Open access

The Crosstalk of c-MET with Related Receptor Tyrosine Kinases in Urothelial Bladder Cancer

Written By

Sheng-Hui Lan, Shan-Ying Wu, Giri Raghavaraju, Nan-Haw Chow and Hsiao-Sheng Liu

Submitted: 28 August 2012 Published: 22 May 2013

DOI: 10.5772/53718

From the Edited Volume

Advances in the Scientific Evaluation of Bladder Cancer and Molecular Basis for Diagnosis and Treatment

Edited by Raj Persad and Weranja Ranasinghe

Chapter metrics overview

2,336 Chapter Downloads

View Full Metrics

1. Introduction

RTKs are often deregulated in human malignancies, contributing to cancer development and progression. Deregulation of RTKs leads to aberrant receptor activity resulting in increased cell proliferation, inhibition of apoptosis, invasion, and enhanced tumor metastases. Because RTKs are membrane proteins, they represent attractive targets for cancer therapy, with a number of agents already approved for clinical use.

c-MET gene, located on chromosome 7q21-q31, encodes a single precursor protein and is post-transcriptionally digested and glycosylated. The mature receptor is composed of a 50 kDa extracellular α-chain and a transmembrane 140 kDa β-chain, which are linked by disulfide bonds [1]. The MET β-chain contains homologous domains that shared with other proteins, including a semaphorin (Sema) domain, a PSI domain (in plexins, semaphorins and integrins), four IPT repeats (in immunoglobulins, plexins and transcription factors), a transmembrane domain, a juxtamembrane domain, a tyrosine kinase domain and a carboxy-terminal tail region [2, 3].

The transforming property of c-MET was initially described in a human osteosarcoma cell line after chemically induced mutagenesis [4]. In this in vitro model, c-MET was found to be constitutively activated by translocation at (1;7), resulting in fused sequences of c-MET gene on chromosome 7q31 to the translocated promoter region on chromosome 1q25 [5]. Since then, support for c-MET signaling in human carcinogenesis comes from data of the cell culture [6], mice [7, 8], and sporadic and hereditary forms of renal carcinoma, where germline and somatic missense mutations were identified in c-MET’s kinase domain [9, 10]. Furthermore, c-MET activity plays a significant role in promoting tumor invasion and metastasis [11, 12]. In summary, c-MET regulates embryonic development and play important roles in the carcinogenesis, tumor progression, and a variety of cellular processes, including migration, proliferation, morphogenesis, and angiogenesis [13, 14].

HGF is predominantly secreted by mesenchymal cells, and c-MET is widely expressed on the surface of epithelial cancer cells [15]. Homodimerization of c-MET after binding to HGR leads to transphosphorylation of cytoplasmic tyrosine kinase domain at two specific sites (Y1234 and Y1235) and activation of down-stream signaling [16]. These events are essential during embryogenesis, and also play a critical role in normal tissue homeostasis of the hepatocytes, renal tubule cells, and myoblasts [17].

The phosphorylation of two tyrosine residues within COOH terminus (Y1349 and Y1356) is necessary and sufficient to mediate biological effects induced by of the c-MET activation [18]. These two residues recruit a number of adapter proteins, including Gab1, Grb2, Shc and the p85 subunit of phosphatidylinositol-3 kinase (PI3K) [17]. The involvement of diverse effectors allows the activation of different downstream pathways, including PI3K-Akt signaling, Ras-mitogen-activated protein kinase (MAPK) pathways, signal transducer and activator of transcription proteins (STATs) and the nuclear factor-kB (NF-kB) complex [17]. These signaling pathways are important during embryogenesis and in normal tissue homeostasis, such as cell proliferation, differentiation, transformation, migration and apoptosis.

Accumulating data have demonstrated that crosstalk between c-MET and other RTKs may contribute to tumor progression in some of human cancers [19-21]. As a result, evaluation of c-MET expression status and its crosstalk partners of RTKs may identify a subset of c-MET-positive cancer patients who may require co-targeting therapy.

Advertisement

2. Role of c-MET in human cancers

Overexpression of c-MET has been reported in different subtypes of lung cancer, including adenocarcinoma (67%), carcinoid (60%), large cell carcinoma (57%), squamous cell carcinoma (57%), and small cell lung cancer (SCLC) (25%) [22]. In terms of functional activity, positive staining could be demonstrated in the subtypes of adenocarcinoma (44%), large cell carcinoma (86%), squamous cell carcinoma (71%), carcinoid (40%), and SCLC (100%), respectively, using antibody for phospho-c-MET at the Y1003 (c-Cbl binding site). On the other hand, positive staining was observed in 33% of adenocarcinomas, 57% of large cell carcinoma and 50% of SCLCs using antibody for autophosphorylation of c-MET at the Y1230/1234/1235 site [22]. Importantly, missense germ-line mutations in the tyrosine kinase domain of c-MET have been described in patients with hereditary papillary renal carcinoma [9]; whereas sporadic mutations in the tyrosine kinase, juxtamembrane, or semaphorin domains of c-MET have been detected in gastric cancer, HCC and SCLCs [23-25]. Concerning biologic significance, activation of HGF/MET signalling pathway was shown to promote cell invasiveness in vivo and trigger tumor metastases through angiogenic pathways [26]. In addition, amplification of c-MET has been detected in the carcinomas of the stomach, esophagus, and colorectum, non–small-cell lung cancer, and glioblastoma, and is usually associated with acquired resistance to anticancer drugs-gefitinib or erlotinib [27-32].

Altered HGF secretion was reported in both solid and hematologic malignancies. Both tumor and mesenchymal cells are responsible for increased HGF production, leading to paracrine and/or autocrine activation of c-MET by HGF [33, 34, 35]. The enhanced c-MET signaling is tumorigenic and could induce tumor metastasis in athymic nude mice [11]. As a result, HGF and/or c-MET overexpression were suggested to be a prognostic biomarker for cancer patients [36-38], although not all studies got the same conclusion [39, 40].

Advertisement

3. Role of c-MET-related RTKs in cancer

In addition to c-MET, coexpression of c-MET and related RTKs was shown to have prognostic relevance in some human cancers [41-45]. For example, RON and MET were overexpressed in 55 % and 56 % of human ovarian cancer, respectively, and 42 % of them have co-expression of RON and MET (P < 0.001) [41]. Coexpression of RON/MET was associated with more aggressive phenotype of node-negative breast cancer patients. The 10-year disease-free survival in RON-/MET- breast cancer is significantly higher than that of RON+/MET+ group (79.3 % vs. 11.8 %) [42]. Furthermore, both MET and EGF family receptors are overexpressed in different human cancers. Coexpression of c-MET and HER2 were observed in breast cancer and cholangiocarcinoma, and is usually associated with poor prognosis [43]. Similarly, coexpression of c-MET and HER2 could be detected in gastric cancer, and activation of c-MET further increases the resistance to EGFR inhibitor-Lapatinib [44, 45].

Advertisement

4. Overexpression of c-MET as a prognostic indicator for urothelial carcinoma of the bladder

High levels of c-MET expression have been correlated with metastatic progression of tumors and poor survival in patients with carcinomas of the breast, extrahepatic biliary tract, stomach, endometrum, liver, colorectum, and kidney [46-53]. c-MET was also reported to play a positive role in the tumorigenesis of human bladder [54, 55]. For example, expression of c-met mRNA tended to positively correlate with differentiation of cancer cell lines in the absence of point mutation [55]. Expression of Met was positively associated with histologic grade, stage classification, tumor size, and nodular tumor growth (P < 0.05, respectively), and is an independent indicators for poor long-term survival (P = 0.04) [55]. Furthermore, pY1349 c-Met was found to be a prognostic marker in predicting metastasis-free and survival of bladder cancer in a large cohort study of 133 non-metastatic specimens of bladder cancer [56]. Taken together, c-met proto-oncogene plays an important role in the progression of bladder carcinogenesis. Evaluation of Met expression could identify a subset of bladder cancer patients who may require a more intensive treatment targeting strategy.

Advertisement

5. The signaling pathway of c-MET

5.1. c-MET-related signaling pathways

The signaling for growth depends on RAS-MAPK signaling pathway and plays an essential role in morphogenesis and epithelial-to-mesenchymal transition that results from loss of intracellular adhesion via cadherins, focal adhesion kinase, and integrins, in association with alteration of cell shape [57]. Activation of HGF/c-MET axis prevents cell apoptosis through PI3 kinase and subsequent Akt signaling events [58-60]. The crosstalk of c-MET and PI3K-Akt pathway with RAS-MAPK pathway has been implicated in patient survival [61, 62].

5.2. Crosstalk with other membrane proteins or receptor tyrosine kinases

c-MET is known to interact with other membrane proteins on the cell surface [63], including laminin receptor-α6β4 integrin, semaphorin receptors of plexin B family, and v6 splice variant of hyaluronan receptor-CD44 [63, 64]. The crosstalk between c-MET and membrane proteins modulates the activation of both c-MET and its partners and allows for integration of signals present in the extracellular environment [65]. Crosstalk between c-MET and epidermal growth factor receptor (EGFR) has been implicated in several biological systems [66]. Furthermore, the crosstalk of c-MET with other RTKs regulates different physiological and/or pathological situations additively or synergistically. This interaction promotes trans-phosphorylation of kinase of each other by directly binding or transducing through their downstream signaling pathways indirectly. We review the potential role of c-MET and related RTKs, including RON, EGFR, Axl and platelet derived growth factor receptor-alpha (PDGFR-α), in urothelial carcinoma of the bladder, either independently or in combination in vivo (crosstalk) (Fig. 1).

Advertisement

6. RON

Recepteur d’Origine Nantais (RON) is a MET RTK subfamily member. Its ligand is macrophage-stimulating protein (MSP) which is expressed by renal tubular cells [67-69]. Activation of RON induces apoptotic resistance, superoxide anion production, and phagocytosis of macrophages through different molecules and related signaling pathways, i.e. Src, ERK, p38 and PI3K/AKT, which are related to tumorigenesis [70-72]. The crosstalk between c-MET and RON has been reported in different in vitro experimental models, and has been confirmed in the human cancers of the liver, ovary, breast and urinary bladder.

Heterodimerization plays a pivotal role in initiating the crosstalk and activation of related signal transduction pathways. Follenzi et al., showed that activated c-MET directly cross-phosphorylates RON, and c-MET/RON heterodimmer activates the catalytic region of c-MET at Y1234/Y1235 and RON at Y1238/Y1239, respectively (Figure 1A). Moreover, both signal transducer docking sites of c-MET at Y1349/Y1356 and RON at Y1353/Y1360 are generated for downstream signaling molecules. Mutation of RON suppresses the transforming phenotype induced by c-MET [73]. In contrast, RON is specifically trans-phosphorylated by MET, but not by EGFR or HER2; and MET-specific kinase inhibitors also suppress the phosphorylation of RON [41]. In addition to HGF, other cytokines, including EGF, interleukin-1, interleukin-6 and tumor necrosis factor alpha (TNF- α), are able to induce the expression of both MET and RON in HCC, suggesting that MET and RON are regulated by similar cytokine networks [42].

Overexpression of RON increases the growth, motility and anti-apoptosis of cancer cells in vitro [74]. In primary human bladder cancer, overexpression of RON is detected in 32.8 % of the tumors, and 23.3 % of these positive tumors also showed high levels of MET expression as well. In addition, co-expressed RON and MET was significantly associated with decreased overall survival (P= 0.005) or metastasis-free survival (P = 0.01) [74]. Overexpression of RON and MET seems to be a universal event in uroepithelial cells [75]. These data support the potential significance of RON/MET crosstalk, and the occurrence as a biomarker in selection of appropriate treatment strategy for cancer patients.

Advertisement

7. EGFR

The EGFR (HER1 or ErbB-1 in humans) belongs to RTKs of ErbB family which consists of EGFR, HER2/c-neu (ErbB-2), Her3 (ErbB-3) and Her4 (ErbB-4) four subfamily members. EGF is the ligand of EGFR [76]. EGFR signaling pathway participates in the growth and progression of urothelial cancers. Mutations affecting EGFR expression or activity may initiate a cascade of events leading to autonomous cell proliferation, migration, invasion and apoptosis inhibition, leading to tumor progression [77, 78].

The crosstalk between EGFR and MET has been reported during development and tumorigenesis. Cooperative action of MET and EGFR controls the number of nephron (the functional unit of the kidney) and maintains the duct morphology during kidney development [79]. Three underlying mechanisms of crosstalk between MET and RTK have been reported: (1) Trans-phophorylation and activation: Both RON and EGFR can bind with MET, and form heterodimeric receptor complex to activate both tyrosine kinases through trans-phosphorylation. The crosstalk of EGFR or RON with c-MET was confirmed by co-immunopreciptation assay (Figure 1A) [66, 80]; (2) c-MET activates EGFR through transcriptional activation of the ligand EGF: c-MET increases the production of EGF through Ras/Erk signaling-mediated promoter activation. EGF then is transported out of the cell to bind with EGFR in an autocrine or paracrine manner (Figure 1B) [81]; (3) EGFR activates c-MET through Ras/Erk MAPK signaling pathway to activate metalloproteinasea (TIMP)-3 which then cleavages the c-MET at ectodomain (Figure 1C). The truncated c-MET protein promotes the proliferation and cell transformation [82, 83].

Naik et al., reported that positive staining for EGFR, HER2 and EGF could be detected in 23%, 60% and 47% of primary bladder cancer specimens, respectively [84]. The HER2/neu gene amplification and protein overexpression were demonstrated in high grade, invasive bladder cancer [85]. Overexpression of EGFR/ERBB2 correlates with higher tumor grading/stage and poorer clinical outcome in bladder cancer patients [86, 87]. These evidences support the selection of EGFR as a molecular marker for diagnosis and/or prognosis of bladder carcinoma [88, 89]. Recently, EGFR inhibitor Iressa has shown a strong protective efficacy through cell cycle regulation in carcinogen induced rat bladder cancer model [90]. Therefore, EGFR, vascular endothelial growth factor (VEGF), mTOR and their-related signaling molecules are excellent therapeutic targets, in combination with cytotoxic chemotherapy, in the design of bladder cancer treatment [91]. Overexpression of RON and EGFR, as well as their crosstalk, has been reported in various human bladder cancer cell lines [74, 92]. It is noteworthy to clarify the potential of RTK co-targeting in the application of EGFR inhibitors in bladder cancer therapy.

Advertisement

8. AXL

AXL is a member of TAM RTK family, including AXL, Tyro3 and Merk. It has a unique structure of extracellular region that juxtaposes IgL and FNIII repeats [93, 94]. The protein S and Gas6 (growth-arrest-specific protein 6) are ligands for TAM receptor [95]. Gas6/AXL controls diverse cellular functions, including proliferation, survival, migration and anti-inflammation through different signaling pathways [96]. Gas6/AXL stimulates cell proliferation through MEK/Erk signaling pathway [97]. Gas6/AXL activates the PI3K/AKT and p38 signaling pathways to enhance the cell survival and migration, respectively [98, 99]. Gas6/AXL also suppresses Toll-like receptor and cytokine receptor signaling in innate immune cells through regulation of STAT1 [100, 101]. Overexpression of AXL has been reported in mesothelioma, NSCLC, breast carcinoma, and bladder cancer [20, 96, 102]. However, AXL can be activated by a ligand-independent manner when AXL interacts with adjacent cells in which AXL was overexpressed, suggesting that overexpression of AXL may be activated per se through auto-activation [103].

Advertisement

9. PDGFR-α

PDGF, a ligand of PDGFR-α and -β, results in auto-phosphorylation and signaling transduction of PDGFR [104]. PDGF/PDGFR signaling is involved in the development of various tissues, and is essential for epithelial-mesenchymal interaction during metamorphic skin remodeling, mesenchymal cell migration and proliferation [105]. In PDGF-α knock-out mice, neural tube and brain are abnormally accompanied by defect of the nervous system [106]. PDGF contributes to the development and progression of cancer by autocrine or paracrine signaling, and further promotes tumorigenesis through proliferation, angiogenesis and tumor stromal interaction [107].

In huamn uroepithelial cells, c-MET is frequently co-expressed with AXL, PDGFR-α, discoidin domain receptor tyrosine kinase 2 (DDR2), and/or insulin-like growth factor I receptor (IGF1R). Overexpression of AXL and PDGFR-α has been detected in various human cancers, and is associated with invasiveness and/or metastasis of carcinoma of the breast, kidney and bladder [20, 108, 109]. Overexpression of c-MET/PDGFR-α was demonstrated in all of 9 human bladder cancer cell lines tested [110]. We identified that both AXL and PDGFR may be c-MET related RTKs in a cDNA microarray analysis [20]. In sharp contrast to crosstalk between c-MET and RON or EGFR, both AXL and PDGFR do not directly bind with c-MET, and is transcriptionally activated by mitogen activated protein kinase/extracellular signal-regulated kinase (MEK/Erk) signaling pathway (Figure 1B) [20].

9.1. The relationship among environmental carcinogens, c-MET and RTKs

The environmental carcinogens, mainly from cigarette smoking, play important roles in the bladder cancer development, specifically urothelial carcinoma [111, 112]. Cigar smoking, pipe smoking, and secondhand smoke are implicated as risk factors for urothelial carcinoma. The incidence of urothelial cancer is approximately 4 times higher in smokers compared with non-smokers [113]. It is also reported that 50 % of all bladder cancers in men and 30 % in women are due to cigarette smoking [114]. All these evidences suggest that smoking is the most important risk factor for bladder cancer development. Genetic damage is the major cause of smoking-related cancer induction by which normal cellular pathways are altered to trigger cell growth and induce tumor formation [115]. In addition to bladder cancer, lung cancer formation is also induced by genetic modifications mostly caused by tobacco smoking [116]. Genetic mutations and amplifications in RTK related signaling, such as c-MET, EGFR, ErbB2, c-Kit, VEGFR, PI3K, and PTEN, contribute to lung cancer development by escaping from normal growth control and transforming into a malignant phenotype [117, 118]. Several autocrine loops, including stem cell factor (SCF)/c-Kit, IGF-I/IGF-IR, and HGF/c-MET, lead to the activation of PI3K/Akt signaling pathway and promote the cell growth, survival, and chemotherapy resistance in lung cancer. During lung cancer development, RTKs and their downstream effectors are selectively up-regulated. It is intriguing to clarify whether crosstalk of c-MET with RTKs in bladder cancer is also related to smoking. Altogether, it is noteworthy to clarify the relationship among smoking, c-MET, RTKs and bladder cancer development in the further study.

Advertisement

10. Conclusion and future direction

Overexpression of multiple RTKs has been reported in many human cancers, including bladder cancer. Cross-connection among individual signaling pathway activated by each RTK forms the signaling networks, which may complicate the development of anticancer strategies. With discussion above, more attention is focused to identify the prognostic targets and development of the targeted therapy for bladder cancer. In this review, we describe the current knowledge of interaction between c-MET and related RTKs. On the basis of complicated signaling network, the multimodal strategies should include systemic chemo- or biological therapies in combination with surgery and/or radiation applicable for invasive/metastatic bladder cancers [91]. Diverse therapeutic strategies have been developed to inhibit the HGF/c-MET signaling, including anti-HGF antibodies, HGF antagonists, anti-c-MET antibodies, and c-MET tyrosine kinase inhibitors. The c-MET pathway inhibitors have been reported to block the activities of other related tyrosine kinases. For example, MP470, a RAD51 inhibitor, suppresses the activity of c-MET and PDGFR [119]. MK-2461 suppresses the activity of both c-MET and RON [120]. BMS-777607 inhibits the activity of c-MET, RON and AXL [119, 121]. Furthermore, Foretinib, an oral multi-kinase inhibitor, inhibits the c-MET activity and its related RTKs (RON, EGFR, AXL and PDGFR) [122, 123]. Altogether, these inhibitors have potential to be used for bladder cancer therapy in the future. Cooperative action of c-MET with RON, EGFR, AXL and PDGFR-α has been reported to play important roles in bladder cancer progression, and thus deserves further investigation as the co-targeting therapy candidates. Understanding of the mechanisms underlying crosstalk of c-MET with RTKs is indispensible in the development of novel strategies against urothelial bladder cancer.

Figure 1.

The crosstalk between c-MET and related receptor tyrosine kinases

A. Trans-phosphorylation by other RTKs. The ligands, such as HGF, MSP and EGF, activate the MET, RON and EGFR, respectively, through tyrosine phosphorylation. The activated receptors (MET, RON or EGFR) cross talk with other RTKs through trans-phosphorylation. B. Activation of other RTKs by c-MET through transcriptional regulation. HGF activates the c-MET and downstream Ras/Erk signaling pathway through tyrosine phosphorylation. Expression of PDGFR, AXL and EGF was enhanced through transcriptional regulation. Overexpression of PDGFR and AXL enhances their binding with cognate ligands (PDGF and GAS6) and activation of their downstream signaling pathways. Overexpression of EGF further enhances the activity of EGFR in an autocrine or paracrine manner. C. Metalloproteinase cleavage regulates c-MET activation. EGF induces the phosphorylation of EGFR and activation of Ras/Erk signaling, and promotes the MET ectodomain shedding by cleavage of TIMP3 sensitive metalloproteinase.

Abbreviations

DDR2 (Discoidin domain receptor tyrosine kinase 2)

EGF (Epidermal growth factor)

EGFR (Epidermal growth factor receptor)

HCC (Hepatocellular carcinoma)

HGF (Hepatocyte growth factor)

HGFR (Hepatocyte growth factor receptor)

IGF1R (Insulin-like Growth Factor I Receptor)

IL-1 (Interleukin-1)

IL-6 (Interleukin-6)

MAPK (Mitogen-activated protein kinase)

MSP (Macrophage-stimulating protein)

NF-κB (Nuclear factor-κB)

PI3K (Phosphatidylinositol-3 kinase)

PDGFR (Platelet-derived growth factor receptor)

PTKs (Protein tyrosine kinases)

RON (Recepteur d’Origine Nantais)

RTKs (Receptor tyrosine kinases)

SCLCs (Small cell lung cancer cells)

STATs (Signal transducer and activator of transcription proteins)

TCC (Transitional cell carcinoma)

TIMPs (Tissue inhibitors of metalloproteinases)

TNF- α (Tumor necrosis factor alpha)

Acknowledgments

This review was supported by the grant from the National Science Council (NSC101-2320-B006-025-MY3)

References

  1. 1. Giordano, S., et al., Tyrosine kinase receptor indistinguishable from the c-met protein. Nature, 1989. 339(6220): p. 155-6.
  2. 2. Maestrini, E., et al., A family of transmembrane proteins with homology to the MET-hepatocyte growth factor receptor. Proc Natl Acad Sci U S A, 1996. 93(2): p. 674-8.
  3. 3. Sattler, M. and R. Salgia, c-Met and hepatocyte growth factor: potential as novel targets in cancer therapy. Curr Oncol Rep, 2007. 9(2): p. 102-8.
  4. 4. Cooper, C.S., et al., Molecular cloning of a new transforming gene from a chemically transformed human cell line. Nature, 1984. 311(5981): p. 29-33.
  5. 5. Park, M., et al., Sequence of MET protooncogene cDNA has features characteristic of the tyrosine kinase family of growth-factor receptors. Proc Natl Acad Sci U S A, 1987. 84(18): p. 6379-83.
  6. 6. Maulik, G., et al., Role of the hepatocyte growth factor receptor, c-Met, in oncogenesis and potential for therapeutic inhibition. Cytokine Growth Factor Rev, 2002. 13(1): p. 41-59.
  7. 7. Takayama, H., et al., Diverse tumorigenesis associated with aberrant development in mice overexpressing hepatocyte growth factor/scatter factor. Proc Natl Acad Sci U S A, 1997. 94(2): p. 701-6.
  8. 8. Wang, R., et al., Activation of the Met receptor by cell attachment induces and sustains hepatocellular carcinomas in transgenic mice. J Cell Biol, 2001. 153(5): p. 1023-34.
  9. 9. Schmidt, L., et al., Germline and somatic mutations in the tyrosine kinase domain of the MET proto-oncogene in papillary renal carcinomas. Nat Genet, 1997. 16(1): p. 68-73.
  10. 10. Schmidt, L., et al., Two North American families with hereditary papillary renal carcinoma and identical novel mutations in the MET proto-oncogene. Cancer Res, 1998. 58(8): p. 1719-22.
  11. 11. Rong, S., et al., Invasiveness and metastasis of NIH 3T3 cells induced by Met-hepatocyte growth factor/scatter factor autocrine stimulation. Proc Natl Acad Sci U S A, 1994. 91(11): p. 4731-5.
  12. 12. Ma, P.C., et al., c-Met: structure, functions and potential for therapeutic inhibition. Cancer Metastasis Rev, 2003. 22(4): p. 309-25.
  13. 13. Coyle, R.C., A. Latimer, and J.R. Jessen, Membrane-type 1 matrix metalloproteinase regulates cell migration during zebrafish gastrulation: evidence for an interaction with non-canonical Wnt signaling. Exp Cell Res, 2008. 314(10): p. 2150-62.
  14. 14. Birchmeier, C., et al., Met, metastasis, motility and more. Nat Rev Mol Cell Biol, 2003. 4(12): p. 915-25.
  15. 15. Prat, M., et al., The receptor encoded by the human c-MET oncogene is expressed in hepatocytes, epithelial cells and solid tumors. Int J Cancer, 1991. 49(3): p. 323-8.
  16. 16. Longati, P., et al., Tyrosines1234-1235 are critical for activation of the tyrosine kinase encoded by the MET proto-oncogene (HGF receptor). Oncogene, 1994. 9(1): p. 49-57.
  17. 17. Furge, K.A., Y.W. Zhang, and G.F. Vande Woude, Met receptor tyrosine kinase: enhanced signaling through adapter proteins. Oncogene, 2000. 19(49): p. 5582-9.
  18. 18. Ponzetto, C., et al., A multifunctional docking site mediates signaling and transformation by the hepatocyte growth factor/scatter factor receptor family. Cell, 1994. 77(2): p. 261-71.
  19. 19. Jung, K.H., B.H. Park, and S.S. Hong, Progress in cancer therapy targeting c-Met signaling pathway. Arch Pharm Res, 2012. 35(4): p. 595-604.
  20. 20. Yeh, C.Y., et al., Transcriptional activation of the Axl and PDGFR-alpha by c-Met through a ras- and Src-independent mechanism in human bladder cancer. BMC Cancer, 2011. 11: p. 139.
  21. 21. Tanizaki, J., et al., Differential roles of trans-phosphorylated EGFR, HER2, HER3, and RET as heterodimerisation partners of MET in lung cancer with MET amplification. Br J Cancer, 2011. 105(6): p. 807-13.
  22. 22. Ma, P.C., et al., Functional expression and mutations of c-Met and its therapeutic inhibition with SU11274 and small interfering RNA in non-small cell lung cancer. Cancer Res, 2005. 65(4): p. 1479-88.
  23. 23. Lee, J.H., et al., A novel germ line juxtamembrane Met mutation in human gastric cancer. Oncogene, 2000. 19(43): p. 4947-53.
  24. 24. Park, W.S., et al., Somatic mutations in the kinase domain of the Met/hepatocyte growth factor receptor gene in childhood hepatocellular carcinomas. Cancer Res, 1999. 59(2): p. 307-10.
  25. 25. Ma, P.C., et al., c-MET mutational analysis in small cell lung cancer: novel juxtamembrane domain mutations regulating cytoskeletal functions. Cancer Res, 2003. 63(19): p. 6272-81.
  26. 26. Zhang, Y.W., et al., Hepatocyte growth factor/scatter factor mediates angiogenesis through positive VEGF and negative thrombospondin 1 regulation. Proc Natl Acad Sci U S A, 2003. 100(22): p. 12718-23.
  27. 27. Smolen, G.A., 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 U S A, 2006. 103(7): p. 2316-21.
  28. 28. Miller, C.T., et al., Genomic amplification of MET with boundaries within fragile site FRA7G and upregulation of MET pathways in esophageal adenocarcinoma. Oncogene, 2006. 25(3): p. 409-18.
  29. 29. Umeki, K., G. Shiota, and H. Kawasaki, Clinical significance of c-met oncogene alterations in human colorectal cancer. Oncology, 1999. 56(4): p. 314-21.
  30. 30. Beroukhim, R., et al., Assessing the significance of chromosomal aberrations in cancer: methodology and application to glioma. Proc Natl Acad Sci U S A, 2007. 104(50): p. 20007-12.
  31. 31. Engelman, J.A., et al., MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science, 2007. 316(5827): p. 1039-43.
  32. 32. Bean, J., et al., MET amplification occurs with or without T790M mutations in EGFR mutant lung tumors with acquired resistance to gefitinib or erlotinib. Proc Natl Acad Sci U S A, 2007. 104(52): p. 20932-7.
  33. 33. Ferracini, R., et al., The Met/HGF receptor is over-expressed in human osteosarcomas and is activated by either a paracrine or an autocrine circuit. Oncogene, 1995. 10(4): p. 739-49.
  34. 34. Furge, K.A., et al., Suppression of Ras-mediated tumorigenicity and metastasis through inhibition of the Met receptor tyrosine kinase. Proc Natl Acad Sci U S A, 2001. 98(19): p. 10722-7.
  35. 35. Moghul, A., et al., Modulation of c-MET proto-oncogene (HGF receptor) mRNA abundance by cytokines and hormones: evidence for rapid decay of the 8 kb c-MET transcript. Oncogene, 1994. 9(7): p. 2045-52.
  36. 36. Shattuck, D.L., et al., Met receptor contributes to trastuzumab resistance of Her2-overexpressing breast cancer cells. Cancer Res, 2008. 68(5): p. 1471-7.
  37. 37. Siegfried, J.M., et al., Association of immunoreactive hepatocyte growth factor with poor survival in resectable non-small cell lung cancer. Cancer Res, 1997. 57(3): p. 433-9.
  38. 38. Sawada, K., et al., c-Met overexpression is a prognostic factor in ovarian cancer and an effective target for inhibition of peritoneal dissemination and invasion. Cancer Res, 2007. 67(4): p. 1670-9.
  39. 39. Nakamura, Y., et al., c-Met activation in lung adenocarcinoma tissues: an immunohistochemical analysis. Cancer Sci, 2007. 98(7): p. 1006-13.
  40. 40. Resnick, M.B., et al., Epidermal growth factor receptor, c-MET, beta-catenin, and p53 expression as prognostic indicators in stage II colon cancer: a tissue microarray study. Clin Cancer Res, 2004. 10(9): p. 3069-75.
  41. 41. Maggiora, P., et al., The RON and MET oncogenes are co-expressed in human ovarian carcinomas and cooperate in activating invasiveness. Exp Cell Res, 2003. 288(2): p. 382-9.
  42. 42. Lee, W.Y., et al., Prognostic significance of co-expression of RON and MET receptors in node-negative breast cancer patients. Clin Cancer Res, 2005. 11(6): p. 2222-8.
  43. 43. Lengyel, E., et al., C-Met overexpression in node-positive breast cancer identifies patients with poor clinical outcome independent of Her2/neu. Int J Cancer, 2005. 113(4): p. 678-82.
  44. 44. Chen, C.T., et al., MET Activation Mediates Resistance to Lapatinib Inhibition of HER2-Amplified Gastric Cancer Cells. Mol Cancer Ther, 2012. 11(3): p. 660-9.
  45. 45. Carneiro, F. and M. Sobrinho-Simoes, The prognostic significance of amplification and overexpression of c-met and c-erb B-2 in human gastric carcinomas. Cancer, 2000. 88(1): p. 238-40.
  46. 46. Ghoussoub, R.A., et al., Expression of c-met is a strong independent prognostic factor in breast carcinoma. Cancer, 1998. 82(8): p. 1513-20.
  47. 47. Hida, Y., et al., Clinical significance of hepatocyte growth factor and c-Met expression in extrahepatic biliary tract cancers. Oncol Rep, 1999. 6(5): p. 1051-6.
  48. 48. Nakajima, M., et al., The prognostic significance of amplification and overexpression of c-met and c-erb B-2 in human gastric carcinomas. Cancer, 1999. 85(9): p. 1894-902.
  49. 49. Taniguchi, K., et al., The relation between the growth patterns of gastric carcinoma and the expression of hepatocyte growth factor receptor (c-met), autocrine motility factor receptor, and urokinase-type plasminogen activator receptor. Cancer, 1998. 82(11): p. 2112-22.
  50. 50. Wagatsuma, S., et al., Tumor angiogenesis, hepatocyte growth factor, and c-Met expression in endometrial carcinoma. Cancer, 1998. 82(3): p. 520-30.
  51. 51. Ueki, T., et al., Expression of hepatocyte growth factor and its receptor c-met proto-oncogene in hepatocellular carcinoma. Hepatology, 1997. 25(4): p. 862-6.
  52. 52. Di Renzo, M.F., et al., Overexpression and amplification of the met/HGF receptor gene during the progression of colorectal cancer. Clin Cancer Res, 1995. 1(2): p. 147-54.
  53. 53. Natali, P.G., et al., Overexpression of the met/HGF receptor in renal cell carcinomas. Int J Cancer, 1996. 69(3): p. 212-7.
  54. 54. Joseph, A., et al., Expression of scatter factor in human bladder carcinoma. J Natl Cancer Inst, 1995. 87(5): p. 372-7.
  55. 55. Cheng, H.L., et al., Overexpression of c-met as a prognostic indicator for transitional cell carcinoma of the urinary bladder. A comparison with p53 nuclear accumulation. J Clin Oncol, 2002, 20(60): p. 1544-50.
  56. 56. Miyata, Y., et al., Phosphorylated hepatocyte growth factor receptor/c-Met is associated with tumor growth and prognosis in patients with bladder cancer: correlation with matrix metalloproteinase-2 and -7 and E-cadherin. Hum Pathol, 2009. 40(4): p. 496-504.
  57. 57. Boccaccio, C. and P.M. Comoglio, Invasive growth: a MET-driven genetic programme for cancer and stem cells. Nat Rev Cancer, 2006. 6(8): p. 637-45.
  58. 58. Fan, S., et al., The cytokine hepatocyte growth factor/scatter factor inhibits apoptosis and enhances DNA repair by a common mechanism involving signaling through phosphatidyl inositol 3' kinase. Oncogene, 2000. 19(18): p. 2212-23.
  59. 59. Derksen, P.W., et al., The hepatocyte growth factor/Met pathway controls proliferation and apoptosis in multiple myeloma. Leukemia, 2003. 17(4): p. 764-74.
  60. 60. Xiao, G.H., et al., Anti-apoptotic signaling by hepatocyte growth factor/Met via the phosphatidylinositol 3-kinase/Akt and mitogen-activated protein kinase pathways. Proc Natl Acad Sci U S A, 2001. 98(1): p. 247-52.
  61. 61. Zeng, Q., et al., Hepatocyte growth factor inhibits anoikis in head and neck squamous cell carcinoma cells by activation of ERK and Akt signaling independent of NFkappa B. J Biol Chem, 2002. 277(28): p. 25203-8.
  62. 62. Tulasne, D. and B. Foveau, The shadow of death on the MET tyrosine kinase receptor. Cell Death Differ, 2008. 15(3): p. 427-34.
  63. 63. Knudsen, B.S. and G. Vande Woude, Showering c-MET-dependent cancers with drugs. Curr Opin Genet Dev, 2008. 18(1): p. 87-96.
  64. 64. Comoglio, P.M., S. Giordano, and L. Trusolino, Drug development of MET inhibitors: targeting oncogene addiction and expedience. Nat Rev Drug Discov, 2008. 7(6): p. 504-16.
  65. 65. Migliore, C. and S. Giordano, Molecular cancer therapy: can our expectation be MET? Eur J Cancer, 2008. 44(5): p. 641-51.
  66. 66. Guo, A., et al., Signaling networks assembled by oncogenic EGFR and c-Met. Proc Natl Acad Sci U S A, 2008. 105(2): p. 692-7.
  67. 67. Ronsin, C., et al., A novel putative receptor protein tyrosine kinase of the met family. Oncogene, 1993. 8(5): p. 1195-202.
  68. 68. Wang, M.H., et al., Identification of the ron gene product as the receptor for the human macrophage stimulating protein. Science, 1994. 266(5182): p. 117-9.
  69. 69. Rampino, T., et al., Macrophage-stimulating protein is produced by tubular cells and activates mesangial cells. J Am Soc Nephrol, 2002. 13(3): p. 649-57.
  70. 70. Brunelleschi, S., et al., Macrophage stimulating protein (MSP) evokes superoxide anion production by human macrophages of different origin. Br J Pharmacol, 2001. 134(6): p. 1285-95.
  71. 71. Chen, Y.Q., J.H. Fisher, and M.H. Wang, Activation of the RON receptor tyrosine kinase inhibits inducible nitric oxide synthase (iNOS) expression by murine peritoneal exudate macrophages: phosphatidylinositol-3 kinase is required for RON-mediated inhibition of iNOS expression. J Immunol, 1998. 161(9): p. 4950-9.
  72. 72. Lutz, M.A. and P.H. Correll, Activation of CR3-mediated phagocytosis by MSP requires the RON receptor, tyrosine kinase activity, phosphatidylinositol 3-kinase, and protein kinase C zeta. J Leukoc Biol, 2003. 73(6): p. 802-14.
  73. 73. Follenzi, A., et al., Cross-talk between the proto-oncogenes Met and Ron. Oncogene, 2000. 19(27): p. 3041-9.
  74. 74. Cheng, H.L., et al., Co-expression of RON and MET is a prognostic indicator for patients with transitional-cell carcinoma of the bladder. Br J Cancer, 2005. 92(10): p. 1906-14.
  75. 75. Comperat, E., et al., Prognostic value of MET, RON and histoprognostic factors for urothelial carcinoma in the upper urinary tract. J Urol, 2008. 179(3): p. 868-72; discussion 872.
  76. 76. Herbst, R.S., Review of epidermal growth factor receptor biology. Int J Radiat Oncol Biol Phys, 2004. 59(2 Suppl): p. 21-6.
  77. 77. Mendelsohn, J., The epidermal growth factor receptor as a target for cancer therapy. Endocr Relat Cancer, 2001. 8(1): p. 3-9.
  78. 78. Kulik, G., A. Klippel, and M.J. Weber, Antiapoptotic signalling by the insulin-like growth factor I receptor, phosphatidylinositol 3-kinase, and Akt. Mol Cell Biol, 1997. 17(3): p. 1595-606.
  79. 79. Ishibe, S., et al., Met and the epidermal growth factor receptor act cooperatively to regulate final nephron number and maintain collecting duct morphology. Development, 2009. 136(2): p. 337-45.
  80. 80. Jo, M., et al., Cross-talk between epidermal growth factor receptor and c-Met signal pathways in transformed cells. J Biol Chem, 2000. 275(12): p. 8806-11.
  81. 81. Reznik, T.E., et al., Transcription-dependent epidermal growth factor receptor activation by hepatocyte growth factor. Mol Cancer Res, 2008. 6(1): p. 139-50.
  82. 82. Nath, D., et al., Shedding of c-Met is regulated by crosstalk between a G-protein coupled receptor and the EGF receptor and is mediated by a TIMP-3 sensitive metalloproteinase. J Cell Sci, 2001. 114(Pt 6): p. 1213-20.
  83. 83. Merlin, S., et al., Deletion of the ectodomain unleashes the transforming, invasive, and tumorigenic potential of the MET oncogene. Cancer Sci, 2009. 100(4): p. 633-8.
  84. 84. Naik, D.S., et al., Epidermal growth factor receptor expression in urinary bladder cancer. Indian J Urol, 2011. 27(2): p. 208-14.
  85. 85. Latif, Z., et al., HER2/neu gene amplification and protein overexpression in G3 pT2 transitional cell carcinoma of the bladder: a role for anti-HER2 therapy? Eur J Cancer, 2004. 40(1): p. 56-63.
  86. 86. Zheng, Y., et al., Dihydrotestosterone upregulates the expression of epidermal growth factor receptor and ERBB2 in androgen receptor-positive bladder cancer cells. Endocr Relat Cancer, 2011. 18(4): p. 451-64.
  87. 87. Jimenez, R.E., et al., Her-2/neu overexpression in muscle-invasive urothelial carcinoma of the bladder: prognostic significance and comparative analysis in primary and metastatic tumors. Clin Cancer Res, 2001. 7(8): p. 2440-7.
  88. 88. Neal, D.E. and K. Mellon, Epidermal growth factor receptor and bladder cancer: a review. Urol Int, 1992. 48(4): p. 365-71.
  89. 89. Kassouf, W., et al., Distinctive expression pattern of ErbB family receptors signifies an aggressive variant of bladder cancer. J Urol, 2008. 179(1): p. 353-8.
  90. 90. Lu, Y., et al., Modulation of gene expression and cell-cycle signaling pathways by the EGFR inhibitor gefitinib (Iressa) in rat urinary bladder cancer. Cancer Prev Res (Phila), 2012. 5(2): p. 248-59.
  91. 91. Vishnu, P., J. Mathew, and W.W. Tan, Current therapeutic strategies for invasive and metastatic bladder cancer. Onco Targets Ther, 2011. 4: p. 97-113.
  92. 92. Hsu, P.Y., et al., Collaboration of RON and epidermal growth factor receptor in human bladder carcinogenesis. J Urol, 2006. 176(5): p. 2262-7.
  93. 93. O'Bryan, J.P., et al., axl, a transforming gene isolated from primary human myeloid leukemia cells, encodes a novel receptor tyrosine kinase. Mol Cell Biol, 1991. 11(10): p. 5016-31.
  94. 94. Hafizi, S., et al., Interaction of Axl receptor tyrosine kinase with C1-TEN, a novel C1 domain-containing protein with homology to tensin. Biochem Biophys Res Commun, 2002. 299(5): p. 793-800.
  95. 95. Faust, M., et al., The murine ufo receptor: molecular cloning, chromosomal localization and in situ expression analysis. Oncogene, 1992. 7(7): p. 1287-93.
  96. 96. Linger, R.M., et al., Taking aim at Mer and Axl receptor tyrosine kinases as novel therapeutic targets in solid tumors. Expert Opin Ther Targets, 2010. 14(10): p. 1073-90.
  97. 97. Fridell, Y.W., et al., Differential activation of the Ras/extracellular-signal-regulated protein kinase pathway is responsible for the biological consequences induced by the Axl receptor tyrosine kinase. Mol Cell Biol, 1996. 16(1): p. 135-45.
  98. 98. Berclaz, G., et al., Estrogen dependent expression of the receptor tyrosine kinase axl in normal and malignant human breast. Ann Oncol, 2001. 12(6): p. 819-24.
  99. 99. Allen, M.P., et al., Novel mechanism for gonadotropin-releasing hormone neuronal migration involving Gas6/Ark signaling to p38 mitogen-activated protein kinase. Mol Cell Biol, 2002. 22(2): p. 599-613.
  100. 100. Sharif, M.N., et al., Twist mediates suppression of inflammation by type I IFNs and Axl. J Exp Med, 2006. 203(8): p. 1891-901.
  101. 101. Rothlin, C.V., et al., TAM receptors are pleiotropic inhibitors of the innate immune response. Cell, 2007. 131(6): p. 1124-36.
  102. 102. Ou, W.B., et al., AXL regulates mesothelioma proliferation and invasiveness. Oncogene, 2011. 30(14): p. 1643-52.
  103. 103. Bellosta, P., et al., The receptor tyrosine kinase ARK mediates cell aggregation by homophilic binding. Mol Cell Biol, 1995. 15(2): p. 614-25.
  104. 104. Farooqi, A.A., et al., PDGF: the nuts and bolts of signalling toolbox. Tumour Biol, 2011. 32(6): p. 1057-70.
  105. 105. Utoh, R., et al., Platelet-derived growth factor signaling as a cue of the epithelial-mesenchymal interaction required for anuran skin metamorphosis. Dev Dyn, 2003. 227(2): p. 157-69.
  106. 106. Fruttiger, M., et al., Defective oligodendrocyte development and severe hypomyelination in PDGF-A knockout mice. Development, 1999. 126(3): p. 457-67.
  107. 107. Yu, J., C. Ustach, and H.R. Kim, Platelet-derived growth factor signaling and human cancer. J Biochem Mol Biol, 2003. 36(1): p. 49-59.
  108. 108. Meric, F., et al., Expression profile of tyrosine kinases in breast cancer. Clin Cancer Res, 2002. 8(2): p. 361-7.
  109. 109. Chung, B.I., et al., Expression of the proto-oncogene Axl in renal cell carcinoma. DNA Cell Biol, 2003. 22(8): p. 533-40.
  110. 110. Black, P.C., et al., Sensitivity to epidermal growth factor receptor inhibitor requires E-cadherin expression in urothelial carcinoma cells. Clin Cancer Res, 2008. 14(5): p. 1478-86.
  111. 111. Kiriluk, K.J., et al., Bladder cancer risk from occupational and environmental exposures. Urol Oncol, 2012. 30(2): p. 199-211.
  112. 112. Wilhelm-Benartzi, C.S., et al., Association of secondhand smoke exposures with DNA methylation in bladder carcinomas. Cancer Causes Control, 2011. 22(8): p. 1205-13.
  113. 113. Burch, J.D., et al., Risk of bladder cancer by source and type of tobacco exposure: a case-control study. Int J Cancer, 1989. 44(4): p. 622-8.
  114. 114. Zeegers, M.P., et al., The impact of characteristics of cigarette smoking on urinary tract cancer risk: a meta-analysis of epidemiologic studies. Cancer, 2000. 89(3): p. 630-9.
  115. 115. Alberg, A.J. and J.R. Hebert, Cigarette smoking and bladder cancer: a new twist in an old saga? J Natl Cancer Inst, 2009. 101(22): p. 1525-6.
  116. 116. Wojtalla, A. and A. Arcaro, Targeting phosphoinositide 3-kinase signalling in lung cancer. Crit Rev Oncol Hematol, 2011. 80(2): p. 278-90.
  117. 117. Hodkinson, P.S., A. Mackinnon, and T. Sethi, Targeting growth factors in lung cancer. Chest, 2008. 133(5): p. 1209-16.
  118. 118. Pisick, E., S. Jagadeesh, and R. Salgia, Receptor tyrosine kinases and inhibitors in lung cancer. ScientificWorldJournal, 2004. 4: p. 589-604.
  119. 119. Liu, X., R.C. Newton, and P.A. Scherle, Development of c-MET pathway inhibitors. Expert Opin Investig Drugs, 2011. 20(9): p. 1225-41.
  120. 120. Pan, B.S., et al., MK-2461, a novel multitargeted kinase inhibitor, preferentially inhibits the activated c-Met receptor. Cancer Res, 2010. 70(4): p. 1524-33.
  121. 121. Schroeder, G.M., et al., Discovery of N-(4-(2-amino-3-chloropyridin-4-yloxy)-3-fluorophenyl)-4-ethoxy-1-(4-fluorophenyl )-2-oxo-1,2-dihydropyridine-3-carboxamide (BMS-777607), a selective and orally efficacious inhibitor of the Met kinase superfamily. J Med Chem, 2009. 52(5): p. 1251-4.
  122. 122. Qian, F., et al., Inhibition of tumor cell growth, invasion, and metastasis by EXEL-2880 (XL880, GSK1363089), a novel inhibitor of HGF and VEGF receptor tyrosine kinases. Cancer Res, 2009. 69(20): p. 8009-16.
  123. 123. Zillhardt, M., et al., Foretinib (GSK1363089), an orally available multikinase inhibitor of c-Met and VEGFR-2, blocks proliferation, induces anoikis, and impairs ovarian cancer metastasis. Clin Cancer Res, 2011. 17(12): p. 4042-51.

Written By

Sheng-Hui Lan, Shan-Ying Wu, Giri Raghavaraju, Nan-Haw Chow and Hsiao-Sheng Liu

Submitted: 28 August 2012 Published: 22 May 2013