An Observatory for the MET Oncogene: A Guide for Targeted Therapies
Abstract
:Simple Summary
Abstract
1. Introduction
2. Structure and Function of the MET Kinase
3. MET Alterations in Tumors and Their Biological Significance
4. MET Amplification
5. Exon14 ‘Skipping’, the Predominant MET Alteration
6. Point Mutations within the MET Coding Sequence
7. Fusion Partners of MET Drive Oncogene ‘Addiction’
8. Genetic Alterations of MET: The Peak of the Iceberg?
9. MET-Targeted Therapies
10. Different MET-Blocking Agents: Advantages and Pitfalls
11. Patient Stratification: A Key for Success in Targeted Therapies
12. Understanding and Overcoming Drug Resistance
13. The Flare Effect: mTOR Pathway Comes to Scene
14. Conclusions
Author Contributions
Funding
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Weidner, K.M.; Hartmann, G.; Naldini, L.; Comoglio, P.M.; Sachs, M.; Fonatsch, C.; Rieder, H.; Birchmeier, W. Molecular characteristics of HGF-SF and its role in cell motility and invasion. Exs 1993, 65, 311–328. [Google Scholar]
- Graveel, C.R.; Tolbert, D.; Vande Woude, G.F. MET: A critical player in tumorigenesis and therapeutic target. Cold Spring Harb. Perspect. Biol. 2013, 5, a009209. [Google Scholar] [CrossRef]
- Comoglio, P.M.; Trusolino, L.; Boccaccio, C. Known and novel roles of the MET oncogene in cancer: A coherent approach to targeted therapy. Nat. Rev. Cancer 2018, 18, 341–358. [Google Scholar] [CrossRef]
- Giordano, S.; Di Renzo, M.F.; Narsimhan, R.P.; Cooper, C.S.; Rosa, C.; Comoglio, P.M. Biosynthesis of the protein encoded by the c-met proto-oncogene. Oncogene 1989, 4, 1383–1388. [Google Scholar] [PubMed]
- Uchikawa, E.; Chen, Z.; Xiao, G.Y.; Zhang, X.; Bai, X.C. Structural basis of the activation of c-MET receptor. Nat. Commun. 2021, 12, 4074. [Google Scholar] [CrossRef] [PubMed]
- Nakamura, T. Structure and function of hepatocyte growth factor. Prog. Growth Factor. Res. 1991, 3, 67–85. [Google Scholar] [CrossRef] [PubMed]
- Tajima, H.; Matsumoto, K.; Nakamura, T. Regulation of cell growth and motility by hepatocyte growth factor and receptor expression in various cell species. Exp. Cell Res. 1992, 202, 423–431. [Google Scholar] [CrossRef]
- Ponzetto, C.; Bardelli, A.; Maina, F.; Longati, P.; Panayotou, G.; Dhand, R.; Waterfield, M.D.; Comoglio, P.M. A novel recognition motif for phosphatidylinositol 3-kinase binding mediates its association with the hepatocyte growth factor/scatter factor receptor. Mol. Cell Biol. 1993, 13, 4600–4608. [Google Scholar]
- Boccaccio, C.; Ando, M.; Tamagnone, L.; Bardelli, A.; Michieli, P.; Battistini, C.; Comoglio, P.M. Induction of epithelial tubules by growth factor HGF depends on the STAT pathway. Nature 1998, 391, 285–288. [Google Scholar] [CrossRef]
- Ponzetto, C.; Bardelli, A.; Zhen, Z.; Maina, F.; dalla Zonca, P.; Giordano, S.; Graziani, A.; Panayotou, G.; Comoglio, P.M. A multifunctional docking site mediates signaling and transformation by the hepatocyte growth factor/scatter factor receptor family. Cell 1994, 77, 261–271. [Google Scholar] [CrossRef]
- Trusolino, L.; Bertotti, A.; Comoglio, P.M. MET signalling: Principles and functions in development, organ regeneration and cancer. Nat. Rev. Mol. Cell Biol. 2010, 11, 834–848. [Google Scholar] [CrossRef]
- Gherardi, E.; Birchmeier, W.; Birchmeier, C.; Vande Woude, G. Targeting MET in cancer: Rationale and progress. Nat. Rev. Cancer 2012, 12, 89–103. [Google Scholar] [CrossRef] [PubMed]
- Boccaccio, C.; Comoglio, P.M. Invasive growth: A MET-driven genetic programme for cancer and stem cells. Nat. Rev. Cancer 2006, 6, 637–645. [Google Scholar] [CrossRef] [PubMed]
- Trusolino, L.; Comoglio, P.M. Scatter-factor and semaphorin receptors: Cell signalling for invasive growth. Nat. Rev. Cancer 2002, 2, 289–300. [Google Scholar] [CrossRef] [PubMed]
- Christofori, G. New signals from the invasive front. Nature 2006, 441, 444–450. [Google Scholar] [CrossRef]
- Frampton, G.M.; Ali, S.M.; Rosenzweig, M.; Chmielecki, J.; Lu, X.; Bauer, T.M.; Akimov, M.; Bufill, J.A.; Lee, C.; Jentz, D.; et al. Activation of MET via diverse exon 14 splicing alterations occurs in multiple tumor types and confers clinical sensitivity to MET inhibitors. Cancer Discov. 2015, 5, 850–859. [Google Scholar] [CrossRef]
- Cerqua, M.; Botti, O.; Arigoni, M.; Gioelli, N.; Serini, G.; Calogero, R.; Boccaccio, C.; Comoglio, P.M.; Altintas, D.M. MET14 promotes a ligand-dependent, AKT-driven invasive growth. Life Sci. Alliance 2022, 5, e202201409. [Google Scholar] [CrossRef]
- Comoglio, P.M.; Giordano, S.; Trusolino, L. Drug development of MET inhibitors: Targeting oncogene addiction and expedience. Nat. Rev. Drug Discov. 2008, 7, 504–516. [Google Scholar] [CrossRef]
- Corso, S.; Migliore, C.; Ghiso, E.; De Rosa, G.; Comoglio, P.M.; Giordano, S. Silencing the MET oncogene leads to regression of experimental tumors and metastases. Oncogene 2008, 27, 684–693. [Google Scholar] [CrossRef]
- Behan, F.M.; Iorio, F.; Picco, G.; Goncalves, E.; Beaver, C.M.; Migliardi, G.; Santos, R.; Rao, Y.; Sassi, F.; Pinnelli, M.; et al. Prioritization of cancer therapeutic targets using CRISPR-Cas9 screens. Nature 2019, 568, 511–516. [Google Scholar] [CrossRef]
- Drilon, A.; Clark, J.W.; Weiss, J.; Ou, S.I.; Camidge, D.R.; Solomon, B.J.; Otterson, G.A.; Villaruz, L.C.; Riely, G.J.; Heist, R.S.; et al. Antitumor activity of crizotinib in lung cancers harboring a MET exon 14 alteration. Nat. Med. 2020, 26, 47–51. [Google Scholar] [CrossRef]
- Wolf, J.; Seto, T.; Han, J.-Y.; Reguart, N.; Garon, E.B.; Groen, H.J.M.; Tan, D.S.-W.; Hida, T.; Jonge, M.J.D.; Orlov, S.V.; et al. Capmatinib (INC280) in METΔex14-mutated advanced non-small cell lung cancer (NSCLC): Efficacy data from the phase II GEOMETRY mono-1 study. J. Clin. Oncol. 2019, 37, 9004. [Google Scholar] [CrossRef]
- Landi, L.; Chiari, R.; Tiseo, M.; D’Inca, F.; Dazzi, C.; Chella, A.; Delmonte, A.; Bonanno, L.; Giannarelli, D.; Cortinovis, D.L.; et al. Crizotinib in MET-Deregulated or ROS1-Rearranged Pretreated Non-Small Cell Lung Cancer (METROS): A Phase II, Prospective, Multicenter, Two-Arms Trial. Clin. Cancer Res. 2019, 25, 7312–7319. [Google Scholar] [CrossRef]
- Cruickshanks, N.; Zhang, Y.; Hine, S.; Gibert, M.; Yuan, F.; Oxford, M.; Grello, C.; Pahuski, M.; Dube, C.; Guessous, F.; et al. Discovery and Therapeutic Exploitation of Mechanisms of Resistance to MET Inhibitors in Glioblastoma. Clin. Cancer Res. 2019, 25, 663–673. [Google Scholar] [CrossRef]
- Kim, S.; Kim, T.M.; Kim, D.W.; Kim, S.; Kim, M.; Ahn, Y.O.; Keam, B.; Heo, D.S. Acquired Resistance of MET-Amplified Non-small Cell Lung Cancer Cells to the MET Inhibitor Capmatinib. Cancer Res. Treat. 2019, 51, 951–962. [Google Scholar] [CrossRef]
- Suzawa, K.; Offin, M.; Lu, D.; Kurzatkowski, C.; Vojnic, M.; Smith, R.S.; Sabari, J.K.; Tai, H.; Mattar, M.; Khodos, I.; et al. Activation of KRAS Mediates Resistance to Targeted Therapy in MET Exon 14-mutant Non-small Cell Lung Cancer. Clin. Cancer Res. 2019, 25, 1248–1260. [Google Scholar] [CrossRef]
- Virzi, A.R.; Gentile, A.; Benvenuti, S.; Comoglio, P.M. Reviving oncogenic addiction to MET bypassed by BRAF (G469A) mutation. Proc. Natl. Acad. Sci. USA 2018, 115, 10058–10063. [Google Scholar] [CrossRef] [PubMed]
- Komada, M.; Hatsuzawa, K.; Shibamoto, S.; Ito, F.; Nakayama, K.; Kitamura, N. Proteolytic processing of the hepatocyte growth factor/scatter factor receptor by furin. FEBS Lett. 1993, 328, 25–29. [Google Scholar] [CrossRef] [PubMed]
- Gherardi, E.; Youles, M.E.; Miguel, R.N.; Blundell, T.L.; Iamele, L.; Gough, J.; Bandyopadhyay, A.; Hartmann, G.; Butler, P.J. Functional map and domain structure of MET, the product of the c-met protooncogene and receptor for hepatocyte growth factor/scatter factor. Proc. Natl. Acad. Sci. USA 2003, 100, 12039–12044. [Google Scholar] [CrossRef] [PubMed]
- Zhen, Z.; Giordano, S.; Longati, P.; Medico, E.; Campiglio, M.; Comoglio, P. Structural and functional domains critical for constitutive activation of the HGF-receptor (Met). Oncogene 1994, 9, 1691–1697. [Google Scholar]
- Altintas, D.M.; Gallo, S.; Basilico, C.; Cerqua, M.; Bocedi, A.; Vitacolonna, A.; Botti, O.; Casanova, E.; Rancati, I.; Milanese, C.; et al. The PSI Domain of the MET Oncogene Encodes a Functional Disulfide Isomerase Essential for the Maturation of the Receptor Precursor. Int. J. Mol. Sci. 2022, 23, 12427. [Google Scholar] [CrossRef] [PubMed]
- Longati, P.; Bardelli, A.; Ponzetto, C.; Naldini, L.; Comoglio, P.M. Tyrosines1234-1235 are critical for activation of the tyrosine kinase encoded by the MET proto-oncogene (HGF receptor). Oncogene 1994, 9, 49–57. [Google Scholar] [PubMed]
- Giordano, S.; Bardelli, A.; Zhen, Z.; Menard, S.; Ponzetto, C.; Comoglio, P.M. A point mutation in the MET oncogene abrogates metastasis without affecting transformation. Proc. Natl. Acad. Sci. USA 1997, 94, 13868–13872. [Google Scholar] [CrossRef] [PubMed]
- Ponzetto, C.; Zhen, Z.; Audero, E.; Maina, F.; Bardelli, A.; Basile, M.L.; Giordano, S.; Narsimhan, R.; Comoglio, P. Specific uncoupling of GRB2 from the Met receptor. Differential effects on transformation and motility. J. Biol. Chem. 1996, 271, 14119–14123. [Google Scholar] [CrossRef] [PubMed]
- Yarden, Y.; Sliwkowski, M.X. Untangling the ErbB signalling network. Nat. Rev. Mol. Cell Biol. 2001, 2, 127–137. [Google Scholar] [CrossRef]
- Peschard, P.; Fournier, T.M.; Lamorte, L.; Naujokas, M.A.; Band, H.; Langdon, W.Y.; Park, M. Mutation of the c-Cbl TKB domain binding site on the Met receptor tyrosine kinase converts it into a transforming protein. Mol. Cell 2001, 8, 995–1004. [Google Scholar] [CrossRef] [PubMed]
- Lefebvre, J.; Ancot, F.; Leroy, C.; Muharram, G.; Lemiere, A.; Tulasne, D. Met degradation: More than one stone to shoot a receptor down. FASEB J. 2012, 26, 1387–1399. [Google Scholar] [CrossRef] [PubMed]
- Abella, J.V.; Peschard, P.; Naujokas, M.A.; Lin, T.; Saucier, C.; Urbe, S.; Park, M. Met/Hepatocyte growth factor receptor ubiquitination suppresses transformation and is required for Hrs phosphorylation. Mol. Cell Biol. 2005, 25, 9632–9645. [Google Scholar] [CrossRef]
- Tan, Y.H.; Krishnaswamy, S.; Nandi, S.; Kanteti, R.; Vora, S.; Onel, K.; Hasina, R.; Lo, F.Y.; El-Hashani, E.; Cervantes, G.; et al. CBL is frequently altered in lung cancers: Its relationship to mutations in MET and EGFR tyrosine kinases. PLoS ONE 2010, 5, e8972. [Google Scholar] [CrossRef]
- Villa-Moruzzi, E.; Puntoni, F.; Bardelli, A.; Vigna, E.; De Rosa, S.; Comoglio, P.M. Protein tyrosine phosphatase PTP-S binds to the juxtamembrane region of the hepatocyte growth factor receptor Met. Biochem. J. 1998, 336 Pt 1, 235–239. [Google Scholar] [CrossRef]
- Lee, C.C.; Yamada, K.M. Identification of a novel type of alternative splicing of a tyrosine kinase receptor. Juxtamembrane deletion of the c-met protein kinase C serine phosphorylation regulatory site. J. Biol. Chem. 1994, 269, 19457–19461. [Google Scholar] [CrossRef]
- Lee, J.H.; Gao, C.F.; Lee, C.C.; Kim, M.D.; Vande Woude, G.F. An alternatively spliced form of Met receptor is tumorigenic. Exp. Mol. Med. 2006, 38, 565–573. [Google Scholar] [CrossRef]
- Blagoev, B.; Ong, S.E.; Kratchmarova, I.; Mann, M. Temporal analysis of phosphotyrosine-dependent signaling networks by quantitative proteomics. Nat. Biotechnol. 2004, 22, 1139–1145. [Google Scholar] [CrossRef] [PubMed]
- Trusolino, L.; Bertotti, A.; Comoglio, P.M. A signaling adapter function for alpha6beta4 integrin in the control of HGF-dependent invasive growth. Cell 2001, 107, 643–654. [Google Scholar] [CrossRef]
- Schlessinger, J. Receptor tyrosine kinases: Legacy of the first two decades. Cold Spring Harb. Perspect. Biol. 2014, 6, a008912. [Google Scholar] [CrossRef]
- Go, H.; Jeon, Y.K.; Park, H.J.; Sung, S.W.; Seo, J.W.; Chung, D.H. High MET gene copy number leads to shorter survival in patients with non-small cell lung cancer. J. Thorac. Oncol. 2010, 5, 305–313. [Google Scholar] [CrossRef]
- Graziano, F.; Galluccio, N.; Lorenzini, P.; Ruzzo, A.; Canestrari, E.; D’Emidio, S.; Catalano, V.; Sisti, V.; Ligorio, C.; Andreoni, F.; et al. Genetic activation of the MET pathway and prognosis of patients with high-risk, radically resected gastric cancer. J. Clin. Oncol. 2011, 29, 4789–4795. [Google Scholar] [CrossRef]
- Zhang, M.; Li, G.; Sun, X.; Ni, S.; Tan, C.; Xu, M.; Huang, D.; Ren, F.; Li, D.; Wei, P.; et al. MET amplification, expression, and exon 14 mutations in colorectal adenocarcinoma. Hum. Pathol. 2018, 77, 108–115. [Google Scholar] [CrossRef]
- Pal, S.K.; Ali, S.M.; Yakirevich, E.; Geynisman, D.M.; Karam, J.A.; Elvin, J.A.; Frampton, G.M.; Huang, X.; Lin, D.I.; Rosenzweig, M.; et al. Characterization of Clinical Cases of Advanced Papillary Renal Cell Carcinoma via Comprehensive Genomic Profiling. Eur. Urol. 2018, 73, 71–78. [Google Scholar] [CrossRef] [PubMed]
- Cheng, F.; Guo, D. MET in glioma: Signaling pathways and targeted therapies. J. Exp. Clin. Cancer Res. 2019, 38, 270. [Google Scholar] [CrossRef] [PubMed]
- Zhou, Y.; Song, K.Y.; Giubellino, A. The Role of MET in Melanoma and Melanocytic Lesions. Am. J. Pathol. 2019, 189, 2138–2148. [Google Scholar] [CrossRef]
- Houldsworth, J.; Cordon-Cardo, C.; Ladanyi, M.; Kelsen, D.P.; Chaganti, R.S. Gene amplification in gastric and esophageal adenocarcinomas. Cancer Res. 1990, 50, 6417–6422. [Google Scholar]
- Kuniyasu, H.; Yasui, W.; Kitadai, Y.; Yokozaki, H.; Ito, H.; Tahara, E. Frequent amplification of the c-met gene in scirrhous type stomach cancer. Biochem. Biophys. Res. Commun. 1992, 189, 227–232. [Google Scholar] [CrossRef] [PubMed]
- Hara, T.; Ooi, A.; Kobayashi, M.; Mai, M.; Yanagihara, K.; Nakanishi, I. Amplification of c-myc, K-sam, and c-met in gastric cancers: Detection by fluorescence in situ hybridization. Lab. Investig. 1998, 78, 1143–1153. [Google Scholar]
- Miller, C.T.; Lin, L.; Casper, A.M.; Lim, J.; Thomas, D.G.; Orringer, M.B.; Chang, A.C.; Chambers, A.F.; Giordano, T.J.; Glover, T.W.; et al. Genomic amplification of MET with boundaries within fragile site FRA7G and upregulation of MET pathways in esophageal adenocarcinoma. Oncogene 2006, 25, 409–418. [Google Scholar] [CrossRef] [PubMed]
- Engelman, J.A.; Zejnullahu, K.; Mitsudomi, T.; Song, Y.; Hyland, C.; Park, J.O.; Lindeman, N.; Gale, C.M.; Zhao, X.; Christensen, J.; et al. MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science 2007, 316, 1039–1043. [Google Scholar] [CrossRef] [PubMed]
- Bubendorf, L.; Dafni, U.; Schobel, M.; Finn, S.P.; Tischler, V.; Sejda, A.; Marchetti, A.; Thunnissen, E.; Verbeken, E.K.; Warth, A.; et al. Prevalence and clinical association of MET gene overexpression and amplification in patients with NSCLC: Results from the European Thoracic Oncology Platform (ETOP) Lungscape project. Lung Cancer 2017, 111, 143–149. [Google Scholar] [CrossRef]
- Bean, J.; Brennan, C.; Shih, J.Y.; Riely, G.; Viale, A.; Wang, L.; Chitale, D.; Motoi, N.; Szoke, J.; Broderick, S.; et al. MET amplification occurs with or without T790M mutations in EGFR mutant lung tumors with acquired resistance to gefitinib or erlotinib. Proc. Natl. Acad. Sci. USA 2007, 104, 20932–20937. [Google Scholar] [CrossRef]
- Bardelli, A.; Corso, S.; Bertotti, A.; Hobor, S.; Valtorta, E.; Siravegna, G.; Sartore-Bianchi, A.; Scala, E.; Cassingena, A.; Zecchin, D.; et al. Amplification of the MET receptor drives resistance to anti-EGFR therapies in colorectal cancer. Cancer Discov. 2013, 3, 658–673. [Google Scholar] [CrossRef]
- Bandyk, M.G.; Zhao, L.; Troncoso, P.; Pisters, L.L.; Palmer, J.L.; von Eschenbach, A.C.; Chung, L.W.; Liang, J.C. Trisomy 7: A potential cytogenetic marker of human prostate cancer progression. Genes. Chromosomes Cancer 1994, 9, 19–27. [Google Scholar] [CrossRef] [PubMed]
- Broberg, K.; Toksvig-Larsen, S.; Lindstrand, A.; Mertens, F. Trisomy 7 accumulates with age in solid tumors and non-neoplastic synovia. Genes. Chromosomes Cancer 2001, 30, 310–315. [Google Scholar] [CrossRef]
- Ried, T.; Knutzen, R.; Steinbeck, R.; Blegen, H.; Schröck, E.; Heselmeyer, K.; du Manoir, S.; Auer, G. Comparative genomic hybridization reveals a specific pattern of chromosomal gains and losses during the genesis of colorectal tumors. Genes Chromosomes Cancer 1996, 15, 234–245. [Google Scholar] [CrossRef]
- Benvenuti, S.; Gentile, A.; Lazzari, L.; Arnesano, A.; Trusolino, L.; Comoglio, P.M. An ‘in-cell trial’ to assess the efficacy of a monovalent anti-MET antibody as monotherapy and in association with standard cytotoxics. Mol. Oncol. 2014, 8, 378–388. [Google Scholar] [CrossRef]
- Sequist, L.V.; Han, J.Y.; Ahn, M.J.; Cho, B.C.; Yu, H.; Kim, S.W.; Yang, J.C.; Lee, J.S.; Su, W.C.; Kowalski, D.; et al. Osimertinib plus savolitinib in patients with EGFR mutation-positive, MET-amplified, non-small-cell lung cancer after progression on EGFR tyrosine kinase inhibitors: Interim results from a multicentre, open-label, phase 1b study. Lancet Oncol. 2020, 21, 373–386. [Google Scholar] [CrossRef] [PubMed]
- Choueiri, T.K.; Heng, D.Y.C.; Lee, J.L.; Cancel, M.; Verheijen, R.B.; Mellemgaard, A.; Ottesen, L.H.; Frigault, M.M.; L’Hernault, A.; Szijgyarto, Z.; et al. Efficacy of Savolitinib vs Sunitinib in Patients With MET-Driven Papillary Renal Cell Carcinoma: The SAVOIR Phase 3 Randomized Clinical Trial. JAMA Oncol. 2020, 6, 1247–1255. [Google Scholar] [CrossRef]
- Awad, M.M.; Oxnard, G.R.; Jackman, D.M.; Savukoski, D.O.; Hall, D.; Shivdasani, P.; Heng, J.C.; Dahlberg, S.E.; Janne, P.A.; Verma, S.; et al. MET Exon 14 Mutations in Non-Small-Cell Lung Cancer Are Associated With Advanced Age and Stage-Dependent MET Genomic Amplification and c-Met Overexpression. J. Clin. Oncol. 2016, 34, 721–730. [Google Scholar] [CrossRef]
- Lennerz, J.K.; Kwak, E.L.; Ackerman, A.; Michael, M.; Fox, S.B.; Bergethon, K.; Lauwers, G.Y.; Christensen, J.G.; Wilner, K.D.; Haber, D.A. MET amplification identifies a small and aggressive subgroup of esophagogastric adenocarcinoma with evidence of responsiveness to crizotinib. J. Clin. Oncol. 2011, 29, 4803. [Google Scholar] [CrossRef]
- Mo, H.N.; Liu, P. Targeting MET in cancer therapy. Chronic Dis. Transl. Med. 2017, 3, 148–153. [Google Scholar] [CrossRef] [PubMed]
- Rosen, E.Y.; Johnson, M.L.; Clifford, S.E.; Somwar, R.; Kherani, J.F.; Son, J.; Bertram, A.A.; Davare, M.A.; Gladstone, E.; Ivanova, E.V.; et al. Overcoming MET-Dependent Resistance to Selective RET Inhibition in Patients with RET Fusion-Positive Lung Cancer by Combining Selpercatinib with Crizotinib. Clin. Cancer Res. 2021, 27, 34–42. [Google Scholar] [CrossRef] [PubMed]
- Pal, S.K.; Tangen, C.; Thompson, I.M., Jr.; Balzer-Haas, N.; George, D.J.; Heng, D.Y.C.; Shuch, B.; Stein, M.; Tretiakova, M.; Humphrey, P.; et al. A comparison of sunitinib with cabozantinib, crizotinib, and savolitinib for treatment of advanced papillary renal cell carcinoma: A randomised, open-label, phase 2 trial. Lancet 2021, 397, 695–703. [Google Scholar] [CrossRef]
- Daud, A.; Kluger, H.M.; Kurzrock, R.; Schimmoller, F.; Weitzman, A.L.; Samuel, T.A.; Moussa, A.H.; Gordon, M.S.; Shapiro, G.I. Phase II randomised discontinuation trial of the MET/VEGF receptor inhibitor cabozantinib in metastatic melanoma. Br. J. Cancer 2017, 116, 432–440. [Google Scholar] [CrossRef]
- Paik, P.K.; Drilon, A.; Fan, P.-D.; Yu, H.; Rekhtman, N.; Ginsberg, M.S.; Borsu, L.; Schultz, N.; Berger, M.F.; Rudin, C.M. Response to MET Inhibitors in Patients with Stage IV Lung Adenocarcinomas Harboring MET Mutations Causing Exon 14 SkippingMET Inhibitors in MET Exon 14 Splice Variant Lung Cancer. Cancer Discov. 2015, 5, 842–849. [Google Scholar] [CrossRef]
- Elisei, R.; Schlumberger, M.J.; Muller, S.P.; Schoffski, P.; Brose, M.S.; Shah, M.H.; Licitra, L.; Jarzab, B.; Medvedev, V.; Kreissl, M.C.; et al. Cabozantinib in progressive medullary thyroid cancer. J. Clin. Oncol. 2013, 31, 3639–3646. [Google Scholar] [CrossRef]
- Shah, M.A.; Wainberg, Z.A.; Catenacci, D.V.; Hochster, H.S.; Ford, J.; Kunz, P.; Lee, F.C.; Kallender, H.; Cecchi, F.; Rabe, D.C.; et al. Phase II study evaluating 2 dosing schedules of oral foretinib (GSK1363089), cMET/VEGFR2 inhibitor, in patients with metastatic gastric cancer. PLoS ONE 2013, 8, e54014. [Google Scholar] [CrossRef] [PubMed]
- Patnaik, A.; Gadgeel, S.; Papadopoulos, K.P.; Rasco, D.W.; Haas, N.B.; Der-Torossian, H.; Faltaos, D.; Potvin, D.; Tassell, V.; Tawashi, M.; et al. Phase I Study of Glesatinib (MGCD265) in Combination with Erlotinib or Docetaxel in Patients with Advanced Solid Tumors. Target. Oncol. 2022, 17, 125–138. [Google Scholar] [CrossRef] [PubMed]
- Kudo, M.; Morimoto, M.; Moriguchi, M.; Izumi, N.; Takayama, T.; Yoshiji, H.; Hino, K.; Oikawa, T.; Chiba, T.; Motomura, K.; et al. A randomized, double-blind, placebo-controlled, phase 3 study of tivantinib in Japanese patients with MET-high hepatocellular carcinoma. Cancer Sci. 2020, 111, 3759–3769. [Google Scholar] [CrossRef] [PubMed]
- Basilico, C.; Pennacchietti, S.; Vigna, E.; Chiriaco, C.; Arena, S.; Bardelli, A.; Valdembri, D.; Serini, G.; Michieli, P. Tivantinib (ARQ197) displays cytotoxic activity that is independent of its ability to bind MET. Clin. Cancer Res. 2013, 19, 2381–2392. [Google Scholar] [CrossRef] [PubMed]
- Calles, A.; Kwiatkowski, N.; Cammarata, B.K.; Ercan, D.; Gray, N.S.; Jänne, P.A. Tivantinib (ARQ 197) efficacy is independent of MET inhibition in non-small-cell lung cancer cell lines. Mol. Oncol. 2015, 9, 260–269. [Google Scholar] [CrossRef] [PubMed]
- Wolf, J.; Seto, T.; Han, J.-Y.; Reguart, N.; Garon, E.B.; Groen, H.J.; Tan, D.S.; Hida, T.; de Jonge, M.; Orlov, S.V. Capmatinib in MET exon 14–mutated or MET-amplified non–small-cell lung cancer. N. Engl. J. Med. 2020, 383, 944–957. [Google Scholar] [CrossRef]
- Wu, Y.L.; Zhang, L.; Kim, D.W.; Liu, X.; Lee, D.H.; Yang, J.C.; Ahn, M.J.; Vansteenkiste, J.F.; Su, W.C.; Felip, E.; et al. Phase Ib/II Study of Capmatinib (INC280) Plus Gefitinib After Failure of Epidermal Growth Factor Receptor (EGFR) Inhibitor Therapy in Patients With EGFR-Mutated, MET Factor-Dysregulated Non-Small-Cell Lung Cancer. J. Clin. Oncol. 2018, 36, 3101–3109. [Google Scholar] [CrossRef]
- Schuler, M.; Berardi, R.; Lim, W.T.; de Jonge, M.; Bauer, T.M.; Azaro, A.; Gottfried, M.; Han, J.Y.; Lee, D.H.; Wollner, M.; et al. Molecular correlates of response to capmatinib in advanced non-small-cell lung cancer: Clinical and biomarker results from a phase I trial. Ann. Oncol. 2020, 31, 789–797. [Google Scholar] [CrossRef] [PubMed]
- Dagogo-Jack, I.; Moonsamy, P.; Gainor, J.F.; Lennerz, J.K.; Piotrowska, Z.; Lin, J.J.; Lennes, I.T.; Sequist, L.V.; Shaw, A.T.; Goodwin, K.; et al. A Phase 2 Study of Capmatinib in Patients With MET-Altered Lung Cancer Previously Treated With a MET Inhibitor. J. Thorac. Oncol. 2021, 16, 850–859. [Google Scholar] [CrossRef]
- Grande, E.; Giovannini, M.; Marriere, E.; Pultar, P.; Quinlan, M.; Chen, X.; Rahmanzadeh, G.; Curigliano, G.; Cui, X. Effect of capmatinib on the pharmacokinetics of digoxin and rosuvastatin administered as a 2-drug cocktail in patients with MET-dysregulated advanced solid tumours: A phase I, multicentre, open-label, single-sequence drug-drug interaction study. Br. J. Clin. Pharmacol. 2021, 87, 2867–2878. [Google Scholar] [CrossRef] [PubMed]
- Paik, P.K.; Felip, E.; Veillon, R.; Sakai, H.; Cortot, A.B.; Garassino, M.C.; Mazieres, J.; Viteri, S.; Senellart, H.; Van Meerbeeck, J.; et al. Tepotinib in Non-Small-Cell Lung Cancer with MET Exon 14 Skipping Mutations. N. Engl. J. Med. 2020, 383, 931–943. [Google Scholar] [CrossRef]
- Wu, Y.L.; Cheng, Y.; Zhou, J.; Lu, S.; Zhang, Y.; Zhao, J.; Kim, D.W.; Soo, R.A.; Kim, S.W.; Pan, H.; et al. Tepotinib plus gefitinib in patients with EGFR-mutant non-small-cell lung cancer with MET overexpression or MET amplification and acquired resistance to previous EGFR inhibitor (INSIGHT study): An open-label, phase 1b/2, multicentre, randomised trial. Lancet Respir. Med. 2020, 8, 1132–1143. [Google Scholar] [CrossRef] [PubMed]
- Decaens, T.; Barone, C.; Assenat, E.; Wermke, M.; Fasolo, A.; Merle, P.; Blanc, J.F.; Grando, V.; Iacobellis, A.; Villa, E.; et al. Phase 1b/2 trial of tepotinib in sorafenib pretreated advanced hepatocellular carcinoma with MET overexpression. Br. J. Cancer 2021, 125, 190–199. [Google Scholar] [CrossRef] [PubMed]
- Dieras, V.; Campone, M.; Yardley, D.A.; Romieu, G.; Valero, V.; Isakoff, S.J.; Koeppen, H.; Wilson, T.R.; Xiao, Y.; Shames, D.S.; et al. Randomized, phase II, placebo-controlled trial of onartuzumab and/or bevacizumab in combination with weekly paclitaxel in patients with metastatic triple-negative breast cancer. Ann. Oncol. 2015, 26, 1904–1910. [Google Scholar] [CrossRef]
- Shah, M.A.; Bang, Y.J.; Lordick, F.; Alsina, M.; Chen, M.; Hack, S.P.; Bruey, J.M.; Smith, D.; McCaffery, I.; Shames, D.S.; et al. Effect of Fluorouracil, Leucovorin, and Oxaliplatin With or Without Onartuzumab in HER2-Negative, MET-Positive Gastroesophageal Adenocarcinoma: The METGastric Randomized Clinical Trial. JAMA Oncol. 2017, 3, 620–627. [Google Scholar] [CrossRef]
- Kishi, K.; Sakai, H.; Seto, T.; Kozuki, T.; Nishio, M.; Imamura, F.; Nokihara, H.; Satouchi, M.; Nakagawa, S.; Tahata, T.; et al. First-line onartuzumab plus erlotinib treatment for patients with MET-positive and EGFR mutation-positive non-small-cell lung cancer. Cancer Treat. Res. Commun. 2019, 18, 100113. [Google Scholar] [CrossRef]
- Martens, T.; Schmidt, N.-O.; Eckerich, C.; Fillbrandt, R.; Merchant, M.; Schwall, R.; Westphal, M.; Lamszus, K. A novel one-armed anti-c-Met antibody inhibits glioblastoma growth in vivo. Clin. Cancer Res. 2006, 12, 6144–6152. [Google Scholar] [CrossRef]
- Harding, J.J.; Zhu, A.X.; Bauer, T.M.; Choueiri, T.K.; Drilon, A.; Voss, M.H.; Fuchs, C.S.; Abou-Alfa, G.K.; Wijayawardana, S.R.; Wang, X.A.; et al. A Phase Ib/II Study of Ramucirumab in Combination with Emibetuzumab in Patients with Advanced Cancer. Clin. Cancer Res. 2019, 25, 5202–5211. [Google Scholar] [CrossRef]
- Scagliotti, G.; Moro-Sibilot, D.; Kollmeier, J.; Favaretto, A.; Cho, E.K.; Grosch, H.; Kimmich, M.; Girard, N.; Tsai, C.M.; Hsia, T.C.; et al. A Randomized-Controlled Phase 2 Study of the MET Antibody Emibetuzumab in Combination with Erlotinib as First-Line Treatment for EGFR Mutation-Positive NSCLC Patients. J. Thorac. Oncol. 2020, 15, 80–90. [Google Scholar] [CrossRef] [PubMed]
- Camidge, D.R.; Moran, T.; Demedts, I.; Grosch, H.; Mileham, K.; Molina, J.; Juan-Vidal, O.; Bepler, G.; Goldman, J.W.; Park, K.; et al. A Randomized, Open-Label Phase II Study Evaluating Emibetuzumab Plus Erlotinib and Emibetuzumab Monotherapy in MET Immunohistochemistry Positive NSCLC Patients with Acquired Resistance to Erlotinib. Clin. Lung Cancer 2022, 23, 300–310. [Google Scholar] [CrossRef] [PubMed]
- Liu, L.; Zeng, W.; Wortinger, M.A.; Yan, S.B.; Cornwell, P.; Peek, V.L.; Stephens, J.R.; Tetreault, J.W.; Xia, J.; Manro, J.R. LY2875358, a neutralizing and internalizing anti-MET bivalent antibody, inhibits HGF-dependent and HGF-independent MET activation and tumor growth. Clin. Cancer Res. 2014, 20, 6059–6070. [Google Scholar] [CrossRef]
- Hultberg, A.; Morello, V.; Huyghe, L.; De Jonge, N.; Blanchetot, C.; Hanssens, V.; De Boeck, G.; Silence, K.; Festjens, E.; Heukers, R. Depleting MET-Expressing Tumor Cells by ADCC Provides a Therapeutic Advantage over Inhibiting HGF/MET SignalingARGX-111 Kills MET-Expressing Cancer Cells by ADCC. Cancer Res. 2015, 75, 3373–3383. [Google Scholar] [CrossRef]
- Lee, J.; Kim, S.T.; Park, S.; Lee, S.; Park, S.H.; Park, J.O.; Lim, H.Y.; Ahn, H.; Bok, H.; Kim, K.M.; et al. Phase I Trial of Anti-MET Monoclonal Antibody in MET-Overexpressed Refractory Cancer. Clin. Color. Cancer 2018, 17, 140–146. [Google Scholar] [CrossRef]
- Lee, B.-S.; Kang, S.; Kim, K.-A.; Song, Y.-J.; Cheong, K.H.; Cha, H.-Y.; Kim, C.H. Met degradation by SAIT301, a Met monoclonal antibody, reduces the invasion and migration of nasopharyngeal cancer cells via inhibition of EGR-1 expression. Cell Death Dis. 2014, 5, e1159. [Google Scholar] [CrossRef]
- Lee, J.M.; Kim, B.; Lee, S.; Jeong, Y.; Oh, Y.; Song, Y.; Jung, S.; Choi, J.; Lee, S.; Cheong, K. Cbl-independent degradation of Met: Ways to avoid agonism of bivalent Met-targeting antibody. Oncogene 2014, 33, 34–43. [Google Scholar] [CrossRef]
- Catenacci, D.V.T.; Tebbutt, N.C.; Davidenko, I.; Murad, A.M.; Al-Batran, S.E.; Ilson, D.H.; Tjulandin, S.; Gotovkin, E.; Karaszewska, B.; Bondarenko, I.; et al. Rilotumumab plus epirubicin, cisplatin, and capecitabine as first-line therapy in advanced MET-positive gastric or gastro-oesophageal junction cancer (RILOMET-1): A randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2017, 18, 1467–1482. [Google Scholar] [CrossRef]
- Affronti, M.L.; Jackman, J.G.; McSherry, F.; Herndon, J.E., 2nd; Massey, E.C., Jr.; Lipp, E.; Desjardins, A.; Friedman, H.S.; Vlahovic, G.; Vredenburgh, J.; et al. Phase II Study to Evaluate the Efficacy and Safety of Rilotumumab and Bevacizumab in Subjects with Recurrent Malignant Glioma. Oncologist 2018, 23, 889-e898. [Google Scholar] [CrossRef] [PubMed]
- Burgess, T.L.; Sun, J.; Meyer, S.; Tsuruda, T.S.; Sun, J.; Elliott, G.; Chen, Q.; Haniu, M.; Barron, W.F.; Juan, T. Biochemical Characterization of AMG 102: A Neutralizing, Fully Human Monoclonal Antibody to Human and Nonhuman Primate Hepatocyte Growth FactorAMG 102 Neutralizes Hepatocyte Growth Factor. Mol. Cancer Ther. 2010, 9, 400–409. [Google Scholar] [CrossRef]
- Mok, T.S.; Geater, S.L.; Su, W.C.; Tan, E.H.; Yang, J.C.; Chang, G.C.; Han, M.; Komarnitsky, P.; Payumo, F.; Garrus, J.E.; et al. A Randomized Phase 2 Study Comparing the Combination of Ficlatuzumab and Gefitinib with Gefitinib Alone in Asian Patients with Advanced Stage Pulmonary Adenocarcinoma. J. Thorac. Oncol. 2016, 11, 1736–1744. [Google Scholar] [CrossRef] [PubMed]
- Patnaik, A.; Weiss, G.J.; Papadopoulos, K.P.; Hofmeister, C.C.; Tibes, R.; Tolcher, A.; Isaacs, R.; Jac, J.; Han, M.; Payumo, F.C.; et al. Phase I ficlatuzumab monotherapy or with erlotinib for refractory advanced solid tumours and multiple myeloma. Br. J. Cancer 2014, 111, 272–280. [Google Scholar] [CrossRef] [PubMed]
- D’Arcangelo, M.; Cappuzzo, F. Focus on the potential role of ficlatuzumab in the treatment of non-small cell lung cancer. Biol. Targets Ther. 2013, 7, 61. [Google Scholar] [CrossRef] [PubMed]
- Du, Z.; Lovly, C.M. Mechanisms of receptor tyrosine kinase activation in cancer. Mol. Cancer 2018, 17, 58. [Google Scholar] [CrossRef] [PubMed]
- Zehir, A.; Benayed, R.; Shah, R.H.; Syed, A.; Middha, S.; Kim, H.R.; Srinivasan, P.; Gao, J.; Chakravarty, D.; Devlin, S.M.; et al. Mutational landscape of metastatic cancer revealed from prospective clinical sequencing of 10,000 patients. Nat. Med. 2017, 23, 703–713. [Google Scholar] [CrossRef] [PubMed]
- Vuong, H.G.; Ho, A.T.N.; Altibi, A.M.A.; Nakazawa, T.; Katoh, R.; Kondo, T. Clinicopathological implications of MET exon 14 mutations in non-small cell lung cancer—A systematic review and meta-analysis. Lung Cancer 2018, 123, 76–82. [Google Scholar] [CrossRef]
- Vigna, E.; Gramaglia, D.; Longati, P.; Bardelli, A.; Comoglio, P.M. Loss of the exon encoding the juxtamembrane domain is essential for the oncogenic activation of TPR-MET. Oncogene 1999, 18, 4275–4281. [Google Scholar] [CrossRef]
- US Food and Drug Administration. TABRECTA (Capmatinib) Prescribing Information; US Food and Drug Administration: Silver Spring, MD, USA, 2022.
- Awad, M.M.; Leonardi, G.C.; Kravets, S.; Dahlberg, S.E.; Drilon, A.; Noonan, S.A.; Camidge, D.R.; Ou, S.-H.I.; Costa, D.B.; Gadgeel, S.M. Impact of MET inhibitors on survival among patients with non-small cell lung cancer harboring MET exon 14 mutations: A retrospective analysis. Lung Cancer 2019, 133, 96–102. [Google Scholar] [CrossRef]
- Lu, S.; Fang, J.; Li, X.; Cao, L.; Zhou, J.; Guo, Q.; Liang, Z.; Cheng, Y.; Jiang, L.; Yang, N.; et al. Phase II study of savolitinib in patients (pts) with pulmonary sarcomatoid carcinoma (PSC) and other types of non-small cell lung cancer (NSCLC) harboring MET exon 14 skipping mutations (METex14+). J. Clin. Oncol. 2020, 38, 9519. [Google Scholar] [CrossRef]
- Engstrom, L.D.; Aranda, R.; Lee, M.; Tovar, E.A.; Essenburg, C.J.; Madaj, Z.; Chiang, H.; Briere, D.; Hallin, J.; Lopez-Casas, P.P.; et al. Glesatinib Exhibits Antitumor Activity in Lung Cancer Models and Patients Harboring MET Exon 14 Mutations and Overcomes Mutation-mediated Resistance to Type I MET Inhibitors in Nonclinical Models. Clin. Cancer Res. 2017, 23, 6661–6672. [Google Scholar] [CrossRef]
- Hu, H.; Mu, Q.; Bao, Z.; Chen, Y.; Liu, Y.; Chen, J.; Wang, K.; Wang, Z.; Nam, Y.; Jiang, B.; et al. Mutational Landscape of Secondary Glioblastoma Guides MET-Targeted Trial in Brain Tumor. Cell 2018, 175, 1665–1678.e18. [Google Scholar] [CrossRef]
- Klempner, S.J.; Borghei, A.; Hakimian, B.; Ali, S.M.; Ou, S.I. Intracranial Activity of Cabozantinib in MET Exon 14-Positive NSCLC with Brain Metastases. J. Thorac. Oncol. 2017, 12, 152–156. [Google Scholar] [CrossRef] [PubMed]
- Moro-Sibilot, D.; Cozic, N.; Perol, M.; Mazieres, J.; Otto, J.; Souquet, P.J.; Bahleda, R.; Wislez, M.; Zalcman, G.; Guibert, S.D.; et al. Crizotinib in c-MET- or ROS1-positive NSCLC: Results of the AcSe phase II trial. Ann. Oncol. 2019, 30, 1985–1991. [Google Scholar] [CrossRef] [PubMed]
- Salgia, R.; Sattler, M.; Scheele, J.; Stroh, C.; Felip, E. The promise of selective MET inhibitors in non-small cell lung cancer with MET exon 14 skipping. Cancer Treat. Rev. 2020, 87, 102022. [Google Scholar] [CrossRef]
- Fujino, T.; Suda, K.; Mitsudomi, T. Lung Cancer with MET exon 14 Skipping Mutation: Genetic Feature, Current Treatments, and Future Challenges. Lung Cancer 2021, 12, 35–50. [Google Scholar] [CrossRef] [PubMed]
- Wang, F.; Liu, Y.; Qiu, W.; Shum, E.; Feng, M.; Zhao, D.; Zheng, D.; Borczuk, A.; Cheng, H.; Halmos, B. Functional Analysis of MET Exon 14 Skipping Alteration in Cancer Invasion and Metastatic Dissemination. Cancer Res. 2022, 82, 1365–1379. [Google Scholar] [CrossRef]
- Rotow, J.K.; Gui, P.; Wu, W.; Raymond, V.M.; Lanman, R.B.; Kaye, F.J.; Peled, N.; Fece de la Cruz, F.; Nadres, B.; Corcoran, R.B.; et al. Co-occurring Alterations in the RAS-MAPK Pathway Limit Response to MET Inhibitor Treatment in MET Exon 14 Skipping Mutation-Positive Lung Cancer. Clin. Cancer Res. 2020, 26, 439–449. [Google Scholar] [CrossRef]
- Jamme, P.; Fernandes, M.; Copin, M.C.; Descarpentries, C.; Escande, F.; Morabito, A.; Gregoire, V.; Jamme, M.; Baldacci, S.; Tulasne, D.; et al. Alterations in the PI3K Pathway Drive Resistance to MET Inhibitors in NSCLC Harboring MET Exon 14 Skipping Mutations. J. Thorac. Oncol. 2020, 15, 741–751. [Google Scholar] [CrossRef]
- Schmidt, L.; Duh, F.M.; Chen, F.; Kishida, T.; Glenn, G.; Choyke, P.; Scherer, S.W.; Zhuang, Z.; Lubensky, I.; Dean, M.; et al. Germline and somatic mutations in the tyrosine kinase domain of the MET proto-oncogene in papillary renal carcinomas. Nat. Genet. 1997, 16, 68–73. [Google Scholar] [CrossRef]
- Schmidt, L.; Junker, K.; Nakaigawa, N.; Kinjerski, T.; Weirich, G.; Miller, M.; Lubensky, I.; Neumann, H.P.; Brauch, H.; Decker, J. Novel mutations of the MET proto-oncogene in papillary renal carcinomas. Oncogene 1999, 18, 2343–2350. [Google Scholar] [CrossRef] [PubMed]
- Zhuang, Z.; Park, W.S.; Pack, S.; Schmidt, L.; Vortmeyer, A.O.; Pak, E.; Pham, T.; Weil, R.J.; Candidus, S.; Lubensky, I.A.; et al. Trisomy 7-harbouring non-random duplication of the mutant MET allele in hereditary papillary renal carcinomas. Nat. Genet. 1998, 20, 66–69. [Google Scholar] [CrossRef]
- Jeffers, M.; Schmidt, L.; Nakaigawa, N.; Webb, C.P.; Weirich, G.; Kishida, T.; Zbar, B.; Vande Woude, G.F. Activating mutations for the met tyrosine kinase receptor in human cancer. Proc. Natl. Acad. Sci. USA 1997, 94, 11445–11450. [Google Scholar] [CrossRef] [PubMed]
- Jeffers, M.; Fiscella, M.; Webb, C.P.; Anver, M.; Koochekpour, S.; Vande Woude, G.F. The mutationally activated Met receptor mediates motility and metastasis. Proc. Natl. Acad. Sci. USA 1998, 95, 14417–14422. [Google Scholar] [CrossRef] [PubMed]
- Graveel, C.; Su, Y.; Koeman, J.; Wang, L.-M.; Tessarollo, L.; Fiscella, M.; Birchmeier, C.; Swiatek, P.; Bronson, R.; Vande Woude, G. Activating Met mutations produce unique tumor profiles in mice with selective duplication of the mutant allele. Proc. Natl. Acad. Sci. USA 2004, 101, 17198–17203. [Google Scholar] [CrossRef]
- Graveel, C.R.; London, C.A.; Woude, G.F.V. A mouse model of activating Met mutations. Cell Cycle 2005, 4, 518–520. [Google Scholar] [CrossRef]
- Park, W.S.; Dong, S.M.; Kim, S.Y.; Na, E.Y.; Shin, M.S.; Pi, J.H.; Kim, B.J.; Bae, J.H.; Hong, Y.K.; Lee, K.S. Somatic mutations in the kinase domain of the Met/hepatocyte growth factor receptor gene in childhood hepatocellular carcinomas. Cancer Res. 1999, 59, 307–310. [Google Scholar]
- Di Renzo, M.F.; Olivero, M.; Martone, T.; Maffe, A.; Maggiora, P.; De Stefani, A.; Valente, G.; Giordano, S.; Cortesina, G.; Comoglio, P. Somatic mutations of the MET oncogene are selected during metastatic spread of human HNSC carcinomas. Oncogene 2000, 19, 1547–1555. [Google Scholar] [CrossRef]
- Aebersold, D.M.; Landt, O.; Berthou, S.; Gruber, G.; Beer, K.T.; Greiner, R.H.; Zimmer, Y. Prevalence and clinical impact of Met Y1253D-activating point mutation in radiotherapy-treated squamous cell cancer of the oropharynx. Oncogene 2003, 22, 8519–8523. [Google Scholar] [CrossRef]
- Lee, J.-H.; Han, S.-U.; Cho, H.; Jennings, B.; Gerrard, B.; Dean, M.; Schmidt, L.; Zbar, B.; Vande Woude, G.F. A novel germ line juxtamembrane Met mutation in human gastric cancer. Oncogene 2000, 19, 4947–4953. [Google Scholar] [CrossRef]
- Ghadjar, P.; Blank-Liss, W.; Simcock, M.; Hegyi, I.; Beer, K.T.; Moch, H.; Aebersold, D.M.; Zimmer, Y. MET Y1253D-activating point mutation and development of distant metastasis in advanced head and neck cancers. Clin. Exp. Metastasis 2009, 26, 809–815. [Google Scholar] [CrossRef]
- Stella, G.M.; Benvenuti, S.; Gramaglia, D.; Scarpa, A.; Tomezzoli, A.; Cassoni, P.; Senetta, R.; Venesio, T.; Pozzi, E.; Bardelli, A. MET mutations in cancers of unknown primary origin (CUPs). Hum. Mutat. 2011, 32, 44–50. [Google Scholar] [CrossRef]
- Neklason, D.W.; Done, M.W.; Sargent, N.R.; Schwartz, A.G.; Anton-Culver, H.; Griffin, C.A.; Ahnen, D.J.; Schildkraut, J.M.; Tomlinson, G.E.; Strong, L.C. Activating mutation in MET oncogene in familial colorectal cancer. BMC Cancer 2011, 11, 424. [Google Scholar] [CrossRef]
- Kong-Beltran, M.; Stamos, J.; Wickramasinghe, D. The Sema domain of Met is necessary for receptor dimerization and activation. Cancer Cell 2004, 6, 75–84. [Google Scholar] [CrossRef] [PubMed]
- Ma, P.C.; Jagadeeswaran, R.; Jagadeesh, S.; Tretiakova, M.S.; Nallasura, V.; Fox, E.A.; Hansen, M.; Schaefer, E.; Naoki, K.; Lader, A. Functional expression and mutations of c-Met and its therapeutic inhibition with SU11274 and small interfering RNA in non–small cell lung cancer. Cancer Res. 2005, 65, 1479–1488. [Google Scholar] [CrossRef] [PubMed]
- de Melo Gagliato, D.; Jardim, D.L.F.; Falchook, G.; Tang, C.; Zinner, R.; Wheler, J.J.; Janku, F.; Subbiah, V.; Piha-Paul, S.A.; Fu, S. Analysis of MET genetic aberrations in patients with breast cancer at MD Anderson Phase I unit. Clin. Breast Cancer 2014, 14, 468–474. [Google Scholar] [CrossRef] [PubMed]
- Sebai, M.; Tulasne, D.; Caputo, S.M.; Verkarre, V.; Fernandes, M.; Guerin, C.; Reinhart, F.; Adams, S.; Maugard, C.; Caron, O.; et al. Novel germline MET pathogenic variants in French patients with papillary renal cell carcinomas type I. Hum. Mutat. 2022, 43, 316–327. [Google Scholar] [CrossRef]
- Verginelli, F.; Pisacane, A.; Gambardella, G.; D’Ambrosio, A.; Candiello, E.; Ferrio, M.; Panero, M.; Casorzo, L.; Benvenuti, S.; Cascardi, E.; et al. Cancer of unknown primary stem-like cells model multi-organ metastasis and unveil liability to MEK inhibition. Nat. Commun. 2021, 12, 2498. [Google Scholar] [CrossRef] [PubMed]
- Wang, R.; Ferrell, L.D.; Faouzi, S.; Maher, J.J.; Bishop, J.M. Activation of the Met receptor by cell attachment induces and sustains hepatocellular carcinomas in transgenic mice. J. Cell Biol. 2001, 153, 1023–1034. [Google Scholar] [CrossRef]
- Jeffers, M.; Rong, S.; Vande Woude, G.F. Hepatocyte growth factor/scatter factor-Met signaling in tumorigenicity and invasion/metastasis. J. Mol. Med. 1996, 74, 505–513. [Google Scholar] [CrossRef]
- Yu, J.; Miehlke, S.; Ebert, M.P.; Hoffmann, J.; Breidert, M.; Alpen, B.; Starzynska, T.; Stolte Prof, M.; Malfertheiner, P.; Bayerdorffer, E. Frequency of TPR-MET rearrangement in patients with gastric carcinoma and in first-degree relatives. Cancer 2000, 88, 1801–1806. [Google Scholar] [CrossRef]
- Stransky, N.; Cerami, E.; Schalm, S.; Kim, J.L.; Lengauer, C. The landscape of kinase fusions in cancer. Nat. Commun. 2014, 5, 4846. [Google Scholar] [CrossRef]
- Bao, Z.S.; Chen, H.M.; Yang, M.Y.; Zhang, C.B.; Yu, K.; Ye, W.L.; Hu, B.Q.; Yan, W.; Zhang, W.; Akers, J.; et al. RNA-seq of 272 gliomas revealed a novel, recurrent PTPRZ1-MET fusion transcript in secondary glioblastomas. Genome Res. 2014, 24, 1765–1773. [Google Scholar] [CrossRef] [PubMed]
- International Cancer Genome Consortium PedBrain Tumor Project. Recurrent MET fusion genes represent a drug target in pediatric glioblastoma. Nat. Med. 2016, 22, 1314–1320. [Google Scholar] [CrossRef] [PubMed]
- Chen, H.M.; Yu, K.; Tang, X.Y.; Bao, Z.S.; Jiang, T.; Fan, X.L.; Chen, X.W.; Su, X.D. Enhanced expression and phosphorylation of the MET oncoprotein by glioma-specific PTPRZ1-MET fusions. FEBS Lett. 2015, 589, 1437–1443. [Google Scholar] [CrossRef]
- Huang, R.; Liu, Y.; Wang, K.; Wang, Z.; Zhang, C.; Zhang, W.; Zhao, Z.; Li, G.; Huang, L.; Chang, Y.; et al. High-sensitive clinical diagnostic method for PTPRZ1-MET and the characteristic protein structure contributing to ligand-independent MET activation. CNS Neurosci. Ther. 2021, 27, 617–628. [Google Scholar] [CrossRef] [PubMed]
- Plenker, D.; Bertrand, M.; de Langen, A.J.; Riedel, R.; Lorenz, C.; Scheel, A.H.; Muller, J.; Bragelmann, J.; Dassler-Plenker, J.; Kobe, C.; et al. Structural Alterations of MET Trigger Response to MET Kinase Inhibition in Lung Adenocarcinoma Patients. Clin. Cancer Res. 2018, 24, 1337–1343. [Google Scholar] [CrossRef]
- Pennacchietti, S.; Michieli, P.; Galluzzo, M.; Mazzone, M.; Giordano, S.; Comoglio, P.M. Hypoxia promotes invasive growth by transcriptional activation of the met protooncogene. Cancer Cell 2003, 3, 347–361. [Google Scholar] [CrossRef]
- De Bacco, F.; Luraghi, P.; Medico, E.; Reato, G.; Girolami, F.; Perera, T.; Gabriele, P.; Comoglio, P.M.; Boccaccio, C. Induction of MET by ionizing radiation and its role in radioresistance and invasive growth of cancer. JNCI J. Natl. Cancer Inst. 2011, 103, 645–661. [Google Scholar] [CrossRef]
- Gambarotta, G.; Pistoi, S.; Giordano, S.; Comoglio, P.M.; Santoro, C. Structure and inducible regulation of the human MET promoter. J. Biol. Chem. 1994, 269, 12852–12857. [Google Scholar] [CrossRef]
- Peters, S.; Adjei, A.A. MET: A promising anticancer therapeutic target. Nat. Rev. Clin. Oncol. 2012, 9, 314–326. [Google Scholar] [CrossRef]
- Linklater, E.S.; Tovar, E.A.; Essenburg, C.J.; Turner, L.; Madaj, Z.; Winn, M.E.; Melnik, M.K.; Korkaya, H.; Maroun, C.R.; Christensen, J.G.; et al. Targeting MET and EGFR crosstalk signaling in triple-negative breast cancers. Oncotarget 2016, 7, 69903–69915. [Google Scholar] [CrossRef] [PubMed]
- Jo, M.; Stolz, D.B.; Esplen, J.E.; Dorko, K.; Michalopoulos, G.K.; Strom, S.C. Cross-talk between epidermal growth factor receptor and c-Met signal pathways in transformed cells. J. Biol. Chem. 2000, 275, 8806–8811. [Google Scholar] [CrossRef]
- Bhanumathy, K.K.; Balagopal, A.; Vizeacoumar, F.S.; Vizeacoumar, F.J.; Freywald, A.; Giambra, V. Protein Tyrosine Kinases: Their Roles and Their Targeting in Leukemia. Cancers 2021, 13, 184. [Google Scholar] [CrossRef] [PubMed]
- Toschi, L.; Janne, P.A. Single-agent and combination therapeutic strategies to inhibit hepatocyte growth factor/MET signaling in cancer. Clin. Cancer Res. 2008, 14, 5941–5946. [Google Scholar] [CrossRef]
- Bell, D.W.; Gore, I.; Okimoto, R.A.; Godin-Heymann, N.; Sordella, R.; Mulloy, R.; Sharma, S.V.; Brannigan, B.W.; Mohapatra, G.; Settleman, J. Inherited susceptibility to lung cancer may be associated with the T790M drug resistance mutation in EGFR. Nat. Genet. 2005, 37, 1315–1316. [Google Scholar] [CrossRef]
- Kobayashi, S.; Boggon, T.J.; Dayaram, T.; Jänne, P.A.; Kocher, O.; Meyerson, M.; Johnson, B.E.; Eck, M.J.; Tenen, D.G.; Halmos, B. EGFR mutation and resistance of non–small-cell lung cancer to gefitinib. N. Engl. J. Med. 2005, 352, 786–792. [Google Scholar] [CrossRef]
- Chen, L.L.; Trent, J.C.; Wu, E.F.; Fuller, G.N.; Ramdas, L.; Zhang, W.; Raymond, A.K.; Prieto, V.G.; Oyedeji, C.O.; Hunt, K.K. A missense mutation in KIT kinase domain 1 correlates with imatinib resistance in gastrointestinal stromal tumors. Cancer Res. 2004, 64, 5913–5919. [Google Scholar] [CrossRef] [PubMed]
- Gorre, M.E.; Mohammed, M.; Ellwood, K.; Hsu, N.; Paquette, R.; Rao, P.N.; Sawyers, C.L. Clinical resistance to STI-571 cancer therapy caused by BCR-ABL gene mutation or amplification. Science 2001, 293, 876–880. [Google Scholar] [CrossRef]
- Chun, S.-Y.; Kwon, Y.-S.; Nam, K.-S.; Kim, S. Lapatinib enhances the cytotoxic effects of doxorubicin in MCF-7 tumorspheres by inhibiting the drug efflux function of ABC transporters. Biomed. Pharmacother. 2015, 72, 37–43. [Google Scholar] [CrossRef]
- Brouwer, K.; Chu, X.; Dahlin, A.; Evers, R.; Fischer, V.; Hillgren, K.; Hoffmaster, K.; Ishikawa, T.; Keppler, D.; Kim, R. Membrane transporters in drug development. Nature 2010, 9, 215–236. [Google Scholar]
- Filomeni, G.; Turella, P.; Dupuis, M.L.; Forini, O.; Ciriolo, M.R.; Cianfriglia, M.; Pezzola, S.; Federici, G.; Caccuri, A.M. 6-(7-Nitro-2, 1, 3-benzoxadiazol-4-ylthio) hexanol, a specific glutathione S-transferase inhibitor, overcomes the multidrug resistance (MDR)-associated protein 1–mediated MDR in small cell lung cancer. Mol. Cancer Ther. 2008, 7, 371–379. [Google Scholar] [CrossRef]
- Rodriguez-Antona, C.; Ingelman-Sundberg, M. Cytochrome P450 pharmacogenetics and cancer. Oncogene 2006, 25, 1679–1691. [Google Scholar] [CrossRef] [PubMed]
- Edwards, D.R.; Handsley, M.M.; Pennington, C.J. The ADAM metalloproteinases. Mol. Asp. Med. 2008, 29, 258–289. [Google Scholar] [CrossRef] [PubMed]
- Vigna, E.; Pacchiana, G.; Mazzone, M.; Chiriaco, C.; Fontani, L.; Basilico, C.; Pennacchietti, S.; Comoglio, P.M. “Active” cancer immunotherapy by anti-Met antibody gene transfer. Cancer Res. 2008, 68, 9176–9183. [Google Scholar] [CrossRef] [PubMed]
- Prat, M.; Crepaldi, T.; Pennacchietti, S.; Bussolino, F.; Comoglio, P.M. Agonistic monoclonal antibodies against the Met receptor dissect the biological responses to HGF. J. Cell Sci. 1998, 111, 237–247. [Google Scholar] [CrossRef]
- Petrelli, A.; Circosta, P.; Granziero, L.; Mazzone, M.; Pisacane, A.; Fenoglio, S.; Comoglio, P.M.; Giordano, S. Ab-induced ectodomain shedding mediates hepatocyte growth factor receptor down-regulation and hampers biological activity. Proc. Natl. Acad. Sci. USA 2006, 103, 5090–5095. [Google Scholar] [CrossRef]
- Schelter, F.; Kobuch, J.; Moss, M.L.; Becherer, J.D.; Comoglio, P.M.; Boccaccio, C.; Krüger, A. A disintegrin and metalloproteinase-10 (ADAM-10) mediates DN30 antibody-induced shedding of the met surface receptor. J. Biol. Chem. 2010, 285, 26335–26340. [Google Scholar] [CrossRef]
- Foveau, B.; Ancot, F.; Leroy, C.; Petrelli, A.; Reiss, K.; Vingtdeux, V.; Giordano, S.; Fafeur, V.; Tulasne, D. Down-regulation of the met receptor tyrosine kinase by presenilin-dependent regulated intramembrane proteolysis. Mol. Biol. Cell 2009, 20, 2495–2507. [Google Scholar] [CrossRef]
- Martinelli, I.; Modica, C.; Chiriaco, C.; Basilico, C.; Hughes, J.M.; Corso, S.; Giordano, S.; Comoglio, P.M.; Vigna, E. hOA-DN30: A highly effective humanized single-arm MET antibody inducing remission of ‘MET-addicted’ cancers. J. Exp. Clin. Cancer Res. 2022, 41, 112. [Google Scholar] [CrossRef] [PubMed]
- Bussolino, F.; Di Renzo, M.F.; Ziche, M.; Bocchietto, E.; Olivero, M.; Naldini, L.; Gaudino, G.; Tamagnone, L.; Coffer, A.; Comoglio, P. Hepatocyte growth factor is a potent angiogenic factor which stimulates endothelial cell motility and growth. J. Cell Biol. 1992, 119, 629–641. [Google Scholar] [CrossRef] [PubMed]
- Wu, X.; Chen, X.; Zhou, Q.; Li, P.; Yu, B.; Li, J.; Qu, Y.; Yan, J.; Yu, Y.; Yan, M. Hepatocyte growth factor activates tumor stromal fibroblasts to promote tumorigenesis in gastric cancer. Cancer Lett. 2013, 335, 128–135. [Google Scholar] [CrossRef] [PubMed]
- Galimi, F.; Cottone, E.; Vigna, E.; Arena, N.; Boccaccio, C.; Giordano, S.; Naldini, L.; Comoglio, P.M. Hepatocyte growth factor is a regulator of monocyte-macrophage function. J. Immunol. 2001, 166, 1241–1247. [Google Scholar] [CrossRef] [PubMed]
- Wang, R.; Zhang, J.; Chen, S.; Lu, M.; Luo, X.; Yao, S.; Liu, S.; Qin, Y.; Chen, H. Tumor-associated macrophages provide a suitable microenvironment for non-small lung cancer invasion and progression. Lung Cancer 2011, 74, 188–196. [Google Scholar] [CrossRef]
- McDermott, U.; Sharma, S.V.; Dowell, L.; Greninger, P.; Montagut, C.; Lamb, J.; Archibald, H.; Raudales, R.; Tam, A.; Lee, D.; et al. Identification of genotype-correlated sensitivity to selective kinase inhibitors by using high-throughput tumor cell line profiling. Proc. Natl. Acad. Sci. USA 2007, 104, 19936–19941. [Google Scholar] [CrossRef]
- Michieli, P.; Mazzone, M.; Basilico, C.; Cavassa, S.; Sottile, A.; Naldini, L.; Comoglio, P.M. Targeting the tumor and its microenvironment by a dual-function decoy Met receptor. Cancer Cell 2004, 6, 61–73. [Google Scholar] [CrossRef]
- Mazzone, M.; Basilico, C.; Cavassa, S.; Pennacchietti, S.; Risio, M.; Naldini, L.; Comoglio, P.M.; Michieli, P. An uncleavable form of pro-scatter factor suppresses tumor growth and dissemination in mice. J. Clin. Investig. 2004, 114, 1418–1432. [Google Scholar] [CrossRef]
- Siravegna, G.; Marsoni, S.; Siena, S.; Bardelli, A. Integrating liquid biopsies into the management of cancer. Nat. Rev. Clin. Oncol. 2017, 14, 531–548. [Google Scholar] [CrossRef]
- Wan, J.; Massie, C.; Garcia-Corbacho, J.; Mouliere, F.; Brenton, J.D.; Caldas, C.; Pacey, S.; Baird, R.; Rosenfeld, N. Liquid biopsies come of age: Towards implementation of circulating tumour DNA. Nat. Rev. Cancer 2017, 17, 223–238. [Google Scholar] [CrossRef]
- Wagle, N.; Emery, C.; Berger, M.F.; Davis, M.J.; Sawyer, A.; Pochanard, P.; Kehoe, S.M.; Johannessen, C.M.; MacConaill, L.E.; Hahn, W.C. Dissecting therapeutic resistance to RAF inhibition in melanoma by tumor genomic profiling. J. Clin. Oncol. 2011, 29, 3085. [Google Scholar] [CrossRef]
- Russo, M.; Siravegna, G.; Blaszkowsky, L.S.; Corti, G.; Crisafulli, G.; Ahronian, L.G.; Mussolin, B.; Kwak, E.L.; Buscarino, M.; Lazzari, L. Tumor Heterogeneity and Lesion-Specific Response to Targeted Therapy in Colorectal CancerLesion-Specific Response to Therapy in Colorectal Cancer. Cancer Discov. 2016, 6, 147–153. [Google Scholar] [CrossRef]
- Tabassum, D.P.; Polyak, K. Tumorigenesis: It takes a village. Nat. Rev. Cancer 2015, 15, 473–483. [Google Scholar] [CrossRef]
- Russo, M.; Pompei, S.; Sogari, A.; Corigliano, M.; Crisafulli, G.; Puliafito, A.; Lamba, S.; Erriquez, J.; Bertotti, A.; Gherardi, M.; et al. A modified fluctuation-test framework characterizes the population dynamics and mutation rate of colorectal cancer persister cells. Nat. Genet. 2022, 54, 976–984. [Google Scholar] [CrossRef] [PubMed]
- Cipponi, A.; Goode, D.L.; Bedo, J.; McCabe, M.J.; Pajic, M.; Croucher, D.R.; Rajal, A.G.; Junankar, S.R.; Saunders, D.N.; Lobachevsky, P. MTOR signaling orchestrates stress-induced mutagenesis, facilitating adaptive evolution in cancer. Science 2020, 368, 1127–1131. [Google Scholar] [CrossRef] [PubMed]
- Russo, M.; Crisafulli, G.; Sogari, A.; Reilly, N.M.; Arena, S.; Lamba, S.; Bartolini, A.; Amodio, V.; Magrì, A.; Novara, L. Adaptive mutability of colorectal cancers in response to targeted therapies. Science 2019, 366, 1473–1480. [Google Scholar] [CrossRef]
- Chaft, J.E.; Oxnard, G.R.; Sima, C.S.; Kris, M.G.; Miller, V.A.; Riely, G.J. Disease flare after tyrosine kinase inhibitor discontinuation in patients with EGFR-mutant lung cancer and acquired resistance to erlotinib or gefitinib: Implications for clinical trial design. Clin. Cancer Res. 2011, 17, 6298–6303. [Google Scholar] [CrossRef] [PubMed]
- Zuniga, R.M.; Torcuator, R.; Jain, R.; Anderson, J.; Doyle, T.; Schultz, L.; Mikkelsen, T. Rebound tumour progression after the cessation of bevacizumab therapy in patients with recurrent high-grade glioma. J. Neurooncol 2010, 99, 237–242. [Google Scholar] [CrossRef]
- West, H.; Oxnard, G.R.; Doebele, R.C. Acquired resistance to targeted therapies in advanced non-small cell lung cancer: New strategies and new agents. Am. Soc. Clin. Oncol. Educ. Book. 2013, 33, e272–e278. [Google Scholar] [CrossRef]
- Pop, O.; Pirvu, A.; Toffart, A.C.; Moro-Sibilot, D. Disease flare after treatment discontinuation in a patient with EML4-ALK lung cancer and acquired resistance to crizotinib. J. Thorac. Oncol. 2012, 7, e1–e2. [Google Scholar] [CrossRef]
- Yap, T.A.; Macklin-Doherty, A.; Popat, S. Continuing EGFR inhibition beyond progression in advanced non-small cell lung cancer. Eur. J. Cancer 2017, 70, 12–21. [Google Scholar] [CrossRef]
- Pupo, E.; Ducano, N.; Lupo, B.; Vigna, E.; Avanzato, D.; Perera, T.; Trusolino, L.; Lanzetti, L.; Comoglio, P.M. Rebound Effects Caused by Withdrawal of MET Kinase Inhibitor Are Quenched by a MET Therapeutic Antibody. Cancer Res. 2016, 76, 5019–5029. [Google Scholar] [CrossRef] [PubMed]
- Altintas, D.M.; Cerqua, M.; De Laurentiis, A.; Trusolino, L.; Boccaccio, C.; Comoglio, P.M. An mTOR feedback loop mediates the ‘flare’ (‘rebound’) response to MET tyrosine kinase inhibition. Sci. Rep. 2023, 13, 1378. [Google Scholar] [CrossRef]
- Guertin, D.A.; Sabatini, D.M. Defining the Role of mTOR in Cancer. Cancer Cell 2007, 12, 9–22. [Google Scholar] [CrossRef] [PubMed]
- Mamane, Y.; Petroulakis, E.; LeBacquer, O.; Sonenberg, N. mTOR, translation initiation and cancer. Oncogene 2006, 25, 6416–6422. [Google Scholar] [CrossRef]
- Huber, A.; Bodenmiller, B.; Uotila, A.; Stahl, M.; Wanka, S.; Gerrits, B.; Aebersold, R.; Loewith, R. Characterization of the rapamycin-sensitive phosphoproteome reveals that Sch9 is a central coordinator of protein synthesis. Genes. Dev. 2009, 23, 1929–1943. [Google Scholar] [CrossRef]
- Thoreen, C.C.; Chantranupong, L.; Keys, H.R.; Wang, T.; Gray, N.S.; Sabatini, D.M. A unifying model for mTORC1-mediated regulation of mRNA translation. Nature 2012, 485, 109–113. [Google Scholar] [CrossRef]
- Lazaris-Karatzas, A.; Montine, K.S.; Sonenberg, N. Malignant transformation by a eukaryotic initiation factor subunit that binds to mRNA 5’ cap. Nature 1990, 345, 544–547. [Google Scholar] [CrossRef]
- Blagosklonny, M.V. Rapalogs in cancer prevention: Anti-aging or anticancer? Cancer Biol. Ther. 2012, 13, 1349–1354. [Google Scholar] [CrossRef] [PubMed]
- Wei, K.; Li, M.; Zöller, M.; Wang, M.; Mehrabi, A.; Hoffmann, K. Targeting c-MET by Tivantinib through synergistic activation of JNK/c-jun pathway in cholangiocarcinoma. Cell Death Dis. 2019, 10, 231. [Google Scholar] [CrossRef]
- Lee, Y.J.; Han, J.-Y.; Lee, G.K.; Shin, J.; Yun, S.A.; Oh, J.Y.; Lee, S.; Kim, H.T.; Lee, J.S. C-MET overexpression as a resistance biomarker to epidermal growth factor receptor tyrosine kinase inhibitors in EGFR-mutant non-small cell lung cancer. J. Clin. Oncol. 2016, 34, e20660. [Google Scholar] [CrossRef]
- Danilkovitch-Miagkova, A.; Zbar, B. Dysregulation of Met receptor tyrosine kinase activity in invasive tumors. J. Clin. Investig. 2002, 109, 863–867. [Google Scholar] [CrossRef] [PubMed]
- Blumenschein, G.R., Jr.; Mills, G.B.; Gonzalez-Angulo, A.M. Targeting the hepatocyte growth factor–cMET axis in cancer therapy. J. Clin. Oncol. 2012, 30, 3287. [Google Scholar] [CrossRef] [PubMed]
Drug | Number of Trials (Phase) * | Cancer Types | Principal Outcome | Notes |
---|---|---|---|---|
Multitarget tyrosine kinase inhibitors | ||||
PF02341066 (Crizotinib) | 51 (early I/I); 62 (II); 18 (III); 5 (IV); 7 (NA) | Breast cancer, renal clear cell cancer, glioblastoma, inflammatory myofibroblastic tumors, lymphoma, papillary renal cancers, MET+ gastric adenocarcinoma, MET+ or RON+ metastatic urothelial cancer and NSCLC | Substantial antitumor activity in patients with MET amplification and/or METΔ14 [66,67,68]. Crizotinib overcomes resistance to selpercatinib in RET-fusion positive NSCLC patients [69]. |
|
XL184 (Cabozantinib) | 54 (early I/I); 157 (II); 19 (III); 2 (IV); 7 (NA) | Breast cancer, glioblastoma, HCC, kidney cancer, medullary thyroid cancer, melanoma, NSCLC, ovarian cancer, and prostate cancer | Cabozantinib significantly improved progression-free survival in patients with metastatic PRCC and melanoma [70,71]. Complete response was reported in one patient with METΔ14 [72]. |
|
GSK1363089 (Foretinib) | 6 (early I/I); 7 (II) | Mixed cancer, breast cancer, gastric cancer, head and neck cancer, liver cancer, NSCLC, and PRCC | No activity in unselected patients [74]. | Targets: MET, RET, and others. |
MGCD265 (Glesatinib) | 4 (early I/I); 2 (II) | Mixed cancer and NSCLC | Results are pending. The safety profile is acceptable in non-genetically selected patients with advanced solid tumors [75]. | Targets: MET and AXL |
MP470 (Amuvatinib) | 2 (early I/I); 1 (II); 1 (NA) | Mixed cancer, gastric cancer, glioblastoma, pancreatic cancer, and SCLC | Results are pending. | Targets: MET, RET, FLT3, and PDGFRA |
E7050 (Golvatinib) | 8 (early I/I) | Mixed cancer, gastric cancer, head and neck cancer, and HCC | Results are pending. | Targets: MET and VEGFR2 |
Specific MET inhibitors (small molecules) | ||||
ARQ197 (Tivantinib) | 25 (early I/I); 21 (II); 4 (III) | Mixed cancer **, colorectal cancer, HCC, liver cancer, mesothelioma, NSCLC, SCLC, and stomach cancer | Tivantinib treatment did not demonstrate efficacy in a Phase III trial for HCC patients with high MET levels (based on staining intensity) [76]. | Specificity to MET is controversial [77,78]. |
INCB28060 (Capmatinib) | 18 (early I/I); 27 (II); 3 (III); 1 (IV) | Mixed cancer, colorectal cancer, glioblastoma, head and neck cancer, HCC, NSCLC, and PRCC | Capmatinib showed substantial antitumor activity in patients with MET amplification or METΔ14 [79,80,81]. Capmatinib induces potentially similar resistance mechanisms as Crizotinib [82] but is a promising option in MET-amplified, EGFR-inhibitor-resistant tumors [80]. | Approved to treat adult patients with metastatic NSCLC with METΔ14 [83]. |
AZD6094 (Savolitinib or Volitinib) | 7 (early I/I); 8 (II); 3 (III) | Mixed cancer, colorectal cancer, gastric cancer, NSCLC, kidney cancer, and PRCC | Encouraging results in EGFR-mutated, MET-amplified tumors [64,65]. | NA |
AMG337 | 3 (early I/I); 5 (II) | Mixed cancer, renal clear cell cancer, esophageal cancer, and stomach cancer | Results are pending. | NA |
MSC2156119J (Tepotinib) | 5 (early I/I); 5 (II) | Mixed cancer, lung cancer, and NSCLC | Partial response in NSCLC patients with METΔ14 [84]. Promising results in patients with MET amplification [85,86]. | NA |
OMO-1 (JNJ-38877618) | 1 (early I/I) | Mixed cancer, lung cancer, and NSCLC | Results are pending. | NA |
MET antibodies | ||||
MetMab (Onartuzumab) | 6 (early I/I); 8 (II); 5 (III) | Mixed cancer, breast cancer, colorectal cancer, glioblastoma, HER2- and MET+ gastric cancer, HCC, and MET+ NSCLC | Some clinical trials were inconclusive due to poor patient selection or the premature termination of the study [87,88,89]. Other results are pending. | One-armed monoclonal antibody [90]. |
LY2875358 (Emibetuzumab) | 1 (early I/I); 2 (II) | Mixed cancer, gastric cancer, and NSCLC | Cytostatic antitumor activity [91]. Significant increase in median progression-free survival for patients with the highest MET expression [92]. It cannot reverse acquired resistance to Erlotinib, an EGFR inhibitor [93]. | Humanized IgG4 bivalent monoclonal antibody [94]. |
ARGX-111 | 1 (early I/I) | Mixed cancer, gastric cancer, glioblastoma, liver cancer, and renal cancer | Results are pending. | Bivalent monoclonal antibody with the property to activate ADCC [95]. |
SAIT301 | 1 (early I/I) | Mixed cancer | Phase I completed, and the recommended dose for phase II was established [96]. | Bivalent monoclonal antibodies targeted against the MET α chain, inducing CBL-independent degradation of MET to circumvent the detrimental agonist effect of other bivalent antibodies [97,98]. |
SF antibodies | ||||
AMG 102 (Rilotumumab) | 6 (early I/I); 13 (II); 3 (III) | Mixed cancer, gastric cancer, glioblastoma, lung cancer, mesothelioma, and prostate cancer | No improvement in the clinical outcome in patients with MET+ gastric cancer [99]. Toxicity issue [100]. | Humanized IgG2 monoclonal antibody [101]. |
AV-299 (Ficlatuzumab) | 6 (early I/I); 4 (II); 3 (III) | AML, head and neck cancer, liver cancer, and NSCLC | Low benefit compared to other drugs [102,103]. | Humanized IgG1 monoclonal antibody [104]. |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Altintas, D.M.; Comoglio, P.M. An Observatory for the MET Oncogene: A Guide for Targeted Therapies. Cancers 2023, 15, 4672. https://doi.org/10.3390/cancers15184672
Altintas DM, Comoglio PM. An Observatory for the MET Oncogene: A Guide for Targeted Therapies. Cancers. 2023; 15(18):4672. https://doi.org/10.3390/cancers15184672
Chicago/Turabian StyleAltintas, Dogus M., and Paolo M. Comoglio. 2023. "An Observatory for the MET Oncogene: A Guide for Targeted Therapies" Cancers 15, no. 18: 4672. https://doi.org/10.3390/cancers15184672
APA StyleAltintas, D. M., & Comoglio, P. M. (2023). An Observatory for the MET Oncogene: A Guide for Targeted Therapies. Cancers, 15(18), 4672. https://doi.org/10.3390/cancers15184672