Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Jan 1.
Published in final edited form as: Pigment Cell Melanoma Res. 2020 Apr 24;34(1):44–58. doi: 10.1111/pcmr.12880

Emerging strategies to treat rare and intractable subtypes of melanoma

Gretchen M Alicea 1, Vito W Rebecca 1
PMCID: PMC7544642  NIHMSID: NIHMS1610868  PMID: 32274887

Abstract

Melanoma is the deadliest form of skin cancer, possessing a diverse landscape of subtypes with distinct molecular signatures and levels of aggressiveness. Although immense progress has been achieved therapeutically for patients with the most common forms of this disease, little is known of how to effectively treat patients with rarer subtypes of melanoma. These subtypes include acral lentiginous (the rarest form of cutaneous melanoma; AL), uveal, and mucosal melanomas, which display variations in distribution across (a) the world, (b) patient age-groups, and (c) anatomic sites. Unfortunately, patients with these relatively rare subtypes of melanoma typically respond worse to therapies approved for the more common, non-AL cutaneous melanoma and do not have effective alternatives, and thus consequently have worse overall survival rates. Achieving durable therapeutic responses in these high-risk melanoma subtypes represents one of the greatest challenges of the field. This review aims to collate and highlight effective preclinical and/or clinical strategies against these rare forms of melanoma.

Keywords: acral lentiginous melanoma, mucosal melanoma, therapy resistance, uveal melanoma

1 |. INTRODUCTION

The melanoma field represents a paradigm for preclinical and clinical advancements in targeted and immune therapy modalities, with 13 new FDA-approved therapies since 2011 (Dummer et al., 2018a, 2018b; Long et al., 2018; Ribas & Flaherty, 2011). The catalyst for the development of targeted therapy modalities was the identification of activating NRAS mutations and BRAF mutations in 1984 (Albino, Le Strange, Oliff, Furth, & Old, 1984; Padua, Barrass, & Currie, 1984) and 2002 (Davies et al., 2002), respectively, which paved the way for molecular stratification of the melanoma patient population. Approximately 45%–50% of non-acral lentiginous (AL) cutaneous melanoma patients have tumors that harbor activating BRAF mutations (Schadendorf et al., 2017), with a single amino acid substitution of valine for glutamic acid at codon 600 (V600E) occurring in 90% of cases (Figure 1a). Activating NRAS mutations at codon 12, 13, or 61 are detectable in 15%–20% of non-AL cutaneous melanoma patients and serve as an independent predictor of worse patient overall survival (Jakob et al., 2012). Mutations of BRAF and NRAS are considered mutually exclusive (Hodis et al., 2012); however, there are rare reports where both mutations exist in different regions of the same tumor or at different metastatic sites of the same patient (Wilmott et al., 2012). To date, it remains unclear whether the same melanoma cell can harbor both a BRAF and an NRAS mutation, or at the single-cell level, these mutations are indeed mutually exclusive.

FIGURE 1.

FIGURE 1

The frequency of activating BRAF, NRAS, and GNAQ/GNA11 mutations in (a) SSM, (b) acral lentiginous, (c) mucosal, (d) uveal, (e) nodular, and (f) lentigo maligna melanoma

With discoveries revealing that ~70% of non-AL cutaneous melanomas contain mutations constitutively activating the mitogen-activated protein kinase (MAPK) pathway came intense development of inhibitors capable of targeting various nodes of the mitogen-activated protein kinase (MAPK) pathway (i.e., BRAF, MEK, and ERK inhibitors) that continues to date (English & Cobb, 2002). The first targeted therapy approved for the treatment of patients with BRAFV600E/K mutant melanoma was the small molecule inhibitor vemurafenib, an agent designed to have high specificity against the mutant V600E, V600K, V600D, and V600R forms of BRAF (Kim et al., 2014). Vemurafenib had response rates of ~48% in phase II and III clinical trials leading to the 2011 Food Drug and Agriculture (FDA) approval. A few years later, the combination of a BRAF inhibitor and a MEK inhibitor was observed to further increase the response rate to ~76% leading to the 2014 FDA approval of dabrafenib and trametinib (Paraiso et al., 2010; Queirolo, Picasso, & Spagnolo, 2015). There are now three BRAF inhibitor plus MEK inhibitor combinations FDA approved for melanoma patients with BRAFV600E/K mutations (dabrafenib/trametinib, vemurafenib/cobimetinib (Larkin et al., 2014; Ribas et al., 2014), encorafenib/binimetinib (Dummer et al., 2018a, 2018b).

For patients with wild-type BRAF, treatment with BRAF inhibitors that specifically target V600E/K mutant BRAF may increase melanoma aggressiveness due to the paradoxical activation of wild-type BRAF and downstream MAPK pathway signaling (Carnahan et al., 2010; Hatzivassiliou et al., 2010; Heidorn et al., 2010; Joseph et al., 2010; Poulikakos, Zhang, Bollag, Shokat, & Rosen, 2010). Preclinically, targeting downstream of BRAF with MEK inhibitors in BRAF-wild-type melanoma cells demonstrates the importance of the MAPK pathway for their survival, with significant anticancer activity (Rebecca, Alicea, et al., 2014). However, clinical trials testing multiple MEK inhibitors (i.e., binimetinib, trametinib) have concluded that although encouraging response rates and small increases in progression-free survival could be achieved in certain trials relative to dacarbazine, no significant increase in overall survival of patients with BRAF-wild-type melanoma was achieved with MEK inhibition (van Herpen et al., 2019). In an effort to increase MEK inhibitor efficacy, combination strategies with other agents (i.e., PI3K inhibitors (Greger et al., 2012), CDK4/6 inhibitors (Kwong et al., 2012)) are being clinically tested in the BRAF-wild-type (i.e., patients with or without NRAS-MT melanoma) setting after failure of immunotherapy (Dummer et al., 2017). ERK inhibitors are also being clinically investigated to see if durable efficacy can be achieved in patients with wild-type BRAF, with reports showing the first-in-class ERK1/2 inhibitor ulixertinib has an acceptable safety profile and early evidence of clinical activity (Moschos et al., 2018; Sullivan et al., 2018). Preclinical evidence suggests that concurrent inhibition of multiple nodes of the MAPK pathway in NRAS-mutant melanoma (i.e., MEK and ERK) may have synergistic activity on par with the BRAF inhibitor and MEK inhibitor combination in BRAF-mutant melanomas, and further studies evaluating this strategy are under way (Rebecca, Alicea, et al., 2014).

In parallel, large strides have been made in the development of immune checkpoint blockade strategies with the FDA approval of antibodies targeting cytotoxic T-lymphocyte antigen 4 (CTLA4, ipilimumab) in 2011 (Ledford, 2011) and programmed cell death 1 (PD1, pembrolizumab, nivolumab) in 2014 (Mahoney, Freeman, & McDermott, 2015; Schachter et al., 2017) and the combination of ipilimumab and nivolumab in 2015 (Camacho, 2015). Immune checkpoint blockade describes the use of therapeutic antibodies that overcome immunosuppressive checkpoints with the goal of unchaining antitumor immune responses (Kalbasi & Ribas, 2020). CTLA4 and PD-1 are both receptors that suppress effector T-cell activity (Blank et al., 2004; Chen et al., 1992; Dong et al., 2002). These immunotherapy-based strategies elicit long-lasting responses in a subset of patients and represent a therapeutic strategy suitable for all genotypes of non-AL cutaneous melanoma. However, the majority of patients treated with immunotherapy progress within 5 years due to poorly understood primary resistance mechanisms, and clinicians still cannot reliably discriminate which patients will respond or not respond. Both tumor intrinsic (i.e., insufficient tumor antigenicity, tumor interferon-γ signaling, tumor stemness) and extrinsic (i.e., regulatory T cells, myeloid-derived suppressor cells) resistance mechanisms have been reported, and there are intense efforts focused on overcoming these therapeutic hurdles to further increase the efficacy of immune checkpoint blockade strategies.

The promising efficacy of these new therapeutic strategies has been demonstrated largely in non-AL cutaneous melanoma patients with either superficial spreading melanoma (SSM), nodular melanoma (NM), or lentigo maligna melanoma (LMM). SSM, NM, and LMM represent the most common forms of melanoma in Caucasians (>85% of cases). It is important to appreciate that most of the recent pivotal discoveries in melanoma were performed on SSM cell lines, short-term cultures, animal models, and tumor biopsies taken from patients with SSM largely due to their greater availability (personal communication from Dr. Meenhard Herlyn). AL melanoma represents the fourth and rarest subtype of cutaneous melanoma. In addition, mucosal melanoma and uveal melanoma are other rare subtypes of melanoma that are non-cutaneous in origin. The efficacy of immune checkpoint blockade is lower in rarer subtypes of melanoma relative to patients with non-AL cutaneous melanoma, which will be discussed later. There is also little information regarding the efficacy of combination BRAF inhibitor and MEK inhibitor therapy in these subtypes. In the remainder of the review, we will flesh out details regarding mutational landscapes, etiology, and efficacy of targeted and immune therapy options for patients with rare subtypes of melanoma and draw attention to developments in each respective case in the way of emerging therapeutic strategies with promise to increase the overall survival of patients with intractable rare subtypes of melanoma.

2 |. ACRAL LENTIGINOUS: THE MOST UNCOMMON OF THE 4 MA JOR CMM SUBTYPES IN CAUCASIANS

2.1 |. Incidence/Presentation

Acral lentiginous melanoma is an uncommon yet relatively aggressive subtype of CMM that accounts for 2%–3% of all melanoma cases. AL melanoma arises on sun-protected, glabrous skin of the soles, palms, and nail beds. AL melanoma has been historically associated with worse 10-year survival rates relative to other forms of CMM (67.5% vs. 87.5%; Bradford, Goldstein, McMaster, & Tucker, 2009). Further, 10-year AL melanoma survival rates are highest in non-Hispanic Whites (69.4%), intermediate in Blacks (71.5%), and lowest in Hispanic Whites (57.3%) and Asian/Pacific Islanders (54.1%), as found by the Surveillance, Epidemiology, and End Results (SEER) Program of the National Cancer Institute evaluating data from 17 population-based cancer registries from 1986 to 2005 (Bradford et al., 2009). Another analysis of AL melanoma prognostic features in a cohort of German, Swiss, and Austrian patients suggests no significant difference exist relative to other subtypes of cutaneous melanoma; however, this conclusion may stem due to differential ethnicity landscapes between this patient cohort and that in the SEER study (Teramoto et al., 2018). There does not appear to be a gender bias, with a similar frequency between men and women and a comparable median age of diagnosis of 63.1 years for men and 62.2 years for women. The incidence of AL melanoma increases with age, and for reasons poorly understood, men are twice as likely to develop AL melanoma relative to women after the age of 80 (American Cancer Society, 2019).

The distribution of AL melanoma varies geographically among populations throughout the world (Figure 2). While AL melanoma represents only ~2%–3% of all melanoma cases in Caucasian populations, AL melanoma makes up 50%–80% of all cases in non-Caucasian individuals in the United States (i.e., those of African, Latin American, and Asian descent) (Cascinelli et al., 1994; Chang et al., 2004; Nakamura & Fujisawa, 2018; Wang, Zhao, & Ma, 2016). Furthermore, the incidence in Hispanic Whites doubles compared to non-Hispanic Whites after the aged of 70 (Bradford et al., 2009). A 2009 SEER study found the overall incidence rates of AL melanoma were similar between non-Hispanic Whites and Blacks; however, Hispanic Whites have statistically higher incidence rates relative to non-Hispanic Whites (Bradford et al., 2009). Updated epidemiological studies should be performed to continue understanding the differential incidence trends that may exist across different ethnicities. Of note, the incidence of other subtypes of cutaneous melanoma (i.e., NM, SSM) is much lower in non-Caucasians relative to Caucasians. As this subtype of melanoma is not related to ultraviolet radiation (UV), there are different theories of the cause of AL melanoma. Some reports state that trauma and pressure in the foot (a predilected area of AL) is causal (Costello, Pittelkow, & Mangold, 2017). However, the hand is also exposed to trauma but its location is less favorable. The main sites of AL melanoma metastases are the lungs, distant lymph nodes, scalp, contralateral limb, and liver (Kwon et al., 2019).

FIGURE 2.

FIGURE 2

A schematic depicting the common sites of superficial spreading, nodular, lentigo maligna, acral lentiginous, mucosal, and uveal melanomas

2.2 |. Mutational/molecular profile

Acral lentiginous melanomas possess a significantly lower mutational burden relative to the more common cutaneous melanoma subtypes, likely due to the sun-protected locations they arise from (Furney et al., 2014). BRAF mutations in are found in 1 in every 5 Al melanoma patients, leaving ~80% ineligible to receive BRAF inhibitor and combination BRAF/MEK inhibitor strategies (Curtin et al., 2005; Figure 1b). Therefore, new targets specific for AL melanoma are needed. 80% of AL melanomas display genetic aberrations of cyclin-dependent kinase 4/6 (CDK4/6) pathway-related genes (i.e., amplification of CDK4 and CCND1, and/or loss of CDK2NA), representing the most frequent copy number alteration detected (Kong et al., 2017). Additionally, activating KIT mutations are present in ~6% of cases (Zaremba et al., 2019). AL melanoma displays similar incidence of NRAS mutations as non-AL cutaneous melanoma, detectable in 15%–28% of AL melanoma patients, and NRAS mutations are an independent prognostic factor of worse overall survival (Kong et al., 2017; Mandala, Merelli, & Massi, 2014; Zaremba et al., 2019).

2.3 |. Treatment/Efficacy

Considerable barriers exist to treat patients with AL melanoma: (a) a contrasting genomic and genetic landscape relative to non-AL cutaneous melanomas, (b) unclear targetable drivers, and (3) sparse experimental models available for preclinical drug development (Turajlic et al., 2012). Unfortunately, FDA-approved targeted therapy strategies for melanoma are not available for the majority of AL melanoma patients (i.e., BRAF inhibitors since AL melanoma has a low frequency of BRAF mutations), and the efficacy of immune checkpoint blockade strategies is not well known in AL melanoma, with differing overall response rates (ORR) differing by country. For example, the ORR of anti-PD-1 in AL melanoma patients was found to be similar to that in non-AL cutaneous melanoma patients within the United States (Shoushtari et al., 2016). In contrast, the ORR was 66.7% for SSM patients and 28.6% of AL melanoma patients in a recent Japanese study, suggesting the efficacy of immune checkpoint blockade may vary with ethnicity (Yamazaki et al., 2019). The lower mutational burden observed in AL melanoma cases is thought to drive the reduced efficacy of immune checkpoint inhibitor strategies (e.g., PD-1 blockade) in patients (Furney et al., 2014). Although AL melanoma patients with Kit mutations can be treated with a KIT inhibitor per National Comprehensive Cancer Network (NCCN) guidelines, resistance mechanisms that reactivate downstream MAPK and PI3K pathway signaling have been suggested to blunt long-term durability (Table 1; Carlino, Todd, & Rizos, 2014). Due to the high percentage of AL melanoma tumors with CDK4/6-pathway aberrations, CDK4/6 inhibition represents one of the most promising targeted therapy strategies for AL melanomas clinically (NCT03454919). However, durable responses are not observed in all patients due to resistance and CDK4/6 inhibitor-based combinations will likely be needed to improve the curative rate for patients with AL melanoma (Table 1). Preclinical investigation to optimize targeted therapy strategies has not been extensively performed in AL melanoma models, but the rich body of literature that exists from studies in non-AL cutaneous melanoma models strongly suggests that single-agent approaches will not be durable due to the nearly universal onset of resistance (Rebecca, Alicea, et al., 2014; Rebecca, Wood, et al., 2014). In SSM models, treatment with a MAPK pathway inhibitor plus a CDK4/6 inhibitor has shown synergistic activity in BRAF-MT (Martin et al., 2018) and BRAF-wild-type settings (Teh et al., 2018); however, residual disease persists. Resistance mechanisms to CDK4/6 inhibitors and/or MEK inhibitors must be delineated to develop combination strategies that produce durable responses in AL melanoma patients.

TABLE 1.

Current clinical trials tailored for patients with acral lentiginous, mucosal, or uveal melanoma

Subtype Agent(s) Agent target(s) ClinicalTrials.gov identifier Phase
Nodular RO4929097 Gamma secretase/Notch signaling pathway NCT01120275 II
Nodular Dinaciclib CDK NCT00937937 II
Nodular RSV-TK, ganciclovir Adenovirus, antiviral NCT00005057 I
Lentigo maligna of head and neck Imiquimod cream + surgery TLR7 NCT01720407 III
Lentigo maligna of the face Picato gel Protein kinase C NCT02723721 II
Lentigo maligna 5-Aminolevulinic acid nanoemulsion Light sensitizing NCT02685592 IV
Acral SHR-1210, apatinib Anti-PD-1, VEGFR2 NCT03955354 II
Mucosal, acral, vulvovaginal Dasatinib BCR/ABL, SRC NCT00700882 II
Acral, mucosal PLX3397 CSF-1 NCT02071940 II
Mucosal, acral, CSD Nilotinib BCR/ABL NCT00788775 II
Acral, mucosal Nilotinib BCR/ABL NCT0135121 II
Acral, mucosal Nivolumab ± ipilimumab Anti-PD-1, anti-CTLA-4 NCT02978443 II
Acral, mucosal Sunitinib VEGFR, PDGFR NCT00577382 II
Acral, mucosal, CSD Imatinib ABL, c-Kit, PDGFR NCT00424515 II
Acral TQB2450 ± anlotinib Anti-PD-L1, VEGFR, FGFR, PDGFR, c-Kit NCT03991975 I, II
Acral Palbociclib CDK4, CDK6 NCT03454919 II
Mucosal of head and neck Radiation DNA NCT03138642 II
Mucosal Toripalimab, IFN a-2B Anti-PD-1 NCT03178123 II
Mucosal CM082 plus Toripalimab Multikinase inhibitor, anti-PD-1 monoclonal NCT03602547 II
Mucosal Ipilimumab, nivolumab anti-CTLA-4, anti-PD-1 NCT03241186 II
Mucosal Apatinib, SHR01210 VEGFR2, c-Kit, c-Src, anti-PD-1 NCT03986515 II
Uveal Vorinostat HDAC NCT03022565 I
Uveal SIR-Spheres® yttrium 90, ipilimumab, nivolumab Radiation, anti-CTLA-4, anti-PD-1 NCT02913417 I/II
Uveal BVD-523 ERK1/2 NCT03417739 II
Uveal Autologous dendritic cells loaded with autologous tumor RNA Tumor vaccine NCT01983748 III
Uveal Selumetinib MEK1/2 NCT02768766 I
Acral SHR-1210, apatinib Anti-PD-1, VEGFR2 NCT03955354 II
Mucosal, acral, vulvovaginal Dasatinib BCR/ABL, SRC NCT00700882 II
Acral, mucosal PLX3397 CSF-1 NCT02071940 II
Mucosal, acral, CSD Nilotinib BCR/ABL NCT00788775 II
Acral, mucosal Nilotinib BCR/ABL NCT0135121 II
Acral, mucosal Nivolumab ± ipilimumab Anti-PD-1, anti-CTLA-4 NCT02978443 II
Acral, mucosal Sunitinib VEGFR, PDGFR NCT00577382 II
Acral, mucosal, CSD Imatinib ABL, c-Kit, PDGFR NCT00424515 II
Acral TQB2450 ± anlotinib Anti-PD-L1, VEGFR, FGFR, NCT03991975 I, II
Acral Palbociclib CDK4, CDK6 NCT03454919 II
Mucosal of head and neck Radiation DNA NCT03138642 II
Mucosal Toripalimab, IFN a-2B Anti-PD-1 NCT03178123 II
Mucosal CM082 plus toripalimab Multikinase inhibitor, anti-PD-1 monoclonal NCT03602547 II
Mucosal Ipilimumab, nivolumab anti-CTLA-4, anti-PD-1 NCT03241186 II
Mucosal Apatinib, SHR01210 VEGFR2, c-Kit, c-Src, anti-PD-1 NCT03986515 II
Uveal Vorinostat HDAC NCT03022565 I
Uveal SIR-Spheres® yttrium 90, ipilimumab, nivolumab Radiation, anti-CTLA-4, anti-PD-1 NCT02913417 I/II
Uveal BVD-523 ERK1/2 NCT03417739 II
Uveal Autologous dendritic cells loaded with autologous tumor RNA Tumor vaccine NCT01983748 III
Uveal Selumetinib MEK1/2 NCT02768766 I

3 |. MUCOSAL MELANOMA: ONE OF THE RAREST FORMS OF MALIGNANT MELANOMA

3.1 |. Incidence/Presentation

Mucosal melanoma (MM) is one of the rarest types of melanoma, accounting for only 1% of all cases (Dumaz et al., 2019; McLaughlin et al., 2005), and has a significantly worse prognosis relative to the other subtypes (Dumaz et al., 2019). Distinct from cutaneous melanoma, MM arises from melanocytes located in mucosal membranes inside the body (i.e., genitourinary, anorectal, nasopharyngeal; Dumaz et al., 2019). The head and neck (55), vulva (18), and anus (24) are the most common observed sites; however, MM can also occur in the gut, lungs, and urinary track (Figure 2; Chang, Karnell, & Menck, 1998). It is rarely diagnosed at early stages due to difficult visual detection, which is much more tractable for cutaneous subtypes of melanoma. The overall median age of diagnosis is 70 years, with the exception of MMs arising in the mouth that manifest more frequently in younger patients (Tyrrell & Payne, 2018). The incidence of MM has been stable for the last few years with the exception of MM in the genital tract, which is higher in females relative to males for reasons not clearly understood (McLaughlin et al., 2005).

3.2 |. Mutational/Molecular profile

Approximately 3%–15% of MMs harbor an activating mutation in BRAF, with ~63% located on the V600 codon and 37% located on a non-V600 codon (Figure 1c; Dumaz et al., 2019). This is in contrast to non-AL cutaneous melanomas where <10% of BRAF mutations are outside of the V600 codon, and more closely resembles the high prevalence of non-V600 mutations found in 48% of lung adenocarcinomas. A closer analysis of the most common non-V600 mutations reveals (a) a difference between the frequency of mutations on D594, G469, and K601 between non-AL cutaneous melanomas and MMs, and (b) convergence in the non-V600 mutational landscape between MM and lung cancers where mutations are often associated with genotoxic agents (Dumaz et al., 2019).

In regard to NRAS mutations, approximately 12% of MMs harbor activating mutations, which is lower relative to cutaneous melanomas where NRAS mutations occur in 15%–20% of cases (Tate et al., 2019). There is also a divergence in the location of NRAS mutations between MM and cutaneous melanoma, with 54% located on codon 61 in MM versus 88% in cutaneous melanoma, and 46% located on codons 12 and 13 in MM versus 12% for cutaneous melanomas. Approximately 7%–22% of MMs have v-kit Hardy-Zuckerman 4 feline sarcoma viral oncogene homolog (KIT) somatic mutations or amplifications (Dumaz et al., 2019; Tyrrell & Payne, 2018). MMs located in the genital area appear to be driven by mutations in SF3B1 which encodes the subunit 1 of splicing factor 3b, a component of the spliceosome that processes pre-mRNA into mature transcripts (Furney et al., 2013; Oiso, Sakai, Yanagihara, Nishio, & Kawada, 2018; Quek et al., 2019). A recent study analyzing the mutational landscape of MM identified IGF2R mutations in 31.7% of MM samples relative to 6.3% of SSM cases (Iida et al., 2018). Interestingly, a lower frequency of UV-induced DNA damage, a lower number of mutations and a link to high tobacco exposure have also been identified in MM (Iida et al., 2018).

3.3 |. Treatment/Efficacy

Unfortunately, MM is typically detected at relatively more advanced states due to difficulty in early detection (Newell et al., 2019). The main treatment for MM differs slightly on where the tumor is located; however, like any other subtype of melanoma, patients are initially treated with surgical excision. MMs arising in the head and neck are treated with complete surgical excision of the tumor when the patient is in stages III and IVA. However, this is associated with a high rate of recurrence (Patel et al., 2002). MMs that have arisen in the vulvovaginal or anorectal area also receive radiation in addition to surgical tumor excision. Therapeutic efficacy may be improved in select patients when treatment is personalized by tumor mutational status. Clinical trials targeting KIT with imatinib show no clear effect in unselected metastatic melanoma patient populations (Kim et al., 2008; Ugurel et al., 2005; Wyman et al., 2006), but encouraging clinical benefit has been observed with KIT inhibition specifically in patients with melanomas harboring KIT mutations (not in patients whose melanoma harbor KIT amplification only; Table 1). Nonetheless, disease progression ultimately occurs in the majority of cases (Cho, Kim, Kwon, Kim, & Lee, 2012; Hodi et al., 2013). These data support the practice of determining KIT mutational status for MM patients to have a higher chance of receiving additional clinical benefit (Lutzky, Bauer, & Bastian, 2008). Subsequent phase II clinical trials now require a KIT alteration for enrollment. For the relatively small number of MM patients whose tumors harbor BRAF mutations (relative to the ~50% in non-AL cutaneous melanoma patients), treatment with combination BRAF inhibitor and MEK inhibitor therapy is available. However, the efficacy of targeted therapy specifically in the MM patient population is not completely understood due to the low number available for analysis.

The efficacy of immune checkpoint inhibitor therapy also remains unclear in MM patients, with conflicting evidence of whether MM patients respond as well as non-AL cutaneous melanoma patients (Table 1). In one multi-institutional analysis of clinical trials focusing on all the subtypes of metastatic melanoma, patients with MM had similar responses compared with non-AL cutaneous melanoma patients when treated with anti-PD-1 single-agent therapy, with a progression-free survival of 3.9 months (Shoushtari et al., 2016). In another pooled analysis, MM patients treated with nivolumab as monotherapy or nivolumab in combination with ipilimumab experienced reduced clinical benefit relative to non-AL cutaneous melanoma patients (D’Angelo et al., 2017). MM patients experienced 50% shorter progression-free survival (3.0 months) relative to patients with non-AL cutaneous melanoma (6.2 months) for monotherapy (nivolumab) and for nivolumab plus ipilimumab (5.9 vs. 11.7 months; D’Angelo et al., 2017). Another recent study combining axitinib (small molecule receptor tyrosine kinase inhibitor) with toripalimab (anti-PD-1) found a median progression-free survival of 7.5 months in among 29 patients with chemotherapy-naïve mucosal melanoma (Sheng et al., 2019). Although these data suggest that MM patients may not achieve as much benefit with immune checkpoint inhibitor therapy as non-AL cutaneous melanoma patients, it should be considered that in each of the pooled analyses, the number of MM cases was only 10% of patients compared to 75% from cutaneous melanoma (D’Angelo et al., 2017). Also notable, another prospective study where 44 patients with unresectable MM were treated with immune checkpoint inhibitors concluded that the site of origin for MM (i.e., vaginal, anal) may not have a significant impact on the objective response rate, which was 8.2% for ipilimumab and 35% for pembrolizumab (Moya-Plana et al., 2019). The lower mutational burden in MM relative to non-AL cutaneous melanoma may explain the decreased efficacy of immune checkpoint blockade in MM (Newell et al., 2019).

4 |. UVEAL MELANOMA: THE MOST FREQUENT FORM OF NON-CUTANEOUS MELANOMA

4.1 |. Incidence/Presentation

Uveal melanoma (UM) is the most common form of ocular melanoma, as well as the most prevalent form of non-cutaneous melanoma, accounting for 5% of all melanomas (Figure 2) (Chang et al., 1998). It most commonly arises in non-Hispanic Whites relative to other races (i.e., African and Asian Americans), with a slight predominance for men (52.3%) relative to women (47.7%; Aronow, Topham, & Singh, 2018). The incidence of UM has remained stable over the last few decades and is diagnosed in 4–5 per million individuals in the United States each year (Aronow et al., 2018; Singh, Turell, & Topham, 2011). The median age of diagnosis is 62, and the incidence of UM increases with age. Early detection of UM provides a favorable 85% survival rate; however, this survival rate significantly decreases to 15% once UM cells have disseminated. Approximately 50% of UM patients develop metastases, and among patients with metastatic disease, 90% have liver involvement and ~70% have liver-only disease (Bedikian et al., 1995). This is a distinct metastatic pattern relative to cutaneous melanoma or mucosal melanoma.

4.2 |. Mutational/Molecular profile

Unlike non-AL cutaneous melanomas, UMs have a much lower mutational burden due to the sun-protected site they arise from within the ocular cavity (Harbour, 2012). Activating mutations in BRAF or NRAS are not detected (extremely rare) in tumor cells of UM patients (Figure 1d; Rimoldi et al., 2003). In contrast, the main drivers for UM are activating mutations of guanine nucleotide-binding protein G (GNAQ/11), splicing factor 3B subunit 1 (SF3B1), eukaryotic translation initiation factor (EIF1AX), and inactivating mutations of the tumor suppressor BRCA-associated protein-1 (BAP1; Dono et al., 2014). The GNAQ/11 genes encode specific GTP binding proteins that mediate signal transduction from the inner cell surface to the MAPK pathway through activation of the protein kinase C (PKC) enzyme. GNAQ and GNA11 mutations are mutually exclusive, and thus in total are detected in 85%–94% of UM across all stages of disease (Figure 1d; Chen et al., 2014; Field & Harbour, 2014; Goldsmith & Dhanasekaran, 2007). Due to their detection in benign uveal nevi, GNAQ/11 mutations are thought to be early mutational events (Harbour, 2012).

BAP1 (located on the short arm of chromosome 3) loss-of-function mutations are posited to serve as a predisposing factor for diverse hereditary cancers including mesothelioma, cutaneous melanoma, renal cell carcinoma, and UM (Harbour et al., 2013). A recent comprehensive review identified that among 174 patients harboring germline BAP1 mutations, 130 developed tumors that were either UM (31% of cases), cutaneous melanoma (13% of cases), renal cell carcinoma (10% of cases), or MM (22% of cases; Masoomian, Shields, & Shields, 2018). In UM, loss of BAP1 returns melanoma cells to a more stem cell-like state as BAP1 is involved in melanocyte differentiation (Matatall et al., 2013). BAP1 is frequently mutated in metastasizing uveal melanomas, which supports the growing evidence that stem-like melanoma cell states drive elements of the metastatic cascade (Harbour et al., 2010; Rambow et al., 2018).

4.3 |. Treatment/Efficacy

There has been a recent decline in UM patients treated solely with surgery due to micrometastases that develop years before primary tumor detection. The current approach for treatment of metastatic UM is radiation; however, the survival rate is not significantly improved relative to what is possible from surgery (Krantz, Dave, Komatsubara, Marr, & Carvajal, 2017). There have been an array of clinical studies trying to identify efficacious therapeutic strategies for patients with metastatic UM (Table 1). UM patients that possess GNAQ or GNA11 mutations can be treated in clinical trials with targeted therapy approaches specific for the MAPK pathway (i.e., MEK inhibitor, ERK inhibitor) as these tumors display elevated MAPK activity. Preclinical studies have shown that treatment of UM with a combination of a MAPK pathway inhibitor and a PKC inhibitor may provide synergistic efficacy relative to what is achievable by either agent alone (Chen et al., 2014). Clinical trials with selumetinib, a MEK inhibitor, reported a higher progression-free survival among UM patients (15.9 vs. 7 weeks); however, no clinically meaningful increase in overall survival was observed in comparison to the chemotherapeutic temozolomide in the metastatic setting (10.8 vs. 9.4 months). Additionally, preclinical studies identified that targeting the PI3K/AKT pathway (in GNAQ and GNA11 mutant xenograft models) in combination with a MEK inhibitor may be an effective treatment strategy for patients with GNAQ or GNA11 mutations; however, clinical trials using this combination have stopped due to low response rates and high toxicity (Khalili et al., 2012; Table 1). Inhibitors against bromodomain and extraterminal (BET) proteins have had encouraging activity preclinically in UM, which could be further increased by concurrent inhibition of escape mechanisms mediated by fibroblast growth factor receptors (Chua et al., 2019). Similarly, targeting microenvironment-derived factors including HGF can also increase MEK inhibitor efficacy against UM cells, preclinically (Cheng et al., 2017). For UM with BAP1 mutations, it has been shown preclinically that treatment with a histone deacetylase (HDAC) inhibitor could be beneficial. Because BAP1 mutations are associated with loss of melanocytic differentiation, treatment with HDAC inhibitors (valproic acid) are postulated to inhibit the growth of uveal melanoma in vivo by inducing morphological differentiation (Landreville et al., 2012).

While immune checkpoint inhibitors are the standard of care for cutaneous melanoma, UM has not yet had a phase III clinical trial for immune therapy (Rossi et al., 2019). Small studies in UM patients (10 patients) treated with pembrolizumab (anti-PD-1) after treatment with ipilimumab reported a median progression-free survival of 18 weeks; ranging from 3.14 to 49.3 weeks (Kottschade et al., 2016). Of the eight evaluable patients, four rapidly progressed, one had stable disease, two had partial responses, and one had a complete response. Although this small study resulted in comparable results seen in patients with non-AL cutaneous melanoma, other studies suggest far lower response rates to single agent anti-PD-1 and combination anti-PD-1 plus anti-CTLA-4 in UM patients. An analysis of Danish UM patients observed partial responses in 7% of patients to anti-PD-1 and 21% to concurrent anti-PD-1 plus anti-CTLA-4 (Bol et al., 2019). Metastatic UM patients treated with ipilimumab from two additional clinical studies had a median overall survival of 9 months (in contrast to 19.9 months in non-AL cutaneous melanoma; Danielli et al., 2012; Luke et al., 2013; Wolchok et al., 2017). Despite the reduced efficacy of immune checkpoint blockade in UM patients, this option may represent the most effective strategy to date.

4.3.1 |. Non-acral lentiginous cutaneous melanoma

For the remainder of the review, we herein describe relatively rarer forms of cutaneous melanoma relative to SSM (Figure 1a), as the reader can find plentiful preclinical and clinical information on SSM biology in the literature (Atay et al., 2019; Garman et al., 2017; Herlyn et al., 1985; Krepler et al., 2016, 2017; Perego et al., 2018; Roesch et al., 2010; Villanueva et al., 2010, 2013).

5 |. NODULAR: THE QUICKEST GROWING OF THE MELANOMA SUBTYPES

5.1 |. Incidence/Presentation

Nodular melanoma represents the second most common subtype of melanoma, responsible for 10%–15% of total melanomas in Caucasians. NM is the melanoma subtype most associated with increased thickness at clinical presentation, which is attributed to the relatively poorer prognosis of patients with NM (Lattanzi et al., 2019). The median age of diagnosis for NM is 53 years, with thicker tumors more common in older patients. NM is more common in women than men for reasons poorly understood and commonly presents de novo on the head, neck, or trunk of patients (Figure 2; Xiong, Charifa, & Chen, 2019).

5.2 |. Mutational/Molecular profile

Activating BRAF mutations are detected in patients with NM at a slightly lower frequency relative to SSM, with 43%–47% of patients possessing mutations mostly (88% of cases) in V600E (Gorden et al., 2003; Shinozaki, Fujimoto, Morton, & Hoon, 2004; Spathis et al., 2019; Figure 1e). A recent study identified evidence that BRAFV600E expression may serve as a prognostic marker in primary NM associated with ulceration and reduced survival (Hugdahl, Kalvenes, Puntervoll, Ladstein, & Akslen, 2016). Preclinically, it was reported that hyperactivation of the downstream MAPK effector ribosomal protein S6 kinase (RSK1) is detectable in metastatic tumor tissues derived from NM to a higher extent relative to SSM (Salhi et al., 2015). Activating NRAS mutations are detected at a significantly elevated frequency in NM relative to SSM in 30%–33% vs. 19% of cases, respectively (Chiappetta et al., 2015; Heppt et al., 2017; Lattanzi et al., 2019). Interestingly, BRAF and NRAS mutations may not be as mutually exclusive in NM relative to SSM, with the identification of both mutations in the same tumor specimens when assessed by laser capture dissection followed by direct sequencing analysis of exons 11 and 15 of the BRAF gene and exons 1 and 2 of the NRAS gene (Chiappetta et al., 2015). Additional high-throughput sequencing of patient-derived samples of single nucleotide variations (SNVs) expected to impact protein coding reveals NOTCH4, RPSKA6, BCL2L12, TERT, ERBB3, ZNF560, SSPO, and SNX31 to be significantly under-mutated in NM relative to SSM (Lattanzi et al., 2019).

5.3 |. Treatment/Efficacy

An analysis of the most recent Surveillance, Epidemiology, and End Results (SEER) cohort and the New York University (NRU) cohort suggests that relative to patients with metastatic SSM treated with BRAF inhibitor (BRAFi) therapy, patients with metastatic NM may respond worse to BRAFi for reasons not completely understood, suggesting the potential existence of distinct clinical and biological properties between NM and SSM (Lattanzi et al., 2019). The observation of activated RSK1 via constitutive phosphorylation at the Ser-380 residue may explain the poorer efficacy of BRAFi and/or BRAFi/MEKi in patients with this melanoma subtype. In contrast, no significant difference in response rates and survival was detected in NM versus SSM among a cohort of 154 patients treated with either anti-CTLA-4, anti-PD-1, or the combination of both immune checkpoint inhibitor approaches. Immune checkpoint blockade may serve an ideal first-line therapy for patients with this subtype.

6 |. LENTIGO MALIGNA: “HUTCHINSON’S MELANOCYTIC FRECKLE”

6.1 |. Incidence/Presentation

Lentigo maligna (LM) is the third most common subtype of melanoma, comprising roughly 4%–15% of all melanoma cases (Kallini, Jain, & Khachemoune, 2013; Xiong et al., 2019), and its incidence has dramatically increased over the past few decades across the United States (Mirzoyev et al., 2014; Swetter, Boldrick, Jung, Egbert, & Harvell, 2005), the Netherlands (Greveling, Wakkee, Nijsten, van den Bos, & Hollestein, 2016), and other regions of the world. LM melanoma typically presents on chronically sun-damaged (CSD) skin of the head and neck (Figure 2), appearing as an irregular brown macule commonly on the head and neck in the elderly (DeWane, Kelsey, Oliviero, Rabinovitz, & Grant-Kels, 2019). In contrast to the mean age of diagnosis of SSM between 40 and 60 years, the mean age of diagnosis for LM melanoma is 66–72 years (Xiong et al., 2019). Credit is given to Sir John Hutchinson for the earliest description of LM melanoma in 1,890. LM melanoma was initially referred to as “Hutchinson’s melanocytic freckle” due to the prevailing thought that it was benign, non-infectious lesion owing to its slow growing nature (Xiong et al., 2019). Critical work by Ackerman and Silvers in the late 1970s–1980s finally led to wide acceptance of LM melanoma as a malignant disease worthy of clinical attention and intervention (Cohen, 1995; Silvers, 1976). Chronic ultraviolet radiation is the major risk factor for the development of LM melanoma, which differs from NM and SSM that are associated with intense intermittent ultraviolet radiation exposure (Elwood & Hislop, 1982; Holman & Armstrong, 1984). LM melanomas arise most frequently on the face and other sites of chronic sun damage which also differs from NM and SSM that arise most commonly on the trunk in men and legs in women (Figure 2). LM melanoma is thought to occur in older patients due to the increased lifetime sun and ultraviolet radiation exposure.

6.2 |. Mutational/Molecular profile

Lentigo maligna melanomas have a relatively high mutational burden compared to other melanoma subtypes due to chronic ultraviolet exposure (Xiong et al., 2019). The frequency of activating BRAF mutations in LM is unclear, with reports finding 16.7%–53.4% of LM patients harboring BRAF mutations (Saldanha, Potter, Daforno, & Pringle, 2006; Spathis et al., 2019; Figure 1f). The large variation may, in part, be attributed to the regional differences among tested patient tissue cohorts. In a Greek cohort, 16.7% of LM melanoma cases expressed BRAF mutations (Spathis et al., 2019) and 50% of LM cases in a Japanese cohort expressed BRAF mutations (Yamazaki et al., 2015). When BRAF mutations are present, the V600K substitution is frequently observed (~77%) relative to the V600E (~23%) as observed in SSM, in this small set of 13 LM patient tumor samples. This finding is consistent with V600K mutations arising on chronically sun-damaged skin (Connolly, Nehal, & Busam, 2015; Stadelmeyer et al., 2014). Activating NRAS mutations have been reported to occur in ~8.1%–16% of LM cases (Akslen et al., 2008; Saldanha et al., 2006).

6.3 |. Treatment/Efficacy

The treatment of choice for patients with localized LM melanoma consists of surgical excision as first line of therapy, followed by radiation therapy with fractionated superficial radiotherapy, or topical imiquimod cream as an alternative to surgery (Marsden et al., 2017; Mora, Karia, & Nguyen, 2015; Xiong et al., 2019). Once LM melanoma metastasizes to visceral organs, the five-year survival is similar to SSM (Kantor & Kantor, 2009). Interestingly, the efficacy of immune checkpoint blockade may be significantly higher in patients with LM melanoma relative to the other subtypes discussed. A study investigating the overall response rate (ORR) of anti-PD-1/PD-L1 in different subtypes of melanoma found patients with melanoma on CSD skin (including LM melanoma, desmoplastic melanoma, and subtype not-specified cases) exhibited an overall response rate of 70% (Kaunitz et al., 2017), which fits the theory that cancer cells with high mutational burdens may be more sensitive to immune checkpoint blockade due to the increased presence of immune-stimulatory neoepitopes (Morrison et al., 2018; Panda et al., 2017). Additional investigations on the efficacy of targeted and immune-based therapy are needed specifically for patients with LM melanoma to ensure the optimal treatment(s) is identified for this cohort and further improved through preclinical experimentation and clinical trials.

7 |. DISCUSSION

The last decade has ushered in an era of immense therapeutic possibility for patients with metastatic melanoma. What was once historically an intractable disease treated with ineffective surgery, radiation, and chemotherapy in the metastatic setting has now become a paradigm for the use of targeted and immune-based therapy. Unfortunately, the increased overall survival benefits of recently approved therapies for melanoma patients is experienced mostly by patients with the most prevalent subtype of non-AL cutaneous melanoma (SSM, NM, LMM). SSM, NM, and LMM are the most common subtypes among Caucasians in the United States and Europeans, however not among individuals of Latin American, Asian, or African descent. In these populations, other subtypes of melanoma are more predominant, and unfortunately, it remains unclear what therapeutic strategies may best cure these at-risk patient populations. Although cutaneous melanomas can be stratified into the SSM, NM, LMM, and ALM subgroups based on the work of Drs. Wallace Clark, Martin Mihm, Vincent McGovern, and Richard Reed (Scolyer, Long, & Thompson, 2011), there was debate on whether their exists justification for the use of this classification system, as early reports suggest no differences in mortality and metastases among the different subtypes. However, large-scale retrospective analyses that have been recently performed demonstrate potential disparities in the efficacy of BRAFi/MEKi and immune checkpoint inhibitor therapy among AL melanoma, as well as non-cutaneous melanoma subtypes (UM, MM) relative to non-AL cutaneous melanomas. It is critical the melanoma community address these therapeutic shortcomings in the near future and engineer adequate experimental models for preclinical investigation of resistance mechanisms to existing standard-of-care therapies to facilitate the development of optimized therapeutic strategies that increase the curative rates for patients with rarer forms of malignant melanoma. Small collections of models of rarer forms of melanoma (i.e., cell lines, patient-derived xenograft) do exist in laboratories across the world, and there has been renewed interest across the melanoma field to focus on these rarer subtypes. Only by providing the level of experimental fervor, clinical attention, and collaboration to ALM, MM, and UM that currently exists for SSM can we best heal patients with these subtypes.

Funding information

This study was supported by 1U54CA224070, Dr. Miriam and Sheldon G. Adelson Medical Research Foundation, Melanoma Research Foundation, PO1CA114046, and RO1 Minority Supplement CA174746-05.

Footnotes

CONFLICT OF INTEREST

The authors have no conflict of interest in relation to this work.

REFERENCES

  1. Akslen LA, Puntervoll H, Bachmann IM, Straume O, Vuhahula E, Kumar R, & Molven A (2008). Mutation analysis of the EGFR-NRAS-BRAF pathway in melanomas from black Africans and other subgroups of cutaneous melanoma. Melanoma Research, 18(1), 29–35. 10.1097/CMR.0b013e3282f32517 [DOI] [PubMed] [Google Scholar]
  2. Albino AP, Le Strange R, Oliff AI, Furth ME, & Old LJ (1984). Transforming ras genes from human melanoma: A manifestation of tumour heterogeneity? Nature, 308(5954), 69–72. 10.1038/308069a0 [DOI] [PubMed] [Google Scholar]
  3. American Cancer Society (2019). Cancer facts and figures 2018. American Cancer Society. https://www.cancer.org/research/cancer-facts-statistics/all-cancer-facts-figures/cancer-facts-figures-2018.html [Google Scholar]
  4. Aronow ME, Topham AK, & Singh AD (2018). Uveal melanoma: 5-year update on incidence, treatment, and survival (SEER 1973–2013). Ocular Oncology and Pathology, 4(3), 145–151. 10.1159/000480640 [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Atay C, Kwak T, Lavilla-Alonso S, Donthireddy L, Richards A, Moberg V, … Gabrilovich DI (2019). BRAF targeting sensitizes resistant melanoma to cytotoxic T cells. Clinical Cancer Research, 25(9), 2783–2794. 10.1158/1078-0432.Ccr-18-2725 [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Bedikian AY, Legha SS, Mavligit G, Carrasco CH, Khorana S, Plager C, … Benjamin RS (1995). Treatment of uveal melanoma metastatic to the liver: a review of the M. D. Anderson Cancer Center experience and prognostic factors. Cancer, 76(9), 1665–1670. . [DOI] [PubMed] [Google Scholar]
  7. Blank C, Brown I, Peterson AC, Spiotto M, Iwai Y, Honjo T, & Gajewski TF (2004). PD-L1/B7H-1 inhibits the effector phase of tumor rejection by T cell receptor (TCR) transgenic CD8+ T cells. Cancer Research, 64(3), 1140–1145. 10.1158/0008-5472.can-03-3259 [DOI] [PubMed] [Google Scholar]
  8. Bol KF, Ellebaek E, Hoejberg L, Bagger MM, Larsen MS, Klausen TW, … Svane IM (2019). Real-world impact of immune checkpoint inhibitors in metastatic uveal melanoma. Cancers (Basel), 11(10), 1489 10.3390/cancers11101489 [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Bradford PT, Goldstein AM, McMaster ML, & Tucker MA (2009). Acral lentiginous melanoma: Incidence and survival patterns in the United States, 1986–2005. Archives of Dermatology, 145(4), 427–434. 10.1001/archdermatol.2008.609 [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Camacho LH (2015). CTLA-4 blockade with ipilimumab: Biology, safety, efficacy, and future considerations. Cancer Medicine, 4(5), 661–672. 10.1002/cam4.371 [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Carlino MS, Todd JR, & Rizos H (2014). Resistance to c-Kit inhibitors in melanoma: Insights for future therapies. Oncoscience, 1(6), 423–426. 10.18632/oncoscience.51 [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Carnahan J, Beltran PJ, Babij C, Le Q, Rose MJ, Vonderfecht S, … Burgess TL (2010). Selective and potent Raf inhibitors paradoxically stimulate normal cell proliferation and tumor growth. Molecular Cancer Therapeutics, 9(8), 2399–2410. 10.1158/1535-7163.Mct-10-0181 [DOI] [PubMed] [Google Scholar]
  13. Cascinelli N, Zurrida S, Galimberti V, Bartoli C, Bufalino R, Del prato I, … Clemente C (1994). Acral lentiginous melanoma. A histological type without prognostic significance. The Journal of Dermatologic Surgery and Oncology, 20(12), 817–822. 10.1111/j.1524-4725.1994.tb03711.x [DOI] [PubMed] [Google Scholar]
  14. Chang AE, Karnell LH, & Menck HR (1998). The National Cancer Data Base report on cutaneous and noncutaneous melanoma: A summary of 84,836 cases from the past decade. The American College of Surgeons Commission on Cancer and the American Cancer Society. Cancer, 83(8), 1664–1678. [DOI] [PubMed] [Google Scholar]
  15. Chang J-C, Yeh K-Y, Wang C-H, Yang T-S, Chiang H-F, Wei F-C, … Yang C-H (2004). Malignant melanoma in Taiwan: A prognostic study of 181 cases. Melanoma Research, 14(6), 537–541. 10.1097/00008390-200412000-00016. [DOI] [PubMed] [Google Scholar]
  16. Chen L, Ashe S, Brady WA, Hellström I, Hellström KE, Ledbetter JA, … Linsley PS (1992). Costimulation of antitumor immunity by the B7 counterreceptor for the T lymphocyte molecules CD28 and CTLA-4. Cell, 71(7), 1093–1102. 10.1016/s0092-8674(05)80059-5 [DOI] [PubMed] [Google Scholar]
  17. Chen X, Wu Q, Tan L, Porter D, Jager MJ, Emery C, & Bastian BC (2014). Combined PKC and MEK inhibition in uveal melanoma with GNAQ and GNA11 mutations. Oncogene, 33(39), 4724–4734. 10.1038/onc.2013.418 [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Cheng H, Chua V, Liao C, Purwin TJ, Terai M, Kageyama K, … Aplin AE (2017). Co-targeting HGF/cMET signaling with MEK inhibitors in metastatic uveal melanoma. Molecular Cancer Therapeutics, 16(3), 516–528. 10.1158/1535-7163.Mct-16-0552 [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Chiappetta C, Proietti I, Soccodato V, Puggioni C, Zaralli R, Pacini L, … Di Cristofano C (2015). BRAF and NRAS mutations are heterogeneous and not mutually exclusive in nodular melanoma. Applied Immunohistochemistry & Molecular Morphology, 23(3), 172–177. 10.1097/pai.0000000000000071 [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Cho JH, Kim KM, Kwon M, Kim JH, & Lee J (2012). Nilotinib in patients with metastatic melanoma harboring KIT gene aberration. Investigational New Drugs, 30(5), 2008–2014. 10.1007/s10637-011-9763-9 [DOI] [PubMed] [Google Scholar]
  21. Chua V, Orloff M, Teh JLF, Sugase T, Liao C, Purwin TJ, … Aplin AE (2019). Stromal fibroblast growth factor 2 reduces the efficacy of bromodomain inhibitors in uveal melanoma. EMBO Molecular Medicine, 11(2). 10.15252/emmm.201809081 [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Cohen LM (1995). Lentigo maligna and lentigo maligna melanoma. Journal of the American Academy of Dermatology, 33(6), 923–936; quiz 937–940. 10.1016/0190-9622(95)90282-1. [DOI] [PubMed] [Google Scholar]
  23. Connolly KL, Nehal KS, & Busam KJ (2015). Lentigo maligna and lentigo maligna melanoma: Contemporary issues in diagnosis and management. Melanoma Management, 2(2), 171–178. 10.2217/mmt.15.3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Costello CM, Pittelkow MR, & Mangold AR (2017). Acral melanoma and mechanical stress on the plantar surface of the foot. New England Journal of Medicine, 377(4), 395–396. 10.1056/NEJMc1706162 [DOI] [PubMed] [Google Scholar]
  25. Curtin JA, Fridlyand J, Kageshita T, Patel HN, Busam KJ, Kutzner H, … Bastian BC (2005). Distinct sets of genetic alterations in melanoma. New England Journal of Medicine, 353(20), 2135–2147. 10.1056/NEJMoa050092 [DOI] [PubMed] [Google Scholar]
  26. D’Angelo SP, Larkin J, Sosman JA, Lebbé C, Brady B, Neyns B, … Wolchok JD (2017). Efficacy and safety of nivolumab alone or in combination with ipilimumab in patients with mucosal melanoma: A pooled analysis. Journal of Clinical Oncology, 35(2), 226–235. 10.1200/jco.2016.67.9258 [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Danielli R, Ridolfi R, Chiarion-Sileni V, Queirolo P, Testori A, Plummer R, … Maio M (2012). Ipilimumab in pretreated patients with metastatic uveal melanoma: Safety and clinical efficacy. Cancer Immunology, Immunotherapy, 61(1), 41–48. 10.1007/s00262-011-1089-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Davies H, Bignell GR, Cox C, Stephens P, Edkins S, Clegg S, … Futreal PA (2002). Mutations of the BRAF gene in human cancer. Nature, 417(6892), 949–954. 10.1038/nature00766 [DOI] [PubMed] [Google Scholar]
  29. DeWane ME, Kelsey A, Oliviero M, Rabinovitz H, & Grant-Kels JM (2019). Melanoma on chronically sun-damaged skin: Lentigo maligna and desmoplastic melanoma. Journal of the American Academy of Dermatology, 81(3), 823–833. 10.1016/j.jaad.2019.03.066 [DOI] [PubMed] [Google Scholar]
  30. Dong H, Strome SE, Salomao DR, Tamura H, Hirano F, Flies DB, … Chen L (2002). Tumor-associated B7–H1 promotes T-cell apoptosis: A potential mechanism of immune evasion. Nature Medicine, 8(8), 793–800. 10.1038/nm730 [DOI] [PubMed] [Google Scholar]
  31. Dono M, Angelini G, Cecconi M, Amaro A, Esposito AI, Mirisola V, … Pfeffer U (2014). Mutation frequencies of GNAQ, GNA11, BAP1, SF3B1, EIF1AX and TERT in uveal melanoma: Detection of an activating mutation in the TERT gene promoter in a single case of uveal melanoma. British Journal of Cancer, 110(4), 1058–1065. 10.1038/bjc.2013.804 [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Dumaz N, Jouenne F, Delyon J, Mourah S, Bensussan A, & Lebbe C (2019). Atypical BRAF and NRAS mutations in mucosal melanoma. Cancers (Basel), 11(8), 1133 10.3390/cancers11081133 [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Dummer R, Ascierto PA, Gogas HJ, Arance A, Mandala M, Liszkay G, … Flaherty KT (2018). Overall survival in patients with BRAF-mutant melanoma receiving encorafenib plus binimetinib versus vemurafenib or encorafenib (COLUMBUS): A multicentre, open-label, randomised, phase 3 trial. The Lancet Oncology, 19(10), 1315–1327. 10.1016/s1470-2045(18)30497-2 [DOI] [PubMed] [Google Scholar]
  34. Dummer R, Ascierto PA, Gogas HJ, Arance A, Mandala M, Liszkay G, … Flaherty KT (2018). Encorafenib plus binimetinib versus vemurafenib or encorafenib in patients with BRAF-mutant melanoma (COLUMBUS): A multicentre, open-label, randomised phase 3 trial. The Lancet Oncology, 19(5), 603–615. 10.1016/s1470-2045(18)30142-6 [DOI] [PubMed] [Google Scholar]
  35. Dummer R, Schadendorf D, Ascierto PA, Arance A, Dutriaux C, Di Giacomo AM, … Flaherty K (2017). Binimetinib versus dacarbazine in patients with advanced NRAS-mutant melanoma (NEMO): A multicentre, open-label, randomised, phase 3 trial. The Lancet Oncology, 18(4), 435–445. 10.1016/s1470-2045(17)30180-8 [DOI] [PubMed] [Google Scholar]
  36. Elwood JM, & Hislop TG (1982). Solar radiation in the etiology of cutaneous malignant melanoma in Caucasians. National Cancer Institute Monograph, 62, 167–171. [PubMed] [Google Scholar]
  37. English JM, & Cobb MH (2002). Pharmacological inhibitors of MAPK pathways. Trends in Pharmacological Sciences, 23(1), 40–45. 10.1016/S0165-6147(00)01865-4 [DOI] [PubMed] [Google Scholar]
  38. Field MG, & Harbour JW (2014). GNAQ/11 mutations in uveal melanoma: Is YAP the key to targeted therapy? Cancer Cell, 25(6), 714–715. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Furney SJ, Pedersen M, Gentien D, Dumont AG, Rapinat A, Desjardins L, … Marais R (2013). SF3B1 mutations are associated with alternative splicing in uveal melanoma. Cancer Discovery, 3(10), 1122–1129. 10.1158/2159-8290.Cd-13-0330 [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Furney SJ, Turajlic S, Stamp G, Thomas JM, Hayes A, Strauss D, … Marais R (2014). The mutational burden of acral melanoma revealed by whole-genome sequencing and comparative analysis. Pigment Cell & Melanoma Research, 27(5), 835–838. 10.1111/pcmr.12279 [DOI] [PubMed] [Google Scholar]
  41. Garman B, Anastopoulos IN, Krepler C, Brafford P, Sproesser K, Jiang Y, … Nathanson KL (2017). Genetic and genomic characterization of 462 melanoma patient-derived xenografts, tumor biopsies, and cell lines. Cell Reports, 21(7), 1936–1952. 10.1016/j.celrep.2017.10.052 [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Goldsmith ZG, & Dhanasekaran DN (2007). G protein regulation of MAPK networks. Oncogene, 26(22), 3122–3142. 10.1038/sj.onc.1210407 [DOI] [PubMed] [Google Scholar]
  43. Gorden A, Osman I, Gai W, He D, Huang W, Davidson A, … Polsky D (2003). Analysis of BRAF and N-RAS mutations in metastatic melanoma tissues. Cancer Research, 63(14), 3955–3957. [PubMed] [Google Scholar]
  44. Greger JG, Eastman SD, Zhang V, Bleam MR, Hughes AM, Smitheman KN, … Gilmer TM (2012). Combinations of BRAF, MEK, and PI3K/mTOR inhibitors overcome acquired resistance to the BRAF inhibitor GSK2118436 dabrafenib, mediated by NRAS or MEK mutations. Molecular Cancer Therapeutics, 11(4), 909–920. 10.1158/1535-7163.Mct-11-0989 [DOI] [PubMed] [Google Scholar]
  45. Greveling K, Wakkee M, Nijsten T, van den Bos RR, & Hollestein LM (2016). Epidemiology of lentigo maligna and lentigo maligna melanoma in the Netherlands, 1989–2013. The Journal of Investigative Dermatology, 136(10), 1955–1960. 10.1016/j.jid.2016.06.014 [DOI] [PubMed] [Google Scholar]
  46. Harbour JW (2012). The genetics of uveal melanoma: An emerging framework for targeted therapy. Pigment Cell & Melanoma Research, 25(2), 171–181. 10.1111/j.1755-148X.2012.00979.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Harbour JW, Onken MD, Roberson EDO, Duan S, Cao L, Worley LA, … Bowcock AM (2010). Frequent mutation of BAP1 in metastasizing uveal melanomas. Science, 330(6009), 1410–1413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Harbour JW, Roberson ED, Anbunathan H, Onken MD, Worley LA, & Bowcock AM (2013). Recurrent mutations at codon 625 of the splicing factor SF3B1 in uveal melanoma. Nature Genetics, 45(2), 133–135. 10.1038/ng.2523 [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Hatzivassiliou G, Song K, Yen I, Brandhuber BJ, Anderson DJ, Alvarado R, … Malek S (2010). RAF inhibitors prime wild-type RAF to activate the MAPK pathway and enhance growth. Nature, 464(7287), 431–435. 10.1038/nature08833 [DOI] [PubMed] [Google Scholar]
  50. Heidorn SJ, Milagre C, Whittaker S, Nourry A, Niculescu-Duvas I, Dhomen N, … Marais R (2010). Kinase-dead BRAF and oncogenic RAS cooperate to drive tumor progression through CRAF. Cell, 140(2), 209–221. 10.1016/j.cell.2009.12.040 [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Heppt MV, Siepmann T, Engel J, Schubert-Fritschle G, Eckel R, Mirlach L, … Berking C (2017). Prognostic significance of BRAF and NRAS mutations in melanoma: A German study from routine care. BMC Cancer, 17(1), 536 10.1186/s12885-017-3529-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Herlyn M, Balaban G, Bennicelli J, Guerry D, Halaban R, Herlyn D, … Koprowski H (1985). Primary melanoma cells of the vertical growth phase: Similarities to metastatic cells. Journal of the National Cancer Institute, 74(2), 283–289. [PubMed] [Google Scholar]
  53. Hodi FS, Corless CL, Giobbie-Hurder A, Fletcher JA, Zhu M, Marino-Enriquez A, … Fisher DE (2013). Imatinib for melanomas harboring mutationally activated or amplified KIT arising on mucosal, acral, and chronically sun-damaged skin. Journal of Clinical Oncology, 31(26), 3182–3190. 10.1200/jco.2012.47.7836 [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Hodis E, Watson IR, Kryukov GV, Arold ST, Imielinski M, Theurillat J-P, … Chin L (2012). A landscape of driver mutations in melanoma. Cell, 150(2), 251–263. 10.1016/j.cell.2012.06.024 [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Holman CD, & Armstrong BK (1984). Cutaneous malignant melanoma and indicators of total accumulated exposure to the sun: An analysis separating histogenetic types. Journal of the National Cancer Institute, 73(1), 75–82. [PubMed] [Google Scholar]
  56. Hugdahl E, Kalvenes MB, Puntervoll HE, Ladstein RG, & Akslen LA (2016). BRAF-V600E expression in primary nodular melanoma is associated with aggressive tumour features and reduced survival. British Journal of Cancer, 114(7), 801–808. 10.1038/bjc.2016.44 [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Iida Y, Salomon MP, Hata K, Tran K, Ohe S, Griffiths CF, … Hoon DSB (2018). Predominance of triple wild-type and IGF2R mutations in mucosal melanomas. BMC Cancer, 18(1), 1054 10.1186/s12885-018-4977-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Jakob JA, Bassett RL, Ng CS, Curry JL, Joseph RW, Alvarado GC, … Davies MA (2012). NRAS mutation status is an independent prognostic factor in metastatic melanoma. Cancer, 118(16), 4014–4023. 10.1002/cncr.26724 [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Joseph EW, Pratilas CA, Poulikakos PI, Tadi M, Wang W, Taylor BS, … Rosen N (2010). The RAF inhibitor PLX4032 inhibits ERK signaling and tumor cell proliferation in a V600E BRAF-selective manner. Proceedings of the National Academy of Sciences United States of America, 107(33), 14903–14908. 10.1073/pnas.1008990107 [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Kalbasi A, & Ribas A (2020). Tumour-intrinsic resistance to immune checkpoint blockade. Nature Reviews Immunology, 20(1), 25–39. 10.1038/s41577-019-0218-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Kallini JR, Jain SK, & Khachemoune A (2013). Lentigo maligna: Review of salient characteristics and management. American Journal of Clinical Dermatology, 14(6), 473–480. 10.1007/s40257-013-0044-6 [DOI] [PubMed] [Google Scholar]
  62. Kantor J, & Kantor DE (2009). Routine dermatologist-performed full-body skin examination and early melanoma detection. Archives of Dermatology, 145(8), 873–876. 10.1001/archdermatol.2009.137 [DOI] [PubMed] [Google Scholar]
  63. Kaunitz GJ, Cottrell TR, Lilo M, Muthappan V, Esandrio J, Berry S, … Taube JM (2017). Melanoma subtypes demonstrate distinct PD-L1 expression profiles. Laboratory Investigation, 97(9), 1063–1071. 10.1038/labinvest.2017.64 [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Khalili JS, Yu X, Wang J, Hayes BC, Davies MA, Lizee G, … Woodman SE (2012). Combination small molecule MEK and PI3K inhibition enhances uveal melanoma cell death in a mutant GNAQ-and GNA11-dependent manner. Clinical Cancer Research, 18(16), 4345–4355. 10.1158/1078-0432.Ccr-11-3227 [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Kim G, McKee AE, Ning Y-M, Hazarika M, Theoret M, Johnson JR, … Pazdur R (2014). FDA approval summary: Vemurafenib for treatment of unresectable or metastatic melanoma with the BRAFV600E mutation. Clinical Cancer Research, 20(19), 4994–5000. 10.1158/1078-0432.Ccr-14-0776 [DOI] [PubMed] [Google Scholar]
  66. Kim KB, Eton O, Davis DW, Frazier ML, McConkey DJ, Diwan AH, … Prieto VG (2008). Phase II trial of imatinib mesylate in patients with metastatic melanoma. British Journal of Cancer, 99(5), 734–740. 10.1038/sj.bjc.6604482 [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Kong Y, Sheng X, Wu X, Yan J, Ma M, Yu J, … Guo J (2017). Frequent genetic aberrations in the CDK4 pathway in acral melanoma indicate the potential for CDK4/6 inhibitors in targeted therapy. Clinical Cancer Research, 23(22), 6946–6957. 10.1158/1078-0432.Ccr-17-0070 [DOI] [PubMed] [Google Scholar]
  68. Kottschade LA, McWilliams RR, Markovic SN, Block MS, Villasboas Bisneto J, Pham AQ, … Dronca RS (2016). The use of pembrolizumab for the treatment of metastatic uveal melanoma. Melanoma Research, 26(3), 300–303. 10.1097/cmr.0000000000000242 [DOI] [PubMed] [Google Scholar]
  69. Krantz BA, Dave N, Komatsubara KM, Marr BP, & Carvajal RD (2017). Uveal melanoma: Epidemiology, etiology, and treatment of primary disease. Clinical Ophthalmology (Auckland, N.Z.), 11, 279–289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Krepler C, Sproesser K, Brafford P, Beqiri M, Garman B, Xiao M, … Herlyn M (2017). A comprehensive patient-derived xenograft collection representing the heterogeneity of melanoma. Cell Reports, 21(7), 1953–1967. 10.1016/j.celrep.2017.10.021 [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Krepler C, Xiao M, Sproesser K, Brafford PA, Shannan B, Beqiri M, … Herlyn M (2016). Personalized preclinical trials in BRAF inhibitor-resistant patient-derived xenograft models identify second-line combination therapies. Clinical Cancer Research, 22(7), 1592–1602. 10.1158/1078-0432.Ccr-15-1762 [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Kwon MR, Choi SH, Jang KT, Kim JH, Mun GH, Lee J, & Lee DY (2019). Acral malignant melanoma; emphasis on the primary metastasis and the usefulness of preoperative ultrasound for sentinel lymph node metastasis. Scientific Reports, 9(1), 15894 10.1038/s41598-019-52180-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Kwong LN, Costello JC, Liu H, Jiang S, Helms TL, Langsdorf AE, … Chin L (2012). Oncogenic NRAS signaling differentially regulates survival and proliferation in melanoma. Nature Medicine, 18(10), 1503–1510. 10.1038/nm.2941 [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Landreville S, Agapova OA, Matatall KA, Kneass ZT, Onken MD, Lee RS, … Harbour JW (2012). Histone deacetylase inhibitors induce growth arrest and differentiation in uveal melanoma. Clinical Cancer Research, 18(2), 408–416. 10.1158/1078-0432.Ccr-11-0946 [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Larkin J, Ascierto PA, Dreno B, Atkinson V, Liszkay G, Maio M, … Ribas A (2014). Combined vemurafenib and cobimetinib in BRAF-mutated melanoma. New England Journal of Medicine, 371(20), 1867– 1876. 10.1056/NEJMoa1408868 [DOI] [PubMed] [Google Scholar]
  76. Lattanzi M, Lee Y, Simpson D, Moran U, Darvishian F, Kim RH, … Osman I (2019). Primary melanoma histologic subtype: Impact on survival and response to therapy. Journal of the National Cancer Institute, 111(2), 180–188. 10.1093/jnci/djy086 [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Ledford H (2011). Melanoma drug wins US approval. Nature, 471(7340), 561 10.1038/471561a [DOI] [PubMed] [Google Scholar]
  78. Long GV, Eroglu Z, Infante J, Patel S, Daud A, Johnson DB, … Flaherty KT (2018). Long-term outcomes in patients with BRAF V600-mutant metastatic melanoma who received dabrafenib combined with trametinib. Journal of Clinical Oncology, 36(7), 667–673. 10.1200/jco.2017.74.1025 [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Luke JJ, Callahan MK, Postow MA, Romano E, Ramaiya N, Bluth M, … Carvajal RD (2013). Clinical activity of ipilimumab for metastatic uveal melanoma: A retrospective review of the Dana-Farber Cancer Institute, Massachusetts General Hospital, Memorial Sloan-Kettering Cancer Center, and University Hospital of Lausanne experience. Cancer, 119(20), 3687–3695. 10.1002/cncr.28282 [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Lutzky J, Bauer J, & Bastian BC (2008). Dose-dependent, complete response to imatinib of a metastatic mucosal melanoma with a K642E KIT mutation. Pigment Cell & Melanoma Research, 21(4), 492–493. 10.1111/j.1755-148X.2008.00475.x [DOI] [PubMed] [Google Scholar]
  81. Mahoney KM, Freeman GJ, & McDermott DF (2015). The next immune-checkpoint inhibitors: PD-1/PD-L1 blockade in melanoma. Clinical Therapeutics, 37(4), 764–782. 10.1016/j.clinthera.2015.02.018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Mandala M, Merelli B, & Massi D (2014). Nras in melanoma: Targeting the undruggable target. Critical Reviews in Oncology Hematology, 92(2), 107–122. 10.1016/j.critrevonc.2014.05.005 [DOI] [PubMed] [Google Scholar]
  83. Marsden JR, Fox R, Boota NM, Cook M, Wheatley K, Billingham LJ, & Steven NM (2017). Effect of topical imiquimod as primary treatment for lentigo maligna: The LIMIT-1 study. British Journal of Dermatology, 176(5), 1148–1154. 10.1111/bjd.15112 [DOI] [PubMed] [Google Scholar]
  84. Martin CA, Cullinane C, Kirby L, Abuhammad S, Lelliott EJ, Waldeck K, … Sheppard KE (2018). Palbociclib synergizes with BRAF and MEK inhibitors in treatment naive melanoma but not after the development of BRAF inhibitor resistance. International Journal of Cancer, 142(10), 2139–2152. 10.1002/ijc.31220 [DOI] [PubMed] [Google Scholar]
  85. Masoomian B, Shields JA, & Shields CL (2018). Overview of BAP1 cancer predisposition syndrome and the relationship to uveal melanoma. Journal of Current Ophthalmology, 30(2), 102–109. 10.1016/j.joco.2018.02.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. Matatall KA, Agapova OA, Onken MD, Worley LA, Bowcock AM, & Harbour JW (2013). BAP1 deficiency causes loss of melanocytic cell identity in uveal melanoma. BMC Cancer, 13, 371–371. 10.1186/1471-2407-13-371 [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. McLaughlin CC, Wu XC, Jemal A, Martin HJ, Roche LM, & Chen VW (2005). Incidence of noncutaneous melanomas in the U.S. Cancer, 103(5), 1000–1007. 10.1002/cncr.20866 [DOI] [PubMed] [Google Scholar]
  88. Mirzoyev SA, Knudson RM, Reed KB, Hou JL, Lohse CM, Frohm ML, … Roenigk RK (2014). Incidence of lentigo maligna in Olmsted County, Minnesota, 1970 to 2007. Journal of the American Academy of Dermatology, 70(3), 443–448. 10.1016/j.jaad.2013.11.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. Mora AN, Karia PS, & Nguyen BM (2015). A quantitative systematic review of the efficacy of imiquimod monotherapy for lentigo maligna and an analysis of factors that affect tumor clearance. Journal of the American Academy of Dermatology, 73(2), 205–212. 10.1016/j.jaad.2015.05.022 [DOI] [PubMed] [Google Scholar]
  90. Morrison C, Pabla S, Conroy JM, Nesline MK, Glenn ST, Dressman D, … Ernstoff MS (2018). Predicting response to checkpoint inhibitors in melanoma beyond PD-L1 and mutational burden. Journal for ImmunoTherapy of Cancer, 6(1), 32 10.1186/s40425-018-0344-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Moschos SJ, Sullivan RJ, Hwu W-J, Ramanathan RK, Adjei AA, Fong PC, … Flaherty KT (2018). Development of MK-8353, an orally administered ERK1/2 inhibitor, in patients with advanced solid tumors. JCI Insight, 3(4). 10.1172/jci.insight.92352 [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Moya-Plana A, Herrera Gómez RG, Rossoni C, Dercle L, Ammari S, Girault I, … Robert C (2019). Evaluation of the efficacy of immunotherapy for non-resectable mucosal melanoma. Cancer Immunology, Immunotherapy, 68(7), 1171–1178. 10.1007/s00262-019-02351-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Nakamura Y, & Fujisawa Y (2018). Diagnosis and management of acral lentiginous melanoma. Current Treatment Options in Oncology, 19(8), 42 10.1007/s11864-018-0560-y [DOI] [PubMed] [Google Scholar]
  94. Newell F, Kong Y, Wilmott JS, Johansson PA, Ferguson PM, Cui C, … Scolyer RA (2019). Whole-genome landscape of mucosal melanoma reveals diverse drivers and therapeutic targets. Nature Communications, 10(1), 3163 10.1038/s41467-019-11107-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Oiso N, Sakai K, Yanagihara S, Nishio K, & Kawada A (2018). Genital mucosal melanoma with somatic SF3B1 R625C mutation. European Journal of Dermatology, 28(3), 414–415. 10.1684/ejd.2018.3281 [DOI] [PubMed] [Google Scholar]
  96. Padua RA, Barrass N, & Currie GA (1984). A novel transforming gene in a human malignant melanoma cell line. Nature, 311(5987), 671–673. 10.1038/311671a0 [DOI] [PubMed] [Google Scholar]
  97. Panda A, Betigeri A, Subramanian K, Ross JS, Pavlick DC, Ali S, … Ganesan S (2017). Identifying a clinically applicable mutational burden threshold as a potential biomarker of response to immune checkpoint therapy in solid tumors. JCO Precision Oncology, 2017. 10.1200/po.17.00146 [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Paraiso KHT, Fedorenko IV, Cantini LP, Munko AC, Hall M, Sondak VK, … Smalley KSM (2010). Recovery of phospho-ERK activity allows melanoma cells to escape from BRAF inhibitor therapy. British Journal of Cancer, 102(12), 1724–1730. 10.1038/sj.bjc.6605714 [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Patel SG, Prasad ML, Escrig M, Singh B, Shaha AR, Kraus DH, … Shah JP (2002). Primary mucosal malignant melanoma of the head and neck. Head and Neck, 24(3), 247–257. 10.1002/hed.10019 [DOI] [PubMed] [Google Scholar]
  100. Perego M, Maurer M, Wang JX, Shaffer S, Müller AC, Parapatics K, … Herlyn M (2018). A slow-cycling subpopulation of melanoma cells with highly invasive properties. Oncogene, 37(3), 302–312. 10.1038/onc.2017.341 [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. Poulikakos PI, Zhang C, Bollag G, Shokat KM, & Rosen N (2010). RAF inhibitors transactivate RAF dimers and ERK signalling in cells with wild-type BRAF. Nature, 464(7287), 427–430. 10.1038/nature08902 [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Queirolo P, Picasso V, & Spagnolo F (2015). Combined BRAF and MEK inhibition for the treatment of BRAF-mutated metastatic melanoma. Cancer Treatment Reviews, 41(6), 519–526. 10.1016/j.ctrv.2015.04.010 [DOI] [PubMed] [Google Scholar]
  103. Quek C, Rawson RV, Ferguson PM, Shang P, Silva I, Saw RPM, … Wilmott JS (2019). Recurrent hotspot SF3B1 mutations at codon 625 in vulvovaginal mucosal melanoma identified in a study of 27 Australian mucosal melanomas. Oncotarget, 10(9), 930–941. 10.18632/oncotarget.26584 [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Rambow F, Rogiers A, Marin-Bejar O, Aibar S, Femel J, Dewaele M, … Marine J-C (2018). Toward minimal residual disease-directed therapy in melanoma. Cell, 174(4), 843–855.e819. 10.1016/j.cell.2018.06.025 [DOI] [PubMed] [Google Scholar]
  105. Rebecca VW, Alicea GM, Paraiso KH, Lawrence H, Gibney GT, & Smalley KS (2014). Vertical inhibition of the MAPK pathway enhances therapeutic responses in NRAS-mutant melanoma. Pigment Cell Melanoma Res, 27(6), 1154–1158. 10.1111/pcmr.12303 [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Rebecca VW, Wood E, Fedorenko IV, Paraiso KHT, Haarberg HE, Chen YI, … Smalley KSM (2014). Evaluating melanoma drug response and therapeutic escape with quantitative proteomics. Molecular & Cellular Proteomics: MCP, 13(7), 1844–1854. 10.1074/mcp.M113.037424 [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Ribas A, & Flaherty KT (2011). BRAF targeted therapy changes the treatment paradigm in melanoma. Nature Reviews Clinical Oncology, 8(7), 426–433. 10.1038/nrclinonc.2011.69 [DOI] [PubMed] [Google Scholar]
  108. Ribas A, Gonzalez R, Pavlick A, Hamid O, Gajewski TF, Daud A, … McArthur GA (2014). Combination of vemurafenib and cobimetinib in patients with advanced BRAF(V600)-mutated melanoma: A phase 1b study. The Lancet Oncology, 15(9), 954–965. 10.1016/s1470-2045(14)70301-8 [DOI] [PubMed] [Google Scholar]
  109. Rimoldi D, Salvi S, Lienard D, Lejeune FJ, Speiser D, Zografos L, & Cerottini JC (2003). Lack of BRAF mutations in uveal melanoma. Cancer Research, 63(18), 5712–5715. [PubMed] [Google Scholar]
  110. Roesch A, Fukunaga-Kalabis M, Schmidt EC, Zabierowski SE, Brafford PA, Vultur A, … Herlyn M (2010). A temporarily distinct subpopulation of slow-cycling melanoma cells is required for continuous tumor growth. Cell, 141(4), 583–594. 10.1016/j.cell.2010.04.020 [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Rossi E, Schinzari G, Zizzari IG, Maiorano BA, Pagliara MM, Sammarco MG, … Tortora G (2019). Immunological backbone of uveal melanoma: Is there a rationale for immunotherapy? Cancers (Basel), 11(8), 1055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. Saldanha G, Potter L, Daforno P, & Pringle JH (2006). Cutaneous melanoma subtypes show different BRAF and NRAS mutation frequencies. Clinical Cancer Research, 12(15), 4499–4505. 10.1158/1078-0432.Ccr-05-2447 [DOI] [PubMed] [Google Scholar]
  113. Salhi A, Farhadian JA, Giles KM, Vega-Saenz de Miera E, Silva IP, Bourque C, … Osman I (2015). RSK1 activation promotes invasion in nodular melanoma. American Journal of Pathology, 185(3), 704–716. 10.1016/j.ajpath.2014.11.021 [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Schachter J, Ribas A, Long GV, Arance A, Grob J-J, Mortier L, … Robert C (2017). Pembrolizumab versus ipilimumab for advanced melanoma: Final overall survival results of a multicentre, randomised, open-label phase 3 study (KEYNOTE-006). Lancet, 390(10105), 1853–1862. 10.1016/s0140-6736(17)31601-x [DOI] [PubMed] [Google Scholar]
  115. Schadendorf D, Long GV, Stroiakovski D, Karaszewska B, Hauschild A, Levchenko E, … Robert C (2017). Three-year pooled analysis of factors associated with clinical outcomes across dabrafenib and trametinib combination therapy phase 3 randomised trials. European Journal of Cancer, 82, 45–55. 10.1016/j.ejca.2017.05.033 [DOI] [PubMed] [Google Scholar]
  116. Scolyer RA, Long GV, & Thompson JF (2011). Evolving concepts in melanoma classification and their relevance to multidisciplinary melanoma patient care. Molecular Oncology, 5(2), 124–136. 10.1016/j.molonc.2011.03.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  117. Sheng X, Yan X, Chi Z, Si L, Cui C, Tang B, … Guo J (2019). Axitinib in combination with toripalimab, a humanized immunoglobulin G4 monoclonal antibody against programmed cell death-1, in patients with metastatic mucosal melanoma: an open-label phase IB trial. Journal of Clinical Oncology, 37(32), 2987–2999. 10.1200/jco.19.00210 [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Shinozaki M, Fujimoto A, Morton DL, & Hoon DS (2004). Incidence of BRAF oncogene mutation and clinical relevance for primary cutaneous melanomas. Clinical Cancer Research, 10(5), 1753–1757. 10.1158/1078-0432.CCR-1169-3 [DOI] [PubMed] [Google Scholar]
  119. Shoushtari AN, Munhoz RR, Kuk D, Ott PA, Johnson DB, Tsai KK, … Postow MA (2016). The efficacy of anti-PD-1 agents in acral and mucosal melanoma. Cancer, 122(21), 3354–3362. 10.1002/cncr.30259 [DOI] [PMC free article] [PubMed] [Google Scholar]
  120. Silvers DN (1976). Focus on melanoma: The therapeutic dilemma of lentigo maligna (Hutchinson’s freckle). The Journal of Dermatologic Surgery and Oncology, 2(4), 301–303. 10.1111/j.1524-4725.1976.tb00226.x [DOI] [PubMed] [Google Scholar]
  121. Singh AD, Turell ME, & Topham AK (2011). Uveal melanoma: Trends in incidence, treatment, and survival. Ophthalmology, 118(9), 1881–1885. 10.1016/j.ophtha.2011.01.040 [DOI] [PubMed] [Google Scholar]
  122. Spathis A, Katoulis A, Damaskou V, Liakou A, Kottaridi C, Leventakou D, … Panayiotides I (2019). BRAF mutation status in primary, recurrent, and metastatic malignant melanoma and its relation to histopathological parameters. Dermatology Practical & Conceptual, 9(1), 54–62. 10.5826/dpc.0901a13 [DOI] [PMC free article] [PubMed] [Google Scholar]
  123. Stadelmeyer E, Heitzer E, Resel M, Cerroni L, Wolf P, & Dandachi N (2014). The BRAF V600K mutation is more frequent than the BRAF V600E mutation in melanoma in situ of lentigo maligna type. The Journal of Investigative Dermatology, 134(2), 548–550. 10.1038/jid.2013.338 [DOI] [PubMed] [Google Scholar]
  124. Sullivan RJ, Infante JR, Janku F, Wong DJL, Sosman JA, Keedy V, … Li BT (2018). First-in-class ERK1/2 inhibitor ulixertinib (BVD-523) in patients with MAPK mutant advanced solid tumors: results of a phase I dose-escalation and expansion study. Cancer Discovery, 8(2), 184–195. 10.1158/2159-8290.Cd-17-1119 [DOI] [PubMed] [Google Scholar]
  125. Swetter SM, Boldrick JC, Jung SY, Egbert BM, & Harvell JD (2005). Increasing incidence of lentigo maligna melanoma subtypes: Northern California and national trends 1990–2000. The Journal of Investigative Dermatology, 125(4), 685–691. 10.1111/j.0022-202X.2005.23852.x [DOI] [PubMed] [Google Scholar]
  126. Tate JG, Bamford S, Jubb HC, Sondka Z, Beare DM, Bindal N, … Forbes SA (2019). COSMIC: The catalogue of somatic mutations in cancer. Nucleic Acids Research, 47(D1), D941–D947. 10.1093/nar/gky1015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  127. Teh JLF, Cheng PF, Purwin TJ, Nikbakht N, Patel P, Chervoneva I, … Aplin AE (2018). In vivo E2F reporting reveals efficacious schedules of MEK1/2-CDK4/6 targeting and mTOR-S6 resistance mechanisms. Cancer Discovery, 8(5), 568–581. 10.1158/2159-8290.Cd-17-0699 [DOI] [PMC free article] [PubMed] [Google Scholar]
  128. Teramoto Y, Keim U, Gesierich A, Schuler G, Fiedler E, Tüting T, … Garbe C (2018). Acral lentiginous melanoma: A skin cancer with unfavourable prognostic features. A study of the German central malignant melanoma registry (CMMR) in 2050 patients. British Journal of Dermatology, 178(2), 443–451. 10.1111/bjd.15803 [DOI] [PubMed] [Google Scholar]
  129. Turajlic S, Furney SJ, Lambros MB, Mitsopoulos C, Kozarewa I, Geyer FC, … Marais R (2012). Whole genome sequencing of matched primary and metastatic acral melanomas. Genome Research, 22(2), 196–207. 10.1101/gr.125591.111 [DOI] [PMC free article] [PubMed] [Google Scholar]
  130. Tyrrell H, & Payne M (2018). Combatting mucosal melanoma: Recent advances and future perspectives. Melanoma Management, 5(3), Mmt11 10.2217/mmt-2018-0003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  131. Ugurel S, Hildenbrand R, Zimpfer A, La Rosée P, Paschka P, Sucker A, … Schadendorf D (2005). Lack of clinical efficacy of imatinib in metastatic melanoma. British Journal of Cancer, 92(8), 1398–1405. 10.1038/sj.bjc.6602529 [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. van Herpen CML, Agarwala SS, Hauschild A, Berking C, Beck JT, Schadendorf D, … Dummer R (2019). Biomarker results from a phase II study of MEK1/2 inhibitor binimetinib (MEK162) in patients with advanced NRAS- or BRAF-mutated melanoma. Oncotarget, 10(19), 1850–1859. 10.18632/oncotarget.26753 [DOI] [PMC free article] [PubMed] [Google Scholar]
  133. Villanueva J, Infante JR, Krepler C, Reyes-Uribe P, Samanta M, Chen H-Y, … Nathanson KL (2013). Concurrent MEK2 mutation and BRAF amplification confer resistance to BRAF and MEK inhibitors in melanoma. Cell Reports, 4(6), 1090–1099. 10.1016/j.celrep.2013.08.023 [DOI] [PMC free article] [PubMed] [Google Scholar]
  134. Villanueva J, Vultur A, Lee JT, Somasundaram R, Fukunaga-Kalabis M, Cipolla AK, … Herlyn M (2010). Acquired resistance to BRAF inhibitors mediated by a RAF kinase switch in melanoma can be overcome by cotargeting MEK and IGF-1R/PI3K. Cancer Cell, 18(6), 683–695. 10.1016/j.ccr.2010.11.023 [DOI] [PMC free article] [PubMed] [Google Scholar]
  135. Wang Y, Zhao Y, & Ma S (2016). Racial differences in six major subtypes of melanoma: Descriptive epidemiology. BMC Cancer, 16, 691 10.1186/s12885-016-2747-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  136. Wilmott JS, Tembe V, Howle JR, Sharma R, Thompson JF, Rizos H, … Long GV (2012). Intratumoral molecular heterogeneity in a BRAF-mutant, BRAF inhibitor-resistant melanoma: A case illustrating the challenges for personalized medicine. Molecular Cancer Therapeutics, 11(12), 2704–2708. 10.1158/1535-7163.Mct-12-0530 [DOI] [PubMed] [Google Scholar]
  137. Wolchok JD, Chiarion-Sileni V, Gonzalez R, Rutkowski P, Grob J-J, Cowey CL, … Larkin J (2017). Overall survival with combined nivolumab and ipilimumab in advanced melanoma. New England Journal of Medicine, 377(14), 1345–1356. 10.1056/NEJMoa1709684 [DOI] [PMC free article] [PubMed] [Google Scholar]
  138. Wyman K, Atkins MB, Prieto V, Eton O, McDermott DF, Hubbard F, … Sosman JA (2006). Multicenter phase II trial of high-dose imatinib mesylate in metastatic melanoma: Significant toxicity with no clinical efficacy. Cancer, 106(9), 2005–2011. 10.1002/cncr.21834 [DOI] [PubMed] [Google Scholar]
  139. Xiong M, Charifa A, & Chen CSJ (2019). Cancer, lentigo maligna melanoma In StatPearls (Ed.), Cancer, Lentigo Maligna Melanoma. Treasure Island, FL: StatPearls Publishing LLC. [PubMed] [Google Scholar]
  140. Yamazaki N, Kiyohara Y, Uhara H, Uehara J, Fujisawa Y, Takenouchi T, … Minami H (2019). Long-term follow up of nivolumab in previously untreated Japanese patients with advanced or recurrent malignant melanoma. Cancer Science, 110(6), 1995–2003. 10.1111/cas.14015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  141. Yamazaki N, Tanaka R, Tsutsumida A, Namikawa K, Eguchi H, Omata W, … Tsuta K (2015). BRAF V600 mutations and pathological features in Japanese melanoma patients. Melanoma Research, 25(1), 9–14. 10.1097/cmr.0000000000000091 [DOI] [PMC free article] [PubMed] [Google Scholar]
  142. Zaremba A, Murali R, Jansen P, Möller I, Sucker A, Paschen A, … Cosgarea I (2019). Clinical and genetic analysis of melanomas arising in acral sites. European Journal of Cancer, 119, 66–76. 10.1016/j.ejca.2019.07.008 [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES