139
Views
82
CrossRef citations to date
0
Altmetric
Review

Emerging molecular targets in oncology: clinical potential of MET/hepatocyte growth-factor inhibitors

, &
Pages 1001-1014 | Published online: 12 Jun 2014

Abstract

The MET/hepatocyte growth-factor (HGF) signaling pathway plays a key role in the processes of embryogenesis, wound healing, and organ regeneration. Aberrant activation of MET/HGF occurs through multiple mechanisms including gene amplification, mutation, protein overexpression, and abnormal gene splicing interrupting autocrine and paracrine regulatory feedback mechanisms. In many cancers including non-small-cell lung cancer, colorectal, gastric, renal, and hepatocellular cancer, dysregulation of MET may lead to a more aggressive cancer phenotype and may be a negative prognostic indicator. Successful therapeutic targeting of the MET/HGF pathway has been achieved using monoclonal antibodies against the MET receptor and its ligand HGF in addition to MET-specific and multitargeted small-molecule tyrosine-kinase inhibitors with several drugs in late-phase clinical trials including onartuzumab, rilotumumab, tivantinib, and cabozantinib. MET frequently interacts with other key oncogenic tyrosine kinases including epidermal growth-factor receptor (EGFR) and HER-3 and these interactions may be responsible for resistance to anti-EGFR therapies. Similarly, resistance to MET inhibition may be mediated through EGFR activation, or alternatively by increasing levels of MET amplification or acquisition of novel “gatekeeper” mutations. In order to optimize development of effective inhibitors of the MET/HGF pathway clinical trials must be enriched for patients with demonstrable MET-pathway dysregulation for which robustly standardized and validated assays are required.

MET signaling pathways and function in healthy tissue

The MET proto-oncogene was first identified in a chemically transformed osteosarcoma-derived cell line in 1984, and its protein product was subsequently found to describe a receptor tyrosine kinase the ligand for which was identified as hepatocyte growth factor (HGF; or scatter factor).Citation1Citation3 Ligand-dependent activation by binding of HGF to MET leads to receptor dimerization and phosphorylation of three kinase-domain tyrosine residues which then initiate the process of autophosphorylation of tyrosine (Tyr) 1349 and Tyr1356 in the bidentate substrate-binding site, facilitating recruitment of cytoplasmic effector proteins and activating transmembrane signaling.Citation4 Downstream signaling effects are transmitted via mitogen-activated protein kinase (MAPK), phosphoinositide 3-kinase (PI3K)/Akt (protein kinase B), signal transducer and activator of transcription proteins (STAT), and nuclear factor-κB.Citation5Citation7 The final output of the terminal effector components of these pathways is activation of cytoplasmic and nuclear processes leading to increases in cell proliferation, survival and mobilization, and invasive capacity.Citation8

The MET/HGF signaling pathway plays a key role in hepatocyte and placental formation during embryogenesis, and additionally in voluntary muscle and central nervous system formation.Citation9Citation12 The effects of MET/HGF are critical for wound healing and organ regeneration; signaling through this pathway encourages proliferation of keratinocytes and their mobilization into de-epithelized zones, and increased levels of HGF produced in response to injury by hepatocytes and renal epithelial cells leads to mitotic and antiapoptotic activity.Citation13Citation15 These constitutive effects of MET on proliferation, apoptosis, and migration are subverted during the process of tumor growth and metastasis leading to an aggressive MET-addicted tumor phenotype.

MET activation in cancer

Aberrant MET signaling is a hallmark of multiple cancer types, and may occur through gene amplification or mutation, protein overexpression, or abnormal gene splicing which interrupt normal autocrine and paracrine regulatory feedback mechanisms.Citation6 Missense mutations of MET have been demonstrated in the germ line of families with a history of hereditary papillary renal cell carcinoma (RCC) and in the tumors of a subset of sporadic papillary renal cancers.Citation16 Production of mouse models with an activating mutation replacing endogenous MET yielded diverse cancers including carcinomas, lymphomas, and sarcomas, providing proof of concept of oncogenic activity for the mutated genotype.Citation17 MET amplification on chromosome 7q31 has been described in gastroesophageal, colorectal, and endometrial carcinoma, medulloblastoma, non-small-cell lung cancer (NSCLC), and glioma.Citation18Citation23 Overexpression of the protein receptor tyrosine kinase is more common than amplification, and has been demonstrated in all tumor types with gene amplification in addition to breast, cervical, head and neck, renal, hepatocellular, melanoma, thyroid, and mesothelioma cancer types.Citation24

MET also interacts with other key oncogenic signaling pathways, in particular HER2 (human epidermal growth factor receptor 2) superfamily members epidermal growth-factor receptor (EGFR) and HER-3. For example, cells that express EGFR and MET demonstrate ligand-independent MET phosphorylation and activation through EGFR, whereas in EGFR-mutant NSCLC, MET amplification leads to escape from gefitinib sensitivity by HER3-mediated activation of PI3K signaling.Citation25,Citation26 In Kirsten rat sarcoma (KRAS) wild-type colorectal cancer cell lines overexpression of the EGFR ligand TGFα (transforming growth factor-α) leads to MET activation and cetuximab resistance, and MET amplification appears to be a resistance mechanism for colorectal cancer patients treated with anti-EGFR antibody therapy.Citation27,Citation28 The MET pathway also increases the malignant potential of tumors through induction of angiogenesis; MET/HGF is a potent inducer of vascular endothelial growth factor (VEGF)-A production and suppressor of thrombspondin-1, and acts synergistically with the VEGF receptor (VEGFR) through common downstream signaling molecules to increase neovascularization activity.Citation7,Citation29 Finally, there appears to be an emerging role for MET/HGF signaling in maintaining the stem cell niche in cancer; Wnt activity in colorectal cancer stem cells has been described to be supported by myofibroblast-secreted HGF.Citation30 These interconnected and diverse functions underlie the key role of the MET/HGF axis in driving tumor growth and supporting an intercellular milieu that is conducive to the metastatic spread of the primary tumor.

Development of MET-inhibitor therapies

Greater understanding of the structure, function, and role of MET/HGF in cancer has led to the development of multiple compounds targeting this pathway. These include monoclonal antibodies targeting the receptor and ligand, and small-molecule tyrosine-kinase inhibitors (TKIs) functional at an intracellular level. Monoclonal antibodies in clinical trials include onartuzumab (MetMab; Roche, Basel, Switzerland), rilotumumab (Amgen, Thousand Oaks, CA, USA) and ficlatuzumab (Aveo Pharmaceuticals, Cambridge, MA, USA). Onartuzumab, a human immunoglobulin (Ig)-G1 antibody with murine variable domains is a potent MET antagonist that competes with HGF for binding at that site.Citation31 Rilotumumab and ficlatuzumab are fully humanized monoclonal anti-HGF antibodies that block HGF binding to MET.Citation32 Onartuzumab and rilotumumab bind to the Sema and SPH (serine protease-homology) domains of MET and HGF respectively, and the monovalent binding design of onartuzumab has been demonstrated to prevent activation of the receptor induced by dimerization which may occur with bivalent antibodies.Citation33

The majority of small-molecule inhibitors of MET may be classified as one of three subtypes each of which impedes adenosine triphosphate (ATP) binding, but of which type II and type III also occupy other distinct binding sites within the MET receptor.Citation6 Most type I inhibitors (eg, crizotinib) preferentially bind to the inactive form of the enzyme and are therefore ineffective against tumors harboring an activating Tyr1230H mutation. Type I inhibitors are most specific for the MET kinase; however, crizotinib has also demonstrated significant efficacy against NSCLC tumors harboring echinoderm microtubule-associated protein-like 4–anaplastic lymphoma kinase (EML4-ALK) fusion and ALK-positive large-cell lymphomas.Citation34,Citation35 Type II inhibitors are more promiscuous: in addition to MET, foretinib is an inhibitor of AXL, RON (Recepteur d’Origine Nantais), VEGFR2, PDGFR (platelet-derived growth-factor receptor-β), and KIT.Citation36,Citation37 Cabozantinib is a multitargeted TKI with activity against MET, VEGFR2, RET, KIT, AXL, tyrosine kinase with immunoglobulin-like and EGF-like domains (TIE)-2 and FLT, and is active against both the active and inactive conformations of MET.Citation38 Tivantinib, which cannot be accurately classified into any of these subtypes, is a non-ATP competitive MET inhibitor that impedes ligand-dependent and -independent activation.Citation39

MET in lung cancer

In NSCLC overexpression of MET occurs in up to 60% of tumor tissues.Citation40 The most common mechanism of MET activation is protein expression secondary to transcriptional upregulation.Citation41 Although less common, amplification and mutation of the MET gene are alternative mechanisms leading to MET activation; MET amplification has been reported in 1%–21% of cases, whereas mutations of the MET gene occur less frequently and usually lead to activation of the HGF/MET pathway through alternative splice forms deleting the juxtamembrane domain.Citation22,Citation42Citation45 Although MET activation does not appear to be associated with specific clinicopathological characteristics studies have consistently reported an association of both MET amplification/overexpression and intratumoral/plasma HGF levels with poor prognosis.Citation43,Citation46Citation51 Moreover, the HGF/MET pathway also plays an important role in mediating resistance to EGFR TKIs through the activation of both PI3K/Akt and extracellular signal-regulated kinase (ERK) signaling.Citation52,Citation53 Preclinical data suggest that combining EGFR TKIs and MET inhibitors is a promising strategy to restore gefitinib sensitivity in cell lines.Citation26,Citation54

In recent years, several inhibitors of the HGF/MET pathway have been investigated in NSCLC, largely in combination with EGFR TKIs. Onartuzumab (the monoclonal antibody that competes with HGF for MET binding) has been examined in a randomized Phase II trial of erlotinib ± onartuzumab in EGFR-unselected, chemorefractory, NSCLC patients (). A total of 137 patients were enrolled, and no differences in survival outcomes were observed in the non-molecularly selected intention-to-treat population.Citation55 However, in a prespecified subgroup analysis of MET-positive patients (n=66), the combination of onartuzumab plus erlotinib was associated with a significant improvement in both progression-free survival (PFS; hazard ratio [HR] 0.53, P=0.04) and overall survival (OS; HR 0.37, P=0.002) compared to erlotinib alone. Outcomes were inferior for MET-negative patients treated with onartuzumab compared to those treated with erlotinib alone (PFS HR 1.71, P=0.06; OS HR 2.61, P=0.004). A randomized Phase III trial of erlotinib ± onartuzumab in MET-positive patients was initiated but recruitment was halted for futility following a planned interim analysis.Citation56 In contrast, a trial of ficlatuzumab, the humanized IgG1 antibody, has provided contrasting results in a similar patient population.Citation57 Despite promising preliminary activity observed in Phase I trials, a randomized Phase II trial of gefitinib ± ficlatuzumab in 188 treatment-naïve Asian patients with high incidence of sensitizing EGFR mutations failed to demonstrate superiority of the combination therapy.Citation58Citation60 PFS was 5.6 months in the combination arm versus 4.7 months in the gefitinib-alone arm. Interestingly, in contrast to the results reported in the trial with onartuzumab, a subgroup analysis showed that the addition of ficlatuzumab to gefitinib seemed to benefit more those patients with low tumoral MET expression (7.3 versus 2.8 months), and this difference appeared to be more pronounced in the presence of tumors with sensitizing EGFR mutations. However, as the patients in this subset analysis were fewer than 20 it is difficult to draw any firm conclusions from these data.

Table 1 Selected MET-inhibitor clinical trials with biomarker-selected results if available

Mixed results have also been demonstrated for tivantinib (ARQ 197; ArQule, Woburn, MA, USA), the non-ATP-competitive small-molecule MET inhibitor that exerts its antitumor activity by stabilizing the inactive conformation of MET.Citation39 A randomized Phase II trial of erlotinib with or without tivantinib in 167 previously treated patients with EGFR TKI-naïve NSCLC failed to meet its primary end point of an improvement in PFS in the intention-to-treat non-biomarker-selected patient population.Citation61 In this study, a numerically better but not statistically significant improvement in PFS (3.8 versus 2.3 months, HR 0.81; P=0.24) was reported for the combination therapy. However, in a prespecified proportional hazards model assessing PFS, this difference was found to be statistically significant (HR 0.68, P=0.04). Moreover, in another prespecified exploratory analysis, a significant improvement in PFS was also observed in the group of patients with KRAS-mutant tumors (HR 0.18, P-value for interaction =0.006). Based on the evidence of a trend toward an increased benefit from tivantinib in patients with nonsquamous histology and tumors with high MET gene copy number, a randomized Phase III trial of erlotinib ± tivantinib in pretreated, nonsquamous NSCLC patients was initiated. Although a significant improvement in PFS for the combination treatment was observed, the trial was discontinued early on the advice of the independent data-monitoring committee for futility, as the desired difference in the primary end point of OS would not be reached.Citation24 Other Phase II and III clinical trials investigating tivantinib in combination with erlotinib in NSCLC are ongoing; these include those in the EGFR wild-typeCitation62 and KRAS-mutantCitation63 populations.

MET in colorectal cancer

Activation of MET is a common feature in colorectal cancer and upregulation of the HGF/MET pathway, as expressed by MET messenger ribonucleic acid (mRNA) or protein overexpression has been consistently reported in this setting.Citation64Citation66 In contrast and in keeping with other malignancies, ligand-independent MET activation by amplification or mutation has only been reported in a minority of cases.Citation41,Citation67 In colorectal cancer MET appears to have an important role in tumor progression and has also been reported to be a negative prognostic indicator.Citation23,Citation68Citation70 Multiple series demonstrate that MET activation in colorectal cancer may provide a selective advantage for the acquisition of an aggressive phenotype that correlates with early stage invasion, liver metastases, and unfavorable clinical outcomes.Citation23,Citation68Citation71

Preclinical data suggest that HGF-induced MET activation may represent an alternative, RAS-independent mechanism of resistance to cetuximab through the reactivation of the MAPK and Akt pathways. Stimulation with HGF was shown to inhibit the antiproliferative effects of EGFR inhibition, while MET inhibition abrogated this effect.Citation72 These preliminary findings of the importance of MET in resistance to anti-EGFR therapy in colorectal cancer have been confirmed in a recent study where MET amplification emerged as a novel mechanism of both primary and secondary resistance to EGFR-targeted antibodies, possibly accounting for >10% of cetuximab-resistant cases that are wild type for KRAS, BRAF, NRAS (neuroblastoma RAS), PIK3CA (phosphatidylinositol-4,5-bisphosphate 3-kinase, catalytic subunit alpha), and HER2.Citation28 Interestingly, MET amplification in patients who progressed on anti-EGFR agents was either a new molecular finding in posttreatment tumor samples or the result of the expansion of a preexisting minor subclonal population of MET-amplified cancer cells under the selective pressure of an EGFR-targeted therapy.

Several MET inhibitors have been tested in colorectal cancer or are currently under investigation; however, most of the available data relate to the monoclonal antibody rilotumumab and the selective, non-ATP-competitive MET TKI tivantinib. In a three-arm, randomized Phase IB/II trial (n=142) of panitumumab (Vectibix®; Amgen) in combination with rilotumumab (anti-HGF antibody), ganitumumab (IGF-1R [insulin-like growth-factor receptor-1 inhibitor]) or placebo, the use of the combination treatment including rilotumumab showed promising activity (overall response rate 31% versus 21%; PFS 5.2 versus 3.7 months) compared to single-agent panitumumab in patients with chemorefractory tumors.Citation73 In a biomarker analysis of 91 of 96 patients allocated to panitumumab ± rilotumumab a correlation between MET expression and activity of rilotumumab was found only when MET-staining intensity (≥2+ versus ≤1+) rather than percentage of MET-positive cells (>50% versus ≤50%) was considered. The anti-MET monoclonal antibody onartuzumab (MetMab) is currently being investigated in a randomized, double-blind, placebo-controlled, Phase II study in conjunction with bevacizumab plus mFOLFOX-6 in chemonaïve metastatic colorectal cancer patients;Citation74 recruitment has completed to this study and results are currently pending.Citation75

As a result of promising signals of activity of the anti-MET TKI tivantinib in a Phase IB study in colorectal cancer where four of nine patients had an objective response, a combination study of irinotecan–cetuximab (Erbitux, Merk-Serono) with or without this drug was investigated in a randomized, Phase II trial in a population of KRAS wild-type metastatic colorectal cancer patients (n=122) who had progressed on or after one line of systemic therapy.Citation76,Citation77 Tivantinib in combination with standard treatment was associated with a higher response rate (45% versus 33.3%) and a slight improvement in PFS (8.3 versus 7.3 months, respectively); however this was not statistically significant (PFS HR 0.85, P=0.38). Surprisingly, but in line with what has been observed with ficlatuzumab in NSCLC, a subgroup analysis conducted in a small number of study patients showed a statistically significant improvement in PFS with tivantinib in patients with low-MET-expressing tumors; however this subgroup contained only 23 patients and requires validation in a larger patient cohort. Tivantinib is currently under investigation in conjunction with cetuximab in a Phase II study in advanced colorectal cancer patients who are refractory to anti-EGFR therapy and who demonstrate high MET (immunohistochemistry [IHC] 2+ or IHC 3+) expression.Citation74

Esophagogastric cancer

The MET pathway is frequently aberrantly activated in gastric carcinoma with evidence of both overexpression and amplification demonstrated in multiple series. Overexpression of MET appears to be more common than amplification, and may be membranous or cytoplasmic. In a recent series of 495 Korean patients membranous and cytoplasmic overexpression of MET was demonstrated in 22% of cases and membranous expression alone in 8%. Simultaneous overexpression correlated well with increased MET mRNA expression, copy-number gain, and amplification, and was prognostic with respect to OS and disease-free survival; however, membranous MET expression alone did not significantly correlate with these end points.Citation78 In another large Asian series (n=438), Lee et al demonstrated a 24% prevalence of overexpression (IHC 2+ and 3+) combined and a 3.4% rate of copy-number gain using silver in situ hybridization. Almost all (>90%) patients with MET IHC 3+ overexpression demonstrated gene amplification, with the remainder demonstrating high gene polysomy.Citation79 Furthermore, all patients with gene amplification demonstrated either IHC 2+ or 3+ overexpression. Therefore it appears that in comparison to overexpression MET amplification in gastric cancer is a relatively infrequent event with rates ranging from 2% to 10%.Citation78Citation81 Of note, earlier studies examining levels of amplification are likely to have overestimated the incidence of this event due to methodological considerations as these studies frequently used Southern blot or quantitative polymerase chain-reaction techniques which are unable to distinguish between true gene amplification and polysomy, whereas this poses less difficulty with current in situ hybridization techniques.Citation18,Citation19,Citation82

There is particular difficulty in assessing the consistency of measurement of MET overexpression across studies as different investigators have used either varying percentage of cell staining alone as a measure, whereas others have used both percentage of cells and intensity of staining and various composites of these to yield an H-score.Citation83 This is also true of amplification as the level of copy-number gain which has been designated as MET high is between two and five times the level of a reference gene depending on the series.Citation79,Citation80 A recent meta-analysis referencing 15 studies and 2,210 patients attempted to synthesize the prognostic effects of both overexpression and amplification across multiple studies. Both overexpression and amplification were demonstrated to be associated with inferior OS, with HR =2.66 and HR =1.66, respectively.Citation83 This was true for Western and Asian populations, and the prognostic effect of MET was also independent of stage. An additional important consideration when applying these data to potential clinical trial design is the fact that the pattern of MET copy-number alteration in gastric cancer (using high-resolution single-nucleotide polymorphism arrays) appears to be predominantly mutually exclusive of amplification of other relevant receptor tyrosine-kinase genes (FGFR, ERBB2, KRAS, and EGFR).Citation84

Abrogation of MET-pathway signaling in gastric cancer has been successful using both small-molecule TKIs and monoclonal antibody therapy. In the initial Phase I study of tivantinib (the orally available tyrosine kinase MET inhibitor) in a non-molecularly selected population minor regression was noted in a patient with gastric cancer with stable disease for 15 weeks duration.Citation85 Early reports of efficacy of crizotinib in a MET-amplified patient cohort were described by Lennerz et al who reported responses in two of four patients treated with crizotinib in a Phase I trial enriched for MET-amplified patients.Citation81 Additionally, a case report detailing a complete and durable response in a female gastric cancer patient with high MET polysomy and MET overexpression was reported during the Phase I trial of onartuzumab.Citation86 This patient was treated with single-agent onartuzumab at a dose of 20 mg/kg every 3 weeks with a complete response demonstrated following four doses. Unsurprisingly, results of MET inhibition have been less promising in unselected patient populations. Foretinib, a multitargeted TKI targeting MET, RON, AXL, TIE-2, and VEGFR2 failed to demonstrate activity in a largely non-MET-amplified gastric cancer patient population previously treated with chemotherapy.Citation87 In this Phase II study, 69 evaluable patients were treated with foretinib either on an intermittent (240 mg/day for 5 consecutive days every 2 weeks) or daily dosing (80 mg/day during each 2-week cycle) schedule until progression. No patient in either cohort demonstrated a complete or partial response and 23% and 20% of patients in the intermittent and daily dosing cohorts respectively had a best response of stable disease. Three patients in this study were MET-amplified by FISH (fluorescence in situ hybridization): one was unevaluable due to toxicity, one had progressive disease, and one had stable disease of short duration (2.1 months). A Phase II study evaluating the addition of the anti-HGF monoclonal antibody rilotumumab to epirubicin + cisplatin + capecitabine (Xeloda®, Roche) (ECX) chemotherapy in a non-MET-selected population has been reported in abstract form. A total of 121 patients with treatment-naïve advanced gastroesophageal cancer were randomized to ECX chemotherapy plus either placebo or rilotumumab at two dose levels (7.5 mg/kg or 15 mg/kg). In the 90 patients with evaluable MET expression, patients with MET-high tumors (>50% cells with MET expression) had superior survival when treated with rilotumumab than those with MET-low tumors (OS 11.1 versus 5.7 months, HR 0.29; P=0.012). Conversely, patients with MET-low tumors treated with chemotherapy plus rilotumumab had a trend toward worse survival.Citation88 An exposure–response analysis presented at the same meeting demonstrated that increased exposure to rilotumumab in MET-high patients was associated with improvements in PFS and OS in that patient group.Citation89 Both onartuzumab and rilotumumab are currently in global Phase III randomized trials in advanced esophagogastric cancer with MET overexpression: onartuzumab in conjunction with FOLFOX chemotherapyCitation90 and rilotumumab with ECX.Citation91 Multiple MET-targeting TKIs are also currently under evaluation in clinical trials in this setting.

Hepatocellular carcinoma

The MET/HGF pathway has been attributed an important role in the genesis and maintenance of hepatocellular carcinoma, and has emerged as an attractive therapeutic target for this disease. In hepatocellular carcinoma MET overexpression has been reported in 20%–48% of cases.Citation92Citation94 This phenomenon has not been consistently associated with gene amplification, suggesting that for hepatocellular carcinoma alternative mechanisms including autocrine or paracrine HGF-induced activation or epigenetic regulation of expression may account for a significant number of MET-overexpressing tumors.Citation95,Citation96 In studies investigating the correlation between MET expression and clinicopathological features or clinical outcome in hepatocellular carcinoma MET has been largely shown to correlate with aggressive tumor phenotype and poor survival in both the early stage and advanced setting.Citation97Citation100 A possible association of MET overexpression with favorable clinical characteristics as suggested by other studies, is likely to be due to the small number of patients analyzed, heterogeneity of the patient populations, or differences in study methodology.Citation96,Citation101

In vitro and in vivo studies demonstrate that MET overexpression is associated with the development of hepatocellular carcinoma, while knockdown of MET leads to the inhibition of tumor growth and regression of advanced tumors.Citation102Citation104 The promising results observed with MET inhibition in preclinical studies of hepatocellular carcinoma raised interest in assessment of MET as a therapeutic target in the clinical setting, in particular because effective systemic treatment options are limited for patients with this disease.Citation39,Citation103,Citation104 Several selective MET inhibitors are under development and being tested in early stage clinical trials; however tivantinib (ARQ197; Aveo) is the agent with the majority of clinical data available. In a randomized, double-blind, placebo-controlled, crossover Phase II trial, 74 patients with advanced, Child–Pugh A hepatocellular carcinoma previously treated with one systemic therapy were randomly allocated in a 2:1 ratio to receive oral tivantinib or placebo.Citation100 Although clinically marginal, a statistically significant improvement in median time to progression (1.6 versus 1.4 months, HR 0.64; P=0.04) was observed in favor of tivantinib. Importantly, a prespecified subgroup analysis indicated that MET overexpression may represent a potential predictive biomarker for tivantinib benefit as the most clinically and statistically significant tivantinib effects in terms of tumor stabilization (50% versus 20%), time to progression (2.7 versus 1.4 months, HR 0.43; P=0.03) and OS (7.2 versus 3.8 months, HR 0.38; P=0.01) were observed in the group of patients with MET-overexpressing tumors. However, given the modest activity of the drug in the unselected population and the small numbers of patients assessed for MET expression in the subgroup analysis (n=22), confirmatory evidence of clinical benefit will be sought in a Phase III randomized trial comparing tivantinib with placebo in pretreated patients with MET-overexpressing tumors.Citation105

Other multitargeted TKIs with activity against MET have also recently been investigated in hepatocellular carcinoma.Citation106Citation108 In particular, in a Phase II randomized discontinuation trial cabozantinib (an oral inhibitor of MET and VEGFR2), was investigated in 41 patients with hepatocellular carcinoma half of whom had been previously treated with sorafenib.Citation106 Although only 5% of patients demonstrated a partial response at 12 weeks prior to the randomization, the overall disease-control rate (partial response + stable disease) at this time point was 68%, and 38% of patients with serial α-fetoprotein measurements demonstrated a decline of >50% from baseline. These encouraging results which may in part have been driven also by the antiangiogenic properties of this drug, have led to the development of a large Phase III controlled trial of cabozantinib versus placebo in hepatocellular carcinoma patients previously treated with sorafenib.Citation109 The monoclonal antibody onartuzumab is also being investigated in conjunction with sorafenib in the first line setting for patients with hepatocellular carcinoma.Citation110

Prostate cancer

MET expression in prostate cancer is associated with high-grade tumors and the presence of metastases, in particular bone metastases, and in prostate cancer cell lines MET expression is inversely correlated with expression of the androgen receptor.Citation111,Citation112 The androgen receptor has been demonstrated to be a negative regulator of MET, and accordingly the effect of small-molecule MET inhibitors has been demonstrated to be more potent in androgen-insensitive prostate cancer cells.Citation113,Citation114 Cabozantinib, an inhibitor of MET, VEGFR, and multiple other tyrosine kinases, was investigated in a randomized discontinuation study in advanced castration-resistant prostate cancer at a dose of 100 mg daily; patients with stable disease by response-evaluation criteria in solid tumors (RECIST) at 12 weeks were randomized to cabozantinib or placebo.Citation115 Recruitment was halted following enrollment of 171 patients due to efficacy in the experimental arm of the trial. Although the overall response rate at 12 weeks was 5%, an additional 75% of patients had stable disease, of whom 31 were randomized at week 12. PFS was 23.9 weeks for men treated with cabozantinib, and 5.9 weeks for those receiving placebo (HR 0.12, P<0.001). Bone pain and narcotic use were also significantly decreased in the majority of patients. Dose reductions were frequent (51% at 12 weeks) in this initial study and a subsequent dose-ranging study demonstrated superior tolerability and comparable efficacy for a 40 mg daily dose which was recommended for subsequent randomized clinical trials.Citation115,Citation116

Significant resolution of bone lesions on bone scan has been a notable effect of cabozantinib in prostate cancer trials; it has recently been demonstrated that in addition to direct cytotoxic effects on prostate cancer cells, cabozantinib has an inhibitory effect on osteoclast production and a biphasic dose-dependent effect on osteoblast activity both mediated through MET and VEGFR signaling.Citation117 Therefore, the effects of cabozantinib on bone scintigraphy are as a result of cytotoxicity in addition to direct effects on bone remodeling.

Cabozantinib is currently under investigation in several large randomized studies in metastatic castration-resistant prostate cancer in previously treated patientsCitation118,Citation119 and in combination with abiraterone in patients who are treatment-naïve.Citation120 However, the addition of rilotumumab to mitoxantrone and prednisolone therapy in metastatic castration-resistant prostate cancer patients previously treated with docetaxel did not result in any improvements in PFS or OS when compared to standard therapy (PFS 3.0 versus 2.9 months, OS 12.2 versus 11.1 months, respectively), including in MET-high (n=38) patients.Citation121 Therefore it was not recommended that rilotumumab proceed to a Phase III trial in this setting.

Renal cell carcinoma

The MET pathway is activated through at least two separate mechanisms in RCC of distinct histological subtypes. In clear-cell RCC inactivation of the VHL gene is common, and preclinical data suggest that this may induce constitutive phosphorylation of MET leading to enhanced cell mobilization and invasive capacity.Citation122 MET expression is common in RCC and associated with a negative prognosis; in a recent study examining MET expression on 330 RCC cores, expression was highest in papillary and sarcomatoid subtypes and those with a higher Fuhrman grade but was also present on clear-cell RCC, and in an analysis limited to clear-cell subtypes remained a negative prognostic marker.Citation123 In MET-activated clear-cell RCC cell lines treatment with tivantinib led to inhibition of cell proliferation providing a clinical rationale for targeting MET-activated clear-cell RCC with these agents.

A Phase II study with the anti-HGF monoclonal antibody rilotumumab was conducted in 61 patients with metastatic RCC of varying histologies (clear-cell 75.4%, papillary 11.5%), the majority of whom had previously received antiangiogenic therapy.Citation124 Although one partial response was maintained for 2.5 years no other responses were seen, median PFS was 3.7 months at 10 mg/kg and 2.0 months at 20 mg/kg rilotumumab doses and tumoral MET expression was not associated with response or survival outcomes. As a result, further development of rilotumumab has not been pursued in this disease.

The antiangiogenic properties of the TKI cabozantinib make this an attractive agent for treatment of RCC. Promising results in clear-cell RCC patients were seen in a drug–drug interaction study examining the effects of rosiglitazone on cabozantinib pharmacokinetics; of 25 patients treated with a median of two prior treatments, 24% had a confirmed partial response by RECIST, and 86% experienced some tumor regression.Citation125 These encouraging results have led to the development of multiple clinical trials investigating cabozantinib in clear-cell RCC: in comparison to everolimus in a Phase II randomized study for patients who have previously progressed following TKI therapy,Citation126 and in comparison to sunitinib in previously untreated patients.Citation127

A second mechanism of MET activation is seen in the papillary subtype of renal cancer, with activating mutations of MET found in the germ line of families with hereditary papillary RCC and in a proportion of sporadic noninherited cases. In a nonrandomized study assessing the effect of the nonselective MET inhibitor foretinib 74 patients with papillary RCC were recruited, eleven of whom had germ-line and five of whom had somatic MET mutations.Citation128 Two patients demonstrated MET amplification with no mutation. Median PFS was 9.3 months and 1-year survival was 70% with median OS not reached. Of the ten patients with a germ-line mutation, half had a partial response and half had stable disease, whereas only one of five patients with a somatic mutation had a response and no MET amplified patient did. Although the trial failed to meet its primary end point of a response rate of >25% the response rate in germ-line-mutant patients is noteworthy, and MET inhibition would appear to be worthwhile in this patient group.

Toxicity of MET inhibition

The extracellular inhibitors of the MET pathway (onartuzumab, rilotumumab, and ficlatuzumab) appear to be well tolerated, with relatively few treatment-related serious adverse events reported in clinical trials to date. In the Phase I studies for both onartuzumab and rilotumumab, the maximum tolerated dose was not reached.Citation129,Citation130 Peripheral edema appears to be a class effect of these compounds, and increased rates of neutropenia have been demonstrated when rilotumumab is used in conjunction with chemotherapy.Citation88 Activation of the MET pathway has been associated with dysregulation of the clotting cascade in preclinical models; however, with the caveat of relatively small control groups treated to date, significant differences in the incidence of thromboembolic disease have not been noted with these drugs.Citation131 Class-effect toxicities associated with nonselective tyrosine kinase inhibition (fatigue, gastrointestinal upset, hepatotoxicity) are frequent but usually mild.Citation87,Citation115 However, awareness of toxicity related to off-target effects, such as those on VEGFR (hypertension, hemorrhage, perforation) is also necessary as these may be significant.Citation115 Additionally, tivantinib appears to have cytotoxic effects that are independent of its MET-inhibitory activity and significant rates of neutropenia and neutropenia-related deaths have been documented with the use of this compound.Citation100,Citation132

Resistance to MET inhibition

Acquisition of novel mutations, redundancy in intracellular signaling pathways, and downregulation of inhibitory feedback mechanisms have been demonstrated to be responsible for de novo and acquired resistance to other TKIs, such as those inhibiting EGFR, BRAF and mitogen-activated protein-kinase kinase (MEK). The mechanisms by which resistance to MET inhibition may occur have recently begun to emerge, and preeminent among these is the interplay between the MET and the EGFR pathways. In MET-amplified gastric cancer lines treated with the MET inhibitor PHA-665752, EGF, and heregulin-dependent activation of EGFR and HER3 led to downstream effects on the MAPK and PI3K pathways and abrogation of the effects of MET inhibition.Citation133 However, combined blockade of MET and EGFR using gefitinib or with MEK and Akt inhibitors led to reversal of MET resistance. In a separate experiment, resistance to MET therapy in SNU6838 cells was mediated through TGFα expression and EGFR activation.Citation134 Similarly, activation of the EGFR pathway has been demonstrated to be responsible for acquired resistance to the MET inhibitor PF2341066 in MET-amplified NSCLC lines and while combination therapy with PF2341066 and the EGFR inhibitor erlotinib did not result in decreased cell proliferation, it did suppress emergence of MET resistance.Citation135

Alternative escape mechanisms from MET inhibition include increased amplification of MET, acquisition of mutations affecting binding-site conformation, and upregulation of non-EGFR-signaling pathways. In MET-amplified gastric (GTL16) and NSCLC (EBC-1) cell lines when initially sensitive cells were treated with either of two MET inhibitors (PHA-665752 or JNJ38877605), the MET gene acquired further amplification with subsequent increased levels of protein expression leading to adequate levels of phosphorylation which successfully maintained enzymatic activity.Citation136 However, at higher levels of drug which overcame the increased MET amplification, amplification and overexpression of KRAS emerged and this remained sensitive to downstream inhibition of MAPK elements using U0126 and PD0325901. An additional pathway by which MET amplified gastric cancer cell line GTL16-acquired resistance to MET inhibition with PF04217903 is the emergence of a novel SND1 (staphylococcal nuclease domain 1)–BRAF fusion protein that uses activated BRAF to escape MET suppression.Citation137 Again, the activity of this resistance mechanism could be suppressed through combined MET and BRAF or MEK inhibition. Further evidence of the efficacy of combination therapy in overcoming resistance is demonstrated by NSCLC cell lines resistant to erlotinib and the MET inhibitor SU11274, which display upregulation of both mTOR (mammalian target of rapamycin) and Wnt pathway components and restoration of sensitivity to EGFR/MET inhibition by the addition of everolimus.Citation138

A final mechanism of resistance in gastric cancer cell lines has been demonstrated when MET-amplified SNU6838 gastric cancer cell lines were treated with the MET inhibitors PHA-665752 and PF2341066; a novel mutation occurred in the activation loop of MET, causing a conformal change that blocked inhibitor binding analogous to the gatekeeper mutations seen in EGFR (T790M) following erlotinib treatment and in ABL (T315I) following imatinib.Citation134 Although the MET Y1230H mutation renders cancers insensitive to type I MET inhibitors, conformal differences between these and type II compounds may allow treatment of MET Y1230H mutant cancers or prevent the emergence of resistance due to the mutation.Citation139,Citation140

Conclusion and future directions

The ubiquity of MET-pathway activation in cancer and the malignant phenotype that it confers on MET-mutated, -amplified, or -overexpressed tumors ensure that this is an attractive therapeutic target for many cancers. Pharmacological inhibition of this pathway has clear benefits in terms of response and survival, albeit in limited numbers to date. It is clear that to optimize these benefits clinical trials must be enriched for patients with demonstrable MET-pathway dysregulation; what is less clear is the best means by which to achieve this. Robust standardization and validation of assay methodology for MET expression is essential in order to confidently address the benefit of MET inhibition across distinct patient populations, and assessment of the correlation between gene amplification, protein expression, and treatment efficacy is also mandated. With respect to clinical trial development, treatment with anti-MET/HGF antibodies and chemotherapy and/or other antibodies appears to be an attractive option given the lack of significant additive toxicities seen for combination regimens, whereas the small-molecule TKIs may potentially be combined with other similar drugs targeting other relevant pathways. These combinatorial approaches may be designed in order to delay or prevent the emergence of resistance to MET inhibition via intimately connected pathways, such as EGFR, HER3, and RAS. Ultimately, collaborative clinical trials and serial tissue collection will be required in order to fully evaluate the impact of inhibition of this promising target on oncology outcomes.

Acknowledgments

We acknowledge support from the National Institute for Health Research Royal Marsden/Institute for Cancer Research Biomedical Research Centre.

Disclosure

Dr Smyth and Dr Sclafani declare no relevant conflicts. Professor Cunningham has received research funding from Roche, Amgen, Celgene, Sanofi, Merck Serono, Novartis, and Astra Zeneca.

References

  • CooperCSParkMBlairDGMolecular cloning of a new transforming gene from a chemically transformed human cell lineNature1984311598129336590967
  • GiordanoSPonzettoCDi RenzoMFCooperCSComoglioPMTyrosine kinase receptor indistinguishable from the c-met proteinNature198933962201551562541345
  • NaldiniLVignaENarsimhanRPHepatocyte growth factor (HGF) stimulates the tyrosine kinase activity of the receptor encoded by the proto-oncogene c-METOncogene1991645015041827664
  • PonzettoCBardelliAZhenZA multifunctional docking site mediates signaling and transformation by the hepatocyte growth factor/scatter factor receptor familyCell19947722612717513258
  • SipekiSBanderEBudayLPhosphatidylinositol 3-kinase contributes to Erk1/Erk2 MAP kinase activation associated with hepatocyte growth factor-induced cell scatteringCell Signal1999111288589010659996
  • GherardiEBirchmeierWBirchmeierCVande WoudeGTargeting MET in cancer: rationale and progressNat Rev Cancer20121228910322270953
  • ZhangYWWangLMJoveRVande WoudeGFRequirement of Stat3 signaling for HGF/SF-Met mediated tumorigenesisOncogene200221221722611803465
  • TrusolinoLBertottiAComoglioPMMET signalling: principles and functions in development, organ regeneration and cancerNat Rev Mol Cell Biol2010111283484821102609
  • SchmidtCBladtFGoedeckeSScatter factor/hepatocyte growth factor is essential for liver developmentNature199537365166997027854452
  • UeharaYMinowaOMoriCPlacental defect and embryonic lethality in mice lacking hepatocyte growth factor/scatter factorNature199537365167027057854453
  • MainaFHiltonMCPonzettoCDaviesAMKleinRMet receptor signaling is required for sensory nerve development and HGF promotes axonal growth and survival of sensory neuronsGenes Dev19971124334133509407027
  • BladtFRiethmacherDIsenmannSAguzziABirchmeierCEssential role for the c-met receptor in the migration of myogenic precursor cells into the limb budNature199537665437687717651534
  • ChmielowiecJBorowiakMMorkelMc-Met is essential for wound healing in the skinJ Cell Biol2007177115116217403932
  • HuhCGFactorVMSánchezAUchidaKConnerEAThorgeirssonSSHepatocyte growth factor/c-met signaling pathway is required for efficient liver regeneration and repairProc Natl Acad Sci U S A2004101134477448215070743
  • LiuYHepatocyte growth factor in kidney fibrosis: therapeutic potential and mechanisms of actionAm J Physiol Renal Physiol20042871F7F1615180923
  • SchmidtLDuhFMChenFGermline and somatic mutations in the tyrosine kinase domain of the MET proto-oncogene in papillary renal carcinomasNat Genet199716168739140397
  • GraveelCRLondonCAVande WoudeGFA mouse model of activating Met mutationsCell Cycle20054451852015738649
  • NakajimaMSawadaHYamadaYThe prognostic significance of amplification and overexpression of c-met and c-erb B-2 in human gastric carcinomasCancer19998591894190210223227
  • KuniyasuHYasuiWKitadaiYYokozakiHItoHTaharaEFrequent amplification of the c-met gene in scirrhous type stomach cancerBiochem Biophys Res Commun199218912272321333188
  • FischerUMüllerHWSattlerHPFeidenKZangKDMeeseEAmplification of the MET gene in gliomaGenes Chromosomes Cancer199512163657534113
  • SamuelsonELevanKAdamovicTLevanGHorvathGRecurrent gene amplifications in human type I endometrial adenocarcinoma detected by fluorescence in situ hybridizationCancer Genet Cytogenet20081811253018262049
  • Beau-FallerMRuppertAMVoegeliACMET gene copy number in non-small cell lung cancer: molecular analysis in a targeted tyrosine kinase inhibitor naïve cohortJ Thorac Oncol20083433133918379349
  • ZengZSWeiserMRKuntzEc-Met gene amplification is associated with advanced stage colorectal cancer and liver metastasesCancer Lett2008265225826918395971
  • ScagliottiGVNovelloSvon PawelJThe emerging role of MET/HGF inhibitors in oncologyCancer Treat Rev201339779380123453860
  • DulakAMGubishCTStabileLPHenryCSiegfriedJMHGF-independent potentiation of EGFR action by c-MetOncogene201130333625363521423210
  • EngelmanJAZejnullahuKMitsudomiTMET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signalingScience200731658271039104317463250
  • TroianiTMartinelliENapolitanoSIncreased TGF-α as a mechanism of acquired resistance to the anti-EGFR inhibitor cetuximab through EGFR–MET interaction and activation of MET signaling in colon cancer cellsClin Cancer Res201319246751676524122793
  • BardelliACorsoSBertottiAAmplification of the MET receptor drives resistance to anti-EGFR therapies in colorectal cancerCancer Discov20133665867323729478
  • SulpiceEDingSMuscatelli-GrouxBCross-talk between the VEGF-A and HGF signalling pathways in endothelial cellsBiol Cell2009101952553919281453
  • VermeulenLDe SousaEMFvan der HeijdenMWnt activity defines colon cancer stem cells and is regulated by the microenvironmentNat Cell Bioly2010125468476
  • JinHYangRZhengZMetMAb, the one-armed 5D5 anti-c-Met antibody, inhibits orthotopic pancreatic tumor growth and improves survivalCan Res2008681143604368
  • BurgessTLSunJMeyerSBiochemical characterization of AMG 102: a neutralizing, fully human monoclonal antibody to human and nonhuman primate hepatocyte growth factorMol Cancer Ther20109240040920124448
  • MerchantMMaXMaunHRMonovalent antibody design and mechanism of action of onartuzumab, a MET antagonist with anti-tumor activity as a therapeutic agentProc Natl Acad Sci U S A201311032E2987E299623882082
  • ShawATKimDWNakagawaKCrizotinib versus chemotherapy in advanced ALK-positive lung cancerN Engl J Med2013368252385239423724913
  • MosseYPLimMSVossSDSafety and activity of crizotinib for paediatric patients with refractory solid tumours or anaplastic large-cell lymphoma: a Children’s Oncology Group phase 1 consortium studyLancet Oncol201314647248023598171
  • QianFEngstSYamaguchiKInhibition of tumor cell growth, invasion, and metastasis by EXEL-2880 (XL880, GSK1363089), a novel inhibitor of HGF and VEGF receptor tyrosine kinasesCancer Res200969208009801619808973
  • EderJPShapiroGIApplemanLJA phase I study of foretinib, a multi-targeted inhibitor of c-Met and vascular endothelial growth factor receptor 2Clin Cancer Res201016133507351620472683
  • YakesFMChenJTanJCabozantinib (XL184), a novel MET and VEGFR2 inhibitor, simultaneously suppresses metastasis, angiogenesis, and tumor growthMol Cancer Ther201110122298230821926191
  • MunshiNJeaySLiYARQ 197, a novel and selective inhibitor of the human c-Met receptor tyrosine kinase with antitumor activityMol Cancer Ther2010961544155320484018
  • IchimuraEMaeshimaANakajimaTNakamuraTExpression of c-met/HGF receptor in human non-small cell lung carcinomas in vitro and in vivo and its prognostic significanceJpn J Cancer Res19968710106310698957065
  • SmolenGASordellaRMuirBAmplification of MET may identify a subset of cancers with extreme sensitivity to the selective tyrosine kinase inhibitor PHA-665752Proc Natl Acad Sci U S A200610372316232116461907
  • OnozatoRKosakaTKuwanoHSekidoYYatabeYMitsudomiTActivation of MET by gene amplification or by splice mutations deleting the juxtamembrane domain in primary resected lung cancersJ Thorac Oncol20094151119096300
  • CappuzzoFJannePASkokanMMET increased gene copy number and primary resistance to gefitinib therapy in non-small-cell lung cancer patientsAnn Oncol200920229830418836087
  • MaPCKijimaTMaulikGc-MET mutational analysis in small cell lung cancer: novel juxtamembrane domain mutations regulating cytoskeletal functionsCancer Res200363196272628114559814
  • BolandJMJangJSLiJMET and EGFR mutations identified in ALK-rearranged pulmonary adenocarcinoma: molecular analysis of 25 ALK-positive casesJ Thorac Oncol20138557458123449277
  • OkudaKSasakiHYukiueHYanoMFujiiYMet gene copy number predicts the prognosis for completely resected non-small cell lung cancerCancer Sci200899112280228519037978
  • LimEHZhangSLLiJLUsing whole genome amplification (WGA) of low-volume biopsies to assess the prognostic role of EGFR, KRAS, p53, and CMET mutations in advanced-stage non-small cell lung cancer (NSCLC)J Thorac Oncol200941122119096301
  • SiegfriedJMWeissfeldLASingh-KawPWeyantRJTestaJRLandreneauRJAssociation of immunoreactive hepatocyte growth factor with poor survival in resectable non-small cell lung cancerCancer Res19975734334399012470
  • OnitsukaTUramotoHOnoKComprehensive molecular analyses of lung adenocarcinoma with regard to the epidermal growth factor receptor, K-ras, MET, and hepatocyte growth factor statusJ Thorac Oncol20105559159620150826
  • HosodaHIzumiHTukadaYPlasma hepatocyte growth factor elevation may be associated with early metastatic disease in primary lung cancer patientsAnn Thorac Cardiovasc Surg20121811721959198
  • UjiieHTomidaMAkiyamaHSerum hepatocyte growth factor and interleukin-6 are effective prognostic markers for non-small cell lung cancerAnticancer Res20123283251325822843899
  • TurkeABZejnullahuKWuYLPreexistence and clonal selection of MET amplification in EGFR mutant NSCLCCancer Cell2010171778820129249
  • YanoSYamadaTTakeuchiSHepatocyte growth factor expression in EGFR mutant lung cancer with intrinsic and acquired resistance to tyrosine kinase inhibitors in a Japanese cohortJ Thorac Oncol20116122011201722052230
  • ZucaliPARuizMGGiovannettiERole of cMET expression in non-small-cell lung cancer patients treated with EGFR tyrosine kinase inhibitorsAnn Oncol20081991605161218467317
  • SpigelDRErvinTJRamlauRARandomized phase II trial of onartuzumab in combination with erlotinib in patients with advanced non-small-cell lung cancerJ Clin Oncol1110201331324105411424101053
  • GenentechA Study of Onartuzumab (MetMAb) in Combination With Tarceva (Erlotinib) in Patients With Met Diagnostic-Positive Non-Small Cell Lung Cancer Who Have Received Chemotherapy For Advanced or Metastatic Disease (MetLung) Available from: http://clinicaltrials.gov/show/NCT01456325. NLM identifier: NCT01456325Accessed March 5, 2014
  • ElezETaberneroJPrudkinLPharmacodynamic (PD)–pharmacokinetic (PK) study of ficlatuzumab (F), a monoclonal antibody (MAB) directed to the hepatocyte growth factor (HGF), in patients (Pts) with advanced solid tumors who have live metastases (Mets)Ann Oncol201223Suppl 9443PD
  • MeetzeKBoudrowAConnolyKAnti-tumor activity of SCH 900105 (AV299), an anti-HGF antibody, in non-small cell lung cancer modelsMol Cancer Ther20098Suppl 12C173
  • PatnaikAWeissGJPapadopoulosKPhase I study of SCH 900105 (SC), an anti-hepatocyte growth factor (HGF) monoclonal antibody (MAb), as a single agent and in combination with erlotinib (E) in patients (pts) with advanced solid tumorsJ Clin Oncol201028Suppl 152525
  • MokTParkKGeaterSLA randomized phase (Ph) 2 study with exploratory biomarker analysis of ficlatuzumab (F) a humanized hepatocyte growth factor (HGF) inhibitory MAB in combination with gefitinib (G) versus G in Asian patients (pts) with lung adenocarcinoma (LA)Ann Oncol201223Suppl 91198P21948813
  • SequistLVvon PawelJGarmeyEGRandomized phase II study of erlotinib plus tivantinib versus erlotinib plus placebo in previously treated non-small-cell lung cancerJ Clin Oncol201129243307331521768463
  • Kyowa Hakko Kirin Company, LimitedA Phase 3, Randomized, Double-Blinded, Placebo-Controlled Study of ARQ 197 Plus Erlotinib Versus Placebo Plus Erlotinib (ATTENTION) Available from: http://clinicaltrials.gov/ct2/show/NCT01377376. NLM identifier: NCT01377376Accessed March 5, 2014
  • ArQuleErlotinib Plus Tivantinib (ARQ 197) Versus Single Agent Chemotherapy in Locally Advanced or Metastatic Non-Small Cell Lung Cancer Available from: http://clinicaltrials.gov/ct2/show/NCT01395758. NLM identifier: NCT01395758Accessed March 5, 2014
  • LiuCParkMTsaoMSOverexpression of c-met proto-oncogene but not epidermal growth factor receptor or c-erbB-2 in primary human colorectal carcinomasOncogene1992711811851741162
  • FujitaSSuganoKExpression of c-met proto-oncogene in primary colorectal cancer and liver metastasesJpn J Clin Oncol19972763783839437998
  • HiscoxSEHallettMBPuntisMCNakamuraTJiangWGExpression of the HGF/SF receptor, c-met, and its ligand in human colorectal cancersCancer Invest19971565135219412656
  • Cancer Genome Atlas NetworkComprehensive molecular characterization of human colon and rectal cancerNature2012487740733033722810696
  • FazekasKCsukaOKövesIRásóETímárJExperimental and clinicopathologic studies on the function of the HGF receptor in human colon cancer metastasisClin Exp Metastasis200018863964911827067
  • TakeuchiHBilchikASahaSc-MET expression level in primary colon cancer: a predictor of tumor invasion and lymph node metastasesClin Cancer Res2003941480148812684423
  • KammulaUSKuntzEJFranconeTDMolecular co-expression of the c-Met oncogene and hepatocyte growth factor in primary colon cancer predicts tumor stage and clinical outcomeCancer Lett2007248221922816945480
  • LeeCTChowNHSuPFLinSCLinPCLeeJCThe prognostic significance of RON and MET receptor coexpression in patients with colorectal cancerDis Colon Rectum20085181268127418536971
  • LiskaDChenCTBachleitner-HofmannTChristensenJGWeiserMRHGF rescues colorectal cancer cells from EGFR inhibition via MET activationClin Cancer Res201117347248221098338
  • EngCVan CutsemENowaraEA randomized, phase Ib/II trial of rilotumumab (AMG 102; ril) or ganitumab (AMG 479; gan) with panitumumab (pmab) versus pmab alone in patients (pts) with wild-type (WT) KRAS metastatic colorectal cancer (mCRC): Primary and biomarker analysesJ Clin Oncol201129Suppl350021844507
  • GenentechFOLFOX/Bevacizumab With Onartuzumab (MetMAb) Versus Placebo as First-Line Treatment in Patients With Metastatic Colorectal Cancer Available from: http://clinicaltrials.gov/ct2/show/NCT01418222. NLM identifier: NCT01418222Accessed March 5, 2014
  • BendellJCErvinTJGallinsonDTreatment rationale and study design for a randomized, double-blind, placebo-controlled phase II study evaluating onartuzumab (MetMAb) in combination with bevacizumab plus mFOLFOX-6 in patients with previously untreated metastatic colorectal cancerClin Colorectal Cancer201312321822223810377
  • EngCHartLLSevertsevAA randomized, placebo-controlled, phase I/II study of tivantinib (ARQ 197) in combination with cetuximab and irinotecan in patients (pts) with KRAS wild-type (WT) metastatic colorectal cancer (CRC) who had received previous front-line systemic therapyJ Clin Oncol2013Suppl 313508
  • EngCBessudoAGabrailNPhase I/II study of tivantinib (ARQ197), irinotecan, and cetuximab in patients with KRAS wild type, previously treated, metastatic colorectal cancer patientsAnn Oncol201223Suppl 4PD-0018
  • HaSYLeeJKangSYMET overexpression assessed by new interpretation method predicts gene amplification and poor survival in advanced gastric carcinomasMod Pathol201326121632164123807774
  • LeeHEKimMALeeHSMET in gastric carcinomas: comparison between protein expression and gene copy number and impact on clinical outcomeBr J Cancer2012107232533322644302
  • GrazianoFGalluccioNLorenziniPGenetic activation of the MET pathway and prognosis of patients with high-risk, radically resected gastric cancerJ Clin Oncol201129364789479522042954
  • LennerzJKKwakELAckermanAMET amplification identifies a small and aggressive subgroup of esophagogastric adenocarcinoma with evidence of responsiveness to crizotinibJ Clin Oncol201129364803481022042947
  • TsujimotoHSugiharaHHagiwaraAHattoriTAmplification of growth factor receptor genes and DNA ploidy pattern in the progression of gastric cancerVirchows Arch199743163833899428926
  • YuSYuYZhaoNCuiJLiWLiuTc-Met as a prognostic marker in gastric cancer: a systematic review and meta-analysisPloS One2013811e7913724223894
  • DengNGohLKWangHA comprehensive survey of genomic alterations in gastric cancer reveals systematic patterns of molecular exclusivity and co-occurrence among distinct therapeutic targetsGut201261567368422315472
  • YapTAOlmosDBrunettoATPhase I trial of a selective c-MET inhibitor ARQ 197 incorporating proof of mechanism pharmacodynamic studiesJ Clin Oncol201129101271127921383285
  • CatenacciDVHendersonLXiaoSYDurable complete response of metastatic gastric cancer with anti-Met therapy followed by resistance at recurrenceCancer Discov20111757357922389872
  • ShahMAWainbergZACatenacciDVPhase II study evaluating 2 dosing schedules of oral foretinib (GSK1363089), cMET/VEGFR2 inhibitor, in patients with metastatic gastric cancerPloS One201383e5401423516391
  • OlinerKSTangRAndersonAEvaluation of MET pathway biomarkers in a phase II study of rilotumumab (R, AMG 102) or placebo (P) in combination with epirubicin, cisplatin, and capecitabine (ECX) in patients (pts) with locally advanced or metastatic gastric (G) or esophagogastric junction (EGJ) cancerJ Clin Oncol201230Suppl4005
  • ZhuMTangRDoshiSExposure-response (E-R) analysis of rilotumumab (R, AMG 102) plus epirubicin/cisplatin/capecitabine (ECX) in patients (pts) with locally advanced or metastatic gastric or esophagogastric junction (G/EGJ) cancerJ Clin Oncol201230Suppl2535
  • Hoffmann-La RocheA Study of Onartuzumab (MetMAb) in Combination With mFOLFOX6 in Patients With Metastatic HER2-Negative And Met-Positive Gastroesophageal Cancer (MetGastric) Available from: http://clinicaltrials.gov/ct2/show/NCT01662869. NLM identifier: NCT01662869Accessed March 5, 2014
  • AmgenFirst-Line Treatment for Locally Advanced or Metastatic Mesenchymal Epithelial Transition Factor (MET) - Positive Gastric, Lower Esophageal, or Gastroesophageal Junction (GEJ) Adenocarcinoma (RILOMET-1) Available from: http://clinicaltrials.gov/ct2/show/NCT01697072. NLM identifier: NCT01697072Accessed March 5, 2014
  • KissAWangNJXieJPThorgeirssonSSAnalysis of transforming growth factor (TGF)-alpha/epidermal growth factor receptor, hepatocyte growth factor/c-met, TGF-beta receptor type II, and p53 expression in human hepatocellular carcinomasClin Cancer Res199737105910669815784
  • LjubimovaJYPetrovicLMWilsonSEGellerSADemetriouAAExpression of HGF, its receptor c-met, c-myc, and albumin in cirrhotic and neoplastic human liver tissueJ Histochem Cytochem199745179879010472
  • OkanoJShiotaGKawasakiHExpression of hepatocyte growth factor (HGF) and HGF receptor (c-met) proteins in liver diseases: an immunohistochemical studyLiver199919215115910220746
  • KondoSOjimaHTsudaHClinical impact of c-Met expression and its gene amplification in hepatocellular carcinomaInt J Clin Oncol201318220721322218908
  • LeeSJLeeJSohnIA Survey of c-MET expression and amplification in 287 patients with hepatocellular carcinomaAnticancer Res201333115179518624222167
  • OsadaSKanematsuMImaiHGoshimaSClinical significance of serum HGF and c-Met expression in tumor tissue for evaluation of properties and treatment of hepatocellular carcinomaHepatogastroenterology20085582–8354454918613405
  • KeAWShiGMZhouJRole of overexpression of CD151 and/or c-Met in predicting prognosis of hepatocellular carcinomaHepatology200949249150319065669
  • WangZLLiangPDongBWYuXLYu deJPrognostic factors and recurrence of small hepatocellular carcinoma after hepatic resection or microwave ablation: a retrospective studyJ Gastrointest Surg200812232733717943391
  • SantoroARimassaLBorbathITivantinib for second-line treatment of advanced hepatocellular carcinoma: a randomised, placebo-controlled phase 2 studyLancet Oncol2013141556323182627
  • AngCSSunMYHuitzil-MelendezDFc-MET and HGF mRNA expression in hepatocellular carcinoma: correlation with clinicopathological features and survivalAnticancer Res20133383241324523898085
  • HoriguchiNTakayamaHToyodaMHepatocyte growth factor promotes hepatocarcinogenesis through c-Met autocrine activation and enhanced angiogenesis in transgenic mice treated with diethylnitrosamineOncogene200221121791179911896611
  • ZhangSZPanFYXuJFKnockdown of c-Met by adenovirus-delivered small interfering RNA inhibits hepatocellular carcinoma growth in vitro and in vivoMol Cancer Ther20054101577158416227408
  • YouHDingWDangHJiangYRountreeCBc-Met represents a potential therapeutic target for personalized treatment in hepatocellular carcinomaHepatology201154387988921618573
  • Daiichi Sankyo IncStudy of Tivantinib in Subjects With Inoperable Hepatocellular Carcinoma Who Have Been Treated With One Prior Therapy (METIV-HCC) Available from: http://clinicaltrials.gov/ct2/show/NCT01755767. NLM identifier: NCT01755767Accessed March 5, 2014
  • VerslypeCCohnALKelleyRKActivity of cabozantinib (XL184) in hepatocellular carcinoma: results from a phase II randomized discontinuation trial (RDT)J Clin Oncol201230Suppl4007
  • YauTCSukeepaisarnjaroenWChaoYA phase I/II study of foretinib, an oral multikinase inhibitor targeting MET, RON, AXL, TIE-2, and VEGFR in advanced hepatocellular carcinoma (HCC)J Clin Oncol201230Suppl4108
  • O’NeilBHBendellJCModianoMRPhase I/II study of E7050 (golvantinib) in combination with sorafenib in patients (pts) with advanced hepatocellular carcinoma (HCC): Phase I resultsJ Clin Oncol201230Suppl 34294
  • ExelixisStudy of Cabozantinib (XL184) vs Placebo in Subjects With Hepatocellular Carcinoma Who Have Received Prior Sorafenib (CELESTIAL) Available from: http://clinicaltrials.gov/ct2/show/NCT01908426. NLM identifier: NCT01908426Accessed March 5, 2014
  • Hoffmann-La RocheA Study of Onartuzumab as Single Agent and in Combination With Sorafenib in Patients With Advanced Hepatocellular Carcinoma Available from: http://clinicaltrials.gov/ct2/show/NCT01897038. NLM identifier: NCT01897038Accessed March 5, 2014
  • KnudsenBSGmyrekGAInraJHigh expression of the Met receptor in prostate cancer metastasis to boneUrology20026061113111712475693
  • PistersLLTroncosoPZhauHELiWvon EschenbachACChungLWC-met proto-oncogene expression in benign and malignant human prostate tissuesJ Urol199515412932987539865
  • TuWHZhuCClarkCChristensenJGSunZEfficacy of c-Met inhibitor for advanced prostate cancerBMC Cancer20101055620946682
  • VerrasMLeeJXueHLiTHWangYSunZThe androgen receptor negatively regulates the expression of c-Met: implications for a novel mechanism of prostate cancer progressionCancer Res200767396797517283128
  • SmithDCSmithMRSweeneyCCabozantinib in patients with advanced prostate cancer: results of a phase II randomized discontinuation trialJ Clin Oncol201331441241923169517
  • LeeRJSaylorPJMichaelsonMDA dose-ranging study of cabozantinib in men with castration-resistant prostate cancer and bone metastasesClin Cancer Res201319113088309423553848
  • DaiJZhangHKaratsinidesACabozantinib inhibits prostate cancer growth and prevents tumor-induced bone lesionsClin Cancer Res201420361763024097861
  • ExelixisStudy of Cabozantinib (XL184) Versus Prednisone in Men With Metastatic Castration-resistant Prostate Cancer Previously Treated With Docetaxel and Abiraterone or MDV3100 (COMET-1) Available from: http://clinicaltrials.gov/ct2/show/NCT01605227. NLM identifier: NCT01605227Accessed March 5, 2014
  • ExelixisStudy of Cabozantinib (XL184) Versus Mitoxantrone Plus Prednisone in Men With Previously Treated Symptomatic Castration-resistant Prostate Cancer (COMET-2) Available from: http://clinicaltrials.gov/ct2/show/NCT01522443. NLM identifier: NCT01522443Accessed March 5, 2014
  • ExelixisStudy of Cabozantinib in Combination With Abiraterone in Chemotherapy-Naïve Subjects With Bone-Metastatic Castration-Resistant Prostate Cancer Available from: http://clinicaltrials.gov/ct2/show/NCT01995058. NLM identifier: NCT01995058Accessed March 5, 2014
  • RyanCJRosenthalMNgSTargeted MET inhibition in castration-resistant prostate cancer: a randomized phase II study and biomarker analysis with rilotumumab plus mitoxantrone and prednisoneClin Cancer Res201319121522423136195
  • NakaigawaNYaoMBabaMInactivation of von Hippel-Lindau gene induces constitutive phosphorylation of MET protein in clear cell renal carcinomaCancer Res20066673699370516585196
  • GibneyGTAzizSACampRLc-Met is a prognostic marker and potential therapeutic target in clear cell renal cell carcinomaAnn Oncol201324234334923022995
  • SchoffskiPGarciaJAStadlerWMA phase II study of the efficacy and safety of AMG 102 in patients with metastatic renal cell carcinomaBJU Int2011108567968621156020
  • ChoueiriTKPalSKMcDermottDFActivity of cabozantinib (XL184) in patients (pts) with metastatic, refractory renal cell carcinoma (RCC)J Clin Oncol201230Suppl 5364
  • ExelixisA Study of Cabozantinib (XL184) vs Everolimus in Subjects With Metastatic Renal Cell Carcinoma (METEOR) Available from: http://clinicaltrials.gov/ct2/show/NCT01865747. NLM identifier: NCT01865747Accessed March 5, 2014
  • National Cancer Institute (NCI)Cabozantinib-s-malate or Sunitinib Malate in Treating Patients With Previously Untreated Locally Advanced or Metastatic Kidney Cancer Available from: http://clinicaltrials.gov/ct2/show/NCT01835158. NLM identifier: NCT01835158Accessed March 5, 2014
  • ChoueiriTKVaishampayanURosenbergJEPhase II and biomarker study of the dual MET/VEGFR2 inhibitor foretinib in patients with papillary renal cell carcinomaJ Clin Oncol201331218118623213094
  • GordonMSSweeneyCJMendelsonDSSafety, 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 tumorsClin Cancer Res201016269971020068101
  • SalgiaRPatelPBothosJPhase I dose-escalation study of onartuzumab as a single agent and in combination with bevacizumab in patients with advanced solid malignanciesClin Cancer Res20142061666167524493831
  • BoccaccioCSabatinoGMedicoEThe MET oncogene drives a genetic programme linking cancer to haemostasisNature2005434703139640015772665
  • BasilicoCPennacchiettiSVignaETivantinib (ARQ197) displays cytotoxic activity that is independent of its ability to bind METClin Cancer Res20131992381239223532890
  • Bachleitner-HofmannTSunMYChenCTHER kinase activation confers resistance to MET tyrosine kinase inhibition in MET oncogene-addicted gastric cancer cellsMol Cancer Ther20087113499350818974395
  • QiJMcTigueMARogersAMultiple mutations and bypass mechanisms can contribute to development of acquired resistance to MET inhibitorsCancer Res20117131081109121266357
  • McDermottUPusapatiRVChristensenJGGrayNSSettlemanJAcquired resistance of non-small cell lung cancer cells to MET kinase inhibition is mediated by a switch to epidermal growth factor receptor dependencyCancer Res20107041625163420124471
  • CeperoVSierraJRCorsoSMET and KRAS gene amplification mediates acquired resistance to MET tyrosine kinase inhibitorsCancer Res201070197580759020841479
  • LeeNVLiraMEPavlicekAA novel SND1-BRAF fusion confers resistance to c-Met inhibitor PF-04217903 in GTL16 cells through [corrected] MAPK activationPloS One201276e3965322745804
  • FongJTJacobsRJMoravecDNAlternative signaling pathways as potential therapeutic targets for overcoming EGFR and c-Met inhibitor resistance in non-small cell lung cancerPloS One2013811e7839824223799
  • PanB-SChanGKYChenardMMK-2461, a novel multitargeted kinase inhibitor, preferentially inhibits the activated c-Met receptorCancer Res20107041524153320145145
  • BellonSFKaplan-LefkoPYangYc-Met inhibitors with novel binding mode show activity against several hereditary papillary renal cell carcinoma-related mutationsJ Biol Chem200828352675268318055465