Skip to main content
Therapeutic Advances in Medical Oncology logoLink to Therapeutic Advances in Medical Oncology
. 2011 Nov;3(1 Suppl):S21–S35. doi: 10.1177/1758834011422557

c-MET as a potential therapeutic target and biomarker in cancer

J Rafael Sierra 1, Ming-Sound Tsao 2,
Editor: Johann S de Bono3
PMCID: PMC3225018  PMID: 22128285

Abstract

The receptor tyrosine kinase c-MET and its ligand, hepatocyte growth factor (HGF), regulate multiple cellular processes that stimulate cell proliferation, invasion and angiogenesis. This review provides an overview of the evidence to support c-MET or the HGF/c-MET signaling pathway as relevant targets for personalized cancer treatment based on high frequencies of c-MET and/or HGF overexpression, activation, amplification in non-small cell lung carcinoma (NSCLC), gastric, ovarian, pancreatic, thyroid, breast, head and neck, colon and kidney carcinomas. Additionally, the current knowledge of small molecule inhibitors (tivantinib [ARQ 197]), c-MET/HGF antibodies (rilotumumab and MetMAb) and mechanisms of resistance to c-MET-targeted therapies are discussed.

Keywords: cancer, c-MET, hepatocyte growth factor, personalized medicine, targeted therapy

Introduction

The MET proto-oncogene encodes for the receptor tyrosine kinase (RTK), c-MET. Cells of epithelial–endothelial origin widely express c-MET, where it is essential for embryonic development [Brand-Saberi et al. 1996; Heymann et al. 1996; Bladt et al. 1995] and tissue repair [Borowiak et al. 2004; Huh et al. 2004]. Hepatocyte growth factor (HGF) is the only known ligand for the c-MET receptor and is expressed mainly in cells of mesenchymal origin, although some epithelial cancer cells appear to express both HGF and c-MET [Ma et al. 2008; Tsao et al. 2001, 1998; To and Tsao, 1998; Tuck et al. 1996; Furukawa et al. 1995]. Under normal conditions c-MET dimerizes and autophosphorylates upon ligand binding, which in turn creates active docking sites for proteins that mediate downstream signaling leading to the activation of the mitogen-activated protein kinase (MAPK), phosphatidylinositol 3-kinase (PI3K)-AKT, v-src sarcoma (Schmidt-Ruppin A-2) viral oncogene homolog (SRC), signal transducer and activator of transcription (STAT) signaling pathways [Organ et al. 2011; Trusolino et al. 2010; Seiden-Long et al. 2008; Peschard and Park, 2007]. Such activation evokes a variety of pleiotropic biological responses leading to increased cell growth, scattering and motility, invasion, protection from apoptosis, branching morphogenesis, and angiogenesis [Sierra et al. 2008; Conrotto et al. 2005; Yi and Tsao, 2000; Silvagno et al. 1995]. However, under pathological conditions improper activation of c-MET may confer proliferative, survival and invasive/metastatic abilities of cancer cells [Benvenuti and Comoglio, 2007; Danilkovitch-Miagkova and Zbar, 2002]. This review article discusses the evidence for the potential role of c-MET as a biomarker and therapeutic target in cancer, especially non-small cell lung carcinoma (NSCLC).

Role of c-MET in cancer

c-MET was first identified in the early 1980s as the product of a chromosomal rearrangement after treatment with the carcinogen N-methyl-N′-nitro-N-nitrosoguanidine [Cooper et al. 1984]. This rearrangement results in a constitutively fused oncogene, TPR-MET, which translates into an oncoprotein following dimerization by a leucine-zipper motif located in the TPR moiety. This provides the structural requirement for c-MET kinase to be constitutively active. TPR-MET has the ability to transform epithelial cells [Rodrigues and Park, 1993; Park et al. 1986], and to induce spontaneous mammary tumors when ubiquitously overexpressed in transgenic mice [Liang et al. 1996]. These findings set the starting point for a currently ongoing effort to unveil all oncogenic abilities of c-MET. It took more than a decade to provide the proof of concept for the role of c-MET in human cancers, which became evident following the identification of activating point mutations in the germline of patients affected by hereditary papillary renal carcinomas [Olivero et al. 1999; Schmidt et al. 1999]. However, spontaneously occurring oncogenic MET mutations remain rare at 2–3% [Schmidt et al. 1999, 1997].

A large number of reports have shown that an altered level of RTK activation can play an important role in the pathophysiology of cancer [Lemmon and Schlessinger, 2010]. Deregulation and the consequent aberrant signaling of c-MET may occur by different mechanisms including gene amplification, overexpression, activating mutations, increased autocrine or paracrine ligand-mediated stimulation, and interaction with other active cell-surface receptors. Many studies have reported that c-MET is overexpressed in a variety of carcinomas including lung, breast, ovary, kidney, colon, thyroid, liver, and gastric carcinomas [Knowles et al. 2009; Lengyel et al. 2005; Tokunou et al. 2001; Ramirez et al. 2000; Tsao et al. 1998; Koochekpour et al. 1997; Olivero et al. 1996; Tuck et al. 1996; Di Renzo et al. 1995c; Furukawa et al. 1995; Liu et al. 1992; Soman et al. 1991; Houldsworth et al. 1990], (Table 1). Such overexpression could be the result of transcriptional activation, hypoxia-induced overexpression [Pennacchietti et al. 2003], or as a result of MET amplification, which is notably observed only in a small subset of cancers [Cappuzzo et al. 2009a, 2009b; Beau-Faller et al. 2008; Miller et al. 2006; Han et al. 2003; Tsugawa et al. 1998; Kuniyasu et al. 1992]. Transgenic mice overexpressing c-MET have been reported to spontaneously develop hepatocellular carcinoma, and when the transgene was inactivated, tumor regression was reported even in large tumors [Wang et al. 2001].

Table 1.

c-MET and hepatocyte growth factor (HGF) are highly expressed in a variety of carcinomas.

MET gene mutation and amplification

As mentioned, somatic mutations on the MET gene are rarely found in patients with nonhereditary cancer. To date, missense mutations and single nucleotide polymorphisms (SNPs) have been found in the SEMA and juxtamembrane domain of MET, whereas, activating mutations have been described mainly in NSCLC, hereditary and spontaneous renal carcinomas, hepatocellular carcinomas, gliomas, gastric, squamous cell carcinoma of the head and neck, and breast carcinomas [Stella et al. 2011; Seiwert et al. 2009; Ma et al. 2008; Giordano et al. 2000; Lee et al. 2000; Park et al. 1999]. Potentially oncogenic mutations involve mainly point mutations that generate an alternative splicing encoding a shorter protein that lacks exon 14, which encodes for the juxtamembrane domain of c-MET [Ma et al. 2008; Lutterbach et al. 2007]; point mutations in the kinase domain that render the enzyme constitutively active [Giordano et al. 2000]; and Y1003 mutations that inactivate the Cbl binding site leading to constitutive c-MET expression [Kong-Beltran et al. 2006; Peschard et al. 2001; Vigna et al. 1999]. In contrast, several other point mutations (i.e. N375S, R988C and T1010I) have been reported as SNPs since they have been found to lack transforming abilities [John et al. 2011; Tyner et al. 2010; Tengs et al. 2006].

The most frequent genetic alteration is gene amplification, and as a consequence high c-MET protein expression and activation which has been reported as associated with a poor prognosis in NSCLC, colorectal and gastric cancers (Table 2) [Go et al. 2010; Cappuzzo et al. 2009a, 2009b; Zeng et al. 2008; Nakajima et al. 1999; Di Renzo et al. 1995a]. There were also reports that MET is more frequently amplified in metastatic tumors, suggesting a role in the late phases of malignant progression [Zeng et al. 2008; Tsugawa et al. 1998; Di Renzo et al. 1995a].

Table 2.

Types of cancers where c-MET/hepatocyte growth factor (HGF) has been reported to be important in primary tumor or metastatic progression.

Primary Metastases Clinical association
NSCLC 4.1% (18/435) Poor prognosis [Cappuzzo et al. 2009b]
3.9% (7/180) [Go et al. 2010]
3.8% (2/53) [Chen et al. 2009]
7% (12/183) [Onitsuka et al. 2010b]
Colorectal 9% (3/32) 89% (8/9) [Di Renzo et al. 1995a]
3.6% (9/247) 18% (25/147) [Zeng et al. 2008]
Gastric 9.4% (13/138) Poorer prognosis [Nakajima et al. 1999]
21.2% (100/472) Poorer prognosis [Lee et al. 2011]
23% (15/64) Advanced stage and poor prognosis [Kuniyasu et al. 1992]
Ovarian 5.6% (5/89) Poor prognosis [Yamamoto et al. 2011]

NSCLC, non-small cell lung carcinoma.

c-MET protein overexpression

Over the years many groups have established that c-MET and HGF are highly expressed in a large number of solid and soft tumors (for a comprehensive list, see www.vai.org/met). The list of tumors in which c-MET is expressed is large, and it has been shown that high levels of c-MET can lead to the constitutive activation of the enzyme, as well as rendering cells sensitive to subthreshold amounts of HGF. Although many of these studies have not identified the level of c-MET receptor activity/phosphorylation or compared the expression level with its normal counterpart, it could be speculated that it is expressed with autocrine loops of HGF/c-MET which increase cell proliferation and metastases [Navab et al. 2009; Yi and Tsao, 2000; Yi et al. 1998; Rong et al. 1994; Tsao et al. 1993]. Furthermore, independent studies have shown that HGF is expressed ubiquitously throughout the body, showing this growth factor to be a systemically available cytokine as well as coming from the tumor stroma [Vuononvirta et al. 2009; Parr et al. 2004; Aguirre Ghiso et al. 1999]. A positive paracrine and autocrine loop of c-MET activation can therefore lead to further MET expression [Boccaccio et al. 1994].

c-MET as prognostic markers

High levels of c-MET and/or HGF expression have been associated with poor patient outcome. Nearly half of lung adenocarcinomas demonstrate high expression of c-MET and HGF [Tsao et al. 2001; Siegfried et al. 1997; Takanami et al. 1996]. Such high expression patterns have been reported to correlate with increased tumor growth rate and metastasis, poor prognosis and resistance to radiotherapy [De Bacco et al. 2011; Matsui et al. 2010; Navab et al. 2009]. High levels of c-MET/HGF in breast carcinoma have been correlated with histological grade, poor prognosis and high proliferative cell index [Garcia et al. 2007a; Edakuni et al. 2001; Yamashita et al. 1994], and even with a greater incidence of metastases [Chen et al. 2007; Garcia et al. 2007b]. In these reports, c-MET overexpression was observed in hypoxic areas and correlated directly with poorer overall survival. Table 3 provides an overview of several reports that have described c-MET and/or HGF as a prognostic marker.

Table 3.

Reports that have considered c-MET and hepatocyte growth factor (HGF) as prognostic markers.

Type of cancer Number of patients studied c-MET HGF
[Ichimura et al. 1996] NSCLC 107 High c-MET protein, worse prognosis
[Nakamura et al. 2007] NSCLC 130 High c-MET associated with tumor differentiation High HGF associated with tumor differentiation
[Takanami et al. 1996] NSCLC c-MET-positive patients, worse prognosis HGF-positive patients, worse prognosis
[Siegfried et al. 1997] NSCLC 56 HGF-positive patients, worse prognosis
[Masuya et al. 2004] NSCLC 88 c-MET-positive patients, worse prognosis
[Iwasaki et al. 2004] NSCLC 71 No correlation
[Aune et al. 2011] Ovarian 123 Patients with high HGF, poor prognosis
[Barakat et al. 2010] Gastrointestinal 118 High HGF discriminates malignant from benign
[Skoldenberg et al. 2009] Neuroblastoma 73 High HGF, higher disease stage
[Toiyama et al. 2009] CRC 184 Patients with high HGF, poor prognosis
[Tanimoto et al. 2008] RCC 45 Patients with high HGF, poor prognosis
[Kammula et al. 2007] CRC 60 Patients with high HGF, poor prognosis

CRC, colorectal cancer; NSCLC, non-small cell lung carcinoma; RCC, renal cell cancer.

Plexins: modulators of c-MET activation

In the last decade several groups have reported that Plexin B family members (Plexin-B1, Plexin-B2, and Plexin-B3) can bind to both HGF RTK family members: c-MET and RON [Conrotto et al. 2004; Giordano et al. 2002]. B-Plexins are single-pass transmembrane receptors shown to share structural similarities with c-MET [Tamagnone and Comoglio, 2000; Tamagnone et al. 1999]. Giordano and colleagues have demonstrated that activation of Plexin-B1 by its high-affinity ligand, Sema4D, can transactivate c-MET's invasive growth program, thus promoting tumor growth, invasion, migration and angiogenesis [Sierra et al. 2008; Conrotto et al. 2005, 2004; Giordano et al. 2002]. Similarly, Swiercz and colleagues [2008, 2004] observed that the Sema4D/Plexin-B1 complex can transactivate ERBB2 promoting tumor growth [Swiercz et al. 2008, 2004].

As with other semaphorins and plexins, Plexin-B1's roles in cancer have been contradictory [Ch'ng and Kumanogoh, 2010; Capparuccia and Tamagnone, 2009]. Sema4D/Plexin-B1 complex has been associated with potentiating tumor progression in gastric [Schroeder, 1992], breast [Sierra et al. 2008; Swiercz et al. 2008], lung [Basile et al. 2006], head and neck [Basile et al. 2006, 2005], cervical [Qiang et al. 2011], ovary [Valente et al. 2009] and pancreatic [Kato et al. 2011] cancers and tumor-associated macrophage proangiogenic effects [Sierra et al. 2008]. Furthermore, Plexin-B1 point mutations found in prostate cancer bone metastases and lymph node metastases of other primary cancers render cells more motile with an increase in invasion, adhesion, and lamellipodia extension [Wong et al. 2007]. Conversely, Plexin-B1 has been shown to play a tumor suppressor role in melanomas. Two different groups have shown that Plexin-B1 expression in melanomas reduces BRAF signaling pathways [Argast et al. 2009] and decreases c-MET expression levels [Stevens et al. 2010]. These results challenge the previously published murine model lacking Plexin-B1, which demonstrated that tumor growth or angiogenesis of human melanoma cells growing in Plexin-B1-lacking environments are not affected [Fazzari et al. 2007].

c-MET as a mechanism of resistance to epidermal growth factor receptor therapies

During the past 5 years, c-MET has gained considerable interest following the report that lung adenocarcinoma cell line HCC827 (bearing the sensitizing mutation in the epidermal growth factor gene) developed resistance by amplification of the MET gene when exposed to increasing concentrations of the epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI), erlotinib, for long periods of time [Engelman et al. 2007]. This finding was further supported by clinical evidence showing lung tumors from four EGFR TKI refractory patients displayed the MET amplification [Engelman et al. 2007]. Engelman's report was also the first to demonstrate that patients could bear two different mechanisms of resistance to an EGFR TKI, as one of the four patients harbored both the MET amplification and the commonly found gatekeeper mutation in EGFR (T790M). The latter has been shown to decrease the sensitivity of the EGFR kinase to the reversible TKI [Yun et al. 2008]. Furthermore, cells with amplified MET were now sensitive to a dual treatment with EGFR and c-MET TKI, suggesting that inhibition of both receptors could result in disease stabilization.

Several investigators have since reported that a large cohort of patients with lung cancer who were treated with EGFR TKI and relapsed (approximately 18%) displayed MET amplification [Turke et al. 2010; Cappuzzo et al. 2009a; Chen et al. 2009; Bean et al. 2007] or high HGF levels [Onitsuka et al. 2010a]. It is important to note that the prevalence of MET amplification in patients with NSCLC who are untreated has been reported to be as low as 4–7% (Table 4). Turke and colleagues have suggested that EGFR TKI-resistant tumors with MET amplification arise in tumors with pre-existing clones of MET-amplified cells, which survive TKI therapy [Turke et al. 2010].

Table 4.

Reports that analyzed MET gene copy number (GCN) in non-small cell lung carcinoma (NSCLC).

Number of patients analyzed Number of patients with high MET GCN
[Engelman et al. 2007] 8 0
[Bean et al. 2007] 62 2 (3%)
[Okuda et al. 2008] 213 12 (5%)
[Beau-Faller et al. 2008] 106 22 (21%)
[Cappuzzo et al. 2009a] 206 12 (6%)
[Cappuzzo et al. 2009b] 435 18 (4%)
[Chen et al. 2009] 53 2 (4%)
[Onitsuka et al. 2010b] 183 12 (7%)
[Go et al. 2010] 180 7 (4%)
Total 1446 87 (6%)

c-MET activation may bypass EGFR TKIs in sensitive cells by two putative mechanisms: c-MET autophosphorylation creates docking sites where downstream signaling proteins can transduce prosurvival signals via the MAPK and PI3K/AKT signaling pathways; and transphosphorylation of other ERBB receptors may amplify the protumorigenic invasive program of c-MET. The latter mechanism is also known as ‘cross talk’ between RTKs. For all these properties, c-MET is believed to play a significant role in tumor progression and metastasis.

c-MET as a therapeutic target

Because of its pleotrophic role in cellular processes important in oncogenesis and cancer progression, c-MET is considered to be an important target in anticancer therapy [Trusolino et al. 2010; Migliore and Giordano, 2008; Peschard and Park, 2007; Corso et al. 2005]. Several molecules targeting c-MET have recently been evaluated in early phase clinical trials. Most of them are small kinase inhibitors, while some are biological antagonists and monoclonal antibodies targeting either the ligand or the receptor [Eder et al. 2009; Comoglio et al. 2008; Toschi and Janne, 2008].

Preclinical studies have shown that in animal models, the inhibition of c-MET or neutralization of its ligand impairs tumorigenic and metastatic properties of cancer cells [Corso et al. 2008; McDermott et al. 2007; Zou et al. 2007; Petrelli et al. 2006; Smolen et al. 2006; Michieli et al. 2004]. Based on this evidence, compounds that abrogate the kinase activity of c-MET have been developed [Porter, 2010]. An important issue relevant to the development of c-MET inhibitors is the identification of a molecular profile predictive of tumors that would benefit from this targeted therapy. Several studies have shown that certain gastric cancer and NSCLC cell lines display an exquisite sensitivity to c-MET inhibitors. Collectively, these studies have analyzed large panels of cell lines with known MET gene copy number and mutation variations. These studies demonstrated that cell lines with activated HGF/c-MET autocrine loop or MET amplification upon treatment with a c-MET TKI undergo apoptosis both in vitro and in vivo [Pan et al. 2010; Corso et al. 2008; Vigna et al. 2008; Lutterbach et al. 2007; Petrelli et al. 2006; Smolen et al. 2006]. The results have identified a subset of tumors based on genetic alterations that appear to be dependent on sustained c-MET activity for their growth and survival, and treatment with a single agent may inhibit tumor growth and induce cell death.

To date, only a few c-MET inhibitors have entered clinical trials. The non-ATP competitive c-MET inhibitor, tivantinib (ARQ 197), has recently completed a phase II clinical trial and shown to produce an increased response rate and overall survival when combined with erlotinib [Schiller et al. 2010]. Prior to this study, a phase I trial showed that 27% (14 out of 51 patients) of patients had stable disease for over 4 months [Yap et al. 2011]. Based on these results, tivantinib has entered a randomized, double-blind, placebo-controlled phase III study in previously treated patients with metastatic NSCLC [DeJager, 2010]. Cabozantinib (XL184), a multikinase inhibitor that targets c-MET, VEGFR2, AXL, KIT, TIE2, FLT3, and RET, has reached phase II/III trials showing reduction of tumor mass in almost 60% of patients treated with glioblastoma and an overall disease control rate of almost 50% in all of the patients who received this inhibitor in phase II studies [Wen, 2010; Salgia et al. 2008]. Cabozantinib has entered a combination regimen with erlotinib as a phase Ib/II trial. Very little is known about the results of this trial, with the exception of a partial response by patients who have MET amplification or EGFR T790M mutation [Wakelee et al. 2010]. Foretinib (XL880) is a multikinase inhibitor that targets c-MET and VEGFR2 at nanomolar concentrations [Qian et al. 2009]. It was found to stabilize the disease in 55% of the patients treated in a phase I trial [Eder et al. 2010] and, similar to other c-MET inhibitors, it recently started a phase II trial that considers the combination of this inhibitor with erlotinib [Seymour, 2010]. Lastly, the dual c-MET and ALK inhibitor, crizotinib (PF02341066), is being studied in a phase III randomized, double-blind, placebo-controlled study following several articles that demonstrated its tumor and metastasis inhibitory effects in both c-MET and ALK-positive patients [Kijima et al. 2011; Kimura et al. 2011; Ou et al. 2011]. This drug has also started a combination phase I study that considers the combination of this multikinase c-MET inhibitor with the irreversible pan-HER inhibitor PF00299804. Other compounds such as JNJ38877605 are in early phase clinical trials, but little is known regarding their results. Several reviews have been published on the subject of c-MET TKI in clinical trials [Eder et al. 2009; Comoglio et al. 2008; Toschi and Janne, 2008].

Another type of c-MET-targeted agent is monoclonal antibodies that have displayed promising results in tumors with high HGF/c-MET levels. Rilotumumab (AMG102) is an anti-HGF monoclonal antibody that interferes with c-MET's activation by HGF [Giordano, 2009]. Rilotumumab is currently evaluated in clinical phase I/II studies alone or in combination with the EGFR-blocking antibody panitumumab [Amgen, 2008]. Previous studies have shown that rilotumumab decreases c-MET phosphorylation and can stabilize the progression of certain solid tumors [Wen et al. 2011; Gordon et al. 2010].

MetMAb (OA-5D5), a human, monovalent antagonistic anti-MET antibody [Jin et al. 2008], in preclinical studies was able to inhibit glioblastoma U87 and pancreatic BxPC3 and KP4 tumor xenografts followed by a decrease in cellular proliferation and motility [Jin et al. 2008; Martens et al. 2006]. A recent phase II clinical trial using MetMAb in combination with erlotinib to treat patients with NSCLC resulted in a doubling of patient survival from 6.4 to 12.4 months [Spigel et al. 2010; Zhou et al. 2010]. In this trial, the authors point out that ‘c-MET diagnostic negative tumors’ when treated with MetMAb and erlotinib had a worse overall survival when compared with the erlotinib plus placebo arm [hazard ratio (HR) = 2.52), pointing out that only c-MET-diagnostic positive tumors benefited from the combinational treatment (HR = 0.56) [Khachatryan et al. 2010]. So far, monoclonal antibodies in preclinical and clinical studies have only demonstrated partial or complete response in patients (or cell lines) with high c-MET levels or an HGF/c-MET autocrine loop [Wen et al. 2011; Gordon et al. 2010; Khachatryan et al. 2010; Jin et al. 2008; Vigna et al. 2008; Martens et al. 2006].

In recent years, the therapeutic aim of finding drugs that selectively target a molecule expressed on neoplastic cells has shifted to finding drugs or combinations of drugs that are able to inhibit multiple pathways both in cancer cells and cells of the microenvironment [Sierra et al. 2010]. We have described multiple clinical trials in which c-MET-specific inhibitors are being combined with other RTK inhibitors. This rationale has gained momentum after clinical experience shows that patients who undergo a single-targeted therapy develop drug resistance and relapse. In addition, we have become more knowledgeable with regard to the notion that the tumor microenvironment plays an important role in maintaining the tumor niche; therefore, combination therapies attempt to inhibit neoplastic cells, the vessels and supporting cells (cancer-associated fibroblasts or tumor-associated macrophages) that transport and/or provide nutrients and growth factors to them [Petrelli and Valabrega, 2009]. Finally, the use of multikinase inhibitors has the potential of delaying the development of resistance, since it is known that neoplastic cells are able to undergo an ‘oncogenic switch’ by which the cell that was originally dependant on a single oncogene can rely on the activation of alternative(s) oncogenes [Cepero et al. 2010b; Sierra et al. 2010].

Blocking HGF or c-MET antibodies and TKI appears promising and it is anticipated that many studies will be initiated in the years to come, since c-MET is highly expressed in a wide variety of tumors. All of the c-MET-targeted therapies previously discussed exhibit the potential to enter a phase III study either alone or in combination with other kinase inhibitors and reach the approval of the US Food and Drug Administration in the future.

Potential resistant factors to c-MET inhibitors

As with other TKI inhibitors, sensitive cells/tumors treated with c-MET inhibitors develop resistance [Cepero et al. 2010b; Sierra et al. 2010]. Since no clinical specimens are still available, only preclinical in vitro systems have been able to predict the possible mechanisms of resistance that patients will develop when exposed to anti-MET treatments. So far, three mechanisms of resistance to c-MET inhibitors have been described. First, cells treated with c-MET TKI at high fixed doses develop a dependency on EGFRs [Corso et al. 2010; McDermott et al. 2010]. Both reports conclude that cells harboring high MET copy number will undergo an oncogenic switch that will create an ERBB tyrosine kinase dependency, similar to the oncogenic switch from EGFR to c-MET in NSCLC cells. The second known mechanism of resistance reported by Cepero et al. [2010a] is that when c-MET-dependent NSCLC and gastric cell lines are exposed to increasing doses of c-MET inhibitors amplify wild-type MET and KRAS, it enables cells to overcome the inhibitory threshold of the compound and still sustain high MAPK and PI3K/AKT activity. The third reported mechanism of resistance is the acquisition of a point mutation in the activation loop of c-MET (Y1230H) [Qi et al. 2011]. This mutation had previously been described as a somatic mutation in hereditary and sporadic renal carcinomas [Giordano et al. 2000]; here it has been shown to overcome the inhibitory effect of any c-MET kinase inhibitor.

Summary

With just over 25 years since its first discovery, the c-MET receptor is emerging as an important target for personalized cancer therapy. Inhibition of c-MET receptor activity in vivo has shown promising results in inhibition of tumor cell growth and in overcoming resistance to anti-EGFR therapy, which has now becoming a standard therapy for patients with advanced NSCLC. Results from early phase clinical trials are starting to demonstrate the importance of c-MET/HGF signaling in cancer biology. The challenge will be to explore and discover further other important crosstalk mechanisms involving this pathway, which could lead to further improvement in the efficacy of novel anticancer therapies and improve patient survival.

Acknowledgements

Matthew Joynson, a medical writer, assisted with the styling of this manuscript. The authors wrote and revised the main draft of the article.

Funding

This manuscript was partially supported by the Ontario Research Fund Research Excellence Award (RE-03-020) from the Ontario Ministry of Research and Innovation, Canadian Institutes of Health Research (grant number MOP-64345) and in part by the Ontario Ministry of Health and Long Term Care. MST is the M. Qasim Choksi Chair in Lung Cancer Translational Research. Editorial assistance was supported by Daiichi Sankyo Europe GmbH.

Conflicts of interest statement

Dr J. Rafael Sierra declares no conflict of interest. Dr Ming-Sound Tsao has received honoraria from Daiichi Sankyo Europe GmbH for speaking at scientific symposia.

References

  1. Aguirre Ghiso J.A., Alonso D.F., Farias E.F., Gomez D.E., de Kier Joffe E.B. (1999) Deregulation of the signaling pathways controlling urokinase production. Its relationship with the invasive phenotype. Eur J Biochem 263: 295–304 [DOI] [PubMed] [Google Scholar]
  2. Amgen (2008) Panitumumab combination study with AMG 102 or AMG 479 in wild-type KRAS mCRC. National Clinical Trial NCT00788957. Available at: http://clinicaltrials.gov/ct2/show/NCT00788957 (accessed 18 August 2011).
  3. Argast G.M., Croy C.H., Couts K.L., Zhang Z., Litman E., Chan D.C., et al. (2009) Plexin B1 is repressed by oncogenic B-Raf signaling and functions as a tumor suppressor in melanoma cells. Oncogene 28: 2697–2709 [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Aune G., Lian A.M., Tingulstad S., Torp S.H., Forsmo S., Reseland J.E., et al. (2011) Increased circulating hepatocyte growth factor (HGF): a marker of epithelial ovarian cancer and an indicator of poor prognosis. Gynecol Oncol 121: 402–406 [DOI] [PubMed] [Google Scholar]
  5. Barakat O., Rodriguez G.C., Raijman I., Allison P.M., Nieto J., Ozaki C.F., et al. (2010) Clinical value of plasma hepatocyte growth factor measurement for the diagnosis of periampullary cancer and prognosis after pancreaticoduodenectomy. J Surg Oncol 102: 816–820 [DOI] [PubMed] [Google Scholar]
  6. Basile J.R., Afkhami T., Gutkind J.S. (2005) Semaphorin 4D/plexin-B1 induces endothelial cell migration through the activation of PYK2, Src, and the phosphatidylinositol 3-kinase-Akt pathway. Mol Cell Biol 25: 6889–6898 [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Basile J.R., Castilho R.M., Williams V.P., Gutkind J.S. (2006) Semaphorin 4D provides a link between axon guidance processes and tumor-induced angiogenesis. Proc Natl Acad Sci U S A 103: 9017–9022 [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Bean J., Brennan C., Shih J.Y., Riely G., Viale A., Wang L., et al. (2007) 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 104: 20932–20937 [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Beau-Faller M., Ruppert A.M., Voegeli A.C., Neuville A., Meyer N., Guerin E., et al. (2008) MET gene copy number in non-small cell lung cancer: molecular analysis in a targeted tyrosine kinase inhibitor naive cohort. J Thorac Oncol 3: 331–339 [DOI] [PubMed] [Google Scholar]
  10. Benvenuti S., Comoglio P.M. (2007) The MET receptor tyrosine kinase in invasion and metastasis. J Cell Physiol 213: 316–325 [DOI] [PubMed] [Google Scholar]
  11. Bladt F., Riethmacher D., Isenmann S., Aguzzi A., Birchmeier C. (1995) Essential role for the c-met receptor in the migration of myogenic precursor cells into the limb bud. Nature 376: 768–771 [DOI] [PubMed] [Google Scholar]
  12. Boccaccio C., Gaudino G., Gambarotta G., Galimi F., Comoglio P.M. (1994) Hepatocyte growth factor (HGF) receptor expression is inducible and is part of the delayed-early response to HGF. J Biol Chem 269: 12846–12851 [PubMed] [Google Scholar]
  13. Borowiak M., Garratt A.N., Wustefeld T., Strehle M., Trautwein C., Birchmeier C. (2004) Met provides essential signals for liver regeneration. Proc Natl Acad Sci U S A 101: 10608–10613 [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Brand-Saberi B., Muller T.S., Wilting J., Christ B., Birchmeier C. (1996) Scatter factor/hepatocyte growth factor (SF/HGF) induces emigration of myogenic cells at interlimb level in vivo. Dev Biol 179: 303–308 [DOI] [PubMed] [Google Scholar]
  15. Capparuccia L., Tamagnone L. (2009) Semaphorin signaling in cancer cells and in cells of the tumor microenvironment–two sides of a coin. J Cell Sci 122: 1723–1736 [DOI] [PubMed] [Google Scholar]
  16. Cappuzzo F., Janne P.A., Skokan M., Finocchiaro G., Rossi E., Ligorio C., et al. (2009a) MET increased gene copy number and primary resistance to gefitinib therapy in non-small-cell lung cancer patients. Ann Oncol 20: 298–304 [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Cappuzzo F., Marchetti A., Skokan M., Rossi E., Gajapathy S., Felicioni L., et al. (2009b) Increased MET gene copy number negatively affects survival of surgically resected non-small-cell lung cancer patients. J Clin Oncol 27: 1667–1674 [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Cepero V., Sierra J.R., Corso S., Ghiso E., Casorzo L., Perera T., et al. (2010a) MET and KRAS gene amplification mediates acquired resistance to MET tyrosine kinase inhibitors. Cancer Res 70: 7580–7590 [DOI] [PubMed] [Google Scholar]
  19. Cepero V., Sierra J.R., Giordano S. (2010b) Tyrosine kinases as molecular targets to inhibit cancer progression and metastasis. Curr Pharm Des 16: 1396–1409 [DOI] [PubMed] [Google Scholar]
  20. Ch'ng E.S., Kumanogoh A. (2010) Roles of Sema4D and Plexin-B1 in tumor progression. Mol Cancer 9: 251–251 [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Chau N.G., Perez-Ordonez B., Zhang K., Pham N.A., Ho J., Zhang T., et al. (2011) The association between EGFR variant III, HPV, p16, c-MET, EGFR gene copy number and response to EGFR inhibitors in patients with recurrent or metastatic squamous cell carcinoma of the head and neck. Head Neck Oncol 3: 11–11 [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Chen H.H., Su W.C., Lin P.W., Guo H.R., Lee W.Y. (2007) Hypoxia-inducible factor-1alpha correlates with MET and metastasis in node-negative breast cancer. Breast Cancer Res Treat 103: 167–175 [DOI] [PubMed] [Google Scholar]
  23. Chen H.J., Mok T.S., Chen Z.H., Guo A.L., Zhang X.C., Su J., et al. (2009) Clinicopathologic and molecular features of epidermal growth factor receptor T790M mutation and c-MET amplification in tyrosine kinase inhibitor-resistant Chinese non-small cell lung cancer. Pathol Oncol Res 15: 651–658 [DOI] [PubMed] [Google Scholar]
  24. Comoglio P.M., Giordano S., Trusolino L. (2008) Drug development of MET inhibitors: targeting oncogene addiction and expedience. Nat Rev Drug Discov 7: 504–516 [DOI] [PubMed] [Google Scholar]
  25. Conrotto P., Corso S., Gamberini S., Comoglio P.M., Giordano S. (2004) Interplay between scatter factor receptors and B plexins controls invasive growth. Oncogene 23: 5131–5137 [DOI] [PubMed] [Google Scholar]
  26. Conrotto P., Valdembri D., Corso S., Serini G., Tamagnone L., Comoglio P.M., et al. (2005) Sema4D induces angiogenesis through Met recruitment by Plexin B1. Blood 105: 4321–4329 [DOI] [PubMed] [Google Scholar]
  27. Cooper C.S., Park M., Blair D.G., Tainsky M.A., Huebner K., Croce C.M., et al. (1984) Molecular cloning of a new transforming gene from a chemically transformed human cell line. Nature 311: 29–33 [DOI] [PubMed] [Google Scholar]
  28. Corso S., Comoglio P.M., Giordano S. (2005) Cancer therapy: can the challenge be MET?. Trends Mol Med 11: 284–292 [DOI] [PubMed] [Google Scholar]
  29. Corso S., Ghiso E., Cepero V., Sierra J.R., Migliore C., Bertotti A., et al. (2010) Activation of HER family members in gastric carcinoma cells mediates resistance to MET inhibition. Mol Cancer 9: 121–121 [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Corso S., Migliore C., Ghiso E., De Rosa G., Comoglio P.M., Giordano S. (2008) Silencing the MET oncogene leads to regression of experimental tumors and metastases. Oncogene 27: 684–693 [DOI] [PubMed] [Google Scholar]
  31. Danilkovitch-Miagkova A., Zbar B. (2002) Dysregulation of Met receptor tyrosine kinase activity in invasive tumors. J Clin Invest 109: 863–867 [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. De Bacco F., Luraghi P., Medico E., Reato G., Girolami F., Perera T., et al. (2011) Induction of MET by ionizing radiation and its role in radioresistance and invasive growth of cancer. J Natl Cancer Inst 103: 645–661 [DOI] [PubMed] [Google Scholar]
  33. DeJager, R. (2010) ARQ 197 plus erlotinib versus placebo plus erlotinib for the treatment of non-squamous, non-small-cell lung cancer. National Clinical Trial NTC01244191. Available at: http://www.clinicaltrials.gov/ct2/show/NCT01244191?term=nct01244191&rank=1 (accessed 18 August 2011)
  34. Di Renzo M.F., Olivero M., Giacomini A., Porte H., Chastre E., Mirossay L., et al. (1995a) Overexpression and amplification of the met/HGF receptor gene during the progression of colorectal cancer. Clin Cancer Res 1: 147–154 [PubMed] [Google Scholar]
  35. Di Renzo M.F., Olivero M., Serini G., Orlandi F., Pilotti S., Belfiore A., et al. (1995b) Overexpression of the c-MET/HGF receptor in human thyroid carcinomas derived from the follicular epithelium. J Endocrinol Invest 18: 134–139 [DOI] [PubMed] [Google Scholar]
  36. Di Renzo M.F., Poulsom R., Olivero M., Comoglio P.M., Lemoine N.R. (1995c) Expression of the Met/hepatocyte growth factor receptor in human pancreatic cancer. Cancer Res 55: 1129–1138 [PubMed] [Google Scholar]
  37. Edakuni G., Sasatomi E., Satoh T., Tokunaga O., Miyazaki K. (2001) Expression of the hepatocyte growth factor/c-Met pathway is increased at the cancer front in breast carcinoma. Pathol Int 51: 172–178 [DOI] [PubMed] [Google Scholar]
  38. Eder J.P., Shapiro G.I., Appleman L.J., Zhu A.X., Miles D., Keer H., et al. (2010) A phase I study of foretinib, a multi-targeted inhibitor of c-Met and vascular endothelial growth factor receptor 2. Clin Cancer Res 16: 3507–3516 [DOI] [PubMed] [Google Scholar]
  39. Eder J.P., Vande Woude G.F., Boerner S.A., LoRusso P.M. (2009) Novel therapeutic inhibitors of the c-Met signaling pathway in cancer. Clin Cancer Res 15: 2207–2214 [DOI] [PubMed] [Google Scholar]
  40. Engelman J.A., Zejnullahu K., Mitsudomi T., Song Y., Hyland C., Park J.O., et al. (2007) MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science 316: 1039–1043 [DOI] [PubMed] [Google Scholar]
  41. Fazzari P., Penachioni J., Gianola S., Rossi F., Eickholt B.J., Maina F., et al. (2007) Plexin-B1 plays a redundant role during mouse development and in tumour angiogenesis. BMC Dev Biol 7: 55–55 [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Furukawa T., Duguid W.P., Kobari M., Matsuno S., Tsao M.S. (1995) Hepatocyte growth factor and Met receptor expression in human pancreatic carcinogenesis. Am J Pathol 147: 889–895 [PMC free article] [PubMed] [Google Scholar]
  43. Garcia S., Dales J.P., Charafe-Jauffret E., Carpentier-Meunier S., Andrac-Meyer L., Jacquemier J., et al. (2007a) Overexpression of c-Met and of the transducers PI3K, FAK and JAK in breast carcinomas correlates with shorter survival and neoangiogenesis. Int J Oncol 31: 49–58 [PubMed] [Google Scholar]
  44. Garcia S., Dales J.P., Charafe-Jauffret E., Carpentier-Meunier S., Andrac-Meyer L., Jacquemier J., et al. (2007b) Poor prognosis in breast carcinomas correlates with increased expression of targetable CD146 and c-Met and with proteomic basal-like phenotype. Hum Pathol 38: 830–841 [DOI] [PubMed] [Google Scholar]
  45. Giordano S. (2009) Rilotumumab, a mAb against human hepatocyte growth factor for the treatment of cancer. Curr Opin Mol Ther 11: 448–455 [PubMed] [Google Scholar]
  46. Giordano S., Corso S., Conrotto P., Artigiani S., Gilestro G., Barberis D., et al. (2002) The semaphorin 4D receptor controls invasive growth by coupling with Met. Nat Cell Biol 4: 720–724 [DOI] [PubMed] [Google Scholar]
  47. Giordano S., Maffe A., Williams T.A., Artigiani S., Gual P., Bardelli A., et al. (2000) Different point mutations in the met oncogene elicit distinct biological properties. FASEB J 14: 399–406 [DOI] [PubMed] [Google Scholar]
  48. Go H., Jeon Y.K., Park H.J., Sung S.W., Seo J.W., Chung D.H. (2010) High MET gene copy number leads to shorter survival in patients with non-small cell lung cancer. J Thorac Oncol 5: 305–313 [DOI] [PubMed] [Google Scholar]
  49. Gordon M.S., Sweeney C.S., Mendelson D.S., Eckhardt S.G., Anderson A., Beaupre D.M., et al. (2010) Safety, pharmacokinetics, and pharmacodynamics of AMG 102, a fully human hepatocyte growth factor-neutralizing monoclonal antibody, in a first-in-human study of patients with advanced solid tumors. Clin Cancer Res 16: 699–710 [DOI] [PubMed] [Google Scholar]
  50. Han S.Y., Druck T., Huebner K. (2003) Candidate tumor suppressor genes at FRA7G are coamplified with MET and do not suppress malignancy in a gastric cancer. Genomics 81: 105–107 [DOI] [PubMed] [Google Scholar]
  51. Heymann S., Koudrova M., Arnold H., Koster M., Braun T. (1996) Regulation and function of SF/HGF during migration of limb muscle precursor cells in chicken. Dev Biol 180: 566–578 [DOI] [PubMed] [Google Scholar]
  52. Houldsworth J., Cordon-Cardo C., Ladanyi M., Kelsen D.P., Chaganti R.S. (1990) Gene amplification in gastric and esophageal adenocarcinomas. Cancer Res 50: 6417–6422 [PubMed] [Google Scholar]
  53. Huh C.G., Factor V.M., Sanchez A., Uchida K., Conner E.A., Thorgeirsson S.S. (2004) Hepatocyte growth factor/c-met signaling pathway is required for efficient liver regeneration and repair. Proc Natl Acad Sci U S A 101: 4477–4482 [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Ichimura E., Maeshima A., Nakajima T., Nakamura T. (1996) Expression of c-met/HGF receptor in human non-small cell lung carcinomas in vitro and in vivo and its prognostic significance. Jpn J Cancer Res 87: 1063–1069 [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Iwasaki A., Kuwahara M., Yoshinaga Y., Shirakusa T. (2004) Basic fibroblast growth factor (bFGF) and vascular endothelial growth factor (VEGF) levels, as prognostic indicators in NSCLC. Eur J Cardiothorac Surg 25: 443–448 [DOI] [PubMed] [Google Scholar]
  56. Jin H., Yang R., Zheng Z., Romero M., Ross J., Bou-Reslan H., et al. (2008) MetMAb, the one-armed 5D5 anti-c-Met antibody, inhibits orthotopic pancreatic tumor growth and improves survival. Cancer Res 68: 4360–4368 [DOI] [PubMed] [Google Scholar]
  57. John T., Kohler D., Pintilie M., Yanagawa N., Pham N.A., Li M., et al. (2011) The ability to form primary tumor xenografts is predictive of increased risk of disease recurrence in early-stage non-small cell lung cancer. Clin Cancer Res 17: 134–141 [DOI] [PubMed] [Google Scholar]
  58. Kammula U.S., Kuntz E.J., Francone T.D., Zeng Z., Shia J., Landmann R.G., et al. (2007) Molecular co-expression of the c-Met oncogene and hepatocyte growth factor in primary colon cancer predicts tumor stage and clinical outcome. Cancer Lett 248: 219–228 [DOI] [PubMed] [Google Scholar]
  59. Kato S., Kubota K., Shimamura T., Shinohara Y., Kobayashi N., Watanabe S., et al. (2011) Semaphorin 4D, a lymphocyte semaphorin, enhances tumor cell motility through binding its receptor, plexinB1, in pancreatic cancer. Cancer Sci, 4 August [Epub ahead of print] [DOI] [PubMed] [Google Scholar]
  60. Khachatryan V., Sirunyan A.M., Tumasyan A., Adam W., Bergauer T., Dragicevic M., et al. (2010) Search for quark compositeness with the dijet centrality ratio in pp collisions at radicals=7 TeV. Phys Rev Lett 105: 262001–262001 [DOI] [PubMed] [Google Scholar]
  61. Kijima T., Takeuchi K., Tetsumoto S., Shimada K., Takahashi R., Hirata H., et al. (2011) Favorable response to crizotinib in three patients with echinoderm microtubule-associated protein-like 4-anaplastic lymphoma kinase fusion-type oncogene-positive non-small cell lung cancer. Cancer Sci 102: 1602–1604 [DOI] [PubMed] [Google Scholar]
  62. Kimura H., Nakajima T., Takeuchi K., Soda M., Mano H., Iizasa T., et al. (2011) ALK fusion gene positive lung cancer and 3 cases treated with an inhibitor for ALK kinase activity. Lung Cancer, 12 July [Epub ahead of print] [DOI] [PubMed] [Google Scholar]
  63. Knowles L.M., Stabile L.P., Egloff A.M., Rothstein M.E., Thomas S.M., Gubish C.T., et al. (2009) HGF and c-Met participate in paracrine tumorigenic pathways in head and neck squamous cell cancer. Clin Cancer Res 15: 3740–3750 [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Kong-Beltran M., Seshagiri S., Zha J., Zhu W., Bhawe K., Mendoza N., et al. (2006) Somatic mutations lead to an oncogenic deletion of met in lung cancer. Cancer Res 66: 283–289 [DOI] [PubMed] [Google Scholar]
  65. Koochekpour S., Jeffers M., Rulong S., Taylor G., Klineberg E., Hudson E.A., et al. (1997) Met and hepatocyte growth factor/scatter factor expression in human gliomas. Cancer Res 57: 5391–5398 [PubMed] [Google Scholar]
  66. Kuniyasu H., Yasui W., Kitadai Y., Yokozaki H., Ito H., Tahara E. (1992) Frequent amplification of the c-met gene in scirrhous type stomach cancer. Biochem Biophys Res Commun 189: 227–232 [DOI] [PubMed] [Google Scholar]
  67. Lee J.H., Han S.U., Cho H., Jennings B., Gerrard B., Dean M., et al. (2000) A novel germ line juxtamembrane Met mutation in human gastric cancer. Oncogene 19: 4947–4953 [DOI] [PubMed] [Google Scholar]
  68. Lemmon M.A., Schlessinger J. (2010) Cell signaling by receptor tyrosine kinases. Cell 141: 1117–1134 [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Lee J., Seo J.W., Jun H.J., Ki C.S., Park S.H., Park Y.S., et al. (2011) Impact of MET amplification on gastric cancer: possible roles as a novel prognostic marker and a potential therapeutic target. Oncol Rep 25: 1517–1524 [DOI] [PubMed] [Google Scholar]
  70. Lengyel E., Prechtel D., Resau J.H., Gauger K., Welk A., Lindemann K., et al. (2005) C-Met overexpression in node-positive breast cancer identifies patients with poor clinical outcome independent of Her2/neu. Int J Cancer 113: 678–682 [DOI] [PubMed] [Google Scholar]
  71. Liang T.J., Reid A.E., Xavier R., Cardiff R.D., Wang T.C. (1996) Transgenic expression of tpr-met oncogene leads to development of mammary hyperplasia and tumors. J Clin Invest 97: 2872–2877 [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Liu C., Park M., Tsao M.S. (1992) Overexpression of c-met proto-oncogene but not epidermal growth factor receptor or c-erbB-2 in primary human colorectal carcinomas. Oncogene 7: 181–185 [PubMed] [Google Scholar]
  73. Liu C., Tsao M.S. (1993a) In vitro and in vivo expressions of transforming growth factor-alpha and tyrosine kinase receptors in human non-small-cell lung carcinomas. Am J Pathol 142: 1155–1162 [PMC free article] [PubMed] [Google Scholar]
  74. Liu C., Tsao M.S. (1993b) Proto-oncogene and growth factor/receptor expression in the establishment of primary human non-small cell lung carcinoma cell lines. Am J Pathol 142: 413–423 [PMC free article] [PubMed] [Google Scholar]
  75. Liu N., Furukawa T., Kobari M., Tsao M.S. (1998) Comparative phenotypic studies of duct epithelial cell lines derived from normal human pancreas and pancreatic carcinoma. Am J Pathol 153: 263–269 [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Lutterbach B., Zeng Q., Davis L.J., Hatch H., Hang G., Kohl N.E., et al. (2007) Lung cancer cell lines harboring MET gene amplification are dependent on Met for growth and survival. Cancer Res 67: 2081–2088 [DOI] [PubMed] [Google Scholar]
  77. Ma P.C., Tretiakova M.S., MacKinnon A.C., Ramnath N., Johnson C., Dietrich S., et al. (2008) Expression and mutational analysis of MET in human solid cancers. Genes Chromosomes Cancer 47: 1025–1037 [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Martens T., Schmidt N.O., Eckerich C., Fillbrandt R., Merchant M., Schwall R., et al. (2006) A novel one-armed anti-c-Met antibody inhibits glioblastoma growth in vivo. Clin Cancer Res 12: 6144–6152 [DOI] [PubMed] [Google Scholar]
  79. Masuya D., Huang C., Liu D., Nakashima T., Kameyama K., Haba R., et al. (2004) The tumour-stromal interaction between intratumoral c-Met and stromal hepatocyte growth factor associated with tumour growth and prognosis in non-small-cell lung cancer patients. Br J Cancer 90: 1555–1562 [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Matsui S., Osada S., Tomita H., Komori S., Mori R., Sanada Y., et al. (2010) Clinical significance of aggressive hepatectomy for colorectal liver metastasis, evaluated from the HGF/c-Met pathway. Int J Oncol 37: 289–297 [DOI] [PubMed] [Google Scholar]
  81. McDermott U., Pusapati R.V., Christensen J.G., Gray N.S., Settleman J. (2010) Acquired resistance of non-small cell lung cancer cells to MET kinase inhibition is mediated by a switch to epidermal growth factor receptor dependency. Cancer Res 70: 1625–1634 [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. McDermott U., Sharma S.V., Dowell L., Greninger P., Montagut C., Lamb J., et al. (2007) Identification of genotype-correlated sensitivity to selective kinase inhibitors by using high-throughput tumor cell line profiling. Proc Natl Acad Sci U S A 104(50): 19936–19941http://www.ncbi.nlm.nih.gov/pubmed/18077425?dopt=Citation [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Michieli P., Mazzone M., Basilico C., Cavassa S., Sottile A., Naldini L., et al. (2004) Targeting the tumor and its microenvironment by a dual-function decoy Met receptor. Cancer Cell 6: 61–73 [DOI] [PubMed] [Google Scholar]
  84. Migliore C., Giordano S. (2008) Molecular cancer therapy: can our expectation be MET?. Eur J Cancer 44: 641–651 [DOI] [PubMed] [Google Scholar]
  85. Miller C.T., Lin L., Casper A.M., Lim J., Thomas D.G., Orringer M.B., et al. (2006) Genomic amplification of MET with boundaries within fragile site FRA7G and upregulation of MET pathways in esophageal adenocarcinoma. Oncogene 25: 409–418 [DOI] [PubMed] [Google Scholar]
  86. Nakajima M., Sawada H., Yamada Y., Watanabe A., Tatsumi M., Yamashita J., et al. (1999) The prognostic significance of amplification and overexpression of c-met and c-erb B-2 in human gastric carcinomas. Cancer 85: 1894–1902 [DOI] [PubMed] [Google Scholar]
  87. Nakamura Y., Niki T., Goto A., Morikawa T., Miyazawa K., Nakajima J., et al. (2007) c-Met activation in lung adenocarcinoma tissues: an immunohistochemical analysis. Cancer Sci 98: 1006–1013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Navab R., Liu J., Seiden-Long I., Shih W., Li M., Bandarchi B., et al. (2009) Co-overexpression of Met and hepatocyte growth factor promotes systemic metastasis in NCI-H460 non-small cell lung carcinoma cells. Neoplasia 11: 1292–1300 [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. Okuda K., Sasaki H., Yukiue H., Yano M., Fujii Y. (2008) Met gene copy number predicts the prognosis for completely resected non-small cell lung cancer. Cancer Sci 99: 2280–2285 [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Olivero M., Rizzo M., Madeddu R., Casadio C., Pennacchietti S., Nicotra M.R., et al. (1996) Overexpression and activation of hepatocyte growth factor/scatter factor in human non-small-cell lung carcinomas. Br J Cancer 74: 1862–1868 [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Olivero M., Valente G., Bardelli A., Longati P., Ferrero N., Cracco C., et al. (1999) Novel mutation in the ATP-binding site of the MET oncogene tyrosine kinase in a HPRCC family. Int J Cancer 82: 640–643 [DOI] [PubMed] [Google Scholar]
  92. Onitsuka T., Uramoto H., Nose N., Takenoyama M., Hanagiri T., Sugio K., et al. (2010a) Acquired resistance to gefitinib: the contribution of mechanisms other than the T790M, MET, and HGF status. Lung Cancer 68: 198–203 [DOI] [PubMed] [Google Scholar]
  93. Onitsuka T., Uramoto H., Ono K., Takenoyama M., Hanagiri T., Oyama T., et al. (2010b) Comprehensive molecular analyses of lung adenocarcinoma with regard to the epidermal growth factor receptor, K-ras, MET, and hepatocyte growth factor status. J Thorac Oncol 5: 591–596 [DOI] [PubMed] [Google Scholar]
  94. Organ S.L., Tong J., Taylor P., St-Germain J.R., Navab R., Moran M.F., et al. (2011) Quantitative phospho-proteomic profiling of hepatocyte growth factor (HGF)-MET signaling in colorectal cancer. J Proteome Res 10: 3200–3211 [DOI] [PubMed] [Google Scholar]
  95. Ou S.H., Kwak E.L., Siwak-Tapp C., Dy J., Bergethon K., Clark J.W., et al. (2011) Activity of crizotinib (PF02341066), a dual mesenchymal-epithelial transition (MET) and anaplastic lymphoma kinase (ALK) inhibitor, in a non-small cell lung cancer patient with de novo MET amplification. J Thorac Oncol 6: 942–946 [DOI] [PubMed] [Google Scholar]
  96. Pan B.S., Chan G.K., Chenard M., Chi A., Davis L.J., Deshmukh S.V., et al. (2010) MK-2461, a novel multitargeted kinase inhibitor, preferentially inhibits the activated c-Met receptor. Cancer Res 70: 1524–1533 [DOI] [PubMed] [Google Scholar]
  97. Park M., Dean M., Cooper C.S., Schmidt M., O'Brien S.J., Blair D.G., et al. (1986) Mechanism of met oncogene activation. Cell 45: 895–904 [DOI] [PubMed] [Google Scholar]
  98. Park W.S., Dong S.M., Kim S.Y., Na E.Y., Shin M.S., Pi J.H., et al. (1999) Somatic mutations in the kinase domain of the Met/hepatocyte growth factor receptor gene in childhood hepatocellular carcinomas. Cancer Res 59: 307–310 [PubMed] [Google Scholar]
  99. Parr C., Watkins G., Mansel R.E., Jiang W.G. (2004) The hepatocyte growth factor regulatory factors in human breast cancer. Clin Cancer Res 10: 202–211 [DOI] [PubMed] [Google Scholar]
  100. Pennacchietti S., Michieli P., Galluzzo M., Mazzone M., Giordano S., Comoglio P.M. (2003) Hypoxia promotes invasive growth by transcriptional activation of the met protooncogene. Cancer Cell 3: 347–361 [DOI] [PubMed] [Google Scholar]
  101. Peschard P., Fournier T.M., Lamorte L., Naujokas M.A., Band H., Langdon W.Y., et al. (2001) Mutation of the c-Cbl TKB domain binding site on the Met receptor tyrosine kinase converts it into a transforming protein. Mol Cell 8: 995–1004 [DOI] [PubMed] [Google Scholar]
  102. Peschard P., Park M. (2007) From Tpr-Met to Met, tumorigenesis and tubes. Oncogene 26: 1276–1285 [DOI] [PubMed] [Google Scholar]
  103. Petrelli A., Circosta P., Granziero L., Mazzone M., Pisacane A., Fenoglio S., et al. (2006) Ab-induced ectodomain shedding mediates hepatocyte growth factor receptor down-regulation and hampers biological activity. Proc Natl Acad Sci U S A 103: 5090–5095 [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Petrelli A., Valabrega G. (2009) Multitarget drugs: the present and the future of cancer therapy. Expert Opin Pharmacother 10: 589–600 [DOI] [PubMed] [Google Scholar]
  105. Porter J. (2010) Small molecule c-Met kinase inhibitors: a review of recent patents. Expert Opin Ther Pat 20: 159–177 [DOI] [PubMed] [Google Scholar]
  106. Qi J., McTigue M.A., Rogers A., Lifshits E., Christensen J.G., Janne P.A., et al. (2011) Multiple mutations and bypass mechanisms can contribute to development of acquired resistance to MET inhibitors. Cancer Res 71: 1081–1091 [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Qian F., Engst S., Yamaguchi K., Yu P., Won K.A., Mock L., et al. (2009) 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 69: 8009–8016 [DOI] [PubMed] [Google Scholar]
  108. Qiang R., Wang F., Shi L.Y., Liu M., Chen S., Wan H.Y., et al. (2011) Plexin-B1 is a target of miR-214 in cervical cancer and promotes the growth and invasion of HeLa cells. Int J Biochem Cell Biol 43: 632–641 [DOI] [PubMed] [Google Scholar]
  109. Ramirez R., Hsu D., Patel A., Fenton C., Dinauer C., Tuttle R.M., et al. (2000) Over-expression of hepatocyte growth factor/scatter factor (HGF/SF) and the HGF/SF receptor (cMET) are associated with a high risk of metastasis and recurrence for children and young adults with papillary thyroid carcinoma. Clin Endocrinol (Oxf) 53: 635–644 [DOI] [PubMed] [Google Scholar]
  110. Rodrigues G.A., Park M. (1993) Dimerization mediated through a leucine zipper activates the oncogenic potential of the met receptor tyrosine kinase. Mol Cell Biol 13: 6711–6722 [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Rong S., Segal S., Anver M., Resau J.H., Vande Woude G.F. (1994) Invasiveness and metastasis of NIH 3T3 cells induced by Met-hepatocyte growth factor/scatter factor autocrine stimulation. Proc Natl Acad Sci U S A 91: 4731–4735 [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. Salgia R., Sherman S., Hong D.S., Ng C.S., Frye J., Janisch L. (2008) A phase I study of XL184, a RET, VEGFR2, and MET kinase inhibitor, in patients (pts) with advanced malignancies, including pts with medullary thyroid cancer (MTC). ASCO Annual Meeting, Chicago, IL, USA, May 30–June 3, 2008 . [Google Scholar]
  113. Schiller J.H., Akerley W.L., Brugger W., Ferrari D., Garmey E.G., Gerber D.E., et al. (2010) Results from ARQ 197-209: a global randomized placebo-controlled phase II clinical trial of erlotinib plus ARQ 197 versus erlotinib plus placebo in previously treated EGFR inhibitor-naive patients with locally advanced or metastatic non-small cell lung cancer (NSCLC). J Clin Oncol 28: LBA7502–LBA7502 [Google Scholar]
  114. Schmidt L., Duh F.M., Chen F., Kishida T., Glenn G., Choyke P., et al. (1997) Germline and somatic mutations in the tyrosine kinase domain of the MET proto-oncogene in papillary renal carcinomas. Nat Genet 16: 68–73 [DOI] [PubMed] [Google Scholar]
  115. Schmidt L., Junker K., Nakaigawa N., Kinjerski T., Weirich G., Miller M., et al. (1999) Novel mutations of the MET proto-oncogene in papillary renal carcinomas. Oncogene 18: 2343–2350 [DOI] [PubMed] [Google Scholar]
  116. Schroeder P. (1992) Six tips for effective presentations. Nurs Qual Connect 2: 4–4 [PubMed] [Google Scholar]
  117. Seiden-Long I.M., Brown K.R., Shih W., Wigle D.A., Radulovich N., Jurisica I., et al. (2006) Transcriptional targets of hepatocyte growth factor signaling and Ki-ras oncogene activation in colorectal cancer. Oncogene 25: 91–102 [DOI] [PubMed] [Google Scholar]
  118. Seiden-Long I., Navab R., Shih W., Li M., Chow J., Zhu C.Q., et al. (2008) Gab1 but not Grb2 mediates tumor progression in Met overexpressing colorectal cancer cells. Carcinogenesis 29: 647–655 [DOI] [PubMed] [Google Scholar]
  119. Seiwert T.Y., Jagadeeswaran R., Faoro L., Janamanchi V., Nallasura V., El Dinali M., et al. (2009) The MET receptor tyrosine kinase is a potential novel therapeutic target for head and neck squamous cell carcinoma. Cancer Res 69: 3021–3031 [DOI] [PMC free article] [PubMed] [Google Scholar]
  120. Seymour, L. (2010) MET/VEGFR2 inhibitor GSK1363089 and erlotinib hydrochloride or erlotinib hydrochloride alone in treating patients with locally advanced or metastatic non-small cell lung cancer that has not responded to previous chemotherapy. National Clinical Trial NCT01068587. Available at: http://www.clinicaltrials.gov/ct2/show/NCT01068587?term=foretinib=rank=10 (accessed 18 August 2011)
  121. Siegfried J.M., Weissfeld L.A., Singh-Kaw P., Weyant R.J., Testa J.R., Landreneau R.J. (1997) Association of immunoreactive hepatocyte growth factor with poor survival in resectable non-small cell lung cancer. Cancer Res 57: 433–439 [PubMed] [Google Scholar]
  122. Sierra J.R., Cepero V., Giordano S. (2010) Molecular mechanisms of acquired resistance to tyrosine kinase targeted therapy. Mol Cancer 9: 75–75 [DOI] [PMC free article] [PubMed] [Google Scholar]
  123. Sierra J.R., Corso S., Caione L., Cepero V., Conrotto P., Cignetti A., et al. (2008) Tumor angiogenesis and progression are enhanced by Sema4D produced by tumor-associated macrophages. J Exp Med 205: 1673–1685 [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. Silvagno F., Follenzi A., Arese M., Prat M., Giraudo E., Gaudino G., et al. (1995) In vivo activation of met tyrosine kinase by heterodimeric hepatocyte growth factor molecule promotes angiogenesis. Arterioscler Thromb Vasc Biol 15: 1857–1865 [DOI] [PubMed] [Google Scholar]
  125. Skoldenberg E.G., Larsson A., Jakobson A., Hedborg F., Kogner P., Christofferson R.H., et al. (2009) The angiogenic growth factors HGF and VEGF in serum and plasma from neuroblastoma patients. Anticancer Res 29: 3311–3319 [PubMed] [Google Scholar]
  126. Smolen G.A., Sordella R., Muir B., Mohapatra G., Barmettler A., Archibald H., et al. (2006) 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 103: 2316–2321 [DOI] [PMC free article] [PubMed] [Google Scholar]
  127. Soman N.R., Correa P., Ruiz B.A., Wogan G.N. (1991) The TPR-MET oncogenic rearrangement is present and expressed in human gastric carcinoma and precursor lesions. Proc Natl Acad Sci U S A 88: 4892–4896 [DOI] [PMC free article] [PubMed] [Google Scholar]
  128. Spigel, D.R., Ervin, T.J., Ramlau, R., Daniel, D.B., Goldschmidt, J.H., Krzakowski, M. et al. (2010) Randomized, phase 2, multicenter, double-blind, placebo-controlled study evaluating MetMAb, an antibody to Met receptor, in combination with erlotinib, in patients with advanced non-small-cell lung cancer. 35th ESMO Conference, Milan, Italy, October 8–12, 2010.
  129. Stella G.M., Benvenut S., Gramaglia D., Scarpa A., Tomezzoli A., Cassoni P., et al. (2011) MET mutations in cancers of unknown primary origin (CUPs). Hum Mutat 32: 44–50 [DOI] [PubMed] [Google Scholar]
  130. Stevens L., McClelland L., Fricke A., Williamson M., Kuo I., Scott G. (2010) Plexin B1 suppresses c-Met in melanoma: a role for plexin B1 as a tumor-suppressor protein through regulation of c-Met. J Invest Dermatol 130: 1636–1645 [DOI] [PMC free article] [PubMed] [Google Scholar]
  131. Swiercz J.M., Kuner R., Offermanns S. (2004) Plexin-B1/RhoGEF-mediated RhoA activation involves the receptor tyrosine kinase ErbB-2. J Cell Biol 165: 869–880 [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. Swiercz J.M., Worzfeld T., Offermanns S. (2008) ErbB-2 and met reciprocally regulate cellular signaling via plexin-B1. J Biol Chem 283: 1893–1901 [DOI] [PubMed] [Google Scholar]
  133. Takanami I., Tanana F., Hashizume T., Kikuchi K., Yamamoto Y., Yamamoto T., et al. (1996) Hepatocyte growth factor and c-Met/hepatocyte growth factor receptor in pulmonary adenocarcinomas: an evaluation of their expression as prognostic markers. Oncology 53: 392–397 [DOI] [PubMed] [Google Scholar]
  134. Tamagnone L., Artigiani S., Chen H., He Z., Ming G.I., Song H., et al. (1999) Plexins are a large family of receptors for transmembrane, secreted, and GPI-anchored semaphorins in vertebrates. Cell 99: 71–80 [DOI] [PubMed] [Google Scholar]
  135. Tamagnone L., Comoglio P.M. (2000) Signalling by semaphorin receptors: cell guidance and beyond. Trends Cell Biol 10: 377–383 [DOI] [PubMed] [Google Scholar]
  136. Tanimoto S., Fukumori T., El-Moula G., Shiirevnyamba A., Kinouchi S., Koizumi T., et al. (2008) Prognostic significance of serum hepatocyte growth factor in clear cell renal cell carcinoma: comparison with serum vascular endothelial growth factor. J Med Invest 55: 106–111 [DOI] [PubMed] [Google Scholar]
  137. Tengs T., Lee J.C., Paez J.G., Zhao X., LaFramboise T., Giannoukos G., et al. (2006) A transforming MET mutation discovered in non-small cell lung cancer using microarray-based resequencing. Cancer Lett 239: 227–233 [DOI] [PubMed] [Google Scholar]
  138. To C.T., Tsao M.S. (1998) The roles of hepatocyte growth factor/scatter factor and met receptor in human cancers [review]. Oncol Rep 5: 1013–1024 [DOI] [PubMed] [Google Scholar]
  139. Toiyama Y., Miki C., Inoue Y., Okugawa Y., Tanaka K., Kusunoki M. (2009) Serum hepatocyte growth factor as a prognostic marker for stage II or III colorectal cancer patients. Int J Cancer 125: 1657–1662 [DOI] [PubMed] [Google Scholar]
  140. Tokunou M., Niki T., Eguchi K., Iba S., Tsuda H., Yamada T., et al. (2001) c-MET expression in myofibroblasts: role in autocrine activation and prognostic significance in lung adenocarcinoma. Am J Pathol 158: 1451–1463 [DOI] [PMC free article] [PubMed] [Google Scholar]
  141. Toschi L., Janne P.A. (2008) Single-agent and combination therapeutic strategies to inhibit hepatocyte growth factor/MET signaling in cancer. Clin Cancer Res 14: 5941–5946 [DOI] [PubMed] [Google Scholar]
  142. Trusolino L., Bertotti A., Comoglio P.M. (2010) MET signalling: principles and functions in development, organ regeneration and cancer. Nat Rev Mol Cell Biol 11: 834–848 [DOI] [PubMed] [Google Scholar]
  143. Tsao M.S., Liu N., Chen J.R., Pappas J., Ho J., To C., et al. (1998) Differential expression of Met/hepatocyte growth factor receptor in subtypes of non-small cell lung cancers. Lung Cancer 20: 1–16 [DOI] [PubMed] [Google Scholar]
  144. Tsao M.S., Yang Y., Marcus A., Liu N., Mou L. (2001) Hepatocyte growth factor is predominantly expressed by the carcinoma cells in non-small-cell lung cancer. Hum Pathol 32: 57–65 [DOI] [PubMed] [Google Scholar]
  145. Tsao M.S., Zhu H., Giaid A., Viallet J., Nakamura T., Park M. (1993) Hepatocyte growth factor/scatter factor is an autocrine factor for human normal bronchial epithelial and lung carcinoma cells. Cell Growth Differ 4: 571–579 [PubMed] [Google Scholar]
  146. Tsugawa K., Yonemura Y., Hirono Y., Fushida S., Kaji M., Miwa K., et al. (1998) Amplification of the c-met, c-erbB-2 and epidermal growth factor receptor gene in human gastric cancers: correlation to clinical features. Oncology 55: 475–481 [DOI] [PubMed] [Google Scholar]
  147. Tuck A.B., Park M., Sterns E.E., Boag A., Elliott B.E. (1996) Coexpression of hepatocyte growth factor and receptor (Met) in human breast carcinoma. Am J Pathol 148: 225–232 [PMC free article] [PubMed] [Google Scholar]
  148. Turke A.B., Zejnullahu K., Wu Y.L., Song Y., Dias-Santagata D., Lifshits E., et al. (2010) Preexistence and clonal selection of MET amplification in EGFR mutant NSCLC. Cancer Cell 17: 77–88 [DOI] [PMC free article] [PubMed] [Google Scholar]
  149. Tyner J.W., Fletcher L.B., Wang E.Q., Yang W.F., Rutenberg-Schoenberg M.L., Beadling C., et al. (2010) MET receptor sequence variants R970C and T992I lack transforming capacity. Cancer Res 70: 6233–6237 [DOI] [PMC free article] [PubMed] [Google Scholar]
  150. Valente G., Nicotra G., Arrondini M., Castino R., Capparuccia L., Prat M., et al. (2009) Co-expression of plexin-B1 and Met in human breast and ovary tumours enhances the risk of progression. Cell Oncol 31: 423–436 [DOI] [PMC free article] [PubMed] [Google Scholar]
  151. Vigna E., Gramaglia D., Longati P., Bardelli A., Comoglio P.M. (1999) Loss of the exon encoding the juxtamembrane domain is essential for the oncogenic activation of TPR-MET. Oncogene 18(29): 4275–4281 [DOI] [PubMed] [Google Scholar]
  152. Vigna E., Pacchiana G., Mazzone M., Chiriaco C., Fontani L., Basilico C., et al. (2008) ‘Active’ cancer immunotherapy by anti-Met antibody gene transfer. Cancer Res 68: 9176–9183 [DOI] [PubMed] [Google Scholar]
  153. Vuononvirta R., Sebire N.J., Messahel B., Perusinghe N., Reis-Filho J.S., Pritchard-Jones K., et al. (2009) Expression of hepatocyte growth factor and its receptor met in Wilms' tumors and nephrogenic rests reflects their roles in kidney development. Clin Cancer Res 15: 2723–2730 [DOI] [PMC free article] [PubMed] [Google Scholar]
  154. Wakelee, H.A., Gettinger, S.N., Engelman, J.A., Janne, P.A., West, H.J., Subramaniam, D.S. et al. (2010) A phase Ib/II study of XL184 (BMS 907351) with and without erlotinib (E) in patients (pts) with non-small cell lung cancer (NSCLC). In: Proceedings of the 2010 ASCO Annual Meeting, J Clin Oncol 28(15): suppl_3017.
  155. Wang N.S., Liu C., Emond J., Tsao M.S. (1992) Annulate lamellae in a large cell lung carcinoma cell line with high expression of tyrosine kinase receptor and proto-oncogenes. Ultrastruct Pathol 16: 439–449 [DOI] [PubMed] [Google Scholar]
  156. Wang R., Ferrell L.D., Faouzi S., Maher J.J., Bishop J.M. (2001) Activation of the Met receptor by cell attachment induces and sustains hepatocellular carcinomas in transgenic mice. J Cell Biol 153: 1023–1034 [DOI] [PMC free article] [PubMed] [Google Scholar]
  157. Wen P.Y. (2010) American Society of Clinical Oncology 2010: report of selected studies from the CNS tumors section. Expert Rev Anticancer Ther 10: 1367–1369 [DOI] [PubMed] [Google Scholar]
  158. Wen P.Y., Schiff D., Cloughesy T.F., Raizer J.J., Laterra J., Smitt M., et al. (2011) A phase II study evaluating the efficacy and safety of AMG 102 (rilotumumab) in patients with recurrent glioblastoma. Neuro Oncol 13: 437–446 [DOI] [PMC free article] [PubMed] [Google Scholar]
  159. Wong O.G., Nitkunan T., Oinuma I., Zhou C., Blanc V., Brown R.S., et al. (2007) Plexin-B1 mutations in prostate cancer. Proc Natl Acad Sci U S A 104: 19040–19045 [DOI] [PMC free article] [PubMed] [Google Scholar]
  160. Yamamoto S., Tsuda H., Miyai K., Takano M., Tamai S., Matsubara O. (2011) Gene amplification and protein overexpression of MET are common events in ovarian clear-cell adenocarcinoma: their roles in tumor progression and prognostication of the patient. Mod Pathol 24: 1146–1155 [DOI] [PubMed] [Google Scholar]
  161. Yamashita J., Ogawa M., Yamashita S., Nomura K., Kuramoto M., Saishoji T., et al. (1994) Immunoreactive hepatocyte growth factor is a strong and independent predictor of recurrence and survival in human breast cancer. Cancer Res 54: 1630–1633 [PubMed] [Google Scholar]
  162. Yap T.A., Olmos D., Brunetto A.T., Tunariu N., Barriuso J., Riisnaes R., et al. (2011) Phase I trial of a selective c-MET inhibitor ARQ 197 incorporating proof of mechanism pharmacodynamic studies. J Clin Oncol 29: 1271–1279 [DOI] [PubMed] [Google Scholar]
  163. Yi S., Chen J.R., Viallet J., Schwall R.H., Nakamura T., Tsao M.S. (1998) Paracrine effects of hepatocyte growth factor/scatter factor on non-small-cell lung carcinoma cell lines. Br J Cancer 77: 2162–2170 [DOI] [PMC free article] [PubMed] [Google Scholar]
  164. Yi S., Tsao M.S. (2000) Activation of hepatocyte growth factor-met autocrine loop enhances tumorigenicity in a human lung adenocarcinoma cell line. Neoplasia 2: 226–234 [DOI] [PMC free article] [PubMed] [Google Scholar]
  165. Yun C.H., Mengwasser K.E., Toms A.V., Woo M.S., Greulich H., Wong K.K., et al. (2008) The T790M mutation in EGFR kinase causes drug resistance by increasing the affinity for ATP. Proc Natl Acad Sci U S A 105: 2070–2075 [DOI] [PMC free article] [PubMed] [Google Scholar]
  166. Zeng Z.S., Weiser M.R., Kuntz E., Chen C.T., Khan S.A., Forslund A., et al. (2008) c-Met gene amplification is associated with advanced stage colorectal cancer and liver metastases. Cancer Lett 265: 258–269 [DOI] [PMC free article] [PubMed] [Google Scholar]
  167. Zhou C., Wu Y.-L., Chen G., et al. (2010) Efficacy results from the randomised phase III OPTIMAL (CTONG 0802) study comparing first-line erlotinib versus carboplatin (CBDCA) plus gemcitabine (GEM), in Chinese advanced non-small-cell lung cancer (NSCLC) patients (pts) with EGFR activating mutations. ESMO 2010 21: LBA12–LBA12 [Google Scholar]
  168. Zou H.Y., Li Q., Lee J.H., Arango M.E., McDonnell S.R., Yamazaki S., et al. (2007) An orally available small-molecule inhibitor of c-Met, PF-2341066, exhibits cytoreductive antitumor efficacy through antiproliferative and antiangiogenic mechanisms. Cancer Res 67: 4408–4417 [DOI] [PubMed] [Google Scholar]

Articles from Therapeutic Advances in Medical Oncology are provided here courtesy of SAGE Publications

RESOURCES