全部 标题 作者
关键词 摘要

OALib Journal期刊
ISSN: 2333-9721
费用:99美元

查看量下载量

相关文章

更多...

Targeting the Cellular Signaling: BRAF Inhibition and Beyond for the Treatment of Metastatic Malignant Melanoma

DOI: 10.1155/2012/259170

Full-Text   Cite this paper   Add to My Lib

Abstract:

Although advances in cytotoxic treatments have been obtained in several neoplasias, in metastatic melanoma there was no drug able to significantly change the natural history of the disease in the last 30 years. In the last decade, translational research identified important mechanisms in malignant transformation, invasion, and progression. Signaling pathways can be abnormally activated by oncogenes. The identification of oncogenic mutated kinases implicated in this process provides an opportunity for new target therapies. The melanoma dependence on BRAF-mutated kinase allowed the development of inhibitors that produced major responses in clinical trials. This is the beginning of a novel class of drugs in metastatic melanoma; the identification of the transduction signaling networking and other “druggable” kinases is in active research. In this paper, we discuss the ongoing research on cellular signaling inhibition, resistance mechanisms, and strategies to overcome treatment failure. 1. Introduction Malignant skin melanoma is one of the most chemoresistant and aggressive human neoplasias. In the last 30 years, no cytotoxic agent was able to importantly change the natural history of disease [1]. Several strategies to overcome resistance to cytotoxic agents have been tested, including combinations of drugs [2], cytokines, and vaccination [1]. With these therapeutic approaches, only a small fraction of the metastatic patients experienced tumour shrinkage, but such effects did not translate into significant clinical benefits in terms of progression-free survival or overall survival [1, 3]. Until recently, no predictive marker of response could be established. This scenario started to change in the last decade. With advances in translational research, it was possible to identify pathways and somatic mutations implicated in the biology of the melanoma. The identification and blockade of abnormal signaling through the mitogen-activated protein kinase (MAPK) pathway is the most promising therapeutic strategy to date. Around 60% of all melanomas express somatic mutations in the BRAF protein, and 90% of these express the oncogenic activating V600E mutation [4]. Vemurafenib, an inhibitor of the V600E BRAF kinase activity, has produced major responses [5] and showed an overall survival advantage as single agent against dacarbazine in a recent phase III trial [6]. Despite the advances, responses are transitory and we have not yet been able to neither cure nor stabilize the disease for long periods. Better understanding of tumoral biology and its adaptations to the

References

[1]  R. Mouawad, M. Sebert, J. Michels, J. Bloch, J. P. Spano, and D. Khayat, “Treatment for metastatic malignant melanoma: old drugs and new strategies,” Critical Reviews in Oncology/Hematology, vol. 74, no. 1, pp. 27–39, 2010.
[2]  G. J. Hill II, G. E. Metter, and E. T. Krementz, “DTIC and combination therapy for melanoma. II. Escalating schedules of DTIC with BCNU, CCNU, and vincristine,” Cancer Treatment Reports, vol. 63, no. 11-12, pp. 1989–1992, 1979.
[3]  E. Bajetta, M. del Vecchio, C. Bernard-Marty et al., “Metastatic melanoma: chemotherapy,” Seminars in Oncology, vol. 29, no. 5, pp. 427–445, 2002.
[4]  H. Davies, G. R. Bignell, C. Cox et al., “Mutations of the BRAF gene in human cancer,” Nature, vol. 417, no. 6892, pp. 949–954, 2002.
[5]  K. T. Flaherty, I. Puzanov, K. B. Kim et al., “Inhibition of mutated, activated BRAF in metastatic melanoma,” The New England Journal of Medicine, vol. 363, no. 9, pp. 809–819, 2010.
[6]  P. B. Chapman, A. Hauschild, C. Robert et al., “Improved survival with vemurafenib in melanoma with BRAF V600E mutation,” The New England Journal of Medicine, vol. 364, no. 26, pp. 2507–2516, 2011.
[7]  V. Gray-Schopfer, C. Wellbrock, and R. Marais, “Melanoma biology and new targeted therapy,” Nature, vol. 445, no. 7130, pp. 851–857, 2007.
[8]  J. Ackermann, M. Frutschi, K. Kaloulis, T. McKee, A. Trumpp, and F. Beermann, “Metastasizing melanoma formation caused by expression of activated N-RasQ61K on an INK4a-deficient background,” Cancer Research, vol. 65, no. 10, pp. 4005–4011, 2005.
[9]  L. Chin, A. Tam, J. Pomerantz et al., “Essential role for oncogenic Ras in tumour maintenance,” Nature, vol. 400, no. 6743, pp. 468–472, 1999.
[10]  M. Eskandarpour, F. Huang, K. A. Reeves, E. Clark, and J. Hansson, “Oncogenic NRAS has multiple effects on the malignant phenotype of human melanoma cells cultured in vitro,” International Journal of Cancer, vol. 124, no. 1, pp. 16–26, 2009.
[11]  S. R. D. Johnston, “Farnesyl transferase inhibitors: a novel targeted therapy for cancer,” The Lancet Oncology, vol. 2, no. 1, pp. 18–26, 2001.
[12]  S. Rao, D. Cunningham, A. de Gramont et al., “Phase III double-blind placebo-controlled study of farnesyl transferase inhibitor R115777 in patients with refractory advanced colorectal cancer,” Journal of Clinical Oncology, vol. 22, no. 19, pp. 3950–3957, 2004.
[13]  “E2902: a phase III randomized study of farnesyl transferase inhibitor R115777 in acute myeloid leukemia patients in second or subsequent remission or in remission after primary induction failure or patients over age 60 in first remission,” Clinical Advances in Hematology & Oncology, vol. 5, no. 1, pp. 13–14, 2007.
[14]  “Phase II study of the farnesyltransferase inhibitor R115777 in advanced melanoma: CALGB 500104,” ASCO, http://www.asco.org/ascov2/Meetings/Abstracts?&vmview=abst_detail_view&confID=40&abstractID=34612.
[15]  K. S. M. Smalley and T. G. Eisen, “Farnesyl transferase inhibitor SCH66336 is cytostatic, pro-apoptotic and enhances chemosensitivity to cisplatin in melanoma cells,” International Journal of Cancer, vol. 105, no. 2, pp. 165–175, 2003.
[16]  J. Dong, R. G. Phelps, R. Qiao et al., “BRAF oncogenic mutations correlate with progression rather than initiation of human melanoma,” Cancer Research, vol. 63, no. 14, pp. 3883–3885, 2003.
[17]  R. Kumar, S. Angelini, E. Snellman, and K. Hemminki, “BRAF mutations are common somatic events in melanocytic nevi,” Journal of Investigative Dermatology, vol. 122, no. 2, pp. 342–348, 2004.
[18]  E. E. Patton, H. R. Widlund, J. L. Kutok et al., “BRAF mutations are sufficient to promote nevi formation and cooperate with p53 in the genesis of melanoma,” Current Biology, vol. 15, no. 3, pp. 249–254, 2005.
[19]  G. Saldanha, D. Purnell, A. Fletcher, L. Potter, A. Gillies, and J. H. Pringle, “High BRAF mutation frequency does not characterize all melanocytic tumor types,” International Journal of Cancer, vol. 111, no. 5, pp. 705–710, 2004.
[20]  N. Dhomen, J. S. Reis-Filho, S. D. R. Dias et al., “Oncogenic braf induces melanocyte senescence and melanoma in mice,” Cancer Cell, vol. 15, no. 4, pp. 294–303, 2009.
[21]  D. Dankort, D. P. Curley, R. A. Cartlidge et al., “BrafV600E cooperates with Pten loss to induce metastatic melanoma,” Nature Genetics, vol. 41, no. 5, pp. 544–552, 2009.
[22]  Y. Chudnovsky, A. E. Adams, P. B. Robbins, Q. Lin, and P. A. Khavari, “Use of human tissue to assess the oncogenic activity of melanoma-associated mutations,” Nature Genetics, vol. 37, no. 7, pp. 745–749, 2005.
[23]  J. A. Curtin, J. Fridlyand, T. Kageshita et al., “Distinct sets of genetic alterations in melanoma,” The New England Journal of Medicine, vol. 353, no. 20, pp. 2135–2147, 2005.
[24]  S. M. Wilhelm, C. Carter, L. Tang et al., “BAY 43-9006 exhibits broad spectrum oral antitumor activity and targets the RAF/MEK/ERK pathway and receptor tyrosine kinases involved in tumor progression and angiogenesis,” Cancer Research, vol. 64, no. 19, pp. 7099–7109, 2004.
[25]  D. F. McDermott, J. A. Sosman, R. Gonzalez et al., “Double-blind randomized phase II study of the combination of sorafenib and dacarbazine in patients with advanced melanoma: a report from the 11715 study group,” Journal of Clinical Oncology, vol. 26, no. 13, pp. 2178–2185, 2008.
[26]  R. K. Amaravadi, L. M. Schuchter, D. F. McDermott et al., “Phase II trial of temozolomide and sorafenib in advanced melanoma patients with or without brain metastases,” Clinical Cancer Research, vol. 15, no. 24, pp. 7711–7718, 2009.
[27]  A. Hauschild, S. S. Agarwala, U. Trefzer et al., “Results of a phase III, randomized, placebo-controlled study of sorafenib in combination with carboplatin and paclitaxel as second-line treatment in patients with unresectable stage III or stage IV melanoma,” Journal of Clinical Oncology, vol. 27, no. 17, pp. 2823–2830, 2009.
[28]  B. Escudier, T. Eisen, W. M. Stadler et al., “Sorafenib for treatment of renal cell carcinoma: final efficacy and safety results of the phase III treatment approaches in renal cancer global evaluation trial,” Journal of Clinical Oncology, vol. 27, no. 20, pp. 3312–3318, 2009.
[29]  T. Eisen, T. Ahmad, K. T. Flaherty et al., “Sorafenib in advanced melanoma: a phase II randomised discontinuation trial analysis,” British Journal of Cancer, vol. 95, no. 5, pp. 581–586, 2006.
[30]  C. Robert, J. P. Arnault, and C. Mateus, “RAF inhibition and induction of cutaneous squamous cell carcinoma,” Current Opinion in Oncology, vol. 23, no. 2, pp. 177–182, 2011.
[31]  R. Kefford, H. Arkenau, M. P. Brown, et al., “Phase I/II study of GSK2118436, a selective inhibitor of oncogenic mutant BRAF kinase, in patients with metastatic melanoma and other solid tumors,” Journal of Clinical Oncology, vol. 28, no. 15, p. 8503, 2010.
[32]  “A Study of GSK2118436 in BRAF Mutatant Metastatic Melanoma to the Brain,” http://clinicaltrials.gov/ct2/show/NCT01266967?term=GSK2118436+melanoma rank=3.
[33]  “A Study Comparing GSK2118436 to Dacarbazine (DTIC) in Previously Untreated Subjects With BRAF Mutation Positive Advanced (Stage III) or Metastatic (Stage IV) Melanoma,” http://clinicaltrials.gov/ct2/show/NCT01227889?term=GSK2118436+melanoma&rank=1.
[34]  D. B. Solit, L. A. Garraway, C. A. Pratilas et al., “BRAF mutation predicts sensitivity to MEK inhibition,” Nature, vol. 439, no. 7074, pp. 358–362, 2006.
[35]  B. S. Jaiswal, V. Janakiraman, N. M. Kljavin et al., “Combined targeting of BRAF and CRAF or BRAF and PI3K effector pathways is required for efficacy in NRAS mutant tumors,” PLoS ONE, vol. 4, no. 5, Article ID e5717, 2009.
[36]  P. M. LoRusso, S. S. Krishnamurthi, J. J. Rinehart et al., “Phase I pharmacokinetic and pharmacodynamic study of the oral MAPK/ERK kinase inhibitor PD-0325901 in patients with advanced cancers,” Clinical Cancer Research, vol. 16, no. 6, pp. 1924–1937, 2010.
[37]  P. M. LoRusso, A. A. Adjei, M. Varterasian et al., “Phase I and pharmacodynamic study of the oral MEK inhibitor CI-1040 in patients with advanced malignancies,” Journal of Clinical Oncology, vol. 23, no. 23, pp. 5281–5293, 2005.
[38]  A. A. Adjei, R. B. Cohen, W. Franklin et al., “Phase I pharmacokinetic and pharmacodynamic study of the oral, small-molecule mitogen-activated protein kinase kinase 1/2 inhibitor AZD6244 (ARRY-142886) in patients with advanced cancers,” Journal of Clinical Oncology, vol. 26, no. 13, pp. 2139–2146, 2008.
[39]  “Search of: AZD6244 melanoma,” http://clinicaltrials.gov/ct2/results?term=AZD6244+melanoma.
[40]  “Investigate Safety, Pharmacokinetics and Pharmacodynamics of GSK2118436 & GSK1120212,” http://clinicaltrials.gov/ct2/show/NCT01072175?term=GSK1120212&rank=3.
[41]  “Study to Determine the Effectiveness of GSK1120212 in BRAF Mutation-positive Melanoma Previously Treated With or Without a BRAF Inhibitor,” http://clinicaltrials.gov/ct2/show/NCT01037127?term=GSK1120212+BRAF+melanoma&rank=1.
[42]  M. Ohori, M. Takeuchi, R. Maruki, H. Nakajima, and H. Miyake, “FR180204, a novel and selective inhibitor of extracellular signal-regulated kinase, ameliorates collagen-induced arthritis in mice,” Naunyn-Schmiedeberg's Archives of Pharmacology, vol. 374, no. 4, pp. 311–316, 2007.
[43]  M. Ohori, “ERK inhibitors as a potential new therapy for rheumatoid arthritis,” Drug News and Perspectives, vol. 21, no. 5, pp. 245–250, 2008.
[44]  M. Cully, H. You, A. J. Levine, and T. W. Mak, “Beyond PTEN mutations: the PI3K pathway as an integrator of multiple inputs during tumorigenesis,” Nature Reviews Cancer, vol. 6, no. 3, pp. 184–192, 2006.
[45]  L. C. Cantley, “The phosphoinositide 3-kinase pathway,” Science, vol. 296, no. 5573, pp. 1655–1657, 2002.
[46]  S. A. Aziz, M. Davies, E. Pick et al., “Phosphatidylinositol-3-kinase as a therapeutic target in melanoma,” Clinical Cancer Research, vol. 15, no. 9, pp. 3029–3036, 2009.
[47]  M. Cheung, A. Sharma, S. V. Madhunapantula, and G. P. Robertson, “Akt3 and mutant V600EB-Raf cooperate to promote early melanoma development,” Cancer Research, vol. 68, no. 9, pp. 3429–3439, 2008.
[48]  J. M. Ma?ecki, A. Bentke, B. Ostrowska, and P. Laidler, “Cytochalasin D, LY294002 and olomoucine synergize in promoting death of melanoma cells through activation of caspase-3 and apoptosis,” Melanoma Research, vol. 20, no. 1, pp. 52–58, 2010.
[49]  B. Bedogni, M. S. O'Neill, S. M. Welford et al., “Topical treatment with inhibitors of the phosphatidylinositol -kinase/Akt and Raf/mitogen-activated protein kinase kinase/extracellular signal-regulated kinase pathways reduces melanoma development in severe combined immunodeficient mice,” Cancer Research, vol. 64, no. 7, pp. 2552–2560, 2004.
[50]  F. Meier, S. Busch, K. Lasithiotakis et al., “Combined targeting of MAPK and AKT signalling pathways is a promising strategy for melanoma treatment,” British Journal of Dermatology, vol. 156, no. 6, pp. 1204–1213, 2007.
[51]  S. A. Aziz, L. B. Jilaveanu, C. Zito et al., “Vertical targeting of the phosphatidylinositol-3 kinase pathway as a strategy for treating melanoma,” Clinical Cancer Research, vol. 16, no. 24, pp. 6029–6039, 2010.
[52]  J. Werzowa, D. Cejka, T. Fuereder et al., “Suppression of mTOR complex 2-dependent AKT phosphorylation in melanoma cells by combined treatment with rapamycin and LY294002,” British Journal of Dermatology, vol. 160, no. 5, pp. 955–964, 2009.
[53]  T. Ran, T. Lu, H. Yuan et al., “A selectivity study on mTOR/PI3Kα inhibitors by homology modeling and 3D-QSAR,” Journal of Molecular Modeling. In press.
[54]  J. M. Stahl, A. Sharma, M. Cheung et al., “Deregulated Akt3 activity promotes development of malignant melanoma,” Cancer Research, vol. 64, no. 19, pp. 7002–7010, 2004.
[55]  J. Downward, “PI 3-kinase, Akt and cell survival,” Seminars in Cell and Developmental Biology, vol. 15, no. 2, pp. 177–182, 2004.
[56]  D. S. Ernst, E. Eisenhauer, N. Wainman et al., “Phase II study of perifosine in previously untreated patients with metastatic melanoma,” Investigational New Drugs, vol. 23, no. 6, pp. 569–575, 2005.
[57]  Y. Cheng, X. Ren, Y. Zhang et al., “eEF-2 kinase dictates cross-talk between autophagy and apoptosis induced by Akt inhibition, thereby modulating cytotoxicity of novel Akt inhibitor MK-2206,” Cancer Research, vol. 71, no. 7, pp. 2654–2663, 2011.
[58]  H. Hirai, H. Sootome, Y. Nakatsuru et al., “MK-2206, an allosteric akt inhibitor, enhances antitumor efficacy by standard chemotherapeutic agents or molecular targeted drugs in vitro and in vivo,” Molecular Cancer Therapeutics, vol. 9, no. 7, pp. 1956–1967, 2010.
[59]  S. K. Pal, K. Reckamp, H. Yu, and R. A. Figlin, “Akt inhibitors in clinical development for the treatment of cancer,” Expert Opinion on Investigational Drugs, vol. 19, no. 11, pp. 1355–1366, 2010.
[60]  “Search of: GSK2141795,” http://clinicaltrials.gov/ct2/results?term=GSK2141795.
[61]  “Search of: MK-2206,” http://clinicaltrials.gov/ct2/results?term=MK-2206.
[62]  “Search of: PBI-05204,” http://clinicaltrials.gov/ct2/results?term=PBI-05204.
[63]  “Search of: RX-0201,” http://clinicaltrials.gov/ct2/results?term=RX-0201.
[64]  M. P. Myers, I. Pass, I. H. Batty et al., “The lipid phosphatase activity of PTEN is critical for its tumor supressor function,” Proceedings of the National Academy of Sciences of the United States of America, vol. 95, no. 23, pp. 13513–13518, 1998.
[65]  T. Maehama and J. E. Dixon, “The tumor suppressor, PTEN/MMAC1, dephosphorylates the lipid second messenger, phosphatidylinositol 3,4,5-trisphosphate,” Journal of Biological Chemistry, vol. 273, no. 22, pp. 13375–13378, 1998.
[66]  J. M. Stahl, M. Cheung, A. Sharma, N. R. Trivedi, S. Shanmugam, and G. P. Robertson, “Loss of PTEN promotes tumor development in malignant melanoma,” Cancer Research, vol. 63, no. 11, pp. 2881–2890, 2003.
[67]  P. Guldberg, P. T. Straten, A. Birck, V. Ahrenkiel, A. F. Kirkin, and J. Zeuthen, “Disruption of the MMAC1/PTEN gene by deletion or mutation is a frequent event in malignant melanoma,” Cancer Research, vol. 57, no. 17, pp. 3660–3663, 1997.
[68]  G. P. Robertson, “Functional and therapeutic significance of Akt deregulation in malignant melanoma,” Cancer and Metastasis Reviews, vol. 24, no. 2, pp. 273–285, 2005.
[69]  H. Huang, J. C. Cheville, Y. Pan, P. C. Roche, L. J. Schmidt, and D. J. Tindall, “PTEN induces chemosensitivity in PTEN-mutated prostate cancer cells by suppression of Bcl-2 expression,” Journal of Biological Chemistry, vol. 276, no. 42, pp. 38830–38836, 2001.
[70]  R. Bianco, T. Troiani, G. Tortora, and F. Ciardiello, “Intrinsic and acquired resistance to EGFR inhibitors in human cancer therapy,” Endocrine-Related Cancer, vol. 12, no. 1, pp. S159–S171, 2005.
[71]  L. A. Julien and P. P. Roux, “mTOR, the mammalian target of rapamycin,” Medecine/Sciences, vol. 26, no. 12, pp. 1056–1060, 2010.
[72]  K. R. Molhoek, D. L. Brautigan, and C. L. Slingluff Jr., “Synergistic inhibition of human melanoma proliferation by combination treatment with B-Raf inhibitor BAY43-9006 and mTOR inhibitor rapamycin,” Journal of Translational Medicine, vol. 3, p. 39, 2005.
[73]  R. Nahta and R. O'Regan, “Evolving strategies for overcoming resistance to HER2-directed therapy: targeting the PI3K/Akt/mTOR pathway,” Clinical Breast Cancer, vol. 10, no. 3, pp. S72–S78, 2010.
[74]  K. Margolin, J. Longmate, T. Baratta et al., “CCI-779 in metastatic melanoma: a phase II trial of the California cancer consortium,” Cancer, vol. 104, no. 5, pp. 1045–1048, 2005.
[75]  Y. Wang, X. Y. Wang, J. R. Subjeck, P. A. Shrikant, and H. L. Kim, “Temsirolimus, an mTOR inhibitor, enhances anti-tumour effects of heat shock protein cancer vaccines,” British Journal of Cancer, vol. 104, no. 4, pp. 643–652, 2011.
[76]  K. R. Molhoek, D. L. Brautigan, and C. L. Slingluff Jr., “Synergistic inhibition of human melanoma proliferation by combination treatment with B-Raf inhibitor BAY43-9006 and mTOR inhibitor rapamycin,” Journal of Translational Medicine, vol. 3, p. 39, 2005.
[77]  “Search of: mtor melanoma,” http://clinicaltrials.gov/ct2/results?term=mtor+melanoma.
[78]  H. Yoshida, T. Kunisada, T. Grimm, E. K. Nishimura, E. Nishioka, and S. I. Nishikawa, “Review: melanocyte migration and survival controlled by SCF/c-kit expression,” Journal of Investigative Dermatology Symposium Proceedings, vol. 6, no. 1, pp. 1–5, 2001.
[79]  J. A. Curtin, K. Busam, D. Pinkel, and B. C. Bastian, “Somatic activation of KIT in distinct subtypes of melanoma,” Journal of Clinical Oncology, vol. 24, no. 26, pp. 4340–4346, 2006.
[80]  L. K. Ashman, “The biology of stem cell factor and its receptor C-kit,” International Journal of Biochemistry and Cell Biology, vol. 31, no. 10, pp. 1037–1051, 1999.
[81]  J. Coffey, R. Linger, J. Pugh et al., “Somatic KIT mutations occur predominantly in seminoma germ cell tumors and are not predictive of bilateral disease: report of 220 tumors and review of literature,” Genes Chromosomes and Cancer, vol. 47, no. 1, pp. 34–42, 2008.
[82]  A. P. D. Tos, L. Laurino, I. Bearzi, L. Messerini, and F. Farinati, “Gastrointestinal stromal tumors: the histology report,” Digestive and Liver Disease, vol. 43, supplement 4, pp. S304–S309, 2011.
[83]  N. Girard, “Thymic tumors: relevant molecular data in the clinic,” Journal of Thoracic Oncology, vol. 5, no. 10, pp. S291–S295, 2010.
[84]  C. A. Torres-Cabala, W. L. Wang, J. Trent et al., “Correlation between KIT expression and KIT mutation in melanoma: a study of 173 cases with emphasis on the acral-lentiginous/mucosal type,” Modern Pathology, vol. 22, no. 11, pp. 1446–1456, 2009.
[85]  D. Handolias, R. Salemi, W. Murray et al., “Mutations in KIT occur at low frequency in melanomas arising from anatomical sites associated with chronic and intermittent sun exposure,” Pigment Cell and Melanoma Research, vol. 23, no. 2, pp. 210–215, 2010.
[86]  K. S. Smalley, V. K. Sondak, and J. S. Weber, “c-KIT signaling as the driving oncogenic event in sub-groups of melanomas,” Histology and Histopathology, vol. 24, no. 5, pp. 643–650, 2009.
[87]  C. D. Mol, D. R. Dougan, T. R. Schneider et al., “Structural basis for the autoinhibition and STI-571 inhibition of c-Kit tyrosine kinase,” Journal of Biological Chemistry, vol. 279, no. 30, pp. 31655–31663, 2004.
[88]  I. Satzger, U. Küttler, B. V?lker, F. Schenck, A. Kapp, and R. Gutzmer, “Anal mucosal melanoma with KIT-activating mutation and response to imatinib therapy—case report and review of the literature,” Dermatology, vol. 220, no. 1, pp. 77–81, 2010.
[89]  D. Handolias, A. L. Hamilton, R. Salemi et al., “Clinical responses observed with imatinib or sorafenib in melanoma patients expressing mutations in KIT,” British Journal of Cancer, vol. 102, no. 8, pp. 1219–1223, 2010.
[90]  F. S. Hodi, P. Friedlander, C. L. Corless et al., “Major response to imatinib mesylate in KIT-mutated melanoma,” Journal of Clinical Oncology, vol. 26, no. 12, pp. 2046–2051, 2008.
[91]  J. Lutzky, J. Bauer, and B. C. Bastian, “Dose-dependent, complete response to imatinib of a metastatic mucosal melanoma with a K642E KIT mutation,” Pigment Cell and Melanoma Research, vol. 21, no. 4, pp. 492–493, 2008.
[92]  A. Ashida, M. Takata, H. Murata, K. Kido, and T. Saida, “Pathological activation of KIT In metastatic tumors of acral and mucosal melanomas,” International Journal of Cancer, vol. 124, no. 4, pp. 862–868, 2009.
[93]  S. Ugurel, R. Hildenbrand, A. Zimpfer et al., “Lack of clinical efficacy of imatinib in metastatic melanoma,” British Journal of Cancer, vol. 92, no. 8, pp. 1398–1405, 2005.
[94]  K. Wyman, M. B. Atkins, V. Prieto et al., “Multicenter phase II trial of high-dose imatinib mesylate in metastatic melanoma: significant toxicity with no clinical efficacy,” Cancer, vol. 106, no. 9, pp. 2005–2011, 2006.
[95]  “Phase II trial of imatinib mesylate (STI-571) in metastatic melanoma (MM),” ASCO, http://www.asco.org/ascov2/Meetings/Abstracts?&vmview=abst_detail_view&confID=26&abstractID=2901.
[96]  “Search of: kit melanoma,” http://clinicaltrials.gov/ct2/results?term=kit+melanoma.
[97]  C. M. Johannessen, J. S. Boehm, S. Y. Kim et al., “COT drives resistance to RAF inhibition through MAP kinase pathway reactivation,” Nature, vol. 468, no. 7326, pp. 968–972, 2010.
[98]  N. Wagle, C. Emery, M. F. Berger et al., “Dissecting therapeutic resistance to RAF inhibition in melanoma by tumor genomic profiling,” Journal of Clinical Oncology, vol. 29, no. 22, pp. 3085–3096, 2011.
[99]  R. Nazarian, H. Shi, Q. Wang et al., “Melanomas acquire resistance to B-RAF(V600E) inhibition by RTK or N-RAS upregulation,” Nature, vol. 468, no. 7326, pp. 973–977, 2010.
[100]  G. Hatzivassiliou, K. Song, I. Yen et al., “RAF inhibitors prime wild-type RAF to activate the MAPK pathway and enhance growth,” Nature, vol. 464, no. 7287, pp. 431–435, 2010.
[101]  P. I. Poulikakos, C. Zhang, G. Bollag, K. M. Shokat, and N. Rosen, “RAF inhibitors transactivate RAF dimers and ERK signalling in cells with wild-type BRAF,” Nature, vol. 464, no. 7287, pp. 427–430, 2010.
[102]  D. B. Solit and N. Rosen, “Resistance to BRAF inhibition in melanomas,” The New England Journal of Medicine, vol. 364, no. 8, pp. 772–774, 2011.
[103]  S. Whittaker, R. Kirk, R. Hayward et al., “Gatekeeper mutations mediate resistance to BRAF-targeted therapies,” Science Translational Medicine, vol. 2, no. 35, Article ID 35ra41, 2010.
[104]  F. S. Hodi, S. J. O'Day, D. F. McDermott et al., “Improved survival with ipilimumab in patients with metastatic melanoma,” The New England Journal of Medicine, vol. 363, no. 8, pp. 711–723, 2010.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133

WeChat 1538708413