291
Views
100
CrossRef citations to date
0
Altmetric
Review

NRAS-mutant melanoma: current challenges and future prospect

, , , &
Pages 3941-3947 | Published online: 08 Aug 2017

Abstract

Melanoma is one of the most common cutaneous cancers worldwide. Activating mutations in RAS oncogenes are found in a third of all human cancers and NRAS mutations are found in 15%–20% of melanomas. The NRAS-mutant subset of melanoma is more aggressive and associated with poorer outcomes, compared to non-NRAS-mutant melanoma. Although immune checkpoint inhibitors and targeted therapies for BRAF-mutant melanoma are transforming the treatment of metastatic melanoma, the ideal treatment for NRAS-mutant melanoma remains unknown. Despite promising preclinical data, current therapies for NRAS-mutant melanoma remain limited, showing a modest increase in progression-free survival but without any benefit in overall survival. Combining MEK inhibitors with agents inhibiting cell cycling and the PI3K–AKT pathway appears to provide additional benefit; in particular, a strategy of MEK inhibition and CDK4/6 inhibition is likely to be a viable treatment option in the future. Patients whose tumors had NRAS mutations had better response to immunotherapy and better outcomes than patients whose tumors had other genetic subtypes, suggesting that immune therapies – especially immune checkpoint inhibitors – may be particularly effective as treatment options for NRAS-mutant melanoma. Improved understanding of NRAS-mutant melanoma will be essential to develop new treatment strategies for this subset of patients with melanoma.

Introduction

Throughout the recent 5 years, there has been a significant shift in the therapeutic management of disseminated melanoma. The clinical success of BRAF-targeted therapy and immunotherapy with high responders and long survivors suggests that short- and long-term disease control can be a reality for the unclearly defined subgroups of patients with melanoma. BRAF and NRAS co-mutations are not mutually exclusive; however, the sole finding of double-mutated cells in a resistant tumor is insufficient to determine follow-up therapy, and combinational therapy targeting different pathways will be necessary.Citation1Citation4

Some specific driver mutations, each with well-known unique clinical and genetic features, have been described; these driver mutations occur in multiple oncogenes including BRAF, NRAS, and CKIT, which are the most commonly described, and may serve as potential therapeutic targets. Despite this progress, few advances have been made in developing targeted therapeutic strategies for the 50% of patients whose melanomas are BRAF wild-type (WT). The best-characterized subgroup of BRAF-WT tumors comprises 15%–20% of all melanomas that harbor activating NRAS mutations. The small GTPase, NRAS, was the first oncogene identified in melanoma and other mutational subtypes of melanoma; patients with mutant NRAS tumors tend to be older and have a history of chronic ultraviolet (UV) exposure.Citation5Citation7 Histologically, mutant NRAS tumors are more aggressive than other subtypes and have thicker lesions, elevated mitotic activity, and higher rates of lymph node metastasis.Citation8,Citation9 Given the more aggressive disease seen with mutant NRAS patients who have not received any specific therapy for the disease, it is not surprising that NRAS mutation status is a predictor of poorer outcomes in patients with melanoma who harbor this mutation, with lower median overall survival (OS) compared to non-NRAS-mutant melanoma.Citation6Citation8 Moreover, and in contrast with BRAF-mutant melanoma, little progress has been made in developing targeted therapeutic strategies for NRAS-mutant melanoma; no effective small-molecule inhibitors have been approved that specifically target NRAS, although MEK inhibitors have demonstrated modest clinical activity in a phase II trial, with clinical benefit in progression-free survival (PFS), but without a clear benefit in OS.Citation3 A better understanding of the biological and signaling characteristics of the NRAS-mutant melanoma will outline some effective therapeutic strategies for its therapeutic management that are now urgently needed.

Ras proteins as an oncogene

Despite the attention focused upon BRAF-mutant melanoma, NRAS was the first melanoma oncogene to be identified. It is known that approximately one-third of all human cancers have oncogenic mutations in the small GTPase RAS family.Citation10 The RAS family of GTPases consists of KRAS, HRAS, and NRAS. Although NRAS, KRAS, and HRAS share structural and functional similarities, mutations in KRAS are the most frequent RAS mutations in human malignant disease; moreover, in melanoma, the most commonly mutated isoform of RAS mutation typically occur at codons 12, 61, or, less frequently, 13, with 15% of cases harboring point mutations.Citation11 Whereas mutant NRAS(Q61) disrupts the GTPase activity of RAS, locking it in its active conformation, NRAS(G12) and NRAS(G13) mutations affect the Walker A-motif (p-loop) of the protein, thus decreasing its sensitivity to GTPase-accelerating proteins.Citation12,Citation13 Mutations in G12/13 and Q61 can all be described as activating; yet, they affect the NRAS protein in a distinct way as they favor the formation of GTP-bound, active RAS proteins.

Genetic evidence in experimental systems provides strong evidence that the RAF–MEK–ERK pathway is critical to the ability of RAS to induce cell proliferation, migration, and survival – highlighting the functional and biochemical relationship between RAS and this pathway in cancer.Citation14 Whereas KRAS mutations are frequent in colorectal, lung, and pancreatic cancers, NRAS mutations are, by far, the predominant alteration among RAS isoforms in melanoma.

Mutations in NRAS constitutively activate intracellular signaling through a variety of pathways – most notably, the RAS–RAF–MAPK and PI3K–AKT pathways. These mutations activate MAPK signaling to a similar degree as BRAF mutations and rarely co-occur with mutations in the PI3K–AKT pathways, suggesting that mutant NRAS drives this pathway as well. These activated signaling pathways induce cell-cycle dysregulation, pro-survival pathways, and cellular proliferation.Citation15

NRAS-mutant melanoma

NRAS was the first melanoma oncogene to be identified in 1984 in a screen of melanoma cell lines for genes that possessed transforming properties and were identified as activating mutations in NRAS in 4/30 samples.Citation16 Currently, mutations of NRAS, KRAS, or HRAS are known to be present in 20%, 2%, and 1% of all melanomas tested, respectively.Citation17 The most common oncogenic change present in >80% of all NRAS mutations is a point mutation leading to the substitution of glutamine to leucine at position 61, with mutations at positions 12 and 13 occurring with less frequency.Citation18

NRAS mutations occur at a fairly consistent rate of 15%–20% at all non-uveal sites of melanoma, including sun-exposed and sun-unexposed skin, mucosal, and acral sites of origin. This distribution contrasts with BRAF mutations, which are more common in intermittently sun-exposed skin, and with KIT mutations, which are present predominantly in mucosal and acral melanomas. Furthermore, in contrast to BRAF, NRAS mutations are rarely present in benign melanocytic nevi – with the exception of congenital nevi.Citation9 The presence of NRAS mutations in melanoma has prognostic significance.Citation12,Citation19 Typical patients harboring NRAS mutations tend to be older (>55 years) than patients with BRAF mutations, with a chronic pattern of UV exposure and lesions are usually located at the extremities and have greater levels of mitosis than BRAF-mutant melanomas. Moreover, NRAS mutations are associated with lower rates of ulceration and thicker primary tumors, with the presence of NRAS mutations an adverse prognostic factor leading to shorter MSS. Several studies examining the effect of NRAS mutations on OS have found different results; when OS was measured from the time of primary disease, NRAS mutations were found to have no impact on OS.Citation20,Citation21 However, in two other studies where OS was measured from the time of biopsy of advanced disease, NRAS mutations were associated with improved OS when compared to tumors with BRAF mutations or those WT for both.Citation22,Citation23

Current management strategies

Improved understanding of genetic and molecular basis of melanoma has revolutionized treatment options for this disease. Genetic profiling of melanomas varies widely between institutions and practices and, in spite of these advances, the treatment of melanoma remains challenging in terms of therapy selection. Because no therapeutic agents have been approved specifically for NRAS-mutant melanoma, due to the fact that several different strategies of directly targeting RAS have not resulted in effective therapeutics, mutational profiling of NRAS is not performed routinely by many clinicians although identifying NRAS mutations may have prognostic implications and facilitate clinical trial enrolment.

Farnesyl transferase inhibitors

Farnesyl transferase inhibitors are a class of drugs designed to prevent the posttranslational modification of Ras and its insertion into the plasma membrane. Although this mechanism of inhibition showed promising preclinical activity, the clinical experience in different clinical trials with these inhibitors has been very disappointing – with several serious side effects and very few responses observed.Citation24Citation26 In melanoma, farnesyl transferase inhibitors were evaluated in a small phase II trial with 14 patients whose NRAS mutation status was unknown; in this trial, none of the patients experienced a clinical response.Citation27 The lack of success observed with these drugs is attributable to the fact that many critical cellular proteins are farnesylated, in addition to Ras. Currently, there are no further ongoing clinical trials with farnesyl transferase inhibitors.

MEK inhibitors

After the lack of success in directly targeting NRAS was objectivized, focus developed toward targeting the critical signal-transduction pathways of the MAPK pathway, using MEK inhibitors. First-generation MEK inhibitors (PD 098059, U0126) showed promising inhibition in preclinical models of melanoma, but they did not progress to clinical trials.Citation28 CI-1040 (PD 184352) and its derivate PD-0325901 (PD-901) were the first MEK inhibitors tested in a clinical trial. CI-1040 was identified as a drug with a favorable safety profile, but with low oral bioavailability and high metabolism that led to plasma drug levels insufficient for antitumor activity.Citation29 PD-901 is a second-generation MEK inhibitor with some clinical response, as demonstrated in a clinical trial with 48 patients with melanoma (three patients experienced partial responses and 10 had temporary stable disease); however, the high incidence of adverse events – particularly ocular and neurologic – were observed, and limited further development of this drug.Citation30 Overall, the results of early clinical trials with early-generation MEK inhibitors were disappointing (10% objective response rate) and retrospective genotyping for the NRAS/BRAF mutations did predict for clinical benefit.

Newer MEK inhibitors have been developed with a better safety profile and antitumor activity. The first of these newer generation MEK inhibitors to be developed was selumetinib (AZD-6244; ARRY-142886) – another second-generation inhibitor of MEK1/2 with potent inhibition of cell lines in both RAS- and RAF-mutant cancers.Citation31 Initially, it was tested in a phase I trial with 11 patients with melanoma, where activity was observed with one partial response (in a NRAS-mutant patient) and seven patients had stable disease; two other clinical trials were performed, but they did not show clear benefit with selumetinib.Citation32 In a phase II trial study with BRAF-WT and NRAS-unselected patients with melanoma, selumetinib was compared with temozolomide; the study reported equivalent/inferior response rates for selumetinib (5.8% vs 9.7%) and no difference in PFS between the two groups [hazard ratio (HR) 1.07].Citation33 In another phase II trial of docetaxel with or without selumetinib in patients with BRAF-WT advanced melanoma, no difference in OS was noted between the two groups.Citation34 This agent was subsequently evaluated only in BRAF V600-mutant melanoma in combination with dacarbazine as compared to dacarbazine alone; although PFS was extended (median 5.6 vs 3.0 months), no improvement in OS was identified (13.9 vs 10.5 months).Citation35 Selumetinib has, moreover, been tested for uveal melanoma in two phase II trials with a PFS advantage, as compared to chemotherapy.Citation36,Citation37 To date, no trials have been specifically conducted for NRAS-mutant melanoma.

Clinical development of third-generation MEK inhibitors led to focused attention in targeting MEK in patients with NRAS-mutant melanoma. Both trametinib (GSK1120212) and MEK 162 (ARRY-438162) are potent inhibitors of MEK1/2, with sustained MAPK pathway inhibition at clinically achievable doses.Citation38,Citation39 Trametinib is an allosteric inhibitor of MEK1/2 that was first tested in a phase I trial among patients with advanced melanoma; efficacy was demonstrated in BRAF-mutant patients, but no responses were seen in NRAS-mutant patients.Citation40 Trametinib has received FDA approval for use in the treatment of BRAF V600-mutant melanoma as a single agent and in combination with dabrafenib. This approval was based on improved OS in a phase III trial for trametinib compared with dacarbazine in BRAF-mutant melanoma,Citation38 and the strong activity demonstrated in another phase III trial among patients with BRAF-mutant melanoma treated in the METRIC study; an overall response rate of 24% and a median PFS of 4.8 months, both significantly better than with standard chemotherapy, were observed.Citation38 Additionally, a phase III trial analyzing the efficacy of BRAF inhibition with dabrafenib alone compared to a combination of dabrafenib and trametinib demonstrated clear benefit with the combination treatment in terms of response rate, PFS, and OS.Citation41 Potential combination strategies of trametinib combined with other agents may play a role in the future of NRAS-mutant patients. Binimetinib (MEK 162) is the MEK1/2 inhibitor that was the first to show significant activity in NRAS-mutant melanoma. In a phase I trial with patients who had advanced solid tumors, MEK 162 showed promising signs of clinical activity.Citation42 In a phase II study of binimetinib in patients with advanced NRAS-mutant melanoma, 20% showed partial responses and 43.3% stable disease, with a median PFS of 3.7 months.Citation43 Based on these promising results, a randomized phase III trial, the NEMO trial was performed. In this trial, binimetinib improved PFS compared with dacarbazine (2.8 months vs 1.5 months, HR 0.62 [95% confidence interval 0.47–0.80]) and was tolerable. Grade 3–4 adverse events seen in at least 5% of patients in either group were increased creatine phosphokinase (19% vs 0%), hypertension (7% vs 2%), anemia (2% vs 5%), and neutropenia (1% vs 9%) in binimetinib group versus dacarbazine group, respectively; serious adverse events (all grades) occurred in 34% patients in the binimetinib group and in 22% patients in the dacarbazine group. Binimetinib might represent a new treatment option for patients with NRAS-mutant melanoma after failed immunotherapy.Citation44

Other MEK inhibitors such as pimasertib (AS703026), cobimetinib (GDC-0973), TAK-733, and RO4987655 have been tested for efficacy in different clinical trials with patients with melanoma. Pimasertib has been evaluated in a phase I trial with 17 NRAS-mutant patients, with two partial and two complete responses.Citation45 Cobimetinib has not been evaluated specifically for the NRAS cohort but, in combination with vemurafenib, has demonstrated a promising activity for patients with BRAF-mutant melanomaCitation46 ().

Table 1 Phase I/II/III trials of MEK inhibitors in NRAS-mutant melanoma

MEK inhibitor combinations

Although the next generation of MEK inhibitors are showing promising clinical efficacy, the relatively suboptimal response rate and PFS has led to interest in different MEK inhibitor-based combinations. The combination of MEK inhibition with RAF, EGFR–PI3K–AKT, and CDK4/6, which are the two particular pathways of interest, are currently being evaluated in clinical trials.

MEK + CDK4-6 inhibitors, which are regulators of the G1/S cell-cycle checkpoint inhibiting cancer cell growth, are being tested in phase I/II trials. Early results for the combination of ribociclib (LEE001) with binimetinib in patients with NRAS-mutant melanoma have shown a partial response in 33% and stable disease in 52% of patients.Citation47 Another phase I/II trial with a combination of trametinib and palbociclib in patients with solid tumors and with a specific cohort for NRAS-mutant melanoma is ongoing.

The combination of MEK + PI3K/AKT inhibitors has been shown to synergistically inhibit the growth of NRAS-mutant melanoma cell lines. In melanoma cell lines where BRAF inhibitor resistance is mediated through an acquired NRAS mutation, the combination of a MEK inhibitor plus a PI3K/mTOR inhibitor was noted to overcome drug resistance and inhibit cell survival.Citation48 Several early-phase studies in solid tumors have been performed, but it is not yet clear what the optimal combination of signal transduction inhibitors will be for NRAS-mutant melanoma. There are multiple phase I/II studies examining these combinations in patients with melanoma.

Other combinations for NRAS-mutant melanoma

Other targets such as polo-like kinase 1 (PLK1), TANK-binding kinase 1 (TBK1), and ROCK 1/2 are overexpressed in NRAS-mutant melanoma and the combination of MEK inhibitors plus specific inhibitors in these targets may show promising results in the different phase I/II trials that are ongoing.Citation49Citation51

Immune-based therapies

Immune therapies are playing an increasing role in the treatment of patients with metastatic melanoma, regardless of its BRAF or NRAS mutation status, particularly when there is no specific targeted therapy available.Citation52

Despite the current lack of effective and specifically approved targeted therapies for NRAS-mutant melanoma, there is some evidence that NRAS mutational status may predict for response to other therapies. Immune-based therapies are the standard of care in melanoma therapeutics, mainly used as first-line therapy particularly in patients with BRAF-WT melanoma, and are used regardless of tumor genotype. Some retrospective data suggest that patients with NRAS-mutant melanoma may have higher response rates to immunotherapies.

A retrospective analysis of patients treated with high-dose interleukin-2 demonstrated that the majority of the responders were patients with NRAS-mutant melanoma, and that those patients with either BRAF-mutant or BRAF/NRAS-WT melanoma were less likely to respond.Citation53 It is not yet clear whether BRAF or NRAS mutational status predicts for better responses in patients with melanoma receiving the antiCTLA-4 antibody ipilimumab or anti-PD-1/PD-L1 antibodies; however, some publications suggest that patients with NRAS mutations respond better to these agents.Citation54,Citation55 In the only published analysis to date, the disease control rate of patients with melanoma on ipilimumab therapy was noted to be 30% and 33% for those with and without BRAF mutations, respectivelyCitation55 ().

Table 2 Response rate and clinical benefit in patients with NRAS-mutant melanoma receiving immunotherapy

Future perspectives

Although the current target and immunotherapeutic agents may offer some hope to patients with NRAS-mutant melanoma, none of these therapies are mutation specific and have shown modest response rates and carry risk of significant toxicities. In contrast with BRAF-mutant melanoma, to date, no effective molecularly targeted therapeutic strategies have been approved for NRAS-mutant or -WT melanoma.

Many targeted strategies are now being evaluated for NRAS-mutant melanoma, although these tumors appear to be more heterogeneous than those with BRAF mutations. The most promising data from clinical investigations are with regard to MEK inhibition; however, the relatively short PFS indicates that either combination strategies or other targeted approaches will be necessary to achieve more clinically important disease responses. Both combinations for pathway interference (MEK + PI3K/mTOR and MEK + CDK 4,6) as well as combination of targeted therapy with immunotherapy are, to date, the most promising strategy to interfere with current targets refractory to chemotherapy, such as NRAS-mutant melanoma.

Conclusion

NRAS-mutant melanoma is a relatively common subtype of this disease (15%–20% of patients harbor this mutation) with a known poor prognosis. Although, currently, there are no targeted therapies that directly target NRAS, a high number of newer targeted therapeutic strategies, particularly mono- and combination therapy with MEK inhibitors, hold promise of being effective treatment strategies in the near future in the several clinical trials that are being conducted. Immune-based therapies are not genotype-specific but appear to be at least as or even more effective in the NRAS-mutant population compared to other melanoma subtypes.

Author contributions

All authors contributed toward data analysis, drafting and revising of the paper, and agree to be accountable for all aspects of the work.

Acknowledgments

The authors thank all patients with melanoma and their families for their help in participating in the investigations into malignant melanoma.

Disclosure

The authors report no conflicts of interest in this work.

References

  • RobertCSchachterJLongGVKEYNOTE-006 investigatorsPembrolizumab versus ipilimumab in advanced melanomaN Engl J Med2015372262521253225891173
  • AsciertoPAMcArthurGADrénoBCobimetinib combined with vemurafenib in advanced BRAF(V600)-mutant melanoma (coBRIM): updated efficacy results from a randomised, double-blind, phase 3 trialLancet Oncol20161791248126027480103
  • DhillonSDabrafenib plus trametinib: a review in advanced melanoma with a BRAF (V600) mutationTarget Oncol201611341742827246822
  • LarkinJChiarion-SileniVGonzalezRCombined nivolumab and ipilimumab or monotherapy in untreated melanomaN Engl J Med20153731233426027431
  • CurtinJAFridlyandJKageshitaTDistinct sets of genetic alterations in melanomaN Engl J Med2005353202135214716291983
  • JakobJABassettRLJrNgCSNRAS mutation status is an independent prognostic factor in metastatic melanomaCancer2012118164014402322180178
  • LeeJHChoiJWKimYSFrequencies of BRAF and NRAS mutations are different in histological types and sites of origin of cutaneous melanoma: a meta-analysisBr J Dermatol2011164477678421166657
  • ThumarJShahbazianDAzizSAJilaveanuLBKlugerHMMEK targeting in N-RAS mutated metastatic melanomaMol Cancer2014134524588908
  • DevittBLiuWSalemiRClinical outcome and pathological features associated with NRAS mutation in cutaneous melanomaPigment Cell Melanoma Res201124466667221615881
  • Diaz-FloresEShannonKTargeting oncogenic RasGenes Dev200721161989199217699748
  • MalumbresMBarbacidMRAS oncogenes: the first 30 yearsNat Rev Cancer20033645946512778136
  • DaudABastianBCBeyond BRAF in melanomaCurr Top Microbiol Immunol20123559911721826607
  • FedorenkoIVGibneyGTSmalleyKSNRAS mutant melanoma: biological behavior and future strategies for therapeutic managementOncogene201332253009301823069660
  • DrostenMDhawahirASumEYGenetic analysis of Ras signalling pathways in cell proliferation, migration and survivalEMBO J20102961091110420150892
  • HodisEWatsonIRKryukovGVA landscape of driver mutations in melanomaCell2012150225126322817889
  • AlbinoAPLe StrangeROliffAIFurthMEOldLJTransforming ras genes from human melanoma: a manifestation of tumour heterogeneity?Nature198430859546972
  • MilagreCDhomenNGeyerFCA mouse model of melanoma driven by oncogenic KRASCancer Res201070135549555720516123
  • BosJLRas oncogenes in human cancer: a reviewCancer Res19894917468246892547513
  • CharbelCFontaineRHMaloufGGNRAS mutation is the sole recurrent somatic mutation in large congenital melanocytic neviJ Invest Dermatol201413441067107424129063
  • AkslenLAAngeliniSStraumeOBRAF and NRAS mutations are frequent in nodular melanoma but are not associated with tumor cell proliferation or patient survivalJ Invest Dermatol2005125231231716098042
  • EllerhorstJAGreeneVREkmekciogluSClinical correlates of NRAS and BRAF mutations in primary human melanomaClin Cancer Res201117222923520975100
  • OmholtKPlatzAKanterLRingborgUHanssonJNRAS and BRAF mutations arise early during melanoma pathogenesis and are preserved throughout tumor progressionClin Cancer Res20039176483648814695152
  • PlatzAEgyhaziSRingborgUHanssonJHuman cutaneous melanoma; a review of NRAS and BRAF mutation frequencies in relation to histogenetic subclass and body siteMol Oncol20081439540519383313
  • KonstantinopoulosPAKaramouzisMVPapavassiliouAGPost-translational modifications and regulation of the RAS superfamily of GTPases as anticancer targetsNat Rev Drug Discov20076754155517585331
  • SmalleyKSEisenTGFarnesyl transferase inhibitor SCH66336 is cytostatic, pro-apoptotic and enhances chemosensitivity to cisplatin in melanoma cellsInt J Cancer2003105216517512673674
  • NiessnerHBeckDSinnbergTThe farnesyl transferase inhibitor lonafarnib inhibits mTOR signaling and enforces sorafenib-induced apoptosis in melanoma cellsJ Invest Dermatol2011131246847920944654
  • GajewskiTFSalamaAKNiedzwieckiDCancer and Leukemia Group BPhase II study of the farnesyltransferase inhibitor R115777 in advanced melanoma (CALGB 500104)J Transl Med20121024623228035
  • FridayBBAdjeiAAAdvances in targeting the Ras/Raf/MEK/Erk mitogen-activated protein kinase cascade with MEK inhibitors for cancer therapyClin Cancer Res200814234234618223206
  • RinehartJAdjeiAALorussoPMMulticenter phase II study of the oral MEK inhibitor, CI-1040, in patients with advanced non-small-cell lung, breast, colon, and pancreatic cancerJ Clin Oncol200422224456446215483017
  • LoRussoPMKrishnamurthiSSRinehartJJPhase I pharmacokinetic and pharmacodynamic study of the oral MAPK/ERK kinase inhibitor PD-0325901 in patients with advanced cancersClin Cancer Res20101661924193720215549
  • YehTCMarshVBernatBABiological characterization of ARRY-142886 (AZD6244), a potent, highly selective mitogen-activated protein kinase kinase 1/2 inhibitorClin Cancer Res20071351576158317332304
  • AdjeiAACohenRBFranklinWPhase 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 cancersJ Clin Oncol200826132139214618390968
  • KirkwoodJMBastholtLRobertCPhase II, open-label, randomized trial of the MEK1/2 inhibitor selumetinib as monotherapy versus temozolomide in patients with advanced melanomaClin Cancer Res201218255556722048237
  • GuptaALoveSSchuhADOC-MEK: a double-blind randomized phase II trial of docetaxel with or without selumetinib in wild-type BRAF advanced melanomaAnn Oncol201425596897424567366
  • RobertCDummerRGutzmerRSelumetinib plus dacarbazine versus placebo plus dacarbazine as first-line treatment for BRAF-mutant metastatic melanoma: a phase 2 double-blind randomised studyLancet Oncol201314873374023735514
  • CarvajalRDSchwartzGKMannHSmithINathanPDStudy design and rationale for a randomised, placebo-controlled, double-blind study to assess the efficacy of selumetinib (AZD6244; ARRY-142886) in combination with dacarbazine in patients with metastatic uveal melanoma (SUMIT)BMC Cancer20151546726059332
  • CarvajalRDSosmanJAQuevedoJFEffect of selumetinib vs chemotherapy on progression-free survival in uveal melanoma: a randomized clinical trialJAMA2014311232397240524938562
  • FlahertyKTRobertCHerseyPMETRIC Study GroupImproved survival with MEK inhibition in BRAF-mutated melanomaN Engl J Med2012367210711422663011
  • GilmartinAGBleamMRGroyAGSK1120212 (JTP-74057) is an inhibitor of MEK activity and activation with favorable pharmacokinetic properties for sustained in vivo pathway inhibitionClin Cancer Res2011175989100021245089
  • FalchookGSLewisKDInfanteJRActivity of the oral MEK inhibitor trametinib in patients with advanced melanoma: a phase 1 dose-escalation trialLancet Oncol201213878278922805292
  • LongGVStroyakovskiyDGogasHCombined BRAF and MEK inhibition versus BRAF inhibition alone in melanomaN Engl J Med2014371201877188825265492
  • BendellJCJavleMBekaii-SaabTSA phase 1 dose-escalation and expansion study of binimetinib (MEK162), a potent and selective oral MEK1/2 inhibitorBr J Cancer2017116557558328152546
  • AsciertoPASchadendorfDBerkingCMEK162 for patients with advanced melanoma harbouring NRAS or Val600 BRAF mutations: a non-randomised, open-label phase 2 studyLancet Oncol201314324925623414587
  • DummerRSchadendorfDAsciertoPABinimetinib versus dacarbazine in patients with advanced NRAS-mutant melanoma (NEMO): a multicentre, open-label, randomised, phase 3 trialLancet Oncol201718443544528284557
  • LeAPimasertib, a selective oral MEK 1/2 inhibitor, shows clinical activity in cutaneous and uveal metastatic melanoma in the phase I program. EADO congress2012 abstract C06
  • LarkinJAsciertoPADrénoBCombined vemurafenib and cobimetinib in BRAF-mutated melanomaN Engl J Med2014371201867187625265494
  • SosmanJAKittanehMLolkemaMPJA phase 1b/2 study of LEE011 in combination with binimetinib (MEK162) in patients with NRAS-mutant melanoma: early encouraging clinical activityJ Clin Oncol201432Suppl 159009
  • GregerJGEastmanSDZhangVCombinations of BRAF, MEK, and PI3K/mTOR inhibitors overcome acquired resistance to the BRAF inhibitor GSK2118436 dabrafenib, mediated by NRAS or MEK mutationsMol Cancer Ther201211490992022389471
  • LinCCSuWCYenCJA phase I study of two dosing schedules of volasertib (BI 6727), an intravenous polo-like kinase inhibitor, in patients with advanced solid malignanciesBr J Cancer2014110102434244024755882
  • VogelCJSmitMAMaddaloGCooperative induction of apoptosis in NRAS mutant melanoma by inhibition of MEK and ROCKPigment Cell Melanoma Res201528330731725728708
  • VuHLAplinAETargeting TBK1 inhibits migration and resistance to MEK inhibitors in mutant NRAS melanomaMol Cancer Res201412101509151924962318
  • JohnsonDBLovlyCMFlavinMImpact of NRAS mutations for patients with advanced melanoma treated with immune therapiesCancer Immunol Res20153328829525736262
  • JosephRWSullivanRJHarrellRCorrelation of NRAS mutations with clinical response to high-dose IL-2 in patients with advanced melanomaJ Immunother2012351667222130161
  • JohnsonDBPuzanovITreatment of NRAS-mutant melanomaCurr Treat Options Oncol20151641525796376
  • ShahabiVWhitneyGHamidOAssessment of association between BRAF-V600E mutation status in melanomas and clinical response to ipilimumabCancer Immunol Immunother201261573373722382362