Review Open Access
Copyright ©2014 Baishideng Publishing Group Inc. All rights reserved.
World J Med Genet. Aug 27, 2014; 4(3): 58-68
Published online Aug 27, 2014. doi: 10.5496/wjmg.v4.i3.58
Molecular genetics of gastric adenocarcinoma in clinical practice
Margaret Cho, Ogechukwu Eze, Ruliang Xu, Department of Pathology, New York University Langone Medical Center, New York, NY 10016, United States
Author contributions: Cho M and Eze O searched for literatures and wrote portions of manuscript; Xu R wrote and edited the majority of manuscript.
Correspondence to: Ruliang Xu, MD, PhD, Department of Pathology, New York University Langone Medical Center, 550 First Avenue, New York, NY 10016, United States. ruliang.xu@nyumc.org
Telephone: +1-212-2630728 Fax: +1-212-2637916
Received: December 24, 2013
Revised: April 16, 2014
Accepted: May 15, 2014
Published online: August 27, 2014

Abstract

The molecular genetics of gastric carcinoma (GC) dictates their biology and clinical behavior. The two morphologically distinct types of gastric carcinoma by Lauren classification, i.e., intestinal and diffuse cell types, have a significant difference in clinical outcome. These two types of GC have different molecular pathogenetic pathways with unique genetic alterations. In addition to environmental and other etiologies, intestinal type GC is associated with Helicobacter pylori (H. pylori) infection and involves a multistep molecular pathway driving the normal epithelium to intestinal metaplasia, dysplasia, and malignant transformation by chromosomal and/or microsatellite instability (MSI), mutation of tumor suppressor genes, and loss of heterozygosity among others. Diffuse type shows no clear causal relationship with H. pylori infection, but is commonly associated with deficiency of cell-cell adhesion due to mutation of the E-cadherin gene (CDH1), and a manifestation of the hereditary gastric cancer syndrome. Thus, detection of CDH1 mutation or loss of expression of E-cadherin may aid in early diagnosis or screening of diffuse type GC. Detection of certain genetic markers, for example, MSI and matrix metalloproteinases, may provide prognostic information, particularly for intestinal type. The common genetic alterations may offer therapeutic targets for treatment of GC. Polymorphisms in Thymidylate synthase to metabolize 5-fluorouracil, glutathione S-transferase for degradation of Cisplatin, and amplification/overexpression of human epidermal growth factor receptor 2 targeted by monoclonal antibody Trastuzumab, are a few examples. P13K/Akt/mTOR pathway, c-Met pathways, epidermal growth factor receptor, insulin-like growth factor receptor, vascular endothelial growth factor receptor fibroblast growth factor receptor, and micro RNAs are several potential therapeutic biomarkers for GC under investigation.

Key Words: Molecular genetics, Lauren classification, Intestinal type gastric cancer, Diffuse type gastric cancer, Molecular Biomarker

Core tip: Intestinal and diffuse cell types of gastric carcinoma have a significant difference in clinical outcome with different molecular pathogenetic pathways. Intestinal type gastric carcinoma (GC) is associated with chromosomal and/or microsatellite instability, mutation of tumor suppressor genes, and loss of heterozygosity. Diffuse type GC is commonly associated with mutation of the E-cadherin gene, and a manifestation of the hereditary gastric cancer syndrome. Detection of certain mutations may aid in early diagnosis, screening, and prognostication of GC, and common genetic alterations may offer therapeutic targets for treatment. Furthermore, potential therapeutic biomarkers for GC are under investigation and may hold future promise.



INTRODUCTION

Gastric carcinoma (GC) is the second major leading cause of cancer-related death and fourth most common cancer worldwide[1]. The relatively unfavorable outcome is largely attributable to complex biology and marginal effectiveness of treatment options, including surgical resection, chemotherapy, and multidisciplinary approach. Even with use of multimodality approaches, overall survival continues to be poor with 30%-36% 5-year survival rates[2,3]. Chemotherapy is the main treatment in cases of metastatic disease, and the median survival time is only 9 to 14 mo[3,4]. However, not all of GCs have the same outcomes. The biological behavior and clinical presentation of GCs differ with their histological and molecular features.

The current World Health Organization classification (2010 edition), classifies GC into many different types based upon the histology combined with molecular genetic information. However, the traditional Lauren classification, purely on histologic basis, is the most commonly used system. It classifies GC into three different groups: (1) intestinal type with glandular differentiation or pattern; (2) diffuse, or poorly differentiated type, including a signet ring cell histology; and (3) mixed or indeterminate type[5]. The first two types of GCs (intestinal and diffuse types) have distinct histogenesis as well as clinical characteristics. Recent data suggest that these two groups largely differ in their molecular genetics. This paper will mainly review the molecular characteristics of non-hereditary intestinal and diffuse types of GC to understand the molecular pathways involved in GC development and to identify molecular targets for diagnosis, therapy, and prognostication.

MOLECULAR BASIS OF PATHOGENESIS
Genetic predisposition of GC

Certain genetic polymorphisms are predisposed to an increased risk for gastric cancer. These polymorphisms were found in genes involved in the inflammatory response to Helicobacter pylori (H. pylori) infection[6-8], prevention of DNA to oxidative damage[9], and mucosal protection against H. pylori infection[10-12], and detoxification[13,14]. Polymorphisms of the interleukin 1 (IL-1β) gene consistently show strong association with GC[15]. The association is also seen with other genes, including IL-1 receptor antagonist genes[15-19], tumor necrosis factor-alpha gene[18,20,21], rs11556218 T ⁄G polymorphism of the IL-16 gene[8], and genes encoding glutathione-S-transferase (GST) (GSTT1 and GSTM1)[22,23]. Many hereditary tumor syndromes increase the risk to develop GC. The high risk association is well-documented in Hereditary diffuse-type gastric cancer syndrome, familial adenomatous polyposis, hereditary nonpolyposis colorectal cancer, Peutz-Jeghers syndrome, Juvenile polyposis and Li-Fraumeni syndrome[24,25].

Molecular pathogenesis of intestinal cell type of GC

The major etiology of the intestinal type includes dietary, environmental factors, and H. pylori infection[26]. There are many good reviews that have discussed the role and significance of dietary habits and environmental factors in gastric carcinogenesis, but H. pylori infection has also been shown to emerge as an important carcinogen in the stomach. The bacterial virulence factors of H. pylori contributing to GC risk include vacuolating cytotoxin A (vacA), blood group antigen binding adhesion 2, outer inflammatory protein, and cytotoxin-associated gene product (cagA) genes[27,28]. Infection with a cagA-positive H.pylori strain in comparison with a cagA-negative strain increases the risk for development of GC[29]. CagA is translocated into host cells and induces a growth factor-like response in gastric epithelial cells by forming a physical complex with the Src homology 2 domain-containing tyrosine phosphatase in a phosphorylation dependent manner[30,31]. In addition, aberrant expression of activation-induced cytidine deaminase, a gene originally linked to immunoglobulin class switching and B lymphocyte hypermutation, results in accumulation of mutations in the p53 tumor suppressor gene[31,32]. A second virulence gene, the vacA, induces gastric epithelial cell apoptosis and interferes with T cell activation which suppresses local immune response[33]. Chronic inflammation also causes genetic instability through the generation of reactive oxygen and nitrogen species which can directly damage the genomic and mitochondrial DNA[31,34].

The development of GC is a multi-step process. Chronic or atrophic gastritis may lead to intestinal metaplasia, subsequently dysplasia, and eventually carcinoma in some patients[26,35]. During the above progression, a series of genetic alterations occur. The inactivation of tumor suppressor gene p53 is involved in early carcinogenesis, and is found in 38% of intestinal metaplasia, 58% of dysplasia, and 38%-71% of GC[36-38]. Mutations occur more commonly in CpG sites of p53 and transition of G:C to A:T at these sites is the most common type of mutation irrespective of the histologic type of GC[39]. Mutation of p73, a member of the p53 family, is indicated in the carcinogenesis of GC associated with H. pylori infection in the mouse model[40].

Chromosomal instability in intestinal-type GC includes gains at 8q, 17q, 20q and losses at 3p and 5q[28,41,42]. Microsatellite instability-high is often seen in intestinal type of GC, largely due to epigenetic effect, (i.e., hypermethylation of the promoter regions of mismatch repair genes, most commonly mutL homolog 1 and mutS homolog 2, and in small percentage of cases, gene mutations[43-45]. The CpG island methylator phenotype was found in 24%-47% of GC, similar to colorectal cancer[43,46-48]. Recent studies have conducted a genome-wide search to identify novel methylation-silenced genes in GC[49,50]. GC cell lines were treated with a demethylating and/or deacetyling agent and were screened for epigenetically silenced genes using oligonucleotide microarrays[49]. The gene encoding serine proteases inhibitor Tissue Factor Pathway Inhibitor 2 was found to be highly methylated (81%) in GC, and its methylation was a significant and independent prognostic indicator in GC[31,49].

Loss of heterozygosity (LOH) or mutation of Adenomatous polyposis coli (APC) gene may be found in approximately 25% of the cancer precursors, adenomas, and in up to 60% of intestinal-type GC[51-53]. Mutation of CTNNB1, which encodes β-catenin, appears to be exclusive to the mutations that inactivate APC protein[54]. β-catenin accumulates in the cytoplasm, binds to members of the Tcf/Lef family of transcription factors, and is translocated to the nucleus where the Tcf/β-catenin complex activates target genes such as MYC and cyclin D1 gene[31,54]. The incidence of CTNNB1 mutations in intestinal- vs diffuse-type GC remains unclear. One study reported no mutations in diffuse-type GC but 27% incidence in intestinal-type GC[55]. Clements et al[56] 2002 found that 26% of tumors with β-catenin nuclear staining contained CTNNB1 mutations, with no difference between diffuse- and intestinal-type GC.

LOH at the bcl-2 locus and amplification of cyclin D1 and E genes are also associated with intestinal-type GCs[57,58]. The oncogene ErbB2 (Her-2/neu) is amplified in approximately 20% of intestinal type GCs[59]. E-cadherin gene (CDH1) mutations have an insignificant association with the development of intestinal-type GC, in contast to diffuse type GC[60,61].

RUNX3 is now accepted as a tumor suppressor gene and first reported in gastric epithelial cells of RUNX3 knockout mice in 2002[62]. Approximately 45%-60% of human GCs show loss of RUNX3 expression due to hemizygous deletion and hypermethylation of the promoter region, and RUNX3 hypermethylation is seen in H. pylori infection, intestinal metaplasia, and gastric adenoma[31,62,63]. In response to transforming growth factor (TGF)-β, RUNX3 inhibits gastric epithelial proliferation by inducing the CDKN1A (p21) gene[64] and also upregulates the expression of proapoptotic gene BCL2L11 (Bim) in gastric cancer cells treated with TGF-β[65]. Restoration of RUNX3 also strongly inhibited peritoneal metastases of GC in an animal model[66]. RUNX3 inhibited the expression of vascular endothelial growth factor A (VEGF-A) and suppressed angiogenesis and metastasis of GCs[67].

Molecular pathogenesis of diffuse type GC

Little is known about the etiology of diffuse type GC. Epidemiological studies did not link H pylori infection to diffuse type of GC. Its association with hereditary gastric cancer predisposition syndrome is well-documented[68]. The unique molecular genetics of this type of GC, in contrast to intestinal-type, is deficiency of the cell-cell adhesion due to genetic or epigenetic inactivation/down regulation of E-cadherin gene (CDH1). Approximately 50% of diffuse-type GC harbor this mutation or inactivation[24,69]. The abnormality of CDH1 gene can be found in early stage of diffuse GC development and loss of E-cadherin expression is seen in invasive and in situ carcinomas[70]. In a model proposed by Carneiro et al[70], the development of diffuse GC in E-cadherin mutation carriers encompasses in situ signet ring carcinoma with pagetoid spread of signet ring cells as pre-invasive lesions. In early hereditary GC, the wild-type CDH1 allele is suppressed or lost in tumor cells with a second hit caused by promoter hypermethylation of CDH1 in at least 50% of cases[71]. Promoter methylation is also part of the major mechanism underlying E-cadherin downregulation in sporadic diffuse gastric cancers[60]. Chromosomal instability in diffuse-type GC include gains at 12q, 13q and losses at 4q, 15q, 16q, and 17p[28,42,72,73]. The diffuse type GC is also associated with the alterations or mutations in other genes or gene products, including the met proto-oncogene encoding the hepatocyte growth factor receptor and the SC-1 antigen ( an apoptosis receptor)[74-76].

Molecular pathogenesis of Epstein-Barr virus-associated GC

Five percent of GC is associated with monoclonal proliferation of Epstein-Barr virus (EBV)-infected epithelial cells and is a specific clinicopathologic subset with characteristics of younger age, male predominance, proximal location, lower rate of lymph node involvement, marked lymphocytic infiltration, and lace pattern within the mucosa[31,77]. EBV maintains its latent infection and expresses viral latent genes which include EBV-determined nuclear antigen 1, EBV-encoded small RNA, latent membrane protein 2A (LMP2A) and Bam H1-A rightward transcripts (BARTs)[31,78]. Frequent loss of p16 (CDKN2A), smad4, Fhit, and CD82 (KAI-1) are seen[79]. Global CpG island methylation in the PTEN promoter region is considered as a characteristic abnormality in EBV-associated GC[80] with viral LMP2A responsible for aberrant hypermethylation by activation of host DNA methyltransferase 1[81]. LMP2A also upregulates Birc5 (survivin) expression through the activation of nuclear factor-κB, activates extracellular signal regulated kinases (ERK/MAPK1), and inhibits TGF-β-induced apoptosis through activation of the Ras/PI3K/Akt pathway[31,82-84].

GENETIC CHANGES ASSOCIATED WITH MUCIN PHENOTYPIC EXPRESSION IN GC

GC can be classified into four phenotypes according to mucin (MUC1, MUC2 and CD10) expression: gastric or foveolar phenotype (G-type), intestinal phenotype (I-type), intestinal and gastric mixed phenotype and neither gastric nor intestinal phenotype[31,85]. Genetic changes associated with mucin phenotypic expression in GC include TP53 mutations in I-type GC and microsatellite instability in G-type. Specific epigenetic alterations include methylation of hMLH1 occurring more frequently in MUC2-negative GC and more frequently methylated MGMT in MUC2-positive GC than in MUC2-negative GC[31,85].

MOLECULAR DIAGNOSIS OF GC

Genetic markers associated with the development of GC are numerous. However, very few have diagnostic utility. CDH1 probably is the best candidate marker for such purpose. About 50 of diffuse type of GC have CDH1 mutation, either complete or partial deletions of exons, in more than 70% of somatic E-cadherin mutations[24,68]. This unique gene alteration may have a diagnostic potential. CDH1 mutations can be detected by polymerase chain reaction on paraffin-embedded tissue. Detection of the germline mutation in CDH1 may help identify asymptomatic mutations carriers of hereditary gastric cancer syndrome and provide molecular basis for prophylactic total gastrectomy[86].

Molecular prognostication

Currently clinical stage is considered to be the gold standard to predict clinical behavior and the most valuable prognostic factor for all GC types. However, clinical stage does not address the issue of tumor heterogeneity. Many studies have investigated molecular biomarkers as alternatives or supplements to the current staging system. There is some success in identifying biomarkers potentially useful in predicting prognosis and therapeutic response.

Microsatellite instability-high is commonly seen in GC located in the distal stomach or antrum, usually intestinal type. It is less frequently associated with metastasis to local lymph nodes[45,87,88]. However, it is still controversial whether patients with MSI-H GC have a favorable long term survival than those with microsatellite instability-low or microsatellite stability GCs[89,90].

Overexpression of matrix metalloproteinases (MMPs) is shown to be related to tumor invasiveness and metastasis[91]. MMP-1-overexpression in GCs has a worse prognosis than tumors without MMP overexpression. VEGF overexpression is associated with shorter survival time attributable to its enhancement of tumor angiogenesis. Amplification or overexpression of cyclin E is correlated with aggressiveness[92]. Amplification/overexpression of the ERBB2 (Her-2/neu) oncogene in general is considered to be an independent, poor prognostic factor[93,94]. Overexpression of EGF-R and abnormal expression of E-cadherin and β-catenin decrease survival or have poor prognosis[95-97]. Abnormal gene expression of IGF2, KIAA1093, OCT2, PCOLCW, PFN2, RBP4, and three genes (BIK, Aurora kinase B and eIF5A2) identified in the primary tumor are related to node metastasis[98,99]. Expression of caudal type homeobox transcription factor 2 (CDX2) and combination of normal expression of E-cadherin and negative expression of the transmembrane protein MUC1 predict a better prognosis for patients with GC[100,101]. Down-regulation of a cyclin dependent kinase inhibitor, P27/Kip1, is a negative prognostic factor[102,103]. Loss of expression of tumor suppressor gene Rb is related to worse overall survival or inversely correlates with tumor invasion[104,105]. Mutation or abnormal expression of p53 may have a reduced cumulative survival, lymph node metastasis, and lower chemosensitivity[100,106,107], but its overall prognostic significance is controversial[108]. Protection of telomere expression levels are also higher in advanced GC[28].

Molecular therapeutic predicators

The management of patients with GC, particularly those in late stage of tumors, usually requires chemotherapy or target therapy as single or one of the components of combined modality. The chemotherapy or target therapy is toxic, and the effectiveness is variable with patients. Molecular biomarkers have been proven to be a useful tool to predicate therapeutic response and may be used clinically to select patients or chemotherapeutic regimen for optimal result.

Predictors for Fluorouracil treatment: (1) Thymidylate synthase (TYMS) is a catabolizing enzyme for fluorouracil (5-FU). Polymorphisms in the gene encoding TYMS affect expression and appear to be associated with poorer response with 5-FU (47 marker) levels[109,110]. A specific polymorphism in the 5’-untranslated regions is correlated with low sensitivity to 5-FU based chemotherapy and decreased survival in a retrospective study[111]; (2) Dihydropyrimidine dehydrogenase (DPD) and thymidine phosphorylase (TP) are two regulatory enzymes involved in the degradation of 5-FU. Low levels of DPD and TP have been shown to be associated with better response[24,112,113]; and (3) The role of other genes or products for predicting 5-FU response has been also investigated, but results are inconclusive. These molecular markers include glutathione S-transferase (GST), vascular endothelial growth factor, and apoptosis-related genes and gene products including Bcl-2, Bax and p53[43,114,115].

Molecular predictors for Cisplatin treatment: Unlike for 5-FU, molecular predicting markers for chemosensitivity of Cisplatin are not well established. GST, an enzyme that degrades Cisplatin, is one of the potential markers. Its activity is affected by polymorphisms in the GSTM1, GSTT1, and GSTP1 genes, which may in turn cause variable catabolism of Cisplatin and prognosis[111,116,117]. GC with a high LOH rate or MSI-high show a better response to a Cisplatin-based chemotherapy[118].

Molecular targeted therapy: Trastuzumab is a monoclonal antibody targeting HER-2 that has shown an overall survival benefit when combined with palliative chemotherapy in patients with HER-2 amplified GC[119]. HER-2 is currently the only validated therapeutic target in GC with guidelines for HER-2 testing established by the ToGA trial[119].

HER-2 expression may be assessed by immunohistochemistry (IHC), with scoring ranging from 0 to 3+, by gene amplification using fluorescence in situ hybridization (FISH) or by silver in situ hybridization[28,119]. The survival benefit associated with trastuzumab is seen greatest in IHC 3+ or IHC 2+ and FISH-positive patients. Complete membranous staining is not a prerequisite for IHC 2+ or IHC 3+ scores in GC as it is for breast cancer since gastric tumor cells may only show HER-2 staining at the basolateral or lateral membrane regions[28,120].

Other potential candidates for targeted GC therapy include P13K/Akt/mTOR pathway, c-Met pathways, epidermal growth factor receptor (EGFR), VEGF receptor, insulin-like growth factor receptor, and fibroblast growth factor receptor[121].

Lapatinib is a dual kinase inhibitor of EGFR and HER-2 under investigation in two ongoing phase III clinical trials in a select group of patients positive HER-2[121]. These include the Lapatinib Optimization Study in HER-2 Positive Gastric Cancer study with capecitabine and cisplatin in the first-line setting and the TYTAN study in second-line therapy using paclitaxel[121-123].

In terms of other agents, targeting human EGFR in GC remains controversial. Cetuximab is targeted against EGFR and is a recombinant human, chimeric IgG1 monoclonal antibody[121,124]. With combined chemotherapy and cetuximab, promising results have been shown in a phase II trial, but when compared to chemotherapy, the EXPAND study (phase III) failed in prolonging the progression free survival (PFS) and overall survival[125]. The REAL-III trial did not show any advantage of adding panitumumab to a combination of chemotherapy and also showed a worse overall survival and PFS[126-128]. The combination with matuzumab and chemotherapy seems more promising but was evaluated only in phase II trials[129]. Thus, additional studies are necessary.

Antiangiogenic therapy has shown minimal effectiveness when compared to existing treatments for GC[130,131]. Biomarkers such as serum VEGF-A and microvessel density still remain unconfirmed as potentially useful predictive markers by phase III trials[121]. However, in advanced cases of GC treated with the VEGF inhibitor bevacizumab, plasma VEGF-A and tumor neuropilin-1 are strong biomarker candidates for predicting clinical outcome[132].

Various ongoing trials are testing potential targeting agents addressed to the downstream components of VEGF-R/EGFR, such as inhibitors of mTOR, c-Met, and Histone deacetylase[121]. The phase III trial (GRANITE-1) of everolimus, an inhibitor of the P13K/Akt/mTOR pathway, has reported prolonged PFS with a 34% reduction of the risk of progression[121,133].

MICRORNAS AS THERAPEUTIC TARGETS

In recent years, microRNAs (miRNAs) have been investigated as potential markers in treatment of GC. MiRNAs are important regulators of genes with critical roles in cell proliferation, differentiation, and survival[134]. MiRNAs play important roles in the pathogenesis of a variety of malignancies[135-139]. Different miRNA methylation profiles are seen in various cancers. MiR-155 is down-regulated and methylated in GC[140]. MiR-155 is up-regulated in breast cancer[141], colorectal cancer[142] and pancreatic ductal adenocarcinoma[143]. Upregulated miRNAs might act as oncogenes and target tumor suppressors, while down-regulated miRNAs might act as tumor suppressors and target oncogenes[144]. Several miRNAs have been found to be deregulated in GC but the specific molecular mechanisms are unknown[144]. DNA hypermethylation in the miRNA 50 regulatory region accounts for the downregulation of miRNA in tumors[145,146], and many miRNAs have been reported to be down-regulated due to hypermethylation of the CpG islands in GC. MiR-124a-1, miR-124a-2 and miR-124a-3 have been found to be methylation-silenced in GC cell lines[144,147]. Such epigenetic changes are reversible, and make them a potential therapeutic target. Silenced miRNAs in GC could be restored by treating with demethylating agents, such as decitabine (5-aza-20-deoxycytidine), which leads to inhibition of growth, invasion, and metastasis of GC cells[148].

Interestingly, studies have shown that the miRNA methylation levels are positively associated with the clinical stage of GC patients[144]. Low expression of miR-34b and miR-129-3p are associated with a poor clinical outcome of GC patients, and hypermethylation of miR-129-2 and miR-34b CpG islands had a tendency to show poor clinicopathological features[144,149]. Thus, specific miRNA methylation levels may be used in the prognosis of GC patients. However, limitations exist as several factors besides methylation can affect miRNA expression levels. As reported by Tsukamoto et al[148], the expression of miR-375 in NUGC3 cells can be significantly increased with either 5-aza-2-deoxycytidine and markedly up-regulated by greater than 20-fold when treated with both 5-aza-2-deoxycytidine and trichostatin A[148]. In addition, H. pylori infection can induce aberrant DNA methylation in gastric epithelial cells[150]. Individuals with H. pylori had 7.8-13.1-fold higher methylation levels than those without H. pylori infection[147,151]. Another limitation is the serious side effects of demethylating drugs. The use of demethylating agents may induce the expression of many otherwise normally silenced genes and cause a variety of diseases. Thus, the use of demethylating agents in restoring the expression of epigenetically silenced miRNA in GC still requires further investigation.

CONCLUSION

The current research has provided some insights to the genetics of gastric cancer. Clinical trials based upon the genetic information have generated promising results. However, up to date, we still do not have an optimal solution for prevention, early diagnosis, and treatment of this disease. The advanced molecular technology, particularly next generation sequencing, may offer hope in deciphering the myth behind the molecular genetics of gastric cancer. Equipped with the advanced technology, together with efforts from clinical oncology and bioinformatics, we have gradually gained much more understanding about the genetic basis of the host-environmental interaction and will have a greater opportunity to identify diagnostic and therapeutic markers for gastric cancer. These advancements have shed light in finding a cure for gastric cancer in the near future.

Footnotes

P- Reviewer: Declich P, Kumar R, Montecucco A S- Editor: Ji FF L- Editor: A E- Editor: Liu SQ

References
1.  Parkin DM, Bray F, Ferlay J, Pisani P. Global cancer statistics, 2002. CA Cancer J Clin. 2005;55:74-108.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13286]  [Cited by in F6Publishing: 13414]  [Article Influence: 706.0]  [Reference Citation Analysis (1)]
2.  Macdonald JS, Smalley SR, Benedetti J, Hundahl SA, Estes NC, Stemmermann GN, Haller DG, Ajani JA, Gunderson LL, Jessup JM. Chemoradiotherapy after surgery compared with surgery alone for adenocarcinoma of the stomach or gastroesophageal junction. N Engl J Med. 2001;345:725-730.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2465]  [Cited by in F6Publishing: 2365]  [Article Influence: 102.8]  [Reference Citation Analysis (0)]
3.  Cunningham D, Starling N, Rao S, Iveson T, Nicolson M, Coxon F, Middleton G, Daniel F, Oates J, Norman AR. Capecitabine and oxaliplatin for advanced esophagogastric cancer. N Engl J Med. 2008;358:36-46.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1579]  [Cited by in F6Publishing: 1611]  [Article Influence: 100.7]  [Reference Citation Analysis (0)]
4.  Wagner AD, Grothe W, Haerting J, Kleber G, Grothey A, Fleig WE. Chemotherapy in advanced gastric cancer: a systematic review and meta-analysis based on aggregate data. J Clin Oncol. 2006;24:2903-2909.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 833]  [Cited by in F6Publishing: 872]  [Article Influence: 48.4]  [Reference Citation Analysis (0)]
5.  Lauren P. The two histological main types of gastric carcinoma: diffuse and so-called intestinal-type carcinoma. an attempt at a histo-clinical classification. Acta Pathol Microbiol Scand. 1965;64:31-49.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Crusius JB, Canzian F, Capellá G, Peña AS, Pera G, Sala N, Agudo A, Rico F, Del Giudice G, Palli D. Cytokine gene polymorphisms and the risk of adenocarcinoma of the stomach in the European prospective investigation into cancer and nutrition (EPIC-EURGAST). Ann Oncol. 2008;19:1894-1902.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 85]  [Cited by in F6Publishing: 100]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
7.  Canedo P, Durães C, Pereira F, Regalo G, Lunet N, Barros H, Carneiro F, Seruca R, Rocha J, Machado JC. Tumor necrosis factor alpha extended haplotypes and risk of gastric carcinoma. Cancer Epidemiol Biomarkers Prev. 2008;17:2416-2420.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 251]  [Reference Citation Analysis (0)]
8.  Gao LB, Rao L, Wang YY, Liang WB, Li C, Xue H, Zhou B, Sun H, Li Y, Lv ML. The association of interleukin-16 polymorphisms with IL-16 serum levels and risk of colorectal and gastric cancer. Carcinogenesis. 2009;30:295-299.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 73]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
9.  Capellá G, Pera G, Sala N, Agudo A, Rico F, Del Giudicce G, Plebani M, Palli D, Boeing H, Bueno-de-Mesquita HB. DNA repair polymorphisms and the risk of stomach adenocarcinoma and severe chronic gastritis in the EPIC-EURGAST study. Int J Epidemiol. 2008;37:1316-1325.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 61]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
10.  Hishida A, Matsuo K, Goto Y, Mitsuda Y, Hiraki A, Naito M, Wakai K, Tajima K, Hamajima N. Toll-like receptor 4 +3725 G/C polymorphism, Helicobacter pylori seropositivity, and the risk of gastric atrophy and gastric cancer in Japanese. Helicobacter. 2009;14:47-53.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 66]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
11.  Zhang Y, Jin M, Liu B, Ma X, Yao K, Li Q, Chen K. Association between H-RAS T81C genetic polymorphism and gastrointestinal cancer risk: a population based case-control study in China. BMC Cancer. 2008;8:256.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 26]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
12.  Sawa T, Mounawar M, Tatemichi M, Gilibert I, Katoh T, Ohshima H. Increased risk of gastric cancer in Japanese subjects is associated with microsatellite polymorphisms in the heme oxygenase-1 and the inducible nitric oxide synthase gene promoters. Cancer Lett. 2008;269:78-84.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 28]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
13.  Shibata T, Arisawa T, Tahara T, Ohkubo M, Yoshioka D, Maruyama N, Fujita H, Kamiya Y, Nakamura M, Nagasaka M. Selenoprotein S (SEPS1) gene -105G>A promoter polymorphism influences the susceptibility to gastric cancer in the Japanese population. BMC Gastroenterol. 2009;9:2.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 44]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
14.  Tripathi S, Ghoshal U, Ghoshal UC, Mittal B, Krishnani N, Chourasia D, Agarwal AK, Singh K. Gastric carcinogenesis: Possible role of polymorphisms of GSTM1, GSTT1, and GSTP1 genes. Scand J Gastroenterol. 2008;43:431-439.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 40]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
15.  McNamara D, El-Omar E. Helicobacter pylori infection and the pathogenesis of gastric cancer: a paradigm for host-bacterial interactions. Dig Liver Dis. 2008;40:504-509.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 59]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
16.  Gehmert S, Velapatiño B, Herrera P, Balqui J, Santivañez L, Cok J, Vargas G, Combe J, Passaro DJ, Wen S. Interleukin-1 beta single-nucleotide polymorphism’s C allele is associated with elevated risk of gastric cancer in Helicobacter pylori-infected Peruvians. Am J Trop Med Hyg. 2009;81:804-810.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 16]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
17.  Kumar S, Kumar A, Dixit VK. Evidences showing association of interleukin-1B polymorphisms with increased risk of gastric cancer in an Indian population. Biochem Biophys Res Commun. 2009;387:456-460.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 28]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
18.  Melo Barbosa HP, Martins LC, Dos Santos SE, Demachki S, Assumpção MB, Aragão CD, de Oliveira Corvelo TC. Interleukin-1 and TNF-alpha polymorphisms and Helicobacter pylori in a Brazilian Amazon population. World J Gastroenterol. 2009;15:1465-1471.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 38]  [Cited by in F6Publishing: 47]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
19.  Persson C, Engstrand L, Nyrén O, Hansson LE, Enroth H, Ekström AM, Ye W. Interleukin 1-beta gene polymorphisms and risk of gastric cancer in Sweden. Scand J Gastroenterol. 2009;44:339-345.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 28]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
20.  Garza-González E, Bosques-Padilla FJ, El-Omar E, Hold G, Tijerina-Menchaca R, Maldonado-Garza HJ, Pérez-Pérez GI. 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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 89]  [Cited by in F6Publishing: 98]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
21.  El-Omar EM, Carrington M, Chow WH, McColl KE, Bream JH, Young HA, Herrera J, Lissowska J, Yuan CC, Rothman N. Interleukin-1 polymorphisms associated with increased risk of gastric cancer. Nature. 2000;404:398-402.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1690]  [Cited by in F6Publishing: 1614]  [Article Influence: 67.3]  [Reference Citation Analysis (0)]
22.  Zendehdel K, Bahmanyar S, McCarthy S, Nyren O, Andersson B, Ye W. Genetic polymorphisms of glutathione S-transferase genes GSTP1, GSTM1, and GSTT1 and risk of esophageal and gastric cardia cancers. Cancer Causes Control. 2009;20:2031-2038.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 43]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
23.  Moy KA, Yuan JM, Chung FL, Wang XL, Van Den Berg D, Wang R, Gao YT, Yu MC. Isothiocyanates, glutathione S-transferase M1 and T1 polymorphisms and gastric cancer risk: a prospective study of men in Shanghai, China. Int J Cancer. 2009;125:2652-2659.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 57]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
24.  Napieralski R, Ott K, Kremer M, Specht K, Vogelsang H, Becker K, Müller M, Lordick F, Fink U, Rüdiger Siewert J. Combined GADD45A and thymidine phosphorylase expression levels predict response and survival of neoadjuvant-treated gastric cancer patients. Clin Cancer Res. 2005;11:3025-3031.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Hamilton JP, Meltzer SJ. A review of the genomics of gastric cancer. Clin Gastroenterol Hepatol. 2006;4:416-425.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 79]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
26.  Kelley JR, Duggan JM. Gastric cancer epidemiology and risk factors. J Clin Epidemiol. 2003;56:1-9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 496]  [Cited by in F6Publishing: 546]  [Article Influence: 26.0]  [Reference Citation Analysis (0)]
27.  Tan IB, Ng I, Tai WM, Tan P. Understanding the genetic basis of gastric cancer: recent advances. Expert Rev Gastroenterol Hepatol. 2012;6:335-341.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 30]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
28.  Zulfiqar M, Bhalla A, Weindel M, Shidham VB. Molecular diagnostics in esophageal and gastric neoplasms. Clin Lab Med. 2013;33:867-873.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 6]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
29.  Parsonnet J, Friedman GD, Orentreich N, Vogelman H. Risk for gastric cancer in people with CagA positive or CagA negative Helicobacter pylori infection. Gut. 1997;40:297-301.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Higashi H, Tsutsumi R, Muto S, Sugiyama T, Azuma T, Asaka M, Hatakeyama M. SHP-2 tyrosine phosphatase as an intracellular target of Helicobacter pylori CagA protein. Science. 2002;295:683-686.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Jang BG, Kim WH. Molecular pathology of gastric carcinoma. Pathobiology. 2011;78:302-310.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 94]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
32.  Matsumoto Y, Marusawa H, Kinoshita K, Endo Y, Kou T, Morisawa T, Azuma T, Okazaki IM, Honjo T, Chiba T. Helicobacter pylori infection triggers aberrant expression of activation-induced cytidine deaminase in gastric epithelium. Nat Med. 2007;13:470-476.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 379]  [Cited by in F6Publishing: 370]  [Article Influence: 21.8]  [Reference Citation Analysis (0)]
33.  Yamasaki E, Wada A, Kumatori A, Nakagawa I, Funao J, Nakayama M, Hisatsune J, Kimura M, Moss J, Hirayama T. Helicobacter pylori vacuolating cytotoxin induces activation of the proapoptotic proteins Bax and Bak, leading to cytochrome c release and cell death, independent of vacuolation. J Biol Chem. 2006;281:11250-11259.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 106]  [Cited by in F6Publishing: 114]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
34.  Farinati F, Cardin R, Cassaro M, Bortolami M, Nitti D, Tieppo C, Zaninotto G, Rugge M. Helicobacter pylori, inflammation, oxidative damage and gastric cancer: a morphological, biological and molecular pathway. Eur J Cancer Prev. 2008;17:195-200.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 58]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
35.  Oberhuber G, Stolte M. Gastric polyps: an update of their pathology and biological significance. Virchows Arch. 2000;437:581-590.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 85]  [Cited by in F6Publishing: 92]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
36.  Shiao YH, Rugge M, Correa P, Lehmann HP, Scheer WD. p53 alteration in gastric precancerous lesions. Am J Pathol. 1994;144:511-517.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Xiangming C, Hokita S, Natsugoe S, Tanabe G, Baba M, Takao S, Kuroshima K, Aikou T. p21 expression is a prognostic factor in patients with p53-negative gastric cancer. Cancer Lett. 2000;148:181-188.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 19]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
38.  Joypaul BV, Newman EL, Hopwood D, Grant A, Qureshi S, Lane DP, Cuschieri A. Expression of p53 protein in normal, dysplastic, and malignant gastric mucosa: an immunohistochemical study. J Pathol. 1993;170:279-283.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 63]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
39.  Fenoglio-Preiser CM, Wang J, Stemmermann GN, Noffsinger A. TP53 and gastric carcinoma: a review. Hum Mutat. 2003;21:258-270.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 172]  [Cited by in F6Publishing: 179]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
40.  Wei J, O’Brien D, Vilgelm A, Piazuelo MB, Correa P, Washington MK, El-Rifai W, Peek RM, Zaika A. Interaction of Helicobacter pylori with gastric epithelial cells is mediated by the p53 protein family. Gastroenterology. 2008;134:1412-1423.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 51]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
41.  Kokkola A, Monni O, Puolakkainen P, Larramendy ML, Victorzon M, Nordling S, Haapiainen R, Kivilaakso E, Knuutila S. 17q12-21 amplicon, a novel recurrent genetic change in intestinal type of gastric carcinoma: a comparative genomic hybridization study. Genes Chromosomes Cancer. 1997;20:38-43.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 2]  [Reference Citation Analysis (0)]
42.  Wu MS, Chang MC, Huang SP, Tseng CC, Sheu JC, Lin YW, Shun CT, Lin MT, Lin JT. Correlation of histologic subtypes and replication error phenotype with comparative genomic hybridization in gastric cancer. Genes Chromosomes Cancer. 2001;30:80-86.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 4]  [Reference Citation Analysis (0)]
43.  Yeh KH, Shun CT, Chen CL, Lin JT, Lee WJ, Lee PH, Chen YC, Cheng AL. High expression of thymidylate synthase is associated with the drug resistance of gastric carcinoma to high dose 5-fluorouracil-based systemic chemotherapy. Cancer. 1998;82:1626-1631.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 2]  [Reference Citation Analysis (0)]
44.  Gu M, Kim D, Bae Y, Choi J, Kim S, Song S. Analysis of microsatellite instability, protein expression and methylation status of hMLH1 and hMSH2 genes in gastric carcinomas. Hepatogastroenterology. 2009;56:899-904.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Seo HM, Chang YS, Joo SH, Kim YW, Park YK, Hong SW, Lee SH. Clinicopathologic characteristics and outcomes of gastric cancers with the MSI-H phenotype. J Surg Oncol. 2009;99:143-147.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 79]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
46.  Oue N, Oshimo Y, Nakayama H, Ito R, Yoshida K, Matsusaki K, Yasui W. DNA methylation of multiple genes in gastric carcinoma: association with histological type and CpG island methylator phenotype. Cancer Sci. 2003;94:901-905.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 74]  [Cited by in F6Publishing: 78]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
47.  Toyota M, Ahuja N, Suzuki H, Itoh F, Ohe-Toyota M, Imai K, Baylin SB, Issa JP. Aberrant methylation in gastric cancer associated with the CpG island methylator phenotype. Cancer Res. 1999;59:5438-5442.  [PubMed]  [DOI]  [Cited in This Article: ]
48.  Zhang KL, Sun Y, Li Y, Liu M, Qu B, Cui SH, Kong QY, Chen XY, Li H, Liu J. Increased frequency of CpG island methylator phenotype and CDH1 methylation in a gastric cancer high-risk region of china. Transl Oncol. 2008;1:28-35.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 15]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
49.  Jee CD, Kim MA, Jung EJ, Kim J, Kim WH. Identification of genes epigenetically silenced by CpG methylation in human gastric carcinoma. Eur J Cancer. 2009;45:1282-1293.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 74]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
50.  Yamashita S, Tsujino Y, Moriguchi K, Tatematsu M, Ushijima T. Chemical genomic screening for methylation-silenced genes in gastric cancer cell lines using 5-aza-2’-deoxycytidine treatment and oligonucleotide microarray. Cancer Sci. 2006;97:64-71.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 191]  [Cited by in F6Publishing: 201]  [Article Influence: 11.2]  [Reference Citation Analysis (0)]
51.  Nakatsuru S, Yanagisawa A, Furukawa Y, Ichii S, Kato Y, Nakamura Y, Horii A. Somatic mutations of the APC gene in precancerous lesion of the stomach. Hum Mol Genet. 1993;2:1463-1465.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 52]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
52.  Nakatsuru S, Yanagisawa A, Ichii S, Tahara E, Kato Y, Nakamura Y, Horii A. Somatic mutation of the APC gene in gastric cancer: frequent mutations in very well differentiated adenocarcinoma and signet-ring cell carcinoma. Hum Mol Genet. 1992;1:559-563.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 165]  [Cited by in F6Publishing: 177]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
53.  Horii A, Nakatsuru S, Miyoshi Y, Ichii S, Nagase H, Kato Y, Yanagisawa A, Nakamura Y. The APC gene, responsible for familial adenomatous polyposis, is mutated in human gastric cancer. Cancer Res. 1992;52:3231-3233.  [PubMed]  [DOI]  [Cited in This Article: ]
54.  Polakis P. Wnt signaling and cancer. Genes Dev. 2000;14:1837-1851.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Park WS, Oh RR, Park JY, Lee SH, Shin MS, Kim YS, Kim SY, Lee HK, Kim PJ, Oh ST. Frequent somatic mutations of the beta-catenin gene in intestinal-type gastric cancer. Cancer Res. 1999;59:4257-4260.  [PubMed]  [DOI]  [Cited in This Article: ]
56.  Clements WM, Wang J, Sarnaik A, Kim OJ, MacDonald J, Fenoglio-Preiser C, Groden J, Lowy AM. beta-Catenin mutation is a frequent cause of Wnt pathway activation in gastric cancer. Cancer Res. 2002;62:3503-3506.  [PubMed]  [DOI]  [Cited in This Article: ]
57.  Ayhan A, Yasui W, Yokozaki H, Seto M, Ueda R, Tahara E. Loss of heterozygosity at the bcl-2 gene locus and expression of bcl-2 in human gastric and colorectal carcinomas. Jpn J Cancer Res. 1994;85:584-591.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 45]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
58.  Müller W, Noguchi T, Wirtz HC, Hommel G, Gabbert HE. Expression of cell-cycle regulatory proteins cyclin D1, cyclin E, and their inhibitor p21 WAF1/CIP1 in gastric cancer. J Pathol. 1999;189:186-193.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 2]  [Reference Citation Analysis (0)]
59.  Wang YL, Sheu BS, Yang HB, Lin PW, Chang YC. Overexpression of c-erb-B2 proteins in tumor and non-tumor parts of gastric adenocarcinoma--emphasis on its relation to H. pylori infection and clinicohistological characteristics. Hepatogastroenterology. 2002;49:1172-1176.  [PubMed]  [DOI]  [Cited in This Article: ]
60.  Machado JC, Oliveira C, Carvalho R, Soares P, Berx G, Caldas C, Seruca R, Carneiro F, Sobrinho-Simöes M. E-cadherin gene (CDH1) promoter methylation as the second hit in sporadic diffuse gastric carcinoma. Oncogene. 2001;20:1525-1528.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 193]  [Cited by in F6Publishing: 210]  [Article Influence: 9.1]  [Reference Citation Analysis (0)]
61.  Mingchao TR, Stockton P, Sun K, Sills RC, Clayton N, Portier M, Flake G. Loss of E-cadherin expression in gastric intestinal metaplasia and later stage p53 altered expression in gastric carcinogenesis. Exp Toxicol Pathol. 2001;53:237-246.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 18]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
62.  Li QL, Ito K, Sakakura C, Fukamachi H, Inoue Ki, Chi XZ, Lee KY, Nomura S, Lee CW, Han SB. Causal relationship between the loss of RUNX3 expression and gastric cancer. Cell. 2002;109:113-124.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 789]  [Cited by in F6Publishing: 820]  [Article Influence: 37.3]  [Reference Citation Analysis (0)]
63.  Kim TY, Lee HJ, Hwang KS, Lee M, Kim JW, Bang YJ, Kang GH. Methylation of RUNX3 in various types of human cancers and premalignant stages of gastric carcinoma. Lab Invest. 2004;84:479-484.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 164]  [Cited by in F6Publishing: 183]  [Article Influence: 9.2]  [Reference Citation Analysis (0)]
64.  Chi XZ, Yang JO, Lee KY, Ito K, Sakakura C, Li QL, Kim HR, Cha EJ, Lee YH, Kaneda A. RUNX3 suppresses gastric epithelial cell growth by inducing p21(WAF1/Cip1) expression in cooperation with transforming growth factor {beta}-activated SMAD. Mol Cell Biol. 2005;25:8097-8107.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 132]  [Cited by in F6Publishing: 144]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
65.  Yano T, Ito K, Fukamachi H, Chi XZ, Wee HJ, Inoue K, Ida H, Bouillet P, Strasser A, Bae SC. The RUNX3 tumor suppressor upregulates Bim in gastric epithelial cells undergoing transforming growth factor beta-induced apoptosis. Mol Cell Biol. 2006;26:4474-4488.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 120]  [Cited by in F6Publishing: 134]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
66.  Sakakura C, Hasegawa K, Miyagawa K, Nakashima S, Yoshikawa T, Kin S, Nakase Y, Yazumi S, Yamagishi H, Okanoue T. Possible involvement of RUNX3 silencing in the peritoneal metastases of gastric cancers. Clin Cancer Res. 2005;11:6479-6488.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 56]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
67.  Peng Z, Wei D, Wang L, Tang H, Zhang J, Le X, Jia Z, Li Q, Xie K. RUNX3 inhibits the expression of vascular endothelial growth factor and reduces the angiogenesis, growth, and metastasis of human gastric cancer. Clin Cancer Res. 2006;12:6386-6394.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 68]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
68.  Guilford P, Hopkins J, Harraway J, McLeod M, McLeod N, Harawira P, Taite H, Scoular R, Miller A, Reeve AE. E-cadherin germline mutations in familial gastric cancer. Nature. 1998;392:402-405.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1135]  [Cited by in F6Publishing: 1093]  [Article Influence: 42.0]  [Reference Citation Analysis (0)]
69.  Berx G, Becker KF, Höfler H, van Roy F. Mutations of the human E-cadherin (CDH1) gene. Hum Mutat. 1998;12:226-237.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 5]  [Reference Citation Analysis (0)]
70.  Carneiro F, Huntsman DG, Smyrk TC, Owen DA, Seruca R, Pharoah P, Caldas C, Sobrinho-Simões M. Model of the early development of diffuse gastric cancer in E-cadherin mutation carriers and its implications for patient screening. J Pathol. 2004;203:681-687.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 196]  [Cited by in F6Publishing: 208]  [Article Influence: 10.4]  [Reference Citation Analysis (0)]
71.  Humar B, Guilford P. Hereditary diffuse gastric cancer: a manifestation of lost cell polarity. Cancer Sci. 2009;100:1151-1157.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 69]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
72.  Weiss MM, Kuipers EJ, Postma C, Snijders AM, Pinkel D, Meuwissen SG, Albertson D, Meijer GA. Genomic alterations in primary gastric adenocarcinomas correlate with clinicopathological characteristics and survival. Cell Oncol. 2004;26:307-317.  [PubMed]  [DOI]  [Cited in This Article: ]
73.  Tsukamoto Y, Uchida T, Karnan S, Noguchi T, Nguyen LT, Tanigawa M, Takeuchi I, Matsuura K, Hijiya N, Nakada C. Genome-wide analysis of DNA copy number alterations and gene expression in gastric cancer. J Pathol. 2008;216:471-482.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 98]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
74.  Kuniyasu H, Yasui W, Kitadai Y, Yokozaki H, Ito H, Tahara E. Frequent amplification of the c-met gene in scirrhous type stomach cancer. Biochem Biophys Res Commun. 1992;189:227-232.  [PubMed]  [DOI]  [Cited in This Article: ]
75.  Vollmers HP, Zimmermann U, Krenn V, Timmermann W, Illert B, Hensel F, Hermann R, Thiede A, Wilhelm M, Rückle-Lanz H. Adjuvant therapy for gastric adenocarcinoma with the apoptosis-inducing human monoclonal antibody SC-1: first clinical and histopathological results. Oncol Rep. 1998;5:549-552.  [PubMed]  [DOI]  [Cited in This Article: ]
76.  Vollmers HP, Hensel F, Hermann R, Dämmrich J, Wozniak E, Gessner P, Herrmann B, Zimmermann U, Müller-Hermelink HK. Tumor-specific apoptosis induced by the human monoclonal antibody SC-1: a new therapeutical approach for stomach cancer. Oncol Rep. 1998;5:35-40.  [PubMed]  [DOI]  [Cited in This Article: ]
77.  Uozaki H, Fukayama M. Epstein-Barr virus and gastric carcinoma--viral carcinogenesis through epigenetic mechanisms. Int J Clin Exp Pathol. 2008;1:198-216.  [PubMed]  [DOI]  [Cited in This Article: ]
78.  Fukayama M. Epstein-Barr virus and gastric carcinoma. Pathol Int. 2010;60:337-350.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 97]  [Cited by in F6Publishing: 102]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
79.  Lee HS, Chang MS, Yang HK, Lee BL, Kim WH. Epstein-barr virus-positive gastric carcinoma has a distinct protein expression profile in comparison with epstein-barr virus-negative carcinoma. Clin Cancer Res. 2004;10:1698-1705.  [PubMed]  [DOI]  [Cited in This Article: ]
80.  Kang GH, Lee S, Kim WH, Lee HW, Kim JC, Rhyu MG, Ro JY. Epstein-barr virus-positive gastric carcinoma demonstrates frequent aberrant methylation of multiple genes and constitutes CpG island methylator phenotype-positive gastric carcinoma. Am J Pathol. 2002;160:787-794.  [PubMed]  [DOI]  [Cited in This Article: ]
81.  Hino R, Uozaki H, Murakami N, Ushiku T, Shinozaki A, Ishikawa S, Morikawa T, Nakaya T, Sakatani T, Takada K. Activation of DNA methyltransferase 1 by EBV latent membrane protein 2A leads to promoter hypermethylation of PTEN gene in gastric carcinoma. Cancer Res. 2009;69:2766-2774.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 258]  [Cited by in F6Publishing: 272]  [Article Influence: 18.1]  [Reference Citation Analysis (0)]
82.  Hino R, Uozaki H, Inoue Y, Shintani Y, Ushiku T, Sakatani T, Takada K, Fukayama M. Survival advantage of EBV-associated gastric carcinoma: survivin up-regulation by viral latent membrane protein 2A. Cancer Res. 2008;68:1427-1435.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 74]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
83.  Chen SY, Lu J, Shih YC, Tsai CH. Epstein-Barr virus latent membrane protein 2A regulates c-Jun protein through extracellular signal-regulated kinase. J Virol. 2002;76:9556-9561.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 65]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
84.  Fukuda M, Longnecker R. Latent membrane protein 2A inhibits transforming growth factor-beta 1-induced apoptosis through the phosphatidylinositol 3-kinase/Akt pathway. J Virol. 2004;78:1697-1705.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 99]  [Cited by in F6Publishing: 97]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
85.  Motoshita J, Oue N, Nakayama H, Kuraoka K, Aung PP, Taniyama K, Matsusaki K, Yasui W. DNA methylation profiles of differentiated-type gastric carcinomas with distinct mucin phenotypes. Cancer Sci. 2005;96:474-479.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 11]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
86.  Fitzgerald RC, Caldas C. E-cadherin mutations and hereditary gastric cancer: prevention by resection? Dig Dis. 2002;20:23-31.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 19]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
87.  Simpson AJ, Caballero OL, Pena SD. Microsatellite instability as a tool for the classification of gastric cancer. Trends Mol Med. 2001;7:76-80.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 41]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
88.  Schneider BG, Bravo JC, Roa JC, Roa I, Kim MC, Lee KM, Plaisance KT, McBride CM, Mera R. Microsatellite instability, prognosis and metastasis in gastric cancers from a low-risk population. Int J Cancer. 2000;89:444-452.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 2]  [Reference Citation Analysis (0)]
89.  Wirtz HC, Müller W, Noguchi T, Scheven M, Rüschoff J, Hommel G, Gabbert HE. Prognostic value and clinicopathological profile of microsatellite instability in gastric cancer. Clin Cancer Res. 1998;4:1749-1754.  [PubMed]  [DOI]  [Cited in This Article: ]
90.  Hayden JD, Cawkwell L, Quirke P, Dixon MF, Goldstone AR, Sue-Ling H, Johnston D, Martin IG. Prognostic significance of microsatellite instability in patients with gastric carcinoma. Eur J Cancer. 1997;33:2342-2346.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 40]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
91.  Noël A, Jost M, Maquoi E. Matrix metalloproteinases at cancer tumor-host interface. Semin Cell Dev Biol. 2008;19:52-60.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 166]  [Cited by in F6Publishing: 184]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
92.  Bani-Hani KE, Almasri NM, Khader YS, Sheyab FM, Karam HN. Combined evaluation of expressions of cyclin E and p53 proteins as prognostic factors for patients with gastric cancer. Clin Cancer Res. 2005;11:1447-1453.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 40]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
93.  Allgayer H, Babic R, Gruetzner KU, Tarabichi A, Schildberg FW, Heiss MM. c-erbB-2 is of independent prognostic relevance in gastric cancer and is associated with the expression of tumor-associated protease systems. J Clin Oncol. 2000;18:2201-2209.  [PubMed]  [DOI]  [Cited in This Article: ]
94.  Parker WB, Cheng YC. Metabolism and mechanism of action of 5-fluorouracil. Pharmacol Ther. 1990;48:381-395.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 310]  [Cited by in F6Publishing: 327]  [Article Influence: 9.6]  [Reference Citation Analysis (0)]
95.  Jonjić N, Kovac K, Krasević M, Valković T, Ernjak N, Sasso F, Melato M. Epidermal growth factor-receptor expression correlates with tumor cell proliferation and prognosis in gastric cancer. Anticancer Res. 1997;17:3883-3888.  [PubMed]  [DOI]  [Cited in This Article: ]
96.  Zhou YN, Xu CP, Han B, Li M, Qiao L, Fang DC, Yang JM. Expression of E-cadherin and beta-catenin in gastric carcinoma and its correlation with the clinicopathological features and patient survival. World J Gastroenterol. 2002;8:987-993.  [PubMed]  [DOI]  [Cited in This Article: ]
97.  Gamboa-Dominguez A, Dominguez-Fonseca C, Quintanilla-Martinez L, Reyes-Gutierrez E, Green D, Angeles-Angeles A, Busch R, Hermannstädter C, Nährig J, Becker KF. Epidermal growth factor receptor expression correlates with poor survival in gastric adenocarcinoma from Mexican patients: a multivariate analysis using a standardized immunohistochemical detection system. Mod Pathol. 2004;17:579-587.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 70]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
98.  Hippo Y, Taniguchi H, Tsutsumi S, Machida N, Chong JM, Fukayama M, Kodama T, Aburatani H. Global gene expression analysis of gastric cancer by oligonucleotide microarrays. Cancer Res. 2002;62:233-240.  [PubMed]  [DOI]  [Cited in This Article: ]
99.  Marchet A, Mocellin S, Belluco C, Ambrosi A, DeMarchi F, Mammano E, Digito M, Leon A, D’Arrigo A, Lise M. Gene expression profile of primary gastric cancer: towards the prediction of lymph node status. Ann Surg Oncol. 2007;14:1058-1064.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 60]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
100.  Fondevila C, Metges JP, Fuster J, Grau JJ, Palacín A, Castells A, Volant A, Pera M. p53 and VEGF expression are independent predictors of tumour recurrence and survival following curative resection of gastric cancer. Br J Cancer. 2004;90:206-215.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 129]  [Cited by in F6Publishing: 145]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
101.  Tanaka M, Kitajima Y, Sato S, Miyazaki K. Combined evaluation of mucin antigen and E-cadherin expression may help select patients with gastric cancer suitable for minimally invasive therapy. Br J Surg. 2003;90:95-101.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 19]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
102.  Chetty R. p27 Protein and cancers of the gastrointestinal tract and liver: an overview. J Clin Gastroenterol. 2003;37:23-27.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 32]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
103.  Catzavelos C, Tsao MS, DeBoer G, Bhattacharya N, Shepherd FA, Slingerland JM. Reduced expression of the cell cycle inhibitor p27Kip1 in non-small cell lung carcinoma: a prognostic factor independent of Ras. Cancer Res. 1999;59:684-688.  [PubMed]  [DOI]  [Cited in This Article: ]
104.  Chou NH, Chen HC, Chou NS, Hsu PI, Tseng HH. Expression of altered retinoblastoma protein inversely correlates with tumor invasion in gastric carcinoma. World J Gastroenterol. 2006;12:7188-7191.  [PubMed]  [DOI]  [Cited in This Article: ]
105.  Feakins RM, Nickols CD, Bidd H, Walton SJ. Abnormal expression of pRb, p16, and cyclin D1 in gastric adenocarcinoma and its lymph node metastases: relationship with pathological features and survival. Hum Pathol. 2003;34:1276-1282.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in F6Publishing: 53]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
106.  Oh SY, Kwon HC, Kim SH, Jang JS, Kim MC, Kim KH, Han JY, Kim CO, Kim SJ, Jeong JS. Clinicopathologic significance of HIF-1alpha, p53, and VEGF expression and preoperative serum VEGF level in gastric cancer. BMC Cancer. 2008;8:123.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 63]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
107.  Pinto-de-Sousa J, Silva F, David L, Leitão D, Seixas M, Pimenta A, Cardoso-de-Oliveira M. Clinicopathological significance and survival influence of p53 protein expression in gastric carcinoma. Histopathology. 2004;44:323-331.  [PubMed]  [DOI]  [Cited in This Article: ]
108.  Anderson C, Nijagal A, Kim J. Molecular markers for gastric adenocarcinoma: an update. Mol Diagn Ther. 2006;10:345-352.  [PubMed]  [DOI]  [Cited in This Article: ]
109.  Marsh S. Thymidylate synthase pharmacogenetics. Invest New Drugs. 2005;23:533-537.  [PubMed]  [DOI]  [Cited in This Article: ]
110.  Kawakami K, Watanabe G. Identification and functional analysis of single nucleotide polymorphism in the tandem repeat sequence of thymidylate synthase gene. Cancer Res. 2003;63:6004-6007.  [PubMed]  [DOI]  [Cited in This Article: ]
111.  Ruzzo A, Graziano F, Kawakami K, Watanabe G, Santini D, Catalano V, Bisonni R, Canestrari E, Ficarelli R, Menichetti ET. Pharmacogenetic profiling and clinical outcome of patients with advanced gastric cancer treated with palliative chemotherapy. J Clin Oncol. 2006;24:1883-1891.  [PubMed]  [DOI]  [Cited in This Article: ]
112.  Terashima M, Irinoda T, Fujiwara H, Nakaya T, Takagane A, Abe K, Yonezawa H, Oyama K, Inaba T, Saito K. Roles of thymidylate synthase and dihydropyrimidine dehydrogenase in tumor progression and sensitivity to 5-fluorouracil in human gastric cancer. Anticancer Res. 2002;22:761-768.  [PubMed]  [DOI]  [Cited in This Article: ]
113.  Hua D, Huang ZH, Mao Y, Deng JZ. Thymidylate synthase and thymidine phosphorylase gene expression as predictive parameters for the efficacy of 5-fluorouracil-based adjuvant chemotherapy for gastric cancer. World J Gastroenterol. 2007;13:5030-5034.  [PubMed]  [DOI]  [Cited in This Article: ]
114.  Giatromanolaki A, Stathopoulos GP, Koukourakis MI, Rigatos S, Vrettou E, Kittas C, Fountzilas G, Sivridis E. Angiogenesis and apoptosis-related protein (p53, bcl-2, and bax) expression versus response of gastric adenocarcinomas to paclitaxel and carboplatin chemotherapy. Am J Clin Oncol. 2001;24:222-226.  [PubMed]  [DOI]  [Cited in This Article: ]
115.  Kikuyama S, Inada T, Shimizu K, Miyakita M, Ogata Y. p53, bcl-2 and thymidine phosphorylase as predictive markers of chemotherapy in patients with advanced and recurrent gastric cancer. Anticancer Res. 2001;21:2149-2153.  [PubMed]  [DOI]  [Cited in This Article: ]
116.  Mannervik B. The isoenzymes of glutathione transferase. Adv Enzymol Relat Areas Mol Biol. 1985;57:357-417.  [PubMed]  [DOI]  [Cited in This Article: ]
117.  Ott K, Lordick F, Becker K, Ulm K, Siewert J, Höfler H, Keller G. Glutathione-S-transferase P1, T1 and M1 genetic polymorphisms in neoadjuvant-treated locally advanced gastric cancer: GSTM1-present genotype is associated with better prognosis in completely resected patients. Int J Colorectal Dis. 2008;23:773-782.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 34]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
118.  Ott K, Vogelsang H, Mueller J, Becker K, Müller M, Fink U, Siewert JR, Höfler H, Keller G. Chromosomal instability rather than p53 mutation is associated with response to neoadjuvant cisplatin-based chemotherapy in gastric carcinoma. Clin Cancer Res. 2003;9:2307-2315.  [PubMed]  [DOI]  [Cited in This Article: ]
119.  Bang YJ, Van Cutsem E, Feyereislova A, Chung HC, Shen L, Sawaki A, Lordick F, Ohtsu A, Omuro Y, Satoh T. Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): a phase 3, open-label, randomised controlled trial. Lancet. 2010;376:687-697.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4615]  [Cited by in F6Publishing: 4831]  [Article Influence: 345.1]  [Reference Citation Analysis (0)]
120.  Bang YJ. Advances in the management of HER2-positive advanced gastric and gastroesophageal junction cancer. J Clin Gastroenterol. 2012;46:637-648.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 67]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
121.  Cidon EU, Ellis SG, Inam Y, Adeleke S, Zarif S, Geldart T. Molecular targeted agents for gastric cancer: a step forward towards personalized therapy. Cancers (Basel). 2013;5:64-91.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 41]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
122.  Satoh T, Bang Y, Wang J, Xu J, Chung HC, Yeh K, Chen J, Mukaiyama A, Yoshida P, Ohtsu A. Interim safety analysis from TYTAN: A phase III Asian study of lapatinib in combination with paclitaxel as second-line therapy in gastric cancer. J Clin Oncol. 2010;28:Abstract 4057.  [PubMed]  [DOI]  [Cited in This Article: ]
123.   Available from: http: //clinicaltrials.gov/ct2/show/NCT00680901?term=LOGiC&rank=8/.  [PubMed]  [DOI]  [Cited in This Article: ]
124.  Martinelli E, De Palma R, Orditura M, De Vita F, Ciardiello F. Anti-epidermal growth factor receptor monoclonal antibodies in cancer therapy. Clin Exp Immunol. 2009;158:1-9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 242]  [Cited by in F6Publishing: 220]  [Article Influence: 14.7]  [Reference Citation Analysis (0)]
125.  Leach B. Cetuximab Fails to Extend PFS in Advanced Gastric Cancer.  Available from: http://www.onclive.com/web-exclusives/Cetuximab-Fails-to-Extend-PFS-in-Advanced-Gastric-Cancer/.  [PubMed]  [DOI]  [Cited in This Article: ]
126.  Okines AF, Ashley SE, Cunningham D, Oates J, Turner A, Webb J, Saffery C, Chua YJ, Chau I. Epirubicin, oxaliplatin, and capecitabine with or without panitumumab for advanced esophagogastric cancer: dose-finding study for the prospective multicenter, randomized, phase II/III REAL-3 trial. J Clin Oncol. 2010;28:3945-3950.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 97]  [Cited by in F6Publishing: 107]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
127.  Chau I, Okines AFC, Gonzalez de Castro D, Saffery C, Barbachano Y, Wotherspoon A, Puckey L, Hulkki Wilson S, Coxon FY, Middleton GW. REAL3: A multicenter randomized phase II/III trial of epirubicin, oxaliplatin, and capecitabine (EOC) versus modified (m) EOC plus panitumumab (P) in advanced oesophagogastric (OG) cancer. Response rate (RR), toxicity, and molecular analysis from phase II. J Clin Oncol. 2011;29:Abstract 4131.  [PubMed]  [DOI]  [Cited in This Article: ]
128.  Waddell TS, Chau I, Barbachano Y, de Castro DG, Wotherspoon A, Saffery C, Middleton GW, Wadsley J, Ferry DR, Mansoor W. A randomized multicenter trial of epirubicin, oxaliplatin, and capecitabine (EOC) plus panitumumab in advanced esophagogastric cancer (REAL3). J Clin Oncol. 2012;30:Abstract 4000.  [PubMed]  [DOI]  [Cited in This Article: ]
129.  Rao S, Starling N, Cunningham D, Sumpter K, Gilligan D, Ruhstaller T, Valladares-Ayerbes M, Wilke H, Archer C, Kurek R. Matuzumab plus epirubicin, cisplatin and capecitabine (ECX) compared with epirubicin, cisplatin and capecitabine alone as first-line treatment in patients with advanced oesophago-gastric cancer: a randomised, multicentre open-label phase II study. Ann Oncol. 2010;21:2213-2219.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 94]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]
130.  Kang Y, Ohtsu A, Van Cutsem E, Rha SY, Sawaki A, Park S, Lim H, Wu J, Langer B, Shah MA. AVAGAST: A randomized, double-blind, placebo-controlled, phase III study of first-line capecitabine and cis plus bevacizumab or placebo in patients with advanced gastric cancer (AGC). J Clin Oncol. 2010;28:Abstract LBA4007.  [PubMed]  [DOI]  [Cited in This Article: ]
131.  Kang Y, Ohtsu A, Van Cutsem E, Roman L, Nunes J, Li C, Otero D, Rivera F, Aprile G, Pimentel Alvarez PR. Survival analysis by pooling risk factors in AVAGAST: First-line capecitabine plus bevacizumab or placebo in patients with advanced gastric cancer. J Clin Oncol. 2011;29:Abstract 4119.  [PubMed]  [DOI]  [Cited in This Article: ]
132.  Van Cutsem E, de Haas S, Kang YK, Ohtsu A, Tebbutt NC, Ming Xu J, Peng Yong W, Langer B, Delmar P, Scherer SJ. Bevacizumab in combination with chemotherapy as first-line therapy in advanced gastric cancer: a biomarker evaluation from the AVAGAST randomized phase III trial. J Clin Oncol. 2012;30:2119-2127.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 355]  [Cited by in F6Publishing: 385]  [Article Influence: 32.1]  [Reference Citation Analysis (0)]
133.  Van Cutsem E, Yeh KH, Bang YJ. Phase III trial of everolimus (EVE) in previously treated patients with advanced gastric cancer (AGC): GRANITE-1. J Clin Oncol. 2012;4:LBA3.  [PubMed]  [DOI]  [Cited in This Article: ]
134.  Ambros V. The functions of animal microRNAs. Nature. 2004;431:350-355.  [PubMed]  [DOI]  [Cited in This Article: ]
135.  Wang H, Li M, Zhang R, Wang Y, Zang W, Ma Y, Zhao G, Zhang G. Effect of miR-335 upregulation on the apoptosis and invasion of lung cancer cell A549 and H1299. Tumour Biol. 2013;34:3101-3109.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 46]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
136.  He H, Di Y, Liang M, Yang F, Yao L, Hao S, Li J, Jiang Y, Jin C, Fu D. The microRNA-218 and ROBO-1 signaling axis correlates with the lymphatic metastasis of pancreatic cancer. Oncol Rep. 2013;30:651-658.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 30]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
137.  Xu YY, Wu HJ, Ma HD, Xu LP, Huo Y, Yin LR. MicroRNA-503 suppresses proliferation and cell-cycle progression of endometrioid endometrial cancer by negatively regulating cyclin D1. FEBS J. 2013;280:3768-3779.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 71]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
138.  Bae HJ, Noh JH, Kim JK, Eun JW, Jung KH, Kim MG, Chang YG, Shen Q, Kim SJ, Park WS. MicroRNA-29c functions as a tumor suppressor by direct targeting oncogenic SIRT1 in hepatocellular carcinoma. Oncogene. 2014;33:2557-2567.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 105]  [Cited by in F6Publishing: 115]  [Article Influence: 10.5]  [Reference Citation Analysis (0)]
139.  Zhao S, Yao DS, Chen JY, Ding N. Aberrant expression of miR-20a and miR-203 in cervical cancer. Asian Pac J Cancer Prev. 2013;14:2289-2293.  [PubMed]  [DOI]  [Cited in This Article: ]
140.  Corral-Fernández NE, Salgado-Bustamante M, Martínez-Leija ME, Cortez-Espinosa N, García-Hernández MH, Reynaga-Hernández E, Quezada-Calvillo R, Portales-Pérez DP. Dysregulated miR-155 expression in peripheral blood mononuclear cells from patients with type 2 diabetes. Exp Clin Endocrinol Diabetes. 2013;121:347-353.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 63]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
141.  Kong W, He L, Richards EJ, Challa S, Xu CX, Permuth-Wey J, Lancaster JM, Coppola D, Sellers TA, Djeu JY. Upregulation of miRNA-155 promotes tumour angiogenesis by targeting VHL and is associated with poor prognosis and triple-negative breast cancer. Oncogene. 2014;33:679-689.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 272]  [Cited by in F6Publishing: 305]  [Article Influence: 27.7]  [Reference Citation Analysis (0)]
142.  Zhang GJ, Xiao HX, Tian HP, Liu ZL, Xia SS, Zhou T. Upregulation of microRNA-155 promotes the migration and invasion of colorectal cancer cells through the regulation of claudin-1 expression. Int J Mol Med. 2013;31:1375-1380.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 85]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
143.  Liu Q, Chen J, Wang J, Amos C, Killary AM, Sen S, Wei C, Frazier ML. Putative tumor suppressor gene SEL1L was downregulated by aberrantly upregulated hsa-mir-155 in human pancreatic ductal adenocarcinoma. Mol Carcinog. 2014;53:711-721.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 29]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
144.  Ma J, Hong L, Chen Z, Nie Y, Fan D. Epigenetic regulation of microRNAs in gastric cancer. Dig Dis Sci. 2014;59:716-723.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 30]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
145.  Krek A, Grün D, Poy MN, Wolf R, Rosenberg L, Epstein EJ, MacMenamin P, da Piedade I, Gunsalus KC, Stoffel M. Combinatorial microRNA target predictions. Nat Genet. 2005;37:495-500.  [PubMed]  [DOI]  [Cited in This Article: ]
146.  Saito Y, Liang G, Egger G, Friedman JM, Chuang JC, Coetzee GA, Jones PA. Specific activation of microRNA-127 with downregulation of the proto-oncogene BCL6 by chromatin-modifying drugs in human cancer cells. Cancer Cell. 2006;9:435-443.  [PubMed]  [DOI]  [Cited in This Article: ]
147.  Ando T, Yoshida T, Enomoto S, Asada K, Tatematsu M, Ichinose M, Sugiyama T, Ushijima T. DNA methylation of microRNA genes in gastric mucosae of gastric cancer patients: its possible involvement in the formation of epigenetic field defect. Int J Cancer. 2009;124:2367-2374.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 211]  [Cited by in F6Publishing: 215]  [Article Influence: 14.3]  [Reference Citation Analysis (0)]
148.  Tsukamoto Y, Nakada C, Noguchi T, Tanigawa M, Nguyen LT, Uchida T, Hijiya N, Matsuura K, Fujioka T, Seto M. MicroRNA-375 is downregulated in gastric carcinomas and regulates cell survival by targeting PDK1 and 14-3-3zeta. Cancer Res. 2010;70:2339-2349.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 320]  [Cited by in F6Publishing: 341]  [Article Influence: 24.4]  [Reference Citation Analysis (0)]
149.  Tsai KW, Wu CW, Hu LY, Li SC, Liao YL, Lai CH, Kao HW, Fang WL, Huang KH, Chan WC. Epigenetic regulation of miR-34b and miR-129 expression in gastric cancer. Int J Cancer. 2011;129:2600-2610.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 141]  [Cited by in F6Publishing: 151]  [Article Influence: 11.6]  [Reference Citation Analysis (0)]
150.  Maekita T, Nakazawa K, Mihara M, Nakajima T, Yanaoka K, Iguchi M, Arii K, Kaneda A, Tsukamoto T, Tatematsu M. High levels of aberrant DNA methylation in Helicobacter pylori-infected gastric mucosae and its possible association with gastric cancer risk. Clin Cancer Res. 2006;12:989-995.  [PubMed]  [DOI]  [Cited in This Article: ]
151.  Kalimutho M, Di Cecilia S, Del Vecchio Blanco G, Roviello F, Sileri P, Cretella M, Formosa A, Corso G, Marrelli D, Pallone F. Epigenetically silenced miR-34b/c as a novel faecal-based screening marker for colorectal cancer. Br J Cancer. 2011;104:1770-1778.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 76]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]