Topic Highlight Open Access
Copyright ©The Author(s) 2016. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Feb 28, 2016; 22(8): 2434-2440
Published online Feb 28, 2016. doi: 10.3748/wjg.v22.i8.2434
Emerging molecular basis of hematogenous metastasis in gastric cancer
Jing Zhong, Yan Chen, Liang-Jing Wang, Department of Gastroenterology, the Second Affiliated Hospital, School of Medicine and Institute of Gastroenterology, Zhejiang University, Hangzhou 310009, Zhejiang Province, China
Author contributions: Zhong J and Chen Y contributed to the literature searching and the writing of the manuscript; Wang LJ contributed to the original idea, polishing and final proof of the manuscript.
Supported by National Natural Science Foundation of China, No. 81472214 and No. 81302070; Science and Technology Innovation Team Fund of Zhejiang Province, No. 2013TD13; Natural Science Foundation of Zhejiang Province, No. LY13H160019; and Science and Technology Fund of Zhejiang Province, No. 2012C37105.
Conflict-of-interest statement: The authors have no conflict of interest to report.
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: Dr. Liang-Jing Wang, Professor, Department of Gastroenterology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, Zhejiang Province, China. wanglj76@hotmail.com
Telephone: +86-571-87783715 Fax: +86-571-86006788
Received: April 28, 2015
Peer-review started: May 7, 2015
First decision: October 14, 2015
Revised: October 29, 2015
Accepted: November 19, 2015
Article in press: November 19, 2015
Published online: February 28, 2016

Abstract

Lymphatic metastasis is commonly observed in gastric cancer (GC), but hematogenous metastasis is more likely responsible for the cancer-related mortality. Since Stephen Paget first introduced the “seed and soil hypothesis” a century ago, growing evidence recognizes that numerous essential secreted factors and signaling pathway effectors participate in the pre-metastatic niche formation and distant organ metastasis. The cross-talk between GC cells and surrounding microenvironment may consist of a series of interrelated steps, including epithelial mesenchymal transition, intravasation into blood vessels, circulating tumor cell translocation, and secondary organ metastasis. Secreted factors including vascular endothelial growth factor (VEGF), matrix metalloproteinases and cancer-derived extracellular vesicles, especially exosomes, are essential in formation of premetastatic niche. Circulating tumor cells and microRNAs represent as ‘‘metastatic intermediates’’ between primary tumors and sites of dissemination. Many biomarkers have been identified as novel metastatic markers and prognostic effectors. In addition, molecular therapy has been designed to target biomarkers such as growth factors (human epidermal growth factor receptor 2, VEGF) and chemokines, although they have not clearly proven to be effective in inhibiting GC metastasis in clinical trials. In this review, we will systematically discuss the emerging molecules and their microenvironment in hematogenous metastasis of GC, which may help us to find new therapeutic strategies in the future.

Key Words: Gastric cancer, Metastasis, Molecular mechanism

Core tip: The premetastatic niche is a novel predictor of cancer metastasis. The following steps including local invasion, intravasation into vessel lumen, survival in the circulation and extravasation also contribute to gastric cancer progression through a variety of mechanisms. This review provides an overview of the complex interaction between tumors and their microenvironment in hematogenous metastasis cascade.



INTRODUCTION

Gastric cancer (GC) is the second leading cause of cancer-related death in the world[1]. Many GC patients are diagnosed at advanced stage with metastasis, and miss the possibility for curative resection. It has been documented that patients with advanced stage GC have a poor prognosis with a five-year survival rate less than 15%[2]. Thus, metastasis is demonstrated to be an essential event in the prognosis of GC. Successful hematogenous metastasis cascade depends on intrinsic factors of the tumor cells and their subsequent communication with the surrounding microenvironment[3]. During metastatic progression, tumor cells possess continuous interdependent strategies[4]. First, tumor cells form a microenvironment, escape from primary site through surrounding extracellular matrix (ECM) and intravasate into the lumina of blood vessels. Translocation system is then formed and circulating tumor cells (CTCs) survive in the circulation and arrest at the secondary organs. Subsequently, tumor cells survive with the microenvironment of distant tissues, thereby micrometastasis and metastatic colonization emerge. In this review, we will discuss the emerging molecular mechanism that regulates hematogenous metastasis in GC.

TUMOR MICROENVIRONMENT FORMATION AS A “PRE-METASTATIC NICHE”

Stephen Paget has introduced the “seed and soil hypothesis” for a century, but emerging evidence recognizes numerous essential secreted factors and cancer-derived extracellular vesicles (EVs) as potential new effectors in pre-metastatic niche formation and metastasis.

Secreted factors

Multiple chemokines and growth factors secreted by cancer cells and their associated stromal cells are involved in recruitment of bone marrow-derived hematopoietic progenitor cells (HPCs) to support the new vessel formation and tumor metastatic microenvironment[5]. Vascular endothelial growth factor (VEGF) is initially identified as an endothelial cell-specific mitogen, which can embed endothelial cells (ECs), stimulate the formation of new blood vessels and induce angiogenesis[6]. VEGF-A, VEGF-B and placental growth factor (PIGF) binding to VEGFR-1 are considered to be key regulators of blood vessel growth, while VEGF-C and VEGF-D binding to VEGFR-2 regulate lymphatic angiogenesis[7]. High expression of VEGF was observed in patients with GC and significantly linked to the microvessel density (MVD) and the presence of vascular invasion[8,9]. Furthermore, VEGF correlated with tumor size, poor TNM stage and overall survival and appears to be a significant prognostic factor for hematogenous metastasis of GC[10]. However, a phase III randomized trial has demonstrated that anti-VEGF-A monoclonal antibody, bevacizumab, failed to improve survival in advanced GC patients[11]. The expression of other angiogenic factors such as platelet-derived growth factor (PDGF-B) was seen with a higher MVD score in 50% of diffuse-type gastric adenocarcinoma cases[12]. Examination of human tissues from relapsed GC patients demonstrated higher VEGFR-1 (FLT-1) expression[13]. Circulating VEGFR-1+ HPCs and VEGFR-2+ ECs may precede the arrival of tumor cells, then VEGFR1+ cells could maintain the expression of primitive cell surface markers, including CD34, CD11b, c-kit, and Sca-1 in the pre-metastatic niche[14]. In a smaller phase I study, there was no significant difference in median overall survival time between the HLA-A*2402-positive and -negative groups that were vaccinated with URLC10 and VEGFR1 peptides[15]. Recently, the phase III trial REGARD[16] has demonstrated that ramucirumab, a fully human IgG1 anti-VEGFR-2 monoclonal antibody, significantly improved median overall survival compared to the placebo group, suggesting VEGFR-2 signaling inhibition as an important therapeutic modality in advanced GC.

Fibroblasts and matrix metalloproteinases (MMPs) probably also contribute to pre-metastatic niche formation by secretion of pro-angiogenic and ECM-remodeling factors[17]. The interaction of VLA-4 (integrin α4β1) with its ligand, fibronectin, is essential for tumor cell migration. VCAM-1, the counter receptor of VLA-4, is significantly increased in serum of GC patients. Concentration of soluble VCAM-1 is positively associated with invasion depth and the presence of distant metastases[18]. MMPs, zinc-dependent endopeptidases capable of degrading ECM proteins, can drive the loss of the basement membrane. The activation of MMPs and urokinase-type plasminogen activator (uPA) is also required for expression of transcription factor Snail-1 which finally inhibits E-cadherin[19]. Recent studies have confirmed that elevated expression of MMP-2, MMP-7, MMP-9 and MT1-MMP in gastrointestinal cancers increases their ability to metastasize[9,20-22]. In particular, MT-MMP immunolocalized in 61% of GC cases is implicated in vascular invasion of the tumor cells through the activation of proMMP-2[23]. Following exposure to Helicobacter pylori (H. pylori) strain 60190, the expression of MMP-7 is upregulated, which is dependent on gastrin[24]. Meta-analyses showed that overexpression of MMPs (such as MMP-2 and MMP-9) is a poor prognostic factor of GC[25,26]. Moreover, human epidermal growth factor receptor 2 (HER2) knockdown resulted in the downregulation of the expression of MMP-9, which abrogates the invasion promoted by HER2 signaling in GC[27]. Clinical trials on MMP inhibitors (MMPIs) in advanced stage cancer patients have yielded inconsistent outcomes[28], however, marimastat as a broad-spectrum MMPI was illustrated to increase overall survival in patients with advanced GC in a phase III randomized trial[29].

Despite that, the release of other soluble secreted factors including lysyl oxidase, FGF, SDF-1[30], tumor necrosis factor (TNF)-α[31] and transforming growth factor (TGF)-β have been shown to play critical roles in the formation of the pre-metastatic niche.

Cancer-derived EVs

Recent research advances provide provocative insights into EVs, which have implications regarding the steps of the tumor pre-metastatic niche formation. According to their origin, EVs can be organized into several categories, including exosomes, tumor-derived microvesicles (TMVs), and large oncosomes. We will focus on the mechanisms that exosomes mediate metastasis by conditioning both the bone marrow and the premetastatic niche.

Exosomes are small, membrane-bound vesicles (40-100 nm) that transfer RNAs and proteins from one cell (site of origin) to distant locations. Several studies have indicated that hypoxia promotes the secretion of exosomes with enhanced angiogenic and metastatic potential in different tumor types[32]. The release mechanisms of exosomes involve the Rab family of small GTPases in vesicle trafficking and micro-vesicle budding pathway[33]. As expected, exosomes derived from tumor cells promote metastasis by conditioning both the bone marrow and the pre-metastatic niche[34]. Peinado et al[17] proposed a model for tumor-derived exosomes as a systemic factor increasing metastatic behavior through educating bone marrow derived cells. Recently, it has been suggested that GC-derived exosomes could promote human umbilical cord-derived MSCs migration[35]. After injecting fluorescently labeled exosomes from metastatic melanoma cells into mice, exosomes were observed to exit the circulation to localize in metastatic sites: the lung, bone marrow, liver and spleen[36]. However, little is known about whether and how GC-derived exosomes could promote distant organ metastasis. Interestingly, microRNAs (miRNAs) detected in human serum and saliva are mostly concentrated in exosomes[37]. Recent evidence has found that AZ-P7a, a metastatic GC cell line, released let-7 miRNAs via exosomes into the extracellular environment to maintain the oncogenesis[38]. The enrichment of let-7 miRNA family in the exosomes from AZ-P7a cells may reflect metastasis in GC. Later on, Melo et al[39] revealed that miRNA biogenesis in exosomes inhibited the expression of their respective mRNA targets, such as phosphatase and tensin homolog (PTEN) and the transcription factor homeobox D10 (HOXD10), indicating the contribution to breast cancer progression.

Cancer stem cells or cancer initiating cells

Accumulating evidence suggested that tumor cell subpopulation in the primary tumor mass termed Cancer stem cells (CSCs) or cancer initiating cells (CICs) has the potential to successfully form metastasis in a distant organ[40]. Since Takaishi et al[41] identified CD44 as a gastric CSC (GCSC) marker, isolation and culture of GCSCs have become a novel model for GC research. Interestingly, chronic H. pylori infection leads to an expansion of the compartment of gastric epithelial stem cells. It has been confirmed that CD44+ cells induced by H. pylori/CagA exhibited the hummingbird phenotype and stimulated the expression of mesenchymal markers[42]. Initial evidence identified the high expression of CD44 and CD133 in precancerous lesions, malignantly transforming tissues and drug-resistant GC tissues[43]. Some other molecules including TR3, Musashi-1, ALDH and Nanog have been proposed as novel metastatic markers on GCSCs, bringing great opportunities for therapy of GC patients[41].

LOCAL INVASION AND INTRAVASATION

After a pre-metastatic niche is established, metastatic carcinoma cells invade locally through surrounding ECM, which is termed as “local invasion”. Subsequently, tumor cells escape from the primary site, and disseminate via the hematogenous circulation into the vessel lumen, which is termed as “intravasation”. In the process of epithelial mesenchymal transition (EMT), tumor cells lose their epithelial cell feature and cell polarity, which makes cell permeability increase. Then the loss of polarity and alteration of the actin cytoskeleton disrupt the cell-cell adhesion and cell-ECM adhesion, mediating the concomitant formation of membrane protrusions required for invasive growth[44].

E-cadherin encoded by the CDH1 gene is essential in the activation of transcriptional regulators which disassemble intercellular junctions. A recent meta-analysis showed that[45] downregulation of E-cadherin is frequently observed in patients with diffuse type GC. The downregulation of E-cadherin results in the loss of connection of the E-cadherin-dependent cytoplasmic cell-adhesion complex and dysregulation of cellular signaling pathways including Wnt signaling, Rho GTPases, and epidermal growth factor receptor (EGFR)[46]. β-catenin acting as a transcription cofactor with T cell factor/lymphoid enhancer factor (TCF/LEF) in Wnt signaling pathway is also down-regulated in 47% of GC cases[47]. Importantly, abnormal expression of E-cadherin and β-catenin was correlated with advanced tumor stage and lower 5-year survival rate of GC patients[45,48]. Evidence suggests that Rac1, RhoA and RhoC were overexpressed in metastases GC tissues and related to higher TNM stage[49,50]. Moreover, siRNA mediated RhoC knockdown inhibits the migration and invasion of GC cells[50]. Recent findings demonstrate that RhoA mutations induced by E-cadherin are quite common in diffuse GC[51,52].

Another signaling pathway involved in EMT is TGF-β, which enhances tumor invasion and metastasis by stimulating angiogenesis and cell motility[53]. During TGF-β-induced EMT, not only the production of potent angiogenic inhibitor such as thrombospondin 1 is induced, but also transmembrane claudins, occludins and scaffold proteins are down-regulated, leading to the degradation of tight junctions[54]. Inactivation of TGF-β signaling components including TGF-β receptors, Smad2 and Smad4 consists in almost all types of GC[53]. Several studies have showed the immunoreactivity for TGF-β in gastric carcinomas, with positive rates ranging from 22.8% to 73.8%[55]. Decreased SMAD4 expression and increased SMAD7 expression have been found in GC, ensuring the inactivation of TGF-β-R1 and TGF-β-R2. Treatment with recombinant TGF-β1 or cagE-positive H. pylori significantly altered EMT-related marker and enhanced the ability of cancer cells to migrate[56]. Moreover, TGF-β is also known to regulate invasion and metastasis of GC cells via ERK, MAPK, BMP, and JNK signal pathways[57]. Significant upregulation of serum concentrations of TGF-β1 in GC patients is correlated with tumor mass and metastasis[58]. In summary, these studies suggest that activation of TGF-β signaling can regulate its associated target genes, enhancing distant metastasis in GC.

SURVIVAL IN THE CIRCULATION AND EXTRAVASATION

After tumor cells intravasate into the circulation, they disseminate widely through the venous and arterial vessels and finally migrate to a limited subset of target organs. Evidence indicates that CTCs and miRNAs represent as ‘‘metastatic intermediates’’ between primary tumors and sites of dissemination in a variety of cancers[4].

CTCs

Since CTCs were first discovered in the nineteenth century, there have been numerous studies on the detection and isolation of CTCs in peripheral blood of patients with various carcinomas[59], including gastrointestinal cancers[60]. In spite of the low count of CTCs in peripheral blood, the advances of molecular and cytological techniques enable us to detect or characterize CTCs[59,61,62]. To date, several clinical studies have demonstrated that CTCs can be a potential prognostic marker or be used to monitor the recurrence of cancers[61,63]. Numerous studies increasingly attach our attention to CTCs because of their significant utilities in the diagnosis, therapy or prognosis of various cancer types, including GC.

CTCs, as the metastatic intermediate, connect primary tumors and metastatic tumors[4,64]. Although these complex processes of translocation of tumor cells have not been fully uncovered, tumor cell shedding from primary tumors and then invading into vessels are regarded as the initiation, in which EMT plays vital roles[65,66]. Once in the blood vessels, CTCs are confronted with several natural obstacles, such as shear forces, the attack of immune cells and anoikis, which result in only 0.01 percent of CTCs alive and impede the metastatic process[67,68]. The phenomenon that CTCs in the hematogenous circulation are arrested by vessels is mainly explained by two hypotheses. One is about physical trapping of CTCs, with large diameters of 20-30 μm, in which carcinoma cells could be arrested by capillaries with the luminal diameter of 8 μm around[4], whereas the other hypothesis is that CTCs favor the trapping of particular tissues through adhesive interactions[69].

A recent meta-analysis indicated that CTCs could be a predictor of the survival of GC patients and the patients with detectable CTCs had shortened recurrence-free survival[61]. It has been reported that the positive rate of CTCs was high in advanced GC patients and indicated a poor prognosis[70]. A study also showed the tumorigenicity of CTCs from advanced GC patients[63].

miRNAs

MiRNAs play critical mechanistic roles in angiogenesis and cancer metastasis. The levels of several individual miRNAs are dysregulated in GC. A study identified that diffuse histologic type, tumor invasion and progressive stage were associated with low expression of miR-200b in patients with GC[71]. miR-200b suppresses expression of ZEB2 EMT-inducing transcription factors and enhances expression of E-cadherin[71]. We have reported that miR-141 and miR-375 were significantly downregulated in GC[73,74]. Snail-regulated miR-375 inhibited the migration and invasion of GC cells partially by targeting JAK2 oncogene[74]. Liu et al[75] reported that miR-10b promoted invasion of gastric cells by activating RhoC-AKT signaling through targeting HOXD10. In addition, miRNA signaling is converged with several crucial molecular signaling pathways. miR-21 up-regulated in GC tissues has been shown to target RECK[76], PTEN[77] and PDCD4[78] tumor suppressor genes which inhibit tumor metastasis and angiogenesis via modulating MMPs and PI3K/Akt pathway.

CONCLUSION

During the past decades, studies have indicated numerous molecular biomarkers involved in progression of the GC metastatic cascade (Table 1). There are complex processes during hematogenous metastasis in GC. In early stage, tumor cells arrive in distant sites termed as “premetastatic niche”. Secreted factors and cancer-derived EVs, especially exosomes, are essential in formation of premetastatic niche. Then these cells arrest in organs, adhere to vessel wall and invade locally through surrounding ECM. During metastatic progression, CTCs and miRNAs represent as metastatic mediators between primary tumors and sites of dissemination. We shall dwell deeper into the mechanisms of GC stem cell self-expansion, as well as crosstalk between GC-derived EVs and CTCs with surrounding microenvironment. Identification of potential molecular targets of GC metastasis will be of great importance for cancer therapy, which is a challenging issue in ameliorating the morbidity and mortality burden of GC.

Table 1 Biomarkers identified as mediators of metastasis in gastric cancer.
BiomarkersTarget gene(s)Ref.
Growth factors
VEGFCD34, CD11b, c-kit, and Sca-1[14]
PDGF-B[13]
EGFEGFR[46]
Chemokines
SDF-1CXCR4[30]
TNF-αTNFR I/II[31]
TGF-βTSP1, SMAD4[55,56]
EVs
ExosomesHOXD10 and PTEN[39]
ECM Proteases
E-cadherin[45]
Rho GTPases: Rac1, RhoA and RhoC[49,50]
MMP-2, MMP-7, MMP-9 and MT1-MMP[20-22]
VLA-4VCAM-1[18]
MicroRNAs
miR-200bZEB1 and ZEB2[71]
miR-141ZEB1[72]
miR-375JAK-2[73,74]
miR-10bHOXD10[75]
miR-21RECK, PTEN and PDCD4[76-78]
Footnotes

P- Reviewer: Koutsilieris M S- Editor: Qi Y L- Editor: Wang TQ E- Editor: Zhang DN

References
1.  Danaei G, Vander Hoorn S, Lopez AD, Murray CJ, Ezzati M. Causes of cancer in the world: comparative risk assessment of nine behavioural and environmental risk factors. Lancet. 2005;366:1784-1793.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 831]  [Cited by in F6Publishing: 745]  [Article Influence: 39.2]  [Reference Citation Analysis (0)]
2.  Hochwald SN, Kim S, Klimstra DS, Brennan MF, Karpeh MS. Analysis of 154 actual five-year survivors of gastric cancer. J Gastrointest Surg. 2000;4:520-525.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Jung HY, Fattet L, Yang J. Molecular pathways: linking tumor microenvironment to epithelial-mesenchymal transition in metastasis. Clin Cancer Res. 2015;21:962-968.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 206]  [Cited by in F6Publishing: 227]  [Article Influence: 22.7]  [Reference Citation Analysis (0)]
4.  Valastyan S, Weinberg RA. Tumor metastasis: molecular insights and evolving paradigms. Cell. 2011;147:275-292.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2388]  [Cited by in F6Publishing: 2701]  [Article Influence: 207.8]  [Reference Citation Analysis (0)]
5.  Kaplan RN, Psaila B, Lyden D. Bone marrow cells in the ‘pre-metastatic niche’: within bone and beyond. Cancer Metastasis Rev. 2006;25:521-529.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 234]  [Cited by in F6Publishing: 235]  [Article Influence: 13.8]  [Reference Citation Analysis (0)]
6.  Abdel-Rahman O. Targeting vascular endothelial growth factor (VEGF) pathway in gastric cancer: preclinical and clinical aspects. Crit Rev Oncol Hematol. 2015;93:18-27.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 41]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
7.  Olsson AK, Dimberg A, Kreuger J, Claesson-Welsh L. VEGF receptor signalling - in control of vascular function. Nat Rev Mol Cell Biol. 2006;7:359-371.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2194]  [Cited by in F6Publishing: 2264]  [Article Influence: 125.8]  [Reference Citation Analysis (0)]
8.  Chen J, Li T, Wu Y, He L, Zhang L, Shi T, Yi Z, Liu M, Pang X. Prognostic significance of vascular endothelial growth factor expression in gastric carcinoma: a meta-analysis. J Cancer Res Clin Oncol. 2011;137:1799-1812.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 31]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
9.  Partyka R, Gonciarz M, Jałowiecki P, Kokocińska D, Byrczek T. VEGF and metalloproteinase 2 (MMP 2) expression in gastric cancer tissue. Med Sci Monit. 2012;18:BR130-BR134.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Ohta M, Konno H, Tanaka T, Baba M, Kamiya K, Syouji T, Kondoh K, Watanabe M, Terada H, Nakamura S. The significance of circulating vascular endothelial growth factor (VEGF) protein in gastric cancer. Cancer Lett. 2003;192:215-225.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Shen L, Li J, Xu J, Pan H, Dai G, Qin S, Wang L, Wang J, Yang Z, Shu Y. Bevacizumab plus capecitabine and cisplatin in Chinese patients with inoperable locally advanced or metastatic gastric or gastroesophageal junction cancer: randomized, double-blind, phase III study (AVATAR study). Gastric Cancer. 2015;18:168-176.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 168]  [Cited by in F6Publishing: 195]  [Article Influence: 21.7]  [Reference Citation Analysis (0)]
12.  Suzuki S, Dobashi Y, Hatakeyama Y, Tajiri R, Fujimura T, Heldin CH, Ooi A. Clinicopathological significance of platelet-derived growth factor (PDGF)-B and vascular endothelial growth factor-A expression, PDGF receptor-β phosphorylation, and microvessel density in gastric cancer. BMC Cancer. 2010;10:659.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 79]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
13.  Suspitsin EN, Kashyap A, Shelekhova KV, Sokolenko AP, Kuligina ESh, Iyevleva AG, Kornilov AV, Ehemann V, Yanus GA, Aleksakhina SN. Evidence for angiogenesis-independent contribution of VEGFR1 (FLT1) in gastric cancer recurrence. Med Oncol. 2013;30:644.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
14.  Kaplan RN, Rafii S, Lyden D. Preparing the “soil”: the premetastatic niche. Cancer Res. 2006;66:11089-11093.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 482]  [Cited by in F6Publishing: 450]  [Article Influence: 26.5]  [Reference Citation Analysis (0)]
15.  Higashihara Y, Kato J, Nagahara A, Izumi K, Konishi M, Kodani T, Serizawa N, Osada T, Watanabe S. Phase I clinical trial of peptide vaccination with URLC10 and VEGFR1 epitope peptides in patients with advanced gastric cancer. Int J Oncol. 2014;44:662-668.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 25]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
16.  Fuchs CS, Tomasek J, Yong CJ, Dumitru F, Passalacqua R, Goswami C, Safran H, dos Santos LV, Aprile G, Ferry DR. Ramucirumab monotherapy for previously treated advanced gastric or gastro-oesophageal junction adenocarcinoma (REGARD): an international, randomised, multicentre, placebo-controlled, phase 3 trial. Lancet. 2014;383:31-39.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1541]  [Cited by in F6Publishing: 1463]  [Article Influence: 146.3]  [Reference Citation Analysis (0)]
17.  Peinado H, Lavotshkin S, Lyden D. The secreted factors responsible for pre-metastatic niche formation: old sayings and new thoughts. Semin Cancer Biol. 2011;21:139-146.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 458]  [Cited by in F6Publishing: 460]  [Article Influence: 35.4]  [Reference Citation Analysis (0)]
18.  Ding YB, Chen GY, Xia JG, Zang XW, Yang HY, Yang L. Association of VCAM-1 overexpression with oncogenesis, tumor angiogenesis and metastasis of gastric carcinoma. World J Gastroenterol. 2003;9:1409-1414.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 105]  [Cited by in F6Publishing: 107]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
19.  Radisky DC, Levy DD, Littlepage LE, Liu H, Nelson CM, Fata JE, Leake D, Godden EL, Albertson DG, Nieto MA. Rac1b and reactive oxygen species mediate MMP-3-induced EMT and genomic instability. Nature. 2005;436:123-127.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 940]  [Cited by in F6Publishing: 935]  [Article Influence: 49.2]  [Reference Citation Analysis (0)]
20.  Tester AM, Ruangpanit N, Anderson RL, Thompson EW. MMP-9 secretion and MMP-2 activation distinguish invasive and metastatic sublines of a mouse mammary carcinoma system showing epithelial-mesenchymal transition traits. Clin Exp Metastasis. 2000;18:553-560.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Yoshikawa T, Yanoma S, Tsuburaya A, Kobayashi O, Sairenji M, Motohashi H, Miyagi Y, Morinaga S, Noguchi Y, Yamamoto Y. Expression of MMP-7 and MT1-MMP in peritoneal dissemination of gastric cancer. Hepatogastroenterology. 2006;53:964-967.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Li W, Li S, Deng L, Yang S, Li M, Long S, Chen S, Lin F, Xiao L. Decreased MT1-MMP in gastric cancer suppressed cell migration and invasion via regulating MMPs and EMT. Tumour Biol. 2015;36:6883-6889.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 21]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
23.  Nomura H, Sato H, Seiki M, Mai M, Okada Y. Expression of membrane-type matrix metalloproteinase in human gastric carcinomas. Cancer Res. 1995;55:3263-3266.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  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)]
25.  Chen J, Chen LJ, Zhou HC, Yang RB, Lu Y, Xia YL, Wu W, Hu LW. Prognostic value of matrix metalloproteinase-9 in gastric cancer: a meta-analysis. Hepatogastroenterology. 2014;61:518-524.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Shen W, Xi H, Wei B, Chen L. The prognostic role of matrix metalloproteinase 2 in gastric cancer: a systematic review with meta-analysis. J Cancer Res Clin Oncol. 2014;140:1003-1009.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 54]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
27.  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)]
28.  Coussens LM, Fingleton B, Matrisian LM. Matrix metalloproteinase inhibitors and cancer: trials and tribulations. Science. 2002;295:2387-2392.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2074]  [Cited by in F6Publishing: 2040]  [Article Influence: 92.7]  [Reference Citation Analysis (0)]
29.  Bramhall SR, Hallissey MT, Whiting J, Scholefield J, Tierney G, Stuart RC, Hawkins RE, McCulloch P, Maughan T, Brown PD. Marimastat as maintenance therapy for patients with advanced gastric cancer: a randomised trial. Br J Cancer. 2002;86:1864-1870.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 196]  [Cited by in F6Publishing: 207]  [Article Influence: 9.4]  [Reference Citation Analysis (0)]
30.  Iwasa S, Yanagawa T, Fan J, Katoh R. Expression of CXCR4 and its ligand SDF-1 in intestinal-type gastric cancer is associated with lymph node and liver metastasis. Anticancer Res. 2009;29:4751-4758.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Mochizuki Y, Nakanishi H, Kodera Y, Ito S, Yamamura Y, Kato T, Hibi K, Akiyama S, Nakao A, Tatematsu M. TNF-alpha promotes progression of peritoneal metastasis as demonstrated using a green fluorescence protein (GFP)-tagged human gastric cancer cell line. Clin Exp Metastasis. 2004;21:39-47.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Azmi AS, Bao B, Sarkar FH. Exosomes in cancer development, metastasis, and drug resistance: a comprehensive review. Cancer Metastasis Rev. 2013;32:623-642.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 757]  [Cited by in F6Publishing: 838]  [Article Influence: 83.8]  [Reference Citation Analysis (0)]
33.  Shen B, Fang Y, Wu N, Gould SJ. Biogenesis of the posterior pole is mediated by the exosome/microvesicle protein-sorting pathway. J Biol Chem. 2011;286:44162-44176.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 70]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
34.  Alderton GK. Metastasis. Exosomes drive premetastatic niche formation. Nat Rev Cancer. 2012;12:447.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 73]  [Cited by in F6Publishing: 75]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
35.  Gu J, Qian H, Shen L, Zhang X, Zhu W, Huang L, Yan Y, Mao F, Zhao C, Shi Y. Gastric cancer exosomes trigger differentiation of umbilical cord derived mesenchymal stem cells to carcinoma-associated fibroblasts through TGF-β/Smad pathway. PLoS One. 2012;7:e52465.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 151]  [Cited by in F6Publishing: 199]  [Article Influence: 16.6]  [Reference Citation Analysis (0)]
36.  Peinado H, Alečković M, Lavotshkin S, Matei I, Costa-Silva B, Moreno-Bueno G, Hergueta-Redondo M, Williams C, García-Santos G, Ghajar C. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat Med. 2012;18:883-891.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2495]  [Cited by in F6Publishing: 2721]  [Article Influence: 226.8]  [Reference Citation Analysis (0)]
37.  Gallo A, Tandon M, Alevizos I, Illei GG. The majority of microRNAs detectable in serum and saliva is concentrated in exosomes. PLoS One. 2012;7:e30679.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 712]  [Cited by in F6Publishing: 801]  [Article Influence: 66.8]  [Reference Citation Analysis (2)]
38.  Ohshima K, Inoue K, Fujiwara A, Hatakeyama K, Kanto K, Watanabe Y, Muramatsu K, Fukuda Y, Ogura S, Yamaguchi K. Let-7 microRNA family is selectively secreted into the extracellular environment via exosomes in a metastatic gastric cancer cell line. PLoS One. 2010;5:e13247.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 424]  [Cited by in F6Publishing: 468]  [Article Influence: 33.4]  [Reference Citation Analysis (0)]
39.  Melo SA, Sugimoto H, O’Connell JT, Kato N, Villanueva A, Vidal A, Qiu L, Vitkin E, Perelman LT, Melo CA. Cancer exosomes perform cell-independent microRNA biogenesis and promote tumorigenesis. Cancer Cell. 2014;26:707-721.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1089]  [Cited by in F6Publishing: 1142]  [Article Influence: 114.2]  [Reference Citation Analysis (0)]
40.  Lorusso G, Rüegg C. The tumor microenvironment and its contribution to tumor evolution toward metastasis. Histochem Cell Biol. 2008;130:1091-1103.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 330]  [Cited by in F6Publishing: 343]  [Article Influence: 21.4]  [Reference Citation Analysis (0)]
41.  Li K, Dan Z, Nie YQ. Gastric cancer stem cells in gastric carcinogenesis, progression, prevention and treatment. World J Gastroenterol. 2014;20:5420-5426.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 48]  [Cited by in F6Publishing: 48]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
42.  Bessède E, Dubus P, Mégraud F, Varon C. Helicobacter pylori infection and stem cells at the origin of gastric cancer. Oncogene. 2015;34:2547-2555.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 63]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
43.  Wang T, Ong CW, Shi J, Srivastava S, Yan B, Cheng CL, Yong WP, Chan SL, Yeoh KG, Iacopetta B. Sequential expression of putative stem cell markers in gastric carcinogenesis. Br J Cancer. 2011;105:658-665.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 85]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
44.  Thiery JP, Sleeman JP. Complex networks orchestrate epithelial-mesenchymal transitions. Nat Rev Mol Cell Biol. 2006;7:131-142.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2893]  [Cited by in F6Publishing: 3008]  [Article Influence: 167.1]  [Reference Citation Analysis (0)]
45.  Li T, Chen J, Liu QL, Huo ZH, Wang ZW. Meta-analysis: E-cadherin immunoexpression as a potential prognosis biomarker related to gastric cancer metastasis in Asian patients. Eur Rev Med Pharmacol Sci. 2014;18:2693-2703.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Liu X, Chu KM. E-cadherin and gastric cancer: cause, consequence, and applications. Biomed Res Int. 2014;2014:637308.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 91]  [Article Influence: 9.1]  [Reference Citation Analysis (1)]
47.  Li LF, Wei ZJ, Sun H, Jiang B. Abnormal β-catenin immunohistochemical expression as a prognostic factor in gastric cancer: a meta-analysis. World J Gastroenterol. 2014;20:12313-12321.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 21]  [Cited by in F6Publishing: 22]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
48.  Di Bartolomeo M, Pietrantonio F, Pellegrinelli A, Martinetti A, Mariani L, Daidone MG, Bajetta E, Pelosi G, de Braud F, Floriani I. Osteopontin, E-cadherin, and β-catenin expression as prognostic biomarkers in patients with radically resected gastric cancer. Gastric Cancer. 2015;Epub ahead of print.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 28]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
49.  Pan Y, Bi F, Liu N, Xue Y, Yao X, Zheng Y, Fan D. Expression of seven main Rho family members in gastric carcinoma. Biochem Biophys Res Commun. 2004;315:686-691.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 134]  [Cited by in F6Publishing: 138]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
50.  Liu N, Zhang G, Bi F, Pan Y, Xue Y, Shi Y, Yao L, Zhao L, Zheng Y, Fan D. RhoC is essential for the metastasis of gastric cancer. J Mol Med (Berl). 2007;85:1149-1156.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 43]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
51.  Zhou J, Hayakawa Y, Wang TC, Bass AJ. RhoA mutations identified in diffuse gastric cancer. Cancer Cell. 2014;26:9-11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 33]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
52.  Ushiku T, Ishikawa S, Kakiuchi M, Tanaka A, Katoh H, Aburatani H, Lauwers GY, Fukayama M. RHOA mutation in diffuse-type gastric cancer: a comparative clinicopathology analysis of 87 cases. Gastric Cancer. 2015;Epub ahead of print.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 43]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
53.  Mishra L, Derynck R, Mishra B. Transforming growth factor-beta signaling in stem cells and cancer. Science. 2005;310:68-71.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 155]  [Cited by in F6Publishing: 156]  [Article Influence: 8.2]  [Reference Citation Analysis (0)]
54.  Ikushima H, Miyazono K. TGFbeta signalling: a complex web in cancer progression. Nat Rev Cancer. 2010;10:415-424.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 843]  [Cited by in F6Publishing: 854]  [Article Influence: 61.0]  [Reference Citation Analysis (0)]
55.  Docea AO, Mitruţ P, Grigore D, Pirici D, Călina DC, Gofiţă E. Immunohistochemical expression of TGF beta (TGF-β), TGF beta receptor 1 (TGFBR1), and Ki67 in intestinal variant of gastric adenocarcinomas. Rom J Morphol Embryol. 2012;53:683-692.  [PubMed]  [DOI]  [Cited in This Article: ]
56.  Chang H, Kim N, Park JH, Nam RH, Choi YJ, Park SM, Choi YJ, Yoon H, Shin CM, Lee DH. Helicobacter pylori Might Induce TGF-β1-Mediated EMT by Means of cagE. Helicobacter. 2015;20:438-448.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 13]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
57.  Wu WK, Cho CH, Lee CW, Fan D, Wu K, Yu J, Sung JJ. Dysregulation of cellular signaling in gastric cancer. Cancer Lett. 2010;295:144-153.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 114]  [Cited by in F6Publishing: 128]  [Article Influence: 9.1]  [Reference Citation Analysis (0)]
58.  Li X, Yue ZC, Zhang YY, Bai J, Meng XN, Geng JS, Fu SB. Elevated serum level and gene polymorphisms of TGF-beta1 in gastric cancer. J Clin Lab Anal. 2008;22:164-171.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 36]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
59.  Takeuchi H, Kitagawa Y. Circulating tumor cells in gastrointestinal cancer. J Hepatobiliary Pancreat Sci. 2010;17:577-582.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 22]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
60.  Chen Q, Ge F, Cui W, Wang F, Yang Z, Guo Y, Li L, Bremner RM, Lin PP. Lung cancer circulating tumor cells isolated by the EpCAM-independent enrichment strategy correlate with Cytokeratin 19-derived CYFRA21-1 and pathological staging. Clin Chim Acta. 2013;419:57-61.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 29]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
61.  Zhang ZY, Dai ZL, Yin XW, Li SH, Li SP, Ge HY. Meta-analysis shows that circulating tumor cells including circulating microRNAs are useful to predict the survival of patients with gastric cancer. BMC Cancer. 2014;14:773.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 17]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
62.  Zhang ZY, Ge HY. Micrometastasis in gastric cancer. Cancer Lett. 2013;336:34-45.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 47]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
63.  Toyoshima K, Hayashi A, Kashiwagi M, Hayashi N, Iwatsuki M, Ishimoto T, Baba Y, Baba H, Ohta Y. Analysis of circulating tumor cells derived from advanced gastric cancer. Int J Cancer. 2015;137:991-998.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 37]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
64.  Vegas H, André T, Bidard FC, Ferrand FR, Huguet F, Mariani P, Pierga JY. [Disseminated and circulating tumor cells in gastrointestinal oncology]. Bull Cancer. 2012;99:535-544.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 2]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
65.  Bonnomet A, Brysse A, Tachsidis A, Waltham M, Thompson EW, Polette M, Gilles C. Epithelial-to-mesenchymal transitions and circulating tumor cells. J Mammary Gland Biol Neoplasia. 2010;15:261-273.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 157]  [Cited by in F6Publishing: 140]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
66.  Książkiewicz M, Markiewicz A, Zaczek AJ. Epithelial-mesenchymal transition: a hallmark in metastasis formation linking circulating tumor cells and cancer stem cells. Pathobiology. 2012;79:195-208.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 129]  [Cited by in F6Publishing: 137]  [Article Influence: 11.4]  [Reference Citation Analysis (0)]
67.  Rhim AD, Mirek ET, Aiello NM, Maitra A, Bailey JM, McAllister F, Reichert M, Beatty GL, Rustgi AK, Vonderheide RH. EMT and dissemination precede pancreatic tumor formation. Cell. 2012;148:349-361.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1400]  [Cited by in F6Publishing: 1521]  [Article Influence: 126.8]  [Reference Citation Analysis (0)]
68.  Joosse SA, Gorges TM, Pantel K. Biology, detection, and clinical implications of circulating tumor cells. EMBO Mol Med. 2015;7:1-11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 338]  [Cited by in F6Publishing: 376]  [Article Influence: 41.8]  [Reference Citation Analysis (0)]
69.  Stanger BZ, Kahn ML. Platelets and tumor cells: a new form of border control. Cancer Cell. 2013;24:9-11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 42]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
70.  Okabe H, Tsunoda S, Hosogi H, Hisamori S, Tanaka E, Tanaka S, Sakai Y. Circulating Tumor Cells as an Independent Predictor of Survival in Advanced Gastric Cancer. Ann Surg Oncol. 2015;22:3954-3961.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 49]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
71.  Kurashige J, Kamohara H, Watanabe M, Hiyoshi Y, Iwatsuki M, Tanaka Y, Kinoshita K, Saito S, Baba Y, Baba H. MicroRNA-200b regulates cell proliferation, invasion, and migration by directly targeting ZEB2 in gastric carcinoma. Ann Surg Oncol. 2012;19 Suppl 3:S656-S664.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 120]  [Cited by in F6Publishing: 133]  [Article Influence: 11.1]  [Reference Citation Analysis (0)]
72.  Du Y, Xu Y, Ding L, Yao H, Yu H, Zhou T, Si J. Down-regulation of miR-141 in gastric cancer and its involvement in cell growth. J Gastroenterol. 2009;44:556-561.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 132]  [Cited by in F6Publishing: 147]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
73.  Ding L, Xu Y, Zhang W, Deng Y, Si M, Du Y, Yao H, Liu X, Ke Y, Si J. MiR-375 frequently downregulated in gastric cancer inhibits cell proliferation by targeting JAK2. Cell Res. 2010;20:784-793.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 260]  [Cited by in F6Publishing: 275]  [Article Influence: 19.6]  [Reference Citation Analysis (0)]
74.  Xu Y, Jin J, Liu Y, Huang Z, Deng Y, You T, Zhou T, Si J, Zhuo W. Snail-regulated MiR-375 inhibits migration and invasion of gastric cancer cells by targeting JAK2. PLoS One. 2014;9:e99516.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 56]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
75.  Liu Z, Zhu J, Cao H, Ren H, Fang X. miR-10b promotes cell invasion through RhoC-AKT signaling pathway by targeting HOXD10 in gastric cancer. Int J Oncol. 2012;40:1553-1560.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 67]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
76.  Zhang Z, Li Z, Gao C, Chen P, Chen J, Liu W, Xiao S, Lu H. miR-21 plays a pivotal role in gastric cancer pathogenesis and progression. Lab Invest. 2008;88:1358-1366.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 339]  [Cited by in F6Publishing: 362]  [Article Influence: 22.6]  [Reference Citation Analysis (0)]
77.  Yang SM, Huang C, Li XF, Yu MZ, He Y, Li J. miR-21 confers cisplatin resistance in gastric cancer cells by regulating PTEN. Toxicology. 2013;306:162-168.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 144]  [Cited by in F6Publishing: 144]  [Article Influence: 13.1]  [Reference Citation Analysis (0)]
78.  Tu H, Sun H, Lin Y, Ding J, Nan K, Li Z, Shen Q, Wei Y. Oxidative stress upregulates PDCD4 expression in patients with gastric cancer via miR-21. Curr Pharm Des. 2014;20:1917-1923.  [PubMed]  [DOI]  [Cited in This Article: ]