Unveiling the Role of HGF/c-Met Signaling in Non-Small Cell Lung Cancer Tumor Microenvironment
Abstract
:1. Introduction
2. Structure and Function of HGF/c-MET Signaling
3. HGF/c-Met Signaling in NSCLC
3.1. Dysregulated HGF/c-MET Signaling in NSCLC
3.2. Role in NSCLC Development
3.3. Drug Development of MET Inhibitors
4. HGF/c-MET Signaling in Microenvironment
4.1. Promotes Angiogenesis
4.2. Effects on Tumor Metabolism
4.3. Interaction with Cancer-Associated Fibroblasts
4.4. Modulating Immune System and Anti-Tumor Immunity
5. Targeting HGF/c-Met Signaling Combined with Immunotherapy in NSCLC
5.1. The Significance of Targeting MET Combined with Immunotherapy
5.2. Current Therapeutic Strategies
6. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Acknowledgments
Conflicts of Interest
References
- Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2016. CA Cancer J. Clin. 2016, 66, 7–30. [Google Scholar] [CrossRef] [PubMed]
- Chen, Z.; Fillmore, C.M.; Hammerman, P.S.; Kim, C.F.; Wong, K.K. Non-small-cell lung cancers: A heterogeneous set of diseases. Nat. Rev. Cancer 2014, 14, 535–546. [Google Scholar] [CrossRef]
- Blume-Jensen, P.; Hunter, T. Oncogenic kinase signalling. Nature 2001, 411, 355–365. [Google Scholar] [CrossRef]
- Sadiq, A.A.; Salgia, R. MET as a possible target for non-small-cell lung cancer. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2013, 31, 1089–1096. [Google Scholar] [CrossRef] [PubMed]
- Li, Q.; Tie, Y.; Alu, A.; Ma, X.; Shi, H. Targeted therapy for head and neck cancer: Signaling pathways and clinical studies. Signal Transduct. Target. Ther. 2023, 8, 31. [Google Scholar] [CrossRef] [PubMed]
- Remon, J.; Hendriks, L.E.L.; Mountzios, G.; García-Campelo, R.; Saw, S.P.L.; Uprety, D.; Recondo, G.; Villacampa, G.; Reck, M. MET alterations in NSCLC-Current Perspectives and Future Challenges. J. Thorac. Oncol. Off. Publ. Int. Assoc. Study Lung Cancer 2023, 18, 419–435. [Google Scholar] [CrossRef] [PubMed]
- Wolf, J.; Seto, T.; Han, J.-Y.; Reguart, N.; Garon, E.B.; Groen, H.J.M.; Tan, D.S.-W.; Hida, T.; De Jonge, M.J.; 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]
- Wu, Y.L.; Guarneri, V.; Voon, P.J.; Lim, B.K.; Yang, J.J.; Wislez, M.; Huang, C.; Liam, C.K.; Mazieres, J.; Tho, L.M.; et al. Tepotinib plus osimertinib in patients with EGFR-mutated non-small-cell lung cancer with MET amplification following progression on first-line osimertinib (INSIGHT 2): A multicentre, open-label, phase 2 trial. Lancet Oncol. 2024, 25, 989–1002. [Google Scholar] [CrossRef]
- Jamme, P.; Fernandes, M.; Copin, M.C.; Descarpentries, C.; Escande, F.; Morabito, A.; Grégoire, 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. Off. Publ. Int. Assoc. Study Lung Cancer 2020, 15, 741–751. [Google Scholar] [CrossRef]
- Guo, R.; Offin, M.; Brannon, A.R.; Chang, J.; Chow, A.; Delasos, L.; Girshman, J.; Wilkins, O.; McCarthy, C.G.; Makhnin, A.; et al. MET Exon 14-altered Lung Cancers and MET Inhibitor Resistance. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2021, 27, 799–806. [Google Scholar] [CrossRef]
- Li, A.; Yang, J.J.; Zhang, X.C.; Zhang, Z.; Su, J.; Gou, L.Y.; Bai, Y.; Zhou, Q.; Yang, Z.; Han-Zhang, H.; et al. Acquired MET Y1248H and D1246N Mutations Mediate Resistance to MET Inhibitors in Non-Small Cell Lung Cancer. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2017, 23, 4929–4937. [Google Scholar] [CrossRef] [PubMed]
- Liang, X.J.; Song, X.Y.; Wu, J.L.; Liu, D.; Lin, B.Y.; Zhou, H.S.; Wang, L. Advances in Multi-Omics Study of Prognostic Biomarkers of Diffuse Large B-Cell Lymphoma. Int. J. Biol. Sci. 2022, 18, 1313–1327. [Google Scholar] [CrossRef] [PubMed]
- Khemlina, G.; Ikeda, S.; Kurzrock, R. The biology of Hepatocellular carcinoma: Implications for genomic and immune therapies. Mol. Cancer 2017, 16, 149. [Google Scholar] [CrossRef] [PubMed]
- Moreno, V.M.; Baeza, A. Bacteria as Nanoparticle Carriers for Immunotherapy in Oncology. Pharmaceutics 2022, 14, 784. [Google Scholar] [CrossRef]
- Raj, S.; Kesari, K.K.; Kumar, A.; Rathi, B.; Sharma, A.; Gupta, P.K.; Jha, S.K.; Jha, N.K.; Slama, P.; Roychoudhury, S.; et al. Molecular mechanism(s) of regulation(s) of c-MET/HGF signaling in head and neck cancer. Mol. Cancer 2022, 21, 31. [Google Scholar] [CrossRef]
- Jiang, Z.; Liang, G.; Xiao, Y.; Qin, T.; Chen, X.; Wu, E.; Ma, Q.; Wang, Z. Targeting the SLIT/ROBO pathway in tumor progression: Molecular mechanisms and therapeutic perspectives. Ther. Adv. Med. Oncol. 2019, 11, 1758835919855238. [Google Scholar] [CrossRef]
- Zhang, Y.; Xia, M.; Jin, K.; Wang, S.; Wei, H.; Fan, C.; Wu, Y.; Li, X.; Li, X.; Li, G.; et al. Function of the c-Met receptor tyrosine kinase in carcinogenesis and associated therapeutic opportunities. Mol. Cancer 2018, 17, 45. [Google Scholar] [CrossRef]
- Zhou, H.; Liu, Z.; Wang, Y.; Wen, X.; Amador, E.H.; Yuan, L.; Ran, X.; Xiong, L.; Ran, Y.; Chen, W.; et al. Colorectal liver metastasis: Molecular mechanism and interventional therapy. Signal Transduct. Target. Ther. 2022, 7, 70. [Google Scholar] [CrossRef]
- Byeon, H.K.; Ku, M.; Yang, J. Beyond EGFR inhibition: Multilateral combat strategies to stop the progression of head and neck cancer. Exp. Mol. Med. 2019, 51, 1–14. [Google Scholar] [CrossRef]
- Sagi, Z.; Hieronymus, T. The Impact of the Epithelial-Mesenchymal Transition Regulator Hepatocyte Growth Factor Receptor/Met on Skin Immunity by Modulating Langerhans Cell Migration. Front. Immunol. 2018, 9, 517. [Google Scholar] [CrossRef]
- Mizuno, S.; Nakamura, T. HGF-MET cascade, a key target for inhibiting cancer metastasis: The impact of NK4 discovery on cancer biology and therapeutics. Int. J. Mol. Sci. 2013, 14, 888–919. [Google Scholar] [CrossRef]
- Linossi, E.M.; Estevam, G.O.; Oshima, M.; Fraser, J.S.; Collisson, E.A.; Jura, N.J.B.S.T. State of the structure address on MET receptor activation by HGF. Biochem. Soc. Trans. 2021, 49, 645–661. [Google Scholar] [CrossRef] [PubMed]
- Organ, S.L.; Tsao, M.S. An overview of the c-MET signaling pathway. Ther. Adv. Med. Oncol. 2011, 3, S7–S19. [Google Scholar] [CrossRef] [PubMed]
- Xu, Y.; Fisher, G.J. Role of met axis in head and neck cancer. Cancers 2013, 5, 1601–1618. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Panganiban, R.A.; Day, R.M. Hepatocyte growth factor in lung repair and pulmonary fibrosis. Acta Pharmacol. Sin. 2011, 32, 12–20. [Google Scholar] [CrossRef]
- Srivastava, A.K.; Navas, T.; Herrick, W.G.; Hollingshead, M.G.; Bottaro, D.P.; Doroshow, J.H.; Parchment, R.E. Effective implementation of novel MET pharmacodynamic assays in translational studies. Ann. Transl. Med. 2017, 5, 3. [Google Scholar] [CrossRef]
- Wang, H.; Rao, B.; Lou, J.; Li, J.; Liu, Z.; Li, A.; Cui, G.; Ren, Z.; Yu, Z. The Function of the HGF/c-Met Axis in Hepatocellular Carcinoma. Front. Cell Dev. Biol. 2020, 8, 55. [Google Scholar] [CrossRef]
- Heldin, C.H. Dimerization of cell surface receptors in signal transduction. Cell 1995, 80, 213–223. [Google Scholar] [CrossRef]
- Tanizaki, J.; Okamoto, I.; Sakai, K.; Nakagawa, K. Differential roles of trans-phosphorylated EGFR, HER2, HER3, and RET as heterodimerisation partners of MET in lung cancer with MET amplification. Br. J. Cancer 2011, 105, 807–813. [Google Scholar] [CrossRef]
- Benvenuti, S.; Lazzari, L.; Arnesano, A.; Li Chiavi, G.; Gentile, A.; Comoglio, P.M. Ron kinase transphosphorylation sustains MET oncogene addiction. Cancer Res. 2011, 71, 1945–1955. [Google Scholar] [CrossRef] [PubMed]
- Kissil, J.L.; Charest, A. New tools for dissecting RON receptor tyrosine kinase oncogenic signaling. Cancer Biol. Ther. 2006, 5, 1187–1188. [Google Scholar] [CrossRef] [PubMed]
- Li, W.; Xiong, X.; Abdalla, A.; Alejo, S.; Zhu, L.; Lu, F.; Sun, H. HGF-induced formation of the MET-AXL-ELMO2-DOCK180 complex promotes RAC1 activation, receptor clustering, and cancer cell migration and invasion. J. Biol. Chem. 2018, 293, 15397–15418. [Google Scholar] [CrossRef]
- Dulak, A.M.; Gubish, C.T.; Stabile, L.P.; Henry, C.; Siegfried, J.M. HGF-independent potentiation of EGFR action by c-Met. Oncogene 2011, 30, 3625–3635. [Google Scholar] [CrossRef] [PubMed]
- Fischer, O.M.; Giordano, S.; Comoglio, P.M.; Ullrich, A. Reactive oxygen species mediate Met receptor transactivation by G protein-coupled receptors and the epidermal growth factor receptor in human carcinoma cells. J. Biol. Chem. 2004, 279, 28970–28978. [Google Scholar] [CrossRef] [PubMed]
- Ariyawutyakorn, W.; Saichaemchan, S.; Varella-Garcia, M. Understanding and Targeting MET Signaling in Solid Tumors—Are We There Yet? J. Cancer 2016, 7, 633–649. [Google Scholar] [CrossRef]
- Dai, L.; Trillo-Tinoco, J.; Cao, Y.; Bonstaff, K.; Doyle, L.; Del Valle, L.; Whitby, D.; Parsons, C.; Reiss, K.; Zabaleta, J.; et al. Targeting HGF/c-MET induces cell cycle arrest, DNA damage, and apoptosis for primary effusion lymphoma. Blood 2015, 126, 2821–2831. [Google Scholar] [CrossRef]
- Ma, P.C.; Tretiakova, M.S.; MacKinnon, A.C.; Ramnath, N.; Johnson, C.; Dietrich, S.; Seiwert, T.; Christensen, J.G.; Jagadeeswaran, R.; Krausz, T.; et al. Expression and mutational analysis of MET in human solid cancers. Genes Chromosomes Cancer 2008, 47, 1025–1037. [Google Scholar] [CrossRef]
- Park, S.; Choi, Y.L.; Sung, C.O.; An, J.; Seo, J.; Ahn, M.J.; Ahn, J.S.; Park, K.; Shin, Y.K.; Erkin, O.C.; et al. High MET copy number and MET overexpression: Poor outcome in non-small cell lung cancer patients. Histol. Histopathol. 2012, 27, 197–207. [Google Scholar] [CrossRef]
- Tsuta, K.; Kozu, Y.; Mimae, T.; Yoshida, A.; Kohno, T.; Sekine, I.; Tamura, T.; Asamura, H.; Furuta, K.; Tsuda, H. c-MET/phospho-MET protein expression and MET gene copy number in non-small cell lung carcinomas. J. Thorac. Oncol. Off. Publ. Int. Assoc. Study Lung Cancer 2012, 7, 331–339. [Google Scholar] [CrossRef]
- Ma, P.C.; Jagadeeswaran, R.; Jagadeesh, S.; Tretiakova, M.S.; Nallasura, V.; Fox, E.A.; Hansen, M.; Schaefer, E.; Naoki, K.; Lader, A.; et al. 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]
- Comprehensive molecular profiling of lung adenocarcinoma. Nature 2014, 511, 543–550. [CrossRef] [PubMed]
- Kalluri, R. The biology and function of fibroblasts in cancer. Nat. Rev. Cancer 2016, 16, 582–598. [Google Scholar] [CrossRef] [PubMed]
- Seo, J.S.; Ju, Y.S.; Lee, W.C.; Shin, J.Y.; Lee, J.K.; Bleazard, T.; Lee, J.; Jung, Y.J.; Kim, J.O.; Shin, J.Y.; et al. The transcriptional landscape and mutational profile of lung adenocarcinoma. Genome Res. 2012, 22, 2109–2119. [Google Scholar] [CrossRef] [PubMed]
- Onozato, R.; Kosaka, T.; Kuwano, H.; Sekido, Y.; Yatabe, Y.; Mitsudomi, T. Activation of MET by gene amplification or by splice mutations deleting the juxtamembrane domain in primary resected lung cancers. J. Thorac. Oncol. Off. Publ. Int. Assoc. Study Lung Cancer 2009, 4, 5–11. [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.; Jänne, 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. Off. J. Am. Soc. Clin. Oncol. 2016, 34, 721–730. [Google Scholar] [CrossRef]
- Schrock, A.B.; Frampton, G.M.; Suh, J.; Chalmers, Z.R.; Rosenzweig, M.; Erlich, R.L.; Halmos, B.; Goldman, J.; Forde, P.; Leuenberger, K.; et al. Characterization of 298 Patients with Lung Cancer Harboring MET Exon 14 Skipping Alterations. J. Thorac. Oncol. Off. Publ. Int. Assoc. Study Lung Cancer 2016, 11, 1493–1502. [Google Scholar] [CrossRef]
- 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]
- Drusbosky, L.M.; Dawar, R.; Rodriguez, E.; Ikpeazu, C.V. Therapeutic strategies in METex14 skipping mutated non-small cell lung cancer. J. Hematol. Oncol. 2021, 14, 129. [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.; et al. Response to MET inhibitors in patients with stage IV lung adenocarcinomas harboring MET mutations causing exon 14 skipping. Cancer Discov. 2015, 5, 842–849. [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] [PubMed]
- Drilon, A.E.; Camidge, D.R.; Ou, S.-H.I.; Clark, J.W.; Socinski, M.A.; Weiss, J.; Riely, G.J.; Winter, M.; Wang, S.C.; Monti, K.; et al. Efficacy and safety of crizotinib in patients (pts) with advanced MET exon 14-altered non-small cell lung cancer (NSCLC). J. Clin. Oncol. 2016, 34, 108. [Google Scholar] [CrossRef]
- Lee, C.; Usenko, D.; Frampton, G.M.; McMahon, C.; Ali, S.M.; Weiss, J. MET 14 Deletion in Sarcomatoid Non-Small-Cell Lung Cancer Detected by Next-Generation Sequencing and Successfully Treated with a MET Inhibitor. J. Thorac. Oncol. Off. Publ. Int. Assoc. Study Lung Cancer 2015, 10, e113–e114. [Google Scholar] [CrossRef] [PubMed]
- Waqar, S.N.; Morgensztern, D.; Sehn, J. MET Mutation Associated with Responsiveness to Crizotinib. J. Thorac. Oncol. Off. Publ. Int. Assoc. Study Lung Cancer 2015, 10, e29–e31. [Google Scholar] [CrossRef]
- Mendenhall, M.A.; Goldman, J.W. MET-Mutated NSCLC with Major Response to Crizotinib. J. Thorac. Oncol. Off. Publ. Int. Assoc. Study Lung Cancer 2015, 10, e33–e34. [Google Scholar] [CrossRef]
- Jenkins, R.W.; Oxnard, G.R.; Elkin, S.; Sullivan, E.K.; Carter, J.L.; Barbie, D.A. Response to Crizotinib in a Patient With Lung Adenocarcinoma Harboring a MET Splice Site Mutation. Clin. Lung Cancer 2015, 16, e101–e104. [Google Scholar] [CrossRef]
- Wolf, J.; Garon, E.B.; Groen, H.J.M.; Tan, D.S.W.; Gilloteau, I.; Le Mouhaer, S.; Hampe, M.; Cai, C.; Chassot-Agostinho, A.; Reynolds, M.; et al. Patient-reported outcomes in capmatinib-treated patients with METex14-mutated advanced NSCLC: Results from the GEOMETRY mono-1 study. Eur. J. Cancer 2022, 183, 98–108. [Google Scholar] [CrossRef]
- Rolfo, C.; Malapelle, U.; Russo, A. Skipping or Not Skipping? That’s the Question! An Algorithm to Classify Novel MET Exon 14 Variants in Non–Small-Cell Lung Cancer. JCO Precis. Oncol. 2023, 7, e2200674. [Google Scholar] [CrossRef]
- Davies, K.D.; Lomboy, A.; Lawrence, C.A.; Yourshaw, M.; Bocsi, G.T.; Camidge, D.R.; Aisner, D.L. DNA-based versus RNA-based detection of MET exon 14 skipping events in lung cancer. J. Thorac. Oncol. 2019, 14, 737–741. [Google Scholar] [CrossRef]
- Moore, D.A.; Benafif, S.; Poskitt, B.; Argue, S.; Lee, S.-M.; Ahmad, T.; Papadatos-Pastos, D.; Jamal-Hanjani, M.; Bennett, P.; Forster, M.D. Optimising fusion detection through sequential DNA and RNA molecular profiling of non-small cell lung cancer. Lung Cancer 2021, 161, 55–59. [Google Scholar] [CrossRef]
- Noonan, S.A.; Berry, L.; Lu, X.; Gao, D.; Barón, A.E.; Chesnut, P.; Sheren, J.; Aisner, D.L.; Merrick, D.; Doebele, R.C.; et al. Identifying the Appropriate FISH Criteria for Defining MET Copy Number-Driven Lung Adenocarcinoma through Oncogene Overlap Analysis. J. Thorac. Oncol. Off. Publ. Int. Assoc. Study Lung Cancer 2016, 11, 1293–1304. [Google Scholar] [CrossRef] [PubMed]
- Jorge, S.E.; Schulman, S.; Freed, J.A.; VanderLaan, P.A.; Rangachari, D.; Kobayashi, S.S.; Huberman, M.S.; Costa, D.B. Responses to the multitargeted MET/ALK/ROS1 inhibitor crizotinib and co-occurring mutations in lung adenocarcinomas with MET amplification or MET exon 14 skipping mutation. Lung Cancer 2015, 90, 369–374. [Google Scholar] [CrossRef] [PubMed]
- Turke, A.B.; Zejnullahu, K.; Wu, Y.L.; Song, Y.; Dias-Santagata, D.; Lifshits, E.; Toschi, L.; Rogers, A.; Mok, T.; Sequist, L.; et al. Preexistence and clonal selection of MET amplification in EGFR mutant NSCLC. Cancer Cell 2010, 17, 77–88. [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. 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]
- Sun, D.; Wu, W.; Wang, L.; Qu, J.; Han, Q.; Wang, H.; Song, S.; Liu, N.; Wang, Y.; Hou, H. Identification of MET fusions as novel therapeutic targets sensitive to MET inhibitors in lung cancer. J. Transl. Med. 2023, 21, 150. [Google Scholar] [CrossRef] [PubMed]
- Yeh, I.; Botton, T.; Talevich, E.; Shain, A.H.; Sparatta, A.J.; de la Fouchardiere, A.; Mully, T.W.; North, J.P.; Garrido, M.C.; Gagnon, A.; et al. Activating MET kinase rearrangements in melanoma and Spitz tumours. Nat. Commun. 2015, 6, 7174. [Google Scholar] [CrossRef]
- Kentsis, A.; Reed, C.; Rice, K.L.; Sanda, T.; Rodig, S.J.; Tholouli, E.; Christie, A.; Valk, P.J.; Delwel, R.; Ngo, V.; et al. Autocrine activation of the MET receptor tyrosine kinase in acute myeloid leukemia. Nat. Med. 2012, 18, 1118–1122. [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]
- Sorokin, M.; Rabushko, E.; Rozenberg, J.M.; Mohammad, T.; Seryakov, A.; Sekacheva, M.; Buzdin, A. Clinically relevant fusion oncogenes: Detection and practical implications. Ther. Adv. Med. Oncol. 2022, 14, 17588359221144108. [Google Scholar] [CrossRef]
- Kazdal, D.; Hofman, V.; Christopoulos, P.; Ilié, M.; Stenzinger, A.; Hofman, P. Fusion-positive non-small cell lung carcinoma: Biological principles, clinical practice, and diagnostic implications. Genes Chromosomes Cancer 2022, 61, 244–260. [Google Scholar] [CrossRef]
- Baldacchino, S.; Grech, G. Somatic copy number aberrations in metastatic patients: The promise of liquid biopsies. Semin. Cancer Biol. 2020, 60, 302–310. [Google Scholar] [CrossRef]
- 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]
- Fresno Vara, J.A.; Casado, E.; de Castro, J.; Cejas, P.; Belda-Iniesta, C.; González-Barón, M. PI3K/Akt signalling pathway and cancer. Cancer Treat. Rev. 2004, 30, 193–204. [Google Scholar] [CrossRef]
- Zhou, J.Y.; Chen, X.; Zhao, J.; Bao, Z.; Chen, X.; Zhang, P.; Liu, Z.F.; Zhou, J.Y. MicroRNA-34a overcomes HGF-mediated gefitinib resistance in EGFR mutant lung cancer cells partly by targeting MET. Cancer Lett. 2014, 351, 265–271. [Google Scholar] [CrossRef]
- Jiao, D.; Chen, J.; Li, Y.; Tang, X.; Wang, J.; Xu, W.; Song, J.; Li, Y.; Tao, H.; Chen, Q. miR-1-3p and miR-206 sensitizes HGF-induced gefitinib-resistant human lung cancer cells through inhibition of c-Met signalling and EMT. J. Cell. Mol. Med. 2018, 22, 3526–3536. [Google Scholar] [CrossRef] [PubMed]
- Ying, L.; Zhu, Z.; Xu, Z.; He, T.; Li, E.; Guo, Z.; Liu, F.; Jiang, C.; Wang, Q. Cancer Associated Fibroblast-Derived Hepatocyte Growth Factor Inhibits the Paclitaxel-Induced Apoptosis of Lung Cancer A549 Cells by Up-Regulating the PI3K/Akt and GRP78 Signaling on a Microfluidic Platform. PLoS ONE 2015, 10, e0129593. [Google Scholar] [CrossRef]
- Yuan, J.; Dong, X.; Yap, J.; Hu, J. The MAPK and AMPK signalings: Interplay and implication in targeted cancer therapy. J. Hematol. Oncol. 2020, 13, 113. [Google Scholar] [CrossRef] [PubMed]
- Onyido, E.K.; Sweeney, E.; Nateri, A.S. Wnt-signalling pathways and microRNAs network in carcinogenesis: Experimental and bioinformatics approaches. Mol. Cancer 2016, 15, 56. [Google Scholar] [CrossRef]
- Maharati, A.; Zanguei, A.S.; Khalili-Tanha, G.; Moghbeli, M. MicroRNAs as the critical regulators of tyrosine kinase inhibitors resistance in lung tumor cells. Cell Commun. Signal. 2022, 20, 27. [Google Scholar] [CrossRef] [PubMed]
- Wang, N.; Ma, T.; Yu, B. Targeting epigenetic regulators to overcome drug resistance in cancers. Signal Transduct. Target. Ther. 2023, 8, 69. [Google Scholar] [CrossRef] [PubMed]
- Breindel, J.L.; Haskins, J.W.; Cowell, E.P.; Zhao, M.; Nguyen, D.X.; Stern, D.F. EGF receptor activates MET through MAPK to enhance non-small cell lung carcinoma invasion and brain metastasis. Cancer Res. 2013, 73, 5053–5065. [Google Scholar] [CrossRef]
- Morgan, R.D.; Ferreras, C.; Peset, I.; Avizienyte, E.; Renehan, A.G.; Edmondson, R.J.; Murphy, A.D.; Nicum, S.; Van Brussel, T.; Clamp, A.R.; et al. c-MET/VEGFR-2 co-localisation impacts on survival following bevacizumab therapy in epithelial ovarian cancer: An exploratory biomarker study of the phase 3 ICON7 trial. BMC Med. 2022, 20, 59. [Google Scholar] [CrossRef]
- Wheeler, D.L.; Huang, S.; Kruser, T.J.; Nechrebecki, M.M.; Armstrong, E.A.; Benavente, S.; Gondi, V.; Hsu, K.T.; Harari, P.M. Mechanisms of acquired resistance to cetuximab: Role of HER (ErbB) family members. Oncogene 2008, 27, 3944–3956. [Google Scholar] [CrossRef]
- Morgillo, F.; Woo, J.K.; Kim, E.S.; Hong, W.K.; Lee, H.Y. Heterodimerization of insulin-like growth factor receptor/epidermal growth factor receptor and induction of survivin expression counteract the antitumor action of erlotinib. Cancer Res. 2006, 66, 10100–10111. [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]
- Agarwal, S.; Zerillo, C.; Kolmakova, J.; Christensen, J.G.; Harris, L.N.; Rimm, D.L.; Digiovanna, M.P.; Stern, D.F. Association of constitutively activated hepatocyte growth factor receptor (Met) with resistance to a dual EGFR/Her2 inhibitor in non-small-cell lung cancer cells. Br. J. Cancer 2009, 100, 941–949. [Google Scholar] [CrossRef] [PubMed]
- Wang, W.; Li, Q.; Takeuchi, S.; Yamada, T.; Koizumi, H.; Nakamura, T.; Matsumoto, K.; Mukaida, N.; Nishioka, Y.; Sone, S.; et al. Met kinase inhibitor E7050 reverses three different mechanisms of hepatocyte growth factor-induced tyrosine kinase inhibitor resistance in EGFR mutant lung cancer. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2012, 18, 1663–1671. [Google Scholar] [CrossRef]
- Benedettini, E.; Sholl, L.M.; Peyton, M.; Reilly, J.; Ware, C.; Davis, L.; Vena, N.; Bailey, D.; Yeap, B.Y.; Fiorentino, M.; et al. Met activation in non-small cell lung cancer is associated with de novo resistance to EGFR inhibitors and the development of brain metastasis. Am. J. Pathol. 2010, 177, 415–423. [Google Scholar] [CrossRef]
- Cui, J.J. Targeting receptor tyrosine kinase MET in cancer: Small molecule inhibitors and clinical progress. J. Med. Chem. 2014, 57, 4427–4453. [Google Scholar] [CrossRef]
- Santarpia, M.; Massafra, M.; Gebbia, V.; D’Aquino, A.; Garipoli, C.; Altavilla, G.; Rosell, R. A narrative review of MET inhibitors in non-small cell lung cancer with MET exon 14 skipping mutations. Transl. Lung Cancer Res. 2021, 10, 1536–1556. [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] [PubMed]
- Landi, L.; Chiari, R.; Tiseo, M.; D’Incà, 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. Off. J. Am. Assoc. Cancer Res. 2019, 25, 7312–7319. [Google Scholar] [CrossRef] [PubMed]
- Camidge, D.R.; Otterson, G.A.; Clark, J.W.; Ou, S.-H.I.; Weiss, J.; Ades, S.; Conte, U.; Tang, Y.; Wang, S.C.-E.; Murphy, D.; et al. Crizotinib in patients (pts) with MET-amplified non-small cell lung cancer (NSCLC): Updated safety and efficacy findings from a phase 1 trial. J. Clin. Oncol. 2018, 36, 9062. [Google Scholar] [CrossRef]
- Moro-Sibilot, D.; Cozic, N.; Pérol, M.; Mazières, 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 AcSé phase II trial. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2019, 30, 1985–1991. [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. Off. J. Am. Soc. Clin. Oncol. 2018, 36, 3101–3109. [Google Scholar] [CrossRef]
- Reckamp, K.L.; Frankel, P.H.; Ruel, N.; Mack, P.C.; Gitlitz, B.J.; Li, T.; Koczywas, M.; Gadgeel, S.M.; Cristea, M.C.; Belani, C.P.; et al. Phase II Trial of Cabozantinib Plus Erlotinib in Patients With Advanced Epidermal Growth Factor Receptor (EGFR)-Mutant Non-small Cell Lung Cancer With Progressive Disease on Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitor Therapy: A California Cancer Consortium Phase II Trial (NCI 9303). Front. Oncol. 2019, 9, 132. [Google Scholar] [CrossRef]
- Neal, J.W.; Dahlberg, S.E.; Wakelee, H.A.; Aisner, S.C.; Bowden, M.; Huang, Y.; Carbone, D.P.; Gerstner, G.J.; Lerner, R.E.; Rubin, J.L.; et al. Erlotinib, cabozantinib, or erlotinib plus cabozantinib as second-line or third-line treatment of patients with EGFR wild-type advanced non-small-cell lung cancer (ECOG-ACRIN 1512): A randomised, controlled, open-label, multicentre, phase 2 trial. Lancet Oncol. 2016, 17, 1661–1671. [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]
- Lu, S.; Fang, J.; Li, X.; Cao, L.; Zhou, J.; Guo, Q.; Liang, Z.; Cheng, Y.; Jiang, L.; Yang, N.; et al. Once-daily savolitinib in Chinese patients with pulmonary sarcomatoid carcinomas and other non-small-cell lung cancers harbouring MET exon 14 skipping alterations: A multicentre, single-arm, open-label, phase 2 study. Lancet Respir. Med. 2021, 9, 1154–1164. [Google Scholar] [CrossRef]
- Choueiri, T.K.; Plimack, E.; Arkenau, H.T.; Jonasch, E.; Heng, D.Y.C.; Powles, T.; Frigault, M.M.; Clark, E.A.; Handzel, A.A.; Gardner, H.; et al. Biomarker-Based Phase II Trial of Savolitinib in Patients With Advanced Papillary Renal Cell Cancer. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2017, 35, 2993–3001. [Google Scholar] [CrossRef]
- Lu, S.; Fang, J.; Cao, L.; Li, X.; Guo, Q.; Zhou, J.; Cheng, Y.; Jiang, L.; Chen, Y.; Zhang, H.; et al. Abstract CT031: Preliminary efficacy and safety results of savolitinib treating patients with pulmonary sarcomatoid carcinoma (PSC) and other types of non-small cell lung cancer (NSCLC) harboring MET exon 14 skipping mutations. Cancer Res. 2019, 79, CT031. [Google Scholar] [CrossRef]
- Paik, P.K.; Veillon, R.; Cortot, A.B.; Felip, E.; Sakai, H.; Mazieres, J.; Griesinger, F.; Horn, L.; Senellart, H.; Van Meerbeeck, J.P.; et al. Phase II study of tepotinib in NSCLC patients with METex14 mutations. J. Clin. Oncol. 2019, 37, 9005. [Google Scholar] [CrossRef]
- Spigel, D.R.; Edelman, M.J.; O’Byrne, K.; Paz-Ares, L.; Mocci, S.; Phan, S.; Shames, D.S.; Smith, D.; Yu, W.; Paton, V.E.; et al. Results From the Phase III Randomized Trial of Onartuzumab Plus Erlotinib Versus Erlotinib in Previously Treated Stage IIIB or IV Non-Small-Cell Lung Cancer: METLung. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2017, 35, 412–420. [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. Off. Publ. Int. Assoc. Study Lung Cancer 2020, 15, 80–90. [Google Scholar] [CrossRef] [PubMed]
- Dersarkissian, M.; Bhak, R.; Lin, H.; Li, S.; Cheng, M.; Lax, A.; Huang, H.; Duh, M.; Ou, S. P2. 01-103 Real-world treatment patterns and survival in non-small cell lung cancer patients with EGFR exon 20 insertion mutations. J. Thorac. Oncol. 2019, 14, S681. [Google Scholar] [CrossRef]
- Cho, B.C.; Ahn, M.; Kim, T.; Kim, C.; Shim, B.; Han, J.; Drilon, A.; Lena, H.; Gomez, J.; Gray, J. 1173P Early safety, tolerability, and efficacy of REGN5093 in patients (pts) with MET-altered advanced non-small cell lung cancer (aNSCLC) from a first in human (FIH) study. Ann. Oncol. 2022, 33, S1085. [Google Scholar] [CrossRef]
- Camidge, D.R.; Janku, F.; Martinez-Bueno, A.; Catenacci, D.V.; Lee, J.; Lee, S.-H.; Dowlati, A.; Rohrberg, K.S.; Navarro, A.; Moon, Y.W. Safety and preliminary clinical activity of the MET antibody mixture, Sym015 in advanced non-small cell lung cancer (NSCLC) patients with MET amplification/exon 14 deletion (MET Amp/Ex14∆). J. Clin. Oncol. 2020, 38, 9510. [Google Scholar] [CrossRef]
- Drilon, A.E.; Awad, M.M.; Gadgeel, S.M.; Villaruz, L.C.; Sabari, J.K.; Perez, J.; Daly, C.; Patel, S.; Li, S.; Seebach, F.A. A phase 1/2 study of REGN5093-M114, a METxMET antibody-drug conjugate, in patients with mesenchymal epithelial transition factor (MET)-overexpressing NSCLC. J. Clin. Oncol. 2022, 40, TPS8593. [Google Scholar] [CrossRef]
- Camidge, D.R.; Bar, J.; Horinouchi, H.; Goldman, J.W.; Moiseenko, F.V.; Filippova, E.; Cicin, I.; Bradbury, P.A.; Daaboul, N.; Tomasini, P. Telisotuzumab vedotin (Teliso-V) monotherapy in patients (pts) with previously treated c-Met–overexpressing (OE) advanced non-small cell lung cancer (NSCLC). J. Clin. Oncol. 2022, 40, 9016. [Google Scholar] [CrossRef]
- Grandal, M.M.; Havrylov, S.; Poulsen, T.T.; Koefoed, K.; Dahlman, A.; Galler, G.R.; Conrotto, P.; Collins, S.; Eriksen, K.W.; Kaufman, D.; et al. Simultaneous Targeting of Two Distinct Epitopes on MET Effectively Inhibits MET- and HGF-Driven Tumor Growth by Multiple Mechanisms. Mol. Cancer Ther. 2017, 16, 2780–2791. [Google Scholar] [CrossRef]
- Zhou, C.; Tang, K.-J.; Cho, B.C.; Liu, B.; Paz-Ares, L.; Cheng, S.; Kitazono, S.; Thiagarajan, M.; Goldman, J.W.; Sabari, J.K. Amivantamab plus chemotherapy in NSCLC with EGFR exon 20 insertions. N. Engl. J. Med. 2023, 389, 2039–2051. [Google Scholar] [CrossRef] [PubMed]
- Shah, P.D.; Huang, A.C.; Xu, X.; Orlowski, R.; Amaravadi, R.K.; Schuchter, L.M.; Zhang, P.; Tchou, J.; Matlawski, T.; Cervini, A.; et al. Phase I trial of autologous RNA-electroporated cMET-directed CAR T cells administered intravenously in patients with melanoma and breast carcinoma. Cancer Res. Commun. 2023, 3, 821–829. [Google Scholar] [CrossRef] [PubMed]
- Passaro, A.; Wang, J.; Wang, Y.; Lee, S.-H.; Melosky, B.; Shih, J.-Y.; Azuma, K.; Juan-Vidal, O.; Cobo, M.; Felip, E. Amivantamab plus chemotherapy with and without lazertinib in EGFR-mutant advanced NSCLC after disease progression on osimertinib: Primary results from the phase III MARIPOSA-2 study. Ann. Oncol. 2024, 35, 77–90. [Google Scholar] [CrossRef]
- Strickler, J.H.; Weekes, C.D.; Nemunaitis, J.; Ramanathan, R.K.; Heist, R.S.; Morgensztern, D.; Angevin, E.; Bauer, T.M.; Yue, H.; Motwani, M. First-in-human phase I, dose-escalation and-expansion study of telisotuzumab vedotin, an antibody–drug conjugate targeting c-Met, in patients with advanced solid tumors. J. Clin. Oncol. 2018, 36, 3298–3306. [Google Scholar] [CrossRef]
- Camidge, D.R.; Morgensztern, D.; Heist, R.S.; Barve, M.; Vokes, E.; Goldman, J.W.; Hong, D.S.; Bauer, T.M.; Strickler, J.H.; Angevin, E. Phase I study of 2-or 3-week dosing of telisotuzumab vedotin, an antibody–drug conjugate targeting c-Met, monotherapy in patients with advanced non–small cell lung carcinoma. Clin. Cancer Res. 2021, 27, 5781–5792. [Google Scholar] [CrossRef] [PubMed]
- Camidge, D.R.; Barlesi, F.; Goldman, J.W.; Morgensztern, D.; Heist, R.; Vokes, E.; Spira, A.; Angevin, E.; Su, W.-C.; Hong, D.S. Phase Ib study of telisotuzumab vedotin in combination with erlotinib in patients with c-met protein–expressing non–small-cell lung cancer. J. Clin. Oncol. 2023, 41, 1105–1115. [Google Scholar]
- Camidge, D.R.; Barlesi, F.; Goldman, J.W.; Morgensztern, D.; Heist, R.; Vokes, E.; Angevin, E.; Hong, D.S.; Rybkin, I.I.; Barve, M.; et al. A Phase 1b study of telisotuzumab vedotin in combination with nivolumab in patients with NSCLC. JTO Clin. Res. Rep. 2022, 3, 100262. [Google Scholar] [CrossRef]
- Valkenburg, K.C.; de Groot, A.E.; Pienta, K.J. Targeting the tumour stroma to improve cancer therapy. Nat. Rev. Clin. Oncol. 2018, 15, 366–381. [Google Scholar] [CrossRef]
- Hinshaw, D.C.; Shevde, L.A. The Tumor Microenvironment Innately Modulates Cancer Progression. Cancer Res. 2019, 79, 4557–4566. [Google Scholar] [CrossRef]
- Quail, D.F.; Joyce, J.A. Microenvironmental regulation of tumor progression and metastasis. Nat. Med. 2013, 19, 1423–1437. [Google Scholar] [CrossRef]
- Bhide, S.A.; Nutting, C.M. Recent advances in radiotherapy. BMC Med. 2010, 8, 25. [Google Scholar] [CrossRef]
- Chandra, R.A.; Keane, F.K.; Voncken, F.E.M.; Thomas, C.R., Jr. Contemporary radiotherapy: Present and future. Lancet 2021, 398, 171–184. [Google Scholar] [CrossRef] [PubMed]
- Wang, B.; Liu, W.; Liu, C.; Du, K.; Guo, Z.; Zhang, G.; Huang, Z.; Lin, S.; Cen, B.; Tian, Y.; et al. Cancer-Associated Fibroblasts Promote Radioresistance of Breast Cancer Cells via the HGF/c-Met Signaling Pathway. Int. J. Radiat. Oncol. Biol. Phys. 2023, 116, 640–654. [Google Scholar] [CrossRef]
- Bhowmick, N.A.; Neilson, E.G.; Moses, H.L. Stromal fibroblasts in cancer initiation and progression. Nature 2004, 432, 332–337. [Google Scholar] [CrossRef] [PubMed]
- Ding, S.; Merkulova-Rainon, T.; Han, Z.C.; Tobelem, G. HGF receptor up-regulation contributes to the angiogenic phenotype of human endothelial cells and promotes angiogenesis in vitro. Blood 2003, 101, 4816–4822. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed]
- Cha, S.T.; Chen, P.S.; Johansson, G.; Chu, C.Y.; Wang, M.Y.; Jeng, Y.M.; Yu, S.L.; Chen, J.S.; Chang, K.J.; Jee, S.H.; et al. MicroRNA-519c suppresses hypoxia-inducible factor-1alpha expression and tumor angiogenesis. Cancer Res. 2010, 70, 2675–2685. [Google Scholar] [CrossRef]
- Roy, R.; Yang, J.; Moses, M.A. Matrix metalloproteinases as novel biomarkers and potential therapeutic targets in human cancer. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2009, 27, 5287–5297. [Google Scholar] [CrossRef]
- Qian, F.; Engst, S.; Yamaguchi, K.; Yu, P.; Won, K.A.; Mock, L.; Lou, T.; Tan, J.; Li, C.; Tam, D.; et al. Inhibition of tumor cell growth, invasion, and metastasis by EXEL-2880 (XL880, GSK1363089), a novel inhibitor of HGF and VEGF receptor tyrosine kinases. Cancer Res. 2009, 69, 8009–8016. [Google Scholar] [CrossRef]
- Crawford, Y.; Ferrara, N. Tumor and stromal pathways mediating refractoriness/resistance to anti-angiogenic therapies. Trends Pharmacol. Sci. 2009, 30, 624–630. [Google Scholar] [CrossRef]
- Cascone, T.; Xu, L.; Lin, H.Y.; Liu, W.; Tran, H.T.; Liu, Y.; Howells, K.; Haddad, V.; Hanrahan, E.; Nilsson, M.B.; et al. The HGF/c-MET Pathway Is a Driver and Biomarker of VEGFR-inhibitor Resistance and Vascular Remodeling in Non-Small Cell Lung Cancer. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2017, 23, 5489–5501. [Google Scholar] [CrossRef] [PubMed]
- Huang, J.; Sun, W.; Wang, Z.; Lv, C.; Zhang, T.; Zhang, D.; Dong, W.; Shao, L.; He, L.; Ji, X.; et al. FTO suppresses glycolysis and growth of papillary thyroid cancer via decreasing stability of APOE mRNA in an N6-methyladenosine-dependent manner. J. Exp. Clin. Cancer Res. 2022, 41, 42. [Google Scholar] [CrossRef]
- Boschert, V.; Klenk, N.; Abt, A.; Janaki Raman, S.; Fischer, M.; Brands, R.C.; Seher, A.; Linz, C.; Müller-Richter, U.D.A.; Bischler, T.; et al. The Influence of Met Receptor Level on HGF-Induced Glycolytic Reprogramming in Head and Neck Squamous Cell Carcinoma. Int. J. Mol. Sci. 2020, 21, 471. [Google Scholar] [CrossRef] [PubMed]
- De Rosa, V.; Iommelli, F.; Monti, M.; Fonti, R.; Votta, G.; Stoppelli, M.P.; Del Vecchio, S. Reversal of Warburg Effect and Reactivation of Oxidative Phosphorylation by Differential Inhibition of EGFR Signaling Pathways in Non-Small Cell Lung Cancer. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2015, 21, 5110–5120. [Google Scholar] [CrossRef]
- Kanaji, N.; Yokohira, M.; Nakano-Narusawa, Y.; Watanabe, N.; Imaida, K.; Kadowaki, N.; Bandoh, S. Hepatocyte growth factor produced in lung fibroblasts enhances non-small cell lung cancer cell survival and tumor progression. Respir. Res. 2017, 18, 118. [Google Scholar] [CrossRef] [PubMed]
- Oliveres, H.; Pineda, E.; Maurel, J. MET inhibitors in cancer: Pitfalls and challenges. Expert Opin. Investig. Drugs 2020, 29, 73–85. [Google Scholar] [CrossRef]
- Yan, B.; Jiang, Z.; Cheng, L.; Chen, K.; Zhou, C.; Sun, L.; Qian, W.; Li, J.; Cao, J.; Xu, Q.; et al. Paracrine HGF/c-MET enhances the stem cell-like potential and glycolysis of pancreatic cancer cells via activation of YAP/HIF-1α. Exp. Cell Res. 2018, 371, 63–71. [Google Scholar] [CrossRef]
- Ilangumaran, S.; Villalobos-Hernandez, A.; Bobbala, D.; Ramanathan, S. The hepatocyte growth factor (HGF)-MET receptor tyrosine kinase signaling pathway: Diverse roles in modulating immune cell functions. Cytokine 2016, 82, 125–139. [Google Scholar] [CrossRef]
- Shen, Z.; Xue, W.; Zheng, Y.; Geng, Q.; Wang, L.; Fan, Z.; Wang, W.; Yue, Y.; Zhai, Y.; Li, L.; et al. Molecular mechanism study of HGF/c-MET pathway activation and immune regulation for a tumor diagnosis model. Cancer Cell Int. 2021, 21, 374. [Google Scholar] [CrossRef]
- Thompson, J.; Dolcet, X.; Hilton, M.; Tolcos, M.; Davies, A.M. HGF promotes survival and growth of maturing sympathetic neurons by PI-3 kinase- and MAP kinase-dependent mechanisms. Mol. Cell. Neurosci. 2004, 27, 441–452. [Google Scholar] [CrossRef]
- Yamaura, K.; Ito, K.; Tsukioka, K.; Wada, Y.; Makiuchi, A.; Sakaguchi, M.; Akashima, T.; Fujimori, M.; Sawa, Y.; Morishita, R.; et al. Suppression of acute and chronic rejection by hepatocyte growth factor in a murine model of cardiac transplantation: Induction of tolerance and prevention of cardiac allograft vasculopathy. Circulation 2004, 110, 1650–1657. [Google Scholar] [CrossRef] [PubMed]
- Pitt, J.M.; Marabelle, A.; Eggermont, A.; Soria, J.C.; Kroemer, G.; Zitvogel, L. Targeting the tumor microenvironment: Removing obstruction to anticancer immune responses and immunotherapy. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2016, 27, 1482–1492. [Google Scholar] [CrossRef] [PubMed]
- Gajewski, T.F.; Schreiber, H.; Fu, Y.X. Innate and adaptive immune cells in the tumor microenvironment. Nat. Immunol. 2013, 14, 1014–1022. [Google Scholar] [CrossRef] [PubMed]
- Maimela, N.R.; Liu, S.; Zhang, Y. Fates of CD8+ T cells in Tumor Microenvironment. Comput. Struct. Biotechnol. J. 2019, 17, 1–13. [Google Scholar] [CrossRef]
- Golstein, P.; Griffiths, G.M. An early history of T cell-mediated cytotoxicity. Nat. Rev. Immunol. 2018, 18, 527–535. [Google Scholar] [CrossRef]
- Schag, K.; Schmidt, S.M.; Müller, M.R.; Weinschenk, T.; Appel, S.; Weck, M.M.; Grünebach, F.; Stevanovic, S.; Rammensee, H.G.; Brossart, P. Identification of C-met oncogene as a broadly expressed tumor-associated antigen recognized by cytotoxic T-lymphocytes. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2004, 10, 3658–3666. [Google Scholar] [CrossRef]
- Kumai, T.; Matsuda, Y.; Ohkuri, T.; Oikawa, K.; Ishibashi, K.; Aoki, N.; Kimura, S.; Harabuchi, Y.; Celis, E.; Kobayashi, H. c-Met is a novel tumor associated antigen for T-cell based immunotherapy against NK/T cell lymphoma. Oncoimmunology 2015, 4, e976077. [Google Scholar] [CrossRef]
- Benkhoucha, M.; Molnarfi, N.; Kaya, G.; Belnoue, E.; Bjarnadóttir, K.; Dietrich, P.Y.; Walker, P.R.; Martinvalet, D.; Derouazi, M.; Lalive, P.H. Identification of a novel population of highly cytotoxic c-Met-expressing CD8(+) T lymphocytes. EMBO Rep. 2017, 18, 1545–1558. [Google Scholar] [CrossRef]
- Benkhoucha, M.; Tran, N.L.; Senoner, I.; Breville, G.; Fritah, H.; Migliorini, D.; Dutoit, V.; Lalive, P.H. c-Met(+) Cytotoxic T Lymphocytes Exhibit Enhanced Cytotoxicity in Mice and Humans In Vitro Tumor Models. Biomedicines 2023, 11, 3123. [Google Scholar] [CrossRef]
- Chen, Q.; Yan, M.; Lin, H.; Lai, J.; Yang, Z.; Hu, D.; Deng, Y.; Shi, S.; Shuai, L.; Zhang, L.; et al. Hepatocyte growth factor-mediated apoptosis mechanisms of cytotoxic CD8(+) T cells in normal and cirrhotic livers. Cell Death Discov. 2023, 9, 13. [Google Scholar] [CrossRef]
- Benkhoucha, M.; Molnarfi, N.; Schneiter, G.; Walker, P.R.; Lalive, P.H. The neurotrophic hepatocyte growth factor attenuates CD8+ cytotoxic T-lymphocyte activity. J. Neuroinflamm. 2013, 10, 154. [Google Scholar] [CrossRef]
- Fischer, K.; Hoffmann, P.; Voelkl, S.; Meidenbauer, N.; Ammer, J.; Edinger, M.; Gottfried, E.; Schwarz, S.; Rothe, G.; Hoves, S.; et al. Inhibitory effect of tumor cell-derived lactic acid on human T cells. Blood 2007, 109, 3812–3819. [Google Scholar] [CrossRef] [PubMed]
- Glodde, N.; Bald, T.; van den Boorn-Konijnenberg, D.; Nakamura, K.; O’Donnell, J.S.; Szczepanski, S.; Brandes, M.; Eickhoff, S.; Das, I.; Shridhar, N.; et al. Reactive Neutrophil Responses Dependent on the Receptor Tyrosine Kinase c-MET Limit Cancer Immunotherapy. Immunity 2017, 47, 789–802.e9. [Google Scholar] [CrossRef] [PubMed]
- Tie, Y.; Tang, F.; Wei, Y.Q.; Wei, X.W. Immunosuppressive cells in cancer: Mechanisms and potential therapeutic targets. J. Hematol. Oncol. 2022, 15, 61. [Google Scholar] [CrossRef] [PubMed]
- Grenier, A.; Chollet-Martin, S.; Crestani, B.; Delarche, C.; El Benna, J.; Boutten, A.; Andrieu, V.; Durand, G.; Gougerot-Pocidalo, M.A.; Aubier, M.; et al. Presence of a mobilizable intracellular pool of hepatocyte growth factor in human polymorphonuclear neutrophils. Blood 2002, 99, 2997–3004. [Google Scholar] [CrossRef]
- He, M.; Peng, A.; Huang, X.Z.; Shi, D.C.; Wang, J.C.; Zhao, Q.; Lin, H.; Kuang, D.M.; Ke, P.F.; Lao, X.M. Peritumoral stromal neutrophils are essential for c-Met-elicited metastasis in human hepatocellular carcinoma. Oncoimmunology 2016, 5, e1219828. [Google Scholar] [CrossRef]
- Wislez, M.; Rabbe, N.; Marchal, J.; Milleron, B.; Crestani, B.; Mayaud, C.; Antoine, M.; Soler, P.; Cadranel, J. Hepatocyte growth factor production by neutrophils infiltrating bronchioloalveolar subtype pulmonary adenocarcinoma: Role in tumor progression and death. Cancer Res. 2003, 63, 1405–1412. [Google Scholar]
- Jing, W.; Wang, G.; Cui, Z.; Li, X.; Zeng, S.; Jiang, X.; Li, W.; Han, B.; Xing, N.; Zhao, Y.; et al. Tumor-neutrophil cross talk orchestrates the tumor microenvironment to determine the bladder cancer progression. Proc. Natl. Acad. Sci. USA 2024, 121, e2312855121. [Google Scholar] [CrossRef]
- Finisguerra, V.; Di Conza, G.; Di Matteo, M.; Serneels, J.; Costa, S.; Thompson, A.A.; Wauters, E.; Walmsley, S.; Prenen, H.; Granot, Z.; et al. MET is required for the recruitment of anti-tumoural neutrophils. Nature 2015, 522, 349–353. [Google Scholar] [CrossRef]
- Liu, Y.; Smith, M.R.; Wang, Y.; D’Agostino, R., Jr.; Ruiz, J.; Lycan, T.; Kucera, G.L.; Miller, L.D.; Li, W.; Chan, M.D.; et al. c-Met Mediated Cytokine Network Promotes Brain Metastasis of Breast Cancer by Remodeling Neutrophil Activities. Cancers 2023, 15, 2626. [Google Scholar] [CrossRef]
- Wu, B.; Shi, X.; Jiang, M.; Liu, H. Cross-talk between cancer stem cells and immune cells: Potential therapeutic targets in the tumor immune microenvironment. Mol. Cancer 2023, 22, 38. [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]
- Chen, P.-M.; Liu, K.-J.; Hsu, P.-J.; Wei, C.-F.; Bai, C.-H.; Ho, L.-J.; Sytwu, H.-K.; Yen, B.L. Induction of immunomodulatory monocytes by human mesenchymal stem cell-derived hepatocyte growth factor through ERK1/2. J. Leukoc. Biol. 2014, 96, 295–303. [Google Scholar] [CrossRef] [PubMed]
- Krug, S.; Abbassi, R.; Griesmann, H.; Sipos, B.; Wiese, D.; Rexin, P.; Blank, A.; Perren, A.; Haybaeck, J.; Hüttelmaier, S.; et al. Therapeutic targeting of tumor-associated macrophages in pancreatic neuroendocrine tumors. Int. J. Cancer 2018, 143, 1806–1816. [Google Scholar] [CrossRef]
- Choi, W.; Lee, J.; Lee, J.; Lee, S.H.; Kim, S. Hepatocyte Growth Factor Regulates Macrophage Transition to the M2 Phenotype and Promotes Murine Skeletal Muscle Regeneration. Front. Physiol. 2019, 10, 914. [Google Scholar] [CrossRef]
- Dong, N.; Shi, X.; Wang, S.; Gao, Y.; Kuang, Z.; Xie, Q.; Li, Y.; Deng, H.; Wu, Y.; Li, M.; et al. M2 macrophages mediate sorafenib resistance by secreting HGF in a feed-forward manner in hepatocellular carcinoma. Br. J. Cancer 2019, 121, 22–33. [Google Scholar] [CrossRef]
- Wang, Q.W.; Sun, L.H.; Zhang, Y.; Wang, Z.; Zhao, Z.; Wang, Z.L.; Wang, K.Y.; Li, G.Z.; Xu, J.B.; Ren, C.Y.; et al. MET overexpression contributes to STAT4-PD-L1 signaling activation associated with tumor-associated, macrophages-mediated immunosuppression in primary glioblastomas. J. Immunother. Cancer 2021, 9, e002451. [Google Scholar] [CrossRef]
- Lu, Z.; Chang, W.; Meng, S.; Xu, X.; Xie, J.; Guo, F.; Yang, Y.; Qiu, H.; Liu, L. Mesenchymal stem cells induce dendritic cell immune tolerance via paracrine hepatocyte growth factor to alleviate acute lung injury. Stem Cell Res. Ther. 2019, 10, 372. [Google Scholar] [CrossRef]
- Okunishi, K.; Dohi, M.; Nakagome, K.; Tanaka, R.; Mizuno, S.; Matsumoto, K.; Miyazaki, J.; Nakamura, T.; Yamamoto, K. A novel role of hepatocyte growth factor as an immune regulator through suppressing dendritic cell function. J. Immunol. 2005, 175, 4745–4753. [Google Scholar] [CrossRef]
- Singhal, E.; Sen, P. Hepatocyte growth factor-induced c-Src-phosphatidylinositol 3-kinase-AKT-mammalian target of rapamycin pathway inhibits dendritic cell activation by blocking IκB kinase activity. Int. J. Biochem. Cell Biol. 2011, 43, 1134–1146. [Google Scholar] [CrossRef]
- Singhal, E.; Kumar, P.; Sen, P. A novel role for Bruton’s tyrosine kinase in hepatocyte growth factor-mediated immunoregulation of dendritic cells. J. Biol. Chem. 2011, 286, 32054–32063. [Google Scholar] [CrossRef]
- Rutella, S.; Bonanno, G.; Procoli, A.; Mariotti, A.; de Ritis, D.G.; Curti, A.; Danese, S.; Pessina, G.; Pandolfi, S.; Natoni, F.; et al. Hepatocyte growth factor favors monocyte differentiation into regulatory interleukin (IL)-10++IL-12low/neg accessory cells with dendritic-cell features. Blood 2006, 108, 218–227. [Google Scholar] [CrossRef] [PubMed]
- Ludewig, B.; Graf, D.; Gelderblom, H.R.; Becker, Y.; Kroczek, R.A.; Pauli, G. Spontaneous apoptosis of dendritic cells is efficiently inhibited by TRAP (CD40-ligand) and TNF-alpha, but strongly enhanced by interleukin-10. Eur. J. Immunol. 1995, 25, 1943–1950. [Google Scholar] [CrossRef] [PubMed]
- Yue, F.Y.; Dummer, R.; Geertsen, R.; Hofbauer, G.; Laine, E.; Manolio, S.; Burg, G. Interleukin-10 is a growth factor for human melanoma cells and down-regulates HLA class-I, HLA class-II and ICAM-1 molecules. Int. J. Cancer 1997, 71, 630–637. [Google Scholar] [CrossRef]
- Youn, J.I.; Collazo, M.; Shalova, I.N.; Biswas, S.K.; Gabrilovich, D.I. Characterization of the nature of granulocytic myeloid-derived suppressor cells in tumor-bearing mice. J. Leukoc. Biol. 2012, 91, 167–181. [Google Scholar] [CrossRef] [PubMed]
- Yen, B.L.; Yen, M.L.; Hsu, P.J.; Liu, K.J.; Wang, C.J.; Bai, C.H.; Sytwu, H.K. Multipotent human mesenchymal stromal cells mediate expansion of myeloid-derived suppressor cells via hepatocyte growth factor/c-met and STAT3. Stem Cell Rep. 2013, 1, 139–151. [Google Scholar] [CrossRef]
- Gabrilovich, D.I.; Nagaraj, S. Myeloid-derived suppressor cells as regulators of the immune system. Nat. Rev. Immunol. 2009, 9, 162–174. [Google Scholar] [CrossRef]
- Liu, Q.; Wang, Y.; Zheng, Q.; Dong, X.; Xie, Z.; Panayi, A.; Bai, X.; Li, Z. MicroRNA-150 inhibits myeloid-derived suppressor cells proliferation and function through negative regulation of ARG-1 in sepsis. Life Sci. 2021, 278, 119626. [Google Scholar] [CrossRef]
- Tanaka, A.; Sakaguchi, S. Regulatory T cells in cancer immunotherapy. Cell Res. 2017, 27, 109–118. [Google Scholar] [CrossRef]
- Chen, Q.H.; Wu, F.; Liu, L.; Chen, H.B.; Zheng, R.Q.; Wang, H.L.; Yu, L.N. Mesenchymal stem cells regulate the Th17/Treg cell balance partly through hepatocyte growth factor in vitro. Stem Cell Res. Ther. 2020, 11, 91. [Google Scholar] [CrossRef]
- Benkhoucha, M.; Santiago-Raber, M.L.; Schneiter, G.; Chofflon, M.; Funakoshi, H.; Nakamura, T.; Lalive, P.H. Hepatocyte growth factor inhibits CNS autoimmunity by inducing tolerogenic dendritic cells and CD25+Foxp3+ regulatory T cells. Proc. Natl. Acad. Sci. USA 2010, 107, 6424–6429. [Google Scholar] [CrossRef] [PubMed]
- Munn, D.H.; Mellor, A.L. IDO in the Tumor Microenvironment: Inflammation, Counter-Regulation, and Tolerance. Trends Immunol. 2016, 37, 193–207. [Google Scholar] [CrossRef]
- Bonanno, G.; Mariotti, A.; Procoli, A.; Folgiero, V.; Natale, D.; De Rosa, L.; Majolino, I.; Novarese, L.; Rocci, A.; Gambella, M.; et al. Indoleamine 2,3-dioxygenase 1 (IDO1) activity correlates with immune system abnormalities in multiple myeloma. J. Transl. Med. 2012, 10, 247. [Google Scholar] [CrossRef] [PubMed]
- Sun, Y.; Guo, J.; Yu, L.; Guo, T.; Wang, J.; Wang, X.; Chen, Y. PD-L1(+) exosomes from bone marrow-derived cells of tumor-bearing mice inhibit antitumor immunity. Cell. Mol. Immunol. 2021, 18, 2402–2409. [Google Scholar] [CrossRef] [PubMed]
- Fehrenbacher, L.; Spira, A.; Ballinger, M.; Kowanetz, M.; Vansteenkiste, J.; Mazieres, J.; Park, K.; Smith, D.; Artal-Cortes, A.; Lewanski, C. Atezolizumab versus docetaxel for patients with previously treated non-small-cell lung cancer (POPLAR): A multicentre, open-label, phase 2 randomised controlled trial. Lancet 2016, 387, 1837–1846. [Google Scholar] [CrossRef] [PubMed]
- Saigi, M.; Alburquerque-Bejar, J.J.; Mc Leer-Florin, A.; Pereira, C.; Pros, E.; Romero, O.A.; Baixeras, N.; Esteve-Codina, A.; Nadal, E.; Brambilla, E.; et al. MET-Oncogenic and JAK2-Inactivating Alterations Are Independent Factors That Affect Regulation of PD-L1 Expression in Lung Cancer. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2018, 24, 4579–4587. [Google Scholar] [CrossRef]
- Ahn, H.K.; Kim, S.; Kwon, D.; Koh, J.; Kim, Y.A.; Kim, K.; Chung, D.H.; Jeon, Y.K. MET Receptor Tyrosine Kinase Regulates the Expression of Co-Stimulatory and Co-Inhibitory Molecules in Tumor Cells and Contributes to PD-L1-Mediated Suppression of Immune Cell Function. Int. J. Mol. Sci. 2019, 20, 4287. [Google Scholar] [CrossRef]
- Peng, S.; Wang, R.; Zhang, X.; Ma, Y.; Zhong, L.; Li, K.; Nishiyama, A.; Arai, S.; Yano, S.; Wang, W. EGFR-TKI resistance promotes immune escape in lung cancer via increased PD-L1 expression. Mol. Cancer 2019, 18, 165. [Google Scholar] [CrossRef]
- Minne, R.L.; Luo, N.Y.; Traynor, A.M.; Huang, M.; DeTullio, L.; Godden, J.; Stoppler, M.; Kimple, R.J.; Baschnagel, A.M. Genomic and Immune Landscape Comparison of MET Exon 14 Skipping and MET-Amplified Non-small Cell Lung Cancer. Clin. Lung Cancer in press. 2024. [Google Scholar] [CrossRef]
- Zhang, Z.S.; Yang, R.H.; Yao, X.; Cheng, Y.Y.; Shi, H.X.; Yao, C.Y.; Gao, Z.X.; Qi, D.F.; Zhang, W.K.; Dou, Y.Y.; et al. HGF/c-MET pathway contributes to cisplatin-mediated PD-L1 expression in hepatocellular carcinoma. Cell Biol. Int. 2021, 45, 2521–2533. [Google Scholar] [CrossRef]
- Xu, R.; Liu, X.; Li, A.; Song, L.; Liang, J.; Gao, J.; Tang, X. c-Met up-regulates the expression of PD-L1 through MAPK/NF-κBp65 pathway. J. Mol. Med. 2022, 100, 585–598. [Google Scholar] [CrossRef] [PubMed]
- Balan, M.; Mier y Teran, E.; Waaga-Gasser, A.M.; Gasser, M.; Choueiri, T.K.; Freeman, G.; Pal, S. Novel roles of c-Met in the survival of renal cancer cells through the regulation of HO-1 and PD-L1 expression. J. Biol. Chem. 2015, 290, 8110–8120. [Google Scholar] [CrossRef] [PubMed]
- Li, E.; Huang, X.; Zhang, G.; Liang, T. Combinational blockade of MET and PD-L1 improves pancreatic cancer immunotherapeutic efficacy. J. Exp. Clin. Cancer Res. 2021, 40, 279. [Google Scholar] [CrossRef]
- Yoshida, R.; Saigi, M.; Tani, T.; Springer, B.F.; Shibata, H.; Kitajima, S.; Mahadevan, N.R.; Campisi, M.; Kim, W.; Kobayashi, Y.; et al. MET-Induced CD73 Restrains STING-Mediated Immunogenicity of EGFR-Mutant Lung Cancer. Cancer Res. 2022, 82, 4079–4092. [Google Scholar] [CrossRef]
- Stagg, J.; Divisekera, U.; Duret, H.; Sparwasser, T.; Teng, M.W.; Darcy, P.K.; Smyth, M.J. CD73-deficient mice have increased antitumor immunity and are resistant to experimental metastasis. Cancer Res. 2011, 71, 2892–2900. [Google Scholar] [CrossRef]
- Vigano, S.; Alatzoglou, D.; Irving, M.; Ménétrier-Caux, C.; Caux, C.; Romero, P.; Coukos, G. Targeting Adenosine in Cancer Immunotherapy to Enhance T-Cell Function. Front. Immunol. 2019, 10, 925. [Google Scholar] [CrossRef] [PubMed]
- Zhang, X.; Cheng, C.; Hou, J.; Qi, X.; Wang, X.; Han, P.; Yang, X. Distinct contribution of PD-L1 suppression by spatial expression of PD-L1 on tumor and non-tumor cells. Cell. Mol. Immunol. 2019, 16, 392–400. [Google Scholar] [CrossRef] [PubMed]
- Sun, X.; Li, C.W.; Wang, W.J.; Chen, M.K.; Li, H.; Lai, Y.J.; Hsu, J.L.; Koller, P.B.; Chan, L.C.; Lee, P.C.; et al. Inhibition of c-MET upregulates PD-L1 expression in lung adenocarcinoma. Am. J. Cancer Res. 2020, 10, 564–571. [Google Scholar]
- Li, H.; Li, C.W.; Li, X.; Ding, Q.; Guo, L.; Liu, S.; Liu, C.; Lai, C.C.; Hsu, J.M.; Dong, Q.; et al. MET Inhibitors Promote Liver Tumor Evasion of the Immune Response by Stabilizing PDL1. Gastroenterology 2019, 156, 1849–1861.e13. [Google Scholar] [CrossRef]
- Yoshimura, K.; Inoue, Y.; Tsuchiya, K.; Karayama, M.; Yamada, H.; Iwashita, Y.; Kawase, A.; Tanahashi, M.; Ogawa, H.; Inui, N.; et al. Elucidation of the relationships of MET protein expression and gene copy number status with PD-L1 expression and the immune microenvironment in non-small cell lung cancer. Lung Cancer 2020, 141, 21–31. [Google Scholar] [CrossRef]
- Sabari, J.K.; Leonardi, G.C.; Shu, C.A.; Umeton, R.; Montecalvo, J.; Ni, A.; Chen, R.; Dienstag, J.; Mrad, C.; Bergagnini, I.; et al. PD-L1 expression, tumor mutational burden, and response to immunotherapy in patients with MET exon 14 altered lung cancers. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2018, 29, 2085–2091. [Google Scholar] [CrossRef] [PubMed]
- Iwai, Y.; Hamanishi, J.; Chamoto, K.; Honjo, T. Cancer immunotherapies targeting the PD-1 signaling pathway. J. Biomed. Sci. 2017, 24, 26. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Y.; Yang, Q.; Zeng, X.; Wang, M.; Dong, S.; Yang, B.; Tu, X.; Wei, T.; Xie, W.; Zhang, C.; et al. MET Amplification Attenuates Lung Tumor Response to Immunotherapy by Inhibiting STING. Cancer Discov. 2021, 11, 2726–2737. [Google Scholar] [CrossRef] [PubMed]
- Lindner, A.K.; Pichler, M.; Thurnher, M.; Pichler, R. Targeting c-Met to Improve Immune Checkpoint Inhibition in Metastatic Renal Cell Carcinoma. Eur. Urol. 2022, 81, 1–2. [Google Scholar] [CrossRef]
- Sun, Z.J.; Wu, Y.; Hou, W.H.; Wang, Y.X.; Yuan, Q.Y.; Wang, H.J.; Yu, M. A novel bispecific c-MET/PD-1 antibody with therapeutic potential in solid cancer. Oncotarget 2017, 8, 29067–29079. [Google Scholar] [CrossRef]
- Li, J.F.; Niu, Y.Y.; Xing, Y.L.; Liu, F. A novel bispecific c-MET/CTLA-4 antibody targetting lung cancer stem cell-like cells with therapeutic potential in human non-small-cell lung cancer. Biosci. Rep. 2019, 39, BSR20171278. [Google Scholar] [CrossRef]
- Huang, L.; Xie, K.; Li, H.; Wang, R.; Xu, X.; Chen, K.; Gu, H.; Fang, J. Suppression of c-Met-Overexpressing Tumors by a Novel c-Met/CD3 Bispecific Antibody. Drug Des. Dev. Ther. 2020, 14, 3201–3214. [Google Scholar] [CrossRef]
- Hou, W.; Yuan, Q.; Yuan, X.; Wang, Y.; Mo, W.; Wang, H.; Yu, M. A novel tetravalent bispecific antibody targeting programmed death 1 and tyrosine-protein kinase Met for treatment of gastric cancer. Investig. New Drugs 2019, 37, 876–889. [Google Scholar] [CrossRef]
- Zheng, P.P.; Kros, J.M.; Li, J. Approved CAR T cell therapies: ICE bucket challenges on glaring safety risks and long-term impacts. Drug Discov. Today 2018, 23, 1175–1182. [Google Scholar] [CrossRef]
- Min, J.; Long, C.; Zhang, L.; Duan, J.; Fan, H.; Chu, F.; Li, Z. c-Met specific CAR-T cells as a targeted therapy for non-small cell lung cancer cell A549. Bioengineered 2022, 13, 9216–9232. [Google Scholar] [CrossRef]
- Thayaparan, T.; Petrovic, R.M.; Achkova, D.Y.; Zabinski, T.; Davies, D.M.; Klampatsa, A.; Parente-Pereira, A.C.; Whilding, L.M.; van der Stegen, S.J.; Woodman, N.; et al. CAR T-cell immunotherapy of MET-expressing malignant mesothelioma. Oncoimmunology 2017, 6, e1363137. [Google Scholar] [CrossRef] [PubMed]
- Yuan, X.; Sun, Z.; Yuan, Q.; Hou, W.; Liang, Q.; Wang, Y.; Mo, W.; Wang, H.; Yu, M. Dual-function chimeric antigen receptor T cells targeting c-Met and PD-1 exhibit potent anti-tumor efficacy in solid tumors. Investig. New Drugs 2021, 39, 34–51. [Google Scholar] [CrossRef] [PubMed]
- Jiang, W.; Li, T.; Guo, J.; Wang, J.; Jia, L.; Shi, X.; Yang, T.; Jiao, R.; Wei, X.; Feng, Z.; et al. Bispecific c-Met/PD-L1 CAR-T Cells Have Enhanced Therapeutic Effects on Hepatocellular Carcinoma. Front. Oncol. 2021, 11, 546586. [Google Scholar] [CrossRef]
- Kang, C.H.; Kim, Y.; Lee, D.Y.; Choi, S.U.; Lee, H.K.; Park, C.H. c-Met-Specific Chimeric Antigen Receptor T Cells Demonstrate Anti-Tumor Effect in c-Met Positive Gastric Cancer. Cancers 2021, 13, 5738. [Google Scholar] [CrossRef] [PubMed]
- Mori, J.I.; Adachi, K.; Sakoda, Y.; Sasaki, T.; Goto, S.; Matsumoto, H.; Nagashima, Y.; Matsuyama, H.; Tamada, K. Anti-tumor efficacy of human anti-c-met CAR-T cells against papillary renal cell carcinoma in an orthotopic model. Cancer Sci. 2021, 112, 1417–1428. [Google Scholar] [CrossRef]
- Huo, Q.; Lv, J.; Zhang, J.; Huang, H.; Hu, H.; Zhao, Y.; Zhang, X.; Wang, Y.; Zhou, Y.; Qiu, J.; et al. c-Met is a chimeric antigen receptor T-cell target for treating recurrent nasopharyngeal carcinoma. Cytotherapy 2023, 25, 1037–1047. [Google Scholar] [CrossRef]
- Chen, C.; Gu, Y.M.; Zhang, F.; Zhang, Z.C.; Zhang, Y.T.; He, Y.D.; Wang, L.; Zhou, N.; Tang, F.T.; Liu, H.J.; et al. Construction of PD1/CD28 chimeric-switch receptor enhances anti-tumor ability of c-Met CAR-T in gastric cancer. Oncoimmunology 2021, 10, 1901434. [Google Scholar] [CrossRef]
- Chiriaco, C.; Donini, C.; Cortese, M.; Ughetto, S.; Modica, C.; Martinelli, I.; Proment, A.; Vitali, L.; Fontani, L.; Casucci, M.; et al. Efficacy of CAR-T immunotherapy in MET overexpressing tumors not eligible for anti-MET targeted therapy. J. Exp. Clin. Cancer Res. 2022, 41, 309. [Google Scholar] [CrossRef]
- Tchou, J.; Zhao, Y.; Levine, B.L.; Zhang, P.J.; Davis, M.M.; Melenhorst, J.J.; Kulikovskaya, I.; Brennan, A.L.; Liu, X.; Lacey, S.F.; et al. Safety and Efficacy of Intratumoral Injections of Chimeric Antigen Receptor (CAR) T Cells in Metastatic Breast Cancer. Cancer Immunol. Res. 2017, 5, 1152–1161. [Google Scholar] [CrossRef]
- Peng, Y.; Zhang, W.; Chen, Y.; Zhang, L.; Shen, H.; Wang, Z.; Tian, S.; Yang, X.; Cui, D.; He, Y.; et al. Engineering c-Met-CAR NK-92 cells as a promising therapeutic candidate for lung adenocarcinoma. Pharmacol. Res. 2023, 188, 106656. [Google Scholar] [CrossRef]
- Chiawpanit, C.; Wathikthinnakorn, M.; Sawasdee, N.; Phanthaphol, N.; Sujjitjoon, J.; Junking, M.; Yamabhai, M.; Panaampon, J.; Yenchitsomanus, P.T.; Panya, A. Precision immunotherapy for cholangiocarcinoma: Pioneering the use of human-derived anti-cMET single chain variable fragment in anti-cMET chimeric antigen receptor (CAR) NK cells. Int. Immunopharmacol. 2024, 136, 112273. [Google Scholar] [CrossRef]
- Liu, B.; Liu, Z.Z.; Zhou, M.L.; Lin, J.W.; Chen, X.M.; Li, Z.; Gao, W.B.; Yu, Z.D.; Liu, T. Development of c-MET-specific chimeric antigen receptor-engineered natural killer cells with cytotoxic effects on human liver cancer HepG2 cells. Mol. Med. Rep. 2019, 20, 2823–2831. [Google Scholar] [CrossRef] [PubMed]
Drug | Trial | Phase | Treatment | Population | Methodological Platforms | N. of Patients | ORR | PFS | DOR | Ref. |
---|---|---|---|---|---|---|---|---|---|---|
Crizotinib | PROFILE 1001 NCT00585195 | I | Crizotinib 250 mg b.i.d. | MET exon 14 skipping | NGS/RT-PCR | 69 | 32% | 7.3 months (95% CI, 5.4–9.1) | 9.1 months (95% CI, 6.4–12.7) | [91] |
METROS NCT02499614 | II | Crizotinib 250 mg b.i.d. | MET exon 14 skipping | Sanger direct sequencing | 26 | 27% | 4.4 months (95% CI, 3.0–5.8) | 3.7 months (95% CI, 1.1–6.3) | [92] | |
NCT00585195 | I | Crizotinib 250 mg b.i.d. | MET amplification | NGS/RT-PCR | Low: 3 Medium: 14 High: 20 | 33% 14.3% 40% | 1.8 months (95% CI, 0.8–14.0) 1.9 months (95% CI, 1.3–5.5) 6.7 months (95% CI, 3.4–7.4) | 12.1 months (95% CI, 12.1–12.1) 3.7 months (95% CI, 3.7–3.7) 5.5 months (95% CI, 3.3–25.8) | [93] | |
AcSé (NCT02034981) | II | Crizotinib 250 mg b.i.d. | c-MET ≥ 6 copies all c-MET-mutations | IHC/FISH/NGS | 25 28 | 16% 10.7% | 3.2 months (95% CI, 1.9–3.7) 2.4 months (95% CI, 1.6–5.9) | Not reported | [94] | |
Capmatinib | GEOMETRY mono-1 NCT02414139 | II | Capmatinib 400 mg b.i.d. | MET exon 14 skipping MET exon 14 skipping MET amplification MET amplification | FISH/RT-PCR | 28 69 69 15 | 68% 41% 29% 40% | 12.4 months (95% CI, 8.2–NR) 5.4 months (95% CI, 4.2–7.0) 4.1 months (95% CI, 2.9–4.8) 4.2 months (95% CI, 1.4–6.90) | 12.6 months (95% CI, 5.6–NR) 9.7 months (95% CI, 5.6–13.0) 8.3 months (95% CI, 4.2–15.4) 7.5 months (95% CI, 2.6–14.3) | [7] |
NCT01610336 | II | Gefitinib 250 mg daily Capmatinib 400 mg b.i.d. | MET amplification copy number < 4 | IHC/FISH | 41 | 12% | Copy number ≥ 6 5.49 months (95% CI: 4.21–7.29) IHC: +3 5.45 months (95% CI: 3.71–7.10) | Not reported | [95] | |
Copy number ≥ 4 < 6 Copy number ≥ 6 IHC: 0 IHC: +1 IHC: +2 IHC: +3 | 18 36 4 2 16 78 | 22% 47% 25% 0% 19% 32% | ||||||||
Cabozantinib | NCT01866410 | II | Cabozantinib 40 mg daily + erlotinib 150 mg daily | Advanced NSCLC with EGFR mutation and progressive disease on EGFR TKI (no MET mutations) | FISH | 37 | 10.8% | 3.6 months (95% CI, 2.0–5.6) | Not reported | [96] |
NCT01708954 | II | Arm A: erlotinib 150 mg daily Arm B: cabozantinib 60 mg daily Arm C: erlotinib 150 mg + cabozantinib 40 mg | Previously treated advanced NSCLC(MET mutations not evaluated) | Not evaluated | 38 38 35 | 3% 11% 3% | 1.8 months (95% CI 1.7–2.2) 4.3 months (95% CI 3.6–7.4) 4.7 months (95% CI 2.4–7.4) | Not reported | [97] | |
Savolitinib | TATTON NCT02143466 | Ib | Savolitinib 300 mg or 600 mg + Osimertinib 80 mg/daily | MET amplification(Post-1st/2nd-generation EGFR TKI T790M-) | FISH | 51 | 65% | 9.0 months (95% CI: 5.5–11.9) | 9.0 months (95% CI: 6.1–22.7 | [98] |
Savolitinib 300 mg + Osimertinib 80 mg/daily | MET amplification Post-1st/2nd-generation EGFR TKI T790M+ Post-3rd-generation EGFR TKI Post-1st/2nd-generation EGFR TKI T790M- | 18 69 36 | 67% 21% 23% | 11.0 months (95% CI: 4.0–NR) 5.4 months (95% CI: 4.1–8.0) 9.1 months (95% CI: 5.4–12.9) | 12.4 months (95% CI: 2.8–NR) 7.9 months (95% CI: 4.0–10.5) 8.0 months (95% CI: 4.5–NR) 8.0 months (95% CI: 4.5–NR) | |||||
NCT02897479 | II | Savolitinib 600 mg for BW ≥ 50 kgor 400 mg for BW < 50 kg | MET exon 14 skipping | NGS | 61 | 49.2% | 6.9 months | Not reported | [99] | |
NCT02127710 | II | Savolitinib 600 mg/daily | MET kinase domain mutant/amplified | FISH/NGS | 41 | 18% | 6.2 months (95% CI: 4.1–7.0) | Range: 2.4–16.4 months | [100] | |
MET kinase domain mutant/amplified | 65 | 0% | 1.4 months (95% CI: 1.4–2.7) | |||||||
NCT02897479 | II | Savolitinib 600 mg/day | MET exon 14 skipping | NGS | 34 | 38.7% | Not reported | 34 weeks (range, 16–96) | [101] | |
Tepotinib | VISION NCT02864992 | II | Tepotinib 500 mg/day | MET exon 14 skipping | NGS | 87 | BIRC: Liquid biopsy: 50% tissue biopsy: 45.1% | BIRC: liquid biopsy: 9.5 months (95% CI, 6.7–NR) tissue biopsy: 10.8 months (95% CI, 6.9–NR) | Not reported | [102] |
Inv: liquid biopsy: 55.3% tissue biopsy: 54.9% BIRC: | Inv: liquid biopsy: 9.5 months (95% CI, 5.3–21.1) tissue biopsy: 12.2 months (95% CI, 6.3–NR) | |||||||||
Foretinib | NCT00726323 | II | Foretinib | MET mutation MET amplification Chromosome 7 polysomy | IHC/FISH | 74 | 13.5% | 9.3 months (95% CI: 6.9–12.9) | 18.5 months |
Drug | Trial | Combination | Phase | Population | Methodological Platforms | Status * | Result | Ref. |
---|---|---|---|---|---|---|---|---|
Sym-015 | NCT02648724 | No | I/II | MET exon 14 skipping or MET amplification | Not evaluated | Completed | ORR 25% DCR 80% PFS 5.5 months (95% CI, 3.5–9.7) | [110] |
Amivantamab | NCT04538664 | Pemetrexed+Carboplatin | III | Advanced/Metastatic NSCLC Exon20 ins EGFR | Not evaluated | Active/Not recruiting | Superior efficacy of the combination versus chemotherapy alone (median PFS: 11.4 vs. 6.7 months; ORR: 73% vs. 47%) | [111] |
NCT04487080 | Lazertinib | III | Advanced/Metastatic NSCLC Exon19 del or Exon 21 L858R EGFR | Not evaluated | Active/Not recruiting | Higher toxicity of the combination vs. monotherapy (≥Grade 3 AEs: 75% vs. 43%); Superior efficacy of the combination versus monotherapy (median PFS: 23.7 vs. 16.6 months) | [112] | |
NCT04988295 | Lazertinib+Pemetrexed+Carboplatin or Pemetrexed+Carboplatin | III | Advanced/Metastatic Non-squamous NSCLC Exon19 del or Exon 21 L858R EGFR; progressed on/after Osimertinib | Not evaluated | Active/Not recruiting | Higher toxicity of the Ami+Laze+chemo vs. Ami+chemo vs. chemo (≥Grade 3 AEs: 92% vs. 72% vs. 48%); Superior efficacy of the Ami+Laze+chemo or Ami+chemo versus chemo (median PFS: 8.3 vs. 6.3 vs. 4.2 months) | [113] | |
Teliso-V | NCT05513703 | No | II | Advanced/Metastatic Non-Squamous NSCLC MET gene amplification | FISH | Active/Not recruiting | Not reported | |
NCT04928846 | No | II | Previously Treated Non-Squamous NSCLC MET overexpression | IHC | Recruiting | Not reported | ||
NCT02099058 | None or Erlotinib or Nivolumab or Osimertinb | I | Advanced NSCLC | IHC | Active/Not recruiting | Safe and tolerated as monotherapy; antitumor activity in MET-positive patients Acceptable toxicity in combination with Erlotinib; encouraging antitumor activity in EGFR TKI pretreated/EGFR-mutated/MET-positive patients Tolerated in combination with Nivolumab; limited antitumor activity in MET-positive patients | [114,115,116,117] | |
NCT06093503 | Osimertinib | III | Advanced/metastatic non-squamous NSCLC MET overexpression | IHC | Not yet recruiting | N/A | ||
REGN5093-M114 | NCT04982224 | None or Cemiplimab | I/II | Advanced NSCLC MET overexpression | IHC | Recruiting | Not reported |
Trial | MET TKI | ICI | Phase | Population | Status * | Result |
---|---|---|---|---|---|---|
NCT05782361 | Tepotinib | Pembrolizumab | I | Advanced cancer/ NSCLC MET ex14 skipping positive | Recruiting | Not reported |
NCT04139317 | Capmatinib | Pembrolizumab | II | Advanced/metastatic NSCLC PD-L1 ≥ 50% | Terminated | Lack of tolerability of the combination |
NCT04323436 | Capmatinib | Spartalizumab | II | Advanced/metastatic NSCLC MET ex14 skipping positive | Terminated | Lack of tolerability of the combination |
NCT03647488 | Capmatinib | Spartalizumab | II | Advanced/metastatic NSCLC | Completed | The study was not opened in the randomized part 67% disease/clinical progression 27% Adverse events |
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. |
© 2024 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
Yao, S.; Liu, X.; Feng, Y.; Li, Y.; Xiao, X.; Han, Y.; Xia, S. Unveiling the Role of HGF/c-Met Signaling in Non-Small Cell Lung Cancer Tumor Microenvironment. Int. J. Mol. Sci. 2024, 25, 9101. https://doi.org/10.3390/ijms25169101
Yao S, Liu X, Feng Y, Li Y, Xiao X, Han Y, Xia S. Unveiling the Role of HGF/c-Met Signaling in Non-Small Cell Lung Cancer Tumor Microenvironment. International Journal of Molecular Sciences. 2024; 25(16):9101. https://doi.org/10.3390/ijms25169101
Chicago/Turabian StyleYao, Shuxi, Xinyue Liu, Yanqi Feng, Yiming Li, Xiangtian Xiao, Yuelin Han, and Shu Xia. 2024. "Unveiling the Role of HGF/c-Met Signaling in Non-Small Cell Lung Cancer Tumor Microenvironment" International Journal of Molecular Sciences 25, no. 16: 9101. https://doi.org/10.3390/ijms25169101