Topic Highlight Open Access
Copyright ©The Author(s) 2015. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Sep 14, 2015; 21(34): 9838-9852
Published online Sep 14, 2015. doi: 10.3748/wjg.v21.i34.9838
Recent advances in the molecular diagnostics of gastric cancer
Mitsuro Kanda, Yasuhiro Kodera, Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
Author contributions: Kanda M wrote the manuscript; and Kodera Y revised the manuscript for important intellectual content.
Conflict-of-interest statement: The authors have no conflict of interest related to the manuscript.
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: Mitsuro Kanda, MD, PhD, Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan. m-kanda@med.nagoya-u.ac.jp
Telephone: +81-52-7442249 Fax: +81-52-7442255
Received: April 5, 2015
Peer-review started: April 7, 2015
First decision: June 2, 2015
Revised: June 15, 2015
Accepted: August 25, 2015
Article in press: August 25, 2015
Published online: September 14, 2015

Abstract

Gastric cancer (GC) is the third most common cause of cancer-related death in the world, representing a major global health issue. Although the incidence of GC is declining, the outcomes for GC patients remain dismal because of the lack of effective biomarkers to detect early GC and predict both recurrence and chemosensitivity. Current tumor markers for GC, including serum carcinoembryonic antigen and carbohydrate antigen 19-9, are not ideal due to their relatively low sensitivity and specificity. Recent improvements in molecular techniques are better able to identify aberrant expression of GC-related molecules, including oncogenes, tumor suppressor genes, microRNAs and long non-coding RNAs, and DNA methylation, as novel molecular markers, although the molecular pathogenesis of GC is complicated by tumor heterogeneity. Detection of genetic and epigenetic alterations from gastric tissue or blood samples has diagnostic value in the management of GC. There are high expectations for molecular markers that can be used as new screening tools for early detection of GC as well as for patient stratification towards personalized treatment of GC through prediction of prognosis and drug-sensitivity. In this review, the studies of potential molecular biomarkers for GC that have been reported in the publicly available literature between 2012 and 2015 are reviewed and summarized, and certain highlighted papers are examined.

Key Words: Gastric cancer, Biomarker, Prognosis, MicroRNA, DNA methylation, Long non-coding RNA

Core tip: Gastric cancer (GC), although declining in incidence in recent decades, is still the fourth most common malignancy and the third leading cause of cancer-related death worldwide. Although reliable biomarkers are necessary to improve the management of GC, conventional tumor markers have insufficient diagnostic performance. Detection of molecular markers in gastric tissue and blood samples may enhance the sensitivity and specificity of diagnostic and prognostic tests for early stage GC and provide a means to monitor recurrence and predict response to treatment. In this review, we introduce recently reported candidates for GC-related biomarkers and overview important findings.



INTRODUCTION

Gastric cancer (GC), the third leading cause of global cancer death, is a malignant disease with a high mortality rate despite declining incidence in the recent decade[1,2]. Multimodal treatment strategies including surgery, chemotherapy and radiotherapy can improve local and regional tumor control and decrease the rate of systemic metastasis[3,4]. However, the overall prognosis for advanced disease remains poor. The high mortality rate attributed to GC is mainly due to the lack of both early detection methods and effective medical treatment for advanced stages of the disease[1,5]. Early diagnosis is beneficial and critical for successful surgical removal of GCs because peritoneal dissemination and local/distal metastases often occur in the late stages of GC and greatly reduce the efficacy of surgical intervention[6,7].

Carcinoembryonic antigen, carbohydrate antigen (CA) 19-9 and CA72-4 are the most commonly used biomarkers for GC[8]. Although widely used, they are not ideal markers because of their relatively low sensitivity and specificity in the diagnosis and prognosis of GC[9,10]. Therefore, identification of more specific and sensitive novel markers for GC is urgently required to establish screening strategies and further stratify patients for individualized therapies[3,11]. Modern biomedical research has explored many potential GC biomarker genes by examining serum protein antigens, oncogenic genes or gene families through improved molecular biological technologies, such as microarray and next-generation sequencing analyses[12]. However, there remains room for improvement of molecular-based diagnosis methods in terms of sensitivity, specificity and accessibility; therefore, they have not been utilized in clinical practice.

Recently, it was demonstrated that microRNA (miRNA) and long non-coding RNA (lncRNA) can be effective candidates for molecular diagnostics in GC in addition to altered expression of oncogenes and tumor suppressor genes (TSGs)[8,13,14]. The search for non-invasive tools for diagnosis has led to the investigation of proteins and circulating nucleic acids, including miRNAs and lncRNAs, in plasma and serum samples[15].

The aim of this review is to provide up-to-date information regarding molecular biomarkers for early detection and risk stratification for patients with GC. A prognostic cancer biomarker provides information on the likely course of the disease. In contrast, a predictive biomarker is defined as a marker that can be used to identify subpopulations of patients most likely to respond to a targeted therapy[8,16]. The search for cancer biomarkers is carried out in order to identify tumor cells at early stages and predict treatment response, ultimately leading to a favorable therapeutic outcome[8,17]. The studies of potential molecular biomarkers for GC that have been reported in the publicly available literature between 2012 and 2015 are reviewed, summarized and categorized by based on their suggested clinical implication; early detection, monitoring recurrences, prediction of survival and prediction of treatment response.

UPDATE ON GENES OVEREXPRESSED IN GC

To date, numerous GC-related oncogenes have been reported. Oncogenes are frequently overexpressed in GC and promote cancer cell growth and cell cycle progression[7,18]. They also inhibit apoptosis by silencing growth-inhibition associated genes[19]. Particularly, when the target molecules are minimally expressed in normal gastric mucosa or blood samples from healthy controls, detection of aberrantly activated oncogenes can be of great diagnostic value. Recently reported genes that are overexpressed in GC are listed in Table 1[20-44], and we review certain highlighted studies.

Table 1 Genes upregulated in gastric cancer.
Symbol (location)Biological functionMaterialsDetection methodsPtSurvivalRelevant clinical factorsFunctional analysesInteracting moleculesIn vivo studyRef.
Early detection
ERCC5 (13q33)DNA repairTissueIHC176OSDepth, smoking, Helicobacter pylori---[27]
IFITM1 (11p15.5)Interferon induced transmembrane proteinTissueIHC151-DifferentiationMigration, invasion--[21]
MMP-9 (20q13.12)Breakdown of extracellular matrixTissue, CirculatingIHC, ELISA45-N, stage---[40]
PTTG1 (5q35.1)Homolog of yeast securin proteinsTissueQPCR, IHC98OS----[43]
STC1 (8p21.2)Regulation of renal and intestinal calcium and cell metabolismTissue, circulatingIHC, ELISA83PFSN, stage---[30]
Monitoring recurrences
CD147 (19p13.3)Plasma membrane protein involved in spermatogenesisTissueIHC223OS, RFSDepth, N, stage---[26]
CEACAM6 (19q13.2)Cell adhesionTissueIHC106-T, N, vascular invasion-HER2-[28]
ERBB3 (12q13)Epidermal growth factor receptor family of receptor tyrosine kinasesTissueQPCR, IHC167OS, RFSDepth-IGF-1R, EphA2-[29]
NUAK1 (12q23.3)Regulator of whole-body and cellular energy homeostasisTissueIHC117OS, RFSDifferentiation, depth, N, stage---[37]
SULF1/2 (8q13.2)Extracellular heparan sulfate endosulfataseTissueQPCR, IHC450OS, RFSSize, depth, N, stageProliferation-Yes[20]
Prediction of survival
B7-H4 (1p13.1)Cell surface protein interacting with ligand bound to T cell receptorsCirculatingELISA132OSSize, depth, N, stage---[35]
CCND1 (11q13)Regulators of cell cycles via CDK kinasesTissueIHC211OSAge---[42]
DPYSL3 (5q32)Cell-adhesion factor involved in the metastatic process of tumor cellsTissueQPCR, IHC238OS, RFSDifferentiation, depth, N, CY, stage-VEGF, FAK, EZR-[33]
IL-17 (6p12)Proinflammatory cytokine produced by activated T cellsTissue, circulatingIHC, ELISA50-N, stage---[22]
KDM4A (1p34.1)Trimethylation-specific demethylaseTissueQPCR, IHC123OS, RFSN, stageProliferation, apoptosis--[32]
MAGED2 (Xp11.2)UnknownTissue, CirculatingQPCR, IHC225OS, RFSN, stage---[44]
MYCL1 (1p34.2)Transcription factor involved in cell differentiation and apoptosisTissueIHC176OSAge, differentiation, stage---[39]
NEDD4 (15q)Regulation of degradation of epithelial sodium channelTissueIHC214OSDepth, N, stageMigration, invasion--[36]
S100A6 (1q21)Regulation of cell cycle progression and differentiationCirculatingELISA103OSVascular invasion, perineural invasion, N, stageProliferation, invasion, apoptosis-Yes[38]
SATB1 (3p23)Matrix protein which binds nuclear matrix and scaffold-associated DNAsTissueIHC175OS, RFSAge, N---[31]
SERPINA1 (14q32.1)Serine protease inhibitorTissueQPCR, IHC400OSAge, size, depth, NMigration, invasionMMP8-[34]
XPO1 (2p15)Mediator of leucine-rich nuclear export signal-dependent proteinTissueIHC120OS, PFSHER2, CEA, stage---[25]
YBX1 (1p34)Modulator of gene transcription and protein translationTissueIHC167-N, perforationMigration, invasion--[41]
Prediction of treatment response
BMP4 (14q22-23)Endochondral bone formationTissueIHC197OSChemoresistance-EMT-[24]
TUBB3 (16q24.3)Formation of microtubulesCirculatingELISA128OSSex, chemoresistance---[23]
Bone morphogenetic protein 4

Bone morphogenetic protein 4 (BMP4) encodes a secreted protein belonging to the TGFb superfamily. BMP4 binds to BMP type I/II receptors, resulting in activation of a signaling cascade that culminates in phosphorylation of SMAD1/5/8 and the regulation of gene expression[45]. Ivanova et al[24] conducted an integrated epigenomics study to identify genes associated with cisplatin resistance in GC and found that BMP4 was an epigenetically regulated gene that is highly expressed in cisplatin-resistant GC cell lines. BMP4 promoter methylation levels were inversely correlated with BMP4 expression, and patients with high BMP4-expressing GC showed significantly worse prognosis. Inhibition of BMP4 resulted in significant sensitization of GC cells to cisplatin, and BMP4-expressing GC cells did not exhibit cross-resistance to oxaliplatin[24]. These results indicated that BMP4 epigenetic and expression status may represent a promising biomarker for cisplatin resistance in GC.

Dihydropyrimidinase-like 3

Dihydropyrimidinase-like 3 (DPYSL3) has been described as a cell-adhesion molecule; it is actively expressed in normal tissues in cardiac myocytes, brain, pineal body, retina and smooth muscle and is moderately expressed in various tissues, including gastric mucosa[46]. DPYSL3 is involved in the metastatic process of tumor cells[47,48]. We recently investigated the expression status of DPYSL3 in GC cells and tissues and found that DPYSL3 mRNA expression levels positively correlated with those of potentially interacting genes (vascular endothelial growth factor, focal adhesion kinase and ezrin)[33,49]. Tissues from patients with stage IV GC showed increased expression of DPYSL3 mRNA. High DPYSL3 mRNA expression in GCs was significantly associated with more malignant phenotypes, including recurrence, and was an independent prognostic factor. The potential of DPYSL3 as a biomarker for the progression of GC was demonstrated.

Erb-b2 receptor tyrosine kinase 3

Erb-b2 receptor tyrosine kinase 3 (ERBB3), alternatively named human epidermal growth factor receptor (HER) 3, is a key member of the ErbB family and preferentially signals through the phosphatidylinositol 3-kinase pathway[50]. ERBB3 heterodimerizes with other HER family members to initiate signal transduction[51]. Ema et al[29] conducted an integrated immunohistochemical analysis of receptor type tyrosine kinases, including ERBB3, in stage II/III GC and found that ERBB3 expression was significantly associated with shorter recurrence-free survival. Additionally, ERBB3 expression was closely correlated with IGF-1R and EphA2 expression levels and was identified as the only independent prognostic factor regardless of the stage[29]. ERBB3 is proposed to be a prognostic marker for GC after curative gastrectomy.

Serpin peptidase inhibitor, clade A member 1

Serpin peptidase inhibitor, clade A member 1 (SERPINA1) is primarily synthesized in the liver and is also produced in certain cells, such as GC, colon cancer and lung cancer cells[34,52]. SERPINA1 has been reported to have major roles in physiologic and pathologic processes, including angiogenesis, intravascular fibrinolysis, wound healing, and tumor invasion and metastasis[52]. Kwon et al[34] evaluated the clinical significance of SERPINA1 expression by immunohistochemical staining in 400 GC tissues and found that SERPINA1 expression was significantly associated with a more aggressive phenotype of GC and shorter overall survival. In the functional analysis, upregulation of SERPINA1 increased the release of metalloproteinase-8, migration and invasion in GC cells.

Extracellular heparan sulfate 6-O-endosulfatase 1

Extracellular heparan sulfate 6-O-endosulfatase 1 (SULF1) has been identified in mammals, and the encoded protein is secreted to the cell surface to modulate the sulfation of heparan sulfate proteoglycans[53,54]. Hur et al[20] conducted an expression analysis on SULF1 in 450 GC tissues to evaluate the potential of SULF1 as a biomarker for GC. The expression of SULF1 was identified as a predictive factor of lymph node metastasis, recurrence and worse prognosis. Moreover, they found that hypomethylation of CpG islands within the SULF1 gene promoter imparts oncogenic potential in GC. Expression level and methylation status of SULF1 are promising biomarkers for patients with GC.

GENES DOWNREGULATED IN GC

Loss of expression of GC-related TSGs leads to accelerated cell growth, cell cycle progression, and impaired inhibition of oncogenic gene expression[7]. Similar to oncogenes, altered expression levels of GC-related TSGs in gastric tissues can be as diagnostic molecular markers for the early detection or progression of GC[13,55]. Table 2 provides a list of updated genes that are suppressed in GC without hypermethylation[56-62], and certain representative genes are reviewed individually.

Table 2 Genes suppressed in gastric cancer.
Symbol (location)Biological functionMaterialsDetection methodsPtSurvivalRelevant clinical factorsFunctional analysesInteracting moleculesIn vivo studyRef.
Early detection
TMEFF2 (2q32.3)UnknownTissueQPCR, IHC105OS-Proliferation, apoptosisSHP-1Yes[61]
Monitoring recurrences
BTG1 (12q22)Regulator of cell growth and differentiationTissueQPCR, IHC233OS, RFSSex, location, size, N, stage---[59]
eIF3f (11p15.4)UnknownTissueIHC195RFSCEA, stage---[57]
Prediction of survival
ITIH5 (10p14)Extracellular matrix stabilizationTissueQPCR, IHC331OSDifferentiation, N, stage---[60]
JAMA (1q21.2-3)Regulator of tight junction assembly in epitheliaTissueIHC167OSSize, lymphovascular invasion, N, stageMigration, invasion--[58]
SEMA3A (7p12.1)Neuronal pattern developmentTissueIHC128OSDifferentiation, depth, N, stageProliferation, migration--[62]
STUB1 (16p13.3)Ubiquitin ligase/cochaperoneTissueIHC493OSSize, depth, N, stageProliferationNF-κB, IL-8Yes[56]
B-cell translocation gene 1

B-cell translocation gene 1 (BTG1) is a translocation partner of the c-Myc gene in the context of B-cell chronic lymphocytic leukemia and belongs to a family of antiproliferative genes[63,64]. BTG1 is constitutively expressed in quiescent cells, and its expression is downregulated as cells enter the growth cycle[65]. In breast and ovarian cancers, artificial expression of BTG1 mediates Bcl-2-regulated apoptosis and suppresses the proliferation of cancer cells[66,67]. We recently evaluated the clinical implication of BTG1 expression in GC and examined the genetic diversity among histopathologic and anatomic subtypes[59]. BTG1 expression was downregulated in the majority of GCs, but promoter hypermethylation events or sequence mutations were not detected[68,69]. Patients with downregulated BTG1 mRNA in GCs had significantly shorter recurrence-free survival and overall survival. BTG1 mRNA expression was more strongly suppressed in proximal non-diffuse and diffuse GC compared with distal non-diffuse GC, and subgroup analysis revealed that BTG1 downregulation led to adverse prognosis, specifically in patients with proximal non-diffuse and diffuse GC[59].

Inter-a-trypsin inhibitor 5

Inter-a-trypsin inhibitor 5 (ITIH5) is a new member of the ITIH family of plasma protease inhibitors and is the only ITIH gene with a CpG-rich promoter region, which contains two domains that are conserved in all known ITIHs[70]. Although the precise function of ITIH5 is unclear, it has been reported that the loss of ITIH5 expression is involved in breast cancer development[71]. Mai et al[60] investigated ITIH5 expression and its predictive value in 331 clinical GC tissues. Low ITIH5 expression was significantly associated with lymph node metastasis and advanced stage, and patients with low ITIH5 expression showed shorter survival times than those with high ITIH5 expression, suggesting that ITIH5 may be a potential prognostic biomarker for GC[60]. The mechanisms of ITIH5 silencing and oncological functions of ITIH5 in GC are expected to be clarified.

STIP1 homology and U-box containing protein 1

STIP1 homology and U-box containing protein 1 (STUB1) includes a tetratricopeptide repeat domain at its amino terminus that interacts with the molecular chaperones Hsc70-Hsp70 and the Hsp90 protein[72]. It also contains a U-box domain at its carboxy terminus with E3 ubiquitin ligase activity, which functions as a link between the chaperone and proteasome systems[73]. STUB1 induces ubiquitination and degradation of several oncogenic proteins, such as mutant p53, estrogen receptor a, c-ErbB2/neu, hypoxia inducible factor 1a and SRC-3[73,74]. Wang et al[56] evaluated the prognostic value of STUB1 expression by immunohistochemical staining in 493 patients and the role of STUB1 in tumorigenicity and angiogenesis in vitro and in vivo. Decreased STUB1 expression in GC tissues was significantly associated with advanced stage and diffuse type and was an independent prognostic factor. Forced expression of STUB1 reduced the formation of anchorage-independent colonies in soft agar, suppressed the growth of xenografts in nude mice and inhibited endothelial cell growth and tube formation by suppressing NF-κB-mediated interleukin 8 expression[56]. STUB1 acts as a TSG and represents a promising biomarker for GC.

METHYLATED MARKERS OF GC

Aberrant DNA methylation is an epigenetic alteration that occurs in an organ-disease-specific manner, and therefore, it has been studied as a molecular diagnostic marker[75,76]. To date, frequent promoter hypermethylation and subsequent loss of protein expression has been demonstrated in GC-related TSGs, and their methylation statuses in gastric tissues and blood samples have been proposed as diagnostic markers[5,77]. Because epigenetic alterations are thought to be an early event that possibly precedes gastric carcinogenesis, DNA hypomethylation and CpG island hypermethylation in pre-neoplastic or early neoplastic stages may serve as indicators or biomarkers for screening patients with an increased risk for GC[78,79]. Novel genes proposed as candidates for methylated markers of GC are listed in Table 3[80-97].

Table 3 Methylated markers in gastric cancer.
Symbol (location)Biological functionMaterialsDetection methodsPtSurvivalRelevant clinical factorsFunctional analysesInteracting moleculesIn vivo studyRef.
Early detection
APBA2 (15q11-12)Neuronal adapter protein interacting with the Alzheimer's disease amyloid precursor proteinTissue, CirculatingMSP90OSSize, differentiation, depth, invasive growth, N, CY, stage---[89]
GADD45 (1p31.2)Responding to environmental stressesTissueQPCR, IHC138OSStage---[84]
OSR1 (2p24.1)Development of intermediate mesodermTissueQPCR, IHC164OS-Proliferation, cell cycle, apoptosisβ-catenin, TCF-1-[94]
RASGRF1 (15q24.2)Mediator of Ras-GEF signaling pathwayTissueQPCR130--Proliferation, invasion--[82]
SPG20 (13q13.3)Regulating endosomal trafficking and mitochondria functionTissue, CirculatingIHC, MSP119OS----[95]
TUSC1 (9p21.2)UnknownTissueQPCR, IHC112OSAge, sex, depth, vascular invasion, N---[92]
XAF1 (17p13.1)Inhibitory factor of inhibitor of apoptosis proteinsTissue, CirculatingQPCR, IHC202OS, RFSSize, depth, N, stage, Helicobacter pylori---[85]
Monitoring recurrences
DENND2D (1p13.3)Membrane trafficking protein regulating Rab GTPasesTissueQPCR, IHC112OS, RFSAge, sex, size, depth, N, stage---[96]
PDSS2 (6q21)Synthesize the prenyl side-chain of coenzyme QTissueQPCR, IHC238OS, RFSCA19-9, N---[91]
Prediction of survival
PAX5 (9p13)B-cell lineage specific activator protein that is expressed at early stages of B-cell differentiationTissueQPCR187OSStageProliferation, migration, invasion, apoptosisp53Yes[81]
PEBP1 (12q24-23)Inhibitor of Raf1-mediated phosphorylationTissueIHC135OSDifferentiation, N, stage---[93]
RASSF5A (1q32.1)Suppressor of cell growth in response to activated Ras familyTissueQPCR, IHC132OSDifferentiation, depth, N, stage---[90]
SOCS4 (14q22.1)suppressor of cytokine signalingTissueQPCR50OSDepth, N---[80]
SOX17 (8q11.23)Transcription factor involved in the regulation of embryonic developmentCirculatingMSP73OSDifferentiation---[83]
TCF21 (6q23.2)Mesoderm specifictranscription factorTissueQPCR, IHC200OSDifferentiation, depth, N---[97]
TFF1 (21q22.3)Protecting the gastrointestinal mucosaTissueQPCR, IHC182OSDepthInvasion--[87,88]
Prediction of treatment response
RPRM (2q23.3)Regulator of cell cycles in a p53-dependent mannerTissueQPCR83OSChemoresistanceProliferation, apoptosis-Yes[86]
Amyloid beta precursor protein-binding, family A, member 2

Amyloid beta precursor protein-binding, family A, member 2 (APBA2) is a multimodular adapter protein encoded by a member of the X11 protein family and functions in membrane transport and organization[98]. Furthermore, APBA2 has been reported to be involved in signal transduction processes and is also regarded as a putative vesicular trafficking protein in the brain that can form a complex with the potential to couple synaptic vesicle exocytosis to neuronal cell adhesion[89]. Han et al[89] performed quantitative methylation-specific PCR analysis to detect APBA2 methylation using gastric tissues, peritoneal lavage fluids and blood samples. Notably, methylation of APBA2 was found in approximately 40% of peritoneal lavage fluids and blood samples from patients with GC, but not in healthy controls. In addition, positive methylation in peritoneal lavage fluids and blood samples was associated with peritoneal dissemination, advanced tumor and poor prognosis[89]. Methylation status of APBA2 can be a good biomarker that is applicable in multiple types of samples.

DENN/MADD domain-containing 2D

DENN/MADD domain-containing 2D (DENND2D) regulates Rab GTPases and represents a newly recognized class of membrane trafficking proteins[99]. DENND2D interacts directly with Rab35 and functions as a guanine nucleotide exchange factor for this GTPase[100]. We evaluated the expression level and methylation status of DENND2D in 112 pairs of gastric tissues and found that GC tissues showed a significantly lower mean mRNA expression level and a higher frequency of promoter hypermethylation of DENND2D than corresponding noncancerous tissues[96]. These findings were independent of tumor differentiation, location, and morphology. Downregulation of DENND2D mRNA in GC tissues was significantly associated with factors related to more advanced GC, recurrence and a subsequent poor prognosis[96]. Expression level and methylation status of DENND2D can serve as novel tumor biomarkers that predict progression and early recurrence of all types of GC.

Paired box gene 5

Paired box gene 5 (PAX5) was recently characterized as the key nuclear protein in the paired box-containing family of transcription factors that are involved in control of organ development and tissue differentiation[101]. PAX5 also plays a role in the early stages of B-cell differentiation, as well as neural development and spermatogenesis[102]. Li et al[81] investigated the expression, methylation and function of PAX5 in GC cells and tissues. PAX5 was frequently downregulated in GC concomitant with promoter hypermethylation. Artificial forced expression of PAX5 inhibited proliferation, migration and invasion of GC cells, arrested the cell cycle, induced apoptosis, and repressed tumorigenicity in mouse xenografts. The antitumorigenic function of PAX5 was shown to be mediated by upregulating downstream targets of p53, p21, and metastasis suppressor 1 and downregulating BCL2, cyclin D1 and mesenchymal–epithelial transition factor. Hypermethylation of PAX5 was detected in approximately 80% of GC tissues and identified as an independent prognostic factor[81]. PAX5 serves as a TSG, and its methylation status would be a prognostic marker for GC.

Reprimo

Reprimo (RPRM) is a highly glycosylated protein localized predominantly in the cytoplasm, and it has been reported to be a mediator of the cell cycle[103]. Forced expression of RPRM induces G2 arrest of the cell cycle by inhibiting Cdc2 activity and nuclear translocation of the Cdc2–cyclin B1 complex in various cell lines[104]. Ooki et al[86] evaluated the epigenetic inactivation of RPRM and its biologic function as well as its clinical relevance in GC. Frequent promoter hypermethylation was specifically detected in GCs. Forced RPRM expression inhibited proliferation, anchorage-independent colony formation of GC cells and enhanced DNA damage-induced apoptosis. Furthermore, the tumor inhibitory effect of RPRM was proven in an in vivo study. Methylation of RPRM was significantly associated with a poor response to chemotherapy and poor patient prognosis[86]. RPRM is a novel putative TSG, and promoter methylation of RPRM may serve as a predictive marker for chemotherapy and the malignant behavior of GC.

DIAGNOSTIC POTENTIAL OF miRNAS IN GC

Extensive studies in the past decade have indicated the existence and importance of an additional epigenetic mechanism for regulation of gene function by means of small non-coding miRNAs[105]. Currently, miRNAs are recognized as one of the major regulatory gatekeepers of protein-coding genes in the human genome[106]. Mature miRNAs measuring 20 to 23 nucleotides in length are incorporated into miRNA-induced silencing complexes[107]. These complexes then bind to imperfect complementary sequences in the 3’-untranslated region of target mRNAs and negatively regulate gene expression through either mRNA degradation or translational inhibition[108]. MiRNAs can be released from cancer cells into body fluids via secreting exosome particles, which could protect them from RNase degradation in the circulation[108]. With the surprising stability of miRNAs in tissues, serum or other body fluids, miRNAs have emerged as a new type of cancer biomarker with immeasurable clinical potential[12]. Here, we introduce newly identified miRNAs that potentially represent biomarkers for GC (Table 4)[109-136].

Table 4 Dysregulated microRNAs in hepatocellular carcinoma.
Symbol (location)MaterialsDetection methodsPtSurvivalRelevant clinical factorsFunctional analysesInteracting moleculesIn vivo studyRef.
Early detection
miR-21 (17q23.1)CirculatingQPCR103-Size, depth---[116]
miR-22 (17p13.3)TissueQPCR32--Proliferation, migration, invasionCD151-[117]
miR-29c (1q32.2)TissueQPCR274--Proliferation, adhesion, invasion, migrationITGB1Yes[135]
miR-30b (8q24.22)TissueQPCR21--ApoptosisPAI-1Yes[134]
miR-106b (7q22.1)CirculatingQPCR40-Age---[132]
miR-129 (7q32.1)Gastric juiceQPCR141-----[118]
miR-141 (12p13.31)TissueQPCR30--Proliferation, migration, invasionHDGF-[119]
miR-148a (7p15.2)TissueQPCR64-Size---[127]
miR-181c (19p13.13)Tissue, circulatingQPCR30-----[113]
miR-191 (3p21.31)Tissue, circulatingQPCR75-T, stageProliferation, migration, invasion, cell cycle--[126]
miR-199a-3p (12)Tissue, circulatingQPCR180-T, N, stage---[115]
miR-223 (Xq12)CirculatingQPCR60-Helicobacter pylori---[109]
miR-233 (X)CirculatingQPCR50-Size, differentiation, stage---[130]
Monitoring recurrences
miR-26a (3p22.2)TissueQPCR40OS, RFSN, stageProliferation, metastasisFGF9Yes[114]
miR-34b/c (11q23.1)Noncancerous tissuePyrosequencing129RFSAge---[128]
miR-185 (22q11.21)TissueQPCR126OS, RFSN, stageProliferation, metastasis-Yes[129]
miR-196a (17q21.32)Tissue, circulatingQPCR72--Migration, invasion--[110]
miR-200c (12p13.31)CirculatingQPCR52OS, PFS----[111]
miR-222 (Xp11.3)CirculatingQPCR114OS, RFSN, stage---[121]
Prediction of survival
miR-25 (7q22.1)Tissue, circulatingQPCR70OSN, stageProliferation, migration, invasionTOB1Yes[124]
miR-132 (17p13.3)TissueQPCR79OSLymphovascular invasion, N, stage---[125]
miR-183 (7q32.2)TissueQPCR80-Depth, N, stageProliferation, migration, invasion, apoptosisPDCD4-[122]
miR-192 (11q13.1)TissueQPCR38-Sex, vascular invasion, NInvasion--[136]
miR-214 (1q24.3)TissueQPCR80-Size, NProliferation, migration, invasionCSF1-[131]
miR-630 (15q24.1)TissueQPCR236OSDepth, N, stage---[120]
Prediction of treatment response
miR-17-5p (13q31.3)CirculatingQPCR65OSDifferentiation, stage---[112]
miR-27a (19p13.13)CirculatingQPCR82OSChemoresistance---[123]
MiR-25

Li et al[124] investigated the expression level of miR-25 in plasma and GC tissues and found overexpression of miR-25 in patients with lymph node metastasis. Inhibition of miR-25 significantly suppressed metastasis, invasion and proliferation in vitro and reduced the metastatic capacity of GC cells in vivo through repression of transducer of ERBB2, 1 expression. Furthermore, patients with high plasma expression of miR-25 had poor prognoses[124]. MiR-25 is related to GC progression through repression of transducer of ERBB2, 1 and may represent a noninvasive biomarker for GC.

MiR-129

It has been reported that miR-129 is a cancer-associated miRNA[137]. In previous studies, miR-129 levels were significantly altered in cancerous tissues, including GC, when compared to noncancerous tissues. MiR-129 has been shown to play an important role in regulating cell proliferation by downregulation of cyclin-dependent kinase 6[118,138]. Yu et al[118] assessed the diagnostic values of miR-129-1/2 in gastric secretion samples to propose a new screening tool for GC. After examining 141 secretion samples, patients with GC showed significantly lower levels of miR-129-1 and miR-129-2. Gastric secretions may be a good resource for the molecular diagnosis of GC.

MiR-199a-3p

Li et al[115] performed microarray profiling of plasma samples to compare expression patterns in GC patients and healthy controls and to identify circulating miRNAs that may be novel diagnostic markers for GC. MiRNA-199a-3p was found to be significantly elevated in GC patients and was reduced after resection of the primary tumors in the training set. In the validation stage in a large cohort, plasma miR-199a-3p was elevated in GC patients compared to healthy controls, with a high under the receiver operating characteristic curve area (0.837), and was significantly associated with tumor depth, lymph node metastasis and stage[115]. Plasma miRNA-199a-3p was shown to be a potential biomarker both for early detection and progression of GC.

MiR-630

MiR-630 has been reported to be elevated in lung, head and neck, and pancreatic cancers, and reports show that it can modulate chemosensitivity[139]. Chu et al[120] examined expression levels of miR-630 in 236 GC and adjacent normal tissues and found that miR-630 was elevated in GC tissues. Increased expression of miR-630 was significantly associated with depth of the tumor, lymph node metastasis, distant metastasis and poor overall survival, indicating that miR-630 may serve as a potential marker for the initiation and progression of GC.

SIGNIFICANCE OF lncRNAS IN GC

The genome sequencing projects revealed that the human genome is composed of less than 2% protein-coding genes and that more than 90% of the genome is transcribed as noncoding RNAs[107,140]. LncRNAs are a class of newly identified noncoding RNAs, > 200 nucleotides in length, that are currently being studied for their roles in cellular processes[141]. Changes in the expression levels of lncRNAs have been increasingly reported in various malignancies, suggesting that lncRNAs may play a role in tumorigenesis and tumor progression[142]. Interestingly, recent studies have suggested that lncRNAs also exist in serum, plasma and other body fluids, and certain lncRNAs have been described as candidate biomarkers[142,143]. Here, we introduce reported GC-related lncRNAs from recent publications (Table 5)[140,142,144-147].

Table 5 Gastric cancer-associated long non cording RNAs.
Symbol (location)MaterialsDetection methodsPtSurvivalRelevant clinical factorsFunctional analysesInteracting moleculesIn vivo studyRef.
Early detection
CUDR (19p13.12)CirculatingQPCR106OS----[142]
HOTAIR (12q13.13)TissueQPCR60-Perineural invasion, stage---[144]
Prediction of survival
BLACAT1 (1q32.1)TissueQPCR85OSSize, N, stageProliferation, migration, invasion--[147]
CCAT1 (8q24.21)TissueQPCR62-Size, stageProliferation, apoptosisERK/MAPK pathway-[140]
GAPLINC (18)TissueMicroarray90OSSize, NProliferation, migrationCD44-[145]
HIF1A-AS2 (14q23.2)TissueQPCR83OSDepth, N, stageProliferation-Yes[146]
Colon cancer associated transcript 1

Colon cancer associated transcript 1 (CCAT1) was found to be generally upregulated in colon cancer and correlated with the rs6983267 allele, which was associated with increased cancer susceptibility[148]. The MYC enhancer region physically interacts with the promoter region of CCAT1, suggesting that the cancer-associated variant rs6983267 as an MYC enhancer could regulate CCAT1 expression[140]. Additionally, CCAT1 was reported to have a role in cell-cycle regulation and development of colon cancer[148]. Zhang et al[140] reported that CCAT1 was upregulated in GC tissues compared to paired adjacent normal tissues and that knockdown of CCAT1 significantly inhibited proliferation of GC cells by inducing G0/G1 cell-cycle arrest, apoptosis and inactivation of the ERK/MAPK signaling pathway. Diagnostic performance of CCAT1 is expected to be evaluated in a large cohort in the future.

Hypoxia inducible factor 1 alpha antisense RNA-2

Hypoxia inducible factor 1 alpha antisense RNA-2 (HIF1A-AS2) is an antisense long noncoding RNA, which is a natural antisense transcript of hypoxia-inducible factor 1alpha (HIF-1α)[149]. Although earlier reports indicated that HIF1A-AS2 plays a crucial role in cancer development, via regulation of the cancer-relevant HIF-1α pathway, its oncological role in GC remains to be determined[150,151]. Chen et al[146] reported that upregulation of HIF1A-AS2 was found in GC tissues and significantly correlated with tumor depth, lymph node metastasis, advanced stage and poor prognosis. Furthermore, knockdown of HIF1A-AS2 in GC cells inhibited proliferation in vitro and tumorigenesis in vivo. HIF1A-AS2 may be considered as a promising biomarker for GC.

Gastric adenocarcinoma predictive long intergenic noncoding RNA

Hu et al[145] conducted global microarray and in situ hybridization analyses to explore novel GC-related lncRNAs and identified gastric adenocarcinoma predictive long intergenic noncoding RNA (GAPLINC) as an aberrantly expressed lncRNA. Suppression of GAPLINC led to alterations in cell migration pathways, particularly in CD44. GAPLINC induced increased the cell migration and proliferation abilities of GC cells, and the positive effects of GAPLINC were neutralized by suppression of CD44[145]. Patients with high GAPLINC expression in GC tissues had a significantly worse prognosis, suggesting that GAPLINC may represent a promising biomarker for GC.

CONCLUSION

Exhaustive research performed over recent years and the development of new genetic technologies have built the foundation for a better understanding of the molecular pathogenesis of GC[18,19]. This review aimed to describe the relevance of genomics as a novel diagnostic and prognostic tool in GC, to give an overview of epigenetics in GC (methylation, miRNA and lncRNA) and to discuss how the application of molecular data to the management of GC might improve the accuracy of prognosis prediction and lead to more efficient personalized treatments for GC.

Improvement of the treatment outcomes for GC in the future is dependent on the development of sophisticated biomarkers[8,17]. High-performance biomarkers for early detection, potential distant metastasis and prediction of chemosensitivity, recurrence and prognosis enable personalized therapy[152]. Even with many putative biomarker molecules identified, the outcomes for GC patients remain dismal due to modest improvements in clinical treatment strategies. Increased translational medicine efforts should be made to globally encourage standardized systematic biomarker validation studies in GC. On the basis of recent data, this review highlights the potential of recently reported molecular markers as biomarkers for GC and explores their relationship to disease susceptibility, diagnosis, prognosis and response to treatment.

Despite these encouraging results, there are still many issues to be resolved in the field of GC-related molecular biomarker research. First, a major challenge to identifying reliable biomarkers is inter-individual variability of expression levels influenced by various factors such as pathology, hypoxia, infection and cytotoxic treatment, response to targeted therapy and drug resistance[8]. Second, there is not yet enough data available on circulating molecular profiles to be used as potential biomarkers for the diagnosis and prognosis of GC. Ultimately, blood samples can represent noninvasive screening tools without other invasive procedures such as endoscopy and surgery. Third, we require more robust platforms and quick analytical methods because DNA/RNA extraction and bisulfite conversion is too time-intensive for clinical use. Finally, most studies demonstrating the diagnostic potential of molecular markers have involved small sample sets. Thus, these candidate molecules must be validated in large independent cohorts to confirm the existence of a predictive value.

Although there are still many challenges in the field of GC-related molecular biomarker research, the accumulation of genetic and epigenetic data is of key importance to improve the diagnosis and management of GC and overcome this disease in the future.

Footnotes

P- Reviewer: Larentzakis A, Li Y S- Editor: Ma YJ L- Editor: A E- Editor: Ma S

References
1.  Liang H, Kim YH. Identifying molecular drivers of gastric cancer through next-generation sequencing. Cancer Lett. 2013;340:241-246.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  GLOBOCAN 2012. Estimated Cancer Incidence, Mortality and Prevalence Worldwide in 2012, Stomach Cancer.  Available from: http://globocan.iarc.fr/Pages/ fact_sheets_cancer.aspx.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Wang F, Sun GP, Zou YF, Hao JQ, Zhong F, Ren WJ. MicroRNAs as promising biomarkers for gastric cancer. Cancer Biomark. 2012;11:259-267.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Hartgrink HH, Jansen EP, van Grieken NC, van de Velde CJ. Gastric cancer. Lancet. 2009;374:477-490.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  González CA, Agudo A. Carcinogenesis, prevention and early detection of gastric cancer: where we are and where we should go. Int J Cancer. 2012;130:745-753.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Kanda M, Kobayashi D, Tanaka C, Iwata N, Yamada S, Fujii T, Nakayama G, Sugimoto H, Koike M, Nomoto S. Adverse prognostic impact of perioperative allogeneic transfusion on patients with stage II/III gastric cancer. Gastric Cancer. 2015;Epub ahead of print.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Cho JY. Molecular diagnosis for personalized target therapy in gastric cancer. J Gastric Cancer. 2013;13:129-135.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Durães C, Almeida GM, Seruca R, Oliveira C, Carneiro F. Biomarkers for gastric cancer: prognostic, predictive or targets of therapy? Virchows Arch. 2014;464:367-378.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Emoto S, Ishigami H, Yamashita H, Yamaguchi H, Kaisaki S, Kitayama J. Clinical significance of CA125 and CA72-4 in gastric cancer with peritoneal dissemination. Gastric Cancer. 2012;15:154-161.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Kanda M, Fujii T, Takami H, Suenaga M, Inokawa Y, Yamada S, Nakayama G, Sugimoto H, Koike M, Nomoto S. The combination of the serum carbohydrate antigen 19-9 and carcinoembryonic antigen is a simple and accurate predictor of mortality in pancreatic cancer patients. Surg Today. 2014;44:1692-1701.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Leung WK, Wu MS, Kakugawa Y, Kim JJ, Yeoh KG, Goh KL, Wu KC, Wu DC, Sollano J, Kachintorn U. Screening for gastric cancer in Asia: current evidence and practice. Lancet Oncol. 2008;9:279-287.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Wu HH, Lin WC, Tsai KW. Advances in molecular biomarkers for gastric cancer: miRNAs as emerging novel cancer markers. Expert Rev Mol Med. 2014;16:e1.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Janjigian YY, Kelsen DP. Genomic dysregulation in gastric tumors. J Surg Oncol. 2013;107:237-242.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Cancer Genome Atlas Research Network. Comprehensive molecular characterization of gastric adenocarcinoma. Nature. 2014;513:202-209.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Liu HS, Xiao HS. MicroRNAs as potential biomarkers for gastric cancer. World J Gastroenterol. 2014;20:12007-12017.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Yasui W, Sentani K, Sakamoto N, Anami K, Naito Y, Oue N. Molecular pathology of gastric cancer: research and practice. Pathol Res Pract. 2011;207:608-612.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Lin LL, Huang HC, Juan HF. Discovery of biomarkers for gastric cancer: a proteomics approach. J Proteomics. 2012;75:3081-3097.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  McLean MH, El-Omar EM. Genetics of gastric cancer. Nat Rev Gastroenterol Hepatol. 2014;11:664-674.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Razzak M. Genetics: new molecular classification of gastric adenocarcinoma proposed by The Cancer Genome Atlas. Nat Rev Clin Oncol. 2014;11:499.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Hur K, Han TS, Jung EJ, Yu J, Lee HJ, Kim WH, Goel A, Yang HK. Up-regulated expression of sulfatases (SULF1 and SULF2) as prognostic and metastasis predictive markers in human gastric cancer. J Pathol. 2012;228:88-98.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Lee J, Goh SH, Song N, Hwang JA, Nam S, Choi IJ, Shin A, Kim IH, Ju MH, Jeong JS. Overexpression of IFITM1 has clinicopathologic effects on gastric cancer and is regulated by an epigenetic mechanism. Am J Pathol. 2012;181:43-52.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Meng XY, Zhou CH, Ma J, Jiang C, Ji P. Expression of interleukin-17 and its clinical significance in gastric cancer patients. Med Oncol. 2012;29:3024-3028.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Yu J, Gao J, Lu Z, Li Y, Shen L. Serum levels of TUBB3 correlate with clinical outcome in Chinese patients with advanced gastric cancer receiving first-line paclitaxel plus capecitabine. Med Oncol. 2012;29:3029-3034.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Ivanova T, Zouridis H, Wu Y, Cheng LL, Tan IB, Gopalakrishnan V, Ooi CH, Lee J, Qin L, Wu J. Integrated epigenomics identifies BMP4 as a modulator of cisplatin sensitivity in gastric cancer. Gut. 2013;62:22-33.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Zhou F, Qiu W, Yao R, Xiang J, Sun X, Liu S, Lv J, Yue L. CRM1 is a novel independent prognostic factor for the poor prognosis of gastric carcinomas. Med Oncol. 2013;30:726.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Chu D, Zhu S, Li J, Ji G, Wang W, Wu G, Zheng J. CD147 expression in human gastric cancer is associated with tumor recurrence and prognosis. PLoS One. 2014;9:e101027.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Deng N, Liu JW, Sun LP, Xu Q, Duan ZP, Dong NN, Yuan Y. Expression of XPG protein in the development, progression and prognosis of gastric cancer. PLoS One. 2014;9:e108704.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Deng X, Liu P, Zhao Y, Wang Q. Expression profiling of CEACAM6 associated with the tumorigenesis and progression in gastric adenocarcinoma. Genet Mol Res. 2014;13:7686-7697.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Ema A, Yamashita K, Ushiku H, Kojo K, Minatani N, Kikuchi M, Mieno H, Moriya H, Hosoda K, Katada N. Immunohistochemical analysis of RTKs expression identified HER3 as a prognostic indicator of gastric cancer. Cancer Sci. 2014;105:1591-1600.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Fang Z, Tian Z, Luo K, Song H, Yi J. Clinical significance of stanniocalcin expression in tissue and serum of gastric cancer patients. Chin J Cancer Res. 2014;26:602-610.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Hedner C, Gaber A, Korkocic D, Nodin B, Uhlén M, Kuteeva E, Johannesson H, Jirström K, Eberhard J. SATB1 is an independent prognostic factor in radically resected upper gastrointestinal tract adenocarcinoma. Virchows Arch. 2014;465:649-659.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Hu CE, Liu YC, Zhang HD, Huang GJ. JMJD2A predicts prognosis and regulates cell growth in human gastric cancer. Biochem Biophys Res Commun. 2014;449:1-7.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Kanda M, Nomoto S, Oya H, Shimizu D, Takami H, Hibino S, Hashimoto R, Kobayashi D, Tanaka C, Yamada S. Dihydropyrimidinase-like 3 facilitates malignant behavior of gastric cancer. J Exp Clin Cancer Res. 2014;33:66.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Kwon CH, Park HJ, Lee JR, Kim HK, Jeon TY, Jo HJ, Kim DH, Kim GH, Park DY. Serpin peptidase inhibitor clade A member 1 is a biomarker of poor prognosis in gastric cancer. Br J Cancer. 2014;111:1993-2002.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Shi H, Ji M, Wu J, Zhou Q, Li X, Li Z, Zheng X, Xu B, Zhao W, Wu C. Serum B7-H4 expression is a significant prognostic indicator for patients with gastric cancer. World J Surg Oncol. 2014;12:188.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Sun A, Yu G, Dou X, Yan X, Yang W, Lin Q. Nedd4-1 is an exceptional prognostic biomarker for gastric cardia adenocarcinoma and functionally associated with metastasis. Mol Cancer. 2014;13:248.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Ye XT, Guo AJ, Yin PF, Cao XD, Chang JC. Overexpression of NUAK1 is associated with disease-free survival and overall survival in patients with gastric cancer. Med Oncol. 2014;31:61.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Zhang J, Zhang K, Jiang X, Zhang J. S100A6 as a potential serum prognostic biomarker and therapeutic target in gastric cancer. Dig Dis Sci. 2014;59:2136-2144.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Chen S, Tang J, Huang L, Lin J. Expression and prognostic value of Mycl1 in gastric cancer. Biochem Biophys Res Commun. 2015;456:879-883.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Chen SZ, Yao HQ, Zhu SZ, Li QY, Guo GH, Yu J. Expression levels of matrix metalloproteinase-9 in human gastric carcinoma. Oncol Lett. 2015;9:915-919.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Guo T, Yu Y, Yip GW, Baeg GH, Thike AA, Lim TK, Tan PH, Matsumoto K, Bay BH. Y-box binding protein 1 is correlated with lymph node metastasis in intestinal-type gastric cancer. Histopathology. 2015;66:491-499.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Ma L, Wang X, Lan F, Yu Y, Ouyang X, Liu W, Xie F, Huang Q. Prognostic value of differential CCND1 expression in patients with resected gastric adenocarcinoma. Med Oncol. 2015;32:338.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Xu MD, Dong L, Qi P, Weng WW, Shen XH, Ni SJ, Huang D, Tan C, Sheng WQ, Zhou XY. Pituitary tumor-transforming gene-1 serves as an independent prognostic biomarker for gastric cancer. Gastric Cancer. 2015;Epub ahead of print.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Kanda M, Nomoto S, Oya H, Takami H, Shimizu D, Hibino S, Hashimoto R, Kobayashi D, Tanaka C, Yamada S. The Expression of Melanoma-Associated Antigen D2 Both in Surgically Resected and Serum Samples Serves as Clinically Relevant Biomarker of Gastric Cancer Progression. Ann Surg Oncol. 2015;Epub ahead of print.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Pinto M, Oliveira C, Cirnes L, Carlos Machado J, Ramires M, Nogueira A, Carneiro F, Seruca R. Promoter methylation of TGFbeta receptor I and mutation of TGFbeta receptor II are frequent events in MSI sporadic gastric carcinomas. J Pathol. 2003;200:32-38.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Oya H, Kanda M, Sugimoto H, Shimizu D, Takami H, Hibino S, Hashimoto R, Okamura Y, Yamada S, Fujii T. Dihydropyrimidinase-like 3 is a putative hepatocellular carcinoma tumor suppressor. J Gastroenterol. 2015;50:590-600.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Rosslenbroich V, Dai L, Baader SL, Noegel AA, Gieselmann V, Kappler J. Collapsin response mediator protein-4 regulates F-actin bundling. Exp Cell Res. 2005;310:434-444.  [PubMed]  [DOI]  [Cited in This Article: ]
48.  Kawahara T, Hotta N, Ozawa Y, Kato S, Kano K, Yokoyama Y, Nagino M, Takahashi T, Yanagisawa K. Quantitative proteomic profiling identifies DPYSL3 as pancreatic ductal adenocarcinoma-associated molecule that regulates cell adhesion and migration by stabilization of focal adhesion complex. PLoS One. 2013;8:e79654.  [PubMed]  [DOI]  [Cited in This Article: ]
49.  Gao X, Pang J, Li LY, Liu WP, Di JM, Sun QP, Fang YQ, Liu XP, Pu XY, He D. Expression profiling identifies new function of collapsin response mediator protein 4 as a metastasis-suppressor in prostate cancer. Oncogene. 2010;29:4555-4566.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Begnami MD, Fukuda E, Fregnani JH, Nonogaki S, Montagnini AL, da Costa WL, Soares FA. Prognostic implications of altered human epidermal growth factor receptors (HERs) in gastric carcinomas: HER2 and HER3 are predictors of poor outcome. J Clin Oncol. 2011;29:3030-3036.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Ocana A, Vera-Badillo F, Seruga B, Templeton A, Pandiella A, Amir E. HER3 overexpression and survival in solid tumors: a meta-analysis. J Natl Cancer Inst. 2013;105:266-273.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Allgayer H, Babic R, Grützner KU, Beyer BC, Tarabichi A, Schildberg FW, Heiss MM. Tumor-associated proteases and inhibitors in gastric cancer: analysis of prognostic impact and individual risk protease patterns. Clin Exp Metastasis. 1998;16:62-73.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  Dhoot GK, Gustafsson MK, Ai X, Sun W, Standiford DM, Emerson CP. Regulation of Wnt signaling and embryo patterning by an extracellular sulfatase. Science. 2001;293:1663-1666.  [PubMed]  [DOI]  [Cited in This Article: ]
54.  Morimoto-Tomita M, Uchimura K, Werb Z, Hemmerich S, Rosen SD. Cloning and characterization of two extracellular heparin-degrading endosulfatases in mice and humans. J Biol Chem. 2002;277:49175-49185.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Jang BG, Kim WH. Molecular pathology of gastric carcinoma. Pathobiology. 2011;78:302-310.  [PubMed]  [DOI]  [Cited in This Article: ]
56.  Wang S, Wu X, Zhang J, Chen Y, Xu J, Xia X, He S, Qiang F, Li A, Shu Y. CHIP functions as a novel suppressor of tumour angiogenesis with prognostic significance in human gastric cancer. Gut. 2013;62:496-508.  [PubMed]  [DOI]  [Cited in This Article: ]
57.  Cheng Y, Jia C, Li G, Li H. Expression of eukaryotic initiation factor 3f is associated with prognosis in gastric carcinomas. Oncol Res Treat. 2014;37:198-202.  [PubMed]  [DOI]  [Cited in This Article: ]
58.  Huang JY, Xu YY, Sun Z, Wang ZN, Zhu Z, Song YX, Luo Y, Zhang X, Xu HM. Low junctional adhesion molecule A expression correlates with poor prognosis in gastric cancer. J Surg Res. 2014;192:494-502.  [PubMed]  [DOI]  [Cited in This Article: ]
59.  Kanda M, Oya H, Nomoto S, Takami H, Shimizu D, Hashimoto R, Sueoka S, Kobayashi D, Tanaka C, Yamada S. Diversity of clinical implication of B-cell translocation gene 1 expression by histopathologic and anatomic subtypes of gastric cancer. Dig Dis Sci. 2015;60:1256-1264.  [PubMed]  [DOI]  [Cited in This Article: ]
60.  Mai C, Zhao JJ, Tang XF, Wang W, Pan K, Pan QZ, Zhang XF, Jiang SS, Zhao BW, Li YF. Decreased ITIH5 expression is associated with poor prognosis in primary gastric cancer. Med Oncol. 2014;31:53.  [PubMed]  [DOI]  [Cited in This Article: ]
61.  Sun T, Du W, Xiong H, Yu Y, Weng Y, Ren L, Zhao H, Wang Y, Chen Y, Xu J. TMEFF2 deregulation contributes to gastric carcinogenesis and indicates poor survival outcome. Clin Cancer Res. 2014;20:4689-4704.  [PubMed]  [DOI]  [Cited in This Article: ]
62.  Tang C, Gao X, Liu H, Jiang T, Zhai X. Decreased expression of SEMA3A is associated with poor prognosis in gastric carcinoma. Int J Clin Exp Pathol. 2014;7:4782-4794.  [PubMed]  [DOI]  [Cited in This Article: ]
63.  Rouault JP, Rimokh R, Tessa C, Paranhos G, Ffrench M, Duret L, Garoccio M, Germain D, Samarut J, Magaud JP. BTG1, a member of a new family of antiproliferative genes. EMBO J. 1992;11:1663-1670.  [PubMed]  [DOI]  [Cited in This Article: ]
64.  Nahta R, Yuan LX, Fiterman DJ, Zhang L, Symmans WF, Ueno NT, Esteva FJ. B cell translocation gene 1 contributes to antisense Bcl-2-mediated apoptosis in breast cancer cells. Mol Cancer Ther. 2006;5:1593-1601.  [PubMed]  [DOI]  [Cited in This Article: ]
65.  Kanda M, Sugimoto H, Nomoto S, Oya H, Hibino S, Shimizu D, Takami H, Hashimoto R, Okamura Y, Yamada S. B-cell translocation gene 1 serves as a novel prognostic indicator of hepatocellular carcinoma. Int J Oncol. 2015;46:641-648.  [PubMed]  [DOI]  [Cited in This Article: ]
66.  Sheng SH, Zhao CM, Sun GG. BTG1 expression correlates with the pathogenesis and progression of breast carcinomas. Tumour Biol. 2014;35:3317-3326.  [PubMed]  [DOI]  [Cited in This Article: ]
67.  Sun GG, Lu YF, Cheng YJ, Hu WN. The expression of BTG1 is downregulated in NSCLC and possibly associated with tumor metastasis. Tumour Biol. 2014;35:2949-2957.  [PubMed]  [DOI]  [Cited in This Article: ]
68.  Kanda M, Matthaei H, Wu J, Hong SM, Yu J, Borges M, Hruban RH, Maitra A, Kinzler K, Vogelstein B. Presence of somatic mutations in most early-stage pancreatic intraepithelial neoplasia. Gastroenterology. 2012;142:730-733.e9.  [PubMed]  [DOI]  [Cited in This Article: ]
69.  Kanda M, Knight S, Topazian M, Syngal S, Farrell J, Lee J, Kamel I, Lennon AM, Borges M, Young A. Mutant GNAS detected in duodenal collections of secretin-stimulated pancreatic juice indicates the presence or emergence of pancreatic cysts. Gut. 2013;62:1024-1033.  [PubMed]  [DOI]  [Cited in This Article: ]
70.  Himmelfarb M, Klopocki E, Grube S, Staub E, Klaman I, Hinzmann B, Kristiansen G, Rosenthal A, Dürst M, Dahl E. ITIH5, a novel member of the inter-alpha-trypsin inhibitor heavy chain family is downregulated in breast cancer. Cancer Lett. 2004;204:69-77.  [PubMed]  [DOI]  [Cited in This Article: ]
71.  Veeck J, Chorovicer M, Naami A, Breuer E, Zafrakas M, Bektas N, Dürst M, Kristiansen G, Wild PJ, Hartmann A. The extracellular matrix protein ITIH5 is a novel prognostic marker in invasive node-negative breast cancer and its aberrant expression is caused by promoter hypermethylation. Oncogene. 2008;27:865-876.  [PubMed]  [DOI]  [Cited in This Article: ]
72.  Esser C, Alberti S, Höhfeld J. Cooperation of molecular chaperones with the ubiquitin/proteasome system. Biochim Biophys Acta. 2004;1695:171-188.  [PubMed]  [DOI]  [Cited in This Article: ]
73.  Connell P, Ballinger CA, Jiang J, Wu Y, Thompson LJ, Höhfeld J, Patterson C. The co-chaperone CHIP regulates protein triage decisions mediated by heat-shock proteins. Nat Cell Biol. 2001;3:93-96.  [PubMed]  [DOI]  [Cited in This Article: ]
74.  Muller P, Hrstka R, Coomber D, Lane DP, Vojtesek B. Chaperone-dependent stabilization and degradation of p53 mutants. Oncogene. 2008;27:3371-3383.  [PubMed]  [DOI]  [Cited in This Article: ]
75.  Bird A. DNA methylation patterns and epigenetic memory. Genes Dev. 2002;16:6-21.  [PubMed]  [DOI]  [Cited in This Article: ]
76.  Bird A. Perceptions of epigenetics. Nature. 2007;447:396-398.  [PubMed]  [DOI]  [Cited in This Article: ]
77.  Calcagno DQ, Gigek CO, Chen ES, Burbano RR, Smith Mde A. DNA and histone methylation in gastric carcinogenesis. World J Gastroenterol. 2013;19:1182-1192.  [PubMed]  [DOI]  [Cited in This Article: ]
78.  Terry MB, Delgado-Cruzata L, Vin-Raviv N, Wu HC, Santella RM. DNA methylation in white blood cells: association with risk factors in epidemiologic studies. Epigenetics. 2011;6:828-837.  [PubMed]  [DOI]  [Cited in This Article: ]
79.  Matsusaka K, Funata S, Fukayama M, Kaneda A. DNA methylation in gastric cancer, related to Helicobacter pylori and Epstein-Barr virus. World J Gastroenterol. 2014;20:3916-3926.  [PubMed]  [DOI]  [Cited in This Article: ]
80.  Kobayashi D, Nomoto S, Kodera Y, Fujiwara M, Koike M, Nakayama G, Ohashi N, Nakao A. Suppressor of cytokine signaling 4 detected as a novel gastric cancer suppressor gene using double combination array analysis. World J Surg. 2012;36:362-372.  [PubMed]  [DOI]  [Cited in This Article: ]
81.  Li X, Cheung KF, Ma X, Tian L, Zhao J, Go MY, Shen B, Cheng AS, Ying J, Tao Q. Epigenetic inactivation of paired box gene 5, a novel tumor suppressor gene, through direct upregulation of p53 is associated with prognosis in gastric cancer patients. Oncogene. 2012;31:3419-3430.  [PubMed]  [DOI]  [Cited in This Article: ]
82.  Takamaru H, Yamamoto E, Suzuki H, Nojima M, Maruyama R, Yamano HO, Yoshikawa K, Kimura T, Harada T, Ashida M. Aberrant methylation of RASGRF1 is associated with an epigenetic field defect and increased risk of gastric cancer. Cancer Prev Res (Phila). 2012;5:1203-1212.  [PubMed]  [DOI]  [Cited in This Article: ]
83.  Balgkouranidou I, Karayiannakis A, Matthaios D, Bolanaki H, Tripsianis G, Tentes AA, Lianidou E, Chatzaki E, Fiska A, Lambropoulou M. Assessment of SOX17 DNA methylation in cell free DNA from patients with operable gastric cancer. Association with prognostic variables and survival. Clin Chem Lab Med. 2013;51:1505-1510.  [PubMed]  [DOI]  [Cited in This Article: ]
84.  Guo W, Dong Z, Guo Y, Chen Z, Kuang G, Yang Z. Methylation-mediated repression of GADD45A and GADD45G expression in gastric cardia adenocarcinoma. Int J Cancer. 2013;133:2043-2053.  [PubMed]  [DOI]  [Cited in This Article: ]
85.  Ling ZQ, Lv P, Lu XX, Yu JL, Han J, Ying LS, Zhu X, Zhu WY, Fang XH, Wang S. Circulating Methylated XAF1 DNA Indicates Poor Prognosis for Gastric Cancer. PLoS One. 2013;8:e67195.  [PubMed]  [DOI]  [Cited in This Article: ]
86.  Ooki A, Yamashita K, Yamaguchi K, Mondal A, Nishimiya H, Watanabe M. DNA damage-inducible gene, reprimo functions as a tumor suppressor and is suppressed by promoter methylation in gastric cancer. Mol Cancer Res. 2013;11:1362-1374.  [PubMed]  [DOI]  [Cited in This Article: ]
87.  Tanaka T, Nakamura J, Kitajima Y, Kai K, Miyake S, Hiraki M, Ide T, Koga Y, Noshiro H. Loss of trefoil factor 1 is regulated by DNA methylation and is an independent predictive factor for poor survival in advanced gastric cancer. Int J Oncol. 2013;42:894-902.  [PubMed]  [DOI]  [Cited in This Article: ]
88.  Feng G, Zhang Y, Yuan H, Bai R, Zheng J, Zhang J, Song M. DNA methylation of trefoil factor 1 (TFF1) is associated with the tumorigenesis of gastric carcinoma. Mol Med Rep. 2014;9:109-117.  [PubMed]  [DOI]  [Cited in This Article: ]
89.  Han J, Lv P, Yu JL, Wu YC, Zhu X, Hong LL, Zhu WY, Yu QM, Wang XB, Li P. Circulating methylated MINT2 promoter DNA is a potential poor prognostic factor in gastric cancer. Dig Dis Sci. 2014;59:1160-1168.  [PubMed]  [DOI]  [Cited in This Article: ]
90.  Han L, Dong Z, Wang C, Guo Y, Shen S, Kuang G, Guo W. Decreased expression and aberrant methylation of RASSF5A correlates with malignant progression of gastric cardia adenocarcinoma. Mol Carcinog. 2014;Epub ahead of print.  [PubMed]  [DOI]  [Cited in This Article: ]
91.  Kanda M, Nomoto S, Oya H, Hashimoto R, Takami H, Shimizu D, Sonohara F, Kobayashi D, Tanaka C, Yamada S. Decreased expression of prenyl diphosphate synthase subunit 2 correlates with reduced survival of patients with gastric cancer. J Exp Clin Cancer Res. 2014;33:88.  [PubMed]  [DOI]  [Cited in This Article: ]
92.  Kanda M, Shimizu D, Nomoto S, Hibino S, Oya H, Takami H, Kobayashi D, Yamada S, Inokawa Y, Tanaka C. Clinical significance of expression and epigenetic profiling of TUSC1 in gastric cancer. J Surg Oncol. 2014;110:136-144.  [PubMed]  [DOI]  [Cited in This Article: ]
93.  Li DX, Cai HY, Wang X, Feng YL, Cai SW. Promoter methylation of Raf kinase inhibitory protein: A significant prognostic indicator for patients with gastric adenocarcinoma. Exp Ther Med. 2014;8:844-850.  [PubMed]  [DOI]  [Cited in This Article: ]
94.  Otani K, Dong Y, Li X, Lu J, Zhang N, Xu L, Go MY, Ng EK, Arakawa T, Chan FK. Odd-skipped related 1 is a novel tumour suppressor gene and a potential prognostic biomarker in gastric cancer. J Pathol. 2014;234:302-315.  [PubMed]  [DOI]  [Cited in This Article: ]
95.  Zhang H, Song Y, Xia P, Cheng Y, Guo Q, Diao D, Wang W, Wu X, Liu D, Dang C. Detection of aberrant hypermethylated spastic paraplegia-20 as a potential biomarker and prognostic factor in gastric cancer. Med Oncol. 2014;31:830.  [PubMed]  [DOI]  [Cited in This Article: ]
96.  Kanda M, Shimizu D, Nomoto S, Takami H, Hibino S, Oya H, Hashimoto R, Suenaga M, Inokawa Y, Kobayashi D. Prognostic impact of expression and methylation status of DENN/MADD domain-containing protein 2D in gastric cancer. Gastric Cancer. 2015;18:288-296.  [PubMed]  [DOI]  [Cited in This Article: ]
97.  Yang Z, Li DM, Xie Q, Dai DQ. Protein expression and promoter methylation of the candidate biomarker TCF21 in gastric cancer. J Cancer Res Clin Oncol. 2015;141:211-220.  [PubMed]  [DOI]  [Cited in This Article: ]
98.  Hao Y, Chai KH, McLoughlin DM, Chan HY, Lau KF. Promoter characterization and genomic organization of the human X11β gene APBA2. Neuroreport. 2012;23:146-151.  [PubMed]  [DOI]  [Cited in This Article: ]
99.  Kanda M, Nomoto S, Oya H, Takami H, Hibino S, Hishida M, Suenaga M, Yamada S, Inokawa Y, Nishikawa Y. Downregulation of DENND2D by promoter hypermethylation is associated with early recurrence of hepatocellular carcinoma. Int J Oncol. 2014;44:44-52.  [PubMed]  [DOI]  [Cited in This Article: ]
100.  Marat AL, Dokainish H, McPherson PS. DENN domain proteins: regulators of Rab GTPases. J Biol Chem. 2011;286:13791-13800.  [PubMed]  [DOI]  [Cited in This Article: ]
101.  Liu W, Li X, Chu ES, Go MY, Xu L, Zhao G, Li L, Dai N, Si J, Tao Q. Paired box gene 5 is a novel tumor suppressor in hepatocellular carcinoma through interaction with p53 signaling pathway. Hepatology. 2011;53:843-853.  [PubMed]  [DOI]  [Cited in This Article: ]
102.  Nutt SL, Urbánek P, Rolink A, Busslinger M. Essential functions of Pax5 (BSAP) in pro-B cell development: difference between fetal and adult B lymphopoiesis and reduced V-to-DJ recombination at the IgH locus. Genes Dev. 1997;11:476-491.  [PubMed]  [DOI]  [Cited in This Article: ]
103.  Ohki R, Nemoto J, Murasawa H, Oda E, Inazawa J, Tanaka N, Taniguchi T. Reprimo, a new candidate mediator of the p53-mediated cell cycle arrest at the G2 phase. J Biol Chem. 2000;275:22627-22630.  [PubMed]  [DOI]  [Cited in This Article: ]
104.  Zhang Z, Li J, Lantry LE, Wang Y, Wiseman RW, Lubet RA, You M. p53 transgenic mice are highly susceptible to 1, 2-dimethylhydrazine-induced uterine sarcomas. Cancer Res. 2002;62:3024-3029.  [PubMed]  [DOI]  [Cited in This Article: ]
105.  Pogribny IP, Rusyn I. Role of epigenetic aberrations in the development and progression of human hepatocellular carcinoma. Cancer Lett. 2014;342:223-230.  [PubMed]  [DOI]  [Cited in This Article: ]
106.  Toiyama Y, Okugawa Y, Goel A. DNA methylation and microRNA biomarkers for noninvasive detection of gastric and colorectal cancer. Biochem Biophys Res Commun. 2014;455:43-57.  [PubMed]  [DOI]  [Cited in This Article: ]
107.  Wang J, Song YX, Wang ZN. Non-coding RNAs in gastric cancer. Gene. 2015;560:1-8.  [PubMed]  [DOI]  [Cited in This Article: ]
108.  Aravalli RN, Steer CJ, Cressman EN. Molecular mechanisms of hepatocellular carcinoma. Hepatology. 2008;48:2047-2063.  [PubMed]  [DOI]  [Cited in This Article: ]
109.  Li BS, Zhao YL, Guo G, Li W, Zhu ED, Luo X, Mao XH, Zou QM, Yu PW, Zuo QF. Plasma microRNAs, miR-223, miR-21 and miR-218, as novel potential biomarkers for gastric cancer detection. PLoS One. 2012;7:e41629.  [PubMed]  [DOI]  [Cited in This Article: ]
110.  Tsai KW, Liao YL, Wu CW, Hu LY, Li SC, Chan WC, Ho MR, Lai CH, Kao HW, Fang WL. Aberrant expression of miR-196a in gastric cancers and correlation with recurrence. Genes Chromosomes Cancer. 2012;51:394-401.  [PubMed]  [DOI]  [Cited in This Article: ]
111.  Valladares-Ayerbes M, Reboredo M, Medina-Villaamil V, Iglesias-Díaz P, Lorenzo-Patiño MJ, Haz M, Santamarina I, Blanco M, Fernández-Tajes J, Quindós M. Circulating miR-200c as a diagnostic and prognostic biomarker for gastric cancer. J Transl Med. 2012;10:186.  [PubMed]  [DOI]  [Cited in This Article: ]
112.  Wang M, Gu H, Wang S, Qian H, Zhu W, Zhang L, Zhao C, Tao Y, Xu W. Circulating miR-17-5p and miR-20a: molecular markers for gastric cancer. Mol Med Rep. 2012;5:1514-1520.  [PubMed]  [DOI]  [Cited in This Article: ]
113.  Cui MH, Hou XL, Lei XY, Mu FH, Yang GB, Yue L, Fu Y, Yi GX. Upregulation of microRNA 181c expression in gastric cancer tissues and plasma. Asian Pac J Cancer Prev. 2013;14:3063-3066.  [PubMed]  [DOI]  [Cited in This Article: ]
114.  Deng M, Tang HL, Lu XH, Liu MY, Lu XM, Gu YX, Liu JF, He ZM. miR-26a suppresses tumor growth and metastasis by targeting FGF9 in gastric cancer. PLoS One. 2013;8:e72662.  [PubMed]  [DOI]  [Cited in This Article: ]
115.  Li C, Li JF, Cai Q, Qiu QQ, Yan M, Liu BY, Zhu ZG. miRNA-199a-3p in plasma as a potential diagnostic biomarker for gastric cancer. Ann Surg Oncol. 2013;20 Suppl 3:S397-S405.  [PubMed]  [DOI]  [Cited in This Article: ]
116.  Song J, Bai Z, Zhang J, Meng H, Cai J, Deng W, Bi J, Ma X, Zhang Z. Serum microRNA-21 levels are related to tumor size in gastric cancer patients but cannot predict prognosis. Oncol Lett. 2013;6:1733-1737.  [PubMed]  [DOI]  [Cited in This Article: ]
117.  Wang W, Li F, Zhang Y, Tu Y, Yang Q, Gao X. Reduced expression of miR-22 in gastric cancer is related to clinicopathologic characteristics or patient prognosis. Diagn Pathol. 2013;8:102.  [PubMed]  [DOI]  [Cited in This Article: ]
118.  Yu X, Luo L, Wu Y, Yu X, Liu Y, Yu X, Zhao X, Zhang X, Cui L, Ye G. Gastric juice miR-129 as a potential biomarker for screening gastric cancer. Med Oncol. 2013;30:365.  [PubMed]  [DOI]  [Cited in This Article: ]
119.  Chen B, Huang T, Jiang J, Lv L, Li H, Xia S. miR-141 suppresses proliferation and motility of gastric cancer cells by targeting HDGF. Mol Cell Biochem. 2014;388:211-218.  [PubMed]  [DOI]  [Cited in This Article: ]
120.  Chu D, Zhao Z, Li Y, Li J, Zheng J, Wang W, Zhao Q, Ji G. Increased microRNA-630 expression in gastric cancer is associated with poor overall survival. PLoS One. 2014;9:e90526.  [PubMed]  [DOI]  [Cited in This Article: ]
121.  Fu Z, Qian F, Yang X, Jiang H, Chen Y, Liu S. Circulating miR-222 in plasma and its potential diagnostic and prognostic value in gastric cancer. Med Oncol. 2014;31:164.  [PubMed]  [DOI]  [Cited in This Article: ]
122.  Gu W, Gao T, Shen J, Sun Y, Zheng X, Wang J, Ma J, Hu XY, Li J, Hu MJ. MicroRNA-183 inhibits apoptosis and promotes proliferation and invasion of gastric cancer cells by targeting PDCD4. Int J Clin Exp Med. 2014;7:2519-2529.  [PubMed]  [DOI]  [Cited in This Article: ]
123.  Huang D, Wang H, Liu R, Li H, Ge S, Bai M, Deng T, Yao G, Ba Y. miRNA27a is a biomarker for predicting chemosensitivity and prognosis in metastatic or recurrent gastric cancer. J Cell Biochem. 2014;115:549-556.  [PubMed]  [DOI]  [Cited in This Article: ]
124.  Li BS, Zuo QF, Zhao YL, Xiao B, Zhuang Y, Mao XH, Wu C, Yang SM, Zeng H, Zou QM. MicroRNA-25 promotes gastric cancer migration, invasion and proliferation by directly targeting transducer of ERBB2, 1 and correlates with poor survival. Oncogene. 2015;34:2556-2565.  [PubMed]  [DOI]  [Cited in This Article: ]
125.  Liu X, Yu H, Cai H, Wang Y. The expression and clinical significance of miR-132 in gastric cancer patients. Diagn Pathol. 2014;9:57.  [PubMed]  [DOI]  [Cited in This Article: ]
126.  Peng WZ, Ma R, Wang F, Yu J, Liu ZB. Role of miR-191/425 cluster in tumorigenesis and diagnosis of gastric cancer. Int J Mol Sci. 2014;15:4031-4048.  [PubMed]  [DOI]  [Cited in This Article: ]
127.  Sun J, Song Y, Wang Z, Wang G, Gao P, Chen X, Gao Z, Xu H. Clinical significance of promoter region hypermethylation of microRNA-148a in gastrointestinal cancers. Onco Targets Ther. 2014;7:853-863.  [PubMed]  [DOI]  [Cited in This Article: ]
128.  Suzuki R, Yamamoto E, Nojima M, Maruyama R, Yamano HO, Yoshikawa K, Kimura T, Harada T, Ashida M, Niinuma T. Aberrant methylation of microRNA-34b/c is a predictive marker of metachronous gastric cancer risk. J Gastroenterol. 2014;49:1135-1144.  [PubMed]  [DOI]  [Cited in This Article: ]
129.  Tan Z, Jiang H, Wu Y, Xie L, Dai W, Tang H, Tang S. miR-185 is an independent prognosis factor and suppresses tumor metastasis in gastric cancer. Mol Cell Biochem. 2014;386:223-231.  [PubMed]  [DOI]  [Cited in This Article: ]
130.  Wang H, Wang L, Wu Z, Sun R, Jin H, Ma J, Liu L, Ling R, Yi J, Wang L. Three dysregulated microRNAs in serum as novel biomarkers for gastric cancer screening. Med Oncol. 2014;31:298.  [PubMed]  [DOI]  [Cited in This Article: ]
131.  Wang YW, Shi DB, Chen X, Gao C, Gao P. Clinicopathological significance of microRNA-214 in gastric cancer and its effect on cell biological behaviour. PLoS One. 2014;9:e91307.  [PubMed]  [DOI]  [Cited in This Article: ]
132.  Zeng Q, Jin C, Chen W, Xia F, Wang Q, Fan F, Du J, Guo Y, Lin C, Yang K. Downregulation of serum miR-17 and miR-106b levels in gastric cancer and benign gastric diseases. Chin J Cancer Res. 2014;26:711-716.  [PubMed]  [DOI]  [Cited in This Article: ]
133.  Zhang R, Wang W, Li F, Zhang H, Liu J. MicroRNA-106b~25 expressions in tumor tissues and plasma of patients with gastric cancers. Med Oncol. 2014;31:243.  [PubMed]  [DOI]  [Cited in This Article: ]
134.  Zhu ED, Li N, Li BS, Li W, Zhang WJ, Mao XH, Guo G, Zou QM, Xiao B. miR-30b, down-regulated in gastric cancer, promotes apoptosis and suppresses tumor growth by targeting plasminogen activator inhibitor-1. PLoS One. 2014;9:e106049.  [PubMed]  [DOI]  [Cited in This Article: ]
135.  Han TS, Hur K, Xu G, Choi B, Okugawa Y, Toiyama Y, Oshima H, Oshima M, Lee HJ, Kim VN. MicroRNA-29c mediates initiation of gastric carcinogenesis by directly targeting ITGB1. Gut. 2015;64:203-214.  [PubMed]  [DOI]  [Cited in This Article: ]
136.  Xu YJ, Fan Y. MiR-215/192 participates in gastric cancer progression. Clin Transl Oncol. 2015;17:34-40.  [PubMed]  [DOI]  [Cited in This Article: ]
137.  Gaur A, Jewell DA, Liang Y, Ridzon D, Moore JH, Chen C, Ambros VR, Israel MA. Characterization of microRNA expression levels and their biological correlates in human cancer cell lines. Cancer Res. 2007;67:2456-2468.  [PubMed]  [DOI]  [Cited in This Article: ]
138.  Wu J, Qian J, Li C, Kwok L, Cheng F, Liu P, Perdomo C, Kotton D, Vaziri C, Anderlind C. miR-129 regulates cell proliferation by downregulating Cdk6 expression. Cell Cycle. 2010;9:1809-1818.  [PubMed]  [DOI]  [Cited in This Article: ]
139.  Farhana L, Dawson MI, Murshed F, Das JK, Rishi AK, Fontana JA. Upregulation of miR-150* and miR-630 induces apoptosis in pancreatic cancer cells by targeting IGF-1R. PLoS One. 2013;8:e61015.  [PubMed]  [DOI]  [Cited in This Article: ]
140.  Zhang Y, Ma M, Liu W, Ding W, Yu H. Enhanced expression of long noncoding RNA CARLo-5 is associated with the development of gastric cancer. Int J Clin Exp Pathol. 2014;7:8471-8479.  [PubMed]  [DOI]  [Cited in This Article: ]
141.  Fatica A, Bozzoni I. Long non-coding RNAs: new players in cell differentiation and development. Nat Rev Genet. 2014;15:7-21.  [PubMed]  [DOI]  [Cited in This Article: ]
142.  Dong L, Qi P, Xu MD, Ni SJ, Huang D, Xu QH, Weng WW, Tan C, Sheng WQ, Zhou XY. Circulating CUDR, LSINCT-5 and PTENP1 long noncoding RNAs in sera distinguish patients with gastric cancer from healthy controls. Int J Cancer. 2015;Epub ahead of print.  [PubMed]  [DOI]  [Cited in This Article: ]
143.  Panzitt K, Tschernatsch MM, Guelly C, Moustafa T, Stradner M, Strohmaier HM, Buck CR, Denk H, Schroeder R, Trauner M. Characterization of HULC, a novel gene with striking up-regulation in hepatocellular carcinoma, as noncoding RNA. Gastroenterology. 2007;132:330-342.  [PubMed]  [DOI]  [Cited in This Article: ]
144.  Emadi-Andani E, Nikpour P, Emadi-Baygi M, Bidmeshkipour A. Association of HOTAIR expression in gastric carcinoma with invasion and distant metastasis. Adv Biomed Res. 2014;3:135.  [PubMed]  [DOI]  [Cited in This Article: ]
145.  Hu Y, Wang J, Qian J, Kong X, Tang J, Wang Y, Chen H, Hong J, Zou W, Chen Y. Long noncoding RNA GAPLINC regulates CD44-dependent cell invasiveness and associates with poor prognosis of gastric cancer. Cancer Res. 2014;74:6890-6902.  [PubMed]  [DOI]  [Cited in This Article: ]
146.  Chen WM, Huang MD, Kong R, Xu TP, Zhang EB, Xia R, Sun M, De W, Shu YQ. Antisense Long Noncoding RNA HIF1A-AS2 Is Upregulated in Gastric Cancer and Associated with Poor Prognosis. Dig Dis Sci. 2015;60:1655-1662.  [PubMed]  [DOI]  [Cited in This Article: ]
147.  Hu Y, Pan J, Wang Y, Li L, Huang Y. Long noncoding RNA linc-UBC1 is negative prognostic factor and exhibits tumor pro-oncogenic activity in gastric cancer. Int J Clin Exp Pathol. 2015;8:594-600.  [PubMed]  [DOI]  [Cited in This Article: ]
148.  Kim T, Cui R, Jeon YJ, Lee JH, Lee JH, Sim H, Park JK, Fadda P, Tili E, Nakanishi H. Long-range interaction and correlation between MYC enhancer and oncogenic long noncoding RNA CARLo-5. Proc Natl Acad Sci USA. 2014;111:4173-4178.  [PubMed]  [DOI]  [Cited in This Article: ]
149.  Thrash-Bingham CA, Tartof KD. aHIF: a natural antisense transcript overexpressed in human renal cancer and during hypoxia. J Natl Cancer Inst. 1999;91:143-151.  [PubMed]  [DOI]  [Cited in This Article: ]
150.  Rossignol F, Vaché C, Clottes E. Natural antisense transcripts of hypoxia-inducible factor 1alpha are detected in different normal and tumour human tissues. Gene. 2002;299:135-140.  [PubMed]  [DOI]  [Cited in This Article: ]
151.  Bertozzi D, Iurlaro R, Sordet O, Marinello J, Zaffaroni N, Capranico G. Characterization of novel antisense HIF-1α transcripts in human cancers. Cell Cycle. 2011;10:3189-3197.  [PubMed]  [DOI]  [Cited in This Article: ]
152.  Resende C, Thiel A, Machado JC, Ristimäki A. Gastric cancer: basic aspects. Helicobacter. 2011;16 Suppl 1:38-44.  [PubMed]  [DOI]  [Cited in This Article: ]