Cancer Letters

Cancer Letters

Volume 225, Issue 1, 8 July 2005, Pages 1-26
Cancer Letters

Mini-review
c-Met as a target for human cancer and characterization of inhibitors for therapeutic intervention

https://doi.org/10.1016/j.canlet.2004.09.044Get rights and content

Abstract

Receptor tyrosine kinase (RTK) targeted agents such as trastuzumab, imatinib, bevacizumab, and gefitinib inhibitors have illustrated the utility of targeting this protein class for treatment of selected cancers. A unique member of the RTK family, c-Met, also represents an intriguing target for cancer therapy that is yet to be explored in a clinical setting. The proto-oncogene, c-Met, encodes the high-affinity receptor for hepatocyte growth factor (HGF) or scatter factor (SF). c-Met and HGF are each required for normal mammalian development and have been shown to be particularly important in cell migration, morphogenic differentiation, and organization of three-dimensional tubular structures (e.g. renal tubular cells, gland formation, etc.) as well as cell growth and angiogenesis. Both c-Met and HGF have been shown to be deregulated in and to correlate with poor prognosis in a number of major human cancers. New data describing the constitutive phosphorylation of c-Met in a number of human tumors is presented here along with a variety of mechanisms by which c-Met can become activated, including mutation and gene amplification. In support of the clinical data implicating c-Met activation in the pathogenesis of human cancers, introduction of c-Met and HGF (or mutant c-Met) into cells conferred the properties of motility, invasiveness, and tumorgenicity to the transformed cells. Conversely, the inhibition of c-Met with a variety of receptor antagonists inhibited the motility, invasiveness, and tumorgenicity of human tumor cell lines. Consistent with this observation, small-molecule inhibitors of c-Met were developed that antagonized c-Met/HGF-dependent phenotypes and tumor growth in mouse models. This review will address the potential for development of c-Met inhibitors for treatment of human cancers with particular emphasis on recent findings with small-molecule inhibitors.

Introduction

Receptor tyrosine kinases (RTKs) regulate many key processes in mammalian development, cell function, and tissue homeostasis. These diverse processes include cell growth and survival, organ morphogenesis, neovascularization, and tissue repair and regeneration, among others. Dysregulation of RTKs by mutation, gene rearrangement, gene amplification, and overexpression of both receptor and ligand have been implicated as causative factors in the development and progression of numerous human cancers. The validity of these targeted therapies is illustrated by the successes of imatinib targeting Bcr-Abl in chronic myelogenous leukemia and mutant c-Kit in gastrointestinal stromal tumors, trastuzumab in HER-2 overexpressing breast cancers, bevacizumab in colorectal carcinoma, and gefitinib in select non-small cell lung cancers [1].

c-Met is the prototypic member of a sub-family of RTKs which also includes RON. The c-Met RTK family is structurally distinct from other RTK families and is the only known high-affinity receptor for hepatocyte growth factor (HGF), also known as scatter factor (SF) [2], [3]. c-Met and HGF are widely expressed in a variety of tissues, and their expression is normally confined to cells of epithelial and mesenchymal origin, respectively [4], [5]. c-Met and HGF are each required for normal mammalian development and abnormalities associated with both Met- and HGF-null-mice are consistent with proximity of embryonic expression and epithelial–mesenchymal transition defects during organ morphogenesis [6], [7], [8]. Consistent with this finding, the transduction of signaling and subsequent biologic effects of HGF by c-Met has been shown to be important in epithelial–mesenchymal interaction and regulation of cell migration, invasion, cell proliferation and survival, angiogenesis, morphogenic differentiation, and organization of three-dimensional (3D) tubular structures (e.g. renal tubular cells, gland formation, etc.) during development and tissue repair [9], [10]. The specific consequence of c-Met signaling in a given cell (e.g. mitogenesis vs. morphogenesis) is dependent upon the cell type as well as the culture conditions of the experiment.

At a molecular level, binding of activated HGF to the c-Met extracellular ligand-binding domain results in receptor multimerization and phosphorylation of multiple tyrosine residues at the intracellular region. Tyrosine phosphorylation at the c-Met juxtamembrane, catalytic, and cytoplasmic tail domains regulate the internalization, catalytic activity, and docking of regulatory substrates, respectively [11], [12], [13], [14]. Activation of c-Met results in the binding and phosphorylation of adaptor proteins such as Gab-1, Grb2, Shc, and c-Cbl and subsequent activation of signal transducers such as PI-3-Kinase, PLC-γ, STATs, ERK 1 and 2, and FAK [15]. Although other RTKs signal through these pathways, c-Met-dependent signaling is distinct from other RTKs due to: (1) the presence of a unique multi-substrate docking site at the C-terminal region of the receptor; and (2) signaling through an adaptor protein unique to c-Met, called Gab-1, that has been demonstrated to mediate most biologically relevant Met-dependent signals [13], [16], [17]. The time dependence of signaling and spatial regulation of the c-Met signaling complex are distinct from other RTKs and have been proposed as key elements in distinguishing c-Met signaling and biologic function from other RTKs. An additional unique facet of c-Met signaling relative to other RTKs is its reported interaction with focal adhesion complexes and non-kinase binding partners such as β4 integrins, CD44, and semaphorins, which may further add to the complexity of regulation of cell function by this receptor [18], [19], [20].

The roles of docking proteins and signaling mediators in the regulation of diverse c-Met-dependent cell functions have been elucidated in studies using genetic, biological, and pharmacological means of selective modulation of these pathways. First, the involvement of the docking adaptor protein, Gab-1, has been shown to be required in the majority of c-Met-dependent biologic functions including mitogenesis, motility, and morphogenesis [15], [21]. Direct recruitment to a specific binding site at the C-terminal tail of the receptor resulting in sustained activity of Gab-1 has been demonstrated as unique to c-Met compared to other RTKs such as EGFR [22]. This Gab-1 interaction with c-Met has also been demonstrated to play a critical role in sustained signaling through other key adaptor and key signaling proteins [15], [21]. Downstream of Gab-1, the regulation of cell motility, cell disassociation, cell adhesion, and invasion by c-Met was shown to be dependent on both the Erk and PI3K pathways in experiments utilizing specific pharmacologic inhibitors of these pathways [23], [24], [25]. PI3K has also been shown to control cell c-Met-dependent cell survival through the Akt/PKB pathway while signaling through Erk controls mitogenesis suggesting that these two key pathways are critical for most biologic functions of c-Met [26], [27]. In addition to the PI3K and Erk pathways, directional cell migration and invasion were shown to require additional distinct pathways defined by Ras (via Cdc42, Rac1, and Paks), Crk family proteins, and c-src/focal adhesion kinase [28], [29], [30], [31], [32], [33], [34], [35], [36]. The most complex biologic response to HGF activation of c-Met, branching morphogenesis, requires the coordinate, time-dependent regulation of each of these aforementioned signaling pathways in addition to the involvement of STAT3 and PLCγ signaling [37], [38]. These signaling pathways are defined in the context of c-Met inhibition in Fig. 1.

While the controlled regulation of c-Met and HGF have been shown to be important in mammalian development, tissue maintenance, and repair; their dysregulation is implicated in the progression of cancers. Evidence linking c-Met and HGF as causative or progression factors in human cancers include: (1) the overexpression of both receptor and ligand in neoplasms relative to surrounding tissues in numerous indications; (2) the correlation of receptor and ligand overexpression with disease severity and outcome in multiple cancer types; (3) genetic alteration of c-Met by mutation of gene amplification in multiple cancer types; (4) introduction of c-Met and HGF (or mutant c-Met) into cell lines conferred the properties of tumorgenicity and metastatic propensity on engineered cells; (5) introduction of the c-Met or HGF as transgenes into the germline of mice resulted in the appearance of an array of primary and secondary neoplasms; and (6) the inhibition of c-Met or HGF function with dominant-negative receptors, antibody antagonists (both Met and HGF), and biologic antagonists (e.g. NK4) have reversed cancer associated phenotypes such as motility, invasion and proliferation of tumor cells and tumor growth and dissemination in vivo.

Recent reviews have comprehensively covered c-Met and HGF structure, biochemistry, signal transduction, and biologic function [39], [40], [41]. This review attempts to address the utility of targeting c-Met in human cancers, cancers where c-Met inhibition may be particularly effective and therapeutic approaches to targeting the c-Met/HGF pathway. The particular focus on therapeutic approaches will focus on recent data with small-molecule inhibitors of c-Met.

Section snippets

Altered regulation of c-Met

There are a number of mechanisms by which c-Met becomes dysregulated and activated in human cancers. These include overexpression and constitutive kinase activation in the presence and absence of gene amplification, both paracrine and autocrine activation of c-Met by HGF, and mutation of c-Met. c-Met is expressed by most carcinomas and its elevated expression relative to normal tissue has been detected in a number of cancers including lung, breast, colorectal, prostate, pancreatic, head and

Consequences of c-Met and HGF overexpression or mutation of c-Met

The mitogenic, invasive, and motogenic properties of HGF on numerous epithelial cell lines of a diverse array of tissue and tumor origins have been well established and are cell-type-dependent. It is believed that the dysregulation of cell proliferation and survival due to alteration of c-Met signaling may impact the growth of neoplasms while dysregulation of motility and invasion may affect the propensity of a given neoplasm to disseminate. The diverse set of cellular functions regulated by

Approaches to therapeutic inhibition of c-Met in human cancer and consequences of inhibition

Due to the vast information supporting the role of c-Met and HGF in the pathogenesis of human cancers along with successes of other RTK inhibitors, a number of approaches have been attempted to inhibit HGF- or c-Met-dependent signaling. These approaches include: (1) c-Met biologic inhibitors (ribozymes, dominant-negative receptors, decoy receptors, peptides); (2) HGF kringle variant antagonists; (3) HGF antagonist antibodies; (4) c-Met antagonist antibodies; and (5) small-molecule c-Met

Conclusions and future directions

The c-Met RTK is an exciting novel drug target due to the successes observed in clinical studies with other RTK inhibitors, its demonstrated role in experimental oncogenesis, and its dysregulation and correlation with disease prognosis in numerous cancers. The mutation or gene amplification of c-Met in selected clinical populations suggest that certain patients may be exquisitely sensitive to targeted therapy. Furthermore, the broad overexpression of both the ligand and receptor, their role in

References (224)

  • S. Pennacchietti et al.

    Hypoxia promotes invasive growth by transcriptional activation of the met protooncogene

    Cancer Cell.

    (2003)
  • H. Kuniyasu et al.

    Frequent amplification of the c-met gene in scirrhous type stomach cancer

    Biochem. Biophys. Res. Commun.

    (1992)
  • E. Gohda et al.

    Induction of hepatocyte growth factor in human skin fibroblasts by epidermal growth factor, platelet-derived growth factor and fibroblast growth factor

    Cytokine

    (1994)
  • M. Tamura et al.

    Enhancement of human hepatocyte growth factor production by interleukin-1 alpha and -1 beta and tumor necrosis factor-alpha by fibroblasts in culture

    J. Biol. Chem.

    (1993)
  • T. Moriyama et al.

    Concomitant expression of hepatocyte growth factor (HGF), HGF activator and c-met genes in human glioma cells in vitro

    Fed. Eur. Biochem. Soc. Lett.

    (1995)
  • M. Borset et al.

    Concomitant expression of hepatocyte growth factor/scatter factor and the receptor c-MET in human myeloma cell lines

    J. Biol. Chem.

    (1996)
  • V. Wallenius et al.

    Overexpression of the hepatocyte growth factor (HGF) receptor (Met) and presence of a truncated and activated intracellular HGF receptor fragment in locally aggressive/malignant human musculoskeletal tumors

    Am. J. Pathol.

    (2000)
  • M. Park et al.

    Mechanism of met oncogene activation

    Cell

    (1986)
  • M. Jo et al.

    Cross-talk between epidermal growth factor receptor and c-Met signal pathways in transformed cells

    J. Biol. Chem.

    (2000)
  • J. Tanyi et al.

    Evaluation of the tyrosine kinase domain of the Met proto-oncogene in sporadic ovarian carcinomas*

    Pathol. Oncol. Res.

    (1999)
  • M. Kong-Beltran et al.

    The Sema domain of Met is necessary for receptor dimerization and activation

    Cancer Cell

    (2004)
  • C. Seidel et al.

    Elevated serum concentrations of hepatocyte growth factor in patients with multiple myeloma. The Nordic Myeloma Study Group

    Blood

    (1998)
  • J.M. Siegfried et al.

    The clinical significance of hepatocyte growth factor for non-small cell lung cancer

    Ann. Thorac. Surg.

    (1998)
  • A.D. Laird et al.

    Small molecule tyrosine kinase inhibitors: clinical development of anticancer agents

    Expert. Opin. Investig. Drugs

    (2003)
  • L. Naldini et al.

    Scatter factor and hepatocyte growth factor are indistinguishable ligands for the MET receptor

    Eur. Mol. Biol. Org. J.

    (1991)
  • D.P. Bottaro et al.

    Identification of the hepatocyte growth factor receptor as the c-met proto-oncogene product

    Science

    (1991)
  • M.F. Di Renzo et al.

    Expression of the Met/HGF receptor in normal and neoplastic human tissues

    Oncogene

    (1991)
  • E. Sonnenberg et al.

    Scatter factor/hepatocyte growth factor and its receptor, the c-met tyrosine kinase, can mediate a signal exchange between mesenchyme and epithelia during mouse development

    J. Cell Biol.

    (1993)
  • F. Bladt et al.

    Essential role for the c-met receptor in the migration of myogenic precursor cells into the limb bud [see comment]

    Nature

    (1995)
  • C. Schmidt et al.

    Scatter factor/hepatocyte growth factor is essential for liver development

    Nature

    (1995)
  • I. Tsarfaty et al.

    The Met proto-oncogene mesenchymal to epithelial cell conversion

    Science

    (1994)
  • L. Naldini et al.

    The tyrosine kinase encoded by the MET proto-oncogene is activated by autophosphorylation

    Mol. Cell. Biol.

    (1991)
  • G.A. Rodrigues et al.

    Autophosphorylation modulates the kinase activity and oncogenic potential of the Met receptor tyrosine kinase

    Oncogene

    (1994)
  • T.M. Fournier et al.

    Cbl-transforming variants trigger a cascade of molecular alterations that lead to epithelial mesenchymal conversion

    Mol. Biol. Cell

    (2000)
  • K.A. Furge et al.

    Met receptor tyrosine kinase: enhanced signaling through adapter proteins

    Oncogene

    (2000)
  • M. Sachs et al.

    Essential role of Gab1 for signaling by the c-Met receptor in vivo

    J. Cell Biol.

    (2000)
  • K.M. Weidner et al.

    Interaction between Gab1 and the c-Met receptor tyrosine kinase is responsible for epithelial morphogenesis

    Nature

    (1996)
  • S. Giordano et al.

    The semaphorin 4D receptor controls invasive growth by coupling with Met

    Nat. Cell Biol.

    (2002)
  • C.R. Maroun et al.

    The Gab1 PH domain is required for localization of Gab1 at sites of cell–cell contact and epithelial morphogenesis downstream from the met receptor tyrosine kinase

    Mol. Cell. Biol.

    (1999)
  • R.M. Day et al.

    Differential signaling by alternative HGF isoforms through c-Met: activation of both MAP kinase and PI 3-kinase pathways is insufficient for mitogenesis

    Oncogene

    (1999)
  • K. Nakanishi et al.

    Hepatocyte growth factor promotes migration of human hepatocellular carcinoma via phosphatidylinositol 3-kinase

    Clin. Exp. Metastatis

    (1999)
  • S. Potempa et al.

    Activation of both MAP kinase and phosphatidylinositide 3-kinase by Ras is required for hepatocyte growth factor/scatter factor-induced adherens junction disassembly

    Mol. Biol. Cell

    (1998)
  • S. Fan 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)
  • G.H. Xiao 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. USA

    (2001)
  • I. Royal et al.

    Activation of cdc42, rac, PAK, and rho-kinase in response to hepatocyte growth factor differentially regulates epithelial cell colony spreading and dissociation

    Mol. Biol. Cell

    (2000)
  • A. Kodama et al.

    Involvement of Cdc42 small G protein in cell–cell adhesion, migration and morphology of MDCK cells

    Oncogene

    (1999)
  • A.J. Ridley et al.

    Regulation of scatter factor/hepatocyte growth factor responses by Ras, Rac, and Rho in MDCK cells

    Mol. Cell. Biol.

    (1995)
  • L. Lamorte et al.

    Crk associates with a multimolecular Paxillin/GIT2/beta-PIX complex and promotes Rac-dependent relocalization of Paxillin to focal contacts

    Mol. Biol. Cell

    (2003)
  • L. Lamorte et al.

    Crk adapter proteins promote an epithelial–mesenchymal-like transition and are required for HGF-mediated cell spreading and breakdown of epithelial adherens junctions

    Mol. Biol. Cell

    (2002)
  • L. Lamorte et al.

    A switch from p130Cas/Crk to Gab1/Crk signaling correlates with anchorage independent growth and JNK activation in cells transformed by the Met receptor oncoprotein

    Oncogene

    (2000)
  • Cited by (0)

    View full text