Mutations in the small GTPase NRAS are the second most common oncogenic driver in cutaneous melanoma, being found in 15%-20% of cases.1 To date, little progress has been made in developing targeted therapy strategies for NRAS-mutant melanoma and most patients are treated with immune checkpoint inhibitors (ICI) such as anti–programmed cell death protein 1.2-4 Individuals with NRAS-mutant melanoma who do not respond to ICI therapy or subsequently develop resistance have few other therapeutic options. Ras proteins have proven difficult to target, and although inhibitors of KRAS have recently been developed with promising clinical activity reported,5-7 equivalent inhibitors of mutant NRAS are lacking. In the accompanying article, de Braud et al8 report encouraging results from the expansion arm of a phase Ib dose escalation trial of the BRAF/CRAF kinase inhibitor naporafenib (LXH254) in combination with the MEK inhibitor trametinib in NRAS-mutant melanoma. In this understanding the pathway article, we review the biology of mutant NRAS in melanoma and discuss strategies for targeting this pathway.
NRAS was first identified as a melanoma oncogene in 1984, predating the discovery of activating BRAF mutations by over 17 years.9 The most common NRAS mutations in melanoma (accounting for > 80%) are at Codon 61 (Q61R, Q61L, and Q61K) and serve to impair GTPase activity, locking the gene in a constitutively ON position.10 Melanoma is somewhat unusual in favoring NRAS mutations over KRAS or HRAS mutations (which are more common in other cancers). Recent work has suggested that the NRAS mutations associated with melanoma development (NRAS Q61R, Q61K, or Q61L) have an enhanced capacity to activate the mitogen-activated protein kinase (MAPK) pathway than the rarer NRAS mutations (G12D, G13D, G13R, Q61H, and Q61P).11 Melanomas are uniquely addicted to the MAPK signaling pathway, a highly amplified signaling cascade comprising multiple kinases (RAF, MEK, and ERK) that link external growth signals (from growth factors and receptor tyrosine kinases [RTKs]) to the transcriptional machinery of the cell (Fig 1). Virtually, all melanomas have constitutive activity in the MAPK pathway that plays a key role in malignant transformation through its effects on cell proliferation (through the maintenance of cyclin D1 expression and downregulation of cell cycle inhibitors), the suppression of apoptosis (decreased BIM expression and BAD phosphorylation), immune escape (decreased major histocompatibility complex (MHC) class 1 expression), and invasion (modulation of the actin cytoskeleton).1,12-14
FIG 1.

Overview of strategies to target mutant NRAS signaling in melanoma. NRAS is a small GTPase that serves to link extracellular growth factor signals through RTKs to cell growth and survival. In its mutated state, NRAS can signal in the absence of upstream growth factor signaling resulting in constitutive activation of (1) the MAPK pathway consisting of RAF/MEK/ERK, (2) the PI3K/AKT pathway consisting of PI3K/AKT/mTOR, (3) RAL-GDS, Ral A and Ral B, and (4) other signaling proteins, including AF6, RIN1, and PLCE. In NRAS-mutant melanoma, activation of MEK is dependent on deregulated adenylate cyclase and PKA signaling that relieves the inhibitory phosphorylation of CRAF at S43 and S233. Various strategies are being explored to target mutant NRAS in melanoma including single MEK inhibition and multiple combinations that target MEK in combination with components of the MAPK pathway (MEK + RAFi), PI3K/AKT (MEKi + AKTi, MEKi + mTORi), and growth pathways (MEKi + CDK4/6i). The combination of naporafenib + trametinib is predicted to work by limiting the relief of feedback inhibition and increased RTK signaling that results from single-agent MEKi therapy. This figure was created with BioRender. AMP, adenosine monophosphate; cAMP, cyclic AMP; MAPK, mitogen-activated protein kinase; MSH, melanocyte stimulating hormone; PDE, phosphodiesterase; PI3K, phospho-inositide-3 kinase; PKA, protein kinase A; RTK, receptor tyrosine kinase.
The majority of oncogenic drivers in melanoma strongly activate MAPK signaling, and mutant NRAS is no exception. NRAS-mutant melanomas and BRAF-mutant melanomas use different mechanisms to activate the MAPK pathways with NRAS-mutant melanomas using CRAF rather than BRAF. Mechanistically, NRAS-mediated MAPK activation involves the inhibition of BRAF activity (through phosphorylation of RAF at its inhibitory sites), and the disruption of the protein kinase-A–mediated inhibition of CRAF via increased phosphodiesterase IV expression and decreased accumulation of cyclic adenosine monophosphate (AMP).15 NRAS mutations activate multiple other signaling pathways in addition to MAPK (Fig 1). These include the phospho-inositide 3-kinase/AKT pathway,1 which contributes to melanoma development through effects upon metabolism (via regulation of mTOR and ribosomal protein S6 kinase beta-1: S6K) and cell cycle entry (through inhibition of glycogen synthase-3 kinase: GSK3beta).16 Additional oncogenic pathways activated through mutant NRAS include the Ral-guanine nucleotide exchange factors (the Ral-GEFs), such as Ral-GDS, RalA and RalB, which drive cell survival, invasion, and cell cycle entry,17,18 as well as other less characterized effectors, such as AF6, Rin1, and PLCE.1
As selective NRAS inhibitors do not yet exist, therapeutic strategies have focused on the targeting of downstream signaling pathways. One of the first approaches to be explored both clinically and preclinically was single-agent MEKi (including selumetinib, trametinib, and binimetinib), with mixed results. In an open-label, phase III, clinical trial of the MEKi binimetinib, the overall response rate was 15% with a progression-free survival (PFS) of 2.8 months.19 Greater benefit was seen in patients who had received prior ICI therapy (PFS of 5.5 months).19 The lack of single-agent MEKi activity led to the preclinical exploration of MEK inhibitor–based combination therapies. These studies were motivated by the observation that MAPK inhibition in melanoma cells (eg, BRAFi in BRAF-mutant melanoma or MEKi in NRAS-mutant melanoma) relieves feedback inhibition within the RAF/MEK/ERK signaling pathway, leading to activation of RTKs (eg, EGFR, c-MET, PDGFR, IGF1R), signaling through BRAF and CRAF and increased output through the MAPK pathway, and other parallel pathways (such as PI3K/AKT).20-22 This rebound signaling can be attenuated through vertical pathway inhibition, in which inhibitors of two pathways components are combined (eg, BRAF + MEK, RTK + MEK or MEK + ERK, etc). In NRAS-mutant melanoma, preclinical studies suggested that inhibition of adaptive growth/survival signaling could be achieved through the combination of MEKi + ERKi, MEKi + PI3K/AKTi, MEKi + mTORi, MEKi + RTKi, MEKi + CDK4i, or MEKi + HSP90i (to diminish RTK, CRAF, and CDK4 expression).23-28 Some of these combinations have been evaluated clinically. In an open-label, phase II, clinical trial, the combination of trametinib with the AKT inhibitor GSK2141795 did not lead to any objective responses in a cohort of patients with NRAS-mutant melanoma.29 By contrast, the MEKi + CDK4/6i combination of binimetinib + ribociclib did show clinical activity and was associated with a response rate of 19% and a PFS of 3.7 months.30 The response rate in patients whose melanomas had NRAS mutations in addition to concurrent CDK2NA, CDK4, or cyclin D1 alterations was > 32%.30
A more promising rational strategy to target MEK and the adaptive signaling response is the combination of a MEKi with a pan-RAFi31 (Fig 1). In preclinical studies, the MEKi + Pan-RAFi combination led to increased apoptosis and cell cycle arrest in melanoma cell lines with NRAS mutations.31 Similar findings were seen in other Ras-mutant tumors when the BRAF/CRAF inhibitor naporafenib was combined with the MEKi trametinib.32 In the phase Ib study of naporafenib + trametinib, reported here by de Braud et al,8 the evidence of antitumor activity was seen in > 46% of patients with heavily pretreated NRAS-mutant melanoma. These encouraging findings suggest that targeted therapies could be developed for NRAS-mutant melanoma. Heading forwards, the sequencing of the naporafenib + trametinib combination could prove critical. There is already evidence that patients who receive ICI as their first-line therapy have better outcomes with subsequent single-agent MEKi therapy.19 Other studies have demonstrated that upfront anti–programmed cell death protein 1 therapy can improve responses to the BRAF-MEKi combination by potentially priming the immune response.25,33 Although much remains to be done, the results of de Braud et al8 offer the possibility of developing effective targeted therapies for patients with NRAS-mutant melanoma.
Keiran S.M. Smalley
Research Funding: Revolution Medicines (Inst)
No other potential conflicts of interest were reported.
See accompanying article on page 2651
SUPPORT
Supported by the National Cancer Institute (R01 CA256193, R01 CA262483, R21 CA256289, R21 CA267141) and the Melanoma Research Foundation and Revolution Medicines.
AUTHOR CONTRIBUTIONS
Conception and design: Keiran S.M. Smalley
Collection and assembly of data: Manali S. Phadke
Manuscript writing: All authors
Final approval of manuscript: All authors
Accountable for all aspects of the work: All authors
AUTHORS' DISCLOSURES OF POTENTIAL CONFLICTS OF INTEREST
Targeting NRAS Mutations in Advanced Melanoma
The following represents disclosure information provided by authors of this manuscript. All relationships are considered compensated unless otherwise noted. Relationships are self-held unless noted. I = Immediate Family Member, Inst = My Institution. Relationships may not relate to the subject matter of this manuscript. For more information about ASCO's conflict of interest policy, please refer to www.asco.org/rwc or ascopubs.org/jco/authors/author-center.
Open Payments is a public database containing information reported by companies about payments made to US-licensed physicians (Open Payments).
Keiran S.M. Smalley
Research Funding: Revolution Medicines (Inst)
No other potential conflicts of interest were reported.
REFERENCES
- 1.Fedorenko IV, Gibney GT, Smalley KSM: NRAS mutant melanoma: Biological behavior and future strategies for therapeutic management. Oncogene 32:3009-3018, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Johnson DB, Puzanov I: Treatment of NRAS-mutant melanoma. Curr Treat Options Oncol 16:15, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Randic T, Kozar I, Margue C, et al. : NRAS mutant melanoma: Towards better therapies. Cancer Treat Rev 99:102238, 2021 [DOI] [PubMed] [Google Scholar]
- 4.Vredeveld LC, Possik PA, Smit MA, et al. : Abrogation of BRAFV600E-induced senescence by PI3K pathway activation contributes to melanomagenesis. Genes Dev 26:1055-1069, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Ostrem JM, Peters U, Sos ML, et al. : K-Ras(G12C) inhibitors allosterically control GTP affinity and effector interactions. Nature 503:548-551, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Hong DS, Fakih MG, Strickler JH, et al. : KRAS(G12C) inhibition with sotorasib in advanced solid tumors. N Engl J Med 383:1207-1217, 2020 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Skoulidis F, Li BT, Dy GK, et al. : Sotorasib for lung cancers with KRAS p.G12C mutation. N Engl J Med 384:2371-2381, 2021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.de Braud F, Dooms C, Heist RS, et al. : Initial evidence for the efficacy of naporafenib in combination with trametinib in NRAS-mutant melanoma: Results from the expansion arm of a phase Ib, open-label study. J Clin Oncol 41:2651-2660, 2023 [DOI] [PubMed] [Google Scholar]
- 9.Albino AP, Strange RL, Oliff AI, et al. : Transforming ras genes from human melanoma: A manifestation of tumour heterogeneity? Nature 308:69-72, 1984 [DOI] [PubMed] [Google Scholar]
- 10.Hodis E, Watson IR, Kryukov GV, et al. : A landscape of driver mutations in melanoma. Cell 150:251-263, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Murphy BM, Terrell EM, Chirasani VR, et al. : Enhanced BRAF engagement by NRAS mutants capable of promoting melanoma initiation. Nat Commun 13:3153, 2022 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Bhatt KV, Spofford LS, Aram G, et al. : Adhesion control of cyclin D1 and p27Kip1 levels is deregulated in melanoma cells through BRAF-MEK-ERK signaling. Oncogene 24:3459-3471, 2005 [DOI] [PubMed] [Google Scholar]
- 13.Cartlidge RA, Thomas GR, Cagnol S, et al. : Oncogenic BRAF(V600E) inhibits BIM expression to promote melanoma cell survival. Pigment Cell Melanoma Res 21:534-544, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Sumimoto H, Imabayashi F, Iwata T, et al. : The BRAF-MAPK signaling pathway is essential for cancer-immune evasion in human melanoma cells. J Exp Med 203:1651-1656, 2006 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Marquette A, Andre J, Bagot M, et al. : ERK and PDE4 cooperate to induce RAF isoform switching in melanoma. Nat Struct Mol Biol 18:584-591, 2011 [DOI] [PubMed] [Google Scholar]
- 16.Madhunapantula SV, Robertson GP: The PTEN-AKT3 signaling cascade as a therapeutic target in melanoma. Pigment Cell Melanoma Res 22:400-419, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Mishra PJ, Ha L, Rieker J, et al. : Dissection of RAS downstream pathways in melanomagenesis: A role for Ral in transformation. Oncogene 29:2449-2456, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Zipfel PA, Brady DC, Kashatus DF, et al. : Ral activation promotes melanomagenesis. Oncogene 29:4859-4864, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Dummer R, Schadendorf D, Ascierto PA, et al. : Binimetinib versus dacarbazine in patients with advanced NRAS-mutant melanoma (NEMO): A multicentre, open-label, randomised, phase 3 trial. Lancet Oncol 18:435-445, 2017 [DOI] [PubMed] [Google Scholar]
- 20.Lito P, Pratilas CA, Joseph EW, et al. : Relief of profound feedback inhibition of mitogenic signaling by RAF inhibitors attenuates their activity in BRAFV600E melanomas. Cancer Cell 22:668-682, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Paraiso KHT, Fedorenko IV, Cantini LP, et al. : Recovery of phospho-ERK activity allows melanoma cells to escape from BRAF inhibitor therapy. Br J Cancer 102:1724-1730, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Pratilas CA, Taylor BS, Ye Q, et al. : V600EBRAF is associated with disabled feedback inhibition of RAF–MEK signaling and elevated transcriptional output of the pathway. Proc Natl Acad Sci USA 106:4519-4524, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Posch C, Moslehi H, Feeney L, et al. : Combined targeting of MEK and PI3K/mTOR effector pathways is necessary to effectively inhibit NRAS mutant melanoma in vitro and in vivo. Proc Natl Acad Sci USA 110:4015-4020, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Rebecca VW, Alicea GM, Paraiso KHT, et al. : Vertical inhibition of the MAPK pathway enhances therapeutic responses in NRAS-mutant melanoma. Pigment Cell Melanoma Res 27:1154-1158, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Phadke MS, Chen Z, Li J, et al. : Targeted therapy given after anti-PD-1 leads to prolonged responses in mouse melanoma models through sustained antitumor immunity. Cancer Immunol Res 9:554-567, 2021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Haarberg HE, Paraiso KH, Wood E, et al. : Inhibition of Wee1, AKT, and CDK4 underlies the efficacy of the HSP90 inhibitor XL888 in an in vivo model of NRAS-mutant melanoma. Mol Cancer Ther 12:901-912, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Teh JLF, Cheng PF, Purwin TJ, et al. : In vivo E2F reporting reveals efficacious schedules of MEK1/2–CDK4/6 targeting and mTOR–S6 resistance mechanisms. Cancer Discov 8:568-581, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Vu HL, Aplin AE: Targeting TBK1 inhibits migration and resistance to MEK inhibitors in mutant NRAS melanoma. Mol Cancer Res 12:1509-1519, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Algazi AP, Esteve-Puig R, Nosrati A, et al. : Dual MEK/AKT inhibition with trametinib and GSK2141795 does not yield clinical benefit in metastatic NRAS-mutant and wild-type melanoma. Pigment Cell Melanoma Res 31:110-114, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Schuler M, Zimmer L, Kim KB, et al. : Phase Ib/II trial of ribociclib in combination with binimetinib in patients with NRAS-mutant melanoma. Clin Cancer Res 28:3002-3010, 2022 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Atefi M, Titz B, Avramis E, et al. : Combination of pan-RAF and MEK inhibitors in NRAS mutant melanoma. Mol Cancer 14:27, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Monaco KA, Delach S, Yuan J, et al. : LXH254, a potent and selective ARAF-sparing inhibitor of BRAF and CRAF for the treatment of MAPK-driven tumors. Clin Cancer Res 27:2061-2073, 2021 [DOI] [PubMed] [Google Scholar]
- 33.Atkins MB, Lee SJ, Chmielowski B, et al. : Combination dabrafenib and trametinib versus combination nivolumab and ipilimumab for patients with advanced BRAF-mutant melanoma: The DREAMseq trial-ECOG-ACRIN EA6134. J Clin Oncol 41:186-197, 2023 [DOI] [PMC free article] [PubMed] [Google Scholar]
