메뉴 건너뛰기




Volumn 5, Issue Aug, 2015, Pages

Multiple molecular pathways in melanomagenesis: Characterization of therapeutic targets

Author keywords

Alternative therapeutic targets; Melanoma pathogenesis; Molecular melanoma classification; Signal transduction cascades; Targeted therapy resistance

Indexed keywords

AURORA KINASE; B RAF KINASE; CYCLIN DEPENDENT KINASE 4; CYCLIN DEPENDENT KINASE INHIBITOR 2A; DABRAFENIB; METFORMIN; MICROPHTHALMIA ASSOCIATED TRANSCRIPTION FACTOR; MITOGEN ACTIVATED PROTEIN KINASE; PHOSPHATIDYLINOSITOL 3 KINASE; PHOSPHATIDYLINOSITOL 4,5 BISPHOSPHATE; PROTEIN MDM2; PROTEIN P53; VEMURAFENIB;

EID: 84940989576     PISSN: None     EISSN: 2234943X     Source Type: Journal    
DOI: 10.3389/fonc.2015.00183     Document Type: Review
Times cited : (85)

References (172)
  • 4
    • 84918815964 scopus 로고    scopus 로고
    • Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012
    • Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer (2015) 136:E359-86. doi:10.1002/ijc.29210.
    • (2015) Int J Cancer , vol.136 , pp. E359-E386
    • Ferlay, J.1    Soerjomataram, I.2    Dikshit, R.3    Eser, S.4    Mathers, C.5    Rebelo, M.6
  • 5
    • 84911866508 scopus 로고    scopus 로고
    • The melanoma revolution: from UV carcinogenesis to a new era in therapeutics
    • Lo JA, Fisher DE. The melanoma revolution: from UV carcinogenesis to a new era in therapeutics. Science (2014) 346:945-9. doi:10.1126/science.1253735.
    • (2014) Science , vol.346 , pp. 945-949
    • Lo, J.A.1    Fisher, D.E.2
  • 6
    • 84934976308 scopus 로고    scopus 로고
    • Long-term outcome in BRAF(V600E) melanoma patients treated with vemurafenib: patterns of disease progression and clinical management of limited progression
    • Puzanov I, Amaravadi RK, McArthur GA, Flaherty KT, Chapman PB, Sosman JA, et al. Long-term outcome in BRAF(V600E) melanoma patients treated with vemurafenib: patterns of disease progression and clinical management of limited progression. Eur J Cancer (2015) 51:1435-43. doi:10.1016/j.ejca.2015.04.010.
    • (2015) Eur J Cancer , vol.51 , pp. 1435-1443
    • Puzanov, I.1    Amaravadi, R.K.2    McArthur, G.A.3    Flaherty, K.T.4    Chapman, P.B.5    Sosman, J.A.6
  • 7
    • 84905032732 scopus 로고    scopus 로고
    • Overall survival in COMBI-d, a randomized, double-blinded, phase III study comparing the combination of dabrafenib and trametinib with dabrafenib and placebo as first-line therapy in patients (pts) with unresectable or metastatic BRAF V600E/K mutation-positive cutaneous melanoma
    • Long GV, Stroyakovsky DL, Gogas H, Levchenko E, de Braud F, Larkin JMG, et al. Overall survival in COMBI-d, a randomized, double-blinded, phase III study comparing the combination of dabrafenib and trametinib with dabrafenib and placebo as first-line therapy in patients (pts) with unresectable or metastatic BRAF V600E/K mutation-positive cutaneous melanoma. J Clin Oncol (2015) 33(Suppl):abstr102.
    • (2015) J Clin Oncol , vol.33
    • Long, G.V.1    Stroyakovsky, D.L.2    Gogas, H.3    Levchenko, E.4    de Braud, F.5    Larkin, J.M.G.6
  • 8
    • 84941010092 scopus 로고    scopus 로고
    • Anti-PD1 and anti-PD-L1 in the treatment of metastatic melanoma
    • Simeone E, Grimaldi AM, Ascierto PA. Anti-PD1 and anti-PD-L1 in the treatment of metastatic melanoma. Melanoma Manag (2015) 2:41-50. doi:10.2217/MMT.14.30.
    • (2015) Melanoma Manag , vol.2 , pp. 41-50
    • Simeone, E.1    Grimaldi, A.M.2    Ascierto, P.A.3
  • 9
    • 0028085975 scopus 로고
    • Analysis of the p16 gene (CDKN2) as a candidate for the chromosome 9p melanoma susceptibility locus
    • Kamb A, Shattuck-Eidens D, Eeles R, Liu Q, Gruis NA, Ding W, et al. Analysis of the p16 gene (CDKN2) as a candidate for the chromosome 9p melanoma susceptibility locus. Nat Genet (1994) 8:23-6. doi:10.1038/ng0994-22.
    • (1994) Nat Genet , vol.8 , pp. 23-26
    • Kamb, A.1    Shattuck-Eidens, D.2    Eeles, R.3    Liu, Q.4    Gruis, N.A.5    Ding, W.6
  • 10
    • 33747587608 scopus 로고    scopus 로고
    • Regulation of the INK4b-ARF-INK4a tumour suppressor locus: all for one or one for all
    • Gil J, Peters G. Regulation of the INK4b-ARF-INK4a tumour suppressor locus: all for one or one for all. Nat Rev Mol Cell Biol (2006) 7:667-77. doi:10.1038/nrm1987.
    • (2006) Nat Rev Mol Cell Biol , vol.7 , pp. 667-677
    • Gil, J.1    Peters, G.2
  • 11
    • 70350152582 scopus 로고    scopus 로고
    • Genetic risk factors for melanoma
    • Meyle KD, Guldberg P. Genetic risk factors for melanoma. Hum Genet (2009) 126:499-510. doi:10.1007/s00439-009-0715-9.
    • (2009) Hum Genet , vol.126 , pp. 499-510
    • Meyle, K.D.1    Guldberg, P.2
  • 13
    • 35748972055 scopus 로고    scopus 로고
    • Issues affecting molecular staging in the management of patients with melanoma
    • Palmieri G, Casula M, Sini MC, Ascierto PA, Cossu A. Issues affecting molecular staging in the management of patients with melanoma. J Cell Mol Med (2007) 11:1052-68. doi:10.1111/j.1582-4934.2007.00091.x.
    • (2007) J Cell Mol Med , vol.11 , pp. 1052-1068
    • Palmieri, G.1    Casula, M.2    Sini, M.C.3    Ascierto, P.A.4    Cossu, A.5
  • 14
    • 54049158957 scopus 로고    scopus 로고
    • Increased cyclin D1 expression can mediate BRAF inhibitor resistance in BRAF V600E-mutated melanomas
    • Smalley KS, Lioni M, Dalla Palma M, Xiao M, Desai B, Egyhazi S, et al. Increased cyclin D1 expression can mediate BRAF inhibitor resistance in BRAF V600E-mutated melanomas. Mol Cancer Ther (2008) 7:2876-83. doi:10.1158/1535-7163.MCT-08-0431.
    • (2008) Mol Cancer Ther , vol.7 , pp. 2876-2883
    • Smalley, K.S.1    Lioni, M.2    Dalla Palma, M.3    Xiao, M.4    Desai, B.5    Egyhazi, S.6
  • 15
    • 52149120730 scopus 로고    scopus 로고
    • The role of p53 in pigmentation, tanning and melanoma
    • Box NF, Terzian T. The role of p53 in pigmentation, tanning and melanoma. Pigment Cell Melanoma Res (2008) 21:525-33. doi:10.1111/j.1755-148X.2008.00495.x.
    • (2008) Pigment Cell Melanoma Res , vol.21 , pp. 525-533
    • Box, N.F.1    Terzian, T.2
  • 16
    • 16844376315 scopus 로고    scopus 로고
    • Oncogenic Ras in tumor progression and metastasis
    • Giehl K. Oncogenic Ras in tumor progression and metastasis. Biol Chem (2005) 386:193-205. doi:10.1515/BC.2005.025.
    • (2005) Biol Chem , vol.386 , pp. 193-205
    • Giehl, K.1
  • 17
    • 33644774541 scopus 로고    scopus 로고
    • Examination of mutations in BRAF, NRAS, and PTEN in primary cutaneous melanoma
    • Goel VK, Lazar AJ, Warneke CL, Redston MS, Haluska FG. Examination of mutations in BRAF, NRAS, and PTEN in primary cutaneous melanoma. J Invest Dermatol (2006) 126:154-60. doi:10.1038/sj.jid.5700026.
    • (2006) J Invest Dermatol , vol.126 , pp. 154-160
    • Goel, V.K.1    Lazar, A.J.2    Warneke, C.L.3    Redston, M.S.4    Haluska, F.G.5
  • 19
    • 84879695087 scopus 로고    scopus 로고
    • ARAF acts as a scaffold to stabilize BRAF:CRAF heterodimers
    • Rebocho AP, Marais R. ARAF acts as a scaffold to stabilize BRAF:CRAF heterodimers. Oncogene (2013) 32:3207-12. doi:10.1038/onc.2012.330.
    • (2013) Oncogene , vol.32 , pp. 3207-3212
    • Rebocho, A.P.1    Marais, R.2
  • 23
    • 13444258032 scopus 로고    scopus 로고
    • BRAF mutations are sufficient to promote nevi formation and cooperate with p53 in the genesis of melanoma
    • Patton EE, Widlund HR, Kutok JL, Kopani KR, Amatruda JF, Murphey RD, et al. BRAF mutations are sufficient to promote nevi formation and cooperate with p53 in the genesis of melanoma. Curr Biol (2005) 15:249-54. doi:10.1016/j.cub.2005.01.031.
    • (2005) Curr Biol , vol.15 , pp. 249-254
    • Patton, E.E.1    Widlund, H.R.2    Kutok, J.L.3    Kopani, K.R.4    Amatruda, J.F.5    Murphey, R.D.6
  • 24
    • 38949143728 scopus 로고    scopus 로고
    • The phosphoinositide 3-kinase pathway in human cancer: genetic alterations and therapeutic implications
    • Arcaro A, Guerreiro AS. The phosphoinositide 3-kinase pathway in human cancer: genetic alterations and therapeutic implications. Curr Genomics (2007) 8:271-306. doi:10.2174/138920207782446160.
    • (2007) Curr Genomics , vol.8 , pp. 271-306
    • Arcaro, A.1    Guerreiro, A.S.2
  • 25
    • 50549104987 scopus 로고    scopus 로고
    • Oncogenic BRAF regulates melanoma proliferation through the lineage specific factor MITF
    • Wellbrock C, Rana S, Paterson H. Oncogenic BRAF regulates melanoma proliferation through the lineage specific factor MITF. PLoS One (2008) 3:2734. doi:10.1371/journal.pone.0002734.
    • (2008) PLoS One , vol.3 , pp. 2734
    • Wellbrock, C.1    Rana, S.2    Paterson, H.3
  • 26
    • 10944234560 scopus 로고    scopus 로고
    • Melanocytes and the microphthalmia transcription factor network
    • Steingrímsson E, Copeland NG, Jenkins NA. Melanocytes and the microphthalmia transcription factor network. Annu Rev Genet (2004) 38:365-411. doi:10.1146/annurev.genet.38.072902.092717.
    • (2004) Annu Rev Genet , vol.38 , pp. 365-411
    • Steingrímsson, E.1    Copeland, N.G.2    Jenkins, N.A.3
  • 27
    • 84877873340 scopus 로고    scopus 로고
    • Molecular network associated with MITF in skin melanoma development and progression
    • Yajima I, Kumasaka MY, Thang ND, Goto Y, Takeda K, Iida M, et al. Molecular network associated with MITF in skin melanoma development and progression. J Skin Cancer (2011) 2011:730170. doi:10.1155/2011/730170.
    • (2011) J Skin Cancer , vol.2011
    • Yajima, I.1    Kumasaka, M.Y.2    Thang, N.D.3    Goto, Y.4    Takeda, K.5    Iida, M.6
  • 28
    • 21844478747 scopus 로고    scopus 로고
    • Integrative genomic analyses identify MITF as a lineage survival oncogene amplified in malignant melanoma
    • Garraway LA, Widlund HR, Rubin MA, Getz G, Berger AJ, Ramaswamy S, et al. Integrative genomic analyses identify MITF as a lineage survival oncogene amplified in malignant melanoma. Nature (2005) 436:117-22. doi:10.1038/nature03664.
    • (2005) Nature , vol.436 , pp. 117-122
    • Garraway, L.A.1    Widlund, H.R.2    Rubin, M.A.3    Getz, G.4    Berger, A.J.5    Ramaswamy, S.6
  • 29
    • 38849203228 scopus 로고    scopus 로고
    • In vivo switching of human melanoma cells between proliferative and invasive states
    • Hoek KS, Eichhoff OM, Schlegel NC, Döbbeling U, Kobert N, Schaerer L, et al. In vivo switching of human melanoma cells between proliferative and invasive states. Cancer Res (2008) 68:650-6. doi:10.1158/0008-5472.CAN-07-2491.
    • (2008) Cancer Res , vol.68 , pp. 650-656
    • Hoek, K.S.1    Eichhoff, O.M.2    Schlegel, N.C.3    Döbbeling, U.4    Kobert, N.5    Schaerer, L.6
  • 30
    • 84939223067 scopus 로고    scopus 로고
    • c-Kit signaling as the driving oncogenic event in sub-groups of melanomas. histology and histopathology cellular and molecular biology
    • Smalley KS, Sondak VK, Weber JS. c-Kit signaling as the driving oncogenic event in sub-groups of melanomas. histology and histopathology cellular and molecular biology. J Pathol (2009) 29:643-50.
    • (2009) J Pathol , vol.29 , pp. 643-650
    • Smalley, K.S.1    Sondak, V.K.2    Weber, J.S.3
  • 34
    • 77949712411 scopus 로고    scopus 로고
    • Targeted inhibition of inducible nitric oxide synthase inhibits growth of human melanoma in vivo and synergizes with chemo-therapy
    • Sikora AG, Gelbard A, Davies A, Sano D, Ekmekcioglu S, Kwon J, et al. Targeted inhibition of inducible nitric oxide synthase inhibits growth of human melanoma in vivo and synergizes with chemo-therapy. Clin Cancer Res (2010) 16:1834-44. doi:10.1158/1078-0432.CCR-09-3123.
    • (2010) Clin Cancer Res , vol.16 , pp. 1834-1844
    • Sikora, A.G.1    Gelbard, A.2    Davies, A.3    Sano, D.4    Ekmekcioglu, S.5    Kwon, J.6
  • 35
    • 80052936747 scopus 로고    scopus 로고
    • Met receptor: a moving target
    • Clague MJ. Met receptor: a moving target. Sci Signal (2011) 4:e40. doi:10.1126/scisignal.2002422.
    • (2011) Sci Signal , vol.4
    • Clague, M.J.1
  • 36
    • 79955606614 scopus 로고    scopus 로고
    • Expression of the c-Met proteins in malignant skin cancers
    • Lee YJ, Kim DH, Lee SH, Kim DW, Nam HS, Cho MK. Expression of the c-Met proteins in malignant skin cancers. Ann Dermatol (2011) 23:33-8. doi:10.5021/ad.2011.23.1.33.
    • (2011) Ann Dermatol , vol.23 , pp. 33-38
    • Lee, Y.J.1    Kim, D.H.2    Lee, S.H.3    Kim, D.W.4    Nam, H.S.5    Cho, M.K.6
  • 37
    • 79952072157 scopus 로고    scopus 로고
    • Transcriptional control of melanoma metastasis: the importance of the tumor microenvironment
    • Braeuer RR, Zigler M, Villares GJ, Dobroff AS, Bar-Eli M. Transcriptional control of melanoma metastasis: the importance of the tumor microenvironment. Semin Cancer Biol (2011) 21:83-8. doi:10.1016/j.semcancer.2010.12.007.
    • (2011) Semin Cancer Biol , vol.21 , pp. 83-88
    • Braeuer, R.R.1    Zigler, M.2    Villares, G.J.3    Dobroff, A.S.4    Bar-Eli, M.5
  • 38
    • 79953087601 scopus 로고    scopus 로고
    • Molecular profiling to identify relevant immune resistance mechanisms in the tumor microenvironment
    • Gajewski TF, Fuertes M, Spaapen R, Zheng Y, Kline J. Molecular profiling to identify relevant immune resistance mechanisms in the tumor microenvironment. Curr Opin Immunol (2011) 23:286-92. doi:10.1016/j.coi.2010.11.013.
    • (2011) Curr Opin Immunol , vol.23 , pp. 286-292
    • Gajewski, T.F.1    Fuertes, M.2    Spaapen, R.3    Zheng, Y.4    Kline, J.5
  • 39
    • 72049114407 scopus 로고    scopus 로고
    • Understanding melanoma signaling networks as the basis for molecular targeted therapy
    • Smalley KS. Understanding melanoma signaling networks as the basis for molecular targeted therapy. J Invest Dermatol (2010) 130:28-37. doi:10.1038/jid.2009.177.
    • (2010) J Invest Dermatol , vol.130 , pp. 28-37
    • Smalley, K.S.1
  • 40
    • 84873819555 scopus 로고    scopus 로고
    • Which drug, and when, for patients with BRAF-mutant melanoma?
    • Jang S, Atkins MB. Which drug, and when, for patients with BRAF-mutant melanoma? Lancet Oncol (2013) 14:e60-9. doi:10.1016/S1470-2045(12)70539-9.
    • (2013) Lancet Oncol , vol.14 , pp. e60-e69
    • Jang, S.1    Atkins, M.B.2
  • 41
    • 84922662201 scopus 로고    scopus 로고
    • Cell cycle control as a promising target in melanoma
    • Lee B, Sandhu S, McArthur G. Cell cycle control as a promising target in melanoma. Curr Opin Oncol (2015) 27:141-50. doi:10.1097/CCO.0000000000000159.
    • (2015) Curr Opin Oncol , vol.27 , pp. 141-150
    • Lee, B.1    Sandhu, S.2    McArthur, G.3
  • 42
    • 19544371601 scopus 로고    scopus 로고
    • Histone deacetylase inhibitors and malignant melanoma
    • Boyle GM, Martyn AC, Parsons PG. Histone deacetylase inhibitors and malignant melanoma. Pigment Cell Res (2005) 18:160-6. doi:10.1111/j.1600-0749.2005.00228.x.
    • (2005) Pigment Cell Res , vol.18 , pp. 160-166
    • Boyle, G.M.1    Martyn, A.C.2    Parsons, P.G.3
  • 43
    • 47249113549 scopus 로고    scopus 로고
    • Pharmacologic suppression of MITF expression via HDAC inhibitors in the melanocyte lineage
    • Yokoyama S, Feige E, Poling LL, Levy C, Widlund HR, Khaled M, et al. Pharmacologic suppression of MITF expression via HDAC inhibitors in the melanocyte lineage. Pigment Cell Melanoma Res (2008) 21:457-63. doi:10.1111/j.1755-148X.2008.00480.x.
    • (2008) Pigment Cell Melanoma Res , vol.21 , pp. 457-463
    • Yokoyama, S.1    Feige, E.2    Poling, L.L.3    Levy, C.4    Widlund, H.R.5    Khaled, M.6
  • 44
    • 84879410422 scopus 로고    scopus 로고
    • NRAS mutant melanoma: biological behavior and future strategies for therapeutic management
    • Fedorenko IV, Gibney GT, Smalley KS. NRAS mutant melanoma: biological behavior and future strategies for therapeutic management. Oncogene (2013) 32:3009-18. doi:10.1038/onc.2012.453.
    • (2013) Oncogene , vol.32 , pp. 3009-3018
    • Fedorenko, I.V.1    Gibney, G.T.2    Smalley, K.S.3
  • 45
    • 84862732834 scopus 로고    scopus 로고
    • Combinations of BRAF, MEK, and PI3K/mTOR inhibitors overcome acquired resistance to the BRAF inhibitor GSK2118436 dabrafenib, mediated by NRAS or MEK mutations
    • Greger JG, Eastman SD, Zhang V, Bleam MR, Hughes AM, Smitheman KN, et al. 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) 11:909-20. doi:10.1158/1535-7163.MCT-11-0989.
    • (2012) Mol Cancer Ther , vol.11 , pp. 909-920
    • Greger, J.G.1    Eastman, S.D.2    Zhang, V.3    Bleam, M.R.4    Hughes, A.M.5    Smitheman, K.N.6
  • 46
    • 84875227164 scopus 로고    scopus 로고
    • Targeting hyperactivation of the AKT survival pathway to overcome therapy resistance of melanoma brain metastases
    • Niessner H, Forschner A, Klumpp B, Honegger JB, Witte M, Bornemann A, et al. Targeting hyperactivation of the AKT survival pathway to overcome therapy resistance of melanoma brain metastases. Cancer Med (2013) 2:76-85. doi:10.1002/cam4.50.
    • (2013) Cancer Med , vol.2 , pp. 76-85
    • Niessner, H.1    Forschner, A.2    Klumpp, B.3    Honegger, J.B.4    Witte, M.5    Bornemann, A.6
  • 47
    • 84884597338 scopus 로고    scopus 로고
    • MEK inhibition in the treatment of advanced melanoma
    • Salama AK, Kim KB. MEK inhibition in the treatment of advanced melanoma. Curr Oncol Rep (2013) 15:473-82. doi:10.1007/s11912-013-0336-2.
    • (2013) Curr Oncol Rep , vol.15 , pp. 473-482
    • Salama, A.K.1    Kim, K.B.2
  • 48
    • 0037264633 scopus 로고    scopus 로고
    • Targeting RAS signalling pathways in cancer therapy
    • Downward J. Targeting RAS signalling pathways in cancer therapy. Nat Rev Cancer (2003) 3:11-22. doi:10.1038/nrc969.
    • (2003) Nat Rev Cancer , vol.3 , pp. 11-22
    • Downward, J.1
  • 50
    • 77951495999 scopus 로고    scopus 로고
    • Mutation-driven drug development in melanoma
    • Flaherty KT, Hodi FS, Bastian BC. Mutation-driven drug development in melanoma. Curr Opin Oncol (2010) 22:178-83. doi:10.1097/CCO.0b013e32833888ee.
    • (2010) Curr Opin Oncol , vol.22 , pp. 178-183
    • Flaherty, K.T.1    Hodi, F.S.2    Bastian, B.C.3
  • 51
    • 33745018419 scopus 로고    scopus 로고
    • Mutually exclusive N-Ras Q61R and BRAF V600E mutations at the single-cell level in the same human melanoma
    • Sensi M, Nicolini G, Petti C, Bersani I, Lozupone F, Molla A, et al. Mutually exclusive N-Ras Q61R and BRAF V600E mutations at the single-cell level in the same human melanoma. Oncogene (2006) 25:3357-64. doi:10.1038/sj.onc.1209379.
    • (2006) Oncogene , vol.25 , pp. 3357-3364
    • Sensi, M.1    Nicolini, G.2    Petti, C.3    Bersani, I.4    Lozupone, F.5    Molla, A.6
  • 52
    • 33745925864 scopus 로고    scopus 로고
    • BRAF somatic mutations in malignant melanoma and melanocytic naevi
    • Thomas NE. BRAF somatic mutations in malignant melanoma and melanocytic naevi. Melanoma Res (2006) 16:97-103. doi:10.1097/01.cmr.0000215035.38436.87.
    • (2006) Melanoma Res , vol.16 , pp. 97-103
    • Thomas, N.E.1
  • 53
    • 84883104349 scopus 로고    scopus 로고
    • Heterogeneous distribution of BRAF/NRAS mutations among Italian patients with advanced melanoma
    • Colombino M, Lissia A, Capone M, De Giorgi V, Massi D, Stanganelli I, et al. Heterogeneous distribution of BRAF/NRAS mutations among Italian patients with advanced melanoma. J Transl Med (2013) 11:202. doi:10.1186/1479-5876-11-202.
    • (2013) J Transl Med , vol.11 , pp. 202
    • Colombino, M.1    Lissia, A.2    Capone, M.3    De Giorgi, V.4    Massi, D.5    Stanganelli, I.6
  • 54
    • 84862584008 scopus 로고    scopus 로고
    • Distinguishing clinicopathologic features of patients with V600E and V600K BRAF-mutant metastatic melanoma
    • Menzies AM, Haydu LE, Visintin L, Carlino MS, Howle JR, Thompson JF, et al. Distinguishing clinicopathologic features of patients with V600E and V600K BRAF-mutant metastatic melanoma. Clin Cancer Res (2012) 18:3242-9. doi:10.1158/1078-0432.CCR-12-0052.
    • (2012) Clin Cancer Res , vol.18 , pp. 3242-3249
    • Menzies, A.M.1    Haydu, L.E.2    Visintin, L.3    Carlino, M.S.4    Howle, J.R.5    Thompson, J.F.6
  • 55
    • 84860214530 scopus 로고    scopus 로고
    • From genes to drugs: targeted strategies for melanoma
    • Keith T, Flaherty KT, Hodi FS, Fisher DE. From genes to drugs: targeted strategies for melanoma. Nat Rev Cancer (2012) 12:349-61. doi:10.1038/nrc3218.
    • (2012) Nat Rev Cancer , vol.12 , pp. 349-361
    • Keith, T.1    Flaherty, K.T.2    Hodi, F.S.3    Fisher, D.E.4
  • 56
    • 84988660611 scopus 로고    scopus 로고
    • Future perspectives in melanoma research: meeting report from the "melanoma bridge", Napoli, December 5th-8th 2013
    • Ascierto PA, Grimaldi AM, Anderson AC, Bifulco C, Cochran A, Garbe C, et al. Future perspectives in melanoma research: meeting report from the "melanoma bridge", Napoli, December 5th-8th 2013. J Transl Med (2014) 12:277. doi:10.1186/s12967-014-0277-z.
    • (2014) J Transl Med , vol.12 , pp. 277
    • Ascierto, P.A.1    Grimaldi, A.M.2    Anderson, A.C.3    Bifulco, C.4    Cochran, A.5    Garbe, C.6
  • 57
    • 84904888606 scopus 로고    scopus 로고
    • Systemic treatment for BRAF-mutant melanoma: where do we go next?
    • Menzies AM, Long GV. Systemic treatment for BRAF-mutant melanoma: where do we go next? Lancet Oncol (2014) 15:e371-81. doi:10.1016/S1470-2045(14)70072-5.
    • (2014) Lancet Oncol , vol.15 , pp. e371-e381
    • Menzies, A.M.1    Long, G.V.2
  • 59
    • 84922246385 scopus 로고    scopus 로고
    • Targeting drivers of melanoma with synthetic small molecules and phytochemicals
    • Strickland LR, Pal HC, Elmets CA, Afaq F. Targeting drivers of melanoma with synthetic small molecules and phytochemicals. Cancer Lett (2015) 359:20-35. doi:10.1016/j.canlet.2015.01.016.
    • (2015) Cancer Lett , vol.359 , pp. 20-35
    • Strickland, L.R.1    Pal, H.C.2    Elmets, C.A.3    Afaq, F.4
  • 61
    • 84908160197 scopus 로고    scopus 로고
    • Profile of selumetinib and its potential in the treatment of melanoma
    • Kim DW, Patel SP. Profile of selumetinib and its potential in the treatment of melanoma. Onco Targets Ther (2014) 7:1631-9. doi:10.2147/OTT.S51596.
    • (2014) Onco Targets Ther , vol.7 , pp. 1631-1639
    • Kim, D.W.1    Patel, S.P.2
  • 62
    • 84906549120 scopus 로고    scopus 로고
    • Role of the MEK inhibitor trametinib in the treatment of metastatic melanoma
    • King JW, Nathan PD. Role of the MEK inhibitor trametinib in the treatment of metastatic melanoma. Future Oncol (2014) 10:1559-70. doi:10.2217/fon.14.89.
    • (2014) Future Oncol , vol.10 , pp. 1559-1570
    • King, J.W.1    Nathan, P.D.2
  • 63
    • 84886087051 scopus 로고    scopus 로고
    • Clinical characteristics and outcomes with specific BRAF and NRAS mutations in patients with metastatic melanoma
    • Bucheit AD, Syklawer E, Jakob JA, Bassett RL Jr, Curry JL, Gershenwald JE, et al. Clinical characteristics and outcomes with specific BRAF and NRAS mutations in patients with metastatic melanoma. Cancer (2013) 119:3821-9. doi:10.1002/cncr.28306.
    • (2013) Cancer , vol.119 , pp. 3821-3829
    • Bucheit, A.D.1    Syklawer, E.2    Jakob, J.A.3    Bassett, R.L.4    Curry, J.L.5    Gershenwald, J.E.6
  • 64
    • 66349098469 scopus 로고    scopus 로고
    • Combined targeting of BRAF and CRAF or BRAF and PI3K effector pathways is required for efficacy in NRAS mutant tumors
    • Jaiswal BS, Janakiraman V, Kljavin NM, Eastham-Anderson J, Cupp JE, Liang Y, et al. Combined targeting of BRAF and CRAF or BRAF and PI3K effector pathways is required for efficacy in NRAS mutant tumors. PLoS One (2009) 4:e5717. doi:10.1371/journal.pone.0005717.
    • (2009) PLoS One , vol.4
    • Jaiswal, B.S.1    Janakiraman, V.2    Kljavin, N.M.3    Eastham-Anderson, J.4    Cupp, J.E.5    Liang, Y.6
  • 65
    • 70350518191 scopus 로고    scopus 로고
    • Polyclonality of BRAF mutations in acquired melanocytic nevi
    • Lin J, Takata M, Murata H, Goto Y, Kido K, Ferrone S, et al. Polyclonality of BRAF mutations in acquired melanocytic nevi. J Natl Cancer Inst (2009) 101:1423-7. doi:10.1093/jnci/djp309.
    • (2009) J Natl Cancer Inst , vol.101 , pp. 1423-1427
    • Lin, J.1    Takata, M.2    Murata, H.3    Goto, Y.4    Kido, K.5    Ferrone, S.6
  • 66
    • 84864042630 scopus 로고    scopus 로고
    • BRAF/NRAS mutation frequencies among primary tumors and metastases in patients with melanoma
    • Colombino M, Capone M, Lissia A, Cossu A, Rubino C, De Giorgi V, et al. BRAF/NRAS mutation frequencies among primary tumors and metastases in patients with melanoma. J Clin Oncol (2012) 30:2522-9. doi:10.1200/JCO.2011.41.2452.
    • (2012) J Clin Oncol , vol.30 , pp. 2522-2529
    • Colombino, M.1    Capone, M.2    Lissia, A.3    Cossu, A.4    Rubino, C.5    De Giorgi, V.6
  • 67
    • 84855302859 scopus 로고    scopus 로고
    • Intra-and inter-tumor heterogeneity of BRAF(V600E))mutations in primary and metastatic melanoma
    • Yancovitz M, Litterman A, Yoon J, Ng E, Shapiro RL, Berman RS, et al. Intra-and inter-tumor heterogeneity of BRAF(V600E))mutations in primary and metastatic melanoma. PLoS One (2012) 7:e29336. doi:10.1371/journal.pone.0029336.
    • (2012) PLoS One , vol.7
    • Yancovitz, M.1    Litterman, A.2    Yoon, J.3    Ng, E.4    Shapiro, R.L.5    Berman, R.S.6
  • 68
    • 84900820504 scopus 로고    scopus 로고
    • Discrepant alterations in main candidate genes among multiple primary melanomas
    • Colombino M, Sini M, Lissia A, De Giorgi V, Stanganelli I, Ayala F, et al. Discrepant alterations in main candidate genes among multiple primary melanomas. J Transl Med (2014) 12:117. doi:10.1186/1479-5876-12-117.
    • (2014) J Transl Med , vol.12 , pp. 117
    • Colombino, M.1    Sini, M.2    Lissia, A.3    De Giorgi, V.4    Stanganelli, I.5    Ayala, F.6
  • 69
  • 70
    • 84908257665 scopus 로고    scopus 로고
    • Combined BRAF and MEK inhibition versus BRAF inhibition alone in melanoma
    • Long GV, Stroyakovskiy D, Gogas H, Levchenko E, de Braud F, Larkin J, et al. Combined BRAF and MEK inhibition versus BRAF inhibition alone in melanoma. N Engl J Med (2014) 371:1877-88. doi:10.1056/NEJMoa1406037.
    • (2014) N Engl J Med , vol.371 , pp. 1877-1888
    • Long, G.V.1    Stroyakovskiy, D.2    Gogas, H.3    Levchenko, E.4    de Braud, F.5    Larkin, J.6
  • 72
    • 84863000299 scopus 로고    scopus 로고
    • The molecular evolution of acquired resistance to targeted EGFR blockade in colorectal cancers
    • Diaz LA Jr, Williams RT, Wu J, Kinde I, Hecht JR, Berlin J, et al. The molecular evolution of acquired resistance to targeted EGFR blockade in colorectal cancers. Nature (2012) 486:537-40. doi:10.1038/nature11219.
    • (2012) Nature , vol.486 , pp. 537-540
    • Diaz, L.A.1    Williams, R.T.2    Wu, J.3    Kinde, I.4    Hecht, J.R.5    Berlin, J.6
  • 73
    • 84856990335 scopus 로고    scopus 로고
    • Mechanisms of resistance to crizotinib in patients with ALK gene rearranged non-small cell lung cancer
    • Doebele RC, Pilling AB, Aisner DL, Kutateladze TG, Le AT, Weickhardt AJ, et al. Mechanisms of resistance to crizotinib in patients with ALK gene rearranged non-small cell lung cancer. Clin Cancer Res (2012) 18:1472-82. doi:10.1158/1078-0432.CCR-11-2906.
    • (2012) Clin Cancer Res , vol.18 , pp. 1472-1482
    • Doebele, R.C.1    Pilling, A.B.2    Aisner, D.L.3    Kutateladze, T.G.4    Le, A.T.5    Weickhardt, A.J.6
  • 74
    • 84862999938 scopus 로고    scopus 로고
    • Emergence of KRAS mutations and acquired resistance to anti-EGFR therapy in colorectal cancer
    • Misale S, Yaeger R, Hobor S, Scala E, Janakiraman M, Liska D, et al. Emergence of KRAS mutations and acquired resistance to anti-EGFR therapy in colorectal cancer. Nature (2012) 486:532-6. doi:10.1038/nature11156.
    • (2012) Nature , vol.486 , pp. 532-536
    • Misale, S.1    Yaeger, R.2    Hobor, S.3    Scala, E.4    Janakiraman, M.5    Liska, D.6
  • 75
    • 84891894051 scopus 로고    scopus 로고
    • The genetic landscape of clinical resistance to RAF inhibition in metastatic melanoma
    • Van Allen EM, Wagle N, Sucker A, Treacy DJ, Johannessen CM, Goetz EM, et al. The genetic landscape of clinical resistance to RAF inhibition in metastatic melanoma. Cancer Discov (2014) 4:94-109. doi:10.1158/2159-8290.CD-13-0617.
    • (2014) Cancer Discov , vol.4 , pp. 94-109
    • Van Allen, E.M.1    Wagle, N.2    Sucker, A.3    Treacy, D.J.4    Johannessen, C.M.5    Goetz, E.M.6
  • 76
    • 79953325104 scopus 로고    scopus 로고
    • PTEN loss confers BRAF inhibitor resistance to melanoma cells through the suppression of BIM expression
    • Paraiso KH, Xiang Y, Rebecca VW, Abel EV, Chen YA, Munko AC, et al. PTEN loss confers BRAF inhibitor resistance to melanoma cells through the suppression of BIM expression. Cancer Res (2011) 71:2750-60. doi:10.1158/0008-5472.CAN-10-2954.
    • (2011) Cancer Res , vol.71 , pp. 2750-2760
    • Paraiso, K.H.1    Xiang, Y.2    Rebecca, V.W.3    Abel, E.V.4    Chen, Y.A.5    Munko, A.C.6
  • 77
    • 84887014270 scopus 로고    scopus 로고
    • The FBXO4 tumor suppressor functions as a barrier to BrafV600E-dependent metastatic melanoma
    • Lee EK, Lian Z, D'Andrea K, Letrero R, Sheng W, Liu S, et al. The FBXO4 tumor suppressor functions as a barrier to BrafV600E-dependent metastatic melanoma. Mol Cell Biol (2013) 33:4422-33. doi:10.1128/MCB.00706-13.
    • (2013) Mol Cell Biol , vol.33 , pp. 4422-4433
    • Lee, E.K.1    Lian, Z.2    D'Andrea, K.3    Letrero, R.4    Sheng, W.5    Liu, S.6
  • 78
    • 84875717480 scopus 로고    scopus 로고
    • A genome-scale RNA interference screen implicates NF1 loss in resistance to RAF inhibition
    • Whittaker SR, Theurillat JP, Van Allen E, Wagle N, Hsiao J, Cowley GS, et al. A genome-scale RNA interference screen implicates NF1 loss in resistance to RAF inhibition. Cancer Discov (2013) 3:350-62. doi:10.1158/2159-8290.CD-12-0470.
    • (2013) Cancer Discov , vol.3 , pp. 350-362
    • Whittaker, S.R.1    Theurillat, J.P.2    Van Allen, E.3    Wagle, N.4    Hsiao, J.5    Cowley, G.S.6
  • 79
    • 79958785634 scopus 로고    scopus 로고
    • Protein kinase D3 sensitizes RAF inhibitor RAF265 in melanoma cells by preventing reactivation of MAPK signaling
    • Chen J, Shen Q, Labow M, Gaither LA. Protein kinase D3 sensitizes RAF inhibitor RAF265 in melanoma cells by preventing reactivation of MAPK signaling. Cancer Res (2011) 71:4280-91. doi:10.1158/0008-5472.CAN-10-3761.
    • (2011) Cancer Res , vol.71 , pp. 4280-4291
    • Chen, J.1    Shen, Q.2    Labow, M.3    Gaither, L.A.4
  • 80
    • 84899470984 scopus 로고    scopus 로고
    • Differential activity of MEK and ERK inhibitors in BRAF inhibitor resistant melanoma
    • Carlino MS, Todd JR, Gowrishankar K, Mijatov B, Pupo GM, Fung C, et al. Differential activity of MEK and ERK inhibitors in BRAF inhibitor resistant melanoma. Mol Oncol (2014) 8:544-54. doi:10.1016/j.molonc.2014.01.003.
    • (2014) Mol Oncol , vol.8 , pp. 544-554
    • Carlino, M.S.1    Todd, J.R.2    Gowrishankar, K.3    Mijatov, B.4    Pupo, G.M.5    Fung, C.6
  • 81
    • 84899513979 scopus 로고    scopus 로고
    • Loss of NF1 in cutaneous melanoma is associated with RAS activation and MEK dependence
    • Nissan MH, Pratilas CA, Jones AM, Ramirez R, Won H, Liu C, et al. Loss of NF1 in cutaneous melanoma is associated with RAS activation and MEK dependence. Cancer Res (2014) 74:2340-50. doi:10.1158/0008-5472.CAN-13-2625.
    • (2014) Cancer Res , vol.74 , pp. 2340-2350
    • Nissan, M.H.1    Pratilas, C.A.2    Jones, A.M.3    Ramirez, R.4    Won, H.5    Liu, C.6
  • 82
    • 83455254767 scopus 로고    scopus 로고
    • RAF inhibitor resistance is mediated by dimerization of aberrantly spliced BRAF(V600E)
    • Poulikakos PI, Persaud Y, Janakiraman M, Kong X, Ng C, Moriceau G, et al. RAF inhibitor resistance is mediated by dimerization of aberrantly spliced BRAF(V600E). Nature (2011) 480:387-90. doi:10.1038/nature10662.
    • (2011) Nature , vol.480 , pp. 387-390
    • Poulikakos, P.I.1    Persaud, Y.2    Janakiraman, M.3    Kong, X.4    Ng, C.5    Moriceau, G.6
  • 83
    • 78650303507 scopus 로고    scopus 로고
    • Melanomas acquire resistance to B-RAF(V600E) inhibition by RTK or N-RAS upregulation
    • Nazarian R, Shi H, Wang Q, Kong X, Koya RC, Lee H, et al. Melanomas acquire resistance to B-RAF(V600E) inhibition by RTK or N-RAS upregulation. Nature (2010) 468:973-7. doi:10.1038/nature09626.
    • (2010) Nature , vol.468 , pp. 973-977
    • Nazarian, R.1    Shi, H.2    Wang, Q.3    Kong, X.4    Koya, R.C.5    Lee, H.6
  • 84
    • 84925303187 scopus 로고    scopus 로고
    • EPHA2 is a mediator of vemurafenib resistance and a novel therapeutic target in melanoma
    • Miao B, Ji Z, Tan L, Taylor M, Zhang J, Choi HG, et al. EPHA2 is a mediator of vemurafenib resistance and a novel therapeutic target in melanoma. Cancer Discov (2015) 5:274-87. doi:10.1158/2159-8290.CD-14-0295.
    • (2015) Cancer Discov , vol.5 , pp. 274-287
    • Miao, B.1    Ji, Z.2    Tan, L.3    Taylor, M.4    Zhang, J.5    Choi, H.G.6
  • 85
    • 84925297806 scopus 로고    scopus 로고
    • Ligand-independent EPHA2 signaling drives the adoption of a targeted therapy-mediated metastatic melanoma phenotype
    • Paraiso KH, Das Thakur M, Fang B, Koomen JM, Fedorenko IV, John JK, et al. Ligand-independent EPHA2 signaling drives the adoption of a targeted therapy-mediated metastatic melanoma phenotype. Cancer Discov (2015) 5:264-73. doi:10.1158/2159-8290.CD-14-0293.
    • (2015) Cancer Discov , vol.5 , pp. 264-273
    • Paraiso, K.H.1    Das Thakur, M.2    Fang, B.3    Koomen, J.M.4    Fedorenko, I.V.5    John, J.K.6
  • 86
    • 84864285794 scopus 로고    scopus 로고
    • Tumour micro-environment elicits innate resistance to RAF inhibitors through HGF secretion
    • Straussman R, Morikawa T, Shee K, Barzily-Rokni M, Qian ZR, Du J, et al. Tumour micro-environment elicits innate resistance to RAF inhibitors through HGF secretion. Nature (2012) 487:500-4. doi:10.1038/nature11183.
    • (2012) Nature , vol.487 , pp. 500-504
    • Straussman, R.1    Morikawa, T.2    Shee, K.3    Barzily-Rokni, M.4    Qian, Z.R.5    Du, J.6
  • 87
    • 84880057441 scopus 로고    scopus 로고
    • Antiproliferative effects of continued mitogen-activated protein kinase pathway inhibition following acquired resistance to BRAF and/or MEK inhibition in melanoma
    • Carlino MS, Gowrishankar K, Saunders CA, Pupo GM, Snoyman S, Zhang XD, et al. Antiproliferative effects of continued mitogen-activated protein kinase pathway inhibition following acquired resistance to BRAF and/or MEK inhibition in melanoma. Mol Cancer Ther (2013) 12:1332-42. doi:10.1158/1535-7163.MCT-13-0011.
    • (2013) Mol Cancer Ther , vol.12 , pp. 1332-1342
    • Carlino, M.S.1    Gowrishankar, K.2    Saunders, C.A.3    Pupo, G.M.4    Snoyman, S.5    Zhang, X.D.6
  • 88
    • 78650008177 scopus 로고    scopus 로고
    • Acquired resistance to BRAF inhibitors mediated by a RAF kinase switch in melanoma can be overcome by cotargeting MEK and IGF-1R/PI3K
    • Villanueva J, Vultur A, Lee JT, Somasundaram R, Fukunaga-Kalabis M, Cipolla AK, et al. Acquired resistance to BRAF inhibitors mediated by a RAF kinase switch in melanoma can be overcome by cotargeting MEK and IGF-1R/PI3K. Cancer Cell (2010) 18:683-95. doi:10.1016/j.ccr.2010.11.023.
    • (2010) Cancer Cell , vol.18 , pp. 683-695
    • Villanueva, J.1    Vultur, A.2    Lee, J.T.3    Somasundaram, R.4    Fukunaga-Kalabis, M.5    Cipolla, A.K.6
  • 89
    • 84880325633 scopus 로고    scopus 로고
    • Stat3-targeted therapies overcome the acquired resistance to vemurafenib in melanomas
    • Liu F, Cao J, Wu J, Sullivan K, Shen J, Ryu B, et al. Stat3-targeted therapies overcome the acquired resistance to vemurafenib in melanomas. J Invest Dermatol (2013) 133:2041-9. doi:10.1038/jid.2013.32.
    • (2013) J Invest Dermatol , vol.133 , pp. 2041-2049
    • Liu, F.1    Cao, J.2    Wu, J.3    Sullivan, K.4    Shen, J.5    Ryu, B.6
  • 90
    • 84898016171 scopus 로고    scopus 로고
    • MEK inhibition affects STAT3 signaling and invasion in human melanoma cell lines
    • Vultur A, Villanueva J, Krepler C, Rajan G, Chen Q, Xiao M, et al. MEK inhibition affects STAT3 signaling and invasion in human melanoma cell lines. Oncogene (2014) 33:1850-61. doi:10.1038/onc.2013.131.
    • (2014) Oncogene , vol.33 , pp. 1850-1861
    • Vultur, A.1    Villanueva, J.2    Krepler, C.3    Rajan, G.4    Chen, Q.5    Xiao, M.6
  • 92
    • 84864994126 scopus 로고    scopus 로고
    • Reactivation of mitogen-activated protein kinase (MAPK) pathway by FGF receptor 3 (FGFR3)/Ras mediates resistance to vemurafenib in human B-RAF V600E mutant melanoma
    • Yadav V, Zhang X, Liu J, Estrem S, Li S, Gong XQ, et al. Reactivation of mitogen-activated protein kinase (MAPK) pathway by FGF receptor 3 (FGFR3)/Ras mediates resistance to vemurafenib in human B-RAF V600E mutant melanoma. J Biol Chem (2012) 287:28087-98. doi:10.1074/jbc.M112.377218.
    • (2012) J Biol Chem , vol.287 , pp. 28087-28098
    • Yadav, V.1    Zhang, X.2    Liu, J.3    Estrem, S.4    Li, S.5    Gong, X.Q.6
  • 93
    • 84894237511 scopus 로고    scopus 로고
    • Dermatologic toxicities to targeted cancer therapy: shared clinical and histologic adverse skin reactions
    • Curry JL, Torres-Cabala CA, Kim KB, Tetzlaff MT, Duvic M, Tsai KY, et al. Dermatologic toxicities to targeted cancer therapy: shared clinical and histologic adverse skin reactions. Int J Dermatol (2014) 53:376-84. doi:10.1111/ijd.12205.
    • (2014) Int J Dermatol , vol.53 , pp. 376-384
    • Curry, J.L.1    Torres-Cabala, C.A.2    Kim, K.B.3    Tetzlaff, M.T.4    Duvic, M.5    Tsai, K.Y.6
  • 94
    • 77949685981 scopus 로고    scopus 로고
    • RAF inhibitors prime wild-type RAF to activate the MAPK pathway and enhance growth
    • Hatzivassiliou G, Song K, Yen I, Brandhuber BJ, Anderson DJ, Alvarado R, et al. RAF inhibitors prime wild-type RAF to activate the MAPK pathway and enhance growth. Nature (2010) 464:431-5. doi:10.1038/nature08833.
    • (2010) Nature , vol.464 , pp. 431-435
    • Hatzivassiliou, G.1    Song, K.2    Yen, I.3    Brandhuber, B.J.4    Anderson, D.J.5    Alvarado, R.6
  • 95
    • 74849109743 scopus 로고    scopus 로고
    • Kinase-dead BRAF and oncogenic RAS cooperate to drive tumor progression through CRAF
    • Heidorn SJ, Milagre C, Whittaker S, Nourry A, Niculescu-Duvas I, Dhomen N, et al. Kinase-dead BRAF and oncogenic RAS cooperate to drive tumor progression through CRAF. Cell (2010) 140:209-21. doi:10.1016/j.cell.2009.12.040.
    • (2010) Cell , vol.140 , pp. 209-221
    • Heidorn, S.J.1    Milagre, C.2    Whittaker, S.3    Nourry, A.4    Niculescu-Duvas, I.5    Dhomen, N.6
  • 96
    • 77949732073 scopus 로고    scopus 로고
    • RAF inhibitors transactivate RAF dimers and ERK signalling in cells with wild-type BRAF
    • Poulikakos PI, Zhang C, Bollag G, Shokat KM, Rosen N. RAF inhibitors transactivate RAF dimers and ERK signalling in cells with wild-type BRAF. Nature (2010) 464:427-30. doi:10.1038/nature08902.
    • (2010) Nature , vol.464 , pp. 427-430
    • Poulikakos, P.I.1    Zhang, C.2    Bollag, G.3    Shokat, K.M.4    Rosen, N.5
  • 97
    • 84881668573 scopus 로고    scopus 로고
    • Targeting BRAF in melanoma: biological and clinical challenges
    • Mandalà M, Voit C. Targeting BRAF in melanoma: biological and clinical challenges. Crit Rev Oncol Hematol (2013) 87:239-55. doi:10.1016/j.critrevonc.2013.01.003.
    • (2013) Crit Rev Oncol Hematol , vol.87 , pp. 239-255
    • Mandalà, M.1    Voit, C.2
  • 98
    • 84886385231 scopus 로고    scopus 로고
    • Identification of multiple mechanisms of resistance to vemurafenib in a patient with BRAFV600E-mutated cutaneous melanoma successfully rechallenged after progression
    • Romano E, Pradervand S, Paillusson A, Weber J, Harshman K, Muehlethaler K, et al. Identification of multiple mechanisms of resistance to vemurafenib in a patient with BRAFV600E-mutated cutaneous melanoma successfully rechallenged after progression. Clin Cancer Res (2013) 19:5749-57. doi:10.1158/1078-0432.CCR-13-0661.
    • (2013) Clin Cancer Res , vol.19 , pp. 5749-5757
    • Romano, E.1    Pradervand, S.2    Paillusson, A.3    Weber, J.4    Harshman, K.5    Muehlethaler, K.6
  • 99
    • 78649436372 scopus 로고    scopus 로고
    • BRAF gene amplification can promote acquired resistance to MEK inhibitors in cancer cells harboring the BRAF V600E mutation
    • Corcoran RB, Dias-Santagata D, Bergethon K, Iafrate AJ, Settleman J, Engelman JA. BRAF gene amplification can promote acquired resistance to MEK inhibitors in cancer cells harboring the BRAF V600E mutation. Sci Signal (2010) 3:ra84. doi:10.1126/scisignal.2001148.
    • (2010) Sci Signal , vol.3
    • Corcoran, R.B.1    Dias-Santagata, D.2    Bergethon, K.3    Iafrate, A.J.4    Settleman, J.5    Engelman, J.A.6
  • 100
    • 84873728334 scopus 로고    scopus 로고
    • Modelling vemurafenib resistance in melanoma reveals a strategy to forestall drug resistance
    • Dhas Thakur M, Salangsang F, Landman AS, Sellers WR, Pryer NK, Levesque MP, et al. Modelling vemurafenib resistance in melanoma reveals a strategy to forestall drug resistance. Nature (2013) 494:251-5. doi:10.1038/nature11814.
    • (2013) Nature , vol.494 , pp. 251-255
    • Dhas Thakur, M.1    Salangsang, F.2    Landman, A.S.3    Sellers, W.R.4    Pryer, N.K.5    Levesque, M.P.6
  • 101
    • 84864286442 scopus 로고    scopus 로고
    • Widespread potential for growth-factor driven resistance to anticancer kinase inhibitors
    • Wilson TR, Fridlyand J, Yan Y, Penuel E, Burton L, Chan E, et al. Widespread potential for growth-factor driven resistance to anticancer kinase inhibitors. Nature (2012) 487:505-9. doi:10.1038/nature11249.
    • (2012) Nature , vol.487 , pp. 505-509
    • Wilson, T.R.1    Fridlyand, J.2    Yan, Y.3    Penuel, E.4    Burton, L.5    Chan, E.6
  • 102
    • 84866266455 scopus 로고    scopus 로고
    • Disruption of the protein interaction between FAK and IGF-1R inhibits melanoma tumor growth
    • Ucar DA, Kurenova E, Garrett TJ, Cance WG, Nyberg C, Cox A, et al. Disruption of the protein interaction between FAK and IGF-1R inhibits melanoma tumor growth. Cell Cycle (2012) 11:3250-9. doi:10.4161/cc.21611.
    • (2012) Cell Cycle , vol.11 , pp. 3250-3259
    • Ucar, D.A.1    Kurenova, E.2    Garrett, T.J.3    Cance, W.G.4    Nyberg, C.5    Cox, A.6
  • 103
    • 84880372025 scopus 로고    scopus 로고
    • A STATement on vemurafenib-resistant melanoma
    • Hartsough EJ, Aplin AE. A STATement on vemurafenib-resistant melanoma. J Invest Dermatol (2013) 133:1928-9. doi:10.1038/jid.2013.136.
    • (2013) J Invest Dermatol , vol.133 , pp. 1928-1929
    • Hartsough, E.J.1    Aplin, A.E.2
  • 104
    • 84896691873 scopus 로고    scopus 로고
    • Mutant B-RAF-Mcl-1 survival signaling depends on the STAT3 transcription factor
    • Becker TM, Boyd SC, Mijatov B, Gowrishankar K, Snoyman S, Pupo GM, et al. Mutant B-RAF-Mcl-1 survival signaling depends on the STAT3 transcription factor. Oncogene (2014) 33:1158-66. doi:10.1038/onc.2013.45.
    • (2014) Oncogene , vol.33 , pp. 1158-1166
    • Becker, T.M.1    Boyd, S.C.2    Mijatov, B.3    Gowrishankar, K.4    Snoyman, S.5    Pupo, G.M.6
  • 105
    • 79956105175 scopus 로고    scopus 로고
    • STAT3 mediates resistance to MEK inhibitor through microRNA miR-17
    • Dai B, Meng J, Peyton M, Girard L, Bornmann WG, Ji L, et al. STAT3 mediates resistance to MEK inhibitor through microRNA miR-17. Cancer Res (2011) 71:3658-68. doi:10.1158/0008-5472.CAN-10-3647.
    • (2011) Cancer Res , vol.71 , pp. 3658-3668
    • Dai, B.1    Meng, J.2    Peyton, M.3    Girard, L.4    Bornmann, W.G.5    Ji, L.6
  • 106
    • 78650309875 scopus 로고    scopus 로고
    • COT drives resistance to RAF inhibition through MAP kinase pathway reactivation
    • Johannessen CM, Boehm JS, Kim SY, Thomas SR, Wardwell L, Johnson LA, et al. COT drives resistance to RAF inhibition through MAP kinase pathway reactivation. Nature (2010) 468:968-72. doi:10.1038/nature09627.
    • (2010) Nature , vol.468 , pp. 968-972
    • Johannessen, C.M.1    Boehm, J.S.2    Kim, S.Y.3    Thomas, S.R.4    Wardwell, L.5    Johnson, L.A.6
  • 108
    • 84862778070 scopus 로고    scopus 로고
    • Role and therapeutic potential of PI3K-mTOR signaling in de novo resistance to BRAF inhibition
    • Deng W, Gopal YN, Scott A, Chen G, Woodman SE, Davies MA. Role and therapeutic potential of PI3K-mTOR signaling in de novo resistance to BRAF inhibition. Pigment Cell Melanoma Res (2012) 25:248-58. doi:10.1111/j.1755-148X.2011.00950.x.
    • (2012) Pigment Cell Melanoma Res , vol.25 , pp. 248-258
    • Deng, W.1    Gopal, Y.N.2    Scott, A.3    Chen, G.4    Woodman, S.E.5    Davies, M.A.6
  • 109
    • 84891898344 scopus 로고    scopus 로고
    • Acquired resistance and clonal evolution in melanoma during BRAF inhibitor therapy
    • Shi H, Hugo W, Kong X, Hong A, Koya RC, Moriceau G, et al. Acquired resistance and clonal evolution in melanoma during BRAF inhibitor therapy. Cancer Discov (2014) 4:80-93. doi:10.1158/2159-8290.CD-13-0642.
    • (2014) Cancer Discov , vol.4 , pp. 80-93
    • Shi, H.1    Hugo, W.2    Kong, X.3    Hong, A.4    Koya, R.C.5    Moriceau, G.6
  • 110
    • 84871998076 scopus 로고    scopus 로고
    • An epithelial-mesenchymal transition gene signature predicts resistance to EGFR and PI3K inhibitors and identifies Axl as a therapeutic target for overcoming EGFR inhibitor resistance
    • Byers LA, Diao L, Wang J, Saintigny P, Girard L, Peyton M, et al. An epithelial-mesenchymal transition gene signature predicts resistance to EGFR and PI3K inhibitors and identifies Axl as a therapeutic target for overcoming EGFR inhibitor resistance. Clin Cancer Res (2013) 19:279-90. doi:10.1158/1078-0432.CCR-12-1558.
    • (2013) Clin Cancer Res , vol.19 , pp. 279-290
    • Byers, L.A.1    Diao, L.2    Wang, J.3    Saintigny, P.4    Girard, L.5    Peyton, M.6
  • 111
    • 84909594601 scopus 로고    scopus 로고
    • Phenotype switching: tumor cell plasticity as a resistance mechanism and target for therapy
    • Kemper K, de Goeje PL, Peeper DS, van Amerongen R. Phenotype switching: tumor cell plasticity as a resistance mechanism and target for therapy. Cancer Res (2014) 74:5937-41. doi:10.1158/0008-5472.CAN-14-1174.
    • (2014) Cancer Res , vol.74 , pp. 5937-5941
    • Kemper, K.1    de Goeje, P.L.2    Peeper, D.S.3    van Amerongen, R.4
  • 113
    • 33747882661 scopus 로고    scopus 로고
    • MITF: master regulator of melanocyte development and melanoma oncogene
    • Levy C, Khaled M, Fisher DE. MITF: master regulator of melanocyte development and melanoma oncogene. Trends Mol Med (2006) 12:406-14. doi:10.1016/j.molmed.2006.07.008.
    • (2006) Trends Mol Med , vol.12 , pp. 406-414
    • Levy, C.1    Khaled, M.2    Fisher, D.E.3
  • 114
    • 10944228764 scopus 로고    scopus 로고
    • Critical role of CDK2 for melanoma growth linked to its melanocyte-specific transcriptional regulation by MITF
    • Du J, Widlund HR, Horstmann MA, Ramaswamy S, Ross K, Huber WE, et al. Critical role of CDK2 for melanoma growth linked to its melanocyte-specific transcriptional regulation by MITF. Cancer Cell (2004) 6:565-76. doi:10.1016/j.ccr.2004.10.014.
    • (2004) Cancer Cell , vol.6 , pp. 565-576
    • Du, J.1    Widlund, H.R.2    Horstmann, M.A.3    Ramaswamy, S.4    Ross, K.5    Huber, W.E.6
  • 115
    • 79952601233 scopus 로고    scopus 로고
    • A proliferative melanoma cell phenotype is responsive to RAF/MEK inhibition independent of BRAF mutation status
    • Zipser MC, Eichhoff OM, Widmer DS, Schlegel NC, Schoenewolf NL, Stuart D, et al. A proliferative melanoma cell phenotype is responsive to RAF/MEK inhibition independent of BRAF mutation status. Pigment Cell Melanoma Res (2011) 24:326-33. doi:10.1111/j.1755-148X.2010.00823.x.
    • (2011) Pigment Cell Melanoma Res , vol.24 , pp. 326-333
    • Zipser, M.C.1    Eichhoff, O.M.2    Widmer, D.S.3    Schlegel, N.C.4    Schoenewolf, N.L.5    Stuart, D.6
  • 116
    • 84863393080 scopus 로고    scopus 로고
    • Intratumor heterogeneity and branched evolution revealed by multiregion sequencing
    • Gerlinger M, Rowan AJ, Horswell S, Larkin J, Endesfelder D, Gronroos E, et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N Engl J Med (2012) 366:883-92. doi:10.1056/NEJMoa1113205.
    • (2012) N Engl J Med , vol.366 , pp. 883-892
    • Gerlinger, M.1    Rowan, A.J.2    Horswell, S.3    Larkin, J.4    Endesfelder, D.5    Gronroos, E.6
  • 117
    • 84954201325 scopus 로고    scopus 로고
    • Intratumoral heterogeneity: clonal cooperation in epithelial-to-mesenchymal transition and metastasis
    • Neelakantan D, Drasin DJ, Ford HL. Intratumoral heterogeneity: clonal cooperation in epithelial-to-mesenchymal transition and metastasis. Cell Adh Migr (2014) 16:0. doi:10.4161/19336918.2014.972761.
    • (2014) Cell Adh Migr , vol.16 , pp. 0
    • Neelakantan, D.1    Drasin, D.J.2    Ford, H.L.3
  • 118
    • 37649005234 scopus 로고    scopus 로고
    • Autophagy in the pathogenesis of disease
    • Levine B, Kroemer G. Autophagy in the pathogenesis of disease. Cell (2008) 132:27-42. doi:10.1016/j.cell.2007.12.018.
    • (2008) Cell , vol.132 , pp. 27-42
    • Levine, B.1    Kroemer, G.2
  • 119
    • 23344446037 scopus 로고    scopus 로고
    • Autophagy
    • Klionsky DJ. Autophagy. Curr Biol (2005) 15:R282-3. doi:10.1016/j.cub.2005.04.013.
    • (2005) Curr Biol , vol.15 , pp. R282-R283
    • Klionsky, D.J.1
  • 120
    • 34848886914 scopus 로고    scopus 로고
    • Autophagosome formation: core machinery and adaptations
    • Xie Z, Klionsky DJ. Autophagosome formation: core machinery and adaptations. Nat Cell Biol (2007) 9:1102-9. doi:10.1038/ncb1007-1102.
    • (2007) Nat Cell Biol , vol.9 , pp. 1102-1109
    • Xie, Z.1    Klionsky, D.J.2
  • 121
    • 77955708390 scopus 로고    scopus 로고
    • Overview of macroautophagy regulation in mammalian cells
    • Mehrpour M, Esclatine A, Beau I, Codogno P. Overview of macroautophagy regulation in mammalian cells. Cell Res (2010) 20:748-62. doi:10.1038/cr.2010.82.
    • (2010) Cell Res , vol.20 , pp. 748-762
    • Mehrpour, M.1    Esclatine, A.2    Beau, I.3    Codogno, P.4
  • 123
    • 77954761710 scopus 로고    scopus 로고
    • AMP-activated kinase (AMPK)-generated signals in malignant melanoma cell growth and survival
    • Woodard J, Platanias LC. AMP-activated kinase (AMPK)-generated signals in malignant melanoma cell growth and survival. Biochem Biophys Res Commun (2010) 398:135-9. doi:10.1016/j.bbrc.2010.06.052.
    • (2010) Biochem Biophys Res Commun , vol.398 , pp. 135-139
    • Woodard, J.1    Platanias, L.C.2
  • 124
    • 77956401999 scopus 로고    scopus 로고
    • Metformin and other biguanides in oncology: advancing the research agenda
    • Pollak M. Metformin and other biguanides in oncology: advancing the research agenda. Cancer Prev Res (Phila) (2010) 3:1060-5. doi:10.1158/1940-6207.CAPR-10-0175.
    • (2010) Cancer Prev Res (Phila) , vol.3 , pp. 1060-1065
    • Pollak, M.1
  • 126
    • 0034614420 scopus 로고    scopus 로고
    • Dimethylbiguanide inhibits cell respiration via an indirect effect targeted on the respiratory chain complex I
    • El-Mir MY, Nogueira V, Fontaine E, Averet N, Rigoulet M, Leverve X. Dimethylbiguanide inhibits cell respiration via an indirect effect targeted on the respiratory chain complex I. J Biol Chem (2000) 275:223-8. doi:10.1074/jbc.275.1.223.
    • (2000) J Biol Chem , vol.275 , pp. 223-228
    • El-Mir, M.Y.1    Nogueira, V.2    Fontaine, E.3    Averet, N.4    Rigoulet, M.5    Leverve, X.6
  • 127
    • 0034659785 scopus 로고    scopus 로고
    • Evidence that metformin exerts its anti-diabetic effects through inhibition of complex 1 of the mitochondrial respiratory chain
    • Owen MR, Doran E, Halestrap AP. Evidence that metformin exerts its anti-diabetic effects through inhibition of complex 1 of the mitochondrial respiratory chain. Biochem J (2000) 348:607-14. doi:10.1042/0264-6021:3480607.
    • (2000) Biochem J , vol.348 , pp. 607-614
    • Owen, M.R.1    Doran, E.2    Halestrap, A.P.3
  • 128
    • 1542618348 scopus 로고    scopus 로고
    • The tumor suppressor LKB1 kinase directly activates AMP-activated kinase and regulates apoptosis in response to energy stress
    • Shaw RJ, Kosmatka M, Bardeesy N, Hurley RL, Witters LA, Depinho RA, et al. The tumor suppressor LKB1 kinase directly activates AMP-activated kinase and regulates apoptosis in response to energy stress. Proc Natl Acad Sci U S A (2004) 101:3329-35. doi:10.1073/pnas.0308061100.
    • (2004) Proc Natl Acad Sci U S A , vol.101 , pp. 3329-3335
    • Shaw, R.J.1    Kosmatka, M.2    Bardeesy, N.3    Hurley, R.L.4    Witters, L.A.5    Depinho, R.A.6
  • 129
    • 77949462458 scopus 로고    scopus 로고
    • AMPK as a metabolic tumor suppressor: control of metabolism and cell growth
    • Luo Z, Zang M, Guo W. AMPK as a metabolic tumor suppressor: control of metabolism and cell growth. Future Oncol (2010) 6:457-70. doi:10.2217/fon.09.174.
    • (2010) Future Oncol , vol.6 , pp. 457-470
    • Luo, Z.1    Zang, M.2    Guo, W.3
  • 130
    • 34547114031 scopus 로고    scopus 로고
    • Systemic treatment with the antidiabetic drug metformin selectively impairs p53-deficient tumor cell growth
    • Buzzai M, Jones RG, Amaravadi RK, Lum JJ, Deberardinis RJ, Zhao F, et al. Systemic treatment with the antidiabetic drug metformin selectively impairs p53-deficient tumor cell growth. Cancer Res (2007) 67:6745-52. doi:10.1158/0008-5472.CAN-06-4447.
    • (2007) Cancer Res , vol.67 , pp. 6745-6752
    • Buzzai, M.1    Jones, R.G.2    Amaravadi, R.K.3    Lum, J.J.4    Deberardinis, R.J.5    Zhao, F.6
  • 131
    • 77950191479 scopus 로고    scopus 로고
    • Targeting cancer cell metabolism: the combination of metformin and 2-deoxyglucose induces p53-dependent apoptosis in prostate cancer cells
    • Ben Sahra I, Laurent K, Giuliano S, Larbret F, Ponzio G, Gounon P, et al. Targeting cancer cell metabolism: the combination of metformin and 2-deoxyglucose induces p53-dependent apoptosis in prostate cancer cells. Cancer Res (2010) 70:2465-75. doi:10.1158/0008-5472.CAN-09-2782.
    • (2010) Cancer Res , vol.70 , pp. 2465-2475
    • Ben Sahra, I.1    Laurent, K.2    Giuliano, S.3    Larbret, F.4    Ponzio, G.5    Gounon, P.6
  • 132
    • 80053417028 scopus 로고    scopus 로고
    • Metformin inhibits melanoma development through autophagy and apoptosis mechanisms
    • Tomic T, Botton T, Cerezo M, Robert G, Luciano F, Puissant A, et al. Metformin inhibits melanoma development through autophagy and apoptosis mechanisms. Cell Death Dis (2011) 2:e199. doi:10.1038/cddis.2011.86.
    • (2011) Cell Death Dis , vol.2
    • Tomic, T.1    Botton, T.2    Cerezo, M.3    Robert, G.4    Luciano, F.5    Puissant, A.6
  • 133
    • 84866256417 scopus 로고    scopus 로고
    • AMPK activators inhibit the proliferation of human melanomas bearing the activated MAPK pathway
    • Petti C, Vegetti C, Molla A, Bersani I, Cleris L, Mustard KJ, et al. AMPK activators inhibit the proliferation of human melanomas bearing the activated MAPK pathway. Melanoma Res (2012) 22:341-50. doi:10.1097/CMR.0b013e3283544929.
    • (2012) Melanoma Res , vol.22 , pp. 341-350
    • Petti, C.1    Vegetti, C.2    Molla, A.3    Bersani, I.4    Cleris, L.5    Mustard, K.J.6
  • 134
    • 84887296747 scopus 로고    scopus 로고
    • Phenformin enhances the therapeutic benefit of BRAF(V600E) inhibition in melanoma
    • Yuan P, Ito K, Perez-Lorenzo R, Del Guzzo C, Lee JH, Shen CH, et al. Phenformin enhances the therapeutic benefit of BRAF(V600E) inhibition in melanoma. Proc Natl Acad Sci U S A (2013) 110:18226-31. doi:10.1073/pnas.1317577110.
    • (2013) Proc Natl Acad Sci U S A , vol.110 , pp. 18226-18231
    • Yuan, P.1    Ito, K.2    Perez-Lorenzo, R.3    Del Guzzo, C.4    Lee, J.H.5    Shen, C.H.6
  • 135
    • 79956260998 scopus 로고    scopus 로고
    • Combination therapy with vemurafenib (PLX4032/RG7204) and metformin in melanoma cell lines with distinct driver mutations
    • Niehr F, von Euw E, Attar N, Guo D, Matsunaga D, Sazegar H, et al. Combination therapy with vemurafenib (PLX4032/RG7204) and metformin in melanoma cell lines with distinct driver mutations. J Transl Med (2011) 9:76. doi:10.1186/1479-5876-9-76.
    • (2011) J Transl Med , vol.9 , pp. 76
    • Niehr, F.1    von Euw, E.2    Attar, N.3    Guo, D.4    Matsunaga, D.5    Sazegar, H.6
  • 136
    • 84878959237 scopus 로고    scopus 로고
    • Overcoming intrinsic multidrug resistance in melanoma by blocking the mitochondrial respiratory chain of slow-cycling JARID1B(high) cells
    • Roesch A, Vultur A, Bogeski I, Wang H, Zimmermann KM, Speicher D, et al. Overcoming intrinsic multidrug resistance in melanoma by blocking the mitochondrial respiratory chain of slow-cycling JARID1B(high) cells. Cancer Cell (2013) 23:811-25. doi:10.1016/j.ccr.2013.05.003.
    • (2013) Cancer Cell , vol.23 , pp. 811-825
    • Roesch, A.1    Vultur, A.2    Bogeski, I.3    Wang, H.4    Zimmermann, K.M.5    Speicher, D.6
  • 137
    • 84891918702 scopus 로고    scopus 로고
    • Secretome from senescent melanoma engages the STAT3 pathway to favor reprogramming of naive melanoma towards a tumor-initiating cell phenotype
    • Ohanna M, Cheli Y, Bonet C, Bonazzi VF, Allegra M, Giuliano S, et al. Secretome from senescent melanoma engages the STAT3 pathway to favor reprogramming of naive melanoma towards a tumor-initiating cell phenotype. Oncotarget (2013) 4:2212-24.
    • (2013) Oncotarget , vol.4 , pp. 2212-2224
    • Ohanna, M.1    Cheli, Y.2    Bonet, C.3    Bonazzi, V.F.4    Allegra, M.5    Giuliano, S.6
  • 138
    • 78049237405 scopus 로고    scopus 로고
    • Cancer stem cells versus phenotype-switching in melanoma
    • Hoek KS, Goding CR. Cancer stem cells versus phenotype-switching in melanoma. Pigment Cell Melanoma Res (2010) 23:746-59. doi:10.1111/j.1755-148X.2010.00757.x.
    • (2010) Pigment Cell Melanoma Res , vol.23 , pp. 746-759
    • Hoek, K.S.1    Goding, C.R.2
  • 139
    • 84919860686 scopus 로고    scopus 로고
    • Is it time to test biguanide metformin in the treatment of melanoma?
    • Cerezo M, Tomic T, Ballotti R, Rocchi S. Is it time to test biguanide metformin in the treatment of melanoma? Pigment Cell Melanoma Res (2015) 28:8-20. doi:10.1111/pcmr.12267.
    • (2015) Pigment Cell Melanoma Res , vol.28 , pp. 8-20
    • Cerezo, M.1    Tomic, T.2    Ballotti, R.3    Rocchi, S.4
  • 141
    • 84908894973 scopus 로고    scopus 로고
    • Genome-wide analysis of noncoding regulatory mutations in cancer
    • Weinhold N, Jacobsen A, Schultz N, Sander C, Lee W. Genome-wide analysis of noncoding regulatory mutations in cancer. Nat Genet (2014) 46:1160-5. doi:10.1038/ng.3101.
    • (2014) Nat Genet , vol.46 , pp. 1160-1165
    • Weinhold, N.1    Jacobsen, A.2    Schultz, N.3    Sander, C.4    Lee, W.5
  • 142
    • 84874191269 scopus 로고    scopus 로고
    • TERT promoter mutations in familial and sporadic melanoma
    • Horn S, Figl A, Rachakonda PS, Fischer C, Sucker A, Gast A, et al. TERT promoter mutations in familial and sporadic melanoma. Science (2013) 339:959-61. doi:10.1126/science.1230062.
    • (2013) Science , vol.339 , pp. 959-961
    • Horn, S.1    Figl, A.2    Rachakonda, P.S.3    Fischer, C.4    Sucker, A.5    Gast, A.6
  • 143
    • 84874189784 scopus 로고    scopus 로고
    • Highly recurrent TERT promoter mutations in human melanoma
    • Huang FW, Hodis E, Xu MJ, Kryukov GV, Chin L, Garraway LA. Highly recurrent TERT promoter mutations in human melanoma. Science (2013) 339:957-9. doi:10.1126/science.1229259.
    • (2013) Science , vol.339 , pp. 957-959
    • Huang, F.W.1    Hodis, E.2    Xu, M.J.3    Kryukov, G.V.4    Chin, L.5    Garraway, L.A.6
  • 144
    • 84865461263 scopus 로고    scopus 로고
    • BAP1 cancer syndrome: malignant mesothelioma, uveal and cutaneous melanoma, and MBAITs
    • Carbone M, Ferris LK, Baumann F, Napolitano A, Lum CA, Flores EG, et al. BAP1 cancer syndrome: malignant mesothelioma, uveal and cutaneous melanoma, and MBAITs. J Transl Med (2012) 10:179. doi:10.1186/1479-5876-10-179.
    • (2012) J Transl Med , vol.10 , pp. 179
    • Carbone, M.1    Ferris, L.K.2    Baumann, F.3    Napolitano, A.4    Lum, C.A.5    Flores, E.G.6
  • 145
    • 84899648964 scopus 로고    scopus 로고
    • Rare missense variants in POT1 predispose to familial cutaneous malignant melanoma
    • Shi J, Yang XR, Ballew B, Rotunno M, Calista D, Fargnoli MC, et al. Rare missense variants in POT1 predispose to familial cutaneous malignant melanoma. Nat Genet (2014) 46:482-6. doi:10.1038/ng.2941.
    • (2014) Nat Genet , vol.46 , pp. 482-486
    • Shi, J.1    Yang, X.R.2    Ballew, B.3    Rotunno, M.4    Calista, D.5    Fargnoli, M.C.6
  • 146
    • 0038451396 scopus 로고    scopus 로고
    • POT1 as a terminal transducer of TRF1 telomere length control
    • Loayza D, De Lange T. POT1 as a terminal transducer of TRF1 telomere length control. Nature (2003) 423:1013-8. doi:10.1038/nature01688.
    • (2003) Nature , vol.423 , pp. 1013-1018
    • Loayza, D.1    De Lange, T.2
  • 147
    • 46249125488 scopus 로고    scopus 로고
    • How shelterin protects mammalian telomeres
    • Palm W, de Lange T. How shelterin protects mammalian telomeres. Annu Rev Genet (2008) 42:301-34. doi:10.1146/annurev.genet.41.110306.130350.
    • (2008) Annu Rev Genet , vol.42 , pp. 301-334
    • Palm, W.1    de Lange, T.2
  • 149
    • 84895925247 scopus 로고    scopus 로고
    • Context-specific regulation of cancer epigenomes by histone and transcription factor methylation
    • Sarris M, Nikolaou K, Talianidis I. Context-specific regulation of cancer epigenomes by histone and transcription factor methylation. Oncogene (2012) 33:1207-17. doi:10.1038/onc.2013.87.
    • (2012) Oncogene , vol.33 , pp. 1207-1217
    • Sarris, M.1    Nikolaou, K.2    Talianidis, I.3
  • 150
    • 84899720407 scopus 로고    scopus 로고
    • SWI/SNF chromatin remodeling enzymes in melanocyte differentiation and melanoma
    • Mehrotra A, Mehta G, Aras S, Trivedi A, de la Serna IL. SWI/SNF chromatin remodeling enzymes in melanocyte differentiation and melanoma. Crit Rev Eukaryot Gene Expr (2014) 24:151-61. doi:10.1615/CritRevEukaryotGeneExpr.2014007882.
    • (2014) Crit Rev Eukaryot Gene Expr , vol.24 , pp. 151-161
    • Mehrotra, A.1    Mehta, G.2    Aras, S.3    Trivedi, A.4    de la Serna, I.L.5
  • 151
    • 84872062248 scopus 로고    scopus 로고
    • JARID1B expression in human melanoma and benign melanocytic skin lesions
    • Kuźbicki L, Lange D, Straczyńska-Niemiec A, Chwirot BW. JARID1B expression in human melanoma and benign melanocytic skin lesions. Melanoma Res (2013) 23:8-12. doi:10.1097/CMR.0b013e32835d5d6f.
    • (2013) Melanoma Res , vol.23 , pp. 8-12
    • Kuźbicki, L.1    Lange, D.2    Straczyńska-Niemiec, A.3    Chwirot, B.W.4
  • 152
    • 84919597213 scopus 로고    scopus 로고
    • miR-203 inhibits melanoma invasive and proliferative abilities by targeting the polycomb group gene BMI1
    • Chang X, Sun Y, Han S, Zhu W, Zhang H, Lian S. miR-203 inhibits melanoma invasive and proliferative abilities by targeting the polycomb group gene BMI1. Biochem Biophys Res Commun (2015) 456:361-6. doi:10.1016/j.bbrc.2014.11.087.
    • (2015) Biochem Biophys Res Commun , vol.456 , pp. 361-366
    • Chang, X.1    Sun, Y.2    Han, S.3    Zhu, W.4    Zhang, H.5    Lian, S.6
  • 153
    • 84919846786 scopus 로고    scopus 로고
    • EZH2: an emerging role in melanoma biology and strategies for targeted therapy
    • Tiffen J, Gallagher SJ, Hersey P. EZH2: an emerging role in melanoma biology and strategies for targeted therapy. Pigment Cell Melanoma Res (2015) 28:21-30. doi:10.1111/pcmr.12280.
    • (2015) Pigment Cell Melanoma Res , vol.28 , pp. 21-30
    • Tiffen, J.1    Gallagher, S.J.2    Hersey, P.3
  • 156
    • 84939894917 scopus 로고    scopus 로고
    • Treatment of resistant metastatic melanoma using sequential epigenetic therapy (decitabine and panobinostat) combined with chemotherapy (temozolomide)
    • Xia C, Leon-Ferre R, Laux D, Deutsch J, Smith BJ, Frees M, et al. Treatment of resistant metastatic melanoma using sequential epigenetic therapy (decitabine and panobinostat) combined with chemotherapy (temozolomide). Cancer Chemother Pharmacol (2014) 74:691-7. doi:10.1007/s00280-014-2501-1.
    • (2014) Cancer Chemother Pharmacol , vol.74 , pp. 691-697
    • Xia, C.1    Leon-Ferre, R.2    Laux, D.3    Deutsch, J.4    Smith, B.J.5    Frees, M.6
  • 158
    • 84857706554 scopus 로고    scopus 로고
    • Melanoma and epigenetic treatment: past and future
    • La Porta CA. Melanoma and epigenetic treatment: past and future. Anticancer Agents Med Chem (2012) 12:202-9. doi:10.2174/187152012800228760.
    • (2012) Anticancer Agents Med Chem , vol.12 , pp. 202-209
    • La Porta, C.A.1
  • 159
    • 84865402404 scopus 로고    scopus 로고
    • Targeting the epigenome for treatment of cancer
    • Geutjes EJ, Bajpe PK, Bernards R. Targeting the epigenome for treatment of cancer. Oncogene (2012) 31:3827-44. doi:10.1038/onc.2011.552.
    • (2012) Oncogene , vol.31 , pp. 3827-3844
    • Geutjes, E.J.1    Bajpe, P.K.2    Bernards, R.3
  • 160
    • 51649095569 scopus 로고    scopus 로고
    • The aurora kinase family in cell division and cancer
    • Vader G, Lens SM. The aurora kinase family in cell division and cancer. Biochim Biophys Acta (2008) 1786:60-72. doi:10.1016/j.bbcan.2008.07.003.
    • (2008) Biochim Biophys Acta , vol.1786 , pp. 60-72
    • Vader, G.1    Lens, S.M.2
  • 161
    • 84857687584 scopus 로고    scopus 로고
    • Pattern formation in centrosome assembly
    • Mahen R, Venkitaraman AR. Pattern formation in centrosome assembly. Curr Opin Cell Biol (2012) 24:14-23. doi:10.1016/j.ceb.2011.12.012.
    • (2012) Curr Opin Cell Biol , vol.24 , pp. 14-23
    • Mahen, R.1    Venkitaraman, A.R.2
  • 162
    • 78649476052 scopus 로고    scopus 로고
    • Shared and separate functions of polo-like kinases and aurora kinases in cancer
    • Lens SM, Voest EE, Medema RH. Shared and separate functions of polo-like kinases and aurora kinases in cancer. Nat Rev Cancer (2010) 10:825-41. doi:10.1038/nrc2964.
    • (2010) Nat Rev Cancer , vol.10 , pp. 825-841
    • Lens, S.M.1    Voest, E.E.2    Medema, R.H.3
  • 164
    • 73549119929 scopus 로고    scopus 로고
    • Aurora-A phosphorylates, activates, and relocalizes the small GTPase RalA
    • Lim KH, Brady DC, Kashatus DF, Ancrile BB, Der CJ, Cox AD, et al. Aurora-A phosphorylates, activates, and relocalizes the small GTPase RalA. Mol Cell Biol (2010) 30:508-23. doi:10.1128/MCB.00916-08.
    • (2010) Mol Cell Biol , vol.30 , pp. 508-523
    • Lim, K.H.1    Brady, D.C.2    Kashatus, D.F.3    Ancrile, B.B.4    Der, C.J.5    Cox, A.D.6
  • 165
    • 33846979444 scopus 로고    scopus 로고
    • Aurora B expression correlates with aggressive behaviour in glioblastoma multiforme
    • Zeng WF, Navaratne K, Prayson RA, Weil RJ. Aurora B expression correlates with aggressive behaviour in glioblastoma multiforme. J Clin Pathol (2007) 60:218-21. doi:10.1136/jcp.2006.036806.
    • (2007) J Clin Pathol , vol.60 , pp. 218-221
    • Zeng, W.F.1    Navaratne, K.2    Prayson, R.A.3    Weil, R.J.4
  • 166
    • 9144251019 scopus 로고    scopus 로고
    • Phosphorylation by aurora kinase A induces Mdm2-mediated destabilization and inhibition of p53
    • Katayama H, Sasai K, Kawai H, Yuan ZM, Bondaruk J, Suzuki F, et al. Phosphorylation by aurora kinase A induces Mdm2-mediated destabilization and inhibition of p53. Nat Genet (2004) 36:55-62. doi:10.1038/ng1279.
    • (2004) Nat Genet , vol.36 , pp. 55-62
    • Katayama, H.1    Sasai, K.2    Kawai, H.3    Yuan, Z.M.4    Bondaruk, J.5    Suzuki, F.6
  • 167
    • 84904845641 scopus 로고    scopus 로고
    • AurkA inhibitors enhance the effects of B-RAF and MEK inhibitors in melanoma treatment
    • Caputo E, Miceli R, Motti ML, Taté R, Fratangelo F, Botti G, et al. AurkA inhibitors enhance the effects of B-RAF and MEK inhibitors in melanoma treatment. J Transl Med (2014) 12:216. doi:10.1186/s12967-014-0216-z.
    • (2014) J Transl Med , vol.12 , pp. 216
    • Caputo, E.1    Miceli, R.2    Motti, M.L.3    Taté, R.4    Fratangelo, F.5    Botti, G.6
  • 168
    • 84926678726 scopus 로고    scopus 로고
    • Beyond BRAF(V600): clinical mutation panel testing by next-generation sequencing in advanced melanoma
    • Siroy AE, Boland GM, Milton DR, Roszik J, Frankian S, Malke J, et al. Beyond BRAF(V600): clinical mutation panel testing by next-generation sequencing in advanced melanoma. J Invest Dermatol (2015) 135:508-15. doi:10.1038/jid.2014.366.
    • (2015) J Invest Dermatol , vol.135 , pp. 508-515
    • Siroy, A.E.1    Boland, G.M.2    Milton, D.R.3    Roszik, J.4    Frankian, S.5    Malke, J.6
  • 170
    • 84905825810 scopus 로고    scopus 로고
    • Combining targeted therapy with immunotherapy in BRAF-mutant melanoma: promise and challenges
    • Hu-Lieskovan S, Robert L, Homet Moreno B, Ribas A. Combining targeted therapy with immunotherapy in BRAF-mutant melanoma: promise and challenges. J Clin Oncol (2014) 32:2248-54. doi:10.1200/JCO.2013.52.1377.
    • (2014) J Clin Oncol , vol.32 , pp. 2248-2254
    • Hu-Lieskovan, S.1    Robert, L.2    Homet Moreno, B.3    Ribas, A.4
  • 171
    • 84937759727 scopus 로고    scopus 로고
    • The next immune-checkpoint inhibitors: PD-1/PD-L1 blockade in melanoma
    • Mahoney KM, Freeman GJ, McDermott DF. The next immune-checkpoint inhibitors: PD-1/PD-L1 blockade in melanoma. Clin Ther (2015) 37(4):764-82. doi:10.1016/j.clinthera.2015.02.018.
    • (2015) Clin Ther , vol.37 , Issue.4 , pp. 764-782
    • Mahoney, K.M.1    Freeman, G.J.2    McDermott, D.F.3
  • 172
    • 84897556601 scopus 로고    scopus 로고
    • Oncogenes in melanoma: an update
    • Kunz M. Oncogenes in melanoma: an update. Eur J Cell Biol (2014) 93:1-10. doi:10.1016/j.ejcb.2013.12.002.
    • (2014) Eur J Cell Biol , vol.93 , pp. 1-10
    • Kunz, M.1


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.