Next Article in Journal
Myeloid-Derived Suppressor Cells: Therapeutic Target for Gastrointestinal Cancers
Next Article in Special Issue
Multi-System-Level Analysis Reveals Differential Expression of Stress Response-Associated Genes in Inflammatory Solar Lentigo
Previous Article in Journal
Bioinformatic Analysis of Sulfotransferases from an Unexplored Gut Microbe, Sutterella wadsworthensis 3_1_45B: Possible Roles towards Detoxification via Sulfonation by Members of the Human Gut Microbiome
Previous Article in Special Issue
The Biology and Genomics of Human Hair Follicles: A Focus on Androgenetic Alopecia
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Molecular Frontiers in Melanoma: Pathogenesis, Diagnosis, and Therapeutic Advances

by
Hyun Jee Kim
1 and
Yeong Ho Kim
2,*
1
Department of Dermatology, International St. Mary’s Hospital, College of Medicine, Catholic Kwandong University, Incheon 22711, Republic of Korea
2
Department of Dermatology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(5), 2984; https://doi.org/10.3390/ijms25052984
Submission received: 2 January 2024 / Revised: 1 March 2024 / Accepted: 2 March 2024 / Published: 4 March 2024
(This article belongs to the Special Issue Molecular Research Progress of Skin and Skin Diseases)

Abstract

:
Melanoma, a highly aggressive skin cancer, is characterized by rapid progression and high mortality. Recent advances in molecular pathogenesis have shed light on genetic and epigenetic changes that drive melanoma development. This review provides an overview of these developments, focusing on molecular mechanisms in melanoma genesis. It highlights how mutations, particularly in the BRAF, NRAS, c-KIT, and GNAQ/GNA11 genes, affect critical signaling pathways. The evolution of diagnostic techniques, such as genomics, transcriptomics, liquid biopsies, and molecular biomarkers for early detection and prognosis, is also discussed. The therapeutic landscape has transformed with targeted therapies and immunotherapies, improving patient outcomes. This paper examines the efficacy, challenges, and prospects of these treatments, including recent clinical trials and emerging strategies. The potential of novel treatment strategies, including neoantigen vaccines, adoptive cell transfer, microbiome interactions, and nanoparticle-based combination therapy, is explored. These advances emphasize the challenges of therapy resistance and the importance of personalized medicine. This review underlines the necessity for evidence-based therapy selection in managing the increasing global incidence of melanoma.

1. Introduction

Melanoma, the deadliest form of skin cancer, presents a significant clinical challenge due to its high metastatic potential and resistance to conventional therapies. It originates from melanocytes and is increasingly prevalent [1].
Melanoma’s incidence surged by 320% from 1975 to 2018, influenced by risk factors such as sun exposure, indoor tanning, family history, and the number of nevi [2]. This type of skin cancer is responsible for nearly 90% of skin cancer deaths, despite constituting a small fraction of skin cancer cases [3]. The disease impacts both older and younger populations, though the increase in incidence is particularly notable among older individuals [3]. The primary cause of death from melanoma is metastatic spread, first to the lymph nodes and, most commonly, to the lungs [3]. Early-stage melanoma (I–II) can be effectively treated with complete surgical excision, boasting an excellent 5-year survival rate of 99.4% [2]. However, the prognosis worsens significantly in advanced stages, with 5-year survival rates dropping to 68% for stage III and 29.8% for stage IV melanoma [2].
The clinical burden of melanoma is growing alongside its rising global incidence, which has been increasing by approximately 3% annually in certain regions [4]. Projections for 2023 estimate 97,620 new cases and 7990 deaths in the United States alone [4]. Meanwhile, the International Agency for Research on Cancer reported an estimated 324,635 new diagnoses and 57,043 deaths from melanoma globally in 2020 [4]. These statistics highlight the critical need for the rational and evidence-based selection of therapies in the treatment of melanoma.
This review focuses on the molecular pathogenesis of melanoma, underlining recent advances in understanding the disease’s genetic and epigenetic landscape. Key genetic drivers central to melanoma development, such as mutations in the BRAF, NRAS, and c-KIT genes, are discussed alongside the latest WHO classification, which categorizes melanomas based on cumulative sun damage (CSD), which correlates with specific molecular alterations. This classification aids in understanding the disease’s molecular diversity [5].
Advances in genomic technologies, especially single-cell sequencing, have significantly improved the characterization of melanoma’s gene signatures and phenotypic subtypes, which are crucial for understanding its aggressiveness and high mortality rate [6].
Despite the high survival rate for localized melanoma, the prognosis for metastatic melanoma remains poor, highlighting the need for effective therapies [2]. The introduction of ipilimumab in 2011 marked a turning point in melanoma treatment [4]. Since then, numerous new drugs for unresectable melanoma have been approved, transforming the therapeutic landscape and improving overall survival (OS) times. This review consolidates recent findings in molecular pathology, diagnostics, and therapeutic strategies for melanoma, emphasizing the challenges of therapy resistance and the potential of personalized medicine and evidence-based therapy selection. Those approaches are essential, given the increasing global incidence and clinical burden of melanoma.

2. Molecular Pathology of Melanoma

The progression from benign melanocytic nevi to malignant melanoma and metastasis involves an interplay of genetic factors and UV-induced damage. Melanocytic nevi, typically benign, can evolve into melanoma through mutations, primarily BRAFV600E in common nevi and various mutations in MAPK signaling, the TERT promoter, and CDKN2A in dysplastic nevi [7]. The progression is marked by additional mutations, such as in NRAS, and is influenced by the activation of the WNT signaling pathway, which is important for metastasis. Genes such as ARID2 and ARID1A are also implicated in melanoma’s progression [7].

2.1. WHO Classification and Molecular Diversity of Melanoma

The 2018 WHO classification of melanocytic lesions provides a refined understanding of melanoma’s molecular diversity, categorizing melanomas based on CSD. This system divides melanomas into:
1. Low-CSD melanomas: these include superficial spreading melanomas, typically associated with less sun damage. They often arise on the trunk and proximal areas of the extremities and primarily feature BRAFV600E mutations. Other mutations in these melanomas include the TERT promoter and CDKN2A, with PTEN and TP53 mutations observed in more advanced stages [5].
2. High-CSD melanomas: comprising lentigo maligna and desmoplastic melanomas, these usually develop on heavily sun-damaged skin, particularly in older individuals. They can have a high mutation load, including NRAS, BRAF non-V600E, or NF1 mutations, and they frequently present TERT promoter mutations, CDKN2A, and occasionally KIT mutations. The mutation count in these melanomas increases with the degree of CSD, with desmoplastic melanomas showing the highest tumor mutation burden [5].
3. Non-CSD-associated melanomas: this category contains Spitz melanomas, acral melanomas, mucosal melanomas, melanomas arising from congenital or blue nevi, and uveal melanomas. These subtypes are typically devoid of BRAF, NRAS, and NF1 mutations (triple wild-type), but can feature KIT mutations, gene amplifications, and structural rearrangements, especially of the CCND1 gene and SF3B1. Acral and mucosal melanomas are biologically distinct from their cutaneous counterparts in sun-exposed areas. Spitz melanomas show tyrosine kinase or serine-threonine kinase fusions, and melanomas in blue nevi and uveal melanomas often contain GNA11 or GNAQ mutations [5].
This classification not only segregates melanomas based on CSD, but also correlates the types with specific molecular alterations, providing crucial insights into the varied molecular pathways and risk factors associated with different subtypes of melanoma.
In addition to these classifications, WHO has introduced the concept of „intermediate” lesions in its latest melanocytic tumor classification, acknowledging the diagnostic challenges of melanocytic tumors. This approach shifts away from viewing melanocytic tumors as simply benign or malignant, suggesting a more nuanced understanding by providing nine categories/pathways, each marked by specific genetic drivers [5].

2.2. Key Genetic Mutations in Melanoma

2.2.1. BRAF Mutations

BRAF mutations are found in approximately 50% of melanomas and play a crucial role in the disease’s pathogenesis (Figure 1). These mutations are predominantly observed in cutaneous melanomas and are frequently associated with UV radiation exposure. This link is evidenced by the high levels of UV radiation signatures, especially C > T substitutions, found in these tumors [8]. Notably, patients with BRAF mutations tend to be younger than those with other melanoma subtypes [8]. The mutation patterns related to UV exposure highlight the interplay between environmental factors and the genetic landscape of melanoma, emphasizing the importance of considering both of them in understanding melanoma development.
The presence of BRAF mutations, particularly V600E, leads to the activation of the MAPK/ERK signaling pathway, a key driver of cell proliferation and survival in melanoma. This understanding has led to the development of targeted therapies such as vemurafenib and dabrafenib, which specifically inhibit the BRAFV600E mutation and have shown significant efficacy in treating patients harboring this genetic alteration.
In clinical practice, the detection of BRAF mutations is a critical factor in treatment decisions, particularly for metastatic melanoma. Testing for the BRAFV600 mutation is recommended for patients with distant metastases, non-resectable regional metastases, or high-risk stage III melanoma post-surgery [9]. Performing this test on metastatic tissue samples is ideal, due to the high concordance of BRAF mutation status in primary and metastatic lesions. The result of this testing significantly informs therapeutic decision-making, underscoring the role of genetic profiling in the personalized treatment of melanoma.

2.2.2. NRAS Mutations

NRAS mutations, which are found in 15–20% of melanoma cases, are significant drivers of the disease, affecting melanoma development through a distinct pathway [9]. These mutations activate the MAPK pathway, albeit through a mechanism different from that of BRAF mutations. NRAS mutations are typically mutually exclusive of BRAF mutations, reinforcing their unique role in melanoma’s molecular pathology [9]. The identification of NRAS mutations is not only essential for understanding melanoma’s genetic diversity, but also plays an important role in clinical decision-making, particularly in cases without BRAF mutations. Despite not being direct targets of current therapies, the presence of NRAS mutations guides the selection and tailoring of treatment strategies. So far, targeted therapies specifically addressing NRAS-mutated melanoma have shown limited success [5]. However, this area remains a significant focus of ongoing research, especially in the context of advanced melanoma that has not responded to standard immunotherapies, including anti-CTLA4 and anti-PD1 antibodies. The development of more effective treatment options for NRAS-mutated melanoma is a key objective of current clinical investigations, reflecting the continuous effort to improve therapeutic outcomes for this challenging subset of melanoma patients.

2.2.3. c-KIT Mutations

c-KIT mutations, while less common than BRAF and NRAS mutations, play a significant role in certain melanoma subtypes, particularly mucosal and acral melanomas. These mutations are predominantly found in melanomas arising from mucosal, acral, and chronically sun-damaged skin, representing a distinct subset within the broader spectrum of melanoma [9].
In the clinical setting, it is recommended to initially test for BRAF and NRAS mutations in acral and mucosal melanomas [5]. If those tests return negative, a further analysis for c-KIT mutations is advised [5]. This stepwise approach to genetic testing ensures a precise and targeted strategy for managing these specific melanoma subtypes.
Therapies targeting c-KIT mutations, although not yet formally approved, have shown promising results in treating melanomas harboring these genetic alterations. Clinical benefits from c-KIT inhibitors have been observed in selected patients, underscoring the importance of these mutations in the therapeutic landscape of melanoma [9]. The ongoing research and development of treatments targeting c-KIT mutations are pivotal in enhancing care and outcomes for patients with these specific melanoma subtypes.

2.2.4. GNAQ/GNA11 Mutations

GNAQ/GNA11 mutations, commonly identified in uveal melanomas [10], represent a distinct genetic subgroup within melanoma. Despite the current limitations of available treatments, these mutations are under active study for targeted therapy options, reflecting a growing interest in developing specific treatments for these subtypes.
In non-uveal melanomas, GNAQ/GNA11 mutations display a unique genetic profile characterized by a lower tumor mutational burden and fewer UV signature mutations than are common in cutaneous melanomas [11]. This suggests significant differences in the genetic landscape of these tumors compared with both cutaneous and uveal melanomas. Additionally, non-uveal melanomas with GNAQ/GNA11 mutations tend to metastasize lymphatically, similar to cutaneous melanoma, rather than the hematogenous metastasis typically seen in uveal melanoma [11]. These findings underline the urgent need for novel therapeutic approaches because non-uveal melanomas with GNAQ/GNA11 mutations respond poorly to existing systemic therapies, including immune checkpoint inhibitors (ICIs) [11]. The rarity and distinct behavior of GNAQ/GNA11 mutant non-uveal melanomas highlight the importance of ongoing research to develop effective treatments for this unique melanoma subgroup.

2.3. Molecular Pathways

2.3.1. MAPK/ERK Pathway

The MAPK/ERK pathway is central to melanoma, with alterations in this pathway often driving tumorigenesis [9]. This pathway, which includes RAS, RAF, ERK, and mitogen-activated extracellular signal-regulated kinase (MEK), is crucial for regulating cellular proliferation [2]. The discovery of activating NRAS mutations in melanoma in the mid-1980s and the subsequent identification of BRAF mutations in 2002 have been significant milestones in understanding melanoma’s molecular pathology [12]. Both of these mutations lead to the overactivation of the MAPK/ERK pathway, promoting melanoma cell proliferation and survival.
The exploration of the MAP kinase pathway as a therapeutic target began with responses observed from MEK inhibitors. However, the development of potent and selective inhibitors such as vemurafenib, dabrafenib, and encorafenib, which target mutated BRAF, has been the major breakthrough in melanoma treatment [12]. The late 2000s saw a pivotal shift with the introduction of these targeted therapies. The initial use of drugs such as sorafenib paved the way for more effective and selective BRAF inhibitors [12].
Combination therapies of BRAF/MEK inhibitors have significantly improved the efficacy of melanoma treatments, with reduced toxicity and longer progression-free survival (PFS) times compared with monotherapies [12]. These advances highlight the significance of the MAPK/ERK pathway in melanoma’s molecular pathology and response to therapy, underscoring the importance of understanding the influence of these mutations when developing effective treatments.

2.3.2. PI3K/AKT/mTOR Pathway

The PI3K/AKT/mTOR pathway plays a central role in melanoma development, affecting cell survival, proliferation, and metastasis. Its activation, often driven by genetic mutations and signaling imbalances, underscores the need for targeted therapeutic interventions. The constitutive activation of this pathway is a hallmark of melanoma’s aggressiveness and contributes to resistance against standard treatments by influencing key cellular processes such as autophagic cell death and cell cycle regulation [13].
A range of targeted therapies focusing on inhibitors of PI3K, AKT, and mTOR is under investigation and showing promising results in clinical trials, both as individual treatments and in combination with other drugs, such as BRAF and MEK inhibitors [13]. The exploration of natural compounds, repurposed drugs, and novel synthetic molecules targeting this pathway, along with the emerging role of miRNA in modulating this pathway, presents new opportunities for treatment and underscores the pathway’s significance for the development of novel, effective, and personalized melanoma therapies [13].

2.4. Melanogenesis and Neuroendocrine Regulation in Melanoma Progression

Recent studies have highlighted the complex roles of melanin and melanogenesis in melanoma, revealing their protective effects against UV radiation and their potential to promote malignant transformation [14]. The synthesis of eumelanin and pheomelanin, influenced by environmental and hormonal factors, offers both defense and risks, with the instability of pheomelanin contributing to a mutagenic environment [15,16,17,18,19,20,21,22,23,24,25]. This duality affects melanoma’s development and response to treatments, with advanced melanomas showing a negative correlation between pigmentation and survival, indicating melanin’s dual impact [26,27,28,29]. Inhibiting melanogenesis could, therefore, enhance therapeutic outcomes, highlighting melanin’s intricate influence on melanoma behavior.
Moreover, melanoma’s ability to influence both local and systemic physiological responses through the secretion of neuroendocrine factors, including proopiomelanocortin (POMC) peptides, corticotropin-releasing hormone (CRH), and glucocorticoids, underscores its complex role in the body’s regulatory systems [30]. POMC peptides, including melanocyte-stimulating hormone (MSH), are immunosuppressive, and an increased expression of POMC peptides was noted during the progression of melanomas to advanced stages [31,32,33,34,35,36,37,38,39]. This capability of melanoma to manipulate the neuroendocrine and immune responses contributes to its survival and progression, altering homeostasis in favor of the tumor [30]. Such interactions necessitate the exploration of therapeutic strategies aimed at targeting these pathways, offering potential to improve treatment outcomes and patient prognosis.

3. Molecular Diagnostic Techniques

3.1. Latest Molecular Diagnostic Methods

Molecular diagnostics for melanoma have made significant strides, largely due to advances in genomics, transcriptomics, and emerging techniques such as liquid biopsies.
Genomic analyses, particularly next-generation sequencing (NGS), have been essential in identifying specific mutations and genomic alterations in melanoma. NGS allows for the comprehensive screening of multiple genes associated with melanoma in a single experiment, enabling the efficient identification of mutations in key genes such as BRAF, NRAS, and c-KIT [9]. Although NGS is currently more prevalent as a research tool, its importance in the diagnostic setting is expected to increase, especially as more actionable mutations are identified and targeted therapies become available.
Recent developments in single-cell transcriptomics have made it possible to analyze the tumor microenvironment in melanoma in detail. This technique elucidates cellular behaviors and interactions within tumors, clarifying melanoma progression and suggesting how it might respond to treatments. By identifying various cellular subtypes and their transcriptional states, it enhances our understanding of tumor heterogeneity, which is useful in assessing treatment responses, especially in the advanced stages of melanoma [40]. Furthermore, this approach helps explain the mechanisms of response and resistance to therapies such as ICIs [40].
Additionally, liquid biopsies are emerging as a valuable non-invasive diagnostic tool. They detect circulating tumor DNA in the blood and offer insights into tumor genetics. This minimally invasive method can monitor treatment responses and disease progression, complementing traditional diagnostic techniques [9].

3.2. Molecular Biomarkers Used for Diagnosing and Prognosticating Melanoma

Biomarkers in melanoma encompass a wide range of indicators, from serum proteins to genetic alterations, pathology findings, and imaging results. The use of molecular biomarkers is increasingly important in the early diagnosis, staging, and prediction of therapy responses in melanoma (Table 1).
One well-known driver mutation is BRAFV600E, which is responsive to BRAF inhibitors [41,42,43]. In contrast, NRAS mutations, although less common, are linked to shorter survival times in stage IV melanoma [44,45,46]. Unlike BRAF and NRAS mutations, c-KIT mutations are not closely associated with histological subtypes or tumor stage. They are more prevalent in older patients, acral mucosal melanoma subtypes, and areas with chronic sun-induced damage [47,48,49,50,51]. Furthermore, melanomas with neurofibromin 1 (NF1) mutations are associated with advanced patient age and have a poorer prognosis compared to other mutation patterns [52,53]. On the other hand, female patients with high plasma membrane calcium-transporting ATPase 4 (PMCA4) transcript levels exhibit longer PFS than other patients [54]. High PMCA4 transcript levels in cutaneous melanoma are also associated with an improved prognosis, especially following PD-1 blockade therapy [54].
The tumor mutational burden (TMB) in melanoma is gaining attention for its potential to predict the response to ICIs. A high TMB might correlate with an increased effectiveness of these treatments, suggesting a potential for enhanced tumor recognition and elimination by the immune system [55,56,57].
Gene expression profiling (GEP), exemplified by tests such as the 31-GEP panel Decision-Dx Melanoma™, is being used to assess melanoma recurrence and metastatic risk. These tests examine the expression patterns of selected genes in the primary tumor, contributing to informed clinical decision-making [58,59].
Recent research underscores that the gut microbiome can influence carcinogenesis by producing pro- or anti-tumor inflammatory environments. Studies in melanoma indicate that the intestinal microbiota composition affects the response to ICI therapy [60]. Greater microbiota diversity, particularly certain Ruminococcaceae subspecies, is associated with better anti-PD-1 therapy outcomes and increased CD8+ T cell infiltration, and specific Bacteroidetes species might reduce the risk of ICI-induced colitis [61].

4. Molecular Therapeutic Strategies

The increase in survival rates seen since the advent of BRAF-MEK inhibitors and immunotherapy signifies a major breakthrough in melanoma therapeutics. These advances have not only improved outcomes, but also reshaped approaches to molecular strategies in melanoma treatment. Additionally, the evolving role of adjuvant therapy in melanoma, particularly with respect to patient selection, underscores the growing importance of personalized medicine in optimizing therapeutic efficacy and minimizing adverse effects. Furthermore, novel approaches to treatment strategies, such as neoantigen vaccines, adoptive cell transfer, and microbiome research, are expanding the horizon of melanoma management, offering new avenues for treatment (Table 2).

4.1. Targeted Therapy

4.1.1. BRAF Inhibitors

In the context of treating BRAFV600E mutation-positive melanoma, vemurafenib and dabrafenib have shown significant efficacy, leading to significant improvements in PFS and OS for patients with BRAF-mutated melanoma [62]. These drugs target the mutated BRAF protein, effectively disrupting the MAPK/ERK signaling pathway essential for tumor growth. Furthermore, vemurafenib, the first BRAF inhibitor approved by the FDA in 2011 for metastatic melanoma with the BRAFV600E mutation, has demonstrated an increase in OS compared with dacarbazine in a phase III (BRIM-3) trial [63,64,65,66].

4.1.2. MEK Inhibitors

MEK inhibitors, which target elements downstream of BRAF in the MAPK signaling pathway, are an important therapeutic option for melanoma treatment. Trametinib, which specifically targets MEK1 and MEK2, has been approved to treat metastatic or unresectable melanoma with BRAF V600E or V600K mutations and as adjuvant therapy [67]. The METRIC study highlighted trametinib’s effectiveness, compared with chemotherapy, in metastatic melanoma, as well as in combination with dabrafenib in brain metastases or adjuvant therapy (COMBI-d and COMBI-AD studies, respectively) [68,69,70,71].

4.1.3. BRAF-MEK Combination Therapy

Combination therapy to block multiple points in the MAPK pathway for melanoma treatment is designed to reduce the risk of resistance development. Clinical trials such as coBRIM and COMBI-d have shown enhanced efficacy with this approach, with increased PFS and a higher overall response rate compared with monotherapy [72,73,74,75,76]. The combination of BRAF and MEK inhibitors (vemurafenib and cobimetinib) was approved in 2015 for treatment of patients with unresectable or metastatic melanoma who harbor a BRAF V600E or V600K mutation [77].

4.2. Immunotherapy

Advances in understanding the molecular pathways involved in melanoma have led to the development of successful immunotherapies for unresectable stage III and IV melanoma. These include ICIs that target PD-1 and CTLA-4, which have revolutionized treatment and prognosis in the past decade [5].

4.2.1. Checkpoint Inhibitors

Both CTLA-4 and PD-1 inhibitors are the two types of checkpoint inhibitors currently available to melanoma patients. Ipilimumab, a CTLA-4 inhibitor, enhances T-cell activity by inhibiting the immunosuppressive interaction between CTLA-4 and B7 [2]. Since its approval in 2011, this immunotherapy has been associated with improved OS rates, particularly when used in conjunction with dacarbazine [78].
PD-1 inhibitors (pembrolizumab and nivolumab) are currently approved for the treatment of metastatic or unresectable melanoma and as adjuvant therapy [67]. These drugs enhance the immune system’s ability to recognize and attack cancer cells by blocking the PD-1 receptor on T cells [67]. Pembrolizumab has been reported to surpass the performance of ipilimumab for the treatment of untreated metastatic or unresectable melanoma (KEYNOTE-006 study) and chemotherapy in patients with previously treated metastatic or unresectable disease (KEYNOTE-002 study) [79,80]. Nivolumab has shown effectiveness both as a monotherapy and in combination with ipilimumab, as evidenced in the CHECKMATE-037, CHECKMATE-066, and CHECKMATE-067 studies [81,82,83,84]. The CHECKMATE-067 trial demonstrated that the combination of CTLA-4 and PD-1 inhibitors (ipilimumab and nivolumab) offers a significant survival advantage in advanced melanoma patients [84,85,86]. This combination strategy enhances the overall response rates and PFS by targeting multiple points in the immune response cascade. Although it enhanced survival outcomes, it is important to note that the combination can also increase the occurrence of adverse effects.

4.2.2. Talimogene Laherparepvec (T-VEC)

T-VEC (IMLYGIC®), the first viral oncolytic immunotherapy approved for unresectable metastatic stage IIIB/C–IVM1a melanoma, represents a notable advance in melanoma treatment [87]. This genetically modified herpes simplex type 1 virus is directly injected into tumors, triggering both local and systemic immune responses that lead to tumor cell destruction and the activation of tumor-specific T cells [88].
Clinical trials have demonstrated T-VEC’s effectiveness both as a monotherapy [89,90,91] and in combination with ICIs such as ipilimumab and pembrolizumab [92,93,94], showcasing its ability to enhance local and systemic anti-tumor responses. T-VEC has shown promising efficacy and tolerable side effects in treating both injected and non-injected melanoma lesions, although its systemic effect as a single-agent therapy is relatively modest [89,90,91]. T-VEC’s innovative method for inducing a comprehensive immune response against melanoma cells, along with its synergistic potential with other immunotherapies, makes it a valuable melanoma treatment option.

4.3. Combination of Targeted Therapy and Immunotherapy

Current research efforts are focusing on integrating BRAF and MEK inhibitors, types of targeted therapies, with immunotherapy agents. The rationale behind this strategy is to combine the direct action of the targeted therapy, which addresses specific genetic mutations in melanoma cells, with the broad-acting capacity of immunotherapy to bolster the immune system’s cancer-fighting abilities. The goal is to potentially increase treatment effectiveness and counteract drug resistance.
One example of such research is a clinical trial investigating the combination of dabrafenib (a BRAF inhibitor) and trametinib (a MEK inhibitor) with pembrolizumab, an anti-PD-1 therapy [95,96]. This trial is particularly focused on evaluating the synergistic effects of these treatments in patients with advanced melanoma. Additionally, the SECOMBIT trial demonstrated that sequential immunotherapy (nivolumab plus ipilimumab) and targeted therapy (encorafenib plus binimetinib) provide clinically significant survival benefits in patients with BRAF-mutant melanoma [97].

4.4. Integration of Surgical and Systemic Therapies

The integration of surgical interventions and systemic therapies, including pre-surgical (neoadjuvant) and post-surgical (adjuvant) treatments, is an area of active research. This multidisciplinary approach aims to improve OS rates, reduce recurrence, and manage metastatic disease more effectively.
Adjuvant therapy plays a crucial role in managing melanoma, affecting both OS and recurrence-free survival times. Although interferon-α was previously a common choice, it has been replaced by safer and more effective options, such as ICIs and targeted therapies. Patients with lymph node involvement often receive BRAF/MEK inhibitors if they have the BRAFV600 E/K mutation. Anti-PD-1 therapies, including pembrolizumab, are also an option regardless of mutation status [67].
Pembrolizumab is FDA-approved as adjuvant therapy for the treatment of stage IIB/IIC and III melanoma [98]. The KEYNOTE-716 trial demonstrated that pembrolizumab has a significant and durable impact on distant-metastasis-free survival and recurrence-free survival in resected stage IIB or IIC melanoma patients [99]. Moreover, the KEYNOTE-942 trial introduced a combination of the investigational mRNA-4157/V940 vaccine and pembrolizumab and found that it shows potential in reducing the risk of disease recurrence after surgery [100].
In addition, Schumann K and colleagues have shown the effectiveness of other adjuvant therapies, such as nivolumab and the combination of dabrafenib and trametinib (D + T), in a broader range of patients than is typically included in clinical trials [101]. This suggests the applicability of clinical trial results to everyday clinical practice and highlights the importance of evaluating these therapies in diverse patient groups.
Additionally, the CHECKMATE-238 trial is examining the long-term effects of adjuvant nivolumab compared with ipilimumab in patients with resected stage III/IV melanoma [102]. This contributes to the growing understanding of effective adjuvant treatments in melanoma.

4.5. Novel Approaches to Treatment Strategies

4.5.1. Neoantigen Vaccines

Renewed interest in cancer vaccines, particularly for melanoma, has focused on personalized neoantigen vaccines. These vaccines are tailored to individual patients and target unique tumor-specific antigens that arise from mutations. Clinical trials of neoantigen vaccines in melanoma have shown them to be safe and capable of inducing specific immune responses against these unique tumor antigens [103,104,105,106,107]. These vaccines are designed based on individual tumor mutations identified through advanced sequencing technologies.
Ongoing clinical trials are testing various vaccine platforms, including dendritic cells, viral vectors, RNA, and peptides [103,104,105,106,107]. The aim is to evaluate the clinical effectiveness of these vaccines, especially in combination with other immunotherapies such as ICIs. This combination strategy is expected to enhance the immune system’s response to melanoma.

4.5.2. Adoptive Cell Transfer

Adoptive cell transfer, especially tumor-infiltrating lymphocyte (TIL) therapy, has emerged as a noteworthy strategy in the management of melanoma, offering a personalized approach to cancer treatment. TIL therapy involves extracting immune cells from a patient’s tumor, expanding and enhancing these cells in a laboratory, and then reintroducing them into the patient to bolster the immune system’s ability to fight cancer. This method leverages the unique ability of TILs to recognize and target tumor-specific antigens, making it a potent form of immunotherapy [108,109,110].
Clinical trials have underscored the efficacy of TIL therapy in patients with metastatic melanoma, particularly those unresponsive to conventional treatments. Approximately 50% of patients achieved a partial response, while around 20% achieved a complete response, showcasing the potential of TIL therapy as a personalized form of immunotherapy [108,111]. Furthermore, patients treated with TIL therapy demonstrated an increase in median OS by 6 to 12 months compared to patients receiving standard treatments, highlighting the promise of TIL therapy in extending the lives of patients with advanced melanoma.
However, challenges remain, including the variability in response rates among patients and the logistical complexities involved in cell extraction and expansion. Moreover, the therapy’s side effects, such as cytokine release syndrome, necessitate careful patient monitoring [112]. Future research is directed towards enhancing the efficacy of TIL therapy through combination treatments, improving patient selection criteria, and understanding the genomic correlates of response to therapy [113,114].

4.5.3. Microbiome and Melanoma Treatment Interactions

Recent research has brought to light the important role of the gut microbiome in the development and treatment of melanoma [115]. The gut microbiome, a complex ecosystem of microorganisms located primarily in the intestinal tract, influences various aspects of health, including immune system regulation and inflammation prevention [116,117].
The skin microbiome’s involvement in melanoma is gaining attention, with specific bacterial compositions linked to melanoma, such as an abundance of Fusobacterium and Trueperella genera [118]. Additionally, certain bacteria like Corynebacterium are associated with acral melanoma progression [119]. The presence of Staphylococcus aureus in other skin cancers suggests broader implications of skin microbiota in skin pathologies [120]. Probiotic and prebiotic applications show potential in mitigating UVR-induced skin damage and modulating the tumor immune microenvironment, indicating a promising, but yet to be clinically validated, approach to melanoma treatment and prevention [120,121].
The interplay between genetic mutations and the microbiome is highlighted in melanomagenesis, with BRAF mutations possibly influencing gut microbiota composition. This relationship, explored in colorectal cancer, suggests the potential for microbiota profiles as biomarkers and therapeutic targets in BRAF-mutated cancers, though evidence in melanoma is limited [122,123].
Immunotherapy has revolutionized melanoma treatment, with research focusing on microbiota’s role in modulating responses to ICIs [124,125,126]. Specific gut bacteria like Akkermansia muciniphila and Bifidobacterium species are associated with positive ICI responses, with studies exploring fecal microbiota transplantation to enhance immunotherapy efficacy [124,125,126]. Conversely, the microbiome may influence ICI-related adverse effects, with certain microbial profiles linked to reduced toxicity, suggesting the potential to tailor treatment and manage side effects through microbiota manipulation [127,128].
Ongoing clinical trials in the UK and the US are investigating the relationship between gut microbiota and melanoma treatment outcomes, focusing on immunotherapy efficacy and side effects [97,98,99]. Additionally, the field of molecular pathological epidemiology (MPE) integrates various disciplines to understand tumor–environment–host interactions, offering insights into melanoma pathogenesis and treatment [100,101,102,103]. Challenges such as validating molecular assays and sample size limitations persist [103].
Clinical trials in the UK and the US are investigating the relationship between gut microbiota and melanoma treatment, with a focus on immunotherapy efficacy and side effects. In the UK, one study is assessing gut microbiota diversity’s impact on immunotherapy outcomes and adverse reactions in stage 3 and 4 melanoma patients [129]. Another initiative aims to determine gut microbiota’s potential as biomarkers for immunotherapy effectiveness and toxicity in patients with unresectable stage 3 and 4 melanoma [130]. Meanwhile, in the US, researchers are exploring the effectiveness of fecal microbiota transplants combined with Pembrolizumab for treating PD-1 resistant/refractory melanoma, representing a significant advancement in melanoma treatment [131].
The interaction between lifestyle, environmental factors, diet, and other external influences on the genomic and metabolomic profiles of cells, including immune cells, is increasingly recognized [132]. Molecular pathological epidemiology (MPE) integrates epidemiology, bioinformatics, and biostatistics to understand tumor–environment–host interactions comprehensively [133]. Leveraging cutting-edge technologies like in vivo pathology and artificial intelligence, MPE investigates exogenous factors’ role in gut microbiota and their influence on melanoma pathogenesis and treatment [134]. This integrative approach facilitates exploring etiologic heterogeneity across diseases and establishing causal links between environmental factors and molecular biomarkers [135].

4.5.4. Nanoparticle-Based Combination Therapy for Melanoma

Nanotechnology in melanoma treatment uses nanoparticles (NPs) to target drug delivery to cancer cells. This approach is significant in cancer treatment and also extends to areas such as diagnosis, gene therapy, biomarker creation, targeted therapy, and imaging. The biological properties of NPs allow them to accurately target cancer cells while minimizing the effects on healthy tissues [136].
In chemotherapy, NPs have been explored for their potential to deliver chemotherapeutic agents directly to melanoma cells. This method reduces systemic toxicity and degradation of the drugs. Specifically, carbon nanotubes have been utilized to deliver doxorubicin, a chemotherapeutic agent, directly to melanoma cells [137]. This approach has shown impressive results, with doxorubicin-loaded carbon nanotubes increasing cell death by 90% through inducing a moderate G2-M phase arrest (17.7 ± 1.1%) and significantly reducing tumor size in mice bearing B16–F10 melanoma [137]. Another approach involves the use of chitosan/alginate NPs for the delivery of doxorubicin. This method has been explored for its ability to control the release rate of doxorubicin, achieving better transport and higher intracellular concentration in melanoma cells [138]. The sustained release provided by chitosan/alginate nanoparticles leads to improved accumulation and prolonged cytotoxic effects of encapsulated doxorubicin in melanoma cell lines, showcasing the versatility and potential of nanoparticle-based delivery systems in improving chemotherapy outcomes.
In immunotherapy, nanovaccines like the DGBA-OVA-CpG (Guanidinobenzoic acid-ovalbumin-cytosine-guanine dinucleotides) nanovaccine have shown potential in increasing the generation of cytotoxic T-lymphocytes and reducing melanoma growth when combined with checkpoint inhibitors [139]. This underscores the ability of nanoparticles to modulate the tumor microenvironment and overcome immunosuppression.
In PDT, NPs improve the delivery and efficacy of photosensitizers. NPs increase PSs’ solubility, permeability, and retention in targeted cells, facilitating deeper penetration and reducing treatment resistance by generating high levels of reactive oxygen species (ROS). Studies have shown that NPs can be used to deliver 5-aminolevulinic acid (5-ALA) and other PSs, like phthalocyanine 4 encapsulated in silica NPs, leading to increased apoptosis and reduced tumor survival in melanoma cell lines [140,141]. Additionally, novel approaches like yttrium oxide NPs combined with X-rays have shown potential in increasing ROS production and causing DNA damage, indicating a promising direction for enhancing PDT’s efficacy [142].
Nanoparticle-based therapies offer a promising approach to melanoma treatment, aiming for more precise and effective strategies with minimal harm to healthy tissue. Ongoing research and clinical trials are crucial for moving these innovations from the lab to practical use, ultimately enhancing treatment outcomes for melanoma patients.

4.5.5. DNA Damage Response Inhibitors

Melanoma is characterized by its high mutational burden and frequent mutations in DDR genes, leading to increased DNA damage and replicative stress [143]. DDR inhibitors, including inhibitors of DNA-PKcs, PARP, ATM, CHK1, WEE1, and ATR, have demonstrated promising results in both preclinical and clinical studies to enhance the efficacy of existing therapies, including chemotherapy, targeted therapy, and immunotherapy [143].
For instance, PARP inhibitors, which act through mechanisms like the inhibition of PARP function and “trapping” PARPs on DNA lesions, have shown potential in reducing migration and invasion and inducing apoptosis in melanoma cell lines [144,145,146]. Clinical trials combining PARP inhibitors with chemotherapy, particularly temozolomide, have shown improvement in progression-free survival (PFS), albeit without reaching statistical significance [147,148].
Targeting WEE1 has been shown to increase DNA damage and cell death in melanoma cell lines, suggesting its potential as a therapeutic target [149]. The small molecule inhibitor adavosertib, targeting WEE1, has demonstrated cytotoxic effects alone and in combination with other treatments in melanoma, highlighting its potential for clinical application [150,151,152,153].
The combination of ATR inhibitors with PARP inhibitors, specifically AZD-6738 and olaparib, has been found to effectively target BRAFV600 mutant melanoma cell lines that have developed either primary or acquired resistance to BRAF and MEK inhibitors, demonstrating a high susceptibility to this treatment strategy [154]. Clinical trials, such as those involving ceralasertib, have demonstrated encouraging results, particularly in melanomas resistant to PD-1 inhibitors, suggesting the value of ATR inhibitors in enhancing immune responses and improving outcomes in melanoma treatment [155,156].
Targeting DNA-PKCS, a key component in the DNA repair process, has emerged as a promising strategy for combating resistance to MAPK inhibitors (MAPKi) in melanoma.
In human melanoma cell lines and patient-derived xenografts (PDX), nonhomologous end-joining (NHEJ) targeting by a DNA-PKCS inhibitor prevents/delays acquired MAPKi resistance by reducing the size of ecDNAs and CGRs early in combination treatment [157].
In conclusion, DDR inhibitors represent a promising therapeutic avenue for melanoma treatment, potentially enhancing the effectiveness of current therapies and overcoming resistance. Ongoing and future clinical trials will be crucial in determining their role in melanoma management.

4.5.6. LAG-3 Inhibitors

LAG-3 (lymphocyte-activation gene 3) inhibitors have become a significant development in melanoma immunotherapy, especially when used in combination with PD-1 inhibitors. These inhibitors, now FDA-approved as first-line therapy for metastatic melanoma, demonstrate considerable efficacy and manageable toxicity levels [158]. LAG-3 operates by attaching to MHC class II molecules on antigen-presenting cells, which suppresses TCR signaling and T cell activation, playing a crucial role in the tumor microenvironment to aid melanoma cells in evading immune detection [158].
The collaboration of relatlimab, an anti-LAG-3 antibody, with nivolumab, a PD-1 inhibitor, represents a significant step forward in the treatment of melanoma. Approved in 2022 for treating metastatic or unresectable melanoma, this duo therapy has led to superior PFS outcomes, particularly highlighted in the RELATIVITY-047 trial [159]. This trial showed a PFS of 10.1 months for the combined therapy, as opposed to 4.6 months for nivolumab alone at a median follow-up of 13.2 months, showcasing the effectiveness of LAG-3 inhibition alongside PD-1 blockade [160].
Table 2. Comprehensive overview of melanoma therapies: from traditional approaches to emerging treatments.
Table 2. Comprehensive overview of melanoma therapies: from traditional approaches to emerging treatments.
TherapyMechanism of ActionClinical OutcomesAdverse EffectsReferences
Targeted therapy (BRAF inhibitors)Disrupts MAPK/ERK signaling pathway by targeting mutated BRAF proteinImproved PFS and OS in BRAF-mutated melanomaSkin rash, headache, fever, joint pain[63,64,65,66]
Targeted therapy (MEK inhibitors)Targets MEK1 and MEK2 downstream of BRAF in MAPK signaling pathwayEnhanced efficacy in metastatic melanomasFatigue, rash, diarrhea, hypertension[68,69,70,71]
BRAF-MEK combination therapyReduces resistance development by blocking multiple points in the MAPK pathwayIncreased PFS and a higher overall response rate compared with monotherapyIncreased liver enzymes, fever, fatigue, dermatitis[72,73,74,75,76]
Checkpoint inhibitors: PD-1 and CTLA-4Enhances immune system’s ability to recognize and attack cancer cells by blocking PD-1 receptor
/by blocking immunosuppressive interaction between CTLA-4 and B7
Significant survival advantage in advanced melanoma, though associated with high toxicity levelsFatigue, skin rash, pruritus, colitis[78,79,80,81,82,83,84]
Talimogene laherparepvec (T-VEC)Genetically modified herpes simplex virus type 1 induces local and systemic immune responsesEffective as monotherapy and in combination with ICIs, in unresectable metastatic stage IIIB/C–IVM1a melanoma melanomasFlu-like symptoms, fatigue, chills, fever[87,88,89,90,91,92,93,94]
Combination of targeted therapy and immunotherapyCombines targeted therapy’s direct action on genetic mutations with immunotherapy’s broad-acting capacityProvides clinically significant survival benefits in patients with BRAF-mutant melanomaVaries based on combination[95,96,97]
Integration of surgical and systemic therapiesCombines surgical interventions with systemic therapies to improve outcomesImproves OS rates, reduces recurrence, and manages metastatic melanoma more effectivelyDependent on specific therapies used, varies[67,98,99,100,101,102]
Neoantigen vaccinesTailored to individual patients targeting unique tumor-specific antigens from mutationsSafe, induces specific immune responses against unique tumor antigensInjection site reactions, flu-like symptoms[103,104,105,106,107]
Adoptive cell transfer (TIL therapy)Uses patient’s own T cells, expanded and enhanced in lab, then reintroduced to patient to fight cancerImproves OS in advanced melanomaCytokine release syndrome, requires monitoring[108,109,110,111,112,113,114,161,162]
Microbiome and melanoma treatment interactionsGut microbiome’s role in treatment responses, leading to new strategiesInfluences outcomes and adverse reactions of immunotherapyVaries[115,116,117,118,119,120,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135]
Nanoparticle-based combination therapyUses nanoparticles for targeted drug delivery to cancer cells, reducing effects on healthy tissuesImproves chemotherapy outcomes and enhances PDT efficacyVaries, specific to nanoparticle type[136,137,138,139,140,141,142]
DNA damage response inhibitorsTargets DNA damage response (DDR) genesEnhances efficacy of existing therapies, including chemotherapy, targeted therapy, and immunotherapyAnemia, nausea, fatigue, neutropenia[143,144,145,146,147,148,149,150,151,152,153,154,155,156,157]
LAG-3 inhibitorsTargets LAG-3 to enhance immune responseSuperior PFS outcomes when used with PD-1 inhibitors in metastatic or unresectable melanomaFatigue, diarrhea, pruritus, rash[158,159,160]

5. Discussion and Conclusions

Melanoma treatment has seen dramatic advancements with the introduction of BRAF and MEK inhibitors, steering away from conventional chemotherapy, such as dacarbazine, towards a more personalized medicine approach. This pivotal shift has facilitated the emergence of immunotherapies, notably pembrolizumab and nivolumab, offering renewed hope for patients with advanced stages of the disease. The evolution towards targeted treatments and immunotherapies represents a significant leap in melanoma care, reflecting a deeper understanding of its molecular underpinnings.
Despite these advancements, the journey is far from over. Challenges such as resistance to targeted therapies and the variable efficacy of immunotherapies persist, underscoring the complexity of melanoma treatment. High costs and limited accessibility further complicate the delivery of these advanced treatments, particularly in resource-poor settings. The management of in-transit metastases also presents an area requiring more targeted research efforts, indicating gaps within current treatment paradigms.
The path forward is multifaceted, highlighting the importance of ongoing research into resistance mechanisms, the development of predictive biomarkers, and the creation of cost-effective care strategies on a global scale. Novel approaches to treatment strategies, such as personalized neoantigen vaccines, adoptive cell transfer, and the exploration of the microbiome’s role in melanoma treatment, underscore the dynamic nature of melanoma research and therapy. These innovative strategies offer the promise of further personalizing cancer care, enhancing the immune system’s response to melanoma, and overcoming the limitations of current treatments.
Reflecting on these innovative approaches reinforces the critical need for integrating emerging therapies into clinical practice. The exploration of treatments such as nanoparticle-based therapies and DNA damage response inhibitors represents a promising frontier in overcoming resistance to current therapies, potentially heralding a shift in how melanoma is treated on a molecular level. Furthermore, the collaboration of LAG-3 inhibitors with PD-1 inhibitors represents a significant advancement in immunotherapy, offering improved outcomes for patients.
In conclusion, while significant progress has been made in understanding and treating melanoma, translating these findings into widespread clinical practice is essential. The commitment to overcoming existing challenges, ensuring equitable access, and focusing on long-term outcomes remains paramount. Future research must continue to evolve, integrating novel approaches to treatment strategies to refine treatment approaches and, ultimately, achieve more personalized and effective care for all melanoma patients.

Author Contributions

Conceptualization, H.J.K. and Y.H.K.; writing—original draft preparation, H.J.K.; writing—review and editing, Y.H.K.; supervision, Y.H.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors have no conflicts of interest to declare.

References

  1. Long, G.V.; Swetter, S.M.; Menzies, A.M.; Gershenwald, J.E.; Scolyer, R.A. Cutaneous melanoma. Lancet 2023, 402, 485–502. [Google Scholar] [CrossRef]
  2. Lazaroff, J.; Bolotin, D. Targeted Therapy and Immunotherapy in Melanoma. Dermatol. Clin. 2023, 41, 65–77. [Google Scholar] [CrossRef]
  3. Di Raimondo, C.; Lozzi, F.; Di Domenico, P.P.; Campione, E.; Bianchi, L. The Diagnosis and Management of Cutaneous Metastases from Melanoma. Int. J. Mol. Sci. 2023, 24, 14535. [Google Scholar] [CrossRef]
  4. Seth, R.; Agarwala, S.S.; Messersmith, H.; Alluri, K.C.; Ascierto, P.A.; Atkins, M.B.; Bollin, K.; Chacon, M.; Davis, N.; Faries, M.B.; et al. Systemic Therapy for Melanoma: ASCO Guideline Update. J. Clin. Oncol. 2023, 41, 4794–4820. [Google Scholar] [CrossRef]
  5. Teixido, C.; Castillo, P.; Martinez-Vila, C.; Arance, A.; Alos, L. Molecular Markers and Targets in Melanoma. Cells 2021, 10, 2320. [Google Scholar] [CrossRef]
  6. Hossain, S.M.; Eccles, M.R. Phenotype Switching and the Melanoma Microenvironment; Impact on Immunotherapy and Drug Resistance. Int. J. Mol. Sci. 2023, 24, 1601. [Google Scholar] [CrossRef]
  7. Chen, H.; Hou, K.; Yu, J.; Wang, L.; Chen, X. Nanoparticle-Based Combination Therapy for Melanoma. Front. Oncol. 2022, 12, 928797. [Google Scholar] [CrossRef]
  8. Yang, T.T.; Yu, S.; Ke, C.K.; Cheng, S.T. The Genomic Landscape of Melanoma and Its Therapeutic Implications. Genes 2023, 14, 1021. [Google Scholar] [CrossRef]
  9. Garbe, C.; Amaral, T.; Peris, K.; Hauschild, A.; Arenberger, P.; Basset-Seguin, N.; Bastholt, L.; Bataille, V.; Del Marmol, V.; Dreno, B.; et al. European consensus-based interdisciplinary guideline for melanoma. Part 1: Diagnostics: Update 2022. Eur. J. Cancer 2022, 170, 236–255. [Google Scholar] [CrossRef]
  10. Silva-Rodríguez, P.; Fernández-Díaz, D.; Bande, M.; Pardo, M.; Loidi, L.; Blanco-Teijeiro, M.J. GNAQ and GNA11 Genes: A Comprehensive Review on Oncogenesis, Prognosis and Therapeutic Opportunities in Uveal Melanoma. Cancers 2022, 14, 3066. [Google Scholar] [CrossRef]
  11. Livingstone, E.; Zaremba, A.; Horn, S.; Ugurel, S.; Casalini, B.; Schlaak, M.; Hassel, J.C.; Herbst, R.; Utikal, J.S.; Weide, B.; et al. GNAQ and GNA11 mutant nonuveal melanoma: A subtype distinct from both cutaneous and uveal melanoma. Br. J. Dermatol. 2020, 183, 928–939. [Google Scholar] [CrossRef] [PubMed]
  12. Flaherty, K.T. A twenty year perspective on melanoma therapy. Pigment. Cell Melanoma Res. 2023, 36, 563–575. [Google Scholar] [CrossRef] [PubMed]
  13. Chamcheu, J.C.; Roy, T.; Uddin, M.B.; Banang-Mbeumi, S.; Chamcheu, R.N.; Walker, A.L.; Liu, Y.Y.; Huang, S. Role and Therapeutic Targeting of the PI3K/Akt/mTOR Signaling Pathway in Skin Cancer: A Review of Current Status and Future Trends on Natural and Synthetic Agents Therapy. Cells 2019, 8, 803. [Google Scholar] [CrossRef]
  14. Slominski, R.M.; Sarna, T.; Plonka, P.M.; Raman, C.; Brozyna, A.A.; Slominski, A.T. Melanoma, Melanin, and Melanogenesis: The Yin and Yang Relationship. Front. Oncol. 2022, 12, 842496. [Google Scholar] [CrossRef]
  15. Pawelek, J.M.; Lerner, A.B. 5, 6-Dihydroxyindole is a melanin precursor showing potent cytotoxicity. Nature 1978, 276, 627–628. [Google Scholar] [CrossRef]
  16. Slominski, A.; PAUs, R.; Mihm, M. Inhibition of melanogenesis as an adjuvant strategy in the treatment of melanotic melanomas: Selective review and hypothesis. Anticancer. Res. 1998, 18, 3709–3715. [Google Scholar]
  17. Miranda, M.; Ligas, C.; Amicarelli, F.; D’Alessandro, E.; Brisdelli, F.; Zarivi, O.; Poma, A. Sister chromatid exchange (SCE) rates in human melanoma cells as an index of mutagenesis. Mutagenesis 1997, 12, 233–236. [Google Scholar] [CrossRef]
  18. Graham, D.G.; Tiffany, S.M.; Vogel, F.S. The toxicity of melanin precursors. J. Investig. Dermatol. 1978, 70, 113–116. [Google Scholar] [CrossRef]
  19. Wick, M.M. Levodopa/dopamine analogs as inhibitors of DNA synthesis in human melanoma cells. J. Investig. Dermatol. 1989, 92, S329–S331. [Google Scholar] [CrossRef]
  20. Wick, M.; Fitzgerald, G. Inhibition of reverse transcriptase by tyrosinase generated quinones related to levodopa and dopamine. Chem.-Biol. Interact. 1981, 38, 99–107. [Google Scholar] [CrossRef]
  21. Kim, E.; Panzella, L.; Napolitano, A.; Payne, G.F. Redox activities of melanins investigated by electrochemical reverse engineering: Implications for their roles in oxidative stress. J. Investig. Dermatol. 2020, 140, 537–543. [Google Scholar] [CrossRef] [PubMed]
  22. Ito, S.; Wakamatsu, K.; Sarna, T. Photodegradation of eumelanin and pheomelanin and its pathophysiological implications. Photochem. Photobiol. 2018, 94, 409–420. [Google Scholar] [CrossRef] [PubMed]
  23. Brash, D.E. UV-induced melanin chemiexcitation: A new mode of melanoma pathogenesis. Toxicol. Pathol. 2016, 44, 552–554. [Google Scholar] [CrossRef] [PubMed]
  24. Premi, S.; Wallisch, S.; Mano, C.M.; Weiner, A.B.; Bacchiocchi, A.; Wakamatsu, K.; Bechara, E.J.; Halaban, R.; Douki, T.; Brash, D.E. Chemiexcitation of melanin derivatives induces DNA photoproducts long after UV exposure. Science 2015, 347, 842–847. [Google Scholar] [CrossRef]
  25. Lembo, S.; Di Caprio, R.; Micillo, R.; Balato, A.; Monfrecola, G.; Panzella, L.; Napolitano, A. Light-independent pro-inflammatory and pro-oxidant effects of purified human hair melanins on keratinocyte cell cultures. Exp. Dermatol. 2017, 26, 592–594. [Google Scholar] [CrossRef]
  26. Brożyna, A.A.; Jóźwicki, W.; Carlson, J.A.; Slominski, A.T. Melanogenesis affects overall and disease-free survival in patients with stage III and IV melanoma. Hum. Pathol. 2013, 44, 2071–2074. [Google Scholar] [CrossRef]
  27. Brozyna, A.; Jozwicki, W.; Roszkowski, K.; Filipiak, J.; Slominski, A. Melanin content in melanoma metastases affects the outcome of radiotherapy. Oncotarget 2016, 7, 17844–17853. [Google Scholar] [CrossRef]
  28. Shields, C.L.; Kaliki, S.; Furuta, M.; Fulco, E.; Alarcon, C.; Shields, J.A. American joint committee on cancer classification of uveal melanoma (anatomic stage) predicts prognosis in 7731 patients: The 2013 zimmerman lecture. Ophthalmology 2015, 122, 1180–1186. [Google Scholar] [CrossRef]
  29. Shields, C.; Kaliki, S.; Cohen, M.; Shields, P.; Furuta, M.; Shields, J. Prognosis of uveal melanoma based on race in 8100 patients: The 2015 Doyne Lecture. Eye 2015, 29, 1027–1035. [Google Scholar] [CrossRef]
  30. Slominski, R.M.; Raman, C.; Chen, J.Y.; Slominski, A.T. How cancer hijacks the body’s homeostasis through the neuroendocrine system. Trends Neurosci. 2023, 46, 263–275. [Google Scholar] [CrossRef] [PubMed]
  31. Böhm, M.; Luger, T.A.; Tobin, D.J.; García-Borrón, J.C. Melanocortin receptor ligands: New horizons for skin biology and clinical dermatology. J. Investig. Dermatol. 2006, 126, 1966–1975. [Google Scholar] [CrossRef]
  32. Slominski, A.; Wortsman, J.; Luger, T.; Paus, R.; Solomon, S. Corticotropin releasing hormone and proopiomelanocortin involvement in the cutaneous response to stress. Physiol. Rev. 2000, 80, 979–1020. [Google Scholar] [CrossRef] [PubMed]
  33. Hedley, S.; Murray, A.; Sisley, K.; Ghanem, G.; Morandini, R.; Gawkrodger, D.; Mac Neil, S. α-Melanocyte stimulating hormone can reduce T-cell interaction with melanoma cells in vitro. Melanoma Res. 2000, 10, 323–330. [Google Scholar] [CrossRef] [PubMed]
  34. Slominski, A.; Wortsman, J.; Mazurkiewicz, J.E.; Matsuoka, L.; Dietrich, J.; Lawrence, K.; Gorbani, A.; Paus, R. Detection of proopiomelanocortin-derived antigens in normal and pathologic human skin. J. Lab. Clin. Med. 1993, 122, 658–666. [Google Scholar]
  35. Nagahama, M.; Funasaka, Y.; Fernandez-Frez, M.; Ohashi, A.; Chakraborty, A.; Ueda, M.; Ichihashi, M. Immunoreactivity of α-melanocyte-stimulating hormone, adrenocorticotrophic hormone and β-endorphin in cutaneous malignant melanoma and benign melanocytic naevi. Br. J. Dermatol. 1998, 138, 981–985. [Google Scholar] [CrossRef]
  36. Ghanem, G.; Lienard, D.; Hanson, P.; Lejeune, F.; Fruhling, J. Increased serum alpha-melanocyte stimulating hormone (alpha-MSH) in human malignant melanoma. Eur. J. Cancer Clin. Oncol. 1986, 22, 535–536. [Google Scholar] [CrossRef]
  37. Loir, B.; Bouchard, B.; Morandini, R.; Marmol, V.D.; Deraemaecker, R.; Garcia-Borron, J.C.; Ghanem, G. Immunoreactive α-melanotropin as an autocrine effector in human melanoma cells. Eur. J. Biochem. 1997, 244, 923–930. [Google Scholar] [CrossRef]
  38. Funasaka, Y.; Sato, H.; Chakraborty, A.K.; Ohashi, A.; Chrousos, G.P.; Ichihashi, M. Expression of proopiomelanocortin, corticotropin-releasing hormone (CRH), and CRH receptor in melanoma cells, nevus cells, and normal human melanocytes. J. Investig. Dermatol. Symp. Proc. 1999, 4, 105–109. [Google Scholar] [CrossRef]
  39. Sato, H.; Nagashima, Y.; Chrousos, G.P.; Ichihashi, M.; Funasaka, Y. The expression of corticotropin-releasing hormone in melanoma. Pigment Cell Res. 2002, 15, 98–103. [Google Scholar] [CrossRef]
  40. Quek, C.; Bai, X.; Long, G.V.; Scolyer, R.A.; Wilmott, J.S. High-Dimensional Single-Cell Transcriptomics in Melanoma and Cancer Immunotherapy. Genes 2021, 12, 1629. [Google Scholar] [CrossRef]
  41. 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]
  42. Ny, L.; Hernberg, M.; Nyakas, M.; Koivunen, J.; Oddershede, L.; Yoon, M.; Wang, X.; Guyot, P.; Geisler, J. BRAF mutational status as a prognostic marker for survival in malignant melanoma: A systematic review and meta-analysis. Acta Oncol. 2020, 59, 833–844. [Google Scholar] [CrossRef]
  43. Wolfe, A.R.; Chablani, P.; Siedow, M.R.; Miller, E.D.; Walston, S.; Kendra, K.L.; Wuthrick, E.; Williams, T.M. BRAF mutation correlates with worse local-regional control following radiation therapy in patients with stage III melanoma. Radiat. Oncol. 2021, 16, 181. [Google Scholar] [CrossRef]
  44. Ellerhorst, J.A.; Greene, V.R.; Ekmekcioglu, S.; Warneke, C.L.; Johnson, M.M.; Cooke, C.P.; Wang, L.E.; Prieto, V.G.; Gershenwald, J.E.; Wei, Q.; et al. Clinical correlates of NRAS and BRAF mutations in primary human melanoma. Clin. Cancer Res. 2011, 17, 229–235. [Google Scholar] [CrossRef]
  45. Jakob, J.A.; Bassett, R.L., Jr.; 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]
  46. Dummer, R.; Schadendorf, D.; Ascierto, P.A.; Arance, A.; Dutriaux, C.; Di Giacomo, A.M.; Rutkowski, P.; Del Vecchio, M.; Gutzmer, R.; Mandala, M. Binimetinib versus dacarbazine in patients with advanced NRAS-mutant melanoma (NEMO): A multicentre, open-label, randomised, phase 3 trial. Lancet Oncol. 2017, 18, 435–445. [Google Scholar] [CrossRef] [PubMed]
  47. Carvajal, R.D.; Antonescu, C.R.; Wolchok, J.D.; Chapman, P.B.; Roman, R.-A.; Teitcher, J.; Panageas, K.S.; Busam, K.J.; Chmielowski, B.; Lutzky, J.; et al. KIT as a Therapeutic Target in Metastatic Melanoma. JAMA 2011, 305, 2327–2334. [Google Scholar] [CrossRef]
  48. 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. Imatinib for melanomas harboring mutationally activated or amplified KIT arising on mucosal, acral, and chronically sun-damaged skin. J. Clin. Oncol. 2013, 31, 3182. [Google Scholar] [CrossRef] [PubMed]
  49. Wei, X.; Mao, L.; Chi, Z.; Sheng, X.; Cui, C.; Kong, Y.; Dai, J.; Wang, X.; Li, S.; Tang, B. Efficacy evaluation of imatinib for the treatment of melanoma: Evidence from a retrospective study. Oncol. Res. 2019, 27, 495. [Google Scholar] [CrossRef] [PubMed]
  50. Guo, J.; Carvajal, R.; Dummer, R.; Hauschild, A.; Daud, A.; Bastian, B.; Markovic, S.; Queirolo, P.; Arance, A.; Berking, C. 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]
  51. Guo, J.; Si, L.; Kong, Y.; Flaherty, K.T.; Xu, X.; Zhu, Y.; Corless, C.L.; Li, L.; Li, H.; Sheng, X.; et al. Phase II, open-label, single-arm trial of imatinib mesylate in patients with metastatic melanoma harboring c-Kit mutation or amplification. J. Clin. Oncol. 2011, 29, 2904–2909. [Google Scholar] [CrossRef]
  52. Thielmann, C.M.; Chorti, E.; Matull, J.; Murali, R.; Zaremba, A.; Lodde, G.; Jansen, P.; Richter, L.; Kretz, J.; Möller, I.; et al. NF1-mutated melanomas reveal distinct clinical characteristics depending on tumour origin and respond favourably to immune checkpoint inhibitors. Eur. J. Cancer 2021, 159, 113–124. [Google Scholar] [CrossRef] [PubMed]
  53. Akbani, R.; Akdemir, K.C.; Aksoy, B.A.; Albert, M.; Ally, A.; Amin, S.B.; Arachchi, H.; Arora, A.; Auman, J.T.; Ayala, B. Genomic classification of cutaneous melanoma. Cell 2015, 161, 1681–1696. [Google Scholar] [CrossRef] [PubMed]
  54. Hegedüs, L.; Livingstone, E.; Bánkfalvi, Á.; Viehof, J.; Enyedi, Á.; Bilecz, Á.; Győrffy, B.; Baranyi, M.; Tőkés, A.M.; Gil, J.; et al. The Prognostic Relevance of PMCA4 Expression in Melanoma: Gender Specificity and Implications for Immune Checkpoint Inhibition. Int. J. Mol. Sci. 2022, 23, 3324. [Google Scholar] [CrossRef]
  55. Forschner, A.; Battke, F.; Hadaschik, D.; Schulze, M.; Weißgraeber, S.; Han, C.T.; Kopp, M.; Frick, M.; Klumpp, B.; Tietze, N.; et al. Tumor mutation burden and circulating tumor DNA in combined CTLA-4 and PD-1 antibody therapy in metastatic melanoma—Results of a prospective biomarker study. J. Immunother. Cancer 2019, 7, 180. [Google Scholar] [CrossRef] [PubMed]
  56. Huang, T.; Chen, X.; Zhang, H.; Liang, Y.; Li, L.; Wei, H.; Sun, W.; Wang, Y. Prognostic Role of Tumor Mutational Burden in Cancer Patients Treated With Immune Checkpoint Inhibitors: A Systematic Review and Meta-Analysis. Front. Oncol. 2021, 11, 706652. [Google Scholar] [CrossRef] [PubMed]
  57. Marcus, L.; Fashoyin-Aje, L.A.; Donoghue, M.; Yuan, M.; Rodriguez, L.; Gallagher, P.S.; Philip, R.; Ghosh, S.; Theoret, M.R.; Beaver, J.A.; et al. FDA Approval Summary: Pembrolizumab for the Treatment of Tumor Mutational Burden-High Solid Tumors. Clin. Cancer Res. 2021, 27, 4685–4689. [Google Scholar] [CrossRef] [PubMed]
  58. Bollard, S.M.; Casalou, C.; Potter, S.M. Gene expression profiling in melanoma: A view from the clinic. Cancer Treat. Res. Commun. 2021, 29, 100447. [Google Scholar] [CrossRef] [PubMed]
  59. Grossman, D.; Okwundu, N.; Bartlett, E.K.; Marchetti, M.A.; Othus, M.; Coit, D.G.; Hartman, R.I.; Leachman, S.A.; Berry, E.G.; Korde, L.; et al. Prognostic Gene Expression Profiling in Cutaneous Melanoma: Identifying the Knowledge Gaps and Assessing the Clinical Benefit. JAMA Dermatol. 2020, 156, 1004–1011. [Google Scholar] [CrossRef]
  60. Gopalakrishnan, V.; Spencer, C.N.; Nezi, L.; Reuben, A.; Andrews, M.C.; Karpinets, T.V.; Prieto, P.A.; Vicente, D.; Hoffman, K.; Wei, S.C.; et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science 2018, 359, 97–103. [Google Scholar] [CrossRef]
  61. Dubin, K.; Callahan, M.K.; Ren, B.; Khanin, R.; Viale, A.; Ling, L.; No, D.; Gobourne, A.; Littmann, E.; Huttenhower, C.; et al. Intestinal microbiome analyses identify melanoma patients at risk for checkpoint-blockade-induced colitis. Nat. Commun. 2016, 7, 10391. [Google Scholar] [CrossRef]
  62. Garbe, C.; Amaral, T.; Peris, K.; Hauschild, A.; Arenberger, P.; Basset-Seguin, N.; Bastholt, L.; Bataille, V.; Del Marmol, V.; Dreno, B.; et al. European consensus-based interdisciplinary guideline for melanoma. Part 2: Treatment—Update 2022. Eur. J. Cancer 2022, 170, 256–284. [Google Scholar] [CrossRef]
  63. Chapman, P.B.; Hauschild, A.; Robert, C.; Haanen, J.B.; Ascierto, P.; Larkin, J.; Dummer, R.; Garbe, C.; Testori, A.; Maio, M.; et al. Improved Survival with Vemurafenib in Melanoma with BRAF V600E Mutation. N. Engl. J. Med. 2011, 364, 2507–2516. [Google Scholar] [CrossRef] [PubMed]
  64. Chapman, P.B.; Robert, C.; Larkin, J.; Haanen, J.B.; Ribas, A.; Hogg, D.; Hamid, O.; Ascierto, P.A.; Testori, A.; Lorigan, P.C.; et al. Vemurafenib in patients with BRAFV600 mutation-positive metastatic melanoma: Final overall survival results of the randomized BRIM-3 study. Ann. Oncol. 2017, 28, 2581–2587. [Google Scholar] [CrossRef]
  65. Luke, J.J. Comprehensive Clinical Trial Data Summation for BRAF-MEK Inhibition and Checkpoint Immunotherapy in Metastatic Melanoma. Oncologist 2019, 24, e1197–e1211. [Google Scholar] [CrossRef]
  66. 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] [PubMed]
  67. Villani, A.; Scalvenzi, M.; Micali, G.; Lacarrubba, F.; Fornaro, L.; Martora, F.; Potestio, L. Management of Advanced Invasive Melanoma: New Strategies. Adv. Ther. 2023, 40, 3381–3394. [Google Scholar] [CrossRef] [PubMed]
  68. Latimer, N.R.; Bell, H.; Abrams, K.R.; Amonkar, M.M.; Casey, M. Adjusting for treatment switching in the METRIC study shows further improved overall survival with trametinib compared with chemotherapy. Cancer Med. 2016, 5, 806–815. [Google Scholar] [CrossRef]
  69. Flaherty, K.T.; Robert, C.; Hersey, P.; Nathan, P.; Garbe, C.; Milhem, M.; Demidov, L.V.; Hassel, J.C.; Rutkowski, P.; Mohr, P.; et al. Improved survival with MEK inhibition in BRAF-mutated melanoma. N. Engl. J. Med. 2012, 367, 107–114. [Google Scholar] [CrossRef]
  70. Dummer, R.; Brase, J.C.; Garrett, J.; Campbell, C.D.; Gasal, E.; Squires, M.; Gusenleitner, D.; Santinami, M.; Atkinson, V.; Mandalà, M.; et al. Adjuvant dabrafenib plus trametinib versus placebo in patients with resected, BRAFV600-mutant, stage III melanoma (COMBI-AD): Exploratory biomarker analyses from a randomised, phase 3 trial. Lancet Oncol. 2020, 21, 358–372. [Google Scholar] [CrossRef]
  71. Robert, C.; Grob, J.J.; Stroyakovskiy, D.; Karaszewska, B.; Hauschild, A.; Levchenko, E.; Chiarion Sileni, V.; Schachter, J.; Garbe, C.; Bondarenko, I. Five-year outcomes with dabrafenib plus trametinib in metastatic melanoma. N. Engl. J. Med. 2019, 381, 626–636. [Google Scholar] [CrossRef]
  72. Larkin, J.; Ascierto, P.A.; Dréno, B.; Atkinson, V.; Liszkay, G.; Maio, M.; Mandalà, M.; Demidov, L.; Stroyakovskiy, D.; Thomas, L.; et al. Combined Vemurafenib and Cobimetinib in BRAF-Mutated Melanoma. N. Engl. J. Med. 2014, 371, 1867–1876. [Google Scholar] [CrossRef]
  73. Ascierto, P.A.; McArthur, G.A.; Dréno, B.; Atkinson, V.; Liszkay, G.; Di Giacomo, A.M.; Mandalà, M.; Demidov, L.; Stroyakovskiy, D.; Thomas, L.; et al. Cobimetinib combined with vemurafenib in advanced BRAF(V600)-mutant melanoma (coBRIM): Updated efficacy results from a randomised, double-blind, phase 3 trial. Lancet Oncol. 2016, 17, 1248–1260. [Google Scholar] [CrossRef]
  74. 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. Dabrafenib and trametinib versus dabrafenib and placebo for Val600 BRAF-mutant melanoma: A multicentre, double-blind, phase 3 randomised controlled trial. Lancet 2015, 386, 444–451. [Google Scholar] [CrossRef]
  75. 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]
  76. Ascierto, P.A.; Dréno, B.; Larkin, J.; Ribas, A.; Liszkay, G.; Maio, M.; Mandalà, M.; Demidov, L.; Stroyakovskiy, D.; Thomas, L. 5-year outcomes with cobimetinib plus vemurafenib in BRAF V600 mutation–positive advanced melanoma: Extended follow-up of the coBRIM study. Clin. Cancer Res. 2021, 27, 5225–5235. [Google Scholar] [CrossRef]
  77. Gouda, M.A.; Subbiah, V. Precision oncology for BRAF-mutant cancers with BRAF and MEK inhibitors: From melanoma to tissue-agnostic therapy. ESMO Open 2023, 8, 100788. [Google Scholar] [CrossRef] [PubMed]
  78. Robert, C.; Thomas, L.; Bondarenko, I.; O’Day, S.; Weber, J.; Garbe, C.; Lebbe, C.; Baurain, J.F.; Testori, A.; Grob, J.J.; et al. Ipilimumab plus dacarbazine for previously untreated metastatic melanoma. N. Engl. J. Med. 2011, 364, 2517–2526. [Google Scholar] [CrossRef] [PubMed]
  79. Schachter, J.; Ribas, A.; Long, G.V.; Arance, A.; Grob, J.-J.; Mortier, L.; Daud, A.; Carlino, M.S.; McNeil, C.; Lotem, M. Pembrolizumab versus ipilimumab for advanced melanoma: Final overall survival results of a multicentre, randomised, open-label phase 3 study (KEYNOTE-006). Lancet 2017, 390, 1853–1862. [Google Scholar] [CrossRef] [PubMed]
  80. Ribas, A.; Puzanov, I.; Dummer, R.; Schadendorf, D.; Hamid, O.; Robert, C.; Hodi, F.S.; Schachter, J.; Pavlick, A.C.; Lewis, K.D. Pembrolizumab versus investigator-choice chemotherapy for ipilimumab-refractory melanoma (KEYNOTE-002): A randomised, controlled, phase 2 trial. Lancet Oncol. 2015, 16, 908–918. [Google Scholar] [CrossRef] [PubMed]
  81. Koppolu, V.; Vasigala, V.K.R. Checkpoint immunotherapy by nivolumab for treatment of metastatic melanoma. J. Cancer Res. Ther. 2018, 14, 1167–1175. [Google Scholar] [CrossRef] [PubMed]
  82. Larkin, J.; Minor, D.; D’Angelo, S.; Neyns, B.; Smylie, M.; Miller Jr, W.H.; Gutzmer, R.; Linette, G.; Chmielowski, B.; Lao, C.D. Overall survival in patients with advanced melanoma who received nivolumab versus investigator’s choice chemotherapy in CheckMate 037: A randomized, controlled, open-label phase III trial. J. Clin. Oncol. 2018, 36, 383. [Google Scholar] [CrossRef]
  83. Robert, C.; Long, G.V.; Brady, B.; Dutriaux, C.; Maio, M.; Mortier, L.; Hassel, J.C.; Rutkowski, P.; McNeil, C.; Kalinka-Warzocha, E. Nivolumab in previously untreated melanoma without BRAF mutation. N. Engl. J. Med. 2015, 372, 320–330. [Google Scholar] [CrossRef]
  84. 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. 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] [PubMed]
  85. Larkin, J.; Chiarion-Sileni, V.; Gonzalez, R.; Grob, J.-J.; Rutkowski, P.; Lao, C.D.; Cowey, C.L.; Schadendorf, D.; Wagstaff, J.; Dummer, R. Five-year survival with combined nivolumab and ipilimumab in advanced melanoma. N. Engl. J. Med. 2019, 381, 1535–1546. [Google Scholar] [CrossRef]
  86. Wolchok, J.D.; Chiarion-Sileni, V.; Gonzalez, R.; Grob, J.-J.; Rutkowski, P.; Lao, C.D.; Cowey, C.L.; Schadendorf, D.; Wagstaff, J.; Dummer, R. CheckMate 067: 6.5-Year Outcomes in Patients (pts) with Advanced Melanoma; Wolters Kluwer Health: Philadelphia, PE, USA, 2021. [Google Scholar]
  87. Johnson, D.B.; Puzanov, I.; Kelley, M.C. Talimogene laherparepvec (T-VEC) for the treatment of advanced melanoma. Immunotherapy 2015, 7, 611–619. [Google Scholar] [CrossRef]
  88. Guo, Z.S.; Thorne, S.H.; Bartlett, D.L. Oncolytic virotherapy: Molecular targets in tumor-selective replication and carrier cell-mediated delivery of oncolytic viruses. Biochim. Biophys. Acta 2008, 1785, 217–231. [Google Scholar] [CrossRef] [PubMed]
  89. Senzer, N.N.; Kaufman, H.L.; Amatruda, T.; Nemunaitis, M.; Reid, T.; Daniels, G.; Gonzalez, R.; Glaspy, J.; Whitman, E.; Harrington, K.; et al. Phase II clinical trial of a granulocyte-macrophage colony-stimulating factor-encoding, second-generation oncolytic herpesvirus in patients with unresectable metastatic melanoma. J. Clin. Oncol. 2009, 27, 5763–5771. [Google Scholar] [CrossRef]
  90. Kaufman, H.L.; Kim, D.W.; DeRaffele, G.; Mitcham, J.; Coffin, R.S.; Kim-Schulze, S. Local and distant immunity induced by intralesional vaccination with an oncolytic herpes virus encoding GM-CSF in patients with stage IIIc and IV melanoma. Ann. Surg. Oncol. 2010, 17, 718–730. [Google Scholar] [CrossRef]
  91. Andtbacka, R.H.I.; Collichio, F.; Harrington, K.J.; Middleton, M.R.; Downey, G.; Öhrling, K.; Kaufman, H.L. Final analyses of OPTiM: A randomized phase III trial of talimogene laherparepvec versus granulocyte-macrophage colony-stimulating factor in unresectable stage III-IV melanoma. J. Immunother. Cancer 2019, 7, 145. [Google Scholar] [CrossRef]
  92. Chesney, J.; Puzanov, I.; Collichio, F.; Singh, P.; Milhem, M.M.; Glaspy, J.; Hamid, O.; Ross, M.; Friedlander, P.; Garbe, C.; et al. Randomized, Open-Label Phase II Study Evaluating the Efficacy and Safety of Talimogene Laherparepvec in Combination With Ipilimumab Versus Ipilimumab Alone in Patients With Advanced, Unresectable Melanoma. J. Clin. Oncol. 2018, 36, 1658–1667. [Google Scholar] [CrossRef]
  93. Ribas, A.; Dummer, R.; Puzanov, I.; VanderWalde, A.; Andtbacka, R.H.I.; Michielin, O.; Olszanski, A.J.; Malvehy, J.; Cebon, J.; Fernandez, E.; et al. Oncolytic Virotherapy Promotes Intratumoral T Cell Infiltration and Improves Anti-PD-1 Immunotherapy. Cell 2017, 170, 1109–1119.e1110. [Google Scholar] [CrossRef]
  94. Long, G.; Dummer, R.; Andtbacka, R.; Johnson, D.; Michielin, O.; Martin-Algarra, S. Follow-up analysis of MASTERKEY-265 phase 1b (ph1b) study of talimogene laherparepvec (T-VEC) in combination (combo) with pembrolizumab (pembro) in patients (pts) with unresectable stage IIIB–IVM1c melanoma (MEL). In Proceedings of the Society for Melanoma Research Fifteenth International Congress, Manchester, UK, 24–27 October 2018; pp. 24–27. [Google Scholar]
  95. Ascierto, P.A.; Ferrucci, P.F.; Fisher, R.; Del Vecchio, M.; Atkinson, V.; Schmidt, H.; Schachter, J.; Queirolo, P.; Long, G.V.; Di Giacomo, A.M.; et al. Dabrafenib, trametinib and pembrolizumab or placebo in BRAF-mutant melanoma. Nat. Med. 2019, 25, 941–946. [Google Scholar] [CrossRef]
  96. Ferrucci, P.F.; Di Giacomo, A.M.; Del Vecchio, M.; Atkinson, V.; Schmidt, H.; Schachter, J.; Queirolo, P.; Long, G.V.; Stephens, R.; Svane, I.M.; et al. KEYNOTE-022 part 3: A randomized, double-blind, phase 2 study of pembrolizumab, dabrafenib, and trametinib in BRAF-mutant melanoma. J. Immunother. Cancer 2020, 8, e001806. [Google Scholar] [CrossRef] [PubMed]
  97. Ascierto, P.A.; Mandalà, M.; Ferrucci, P.F.; Guidoboni, M.; Rutkowski, P.; Ferraresi, V.; Arance, A.; Guida, M.; Maiello, E.; Gogas, H.; et al. Sequencing of Ipilimumab Plus Nivolumab and Encorafenib Plus Binimetinib for Untreated BRAF-Mutated Metastatic Melanoma (SECOMBIT): A Randomized, Three-Arm, Open-Label Phase II Trial. J. Clin. Oncol. 2023, 41, 212–221. [Google Scholar] [CrossRef] [PubMed]
  98. Favre-Bulle, A.; Bencina, G.; Zhang, S.; Jiang, R.; Andritschke, D.; Bhadhuri, A. Cost-effectiveness of pembrolizumab as an adjuvant treatment for patients with resected stage IIB or IIC melanoma in Switzerland. J. Med. Econ. 2023, 26, 283–292. [Google Scholar] [CrossRef] [PubMed]
  99. Luke, J.J.; Ascierto, P.A.; Khattak, M.A.; Merino, L.d.l.C.; Vecchio, M.D.; Rutkowski, P.; Spagnolo, F.; Mackiewicz, J.; Chiarion-Sileni, V.; Kirkwood, J.M.M.; et al. Pembrolizumab versus placebo as adjuvant therapy in stage IIB or IIC melanoma: Final analysis of distant metastasis-free survival in the phase 3 KEYNOTE-716 study. J. Clin. Oncol. 2023, 41, LBA9505. [Google Scholar] [CrossRef]
  100. Poh, A. mRNA Vaccine Slows Melanoma Recurrence. Cancer Discov. 2023, 13, 1278. [Google Scholar] [CrossRef]
  101. Nikolaou, V.; Stratigos, A.J. Adjuvant treatment in advanced melanoma: How far have we come? J. Eur. Acad. Dermatol. Venereol. 2023, 37, 851–852. [Google Scholar] [CrossRef] [PubMed]
  102. Larkin, J.; Del Vecchio, M.; Mandalá, M.; Gogas, H.; Arance Fernandez, A.M.; Dalle, S.; Cowey, C.L.; Schenker, M.; Grob, J.J.; Chiarion-Sileni, V.; et al. Adjuvant Nivolumab versus Ipilimumab in Resected Stage III/IV Melanoma: 5-Year Efficacy and Biomarker Results from CheckMate 238. Clin. Cancer Res. 2023, 29, 3352–3361. [Google Scholar] [CrossRef]
  103. Carreno, B.M.; Magrini, V.; Becker-Hapak, M.; Kaabinejadian, S.; Hundal, J.; Petti, A.A.; Ly, A.; Lie, W.R.; Hildebrand, W.H.; Mardis, E.R.; et al. Cancer immunotherapy. A dendritic cell vaccine increases the breadth and diversity of melanoma neoantigen-specific T cells. Science 2015, 348, 803–808. [Google Scholar] [CrossRef] [PubMed]
  104. Hu, Z.; Leet, D.E.; Allesøe, R.L.; Oliveira, G.; Li, S.; Luoma, A.M.; Liu, J.; Forman, J.; Huang, T.; Iorgulescu, J.B.; et al. Personal neoantigen vaccines induce persistent memory T cell responses and epitope spreading in patients with melanoma. Nat. Med. 2021, 27, 515–525. [Google Scholar] [CrossRef] [PubMed]
  105. Ott, P.A.; Hu, Z.; Keskin, D.B.; Shukla, S.A.; Sun, J.; Bozym, D.J.; Zhang, W.; Luoma, A.; Giobbie-Hurder, A.; Peter, L.; et al. An immunogenic personal neoantigen vaccine for patients with melanoma. Nature 2017, 547, 217–221. [Google Scholar] [CrossRef] [PubMed]
  106. Sahin, U.; Derhovanessian, E.; Miller, M.; Kloke, B.P.; Simon, P.; Löwer, M.; Bukur, V.; Tadmor, A.D.; Luxemburger, U.; Schrörs, B.; et al. Personalized RNA mutanome vaccines mobilize poly-specific therapeutic immunity against cancer. Nature 2017, 547, 222–226. [Google Scholar] [CrossRef]
  107. Lopez, J.S.; Camidge, R.; Iafolla, M.; Rottey, S.; Schuler, M.; Hellmann, M.; Balmanoukian, A.; Dirix Gordon, L.; Sullivan, R.; Henick, B. A phase Ib study to evaluate RO7198457, an individualized Neoantigen Specific immunoTherapy (iNeST), in combination with atezolizumab in patients with locally advanced or metastatic solid tumors. Cancer Res. 2020, 80, CT301. [Google Scholar] [CrossRef]
  108. Sarnaik, A.A.; Hamid, O.; Khushalani, N.I.; Lewis, K.D.; Medina, T.; Kluger, H.M.; Thomas, S.S.; Domingo-Musibay, E.; Pavlick, A.C.; Whitman, E.D.; et al. Lifileucel, a Tumor-Infiltrating Lymphocyte Therapy, in Metastatic Melanoma. J. Clin. Oncol. 2021, 39, 2656–2666. [Google Scholar] [CrossRef] [PubMed]
  109. Dafni, U.; Michielin, O.; Lluesma, S.M.; Tsourti, Z.; Polydoropoulou, V.; Karlis, D.; Besser, M.J.; Haanen, J.; Svane, I.M.; Ohashi, P.S.; et al. Efficacy of adoptive therapy with tumor-infiltrating lymphocytes and recombinant interleukin-2 in advanced cutaneous melanoma: A systematic review and meta-analysis. Ann. Oncol. 2019, 30, 1902–1913. [Google Scholar] [CrossRef]
  110. Rosenberg, S.A.; Restifo, N.P. Adoptive cell transfer as personalized immunotherapy for human cancer. Science 2015, 348, 62–68. [Google Scholar] [CrossRef]
  111. Chesney, J.A. Tumor-infiltrating lymphocyte therapy in metastatic melanoma. Clin. Adv. Hematol. Oncol. 2023, 21, 49–51. [Google Scholar]
  112. Betof Warner, A.; Corrie, P.G.; Hamid, O. Tumor-Infiltrating Lymphocyte Therapy in Melanoma: Facts to the Future. Clin. Cancer Res. 2023, 29, 1835–1854. [Google Scholar] [CrossRef]
  113. Creasy, C.A.; Meng, Y.J.; Forget, M.A.; Karpinets, T.; Tomczak, K.; Stewart, C.; Torres-Cabala, C.A.; Pilon-Thomas, S.; Sarnaik, A.A.; Mulé, J.J.; et al. Genomic Correlates of Outcome in Tumor-Infiltrating Lymphocyte Therapy for Metastatic Melanoma. Clin. Cancer Res. 2022, 28, 1911–1924. [Google Scholar] [CrossRef]
  114. Kristensen, N.P.; Heeke, C.; Tvingsholm, S.A.; Borch, A.; Draghi, A.; Crowther, M.D.; Carri, I.; Munk, K.K.; Holm, J.S.; Bjerregaard, A.M.; et al. Neoantigen-reactive CD8+ T cells affect clinical outcome of adoptive cell therapy with tumor-infiltrating lymphocytes in melanoma. J. Clin. Investig. 2022, 132, e150535. [Google Scholar] [CrossRef] [PubMed]
  115. Makaranka, S.; Scutt, F.; Frixou, M.; Wensley, K.E.; Sharma, R.; Greenhowe, J. The gut microbiome and melanoma: A review. Exp. Dermatol. 2022, 31, 1292–1301. [Google Scholar] [CrossRef] [PubMed]
  116. Bhatt, A.P.; Redinbo, M.R.; Bultman, S.J. The role of the microbiome in cancer development and therapy. CA Cancer J. Clin. 2017, 67, 326–344. [Google Scholar] [CrossRef] [PubMed]
  117. Shreiner, A.B.; Kao, J.Y.; Young, V.B. The gut microbiome in health and in disease. Curr. Opin. Gastroenterol. 2015, 31, 69–75. [Google Scholar] [CrossRef] [PubMed]
  118. Mrázek, J.; Mekadim, C.; Kučerová, P.; Švejstil, R.; Salmonová, H.; Vlasáková, J.; Tarasová, R.; Čížková, J.; Červinková, M. Melanoma-related changes in skin microbiome. Folia Microbiol. 2019, 64, 435–442. [Google Scholar] [CrossRef] [PubMed]
  119. Mizuhashi, S.; Kajihara, I.; Sawamura, S.; Kanemaru, H.; Makino, K.; Aoi, J.; Makino, T.; Masuguchi, S.; Fukushima, S.; Ihn, H. Skin microbiome in acral melanoma: Corynebacterium is associated with advanced melanoma. J. Dermatol. 2021, 48, e15–e16. [Google Scholar] [CrossRef] [PubMed]
  120. Patra, V.; Gallais Sérézal, I.; Wolf, P. Potential of Skin Microbiome, Pro- and/or Pre-Biotics to Affect Local Cutaneous Responses to UV Exposure. Nutrients 2020, 12, 1795. [Google Scholar] [CrossRef]
  121. Sherwani, M.A.; Tufail, S.; Muzaffar, A.F.; Yusuf, N. The skin microbiome and immune system: Potential target for chemoprevention? Photodermatol. Photoimmunol. Photomed. 2018, 34, 25–34. [Google Scholar] [CrossRef]
  122. 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]
  123. Trivieri, N.; Pracella, R.; Cariglia, M.G.; Panebianco, C.; Parrella, P.; Visioli, A.; Giani, F.; Soriano, A.A.; Barile, C.; Canistro, G.; et al. BRAF(V600E) mutation impinges on gut microbial markers defining novel biomarkers for serrated colorectal cancer effective therapies. J. Exp. Clin. Cancer Res. 2020, 39, 285. [Google Scholar] [CrossRef]
  124. Routy, B.; Le Chatelier, E.; Derosa, L.; Duong, C.P.M.; Alou, M.T.; Daillère, R.; Fluckiger, A.; Messaoudene, M.; Rauber, C.; Roberti, M.P.; et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science 2018, 359, 91–97. [Google Scholar] [CrossRef] [PubMed]
  125. Matson, V.; Fessler, J.; Bao, R.; Chongsuwat, T.; Zha, Y.; Alegre, M.L.; Luke, J.J.; Gajewski, T.F. The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients. Science 2018, 359, 104–108. [Google Scholar] [CrossRef]
  126. Gopalakrishnan, V.; Spencer, C.; McQuade, J.; Andrews, M.; Helmink, B.; Cogdill, A. The gut microbiome of metastatic melanoma patients initiating systemic therapy is influenced by host factors including diet, probiotic and antibiotic use. In Proceedings of the Annual Meeting of the Society for Immunotherapy of Cancer SITC, San Diego, CA, USA, 1–5 November 2023; pp. 7–11. [Google Scholar]
  127. Vétizou, M.; Pitt, J.M.; Daillère, R.; Lepage, P.; Waldschmitt, N.; Flament, C.; Rusakiewicz, S.; Routy, B.; Roberti, M.P.; Duong, C.P.; et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science 2015, 350, 1079–1084. [Google Scholar] [CrossRef] [PubMed]
  128. Chaput, N.; Lepage, P.; Coutzac, C.; Soularue, E.; Le Roux, K.; Monot, C.; Boselli, L.; Routier, E.; Cassard, L.; Collins, M.; et al. Baseline gut microbiota predicts clinical response and colitis in metastatic melanoma patients treated with ipilimumab. Ann. Oncol. 2017, 28, 1368–1379. [Google Scholar] [CrossRef]
  129. Clinicaltrials.Gov. Identifier NCT03643289, Predicting Response to Immunotherapy for Melanoma with Gut Microbiome and Metabolomics (PRIMM). Available online: https://clinicaltrials.gov/study/NCT03643289 (accessed on 13 February 2024).
  130. Clinicaltrials.Gov. Identifier NCT04107168, Microbiome Immunotherapy Toxicity and Response Evaluation. Available online: https://clinicaltrials.gov/study/NCT04107168 (accessed on 13 February 2024).
  131. Clinicaltrials.Gov. Identifier NCT03341143, Fecal Microbiota Transplant (FMT) in Melanoma Patients. Available online: https://clinicaltrials.gov/study/NCT03341143 (accessed on 13 February 2024).
  132. Ogino, S.; Nowak, J.A.; Hamada, T.; Milner, D.A., Jr.; Nishihara, R. Insights into Pathogenic Interactions Among Environment, Host, and Tumor at the Crossroads of Molecular Pathology and Epidemiology. Annu. Rev. Pathol. 2019, 14, 83–103. [Google Scholar] [CrossRef]
  133. Clinicaltrials.Gov. Identifier NCT03817125, Melanoma Checkpoint and Gut Microbiome Alteration with Microbiome Intervention (MCGRAW). Available online: https://clinicaltrials.gov/study/NCT03817125?cond=NCT03817125&rank=1 (accessed on 13 February 2024).
  134. Mima, K.; Kosumi, K.; Baba, Y.; Hamada, T.; Baba, H.; Ogino, S. The microbiome, genetics, and gastrointestinal neoplasms: The evolving field of molecular pathological epidemiology to analyze the tumor-immune-microbiome interaction. Hum. Genet. 2021, 140, 725–746. [Google Scholar] [CrossRef] [PubMed]
  135. Ogino, S.; Nishihara, R.; VanderWeele, T.J.; Wang, M.; Nishi, A.; Lochhead, P.; Qian, Z.R.; Zhang, X.; Wu, K.; Nan, H.; et al. Review Article: The Role of Molecular Pathological Epidemiology in the Study of Neoplastic and Non-neoplastic Diseases in the Era of Precision Medicine. Epidemiology 2016, 27, 602–611. [Google Scholar] [CrossRef]
  136. Chaturvedi, V.K.; Singh, A.; Singh, V.K.; Singh, M.P. Cancer Nanotechnology: A New Revolution for Cancer Diagnosis and Therapy. Curr. Drug Metab. 2019, 20, 416–429. [Google Scholar] [CrossRef]
  137. Chaudhuri, P.; Soni, S.; Sengupta, S. Single-walled carbon nanotube-conjugated chemotherapy exhibits increased therapeutic index in melanoma. Nanotechnology 2010, 21, 025102. [Google Scholar] [CrossRef]
  138. Yoncheva, K.; Merino, M.; Shenol, A.; Daskalov, N.T.; Petkov, P.S.; Vayssilov, G.N.; Garrido, M.J. Optimization and in-vitro/in-vivo evaluation of doxorubicin-loaded chitosan-alginate nanoparticles using a melanoma mouse model. Int. J. Pharm. 2019, 556, 1–8. [Google Scholar] [CrossRef]
  139. Xu, J.; Wang, H.; Xu, L.; Chao, Y.; Wang, C.; Han, X.; Dong, Z.; Chang, H.; Peng, R.; Cheng, Y.; et al. Nanovaccine based on a protein-delivering dendrimer for effective antigen cross-presentation and cancer immunotherapy. Biomaterials 2019, 207, 1–9. [Google Scholar] [CrossRef]
  140. Cai, J.; Zheng, Q.; Huang, H.; Li, B. 5-aminolevulinic acid mediated photodynamic therapy inhibits survival activity and promotes apoptosis of A375 and A431 cells. Photodiagnosis Photodyn. Ther. 2018, 21, 257–262. [Google Scholar] [CrossRef]
  141. Zhao, B.; Yin, J.J.; Bilski, P.J.; Chignell, C.F.; Roberts, J.E.; He, Y.Y. Enhanced photodynamic efficacy towards melanoma cells by encapsulation of Pc4 in silica nanoparticles. Toxicol. Appl. Pharmacol. 2009, 241, 163–172. [Google Scholar] [CrossRef]
  142. Porosnicu, I.; Butnaru, C.M.; Tiseanu, I.; Stancu, E.; Munteanu, C.V.A.; Bita, B.I.; Duliu, O.G.; Sima, F. Y2O3 Nanoparticles and X-ray Radiation-Induced Effects in Melanoma Cells. Molecules 2021, 26, 3403. [Google Scholar] [CrossRef]
  143. Maresca, L.; Stecca, B.; Carrassa, L. Novel Therapeutic Approaches with DNA Damage Response Inhibitors for Melanoma Treatment. Cells 2022, 11, 1466. [Google Scholar] [CrossRef]
  144. Fratangelo, F.; Camerlingo, R.; Carriero, M.V.; Pirozzi, G.; Palmieri, G.; Gentilcore, G.; Ragone, C.; Minopoli, M.; Ascierto, P.A.; Motti, M.L. Effect of ABT-888 on the apoptosis, motility and invasiveness of BRAFi-resistant melanoma cells. Int. J. Oncol. 2018, 53, 1149–1159. [Google Scholar] [CrossRef]
  145. Raineri, A.; Prodomini, S.; Fasoli, S.; Gotte, G.; Menegazzi, M. Influence of onconase in the therapeutic potential of PARP inhibitors in A375 malignant melanoma cells. Biochem. Pharmacol. 2019, 167, 173–181. [Google Scholar] [CrossRef]
  146. Rodríguez, M.I.; Peralta-Leal, A.; O’Valle, F.; Rodriguez-Vargas, J.M.; Gonzalez-Flores, A.; Majuelos-Melguizo, J.; López, L.; Serrano, S.; de Herreros, A.G.; Rodríguez-Manzaneque, J.C.; et al. PARP-1 regulates metastatic melanoma through modulation of vimentin-induced malignant transformation. PLoS Genet. 2013, 9, e1003531. [Google Scholar] [CrossRef] [PubMed]
  147. Middleton, M.R.; Friedlander, P.; Hamid, O.; Daud, A.; Plummer, R.; Falotico, N.; Chyla, B.; Jiang, F.; McKeegan, E.; Mostafa, N.M.; et al. Randomized phase II study evaluating veliparib (ABT-888) with temozolomide in patients with metastatic melanoma. Ann. Oncol. 2015, 26, 2173–2179. [Google Scholar] [CrossRef] [PubMed]
  148. Plummer, R.; Lorigan, P.; Steven, N.; Scott, L.; Middleton, M.R.; Wilson, R.H.; Mulligan, E.; Curtin, N.; Wang, D.; Dewji, R.; et al. A phase II study of the potent PARP inhibitor, Rucaparib (PF-01367338, AG014699), with temozolomide in patients with metastatic melanoma demonstrating evidence of chemopotentiation. Cancer Chemother. Pharmacol. 2013, 71, 1191–1199. [Google Scholar] [CrossRef] [PubMed]
  149. Magnussen, G.I.; Holm, R.; Emilsen, E.; Rosnes, A.K.R.; Slipicevic, A.; Flørenes, V.A. High Expression of Wee1 Is Associated with Poor Disease-Free Survival in Malignant Melanoma: Potential for Targeted Therapy. PLoS ONE 2012, 7, e38254. [Google Scholar] [CrossRef] [PubMed]
  150. Guertin, A.D.; Li, J.; Liu, Y.; Hurd, M.S.; Schuller, A.G.; Long, B.; Hirsch, H.A.; Feldman, I.; Benita, Y.; Toniatti, C.; et al. Preclinical evaluation of the WEE1 inhibitor MK-1775 as single-agent anticancer therapy. Mol. Cancer Ther. 2013, 12, 1442–1452. [Google Scholar] [CrossRef] [PubMed]
  151. Hirai, H.; Arai, T.; Okada, M.; Nishibata, T.; Kobayashi, M.; Sakai, N.; Imagaki, K.; Ohtani, J.; Sakai, T.; Yoshizumi, T.; et al. MK-1775, a small molecule Wee1 inhibitor, enhances anti-tumor efficacy of various DNA-damaging agents, including 5-fluorouracil. Cancer Biol. Ther. 2010, 9, 514–522. [Google Scholar] [CrossRef]
  152. Bridges, K.A.; Hirai, H.; Buser, C.A.; Brooks, C.; Liu, H.; Buchholz, T.A.; Molkentine, J.M.; Mason, K.A.; Meyn, R.E. MK-1775, a Novel Wee1 Kinase Inhibitor, Radiosensitizes p53-Defective Human Tumor Cells. Clin. Cancer Res. 2011, 17, 5638–5648. [Google Scholar] [CrossRef]
  153. Hirai, H.; Iwasawa, Y.; Okada, M.; Arai, T.; Nishibata, T.; Kobayashi, M.; Kimura, T.; Kaneko, N.; Ohtani, J.; Yamanaka, K.; et al. Small-molecule inhibition of Wee1 kinase by MK-1775 selectively sensitizes p53-deficient tumor cells to DNA-damaging agents. Mol. Cancer Ther. 2009, 8, 2992–3000. [Google Scholar] [CrossRef]
  154. Giunta, E.; Belli, V.; Napolitano, S.; De Falco, V.; Vitiello, P.; Terminiello, M.; Caputo, V.; Vitale, P.; Zanaletti, N.; Ciardiello, D. 13P Synergistic activity of PARP inhibitor and ATR inhibitor in melanoma cell lines may depend on BRAF-V600 mutation status. Ann. Oncol. 2020, 31, S248–S249. [Google Scholar] [CrossRef]
  155. Kim, S.T.; Smith, S.A.; Mortimer, P.; Loembé, A.-B.; Cho, H.; Kim, K.-M.; Smith, C.; Willis, S.; Irurzun-Arana, I.; Berges, A. Phase I study of ceralasertib (AZD6738), a novel DNA damage repair agent, in combination with weekly paclitaxel in refractory cancer. Clin. Cancer Res. 2021, 27, 4700–4709. [Google Scholar] [CrossRef]
  156. Ashworth, A. ATR inhibitors and paclitaxel in melanoma. Clin. Cancer Res. 2021, 27, 4667–4668. [Google Scholar] [CrossRef]
  157. Dharanipragada, P.; Zhang, X.; Liu, S.; Lomeli, S.H.; Hong, A.; Wang, Y.; Yang, Z.; Lo, K.Z.; Vega-Crespo, A.; Ribas, A.; et al. Blocking Genomic Instability Prevents Acquired Resistance to MAPK Inhibitor Therapy in Melanoma. Cancer Discov. 2023, 13, 880–909. [Google Scholar] [CrossRef] [PubMed]
  158. Kreidieh, F.Y.; Tawbi, H.A. The introduction of LAG-3 checkpoint blockade in melanoma: Immunotherapy landscape beyond PD-1 and CTLA-4 inhibition. Ther. Adv. Med. Oncol. 2023, 15, 17588359231186027. [Google Scholar] [CrossRef]
  159. Lipson, E.J.; Tawbi, H.A.-H.; Schadendorf, D.; Ascierto, P.A.; Matamala, L.; Gutiérrez, E.C.; Rutkowski, P.; Gogas, H.; Lao, C.D.; Janoski de Menezes, J.; et al. Relatlimab (RELA) plus nivolumab (NIVO) versus NIVO in first-line advanced melanoma: Primary phase III results from RELATIVITY-047 (CA224-047). J. Clin. Oncol. 2021, 39, 9503. [Google Scholar] [CrossRef]
  160. Tawbi, H.A.; Schadendorf, D.; Lipson, E.J.; Ascierto, P.A.; Matamala, L.; Castillo Gutiérrez, E.; Rutkowski, P.; Gogas, H.J.; Lao, C.D.; De Menezes, J.J.; et al. Relatlimab and Nivolumab versus Nivolumab in Untreated Advanced Melanoma. N. Engl. J. Med. 2022, 386, 24–34. [Google Scholar] [CrossRef] [PubMed]
  161. Jaiswal, A.; Verma, A.; Dannenfelser, R.; Melssen, M.; Tirosh, I.; Izar, B.; Kim, T.G.; Nirschl, C.J.; Devi, K.S.P.; Olson, W.C., Jr.; et al. An activation to memory differentiation trajectory of tumor-infiltrating lymphocytes informs metastatic melanoma outcomes. Cancer Cell 2022, 40, 524–544.e525. [Google Scholar] [CrossRef] [PubMed]
  162. Julve, M.; Furness, A.J. Advances in the development of tumor-infiltrating lymphocyte therapy for advanced melanoma. Expert. Opin. Biol. Ther. 2023, 23, 319–323. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Genetic mutations in melanoma.
Figure 1. Genetic mutations in melanoma.
Ijms 25 02984 g001
Table 1. Molecular Biomarkers in Melanoma.
Table 1. Molecular Biomarkers in Melanoma.
BiomarkerCommentsReferences
BRAFV600E MutationResponsive to BRAF inhibitors. Indicates early diagnosis, staging, and prediction of therapy responses.[41,42,43]
NRAS MutationLinked to shorter survival times in stage IV melanoma. Less common, but significant.[44,45,46]
c-KIT MutationNot closely associated with histological subtypes or tumor stage. Prevalent in older patients, acral mucosal melanoma, and sun-damaged areas.[47,48,49,50,51]
NF1 MutationLinked to a poorer prognosis than other mutation patterns.[52,53]
PMCA4 Transcript LevelsHigh levels in females are associated with longer progression-free survival (PFS) and improved prognosis, especially following PD-1 blockade therapy.[54]
Tumor Mutational Burden (TMB)High TMB might correlate with increased effectiveness of immune checkpoint inhibitor (ICI) treatments.[55,56,57]
Gene Expression Profiling (GEP)GEP tests in melanoma provide prognostic data on recurrence and metastasis risk.[58,59]
Gut MicrobiomeIntestinal microbiota composition affects response to ICI therapy. Greater diversity, particularly Ruminococcaceae subspecies, is associated with better outcomes.[60,61]
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

Kim, H.J.; Kim, Y.H. Molecular Frontiers in Melanoma: Pathogenesis, Diagnosis, and Therapeutic Advances. Int. J. Mol. Sci. 2024, 25, 2984. https://doi.org/10.3390/ijms25052984

AMA Style

Kim HJ, Kim YH. Molecular Frontiers in Melanoma: Pathogenesis, Diagnosis, and Therapeutic Advances. International Journal of Molecular Sciences. 2024; 25(5):2984. https://doi.org/10.3390/ijms25052984

Chicago/Turabian Style

Kim, Hyun Jee, and Yeong Ho Kim. 2024. "Molecular Frontiers in Melanoma: Pathogenesis, Diagnosis, and Therapeutic Advances" International Journal of Molecular Sciences 25, no. 5: 2984. https://doi.org/10.3390/ijms25052984

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