Topic Highlight Open Access
Copyright ©The Author(s) 2016. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Feb 28, 2016; 22(8): 2415-2423
Published online Feb 28, 2016. doi: 10.3748/wjg.v22.i8.2415
Fibroblast growth factor receptor signaling as therapeutic targets in gastric cancer
Masakazu Yashiro, Tasuku Matsuoka, Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka 545-8585, Japan
Masakazu Yashiro, Oncology Institute of Geriatrics and Medical Science, Osaka City University Graduate School of Medicine, Osaka 545-8585, Japan
Author contributions: Yashiro M and Matsuoka T designed this review; Yashiro M wrote and edited the manuscript.
Supported by KAKENHI (partially, Grant-in-Aid for Scientific Research, No. 23390329).
Conflict-of-interest statement: There are not any financial or other interests with regard to the submitted manuscript that might be construed as a conflict of interest.
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: Masakazu Yashiro, MD, Department of Surgical Oncology, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka 545-8585, Japan. m9312510@med.osaka-cu.ac.jp
Telephone: +81-6-66453838 Fax: +81-6-66466450
Received: April 27, 2015
Peer-review started: May 1, 2015
First decision: May 18, 2015
Revised: May 25, 2015
Accepted: December 8, 2015
Article in press: December 8, 2015
Published online: February 28, 2016

Abstract

Fibroblast growth factor receptors (FGFRs) regulate a variety of cellular functions, from embryogenesis to adult tissue homeostasis. FGFR signaling also plays significant roles in the proliferation, invasion, and survival of several types of tumor cells. FGFR-induced alterations, including gene amplification, chromosomal translocation, and mutations, have been shown to be associated with the tumor initiation and progression of gastric cancer, especially in diffuse-type cancers. Therefore, the FGFR signaling pathway might be one of the therapeutic targets in gastric cancer. This review aims to provide an overview of the role of FGFR signaling in tumorigenesis, tumor progression, proliferation, and chemoresistance. We also discuss the accumulating evidence that demonstrates the effectiveness of using clinical therapeutic agents to inhibit FGFR signaling for the treatment of gastric cancer.

Key Words: Fibroblast growth factor receptor, Gastric cancer, Signaling pathway, Targeted therapy

Core tip: Fibroblast growth factor receptor (FGFR) is one of the drivers signaling in the development of gastric cancer. The use of molecular agents as targets of the FGFRs pathway has currently been approved in experimental and clinical trials as a mono-targeted approach or in combination with chemotherapeutic agents. FGFRs might be a promising therapeutic target for the treatment of gastric cancer.



INTRODUCTION

Gastric cancer is one of the most common malignancies in the world. Despite considerable improvements in clinical diagnostics and surgical skills, the prognosis of advanced gastric cancer with distant metastasis is still poor with a median overall survival of approximately less than 1 year[1,2]. To date, the molecular heterogeneity underlining tumor initiation and progression has been elucidated[3]. Following the understanding of gastric cancer biology, targeted therapies have been evaluated in experimental studies and transferred promptly to clinical trials. The use of molecular agents to target the signaling pathways of several receptors, such as epidermal growth factor receptor (EGFR), human epidermal growth factor 2 (HER2), and vascular endothelial growth factor receptor (VEGFR), has been approved for the treatment of gastric cancer as a mono-targeted approach or in combination with cytotoxic chemotherapeutic drugs[4]. Fibroblast growth factor receptor (FGFR) signaling also plays an important role in the development of various types of malignancies, including gastric cancer through the regulation of many other signaling pathways[5]. The FGFR2 gene, also known as the K-samII gene, has been shown to be amplified and overexpressed in gastric cancer, preferentially in diffuse-type cancers[6-9].

In this review, we discuss the genetic alterations induced by FGFR signaling and the role of FGFR signaling in gastric cancer. Subsequently, we review the possibility of a rational therapy through FGFR signaling inhibition.

FGFR CONSTRUCTION AND SIGNALING PATHWAY

The FGFR is a transmembrane tyrosine kinase receptor involved in signaling via interaction with members of the FGFR family (Figure 1). Four members of the FGFR family, FGFR1, FGFR2, FGFR3, and FGFR4, have been identified. These receptors are diverse depending on their ligand, binding affinity, and tissue distribution. FGFR consists of an extracellular domain, a transmembrane domain, and an intracellular domain[10]. The extracellular ligand domain is composed of three immunoglobulin (Ig)-like domains: D1, D2, and D3. The FGFR family binds to their ligands, the fibroblast growth factors (FGFs), with high affinity[11]. FGFR1, FGFR2, and FGFR3 are divided into types IIIb and IIIc based on the alternative splicing within the C-terminal half of the third Ig loop (D3) in the extracellular FGF binding domain. Exon 8 produces the IIIb isoform, whereas exon 9 produces the IIIc isoform. The IIIb isoform is mainly expressed in epithelial cells, while the IIIc isoform is preferentially expressed in mesenchymal cells.

Figure 1
Figure 1 The fiblobrast growth factor structure and signaling network. The Fibroblast growth factor (FGF)-FGFR complex consists of two receptor molecules, two FGFs, and a heparan sulfate proteoglycan (HSPG). The HSPG stabilizes and sequesters the FGFs. The FGFR comprises three extracellular Ig-like domains (D1-3), a transmembrane helix, and an intracellular split tyrosine kinase domain. D1 and D3 are separated by an acidic box. Ligand specificity of FGFR is regulated by alternative splicing within the C-terminal half of the third Ig loop (D3) in the ligand binding domain. Ligand binding and FGFR dimerization trigger the kinase domain phosphorylation, which leads to the docking of adapter residues and the activation of downstream pathways. Ligand-stimulated FGFRs phosphorylate the FGFR-associated cytosolic docking protein, FRS2. FRS2 activates SOS and GRB2, which in turn activate the Ras-Raf-MAPK pathway. A different complex involves GAB1, which activates the PI3K-Akt pathway. PLCγ hydrolyzes PIP2 to IP3 and DAG. PIP3 produces calcium. DAG also activates PKC, which results in the activation of the MAPK pathway by phosphorylating Raf. HSPG: Heparan sulfate proteoglycan; DAG: Diacylglycerol; FRS2α: FGFR substrate 2α; GRB2: Growth factor receptor-bound 2; IP3: Inositol triphosphate; PI3K: Phosphoinositide 3-kinase; PIP: 2-phosphatidylinositol-4,5-biphosphate; PKC: Protein kinase C; SOS: Son of sevenless; GAB1: GRB2-associated binding protein 1; PLCγ: Phospholipase Cγ.

FGF ligands are a family of 22 structurally related proteins that are further divided into subfamilies according to their sequence homology. The secreted-type ligands of the FGF family, including FGF1-10 and FGF16-23, bind to multiple FGFRs to transduce signals in target cells. The FGF-FGFR complex comprises two receptor molecules, two FGFs, and a heparan sulfate proteoglycan. Alternative splicing of D3 in the ligand binding domain mediates the ligand specificity. The IIIb isoform preferentially binds secreted FGF ligands from adjacent mesenchyme such as FGF7, FGF10, and FGF22; the IIIc isoform usually binds ligands secreted from the adjacent epithelium.

A major downstream signaling route of the FGFR family is via the Ras-Raf-mitogen-activated-protein-kinase (MAPK) pathway. Activation of Ras initiates a multistep phosphorylation cascade that leads to the activation of such MAPK as extracellular signal-regulated kinase 1 (ERK1) and ERK2. ERK1 and 2 regulate the transcription of molecules linked to cell proliferation, survival, and transformation[12]. Another important cascade in FGFR signaling is phosphatidylinositol 3-kinase (PI3K) and the downstream protein-serine/threonine kinase Akt; the latter transduces signals triggering a cascade of responses that promote cell growth and proliferation, survival, and motility[13]. Taken together, the activation of FGFR results in the activation of protein kinase C, the activators of transcription (STAT), and inositol-triphospate (IP3)-mediated Ca2+ release[11]. FGF signaling cascades also interact with Notch, Wnt, Hedgehog, and bone morphogenetic protein (BMP) signaling cascades to maintain the homeostasis among stem and progenitor cells[14]. These FGFR signaling pathways have been shown to mediate not only a variety of fundamental diverse cellular behaviors including embryogenesis and adult tissue homeostasis[11], but also oncogenesis, such as mitogenesis, differentiation, cell proliferation, angiogenesis, and invasion. The aberrant regulation of this pathway has been implicated in anti-apoptosis, drug resistance, and epithelial-to-mesenchymal transition (EMT).

FGFR ALTERATIONS IN GASTRIC CANCER

Genetic modification or overexpression of FGFRs has been associated with the tumor initiation and progression of several types of malignancies due to gene amplification, translocation, and mutations leading to enhanced kinase activity[7,11,15,16]. FGFR gene abnormalities have been reported in various cancers[17-19], including non-small cell lung cancer (FGFR1 amplification or FGFR2 mutation), breast cancer (FGFR1 and 2 amplification), squamous cell carcinoma (FGFR1 amplification), gastric cancer (FGFR2 amplification), bladder cancer (FGFR3 mutation or gene translocation), endometrial carcinoma (FGFR2 mutation), glioblastoma (FGFR3 gene translocation), and rhabdomyosarcoma (FGFR4-activating mutation).

In gastric cancer, FGFR2 amplification has been reported to occur in 2% to 9% of patients and more frequently in patients with diffuse-subtype cancers[20-24]. FGFR2 is identical to the product of the K-samII gene[25,26], which is cloned from a diffuse-type gastric cancer cell line, KATO-III[25,27], A previous paper has asserted that FGFR2 amplification in gastric cancer cell lines is dependent on the overexpressed and activated FGFR2 receptor for cell growth. Interestingly, FGFR2-amplified cell lines contain elevated EGFR, HER2, and ErbB3 phosphorylation that are regulated by FGFR2 kinase activity[23]. Meanwhile, a recent paper has revealed that aberrations in the FGFR2-ErbB3-PI3 kinase pathway are found in diffuse-type gastric cancers, while intestinal-type gastric cancer has an activated ErbB2 oncogenic pathway[28], FGFR2 amplification has been found to be associated with a higher pT stage, a higher pN stage, and lymph node metastasis, as well as related to poor overall survival[9,21,22,29]. Interestingly, cancer genomic profiling studies have shown that while the amplification of the FGFR2 gene is prompted in the primary diffuse-gastric tumor, the FGFR2 locus is not amplified in the metastatic tumor[30].

Several papers have presented the role of FGFR4 in gastric cancer. The FGFR4 expression rate in gastric cancer specimens is 38% to 44%, and it has been associated with lymph node metastasis and poor prognosis. Silencing FGFR4 expression in gastric cancer cells results in decreased growth and an increased rate of apoptosis, which is correlated with caspase 3 and Bcl-xL activity[31,32]. One report has recently asserted that the co-expression of several FGFRs might be a useful prognostic factor. Patients with gastric cancer, who have elevated levels of FGFR1, FGFR2, or FGFR4, have a significantly shorter life expectancy. Furthermore, the co-overexpression of all three FGFRs results in a poorer disease-specific survival, compared to the expression of none or only one of the FGFRs, and serves as an independent prognostic factor[33]. Diagnostic detection of tumors with FGFR genetic alterations in the primary and secondary lesions could be useful for selecting patients for FGFR-targeted therapies.

FUNCTIONAL ROLE OF FGFR IN GASTRIC CANCER

FGFR2 mRNA is amplified in scirrhous gastric cancer cells, and ligand FGF7, also designated as keratinocyte growth factor (KGF), is produced by gastric fibroblasts. KGF secreted by fibroblasts has a pivotal role in the development of scirrhous gastric cancer with K-samII amplification in a paracrine manner[27]. Scirrhous-type gastric cancer, also designated as linitis plastica, is diffuse and biologically aggressive, typically infiltrating into the gastric wall with a high frequency of peritoneal metastasis. The proliferative and infiltrative ability of scirrhous gastric cancer cells is closely correlated with the growth factors produced by organ-specific fibroblasts[34]. We previously reported that the conditioned medium derived from gastric fibroblasts stimulates the growth of gastric cancer cells, which are mediated by FGF7/FGFR2 signaling.

EMT is a critical process in cancer progression that provides cancer cells with the ability to escape from the primary foci, invade stromal tissues, and metastasize to secondary regions due to decreased cell-cell adhesion. Most EMT processes are regulated by extracellular matrix (ECM) components and soluble growth factors. Among these, FGF/FGFRs signaling is positively associated with EMT[11]. The FGF/FGFRs signaling pathway has been shown to be associated with WNT signaling, which results in the regulation of cellular phenotype and migration, and subsequently leads to EMT. Twist-related protein 1 (Twist1), a transcriptional factor, has been reported to have an important role in cell lineage determination and differentiation. One paper has asserted that Twist1 prompts the expression of FGFR2 in gastric cancer cell lines, and that FGFR2 mediates Twist 1-induced invasion and EMT, suggesting that the dual inhibition of these factors can become a novel tool for the targeted therapy of gastric cancer[35].

EXPERIMENTAL STUDIES TARGETING FGFR SIGNALING IN GASTRIC CANCER

In ToGA trials, trastuzumab, an anti-HER2 targeting antibody, prolonged the overall survival of patients with HER2-positive gastric tumor when combined with conventional chemotherapy[36]. However, considering the low expression rate (6%-23%) of HER2 and the modest effect of the ToGA trial (only 2.7 mo of prolonged survival) in gastric cancer patients, the development of a novel targeted therapy with higher potency is still required. In contrast, therapies targeting VEGFR and EGFR have failed to translate their preclinical efficacy into improved patient outcomes in several trials for gastric cancer[37-39]. As FGFRs have an important role in the progression of gastric cancer, as mentioned previously, their use as a therapeutic candidate for the development of targeted anticancer agents should attract substantial attention. In this section, relevant literatures will be reviewed to identify the current and future roles of the FGFR family as a potential targeted signaling pathway for gastric cancer treatment (Table 1 and Figure 2). Preclinical studies have established that the inhibition of the FGFRs pathway, alone or in combination with other signaling or chemotherapy, reveals the antitumor activity in gastric cancer in vitro and in vivo.

Table 1 Recent preclinical studies of flobrast growth factor receptor-targeted therapy in gastric cancer.
FGFRs targeted therapeutics (Campany)Study (Ref.)Main results
Multikinase inhibitors
Ki23057 (KIRIN Brewery)Nakamura et al[42]Ki23057 significantly inhibited the proliferation and induced apoptosis in scirrhous gastric cancer lines
Dovitinib (Novartis)Deng et al[20]Dovitinib specifically inhibited cell proliferation and induced apoptosis in gastric cancer cells with potent amplification of FGFR1
S49076Burbridge et al[43]S49076 impaired the growth of MET- and FGFR2-dependent gastric cancer cells and showed good synergistic inhibition of MET and FGFR2 in vivo medel
Foretinib (GlaxoSmithKline)Kataoka et al[47]Foretinib inhibits the cell growth of gastric cancer cell lines by inhibiting not only MET but also FGFR kinase
FGFRs specific inhibitors
AZD4547 (AstraZeneca)Xie et al[50]AZD4547 application resulted in tumor growth inhibition in FGFR2-amplified gastric tumor xenograft and induction of apoptosis by impairing downstream signaling of FGFR2
PD173074Kunii et al[23]FGFR2 inhibition by PD173074 displayed potent and selective growth inhibition in FGFR2-amplified gastric cancer cell lines
LY2874455 (Eli Lilly and Company)Zhao et al[51]LY2874455 is more potent at inhibiting the growth of gastric cancer cells with an increased FGFRs-signaling activity without high blood pressure
RNA based therapy
siRNAZhou et al[54]Treatment with siRNA resulted in reduced proliferation and prompted apoptosis, which accompanied the reduction of VEGFR expression and increase of apoptosis-related proteins
miR-133bWen et al[55]Some micro RNAs (miR-133b) may contribute to the regulation of FGFR-1 receptor in gastric carcinomas such that FGFR1 expression is inversely correlated with miR- 133b expression
FGFRs antibodies
Gal-FR21, 22 and 23Zhao et al[52]Highly specific monoclonal antibodies against FGFR2 strongly depressed growth of xenografts from 2 gastric cancer cell lines, OCUM-2M and SNU-16
GP369Bai et al[53]GP369, an FGFR2-IIIb-specificantibody, exhibited anti-proliferative activity against gastric cancers driven FGFR2 overexpression
Figure 2
Figure 2 Fiblobrast growth factor receptor-targeted therapeutics. Fibroblast growth factor receptors (FGFRs)-targeted tyrosine kinase inhibitors are small molecule ATP-competitive agents that bind to the ATP-binding site of the intracellular tyrosine kinase domain of FGFRs. Monoclonal antibody targets the extracellular domain of the FGFR and blocks ligand binding. RNA interference suppresses the expression of various genes. RNAi: RNA interference.
FGFR tyrosine kinase inhibitor

FGFR tyrosine kinase inhibitors (TKIs) demonstrate their therapeutic effectiveness on cancer cells in vitro and in vivo[40,41]. Most of the FGFR TKIs include Ki23057[42], dovitinib, S49076[43], Pazopanib[44], foretinib[45-47], and Dovitinib (TKI258)[20,48,49], are not specific to FGFRs along with other kinases. Although the multi-kinase inhibitor compounds successfully inhibit the proliferation of FGFRs-amplified gastric cancer cells following the attenuation of multiple serine/threonine and tyrosine kinases, it is not clear whether specific FGFR2 inhibition alone is sufficient. AZD4547 [N-[5-[2-(3,5-dimethoxyphenyl)ethyl]-2Hpyrazol- 3-yl]-4-(3,5 diemthylpiperazin-1-yl) benzamide] is a selective ATP-competitive receptor tyrosine kinase inhibitor that interrupts recombinant FGFR1-3 activities. A previous paper has shown that AZD4547 application results in tumor growth inhibition in FGFR2-amplified gastric tumor xenografts and the induction of apoptosis by impairing the downstream signaling of FGFR2[50]. As a selective FGFR1-3 inhibitor, PD173074 has been evaluated for its anti-tumor efficacy in gastric cancer cell lines[23]. FGFR2 inhibition by PD173074 has displayed potent and selective growth inhibition in FGFR2-amplified gastric cancer cell lines. Interestingly, the inhibition of ErbB3 has also inhibited growth in FGFR2-amplified cell lines. Inhibitors of FGFR2 or ErbB3 signaling may have therapeutic efficacy in the subset of gastric cancers containing FGFR2 amplification. LY2874455 is an orally bioavailable, FGFR-dominant kinase inhibitor lacking significant activity against VEGFR2 in vivo. This molecule is effective on all FGFRs equally. LY2874455 is more potent at inhibiting the growth of gastric cancer cells with increased FGFR-signaling activity in vitro and in vivo. These results demonstrate the potential of FGFR inhibitors as attractive molecular targeted agents for patients for whom anti-VEGF/VEGFR therapies have been inactive[51].

FGFRs antibodies

As mentioned above, small molecule antagonists of FGFRs demonstrate significant efficacy for the treatment of gastric cancer. Highly specific monoclonal antibody against FGFR2 generated from mouse melanoma cells (GAL-FR21 and -22) has been found to strongly depress the growth of xenografts from two gastric cancer cell lines, OCUM-2M and SNU-16[52]. These results suggest that the anti-FGFR2 antibody would be an attractive clinical candidate for gastric cancer therapy[52]. At the same time, GP369, an FGFR2-IIIb-specific antibody, has exhibited anti-proliferative activity against gastric cancers driven by FGFR2 overexpression[53].

FPA144, an FGFR2b-specific antibody, is developed to treat patients with gastric cancers bearing an amplification of the FGFR2 gene (J Clin Oncol 2014; 32 suppl; abstr e15074). FPA144 is glycol-engineered for enhanced antibody-dependent cell-mediated cytotoxicity, a process in which cells of the immune system kill the tumor cell recognized by the antibody. FPA144 may act to prevent the binding of FGFs, such as FGF7, FGF10, and FGF22, to FGFR2b, as well as inhibit their ability to promote the growth of tumor cells. FP-1039, also known as GSK3052230, is a protein drug designed to intervene in the FGF signaling through FGFR1 that stimulates cancer cell growth and angiogenesis. FP-1039 can bind to FGF ligands circulating in the extracellular space, preventing these signaling proteins from reaching FGFR1 on the surface of tumor cells. A phase IB trial to evaluate FP-1039 in combination with paclitaxel and carboplatin, or docetaxel in subjects with solid tumor, is ongoing (NCT01868022).

RNA interference-based therapeutics

RNA interferences a recent potent technology for the treatment of malignancies. Small interfering RNA (siRNA) plays an integral role in RNA silencing therapy due to its stability and lower toxicity. A recent study has examined the effect of FGFR-targeting siRNA on the proliferation of gastric cancer cell lines[54]. Treatment with siRNA has been shown to result in reduced proliferation and enhanced apoptosis, which accompany the reduction of VEGFR expression and the increase of apoptosis-related proteins. Some microRNAs (miR-133b) may contribute to the regulation of FGFR1 in gastric carcinomas such that FGFR1 expression is inversely correlated with miR-133b expression, suggesting that miR-133b might act as a novel tumor suppressor mechanism in gastric cancer cases[55].

Chemoresistance

The development of drug resistant clones is one of the most serious challenges in the field of clinical oncology[56]. FGF family has been shown to be associated with drug resistance in some type of cancer cells to which a variety of conventional chemotherapy has been applied[57-59]. We previously reported that the combination of Ki23057 and 5-FU or S1 shows synergistic anti-tumor effects on diffuse-type gastric cancer cells by decreasing dihydropyrimidine dehydrogenase expression and increasing p21 expression[34]. Accordingly, we explored the effects of Ki23057, combined with various known cytotoxic agents such as irinotecan, paclitaxel, etoposide, oxaliplatin, and gemcitabine, respectively, in cultures of a gastric cancer cell line and its drug-resistant clones[60]. FGFR2 inhibitor could be the therapeutic agent for treating drug-resistant gastric cancer cells. In terms of the clinical trials of FGFRs inhibitors, rationally designed combination trials would be needed to abrogate the resistance to FGFRs targeted therapy and further improve clinical outcomes.

CLINICAL THERAPEUTIC TRIALS AGAINST FGFR SIGNALING IN GASTRIC CANCER

According to the positive results from preclinical experiments, several FGFRs inhibitors are being tested in clinical trials in patients with gastric cancer. To date, five trials are ongoing, as displayed in Table 2. A phase I expansion cohort study of AZD4547 for the treatment of advanced gastric and gastroesophageal cancer was conducted. All patients included were selected prospectively for FGFR2 amplification using the fluorescent in situ hybridization (FISH) analysis of archival or fresh tumors. Preliminary analysis showed that one partial response was observed in a patient with a tumor that had clusters of FGFR gene amplification and that four patients had a stable disease (three with high FGFR amplification and one with clusters) (2014 ASCO Annual Meeting). A phase II study, comparing AZD4547 with paclitaxel in the second-line setting for patients with FGFR polysomy or amplification, is ongoing (NCT01457846, SHINE trial). A phase II trial of dovitinib monotherapy, as a salvage treatment in patients with metastatic or unresectable gastric cancer harboring FGFR2 amplification after failure of first- or second-line chemotherapy, is ongoing (NCT01719549). An open labeled phase I/II trial of docetaxel plus dovitinib, as the second-line chemotherapy for patients with metastatic or unresectable gastric cancer after failure of first-line chemotherapy, is currently underway (NCT01921673). FGFR inhibitor TAS-120 selectively and irreversibly binds to and inhibits FGFR. A phase II trial studying the efficacy of TAS-120 monotherapy in patients with advanced solid tumors or multiple myeloma with FGF/FGFR-related abnormalities is ongoing (NCT02052778). A phase I open label, dose-finding study evaluating the safety and pharmacokinetics of FPA144, an FGFR2b-specific antibody, is currently underway for patients with advanced solid tumors with FGFR2b overexpression and FGFR2 amplification (NCT02318329).

Table 2 Clinical trials of fiblobrast growth factor receptor 2-targeted therapy in gastric cancer.
TrialStudy designFGFRs statusRegimenResponse rateStatus
NCT01457846Phase I/1stFGFR2amplification FISH +AZD454738.5% (contained SD)Conducted
SHINEPhase II/1stFGFR2amplification FISH +AZD4547 paclitaxelComputed accural
NCT01719549Phase II/2ndFGFR2 amplificationDovitinibOngoing
NCT01921673Phase I/II/2ndFGFR2 amplificationDovitinib, docetaxelOngoing
NCT02052778Phase I/II/2ndFGF/FGFR-Related AbnormalitiesTAS-120Ongoing
NCT02318329Phase I/1stFGFR2b overexpression and FGFR2 amplificationFPA144Ongoing
CONCLUSION AND FUTURE PERSPECTIVES

The activating FGFR signaling in FGFR-amplified or FGFR-overexpressed tumors is an attractive therapeutic target for gastric cancer. Novel FGFR inhibitors have recently been developed, and some of them are undergoing clinical trials for treating gastric cancers associated with aberrant FGFR signaling. Combination therapy of FGFR inhibitors and conventional cytotoxic agents may provide a synergistic anti-tumor effect and the possibility of overcoming chemoresistance in gastric cancer.

Although several evidences have shown the effectiveness of FGFR therapeutics, no clinical FGFR inhibitor has been approved. The main hurdle facing the effective usage of FGFR-targeted agents for cancer treatment is the development of a predictive biomarker that would allow optimal patient selection. There is a need for the establishment of biomarker-based patient selection, as it would most likely benefit FGFR-targeted therapy. Secondly, there are insufficient data to identify any one of the FGFR-targeted therapies as superior to the others. A recent high-resolution genomic analysis has presented a comprehensive survey of the genomic alterations in gastric cancer; this has revealed several promising targets for subtype-specific therapies[20]. RTK/Ras signaling are frequently altered and mutually exclusive to one another in gastric cancer. FGFR2 is amplified at frequencies comparable to ERBB2, and KRAS amplification is a prevalent event in gastric cancer[61]. Classifying gastric cancer patients with signature genomic alterations may facilitate patient allocations to the most appropriate clinical trials. Whereas several obstacles may exist and must wait to be overcome, we believe that FGFR-targeted therapy can be a promising candidate for the development of future therapies for patients with certain subtypes of gastric cancer.

ACKNOWLEDGMENTS

We thank our colleagues in our laboratory for helpful discussions. We apologize for not being able to cite all the relevant publications.

Footnotes

P- Reviewer: Blanco Luz P, Yao HR S- Editor: Yu J L- Editor: A E- Editor: Zhang DN

References
1.  Brenner H, Rothenbacher D, Arndt V. Epidemiology of stomach cancer. Methods Mol Biol. 2009;472:467-477.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Hartgrink HH, Jansen EP, van Grieken NC, van de Velde CJ. Gastric cancer. Lancet. 2009;374:477-490.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Holbrook JD, Parker JS, Gallagher KT, Halsey WS, Hughes AM, Weigman VJ, Lebowitz PF, Kumar R. Deep sequencing of gastric carcinoma reveals somatic mutations relevant to personalized medicine. J Transl Med. 2011;9:119.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  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: ]
5.  Katoh M, Katoh M. FGF signaling network in the gastrointestinal tract (review). Int J Oncol. 2006;29:163-168.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Katoh Y, Katoh M. FGFR2-related pathogenesis and FGFR2-targeted therapeutics (Review). Int J Mol Med. 2009;23:307-311.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Grose R, Dickson C. Fibroblast growth factor signaling in tumorigenesis. Cytokine Growth Factor Rev. 2005;16:179-186.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Moffa AB, Tannheimer SL, Ethier SP. Transforming potential of alternatively spliced variants of fibroblast growth factor receptor 2 in human mammary epithelial cells. Mol Cancer Res. 2004;2:643-652.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Toyokawa T, Yashiro M, Hirakawa K. Co-expression of keratinocyte growth factor and K-sam is an independent prognostic factor in gastric carcinoma. Oncol Rep. 2009;21:875-880.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Wesche J, Haglund K, Haugsten EM. Fibroblast growth factors and their receptors in cancer. Biochem J. 2011;437:199-213.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Katoh M, Nakagama H. FGF receptors: cancer biology and therapeutics. Med Res Rev. 2014;34:280-300.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Kamata T, Hattori Y, Hamada H, Kizaki M, Terada M, Ikeda Y. Keratinocyte growth factor regulates proliferation and differentiation of hematopoietic cells expressing the receptor gene K-sam. Exp Hematol. 2002;30:297-305.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Schlessinger J. Cell signaling by receptor tyrosine kinases. Cell. 2000;103:211-225.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Katoh M. FGFR2 abnormalities underlie a spectrum of bone, skin, and cancer pathologies. J Invest Dermatol. 2009;129:1861-1867.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Greulich H, Pollock PM. Targeting mutant fibroblast growth factor receptors in cancer. Trends Mol Med. 2011;17:283-292.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Brooks AN, Kilgour E, Smith PD. Molecular pathways: fibroblast growth factor signaling: a new therapeutic opportunity in cancer. Clin Cancer Res. 2012;18:1855-1862.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Kelleher FC, O’Sullivan H, Smyth E, McDermott R, Viterbo A. Fibroblast growth factor receptors, developmental corruption and malignant disease. Carcinogenesis. 2013;34:2198-2205.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 91]  [Cited by in F6Publishing: 96]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
18.  Dienstmann R, Rodon J, Prat A, Perez-Garcia J, Adamo B, Felip E, Cortes J, Iafrate AJ, Nuciforo P, Tabernero J. Genomic aberrations in the FGFR pathway: opportunities for targeted therapies in solid tumors. Ann Oncol. 2014;25:552-563.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 260]  [Cited by in F6Publishing: 268]  [Article Influence: 26.8]  [Reference Citation Analysis (0)]
19.  Katoh M. Dysregulation of stem cell signaling network due to germline mutation, SNP, Helicobacter pylori infection, epigenetic change and genetic alteration in gastric cancer. Cancer Biol Ther. 2007;6:832-839.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Deng N, Goh LK, Wang H, Das K, Tao J, Tan IB, Zhang S, Lee M, Wu J, Lim KH. A comprehensive survey of genomic alterations in gastric cancer reveals systematic patterns of molecular exclusivity and co-occurrence among distinct therapeutic targets. Gut. 2012;61:673-684.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Jung EJ, Jung EJ, Min SY, Kim MA, Kim WH. Fibroblast growth factor receptor 2 gene amplification status and its clinicopathologic significance in gastric carcinoma. Hum Pathol. 2012;43:1559-1566.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Liu YJ, Shen D, Yin X, Gavine P, Zhang T, Su X, Zhan P, Xu Y, Lv J, Qian J. HER2, MET and FGFR2 oncogenic driver alterations define distinct molecular segments for targeted therapies in gastric carcinoma. Br J Cancer. 2014;110:1169-1178.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Kunii K, Davis L, Gorenstein J, Hatch H, Yashiro M, Di Bacco A, Elbi C, Lutterbach B. FGFR2-amplified gastric cancer cell lines require FGFR2 and Erbb3 signaling for growth and survival. Cancer Res. 2008;68:2340-2348.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Das K, Gunasegaran B, Tan IB, Deng N, Lim KH, Tan P. Mutually exclusive FGFR2, HER2, and KRAS gene amplifications in gastric cancer revealed by multicolour FISH. Cancer Lett. 2014;353:167-175.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Hattori Y, Odagiri H, Nakatani H, Miyagawa K, Naito K, Sakamoto H, Katoh O, Yoshida T, Sugimura T, Terada M. K-sam, an amplified gene in stomach cancer, is a member of the heparin-binding growth factor receptor genes. Proc Natl Acad Sci USA. 1990;87:5983-5987.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Katoh M, Hattori Y, Sasaki H, Tanaka M, Sugano K, Yazaki Y, Sugimura T, Terada M. K-sam gene encodes secreted as well as transmembrane receptor tyrosine kinase. Proc Natl Acad Sci USA. 1992;89:2960-2964.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Nakazawa K, Yashiro M, Hirakawa K. Keratinocyte growth factor produced by gastric fibroblasts specifically stimulates proliferation of cancer cells from scirrhous gastric carcinoma. Cancer Res. 2003;63:8848-8852.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Yamashita K, Sakuramoto S, Watanabe M. Genomic and epigenetic profiles of gastric cancer: potential diagnostic and therapeutic applications. Surg Today. 2011;41:24-38.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Betts G, Valentine H, Pritchard S, Swindell R, Williams V, Morgan S, Griffiths EA, Welch I, West C, Womack C. FGFR2, HER2 and cMet in gastric adenocarcinoma: detection, prognostic significance and assessment of downstream pathway activation. Virchows Arch. 2014;464:145-156.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Nadauld LD, Garcia S, Natsoulis G, Bell JM, Miotke L, Hopmans ES, Xu H, Pai RK, Palm C, Regan JF. Metastatic tumor evolution and organoid modeling implicate TGFBR2 as a cancer driver in diffuse gastric cancer. Genome Biol. 2014;15:428.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Ye YW, Zhou Y, Yuan L, Wang CM, Du CY, Zhou XY, Zheng BQ, Cao X, Sun MH, Fu H. Fibroblast growth factor receptor 4 regulates proliferation and antiapoptosis during gastric cancer progression. Cancer. 2011;117:5304-5313.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Chen H, Shen DP, Zhang ZZ, Liu JH, Shen YY, Ni XZ. Fibroblast growth factor receptor 4 protein expression and clinicopathological features in gastric cancer. World J Gastroenterol. 2015;21:1838-1844.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 8]  [Cited by in F6Publishing: 10]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
33.  Murase H, Inokuchi M, Takagi Y, Kato K, Kojima K, Sugihara K. Prognostic significance of the co-overexpression of fibroblast growth factor receptors 1, 2 and 4 in gastric cancer. Mol Clin Oncol. 2014;2:509-517.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Yashiro M, Shinto O, Nakamura K, Tendo M, Matsuoka T, Matsuzaki T, Kaizaki R, Miwa A, Hirakawa K. Synergistic antitumor effects of FGFR2 inhibitor with 5-fluorouracil on scirrhous gastric carcinoma. Int J Cancer. 2010;126:1004-1016.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Zhu DY, Guo QS, Li YL, Cui B, Guo J, Liu JX, Li P. Twist1 correlates with poor differentiation and progression in gastric adenocarcinoma via elevation of FGFR2 expression. World J Gastroenterol. 2014;20:18306-18315.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 22]  [Cited by in F6Publishing: 22]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
36.  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: ]
37.  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: ]
38.  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: ]
39.  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: ]
40.  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: ]
41.  Gavine PR, Mooney L, Kilgour E, Thomas AP, Al-Kadhimi K, Beck S, Rooney C, Coleman T, Baker D, Mellor MJ. AZD4547: an orally bioavailable, potent, and selective inhibitor of the fibroblast growth factor receptor tyrosine kinase family. Cancer Res. 2012;72:2045-2056.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  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: ]
43.  Burbridge MF, Bossard CJ, Saunier C, Fejes I, Bruno A, Léonce S, Ferry G, Da Violante G, Bouzom F, Cattan V. S49076 is a novel kinase inhibitor of MET, AXL, and FGFR with strong preclinical activity alone and in association with bevacizumab. Mol Cancer Ther. 2013;12:1749-1762.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Kim ST, Jang HL, Lee SJ, Lee J, Choi YL, Kim KM, Cho J, Park SH, Park YS, Lim HY. Pazopanib, a novel multitargeted kinase inhibitor, shows potent in vitro antitumor activity in gastric cancer cell lines with FGFR2 amplification. Mol Cancer Ther. 2014;13:2527-2536.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 32]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
45.  Mohammadi M, Froum S, Hamby JM, Schroeder MC, Panek RL, Lu GH, Eliseenkova AV, Green D, Schlessinger J, Hubbard SR. Crystal structure of an angiogenesis inhibitor bound to the FGF receptor tyrosine kinase domain. EMBO J. 1998;17:5896-5904.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Daniele G, Corral J, Molife LR, de Bono JS. FGF receptor inhibitors: role in cancer therapy. Curr Oncol Rep. 2012;14:111-119.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  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: ]
48.  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: ]
49.  Chowbay B, Jada SR, Wan Teck DL. Correspondence re: Cecchin et al., Carboxylesterase isoform 2 mRNA expression in peripheral blood mononuclear cells is a predictive marker of the irinotecan to SN38 activation step in colorectal cancer patients. Clin Cancer Res 2005; 11: 6901-7. Clin Cancer Res. 2006;12:1942; author reply 1942-1943.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Xie L, Su X, Zhang L, Yin X, Tang L, Zhang X, Xu Y, Gao Z, Liu K, Zhou M. FGFR2 gene amplification in gastric cancer predicts sensitivity to the selective FGFR inhibitor AZD4547. Clin Cancer Res. 2013;19:2572-2583.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Zhao G, Li WY, Chen D, Henry JR, Li HY, Chen Z, Zia-Ebrahimi M, Bloem L, Zhai Y, Huss K. A novel, selective inhibitor of fibroblast growth factor receptors that shows a potent broad spectrum of antitumor activity in several tumor xenograft models. Mol Cancer Ther. 2011;10:2200-2210.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Zhao WM, Wang L, Park H, Chhim S, Tanphanich M, Yashiro M, Kim KJ. Monoclonal antibodies to fibroblast growth factor receptor 2 effectively inhibit growth of gastric tumor xenografts. Clin Cancer Res. 2010;16:5750-5758.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 62]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
53.  Bai A, Meetze K, Vo NY, Kollipara S, Mazsa EK, Winston WM, Weiler S, Poling LL, Chen T, Ismail NS. GP369, an FGFR2-IIIb-specific antibody, exhibits potent antitumor activity against human cancers driven by activated FGFR2 signaling. Cancer Res. 2010;70:7630-7639.  [PubMed]  [DOI]  [Cited in This Article: ]
54.  Zhou D, Jiang X, Ding W, Zheng L, Yang L, Zheng C, Lu L. siRNA-participated chemotherapy: an efficient and specific therapeutic against gastric cancer. J Cancer Res Clin Oncol. 2013;139:2057-2070.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Wen D, Li S, Ji F, Cao H, Jiang W, Zhu J, Fang X. miR-133b acts as a tumor suppressor and negatively regulates FGFR1 in gastric cancer. Tumour Biol. 2013;34:793-803.  [PubMed]  [DOI]  [Cited in This Article: ]
56.  Bertolini F, Marighetti P, Martin-Padura I, Mancuso P, Hu-Lowe DD, Shaked Y, D’Onofrio A. Anti-VEGF and beyond: shaping a new generation of anti-angiogenic therapies for cancer. Drug Discov Today. 2011;16:1052-1060.  [PubMed]  [DOI]  [Cited in This Article: ]
57.  Levina V, Su Y, Nolen B, Liu X, Gordin Y, Lee M, Lokshin A, Gorelik E. Chemotherapeutic drugs and human tumor cells cytokine network. Int J Cancer. 2008;123:2031-2040.  [PubMed]  [DOI]  [Cited in This Article: ]
58.  Chell V, Balmanno K, Little AS, Wilson M, Andrews S, Blockley L, Hampson M, Gavine PR, Cook SJ. Tumour cell responses to new fibroblast growth factor receptor tyrosine kinase inhibitors and identification of a gatekeeper mutation in FGFR3 as a mechanism of acquired resistance. Oncogene. 2013;32:3059-3070.  [PubMed]  [DOI]  [Cited in This Article: ]
59.  Harbinski F, Craig VJ, Sanghavi S, Jeffery D, Liu L, Sheppard KA, Wagner S, Stamm C, Buness A, Chatenay-Rivauday C. Rescue screens with secreted proteins reveal compensatory potential of receptor tyrosine kinases in driving cancer growth. Cancer Discov. 2012;2:948-959.  [PubMed]  [DOI]  [Cited in This Article: ]
60.  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: ]
61.  Lee J, Ou SH. Towards the goal of personalized medicine in gastric cancer--time to move beyond HER2 inhibition. Part I: Targeting receptor tyrosine kinase gene amplification. Discov Med. 2013;15:333-341.  [PubMed]  [DOI]  [Cited in This Article: ]