DOI:10.1093/jnci/djt075 © The Author 2013. Published by Oxford University Press. All rights reserved. For Permissions, please e-mail: [email protected]. Taming the Wild-Types: Targeting PAK1 in Melanomas That Lack BRAF Mutations Keiran S. M. Smalley, Jeffrey S. Weber

Correspondence to: Jeffrey Weber, H. Lee Moffitt Cancer Center, Department of Cutaneous Oncology, 12902 Magnolia Dr–SRB2, Tampa, FL 33612 (e-mail: [email protected]).

Remarkable progress has been made recently in identifying the PAK1 signaling for their survival (9). PAK1 is widely up-regulated molecular drivers of melanomagenesis. High-throughput genomic in a number of other human malignancies, including hormone- screening has revealed melanoma to be a diverse collection of dependent tumors such as , squamous cancer of the tumors initiated by distinct oncogenes (1). About half of all cutane- lung and skin, and melanoma (10). In uveal melanoma cell lines ous melanomas have activating mutations in the / (which typically lack BRAF mutations), elevated PAK1 expression

BRAF that drives tumor initiation and progression through correlates with invasive potential, an effect that is reversed upon its Downloaded from https://academic.oup.com/jnci/article/105/9/591/987409 by guest on 01 October 2021 the mitogen-activated kinase (MAPK) pathway. The next knockdown by small interfering RNA (siRNA) (11). most prevalent melanoma oncogene is the GTPase NRAS, which Many melanomas are “addicted” to signals through the MAPK is found in 15%-20% of cases (2). Other histological subtypes of pathway for their initiation, growth, and survival (12). The mech- melanoma, such as acral lentiginous lesions arising subungually or anism by which the MAPK pathway is activated in melanoma is on the palms or soles of the feet, or mucosal melanomas, tend not dependent upon the driving oncogene. In the majority of mela- to harbor BRAF mutations and are sometimes dependent upon the nomas, MAPK signaling is activated directly via oncogenic BRAF, genetic amplification of and/or activating mutations in the recep- which when mutated, directly phosphorylates MEK (12). In mela- tor tyrosine kinase c-KIT. Ocular melanomas lack oncogenic BRAF nomas with NRAS mutations, MAPK activity proceeds via CRAF and are associated with deleterious mutations in the G- and is dependent upon the simultaneous deregulation of protein GNAQ and GNA11 (3). Although driving oncogenic events have kinase A signaling (which would otherwise phosphorylate and inac- been identified for approximately 70% of all cutaneous melanomas, tivate CRAF) (2,13). In contrast, PAK1 activates MAPK signaling there remains a group of approximately 30% for which the initiat- through the phosphorylation of both CRAF at Ser338 and MEK1 ing event has not been determined. at Ser298 (9). Consistent with these findings, Ong and colleagues In this issue of the Journal, Ong and colleagues identify -acti- observed that siRNA knockdown of PAK1 inhibited the phospho- vating kinase 1 (PAK1) as a potential driver of and therapeutic tar- rylation of MEK and ERK in some BRAF WT cell lines (3 BRAF/ get in a subset of BRAF wild-type (WT) melanomas (4). Through NRAS WT, 1 NRAS mutant), which was associated with reduced an initial genetic and immunohistochemical analysis, PAK1 was cell viability. In a wider panel of NRAS mutant melanoma cell lines, observed to be either amplified or overexpressed in 9% and 26% PAK1 siRNA showed variable inhibition of growth. From a func- of melanomas. Segregation of the specimens on the basis of muta- tional standpoint, PAK1 knockdown was associated with inhibi- tional status revealed those with PAK1 amplification to be BRAF tion of both MEK1 and MEK2 signaling and suppression of WT, with no BRAF mutant tumor samples exhibiting copy number D1 expression. Although PAK1 has been shown to phosphorylate gain (4). When the authors considered mutational status and PAK1 MEK1 at S298 (a site that is lacking in MEK2), Ong et al. showed protein expression, the picture was more complex. Although good PAK1 knockdown to inhibit phosphorylation of MEK1 and MEK2 correlation was seen between PAK1 expression and BRAF WT at S217/S221, suggesting the inhibitory effect was mediated at the status, this was not absolute and some BRAF mutant melanomas level of CRAF (4). (4/40) showed elevation of PAK1 (4). Further analysis of the link Mutated BRAF is a bona fide therapeutic target in the majority between genotype and PAK1 expression showed a non–statistically of melanomas, and impressive clinical responses can be seen significant negative trend with NRAS mutation. Intriguingly, a small following treatment with small-molecule BRAF inhibitors such cohort of melanomas was identified that harbored both NRAS and as vemurafenib and dabrafenib (14, 15). Other targeted therapies BRAF mutations and lacked PAK1 expression (4). are being developed for other melanoma genotypes, with clinical PAK1 was first identified as a downstream effector of the small activity recently being reported for single agent MEK inhibitors Rac1 and CDC42 (5). It regulates the actin , in the 15%–20% of patients whose melanomas harbor NRAS where it helps coordinate the many signals required to control cell mutations (16). The findings of Ong et al. add PAK1 to a potential shape, adhesion dynamics, and migration (5). PAK1 is an important list of therapeutic targets for BRAF WT melanoma (4). In mediator of cell survival through the phosphorylation (and inac- agreement with their genetic studies, Ong et al. showed the PAK1 tivation of) the proapoptotic protein BAD (6, 7). A role for PAK1 inhibitor PF-3758309 to inhibit the phosphorylation of CRAF and in cancer initiation and progression has also been demonstrated, MEK in PAK1 overexpressing (BRAF WT) melanoma xenografts, with studies showing its overexpression to promote the anchorage- an effect associated with tumor stasis. Although efficacy was clearly independent growth of, and genetic abnormalities in, immortalized seen following PAK1 inhibition, the inhibition of CRAF and breast epithelial cells (8). Recent studies have also shown PAK1 to ERK phosphorylation was incomplete, raising questions over the be amplified and overexpressed in subtypes of breast and lung can- potential utility of this strategy in vivo (4). The evolving experience cers, with further work showing these tumors to be dependent upon with vemurafenib in BRAF mutant melanoma has demonstrated jnci.oxfordjournals.org JNCI | Editorials 591 that near total pathway inhibition (>90%) is required for efficacy 6. Schurmann A, Mooney AF, Sanders LC, et al. p21-activated kinase 1 phos- in patients, with recovery of MAPK pathway signaling common phorylates the death agonist bad and protects cells from apoptosis. Mol Cell Biol. 2000;20:453–461. to most mechanisms of therapeutic escape reported (17). The 7. Jin S, Zhuo Y, Guo W, Field J. p21-activated linase 1 (Pak1)-dependent phos- dependence of PAK1 overexpressing BRAF WT melanomas upon phorylation of Raf-1 regulates its mitochondrial localization, phosphoryla- CRAF/MAPK signaling may have identified another subgroup tion of BAD, and Bcl-2 association. J Biol Chem. 2005;280:24698–24705. of patients who may benefit from the new generation of highly 8. Vadlamudi RK, Adam L, Wang RA, et al. Regulatable expression of potent allosteric MEK inhibitors, such as trametinib and MEK- p21-activated kinase-1 promotes anchorage-independent growth and abnormal organization of mitotic spindles in human epithelial breast 162, or even pan RAF inhibitors. These observations fit with cancer cells. J Biol Chem. 2000;275:36238–36244. earlier work demonstrating that some melanomas lacking BRAF- 9. Ong CC, Jubb AM, Haverty PM, Zhou W, Tran V, Truong T, et al. V600E mutations are dependent on CRAF for their survival and Targeting p21-activated kinase 1 (PAK1) to induce apoptosis of tumor that knockdown/inhibition of CRAF leads to apoptosis associated cells. Proc Natl Acad Sci U S A. 2011;108:7177–7182. with inhibition of phospho-BAD/Bcl-2 (18, 19). Whether the 10. Eswaran J, Li DQ, Shah A, Kumar R Molecular pathways: targeting p21-activated kinase 1 signaling in cancer—opportunities, challenges, and PAK1 overexpressing melanomas identified by Ong et al. are part limitations. Clin Cancer Res. 2012;18(14):3743–3749. of a larger group of tumors with CRAF dependency remains to be 11. Pavey S, Zuidervaart W, van Nieuwpoort F, et al. Increased p21-activated determined. kinase-1 expression is associated with invasive potential in uveal mela- Downloaded from https://academic.oup.com/jnci/article/105/9/591/987409 by guest on 01 October 2021 Appropriate caution should be shown when contemplating the noma. Melanoma Res. 2006;16:285–296. testing of PAK1 inhibition in patients since its signaling may be 12. Smalley KS. Understanding melanoma signaling networks as the basis for molecular targeted therapy. J Invest Dermatol. 2010;130(1):28–37. important for leukocyte chemotaxis, as well as macrophage and 13. Marquette A, Andre J, Bagot M, Bensussan A, Dumaz N. ERK and PDE4 T-cell activation (20). PAK1 signaling also appears to play a role in cooperate to induce RAF isoform switching in melanoma. Nat Struct Mol neuronal migration and polarization, and its absence may be associ- Biol. 2011;18:584–591. ated with the onset of neurodegenerative diseases, suggesting that 14. Chapman PB, Hauschild A, Robert C, et al. Improved survival with neurotoxicity may be a limiting side effect of its inhibition (21). As vemurafenib in melanoma with BRAF V600E mutation. New Engl J Med. 2011;364:2507–2516. we continue to optimize targeted therapy in melanoma, resistance 15. Hauschild A, Grob JJ, Demidov LV, et al. Dabrafenib in BRAF-mutated to single-agent therapies appears inevitable. The observation by metastatic melanoma: a multicentre, open-label, phase 3 randomised con- Ong et al that PF-3758309 induced stasis and not tumor regres- trolled trial. Lancet. 2012;380:358–365. sion suggests that combination therapy may be required for PAK1- 16. Ascierto PA, Schadendorf D, Berking C, et al. MEK 162 for patients with dependent melanoma. A PAK inhibitor, possibly not alone but in advanced melanoma harbouring NRAS or Val600 BRAF mutations: a non- randomised, open-label phase 2 study. Lancet Oncol. 2013;14(3):249–256. combination with a MEK inhibitor, might have clinical activity in 17. Bollag G, Hirth P, Tsai J, Zhang J, Ibrahim PN, Cho H, et al. Clinical BRAF WT melanoma, according to the authors’ data. The authors efficacy of a RAF inhibitor needs broad target blockade in BRAF-mutant correctly assess that this constitutes an important unmet need. melanoma. Nature. 2010;467:596–599. There is some evidence that BRAF resistance to current inhibitors 18. Smalley KS, Xiao M, Villanueva J, et al. CRAF inhibition induces apoptosis in may be reversed by the use of PAK inhibitors. There is already melanoma cells with non-V600E BRAF mutations. Oncogene. 2009;28:85–94. 19. Jilaveanu LB, Zito CR, Aziz SA, et al. C-Raf is associated with disease pro- a wealth of evidence demonstrating that the PI3K/AKT/mTOR gression and cell proliferation in a subset of melanomas. Clin Cancer Res. pathway is constitutively activated in most melanomas, with pre- 2009;15:5704–5713. clinical studies showing dual MAPK and PI3K/AKT inhibition to 20. Pacheco A, Chernoff J. Group I p21-activated : emerging roles in immune be more effective than single inhibitor therapy (22,23). In addition, function and viral pathogenesis. Int J Biochem Cell Biol. 2010;42(1):13–16. other recent studies have shown BRAF WT melanomas (specifi- 21. Kreis P, Barnier JV. PAK signalling in neuronal physiology. Cell Signal. 2009; 21(3):384–393. cally melanomas with NRAS mutations) to be highly susceptible 22. Bedogni B, Welford SM, Kwan AC, Ranger-Moore J, Saboda K, Powell to the combination of a MEK and a CDK4 inhibitor (24). It is MB. Inhibition of phosphatidylinositol-3-kinase and mitogen-activated expected that ongoing progress in this area will provide critical new kinase 1/2 prevents melanoma development and promotes information that will guide future clinical trial design and bring melanoma regression in the transgenic TPRas mouse model. Mol Cancer greater benefits to melanoma patients. Ther. 2006;5:3071–3077. 23. Smalley KS, Haass NK, Brafford PA, Lioni M, Flaherty KT, Herlyn M. References Multiple signaling pathways must be targeted to overcome drug resist- ance in cell lines derived from melanoma metastases. Mol Cancer Ther. 1. Hodis E, Watson IR, Kryukov GV, et al. A Landscape of driver mutations 2006;5:1136–1144. in melanoma. Cell. 2012;150:251–263. 24. Kwong LN, Costello JC, Liu H, et al. Oncogenic NRAS signaling dif- 2. Fedorenko IV, Gibney GT, Smalley KS. NRAS mutant melanoma: bio- ferentially regulates survival and proliferation in melanoma. Nature Med. logical behavior and future strategies for therapeutic management [pub- 2012; 18(10):1503–1510. lished online ahead of print October 15, 2012]. Oncogene. doi:10.1038/ onc.2012.453 3. Van Raamsdonk CD, Bezrookove V, Green G, et al. Frequent somatic muta- Notes tions of GNAQ in uveal melanoma and blue naevi. Nature. 2009;457:599–602. JW has been a paid consultant to Glaxo SmithKline Inc. and Genetach Inc, and 4. Ong CC, Jubb AM, Jakubiak D, et al. P21-activated kinase 1 (PAK1) is a he has been a member of a speaker’s bureau for Genentech, Inc. therapeutic target in BRAF wild-type melanoma. J Natl Cancer Inst. 2013. 5. Sells MA, Knaus UG, Bagrodia S, Ambrose DM, Bokoch GM, Chernoff J. Affiliation of authors: Department of Cutaneous Oncology (KSMS, JSW), Human p21-activated kinase (Pak1) regulates actin organization in mam- Department of Molecular Oncology (KSMS), and Melanoma Research Center malian cells. Curr Biol. 1997;7:202–210. of Excellence, Moffitt Cancer Center, Tampa, FL (KSMS, JSW).

592 Editorials | JNCI Vol. 105, Issue 9 | May 1, 2013