Skip to main content
Top
Published in: Molecular Cancer 1/2018

Open Access 01-12-2018 | Review

Role of the HGF/c-MET tyrosine kinase inhibitors in metastasic melanoma

Authors: Lucia Demkova, Lucia Kucerova

Published in: Molecular Cancer | Issue 1/2018

Login to get access

Abstract

Metastatic disease in a cancer patient still remains a therapeutic challenge. Metastatic process involves many steps, during which malignant cells succeed to activate cellular pathways promoting survival in hostile environment, engraftment and growth at the distant site from the primary tumor. Melanoma is known for its high propensity to produce metastases even at the early stages of the disease. Here we summarize the most important molecular mechanisms which were associated with the melanoma metastasis. Then, we specifically focus on the signaling pathway mediated by hepatocyte growth factor (HGF) and its receptor c-Met, which play an important role during physiological processes and were been associated with tumorigenesis. We also focus on the effect of the small molecule inhibitors of the tyrosine kinase domain of the c-Met receptor and its effects on properties of melanoma cell. We summarize recent studies, which involved inhibition of the HGF/c-Met signaling in order to decrease melanoma growth and metastatic capacity.
Literature
2.
go back to reference Garbe C, Peris K, Hauschild A, Saiag P, Middleton M, Bastholt L, Grob JJ, Malvehy J, Newton-Bishop J, Stratigos AJ, et al. Diagnosis and treatment of melanoma. European consensus-based interdisciplinary guideline - update 2016. Eur J Cancer. 2016;63:201–17.CrossRefPubMed Garbe C, Peris K, Hauschild A, Saiag P, Middleton M, Bastholt L, Grob JJ, Malvehy J, Newton-Bishop J, Stratigos AJ, et al. Diagnosis and treatment of melanoma. European consensus-based interdisciplinary guideline - update 2016. Eur J Cancer. 2016;63:201–17.CrossRefPubMed
3.
go back to reference Murárová Z, Borecká D. Malígny melanóm – etiopatogenéza, klinický obraz, diagnostika a manažment pacienta. Onkológia. 2016;11:342–7. Murárová Z, Borecká D. Malígny melanóm – etiopatogenéza, klinický obraz, diagnostika a manažment pacienta. Onkológia. 2016;11:342–7.
4.
go back to reference Bobrovská M, Minariková E, Plank L. Diagnostika malígneho melanómu. Onkológia. 2009;4:144–8. Bobrovská M, Minariková E, Plank L. Diagnostika malígneho melanómu. Onkológia. 2009;4:144–8.
5.
go back to reference Garbe C, Peris K, Hauschild A, Saiag P, Middleton M, Spatz A, Grob J-J, Malvehy J, Newton-Bishop J, Stratigos A, et al. Diagnosis and treatment of melanoma: European consensus-based interdisciplinary guideline. Eur J Cancer. 2010;46:270–83.CrossRefPubMed Garbe C, Peris K, Hauschild A, Saiag P, Middleton M, Spatz A, Grob J-J, Malvehy J, Newton-Bishop J, Stratigos A, et al. Diagnosis and treatment of melanoma: European consensus-based interdisciplinary guideline. Eur J Cancer. 2010;46:270–83.CrossRefPubMed
6.
go back to reference Benca J, Havránková M, Hlavatá Z, Pobijáková M, Sieberová G, Šálek T. Manažment pacientov s malígnym melanómom kože. In: Metodický list Ústrednej komisie racionálnej farmakoterapie a liekovej politiky MZ SR, vol. 152012. 2012;15:1-8. Benca J, Havránková M, Hlavatá Z, Pobijáková M, Sieberová G, Šálek T. Manažment pacientov s malígnym melanómom kože. In: Metodický list Ústrednej komisie racionálnej farmakoterapie a liekovej politiky MZ SR, vol. 152012. 2012;15:1-8.
7.
go back to reference Dummer R, Hauschild A, Lindenblatt N, Pentheroudakis G, Keilholz U. Cutaneous melanoma: ESMO clinical practice guidelines for diagnosis, treatment and follow-up†. Ann Oncol. 2015;26:v126–32.CrossRefPubMed Dummer R, Hauschild A, Lindenblatt N, Pentheroudakis G, Keilholz U. Cutaneous melanoma: ESMO clinical practice guidelines for diagnosis, treatment and follow-up†. Ann Oncol. 2015;26:v126–32.CrossRefPubMed
8.
go back to reference Eggermont AMM, Chiarion-Sileni V, Grob J-J, Dummer R, Wolchok JD, Schmidt H, Hamid O, Robert C, Ascierto PA, Richards JM, et al. Adjuvant ipilimumab versus placebo after complete resection of high-risk stage III melanoma (EORTC 18071): a randomised, double-blind, phase 3 trial. Lancet Oncol. 2015;16:522-30. Eggermont AMM, Chiarion-Sileni V, Grob J-J, Dummer R, Wolchok JD, Schmidt H, Hamid O, Robert C, Ascierto PA, Richards JM, et al. Adjuvant ipilimumab versus placebo after complete resection of high-risk stage III melanoma (EORTC 18071): a randomised, double-blind, phase 3 trial. Lancet Oncol. 2015;16:522-30.
9.
go back to reference Urbanček S. Stručný prehľad farmakoterapie metastatického melanómu. Onkológia. 2016;11:351–6. Urbanček S. Stručný prehľad farmakoterapie metastatického melanómu. Onkológia. 2016;11:351–6.
10.
go back to reference Kircher DA, Silvis MR, Cho JH, Holmen SL. Melanoma brain metastasis: mechanisms, models, and medicine. Int J Mol Sci. 2016;17:1-29. Kircher DA, Silvis MR, Cho JH, Holmen SL. Melanoma brain metastasis: mechanisms, models, and medicine. Int J Mol Sci. 2016;17:1-29.
11.
go back to reference Nguyen DX, Bos PD, Massague J. Metastasis: from dissemination to organ-specific colonization. Nat Rev Cancer. 2009;9:274–84.CrossRefPubMed Nguyen DX, Bos PD, Massague J. Metastasis: from dissemination to organ-specific colonization. Nat Rev Cancer. 2009;9:274–84.CrossRefPubMed
12.
go back to reference Yilmaz M, Christofori G, Lehembre F. Distinct mechanisms of tumor invasion and metastasis. Trends Mol Med. 2007;13:535–41.CrossRefPubMed Yilmaz M, Christofori G, Lehembre F. Distinct mechanisms of tumor invasion and metastasis. Trends Mol Med. 2007;13:535–41.CrossRefPubMed
13.
go back to reference Geiger TR, Peeper DS. Metastasis mechanisms. Biochim Biophys Acta. 2009;1796:293–308.PubMed Geiger TR, Peeper DS. Metastasis mechanisms. Biochim Biophys Acta. 2009;1796:293–308.PubMed
14.
go back to reference Lim YC, Kang HJ, Moon JH. C-met pathway promotes self-renewal and tumorigenecity of head and neck squamous cell carcinoma stem-like cell. Oral Oncol. 2014;50:633–9.CrossRefPubMed Lim YC, Kang HJ, Moon JH. C-met pathway promotes self-renewal and tumorigenecity of head and neck squamous cell carcinoma stem-like cell. Oral Oncol. 2014;50:633–9.CrossRefPubMed
15.
go back to reference Clark AG, Vignjevic DM. Modes of cancer cell invasion and the role of the microenvironment. Curr Opin Cell Biol. 2015;36:13–22.CrossRefPubMed Clark AG, Vignjevic DM. Modes of cancer cell invasion and the role of the microenvironment. Curr Opin Cell Biol. 2015;36:13–22.CrossRefPubMed
16.
go back to reference Yilmaz M, Christofori G. Mechanisms of motility in metastasizing cells. Mol Cancer Res. 2010;8:629–42.CrossRefPubMed Yilmaz M, Christofori G. Mechanisms of motility in metastasizing cells. Mol Cancer Res. 2010;8:629–42.CrossRefPubMed
17.
go back to reference Spano D, Heck C, De Antonellis P, Christofori G, Zollo M. Molecular networks that regulate cancer metastasis. Semin Cancer Biol. 2012;22:234–49.CrossRefPubMed Spano D, Heck C, De Antonellis P, Christofori G, Zollo M. Molecular networks that regulate cancer metastasis. Semin Cancer Biol. 2012;22:234–49.CrossRefPubMed
18.
go back to reference Friedl P, Alexander S. Cancer invasion and the microenvironment: plasticity and reciprocity. Cell. 2011;147:992–1009.CrossRefPubMed Friedl P, Alexander S. Cancer invasion and the microenvironment: plasticity and reciprocity. Cell. 2011;147:992–1009.CrossRefPubMed
20.
go back to reference Wan L, Pantel K, Kang Y. Tumor metastasis: moving new biological insights into the clinic. Nat Med. 2013;19:1450–64.CrossRefPubMed Wan L, Pantel K, Kang Y. Tumor metastasis: moving new biological insights into the clinic. Nat Med. 2013;19:1450–64.CrossRefPubMed
21.
go back to reference Hoshino A, Costa-Silva B, Shen T-L, Rodrigues G, Hashimoto A, Tesic Mark M, Molina H, Kohsaka S, Di Giannatale A, Ceder S, et al. Tumour exosome integrins determine organotropic metastasis. Nature. 2015;527:329–35.CrossRefPubMedPubMedCentral Hoshino A, Costa-Silva B, Shen T-L, Rodrigues G, Hashimoto A, Tesic Mark M, Molina H, Kohsaka S, Di Giannatale A, Ceder S, et al. Tumour exosome integrins determine organotropic metastasis. Nature. 2015;527:329–35.CrossRefPubMedPubMedCentral
22.
go back to reference Pani G, Galeotti T, Chiarugi P. Metastasis: cancer cell's escape from oxidative stress. Cancer Metastasis Rev. 2010;29:351–78.CrossRefPubMed Pani G, Galeotti T, Chiarugi P. Metastasis: cancer cell's escape from oxidative stress. Cancer Metastasis Rev. 2010;29:351–78.CrossRefPubMed
23.
go back to reference Tacconelli A, Farina AR, Cappabianca L, DeSantis G, Tessitore A, Vetuschi A, Sferra R, Rucci N, Argenti B, Screpanti I, et al. TrkA alternative splicing: a regulated tumor-promoting switch in human neuroblastoma. Cancer Cell. 2004;6:347–60.CrossRefPubMed Tacconelli A, Farina AR, Cappabianca L, DeSantis G, Tessitore A, Vetuschi A, Sferra R, Rucci N, Argenti B, Screpanti I, et al. TrkA alternative splicing: a regulated tumor-promoting switch in human neuroblastoma. Cancer Cell. 2004;6:347–60.CrossRefPubMed
24.
go back to reference Hernández-Caballero ME. Molecular Mechanisms of Metastasis: Epithelial-Mesenchymal Transition, Anoikis and Loss of Adhesion, Carcinogenesis, Dr. Kathryn Tonissen (Ed.), InTech, London, SE19SG United Kingdom. https://doi.org/10.5772/55399. Hernández-Caballero ME. Molecular Mechanisms of Metastasis: Epithelial-Mesenchymal Transition, Anoikis and Loss of Adhesion, Carcinogenesis, Dr. Kathryn Tonissen (Ed.), InTech, London, SE19SG United Kingdom. https://​doi.​org/​10.​5772/​55399.
25.
go back to reference Shibue T, Weinberg RA. EMT, CSCs, and drug resistance: the mechanistic link and clinical implications. Nat Rev Clin Oncol. 2017;14:611–29.CrossRefPubMed Shibue T, Weinberg RA. EMT, CSCs, and drug resistance: the mechanistic link and clinical implications. Nat Rev Clin Oncol. 2017;14:611–29.CrossRefPubMed
26.
go back to reference Girouard SD, Murphy GF. Melanoma stem cells: not rare, but well done. Lab Investig. 2011;91:647–64.CrossRefPubMed Girouard SD, Murphy GF. Melanoma stem cells: not rare, but well done. Lab Investig. 2011;91:647–64.CrossRefPubMed
27.
go back to reference Murphy MJ. Diagnostic and prognostic biomarkers and therapeutic targets in melanoma. New York: Springer; 2012.CrossRef Murphy MJ. Diagnostic and prognostic biomarkers and therapeutic targets in melanoma. New York: Springer; 2012.CrossRef
28.
go back to reference Kozovska Z, Gabrisova V, Kucerova L. Malignant melanoma: diagnosis, treatment and cancer stem cells. Neoplasma. 2016;63:510–7.CrossRefPubMed Kozovska Z, Gabrisova V, Kucerova L. Malignant melanoma: diagnosis, treatment and cancer stem cells. Neoplasma. 2016;63:510–7.CrossRefPubMed
29.
go back to reference Brinckerhoff CE. Cancer stem cells (CSCs) in melanoma: There's smoke, but is there fire? J Cell Physiol. 2017;232:2674-78. Brinckerhoff CE. Cancer stem cells (CSCs) in melanoma: There's smoke, but is there fire? J Cell Physiol. 2017;232:2674-78.
30.
go back to reference Schatton T, Murphy GF, Frank NY, Yamaura K, Waaga-Gasser AM, Gasser M, Zhan Q, Jordan S, Duncan LM, Weishaupt C, et al. Identification of cells initiating human melanomas. Nature. 2008;451:345–9.CrossRefPubMedPubMedCentral Schatton T, Murphy GF, Frank NY, Yamaura K, Waaga-Gasser AM, Gasser M, Zhan Q, Jordan S, Duncan LM, Weishaupt C, et al. Identification of cells initiating human melanomas. Nature. 2008;451:345–9.CrossRefPubMedPubMedCentral
31.
go back to reference Fusi A, Reichelt U, Busse A, Ochsenreither S, Rietz A, Maisel M, Keilholz U. Expression of the stem cell markers nestin and CD133 on circulating melanoma cells. J Investig Dermatol. 2010;131:487–94.CrossRefPubMed Fusi A, Reichelt U, Busse A, Ochsenreither S, Rietz A, Maisel M, Keilholz U. Expression of the stem cell markers nestin and CD133 on circulating melanoma cells. J Investig Dermatol. 2010;131:487–94.CrossRefPubMed
32.
go back to reference Zwick E, Bange J, Ullrich A. Receptor tyrosine kinase signalling as a target for cancer intervention strategies. Endocr Relat Cancer. 2001;8:161–73.CrossRefPubMed Zwick E, Bange J, Ullrich A. Receptor tyrosine kinase signalling as a target for cancer intervention strategies. Endocr Relat Cancer. 2001;8:161–73.CrossRefPubMed
37.
go back to reference Li Y, Li A, Glas M, Lal B, Ying M, Sang Y, Xia S, Trageser D, Guerrero-Cazares H, Eberhart CG, et al. C-met signaling induces a reprogramming network and supports the glioblastoma stem-like phenotype. Proc Natl Acad Sci U S A. 2011;108:9951–6.CrossRefPubMedPubMedCentral Li Y, Li A, Glas M, Lal B, Ying M, Sang Y, Xia S, Trageser D, Guerrero-Cazares H, Eberhart CG, et al. C-met signaling induces a reprogramming network and supports the glioblastoma stem-like phenotype. Proc Natl Acad Sci U S A. 2011;108:9951–6.CrossRefPubMedPubMedCentral
38.
go back to reference Yano S, Takeuchi S, Nakagawa T, Yamada T. Ligand-triggered resistance to molecular targeted drugs in lung cancer: roles of hepatocyte growth factor and epidermal growth factor receptor ligands. Cancer Sci. 2012;103:1189–94.CrossRefPubMed Yano S, Takeuchi S, Nakagawa T, Yamada T. Ligand-triggered resistance to molecular targeted drugs in lung cancer: roles of hepatocyte growth factor and epidermal growth factor receptor ligands. Cancer Sci. 2012;103:1189–94.CrossRefPubMed
39.
go back to reference Daud A, Kluger HM, Kurzrock R, Schimmoller F, Weitzman AL, Samuel TA, Moussa AH, Gordon MS, Shapiro GI. Phase II randomised discontinuation trial of the MET/VEGF receptor inhibitor cabozantinib in metastatic melanoma. Br J Cancer. 2017;116:432–40.CrossRefPubMedPubMedCentral Daud A, Kluger HM, Kurzrock R, Schimmoller F, Weitzman AL, Samuel TA, Moussa AH, Gordon MS, Shapiro GI. Phase II randomised discontinuation trial of the MET/VEGF receptor inhibitor cabozantinib in metastatic melanoma. Br J Cancer. 2017;116:432–40.CrossRefPubMedPubMedCentral
40.
go back to reference Bendinelli P, Maroni P, Matteucci E, Desiderio MA. Epigenetic regulation of HGF/met receptor axis is critical for the outgrowth of bone metastasis from breast carcinoma. Cell Death Dis. 2017;8:e2578.CrossRefPubMedPubMedCentral Bendinelli P, Maroni P, Matteucci E, Desiderio MA. Epigenetic regulation of HGF/met receptor axis is critical for the outgrowth of bone metastasis from breast carcinoma. Cell Death Dis. 2017;8:e2578.CrossRefPubMedPubMedCentral
41.
go back to reference Birchmeier C, Birchmeier W, Gherardi E, Vande Woude GF. Met, metastasis, motility and more. Nat Rev Mol Cell Biol. 2003;4:915–25.CrossRefPubMed Birchmeier C, Birchmeier W, Gherardi E, Vande Woude GF. Met, metastasis, motility and more. Nat Rev Mol Cell Biol. 2003;4:915–25.CrossRefPubMed
42.
go back to reference Bladt F, Riethmacher D, Isenmann S, Aguzzi A, Birchmeier C. Essential role for the c-met receptor in the migration of myogenic precursor cells into the limb bud. Nature. 1995;376:768–71.CrossRefPubMed Bladt F, Riethmacher D, Isenmann S, Aguzzi A, Birchmeier C. Essential role for the c-met receptor in the migration of myogenic precursor cells into the limb bud. Nature. 1995;376:768–71.CrossRefPubMed
43.
go back to reference Jiang WG, Hiscox S. Hepatocyte growth factor/scatter factor, a cytokine playing multiple and converse roles. Histol Histopathol. 1997;12:537–55.PubMed Jiang WG, Hiscox S. Hepatocyte growth factor/scatter factor, a cytokine playing multiple and converse roles. Histol Histopathol. 1997;12:537–55.PubMed
44.
go back to reference Nakamura T. Structure and function of hepatocyte growth factor. Progress in growth factor research. 1991;3:65–87.CrossRef Nakamura T. Structure and function of hepatocyte growth factor. Progress in growth factor research. 1991;3:65–87.CrossRef
45.
go back to reference Barrow-McGee R, Kermorgant S. Met endosomal signalling: in the right place, at the right time. Int J Biochem Cell Biol. 2014;49:69–74.CrossRefPubMed Barrow-McGee R, Kermorgant S. Met endosomal signalling: in the right place, at the right time. Int J Biochem Cell Biol. 2014;49:69–74.CrossRefPubMed
46.
go back to reference Seki T, Hagiya M, Shimonishi M, Nakamura T, Shimizu S. Organization of the human hepatocyte growth factor-encoding gene. Gene. 1991;102:213–9.CrossRefPubMed Seki T, Hagiya M, Shimonishi M, Nakamura T, Shimizu S. Organization of the human hepatocyte growth factor-encoding gene. Gene. 1991;102:213–9.CrossRefPubMed
47.
go back to reference Maulik G, Shrikhande A, Kijima T, Ma PC, Morrison PT, Salgia R. Maulik role of the hepatocyte growth factor receptor, c-met, in oncogenetics and potencial for therapeucic inhibition. Cytokine Growth Factor Rev. 2002;13:41–59.CrossRefPubMed Maulik G, Shrikhande A, Kijima T, Ma PC, Morrison PT, Salgia R. Maulik role of the hepatocyte growth factor receptor, c-met, in oncogenetics and potencial for therapeucic inhibition. Cytokine Growth Factor Rev. 2002;13:41–59.CrossRefPubMed
48.
go back to reference Tan Q, Hu J, Yu X, Guan W, Lu H, Yu Y, Zang G, Tang Z. The role of IL-1 family members and Kupffer cells in liver regeneration. Biomed Res Int. 2016;2016:6495793.PubMedPubMedCentral Tan Q, Hu J, Yu X, Guan W, Lu H, Yu Y, Zang G, Tang Z. The role of IL-1 family members and Kupffer cells in liver regeneration. Biomed Res Int. 2016;2016:6495793.PubMedPubMedCentral
49.
go back to reference Liu Y. The human hepatocyte growth factor receptor gene: complete structural organization and promoter characterization. Gene. 1998;215:159–69.CrossRefPubMed Liu Y. The human hepatocyte growth factor receptor gene: complete structural organization and promoter characterization. Gene. 1998;215:159–69.CrossRefPubMed
50.
go back to reference Samame Perez-Vargas JC, Biondani P, Maggi C, Gariboldi M, Gloghini A, Inno A, Volpi CC, Gualeni AV, di Bartolomeo M, de Braud F, et al. Role of cMET in the development and progression of colorectal cancer. Int J Mol Sci. 2013;14:18056–77.CrossRefPubMed Samame Perez-Vargas JC, Biondani P, Maggi C, Gariboldi M, Gloghini A, Inno A, Volpi CC, Gualeni AV, di Bartolomeo M, de Braud F, et al. Role of cMET in the development and progression of colorectal cancer. Int J Mol Sci. 2013;14:18056–77.CrossRefPubMed
51.
go back to reference Garajová I, Giovannetti E, Biasco G, Peters GJ. C-met as a target for personalized therapy. Translational Oncogenomics. 2015;7:13–31.PubMedPubMedCentral Garajová I, Giovannetti E, Biasco G, Peters GJ. C-met as a target for personalized therapy. Translational Oncogenomics. 2015;7:13–31.PubMedPubMedCentral
52.
go back to reference Gherardi E, Birchmeier W, Birchmeier C, Vande Woude G. Targeting MET in cancer: rationale and progress. Nat Rev Cancer. 2012;12:89–103.CrossRefPubMed Gherardi E, Birchmeier W, Birchmeier C, Vande Woude G. Targeting MET in cancer: rationale and progress. Nat Rev Cancer. 2012;12:89–103.CrossRefPubMed
53.
go back to reference Zhu H, Naujokas MA, Park M. Receptor chimeras indicate that the met tyrosine kinase mediates the motility and morphogenic responses of hepatocyte growth/scatter factor. Cell Growth Differ. 1994;5:359–66.PubMed Zhu H, Naujokas MA, Park M. Receptor chimeras indicate that the met tyrosine kinase mediates the motility and morphogenic responses of hepatocyte growth/scatter factor. Cell Growth Differ. 1994;5:359–66.PubMed
54.
go back to reference Kmiecik TE, Keller JR, Rosen E, Vande Woude GF. Hepatocyte growth factor is a synergistic factor for the growth of hematopoietic progenitor cells. Blood. 1992;80:2454–7.PubMed Kmiecik TE, Keller JR, Rosen E, Vande Woude GF. Hepatocyte growth factor is a synergistic factor for the growth of hematopoietic progenitor cells. Blood. 1992;80:2454–7.PubMed
55.
go back to reference Böhm F, Köhler UA, Speicher T, Werner S. Regulation of liver regeneration by growth factors and cytokines. EMBO Molecular Medicine. 2010;2:294–305.CrossRefPubMedPubMedCentral Böhm F, Köhler UA, Speicher T, Werner S. Regulation of liver regeneration by growth factors and cytokines. EMBO Molecular Medicine. 2010;2:294–305.CrossRefPubMedPubMedCentral
56.
go back to reference Chmielowiec J, Borowiak M, Morkel M, Stradal T, Munz B, Werner S, Wehland J, Birchmeier C, Birchmeier W. C-met is essential for wound healing in the skin. J Cell Biol. 2007;177:151–62.CrossRefPubMedPubMedCentral Chmielowiec J, Borowiak M, Morkel M, Stradal T, Munz B, Werner S, Wehland J, Birchmeier C, Birchmeier W. C-met is essential for wound healing in the skin. J Cell Biol. 2007;177:151–62.CrossRefPubMedPubMedCentral
57.
go back to reference Maroun CR, Rowlands T. The met receptor tyrosine kinase: a key player in oncogenesis and drug resistance. Pharmacol Ther. 2014;142:316–38.CrossRefPubMed Maroun CR, Rowlands T. The met receptor tyrosine kinase: a key player in oncogenesis and drug resistance. Pharmacol Ther. 2014;142:316–38.CrossRefPubMed
59.
go back to reference Cao HH, Cheng CY, Su T, Fu XQ, Guo H, Li T, Tse AK, Kwan HY, Yu H, Yu ZL. Quercetin inhibits HGF/c-met signaling and HGF-stimulated melanoma cell migration and invasion. Mol Cancer. 2015;14:103.CrossRefPubMedPubMedCentral Cao HH, Cheng CY, Su T, Fu XQ, Guo H, Li T, Tse AK, Kwan HY, Yu H, Yu ZL. Quercetin inhibits HGF/c-met signaling and HGF-stimulated melanoma cell migration and invasion. Mol Cancer. 2015;14:103.CrossRefPubMedPubMedCentral
60.
go back to reference Kucerova L, Skolekova S, Demkova L, Bohovic R, Matuskova M. Long-term efficiency of mesenchymal stromal cell-mediated CD-MSC/5FC therapy in human melanoma xenograft model. Gene Ther. 2014;21:874–87.CrossRefPubMed Kucerova L, Skolekova S, Demkova L, Bohovic R, Matuskova M. Long-term efficiency of mesenchymal stromal cell-mediated CD-MSC/5FC therapy in human melanoma xenograft model. Gene Ther. 2014;21:874–87.CrossRefPubMed
61.
go back to reference Parikh RA, Wang P, Beumer JH, Chu E, Appleman LJ. The potential roles of hepatocyte growth factor (HGF)-MET pathway inhibitors in cancer treatment. Onco Targets Ther. 2014;7:969–83.PubMedPubMedCentral Parikh RA, Wang P, Beumer JH, Chu E, Appleman LJ. The potential roles of hepatocyte growth factor (HGF)-MET pathway inhibitors in cancer treatment. Onco Targets Ther. 2014;7:969–83.PubMedPubMedCentral
62.
go back to reference Zenali M. deKay J, Liu Z, Hamilton S, Zuo Z, Lu X, Bakkar R, Mills G, Broaddus R: retrospective review of MET gene mutations. Oncoscience. 2015;2:533–41.CrossRefPubMedPubMedCentral Zenali M. deKay J, Liu Z, Hamilton S, Zuo Z, Lu X, Bakkar R, Mills G, Broaddus R: retrospective review of MET gene mutations. Oncoscience. 2015;2:533–41.CrossRefPubMedPubMedCentral
63.
go back to reference Gentile A, Trusolino L, Comoglio PM. The met tyrosine kinase receptor in development and cancer. Cancer Metastasis Rev. 2008;27:85–94.CrossRefPubMed Gentile A, Trusolino L, Comoglio PM. The met tyrosine kinase receptor in development and cancer. Cancer Metastasis Rev. 2008;27:85–94.CrossRefPubMed
64.
go back to reference Sattler M, Pride YB, Ma P, Gramlich JL, Chu SC, Quinnan LA, Shirazian S, Liang C, Podar K, Christensen JG, Salgia R. A novel small molecule met inhibitor induces apoptosis in cells transformed by the Oncogenic TPR-MET tyrosine Kinase. Cancer Res. 2003;63:5462–9.PubMed Sattler M, Pride YB, Ma P, Gramlich JL, Chu SC, Quinnan LA, Shirazian S, Liang C, Podar K, Christensen JG, Salgia R. A novel small molecule met inhibitor induces apoptosis in cells transformed by the Oncogenic TPR-MET tyrosine Kinase. Cancer Res. 2003;63:5462–9.PubMed
65.
go back to reference Puri N, Ahmed S, Janamanchi V, Tretiakova M, Zumba O, Krausz T, Jagadeeswaran R, Salgia R. C-met is a potentially new therapeutic target for treatment of human melanoma. Clin Cancer Res. 2007;13:2246–53.CrossRefPubMed Puri N, Ahmed S, Janamanchi V, Tretiakova M, Zumba O, Krausz T, Jagadeeswaran R, Salgia R. C-met is a potentially new therapeutic target for treatment of human melanoma. Clin Cancer Res. 2007;13:2246–53.CrossRefPubMed
66.
go back to reference Etnyre D, Stone AL, Fong JT, Jacobs RJ, Uppada SB, Botting GM, Rajanna S, Moravec DN, Shambannagari MR, Crees Z, et al. Targeting c-Met in melanoma. Cancer Biology & Therapy. 2014;15:1129–41.CrossRef Etnyre D, Stone AL, Fong JT, Jacobs RJ, Uppada SB, Botting GM, Rajanna S, Moravec DN, Shambannagari MR, Crees Z, et al. Targeting c-Met in melanoma. Cancer Biology & Therapy. 2014;15:1129–41.CrossRef
67.
go back to reference Ma PC, Jagadeeswaran R, Jagadeesh S, Tretiakova MS, Nallasura V, Fox EA, 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;65:1479–88.CrossRefPubMed Ma PC, Jagadeeswaran R, Jagadeesh S, Tretiakova MS, Nallasura V, Fox EA, 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;65:1479–88.CrossRefPubMed
68.
go back to reference Kenessey I, Keszthelyi M, Kramer Z, Berta J, Adam A, Dobos J, Mildner M, Flachner B, Cseh S, Barna G, et al. Inhibition of c-met with the specific small molecule tyrosine Kinase inhibitor SU11274 decreases growth and metastasis formation of experimental human melanoma. Curr Cancer Drug Targets. 2010;10:332–42.CrossRefPubMed Kenessey I, Keszthelyi M, Kramer Z, Berta J, Adam A, Dobos J, Mildner M, Flachner B, Cseh S, Barna G, et al. Inhibition of c-met with the specific small molecule tyrosine Kinase inhibitor SU11274 decreases growth and metastasis formation of experimental human melanoma. Curr Cancer Drug Targets. 2010;10:332–42.CrossRefPubMed
69.
go back to reference Kucerova L, Demkova L, Skolekova S, Bohovic R, Matuskova M. Tyrosine kinase inhibitor SU11274 increased tumorigenicity and enriched for melanoma-initiating cells by bioenergetic modulation. BMC Cancer. 2016;16:308.CrossRefPubMedPubMedCentral Kucerova L, Demkova L, Skolekova S, Bohovic R, Matuskova M. Tyrosine kinase inhibitor SU11274 increased tumorigenicity and enriched for melanoma-initiating cells by bioenergetic modulation. BMC Cancer. 2016;16:308.CrossRefPubMedPubMedCentral
70.
go back to reference Kazandjian D, Blumenthal GM, Chen HY, He K, Patel M, Justice R, Keegan P, Pazdur R. FDA approval summary: crizotinib for the treatment of metastatic non-small cell lung cancer with anaplastic lymphoma kinase rearrangements. Oncologist. 2014;19:e5–11.CrossRefPubMedPubMedCentral Kazandjian D, Blumenthal GM, Chen HY, He K, Patel M, Justice R, Keegan P, Pazdur R. FDA approval summary: crizotinib for the treatment of metastatic non-small cell lung cancer with anaplastic lymphoma kinase rearrangements. Oncologist. 2014;19:e5–11.CrossRefPubMedPubMedCentral
71.
go back to reference Zou HY, Li Q, Lee JH, Arango ME, McDonnell SR, Yamazaki S, Koudriakova TB, Alton G, Cui JJ, Kung P-P, et al. An orally available small-molecule inhibitor of c-met, PF-2341066, exhibits Cytoreductive antitumor efficacy through Antiproliferative and Antiangiogenic mechanisms. Cancer Res. 2007;67:4408–17.CrossRefPubMed Zou HY, Li Q, Lee JH, Arango ME, McDonnell SR, Yamazaki S, Koudriakova TB, Alton G, Cui JJ, Kung P-P, et al. An orally available small-molecule inhibitor of c-met, PF-2341066, exhibits Cytoreductive antitumor efficacy through Antiproliferative and Antiangiogenic mechanisms. Cancer Res. 2007;67:4408–17.CrossRefPubMed
72.
go back to reference Christensen JG, Zou HY, Arango ME, Li Q, Lee JH, McDonnell SR, Yamazaki S, Alton GR, Mroczkowski B, Los G. Cytoreductive antitumor activity of PF-2341066, a novel inhibitor of anaplastic lymphoma kinase and c-met, in experimental models of anaplastic large-cell lymphoma. Mol Cancer Ther. 2007;6:3314–22.CrossRefPubMed Christensen JG, Zou HY, Arango ME, Li Q, Lee JH, McDonnell SR, Yamazaki S, Alton GR, Mroczkowski B, Los G. Cytoreductive antitumor activity of PF-2341066, a novel inhibitor of anaplastic lymphoma kinase and c-met, in experimental models of anaplastic large-cell lymphoma. Mol Cancer Ther. 2007;6:3314–22.CrossRefPubMed
73.
go back to reference Hughes PE, Rex K, Caenepeel S, Yang Y, Zhang Y, Broome MA, Kha HT, Burgess TL, Amore B, Kaplan-Lefko PJ, et al. <em>in vitro</em> and <em>in vivo</em> activity of AMG 337, a potent and selective MET Kinase inhibitor, in MET-dependent cancer models. Mol Cancer Ther. 2016;15:1568–79.CrossRefPubMed Hughes PE, Rex K, Caenepeel S, Yang Y, Zhang Y, Broome MA, Kha HT, Burgess TL, Amore B, Kaplan-Lefko PJ, et al. <em>in vitro</em> and <em>in vivo</em> activity of AMG 337, a potent and selective MET Kinase inhibitor, in MET-dependent cancer models. Mol Cancer Ther. 2016;15:1568–79.CrossRefPubMed
74.
go back to reference Liu L, Siegmund A, Xi N, Kaplan-Lefko P, Rex K, Chen A, Lin J, Moriguchi J, Berry L, Huang L, et al. Discovery of a potent, selective, and orally bioavailable c-met inhibitor: 1-(2-Hydroxy-2-methylpropyl)-N-(5-(7-methoxyquinolin-4-yloxy)pyridin-2-yl)-5-methyl-3-oxo-2-phenyl-2,3-dihydro-1H-pyrazole-4-carboxamide (AMG 458). J Med Chem. 2008;51:3688–91.CrossRefPubMed Liu L, Siegmund A, Xi N, Kaplan-Lefko P, Rex K, Chen A, Lin J, Moriguchi J, Berry L, Huang L, et al. Discovery of a potent, selective, and orally bioavailable c-met inhibitor: 1-(2-Hydroxy-2-methylpropyl)-N-(5-(7-methoxyquinolin-4-yloxy)pyridin-2-yl)-5-methyl-3-oxo-2-phenyl-2,3-dihydro-1H-pyrazole-4-carboxamide (AMG 458). J Med Chem. 2008;51:3688–91.CrossRefPubMed
75.
go back to reference Li B, Torossian A, Sun Y, Du R, Dicker AP, Lu B. Higher levels of c-met expression and phosphorylation identify cell lines with increased sensitivity to AMG-458, a novel selective c-met inhibitor with radiosensitizing effects. Int J Radiat Oncol Biol Phys. 2012;84:e525-31. Li B, Torossian A, Sun Y, Du R, Dicker AP, Lu B. Higher levels of c-met expression and phosphorylation identify cell lines with increased sensitivity to AMG-458, a novel selective c-met inhibitor with radiosensitizing effects. Int J Radiat Oncol Biol Phys. 2012;84:e525-31.
76.
go back to reference Kataoka Y, Mukohara T, Tomioka H, Funakoshi Y, Kiyota N, Fujiwara Y, Yashiro M, Hirakawa K, Hirai M, Minami H. Foretinib (GSK1363089), a multi-kinase inhibitor of MET and VEGFRs, inhibits growth of gastric cancer cell lines by blocking inter-receptor tyrosine kinase networks. Investig New Drugs. 2012;30:1352–60.CrossRef Kataoka Y, Mukohara T, Tomioka H, Funakoshi Y, Kiyota N, Fujiwara Y, Yashiro M, Hirakawa K, Hirai M, Minami H. Foretinib (GSK1363089), a multi-kinase inhibitor of MET and VEGFRs, inhibits growth of gastric cancer cell lines by blocking inter-receptor tyrosine kinase networks. Investig New Drugs. 2012;30:1352–60.CrossRef
77.
go back to reference Chen H-M, Tsai C-H, Hung W-C. Foretinib inhibits angiogenesis, lymphangiogenesis and tumor growth of pancreatic cancer in vivo by decreasing VEGFR-2/3 and TIE-2 signaling. Oncotarget. 2015;6:14940–52.PubMedPubMedCentral Chen H-M, Tsai C-H, Hung W-C. Foretinib inhibits angiogenesis, lymphangiogenesis and tumor growth of pancreatic cancer in vivo by decreasing VEGFR-2/3 and TIE-2 signaling. Oncotarget. 2015;6:14940–52.PubMedPubMedCentral
78.
go back to reference 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;69:8009–16.CrossRefPubMed 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;69:8009–16.CrossRefPubMed
79.
go back to reference Torres KE, Zhu Q-S, Bill K, Lopez G, Ghadimi MP, Xie X, Young ED, Liu J, Nguyen T, Bolshakov S, et al. Activated MET is a molecular prognosticator and potential therapeutic target for malignant peripheral nerve sheath tumors. Clin Cancer Res. 2011;17:3943–55.CrossRefPubMedPubMedCentral Torres KE, Zhu Q-S, Bill K, Lopez G, Ghadimi MP, Xie X, Young ED, Liu J, Nguyen T, Bolshakov S, et al. Activated MET is a molecular prognosticator and potential therapeutic target for malignant peripheral nerve sheath tumors. Clin Cancer Res. 2011;17:3943–55.CrossRefPubMedPubMedCentral
80.
go back to reference Christensen JG, Schreck R, Burrows J, Kuruganti P, Chan E, Le P, Chen J, Wang X, Ruslim L, Blake R, et al. A selective small molecule inhibitor of c-met Kinase inhibits c-met-dependent phenotypes in vitro and exhibits Cytoreductive antitumor activity in vivo. Cancer Res. 2003;63:7345–55.PubMed Christensen JG, Schreck R, Burrows J, Kuruganti P, Chan E, Le P, Chen J, Wang X, Ruslim L, Blake R, et al. A selective small molecule inhibitor of c-met Kinase inhibits c-met-dependent phenotypes in vitro and exhibits Cytoreductive antitumor activity in vivo. Cancer Res. 2003;63:7345–55.PubMed
81.
go back to reference Chattopadhyay C, Ellerhorst JA, Ekmekcioglu S, Greene VR, Davies MA, Grimm EA. Association of Activated c-met with NRAS-mutated human melanomas: a possible avenue for targeting. Int J Cancer. 2012;131:E56–65.CrossRefPubMedPubMedCentral Chattopadhyay C, Ellerhorst JA, Ekmekcioglu S, Greene VR, Davies MA, Grimm EA. Association of Activated c-met with NRAS-mutated human melanomas: a possible avenue for targeting. Int J Cancer. 2012;131:E56–65.CrossRefPubMedPubMedCentral
82.
go back to reference Byeon HK, Na HJ, Yang YJ, Kwon HJ, Chang JW, Ban MJ, Kim WS, Shin DY, Lee EJ, Koh YW, et al. C-met-mediated reactivation of PI3K/AKT signaling contributes to insensitivity of BRAF(V600E) mutant thyroid cancer to BRAF inhibition. Mol Carcinog. 2016;55:1678–87.CrossRefPubMed Byeon HK, Na HJ, Yang YJ, Kwon HJ, Chang JW, Ban MJ, Kim WS, Shin DY, Lee EJ, Koh YW, et al. C-met-mediated reactivation of PI3K/AKT signaling contributes to insensitivity of BRAF(V600E) mutant thyroid cancer to BRAF inhibition. Mol Carcinog. 2016;55:1678–87.CrossRefPubMed
83.
go back to reference Pan B-S, Chan GKY, Chenard M, Chi A, Davis LJ, Deshmukh SV, Gibbs JB, Gil S, Hang G, Hatch H, et al. MK-2461, a novel multitargeted Kinase inhibitor, preferentially inhibits the activated c-met receptor. Cancer Res. 2010;70:1524–33.CrossRefPubMed Pan B-S, Chan GKY, Chenard M, Chi A, Davis LJ, Deshmukh SV, Gibbs JB, Gil S, Hang G, Hatch H, et al. MK-2461, a novel multitargeted Kinase inhibitor, preferentially inhibits the activated c-met receptor. Cancer Res. 2010;70:1524–33.CrossRefPubMed
84.
go back to reference 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;73:3692–703.CrossRefPubMedPubMedCentral 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;73:3692–703.CrossRefPubMedPubMedCentral
85.
go back to reference Basilico C, Pennacchietti S, Vigna E, Chiriaco C, Arena S, Bardelli A, Valdembri D, Serini G, Michieli P. Tivantinib (ARQ197) displays Cytotoxic activity that is independent of its ability to bind MET. Clin Cancer Res. 2013;19:2381–92.CrossRefPubMed Basilico C, Pennacchietti S, Vigna E, Chiriaco C, Arena S, Bardelli A, Valdembri D, Serini G, Michieli P. Tivantinib (ARQ197) displays Cytotoxic activity that is independent of its ability to bind MET. Clin Cancer Res. 2013;19:2381–92.CrossRefPubMed
86.
go back to reference Obenauf AC, Zou Y, Ji AL, Vanharanta S, Shu W, Shi H, Kong X, Bosenberg MC, Wiesner T, Rosen N, et al. Therapy-induced tumour secretomes promote resistance and tumour progression. Nature. 2015;520:368–72.CrossRefPubMedPubMedCentral Obenauf AC, Zou Y, Ji AL, Vanharanta S, Shu W, Shi H, Kong X, Bosenberg MC, Wiesner T, Rosen N, et al. Therapy-induced tumour secretomes promote resistance and tumour progression. Nature. 2015;520:368–72.CrossRefPubMedPubMedCentral
87.
go back to reference Schöffski P, Gordon M, Smith DC, Kurzrock R, Daud A, Vogelzang NJ, Lee Y, Scheffold C, Shapiro GI. Phase II randomised discontinuation trial of cabozantinib in patients with advanced solid tumours. Eur J Cancer. 2017;86:296–304.CrossRefPubMed Schöffski P, Gordon M, Smith DC, Kurzrock R, Daud A, Vogelzang NJ, Lee Y, Scheffold C, Shapiro GI. Phase II randomised discontinuation trial of cabozantinib in patients with advanced solid tumours. Eur J Cancer. 2017;86:296–304.CrossRefPubMed
88.
go back to reference Gordon MS, Kluger HM, Shapiro G, Kurzrock R, Edelman G, Samuel TA, Moussa AH, Ramies DA, Laird AD, Schimmoller F, et al. Activity of cabozantinib (XL184) in metastatic melanoma: results from a phase II randomized discontinuation trial (RDT). J Clin Oncol. 2012;30:8531. Gordon MS, Kluger HM, Shapiro G, Kurzrock R, Edelman G, Samuel TA, Moussa AH, Ramies DA, Laird AD, Schimmoller F, et al. Activity of cabozantinib (XL184) in metastatic melanoma: results from a phase II randomized discontinuation trial (RDT). J Clin Oncol. 2012;30:8531.
89.
go back to reference Surriga O, Rajasekhar VK, Ambrosini G, Dogan Y, Huang R, Schwartz GK. Crizotinib, a c-met inhibitor, prevents metastasis in a metastatic Uveal melanoma model. Mol Cancer Ther. 2013;12:2817–26.CrossRefPubMed Surriga O, Rajasekhar VK, Ambrosini G, Dogan Y, Huang R, Schwartz GK. Crizotinib, a c-met inhibitor, prevents metastasis in a metastatic Uveal melanoma model. Mol Cancer Ther. 2013;12:2817–26.CrossRefPubMed
90.
go back to reference Shojaei F, Lee JH, Simmons BH, Wong A, Esparza CO, Plumlee PA, Feng J, Stewart AE, Hu-Lowe DD, Christensen JG. HGF/c-met acts as an alternative Angiogenic pathway in Sunitinib-resistant tumors. Cancer Res. 2010;70:10090–100.CrossRefPubMed Shojaei F, Lee JH, Simmons BH, Wong A, Esparza CO, Plumlee PA, Feng J, Stewart AE, Hu-Lowe DD, Christensen JG. HGF/c-met acts as an alternative Angiogenic pathway in Sunitinib-resistant tumors. Cancer Res. 2010;70:10090–100.CrossRefPubMed
Metadata
Title
Role of the HGF/c-MET tyrosine kinase inhibitors in metastasic melanoma
Authors
Lucia Demkova
Lucia Kucerova
Publication date
01-12-2018
Publisher
BioMed Central
Published in
Molecular Cancer / Issue 1/2018
Electronic ISSN: 1476-4598
DOI
https://doi.org/10.1186/s12943-018-0795-z

Other articles of this Issue 1/2018

Molecular Cancer 1/2018 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine