Skip to main content
Gastrointestinal Cancer Research : GCR logoLink to Gastrointestinal Cancer Research : GCR
. 2007 Jan-Feb;1(1):12–19.

Molecular Epidemiology of Gastric Cancer: Current Status and Future Prospects

Zhibin Hu 1,2, Jaffer A Ajani 1,2, Qingyi Wei 1,2
PMCID: PMC2632516  PMID: 19262698

Abstract

Gene-environment interaction appears to contribute to the etiology of gastric cancer, as suggested by the varying geographic patterns of gastric cancer incidence. Even in areas with a high rate Helicobacter pylori (H. pylori) infection, only a small proportion of infected individuals develop gastric cancer. It is likely that genetic factors, particularly relatively common genetic variants, such as single nucleotide polymorphisms (SNPs), may modulate the effects of environmental risk factors by regulating multiple biologic pathways involved in gastric carcinogenesis. Thus, common genetic variants can pose a substantial influence on the population attributable risk, even though the absolute risk associated with each of these variants may be low. Remarkable progress has been made in the field of molecular epidemiology, but it appears that an initial view on the magnitude of the effects of inherited variants was overestimated. Nevertheless, evidence suggests that genetic variants may contribute to the etiology of gastric cancer, particularly those SNPs in genes that are involved in inflammatory response, metabolism of chemical carcinogens, DNA repair, and tumor suppression. Although previous molecular epidemiologic studies of potentially functional polymorphisms in candidate genes and gastric cancer susceptibility lack consistency, they have advanced our knowledge of the role of genetic susceptibility in the etiology of gastric cancer. Future, welldesigned large population-based studies will validate current findings and provide the rationale for identifying at-risk subpopulations for primary prevention of gastric cancer.


Although the incidence of gastric cancer has steadily declined in past decades, this disease remains a significant global health problem. Worldwide, cancer of the stomach ranks fourth in frequency (after cancers of the lung, breast, and colon and rectum), with an estimated 934,000 new cases per year in 2002, and it is the second most common cause of cancer death (approximately 700,000 deaths annually).1 Although diagnostic and therapeutic advances have occurred during the past 10 years, the prognosis of late-stage gastric cancer continues to be bleak, and conventional treatments have little effect on survival.2 Accordingly, prevention remains the best strategy for controlling this life-threatening disease.

The etiology of gastric cancer involves a strong environmental component, and its global distribution is characterized by a wide geographic variation in incidence. Particularly high-risk areas include East Asia (especially China and Japan), Eastern Europe, and parts of Central and South America.1 First-line evidence of environmental risk factors is drawn from migration studies, in which populations from high-risk regions of the world were found to have a markedly diminished risk when they moved to lower-risk areas.35

Since 1991, sufficient evidence has emerged to support the hypothesis that Helicobacter pylori (H. pylori) colonization might play a major role in the development of gastric cancer, and it has been classified as a Group 1 carcinogen by the International Agency for Research on Cancer (IARC) and the World Health Organization (WHO).6 Dietary factors, such as the consumption of salted and nitrated foods, are also believed to be responsible for the high incidence and mortality of gastric cancer observed primarily in Asian countries.7,8 On the other hand, risk of gastric cancer is dramatically decreased in populations whose diet includes a high intake of fruits and vegetables,9 which may be partly attributable to the consumption of antioxidant micronutrients.10 Tobacco smoking is also considered a known risk factor for gastric cancer.11

In addition to the aforementioned modifiable environmental factors, genetic factors also appear to play an important role in the etiology of gastric cancer. Data show, for example, that in certain regions of the world where nearly 100% of the populace tests positive for H. pylori, only a small fraction of those infected develop gastric cancer.12 This observation suggests that genetic variations in susceptibility may constitute an underlying mechanism of gastric carcinogenesis.

Family-based genetic analysis revealed the presence of rare, highly penetrant mutations in several genes, such as E-cadherin, that may confer a high individual risk, but they account for only a small percentage of gastric cancer.13,14

In contrast, relatively common genetic variants, such as single nucleotide polymorphisms (SNPs), may modulate the effects of environmental risk factors by regulating multiple biologic pathways involved in gastric carcinogenesis. Thus, common genetic variants can contribute substantially to population attributable risk, even though the absolute risk associated with each of these variants may be low. Remarkable progress has been made in this field of study, but it appears that initial theories might have overestimated the magnitude of the effects of inherited variants.

This review summarizes a number of published association studies using several well-characterized variants or SNPs in genes involved in multiple biologic pathways related to the etiology of gastric cancer. Emphasis is placed on the functional relevance of each genetic variant or SNP, rather than the hypothesis-driven selection of disease-related biologic pathways. Although the significance of most SNPs is still largely unknown, some are more likely to be a-priori disease-causing entities than others. One implication is that SNPs that cause amino acid substitutions (ie, non-synonymous) or that are located at regulatory regions (ie, promoters) may influence disease outcomes by affecting the expression and functions of proteins.

Because relatively small, single studies published to date may not have been sufficiently powered to detect the effect of genetic polymorphisms in low-penetrance genes, we performed a meta-analysis by incorporating data available from numerous published studies to better address the association between SNPs and gastric cancer risk. We identified studies eligible for inclusion in this metaanalysis by conducting an electronic search of the literature (MEDLINE) to select relevant reports. Additional studies were identified by a manual search of references cited in original studies or review articles on similar topics. All analyses were performed with Statistical Analysis System software (v.9.1.3; SAS Institute, Cary, NC) and Review Manage Software (v.4.2; Oxford, England) as described elsewhere.15

MOLECULAR EPIDEMIOLOGIC STUDIES

Mucosal Protection Against H. Pylori Infection

H. pylori infection is associated with diverse clinical outcomes that range from simple asymptomatic gastritis to more serious conditions, such as peptic ulcer disease and gastric neoplasia. Key determinants of these outcomes are severity and distribution of the H. pylori-induced gastritis.16

When H. pylori challenges gastric mucosa, a vigorous inflammatory response is triggered that involves a complex network of inflammatory mediators, especially pro-inflammatory cytokines (eg, interleukin-1beta [IL-1β] and tumor necrosis factor-alpha [TNF-α]), which may help eradicate H. pylori organisms. Concomitant inhibition of acid secretion, however, may extend the area of colonization, resulting in damage-induced inflammation of the corpus mucosa, leading to an early onset of gastric atrophy and malignant transformation. 16 Therefore, individual differences in the intensity of the inflammatory response may contribute to variation in the likelihood of malignant transformation of gastric mucosa, which may be modulated by polymorphisms in genes that code for key inflammatory molecules.

IL1B and IL1RN

IL-1β, encoded by the IL1B gene, is a potent pro-inflammatory cytokine and an inhibitor of gastric acid secretion. Thus, it plays a key role in modulating the inflammatory response to H. pylori infection.17 The interleukin-1–receptor antagonist (IL-1ra), encoded by the IL1RN gene, is an anti-inflammatory cytokine that competitively binds to the IL-1β receptors, thereby modulating the pro-inflammatory effects of IL-1β.18 Inter-individual variation in IL-1β and IL-1ra protein levels appears to be determined by functional polymorphisms in transcription regulatory regions of their respective genes.1921 Case-control studies of diverse ethnic populations have been conducted to determine the roles these polymorphisms play in the development of gastric cancer.2248

As shown in Table 1, the IL1B-511T and IL1RN-S alleles, which are reportedly associated with increased levels of IL-1β production, have been found to confer an increased risk of gastric cancer. However, significant between-study heterogeneity was revealed in the meta-analysis, regardless of which genetic model was used, suggesting that confounders or cofactors may play important roles in determining gastric cancer risk. In subgroup analysis, a significantly elevated risk (assuming a dominant model; OR = 1.37, 95% CI = 1.13–1.67, P = .54 for the heterogeneity test) associated with the IL1RN-S allele was only evident among studies conducted in the United States24,33,44,45,47 but not in Europe (assuming a dominant model; OR = 1.25, 95% CI = 0.93–1.69, P = .0002 for the heterogeneity test),22,27,35,38,40,42,43 nor in Asia (assuming a dominant model; OR = 1.11, 95% CI = 0.75–1.64, P = .0002 for the heterogeneity test).23,25,26,29,31,34,37,39,41,46 Further, a significantly elevated risk (assuming a dominant model; OR = 1.78, 95% CI = 1.01–3.13, P < .0001 for the heterogeneity test) associated with the IL1RN-S allele was only evident among studies of intestinal gastric cancer31,33,35,40,42,47 but not for diffuse gastric cancer (assuming a dominant model; OR = 1.18, 95% CI = 0.87–1.62, P = .27 for the heterogeneity test).31,33,35,40,47 These results suggested that histologic types of the disease might contribute to the study heterogeneity. Therefore, different genetic backgrounds and local environmental factors between populations must be taken into account in such association studies.

Table 1.

Summary of meta-analyses of published studies on selected variants and gastric cancer risk.

Genes and variants Modelsa No. of Cases/Controls OR (95% CI) References
IL1B-31 (T→C) Homozygote comparison 1883/3220 0.95 (0.76–1.18) 22, 23, 25, 26, 29, 30,32–38, 40, 41, 43–48
Dominant model 3476/5788 0.96 (0.80–1.14)
Recessive model 3476/5788 0.98 (0.83–1.16)

IL1B-511 (C→T) Homozygote comparison 1813/2370 1.19 (0.93–1.53) 22, 24, 26–35, 37,39–42, 44, 46, 48
Dominant model 3663/4594 1.10 (0.92–1.32)
Recessive model 3663/4594 1.15 (0.94–1.40)


IL1RN (L→S)b SS vs. LL 2799/4355 1.42 (0.90–2.25) 22–27, 29, 31, 33–35, 37–47
Dominant model 3749/5905 1.23 (1.02–1.48)
Recessive model 3749/5905 1.33 (0.82–2.15)

TNF-α-308 (G→A) Homozygote comparison 2279/3721 1.27 (0.91–1.77)c 24, 27, 29, 35, 38, 42, 45, 46, 48, 55–61
Dominant model 2789/4497 1.06 (0.87–1.28)
Recessive model 2789/4497 1.23 (0.89–1.72)c
Homozygote comparison 792/930 0.92 (0.59–1.42)c,d 64–72

CYP2E1-1053 (C→T) Dominant model 1248/1762 0.78 (0.67–0.92)c
Recessive model 995/1368 0.98 (0.64–1.50)c,d
GSTM1 null genotype Null genotype vs. non-null genotype 3339/6273 1.33 (1.16–1.52) 72, 74–97
P53 R72P (G→C) Homozygote comparison 534/731 0.86 (0.66–1.11)c 121–128
Dominant model 1077/1387 0.98 (0.82–1.18)c
Recessive model 1077/1387 0.81 (0.65–1.00)c
Homozygote comparison 740/722 1.04 (0.51–2.11) 131–136
CDH1-160 (C→A) Dominant model 1287/1240 1.05 (0.81–1.37)
Recessive model 1287/1240 0.99 (0.52–1.87)

a Homozygote comparison: variant homozygotes were compared with wild-type homozygotes; dominant model: a single variant allele was assumed to have a dominant effect; ie, both heterozygotes and variant homozygotes were at risk, compared with wild-type homozygotes; recessive model: only variant homozygotes (having both variant alleles) were at risk, compared with variant heterozygotes and wild-type homozygotes. [143]

b S: ILRN*2; L: other alleles.

c The fixed model (P > .05 for heterogeneity test); otherwise, the random model was used.

d CYP2E1-1053 (C→T): recessive model/homozygote comparison [64–68, 70–71]

TNF-α

TNF-α, encoded by the TNF-α gene, is another potent pro-inflammatory cytokine and acid inhibitor highly expressed in H. pylori-induced gastritis, albeit the acid inhibitory properties are weaker than IL-1β.4951 Several polymorphisms have been reported in the TNF-α promoter, but the majority of published studies have focused on the G→A SNP at position –308, because most of the other SNPs are functionally silent.

The TNF-α-308A allele is thought to increase transcriptional activity of TNF-α 52 and was found to be associated with a higher concentration of TNF-α in patients with malignant tumors.53,54 However, evidence from our meta-analysis with 2,789 cases and 4,497 controls24,27,29,35,38,42,45,46,48,5561 is not supportive (Table 1), suggesting that these results need to be verified by additional large, well-designed studies.

Metabolism of Carcinogens

Inherited polymorphisms in metabolic enzymes contribute to variability in the metabolism of xenobiotics and carcinogens, a well-recognized mechanism underlying the initiation of multiple cancers. However, the metabolic system is rather nonspecific to permit high efficiency in dealing with a wide spectrum of substrates. A large number of metabolic enzymes can be grouped into two families. Phase I enzymes (like the cytochrome P450 superfamily, CYP) catabolize oxidative reactions that introduce electrophilic groups to the molecules and make them more reactive, usually leading to carcinogen activation. Phase II enzymes (like the glutathione S-transferases superfamily, GST) introduce a hydrophilic group into the intermediate molecules, usually resulting in detoxification of activated carcinogens.

CYP2E1

CYP2E1, the only member of the CYP2E subfamily identified so far, catalyzes various exogenous N-nitrosamines, including N-nitroso-dialkyiamines and tobaccosmoke-related nitrosamine.62 Considerable evidence supports the view that carcinogenic N-nitrosamine derivatives are important in the etiology of human cancers, including gastric cancer. However, large inter-individual variation in the activity of CYP2E1 has been observed, suggesting that genetic polymorphisms may play a role in individual capacity of metabolizing carcinogens. The-1053C→T SNP located in the 5’-flanking region of the CYP2E1 gene was reported to affect its binding of trans-acting factors and change its transcriptional regulation, resulting in different expression levels of the CYP2E1 mRNA.63 Although relatively few published studies have investigated this SNP and risk of gastric cancer, there clearly is a dominant protective effect of the variant allele without between-study heterogeneity as shown in Table 1.6472

GSTM1

GSTM1 is a main component of the GST families that facilitate the binding of glutathione (GSH), a nucleophilic tripeptide, to carcinogens, leading to detoxification of several known chemical compounds. The absence of GSTM1 expression due to an inherited, homozygous deletion of the GSTM1 gene in the general population may confer an increased cancer risk because of the resultant low ability to detoxify several xenobiotics, causing a decreased defense against cellular damage, such as oxidative stress. In vivo studies have shown that H. pylori causes oxidative damage in gastric epithelial cells,73 and the GSTM1 null genotype is very likely to be associated with compromised antioxidant capacity in situ, especially in the presence of H. pylori infection, and therefore may be considered a risk factor for gastric cancer.

Twenty-five studies have investigated the role of the GSTM1 null genotype in gastric cancer susceptibility, and the metaanalysis showed a significant overall 1.33-fold increased risk.72,7497 However, there was substantial heterogeneity among these 25 studies (P = .003). When we evaluated the source of heterogeneity by ethnicity (Chinese populations, 11 studies of 1,107 cases and 2,206 controls; other Asian populations, 7 studies of 1,306 cases and 1,999 controls; white populations, 7 studies of 926 cases and 2,068 controls), we found no between-study heterogeneity in each subgroup of ethnicity (data not shown).

The increased risk associated with the GSTM1 null genotype was significant in both Chinese (OR = 1.58, 95% CI = 1.35–1.85) and other Asian populations (OR = 1.17, 95% CI = 1.01–1.36) but not in white populations (OR = 1.03, 95% CI = 0.88–1.21). Furthermore, we used the Egger’s test to provide statistical evidence for the funnel plot symmetry on any publication bias.98 In the linear regression analysis, the intercept values were all significantly deviated from zero for both overall and subgroup tests (data not shown), suggesting some publication bias may be a source of possible bias in the observed associations between the GSTM1 null genotype and gastric cancer risk.

Deoxynucleotide Synthesis and DNA Repair

Studies showed that high consumption of vegetables and fruits was associated with a reduced risk of gastric cancer,99,100 partly due to a sufficient supplement of folate.101,102 An important function of folate is to provide methyl groups required for intracellular methylation reactions and de novo deoxynucleotide synthesis. Chronic folate/methyl deficiency in vivo and in vitro has been associated with abnormal DNA methylation, 103,104 DNA strand breaks, and chromosomal instability.105,106 Moreover, folate depletion may impair DNA excision repair in rat colonic mucosa but not mismatch repair.107 Therefore, it is conceivable that diminished activity of enzymes involved in folate metabolism and DNA strand break repair due functional polymorphisms may confer an increased risk of gastric cancer.

MTHFR

5,10-Methylenetetrahydrofolate reductase (MTHFR) is a central regulatory enzyme in folate metabolism. It catalyzes the reduction of 5,10-methylenetetrahydrofolate (methylene-THF) to 5-methyltetrahydrofolate (methyl-THF), the predominant circulatory form of folate and carbon donor for the remethylation of homocysteine to methionine. Two main non-synonymous SNPs (nsSNPs), 677C→T and 1298A→C of the MTHFR gene, have been identified. For example, the 677C→T nucleotide change at codon 222 results in an alanine-to-valine substitution that was found to induce a thermolabile variant of the MTHFR enzyme with a reduced activity.108 The roles of the MTHFR 677C→T and 1298A→C SNPs in gastric cancer susceptibility were recently summarized by Zintzaras et al.109 In that metaanalysis, theMTHFR677C→T,not the1298A→C variant, was shown to be associated with gastric cancer risk in all geneticmodels tested.109

XRCC1

Among the main DNA maintenance mechanisms operating in mammals, base excision repair (BER) is the primary guardian against damage that results from cellular metabolism, including reactive oxygen species, methylation, deamination, and hydroxylation. The x-ray repair cross complementing group 1 gene (XRCC1), one of the over twenty genes that participate in the BER pathway, encodes a scaffolding protein that functions in the repair of single-strand breaks (SSBs).110 Both biologic and biochemical evidence indicates a direct role of XRCC1 in BER, because it interacts with a complex of DNA repair proteins, including poly(ADP-ribose) polymerase (PARP), DNA ligase 3 (LIG3), and DNA polymerase-β.110,111

Several common nsSNPs in the XRCC1 gene have been reported, including Arg399Gln in exon 10 and Arg194Trp in exon 6. The Arg399Gln is located in the region of the BRCT-I interaction domain of XRCC1 with poly(ADP-ribose) polymerase, while the Arg194Trp variant occurs in the PCNA binding region. These two SNPs have been extensively investigated both in their functions and associations with cancer risk.15 For gastric cancer, however, only five studies have been reported, with conflicting results.112116 This suggests the need for more rigorously designed studies with large sample sizes.

Selected Tumor Suppressor Genes : TP53

The tumor protein 53 gene (TP53 or p53), the most frequently studied tumor suppressor gene, plays a number of roles in carcinogenesis in response to cellular stresses.117 TP53 is the most frequently mutated gene in human cancers, and some of these mutations have been correlated to specific carcinogen exposures and clinical phenotypes. Therefore, it is conceivable that functional genetic variants in the TP53 gene may be associated with the development of certain cancers. One well-known common nsSNP results in a non-conservative change of an arginine (R72) to a proline (P72) at amino acid 72 in a prolinerich region of p53, which may be important for the growth suppression and apoptotic functions of this protein.118,119

Recently, Pietsch reviewed the existing evidence of biochemical and biologic differences between the R72 and P72 isoforms of p53.120 The R72 variant, when found in a mutant form of p53, may enhance tumor development (eg, through increased inactivation of p73), but, when found in the wild-type form of p53, it may better inhibit tumor development (eg, through increased apoptotic ability), whereas the P72 variant may facilitate enhanced growth arrest.120 Several groups performed association studies on the p53 R72P SNP and gastric cancer risk.121128 As shown in Table 1, the 72PP variant homozygote was associated with a borderline decreased gastric cancer risk in a recessive model without between-study heterogeneity, suggesting a role for the p53 R72P SNP in regulating growth arrest in the initiation of gastric cancer.

E-Cadherin

The E-cadherin gene (CDH1) encodes a transmembrane cellular adhesion protein acting as a mediator of homophilic recognition signals, leading to cell-cell contact inhibition. Significant familial clustering of diffuse gastric cancer was found to be attributable to germline mutations in CDH1.129 The majority of CDH1 mutation carriers were considered susceptible to this inherited cancer syndrome dominated by diffuse gastric cancer,129 suggesting a central role for this gene as a tumor suppressor in diffuse gastric cancer.

Mutation-specific genetic testing for the CDH1 gene is now available, mainly for missense mutations and intragenic inframe deletions.129 The hypothesis that decreased expression without mutations in CDH1 in the general population may contribute to gastric cancer risk with a low penetrance has led to a wave of association studies of the CDH1 promoter variant and gastric cancer risk. A C→A SNP located at 160 bp upstream from the CDH1 transcription start site was identified, and the A allele was found to be correlated to a reduced transcriptional factor binding strength and transcriptional activity.130 However, epidemiologic studies failed to demonstrate an association between the promoter CDH1 variant and gastric cancer susceptibility (Table 1).131136

CONCLUSIONS AND PERSPECTIVES

In summary, evidence suggests that genetic variants may contribute to the etiology of gastric cancer, particularly those SNPs in genes that are involved in inflammatory response, metabolism of chemical carcinogens, DNA repair, and tumor suppression. Although previous molecular epidemiologic studies of potentially functional polymorphisms in candidate genes and gastric cancer susceptibility lack consistency, they have advanced our knowledge of the role of genetic susceptibility in the etiology of gastric cancer. For the association of low-penetrance genetic variants and gastric cancer risk, heterogeneity among published studies poses a great challenge, underscoring a need for more careful study design, study execution, and data analysis.

Heterogeneity between studies may arise from the disease itself due to different histological types and anatomic locations that may involve different etiologies and genetic predispositions.137 Lauren proposed categorizing gastric cancers as either intestinal or diffuse types in 1965.138 This method of classification is still useful, because it reflects a fundamental difference in gastric cancer subtypes with regard to their etiology and tumor biologic behaviors. 139 However, new disease classifications based on genetic markers rather than traditional morphologic features are warranted in the future to improve the designs of genetic and molecular epidemiologic studies.

Another source of the heterogeneity is the prevalence of confounders or cofactors. As high-throughput genotyping methods become available technically mature, and more affordable, genome-wide association approaches will be conducted more frequently in the years to come, which may provide the opportunity to develop a comprehensive genetic view of the disease.

The ability to perform related data mining and statistical analyses in terms of networking of the genes presents a challenge. Knowledge of the functional relevance of SNPs will be more critical when the issues of multiple testing can be dealt with using more sophisticated methods.

Currently, most published studies have failed to present sufficient information on environmental exposure in the early stages of the genome-wide studies. This is obviously contrary to the notion of “common variants and common disease,” the basis of the HapMap project and genome-wide scan strategy,140141 because the low-penetrant genetic effects of common SNPs may largely depend on interaction with a particular environmental exposure in multiple stages of gastric cancer carcinogenesis, such as H. pylori infection (eg, pro-inflammation genes and DNA repair genes) and dietary factors (eg, carcinogens, metabolic genes, and folate metabolic genes).

A critical area for further development in molecular epidemiologic studies on gastric cancer susceptibility would be the incorporation of novel technologic advances for refining the assessment of continuous, daily exposure to dietary factors. It is also important to develop phenotypic assays that can provide overall measurements of well-defined biologic pathways to assess correlations or associations between genetic variants and the phenotypes in appropriate tissues.

Another important aspect of epidemiologic studies is the ability to form improved multicenter research consortia,142 which may be more advantageous compared with individual studies, not only in terms of study sample sizes but also finance, data quality, and generalizable findings and conclusions.

Acknowledgments

This study was supported by National Institutes of Health grants R01 ES11740 and R01 CA100264.

Footnotes

Disclosures of Potential Conflicts of Interest

The authors declared no known financial interests or personal or professional relationships that could be perceived as presenting a potential conflict of interest that might have a bearing on the material in this paper.

References

  • 1.Parkin DM, Bray F, Ferlay J. Global cancer statistics, 2002. CA Cancer J Clin. 2005;55:74–108. doi: 10.3322/canjclin.55.2.74. [DOI] [PubMed] [Google Scholar]
  • 2.Correa P. Is gastric cancer preventable? Gut. 2004;53:1217–1219. doi: 10.1136/gut.2004.039834. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Haenszel W, Kurihara M. Studies of Japanese migrants. I Mortality from cancer and other diseases among Japanese in the United States. J Natl Cancer Inst. 1968;40:43–68. [PubMed] [Google Scholar]
  • 4.Buell P, Dunn JE., Jr Cancer mortality among Japanese Issei and Nisei of California. Cancer. 1965;18:656–664. doi: 10.1002/1097-0142(196505)18:5<656::aid-cncr2820180515>3.0.co;2-3. [DOI] [PubMed] [Google Scholar]
  • 5.McMichael AJ, McCall MG, Hartshorne JM, et al. Patterns of gastro-intestinal cancer in European migrants to Australia: the role of dietary change. Int J Cancer. 1980;25:431–437. doi: 10.1002/ijc.2910250402. [DOI] [PubMed] [Google Scholar]
  • 6.IARC Working Group on the Evaluation of Carcinogenic Risks to Humans Infection with Helicobacter pylori. IARC Monogr Eval Carcinog Risks Hum. 1994;61:177–240. [PMC free article] [PubMed] [Google Scholar]
  • 7.Correa P. Human gastric carcinogenesis: a multistep and multifactorial process--First American Cancer Society Award Lecture on Cancer Epidemiology and Prevention. Cancer Res. 1992;52:6735–6740. [PubMed] [Google Scholar]
  • 8.Stadtlander CT, Waterbor JW. Molecular epidemiology, pathogenesis and prevention of gastric cancer. Carcinogenesis. 1999;20:2195–2208. doi: 10.1093/carcin/20.12.2195. [DOI] [PubMed] [Google Scholar]
  • 9.Palli D. Epidemiology of gastric cancer: an evaluation of available evidence. J Gastroenterol. 2000;35(suppl 12):84–89. [PubMed] [Google Scholar]
  • 10.Correa P, Fontham ET, Bravo JC, et al. Chemoprevention of gastric dysplasia: randomized trial of antioxidant supplements and anti-helicobacter pylori therapy. J Natl Cancer Inst. 2000;92:1881–1888. doi: 10.1093/jnci/92.23.1881. [DOI] [PubMed] [Google Scholar]
  • 11.IARC Working Group on the Evaluation of Carcinogenic Risks to Humans Tobacco smoke and involuntary smoking. IARC Monogr Eval Carcinog Risks Hum. 2004;83:1–1438. [PMC free article] [PubMed] [Google Scholar]
  • 12.Holcombe C. Helicobacter pylori: the African enigma. Gut. 1992;33:429–431. doi: 10.1136/gut.33.4.429. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Guilford P, Hopkins J, Harraway J, et al. E-cadherin germline mutations in familial gastric cancer. Nature. 1998;392:402–405. doi: 10.1038/32918. [DOI] [PubMed] [Google Scholar]
  • 14.Huntsman DG, Carneiro F, Lewis FR, et al. Early gastric cancer in young, asymptomatic carriers of germ-line E-cadherin mutations. N Engl J Med. 2001;344:1904–1909. doi: 10.1056/NEJM200106213442504. [DOI] [PubMed] [Google Scholar]
  • 15.Hu Z, Ma H, Chen F, et al. XRCC1 polymorphisms and cancer risk: a meta-analysis of 38 case-control studies. Cancer Epidemiol Biomarkers Prev. 2005;14:1810–1818. doi: 10.1158/1055-9965.EPI-04-0793. [DOI] [PubMed] [Google Scholar]
  • 16.El-Omar EM. The importance of interleukin 1‚ in Helicobacter pylori associated disease. Gut. 2001;48:743–747. doi: 10.1136/gut.48.6.743. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Dinarello CA. Biologic basis for interleukin-1 in disease. Blood. 1996;87:2095–2147. [PubMed] [Google Scholar]
  • 18.Arend WP, Malyak M, Guthridge CJ, et al. Interleukin-1 receptor antagonist: role in biology. Annu Rev Immunol. 1998;16:27–55. doi: 10.1146/annurev.immunol.16.1.27. [DOI] [PubMed] [Google Scholar]
  • 19.Hwang IR, Kodama T, Kikuchi S, et al. Effect of interleukin 1 polymorphisms on gastric mucosal interleukin 1beta production in Helicobacter pylori infection. Gastroenterology. 2002;123:1793–1803. doi: 10.1053/gast.2002.37043. [DOI] [PubMed] [Google Scholar]
  • 20.Danis VA, Millington M, Hyland VJ, et al. Cytokine production by normal human monocytes: inter-subject variation and relationship to an IL-1 receptor antagonist (IL-1Ra) gene polymorphism. Clin Exp Immunol. 1995;99:303–310. doi: 10.1111/j.1365-2249.1995.tb05549.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Santtila S, Savinainen K, Hurme M. Presence of the IL-1RA allele 2 (IL1RN*2) is associated with enhanced IL-1beta production in vitro. Scand J Immunol. 1998;47:195–198. doi: 10.1046/j.1365-3083.1998.00300.x. [DOI] [PubMed] [Google Scholar]
  • 22.El-Omar EM, Carrington M, Chow WH, et al. Interleukin-1 polymorphisms associated with increased risk of gastric cancer. Nature. 2000;404:398–402. doi: 10.1038/35006081. [DOI] [PubMed] [Google Scholar]
  • 23.He X, Jiang L, Fu B, et al. Relationship between interleukin-1B and interleukin-1 receptor antagonist gene polymorphisms and susceptibility to gastric cancer. Zhonghua Yi Xue Za Zhi. 2002;82:685–688. [PubMed] [Google Scholar]
  • 24.El-Omar EM, Rabkin CS, Gammon MD, et al. Increased risk of noncardia gastric cancer associated with proinflammatory cytokine gene polymorphisms. Gastroenterology. 1993;124:1193–1201. doi: 10.1016/s0016-5085(03)00157-4. [DOI] [PubMed] [Google Scholar]
  • 25.Lee SG, Kim B, Choi W, et al. Lack of association between pro-inflammatory genotypes of the interleukin-1 (IL-1B -31 C/+ and IL-1RN *2/*2) and gastric cancer/duodenal ulcer in Korean population. Cytokine. 2003;21:167–171. doi: 10.1016/s1043-4666(03)00032-2. [DOI] [PubMed] [Google Scholar]
  • 26.Zeng ZR, Hu PJ, Hu S, et al. Association of interleukin 1B gene polymorphism and gastric cancers in high and low prevalence regions in China. Gut. 2003;52:1684–1689. doi: 10.1136/gut.52.12.1684. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Machado JC, Figueiredo C, Canedo P, et al. A proinflammatory genetic profile increases the risk for chronic atrophic gastritis and gastric carcinoma. Gastroenterology. 2003;125:364–371. doi: 10.1016/s0016-5085(03)00899-0. [DOI] [PubMed] [Google Scholar]
  • 28.zur Hausen A, Crusius JB, Murillo LS, et al. IL-1B promoter polymorphism and Epstein-Barr virus in Dutch patients with gastric carcinoma. Int J Cancer. 2003;107:866–867. doi: 10.1002/ijc.11468. [DOI] [PubMed] [Google Scholar]
  • 29.Wu MS, Wu CY, Chen CJ, et al. Interleukin-10 genotypes associate with the risk of gastric carcinoma in Taiwanese Chinese. Int J Cancer. 2003;104:617–623. doi: 10.1002/ijc.10987. [DOI] [PubMed] [Google Scholar]
  • 30.Lee KA, Ki CS, Kim HJ, et al. Novel interleukin 1beta polymorphism increased the risk of gastric cancer in a Korean population. J Gastroenterol. 2004;39:429–433. doi: 10.1007/s00535-003-1315-4. [DOI] [PubMed] [Google Scholar]
  • 31.Chen A, Li CN, Hsu PI, et al. Risks of interleukin-1 genetic polymorphisms and Helicobacter pylori infection in the development of gastric cancer. Aliment Pharmacol Ther. 2004;20:203–211. doi: 10.1111/j.1365-2036.2004.01826.x. [DOI] [PubMed] [Google Scholar]
  • 32.Yang J, Hu Z, Xu Y, et al. Interleukin-1B gene promoter variants are associated with an increased risk of gastric cancer in a Chinese population. Cancer Lett. 2004;215:191–198. doi: 10.1016/j.canlet.2004.07.012. [DOI] [PubMed] [Google Scholar]
  • 33.Gatti LL, Burbano RR, de Assumpcao PP, et al. Interleukin-1beta polymorphisms, Helicobacter pylori infection in individuals from Northern Brazil with gastric adenocarcinoma. Clin Exp Med. 2004;4:93–98. doi: 10.1007/s10238-004-0043-2. [DOI] [PubMed] [Google Scholar]
  • 34.Hu S, Song QB, Yu D, et al. Association of interleukin-1 gene polymorphism with gastric cancer in a high-risk area of China. Di Yi Jun Yi Da Xue Xue Bao. 2004;24:1171–1173. [PubMed] [Google Scholar]
  • 35.Glas J, Torok HP, Schneider A, et al. Allele 2 of the interleukin-1 receptor antagonist gene is associated with early gastric cancer. J Clin Oncol. 2004;22:4746–4752. doi: 10.1200/JCO.2004.03.034. [DOI] [PubMed] [Google Scholar]
  • 36.Garza-Gonzalez E, Bosques-Padilla FJ, El-Omar E, et al. Role of the polymorphic IL-1B, IL-1RN and TNF-A genes in distal gastric cancer in Mexico. Int J Cancer. 2005;114:237–241. doi: 10.1002/ijc.20718. [DOI] [PubMed] [Google Scholar]
  • 37.Chang YW, Jang JY, Kim NH, et al. Interleukin-1B (IL-1B) polymorphisms and gastric mucosal levels of IL-1beta cytokine in Korean patients with gastric cancer. Int J Cancer. 2005;114:465–471. doi: 10.1002/ijc.20724. [DOI] [PubMed] [Google Scholar]
  • 38.Zambon CF, Basso D, Navaglia F, et al. Proand anti-inflammatory cytokines gene polymorphisms and Helicobacter pylori infection: interactions influence outcome. Cytokine. 2005;29:141–152. doi: 10.1016/j.cyto.2004.10.013. [DOI] [PubMed] [Google Scholar]
  • 39.Sakuma K, Uozaki H, Chong JM, et al. Cancer risk to the gastric corpus in Japanese, its correlation with interleukin-1beta gene polymorphism (+3953*T) and Epstein-Barr virus infection. Int J Cancer. 2005;115:93–97. doi: 10.1002/ijc.20903. [DOI] [PubMed] [Google Scholar]
  • 40.Ruzzo A, Graziano F, Pizzagalli F, et al. Interleukin 1B gene (IL-1B) and interleukin 1 receptor antagonist gene (IL-1RN) polymorphisms in Helicobacter pylori-negative gastric cancer of intestinal and diffuse histotype. Ann Oncol. 2005;16:887–892. doi: 10.1093/annonc/mdi184. [DOI] [PubMed] [Google Scholar]
  • 41.Zhang WH, Wang XL, Zhou J, et al. Association of interleukin-1B (IL-1B) gene polymorphisms with risk of gastric cancer in Chinese population. Cytokine. 2005;30:378–381. doi: 10.1016/j.cyto.2005.02.002. [DOI] [PubMed] [Google Scholar]
  • 42.Perri F, Piepoli A, Bonvicini C, et al. Cytokine gene polymorphisms in gastric cancer patients from two Italian areas at high and low cancer prevalence. Cytokine. 2005;30:293–302. doi: 10.1016/j.cyto.2005.01.011. [DOI] [PubMed] [Google Scholar]
  • 43.Palli D, Saieva C, Luzzi I, et al. Interleukin-1 gene polymorphisms and gastric cancer risk in a high-risk Italian population. Am J Gastroenterol. 2005;100:1941–1948. doi: 10.1111/j.1572-0241.2005.50084.x. [DOI] [PubMed] [Google Scholar]
  • 44.Alpizar-Alpizar W, Perez-Perez GI, Une C, et al. Association of interleukin-1B and interleukin-1RN polymorphisms with gastric cancer in a high-risk population of Costa Rica. Clin Exp Med. 2005;5:169–176. doi: 10.1007/s10238-005-0082-3. [DOI] [PubMed] [Google Scholar]
  • 45.Rocha GA, Guerra JB, Rocha AM, et al. IL1RN polymorphic gene and cagA-positive status independently increase the risk of noncardia gastric carcinoma. Int J Cancer. 2005;115:678–683. doi: 10.1002/ijc.20935. [DOI] [PubMed] [Google Scholar]
  • 46.Lu W, Pan K, Zhang L, et al. Genetic polymorphisms of interleukin (IL)-1B, IL-1RN, IL-8, IL-10 and tumor necrosis factor {alpha} and risk of gastric cancer in a Chinese population. Carcinogenesis. 2005;26:631–636. doi: 10.1093/carcin/bgh349. [DOI] [PubMed] [Google Scholar]
  • 47.Sicinschi LA, Lopez-Carrillo L, Camargo MC, et al. Gastric cancer risk in a Mexican population: role of Helicobacter pylori CagA positive infection and polymorphisms in interleukin-1 and - 10 genes. Int J Cancer. 2006;118:649–657. doi: 10.1002/ijc.21364. [DOI] [PubMed] [Google Scholar]
  • 48.Kamangar F, Abnet CC, Hutchinson AA, et al. Polymorphisms in inflammation-related genes and risk of gastric cancer (Finland) Cancer Causes Control. 2006;17:117–125. doi: 10.1007/s10552-005-0439-7. [DOI] [PubMed] [Google Scholar]
  • 49.Noach LA, Basma NB, Jansen J, et al. Mucosal tumor necrosis factor-a, interleukin-1b, and interleukin-8 production in patients with Helicobacter pylori infection. Scand J Gastroenterol. 1994;29:425–429. doi: 10.3109/00365529409096833. [DOI] [PubMed] [Google Scholar]
  • 50.Beales IL, Calam J. Interleukin 1 beta and tumour necrosis factor alpha inhibit acid secretion in cultured rabbit parietal cells by multiple pathways. Gut. 1998;42:227–234. doi: 10.1136/gut.42.2.227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Wolfe MM, Nompleggi DJ. Cytokine inhibition of gastric acid secretion—a little goes a long way. Gastroenterology. 1992;102:2177–2178. doi: 10.1016/0016-5085(92)90360-b. [DOI] [PubMed] [Google Scholar]
  • 52.Wilson AG, Symons JA, McDowell TL, et al. Effects of a polymorphism in the human tumor necrosis factor alpha promoter on transcriptional activation. Proc Natl Acad Sci U S A. 1997;94:3195–3199. doi: 10.1073/pnas.94.7.3195. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Forones NM, Mandowsky SV, Lourenco LG. Serum levels of interleukin-2 and tumor necrosis factor-alpha correlate to tumor progression in gastric cancer. Hepatogastroenterology. 2001;48:1199–1201. [PubMed] [Google Scholar]
  • 54.Izutani R, Katoh M, Asano S, et al. Enhanced expression of manganese superoxide dismutase mRNA and increased TNFalpha mRNA expression by gastric mucosa in gastric cancer. World J Surg. 1996;20:228–233. doi: 10.1007/s002689900035. [DOI] [PubMed] [Google Scholar]
  • 55.Jang WH, Yang YI, Yea SS, et al. The-238 tumor necrosis factor-alpha promoter polymorphism is associated with decreased susceptibility to cancers. Cancer Lett. 2001;166:41–46. doi: 10.1016/s0304-3835(01)00438-4. [DOI] [PubMed] [Google Scholar]
  • 56.Wu MS, Huang SP, Chang YT, et al. Tumor necrosis factor-alpha and interleukin-10 promoter polymorphisms in Epstein-Barr virusassociated gastric carcinoma. J Infect Dis. 2002;185:106–109. doi: 10.1086/324771. [DOI] [PubMed] [Google Scholar]
  • 57.Lee SG, Kim B, Yook JH, et al. TNF/LTA polymorphisms and risk for gastric cancer/duodenal ulcer in the Korean population. Cytokine. 2004;28:75–82. doi: 10.1016/j.cyto.2004.06.009. [DOI] [PubMed] [Google Scholar]
  • 58.Fei BY, Xia B, Deng CS, et al. Association of tumor necrosis factor genetic polymorphism with chronic atrophic gastritis and gastric adenocarcinoma in Chinese Han population. World J Gastroenterol. 2004;10:1256–1261. doi: 10.3748/wjg.v10.i9.1256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Li C, Xia B, Yang Y, et al. TNF gene polymorphisms and Helicobacter Pylori infection in gastric carcinogenesis in Chinese population. Am J Gastroenterol. 2005;100:290–294. doi: 10.1111/j.1572-0241.2005.40806.x. [DOI] [PubMed] [Google Scholar]
  • 60.Lee JY, Kim HY, Kim KH, et al. Association of polymorphism of IL-10 and TNF-A genes with gastric cancer in Korea. Cancer Lett. 2005;225:207–214. doi: 10.1016/j.canlet.2004.11.028. [DOI] [PubMed] [Google Scholar]
  • 61.Guo W, Wang N, Li Y, et al. Polymorphisms in tumor necrosis factor genes and susceptibility to esophageal squamous cell carcinoma and gastric cardiac adenocarcinoma in a population of high incidence region of North China. Chin Med J (Engl) 2005;118:1870–1878. [PubMed] [Google Scholar]
  • 62.Yang CS, Yoo JS, Ishizaki H, et al. Cytochrome P450IIE1: roles in nitrosamine metabolism and mechanisms of regulation. Drug Metab Rev. 1990;22:147–159. doi: 10.3109/03602539009041082. [DOI] [PubMed] [Google Scholar]
  • 63.Hayashi S, Watanabe J, Kawajiri K. Genetic polymorphisms in the 5’-flanking region change transcriptional regulation of the human cytochrome P450IIE1 gene. J Biochem (Tokyo) 1991;110:559–565. doi: 10.1093/oxfordjournals.jbchem.a123619. [DOI] [PubMed] [Google Scholar]
  • 64.Kato S, Onda M, Matsukura N, et al. Cytochrome P4502E1 (CYP2E1) genetic polymorphism in a case-control study of gastric cancer and liver disease. Pharmacogenetics. 1995;5:S141–144. doi: 10.1097/00008571-199512001-00016. [DOI] [PubMed] [Google Scholar]
  • 65.Nishimoto IN, Hanaoka T, Sugimura H, et al. 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] [Google Scholar]
  • 66.Gao C, Takezaki T, Wu J, et al. 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] [Google Scholar]
  • 67.Tsukino H, Kuroda Y, Qui D, et al. Effects of cytochrome P450 (CYP) 2A6 gene deletion and CYP2E1 genotypes on gastric adenocarcinoma. Int J Cancer. 2002;100:425–428. doi: 10.1002/ijc.10492. [DOI] [PubMed] [Google Scholar]
  • 68.Park GT, Lee OY, Kwon SJ, et al. Analysis of CYP2E1 polymorphism for the determination of genetic susceptibility to gastric cancer in Koreans. J Gastroenterol Hepatol. 2003;18:1257–1263. doi: 10.1046/j.1440-1746.2003.03167.x. [DOI] [PubMed] [Google Scholar]
  • 69.Suzuki S, Muroishi Y, Nakanishi I, et al. Relationship between genetic polymorphisms of drug-metabolizing enzymes (CYP1A1, CYP2E1, GSTM1, and NAT2), drinking habits, histological subtypes, and p53 gene point mutations in Japanese patients with gastric cancer. J Gastroenterol. 2004;39:220–230. doi: 10.1007/s00535-003-1281-x. [DOI] [PubMed] [Google Scholar]
  • 70.Colombo J, Rossit AR, Caetano A, et al. GSTT1, GSTM1 and CYP2E1 genetic polymorphisms in gastric cancer and chronic gastritis in a Brazilian population. World J Gastroenterol. 2004;10:1240–1245. doi: 10.3748/wjg.v10.i9.1240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Cai L, Zheng ZL, Zhang ZF. Cytochrome p450 2E1 polymorphisms and the risk of gastric cardia cancer. World J Gastroenterol. 2005;11:1867–1871. doi: 10.3748/wjg.v11.i12.1867. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Nan HM, Song YJ, Yun HY, et al. Effects of dietary intake and genetic factors on hypermethylation of the hMLH1 gene promoter in gastric cancer. World J Gastroenterol. 2005;11:3834–3841. doi: 10.3748/wjg.v11.i25.3834. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Obst B, Wagner S, Sewing KF, et al. Helicobacter pylori causes DNA damage in gastric epithelial cells. Carcinogenesis. 2000;21:1111–1115. [PubMed] [Google Scholar]
  • 74.Deakin M, Elder J, Hendrickse C, et al. Glutathione S-transferase GSTT1 genotypes and susceptibility to cancer: studies of interactions with GSTM1 in lung, oral, gastric and colorectal cancers. Carcinogenesis. 1996;17:881–884. doi: 10.1093/carcin/17.4.881. [DOI] [PubMed] [Google Scholar]
  • 75.Kato S, Onda M, Matsukura N, et al. Genetic polymorphisms of the cancer related gene and Helicobacter pylori infection in Japanese gastric cancer patients. An age and gender matched case-control study. Cancer. 1996;77(8 suppl):1654–1661. doi: 10.1002/(SICI)1097-0142(19960415)77:8<1654::AID-CNCR35>3.0.CO;2-X. [DOI] [PubMed] [Google Scholar]
  • 76.Katoh T, Nagata N, Kuroda Y, et al. Glutathione S-transferase M1 (GSTM1) and T1 (GSTT1) genetic polymorphism and susceptibility to gastric and colorectal adenocarcinoma. Carcinogenesis. 1996;17:1855–1659. doi: 10.1093/carcin/17.9.1855. [DOI] [PubMed] [Google Scholar]
  • 77.Martins G, Alves M, Dias J, et al. Glutathione S transferase mu polymorphism and gastric cancer in the Portuguese population. Biomarkers. 1998;3:441–447. doi: 10.1080/135475098231084. [DOI] [PubMed] [Google Scholar]
  • 78.Oda Y, Kobayashi M, Ooi A, et al. Genotypes of glutathione S-transferase M1 and N-acetyl-transferase 2 in Japanese patients with gastric cancer. Gastric Cancer. 1999;2:158–164. doi: 10.1007/s101200050040. [DOI] [PubMed] [Google Scholar]
  • 79.Jiang YH, Ju ZY, Ren CS, et al. Study on the relationship between the glutathiones-transferase Gene deletion environmental factors and susceptibility to gastric carcinoma. Chin J Public Health. 2000;16:877–879. [Google Scholar]
  • 80.Liu Y, Xu RT, Sun GF, et al. The relationship of gstm1 gene homozygous deletion polymorphism and occurrence of gastric cancer. J Chin Med Univ. 2000;29:287–289. [Google Scholar]
  • 81.Cai L, Yu SZ, Zhang ZF. Glutathione S-transferases M1, T1 genotypes and the risk of gastric cancer: a case-control study. World J Gastroenterol. 2001;7:506–509. doi: 10.3748/wjg.v7.i4.506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Lan Q, Chow WH, Lissowska J, et al. Glutathione S-transferase genotypes and stomach cancer in a population-based case-control study in Warsaw, Poland. Pharmacogenetics. 2001;11:655–661. doi: 10.1097/00008571-200111000-00003. [DOI] [PubMed] [Google Scholar]
  • 83.Saadat I, Saadat M. Glutathione S-transferase M1 and T1 null genotypes and the risk of gastric and colorectal cancers. Cancer Lett. 2001;169:21–26. doi: 10.1016/s0304-3835(01)00550-x. [DOI] [PubMed] [Google Scholar]
  • 84.Gao CM, Takezaki T, Wu JZ, et al. 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. doi: 10.1016/s0304-3835(02)00115-5. [DOI] [PubMed] [Google Scholar]
  • 85.Wu MS, Chen CJ, Lin MT, et al. Genetic polymorphisms of cytochrome p450 2E1, glutathione S-transferase M1 and T1, and susceptibility to gastric carcinoma in Taiwan. Int J Colorectal Dis. 2002;17:338–343. doi: 10.1007/s00384-001-0383-2. [DOI] [PubMed] [Google Scholar]
  • 86.Sgambato A, Campisi B, Zupa A, et al. Glutathione S-transferase (GST) polymorphisms as risk factors for cancer in a highly homogeneous population from southern Italy. Anticancer Res. 2002;22:3647–3652. [PubMed] [Google Scholar]
  • 87.Zheng TR, Zheng QH, Gong FS, et al. Gene deletion polymorphisms of GSTT1 and GSTM1 and susceptibility to stomach neoplasm. Shi Yong Zhong Liu Xue Za Zhi. 2002;17:155–157. [Google Scholar]
  • 88.Gong L, Sun HL, Xu YQ. The study of correlations between the deletion of gstm1 gene and gastric cancer. Wan Nan Yi Xue Yuan Xue Bao. 2002;21:181–183. [Google Scholar]
  • 89.Choi SC, Yun KJ, Kim TH, et al. Prognostic potential of glutathione S-transferase M1 and T1 null genotypes for gastric cancer progression. Cancer Lett. 2003;195:169–175. doi: 10.1016/s0304-3835(03)00158-7. [DOI] [PubMed] [Google Scholar]
  • 90.Zhang YC, Deng CS, Zhou Y, et al. Association of glutathione S-transferase M1 and T1 genetic polymorphisms with Helicobacter pylori infection and gastric adenocarcinoma. Shi Jie Hua Ren Xiao Hua Za Zhi. 2003;11:1306–1309. [Google Scholar]
  • 91.Shen XB, Zhang J, Zhu LJ, et al. Relationship between glutathione S-transferase M1, T1 genetic polymorphisms, smoking and alcohol consumption and susceptibility to stomach cancer. J Environ Health. 2004;21:210–214. [Google Scholar]
  • 92.Lai KC, Chen WC, Tsai FJ, et al. Glutathione S-transferase M1 gene null genotype and gastric cancer risk in Taiwan. Hepatogastroenterology. 2005;52:1916–1919. [PubMed] [Google Scholar]
  • 93.Shen J, Wang RT, Xu YC, et al. Interaction models of CYP1A1, GSTM1 polymorphisms and tobacco smoking in intestinal gastric cancer. World J Gastroenterol. 2005;11:6056–6060. doi: 10.3748/wjg.v11.i38.6056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Li H, Chen XL, Li HQ. Polymorphism of CYPIA1 and GSTM1 genes associated with susceptibility of gastric cancer in Shandong Province of China. World J Gastroenterol. 2005;11:5757–5762. doi: 10.3748/wjg.v11.i37.5757. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Mu LN, Lu QY, Yu SZ, et al. Green tea drinking and multigenetic index on the risk of stomach cancer in a Chinese population. Int J Cancer. 2005;116:972–983. doi: 10.1002/ijc.21137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Palli D, Saieva C, Gemma S, et al. GSTT1 and GSTM1 gene polymorphisms and gastric cancer in a high-risk Italian population. Int J Cancer. 2005;115:284–289. doi: 10.1002/ijc.20864. [DOI] [PubMed] [Google Scholar]
  • 97.Tamer L, Ates NA, Ates C, et al. Glutathione Stransferase M1, T1 and P1 genetic polymorphisms, cigarette smoking and gastric cancer risk. Cell Biochem Funct. 2005;23:267–272. doi: 10.1002/cbf.1148. [DOI] [PubMed] [Google Scholar]
  • 98.Egger M, Davey Smith G, Schneider M, et al. Bias in meta-analysis detected by a simple, graphical test. BMJ. 1997;315:629–634. doi: 10.1136/bmj.315.7109.629. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Kobayashi M, Tsubono Y, Sasazuki S, et al. JPHC Study Group: Vegetables, fruit and risk of gastric cancer in Japan: a 10-year follow-up of the JPHC Study Cohort I. Int J Cancer. 2002;102:39–44. doi: 10.1002/ijc.10659. [DOI] [PubMed] [Google Scholar]
  • 100.Terry P, Nyren O, Yuen J. Protective effect of fruits and vegetables on stomach cancer in a cohort of Swedish twins. Int J Cancer. 1998;76:35–37. doi: 10.1002/(sici)1097-0215(19980330)76:1<35::aid-ijc7>3.0.co;2-z. [DOI] [PubMed] [Google Scholar]
  • 101.Mayne ST, Risch HA, Dubrow R, et al. Nutrient intake and risk of subtypes of esophageal and gastric cancer. Cancer Epidemiol Biomarkers Prev. 2001;10:1055–1062. [PubMed] [Google Scholar]
  • 102.Zhang ZF, Kurtz RC, Yu GP, et al. Adenocarcinomas of the esophagus and gastric cardia: the role of diet. Nutr Cancer. 1997;27:298–309. doi: 10.1080/01635589709514541. [DOI] [PubMed] [Google Scholar]
  • 103.Pogribny IP, Basnakian AG, Miller BJ, et al. Breaks in genomic DNA and within the p53 gene are associated with hypomethylation in livers of folate/methyl-deficient rats. Cancer Res. 1995;55:1894–1901. [PubMed] [Google Scholar]
  • 104.Fowler BM, Giuliano AR, Piyathilake C, et al. Hypomethylation in cervical tissue: is there a correlation with folate status? Cancer Epidemiol Biomarkers Prev. 1998;7:901–906. [PubMed] [Google Scholar]
  • 105.Blount BC, Mack MM, Wehr CM, et al. Folate deficiency causes uracil misincorporation into human DNA and chromosome breakage: implications for cancer and neuronal damage. Proc Natl Acad Sci U S A. 1997;94:3290–3295. doi: 10.1073/pnas.94.7.3290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Duthie SJ. Folic acid deficiency and cancer: mechanisms of DNA instability. Br Med Bull. 1999;55:578–592. doi: 10.1258/0007142991902646. [DOI] [PubMed] [Google Scholar]
  • 107.Choi SW, Kim YI, Weitzel JN, et al. Folate depletion impairs DNA excision repair in the colon of the rat. Gut. 1998;43:93–99. doi: 10.1136/gut.43.1.93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Frosst P, Blom HJ, Milos R, et al. A candidate genetic risk factor for vascular disease: a common mutation in methylenetetrahydrofolate reductase. Nat Genet. 1995;10:111–113. doi: 10.1038/ng0595-111. [DOI] [PubMed] [Google Scholar]
  • 109.Zintzaras E. Association of methylenetetrahydrofolate reductase (MTHFR) polymorphisms with genetic susceptibility to gastric cancer: a meta-analysis. J Hum Genet. 2006;51:618–624. doi: 10.1007/s10038-006-0405-6. [DOI] [PubMed] [Google Scholar]
  • 110.Caldecott KW, Tucker JD, Stanker LH, et al. Characterization of the XRCC1-DNA ligase III complex in vitro and its absence from mutant hamster cells. Nucleic Acids Res. 1995;23:4836–4843. doi: 10.1093/nar/23.23.4836. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Dianov GL, Prasad R, Wilson SH, et al. Role of DNA polymerase beta in the excision step of long patch mammalian base excision repair. J Biol Chem. 1999;274:13741–13743. doi: 10.1074/jbc.274.20.13741. [DOI] [PubMed] [Google Scholar]
  • 112.Shen H, Xu Y, Qian Y, et al. Polymorphisms of the DNA repair gene XRCC1 and risk of gastric cancer in a Chinese population. Int J Cancer. 2000;88:601–606. doi: 10.1002/1097-0215(20001115)88:4<601::aid-ijc13>3.0.co;2-c. [DOI] [PubMed] [Google Scholar]
  • 113.Lee SG, Kim B, Choi J, et al. Genetic polymorphisms of XRCC1 and risk of gastric cancer. Cancer Lett. 2002;187:53–60. doi: 10.1016/s0304-3835(02)00381-6. [DOI] [PubMed] [Google Scholar]
  • 114.Ratnasinghe LD, Abnet C, Qiao YL, et al. Polymorphisms of XRCC1 and risk of esophageal and gastric cardia cancer. Cancer Lett. 2004;216:157–164. doi: 10.1016/j.canlet.2004.03.012. [DOI] [PubMed] [Google Scholar]
  • 115.Huang WY, Chow WH, Rothman N, et al. Selected DNA repair polymorphisms and gastric cancer in Poland. Carcinogenesis. 2005;26:1354–1359. doi: 10.1093/carcin/bgi084. [DOI] [PubMed] [Google Scholar]
  • 116.Duarte MC, Colombo J, Rossit AR, et al. Polymorphisms of DNA repair genes XRCC1 and XRCC3, interaction with environmental exposure and risk of chronic gastritis and gastric cancer. World J Gastroenterol. 2005;11:6593–6600. doi: 10.3748/wjg.v11.i42.6593. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Braithwaite AW, Royds JA, Jackson P. The p53 story: layers of complexity. Carcinogenesis. 2005;26:1161–1169. doi: 10.1093/carcin/bgi091. [DOI] [PubMed] [Google Scholar]
  • 118.Walker KK, Levine AJ. Identification of a novel p53 functional domain that is necessary for efficient growth suppression. Proc Natl Acad Sci U S A. 1996;93:15335–15340. doi: 10.1073/pnas.93.26.15335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Sakamuro D, Sabbatini P, White E, et al. The polyproline region of p53 is required to activate apoptosis but not growth arrest. Oncogene. 1997;15:887–898. doi: 10.1038/sj.onc.1201263. [DOI] [PubMed] [Google Scholar]
  • 120.Pietsch EC, Humbey O, Murphy ME. Polymorphisms in the p53 pathway. Oncogene. 2006;25:1602–1611. doi: 10.1038/sj.onc.1209367. [DOI] [PubMed] [Google Scholar]
  • 121.Hiyama T, Tanaka S, Kitadai Y, et al. p53 Codon 72 polymorphism in gastric cancer susceptibility in patients with Helicobacter pylori-associated chronic gastritis. Int J Cancer. 2002;100:304–308. doi: 10.1002/ijc.10483. [DOI] [PubMed] [Google Scholar]
  • 122.Zhang ZW, Newcomb P, Hollowood A, et al. Age-associated increase of codon 72 Arginine p53 frequency in gastric cardia and non-cardia adenocarcinoma. Clin Cancer Res. 2003;9:2151–2156. [PubMed] [Google Scholar]
  • 123.Shen H, Solari A, Wang X, et al. P53 codon 72 polymorphism and risk of gastric cancer in a Chinese population. Oncol Rep. 2004;11:1115–1120. [PubMed] [Google Scholar]
  • 124.Wu MT, Chen MC, Wu DC. Influences of lifestyle habits and p53 codon 72 and p21 codon 31 polymorphisms on gastric cancer risk in Taiwan. Cancer Lett. 2004;205:61–68. doi: 10.1016/j.canlet.2003.11.026. [DOI] [PubMed] [Google Scholar]
  • 125.Perez-Perez GI, Bosques-Padilla FJ, Crosatti ML, et al. Role of p53 codon 72 polymorphism in the risk of development of distal gastric cancer. Scand J Gastroenterol. 2005;40:56–60. doi: 10.1080/00365520410009456. [DOI] [PubMed] [Google Scholar]
  • 126.Lai KC, Chen WC, Jeng LB, et al. Association of genetic polymorphisms of MK, IL-4, p16, p21, p53 genes and human gastric cancer in Taiwan. Eur J Surg Oncol. 2005;31:1135–1140. doi: 10.1016/j.ejso.2005.07.005. [DOI] [PubMed] [Google Scholar]
  • 127.Chung WC, Lee KM, Lee BI, et al. P53 genetic polymorphism of gastric cancer in Korea. Korean J Intern Med. 2006;21:28–32. doi: 10.3904/kjim.2006.21.1.28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Sul J, Yu GP, Lu QY, et al. P53 Codon 72 polymorphisms: a case-control study of gastric cancer and potential interactions. Cancer Lett. 2005;238:210–223. doi: 10.1016/j.canlet.2005.07.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Guilford PJ, Hopkins JB, Grady WM, et al. Ecadherin germline mutations define an inherited cancer syndrome dominated by diffuse gastric cancer. Hum Mutat. 1999;14:249–255. doi: 10.1002/(SICI)1098-1004(1999)14:3<249::AID-HUMU8>3.0.CO;2-9. [DOI] [PubMed] [Google Scholar]
  • 130.Li LC, Chui RM, Sasaki M, et al. A single nucleotide polymorphism in the E-cadherin gene promoter alters transcriptional activities. Cancer Res. 2006;60:873–876. [PubMed] [Google Scholar]
  • 131.Pharoah PD, Oliveira C, Machado JC, et al. CDH1 c-160a promotor polymorphism is not associated with risk of stomach cancer. Int J Cancer. 2002;101:196–197. doi: 10.1002/ijc.10590. [DOI] [PubMed] [Google Scholar]
  • 132.Wu MS, Huang SP, Chang YT, et al. Association of the -160 C → a promoter polymorphism of E-cadherin gene with gastric carcinoma risk. Cancer. 2002;94:1443–1448. doi: 10.1002/cncr.10371. [DOI] [PubMed] [Google Scholar]
  • 133.Humar B, Graziano F, Cascinu S, et al. Association of CDH1 haplotypes with susceptibility to sporadic diffuse gastric cancer. Oncogene. 2002;21:8192–8195. doi: 10.1038/sj.onc.1205921. [DOI] [PubMed] [Google Scholar]
  • 134.Park WS, Cho YG, Park JY, et al. A single nucleotide polymorphism in the E-cadherin gene promoter-160 is not associated with risk of Korean gastric cancer. J Korean Med Sci. 2003;18:501–504. doi: 10.3346/jkms.2003.18.4.501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Lu Y, Xu YC, Shen J, et al. E-cadherin gene C-160A promoter polymorphism and risk of noncardia gastric cancer in a Chinese population. World J Gastroenterol. 2005;11:56–60. doi: 10.3748/wjg.v11.i1.56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Song CG, Huang CM, Liu X, et al. Association of-160(C→A) polymorphism in CDH1 gene with gastric cancer risk in Fujian Chinese population. Zhonghua Yi Xue Yi Chuan Xue Za Zhi. 2005;22:557–559. [PubMed] [Google Scholar]
  • 137.Gonzalez CA, Sala N, Capella G. Genetic susceptibility and gastric cancer risk. Int J Cancer. 2002;100:249–260. doi: 10.1002/ijc.10466. [DOI] [PubMed] [Google Scholar]
  • 138.Lauren P. The two histological main types of gastric carcinoma: diffuse and so-called intestinal-type carcinoma. An attempt at a histoclinical classification. Acta Pathol Microbiol Scand. 1965;64:31–49. doi: 10.1111/apm.1965.64.1.31. [DOI] [PubMed] [Google Scholar]
  • 139.Hermanek P, Wittekind C. News of TNM and its use for classification of gastric cancer. World J Surg. 1995;19:491–495. doi: 10.1007/BF00294707. [DOI] [PubMed] [Google Scholar]
  • 140.Collins F, Galas D. A new five-year plan for the U.S. Human Genome Project. Science. 1993;262:43–46. doi: 10.1126/science.8211127. [DOI] [PubMed] [Google Scholar]
  • 141.The International HapMap Consortium The International HapMap Project. Nature. 2003;426:789–796. doi: 10.1038/nature02168. [DOI] [PubMed] [Google Scholar]
  • 142.Greene SM, Hart G, Wagner EH. Measuring and improving performance in multicenter research consortia. Natl Cancer Inst Monogr. 2005;35:26–32. doi: 10.1093/jncimonographs/lgi034. [DOI] [PubMed] [Google Scholar]
  • 143.Hao K, Xu X, Laird N, et al. Power estimation of multiple SNP association test of case-control study and application. Genet Epidemiol. 2004;26:22–30. doi: 10.1002/gepi.10293. [DOI] [PubMed] [Google Scholar]

Articles from Gastrointestinal Cancer Research : GCR are provided here courtesy of International Society of Gastrointestinal Oncology

RESOURCES