Generic placeholder image

Current Enzyme Inhibition

Editor-in-Chief

ISSN (Print): 1573-4080
ISSN (Online): 1875-6662

Review Article

HGF/c-MET: A Potential Target for the Treatment of Various Cancers

Author(s): Kuttiappan Anitha, Kamal Dua, Dinesh Kumar Chellappan, Gaurav Gupta, Sachin Kumar Singh, Sabapathi Mohana Lakshmi and Shvetank Bhatt*

Volume 19, Issue 2, 2023

Published on: 04 May, 2023

Page: [71 - 80] Pages: 10

DOI: 10.2174/1573408019666230227101036

Price: $65

Abstract

Background: Cancer is the abnormal growth of cells in the body due to an imbalance in the normal apoptotic pathways. The abnormality in the cancer cells makes them malignant. Various types of treatment, including chemotherapy, radiation therapy, targeted therapy, and immunotherapy (IMT), are used for cancer. Mesenchymal-epithelial transition factor (c-Met) belongs to the tyrosine kinase receptor family and is overexpressed in various types of cancers. c-Met is a proto-oncogene and facilitates a wide range of biological functions, including cell proliferation, growth, migration, invasion, and angiogenesis, through interaction with its sole ligand hepatocyte growth factor (HGF). Currently, various c-mesenchymal-epithelial transition (c-MET) inhibitors and antibodies are in human trials for their anti-cancer activity.

Introduction: The c-MET is a kinase receptor for hepatocyte growth factor (HGF). It is wellrecognized for its tumorigenic potential. HGF binding with c-Met leads to c-Met dimerization and c- Met phosphorylation, which in turn activates many intracellular signalling pathways, including ERK1/2, MAPK, STAT3, Rac1, and PI3K/AKT. These pathways regulate the proliferation, invasion, and migration of cancer cells. Upon binding of HGF to c-MET, a series of phosphorylation reactions get started, which leads to transcription and translation of various proteins, followed by abnormal growth of cancerous tissues due to dysregulation of the cell cycle. The HGF/c-MET signalling pathways have shown their potential in the development of many cancers, including gastric cancer (GC). Several clinical trials have evaluated the therapeutic benefits of MET-targeted therapies involving various agents, such as anti-MET antibodies, anti-HGF antibodies, and tyrosine kinase inhibitors (TKIs). Various c-MET inhibitors are in clinical trials. The current review is focussed on the critical role of the HGF/c-MET pathways in the progression of various cancers, including GC. In addition, this review will also focus on the combination potential of c-MET inhibitors with immuno-oncology drugs, such as programmed cell death protein 1 (PD-1) inhibitors, cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) inhibitors, tumor necrosis factor (TNF) receptor (OX-40) agonists, etc.

Materials and Methods: The method adopted for the study was primarily based on the secondary search through a systematic review of the literature.

Results: A study of recent literature and various preclinical and human trials suggests the effectiveness of c-MET inhibitors (alone or in combination) in different cancer types, including GC. In this paper, the results have been elaborated as to how many papers/manuscripts/publications on the topic are present, how many drugs are in Phase I/II/ III/RCT, etc., and that how many papers report on the clinical outcomes of which agent/drug (mentioned in percentage).

Conclusion: The use of c-MET inhibitors and antibodies has emerged as a latent therapeutic approach for the treatment of various types of cancer. The c-MET inhibitors can also be used in combination with various immunotherapeutic drugs, like PD-1 inhibitors, OX-40 agonists, etc.

Keywords: c-MET, HGF, kinase, PD-1 inhibitors, tumor, immunotherapy, TNF.

Graphical Abstract
[1]
Basilico C, Hultberg A, Blanchetot C, et al. Four individually druggable MET hotspots mediate HGF-driven tumor progression. J Clin Invest 2014; 124(7): 3172-86.
[http://dx.doi.org/10.1172/JCI72316] [PMID: 24865428]
[2]
Abounader R, Laterra J. Scatter factor/hepatocyte growth factor in brain tumor growth and angiogenesis. Neuro-oncol 2005; 7(4): 436-51.
[http://dx.doi.org/10.1215/S1152851705000050] [PMID: 16212809]
[3]
Abella JV, Peschard P, Naujokas MA, et al. Met/Hepatocyte growth factor receptor ubiquitination suppresses transformation and is required for Hrs phosphorylation. Mol Cell Biol 2005; 25(21): 9632-45.
[http://dx.doi.org/10.1128/MCB.25.21.9632-9645.2005] [PMID: 16227611]
[4]
Boccaccio C, Comoglio PM. Invasive growth: a MET-driven genetic programme for cancer and stem cells. Nat Rev Cancer 2006; 6(8): 637-45.
[http://dx.doi.org/10.1038/nrc1912] [PMID: 16862193]
[5]
Giordano S, Ponzetto C, Renzo MFD, Cooper CS, Comoglio PM. Tyrosine kinase receptor indistinguishable from the c-met protein. Nature 1989; 339(6220): 155-6.
[http://dx.doi.org/10.1038/339155a0] [PMID: 2541345]
[6]
Jiang WG, Martin TA, Parr C, Davies G, Matsumoto K, Nakamura T. Hepatocyte growth factor, its receptor, and their potential value in cancer therapies. Crit Rev Oncol Hematol 2005; 53(1): 35-69.
[http://dx.doi.org/10.1016/j.critrevonc.2004.09.004] [PMID: 15607934]
[7]
Corso S, Giordano S. Cell-autonomous and non-cell-autonomous mechanisms of HGF/MET-driven resistance to targeted therapies: from basic research to a clinical perspective. Cancer Discov 2013; 3(9): 978-92.
[http://dx.doi.org/10.1158/2159-8290.CD-13-0040] [PMID: 23901039]
[8]
Lokker NA, Mark MR, Luis EA, et al. Structure-function analysis of hepatocyte growth factor: identification of variants that lack mitogenic activity yet retain high affinity receptor binding. EMBO J 1992; 11(7): 2503-10.
[http://dx.doi.org/10.1002/j.1460-2075.1992.tb05315.x] [PMID: 1321034]
[9]
Jeffers M, Rong S, Anver M, Vande Woude GF. Autocrine hepatocyte growth factor/scatter factor-Met signaling induces transformation and the invasive/metastastic phenotype in C127 cells. Oncogene 1996; 13(4): 853-6.
[PMID: 8761307]
[10]
Trusolino L, Comoglio PM. Scatter-factor and semaphorin receptors: cell signalling for invasive growth. Nat Rev Cancer 2002; 2(4): 289-300.
[http://dx.doi.org/10.1038/nrc779] [PMID: 12001990]
[11]
Nakamura T, Sakai K, Nakamura T, Matsumoto K. Hepatocyte growth factor twenty years on: Much more than a growth factor. J Gastroenterol Hepatol 2011; 26(1) (Suppl. 1): 188-202.
[http://dx.doi.org/10.1111/j.1440-1746.2010.06549.x] [PMID: 21199531]
[12]
Zhu GH, Huang C, Qiu ZJ, et al. Expression and prognostic significance of CD151, c-Met, and integrin alpha3/alpha6 in pancreatic ductal adenocarcinoma. Dig Dis Sci 2011; 56(4): 1090-8.
[http://dx.doi.org/10.1007/s10620-010-1416-x] [PMID: 20927591]
[13]
Aparicio IM, Garcia-Marin LJ, Andreolotti AG, Bodega G, Jensen RT, Bragado MJ. Hepatocyte growth factor activates several transduction pathways in rat pancreatic acini. Biochim Biophys Acta Mol Cell Res 2003; 1643(1-3): 37-46.
[http://dx.doi.org/10.1016/j.bbamcr.2003.08.007] [PMID: 14654226]
[14]
Trusolino L, Bertotti A, Comoglio PM. MET signalling: principles and functions in development, organ regeneration and cancer. Nat Rev Mol Cell Biol 2010; 11(12): 834-48.
[http://dx.doi.org/10.1038/nrm3012] [PMID: 21102609]
[15]
Mellado-Gil J, Rosa TC, Demirci C, et al. Disruption of hepatocyte growth factor/c-Met signaling enhances pancreatic beta-cell death and accelerates the onset of diabetes. Diabetes 2011; 60(2): 525-36.
[http://dx.doi.org/10.2337/db09-1305] [PMID: 20980460]
[16]
Demirci C, Ernst S, Alvarez-Perez JC, et al. Loss of HGF/c-Met signaling in pancreatic β-cells leads to incomplete maternal β-cell adaptation and gestational diabetes mellitus. Diabetes 2012; 61(5): 1143-52.
[http://dx.doi.org/10.2337/db11-1154] [PMID: 22427375]
[17]
Sennino B, Ishiguro-Oonuma T, Schriver BJ, Christensen JG, McDonald DM. Inhibition of c-Met reduces lymphatic metastasis in RIP-Tag2 transgenic mice. Cancer Res 2013; 73(12): 3692-703.
[http://dx.doi.org/10.1158/0008-5472.CAN-12-2160] [PMID: 23576559]
[18]
Birchmeier C, Birchmeier W, Gherardi E, Vande Woude GF. Met, metastasis, motility and more. Nat Rev Mol Cell Biol 2003; 4(12): 915-25.
[http://dx.doi.org/10.1038/nrm1261] [PMID: 14685170]
[19]
Lutterbach B, Zeng Q, Davis LJ, et al. Lung cancer cell lines harboring MET gene amplification are dependent on Met for growth and survival. Cancer Res 2007; 67(5): 2081-8.
[http://dx.doi.org/10.1158/0008-5472.CAN-06-3495] [PMID: 17332337]
[20]
Smolen GA, Sordella R, Muir B, et al. Amplification of MET may identify a subset of cancers with extreme sensitivity to the selective tyrosine kinase inhibitor PHA-665752. Proc Natl Acad Sci 2006; 103(7): 2316-21.
[http://dx.doi.org/10.1073/pnas.0508776103] [PMID: 16461907]
[21]
Park CH, Cho SY, Ha JD, et al. Novel c-MET inhibitor suppresses the growth of c-MET-addicted gastric cancer cells. BMC Cancer 2016; 22: 16-35.
[http://dx.doi.org/10.1186/s12885-016-2058-y]
[22]
Titmarsh HF, O’Connor R, Dhaliwal K, Akram AR. The emerging role of the c-MET-HGF axis in non-small cell lung cancer tumor immunology and immunotherapy. Front Oncol 2020; 10: 54.
[http://dx.doi.org/10.3389/fonc.2020.00054] [PMID: 32117721]
[23]
Sharpe AH, Pauken KE. The diverse functions of the PD1 inhibitory pathway. Nat Rev Immunol 2018; 18(3): 153-67.
[http://dx.doi.org/10.1038/nri.2017.108] [PMID: 28990585]
[24]
Yuan X, Sun Z, Yuan Q, et al. Dual-function chimeric antigen receptor T cells targeting c-Met and PD-1 exhibit potent anti-tumor efficacy in solid tumors. Invest New Drugs 2021; 39(1): 34-51.
[http://dx.doi.org/10.1007/s10637-020-00978-3] [PMID: 32772342]
[25]
Saigi M, Alburquerque-Bejar JJ, Mc Leer-Florin A, et al. MET-oncogenic and JAK2-inactivating alterations are independent factors that affect regulation of PD-L1 expression in lung cancer. Clin Cancer Res 2018; 24(18): 4579-87.
[http://dx.doi.org/10.1158/1078-0432.CCR-18-0267] [PMID: 29898990]
[26]
Engelman JA, Zejnullahu K, Mitsudomi T, et al. MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science 2007; 316(5827): 1039-43.
[http://dx.doi.org/10.1126/science.1141478] [PMID: 17463250]
[27]
Morgillo F, Della Corte CM, Fasano M, Ciardiello F. Mechanisms of resistance to EGFR-targeted drugs: lung cancer. ESMO Open 2016; 1(3): e000060.
[http://dx.doi.org/10.1136/esmoopen-2016-000060] [PMID: 27843613]
[28]
Demuth C, Andersen MN, Jakobsen KR, Madsen AT, Sørensen BS. Increased PD-L1 expression in erlotinib-resistant NSCLC cells with MET gene amplification is reversed upon MET-TKI treatment. Oncotarget 2017; 8(40): 68221-9.
[http://dx.doi.org/10.18632/oncotarget.19920] [PMID: 28978110]
[29]
Garcia-Diaz A, Shin DS, Moreno BH, et al. Interferon receptor signaling pathways regulating PD-L1 and PD-L2 expression. Cell Rep 2017; 19(6): 1189-201.
[http://dx.doi.org/10.1016/j.celrep.2017.04.031] [PMID: 28494868]
[30]
Frumento G, Rotondo R, Tonetti M, Damonte G, Benatti U, Ferrara GB. Tryptophan-derived catabolites are responsible for inhibition of T and natural killer cell proliferation induced by indoleamine 2,3-dioxygenase. J Exp Med 2002; 196(4): 459-68.
[http://dx.doi.org/10.1084/jem.20020121] [PMID: 12186838]
[31]
Gardian K, Janczewska S, Durlik M. Microenvironment elements involved in the development of pancreatic cancer tumor. Gastroenterol Res Pract 2012; 2012: 1-6.
[http://dx.doi.org/10.1155/2012/585674] [PMID: 23304126]
[32]
Yu J, Ohuchida K, Mizumoto K, et al. Overexpression of c-met in the early stage of pancreatic carcinogenesis; altered expression is not sufficient for progression from chronic pancreatitis to pancreatic cancer. World J Gastroenterol 2006; 12(24): 3878-82.
[http://dx.doi.org/10.3748/wjg.v12.i24.3878] [PMID: 16804974]
[33]
Modica C, Tortarolo D, Comoglio P, Basilico C, Vigna E. MET/HGF Co-targeting in pancreatic cancer: A tool to provide insight into the tumor/stroma crosstalk. Int J Mol Sci 2018; 19(12): 3920.
[http://dx.doi.org/10.3390/ijms19123920] [PMID: 30544501]
[34]
Patel MB, Pothula SP, Xu Z, et al. The role of the hepatocyte growth factor/c-MET pathway in pancreatic stellate cell–endothelial cell interactions: antiangiogenic implications in pancreatic cancer. Carcinogenesis 2014; 35(8): 1891-900.
[http://dx.doi.org/10.1093/carcin/bgu122] [PMID: 24876152]
[35]
Ide T, Kitajima Y, Miyoshi A, et al. Tumor–stromal cell interaction under hypoxia increases the invasiveness of pancreatic cancer cells through the hepatocyte growth factor/c-Met pathway. Int J Cancer 2006; 119(12): 2750-9.
[http://dx.doi.org/10.1002/ijc.22178] [PMID: 16998831]
[36]
Avan A, Caretti V, Funel N, et al. Crizotinib inhibits metabolic inactivation of gemcitabine in c-Met-driven pancreatic carcinoma. Cancer Res 2013; 73(22): 6745-56.
[http://dx.doi.org/10.1158/0008-5472.CAN-13-0837] [PMID: 24085787]
[37]
Jin H, Yang R, Zheng Z, et al. MetMAb, the one-armed 5D5 anti-c-Met antibody, inhibits orthotopic pancreatic tumor growth and improves survival. Cancer Res 2008; 68(11): 4360-8.
[http://dx.doi.org/10.1158/0008-5472.CAN-07-5960] [PMID: 18519697]
[38]
Ireland L, Santos A, Ahmed MS, et al. Chemoresistance in pancreatic cancer is driven by stroma-derived insulin-like growth factors. Cancer Res 2016; 76(23): 6851-63.
[http://dx.doi.org/10.1158/0008-5472.CAN-16-1201] [PMID: 27742686]
[39]
Sherman MH, Yu RT, Tseng TW, et al. Stromal cues regulate the pancreatic cancer epigenome and metabolome. Proc Natl Acad Sci 2017; 114(5): 1129-34.
[http://dx.doi.org/10.1073/pnas.1620164114] [PMID: 28096419]
[40]
Xu Z, Pang TCY, Liu AC, et al. Targeting the HGF/c-MET pathway in advanced pancreatic cancer: a key element of treatment that limits primary tumour growth and eliminates metastasis. Br J Cancer 2020; 122(10): 1486-95.
[http://dx.doi.org/10.1038/s41416-020-0782-1] [PMID: 32203220]
[41]
Rucki AA, Xiao Q, Muth S, et al. Dual inhibition of Hedgehog and c-MET pathways for pancreatic cancer treatment. Mol Cancer Ther 2017; 16(11): 2399-409.
[http://dx.doi.org/10.1158/1535-7163.MCT-16-0452]
[42]
Noguchi K, Konno M, Eguchi H, et al. c MET affects gemcitabine resistance during carcinogenesis in a mouse model of pancreatic cancer. Oncol Lett 2018; 16(2): 1892-8.
[43]
Pang TC, Xu Z, Mekapogu AR, et al. HGF/c-MET inhibition as adjuvant therapy improves outcomes in an orthotopic mouse model of pancreatic cancer. Cancers 2021; 13(11): 2763.
[44]
Firuzi O, Che PP, El Hassouni B, et al. Role of c-MET inhibitors in overcoming drug resistance in spheroid models of primary human pancreatic cancer and stellate cells. Cancers 2019; 11(5): 638.
[http://dx.doi.org/10.3390/cancers11050638] [PMID: 31072019]
[45]
Goyal L, Muzumdar MD, Zhu AX. Targeting the HGF/c-MET pathway in hepatocellular carcinoma. Clin Cancer Res 2013; 19(9): 2310-8.
[http://dx.doi.org/10.1158/1078-0432.CCR-12-2791] [PMID: 23388504]
[46]
Munshi N, Jeay S, Li Y, et al. ARQ 197, a novel and selective inhibitor of the human c-Met receptor tyrosine kinase with antitumor activity. Mol Cancer Ther 2010; 9(6): 1544-53.
[http://dx.doi.org/10.1158/1535-7163.MCT-09-1173] [PMID: 20484018]
[47]
Schiffer E, Housset C, Cacheux W, et al. Gefitinib, an EGFR inhibitor, prevents hepatocellular carcinoma development in the rat liver with cirrhosis. Hepatology 2005; 41(2): 307-14.
[http://dx.doi.org/10.1002/hep.20538] [PMID: 15660382]
[48]
Liu L, Wu N, Li J. Novel targeted agents for gastric cancer. J Hematol Oncol 2012; 5(1): 31.
[http://dx.doi.org/10.1186/1756-8722-5-31] [PMID: 22709792]
[49]
Koustas E, Karamouzis MV, Sarantis P, Schizas D, Papavassiliou AG. Inhibition of c‐MET increases the antitumour activity of PARP inhibitors in gastric cancer models. J Cell Mol Med 2020; 24(18): 10420-31.
[http://dx.doi.org/10.1111/jcmm.15655] [PMID: 32686903]
[50]
Janjigian YY, Tang LH, Coit DG, et al. MET expression and amplification in patients with localized gastric cancer. Cancer Epidemiol Biomarkers Prev 2011; 20(5): 1021-7.
[http://dx.doi.org/10.1158/1055-9965.EPI-10-1080] [PMID: 21393565]
[51]
Lee HE, Kim MA, Lee HS, et al. MET in gastric carcinomas: comparison between protein expression and gene copy number and impact on clinical outcome. Br J Cancer 2012; 107(2): 325-33.
[http://dx.doi.org/10.1038/bjc.2012.237] [PMID: 22644302]
[52]
Sun Y, Tian M, Zhou L, You W, Li J. Value of c-Met for predicting progression of precancerous gastric lesions in rural Chinese population. Chin J Cancer Res 2012; 24(1): 18-22.
[http://dx.doi.org/10.1007/s11670-012-0018-x] [PMID: 23359758]
[53]
El Darsa H, El Sayed R, Abdel-Rahman O. MET inhibitors for the treatment of gastric cancer: What’s their potential? J Exp Pharmacol 2020; 12: 349-61.
[http://dx.doi.org/10.2147/JEP.S242958] [PMID: 33116950]
[54]
Dong Y, Xu J, Sun B, Wang J, Wang Z. MET-targeted therapies and clinical outcomes: A systematic literature review. Mol Diagn Ther 2022; 26(2): 203-27.
[http://dx.doi.org/10.1007/s40291-021-00568-w] [PMID: 35266116]
[55]
Marano L, Chiari R, Fabozzi A, et al. c-MET targeting in advanced gastric cancer: An open challenge. Cancer Lett 2015; 365(1): 30-6.
[56]
Huang KH, Sung IC, Fang WL, et al. Correlation between HGF/c-Met and Notch1 signaling pathways in human gastric cancer cells. Oncol Rep 2018; 40(1): 294-302.
[http://dx.doi.org/10.3892/or.2018.6447] [PMID: 29781036]
[57]
Wang H, Rao B, Lou J, et al. The function of the HGF/c-Met axis in hepatocellular carcinoma. Front Cell Dev Biol 2020; 8: 55.
[http://dx.doi.org/10.3389/fcell.2020.00055] [PMID: 32117981]
[58]
Kang YK, Muro K, Ryu MH, et al. A phase II trial of a selective c-Met inhibitor tivantinib (ARQ 197) monotherapy as a second- or third-line therapy in the patients with metastatic gastric cancer. Invest New Drugs 2014; 32(2): 355-61.
[http://dx.doi.org/10.1007/s10637-013-0057-2] [PMID: 24337769]
[59]
Yap TA, Olmos D, Brunetto AT, et al. Phase I trial of a selective c-MET inhibitor ARQ 197 incorporating proof of mechanism pharmacodynamic studies. J Clin Oncol 2011; 29(10): 1271-9.
[http://dx.doi.org/10.1200/JCO.2010.31.0367] [PMID: 21383285]
[60]
Van Cutsem E, Karaszewska B, Kang YK, et al. A multicenter phase II study of AMG 337 in patients with MET -amplified gastric/gastroesophageal junction/esophageal adenocarcinoma and other MET -amplified solid tumors. Clin Cancer Res 2019; 25(8): 2414-23.
[http://dx.doi.org/10.1158/1078-0432.CCR-18-1337] [PMID: 30366938]
[61]
Gavine PR, Ren Y, Han L, et al. Volitinib, a potent and highly selective c-Met inhibitor, effectively blocks c-Met signaling and growth in c-MET amplified gastric cancer patient-derived tumor xenograft models. Mol Oncol 2015; 9(1): 323-33.
[http://dx.doi.org/10.1016/j.molonc.2014.08.015] [PMID: 25248999]
[62]
Lee J, Kim ST, Kim K, et al. Tumor genomic profiling guides patients with metastatic gastric cancer to targeted treatment: The VIKTORY umbrella trial. Cancer Discov 2019; 9(10): 1388-405.
[http://dx.doi.org/10.1158/2159-8290.CD-19-0442] [PMID: 31315834]
[63]
Ye W, He L, Su L, Zheng Z, Ding M, Ye S. Case Report: Prompt response to savolitinib in a case of advanced gastric cancer with bone marrow invasion and met abnormalities. Front Oncol 2022; 12: 868654.
[http://dx.doi.org/10.3389/fonc.2022.868654] [PMID: 35444940]
[64]
Shah MA, Wainberg ZA, Catenacci DVT, et al. Phase II study evaluating 2 dosing schedules of oral foretinib (GSK1363089), cMET/VEGFR2 inhibitor, in patients with metastatic gastric cancer. PLoS One 2013; 8(3): e54014.
[http://dx.doi.org/10.1371/journal.pone.0054014] [PMID: 23516391]
[65]
Hou GX, Song BB. Gastric cancer patient with c-MET amplification treated with crizotinib after failed multi-line treatment: A case report and literature review. Math Biosci Eng 2019; 16(5): 5923-30.
[http://dx.doi.org/10.3934/mbe.2019296]
[66]
Iveson T, Donehower RC, Davidenko I, et al. Rilotumumab in combination with epirubicin, cisplatin, and capecitabine as first-line treatment for gastric or oesophagogastric junction adenocarcinoma: an open-label, dose de-escalation phase 1b study and a double-blind, randomised phase 2 study. Lancet Oncol 2014; 15(9): 1007-18.
[http://dx.doi.org/10.1016/S1470-2045(14)70023-3] [PMID: 24965569]
[67]
Shah MA, Cho JY, Tan IB, et al. A randomized phase II study of FOLFOX with or without the MET inhibitor onartuzumab in advanced adenocarcinoma of the stomach and gastroesophageal junction. Oncologist 2016; 21(9): 1085-90.
[http://dx.doi.org/10.1634/theoncologist.2016-0038] [PMID: 27401892]
[68]
Silva Paiva R, Gomes I, Casimiro S, Fernandes I, Costa L. c-Met expression in renal cell carcinoma with bone metastases. J Bone Oncol 2020; 25: 100315.
[http://dx.doi.org/10.1016/j.jbo.2020.100315] [PMID: 33024658]
[69]
Gibney GT, Aziz SA, Camp RL, et al. c-Met is a prognostic marker and potential therapeutic target in clear cell renal cell carcinoma. Ann Oncol 2013; 24(2): 343-9.
[http://dx.doi.org/10.1093/annonc/mds463] [PMID: 23022995]
[70]
Arnold L, Enders J, Thomas S. Activated HGF-c-met axis in head and neck cancer. Cancers 2017; 9(12): 169.
[http://dx.doi.org/10.3390/cancers9120169] [PMID: 29231907]
[71]
Seiwert TY, Jagadeeswaran R, Faoro L, et al. The MET receptor tyrosine kinase is a potential novel therapeutic target for head and neck squamous cell carcinoma. Cancer Res 2009; 69(7): 3021-31.
[http://dx.doi.org/10.1158/0008-5472.CAN-08-2881] [PMID: 19318576]
[72]
Kim JH, Kim BJ, Kim HS. Clinicopathological impacts of high c-Met expression in head and neck squamous cell carcinoma: a meta-analysis and review. Oncotarget 2017; 8(68): 113120-8.
[http://dx.doi.org/10.18632/oncotarget.21303] [PMID: 29348891]
[73]
Stabile LP, He G, Lui VWY, et al. c-Src activation mediates erlotinib resistance in head and neck cancer by stimulating c-Met. Clin Cancer Res 2013; 19(2): 380-92.
[http://dx.doi.org/10.1158/1078-0432.CCR-12-1555] [PMID: 23213056]
[74]
Sun ZJ, Wu Y, Hou WH, et al. A novel bispecific c-MET/PD-1 antibody with therapeutic potential in solid cancer. Oncotarget 2017; 8(17): 29067-79.
[http://dx.doi.org/10.18632/oncotarget.16173] [PMID: 28404966]
[75]
Thayaparan T, Petrovic RM, Achkova DY, et al. CAR T-cell immunotherapy of MET-expressing malignant mesothelioma. OncoImmunology 2017; 6(12): e1363137.
[http://dx.doi.org/10.1080/2162402X.2017.1363137] [PMID: 29209570]
[76]
Hass R, Jennek S, Yang Y, Friedrich K. c-Met expression and activity in urogenital cancers-novel aspects of signal transduction and medical implications. Cell Commun Signal 2017; 15(1): 10.
[http://dx.doi.org/10.1186/s12964-017-0165-2] [PMID: 28212658]
[77]
Goździk-Spychalska J, Szyszka-Barth K, Spychalski Ł, et al. CMET inhibitors in the treatment of lung cancer. Curr Treat Options Oncol 2014; 15(4): 670-82.
[http://dx.doi.org/10.1007/s11864-014-0313-5] [PMID: 25266653]
[78]
Pasquini G, Giaccone G. C-MET inhibitors for advanced non-small cell lung cancer. Expert Opin Investig Drugs 2018; 27(4): 363-75.
[http://dx.doi.org/10.1080/13543784.2018.1462336] [PMID: 29621416]
[79]
Albitar M, Sudarsanam S, Ma W, et al. Correlation of MET gene amplification and TP53 mutation with PD-L1 expression in non-small cell lung cancer. Oncotarget 2018; 9(17): 13682-93.
[http://dx.doi.org/10.18632/oncotarget.24455] [PMID: 29568386]
[80]
Flaherty KT. Chemotherapy and targeted therapy combinations in advanced melanoma. Clin Cancer Res 2006; 12(7): 2366s-70s.
[http://dx.doi.org/10.1158/1078-0432.CCR-05-2505] [PMID: 16609060]
[81]
Straussman R, Morikawa T, Shee K, et al. Tumour micro-environment elicits innate resistance to RAF inhibitors through HGF secretion. Nature 2012; 487(7408): 500-4.
[http://dx.doi.org/10.1038/nature11183] [PMID: 22763439]
[82]
Demkova L, Kucerova L. Role of the HGF/c-MET tyrosine kinase inhibitors in metastasic melanoma. Mol Cancer 2018; 17(1): 26.
[http://dx.doi.org/10.1186/s12943-018-0795-z] [PMID: 29455657]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy