Graphical Abstract

Cutaneous melanoma is a devastating form of skin cancer and its incidence is increasing faster than any other preventable cancer in the United States (Jemal et al, 2013). The small GTPase, NRAS, was the first oncogene identified in melanoma and is mutated in 20% of cases (Milagre et al, 2010). Compared to other mutational subtypes of melanoma, patients with mutant NRAS tumors tend to be older and have a history of chronic UV exposure (Curtin et al, 2005, Lee et al, 2011, Jakob et al, 2012, Thumar et al, 2014). Histologically, the mutant NRAS tumors are more aggressive than other subtypes with thicker lesions, elevated mitotic activity, and higher rates of lymph node metastasis (Thumar et al, 2014, Devitt et al, 2011, Ellerhorst et al, 2011). Given the more aggressive disease seen with mutant NRAS patients, it is not surprising that NRAS mutation status is a predictor of poorer outcomes with lower median overall survival compared to non-NRAS mutant melanoma (Jakob et al, 2012, Thumar et al, 2014).
In normal cells, RAS proteins switch between active GTP-bound forms and inactive GDP-bound forms. The transition between the active and inactive state is mediated by GTPase-activating proteins (GAPs). When examining the types of mutations within NRAS in melanoma patients, approximately 80–90% occur at the Q61 locus and less frequently at the G12 or G13 loci (Colombino et al, 2012). Oncogenic NRAS mutations of Q61 impair the GTP hydrolysis reaction by interfering with the coordination of a catalytic water molecule, which is required for the nucleophilic attack on the gamma-phosphate of GTP (Scheidig et al, 1999, Buhrman et al, 2010). Similarly, oncogenic substitutions in residues G12 and G13 prevent the formation of van der Waals bonds through steric hindrance between NRAS and (GAPs). This perturbs the proper orientation of the catalytic Q61 residue in NRAS, resulting in pronounced attenuation of GTP hydrolysis (Scheffzek et al, 1997). Thus, the outcome of these substitutions is the persistence of the GTP-bound state of NRAS that drives melanoma by promoting cell growth, survival and invasion (Malumbres and Barbacid, 2003). As attempts to target activated RAS directly have not led to clinical therapies, the current focus is to target RAS effector pathways. The major downstream effectors of NRAS relevant in melanoma are RAFs (ARAF, BRAF, and CRAF), phosphatidylinositol 3-kinase (PI3K), and the RAS-like protein (RAL) GEFs (Figure 1) (Downward, 2003). Although 20% of melanomas have activating NRAS mutations, the aggressive nature and complex molecular signaling conferred by this transformation has evaded clinically effective treatment options. This review examines the advances made in targeted therapies that focus on the effector pathways of NRAS.
Figure 1.
NRAS downstream effector pathways.
RAF-MEK Pathway
The RAF-MEK-ERK1/2 pathway is a well-established driver of melanoma. Upon activation, RAS recruits the serine/threonine kinase RAF to the cell membrane, leading to RAF activation. The three RAF isoforms are ARAF, BRAF, and CRAF (also known as RAF1). Activating mutations in BRAF are found in approximately 50% of melanoma patients but in a mutually exclusive manner from NRAS mutations (Davies et al, 2002). RAF kinases activate the dual specificity serine/threonine kinases MEK1 and MEK2 through phosphorylation at the S218 and S222 residues (Hanks and Quinn, 1991). MEK1 and MEK2 share 79% amino acid identity and are equally competent to phosphorylate ERK1 and ERK2 (Dhanasekaran and Premkumar Reddy, 1998), with sequential phosphorylation of ERK1 at T202 and Y204 (T185 and Y187 in ERK2). ERK1/2 signaling impacts diverse cell functions, including regulation of cell proliferation, survival, mitosis, and migration.
PI3K-AKT Pathway
The second best validated class of RAS effectors is class I PI3Ks, which is comprised of a regulatory subunit p85 and a catalytic subunit p110 with α, β, γ and δ isoforms (Mahajan and Mahajan, 2012). RAS binds and activates p110 (Castellano et al, 2013). Upon activation, all type 1 PI3Ks phosphorylate PIP2 (phosphatidylinositol (4,5)-diphosphate), a constitutive membrane component, to create the lipid second messenger PIP3 (phosphatidylinositol (3,4,5)-triphosphate). Point mutations in PIK3CA, which encodes the catalytic subunit p110α of PI3K, are detected in 2–6% of melanomas (Curtin et al, 2006, Feldman et al, 2005). PTEN (phosphatase and tensin homolog) has D3 phosphoinositide phosphatase activity and acts as a negative regulator of PI3K by removing the 3′ phosphate from its products (Feldman et al, 2005). Loss of function mutations in PTEN occur in 10–30% of melanomas (Stahl et al, 2004).
PIP3 initiates signals through binding to the plextrin homology (PH) domain of target proteins, which induces their membrane translocation and/or conformational changes. PIP3 binding to the PH domain of AKT (v-akt murine thymoma viral oncogene homolog) leads to a conformational change that enables mTORC2 (mammalian target of rapamycin 2) to directly phosphorylate AKT S473, which in turn promotes direct phosphorylation of T308 by PDK1 (3-phosphoinositide dependent protein kinase 1). The AKT protein kinase family consists of three members, AKT1-3, and they are involved in cell growth, proliferation, survival and immunity.
RALGEF-RALA/B Pathway
Other RAS effectors are members for the Ral-GEF that activate two highly similar GTPase isoforms, RALA and RALB. Although understudied, the RALA/B-mediated pathways play a key role in mutant NRAS melanomas (Mishra et al, 2010, Zipfel et al, 2010). The best-characterized effector of RAL GTPases is an octameric protein complex termed the exocyst, a large multiprotein complex responsible for the appropriate targeting and tethering of a subset of secretory vesicles to specific dynamic plasma membrane domains during exocytosis (Moskalenko et al, 2002). In addition to its involvement in cell-specific secretory products, the exocyst is also involved in the maintenance of epithelial cell polarity, cell motility and cytokinesis (Grindstaff et al, 1998, Gromley et al, 2005, Spiczka and Yeaman, 2008). In cancer cells, RAL GTPases are chronically activated. The exocyst components SEC5 and EXO84 directly interact with RAL proteins and have tethering-independent functions that support cancer cell survival. RALA regulates anchorage-independent proliferation through an unknown mechanism (Zipfel et al, 2010). RALB activates cell survival and autophagy through its interactions with the SEC5 and EXO84 exocyst components, respectively.
RAF inhibitors
As the RAF-MEK-ERK pathway is a well-established driver in melanoma, several RAF and MEK inhibitors have been evaluated. The ATP-competitive inhibitors of BRAF, vemurafenib (PLX-4032) and dabrafenib (GSK2118436), have received FDA-approval for unresectable or metastatic BRAF V600E melanoma patients. Although BRAF inhibitors could be hypothetically used in mutant NRAS melanoma to target the pathway downstream of NRAS, they are contraindicated due to paradoxical activation of MEK-ERK1/2 signaling in the setting of activated RAS signaling (Hatzivassiliou et al, 2010, Heidorn et al, 2010, Poulikakos et al, 2010, Kaplan et al, 2011). In BRAF wild-type tumors, the addition of selective BRAF inhibitors leads to BRAF recruitment to the plasma membrane in a RAS-dependent manner. Inhibited BRAF dimerizes with CRAF to form a stable complex, leading to CRAF activation and subsequent signaling downstream to MEK-ERK1/2. Therefore, in mutant NRAS tumors, inhibition of BRAF is ineffective due to inhibitor-mediated priming of signaling through BRAF-CRAF heterodimers (Hatzivassiliou et al, 2010, Heidorn et al, 2010, Poulikakos et al, 2010).
The development of pan-RAF kinase inhibitors (Compound A, TAK-632, and LY3009120) that are active against mutant BRAF as well as wild type BRAF and CRAF do not elicit paradoxical RAF-MEK signaling in RAS-transformed cancer cells (Henry et al, 2015). Compound A has been shown to block the proliferation of NRAS mutant melanomas with dependence on the MAPK pathway in vitro (Atefi et al, 2015). Both TAK-632 and LY3009120 inhibit mutant NRAS cell line xenograft in vivo as monotherapy (Nakamura et al, 2013, Peng et al, 2015). Based on the promising preclinical data, LY3009120 is currently being evaluated in patients with solid tumors in a phase I study (NCT02014116).
MEK inhibitors
Prior to the advent of pan-RAF kinase inhibitors, the primary focus in mutant NRAS tumors has been to target downstream MEK. The first generation of MEK inhibitors (PD 098059 and U0126) showed promising inhibition of growth in preclinical models of melanoma. However, due to undesirable pharmaceutical properties, the early MEK inhibitors did not progress to clinical trials (Friday and Adjei, 2008). CI-1040 (PD 184352) and its derivative PD-0325901 (PD-901) were the first MEK inhibitors to be utilized in human patients. Despite a good safety profile, the low oral bioavailability and high metabolism of CI-1040 resulted in insufficient plasma drug levels for antitumor activity (Rinehart et al, 2004). PD-901 had some clinical responses, but several adverse effects, including retinal vein occlusion and neuropathy, led to its cessation in phase II studies (LoRusso et al, 2010).
Since CI-1040 and PD-901, newer MEK inhibitors have been developed with better safety profiles and antitumor activity. The two main MEK inhibitors currently utilized in clinical trials are selumetinib (ARRY-142886, AZD6244) and trametinib (GSK1120212). Selumetinib is a selective allosteric inhibitor of MEK1/2 with an IC50 of 14 nM against purified MEK1 (Yeh et al, 2007). Due to the weak interaction between the inhibitor and MEK1 S212, treatment with selumetinib is effective in inhibiting ERK1/2 phosphorylation but leads to increased phospho-MEK (Yeh et al, 2007, Hatzivassiliou et al, 2013). In a phase II study of patients with advanced melanoma treated with either temozolomide (TMZ), an alkylating agent, or selumetinib, no difference in progression-free survival was observed between the two arms (Kirkwood et al, 2012). In a phase II study of the anti-mitotic chemotherapeutic, docetaxel, with or without selumetinib in patients with wild-type BRAF advanced melanoma, there was no difference in overall survival between the two groups (Gupta et al, 2014).
Trametinib (GSK1120212) was identified in a high-throughput screen for antiproliferative compounds that can induce expression of the CDK inhibitor, p15INK4b, as a strong and selective MEK1/2 inhibitor (Yamaguchi et al, 2011). Unlike other MEK inhibitors, trametinib preferentially binds to unphosphorylated MEK and, upon binding, has a low dissociation rate that stabilizes unphosphorylated MEK (Yoshida et al, 2012). Trametinib effectively inhibits phospho-ERK1/2 in all cells but has variable effects in proliferation assays (Gilmartin et al, 2011). A phase I trial of trametinib in patients with advanced melanoma demonstrated efficacy in mutant BRAF patients but no responses were seen in mutant NRAS patients (Falchook et al, 2012). A phase 3 trial comparing trametinib to the chemotherapeutics dacarbazine or paclitaxel in metastatic BRAF V600E/K melanoma patients showed clinical efficacy (Flaherty et al, 2012b). Trametinib received FDA approval for the treatment of BRAF V600E/K unresectable or metastatic melanoma in May 2013. Additionally, a phase 1 and 2 trial analyzing the efficacy of BRAF inhibition with dabrafenib alone compared to the combination of dabrafenib and trametinib demonstrated efficacy with the combination (Flaherty et al, 2012a). The promising results in early phase trials led to the FDA accelerated approval of the trametinib and dabrafenib combination in unresectable or metastatic melanoma with mutant BRAF V600E/K.
Next-generation MEK inhibitors
Despite the disappointing clinical trials with selumetinib and trametinib in the mutant NRAS melanoma subset (Falchook et al, 2012), newer MEK inhibitors have shown promising pre-clinical and early clinical results in RAS-driven tumors. The compounds CH5126766 (RO5126766), GDC-0623, and G-573 are dominant negative inhibitors of MEK. These dominant negative MEK inhibitors lead to the formation of a stable MEK-RAF complex, where MEK cannot be phosphorylated and released from RAF. The blockade of MEK feedback phosphorylation by RAS-activated RAF makes these inhibitors more likely to be effective in mutant RAS cancers (Hatzivassiliou et al, 2013). A phase 1 study of GDC-0623 in patients with locally advanced or metastatic solid tumors (NCT01106599) was completed but the results have not been published. In phase I studies of CH5126766, there have been responses noted in mutant NRAS melanoma patients (Martinez-Garcia et al, 2012, Zimmer et al, 2014). Of the 8 evaluable mutant NRAS melanoma patients, there were 2 with stable disease and 1 partial response.
Binimetinib (MEK162) and pimasertib (AS703026) are allosteric MEK inhibitors that have shown promise in mutant NRAS melanomas and are currently being investigated in phase 2 and 3 trials selectively in mutant NRAS melanoma. The safety profile of binimetinib and preliminary signs of antitumor activity were reported in a phase 1 trial in patients with advanced solid tumors (Bendell et al, 2011, Finn et al, 2012). In a phase 2 study of binimetinib in advanced mutant NRAS melanoma patients, 6/30 (20%) elicited partial responses and 13/30 (43.3%) showed stable disease (Ascierto et al, 2013). Based on these promising responses, this study was amended to enroll an additional 70 patients with mutant NRAS melanoma (NCT01320085). Additionally, there is an ongoing phase 3 trial in unresectable or metastatic mutant NRAS melanoma patients comparing binimetinib to the chemotherapeutic agent dacarbazine (NCT01763164). In preclinical studies, pimasertib demonstrated strong antiproliferative effects on tumor cell lines with high MEK-ERK1/2 pathway activation but little or no effect in other cells (Goutopoulos et al, 2009). In a phase 1 trial of metastatic melanoma patients, treatment of 17 mutant NRAS patients with pimasertib resulted in 2 partial responses and 2 complete responses (Lebbe et al, 2012). Based on the phase I study, there is an ongoing phase 2 trial of pimasertib compared to dacarbazine in mutant NRAS cutaneous melanoma (NCT01693068). The results from these trials may confirm the efficacy of binimetinib and pimasertib in mutant NRAS melanoma patients.
TAK-733 was discovered as a selective MEK inhibitor using structure-based drug design (Dong et al, 2011). It was found to have favorable pharmacokinetics in several animal models that support once-daily oral dosing in humans (Dong et al, 2011). In vitro testing of melanoma cell lines showed sensitivity in 7/12 mutant BRAF, 4/10 mutant NRAS, 3/5 WT/WT, and all 5 uveal cell lines (von Euw et al, 2012). In additional in vivo murine models utilizing melanoma patient-derived xenografts (PDX), TAK-733 led to tumor growth inhibition or tumor regression wild-type BRAF PDX (Micel et al, 2015). These studies provide support for the use of TAK-733 in melanomas that were wild-type for BRAF.
Combinatorial Therapies in Mutant NRAS Melanoma
Although the next generation of MEK inhibitors are showing promising clinical efficacy in an activating NRAS background, their use will most likely be optimized when combined with other targeted therapies. The combination of MEK inhibition with RAF, EGFR/PI3K/AKT, and CDK4/6 are currently being evaluated in clinical trials. Preclinical studies support the combination of MEK inhibitors with inhibitors of PLK1, TBK1, and ROCK.
RAF and ERK inhibition
For complete blockade of the MAPK pathway, targeting MEK in combination with its upstream effector, RAF, or with its downstream target, ERK1/2, has been examined. RO5126766 (CH5126766) is a potent and selective dual RAF/MEK inhibitor (Wada et al, 2014). In mutant NRAS melanoma, RO5126766 suppressed the RAF-mediated MEK phosphorylation seen with MEK inhibition alone and suppressed tumor growth in vivo. A phase I study of RO5126766 in solid tumors, including melanoma, was recently completed but results remain to be published (NCT00773526). SCH772984 is an ERK1/2 inhibitor with efficacy in melanoma cell lines, regardless of mutational status (Morris et al, 2013). The addition of SCH772984 to MEK inhibition increased apoptosis and delayed resistance in mutant NRAS melanoma cell lines (Rebecca et al, 2014).
EGFR/PI3K/AKT inhibition
EGFR is a receptor tyrosine kinase member of the ERRB receptors family, which signals through the PI3K-AKT pathway to promote survival and maintain. EGFR is frequently expressed in melanoma, and its role in mediating metastasis is well characterized (Rodeck et al, 1991). The compensatory up-regulation of AKT signaling following MEK inhibition is well characterized (Gopal et al, 2010, Deuker et al, 2015). A phospho-proteomic analysis of a MEK-inhibited mutant NRAS melanoma cell line identified up-regulation of EGFR signaling (Fedorenko et al, 2015). A high throughput antibody-based analysis of changes in MEK signal transduction in mutant NRAS melanoma cell lines identified a decrease in MIG6 (Vu et al, 2016). MIG6 is an immediate early response gene that acts as a negative regulator of ERBB receptors (Hackel et al, 2001, Ferby et al, 2006). Functionally, MIG6 was shown to negatively regulate migration and, thus, its down-regulation is likely to promote a pro-migratory phenotype in tumor microenvironments containing high levels of EGF (Vu et al, 2016).
Targeting EGFR signaling in combination with MEK in mutant NRAS melanoma may lead to more durable responses through decreased metastasis. There are few examples of EGFR inhibition in wild-type BRAF melanoma. In an in vivo xenograft model of a mutant NRAS melanoma cell line, the EGFR inhibitor, erlotinib, in combination with a VEGF binding antibody, bevacizumab, reduced metastasis to regional lymph nodes and the lungs (Schicher et al, 2009). The EGFR inhibitor gefitinib failed to show significant clinical efficacy as a single-agent therapy for unselected patients with metastatic melanoma (Patel et al, 2011). In vivo studies with the combination of MEK and EGFR inhibitors in mutant NRAS melanoma are needed to determine the anti-tumor and anti-metastasis response.
More studies have examined the combined inhibition of MEK and PI3K pathways. MEK and PI3K/mTOR inhibition has been shown to synergistically inhibit the growth of mutant NRAS melanoma cell lines in vitro and in xenograft models in vivo (Posch et al, 2013). Similarly, in a RAS-driven melanoma genetically-engineered mouse (GEM) model, the combination of selumetinib and the PI3K/mTOR inhibitor BEZ235 exhibited significant antitumor activity (Roberts et al, 2012). There is a phase II clinical study of trametinib in combination with GSK2141795, an AKT inhibitor, in wild-type BRAF melanoma (NCT01941927). Additionally, there are multiple phase I studies examining the combination of binimetinib with PI3K inhibitors in solid tumors, including melanoma (NCT01363232, NCT01337765, NCT01449058).
CDK4/6 inhibition
In an inducible NRAS-driven GEM model of melanoma, monotherapy with MEK inhibitors (selumetinib or trametinib) was not sufficient to drive tumor regression but the combination of MEK with cyclin-dependent kinase 4/6 (CDK4/6) inhibition (PD-0332991/palbociclib) resulted in tumor regression (Kwong et al, 2012). Inhibition of CDK4 and 6, regulators of the G1/S cell cycle checkpoint, arrests the cell cycle in the G1 phase, suppresses DNA synthesis, and inhibits cancer cell growth. There is an ongoing phase 1/2 trial of binimetinib with the CDK4/6 inhibitor ribociclib (LEE011) in mutant NRAS melanoma patients (NCT01781572). Early results from the 21 evaluable patients were very promising, with 7 patients achieving a partial response (33%) and 11 patients with stable disease (52%) (Sosman et al, 2014). There is also an ongoing phase 1/2 trial of trametinib with palbociclib, the CDK4/6 inhibitor, in patients with solid tumors (NCT02065063).
PLK1 inhibition
Polo-like kinase 1 (PLK1) is overexpressed in mutant NRAS melanoma (Jalili et al, 2011). As a regulator of mitosis, PLK1 influences the activation of CDK1/cyclin B, centrosome maturation, spindle formation, kinetochore assembly, regulation of microtubule nucleation, chromosome segregation and cytokinesis (Cholewa et al, 2014). In clinical trials, the PLK inhibitor volasertib was well tolerated but had poor tumor responses (Lin et al, 2014). While the clinical effects of monotherapy with PLK1 inhibitors has been disappointing, the addition of MEK inhibitors induced apoptosis in mutant NRAS melanoma both in vitro and in vivo (Posch et al, 2015).
TBK1 inhibition
TANK-binding kinase 1 (TBK1), an atypical IκB kinase family member, is activated downstream of RALB and SEC5, a component of the exocyst (Chien et al, 2006). The RALB/SEC5/TBK1 activation complex supports the host innate immune response triggered by either extracellular double-stranded RNA or exposure to Sendai virus downstream of toll-like receptor (TLR) 3 and 4 (Chien et al, 2006). Additionally, TBK1 is an essential contributor to cancer transformation in RAS-transformed tumors (Barbie et al, 2009) (Chien et al, 2006) and promotes migration in mutant NRAS melanoma (Vu and Aplin, 2014). In mutant NRAS melanoma, targeting TBK1 with small interfering RNA transfection or small molecule inhibitors (BX795 and AZ13102909) cooperates with MEK inhibition to elicit apoptosis in 3D in vitro models (Vu and Aplin, 2014).
ROCK inhibition
ROCK1/2 are RHO GTPase-activated serine/threonine kinases that regulate migration, invasion, and metastasis (Amano et al, 2010). In addition to its role in metastasis, ROCK1/2 also has a role in tumor proliferation and survival (Street and Bryan, 2011). In mutant NRAS melanoma, ROCK inhibitors (GSK269962A and fasudil), in combination with trametinib-induced cell death in vitro and suppressed the growth of established tumors in vivo (Vogel et al, 2015). While there are no clinical trials evaluating fasudil in melanoma, it is a well-tolerated agent that is approved in Japan and China for other clinical indications (Feng et al, 2015).
Conclusion
This is an exciting time for targeted therapies in mutant NRAS melanomas with several clinical trials focusing specifically on this subset. While new MEK inhibitors may prove to be efficacious as monotherapies, their combination with inhibitors of pathways known to cooperate with MEK inhibition will likely maximize antitumor activity. Furthermore, utilization of targeted agents is likely in combination with immune checkpoint inhibitors such as PD-1 antibodies, which alone have shown promising results in mutant NRAS melanoma patients (Johnson et al, 2015). Finally, attempts to directly target forms of active RAS are advancing. For example, a recently identified compound, ARS853, binds to KRAS G12C mutant when in a GDP-bound form and inhibits KRAS G12C activation of the MEK-ERK1/2 and AKT pathways (Ostrem et al, 2013, Lito et al, 2016). These studies highlight the possibility of utilizing structural information to design novel inhibitors to other mutationally activated forms of RAS. Also, recent developments have led to antibodies with some selectivity for Q61R forms of NRAS that may have diagnostic purposes (Dias-Santagata et al, 2016). Improved detection and results from current and future clinical trials will hopefully provide new effective therapeutics options for mutant NRAS melanoma patients.
Acknowledgments
Grant Support
H.L. Vu is supported by the National Institutes of Health (NIH) grant F31-CA174331. A.E. Aplin is supported by the NIH under award numbers: CA196278, CA160495, and CA182635, and grants from the Dr. Miriam and Sheldon G. Adelson Medical Research Foundation and the Melanoma Research Alliance.
We are grateful to Dr. Ed Hartsough and Michael Vido for critical reading of this manuscript.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Bibliography
- Amano M, Nakayama M, Kaibuchi K. Rho-kinase/ROCK: A key regulator of the cytoskeleton and cell polarity. Cytoskeleton. 2010;67:545–54. doi: 10.1002/cm.20472. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ascierto PA, Schadendorf D, Berking C, Agarwala SS, van Herpen CM, Queirolo P, et al. MEK162 for patients with advanced melanoma harbouring NRAS or Val600 BRAF mutations: A non-randomised, open-label phase 2 study. Lancet Oncol. 2013;14:249–56. doi: 10.1016/S1470-2045(13)70024-X. [DOI] [PubMed] [Google Scholar]
- Atefi M, Titz B, Avramis E, Ng C, Wong DJ, Lassen A, et al. Combination of pan-RAF and MEK inhibitors in NRAS mutant melanoma. Mol Cancer. 2015;14:27 015–0293-5. doi: 10.1186/s12943-015-0293-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Barbie DA, Tamayo P, Boehm JS, Kim SY, Moody SE, Dunn IF, et al. Systematic RNA interference reveals that oncogenic KRAS-driven cancers require TBK1. Nature. 2009;462:108–12. doi: 10.1038/nature08460. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bendell JC, Papadopoulos K, Jones SF, Barrett E, Guthrie K, Kass CL, et al. Phase 1 dose-escalation study of MEK inhibitor MEK162 (ARRY-438162) in patients with advanced solid tumors. AACR-NCI-EORTC. 2011:abstr B243. [Google Scholar]
- Buhrman G, Holzapfel G, Fetics S, Mattos C. Allosteric modulation of ras positions Q61 for a direct role in catalysis. Proc Natl Acad Sci U S A. 2010;107:4931–6. doi: 10.1073/pnas.0912226107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Castellano E, Sheridan C, Thin MZ, Nye E, Spencer-Dene B, Diefenbacher ME, et al. Requirement for interaction of PI3-kinase p110alpha with RAS in lung tumor maintenance. Cancer Cell. 2013;24:617–30. doi: 10.1016/j.ccr.2013.09.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chien Y, Kim S, Bumeister R, Loo YM, Kwon SW, Johnson CL, et al. RalB GTPase-mediated activation of the IkappaB family kinase TBK1 couples innate immune signaling to tumor cell survival. Cell. 2006;127:157–70. doi: 10.1016/j.cell.2006.08.034. [DOI] [PubMed] [Google Scholar]
- Cholewa BD, Pellitteri-Hahn MC, Scarlett CO, Ahmad N. Large-scale label-free comparative proteomics analysis of polo-like kinase 1 inhibition via the small-molecule inhibitor BI 6727 (volasertib) in BRAF(V600E) mutant melanoma cells. J Proteome Res. 2014;13:5041–50. doi: 10.1021/pr5002516. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Colombino M, Capone M, Lissia A, Cossu A, Rubino C, De Giorgi V, et al. BRAF/NRAS mutation frequencies among primary tumors and metastases in patients with melanoma. J Clin Oncol. 2012;30:2522–9. doi: 10.1200/JCO.2011.41.2452. [DOI] [PubMed] [Google Scholar]
- Curtin JA, Fridlyand J, Kageshita T, Patel HN, Busam KJ, Kutzner H, et al. Distinct sets of genetic alterations in melanoma. N Engl J Med. 2005;353:2135–47. doi: 10.1056/NEJMoa050092. [DOI] [PubMed] [Google Scholar]
- Curtin JA, Stark MS, Pinkel D, Hayward NK, Bastian BC. PI3-kinase subunits are infrequent somatic targets in melanoma. J Invest Dermatol. 2006;126:1660–3. doi: 10.1038/sj.jid.5700311. [DOI] [PubMed] [Google Scholar]
- Davies H, Bignell GR, Cox C, Stephens P, Edkins S, Clegg S, et al. Mutations of the BRAF gene in human cancer. Nature. 2002;417:949–54. doi: 10.1038/nature00766. [DOI] [PubMed] [Google Scholar]
- Deuker MM, Marsh Durban V, Phillips WA, McMahon M. PI3′-kinase inhibition forestalls the onset of MEK1/2 inhibitor resistance in BRAF-mutated melanoma. Cancer Discov. 2015;5:143–53. doi: 10.1158/2159-8290.CD-14-0856. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Devitt B, Liu W, Salemi R, Wolfe R, Kelly J, Tzen CY, et al. Clinical outcome and pathological features associated with NRAS mutation in cutaneous melanoma. Pigment Cell Melanoma Res. 2011;24:666–72. doi: 10.1111/j.1755-148X.2011.00873.x. [DOI] [PubMed] [Google Scholar]
- Dhanasekaran N, Premkumar Reddy E. Signaling by dual specificity kinases. Oncogene. 1998;17:1447–55. doi: 10.1038/sj.onc.1202251. [DOI] [PubMed] [Google Scholar]
- Dias-Santagata D, Su Y, Hoang MP. Immunohistochemical detection of NRASQ61R mutation in diverse tumor types. Am J Clin Pathol. 2016;145:29–34. doi: 10.1093/ajcp/aqv015. [DOI] [PubMed] [Google Scholar]
- Dong Q, Dougan DR, Gong X, Halkowycz P, Jin B, Kanouni T, et al. Discovery of TAK-733, a potent and selective MEK allosteric site inhibitor for the treatment of cancer. Bioorg Med Chem Lett. 2011;21:1315–9. doi: 10.1016/j.bmcl.2011.01.071. [DOI] [PubMed] [Google Scholar]
- Downward J. Targeting RAS signalling pathways in cancer therapy. Nat Rev Cancer. 2003;3:11–22. doi: 10.1038/nrc969. [DOI] [PubMed] [Google Scholar]
- Ellerhorst JA, Greene VR, Ekmekcioglu S, Warneke CL, Johnson MM, Cooke CP, et al. Clinical correlates of NRAS and BRAF mutations in primary human melanoma. Clin Cancer Res. 2011;17:229–35. doi: 10.1158/1078-0432.CCR-10-2276. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Falchook GS, Lewis KD, Infante JR, Gordon MS, Vogelzang NJ, DeMarini DJ, et al. Activity of the oral MEK inhibitor trametinib in patients with advanced melanoma: A phase 1 dose-escalation trial. Lancet Oncol. 2012;13:782–9. doi: 10.1016/S1470-2045(12)70269-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fedorenko IV, Fang B, Munko AC, Gibney GT, Koomen JM, Smalley KS. Phosphoproteomic analysis of basal and therapy-induced adaptive signaling networks in BRAF and NRAS mutant melanoma. Proteomics. 2015;15:327–39. doi: 10.1002/pmic.201400200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Feldman RI, Wu JM, Polokoff MA, Kochanny MJ, Dinter H, Zhu D, et al. Novel small molecule inhibitors of 3-phosphoinositide-dependent kinase-1. J Biol Chem. 2005;280:19867–74. doi: 10.1074/jbc.M501367200. [DOI] [PubMed] [Google Scholar]
- Feng Y, LoGrasso PV, Defert O, Li R. Rho kinase (ROCK) inhibitors and their therapeutic potential. J Med Chem. 2015 doi: 10.1021/acs.jmedchem.5b00683. Epub ahead of print. [DOI] [PubMed] [Google Scholar]
- Ferby I, Reschke M, Kudlacek O, Knyazev P, Pante G, Amann K, et al. Mig6 is a negative regulator of EGF receptor-mediated skin morphogenesis and tumor formation. Nat Med. 2006;12:568–73. doi: 10.1038/nm1401. [DOI] [PubMed] [Google Scholar]
- Finn RS, Javle MM, Tan BR. A phase 1 study of MEK inhibitor MEK162 (ARRY-438162) in patients with biliary tract cancer. ASCO Gastrointestinal Cancers Symposium. 2012:abstr 220. [Google Scholar]
- Flaherty KT, Infante JR, Daud A, Gonzalez R, Kefford RF, Sosman J, et al. Combined BRAF and MEK inhibition in melanoma with BRAF V600 mutations. N Engl J Med. 2012a;367:1694–703. doi: 10.1056/NEJMoa1210093. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Flaherty KT, Robert C, Hersey P, Nathan P, Garbe C, Milhem M, et al. Improved survival with MEK inhibition in BRAF-mutated melanoma. N Engl J Med. 2012b;367:107–14. doi: 10.1056/NEJMoa1203421. [DOI] [PubMed] [Google Scholar]
- Friday BB, Adjei AA. Advances in targeting the Ras/Raf/MEK/Erk mitogen-activated protein kinase cascade with MEK inhibitors for cancer therapy. Clin Cancer Res. 2008;14:342–6. doi: 10.1158/1078-0432.CCR-07-4790. [DOI] [PubMed] [Google Scholar]
- Gilmartin AG, Bleam MR, Groy A, Moss KG, Minthorn EA, Kulkarni SG, et al. GSK1120212 (JTP-74057) is an inhibitor of MEK activity and activation with favorable pharmacokinetic properties for sustained in vivo pathway inhibition. Clin Cancer Res. 2011;17:989–1000. doi: 10.1158/1078-0432.CCR-10-2200. [DOI] [PubMed] [Google Scholar]
- Gopal YN, Deng W, Woodman SE, Komurov K, Ram P, Smith PD, et al. Basal and treatment-induced activation of AKT mediates resistance to cell death by AZD6244 (ARRY-142886) in braf-mutant human cutaneous melanoma cells. Cancer Res. 2010;70:8736–47. doi: 10.1158/0008-5472.CAN-10-0902. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Goutopoulos A, Askew BC, Bankston D, Clark A, Dhanabal M, Dong R, et al. AS703026: A novel allosteric MEK inhibitor. AACR annual meeting. AACR Annual meeting. 2009:abstr 4776. [Google Scholar]
- Grindstaff KK, Yeaman C, Anandasabapathy N, Hsu SC, Rodriguez-Boulan E, Scheller RH, et al. Sec6/8 complex is recruited to cell-cell contacts and specifies transport vesicle delivery to the basal-lateral membrane in epithelial cells. Cell. 1998;93:731–40. doi: 10.1016/s0092-8674(00)81435-x. [DOI] [PubMed] [Google Scholar]
- Gromley A, Yeaman C, Rosa J, Redick S, Chen CT, Mirabelle S, et al. Centriolin anchoring of exocyst and SNARE complexes at the midbody is required for secretory-vesicle-mediated abscission. Cell. 2005;123:75–87. doi: 10.1016/j.cell.2005.07.027. [DOI] [PubMed] [Google Scholar]
- Gupta A, Love S, Schuh A, Shanyinde M, Larkin JM, Plummer R, et al. DOC-MEK: A double blind randomized phase 2 trial of docetaxel with or without selumetinib in wild-type BRAF advanced melanoma. Ann Oncol. 2014;25:968–74. doi: 10.1093/annonc/mdu054. [DOI] [PubMed] [Google Scholar]
- Hackel PO, Gishizky M, Ullrich A. Mig-6 is a negative regulator of the epidermal growth factor receptor signal. Biol Chem. 2001;382:1649–62. doi: 10.1515/BC.2001.200. [DOI] [PubMed] [Google Scholar]
- Hanks SK, Quinn AM. Protein kinase catalytic domain sequence database: Identification of conserved features of primary structure and classification of family members. Methods Enzymol. 1991;200:38–62. doi: 10.1016/0076-6879(91)00126-h. [DOI] [PubMed] [Google Scholar]
- Hatzivassiliou G, Haling JR, Chen H, Song K, Price S, Heald R, et al. Mechanism of MEK inhibition determines efficacy in mutant KRAS- versus BRAF-driven cancers. Nature. 2013;501:232–6. doi: 10.1038/nature12441. [DOI] [PubMed] [Google Scholar]
- Hatzivassiliou G, Song K, Yen I, Brandhuber BJ, Anderson DJ, Alvarado R, et al. RAF inhibitors prime wild-type RAF to activate the MAPK pathway and enhance growth. Nature. 2010;464:431–5. doi: 10.1038/nature08833. [DOI] [PubMed] [Google Scholar]
- Heidorn SJ, Milagre C, Whittaker S, Nourry A, Niculescu-Duvas I, Dhomen N, et al. Kinase-dead BRAF and oncogenic RAS cooperate to drive tumor progression through CRAF. Cell. 2010;140:209–21. doi: 10.1016/j.cell.2009.12.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Henry JR, Kaufman MD, Peng SB, Ahn YM, Caldwell TM, Vogeti L, et al. Discovery of 1-(3,3-dimethylbutyl)-3-(2-fluoro-4-methyl-5-(7-methyl-2-(methylamino)pyrido[2,3- d]pyrimidin-6-yl)phenyl)urea (LY3009120) as a pan-RAF inhibitor with minimal paradoxical activation and activity against BRAF or RAS mutant tumor cells. J Med Chem. 2015;58:4165–79. doi: 10.1021/acs.jmedchem.5b00067. [DOI] [PubMed] [Google Scholar]
- Jakob JA, Bassett RL, Jr, Ng CS, Curry JL, Joseph RW, Alvarado GC, et al. NRAS mutation status is an independent prognostic factor in metastatic melanoma. Cancer. 2012;118:4014–23. doi: 10.1002/cncr.26724. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jalili A, Moser A, Pashenkov M, Wagner C, Pathria G, Borgdorff V, et al. Polo-like kinase 1 is a potential therapeutic target in human melanoma. J Invest Dermatol. 2011;131:1886–95. doi: 10.1038/jid.2011.136. [DOI] [PubMed] [Google Scholar]
- Jemal A, Simard EP, Dorell C, Noone AM, Markowitz LE, Kohler B, et al. Annual report to the nation on the status of cancer, 1975–2009, featuring the burden and trends in human papillomavirus(HPV)-associated cancers and HPV vaccination coverage levels. J Natl Cancer Inst. 2013;105:175–201. doi: 10.1093/jnci/djs491. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Johnson DB, Lovly CM, Flavin M, Panageas KS, Ayers GD, Zhao Z, et al. Impact of NRAS mutations for patients with advanced melanoma treated with immune therapies. Cancer Immunol Res. 2015;3:288–95. doi: 10.1158/2326-6066.CIR-14-0207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kaplan FM, Shao Y, Mayberry MM, Aplin AE. Hyperactivation of MEK-ERK1/2 signaling and resistance to apoptosis induced by the oncogenic B-RAF inhibitor, PLX4720, in mutant N-RAS melanoma cells. Oncogene. 2011;30:366–71. doi: 10.1038/onc.2010.408. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kirkwood JM, Bastholt L, Robert C, Sosman J, Larkin J, Hersey P, et al. Phase II, open-label, randomized trial of the MEK1/2 inhibitor selumetinib as monotherapy versus temozolomide in patients with advanced melanoma. Clin Cancer Res. 2012;18:555–67. doi: 10.1158/1078-0432.CCR-11-1491. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kwong LN, Costello JC, Liu H, Jiang S, Helms TL, Langsdorf AE, et al. Oncogenic NRAS signaling differentially regulates survival and proliferation in melanoma. Nat Med. 2012;18:1503–10. doi: 10.1038/nm.2941. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lebbe C, Lesimple T, Raymond E, et al. Pimasertib (MSC1936369/AS703026), a selective oral MEK1/2 inhibitor, shows clinical activity in cutaneous and uveal metastatic melanoma in the phase I program. European Association of Dermato-Oncology (EADO) Congress. 2012:abstr C06. [Google Scholar]
- Lee JH, Choi JW, Kim YS. Frequencies of BRAF and NRAS mutations are different in histological types and sites of origin of cutaneous melanoma: A meta-analysis. Br J Dermatol. 2011;164:776–84. doi: 10.1111/j.1365-2133.2010.10185.x. [DOI] [PubMed] [Google Scholar]
- Lin CC, Su WC, Yen CJ, Hsu CH, Su WP, Yeh KH, et al. A phase I study of two dosing schedules of volasertib (BI 6727), an intravenous polo-like kinase inhibitor, in patients with advanced solid malignancies. Br J Cancer. 2014;110:2434–40. doi: 10.1038/bjc.2014.195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lito P, Solomon M, Li LS, Hansen R, Rosen N. Allele-specific inhibitors inactivate mutant KRAS G12C by a trapping mechanism. Science. 2016 doi: 10.1126/science.aad6204. Epub ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
- LoRusso PM, Krishnamurthi SS, Rinehart JJ, Nabell LM, Malburg L, Chapman PB, et al. Phase I pharmacokinetic and pharmacodynamic study of the oral MAPK/ERK kinase inhibitor PD-0325901 in patients with advanced cancers. Clin Cancer Res. 2010;16:1924–37. doi: 10.1158/1078-0432.CCR-09-1883. [DOI] [PubMed] [Google Scholar]
- Mahajan K, Mahajan NP. PI3K-independent AKT activation in cancers: A treasure trove for novel therapeutics. J Cell Physiol. 2012;227:3178–84. doi: 10.1002/jcp.24065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Malumbres M, Barbacid M. RAS oncogenes: The first 30 years. Nat Rev Cancer. 2003;3:459–65. doi: 10.1038/nrc1097. [DOI] [PubMed] [Google Scholar]
- Martinez-Garcia M, Banerji U, Albanell J, Bahleda R, Dolly S, Kraeber-Bodere F, et al. First-inhuman, phase I dose-escalation study of the safety, pharmacokinetics, and pharmacodynamics of RO5126766, a first-in-class dual MEK/RAF inhibitor in patients with solid tumors. Clin Cancer Res. 2012;18:4806–19. doi: 10.1158/1078-0432.CCR-12-0742. [DOI] [PubMed] [Google Scholar]
- Micel LN, Tentler JJ, Tan AC, Selby HM, Brunkow KL, Robertson KM, et al. Antitumor activity of the MEK inhibitor TAK-733 against melanoma cell lines and patient-derived tumor explants. Mol Cancer Ther. 2015;14:317–25. doi: 10.1158/1535-7163.MCT-13-1012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Milagre C, Dhomen N, Geyer FC, Hayward R, Lambros M, Reis-Filho JS, et al. A mouse model of melanoma driven by oncogenic KRAS. Cancer Res. 2010;70:5549–57. doi: 10.1158/0008-5472.CAN-09-4254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mishra PJ, Ha L, Rieker J, Sviderskaya EV, Bennett DC, Oberst MD, et al. Dissection of RAS downstream pathways in melanomagenesis: A role for ral in transformation. Oncogene. 2010;29:2449–56. doi: 10.1038/onc.2009.521. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Morris EJ, Jha S, Restaino CR, Dayananth P, Zhu H, Cooper A, et al. Discovery of a novel ERK inhibitor with activity in models of acquired resistance to BRAF and MEK inhibitors. Cancer Discov. 2013;3:742–50. doi: 10.1158/2159-8290.CD-13-0070. [DOI] [PubMed] [Google Scholar]
- Moskalenko S, Henry DO, Rosse C, Mirey G, Camonis JH, White MA. The exocyst is a ral effector complex. Nat Cell Biol. 2002;4:66–72. doi: 10.1038/ncb728. [DOI] [PubMed] [Google Scholar]
- Nakamura A, Arita T, Tsuchiya S, Donelan J, Chouitar J, Carideo E, et al. Antitumor activity of the selective pan-RAF inhibitor TAK-632 in BRAF inhibitor-resistant melanoma. Cancer Res. 2013;73:7043–55. doi: 10.1158/0008-5472.CAN-13-1825. [DOI] [PubMed] [Google Scholar]
- Ostrem JM, Peters U, Sos ML, Wells JA, Shokat KM. K-ras(G12C) inhibitors allosterically control GTP affinity and effector interactions. Nature. 2013;503:548–51. doi: 10.1038/nature12796. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Patel SP, Kim KB, Papadopoulos NE, Hwu WJ, Hwu P, Prieto VG, et al. A phase II study of gefitinib in patients with metastatic melanoma. Melanoma Res. 2011;21:357–63. doi: 10.1097/CMR.0b013e3283471073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Peng SB, Henry JR, Kaufman MD, Lu WP, Smith BD, Vogeti S, et al. Inhibition of RAF isoforms and active dimers by LY3009120 leads to anti-tumor activities in RAS or BRAF mutant cancers. Cancer Cell. 2015;28:384–98. doi: 10.1016/j.ccell.2015.08.002. [DOI] [PubMed] [Google Scholar]
- Posch C, Cholewa BD, Vujic I, Sanlorenzo M, Ma J, Kim ST, et al. Combined inhibition of MEK and Plk1 has synergistic antitumor activity in NRAS mutant melanoma. J Invest Dermatol. 2015;135:2475–83. doi: 10.1038/jid.2015.198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Posch C, Moslehi H, Feeney L, Green GA, Ebaee A, Feichtenschlager V, 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 U S A. 2013;110:4015–20. doi: 10.1073/pnas.1216013110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Poulikakos PI, Zhang C, Bollag G, Shokat KM, Rosen N. RAF inhibitors transactivate RAF dimers and ERK signalling in cells with wild-type BRAF. Nature. 2010;464:427–30. doi: 10.1038/nature08902. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rebecca VW, Alicea GM, Paraiso KH, Lawrence H, Gibney GT, Smalley KS. Vertical inhibition of the MAPK pathway enhances therapeutic responses in NRAS-mutant melanoma. Pigment Cell Melanoma Res. 2014;27:1154–8. doi: 10.1111/pcmr.12303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rinehart J, Adjei AA, Lorusso PM, Waterhouse D, Hecht JR, Natale RB, et al. Multicenter phase II study of the oral MEK inhibitor, CI-1040, in patients with advanced non-small-cell lung, breast, colon, and pancreatic cancer. J Clin Oncol. 2004;22:4456–62. doi: 10.1200/JCO.2004.01.185. [DOI] [PubMed] [Google Scholar]
- Roberts PJ, Usary JE, Darr DB, Dillon PM, Pfefferle AD, Whittle MC, et al. Combined PI3K/mTOR and MEK inhibition provides broad antitumor activity in faithful murine cancer models. Clin Cancer Res. 2012;18:5290–303. doi: 10.1158/1078-0432.CCR-12-0563. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rodeck U, Melber K, Kath R, Menssen HD, Varello M, Atkinson B, et al. Constitutive expression of multiple growth factor genes by melanoma cells but not normal melanocytes. J Invest Dermatol. 1991;97:20–6. doi: 10.1111/1523-1747.ep12477822. [DOI] [PubMed] [Google Scholar]
- Scheffzek K, Ahmadian MR, Kabsch W, Wiesmuller L, Lautwein A, Schmitz F, et al. The ras-RasGAP complex: Structural basis for GTPase activation and its loss in oncogenic ras mutants. Science. 1997;277:333–8. doi: 10.1126/science.277.5324.333. [DOI] [PubMed] [Google Scholar]
- Scheidig AJ, Burmester C, Goody RS. The pre-hydrolysis state of p21(ras) in complex with GTP: New insights into the role of water molecules in the GTP hydrolysis reaction of ras-like proteins. Structure. 1999;7:1311–24. doi: 10.1016/s0969-2126(00)80021-0. [DOI] [PubMed] [Google Scholar]
- Schicher N, Paulitschke V, Swoboda A, Kunstfeld R, Loewe R, Pilarski P, et al. Erlotinib and bevacizumab have synergistic activity against melanoma. Clin Cancer Res. 2009;15:3495–502. doi: 10.1158/1078-0432.CCR-08-2407. [DOI] [PubMed] [Google Scholar]
- Sosman JA, Kittaneh M, Lolkema, Martijn PJK, Postow MA, Schwartz G, Franklin C, et al. A phase 1b/2 study of LEE011 in combination with binimetinib (MEK162) in patients with NRAS-mutant melanoma: Early encouraging clinical activity. J Clin Oncol. 2014;32:5s, abstr 9009. [Google Scholar]
- Spiczka KS, Yeaman C. Ral-regulated interaction between Sec5 and paxillin targets exocyst to focal complexes during cell migration. J Cell Sci. 2008;121:2880–91. doi: 10.1242/jcs.031641. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stahl JM, Sharma A, Cheung M, Zimmerman M, Cheng JQ, Bosenberg MW, et al. Deregulated Akt3 activity promotes development of malignant melanoma. Cancer Res. 2004;64:7002–10. doi: 10.1158/0008-5472.CAN-04-1399. [DOI] [PubMed] [Google Scholar]
- Street CA, Bryan BA. Rho kinase proteins--pleiotropic modulators of cell survival and apoptosis. Anticancer Res. 2011;31:3645–57. [PMC free article] [PubMed] [Google Scholar]
- Thumar J, Shahbazian D, Aziz SA, Jilaveanu LB, Kluger HM. MEK targeting in N-RAS mutated metastatic melanoma. Mol Cancer. 2014;13:45, 4598-13-45. doi: 10.1186/1476-4598-13-45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vogel CJ, Smit MA, Maddalo G, Possik PA, Sparidans RW, van der Burg SH, et al. Cooperative induction of apoptosis in NRAS mutant melanoma by inhibition of MEK and ROCK. Pigment Cell Melanoma Res. 2015;28:307–17. doi: 10.1111/pcmr.12364. [DOI] [PubMed] [Google Scholar]
- von Euw E, Atefi M, Attar N, Chu C, Zachariah S, Burgess BL, et al. Antitumor effects of the investigational selective MEK inhibitor TAK733 against cutaneous and uveal melanoma cell lines. Mol Cancer. 2012;11:22. doi: 10.1186/1476-4598-11-22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vu HL, Rosenbaum S, Capparelli C, Purwin TJ, Davies MA, Berger AC, et al. MIG6 is MEK-regulated and affects EGF-induced migration in mutant NRAS melanoma. J Invest Dermatol. 2016 doi: 10.1016/j.jid.2015.11.012. Epub ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vu HL, Aplin AE. Targeting TBK1 inhibits migration and resistance to MEK inhibitors in mutant NRAS melanoma. Mol Cancer Res. 2014;12:1509–19. doi: 10.1158/1541-7786.MCR-14-0204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wada M, Horinaka M, Yamazaki T, Katoh N, Sakai T. The dual RAF/MEK inhibitor CH5126766/RO5126766 may be a potential therapy for RAS-mutated tumor cells. PLoS One. 2014;9:e113217. doi: 10.1371/journal.pone.0113217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yamaguchi T, Kakefuda R, Tajima N, Sowa Y, Sakai T. Antitumor activities of JTP-74057 (GSK1120212), a novel MEK1/2 inhibitor, on colorectal cancer cell lines in vitro and in vivo. Int J Oncol. 2011;39:23–31. doi: 10.3892/ijo.2011.1015. [DOI] [PubMed] [Google Scholar]
- Yeh TC, Marsh V, Bernat BA, Ballard J, Colwell H, Evans RJ, et al. Biological characterization of ARRY-142886 (AZD6244), a potent, highly selective mitogen-activated protein kinase kinase 1/2 inhibitor. Clin Cancer Res. 2007;13:1576–83. doi: 10.1158/1078-0432.CCR-06-1150. [DOI] [PubMed] [Google Scholar]
- Yoshida T, Kakegawa J, Yamaguchi T, Hantani Y, Okajima N, Sakai T, et al. Identification and characterization of a novel chemotype MEK inhibitor able to alter the phosphorylation state of MEK1/2. Oncotarget. 2012;3:1533–45. doi: 10.18632/oncotarget.747. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zimmer L, Barlesi F, Martinez-Garcia M, Dieras V, Schellens JH, Spano JP, et al. Phase I expansion and pharmacodynamic study of the oral MEK inhibitor RO4987655 (CH4987655) in selected patients with advanced cancer with RAS-RAF mutations. Clin Cancer Res. 2014;20:4251–61. doi: 10.1158/1078-0432.CCR-14-0341. [DOI] [PubMed] [Google Scholar]
- Zipfel PA, Brady DC, Kashatus DF, Ancrile BD, Tyler DS, Counter CM. Ral activation promotes melanomagenesis. Oncogene. 2010;29:4859–64. doi: 10.1038/onc.2010.224. [DOI] [PMC free article] [PubMed] [Google Scholar]

