Topic Highlight Open Access
Copyright ©The Author(s) 2016. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Jul 14, 2016; 22(26): 5896-5908
Published online Jul 14, 2016. doi: 10.3748/wjg.v22.i26.5896
Exploring the role of molecular biomarkers as a potential weapon against gastric cancer: A review of the literature
Marwa Matboli, Nourhan Hossam, Alaa Habieb, Oncology Diagnostic Unit, Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Ain Shams University, Cairo 113331, Egypt
Sarah El-Nakeep, Hepatology and Gastroenterology Unit, Internal Medicine Department, Faculty of Medicine, Ain Shams University, Cairo 113331, Egypt
Ali E Ebrahim, Ziad Nagy, Ahmed EM Azazy, Armed Forces College of Medicine, Cairo 113331, Egypt
Omar Abdel-Rahman, Clinical Oncology Department, Faculty of Medicine, Ain Shams University, Cairo 113331, Egypt
Author contributions: Matboli M and El-Nakeep S planned the study and drafted the review; HossamN, Habieb A, Azazy AEM, Ebrahim AE and Nagy Z have collected the data and drafted the review; Abdel-Rahman O has revised the review and analyzed the collected literature.
Conflict-of-interest statement: The authors declare that they have no competing interests.
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. Omar Abdel-Rahman, Clinical Oncology and Nuclear Medicine Department, Faculty of Medicine, Ain Shams University, Cairo 113331, Egypt. omar.abdelrhman@med.asu.edu.eg
Telephone: +2-100-8541806 Fax: +2-100-26858397
Received: March 15, 2016
Peer-review started: March 18, 2016
First decision: May 12, 2016
Revised: May 25, 2016
Accepted: June 13, 2016
Article in press: June 13, 2016
Published online: July 14, 2016

Abstract

Gastric cancer (GC) is a global health problem and a major cause of cancer-related death with high recurrence rates ranging from 25% to 40% for GC patients staging II-IV. Unfortunately, while the majority of GC patients usually present with advanced tumor stage; there is still limited evidence-based therapeutic options. Current approach to GC management consists mainly of; endoscopy followed by, gastrectomy and chemotherapy or chemo-radiotherapy. Recent studies in GC have confirmed that it is a heterogeneous disease. Many molecular characterization studies have been performed in GC. Recent discoveries of the molecular pathways underlying the disease have opened the door to more personalized treatment and better predictable outcome. The identification of molecular markers is a useful tool for clinical managementin GC patients, assisting in diagnosis, evaluation of response to treatment and development of novel therapeutic modalities. While chemotherapeutic agents have certain physiological effects on the tumor cells, the prediction of the response is different from one type of tumor to the other. The specificity of molecular biomarkers is a principal feature driving their application in anticancer therapies. Here we are trying to focus on the role of molecular pathways of GC and well-established molecular markers that can guide the therapeutic management.

Key Words: Gastric cancer, Molecular therapy, Targeted therapy, Biomarkers, Bioinformatics

Core tip: We tried to highlight the role of molecular biomarkers as a predictor to chemotherapeutic response to existing regimens, aiming for better personalized therapy. Also we provided a summary of molecular markers that may aid in the development of rational therapeutic options for gastric cancer (GC) patients, aiming for improving their outcomes. However, among the plethora of agents targeting VEGF, EGFR, HER-2, IGF and mTOR pathways, trastuzumab and ramucirumab have been the only approved therapeutic options for use in advanced GC. Despite having many promising studies in their early stages, a lot have failed to prove their effectiveness in GC on the long run.



INTRODUCTION

Gastric cancer (GC) is the second cause of cancer mortality[1] and the fifth most common malignancy in the world, fifty percent of the cases are from Eastern Asia[2], where China has the highest incidence[3]. These statistics are considered to be an improvement compared to the very first estimates in 1975 where GC was the most common neoplasm[4]. In spite of the apparent global decline in incidence and mortality estimates of GC in age-standardized figures, the absolute number of GC cases remains stable or even increasing[5].

The five year survival rates remain disappointing despite improvements in the diagnosis and treatment of GC cases because they are usually diagnosed at an advanced stage which is rarely curable, this is in addition to the quite high rate of recurrence[6]. Surgery has been the cornerstone in GC treatment[7], however a high rate of intra-abdominal metastasis (80%), loco-regional (40%-80%) as well as distant (20%-40%) recurrences has been reported and luckily a survival benefit has been observed from the addition of chemotherapy or radiotherapy to surgery[8-12].

The treatment of GC is dependent on the type of cancer tissue, the TNM staging and the general condition of the patient[1]. Metastatic GC gives us fewer options in dealing with the disease, aiming for palliative rather than curative goal[3].

The standard treatment options for GC include mainly: adjuvant chemotherapy or adjuvant chemo-radiation with perioperative chemotherapy. On the other hand, several recent studies evaluated the use of neoadjuvant chemotherapy solely in GC[13], which plays a role in down-staging the disease, in addition to the eradication of any possible micrometastasis[14-16].

While the chemotherapeutic agents that have proven effectiveness in the treatment of GC are more than a few, we cannot ignore the fact that the targeted therapy for GC is still very limited, mainly to vascular endothelial growth factor (VEGF) pathway - and HER2 - targeted agents. Recent achievements in the field of epigenetics and genetic background of the disease may enhance our chances of targeted therapeutic options in GC[17].

On the other hand, multiple molecular biomarkers had shown their potential efficacy as diagnostic and prognostic tools in GC but they still need further validation to be used in the day-to-day clinical practice. Up till the time being, the only used markers for GC in clinical practice are carcino-embryonic antigens; CA 19-9, CA-50[18] and CA-72[19], which lack the high sensitivity and specificity that is needed in assessing diagnosis and prognosis of GC, making their efficacy questionable.

But as the link between the new era of the molecular markers and the treatment options is increasing; where some predict response to chemotherapy while others predict the post treatment survival or recurrence; the current review focuses on the role of molecular aberrations in affecting the therapeutic guidelines, either used for predicting the outcome of specific therapeutic agents or exploited as a therapeutic target in the tumor cells.

MOLECULAR BIOMARKERS PREDICTING THE TREATMENT RESPONSE

Prediction of chemo-resistance is an important goal in personalized medicine, where each patient is treated according to his epigenetic and genetic background. Pharmacogenomics is a rapidly growing field with hope of decreasing the burden on both the patient; by adjusting the dose, type and combination of drugs used; and the burden of cost on the healthcare system[20,21].

Multiple genetic and epigenetic markers have been shown to have a predictive value in GC therapy, although their use is limited in the routine management of the disease, where treatment decision depends mainly on the clinical staging of the patient[20] (Table 1).

Table 1 Showing the molecular markers used in gastric cancer pharmacogenomics.
Marker typeNameDrug predictedPredicted drug effect
Genetic markers13 gene signature[48]5-FUSensitivity or resistance to 5-FU
Genetic markersMRP4[49]CisplatinDDP resistance
Genetic markersMetallothionein-IG and HBEGF[25]CisplatinDDP resistance
Genetic markersDihydropyrimidine5-FU5-FU resistance
Dehydrogenase and HB-EGF-like growth factor genes[25]
Genetic markersPanel of genes[26]DoxorubicinPredicts response to chemotherapy
Genetic markersDihydropyrimidine5-FU5-FU resistance
Dehydrogenase and HB-EGF-like growth factor genes[25]
Genetic markersTP53 codon 72 polymorphism[50]Paclitaxel and cisplatinCertain genotypes predict response to combination therapy
lncRNAlncRNA MRUL[36]Multiple chemotherapeutic drugsMultidrug resistance
Epigenetic MarkersMethylation BMP4[38]CisplatinHigh expression predicts resistance to the drug
Epigenetic MarkersPromoter methylation of RPRM[39]CDDP and 5-FUPrediction of response to treatment
Epigenetic markersMethylation of BNIP3 and DAPK[37]Fluoropyrimidine-based chemotherapyMethylation predicts lower response to chemotherapy
miRNAmiRNA27a[34]Fluoropyrimidine combined with oxaliplatin or paclitaxilPrediction of response to treatment
MicroRNA58 signature mi-RNA; among them: let-7g, miR-342, miR-16, miR-181, miR-1, and miR-34[33]Cisplatin and 5-FUChemotherapeutic response
Protein markersThymidylate synthetase (TS) and Dihydropyrimidine dehydrogenase (DPD)[27,40]5-FUCorrelation with tumor sensitivity to 5-FU
Serum proteinAMBP[41]paclitaxel–capecitabinePredicts response to chemotherapy
Tissue proteinFOXM1[43]DocetaxelResistance to Docetaxel
Transcription factor
Protein markersRibosomal proteins S13 and L23[47]vincristine, adriamycin, and 5-FUMultidrug resistance by inhibition of chemotherapy related cell death and detoxification system
Serum protein (ELISA)REG4[42]5-FUResistance to 5-FU containing regimens
Protein markersClass III β tubulin serum level[45,46]Paclitaxel plus capecitabinePrediction of response to treatment
Genetic markers

Lin et al[22] compared the already published gene expression profiling signatures in GC as well as the more integrated genomic features of GC from gene expression, chromosomal instability, somatic mutation, and DNA methylation. Moreover, they identified the consensus patterns across these signatures, the biological functions and the underlying molecular pathways[22,23]. Tanaka et al[23] identified the precise prediction models of in vitro activity for 8 anticancer drugs (5-FU, TXL, CDDP, DOX, CPT-11, MMC, SN-38, and TXT), along with individual clinical responses to 5-FU using cDNA microarray analysis[24].

Suganuma et al[25] reported that metallothionein-IG and heparin-binding epidermal growth factor-like growth factor (HB-EGF), glutathione-S-transferase and cyclooxygenase-2 genes were potential candidate cisplatin-resistance-related genes by oligonucleotide microarrays. For 5-FU resistance, dihydropyrimidine dehydrogenase (DPD) and HB-EGF-like growth factor genes were also suggested to be resistance-related genes[25]. Doxorubicin response has also been linked to panel of genes including; ADAM22, CYR61, FN1, SPHK1 and GNAI1 by real-time RT-PCR in one study[26], but the main concern of most of these genetic signature studies is that the number of GC samples used for validation were always small in number, which means that there is a long way to go till the actual incorporation of these markers into the clinical practice.

A link was discovered between the response to cisplatin therapy and Multi-drug resistance-associated protein (MRP): when the phenotype of DDP resistance was reversed by lowering the MRP4 expression with small interfering RNA technique in GC cell line[27].

Moreover, genetic polymorphism was linked to the response of 5-FU and cisplatin in two studies, where rs715572 and rs5754312 were linked to survival of patients treated with 5-FU + cisplatin[28]. Also paclitaxel and cisplatin treatment response was predicted with TP53 codon 72 SNP[29].

A group of researchers used gene expression data to describe four molecular subtypes of GC linked to disease progression and prognosis. The mesenchymal-like type with highest recurrence frequency (63%) of the four subtypes; microsatellite-unstable tumors are hyper-mutated displaying the best overall prognosis and the lowest frequency of recurrence (22%) of the four subtypes; tumor protein 53 (TP53)-active and TP53-inactive types include patients with intermediate prognosis and recurrence rates (with respect to the other two subtypes)[30].

Scientists proposed a molecular classification dividing GC into four subtypes: Epstein-Barr virus positive tumors with recurrent PIK3CA mutations, extreme DNA hypermethylation, and amplification of JAK2, CD274 and PDCD1LG2; microsatellite unstable tumors with elevated mutation rates, including mutations of genes encoding targetable oncogenic signaling proteins; genomically stable tumors, with mutations of RHO-family GTPase-activating proteins; and tumors with chromosomal instability with marked aneuploidy and amplification of receptor tyrosine kinases. Identification of these molecular subtypes provides an efficient roadmap for patient stratification and targeted therapies[31].

Epigenetic markers

MicroRNA: MicroRNA was linked to the resistance to trastuzumab in one study, where it was shown that miR-21/PTEN pathway may have a regulatory effect on the treatment response[32]. MicroRNA let-7i might predict the pathologic response to neoadjuvant chemotherapy[29]. Also 58 signature mi-RNAs were found to predict the chemotherapeutic response of cisplatin/fluorouracil; among the apoptosis inducers are let-7g, miR-342, miR-16, miR-181, miR-1, and miR-34[33]. miRNA-27a higher expression predicts resistance to treatment with fluoropyrimidine-containing therapy[34].

Long non coding RNAs: Long non coding RNAs (lncRNAs) are potential biomarkers for GC especially those in blood and gastric secretions which offer a minimally invasive route[35]. But the tissue samples are still the main site of research; where lncRNA MRUL (MDR-related and up regulated lncRNA) was associated with multi-drug chemotherapeutic resistance[36].

Methylation related biomarkers: Bcl-2/adenovirus E1B 19 kDa-interacting protein 3 and death associated protein kinase DAPK methylation predicts lower response to fluoropyrimidine-based chemotherapy[37].

Decreased methylation of the Bone morphogenic protein 4 (BMP4) genes will lead to increased expression of the secreted protein, which is correlated with cisplatin resistance. BMP4 is highly expressed in cisplatin-resistant tissues and cisplatin sensitization was markedly increased with genetically targeting of BMP4 resulting in its inhibition[38].

A study showed that increased promoter methylation will cause increased expression of Reprimo (a highly glycosylated cellular protein) which was associated with a lower response to cisplatin and 5-FU chemotherapy, in addition the Reprimo Knockdown is associated with tumor suppression effect[39].

Protein markers

Cellular enzymatic activity: Cellular enzymatic activity was linked to the chemotherapeutic resistance where thymidylate synthetase (TS) and DPD were associated with 5-FU tumor sensitivity[27,40].

Cellular proteins: (1) AMBP (Alpha-1-Microglobulin/Bikunin Precursor) protein in serum was shown to predict the chemotherapeutic response to paclitaxel-capecitabine[41]; (2) Regenerating gene family, member 4 (Reg IV or REG4) predicted resistance to 5-FU containing regimens[42]; (3) As for tissue proteins; Forkhead Box M1 Transcription Factor (FOXM1) was shown to predict resistance to docetaxel[43]; (4) Increased expression of β-tubulin III protein (TUBB3) in serum has been linked to taxane resistance in non small cell lung cancer[24] and ovarian carcinoma[44]; moreover, in a study on Chinese patients with advanced GC it was shown to predict lower response of GC to paclitaxel plus capecitabine[45] which was confirmed later in another study[46]; and (5) Ribosomal proteins: It was found that genetically unregulated ribosomal proteins S13 and L23 enhances vincristine, adriamycin, and 5-FU resistance by inhibition of cell death and detoxification systems induced by chemotherapy[47].

Thus, a number of predictive biomarkers have been extensively evaluated in the setting of GC systemic therapy. However, the vast majority of these markers were derived from small scale retrospective studies; and thus, we cannot recommend incorporating any of these markers into routine practice except after careful assessment within the setting of prospective controlled clinical trials.

MOLECULAR ABERRATIONS AS POTENTIAL THERAPEUTIC TARGETS
Tumor angiogenesis inhibition

Anti-VEGFR mAbs (Ramucirumab): VEGF has long been recognized as a key regulatory pathway of angiogenesis and thus several therapeutic agents were developed to target VEGF including neutralizing antibodies to VEGF or its receptor in GC[48-52] (Table 1). Several studies reported that the expression of VEGF and SSTR was associated with progression of GC[53,54]. A research group used a mouse model in which VEGF-A was expressed via adenovirus, enabling a stromal response marked by immune infiltration and angiogenesis, and identified specific stromal gene expression signatures to discover predictive biomarkers of therapeutic response, especially to immunotherapy and antiangiogenic agents[55]. Ramucirumab is a fully humanized IgG1 monoclonal antibody targeting VEGFR2 thus antagonizing VEGF-A, VEGF-C and VEGF-D leading to a decrease of endothelial cell proliferation, migration and tumor vascularity, also decreasing lymphatic penetration, growth and metastasis to regional lymph node[56,57]. Its efficacy as a second line treatment of advanced GC has been proven following the recent publication of two phase III studies (either alone or in combination with paclitaxel), and in both studies it had shown a clear overall survival benefit vs the control regimen (placebo in one study and paclitaxel monotherapy in another study)[58,59]. Several trials are still ongoing to validate its effect in earlier stages of GC[60].

Anti-VEGF mABs (Bevacizumab): Bevacizumab has been evaluated for advanced GC in multiple phase II and III studies (3 phase II and 2 phase III); but unfortunately, the results were disappointingly negative in all these studies[61-65].

Direct multi-Tyrosine kinase inhibitors (sorafenib, regorafenib, suntinib, axitinib, dovitinib, apatinib, erlotinib, gefitinib, dacomitinib, afatinib)

Tyrosine kinase (TK) inhibition can be conducted by various drugs and most of the angiogenic factors and epidermal growth factors share a common end-pathway incorporating the TK in their receptors[20]. Several research groups found that tyrosine kinase with immunoglobulin-like and EGF-like domains 1 (TIE-1) and mitogen-activated protein kinase kinase 4, might serve as promising molecular biomarkers for GC prognosis. On the other hand, overexpression of TIE-1 kinase in GC patients was associated with reduced survival rates[66-68].

Here, we give an overview on the multi-kinase TKIs which are mostly oral drugs (Table 2). Overall, they have weak to moderate activity and none of them has been approved yet in GC[69,70].

Table 2 Molecularly-targeted drugs evaluated in clinical trials for gastric cancer.
Drug nameTypeMolecular effectPrimary cancer which it is usedEffect on gastric cancer in studies
Trastuzumab[17]Fully humanized monoclonal antibodyAnti-HER-2 receptor proteinBreast cancerEffective
First approved molecular therapy
Sunitinib[17]Oral multi-tyrosine kinase inhibitorAnti- VEGF, PDGF and KIT receptorsGastrintestinal stromal tumors, renal cell carcinoma and pancreatic neuroendocrine tumorsLimited therapeutic effect
Bevacizumab[131,132]Fully humanized monoclonal antibodyAnti-VEGFColorectal cancer, non small cell lung cancer and breast cancerGives better survival in peritoneal metastatic disease or combined with anti-HER-2 therapy
Lapatinib[17]Oral dual tyrosine kinase inhibitorAnti-EGFR and HER-2HER-2 positive advanced breast cancerNot effective
Everolimus[17]Oral mTOR inhibitorAnti-intracellular receptor FKBP12Renal cancerEffective in advanced gastric cancer
Ramucirumab[17]Fully humanized IgG1 monoclonal antibodyAnti-VEGFR-2Gastric and lung cancerEffective approved
Cetuximab[17]Monoclonal IgG antibodyAnti-EGFRColorectal cancerNot effective
Panitumumab[17]Fully humanized IgG2 monoclonal antibodyAnti-EGFRAdvanced colorectal cancerNot effective
Gefitinib[133]Tyrosine kinase inhibitorAnti- EGFREGFR mutation positive lung cancerNot effective
Matuzumab[134]Fully humanized monoclonal antibodyAnti-EGFRNot yet approved in any other indicationModerately effective
Tivantinib[94]Tyrosine kinase inhibitorSelective c-Met inhibitorNot yet approved in any other indicationmoderately effective
Onartuzumab[135]Fully humanized monoclonal antibodyAnti-extracellulardomain of the tyrosine kinase receptor METNot yet approved in any other indicationNot effective
Regorafenib[73]Tyrosine kinase inhibitorAnti-angiogenic factorGastrointestinal stromal tumorsFound effective when tested on xenograft model with GC
Pembrolizumab[100]Monoclonal antibodyPD-1 inhibitorAdvanced melanoma, advanced lung cancerPromising phase IB results.
Phase III results are awaited
Apatinib[72]Tyrosine kinase inhibitorMultikinase inhibitorNot yet approved in any other indicationShown to be effective in a phase III Chinese study

The only successful exception of this group of agents has been Apatinib, which is a selective tyrosine kinase inhibitor against VEGF-2. A phase II trial of monotherapy in GC showed a favorable response[71]. Later in the phase III trial, the drug showed a good safety profile and beneficial effect with improved overall survival and progression-free survival in advanced GC that was refractory to other lines of therapy[72].

Regorafenib which is a multi-kinase inhibitor used in advanced cancers, was tested on xenograft model of patients with GC, it gave positive results regarding effectiveness for further research[73].

Suntinib and sorafenib are multi-kinase inhibitors (VEGF, PDGF, KIT) that have proven to be effective in a number of solid tumors, but when tested as a monotherapy or in combination in advanced GC showed a limited - if any - efficacy[71].

Epidermal growth factor receptor inhibition based agents

EGFR is a cell surface receptor that is activated by EGF and transforming growth factor alpha. Upon activation it initiates a downstream signaling through intracellular tyrosine kinase domain resulting in DNA synthesis and cell proliferation. Among the family of EGFRs; EGFR-1 and HER-2 which are currently targets for development of drugs for GC treatment[60]. Recent studies reported that serum HER2 levels are highly specific and demonstrated moderate diagnostic performance for HER2 tissue status in GC[74-76].

Anti-EGFR mAbs (cetuximab/panitumumab): EGFR is commonly over expressed in gastrointestinal malignancies. Its over expression is associated with a more aggressive phenotype and poorer survival, which suggests that EGFR may be a rational therapeutic target[77]. Cetuximab is a humanized monoclonal antibody against EGFR and it is the most investigated in GC[78]. Several randomized studies comparing the addition of cetuximab to conventional chemotherapies concluded that there was no clinically significant benefit associated with adding cetuximab to the conventional chemotherapies[78].

Anti-HER-2 mAbs (Trastuzumab): HER-2, also called ERB-2; is a tyrosine kinase receptor that when mutated exerts an oncogenic effect on cell proliferation, differentiation, programmed death and mobility; while mostly connected to breast cancer, it has proven to be the culprit in other tumors as well[79]. HER-2 is related to increased invasiveness and metastatic potential of the tumor[17].

HER-2 over expression has been reported in 10%-38% of GC patients[80]. Trastuzumab is a fully humanized anti-HER-2 monoclonal antibody that is already widely accepted as a standard agent for HER-2-positive breast cancer[81]. It is the first biological treatment to show improved survival in case of GC, immunohistochemistry score of more than +3 should receive the treatment, while > +2 should repeat the test using in situ hybridization[17].

Lapatinib is another HER-2 antagonist that is used in breast cancer resistant to trastuzumab therapy, its use in GC is not supported by evidence as it did not show any activity in GC patients in many studies[17].

PI3K/AKT/mTOR (mammalian target of rapamycin) pathway inhibitors

mTOR is active in 60% of GC cases while PI3K/Akt is active in 30% of GC cases[82]. Intriguingly, most of the key mentioned growth factor receptors affected in GC share this pathway for signal transduction[73], so it is expected to examine the effect of its inhibition in treatment of GC. Unfortunately, most of the inhibitors have shown low to moderate efficacy if any, despite being theoretically eligible targets, and further research is needed to determine the best drug to be used.

mTOR inhibitors including Rapamycin and its derivative everolimus have shown their effectiveness in various preclinical and early clinical trials, while their phase III trials showed disappointing outcomes[83].

Everolimus: First introduced to GC in 2008, upon the suggestion that cisplatin-induced hypoxia will activate hypoxia inducible factor 1 alpha, and VEGF; the addition of everolimus has proven its efficacy in vivo in inhibiting these alternative pathways[84].

Recently, the results of the Granite study evaluating everolimus for previously treated advanced GC have been reported and it was disappointingly negative; however, this same study showed that PIK3CA mutation and pS6 increased expression could be clues to everolimus effective salvage therapy but further prospective assessment for this point is needed[82,85].

Rapamycin: mTOR increased expression raises the risk of recurrence by three folds, but its definite role in activation and progression of GC is not fully comprehended[86].

Rapamycin-first known for its antifungal activity- has also anticancer activity and antiangiogenic properties. It was highly effective in preclinical trials and animal models against GC; in addition it has been shown to increase the effectiveness of chemotherapeutic drugs against GC[83,87]. However, clear level I evidence supporting its use in GC is lacking.

Mesenchymal epithelial transition factor inhibitors

Many studies have suggested that mesenchymal epithelial transition (MET) protein was over expressed in GC patients[88-90]. Aberrant gastric MET activation can lead to increased mesenchymal characteristics and less epithelial features, and promote cancer cell stemness, invasion, metastasis, and chemo-resistance with repressed E-cadherin; which allows tumor cells to disseminate and spread throughout the body. Stress, and hypoxia could aggravate GC via MET, which was significantly correlated with disease prognosis[91].

Rilotumumab: It is a monoclonal antibody against hepatocyte growth factor receptor, thus inhibiting the MET pathway responsible for cell invasion and proliferation[71], it affects mainly MET-positive GC patients with good safety profile in phase II studies[92]. Phase III studies are currently ongoing to better delineate its position in the treatment armamentarium of GC[93].

Tivantinib and foretinib: A phase II trial to test the efficacy of tivantinib (a selective c-MET inhibitor) as a monotherapy in GC in Asian population concluded that the drug has a modest effect as a second or third choice in metastatic cases[94]. Foretinib on the other hand; which is an oral multi-kinase inhibitor showed no benefit as a monotherapy in metastatic GC[71].

Onartuzumab: Onartuzumab is a monoclonal antibody inhibiting the MET pathway, used mainly in advanced solid tumors either as a single agent or in addition to bevacizumab[95], it could also be of benefit in GC; however, randomized evidence is not yet mature to support its use.

Targeting immune checkpoints

Cytotoxic T-lymphocyte antigen 4 and programmed cell death protein 1 (PD-1) are both inhibitory receptors expressed by T cells. These molecules usually appear on the surface of T cells after their activation and send an inhibitory signal[96]. In GC, PD-1 expression on CD8+ lymphocytes is significantly higher than that of normal gastric mucosa and peripheral blood[97]. PD-L1 overexpression, may also serve as a predictive biomarker in GC[98].

Immunotherapy in general and immune checkpoint inhibitors - in particular - has achieved major breakthroughs in the management of a number of difficult to treat cancers like melanoma and non small cell lung cancer[99]. For GC, a phase IB study has assessed the safety, and antitumor activity of pembrolizumab (PD-1 inhibitor) in advanced GC. Overall response rate was 32% in Asian pacific patients and 30% in rest of the world[100]. This has lead to the launch of a number of randomized studies to further evaluate the role of this new group of agents in the management of this disease[101].

Other pathways like insulin like growth factor-1

Insulin like growth factor (IGF)-1 gene expression may be associated with GC susceptibility and a research group showed that serum IGF-I and IGFBP-3 levels in GC patients were significantly decreased compared to the controls[102]. But unfortunately Figitumumab; a monoclonal antibody against IGF-1; was withdrawn from phase III clinical trial of treating lung cancer due to excessive deaths, and showed no benefit in breast cancer treatment[103]. In GC, we did not have randomized evidence supporting the use of this agent or any other agents targeting the IGF-1 pathway till now and their use should be restricted to controlled clinical studies[104].

FUTURE CANDIDATE MOLECULARMARKERS TO BE EXPLOITED AS CANDIDATE THERAPEUTIC TARGETS
Gastrokine 1

Certain studies suggested that Gastrokine 1 (GKN1) role in normal cells is to maintain integrity and mediate repair of gastric epithelium[105]. Rippa et al[103] demonstrated that GKN1 mRNA present in normal gastric cells more than adenocarcinoma cells[106]. More recent study by Xing et al[104] discovered role of GKN1 gene in GC progression, GKN1 could prevent epithelial to mesenchymal transition, decrease level of reactive oxygen species, re-expression of E-cadherin and decrease phosphatidylinositol 3-kinase (PI3K)/AKt pathway protein and so decrease metastasis in GC cell line[107].

HDAC inhibitors

Another epigenetic mechanism that plays a role in GC is histone modifications in the form of histone post translationalmodifications[108]. Histone deacetylase is thus a major target for therapeutic epigenetic inhibition.

Cancer cells are characterized by over expression of histone deacetylases (HDACs) and dysregulation of histone methyltransferases/demethylases where HDAC inhibitors have a metal binding domain that block the Zn chelation at the HDAC active sites[109]. HDAC inhibitors represent a potential approach for cancer treatment where they act mainly through cell cycle arrest at G1 or G2-M phase together with induction of apoptosis and inhibiting angiogenesis[110]. HDAC inhibitors are classified according to their structure into four groups: short chain fatty acids (e.g., phenylacetate and valproate), hydroxamic acids (e.g., panobinostat and belinostat), cyclicpeptides (e.g., romidepsin) and benzamide derivatives (e.g., MS-275)[108]. HDAC inhibitors include also sodium butyrate[111].

lncRNAs

lncRNAs play a significant role in GC progression. As lncRNAs regulate genes at different levels; namely: transcriptional, posttranscriptional and epigenetic, some of lncRNAs may have an oncogenic while other may have a tumor suppressor action[112-115]. lncRNAs with oncogenic function show high expression in GC and its expression was correlated to TNM staging and overall survival, including HOTAIR, ANRIL, H19, GHET1, CCAT1, MALTA1, HULC and MRUL which were associated with multidrug resistance and failure of chemotherapy in GC[116-124]. Tumor suppressor lncRNAs which were less expressed in GC cell lines include FENDRR, GAS5 and MEG3[125-127]. lncRNAs in GC may act as competing endogenous RNA to antagonize miRNA and relieve its repressing effect on target mRNA[128].

Like lncRNAs; miRNA regulates gene expression at different levels[129]. Several studies suggested that miRNA has a versatile role in GC[115]. In an interesting study by Kim et al[33], they demonstrated a specific miRNA signature which was also related to GC chemo resistance. Also miR-610 might have a role in preventing cancer metastasis through inhibiting actin binding protein[130].

The exact mechanism and role of lncRNAs and miRNA in GC is still unclear thus further studies are required to confirm their role in GC and consequently devising suitable therapeutic agents targeting them.

CONCLUSION

GC is a global health problem that necessitates exploiting all the available scientific advances to improve the outcome of GC patients. The use of molecular markers to predict response to GC systemic therapy has been experimented extensively. However, most of the available data were derived from small-scale retrospective analyses which do not translate to the routine use of any of these markers in day-to-day clinical practice till the time-being. On the other hand, the exploitation from our better understanding of the biology of GC has paved the way to evaluating novel agents targeting potentially carcinogenic pathways in this disease. Thus, among the plethora of agents targeting VEGF, EGFR, HER-2, IGF and mTOR pathways, only trastuzumab and ramucirumab have been approved for clinical use in advanced GC. So, while Apatinib showed impressive activity in a phase III Chinese study and may progress further to approval and pembrolizumab showed very encouraging results in early phase clinical studies, with phase III studies are ongoing, but we still have to wait to the final results because many drugs has lost the battle of approval before in GC.

We believe that properly conducted prospective randomized studies are the key to improve the outcomes of GC cases; and thus, this has to be endorsed by all scientific entities involved in GC research.

Footnotes

Manuscript source: Invited manuscript

P- Reviewer: Ribas G, Terashima M S- Editor: Gong ZM L- Editor: A E- Editor: Ma S

References
1.  Shi C, Liu B, Yan J, Liu H, Pan Z, Yao W, Yan F, Zhang H. Gastric Cancer: Preoperative TNM Staging With Individually Adjusted Computed Tomography Scanning Phase. J Comput Assist Tomogr. 2016;40:160-166.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 8]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
2.  Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, Forman D, Bray F. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 2015;136:E359-E386.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20108]  [Cited by in F6Publishing: 19874]  [Article Influence: 2208.2]  [Reference Citation Analysis (18)]
3.  Cancer NgfG. NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®) Gastric Cancer, Version 3. NCCNorg. 2015; Available from: http://nccn.org/.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Parkin DM, Stjernswärd J, Muir CS. Estimates of the worldwide frequency of twelve major cancers. Bull World Health Organ. 1984;62:163-182.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Parkin DM. The global health burden of infection-associated cancers in the year 2002. Int J Cancer. 2006;118:3030-3044.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1946]  [Cited by in F6Publishing: 1904]  [Article Influence: 105.8]  [Reference Citation Analysis (0)]
6.  Yalçin S. The increasing role of pharmacogenetics in the treatment of gastrointestinal cancers. Gastrointest Cancer Res. 2009;3:197-203.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Nashimoto A, Akazawa K, Isobe Y, Miyashiro I, Katai H, Kodera Y, Tsujitani S, Seto Y, Furukawa H, Oda I. Gastric cancer treated in 2002 in Japan: 2009 annual report of the JGCA nationwide registry. Gastric Cancer. 2013;16:1-27.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 312]  [Cited by in F6Publishing: 347]  [Article Influence: 31.5]  [Reference Citation Analysis (0)]
8.  Ko KJ, Shim JH, Yoo HM, O SI, Jeon HM, Park CH, Jeon DJ, Song KY. The clinical value of non-curative resection followed by chemotherapy for incurable gastric cancer. World J Surg. 2012;36:1800-1805.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 11]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
9.  Hallissey MT, Dunn JA, Ward LC, Allum WH. The second British Stomach Cancer Group trial of adjuvant radiotherapy or chemotherapy in resectable gastric cancer: five-year follow-up. Lancet. 1994;343:1309-1312.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  A comparison of combination chemotherapy and combined modality therapy for locally advanced gastric carcinoma. Gastrointestinal Tumor Study Group. Cancer. 1982;49:1771-1777.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Schwarz RE, Zagala-Nevarez K. Recurrence patterns after radical gastrectomy for gastric cancer: prognostic factors and implications for postoperative adjuvant therapy. Ann Surg Oncol. 2002;9:394-400.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Watt I, Stewart I, Anderson D, Bell G, Anderson JR. Laparoscopy, ultrasound and computed tomography in cancer of the oesophagus and gastric cardia: a prospective comparison for detecting intra-abdominal metastases. Br J Surg. 1989;76:1036-1039.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Young KM, Cunningham D, Chau I. Neoadjuvant chemotherapy for gastric cancer. Transl Gastrointest Cancer. 2012;1:202-204.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Orditura M, Galizia G, Sforza V, Gambardella V, Fabozzi A, Laterza MM, Andreozzi F, Ventriglia J, Savastano B, Mabilia A. Treatment of gastric cancer. World J Gastroenterol. 2014;20:1635-1649.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 404]  [Cited by in F6Publishing: 451]  [Article Influence: 45.1]  [Reference Citation Analysis (5)]
15.  Vesell ES. Pharmacogenetic perspectives gained from twin and family studies. Pharmacol Ther. 1989;41:535-552.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Yong WP, Innocenti F, Ratain MJ. The role of pharmacogenetics in cancer therapeutics. Br J Clin Pharmacol. 2006;62:35-46.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 50]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
17.  Aoyagi K, Kouhuji K, Kizaki J, Isobe T, Hashimoto K, Shirouzu K. Molecular targeting to treat gastric cancer. World J Gastroenterol. 2014;20:13741-13755.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 23]  [Cited by in F6Publishing: 23]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
18.  Pectasides D, Mylonakis A, Kostopoulou M, Papadopoulou M, Triantafillis D, Varthalitis J, Dimitriades M, Athanassiou A. CEA, CA 19-9, and CA-50 in monitoring gastric carcinoma. Am J Clin Oncol. 1997;20:348-353.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Aloe S, D’Alessandro R, Spila A, Ferroni P, Basili S, Palmirotta R, Carlini M, Graziano F, Mancini R, Mariotti S. Prognostic value of serum and tumor tissue CA 72-4 content in gastric cancer. Int J Biol Markers. 2003;18:21-27.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Cho JY. Molecular diagnosis for personalized target therapy in gastric cancer. J Gastric Cancer. 2013;13:129-135.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 48]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
21.  Nishiyama M, Eguchi H. Pharmacokinetics and pharmacogenomics in gastric cancer chemotherapy. Adv Drug Deliv Rev. 2009;61:402-407.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 24]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
22.  Lin X, Zhao Y, Song WM, Zhang B. Molecular classification and prediction in gastric cancer. Comput Struct Biotechnol J. 2015;13:448-458.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 48]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
23.  Tanaka T, Tanimoto K, Otani K, Satoh K, Ohtaki M, Yoshida K, Toge T, Yahata H, Tanaka S, Chayama K. Concise prediction models of anticancer efficacy of 8 drugs using expression data from 12 selected genes. Int J Cancer. 2004;111:617-626.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 31]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
24.  Sève P, Mackey J, Isaac S, Trédan O, Souquet PJ, Pérol M, Lai R, Voloch A, Dumontet C. Class III beta-tubulin expression in tumor cells predicts response and outcome in patients with non-small cell lung cancer receiving paclitaxel. Mol Cancer Ther. 2005;4:2001-2007.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 186]  [Cited by in F6Publishing: 198]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
25.  Suganuma K, Kubota T, Saikawa Y, Abe S, Otani Y, Furukawa T, Kumai K, Hasegawa H, Watanabe M, Kitajima M. Possible chemoresistance-related genes for gastric cancer detected by cDNA microarray. Cancer Sci. 2003;94:355-359.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Liu H, Li N, Yao L, Jiang L, Bao G, Li J, Ma Q, Liu Z. Prediction of doxorubicin sensitivity in gastric cancers based on a set of novel markers. Oncol Rep. 2008;20:963-969.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Ishikawa Y, Kubota T, Otani Y, Watanabe M, Teramoto T, Kumai K, Kitajima M, Takechi T, Okabe H, Fukushima M. Dihydropyrimidine dehydrogenase activity and messenger RNA level may be related to the antitumor effect of 5-fluorouracil on human tumor xenografts in nude mice. Clin Cancer Res. 1999;5:883-889.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Bashash M, Shah A, Hislop G, Treml M, Bretherick K, Janoo-Gilani R, Leach S, Le N, Bajdik C, Brooks-Wilson A. Genetic polymorphisms at TIMP3 are associated with survival of adenocarcinoma of the gastroesophageal junction. PLoS One. 2013;8:e59157.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 17]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
29.  Liu K, Qian T, Tang L, Wang J, Yang H, Ren J. Decreased expression of microRNA let-7i and its association with chemotherapeutic response in human gastric cancer. World J Surg Oncol. 2012;10:225.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 41]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
30.  Cristescu R, Lee J, Nebozhyn M, Kim KM, Ting JC, Wong SS, Liu J, Yue YG, Wang J, Yu K. Molecular analysis of gastric cancer identifies subtypes associated with distinct clinical outcomes. Nat Med. 2015;21:449-456.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1071]  [Cited by in F6Publishing: 1350]  [Article Influence: 150.0]  [Reference Citation Analysis (0)]
31.  Cancer Genome Atlas Research Network. Comprehensive molecular characterization of gastric adenocarcinoma. Nature. 2014;513:202-209.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4230]  [Cited by in F6Publishing: 4373]  [Article Influence: 437.3]  [Reference Citation Analysis (2)]
32.  Eto K, Iwatsuki M, Watanabe M, Ida S, Ishimoto T, Iwagami S, Baba Y, Sakamoto Y, Miyamoto Y, Yoshida N. The microRNA-21/PTEN pathway regulates the sensitivity of HER2-positive gastric cancer cells to trastuzumab. Ann Surg Oncol. 2014;21:343-350.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 77]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
33.  Kim CH, Kim HK, Rettig RL, Kim J, Lee ET, Aprelikova O, Choi IJ, Munroe DJ, Green JE. miRNA signature associated with outcome of gastric cancer patients following chemotherapy. BMC Med Genomics. 2011;4:79.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 97]  [Cited by in F6Publishing: 113]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
34.  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: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 59]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
35.  Yang Z, Guo X, Li G, Shi Y, Li L. Long noncoding RNAs as potential biomarkers in gastric cancer: Opportunities and challenges. Cancer Lett. 2016;371:62-70.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 49]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
36.  Wang Y, Zhang D, Wu K, Zhao Q, Nie Y, Fan D. Long noncoding RNA MRUL promotes ABCB1 expression in multidrug-resistant gastric cancer cell sublines. Mol Cell Biol. 2014;34:3182-3193.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 108]  [Cited by in F6Publishing: 125]  [Article Influence: 12.5]  [Reference Citation Analysis (0)]
37.  Sugita H, Iida S, Inokuchi M, Kato K, Ishiguro M, Ishikawa T, Takagi Y, Enjoji M, Yamada H, Uetake H. Methylation of BNIP3 and DAPK indicates lower response to chemotherapy and poor prognosis in gastric cancer. Oncol Rep. 2011;25:513-518.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 52]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
38.  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: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 80]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
39.  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: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 32]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
40.  Ishikawa Y, Kubota T, Otani Y, Watanabe M, Teramoto T, Kumai K, Takechi T, Okabe H, Fukushima M, Kitajima M. Dihydropyrimidine dehydrogenase and messenger RNA levels in gastric cancer: possible predictor for sensitivity to 5-fluorouracil. Jpn J Cancer Res. 2000;91:105-112.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Huang H, Han Y, Gao J, Feng J, Zhu L, Qu L, Shen L, Shou C. High level of serum AMBP is associated with poor response to paclitaxel-capecitabine chemotherapy in advanced gastric cancer patients. Med Oncol. 2013;30:748.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 20]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
42.  Mitani Y, Oue N, Matsumura S, Yoshida K, Noguchi T, Ito M, Tanaka S, Kuniyasu H, Kamata N, Yasui W. Reg IV is a serum biomarker for gastric cancer patients and predicts response to 5-fluorouracil-based chemotherapy. Oncogene. 2007;26:4383-4393.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 89]  [Cited by in F6Publishing: 101]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
43.  Okada K, Fujiwara Y, Takahashi T, Nakamura Y, Takiguchi S, Nakajima K, Miyata H, Yamasaki M, Kurokawa Y, Mori M. Overexpression of forkhead box M1 transcription factor (FOXM1) is a potential prognostic marker and enhances chemoresistance for docetaxel in gastric cancer. Ann Surg Oncol. 2013;20:1035-1043.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 73]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
44.  Ferrandina G, Zannoni GF, Martinelli E, Paglia A, Gallotta V, Mozzetti S, Scambia G, Ferlini C. Class III beta-tubulin overexpression is a marker of poor clinical outcome in advanced ovarian cancer patients. Clin Cancer Res. 2006;12:2774-2779.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 173]  [Cited by in F6Publishing: 176]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
45.  Gao J, Lu M, Yu JW, Li YY, Shen L. Thymidine Phosphorylase/β-tubulin III expressions predict the response in Chinese advanced gastric cancer patients receiving first-line capecitabine plus paclitaxel. BMC Cancer. 2011;11:177.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 22]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
46.  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: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 17]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
47.  Shi Y, Zhai H, Wang X, Han Z, Liu C, Lan M, Du J, Guo C, Zhang Y, Wu K. Ribosomal proteins S13 and L23 promote multidrug resistance in gastric cancer cells by suppressing drug-induced apoptosis. Exp Cell Res. 2004;296:337-346.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 81]  [Cited by in F6Publishing: 86]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
48.  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)]
49.  ElHalawani H, Abdel-Rahman O. Critical evaluation of ramucirumab in the treatment of advanced gastric and gastroesophageal cancers. Ther Clin Risk Manag. 2015;11:1123-1132.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.1]  [Reference Citation Analysis (0)]
50.  Basilio-de-Oliveira RP, Pannain VL. Prognostic angiogenic markers (endoglin, VEGF, CD31) and tumor cell proliferation (Ki67) for gastrointestinal stromal tumors. World J Gastroenterol. 2015;21:6924-6930.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 54]  [Cited by in F6Publishing: 55]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
51.  Zhao DQ, Chen J, Wu YF, Tian D, Zhou RX. Correlation between Vascular Endothelial Growth Factor and Somatostatin Receptor with Progression and Prognosis in Gastric Cancer. Hepatogastroenterology. 2014;61:1154-1158.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Uhlik MT, Liu J, Falcon BL, Iyer S, Stewart J, Celikkaya H, O’Mahony M, Sevinsky C, Lowes C, Douglass L. Stromal-Based Signatures for the Classification of Gastric Cancer. Cancer Res. 2016;76:2573-2586.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 28]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
53.  Company ELa. CYRAMZA (ramucirumab) injection, for intravenous infusion. : Highlights of Prescribing Information 2014; .  [PubMed]  [DOI]  [Cited in This Article: ]
54.  Park do J, Thomas NJ, Yoon C, Yoon SS. Vascular endothelial growth factor a inhibition in gastric cancer. Gastric Cancer. 2015;18:33-42.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 43]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
55.  Wilke H, Muro K, Van Cutsem E, Oh SC, Bodoky G, Shimada Y, Hironaka S, Sugimoto N, Lipatov O, Kim TY. Ramucirumab plus paclitaxel versus placebo plus paclitaxel in patients with previously treated advanced gastric or gastro-oesophageal junction adenocarcinoma (RAINBOW): a double-blind, randomised phase 3 trial. Lancet Oncol. 2014;15:1224-1235.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1613]  [Cited by in F6Publishing: 1604]  [Article Influence: 160.4]  [Reference Citation Analysis (0)]
56.  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: 1471]  [Article Influence: 147.1]  [Reference Citation Analysis (0)]
57.  Kim JG. Molecular targeted therapy for advanced gastric cancer. Korean J Intern Med. 2013;28:149-155.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 25]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
58.  Ohtsu A, Shah MA, Van Cutsem E, Rha SY, Sawaki A, Park SR, Lim HY, Yamada Y, Wu J, Langer B. Bevacizumab in combination with chemotherapy as first-line therapy in advanced gastric cancer: a randomized, double-blind, placebo-controlled phase III study. J Clin Oncol. 2011;29:3968-3976.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 795]  [Cited by in F6Publishing: 887]  [Article Influence: 68.2]  [Reference Citation Analysis (0)]
59.  Shah MA, Jhawer M, Ilson DH, Lefkowitz RA, Robinson E, Capanu M, Kelsen DP. Phase II study of modified docetaxel, cisplatin, and fluorouracil with bevacizumab in patients with metastatic gastroesophageal adenocarcinoma. J Clin Oncol. 2011;29:868-874.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 131]  [Cited by in F6Publishing: 157]  [Article Influence: 11.2]  [Reference Citation Analysis (0)]
60.  Shah MA, Ramanathan RK, Ilson DH, Levnor A, D’Adamo D, O’Reilly E, Tse A, Trocola R, Schwartz L, Capanu M. Multicenter phase II study of irinotecan, cisplatin, and bevacizumab in patients with metastatic gastric or gastroesophageal junction adenocarcinoma. J Clin Oncol. 2006;24:5201-5206.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 303]  [Cited by in F6Publishing: 346]  [Article Influence: 19.2]  [Reference Citation Analysis (0)]
61.  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)]
62.  El-Rayes BF, Zalupski M, Bekai-Saab T, Heilbrun LK, Hammad N, Patel B, Urba S, Shields AF, Vaishampayan U, Dawson S. A phase II study of bevacizumab, oxaliplatin, and docetaxel in locally advanced and metastatic gastric and gastroesophageal junction cancers. Ann Oncol. 2010;21:1999-2004.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 73]  [Cited by in F6Publishing: 85]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
63.  Lin WC, Li AF, Chi CW, Chung WW, Huang CL, Lui WY, Kung HJ, Wu CW. tie-1 protein tyrosine kinase: a novel independent prognostic marker for gastric cancer. Clin Cancer Res. 1999;5:1745-1751.  [PubMed]  [DOI]  [Cited in This Article: ]
64.  Wu CW, Li AF, Chi CW, Huang CL, Shen KH, Liu WY, Lin W. Human gastric cancer kinase profile and prognostic significance of MKK4 kinase. Am J Pathol. 2000;156:2007-2015.  [PubMed]  [DOI]  [Cited in This Article: ]
65.  Wu CW, Li AF, Chi CW, Lai CH, Huang CL, Lo SS, Lui WY, Lin WC. Clinical significance of AXL kinase family in gastric cancer. Anticancer Res. 2002;22:1071-1078.  [PubMed]  [DOI]  [Cited in This Article: ]
66.  Morishita A, Gong J, Masaki T. Targeting receptor tyrosine kinases in gastric cancer. World J Gastroenterol. 2014;20:4536-4545.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 43]  [Cited by in F6Publishing: 35]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
67.  Wilhelm SM, Adnane L, Newell P, Villanueva A, Llovet JM, Lynch M. Preclinical overview of sorafenib, a multikinase inhibitor that targets both Raf and VEGF and PDGF receptor tyrosine kinase signaling. Mol Cancer Ther. 2008;7:3129-3140.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1000]  [Cited by in F6Publishing: 1058]  [Article Influence: 66.1]  [Reference Citation Analysis (0)]
68.  Schulte N, Ebert MP, Härtel N. Gastric Cancer: New Drugs - New Strategies. Gastrointest Tumors. 2014;1:180-194.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 13]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
69.  Li J, Qin S, Xu J, Xiong J, Wu C, Bai Y, Liu W, Tong J, Liu Y, Xu R. Randomized, Double-Blind, Placebo-Controlled Phase III Trial of Apatinib in Patients With Chemotherapy-Refractory Advanced or Metastatic Adenocarcinoma of the Stomach or Gastroesophageal Junction. J Clin Oncol. 2016;34:1448-1454.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 551]  [Cited by in F6Publishing: 663]  [Article Influence: 82.9]  [Reference Citation Analysis (0)]
70.  Huynh H, Ong R, Zopf D. Antitumor activity of the multikinase inhibitor regorafenib in patient-derived xenograft models of gastric cancer. J Exp Clin Cancer Res. 2015;34:132.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 36]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
71.  Zhang K, Cui J, Xi H, Bian S, Ma L, Shen W, Li J, Wang N, Wei B, Chen L. Serum HER2 Is a Potential Surrogate for Tissue HER2 Status in Gastric Cancer: A Systematic Review and Meta-Analysis. PLoS One. 2015;10:e0136322.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 13]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
72.  Zhou Z, Xie J, Cai Y, Yang S, Chen Y, Wu H. The significance of NTR1 expression and its correlation with β-catenin and EGFR in gastric cancer. Diagn Pathol. 2015;10:128.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 8]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
73.  Baykara M, Benekli M, Ekinci O, Irkkan SC, Karaca H, Demirci U, Akinci MB, Unal OU, Dane F, Turkoz FP. Clinical Significance of HER2 Overexpression in Gastric and Gastroesophageal Junction Cancers. J Gastrointest Surg. 2015;19:1565-1571.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 10]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
74.  Kitagawa Y, Ueda M, Ando N, Ozawa S, Shimizu N, Kitajima M. Further evidence for prognostic significance of epidermal growth factor receptor gene amplification in patients with esophageal squamous cell carcinoma. Clin Cancer Res. 1996;2:909-914.  [PubMed]  [DOI]  [Cited in This Article: ]
75.  Pinto C, Di Fabio F, Barone C, Siena S, Falcone A, Cascinu S, Rojas Llimpe FL, Stella G, Schinzari G, Artale S. Phase II study of cetuximab in combination with cisplatin and docetaxel in patients with untreated advanced gastric or gastro-oesophageal junction adenocarcinoma (DOCETUX study). Br J Cancer. 2009;101:1261-1268.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 108]  [Cited by in F6Publishing: 125]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
76.  Gala K, Chandarlapaty S. Molecular pathways: HER3 targeted therapy. Clin Cancer Res. 2014;20:1410-1416.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 127]  [Cited by in F6Publishing: 130]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
77.  Jaehne J, Urmacher C, Thaler HT, Friedlander-Klar H, Cordon-Cardo C, Meyer HJ. Expression of Her2/neu oncogene product p185 in correlation to clinicopathological and prognostic factors of gastric carcinoma. J Cancer Res Clin Oncol. 1992;118:474-479.  [PubMed]  [DOI]  [Cited in This Article: ]
78.  Bang YJ, Van Cutsem E, Feyereislova A, Chung HC, Shen L, Sawaki A, Lordick F, Ohtsu A, Omuro Y, Satoh T. Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): a phase 3, open-label, randomised controlled trial. Lancet. 2010;376:687-697.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4615]  [Cited by in F6Publishing: 4894]  [Article Influence: 349.6]  [Reference Citation Analysis (1)]
79.  Park JH, Ryu MH, Park YS, Park SR, Na YS, Rhoo BY, Kang YK. Successful control of heavily pretreated metastatic gastric cancer with the mTOR inhibitor everolimus (RAD001) in a patient with PIK3CA mutation and pS6 overexpression. BMC Cancer. 2015;15:119.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 22]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
80.  Cao Y, Qu J, Li C, Yang D, Hou K, Zheng H, Liu Y, Qu X. Celecoxib sensitizes gastric cancer to rapamycin via inhibition of the Cbl-b-regulated PI3K/Akt pathway. Tumour Biol. 2015;36:5607-5615.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 10]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
81.  Cejka D, Preusser M, Woehrer A, Sieghart W, Strommer S, Werzowa J, Fuereder T, Wacheck V. Everolimus (RAD001) and anti-angiogenic cyclophosphamide show long-term control of gastric cancer growth in vivo. Cancer Biol Ther. 2008;7:1377-1385.  [PubMed]  [DOI]  [Cited in This Article: ]
82.  Ohtsu A, Ajani JA, Bai YX, Bang YJ, Chung HC, Pan HM, Sahmoud T, Shen L, Yeh KH, Chin K. Everolimus for previously treated advanced gastric cancer: results of the randomized, double-blind, phase III GRANITE-1 study. J Clin Oncol. 2013;31:3935-3943.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 332]  [Cited by in F6Publishing: 357]  [Article Influence: 32.5]  [Reference Citation Analysis (0)]
83.  Yu G, Wang J, Chen Y, Wang X, Pan J, Li G, Jia Z, Li Q, Yao JC, Xie K. Overexpression of phosphorylated mammalian target of rapamycin predicts lymph node metastasis and prognosis of chinese patients with gastric cancer. Clin Cancer Res. 2009;15:1821-1829.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 106]  [Cited by in F6Publishing: 119]  [Article Influence: 7.9]  [Reference Citation Analysis (0)]
84.  Shigematsu H, Yoshida K, Sanada Y, Osada S, Takahashi T, Wada Y, Konishi K, Okada M, Fukushima M. Rapamycin enhances chemotherapy-induced cytotoxicity by inhibiting the expressions of TS and ERK in gastric cancer cells. Int J Cancer. 2010;126:2716-2725.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 9]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
85.  Lee HE, Kim MA, Lee HS, Jung EJ, Yang HK, Lee BL, Bang YJ, Kim WH. MET in gastric carcinomas: comparison between protein expression and gene copy number and impact on clinical outcome. Br J Cancer. 2012;107:325-333.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 127]  [Cited by in F6Publishing: 144]  [Article Influence: 12.0]  [Reference Citation Analysis (0)]
86.  Ma J, Ma J, Meng Q, Zhao ZS, Xu WJ. Prognostic value and clinical pathology of MACC-1 and c-MET expression in gastric carcinoma. Pathol Oncol Res. 2013;19:821-832.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 30]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
87.  Ha SY, Lee J, Kang SY, Do IG, Ahn S, Park JO, Kang WK, Choi MG, Sohn TS, Bae JM. MET overexpression assessed by new interpretation method predicts gene amplification and poor survival in advanced gastric carcinomas. Mod Pathol. 2013;26:1632-1641.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 92]  [Cited by in F6Publishing: 101]  [Article Influence: 9.2]  [Reference Citation Analysis (0)]
88.  Huang L, Wu RL, Xu AM. Epithelial-mesenchymal transition in gastric cancer. Am J Transl Res. 2015;7:2141-2158.  [PubMed]  [DOI]  [Cited in This Article: ]
89.  Zhu M, Tang R, Doshi S, Oliner KS, Dubey S, Jiang Y, Donehower RC, Iveson T, Loh EY, Zhang Y. Exposure-response analysis of rilotumumab in gastric cancer: the role of tumour MET expression. Br J Cancer. 2015;112:429-437.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 37]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
90.  Abdel-Rahman O. Targeting the hepatocyte growth factor/mesenchymal epithelial transition pathway in gastric cancer: biological rationale and clinical applications. Expert Rev Anticancer Ther. 2015;15:235-245.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 6]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
91.  Kang YK, Muro K, Ryu MH, Yasui H, Nishina T, Ryoo BY, Kamiya Y, Akinaga S, Boku N. A phase II trial of a selective c-Met inhibitor tivantinib (ARQ 197) monotherapy as a second- or third-line therapy in the patients with metastatic gastric cancer. Invest New Drugs. 2014;32:355-361.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 83]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
92.  Borin MT, Chen M, Mocci S, Rubets I, Chittenden J, Aldairy W, Stroh M. Onartuzumab with or without bevacizumab in combination with weekly paclitaxel does not prolong QTc or adversely affect other ECG parameters in patients with locally recurrent or metastatic triple-negative breast cancer. Cancer Chemother Pharmacol. 2015;75:401-410.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 4]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
93.  Peggs KS, Quezada SA, Chambers CA, Korman AJ, Allison JP. Blockade of CTLA-4 on both effector and regulatory T cell compartments contributes to the antitumor activity of anti-CTLA-4 antibodies. J Exp Med. 2009;206:1717-1725.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 644]  [Cited by in F6Publishing: 682]  [Article Influence: 45.5]  [Reference Citation Analysis (0)]
94.  Saito H, Kuroda H, Matsunaga T, Osaki T, Ikeguchi M. Increased PD-1 expression on CD4+ and CD8+ T cells is involved in immune evasion in gastric cancer. J Surg Oncol. 2013;107:517-522.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 67]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
95.  Bang YJ, Chung HC, Shankaran V, Geva R, Catenacci DV, Gupta S, Eder JP, Berger R, Gonzalez EJ, Ray A. Relationship between PD-L1 expression and clinical outcomes in patients (Pts) with advanced gastric cancer treated with the anti-PD-1 monoclonal antibody pembrolizumab (Pembro; MK-3475) in KEYNOTE-012. J Clin Oncol. 2015;43:4001.  [PubMed]  [DOI]  [Cited in This Article: ]
96.  Abdel-Rahman O. Correlation between PD-L1 expression and outcome of NSCLC patients treated with anti-PD-1/PD-L1 agents: A meta-analysis. Crit Rev Oncol Hematol. 2016;101:75-85.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 90]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
97.  Muro K, Bang Y, Shankaran V, Geva R, Catenacci DV, Gupta S, Chung HC. LBA15A phase 1B study of pembrolizumab (PEMBRO; MK-3475) inpatients (PTS) with advanced gastric cancer. Ann Oncol. 2014;25:mdu438-15.  [PubMed]  [DOI]  [Cited in This Article: ]
98.  Abdel-Rahman O. Immune checkpoints aberrations and gastric cancer; assessment of prognostic value and evaluation of therapeutic potentials. Crit Rev Oncol Hematol. 2016;97:65-71.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 26]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
99.  Altinkaynak K, Bilici M, Bakan N, Akçay F. Circulating levels of IGF-I and IGFBP-3 in gastric cancer. Turk J Med Sci. 2012;42:1458-1462.  [PubMed]  [DOI]  [Cited in This Article: ]
100.  Yang Y, Yee D. Targeting insulin and insulin-like growth factor signaling in breast cancer. J Mammary Gland Biol Neoplasia. 2012;17:251-261.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 57]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
101.  Abdel-Rahman O. Insulin-like growth factor pathway aberrations and gastric cancer; evaluation of prognostic significance and assessment of therapeutic potentials. Med Oncol. 2015;32:431.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 10]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
102.  Oien KA, McGregor F, Butler S, Ferrier RK, Downie I, Bryce S, Burns S, Keith WN. Gastrokine 1 is abundantly and specifically expressed in superficial gastric epithelium, down-regulated in gastric carcinoma, and shows high evolutionary conservation. J Pathol. 2004;203:789-797.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 82]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
103.  Rippa E, Martin G, Rocco A, La Monica G, Fiengo A, Siciliano RA, Cacace G, Malori A, Nardone G, Arcari P. Changes of protein expression in Helicobacter pylori-infected human gastric mucosa. Curr Top Peptide Protein Res. 2007;8:35-43.  [PubMed]  [DOI]  [Cited in This Article: ]
104.  Xing R, Li W, Cui J, Zhang J, Kang B, Wang Y, Wang Z, Liu S, Lu Y. Gastrokine 1 induces senescence through p16/Rb pathway activation in gastric cancer cells. Gut. 2012;61:43-52.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 49]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
105.  Ramachandran S, Ient J, Göttgens EL, Krieg AJ, Hammond EM. Epigenetic Therapy for Solid Tumors: Highlighting the Impact of Tumor Hypoxia. Genes (Basel). 2015;6:935-956.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 40]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
106.  Keppler BR, Archer TK. Chromatin-modifying enzymes as therapeutic targets--Part 1. Expert Opin Ther Targets. 2008;12:1301-1312.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 62]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
107.  Marchion D, Münster P. Development of histone deacetylase inhibitors for cancer treatment. Expert Rev Anticancer Ther. 2007;7:583-598.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 122]  [Cited by in F6Publishing: 126]  [Article Influence: 12.6]  [Reference Citation Analysis (0)]
108.  Drottar M, Liberman MC, Ratan RR, Roberson DW. The histone deacetylase inhibitor sodium butyrate protects against cisplatin-induced hearing loss in guinea pigs. Laryngoscope. 2006;116:292-296.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 37]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
109.  Song H, Sun W, Ye G, Ding X, Liu Z, Zhang S, Xia T, Xiao B, Xi Y, Guo J. Long non-coding RNA expression profile in human gastric cancer and its clinical significances. J Transl Med. 2013;11:225.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 166]  [Cited by in F6Publishing: 185]  [Article Influence: 16.8]  [Reference Citation Analysis (0)]
110.  Cao WJ, Wu HL, He BS, Zhang YS, Zhang ZY. Analysis of long non-coding RNA expression profiles in gastric cancer. World J Gastroenterol. 2013;19:3658-3664.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 144]  [Cited by in F6Publishing: 163]  [Article Influence: 14.8]  [Reference Citation Analysis (0)]
111.  Lin XC, Zhu Y, Chen WB, Lin LW, Chen DH, Huang JR, Pan K, Lin Y, Wu BT, Dai Y. Integrated analysis of long non-coding RNAs and mRNA expression profiles reveals the potential role of lncRNAs in gastric cancer pathogenesis. Int J Oncol. 2014;45:619-628.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 46]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
112.  Mattie MD, Benz CC, Bowers J, Sensinger K, Wong L, Scott GK, Fedele V, Ginzinger D, Getts R, Haqq C. Optimized high-throughput microRNA expression profiling provides novel biomarker assessment of clinical prostate and breast cancer biopsies. Mol Cancer. 2006;5:24.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 535]  [Cited by in F6Publishing: 538]  [Article Influence: 29.9]  [Reference Citation Analysis (0)]
113.  Liu XH, Sun M, Nie FQ, Ge YB, Zhang EB, Yin DD, Kong R, Xia R, Lu KH, Li JH. Lnc RNA HOTAIR functions as a competing endogenous RNA to regulate HER2 expression by sponging miR-331-3p in gastric cancer. Mol Cancer. 2014;13:92.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 659]  [Cited by in F6Publishing: 763]  [Article Influence: 76.3]  [Reference Citation Analysis (0)]
114.  Okugawa Y, Toiyama Y, Hur K, Toden S, Saigusa S, Tanaka K, Inoue Y, Mohri Y, Kusunoki M, Boland CR. Metastasis-associated long non-coding RNA drives gastric cancer development and promotes peritoneal metastasis. Carcinogenesis. 2014;35:2731-2739.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 168]  [Cited by in F6Publishing: 214]  [Article Influence: 21.4]  [Reference Citation Analysis (0)]
115.  Endo H, Shiroki T, Nakagawa T, Yokoyama M, Tamai K, Yamanami H, Fujiya T, Sato I, Yamaguchi K, Tanaka N. Enhanced expression of long non-coding RNA HOTAIR is associated with the development of gastric cancer. PLoS One. 2013;8:e77070.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 177]  [Cited by in F6Publishing: 202]  [Article Influence: 18.4]  [Reference Citation Analysis (0)]
116.  Zhang EB, Kong R, Yin DD, You LH, Sun M, Han L, Xu TP, Xia R, Yang JS, De W. Long noncoding RNA ANRIL indicates a poor prognosis of gastric cancer and promotes tumor growth by epigenetically silencing of miR-99a/miR-449a. Oncotarget. 2014;5:2276-2292.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 259]  [Cited by in F6Publishing: 299]  [Article Influence: 33.2]  [Reference Citation Analysis (0)]
117.  Zhuang M, Gao W, Xu J, Wang P, Shu Y. The long non-coding RNA H19-derived miR-675 modulates human gastric cancer cell proliferation by targeting tumor suppressor RUNX1. Biochem Biophys Res Commun. 2014;448:315-322.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 142]  [Cited by in F6Publishing: 162]  [Article Influence: 16.2]  [Reference Citation Analysis (0)]
118.  Yang F, Xue X, Zheng L, Bi J, Zhou Y, Zhi K, Gu Y, Fang G. Long non-coding RNA GHET1 promotes gastric carcinoma cell proliferation by increasing c-Myc mRNA stability. FEBS J. 2014;281:802-813.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 164]  [Cited by in F6Publishing: 178]  [Article Influence: 17.8]  [Reference Citation Analysis (0)]
119.  Yang F, Xue X, Bi J, Zheng L, Zhi K, Gu Y, Fang G. Long noncoding RNA CCAT1, which could be activated by c-Myc, promotes the progression of gastric carcinoma. J Cancer Res Clin Oncol. 2013;139:437-445.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 199]  [Cited by in F6Publishing: 224]  [Article Influence: 18.7]  [Reference Citation Analysis (0)]
120.  Wang J, Su L, Chen X, Li P, Cai Q, Yu B, Liu B, Wu W, Zhu Z. MALAT1 promotes cell proliferation in gastric cancer by recruiting SF2/ASF. Biomed Pharmacother. 2014;68:557-564.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 116]  [Cited by in F6Publishing: 128]  [Article Influence: 12.8]  [Reference Citation Analysis (0)]
121.  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: ]  [Cited by in Crossref: 587]  [Cited by in F6Publishing: 613]  [Article Influence: 36.1]  [Reference Citation Analysis (0)]
122.  Xu TP, Huang MD, Xia R, Liu XX, Sun M, Yin L, Chen WM, Han L, Zhang EB, Kong R. Decreased expression of the long non-coding RNA FENDRR is associated with poor prognosis in gastric cancer and FENDRR regulates gastric cancer cell metastasis by affecting fibronectin1 expression. J Hematol Oncol. 2014;7:63.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 181]  [Cited by in F6Publishing: 216]  [Article Influence: 21.6]  [Reference Citation Analysis (0)]
123.  Sun M, Jin FY, Xia R, Kong R, Li JH, Xu TP, Liu YW, Zhang EB, Liu XH, De W. Decreased expression of long noncoding RNA GAS5 indicates a poor prognosis and promotes cell proliferation in gastric cancer. BMC Cancer. 2014;14:319.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 216]  [Cited by in F6Publishing: 256]  [Article Influence: 25.6]  [Reference Citation Analysis (0)]
124.  Sun M, Xia R, Jin F, Xu T, Liu Z, De W, Liu X. Downregulated long noncoding RNA MEG3 is associated with poor prognosis and promotes cell proliferation in gastric cancer. Tumour Biol. 2014;35:1065-1073.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 208]  [Cited by in F6Publishing: 200]  [Article Influence: 18.2]  [Reference Citation Analysis (0)]
125.  Xia T, Liao Q, Jiang X, Shao Y, Xiao B, Xi Y, Guo J. Long noncoding RNA associated-competing endogenous RNAs in gastric cancer. Sci Rep. 2014;4:6088.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 246]  [Cited by in F6Publishing: 322]  [Article Influence: 32.2]  [Reference Citation Analysis (0)]
126.  Garzon R, Calin GA, Croce CM. MicroRNAs in Cancer. Annu Rev Med. 2009;60:167-179.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1354]  [Cited by in F6Publishing: 1476]  [Article Influence: 98.4]  [Reference Citation Analysis (0)]
127.  Wang J, Zhang J, Wu J, Luo D, Su K, Shi W, Liu J, Tian Y, Wei L. MicroRNA-610 inhibits the migration and invasion of gastric cancer cells by suppressing the expression of vasodilator-stimulated phosphoprotein. Eur J Cancer. 2012;48:1904-1913.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 40]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
128.  Park JS, Young Yoon S, Kim JM, Yeom YI, Kim YS, Kim NS. Identification of novel genes associated with the response to 5-FU treatment in gastric cancer cell lines using a cDNA microarray. Cancer Lett. 2004;214:19-33.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 35]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
129.  Zhang YH, Wu Q, Xiao XY, Li DW, Wang XP. Silencing MRP4 by small interfering RNA reverses acquired DDP resistance of gastric cancer cell. Cancer Lett. 2010;291:76-82.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 31]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
130.  Kim JG, Sohn SK, Chae YS, Song HS, Kwon KY, Do YR, Kim MK, Lee KH, Hyun MS, Lee WS. TP53 codon 72 polymorphism associated with prognosis in patients with advanced gastric cancer treated with paclitaxel and cisplatin. Cancer Chemother Pharmacol. 2009;64:355-360.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 32]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
131.  Meulendijks D, Beerepoot LV, Boot H, de Groot JW, Los M, Boers JE, Vanhoutvin SA, Polee MB, Beeker A, Portielje JE. Trastuzumab and bevacizumab combined with docetaxel, oxaliplatin and capecitabine as first-line treatment of advanced HER2-positive gastric cancer: a multicenter phase II study. Invest New Drugs. 2016;34:119-128.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 29]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
132.  Aoyagi K, Kouhuji K, Miyagi M, Kizaki J, Isobe T, Hashimoto K, Shirouzu K. Molecular targeting therapy using bevacizumab for peritoneal metastasis from gastric cancer. World J Crit Care Med. 2013;2:48-55.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 12]  [Cited by in F6Publishing: 11]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
133.  Sun T, Jia Y, Xiao D. Interference of STAT 5b expression enhances the chemo-sensitivity of gastric cancer cells to gefitinib by promoting mitochondrial pathway-mediated cell apoptosis. Oncol Rep. 2015;34:227-234.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 5]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
134.  Rao S, Starling N, Cunningham D, Sumpter K, Gilligan D, Ruhstaller T, Valladares-Ayerbes M, Wilke H, Archer C, Kurek R. Matuzumab plus epirubicin, cisplatin and capecitabine (ECX) compared with epirubicin, cisplatin and capecitabine alone as first-line treatment in patients with advanced oesophago-gastric cancer: a randomised, multicentre open-label phase II study. Ann Oncol. 2010;21:2213-2219.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 102]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
135.  Shah MA, Bang YJ, Lordick F, Tabernero J, Chen M, Hack SP, Phan SC, Shames DS, Cunningham D. METGastric: A phase III study of onartuzumab plus mFOLFOX6 in patients with metastatic HER2-negative (HER2-) and MET-positive (MET) adenocarcinoma of the stomach or gastroesophageal junction (GEC). J Clin Oncol. 2015;33:4012.  [PubMed]  [DOI]  [Cited in This Article: ]