Next Article in Journal
Pathology of Urologic Cancers
Next Article in Special Issue
Golgi-Targeting Anticancer Natural Products
Previous Article in Journal
Linear Tumor Regression of Rectal Cancer in Daily MRI during Preoperative Chemoradiotherapy: An Insight of Tumor Regression Velocity for Personalized Cancer Therapy
Previous Article in Special Issue
The CBP/β-Catenin Antagonist, ICG-001, Inhibits Tumor Metastasis via Blocking of the miR-134/ITGB1 Axis-Mediated Cell Adhesion in Nasopharyngeal Carcinoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Receptor Tyrosine Kinases Amplified in Diffuse-Type Gastric Carcinoma: Potential Targeted Therapies and Novel Downstream Effectors

Department of Cancer Cell Research, Sasaki Institute, Sasaki Foundation, Tokyo 101-0062, Japan
*
Author to whom correspondence should be addressed.
Cancers 2022, 14(15), 3750; https://doi.org/10.3390/cancers14153750
Submission received: 29 June 2022 / Revised: 28 July 2022 / Accepted: 28 July 2022 / Published: 1 August 2022
(This article belongs to the Special Issue A Deeper Dive into Signaling Pathways in Cancers)

Abstract

:

Simple Summary

Diffuse-type gastric carcinoma (DGC) is an aggressive subtype of gastric carcinoma with an extremely poor prognosis due to frequent peritoneal metastasis and high probability of recurrence. Its pathogenesis is poorly understood, and consequently, no effective molecular targeted therapy is available. The importance of oncogenic receptor tyrosine kinase (RTK) signaling has been recently demonstrated in the malignant progression of DGC. In particular, RTK gene amplification appears to accelerate peritoneal metastasis. In this review, we provide an overview of RTK gene amplification in DGC and the potential of related targeted therapies.

Abstract

Gastric cancer (GC) is a major cause of cancer-related death worldwide. Patients with an aggressive subtype of GC, known as diffuse-type gastric carcinoma (DGC), have extremely poor prognoses. DGC is characterized by rapid infiltrative growth, massive desmoplastic stroma, frequent peritoneal metastasis, and high probability of recurrence. These clinical features and progression patterns of DGC substantially differ from those of other GC subtypes, suggesting the existence of specific oncogenic signals. The importance of gene amplification and the resulting aberrant activation of receptor tyrosine kinase (RTK) signaling in the malignant progression of DGC is becoming apparent. Here, we review the characteristics of RTK gene amplification in DGC and its importance in peritoneal metastasis. These insights may potentially lead to new targeted therapeutics.

1. Introduction

Gastric cancer (GC) has the fifth-highest incidence among cancers, and it is the fourth leading cause of cancer-related death worldwide [1]. According to Lauren’s classification, GC is histologically categorized into two main subtypes: intestinal-type gastric carcinoma (IGC) and diffuse-type gastric carcinoma (DGC) [2]. DGC is an aggressive subtype with an extremely poor prognosis owing to rapid infiltrative invasion within the submucosa and frequent occurrence of peritoneal dissemination and high probability of recurrence [3]. DGC contains poorly differentiated and signet ring carcinoma cells that solitarily exist within a dense tumor stroma because of the lack of cell–cell adhesion. Scirrhous gastric carcinoma (SGC), also referred to as linitis plastica, is a subtype of DGC that is characterized by an extensive desmoplastic reaction [4].
Peritoneal metastasis is the dissemination of cancer to the peritoneum, which covers the abdominal cavity and the intra-abdominal organs, and it is often associated with the formation of malignant ascites. The quality of life and survival rate of the patients are both greatly reduced by peritoneal metastasis. However, no effective molecular targeted therapy is currently available. Because the pathological and histological features of DGC are quite different from those of IGC, studies focusing on DGC are necessary to understand its molecular basis and to develop effective molecular targeted therapy. The importance of oncogenic receptor tyrosine kinase (RTK) signaling has been recently demonstrated in the malignant progression of DGC. In particular, RTK gene amplification appears to accelerate peritoneal metastasis, and thus it is considered a promising therapeutic target. In this review, we provide an overview of RTK gene amplification in DGC and the potential of related targeted therapies. The recent literature regarding the downstream effectors of RTKs is summarized. Finally, we highlight issues that should be addressed to effectively target amplified RTK in DGC.

2. Gene Amplification of RTKs in DGC

RTKs are cell surface receptors that regulate various cellular processes, including proliferation, survival, metabolism, differentiation, migration, and invasion. Upon binding to specific ligands, two RTK molecules dimerize and tyrosine-phosphorylate each other, and recruit and activate a variety of intracellular signaling molecules to trigger the activation of downstream signaling pathways, such as Ras/MAPK, PI3-kinase/Akt, JAK/Stat, and NF-κB pathways (Figure 1). RTKs are well-known oncogenic drivers that are aberrantly activated in a wide range of human cancers. Oncogenic activation of RTK signaling is caused in several ways, including overexpression, activating mutations, fusions, and ligand dysregulation [5]. RTK overexpression is commonly caused by gene amplification that provokes ligand-independent dimerization owing to local enrichment, causing constitutive activation of downstream signaling pathways. Cancer cells harboring gene amplification of RTKs often exhibit ‘oncogene addiction’, and their growth and survival are highly dependent on the activity of amplified RTKs. Hence, RTKs amplified in cancers have been considered promising therapeutic targets; many RTK inhibitors have been developed, evaluated in preclinical and clinical trials, and used in the clinic.
The amplification of RTK genes has been identified in GC, including EGFR encoding epidermal growth factor receptor (EGFR), ERBB2 encoding human epidermal growth factor receptor 2 (HER2, also known as ERBB2), MET encoding Met (also known as c-Met), and FGFR2 encoding fibroblast growth factor receptor 2 (FGFR2), as summarized in Table 1. We also performed gene-amplification analysis of these RTKs in GC using cBioPortal and several publicly available datasets (Table 2). The frequencies of gene amplification were as follows: EGFR, 2.7–14.7%; ERBB2, 8–22.0%; MET, 2.2–8.8%; FGFR2, 2–5.4%. This is roughly consistent with other large cohort studies and confirms that a subset of GC harbors amplification of these RTK genes.
Tsujino et al. reported that amplification of EGFR and ERBB2 occurs in poorly differentiated adenocarcinoma and signet ring cell carcinoma, i.e., DGC [6]. The incidence of EGFR amplification is higher in metastatic tumors than that in primary tumors. Moreover, EGFR overexpression is frequently observed in GC and is associated with worse prognosis (Table 1). A large cohort study reported that ERBB2 is amplified in approximately 23% of GC cases [17]. The rate of ERBB2 amplification was substantially higher in patients with IGC than that in those with DGC in several studies that analyzed different large cohorts (Table 1). Therefore, HER2 is less likely to play a specific role in DGC but is generally implicated in the progression of GC.
Met is a receptor for hepatocyte growth factor (HGF) and encoded by MET proto-oncogene [40]. Physiologically, Met plays an essential role in embryogenesis and tissue regeneration [41]. Met is aberrantly activated in various cancer types owing to gene amplification, point mutations, rearrangement, overexpression, and aberrant splicing [42]. Oncogenic activation of Met promotes diverse malignant aspects of tumors, including invasion, metastasis, drug resistance, and angiogenesis [41]. Kuniyasu et al. reported that MET amplification frequently occurs in SGC cell lines and tumor tissues [21]. Following studies also reported MET amplification in GC, although the rate differs among studies, and its association with DGC and unfavorable outcomes (Table 1).
FGFR2 is a receptor for fibroblast growth factors, and it is oncogenically activated by genetic alterations in cancer [43]. FGFR2 is identical to the product of the K-sam gene that was cloned as a gene amplified in the KATO-III SGC cell line [44]. FGFR2 overexpression caused by gene amplification is substantially more frequent in DGC and metastatic GCs than in primary GCs, and it is associated with tumor progression and poor patient survival [43] (Table 1).
The Cancer Genome Atlas (TCGA) project classified GC into four molecular subtypes: Epstein–Barr virus-positive, microsatellite-unstable, genomically stable, and chromosomally instable [10]. DGC is enriched in the genomically stable subtype and is characterized by recurrent E-cadherin and RhoA mutations or fusions of Rho GAPs. However, alterations in RTK genes are rare in the genomically stable subtype: EGFR (2%), ERBB2 (7%), FGFR2 (9%), and MET (0%). In contrast, a recent multiomics study demonstrated that alterations in RTK and downstream MAPK signaling pathways, mostly gene amplification of KRAS (19.4%), FGFR2 (11.2%), MET (7.1%), ERBB2 (5.1%), and EGFR (4.1%), occur more frequently in cancer cells within malignant ascites than those in primary DGC [12] (Figure 1). In addition, RTK alterations occur in a mutually exclusive manner [8,9,12]. In contrast, MET amplification has been reported to coexist with HER2 amplification in several tumor samples [18]. Interestingly, Tajiri et al. reported that HER2 was coamplified with EGFR, FGFR, and MET in some tumors, but in mutually exclusive cells [15]. Thus, the amplification of one RTK signaling component may be sufficient to drive tumor progression and can occur in a mutually exclusive manner at the cellular level. Taken together, RTK amplification is most likely a key event in the acquisition of malignant and metastatic phenotypes in DGC cells.
Loss of E-cadherin function that weakens cell–cell adhesion is a hallmark of DGC. Germline mutation of CDH1 gene encoding E-cadherin causes hereditary DGC [45]. Previous multiomics studies did not show any correlation between CDH1 mutation and RTK gene amplification [10,12]. Nevertheless, RTK signaling can negatively regulate E-cadherin function via downregulation/degradation of E-cadherin and disassembly of E-cadherin complexes [46]. Thus, RTK gene amplification and resulting oncogenic activation may contribute to the loss of E-cadherin function in DGC without CDH1 mutation.

3. Targeting RTKs for Peritoneal Dissemination of DGC

In general, DGC is less sensitive to cytotoxic chemotherapy than IGC [3,47]. Although the anti-HER2 antibody, trastuzumab, in combination with chemotherapy improved the survival of patients with HER2-positive advanced IGC, its efficacy was limited in patients with DGC [48]. This is most likely because the rate of HER2 amplification was low in DGC. In contrast, dozens of RTK inhibitors targeting MET, FGFR2, and EGF exhibited efficacy against DGC harboring RTK gene amplification, at least in vitro and in preclinical models (Table 3).
We and others have shown that Met inhibitors, including PHA-665752, capmatinib, crizotinib, and E7050, exhibit remarkable antitumor and/or antiperitoneal dissemination activities against DGC cells that are positive for MET amplification in mouse xenograft models [8,12,54,55,56]. Some Met inhibitors, such as crizotinib, AMG 337, and Savolitinib, showed antitumor activity in patients with MET-amplified GC in clinical trials [8,51,64]. The monoclonal antibody, ABT-700, which disrupts Met dimerization, also showed efficacy against MET-amplified DGC [18]. Moreover, P3D12-vc-MMAF, which is a conjugate of a Met-degrading antibody and the tubulin inhibitor, MMAF, exhibited a drastic antitumor efficacy in DGC with MET amplification [59].
Gene alterations in the FGFR family (FGFR1, FGFR2, FGFR3, and FGFR4) have been reported in various cancers. Thus, pan-FGFR inhibitors have been developed and show antitumor effects against DGC in preclinical studies, and some of them have been tested in clinical trials [43,84]. As gene amplification of FGFR2 is the predominant FGFR alteration in DGC, the development of FGFR2-specific inhibitors may improve efficacy and reduce adverse effects. In this regard, a unique selective allosteric inhibitor of FGFR2, alofanib (RPT835), was reported [85]. Although its efficacy against DGC is currently unclear, clinical studies on alofanib in patients with advanced or metastatic GC are ongoing. Other therapeutic modalities targeting FGFR2, such as a bivalent degrader, DGY-09-192 [78], a neutralizing monoclonal antibody, PRO-007 [80], and an antibody–drug conjugate, BAY 1187982 [81], may have better efficacy and selectivity than that of small-molecule inhibitors.
Although DGC cells addicted to amplified RTK signaling firstly showed high sensitivity to the RTK inhibitors, resistant cells emerge upon their continuous exposure to RTK inhibitors. As this is a serious issue in clinical usage of RTK inhibitors, the underlying molecular mechanisms have been extensively studied. Coamplification of RTKs such as HER2 and/or EGFR contribute to therapeutic resistance to Met inhibitor in DGC harboring Met amplification [86]. A patient with concurrent MET and HER2 amplification responded to combined MET/HER2 inhibition. It was reported that FGFR2 overexpression is responsible for Met inhibitor resistance in MET-amplified PDX tumors of GC [28]. In this case, treatment with FGFR2 and Met inhibitors blocked the tumor growth. Overexpression of phosphoinositide 3-kinase (PI3K) p110α contributes to acquired resistance to Met inhibitor in DGC cells with MET amplification [83]. PI-103, a PI3K inhibitor, in combination with Met inhibitor showed antitumor effects against the Met-inhibitor-resistant DGC cells. Truncated forms of RAFs were also reported to confer resistance to Met inhibition in DGC cells with MET amplification [87]. FGFR2-ACSL5 fusion was found in a patient with FGFR2-amplified GC that acquired resistance to FGFR inhibitor [88]. Acquired resistance to FGFR inhibitor also occurs via PKC-mediated GSK3β phosphorylation in DGC-derived PDX tumors with FGFR2 amplification [89]. PKC inhibitors reversed the resistance of FGFR2-addicted tumors to FGFR inhibitors in vivo. Futibatinib, a unique irreversible pan-FGFR inhibitor that binds to the FGFR kinase domain, demonstrates antitumor activity against DGC with FGFR2 amplification [70,90]. Notably, futibatinib can inhibit FGFR2 mutants resistant to ATP-competitive FGFR inhibitors [70]. Taken together, the identification of responsible and druggable molecules and the development of inhibitors with different modes of action are critical to overcoming resistance to RTK inhibitors in RTK-addicted DGC.
RTK signaling plays a pivotal tumor-supporting role not only in cancer cells, but also in the tumor microenvironment. For example, vascular endothelial growth factor receptor (VEGFR) expressed in endothelial cells promotes tumor angiogenesis. Accordingly, dual- or multitarget tyrosine kinase inhibitors that inhibit both VEGFR in endothelial cells and other RTKs in cancer cells may have better therapeutic effects against RTK-addicted tumors. Several inhibitors targeting both VEGFR and Met or FGFR2 have been developed and tested in preclinical and clinical trials (Table 3). However, targeting multiple RTKs may increase the risk of adverse effects.

4. Novel Downstream Effectors of RTK in DGC

As described above, the use of RTK inhibitors inevitably results in acquired resistance in tumors. Therefore, understanding the downstream signaling of RTK is critical for the development of novel and alternative targeting approaches. PLEKHA5 (Pleckstrin homology domain containing A5) is a member of the PLEKHA family of proteins that contain a PH domain. Using a phosphoproteomic approach, we identified PLEKHA5 as a protein that is tyrosine-phosphorylated downstream of Met signaling (Figure 2) [91]. PLEKHA5 silencing selectively blocked the growth of DGC cells addicted to amplified Met, even when they acquired resistance to Met inhibitors. PLEKHA5 knockdown dysregulates glycolysis, leading to JNK activation and apoptotic cell death. In a mouse xenograft model, PLEKHA5 silencing markedly suppressed the peritoneal dissemination of MET-amplified DGC cells. Although its precise cellular functions remain to be elucidated, PLEKHA5 may be a biomarker for Met addiction, as well as a potential therapeutic target.
SHP2 (Src homology region 2 domain-containing phosphatase 2, also known as PTPN11, protein-tyrosine phosphatase nonreceptor type 11) is an oncogenic nonreceptor-type tyrosine phosphatase that regulates ERK activation downstream of RTK signaling [92]. Recurrent mutations in the PTPN11 gene encoding SHP2 have been observed in a variety of human cancers; therefore, SHP2 is thought to be a promising therapeutic target. Consequently, effective and selective allosteric SHP2 inhibitors, such as SHP099, have been developed recently [93]. We identified SHP2 by phosphoproteomic analysis of DGC cells with MET amplification [82] (Figure 2). SHP2 was tyrosine-phosphorylated in DGC cells with either MET or FGFR2 gene amplification. The growth of these cells was severely impaired by the knockdown or pharmacological inhibition of SHP2, even in Met-addicted DGC cells that acquired resistance to Met inhibitors. Furthermore, inhibition of SHP2 markedly suppressed the peritoneal dissemination of DGC cells harboring MET amplification. As SHP2 serves as a common signaling node downstream of multiple RTKs, targeting SHP2 may be an attractive alternative approach for the treatment of DGC with RTK amplification. As SHP2 is also involved in the immune checkpoint downstream of PD-1 [94], blockage of SHP2 may show additional efficacy through dual inhibition of RTK signaling and immune suppression.
Transferrin receptor 1 (TfR1) is a ubiquitously expressed membrane protein necessary for the cellular uptake of iron-loaded transferrin. TfR1 is upregulated in a variety of cancers and supports cancer cell growth by fulfilling an increased iron demand [95]. Shirakihara et al. recently identified TfR1 as a tyrosine-phosphorylated protein associated with FGFR2 in DGC harboring FGFR2 amplification [96] (Figure 2). TfR1 knockdown or FGFR2 inhibition impaired iron uptake and proliferation in DGC cells with FGFR2 amplification. Furthermore, TfR1 knockdown suppressed peritoneal metastasis of FGFR2-amplified DGC and improved survival in a mouse xenograft model. Thus, TfR1 plays a pivotal role in the oncogenic signaling of FGFR2; therefore, it may be a therapeutic target for FGFR2-addicted DGC. This finding provides a strong rationale for the clinical evaluation of TfR1 inhibitors, such as monoclonal antibodies against TfR1 [97].

5. Perspective

RTK amplification is a key event in the malignant progression of DGC, and it is a potential therapeutic target. However, several obstacles hinder the clinical application of RTK inhibitors in the treatment of DGC. For example, despite obvious antitumor effects in preclinical models, several clinical trials have shown insufficient benefits of RTK inhibitors in the treatment of DGC [98]. This is most likely owing to the lack of biomarkers to select patients with tumors that are addicted to RTK signaling. In addition to assessing the amount of RTK proteins or genes, evaluating the activation status of downstream effectors and/or gene expression signatures may be necessary to select RTK-addicted tumors. Detailed tumor genome profiling may enable the accurate identification of RTK-addicted tumors and patients who can benefit from RTK-targeting therapies [64].
From a biological perspective, understanding the precise oncogenic RTK functions is important for the development of effective therapies. For instance, it remains unclear whether the specificity of downstream signaling exists among the different RTKs. FGFR2 and Met may have specific and overlapping roles, which may not be shared by other RTKs, such as HER2, in tumor malignancies, according to their embryogenic and morphogenic functions. This may confer biological advantages to DGC cells by reinforcing their invasive and metastatic phenotypes. Additionally, it remains unclear why RTK activation mechanisms differ between cancer types. In the case of Met, point mutations are the major activation mechanisms in renal cancer, whereas gene amplification is predominant in DGC. Similarly, Ras gene amplification is more prevalent in DGC, whereas point mutations are dominant in lung and pancreatic cancers. These facts raise the possibility that the signaling downstream of RTK is different between activation patterns and/or is cell-context dependent. It is also critical to understand whether oncogenic RTK signaling caused by gene amplification elicits activation of specific downstream pathways. If downstream signaling differs between oncogenic and physiological RTK activation, targeting only oncogenic signaling would be an ideal therapeutic approach with minimal adverse effects. Thus, further detailed biological studies are necessary to develop effective targeted therapies against DGC harboring RTK gene amplification.

Author Contributions

Conceptualization, H.Y.; data curation, H.Y. and M.M.; funding acquisition, H.Y.; writing—original draft, H.Y.; writing—review and editing, H.Y., Y.N. and M.M. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by JSPS KAKENHI (18K07219 and 21K06879).

Acknowledgments

We apologize to researchers whose work was not cited due to space limitations.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  2. Lauren, P. The two histological main types of gastric carcinoma: Diffuse and so-called intestinal-type carcinoma. An attempt at a histo-clinical classification. Acta Pathol. Microbiol. Scand. 1965, 64, 31–49. [Google Scholar] [CrossRef] [PubMed]
  3. Ooki, A.; Yamaguchi, K. The dawn of precision medicine in diffuse-type gastric cancer. Ther. Adv. Med. Oncol. 2022, 14, 17588359221083049. [Google Scholar] [CrossRef]
  4. Yashiro, M.; Hirakawa, K. Cancer-stromal interactions in scirrhous gastric carcinoma. Cancer Microenviron. 2010, 3, 127–135. [Google Scholar] [CrossRef] [Green Version]
  5. Saraon, P.; Pathmanathan, S.; Snider, J.; Lyakisheva, A.; Wong, V.; Stagljar, I. Receptor tyrosine kinases and cancer: Oncogenic mechanisms and therapeutic approaches. Oncogene 2021, 40, 4079–4093. [Google Scholar] [CrossRef] [PubMed]
  6. Tsujino, T.; Yoshida, K.; Nakayama, H.; Ito, H.; Shimosato, T.; Tahara, E. Alterations of oncogenes in metastatic tumours of human gastric carcinomas. Br. J. Cancer 1990, 62, 226–230. [Google Scholar] [CrossRef] [Green Version]
  7. Tsugawa, K.; Yonemura, Y.; Hirono, Y.; Fushida, S.; Kaji, M.; Miwa, K.; Miyazaki, I.; Yamamoto, H. Amplification of the c-met, c-erbB-2 and epidermal growth factor receptor gene in human gastric cancers: Correlation to clinical features. Oncology 1998, 55, 475–481. [Google Scholar] [CrossRef] [PubMed]
  8. Lennerz, J.K.; Kwak, E.L.; Ackerman, A.; Michael, M.; Fox, S.B.; Bergethon, K.; Lauwers, G.Y.; Christensen, J.G.; Wilner, K.D.; Haber, D.A.; et al. MET amplification identifies a small and aggressive subgroup of esophagogastric adenocarcinoma with evidence of responsiveness to crizotinib. J. Clin. Oncol. 2011, 29, 4803–4810. [Google Scholar] [CrossRef] [Green Version]
  9. Deng, N.; Goh, L.K.; Wang, H.; Das, K.; Tao, J.; Tan, I.B.; Zhang, S.; Lee, M.; Wu, J.; Lim, K.H.; et al. 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. [Google Scholar] [CrossRef] [PubMed]
  10. The Cancer Genome Atlas Research Network. Comprehensive molecular characterization of gastric adenocarcinoma. Nature 2014, 513, 202–209. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Nagatsuma, A.K.; Aizawa, M.; Kuwata, T.; Doi, T.; Ohtsu, A.; Fujii, H.; Ochiai, A. Expression profiles of HER2, EGFR, MET and FGFR2 in a large cohort of patients with gastric adenocarcinoma. Gastric Cancer 2015, 18, 227–238. [Google Scholar] [CrossRef] [Green Version]
  12. Tanaka, Y.; Chiwaki, F.; Kojima, S.; Kawazu, M.; Komatsu, M.; Ueno, T.; Inoue, S.; Sekine, S.; Matsusaki, K.; Matsushita, H.; et al. Multi-omic profiling of peritoneal metastases in gastric cancer identifies molecular subtypes and therapeutic vulnerabilities. Nat. Camcer 2021, 2, 962–977. [Google Scholar] [CrossRef] [PubMed]
  13. 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. [Google Scholar] [CrossRef]
  14. Liu, Y.J.; Shen, D.; Yin, X.; Gavine, P.; Zhang, T.; Su, X.; Zhan, P.; Xu, Y.; Lv, J.; Qian, J.; et al. HER2, MET and FGFR2 oncogenic driver alterations define distinct molecular segments for targeted therapies in gastric carcinoma. Br. J. Cancer 2014, 110, 1169–1178. [Google Scholar] [CrossRef]
  15. Tajiri, R.; Ooi, A.; Fujimura, T.; Dobashi, Y.; Oyama, T.; Nakamura, R.; Ikeda, H. Intratumoral heterogeneous amplification of ERBB2 and subclonal genetic diversity in gastric cancers revealed by multiple ligation-dependent probe amplification and fluorescence in situ hybridization. Hum. Pathol. 2014, 45, 725–734. [Google Scholar] [CrossRef] [Green Version]
  16. Su, X.; Zhan, P.; Gavine, P.R.; Morgan, S.; Womack, C.; Ni, X.; Shen, D.; Bang, Y.J.; Im, S.A.; Ho Kim, W.; et al. FGFR2 amplification has prognostic significance in gastric cancer: Results from a large international multicentre study. Br. J. Cancer 2014, 110, 967–975. [Google Scholar] [CrossRef] [PubMed]
  17. Van Cutsem, E.; Bang, Y.J.; Feng-Yi, F.; Xu, J.M.; Lee, K.W.; Jiao, S.C.; Chong, J.L.; Lopez-Sanchez, R.I.; Price, T.; Gladkov, O.; et al. HER2 screening data from ToGA: Targeting HER2 in gastric and gastroesophageal junction cancer. Gastric Cancer 2015, 18, 476–484. [Google Scholar] [CrossRef]
  18. Wang, J.; Goetsch, L.; Tucker, L.; Zhang, Q.; Gonzalez, A.; Vaidya, K.S.; Oleksijew, A.; Boghaert, E.; Song, M.; Sokolova, I.; et al. Anti-c-Met monoclonal antibody ABT-700 breaks oncogene addiction in tumors with MET amplification. BMC Cancer 2016, 16, 105. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Phan, D.A.T.; Nguyen, V.T.; Hua, T.N.H.; Ngo, Q.D.; Doan, T.P.T.; Nguyen, S.T.; Thai, A.T.; Nguyen, V.T. HER2 status and its heterogeneity in gastric carcinoma of Vietnamese patient. J. Pathol. Transl. Med. 2017, 51, 396–402. [Google Scholar] [CrossRef]
  20. Rajadurai, P.; Fatt, H.K.; Ching, F.Y. Prevalence of HER2 positivity and its clinicopathological correlation in locally advanced/metastatic gastric cancer patients in Malaysia. J. Gastrointest. Cancer 2018, 49, 150–157. [Google Scholar] [CrossRef] [Green Version]
  21. Kuniyasu, H.; Yasui, W.; Kitadai, Y.; Yokozaki, H.; Ito, H.; Tahara, E. Frequent amplification of the c-met gene in scirrhous type stomach cancer. Biochem. Biophys. Res. Commun. 1992, 189, 227–232. [Google Scholar] [CrossRef]
  22. Hara, T.; Ooi, A.; Kobayashi, M.; Mai, M.; Yanagihara, K.; Nakanishi, I. Amplification of c-myc, K-sam, and c-met in gastric cancers: Detection by fluorescence in situ hybridization. Lab. Investig. 1998, 78, 1143–1153. [Google Scholar] [PubMed]
  23. Graziano, F.; Galluccio, N.; Lorenzini, P.; Ruzzo, A.; Canestrari, E.; D’Emidio, S.; Catalano, V.; Sisti, V.; Ligorio, C.; Andreoni, F.; et al. Genetic activation of the MET pathway and prognosis of patients with high-risk, radically resected gastric cancer. J. Clin. Oncol. 2011, 29, 4789–4795. [Google Scholar] [CrossRef]
  24. Janjigian, Y.Y.; Tang, L.H.; Coit, D.G.; Kelsen, D.P.; Francone, T.D.; Weiser, M.R.; Jhanwar, S.C.; Shah, M.A. MET expression and amplification in patients with localized gastric cancer. Cancer Epidemiol. Biomarkers Prev. 2011, 20, 1021–1027. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Lee, J.; Seo, J.W.; Jun, H.J.; Ki, C.S.; Park, S.H.; Park, Y.S.; Lim, H.Y.; Choi, M.G.; Bae, J.M.; Sohn, T.S.; et al. Impact of MET amplification on gastric cancer: Possible roles as a novel prognostic marker and a potential therapeutic target. Oncol. Rep. 2011, 25, 1517–1524. [Google Scholar] [CrossRef] [PubMed]
  26. Kawakami, H.; Okamoto, I.; Arao, T.; Okamoto, W.; Matsumoto, K.; Taniguchi, H.; Kuwata, K.; Yamaguchi, H.; Nishio, K.; Nakagawa, K.; et al. MET amplification as a potential therapeutic target in gastric cancer. Oncotarget 2012, 4, 9–17. [Google Scholar] [CrossRef] [Green Version]
  27. Shi, J.; Yao, D.; Liu, W.; Wang, N.; Lv, H.; He, N.; Shi, B.; Hou, P.; Ji, M. Frequent gene amplification predicts poor prognosis in gastric cancer. Int. J. Mol. Sci. 2012, 13, 4714–4726. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Liu, K.; Song, X.; Zhu, M.; Ma, H. Overexpression of FGFR2 contributes to inherent resistance to MET inhibitors in MET-amplified patient-derived gastric cancer xenografts. Oncol. Lett. 2015, 10, 2003–2008. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Peng, Z.; Li, Z.; Gao, J.; Lu, M.; Gong, J.; Tang, E.T.; Oliner, K.S.; Hei, Y.J.; Zhou, H.; Shen, L. Tumor MET expression and gene amplification in Chinese patients with locally advanced or metastatic gastric or gastroesophageal junction cancer. Mol. Cancer Ther. 2015, 14, 2634–2641. [Google Scholar] [CrossRef] [Green Version]
  30. Liao, H.; Tian, T.; Sheng, Y.; Peng, Z.; Li, Z.; Wang, J.; Li, Y.; Zhang, C.; Gao, J. The significance of MET expression and strategies of targeting MET treatment in advanced gastric cancer. Front. Oncol. 2021, 11, 719217. [Google Scholar] [CrossRef]
  31. Nakatani, H.; Sakamoto, H.; Yoshida, T.; Yokota, J.; Tahara, E.; Sugimura, T.; Terada, M. Isolation of an amplified DNA sequence in stomach cancer. Jpn. J. Cancer Res. 1990, 81, 707–710. [Google Scholar] [CrossRef]
  32. Jung, E.J.; Jung, E.J.; Min, S.Y.; Kim, M.A.; Kim, W.H. Fibroblast growth factor receptor 2 gene amplification status and its clinicopathologic significance in gastric carcinoma. Hum. Pathol. 2012, 43, 1559–1566. [Google Scholar] [CrossRef] [PubMed]
  33. Xie, L.; Su, X.; Zhang, L.; Yin, X.; Tang, L.; Zhang, X.; Xu, Y.; Gao, Z.; Liu, K.; Zhou, M.; et al. FGFR2 gene amplification in gastric cancer predicts sensitivity to the selective FGFR inhibitor AZD4547. Clin. Cancer Res. 2013, 19, 2572–2583. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Betts, G.; Valentine, H.; Pritchard, S.; Swindell, R.; Williams, V.; Morgan, S.; Griffiths, E.A.; 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. [Google Scholar] [CrossRef]
  35. Kim, S.T.; Jang, H.L.; Lee, S.J.; Lee, J.; Choi, Y.L.; Kim, K.M.; Cho, J.; Park, S.H.; Park, Y.S.; Lim, H.Y.; et al. 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Han, N.; Kim, M.A.; Lee, H.S.; Kim, W.H. Evaluation of fibroblast growth factor receptor 2 expression, heterogeneity and clinical significance in gastric cancer. Pathobiology 2015, 82, 269–279. [Google Scholar] [CrossRef] [Green Version]
  37. Ahn, S.; Lee, J.; Hong, M.; Kim, S.T.; Park, S.H.; Choi, M.G.; Lee, J.H.; Sohn, T.S.; Bae, J.M.; Kim, S.; et al. FGFR2 in gastric cancer: Protein overexpression predicts gene amplification and high H-index predicts poor survival. Mod. Pathol. 2016, 29, 1095–1103. [Google Scholar] [CrossRef]
  38. Teles, S.P.; Oliveira, P.; Ferreira, M.; Carvalho, J.; Ferreira, P.; Oliveira, C. Integrated analysis of structural variation and RNA expression of FGFR2 and its splicing modulator ESRP1 highlight the ESRP1(amp)-FGFR2(norm)-FGFR2-IIIc(high) axis in diffuse gastric cancer. Cancers 2019, 12, 70. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Schrumpf, T.; Behrens, H.M.; Haag, J.; Kruger, S.; Rocken, C. FGFR2 overexpression and compromised survival in diffuse-type gastric cancer in a large central European cohort. PLoS ONE 2022, 17, e0264011. [Google Scholar] [CrossRef]
  40. Fu, J.; Su, X.; Li, Z.; Deng, L.; Liu, X.; Feng, X.; Peng, J. HGF/c-MET pathway in cancer: From molecular characterization to clinical evidence. Oncogene 2021, 40, 4625–4651. [Google Scholar] [CrossRef]
  41. Trusolino, L.; Bertotti, A.; Comoglio, P.M. MET signalling: Principles and functions in development, organ regeneration and cancer. Nat. Rev. Mol. Cell Biol. 2010, 11, 834–848. [Google Scholar] [CrossRef] [PubMed]
  42. Recondo, G.; Che, J.; Janne, P.A.; Awad, M.M. Targeting MET dysregulation in cancer. Cancer Discov. 2020, 10, 922–934. [Google Scholar] [CrossRef] [PubMed]
  43. Yashiro, M.; Matsuoka, T. Fibroblast growth factor receptor signaling as therapeutic targets in gastric cancer. World J. Gastroenterol. 2016, 22, 2415–2423. [Google Scholar] [CrossRef]
  44. 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. [Google Scholar] [CrossRef] [Green Version]
  45. Guilford, P.; Hopkins, J.; Harraway, J.; McLeod, M.; McLeod, N.; Harawira, P.; Taite, H.; Scoular, R.; Miller, A.; Reeve, A.E. E-cadherin germline mutations in familial gastric cancer. Nature 1998, 392, 402–405. [Google Scholar] [CrossRef] [PubMed]
  46. Graziano, F.; Humar, B.; Guilford, P. The role of the E-cadherin gene (CDH1) in diffuse gastric cancer susceptibility: From the laboratory to clinical practice. Ann. Oncol. 2003, 14, 1705–1713. [Google Scholar] [CrossRef]
  47. Wang, K.; Li, E.; Busuttil, R.A.; Kong, J.C.; Pattison, S.; Sung, J.J.Y.; Yu, J.; El-Omar, E.M.; Simpson, J.A.; Boussioutas, A. A cohort study and meta-analysis of the evidence for consideration of Lauren subtype when prescribing adjuvant or palliative chemotherapy for gastric cancer. Ther. Adv. Med. Oncol. 2020, 12, 1758835920930359. [Google Scholar] [CrossRef] [PubMed]
  48. Bang, Y.J.; Van Cutsem, E.; Feyereislova, A.; Chung, H.C.; Shen, L.; Sawaki, A.; Lordick, F.; Ohtsu, A.; Omuro, Y.; Satoh, T.; et al. 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. [Google Scholar] [CrossRef]
  49. Kim, J.; Park, K.E.; Jeong, Y.S.; Kim, Y.; Park, H.; Nam, J.H.; Jung, K.; Son, W.S.; Jung, H.S.; Lee, J.H.; et al. Therapeutic efficacy of ABN401, a highly potent and selective MET inhibitor, based on diagnostic biomarker test in MET-addicted cancer. Cancers 2020, 12, 1575. [Google Scholar] [CrossRef] [PubMed]
  50. Hughes, P.E.; Rex, K.; Caenepeel, S.; Yang, Y.; Zhang, Y.; Broome, M.A.; Kha, H.T.; Burgess, T.L.; Amore, B.; Kaplan-Lefko, P.J.; et al. In vitro and in vivo activity of AMG 337, a potent and selective MET kinase inhibitor, in MET-dependent cancer models. Mol. Cancer Ther. 2016, 15, 1568–1579. [Google Scholar] [CrossRef] [Green Version]
  51. Van Cutsem, E.; Karaszewska, B.; Kang, Y.K.; Chung, H.C.; Shankaran, V.; Siena, S.; Go, N.F.; Yang, H.; Schupp, M.; Cunningham, D. A multicenter phase II study of AMG 337 in patients with MET-amplified gastric/gastroesophageal junction/esophageal adenocarcinoma and other MET-amplified solid tumors. Clin. Cancer Res. 2019, 25, 2414–2423. [Google Scholar] [CrossRef] [Green Version]
  52. Kasai, S.; Kuwayama, N.; Motoo, Y.; Kawashima, A.; Matsumoto, K.; Yano, S.; Matsushima, K.; Yasumoto, K. Dual blockade of MET and VEGFR2 signaling pathways as a potential therapeutic maneuver for peritoneal carcinomatosis in scirrhous gastric cancer. Biochem. Biophys. Res. Commun. 2022, 600, 80–86. [Google Scholar] [CrossRef]
  53. Sohn, S.H.; Kim, B.; Sul, H.J.; Kim, Y.J.; Kim, H.S.; Kim, H.; Seo, J.B.; Koh, Y.; Zang, D.Y. INC280 inhibits Wnt/beta-catenin and EMT signaling pathways and its induce apoptosis in diffuse gastric cancer positive for c-MET amplification. BMC Res. Notes 2019, 12, 125. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Okamoto, W.; Okamoto, I.; Arao, T.; Kuwata, K.; Hatashita, E.; Yamaguchi, H.; Sakai, K.; Yanagihara, K.; Nishio, K.; Nakagawa, K. Antitumor action of the MET tyrosine kinase inhibitor crizotinib (PF-02341066) in gastric cancer positive for MET amplification. Mol. Cancer Ther. 2012, 11, 1557–1564. [Google Scholar] [CrossRef] [Green Version]
  55. Yamaguchi, H.; Takanashi, M.; Yoshida, N.; Ito, Y.; Kamata, R.; Fukami, K.; Yanagihara, K.; Sakai, R. Saracatinib impairs the peritoneal dissemination of diffuse-type gastric carcinoma cells resistant to Met and fibroblast growth factor receptor inhibitors. Cancer Sci. 2014, 105, 528–536. [Google Scholar] [CrossRef] [Green Version]
  56. Nakagawa, T.; Tohyama, O.; Yamaguchi, A.; Matsushima, T.; Takahashi, K.; Funasaka, S.; Shirotori, S.; Asada, M.; Obaishi, H. E7050: A dual c-Met and VEGFR-2 tyrosine kinase inhibitor promotes tumor regression and prolongs survival in mouse xenograft models. Cancer Sci. 2010, 101, 210–215. [Google Scholar] [CrossRef]
  57. 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. Investig. New Drugs 2012, 30, 1352–1360. [Google Scholar] [CrossRef]
  58. Kim, H.J.; Kang, S.K.; Kwon, W.S.; Kim, T.S.; Jeong, I.; Jeung, H.C.; Kragh, M.; Horak, I.D.; Chung, H.C.; Rha, S.Y. Forty-nine gastric cancer cell lines with integrative genomic profiling for development of c-MET inhibitor. Int. J. Cancer 2018, 143, 151–159. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Fujita, R.; Blot, V.; Wong, E.; Stewart, C.; Lieuw, V.; Richardson, R.; Banah, A.; Villicana, J.; Timmer, A.; Coronella, J.; et al. A novel non-agonist c-Met antibody drug conjugate with superior potency over a c-Met tyrosine kinase inhibitor in c-Met amplified and non-amplified cancers. Cancer Biol. Ther. 2020, 21, 549–559. [Google Scholar] [CrossRef]
  60. Smolen, G.A.; Sordella, R.; Muir, B.; Mohapatra, G.; Barmettler, A.; Archibald, H.; Kim, W.J.; Okimoto, R.A.; Bell, D.W.; Sgroi, D.C.; et al. Amplification of MET may identify a subset of cancers with extreme sensitivity to the selective tyrosine kinase inhibitor PHA-665752. Proc. Natl. Acad. Sci. USA 2006, 103, 2316–2321. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Burbridge, M.F.; Bossard, C.J.; Saunier, C.; Fejes, I.; Bruno, A.; Leonce, S.; Ferry, G.; Da Violante, G.; Bouzom, F.; Cattan, V.; et al. 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Jones, R.D.O.; Grondine, M.; Borodovsky, A.; San Martin, M.; DuPont, M.; D’Cruz, C.; Schuller, A.; Henry, R.; Barry, E.; Castriotta, L.; et al. A pharmacokinetic-pharmacodynamic model for the MET tyrosine kinase inhibitor, savolitinib, to explore target inhibition requirements for anti-tumour activity. Br. J. Pharmacol. 2021, 178, 600–613. [Google Scholar] [CrossRef]
  63. Gavine, P.R.; Ren, Y.; Han, L.; Lv, J.; Fan, S.; Zhang, W.; Xu, W.; Liu, Y.J.; Zhang, T.; Fu, H.; et al. Volitinib, a potent and highly selective c-Met inhibitor, effectively blocks c-Met signaling and growth in c-MET amplified gastric cancer patient-derived tumor xenograft models. Mol. Oncol. 2015, 9, 323–333. [Google Scholar] [CrossRef] [PubMed]
  64. Lee, J.; Kim, S.T.; Kim, K.; Lee, H.; Kozarewa, I.; Mortimer, P.G.S.; Odegaard, J.I.; Harrington, E.A.; Lee, J.; Lee, T.; et al. Tumor genomic profiling guides patients with metastatic gastric cancer to targeted treatment: The VIKTORY umbrella trial. Cancer Discov. 2019, 9, 1388–1405. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Toiyama, Y.; Yasuda, H.; Saigusa, S.; Matushita, K.; Fujikawa, H.; Tanaka, K.; Mohri, Y.; Inoue, Y.; Goel, A.; Kusunoki, M. Co-expression of hepatocyte growth factor and c-Met predicts peritoneal dissemination established by autocrine hepatocyte growth factor/c-Met signaling in gastric cancer. Int. J. Cancer 2012, 130, 2912–2921. [Google Scholar] [CrossRef] [PubMed]
  66. Kim, B.J.; Kim, Y.J.; Sohn, S.H.; Kim, B.; Sul, H.J.; Kim, H.S.; Zang, D.Y. Tivantinib inhibits the VEGF signaling pathway and induces apoptosis in gastric cancer cells with c-MET or VEGFA amplification. Investig. New Drugs 2020, 38, 1633–1640. [Google Scholar] [CrossRef]
  67. Okuno, T.; Yashiro, M.; Masuda, G.; Togano, S.; Kuroda, K.; Miki, Y.; Hirakawa, K.; Ohsawa, M.; Wanibuchi, H.; Ohira, M. Establishment of a new scirrhous gastric cancer cell line with FGFR2 overexpression, OCUM-14. Ann. Surg. Oncol. 2019, 26, 1093–1102. [Google Scholar] [CrossRef]
  68. Ran, K.; Zeng, J.; Wan, G.; He, X.; Feng, Z.; Xiang, W.; Wei, W.; Hu, X.; Wang, N.; Liu, Z.; et al. Design, synthesis and biological evaluations of a series of Pyrido[1,2-a]pyrimidinone derivatives as novel selective FGFR inhibitors. Eur. J. Med. Chem. 2021, 220, 113499. [Google Scholar] [CrossRef]
  69. Perera, T.P.S.; Jovcheva, E.; Mevellec, L.; Vialard, J.; De Lange, D.; Verhulst, T.; Paulussen, C.; Van De Ven, K.; King, P.; Freyne, E.; et al. Discovery and pharmacological characterization of JNJ-42756493 (erdafitinib), a functionally selective small-molecule FGFR family inhibitor. Mol. Cancer Ther. 2017, 16, 1010–1020. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Sootome, H.; Fujita, H.; Ito, K.; Ochiiwa, H.; Fujioka, Y.; Ito, K.; Miura, A.; Sagara, T.; Ito, S.; Ohsawa, H.; et al. Futibatinib is a novel irreversible FGFR 1-4 inhibitor that shows selective antitumor activity against FGFR-deregulated tumors. Cancer Res. 2020, 80, 4986–4997. [Google Scholar] [CrossRef] [PubMed]
  71. 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. [Google Scholar] [CrossRef] [PubMed]
  72. 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. [Google Scholar] [CrossRef]
  73. Zhao, G.; Li, W.Y.; Chen, D.; Henry, J.R.; Li, H.Y.; Chen, Z.; Zia-Ebrahimi, M.; Bloem, L.; Zhai, Y.; Huss, K.; et al. 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Hilberg, F.; Tontsch-Grunt, U.; Baum, A.; Le, A.T.; Doebele, R.C.; Lieb, S.; Gianni, D.; Voss, T.; Garin-Chesa, P.; Haslinger, C.; et al. Triple angiokinase inhibitor nintedanib directly inhibits tumor cell growth and induces tumor shrinkage via blocking oncogenic receptor tyrosine kinases. J. Pharmacol. Exp. Ther. 2018, 364, 494–503. [Google Scholar] [CrossRef] [PubMed]
  75. 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Gozgit, J.M.; Wong, M.J.; Moran, L.; Wardwell, S.; Mohemmad, Q.K.; Narasimhan, N.I.; Shakespeare, W.C.; Wang, F.; Clackson, T.; Rivera, V.M. Ponatinib (AP24534), a multitargeted pan-FGFR inhibitor with activity in multiple FGFR-amplified or mutated cancer models. Mol. Cancer Ther. 2012, 11, 690–699. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Jiang, X.F.; Dai, Y.; Peng, X.; Shen, Y.Y.; Su, Y.; Wei, M.M.; Liu, W.R.; Ding, Z.B.; Zhang, A.; Shi, Y.H.; et al. SOMCL-085, a novel multi-targeted FGFR inhibitor, displays potent anticancer activity in FGFR-addicted human cancer models. Acta Pharmacol. Sin. 2018, 39, 243–250. [Google Scholar] [CrossRef] [Green Version]
  78. Du, G.; Jiang, J.; Wu, Q.; Henning, N.J.; Donovan, K.A.; Yue, H.; Che, J.; Lu, W.; Fischer, E.S.; Bardeesy, N.; et al. Discovery of a potent degrader for fibroblast growth factor receptor 1/2. Angew. Chem. Int. Ed. Engl. 2021, 60, 15905–15911. [Google Scholar] [CrossRef]
  79. Xiang, H.; Chan, A.G.; Ahene, A.; Bellovin, D.I.; Deng, R.; Hsu, A.W.; Jeffry, U.; Palencia, S.; Powers, J.; Zanghi, J.; et al. Preclinical characterization of bemarituzumab, an anti-FGFR2b antibody for the treatment of cancer. MAbs 2021, 13, 1981202. [Google Scholar] [CrossRef]
  80. Kim, S.T.; Lee, I.K.; Rom, E.; Sirkis, R.; Park, S.H.; Park, J.O.; Park, Y.S.; Lim, H.Y.; Kang, W.K.; Kim, K.M.; et al. Neutralizing antibody to FGFR2 can act as a selective biomarker and potential therapeutic agent for gastric cancer with FGFR2 amplification. Am. J. Transl. Res. 2019, 11, 4508–4515. [Google Scholar]
  81. Sommer, A.; Kopitz, C.; Schatz, C.A.; Nising, C.F.; Mahlert, C.; Lerchen, H.G.; Stelte-Ludwig, B.; Hammer, S.; Greven, S.; Schuhmacher, J.; et al. Preclinical efficacy of the auristatin-based antibody-drug conjugate BAY 1187982 for the treatment of FGFR2-positive solid tumors. Cancer Res. 2016, 76, 6331–6339. [Google Scholar] [CrossRef] [Green Version]
  82. Nagamura, Y.; Miyazaki, M.; Nagano, Y.; Tomiyama, A.; Ohki, R.; Yanagihara, K.; Sakai, R.; Yamaguchi, H. SHP2 as a potential therapeutic target in diffuse-type gastric carcinoma addicted to receptor tyrosine kinase signaling. Cancers 2021, 13, 4309. [Google Scholar] [CrossRef] [PubMed]
  83. Ji, F.; Liu, X.; Wu, Y.; Fang, X.; Huang, G. Overexpression of PI3K p110alpha contributes to acquired resistance to MET inhibitor, in MET-amplified SNU-5 gastric xenografts. Drug Des. Dev. Ther. 2015, 9, 5697–5704. [Google Scholar] [CrossRef] [Green Version]
  84. Lengyel, C.G.; Hussain, S.; Seeber, A.; Jamil Nidhamalddin, S.; Trapani, D.; Habeeb, B.S.; Elfaham, E.; Mazher, S.A.; Seid, F.; Khan, S.Z.; et al. FGFR pathway inhibition in gastric cancer: The golden era of an old target? Life 2022, 12, 81. [Google Scholar] [CrossRef] [PubMed]
  85. Tsimafeyeu, I.; Ludes-Meyers, J.; Stepanova, E.; Daeyaert, F.; Khochenkov, D.; Joose, J.B.; Solomko, E.; Van Akene, K.; Peretolchina, N.; Yin, W.; et al. Targeting FGFR2 with alofanib (RPT835) shows potent activity in tumour models. Eur. J. Cancer 2016, 61, 20–28. [Google Scholar] [CrossRef] [PubMed]
  86. Kwak, E.L.; Ahronian, L.G.; Siravegna, G.; Mussolin, B.; Borger, D.R.; Godfrey, J.T.; Jessop, N.A.; Clark, J.W.; Blaszkowsky, L.S.; Ryan, D.P.; et al. Molecular heterogeneity and receptor coamplification drive resistance to targeted therapy in MET-amplified esophagogastric cancer. Cancer Discov. 2015, 5, 1271–1281. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Petti, C.; Picco, G.; Martelli, M.L.; Trisolini, E.; Bucci, E.; Perera, T.; Isella, C.; Medico, E. Truncated RAF kinases drive resistance to MET inhibition in MET-addicted cancer cells. Oncotarget 2015, 6, 221–233. [Google Scholar] [CrossRef] [Green Version]
  88. Kim, S.Y.; Ahn, T.; Bang, H.; Ham, J.S.; Kim, J.; Kim, S.T.; Jang, J.; Shim, M.; Kang, S.Y.; Park, S.H.; et al. Acquired resistance to LY2874455 in FGFR2-amplified gastric cancer through an emergence of novel FGFR2-ACSL5 fusion. Oncotarget 2017, 8, 15014–15022. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Lau, W.M.; Teng, E.; Huang, K.K.; Tan, J.W.; Das, K.; Zang, Z.; Chia, T.; Teh, M.; Kono, K.; Yong, W.P.; et al. Acquired resistance to FGFR inhibitor in diffuse-type gastric cancer through an AKT-independent PKC-mediated phosphorylation of GSK3beta. Mol. Cancer Ther. 2018, 17, 232–242. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Meric-Bernstam, F.; Bahleda, R.; Hierro, C.; Sanson, M.; Bridgewater, J.; Arkenau, H.T.; Tran, B.; Kelley, R.K.; Park, J.O.; Javle, M.; et al. Futibatinib, an irreversible FGFR1-4 inhibitor, in patients with advanced solid tumors harboring FGF/FGFR aberrations: A phase I dose-expansion study. Cancer Discov. 2022, 12, 402–415. [Google Scholar] [CrossRef]
  91. Nagamura, Y.; Miyazaki, M.; Nagano, Y.; Yuki, M.; Fukami, K.; Yanagihara, K.; Sasaki, K.; Sakai, R.; Yamaguchi, H. PLEKHA5 regulates the survival and peritoneal dissemination of diffuse-type gastric carcinoma cells with Met gene amplification. Oncogenesis 2021, 10, 25. [Google Scholar] [CrossRef] [PubMed]
  92. Matozaki, T.; Murata, Y.; Saito, Y.; Okazawa, H.; Ohnishi, H. Protein tyrosine phosphatase SHP-2: A proto-oncogene product that promotes Ras activation. Cancer Sci. 2009, 100, 1786–1793. [Google Scholar] [CrossRef]
  93. Chen, Y.N.; LaMarche, M.J.; Chan, H.M.; Fekkes, P.; Garcia-Fortanet, J.; Acker, M.G.; Antonakos, B.; Chen, C.H.; Chen, Z.; Cooke, V.G.; et al. Allosteric inhibition of SHP2 phosphatase inhibits cancers driven by receptor tyrosine kinases. Nature 2016, 535, 148–152. [Google Scholar] [CrossRef] [PubMed]
  94. Zhao, M.; Guo, W.; Wu, Y.; Yang, C.; Zhong, L.; Deng, G.; Zhu, Y.; Liu, W.; Gu, Y.; Lu, Y.; et al. SHP2 inhibition triggers anti-tumor immunity and synergizes with PD-1 blockade. Acta Pharm. Sin. B 2019, 9, 304–315. [Google Scholar] [CrossRef] [PubMed]
  95. Shen, Y.; Li, X.; Dong, D.; Zhang, B.; Xue, Y.; Shang, P. Transferrin receptor 1 in cancer: A new sight for cancer therapy. Am. J. Cancer Res. 2018, 8, 916–931. [Google Scholar] [PubMed]
  96. Shirakihara, T.; Yamaguchi, H.; Kondo, T.; Yashiro, M.; Sakai, R. Transferrin receptor 1 promotes the fibroblast growth factor receptor-mediated oncogenic potential of diffused-type gastric cancer. Oncogene 2022, 41, 2587–2596. [Google Scholar] [CrossRef] [PubMed]
  97. Candelaria, P.V.; Leoh, L.S.; Penichet, M.L.; Daniels-Wells, T.R. Antibodies targeting the transferrin receptor 1 (TfR1) as direct anti-cancer agents. Front. Immunol. 2021, 12, 607692. [Google Scholar] [CrossRef] [PubMed]
  98. Reddavid, R.; Dagatti, S.; Franco, C.; Puca, L.; Tomatis, M.; Corso, S.; Giordano, S.; Degiuli, M. Molecularly targeted therapies for gastric cancer. State of the art. Cancers 2021, 13, 4094. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Receptor tyrosine kinase (RTK) signaling and gene alterations found in diffuse-type gastric carcinoma (DGC). Upon ligand binding, RTK molecules dimerize and transphosphorylate, which in turn, recruit a variety of intracellular signaling proteins. For example, Grb2/Sos bind to phosphorylated RTK and activate Ras signaling for cell growth. Analogously, phosphoinositide 3-kinase (PI3K) is recruited to activated RTK and generates phosphatidylinositol-3,4,5-triphosphate (PIP3), which is counteracted by PTEN, and activates Akt signaling for cell survival. Activation of these signaling pathways contributes to peritoneal metastasis. Genes encoding RTK signaling components altered in DGC are shown in bold and in oblique characters. Genes amplified in DGC are highlighted in red.
Figure 1. Receptor tyrosine kinase (RTK) signaling and gene alterations found in diffuse-type gastric carcinoma (DGC). Upon ligand binding, RTK molecules dimerize and transphosphorylate, which in turn, recruit a variety of intracellular signaling proteins. For example, Grb2/Sos bind to phosphorylated RTK and activate Ras signaling for cell growth. Analogously, phosphoinositide 3-kinase (PI3K) is recruited to activated RTK and generates phosphatidylinositol-3,4,5-triphosphate (PIP3), which is counteracted by PTEN, and activates Akt signaling for cell survival. Activation of these signaling pathways contributes to peritoneal metastasis. Genes encoding RTK signaling components altered in DGC are shown in bold and in oblique characters. Genes amplified in DGC are highlighted in red.
Cancers 14 03750 g001
Figure 2. Cellular functions of PLEKHA5, SHP2, and Transferrin receptor 1 (TfR1 downstream of amplified RTKs in DGC. PLEKHA5 is tyrosine-phosphorylated downstream of amplified Met. Downregulation of PLEKHA5 induces apoptosis via JNK activation and blocks cell migration, invasion, and peritoneal metastasis in Met-addicted DGC cells. SHP2 is also tyrosine-phosphorylated downstream of amplified Met and FGFR. Inhibition of SHP2 blocks growth, migration, invasion, and peritoneal dissemination of Met-addicted DGC. TfR1 associates with FGFR2 and is tyrosine-phosphorylated. TfR1 promotes transferrin-mediated iron uptake, which is required for growth, survival, and peritoneal metastasis of FGFR2-addicted DGC cells.
Figure 2. Cellular functions of PLEKHA5, SHP2, and Transferrin receptor 1 (TfR1 downstream of amplified RTKs in DGC. PLEKHA5 is tyrosine-phosphorylated downstream of amplified Met. Downregulation of PLEKHA5 induces apoptosis via JNK activation and blocks cell migration, invasion, and peritoneal metastasis in Met-addicted DGC cells. SHP2 is also tyrosine-phosphorylated downstream of amplified Met and FGFR. Inhibition of SHP2 blocks growth, migration, invasion, and peritoneal dissemination of Met-addicted DGC. TfR1 associates with FGFR2 and is tyrosine-phosphorylated. TfR1 promotes transferrin-mediated iron uptake, which is required for growth, survival, and peritoneal metastasis of FGFR2-addicted DGC cells.
Cancers 14 03750 g002
Table 1. Gene amplification of RTKs in gastric cancer (GC).
Table 1. Gene amplification of RTKs in gastric cancer (GC).
GeneSample (1)Frequency (%) (2)Technique (3)Classification (4)Associated Phenotypes (5)Ref.
EGFREarly GC
Advanced GC
Metastatic GC
0/20 (0%)
1/69 (1.4%)
3/32 (9.3%)
Southern blot Metastatic tumor[6]
EGFRGC6/70 (8.5%)Slot blot>2-foldLarge tumor, advanced stage, poor survival[7]
EGFRGEC23/489 (4.7%)FISHEGFR/CEP7 > 2.2Squamous cell carcinoma, poor survival[8]
EGFRGC15/193 (7.7%)SNP arrayCNA [9]
EGFRGC17/293 (5.8%)SNP arrayCNA [10]
EGFRGC23/950 (2.4%)FISHEGFR/CEP7 ≥ 2 [11]
EGFRGC ascites4/98 (4.0%)WGSCNA > 5 × ploidy [12]
HER2Early GC
Advanced GC
Metastatic GC
0/20 (0%)
4/69 (5.7%)
8/32 (25%)
Southern blot Metastatic tumor[6]
HER2GC9/70 (12.8%)Slot blot>2-foldLymph node metastasis, poor survival[7]
HER2GC15/128 (11.7%)Southern blot>2-foldIGC, poor survival[13]
HER2GEC45/489 (9.2%)FISHHER2/CEP17 > 2.2 [8]
HER2GC14/193 (7.2%)SNP arrayCNAPoor survival[9]
HER2GC38/293 (12.9%)SNP arrayCNA [10]
HER2Chinese GC33/219 (15.0%)FISHHER2/CEP17 > 2 [14]
HER2GC51/475 (10.7%)FISHHER2/CEP17 > 2.2Differentiated[15]
HER2Chinese GC
Korean GC
30/204 (14.7%)
27/338 (7.9%)
FISHHER2/CEP17 ≥ 2 [16]
HER2GC90/950 (9.4%)FISHHER2/CEP17 ≥ 2 [11]
HER2GC/GEJC756/3280 (23.0%)FISHHER2/CEP17 ≥ 2IGC[17]
HER2Asian GC32/134 (23.8%)FISHHER2/CEP17 ≥ 29/32 have Met coamplification[18]
HER2GC33/208 (15.8%)FISH/SISHHER2/CEP17 ≥ 2IGC, differentiated, heterogeneity is associated with DGC[19]
HER2GC/GEC40/228 (17.5%)FISH [20]
HER2GC ascites5/98 (5.1%)WGSCNA > 5 × ploidy [12]
METGC cell line
Early GC
Advanced GC
SGC
6/11 (54.5%)
0/11 (0%)
15/64 (23.4%)
5/13 (38.4%)
Southern blot≥3-fold [21]
METGC6/154 (3.8%)FISH [22]
METGC7/70 (10%)Slot blot>2-foldInfiltrative invasion, peritoneal dissemination, poor survival[7]
METGC13/128 (10.1%)Southern blot>2-foldLymph node metastasis, poor survival[13]
METStage II/III GC21/216 (9.7%)qPCR≥5 copiesPoor survival[23]
METWestern GC0/38 (0%)FISHMET/CEP7 > 2 [24]
METGC100/472 (21.1%)qPCR>4 copiesPoor survival[25]
METGEC10/489 (2.0%)FISHMET/CEP7 > 2.2High-grade, advanced stages, poor survival[8]
METGC8/193 (4.1%)SNP arrayCNAPoor survival[9]
METGC
GC cell line
4/266 (1.5%)
3/11 (27.2%)
qPCR/FISH≥4 copies [26]
METGC39/128 (30.4%)qPCR≥4 copiesInvasion, poor survival[27]
METGC12/293 (4.1%)SNP arrayCNA [10]
METChinese GC12/196 (6.1%)FISHMET/CEP7 > 2Lymph node and distant metastasis, Poor survival[14]
METGC xenograft5/30 (16.6%)SNP arrayCNA [28]
METGC12/950 (1.2%)FISHMET/CEP7 ≥ 2 [11]
METChinese advanced or metastatic GC or GEJC8/113 (7.0%)FISHMET/CEP7 > 2DGC[29]
METAsian GC13/134 (9.7%)FISHMET/CEP7 ≥ 29/13 have HER2 coamplification[18]
METGC7/49 (14.2%)CISHMET/CEP7 ≥ 2 [30]
METGC ascites7/98 (7.1%)WGSCNA > 5 × ploidy [12]
FGFR2GC
GC xenograft
3/24 (12.5%)
2/13 (15.3%)
Southern blot [31]
FGFR2GC3/154 (1.9%)FISH [22]
FGFR2GC18/193 (9.3%)SNP arrayCNA [9]
FGFR2GC14/313 (4.4%)FISHFGFR2/CEP10 ≥ 2Invasion, metastasis, poor survival[32]
FGFR2Chinese GC
Chinese GC
Caucasian GC
3/131 (2.2%)
9/197 (4.5%)
7/97 (7.2%)
aCGH
FISH
log ratio > 0.8
FGFR2/CEP10 ≥ 2
[33]
FGFR2GC3/171 (1.7%)FISHFGFR2/CEP10 ≥ 2Poor survival[34]
FGFR2GC15/293 (5.1%)SNP arrayCNA [10]
FGFR2GC cell line
GC
4/38 (10.5%)
24/482 (4.9%)
FISH
qRT-PCR
FGFR2/CEP10 ≥ 2
> 4 copies
[35]
FGFR2Chinese GC10/198 (5.0%)FISHFGFR2/CEP10 > 2 [14]
FGFR2UK GC
Chinese GC
Korean GC
30/408 (7.3%)
9/197 (4.4%)
15/356 (4.2%)
FISHFGFR2/CEP10 ≥ 2Lymph node metastasis and poor survival[16]
FGFR2GC5/188 (2.6%)FISHFGFR2/CEP10 ≥ 2 [36]
FGFR2GC67/1974 (3.3%)FISHFGFR2/CEP10 > 2 [37]
FGFR2GC (TCGA)63/338 (18.6%)WGSCNA [38]
FGFR2GC ascites11/98 (11.2%)WGSCNA > 5 × ploidy [12]
FGFR2Non-Asian GC20/493 (4.0%)CISHFGFR2/CEP10 > 2 [39]
(1) GC, gastric cancer; GEC, gastroesophageal cancer; GEJC, gastroesophageal junction cancer; SGC, scirrhous gastric cancer; TCGA, The Cancer Genome Atlas. (2) Numbers denote positive cases/total cases. (3) aCGH, array comparative genomic hybridization; CISH, chromogenic in situ hybridization; FISH, fluorescence in situ hybridization; qPCR, quantitative polymerase chain reaction; SISH, silver in situ hybridization; SNP, single nucleotide polymorphism; WGS, whole-genome sequencing. (4) CEP, chromosome enumerating probe; CNA, copy number alteration. (5) DGC, diffuse-type gastric cancer; IGC, intestinal-type gastric cancer.
Table 2. Gene amplification of EGFR, ERBB2, MET, and FGFR2 in GC in publicly available datasets.
Table 2. Gene amplification of EGFR, ERBB2, MET, and FGFR2 in GC in publicly available datasets.
GeneDatasetAmplified/Total TumorsFrequency
EGFRICGC_TCGA202010/6814.7%
MSKCC20176/1006%
OrigiMed202023/8502.7%
TCGA_PanCancerAtlas_STAD23/4385.2%
MSK202116/3205%
TCGA201417/2935.8%
ERBB2ICGC_TCGA202015/6822.0%
MSKCC201718/10018%
OrigiMed202068/8508%
TCGA_PanCancerAtlas_STAD58/43813.2%
MSK202137/32011.5%
TCGA201438/29312.9%
METICGC_TCGA20206/688.8%
MSKCC20174/1004%
OrigiMed202019/8502.2%
TCGA_PanCancerAtlas_STAD12/4382.7%
MSK202111/3203.4%
TCGA201412/2934.0%
FGFR2ICGC_TCGA20202/682.9%
MSKCC20172/1002%
OrigiMed202046/8505.4%
TCGA_PanCancerAtlas_STAD19/4384.3%
MSK202112/3203.7%
TCGA201415/2935.1%
Gene-amplification analysis of the indicated stomach adenocarcinoma datasets was performed using the cBioPortal (https://www.cbioportal.org/, accessed on 18 May 2022).
Table 3. Drugs targeting RTK signaling that are effective in DGC harboring gene amplification of RTKs in vitro or in preclinical models.
Table 3. Drugs targeting RTK signaling that are effective in DGC harboring gene amplification of RTKs in vitro or in preclinical models.
DrugType (1)TargetInhibited Functions and PhenotypesRefs.
ABN401SMIMetCell growth, survival, tumor growth[49]
AMG 337SMIMetCell growth, survival, tumor growth[50,51]
CabozantinibSMIMet/VEGFR2Cell growth[52]
Capmatinib/ INC280SMIMetCell growth, peritoneal metastasis[12,53]
Crizotinib/PF-02341066SMIMet/ALKCell growth, survival, tumor growth[54,55]
E7050SMIMet/VEGFR2Cell growth, tumor growth, angiogenesis, peritoneal metastasis[56]
Foretinib/GSK1363089SMIMet/VEGFR/PDGFRβ/Tie-2/RON/AXLCell growth[57,58]
JNJ38877605SMIMetCell growth, survival[26,55]
PHA-665752SMIMetCell growth, survival, tumor growth, peritoneal metastasis, ascites formation[25,55,59,60]
S49076SMIMet/FGFR1-3/AXLCell growth, tumor growth[61]
Savolitinib/VolitinibSMIMetCell growth, tumor growth[30,62,63,64]
SGX523SMIMetCell growth, survival[26]
SU11274SMIMetCell growth, survival, migration, peritoneal metastasis[65]
Tivantinib/ARQ197SMIMetCell growth, survival[58,66]
ABT-700mAbMetCell growth, survival, tumor growth[18]
SAIT301mAbMetCell growth[58]
Sym015mAbMetCell growth[58]
P3D12-vc-MMAFADCMetCell survival, tumor growth[59]
AZD4547SMIFGFR1-3Cell growth, tumor growth[33,67]
Compound 23dSMIFGFR1-4Cell growth, survival, tumor growth[68]
DovitinibSMIFGFR/VEGFRCell growth, survival, tumor growth[9]
Erdafitinib/JNJ-42756493SMIFGFR1-4Cell growth, tumor growth[68,69]
FutibatinibSMIFGFR1-4Cell growth, tumor growth[70]
Infigratinib/BGJ398SMIFGFR1-3Cell growth, peritoneal metastasis[12,67]
Ki23057SMIFGFR1, 2/VEGFR/PDGFR/c-KitCell growth, survival, tumor growth, peritoneal metastasis, lymph node metastasis, ascites formation[71,72]
LY2874455SMIFGFR1-4Tumor growth[73]
NintedanibSMIFGFR1-3/VEGFR1-3/PDGFRα, βCell growth[74]
PazopanibSMIFGFR/VEGFR/PDGFR/c-KitCell growth, cell survival[35]
PD173074SMIFGFR1-3Cell growth, survival[35,55,75]
Ponatinib/AP24534SMIFGFR/Bcr-Abl/VEGFR/PDGFR/SrcCell growth, tumor growth[76]
SOMCL-085SMIFGFR/VEGFR/PDGFRCell growth, tumor growth[77]
DGY-09-192PROTACFGFR1, 2Cell growth[78]
BemarituzumabmAbFGFR2bCell growth, tumor growth[79]
PRO-007mAbFGFR2Cell growth, invasion[80]
BAY 1187982ADCFGFR2Tumor growth[81]
OsimertinibSMIEGFRCell growth[12]
SHP099SMISHP2Cell growth, migration, invasion, peritoneal metastasis, ascites formation[82]
PI-103SMIPI3KTumor growth[83]
(1) SMI, small molecule inhibitor; mAb, monoclonal antibody; ADC, antibody–drug conjugate; PROTAC, proteolysis targeting chimera.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Yamaguchi, H.; Nagamura, Y.; Miyazaki, M. Receptor Tyrosine Kinases Amplified in Diffuse-Type Gastric Carcinoma: Potential Targeted Therapies and Novel Downstream Effectors. Cancers 2022, 14, 3750. https://doi.org/10.3390/cancers14153750

AMA Style

Yamaguchi H, Nagamura Y, Miyazaki M. Receptor Tyrosine Kinases Amplified in Diffuse-Type Gastric Carcinoma: Potential Targeted Therapies and Novel Downstream Effectors. Cancers. 2022; 14(15):3750. https://doi.org/10.3390/cancers14153750

Chicago/Turabian Style

Yamaguchi, Hideki, Yuko Nagamura, and Makoto Miyazaki. 2022. "Receptor Tyrosine Kinases Amplified in Diffuse-Type Gastric Carcinoma: Potential Targeted Therapies and Novel Downstream Effectors" Cancers 14, no. 15: 3750. https://doi.org/10.3390/cancers14153750

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop