Elsevier

Pharmacological Research

Volume 102, December 2015, Pages 90-106
Pharmacological Research

Review
MET/HGF targeted drugs as potential therapeutic strategies in non-small cell lung cancer

https://doi.org/10.1016/j.phrs.2015.09.016Get rights and content

Introduction

Lung cancer is the tumor with the second highest incidence (≈14%) in both genders after prostate cancer in men and breast cancer in women [1]. According to the latest available statistics, more than 221,200 new cases are estimated for 2015 in the USA alone [1]. Lung cancer is also the first cause of death from cancer worldwide, with the prediction of 158,040 deaths in 2015 [1].

Non-small cell lung cancer (NSCLC) represents around 80% of lung cancer cases and is classified in three subtypes: adenocarcinoma, squamous cell (epidermoid) carcinoma, and large-cell carcinoma. Approximately 70% of NSCLC patients are diagnosed in an advanced stage (IIIB-IV), whose standard treatment, based on platinum chemotherapy [2], [3], [4], shows poor response rates (RR) of 17–32%, progression-free survival (PFS) of 3.1–3.5 months, and overall survival (OS) of 7.4–11.3 months [5], [6], [7], [8].

The biology and treatment of NSCLC has been revolutionized over the past few years due to the identification of druggable genetic alterations, such as epidermal growth factor receptor (EGFR) mutations or echinoderm microtubule-associated protein-like 4-anaplastic lymphoma kinase (EML4-ALK) translocations. EGFR is mutated in up to 59.7% of NSCLC tumors from Asian patients and in around 16.7% of those from Caucasian patients. EGFR mutations are more frequent in women and non-smokers [9], [10]. Most (90%) of the activating mutations correspond to leucine-to-arginine substitutions at position 858 (L858R) or deletions in exon 19, which affect the conserved sequence ELREA (delE746-A750). There have also been reports of a much less frequent mutation in exon 18, encompassing a substitution of glycine for alanine, cysteine or serine at codon 719 (G719X) [10], [11], [12], [13], [14], [15]. Novel therapeutic tyrosine kinase inhibitors (TKIs) targeted to these mutations, such as erlotinib (Tarceva®, Roche), gefitinib (Iressa®, AstraZeneca), or afatinib (Giotrif®, Boehringer Ingelheim), have shown RR of up to 75% [9], [16]. However, all patients with activating mutations in EGFR ultimately develop resistance to TKIs after a median of 12 months [10], [17], [18]. The most important resistance mechanism is a secondary point mutation in exon 20 of EGFR (T790 M), which replaces methionine with threonine at amino acid position 790. Other mechanisms reported in the literature include: MET protooncogen (MET) amplification and protein overexpression of its ligand, the hepatocyte growth factor (HGF); phosphatase and tensin homolog (PTEN) loss; kirsten rat sarcoma viral oncogene homolog (KRAS) mutations; v-raf murine sarcoma viral oncogene homolog B (BRAF) mutations; human EGF receptor 2 (HER-2) mutations, epithelial-to-mesenchymal transition (EMT); AXL receptor tyrosine kinase (AXL) overexpression; and small-cell transformation [19], [20], [21], [22], [23], [24], [25], [26], [27], [28], [29], [30], [31], [32], [33], [34].

Druggable alterations in NSCLC also include EML4-ALK translocations [35], [36], which have been reported in 3–7% of NSCLC patients, most frequently in younger patients with a history of no or light smoking and adenocarcinoma histology [37], [38]. Patients with this genetic alteration can benefit from crizotinib (Xalkori®, Pfizer), a potent TKI that is also active against MET and ROS1 (c-ROS oncogene) tyrosine kinases, which has shown overall response rates (ORR) of approximately 60% [39], [40] and PFS of 8–10 months [41]. As in the case of EGFR inhibitors, multiple resistance mechanisms have also been described, such as mutations in the ALK kinase domain (C1156Y, L1196M, G1296A) [42], [43], [44], [45], [46], [47], [48], [49].

The MET protooncogene encodes the c-Met protein, a tyrosine kinase receptor that binds to hepatocyte growth factor (HGF). Together with its ligand, c-Met plays a key role in several processes, such as proliferation, survival, motility, invasiveness, and stimulation of angiogenesis cell survival and proliferation through the PI3K/AKT, RAS-RAF-ERK1/2, and STAT3 pathways [50], [51], [52]. Alterations in the MET pathway, such as MET gene amplification and activating mutations or HGF overexpression, have been associated with a poor clinical outcome and EGFR-TKIs resistance in NSCLC patients [21], [32], [52], [53], [54], [55], [56] and MET inhibition is a promising strategy to overcome resistance to therapy in tumors with deregulated MET signaling [57].

Several MET inhibitors have been developed to block the HGF/MET signaling pathway (Fig. 1), including selective and non-selective MET TKIs, and monoclonal antibodies designed to prevent MET-HGF binding. Some of these inhibitors have been tested in clinical trials as single agents or in association with other targeted drugs, chemotherapy, or radiotherapy. In this review, we focus on these different strategies to prevent MET pathway activation.

Section snippets

MET pathway signaling and dysregulation

The MET tyrosine kinase (TK) receptor is a heterodimer that comprises an extracellular α-chain and transmembrane β-chain linked by a disulphide bond [58], [59]. The extracellular portion is constituted by a semaphorin (SEMA) domain, which is essential for ligand binding and receptor dimerization. A TK domain with four key tyrosine residues involved in MET pathway activation after HGF binding composes the intracellular portion, which is connected with the extracellular portion by juxtamembrane

MET: targeted drugs

Various clinical trials have tested molecules targeting HGF/MET signaling in treatment-naïve or TKI-resistant NSCLC patients [40], [97], [98], [105], [106], [107], [108], [109]. The results of some of those trials are already available and are summarized in Table 1, Table 2. Ongoing studies are reported in Table 3.

Conclusions

The stratification of NSCLC into different molecular subsets has revolutionized the treatment of this malignancy. It has led to the widespread clinical use of TKIs such as gefitinib and erlotinib (FDA-approved for patients with activating EGFR mutations) and crizotinib (FDA-approved for EML4-ALK translocated tumors). MET/HGF signaling also represents an important target in NSCLC. Through its activation of the PI3K/AKT, RAS-RAF-ERK1/2 and STAT3 pathways, MET plays a key role in several processes

Conflict of interest

The authors declare that there is not conflict of interest that could be perceived as prejudicing the impartiality of the research reported and there is not any competing financial interest in relation to the work described in this article.

Funding

This work was partly supported by a contract for Marisa Cañadas-Garre (Técnicos de Apoyo Subprogram. CA12/00097) from Instituto de Salud Carlos III, Ministerio de Economía y Competitividad and a research grant for Cristina Pérez-Ramírez (FPU12/04722), from Ministerio de Educación, Cultura y Deporte.

Acknowledgment

The results of this investigation are part of the doctoral thesis presented by Cristina Pérez-Ramírez at the University of Granada.

First page preview

First page preview
Click to open first page preview

References (202)

  • S. Kim et al.

    Heterogeneity of genetic changes associated with acquired crizotinib resistance in alk-rearranged lung cancer

    J. Thorac. Oncol.

    (2013)
  • Y. Liu

    The human hepatocyte growth factor receptor gene: complete structural organization and promoter characterization

    Gene

    (1998)
  • N.A. Cipriani et al.

    Met as a target for treatment of chest tumors

    Lung Cancer

    (2009)
  • C.M. Stellrecht et al.

    Met receptor tyrosine kinase as a therapeutic anticancer target

    Cancer Lett.

    (2009)
  • L.S. Lock et al.

    Grb2-independent recruitment of gab1 requires the c-terminal lobe and structural integrity of the met receptor kinase domain

    J. Biol. Chem.

    (2003)
  • L. Lamorte et al.

    Crk synergizes with epidermal growth factor for epithelial invasion and morphogenesis and is required for the met morphogenic program

    J. Biol. Chem.

    (2002)
  • C. Ponzetto et al.

    A multifunctional docking site mediates signaling and transformation by the hepatocyte growth factor/scatter factor receptor family

    Cell

    (1994)
  • H.C. Chen et al.

    Tyrosine phosphorylation of focal adhesion kinase stimulated by hepatocyte growth factor leads to mitogen-activated protein kinase activation

    J. Biol. Chem.

    (1998)
  • J.F. Lai et al.

    Involvement of focal adhesion kinase in hepatocyte growth factor-induced scatter of madin-darby canine kidney cells

    J. Biol. Chem.

    (2000)
  • D. Sakkab et al.

    Signaling of hepatocyte growth factor/scatter factor (hgf) to the small gtpase rap1 via the large docking protein gab1 and the adapter protein crkl

    J. Biol. Chem.

    (2000)
  • F. Cappuzzo et al.

    Met increased gene copy number and primary resistance to gefitinib therapy in non-small-cell lung cancer patients

    Ann. Oncol.

    (2009)
  • W. Jiang et al.

    Hepatocyte growth factor/scatter factor, its molecular, cellular and clinical implications in cancer

    Crit. Rev. Oncol. Hematol.

    (1999)
  • R.L. Siegel et al.

    Cancer statistics

    CA: Cancer J. Clin.

    (2015)
  • S. Edge et al.

    Ajcc Cancer Staging Manual

    (2010)
  • R.S.H.J. Herbst et al.

    Lung cancer

    N. Engl. J. Med.

    (2008)
  • F. Fossella et al.

    Randomized, multinational, phase III study of docetaxel plus platinum combinations versus vinorelbine plus cisplatin for advanced non-small-cell lung cancer: the tax 326 study group

    J. Clin. Oncol.

    (2003)
  • K. Kelly et al.

    Randomized phase III trial of paclitaxel plus carboplatin versus vinorelbine plus cisplatin in the treatment of patients with advanced non-small-cell lung cancer: a southwest oncology group trial

    J. Clin. Oncol.

    (2001)
  • G.V. Scagliotti et al.

    Phase iii randomized trial comparing three platinum-based doublets in advanced non-small-cell lung cancer

    J. Clin. Oncol.

    (2002)
  • J.H. Schiller et al.

    Comparison of four chemotherapy regimens for advanced non-small-cell lung cancer

    N. Engl. J. Med.

    (2002)
  • T.S.W.Y. Mok et al.

    Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma

    N. Engl. J. Med.

    (2009)
  • R. Rosell et al.

    Screening for epidermal growth factor receptor mutations in lung cancer

    N. Engl. J. Med.

    (2009)
  • T. Kosaka et al.

    Mutations of the epidermal growth factor receptor gene in lung cancer: biological and clinical implications

    Cancer Res.

    (2004)
  • T.J. Lynch et al.

    Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib

    New Engl. J. Med.

    (2004)
  • J.G. Paez et al.

    Egfr mutations in lung cancer: Correlation with clinical response to gefitinib therapy

    Science (New York, NY)

    (2004)
  • W. Pao et al.

    Egf receptor gene mutations are common in lung cancers from never smokers and are associated with sensitivity of tumors to gefitinib and erlotinib

    Proc. Natl. Acad. Sci. U. S. A.

    (2004)
  • H. Shigematsu et al.

    Clinical and biological features associated with epidermal growth factor receptor gene mutations in lung cancers

    J. Natl. Cancer Inst.

    (2005)
  • M.E. Arcila et al.

    Prevalence, clinicopathologic associations, and molecular spectrum of erbb2 (her2) tyrosine kinase mutations in lung adenocarcinomas

    Clin. Cancer Res.

    (2012)
  • L.A. Byers et al.

    An epithelial-mesenchymal transition gene signature predicts resistance to egfr and pi3k inhibitors and identifies axl as a therapeutic target for overcoming egfr inhibitor resistance

    Clin. Cancer Res.

    (2013)
  • J.A. Engelman et al.

    Met amplification leads to gefitinib resistance in lung cancer by activating erbb3 signaling

    Science (New York, NY)

    (2007)
  • S.B.T. Kobayashi et al.

    Egfr mutation and resistance of non-small-cell lung cancer to gefitinib

    New Engl. J. Med.

    (2005)
  • W.T. Lim et al.

    Pten and phosphorylated akt expression and prognosis in early- and late-stage non-small cell lung cancer

    Oncol. Rep.

    (2007)
  • E. Massarelli et al.

    Kras mutation is an important predictor of resistance to therapy with epidermal growth factor receptor tyrosine kinase inhibitors in non-small-cell lung cancer

    Clin. Cancer Res.

    (2007)
  • K. Ohashi et al.

    Lung cancers with acquired resistance to egfr inhibitors occasionally harbor braf gene mutations but lack mutations in kras, nras, or mek1

    Proc. Natl. Acad. Sci. U. S. A.

    (2012)
  • W. Pao et al.

    Acquired resistance of lung adenocarcinomas to gefitinib or erlotinib is associated with a second mutation in the egfr kinase domain

    PLoS Med.

    (2005)
  • W.W.T. Pao et al.

    Kras mutations and primary resistance of lung adenocarcinomas to gefitinib or erlotinib

    PLoS Med.

    (2005)
  • S. Thomson et al.

    Epithelial to mesenchymal transition is a determinant of sensitivity of non-small-cell lung carcinoma cell lines and xenografts to epidermal growth factor receptor inhibition

    Cancer Res.

    (2005)
  • F. Wu et al.

    The role of axl in drug resistance and epithelial-to-mesenchymal transition of non-small cell lung carcinoma

    Int. J. Clin. Exp. Pathol.

    (2014)
  • S. Yano et al.

    Hepatocyte growth factor induces gefitinib resistance of lung adenocarcinoma with epidermal growth factor receptor-activating mutations

    Cancer Res.

    (2008)
  • Z. Zhang et al.

    Activation of the axl kinase causes resistance to egfr-targeted therapy in lung cancer

    Nat. Genet.

    (2012)
  • C. Perez-Ramirez et al.

    A new promising biomarker in non-small-cell lung carcinoma

    Pharmacogenomics

    (2015)
  • View full text