer (e.g. ‘Hepatocellular carcinoma’, ‘Liver neoplasm’, ‘Liver cancer’) and polymorphism or variation. Furthermore, reference lists of main reports and review articles were also reviewed by a manual search to identify additional relevant publications.

The included studies have to meet the following criteria: (1) original papers containing independent data, (2) identification of HCC patients was confirmed histologically or pathologically, (3) genotype distribution information or odds ratio (OR) with its 95% confidence interval (CI) and P-value, (4) case–control or cohort studies. The major reasons for exclusion of studies were (1) overlapping data and (2) case-only studies, family based studies, and review articles.

Eligible Studies and Data Extraction

Data extraction was performed independently by two reviewers and differences were resolved by further discussion among all authors. For each included study, the following information was extracted from each report according to a fixed protocol: first author, publication year, definition and numbers of cases and controls, diagnostic criterion, frequency of genotypes, age, sex, cigarette smoking status, alcohol drinking, ethnicity and genotyping method.

Statistical Methods

For the GSTM1 and GSTT1 gene, we estimated the risks of the null genotype on HCC, compared with the non-null genotypes in the recessive model. The strength of the association between the GSTM1 and GSTT1 gene and HCC risk was measured by odds ratios (ORs) with 95% confidence intervals (CIs).

Heterogeneity across individual studies was calculated using the Cochran chi-square Q test followed by subsidiary analysis or by random-effects regression models with restricted maximum likelihood estimation [9], [10]. Random-effects and fixed-effect summary measures were calculated as inverse variance-weighted average of the log OR. The results of random-effects summary were reported in the text because it takes into account the variation between studies. In addition, sources of heterogeneity were investigated by stratified meta-analyses based on ethnicity, source of controls (population or hospital based) and sample size (No. cases ≥200 or <200). Ethnic group was defined as East Asians, Caucasians (i.e. people of European origin), Indians and Africans. 95% CIs were constructed using Woolf’s method [11]. The significance of the overall OR was determined by the Z-test. Ethnicity, sample size, control source, sex distribution among cases and controls were analyzed as covariates in meta-regression. Funnel plots and Egger’s linear regression test were used to assess evidence for potential publication bias [12]. In order to assess the stability of the result, sensitivity analyses were performed, each study in turn was removed from the total, and the remaining were reanalyzed. All statistical analyses were carried out with the Stata software version 10.0 (Stata Corporation, College Station, TX, USA). The type I error rate was set at 0.05 for two-sided analysis.

Results

Characteristics of Studies

The combined search yielded 287 references. 254 articles were excluded because they clearly did not meet the criteria or overlapping references (Figure S1). Finally, a total of 34 studies were retrieved based on the search criteria for HCC susceptibility related to the GST polymorphisms [13][46]. Study quality was assessed according to the score scale for randomized controlled association study proposed by Clark et al [47]. The main study characteristics were summarized in Table 1. There are 33 studies with 4412 cases and 6804 controls concerning GSTM1 and 28 studies with 3892 HCC cases and 6117 controls concerning GSTT1. Of the cases, 74% were East Asians, 10% were Caucasians, 9% were Indians and 7% were African.

thumbnail
Table 1. Characteristics of the studies included in the meta-analysis.

https://doi.org/10.1371/journal.pone.0048924.t001

Association of GSTM1null Polymorphism with HCC

Overall, there was evidence of an association between the increased risk of HCC and the null genotype of GSTM1 when all eligible studies were pooled into the meta-analysis. Using random effect model, the summary OR of GSTM1 null genotype was 1.29 (95% CI: 1.06–1.58, P = 0.01, Figure 1) with statistically significant between-study heterogeneity (P<10−5). This analysis is based on pooling of data from a number of different ethnic populations. When stratifying for ethnicity, significant associations were found for East Asians and Indians with OR of 1.28 (95% CI: 1.02–1.61, P = 0.03) and of 2.45 (95% CI: 1.25–4.79, P = 0.009), respectively. Subsidiary analyses of control source yielded an OR for hospital-based controls of 1.44 (95% CI: 1.02–2.02) and for population-based controls of 1.19 (95% CI: 0.95–1.50). When studies were stratified for sample size, significant risk was found in small studies with OR of 1.30 (95% CI: 1.04–1.62, P = 0.02). However, no significant association was detected among seven large studies (Table 2). In meta-regression analysis, ethnicity (P = 0.63), sample size (P = 0.29), source of controls (P = 0.45), sex distribution in cases (P = 0.27) and controls (P = 0.47) did not significantly explained such heterogeneity.

thumbnail
Figure 1. Meta-analysis of GSTM1 null genotype associated with HCC.

https://doi.org/10.1371/journal.pone.0048924.g001

thumbnail
Table 2. Main results of pooled odds ratios (ORs) with confidence interval (CI) in the meta-analysis.

https://doi.org/10.1371/journal.pone.0048924.t002

Association of GSTT1null Polymorphism with HCC

Significant heterogeneity was present among the 28 studies (P<10−5). In a meta-regression analysis, ethnicity (P = 0.53), sample size (P = 0.32), source of controls (P = 0.14), and gender distribution among cases (P = 0.30) and controls (P = 0.38) explained little heterogeneity. Overall, significantly increased HCC risk was found for GSTT1 null genotype (OR = 1.43, 95% CI: 1.22–1.68, P<10−5; Figure 2).

thumbnail
Figure 2. Meta-analysis of GSTT1 null genotype associated with HCC.

https://doi.org/10.1371/journal.pone.0048924.g002

In the subgroup analyses by ethnicity, significant risks were found among East Asians with OR of 1.40 (95% CI: 1.18–1.66, P<10−4). However, no such associations were detected among Caucasians, African and Indian populations (Table 2). Subsidiary analyses of sample size yielded an OR for small studies of 1.48 (95% CI: 1.24–1.75, P<10−5), while no significant results were found for large studies. By considering control source subgroups, the OR was 1.28 (95% CI: 1.05–1.55, P = 0.01) in population-based controls, compared to 1.66 (95% CI: 1.30–2.12, P<10−5) in hospital controls.

Gene–gene Interaction

The effect of each genotype of GSTs was independently assessed. Significant association was established between the null genotype of GSTM1, GSTT1 and HCC. The data on both null genotype of GSTs among cases and controls were available in 12 studies which included 1762 cases and 2443 controls. The interaction between GSTM1 null and GSTT1 null, for which an OR of 1.88 (95% CI: 1.41–2.50, P<10−4; figure3) for HCC appeared in compared with individuals with the positive genotypes.

thumbnail
Figure 3. Forest plot for association of dual null genotype of GSTM1/GSTT1 and HCC risk.

https://doi.org/10.1371/journal.pone.0048924.g003

Sensitivity Analyses and Publication Bias

A single study involved in the meta-analysis was deleted each time to reflect the influence of the individual data-set to the pooled ORs, and the corresponding pooled ORs were not qualitatively altered. Begg’s funnel plot and Egger’s test were performed to access the publication bias of the literatures. The shape of the funnel plots was symmetrical for these polymorphisms (Figure S2 and S3). The statistical results still did not show publication bias in these studies for GSTM1 (Egger test, P = 0.90) and GSTT1 (Egger test, P = 0.55).

Discussion

Large sample and unbiased epidemiological studies of predisposition genes polymorphisms could provide insight into the in vivo relationship between candidate genes and diseases. This is the most comprehensive meta-analysis that examined the GSTM1, GSTT1 null polymorphism and the relationship to susceptibility for hepatocellular carcinoma. Its strength was based on the accumulation of published data giving greater information to detect significant differences. In total, the meta-analysis involved 34 studies for hepatocellular carcinoma, which provided 4463 cases and 6857 controls.

Our results demonstrated that the GSTM1 and GSTT1 null polymorphism is a risk factor for developing hepatocellular carcinoma. However, this association became non-significant when the meta-analysis was restricted to larger studies, suggesting potential small studies effects. In the stratified analysis by ethnicity, significant associations were found in East Asians for the two polymorphisms. Significant result was also found among Indians for GSTT1 null polymorphism. However, no significant associations were detected among Caucasian and African populations for the two polymorphisms. There are several possible reasons for such differences. Firstly, the frequencies of the risk-association homozygous null genotype vary between different races. For example, the homozygous null genotype distributions of the GSTT1 polymorphism varies between East Asian, Indian and African populations, with a prevalence of ∼52%, ∼29%, and ∼31%, respectively [19], [22], [39]. Therefore, additional studies are warranted to further validate ethnic difference in the effect of these polymorphisms on HCC risk. Secondly, such different results could also be explained by study design or sample size. Besides, other confounding factors, such as age, sex, life style should also be considered. Thus, further prospective researches are needed to examine the effect of this variation on HCC to make a comprehensive and true conclusion [18]. Other confounding factors, such as age, life style (e.g. smoking, alcohol consumption) should also be considered [22], [27], [36]. Nevertheless, owing to the limited number of relevant studies among Caucasians and Africans included in this meta-analysis, the observed ethnic difference in this meta-analysis is also likely to be caused by chance because studies with small sample sizes may have insufficient statistical power to detect a slight effect or may have generated a fluctuated risk estimate. Thus, further studies including a wider spectrum of subjects to investigate the role of this variant in these populations will be needed.

Our results indicated that significantly increased HCC risk in GSTM1 null genotype was found among the hospital-based studies but not in population-based studies. Possible reason could be that the hospital-based studies have some biases because such controls may just represent a sample of ill-defined reference population, and may not be representative of the general population very well, particularly when the genotypes under investigation were associated with the disease conditions that the hospital-based controls may have. Therefore, using a proper and representative population-based control subjects is very important to reduce biases in such genetic association studies.

If genetic susceptibility to HCC is, in part, mediated through metabolic gene polymorphisms, it is possible that the combinations of certain genotypes may be more discriminating as risk factors for HCC than a single locus genotype. Among the 34 studies included in the present meta-analysis, 12 studies investigated the interaction between GSTM1 and GSTT1 polymorphism. By pooling the collected data on GSTM1 and GSTT1 genotypes, a statistically significant 1.88-fold increased risk for HCC appeared for individuals with combined deletion mutations in GSTT1 and GSTM1 genes in comparison with individuals with the positive genotypes. This result suggests that in the presence of both of the two risk factors, an important number of HCC cases would occur.

It is well accepted that the strength of an association is not an inherent biologic property with small associations potentially reflecting important causal relations. It may take relatively few common genetic variants, each conveying only small to modest excess risk, to account for a sizable portion of the population attributable fraction for common diseases (e.g., 10–18 genes, each with a 20–30 percent prevalence and conveying an odds ratio of only 1.2–1.5 to explain between 30 and 50 percent of the population attributable fraction [48]. Recently, several other drug metabolizing genes were found to be associated with HCC susceptibility. N-acetyltransferase 2 and CYP1A1 genotype were found to be associated with increased risk of HCC among smokers [49], [50]. Liu et al. reported that CYP2E1 PstI/RsaI polymorphism was significantly associated with increased HCC susceptibility among alcohol drinkers [51]. Therefore, these gene/polymorphisms which works in close network with each other should be address in future studies.

A number of factors predict HCC, however, detailed pathogenesis mechanisms of HCC remain a matter of speculation. Members of the GST family are important candidates for involvement in susceptibility to commonly occurring forms of cancer, because they may regulate an individual’s ability to metabolize environmental carcinogens [52]. Normal or increased GST enzyme activity or levels may protect susceptible tissues from somatic mutations in DNA by facilitating the conjugation and subsequent elimination of electrophilic carcinogens [53]. Absent or deficient GST enzyme activity may result in poorer elimination of electrophilic carcinogens, particularly in the presence of very active electrophilic activation by phase I enzymes. If an individual’s inherited genotype at a GST locus does not permit the efficient metabolism of compounds involved in carcinogens, then that individual may be at increased cancer risk. GSTM1 and GSTT1 are two important members of GSTs supergene family, which plays an important role in the second stage of biotransformation by conjugating extraneous chemicals with glutathione [54]. Previous some studies have showed that the deficient types of GSTM1 and GSTT1, namely GSTM1-null and GSTT1-null genotypes are completely lack of respective enzyme activity and cannot metabolize transfer AFB1-epoxide, the most important metabolic carcinogen of AFB1, to un-toxic metabolite which is highly soluble and can be excreted out of body, which may be associated with the increased risk of HCC [18], [32].

Compared with the previous meta-analysis [55][57], the present study is much larger. In addition, we assessed not only the effect of each genotype of GSTs and HCC independently but also the interaction effect between GSTM1 and GSTT1. Furthermore, we also performed meta-regression to evaluate the effect of several preselected factors on the observed variability among studies evaluating GSTM1 or GSTT1 null genotype. Our results suggest an overestimation of the true genetic association by small studies, consistent with the phenomenon known as “winner’s curse” [58], [59].

In interpreting the results, some limitations of this meta-analysis should be addressed. Firstly, heterogeneity is a potential problem when interpreting all the results of meta-analysis. Although we minimized the likelihood by performing a careful search for published studies, using the explicit criteria for study inclusion, the significant between-study heterogeneity still existed in most of comparison. The presence of heterogeneity can result from differences in the age distribution, selection of controls, prevalence lifestyle factors and so on. Secondly, the subgroup meta-analyses considering interactions between GSTM1, GSTT1 null genotype and ethnic difference, as well as between GSTT1 null and GSTM1 null genotypes, were performed on the basis of a fraction of all the possible data to be pooled, so selection bias may have occurred and our results may be over inflated. In this context, more reliable results can be expected if individual data are available for a pooled analysis. Finally, lacking the original data of the reviewed studies limited our further evaluation of potential interactions because the interactions between gene-environment, such aflatoxin B1 exposure, HBV or HCV infection, smoking and alcohol abuse.

Despite these limitations, this meta-analysis suggests that the null genotypes of GSTM1 and GSTT1 may increase the risk of HCC, particularly in East Asian population. In addition, the combination of unfavourable genotypes of GSTM1 and GSTT1 may result in an additional risk of HCC. For future association studies, strict selection of patients and controls, larger studies of different ethnic populations will be required. Moreover, gene–gene and gene–environment interactions should also be considered in future studies.

Supporting Information

Figure S1.

Flow diagram of the study selection process.

https://doi.org/10.1371/journal.pone.0048924.s001

(TIF)

Figure S2.

Begg’s funnel plot for publication bias in selection of studies on GSTM1 polymorphism.

https://doi.org/10.1371/journal.pone.0048924.s002

(TIF)

Figure S3.

Begg’s funnel plot for publication bias in selection of studies on GSTT1 polymorphism.

https://doi.org/10.1371/journal.pone.0048924.s003

(TIF)

Author Contributions

Conceived and designed the experiments: YC. Performed the experiments: KS JYY XZS YC. Analyzed the data: KS JYY XZS YC. Contributed reagents/materials/analysis tools: KS JYY XZS. Wrote the paper: KS JYY YC.

References

  1. 1. Parkin DM, Bray F, Ferlay J, Pisani P (2001) Estimating the world cancer burden: GLOBOCAN 2000. Int J Cancer 94: 153–156.
  2. 2. Montesano R, Hainaut P, Wild CP (1997) Hepatocellular carcinoma: from gene to public health. J Natl Cancer Inst 89: 1844–1851.
  3. 3. Bruix J, Barrera JM, Calvet X, Ercilla G, Costa J, et al. (1989) Prevalence of antibodies to hepatitis C virus in Spanish patients with hepatocellular carcinoma and hepatic cirrhosis. Lancet 2: 1004–1006.
  4. 4. Ross RK, Yuan JM, Yu MC, Wogan GN, Qian GS, et al. (1992) Urinary aflatoxin biomarkers and risk of hepatocellular carcinoma. Lancet 339: 943–946.
  5. 5. Marrero JA, Fontana RJ, Fu S, Conjeevaram HS, Su GL, et al. (2005) Alcohol, tobacco and obesity are synergistic risk factors for hepatocellular carcinoma. J Hepatol 42: 218–224.
  6. 6. Wang JS, Groopman JD (1999) DNA damage by mycotoxins. Mutat Res 424: 167–181.
  7. 7. Hayes J, Pulford D (1995) The glutathione S-transferase supergene family: Regulation of GST and the contribution of the isoenzymes to cancer chemoprevention and drug resistance. Crit Rev Biochem Mol Biol 30: 445–600.
  8. 8. Hayes JD, Flanagan JU, Jowsey IR (2005) Glutathione transferases. Annu Rev Pharmacol Toxicol 45: 51–88.
  9. 9. Cochran WG (1954) The combination of estimates from different experiments. Biometrics 10: 101–129.
  10. 10. DerSimonian R, Laird N (1986) Meta-analysis in clinical trials. Control Clin Trials 7: 177–188.
  11. 11. Woolf B (1955) On estimating the relation between blood group and disease. Ann Hum Genet 19: 251–253.
  12. 12. Egger M, Davey Smith G, Schneider M, Minder C (1997) Bias in meta-analysis detected by a simple, graphical test. British Med J 315: 629–634.
  13. 13. Dong CH, Yu SC, Chen GC, Zhao DM, Fu YP (1997) Polymorphisms of GSTT 1 and M 1 genotypes and their effects on elevated Aflatoxin exposure and increased risk of hepatocellular carcinoma. Zhong Liu Fang Zhi Yan Jiu 24: 327–329.
  14. 14. Yu MW, Chiu YH, Chiang YC, Chen CH, Lee TH, et al. (1999) Plasma carotenoids, glutathione S-transferase M1 and T1 genetic polymorphisms, and risk of hepatocellular carcinoma: independent and interactive effects. Am J Epidemiol 149: 621–629.
  15. 15. Wu HL, Chen MN, Liu PX, Zhang RN (2000) Relationship between GSTM1 gene polymorphism and genetic susceptibility to primary hepatocellular carcinoma. Pract J Cancer 15: 463–465.
  16. 16. Huang TR, Feng L, Shi JJ, Liang RX, Wei ZL, et al. (2000) A case-control study for detecing risk factors of primary liver cancer in Fusui, Guangxi. Guang Xi Yi Ke Da Xue Xue Bao 17: 758–760.
  17. 17. Zhu WC, Chen Q, Luo CL, Chu XW, Wu M (2001) Relationship study between gene polymorphism of CYP1A1, GSTM l, and genetic susceptibility of primary hepatocellular carcinoma. Zhong Liu Fang Zhi Yan Jiu 8: 572–574.
  18. 18. Sun CA, Wang LY, Chen CJ, Lu SN, You SL, et al. (2001) Genetic polymorphisms of glutathione S-transferases M1 and T1 associated with susceptibility to aflatoxin-related hepatocarcinogenesis among chronic hepatitis B carriers: a nested case–control study in Taiwan. Carcinogenesis 22: 1289–1294.
  19. 19. Tiemersma EW, Omer RE, Bunschoten A, van’t Veer P, Kok FJ, et al. (2001) Role of genetic polymorphism of glutathione-S-transferase T1 and microsomal epoxide hydrolase in aflatoxin-associated hepatocellular carcinoma. Cancer Epidemiol Biomarkers Prev 10: 785–791.
  20. 20. Liu CZ, Bian JC, Jiang F, Shen FM (2002) Genetic polymorphisms of glutathione S-transferases M1, T1, P1 on susceptibility hepatocellular carcinoma. China Public Health 18: 935–936.
  21. 21. Chen SY, Wang LY, Lunn RM, Tsai WY, Lee PH, et al. (2002) Polycyclic aromatic hydrocarbon-DNA adducts in liver tissues of hepatocellular carcinoma patients and controls. Int J Cancer 99: 14–21.
  22. 22. Munaka M, Kohshi K, Kawamoto T, Takasawa S, Nagata N, et al. (2003) Genetic polymorphisms of tobacco- and alcohol-related metabolizing enzymes and the risk of hepatocellular carcinoma. J Cancer Res Clin Oncol 129: 355–360.
  23. 23. Wu YQ, Lu PX, Wang JS, Wang JB, Wan SG, et al. (2003) Correlation between the genetic polymorphism of EPHX and susceptibility to hepatocellular carcinoma. Zhong Liu 23: 287–289.
  24. 24. Liu ZG, Wei YP, Ma Y, Deng ZL (2003) Population with GSTT1 gene deletion and the relationship to hepatocellular carcinoma from Guangxi. Guang Xi Yi Ke Da Xue Xue Bao 20: 161–163.
  25. 25. Yu MW, Yang SY, Pan IJ, Lin CL, Liu CJ, et al. (2003) Polymorphisms in XRCC1 and glutathione S-transferase genes and hepatitis B-related hepatocellular carcinoma. J Natl Cancer Inst 95: 1485–1488.
  26. 26. McGlynn KA, Hunter K, LeVoyer T, Roush J, Wise P, et al. (2003) Susceptibility to aflatoxin B1-related primary hepatocellular carcinoma in mice and humans. Cancer Res 63: 4594–4601.
  27. 27. Li SP, Wu JZ, Ding JH, Gao CM, Cao HX, et al. (2004) Impact of genetic polymorphisms of glutathione S-transferase T1, Ml on the risk of primary hepatocellular carcinoma in alcohol drinkers. Pract J Cancer 19: 229–232.
  28. 28. Deng ZL, Wei YP, Ma Y (2005) Polymorphism of glutathione S-transferase mu 1 and theta 1 genes and hepatocellular carcinoma in southern Guangxi, China. World J Gastroenterol 11: 272–274.
  29. 29. Covolo L, Gelatti U, Talamini R, Garte S, Trevisi P, et al. (2005) Alcohol dehydrogenase 3, glutathione S-transferase M1 and T1 polymorphisms, alcohol consumption and hepatocellular carcinoma (Italy). Cancer Causes Control 16: 831–838.
  30. 30. Zhu MH, Chen XH, Zhou LF (2005) Association of genetic polymorphisms in glutathione S-transferases M1 with hepatitis beta-related hepatocellular carcinoma. Zhejiang Da Xue Xue Bao Yi Xue Ban 34: 126–130.
  31. 31. Ma DL, Chen YX, Li Y, Zhao HT, Xie XM, et al. (2005) Glutathione-S-transferase M1 and T1 polymorphisms (deficiency) and susceptibility to liver cancer in hepatitis B surface antigen positive (HBsAg positive) population. Guangxi Yi Xue 27: 656–657.
  32. 32. Kirk GD, Turner PC, Gong Y, Lesi OA, Mendy M, et al. (2005) Hepatocellular carcinoma and polymorphisms in carcinogen-metabolizing and DNA repair enzymes in a population with aflatoxin exposure and hepatitis B virus endemicity. Cancer Epidemiol Biomarkers Prev 14: 373–379.
  33. 33. Long XD, Ma Y, Wei YP, Dong ZL (2005) Study on the detoxication gene gstM1-, gstT1-null and susceptibility to aflatoxin B1 related hepatocellular carcinoma in Guangxi. Zhonghua Liu Xing Bing Xue Za Zhi 26: 777–781.
  34. 34. Zhang YC, Deng CS, Zhu YQ (2005) Study of genetic polymorphisms of xenobiotica metabolizing enzymes in hepatitis B virus-associated hepatic diseases. J Wenzhou Med College 35: 464–467.
  35. 35. Guo HY, Bian JC, Jiang F, Wang QM, Zhang ZM, et al. (2005) The null genotype of GSTM1 and GSTT1 and the genetic susceptibility of primary liver cancer in luoyang China. Zhong Liu 25: 58–61.
  36. 36. Ladero JM, Martínez C, García-Martín E, Ropero P, Briceño O, et al. (2006) Glutathione S-transferase M1 and T1 genetic polymorphisms are not related to the risk of hepatocellular carcinoma: a study in the Spanish population. Eur J Cancer 42: 73–77.
  37. 37. Long XD, Ma Y, Wei YP, Deng ZL (2006) The polymorphisms of GSTM1, GSTT1, HYL1*2, and XRCC1, and aflatoxin B1-related hepatocellular carcinoma in Guangxi population, China. Hepatol Res 36: 48–55.
  38. 38. Borentain P, Gérolami V, Ananian P, Garcia S, Noundou A, et al. (2007) DNA-repair and carcinogen-metabolising enzymes genetic polymorphisms as an independent risk factor for hepatocellular carcinoma in Caucasian liver-transplanted patients. Eur J Cancer 43: 2479–2486.
  39. 39. Kiran M, Chawla YK, Kaur J (2008) Glutathione-S-transferase and microsomal epoxide hydrolase polymorphism and viral-related hepatocellular carcinoma risk in India. DNA Cell Biol 27: 687–694.
  40. 40. He LJ, Gu YY, Liao ZH (2008) Glutathione S-transferase M1 genetic polymorphism, alcohol consumption, cigarette smoking and hepatocellular carcinoma. Guangxi Yi Ke Da Xue Xue Bao 25: 567–568.
  41. 41. Abd El-Moneim E, Younis FA, Allam N, Gameel K, Osman M (2008) Gene deletion of glutathione S-transferase M1 and T1 and risk factors of hepatocellular carcinoma in Egyptian patients. Egypt J Immunol 15: 125–134.
  42. 42. Imaizumi T, Higaki Y, Hara M, Sakamoto T, Horita M, et al. (2009) Interaction between cytochrome P450 1A2 genetic polymorphism and cigarette smoking on the risk of hepatocellular carcinoma in a Japanese population. Carcinogenesis 30: 1729–1734.
  43. 43. Yang ZG, Xie YA, Kuang ZP, Luo XL, Zhang WM, et al. (2009) Relationship between genetic polymorphisms of glutathione-S-transferase M1, T1 genes and susceptibility to hepatocellular carcinoma in population of fusui district of guangxi zhuang autonomous region. Zhonghua Zhong Liu 16: 970–973.
  44. 44. Asim M, Khan LA, Husain SA, Husain S, Sarma MP, et al. (2010) Genetic polymorphism of glutathione S transferases M1 and T1 in Indian patients with hepatocellular carcinoma. Dis Markers 28: 369–76.
  45. 45. Xiao KY, Li LQ, Peng MH, Qin X, Peng T, et al. (2011) Gene polymorphisms of GSTM1 and GSTT1 in the clustering families of hapatocellular carninoma. Zhong Guo Ai Zheng Fang Zhi Za Zhi 3: 287–290.
  46. 46. Sarma MP, Asim M, Medhi S, Bharathi T, Kar P (2012) Hepatitis C virus related hepatocellular carcinoma: a case control study from India. J Med Virol 84: 1009–1017.
  47. 47. Clark MF, Baudouin SV (2006) A systematic review of the quality of genetic association studies in human sepsis. Intensive Care Med 32: 1706–1712.
  48. 48. Yang Q, Khoury MJ, Friedman J, Little J, Flanders WD (2005) How many genes underlie the occurrence of common complex diseases in the population? Int J Epidemiol 34: 1129–1137.
  49. 49. Zhang J, Xu F, Ouyang C (2012) Joint effect of polymorphism in the N-acetyltransferase 2 gene and smoking on hepatocellular carcinoma. Tumour Biol 33: 1059–1063.
  50. 50. Yu L, Sun L, Jiang YF, Lu BL, Sun DR, et al. (2012) Interactions between CYP1A1 polymorphisms and cigarette smoking are associated with the risk of hepatocellular carcinoma: evidence from epidemiological studies. Mol Biol Rep 39: 6641–6646.
  51. 51. Liu C, Wang H, Pan C, Shen J, Liang Y (2012) CYP2E1 PstI/RsaI polymorphism and interaction with alcohol consumption in hepatocellular carcinoma susceptibility: evidence from 1,661 cases and 2,317 controls. Tumour Biol 33: 979–984.
  52. 52. Li R, Boerwinkle E, Olshan AF, Chambless LE, Pankow JS, et al. (2000) Glutathione S-transferase genotype as a susceptibility factor in smoking-related coronary heart disease. Atherosclerosis 149: 451–462.
  53. 53. Hayes JD, Pulford DJ (1995) The glutathione S-transferase supergene family: regulation of GST and the contribution of the isoenzymes to cancer chemoprevention and drug resistance. Crit Rev Biochem Mol Biol 30: 445–600.
  54. 54. Sheehan D, Meade G, Foley VM, Dowd CA (2001) Structure, function and evolution of glutathione transferases: implications for classification of non-mammalian members of an ancient enzyme superfamily. Biochem J 360(Pt 1): 1–16.
  55. 55. White DL, Li D, Nurgalieva Z, El-Serag HB (2008) Genetic variants of glutathione S-transferase as possible risk factors for hepatocellular carcinoma: a HuGE systematic review and meta-analysis. Am J Epidemiol 167: 377–389.
  56. 56. Wang B, Huang G, Wang D, Li A, Xu Z, et al. (2010) Null genotypes of GSTM1 and GSTT1 contribute to hepatocellular carcinoma risk: evidence from an updated meta-analysis. J Hepatol 53: 508–518.
  57. 57. Yu L, Wang CY, Xi B, Sun L, Wang RQ, et al. (2011) GST polymorphisms are associated with hepatocellular carcinoma risk in Chinese population. World J Gastroenterol 17: 3248–3256.
  58. 58. Lohmueller KE, Pearce CL, Pike M, Lander ES, Hirschhorn JN (2003) Meta-analysis of genetic association studies supports a contribution of common variants to susceptibility to common disease. Nat Genet 33: 177–182.
  59. 59. Morgan TM, Coffey CS, Krumholz HM (2003) Overestimation of genetic risks owing to small sample sizes in cardiovascular studies. Clin Genet 64: 7–17.