Melanoma Targeted Therapies beyond BRAF-Mutant Melanoma: Potential Druggable Mutations and Novel Treatment Approaches
Abstract
:Simple Summary
Abstract
1. Introduction
2. The Mutational Landscape in Metastatic Melanomas
3. Targeting Activating Mutations in the KIT Oncogene
4. Targeting Activating Mutations in RAS Oncogenes
4.1. NRAS
4.1.1. NRAS Biology
4.1.2. The Frequency in Melanomas and Characteristics
4.1.3. The Prognostic Impact of NRAS in Melanomas
4.1.4. Targeting Upstream Effectors of NRAS
4.1.5. Targeting NRAS
4.1.6. Post-Translational Targets in RAS Proteins
4.1.7. Targeting the Downstream Effectors of NRAS
4.2. Targeting KRAS/HRAS
5. Targeting Tumor Suppressor Gene Alterations
5.1. Targeting NF1 in Melanomas
5.2. Targeting TP53
5.3. Targeting CDKN2A and CDK4/6 Networks
6. Targeting Fusion Gene Alterations
7. Targeting Epigenetic Regulators
8. Targeting Homologous Recombination Deficiency and the DNA Damage Response Pathway
9. Conclusions
Author Contributions
Funding
Conflicts of Interest
Abbreviations
References
- Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef]
- Karimkhani, C.; Green, A.; Nijsten, T.; Weinstock, M.; Dellavalle, R.; Naghavi, M.; Fitzmaurice, C. The global burden of melanoma: Results from the Global Burden of Disease Study. Br. J. Dermatol. 2017, 177, 134–140. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Guy, G.P., Jr.; Thomas, C.C.; Thompson, T.; Watson, M.; Massetti, G.M.; Richardson, L.C. Centers for Disease Control and Prevention (CDC). Vital signs: Melanoma incidence and mortality trends and projections—United States, 1982–2030. MMWR Morb Mortal Wkly Rep. 2015, 64, 591. [Google Scholar]
- Garbe, C.; Keim, U.; Gandini, S.; Amaral, T.; Katalinic, A.; Hollezcek, B.; Martus, P.; Flatz, L.; Leiter, U.; Whiteman, D. Epidemiology of cutaneous melanoma and keratinocyte cancer in white populations 1943–2036. Eur. J. Cancer 2021, 152, 18–25. [Google Scholar] [CrossRef] [PubMed]
- Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer Statistics. CA Cancer J. Clin. 2021, 71, 7–33. [Google Scholar] [CrossRef]
- Davies, H.; Bignell, G.R.; Cox, C.; Stephens, P.; Edkins, S.; Clegg, S.; Teague, J.; Woffendin, H.; Garnett, M.J.; Bottomley, W.; et al. Mutations of the BRAF gene in human cancer. Nature 2002, 417, 949–954. [Google Scholar] [CrossRef]
- Krummel, M.F.; Allison, J. CD28 and CTLA-4 have opposing effects on the response of T cells to stimulation. J. Exp. Med. 1995, 182, 459–465. [Google Scholar] [CrossRef] [Green Version]
- Leach, D.R.; Krummel, M.F.; Allison, J.P. Enhancement of Antitumor Immunity by CTLA-4 Blockade. Science 1996, 271, 1734–1736. [Google Scholar] [CrossRef] [Green Version]
- Snyder, A.; Makarov, V.; Merghoub, T.; Yuan, J.; Zaretsky, J.M.; Desrichard, A.; Walsh, L.A.; Postow, M.A.; Wong, P.; Ho, T.S.; et al. Genetic Basis for Clinical Response to CTLA-4 Blockade in Melanoma. N. Engl. J. Med. 2014, 371, 2189–2199. [Google Scholar] [CrossRef] [Green Version]
- Ishida, Y.; Agata, Y.; Shibahara, K.; Honjo, T. Induced expression of PD-1, a novel member of the immunoglobulin gene super-family, upon programmed cell death. EMBO J. 1992, 11, 3887–3895. [Google Scholar] [CrossRef] [PubMed]
- Blank, C.; Brown, I.; Peterson, A.C.; Spiotto, M.; Iwai, Y.; Honjo, T.; Gajewski, T.F. PD-L1/B7H-1 Inhibits the Effector Phase of Tumor Rejection by T Cell Receptor (TCR) Transgenic CD8+ T Cells. Cancer Res. 2004, 64, 1140–1145. [Google Scholar] [CrossRef] [Green Version]
- Chapman, P.B.; Hauschild, A.; Robert, C.; Haanen, J.B.; Ascierto, P.; Larkin, J.; Dummer, R.; Garbe, C.; Testori, A.; Maio, M.; et al. Improved Survival with Vemurafenib in Melanoma with BRAF V600E Mutation. N. Engl. J. Med. 2011, 364, 2507–2516. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Paraiso, K.H.T.; Fedorenko, I.V.; Cantini, L.P.; Munko, A.C.; Hall, M.; Sondak, V.K.; Messina, J.L.; Flaherty, K.T.; Smalley, K.S.M. Recovery of phospho-ERK activity allows melanoma cells to escape from BRAF inhibitor therapy. Br. J. Cancer 2010, 102, 1724–1730. [Google Scholar] [CrossRef] [PubMed]
- Robert, C.; Karaszewska, B.; Schachter, J.; Rutkowski, P.; Mackiewicz, A.; Stroiakovski, D.; Lichinitser, M.; Dummer, R.; Grange, F.; Mortier, L.; et al. Improved Overall Survival in Melanoma with Combined Dabrafenib and Trametinib. N. Engl. J. Med. 2015, 372, 30–39. [Google Scholar] [CrossRef] [Green Version]
- Larkin, J.; Ascierto, P.A.; Dréno, B.; Atkinson, V.; Liszkay, G.; Maio, M.; Mandalà, M.; Demidov, L.; Stroyakovskiy, D.; Thomas, L.; et al. Combined Vemurafenib and Cobimetinib in BRAF-Mutated Melanoma. N. Engl. J. Med. 2014, 371, 1867–1876. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dummer, R.; Ascierto, P.A.; Gogas, H.J.; Arance, A.; Mandala, M.; Liszkay, G.; Garbe, C.; Schadendorf, D.; Krajsova, I.; Gutzmer, R.; et al. Encorafenib plus binimetinib versus vemurafenib or encorafenib in patients with BRAF -mutant melanoma (COLUMBUS): A multicentre, open-label, randomised phase 3 trial. Lancet Oncol. 2018, 19, 603–615. [Google Scholar] [CrossRef] [Green Version]
- Schachter, J.; Ribas, A.; Long, G.V.; Arance, A.; Grob, J.-J.; Mortier, L.; Daud, A.; Carlino, M.S.; McNeil, C.; Lotem, M.; et al. Pembrolizumab versus ipilimumab for advanced melanoma: Final overall survival results of a multicentre, randomised, open-label phase 3 study (KEYNOTE-006). Lancet 2017, 390, 1853–1862. [Google Scholar] [CrossRef]
- Robert, C.; Long, G.V.; Brady, B.; Dutriaux, C.; Maio, M.; Mortier, L.; Hassel, J.C.; Rutkowski, P.; McNeil, C.; Kalinka-Warzocha, E.; et al. Nivolumab in Previously Untreated Melanoma without BRAF Mutation. N. Engl. J. Med. 2015, 372, 320–330. [Google Scholar] [CrossRef] [Green Version]
- Weber, J.S.; D’Angelo, S.P.; Minor, D.; Hodi, F.S.; Gutzmer, R.; Neyns, B.; Hoeller, C.; Khushalani, N.I.; Miller, W.H.; Lao, C.D.; et al. Nivolumab versus chemotherapy in patients with advanced melanoma who progressed after anti-CTLA-4 treatment (CheckMate 037): A randomised, controlled, open-label, phase 3 trial. Lancet Oncol. 2015, 16, 375–384. [Google Scholar] [CrossRef]
- Wolchok, J.D.; Chiarion-Sileni, V.; Gonzalez, R.; Rutkowski, P.; Grob, J.-J.; Cowey, C.L.; Lao, C.D.; Wagstaff, J.; Schadendorf, D.; Ferrucci, P.F.; et al. Overall Survival with Combined Nivolumab and Ipilimumab in Advanced Melanoma. N. Engl. J. Med. 2017, 377, 1345–1356. [Google Scholar] [CrossRef] [PubMed]
- Robert, C.; Grob, J.J.; Stroyakovskiy, D.; Karaszewska, B.; Hauschild, A.; Levchenko, E.; Sileni, V.C.; Schachter, J.; Garbe, C.; Bondarenko, I.; et al. Five-Year Outcomes with Dabrafenib plus Trametinib in Metastatic Melanoma. N. Engl. J. Med. 2019, 381, 626–636. [Google Scholar] [CrossRef] [PubMed]
- Larkin, J.; Chiarion-Sileni, V.; Gonzalez, R.; Grob, J.-J.; Rutkowski, P.; Lao, C.D.; Cowey, C.L.; Schadendorf, D.; Wagstaff, J.; Dummer, R.; et al. Five-Year Survival with Combined Nivolumab and Ipilimumab in Advanced Melanoma. N. Engl. J. Med. 2019, 381, 1535–1546. [Google Scholar] [CrossRef] [Green Version]
- Hamid, O.; Robert, C.; Daud, A.; Hodi, F.S.; Hwu, W.J.; Kefford, R.; Wolchok, J.D.; Hersey, P.; Joseph, R.; Weber, J.S.; et al. Five-year survival outcomes for patients with advanced melanoma treated with pembrolizumab in KEYNOTE. Ann. Oncol. 2019, 30, 582–588. [Google Scholar] [CrossRef] [PubMed]
- Robert, C.; Long, G.V.; Brady, B.; Dutriaux, C.; di Giacomo, A.M.; Mortier, L.; Rutkowski, P.; Hassel, J.C.; McNeil, C.M.; Kalinka, E.A.; et al. Five-Year Outcomes With Nivolumab in Patients With Wild-Type BRAF Advanced Melanoma. J. Clin. Oncol. 2020, 38, 3937–3946. [Google Scholar] [CrossRef] [PubMed]
- Lawrence, M.S.; Stojanov, P.; Polak, P.; Kryukov, G.V.; Cibulskis, K.; Sivachenko, A.; Carter, S.L.; Stewart, C.; Mermel, C.H.; Roberts, S.A.; et al. Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nat. Cell Biol. 2013, 499, 214–218. [Google Scholar] [CrossRef] [PubMed]
- Hodis, E.; Watson, I.R.; Kryukov, G.V.; Arold, S.T.; Imielinski, M.; Theurillat, J.-P.; Nickerson, E.; Auclair, D.; Li, L.; Place, C.; et al. A Landscape of Driver Mutations in Melanoma. Cell 2012, 150, 251–263. [Google Scholar] [CrossRef] [Green Version]
- Akbani, R.; Akdemir, K.C.; Aksoy, B.A.; Albert, M.; Ally, A.; Amin, S.; Arachchi, H.; Arora, A.; Auman, J.T.; Ayala, B.; et al. Genomic Classification of Cutaneous Melanoma. Cell 2015, 161, 1681–1696. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Raaijmakers, M.I.G.; Widmer, D.S.; Narechania, A.; Eichhoff, O.; Freiberger, S.N.; Wenzina, J.; Cheng, P.; Mihic-Probst, D.; DeSalle, R.; Dummer, R.; et al. Co-existence of BRAF and NRAS driver mutations in the same melanoma cells results in heterogeneity of targeted therapy resistance. Oncotarget 2016, 7, 77163–77174. [Google Scholar] [CrossRef] [Green Version]
- The ICGC/TCGA Pan-Cancer Analysis of Whole Genomes Consortium. Pan-cancer analysis of whole genomes. Nature 2020, 578, 82–93. [Google Scholar] [CrossRef] [Green Version]
- Zehir, A.; Benayed, R.; Shah, R.H.; Syed, A.; Middha, S.; Kim, H.R.; Srinivasan, P.; Gao, J.; Chakravarty, D.; Devlin, S.M.; et al. Mutational landscape of metastatic cancer revealed from prospective clinical sequencing of 10,000 patients. Nat. Med. 2017, 23, 703–713. [Google Scholar] [CrossRef] [PubMed]
- Yeh, I.; Jorgenson, E.; Shen, L.; Xu, M.; North, J.P.; Shain, A.H.; Reuss, D.; Wu, H.; Robinson, W.; Olshen, A.; et al. Targeted Genomic Profiling of Acral Melanoma. J. Natl. Cancer Inst. 2019, 111, 1068–1077. [Google Scholar] [CrossRef] [PubMed]
- Vanni, I.; Tanda, E.T.; Dalmasso, B.; Pastorino, L.; Andreotti, V.; Bruno, W.; Boutros, A.; Spagnolo, F.; Ghiorzo, P. Non-BRAF Mutant Melanoma: Molecular Features and Therapeutical Implications. Front. Mol. Biosci. 2020, 7, 172. [Google Scholar] [CrossRef] [PubMed]
- Uribe, P.; Wistuba, I.I.; González, S. BRAF Mutation. Am. J. Dermatopathol. 2003, 25, 365–370. [Google Scholar] [CrossRef] [PubMed]
- Bauer, J.; Curtin, J.; Pinkel, D.; Bastian, B.C. Congenital Melanocytic Nevi Frequently Harbor NRAS Mutations but no BRAF Mutations. J. Investig. Dermatol. 2007, 127, 179–182. [Google Scholar] [CrossRef]
- Zhang, T.; Dutton-Regester, K.; Brown, K.M.; Hayward, N.K. The genomic landscape of cutaneous melanoma. Pigment. Cell Melanoma Res. 2016, 29, 266–283. [Google Scholar] [CrossRef]
- Besmer, P.; Murphy, J.E.; George, P.C.; Qiu, F.; Bergold, P.; Lederman, L.; Snyder, H.W., Jr.; Brodeur, D.; Zuckerman, E.E.; Hardy, W.D. A new acute transforming feline retrovirus and relationship of its oncogene v-kit with the protein kinase gene family. Nat. Cell Biol. 1986, 320, 415–421. [Google Scholar] [CrossRef]
- Yarden, Y.; Kuang, W.J.; Yang-Feng, T.; Coussens, L.; Munemitsu, S.; Dull, T.J.; Chen, E.; Schlessinger, J.; Francke, U.; Ullrich, A. Human proto-oncogene c-kit: A new cell surface receptor tyrosine kinase for an unidentified ligand. EMBO J. 1987, 6, 3341–3351. [Google Scholar] [CrossRef]
- Qiu, F.H.; Ray, P.; Brown, K.; Barker, P.E.; Jhanwar, S.; Ruddle, F.H.; Besmer, P. Primary structure of c-kit: Relationship with the CSF-1/PDGF receptor kinase family—oncogenic activation of v-kit involves deletion of extracellular domain and C terminus. EMBO J. 1988, 7, 1003–1011. [Google Scholar] [CrossRef]
- Ullrich, A.; Schlessinger, J. Signal transduction by receptors with tyrosine kinase activity. Cell 1990, 61, 203–212. [Google Scholar] [CrossRef]
- Zhang, Z.; Zhang, R.; Joachimiak, A.; Schlessinger, J.; Kong, X.-P. Crystal structure of human stem cell factor: Implication for stem cell factor receptor dimerization and activation. Proc. Natl. Acad. Sci. USA 2000, 97, 7732–7737. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Huang, E.; Nocka, K.; Beier, D.R.; Chu, T.-Y.; Buck, J.; Lahm, H.-W.; Wellner, D.; Leder, P.; Besmer, P. The hematopoietic growth factor KL is encoded by the SI locus and is the ligand of the c-kit receptor, the gene product of the W locus. Cell 1990, 63, 225–233. [Google Scholar] [CrossRef]
- Lemmon, M.; Pinchasi, D.; Zhou, M.; Lax, I.; Schlessinger, J. Kit Receptor Dimerization Is Driven by Bivalent Binding of Stem Cell Factor. J. Biol. Chem. 1997, 272, 6311–6317. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Verstraete, K.; Savvides, S.N. Extracellular assembly and activation principles of oncogenic class III receptor tyrosine kinases. Nat. Rev. Cancer 2012, 12, 753–766. [Google Scholar] [CrossRef] [PubMed]
- Linnekin, D. Early signaling pathways activated by c-Kit in hematopoietic cells. Int. J. Biochem. Cell Biol. 1999, 31, 1053–1074. [Google Scholar] [CrossRef] [Green Version]
- Kitamura, Y.; Hirotab, S. Kit as a Human Oncogenic tyrosine kinase. Cell. Mol. Life Sci. 2004, 61, 2924–2931. [Google Scholar] [CrossRef]
- Pham, D.M.; Guhan, S.; Tsao, H. KIT and Melanoma: Biological Insights and Clinical Implications. Yonsei Med. J. 2020, 61, 562–571. [Google Scholar] [CrossRef]
- Willmore-Payne, C.; Holden, J.A.; Tripp, S.; Layfield, L.J. Human malignant melanoma: Detection of BRAF- and c-kit–activating mutations by high-resolution amplicon melting analysis. Hum. Pathol. 2005, 36, 486–493. [Google Scholar] [CrossRef]
- Went, P.T.; Dirnhofer, S.; Bundi, M.; Mirlacher, M.; Schraml, P.; Mangialaio, S.; Dimitrijevic, S.; Kononen, J.; Lugli, A.; Simon, R.; et al. Prevalence of KIT Expression in Human Tumors. J. Clin. Oncol. 2004, 22, 4514–4522. [Google Scholar] [CrossRef]
- Beadling, C.; Jacobson-Dunlop, E.; Hodi, F.S.; Le, C.; Warrick, A.; Patterson, J.; Town, A.; Harlow, A.; Cruz, F.; Azar, S.; et al. KIT Gene Mutations and Copy Number in Melanoma Subtypes. Clin. Cancer Res. 2008, 14, 6821–6828. [Google Scholar] [CrossRef] [Green Version]
- Curtin, J.A.; Busam, K.; Pinkel, D.; Bastian, B.C. Somatic Activation of KIT in Distinct Subtypes of Melanoma. J. Clin. Oncol. 2006, 24, 4340–4346. [Google Scholar] [CrossRef]
- Kong, Y.; Si, L.; Zhu, Y.; Xu, X.; Corless, C.L.; Flaherty, K.T.; Li, L.; Li, H.; Sheng, X.; Cui, C.; et al. Large-Scale Analysis of KIT Aberrations in Chinese Patients with Melanoma. Clin. Cancer Res. 2011, 17, 1684–1691. [Google Scholar] [CrossRef] [Green Version]
- Hayward, N.K.; Wilmott, J.S.; Waddell, N.; Johansson, P.A.; Field, M.A.; Nones, K.; Patch, A.-M.; Kakavand, H.; Alexandrov, L.B.; Burke, H.; et al. Whole-genome landscapes of major melanoma subtypes. Nature 2017, 545, 175–180. [Google Scholar] [CrossRef] [PubMed]
- Jenkins, R.W.; Fisher, D.E. Treatment of Advanced Melanoma in 2020 and Beyond. J. Investig. Dermatol. 2021, 141, 23–31. [Google Scholar] [CrossRef] [PubMed]
- McKean, M.; Oba, J.; Ma, J.; Roth, K.G.; Wang, W.-L.; Macedo, M.P.; Carapeto, F.C.L.; Haydu, L.E.; Siroy, A.E.; Vo, P.; et al. Tyrosine Kinase Inhibitor and Immune Checkpoint Inhibitor Responses in KIT-Mutant Metastatic Melanoma. J. Investig. Dermatol. 2019, 139, 728–731. [Google Scholar] [CrossRef] [Green Version]
- Huang, L.; Jiang, S.; Shi, Y. Tyrosine kinase inhibitors for solid tumors in the past 20 years (2001–2020). J. Hematol. Oncol. 2020, 13, 143. [Google Scholar] [CrossRef] [PubMed]
- Wyman, K.; Atkins, M.B.; Prieto, V.; Eton, O.; McDermott, D.F.; Hubbard, F.; Byrnes, C.; Sanders, K.; Sosman, J.A. Multicenter Phase II trial of high-dose imatinib mesylate in metastatic melanoma: Significant Toxicity with No Clinical Efficacy. Cancer 2006, 106, 2005–2011. [Google Scholar] [CrossRef] [PubMed]
- Ugurel, S.; Hildenbrand, R.; Zimpfer, A.; La Rosée, P.; Paschka, P.; Sucker, A.; Keikavoussi, P.; Becker, J.C.; Rittgen, W.; Hochhaus, A.; et al. Lack of clinical efficacy of imatinib in metastatic melanoma. Br. J. Cancer 2005, 92, 1398–1405. [Google Scholar] [CrossRef] [Green Version]
- Kim, K.B.; Eton, O.; Davis, D.W.; Frazier, M.L.; McConkey, D.J.; Diwan, A.H.; Papadopoulos, N.E.; Bedikian, A.Y.; Camacho, L.H.; Ross, M.I.; et al. Phase II trial of imatinib mesylate in patients with metastatic melanoma. Br. J. Cancer 2008, 99, 734–740. [Google Scholar] [CrossRef]
- Carvajal, R.D.; Antonescu, C.R.; Wolchok, J.D.; Chapman, P.B.; Roman, R.-A.; Teitcher, J.; Panageas, K.S.; Busam, K.J.; Chmielowski, B.; Lutzky, J.; et al. KIT as a Therapeutic Target in Metastatic Melanoma. JAMA 2011, 305, 2327–2334. [Google Scholar] [CrossRef] [Green Version]
- Guo, J.; Si, L.; Kong, Y.; Flaherty, K.T.; Xu, X.; Zhu, Y.; Corless, C.L.; Li, L.; Li, H.; Sheng, X.; et al. Phase II, Open-Label, Single-Arm Trial of Imatinib Mesylate in Patients with Metastatic Melanoma Harboring c-Kit Mutation or Amplification. J. Clin. Oncol. 2011, 29, 2904–2909. [Google Scholar] [CrossRef] [PubMed]
- Hodi, F.S.; Corless, C.L.; Giobbie-Hurder, A.; Fletcher, J.A.; Zhu, M.; Marino-Enriquez, A.; Friedlander, P.; Gonzalez, R.; Weber, J.S.; Gajewski, T.F.; et al. Imatinib for Melanomas Harboring Mutationally Activated or Amplified KIT Arising on Mucosal, Acral, and Chronically Sun-Damaged Skin. J. Clin. Oncol. 2013, 31, 3182–3190. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wei, X.; Mao, L.; Chi, Z.; Sheng, X.; Cui, C.; Kong, Y.; Dai, J.; Wang, X.; Li, S.; Tang, B.; et al. Efficacy Evaluation of Imatinib for the Treatment of Melanoma: Evidence from a Retrospective Study. Oncol. Res. 2019, 27, 495–501. [Google Scholar] [CrossRef] [PubMed]
- Cho, J.H.; Kim, K.M.; Kwon, M.; Kim, J.H.; Lee, J. Nilotinib in patients with metastatic melanoma harboring KIT gene aberration. Investig. New Drugs 2012, 30, 2008–2014. [Google Scholar] [CrossRef]
- Carvajal, R.D.; Lawrence, D.P.; Weber, J.S.; Gajewski, T.F.; Gonzalez, R.; Lutzky, J.; O’Day, S.J.; Hamid, O.; Wolchok, J.D.; Chapman, P.B.; et al. Phase II Study of Nilotinib in Melanoma Harboring KIT Alterations Following Progression to Prior KIT Inhibition. Clin. Cancer Res. 2015, 21, 2289–2296. [Google Scholar] [CrossRef] [Green Version]
- Lee, S.J.; Kim, T.M.; Kim, Y.J.; Jang, K.-T.; Lee, H.J.; Lee, S.N.; Ahn, M.S.; Hwang, I.G.; Lee, S.; Lee, M.-H.; et al. Phase II Trial of Nilotinib in Patients with Metastatic Malignant Melanoma Harboring KIT Gene Aberration: A Multicenter Trial of Korean Cancer Study Group (UN10-06). Oncologist 2015, 20, 1312–1319. [Google Scholar] [CrossRef] [Green Version]
- Guo, J.; Carvajal, R.D.; Dummer, R.; Hauschild, A.; Daud, A.; Bastian, B.C.; Markovic, S.N.; Queirolo, P.; Arance, A.; Berking, C.; et al. Efficacy and safety of nilotinib in patients with KIT-mutated metastatic or inoperable melanoma: Final results from the global, single-arm, phase II TEAM trial. Ann. Oncol. 2017, 28, 1380–1387. [Google Scholar] [CrossRef]
- Delyon, J.; Chevret, S.; Jouary, T.; Dalac, S.; Dalle, S.; Guillot, B.; Arnault, J.-P.; Avril, M.-F.; Bedane, C.; Bens, G.; et al. STAT3 Mediates Nilotinib Response in KIT-Altered Melanoma: A Phase II Multicenter Trial of the French Skin Cancer Network. J. Investig. Dermatol. 2018, 138, 58–67. [Google Scholar] [CrossRef] [Green Version]
- Kalinsky, K.; Lee, S.; Rubin, K.; Lawrence, D.P.; Iafrarte, A.J.; Borger, D.R.; Margolin, K.A.; Leitao, M.M.; Tarhini, A.A.; Koon, H.B.; et al. A Phase II Trial of Dasatinib in Patients with Locally Advanced or Stage IV Mucosal, Acral and Vulvovaginal Melanoma: A Trial of the ECOG-ACRIN Cancer Research Group (E2607). Cancer 2017, 123, 2688–2697. [Google Scholar] [CrossRef]
- Minor, D.R.; Kashani-Sabet, M.; Garrido, M.; O’Day, S.J.; Hamid, O.; Bastian, B.C. Sunitinib Therapy for Melanoma Patients with KIT Mutations. Clin. Cancer Res. 2012, 18, 1457–1463. [Google Scholar] [CrossRef] [Green Version]
- Kluger, H.M.; Dudek, A.Z.; McCann, C.; Ritacco, J.; Southard, N.; Jilaveanu, L.B.; Molinaro, A.; Sznol, M. A phase II trial of dasatinib in advanced melanoma. Cancer 2011, 117, 2202–2208. [Google Scholar] [CrossRef]
- Quintás-Cardama, A.; Lazar, A.J.; Woodman, S.E.; Kim, K.; Ross, M.; Hwu, P.; Quint, A. Complete response of stage IV anal mucosal melanoma expressing KIT Val560Asp to the multikinase inhibitor sorafenib. Nat. Clin. Pract. Oncol. 2008, 5, 737–740. [Google Scholar] [CrossRef]
- Prosvicova, J.; Lukesova, S.; Kopecky, J.; Grim, J.; Papik, Z.; Kolarova, R.; Navratilova, B.; Dubreuil, P.; Agopian, J.; Mansfield, C.D.; et al. Rapid and clinically significant response to masitinib in the treatment of mucosal primary esophageal melanoma with somatic KIT exon 11 mutation involving brain metastases: A case report. Biomed. Pap. 2015, 159, 695–697. [Google Scholar] [CrossRef] [PubMed]
- Oba, J.; Kim, S.-H.; Wang, W.-L.; Macedo, M.P.; Carapeto, F.; Mckean, M.A.; van Arnam, J.; Eterovic, A.K.; Sen, S.; Kale, C.R.; et al. Targeting the HGF/MET Axis Counters Primary Resistance to KIT Inhibition in KIT-Mutant Melanoma. JCO Precis. Oncol. 2018, 2, 1–8. [Google Scholar] [CrossRef] [PubMed]
- Han, Y.; Gu, Z.; Wu, J.; Huang, X.; Zhou, R.; Shi, C.; Tao, W.; Wang, L.; Wang, Y.; Zhou, G.; et al. Repurposing Ponatinib as a Potent Agent against KIT Mutant Melanomas. Theranostics 2019, 9, 1952–1964. [Google Scholar] [CrossRef]
- Flaherty, K.T.; Hamilton, B.K.; Rosen, M.A.; Amaravadi, R.K.; Schuchter, L.M.; Gallagher, M.; Chen, H.; Sehgal, C.; O’Dwyer, P.J. Phase I/II Trial of Imatinib and Bevacizumab in Patients with Advanced Melanoma and Other Advanced Cancers. Oncologist 2015, 20, 952–959. [Google Scholar] [CrossRef] [Green Version]
- Reilley, M.J.; Bailey, A.; Subbiah, V.; Janku, F.; Naing, A.; Falchook, G.; Karp, D.; Piha-Paul, S.; Tsimberidou, A.; Fu, S.; et al. Phase I clinical trial of combination imatinib and ipilimumab in patients with advanced malignancies. J. Immunother. Cancer 2017, 5, 35. [Google Scholar] [CrossRef]
- Abdou, Y.; Kapoor, A.; Hamad, L.; Ernstoff, M.S. Combination of pembrolizumab and imatinib in a patient with double KIT mutant melanoma. Medicine 2019, 98, e17769. [Google Scholar] [CrossRef] [Green Version]
- Padua, R.A.; Barrass, N.; Currie, G.A. A novel transforming gene in a human malignant melanoma cell line. Nat. Cell Biol. 1984, 311, 671–673. [Google Scholar] [CrossRef]
- Fedorenko, I.V.; Gibney, G.T.; Smalley, K.S.M. NRAS mutant melanoma: Biological behavior and future strategies for therapeutic management. Oncogene 2013, 32, 3009–3018. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yin, C.; Zhu, B.; Zhang, T.; Liu, T.; Chen, S.; Liu, Y.; Li, X.; Miao, X.; Li, S.; Mi, X.; et al. Pharmacological Targeting of STK19 Inhibits Oncogenic NRAS-Driven Melanomagenesis. Cell 2019, 176, 1113–1127.e16. [Google Scholar] [CrossRef] [Green Version]
- Curtin, J.A.; Fridlyand, J.; Kageshita, T.; Patel, H.N.; Busam, K.J.; Kutzner, H.; Cho, K.-H.; Aiba, S.; Bröcker, E.-B.; LeBoit, P.E.; et al. Distinct Sets of Genetic Alterations in Melanoma. N. Engl. J. Med. 2005, 353, 2135–2147. [Google Scholar] [CrossRef] [PubMed]
- Jakob, J.A.; Bassett, R.L.; Ng, C.S.; Curry, J.L.; Joseph, R.; Alvarado, G.C.; Apn, M.L.R.; Richard, J.; Gershenwald, J.E.; Kim, K.B.; et al. NRAS mutation status is an independent prognostic factor in metastatic melanoma. Cancer 2012, 118, 4014–4023. [Google Scholar] [CrossRef]
- Akslen, L.A.; Angelini, S.; Straume, O.; Bachmann, I.M.; Molven, A.; Hemminki, K.; Kumar, R. BRAF and NRAS Mutations Are Frequent in Nodular Melanoma but Are not Associated with Tumor Cell Proliferation or Patient Survival. J. Investig. Dermatol. 2005, 125, 312–317. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ellerhorst, J.A.; Greene, V.R.; Ekmekcioglu, S.; Warneke, C.L.; Johnson, M.M.; Cooke, C.P.; Wang, L.-E.; Prieto, V.G.; Gershenwald, J.E.; Wei, Q.; et al. Clinical Correlates ofNRASandBRAFMutations in Primary Human Melanoma. Clin. Cancer Res. 2010, 17, 229–235. [Google Scholar] [CrossRef] [Green Version]
- Moore, A.R.; Rosenberg, S.C.; McCormick, F.; Malek, S. RAS-targeted therapies: Is the undruggable drugged? Nat. Rev. Drug Discov. 2020, 19, 533–552. [Google Scholar] [CrossRef]
- Burd, C.E.; Liu, W.; Huynh, M.V.; Waqas, M.A.; Gillahan, J.E.; Clark, K.S.; Fu, K.; Martin, B.L.; Jeck, W.R.; Souroullas, G.P.; et al. Mutation-Specific RAS Oncogenicity Explains NRAS Codon 61 Selection in Melanoma. Cancer Discov. 2014, 4, 1418–1429. [Google Scholar] [CrossRef] [Green Version]
- Kirchberger, M.C.; Ugurel, S.; Mangana, J.; Heppt, M.V.; Eigentler, T.K.; Berking, C.; Schadendorf, D.; Schuler, G.; Dummer, R.; Heinzerling, L. MEK inhibition may increase survival of NRAS-mutated melanoma patients treated with checkpoint blockade: Results of a retrospective multicentre analysis of 364 patients. Eur. J. Cancer 2018, 98, 10–16. [Google Scholar] [CrossRef] [PubMed]
- Sadetsky, N.; Lambert, P.; Julian, C.; Chen, J.; Yan, Y. Comprehensive genomic profiling and outcomes among metastatic melanoma patients (pts) treated with first-line cancer immunotherapy (CIT) in a real-world setting. Ann. Oncol. 2019, 30, vii2. [Google Scholar] [CrossRef]
- Mangana, J.; Cheng, P.F.; Schindler, K.; Weide, B.; Held, U.; Frauchiger, A.L.; Romano, E.; Kähler, K.C.; Rozati, S.; Rechsteiner, M.; et al. Analysis of BRAF and NRAS Mutation Status in Advanced Melanoma Patients Treated with Anti-CTLA-4 Antibodies: Association with Overall Survival? PLoS ONE 2015, 10, e0139438. [Google Scholar] [CrossRef] [Green Version]
- Means-Powell, J.A.; Adjei, A.A.; Puzanov, I.; Dy, G.K.; Goff, L.W.; Ma, W.W.; Fetterly, G.J.; Michael, S.A.; Chai, F.; Lamar, M.; et al. Safety and efficacy of MET inhibitor tivantinib (ARQ 197) combined with sorafenib in patients (pts) with NRAS wild-type or mutant melanoma from a phase I study. J. Clin. Oncol. 2012, 30, 8519. [Google Scholar] [CrossRef]
- Niessner, H.; Beck, D.; Sinnberg, T.; Lasithiotakis, K.; Maczey, E.; Gogel, J.; Venturelli, S.; Berger, A.; Mauthe, M.; Toulany, M.; et al. The Farnesyl Transferase Inhibitor Lonafarnib Inhibits mTOR Signaling and Enforces Sorafenib-Induced Apoptosis in Melanoma Cells. J. Investig. Dermatol. 2011, 131, 468–479. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gajewski, T.F.; Salama, A.K.; Niedzwiecki, D.; Johnson, J.; Linette, G.; Bucher, C.; Blaskovich, M.A.; Sebti, S.M.; Haluska, F. Phase II study of the farnesyltransferase inhibitor R115777 in advanced melanoma (CALGB 500104). J. Transl. Med. 2012, 10, 246. [Google Scholar] [CrossRef] [Green Version]
- Dorard, C.; Estrada, C.; Barbotin, C.; Larcher, M.; Garancher, A.; Leloup, J.; Beermann, F.; Baccarini, M.; Pouponnot, C.; LaRue, L.; et al. RAF proteins exert both specific and compensatory functions during tumour progression of NRAS-driven melanoma. Nat. Commun. 2017, 8, 15262. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dumaz, N.; Hayward, R.; Martin, J.; Ogilvie, L.; Hedley, D.; Curtin, J.A.; Bastian, B.C.; Springer, C.; Marais, R. In Melanoma, RAS Mutations are Accompanied by Switching Signaling from BRAF to CRAF and Disrupted Cyclic AMP Signaling. Cancer Res. 2006, 66, 9483–9491. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Solit, D.B.; Garraway, L.A.; Pratilas, C.A.; Sawai, A.; Getz, G.; Basso, A.; Ye, Q.; Lobo, J.M.; She, Y.; Osman, I.; et al. BRAF mutation predicts sensitivity to MEK inhibition. Nat. Cell Biol. 2005, 439, 358–362. [Google Scholar] [CrossRef] [Green Version]
- Kim, T.W.; Lee, J.; Shin, S.J.; Kim, J.-S.; Kim, Y.J.; Han, H.S.; Lee, S.J.; Lim, H.-S.; Hong, Y.-H.; Noh, Y.S.; et al. Belvarafenib, a novel pan-RAF inhibitor, in solid tumor patients harboring BRAF, KRAS, or NRAS mutations: Phase I study. J. Clin. Oncol. 2019, 37, 3000. [Google Scholar] [CrossRef]
- Atefi, M.; Titz, B.; Avramis, E.; Ng, C.; Wong, D.J.; Lassen, A.; Cerniglia, M.; Escuin-Ordinas, H.; Foulad, D.; Comin-Anduix, B.; et al. Combination of pan-RAF and MEK inhibitors in NRAS mutant melanoma. Mol. Cancer 2015, 14, 27. [Google Scholar] [CrossRef] [Green Version]
- Kirkwood, J.M.; Bastholt, L.; Robert, C.; Sosman, J.A.; Larkin, J.; Hersey, P.; Middleton, M.R.; Cantarini, M.V.; Zazulina, V.; Kemsley, K.; et al. Phase II, Open-Label, Randomized Trial of the MEK1/2 Inhibitor Selumetinib as Monotherapy versus Temozolomide in Patients with Advanced Melanoma. Clin. Cancer Res. 2012, 18, 555–567. [Google Scholar] [CrossRef] [Green Version]
- Gupta, A.; Love, S.; Schuh, A.; Shanyinde, M.; Larkin, J.M.; Plummer, R.; Nathan, P.D.; Danson, S.; Ottensmeier, C.H.; Lorigan, P.; et al. DOC-MEK: A double-blind randomized phase II trial of docetaxel with or without selumetinib in wild-type BRAF advanced melanoma. Ann. Oncol. 2014, 25, 968–974. [Google Scholar] [CrossRef]
- Dummer, R.; Schadendorf, D.; Ascierto, P.A.; Arance, A.; Dutriaux, C.; di Giacomo, A.M.; Rutkowski, P.; del Vecchio, M.; Gutzmer, R.; Mandala, M.; et al. Binimetinib versus dacarbazine in patients with advanced NRAS-mutant melanoma (NEMO): A multicentre, open-label, randomised, phase 3 trial. Lancet Oncol. 2017, 18, 435–445. [Google Scholar] [CrossRef]
- Lebbé, C.; Dutriaux, C.; Lesimple, T.; Kruit, W.; Kerger, J.; Thomas, L.; Guillot, B.; de Braud, F.; Garbe, C.; Grob, J.-J.; et al. Pimasertib Versus Dacarbazine in Patients with Unresectable NRAS-Mutated Cutaneous Melanoma: Phase II, Randomized, Controlled Trial with Crossover. Cancers 2020, 12, 1727. [Google Scholar] [CrossRef] [PubMed]
- Greger, J.G.; Eastman, S.D.; Zhang, V.; Bleam, M.R.; Hughes, A.M.; Smitheman, K.N.; Dickerson, S.H.; Laquerre, S.G.; Liu, L.; Gilmer, T.M. Combinations of BRAF, MEK, and PI3K/mTOR Inhibitors Overcome Acquired Resistance to the BRAF Inhibitor GSK2118436 Dabrafenib, Mediated by NRAS or MEK Mutations. Mol. Cancer Ther. 2012, 11, 909–920. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Posch, C.; Moslehi, H.; Feeney, L.; Green, G.A.; Ebaee, A.; Feichtenschlager, V.; Chong, K.; Peng, L.; Dimon, M.T.; Phillips, T.; et al. Combined targeting of MEK and PI3K/mTOR effector pathways is necessary to effectively inhibit NRAS mutant melanoma in vitro and in vivo. Proc. Natl. Acad. Sci. USA 2013, 110, 4015–4020. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Algazi, A.P.; Esteve-Puig, R.; Nosrati, A.; Hinds, B.; Hobbs-Muthukumar, A.; Nandoskar, P.; Ortiz-Urda, S.; Chapman, P.B.; Daud, A. Dual MEK/AKT inhibition with trametinib and GSK2141795 does not yield clinical benefit in metastatic NRAS-mutant and wild-type melanoma. Pigment. Cell Melanoma Res. 2018, 31, 110–114. [Google Scholar] [CrossRef] [PubMed]
- Garcia-Alvarez, A.; Ortiz, C.; Muñoz-Couselo, E. Current Perspectives and Novel Strategies of NRAS-Mutant Melanoma. OncoTargets Ther. 2021, 14, 3709–3719. [Google Scholar] [CrossRef]
- Sosman, J.A.; Kittaneh, M.; Lolkema, M.P.J.K.; Postow, M.A.; Schwartz, G.; Franklin, C.; Matano, A.; Bhansali, S.; Parasuraman, S.; Kim, K. A phase 1b/2 study of LEE011 in combination with binimetinib (MEK162) in patients with NRAS-mutant melanoma: Early encouraging clinical activity. J. Clin. Oncol. 2014, 32, 9009. [Google Scholar] [CrossRef]
- Vogel, C.J.; Smit, M.A.; Maddalo, G.; Possik, P.A.; Sparidans, R.W.; van der Burg, S.H.; Verdegaal, E.M.; Heck, A.J.R.; Samatar, A.A.; Beijnen, J.H.; et al. Cooperative induction of apoptosis in NRAS mutant melanoma by inhibition of MEK and ROCK. Pigment Cell Melanoma Res. 2015, 28, 307–317. [Google Scholar] [CrossRef]
- Prior, I.A.; Hood, F.E.; Hartley, J.L. The Frequency of Ras Mutations in Cancer. Cancer Res. 2020, 80, 2969–2974. [Google Scholar] [CrossRef] [Green Version]
- Skoulidis, F.; Li, B.T.; Dy, G.K.; Price, T.J.; Falchook, G.S.; Wolf, J.; Italiano, A.; Schuler, M.; Borghaei, H.; Barlesi, F.; et al. Sotorasib for Lung Cancers with KRAS p.G12C Mutation. N. Engl. J. Med. 2021, 384, 2371–2381. [Google Scholar] [CrossRef]
- Hong, D.S.; Fakih, M.G.; Strickler, J.H.; Desai, J.; Durm, G.A.; Shapiro, G.I.; Falchook, G.S.; Price, T.J.; Sacher, A.; Denlinger, C.S.; et al. KRASG12C Inhibition with Sotorasib in Advanced Solid Tumors. N. Engl. J. Med. 2020, 383, 1207–1217. [Google Scholar] [CrossRef] [PubMed]
- Morris, L.G.T.; Chan, T.A. Therapeutic targeting of tumor suppressor genes. Cancer 2015, 121, 1357–1368. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kiuru, M.; Busam, K.J. The NF1 gene in tumor syndromes and melanoma. Lab. Investig. 2017, 97, 146–157. [Google Scholar] [CrossRef] [Green Version]
- Krauthammer, M.; Kong, Y.; Bacchiocchi, A.; Evans, P.; Pornputtapong, N.; Wu, C.; McCusker, J.; Ma, S.; Cheng, E.; Straub, R.; et al. Exome sequencing identifies recurrent mutations in NF1 and RASopathy genes in sun-exposed melanomas. Nat. Genet. 2015, 47, 996–1002. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wiesner, T.; Kiuru, M.; Scott, S.N.; Arcila, M.; Halpern, A.C.; Hollmann, T.; Berger, M.F.; Busam, K.J. NF1 Mutations Are Common in Desmoplastic Melanoma. Am. J. Surg. Pathol. 2015, 39, 1357–1362. [Google Scholar] [CrossRef] [Green Version]
- Nissan, M.H.; Pratilas, C.A.; Jones, A.M.; Ramirez, R.; Won, H.; Liu, C.; Tiwari, S.; Kong, L.; Hanrahan, A.J.; Yao, Z.; et al. Loss of NF1 in Cutaneous Melanoma Is Associated with RAS Activation and MEK Dependence. Cancer Res. 2014, 74, 2340–2350. [Google Scholar] [CrossRef] [Green Version]
- Ranzani, M.; Alifrangis, C.; Perna, D.; Dutton-Regester, K.; Pritchard, A.; Wong, K.; Rashid, M.; Espinoza, C.D.R.; Hayward, N.; McDermott, U.; et al. BRAF / NRAS wild-type melanoma, NF 1 status and sensitivity to trametinib. Pigment. Cell Melanoma Res. 2015, 28, 117–119. [Google Scholar] [CrossRef] [Green Version]
- Py, C.; Christinat, Y.; Kreutzfeldt, M.; McKee, T.A.; Dietrich, P.-Y.; Tsantoulis, P. Response of NF1-Mutated Melanoma to an MEK Inhibitor. JCO Precis. Oncol. 2018, 2, 1–11. [Google Scholar] [CrossRef]
- Falchook, G.S.; Lewis, K.D.; Infante, J.R.; Gordon, M.S.; Vogelzang, N.J.; DeMarini, D.J.; Sun, P.; Moy, C.; Szabo, S.A.; Roadcap, L.T.; et al. Activity of the oral MEK inhibitor trametinib in patients with advanced melanoma: A phase 1 dose-escalation trial. Lancet Oncol. 2012, 13, 782–789. [Google Scholar] [CrossRef] [Green Version]
- Alon, M.; Arafeh, R.; Lee, J.S.; Madan, S.; Kalaora, S.; Nagler, A.; Abgarian, T.; Greenberg, P.; Ruppin, E.; Samuels, Y. CAPN1 is a novel binding partner and regulator of the tumor suppressor NF1 in melanoma. Oncotarget 2018, 9, 31264–31277. [Google Scholar] [CrossRef]
- Vogelstein, B.; Lane, D.; Levine, A.J. Surfing the p53 network. Nat. Cell Biol. 2000, 408, 307–310. [Google Scholar] [CrossRef]
- Liu, Y.; Chen, C.; Xu, Z.; Scuoppo, C.; Rillahan, C.D.; Gao, J.; Spitzer, B.; Bosbach, B.; Kastenhuber, E.R.; Baslan, T.; et al. Deletions linked to TP53 loss drive cancer through p53-independent mechanisms. Nat. Cell Biol. 2016, 531, 471–475. [Google Scholar] [CrossRef] [Green Version]
- Viros, A.; Sanchez-Laorden, B.; Pedersen, M.; Furney, S.; Rae, J.; Hogan, K.; Ejiama, S.; Girotti, M.R.; Cook, M.; Dhomen, N.; et al. Ultraviolet radiation accelerates BRAF-driven melanomagenesis by targeting TP53. Nat. Cell Biol. 2014, 511, 478–482. [Google Scholar] [CrossRef]
- Shadfan, M.; Lopez-Pajares, V.; Yuan, Z.-M. MDM2 and MDMX: Alone and together in regulation of p53. Transl. Cancer Res. 2012, 1, 88–89. [Google Scholar] [PubMed]
- Loureiro, J.; Abrantes, M.; Oliveira, P.; Saraiva, L. P53 in skin cancer: From a master player to a privileged target for prevention and therapy. Biochim. Biophys. Acta (BBA)-Bioenerg. 2020, 1874, 188438. [Google Scholar] [CrossRef] [PubMed]
- Gembarska, A.; Luciani, F.; Fedele, C.; Russell, E.A.; Dewaele, M.; Villar, S.; Zwolinska, A.; Haupt, S.; de Lange, J.; Yip, D.; et al. MDM4 is a key therapeutic target in cutaneous melanoma. Nat. Med. 2012, 18, 1239–1247. [Google Scholar] [CrossRef] [Green Version]
- Wu, C.-E.; Esfandiari, A.; Ho, Y.-H.; Wang, N.; Mahdi, A.; Aptullahoglu, E.; Lovat, P.; Lunec, J. Targeting negative regulation of p53 by MDM2 and WIP1 as a therapeutic strategy in cutaneous melanoma. Br. J. Cancer 2018, 118, 495–508. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lu, M.; Breyssens, H.; Salter, V.; Zhong, S.; Hu, Y.; Baer, C.; Ratnayaka, I.; Sullivan, A.; Brown, N.R.; Endicott, J.; et al. Restoring p53 Function in Human Melanoma Cells by Inhibiting MDM2 and Cyclin B1/CDK1-Phosphorylated Nuclear iASPP. Cancer Cell 2013, 23, 618–633. [Google Scholar] [CrossRef] [Green Version]
- Loureiro, J.; Raimundo, L.; Calheiros, J.; Carvalho, C.; Barcherini, V.; Lima, N.; Gomes, C.; Almeida, M.; Alves, M.; Costa, J.; et al. Targeting p53 for Melanoma Treatment: Counteracting Tumour Proliferation, Dissemination and Therapeutic Resistance. Cancers 2021, 13, 1648. [Google Scholar] [CrossRef]
- Moschos, S.J.; Sandhu, S.K.; Lewis, K.D.; Sullivan, R.J.; Johnson, D.B.; Zhang, Y.; Rasmussen, E.; Henary, H.A.; Long, G.V. Phase 1 study of the p53-MDM2 inhibitor AMG 232 combined with trametinib plus dabrafenib or trametinib in patients (Pts) with TP53 wild type (TP53WT) metastatic cutaneous melanoma (MCM). J. Clin. Oncol. 2017, 35, 2575. [Google Scholar] [CrossRef]
- Sallman, D.A.; DeZern, A.E.; Garcia-Manero, G.; Steensma, D.P.; Roboz, G.J.; Sekeres, M.A.; Cluzeau, T.; Sweet, K.L.; McLemore, A.; McGraw, K.L.; et al. Eprenetapopt (APR-246) and Azacitidine in TP53-Mutant Myelodysplastic Syndromes. J. Clin. Oncol. 2021, 39, 1584–1594. [Google Scholar] [CrossRef] [PubMed]
- Cluzeau, T.; Sebert, M.; Rahmé, R.; Cuzzubbo, S.; Lehmann-Che, J.; Madelaine, I.; Peterlin, P.; Bève, B.; Attalah, H.; Chermat, F.; et al. Eprenetapopt Plus Azacitidine in TP53-Mutated Myelodysplastic Syndromes and Acute Myeloid Leukemia: A Phase II Study by the Groupe Francophone des Myélodysplasies (GFM). J. Clin. Oncol. 2021, 39, 1575–1583. [Google Scholar] [CrossRef] [PubMed]
- Goldstein, A.M.; Chan, M.; Harland, M.; Gillanders, E.M.; Hayward, N.K.; Avril, M.-F.; Azizi, E.; Scarrà, G.B.; Bishop, D.T.; Paillerets, B.B.-D.; et al. High-risk Melanoma Susceptibility Genes and Pancreatic Cancer, Neural System Tumors, and Uveal Melanoma across GenoMEL. Cancer Res. 2006, 66, 9818–9828. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, S.; Ramsay, E.S.; Mock, B.A. Cdkn2a, the cyclin-dependent kinase inhibitor encoding p16INK4a and p19ARF, is a candidate for the plasmacytoma susceptibility locus, Pctr1. Proc. Natl. Acad. Sci. USA 1998, 95, 2429–2434. [Google Scholar] [CrossRef] [Green Version]
- Martin, C.A.; Cullinane, C.; Kirby, L.; AbuHammad, S.; Lelliott, E.J.; Waldeck, K.; Young, R.J.; Brajanovski, N.; Cameron, D.P.; Walker, R.; et al. Palbociclib synergizes with BRAF and MEK inhibitors in treatment naïve melanoma but not after the development of BRAF inhibitor resistance. Int. J. Cancer 2018, 142, 2139–2152. [Google Scholar] [CrossRef] [Green Version]
- Hayes, T.K.; Luo, F.; Cohen, O.; Goodale, A.B.; Lee, Y.; Pantel, S.; Bagul, M.; Piccioni, F.; Root, D.E.; Garraway, L.A.; et al. A Functional Landscape of Resistance to MEK1/2 and CDK4/6 Inhibition in NRAS-Mutant Melanoma. Cancer Res. 2019, 79, 2352–2366. [Google Scholar] [CrossRef] [Green Version]
- Kwong, L.N.; Costello, J.; Liu, H.; Jiang, S.; Helms, T.L.; Langsdorf, A.E.; Jakubosky, D.; Genovese, G.; Muller, F.; Jeong, J.H.; et al. Oncogenic NRAS signaling differentially regulates survival and proliferation in melanoma. Nat. Med. 2012, 18, 1503–1510. [Google Scholar] [CrossRef] [Green Version]
- Patnaik, A.; Rosen, L.S.; Tolaney, S.M.; Tolcher, A.W.; Goldman, J.W.; Gandhi, L.; Papadopoulos, K.P.; Beeram, M.; Rasco, D.W.; Hilton, J.F.; et al. Efficacy and Safety of Abemaciclib, an Inhibitor of CDK4 and CDK6, for Patients with Breast Cancer, Non-Small Cell Lung Cancer, and Other Solid Tumors. Cancer Discov. 2016, 6, 740–753. [Google Scholar] [CrossRef] [Green Version]
- Sahebjam, S.; Le Rhun, E.; Queirolo, P.; Jerusalem, G.; Subramaniam, D.; Bear, M.; Yang, Z.; Chen, Y.; Conte, P. A phase II study of abemaciclib in patients (pts) with brain metastases (BM) secondary to non-small cell lung cancer (NSCLC) or melanoma (MEL). Ann. Oncol. 2019, 30, v117. [Google Scholar] [CrossRef]
- Infante, J.R.; Cassier, P.A.; Gerecitano, J.F.; Witteveen, P.O.; Chugh, R.; Ribrag, V.; Chakraborty, A.; Matano, A.; Dobson, J.R.; Crystal, A.S.; et al. A Phase I Study of the Cyclin-Dependent Kinase 4/6 Inhibitor Ribociclib (LEE011) in Patients with Advanced Solid Tumors and Lymphomas. Clin. Cancer Res. 2016, 22, 5696–5705. [Google Scholar] [CrossRef] [Green Version]
- Mao, L.; Dai, J.; Cao, Y.; Bai, X.; Sheng, X.; Chi, Z.; Cui, C.; Kong, Y.; Zhang, Y.; Wu, L.; et al. Palbociclib in advanced acral melanoma with genetic aberrations in the cyclin-dependent kinase 4 pathway. Eur. J. Cancer 2021, 148, 297–306. [Google Scholar] [CrossRef] [PubMed]
- Garutti, M.; Targato, G.; Buriolla, S.; Palmero, L.; Minisini, A.; Puglisi, F. CDK4/6 Inhibitors in Melanoma: A Comprehensive Review. Cells 2021, 10, 1334. [Google Scholar] [CrossRef] [PubMed]
- Mertens, F.; Johansson, B.; Fioretos, T.; Mitelman, F. The emerging complexity of gene fusions in cancer. Nat. Rev. Cancer 2015, 15, 371–381. [Google Scholar] [CrossRef] [PubMed]
- Mitelman, F.; Johansson, B.; Mertens, F. The impact of translocations and gene fusions on cancer causation. Nat. Rev. Cancer 2007, 7, 233–245. [Google Scholar] [CrossRef] [PubMed]
- Wiesner, T.; He, J.; Yelensky, R.; Esteve-Puig, R.; Botton, T.; Yeh, I.; Lipson, D.; Otto, G.; Brennan, K.; Murali, R.; et al. Kinase fusions are frequent in Spitz tumours and spitzoid melanomas. Nat. Commun. 2014, 5, 3116. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Newman, S.; Fan, L.; Pribnow, A.; Silkov, A.; Rice, S.V.; Lee, S.; Shao, Y.; Shaner, B.; Mulder, H.; Nakitandwe, J.; et al. Clinical genome sequencing uncovers potentially targetable truncations and fusions of MAP3K8 in spitzoid and other melanomas. Nat. Med. 2019, 25, 597–602. [Google Scholar] [CrossRef]
- Niu, H.-T.; Zhou, Q.-M.; Wang, F.; Shao, Q.; Guan, Y.-X.; Wen, X.-Z.; Chen, L.-Z.; Feng, Q.-S.; Li, W.; Zeng, Y.-X.; et al. Identification of anaplastic lymphoma kinase break points and oncogenic mutation profiles in acral/mucosal melanomas. Pigment Cell Melanoma Res. 2013, 26, 646–653. [Google Scholar] [CrossRef]
- Busam, K.J.; Vilain, R.E.; Lum, T.; Busam, J.A.; Hollmann, T.J.; Saw, R.P.; Coit, D.C.; Scolyer, R.A.; Wiesner, T. Primary and Metastatic Cutaneous Melanomas Express ALK Through Alternative Transcriptional Initiation. Am. J. Surg. Pathol. 2016, 40, 786–795. [Google Scholar] [CrossRef] [Green Version]
- Turner, J.; Couts, K.; Sheren, J.; Saichaemchan, S.; Ariyawutyakorn, W.; Avolio, I.; Cabral, E.; Glogowska, M.; Amato, C.; Robinson, S.; et al. Kinase gene fusions in defined subsets of melanoma. Pigment Cell Melanoma Res. 2017, 30, 53–62. [Google Scholar] [CrossRef] [Green Version]
- Stransky, N.; Cerami, E.; Schalm, S.; Kim, J.L.; Lengauer, C. The landscape of kinase fusions in cancer. Nat. Commun. 2014, 5, 4846. [Google Scholar] [CrossRef] [Green Version]
- Lezcano, C.; Shoushtari, A.N.; Ariyan, C.; Hollmann, T.J.; Busam, K.J. Primary and Metastatic Melanoma with NTRK Fusions. Am. J. Surg. Pathol. 2018, 42, 1052–1058. [Google Scholar] [CrossRef]
- Drilon, A.; Laetsch, T.W.; Kummar, S.; Dubois, S.G.; Lassen, U.N.; Demetri, G.D.; Nathenson, M.; Doebele, R.C.; Farago, A.F.; Pappo, A.S.; et al. Efficacy of Larotrectinib inTRKFusion–Positive Cancers in Adults and Children. N. Engl. J. Med. 2018, 378, 731–739. [Google Scholar] [CrossRef]
- Hong, D.S.; DuBois, S.G.; Kummar, S.; Farago, A.F.; Albert, C.M.; Rohrberg, K.S.; van Tilburg, C.M.; Nagasubramanian, R.; Berlin, J.D.; Federman, N.; et al. Larotrectinib in patients with TRK fusion-positive solid tumours: A pooled analysis of three phase 1/2 clinical trials. Lancet Oncol. 2020, 21, 531–540. [Google Scholar] [CrossRef]
- Doebele, R.C.; Drilon, A.; Paz-Ares, L.; Siena, S.; Shaw, A.T.; Farago, A.F.; Blakely, C.M.; Seto, T.; Cho, B.C.; Tosi, D.; et al. Entrectinib in patients with advanced or metastatic NTRK fusion-positive solid tumours: Integrated analysis of three phase 1–2 trials. Lancet Oncol. 2020, 21, 271–282. [Google Scholar] [CrossRef]
- Couts, K.L.; McCoach, C.E.; Murphy, D.; Christiansen, J.; Turner, J.; Lewis, K.D.; Robinson, W.A.; Doebele, R.C. Acral Lentiginous Melanoma Harboring a ROS1 Gene Fusion With Clinical Response to Entrectinib. JCO Precis. Oncol. 2017, 1, 1–7. [Google Scholar] [CrossRef] [Green Version]
- Couts, K.L.; Bemis, J.; Turner, J.A.; Bagby, S.M.; Murphy, D.; Christiansen, J.; Hintzsche, J.D.; Danielle, M.; Pitts, T.M.; Wells, K.; et al. ALK Inhibitor Response in Melanomas Expressing EML4-ALK Fusions and Alternate ALK Isoforms. Mol. Cancer Ther. 2018, 17, 222–231. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sharma, S.; Kelly, T.K.; Jones, P.A. Epigenetics in cancer. Carcinogenesis 2010, 31, 27–36. [Google Scholar] [CrossRef] [PubMed]
- Bates, S.E. Epigenetic Therapies for Cancer. N. Engl. J. Med. 2020, 383, 650–663. [Google Scholar] [CrossRef] [PubMed]
- Duan, R.; Du, W.; Guo, W. EZH2: A novel target for cancer treatment. J. Hematol. Oncol. 2020, 13, 104. [Google Scholar] [CrossRef] [PubMed]
- Gao, J.; Aksoy, B.A.; Dogrusoz, U.; Dresdner, G.; Gross, B.; Sumer, S.O.; Sun, Y.; Jacobsen, A.; Sinha, R.; Larsson, E.; et al. Integrative Analysis of Complex Cancer Genomics and Clinical Profiles Using the cBioPortal. Sci. Signal. 2013, 6, pl1. [Google Scholar] [CrossRef] [Green Version]
- Tiffen, J.C.; Gallagher, S.J.; Tseng, H.-Y.; Filipp, F.V.; Groth, B.F.D.S.; Hersey, P. EZH2 as a mediator of treatment resistance in melanoma. Pigment Cell Melanoma Res. 2016, 29, 500–507. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zingg, D.; Debbache, J.; Schaefer, S.M.; Tuncer, E.; Frommel, S.C.; Cheng, P.; Arenas-Ramirez, N.; Haeusel, J.; Zhang, Y.; Bonalli, M.; et al. The epigenetic modifier EZH2 controls melanoma growth and metastasis through silencing of distinct tumour suppressors. Nat. Commun. 2015, 6, 6051. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bachmann, I.M.; Halvorsen, O.J.; Collett, K.; Stefansson, I.M.; Straume, O.; Haukaas, S.A.; Salvesen, H.B.; Otte, A.P.; Akslen, L.A. EZH2 expression is associated with high proliferation rate and aggressive tumor subgroups in cutaneous melanoma and cancers of the endometrium, prostate, and breast. J. Clin. Oncol. 2006, 24, 268–273. [Google Scholar] [CrossRef]
- Tiffen, J.C.; Gunatilake, D.; Gallagher, S.; Gowrishankar, K.; Heinemann, A.; Cullinane, C.; Dutton-Regester, K.; Pupo, G.M.; Strbenac, D.; Yang, J.; et al. Targeting activating mutations of EZH2 leads to potent cell growth inhibition in human melanoma by derepression of tumor suppressor genes. Oncotarget 2015, 6, 27023–27036. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Unruh, D.; Zewde, M.; Buss, A.; Drumm, M.R.; Tran, A.N.; Scholtens, D.M.; Horbinski, C. Methylation and transcription patterns are distinct in IDH mutant gliomas compared to other IDH mutant cancers. Sci. Rep. 2019, 9, 8946. [Google Scholar] [CrossRef] [PubMed]
- Dang, L.; White, D.W.; Gross, S.; Bennett, B.D.; Bittinger, M.A.; Driggers, E.M.; Fantin, V.R.; Jang, H.G.; Jin, S.; Keenan, M.C.; et al. Cancer-associated IDH1 mutations produce 2-hydroxyglutarate. Nature 2009, 462, 739–744. [Google Scholar] [CrossRef] [Green Version]
- Mellinghoff, I.K.; Ellingson, B.M.; Touat, M.; Maher, E.; De La Fuente, M.I.; Holdhoff, M.; Cote, G.M.; Burris, H.; Janku, F.; Young, R.J.; et al. Ivosidenib in Isocitrate Dehydrogenase 1–Mutated Advanced Glioma. J. Clin. Oncol. 2020, 38, 3398–3406. [Google Scholar] [CrossRef]
- Abou-Alfa, G.K.; Macarulla, T.; Javle, M.M.; Kelley, R.K.; Lubner, S.J.; Adeva, J.; Cleary, J.M.; Catenacci, D.V.; Borad, M.J.; Bridgewater, J.; et al. Ivosidenib in IDH1-mutant, chemotherapy-refractory cholangiocarcinoma (ClarIDHy): A multicentre, randomised, double-blind, placebo-controlled, phase 3 study. Lancet Oncol. 2020, 21, 796–807. [Google Scholar] [CrossRef]
- Roboz, G.J.; Dinardo, C.D.; Stein, E.M.; de Botton, S.; Mims, A.S.; Prince, G.T.; Altman, J.K.; Arellano, M.L.; Donnellan, W.; Erba, H.P.; et al. Ivosidenib induces deep durable remissions in patients with newly diagnosed IDH1-mutant acute myeloid leukemia. Blood 2020, 135, 463–471. [Google Scholar] [CrossRef] [Green Version]
- Stein, E.M.; DiNardo, C.D.; Pollyea, D.A.; Fathi, A.T.; Roboz, G.J.; Altman, J.K.; Stone, R.M.; DeAngelo, D.J.; Levine, R.L.; Flinn, I.W.; et al. Enasidenib in mutant IDH2 relapsed or refractory acute myeloid leukemia. Blood 2017, 130, 722–731. [Google Scholar] [CrossRef]
- Lopez, G.Y.; Reitman, Z.J.; Solomon, D.; Waldman, T.; Bigner, D.D.; McLendon, R.E.; Rosenberg, S.A.; Samuels, Y.; Yan, H. IDH1R132 mutation identified in one human melanoma metastasis, but not correlated with metastases to the brain. Biochem. Biophys. Res. Commun. 2010, 398, 585–587. [Google Scholar] [CrossRef] [Green Version]
- Shibata, T.; Kokubu, A.; Miyamoto, M.; Sasajima, Y.; Yamazaki, N. Mutant IDH1 Confers an in Vivo Growth in a Melanoma Cell Line with BRAF Mutation. Am. J. Pathol. 2011, 178, 1395–1402. [Google Scholar] [CrossRef] [PubMed]
- Pearl, L.H.; Schierz, A.C.; Ward, S.E.; Al-Lazikani, B.; Pearl, F.M. Therapeutic opportunities within the DNA damage response. Nat. Rev. Cancer 2015, 15, 166–180. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chatterjee, N.; Walker, G.C. Mechanisms of DNA damage, repair, and mutagenesis. Environ. Mol. Mutagen. 2017, 58, 235–263. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kaufman, B.; Shapira-Frommer, R.; Schmutzler, R.K.; Audeh, M.W.; Friedlander, M.; Balmaña, J.; Mitchell, G.; Fried, G.; Stemmer, S.M.; Hubert, A.; et al. Olaparib Monotherapy in Patients with Advanced Cancer and a Germline BRCA1/2 Mutation. J. Clin. Oncol. 2015, 33, 244–250. [Google Scholar] [CrossRef]
- Moore, K.; Colombo, N.; Scambia, G.; Kim, B.-G.; Oaknin, A.; Friedlander, M.; Lisyanskaya, A.; Floquet, A.; Leary, A.; Sonke, G.S.; et al. Maintenance Olaparib in Patients with Newly Diagnosed Advanced Ovarian Cancer. N. Engl. J. Med. 2018, 379, 2495–2505. [Google Scholar] [CrossRef]
- Mirza, M.R.; Monk, B.J.; Herrstedt, J.; Oza, A.M.; Mahner, S.; Redondo, A.; Fabbro, M.; Ledermann, J.A.; Lorusso, D.; Vergote, I.; et al. Niraparib Maintenance Therapy in Platinum-Sensitive, Recurrent Ovarian Cancer. N. Engl. J. Med. 2016, 375, 2154–2164. [Google Scholar] [CrossRef]
- Litton, J.K.; Rugo, H.S.; Ettl, J.; Hurvitz, S.A.; Gonçalves, A.; Lee, K.-H.; Fehrenbacher, L.; Yerushalmi, R.; Mina, L.A.; Martin, M.; et al. Talazoparib in Patients with Advanced Breast Cancer and a Germline BRCA Mutation. N. Engl. J. Med. 2018, 379, 753–763. [Google Scholar] [CrossRef] [PubMed]
- de Bono, J.S.; Mateo, J.; Fizazi, K.; Saad, F.; Shore, N.; Sandhu, S.; Chi, K.N.; Sartor, O.; Agarwal, N.; Olmos, D.; et al. Olaparib for Metastatic Castration-Resistant Prostate Cancer. N. Engl. J. Med. 2020, 382, 2091–2102. [Google Scholar] [CrossRef]
- Golan, T.; Hammel, P.; Reni, M.; van Cutsem, E.; Macarulla, T.; Hall, M.J.; Park, J.-O.; Hochhauser, D.; Arnold, D.; Oh, D.-Y.; et al. Maintenance Olaparib for Germline BRCA-Mutated Metastatic Pancreatic Cancer. N. Engl. J. Med. 2019, 381, 317–327. [Google Scholar] [CrossRef]
- O’Connor, M.J. Targeting the DNA Damage Response in Cancer. Mol. Cell 2015, 60, 547–560. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Knijnenburg, T.A.; Wang, L.; Zimmermann, M.T.; Chambwe, N.; Gao, G.F.; Cherniack, A.D.; Fan, H.; Shen, H.; Way, G.P.; Greene, C.S.; et al. Genomic and Molecular Landscape of DNA Damage Repair Deficiency across The Cancer Genome Atlas. Cell Rep. 2018, 23, 239–254. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pilié, P.G.; Tang, C.; Mills, G.B.; Yap, T.A. State-of-the-art strategies for targeting the DNA damage response in cancer. Nat. Rev. Clin. Oncol. 2019, 16, 81–104. [Google Scholar] [CrossRef] [PubMed]
- Heeke, A.L.; Pishvaian, M.J.; Lynce, F.; Xiu, J.; Brody, J.R.; Chen, W.-J.; Baker, T.M.; Marshall, J.L.; Isaacs, C. Prevalence of Homologous Recombination–Related Gene Mutations Across Multiple Cancer Types. JCO Precis. Oncol. 2018, 2, 1–13. [Google Scholar] [CrossRef] [PubMed]
- Chan, W.Y.; Brown, L.J.; Reid, L.; Joshua, A.M. PARP Inhibitors in Melanoma—An Expanding Therapeutic Option? Cancers 2021, 13, 4520. [Google Scholar] [CrossRef] [PubMed]
- Makino, E.; Fröhlich, L.M.; Sinnberg, T.; Kosnopfel, C.; Sauer, B.; Garbe, C.; Schittek, B. Targeting Rad51 as a strategy for the treatment of melanoma cells resistant to MAPK pathway inhibition. Cell Death Dis. 2020, 11, 581. [Google Scholar] [CrossRef] [PubMed]
- Kim, K.B.; Soroceanu, L.; de Semir, D.; Millis, S.Z.; Ross, J.; Vosoughi, E.; Dar, A.A.; Nosrati, M.; Desprez, P.-Y.; Ice, R.; et al. Prevalence of Homologous Recombination Pathway Gene Mutations in Melanoma: Rationale for a New Targeted Therapeutic Approach. J. Investig. Dermatol. 2021, 141, 2028–2036. [Google Scholar] [CrossRef]
- Chabanon, R.M.; Muirhead, G.; Krastev, D.B.; Adam, J.; Morel, D.; Garrido, M.; Lamb, A.; Hénon, C.; Dorvault, N.; Rouanne, M.; et al. PARP inhibition enhances tumor cell–intrinsic immunity in ERCC1-deficient non–small cell lung cancer. J. Clin. Investig. 2019, 129, 1211–1228. [Google Scholar] [CrossRef] [PubMed]
- Peyraud, F.; Italiano, A. Combined PARP Inhibition and Immune Checkpoint Therapy in Solid Tumors. Cancers 2020, 12, 1502. [Google Scholar] [CrossRef] [PubMed]
- Lau, B.; Menzies, A.; Joshua, A. Ongoing partial response at 6 months to olaparib for metastatic melanoma with somatic PALB2 mutation after failure of immunotherapy: A case report. Ann. Oncol. 2021, 32, 280–282. [Google Scholar] [CrossRef]
- Khaddour, K.; Ansstas, M.; Visconti, J. Mutation clearance and complete radiologic resolution of immunotherapy relapsed metastatic melanoma after treatment with nivolumab and olaparib in a patient with homologous recombinant deficiency: Any role for PARP inhibitors and checkpoint blockade? Ann. Oncol. 2021, 32, 279–280. [Google Scholar] [CrossRef] [PubMed]
- Khaddour, K.; Ansstas, M.; Ansstas, G. Clinical outcomes and longitudinal circulating tumor DNA changes after treatment with nivolumab and olaparib in immunotherapy relapsed melanoma with detected homologous recombination deficiency. Cold Spring Harb Mol. Case Stud. 2021, 7, a006129. [Google Scholar] [CrossRef] [PubMed]
Study Design | Study Drug | Dose | Number of Patients | Type of Mutations | Special Characteristics | Overall Response Rate | Duration of Response | Median Progression Free Survival | Median Overall Survival | Reference |
---|---|---|---|---|---|---|---|---|---|---|
Single group, open label, multicenter, phase 2 trial | Imatinib | 400 mg twice daily | 28 | Mutation and/or amplification | 328 patients screened, 51 had KIT alterations, 28 were treated, 25 analyzed (3 excluded due to toxicity) | 16% | 2 CR (94 and 95 weeks), 2 PR (53 and 89 weeks) and 2 transient PR (12 and 18 weeks) | 12 weeks | 46.3 weeks | [60] |
Single arm, open label, single center, phase 2 trial | Imatinib | 400 mg daily | 43 | Mutation and/or amplification | Dose allowed to be escalated to 600 mg or 800 mg daily if POD | 22% | Not available | 3.5 months | 14 months | [61] |
Single arm, open label, multicenter, phase 2 trial | Imatinib | 400 mg daily | 25 | Mutation and/or amplification | Dose escalated to 400 mg twice daily if no response | 29% (21% excluding non-confirmed responses) | Not available | 3.7 months | 12.5 months | [62] |
Retrospective chart review, single center | Imatinib | 400 mg daily | 78 | Mutation and/or amplification | Largest study, but retrospective | 21.8% | Not available | 4.2 months | 13.1 months | [63] |
Single arm, open label, single center, phase 2 trial | Nilotinib | 400 mg twice daily | 11 | Mutation and/or amplification | Of the 11 patients, 9 were evaluated for a response. Most patients had M1c disease | 22.2% | 2 w/KIT mutations responded for 8.4 and 10.0 months. 1 w/amplification had SD for 6 months | 2.5 months | 7.7 months | [64] |
Phase 2 trial with 2 study groups, open label | Nilotinib | 400 mg twice daily | 19 | Mutation and/or amplification | 20 enrolled, 19 treated. 4 were not evaluated for radiographic responses to therapy. 2 cohorts: (A) those refractory or intolerant to a prior KIT inhibitor; and B) those with brain metastases | 10.5% overall (18.1% in cohort A, 2 PR; 0 in cohort B) | One patient in cohort A had ongoing response for 34.5 months. One in cohort B had a 3.9-month response in CNS disease | 3.3 months | 9.1 months | [65] |
Open label, single arm, multicenter, phase 2 | Nilotinib | 400 mg twice daily | 42 | Mutation and/or amplification | 176 patients screened, 42 enrolled. PFS 8.5 months in CR/PR/SD vs. 7 weeks in PD | 16.7% | 34 weeks | 3.3 months | 11.9 months | [66] |
Open label, single arm, multicenter, phase 2 | Nilotinib | 400 mg twice daily | 42 | Mutation only | First to evaluate nilotinib without prior KIT inhibitor therapy. Originally designed for 2 groups (prior dacarbazine vs. nilotinib), but not enough patients | 26.2% | 7.1 months | 4.2 months | 18.0 months | [67] |
Open label, single arm, multicenter, phase 2 | Nilotinib | 400 mg twice daily | 25 | Mutation and/or amplification | 4 patients exhibited durable response, 3 persisting (3.6 and 2.8 years for 2 patients with stage IIIC and 2.5 years for 1 with IVM1b) | 20% | In patients with CR and PR, 46.8 months | 6.0 months | 13.2 months | [68] |
Two-stage, open label, single arm, single center, phase 2 | Dasatinib | 70 mg twice daily | 30 in stage 2 | Mutation only | 2 stages: (1) both KIT+ and KIT-wt w/ mucosal, acral and CSD melanoma; 57 patients, 51 analyzed; (2) trial amended for KIT+ only and added vulvo-vaginal and excluded CSD melanoma; 30 patients, 22 analyzed | 18.2% in stage 2 5.9% in stage 1 | 4.2 months in stage 2 | 2.1 months (stage 1 and 2 combined) 2.7 months in KIT+ patients (both stages) | 7.5 months (stage 1 and 2 combined) 11.8 months in KIT+ patients (both stages) | [69] |
Open label, single arm, single center, phase 2 | Sunitinib | 50 mg daily | 12 (10 analyzed) | Mutation, amplification or over-expression | 90 enrolled, 12 treated with sunitib (KIT alterations). Sunitib given 4 weeks on, 2 off. Dose reduced to 37.5 or 25 mg if AEs | 40% | 1 CR for 15 months, 2 PR (1 month and 7 months) in patients with mutations. 1 PR in amplification or over-expression. | Not available | Not available | [70] |
Drug(s) | Study Phase | Target Population | Sample Size | ClinicalTrials.gov (Accessed on 1 September 2021) Registration |
---|---|---|---|---|
MEK + Autophagy inhibitor Trametinib + Hydroxychloroquine | Phase 1b/2 | Metastatic or locally advanced unresectable NRAS melanoma | 29 | NCT03979651 |
MEK inhibitor Binimetinib | Phase 2 | BRAF or NRAS mutant locally advanced or metastatic melanoma | 183 | NCT01320085 |
MEK inhibitor HL-085 | Phase 1/2 | Stage III or Stage IV NRAS mutated melanoma | 54 | NCT03973151 |
MEK inhibitor FCN-159 | Phase 1a/1b | NRAS aberrant and NRAS mutated melanoma | 37 | NCT03932253 |
RAF inhibitor + ERK inhibitor +/− other agents Encorafenib + LY3214996 | Phase 1 | Part B: BRAF mutant metastatic melanoma refractory or relapse, NRAS metastatic melanoma, BRAF mutant NSCLC | 245 | NCT02857270 |
ERK inhibitor ASN007 | Phase 1 | NRAS/BRAF mutant melanoma | 49 | NCT03415126 |
ARF-sparing inhibitor of BRAF and CRAF + ERK inhibitor or MEK inhibitor or CD4/6 inhibitor LXH254 + LTT462 or Trametinib or Ribociclib | Phase 1b | KRAS/BRAF NSCLC | 331 | NCT02974725 |
Phase 2 | NRAS mutant melanoma | 320 | NCT04417621 | |
ERBB3 antibody + MEK inhibitor CDX-3370 + Trametinib | Phase 1b/2 Terminated | NRAS melanoma | 3 | NCT03580382 |
RAF Inhibitor + MEK inhibitor + immune check point inhibitor | Phase 1b | Stage IV or Stage III NRAS melanoma relapse after check point inhibitor | 83 | NCT04835805 |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Khaddour, K.; Maahs, L.; Avila-Rodriguez, A.M.; Maamar, Y.; Samaan, S.; Ansstas, G. Melanoma Targeted Therapies beyond BRAF-Mutant Melanoma: Potential Druggable Mutations and Novel Treatment Approaches. Cancers 2021, 13, 5847. https://doi.org/10.3390/cancers13225847
Khaddour K, Maahs L, Avila-Rodriguez AM, Maamar Y, Samaan S, Ansstas G. Melanoma Targeted Therapies beyond BRAF-Mutant Melanoma: Potential Druggable Mutations and Novel Treatment Approaches. Cancers. 2021; 13(22):5847. https://doi.org/10.3390/cancers13225847
Chicago/Turabian StyleKhaddour, Karam, Lucas Maahs, Ana Maria Avila-Rodriguez, Yazan Maamar, Sami Samaan, and George Ansstas. 2021. "Melanoma Targeted Therapies beyond BRAF-Mutant Melanoma: Potential Druggable Mutations and Novel Treatment Approaches" Cancers 13, no. 22: 5847. https://doi.org/10.3390/cancers13225847
APA StyleKhaddour, K., Maahs, L., Avila-Rodriguez, A. M., Maamar, Y., Samaan, S., & Ansstas, G. (2021). Melanoma Targeted Therapies beyond BRAF-Mutant Melanoma: Potential Druggable Mutations and Novel Treatment Approaches. Cancers, 13(22), 5847. https://doi.org/10.3390/cancers13225847