Next Article in Journal
Chromosome Asynapsis Is the Main Cause of Male Sterility in the Interspecies Hybrids of East Asian Voles (Alexandromys, Rodentia, Arvicolinae)
Next Article in Special Issue
Association between IL-17F, IL-17RA Gene Polymorphisms and Response to Biological Drugs in Psoriasis and Beyond
Previous Article in Journal
Metabarcoding of Antarctic Lichens from Areas with Different Deglaciation Times Reveals a High Diversity of Lichen-Associated Communities
Previous Article in Special Issue
Systemic Retinoids for Generalized Verrucosis Due to Congenital Immunodeficiency: Case Reports and Review of the Literature
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Genomic Landscape of Melanoma and Its Therapeutic Implications

1
Department of Dermatology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
2
Department of Dermatology, Pingtung Hospital, Ministry of Health and Welfare, Pingtung 900, Taiwan
3
Department of Dermatology, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
4
Neuroscience Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
5
Department of Psychiatry, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
6
Department of Psychiatry, Kaohsiung Municipal SiaoGang Hospital, Kaohsiung Medical University, Kaohsiung 812, Taiwan
*
Author to whom correspondence should be addressed.
Genes 2023, 14(5), 1021; https://doi.org/10.3390/genes14051021
Submission received: 25 February 2023 / Revised: 25 March 2023 / Accepted: 28 April 2023 / Published: 29 April 2023
(This article belongs to the Special Issue Genetics of Complex Cutaneous Disorders)

Abstract

:
Melanoma is one of the most aggressive malignancies of the skin. The genetic composition of melanoma is complex and varies among different subtypes. With the aid of recent technologies such as next generation sequencing and single-cell sequencing, our understanding of the genomic landscape of melanoma and its tumor microenvironment has become increasingly clear. These advances may provide explanation to the heterogenic treatment outcomes of melanoma patients under current therapeutic guidelines and provide further insights to the development of potential new therapeutic targets. Here, we provide a comprehensive review on the genetics related to melanoma tumorigenesis, metastasis, and prognosis. We also review the genetics affecting the melanoma tumor microenvironment and its relation to tumor progression and treatment.
Keywords:
melanoma; gene; mutation

1. Introduction

Melanoma is one of the most aggressive cancer types with an estimated incidence of 325,000 worldwide in the year 2020 and is estimated to continue to grow in the future [1]. Melanoma can be classified into cutaneous melanoma, acral melanoma, mucosal melanoma, and uveal melanoma according to the location of its primary origin. Cutaneous melanoma involving non-glabrous skin is the most common melanoma subtype. It is associated with ultraviolet (UV) radiation exposure and is more common in lighter skin types [1]. Acral melanoma occurs on glabrous skin not commonly exposed to UV radiation and is the most common melanoma subtype observed in patients with darker skin types [2]. Mucosal melanoma is a rare (1.3% of melanomas) but aggressive melanoma subtype originating from melanocytes of mucous membranes [3]. Lastly, uveal melanoma, melanoma arising from the choroid, ciliary body, or iris, represents 5% of melanoma and is more prevalent in populations with lighter skin types [4,5]. The mortality of melanoma remains high, especially for thick lesions and metastatic diseases [6].
The genetic composition of melanoma is complex and varies among different subtypes and individuals, resulting in heterogenous treatment outcomes. Recently, a more in-depth understanding of the genomic landscape of melanoma has been made possible with the aid of next generation sequencing, a sequencing technology making large-scale whole genome sequencing more efficient and practical. Another recent technical advancement is single-cell sequencing, which provides genetic information at the resolution of a single cell. A more thorough understanding of tumor heterogeneity and its microenvironment has been made possible with this technology. The tumor microenvironment, especially immune cells, were shown to profoundly affect survival of melanoma patients [7]. This has become increasingly important as immune checkpoint therapy has become standard treatment for advanced melanoma. However, not all patients respond to immune checkpoint therapy [8]. As our understanding of the complex interaction of tumor cells and the microenvironment increases, opportunities to identify favorable treatment responders have emerged.
In this review, we aim to provide a comprehensive review on the genetic alterations related to melanoma tumorigenesis among different melanoma subtypes, gene mutations related to metastatic melanoma, and the treatment implications of different mutated genes.

2. Driver Mutations

Driver mutations occur in the early stages of oncogenesis (i.e., process of normal cells turning into malignant cells) and provide growth advantages to the cancer cells [9]. Identifying these mutations provides potential targets for therapy and information regarding prognosis. Four major genetic subtypes of cutaneous melanoma have been proposed by The Cancer Genome Atlas according to frequently detected hot-spot mutations: mutant BRAF, mutant RAS, mutant NF1, and triple wild-type (TWT), characterized by a lack of BRAF, RAS, or NF1 mutations [10]. The most commonly observed BRAF and NRAS mutations are mutually exclusive [10]. These hot-spot mutations (i.e., mutations observed in significantly increased frequencies compared to normal samples) are all regulators of the mitogen-activated protein kinase (MAPK) pathway involving Ras/RAF/MEK/ERK signaling. Within the TWT subgroup, various driver mutations were observed, including KIT, CTNNB1, GNA11, GNAQ, and EZH2 [10]. Similarly, most driver mutations in the TWT subgroup are MAPK-activating mutations, highlighting the critical role of the MAPK pathway in the pathogenesis of melanoma.
Other frequently involved mutated genes and molecular pathways in the carcinogenesis of melanoma are summarized in Table 1.

2.1. Cutaneous Melanomas

Cutaneous melanomas include superficial spreading melanoma (SSM) and lentigo maligna melanoma (LMM). These melanoma subtypes arise from areas of skin exposed to ultraviolet (UV) radiation, with SSM more common on skin with intermittent high-intensity UV radiation while LMM more commonly arises on areas with chronic UV radiation. These subtypes are the most common melanoma subtypes and are more commonly seen in patients with lighter skin types [11]. Due to their relation with UV radiation exposure, they frequently exhibit a high burden of somatic mutation showing high levels of UV radiation signatures, especially C > T substitutions [12]. In comparison, the genetic aberrations in acral and mucosal melanomas are dominated by structural variants [12]. About 50% of UV-related melanomas harbor BRAF mutations, with valine (V) to glutamic acid (E) substitution in codon 600 (p.V600E) being the most common mutation [10,12,13,14]. Clinically, patients with BRAF mutations are younger than other subtypes [10,13,14,15]. The second most frequently observed is the N/K/H-RAS mutated subtype (20–28%) [10,16]. Contrary to BRAF-mutant melanomas, patients with NRAS-mutant melanoma tend to be older and have chronic sun UV exposure [17]. Following RAS mutation, the third common is the NF1 mutated subtype (14%) [10]. Lastly, 5–10% are TWT harboring less frequently observed driver mutations [10]. Compared to melanomas with hot-spot mutations, TWT melanomas harbor UV signature mutations less frequently [10]. Different from SSM, LMM was shown to have an increased frequency of non-p.V600E BRAF, NF1, and TP53 mutations [15,18].

2.2. Acral Melanoma

Acral melanoma, melanomas arising on sun-protected areas such as the palms, soles, and nail units, is the most common melanoma subtype observed in darker skin types [2]. Compared to UV-related cutaneous melanomas, acral melanomas tend to be diagnosed at a more advanced stage and have worse prognosis even after adjusting for Breslow thickness [19]. Acral melanomas share many similar driver mutations with cutaneous melanomas, with BRAF (10–34.4%) being the most frequently mutated gene in both Asian and Caucasian cohorts [20,21,22,23]. Most BRAF mutations are a p.V600E substitution and only a few cases are valine (V) to aspartate (D) (p.V600D) substitution or valine (V) to leucine (L) (p.V600L) in codon 600 [15,23,24]. Approximately 9–21.9% acral melanomas have NRAS mutations and 11–23% acral melanomas harbor NF1 mutations [22]. Acral melanomas have a higher proportion of triple-negative melanomas (45–58%) compared to UV-related melanomas [12,15,24]. These triple-negative melanomas most commonly contain KIT (10.9–24.4%), GNAQ (18.6%), TYRP1 mutations [15,20,21,22,23]. Mutations in TP53, PTEN, DDX3X, RASA2, PP6C, RAC1, or RB1 were less frequently observed in acral melanomas and only reported in limited studies [12,20].
Compared to UV-related melanomas, acral melanomas exhibit a higher frequency of genomic structural variations and lower point mutation burden. Up to 75% of acral melanomas were found to have copy number variations (CNVs), and most AMs exhibit at least one structural variation (SV), including deletions, duplications, tandem duplications, and foldback inversions [12,20,22]. These CNVs and SVs mostly involve cell-cycle aberrations (CDK4/6, CCND1/2, CDKN2A) and gains in receptor tyrosine kinase (RTK) related genes and antiapoptosis genes (BIRC2, BIRC3, BIRC5) [15].

2.3. Mucosal Melanoma

Other rare melanoma subtypes unrelated to UV radiation include uveal melanoma and mucosal melanoma. Mucosal melanomas (MMs) are more prevalent in Asian populations with a slight female predominance due to increased melanomas involving the female genital tract [25]. Compared to cutaneous melanomas; patients with MM have a significantly lower 5-year survival rate (25%) due to frequent late diagnosis [25,26,27]. KIT mutation (19.1–23.1%) was the most frequently found driver mutation in mucosal melanomas followed by NF1 (7.8–16.4%), RAS (6.2–17.9%), and BRAF (3.1–16.4%) mutations [27,28,29]. Other significantly mutated genes in MM include SF3B1 (8.1–11.9%), SPRED1 (4.0–7.5%), and ATRX (6.0%) [28,29]. G-alpha protein GNAQ/GNA11 has been described to occur in up to 9.5% of MM [30]. Atypical BRAF mutations other than the p.V600 hotspot are more frequent in MM [28]. Similar to acral melanoma; MM harbors less mutational burden compared to melanoma associated with UV radiation and exhibits more structural variations and copy number alterations [27,28,29].

2.4. Uveal Melanoma

Uveal melanomas (UMs) are melanomas arising from the uveal tract and are the most common tumor of the eye in adults. The overall survival is poor for metastatic UM [4]. Similar to cutaneous melanomas, UMs are more common in patients with lighter skin types [4,31]. However, the genomic landscape for UM is completely different from other melanoma subtypes. UMs typically lack common driver mutations identified in cutaneous melanomas [32]. Significantly mutated driver mutations for UM include GNAAQ/GNA11 (88–92.5%), BAP1 (45%), SF3B1 (24%), EIEF1AX (7–17%), CYSLTR2, SRSF2 (4%), MAPKAPK5, and PLCB4 (2.5%) [32,33,34]. These significantly mutated genes had not been identified in CMs [35]. Chromosome copy number aberrations play an important role in risk stratification of UM. Currently, four distinct subtypes related to prognosis are identified based on copy number loss of chromosome 3 and gain of chromosomes 6p and 8q [32].
Table 2 summarizes genetic alteration in different melanoma subtypes.

3. Telomere and Melanoma Tumorigenesis

Telomeres are DNA sequences located at the end of chromosomes. Its integrity is crucial in maintaining genome stability and is regulated by telomerase, a ribonucleoprotein enzyme that synthesizes telomeric DNA [43]. Telomerase reverse transcriptase (TERT) aberrations are the most common noncoding mutation (i.e., mutation of regions of DNA that do not code for amino acids) in melanoma expressed in early melanoma progression following MAPK pathway activation [44]. TERT promoter is an important regulator of telomerase, whose mutation alters telomere length and promotes tumor cell immortalization [45]. Short telomeres and TERT aberrations are associated with poor survival in melanoma [46,47,48]. In a study including 1019 patients on early melanoma (stage I and II disease) by Rachakonda et al., the hazard ratio for poor melanoma-specific survival was 2.05 (95% CI = 1.33–3.16) for short telomers compared to long telomers [46]. The effect of short telomers on survival was especially prominent in patients under 30 years of age [46]. Patients with TERT promotor mutation and concomitant BRAF or NRAS mutations were shown to have worse disease-free survival compared to patients with TERT promotor mutation or BRAF/NRAS mutations alone [47]. TERT mutation can be used to identify melanoma patients with poor prognosis, including relapsing, disease, and death [47]. Over 60% of UV-related melanomas exhibit TERT promoter mutations [10]. Within the four major genetic subtypes of cutaneous melanomas, TERT promoter mutation is observed in 75% of tumors with BRAF mutation but only observed in 6.7% of TWT melanomas [10]. Although less frequent than UV-related melanomas, 9–41% of acral melanomas [21,39] and 30% of mucosal melanomas [29,40] exhibit TERT alterations. These are more commonly copy number gains (i.e., gene amplification due to increased gene copies) rather than promoter mutations [40]. Different from other melanoma subtypes, TERT alterations are rare in uveal melanomas [41,42].

4. Cell Cycle Regulators and Tumor Suppressor Genes

4.1. Cell Cycle Regulators

DNA replication in the process of cell proliferation of the human cell is regulated by several cell cycle regulators. These important players in cell regulation include cyclins, cyclin-dependent kinases (CDKs), CDK inhibitors, and the tumor suppressor retinoblastoma (RB) family of proteins [49]. Mutations in genes encoding these regulators may result in aberrant cell proliferation and cancer [49].
As driver mutations occur at the early stages of tumor development, cell cycle regulators play an important role in sustaining cell proliferation in carcinogenesis, resulting in tumor progression. For example, BRAF mutation alone is not sufficient for inducing melanoma as BRAF mutations are frequently observed in benign nevi [50,51]. Additional genetic alterations in BRAF-mutants are required for cancer development [50]. Commonly, aberrations in cell cycle regulation collaborate with driver mutations in the carcinogenesis of melanoma. Significant players in melanoma cell cycle regulation include the CDKN2A (cyclin dependent kinase inhibitor 2A) gene which encodes the tumor suppressor p16INK4A, p14ARF, CDKs controlling specific check points of the cell cycle, and the tumor suppressor retinoblastoma protein (RB). Aberrations of the p16INK4A:cyclin D-CK4/6:RB pathway is frequently observed in both cutaneous melanomas and acral melanomas with a prevalence of 68.8–82.7% [10,15,17,52,53]. CDKN2A is also the most significant germline mutation with high penetrance in familial melanoma [54].
Mutations of cell cycle regulators CDKN2A and CDKs were less common in acral melanoma compared with cutaneous melanoma [20]. Melanomas with cell-cycle aberrations are associated with a higher frequency of clinical ulceration, which is associated with worse prognosis [15].

4.2. Tumor Suppressor Genes

TP53 is one of the major tumor suppressor genes (TSGs) regulating cell division and is the most commonly altered gene in human malignancies [55,56]. Its encoded protein p53 plays a critical role in maintaining genomic integrity by promoting cell arrest and apoptosis. Mutations of TP53 result in rapid tumor progression and metastasis [56]. Despite being the most frequently mutated gene in cutaneous melanoma, BRAF hyperactivity alone is not sufficient for melanoma development without concurrent p53 inactivation [57]. Although frequently observed in various human cancers, the incidence of TP53 mutation (11–30%) is relatively lower in melanoma [58,59], indicating that alterations of other cell cycle regulators, including CDKN2A and its products p14ARF and p16INK4A, are more frequently involved in melanoma tumorigenesis [60]. Melanoma patients with TP53 mutations were shown to have worse prognosis compared to those without the mutation [61,62]. Moreover, TP53 mutation was shown to be associated with poorer response and poorer overall survival to cytotoxic T-lymphocyte antigen-4 (CTLA-4) blockade immunotherapy in metastatic melanoma due to impaired cytotoxic T-cell-induced apoptosis of tumor cells [61].
Similar to TP53, PTEN is also a critical tumor suppressor gene in melanoma development. It inhibits cell cycle progression by blocking the PI3K pathway [63]. PTEN inactivating mutations occur in the later stages of primary melanoma and are associated with poorer overall survival [63]. PTEN loss is also associated with impaired host T cell responses [63,64]. Hence, similar to TP53 mutation, PTEN loss melanomas also show inferior response rate to immune blockade therapies [65,66].

5. Metastatic Melanoma

Melanoma is an aggressive cancer and has the tendency to metastasize, leading to the patient’s death [67]. Alterations of the MAPK pathway occur early in cutaneous melanoma tumorigenesis and continue to be amplified in later stages, predominantly by gene duplication as the tumor progresses [44,68]. Currently, there are no genetic mutations exclusively found in metastatic melanoma [44,68,69]. However, several hub genes associated with metastatic melanoma were identified using bioinformatic approaches. These genes include CDK1, BAP1, CXCL8, THBS1, KIT, DSG1, FLG, and PKP1 [70,71,72,73]. Within the identified hub genes, CDK1 was found to be associated with favorable prognosis [70]. CDK1 was also associated with increased lymphocyte infiltration, which is also an indicator of favorable prognosis [70]. On the contrary, DSG1, FLG, and PKP1 were poor prognostic factors [72]. Loss-of-function mutation of BAP-1 was associated with poor overall survival [71]. Recently, melanoblast-specific genes, such as KDELR3 and KDELR1, were identified to contribute to the tumor’s ability to metastasize [74]. These findings may provide potential targets for melanoma treatment. Currently, CDK1 inhibition shows preliminary effects in inducing tumor cell apoptosis in colorectal cancer [75].

6. Tumor-Infiltrating Lymphocytes

The behavior of tumor cells is influenced by the surrounding tumor microenvironment, which consists of host immune cells, extracellular matrix, blood vessels, and stroma cells [76]. An important factor influencing melanoma prognosis is the interaction between the host immune cells and tumor cells (i.e., tumor stroma immunobiology). The prognosis of melanoma is more heavily influenced by the tumor stroma immunobiology than the genetic subtypes of driver mutations [10]. Tumor-infiltrating lymphocytes (TIL) have first been shown to be an independent positive prognostic factor for melanoma by Larsen and Grude in 1978 and confirmed by many later studies [77,78,79]. Consequently, as shown in The Cancer Genome Atlas (TCGA) program, advanced cutaneous melanomas expressing genes associated with increased immune cell infiltration and signaling are associated with a favorable survival after adjusting for potential contamination by lymph node tissues [10]. However, it is worth noting that the subclass of lymphocytes also affects prognosis. For example, increased infiltration of regulatory T cells is associated with worse prognosis [80]. Nonetheless, lymphocyte subclass analysis was only performed in limited studies, which may lead to potential biases [80]. Interestingly, in contrast with other melanoma subtypes, increased tumor infiltrating in UM is associated with worse prognosis [32], indicating that UM is a unique subset of melanoma.

7. Therapeutic and Prognostic Implications

7.1. Targeting the MAPK Pathway

Early studies have shown that BRAF-mutant tumors have worse prognosis (median survival 5.7 vs. 8.5 months) in metastatic UV-related melanomas [13]. However, this has not been confirmed by more recent larger studies [10,13,15]. As the most frequently mutated gene in cutaneous melanoma, BRAF has become a popular target for melanoma treatment. BRAF inhibitors (BRAFi) are currently approved for BRAF V600E and V600K mutant melanoma and successfully improved the progression-free survival and overall survival of patients with these mutations [81,82]. However, the clinical benefits of BRAFi were shown to be short-lived due to drug resistance. The median progression-free survival was 5.1 and 6.8 months for dabrafenib and vemurafenib, respectively [83,84]. MAPK pathway activation by bypassing BRAF is the most common mechanism of BRAFi resistance. Hence, by combining BRAF and MEK (mitogen-activated protein kinase kinase) inhibitors, the survival benefits of these targeted therapies have been prolonged [81,82,85]. Although both BRAF V600E and V600K mutant melanoma benefit from BRAFi, melanomas with BRAF V600K mutations have shorter progression-free survival under BRAF inhibitors but superior response to immune checkpoint inhibitors compared to V600E mutants [10,14,15,18,86]. The progression-free survival for BRAF V600E and V600K mutant melanomas was 6.9 and 5.9 months for vemurafenib and 6.3 and 4.5 months in the Phase III trial of dabrafenib [87,88]. Among the different melanoma subtypes, BRAFi benefits cutaneous melanomas the most, as they have the highest frequency of BRAF V600 mutations. Mucosal melanoma frequently harbors atypical BRAF mutations other than the p.V600 hot spot (such as p.G469A, p.L597Q, p.N581S, p.T599I, and p.G596R), rendering BRAF inhibitors that revolutionized melanoma treatment not applicable in this population [28].
NRAS induces cell-cycle dysregulation and cell proliferation by activating downstream signaling of the MAPK pathway [89]. MEK inhibitors has been used to treat advanced NRAS-mutant melanoma. However, the results were short-lived, and only resulted in improvement in median progression-free survival for approximately 3 months [90,91,92]. Strategies directly targeting NRAS has been attempted by inhibiting farnesyltransferase, a key enzyme in RAS maturation, but have not been successfully applied in a clinical setting due to alternative prenylation via geranylgeranyl transferase type 1 [93,94,95,96].

7.2. KIT as a Potential Treatment Target

As KIT mutations are one of the most frequently mutated genes in melanomas, it is a potential target for advanced melanoma treatment. Although currently there are no approved KIT inhibitors for melanoma, KIT inhibitor imatinib has shown to provide a durable response to a subset of patients with KIT mutant metastatic melanoma with a complete response lasting for more than 1.5 years in a subset of patients [97,98]. Studies on the effeteness of other KIT inhibitors including nilotinib and ripretinib are still ongoing [99,100]. KIT mutation is relatively rare in uveal melanoma [36]. Nonetheless, most primary uveal melanomas express KIT protein on tumor immunohistochemical analysis, making KIT a potential target for treatment. However, the results of KIT inhibition by imatinib on UM are disappointing, as no patient showed a response to imatinib in early studies [101,102].

7.3. Microphthalmia-Associated Transcription Factor

Microphthalmia-associated transcription factor (MITF) is an important regulator in melanoma development and progression. MITF mutation is frequently observed in melanoma [10]. The role of MITF in melanoma is paradoxical as it is an oncogene but also has properties in suppressing melanoma invasion and metastasis [103,104]. Compared to melanomas with high levels of MITF expression (MITF-high), MITF-low melanomas are shown to be more invasive [104]. Moreover, melanoma with low MITF expression is associated with resistance to BRAF inhibition therapy due to reactivation of ERK in the MAPK pathway [104]. The expression of MITF is negatively correlated with the expression of AXL, a receptor tyrosine kinase. MITF-low/AXL-high melanomas, characterized by low MITF expression and high AXL markers, are associated with intrinsic resistance to RAF/MEK inhibition therapy. Although melanoma can be classified into MITF-low/AXL-high or MITF-high/AXL-low melanoma at the tumor level, subclones with AXL-high and MITF-high expression co-exist in melanoma at the single cell level [105]. The application of BRAFi selectively selects in favor of AXL-high subclones and contributes to subsequent drug resistance [105].

7.4. TERT Promoter Aberrations

TERT promoter mutations are associated with poorer survival and higher metastatic rate in both UV-related and acral melanomas [47,106,107]. Due to the high prevalence of TERT promoter aberrations in various melanoma subtypes, it is a potential target for melanoma treatment. As TERT alterations are common in BRAF V600 mutant melanomas and plays an important role in controlling the apoptosis of cancer cells, combined therapies targeting both BRAF and TERT may potentially improvement the survival of these patients [108]. Similarly, a high frequency of TERT promoter aberrations is also found in NRAS mutant melanomas, making TERT activity inhibition a potential therapeutic target in this population [109]. TERT mutation status has also been used as a potential biomarker indicating favorable response to anti-cytotoxic T lymphocyte-associated antigen 4 (anti-CTLA4) treatment [110].

7.5. Cell-Cycle Aberrations

Cell-cycle aberrations are an independent prognostic factor for melanoma-specific survival indicating worse prognosis [15]. As cell-cycle aberrations frequently occur in melanoma, its inhibitors are possible treatment opportunities for these patients. Recently, palbociclib, an FDA-approved CDK4/6 inhibitor for treating metastatic breast cancer, has been demonstrated to show antitumor effects by inhibiting tumor growth in acral melanoma and mucosal melanoma with CDK4 amplifications in patient-derived xenograft models [27,52,111]. However, palbociclib only showed a modest effect (20% patients had tumor shrinkage at 8 weeks) as monotherapy in advanced acral melanoma with CDK4 pathway gene aberrations in a phase II clinical study [112]. Further clinical studies are required to evaluate its effectiveness on melanoma patients. Other CDK4/6 inhibitors include ribociclib and abemaciclib. Similarly, these agents are currently applied to a limited proportion of melanoma patients and further clinical studies are required to evaluate their effectiveness [113]. Cell-cycle inhibitors combined with targeted therapies against the MAPK pathway have been shown to demonstrate a synergic effect in treating melanoma [111,114,115].

7.6. Immune Checkpoint Inhibitors

Immune checkpoint inhibitors (ICIs) targeting programmed cell death protein 1 (PD-1) and cytotoxic T-lymphocyte–associated antigen 4 (CTLA-4) have become the standard care of advanced melanoma in the past 10 years and revolutionized the treatment of melanoma by significantly increasing the overall survival of melanoma patients [116,117,118,119,120]. Not all melanomas respond to ICIs. Currently, only approximately one-third of melanomas respond to ICI treatment [121,122,123]. Within different melanoma subtypes, the response rate to ICIs is lower in acral melanoma compared to cutaneous melanomas. Genetic markers indicating favorable ICI responders in melanoma include high tumor mutational burden [98,124], BRAF wild type [98], and increased gene expression of inflammation and T-cell population [10,98,125]. Tumor programmed death-1 ligand 1 (PD-L1) expression is also an important factor influencing therapeutic response to anti-PD-1 immunotherapy, with PD-L1-positive melanoma demonstrating a higher response rate to anti-PD-1 therapy compared to their PD-L1-negative counterpart [126]. Overall, 35% of melanoma express PD-L1, with chronic sun-damaged melanoma showing the highest percentage of PD-L1-positive tumors (62%), followed by mucosal melanoma (44%), acral melanoma (31%), and uveal melanoma (10%) [127]. PD-L1 expression is independent of melanoma driver mutation status (BRAF/RAS/NF1) [128]. A more accurate prediction for ICI therapy response may be achieved by combining the above factors.

8. Conclusions

The frequencies of key driver gene and pathway alterations differ among different melanoma subtypes, reflecting their heterogenic background in pathogenesis and prognosis. In this era of personal medicine, an in-depth understanding of the genetics of melanoma is crucial in developing personalized treatment and clinical decision-making for targeted therapies of specific genes. Currently, BRAF-targeted therapy has been successful in significantly improving overall survival in BRAF-mutant melanomas and c-kit inhibitors have been applied in melanomas with the corresponding mutation. Other potential targeted therapies are under development as more significantly mutated genes and pathways have been identified. More recently, ICI therapy has become the standard treatment for advanced melanoma and has significantly improved the survival of these patients. Since only a proportion of patients respond to this revolutionizing therapy, the identification of potential genetic markers indicating favorable responses has become increasingly important. A deepened understanding of the genetic background of melanoma allows us to utilize the best combination of treatments according to the tumor genetic background and predict treatment response more accurately. However, many recently developed targeted therapies are applied only on a limited number of melanoma patients and larger clinical studies are required to verify their effectiveness in this population. It is also worth mentioning that whole genome sequencing is only applied in a limited number of studies and important regulatory noncoding alterations and single-nucleotide variants may be overlooked under whole exome sequencing or low-pass whole genome sequencing.

Author Contributions

Conceptualization, T.-T.Y., S.Y. and S.-T.C.; literature review, T.-T.Y., S.Y., C.-L.K.K. and S.-T.C.; writing—original draft preparation, T.-T.Y.; writing—review and editing, T.-T.Y. and S.-T.C.; supervision, S.Y. and S.-T.C.; project administration, S.-T.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors thank Yi-Shan Teng for insightful discussions and comments on the manuscript.

Conflicts of Interest

Sebastian Yu, is a guest editor of the Special Issue: Genetics of Complex Cutaneous Disorders, had no role in the peer review process or decision to publish this article.

References

  1. Arnold, M.; Singh, D.; Laversanne, M.; Vignat, J.; Vaccarella, S.; Meheus, F.; Cust, A.E.; de Vries, E.; Whiteman, D.C.; Bray, F. Global Burden of Cutaneous Melanoma in 2020 and Projections to 2040. JAMA Dermatol. 2022, 158, 495. [Google Scholar] [CrossRef] [PubMed]
  2. Chang, J.W.-C. Acral Melanoma: A Unique Disease in Asia. JAMA Dermatol. 2013, 149, 1272. [Google Scholar] [CrossRef]
  3. McLaughlin, C.C.; Wu, X.-C.; Jemal, A.; Martin, H.J.; Roche, L.M.; Chen, V.W. Incidence of Noncutaneous Melanomas in the U.S. Cancer 2005, 103, 1000–1007. [Google Scholar] [CrossRef] [PubMed]
  4. Kaliki, S.; Shields, C.L. Uveal Melanoma: Relatively Rare but Deadly Cancer. Eye 2017, 31, 241–257. [Google Scholar] [CrossRef]
  5. Chattopadhyay, C.; Kim, D.W.; Gombos, D.S.; Oba, J.; Qin, Y.; Williams, M.D.; Esmaeli, B.; Grimm, E.A.; Wargo, J.A.; Woodman, S.E.; et al. Uveal Melanoma: From Diagnosis to Treatment and the Science in between: Uveal Melanoma Review. Cancer 2016, 122, 2299–2312. [Google Scholar] [CrossRef]
  6. Baade, P.D.; Whiteman, D.C.; Janda, M.; Cust, A.E.; Neale, R.E.; Smithers, B.M.; Green, A.C.; Khosrotehrani, K.; Mar, V.; Soyer, H.P.; et al. Long-Term Deaths from Melanoma According to Tumor Thickness at Diagnosis. Int. J. Cancer 2020, 147, 1391–1396. [Google Scholar] [CrossRef] [PubMed]
  7. Azimi, F.; Scolyer, R.A.; Rumcheva, P.; Moncrieff, M.; Murali, R.; McCarthy, S.W.; Saw, R.P.; Thompson, J.F. Tumor-Infiltrating Lymphocyte Grade Is an Independent Predictor of Sentinel Lymph Node Status and Survival in Patients with Cutaneous Melanoma. J. Clin. Oncol. 2012, 30, 2678–2683. [Google Scholar] [CrossRef] [PubMed]
  8. Huang, A.C.; Zappasodi, R. A Decade of Checkpoint Blockade Immunotherapy in Melanoma: Understanding the Molecular Basis for Immune Sensitivity and Resistance. Nat. Immunol. 2022, 23, 660–670. [Google Scholar] [CrossRef]
  9. Stratton, M.R.; Campbell, P.J.; Futreal, P.A. The Cancer Genome. Nature 2009, 458, 719–724. [Google Scholar] [CrossRef]
  10. Akbani, R.; Akdemir, K.C.; Aksoy, B.A.; Albert, M.; Ally, A.; Amin, S.B.; Arachchi, H.; Arora, A.; Auman, J.T.; Ayala, B.; et al. Genomic Classification of Cutaneous Melanoma. Cell 2015, 161, 1681–1696. [Google Scholar] [CrossRef]
  11. Whiteman, D.C.; Pavan, W.J.; Bastian, B.C. The Melanomas: A Synthesis of Epidemiological, Clinical, Histopathological, Genetic, and Biological Aspects, Supporting Distinct Subtypes, Causal Pathways, and Cells of Origin: The Melanomas. Pigment Cell Melanoma Res. 2011, 24, 879–897. [Google Scholar] [CrossRef] [PubMed]
  12. Hayward, N.K.; Wilmott, J.S.; Waddell, N.; Johansson, P.A.; Field, M.A.; Nones, K.; Patch, A.-M.; Kakavand, H.; Alexandrov, L.B.; Burke, H.; et al. Whole-Genome Landscapes of Major Melanoma Subtypes. Nature 2017, 545, 175–180. [Google Scholar] [CrossRef]
  13. Long, G.V.; Menzies, A.M.; Nagrial, A.M.; Haydu, L.E.; Hamilton, A.L.; Mann, G.J.; Hughes, T.M.; Thompson, J.F.; Scolyer, R.A.; Kefford, R.F. Prognostic and Clinicopathologic Associations of Oncogenic BRAF in Metastatic Melanoma. J. Clin. Oncol. 2011, 29, 1239–1246. [Google Scholar] [CrossRef]
  14. Bauer, J.; Büttner, P.; Murali, R.; Okamoto, I.; Kolaitis, N.A.; Landi, M.T.; Scolyer, R.A.; Bastian, B.C. BRAF Mutations in Cutaneous Melanoma Are Independently Associated with Age, Anatomic Site of the Primary Tumor, and the Degree of Solar Elastosis at the Primary Tumor Site. Pigment Cell Melanoma Res. 2011, 24, 345–351. [Google Scholar] [CrossRef]
  15. Sheen, Y.-S.; Tan, K.-T.; Tse, K.-P.; Liao, Y.-H.; Lin, M.-H.; Chen, J.-S.; Liau, J.-Y.; Tseng, Y.-J.; Lee, C.-H.; Hong, C.-H.; et al. Genetic Alterations in Primary Melanoma in Taiwan. Br. J. Dermatol. 2020, 182, 1205–1213. [Google Scholar] [CrossRef]
  16. Jakob, J.A.; Bassett, R.L.; Ng, C.S.; Curry, J.L.; Joseph, R.W.; Alvarado, G.C.; Rohlfs, M.L.; Richard, J.; Gershenwald, J.E.; Kim, K.B.; et al. NRAS Mutation Status Is an Independent Prognostic Factor in Metastatic Melanoma. Cancer 2012, 118, 4014–4023. [Google Scholar] [CrossRef] [PubMed]
  17. Curtin, J.A.; Fridlyand, J.; Kageshita, T.; Patel, H.N.; Busam, K.J.; Kutzner, H.; Cho, K.-H.; Aiba, S.; Bröcker, E.-B.; LeBoit, P.E.; et al. Distinct Sets of Genetic Alterations in Melanoma. N. Engl. J. Med. 2005, 353, 2135–2147. [Google Scholar] [CrossRef] [PubMed]
  18. Sanna, A.; Harbst, K.; Johansson, I.; Christensen, G.; Lauss, M.; Mitra, S.; Rosengren, F.; Häkkinen, J.; Vallon-Christersson, J.; Olsson, H.; et al. Tumor Genetic Heterogeneity Analysis of Chronic Sun-damaged Melanoma. Pigment Cell Melanoma Res. 2020, 33, 480–489. [Google Scholar] [CrossRef] [PubMed]
  19. Bradford, P.T.; Goldstein, A.M.; McMaster, M.L.; Tucker, M.A. Acral Lentiginous Melanoma: Incidence and Survival Patterns in the United States, 1986–2005. Arch. Dermatol. 2009, 145, 427–434. [Google Scholar] [CrossRef] [PubMed]
  20. Moon, K.R.; Choi, Y.D.; Kim, J.M.; Jin, S.; Shin, M.-H.; Shim, H.-J.; Lee, J.-B.; Yun, S.J. Genetic Alterations in Primary Acral Melanoma and Acral Melanocytic Nevus in Korea: Common Mutated Genes Show Distinct Cytomorphological Features. J. Investig. Dermatol. 2018, 138, 933–945. [Google Scholar] [CrossRef] [PubMed]
  21. Zaremba, A.; Murali, R.; Jansen, P.; Möller, I.; Sucker, A.; Paschen, A.; Zimmer, L.; Livingstone, E.; Brinker, T.J.; Hadaschik, E.; et al. Clinical and Genetic Analysis of Melanomas Arising in Acral Sites. Eur. J. Cancer 2019, 119, 66–76. [Google Scholar] [CrossRef] [PubMed]
  22. 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]
  23. Elefanti, L.; Zamuner, C.; Del Fiore, P.; Stagni, C.; Pellegrini, S.; Dall’Olmo, L.; Fabozzi, A.; Senetta, R.; Ribero, S.; Salmaso, R.; et al. The Molecular Landscape of Primary Acral Melanoma: A Multicenter Study of the Italian Melanoma Intergroup (IMI). Int. J. Mol. Sci. 2021, 22, 3826. [Google Scholar] [CrossRef]
  24. Gao, H.-W.; Tsai, W.-C.; Perng, C.-L.; Wang, W.-M.; Chiang, C.-P. Distinct MAPK and PI3K Pathway Mutations in Different Melanoma Types in Taiwanese Individuals. Eur. J. Dermatol. EJD 2018, 28, 509–518. [Google Scholar] [CrossRef] [PubMed]
  25. Patrick, R.J.; Fenske, N.A.; Messina, J.L. Primary Mucosal Melanoma. J. Am. Acad. Dermatol. 2007, 56, 828–834. [Google Scholar] [CrossRef]
  26. Hahn, H.; Lee, K.; Choi, W.; Cheong, S.; Myung, K.; Hahn, H. An Updated Review of Mucosal Melanoma: Survival Meta-analysis. Mol. Clin. Oncol. 2019, 11, 116–126. [Google Scholar] [CrossRef]
  27. Zhou, R.; Shi, C.; Tao, W.; Li, J.; Wu, J.; Han, Y.; Yang, G.; Gu, Z.; Xu, S.; Wang, Y.; et al. Analysis of Mucosal Melanoma Whole-Genome Landscapes Reveals Clinically Relevant Genomic Aberrations. Clin. Cancer Res. 2019, 25, 3548–3560. [Google Scholar] [CrossRef]
  28. 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]
  29. Newell, F.; Kong, Y.; Wilmott, J.S.; Johansson, P.A.; Ferguson, P.M.; Cui, C.; Li, Z.; Kazakoff, S.H.; Burke, H.; Dodds, T.J.; et al. Whole-Genome Landscape of Mucosal Melanoma Reveals Diverse Drivers and Therapeutic Targets. Nat. Commun. 2019, 10, 3163. [Google Scholar] [CrossRef]
  30. Sheng, X.; Kong, Y.; Li, Y.; Zhang, Q.; Si, L.; Cui, C.; Chi, Z.; Tang, B.; Mao, L.; Lian, B.; et al. GNAQ and GNA11 Mutations Occur in 9.5% of Mucosal Melanoma and Are Associated with Poor Prognosis. Eur. J. Cancer 2016, 65, 156–163. [Google Scholar] [CrossRef] [PubMed]
  31. Weis, E. The Association between Host Susceptibility Factors and Uveal Melanoma: A Meta-Analysis. Arch. Ophthalmol. 2006, 124, 54. [Google Scholar] [CrossRef] [PubMed]
  32. Robertson, A.G.; Shih, J.; Yau, C.; Gibb, E.A.; Oba, J.; Mungall, K.L.; Hess, J.M.; Uzunangelov, V.; Walter, V.; Danilova, L.; et al. Integrative Analysis Identifies Four Molecular and Clinical Subsets in Uveal Melanoma. Cancer Cell 2017, 32, 204–220.e15. [Google Scholar] [CrossRef] [PubMed]
  33. Decatur, C.L.; Ong, E.; Garg, N.; Anbunathan, H.; Bowcock, A.M.; Field, M.G.; Harbour, J.W. Driver Mutations in Uveal Melanoma: Associations with Gene Expression Profile and Patient Outcomes. JAMA Ophthalmol. 2016, 134, 728. [Google Scholar] [CrossRef]
  34. Johansson, P.A.; Brooks, K.; Newell, F.; Palmer, J.M.; Wilmott, J.S.; Pritchard, A.L.; Broit, N.; Wood, S.; Carlino, M.S.; Leonard, C.; et al. Whole Genome Landscapes of Uveal Melanoma Show an Ultraviolet Radiation Signature in Iris Tumours. Nat. Commun. 2020, 11, 2408. [Google Scholar] [CrossRef] [PubMed]
  35. Johnson, D.B.; Roszik, J.; Shoushtari, A.N.; Eroglu, Z.; Balko, J.M.; Higham, C.; Puzanov, I.; Patel, S.P.; Sosman, J.A.; Woodman, S.E. Comparative Analysis of the GNAQ, GNA11, SF3B1, and EIF1AX Driver Mutations in Melanoma and across the Cancer Spectrum. Pigment Cell Melanoma Res. 2016, 29, 470–473. [Google Scholar] [CrossRef]
  36. Wallander, M.L.; Layfield, L.J.; Emerson, L.L.; Mamalis, N.; Davis, D.; Tripp, S.R.; Holden, J.A. KIT Mutations in Ocular Melanoma: Frequency and Anatomic Distribution. Mod. Pathol. 2011, 24, 1031–1035. [Google Scholar] [CrossRef] [PubMed]
  37. Tsao, H.; Mihm, M.C.; Sheehan, C. PTEN Expression in Normal Skin, Acquired Melanocytic Nevi, and Cutaneous Melanoma. J. Am. Acad. Dermatol. 2003, 49, 865–872. [Google Scholar] [CrossRef]
  38. Li, Y.; Shi, J.; Yang, J.; Ge, S.; Zhang, J.; Jia, R.; Fan, X. Uveal Melanoma: Progress in Molecular Biology and Therapeutics. Ther. Adv. Med. Oncol. 2020, 12, 175883592096585. [Google Scholar] [CrossRef] [PubMed]
  39. Rabbie, R.; Ferguson, P.; Molina-Aguilar, C.; Adams, D.J.; Robles-Espinoza, C.D. Melanoma Subtypes: Genomic Profiles, Prognostic Molecular Markers and Therapeutic Possibilities. J. Pathol. 2019, 247, 539–551. [Google Scholar] [CrossRef]
  40. Ma, Y.; Xia, R.; Ma, X.; Judson-Torres, R.L.; Zeng, H. Mucosal Melanoma: Pathological Evolution, Pathway Dependency and Targeted Therapy. Front. Oncol. 2021, 11, 702287. [Google Scholar] [CrossRef]
  41. Griewank, K.G.; Murali, R.; Schilling, B.; Scholz, S.; Sucker, A.; Song, M.; Süsskind, D.; Grabellus, F.; Zimmer, L.; Hillen, U.; et al. TERT Promoter Mutations in Ocular Melanoma Distinguish between Conjunctival and Uveal Tumours. Br. J. Cancer 2013, 109, 497–501. [Google Scholar] [CrossRef] [PubMed]
  42. Dono, M.; Angelini, G.; Cecconi, M.; Amaro, A.; Esposito, A.I.; Mirisola, V.; Maric, I.; Lanza, F.; Nasciuti, F.; Viaggi, S.; et al. 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. Br. J. Cancer 2014, 110, 1058–1065. [Google Scholar] [CrossRef]
  43. Maciejowski, J.; de Lange, T. Telomeres in Cancer: Tumour Suppression and Genome Instability. Nat. Rev. Mol. Cell Biol. 2017, 18, 175–186. [Google Scholar] [CrossRef] [PubMed]
  44. Shain, A.H.; Joseph, N.M.; Yu, R.; Benhamida, J.; Liu, S.; Prow, T.; Ruben, B.; North, J.; Pincus, L.; Yeh, I.; et al. Genomic and Transcriptomic Analysis Reveals Incremental Disruption of Key Signaling Pathways during Melanoma Evolution. Cancer Cell 2018, 34, 45–55.e4. [Google Scholar] [CrossRef]
  45. Chiba, K.; Johnson, J.Z.; Vogan, J.M.; Wagner, T.; Boyle, J.M.; Hockemeyer, D. Cancer-Associated TERT Promoter Mutations Abrogate Telomerase Silencing. eLife 2015, 4, e07918. [Google Scholar] [CrossRef]
  46. Rachakonda, S.; Srinivas, N.; Mahmoudpour, S.H.; Garcia-Casado, Z.; Requena, C.; Traves, V.; Soriano, V.; Cardelli, M.; Pjanova, D.; Molven, A.; et al. Telomere Length and Survival in Primary Cutaneous Melanoma Patients. Sci. Rep. 2018, 8, 10947. [Google Scholar] [CrossRef]
  47. Nagore, E.; Heidenreich, B.; Rachakonda, S.; Garcia-Casado, Z.; Requena, C.; Soriano, V.; Frank, C.; Traves, V.; Quecedo, E.; Sanjuan-Gimenez, J.; et al. TERT Promoter Mutations in Melanoma Survival: TERT Promoter Mutations in Melanoma Survival. Int. J. Cancer 2016, 139, 75–84. [Google Scholar] [CrossRef]
  48. Griewank, K.G.; Murali, R.; Puig-Butille, J.A.; Schilling, B.; Livingstone, E.; Potrony, M.; Carrera, C.; Schimming, T.; Möller, I.; Schwamborn, M.; et al. TERT Promoter Mutation Status as an Independent Prognostic Factor in Cutaneous Melanoma. JNCI J. Natl. Cancer Inst. 2014, 106, dju246. [Google Scholar] [CrossRef] [PubMed]
  49. Leal-Esteban, L.C.; Fajas, L. Cell Cycle Regulators in Cancer Cell Metabolism. Biochim. Biophys. Acta BBA Mol. Basis Dis. 2020, 1866, 165715. [Google Scholar] [CrossRef] [PubMed]
  50. Paluncic, J.; Kovacevic, Z.; Jansson, P.J.; Kalinowski, D.; Merlot, A.M.; Huang, M.L.-H.; Lok, H.C.; Sahni, S.; Lane, D.J.R.; Richardson, D.R. Roads to Melanoma: Key Pathways and Emerging Players in Melanoma Progression and Oncogenic Signaling. Biochim. Biophys. Acta BBA Mol. Cell Res. 2016, 1863, 770–784. [Google Scholar] [CrossRef] [PubMed]
  51. Pollock, P.M.; Harper, U.L.; Hansen, K.S.; Yudt, L.M.; Stark, M.; Robbins, C.M.; Moses, T.Y.; Hostetter, G.; Wagner, U.; Kakareka, J.; et al. High Frequency of BRAF Mutations in Nevi. Nat. Genet. 2003, 33, 19–20. [Google Scholar] [CrossRef] [PubMed]
  52. Kong, Y.; Sheng, X.; Wu, X.; Yan, J.; Ma, M.; Yu, J.; Si, L.; Chi, Z.; Cui, C.; Dai, J.; et al. Frequent Genetic Aberrations in the CDK4 Pathway in Acral Melanoma Indicate the Potential for CDK4/6 Inhibitors in Targeted Therapy. Clin. Cancer Res. 2017, 23, 6946–6957. [Google Scholar] [CrossRef]
  53. Young, R.J.; Waldeck, K.; Martin, C.; Foo, J.H.; Cameron, D.P.; Kirby, L.; Do, H.; Mitchell, C.; Cullinane, C.; Liu, W.; et al. Loss of CDKN2A Expression Is a Frequent Event in Primary Invasive Melanoma and Correlates with Sensitivity to the CDK4/6 Inhibitor PD0332991 in Melanoma Cell Lines. Pigment Cell Melanoma Res. 2014, 27, 590–600. [Google Scholar] [CrossRef] [PubMed]
  54. Zocchi, L.; Lontano, A.; Merli, M.; Dika, E.; Nagore, E.; Quaglino, P.; Puig, S.; Ribero, S. Familial Melanoma and Susceptibility Genes: A Review of the Most Common Clinical and Dermoscopic Phenotypic Aspect, Associated Malignancies and Practical Tips for Management. J. Clin. Med. 2021, 10, 3760. [Google Scholar] [CrossRef] [PubMed]
  55. Hollstein, M.; Sidransky, D.; Vogelstein, B.; Harris, C.C. P53 Mutations in Human Cancers. Science 1991, 253, 49–53. [Google Scholar] [CrossRef] [PubMed]
  56. Kastenhuber, E.R.; Lowe, S.W. Putting P53 in Context. Cell 2017, 170, 1062–1078. [Google Scholar] [CrossRef]
  57. Patton, E.E.; Widlund, H.R.; Kutok, J.L.; Kopani, K.R.; Amatruda, J.F.; Murphey, R.D.; Berghmans, S.; Mayhall, E.A.; Traver, D.; Fletcher, C.D.M.; et al. BRAF Mutations Are Sufficient to Promote Nevi Formation and Cooperate with P53 in the Genesis of Melanoma. Curr. Biol. CB 2005, 15, 249–254. [Google Scholar] [CrossRef]
  58. Weiss, J.; Heine, M.; Arden, K.C.; Körner, B.; Pilch, H.; Herbst, R.A.; Jung, E.G. Mutation and Expression of TP53 in Malignant Melanomas. In Skin Cancer: Basic Science, Clinical Research and Treatment; Garbe, C., Schmitz, S., Orfanos, C.E., Eds.; Recent Results in Cancer Research; Springer: Berlin/Heidelberg, Germany, 1995; Volume 139, pp. 137–154. ISBN 978-3-642-78773-7. [Google Scholar]
  59. Hussein, M.R. The TP53 Tumor Suppressor Gene and Melanoma Tumorigenesis: Is There a Relationship? Tumor Biol. 2004, 25, 200–207. [Google Scholar] [CrossRef] [PubMed]
  60. Hocker, T.L.; Singh, M.K.; Tsao, H. Melanoma Genetics and Therapeutic Approaches in the 21st Century: Moving from the Benchside to the Bedside. J. Investig. Dermatol. 2008, 128, 2575–2595. [Google Scholar] [CrossRef]
  61. Xiao, W.; Du, N.; Huang, T.; Guo, J.; Mo, X.; Yuan, T.; Chen, Y.; Ye, T.; Xu, C.; Wang, W.; et al. TP53 Mutation as Potential Negative Predictor for Response of Anti-CTLA-4 Therapy in Metastatic Melanoma. EBioMedicine 2018, 32, 119–124. [Google Scholar] [CrossRef]
  62. Gould Rothberg, B.E.; Berger, A.J.; Molinaro, A.M.; Subtil, A.; Krauthammer, M.O.; Camp, R.L.; Bradley, W.R.; Ariyan, S.; Kluger, H.M.; Rimm, D.L. Melanoma Prognostic Model Using Tissue Microarrays and Genetic Algorithms. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2009, 27, 5772–5780. [Google Scholar] [CrossRef]
  63. Cabrita, R.; Mitra, S.; Sanna, A.; Ekedahl, H.; Lövgren, K.; Olsson, H.; Ingvar, C.; Isaksson, K.; Lauss, M.; Carneiro, A.; et al. The Role of PTEN Loss in Immune Escape, Melanoma Prognosis and Therapy Response. Cancers 2020, 12, 742. [Google Scholar] [CrossRef]
  64. Dong, Y.; Richards, J.-A.; Gupta, R.; Aung, P.P.; Emley, A.; Kluger, Y.; Dogra, S.K.; Mahalingam, M.; Wajapeyee, N. PTEN Functions as a Melanoma Tumor Suppressor by Promoting Host Immune Response. Oncogene 2014, 33, 4632–4642. [Google Scholar] [CrossRef]
  65. Peng, W.; Chen, J.Q.; Liu, C.; Malu, S.; Creasy, C.; Tetzlaff, M.T.; Xu, C.; McKenzie, J.A.; Zhang, C.; Liang, X.; et al. Loss of PTEN Promotes Resistance to T Cell–Mediated Immunotherapy. Cancer Discov. 2016, 6, 202–216. [Google Scholar] [CrossRef] [PubMed]
  66. Vidotto, T.; Melo, C.M.; Castelli, E.; Koti, M.; dos Reis, R.B.; Squire, J.A. Emerging Role of PTEN Loss in Evasion of the Immune Response to Tumours. Br. J. Cancer 2020, 122, 1732–1743. [Google Scholar] [CrossRef]
  67. Braeuer, R.R.; Watson, I.R.; Wu, C.-J.; Mobley, A.K.; Kamiya, T.; Shoshan, E.; Bar-Eli, M. Why Is Melanoma So Metastatic? Pigment Cell Melanoma Res. 2014, 27, 19–36. [Google Scholar] [CrossRef]
  68. Vergara, I.A.; Mintoff, C.P.; Sandhu, S.; McIntosh, L.; Young, R.J.; Wong, S.Q.; Colebatch, A.; Cameron, D.L.; Kwon, J.L.; Wolfe, R.; et al. Evolution of Late-Stage Metastatic Melanoma Is Dominated by Aneuploidy and Whole Genome Doubling. Nat. Commun. 2021, 12, 1434. [Google Scholar] [CrossRef] [PubMed]
  69. Manca, A.; Paliogiannis, P.; Colombino, M.; Casula, M.; Lissia, A.; Botti, G.; Caracò, C.; Ascierto, P.A.; Sini, M.C.; Palomba, G.; et al. Mutational Concordance between Primary and Metastatic Melanoma: A Next-Generation Sequencing Approach. J. Transl. Med. 2019, 17, 289. [Google Scholar] [CrossRef]
  70. Luan, H.; Jian, L.; He, Y.; Zhang, T.; Zhou, L. Exploration and Validation of Metastasis-Associated Genes for Skin Cutaneous Melanoma. Sci. Rep. 2022, 12, 13002. [Google Scholar] [CrossRef] [PubMed]
  71. Chen, J.; Wu, F.; Shi, Y.; Yang, D.; Xu, M.; Lai, Y.; Liu, Y. Identification of Key Candidate Genes Involved in Melanoma Metastasis. Mol. Med. Rep. 2019, 20, 903–914. [Google Scholar] [CrossRef]
  72. Jia, C.-L.; Yang, F.; Li, R.-N. Identification of Potential Core Genes Between Primary and Metastatic Malignant Melanoma and Analysis of Their Immune Correlation. Int. J. Gen. Med. 2022, 15, 379–391. [Google Scholar] [CrossRef] [PubMed]
  73. Su, W.; Guan, Y.; Huang, B.; Wang, J.; Wei, Y.; Zhao, Y.; Jiao, Q.; Ji, J.; Yu, D.; Xu, L. Bioinformatic Analysis Reveals Hub Genes and Pathways That Promote Melanoma Metastasis. BMC Cancer 2020, 20, 863. [Google Scholar] [CrossRef]
  74. Marie, K.L.; Sassano, A.; Yang, H.H.; Michalowski, A.M.; Michael, H.T.; Guo, T.; Tsai, Y.C.; Weissman, A.M.; Lee, M.P.; Jenkins, L.M.; et al. Melanoblast Transcriptome Analysis Reveals Pathways Promoting Melanoma Metastasis. Nat. Commun. 2020, 11, 333. [Google Scholar] [CrossRef] [PubMed]
  75. Zhang, P.; Kawakami, H.; Liu, W.; Zeng, X.; Strebhardt, K.; Tao, K.; Huang, S.; Sinicrope, F.A. Targeting CDK1 and MEK/ERK Overcomes Apoptotic Resistance in BRAF-Mutant Human Colorectal Cancer. Mol. Cancer Res. 2018, 16, 378–389. [Google Scholar] [CrossRef] [PubMed]
  76. Anderson, N.M.; Simon, M.C. The Tumor Microenvironment. Curr. Biol. 2020, 30, R921–R925. [Google Scholar] [CrossRef]
  77. Lee, N.; Zakka, L.R.; Mihm, M.C.; Schatton, T. Tumour-Infiltrating Lymphocytes in Melanoma Prognosis and Cancer Immunotherapy. Pathology 2016, 48, 177–187. [Google Scholar] [CrossRef]
  78. Clemente, C.G.; Mihm, M.C.; Bufalino, R.; Zurrida, S.; Collini, P.; Cascinelli, N. Prognostic Value of Tumor Infiltrating Lymphocytes in the Vertical Growth Phase of Primary Cutaneous Melanoma. Cancer 1996, 77, 1303–1310. [Google Scholar] [CrossRef]
  79. Mihm, M.C.; Mulé, J.J. Reflections on the Histopathology of Tumor-Infiltrating Lymphocytes in Melanoma and the Host Immune Response. Cancer Immunol. Res. 2015, 3, 827–835. [Google Scholar] [CrossRef]
  80. Fridman, W.H.; Zitvogel, L.; Sautès–Fridman, C.; Kroemer, G. The Immune Contexture in Cancer Prognosis and Treatment. Nat. Rev. Clin. Oncol. 2017, 14, 717–734. [Google Scholar] [CrossRef]
  81. Long, G.V.; Hauschild, A.; Santinami, M.; Atkinson, V.; Mandalà, M.; Chiarion-Sileni, V.; Larkin, J.; Nyakas, M.; Dutriaux, C.; Haydon, A.; et al. Adjuvant Dabrafenib plus Trametinib in Stage III BRAF-Mutated Melanoma. N. Engl. J. Med. 2017, 377, 1813–1823. [Google Scholar] [CrossRef] [PubMed]
  82. Long, G.V.; Flaherty, K.T.; Stroyakovskiy, D.; Gogas, H.; Levchenko, E.; de Braud, F.; Larkin, J.; Garbe, C.; Jouary, T.; Hauschild, A.; et al. Dabrafenib plus Trametinib versus Dabrafenib Monotherapy in Patients with Metastatic BRAF V600E/K-Mutant Melanoma: Long-Term Survival and Safety Analysis of a Phase 3 Study. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2017, 28, 1631–1639. [Google Scholar] [CrossRef] [PubMed]
  83. Hauschild, A.; Grob, J.-J.; Demidov, L.V.; Jouary, T.; Gutzmer, R.; Millward, M.; Rutkowski, P.; Blank, C.U.; Miller, W.H.; Kaempgen, E.; et al. Dabrafenib in BRAF-Mutated Metastatic Melanoma: A Multicentre, Open-Label, Phase 3 Randomised Controlled Trial. Lancet 2012, 380, 358–365. [Google Scholar] [CrossRef] [PubMed]
  84. Sosman, J.A.; Kim, K.B.; Schuchter, L.; Gonzalez, R.; Pavlick, A.C.; Weber, J.S.; McArthur, G.A.; Hutson, T.E.; Moschos, S.J.; Flaherty, K.T.; et al. Survival in BRAF V600–Mutant Advanced Melanoma Treated with Vemurafenib. N. Engl. J. Med. 2012, 366, 707–714. [Google Scholar] [CrossRef] [PubMed]
  85. Long, G.V.; Stroyakovskiy, D.; Gogas, H.; Levchenko, E.; de Braud, F.; Larkin, J.; Garbe, C.; Jouary, T.; Hauschild, A.; Grob, J.J.; et al. Combined BRAF and MEK Inhibition versus BRAF Inhibition Alone in Melanoma. N. Engl. J. Med. 2014, 371, 1877–1888. [Google Scholar] [CrossRef] [PubMed]
  86. Pires da Silva, I.; Wang, K.Y.X.; Wilmott, J.S.; Holst, J.; Carlino, M.S.; Park, J.J.; Quek, C.; Wongchenko, M.; Yan, Y.; Mann, G.; et al. Distinct Molecular Profiles and Immunotherapy Treatment Outcomes of V600E and V600K BRAF-Mutant Melanoma. Clin. Cancer Res. 2019, 25, 1272–1279. [Google Scholar] [CrossRef]
  87. McArthur, G.A.; Chapman, P.B.; Robert, C.; Larkin, J.; Haanen, J.B.; Dummer, R.; Ribas, A.; Hogg, D.; Hamid, O.; Ascierto, P.A.; et al. Safety and Efficacy of Vemurafenib in BRAF(V600E) and BRAF(V600K) Mutation-Positive Melanoma (BRIM-3): Extended Follow-up of a Phase 3, Randomised, Open-Label Study. Lancet Oncol. 2014, 15, 323–332. [Google Scholar] [CrossRef]
  88. Ascierto, P.A.; Minor, D.; Ribas, A.; Lebbe, C.; O’Hagan, A.; Arya, N.; Guckert, M.; Schadendorf, D.; Kefford, R.F.; Grob, J.-J.; et al. Phase II Trial (BREAK-2) of the BRAF Inhibitor Dabrafenib (GSK2118436) in Patients with Metastatic Melanoma. J. Clin. Oncol. 2013, 31, 3205–3211. [Google Scholar] [CrossRef]
  89. Simanshu, D.K.; Nissley, D.V.; McCormick, F. RAS Proteins and Their Regulators in Human Disease. Cell 2017, 170, 17–33. [Google Scholar] [CrossRef] [PubMed]
  90. Ascierto, P.A.; Schadendorf, D.; Berking, C.; Agarwala, S.S.; van Herpen, C.M.; Queirolo, P.; Blank, C.U.; Hauschild, A.; Beck, J.T.; St-Pierre, A.; et al. MEK162 for Patients with Advanced Melanoma Harbouring NRAS or Val600 BRAF Mutations: A Non-Randomised, Open-Label Phase 2 Study. Lancet Oncol. 2013, 14, 249–256. [Google Scholar] [CrossRef]
  91. 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]
  92. Lebbé, C.; Dutriaux, C.; Lesimple, T.; Kruit, W.; Kerger, J.; Thomas, L.; Guillot, B.; de 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]
  93. Cox, A.D.; Der, C.J.; Philips, M.R. Targeting RAS Membrane Association: Back to the Future for Anti-RAS Drug Discovery? Clin. Cancer Res. 2015, 21, 1819–1827. [Google Scholar] [CrossRef] [PubMed]
  94. Niessner, H.; Beck, D.; Sinnberg, T.; Lasithiotakis, K.; Maczey, E.; Gogel, J.; Venturelli, S.; Berger, A.; Mauthe, M.; Toulany, M.; et al. The Farnesyl Transferase Inhibitor Lonafarnib Inhibits MTOR Signaling and Enforces Sorafenib-Induced Apoptosis in Melanoma Cells. J. Investig. Dermatol. 2011, 131, 468–479. [Google Scholar] [CrossRef] [PubMed]
  95. Randic, T.; Kozar, I.; Margue, C.; Utikal, J.; Kreis, S. NRAS Mutant Melanoma: Towards Better Therapies. Cancer Treat. Rev. 2021, 99, 102238. [Google Scholar] [CrossRef] [PubMed]
  96. Ryan, M.B.; Corcoran, R.B. Therapeutic Strategies to Target RAS-Mutant Cancers. Nat. Rev. Clin. Oncol. 2018, 15, 709–720. [Google Scholar] [CrossRef]
  97. Carvajal, R.D. KIT as a Therapeutic Target in Metastatic Melanoma. JAMA 2011, 305, 2327. [Google Scholar] [CrossRef]
  98. Hodi, F.S.; Corless, C.L.; Giobbie-Hurder, A.; Fletcher, J.A.; Zhu, M.; Marino-Enriquez, A.; Friedlander, P.; Gonzalez, R.; Weber, J.S.; Gajewski, T.F.; et al. Imatinib for Melanomas Harboring Mutationally Activated or Amplified KIT Arising on Mucosal, Acral, and Chronically Sun-Damaged Skin. J. Clin. Oncol. 2013, 31, 3182–3190. [Google Scholar] [CrossRef]
  99. Guo, J.; Carvajal, R.D.; Dummer, R.; Hauschild, A.; Daud, A.; Bastian, B.C.; Markovic, S.N.; Queirolo, P.; Arance, A.; Berking, C.; et al. Efficacy and Safety of Nilotinib in Patients with KIT-Mutated Metastatic or Inoperable Melanoma: Final Results from the Global, Single-Arm, Phase II TEAM Trial. Ann. Oncol. 2017, 28, 1380–1387. [Google Scholar] [CrossRef]
  100. Janku, F.; Bauer, S.; Shoumariyeh, K.; Jones, R.L.; Spreafico, A.; Jennings, J.; Psoinos, C.; Meade, J.; Ruiz-Soto, R.; Chi, P. Efficacy and Safety of Ripretinib in Patients with KIT-Altered Metastatic Melanoma. ESMO Open 2022, 7, 100520. [Google Scholar] [CrossRef]
  101. Hofmann, U.B.; Kauczok-Vetter, C.S.; Houben, R.; Becker, J.C. Overexpression of the KIT/SCF in Uveal Melanoma Does Not Translate into Clinical Efficacy of Imatinib Mesylate. Clin. Cancer Res. 2009, 15, 324–329. [Google Scholar] [CrossRef]
  102. Calipel, A.; Landreville, S.; De La Fouchardière, A.; Mascarelli, F.; Rivoire, M.; Penel, N.; Mouriaux, F. Mechanisms of Resistance to Imatinib Mesylate in KIT-Positive Metastatic Uveal Melanoma. Clin. Exp. Metastasis 2014, 31, 553–564. [Google Scholar] [CrossRef]
  103. Garraway, L.A.; Widlund, H.R.; Rubin, M.A.; Getz, G.; Berger, A.J.; Ramaswamy, S.; Beroukhim, R.; Milner, D.A.; Granter, S.R.; Du, J.; et al. Integrative Genomic Analyses Identify MITF as a Lineage Survival Oncogene Amplified in Malignant Melanoma. Nature 2005, 436, 117–122. [Google Scholar] [CrossRef]
  104. Hoek, K.S.; Schlegel, N.C.; Brafford, P.; Sucker, A.; Ugurel, S.; Kumar, R.; Weber, B.L.; Nathanson, K.L.; Phillips, D.J.; Herlyn, M.; et al. Metastatic Potential of Melanomas Defined by Specific Gene Expression Profiles with No BRAF Signature. Pigment Cell Res. 2006, 19, 290–302. [Google Scholar] [CrossRef] [PubMed]
  105. Tirosh, I.; Izar, B.; Prakadan, S.M.; Wadsworth, M.H.; Treacy, D.; Trombetta, J.J.; Rotem, A.; Rodman, C.; Lian, C.; Murphy, G.; et al. Dissecting the Multicellular Ecosystem of Metastatic Melanoma by Single-Cell RNA-Seq. Science 2016, 352, 189–196. [Google Scholar] [CrossRef] [PubMed]
  106. Hugdahl, E.; Kalvenes, M.B.; Mannelqvist, M.; Ladstein, R.G.; Akslen, L.A. Prognostic Impact and Concordance of TERT Promoter Mutation and Protein Expression in Matched Primary and Metastatic Cutaneous Melanoma. Br. J. Cancer 2018, 118, 98–105. [Google Scholar] [CrossRef]
  107. Diaz, A.; Puig-Butillé, J.A.; Muñoz, C.; Costa, D.; Díez, A.; Garcia-Herrera, A.; Carrera, C.; Badenas, C.; Solé, F.; Malvehy, J.; et al. TERT Gene Amplification Is Associated with Poor Outcome in Acral Lentiginous Melanoma. J. Am. Acad. Dermatol. 2014, 71, 839–841. [Google Scholar] [CrossRef]
  108. Del Bianco, P.; Stagni, C.; Giunco, S.; Fabozzi, A.; Elefanti, L.; Pellegrini, S.; Vecchiato, A.; Pigozzo, J.; Zamuner, C.; De Rossi, A.; et al. TERT Promoter Mutations Differently Correlate with the Clinical Outcome of MAPK Inhibitor-Treated Melanoma Patients. Cancers 2020, 12, 946. [Google Scholar] [CrossRef]
  109. Reyes-Uribe, P.; Adrianzen-Ruesta, M.P.; Deng, Z.; Echevarria-Vargas, I.; Mender, I.; Saheb, S.; Liu, Q.; Altieri, D.C.; Murphy, M.E.; Shay, J.W.; et al. Exploiting TERT Dependency as a Therapeutic Strategy for NRAS-Mutant Melanoma. Oncogene 2018, 37, 4058–4072. [Google Scholar] [CrossRef] [PubMed]
  110. Li, H.; Li, J.; Zhang, C.; Zhang, C.; Wang, H. TERT Mutations Correlate with Higher TMB Value and Unique Tumor Microenvironment and May Be a Potential Biomarker for Anti-CTLA4 Treatment. Cancer Med. 2020, 9, 7151–7160. [Google Scholar] [CrossRef]
  111. Lee, B.; Sandhu, S.; McArthur, G. Cell Cycle Control as a Promising Target in Melanoma. Curr. Opin. Oncol. 2015, 27, 141–150. [Google Scholar] [CrossRef]
  112. Mao, L.; Dai, J.; Cao, Y.; Bai, X.; Sheng, X.; Chi, Z.; Cui, C.; Kong, Y.; Zhang, Y.; Wu, L.; et al. Palbociclib in Advanced Acral Melanoma with Genetic Aberrations in the Cyclin-Dependent Kinase 4 Pathway. Eur. J. Cancer 2021, 148, 297–306. [Google Scholar] [CrossRef] [PubMed]
  113. Garutti, M.; Targato, G.; Buriolla, S.; Palmero, L.; Minisini, A.M.; Puglisi, F. CDK4/6 Inhibitors in Melanoma: A Comprehensive Review. Cells 2021, 10, 1334. [Google Scholar] [CrossRef] [PubMed]
  114. Kwong, L.N.; Costello, J.C.; Liu, H.; Jiang, S.; Helms, T.L.; Langsdorf, A.E.; Jakubosky, D.; Genovese, G.; Muller, F.L.; Jeong, J.H.; et al. Oncogenic NRAS Signaling Differentially Regulates Survival and Proliferation in Melanoma. Nat. Med. 2012, 18, 1503–1510. [Google Scholar] [CrossRef] [PubMed]
  115. Schuler, M.; Zimmer, L.; Kim, K.B.; Sosman, J.A.; Ascierto, P.A.; Postow, M.A.; De Vos, F.Y.F.L.; van Herpen, C.M.L.; Carlino, M.S.; Johnson, D.B.; et al. Phase Ib/II Trial of Ribociclib in Combination with Binimetinib in Patients with NRAS-Mutant Melanoma. Clin. Cancer Res. 2022, 28, 3002–3010. [Google Scholar] [CrossRef]
  116. Hodi, F.S.; O’Day, S.J.; McDermott, D.F.; Weber, R.W.; Sosman, J.A.; Haanen, J.B.; Gonzalez, R.; Robert, C.; Schadendorf, D.; Hassel, J.C.; et al. Improved Survival with Ipilimumab in Patients with Metastatic Melanoma. N. Engl. J. Med. 2010, 363, 711–723. [Google Scholar] [CrossRef]
  117. Arheden, A.; Skalenius, J.; Bjursten, S.; Stierner, U.; Ny, L.; Levin, M.; Jespersen, H. Real-World Data on PD-1 Inhibitor Therapy in Metastatic Melanoma. Acta Oncol. 2019, 58, 962–966. [Google Scholar] [CrossRef]
  118. Hamid, O.; Robert, C.; Daud, A.; Hodi, F.S.; Hwu, W.J.; Kefford, R.; Wolchok, J.D.; Hersey, P.; Joseph, R.; Weber, J.S.; et al. Five-Year Survival Outcomes for Patients with Advanced Melanoma Treated with Pembrolizumab in KEYNOTE-001. Ann. Oncol. 2019, 30, 582–588. [Google Scholar] [CrossRef]
  119. Hodi, F.S.; Chiarion-Sileni, V.; Gonzalez, R.; Grob, J.-J.; Rutkowski, P.; Cowey, C.L.; Lao, C.D.; Schadendorf, D.; Wagstaff, J.; Dummer, R.; et al. Nivolumab plus Ipilimumab or Nivolumab Alone versus Ipilimumab Alone in Advanced Melanoma (CheckMate 067): 4-Year Outcomes of a Multicentre, Randomised, Phase 3 Trial. Lancet Oncol. 2018, 19, 1480–1492. [Google Scholar] [CrossRef]
  120. Schadendorf, D.; Hodi, F.S.; Robert, C.; Weber, J.S.; Margolin, K.; Hamid, O.; Patt, D.; Chen, T.-T.; Berman, D.M.; Wolchok, J.D. Pooled Analysis of Long-Term Survival Data from Phase II and Phase III Trials of Ipilimumab in Unresectable or Metastatic Melanoma. J. Clin. Oncol. 2015, 33, 1889–1894. [Google Scholar] [CrossRef]
  121. Brahmer, J.R.; Tykodi, S.S.; Chow, L.Q.M.; Hwu, W.-J.; Topalian, S.L.; Hwu, P.; Drake, C.G.; Camacho, L.H.; Kauh, J.; Odunsi, K.; et al. Safety and Activity of Anti-PD-L1 Antibody in Patients with Advanced Cancer. N. Engl. J. Med. 2012, 366, 2455–2465. [Google Scholar] [CrossRef]
  122. Hamid, O.; Robert, C.; Daud, A.; Hodi, F.S.; Hwu, W.-J.; Kefford, R.; Wolchok, J.D.; Hersey, P.; Joseph, R.W.; Weber, J.S.; et al. Safety and Tumor Responses with Lambrolizumab (Anti-PD-1) in Melanoma. N. Engl. J. Med. 2013, 369, 134–144. [Google Scholar] [CrossRef] [PubMed]
  123. Topalian, S.L.; Hodi, F.S.; Brahmer, J.R.; Gettinger, S.N.; Smith, D.C.; McDermott, D.F.; Powderly, J.D.; Carvajal, R.D.; Sosman, J.A.; Atkins, M.B.; et al. Safety, Activity, and Immune Correlates of Anti-PD-1 Antibody in Cancer. N. Engl. J. Med. 2012, 366, 2443–2454. [Google Scholar] [CrossRef] [PubMed]
  124. Goodman, A.M.; Kato, S.; Bazhenova, L.; Patel, S.P.; Frampton, G.M.; Miller, V.; Stephens, P.J.; Daniels, G.A.; Kurzrock, R. Tumor Mutational Burden as an Independent Predictor of Response to Immunotherapy in Diverse Cancers. Mol. Cancer Ther. 2017, 16, 2598–2608. [Google Scholar] [CrossRef] [PubMed]
  125. Patterson, A.; Auslander, N. Mutated Processes Predict Immune Checkpoint Inhibitor Therapy Benefit in Metastatic Melanoma. Nat. Commun. 2022, 13, 5151. [Google Scholar] [CrossRef] [PubMed]
  126. Gandini, S.; Massi, D.; Mandalà, M. PD-L1 Expression in Cancer Patients Receiving Anti PD-1/PD-L1 Antibodies: A Systematic Review and Meta-Analysis. Crit. Rev. Oncol. Hematol. 2016, 100, 88–98. [Google Scholar] [CrossRef]
  127. Kaunitz, G.J.; Cottrell, T.R.; Lilo, M.; Muthappan, V.; Esandrio, J.; Berry, S.; Xu, H.; Ogurtsova, A.; Anders, R.A.; Fischer, A.H.; et al. Melanoma Subtypes Demonstrate Distinct PD-L1 Expression Profiles. Lab. Investig. 2017, 97, 1063–1071. [Google Scholar] [CrossRef]
  128. Danilova, L.; Wang, H.; Sunshine, J.; Kaunitz, G.J.; Cottrell, T.R.; Xu, H.; Esandrio, J.; Anders, R.A.; Cope, L.; Pardoll, D.M.; et al. Association of PD-1/PD-L Axis Expression with Cytolytic Activity, Mutational Load, and Prognosis in Melanoma and Other Solid Tumors. Proc. Natl. Acad. Sci. USA 2016, 113, E7769–E7777. [Google Scholar] [CrossRef] [PubMed]
Table 1. Frequently altered pathways and genes in melanoma.
Table 1. Frequently altered pathways and genes in melanoma.
PathwaysGenes Related to MelanomaFunctionStage of Involvement in Melanoma
MAPKBRAF, RAS, NF1Cell cycle proliferation regulationEarly initiation
TelomeraseTERT promoterGene stabilization Early initiation
Cell-cycleRB1, CDKN2ACell cycle regulationThin, intermediate melanoma
Apoptosis MDM2, TP53Cell apoptosisThick melanoma
PTEN/PI3K/AKTPTEN, PI3KCell survival and proliferation Thick melanoma
MITFMITFPigmentation, Melanocyte development and differentiationThick, advanced melanoma
Table 2. Frequency of gene alterations in common melanoma subtypes.
Table 2. Frequency of gene alterations in common melanoma subtypes.
GenesUV-Related
Melanoma
Acral MelanomaMucosal
Melanoma
Uveal Melanoma
BRAF~50% [10,13,14]10–34.4% [20,21,22,23]3.1–16.4%
Mutations other than p.v600 codon [27,28,29]
Rare
[32,33,34]
N/H/K-RAS20–28% [10,16]9–21.9% [22]6.2–17.9%
[27,28,29]
Rare
[10]
NF114% [10]11–23% [22]7.8–16.4%
[27,28,29]
Rare
[32,33,34]
KIT1.8% [10]10.9–24.4% [15,20,21,22,23]19.1–23.1%
[27,28,29]
~9% [36]
PTEN/PI3K/AKT~40–63% [10,37]Rare [12]Rare [12]59%
[38]
TERT>60%–86% [10,12]9–41% [21,39]30% [29,40]Rare [41,42]
GNAAQ/GNA110.9% [10]18.6% [15,20,21,22,23]~9.5% [30]89–92.5% [32,33,34,35]
Other genetic alterations--SF3B1 (8.1–11.9%), SPRED1 (4.0–7.5%), ATRX (6%)
[28,29]
BAP1 (45%), SF3B1 (24%),
Most significantly mutated genes are not identified in other melanoma subtypes
[32,33,34]
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

Yang, T.-T.; Yu, S.; Ke, C.-L.K.; Cheng, S.-T. The Genomic Landscape of Melanoma and Its Therapeutic Implications. Genes 2023, 14, 1021. https://doi.org/10.3390/genes14051021

AMA Style

Yang T-T, Yu S, Ke C-LK, Cheng S-T. The Genomic Landscape of Melanoma and Its Therapeutic Implications. Genes. 2023; 14(5):1021. https://doi.org/10.3390/genes14051021

Chicago/Turabian Style

Yang, Ting-Ting, Sebastian Yu, Chiao-Li Khale Ke, and Shih-Tsung Cheng. 2023. "The Genomic Landscape of Melanoma and Its Therapeutic Implications" Genes 14, no. 5: 1021. https://doi.org/10.3390/genes14051021

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