Next Article in Journal
Generation of Human Regulatory Dendritic Cells from Cryopreserved Healthy Donor Cells and Hematopoietic Stem Cell Transplant Recipients
Next Article in Special Issue
CAR NK Cell Therapy for the Treatment of Metastatic Melanoma: Potential & Prospects
Previous Article in Journal
Novel In Vitro Models for Cell Differentiation and Drug Transport Studies of the Human Intestine
Previous Article in Special Issue
The Development of Nonthermal Plasma and Tirapazamine as a Novel Combination Therapy to Treat Melanoma In Situ
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

PAK1 and Therapy Resistance in Melanoma

1
Department of Immunology, Roswell Park Comprehensive Cancer Center, Elm & Carlton St., Buffalo, NY 14263, USA
2
Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Elm & Carlton St., Buffalo, NY 14263, USA
*
Author to whom correspondence should be addressed.
Cells 2023, 12(19), 2373; https://doi.org/10.3390/cells12192373
Submission received: 18 August 2023 / Revised: 23 September 2023 / Accepted: 27 September 2023 / Published: 28 September 2023
(This article belongs to the Special Issue Recent Advances in the Field of Metastatic Melanoma)

Abstract

:
Malignant melanoma claims more lives than any other skin malignancy. While primary melanomas are usually cured via surgical excision, the metastatic form of the disease portents a poor prognosis. Decades of intense research has yielded an extensive armamentarium of anti-melanoma therapies, ranging from genotoxic chemo- and radiotherapies to targeted interventions in specific signaling pathways and immune functions. Unfortunately, even the most up-to-date embodiments of these therapies are not curative for the majority of metastatic melanoma patients, and the need to improve their efficacy is widely recognized. Here, we review the reports that implicate p21-regulated kinase 1 (PAK1) and PAK1-related pathways in the response of melanoma to various therapeutic modalities. Ample data suggest that PAK1 may decrease cell sensitivity to programmed cell death, provide additional stimulation to growth-promoting molecular pathways, and contribute to the creation of an immunosuppressive tumor microenvironment. Accordingly, there is mounting evidence that the concomitant inhibition of PAK1 enhances the potency of various anti-melanoma regimens. Overall, the available information suggests that a safe and effective inhibition of PAK1-dependent molecular processes would enhance the potency of the currently available anti-melanoma treatments, although considerable challenges in implementing such strategies still exist.

1. The Therapy of Metastatic Melanoma: An Unmet Need Remains despite the Recent Progress

Melanoma is a cancer originating from melanocytes. The most prevalent variety of melanoma (cutaneous melanoma) originates from the pigment-producing cells in the epidermis. Cutaneous melanoma is the deadliest skin malignancy: the American Cancer Society estimates the yearly death toll of this disease to be at nearly eight thousand people in the USA alone [1]. While early-stage cutaneous melanoma is curable in >99% of cases, it is also known to metastasize relatively early, and fewer than a third of the patients with distal metastases survive for 5 years after diagnosis [1]. Melanomas may also arise from melanocytes at other locations, albeit with a much lower incidence. An example of that is a relatively rare uveal melanoma, which, nevertheless, is the most common type of eye cancer in adults. Uveal melanoma is commonly diagnosed when it is relatively large [2]. In this disease, the overall long-term rate of metastasis exceeds 60% [3], and the outcomes for patients who present with metastatic disease are dismal [4]. The difficulty of properly diagnosing melanomas at uncommon sites or in uncommon patient populations (e.g., children) presents additional challenges to effective management of the primary disease before the onset of metastasis [5].
The treatment of metastatic melanoma necessitates strategies beyond surgical removal. Unfortunately, historically, melanoma has proved to be notoriously resistant to conventional chemotherapy, with low response rates and a rapid emergence of therapeutic resistance. The response rates to various chemotherapeutic regimens generally remained below 20% [6,7], with severe adverse effects being relatively common and the overall survival benefits remaining uncertain [8]. The five-year survival percentage among such treated patients did not exceed single digits [9].
A better understanding of the underlying oncogenic drivers in melanoma lead to the advent of targeted therapies [10,11,12], which, to date, have shown the best results in BRAF-driven cases. However, an effective targeted therapy is still lacking for a large cohort of cases driven by other oncogenic events, while even the BRAF-driven cases commonly develop resistance in the course of treatment.
Encouragingly, cutaneous melanoma is one of the most immunogenic tumors, which made it an attractive target for immunotherapy [13,14]. A direct comparison of immunotherapy to genotoxic chemotherapy reveals that the former yields superior long-term outcomes, including in BRAF wild-type cases, where common targeted therapies would be inapplicable [15]. Indeed, over a third of patients with advanced disease can reach the 5-year survival milestone when treated with individual immune checkpoint inhibitors [16,17], and this could be increased to about half of the patients when the inhibitors are used in combination [17]. The long-term survival of some patients indicates that this approach can be curative. However, the efficacy of these drugs comes at the cost of relatively common severe adverse effects, while most of the patients still progress while under treatment. Furthermore, the efficacy of checkpoint inhibitors in uveal melanoma is much lower than in the cutaneous disease [18,19]. Most recently, tebentafusp, an ImmTAC (Immune mobilizing monoclonal T-cell receptor Against Cancer) demonstrated a statistically significant benefit in uveal melanoma [20]. However, this treatment is limited to HLA-A*02:01-positive patients and yields a rather modest increase in median survival: overall by <6 months, progression-free by <0.5 month, with a ~9% response rate [20].
Overall, despite the veritable progress in the management of melanoma, it is obvious that the currently available treatments for metastatic melanoma leave a lot of room for improvement. A better understanding of the molecular mechanisms of sensitivity and resistance to the currently available therapies leads to the identification of potentially druggable factors that may serve as intervention points to improve the efficacy of these treatments. One such factor, implicated in cancer responses to multiple treatment modalities, is p21RAC1-activated kinase 1 (PAK1) (Figure 1).

2. PAK1 Structure and Expression in Melanoma

PAK1 is a serine-threonine kinase conserved among eukaryotes as diverse as yeast, mammals, and flowering plants. Mammalian PAK1 was originally identified as a kinase, activated in the brain by Rho-family GTPases [21]. Subsequent research revealed that several homologous kinases are encoded by human and mouse genomes. Currently, this enzyme family consists of PAK1, PAK2, PAK3, PAK4, PAK5, and PAK6. Based on the degree of similarity within the family, the first three of these proteins are classified as Group I PAKs, while the rest—Group II.
The human PAK1 gene occupies over 150 kbps on the longer arm of chromosome 11. NCBI GenBank (https://www.ncbi.nlm.nih.gov/gene/5058; accessed on 19 July 2023) currently holds several dozen different transcripts of this gene, with this diversity being attributable to alternative splicing and alternative transcription initiation. Not all of these transcripts are equally well supported by experimental evidence, and some encode identical proteins despite the differences in the non-coding regions. Among the different encoded proteins, Isoform 1, consisting of 553 amino acids (Figure 2A), is the most commonly studied human variant of human PAK1. Several shorter isoforms of this protein have been reported as well. There is evidence that PAK1 isoforms may be functionally distinct [22,23] and that in melanoma, the predominant splicing pattern favors the creation of an enzymatically active PAK1 over a product of alternative splicing that encodes a truncated kinase domain (Isoform 2; Figure 2A) [24]. There is also some evidence that additional variation among the PAK1 transcripts may be introduced via alternative polyadenylation [25]. This potentially could change the susceptibility of PAK1 expression to the control of miRNAs [26]. Overall, the significance and mechanisms of the generation of the alternative PAK1 mRNAs, as well as the biological functions of the individual protein isoforms, still remain as areas in need of further exploration.
Based primarily on the structure of its kinase domain, human PAK1 is classified together with other PAKs within the STE20 family from the STE group of kinases [27] (Figure 2B). In addition to the kinase domain, PAK1 protein includes a p21RAC-binding domain (PBD), which overlaps with an auto-inhibitory domain (AID), as well as several conserved binding sites for adaptor proteins and guanine nucleotide exchange factors [28,29] (Figure 2A). Immediately preceding the PBD is a stretch of basic amino acids, which allows PAK1 to bind to certain lipids [30].
Figure 2. PAK1 structure and position within human kinome. (A) Conserved elements within PAK1 isoforms 1 and 2. (B) Position of PAK proteins on the evolutionary tree of human kinases. The evolutionary tree of kinases was drawn using KinMap [31].
Figure 2. PAK1 structure and position within human kinome. (A) Conserved elements within PAK1 isoforms 1 and 2. (B) Position of PAK proteins on the evolutionary tree of human kinases. The evolutionary tree of kinases was drawn using KinMap [31].
Cells 12 02373 g002aCells 12 02373 g002b
In its inactive state, PAK1 is a “head-to-tail” dimer, where the AIDs are bound to and inhibit the kinase domains [29]. The best-known mode of PAK1 regulation is its activation by the small GTPases of the Rho family, such as p21RAC1, from which the name of PAK1 is derived. These small GTPases gain affinity to PAK1 while bound to GTP [32], and this interaction relieves the kinase domain from the AID. This, in turn, facilitates the formation of a “face-to-face” dimer, where the monomers cross-phosphorylate each other, thus achieving full activation [33,34]. Importantly, constitutively active mutants of these GTPases demonstrate an enhanced affinity to PAK1 [35]. This mode of regulation also implies that other potential interactors of the autoinhibitory domain that diminish its affinity for the kinase domain would act as activators of PAK1. The interaction of PAK1 with certain lipids may also contribute to its activation, as well as to membrane localization [30,36].
Activated PAK1 is known to phosphorylate a great number of targets. While their complete inventory is beyond the scope of this review, the numerous biochemical and biological consequences of PAK1 activity have been extensively reviewed elsewhere [28,37,38,39,40]. The involvement of many of the PAK1-regulated factors in such phenomena as cell motility, cell proliferation, and survival support the continuous interest in PAK1 among cancer researchers and has led to the recognition of this enzyme as a bona fide target for anti-cancer therapy [28]. Indeed, there is abundant evidence of PAK1 being essential for the survival and proliferation of at least some cancer types, and this dependence correlates with the nature of oncogenic drivers in these malignancies. Melanomas commonly display elevated levels of PAK1 expression as well as its activity, with the latter being judged by the abundance of the appropriately phosphorylated form [41,42,43]. As mentioned above, PAK1 mRNA splicing in melanoma is skewed towards producing the kinase-proficient isoform of the protein [24]. The PAK1 gene is amplified in 5–9% of cutaneous melanomas [41,43]. The incidence is higher in acral melanomas [44,45], which also tend to retain wild-type BRAF and portent worse outcomes [46]. Importantly, the amplicon often includes other cancer-relevant genes (e.g., CCND1 and GAB2), which might further cooperate with PAK1 in oncogenesis. Nevertheless, gene amplification may explain only a fraction of the cases of PAK1 overexpression, and additional mechanisms that account for higher PAK1 levels are yet to be firmly established. The hyperactivation of PAK1 in cutaneous melanomas may be further attributed to activating mutations in RAC1 [47,48], but also to even more common activating mutations in NRAS and the inactivation of RAS inhibitor NF1 [47], as RAS-induced transformation involves and depends on the activation of RAC1 and PAK1 [49,50,51,52]. Activating mutations in KRAS and HRAS are also expected to activate PAK1, although these mutations are present in less than 2% of melanomas each [53]. Frequent mutations in PREX2 [54] which increase its capacity to act as a guanine nucleotide exchange factor (GEF) for RAC1 [55] are also expected to upregulate PAK1. Mutations in the KIT gene, encoding receptor tyrosine kinase c-KIT, are typically found in acral and mucosal melanomas [56,57] and may be predicted to activate a PAK1-mediated oncogenic pathway, as was documented in other malignancies [58]. The co-occurrence of several mutations that are potentially capable of activating PAK1 is not uncommon in cutaneous melanoma [59]. In a small subset of cutaneous melanomas, and in the vast majority of uveal melanomas, the activation of PAK1 is expected as a consequence of activating mutations in the GNAQ and GNA11 oncogenes, which reportedly signal through RAC1 and RAS [60,61]. Finally, there are intriguing observations that BRAF can directly activate PAK1 in the context of thyroid cancer [62]. If confirmed in the context of cutaneous melanoma, this phenomenon might be of great importance due to the high prevalence of BRAF mutations in this disease.

3. PAK1 and Genotoxic Therapy in Melanoma

In general, melanoma is relatively resistant to genotoxic chemo- and radiotherapy. Various conventional genotoxic chemotherapeutic regimens, primarily based on the alkylating agent dacarbazine, are known to achieve response in ~20% of metastatic cutaneous melanoma cases, with responses usually lasting for several months [9,63]. Prior attempts to improve the efficacy of such regimens by combining multiple genotoxic agents failed to yield a significant clinical benefit despite noticeably increasing the incidence of adverse effects [63].
Radiation therapy in melanoma is used to manage some primary tumors with limited surgical options, with acceptable long-term control in most lentigo maligna and uveal melanomas [64,65], although with less encouraging results in the mucosal form of the disease [66]. As an adjuvant for lymphadenectomy, radiation therapy offers short-term benefits, but does not improve the overall survival [67]. Radiation therapy is also commonly used for metastatic melanoma, where the effect is often palliative, but hardly ever curative [68]. An obvious drawback of radiation therapy is the damage to normal tissues, including vital organs [69].
There is sufficient evidence to implicate PAK1 in the response of cancer cells to genotoxic impacts, including those delivered in the course of therapy. Reportedly, PAK1 can be activated by at least some types of DNA damage [70]. Various lines of evidence point to the role of PAK1 in DNA repair [71]. A comparison between PAK1 wild-type and knockout cells following ionizing radiation suggested that PAK1 affects a wide array of genes and proteins with known roles in the DNA repair and DNA damage responses [72]. Those experiments were conducted on a single pair of mouse embryonic fibroblast cell lines, so their direct relevance to human melanoma cannot be taken for granted. Importantly, it was also reported that PAK1 can suppress the sensing of DNA damage and increase the tolerance of melanoma cells to genotoxic treatment [73]. In particular, PAK1 appears to attenuate p53 activation in human melanoma cells with wild-type p53 [73], and this parallels the de-repression of some of the p53 targets observed in PAK1-deficient mouse fibroblasts [72].
These observations add to the list of several previously reported avenues through which PAK1 may affect the pathway of programmed cell death: the inactivation of pro-apoptotic proteins BIM [74], BAD [75,76], and FOXO1 [77], and the stimulation of a pro-survival transcription factor, NF-κB [78]. Notably, some of the NF-κB targets are secreted factors that provide additional signals to further activate NF-κB [79]. Therefore, some of the consequences of PAK1 activation may not be cell-autonomous. Admittedly, these connections between PAK1 and the apoptotic machinery have been predominantly investigated in non-melanoma models, and their relevance to life/death decisions in melanoma cells is yet to be rigorously established. Nevertheless, the presence of the same factors and the corresponding pathways in melanoma cells makes it possible that these observations are relevant to melanoma as well.
The drive to increase the efficacy of conventional genotoxic therapy for melanoma has been somewhat obscured by the successes of other treatment modalities. However, the frequent eventual failure of the latter leaves unmet the need for effective care for heavily pre-treated recurring disease. In this context, there may be an opportunity to exploit PAK1 inhibition in conjunction with chemo- or radiotherapy to improve the plight of this category of patients.

4. PAK1 and Targeted Therapies in Melanoma

PAK1 is positioned amidst multiple intracellular signaling pathways, including the ones that control cancer-relevant traits such as proliferation, resistance to apoptosis, invasiveness, motility, etc. [28]. In the context of melanoma, the so-called MAPK cascade is of a particular importance. This cascade normally transmits and amplifies the signal from the plasma membrane to intracellular components, including the nucleus. The pathway not only promotes cell growth and survival, but also imparts widespread changes in cell metabolism [80]. In the canonical scheme, the engagement of a receptor tyrosine kinase eventually leads to the activation of an RAS family protein, which in turn recruits to the plasma membrane and activates an RAF family member, which phosphorylates a kinase of the MEK family, whose targets are the MAP kinases, including those known as ERKs [81]. There are numerous variations on this theme, with various members of the RAS, RAF, MEK, and MAPK/ERK families playing different roles in different contexts. The abovementioned activating mutations in BRAF, NRAS, HRAS, KRAS, KIT, GNAQ, and GNA11 are believed to exert their oncogenic effects, either fully or in part, through the MAPK cascade. The important modulation of this cascade is provided by the additional regulators of the key players. This includes the abovementioned negative regulator of RAS: NF1, which is lost in some cutaneous melanomas [47]. Another notable example is the phosphorylation of both CRAF and MEK1 proteins by PAK1, which reportedly provides a co-stimulatory signal [82,83,84] and facilitates the association between MEK and ERK proteins [85]. Common mutations in RAC1 and PREX2, which eventually activate PAK1, are expected to have the same effect. Mucosal melanomas, which are a very rare type of the disease, have a mutation landscape that is quite diverse and somewhat distinct from those of the cutaneous and uveal forms, but the majority of its cases are still expected to carry an activated MAPK cascade [86,87].
Targeting the MAPK cascade has emerged as a standard of care for BRAF-mutant melanoma, where combinations of BRAF and MEK inhibitors have proven to be especially effective at prolonging survival [88,89,90]. Unfortunately, even under a combined therapy, complete responses are achieved in the minority of cases, and are commonly followed by disease recurrence. The responsiveness of NRAS-driven melanomas to targeted therapies is even worse, as BRAF inhibitors are generally inapplicable in this scenario, while MEK inhibitors alone do not offer significant improvements over conventional chemotherapy in the overall survival or the safety profiles [91,92]. Similarly, a MEK inhibitor failed to significantly improve the overall survival in GNAQ- and GNA11-driven uveal melanoma, either when used alone or added to conventional chemotherapy [93,94].
The mechanisms of resistance to the inhibitors of the MAPK cascade have received considerable attention. Predictably, activating mutations in NRAS oncogenes provide an alternative pathway to MEK activation, negating the utility of BRAF inhibitors, and are present in therapy-resistant cases [95,96]. Among the earliest recognized resistance mechanisms was also the activation of the PI3K/AKT pathway [41,97,98]. This pathway is long-known to have oncogenic properties [99], and to increase the tolerance to a variety of potentially lethal impacts [100,101,102]. In the context of cancer, the activation of this pathway occasionally may be achieved via activating mutations or the amplification of individual AKT isoforms or, more commonly, via activating mutations in the catalytic subunits of PI3K, via the loss of the tumor suppressor PTEN, or via the activation of receptor tyrosine kinases [96,103] that serve as upstream regulators of PI3K. A number of additional resistance mechanisms have been noted, such as those provided by the Hippo pathway [23] and the kinase PLK3 [104].
PAK1 is an evolutionarily conserved regulator of the MAPK cascade; even in yeast, this cascade is regulated by a PAK1 homologue, Ste20 [105]. Importantly, the role of PAK1 as a co-activator of the MAPK cascade, as well as its close interaction with the PI3K/AKT pathway [106,107], positions PAK1 as a candidate modulator of cell responses to targeted therapies. Singhal et al. discovered that the sensitivity to PAK inhibition distinguishes NRAS- from BRAF-mutant melanomas [108], and this observation was later extended by Ong et al. [43]. The dependence of RAS-mediated transformation on RAC is also known from other systems [50,51,52], but the exact reason for the different drug sensitivity is yet to be firmly established. Of note, it was reported that BRAF, and especially its activated mutant, differ from CRAF based on a reduced dependence on the status of the PAK1 phosphorylation site [84]. Interestingly, it was noted that the inhibition of PAK1 also made NRAS-mutant cells more sensitive to the inhibitors of the MAPK cascade [108], suggesting an avenue to sensitize to targeted therapy this otherwise relatively resistant tumor variant.
The ability of PAK1 to provide additional stimulation to CRAF and MEKs points to a possibility that PAK1 may offer an alternative route for maintaining the minimally required level of ERK activity in BRAF-mutant cells treated with a BRAF inhibitor. Indeed, Babagana et al. demonstrated that in BRAF-mutant cells, the inhibition of PAK1 decreases, while its activation increases the tolerance to BRAF and MEK inhibitors [41]. This phenomenon was later confirmed in an extensive study by Lu et al. [109]. Accordingly, the resistant phenotype can be rendered not only by PAK1 itself, but also by PAK1 activators, such as RAC1 [41,110] and RHOJ [111], and such resistance is negated by PAK1 inhibition. This is in line with the observation that RAC1-driven oncogenesis and metastasis depend on PAK1 [112,113]. Interestingly, the inhibition of PAK1 can also negate the resistance to BRAF inhibitors that is afforded by hyperactive AKT [41]. As mentioned above, AKT is implicated as a key downstream effector of multiple other resistance factors, so the ability to negate its effect potentially expands the utility of PAK1 inhibition as a countermeasure for acquired drug resistance.
The recognition of the role of PAK1 in many BRAF wild-type cutaneous melanomas has brought about the question of whether it plays a similar role in the GNA-driven uveal form of the disease [114]. Indeed, as mentioned above, there is evidence that GNA-driven oncogenesis involves the activation of RAS, RAC1, and PAK1. Accordingly, one may predict that, akin to the RAS-driven cases, PAK inhibition in GNA-driven melanomas would enhance the efficacy of targeted therapies. Indeed, a significant synergy between MEK and PAK inhibitors is seen in cultured uveal melanoma cells (Figure 3). Importantly, the synergy is seen even between doses of the drugs that are largely ineffectual when used alone. This provides the hope that these types of drugs, which have not seen a great success in single-drug trials yet, might be more useful in a combined regimen.
It is worth noting that the role of PAK1 in the resistance to targeted therapies is not necessarily limited to its direct effects on the MAPK and AKT pathways. The anti-apoptotic activity of PAK1, including its inhibitory phosphorylation of BAD, may contribute to the phenomenon as well [111]. Moreover, PAK1 has a mutually inhibitory relationship with the tumor suppressor NF2 (aka Merlin) [118,119,120]. The loss of NF2 activity inhibits LATS kinases and consequently relieves from inhibition their target YAP [121], a known determinant of resistance to targeted therapies in melanoma [23].
Overall, ample evidence suggests that PAK1 by itself is a critical vulnerability in a sizeable subset of melanomas, while its co-targeting has the potential to enhance MAPK-directed targeted therapies and circumvent many common modes of resistance to the latter.

5. PAK1 and Immunotherapy of Melanoma

The immune system presents a major anti-cancer defense mechanism, but its effectiveness is often attenuated by the immunosuppressive microenvironment created within tumors, as well as the reduced propensity of tumor cells to present their antigens for immunosurveillance. Modern immune therapies most commonly target the “immune checkpoints”, which are exploited by melanoma cells to curtail the activity of cytotoxic T-cells [14,122]. Another type of immunotherapeutic, which to date has shown some efficacy in uveal melanoma, is the ImmTAC (Immune-mobilizing monoclonal T-cell receptor Against Cancer). An ImmTAC acts by tethering T-cells to target cancer cells. A clinically effective [20] ImmTAC, tebentafusp, combines a soluble T-cell receptor that is specific for HLA-bound melanocyte lineage-specific antigen gp100 with an antibody fragment that engages the CD3 receptors on T-cells [123]. Importantly, both the “checkpoint inhibitors” and ImmTAC depend on the successful penetration of T-cells into the tumor and on the presentation of tumor-specific antigens on the surface of tumor cells. The factors that affect these two parameters are expected to affect the efficacy of immunotherapy. This consideration provides some insights into the frequent failure of this therapeutic modality, as well as the avenues to increase its efficacy.
Multiple lines of investigation point to the effects of PAK1 on tumors’ interactions with the immune system and on the various aspects of immunotherapy. Not all of the observations were validated in the context of melanoma, and the relative contribution of various phenomena to the clinical outcomes is yet to be established. The high significance of the topic makes it a very active field of research.
The chemical inhibition or genetic inactivation of PAK reportedly enhanced immunity in a genetically engineered mouse model of intestinal tumorigenesis [124]. Importantly, the absence of PAK1 led to an increase in splenic lymphocytes only in tumor-bearing mice, but not in their tumor-free counterparts, suggesting that the effect is specific for anti-tumor immunity [124]. Interestingly, in this model, the tumor itself had an immunosuppressive effect, which was negated by PAK1 inactivation [124]. The exact mechanism of this phenomenon remains unknown. In part, the role of PAK1 in immune evasion may be expected based on its contribution to the MAPK cascade. Numerous reports suggest that the MAPK cascade contributes to immune evasion in various systems [125,126,127,128] and its inhibition augments immune therapy [129,130,131]. In particular, this pathway appears to enhance the secretion of immunosuppressive cytokines by cancer cells [132] and to limit the ability of cancer cells to present their potential antigens on cell surfaces, while the corresponding inhibitors upregulate HLA class I molecules [125,127]. Intriguingly, a direct interaction was reported between PAK1 and HLA-A, -B, and -C proteins [133], although the biological significance of this phenomenon is still unexplored. Furthermore, the inhibition of the MAPK cascade increases the expression of melanocyte differentiation antigens, which serve as important recognition targets for immunotherapy [126,128]. Thus, the combination of higher-expressed antigens and more effective antigen presentation could make tumors a better target for the immune system. Interestingly, the MAPK cascade in melanoma cells also contributes to the production of IL-1, which stimulates the release of immunosuppressive secreted factors by tumor-associate fibroblasts, thus contributing to the immunosuppressive tumor microenvironment [134]. Furthermore, it was reported that a melanoma-derived mutant RAC1 (RAC1 P29S) selectively drives the expression of PD-L1 [135], a major immune-evasive molecule [136]. However, the molecular mechanism of this phenomenon remains unexplored, and it is peculiar that the effect was limited to this, but not the other (RAC1 Q61L) constitutively active RAC1 variant [135]. Thus, it is uncertain whether PAK1 mediates the control of PD-L1 by RAC1 in that system. However, the role of PAK1 in PD-L1 control has been shown more convincingly in pancreatic cancer models, where the loss of PAK1 decreased PD-L1 expression while increasing tumor infiltration by CD4+ and CD8+ T-cells [137].
The expression of FasL by melanoma cells was proposed as one of the mechanisms that protects the tumors from the immune system by killing vulnerable T-cells [138,139]. The actual incidence of FasL expression in melanomas is a subject of a controversy [140], but it is noteworthy that FasL is also produced by the endothelial cells in the tumor [141]. Interestingly, the death of T-cells from FasL depends on the function of RAC proteins [142]. It is thus possible that the inhibition of RAC signaling would improve anti-tumor immunity and the efficacy of immune therapy, but the role of PAK1 in FasL-mediated killing is yet to be established.
Importantly, the systemic inhibition of the MAPK cascade is also known to affect the immune system directly, wherein it is expected to enhance the anti-tumor response. This is especially relevant to MEK inhibitors, which can suppress B-regulatory cells [143] and increase the number of effector-phenotype antigen-specific CD8-positive T-cells within the tumor [129]. These inhibitors also extend the life of tumor-infiltrating T-cells, while sparing their cytotoxic activity [129]. Overall, the efficacy of immune therapy in preclinical models is enhanced by a more efficient inhibition of the MAPK cascade [125,126,127,129]. It is therefore likely that further suppression of this pathway via a concomitant inhibition of PAK1 would be beneficial for immune therapy.
An obvious concern is whether some MAPK-independent functions of PAK1 are critically essential for the immune system, so that PAK1 inhibition would be immunosuppressive. The observations in the PAK1-deficient mouse model seem to dispel this concern, as the knockout animals demonstrated heightened, rather than decreased, immunity [124,137]. This supports the enthusiasm for a further investigation of PAK1 inhibition as an auxiliary for immune therapy.

6. Future Directions

While the preponderance of evidence points to PAK1 inhibition as a promising strategy to augment various types of anti-melanoma therapies, the need for a clinically usable approach to PAK1 regulation remains unmet. To date, there is a multitude of chemical agents that can inhibit this enzyme, yet none have received an approval for clinical use. The histories, chemical structures, specificities, and investigational status of these molecules have been extensively reviewed by others elsewhere [39,40,144]. The hurdles for the currently available compounds range from chemical instability and poor bioavailability to off-target effects and expected toxicity. Several directions forward can be envisioned. The development of new compounds remains a possibility. A critical issue on this path remains to be the balance between isoform specificity and anti-cancer activity. PAK1-deficient animals have a relatively mild phenotype [145], although they are prone to cardiac problems under specific types of stress [146]. Non-transformed cells have a higher tolerance to PAK1 inhibition [50], and may even benefit from it under certain conditions [147]. In contrast, PAK2 deficiency is embryonically lethal [148,149], while PAK3-deificent mice display deficits in some memory- and learning-related tasks [150]. Concededly, the phenotypes of the knockout animals may only imperfectly recapitulate the effects of systemic target inhibition in an adult organism. Nevertheless, it is reasonable to assume that a PAK1-specific inhibitor would be less prone to off-target toxicity than the one targeting all group 1 PAKs, not to mention the inhibitors with even broader specificity. Toxic side effects may be further compounded by the spurious inhibition of more distantly related enzymes. A noteworthy recent development [151] is the report of a PAK1-specific degrader molecule (BJG-05-039), which combines a highly selective, but only modestly potent, PAK1 inhibitor, NVS-PAK1-1 [152] with lenalidomide, a recruiter of the CRL4CRBN E3 ubiquitin ligase [153]. The approach results in a significant reduction of PAK1 protein levels in treated cells, and potentially provides a concurrent suppression of the enzymatic and the scaffolding functions of PAK1 [151]. The high specificity and potency of BJG-05-039 against PAK1 suggest an opportunity to avoid the off-target toxicity of less specific PAK inhibitors. On the other hand, there is some functional overlap between PAKs, which brings up the possibility that a treated cancer may evolve a resistance to complete PAK1 inhibition by elevating the activity of other isoforms [49]. Thus, a broader-specificity inhibitor might be needed for a more robust anti-cancer effect.
Conceivably, a practical resolution might emerge from the use of PAK inhibitors not as monotherapies, but in synergistic combinations with other anti-cancer treatments. In general, the search for a synergistic drug combination for melanoma treatment remains an active area of research [154]. Some examples of combinations, where the synergy of other therapeutics with PAK1 inhibitors is documented or may be predicted, are discussed above. Each component in such a combination may have some preferential toxicity to cancer cells, albeit with some dose-dependent side effects. In an ideal case, synergy would allow the combination to achieve a therapeutic effect even when the individual components are delivered at doses that are safe but ineffective upon individual administration, i.e., when the respective targets are only partially inhibited. In this scenario, even an inhibitor with otherwise non-ideal pharmacokinetics or pharmacodynamics might prove useful, as long as it potently synergizes with other components of the combination regimen.
Another possible avenue for drug improvement is the optimization of the delivery options for the existing compounds. For example, IPA3, a pioneering compound that selectively disrupts the interaction between group 1 PAKs and small GTPases [155], was considered to be sub-optimal for clinical use due to its perceived metabolically labile nature [156]. However, more recent studies suggest that at least some of its shortfalls can be overcome by formulating it in liposomes [157,158].
It is also worth mentioning that PAK1 can be regulated indirectly by preventing its activation by cancer-relevant small GTPases. Targeting a particular cancer-relevant GTPase may prevent the pathological activation of a PAK enzyme, while preserving at least some of its normal physiological functions. The inhibition of small GTPases is a burgeoning area of pharmacological research. For example, there are already a number of potent and relatively specific inhibitors of RAC1 [159], the eponymous activator of PAK1. However, the hurdles in the clinical translation of these discoveries still remain [159], and none have been approved as an anti-melanoma therapy. Previously, the research into the regulation of small GTPases was mostly focused on their interactions with guanine nucleotide exchange factors, guanine nucleotide dissociation inhibitors, and GTPase-activating proteins [160]. Recent findings suggest that the activity of RAC1 can also be manipulated by controlling the local availability of GTP [161]. The clinical effectiveness of this strategy is yet to be proven, but multiple inhibitors of GTP biosynthesis are already approved for human use.
Another approach, yet to be implemented in practice, is to exploit the higher activity of PAK1 in treatment-resistant cells as a source of a specific vulnerability. One possible line of exploration is the intracellular stress of the hyperactivation of PAK1. The hyperactivity of oncogenic pathways is often damaging to a cell, making it exceptionally dependent on cytoprotective stress-response pathways and sensitive to further impacts that elevate the same type of damage. Paradoxically, this scenario applies even to oncogenes that are otherwise known to protect cancer cells from common therapies, as exemplified by AKT [116,162]. Another way of exploiting PAK1 activation may be to rely on its effect on DNA damage response. While its ability to impair damage sensing mechanisms may increase cell tolerance to individual genotoxic compounds [73], the failure of cell cycle checkpoints may also sensitize a cell to conditions which a normal cell can survive by arresting its cell cycle progression. Indeed, such strategies to exploit cell cycle checkpoint deficiencies, albeit not in a PAK1-dependent manner, have been proposed for melanoma already [163,164].
Overall, the available information on the functions of PAK1, the experimental data from preclinical melanoma models, and the evidence from similarly treated non-melanoma cancers all point to PAK1 as a modulator of melanoma’s response to multiple types of anti-cancer therapy. The co-targeting of PAK1 in the context of other melanoma treatments appears as a worthwhile direction for future exploration, with a potential to tangibly improve the management of this deadly malignancy.

Author Contributions

All authors contributed to the conceptualization, data curation, writing, review, and editing of this manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

The work at Eugene Kandel’s laboratory is supported by the Roswell Park Alliance Foundation. The research activities at the Roswell Park Comprehensive Cancer Center are supported in part by the NCI grant P30CA16056.

Data Availability Statement

The data presented in this study are available on request from the corresponding author.

Acknowledgments

The authors would like to thank the current and past members of the Department of Cell Stress Biology at the Roswell Park Comprehensive Cancer Center for many helpful discussions on the topic of this review.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Siegel, R.L.; Miller, K.D.; Wagle, N.S.; Jemal, A. Cancer statistics, 2023. CA Cancer J. Clin. 2023, 73, 17–48. [Google Scholar] [CrossRef] [PubMed]
  2. Kaliki, S.; Shields, C.L. Uveal melanoma: Relatively rare but deadly cancer. Eye 2017, 31, 241–257. [Google Scholar] [CrossRef] [PubMed]
  3. Kujala, E.; Makitie, T.; Kivela, T. Very long-term prognosis of patients with malignant uveal melanoma. Investig. Ophthalmol. Vis. Sci. 2003, 44, 4651–4659. [Google Scholar] [CrossRef] [PubMed]
  4. Johnson, D.B.; Daniels, A.B. Continued Poor Survival in Metastatic Uveal Melanoma: Implications for Molecular Prognostication, Surveillance Imaging, Adjuvant Therapy, and Clinical Trials. JAMA Ophthalmol. 2018, 136, 986–988. [Google Scholar] [CrossRef] [PubMed]
  5. Conway, J.; Bellet, J.S.; Rubin, A.I.; Lipner, S.R. Adult and Pediatric Nail Unit Melanoma: Epidemiology, Diagnosis, and Treatment. Cells 2023, 12, 964. [Google Scholar] [CrossRef] [PubMed]
  6. Chapman, P.B.; Einhorn, L.H.; Meyers, M.L.; Saxman, S.; Destro, A.N.; Panageas, K.S.; Begg, C.B.; Agarwala, S.S.; Schuchter, L.M.; Ernstoff, M.S.; et al. Phase III multicenter randomized trial of the Dartmouth regimen versus dacarbazine in patients with metastatic melanoma. J. Clin. Oncol. 1999, 17, 2745–2751. [Google Scholar] [CrossRef] [PubMed]
  7. Avril, M.F.; Aamdal, S.; Grob, J.J.; Hauschild, A.; Mohr, P.; Bonerandi, J.J.; Weichenthal, M.; Neuber, K.; Bieber, T.; Gilde, K.; et al. Fotemustine compared with dacarbazine in patients with disseminated malignant melanoma: A phase III study. J. Clin. Oncol. 2004, 22, 1118–1125. [Google Scholar] [CrossRef] [PubMed]
  8. Brown, C.K.; Kirkwood, J.M. Medical management of melanoma. Surg. Clin. N. Am. 2003, 83, 283–322. [Google Scholar] [CrossRef]
  9. Bajetta, E.; Del Vecchio, M.; Bernard-Marty, C.; Vitali, M.; Buzzoni, R.; Rixe, O.; Nova, P.; Aglione, S.; Taillibert, S.; Khayat, D. Metastatic melanoma: Chemotherapy. Semin. Oncol. 2002, 29, 427–445. [Google Scholar] [CrossRef]
  10. Guo, W.; Wang, H.; Li, C. Signal pathways of melanoma and targeted therapy. Signal Transduct. Target. Ther. 2021, 6, 424. [Google Scholar] [CrossRef]
  11. Caksa, S.; Baqai, U.; Aplin, A.E. The future of targeted kinase inhibitors in melanoma. Pharmacol. Ther. 2022, 239, 108200. [Google Scholar] [CrossRef] [PubMed]
  12. Fernandez, M.F.; Choi, J.; Sosman, J. New Approaches to Targeted Therapy in Melanoma. Cancers 2023, 15, 3224. [Google Scholar] [CrossRef] [PubMed]
  13. Passarelli, A.; Mannavola, F.; Stucci, L.S.; Tucci, M.; Silvestris, F. Immune system and melanoma biology: A balance between immunosurveillance and immune escape. Oncotarget 2017, 8, 106132–106142. [Google Scholar] [CrossRef] [PubMed]
  14. Knight, A.; Karapetyan, L.; Kirkwood, J.M. Immunotherapy in Melanoma: Recent Advances and Future Directions. Cancers 2023, 15, 1106. [Google Scholar] [CrossRef] [PubMed]
  15. Ascierto, P.A.; Long, G.V.; Robert, C.; Brady, B.; Dutriaux, C.; Di Giacomo, A.M.; Mortier, L.; Hassel, J.C.; Rutkowski, P.; McNeil, C.; et al. Survival Outcomes in Patients With Previously Untreated BRAF Wild-Type Advanced Melanoma Treated With Nivolumab Therapy: Three-Year Follow-up of a Randomized Phase 3 Trial. JAMA Oncol. 2019, 5, 187–194. [Google Scholar] [CrossRef] [PubMed]
  16. Robert, C.; Ribas, A.; Schachter, J.; Arance, A.; Grob, J.J.; Mortier, L.; Daud, A.; Carlino, M.S.; McNeil, C.M.; Lotem, M.; et al. Pembrolizumab versus ipilimumab in advanced melanoma (KEYNOTE-006): Post-hoc 5-year results from an open-label, multicentre, randomised, controlled, phase 3 study. Lancet. Oncol. 2019, 20, 1239–1251. [Google Scholar] [CrossRef] [PubMed]
  17. Larkin, J.; Chiarion-Sileni, V.; Gonzalez, R.; Grob, J.J.; Rutkowski, P.; Lao, C.D.; Cowey, C.L.; Schadendorf, D.; Wagstaff, J.; Dummer, R.; et al. Five-Year Survival with Combined Nivolumab and Ipilimumab in Advanced Melanoma. N. Engl. J. Med. 2019, 381, 1535–1546. [Google Scholar] [CrossRef]
  18. Piulats, J.M.; Espinosa, E.; de la Cruz Merino, L.; Varela, M.; Alonso Carrion, L.; Martin-Algarra, S.; Lopez Castro, R.; Curiel, T.; Rodriguez-Abreu, D.; Redrado, M.; et al. Nivolumab Plus Ipilimumab for Treatment-Naive Metastatic Uveal Melanoma: An Open-Label, Multicenter, Phase II Trial by the Spanish Multidisciplinary Melanoma Group (GEM-1402). J. Clin. Oncol. 2021, 39, 586–598. [Google Scholar] [CrossRef]
  19. Pelster, M.S.; Gruschkus, S.K.; Bassett, R.; Gombos, D.S.; Shephard, M.; Posada, L.; Glover, M.S.; Simien, R.; Diab, A.; Hwu, P.; et al. Nivolumab and Ipilimumab in Metastatic Uveal Melanoma: Results From a Single-Arm Phase II Study. J. Clin. Oncol. 2021, 39, 599–607. [Google Scholar] [CrossRef]
  20. Nathan, P.; Hassel, J.C.; Rutkowski, P.; Baurain, J.F.; Butler, M.O.; Schlaak, M.; Sullivan, R.J.; Ochsenreither, S.; Dummer, R.; Kirkwood, J.M.; et al. Overall Survival Benefit with Tebentafusp in Metastatic Uveal Melanoma. N. Engl. J. Med. 2021, 385, 1196–1206. [Google Scholar] [CrossRef]
  21. Manser, E.; Leung, T.; Salihuddin, H.; Zhao, Z.S.; Lim, L. A brain serine/threonine protein kinase activated by Cdc42 and Rac1. Nature 1994, 367, 40–46. [Google Scholar] [CrossRef] [PubMed]
  22. Grebenova, D.; Holoubek, A.; Roselova, P.; Obr, A.; Brodska, B.; Kuzelova, K. PAK1, PAK1Delta15, and PAK2: Similarities, differences and mutual interactions. Sci. Rep. 2019, 9, 17171. [Google Scholar] [CrossRef] [PubMed]
  23. Lin, L.; Sabnis, A.J.; Chan, E.; Olivas, V.; Cade, L.; Pazarentzos, E.; Asthana, S.; Neel, D.; Yan, J.J.; Lu, X.; et al. The Hippo effector YAP promotes resistance to RAF- and MEK-targeted cancer therapies. Nat. Genet. 2015, 47, 250–256. [Google Scholar] [CrossRef] [PubMed]
  24. Liu, X.; Si, W.; Liu, X.; He, L.; Ren, J.; Yang, Z.; Yang, J.; Li, W.; Liu, S.; Pei, F.; et al. JMJD6 promotes melanoma carcinogenesis through regulation of the alternative splicing of PAK1, a key MAPK signaling component. Mol. Cancer 2017, 16, 175. [Google Scholar] [CrossRef]
  25. Chu, Y.; Elrod, N.; Wang, C.; Li, L.; Chen, T.; Routh, A.; Xia, Z.; Li, W.; Wagner, E.J.; Ji, P. Nudt21 regulates the alternative polyadenylation of Pak1 and is predictive in the prognosis of glioblastoma patients. Oncogene 2019, 38, 4154–4168. [Google Scholar] [CrossRef] [PubMed]
  26. Gartel, A.L.; Kandel, E.S. miRNAs: Little known mediators of oncogenesis. Semin. Cancer Biol. 2008, 18, 103–110. [Google Scholar] [CrossRef] [PubMed]
  27. Strange, K.; Denton, J.; Nehrke, K. Ste20-type kinases: Evolutionarily conserved regulators of ion transport and cell volume. Physiology 2006, 21, 61–68. [Google Scholar] [CrossRef]
  28. Kichina, J.V.; Goc, A.; Al-Husein, B.; Somanath, P.R.; Kandel, E.S. PAK1 as a therapeutic target. Expert Opin. Ther. Targets 2010, 14, 703–725. [Google Scholar] [CrossRef]
  29. Lei, M.; Lu, W.; Meng, W.; Parrini, M.C.; Eck, M.J.; Mayer, B.J.; Harrison, S.C. Structure of PAK1 in an autoinhibited conformation reveals a multistage activation switch. Cell 2000, 102, 387–397. [Google Scholar] [CrossRef]
  30. Strochlic, T.I.; Viaud, J.; Rennefahrt, U.E.; Anastassiadis, T.; Peterson, J.R. Phosphoinositides are essential coactivators for p21-activated kinase 1. Mol. Cell 2010, 40, 493–500. [Google Scholar] [CrossRef]
  31. Eid, S.; Turk, S.; Volkamer, A.; Rippmann, F.; Fulle, S. KinMap: A web-based tool for interactive navigation through human kinome data. BMC Bioinform. 2017, 18, 16. [Google Scholar] [CrossRef] [PubMed]
  32. Knaus, U.G.; Morris, S.; Dong, H.J.; Chernoff, J.; Bokoch, G.M. Regulation of human leukocyte p21-activated kinases through G protein—coupled receptors. Science 1995, 269, 221–223. [Google Scholar] [CrossRef] [PubMed]
  33. Pirruccello, M.; Sondermann, H.; Pelton, J.G.; Pellicena, P.; Hoelz, A.; Chernoff, J.; Wemmer, D.E.; Kuriyan, J. A dimeric kinase assembly underlying autophosphorylation in the p21 activated kinases. J. Mol. Biol. 2006, 361, 312–326. [Google Scholar] [CrossRef] [PubMed]
  34. Wang, J.; Wu, J.W.; Wang, Z.X. Structural insights into the autoactivation mechanism of p21-activated protein kinase. Structure 2011, 19, 1752–1761. [Google Scholar] [CrossRef] [PubMed]
  35. Thompson, G.; Owen, D.; Chalk, P.A.; Lowe, P.N. Delineation of the Cdc42/Rac-binding domain of p21-activated kinase. Biochemistry 1998, 37, 7885–7891. [Google Scholar] [CrossRef] [PubMed]
  36. Bokoch, G.M.; Reilly, A.M.; Daniels, R.H.; King, C.C.; Olivera, A.; Spiegel, S.; Knaus, U.G. A GTPase-independent mechanism of p21-activated kinase activation. Regulation by sphingosine and other biologically active lipids. J. Biol. Chem. 1998, 273, 8137–8144. [Google Scholar] [CrossRef] [PubMed]
  37. Kumar, R.; Sanawar, R.; Li, X.; Li, F. Structure, biochemistry, and biology of PAK kinases. Gene 2017, 605, 20–31. [Google Scholar] [CrossRef] [PubMed]
  38. Rane, C.K.; Minden, A. P21 activated kinase signaling in cancer. Semin. Cancer Biol. 2019, 54, 40–49. [Google Scholar] [CrossRef]
  39. Yao, D.; Li, C.; Rajoka, M.S.R.; He, Z.; Huang, J.; Wang, J.; Zhang, J. P21-Activated Kinase 1: Emerging biological functions and potential therapeutic targets in Cancer. Theranostics 2020, 10, 9741–9766. [Google Scholar] [CrossRef]
  40. Somanath, P.R.; Chernoff, J.; Cummings, B.S.; Prasad, S.M.; Homan, H.D. Targeting P21-Activated Kinase-1 for Metastatic Prostate Cancer. Cancers 2023, 15, 2236. [Google Scholar] [CrossRef]
  41. Babagana, M.; Johnson, S.; Slabodkin, H.; Bshara, W.; Morrison, C.; Kandel, E.S. P21-activated kinase 1 regulates resistance to BRAF inhibition in human cancer cells. Mol. Carcinog. 2017, 56, 1515–1525. [Google Scholar] [CrossRef] [PubMed]
  42. Pavey, S.; Zuidervaart, W.; van Nieuwpoort, F.; Packer, L.; Jager, M.; Gruis, N.; Hayward, N. Increased p21-activated kinase-1 expression is associated with invasive potential in uveal melanoma. Melanoma Res. 2006, 16, 285–296. [Google Scholar] [CrossRef] [PubMed]
  43. Ong, C.C.; Jubb, A.M.; Jakubiak, D.; Zhou, W.; Rudolph, J.; Haverty, P.M.; Kowanetz, M.; Yan, Y.; Tremayne, J.; Lisle, R.; et al. P21-activated kinase 1 (PAK1) as a therapeutic target in BRAF wild-type melanoma. J. Natl. Cancer Inst. 2013, 105, 606–607. [Google Scholar] [CrossRef] [PubMed]
  44. Wang, M.; Banik, I.; Shain, A.H.; Yeh, I.; Bastian, B.C. Integrated genomic analyses of acral and mucosal melanomas nominate novel driver genes. Genome Med. 2022, 14, 65. [Google Scholar] [CrossRef] [PubMed]
  45. Newell, F.; Wilmott, J.S.; Johansson, P.A.; Nones, K.; Addala, V.; Mukhopadhyay, P.; Broit, N.; Amato, C.M.; Van Gulick, R.; Kazakoff, S.H.; et al. Whole-genome sequencing of acral melanoma reveals genomic complexity and diversity. Nat. Commun. 2020, 11, 5259. [Google Scholar] [CrossRef] [PubMed]
  46. Gumaste, P.V.; Fleming, N.H.; Silva, I.; Shapiro, R.L.; Berman, R.S.; Zhong, J.; Osman, I.; Stein, J.A. Analysis of recurrence patterns in acral versus nonacral melanoma: Should histologic subtype influence treatment guidelines? J. Natl. Compr. Cancer Netw. JNCCN 2014, 12, 1706–1712. [Google Scholar] [CrossRef] [PubMed]
  47. Hodis, E.; Watson, I.R.; Kryukov, G.V.; Arold, S.T.; Imielinski, M.; Theurillat, J.P.; Nickerson, E.; Auclair, D.; Li, L.; Place, C.; et al. A landscape of driver mutations in melanoma. Cell 2012, 150, 251–263. [Google Scholar] [CrossRef] [PubMed]
  48. Krauthammer, M.; Kong, Y.; Ha, B.H.; Evans, P.; Bacchiocchi, A.; McCusker, J.P.; Cheng, E.; Davis, M.J.; Goh, G.; Choi, M.; et al. Exome sequencing identifies recurrent somatic RAC1 mutations in melanoma. Nat. Genet. 2012, 44, 1006–1014. [Google Scholar] [CrossRef]
  49. Chow, H.Y.; Jubb, A.M.; Koch, J.N.; Jaffer, Z.M.; Stepanova, D.; Campbell, D.A.; Duron, S.G.; O’Farrell, M.; Cai, K.Q.; Klein-Szanto, A.J.; et al. p21-Activated kinase 1 is required for efficient tumor formation and progression in a Ras-mediated skin cancer model. Cancer Res. 2012, 72, 5966–5975. [Google Scholar] [CrossRef]
  50. Nheu, T.; He, H.; Hirokawa, Y.; Walker, F.; Wood, J.; Maruta, H. PAK is essential for RAS-induced upregulation of cyclin D1 during the G1 to S transition. Cell Cycle 2004, 3, 71–74. [Google Scholar] [CrossRef]
  51. Tang, Y.; Yu, J.; Field, J. Signals from the Ras, Rac, and Rho GTPases converge on the Pak protein kinase in Rat-1 fibroblasts. Mol. Cell. Biol. 1999, 19, 1881–1891. [Google Scholar] [CrossRef] [PubMed]
  52. Qiu, R.G.; Chen, J.; Kirn, D.; McCormick, F.; Symons, M. An essential role for Rac in Ras transformation. Nature 1995, 374, 457–459. [Google Scholar] [CrossRef] [PubMed]
  53. Vanni, I.; Tanda, E.T.; Dalmasso, B.; Pastorino, L.; Andreotti, V.; Bruno, W.; Boutros, A.; Spagnolo, F.; Ghiorzo, P. Non-BRAF Mutant Melanoma: Molecular Features and Therapeutical Implications. Front. Mol. Biosci. 2020, 7, 172. [Google Scholar] [CrossRef] [PubMed]
  54. Berger, M.F.; Hodis, E.; Heffernan, T.P.; Deribe, Y.L.; Lawrence, M.S.; Protopopov, A.; Ivanova, E.; Watson, I.R.; Nickerson, E.; Ghosh, P.; et al. Melanoma genome sequencing reveals frequent PREX2 mutations. Nature 2012, 485, 502–506. [Google Scholar] [CrossRef] [PubMed]
  55. Lissanu Deribe, Y.; Shi, Y.; Rai, K.; Nezi, L.; Amin, S.B.; Wu, C.C.; Akdemir, K.C.; Mahdavi, M.; Peng, Q.; Chang, Q.E.; et al. Truncating PREX2 mutations activate its GEF activity and alter gene expression regulation in NRAS-mutant melanoma. Proc. Natl. Acad. Sci. USA 2016, 113, E1296–E1305. [Google Scholar] [CrossRef] [PubMed]
  56. Curtin, J.A.; Busam, K.; Pinkel, D.; Bastian, B.C. Somatic activation of KIT in distinct subtypes of melanoma. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2006, 24, 4340–4346. [Google Scholar] [CrossRef] [PubMed]
  57. Beadling, C.; Jacobson-Dunlop, E.; Hodi, F.S.; Le, C.; Warrick, A.; Patterson, J.; Town, A.; Harlow, A.; Cruz, F., 3rd; Azar, S.; et al. KIT gene mutations and copy number in melanoma subtypes. Clin. Cancer Res. 2008, 14, 6821–6828. [Google Scholar] [CrossRef]
  58. McDaniel, A.S.; Allen, J.D.; Park, S.J.; Jaffer, Z.M.; Michels, E.G.; Burgin, S.J.; Chen, S.; Bessler, W.K.; Hofmann, C.; Ingram, D.A.; et al. Pak1 regulates multiple c-Kit mediated Ras-MAPK gain-in-function phenotypes in Nf1+/- mast cells. Blood 2008, 112, 4646–4654. [Google Scholar] [CrossRef]
  59. Lodde, G.C.; Jansen, P.; Herbst, R.; Terheyden, P.; Utikal, J.; Pfohler, C.; Ulrich, J.; Kreuter, A.; Mohr, P.; Gutzmer, R.; et al. Characterisation and outcome of RAC1 mutated melanoma. Eur. J. Cancer 2023, 183, 1–10. [Google Scholar] [CrossRef]
  60. Vaque, J.P.; Dorsam, R.T.; Feng, X.; Iglesias-Bartolome, R.; Forsthoefel, D.J.; Chen, Q.; Debant, A.; Seeger, M.A.; Ksander, B.R.; Teramoto, H.; et al. A genome-wide RNAi screen reveals a Trio-regulated Rho GTPase circuitry transducing mitogenic signals initiated by G protein-coupled receptors. Mol. Cell 2013, 49, 94–108. [Google Scholar] [CrossRef]
  61. Moore, A.R.; Ran, L.; Guan, Y.; Sher, J.J.; Hitchman, T.D.; Zhang, J.Q.; Hwang, C.; Walzak, E.G.; Shoushtari, A.N.; Monette, S.; et al. GNA11 Q209L Mouse Model Reveals RasGRP3 as an Essential Signaling Node in Uveal Melanoma. Cell Rep. 2018, 22, 2455–2468. [Google Scholar] [CrossRef] [PubMed]
  62. McCarty, S.K.; Saji, M.; Zhang, X.; Knippler, C.M.; Kirschner, L.S.; Fernandez, S.; Ringel, M.D. BRAF activates and physically interacts with PAK to regulate cell motility. Endocr.-Relat. Cancer 2014, 21, 865–877. [Google Scholar] [CrossRef] [PubMed]
  63. Serrone, L.; Zeuli, M.; Sega, F.M.; Cognetti, F. Dacarbazine-based chemotherapy for metastatic melanoma: Thirty-year experience overview. J. Exp. Clin. Cancer Res. CR 2000, 19, 21–34. [Google Scholar] [PubMed]
  64. Melia, B.M.; Abramson, D.H.; Albert, D.M.; Boldt, H.C.; Earle, J.D.; Hanson, W.F.; Montague, P.; Moy, C.S.; Schachat, A.P.; Simpson, E.R.; et al. Collaborative ocular melanoma study (COMS) randomized trial of I-125 brachytherapy for medium choroidal melanoma. I. Visual acuity after 3 years COMS report no. 16. Ophthalmology 2001, 108, 348–366. [Google Scholar] [CrossRef] [PubMed]
  65. Hendrickx, A.; Cozzio, A.; Plasswilm, L.; Panje, C.M. Radiotherapy for lentigo maligna and lentigo maligna melanoma—A systematic review. Radiat. Oncol. 2020, 15, 174. [Google Scholar] [CrossRef] [PubMed]
  66. Spencer, K.R.; Mehnert, J.M. Mucosal Melanoma: Epidemiology, Biology and Treatment. Cancer Treat. Res. 2016, 167, 295–320. [Google Scholar] [CrossRef] [PubMed]
  67. Henderson, M.A.; Burmeister, B.H.; Ainslie, J.; Fisher, R.; Di Iulio, J.; Smithers, B.M.; Hong, A.; Shannon, K.; Scolyer, R.A.; Carruthers, S.; et al. Adjuvant lymph-node field radiotherapy versus observation only in patients with melanoma at high risk of further lymph-node field relapse after lymphadenectomy (ANZMTG 01.02/TROG 02.01): 6-year follow-up of a phase 3, randomised controlled trial. Lancet Oncol. 2015, 16, 1049–1060. [Google Scholar] [CrossRef] [PubMed]
  68. Fogarty, G.B.; Hong, A. Radiation therapy for advanced and metastatic melanoma. J. Surg. Oncol. 2014, 109, 370–375. [Google Scholar] [CrossRef]
  69. Zhou, Y.J.; Tang, Y.; Liu, S.J.; Zeng, P.H.; Qu, L.; Jing, Q.C.; Yin, W.J. Radiation-induced liver disease: Beyond DNA damage. Cell Cycle 2023, 22, 506–526. [Google Scholar] [CrossRef] [PubMed]
  70. Roig, J.; Traugh, J.A. p21-activated protein kinase gamma-PAK is activated by ionizing radiation and other DNA-damaging agents. Similarities and differences to alpha-PAK. J. Biol. Chem. 1999, 274, 31119–31122. [Google Scholar] [CrossRef]
  71. Perez-Yepez, E.A.; Saldivar-Ceron, H.I.; Villamar-Cruz, O.; Perez-Plasencia, C.; Arias-Romero, L.E. p21 Activated kinase 1: Nuclear activity and its role during DNA damage repair. DNA Repair 2018, 65, 42–46. [Google Scholar] [CrossRef]
  72. Motwani, M.; Li, D.Q.; Horvath, A.; Kumar, R. Identification of novel gene targets and functions of p21-activated kinase 1 during DNA damage by gene expression profiling. PLoS ONE 2013, 8, e66585. [Google Scholar] [CrossRef]
  73. Ho, H.; Aruri, J.; Kapadia, R.; Mehr, H.; White, M.A.; Ganesan, A.K. RhoJ regulates melanoma chemoresistance by suppressing pathways that sense DNA damage. Cancer Res. 2012, 72, 5516–5528. [Google Scholar] [CrossRef] [PubMed]
  74. Vadlamudi, R.K.; Bagheri-Yarmand, R.; Yang, Z.; Balasenthil, S.; Nguyen, D.; Sahin, A.A.; den Hollander, P.; Kumar, R. Dynein light chain 1, a p21-activated kinase 1-interacting substrate, promotes cancerous phenotypes. Cancer Cell 2004, 5, 575–585. [Google Scholar] [CrossRef]
  75. Schurmann, A.; Mooney, A.F.; Sanders, L.C.; Sells, M.A.; Wang, H.G.; Reed, J.C.; Bokoch, G.M. p21-activated kinase 1 phosphorylates the death agonist bad and protects cells from apoptosis. Mol. Cell. Biol. 2000, 20, 453–461. [Google Scholar] [CrossRef] [PubMed]
  76. Jin, S.; Zhuo, Y.; Guo, W.; Field, J. p21-activated Kinase 1 (Pak1)-dependent phosphorylation of Raf-1 regulates its mitochondrial localization, phosphorylation of BAD, and Bcl-2 association. J. Biol. Chem. 2005, 280, 24698–24705. [Google Scholar] [CrossRef] [PubMed]
  77. Mazumdar, A.; Kumar, R. Estrogen regulation of Pak1 and FKHR pathways in breast cancer cells. FEBS Lett. 2003, 535, 6–10. [Google Scholar] [CrossRef]
  78. Frost, J.A.; Swantek, J.L.; Stippec, S.; Yin, M.J.; Gaynor, R.; Cobb, M.H. Stimulation of NFkappa B activity by multiple signaling pathways requires PAK1. J. Biol. Chem. 2000, 275, 19693–19699. [Google Scholar] [CrossRef]
  79. Kandel, E.S.; Lu, T.; Wan, Y.; Agarwal, M.K.; Jackson, M.W.; Stark, G.R. Mutagenesis by reversible promoter insertion to study the activation of NF-kappaB. Proc. Natl. Acad. Sci. USA 2005, 102, 6425–6430. [Google Scholar] [CrossRef]
  80. Huang, C.; Radi, R.H.; Arbiser, J.L. Mitochondrial Metabolism in Melanoma. Cells 2021, 10, 3197. [Google Scholar] [CrossRef]
  81. Dillon, M.; Lopez, A.; Lin, E.; Sales, D.; Perets, R.; Jain, P. Progress on Ras/MAPK Signaling Research and Targeting in Blood and Solid Cancers. Cancers 2021, 13, 5059. [Google Scholar] [CrossRef] [PubMed]
  82. Beeser, A.; Jaffer, Z.M.; Hofmann, C.; Chernoff, J. Role of group A p21-activated kinases in activation of extracellular-regulated kinase by growth factors. J. Biol. Chem. 2005, 280, 36609–36615. [Google Scholar] [CrossRef] [PubMed]
  83. Frost, J.A.; Xu, S.; Hutchison, M.R.; Marcus, S.; Cobb, M.H. Actions of Rho family small G proteins and p21-activated protein kinases on mitogen-activated protein kinase family members. Mol. Cell. Biol. 1996, 16, 3707–3713. [Google Scholar] [CrossRef] [PubMed]
  84. Tran, N.H.; Wu, X.; Frost, J.A. B-Raf and Raf-1 are regulated by distinct autoregulatory mechanisms. J. Biol. Chem. 2005, 280, 16244–16253. [Google Scholar] [CrossRef] [PubMed]
  85. Eblen, S.T.; Slack, J.K.; Weber, M.J.; Catling, A.D. Rac-PAK signaling stimulates extracellular signal-regulated kinase (ERK) activation by regulating formation of MEK1-ERK complexes. Mol. Cell. Biol. 2002, 22, 6023–6033. [Google Scholar] [CrossRef] [PubMed]
  86. Broit, N.; Johansson, P.A.; Rodgers, C.B.; Walpole, S.T.; Newell, F.; Hayward, N.K.; Pritchard, A.L. Meta-Analysis and Systematic Review of the Genomics of Mucosal Melanoma. Mol. Cancer Res. 2021, 19, 991–1004. [Google Scholar] [CrossRef] [PubMed]
  87. Wróblewska, J.P.; Dias-Santagata, D.; Ustaszewski, A.; Wu, C.L.; Fujimoto, M.; Selim, M.A.; Biernat, W.; Ryś, J.; Marszalek, A.; Hoang, M.P. Prognostic Roles of BRAF, KIT, NRAS, IGF2R and SF3B1 Mutations in Mucosal Melanomas. Cells 2021, 10, 2216. [Google Scholar] [CrossRef] [PubMed]
  88. Dummer, R.; Flaherty, K.T.; Robert, C.; Arance, A.; de Groot, J.W.B.; Garbe, C.; Gogas, H.J.; Gutzmer, R.; Krajsova, I.; Liszkay, G.; et al. COLUMBUS 5-Year Update: A Randomized, Open-Label, Phase III Trial of Encorafenib Plus Binimetinib Versus Vemurafenib or Encorafenib in Patients With BRAF V600-Mutant Melanoma. J. Clin. Oncol. 2022, 40, 4178–4188. [Google Scholar] [CrossRef]
  89. Ascierto, P.A.; Dreno, B.; Larkin, J.; Ribas, A.; Liszkay, G.; Maio, M.; Mandala, M.; Demidov, L.; Stroyakovskiy, D.; Thomas, L.; et al. 5-Year Outcomes with Cobimetinib plus Vemurafenib in BRAFV600 Mutation-Positive Advanced Melanoma: Extended Follow-up of the coBRIM Study. Clin. Cancer Res. 2021, 27, 5225–5235. [Google Scholar] [CrossRef]
  90. Robert, C.; Grob, J.J.; Stroyakovskiy, D.; Karaszewska, B.; Hauschild, A.; Levchenko, E.; Chiarion Sileni, V.; Schachter, J.; Garbe, C.; Bondarenko, I.; et al. Five-Year Outcomes with Dabrafenib plus Trametinib in Metastatic Melanoma. N. Engl. J. Med. 2019, 381, 626–636. [Google Scholar] [CrossRef]
  91. Lebbe, C.; Dutriaux, C.; Lesimple, T.; Kruit, W.; Kerger, J.; Thomas, L.; Guillot, B.; Braud, F.; Garbe, C.; Grob, J.J.; et al. Pimasertib Versus Dacarbazine in Patients With Unresectable NRAS-Mutated Cutaneous Melanoma: Phase II, Randomized, Controlled Trial with Crossover. Cancers 2020, 12, 1727. [Google Scholar] [CrossRef] [PubMed]
  92. Dummer, R.; Schadendorf, D.; Ascierto, P.A.; Arance, A.; Dutriaux, C.; Di Giacomo, A.M.; Rutkowski, P.; Del Vecchio, M.; Gutzmer, R.; Mandala, M.; et al. Binimetinib versus dacarbazine in patients with advanced NRAS-mutant melanoma (NEMO): A multicentre, open-label, randomised, phase 3 trial. Lancet Oncol. 2017, 18, 435–445. [Google Scholar] [CrossRef] [PubMed]
  93. Carvajal, R.D.; Piperno-Neumann, S.; Kapiteijn, E.; Chapman, P.B.; Frank, S.; Joshua, A.M.; Piulats, J.M.; Wolter, P.; Cocquyt, V.; Chmielowski, B.; et al. Selumetinib in Combination With Dacarbazine in Patients With Metastatic Uveal Melanoma: A Phase III, Multicenter, Randomized Trial (SUMIT). J. Clin. Oncol. 2018, 36, 1232–1239. [Google Scholar] [CrossRef] [PubMed]
  94. Khan, S.; Patel, S.P.; Shoushtari, A.N.; Ambrosini, G.; Cremers, S.; Lee, S.; Franks, L.; Singh-Kandah, S.; Hernandez, S.; Sender, N.; et al. Intermittent MEK inhibition for the treatment of metastatic uveal melanoma. Front. Oncol. 2022, 12, 975643. [Google Scholar] [CrossRef] [PubMed]
  95. Trunzer, K.; Pavlick, A.C.; Schuchter, L.; Gonzalez, R.; McArthur, G.A.; Hutson, T.E.; Moschos, S.J.; Flaherty, K.T.; Kim, K.B.; Weber, J.S.; et al. Pharmacodynamic effects and mechanisms of resistance to vemurafenib in patients with metastatic melanoma. J. Clin. Oncol. 2013, 31, 1767–1774. [Google Scholar] [CrossRef] [PubMed]
  96. Nazarian, R.; Shi, H.; Wang, Q.; Kong, X.; Koya, R.C.; Lee, H.; Chen, Z.; Lee, M.K.; Attar, N.; Sazegar, H.; et al. Melanomas acquire resistance to B-RAF(V600E) inhibition by RTK or N-RAS upregulation. Nature 2010, 468, 973–977. [Google Scholar] [CrossRef] [PubMed]
  97. Perna, D.; Karreth, F.A.; Rust, A.G.; Perez-Mancera, P.A.; Rashid, M.; Iorio, F.; Alifrangis, C.; Arends, M.J.; Bosenberg, M.W.; Bollag, G.; et al. BRAF inhibitor resistance mediated by the AKT pathway in an oncogenic BRAF mouse melanoma model. Proc. Natl. Acad. Sci. USA 2015, 112, E536–E545. [Google Scholar] [CrossRef]
  98. Shao, Y.; Aplin, A.E. Akt3-mediated resistance to apoptosis in B-RAF-targeted melanoma cells. Cancer Res. 2010, 70, 6670–6681. [Google Scholar] [CrossRef]
  99. Kandel, E.S.; Hay, N. The regulation and activities of the multifunctional serine/threonine kinase Akt/PKB. Exp. Cell Res. 1999, 253, 210–229. [Google Scholar] [CrossRef]
  100. Gottlob, K.; Majewski, N.; Kennedy, S.; Kandel, E.; Robey, R.B.; Hay, N. Inhibition of early apoptotic events by Akt/PKB is dependent on the first committed step of glycolysis and mitochondrial hexokinase. Genes Dev. 2001, 15, 1406–1418. [Google Scholar] [CrossRef]
  101. Kandel, E.S.; Skeen, J.; Majewski, N.; Di Cristofano, A.; Pandolfi, P.P.; Feliciano, C.S.; Gartel, A.; Hay, N. Activation of Akt/protein kinase B overcomes a G(2)/m cell cycle checkpoint induced by DNA damage. Mol. Cell. Biol. 2002, 22, 7831–7841. [Google Scholar] [CrossRef] [PubMed]
  102. Kennedy, S.G.; Kandel, E.S.; Cross, T.K.; Hay, N. Akt/Protein kinase B inhibits cell death by preventing the release of cytochrome c from mitochondria. Mol. Cell. Biol. 1999, 19, 5800–5810. [Google Scholar] [CrossRef] [PubMed]
  103. Zuo, Q.; Liu, J.; Huang, L.; Qin, Y.; Hawley, T.; Seo, C.; Merlino, G.; Yu, Y. AXL/AKT axis mediated-resistance to BRAF inhibitor depends on PTEN status in melanoma. Oncogene 2018, 37, 3275–3289. [Google Scholar] [CrossRef] [PubMed]
  104. Babagana, M.; Kichina, J.V.; Slabodkin, H.; Johnson, S.; Maslov, A.; Brown, L.; Attwood, K.; Nikiforov, M.A.; Kandel, E.S. The role of polo-like kinase 3 in the response of BRAF-mutant cells to targeted anticancer therapies. Mol. Carcinog. 2020, 59, 5–14. [Google Scholar] [CrossRef] [PubMed]
  105. Elion, E.A. Pheromone response, mating and cell biology. Curr. Opin. Microbiol. 2000, 3, 573–581. [Google Scholar] [CrossRef] [PubMed]
  106. Somanath, P.R.; Vijai, J.; Kichina, J.V.; Byzova, T.; Kandel, E.S. The role of PAK-1 in activation of MAP kinase cascade and oncogenic transformation by Akt. Oncogene 2009, 28, 2365–2369. [Google Scholar] [CrossRef] [PubMed]
  107. Tang, Y.; Zhou, H.; Chen, A.; Pittman, R.N.; Field, J. The Akt proto-oncogene links Ras to Pak and cell survival signals. J. Biol. Chem. 2000, 275, 9106–9109. [Google Scholar] [CrossRef]
  108. Singhal, R.; Kandel, E.S. The response to PAK1 inhibitor IPA3 distinguishes between cancer cells with mutations in BRAF and Ras oncogenes. Oncotarget 2012, 3, 700–708. [Google Scholar] [CrossRef]
  109. Lu, H.; Liu, S.; Zhang, G.; Bin, W.; Zhu, Y.; Frederick, D.T.; Hu, Y.; Zhong, W.; Randell, S.; Sadek, N.; et al. PAK signalling drives acquired drug resistance to MAPK inhibitors in BRAF-mutant melanomas. Nature 2017, 550, 133–136. [Google Scholar] [CrossRef]
  110. Watson, I.R.; Li, L.; Cabeceiras, P.K.; Mahdavi, M.; Gutschner, T.; Genovese, G.; Wang, G.; Fang, Z.; Tepper, J.M.; Stemke-Hale, K.; et al. The RAC1 P29S hotspot mutation in melanoma confers resistance to pharmacological inhibition of RAF. Cancer Res. 2014, 74, 4845–4852. [Google Scholar] [CrossRef]
  111. Ruiz, R.; Jahid, S.; Harris, M.; Marzese, D.M.; Espitia, F.; Vasudeva, P.; Chen, C.F.; de Feraudy, S.; Wu, J.; Gillen, D.L.; et al. The RhoJ-BAD signaling network: An Achilles’ heel for BRAF mutant melanomas. PLoS Genet. 2017, 13, e1006913. [Google Scholar] [CrossRef] [PubMed]
  112. Araiza-Olivera, D.; Feng, Y.; Semenova, G.; Prudnikova, T.Y.; Rhodes, J.; Chernoff, J. Suppression of RAC1-driven malignant melanoma by group A PAK inhibitors. Oncogene 2018, 37, 944–952. [Google Scholar] [CrossRef] [PubMed]
  113. Stejerean-Todoran, I.; Gimotty, P.A.; Watters, A.; Brafford, P.; Krepler, C.; Godok, T.; Li, H.; Bonilla Del Rio, Z.; Zieseniss, A.; Katschinski, D.M.; et al. A distinct pattern of growth and RAC1 signaling in melanoma brain metastasis cells. Neuro-Oncology 2023, 25, 674–686. [Google Scholar] [CrossRef] [PubMed]
  114. Smalley, K.S.; Weber, J.S. Taming the wild-types: Targeting PAK1 in melanomas that lack BRAF mutations. J. Natl. Cancer Inst. 2013, 105, 591–592. [Google Scholar] [CrossRef]
  115. Verbik, D.J.; Murray, T.G.; Tran, J.M.; Ksander, B.R. Melanomas that develop within the eye inhibit lymphocyte proliferation. Int. J. Cancer 1997, 73, 470–478. [Google Scholar] [CrossRef]
  116. Babagana, M.; Brown, L.R.; Slabodkin, H.Z.; Kichina, J.V.; Kandel, E.S. Proteotoxic Stress as an Exploitable Vulnerability in Cells with Hyperactive AKT. Int. J. Mol. Sci. 2021, 22, 11367. [Google Scholar] [CrossRef] [PubMed]
  117. Ianevski, A.; Giri, A.K.; Aittokallio, T. SynergyFinder 3.0: An interactive analysis and consensus interpretation of multi-drug synergies across multiple samples. Nucleic Acids Res. 2022, 50, W739–W743. [Google Scholar] [CrossRef]
  118. Kissil, J.L.; Johnson, K.C.; Eckman, M.S.; Jacks, T. Merlin phosphorylation by p21-activated kinase 2 and effects of phosphorylation on merlin localization. J. Biol. Chem. 2002, 277, 10394–10399. [Google Scholar] [CrossRef]
  119. Kissil, J.L.; Wilker, E.W.; Johnson, K.C.; Eckman, M.S.; Yaffe, M.B.; Jacks, T. Merlin, the product of the Nf2 tumor suppressor gene, is an inhibitor of the p21-activated kinase, Pak1. Mol. Cell 2003, 12, 841–849. [Google Scholar] [CrossRef]
  120. Xiao, G.H.; Beeser, A.; Chernoff, J.; Testa, J.R. p21-activated kinase links Rac/Cdc42 signaling to merlin. J. Biol. Chem. 2002, 277, 883–886. [Google Scholar] [CrossRef]
  121. Sabra, H.; Brunner, M.; Mandati, V.; Wehrle-Haller, B.; Lallemand, D.; Ribba, A.S.; Chevalier, G.; Guardiola, P.; Block, M.R.; Bouvard, D. beta1 integrin-dependent Rac/group I PAK signaling mediates YAP activation of Yes-associated protein 1 (YAP1) via NF2/merlin. J. Biol. Chem. 2017, 292, 19179–19197. [Google Scholar] [CrossRef] [PubMed]
  122. Chocarro, L.; Bocanegra, A.; Blanco, E.; Fernández-Rubio, L.; Arasanz, H.; Echaide, M.; Garnica, M.; Ramos, P.; Piñeiro-Hermida, S.; Vera, R.; et al. Cutting-Edge: Preclinical and Clinical Development of the First Approved Lag-3 Inhibitor. Cells 2022, 11, 2351. [Google Scholar] [CrossRef]
  123. Damato, B.E.; Dukes, J.; Goodall, H.; Carvajal, R.D. Tebentafusp: T Cell Redirection for the Treatment of Metastatic Uveal Melanoma. Cancers 2019, 11, 971. [Google Scholar] [CrossRef] [PubMed]
  124. Huynh, N.; Wang, K.; Yim, M.; Dumesny, C.J.; Sandrin, M.S.; Baldwin, G.S.; Nikfarjam, M.; He, H. Depletion of p21-activated kinase 1 up-regulates the immune system of APC(∆14/+) mice and inhibits intestinal tumorigenesis. BMC Cancer 2017, 17, 431. [Google Scholar] [CrossRef]
  125. Mimura, K.; Kua, L.F.; Shiraishi, K.; Kee Siang, L.; Shabbir, A.; Komachi, M.; Suzuki, Y.; Nakano, T.; Yong, W.P.; So, J.; et al. Inhibition of mitogen-activated protein kinase pathway can induce upregulation of human leukocyte antigen class I without PD-L1-upregulation in contrast to interferon-gamma treatment. Cancer Sci. 2014, 105, 1236–1244. [Google Scholar] [CrossRef] [PubMed]
  126. Frederick, D.T.; Piris, A.; Cogdill, A.P.; Cooper, Z.A.; Lezcano, C.; Ferrone, C.R.; Mitra, D.; Boni, A.; Newton, L.P.; Liu, C.; et al. BRAF inhibition is associated with enhanced melanoma antigen expression and a more favorable tumor microenvironment in patients with metastatic melanoma. Clin. Cancer Res. 2013, 19, 1225–1231. [Google Scholar] [CrossRef] [PubMed]
  127. Sapkota, B.; Hill, C.E.; Pollack, B.P. Vemurafenib enhances MHC induction in BRAF(V600E) homozygous melanoma cells. Oncoimmunology 2013, 2, e22890. [Google Scholar] [CrossRef]
  128. Boni, A.; Cogdill, A.P.; Dang, P.; Udayakumar, D.; Njauw, C.N.; Sloss, C.M.; Ferrone, C.R.; Flaherty, K.T.; Lawrence, D.P.; Fisher, D.E.; et al. Selective BRAFV600E inhibition enhances T-cell recognition of melanoma without affecting lymphocyte function. Cancer Res. 2010, 70, 5213–5219. [Google Scholar] [CrossRef]
  129. Ebert, P.J.R.; Cheung, J.; Yang, Y.; McNamara, E.; Hong, R.; Moskalenko, M.; Gould, S.E.; Maecker, H.; Irving, B.A.; Kim, J.M.; et al. MAP Kinase Inhibition Promotes T Cell and Anti-tumor Activity in Combination with PD-L1 Checkpoint Blockade. Immunity 2016, 44, 609–621. [Google Scholar] [CrossRef]
  130. Wang, Y.; Liu, S.; Yang, Z.; Algazi, A.P.; Lomeli, S.H.; Wang, Y.; Othus, M.; Hong, A.; Wang, X.; Randolph, C.E.; et al. Anti-PD-1/L1 lead-in before MAPK inhibitor combination maximizes antitumor immunity and efficacy. Cancer Cell 2021, 39, 1375–1387.e1376. [Google Scholar] [CrossRef]
  131. Phadke, M.S.; Chen, Z.; Li, J.; Mohamed, E.; Davies, M.A.; Smalley, I.; Duckett, D.R.; Palve, V.; Czerniecki, B.J.; Forsyth, P.A.; et al. Targeted Therapy Given after Anti-PD-1 Leads to Prolonged Responses in Mouse Melanoma Models through Sustained Antitumor Immunity. Cancer Immunol. Res. 2021, 9, 554–567. [Google Scholar] [CrossRef] [PubMed]
  132. Sumimoto, H.; Imabayashi, F.; Iwata, T.; Kawakami, Y. The BRAF-MAPK signaling pathway is essential for cancer-immune evasion in human melanoma cells. J. Exp. Med. 2006, 203, 1651–1656. [Google Scholar] [CrossRef]
  133. Kennedy, S.A.; Jarboui, M.A.; Srihari, S.; Raso, C.; Bryan, K.; Dernayka, L.; Charitou, T.; Bernal-Llinares, M.; Herrera-Montavez, C.; Krstic, A.; et al. Extensive rewiring of the EGFR network in colorectal cancer cells expressing transforming levels of KRAS(G13D). Nat. Commun. 2020, 11, 499. [Google Scholar] [CrossRef] [PubMed]
  134. Khalili, J.S.; Liu, S.; Rodriguez-Cruz, T.G.; Whittington, M.; Wardell, S.; Liu, C.; Zhang, M.; Cooper, Z.A.; Frederick, D.T.; Li, Y.; et al. Oncogenic BRAF(V600E) promotes stromal cell-mediated immunosuppression via induction of interleukin-1 in melanoma. Clin. Cancer Res. 2012, 18, 5329–5340. [Google Scholar] [CrossRef]
  135. Vu, H.L.; Rosenbaum, S.; Purwin, T.J.; Davies, M.A.; Aplin, A.E. RAC1 P29S regulates PD-L1 expression in melanoma. Pigment Cell Melanoma Res. 2015, 28, 590–598. [Google Scholar] [CrossRef] [PubMed]
  136. Tang, Q.; Chen, Y.; Li, X.; Long, S.; Shi, Y.; Yu, Y.; Wu, W.; Han, L.; Wang, S. The role of PD-1/PD-L1 and application of immune-checkpoint inhibitors in human cancers. Front. Immunol. 2022, 13, 964442. [Google Scholar] [CrossRef] [PubMed]
  137. Wang, K.; Zhan, Y.; Huynh, N.; Dumesny, C.; Wang, X.; Asadi, K.; Herrmann, D.; Timpson, P.; Yang, Y.; Walsh, K.; et al. Inhibition of PAK1 suppresses pancreatic cancer by stimulation of anti-tumour immunity through down-regulation of PD-L1. Cancer Lett. 2020, 472, 8–18. [Google Scholar] [CrossRef]
  138. Hahne, M.; Rimoldi, D.; Schroter, M.; Romero, P.; Schreier, M.; French, L.E.; Schneider, P.; Bornand, T.; Fontana, A.; Lienard, D.; et al. Melanoma cell expression of Fas(Apo-1/CD95) ligand: Implications for tumor immune escape. Science 1996, 274, 1363–1366. [Google Scholar] [CrossRef]
  139. Andreola, G.; Rivoltini, L.; Castelli, C.; Huber, V.; Perego, P.; Deho, P.; Squarcina, P.; Accornero, P.; Lozupone, F.; Lugini, L.; et al. Induction of lymphocyte apoptosis by tumor cell secretion of FasL-bearing microvesicles. J. Exp. Med. 2002, 195, 1303–1316. [Google Scholar] [CrossRef]
  140. Chappell, D.B.; Zaks, T.Z.; Rosenberg, S.A.; Restifo, N.P. Human melanoma cells do not express Fas (Apo-1/CD95) ligand. Cancer Res. 1999, 59, 59–62. [Google Scholar]
  141. Motz, G.T.; Santoro, S.P.; Wang, L.P.; Garrabrant, T.; Lastra, R.R.; Hagemann, I.S.; Lal, P.; Feldman, M.D.; Benencia, F.; Coukos, G. Tumor endothelium FasL establishes a selective immune barrier promoting tolerance in tumors. Nat. Med. 2014, 20, 607–615. [Google Scholar] [CrossRef] [PubMed]
  142. Ramaswamy, M.; Dumont, C.; Cruz, A.C.; Muppidi, J.R.; Gomez, T.S.; Billadeau, D.D.; Tybulewicz, V.L.; Siegel, R.M. Cutting edge: Rac GTPases sensitize activated T cells to die via Fas. J. Immunol. 2007, 179, 6384–6388. [Google Scholar] [CrossRef] [PubMed]
  143. Yarchoan, M.; Mohan, A.A.; Dennison, L.; Vithayathil, T.; Ruggieri, A.; Lesinski, G.B.; Armstrong, T.D.; Azad, N.S.; Jaffee, E.M. MEK inhibition suppresses B regulatory cells and augments anti-tumor immunity. PLoS ONE 2019, 14, e0224600. [Google Scholar] [CrossRef] [PubMed]
  144. Semenova, G.; Chernoff, J. Targeting PAK1. Biochem. Soc. Trans. 2017, 45, 79–88. [Google Scholar] [CrossRef] [PubMed]
  145. Allen, J.D.; Jaffer, Z.M.; Park, S.J.; Burgin, S.; Hofmann, C.; Sells, M.A.; Chen, S.; Derr-Yellin, E.; Michels, E.G.; McDaniel, A.; et al. p21-activated kinase regulates mast cell degranulation via effects on calcium mobilization and cytoskeletal dynamics. Blood 2009, 113, 2695–2705. [Google Scholar] [CrossRef] [PubMed]
  146. Ke, Y.; Lei, M.; Wang, X.; Solaro, R.J. Novel roles of PAK1 in the heart. Cell. Logist. 2012, 2, 89–94. [Google Scholar] [CrossRef] [PubMed]
  147. Zynda, E.R.; Maloy, M.H.; Kandel, E.S. The role of PAK1 in the sensitivity of kidney epithelial cells to ischemia-like conditions. Cell Cycle 2019, 18, 596–604. [Google Scholar] [CrossRef]
  148. Marlin, J.W.; Chang, Y.W.; Ober, M.; Handy, A.; Xu, W.; Jakobi, R. Functional PAK-2 knockout and replacement with a caspase cleavage-deficient mutant in mice reveals differential requirements of full-length PAK-2 and caspase-activated PAK-2p34. Mamm. Genome 2011, 22, 306–317. [Google Scholar] [CrossRef]
  149. Hofmann, C.; Shepelev, M.; Chernoff, J. The genetics of Pak. J. Cell Sci. 2004, 117, 4343–4354. [Google Scholar] [CrossRef]
  150. Meng, J.; Meng, Y.; Hanna, A.; Janus, C.; Jia, Z. Abnormal long-lasting synaptic plasticity and cognition in mice lacking the mental retardation gene Pak3. J. Neurosci. 2005, 25, 6641–6650. [Google Scholar] [CrossRef]
  151. Chow, H.Y.; Karchugina, S.; Groendyke, B.J.; Toenjes, S.; Hatcher, J.; Donovan, K.A.; Fischer, E.S.; Abalakov, G.; Faezov, B.; Dunbrack, R.; et al. Development and Utility of a PAK1-Selective Degrader. J. Med. Chem. 2022, 65, 15627–15641. [Google Scholar] [CrossRef] [PubMed]
  152. Karpov, A.S.; Amiri, P.; Bellamacina, C.; Bellance, M.H.; Breitenstein, W.; Daniel, D.; Denay, R.; Fabbro, D.; Fernandez, C.; Galuba, I.; et al. Optimization of a Dibenzodiazepine Hit to a Potent and Selective Allosteric PAK1 Inhibitor. ACS Med. Chem. Lett. 2015, 6, 776–781. [Google Scholar] [CrossRef] [PubMed]
  153. Bricelj, A.; Steinebach, C.; Kuchta, R.; Gütschow, M.; Sosič, I. E3 Ligase Ligands in Successful PROTACs: An Overview of Syntheses and Linker Attachment Points. Front. Chem. 2021, 9, 707317. [Google Scholar] [CrossRef] [PubMed]
  154. Thompson, Z.J.; Teer, J.K.; Li, J.; Chen, Z.; Welsh, E.A.; Zhang, Y.; Ayoubi, N.; Eroglu, Z.; Tan, A.C.; Smalley, K.S.M.; et al. Drepmel-A Multi-Omics Melanoma Drug Repurposing Resource for Prioritizing Drug Combinations and Understanding Tumor Microenvironment. Cells 2022, 11, 2894. [Google Scholar] [CrossRef] [PubMed]
  155. Deacon, S.W.; Beeser, A.; Fukui, J.A.; Rennefahrt, U.E.; Myers, C.; Chernoff, J.; Peterson, J.R. An isoform-selective, small-molecule inhibitor targets the autoregulatory mechanism of p21-activated kinase. Chem. Biol. 2008, 15, 322–331. [Google Scholar] [CrossRef] [PubMed]
  156. Rudolph, J.; Crawford, J.J.; Hoeflich, K.P.; Wang, W. Inhibitors of p21-activated kinases (PAKs). J. Med. Chem. 2015, 58, 111–129. [Google Scholar] [CrossRef] [PubMed]
  157. Al-Azayzih, A.; Missaoui, W.N.; Cummings, B.S.; Somanath, P.R. Liposome-mediated delivery of the p21 activated kinase-1 (PAK-1) inhibitor IPA-3 limits prostate tumor growth in vivo. Nanomed. Nanotechnol. Biol. Med. 2016, 12, 1231–1239. [Google Scholar] [CrossRef] [PubMed]
  158. Najahi-Missaoui, W.; Quach, N.D.; Somanath, P.R.; Cummings, B.S. Liposomes Targeting P21 Activated Kinase-1 (PAK-1) and Selective for Secretory Phospholipase A(2) (sPLA(2)) Decrease Cell Viability and Induce Apoptosis in Metastatic Triple-Negative Breast Cancer Cells. Int. J. Mol. Sci. 2020, 21, 9396. [Google Scholar] [CrossRef]
  159. Bailly, C.; Beignet, J.; Loirand, G.; Sauzeau, V. Rac1 as a therapeutic anticancer target: Promises and limitations. Biochem. Pharmacol. 2022, 203, 115180. [Google Scholar] [CrossRef]
  160. Mosaddeghzadeh, N.; Ahmadian, M.R. The RHO Family GTPases: Mechanisms of Regulation and Signaling. Cells 2021, 10, 1831. [Google Scholar] [CrossRef]
  161. Bianchi-Smiraglia, A.; Wolff, D.W.; Marston, D.J.; Deng, Z.; Han, Z.; Moparthy, S.; Wombacher, R.M.; Mussell, A.L.; Shen, S.; Chen, J.; et al. Regulation of local GTP availability controls RAC1 activity and cell invasion. Nat. Commun. 2021, 12, 6091. [Google Scholar] [CrossRef]
  162. Nogueira, V.; Park, Y.; Chen, C.C.; Xu, P.Z.; Chen, M.L.; Tonic, I.; Unterman, T.; Hay, N. Akt determines replicative senescence and oxidative or oncogenic premature senescence and sensitizes cells to oxidative apoptosis. Cancer Cell 2008, 14, 458–470. [Google Scholar] [CrossRef]
  163. Barnaba, N.; LaRocque, J.R. Targeting cell cycle regulation via the G2-M checkpoint for synthetic lethality in melanoma. Cell Cycle 2021, 20, 1041–1051. [Google Scholar] [CrossRef]
  164. Maresca, L.; Stecca, B.; Carrassa, L. Novel Therapeutic Approaches with DNA Damage Response Inhibitors for Melanoma Treatment. Cells 2022, 11, 1466. [Google Scholar] [CrossRef]
Figure 1. Proposed mechanisms linking PAK1 activity and melanoma resistance to various therapeutic modalities. See text for details.
Figure 1. Proposed mechanisms linking PAK1 activity and melanoma resistance to various therapeutic modalities. See text for details.
Cells 12 02373 g001
Figure 3. Examples of synergistic suppression of uveal melanoma cells via combinations of PAK inhibitors and a MEK inhibitor, selumetinib. Mel202 uveal melanoma cells [115] (a gift from Dr. Harbour, University of Texas Southwestern) were exposed to a range of doses of selumetinib in combination with various doses of PAK inhibitors PF3758309 (A) or IPA3 (B). After 5 days of treatment, the numbers of remaining cells were compared using methylene blue staining and an extraction method as described earlier [116], and drug synergy according to the Bliss synergy model was analyzed using SynegyFinder software (version #: 6.04.2023-R-3.6.3-dev) [117].
Figure 3. Examples of synergistic suppression of uveal melanoma cells via combinations of PAK inhibitors and a MEK inhibitor, selumetinib. Mel202 uveal melanoma cells [115] (a gift from Dr. Harbour, University of Texas Southwestern) were exposed to a range of doses of selumetinib in combination with various doses of PAK inhibitors PF3758309 (A) or IPA3 (B). After 5 days of treatment, the numbers of remaining cells were compared using methylene blue staining and an extraction method as described earlier [116], and drug synergy according to the Bliss synergy model was analyzed using SynegyFinder software (version #: 6.04.2023-R-3.6.3-dev) [117].
Cells 12 02373 g003
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kichina, J.V.; Maslov, A.; Kandel, E.S. PAK1 and Therapy Resistance in Melanoma. Cells 2023, 12, 2373. https://doi.org/10.3390/cells12192373

AMA Style

Kichina JV, Maslov A, Kandel ES. PAK1 and Therapy Resistance in Melanoma. Cells. 2023; 12(19):2373. https://doi.org/10.3390/cells12192373

Chicago/Turabian Style

Kichina, Julia V., Alexei Maslov, and Eugene S. Kandel. 2023. "PAK1 and Therapy Resistance in Melanoma" Cells 12, no. 19: 2373. https://doi.org/10.3390/cells12192373

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop