Topic Highlight Open Access
Copyright ©The Author(s) 2016. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Aug 7, 2016; 22(29): 6619-6628
Published online Aug 7, 2016. doi: 10.3748/wjg.v22.i29.6619
Inflammatory microenvironment contributes to epithelial-mesenchymal transition in gastric cancer
Hui-Ying Ma, Xin-Zhou Liu, Chun-Min Liang, Lab of Tumor Immunology, Department of Anatomy and Histology and Embryology, Shanghai Medical College of Fudan University, Shanghai 200032, China
Author contributions: Ma HY and Liu XZ collected data and drafted the manuscript; Liang CM supervised and revised the manuscript.
Supported by National Science Foundation of China, No. 31471147.
Conflict-of-interest statement: The authors certify that there is no actual or potential conflict of interest and publication copyright in relation to this article.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Chun-Min Liang, MD, PhD, Lab of Tumor Immunology, Department of Anatomy and Histology and Embryology, Shanghai Medical College of Fudan University, 138 Yixueyuan Road, Shanghai 200032, China. cmliang@fudan.edu.cn
Telephone: +86-21-54237027 Fax: +86-21-54237027
Received: March 28, 2016
Peer-review started: March 28, 2016
First decision: May 30, 2016
Revised: June 12, 2016
Accepted: July 6, 2016
Article in press: July 6, 2016
Published online: August 7, 2016

Abstract

Gastric cancer (GC) is the fifth most common malignancy in the world. The major cause of GC is chronic infection with Helicobacter pylori (H. pylori). Infection with H. pylori leads to an active inflammatory microenvironment that is maintained by immune cells such as T cells, macrophages, natural killer cells, among other cells. Immune cell dysfunction allows the initiation and accumulation of mutations in GC cells, inducing aberrant proliferation and protection from apoptosis. Meanwhile, immune cells can secrete certain signals, including cytokines, and chemokines, to alter intracellular signaling pathways in GC cells. Thus, GC cells obtain the ability to metastasize to lymph nodes by undergoing the epithelial-mesenchymal transition (EMT), whereby epithelial cells lose their epithelial attributes and acquire a mesenchymal cell phenotype. Metastasis is a leading cause of death for GC patients, and the involved mechanisms are still under investigation. In this review, we summarize the current research on how the inflammatory environment affects GC initiation and metastasis via EMT.

Key Words: Gastric cancer, Inflammation, Epithelial-mesenchymal transition, Microenvironment, Immune cells

Core tip: The major cause of gastric cancer (GC) is Helicobacter pylori infection, resulting in an inflammatory microenvironment in GC. Meanwhile, the leading cause of death for GC patients is metastasis. The major pathway for metastasis is the epithelial-mesenchymal transition (EMT). Therefore, a thorough understanding of how the inflammatory microenvironment contributes to the promotion of the EMT is indispensable for developing new treatments. In this review, we summarize the mechanisms of inflammatory mediators, divided among immune cells and molecules, on the prognosis of GC patients and EMT, which suggests that a combination of immunotherapy and anti-EMT treatments may be encouraging for the treatment of GC.



INTRODUCTION

Gastric cancer (GC) was the fifth most common malignancy and the third leading cause of cancer worldwide in 2012. Almost one million new cases were estimated to have occurred annually. More than 70% of these occurred in developing countries, and approximately half of all cases worldwide (405000 cases) were diagnosed in China[1]. Two histologically distinct types of GC have described: diffuse-type, in which infiltrating neoplastic cells exists individually, and intestinal-type, which initiates from normal mucosa, transiting to chronic superficial gastritis, atrophic gastritis, intestinal metaplasia, and finally to dysplasia and adenocarcinoma[2]. In recent years, improvements in endoscopic detection and treatment strategies such as surgical resection and chemotherapy have contributed to 5-year survival rates of approximately 60% in Japan[3]. However, despite multimodal therapy, the average overall 5-year survival worldwide still remain at 40%[4,5], while in the United States, the 5-year survival rate is only 26%[6], and more than 60% of patients will develop local relapse or metastatic disease[4]. Therefore, investigating the mechanisms underlying the initiation and progression of GC will help improve early detection and treatment efficiency.

GC is the result of the accumulation of genomic damage that affects cellular functions essential for cancer development[7]. The major cause of GC is chronic infection with the Gram-negative bacterium Helicobacter pylori (H. pylori), which contributes to more than 75% of GC cases[8]. Although in the past, H. pylori infection has been regarded as a risk factor for GC and is categorized as a Group 1 carcinogen for humans[9-11], only a small number of infected individuals develop GC (approximately 2%-3% of the total infected individuals)[12], which makes H. pylori status an unclear predictor of GC prognosis. Some studies have yielded contrasting findings, showing that GC patients with positive H. pylori infection have better disease-free survival and overall survival, whereas negative H. pylori infection indicates poor prognosis in GC patients[13,14]. Recent meta-analyses further showed that instead of serving as a risk factor, H. pylori status could act as a protective factor in predicting GC progression[15], leading to further confusion. Another key factor leading to approximately 10% of GC cases is Epstein-Barr virus (EBV) infection[16]. A meta-analysis of 13 studies showed that EBV-positive patients have decreased survival, which indicates that EBV might serve as a predictive factor[17]. However, studies on the role of EBV are still in their infancy. Although the relationship between H. pylori or EBV and the prognosis of GC patients is unclear, the fact remains that these infections can induce physiological and morphological changes within the gastric epithelium, resulting in an increased risk of neoplastic transformations such as hypochlorhydria and gastric atrophy, which are precursors of GC. The induced inflammatory microenvironment recruits more immune cells that secrete aberrant factors such TGF-β, which may further lead to tumor cell metastasis, which is a major factor in the poor survival of GC patients[18].

METASTASIS AND EPITHELIAL-MESENCHYMAL TRANSITION

A key process in promoting tumor cells metastasis is the epithelial-mesenchymal transition (EMT), which is a process by which epithelial cells lose their epithelial attributes and acquire a mesenchymal cell phenotype[19,20]. During this process, epithelial tumor cells are endowed with three main changes. First, cell morphology changes from a cobblestone-like monolayer of epithelial cells with apical-basal polarity to spindle-shaped mesenchymal cells with migratory pseudopodia or filopodia structures. Second, the cytoskeleton and intercellular junctions are reorganized with changes in differentiation markers such as the loss of E-cadherin and increased expression of Vimentin and Fibronectin. Third, functional changes are shown to potentiate angiogenesis and intrastation through enhanced protease expression, allowing invasion through the extracellular matrix (ECM)[21,22]. EMT also increases resistance to apoptosis and contributes to the survival of circulating tumor cells[23]. Not all of these changes are invariably observed during EMT; however, the ability to migrate and invade the ECM as a single cell is regarded as marking the functional completion of the EMT program.

The development of EMT involves many different signaling pathways. Transforming growth factor-β (TGF-β) is recognized as a potent inducer of the EMT, acting at translational, post-translational, transcriptional and post-transcriptional levels[24]. After signal binds to TGF-β receptors, the EMT is initiated by either the phosphorylation of Smad2/3/4, which induces the transcription of Snail or Slug[25,26], or through non-Smad signaling pathways, including the PI3K/Akt-mTOR pathway[27,28], the RHO-GTPase pathway[29,30], and the ERK, p38 and JUN N-terminal kinase (JNK) MAPK pathways[31-33]. Aside from TGF-β receptors, receptor tyrosine kinases also contribute to the initiation of the EMT through the activation of the PI3K/Akt and ERK signaling pathways, which promotes cell mobility and invasive behavior[34-36]. Some studies have also found that the frizzled receptor, Notch receptor, and patched (PTC) receptors as well as the IL-6 receptor can participate in EMT progression by activating the Wnt and STAT signaling, among other pathways[37-40]. To activate these pathways, ligands must first bind to membrane receptors, for example, cytokines found in the GC microenvironment, such as TGF-β1 and IL-1β. These inflammatory cytokines are thought to be released from recruited immune cells, endothelial cells and fibroblasts[41], indicating that members of the microenvironment regulate EMT progression.

INFLAMMATION AND EMT IN GC

The connection between inflammation and cancer has been studied for years, and chronic inflammation is thought to be a key contributor to tumor development[42]. Chronic inflammation is a protective response to damage to tissue homeostasis, inducing a prolonged, aberrant form called a “wound”[43]. The so-called “wound” continuously recruits immune cells and other protective cells and induces their secretion of inflammatory mediators. During this state, despite the excessive mediators, the damaged cells will amplify and divide frequently, leading the microenvironment to become oxidative and thus increasing the likelihood of DNA damage and mutations. Once the key damage or mutation occurs, the damaged cells start to secret pro-inflammatory cytokines to keep them active and revert themselves to tumor cells. Meanwhile, these cells manage to escape from immune surveillance and modify infiltrating immune cells into tumor-associated immune cells, which assists tumor progression rather than immune inhibition. These changes result in a “wound” that never heals and promote tumor initiation, progression and metastasis[44-47]. Given that more than 85% of GC is caused by infection, which induces inflammation, inflammation is accepted as a major diver of gastric carcinogenesis[48,49].

The tumor-associated microenvironment is characterized by tumor infiltrating lymphocytes (TILs), the secretion of inflammatory mediators and angiogenesis. TILs interact with tumor cells via inflammatory molecules such as cytokines (TGF-β, TNF-α, IL-6, IL-1β), chemokines (CC- and CXC- receptors) and matrix metalloproteinases (MMPs), which form an inflammatory network[42]. Unfortunately, these molecules are also inducers of the EMT (Table 1), which may explain how inflammation contributes to GC cells metastasis. Upon infection by H. pylori, the level of soluble HB-EGF shedding is up-regulated, which further induces GC cells to undergo the EMT. This process partially relies on the expression of gastrin and MMP7[50,51]. GC EMT could also be induced by H. pylori cytotoxin-associated gene A (CagA), which downregulates E-cadherin expression and increases the expression of vimentin and twist[52]. Eradication of H. pylori reduces the expression of TGF-β1 while increasing E-cadherin expression, indicating that H. pylori may trigger TGF-β1-induced EMT[53]. The development and metastasis of tumor cells may occur because GC cells escape immune surveillance or because immune cells become helpers for GC cells. Therefore, the tumor-related inflammatory microenvironment has an important role in regulating GC EMT, mainly through interactions with infiltrating immune cells.

Table 1 Important inflammatory mediators involved in the EMT in GC.
CategoriesFactorsRef
CytokinesTGF-β1[103,104]
TNF-α[99,100]
TGF-α[108]
IL-6[109,110]
IL-8[102]
Chemokines and receptorsCCL5[84,131]
CCL18[132,133]
CCR2[134]
CCR7[135]
CCL20-CCR6[136,137]
CXCR1[138,139]
CXCR3[140]
CXCL12-CXCR4[108,117]
Immune cellsCD8+ cytotoxic T cells[56,60]
Th1/Th2 cells ratio[63]
Th17 cells[64,65,67]
Foxp3+ Tregs[68-70]
Foxp3+/CD4+ ratio[71]
Foxp3+/CD8+ ratio[68]
NK cells[87-89]
TAMs[78,79]
DCs[93-95]
TAM/Foxp3+ ratio[141]
MMPsMMP-2[125]
MMP-9[124-126,142]
MMP-7[51,127,128]
Immune cells and EMT

The major infiltrating functional immune cells in GC are T cells, macrophages, NK cells, DCs and MDSCs[54].

T cells are mainly divided into CD8+ cytotoxic T cells and CD4+ T helper cells[55]. CD8+ cytotoxic T cells (CTLs) exert active antitumor effects, and previous work has shown that GC patients with high CD8+ CTL infiltration display better prognoses[56]. However, other work has shown that higher CD8+ CTLs do not indicate good outcomes with metastasis due to the occurrence of adaptive immune resistance, such as the ratio of CD8+ CTLs with programmed death-ligand 1 (PD-L1)[57]. Meanwhile, CTLs can also produce IL-17 to promote inflammation and result in a poor prognosis[58]. EBV-specific CD8+ CTL injection significantly reduced tumor growth and metastasis in mouse models of GC[59]. Thus, treatment with autologous CD8+ CTL injection for GC patients and patients with metastatic GC seems promising[60].

Naïve CD4+ T helper cells can differentiate into several subsets, including Th1, Th2, Treg, and Th17, by secreting various cytokines such as TGF-β, IL-10, and IFN-γ, which are also inducers of the EMT[55,61]. CD4+ T cell subsets are found at significantly lower levels in metastatic tumor draining lymph nodes (TDLNs) than in metastasis-free TDLNs, which indicates that metastasis is a consequence of the loss of CD4+ T cells[62]. Th1 (IFN-γ producing) and Th2 (IL-4 producing) cells play key roles in anti-tumor immunity. The balance between these two cell types can alter antitumor activity, as shown in human peripheral blood: a high Th1/Th2 ratio correlates with a better prognosis and less metastasis[63]. An expansion of Th17 cells is found in GC patients’ tissues and peripheral blood, especially in patients with metastasis. High levels of IL-1β, IL-21, IL-17 and TGF-β expression are also observed, which will induce macrophages to produce more IL-6 and IL-8 to activate the NF-κB pathway and might be a reason why metastasis occurs through the induction of the EMT[64-67]. Another important CD4+ T cell subset related to GC progression at CD4+ suppressor T lymphocytes, or Tregs, that express Foxp3. Higher Foxp3+ Treg infiltration is correlated with GC metastasis and poor prognosis[68-70]. Similar to Th1 and Th2, the ratio of Foxp3+/CD4+ and Foxp3+/CD8+ cells is very important for the suppression of metastasis[68,71].

Macrophages are among the most important immune cells that infiltrate the tumor microenvironment and include the following two phenotypes: M1 macrophages, which facilitate anti-tumor activity, and M2 macrophages, or tumor-associated macrophages (TAMs), which promote tumor progression[72]. Although macrophages can secrete cytokines such as IL-25 to hamper tumor growth and metastasis, large amounts of infiltration by TAMs disrupt this process[73]. TAM infiltration in GC can promote angiogenesis and lymphangiogenesis and predict poor overall survival[74-76]; hence, TAMs are regarded as a promising therapeutic target[77]. When TAMs are co-cultured with GC cells, the metastatic ability of GC cells increases, which might be the result of TGF-β1 secretion activating the TGF-β and NF-κB signaling pathways[78,79]. IL-8, which is secreted by surrounding TAMs, could also be an inducer of GC cell metastasis, especially under hypoxic conditions[80-82]. Meanwhile, chemokine factors can affect the relation between TAMs and GC cells. High CXCL12 expression on GC cells can recruit TAMs[83]. Recruited TAMs then secret CCL5, which activates the STAT3 signaling pathway, leading to tumor growth and invasion[84]. Activation of the NF-κB or STAT3 signaling pathway can elevate the expression of certain proteins related to mesenchymal phenotypes, such as Vimentin. In this way, GC cells start to undergo the EMT, which ultimately assists in metastasis[85,86].

NK cells play an important role in regulating GC development and metastasis by directly clearing tumor cells. Previous studies found that in GC patients, the expression of NKG2D, an activating receptor specifically expressed on NK cells, is higher compared with healthy controls, with the same trend observed when comparing GC patients with and without lymph node metastasis[87,88]. This NK cell dysfunction may be related to TGF-β1 levels[89]. These groups of immune cells are unable to inhibit GC progression mainly due to their loss or dysfunction.

DCs are the cells that process and present antigens to T cells[90]. However, their numbers still make a difference in controlling GC progression. Patients with lower DC infiltration have less lymph node metastasis and show a favorable prognosis[91-93]. This effect might due to the elevated IL-1β expression and decreased IL-10 expression produced by DCs through the activation of the NF-κB signaling pathway[94,95], which further affects the metastatic ability of GC cells. MDSCs are a relatively heterogeneous population of cells. Their expansion during cancer is associated with advanced GC stages and indicates poor prognosis[96,97]. However, studies of MDSC function in GC are still very limited.

Inflammatory molecules and EMT

Inflammatory mediators are factors that act directly on tumor cells and are secreted by both GC cells and infiltrating cells in the surrounding microenvironment. These mediators can be divided into three groups: cytokines, chemokines and MMPs.

Cytokines can be secreted by all constituents of the tumor microenvironment and appears to modify the EMT of GC cells, including TNF-α, IL-8, TGFβ, TGF-α, and IL-6[98]. TNF-α levels are increased by TNF-α-inducing protein (Tipα), which is released by H. pylori. The binding of Tipα to its membrane receptor activates the NF-κB signaling pathway, resulting in the transcription of TNF-α, which further increases the expression of N-cadherin and vimentin to enable GC cell migration and metastasis[99-101]. Increased of IL-8 levels promote the EMT in GC cells at early stages of GC progression through the activation of the NF-κB pathway[102]. TGFβ is the most potent and common inducer of the EMT. High TGF-β1 expression indicates poor prognosis in GC patients and is related to lymph node metastasis through the activation of the TGF-β signaling pathway[103,104]. Inhibition of this pathway can inhibit EMT-mediated migration and invasion[105-107]. TGF-α is involved in the EMT and is associated with poor OS in GC patients[108]. IL-6 can rescue GC cell resistance to anti-tumor drugs and EMT by activating the STAT3 pathway[109,110].

Chemokines are a group of secreted proteins that are produced in response to pro-inflammatory stimuli and most commonly participate in the chemotaxis of leukocyte trafficking and positioning. Current studies show that chemokines are also involved in tumor growth, angiogenesis, EMT, metastasis and immune evasion[111-113]. The two most important chemokine receptors in GC are CXCR4 and CCR7. CXCR4 expression is associated with aggressive tumor behaviors such as invasion and metastasis[114]. After binding its ligand CXCL12, actin polymerization is activated to induce cell motility and the EMT[108,115,116]. The CXCL12-CXCR4 axis alters the migratory and invasive ability of GC cells by upregulating the expression of MMP-2 and MMP-7 to assist EMT progression[108,117]. Meanwhile, CXCL12 can recruit myeloid-derived suppressor cells (MDSCs) into the tumor microenvironment to promote the progression of gastric cancer[58]. CCR7 is another important chemokine receptor in the progression of GC. CCR7 is associated with lymph node metastasis in GC patients[70,118]. Through the activation of CCR7 signaling or the TGF-β1 signaling pathway, GC cells initiate the EMT by altering the expression of E-cadherin, MMP-9, and Snail, which enable them to metastasize, and led by CCR7, they metastasize toward lymph vessels, which is why GC cells metastasize to lymph nodes[119,120].

After infection by pathogens such as H. pylori, the expression of matrix metalloproteinase (MMP) family is upregulated because the pathogens need to secrete proteins to assist their adherence to epithelial gastric cells[121,122]. The MMP family is among the most important inducers of the EMT through the degradation of the extracellular matrix (ECM) and basement membrane barriers[123]. Increased expression of MMP-2 and MMP-9 is reported to enhance the invasion ability of GC cells and correlates with metastatic GC[124-126]. Elevated MMP-7 levels can be used as a biomarker for H. pylori-related GC and potentially regulate the progression of GC through the EMT[51,127,128]. MMP7-/- mice infected with H. pylori show increased levels of M1 macrophages, which enhance the inflammatory response[129,130]. However, the precise mechanism of how MMPs regulate the EMT of GC needs to be clarified in the future.

CONCLUSION

The progression of GC is mainly caused by microbial pathogens and is closely related to host inflammatory factors. The inflammatory microenvironment enables the host immune system to not only combat pathogens but also to secrete cytokines to stimulate normal gastric epithelial cells to protect themselves. During this process, the altered microenvironment may cause random mutations to occur in gastric cells. Once these mutations accumulate to a certain level, the process will continue without restoring normal homeostasis. Thus, the infection starts to become an adenoma followed by a carcinoma. Meanwhile, in the gastric cancer microenvironment, the aberrant secretion by immune cells might lead to dysfunction and also stimulate GC cells to become resistant. In this way, GC cells are likely to gain the ability to continuously proliferate, become protected from apoptosis and escape immune surveillance. Through alterations in their signaling pathways, GC cells begin to translate more mesenchymal proteins such as MMP and vimentin, allowing them to migrate and invade into the blood and lymph vessels to metastasize, otherwise known as the EMT. Current studies mainly focus on the immune cells and GC prognosis and the effects on metastasis. However, studies on the mechanisms by which immune cells alter GC cells undergoing the EMT in the inflammatory microenvironment are still very limited. As long as GC metastasis is a major cause of death, targeting the EMT combined with immunotherapy shows promising results for the treatment of GC in the future.

ACKNOWLEDGMENTS

The authors thank Jessie Yang for assistance in recording the audio core tip.

Footnotes

Manuscript source: Invited manuscript

Specialty type: Gastroenterology and hepatology

Country of origin: China

Peer-review report classification

Grade A (Excellent): 0

Grade B (Very good): B

Grade C (Good): C, C

Grade D (Fair): 0

Grade E (Poor): 0

P- Reviewer: Bi J, Cao XC, Wang WH S- Editor: Yu J L- Editor: A E- Editor: Ma S

References
1.  Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A. Global cancer statistics, 2012. CA Cancer J Clin. 2015;65:87-108.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18694]  [Cited by in F6Publishing: 20810]  [Article Influence: 2312.2]  [Reference Citation Analysis (1)]
2.  Correa P. Human gastric carcinogenesis: a multistep and multifactorial process--First American Cancer Society Award Lecture on Cancer Epidemiology and Prevention. Cancer Res. 1992;52:6735-6740.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Kim HS, Lee H, Jeung HC, Noh SH, Chung HC, Roh JK, Nam CM, Rha SY. Advanced detection of recent changing trends in gastric cancer survival: up-to-date comparison by period analysis. Jpn J Clin Oncol. 2011;41:1344-1350.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 13]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
4.  Cunningham D, Allum WH, Stenning SP, Thompson JN, Van de Velde CJ, Nicolson M, Scarffe JH, Lofts FJ, Falk SJ, Iveson TJ. Perioperative chemotherapy versus surgery alone for resectable gastroesophageal cancer. N Engl J Med. 2006;355:11-20.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4120]  [Cited by in F6Publishing: 4221]  [Article Influence: 234.5]  [Reference Citation Analysis (0)]
5.  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: 2362]  [Article Influence: 102.7]  [Reference Citation Analysis (0)]
6.  Siegel R, Naishadham D, Jemal A. Cancer statistics, 2013. CA Cancer J Clin. 2013;63:11-30.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9215]  [Cited by in F6Publishing: 9710]  [Article Influence: 882.7]  [Reference Citation Analysis (3)]
7.  Guggenheim DE, Shah MA. Gastric cancer epidemiology and risk factors. J Surg Oncol. 2013;107:230-236.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 296]  [Cited by in F6Publishing: 350]  [Article Influence: 29.2]  [Reference Citation Analysis (0)]
8.  Herrera V, Parsonnet J. Helicobacter pylori and gastric adenocarcinoma. Clin Microbiol Infect. 2009;15:971-976.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 154]  [Cited by in F6Publishing: 172]  [Article Influence: 12.3]  [Reference Citation Analysis (0)]
9.  Forman D, Newell DG, Fullerton F, Yarnell JW, Stacey AR, Wald N, Sitas F. Association between infection with Helicobacter pylori and risk of gastric cancer: evidence from a prospective investigation. BMJ. 1991;302:1302-1305.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Helicobacter and Cancer Collaborative Group. Gastric cancer and Helicobacter pylori: a combined analysis of 12 case control studies nested within prospective cohorts. Gut. 2001;49:347-353.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Parsonnet J, Friedman GD, Vandersteen DP, Chang Y, Vogelman JH, Orentreich N, Sibley RK. Helicobacter pylori infection and the risk of gastric carcinoma. N Engl J Med. 1991;325:1127-1131.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2805]  [Cited by in F6Publishing: 2664]  [Article Influence: 80.7]  [Reference Citation Analysis (0)]
12.  Conteduca V, Sansonno D, Lauletta G, Russi S, Ingravallo G, Dammacco F. H. pylori infection and gastric cancer: state of the art (review). Int J Oncol. 2013;42:5-18.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 135]  [Cited by in F6Publishing: 147]  [Article Influence: 12.3]  [Reference Citation Analysis (0)]
13.  Meimarakis G, Winter H, Assmann I, Kopp R, Lehn N, Kist M, Stolte M, Jauch KW, Hatz RA. Helicobacter pylori as a prognostic indicator after curative resection of gastric carcinoma: a prospective study. Lancet Oncol. 2006;7:211-222.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 73]  [Cited by in F6Publishing: 79]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
14.  Marrelli D, Pedrazzani C, Berardi A, Corso G, Neri A, Garosi L, Vindigni C, Santucci A, Figura N, Roviello F. Negative Helicobacter pylori status is associated with poor prognosis in patients with gastric cancer. Cancer. 2009;115:2071-2080.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 73]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
15.  Wang F, Sun G, Zou Y, Zhong F, Ma T, Li X. Protective role of Helicobacter pylori infection in prognosis of gastric cancer: evidence from 2,454 patients with gastric cancer. PLoS One. 2013;8:e62440.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 23]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
16.  Iizasa H, Nanbo A, Nishikawa J, Jinushi M, Yoshiyama H. Epstein-Barr Virus (EBV)-associated gastric carcinoma. Viruses. 2012;4:3420-3439.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Camargo MC, Kim WH, Chiaravalli AM, Kim KM, Corvalan AH, Matsuo K, Yu J, Sung JJ, Herrera-Goepfert R, Meneses-Gonzalez F. Improved survival of gastric cancer with tumour Epstein-Barr virus positivity: an international pooled analysis. Gut. 2014;63:236-243.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 279]  [Cited by in F6Publishing: 271]  [Article Influence: 27.1]  [Reference Citation Analysis (0)]
18.  Li N, Xie C, Lu NH. Transforming growth factor-β: an important mediator in Helicobacter pylori-associated pathogenesis. Front Cell Infect Microbiol. 2015;5:77.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 19]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
19.  Thiery JP. Epithelial-mesenchymal transitions in tumour progression. Nat Rev Cancer. 2002;2:442-454.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4877]  [Cited by in F6Publishing: 4952]  [Article Influence: 225.1]  [Reference Citation Analysis (0)]
20.  Greenburg G, Hay ED. Epithelia suspended in collagen gels can lose polarity and express characteristics of migrating mesenchymal cells. J Cell Biol. 1982;95:333-339.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Boyer B, Thiery JP. Epithelium-mesenchyme interconversion as example of epithelial plasticity. APMIS. 1993;101:257-268.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Hay ED. An overview of epithelio-mesenchymal transformation. Acta Anat (Basel). 1995;154:8-20.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Thiery JP, Acloque H, Huang RY, Nieto MA. Epithelial-mesenchymal transitions in development and disease. Cell. 2009;139:871-890.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6805]  [Cited by in F6Publishing: 7342]  [Article Influence: 489.5]  [Reference Citation Analysis (0)]
24.  Lamouille S, Xu J, Derynck R. Molecular mechanisms of epithelial-mesenchymal transition. Nat Rev Mol Cell Biol. 2014;15:178-196.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4715]  [Cited by in F6Publishing: 5563]  [Article Influence: 556.3]  [Reference Citation Analysis (0)]
25.  Mani SA, Guo W, Liao MJ, Eaton EN, Ayyanan A, Zhou AY, Brooks M, Reinhard F, Zhang CC, Shipitsin M. The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell. 2008;133:704-715.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6211]  [Cited by in F6Publishing: 6520]  [Article Influence: 407.5]  [Reference Citation Analysis (0)]
26.  Feng XH, Derynck R. Specificity and versatility in tgf-beta signaling through Smads. Annu Rev Cell Dev Biol. 2005;21:659-693.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1427]  [Cited by in F6Publishing: 1459]  [Article Influence: 76.8]  [Reference Citation Analysis (0)]
27.  Lamouille S, Connolly E, Smyth JW, Akhurst RJ, Derynck R. TGF-β-induced activation of mTOR complex 2 drives epithelial-mesenchymal transition and cell invasion. J Cell Sci. 2012;125:1259-1273.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 211]  [Cited by in F6Publishing: 243]  [Article Influence: 20.3]  [Reference Citation Analysis (0)]
28.  Lamouille S, Derynck R. Cell size and invasion in TGF-beta-induced epithelial to mesenchymal transition is regulated by activation of the mTOR pathway. J Cell Biol. 2007;178:437-451.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 420]  [Cited by in F6Publishing: 451]  [Article Influence: 26.5]  [Reference Citation Analysis (0)]
29.  Bhowmick NA, Ghiassi M, Bakin A, Aakre M, Lundquist CA, Engel ME, Arteaga CL, Moses HL. Transforming growth factor-beta1 mediates epithelial to mesenchymal transdifferentiation through a RhoA-dependent mechanism. Mol Biol Cell. 2001;12:27-36.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Tsapara A, Luthert P, Greenwood J, Hill CS, Matter K, Balda MS. The RhoA activator GEF-H1/Lfc is a transforming growth factor-beta target gene and effector that regulates alpha-smooth muscle actin expression and cell migration. Mol Biol Cell. 2010;21:860-870.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 72]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
31.  Yamashita M, Fatyol K, Jin C, Wang X, Liu Z, Zhang YE. TRAF6 mediates Smad-independent activation of JNK and p38 by TGF-beta. Mol Cell. 2008;31:918-924.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 408]  [Cited by in F6Publishing: 442]  [Article Influence: 27.6]  [Reference Citation Analysis (0)]
32.  Xie L, Law BK, Chytil AM, Brown KA, Aakre ME, Moses HL. Activation of the Erk pathway is required for TGF-beta1-induced EMT in vitro. Neoplasia. 2004;6:603-610.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 366]  [Cited by in F6Publishing: 406]  [Article Influence: 21.4]  [Reference Citation Analysis (0)]
33.  Marchetti A, Colletti M, Cozzolino AM, Steindler C, Lunadei M, Mancone C, Tripodi M. ERK5/MAPK is activated by TGFbeta in hepatocytes and required for the GSK-3beta-mediated Snail protein stabilization. Cell Signal. 2008;20:2113-2118.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 37]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
34.  Doehn U, Hauge C, Frank SR, Jensen CJ, Duda K, Nielsen JV, Cohen MS, Johansen JV, Winther BR, Lund LR. RSK is a principal effector of the RAS-ERK pathway for eliciting a coordinate promotile/invasive gene program and phenotype in epithelial cells. Mol Cell. 2009;35:511-522.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 159]  [Cited by in F6Publishing: 175]  [Article Influence: 11.7]  [Reference Citation Analysis (0)]
35.  Graham TR, Zhau HE, Odero-Marah VA, Osunkoya AO, Kimbro KS, Tighiouart M, Liu T, Simons JW, O’Regan RM. Insulin-like growth factor-I-dependent up-regulation of ZEB1 drives epithelial-to-mesenchymal transition in human prostate cancer cells. Cancer Res. 2008;68:2479-2488.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 265]  [Cited by in F6Publishing: 277]  [Article Influence: 17.3]  [Reference Citation Analysis (0)]
36.  Lo HW, Hsu SC, Xia W, Cao X, Shih JY, Wei Y, Abbruzzese JL, Hortobagyi GN, Hung MC. Epidermal growth factor receptor cooperates with signal transducer and activator of transcription 3 to induce epithelial-mesenchymal transition in cancer cells via up-regulation of TWIST gene expression. Cancer Res. 2007;67:9066-9076.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 480]  [Cited by in F6Publishing: 506]  [Article Influence: 29.8]  [Reference Citation Analysis (0)]
37.  Brabletz T, Jung A, Reu S, Porzner M, Hlubek F, Kunz-Schughart LA, Knuechel R, Kirchner T. Variable beta-catenin expression in colorectal cancers indicates tumor progression driven by the tumor environment. Proc Natl Acad Sci USA. 2001;98:10356-10361.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 807]  [Cited by in F6Publishing: 833]  [Article Influence: 36.2]  [Reference Citation Analysis (0)]
38.  Li X, Deng W, Nail CD, Bailey SK, Kraus MH, Ruppert JM, Lobo-Ruppert SM. Snail induction is an early response to Gli1 that determines the efficiency of epithelial transformation. Oncogene. 2006;25:609-621.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 138]  [Cited by in F6Publishing: 149]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
39.  Xie M, Zhang L, He CS, Xu F, Liu JL, Hu ZH, Zhao LP, Tian Y. Activation of Notch-1 enhances epithelial-mesenchymal transition in gefitinib-acquired resistant lung cancer cells. J Cell Biochem. 2012;113:1501-1513.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 85]  [Article Influence: 7.1]  [Reference Citation Analysis (0)]
40.  Sullivan NJ, Sasser AK, Axel AE, Vesuna F, Raman V, Ramirez N, Oberyszyn TM, Hall BM. Interleukin-6 induces an epithelial-mesenchymal transition phenotype in human breast cancer cells. Oncogene. 2009;28:2940-2947.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 510]  [Cited by in F6Publishing: 548]  [Article Influence: 36.5]  [Reference Citation Analysis (0)]
41.  Hanahan D, Coussens LM. Accessories to the crime: functions of cells recruited to the tumor microenvironment. Cancer Cell. 2012;21:309-322.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2921]  [Cited by in F6Publishing: 3020]  [Article Influence: 251.7]  [Reference Citation Analysis (0)]
42.  Elinav E, Nowarski R, Thaiss CA, Hu B, Jin C, Flavell RA. Inflammation-induced cancer: crosstalk between tumours, immune cells and microorganisms. Nat Rev Cancer. 2013;13:759-771.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1198]  [Cited by in F6Publishing: 1304]  [Article Influence: 118.5]  [Reference Citation Analysis (0)]
43.  Dvorak HF. Tumors: wounds that do not heal. Similarities between tumor stroma generation and wound healing. N Engl J Med. 1986;315:1650-1659.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3033]  [Cited by in F6Publishing: 2969]  [Article Influence: 78.1]  [Reference Citation Analysis (0)]
44.  Medzhitov R. Origin and physiological roles of inflammation. Nature. 2008;454:428-435.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3548]  [Cited by in F6Publishing: 3856]  [Article Influence: 241.0]  [Reference Citation Analysis (0)]
45.  Coussens LM, Werb Z. Inflammation and cancer. Nature. 2002;420:860-867.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10123]  [Cited by in F6Publishing: 10542]  [Article Influence: 479.2]  [Reference Citation Analysis (0)]
46.  de Visser KE, Eichten A, Coussens LM. Paradoxical roles of the immune system during cancer development. Nat Rev Cancer. 2006;6:24-37.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1633]  [Cited by in F6Publishing: 1589]  [Article Influence: 88.3]  [Reference Citation Analysis (0)]
47.  Virchow R. An Address on the Value of Pathological Experiments. Br Med J. 1881;2:198-203.  [PubMed]  [DOI]  [Cited in This Article: ]
48.  Hardbower DM, de Sablet T, Chaturvedi R, Wilson KT. Chronic inflammation and oxidative stress: the smoking gun for Helicobacter pylori-induced gastric cancer? Gut Microbes. 2013;4:475-481.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 93]  [Cited by in F6Publishing: 86]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
49.  Wang F, Meng W, Wang B, Qiao L. Helicobacter pylori-induced gastric inflammation and gastric cancer. Cancer Lett. 2014;345:196-202.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 375]  [Cited by in F6Publishing: 478]  [Article Influence: 47.8]  [Reference Citation Analysis (1)]
50.  McCaig C, Duval C, Hemers E, Steele I, Pritchard DM, Przemeck S, Dimaline R, Ahmed S, Bodger K, Kerrigan DD. The role of matrix metalloproteinase-7 in redefining the gastric microenvironment in response to Helicobacter pylori. Gastroenterology. 2006;130:1754-1763.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 74]  [Cited by in F6Publishing: 77]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
51.  Yin Y, Grabowska AM, Clarke PA, Whelband E, Robinson K, Argent RH, Tobias A, Kumari R, Atherton JC, Watson SA. Helicobacter pylori potentiates epithelial: mesenchymal transition in gastric cancer: links to soluble HB-EGF, gastrin and matrix metalloproteinase-7. Gut. 2010;59:1037-1045.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 90]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
52.  Yu H, Zeng J, Liang X, Wang W, Zhou Y, Sun Y, Liu S, Li W, Chen C, Jia J. Helicobacter pylori promotes epithelial-mesenchymal transition in gastric cancer by downregulating programmed cell death protein 4 (PDCD4). PLoS One. 2014;9:e105306.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 47]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
53.  Choi YJ, Kim N, Chang H, Lee HS, Park SM, Park JH, Shin CM, Kim JM, Kim JS, Lee DH. Helicobacter pylori-induced epithelial-mesenchymal transition, a potential role of gastric cancer initiation and an emergence of stem cells. Carcinogenesis. 2015;36:553-563.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 64]  [Article Influence: 7.1]  [Reference Citation Analysis (0)]
54.  Diakos CI, Charles KA, McMillan DC, Clarke SJ. Cancer-related inflammation and treatment effectiveness. Lancet Oncol. 2014;15:e493-e503.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 951]  [Cited by in F6Publishing: 1334]  [Article Influence: 148.2]  [Reference Citation Analysis (0)]
55.  Subhash VV, Yeo MS, Tan WL, Yong WP. Strategies and Advancements in Harnessing the Immune System for Gastric Cancer Immunotherapy. J Immunol Res. 2015;2015:308574.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 27]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
56.  Lee HE, Chae SW, Lee YJ, Kim MA, Lee HS, Lee BL, Kim WH. Prognostic implications of type and density of tumour-infiltrating lymphocytes in gastric cancer. Br J Cancer. 2008;99:1704-1711.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 198]  [Cited by in F6Publishing: 229]  [Article Influence: 14.3]  [Reference Citation Analysis (0)]
57.  Thompson ED, Zahurak M, Murphy A, Cornish T, Cuka N, Abdelfatah E, Yang S, Duncan M, Ahuja N, Taube JM. Patterns of PD-L1 expression and CD8 T cell infiltration in gastric adenocarcinomas and associated immune stroma. Gut. 2016;.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 289]  [Cited by in F6Publishing: 326]  [Article Influence: 46.6]  [Reference Citation Analysis (0)]
58.  Zhuang Y, Peng LS, Zhao YL, Shi Y, Mao XH, Chen W, Pang KC, Liu XF, Liu T, Zhang JY. CD8(+) T cells that produce interleukin-17 regulate myeloid-derived suppressor cells and are associated with survival time of patients with gastric cancer. Gastroenterology. 2012;143:951-62.e8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 112]  [Cited by in F6Publishing: 123]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
59.  Choi BK, Lee SC, Lee MJ, Kim YH, Kim YW, Ryu KW, Lee JH, Shin SM, Lee SH, Suzuki S. 4-1BB-based isolation and expansion of CD8+ T cells specific for self-tumor and non-self-tumor antigens for adoptive T-cell therapy. J Immunother. 2014;37:225-236.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 22]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
60.  Turcotte S, Gros A, Tran E, Lee CC, Wunderlich JR, Robbins PF, Rosenberg SA. Tumor-reactive CD8+ T cells in metastatic gastrointestinal cancer refractory to chemotherapy. Clin Cancer Res. 2014;20:331-343.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 47]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
61.  Lee K, Hwang H, Nam KT. Immune response and the tumor microenvironment: how they communicate to regulate gastric cancer. Gut Liver. 2014;8:131-139.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 89]  [Cited by in F6Publishing: 107]  [Article Influence: 10.7]  [Reference Citation Analysis (0)]
62.  Okita Y, Ohira M, Tanaka H, Tokumoto M, Go Y, Sakurai K, Toyokawa T, Kubo N, Muguruma K, Sawada T. Alteration of CD4 T cell subsets in metastatic lymph nodes of human gastric cancer. Oncol Rep. 2015;34:639-647.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 12]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
63.  Ubukata H, Motohashi G, Tabuchi T, Nagata H, Konishi S, Tabuchi T. Evaluations of interferon-γ/interleukin-4 ratio and neutrophil/lymphocyte ratio as prognostic indicators in gastric cancer patients. J Surg Oncol. 2010;102:742-747.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 123]  [Cited by in F6Publishing: 131]  [Article Influence: 10.1]  [Reference Citation Analysis (0)]
64.  Liu T, Peng L, Yu P, Zhao Y, Shi Y, Mao X, Chen W, Cheng P, Wang T, Chen N. Increased circulating Th22 and Th17 cells are associated with tumor progression and patient survival in human gastric cancer. J Clin Immunol. 2012;32:1332-1339.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 77]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
65.  Su Z, Sun Y, Zhu H, Liu Y, Lin X, Shen H, Chen J, Xu W, Xu H. Th17 cell expansion in gastric cancer may contribute to cancer development and metastasis. Immunol Res. 2014;58:118-124.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 42]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
66.  Fossiez F, Djossou O, Chomarat P, Flores-Romo L, Ait-Yahia S, Maat C, Pin JJ, Garrone P, Garcia E, Saeland S. T cell interleukin-17 induces stromal cells to produce proinflammatory and hematopoietic cytokines. J Exp Med. 1996;183:2593-2603.  [PubMed]  [DOI]  [Cited in This Article: ]
67.  Bizama C, Benavente F, Salvatierra E, Gutiérrez-Moraga A, Espinoza JA, Fernández EA, Roa I, Mazzolini G, Sagredo EA, Gidekel M. The low-abundance transcriptome reveals novel biomarkers, specific intracellular pathways and targetable genes associated with advanced gastric cancer. Int J Cancer. 2014;134:755-764.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 20]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
68.  Shen Z, Zhou S, Wang Y, Li RL, Zhong C, Liang C, Sun Y. Higher intratumoral infiltrated Foxp3+ Treg numbers and Foxp3+/CD8+ ratio are associated with adverse prognosis in resectable gastric cancer. J Cancer Res Clin Oncol. 2010;136:1585-1595.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 143]  [Cited by in F6Publishing: 158]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
69.  Lee HE, Park DJ, Kim WH, Kim HH, Lee HS. High FOXP3+ regulatory T-cell density in the sentinel lymph node is associated with downstream non-sentinel lymph-node metastasis in gastric cancer. Br J Cancer. 2011;105:413-419.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 32]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
70.  Zhou S, Shen Z, Wang Y, Ma H, Xu S, Qin J, Chen L, Tao H, Zhen Z, Chen G. CCR7 expression and intratumoral FOXP3+ regulatory T cells are correlated with overall survival and lymph node metastasis in gastric cancer. PLoS One. 2013;8:e74430.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 36]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
71.  Kim HI, Kim H, Cho HW, Kim SY, Song KJ, Hyung WJ, Park CG, Kim CB. The ratio of intra-tumoral regulatory T cells (Foxp3+)/helper T cells (CD4+) is a prognostic factor and associated with recurrence pattern in gastric cardia cancer. J Surg Oncol. 2011;104:728-733.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 41]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
72.  Rojas A, Delgado-López F, Gonzalez I. Tumor-associated macrophages in gastric cancer: more than bystanders in tumor microenvironment. Gastric Cancer. 2016; Epub ahead of print.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 9]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
73.  Li J, Liao Y, Ding T, Wang B, Yu X, Chu Y, Xu J, Zheng L. Tumor-infiltrating macrophages express interleukin-25 and predict a favorable prognosis in patients with gastric cancer after radical resection. Oncotarget. 2016;7:11083-11093.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 11]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
74.  Ohta M, Kitadai Y, Tanaka S, Yoshihara M, Yasui W, Mukaida N, Haruma K, Chayama K. Monocyte chemoattractant protein-1 expression correlates with macrophage infiltration and tumor vascularity in human gastric carcinomas. Int J Oncol. 2003;22:773-778.  [PubMed]  [DOI]  [Cited in This Article: ]
75.  Wu H, Xu JB, He YL, Peng JJ, Zhang XH, Chen CQ, Li W, Cai SR. Tumor-associated macrophages promote angiogenesis and lymphangiogenesis of gastric cancer. J Surg Oncol. 2012;106:462-468.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 88]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
76.  Zhang H, Wang X, Shen Z, Xu J, Qin J, Sun Y. Infiltration of diametrically polarized macrophages predicts overall survival of patients with gastric cancer after surgical resection. Gastric Cancer. 2015;18:740-750.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 92]  [Cited by in F6Publishing: 99]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
77.  Yamaguchi T, Fushida S, Yamamoto Y, Tsukada T, Kinoshita J, Oyama K, Miyashita T, Tajima H, Ninomiya I, Munesue S. Tumor-associated macrophages of the M2 phenotype contribute to progression in gastric cancer with peritoneal dissemination. Gastric Cancer. 2015; Epub ahead of print.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 146]  [Cited by in F6Publishing: 175]  [Article Influence: 21.9]  [Reference Citation Analysis (0)]
78.  Luo H, Hao Y, Tang B, Zeng D, Shi Y, Yu P. Mouse forestomach carcinoma cells immunosuppress macrophages through TGF-β1. Turk J Gastroenterol. 2012;23:658-665.  [PubMed]  [DOI]  [Cited in This Article: ]
79.  Shen Z, Kauttu T, Cao J, Seppänen H, Vainionpää S, Ye Y, Wang S, Mustonen H, Puolakkainen P. Macrophage coculture enhanced invasion of gastric cancer cells via TGF-β and BMP pathways. Scand J Gastroenterol. 2013;48:466-472.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 14]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
80.  Shen Z, Seppänen H, Vainionpää S, Ye Y, Wang S, Mustonen H, Puolakkainen P. IL10, IL11, IL18 are differently expressed in CD14+ TAMs and play different role in regulating the invasion of gastric cancer cells under hypoxia. Cytokine. 2012;59:352-357.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 27]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
81.  Shen Z, Ye Y, Kauttu T, Seppänen H, Vainionpää S, Wang S, Mustonen H, Puolakkainen P. The novel focal adhesion gene kindlin-2 promotes the invasion of gastric cancer cells mediated by tumor-associated macrophages. Oncol Rep. 2013;29:791-797.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 18]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
82.  Shen Z, Kauttu T, Seppänen H, Vainionpää S, Ye Y, Wang S, Mustonen H, Puolakkainen P. Both macrophages and hypoxia play critical role in regulating invasion of gastric cancer in vitro. Acta Oncol. 2013;52:852-860.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 27]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
83.  Park JY, Sung JY, Lee J, Park YK, Kim YW, Kim GY, Won KY, Lim SJ. Polarized CD163+ tumor-associated macrophages are associated with increased angiogenesis and CXCL12 expression in gastric cancer. Clin Res Hepatol Gastroenterol. 2016;40:357-365.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 71]  [Article Influence: 8.9]  [Reference Citation Analysis (0)]
84.  Ding H, Zhao L, Dai S, Li L, Wang F, Shan B. CCL5 secreted by tumor associated macrophages may be a new target in treatment of gastric cancer. Biomed Pharmacother. 2016;77:142-149.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 34]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
85.  Zhang J, Yan Y, Yang Y, Wang L, Li M, Wang J, Liu X, Duan X, Wang J. High Infiltration of Tumor-Associated Macrophages Influences Poor Prognosis in Human Gastric Cancer Patients, Associates With the Phenomenon of EMT. Medicine (Baltimore). 2016;95:e2636.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 72]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
86.  Yang T, Zhang X, Wang M, Zhang J, Huang F, Cai J, Zhang Q, Mao F, Zhu W, Qian H. Activation of mesenchymal stem cells by macrophages prompts human gastric cancer growth through NF-κB pathway. PLoS One. 2014;9:e97569.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 33]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
87.  Saito H, Osaki T, Ikeguchi M. Decreased NKG2D expression on NK cells correlates with impaired NK cell function in patients with gastric cancer. Gastric Cancer. 2012;15:27-33.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 45]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
88.  Nio Y, Shiraishi T, Imai S, Tsubono M, Morimoto H, Tseng CC, Tobe T. The clinical status and histopathological factors affecting natural killer cells of peripheral blood lymphocytes in patients with gastric cancer. J Clin Lab Immunol. 1991;35:97-108.  [PubMed]  [DOI]  [Cited in This Article: ]
89.  Yoon SJ, Heo DS, Kang SH, Lee KH, Kim WS, Kim GP, Lee JA, Lee KS, Bang YJ, Kim NK. Natural killer cell activity depression in peripheral blood and ascites from gastric cancer patients with high TGF-beta 1 expression. Anticancer Res. 1998;18:1591-1596.  [PubMed]  [DOI]  [Cited in This Article: ]
90.  Rhee I, Zhong MC, Reizis B, Cheong C, Veillette A. Control of dendritic cell migration, T cell-dependent immunity, and autoimmunity by protein tyrosine phosphatase PTPN12 expressed in dendritic cells. Mol Cell Biol. 2014;34:888-899.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 37]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
91.  Ishigami S, Natsugoe S, Tokuda K, Nakajo A, Xiangming C, Iwashige H, Aridome K, Hokita S, Aikou T. Clinical impact of intratumoral natural killer cell and dendritic cell infiltration in gastric cancer. Cancer Lett. 2000;159:103-108.  [PubMed]  [DOI]  [Cited in This Article: ]
92.  Ananiev J, Gulubova MV, Manolova IM. Prognostic significance of CD83 positive tumor-infiltrating dendritic cells and expression of TGF-beta 1 in human gastric cancer. Hepatogastroenterology. 2011;58:1834-1840.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 18]  [Reference Citation Analysis (0)]
93.  Hu M, Li K, Maskey N, Xu Z, Peng C, Wang B, Li Y, Yang G. Decreased intratumoral Foxp3 Tregs and increased dendritic cell density by neoadjuvant chemotherapy associated with favorable prognosis in advanced gastric cancer. Int J Clin Exp Pathol. 2014;7:4685-4694.  [PubMed]  [DOI]  [Cited in This Article: ]
94.  Kim JE, Lee JY, Kang MJ, Jeong YJ, Choi JA, Oh SM, Lee KB, Park JH. Withaferin A Inhibits Helicobacter pylori-induced Production of IL-1β in Dendritic Cells by Regulating NF-κB and NLRP3 Inflammasome Activation. Immune Netw. 2015;15:269-277.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 29]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
95.  Chang LL, Wang SW, Wu IC, Yu FJ, Su YC, Chen YP, Wu DC, Kuo CH, Hung CH. Impaired dendritic cell maturation and IL-10 production following H. pylori stimulation in gastric cancer patients. Appl Microbiol Biotechnol. 2012;96:211-220.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 23]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
96.  Wang L, Chang EW, Wong SC, Ong SM, Chong DQ, Ling KL. Increased myeloid-derived suppressor cells in gastric cancer correlate with cancer stage and plasma S100A8/A9 proinflammatory proteins. J Immunol. 2013;190:794-804.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 181]  [Cited by in F6Publishing: 189]  [Article Influence: 15.8]  [Reference Citation Analysis (0)]
97.  Gabitass RF, Annels NE, Stocken DD, Pandha HA, Middleton GW. Elevated myeloid-derived suppressor cells in pancreatic, esophageal and gastric cancer are an independent prognostic factor and are associated with significant elevation of the Th2 cytokine interleukin-13. Cancer Immunol Immunother. 2011;60:1419-1430.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 408]  [Cited by in F6Publishing: 445]  [Article Influence: 34.2]  [Reference Citation Analysis (0)]
98.  Sansone P, Bromberg J. Environment, inflammation, and cancer. Curr Opin Genet Dev. 2011;21:80-85.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 46]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
99.  Suganuma M, Kurusu M, Suzuki K, Nishizono A, Murakami K, Fujioka T, Fujiki H. New tumor necrosis factor-alpha-inducing protein released from Helicobacter pylori for gastric cancer progression. J Cancer Res Clin Oncol. 2005;131:305-313.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 52]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
100.  Watanabe T, Takahashi A, Suzuki K, Kurusu-Kanno M, Yamaguchi K, Fujiki H, Suganuma M. Epithelial-mesenchymal transition in human gastric cancer cell lines induced by TNF-α-inducing protein of Helicobacter pylori. Int J Cancer. 2014;134:2373-2382.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 50]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
101.  Watanabe T, Tsuge H, Imagawa T, Kise D, Hirano K, Beppu M, Takahashi A, Yamaguchi K, Fujiki H, Suganuma M. Nucleolin as cell surface receptor for tumor necrosis factor-alpha inducing protein: a carcinogenic factor of Helicobacter pylori. J Cancer Res Clin Oncol. 2010;136:911-921.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 48]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
102.  Chung HW, Jang S, Kim H, Lim JB. Combined targeting of high-mobility group box-1 and interleukin-8 to control micrometastasis potential in gastric cancer. Int J Cancer. 2015;137:1598-1609.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 30]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
103.  Ma H, Wei Y, Leng Y, Li S, Gao L, Hu H, Chen L, Wang F, Xiao H, Zhu C. TGF-β1-induced expression of Id-1 is associated with tumor progression in gastric cancer. Med Oncol. 2014;31:19.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 12]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
104.  Hu WQ, Wang LW, Yuan JP, Yan SG, Li JD, Zhao HL, Peng CW, Yang GF, Li Y. High expression of transform growth factor beta 1 in gastric cancer confers worse outcome: results of a cohort study on 184 patients. Hepatogastroenterology. 2014;61:245-250.  [PubMed]  [DOI]  [Cited in This Article: ]
105.  Matsuoka J, Yashiro M, Doi Y, Fuyuhiro Y, Kato Y, Shinto O, Noda S, Kashiwagi S, Aomatsu N, Hirakawa T. Hypoxia stimulates the EMT of gastric cancer cells through autocrine TGFβ signaling. PLoS One. 2013;8:e62310.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 75]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
106.  Shinto O, Yashiro M, Kawajiri H, Shimizu K, Shimizu T, Miwa A, Hirakawa K. Inhibitory effect of a TGFbeta receptor type-I inhibitor, Ki26894, on invasiveness of scirrhous gastric cancer cells. Br J Cancer. 2010;102:844-851.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 70]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
107.  Chen F, Zhuang M, Peng J, Wang X, Huang T, Li S, Lin M, Lin H, Xu Y, Li J. Baicalein inhibits migration and invasion of gastric cancer cells through suppression of the TGF-β signaling pathway. Mol Med Rep. 2014;10:1999-2003.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 34]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
108.  Fanelli MF, Chinen LT, Begnami MD, Costa WL, Fregnami JH, Soares FA, Montagnini AL. The influence of transforming growth factor-α, cyclooxygenase-2, matrix metalloproteinase (MMP)-7, MMP-9 and CXCR4 proteins involved in epithelial-mesenchymal transition on overall survival of patients with gastric cancer. Histopathology. 2012;61:153-161.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 61]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
109.  Li P, Shan JX, Chen XH, Zhang D, Su LP, Huang XY, Yu BQ, Zhi QM, Li CL, Wang YQ. Epigenetic silencing of microRNA-149 in cancer-associated fibroblasts mediates prostaglandin E2/interleukin-6 signaling in the tumor microenvironment. Cell Res. 2015;25:588-603.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 103]  [Cited by in F6Publishing: 124]  [Article Influence: 13.8]  [Reference Citation Analysis (0)]
110.  Yang Z, Guo L, Liu D, Sun L, Chen H, Deng Q, Liu Y, Yu M, Ma Y, Guo N. Acquisition of resistance to trastuzumab in gastric cancer cells is associated with activation of IL-6/STAT3/Jagged-1/Notch positive feedback loop. Oncotarget. 2015;6:5072-5087.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 76]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
111.  Sarvaiya PJ, Guo D, Ulasov I, Gabikian P, Lesniak MS. Chemokines in tumor progression and metastasis. Oncotarget. 2013;4:2171-2185.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 229]  [Cited by in F6Publishing: 252]  [Article Influence: 25.2]  [Reference Citation Analysis (0)]
112.  Liang CM, Chen L, Hu H, Ma HY, Gao LL, Qin J, Zhong CP. Chemokines and their receptors play important roles in the development of hepatocellular carcinoma. World J Hepatol. 2015;7:1390-1402.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 37]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
113.  Lee HJ, Song IC, Yun HJ, Jo DY, Kim S. CXC chemokines and chemokine receptors in gastric cancer: from basic findings towards therapeutic targeting. World J Gastroenterol. 2014;20:1681-1693.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 60]  [Cited by in F6Publishing: 69]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
114.  Lee HJ, Kim SW, Kim HY, Li S, Yun HJ, Song KS, Kim S, Jo DY. Chemokine receptor CXCR4 expression, function, and clinical implications in gastric cancer. Int J Oncol. 2009;34:473-480.  [PubMed]  [DOI]  [Cited in This Article: ]
115.  Chen G, Chen SM, Wang X, Ding XF, Ding J, Meng LH. Inhibition of chemokine (CXC motif) ligand 12/chemokine (CXC motif) receptor 4 axis (CXCL12/CXCR4)-mediated cell migration by targeting mammalian target of rapamycin (mTOR) pathway in human gastric carcinoma cells. J Biol Chem. 2012;287:12132-12141.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 68]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
116.  Oh YS, Kim HY, Song IC, Yun HJ, Jo DY, Kim S, Lee HJ. Hypoxia induces CXCR4 expression and biological activity in gastric cancer cells through activation of hypoxia-inducible factor-1α. Oncol Rep. 2012;28:2239-2246.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 41]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
117.  Hashimoto I, Koizumi K, Tatematsu M, Minami T, Cho S, Takeno N, Nakashima A, Sakurai H, Saito S, Tsukada K. Blocking on the CXCR4/mTOR signalling pathway induces the anti-metastatic properties and autophagic cell death in peritoneal disseminated gastric cancer cells. Eur J Cancer. 2008;44:1022-1029.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 101]  [Cited by in F6Publishing: 107]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]
118.  Mashino K, Sadanaga N, Yamaguchi H, Tanaka F, Ohta M, Shibuta K, Inoue H, Mori M. Expression of chemokine receptor CCR7 is associated with lymph node metastasis of gastric carcinoma. Cancer Res. 2002;62:2937-2941.  [PubMed]  [DOI]  [Cited in This Article: ]
119.  Zhang J, Zhou Y, Yang Y. CCR7 pathway induces epithelial-mesenchymal transition through up-regulation of Snail signaling in gastric cancer. Med Oncol. 2015;32:467.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 19]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
120.  Ma H, Gao L, Li S, Qin J, Chen L, Liu X, Xu P, Wang F, Xiao H, Zhou S. CCR7 enhances TGF-β1-induced epithelial-mesenchymal transition and is associated with lymph node metastasis and poor overall survival in gastric cancer. Oncotarget. 2015;6:24348-24360.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 42]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
121.  Wroblewski LE, Noble PJ, Pagliocca A, Pritchard DM, Hart CA, Campbell F, Dodson AR, Dockray GJ, Varro A. Stimulation of MMP-7 (matrilysin) by Helicobacter pylori in human gastric epithelial cells: role in epithelial cell migration. J Cell Sci. 2003;116:3017-3026.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 116]  [Cited by in F6Publishing: 115]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
122.  Bebb JR, Letley DP, Thomas RJ, Aviles F, Collins HM, Watson SA, Hand NM, Zaitoun A, Atherton JC. Helicobacter pylori upregulates matrilysin (MMP-7) in epithelial cells in vivo and in vitro in a Cag dependent manner. Gut. 2003;52:1408-1413.  [PubMed]  [DOI]  [Cited in This Article: ]
123.  Orlichenko LS, Radisky DC. Matrix metalloproteinases stimulate epithelial-mesenchymal transition during tumor development. Clin Exp Metastasis. 2008;25:593-600.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 161]  [Cited by in F6Publishing: 157]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
124.  Shan YQ, Ying RC, Zhou CH, Zhu AK, Ye J, Zhu W, Ju TF, Jin HC. MMP-9 is increased in the pathogenesis of gastric cancer by the mediation of HER2. Cancer Gene Ther. 2015;22:101-107.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 32]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
125.  Hwang TL, Changchien TT, Wang CC, Wu CM. Claudin-4 expression in gastric cancer cells enhances the invasion and is associated with the increased level of matrix metalloproteinase-2 and -9 expression. Oncol Lett. 2014;8:1367-1371.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 29]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
126.  Al-Batran SE, Pauligk C, Wirtz R, Werner D, Steinmetz K, Homann N, Schmalenberg H, Hofheinz RD, Hartmann JT, Atmaca A. The validation of matrix metalloproteinase-9 mRNA gene expression as a predictor of outcome in patients with metastatic gastric cancer. Ann Oncol. 2012;23:1699-1705.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 18]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
127.  Yeh YC, Sheu BS, Cheng HC, Wang YL, Yang HB, Wu JJ. Elevated serum matrix metalloproteinase-3 and -7 in H. pylori-related gastric cancer can be biomarkers correlating with a poor survival. Dig Dis Sci. 2010;55:1649-1657.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 54]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
128.  Sakamoto N, Naito Y, Oue N, Sentani K, Uraoka N, Zarni Oo H, Yanagihara K, Aoyagi K, Sasaki H, Yasui W. MicroRNA-148a is downregulated in gastric cancer, targets MMP7, and indicates tumor invasiveness and poor prognosis. Cancer Sci. 2014;105:236-243.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 74]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
129.  Ogden SR, Noto JM, Allen SS, Patel DA, Romero-Gallo J, Washington MK, Fingleton B, Israel DA, Lewis ND, Wilson KT. Matrix metalloproteinase-7 and premalignant host responses in Helicobacter pylori-infected mice. Cancer Res. 2010;70:30-35.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 15]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
130.  Krakowiak MS, Noto JM, Piazuelo MB, Hardbower DM, Romero-Gallo J, Delgado A, Chaturvedi R, Correa P, Wilson KT, Peek RM. Matrix metalloproteinase 7 restrains Helicobacter pylori-induced gastric inflammation and premalignant lesions in the stomach by altering macrophage polarization. Oncogene. 2015;34:1865-1871.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 31]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
131.  Kuo CH, Liu CJ, Lu CY, Hu HM, Kuo FC, Liou YS, Yang YC, Hsieh MC, Lee OK, Wu DC. 17β-estradiol inhibits mesenchymal stem cells-induced human AGS gastric cancer cell mobility via suppression of CCL5- Src/Cas/Paxillin signaling pathway. Int J Med Sci. 2014;11:7-16.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 10]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
132.  Hou X, Zhang Y, Qiao H. CCL18 promotes the invasion and migration of gastric cancer cells via ERK1/2/NF-κB signaling pathway. Tumour Biol. 2016;37:641-651.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 16]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
133.  Leung SY, Yuen ST, Chu KM, Mathy JA, Li R, Chan AS, Law S, Wong J, Chen X, So S. Expression profiling identifies chemokine (C-C motif) ligand 18 as an independent prognostic indicator in gastric cancer. Gastroenterology. 2004;127:457-469.  [PubMed]  [DOI]  [Cited in This Article: ]
134.  Li R, Zhang H, Liu H, Lin C, Cao Y, Zhang W, Shen Z, Xu J. High expression of C-C chemokine receptor 2 associates with poor overall survival in gastric cancer patients after surgical resection. Oncotarget. 2016; Epub ahead of print.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 9]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
135.  Du P, Liu Y, Ren H, Zhao J, Zhang X, Patel R, Hu C, Gan J, Huang G. Expression of chemokine receptor CCR7 is a negative prognostic factor for patients with gastric cancer: a meta-analysis. Gastric Cancer. 2016; Epub ahead of print.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 17]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
136.  Han G, Wu D, Yang Y, Li Z, Zhang J, Li C. CrkL meditates CCL20/CCR6-induced EMT in gastric cancer. Cytokine. 2015;76:163-169.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 41]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
137.  Ohtani H, Nakayama T, Yoshie O. In situ expression of the CCL20-CCR6 axis in lymphocyte-rich gastric cancer and its potential role in the formation of lymphoid stroma. Pathol Int. 2011;61:645-651.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 13]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
138.  Wang J, Hu W, Wu X, Wang K, Yu J, Luo B, Luo G, Wang W, Wang H, Li J. CXCR1 promotes malignant behavior of gastric cancer cells in vitro and in vivo in AKT and ERK1/2 phosphorylation. Int J Oncol. 2016;48:2184-2196.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 13]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
139.  Li Z, Wang Y, Dong S, Ge C, Xiao Y, Li R, Ma X, Xue Y, Zhang Q, Lv J. Association of CXCR1 and 2 expressions with gastric cancer metastasis in ex vivo and tumor cell invasion in vitro. Cytokine. 2014;69:6-13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 29]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
140.  Li K, Zhu Z, Luo J, Fang J, Zhou H, Hu M, Maskey N, Yang G. Impact of chemokine receptor CXCR3 on tumor-infiltrating lymphocyte recruitment associated with favorable prognosis in advanced gastric cancer. Int J Clin Exp Pathol. 2015;8:14725-14732.  [PubMed]  [DOI]  [Cited in This Article: ]
141.  Haas M, Dimmler A, Hohenberger W, Grabenbauer GG, Niedobitek G, Distel LV. Stromal regulatory T-cells are associated with a favourable prognosis in gastric cancer of the cardia. BMC Gastroenterol. 2009;9:65.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 107]  [Cited by in F6Publishing: 119]  [Article Influence: 7.9]  [Reference Citation Analysis (0)]
142.  Yoo YA, Kang MH, Lee HJ, Kim BH, Park JK, Kim HK, Kim JS, Oh SC. Sonic hedgehog pathway promotes metastasis and lymphangiogenesis via activation of Akt, EMT, and MMP-9 pathway in gastric cancer. Cancer Res. 2011;71:7061-7070.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 237]  [Cited by in F6Publishing: 266]  [Article Influence: 20.5]  [Reference Citation Analysis (0)]