108
Views
9
CrossRef citations to date
0
Altmetric
Review

Molecular targeted therapies in metastatic melanoma

, &
Pages 49-56 | Published online: 07 Jun 2013

Abstract

The advent of personalized medicine has ushered in a new era for cancer therapy with a significant impact on the management of advanced melanoma. Molecular targeted therapies have shown promise in the management of various malignancies, including melanoma, with lower toxicity profiles and better overall survival as compared with conventional therapy. The discovery of BRAF mutations in melanoma led to the development of BRAF inhibitors for the treatment of advanced melanoma. However, growing concerns over drug resistance to molecular targeted therapies including BRAF inhibitors, have spurred efforts to elucidate additional molecular targets for the treatment of advanced melanoma. In this review, we discuss the known molecular aberrations in melanoma, current and novel targeted approaches in its treatment, and drug resistance patterns.

Introduction

Malignant melanoma is the fifth and sixth most common new skin cancer diagnosis in men and women, respectively, in the United States. Among the skin cancers, melanoma has the greatest metastatic potential, with metastatic disease occurring in 10%–15% of patients at diagnosis.Citation1,Citation2 Metastatic melanoma has a dismal prognosis, with a five-year overall survival of 15%. Over the past 40 years, limited progress has been made in the treatment of metastatic melanoma through the use of chemotherapy, immunotherapy, biochemotherapy, and combinations thereof.Citation3,Citation4

Conventional chemotherapy with dacarbazine and temozolomide has yielded poor response rates of 7%–20% and a median survival of nine months, with mild toxicity profiles.Citation5,Citation6 Immunotherapies such as interleukin-2, while achieving durable responses (response rate 16%, median duration of response 8.9 months) in metastatic melanoma, are associated with significant toxicityCitation3 and offer limited options for effective and safe therapies for management of metastatic melanoma.Citation7,Citation8

Two new immunotherapeutic agents, ie, ipilimumab (recombinant, fully human IgG1 monoclonal antibody against cytotoxic T lymphocyte-associated antigen 4 [CTLA-4]) and anti-programmed cell death 1 [PD-1], show promise as potentially effective therapies with manageable side effect profiles in metastatic melanoma. Ipilimumab has an overall response rate of 10.9%, and in those patients who respond, over half have a durable response.Citation9,Citation10 The major limitations are that at this time there is no way to predict these responders, and side effects include numerous immune-mediated toxicities. A T cell regulator that functions similarly to CTLA-4 is PD-1. The PD-1 ligand allows tumors to evade the host immune response. PD-1 ligand antibodies have been shown to enhance tumor immune response in patients with melanoma.Citation11 Other promising therapies include several angiogenesis-promoting molecules, such as vascular endothelial growth factor.Citation12 In spite of recent advances in immune-based therapy, and given the absence of long-term remissions in the majority of treated patients, new treatments for metastatic melanoma are needed.

Recent advances in molecular biology and genomics have uncovered the molecular heterogeneity of tumors and facilitated a shift in anticancer therapy strategies from the traditional “one-size-fits-all” approach to an individualized approach to therapy.Citation13,Citation14 Key molecular drivers of tumor oncogenesis and mechanisms of tumor resistance have been uncovered, revealing the limitations of reliance solely on the clinical and pathological classification of tumors. This knowledge has resulted in the development of new treatment strategies that rely on therapy targeted towards identified functional genetic mutations, resulting in improved tumor response rates and relatively tolerable side effect profiles.Citation15

The discovery of activating mutations in serine/threonine kinase, BRAF (v-raf murine sarcoma viral oncogene homolog B1) in 50%–60% of melanomas (superficial spreading type) in 2002 spurred investigations into the development of targeted therapies. This ultimately resulted in the approval of vemurafenib, a BRAF inhibitor, by the US Food and Drug Administration in August 2011 for the treatment of locally advanced/unresectable or metastatic BRAF-mutated malignant melanoma.Citation16,Citation17 The purpose of this review is to discuss the conventional and novel molecular targeted treatment approaches for the management of advanced melanoma and show the major drug resistance patterns associated with BRAF inhibitor therapies.

Molecular pathogenesis of melanoma and implications for targeted therapy

Melanoma is a heterogeneous disease reflected by its complex pathobiology. Recent advances in molecular genomic techniques have enabled the elucidation of functionally relevant cellular processes implicated in the oncogenesis of melanoma. Dysregulation of the cell growth cycle and signaling represent key mechanisms for tumor growth and persistence in melanoma and are the predominant molecular events in the majority of cases.

Cell cycle changes

Cell cycle dysregulation in melanoma represents one of the most important pathogenetic mechanisms for its oncogenesis, resulting in uncontrolled cellular proliferation. The most prominent molecular target is the CDKN2A locus (chromosome 9p21) that acts as a tumor suppressor in melanoma. Germline and somatic mutations in CDKN2A account for 10%–40% of familial melanoma,Citation18 and 10% of all melanomas are familial in origin.Citation19 The absolute risk for melanoma in individuals with the CDKN2A mutation is modulated by identifiable heritable traits (skin, hair, and eye color, large numbers of benign and atypical nevi, giant congenital nevi or a family history of melanoma) and environmental factors (history of sunlight exposure).Citation20,Citation21Citation24 In familial cases, the risk for development of melanoma by the age of 50 years is 50% in the United States, and 76% by the age of 80 years.Citation25 In sporadic CDKN2A mutation carriers, the risk of melanoma is much lower, at 14%, 24%, and 28% by the ages of 50, 70, and 80 years.Citation26

CDKN2A encodes two distinct proteins, p16INK4A and p14ARF, which both act as tumor suppressors by inhibiting progression of the cell cycle through negative regulation of the RB1 and p53 pathways, respectively. Therefore, genetic aberrations that lead to functional loss of either of these proteins (p16INK4A and p14ARF) will ultimately result in uncontrolled cellular proliferation. While initial studies of first-generation CDK inhibitors, such as flavopiridol, failed to demonstrate efficacy in preclinical studies, second-generation CDK inhibitors (SCH 727965), have shown more promising results in halting melanoma progression in mouse xenografts. This effect is potentiated when CDK inhibitors are combined with paclitaxel.Citation27,Citation28

RB1 is the central piece of the pathway controlled by p16INK4A, serving in its unphosphorylated form to sequester E2F transcription factor, preventing it from inducing gene expression critical for transition from the G1 to the S phase of the cell cycle. RB1 phosphorylation leads to release of E2F, enabling it to induce expression of the target genes necessary for progression from the G1 to S phase of the cell cycle. Phosphorylation of RB1 is performed by a catalytic complex composed of cyclin D1 and CDK 4 or 6. The activity of this catalytic complex is dependent on levels of p16INK4A. High levels of p16INK4A lead to suppression of the activity of the cyclin D1-CDK4/6 complex, with resulting suppression of RB1 phosphorylation and suppression of release of EF2 sequestration and ultimately cell cycle arrest at the G1/S step. However, when levels of p16INK4A are low, inhibition of the catalytic complex is released, resulting in progression of the cell cycle. Genetic mutations that affect the CDKN2A locus occur as deletions of p16INK4A in 50% of melanomas and as inactivating point mutations in 9% of melanomas.Citation29 This suggests that p16INK4A serves a critical role in cell cycle regulation in melanocytes. Less common mutations include amplifications and point mutations of the CDK4 gene, resulting in constitutive activation of the CDK4/cyclin D1 complex, overexpression of CDK6, and inactivating mutations in the RB1 gene (6% of melanomas).Citation29

TP53 is the most common gene mutation in human cancer and p53 transcription factor controls various genes responsible for cell cycle arrest, senescence, DNA repair, and cell death. However, TP53 mutations represent a low frequency event in melanoma, occurring in only 9% of melanomas.Citation30 MDM2 is one of the negative regulators of p53 and is regulated by p14ARF (one of the protein products of CDKN2A) which, when bound to MDM2, inhibits its function and stabilizes p53.Citation20 Mutations in p14ARF result in restoration of MDM2 activity and ubiquitination of p53 by MDM2, with resulting degradation and destabilization of p53.Citation31 This loss of p53 through mutations in p14ARF and active MDM2 eventually leads to cell cycle progression. Therefore, the MDM2-p53 interaction is a possible treatment target for tumors because blocking MDM2 stabilizes and reactivates p53, allowing for tumor suppression.Citation32 MDM2 antagonists are just beginning clinical trials.

Cell signaling changes

Dysregulation of the signal transduction pathway for mitogen-activated protein kinase (MAPK), also known as RAS/RAF/MEK, plays a key role in multiple human cancers, including melanoma. Activation of MAPK signaling by mutations is implicated in 90% of melanomas.Citation33 Signaling through the MAPK pathway occurs through extracellular signals that lead to the binding of a broad array of receptor tyrosine kinases, which results in activation of Ras, a small G-protein with three isoforms, HRAS, KRAS, and NRAS, with resultant downstream effects of cellular proliferation and survival. Al-Mulla et al demonstrated the variable effects of Ras mutations on the invasiveness of tumors in vitro, with implications for the biologic behavior of these tumors in vivo.Citation34 The receptor tyrosine kinases include growth factor receptors, such as epidermal growth factor receptor, c-KIT, platelet-derived growth factor receptor, vascular endothelial growth factor receptor, and fibroblast growth factor receptor.Citation35 Binding of growth factors to receptor tyrosine kinases leads to activation of Ras, resulting in formation of a complex between Ras and one of the RAF serine/threonine kinase isoforms, ie, ARAF, BRAF, or RAF1 (CRAF). Formation of the Ras-RAF complex leads to activation of RAF and phosphorylation and activation of MEK that, in turn, activates MAPK isoforms (also known as ERK), including MAPK3 and MAPK1. MAPK3 and MAPK1 activation results in an array of downstream effects, including increased proliferation, protection from apoptosis, and increased survival through induction of transcription factors and cell cycle proteins in the nucleus.Citation20,Citation35 Ras activation ultimately results in stimulation of multiple intracellular signaling pathways, including the MAPK pathway, Ral guanine nucleotide exchange factors, and the phosphoinositide 3-kinase (P13K/AKT) pathway.Citation36

The predominant mutations in the MAPK pathway leading to its constitutive activation are mutations in BRAF. Of all malignancies, activating mutations in BRAF are present at the highest frequency (27%–68%) in melanomas.Citation16,Citation37 The mutation that accounts for the majority (60%–100%) of all BRAF mutations in melanoma involves substitution of a glutamate for valine at position 600 (V600E).Citation38,Citation39 This results in downstream activation of MAPK and ultimately proliferation and survival of melanoma. Hence BRAF V600E represents an attractive molecular target for treatment of melanoma. The BRAF V600E mutation has also been described in benign melanocytic proliferations, suggesting that this mutational event alone is not sufficient for tumorigenesis and that additional genetic insults are required for transformation to melanoma.Citation20 Intermittently sun-exposed skin, as well as acral and mucosal melanomas, commonly bear BRAF mutations.Citation40 MAPK signaling in melanocytes via growth factor-mediated activation of adenylate cyclase primarily occurs through BRAF and likely explains the high frequency of BRAF mutations seen in melanoma. An increase in adenylate cyclase activity results in accumulation of cyclic AMP and activation of protein kinase. Activated protein kinase A inhibits CRAF, enabling signaling to proceed via BRAF.Citation41

Mutations in the Ras proteins, NRAS, HRAS, and KRAS, are less common events in melanoma, accounting for 20%, 2%, and 1% of all melanomas, respectively. These Ras mutations appear to represent early events in the oncogenesis of melanoma, with additional mutational events necessary to initiate oncogenic transformation.Citation42 The most common (>80%) mutation in NRAS is a point mutation that results in substitution of glutamine for leucine at position 61.Citation43 This point mutation leads to dysfunctional GTPase activity that maintains the Ras protein in an activated (GTP-bound) state. NRAS-mutated melanomas appear to have distinctive clinical, histopathologic, and prognostic features. The typical clinical presentation is in older individuals (>55 years of age) on chronically photoexposed skin of the extremities. Histopathologic features include thicker tumors without ulceration and higher mitotic rates compared with BRAF-mutant melanomas. Mutant NRAS melanoma has a worse overall survival than wild-type NRAS melanoma.Citation44Citation46 Mutant NRAS melanoma is dependent on CRAF and not BRAF signaling for growth factor-mediated MAPK signaling. This pathway relies on two parallel mechanisms, including Ras isoform switching, that permits inactivation of BRAF by causing its phosphorylation thereby preventing Ras/BRAF association, and increased expression of the cyclic AMP degrading enzyme, phosphodiesterase IV, which restricts protein kinase A activity and ultimately prevents phosphorylation of CRAF at its inhibitory sites, promoting CRAF-mediated MAPK signaling.Citation41 The latter mechanism presents an opportunity for therapeutic targeting of phosphodiesterase IV through its inhibition in melanomas with Ras mutations that are resistant to BRAF V600E inhibitors.Citation47 While phosphodiesterase IV antibodies have demonstrated therapeutic potential for the management of chronic obstructive pulmonary disease and asthma,Citation48,Citation49 there are no definitive studies evaluating their clinical efficacy in mutant NRAS or wild-type BRAF melanoma. However, preliminary findings have demonstrated inhibition of growth potential and increased apoptosis of mutant NRAS melanoma cell lines.Citation47

BRAF inhibitors: mechanisms of action and drug resistance patterns

Recognition of multiple mutations in melanoma within components of the MAPK signaling pathway has led to interest in targeted therapies, especially given the lack of evidence for improved overall survival rates with conventional therapies, such as interleukin-2 and chemotherapy.Citation50 While mutations in both NRAS and BRAF have been identified in melanoma,Citation16,Citation51 therapies targeting the MAPK pathway have focused on inhibition of BRAF and MEK. Initial attempts to inhibit BRAF in melanoma used sorafenib, a tyrosine kinase inhibitor that inhibits multiple tyrosine kinases, including BRAF. However, sorafenib does not block the V600E mutation, and therapy alone or in combination with chemotherapy did not demonstrate benefit.Citation52Citation54 More selective BRAF inhibitors were developed, and in 2011 the US Food and Drug Administration approved the selective inhibitor, vemurafenib, for patients with malignant melanoma bearing the activating BRAF (V600E) mutation. In a Phase I trial, 81% of patients with V600E-positive metastatic melanoma responded to treatment. Overall, 26 of 32 patients showed a partial response (defined as a decrease by at least 30% in the sum of the largest diameter of each target lesion), including two with complete resolution.Citation55 A randomized controlled Phase III trial compared vemurafenib with a commonly used standard chemotherapy agent, dacarbazine, in 675 patients with untreated V600E-positive metastatic melanoma. At six months, overall survival was 84% in the vemurafenib group and 64% in the dacarbazine group.Citation17 After the interim analysis, crossover to vemurafenib from dacarbazine was recommended, and an updated analysis continues to show improvement in overall survival and progression-free survival.Citation17 While the initial response can be dramatic, progression-free survival has ranged from 5–7 months,Citation17,Citation57 leading to concern about drug resistance. Indeed, secondary mutations in addition to BRAF have been observed with progressive disease.Citation58,Citation59

Dabrafenib is another selective BRAF inhibitor that has shown significant activity in patients with metastatic melanoma in Phase I/II studies.Citation60 Further clinical trials are underway with dabrafenib. It is important to note that the BRAF inhibitors, vemurafenib and dabrafenib, are the first treatments to show benefit in patients with metastasis to the brain.Citation61,Citation62 In addition, while vemurafenib has been studied in patients with the V600E mutation, studies with dabrafenib are examining activity in non-V600E and V600K mutations.Citation56,Citation62

Importantly, vemurafenib and dabrafenib have been well tolerated overall, with the most common side effects being cutaneous. Patients can experience fatigue and arthralgias with both agents, but unlike vemurafenib, approximately 10% of patients on dabrafenib also develop pyrexia.Citation63 Patients can experience significant photosensitivity and rash that can require dose reduction, and epithelial neoplasms ranging from benign keratosis to keratoacanthomas and squamous cell carcinomas are common, and can affect up to 20%–30% of patients on vemurafenib.Citation17,Citation64 The mechanism underlying increased rates of malignant squamous proliferations with BRAF inhibitors is thought to involve disruption of the MAPK pathway, where inhibition of RAF activity leads to increased activity in RAS. RAS mutations are common in cutaneous squamous cell carcinomas and keratoacanthomas, and the paradoxical activation of RAS seen with BRAF inhibition accelerates the oncogenic process.Citation65 There is also concern that BRAF inhibition may induce carcinogenesis in other melanocytic proliferations or other organs through a similar mechanism. For example, new wild-type BRAF primary melanomas have been found to arise in patients subsequent to treatment with BRAF inhibitors.Citation66

Understanding drug resistance for selective BRAF inhibitors remains a major concern and area of interest. Although the pathway that leads to squamous cell carcinoma with BRAF inhibitors seems to have been elucidated, studies suggest resistance is due to more complex compensatory activation of numerous components of the MAPK pathway. There can be upregulation of receptor tyrosine kinases, such as platelet-derived growth factor receptor-β and insulin-like growth factors, secondary NRAS mutations, and activation of MEK.Citation58,Citation59 MEK activation can occur through various mutations in MEK1.Citation67 COT kinase is an activator of the MAPK pathway that does not require RAF signaling, and upregulation of COT kinase has been shown to promote resistance to BRAF inhibitors.Citation68 In addition, increased phosophorylated ERK1/2 levels have been observed in melanomas with acquired resistance to vemurafenib.Citation67 Finally, BRAF V600E splice variants, reported in colon and thyroid cancer, lead to reactivation of feedback in the MAPK pathway via MEK and the receptor tyrosine kinase, epidermal growth factor receptor.Citation69Citation71 The possibilities for potential combinations allowing for drug resistance are clearly numerous and complex. These resistance patterns support consideration of combination therapies, including BRAF inhibitors and drugs that target other members of the MAPK pathway.

Future directions in molecular-targeted therapy for melanoma

With limited disease-free survival rates and drug resistance following treatment with BRAF inhibitors, additional treatment options are needed. Efforts are underway to find other targeted therapies within the MAPK pathway that could be used alone or in combination with BRAF inhibitors. There has been significant investigation into MEK inhibition. Phase III trials comparing trametinib, a MEK inhibitor, with chemotherapy in patients with BRAF V600E/K mutant malignant melanoma show improved overall survival and progression-free survival.Citation72 The combination of dabrafenib and trametinib has shown improved progression-free survival as well as reduction in the rate of secondary cutaneous neoplasms (such as squamous cell carcinoma).Citation73 Based on the numerous pathways for resistance, MEK inhibition alone is unlikely to be the only answer to BRAF resistance. Nonetheless, MEK inhibitors have shown promise.

Another option being explored for targeted therapy in melanoma is the receptor tyrosine kinase, c-KIT (or CD117). Activating c-KIT mutations have been reported in approximately 20%–30% of certain subtypes of melanoma, including acral melanomas and mucosal melanomas, and melanomas that develop on photodamaged skin.Citation74 The most common point mutation is L576P in exon 11, but point mutations also occur in exons 13, 17, and 18.Citation75 Other tumors, including gastrointestinal stromal tumors with c-KIT mutations have been responsive to the tyrosine kinase inhibitor, imatinib.Citation76 Therefore, Phase II trials were conducted with imatinib in patients with acral or mucosal melanoma or melanomas on chronically sun-damaged skin that harbored KIT mutations or amplifications. Response rates of 16%–23% with a small number of complete long-term responses have been seen, with no difference in response rates between the various melanoma subtypes.Citation77Citation79 Notably, the same KIT mutations (K642E and N822K) that have shown response to treatment in gastrointestinal stromal tumors also show response in the treatment of melanoma. Meanwhile, resistance to specific KIT mutations (V654A and D820Y) are observed in both gastrointestinal stromal tumors and melanoma.Citation77 There are numerous tyrosine kinase inhibitors, and trials involving other KIT-directed therapies are underway in patients with melanomas harboring c-KIT mutations.

Attempts have been made to target Ras indirectly by blocking important post-translation modification. Farnesyl transferase inhibitors, such as lonafarnib, block Ras by inhibiting its farnesylation and blocking translocation of Ras to the plasma membrane. Lonafarnib, in combination with other chemotherapeutic agents such as sorafenib and cisplatin, has demonstrated encouraging results in metastatic melanoma in vitro.Citation80,Citation81 A single clinical trial attempted to treat melanoma by inhibiting Ras via farnesyl transferase suppression, and showed significant toxicity and a lack of efficacy.Citation82 However, it should be noted that the patients in this study were not selected based on the presence of NRAS mutation.

Conclusion

The advent of oncologic molecular typing has galvanized the discipline of personalized medicine. Recognition of common mutations within particular tumors has shown and holds tremendous promise for targeted individualized therapies. In melanoma, studies show an early favorable response to BRAF V600E inhibitors in treating BRAF V600E mutant melanomas. However, many challenges remain. The first and foremost is the other half of metastatic melanomas which do not harbor BRAF mutations. Another challenge is that although overall survival increases for those using BRAF inhibitors, complete resolution only occurs in a minority and most cases relapse through secondary resistance. Other targeted therapies, such as tyrosine kinase inhibitors, are being explored in melanomas with activating c-KIT mutations, and therapies such as MEK inhibitors are being developed to exploit regulation of the MAPK pathway. Immunomodulation has shown potential, but the ability to provide targeted immunomodulating therapy has yet to be achieved. The limitations of BRAF inhibitors and other targeted therapies reinforce the complexity of these tumors and the host response. Combinations of existing and new therapeutic options will need to be explored. However, advances made over the last few years have generated new hope for effective treatments.

Disclosure

The authors report no conflicts of interest in this work.

References

  • National Cancer InstituteA snapshot of melanoma2012 Available from: http://www.cancer.gov/researchandfunding/snapshots/pdf/Melanoma-Snapshots.pdf. Accessed January 16, 2013.
  • National Cancer InstituteSEER Stats Facts sheets. Melanoma of the skin Available from: http://seer.cancer.gov/statfacts/html/melan.html. Accessed April 16, 2013
  • EggermontAMSchadendorfDMelanoma and immunotherapyHematol Oncol Clin North Am20092354756419464602
  • EggermontAMKirkwoodJMRe-evaluating the role of dacarbazine in metastatic melanoma: what have we learned in 30 years?Eur J Cancer2004401825183615288283
  • ChapmanPBEinhornLHMeyersMLPhase III multicenter randomized trial of the Dartmouth regimen versus dacarbazine in patients with metastatic melanomaJ Clin Oncol1999172745275110561349
  • MiddletonMRGrobbJJAaronsonNRandomized phase III study of temozolomide versus dacarbazine in the treatment of patients with advanced metastatic malignant melanomaJ Clin Oncol20001815816610623706
  • AtkinsMBLotzeMTDutcherJPHigh-dose recombinant interleukin-2 therapy for patients with metastatic melanoma: analysis of 270 patients treated between 1985 and 1993J Clin Oncol1999172105211610561265
  • TsaoHAtkinsMBSoberAJManagement of cutaneous melanomaN Engl J Med2004351998101215342808
  • HodiFSO’DaySJMacDermottDFImproved survival with ipilimumab in patients with metastatic melanomaN Engl J Med201036371172320525992
  • RobertCThomasLBondarenkoIIpilimumab plus dacarbazine for previously untreated metastatic melanomaN Engl J Med20113642517252621639810
  • BrahmerJRTykodiSSChowLQSafety and activity of anti-PD-L1 antibody in patients with advanced cancerN Engl J Med20123662455246522658128
  • DeanELoriganPAdvances in the management of melanoma: targeted therapy, immunotherapy and future directionsExpert Rev Anticancer Ther2012121437144823249108
  • MidorikawaYTsujiSTakayamaTAburataniHGenomic approach towards personalized anti-cancer drug therapyPharmacogenomics20121319119922256868
  • AwadaAVandoneAMAftimosPPersonalized management of patients with solid cancers: moving from patient characteristics to tumor biologyCurr Opin Oncol20122429730422410457
  • FinnLMarkovicSNJosephRWTherapy for metastatic melanoma: the past, the present and futureBMC Med2012102322385436
  • DaviesHBignellGRCoxCMutations of the BRAF gene in human cancerNature200241794995412068308
  • ChapmanPBHauschildARobertCBRIM-3 Study GroupImproved survival with vemurafenib in melanoma with BRAF V600E mutationN Engl J Med20113642507251621639808
  • GoldsteinAMChanMHarlandMLund Melanoma Study Group, Melanoma Genetics Consortium (GenoMEL)Features associated with germline CDKN2A mutations: a GenoMEL study of melanoma-prone families from three continentsJ Med Genet2007449910616905682
  • MeyerLJZoneJHGenetics of cutaneous melanomaJ Invest Dermatol1994103Suppl 5112S116S7963671
  • SekulicAHaluskaPJrMillerAJMelanoma Study Group of Mayo Clinic Cancer CenterMalignant melanoma in the 21st century: the emerging molecular landscapeMayo Clin Proc20088382584618613999
  • GilchrestBAEllerMSGellerACThe pathogenesis of melanoma induced by ultraviolet radiationN Engl J Med19993401341134810219070
  • GandiniSSeraFCattaruzzaMSMeta-analysis of risk factors for cutaneous melanoma: I. Common and atypical neviEur J Cancer200541284415617989
  • TannousZSMihmMCJrSoberAJDuncanLMCongenital melanocytic nevi: clinical and histopathologic features, risk of melanoma, and clinical managementJ Am Acad Dermatol20055219720315692463
  • WattAJKotsisSVChungKCRisk of melanoma arising in large congenital melanocytic nevi: a systematic reviewPlast Reconstr Surg20041131968197415253185
  • BishopDTDemenaisFGoldsteinAMMelanoma Genetics Consortium. Geographical variation in the penetrance of CDKN2A mutations for melanomaJ Natl Cancer Inst20029489490312072543
  • BeggCBOrlowIHummerAJGenes Environment and Melanoma Study GroupLifetime risk of melanoma in CDKN2A mutation carriers in a population-based sampleJ Natl Cancer Inst2005971507151516234564
  • AbdullahCWangXBeckerDExpression analysis and molecular targeting of cyclin-dependent kinases in advanced melanomaCell Cycle20111097798821358262
  • MannMBKaldisPCell cycle transitions and Cdk inhibition in melanoma therapy: cyclin’ through the optionsCell Cycle201110134921464616
  • BennettDCHow to make a melanoma: what do we know of the primary clonal events?Pigment Cell Melanoma Res200821273818353141
  • PetitjeanAMatheEKatoSImpact of mutant p53functional properties on TP53 mutation patterns and tumor phenotype: lessons from recent developments in the IARC TP53 databaseHum Mutat20072862262917311302
  • KamijoTWeberJDZambettiGZindyFRousselMFSherrCJFunctional and physical interactions of the ARF tumor suppressor with p53 and Mdm2Proc Natl Acad Sci U S A199895829282979653180
  • ShangarySWangSTargeting the MDM-p53 interaction for cancer therapyClin Cancer Res2008145318532418765522
  • NikolaouVAStratigosAJFlahertyKTTsaoHMelanoma: new insights and new therapiesJ Invest Dermatol201213285486322217739
  • Al-MullaFMacKenzieEMDifferences in in vitro invasive capacity induced by differences in Ki-Ras protein mutationsJ Pathol200119554955611745690
  • FecherLAAmaravadiRKFlahertyKTThe MAPK pathway in melanomaCurr Opin Oncol20082018318918300768
  • DownwardJTargeting RAS signaling pathways in cancer therapyNat Rev Cancer20033112212509763
  • HoubenRBeckerJCKappelAConstitutive activation of the Ras-Raf signaling pathway in metastatic melanoma is associated with poor prognosisJ Carcinog20043615046639
  • PlatzAEgyhaziSRingborgUHanssonJHuman cutaneous melanoma: a review of NRAS and BRAF mutation frequencies in relation to histogenetic subclass and body siteMol Oncol2008139540519383313
  • SclafaniFGulloGSheahanKBRAF mutations in melanoma and colorectal cancer: A single oncogenic mutation with different tumor phenotypes and clinical implicationsCrit Rev Oncol Hematol12112012 [Epub ahead of print.]
  • LongGVMenziesAMNagrialAMPrognostic and clinicopathologic associations of oncogenic BRAF in metastatic melanomaJ Clin Oncol2011291239124621343559
  • DumazNHaywardRMartinJIn melanoma, RAS mutations are accompanied by switching signaling from BRAF to CRAF and disrupted cyclic AMP signalingCancer Res2006669483949117018604
  • MooiWJPeeperDSOncogene-induced cell senescence – halting on the road to cancerN Engl J Med20063551037104616957149
  • FedorenkoIVGibneyGTSmalleyKSNRAS mutant melanoma: biological behavior and future strategies for therapeutic managementOncogene10152012 [Epub ahead of print.]
  • DevittBLiuWSalemiRClinical outcome and pathological features associated with NRAS mutation in cutaneous melanomaPigment Cell Melanoma Res20112466667221615881
  • JakobJABassettRLJrNgCSNRAS mutation status is an independent prognostic factor in metastatic melanomaCancer20121184014402322180178
  • EllerhorstJAGreeneVREkmekciogluSClinical correlates of NRAS and BRAF mutations in primary human melanomaClin Cancer Res20111722923520975100
  • MarquetteAAndréJBagotMBensussanADumazNERK and PDE4 cooperate to induce RAF isoform switching in melanomaNat Struct Mol Biol20111858459121478863
  • O’ByrnePMGauvreauGPhosphodiesterase-4-inhibition in COPDLancet200937466566719716944
  • NgkeloAAdcockIMNew treatments for COPDCurr Opin Pharmacol4182013 [Epub ahead of print.]
  • CrosbyTFishRColesBMasonMDSystemic treatments for metastatic cutaneous melanomaCochrane Database Syst Rev20002CD00121510796759
  • DemunterAStasMDegreefHDe Wolf-PeetersCvan den OordJJAnalysis of N- and K-ras mutations in the distinctive tumor progression phases of melanomaJ Invest Dermatol20011171483148911886512
  • EisenTAhmadTFlahertyKTSorafenib in advanced melanoma: a Phase II randomised discontinuation trial analysisBr J Cancer20069558158616880785
  • McDermottDFSosmanJADouble-blind randomized phase II study of the combination of sorafenib and dacarbazine in patients with advanced melanoma: a report from the 11715 Study GroupJ Clin Oncol2008262178218518445842
  • HauschildAAgarwalaSSTrefzerUResults 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 melanomaJ Clin Oncol2009272823283019349552
  • FlahertyKTPuzanovIKimKBInhibition of mutated, activated BRAF in metastatic melanomaN Engl J Med201036380981920818844
  • ChapmanPHauschildARobertCUpdated overall survival (OS) results for BRIM-3, a Phase III randomized, open-label, multicenter trial comparing BRAF inhibitor vemurafenib (vem) with dacarbazine (DTIC) in previously untreated patients with BRAFV600E-mutated melanomaJ Clin Oncol201230Suppl Abstr 8502
  • SosmanJAKimKBSchuchterLSurvival in BRAF V600-mutant advanced melanoma treated with vemurafenibN Engl J Med201236670771422356324
  • NazarianRShiHWangQMelanomas acquire resistance to B-RAF (V600E) inhibition by RTK or N-RAS upregulationNature201046897397721107323
  • WagleNEmeryCBergerMFDissecting therapeutic resistance to RAF inhibition in melanoma by tumor genomic profilingJ Clin Oncol2011293085309621383288
  • KeffordRArkenauHBrownMPPhase I/II study of dabrafenib, a selective inhibitor of oncogenic mutant BRAF kinase, in patients with metastatic melanoma and other solid tumorsJ Clin Oncol201028Suppl Abstr 8503
  • KirkwoodJMLongGVTretzerUBREAK-MB: a Phase II study assessing overall intracranial response rate to dabrafenib (GSK2118436) in patient with BRAFV600E/K mutation-positive melanoma with brain metastasesJ Clin Oncol201230Suppl Abstr 8501
  • DummerRRinderknechtJGoldingerSMAn open-label pilot study of vemurafenib in previously treated metastastic melanoma patients with brain metastasesJ Clin Oncol201129Suppl Abstr 8548
  • HauschildAGrobJJDemidovLVDabrafenib in BRAF-mutated metastatic melanoma: a multicentre, open-label, phase 3 randomised controlled trialLancet201238035836522735384
  • ChuEYWanatKAMillerCJDiverse cutaneous side effects associated with BRAF inhibitor therapy: a clinicopathologic studyJ Am Acad Dermatol2012671265127222609219
  • SuFVirosAMilagreCRAS mutations in cutaneous squamous-cell carcinomas in patients treated with BRAF inhibitorsN Engl J Med201236620721522256804
  • ZimmerLHillenULivingstoneEAtypical melanocytic proliferations and new primary melanomas in patients with advanced melanoma undergoing selective BRAF inhibitionJ Clin Oncol2012302375238322614973
  • TrunzerKPavlickACSchuchterLPharmacodynamic effects and mechanisms of resistance to vemurafenib in patients with metastatic melanomaJ Clin Oncol482013 [Epub ahead of print.]
  • JohannessenCMBoehmJSKimSYCOT drives resistance to RAF inhibition through MAP kinase pathway reactivationNature201046896897221107320
  • PrahalladASunCHuangSUnresponsiveness of colon cancer to BRAF (V600E) inhibition through feedback activation of EGFRNature201248310010322281684
  • SethRCrookSIbrahemSConcomitant mutations and splice variants in KRAS and BRAF demonstrate complex peturbation of the Ras/Raf signaling pathway in advanced colorectal cancerGut2009581234124119474002
  • BaiteiEYZouMAl-MohannaFAberrant BRAF splicing as an alternative mechanism for oncogenic B-Raf activation in thyroid carcinomaJ Pathol20092170771519156774
  • FlahertyKTRobertCHerseyPImproved survival with MEK inhibition in BRAF-mutated melanomaN Engl J Med201236710711422663011
  • FlahertyKTInfanteJRDaudACombined BRAF and MEK inhibition in melanoma with BRAF V600 mutationsN Engl J Med20123671694170323020132
  • CurtinJABusamKPinkelDSomatic activation of KIT in distinct subtypes of melanomaJ Clin Oncol2006244340434616908931
  • BeadlingCJacobson-DunlopEHodiFSKIT gene mutations and copy number in melanoma subtypesClin Cancer Res2008146821682818980976
  • DemetriGDvon MehrenMBlankeCDEfficacy and safety of imatinib mesylate in advanced gastrointestinal stromal tumorsN Engl J Med200234747248012181401
  • CarvajalRDAntonescuCRWolchokJDKIT as a therapeutic target in metastatic melanomaJAMA20113052327233421642685
  • GuoJSiLKongYPhase II, open-label, single-arm trial of imatinib mesylate in patients with metastatic melanoma harboring c-Kit mutation or amplificationJ Clin Oncol2011292904290921690468
  • HodiFSFriedlanderPCorlessCLMajor response to imatinib mesylate in KIT-mutated melanomaJ Clin Oncol2008262046205118421059
  • SmalleyKSEisenTGFarnesyl transferase inhibitor SCH66336 is cytostatic, pro-apoptotic and enhances chemosensitivity to cisplatin in melanoma cellsInt J Cancer200310516517512673674
  • NiessnerHBeckDSinnbergTThe farnesyl transferase inhibitor lonafarnib inhibits mTOR signaling and enforces sorafenib-induced apoptosis in melanoma cellsJ Invest Dermatol201113146847920944654
  • GajewskiTFSalamaAKSNiedzwieckiDPhase II study of the farnesyltransferase inhibitor R115777 in advanced melanoma (CALGB 500104)J Transl Med201212101024623228035