Abstract
The c-Met (mesenchymal-epithelial transition factor) receptor tyrosine kinase, upon activation by its ligand, hepatocyte growth factor (HGF), plays an important role in regulating cellular processes such as proliferation, survival, and metastasis. Dysregulation of c-Met signaling, including overexpression, gene amplification, and mutations, is associated with the development and progression of various cancers, such as non-small cell lung cancer (NSCLC), gastric cancer, and hepatocellular carcinoma. This review explores the oncogenic mechanisms involving c-Met altered cancers, highlighting how dysregulated c-Met signaling promotes tumor growth and metastasis. This work offers a detailed analysis of c-Met expression patterns in different tissues and examines the frequency of c-Met alterations across various cancer types. Advancements in molecular diagnostics have significantly improved the clinical detection of c-Met alterations, facilitating the precise identification of patients eligible for c-Met-targeted therapies. While initial clinical trials of c-Met inhibitors demonstrated promising outcomes, the emergence of resistance remains a major hurdle. This review also covers the mechanisms contributing to resistance, such as secondary mutations, activation of alternative signaling pathways, and tumor cell phenotypic changes. Additionally, it examines current therapeutic approaches targeting c-Met, including small molecule inhibitors, monoclonal antibodies, and combination therapies designed to overcome resistance. The discussion extends to future challenges in c-Met inhibition, highlighting the need for innovative therapeutic strategies and combination regimens to enhance efficacy and mitigate resistance and thus the review emphasizes the transformative potential of c-Met-targeted therapies in advancing cancer treatment.
Similar content being viewed by others
Data availability
Not applicable.
References
Mohan CD, Shanmugam MK, Gowda SG, Chinnathambi A, Rangappa KS, Sethi G (2024) C-MET pathway in human malignancies and its targeting by natural compounds for cancer therapy. Phytomedicine 128:155379
Faiella A, Riccardi F, Cartenì G, Chiurazzi M, Onofrio L (2022) The emerging role of c-met in carcinogenesis and clinical implications as a possible therapeutic target. J Oncol 2022(1):5179182
Zhang Y, Xia M, Jin K, Wang S, Wei H, Fan C, Wu Y, Li X, Li X, Li G, Zeng Z (2018) Function of the c-Met receptor tyrosine kinase in carcinogenesis and associated therapeutic opportunities. Mol Cancer 17:1–4
Gherardi E, Birchmeier W, Birchmeier C, Vande Woude G (2012) Targeting MET in cancer: rationale and progress. Nat Rev Cancer 12(2):89–103. https://doi.org/10.1038/nrc3205
European Bioinformatics Institute ChEMBL database [Internet]. Hinxton (UK): EMBL-EBI; [cited 2025 Mar 31]. Available from: https://www.ebi.ac.uk/chembl/
Comoglio PM, Giordano S, Trusolino L (2008) Drug development of MET inhibitors: targeting oncogene addiction and expedience. Nat Rev Drug Discov 7:504–516
Organ SL, Tsao MS (2011) An overview of the c-MET signaling pathway. Ther Adv Med Oncol 3(1suppl):S7-19
Ariyawutyakorn W, Saichaemchan S, Garcia MV, Understanding and targeting MET signaling in solid tumors - are we there yet? J. Cancer. 7 633–649. doi:, Petrini ANDI (2016) Biology of MET: a double life between normal tissue repair and tumor progression, Ann. Transl. Med. 3 (2015) 82. doi: 10.3978/j.issn.2305-5839.2015.03.58
Barrow-Mcgee R, Kermorgant S (2014) Met endosomal signalling: in the right place, at the right time. Int J Biochem Cell Biol 49:69–74. https://doi.org/10.1016/j.biocel.2014.01.009
Parikh RA, Wang P, Beumer JH, Chu E, Appleman LJ (2014) The potential roles of hepatocyte growth factor (HGF)-MET pathway inhibitors in cancer treatment. Onco Targets Ther 4:969–983
Pasquini G, Giaccone G (2018) C-MET inhibitors for advanced non-small cell lung cancer. Expert Opin Investig Drugs 27(4):363–375
NCBI MET proto-oncogene, receptor tyrosine kinase [Internet]. Bethesda (MD): National Library of Medicine (US), National Center for Biotechnology Information; updated 6 Jul 2025 [cited 2025 Jul 13]. Gene ID: 4233. Available from: https://www.ncbi.nlm.nih.gov/gene/4233
Uhlén M, Fagerberg L, Hallström BM et al (2015) Tissue-based map of the human proteome. Science. ;347(6220):1260419. 10.1126/science.1260419.The Human Protein Atlas. RNA expression data in human tissues (version 24.0). Available from https://www.proteinatlas.org/humanproteome/tissue/data (accessed July 2025)
Wolf J, Hochmair M, Han JY, Reguart N, Souquet PJ, Smit EF, Orlov SV, Vansteenkiste J, Nishio M, de Jonge M, Akerley W (2024) Capmatinib in MET exon 14-mutated non-small-cell lung cancer: final results from the open-label, phase 2 GEOMETRY mono-1 trial. Lancet Oncol 25(10):1357–1370
Paik PK, Felip E, Veillon R, Sakai H, Cortot AB, Garassino MC, Mazieres J, Viteri S, Senellart H, Van Meerbeeck J, Raskin J (2020) Tepotinib in non–small-cell lung cancer with MET exon 14 skipping mutations. N Engl J Med 383(10):931–943
Zhang H, Song Y, Xia F, Liu Y, Zhang L, Yang J, Tu H, Long B, Sui J, Wang Y (2025) Adverse event profile of Crizotinib in real-world from the FAERS database: a 12-year pharmacovigilance study. BMC Pharmacol Toxicol 26(1):61
Choueiri TK, Heng DY, Lee JL, Cancel M, Verheijen RB, Mellemgaard A, Ottesen LH, Frigault MM, L’Hernault A, Szijgyarto Z, Signoretti S (2020) Efficacy of savolitinib vs sunitinib in patients with MET-driven papillary renal cell carcinoma: the SAVOIR phase 3 randomized clinical trial. JAMA Oncol 6(8):1247–1255
Choueiri TK, Vaishampayan U, Rosenberg JE, Logan TF, Harzstark AL, Bukowski RM, Rini BI, Srinivas S, Stein MN, Adams LM, Ottesen LH (2013) Phase II and biomarker study of the dual MET/VEGFR2 inhibitor foretinib in patients with papillary renal cell carcinoma. J Clin Oncol 31(2):181–186
McGregor B, Mortazavi A, Cordes L, Salabao C, Vandlik S, Apolo AB (2022) Management of adverse events associated with cabozantinib plus nivolumab in renal cell carcinoma: a review. Cancer Treat Rev 103:102333
Dong Y, Xu J, Sun B, Wang J, Wang Z. (2022). MET-targeted therapies and clinical outcomes: A systematic literature review. Molecular Diagnosis & Therapy 26:203–227.
Watermann I, Schmitt B, Stellmacher F et al (2015) Improved diagnostics targeting c-MET in non-small cell lung cancer: expression, amplification and activation? Diagn Pathol 10:130
Recondo G, Che J, Jänne PA, Awad MM (2020) Targeting MET dysregulation in cancer. Cancer Discov 10(7):922–934. https://doi.org/10.1158/2159-8290.CD-19-1446
Guo R, Berry LD, Aisner DL et al (2019) MET IHC is a poor screen for MET amplification or MET exon 14 mutations in lung adenocarcinomas: data from a tri-institutional cohort of the lung cancer mutation consortium. J Thorac Oncol 14:1666–1671
Zhou J, Zhang XC, Xue S (2023) SYK-mediated epithelial cell state is associated with response to c-Met inhibitors in c-Met-overexpressing lung cancer. Signal Transduct Target Ther 8:185
Bubendorf L, Dafni U, Schobel M et al (2017) 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 111:143–149
Sun D, Wu W, Wang L, Qu J, Han Q, Wang H, Song S, Liu N, Wang Y, Hou H (2023) Identification of MET fusions as novel therapeutic targets sensitive to MET inhibitors in lung cancer. J Transl Med 21(1):150. https://doi.org/10.1186/s12967-023-03999-7
Reis H, Metzenmacher M, Goetz M et al (2018) MET expression in advanced non-small-cell lung cancer: effect on 10 Han et al JTO clinical and research reports 5 No. clinical outcomes of chemotherapy, targeted therapy, and immunotherapy. Clin Lung Cancer 19:e441e463
Janjigian YY, Tang LH, Coit DG et al (2011) MET expression and amplification in patients with localized gastric cancer. Cancer Epidemiol Biomarkers Prev 20(5):1021–1027. https://doi.org/10.1158/1055-9965.EPI-10-1080
Lee HE, Kim MA, Lee HS et al (2012) MET in gastric carcinomas: comparison between protein expression and gene copy number and impact on clinical outcome. Br J Cancer 107(2):325–333. https://doi.org/10.1038/bjc.2012.237
Sun Y, Tian -M-M, Zhou L-X, You W-C, Li J-Y (2012) Value of c-Met for predicting progression of precancerous gastric lesions in rural Chinese population. Chin J Cancer Res 24(1):18–22. https://doi.org/10.1007/s11670-012-0018-x
Chen C, Kim H, Liska D et al (2012) MET activation mediates resistance to lapatinib inhibition of HER2-amplified gastric cancer cells. Mol Cancer Ther 11(3):660–669. https://doi.org/10.1158/1535-7163.MCT-11-0754
Kim J, Fox C, Peng S (2014) Preexisting oncogenic events impact trastuzumab sensitivity in ERBB2-amplified gastroesophageal adenocarcinoma. J Clin Invest 124(12):5145–5158. https://doi.org/10.1172/JCI75200
Zhang J, Jiang X, Jiang Y et al (2016) Recent advances in the development of dual VEGFR and c-Met small molecule inhibitors as anticancer drugs. Eur J Med Chem 108:495–504
Jo M, Stolz DB, Esplen JE, Dorko K, Michalopoulos GK, Strom SC (2000) Crosstalk between epidermal growth factor receptor and cMet signal pathways in transformed cells. J Biol Chem 275:8806–8811
Zhang Y, Xia M, Jin K et al (2018) Function of the c-Met receptor tyrosine kinase in carcinogenesis and associated therapeutic opportunities. Mol Cancer 17:45
Huang K, Sung I, Fang W et al (2018) Correlation between HGF/c-Met and Notch1 signaling pathways in human gastric cancer cells. Oncol Rep 40:294–230
Wang H, Rao B, Lou J et al (2020) The function of the HGF/c-Met axis in hepatocellular carcinoma. Front Cell Dev Biol 8:55
Zhang Z, Miao L, Wang S et al (2022) Study on the expression of c-Met in gastric cancer and its correlation with preoperative serum tumor markers and prognosis. World J Surg Oncol 20:204. https://doi.org/10.1186/s12957-022-02659-2
Lai X, Dong Q, Xu F, Wu S, Yang D, Liu C, Li Y, Li Z, Ma D (2021) Correlation of c-MET expression with clinical characteristics and the prognosis of colorectal cancer. J Gastrointest Oncol 12(5):2203–2210. https://doi.org/10.21037/jgo-21-536
Schmidt L, Duh FM, Chen F, Kishida T, Glenn G, Choyke P, Scherer SW, Zhuang Z, Lubensky I, Dean M, Allikmets R, Chidambaram A, Bergerheim UR, Feltis JT, Casadevall C, Zamarron A, Bernues M, Richard S, Lips CJ, Walther MM, Tsui LC, Geil L, Orcutt ML, Stackhouse T, Lipan J, Slife L, Brauch H, Decker J, Niehans G, Hughson MD, Moch H, Storkel S, Lerman MI, Linehan WM, Zbar B (1997) Germline and somatic mutations in the tyrosine kinase domain of the MET proto-oncogene in papillary renal carcinomas. Nat Genet 16(1):68–73. https://doi.org/10.1038/ng0597-68
Schmidt L, Junker K, Weirich G, Glenn G, Choyke P, Lubensky I, Zhuang Z, Jeffers M, Vande Woude G, Neumann H, Walther M, Linehan WM, Zbar B (1998) Two North American families with hereditary papillary renal carcinoma and identical novel mutations in the MET proto-oncogene. Cancer Res 58(8):1719–1722 PMID: 9563489
Zhuang Z, Park WS, Pack S, Schmidt L, Vortmeyer AO, Pak E, Pham T, Weil RJ, Candidus S, Lubensky IA, Linehan WM, Zbar B, Weirich G (1998) Trisomy 7-harbouring non-random duplication of the mutant MET allele in hereditary papillary renal carcinomas. Nat Genet 20(1):66–9. https://doi.org/10.1038/1727
Sebai M, Tulasne D, Caputo SM, Verkarre V, Fernandes M, Guérin C, Reinhart F, Adams S, Maugard C, Caron O, Guillaud-Bataille M, Berthet P, Bignon YJ, Bressac-de Paillerets B, Burnichon N, Chiesa J, Giraud S, Lejeune S, Limacher JM, de Pauw A, Stoppa-Lyonnet D, Zattara-Cannoni H, Deveaux S, Lidereau R, Richard S, Rouleau E (2022) Novel germline MET pathogenic variants in French patients with papillary renal cell carcinomas type I. Hum Mutat 43(3):316–327. https://doi.org/10.1002/humu.24313Epub 2022 Jan 19. PMID: 34882875
Jeffers M, Schmidt L, Nakaigawa N, Webb CP, Weirich G, Kishida T, Zbar B, Vande Woude GF (1997) Activating mutations for the Met tyrosine kinase receptor in human cancer. Proc Natl Acad Sci U S A 94(21):11445–11450. https://doi.org/10.1073/pnas.94.21.11445
Bardelli A, Longati P, Gramaglia D, Basilico C, Tamagnone L, Giordano S, Ballinari D, Michieli P, Comoglio PM (1998) Uncoupling signal transducers from oncogenic MET mutants abrogates cell transformation and inhibits invasive growth. Proc Natl Acad Sci U S A 95(24):14379–14383. https://doi.org/10.1073/pnas.95.24.14379
Michieli P, Basilico C, Pennacchietti S, Maffè A, Tamagnone L, Giordano S, Bardelli A, Comoglio PM (1999) Mutant Met-mediated transformation is ligand-dependent and can be inhibited by HGF antagonists. Oncogene 18(37):5221–31. https://doi.org/10.1038/sj.onc.1202899
Park WS et al (1999) Somatic mutations in the kinase domain of the Met/hepatocyte growth factor receptor gene in childhood hepatocellular carcinomas. Cancer Res 59(2):307–310
Hara S et al (2006) Hypoxia enhances c-Met/HGF receptor expression and signaling by activating HIF-1α in human salivary gland cancer cells. Oral Oncol 42(6):593–598. https://doi.org/10.1016/j.oraloncology.2005.10.016
Klosek SK et al (2005) CD151 forms a functional complex with c-Met in human salivary gland cancer cells. Biochemical and biophysical research communications vol. 336,2 : 408 – 16. https://doi.org/10.1016/j.bbrc.2005.08.106
Hara S, Nakashiro KI, Goda H, Hamakawa H (2008) Role of Akt isoforms in HGF-induced invasive growth of human salivary gland cancer cells. Biochem Biophys Res Commun 370(1):123–128
Kim CH, Moon SK, Bae JH, Ho Lee J, Ho Han J, Kim K, Chang Choi E (2006) Expression of hepatocyte growth factor and c-Met in hypopharyngeal squamous cell carcinoma. Acta Otolaryngol 126(1):88–94
Segovia R, Arroyo G, Ituarte C, Inklemona C, Rojo S, Salvatierra A, Berlinghieri G, Saucedo S, Monteros Alvi M, Lamas R (2019) Over-expression of c-MET in biliary tract cancer: prevalence and clinical-molecular correlation (ILOGI). Ann Oncol 30(Suppl 4):iv72 Abstract P-263
Gray MJ et al (2015) Mutations preventing regulated exon skipping in MET cause osteofibrous dysplasia. Am J Hum Genet 97(6):837–847. https://doi.org/10.1016/j.ajhg.2015.11.001
Ma PC, Kijima T, Maulik G et al (2003) c-MET mutational analysis in small cell lung cancer: novel juxtamembrane domain mutations regulating cytoskeletal functions. Cancer Res 63:6272–6281
Voortman J, Harada T, Chang RP, Killian JK, Suuriniemi M, Smith WI, Meltzer PS, Lucchi M, Wang Y, Giaccone G (2013) Detection and therapeutic implications of c-Met mutations in small cell lung cancer and neuroendocrine tumors. Curr Pharm Des 19(5):833–840
Li Y, Gao L, Ma D, Qiu T, Li W, Li W, Guo L, Xing P, Liu B, Deng L, Fu J (2018) Identification of MET exon14 skipping by targeted DNA-and RNA-based next-generation sequencing in pulmonary sarcomatoid carcinomas. Lung Cancer 122:113–119
Davies KD, Lomboy A, Lawrence CA, Yourshaw M, Bocsi GT, Camidge DR, Aisner DL (2019) DNA-based versus RNA-based detection of MET exon 14 skipping events in lung cancer. J Thorac Oncol 14(4):737–741
Jurkiewicz, M., Saqi, A., Mansukhani, M. M., et al. (2020). Efficacy of DNA versus RNA NGS-based Methods in MET Exon 14 skipping mutation detection. Journal of Clinical Oncology 38(15_suppl):9036–9036.
Farragher SM, Tanney A, Kennedy RD, Paul Harkin D (2008) RNA expression analysis from formalin fixed paraffin embedded tissues. Histochem Cell Biol 130(3):435–445
McCombie WR, McPherson JD, Mardis ER (2019) Next-generation sequencing technologies. Cold Spring Harb Perspect Med 9(11):a036798
Poirot B, Doucet L, Benhenda S, Champ J, Meignin V, Lehmann-Che J (2017) MET exon 14 alterations and new resistance mutations to tyrosine kinase inhibitors: risk of inadequate detection with current amplicon-based NGS panels. J Thorac Oncol 12(10):1582–1587
Guo R, Luo J, Chang J, Rekhtman N, Arcila M, Drilon A (2020) MET-dependent solid tumors—molecular diagnosis and targeted therapy. Nat Rev Clin Oncol 17(9):569–587
Drilon A, Wang L, Arcila ME (2015) Broad, hybrid capture-based next-generation sequencing identifies actionable genomic alterations in lung adenocarcinomas otherwise negative for such alterations by other genomic testing approaches. Clin Cancer Res 21:3631–3639
Sanger F, Coulson AR (1975) A rapid method for determining sequences in DNA by primed synthesis with DNA polymerase. J Mol Biol 94(3):441–448
Sanger F, Nicklen S, Coulson AR (1977) DNA sequencing with chain-terminating inhibitors. Proc Natl Acad Sci U S A 74(12):5463–7
Das R, Jakubowski MA, Spildener J, Cheng YW (2022) Identification of novel MET exon 14 skipping variants in non-small cell lung cancer patients: a prototype workflow involving in silico prediction and RT-PCR. Cancers 14(19):4814. https://doi.org/10.3390/cancers14194814
Levsky JM, Singer RH (2003) Fluorescence in situ hybridization: past, present and future. J Cell Sci 116(14):2833–2838
Yuan P, Xue X, Qiu T, Ying J (2024) MET alterations detection platforms and clinical implications in solid tumors: a comprehensive review of literature. Ther Adv Med Oncol 16:17588359231221910
Lee J, Kim ST, Kim K, Lee H, Kozarewa I, Mortimer PG, Odegaard JI, Harrington EA, Lee J, Lee T, Oh SY (2019) Tumor genomic profiling guides patients with metastatic gastric cancer to targeted treatment: the VIKTORY umbrella trial. Cancer Discov 9(10):1388–1405
Heydt C, Becher AK, Wagener-Ryczek S (2019) Comparison of in situ and extraction-based methods for the detection of MET amplifications in solid tumors. Comput Struct Biotechnol J 17:1339–1347
Fan Y, Sun R, Wang Z, Zhang Y, Xiao X, Liu Y, Xin B, Xiong H, Lu D, Ma J (2023) Detection of MET amplification by droplet digital PCR in peripheral blood samples of non-small cell lung cancer. J Cancer Res Clin Oncol 149(5):1667–1677
Guo R, Luo J, Chang J et al (2020) MET-dependent solid tumors - molecular diagnosis and targeted therapy. Nat Rev Clin Oncol 17:569–587
Spigel DR, Ervin TJ, Ramlau RA et al (2013) Randomized phase II trial of onartuzumab in combination with erlotinib in patients with advanced non-small-cell lung cancer. J Clin Oncol 31:4105–4114
score MS, Post-Calculating CASCO (2015) H-Score. https://www.ascopost.com/issues/april-10-2015/calculating-h-score/
Seager M, Aisner DL, Davies KD (2019) Oncogenic gene fusion detection using anchored multiplex polymerase chain reaction followed by next generation sequencing. JoVE (Journal Visualized Experiments) 5(149):e59895
Li Y, Gao L, Ma D, Qiu T, Li W, Li W, Guo L, Xing P, Liu B, Deng L, Fu J, Li J, Yu Y, Ying J (2018) Identification of MET exon14 skipping by targeted DNAand RNA-based next-generation sequencing in pulmonary sarcomatoid carcinomas. Lung Cancer. https://doi.org/10.1016/j.lungcan.2018.06.001
Song Z, Xu C, He Y, Li F, Wang W, Zhu Y, Gao Y, Ji M, Chen M, Lai J, Cheng W (2020) Simultaneous detection of gene fusions and base mutations in cancer tissue biopsies by sequencing dual nucleic acid templates in unified reaction. Clin Chem 66(1):178–187
Wei J, Rybczynska AA, Meng P, Terpstra M, Saber A, Sietzema J, Timens W, Schuuring E, Hiltermann TJ, Groen HJ, van der Wekken AJ (2020) An all-in-one transcriptome-based assay to identify therapy-guiding genomic aberrations in nonsmall cell lung cancer patients. Cancers 12(10):2843
Huang K, Sung I, Fang W et al (2018) Correlation between HGF/c-Met and Notch1 signaling pathways in human gastric cancer cells. Oncol Rep 40:294–302
Funakoshi Y, Mukohara T, Ekyalongo RC, Tomioka H, Kataoka Y, Shimono Y, Chayahara N, Toyoda M, Kiyota N, Fujiwara Y, Minami H (2013) Regulation of MET kinase inhibitor resistance by copy number of MET in gastric carcinoma cells. Oncol Res 21(6):287–293
Funakoshi Y, Mukohara T, Tomioka H, Ekyalongo RC, Kataoka Y, Inui Y, Kawamori Y, Toyoda M, Kiyota N, Fujiwara Y, Minami H (2013) Excessive MET signaling causes resistance and addiction to MET inhibitors in the MKN45 gastric cancer cell line. Investig New Drugs 31:1158–1168
Li A, Yang JJ, Zhang XC, Zhang Z, Su J, Gou LY, Bai Y, Zhou Q, Yang Z, Han-Zhang H, Zhong WZ (2017) Acquired MET Y1248H and D1246N mutations mediate resistance to MET inhibitors in non–small cell lung cancer. Clin Cancer Res 23(16):4929–4937
Deng N, Goh LK, Wang H et al (2012) A comprehensive survey of genomic alterations in gastric cancer reveals systematic patterns of molecular exclusivity and co-occurrence among distinct therapeutic targets. Gut 61:673–684
Ko J, Jung J, Kim ST, Hong JY, Park S, Park JO, Park YS, Lim HY, Ahn S, Kim KM, Kang WK (2022) MET gene alterations predict poor survival following chemotherapy in patients with advanced cancer. Pathol Oncol Res. 1610697. https://doi.org/10.3389/pore.2022.1610697
Yang M, Mandal E, Liu FX, O’Hara Jr RM, Lesher B, Sanborn RE (2024) Non-small cell lung cancer with MET amplification: review of epidemiology, associated disease characteristics, testing procedures, burden, and treatments. Front Oncol 13:1241402
Zhu C, Xu J, Li M, Zhao G, Cao H (2015) Heterogeneity of c-Met expression in Chinese gastric cancer patients. Hum Pathol 46(12):1901–1907
Bahcall M, Awad MM, Sholl LM, Wilson FH, Xu M, Wang S, Palakurthi S, Choi J, Ivanova EV, Leonardi GC, Ulrich BC (2018) Amplification of wild-type KRAS imparts resistance to Crizotinib in MET exon 14 mutant non–small cell lung cancer. Clin Cancer Res 24(23):5963–5976
Jin W, Shan B, Liu H, Zhou S, Li W, Pan J, Lin L, Hu D, Pan Y (2019) Acquired mechanism of Crizotinib resistance in NSCLC with MET exon 14 skipping. J Thorac Oncol 14(7):e137–e139
Harbinski F, Craig VJ, Sanghavi S, Jeffery D, Liu L, Sheppard KA, Wagner S, Stamm C, Buness A, Chatenay-Rivauday C, Yao Y (2012) Rescue screens with secreted proteins reveal compensatory potential of receptor tyrosine kinases in driving cancer growth. Cancer Discov 2(10):948–959
Straussman R, Morikawa T, Shee K, Barzily-Rokni M, Qian ZR, Du J, Davis A, Mongare MM, Gould J, Frederick DT, Cooper ZA (2012) Tumor micro-environment elicits innate resistance to RAF inhibitors through HGF secretion. Nature 487(7408):500–504
Wilson TR, Fridlyand J, Yan Y, Penuel E, Burton L, Chan E, Peng J, Lin E, Wang Y, Sosman J, Ribas A (2012) Widespread potential for growth-factor-driven resistance to anticancer kinase inhibitors. Nature 487(7408):505–509
Marsh T, Pietras K, McAllister SS (2013) Fibroblasts as architects of cancer pathogenesis. Biochimica et biophysica acta (BBA)-Molecular basis of disease 1832(7):1070–1078
Ahn SY, Kim J, Kim MA, Choi J, Kim WH (2017) Increased HGF expression induces resistance to c-MET tyrosine kinase inhibitors in gastric cancer. Anticancer Res 37(3):1127–1138
Cui J, Xia T, Xie D, Gao Y, Jia Z, Wei D, Wang L, Huang S, Quan M, Xie K (2016) HGF/Met and FOXM1 form a positive feedback loop and render pancreatic cancer cells resistance to Met inhibition and aggressive phenotypes. Oncogene 35(36):4708–4718
Apicella M, Giannoni E, Fiore S, Ferrari KJ, Fernández-Pérez D, Isella C, Granchi C, Minutolo F, Sottile A, Comoglio PM, Medico E (2018) Increased lactate secretion by cancer cells sustains non-cell-autonomous adaptive resistance to MET and EGFR targeted therapies. Cell Metab 28(6):848–865
Mekki MS, Mougel A, Vinchent A, Paquet C, Copin MC, Leroy C, Kherrouche Z, Bonte JP, Melnyk O, Vicogne J, Tulasne D (2018) Hypoxia leads to decreased autophosphorylation of the MET receptor but promotes its resistance to tyrosine kinase inhibitors. Oncotarget 9(43):27039
Sun X, Li CW, Wang WJ, Chen MK, Li H, Lai YJ, Hsu JL, Koller PB, Chan LC, Lee PC, Cheng FJ (2020) Inhibition of c-MET upregulates PD-L1 expression in lung adenocarcinoma. Am J Cancer Res 10(2):564
Yi M, Zheng X, Niu M et al (2022) Combination strategies with PD-1/PD-L1 blockade: current advances and future directions. Mol Cancer 21:28. https://doi.org/10.1186/s12943-021-01489-2
Lin X, Peng Z, Wang X et al (2019) Targeting autophagy potentiates antitumor activity of Met-TKIs against Met-amplified gastric cancer. Cell Death Dis 10:139. https://doi.org/10.1038/s41419-019-1314-x
Gao S, Li N, Zhang X, Chen J, Ko BC, Zhao Y (2023) An autophagy-inducing stapled peptide promotes c-MET degradation and overrides adaptive resistance to Sorafenib in c-MET + hepatocellular carcinoma. Biochem Biophys Rep 33:101412
Wang Y, Zhang H, Zhang Y, Li X, Hu X, Wang X (2020) Decorin promotes apoptosis and autophagy via suppressing c-Met in HTR-8 trophoblasts. Reproduction 159(6):669–677
Wang R, Deng Z, Zhu Z, Wang J, Yang X, Xu M, Wang X, Tang Q, Zhou Q, Wan X, Wu W (2023) Kaempferol promotes non-small cell lung cancer cell autophagy via restricting Met pathway. Phytomedicine 121:155090
Schroeder RD, Choi W, Hong DS, McConkey DJ (2017) Autophagy is required for crizotinib-induced apoptosis in MET-amplified gastric cancer cells. Oncotarget 8(31):51675
Liu Y, Liu JH, Chai K, Tashiro SI, Onodera S, Ikejima T (2013) Inhibition of c-Met promoted apoptosis, autophagy and loss of the mitochondrial transmembrane potential in oridonin-induced A549 lung cancer cells. J Pharm Pharmacol 65(11):1622–1642
Lyu P, Gu X, Wang F, Sun H, Zhou Q, Yang S, Yuan W (2024) Advances in targeting cancer-associated fibroblasts through single-cell spatial transcriptomic sequencing. Biomark Res 12(1):73
Mahrous M, Jebriel AO, Allehebi A, Shafik A, El Karak F, Venturini F, Alhusaini H, Meergans M, Sendur MA, Ouda M, Al-Nassar M (2023) Consensus recommendations for the Diagnosis, biomarker Testing, and clinical management of advanced or metastatic Non-small cell lung cancer with Mesenchymal-Epithelial transition exon 14 skipping mutations in the middle East, Africa, and Russia. Cureus 15(7):e41992.
Simard MA, Cabrera-Galvez C, Viteri S, Geist F, Reischmann N, Zühlsdorf M, Karachaliou N (2024) Spatial profiling of METex14-altered NSCLC under Tepotinib treatment: shifting the immunosuppressive landscape. Neoplasia 57:101063
Wang C, Li J, Chen J, Wang Z, Zhu G, Song L, Wu J, Li C, Qiu R, Chen X, Zhang L (2025) Multi-omics analyses reveal biological and clinical insights in recurrent stage I non-small cell lung cancer. Nat Commun 16(1):1477
Silva VR, Neves SP, Santos LD, Dias RB, Bezerra DP (2020) Challenges and therapeutic opportunities of autophagy in cancer therapy. Cancers 12(11):3461
Wang J, Wang S, Sun J, Qiu L (2021) Expression of c-MET, EGFR and HER-2 in gastric adenocarcinoma tissue and its relationship with clinicopathological characteristics. Am J Transl Res 13(9):10856
Naguib EM, Ismail EF, Badran DI et al (2024) Double trouble: how c-MET and HER2 fuel bladder cancer progression. Egypt J Med Hum Genet 25:147. https://doi.org/10.1186/s43042-024-00618-y
Bhardwaj V, Zhan Y, Cortez MA, Ang KK, Molkentine D, Munshi A, Raju U, Komaki R, Heymach JV, Welsh J (2012) C-Met inhibitor MK-8003 radiosensitizes c-Met–expressing non–small-cell lung cancer cells with radiation-induced c-Met–expression. J Thorac Oncol 7(8):1211–1217
Okuda T, Tasaki T, Nakata S, Yamashita K, Yoshioka H, Izumoto S, Kato A, Fujita M (2017) Efficacy of combination therapy with MET and VEGF inhibitors for MET-overexpressing glioblastoma. Anticancer Res 37(7):3871–3876
Jo EB, Lee YS, Lee H, Park JB, Park H, Choi YL, Hong D, Kim SJ (2019) Combination therapy with c-met inhibitor and TRAIL enhances apoptosis in dedifferentiated liposarcoma patient-derived cells. BMC Cancer 19:1–2
Huang S, Long Y, Gao Y, Lin W, Wang L, Jiang J, Yuan X, Chen Y, Zhang P, Chu Q (2024) Combined inhibition of MET and VEGF enhances therapeutic efficacy of EGFR TKIs in EGFR-mutant non-small cell lung cancer with concomitant aberrant MET activation. Exp Hematol Oncol 13(1):97
Puccini A, Marín-Ramos NI, Bergamo F, Schirripa M, Lonardi S, Lenz HJ, Loupakis F, Battaglin F (2019) Safety and tolerability of c-MET inhibitors in cancer. Drug Saf 42:211–233
Catenacci DV, Tebbutt NC, Davidenko I, Murad AM, Al-Batran SE, Ilson DH, Tjulandin S, Gotovkin E, Karaszewska B, Bondarenko I, Tejani MA (2017) 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 18(11):1467–1482
Wei WJ, Hong YL, Deng Y, Wang GL, Qiu JT, Pan F (2024) Research progress on the development of hepatocyte growth factor/c-Met signaling pathway in gastric cancer: a review. World J Gastrointest Oncol 16(8):3397
Liao H, Tian T, Sheng Y, Peng Z, Li Z, Wang J, Li Y, Zhang C, Gao J (2021) The significance of MET expression and strategies of targeting MET treatment in advanced gastric cancer. Front Oncol 11:719217
Merchant M, Ma X, Maun HR, Zheng Z, Peng J, Romero M, Huang A, Yang NY, Nishimura M, Greve J, Santell L (2013) Monovalent antibody design and mechanism of action of onartuzumab, a MET antagonist with anti-tumor activity as a therapeutic agent. Proc Natl Acad Sci U S A 110(32):E2987-96
Wakelee H, Zvirbule Z, De Braud F, Kingsley CD, Mekhail T, Lowe T, Schütte W, Lena H, Lawler W, Braiteh F, Cosgriff T (2017) Efficacy and safety of onartuzumab in combination with first-line bevacizumab-or pemetrexed-based chemotherapy regimens in advanced non-squamous non–small-cell lung cancer. Clin Lung Cancer 18(1):50–59
DiCara DM, Chirgadze DY, Pope AR, Karatt-Vellatt A, Winter A, Slavny P, Van Den Heuvel J, Parthiban K, Holland J, Packman LC, Mavria G (2017) Characterization and structural determination of a new anti-MET function-blocking antibody with binding epitope distinct from the ligand binding domain. Sci Rep 7(1):9000
Diéras V, Campone M, Yardley DA, Romieu G, Valero V, Isakoff SJ, Koeppen H, Wilson TR, Xiao Y, Shames DS, Mocci S (2015) 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 26(9):1904–1910
Liu Y, Yang Y, Ye YC, Shi QF, Chai K, Tashiro SI, Onodera S, Ikejima T (2012) Activation of ERK–p53 and ERK-mediated phosphorylation of Bcl-2 are involved in autophagic cell death induced by the c-Met inhibitor SU11274 in human lung cancer A549 cells. J Pharmacol Sci 118(4):423–432
Jia J, Moyer A, Lowe M, Bolch E, Kortmansky J, Cho M, Lenz HJ, Kalyan A, Niedzwiecki D, Strickler JH (2025) A phase 2 study of Savolitinib in patients with MET amplified metastatic colorectal cancer. J Gastrointest Cancer 56(1):1–7
Eng C, Bessudo A, Hart LL, Severtsev A, Gladkov O, Müller L, Kopp MV, Vladimirov V, Langdon R, Kotiv B, Barni S (2016) A randomized, placebo-controlled, phase 1/2 study of Tivantinib (ARQ 197) in combination with Irinotecan and cetuximab in patients with metastatic colorectal cancer with wild‐type KRAS who have received first‐line systemic therapy. Int J Cancer 139(1):177–186
ClinicalTrials.gov Lung-MAP trial: Capmatinib, Osimertinib, and Ramucirumab for NSCLC (NCT05642572). U.S. National Library of Medicine. Available from: https://clinicaltrials.gov/ct2/show/NCT05642572
Combining targeted drugs for advanced non-small cell lung cancer with EGFR mutations and MET amplification. Available from: https://www.swog.org/patients/trials-open-patients/combining-targeted-drugs-advanced-non-small-cell-lung-cancer-has-egfr
UAMS Cancer Institute NCT06031688: Expanded Lung-MAP Trial Investigating Tepotinib with or Without Ramucirumab in Advanced NSCLC. Available from: https://cancer.uams.edu/kb/nct06031688/
Memorial Sloan Kettering Cancer Center Clinical Trial 24 – 016: Investigating Targeted Therapies in Cancer Treatment. Available from: https://www.mskcc.org/cancer-care/clinical-trials/24-016
Drilon A, Rekhtman N, Arcila M, Wang L, Ni A, Albano M, Van Voorthuysen M, Somwar R, Smith RS, Montecalvo J, Plodkowski A, Ginsberg MS, Riely GJ, Rudin CM, Ladanyi M, Kris MG (2016) Cabozantinib in patients with advanced RET-rearranged non-small-cell lung cancer: an open-label, single-centre, phase 2, single-arm trial. Lancet Oncol 17(12):1653–1660 Epub 2016 Nov 4. PMID: 27825636; PMCID: PMC5143197
MyCancerGenome, NCT01639508 : Clinical trial investigating targeted therapies in cancer. Available from: https://www.mycancergenome.org/content/clinical_trials/NCT01639508/
ClinicalTrials.gov NCT03175224: APL-101 in MET-Altered Tumors. U.S. National Library of Medicine. Available from: https://clinicaltrials.gov/study/NCT03175224
National Cancer Institute NCI-2023-04100: Amivantamab in Combination with Tyrosine Kinase Inhibitors for the Treatment of Non-Small Cell Lung Cancer. Available from: https://www.cancer.gov/about-cancer/treatment/clinical-trials/search/v?id=NCI-2023-04100
National Cancer Institute NCI-2023-08689: Clinical trial investigating C-MET inhibitors in cancer treatment. Available from: https://www.cancer.gov/research/participate/clinical-trials-search/v?id=NCI-2023-08689loc=0q=c-met%20inhibitorsrl=1
Cho BC et al (2020) 1258O Amivantamab (JNJ-61186372), an EGFR-MET bispecific antibody, in combination with lazertinib, a 3rd-generation tyrosine kinase inhibitor (TKI), in advanced EGFR NSCLC. Ann Oncol 31(Suppl 4):S813
Acknowledgements
Namy George, gratefully acknowledges School of Pharmaceutical Sciences, Jaipur National University, Jaipur, India, for academic guidance during her doctoral studies.The authors would like to acknowledge the College of Pharmacy, National University of Science and Technology, Muscat, Oman, for providing the necessary research facilities. This work was supported by The Research Council (TRC) of Oman under the Graduate Research Grant (GRG) program, Grant ID-BFP/GRG/CBS/23/071.
Funding
This work was supported by the College of Pharmacy, National University of Science and Technology, Muscat, Oman. Author Bushara Al Sabahi has received research support from The Research Council (TRC), Oman under the Graduate Research Grant (GRG) program (Grant number BFP/GRG/CBS/23/071).
Author information
Authors and Affiliations
Contributions
All authors contributed to the study “Targeting c-Met Signaling in Cancer: From MET Alteration to Clinical Advances and Future Prospects”. Material preparation, data collection and analysis were performed by Namy George. The first draft of the manuscript was written by Namy George and all authors commented on previous versions of the manuscript. All authors read and approved the final manuscript.
Corresponding authors
Ethics declarations
Competing interests
The authors declare no competing interests.
Ethics approval
This review article is based exclusively on previously published literature and does not involve any studies with human participants or animals conducted by the authors. Hence, ethical approval was not required.
Consent to publish
Not applicable.
Consent to participate
Not applicable.
Additional information
Publisher’s note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Supplementary Information
Below is the link to the electronic supplementary material.
Rights and permissions
Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.
About this article
Cite this article
George, N., Bajaj, S., Al Sabahi, B. et al. Targeting mesenchymal-epithelial transition factor signaling in cancer: from genetic alterations to clinical advances and future prospects. Mol Biol Rep 52, 1041 (2025). https://doi.org/10.1007/s11033-025-11127-5
Received:
Accepted:
Published:
DOI: https://doi.org/10.1007/s11033-025-11127-5