PSCA、MUC1、SMAD7、TNF-α及SEPS1基因单核苷酸多态性与湖南人群胃癌患病风险的相关性研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
背景:胃癌是世界第四大常见恶性肿瘤,居肿瘤相关死因的第二位。流行病学研究发现了胃癌的许多危险因素,如:幽门螺杆菌感染,纤维素摄入量少,吸烟等,但仅有少部分暴露于这些危险因素的人患病的事实表明胃癌的遗传易感性在胃癌发病中起至关重要的作用。研究报道前列腺干细胞抗原(prostate stem cell antigen, PSCA)、粘蛋白(mucin1, MUC1)、肿瘤坏死因子-α(tumor necrosis factor-α, TNF-α)、Selenoprotein S(SEPS1)基因多态性与胃癌的发生相关,mothers against decapentaplegic homolog7(SMAD7)基因多态性与西方人群结肠癌的发生相关。
     目的:(1)分析300例湖南地区胃癌病例的临床及其病理类型资料,了解目前本地区胃癌发病的特征;(2)研究PSCA rs2294008、MUC1rs2070803、SMAD7rs12953717、TNF-αrs361525、SEPS1rs28665122单核苷酸多态性与湖南地区人群胃癌易感性的关系。
     方法:收集300例2011年6月-2011年11月中南大学湘雅医院、中南大学湘雅二医院、湖南省肿瘤医院病理确诊的胃癌病例和300例相同地区同期健康对照的血液标本,分析胃癌病例的临床及其病理类型资料。采用基质辅助激光解吸附电离-飞行时间质谱技术(matrix-assisted laser desorption/ionization time-of-flight mass spectrometry, MALDI-TOF-MS)对300例胃癌病例及300例健康对照者的血液标本进行基因分型,胃癌组与对照组之间年龄、性别及临床病理学特征的差异采用X2检验进行比较确定有无显著性意义。采用X2检验分析5个SNP位点基因型分布在胃癌组和对照组是否符合Hardy-Weinberg遗传平衡。采用非条件Logistic回归分析PSCA rs2294008、MUC1rs2070803、SMAD7rs12953717、TNF-ars361525、 SEPS1rs28665122胃癌组和对照组间等位基因和基因型频率差别及各基因型频率与胃癌的相关性,计算OR值(Odds ratio OR)和95%可信区间(95%Confidence interval,95%CI), OR值经性别和年龄校正,统计学分析通过SPSS13.0统计软件完成(SPSS, Chicago, IL), P<0.05为差异有统计学意义。
     结果:(1)本组胃癌的临床病理学特点为:在300例胃癌中,贲门癌79例,占全部胃癌的26.3%,非贲门癌221例,占73.7%。性别构成比:胃癌的男/女性别构成比为1.8/1,责门癌男/女构成比显著高于非贲门癌(2.7/1vs1.5/1,P=0.04)。年龄:平均年龄为52.2±11.8岁,贲门癌患者的平均年龄(51.0±12.2岁)与非贲门癌患者相近(52.5±11.4岁)。贲门癌组65岁以上患者所占的比例显著高于非贲门癌组(24.1%vs12.7%,P=0.020)。Lauren病理分型:本组胃癌以肠型为主,占70.3%,贲门癌患者肠型胃癌比例显著高于非责门胃癌(82.4%vs70.1%,P=0.035)。分化程度:本组胃癌病例以低分化为主,占69.6%,其中贲门癌低分化类型所占的比例显著低于非贲门胃癌(56.0%vs74.5%,P<0.001)。TNM分期:本组胃癌以中晚期为主,III、IV期患者占69.1%;贲门癌及非贲门癌两组患者TNM分期无统计学差别。与国内外研究结果相比较:本研究贲门癌占胃癌的比例与陕西、沈阳及葡萄牙、英国的研究结果接近(21.5%-29.5%),但高于韩国及日本(低于10%);本研究在性别构成比及发病年龄等方面与这些地区的较为一致(2.7/1VS1.66-3.7/1,平均年龄51.0-63.0岁):本研究与葡萄牙、英国的结果在Lauren病理分型上较一致,均以肠型为主。TNM分期均以中晚期(Ⅲ+Ⅳ)患者为主,显著高于沈阳、韩国和日本(62.1%vs41.3%-56.4%)。(2)PSCA rs2294008多态性与胃癌发病风险增加有关。与CC基因型相比,携带rs2294008TT、TC基因型显著增加胃癌发病风险,OR值分别为2.26(95%CI1.25-4.07,P=0.007)、1.72(95%CI1.23-2.42,P=0.002),进一步分析发现:携带TT及TC基因型者显著增加肠型和弥漫型胃癌的发病风险的作用一致。(3) MUC1rs2070803多态性与胃癌的遗传易感性有关:等位基因A对胃癌的发生有保护性作用。与GG基因型相比,携带rs2070803GA基因型显著降低胃癌发病风险,OR值为0.42(95%CI0.28-0.62,P<0.0011,进一步分析发现:携带GA基因型降低胃癌发病风险的作用在肠型和弥漫型胃癌中是一致的。(4)胃癌组和对照组SMAD7rsl2953717等位基因、各基因型的分布差异无统计学意义,各基因型分布差异在肠型胃癌组和弥漫型胃癌组无统计学意义。(5)携带TNF-a rs361525AA基因型显著增加胃癌的发病风险,OR值为2.15(95%CI:1.06-4.37,P=0.035),携带AA基因型显著降低肠型胃癌发病风险(OR=0.42,95%CI:0.20-0.89,P=0.023),而弥漫型胃癌组与对照组相比较,基因型频率无统计学意义差异。(6)携带SEPS1rs28665122GA基因型显著降低胃癌的发病风险,OR值为0.63(95%CI:0.40-0.99,P=0.044)。携带AA基因型显著增加肠型胃癌发病风险(OR=1.71,95%CI:1.02-2.84,P=0.041),而弥漫型胃癌组与对照组相比较,基因型频率无统计学意义差异。
     结论:(1)本组胃癌的临床病理学特点为:男性多于女性、发病年龄晚、病理类型以肠型胃癌为主、低分化多见、TNM分期多为中晚期;与非贲门胃癌相比,贲门癌具有以男性比例更高、更倾向于肠型及高分化的特点。(2) PSCA rs2294008和MUC1rs2070803多态性与胃癌发生有关,PSCA rs2294008TT、TC基因型是胃癌的危险因素,而MUC1rs2070803GA基因型是胃癌的保护性因素。(3) SMAD7rsl2953717多态性与胃癌发病无关。(4)携带TNF-α rs361525AA基因型显著增加胃癌的发病风险,携带AA基因型显著增加降低肠型胃癌发病风险。(5)携带SEPS1rs28665122GA基因型显著降低胃癌的发病风险,携带AA基因型显著增加肠型胃癌发病风险。
Background Gastric cancer is the fourth most common cancer and the second leading cause of cancer-related death worldwide. Epidemiological studies have identified many risk factors for gastric cancer, such as Helicobacter pylori (H. pylori) infection, low fiber intake, and tobacco smoking, however, only a fraction of individuals exposed to these factors develop gastric cancer during their lifetime, which suggests that genetic susceptibility plays an important role in gastric carcinogenesis. It has been reported that polymorphisms in prostate stem cell antigen (PSCA), mucin1gene(MUC1), Selenoprotein S(SEPS1) and tumor necrosis factor-a(TNF-a)genes were associated with the risk of gastric cancer, and polymorphism in mothers against decapentaplegic homolog7(SMAD7)gene was associated with colorectal cancer in a Western population.
     Objective (1) Analyzed the clinical and pathological data of300gastric cancers by comparing the cardia and non-cardia gastric cancers in clinicopathological profiles, in order to give a better understanding of the profiles of gastric cancer in our region.(2) Analyzed the relationship of single nucleotide polymorphisms in PSCA rs2294008, MUC1rs2070803, SMAD7rsl2953717, TNF-a rs361525, SEPS1rs28665122and susceptibility to gastric cancer with different clinic pathological profiles in a population from Hunan province.
     Methods300pathologically confirmed gastric cancer patients were enrolled at the Xiangya Hospital of Central South University, The Second-Xiangya Hospital of Central South University and Tumor Hospital of Hunan Province, between June2011and November2011.300healthy controls were selected randomly from the same geographic region as the patients with gastric cancer. First analyze the clinical and pathological data of gastric cancer, then300cases of gastric cancer and300controls were genetyped for PSCA rs2294008, MUC1rs2070803, SMAD7rsl2953717, TNF-a rs361525, SEPS1rs28665122by means of Matrix-assisted laser desorptionionization time-of-flight mass spectrometry(MALDI-TOF-MS). Chi-square test was used to compare demographic distributions and pathological profiles between cases and controls. Hardy-Weinberg equilibrium was checked for controls using the goodness-of-fit χ2test. The associations between the genotypes and risk of gastric cancer were estimated by calculating the odds ratios (ORs) with95%confidence intervals (CIs) using unconditional logistic regression analyses adjusted for age and gender. All tests were conducted at the P=0.05level of significance, using SPSS13.0software package (SPSS, Chicago, IL).
     Results (1) The main clinicopathological profiles of gastric cancer in our region:gastric cardia canceraccounted for26.3%, while non-cardia cancer accounted for73.7%of total300patients. There was a significant difference in the gender composition between cardia and non-cardia cancer, the male/female ratio was significantly higher in cardia cancer than that of non-cardia gastric cancer (2.7/1vsl.5/1, P=0.04). There was no significant difference on average age between cardia cancer patients (51.2±12.2years) and non-cardia gastric cancer group(52.5±11.4years). The proportion of patients over65years of age in cardia cancer group was significantly higher than that of non-cardia gastric cancer group (24.1%vs12.7%, P=0.020). According to Lauren classification, the proportion of intestinal-type gastric cancer in cardia cancer was significantly higher than that of non-cardia gastric cancer(82.4%vs70.1%, P=0.035). Classification according to the degree of differentiation, the proportion of poorly differentiated type in cardia cancer was significantly lower than that of non-cardia gastriccancer (56.0%vs74.5%, P<0.001). The cases are mostly advanced gastric cancer and TNM staging were not significant different between cardia and non-cardia gastric cancer(P=0.160). To compare the findings with other studies in othe countries or regions, we found that the proportion of gastric cardia cancer was consistent with the results from Shanxi, Shenyang, Portugal and England (21.5%-29.5%), but higher than results from SouthKorea and Japan, which was reported to be less than10%. Consistent results in gender composition and age of onset were observed in them(2.7/1VS1.66-3.7/1, the average age of onset:52.1-63.0y). This study was more consistent with Portugal and England with respect to the Lauren classification and TNM staging, as both were mainly intestinal type and with advanced TNM stages(Ⅲ-Ⅳ), whose proportions were significantly higher than Shenyang, SouthKorea and Japan (62.1%VS41.3%-56.4%).
     Conclusion (1) The main clinic pathological profiles of gastric cancer in our region were susceptible with men, late age of onset, intestinal type, poorly differentiated and late TNM staging; compared with non-cardia gastric cancer, cardia cancer tends to be male-dominated, more inclined to well-differentiated and intestinal type.(2) PSCA rs2294008was associated with increased risk of gastric cancer. TT genotype increased gastric cancer risk to an odd of2.26(95%CI1.25-4.07, P=0.007), TC to1.72(95%CI1.23-2.42, P=0.002) compared with CC genetype. Further stratification analysis revealed that rs2294008TT and TC genotypes had a consistently increased risk on both intestinal and diffuse-type gastric cancer.(3) MUC1rs2070803was associated with the susceptibility to gastric cancer. GA genotype was associated with decreased gastric cancer risk to an odd of0.42(95%CI0.28-0.62) compared with GG genetype. Further stratification analysis revealed that the effect of rs2070803in MUC1was consistent in both intestinal and diffuse-subtypes.(4) There were no statistical differences on SMAD7 rs12953717genotypes frequencies between gastric cancer cases and controls, and between the two pathologic subtypes of gastric cancer.(5) TNF-a rs361525AA genotype was significantly associated with increased gastric cancer risk (OR=2.15,95%CI1.06-4.37, P=0.035). AA genotype was significantly associated with decreased intestinal-type gastric cancer risk (OR=0.42,95%CI:0.20-0.89, P=0.023), however, there were no statistical differences on TNF-a rs361525genotype frequencies between intestinal gastric cancer and controls.(6) SEPS1rs28665122GA genotype was significantly associated with decreased gastric cancer risk (OR=0.63,95%CI:0.40-0.99, P=0.044). AA genotype was significantly associated with increased intestinal-type gastric cancer risk (OR=1.71,95%CI:1.02-2.84, P=0.041), however, there were no statistical differences on genotypes frequencies between intestinal gastric cancer and controls.
引文
[1]Parkin DM, Bray F, Ferlay J, et al.Global cancer statistics,2002. CA Cancer J Clin,2005; 55:74-108.
    [2]AM DC Rogaska, Rogaska Slatina. Can gastric cancer be prevented? Journal of Physiology and Pharmacology,2009; 60:(Suppl 7),71-77.
    [3]Yuasa Y. DNA methylation status is inversely correlated with green tea intake and physical activity in gastric cancer patients. Int J Cancer,2009; 124: 2677-2682.
    [4]Pelucchi C. Dietary intake of selected micronutrients and gastric cancer risk: an Italian case-control study. Ann Oncol,2009; 20:160-165.
    [5]Sipponen P, Kekki M, Haapakoski J, et al. Gastric cancer risk in chronic atrophic gastritis:statistical calculations of cross-sectional data. Int J Cancer, 1985; 35:173-177.
    [6]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.
    [7]Kubo A, Corley DA. Body mass index and adenocarcinomas of the esophagus or gastric cardia:asystematic reviewand meta-analysis. Cancer Epidemiol Biomarkers Prev,2006; 15:872-8.
    [8]McColl KE.Cancer of the gastric cardia.Best Pract Res Clin Gastroenterol, 2006; 20:687-96.
    [9]张林,王盂薇,吴本俨,尤纬缔,申明识.责门胃底部癌与胃体、胃窦部癌临床及病理学特点的比较分析.中国综合临床,2006;22:364-367.
    [10]NodaS, Soejima K, Inokuchi K. Clinicopathological analysis of the intestinal type and diffuse type of gastric carcinoma. Jpn J Surg,1980; 10:277-83.
    [11]Ribeiro MM, Sarmento JA, Sobrinho Simoes MA. Prognostic significance of Lauren and Ming classificationsandother pathologic parameters in gastric carcinoma.Cancer.1981; 47:780-4.
    [12]YooCH, Noh SH, KimYI, Min JS. Comparison of prognostic significance of nodal staging betweenold(4thedition)andnew(Sthedition)UICC TNM classification for gastric carcinoma. International Union Against Cancer.World J Surg,1999; 23:492-7:discussion 497-8.
    [13]Sobin LH, Fleming ID. TNM Classification of Mal ignant Tumors, fifth edition(1997).Union Internationale Contre le Cancer and the American Joint Committee on Cancer, Cancer 1997; 80:1803-4.
    [14]Shiraishi N, InomataM, OsawaN. Early and late recurrence after gastrectomy for gastric carcinoma. Univariate and multivariate analyses. Cancer 2000: 89:255-61.
    [15]王振宁,徐惠绵,王舒宝等.胃癌新TNM分期与其生物学行为及预后的关系.中华外科杂志,2000,38(7):493-495.
    [16]MarugameT, Dongmei Q. Comparison of time trends in stomach cancer incidence(1973-1997)in East Asia, Europe and USA, from Cancer Incidence in Five Continents VOL IV-VIII. Jpn J Clin Oncol,2007; 37:242-3.
    [17]Blot WJ, DevesaSS, Kneller RW. Rising incidence of adenocarcinoma of the esophagus and gastric cardia. JAMA,1991,0265:1287-9.
    [18]Inoue M, Tsugane S. Epidemiology of gastric cancerinJapan. Postgrad Med J, 2005; 81:419-24.
    [19]HeYT, HouJ, Chen ZF, et al.Trends in incidence of esophageal and gastric cardia cancer in high-risk areas in China. Eur J Cancer Prev,2008; 17:71-6.
    [20]Wijnhoven BP, Louwman MW, Tilanus Hw. Increased incidence of adenocarcinomas at the gastro-oesophageal junction in Dutch males since the 1990s. Eur J Gastroenterol Hepatol,2002:14:115-22.
    [21]Pera M, Cameron AJ, Trastek VF. Increasing incidence of adenocarcinoma of the esophagus and esophagogastric junction. Gastroenterology,1993:104: 510-3.
    [22]DevesaSS, Blot WJ, Fraumeni JFJr. Changing patterns in the incidence of esophageal and gastric carcinoma in the United States. Cancer,1998; 83: 2049-53.
    [23]VizcainoAP, Moreno V, Lambert R. Timetrends incidence of both majorhistologic types of esophageal carcinomas in selected countries. 1973-1995. Int J Cancer,2002:99:860-8.
    [24]Bai JG, Lv Y, Dang CX. Adenocarcinoma of the Esophagogastric Junction in China according to Siewert'S classification. Jpn J Clin Oncol,2006; 36: 364-7.
    [25]Kamangar F, Qiao YL, Blaser MJ, et al. Helicobacter pylori and oesophageal and gastric cancers in a prospective study in China. Br J Cancer,2007; 96:172-6.
    [26]Stephen B, David R,Cancer Staging Handbook, American Joint Committee on Cancer(AJCC).7th ed. Chicago:Springer 2009; page:145-171.
    [27]Siewert JR, Stein HJ. Classification of adenocarcinoma of the oesophagogastric junction.Br J Surg,1998:85:1457-9.
    [28]赵东晖,张世浦,徐惠绵.贲门癌与胃远端癌临床病理因素的对比分析.沈阳医学院学报,2002,(3):132-133.
    [29]Kim MA, Lee HS, Yang HK. Clinicopathologic and protein expression differences between cardia carcinoma and noncardia carcinoma of the stomach. Cancer,2005:103:1439-46.
    [30]Maeda H, Okabayashi T, Nishimori I, et al. Clinicopathologic features of adenocarcinoma at the gastric cardia:is it different from distal cancer of the stomache. J Am Coll Surg,2008; 206:306-10.
    [31]Pinto-De-Sousa J, David L, Seixas M. Clinicopathologic profiles and prognosis of gastric carcinomas from the cardia, fundus/body and antrum. Dig Surg,2001; 18:102-10.
    [32]Derakhshan MH, Liptrot S, Paul J. Oesophageal and gastric intestinal-type adenocarcinomas show the same male predominance duetoa 17 year delayeddevelopment in females. Gut 2009:58:16-23.
    [33]郝希山.肿瘤学[M]人民卫生出版社,2010:65.
    [34]Spechler SJ, Dixon MF, Genta R, et al.Adenocarcinoma of the esophago-gastric junction. In Hamilton SR, Aaltonen LA, eds. Pathology and GeneticsofTumours of the Digestive System. Lyon, France:IARC Press: 2000; 31-36. World Health Organization Classification of Tumours:vol 2.
    [35]陈龙奇.贲门癌、食管腺癌与Barrett食管:食管胃交界区的困惑.河北医科大学学报,2006,27(3):228-229.
    [36]Chungjw, Lee GH, Choi KS, et al. Unchanging trend of esophagogastric junctionadenocarcinomain Korea:experience at a single institution based on Siewert'S classification. Dis Esophagus,2009.
    [37]Chang SS, Lu CL, ChaoJg, et al. Unchanging trend of adenocarcinoma of the esophagus and gastric cardia in Taiwan:a 15-year experience in a single center. Dig Dis Sci,2002; 47:735-40.
    [38]Kajiyama Y, Tsurumaru M, Udagawa H, et al. Prognostic factors in adenocarcinoma of the gastriccardia:pathologicstage analysis and multivariate regression analysis. J Clin Oncol,1997; 15:2015-21.
    [39]Collins FS, Guyer MS, Charkrabarri A. Variations on a thema:caraloging human DNA sequence variation. Science,1997,278:1580.
    [40]Sachidananda JI1R, Weissman D, Schmidt SC, et al. A map of human genome sequence variation containing 1.42 million single nucleotide polymorphisms. Nature,2001; 409:928-33.
    [41]严卫丽.复杂疾病全基因组关联研究进展——遗传统计分析.遗传,2008,30:543-549。
    [42]Kraft P, Cox DG. Study designs for genome-wide association studies. Advances in genetics.2008; 60:465-504.
    [43]Tenesa A, Farrington SM, Prendergast JG, et al. Genome-wide association scan identifies a colorectal cancer susceptibility locus on 11q23 and replicates risk loci at 8q24 and 18q21. Nat Genet,2008; 40:631-637.
    [44]Abnet CC, Freedman ND, Hu N, et al. A shared susceptibility locus in PLCE1 at 10q23 for gastric adenocarcinoma and esophageal squamous cell carcinoma. Nat Genet,2010; 42:764-767.
    [45]Cui R, Kamatani Y, Takahashi A, et al. Functional variants in ADH1B and ALDH2 coupled with alcohol and smoking synergistically enhance esophageal cancer risk. Gastroenterology,2009; 137:1768-1775.
    [46]Gendler SJ. MUC1, the renaissance molecule. J Mammary Gland Biol Neoplasia,2001; 6:339-353.
    [47]Singh PK, Hollingsworth MA. Cell surface-associated mucins in signal transduction. Trends Cell Biol,2006; 16:467-476.
    [48]Baruch A, Hartmann M, Zrihan-Licht S, et al. Preferential expression of novel MUC1 tumor antigen isoforms in human epithelial tumors and their tumor-potentiating function. Int J Cancer,1997; 71:741-749.
    [49]Study Group of Millennium Genome Project for Cancer. Genetic variation in PSCA is associated with susceptibility to diffuse-type gastric cancer. Nat Genet,2008; 40:730-740.
    [50]Silva E, Teixeira A, David L, et al. Mucins as key molecules for the classification of intestinal metaplasia of the stomach. Virchows Arch,2002; 440:311-317.
    [51]Saeki, N., A. Saito, et al. A Functional Single Nucleotide Polymorphism in Mucin 1, at Chromosome 1q22, Determines Susceptibility to Diffuse-Type Gastric Cancer. Gastroenterology,2011,140:892-902.
    [52]Massague J. TGF-beta in cancer. Cell,2008; 134:215-230
    [53]Xu Y, Pasche B. TGF-beta signaling alterations and susceptibility to colorectal cancer. Hum Mol Genet,2007; 16(Spec No 1):R14-R20
    [54]Nedwin GE, Naylor SL, Sakaguchi AY, et al. Human lymphotoxin and tumor necrosis factor genes:structure, homology and chromosomal localization. Nucleic Acids Res,1985; 13:6361-73.
    [55]Weber M, Hellmann I, Stadler MB, et al. Distribution, silencing potential and evolutionary impact of promoter DNA methylation in the human genome. Nat Genet,2007; 39:457-66.
    [56]van Heel DA, Udalova IA, De Silva AP, et al. Inflammatory bowel disease is associated with a TNF polymorphism that affects an interaction between the OCT1 and NF-kB transcription factors. Hum Mol Genet,2002; 11:1281-9.
    [57]Asghar T, Yoshida S, Kennedy S, et al. The tumor necrosis factor-a promoter-1031C polymorphism is associated with decreased risk of endometriosis in a Japanese population. Hum Reprod,2004; 19:2509-14.
    [58]Negoro K, Kinouchi Y, Hiwatashi N, et al. Crohn"s disease is associated with Novel polymorphisms in the 50-flanking region of the tumor necrosis factor gene. Gastroenterology,1999; 117:1062-7.
    [59]Wilson AG, di Giovine FS, Blakemore Al, et al. Single base polymorphism in the human tumor necrosis factor alpha (TNF-alpha) gene detectable by Ncol restriction of PCR product. Hum Mol Genet,1992; 1:353.
    [60]Orchard TR, Chua CN, Ahmad T, et al. Uveitis and erythema nodosum in inflammatory bowel disease:clinical features and the role of HLA genes. Gastroenterology,2002; 123:714-8.
    [61]Machado JC, Figueiredo C, Canedo P, et al. A proinflammatory genetic profile increases the risk for chronic atrophic gastritis and gastric carcinoma. Gastroenterology,2003; 125:364-71.
    [62]Gao, Y. Elevation in Tanis expression alters glucose metabolism and insulin sensitivity in H4IIE cells. Diabetes,2003; 52,929-934.
    [63]Kryukov, G.V. Characterization of mammalian selenoproteomes. Science, 2003; 300:1439-1443.
    [64]Pahl. H.L.& Baeuerle, P.A. The ER-overload response:activation of NF-kappa B.Trends Biochem. Sci,1997; 22:63-67.
    [65]Shibata T, Arisawa T, Tahara T, et al. Selenoprotein s (sepsl) gene-105G>A promoter polymorphism influences the susceptibility to gastric cancer in the japanese population. BMC gastroenterology,2009; 9:2.
    [66]Guilford P, Hopkins J, Harraway J, et al. E-cadherin germline mutations in familial gastric cancer. Nature,998,392:402-5.
    [67]Sakamoto H, Yoshimura K, Saeki N, et al. Genetic variation in PSCA is associated with susceptibility to diffuse-type gastric cancer. Nat Genet,2008; 40:730-40.
    [68]Sato Y, Suganami H, Hamada C, et al. Designing a multistage, SNP-based, genome screen for common diseases. J Hum Genet,2004; 49:669-76.
    [69]Matsuo K, Tajima K, Suzuki T, et al. Association of prostate stem cell antigen gene polymorphisms with the risk of stomach cancer in Japanese. Int J Cancer, 2009; 125:1961-4.
    [70]Sala, N., X. Munoz. Prostate stem-cell antigen gene is associated with diffuse and intestinal gastric cancer in Caucasians:Results from the EPIC-EURGAST study. International Journal of Cancer,2011.
    [71]Wu X, Ye Y, Kiemeney LA, et al. Genetic variation in the prostate stem cell antigen gene PSCA confers susceptibility to urinary bladder cancer. Nat Genet, 2009; 41:991-5.
    [72]Schier S, Wright NA. Stem cell relationships and the origin of gastrointestinal cancer. Oncology,2005; 1 (Suppl 1):9-13.
    [73]Hartman M, Loy EY, Ku CS, et al. Molecular epidemiology and its current clinical use in cancer management. The lancet oncology,2010; 11:383-390.
    [74]Reiter RE, Gu Z, Watabe T, et al. Prostate stem cell antigen:a cell surface marker overexpressed in prostate cancer. Proceedings of the National Academy of Sciences of the United States of America,1998; 95:1735-1740.
    [75]Bahrenberg G, Brauers A, Joost HG, et al. Reduced expression of PSCA, a member of the LY-6 family of cell surface antigens, in bladder, esophagus, and stomach, tumors Biochemical and biophysical research communications, 2000; 275:783-788.
    [76]Kufe DW. Mucins in cancer:function, prognosis and therapy. Nature reviews.Cancer,2009; 9:874-885.
    [77]Shin K, Fogg VC, Margolis B. Tight junctions and cell polarity. Annu Rev Cell Dev Biol,2006; 22:207-235.
    [78]Vermeer PD. Segregation of receptor and ligand regulates activation of epithelial growth factor receptor. Nature,2003; 422:322-326.
    [79]Pokutta S, Weis WI. Structure and mechanism of cadherins and catenins in cell-cell contacts. Annu Rev Cell Dev Biol,2007; 23:237-261.
    [80]Yamamoto M, Bharti A, Li Y, et al. Interaction of the DF3/MUC1 breast carcinoma-associated antigen and P-catenin in cell adhesion. J Biol Chem, 1997; 272:12492-12494.
    [81]Klaus A, Birchmeier W. Wnt signalling and its impact on development and cancer. Nature Rev Cancer,2008; 8:387-398.
    [82]Linden SK, Florin TH, McGuckin MA. Mucin dynamics in intestinal bacterial infection. PLoS One 2008; 3:e3952.
    [83]Vinall LE. Altered expression and allelic association of the hypervariable membrane mucin MUC1 in Helicobacter pylori gastritis. Gastroenterology, 2002; 123:41-49.
    [84]Kondo S. MUC1 induced by Epstein-Barr virus latent membrane protein 1 causes dissociation of the cell-matrix interaction and cellular invasiveness via STAT signaling. J Virol,2007; 81:1554-1562.
    [85]Ahmad R. MUC1-C oncoprotein functions as a direct activator of the NF-κB p65 transcription factor. Cancer Res,2009; 69:7013-7021.
    [86]Pochampalli MR, Bitler BG, Schroeder JA. Transforming growth factora-dependent cancer progression is modulated by MUC1. Cancer Res, 2007; 67:6591-6598.
    [87]Lapointe J. Gene expression profiling identifies clinically relevant subtypes of prostate cancer. Proc Natl Acad Sci USA,2004; 101:811-816.
    [88]Wreesmann VB. Genome-wide profiling of papillary thyroid cancer identifies MUC1 as an independent prognostic marker. Cancer Res,2004; 64: 3780-3789.
    [89]Jia,Y. A comprehensive analysis of common genetic variation in MUC1, MUC5AC, MUC6 genes and risk of stomach cancer. Cancer Causes Control, 2010; 21:313-321.
    [90]Xu,Q. Risk of gastric cancer is associated with the MUC1568 A/G polymorphism. Int. J. Oncol,2009,35; 1313-1320.
    [91]Abnet, C.C. A shared susceptibility locus in PLCE1 at 10q23 for gastric adenocarcinoma and esophageal squamous cell carcinoma. Nat.Genet,2010, 42; 764-767.
    [92]Dong-Mei Wu, Hai-Xia Zhu, Qing-Hong Zhao, et al. Genetic variations in the SMAD4 gene and gastric cancer Susceptibility. World J Gastroenterol,2010; 16:5635-564.
    [93]Yan X, Liu Z, Chen Y. Regulation of tgf-beta signaling by smad7. Acta biochimica et biophysica Sinica.2009; 41:263-72.
    [94]Yin JJ, Selander K, Chirgwin JM, et al. TGF-beta signaling blockade inhibits PTHrP secretion by breast cancer cells and bone metastases development. J Clin Invest,1999; 103:197-206.
    [95]Javelaud D, Mohammad KS, McKenna CR, et al. Stable overexpression of Smad7 in human melanoma cells impairs bone metastasis. Cancer Res,2007; 67:2317-2324.
    [96]Javelaud D, Delmas V, Moller M, et al. Stable overexpression of Smad7 in human melanoma cells inhibits their tumorigenicity in vitro and in vivo. Oncogene,2005; 24:7624-7629.
    [97]Cerutti JM, Ebina KN, Matsuo SE, et al. Expression of Smad4 and Smad7 in human thyroid follicular carcinoma cell lines. J Endocrinol Invest,2003; 26: 516-521.
    [98]Dowdy SC, Mariani A, Reinholz MM, et al. Overexpression of the TGF-beta antagonist Smad7 in endometrial cancer. Gynecol Oncol,2005; 96:368-373.
    [99]Mikula M, Proell V, Fischer AN, et al. Activated hepatic stellate cells induce tumor progression of neoplastic hepatocytes in a TGF-beta dependent fashion. J Cell Physiol,2006; 209:560-567.
    [100]Azuma H, Ehata S, Miyazaki H, et al. Effect of Smad7 expression on metastasis of mouse mammary carcinoma JygMC(A) cells. J Natl Cancer Inst, 2005; 97:1734-1746.
    [101]Halder SK, Beauchamp RD and Datta PK. Smad7 induces tumorigenicity by blocking TGF-beta-induced growth inhibition and apoptosis. Exp Cell Res, 2005; 307:231-246.
    [102]Kleeff J, Ishiwata T, Maruyama H, et al. The TGF-beta signaling inhibitor Smad7 enhances tumorigenicity in pancreatic cancer. Oncogene,1999; 18:5363-5372.
    [103]Liu X, Lee J, Cooley M, et al. Smad7 but not Smad6 cooperates with oncogenic ras to cause malignant conversion in a mouse model for squamous cell carcinoma. Cancer Res,2003; 63:7760-7768.
    [104]Hong S, Lim S, Li AG, et al. Smad7 binds to the adaptors TAB 2 and TAB 3 to block recruitment of the kinase TAK1 to the adaptor TRAF2. Nat Immunol, 2007; 8:504-513.
    [105]Gibson L, Johnson N, Fraser A, et al. No breast cancer association for transforming growth factor-beta pathway colorectal cancer single nucleotide polymorphisms. Cancer Epidemiol Biomarkers Prev,2009; 18:1934-1936.
    [106]Qidwai T, Khan F. Tumour necrosis factor gene polymorphism and disease prevalence. Scandinavian journal of immunology.2011; 74:522-47.
    [107]Azmy IA, Balasubramanian SP, Wilson AG et al. Role of tumour necrosis factor gene polymorphisms(-308 and-238) in breast cancer susceptibility and severity. Breast Cancer Res,2004; 6:395-400.
    [108]Deshpande A, Nolan JP, White PS, et al. TNF-alpha promoter polymorphisms and susceptibility to human papillomavirus 16-associated cervical cancer. J Infect Dis,2005; 191:969-76.
    [109]Danforth KN, Rodriguez C, Hayes RB, et al. TNF polymorphisms and prostate cancer risk. Prostate,2008; 68:400-7.
    [110]Seidemann K, Zimmermann M, Book M, et al. Tumor necrosis factor and lymphotoxin alfa genetic polymorphisms and outcome in pediatric patients with non-Hodgkin"s lymphoma:results from Berlin-Frankfurt-Munster Trial NHL-BFM 95. J Clin Oncol,2005; 23:8414-21.
    [111]Duell EJ, Casella DP, Burk RD, et al. Inflammation, genetic polymorphisms in proinflammatory genes TNF-A,RANTES, and CCR5, and risk of pancreatic adenocarcinoma. Cancer Epidemiol Biomarkers Prev.2006; 15:726.
    [112]Shih CM, Lee YL, Chiou HL, et al. Association of TNF-alpha polymorphism with susceptibility to and severity of non-small cell lung cancer. Lung Cancer 2006;52:15-20.
    [113]Basturk B, Yavasc,aog lu I, Vuruskan H, et al. Cytokine gene polymorphisms as potential risk and protective factors in renal cell carcinoma. Cytokine,2005; 30:41-5.
    [114]Onishi T. Ohishi Y, Goto H, et al. Study on the relationship between tumour necrosis factor gene polymorphism and prognosis in the patients with renal cell carcinoma. Nihon Hinyokika Gakkai Zasshi,1998; 89:413-20.
    [115]Akkiz H, Bayram S, Bekar A, et al. G-308A TNF-alpha polymorphism is associated with an increased risk of hepatocellular carcinoma in the Turkish population:case-control study. Cancer Epidemiol,2009; 33:261-4.
    [116]Machado JC, Figueiredo C, Canedo P, et al. A proinflammatory genetic profile increases the risk for chronic atrophic gastritis and gastric carcinoma. Gastroenterology,2003; 125:364-71.
    [117]El-Omar EM, Rabkin CS, Gammon MD, et al. Increased risk of noncardia gastric cancer associated with proinflammatory cytokine gene polymorphisms. Gastroenterology,2003;124:1193-201.
    [118]Wu MS, Chen LT, Shun CT, et al. Promoter polymorphisms of tumor necrosis factor-alpha are associated with risk of gastric mucosa-associated lymphoid tissue lymphoma. Int J Cancer,2004; 110:695-700.
    [119]Ye Y, Shibata Y, Yun C, et al. A membrane protein complex mediates retro-translocation from the ER lumen into the cytosol. Nature,2004; 429: 841-847.
    [120]Gao Y, Hannan NR, Wanyonyi S, et al. Activation of the selenoprotein SEPS1 gene expression by pro-inflammatory cytokines in HepG2 cells. Cytokine, 2006:33:246-251.
    [121]Alanne M, Kristiansson K, Auro K, et al. Variation in the selenoprotein S gene locus is associated with coronary heart disease and ischemic stroke in two independent Finnish cohorts. Hum Genet,2007; 122:355-365.
    [122]Moses EK, Johnson MP, Tommerdal L, et al. Genetic association of preeclampsia to the inflammatory response gene SEPS1. Am J Obstet Gynecol,2008; 198:336.el-336.e5.
    [123]Seiderer J, Dambacher J, Kuhnlein B, et al. The role of the selenoprotein S (SELS) gene-105G>A promoter polymorphism in inflammatory bowel disease and regulation of SELS gene expression in intestinal inflammation. Tissue Antigens,2007; 70:238-246.
    [124]Hyrenbach S, Pezzini A, del Zotto E, et al. Grond-Ginsbach C:No association of the-105 promoter polymorphism of the selenoprotein S encoding gene SEPS1 with cerebrovascular disease. Eur J Neurol,2007; 14:1173-1175.
    [125]El-Omar EM, Carrington M, Chow WH, et al. The role of interleukin-1 polymorphisms in the pathogenesis of gastric cancer. Nature,2001; 412:99.
    [1]Gonzalez CA, Sala N, Capella G:Genetic susceptibility and gastric cancer risk. Int J Cancer,2002; 100:249-260.
    [2]Crew KD, Neugut AI. Epidemiology of gastric cancer. World J Gastroenterol, 2006; 21:354-362.
    [3]Forman D, Burley VJ. Gastric cancer:global pattern of the disease and an overview of environmental risk factors. Best Pract Res Clin Gastroenterol,2006; 20:633-649.
    [4]Parkin DM, Bray F, Ferlay J, et al. Global Cancer Statistics,2002. CA Cancer J Clin,2005; 55:74-108.
    [5]Eslick GD. Helicobacter pylori infection causes gastric cancer? A review of the epidemiological, meta-analytic, and experimental evidence. World J Gastroenterol,2006; 12:2991-2999.
    [6]The International HapMap Project. Nature,2003:426:789-796.
    [7]Naito Y, Yoshikawa T. Molecular and cellular mechanisms involved in Helicobacter pylori-induced inflammation and oxidative stress. Free Radic Biol Med,2002; 33:323-336.
    [8]Suerbaum S, Josenhans C, Claus H. et al. Bacterial genomics:seven years on. Trends Microbiol 2002; 10:351-353.
    [9]Stein M, Rappuoli R, Covacci A. Tyrosine phosphorylation of the Helicobacter pylori CagA antigen after cag-driven host cell translocation. Proc Natl Acad Sci USA,2000:97:1263-1268.
    [10]Parsonnet J, Friedman GD, Orentreich N, et al. Risk for gastric cancer in people with CagA positive or CagA negative Helicobacter pylori infection. Gut,1997; 40:297-301.
    [11]Ilver D, Arnqvist A, Ogren J, et al. Helicobacter pylori adhesin binding fucosylated histo-blood group antigens revealed by retagging. Science,1998; 279:373-377.
    [12]Figueiredo C, Machado JC, Pharoah P, et al. Helicobacter pylori and interleukin 1 genotyping:an opportunity to identify high-risk individuals for gastric carcinoma. J Natl Cancer Inst,2002; 94:1680-1687.
    [13]Gonzalez CA, Pera G, Agudo A, et al. Fruit and vegetable intake and the risk of stomach and oesophagus adenocarcinoma in the European Prospective Investigation into Cancer and Nutrition (EPIC-EURGAST). Int J Cancer,2006; 118:2559-2566.
    [14]Jenab M, Riboli E, Ferrari P, et al. Plasma and dietary vitamin C levels and risk of gastric cancer in the European Prospective Investigation into Cancer and Nutrition (EPIC-EURGAST). Carcinogenesis,2006,27,2250-2257.
    [15]Bjelakovic G, Nikolova D, Simonetti RG, et al. Antioxidant supplements for preventing gastrointestinal cancers. Cochrane Database of Systematic Reviews, 2004, Issue 4.
    [16]Shikata K, Kiyohara Y, Kubo M, et al. A prospective study of dietary salt intake and gastric cancer incidence in a defined Japanese population:the Hisayama study. Int J Cancer,2006; 119:196-201.
    [17]E. Tahara, Genetic pathways of two types of gastric cancer, IARC Sci. Publ. 2004; 157:327-349.
    [18]El-Omar EM, Oien K, Murray LS, et al. Increased prevalence of precancerous changes in relatives of gastric cancer patients:critical role of H pylori. Gastroenterology,2000; 118:22-30.
    [19]El-Omar EM. The importance of interleukin lbeta in Helicobacter pylori associated disease. Gut,2001; 48:743-747.
    [20]El-Omar EM, Carrington M, Chow WH, et al. Interleukin-1 polymorphisms associated with increased risk of gastric cancer. Nature,2000; 404:398-402
    [21]Persson C, Canedo P, Machado JC, et al. Polymorphisms in inflammatory response genes and their association with gastric cancer:a HuGE systematic review and meta-analyses. Am J Epidemiol,2011; 173:259-70.
    [22]El-Omar EM, Rabkin CS, Gammon MD, et al. Increased risk of noncardia gastric cancer associated with proinfl ammatory cytokine gene polymorphisms. Gastroenterology,2003; 124:1193-1201.
    [23]Zhuang W, Wu XT, Zhou Y, et al. Interleukin10-592 promoter polymorphism associated with gastric cancer among Asians:a meta-analysis of epidemiologic studies. Dig Dis Sci,2010;55:1525-32.
    [24]Won HH, Kim JW, Kim MJ, et al. Interleukin 10 polymorphisms differentially influence the risk of gastric cancer in East Asians and Caucasians. Cytokine, 2010;51:73-7.
    [25]Wu X, Zeng Z, Chen B, et al. Association between polymorphisms in interleukin-17A and interleukin-17F genes and risks of gastric cancer. Int J Cancer,2010; 127:86-92.
    [26]Saeki N, Saito A, Choi IJ, et al. A functional single nucleotide polymorphism in mucin 1, at chromosome Iq22. determines susceptibility to diffuse-type gastric cancer. Gastroenterology,2011;140:892-902.
    [27]Kwon JA, Lee SY, Ahn EK, et al. Short rare muc6 minisatellites-5 alleles influence susceptibility to gastric carcinoma by regulating gene expression. Hum Mutat,2010; 31:942-9.
    [28]Savage SA, Abnet CC, Haque K, et al. Polymorphisms in interleukin-2,-6, and-10 are not associated with gastric cardia or esophageal cancer in a high-risk Chinese population. Cancer Epidemiol Biomarkers Prev,2004; 13:1547-1549.
    [29]Mori N, Wada A, Hirayama T, et al. Activation of intercellular adhesion molecule 1 expression by Helicobacter pylori is regulated by NF-kappaB in gastric epithelial cancer cells. Infect Immun,2000; 68:1806-1814.
    [30]Maeda S, Yoshida H, Ogura K, et al. H pylori activates NF-kap paB through a signaling pathway involving IkappaB kinases, NF-kappaB-inducing kinase, TRAF2, and TRAF6 in gastric cancer cells. Gastroenterology,2000; 119: 97-108.
    [31]Isomoto H, Mizuta Y, Miyazaki M. et al. Implication of NF-kappaB in Helicobacter pylori-associated gastritis. Am J Gastroenterol,2000; 95: 2768-2776.
    [32]Hatz RA, Rieder G, Stolte M, et al. Pattern of adhesion molecule expression on vascular endothelium in Helicobacter pylori associated antral gastritis. Gastroenterology,1997; 112:1908-1919.
    [33]Sakamoto H, Yoshimura K, Saeki N, et al. Genetic variation in PSCA is associated with susceptibility to diffuse type gastric cancer. Nat Genet,2008, 40:730-740.
    [34]Guan X, Zhao H, Niu J, et al. The VEGF 634G>C promoter polymorphism is associated with risk of gastric cancer. BMC Gastroenterol,2009; 9:77.
    [35]Tahara T, Shibata T, Nakamura M, et al. Effect of polymorphisms in the 30 untranslated region (30-UTR) of vascular endothelial growth factor gene on gastric cancer and peptic ulcer diseases in Japan. Mol Carcinog,2009; 48:1030-7.
    [36]Corso G, Marrelli D, Pedrazzani C, et al. Gastric cardia carcinoma is associated with the promoter-77T>C gene polymorphism of X-ray cross-complementing group 1 (XRCC1). J Gastrointest Surg,2009;13:2233-8.
    [37]Duthie SJ:Folic acid deficiency and cancer:mechanisms of DNA instability. Br Med Bull,1999; 55:578-592.
    [38]Boccia S, Hung R, Ricciardi G, et al. Meta and Pooled Analyses of the Methylenetetrahydrofolate Reductase C677T and A1298C Polymorphisms and Gastric Cancer Risk:A Huge-GSEC Review. Am J Epidemiol,2008; 167: 505-516.
    [39]Wang LH, Kim SH, Lee YL, et al. Inactivation of SMAD4 tumor suppressor gene during gastric carcinoma progression. Clin Cancer Res,2007; 13:102-10.
    [40]Wu DM, Zhu HX, et al. Genetic variations in the SMAD4 gene and gastric cancer susceptibility.World J Gastroenterol,2010; 16:5635-6341.
    [41]Ju H, Lim B, Kim M, et al. SERPINE1 intron polymorphisms affecting gene expression are associated with diffuse-type gastric cancer susceptibility. Cancer, 2010; 116:4248-55.
    [42]Guan X, Zhao H, Niu J, et al. The VEGF 634G>C promoter polymorphism is associated with risk of gastric cancer. BMC Gastroenterol,2009; 9:77.
    [43]Tahara T, Shibata T, Nakamura M, et al. Effect of polymorphisms in the 30 untranslated region (3c-UTR) of vascular endothelial growth factor gene on gastric cancer and peptic ulcer diseases in Japan. Mol Carcinog,2009; 48: 1030-7.
    [44]Zheng HC, Xu XY, Xia P, et al. Involvement of inactive GSK3b overexpression in tumorigenesis and progression of gastric carcinomas. Hum Pathol,2010; 41:1255-64
    [45]Mishra P, Senthivinayagam S, et al. Glycoden synthase kinase-3beta regulates Snail and beta-catenin during gastrin-induced migration of gastric cancer cells. J Mol Signal,2010; 5:1-10
    [46]Zhao P, Li Y, Lu Y. Aberrant expression of CD133 protein correlates with Ki-67 expression and is a prognostic marker in gastric adenocarcinoma. BMC Cancer,2010; 10:218.
    [47]Anami K, Oue N, Nogushi T, et al. Search for transmembrane protein in gastric cancer by the Escherichia coli ampicillin secretion trap:expression of DSC2 in gastric cancer with intestinal phenotype. J Pathol,2010; 221:275-84.
    [48]Kim MA, Jung EJ, Lee HS, et al. P-cadherin expression in gastric carcinoma:ist regulation mechanism and prognostic significance. Hum Pathol,2010; 41: 877-85.
    [49]Lee HJ, Nam KT, Park HS, et al. Gene expression profiling of metastatic lineages identifies CDH17 as a prognostic marker in early stage gastric cancer. Gastroenterology,2010; 139:213-25.
    [50]da Cunha CB, Oliveira C, Wen X, et al. De novo expression of CD44 variants in sporadic and hereditary gastric cancer. Lab Invest,2010; 90:1604-14.
    [51]Ishimoto T, Nagano O, Yae T, et al. CD44 variant regulates redox status in cancer cells by stabilizing the xCT subunit of system xc and thereby promotes tumor growth. Cancer Cell,2011; 19:387-400.
    [52]Koskensalo S, Mrena J, Wiksten JP, et al. MMP-7 overexpression is an independent prognostic marker in gastric cancer. Tumor Biol,2010; 31:149-55.
    [53]Zhao ZS, Chu YQ, Ye ZY, et al. Overexpression of matrix metalloproteinase 11 in human gastric carcinoma and its clinicopathologic significance. Hum Pathol, 2010; 41:686-96.
    [54]Liu R, Zhang C, Hu Z, et al. A five-microRNA signature identified from genome-wide serum microRNA expression profiling serves as fingerprint for gastric cancer diagnosis. Eur J Cancer,2011; 47:784-91.
    [55]Li X, Zhang Y, Zhang Y, et al. Survival prediction of gastric cancer by a seven-microRNA signature. Gut,2010; 59:579-85.
    [56]Pan G, Lv Hui, Ren H, et al. Elevated expression of semaphoring 5A in human gastric cancer and its implication in carcinogenesis. Life Sci.2010; 86:139-44.
    [57]Florou D, Papadopoulos IN, Scorilas A. Molecular analysis and prognostic impact of the novel apoptotic gene BCL2L12 in gastric cancer. Biochem Biophys Res Commun,2010; 391:214-8.
    [58]Zeng J. Ge Z, Wang L, et al. The histone demethylase RBP2 is overexpressed in gastric cancer and its inhibition triggers senescence of cancer cells. Gastroenterology,2010; 138:981-92.
    [59]Ando K, Kakeji Y, Kitao H, et al. High expression of BUBR1 is one of the factors for inducing DNA aneuploidy and progression in gastric cancer. Cancer Sci,2010; 101:639-45.
    [60]H. Kuniyasu, W. Yasui, H. Yokozaki, et al. Aberrant expression of c-met mRNA in human gastric carcinomas. Int. J. Cancer,1993; 55:72-75.
    [61]M.G. Smith, G.L. Hold, E. Tahara, et al. Cellular and molecular aspects of gastric cancer. World J.Gastroenterol,2006; 12:2979-2990.
    [62]Varis. A. Zaika. P. Puolakkainen, et al. Coamplified and overexpressed genes at ERBB2 locus in gastric cancer, Int. J. Cancer,2004; 109:548-553.
    [63]R.R. Burbano, P.P. Assumpcao, M.F. Leal, et al. C-Myc locus amplification as metastasis predictor in intestinal-type gastric adenocarcinomas:CGH study in Brazil. Anticancer Res,2006; 26:2909-2914.
    [64]H.-K. Lee, H.S. Lee, H.-K. Yang, et al. Prognostic significance of bcl-2 and p53 expression in gastric cancer, Int. J. Colorectal Dis,2003; 18:518-525.
    [65]An, I.-S. Choi, J.C. Yao, et al. Prognostic significance of CpG island methylator phenotype and microsatellite instability in gastric carcinoma. Clin. Cancer Res, 2005; 11:656-663.
    [66]X.-S. He, Y.-H. Rong, Q. Su, et al.Expression of p16 gene and Rb protein in gastric carcinoma and their clinicopathological significance. World J. Gastroenterol,2005,11,2218-2223.
    [67]Hasegawa S, Furukawa Y, Li M, et al. Genome-wide analysis of gene expression in intestinal-type gastric cancers using a complementary DNA microarray representing 23,040 genes. Cancer Res,2002; 62:7012-7017.
    [68]Boussioutas A, Li H, Liu J, et al. Distinctive patterns of gene expression in premalignant gastric mucosa and gastric cancer. Cancer Res,2003; 63: 2569-2577.
    [69]Hippo Y, Taniguchi H, Tsutsumi S, et al. Global gene expression analysis of gastric cancer by oligonucleotide microarrays. Cancer Res,2002; 62:233-240.
    [70]Lee S, Baek M, Yang H, et al. Identification of genes differentially expressed between gastric cancers and normal gastric mucosa with cDNA microarrays. Cancer Lett,2002; 184:197-206.
    [71]Tomita H, Takaishi S, Menheniott TR, et al. Inhibition of gastric carcinogenesis by the hormone gastrin is mediated by suppressor of TFF1 epigenetic silencing. Gastroenterology,2011; 140:879-91.
    [72]Lai KW, Koh KX, Loh M, et al. MicroRNA-130b regulates the tumour suppressor RUNX3 in gastric cancer. Eur J Cancer,2010; 46:1456-63.
    [73]Tsang YH, Lamb A, Romero-Gallo J, et al. Helicobacter pylori CagA targets gastric tumor suppressor RUNX3 for proteasome-mediated degradation. Oncogene,2010; 29:5643-50.
    [74]Kim MA, Jung EJ. Lee HS, et al. P-cadherin expression in gastric carcinoma:ist regulation mechanism and prognostic significance. Hum Pathol,2010; 41: 877-85.
    [75]K. Hayashi, H. Yokozaki, S. Gudison, et al. Inactivation of retinoid acid receptor beta by promoter CpG hypermethylation in gastric cancer, Differentiation,2001; 68:3-21.
    [76]Thiagalingam S, Foy RL, Cheng KH, et al. Loss of heterozygosity as a predictor to map tumor suppressor genes in cancer:molecular basis of its occurrence. Curr Opin Oncol,2002; 14:65-72.
    [77]A.J. French, G. Petroni, S.N. Thibideau, et al. Allelic imbalance of 8p indicates poor survival in gastric cancer, J. Mol. Diagnost,2004; 6:243-252.
    [78]S.H. Koo, T.E. Jeong, J.U. Kang, et al. Prognostic implications for gastric carcinoma based on loss of heterozygosity genotypes correlation with clinicopathologic variables, Cancer Genet. Cytogenet,2004; 153:26-31.
    [79]S. Suzuki, K. Egami, K. Sasajima, et al. Comparative study between DNA copy number aberrations determined by quantitative microsatellite analysis and clinical outcome in patients with stomach cancer. Clin. Cancer Res,2004; 10: 3013-3019.
    [80]McKie AB, Filipe MI & Lemoine NR. Abnormalities affecting the APC and MCC tumour suppressor gene loci on chromosome 5q occur frequently in gastric cancer but not in pancreatic cancer. Int J Cancer,1993; 55:598-603.
    [81]Tamura G, Sato K, Akiyama S, et al. Molecular characterization of undifferentiated-type gastric carcinoma. Lab Invest,2001; 81:593-598.
    [82]Keller G, Rotter M, Vogelsang H, et al. Microsatellite instability in adenocarcinomas of the upper gastrointestinal tract. Relation to clinicopathological data and family history. Am J Pathol,1995; 147:593-600.670
    [83]Vauhkonen M, Vauhkonen H, Sajantila A, et al. Intestinal and diffuse type of gastric cancer represent different genetic integrity as assessed with non-cancer related tetranucleotide microsatellite markers.Gastric Cancer,2005; 8:238-244.
    [84]Buonsanti G, Calistri D, Padovan L, et al. Microsatellite instability in intestinal-and diffuse-type gastric carcinoma. J Pathol,1997; 182:167-173.
    [85]Pinto M. Oliveira C, Machado JC, et al. MSI-L gastric carcinomas share the hMLHl methylation status of MSI-H carcinomas but not their clinicopathological profile. Lab Invest,2000; 80:1915-1923.
    [86]Lee HS, Choi SI, Lee HK, et al. Distinct clinical features and outcomes of gastric cancers with microsatellite instability. Mod Pathol,2002; 15:632-640.
    [87]Mori Y. Sato F, Selaru FM, et al. Instabilotyping reveals unique mutational spectra in microsatellite unstable gastric cancers. Cancer Res,2002; 62: 3641-6345.
    [88]Falchetti M. Gastric cancer with high-level microsatellite instability:target gene mutations, clinicopathologic features, and long-term survival. Hum Pathol 2008,39(6):925-932
    [89]Kim MS. Frameshift mutations of Wnt pathway genes AXIN2 and TCF7L2 in gastric carcinomas with high microsatellite instability. Hum Pathol 2009,40(1): 58-64
    [90]Wu MS. Infrequent hMSH2 mutations in sporadic gastric adenocarcinoma with microsatellite instability. Cancer Lett,1997; 112:161-166
    [91]Pinto M. Somatic mutations in mismatch repair genes in sporadic gastric carcinomas are not a cause but a consequence of the mutator phenotype. Cancer Genet Cytogenet,2008; 180:110-114
    [92]H.S. Lee, S.I. Choi, H.K. Lee, et al. Distinct clinical features and outcomes of gastric cancers with microsatellite instability, Mod. Pathol,2002; 15:632-640.
    [93]S. Beghelli, G. de Manzoni, S. Barbi, et al. Microsatellite instability in gastric cancer is associated with better prognosis in only stage Ⅱ cancers, Surgery,2006; 139:347-356.
    [94]Herman JG & Baylin SB. Gene silencing in cancer in association with promoter hypermethylation. N Engl J Med,2003; 349:2042-2054.
    [95]Guilleret I, Yan P, Grange F, et al. Hypermethylation of the human telomerase catalytic subunit (hTERT) gene correlates with telomerase activity. Int J Cancer, 2002; 10:335-341.
    [96]Wu MS, Wang HP, Lin CC, et al. Loss of imprinting and over-expression of IGF2 gene in gastric adenocarcinoma. Cancer Lett,1997; 120:9-14.
    [97]Akiyama Y, Maesawa C, Ogasawara S, et al. Cell-type-specific repression of the Maspin gene is disrupted frequently by demethylation at the promoter region in gastric intestinal metaplasia and cancer cells. Am J Pathol,2003; 163: 1911-1919.
    [98]Oshimo Y, Nakayama H, Ito R, et al. Promoter methylation of cyclin D2 gene in gastric carcinoma. Int J Oncol,2003; 23:1663-1670.
    [99]Hamai Y, Oue N, Mitani Y, et al. DNA hypermethylation and histone hypoacetylation of the HLTF gene are associated with reduced expression in gastric carcinoma. Cancer Sci,2003; 94:692-698.
    [100]Yasui W, Oue N, Ono S, et al. Histone acetylation and gastrointestinal carcinogenesis. Ann N Y Acad Sci,2003; 983:220-231.
    [101]Hong SH, Kim HG, Chung WB, et al. DNA hypermethylation of tumor-related genes in gastric carcinoma. J Korean Med Sci,2005; 20:236-241.
    [102]Tamura G, Yin J, Wang S, et al. E-cadherin gene promoter hypermethylation in primary human gastric carcinomas. J Natl Cancer Inst,2000; 92:569-573.
    [103]Oue N, Shigeishi H, Kuniyasu H, et al. Promoter hypermethylation of MGMT is associated with protein loss in gastric carcinoma. Int J Cancer,2001; 93: 805-809.
    [104]C.M. Fenoglio-Preiser, J. Wang, G.N. Stemmermann, A.Noffsinger, TP53 and gastric carcinoma:a review. Hum. Mutat,2003; 21:258-270.
    [105]X.P. Liu, K. Tsushimi, M. Tsushimi, et al. Expression of p21(WAF1/CIP1) and p53 proteins in gastric carcinoma:its relationship with cell proliferation activity and prognosis. Cancer Lett,2001;170:183-189.
    [106]Y.-E. Joo, I.-J. Chung, Y.-K. Park, et al. Expression of cyclooxygenase-2, p53 and Ki-67 in gastric cancer. J. Korean Med. Sci,2006; 21:871-876.
    [107]Rothwell PM, Fowkes FG, Belch JF, et al. Effect of daily aspirin on long-term risk of death due to cancer:analysis of individual patient data from randomised trials. Lancet,2011; 377:31-41.
    [108]Dong J, Dai J, Zhang M, et al. Potentially functional COX-2-1195G>A polymorphism increases the risk of digestive system cancers:a meta-analysis. J Gastroenterol Hepatol,2010; 25:1042-50.
    [109]Yao L, Liu F, Hong L, et al. The function and mechanism of COX-2 in angiogenesis of gastric cancer cells. J Exp Clin Cancer Res,2011; 30:13.
    [110]Mrena J, Wiksten JP, Kokkola A, et al. COX-2 is associated with proliferation and apoptosis markers and serves as an independent prognostic factor in gastric cancer. Tumour Biol,2010; 31:1-7.
    [111]Menter DG, Schilsky RL, DuBois RN. Cyclooxygenase-2 and cancer treatment: Understanding the risk should be worth the reward. Clin Cancer Res,2010; 16: 1384-90.
    [112]Wang HJ, Ruan HJ, He XJ, et al. MicroRNA-101 is down-regulated in gastric cancer and involved in cell migration and invasion. Eur J Cancer,2010; 46: 2295-303.
    [113]Oshima H, Popivanova BK, Oguma K, et al. Activation of epidermal growth factor receptor signaling by the prostaglandin E(2) receptor EP4 pathway during gastric tumorigenesis. Cancer Sci,2011; 102:713-9.
    [114]Arrington AK, Davydova J, Vickers SM, et al. Anti-ERBB2 sh-RNA suppress both cell growth and tumour growth in ERBB2-overexpressing upper gastrointestinal adenocarcinomas. J Gastrointest Surg,2009; 13:1754-61.
    [115]Tanizaki J, Okamoto I, Takezawa K, et al. Synergistic antitumor effect of S-1 and HER2-targeting agents in gastric cancer with HER2 amplification. Mol Cancer Ther,2010; 9:1198-207.
    [116]Wainberg ZA, Anghel A, Desai AJ, et al. Lapatinib, a dual EGFR and HER2 kinase inhibitor, selectively inhibits HER2-amplified human gastric cancer cells and is synergistic with trastuzumab in vitro and in vivo. Clin Cancer Res,2010; 16:1509-19.
    [117]Hofmann M, Stoss O, Shi D, et al. Assessment of a HER2 scoring system for gastric cancer:results from a validation study. Histopathology,2008; 52: 797-805.
    [118]Van Cutsem E, Kang Y. Chung H, et al. Efficacy results from the ToGA trial:a phase Ill study of trastuzumab added to standard chemotherapy in frist-line human epidermal growth factor receptor 2 positive advanced gastric cancer. J Clin Oncol,2009; 27:(suppl; abstr LBA4509).
    [119]Bang Y, Chung H, Xu J, et al. Pathological features of advanced gastric cancer (GC):relationship to human epidermal growth factor receptor 2 (HER2) positivity in the global screening programme of the ToGA trial. J Clin Oncol, 2009; 27:(suppl; abstr 4556).
    [120]Uemura N, Mukai T, Okamoto S, et al. Effect of Helicobacter pylori eradication on subsequent development of cancer after endoscopic resection of early gastric cancer. Cancer Epidemiol Biomarkers Prev,1997; 6:639-642.
    [121]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.
    [122]Wong BC, Lam SK, Wong WM, et al. Helicobacter pylori eradication to prevent gastric cancer in a high-risk region of China:a randomized controlled trial. JAMA 2004; 291:187-194.
    [123]Yuan JM, Ross RK, Gao YT, et al. Prediagnostic levels of serum micronutrients in relation to risk of gastric cancer in Shanghai, China. Cancer Epidemiol Biomarkers Prev 2004; 13:1772-1780.
    [124]Blot WJ, Li JY, Taylor PR, et al. Nutrition intervention trials in Linxian, China: supplementation with specific vitamin/mineral combinations, cancer incidence, and disease-specific mortality in the general population. J Natl Cancer Inst 1993; 85:1483-1492.
    [125]Varis K, Taylor PR, Sipponen P, et al. Gastric cancer and premalignant lesions in atrophic gastritis:a controlled trial on the effect of supplementation with alphatocopherol and beta-carotene. The Helsinki Gastritis Study Group. Scand J Gastroenterol 1998; 33:294-300.
    [126]Jacobs EJ, Connell CJ, McCullough ML, et al. Vitamin C, vitamin E, and multivitamin supplement use and stomach cancer mortality in the Cancer Prevention Study Ⅱ cohort. Cancer Epidemiol Biomarkers Prev 2002; 11: 35-41.
    [127]Ⅰ ARC Unit of Descriptive Epidemiology:WHO cancer mortality databank. Cancer Mondial.2001; Available from URL:www-dep.iarc.fr/ataava/globocan /who. Htm.

© 2004-2018 中国地质图书馆版权所有 京ICP备05064691号 京公网安备11010802017129号

地址:北京市海淀区学院路29号 邮编:100083

电话:办公室:(+86 10)66554848;文献借阅、咨询服务、科技查新:66554700