Review Open Access
Copyright ©The Author(s) 2016. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Aug 14, 2016; 22(30): 6776-6799
Published online Aug 14, 2016. doi: 10.3748/wjg.v22.i30.6776
New advances in targeted gastric cancer treatment
Daniela Cornelia Lazăr, Adrian Goldiş, Department of Gastroenterology and Hepatology, University of Medicine and Pharmacy, “Victor Babeş”, 300041 Timişoara, Timis County, Romania
Sorina Tăban, Marioara Cornianu, Alexandra Faur, Department of Pathology, University of Medicine and Pharmacy, “Victor Babeş”, 300041 Timişoara, Timis County, Romania
Author contributions: All of the authors contributed equally to this work.
Conflict-of-interest statement: The authors have no conflicts of interest to report.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Daniela Cornelia Lazăr, MD, PhD, Assistant Professor, Department of Gastroenterology and Hepatology, University of Medicine and Pharmacy, “Victor Babeş”, Piata Eftimie Murgu No. 2, 300041 Timişoara, Timis County, Romania. lazar_daniela@yahoo.com
Telephone: +40-356-433121 Fax: +40-256-486956
Received: March 29, 2016
Peer-review started: April 4, 2016
First decision: May 12, 2016
Revised: June 13, 2016
Accepted: July 6, 2016
Article in press: July 6, 2016
Published online: August 14, 2016

Abstract

Despite a decrease in incidence over past decades, gastric cancer remains a major global health problem. In the more recent period, survival has shown only minor improvement, despite significant advances in diagnostic techniques, surgical and chemotherapeutic approaches, the development of novel therapeutic agents and treatment by multidisciplinary teams. Because multiple genetic mutations, epigenetic alterations, and aberrant molecular signalling pathways are involved in the development of gastric cancers, recent research has attempted to determine the molecular heterogeneity responsible for the processes of carcinogenesis, spread and metastasis. Currently, some novel agents targeting a part of these dysfunctional molecular signalling pathways have already been integrated into the standard treatment of gastric cancer, whereas others remain in phases of investigation within clinical trials. It is essential to identify the unique molecular patterns of tumours and specific biomarkers to develop treatments targeted to the individual tumour behaviour. This review analyses the global impact of gastric cancer, as well as the role of Helicobacter pylori infection and the efficacy of bacterial eradication in preventing gastric cancer development. Furthermore, the paper discusses the currently available targeted treatments and future directions of research using promising novel classes of molecular agents for advanced tumours.

Key Words: Gastric cancer, Helicobacter pylori infection, Chemotherapy, Targeted therapy, Clinical trials, New treatment advances

Core tip: Recent research attempts to determine the molecular heterogeneity of gastric tumours. Currently, some novel agents targeting aberrant molecular signalling pathways are already part of the standard treatments for gastric cancer, whereas others remain in phases of clinical trials. By identifying the unique molecular patterns of tumours, new horizons in gastric cancer treatment towards personalized medicine will emerge. This review analyses the role of Helicobacter pylori infection and the efficacy of bacterial eradication in gastric cancer prevention, as well as the currently available targeted treatments and future directions of research using promising novel classes of molecular agents.



INTRODUCTION

Gastric cancer represents a major health problem worldwide[1,2]. Despite a decrease in incidence in past decades, stomach cancer remains the fifth most common type of cancer and the third leading cause of cancer-related mortality worldwide[3]. There is broad variation in the geographic distribution of gastric carcinoma, with this neoplasia being the most common malignancy in some regions, such as Japan.

In developed countries, the incidence of cardia tumours has paralleled the trend in oesophageal cancer, while distal cancers have tended to decrease in incidence[4,5]. In contrast, the incidence of non-cardia neoplasias remains high in Japan and other parts of the world[6].

There are differences in environmental factors, such as dietary patterns and salt intake, the prevalence of Helicobacter pylori (H. pylori) infection and the virulence of strains, as well as host factors, which determine the regional variations in the incidence of stomach cancer.

Data from the literature, based on epidemiological data and observational and therapeutic trials, as well as in vitro and in vivo models, have revealed that H. pylori infection is the most important proven risk factor for human non-cardia gastric neoplasia. The risk of developing stomach cancer proved to be 20-fold higher or more in the presence of H. pylori infection[7-10].

H. pylori prevalence has demonstrated great variability determined by factors such as geographic location, age, ethnicity and socioeconomic conditions. For these reasons, its prevalence is usually high in developing regions, where H. pylori infection represents a public-health issue, and lower in developed countries. Its prevalence can show variability within regions of different countries and between more crowded urban populations and rural populations, mostly due to socioeconomic differences between populations.

H. pylori eradication is an effective method for preventing gastric cancer if it is performed before the development of premalignant lesions. There have been numerous studies suggesting that H. pylori eradication is the most effective approach to gastric cancer prevention, but we should bear in mind that this intervention is more effective in patients who do not have atrophic gastritis or IM at the same time[11-13]. In a pooled analysis of six studies including 6695 patients (most of them from Asia) followed up for 4-10 years, Fuccio and collaborators showed that the relative risk for gastric cancer after H. pylori eradication was 0.65 (95%CI: 0.43-0.98)[14].

De Vries et al[15] found out that the incidence of premalignant gastric lesions was declining, and they concluded that a further decrease of at least 24% in the incidence of gastric cancer in the coming decade could be anticipated in Western countries.

The study by Lee showed that mass eradication of H. pylori infection led to an important reduction in the incidence of gastric atrophy shortly after implementation, supporting the use of this strategy to prevent gastric cancer in populations in whom H. pylori is endemic and the incidence of gastric cancer is high[16].

Usually, the diagnosis of gastric cancer is delayed by a lack of early specific symptoms, and most patients are diagnosed in advanced stages, resulting in poor 5-year survival rates[17], with median survival of less than 1 year for metastatic disease[18-20].

Based on the evidence that exists regarding prognostic factors and the management of specific types of gastric cancer, there is an imperative need to improve the tumour node metastasis (TNM) staging system. The 7th edition of the TNM staging system emphasizes the importance of both depth of invasion and the number of locoregional lymph nodes involved as major prognostic factors, as a consensus approach of Eastern and Western countries[21,22]. Currently, gastro-oesophageal (GE) junction cancers are now classified as oesophageal cancers because of the similarity. However, we should bear in mind that it is sometimes difficult to interpret data from the literature and to implement them because many gastric cancer clinical trials include a significant proportion of GE junction tumours, while many trials designed for oesophageal cancer have also included some proximal gastric tumours.

Surgical resection is the mainstay of stomach cancer treatment, with adjuvant chemotherapy or chemoradiation[23,24]. In recent years, the survival rate has shown only minor improvement despite significant progress in diagnostic techniques, surgical and chemotherapy approaches, the discovery of novel therapeutic agents and treatment of gastric cancer patients by multidisciplinary teams. In patients with locally advanced or metastatic cancers, treatment relies mainly on chemotherapy, although the results are often limited by the high grade toxicity of aggressive regimens associating three agents or by the poor performance status of patients[25]. In such cases, palliatives and BSC are unfortunately the only appropriate treatments.

Because the pathogenesis of gastric cancers involves many different genetic mutations, as well as epigenetic alterations, and the dysfunction of molecular signalling pathways, many efforts have been undertaken in recent years to emphasize the molecular heterogeneity responsible not only for the process of carcinogenesis but also for cancer spread and metastasis. Each of these molecular alterations is involved in a different stage of cancerous disease. Currently, some of these aberrant molecular signalling pathways are used as targets of interventions with novel therapeutic agents, some of which are already approved for the treatment of gastric cancer, while others remain in the phase of clinical trials[26]. It is essential to identify the unique molecular patterns of tumour carcinogenesis and progression in to develop specific treatments targeted to the individual tumour biology and behaviour.

METHODS

A literature search modality was applied for all English language literature published in the last 16 years, before March 2016, by assessing the PubMed electronic database. The keywords used for our research purposes were “gastric cancer”, “stomach neoplasm”, “treatment”, “targeted treatment”, and “molecular treatment”. The specific search was also performed to identify clinical studies involving novel agents for gastric cancer treatment using the ClinicalTrials.gov database. Furthermore, the search was also performed on different cancer-related Web sites.

TARGETED THERAPIES

Numerous targeted therapies belonging to different classes of drugs have been investigated as therapeutics in gastric cancer, starting with preclinical studies and continuing into clinical trials, such as human epidermal growth factor receptor type 2 (HER2), vascular endothelial growth factor (VEGF) receptor, epidermal growth factor receptor (EGFR), the insulin-like growth factor receptor, phosphatidylinositol 3-kinase (PI3k)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) pathway, c-MET, fibroblast growth factor receptor (FGFR), poly [adenosine diphosphate (ADP)]-ribose polymerase (PARP) inhibitors, and immunotherapies[26] (Table 1).

Table 1 Targeted agents that have been studied for the treatment of gastric neoplasia.
Class (target)TypeAgentCurrent status in GC treatmentIndicationsTreatment benefits
HER2 inhibitorsmAbTrastuzumabFDA approval 2010HER2+ metastatic GC↑ OS, ORR, PFS[30]
mAb second generationPertuzumabPhase III clinical trials ongoingHER2+ metastatic/locally advanced unresectable GCUnder investigation[38]
Antibody-drug conjugateTrastuzumab etansinePhase II/III clinical trials ongoingHER2+ advanced GCUnder investigation[39]
TKILapatinibPhase III clinical trialsFirst-line and second-line HER2+ advanced/metastatic GCBenefits in some subgroups of patients; ↑ RR in second-line setting[42,43]
Phase III clinical trials ongoingResectable GCUnder investigation[44]
EGFR inhibitorsmAbCetuximabPhase II/III clinical trialsAdvanced/metastatic GCNo benefits in first-line setting; modest benefit, reduced toxicity in second-line setting[52,57-59]
Humanized mAbPanitumumabPhase III clinical trialsAdvanced GCNo benefits[60]
NimotuzumabPhase II clinical trialsAdvanced GCPromising results in EGFR2+/3+ subgroups of patients[69]
TKIGefitinibPhase I/II/III clinical trials ongoingResectable/advanced and metastatic GCNo benefits[72-74]
ErlotinibPhase IIAdvanced/metastatic GCNo benefits so far; phase II trials pending results[75]
VEGF/VEGFR inhibitorsAnti-VEGF humanized mAbBevacizumabPhase III clinical trialsAdvanced GC, first-line treatment↑ PFS, ORR, benefits in specific subgroups of patients[106-108]
Phase III clinical trialsPerioperative chemotherapy in resectable GCUnder investigation[44,110]
Anti-VEGF humanized mAbRamucirumabFDA approval 2014Advanced GC, second-line treatment↑ OS, PFS[119-122]
TKIApatinibCFDA approval 2014Metastatic GC, third-line treatment↑ OS, PFS[127,129,131]
VEGFR, PDGFR, c-KIT, FMS, FLT inhibitorTKISunitinibPhase I/II clinical trialsAdvanced GC↑ OS, ORR (second-line); promising results[133-136]
RAF, VEGFR, PDGFR inhibitorTKISorafenibPhase I/II clinical trialsAdvanced GC↑ PFS, OS (second-line)[143]; conflicting data[144,145]
VEGFR, PDGFR, c-KIT inhibitorTKIPazopanibPhase I/II clinical trialsAdvanced GCUnder investigation[150-152]
VEGFR, RAF inhibitorTKIRegorafenibPhase I/II ongoing clinical trialsRefractory advanced GC↑ PFS, OS, drawback: drug-related toxicity[156]; under investigation[157-159]
IGF-1 inhibitorHumanized mAbFigitumumabPhase I ongoing clinical trialsAdvanced GCUnder investigation[167,168]
FGF- inhibitorsTKIAZD2171Phase II clinical trialsAdvanced GC, second-line treatmentPromising results[172]
JNJ-42756493Phase I ongoing clinical trialsAdvanced GCPromising results[173]
DovitinibPhase I/II ongoing clinical trialsAdvanced GCUnder investigation[183-185]
Brivanib
INCB054828
HGF/c-MET inhibitorsHumanized mAbRilotumumabPhase II clinical trialsAdvanced/metastatic GC↑ OS in MET (+) subset of patients[188,189]
Phase III clinical trialsIncreased toxicity, ↓ OS, PFS
OnartuzumabPhase III clinical trialsMET(+), HER2(-) GCNegative results
TKIForetonibPhase II clinical trialsMetastatic GCMinimal efficacy[194]
PI3 kinase/mTOR inhibitorsInhibitorEverolimusPhase III clinical trialsAdvanced GC, second/third-line treatmentNegative results[207,208]
SirolimusPhase I ongoing clinical trialsAdvanced GCUnder investigation[209]
PARP inhibitorsInhibitorOlaparibPhase II clinical trialsMetastatic/recurrent GC, second-line treatment↑ OS in specific subgroup of patients; under investigation[211-213]
VeliparibPhase I ongoing clinical trialsAdvanced GCUnder investigation[216]
Immuno-therapy/Immuno-checkpoint blockadeCTLA-4 inhibitorIpilimumabPhase II clinical trialsAdvanced/metastatic GCNegative results[224]
CTLA-4 inhibitorTremalimu-mabPhase I/II ongoing clinical trialsAdvanced GCSynergistic effects for Tremelimumab and Durvalumab combination; under investigation[240,241]
Binding of PD-1 with PD-L1 blockadeNivolumabPhase I/II ongoing clinical trialsAdvanced/metastatic GCPromising results[228]
Pembrolizu-mabPhase I-III ongoing clinical trialsAdvanced/metastatic GC, naive patients and progression under treatmentPromising results[230]; under investigation[231-238,243,244]
PD-L1 inhibitorDurvalumabPhase I clinical trialsAdvanced GCPromising results[239]
GCC inhibitorHumanized mAbMLN0264Phase I/II ongoing clinical trialsAdvanced/metastatic GC, GCC (+)Promising results[247,248]
Tumour cell cycle inhibitorCKIFlavopiridolPhase II clinical trialsAdvanced GCNegative results[251,252]
Tumour cell apoptosisProteasome inhibitorBortezomibPhase II clinical trialsAdvanced/metastatic GCNegative results[262]
MMP inhibitorsInhibitorMarimastatRandomized, double-blind, placebo-controlled studyNon-resectable GC, maintenance treatmentSurvival benefit[265]
Anti-HER2 therapies

The HER2 receptor belongs to the EGFR/HER family and is involved in signal transduction, cell growth and differentiation. The HER2 proto-oncogene encodes for a 185 kDa transmembrane glycoprotein receptor with intracellular tyrosine kinase activity[27]. Data from the literature have revealed HER2 over-expression and amplification in 7%-34% of patients with gastro-oesophageal adenocarcinomas. HER2 immunoreaction is considered to be negative if the immunohistochemistry (IHC) is 0 or 1+ and positive if the IHC 3+. IHC of 2+ requires confirmatory testing with fluorescence in situ hybridization (FISH)[28].

Anti-HER2 therapies have demonstrated efficiency for both in vitro and in vivo gastric cancer models[29]. Therapies evaluated in clinical trials of patients with gastric cancer have included inhibition by monoclonal antibodies (trastuzumab and pertuzumab) and tyrosine kinase inhibitors (TKIs) (lapatinib).

Monoclonal antibodies targeting HER-2

Trastuzumab (trade names Herclon, Herceptin) is a humanized monoclonal antibody that targets the extracellular binding domain of the HER2 receptor; the first therapeutic indication for this agent was for breast cancer (FDA approval since 1998). The efficacy of trastuzumab for gastric cancer was assessed in an international, open-label, phase III trial (ToGA) that randomized naïve patients with metastatic or locally advanced unresectable gastric or gastro-oesophageal junction adenocarcinoma with over-expressed HER2 to chemotherapy associated with trastuzumab vs chemotherapy alone[30]. Because this study demonstrated an improvement in median overall survival (OS) (2.7 mo), as well as significantly better response rates, time to progression of disease, and duration of response for patients who received trastuzumab, the FDA approved this drug as a targeted therapy for gastric and gastro-oesophageal junction adenocarcinoma in 2010. The results of this study demonstrated that adding trastuzumab to classic chemotherapy could increase the OS of patients with advanced gastric cancer to more than 1 year, thus proving the essential role of trastuzumab in the treatment of patients with this advanced stage of disease. Currently, trastuzumab in combination with capecitabine or 5-fluorouracil and cisplatin is indicated for the treatment of patients with HER2-positive metastatic adenocarcinoma of the stomach or gastro-oesophageal junction; it is approved for use only in patients with HER2 overexpression, which is defined as an IHC 3 positive result or an IHC 2 and FISH double-positive result.

There are several other ongoing studies of trastuzumab, such as the HELOISE trial (trastuzumab plus chemotherapy in patients with HER2-positive metastatic gastric or gastro-oesophageal junction cancer)[31], the phase II study NCT01130337 in non-metastatic gastric cancer using trastuzumab and chemotherapy in perioperative setting[32], the TOXAG study (a combination of chemotherapeutic agents plus trastuzumab and chemoradiotherapy in an adjuvant setting in operated patients with HER2+ gastric or gastro-oesophageal junction cancer)[33], the HER-FLOT study (chemotherapy plus trastuzumab as a perioperative treatment for patients with HER2-positive locally advanced oesophago-gastric adenocarcinoma)[34] and the phase III trial RTOG 1010 for locally advanced oesophageal or gastro-oesophageal junction adenocarcinoma (chemoradiation ± trastuzumab prior to surgery)[35]. Patients with cancer have shown good drug tolerance.

The results of two phase II clinical trials assessing the efficacy and safety of trastuzumab plus XELOX (capecitabine/oxaliplatin)[36] and SP (S-1/cisplatin)[37], respectively, for advanced gastric cancer therapy confirmed that trastuzumab associated with these chemotherapy regimens was efficient in treating gastric cancer.

The second generation of anti-HER2 agents was developed because of emerging resistance to anti-HER2 therapy. Pertuzumab (trade name Perjeta), which binds to a distinct site on the HER2 (and potentially HER3) receptor and has demonstrated efficacy in preclinical studies of oesophago-gastric adenocarcinoma, as well as in combination with trastuzumab in breast cancer, is currently being investigated in the phase III JACOB study (first-line treatment with trastuzumab and chemotherapy ± pertuzumab) in patients with HER2-positive metastatic or locally advanced unresectable gastro-oesophageal junction or gastric cancer[38]. Trastuzumab emtansine is an antibody-drug conjugate that utilizes HER2 overexpression to deliver a cytotoxic agent directly into neoplastic cells, and it is currently being evaluated in a second-line phase II/III trial in advanced gastric cancer[39].

TKIs of HER2: Lapatinib (Tykerb/Tyverb) is an oral small molecule dual TKI of EGFR and HER2 that has been approved for the treatment of HER2-positive advanced breast cancer progressing under trastuzumab and in association with hormonal therapy for triple positive metastatic breast cancer[40,41]. Lapatinib was studied in combination with standard chemotherapy in patients with HER2-positive advanced gastric and gastro-oesophageal junction adenocarcinomas in the phase III LOGIC study[42] without demonstrating an improvement in OS, although certain subgroups were shown to have a benefit, nor did the phase III Asian TyTAN trial demonstrate an OS or progression free survival (PFS) benefit for the lapatinib group as a second-line treatment, although there was a statistically significantly increased response rate[43].

The MAGIC-B trial is evaluating the addition of the HER2 TKI lapatinib to perioperative epirubicin, cisplatin, capecitabine (ECX) chemotherapy in a subgroup of patients whose tumours demonstrate HER2 overexpression[44].

EGFR inhibition

EGFR is a transmembrane glycoprotein[45] overexpressed in several gastrointestinal malignancies. Ligand binding to the extracellular domain determines EGFR activation and phosphorylation of the intracellular tyrosine kinase, leading to activation of Ras/Raf/mitogen-activated protein kinase or the Akt/mTOR pathway[46]. EGFR overexpression is found in 30%-50% of gastro-oesophageal malignancies, associated with a more aggressive histology and advanced stage[47]. In gastric cancer, EGFR is expressed at a relatively high level, whereas the mutation rate of KRAS is much lower than that in colorectal cancer (4.2%)[48].

Inhibition of EGFR can be undertaken using monoclonal antibodies (i.e., cetuximab and panitumumab) or TKIs (i.e., gefitinib, erlotinib).

Anti-EGFR monoclonal antibodies: Cetuximab (trade name Erbitux) is a chimeric monoclonal antibody that binds to the extracellular domain of human EGFR and inhibits ligand-induced EGFR tyrosine kinase activation[49]; it is currently approved for the treatment of advanced KRAS wild-type colorectal cancer, as well as squamous cell head and neck cancers[50,51]. Unfortunately, the phase III EXPAND trial (chemotherapy associated with cetuximab in advanced gastro-oesophageal cancers) did not show a PFS or OS benefit for cetuximab in the treatment of gastric cancer[52]. Other clinical trials evaluating cetuximab in gastric cancer settings have shown no benefits in PFS and OS, or they included insufficient numbers of patients[53-55], whereas others are pending results[56].

Based on numerous studies, there is no benefit of adding cetuximab to a first-line chemotherapy regimen for advanced gastric cancer, compared to a single-agent chemotherapeutic regimen; the former treatment represents an alternative choice for first-line treatment because of its lower toxicity. As a second-line treatment, combination regimens including cetuximab are advantageous; however, it is debatable whether these regimens will be used as standard treatment because of economic constraints[57-59].

Panitumumab (trade name Vectibix) is a fully human monoclonal anti-EGFR antibody. The REAL-3 study, a trial of the efficacy of chemotherapy with or without panitumumab in naïve patients with advanced oesophago-gastric cancer, did not show any benefit[60]. Other trials of panitumumab associated with different chemotherapy schemes were stopped early due to poor tolerability[61] or preliminary results from other trials[62]. There are still other trials ongoing using panitumumab[63,64].

The failure of the EXPAND and REAL3 trials could be explained by EGFR perhaps not being the major oncogenic pathway in advanced gastric cancer, and neither study evaluated patients for EGFR expression by IHC/FISH to select the treatment group. Therefore, it is essential to identify predictive markers to determine the population group that would most likely benefit from anti-EGFR therapy[65].

Nimotuzumab (h-R3) is a recombinant humanized monoclonal antibody with high binding specificity to human extracellular EGFR, with strong destructive effects on EGFR(+) cancer cells and showing some advantages compared to cetuximab, such as a longer half-life, higher dose-effect rate, and less severe dermatological toxicity[66]. In vitro, nimotuzumab showed effects of antiproliferation, antiangiogenesis and apoptosis on EGFR-overexpressed tumour cells[67,68]. A double-blind phase II trial[69] evaluated irinotecan-naïve patients with advanced gastric cancer who received nimotuzumab plus irinotecan vs irinotecan alone, and it showed no difference in PFS or OS between these two groups. However, it should be noted that a significant benefit was detected in EGFR2+/3+ subgroups. The phase III clinical trial NCT01813253 of nimotuzumab and irinotecan as a second line treatment for advanced or recurrent gastric or gastro-oesophageal cancer is currently recruiting patients[70] to determine the efficacy of nimotuzumab in these conditions. The efficacy of nimotuzumab seems to be proportional to the tumour surface EGFR density in patients, and the treatment is associated with a low adverse events rate.

A recent randomized phase II trial performed to test the effect of adding nimotuzumab to cisplatin and S-1 in untreated advanced gastric carcinoma showed no additional benefit over nimotuzumab, raising the hypothesis that a negative interaction could exist between nimotuzumab and S-1 chemotherapy[71].

TKIs of EGFR: Gefitinib (trade name Iressa) is an oral EGFR TKI with promising results against several types of malignancies in early phase trials. An open, single centre, phase I/II study of gefitinib in combination with chemoradiation in subjects with resectable gastric cancer (NCT00237900) is awaiting results[72]. Also, a multicentre phase II study was designed to evaluate tumour response rates in patients with surgically unresectable and/or metastatic oesophageal or gastric carcinomas treated with induction chemotherapy followed by gefitinib[73], and a phase III trial (NCT01243398) randomized patients with advanced oesophageal or gastro-oesophageal junction cancers to gefitinib vs placebo after progression on chemotherapy[74]. The pending results of these studies could help to define the role of gefitinib in gastric cancer.

Erlotinib (trade name Tarceva) is an oral EGFR TKI, which has been approved for the treatment of lung and pancreatic cancers. A phase II trial revealed that erlotinib was active in patients with gastro-oesophageal cancer (response rate of 9%), but it showed no responses in gastric cancer[75].

In conclusion, data from numerous current studies, using either small molecule TKI or monoclonal anti-EGFR antibodies, have revealed no significant curative effects of these agents as treatments for metastatic gastric cancer.

VEGF/VEGF receptor inhibition

Angiogenesis is a key element in tumour growth and spread. The importance of angiogenesis in tumour growth and dissemination was first discovered in 1971, when Folkman[76] proposed for the first time the hypothesis according to which “the solid tumours’ growth and the development of metastases are dependent of the formation of new blood vessels”[77,78].

Cancer development is vascular-dependent, and beyond a tumour volume of 2 mm3, tumour cells start to secrete a broad range of proangiogenic factors due to hypoxia to allow for growth and invasion. This fact provides the rationale for targeted antiangiogenic agents becoming a principal strategy in cancer therapy. Therefore, solid tumour growth depends on the development of new blood vessels[79]. To maintain unlimited tumour growth, the tumour tissue depends on the sustained formation of new blood vessels, which is essential for tumours’ growth beyond microscopic size, thus ensuring oxygenation and nutrient perfusion, as well as the elimination of the products of metabolism. Different from normal tissues, in which angiogenesis is strictly controlled, in neoplastic tissue, angiogenesis proved to be uncontrolled and immature[80].

Dvorak showed for the first time an association between tumour angiogenesis and microvascular permeability growth, which led to the identification of vascular permeability factor[81], which in turn was subsequently proved by Ferrara to be a specific angiogenesis inductor, known as VEGF[82].

VEGF is a mitogen and a stimulator of the endothelial cells with angiogenic effects in vivo[83,84], and its expression is correlated with the growth of blood vessels during angiogenesis[85,86], including angiogenesis in the female genital tract and with the development of the tumours. VEGF is a homo-dimeric protein of 40-45 kDa, secreted by a large variety of cells and by most tumour cells, existing in 5 different isomorphic forms (VEGF-A, B, C, D and E) and presenting two specific receptors in endothelial cells[87-90]. VEGF-A is an essential mediator of physiologic and pathologic angiogenesis[91]. VEGF-C is linked to Flt-4, preferably expressed by the lymphatic endothelium[92,93]. VEGF represents one of the most important tumour proangiogenic cytokines. VEGF expression is stimulated by the presence of hypoxia, and it is frequently higher in the proximity of necrosed areas[94,95]. Other effects of VEGF include inducing fenestrations in the endothelium of the small venules and capillaries, even in tissues in which microvascularization does not present normal fenestrations, which partially explains the enhanced permeability of tumour vessels[96,97].

VEGF has demonstrated a pivotal role in tumour angiogenesis, growth, and metastasis in numerous neoplasias, including gastric cancer[98], and for this reason, it is considered an essential therapeutic target for many tumours. Studies have shown that VEGF expression is related to the invasiveness, clinical stage and prognosis of gastric cancer[99].

Multiple anti-VEGF agents have been developed, including monoclonal antibodies (such as bevacizumab) and TKIs (such as apatinib) to inhibit the proangiogenic effects of VEGF or monoclonal antibodies directed against VEGF receptor (such as ramucirumab) to inhibit its function. Another antiangiogenic drug is recombinant human endostatin[68].

Anti-VEGF monoclonal antibodies: Bevacizumab (trade name Avastin) is the first FDA-approved anti-VEGF monoclonal recombinant humanized antibody for cancer treatment. This monoclonal antibody has been acknowledged for clinical use to treat numerous tumours, such as colorectal, breast, lung (non-small cell lung cancer, NSCLC), renal, and ovarian cancers and glioblastoma[100-105]. Bevacizumab treatment is associated with side effects such as thromboembolic events, gastrointestinal perforation, hypertension and proteinuria. The phase III multinational AVAGAST trial (Avastin in gastric cancer)[106], designed to evaluate the efficacy of adding bevacizumab to cisplatin-based chemotherapy as a first-line treatment for advanced gastric cancer, demonstrated that both median PFS and overall response rate were significantly improved in the bevacizumab group, without a significant benefit in OS.

This study evaluated a panel of tumour angiogenic factors, including neuropilin, to find the most appropriate group of patients who might benefit from antiangiogenic treatment[107,108]. Neuropilin, as well as baseline plasma VEGF-A, seemed to be a promising prognostic biomarker, and with the potentially predictive properties for bevacizumab, its role is still under research.

There was another phase III study, AVATAR, performed in China, which also found that bevacizumab added to capecitabine/cisplatin chemotherapy did not significantly improve OS in patients with advanced gastric cancer[109].

Unfortunately, the negative results of the AVAGAST and AVATAR studies might have derived from not having selected the most suitable gastric cancer patients for bevacizumab treatment; therefore, biomarkers and more fundamental research on the molecular typing of gastric tumours are needed for this purpose.

The MAGIC-B study (medical research council adjuvant gastric infusional chemotherapy) from United Kingdom is currently assessing the role of bevacizumab for peri-operative chemotherapy in resectable adenocarcinoma of the stomach and gastro-oesophageal junction, randomizing patients to receive the perioperative regimen administered in the original MAGIC trial[110], namely ECX with or without bevacizumab[44]. Hopefully, this trial will allow for the detection of predictive biomarkers that could identify the subset of patients with the greatest potential benefit from the use of perioperative VEGF-A inhibitory monoclonal antibody[111].

Currently, the safety and efficacy of adding bevacizumab to taxane-based chemotherapeutic regimens or irinotecan in advanced/metastatic gastric cancer is being evaluated in several clinical trials with pending results[112-114]. Also, studies are investigating the associated administration of bevacizumab plus anti-Her2-targeted treatment (trastuzumab) in the Her2(+) subset of gastric cancer patients[115,116]. Furthermore, several trials are assessing the benefit of administrating bevacizumab in neoadjuvant settings[117,118].

Anti-VEGF receptor monoclonal antibodies: Ramucirumab (trade name Cyramza) is a fully human monoclonal antibody that potently inhibits VEGFR-2. Based on the phase III REGARD study (ramucirumab monotherapy for previously treated advanced gastric or gastro-oesophageal junction adenocarcinoma), which found significantly longer OS for ramucirumab used as a second line chemotherapy in gastric or gastro-oesophageal junction adenocarcinoma patients vs best supportive care (BSC)[119], the FDA approved this antibody for use as a single agent in gastric and gastro-oesophageal junction cancer after progression on a platinum- or fluoropyrimidine-containing regimen (2014)[120].

A phase III clinical trial of ramucirumab plus paclitaxel vs placebo plus paclitaxel in the treatment of metastatic gastro-oesophageal junction and gastric adenocarcinoma following disease progression on first-line platinum- and fluoropyrimidine-containing combination (RAINBOW trial) demonstrated significantly longer PFS and OS for the ramucirumab group[121], also leading to approval by the FDA of ramucirumab in combination with paclitaxel as a second-line therapy. Ramucirumab is currently the only antiangiogenic agent that has been approved for the treatment of advanced gastric cancer[122]. However, ramucirumab treatment has been associated with significant adverse reactions, such as neutropenia, leukopenia, hypertension and decreased strength, which must be considered. Unfortunately, a phase II study of ramucirumab combined with the FOLFOX regimen did not show an improvement of early cure as the primary endpoint of PFS in advanced gastric cancer, once again emphasizing the urgent need for molecular biomarkers to select the most suitable patients for antiangiogenic treatment[123].

Endostar (YH-16, recombinant human endostatin) is a novel recombinant human endostatin, found to inhibit significantly VEGF-induced tumour cell proliferation and migration and to suppress VEGFR-2 expression[124]. This agent was approved for the treatment of non-small-cell lung cancer by China’s Food and Drug Administration (CFDA) (2005). Xu et al[125] investigated the efficacy and safety of Endostar combined with SOX (S-1/oxaliplatin) for the first-line treatment of patients with advanced gastric cancer; although the curative effect was not significantly different, their results showed significantly better PFS for the group including Endostar, without the addition of severe toxicity. More trials of the efficacy and safety of Endostatin in gastric cancer settings are needed.

TKIs of VEGF: Apatinib (YN968D1) is an anti-VEGF-2 small molecule TKI developed in China[126]. Li et al[127] reported a phase II, randomized, placebo-controlled study that treated patients with metastatic adenocarcinoma gastric cancer with apatinib monotherapy who did not respond at least two chemotherapeutic regimens. There were statistically significant differences between the Apatinib and placebo groups for both OS and PFS, with adverse effects, such as handfoot syndrome, proteinuria and hypertension, which were clinically managed. The results of a phase III, randomized, double-blind, placebo-controlled clinical study treating advanced or metastatic adenocarcinoma of the stomach or gastro-oesophageal junction with apatinib[128], which included patients from 32 centres in China with advanced gastric cancer who did not respond to second-line chemotherapy treatment, showed that median OS was significantly longer (by 55 d), and PFS was also significantly prolonged in the Apatinib group vs placebo. The disease control rate was approximately 50% in this group, although the efficiency of treatment was not as high.

Studies have demonstrated that apatinib was the first discovered anti-VEGF-2 small molecule TKI with benefits for Asian patients with advanced gastric cancer, representing remarkable progress for third-line treatment in this setting, although it prolonged OS by less than 2 mo. Further data are needed to assess the efficacy and safety of this molecule in other ethnic groups (e.g., Caucasians).

This phase III study of apatinib reassured scholars regarding the existence of a favourable safety profile of the agent compared to other antiangiogenic molecules, especially with regard to cardiovascular toxicity, making it a promising option for recurrent gastric cancer[129]. Data have proved that biomarkers could identify the subset of patients most likely to benefit from the antiangiogenic agents. The AVAGAST trial demonstrated a positive correlation of bevacizumab response with the levels of VEGF-A in serum and neuropilin-1 in tumour tissue in non-Asian patients[108], providing evidence to support further biomarker investigation for antiangiogenic treatment. A biomarker study of apatinib in patients with breast cancer revealed that both hypertension and high expression of phosphorylated VEGFR-2 could serve as potential markers for assessing treatment response[130].

Based on the positive results from the phase III study in stomach cancer patients, apatinib was approved in October 2014 by the CFDA for metastatic gastric or gastro-oesophageal junction adenocarcinoma after second-line chemotherapy progression[131].

Sunitinib (trade name Sutent, previously known as SU11248) represents an oral multi-targeted TKI of VEGFR, platelet-derived growth factor receptor (PDGFR), c-KIT (stem cell factor receptor), rearranged during transfection, and FMS-like tyrosine kinase-3 receptor, exerting antitumor effects. It is currently indicated for the treatment of advanced renal cell carcinoma, gastrointestinal stromal tumours showing imatinib resistance/intolerance and progressive unresectable/metastatic neuroendocrine pancreatic tumours[132].

A phase II trial of single agent sunitinib as a second line monotherapy in patients with advanced gastric and gastroesophageal cancer, showed a median OS of 6.8 mo[133]. Sunitinib was also evaluated in combination with chemotherapy (Docetaxel) in patients with metastatic gastric cancer, showing a similar time to progression but an increased response rate of 41.4% vs 14.3%[134].

Recent phase I and II clinical trials have confirmed the efficacy and tolerability of sunitinib combined with standard chemotherapies. Its clinical benefit in advanced gastric cancer treatment requires further studies[135,136].

Sorafenib (trade name Nexavar) is a multitargeted TKI. It is an inhibitor of the RAF tyrosine kinase and several other receptor tyrosine kinases (VEGFR-2, VEGFR-3, and PDGFR-β), and it is approved for the treatment of advanced hepatocellular carcinoma (HCC) and clear-cell renal-cell carcinoma[137,138]. In animal models of gastric cancer, sorafenib proved to inhibit tumour growth and angiogenesis[139]. Phase I and II trials are evaluating the efficacy of sorafenib for the treatment of advanced gastro-oesophageal junction carcinoma in combination with chemotherapy (FOLFOX4, S1 + cisplatin)/as a single agent[140-142]. A phase II study including gastric cancer patients treated with sorafenib combined with docetaxel and cisplatin (second line) showed very long median PFS and median OS (5.8 and 13.6 mo, respectively)[143], although other studies have been terminated early because of low response rates[144,145].

A phase I-II study of sorafenib + capecitabine and cisplatin, including 21 patients with advanced gastric cancer, obtained a median PFS of 10 mo, a response rate of 63% and a median OS of 14.7 mo[146].

Pazopanib (trade name Votrient) is an oral agent that inhibits angiogenesis through multiple pathways (VEGFR, PDGFR, c-KIT), and it is approved by the FDA for use in the treatment of both metastatic renal cell carcinoma and soft tissue sarcoma[147,148]. A phase I study of pazopanib in combination with cisplatin in patients with advanced solid tumours found that, despite activity observed in some patients, the potential to combine pazopanib and cisplatin appeared limited due to the safety profile and the pharmacokinetic interactions within the combination[149]. Another phase I study evaluating the administration of pazopanib and a MET inhibitor in advanced solid tumours[150] is pending results.

There are two ongoing phase II trials investigating pazopanib in advanced gastric tumours: the PaFLO trial (FLO ± pazopanib as first-line treatment)[151] and another trial adding pazopanib to capecitabine and oxaliplatin[152]. We are hoping that the results of these studies will determine whether there is a role for pazopanib in the treatment of advanced gastric cancer.

Regorafenib (BAY 73-4506) is an oral multikinase inhibitor that activates protein kinases, such as VEGFR and RAF[153], demonstrating efficacy in numerous tumours in different clinical trials. It has received approval for the treatment of advanced colorectal cancer (2012) and also for advanced gastrointestinal stromal tumours (2013)[154]. The antiangiogenic effect of regorafenib might occur via the principal antitumor pathway in gastric tumours; trials using xenograft models of gastric cancer have demonstrated a decreased level of VEGFR-2 proteins secondary to regorafenib[155].

A phase II, double-blind, placebo-controlled trial investigating the efficacy of regorafenib in the treatment of refractory advanced esophagogastric cancer noted a significantly longer median PFS for the regorafenib group vs the placebo group (11 wk vs 3.9 wk) and a superior median OS (25 wk vs 19.4 wk)[156].

Although current data from the literature show a benefit of regorafenib in the second-line treatment of advanced gastric cancer, drug-related toxicity represents a serious problem for patients. The ongoing phase I and II trials assessing the role of regorafenib as single agent or in combination with other chemotherapeutics, such as FOLFOX or paclitaxel[157-159], mainly in a second-line setting, will better delineate the role of regorafenib in advanced gastric cancer.

IGF-1 inhibition

IGF-1 receptor (IGF-1R) is a transmembrane tyrosine kinase receptor that is activated after binding to IGF-1 and IGF-2, promoting tumour angiogenesis, growth, progression, anti-apoptosis and metastasis in several tumours, including gastro-oesophageal cancers[160]. Matsubara et al[161] found that IGF-1R expression in resected gastric tumours was correlated with poorer clinical outcomes. It was shown that patients with gastric cancers expressing low levels of both IGF-1R and EGFR had significantly longer OS. IGF-1R signalling has been associated with resistance to cytotoxic therapy, and inhibition of IGF-1R enhanced tumour cell apoptosis in numerous models[162]. The IGF-1R pathway can be targeted through monoclonal antibodies, IGF-1R antisense/small interfering ribonucleic acid (siRNA), and receptor tyrosine kinases.

Figitumumab (CP-751871) represents a completely humanized IgG2 monoclonal antibody against IGF-1R. A phase I clinical study with figitumumab in combination with docetaxel demonstrated positive results[163], and phase I and II trials have also confirmed roles for this agent in Ewing’s sarcoma and NSCLC; however, a phase III trial investigating NSCLC was terminated early due to not reaching the endpoint[164-166]. Some phase I clinical studies have evaluated the overall safety, tolerability and pharmacokinetic profile of figitumumab administrated in different chemotherapeutic combinations in patients with advanced solid tumours[167,168].

The role of figitumumab in gastric cancer treatment requires further research.

Fibroblast growth factor TKIs

Fibroblast growth factor (FGF) and its signalling receptors are involved in cell proliferation, differentiation, and transformation[169]. FGFR2 amplification occurs in approximately 5%-10% and polysomy in > 20% of gastric cancers, mostly in poorly differentiated gastric cancer (scirrhous cancer)[170].

In animal models of gastric cancer, AZD2171 (AZD), a potent oral VEGF, FGFR1, PDGFRB, and VEGFR-2 TKI, led to tumour inhibition, with the most potent activity seen in tumours over-expressing FGFR2, suggesting a possible clinical benefit of AZD2171 in patients with FGFR2(+) gastric tumours[171]. At the 2015 ASCO meeting, Bang et al[172] presented the results of a phase II study of AZD2171 vs paclitaxel in previously treated patients with advanced gastric cancer. They revealed that AZD was well-tolerated, but there was no statistically significant difference in PFS in favour of the AZD group in FGFR2 amplified or polysomy patients selected by FISH. Marked intra-tumour heterogeneity of FGFR2 amplification and low concordance with elevated FGFR2 expression were reported. The study provided evidence that AZD at this dose and with this schedule caused pharmacologic target inhibition.

The pan-FGFR TKI JNJ-42756493 is an FGFR 1, 2, 3, and 4 inhibitor with nanomolar affinity; orally bioavailable, it has demonstrated broad spectrum antitumor activity in cell line, xenograft and patient-derived explant models with abnormality in the FGFR signalling pathway, such as FGFR gene amplification, mutation and translocation. A multipart, phase I, first in humans study of JNJ-42756493 was initiated in advanced solid tumour patients (including gastric cancer) (NCT01962532); promising interim data showed that this agent had excellent pharmaceutical properties and appeared safe with manageable side effects at dose levels that elicited anti-tumour activity[173].

Ki23057 is an oral broad-range TKI of FGFR2, FGFR1 and VEGF2 that inhibits the proliferation of gastric scirrhous cancer cells presenting FGFR2 gene amplification. Nakamura and collaborators revealed that Ki23057 inhibited the growth and peritoneal dissemination of gastric cancer cells, mainly through FGFR2-RAS/extracellular-regulated kinase (ERK) inhibition[174]. The study of Qiu et al[175] found that the FGFR2 inhibitor Ki23057 might be therapeutically promising for treating drug-resistant gastric cancer cells, especially when used in combination with other chemotherapeutic drugs. The synergistic effects of these combinations might be driven by the apoptosis process and the involvement of the ERCC1 and p53 genes[175].

Dovitinib (TKI 258) strongly inhibits the phosphorylation of FGFR3, leading to the inhibition of tumour cell proliferation and the induction of tumour cell death. Furthermore, this agent might inhibit other members of the RTK superfamily, including VEGFR, FGFR1, PDGFR type 3, FMS-like tyrosine kinase 3, stem cell factor receptor (c-KIT), and colony-stimulating factor receptor 1, resulting in an additional reduction in cellular proliferation and angiogenesis and the induction of tumour cell apoptosis. The activation of FGFR3 has been demonstrated to be associated with cell proliferation and survival in certain cancer cell types[176,177].

Brivanib is the alanine ester of the VEGFR-2 inhibitor BMS-540215, which is a dual TKI that has shown potent and selective inhibition of VEGFR and FGFR tyrosine kinases[178,179]. Furthermore, BMS-540215 has been shown to inhibit selectively the proliferation of endothelial cells stimulated by VEGF and FGF in vitro. It has also shown broad-spectrum in vivo antitumor activity, with a suggested role in the treatment of HCC[180-182].

The FGFR inhibitor INCB054828 is an orally bioavailable inhibitor of FGFR types 1, 2, and 3 (FGFR1/2/3), with potential antineoplastic activity by inhibiting proliferation in FGFR1/2/3-overexpressing tumour cells.

Phase I and II clinical trials using dovitinib, brivanib and INCB054828 in patients with advanced gastric cancer are ongoing; we expect this research to add new data regarding the role of FGF inhibitors in this type of tumour[183-185].

Hepatocyte growth factor/c-MET (mesenchymal-epithelial transition factor receptor) inhibitors

C-MET represents a receptor tyrosine kinase that is expressed in both epithelium and endothelial cells; its overexpression and activating mutations have been demonstrated in many tumour types, including gastric cancer, and it has been correlated with poor prognosis[186]. Its ligand is represented by hepatocyte growth factor (HGF), which is expressed by cells of mesenchymal origin. c-MET and its signal pathway activation induced gastric cancer cell proliferation, survival, and migration[187]. There are various types of HGF/MET inhibitors, such as monoclonal antibodies (rilotumumab and onartuzumab) and small molecule tyrosine kinase (foretinib).

Anti-HGF/c-MET monoclonal antibodies: Rilotumumab (AMG102) is a human monoclonal antibody directed against HGF, and it was demonstrated by a phase II study to show efficacy in a subset of locally advanced/metastatic gastric or oesophago-gastric junction cancer patients with MET overexpression by IHC. Data have shown that adding rilotumumab to standard chemotherapy regimens provided some survival benefit[188,189]. The results of the phase III RILOMET-1 trial, an international, phase III, multicentre, randomized, double-blind, placebo-controlled trial of rilotumumab plus ECX as a first-line therapy in patients with advanced MET-positive gastric or gastro-oesophageal junction adenocarcinoma, were presented at the ASCO 2015 annual meeting, revealing that both OS and PFS were statistically worse in the rilotumumab group, independent of MET expression[190]. Due to the increased toxicity of the drug and treatment-related deaths in the combination group in the RILOMET-1 trial, all of the clinical trials investigating the role of rilotumumab in gastric cancer, including the phase III RILOMET-1 (with ECX) and RILOMET-2 (with cisplatin and capecitabine) trials, were terminated early.

A multicentre, open-label, randomized, phase II trial is ongoing in 30 centres in France, investigating the efficacy of FOLFOX alone or in combination with rilotumumab (AMG 102) or panitumumab as a first-line treatment for patients with advanced gastroesophageal adenocarcinoma[64].

Onartuzumab is a humanized monovalent antibody directed against MET that is also being evaluated in a first line, randomized, phase III trial in MET-positive, HER2-negative GE patients in combination with mFOLFOX6. Data regarding this study were presented at the 2015 ASCO annual meeting, revealing unfortunately that the combination treatment could not prolong OS, not even in the group with MET highly expressed (MET 2+/3+). Severe adverse reactions, such as the reduction of neutrophils, thrombocytopenia, oedema, and pulmonary embolism, were not significantly increased in the combined group[191].

The possible explanations for obtaining negative results with these monoclonal antibodies might be that the MET gene is not the driver gene of gastric cancer; currently, there is not an accurate assessment of MET protein expression or a demonstration that gastric tumours present c-MET heterogeneity.

Anti-HGF/c-MET tyrosine kinase: Foretonib (GSK1363089) is an oral small molecule inhibitor of c-MET and VEGFR-2A. Kwak[192] reported good efficacy and tolerability of this agent in a phase I clinical study, leading to FDA approval of the drug to enter directly into phase III trials for the treatment of NSCLC.

Foretonib was demonstrated in preclinical studies to inhibit effectively the growth of gastric cancer cells[193]. It was also investigated in a phase II study as a single agent in patients with metastatic gastric adenocarcinoma (c-MET unselected subjects), showing good tolerability but only minimal antitumor efficacy[194]; these results indicated that few gastric carcinomas are driven solely by MET and VEGFR2, emphasizing the diverse molecular oncogenesis of this disease[195].

PI3 kinase/mammalian target of the rapamycin pathway inhibition

Studies have shown that PI3K enzymes play a role in the phosphorylation of membrane inositol lipids[196,197]. Upregulation of the PI3k/Akt/mTOR pathway was correlated with poor prognosis and could contribute to the chemoresistance of gastric tumours[198]. Data showed that the mTOR signalling pathway was frequently used in gastric tumoural cells. Studies in vitro and animal models reported that blockage of the mTOR signalling pathway could inhibit the proliferation and metastasis of gastric cancer cells[199].

Everolimus (RAD-001, Afinitor) is an oral mTOR inhibitor that prevents the phosphorylation of p70S6K and 4E-BP1 mediated by mTOR, resulting in G0/G1 arrest[200]. The FDA approved everolimus for the treatment of non-responding advanced renal cell carcinomas (2009)[201], treatment of progressive neuroendocrine tumours of pancreatic origin (2011)[202], paediatric and adult patients with subependymal giant cell astrocytoma[203] and hormone receptor-positive and HER2-negative advanced postmenopausal breast cancer patients (2012)[204]. Anticancer activity has been shown in both phase I and phase II studies, which concluded that everolimus monotherapy as a second-line treatment had a good disease control rate for advanced gastric cancer patients[205,206].

Unfortunately, the phase III GRANITE-1 trial investigating the safety and efficacy of everolimus monotherapy as a second or third line in patients with advanced gastric cancer did not show a survival benefit. Furthermore, grade 3-4 adverse reactions, including anaemia, loss of appetite, and fatigue, were observed in these patients[207,208]. The use of everolimus in the treatment of advanced gastric cancer must be further investigated.

Sirolimus (also known as rapamycin) has potent immunosuppressive and antiproliferative properties due to its ability to inhibit mTOR. A phase I trial is ongoing of mTOR inhibition with rapamycin (sirolimus) for enhancing intranodal dendritic cell vaccine-induced anti-tumour immunity in patients with cancer-testis antigen (NY-ESO-1)-expressing solid tumours, including gastric cancer. Biological therapies, such as sirolimus, might stimulate the immune system and stop tumour cells from growing. Vaccines made from a person’s white blood cells mixed with tumour proteins might help the patient to build an effective immune response to kill tumour cells that express NY-ESO-1[209].

PARP inhibitors

The DNA damage response is the coordinated activation of sensors and effectors, inducing cell cycle arrest, activation of DNA repair, and, if necessary, subsequent induction of apoptosis.

PARP inhibitors prevent the cancer cell’s single stranded break repair mechanism, allowing tumour cell death to occur[210]. These inhibitors have shown some efficacy in BRCA1- and BRCA2-deficient breast and ovarian cancers.

A phase II trial in metastatic/recurrent gastro-oesophageal cancer studied the benefit of administrating the PARP inhibitor olaparib (AZD-2281, trade name Lynparza). Patients received as a second-line treatment paclitaxel with or without olaparib[211]; the results demonstrated improved OS in the olaparib group, although PFS was not significantly different. In accordance with preclinical data showing greater olaparib sensitivity in patients with low ataxia telangiectasia-mutated (ATM) protein[212,213], this study found that low ATM patients had improved OS with olaparib. A phase III study of second line treatment using paclitaxel with or without olaparib in advanced gastric cancer patients is currently ongoing[214].

Olaparib is also being investigated in a trial evaluating the role of some novel antitumoural agents. This is a phase I, open-label, multicentre study of AZD6738 administered orally in combination with cytotoxic chemotherapy regimens (carboplatin) and/or novel anti-cancer agents (olaparib, durvalumab) to patients with advanced malignancies, including gastric cancer. It is currently recruiting patients[215]. AZD6738 is an ataxia, telangiectasia and Rad3-related (ATR) serine/threonine protein kinase inhibitor. ATR is a member of the PI3K-like kinase family, activated by replication-associated DNA damage. AZD6738 induces S-phase cell cycle delay consistent with replication stress and increased sub-G1 “cell death” populations in vitro. Durvalumab is an Fc-optimized monoclonal antibody directed against programmed cell death-1 ligand 1 (PD-L1; B7 homolog 1; B7H1), with potential immune checkpoint inhibitory and antineoplastic activities.

Veliparib (ABT-888) is another PARP inhibitor developed to help prevent DNA repair in cancer cells and to increase the effectiveness of common DNA-damaging therapies, such as chemotherapy or radiation. Veliparib is currently being studied in many cancers and tumour types, including phase III studies of breast cancer and NSCLC. A study of the efficacy of veliparib combined with the FOLFIRI regimen in gastric cancer is awaiting results[216].

The influence on DNA repair mechanisms in tumours opens a new dimension of research in the targeted therapy of gastric cancer[217].

Immunotherapy/Immuno-checkpoint blockade

Because tumours evade host immune recognition through a multitude of mechanisms[218], immunotherapy has developed as a novel field of anticancer treatment, which acts by increasing immune activity and using the blockage mechanism of the inhibitory immune regulatory pathways. Recent data have shown that programmed cell death-1 (PD-1) represents an immunoinhibitory receptor belonging to the CD28 family that plays a crucial role in the phenomenon of immune escape of tumours[219]; one of the most important mechanisms consists of activating immune checkpoints that block host T-cell activation. The presence of PD-L1 is needed for tumour cells to escape the cytotoxic effects of T cells. Promising new agents targeting immune checkpoints (PD-1 and PD-L1) have been recently developed.

Ipilimumab (trade name Yervoy, formerly known as MDX-010 and MDX-101) releases a negative immune regulatory pathway by blocking the inhibitory receptor called cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4). The FDA approved ipilimumab for unresectable/metastatic melanoma patients who received at least one prior systemic treatment for melanoma (2011)[220]. It is undergoing clinical trials for the treatment of NSCLC, small cell lung cancer (SCLC)[221], bladder cancer[222] and metastatic hormone-refractory prostate cancer[223]. As a new maintenance concept, sequential administration of immunotherapy might prolong the clinical benefit of first-line chemotherapy before disease progression. Unfortunately, a phase II trial comparing the efficacy of ipilimumab and BSC after first-line chemotherapy in unresectable locally advanced or metastatic gastric/gastro-oesophageal cancer patients revealed detrimental PFS with ipilimumab vs the BSC group (2.7 mo vs 4.8 mo, P = 0.03), as well as no statistically significant improvement in OS (16.7 mo vs 12 mo, P = 0.64)[224].

Nivolumab (Opdivo, anti-PD-1, BMS-936558, ONO-4538) acts by blocking the binding of receptor inhibitor PD-1, which is expressed on T-cells, to PDL-1 which prevents T cell death. Blockade of interactions between PD-1 and PD-L1 stimulated immune function in vitro and mediated antitumor activity in preclinical models. A phase I trial of nivolumab in advanced solid tumours also included patients with gastric adenocarcinoma. Antibody-mediated blockade of PD-L1 induced durable tumour regression and prolonged the stabilization of disease in patients with advanced cancers. Unfortunately, gastric cancer patients were not included in the efficacy analysis[225,226].

A phase I/II study of nivolumab monotherapy or nivolumab combined with ipilimumab in subjects with advanced or metastatic solid tumours, including gastric cancer, is still recruiting patients[227]. Available data for the group using the single agent nivolumab revealed that nivolumab monotherapy was well tolerated, and it demonstrated encouraging antitumor activity in heavily pretreated gastric cancer patients. Objective responses occurred in patients with both PD-L1-positive and -negative tumours[228].

Also, a phase III trial is currently investigating the safety and efficacy of nivolumab in patients with unresectable advanced or recurrent gastric cancer refractory to or intolerant of standard therapy[229].

Pembrolizumab (MK-3475) is an agent that blocks the binding of PD-1 to PDL-1 (as well as PDL-2). A phase 1 study of pembrolizumab in recurrent and metastatic gastric and gastro-oesophageal junction adenocarcinoma patients with PD-L1 tumour positivity demonstrated good tolerability, as well as anti-tumour activity[230]. Other phase I-III trials of pembrolizumab are investigating this agent in advanced gastric cancer, administered as monotherapy or associated with chemotherapy in naïve patients/after progression under treatment[231,232], with the aim of integrating the molecular subtypes of gastric cancer through integrative genomic analysis[233]. Other phase I/II studies are evaluating the efficacy of pembrolizumab in combination with other classes of agents, such as anti-HER2 monoclonal antibodies[234,235], anti-VEGFR monoclonal antibodies[236] or multitargeted TKIs (PLX 3397)[237]. An innovative approach in gastric cancer treatment, included in a phase I study, consists of administering of pembrolizumab in combination with PEPGH20, a new molecular entity for tumours rich in hyaluronan (HA) that form a coating over the surface of the tumour cell; this agent removes the HA coating[238].

Durvalumab (MEDI4736), an antiPDL-1 agent, has shown some efficacy in gastric cancer[239].

Based on previous data showing a synergistic effect of the combination of CTLA-4 and PDL-1 blocking agents, the combination of durvalumab and tremelimumab (anti-CTLA-4) plus first-line chemotherapy is being investigated in advanced solid tumours, including gastric neoplasms[240,241]. A phase I/II study is currently assessing the safety, tolerability, pharmacokinetics, immunogenicity and preliminary efficacy of INCB024360 in combination with MEDI4736 in subjects with selected advanced solid tumours, including gastric cancer[242]. Epacadostat (INCB 024360) is a potent and novel indoleamine-2,3 dioxygenase (IDO) inhibitor. IDO is an immunomodulatory enzyme produced by some alternatively activated immunoregulatory cells, and it is used as an immune subversion strategy by many tumours. The molecule exhibits potent in vitro and in vivo immunomodulating and antineoplastic activities.

Other ongoing trials are evaluating pembrolizumab in association with other classes of targeted therapies in advanced gastric tumours, such as anti-HER2 molecules as maintenance treatment[243], anti-VEGFR antibodies[244], and ATR serine/threonine protein kinase inhibitors[215].

Guanylyl cyclase C inhibitors

Guanylyl cyclase C (GCC) is a trans-membrane cell surface receptor, expressed not only on normal intestinal tissue but also on the tumour cells of patients with gastrointestinal neoplasias, precisely in approximately 95% of cases of metastatic colorectal cancer, and subsets of gastric and pancreatic tumours. Because the epithelial tight junctions are altered in tumour tissue, systemically delivered GCC-targeting agents would have access only to GCC receptors expressed by the tumour tissue. Tumour GCC expression seems be a good prognostic marker.

MLN0264 consists of a fully human monoclonal antibody targeting GCC that has shown antitumor efficacy in xenograft models of GCC-expressing tumours. A phase I trial evaluating the safety, maximum tolerated dose, and clinical pharmacokinetics of MLN0264 in patients with gastrointestinal malignancies expressing GCC showed promising results, demonstrating good tolerability of the molecule[245,246]. A phase II study of MLN 0264 in previously treated patients with gastric and gastro-oesophageal junction cancers whose tumours express GCC by IHC is currently recruiting patients[247]. Another phase I/II study is evaluating the effects of MLN0264 in previously treated Asian patients with advanced gastrointestinal carcinoma or metastatic/recurrent gastric or gastro-oesophageal junction adenocarcinoma expressing GCC; we are waiting to confirm the benefit of this agent in the treatment of gastric malignancies[248].

Inhibitors of the tumour cell cycle

In the tumour setting, there is an alteration of cell cycle regulatory mechanisms accomplished by molecules such as cyclins, cyclin-dependent kinases (CDKs) and CDK inhibitors (CKIs). CDKs bind to cyclins, facilitating cell cycle progression, whereas the combination of CDKs with CKIs leads to inhibition of cell cycle progression/apoptosis[249].

Flavopiridol (Alvocidib, HMR-1275), a semi-synthetic flavonoid, CKI, was the first inhibitor of the cell cycle investigated in a clinical trial. This inhibitor is in development as a frontline combination therapy for acute myeloid leukaemia and relapsed/refractory chronic lymphocytic leukaemia and other haematological disorders. Flavopiridol suppresses messenger RNA translation, stopping cell proliferation-related protein expression[250]. Unfortunately, flavopiridol demonstrated low efficacy and serious adverse reactions in gastric cancer[251]. Because, as a single agent, it seems to have poor anti-tumour activity, it must be investigated in combination with other chemotherapeutic agents. Its efficacy was assessed in combination with irinotecan in advanced gastric tumours in a phase II clinical trial, but there was only a small number of patients included[252].

Agents inducing tumour cell apoptosis

The induction of tumour cell apoptosis is a promising target in cancer treatment.

Tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) induces apoptosis in many tumours. Although gastric tumour cells often exhibit resistance to TRAIL, studies have demonstrated that some standard chemotherapeutics might enhance sensitivity to TRAIL[253]; therefore, the association of TRAIL with different chemotherapeutics for the treatment of gastric cancer is worth investigating.

NF-κB expression is positively correlated with the degree of malignancy and is negatively correlated with cancer prognosis. Bortezomib (PS-341, Velcade) is a highly selective and potent, reversible proteasome inhibitor that specifically inhibits the chymotrypsin activity of the 26S proteasome, subsequently inhibiting activation of the NF-κB signalling pathway. The agent has a broad spectrum of in vitro and in vivo activity[254,255]. It has FDA approval for multiple myeloma and mantle cell lymphoma[256,257]. Bortezomib demonstrates anti-cancer activity in gastric cancer cell lines, potentially mediated by the inhibition of the ERK1/2 and Akt signalling pathways. In gastric cancer, preclinical studies demonstrated synergistic growth inhibition of this agent in combination with conventional chemotherapy[258,259]. However, the combination of irinotecan and bortezomib was assessed in a multicentre phase II study, which concluded that bortezomib was not effective for the treatment of advanced adenocarcinoma of the stomach, whether used alone or in combination with irinotecan[260]. Also, another phase II study of metastatic gastro-oesophageal neoplasias explored the response rate with a first-line, three-drug regimen that consisted of bortezomib in combination with paclitaxel and carboplatin; the scheme was considered inactive, prompting premature study closure[261]. Based on these data, a multi-institution phase II study of single agent bortezomib in gastric adenocarcinoma was performed to determine anti-tumour efficacy[262]. Unfortunately, this trial showed no benefit of bortezomib monotherapy in locally advanced/metastatic gastric neoplasia.

Future studies of the class of proteasome inhibition in gastric setting are needed and should include combination regimens with targeted agents acting on different oncogenic pathways.

Matrix metalloproteinase inhibitors

The matrix metalloproteinase (MMPs) constitute a family of proteolytic enzymes that cause the breakdown of connective tissue proteins and that play major roles in the normal processes of growth, differentiation and repair. There are tissue inhibitors (TIMPs) that tightly regulate their activity. The aberrant synthesis of MMPs determines local tumour invasion by destroying the extracellular matrix and the basement membrane. Studies have associated the high expression of some MMPs with a poor prognosis of gastric cancer[263,264]; therefore, it could be feasible that specific MMP inhibitors might restore the normal balance of proteolytic activity, preventing tumour growth and metastasis.

Marimastat (BB-2516, TA-2516) is a broad-spectrum MMP inhibitor that proved to exert anti-tumour activity in gastric cancer. A randomized, double-blind, placebo-controlled study designed to evaluate the ability of marimastat to prolong survival in patients with non-resectable gastric and gastro-oesophageal adenocarcinoma provided the first indication of a survival benefit for an MMP inhibitor, supporting a possible role for this inhibitor as a maintenance treatment with response/stable disease following chemotherapy[265].

The correlation of proteolytic gene expression (MMP and TIMP) profiles with clinicopathological features of the tumour could aid in revealing tumour behaviour and could guide management strategies in gastric cancer[266].

CONCLUSION

Despite the decrease in incidence in past decades, gastric cancer remains a major global health problem; therefore, efficient treatments are needed to achieve increased OS and improved prognosis of patients.

Data from the literature have indicated that eradication of H. pylori could prevent gastric cancers. However, carcinogenesis is a multistep process, and it is unclear at which step H. pylori eradication would prevent gastric cancer onset. There may be a so-called “point of no return” in the histological cascade from chronic gastritis to intestinal-type adenocarcinoma after which H. pylori therapy is unlikely to prevent stomach cancer development. This “point of no return” may be represented by the appearance of gastric preneoplastic lesions. Thus, H. pylori eradication may have a decreased efficacy in preventing stomach neoplasia once these lesions appear.

For some decades, standard chemotherapeutic agents have been the mainstay of systemic treatment, unfortunately leading to reduced rates of response, poor clinical outcomes and compromised quality of life due to their significant associated toxicities.

Due to major advances in cancer research, numerous molecular alterations underlying gastric carcinogenesis and multiple signalling pathways involved in gastric cancer development and metastasis have been identified.

Along with these major discoveries, numerous drugs targeting genes and molecular signalling pathways have been investigated in clinical trials. The positive results obtained with some of these agents in gastric cancer treatment have led to improved outcomes, especially in advanced cancers, by incorporating them into the standard treatments for patients. The 2015 edition of the gastric cancer guideline from the United States National Comprehensive Cancer Network therefore recommends trastuzumab as the first line and ramucirumab as the second line for the treatment of advanced gastric tumours. Moreover, based on positive results from a phase III study, apatinib was approved by the CFDA for metastatic gastric/gastro-oesophageal junction tumours as the third line treatment.

Because of the complexity of the molecular alterations and the simultaneous activation of multiple signalling pathways in gastric cancer, targeting a single molecular pathway using one of the novel agents as a monotherapy or even in combination with standard chemotherapeutic regimens might be insufficient to provide a significant benefit to patient survival. One potential option would be to combine targeted drugs that act on different molecular pathways with the goal of achieving a synergistic effect, but our knowledge about these schemes currently remains limited, although they are being investigated in clinical trials that that have the purpose of determining the efficacy, safety and possible increased toxicities of these associations. Perhaps a possible solution to improve efficacy would be the development of novel multi-targeted molecules. Another drawback is represented by the very high costs of these molecular agents, which must decrease to be used widely.

There are impressive emerging of new agents involved in different stages of clinical studies, waiting to demonstrate their roles in the treatment of gastric neoplasias. Future directions in gastric cancer treatment include novel classes of targeted agents, such as c-MET and FGFR inhibitors, epithelial cell adhesion molecule, IGF-1R inhibitors, mTOR pathway and MMP inhibitors, blockade of the tumour cell cycle, proteasomes, histone deacetylases, chaperone proteins, and promising immunotherapy.

Due to the molecular heterogeneity of tumours and the existence of multiple aberrant molecular pathways involved in gastric cancer development, the discovery of the exact molecular profile of the tumour and the detection of specific biomarkers and genes will help to identify the specific subsets of patients who might benefit from certain molecular therapies. This approach will open new horizons in gastric cancer treatment, allowing for personalized therapy using drugs directly targeting the particularly altered molecular pathways of the tumour.

Footnotes

Manuscript source: Invited manuscript

Specialty type: Gastroenterology and hepatology

Country of origin: Romania

Peer-review report classification

Grade A (Excellent): 0

Grade B (Very good): B

Grade C (Good): C

Grade D (Fair): 0

Grade E (Poor): 0

P- Reviewer: Chen MJ, Cho JY S- Editor: Yu J L- Editor: A E- Editor: Ma S

References
1.  Ferro A, Peleteiro B, Malvezzi M, Bosetti C, Bertuccio P, Levi F, Negri E, La Vecchia C, Lunet N. Worldwide trends in gastric cancer mortality (1980-2011), with predictions to 2015, and incidence by subtype. Eur J Cancer. 2014;50:1330-1344.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 403]  [Cited by in F6Publishing: 474]  [Article Influence: 47.4]  [Reference Citation Analysis (0)]
2.  Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D. Global cancer statistics. CA Cancer J Clin. 2011;61:69-90.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23762]  [Cited by in F6Publishing: 25159]  [Article Influence: 1935.3]  [Reference Citation Analysis (3)]
3.  Globocan 2012: Estimated cancer incidence, mortality and prevalence worldwide in 2012.  Available from: http://globocan.iarc.fr./Pages/fact sheets cancer.aspx.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Crew KD, Neugut AI. Epidemiology of upper gastrointestinal malignancies. Semin Oncol. 2004;31:450-464.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 229]  [Cited by in F6Publishing: 247]  [Article Influence: 12.4]  [Reference Citation Analysis (0)]
5.  Kubo A, Corley DA. Marked regional variation in adenocarcinomas of the esophagus and the gastric cardia in the United States. Cancer. 2002;95:2096-2102.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 83]  [Cited by in F6Publishing: 83]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
6.  Parkin DM, Muir CS. Cancer Incidence in Five Continents. Comparability and quality of data. IARC Sci Publ. 1992;45-173.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Malfertheiner P, Bornschein J, Selgrad M. Role of Helicobacter pylori infection in gastric cancer pathogenesis: a chance for prevention. J Dig Dis. 2010;11:2-11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 22]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
8.  Polk DB, Peek RM. Helicobacter pylori: gastric cancer and beyond. Nat Rev Cancer. 2010;10:403-414.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 718]  [Cited by in F6Publishing: 770]  [Article Influence: 55.0]  [Reference Citation Analysis (0)]
9.  Brenner H, Arndt V, Stegmaier C, Ziegler H, Rothenbacher D. Is Helicobacter pylori infection a necessary condition for noncardia gastric cancer? Am J Epidemiol. 2004;159:252-258.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 129]  [Cited by in F6Publishing: 142]  [Article Influence: 7.1]  [Reference Citation Analysis (0)]
10.  Bornschein J, Selgrad M, Warnecke M, Kuester D, Wex T, Malfertheiner P. H. pylori infection is a key risk factor for proximal gastric cancer. Dig Dis Sci. 2010;55:3124-3131.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 59]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
11.  Take S, Mizuno M, Ishiki K, Nagahara Y, Yoshida T, Yokota K, Oguma K, Okada H, Shiratori Y. The effect of eradicating helicobacter pylori on the development of gastric cancer in patients with peptic ulcer disease. Am J Gastroenterol. 2005;100:1037-1042.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 149]  [Cited by in F6Publishing: 159]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
12.  Mera R, Fontham ET, Bravo LE, Bravo JC, Piazuelo MB, Camargo MC, Correa P. Long term follow up of patients treated for Helicobacter pylori infection. Gut. 2005;54:1536-1540.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 239]  [Cited by in F6Publishing: 259]  [Article Influence: 13.6]  [Reference Citation Analysis (0)]
13.  You WC, Brown LM, Zhang L, Li JY, Jin ML, Chang YS, Ma JL, Pan KF, Liu WD, Hu Y. Randomized double-blind factorial trial of three treatments to reduce the prevalence of precancerous gastric lesions. J Natl Cancer Inst. 2006;98:974-983.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 310]  [Cited by in F6Publishing: 297]  [Article Influence: 16.5]  [Reference Citation Analysis (0)]
14.  Fuccio L, Zagari RM, Eusebi LH, Laterza L, Cennamo V, Ceroni L, Grilli D, Bazzoli F. Meta-analysis: can Helicobacter pylori eradication treatment reduce the risk for gastric cancer? Ann Intern Med. 2009;151:121-128.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 246]  [Cited by in F6Publishing: 279]  [Article Influence: 18.6]  [Reference Citation Analysis (0)]
15.  de Vries AC, Meijer GA, Looman CW, Casparie MK, Hansen BE, van Grieken NC, Kuipers EJ. Epidemiological trends of pre-malignant gastric lesions: a long-term nationwide study in the Netherlands. Gut. 2007;56:1665-1670.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 62]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
16.  Lee YC, Chen TH, Chiu HM, Shun CT, Chiang H, Liu TY, Wu MS, Lin JT. The benefit of mass eradication of Helicobacter pylori infection: a community-based study of gastric cancer prevention. Gut. 2013;62:676-682.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 254]  [Cited by in F6Publishing: 269]  [Article Influence: 24.5]  [Reference Citation Analysis (0)]
17.  Correa P. Is gastric cancer preventable? Gut. 2004;53:1217-1219.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 69]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
18.  Cervantes A, Roda D, Tarazona N, Roselló S, Pérez-Fidalgo JA. Current questions for the treatment of advanced gastric cancer. Cancer Treat Rev. 2013;39:60-67.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 118]  [Cited by in F6Publishing: 114]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
19.  Siegel R, Ma J, Zou Z, Jemal A. Cancer statistics, 2014. CA Cancer J Clin. 2014;64:9-29.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8789]  [Cited by in F6Publishing: 9407]  [Article Influence: 940.7]  [Reference Citation Analysis (0)]
20.  Jemal A, Siegel R, Ward E, Hao Y, Xu J, Thun MJ. Cancer statistics, 2009. CA Cancer J Clin. 2009;59:225-249.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Biondi A, Hyung WJ. Seventh edition of TNM classification for gastric cancer. J Clin Oncol. 2011;29:4338-4339; author reply 4340-4342.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Washington K. 7th edition of the AJCC cancer staging manual: stomach. Ann Surg Oncol. 2010;17:3077-3079.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 702]  [Cited by in F6Publishing: 796]  [Article Influence: 61.2]  [Reference Citation Analysis (0)]
23.  Sasako M, Sakuramoto S, Katai H, Kinoshita T, Furukawa H, Yamaguchi T, Nashimoto A, Fujii M, Nakajima T, Ohashi Y. Five-year outcomes of a randomized phase III trial comparing adjuvant chemotherapy with S-1 versus surgery alone in stage II or III gastric cancer. J Clin Oncol. 2011;29:4387-4393.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 869]  [Cited by in F6Publishing: 997]  [Article Influence: 76.7]  [Reference Citation Analysis (0)]
24.  Bang YJ, Kim YW, Yang HK, Chung HC, Park YK, Lee KH, Lee KW, Kim YH, Noh SI, Cho JY. Adjuvant capecitabine and oxaliplatin for gastric cancer after D2 gastrectomy (CLASSIC): a phase 3 open-label, randomised controlled trial. Lancet. 2012;379:315-321.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1055]  [Cited by in F6Publishing: 1171]  [Article Influence: 97.6]  [Reference Citation Analysis (0)]
25.  Ajani JA, Moiseyenko VM, Tjulandin S, Majlis A, Constenla M, Boni C, Rodrigues A, Fodor M, Chao Y, Voznyi E. Quality of life with docetaxel plus cisplatin and fluorouracil compared with cisplatin and fluorouracil from a phase III trial for advanced gastric or gastroesophageal adenocarcinoma: the V-325 Study Group. J Clin Oncol. 2007;25:3210-3216.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 188]  [Cited by in F6Publishing: 208]  [Article Influence: 12.2]  [Reference Citation Analysis (0)]
26.  Kothari N, Almhanna K. Current status of novel agents in advanced gastroesophageal adenocarcinoma. J Gastrointest Oncol. 2015;6:60-74.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 12]  [Reference Citation Analysis (0)]
27.  Coussens L, Yang-Feng TL, Liao YC, Chen E, Gray A, McGrath J, Seeburg PH, Libermann TA, Schlessinger J, Francke U. Tyrosine kinase receptor with extensive homology to EGF receptor shares chromosomal location with neu oncogene. Science. 1985;230:1132-1139.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1199]  [Cited by in F6Publishing: 1217]  [Article Influence: 31.2]  [Reference Citation Analysis (0)]
28.  Stintzing S, Jung A, Rossius L, Modest DP, von Weikersthal LF, Decker T, Kiani A, Al-Batran SE, Vehling-Kaiser U, Heintges T. Mutations within the EGFR signaling pathway: Influence on efficacy in FIRE-3-A randomized phase III study of FOLFIRI plus cetuximab or bevacizumab as first-line treatment for wild-type (WT) KRAS (exon 2) metastatic colorectal cancer (mCRC) patients. J Clin Oncol. 2014;32 Suppl 3:abstr 445.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Matsui Y, Inomata M, Tojigamori M, Sonoda K, Shiraishi N, Kitano S. Suppression of tumor growth in human gastric cancer with HER2 overexpression by an anti-HER2 antibody in a murine model. Int J Oncol. 2005;27:681-685.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 3]  [Reference Citation Analysis (0)]
30.  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: 4813]  [Article Influence: 343.8]  [Reference Citation Analysis (0)]
31.  Clinical trials.gov. A study of trastuzumab emtansine versus taxane in patients with advanced gastric cancer.  Available from: http://clinicaltrials.gov/show/NCT01641939.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Clinical trials.gov. A study of capecitabine [xeloda] in combination with trastuzumab [herceptin] and oxaliplatine in patients with resectable gastric cancer.  Available from: http://clinicaltrials.gov/show/NCT01130337.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Clinical trials.gov. A study of the combination of oxaliplatin, capecitabine and herceptin (trastuzumab) and chemoradiotherapy in the adjuvant setting in operated patients with HER2 gastric or gastro-esophageal junction cancer (TOXAG study).  Available from: http://www.clinicaltrials.gov/show/NCT01748773.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Clinical trials.gov. Explorative phase II study of perioperative treatment in patients with adenocarcinoma of the gastroesophageal junction or stomach (HerFLOT).  Available from: http://www.clinicaltrials.gov/ct2/show/NCT01472029.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Clinical trials.gov. Radiation therapy, paclitaxel, and carboplatin with or without trastuzumab in treating patients with esophageal cancer.  Available from: http://clinicaltrials.gov/show/NCT01196390.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Ryu MH, Yoo C, Kim JG, Ryoo BY, Park YS, Park SR, Han HS, Chung IJ, Song EK, Lee KH. Multicenter phase II study of trastuzumab in combination with capecitabine and oxaliplatin for advanced gastric cancer. Eur J Cancer. 2015;51:482-488.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 81]  [Cited by in F6Publishing: 90]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
37.  Kurokawa Y, Sugimoto N, Miwa H, Tsuda M, Nishina S, Okuda H, Imamura H, Gamoh M, Sakai D, Shimokawa T. Phase II study of trastuzumab in combination with S-1 plus cisplatin in HER2-positive gastric cancer (HERBIS-1). Br J Cancer. 2014;110:1163-1168.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 97]  [Cited by in F6Publishing: 103]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
38.  Hoff P, Tabernero J, Shen L. P-0111 Pertuzumab, trastuzumab and chemotherapy in HER2-positive metastatic gastric or gastro-oesophageal junction cancer: an international phase III study (JACOB). Ann Oncol. 2013;24:iv67.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 10]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
39.  Clinical trials.gov. A study of trastuzumab emtansine versus taxane in patients with advanced gastric cancer.  Available from: http://clinicaltrials.gov/show/NCT01641939..  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Cameron D, Casey M, Press M, Lindquist D, Pienkowski T, Romieu CG, Chan S, Jagiello-Gruszfeld A, Kaufman B, Crown J. A phase III randomized comparison of lapatinib plus capecitabine versus capecitabine alone in women with advanced breast cancer that has progressed on trastuzumab: updated efficacy and biomarker analyses. Breast Cancer Res Treat. 2008;112:533-543.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 584]  [Cited by in F6Publishing: 558]  [Article Influence: 34.9]  [Reference Citation Analysis (0)]
41.  Accessdata.fda.gov. Administration US FDA. Lapatinib.  Available from: http://www.accessdata.fda.gov/drugsatfda_docs/label/2013/022059s016s017lbl.pdf 21-23.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Hecht JR, Bang YJ, Qin SK, Chung HC, Xu JM, Park JO, Jeziorski K, Shparyk Y, Hoff PM, Sobrero A. Lapatinib in Combination With Capecitabine Plus Oxaliplatin in Human Epidermal Growth Factor Receptor 2-Positive Advanced or Metastatic Gastric, Esophageal, or Gastroesophageal Adenocarcinoma: TRIO-013/LOGiC--A Randomized Phase III Trial. J Clin Oncol. 2016;34:443-451.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 343]  [Cited by in F6Publishing: 397]  [Article Influence: 49.6]  [Reference Citation Analysis (0)]
43.  Satoh T, Xu RH, Chung HC, Sun GP, Doi T, Xu JM, Tsuji A, Omuro Y, Li J, Wang JW. Lapatinib plus paclitaxel versus paclitaxel alone in the second-line treatment of HER2-amplified advanced gastric cancer in Asian populations: TyTAN--a randomized, phase III study. J Clin Oncol. 2014;32:2039-2049.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 437]  [Cited by in F6Publishing: 449]  [Article Influence: 44.9]  [Reference Citation Analysis (0)]
44.  Clinical trials.gov. Cunningham D. Chemotherapy With or Without Bevacizumab or Lapatinib to Treat Operable Oesophagogastric Cancer (ST03).  Available from: https://clinicaltrials.gov/ct2/show/NCT00450203.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Herbst RS. Review of epidermal growth factor receptor biology. Int J Radiat Oncol Biol Phys. 2004;59:21-26.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 825]  [Cited by in F6Publishing: 887]  [Article Influence: 44.4]  [Reference Citation Analysis (0)]
46.  Oda K, Matsuoka Y, Funahashi A, Kitano H. A comprehensive pathway map of epidermal growth factor receptor signaling. Mol Syst Biol. 2005;1:2005.0010.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 617]  [Cited by in F6Publishing: 695]  [Article Influence: 36.6]  [Reference Citation Analysis (0)]
47.  Lieto E, Ferraraccio F, Orditura M, Castellano P, Mura AL, Pinto M, Zamboli A, De Vita F, Galizia G. Expression of vascular endothelial growth factor (VEGF) and epidermal growth factor receptor (EGFR) is an independent prognostic indicator of worse outcome in gastric cancer patients. Ann Surg Oncol. 2008;15:69-79.  [PubMed]  [DOI]  [Cited in This Article: ]
48.  van Grieken NC, Aoyama T, Chambers PA, Bottomley D, Ward LC, Inam I, Buffart TE, Das K, Lim T, Pang B. KRAS and BRAF mutations are rare and related to DNA mismatch repair deficiency in gastric cancer from the East and the West: results from a large international multicentre study. Br J Cancer. 2013;108:1495-1501.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 71]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
49.  Martinelli E, De Palma R, Orditura M, De Vita F, Ciardiello F. Anti-epidermal growth factor receptor monoclonal antibodies in cancer therapy. Clin Exp Immunol. 2009;158:1-9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 242]  [Cited by in F6Publishing: 219]  [Article Influence: 14.6]  [Reference Citation Analysis (0)]
50.  Saltz LB, Lenz HJ, Kindler HL, Hochster HS, Wadler S, Hoff PM, Kemeny NE, Hollywood EM, Gonen M, Quinones M. Randomized phase II trial of cetuximab, bevacizumab, and irinotecan compared with cetuximab and bevacizumab alone in irinotecan-refractory colorectal cancer: the BOND-2 study. J Clin Oncol. 2007;25:4557-4561.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 300]  [Cited by in F6Publishing: 301]  [Article Influence: 17.7]  [Reference Citation Analysis (0)]
51.  Vermorken JB, Mesia R, Rivera F, Remenar E, Kawecki A, Rottey S, Erfan J, Zabolotnyy D, Kienzer HR, Cupissol D. Platinum-based chemotherapy plus cetuximab in head and neck cancer. N Engl J Med. 2008;359:1116-1127.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2546]  [Cited by in F6Publishing: 2356]  [Article Influence: 147.3]  [Reference Citation Analysis (0)]
52.  Lordick F, Kang YK, Chung HC, Salman P, Oh SC, Bodoky G, Kurteva G, Volovat C, Moiseyenko VM, Gorbunova V. Capecitabine and cisplatin with or without cetuximab for patients with previously untreated advanced gastric cancer (EXPAND): a randomised, open-label phase 3 trial. Lancet Oncol. 2013;14:490-499.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 589]  [Cited by in F6Publishing: 640]  [Article Influence: 58.2]  [Reference Citation Analysis (0)]
53.  Clinical trials.gov. Use of Cetuximab for Unresectable or Metastatic Esophageal and Gastric Cancer.  Available from: https://www.clinicaltrials.gov/ct2/show/NCT00130689.  [PubMed]  [DOI]  [Cited in This Article: ]
54.  Clinical trials.gov. Cetuximab, Cisplatin, and Irinotecan in Treating Patients With Metastatic Esophageal Cancer, Gastroesophageal Junction Cancer, or Gastric Cancer That Did Not Respond to Previous Irinotecan and Cisplatin.  Available from: https://www.clinicaltrials.gov/ct2/show/NCT00397904.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Clinical trials.gov. Pre-operation Chemo and Antibody Therapy Followed by Surgical Resection and Adjuvant Chemoradiation for Gastric Cancer.  Available from: https://www.clinicaltrials.gov/ct2/show/NCT00857246.  [PubMed]  [DOI]  [Cited in This Article: ]
56.  Clinical trials.gov. Erlotinib Hydrochloride and Cetuximab in Treating Patients With Advanced Gastrointestinal Cancer, Head and Neck Cancer, Non-Small Cell Lung Cancer, or Colorectal Cancer.  Available from: https://www.clinicaltrials.gov/ct2/show/study/NCT00397384.  [PubMed]  [DOI]  [Cited in This Article: ]
57.  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)]
58.  Tebbutt NC, Parry MM, Zannino D, Strickland AH, Van Hazel GA, Pavlakis N, Ganju V, Mellor D, Dobrovic A, Gebski VJ. Docetaxel plus cetuximab as second-line treatment for docetaxel-refractory oesophagogastric cancer: the AGITG ATTAX2 trial. Br J Cancer. 2013;108:771-774.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 16]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
59.  Kim C, Lee JL, Ryu MH, Chang HM, Kim TW, Lim HY, Kang HJ, Park YS, Ryoo BY, Kang YK. A prospective phase II study of cetuximab in combination with XELOX (capecitabine and oxaliplatin) in patients with metastatic and/or recurrent advanced gastric cancer. Invest New Drugs. 2011;29:366-373.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 66]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
60.  Waddell T, Chau I, Cunningham D, Gonzalez D, Okines AF, Okines C, Wotherspoon A, Saffery C, Middleton G, Wadsley J. Epirubicin, oxaliplatin, and capecitabine with or without panitumumab for patients with previously untreated advanced oesophagogastric cancer (REAL3): a randomised, open-label phase 3 trial. Lancet Oncol. 2013;14:481-489.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 504]  [Cited by in F6Publishing: 554]  [Article Influence: 50.4]  [Reference Citation Analysis (0)]
61.  Clinical trials.gov. Perioperative Panitumumab and Epirubicin, Oxaliplatin and Xeloda (EOX) in Patients With Gastroesophageal Adenocarcinoma (EOXP).  Available from: https://www.clinicaltrials.gov/ct2/show/results/NCT00667420.  [PubMed]  [DOI]  [Cited in This Article: ]
62.  Clinical trials.gov. Panitumumab, Paclitaxel, Carboplatin and 5FU in the Treatment of Potentially Resectable Gastroesophageal Adenocarcinoma.  Available from: https://www.clinicaltrials.gov/ct2/show/NCT01182610.  [PubMed]  [DOI]  [Cited in This Article: ]
63.  Clinical trials.gov. ECX Panitumumab vs ECX Alone in Locally Advanced Gastric Cancer or Cancer of the Gastroesophageal Junction.  Available from: https://www.clinicaltrials.gov/ct2/show/NCT01234324.  [PubMed]  [DOI]  [Cited in This Article: ]
64.  Clinical trials.gov. MEGA (Met or EGFR Inhibition in Gastroesophageal Adenocarcinoma): FOLFOX Alone or in Combination With AMG 102 or Panitumumab as First-line Treatment in Patients With Advanced Gastroesophageal Adenocarcinoma.  Available from: https://www.clinicaltrials.gov/ct2/show/NCT01443065.  [PubMed]  [DOI]  [Cited in This Article: ]
65.  Xu W, Yang Z, Lu N. Molecular targeted therapy for the treatment of gastric cancer. J Exp Clin Cancer Res. 2016;35:1.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 93]  [Cited by in F6Publishing: 116]  [Article Influence: 14.5]  [Reference Citation Analysis (0)]
66.  Crombet T, Torres L, Neninger E, Catalá M, Solano ME, Perera A, Torres O, Iznaga N, Torres F, Pérez R. Pharmacological evaluation of humanized anti-epidermal growth factor receptor, monoclonal antibody h-R3, in patients with advanced epithelial-derived cancer. J Immunother. 2003;26:139-148.  [PubMed]  [DOI]  [Cited in This Article: ]
67.  Crombet-Ramos T, Rak J, Pérez R, Viloria-Petit A. Antiproliferative, antiangiogenic and proapoptotic activity of h-R3: A humanized anti-EGFR antibody. Int J Cancer. 2002;101:567-575.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 117]  [Cited by in F6Publishing: 126]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
68.  Li K, Li J. Current Molecular Targeted Therapy in Advanced Gastric Cancer: A Comprehensive Review of Therapeutic Mechanism, Clinical Trials, and Practical Application. Gastroenterol Res Pract. 2016;2016:4105615.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 25]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
69.  Satoh T, Lee KH, Rha SY, Sasaki Y, Park SH, Komatsu Y, Yasui H, Kim TY, Yamaguchi K, Fuse N. Randomized phase II trial of nimotuzumab plus irinotecan versus irinotecan alone as second-line therapy for patients with advanced gastric cancer. Gastric Cancer. 2015;18:824-832.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 78]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
70.  Clinical trials.gov. Phase 3 Study of Nimotuzumab and Irinotecan as Second Line With Advanced or Recurrect Gastric and Gastroesophageal Junction Cancer.  Available from: https://www.clinicaltrials.gov/ct2/show/NCT01813253.  [PubMed]  [DOI]  [Cited in This Article: ]
71.  Du F, Zheng Z, Shi S, Jiang Z, Qu T, Yuan X, Sun Y, Song Y, Yang L, Zhao J. S-1 and Cisplatin With or Without Nimotuzumab for Patients With Untreated Unresectable or Metastatic Gastric Cancer: A Randomized, Open-Label Phase 2 Trial. Medicine (Baltimore). 2015;94:e958.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 37]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
72.  Clinical trials.gov. Gefitinib in Combination With Chemoradiation in Resectable Gastric Cancer.  Available from: https://www.clinicaltrials.gov/ct2/show?term=gefitinib, gastric cancer&rank=2.  [PubMed]  [DOI]  [Cited in This Article: ]
73.  Clinical trials.gov. Cisplatin and Irinotecan Chemotherapy, Followed by ZD 1839 (Iressa) in Patients With Esophageal or Gastric Carcinomas.  Available from: https://www.clinicaltrials.gov/ct2/show/NCT00215995.  [PubMed]  [DOI]  [Cited in This Article: ]
74.  Clinical trials.gov. Gefitinib in treating pacients with esophageal cancer that is progressing after chemotherapy.  Available from: http://www.clinicaltrials.gov/ct2/ show/NCT01243398.  [PubMed]  [DOI]  [Cited in This Article: ]
75.  Dragovich T, McCoy S, Fenoglio-Preiser CM, Wang J, Benedetti JK, Baker AF, Hackett CB, Urba SG, Zaner KS, Blanke CD. Phase II trial of erlotinib in gastroesophageal junction and gastric adenocarcinomas: SWOG 0127. J Clin Oncol. 2006;24:4922-4927.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 238]  [Cited by in F6Publishing: 238]  [Article Influence: 13.2]  [Reference Citation Analysis (0)]
76.  Folkman J. Tumor angiogenesis: therapeutic implications. N Engl J Med. 1971;285:1182-1186.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5115]  [Cited by in F6Publishing: 5632]  [Article Influence: 106.3]  [Reference Citation Analysis (0)]
77.  Folkman J, Cole P, Zimmerman S. Tumor behavior in isolated perfused organs: in vitro growth and metastases of biopsy material in rabbit thyroid and canine intestinal segment. Ann Surg. 1966;164:491-502.  [PubMed]  [DOI]  [Cited in This Article: ]
78.  Folkman J. The vascularization of tumors. Sci Am. 1976;234:58-64, 70-3.  [PubMed]  [DOI]  [Cited in This Article: ]
79.  Folkman J. Anti-angiogenesis: new concept for therapy of solid tumors. Ann Surg. 1972;175:409-416.  [PubMed]  [DOI]  [Cited in This Article: ]
80.  Folkman J, Long DM, Becker FF. Growth and metastasis of tumor in organ culture. Cancer. 1963;16:453-467.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 3]  [Reference Citation Analysis (0)]
81.  Dvorak HF, Sioussat TM, Brown LF, Berse B, Nagy JA, Sotrel A, Manseau EJ, Van de Water L, Senger DR. Distribution of vascular permeability factor (vascular endothelial growth factor) in tumors: concentration in tumor blood vessels. J Exp Med. 1991;174:1275-1278.  [PubMed]  [DOI]  [Cited in This Article: ]
82.  Ferrara N, Houck K, Jakeman L, Leung DW. Molecular and biological properties of the vascular endothelial growth factor family of proteins. Endocr Rev. 1992;13:18-32.  [PubMed]  [DOI]  [Cited in This Article: ]
83.  Dvorak HF, Brown LF, Detmar M, Dvorak AM. Vascular permeability factor/vascular endothelial growth factor, microvascular hyperpermeability, and angiogenesis. Am J Pathol. 1995;146:1029-1039.  [PubMed]  [DOI]  [Cited in This Article: ]
84.  Ferrara N, Davis-Smyth T. The biology of vascular endothelial growth factor. Endocr Rev. 1997;18:4-25.  [PubMed]  [DOI]  [Cited in This Article: ]
85.  Peters KG, De Vries C, Williams LT. Vascular endothelial growth factor receptor expression during embryogenesis and tissue repair suggests a role in endothelial differentiation and blood vessel growth. Proc Natl Acad Sci USA. 1993;90:8915-8919.  [PubMed]  [DOI]  [Cited in This Article: ]
86.  Gerber HP, Hillan KJ, Ryan AM, Kowalski J, Keller GA, Rangell L, Wright BD, Radtke F, Aguet M, Ferrara N. VEGF is required for growth and survival in neonatal mice. Development. 1999;126:1149-1159.  [PubMed]  [DOI]  [Cited in This Article: ]
87.  Otani N, Minami S, Yamoto M, Shikone T, Otani H, Nishiyama R, Otani T, Nakano R. The vascular endothelial growth factor/fms-like tyrosine kinase system in human ovary during the menstrual cycle and early pregnancy. J Clin Endocrinol Metab. 1999;84:3845-3851.  [PubMed]  [DOI]  [Cited in This Article: ]
88.  Brown LF, Berse B, Jackman RW, Tognazzi K, Manseau EJ, Dvorak HF, Senger DR. Increased expression of vascular permeability factor (vascular endothelial growth factor) and its receptors in kidney and bladder carcinomas. Am J Pathol. 1993;143:1255-1262.  [PubMed]  [DOI]  [Cited in This Article: ]
89.  Inoue M, Hager JH, Ferrara N, Gerber HP, Hanahan D. VEGF-A has a critical, nonredundant role in angiogenic switching and pancreatic beta cell carcinogenesis. Cancer Cell. 2002;1:193-202.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 301]  [Cited by in F6Publishing: 314]  [Article Influence: 14.3]  [Reference Citation Analysis (0)]
90.  Gerber HP, Kowalski J, Sherman D, Eberhard DA, Ferrara N. Complete inhibition of rhabdomyosarcoma xenograft growth and neovascularization requires blockade of both tumor and host vascular endothelial growth factor. Cancer Res. 2000;60:6253-6258.  [PubMed]  [DOI]  [Cited in This Article: ]
91.  Ferrara N, Gerber HP, LeCouter J. The biology of VEGF and its receptors. Nat Med. 2003;9:669-676.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6747]  [Cited by in F6Publishing: 6654]  [Article Influence: 316.9]  [Reference Citation Analysis (0)]
92.  Kaipainen A, Korhonen J, Mustonen T, van Hinsbergh VW, Fang GH, Dumont D, Breitman M, Alitalo K. Expression of the fms-like tyrosine kinase 4 gene becomes restricted to lymphatic endothelium during development. Proc Natl Acad Sci USA. 1995;92:3566-3570.  [PubMed]  [DOI]  [Cited in This Article: ]
93.  Chang L, Kaipainen A, Folkman J. Lymphangiogenesis new mechanisms. Ann N Y Acad Sci. 2002;979:111-119.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 56]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
94.  Levy AP, Levy NS, Goldberg MA. Post-transcriptional regulation of vascular endothelial growth factor by hypoxia. J Biol Chem. 1996;271:2746-2753.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 485]  [Cited by in F6Publishing: 497]  [Article Influence: 17.8]  [Reference Citation Analysis (0)]
95.  Dor Y, Keshet E. Ischemia-driven angiogenesis. Trends Cardiovasc Med. 1997;7:289-294.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 54]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
96.  Ferrara N. Role of vascular endothelial growth factor in regulation of angiogenesis. Antiangiogenic agents in cancer therapy. New Jersey: Humana Press 1999; 119-142.  [PubMed]  [DOI]  [Cited in This Article: ]
97.  Roberts WG, Palade GE. Neovasculature induced by vascular endothelial growth factor is fenestrated. Cancer Res. 1997;57:765-772.  [PubMed]  [DOI]  [Cited in This Article: ]
98.  Carmeliet P. Angiogenesis in health and disease. Nat Med. 2003;9:653-660.  [PubMed]  [DOI]  [Cited in This Article: ]
99.  Grigore D, Simionescu CE, Stepan A, Mărgăritescu C, Bălăşoiu M, Georgescu CC, Cernea D, Dumitrescu D. Assessment of CD105, α-SMA and VEGF expression in gastric carcinomas. Rom J Morphol Embryol. 2013;54:701-707.  [PubMed]  [DOI]  [Cited in This Article: ]
100.  Hurwitz H, Fehrenbacher L, Novotny W, Cartwright T, Hainsworth J, Heim W, Berlin J, Baron A, Griffing S, Holmgren E. Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. N Engl J Med. 2004;350:2335-2342.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7832]  [Cited by in F6Publishing: 7513]  [Article Influence: 375.7]  [Reference Citation Analysis (1)]
101.  Miller K, Wang M, Gralow J, Dickler M, Cobleigh M, Perez EA, Shenkier T, Cella D, Davidson NE. Paclitaxel plus bevacizumab versus paclitaxel alone for metastatic breast cancer. N Engl J Med. 2007;357:2666-2676.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2393]  [Cited by in F6Publishing: 2249]  [Article Influence: 132.3]  [Reference Citation Analysis (0)]
102.  Sandler A, Gray R, Perry MC, Brahmer J, Schiller JH, Dowlati A, Lilenbaum R, Johnson DH. Paclitaxel-carboplatin alone or with bevacizumab for non-small-cell lung cancer. N Engl J Med. 2006;355:2542-2550.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4457]  [Cited by in F6Publishing: 4342]  [Article Influence: 241.2]  [Reference Citation Analysis (0)]
103.  Cannistra SA, Matulonis UA, Penson RT, Hambleton J, Dupont J, Mackey H, Douglas J, Burger RA, Armstrong D, Wenham R. Phase II study of bevacizumab in patients with platinum-resistant ovarian cancer or peritoneal serous cancer. J Clin Oncol. 2007;25:5180-5186.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 573]  [Cited by in F6Publishing: 548]  [Article Influence: 32.2]  [Reference Citation Analysis (0)]
104.  Escudier B, Pluzanska A, Koralewski P, Ravaud A, Bracarda S, Szczylik C, Chevreau C, Filipek M, Melichar B, Bajetta E. Bevacizumab plus interferon alfa-2a for treatment of metastatic renal cell carcinoma: a randomised, double-blind phase III trial. Lancet. 2007;370:2103-2111.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1757]  [Cited by in F6Publishing: 1684]  [Article Influence: 99.1]  [Reference Citation Analysis (0)]
105.  Takano S, Ishikawa E, Nakai K, Matsuda M, Masumoto T, Yamamoto T, Matsumura A. Bevacizumab in Japanese patients with malignant glioma: from basic research to clinical trial. Onco Targets Ther. 2014;7:1551-1562.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 11]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
106.  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)]
107.  Shah M, Kang Y, Ohtsu A. Tumor and blood plasma biomarker analyses in the AVAGAST phase III randomized study of first-line bevacizumab capecitabine/cisplatin in patients with advanced gastric cancer. Ann Oncol. 2010;20:abstr 174PD.  [PubMed]  [DOI]  [Cited in This Article: ]
108.  Van Cutsem E, de Haas S, Kang YK, Ohtsu A, Tebbutt NC, Ming Xu J, Peng Yong W, Langer B, Delmar P, Scherer SJ. Bevacizumab in combination with chemotherapy as first-line therapy in advanced gastric cancer: a biomarker evaluation from the AVAGAST randomized phase III trial. J Clin Oncol. 2012;30:2119-2127.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 355]  [Cited by in F6Publishing: 385]  [Article Influence: 32.1]  [Reference Citation Analysis (0)]
109.  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)]
110.  Cunningham D, Allum WH, Stenning SP, Thompson JN, Van de Velde CJ, Nicolson M, Scarffe JH, Lofts FJ, Falk SJ, Iveson TJ. Perioperative chemotherapy versus surgery alone for resectable gastroesophageal cancer. N Engl J Med. 2006;355:11-20.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4120]  [Cited by in F6Publishing: 4221]  [Article Influence: 234.5]  [Reference Citation Analysis (0)]
111.  Choi AH, Kim J, Chao J. Perioperative chemotherapy for resectable gastric cancer: MAGIC and beyond. World J Gastroenterol. 2015;21:7343-7348.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 61]  [Cited by in F6Publishing: 67]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
112.  Clinical trials.gov. Docetaxel, Cisplatin, Irinotecan and Bevacizumab (TPCA) in Metastatic Esophageal and Gastric Cancer.  Available from: https://clinicaltrials.gov/ct2/show/NCT00394433.  [PubMed]  [DOI]  [Cited in This Article: ]
113.  Clinical trials.gov. Efficacy of Docetaxel, Capecitabine, Cisplatin, and Bevacizumab in Patients With Unresectable Advanced Gastric Cancer.  Available from: https://clinicaltrials.gov/ct2/show/NCT01471470.  [PubMed]  [DOI]  [Cited in This Article: ]
114.  Clinical trials.gov. Cisplatin, Irinotecan and Bevacizumab (PCA) Versus Docetaxel, Cisplatin, Irinotecan and Bevacizumab (TPCA) in Metastatic Esophageal and Gastric Cancer.  Available from: https://clinicaltrials.gov/ct2/show/NCT00911820.  [PubMed]  [DOI]  [Cited in This Article: ]
115.  Clinical trials.gov. Docetaxel, Oxaliplatin, Capecitabine, Bevacizumab and Trastuzumab in Patients With Locally Advanced or Metastatic Gastric Cancer (B-DOCT).  Available from: https://clinicaltrials.gov/ct2/show/NCT01359397.  [PubMed]  [DOI]  [Cited in This Article: ]
116.  Clinical trials.gov. CAPOX, Bevacizumab and Trastuzumab for Patients With HER2-Positive Metastatic Esophagogastric Cancer.  Available from: https://clinicaltrials.gov/ct2/show/NCT01191697.  [PubMed]  [DOI]  [Cited in This Article: ]
117.  Clinical trials.gov. Bevacizumab and Combination Chemotherapy Before Surgery in Treating Patients With Locally Advanced Esophageal or Stomach Cancer.  Available from: https://clinicaltrials.gov/ct2/show/NCT01212822.  [PubMed]  [DOI]  [Cited in This Article: ]
118.  Clinical trials.gov. Pre-operative Chemotherapy Plus Bevacizumab With Early Salvage Therapy Based on PET Assessment of Response in Patients With Locally Advanced But Resectable Gastric and GEJ Adenocarcinoma.  Available from: https://clinicaltrials.gov/ct2/show/NCT00737438.  [PubMed]  [DOI]  [Cited in This Article: ]
119.  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: 1459]  [Article Influence: 145.9]  [Reference Citation Analysis (0)]
120.  Accessdata.fda.gov. Administration US FDA. Ramucirumab.  Available from: http://www.accessdata.fda.gov/drugsatfda_docs/label/2014/125477lbl.pdf.  [PubMed]  [DOI]  [Cited in This Article: ]
121.  Wilke H, Van Cutsem E, Oh SC. RAINBOW: A global, phase 3, randomized, double-blind study of ramucirumab plus paclitaxel versus placebo plus paclitaxel in the treatment of metastatic gastric adenocarcinoma following disease progression on first-line platinum- and fluoropyrimidinecontaining combination therapy: Results of a multiple Cox regression analysis adjusting for prognostic factors. J Clin Oncol. 2014;32:abstr 4076.  [PubMed]  [DOI]  [Cited in This Article: ]
122.  Casak SJ, Fashoyin-Aje I, Lemery SJ, Zhang L, Jin R, Li H, Zhao L, Zhao H, Zhang H, Chen H. FDA Approval Summary: Ramucirumab for Gastric Cancer. Clin Cancer Res. 2015;21:3372-3376.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 110]  [Cited by in F6Publishing: 396]  [Article Influence: 30.5]  [Reference Citation Analysis (0)]
123.  Yoon HH, Bendell JC, Braiteh FS. Ramucirumab (RAM) plus FOLFOX as front-line therapy (Rx) for advanced gastric or esophageal adenocarcinoma (GE-AC): Randomized, double-blind, multicenter phase 2 trial. J Clin Oncol. 2014;32:abstr 4004.  [PubMed]  [DOI]  [Cited in This Article: ]
124.  Ling Y, Yang Y, Lu N, You QD, Wang S, Gao Y, Chen Y, Guo QL. Endostar, a novel recombinant human endostatin, exerts antiangiogenic effect via blocking VEGF-induced tyrosine phosphorylation of KDR/Flk-1 of endothelial cells. Biochem Biophys Res Commun. 2007;361:79-84.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 159]  [Cited by in F6Publishing: 179]  [Article Influence: 10.5]  [Reference Citation Analysis (0)]
125.  Xu R, Ma N, Wang F, Ma L, Chen R, Chen R, Kebinu M, Ma L, Han Z, Ayixiamu M. Results of a randomized and controlled clinical trial evaluating the efficacy and safety of combination therapy with Endostar and S-1 combined with oxaliplatin in advanced gastric cancer. Onco Targets Ther. 2013;6:925-929.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 22]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
126.  Geng R, Li J. Apatinib for the treatment of gastric cancer. Expert Opin Pharmacother. 2015;16:117-122.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in F6Publishing: 74]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
127.  Li J, Qin S, Xu J, Guo W, Xiong J, Bai Y, Sun G, Yang Y, Wang L, Xu N. Apatinib for chemotherapy-refractory advanced metastatic gastric cancer: results from a randomized, placebo-controlled, parallel-arm, phase II trial. J Clin Oncol. 2013;31:3219-3225.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 316]  [Cited by in F6Publishing: 385]  [Article Influence: 35.0]  [Reference Citation Analysis (0)]
128.  Qin S, Li J, Xu J. Phase III study of apatinib in advanced gastric cancer: a randomized, double-blind, placebo-controlled trial. J Clin Oncol. 2014;32 Suppl 5:abstr 4003.  [PubMed]  [DOI]  [Cited in This Article: ]
129.  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: 651]  [Article Influence: 81.4]  [Reference Citation Analysis (0)]
130.  Fan M, Zhang J, Wang Z, Wang B, Zhang Q, Zheng C, Li T, Ni C, Wu Z, Shao Z. Phosphorylated VEGFR2 and hypertension: potential biomarkers to indicate VEGF-dependency of advanced breast cancer in anti-angiogenic therapy. Breast Cancer Res Treat. 2014;143:141-151.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 56]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
131.  Apatinib got CFDA approval.  Available from: http://www. inyaohui.com/news/201502/05/ 5059.html.  [PubMed]  [DOI]  [Cited in This Article: ]
132.  Sutent (sunitinib maleate) FDA Approval History.  Available from: http://www.drugs.com/history/sutent.html.  [PubMed]  [DOI]  [Cited in This Article: ]
133.  Bang YJ, Kang YK, Kang WK, Boku N, Chung HC, Chen JS, Doi T, Sun Y, Shen L, Qin S. Phase II study of sunitinib as second-line treatment for advanced gastric cancer. Invest New Drugs. 2011;29:1449-1458.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 137]  [Cited by in F6Publishing: 135]  [Article Influence: 9.6]  [Reference Citation Analysis (0)]
134.  Yi JH, Lee J, Lee J, Park SH, Park JO, Yim DS, Park YS, Lim HY, Kang WK. Randomised phase II trial of docetaxel and sunitinib in patients with metastatic gastric cancer who were previously treated with fluoropyrimidine and platinum. Br J Cancer. 2012;106:1469-1474.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 2]  [Reference Citation Analysis (0)]
135.  Gómez-Martín C, Salazar R, Montagut C, Gil-Martín M, Núñez JA, Puig M, Lin X, Khosravan R, Tursi JM, Lechuga MJ. A phase I, dose-finding study of sunitinib combined with cisplatin and 5-fluorouracil in patients with advanced gastric cancer. Invest New Drugs. 2013;31:390-398.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 13]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
136.  Lee KW, Park SR, Oh DY, Park YI, Khosravan R, Lin X, Lee SY, Roh EJ, Valota O, Lechuga MJ. Phase I study of sunitinib plus capecitabine/cisplatin or capecitabine/oxaliplatin in advanced gastric cancer. Invest New Drugs. 2013;31:1547-1558.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 19]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
137.  Llovet JM, Ricci S, Mazzaferro V, Hilgard P, Gane E, Blanc JF, de Oliveira AC, Santoro A, Raoul JL, Forner A. Sorafenib in advanced hepatocellular carcinoma. N Engl J Med. 2008;359:378-390.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9016]  [Cited by in F6Publishing: 9474]  [Article Influence: 592.1]  [Reference Citation Analysis (1)]
138.  Escudier B, Eisen T, Stadler WM, Szczylik C, Oudard S, Siebels M, Negrier S, Chevreau C, Solska E, Desai AA. Sorafenib in advanced clear-cell renal-cell carcinoma. N Engl J Med. 2007;356:125-134.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3788]  [Cited by in F6Publishing: 3689]  [Article Influence: 217.0]  [Reference Citation Analysis (0)]
139.  Yang S, Ngo VC, Lew GB, Chong LW, Lee SS, Ong WJ, Lam WL, Thng CH, Koong HN, Ong HS. AZD6244 (ARRY-142886) enhances the therapeutic efficacy of sorafenib in mouse models of gastric cancer. Mol Cancer Ther. 2009;8:2537-2545.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 26]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
140.  Clinical trials.gov. Phase I Study of Sorafenib With Folfox4 as First-line Treatment in Advanced/Metastatic Gastric Cancer.  Available from: https://clinicaltrials.gov/ct2/show/NCT02209441.  [PubMed]  [DOI]  [Cited in This Article: ]
141.  Clinical trials.gov. Sorafenib Gastric Cancer Asian Phase I Study.  Available from: https://clinicaltrials.gov/ct2/show/NCT00663741.  [PubMed]  [DOI]  [Cited in This Article: ]
142.  Clinical trials.gov. Sorafenib. ICORG 06-41, V4.  Available from: https://clinicaltrials.gov/ct2/show/NCT01158287.  [PubMed]  [DOI]  [Cited in This Article: ]
143.  Sun W, Powell M, O’Dwyer PJ, Catalano P, Ansari RH, Benson AB. Phase II study of sorafenib in combination with docetaxel and cisplatin in the treatment of metastatic or advanced gastric and gastroesophageal junction adenocarcinoma: ECOG 5203. J Clin Oncol. 2010;28:2947-2951.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 148]  [Cited by in F6Publishing: 177]  [Article Influence: 12.6]  [Reference Citation Analysis (0)]
144.  Clinical trials.gov. Sorafenib as a second line treatment in patients with advanced or metastatic gastric cancer.  Available from: http://www.clinicaltrials.gov/ct2/show/NCT00595985.  [PubMed]  [DOI]  [Cited in This Article: ]
145.  Martin-Richard M, Gallego R, Pericay C, Garcia Foncillas J, Queralt B, Casado E, Barriuso J, Iranzo V, Juez I, Visa L. Multicenter phase II study of oxaliplatin and sorafenib in advanced gastric adenocarcinoma after failure of cisplatin and fluoropyrimidine treatment. A GEMCAD study. Invest New Drugs. 2013;31:1573-1579.  [PubMed]  [DOI]  [Cited in This Article: ]
146.  Clinical trials.gov. Capecitabine and Cisplatin (XP) Sorafenib in Advanced Gastric Cancer (AGC) (XP Sorafenib).  Available from: https://clinicaltrials.gov/ct2/show/results/NCT00565370.  [PubMed]  [DOI]  [Cited in This Article: ]
147.  Sternberg CN, Davis ID, Mardiak J, Szczylik C, Lee E, Wagstaff J, Barrios CH, Salman P, Gladkov OA, Kavina A. Pazopanib in locally advanced or metastatic renal cell carcinoma: results of a randomized phase III trial. J Clin Oncol. 2010;28:1061-1068.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1822]  [Cited by in F6Publishing: 1853]  [Article Influence: 132.4]  [Reference Citation Analysis (0)]
148.  van der Graaf WT, Blay JY, Chawla SP, Kim DW, Bui-Nguyen B, Casali PG, Schöffski P, Aglietta M, Staddon AP, Beppu Y. Pazopanib for metastatic soft-tissue sarcoma (PALETTE): a randomised, double-blind, placebo-controlled phase 3 trial. Lancet. 2012;379:1879-1886.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1455]  [Cited by in F6Publishing: 1459]  [Article Influence: 121.6]  [Reference Citation Analysis (0)]
149.  Dieras V, Bachelot T, Campone M, Isambert N, Joly F, Le Tourneau C, Cassier PA, Bompas E, Fumoleau P, Noal S. Pazopanib (P) and cisplatin (CDDP) in patients with advanced solid tumors (PACIFIK): A UNICANCER phase I study. J Clin Oncol. 2014;32 suppl:abstr 2583.  [PubMed]  [DOI]  [Cited in This Article: ]
150.  Clinical trials.gov. Pazopanib and ARQ 197 for Advanced Solid Tumors.  Available from: https://www.clinicaltrials.gov/ct2/show/NCT01468922.  [PubMed]  [DOI]  [Cited in This Article: ]
151.  Clinical trials.gov. FLO/- pazopanib as first-line treatment in advanced gastric cancer (PaFLO).  Available from: http://clinicaltrials.gov/ct2/show/NCT01503372.  [PubMed]  [DOI]  [Cited in This Article: ]
152.  Clinical trials.gov. A study of pazopanib with CAPEOX in AGC patients.  Available from: http://clinicaltrials.gov/ct2/show/NCT01130805.  [PubMed]  [DOI]  [Cited in This Article: ]
153.  Demetri GD, Reichardt P, Kang YK, Blay JY, Rutkowski P, Gelderblom H, Hohenberger P, Leahy M, von Mehren M, Joensuu H. Efficacy and safety of regorafenib for advanced gastrointestinal stromal tumours after failure of imatinib and sunitinib (GRID): an international, multicentre, randomised, placebo-controlled, phase 3 trial. Lancet. 2013;381:295-302.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 942]  [Cited by in F6Publishing: 935]  [Article Influence: 85.0]  [Reference Citation Analysis (0)]
154.  Stivarga (Regorafenib) FDA approval history.  Available from: http://www.drugs.com/history/stivarga.html.  [PubMed]  [DOI]  [Cited in This Article: ]
155.  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: 30]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
156.  Pavlakis N, Sjoquist KM, Tsobanis E. INTEGRATE: a randomized phase II double-blind placebo-controlled study of regorafenib (REG) in refractory advanced esophagogastric cancer (AOGC)-a study by the Australasian Gastrointestinal Trials Group (AGITG): final overall and subgroup results. Ann Oncol. 2015;26 Suppl t4:119.  [PubMed]  [DOI]  [Cited in This Article: ]
157.  Clinical trials.gov. Regorafenib Second Line Treatment of Metastatic or Advanced Upper GI Cancers.  Available from: https://www.clinicaltrials.gov/ct2/show/NCT02241720.  [PubMed]  [DOI]  [Cited in This Article: ]
158.  Clinical trials.gov. FOLFOX Plus Regorafenib in Patients With Unresectable or Metastatic Esophagogastric Cancer.  Available from: https://www.clinicaltrials.gov/ct2/show/NCT01913639.  [PubMed]  [DOI]  [Cited in This Article: ]
159.  Clinical trials.gov. Regorafenib in Combination With Paclitaxel in Advanced Oesophagogastric Carcinoma (REPEAT).  Available from: https://www.clinicaltrials.gov/ct2/show/NCT02406170.  [PubMed]  [DOI]  [Cited in This Article: ]
160.  Foulstone E, Prince S, Zaccheo O, Burns JL, Harper J, Jacobs C, Church D, Hassan AB. Insulin-like growth factor ligands, receptors, and binding proteins in cancer. J Pathol. 2005;205:145-153.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 186]  [Cited by in F6Publishing: 189]  [Article Influence: 9.9]  [Reference Citation Analysis (0)]
161.  Matsubara J, Yamada Y, Nakajima TE, Kato K, Hamaguchi T, Shirao K, Shimada Y, Shimoda T. Clinical significance of insulin-like growth factor type 1 receptor and epidermal growth factor receptor in patients with advanced gastric cancer. Oncology. 2008;74:76-83.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 46]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
162.  Baserga R, Peruzzi F, Reiss K. The IGF-1 receptor in cancer biology. Int J Cancer. 2003;107:873-877.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 475]  [Cited by in F6Publishing: 496]  [Article Influence: 24.8]  [Reference Citation Analysis (0)]
163.  Attard G, Fong PC, Molife R. Phase I trial involving the pharmacodynamic (PD) study of circulating tumour cells, of CP-751,871 (C), a monoclonal antibody against the insulin-like growth factor 1 receptor (IGF-1R), with docetaxel (D) in patients (p) with advanced cancer. J Clin Oncol. 2006;24:abstr 3023.  [PubMed]  [DOI]  [Cited in This Article: ]
164.  Juergens H, Daw NC, Geoerger B, Ferrari S, Villarroel M, Aerts I, Whelan J, Dirksen U, Hixon ML, Yin D. Preliminary efficacy of the anti-insulin-like growth factor type 1 receptor antibody figitumumab in patients with refractory Ewing sarcoma. J Clin Oncol. 2011;29:4534-4540.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 161]  [Cited by in F6Publishing: 173]  [Article Influence: 13.3]  [Reference Citation Analysis (0)]
165.  Goto Y, Sekine I, Tanioka M, Shibata T, Tanai C, Asahina H, Nokihara H, Yamamoto N, Kunitoh H, Ohe Y. Figitumumab combined with carboplatin and paclitaxel in treatment-naïve Japanese patients with advanced non-small cell lung cancer. Invest New Drugs. 2012;30:1548-1556.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 19]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
166.  Langer CJ, Novello S, Park K, Krzakowski M, Karp DD, Mok T, Benner RJ, Scranton JR, Olszanski AJ, Jassem J. Randomized, phase III trial of first-line figitumumab in combination with paclitaxel and carboplatin versus paclitaxel and carboplatin alone in patients with advanced non-small-cell lung cancer. J Clin Oncol. 2014;32:2059-2066.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 104]  [Cited by in F6Publishing: 117]  [Article Influence: 11.7]  [Reference Citation Analysis (0)]
167.  Clinical trials.gov. Study Of CP-751,871 In Combination With Sunitinib In Patients With Advanced Solid Tumors.  Available from: https://www.clinicaltrials.gov/ct2/show/study/NCT00729833.  [PubMed]  [DOI]  [Cited in This Article: ]
168.  Clinical trials.gov. CP-751,871 In Combination With Docetaxel In Advance Non-hematologic Malignancies.  Available from: https://www.clinicaltrials.gov/ct2/show/NCT01653158.  [PubMed]  [DOI]  [Cited in This Article: ]
169.  Grose R, Dickson C. Fibroblast growth factor signaling in tumorigenesis. Cytokine Growth Factor Rev. 2005;16:179-186.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 296]  [Cited by in F6Publishing: 314]  [Article Influence: 16.5]  [Reference Citation Analysis (0)]
170.  Hattori Y, Itoh H, Uchino S, Hosokawa K, Ochiai A, Ino Y, Ishii H, Sakamoto H, Yamaguchi N, Yanagihara K. Immunohistochemical detection of K-sam protein in stomach cancer. Clin Cancer Res. 1996;2:1373-1381.  [PubMed]  [DOI]  [Cited in This Article: ]
171.  Takeda M, Arao T, Yokote H, Komatsu T, Yanagihara K, Sasaki H, Yamada Y, Tamura T, Fukuoka K, Kimura H. AZD2171 shows potent antitumor activity against gastric cancer over-expressing fibroblast growth factor receptor 2/keratinocyte growth factor receptor. Clin Cancer Res. 2007;13:3051-3057.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 97]  [Cited by in F6Publishing: 103]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
172.  Bang YJ, Van Cutsem E, Mansoor W. A randomized, open-label phase II study of AZD4547 (AZD) versus Paclitaxel (P) in previously treated patients with advanced gastric cancer (AGC) with Fibroblast Growth Factor Receptor 2 (FGFR2) polysomy or gene amplification (amp): SHINE study. J Clin Oncol. 2015;33 Suppl:abstr 4014.  [PubMed]  [DOI]  [Cited in This Article: ]
173.  Dienstmann R, Bahleda R, Adamo B, Rodon J, Varga A, Gazzah A, Platero S, Smit H, Perera T, Zhong B. Abstract CT325: First in human study of JNJ-42756493, a potent pan fibroblast growth factor receptor (FGFR) inhibitor in patients with advanced solid tumors. Proceedings: AACR Annual Meeting 2014; April 5-9, 2014; San Diego, CA. Cancer Res. 2014;74:CT325.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 15]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
174.  Nakamura K, Yashiro M, Matsuoka T, Tendo M, Shimizu T, Miwa A, Hirakawa K. A novel molecular targeting compound as K-samII/FGF-R2 phosphorylation inhibitor, Ki23057, for Scirrhous gastric cancer. Gastroenterology. 2006;131:1530-1541.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 61]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
175.  Qiu H, Yashiro M, Zhang X, Miwa A, Hirakawa K. A FGFR2 inhibitor, Ki23057, enhances the chemosensitivity of drug-resistant gastric cancer cells. Cancer Lett. 2011;307:47-52.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 30]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
176.  Trudel S, Li ZH, Wei E, Wiesmann M, Chang H, Chen C, Reece D, Heise C, Stewart AK. CHIR-258, a novel, multitargeted tyrosine kinase inhibitor for the potential treatment of t(4; 14) multiple myeloma. Blood. 2005;105:2941-2948.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 228]  [Cited by in F6Publishing: 215]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
177.  Huynh H, Chow PK, Tai WM, Choo SP, Chung AY, Ong HS, Soo KC, Ong R, Linnartz R, Shi MM. Dovitinib demonstrates antitumor and antimetastatic activities in xenograft models of hepatocellular carcinoma. J Hepatol. 2012;56:595-601.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 42]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
178.  Huynh H, Ngo VC, Fargnoli J, Ayers M, Soo KC, Koong HN, Thng CH, Ong HS, Chung A, Chow P. Brivanib alaninate, a dual inhibitor of vascular endothelial growth factor receptor and fibroblast growth factor receptor tyrosine kinases, induces growth inhibition in mouse models of human hepatocellular carcinoma. Clin Cancer Res. 2008;14:6146-6153.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 172]  [Cited by in F6Publishing: 175]  [Article Influence: 10.9]  [Reference Citation Analysis (0)]
179.  Cai ZW, Zhang Y, Borzilleri RM, Qian L, Barbosa S, Wei D, Zheng X, Wu L, Fan J, Shi Z. Discovery of brivanib alaninate ((S)-((R)-1-(4-(4-fluoro-2-methyl-1H-indol-5-yloxy)-5-methylpyrrolo[2,1-f][1,2,4]triazin-6-yloxy)propan-2-yl)2-aminopropanoate), a novel prodrug of dual vascular endothelial growth factor receptor-2 and fibroblast growth factor receptor-1 kinase inhibitor (BMS-540215). J Med Chem. 2008;51:1976-1980.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 106]  [Cited by in F6Publishing: 113]  [Article Influence: 7.1]  [Reference Citation Analysis (0)]
180.  Ayers M, Fargnoli J, Lewin A, Wu Q, Platero JS. Discovery and validation of biomarkers that respond to treatment with brivanib alaninate, a small-molecule VEGFR-2/FGFR-1 antagonist. Cancer Res. 2007;67:6899-6906.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 43]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
181.  Bhide RS, Cai ZW, Zhang YZ, Qian L, Wei D, Barbosa S, Lombardo LJ, Borzilleri RM, Zheng X, Wu LI. Discovery and preclinical studies of (R)-1-(4-(4-fluoro-2-methyl-1H-indol-5-yloxy)-5- methylpyrrolo[2,1-f][1,2,4]triazin-6-yloxy)propan- 2-ol (BMS-540215), an in vivo active potent VEGFR-2 inhibitor. J Med Chem. 2006;49:2143-2146.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 107]  [Cited by in F6Publishing: 107]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
182.  Finn RS, Kang YK, Mulcahy M, Polite BN, Lim HY, Walters I, Baudelet C, Manekas D, Park JW. Phase II, open-label study of brivanib as second-line therapy in patients with advanced hepatocellular carcinoma. Clin Cancer Res. 2012;18:2090-2098.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 112]  [Cited by in F6Publishing: 124]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
183.  Clinical trials.gov. Dovitinib for Gastric Cancer With FGFR2 Amplification.  Available from: https://www.clinicaltrials.gov/ct2/show/NCT01719549.  [PubMed]  [DOI]  [Cited in This Article: ]
184.  Clinical trials.gov. Open-Label, Dose-Escalation Study of INCB054828 in Subjects With Advanced Malignancies.  Available from: https://www.clinicaltrials.gov/ct2/show/NCT02393248.  [PubMed]  [DOI]  [Cited in This Article: ]
185.  Clinical trials.gov. Combination of Brivanib With 5-Fluorouracil/Leucovorin (5FU/LV) and5-Fluorouracil/Leucovorin/Irinotecan(FOLFIRI).  Available from: https://www.clinicaltrials.gov/ct2/show/NCT01046864.  [PubMed]  [DOI]  [Cited in This Article: ]
186.  Nakajima M, Sawada H, Yamada Y, Watanabe A, Tatsumi M, Yamashita J, Matsuda M, Sakaguchi T, Hirao T, Nakano H. The prognostic significance of amplification and overexpression of c-met and c-erb B-2 in human gastric carcinomas. Cancer. 1999;85:1894-1902.  [PubMed]  [DOI]  [Cited in This Article: ]
187.  Lordick F. Targeting the HGF/MET pathway in gastric cancer. Lancet Oncol. 2014;15:914-916.  [PubMed]  [DOI]  [Cited in This Article: ]
188.  Oliner KS, Tang R, Anderson A, Lan Y, Iveson T, Donehower RC, Jiang Y, Dubey S, Loh E. Evaluation of MET pathway biomarkers in a phase II study of rilotumumab (R, AMG 102) or placebo (P) in combination with epirubicin, cisplatin, and capecitabine (ECX) in patients (pts) with locally advanced or metastatic gastric (G) or esophagogastric junction (EGJ) cancer. J Clin Oncol. 2012;30:abstr 4005.  [PubMed]  [DOI]  [Cited in This Article: ]
189.  Iveson T, Donehower RC, Davidenko I, Tjulandin S, Deptala A, Harrison M, Nirni S, Lakshmaiah K, Thomas A, Jiang Y. Rilotumumab in combination with epirubicin, cisplatin, and capecitabine as first-line treatment for gastric or oesophagogastric junction adenocarcinoma: an open-label, dose de-escalation phase 1b study and a double-blind, randomised phase 2 study. Lancet Oncol. 2014;15:1007-1018.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 201]  [Cited by in F6Publishing: 212]  [Article Influence: 21.2]  [Reference Citation Analysis (0)]
190.  Cunningham D, Tebbutt NC, Davidenko I, Murad AM, AI-Batran SE, IIson DH, Tjulandin S, Gotovkin E, Karaszewska B, Bondarenko I. Phase III, randomized, double-blind, multicenter, placebo (P)-controlled trial of rilotumumab (R) plus epirubicin, cisplatin and capecitabine (ECX) as first-line therapy in patients (pts) with advanced MET-positive (pos) gastric or gastroesophageal junction (G/GEJ) cancer: RILOMET-1 study. J Clin Oncol. 2015;33 Suppl:abstr 4000.  [PubMed]  [DOI]  [Cited in This Article: ]
191.  Shah MA, Bang YJ, Lordicketal F. MET Gastric: 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 Suppl:abstr 4012.  [PubMed]  [DOI]  [Cited in This Article: ]
192.  Kwak F. Clinical activity observed in a phase I dose escalation trial of an oral c-met and ALK inhibitor. J Clin Oncol. 2009;27 Suppl:abstr 15.  [PubMed]  [DOI]  [Cited in This Article: ]
193.  Kataoka Y, Mukohara T, Tomioka H, Funakoshi Y, Kiyota N, Fujiwara Y, Yashiro M, Hirakawa K, Hirai M, Minami H. Foretinib (GSK1363089), a multi-kinase inhibitor of MET and VEGFRs, inhibits growth of gastric cancer cell lines by blocking inter-receptor tyrosine kinase networks. Invest New Drugs. 2012;30:1352-1360.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 83]  [Cited by in F6Publishing: 89]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
194.  Jhawer M, Kindler HL, Wainberg Z, Ford J, Kunz P, Tang L, McCallum S, Kallender H, Shah MA. Assessment of two dosing schedules of GSK1363089 (GSK089), a dual MET/VEGFR2 inhibitor, in metastatic gastric cancer (GC): Interim results of a multicenter phase II study. J Clin Oncol. 2009;27:abstr 4502.  [PubMed]  [DOI]  [Cited in This Article: ]
195.  Shah MA, Wainberg ZA, Catenacci DV, Hochster HS, Ford J, Kunz P, Lee FC, Kallender H, Cecchi F, Rabe DC. Phase II study evaluating 2 dosing schedules of oral foretinib (GSK1363089), cMET/VEGFR2 inhibitor, in patients with metastatic gastric cancer. PLoS One. 2013;8:e54014.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 131]  [Cited by in F6Publishing: 154]  [Article Influence: 14.0]  [Reference Citation Analysis (0)]
196.  Vivanco I, Sawyers CL. The phosphatidylinositol 3-Kinase AKT pathway in human cancer. Nat Rev Cancer. 2002;2:489-501.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4468]  [Cited by in F6Publishing: 4455]  [Article Influence: 202.5]  [Reference Citation Analysis (0)]
197.  Yap TA, Garrett MD, Walton MI, Raynaud F, de Bono JS, Workman P. Targeting the PI3K-AKT-mTOR pathway: progress, pitfalls, and promises. Curr Opin Pharmacol. 2008;8:393-412.  [PubMed]  [DOI]  [Cited in This Article: ]
198.  Yu HG, Ai YW, Yu LL, Zhou XD, Liu J, Li JH, Xu XM, Liu S, Chen J, Liu F. Phosphoinositide 3-kinase/Akt pathway plays an important role in chemoresistance of gastric cancer cells against etoposide and doxorubicin induced cell death. Int J Cancer. 2008;122:433-443.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 143]  [Cited by in F6Publishing: 161]  [Article Influence: 10.1]  [Reference Citation Analysis (0)]
199.  Al-Batran SE, Ducreux M, Ohtsu A. mTOR as a therapeutic target in patients with gastric cancer. Int J Cancer. 2012;130:491-496.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 74]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
200.  Alvarado Y, Mita MM, Vemulapalli S, Mahalingam D, Mita AC. Clinical activity of mammalian target of rapamycin inhibitors in solid tumors. Target Oncol. 2011;6:69-94.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 28]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
201.  Dabney R, Devine R, Sein N, George B. New agents in renal cell carcinoma. Target Oncol. 2014;9:183-193.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 18]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
202.  Yao JC, Shah MH, Ito T, Bohas CL, Wolin EM, Van Cutsem E, Hobday TJ, Okusaka T, Capdevila J, de Vries EG. Everolimus for advanced pancreatic neuroendocrine tumors. N Engl J Med. 2011;364:514-523.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2039]  [Cited by in F6Publishing: 1969]  [Article Influence: 151.5]  [Reference Citation Analysis (0)]
203.  Franz DN, Belousova E, Sparagana S, Bebin EM, Frost M, Kuperman R, Witt O, Kohrman MH, Flamini JR, Wu JY. Efficacy and safety of everolimus for subependymal giant cell astrocytomas associated with tuberous sclerosis complex (EXIST-1): a multicentre, randomised, placebo-controlled phase 3 trial. Lancet. 2013;381:125-132.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 571]  [Cited by in F6Publishing: 556]  [Article Influence: 50.5]  [Reference Citation Analysis (0)]
204.  Hart CD, Migliaccio I, Malorni L, Guarducci C, Biganzoli L, Di Leo A. Challenges in the management of advanced, ER-positive, HER2-negative breast cancer. Nat Rev Clin Oncol. 2015;12:541-552.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 85]  [Cited by in F6Publishing: 105]  [Article Influence: 11.7]  [Reference Citation Analysis (0)]
205.  Okamoto I, Doi T, Ohtsu A, Miyazaki M, Tsuya A, Kurei K, Kobayashi K, Nakagawa K. Phase I clinical and pharmacokinetic study of RAD001 (everolimus) administered daily to Japanese patients with advanced solid tumors. Jpn J Clin Oncol. 2010;40:17-23.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 60]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
206.  Doi T, Muro K, Boku N, Yamada Y, Nishina T, Takiuchi H, Komatsu Y, Hamamoto Y, Ohno N, Fujita Y. Multicenter phase II study of everolimus in patients with previously treated metastatic gastric cancer. J Clin Oncol. 2010;28:1904-1910.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 151]  [Cited by in F6Publishing: 173]  [Article Influence: 12.4]  [Reference Citation Analysis (0)]
207.  Van Cutsem E, Yeh KH, Bang YJ, Shen L, Ajani JA, Bai YX, Chung HC, Pan HM, Chin K, Muro K. Phase III trial of everolimus (EVE) in previously treated patients with advanced gastric cancer (AGC): GRANITE-1. J Clin Oncol. 2012;30:abstr LBA3.  [PubMed]  [DOI]  [Cited in This Article: ]
208.  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)]
209.  Clinical trials.gov. Vaccine Therapy With or Without Sirolimus in Treating Patients With NY-ESO-1 Expressing Solid Tumors.  Available from: https://www.clinicaltrials.gov/ct2/show/NCT01522820.  [PubMed]  [DOI]  [Cited in This Article: ]
210.  Underhill C, Toulmonde M, Bonnefoi H. A review of PARP inhibitors: from bench to bedside. Ann Oncol. 2011;22:268-279.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 154]  [Cited by in F6Publishing: 162]  [Article Influence: 11.6]  [Reference Citation Analysis (0)]
211.  Bang YJ, Im SA, Lee KW, Cho JY, Song EK, Lee KH, Kim YH, Park JO, Chun HG, Zhang DY. Olaparib plus paclitaxel in patients with recurrent or metastatic gastric cancer: A randomized, double-blind phase II study. J Clin Oncol. 2013;31:abstr 4013.  [PubMed]  [DOI]  [Cited in This Article: ]
212.  Bang YJ, Im SA, Lee KW, Cho JY, Song EK, Lee KH, Kim YH, Park JO, Chun HG, Zang DY. Randomized, Double-Blind Phase II Trial With Prospective Classification by ATM Protein Level to Evaluate the Efficacy and Tolerability of Olaparib Plus Paclitaxel in Patients With Recurrent or Metastatic Gastric Cancer. J Clin Oncol. 2015;33:3858-3865.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 171]  [Cited by in F6Publishing: 213]  [Article Influence: 23.7]  [Reference Citation Analysis (0)]
213.  Kubota E, Williamson CT, Ye R, Elegbede A, Peterson L, Lees-Miller SP, Bebb DG. Low ATM protein expression and depletion of p53 correlates with olaparib sensitivity in gastric cancer cell lines. Cell Cycle. 2014;13:2129-2137.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 96]  [Cited by in F6Publishing: 104]  [Article Influence: 10.4]  [Reference Citation Analysis (0)]
214.  Clinical trials.gov. Efficacy and Safety Study of Olaparib in Combination With Paclitaxel to Treat Advanced Gastric Cancer.  Available from: https://www.clinicaltrials.gov/ct2/show/NCT01924533.  [PubMed]  [DOI]  [Cited in This Article: ]
215.  Clinical trials.gov. Ascending Doses of AZD6738 in Combination With Chemotherapy and/or Novel Anti Cancer Agents.  Available from: https://clinicaltrials.gov/ct2/show/NCT02264678.  [PubMed]  [DOI]  [Cited in This Article: ]
216.  Clinical trials.gov. Evaluating the Safety and Tolerability of the Poly-ADP Ribose (PARP) Inhibitor With FOLFIRI in Subjects With Solid Tumor.  Available from: https://www.clinicaltrials.gov/ct2/show/NCT01123876.  [PubMed]  [DOI]  [Cited in This Article: ]
217.  Kanagavel D, Fedyanin M, Tryakin A, Tjulandin S. Second-line treatment of metastatic gastric cancer: Current options and future directions. World J Gastroenterol. 2015;21:11621-11635.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 27]  [Cited by in F6Publishing: 27]  [Article Influence: 3.0]  [Reference Citation Analysis (3)]
218.  Hamid O, Robert C, Daud A, Hodi FS, Hwu WJ, Kefford R, Wolchok JD, Hersey P, Joseph RW, Weber JS. Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. N Engl J Med. 2013;369:134-144.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2597]  [Cited by in F6Publishing: 2629]  [Article Influence: 239.0]  [Reference Citation Analysis (0)]
219.  Keir ME, Liang SC, Guleria I, Latchman YE, Qipo A, Albacker LA, Koulmanda M, Freeman GJ, Sayegh MH, Sharpe AH. Tissue expression of PD-L1 mediates peripheral T cell tolerance. J Exp Med. 2006;203:883-895.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 839]  [Cited by in F6Publishing: 923]  [Article Influence: 51.3]  [Reference Citation Analysis (0)]
220.  Culver ME, Gatesman ML, Mancl EE, Lowe DK. Ipilimumab: a novel treatment for metastatic melanoma. Ann Pharmacother. 2011;45:510-519.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 38]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
221.  Clinical trials.gov. Phase II Study for Previously Untreated Subjects With Non Small Cell Lung Cancer (NSCLC) or Small Cell Lung Cancer (SCLC).  Available from: https://www.clinicaltrials.gov/show/NCT00527735.  [PubMed]  [DOI]  [Cited in This Article: ]
222.  Clinical trials.gov. First-Line Gemcitabine, Cisplatin Ipilimumab for Metastatic Urothelial Carcinoma.  Available from: https://www.clinicaltrials.gov/show/NCT01524991.  [PubMed]  [DOI]  [Cited in This Article: ]
223.  Clinical trials.gov. Study of MDX-010 in Patients With Metastatic Hormone-Refractory Prostate Cancer.  Available from: https://www.clinicaltrials.gov/show/NCT00323882.  [PubMed]  [DOI]  [Cited in This Article: ]
224.  Clinical trials.gov. An Efficacy Study in Gastric and Gastroesophageal Junction Cancer Comparing Ipilimumab Versus Standard of Care Immediately Following First Line Chemotherapy.  Available from: https://www.clinicaltrials.gov/ct2/show/NCT01585987.  [PubMed]  [DOI]  [Cited in This Article: ]
225.  Brahmer JR, Tykodi SS, Chow LQ, Hwu WJ, Topalian SL, Hwu P, Drake CG, Camacho LH, Kauh J, Odunsi K. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med. 2012;366:2455-2465.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5599]  [Cited by in F6Publishing: 5869]  [Article Influence: 489.1]  [Reference Citation Analysis (0)]
226.  Clinical trials.gov. Multiple Ascending Dose (MDX1105-01) (Anti-PDL1).  Available from: https://clinicaltrials.gov/show/NCT00729664.  [PubMed]  [DOI]  [Cited in This Article: ]
227.  Clinical trials.gov. A Phase 1/2, Open-label Study of Nivolumab Monotherapy or Nivolumab Combined With Ipilimumab in Subjects With Advanced or Metastatic Solid Tumors.  Available from: https://www.clinicaltrials.gov/ct2/show/NCT01928394.  [PubMed]  [DOI]  [Cited in This Article: ]
228.  Le DT, Bendell JC, Calvo E, Kim JW, Ascierto PA, Sharma P, Ott PA, Bono P, Jaeger D, Evans TRJ. Safety and activity of nivolumab monotherapy in advanced and metastatic (A/M) gastric or gastroesophageal junction cancer (GC/GEC): Results from the CheckMate-032 study. 2016 Gastrointestinal Cancers Symposium. J Clin Oncol. 2016;34 Suppl 4S:abstr 6.  [PubMed]  [DOI]  [Cited in This Article: ]
229.  Clinical trials.gov. Study of ONO-4538 in Unresectable Advanced or Recurrent Gastric Cancer.  Available from: https://www.clinicaltrials.gov/ct2/show/NCT02267343.  [PubMed]  [DOI]  [Cited in This Article: ]
230.  Muro K, Bang Y, Shankaran V. LBA15 A phase 1b study of pembrolizumab (PEMBRO; MK-3475) in patients (PTS) with advanced gastric cancer. Ann Oncol. 2014;25:1-41.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 38]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
231.  Clinical trials.gov. Study of Pembrolizumab (MK-3475) as First-Line Monotherapy and Combination Therapy for Treatment of Advanced Gastric or Gastroesophageal Junction Adenocarcinoma (MK-3475-062/KEYNOTE-062).  Available from: https://clinicaltrials.gov/ct2/show/NCT02494583.  [PubMed]  [DOI]  [Cited in This Article: ]
232.  Clinical trials.gov. A Study of Pembrolizumab (MK-3475) in Participants With Recurrent or Metastatic Gastric or Gastroesophageal Junction Adenocarcinoma (MK-3475-059/KEYNOTE-059).  Available from: https://clinicaltrials.gov/ct2/show/NCT02335411.  [PubMed]  [DOI]  [Cited in This Article: ]
233.  Clinical trials.gov. Study of Pembrolizumab in Subjects With Advanced Gastric or Gastroesophageal Junction Adenocarcinoma Who Progressed After First-Line Therapy With Platinum and Fluoropyrimidine: Integration of Molecular Subtypes Through Integrative Genomic Analysis.  Available from: https://clinicaltrials.gov/ct2/show/NCT02589496.  [PubMed]  [DOI]  [Cited in This Article: ]
234.  Clinical trials.gov. Combination Margetuximab and Pembrolizumab for Advanced, Metastatic HER2(+) Gastric or Gastroesophageal Junction Cancer.  Available from: https://clinicaltrials.gov/ct2/show/NCT02689284.  [PubMed]  [DOI]  [Cited in This Article: ]
235.  Clinical trials.gov. Pembrolizumab and Monoclonal Antibody Therapy in Advanced Cancer (PembroMab).  Available from: https://clinicaltrials.gov/ct2/show/NCT02318901.  [PubMed]  [DOI]  [Cited in This Article: ]
236.  Clinical trials.gov. A Study of Ramucirumab Plus Pembrolizumab in Participants With Gastric or GEJ Adenocarcinoma, NSCLC or Transitional Cell Carcinoma of the Urothelium.  Available from: https://clinicaltrials.gov/ct2/show/NCT02443324.  [PubMed]  [DOI]  [Cited in This Article: ]
237.  Clinical trials.gov. A Combination Clinical Study of PLX3397 and Pembrolizumab To Treat Advanced Melanoma and Other Solid Tumors.  Available from: https://clinicaltrials.gov/ct2/show/NCT02452424.  [PubMed]  [DOI]  [Cited in This Article: ]
238.  Clinical trials.gov. Phase 1b Open-Label Study of PEGPH20 With Pembrolizumab.  Available from: https://clinicaltrials.gov/ct2/show/NCT02563548.  [PubMed]  [DOI]  [Cited in This Article: ]
239.  Lutzky J, Antonia SJ, Blake-Haskins A, Li X, Robbins PB, Shalabi AM, Vasselli J, Ibrahim RA, Khleif S, Segal NH. A phase 1 study of MEDI4736, an anti–PD-L1 antibody, in patients with advanced solid tumors. J Clin Oncol. 2014;32:abstr 3001.  [PubMed]  [DOI]  [Cited in This Article: ]
240.  Clinical trials.gov. Durvalumab and Tremelimumab in Combination With First-Line Chemotherapy in Advanced Solid Tumors.  Available from: https://clinicaltrials.gov/ct2/show/NCT02658214.  [PubMed]  [DOI]  [Cited in This Article: ]
241.  Clinical trials.gov. A Phase 1b/2 Study of MEDI4736 With Tremelimumab, MEDI4736 or Tremelimumab Monotherapy in Gastric or GEJ Adenocarcinoma.  Available from: https://clinicaltrials.gov/ct2/show/NCT02340975.  [PubMed]  [DOI]  [Cited in This Article: ]
242.  Clinical trials.gov. A Study of Epacadostat (INCB024360) in Combination With Durvalumab (MEDI4736) in Subjects With Selected Advanced Solid Tumors.  Available from: https://clinicaltrials.gov/ct2/show/NCT02318277.  [PubMed]  [DOI]  [Cited in This Article: ]
243.  Clinical trials.gov. Planning Treatment for Oesophago-gastric Cancer: a Maintenance Therapy Trial (PLATFORM).  Available from: https://clinicaltrials.gov/ct2/show/NCT02678182.  [PubMed]  [DOI]  [Cited in This Article: ]
244.  Clinical trials.gov. A Study of Ramucirumab (LY3009806) Plus MEDI4736 in Participants With Advanced Gastrointestinal or Thoracic Malignancies.  Available from: https://clinicaltrials.gov/ct2/show/NCT02572687.  [PubMed]  [DOI]  [Cited in This Article: ]
245.  Clinical trials.gov. Phase 1 Study of MLN0264 in Adult Patients With Advanced Gastrointestinal Malignancies Expressing Guanylyl Cyclase C.  Available from: https://clinicaltrials.gov/show/NCT01577758.  [PubMed]  [DOI]  [Cited in This Article: ]
246.  Messersmith W, Almhanna K, Rodon J, Cruz C, Ryan D, Jung JA, Fasanmade A, Wyant T, Kalebic T. PD-0032MLN0264, an investigational, first-in-class antibodydrug conjugate targeting guanylyl cyclase C (GCC): first in-human study in patients with advanced gastrointestinal malignancies. Ann Oncol. 2013;24:piv36.  [PubMed]  [DOI]  [Cited in This Article: ]
247.  Clinical trials.gov. A Study of MLN0264 in Patients With Cancer of the Stomach or Gastroesophageal Junction.  Available from: https://clinicaltrials.gov/ct2/show/NCT02202759.  [PubMed]  [DOI]  [Cited in This Article: ]
248.  Clinical trials.gov. MLN0264 in Previously Treated Asian Patients With Advanced Gastrointestinal Carcinoma or Metastatic or Recurrent Gastric or Gastroesophageal Junction Adenocarcinoma Expressing Guanylyl Cyclase C.  Available from: https://clinicaltrials.gov/ct2/show/NCT02391038.  [PubMed]  [DOI]  [Cited in This Article: ]
249.  Lim S, Kaldis P. Cdks, cyclins and CKIs: roles beyond cell cycle regulation. Development. 2013;140:3079-3093.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 931]  [Cited by in F6Publishing: 1205]  [Article Influence: 109.5]  [Reference Citation Analysis (0)]
250.  Holkova B, Supko JG, Ames MM, Reid JM, Shapiro GI, Perkins EB, Ramakrishnan V, Tombes MB, Honeycutt C, McGovern RM. A phase I trial of vorinostat and alvocidib in patients with relapsed, refractory, or poor prognosis acute leukemia, or refractory anemia with excess blasts-2. Clin Cancer Res. 2013;19:1873-1883.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 26]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
251.  Schwartz GK, Ilson D, Saltz L, O’Reilly E, Tong W, Maslak P, Werner J, Perkins P, Stoltz M, Kelsen D. Phase II study of the cyclin-dependent kinase inhibitor flavopiridol administered to patients with advanced gastric carcinoma. J Clin Oncol. 2001;19:1985-1992.  [PubMed]  [DOI]  [Cited in This Article: ]
252.  Clinical trials.gov. Irinotecan Hydrochloride With or Without Alvocidib in Treating Patients With Advanced Stomach or Gastroesophageal Junction Cancer That Cannot Be Removed By Surgery.  Available from: https://clinicaltrials.gov/ct2/show/NCT00991952.  [PubMed]  [DOI]  [Cited in This Article: ]
253.  Xu L, Qu X, Luo Y, Zhang Y, Liu J, Qu J, Zhang L, Liu Y. Epirubicin enhances TRAIL-induced apoptosis in gastric cancer cells by promoting death receptor clustering in lipid rafts. Mol Med Rep. 2011;4:407-411.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 18]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
254.  Cusack JC. Rationale for the treatment of solid tumors with the proteasome inhibitor bortezomib. Cancer Treat Rev. 2003;29 Suppl 1:21-31.  [PubMed]  [DOI]  [Cited in This Article: ]
255.  Karin M. Nuclear factor-kappaB in cancer development and progression. Nature. 2006;441:431-436.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2609]  [Cited by in F6Publishing: 2674]  [Article Influence: 148.6]  [Reference Citation Analysis (0)]
256.  Berenson JR, Yang HH, Sadler K, Jarutirasarn SG, Vescio RA, Mapes R, Purner M, Lee SP, Wilson J, Morrison B. Phase I/II trial assessing bortezomib and melphalan combination therapy for the treatment of patients with relapsed or refractory multiple myeloma. J Clin Oncol. 2006;24:937-944.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 164]  [Cited by in F6Publishing: 167]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
257.  Kane RC, Dagher R, Farrell A, Ko CW, Sridhara R, Justice R, Pazdur R. Bortezomib for the treatment of mantle cell lymphoma. Clin Cancer Res. 2007;13:5291-5294.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 262]  [Cited by in F6Publishing: 276]  [Article Influence: 16.2]  [Reference Citation Analysis (0)]
258.  Fujita T, Doihara H, Washio K, Ino H, Murakami M, Naito M, Shimizu N. Antitumor effects and drug interactions of the proteasome inhibitor bortezomib (PS341) in gastric cancer cells. Anticancer Drugs. 2007;18:677-686.  [PubMed]  [DOI]  [Cited in This Article: ]
259.  Bae SH, Ryoo HM, Kim MK, Lee KH, Sin JI, Hyun MS. Effects of the proteasome inhibitor bortezomib alone and in combination with chemotherapeutic agents in gastric cancer cell lines. Oncol Rep. 2008;19:1027-1032.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
260.  Ocean AJ, Christos P, Sparano JA, Shah MA, Yantiss RK, Cheng J, Lin J, Papetti M, Matulich D, Schnoll-Sussman F. Phase II trial of bortezomib alone or in combination with irinotecan in patients with adenocarcinoma of the gastroesophageal junction or stomach. Invest New Drugs. 2014;32:542-548.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 8]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
261.  Jatoi A, Dakhil SR, Foster NR, Ma C, Rowland KM, Moore DF, Jaslowski AJ, Thomas SP, Hauge MD, Flynn PJ. Bortezomib, paclitaxel, and carboplatin as a first-line regimen for patients with metastatic esophageal, gastric, and gastroesophageal cancer: phase II results from the North Central Cancer Treatment Group (N044B). J Thorac Oncol. 2008;3:516-520.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 27]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
262.  Shah MA, Power DG, Kindler HL, Holen KD, Kemeny MM, Ilson DH, Tang L, Capanu M, Wright JJ, Kelsen DP. A multicenter, phase II study of bortezomib (PS-341) in patients with unresectable or metastatic gastric and gastroesophageal junction adenocarcinoma. Invest New Drugs. 2011;29:1475-1481.  [PubMed]  [DOI]  [Cited in This Article: ]
263.  Zhang QW, Liu L, Chen R, Wei YQ, Li P, Shi HS, Zhao YW. Matrix metalloproteinase-9 as a prognostic factor in gastric cancer: a meta-analysis. Asian Pac J Cancer Prev. 2012;13:2903-2908.  [PubMed]  [DOI]  [Cited in This Article: ]
264.  He L, Chu D, Li X, Zheng J, Liu S, Li J, Zhao Q, Ji G. Matrix metalloproteinase-14 is a negative prognostic marker for patients with gastric cancer. Dig Dis Sci. 2013;58:1264-1270.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 30]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
265.  Bramhall SR, Hallissey MT, Whiting J, Scholefield J, Tierney G, Stuart RC, Hawkins RE, McCulloch P, Maughan T, Brown PD. Marimastat as maintenance therapy for patients with advanced gastric cancer: a randomised trial. Br J Cancer. 2002;86:1864-1870.  [PubMed]  [DOI]  [Cited in This Article: ]
266.  Sampieri CL, León-Córdoba K, Remes-Troche JM. Matrix metalloproteinases and their tissue inhibitors in gastric cancer as molecular markers. J Cancer Res Ther. 2013;9:356-363.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 32]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]