Next Article in Journal
Molecular Characterization of Atypical Fibroxanthoma and Pleomorphic Dermal Sarcoma
Previous Article in Journal
The Roles of STAT3 and STAT5 in Breast Cancer
Previous Article in Special Issue
Prognostic Role of Clinicopathological Characteristics and Serum Markers in Metastatic Melanoma Patients Treated with BRAF and MEK Inhibitors
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Skin Photodamage and Melanomagenesis: A Comprehensive Review

by
Michele Manganelli
1,†,
Giorgio Stabile
2,3,†,
Camila Scharf
4,*,
Antonio Podo Brunetti
2,3,
Giovanni Paolino
3,5,
Roberta Giuffrida
6,
Gianmarco Diego Bigotto
2,3,
Giuseppe Damiano
7,
Santo Raffaele Mercuri
3,5,
Fabio Sallustio
8,
Eleonora Mangano
9,
Roberta Bordoni
9,
Paola De Nardi
10,
Gabriella Guida
1,
Caterina Foti
11,
Giuseppe Argenziano
4,
Caterina Longo
12,13,
Giovanni Pellacani
14,
Nathalie Rizzo
15,
Vincenzo Russo
7,
Stefania Guida
2,3,‡ and
Franco Rongioletti
2,3,‡
add Show full author list remove Hide full author list
1
Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, 70121 Bari, Italy
2
Dermatology Clinic, IRCCS San Raffaele Hospital, 20132 Milan, Italy
3
Faculty of Medicine, Vita-Salute San Raffaele University, 20132 Milan, Italy
4
Dermatology Unit, Department of Mental and Physical Health and Preventive Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
5
Dermatology and Cosmetology, San Raffaele Hospital, 20132 Milano, Italy
6
Department of Clinical & Experimental Medicine, Section of Dermatology, University of Messina, 98122 Messina, Italy
7
Immuno-Biotherapy of Melanoma and Solid Tumors Unit, Division of Experimental Oncology, San Raffaele Scientific Institute, DIBIT, Via Olgettina 58, 20132 Milan, Italy
8
Department of Precision and Regenerative Medicine and Ionian Area, Aldo Moro University of Bari, 70121 Bari, Italy
9
Institute of Biomedical Technologies (ITB), National Research Council (CNR), Segrate, 20054 Milan, Italy
10
Colorectal Surgery, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
11
Section of Dermatology and Venereology, Department of Precision and Regenerative Medicine and Jonian Area, University of Bari “Aldo Moro”, 70124 Bari, Italy
12
Department of Dermatology, University of Modena and Reggio Emilia, 41121 Modena, Italy
13
Skin Cancer Center, Azienda Unità Sanitaria Locale, IRCCS di Reggio Emilia, 42122 Reggio Emilia, Italy
14
Department of Dermatology, Sapienza University, 00185 Rome, Italy
15
Department of Pathology, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
These authors contributed equally to this work.
Cancers 2025, 17(11), 1784; https://doi.org/10.3390/cancers17111784
Submission received: 23 April 2025 / Revised: 18 May 2025 / Accepted: 21 May 2025 / Published: 26 May 2025
(This article belongs to the Special Issue Novel Research on the Diagnosis and Treatment of Melanoma)

Simple Summary

Melanoma is the most aggressive form of skin cancer, and its increasing incidence is a major public health concern. One of the main causes of melanoma is exposure to ultraviolet radiation from the sun, which damages the DNA in skin cells and increases the risk of cancer. However, genetics also play a role in determining who is more likely to develop this disease. This overview aims to explore how ultraviolet radiation damages skin cells at a molecular level, how the body tries to repair this damage, and why these repair processes sometimes fail, leading to melanoma. The study also examines different types of melanoma that develop in sun-exposed skin and how they are classified based on the amount of sun damage they have accumulated over time. By improving our understanding of these processes, this research could help develop better strategies for preventing melanoma and identifying individuals at higher risk, ultimately leading to earlier detection and improved treatment decisions.

Abstract

Melanoma, the most aggressive form of skin cancer, still represents a significant and growing public health concern. Ultraviolet radiation (UVR) is considered the primary driver of melanoma, although genetic predisposition also plays a critical role. This review explores the intricate molecular mechanisms by which UVR-induced photodamage contributes to melanoma development. We examine epidemiological evidence linking UV exposure to increased risk, detailing how UVR damage to DNA triggers inflammatory responses and impairs DNA repair mechanisms. Specifically, we discuss the roles of nucleotide excision repair (NER) and base excision repair (BER) in mitigating UV damage. The review further explores diagnostic and surgical implications for melanomas arising on sun-exposed skin. By synthesizing current evidence, this overview aims to deepen understanding of the complex relationship between UVR, photodamage, and melanoma, shedding light on the need for personalized preventive strategies to better stratify the risk and introduce behavioral changes to reduce skin photodamage.

1. Introduction

Melanoma is one of the most aggressive and rapidly increasing skin cancers, originating from melanocytes, the pigment-producing cells of the skin. While a subset of melanomas develops in sun-protected areas, and approximately 10% of cases have a genetic predisposition, ultraviolet radiation (UVR) remains the leading environmental factor in cutaneous melanoma (CM) development, accounting for 60–70% of cases [1].
UVR, including UVA and UVB radiation, is a well-established carcinogen that induces extensive skin cell damage and plays a pivotal role in melanomagenesis [1]. They have distinct but overlapping carcinogenic roles: while UVB directly causes DNA mutations, UVA contributes through oxidative stress and immune evasion. Recent studies have elucidated the molecular pathways by which UV-induced photodamage contributes to tumorigenesis. These processes involve both direct DNA damage and subsequent inflammatory responses, leading to a cascade of events that promote melanocyte transformation [2,3].
DNA mutations accumulate in key regulatory genes, disrupting cellular processes such as cell cycle regulation, apoptosis, and DNA repair [4]. In response to UV-induced damage, skin cells employ repair mechanisms such as nucleotide excision repair (NER) and base excision repair (BER) pathways [5,6,7,8,9,10,11,12]. However, chronic UV exposure can overwhelm these pathways, leading to persistent inflammation, microenvironmental changes, and immune evasion, all of which contribute to melanoma development [13]. Molecular mechanisms underlying melanoma progression highlighted the pivotal role of V600BRAF mutation in switching on metabolic reprogramming [14]. Furthermore, the induction of autophagy has been suggested to be a pro-survival mechanism for melanoma cells [15,16,17]. While an initial increase in autophagy due to UVR might be a cellular defense mechanism, a dysregulated or sustained high level of autophagic flux in melanoma cells could paradoxically contribute to their survival, growth, and spread [1].
Usually, melanoma progresses following a characteristic sequence from the radial growth phase (RGP) to the vertical growth phase (VGP). In the RGP, atypical melanocytes proliferate laterally within the epidermis or superficial dermis, remaining in situ and non-invasive. The transition to the VGP marks a critical event, where melanoma cells gain the ability to invade deeper dermal layers and potentially metastasize. This shift is often driven by cumulative UVR-induced mutations and microenvironmental changes. Understanding the molecular signals regulating this progression is essential for early diagnosis and therapeutic intervention [18].
This review will explore the mechanisms linking UV-induced photodamage to melanoma, focusing on key epidemiological, molecular, and pathophysiological aspects. Additionally, we will examine diagnostic challenges, surgical considerations, and controversies regarding the role of chronic sun exposure in melanoma pathogenesis. By synthesizing epidemiological trends, DNA repair mechanisms, inflammation, and immunological consequences of UVR exposure, this review provides an integrated framework connecting photodamage to melanomagenesis. Particular emphasis is placed on the translational relevance of molecular alterations, including their implications for diagnosis, biomarker development, and therapeutic responsiveness. This comprehensive approach aims to support a deeper understanding of the complex interplay between UVR, skin aging, and melanoma, ultimately informing both preventive strategies and personalized clinical management.

2. Epidemiology

The incidence of CM has risen dramatically in recent decades, posing an increasing public health concern. CM is most commonly diagnosed between the ages of 50 and 60, with a mean age of 59 years [19]. Gender differences are evident, with men more frequently developing melanoma on the trunk and upper limbs, whereas women are more prone to melanomas on the lower extremities [20]. Skin type is also a key risk factor, with Fitzpatrick type I individuals—who are more susceptible to photodamage—at the highest risk [21].
Large-scale epidemiological studies, such as GLOBOCAN [22] and the Nurses’ Health Study [23], have demonstrated a strong correlation between sun exposure and melanoma incidence. According to GLOBOCAN 2020, melanoma accounts for over 324,000 new cases and 57,000 deaths globally each year. European incidence rates vary widely, with the highest observed in northern countries such as Norway and Sweden, and comparatively lower—but steadily increasing—rates in southern countries. In Italy, data from the AIRTUM registry (Associazione Italiana Registri Tumori) estimate an incidence of approximately 12–15 new melanoma cases per 100,000 inhabitants annually, with an upward trend particularly among men and older individuals [24]. These findings highlight the need for region-specific strategies to support targeted prevention campaigns and early detection, particularly in aging populations with high cumulative UVR exposure.
Chronic UV exposure, particularly in populations with high cumulative exposure, is associated with a significant increase in melanoma risk, especially in situ and head and neck melanomas, mainly those classified as lentigo maligna/lentigo maligna melanoma (LM/LMM) [25]. However, some studies suggest a paradoxical protective effect of continuous occupational sun exposure, (OR = 0.57) [26] possibly due to increased melanin production and epidermal thickness, through adaptive photoprotection mechanisms.
Instead, intermittent sun exposure, particularly episodes of intense exposure leading to sunburns, has been identified as a stronger risk factor than chronic exposure. Studies have reported an odds ratio (OR) of 3.00 for individuals with frequent sunbathing habits and 3.90 for those with a history of sunburns [26]. These findings highlight the complex interplay between different patterns of sun exposure and melanoma risk and its understanding is crucial to refining preventive strategies and improving early diagnosis.

3. UVR-Induced DNA Damage

UVR is the most significant environmental factor driving melanoma development, with its ability to induce DNA damage and disrupt cellular homeostasis in the skin. The UVR spectrum comprises three main wavebands: UVA (315–400 nm), UVB (280–315 nm), and UVC (200–280 nm). While UVC is completely absorbed by the stratospheric ozone layer (O3) and thus biologically irrelevant, both UVA and UVB reach the Earth’s surface and contribute to skin carcinogenesis [27].
The effects of UVR on DNA are well-documented, yet the distinct pathways through which UVA and UVB contribute to melanomagenesis remain a topic of ongoing research. UVB, with its higher energy, directly affects DNA by inducing cyclobutane pyrimidine dimers (CPDs) and 6-4 photoproducts (6-4PPs), leading to helix distortions that impair normal replication and transcription [28,29,30,31]. If left unrepaired, these lesions accumulate and generate UVB signature mutations, specifically C>T and CC>TT transitions, frequently observed in key tumor suppressor genes such as TP53 and CDKN2A [32,33,34,35].
On the other hand, UVA radiation, though less energetic, penetrates deeper into the dermis, and exerts its carcinogenic effects through reactive oxygen species (ROS) generation. The increase in superoxide radical ions (O2) upon UVA exposure sets off a cascade of oxidative stress, leading to secondary DNA damage [36]. The enzymatic activity of NADPH oxidase (NOX) further contributes to this oxidative burst, while inducible nitric oxide synthase (iNOS) generates nitric oxide (NO), which reacts with superoxide to produce peroxynitrite (ONOO), a highly reactive molecule that diffuses into the cell and induces damage [37]. The interaction between ONOO and melanin monomers leads to the formation of dioxetane intermediates, which undergo spontaneous thermolysis, producing highly energetic carbonyl compounds. These compounds, in turn, transfer UV energy directly to DNA, leading to additional CPD formation [38].
The role of melanin in this process is particularly intriguing. While eumelanin, the black-brown pigment, provides a degree of photoprotection by absorbing and dissipating UV energy, pheomelanin, the red-yellow pigment, contributes to ROS generation, paradoxically enhancing UVR-induced DNA damage [39]. Studies have demonstrated that melanocytes rich in pheomelanin accumulate twice as many CPDs as their eumelanin-producing counterparts, a phenomenon particularly relevant in individuals with red hair. These individuals frequently carry MC1R polymorphisms, which not only reduce eumelanin production but also impair DNA repair mechanisms, further amplifying melanoma risk [40]. Beyond its photoprotective limitations, pheomelanin also participates in melanin-mediated photochemistry, contributing to the generation of “dark CPDs”, a unique class of lesions that persist long after UV exposure has ceased, extending the window of potential DNA damage and mutation accumulation [41].

3.1. UVB Radiation in Melanoma Development

UVB radiation plays a pivotal role in melanoma development, with its direct DNA-damaging effects and disruption of repair pathways acting as key carcinogenic mechanisms. In addition to CPD and 6-4PP formation, UVB-induced mutations in TP53 and CDKN2A disrupt critical tumor suppressor pathways, promoting unchecked melanocyte proliferation [42,43,44]. Moreover, MC1R signaling, a crucial pathway for both pigmentation and DNA repair, is significantly affected by UVB exposure [41]. When functional, MC1R enhances the efficiency of NER by recruiting repair proteins to sites of photodamage. However, in individuals carrying loss-of-function MC1R variants, this protective mechanism is weakened, leading to inefficient repair of UVB-induced mutations and an increased susceptibility to melanoma [40].

3.2. UVA Radiation in Melanoma Development

While UVB initiates melanoma by directly damaging DNA, UVA, in turn, contributes to melanomagenesis through oxidative stress and mitochondrial dysfunction [45]. The generation of ROS and oxidative DNA lesions, such as 7,8-dihydro-8-oxyguanine (8-oxoG), frequently leads to G>C to T>A transversions, a mutation pattern commonly found in melanoma genomes [46,47,48,49]. Furthermore, UVA exposure promotes the photoisomerization of 6-4PPs into Dewar valence isomers, extending the persistence of DNA damage beyond the initial UV insult [50,51]. Another significant aspect of UVA-induced damage is its impact on mitochondrial integrity. By disrupting mitochondrial function, UVA radiation can trigger the release of pro-apoptotic factors such as cytochrome c, activating intrinsic apoptotic pathways. However, in melanocytes that evade apoptosis, persistent mitochondrial dysfunction fosters genomic instability, a hallmark of tumor initiation and progression [52].

4. Infrared Radiation (IRA)/UVR Cross-Talk in Melanoma Development

Beyond the contributions of UVA and UVB, infrared radiation (IR) has also been implicated in modifying UVR-induced skin cancers. Although IR itself is not inherently mutagenic, evidence suggests that infrared A (IRA, 780–1400 nm), the predominant terrestrial IR component, may interfere with apoptosis and enhance the survival of UV-damaged melanocytes [53]. IRA has been shown to reduce UVB-induced apoptosis by modulating the extrinsic apoptotic pathway, inhibiting caspase-8 activation, and altering the expression of apoptosis-related proteins (BID, BAX, Bcl2, and FLIPL). While this may protect normal skin cells from excessive UVR-induced damage, it also allows DNA-damaged melanocytes to evade cell death, increasing the likelihood of mutation accumulation and malignant transformation. Notably, IRA does not appear to affect UVB-induced DNA repair, meaning that while damaged cells survive longer, their DNA remains unrepaired, further promoting melanomagenesis [54,55,56]. The influence of IRA on UVR-induced ROS in human melanocytes needs further investigation.

5. Genetic Predisposition

While UVR-induced DNA damage is a major driver of melanoma, genetic factors also play a significant role in determining individual susceptibility to the disease. Melanoma arises from a complex interplay between inherited (germline) mutations and acquired (somatic) mutations that are pivotal in predisposing individuals to melanoma (Table 1) [40,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103]. These mutations affect a variety of cellular components, including genes involved in pigmentation, key tumor suppressor genes and oncogenes, crucial signaling pathways, and other important players, such as transcription factors and epigenetic modifications [104,105,106,107].
Mutations in the MC1R gene are among the most well-established genetic risk factors for melanoma. They not only increase UV sensitivity but also impair DNA repair, leading to a twofold increase in melanoma risk. Moreover, MC1R mutations have a synergistic effect with other melanoma-associated mutations, particularly in CDKN2A and BRAF, further amplifying melanoma risk [40,58].
Nonetheless, TP53, one of the most frequently mutated genes in cancer, is also commonly affected in melanoma. UVR-induced mutations in TP53 impair its function as a tumor suppressor, allowing for unchecked cell proliferation and increased survival of UV-damaged melanocytes [108,109]. Additionally, mutations in the NF1 gene, often observed in melanomas arising on chronically sun-exposed skin [77], lead to hyperactivation of the NRAS–MAPK pathway and increased resistance to apoptosis [78].
Another critical pathway involved in melanoma pathogenesis is the RAS–RAF–MEK–ERK (MAPK) signaling pathway. Activating mutations in BRAF, particularly the V600E mutation, are found in approximately 50% of melanomas. This mutation drives constitutive activation of MEK and ERK, promoting melanocyte proliferation and survival. While BRAF mutation is a frequent initiating event that activates the MAPK pathway, the inactivation of key tumor suppressor genes, such as CDKN2A, PTEN, and TP53, is crucial for overcoming BRAF-induced senescence and allowing the uncontrolled proliferation and survival that characterize melanoma. Importantly, UVR exposure leads to impaired TP53 function, accelerating the progression of BRAF-driven melanoma [108,110].
Mutations in NRAS and KIT similarly enhance MAPK signaling, contributing to tumor growth and metastasis [111].
Finally, epigenetic modifications, such as DNA methylation and histone modification, further complicate the genetic landscape of melanoma. Aberrant methylation of tumor suppressor genes and oncogenes has been implicated in melanoma progression, with specific methylation signatures correlating with increased metastatic potential [112].

6. Cellular Response to UV-Induced DNA Damage

DNA damage can be addressed through two fundamental repair mechanisms: (1) direct damage repair and (2) removal and replacement of damaged DNA regions. While photoreactivation (a FAD-dependent direct repair mechanism) occurs in bacteria [105], human cells rely primarily on excision and replacement strategies (Figure 1). Interestingly, UVR triggers α-MSH release from keratinocytes and melanocytes [106,107,108,109,110,111], thus contributing to the activation of the downstream signaling pathways that modulate NER and BER to enhance genomic stability and resist UV-mediated apoptosis [5,6,7,8,9,10,11,12].

6.1. Nucleotide Excision Repair (NER)

NER serves as the primary repair mechanism for UV-induced DNA damage through two sub-pathways: (1) Global Genomic NER (GG-NER), in which the UV-DDB initially recognizes DNA lesions and recruits the XPC-HR23B complex to the site of the lesion; and (2) Transcription-Coupled NER (TC-NER), in which stalled RNA polymerase II recruits repair machinery [112].
The repair process involves unwinding the DNA helix by XPD (ERCC2) and XPB (ERCC3) helicases, excision of the damaged strand by XPF-ERCC1 and XPG endonucleases, followed by DNA synthesis using the undamaged strand as a template and ligation.
NER regulation involves damage sensors (XPC-HR23B, RNAPII/CSB, ATM/ATR) that trigger downstream signaling through p53, CHK1/2, and BRCA1, affecting cell cycle checkpoints and repair [113]. Post-translational modifications (phosphorylation, ubiquitination, acetylation) coordinate repair protein activity. NER integrates with cell cycle checkpoints through ATM/ATR activation of TP53 and CHK1/CHK2, arresting cells at G1/S, while BRCA1/2 proteins facilitate repair during the S and G2/M phases [114,115].

6.2. Base Excision Repair (BER)

Although UVR primarily causes pyrimidine dimers (repaired by NER), it can also induce oxidative stress, leading to base lesions repaired by BER. It operates through two sub-pathways: (1) Short-Patch BER, which replaces a single nucleotide using DNA polymerase β and DNA ligase III/XRCC1; and (2) Long-Patch BER, which synthesizes 2–10 nucleotides using polymerases δ/ε, PCNA, FEN1, and DNA ligase I [116,117,118]. BER is tightly regulated through DNA damage sensors, repair proteins, and post-translational modifications [119].
This process involves (1) DNA glycosylases removing damaged bases to generate AP sites; (2) APE1 creating single-strand breaks at AP sites [120]; (3) DNA polymerase β filling the nucleotide gap [121]; and (4) DNA ligase sealing the strand break.
Also, for oxidative damage like 8-oxoG, which pairs with adenine and causes mutations, MUTYH removes incorrect adenines while OGG1 excises 8-oxoG paired with cytosine [1]. PNKP processes damaged DNA ends through 3′-phosphatase and 5′-kinase activities to generate suitable ends for repair [122].

6.3. Translesion Synthesis (TLS)

TLS is a DNA damage tolerance mechanism that allows replication to bypass lesions (like UV-induced thymine dimers or AP sites) that would otherwise stall replication, leading to genomic instability, cell death, or mutations. It uses specialized, error-prone TLS polymerases to allow replication to proceed despite the presence of lesions [123]. When replicative polymerase encounters a lesion, PCNA is ubiquitinated by RAD6/RAD18, triggering polymerase switching to specialized TLS polymerases. Several TLS polymerases exist, each with specific properties. For instance, Pol ζ bypasses 6-4 photoproducts with high fidelity [124], while Pol η efficiently bypasses CPDs, though deaminated cytosine residues in CPDs lead to C→T signature mutations [125].
The incorporation of incorrect nucleotide bases across from UVR-induced DNA lesions by error-prone DNA polymerases during TLS is mutagenic. The mismatch repair proteins Msh2/Msh6 recognize incorrect nucleotides incorporated during TLS and trigger their removal, creating single-stranded DNA patches that must be filled before replication to prevent double-strand breaks and apoptosis [126]. Moreover, Msh2/Msh6 deficiency increases mutation frequency.

7. UVR-Mediated Inflammation and Immunosuppression in Skin Carcinogenesis

Chronic UVR exposure causes skin aging and significantly increases the risk of skin cancer. UVR-absorbing chromophores initiate a series of biochemical and immunologic events that lead to UVR-induced injury. In the initial phase, keratinocytes undergo apoptosis in large numbers primarily by the activation TP53 [127] and the death receptor CD95/Fas [128]. These apoptotic keratinocytes release pro-inflammatory cytokines into the skin microenvironment, including TNF-α, IL-1α, IL-1β, GM-CSF, IL-6, IL-8, and IL-10 [129]. Along with these, alarmins also contribute to inflammation. Alarmins are intracellular molecules that act as pro-inflammatory mediators in the extracellular milieu upon damage. When released, alarmins bind pattern recognition receptors (PRRs) or other surface receptors, resulting in NF-κB activation and subsequent pro-inflammatory cytokine production. Indeed, evidence has highlighted that UV radiation stimulates HMGB1 release in keratinocytes in vitro and HMGB1 is expressed in skin tumors after chronic radiation [130,131]. These cytokines play a crucial role in orchestrating the inflammatory response and subsequent tissue repair. The recruitment of macrophages via the CCR2/CCL2 axis (chemokine receptor CCR2 and its ligand CCL2) is a critical step in establishing a pro-tumorigenic microenvironment [132].
In addition to its inflammatory effects, UVR is a potent immunosuppressant in the skin. Immunosuppression begins locally in the irradiated area but can have systemic effects [133]. Both UVA and UVB radiation impact the immune system, although most studies on UVR-induced immunosuppression have focused on UVB [134,135]. Under normal circumstances, the transformed cells are recognized and eliminated by anti-tumor immune responses elicited by the host. However, the strong immunosuppressive effects of UVR can allow these transformed cells to evade immune surveillance [133,136,137]. Furthermore, during melanoma progression, melanoma cells are able to avoid immune response via the programmed cell death-1/programmed cell death ligand-1 (PD-1/PD-L1) pathway. Activated T cells and tumor-infiltrating lymphocytes are blocked by the binding of PD-1 with PD-L1 induced by tumor cells, which act as a negative factor for immunomodulation [138].

8. Pathogenesis of Melanoma: Diagnostic and Surgical Controversies

8.1. Diagnostic Aspects

In the diagnostic context of melanoma, factors related to UV exposure and skin phenotype, as well as genetic factors, are critical to establishing screening schedules for early melanoma detection [139]. Considering the aggressive nature of invasive melanoma, timely diagnosis significantly improves patient outcomes. Risk assessment should include sun exposure history, particularly childhood sunburns and adult tanning habits; phenotypic traits, as fair skin, red or blonde hair, and light eyes indicate increased susceptibility; and high nevus count and genetic factors, as, for instance, MC1R gene variants, associated with red hair phenotype, confer increased melanoma risk due to reduced protective pigmentation [33]. Multivariate risk models that integrate these factors enhance stratification accuracy, though reliance on self-reported exposure may introduce bias [140,141].
In this context, dysplastic nevi—while not direct melanoma precursors—are considered important clinical markers of elevated melanoma risk. They may display some architectural disorder and cytologic atypia, but most do not progress to melanoma. Their presence, especially in patients with multiple lesions or a family history of melanoma, supports intensified surveillance strategies. UVR exposure in these individuals contributes to cumulative mutational burden rather than a linear dysplastic nevus–melanoma progression pathway.
Advanced non-invasive imaging techniques, such as dermoscopy and reflectance confocal microscopy (RCM), have transformed non-invasive melanoma diagnosis. They have improved early melanoma detection rates, particularly for atypical presentations on photodamaged skin through widely consolidated assessment algorithms [142,143,144,145]. RCM offers cellular-level visualization, and it increases sensitivity and confidence in diagnosis, as proven by a recent randomized controlled trial [146]. However, diagnosis can be challenging due to skin variations associated with skin photodamage and potential overlapping features between benign and malignant lesions. Nevertheless, histopathological analysis represents the gold standard for diagnosing melanocytic lesions, although distinguishing lesions arising on photoexposed skin remains challenging [147,148].
Currently, artificial intelligence (AI) applications in melanoma detection have shown promising results, with convolutional neural networks demonstrating diagnostic accuracy comparable or superior to dermatologists in controlled studies [149]. These systems analyze dermoscopic images using deep learning algorithms trained on extensive datasets. Total-body photography combined with AI-assisted sequential digital dermoscopy enables comprehensive monitoring of high-risk patients, facilitating detection of subtle changes indicative of early melanoma. Mobile applications leveraging these technologies may expand access to preliminary melanoma screening, though concerns regarding algorithmic bias, data privacy, and clinical integration remain significant challenges.

8.2. Molecular and Histopathologic Characteristics and Their Implications in Clinical Practice

8.2.1. Molecular Biomarkers in Clinical Practice

Recent advances in molecular diagnostics have led to the integration of several key genetic and circulating biomarkers in melanoma care.
These include BRAF, NRAS, and NF1 mutations, which provide insight into tumor biology and guide therapeutic decisions. Liquid biopsy tools—such as circulating tumor DNA (ctDNA), microRNAs (miRs), and exosomes—allow for non-invasive monitoring. ctDNA levels correlate with tumor burden and treatment response, while miR signatures (e.g., miR-15b, miR-150, miR-425) are under investigation for early detection and prognostication.

8.2.2. Immunohistochemical Biomarkers

Immunohistochemistry (IHC) remains a cornerstone in melanoma diagnosis and classification. Markers such as PD-L1 help stratify patients for immune checkpoint therapies. Additionally, S100B and LDH serve as serum markers for disease monitoring in advanced stages, although their sensitivity in early-stage melanoma is limited. These molecular and IHC biomarkers not only aid in diagnosis but also reflect the tumor’s biological behavior and immune interactions.

8.2.3. Etiopathogenic and Molecular Divergence by Sun Exposure Pattern

These biomarker profiles also mirror the underlying etiopathogenic differences between melanomas arising on chronically sun-damaged (CSD) versus non-sun-damaged (non-CSD) skin. CSD melanomas exhibit higher mutational burdens dominated by UV signature mutations (C→T transitions), frequent activating mutations in NRAS and NF1 and the inactivation of tumor suppressors such as CDKN2A and TP53. These tumors typically develop through a gradual progression model from precursor lesions. In contrast, non-CSD melanomas show lower mutational burden, prevalent BRAFV600E mutations, and less genomic complexity, often arising without identifiable precursor lesions. These molecular distinctions have significant implications for prognosis and treatment response, with CSD melanomas generally demonstrating greater heterogeneity and potential for immune recognition [150,151,152,153,154,155,156,157,158,159].
This molecular divergence aligns with the recent World Health Organization (WHO) histopathological classification, which categorizes melanoma into high and low CSD subtypes. High CSD melanoma includes lentigo maligna and desmoplastic melanoma, while low CSD melanoma primarily includes superficial spreading melanoma, further highlighting distinct etiopathogenic and genomic profiles [160,161].
For instance, tumor mutational burden (TMB) and immunotherapy response in melanoma represent a critical aspect of precision oncology. UV-induced melanomas typically exhibit high TMB, generating numerous neoantigens that serve as targets for immune recognition. Clinical evidence demonstrates superior responses to immune checkpoint inhibitors (anti-PD-1/PD-L1, anti-CTLA-4) in melanomas with higher TMB, particularly those with UV signature mutations. This correlation provides a mechanistic explanation for the paradoxical observation that melanomas with indicators of greater sun damage often show improved survival outcomes with immunotherapy [162]. However, response patterns remain complex, with factors beyond TMB, including tumor microenvironment composition and immune exclusion mechanisms, significantly modulating treatment efficacy. Ongoing research aims to develop predictive biomarkers that integrate mutational signatures with immune infiltration patterns to optimize immunotherapeutic approaches.

8.3. Surgical Management

Furthermore, surgical management of melanoma in photodamaged skin requires tailored approaches, particularly in chronically sun-exposed regions such as the face, upper limbs, and shoulders, due to cosmetic and functional reasons [163]. Accurate assessment of lesion depth, evaluation of margins, and surveillance after excision are essential in managing melanoma, especially those arising in chronically sun-damaged skin [164].

8.4. Controversies

Despite significant progress in understanding the link between melanoma and photodamage, controversies persist, particularly regarding the role of sun exposure in melanoma pathogenesis. While intermittent, high-intensity UV exposure—especially in childhood—clearly elevates melanoma risk, chronic sun exposure in outdoor workers has shown inconsistent associations with melanoma development. Paradoxically, melanoma incidence has increased more among indoor workers, leading researchers to explore other contributing factors like genetic predisposition and artificial tanning [165]. Another point of debate concerns the fact that, despite the increase in solar exposure over the years, only a small portion of new melanoma diagnoses are classified as LM, the subtype most closely associated with sun exposure. This can be explained from various points of view. LM typically develops over many years due to chronic sun exposure. Consequently, even with increased exposure, it may take a long time for it to manifest. This means that the current diagnosis rates may not fully reflect the recent increases in UV exposure.
However, it is important to note that primary prevention efforts targeting UV exposure have shown mixed results in modulating melanoma incidence trends. Educational campaigns promoting sun-protective behaviors have increased public awareness but demonstrated limited impact on behavior modification. Structural interventions including shade provision in public spaces and UV index warnings have shown greater effectiveness. Also, school-based programs targeting children have demonstrated promising results in establishing early sun-protective habits, while workplace policies for outdoor workers remain inconsistently implemented. Notably, Australia’s comprehensive “SunSmart” program, combining education, environmental changes, and policy development, has achieved stabilization of melanoma rates among younger cohorts, providing evidence that long-term, multi-level interventions can effectively reduce melanoma incidence [166].
Finally, other factors, such as genetic mutations and environmental factors, may contribute to the prevalence of different melanoma subtypes, leading to a more significant number of cases that are not classified as LM. Finally, while overall UV exposure may have increased, changes in lifestyle and sun protection behaviors can impact the incidence of specific melanoma subtypes. For instance, using sunscreen, protective clothing, and shade-seeking behaviors may contribute to a lower incidence of LM, even in those with considerable sun exposure. In summary, while increased solar exposure contributes to the risk of melanoma, the relationship between UV exposure and specific melanoma subtypes is complex and influenced by various biological, behavioral, and environmental factors.

8.5. Future Directions and Clinical Perspectives

Despite ongoing debate around the precise role of chronic sun exposure in melanoma pathogenesis, future research should aim to delineate the molecular events occurring during the earliest phases of UV-induced photodamage. Markers of oxidative DNA damage, such as 8-oxoG and persistent CPDs, have been proposed as indicators of photogenotoxic stress and mutational load in pre-neoplastic skin [167]. Additionally, pro-inflammatory alarmins (e.g., HMGB1) and cytokines such as IL-1β, TNF-α, and IL-6 are increasingly recognized as molecular mediators of the tumor-promoting microenvironment following chronic UV exposure [13,130]. Alterations in immunosenescence-related pathways and Langerhans cell depletion have also been implicated in the failure of immune surveillance in photoexposed skin [168].
Clinically, the integration of liquid biopsy technologies—including ctDNA, UV-induced miRNAs, and exosomal RNA profiles—with non-invasive diagnostic imaging and AI-supported skin surveillance tools could enable earlier melanoma detection in high-risk individuals [169]. Moving forward, the development of integrated biomarker panels, combining genomic, epigenomic, inflammatory, and immune features, may enable a personalized prevention and early detection model, particularly in populations with high cumulative sun damage.

9. Conclusions

UVR plays a pivotal role in melanomagenesis by inducing DNA damage, promoting oxidative stress, and creating a pro-inflammatory and immunosuppressive skin microenvironment. Despite the presence of DNA repair systems such as NER and BER, chronic UVR exposure often overwhelms these defenses, contributing to mutational burden, immune evasion, and the transformation of melanocytes. Recent insights into the molecular and histopathologic divergence between melanomas arising on CSD versus non-sun-damaged skin have revealed important genomic and immunologic differences, with significant implications for prognosis and therapeutic responsiveness—particularly to immunotherapies.
Advanced molecular and immunohistochemical biomarkers, including ctDNA, miRNAs, BRAF/NRAS/NF1 mutations, and PD-L1 expression, are increasingly integrated into diagnostic and management pathways. These tools, together with non-invasive imaging and AI-supported surveillance, pave the way for earlier detection and personalized prevention strategies.
Controversies persist regarding the role of chronic sun exposure and melanoma subtype prevalence, but ongoing research into photodamage-associated biomarkers and UVR-driven molecular signatures holds promise for resolving these uncertainties. Looking ahead, the development of integrated biomarker panels, capable of detecting early photoinduced molecular alterations, will be critical to improving outcomes. A refined understanding of the intersection between UVR, genetic susceptibility, immune modulation, and diagnostic technologies is essential to advancing melanoma prevention, risk stratification, and treatment.

Author Contributions

Conceptualization, S.G. and F.R.; methodology, C.S.; software, P.D.N., G.P. (Giovanni Paolino) and R.B.; validation, G.G., V.R. and G.D.; formal analysis, G.S., A.P.B., G.D.B. and C.F.; investigation, G.S., A.P.B., G.D.B., N.R. and R.G.; resources, G.S., A.P.B. and G.D.B.; data curation, G.S., A.P.B. and G.D.B.; writing—original draft preparation, G.S. and M.M.; writing—review and editing, S.G., C.S. and M.M.; visualization, P.D.N., S.R.M., F.S., E.M., G.P. (Giovanni Pellacani) and C.L.; supervision, G.A. and F.R.; project administration, F.R. 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 declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
6-4PPs6-4 photoproducts
8-oxodGuo7,8-dihydro-8-oxyguanine
AIArtificial Intelligence
BERBase Excision Repair
CDPsCyclobutane Pyrimidine Dimers
CMCutaneous Melanoma
CSDChronically Sun-Damaged
ctDNACirculating Tumor DNA
iNOSInducible Nitric Oxide Synthase
IRInfrared Radiation
LMLentigo Maligna
LMMLentigo Maligna Melanoma
NONitric Oxide
NOXNADPH Oxidase
NERNucleotide Excision Repair
O2Superoxide Radical Ions
O3Ozone
ONOOPeroxynitrite
RCMReflectance Confocal Microscopy
ROSReactive Oxygen Species
TLSTranslesion Synthesis
TMBTumoral Mutation Burden
UVRUltraviolet Radiation

References

  1. Sample, A.; He, Y.Y. Mechanisms and Prevention of UV-Induced Melanoma. Photodermatol. Photoimmunol. Photomed. 2018, 34, e12329. [Google Scholar] [CrossRef] [PubMed]
  2. Guida, S.; Ciardo, S.; De Pace, B.; De Carvalho, N.; Peccerillo, F.; Manfredini, M.; Farnetani, F.; Chester, J.; Kaleci, S.; Manganelli, M.; et al. The Influence of MC1R on Dermal Morphological Features of Photo-Exposed Skin in Women Revealed by Reflectance Confocal Microscopy and Optical Coherence Tomography. Exp. Dermatol. 2019, 28, 1321–1327. [Google Scholar] [CrossRef]
  3. Guida, S.; Ciardo, S.; De Pace, B.; De Carvalho, N.; Farnetani, F.; Pezzini, C.; Chester, J.; Shaniko, K.; Manganelli, M.; Guida, G.; et al. Atrophic and Hypertrophic Skin Photoaging and Melanocortin-1 Receptor (MC1R): The Missing Link. J. Am. Acad. Dermatol. 2021, 84, 187–190. [Google Scholar] [CrossRef]
  4. Tang, X.; Yang, T.; Yu, D.; Xiong, H.; Zhang, S. Current Insights and Future Perspectives of Ultraviolet Radiation (UV) Exposure: Friends and Foes to the Skin and Beyond the Skin. Environ. Int. 2024, 185, 108535. [Google Scholar] [CrossRef] [PubMed]
  5. Kadekaro, A.L.; Wakamatsu, K.; Ito, S.; Abdel-Malek, Z.A. Cutaneous Photoprotection and Melanoma Susceptibility: Reaching Beyond Melanin Content to the Frontiers of DNA Repair. Front. Biosci. 2006, 11, 2157–2173. [Google Scholar] [CrossRef]
  6. Schärer, O.D. Nucleotide Excision Repair in Eukaryotes. Cold Spring Harb. Perspect. Biol. 2013, 5, a012609. [Google Scholar] [CrossRef] [PubMed]
  7. Sugasawa, K. Molecular Mechanisms of DNA Damage Recognition for Mammalian Nucleotide Excision Repair. DNA Repair 2016, 44, 110–117. [Google Scholar] [CrossRef]
  8. Gillet, L.C.J.; Schärer, O.D. Molecular Mechanisms of Mammalian Global Genome Nucleotide Excision Repair. Chem. Rev. 2006, 106, 253–276. [Google Scholar] [CrossRef]
  9. Spivak, G. Nucleotide Excision Repair in Humans. DNA Repair 2015, 36, 13–19. [Google Scholar] [CrossRef]
  10. Li, X.; Mao, W.; Chen, J.; Goding, C.R.; Cui, R.; Xu, Z.X.; Miao, X. The Protective Role of MC1R in Chromosome Stability and Centromeric Integrity in Melanocytes. Cell Death Discov. 2021, 7, 89. [Google Scholar] [CrossRef]
  11. Jhappan, C.; Noonan, F.P.; Merlino, G. Ultraviolet Radiation and Cutaneous Malignant Melanoma. Oncogene 2003, 22, 3092–3099. [Google Scholar] [CrossRef] [PubMed]
  12. Khan, A.Q.; Travers, J.B.; Kemp, M.G. Roles of UVA Radiation and DNA Damage Responses in Melanoma Pathogenesis. Environ. Mol. Mutagen. 2018, 59, 506–521. [Google Scholar] [CrossRef]
  13. Ciążyńska, M.; Olejniczak-Staruch, I.; Sobolewska-Sztychny, D.; Narbutt, J.; Skibińska, M.; Lesiak, A. Ultraviolet Radiation and Chronic Inflammation—Molecules and Mechanisms Involved in Skin Carcinogenesis: A Narrative Review. Life 2021, 11, 326. [Google Scholar] [CrossRef] [PubMed]
  14. Ferretta, A.; Maida, I.; Guida, S.; De Benedetto, G.; Menga, A.; Castegna, A. New Insight into the Role of Metabolic Reprogramming in Melanoma Cells Harboring BRAF Mutations. Biochim. Biophys. Acta (BBA) Mol. Cell Res. 2016, 1863, 2710–2718. [Google Scholar] [CrossRef]
  15. Ma, X.H.; Piao, S.; Wang, D.; McAfee, Q.; Nathanson, K.L.; Lum, J.J.; Li, L.Z.; Hu, J.; Lindsten, T.; Thompson, C.B.; et al. Measurements of Tumor Cell Autophagy Predict Invasiveness, Resistance to Chemotherapy, and Survival in Melanoma. Clin. Cancer Res. 2011, 17, 3478–3489. [Google Scholar] [CrossRef] [PubMed]
  16. Ma, X.H.; Piao, S.F.; Dey, S.; McAfee, Q.; Karakousis, G.; Villanueva, J.; Hart, L.S.; Levi, S.; Hu, J.; Zhang, G.; et al. Targeting ER Stress-Induced Autophagy Overcomes BRAF Inhibitor Resistance in Melanoma. J. Clin. Investig. 2014, 124, 1406–1417. [Google Scholar] [CrossRef]
  17. Rebecca, V.W.; Massaro, R.R.; Fedorenko, I.V.; Fang, B.; Wood, E.R.; Goodison, S.; Fulp, W.J.; Sondak, V.K.; Smalley, K.S.M. Inhibition of Autophagy Enhances the Effects of the AKT Inhibitor MK-2206 When Combined with Paclitaxel and Carboplatin in BRAF Wild-Type Melanoma. Pigment. Cell Melanoma Res. 2014, 27, 465–478. [Google Scholar] [CrossRef]
  18. Testa, U.; Castelli, G.; Pelosi, E. Melanoma: Genetic Abnormalities, Tumor Progression, Clonal Evolution and Tumor Initiating Cells. Med. Sci. 2017, 5, 28. [Google Scholar] [CrossRef]
  19. Conforti, C.; Zalaudek, I. Epidemiology and Risk Factors of Melanoma: A Review. Dermatol. Pract. Concept. 2021, 11, 2021161S. [Google Scholar] [CrossRef]
  20. Schwartz, M.R.; Luo, L.; Berwick, M. Sex Differences in Melanoma. Curr. Epidemiol. Rep. 2019, 6, 112–118. [Google Scholar] [CrossRef]
  21. MacKie, R.M.; Hauschild, A.; Eggermont, A.M.M. Epidemiology of Invasive Cutaneous Melanoma. Ann. Oncol. 2009, 20 (Suppl. S4), vi1–vi7. [Google Scholar] [CrossRef]
  22. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef]
  23. Walls, A.C.; Han, J.; Li, T.; Qureshi, A.A. Host Risk Factors, Ultraviolet Index of Residence, and Incident Malignant Melanoma in Situ Among US Women and Men. Am. J. Epidemiol. 2013, 177, 997–1005. [Google Scholar] [CrossRef] [PubMed]
  24. Airtum Working Group. I Numeri del Cancro in Italia 2020; Intermedia Editore: Milan, Italy, 2020; ISBN 9788897714785. [Google Scholar]
  25. Worrede, A.; Douglass, S.M.; Weeraratna, A.T. The Dark Side of Daylight: Photoaging and the Tumor Microenvironment in Melanoma Progression. J. Clin. Investig. 2021, 131, e143763. [Google Scholar] [CrossRef]
  26. Nelemans, P.J.; Groenendal, H.; Kiemeney, L.A.; Rampen, F.H.; Ruiter, D.J.; Verbeek, A.L. Effect of Intermittent Exposure to Sunlight on Melanoma Risk Among Indoor Workers and Sun-Sensitive Individuals. Environ. Health Perspect. 1993, 101, 252–255. [Google Scholar] [CrossRef]
  27. McKenzie, R.L.; Aucamp, P.J.; Bais, A.F.; Björn, L.O.; Ilyas, M. Changes in Biologically Active Ultraviolet Radiation Reaching the Earth’s Surface. Photochem. Photobiol. Sci. 2007, 6, 218–231. [Google Scholar] [CrossRef] [PubMed]
  28. Castellani, G.; Buccarelli, M.; Arasi, M.B.; Rossi, S.; Pisanu, M.E.; Bellenghi, M.; Lintas, C.; Tabolacci, C. BRAF Mutations in Melanoma: Biological Aspects, Therapeutic Implications, and Circulating Biomarkers. Cancers 2023, 15, 4026. [Google Scholar] [CrossRef] [PubMed]
  29. De Fabo, E.C.; Noonan, F.P.; Fears, T.; Merlino, G. Ultraviolet B but not Ultraviolet A Radiation Initiates Melanoma. Cancer Res. 2004, 64, 6372–6376. [Google Scholar] [CrossRef]
  30. von Thaler, A.K.; Kamenisch, Y.; Berneburg, M. The Role of Ultraviolet Radiation in Melanomagenesis. Exp. Dermatol. 2010, 19, 81–88. [Google Scholar] [CrossRef] [PubMed]
  31. Hennessey, R.C.; Holderbaum, A.M.; Bonilla, A.; Delaney, C.; Gillahan, J.E.; Tober, K.L.; Oberyszyn, T.M.; Zippin, J.H.; Burd, C.E. Ultraviolet Radiation Accelerates NRas-Mutant Melanomagenesis: A Cooperative Effect Blocked by Sunscreen. Pigment Cell Melanoma Res. 2017, 30, 477–487. [Google Scholar] [CrossRef]
  32. Besaratinia, A.; Yoon, J.; Schroeder, C.; Bradforth, S.E.; Cockburn, M.; Pfeifer, G.P. Wavelength Dependence of Ultraviolet Radiation-Induced DNA Damage as Determined by Laser Irradiation Suggests That Cyclobutane Pyrimidine Dimers Are the Principal DNA Lesions Produced by Terrestrial Sunlight. FASEB J. 2011, 25, 3079–3091. [Google Scholar] [CrossRef] [PubMed]
  33. You, Y.H.; Lee, D.H.; Yoon, J.H.; Nakajima, S.; Yasui, A.; Pfeifer, G.P. Cyclobutane Pyrimidine Dimers Are Responsible for the Vast Majority of Mutations Induced by UVB Irradiation in Mammalian Cells. J. Biol. Chem. 2001, 276, 44688–44694. [Google Scholar] [CrossRef] [PubMed]
  34. Jackson, S.P.; Bartek, J. The DNA-Damage Response in Human Biology and Disease. Nature 2009, 461, 1071–1078. [Google Scholar] [CrossRef] [PubMed]
  35. Pleasance, E.D.; Cheetham, R.K.; Stephens, P.J.; McBride, D.J.; Humphray, S.J.; Greenman, C.D.; Varela, I.; Lin, M.L.; Ordóñez, G.R.; Bignell, G.R.; et al. A Comprehensive Catalogue of Somatic Mutations from a Human Cancer Genome. Nature 2010, 463, 191–196. [Google Scholar] [CrossRef] [PubMed]
  36. Chedekel, M.R.; Smith, S.K.; Post, P.W.; Pokora, A.; Vessell, D.L. Photodestruction of Pheomelanin: Role of Oxygen. Proc. Natl. Acad. Sci. USA 1978, 75, 5395–5399. [Google Scholar] [CrossRef]
  37. Roméro-Graillet, C.; Aberdam, E.; Biagoli, N.; Massabni, W.; Ortonne, J.P.; Ballotti, R. Ultraviolet B Radiation Acts through the Nitric Oxide and cGMP Signal Transduction Pathway to Stimulate Melanogenesis in Human Melanocytes. J. Biol. Chem. 1996, 271, 28052–28056. [Google Scholar] [CrossRef]
  38. Lamola, A.A. Production of Pyrimidine Dimers in DNA in the Dark. Biochem. Biophys. Res. Commun. 1971, 43, 893–898. [Google Scholar] [CrossRef]
  39. Premi, S.; Wallisch, S.; Mano, C.M.; Weiner, A.B.; Bacchiocchi, A.; Wakamatsu, K.; Bechara, E.J.H.; 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]
  40. Tagliabue, E.; Gandini, S.; Bellocco, R.; Maisonneuve, P.; Newton-Bishop, J.; Polsky, D.; Lazovich, D.; Kanetsky, P.; Ghiorzo, P.; Gruis, N.; et al. MC1R Variants as Melanoma Risk Factors Independent of At-Risk Phenotypic Characteristics: A Pooled Analysis from the M-SKIP Project. Cancer Manag. Res. 2018, 10, 1143–1154. [Google Scholar] [CrossRef]
  41. Jarrett, S.G.; Wolf Horrell, E.M.; Boulanger, M.C.; D’Orazio, J.A. Defining the Contribution of MC1R Physiological Ligands to ATR Phosphorylation at Ser435, a Predictor of DNA Repair in Melanocytes. J. Investig. Dermatol. 2015, 135. [Google Scholar] [CrossRef]
  42. Tu, Y.; Dammann, R.; Pfeifer, G.P. Sequence and Time-Dependent Deamination of Cytosine Bases in UVB-Induced Cyclobutane Pyrimidine Dimers in Vivo. J. Mol. Biol. 1998, 284, 297–311. [Google Scholar] [CrossRef] [PubMed]
  43. Rivlin, N.; Brosh, R.; Oren, M.; Rotter, V. Mutations in the p53 Tumor Suppressor Gene: Important Milestones at the Various Steps of Tumorigenesis. Genes Cancer 2011, 2, 466–474. [Google Scholar] [CrossRef] [PubMed]
  44. Sun, X.; Zhang, N.; Yin, C.; Zhu, B.; Li, X. Ultraviolet Radiation and Melanomagenesis: From Mechanism to Immuno-Therapy. Front. Oncol. 2020, 10, 373. [Google Scholar] [CrossRef]
  45. Fadadu, R.P.; Wei, M.L. Ultraviolet A Radiation Exposure and Melanoma: A Review. Melanoma Res. 2022, 32, 166–172. [Google Scholar] [CrossRef]
  46. Noonan, F.P.; Zaidi, M.R.; Wolnicka-Glubisz, A.; Anver, M.R.; Bahn, J.; Wielgus, A.; Cadet, J.; Douki, T.; Mouret, S.; Tucker, M.A.; et al. Melanoma Induction by Ultraviolet A but Not Ultraviolet B Radiation Requires Melanin Pigment. Nat. Commun. 2012, 3, 884. [Google Scholar] [CrossRef]
  47. Beani, J.C. Ultraviolet A-induced DNA Damage: Role in Skin Cancer. Bull. Acad. Natl. Med. 2014, 198, 273–295. [Google Scholar]
  48. Kvam, E.; Tyrrell, R.M. The Role of Melanin in the Induction of Oxidative DNA Base Damage by Ultraviolet A Irradiation of DNA or Melanoma Cells. J. Investig. Dermatol. 1999, 113, 209–213. [Google Scholar] [CrossRef]
  49. Bachelor, M.A.; Bowden, G.T. UVA-Mediated Activation of Signaling Pathways Involved in Skin Tumor Promotion and Progression. Semin. Cancer Biol. 2004, 15, 25–32. [Google Scholar] [CrossRef]
  50. Cadet, J.; Sage, E.; Douki, T. Ultraviolet Radiation-Mediated Damage to Cellular DNA. Mutat. Res. 2005, 571, 3–17. [Google Scholar] [CrossRef]
  51. Cadet, J.; Grand, A.; Douki, T. Solar UV Radiation-Induced DNA Bipyrimidine Photoproducts: Formation and Mechanistic Insights. In Photoinduced Phenomena in Nucleic Acids II; Topics in Current Chemistry; Springer: Berlin/Heidelberg, Germany, 2015; Volume 356. [Google Scholar] [CrossRef]
  52. Zhang, X.; Li, H.; Liu, C.; Yuan, X. Role of ROS Mediated Autophagy in Melanoma (Review). Mol. Med. Rep. 2022, 26, 303. [Google Scholar] [CrossRef]
  53. Calles, C.; Schneider, M.; MacAluso, F.; Benesova, T.; Krutmann, J.; Schroeder, P. Infrared A Radiation Influences the Skin Fibroblast Transcriptome: Mechanisms and Consequences. J. Investig. Dermatol. 2010, 130, 1524–1536. [Google Scholar] [CrossRef] [PubMed]
  54. Cho, S.; Shin, M.H.; Kim, Y.K.; Seo, J.E.; Lee, Y.M.; Park, C.H.; Chung, J.H. Effects of Infrared Radiation and Heat on Human Skin Aging in Vivo. J. Investig. Dermatol. Symp. Proc. 2009, 14, 15–19. [Google Scholar] [CrossRef] [PubMed]
  55. Jantschitsch, C.; Majewski, S.; Maeda, A.; Schwarz, T.; Schwarz, A. Infrared Radiation Confers Resistance to UV-Induced Apoptosis via Reduction of DNA Damage and Upregulation of Antiapoptotic Proteins. J. Investig. Dermatol. 2009, 129, 1271–1279. [Google Scholar] [CrossRef]
  56. Kimeswenger, S.; Schwarz, A.; Födinger, D.; Müller, S.; Pehamberger, H.; Schwarz, T.; Jantschitsch, C. Infrared A Radiation Promotes Survival of Human Melanocytes Carrying Ultraviolet Radiation-Induced DNA Damage. Exp. Dermatol. 2016, 25, 447–452. [Google Scholar] [CrossRef]
  57. Gudbjartsson, D.F.; Sulem, P.; Stacey, S.N.; Goldstein, A.M.; Rafnar, T.; Sigurgeirsson, B.; Benediktsdottir, K.R.; Thorisdottir, K.; Ragnarsson, R.; Sveinsdottir, S.G.; et al. ASIP and TYR Pigmentation Variants Associate with Cutaneous Melanoma and Basal Cell Carcinoma. Nat. Genet. 2008, 40, 886–891. [Google Scholar] [CrossRef]
  58. Puig-Butille, J.A.; Escámez, M.J.; Garcia-Garcia, F.; Tell-Marti, G.; Fabra, À.; Martínez-Santamaría, L.; Badenas, C.; Aguilera, P.; Pevida, M.; Dopazo, J.; et al. Capturing the Biological Impact of CDKN2A and MC1R Genes as an Early Predisposing Event in Melanoma and Non-Melanoma Skin Cancer. Oncotarget 2014, 5, 1439–1451. [Google Scholar] [CrossRef] [PubMed]
  59. Hussussian, C.J.; Struewing, J.P.; Goldstein, A.M.; Higgins, P.A.T.; Ally, D.S.; Sheahan, M.D.; Clark, W.H.; Tucker, M.A.; Dracopoli, N.C. Germline p16 Mutations in Familial Melanoma. Nat. Genet. 1994, 8, 15–21. [Google Scholar] [CrossRef]
  60. De Summa, S.; Lasorella, A.; Strippoli, S.; Giudice, G.; Guida, G.; Elia, R.; Nacchiero, E.; Azzariti, A.; Silvestris, N.; Guida, M.; et al. The Genetic Germline Background of Single and Multiple Primary Melanomas. Front. Mol. Biosci. 2021, 7, 555630. [Google Scholar] [CrossRef]
  61. Puig, S.; Malvehy, J.; Badenas, C.; Ruiz, A.; Jimenez, D.; Cuellar, F.; Azon, A.; Gonzàlez, U.; Castel, T.; Campoy, A.; et al. Role of the CDKN2A Locus in Patients with Multiple Primary Melanomas. J. Clin. Oncol. 2005, 23, 3043–3051. [Google Scholar] [CrossRef]
  62. Liontos, M.; Pateras, S.; Evangelou, K.; Gorgoulis, V. The Tumor Suppressor Gene ARF as a Sensor of Oxidative Stress. Curr. Mol. Med. 2012, 12, 704–715. [Google Scholar] [CrossRef]
  63. Bishop, D.T.; Demenais, F.; Goldstein, A.M.; Bergman, W.; Bishop, J.N.; Bressac-De Paillerets, B.; Chompret, A.; Ghiorzo, P.; Gruis, N.; Hansson, J.; et al. Geographical Variation in the Penetrance of CDKN2A Mutations for Melanoma. J. Natl. Cancer Inst. 2002, 94, 894–903. [Google Scholar] [CrossRef] [PubMed]
  64. Goldstein, A.M.; Chan, M.; Harland, M.; Hayward, N.K.; Demenais, F.; Bishop, D.T.; Azizi, E.; Bergman, W.; Bianchi-Scarra, G.; Bruno, W.; et al. Features Associated with Germline CDKN2A Mutations: A GenoMEL Study of Melanoma-Prone Families from Three Continents. J. Med. Genet. 2007, 44, 99–106. [Google Scholar] [CrossRef] [PubMed]
  65. Lang, J.; Boxer, M.; MacKie, R.M. CDKN2A Mutations in Scottish Families with Cutaneous Melanoma: Results from 32 Newly Identified Families. Br. J. Dermatol. 2005, 153, 1121–1125. [Google Scholar] [CrossRef]
  66. Borges, A.L.; Cuéllar, F.; Puig-Butillé, J.A.; Scarone, M.; Delgado, L.; Badenas, C.; Milà, M.; Malvehy, J.; Barquet, V.; Núñez, J.; et al. CDKN2A Mutations in Melanoma Families from Uruguay. Br. J. Dermatol. 2009, 161, 536–541. [Google Scholar] [CrossRef]
  67. Yakobson, E.; Eisenberg, S.; Isacson, R.; Halle, D.; Levy-Lahad, E.; Catane, R.; Safro, M.; Sobolev, V.; Huot, T.; Peters, G.; et al. A Single Mediterranean, Possibly Jewish, Origin for the Val59Gly CDKN2A Mutation in Four Melanoma-Prone Families. Eur. J. Hum. Genet. 2003, 11, 288–296. [Google Scholar] [CrossRef] [PubMed]
  68. Majore, S.; De Simone, P.; Crisi, A.; Eibenschutz, L.; Binni, F.; Antigoni, I.; De Bernardo, C.; Catricalà, C.; Grammatico, P. CDKN2A/CDK4 Molecular Study on 155 Italian Subjects with Familial and/or Primary Multiple Melanoma. Pigment. Cell Melanoma Res. 2008, 21, 209–211. [Google Scholar] [CrossRef]
  69. Eliason, M.J.; Larson, A.A.; Florell, S.R.; Zone, J.J.; Cannon-Albright, L.A.; Samlowski, W.E.; Leachman, S.A. Population-Based Prevalence of CDKN2A Mutations in Utah Melanoma Families. J. Investig. Dermatol. 2006, 126, 660–666. [Google Scholar] [CrossRef]
  70. Box, N.F.; Duffy, D.L.; Chen, W.; Stark, M.; Martin, N.G.; Sturm, R.A.; Hayward, N.K. MC1R Genotype Modifies Risk of Melanoma in Families Segregating CDKN2A Mutations. Am. J. Hum. Genet. 2001, 69, 765–773. [Google Scholar] [CrossRef]
  71. Peters, G. Tumor Suppression for ARFicionados: The Relative Contributions of p16 INK4a and p14ARF in Melanoma. J. Natl. Cancer Inst. 2008, 100, 757–759. [Google Scholar] [CrossRef]
  72. Burkhart, D.L.; Sage, J. Cellular Mechanisms of Tumour Suppression by the Retinoblastoma Gene. Nat. Rev. Cancer 2008, 8, 671–682. [Google Scholar] [CrossRef]
  73. Li, W.; Sanki, A.; Karim, R.Z.; Thompson, J.F.; Lee, C.S.; Zhuang, L.; McCarthy, S.W.; Scolyer, R.A. The Role of Cell Cycle Regulatory Proteins in the Pathogenesis of Melanoma. Pathology 2006, 38, 287–301. [Google Scholar] [CrossRef] [PubMed]
  74. Aguissa-Touré, A.H.; Li, G. Genetic Alterations of PTEN in Human Melanoma. Cell. Mol. Life Sci. 2012, 69, 1475–1491. [Google Scholar] [CrossRef] [PubMed]
  75. Goel, V.K.; Lazar, A.J.F.; Warneke, C.L.; Redston, M.S.; Haluska, F.G. Examination of Mutations in BRAF, NRAS, and PTEN in Primary Cutaneous Melanoma. J. Investig. Dermatol. 2006, 126, 154–160. [Google Scholar] [CrossRef] [PubMed]
  76. Whittaker, S.R.; Theurillat, J.P.; Van Allen, E.; Wagle, N.; Hsiao, J.; Cowley, G.S.; Schadendorf, D.; Root, D.E.; Garraway, L.A. A Genome-Scale RNA Interference Screen Implicates NF1 Loss in Resistance to RAF Inhibition. Cancer Discov. 2013, 3, 350–362. [Google Scholar] [CrossRef]
  77. Nissan, M.H.; Pratilas, C.A.; Jones, A.M.; Ramirez, R.; Won, H.; Liu, C.; Tiwari, S.; Kong, L.; Hanrahan, A.J.; Yao, Z.; et al. Loss of NF1 in Cutaneous Melanoma Is Associated with RAS Activation and MEK Dependence. Cancer Res. 2014, 74, 2340–2350. [Google Scholar] [CrossRef]
  78. Kanavy, H.E.; Gerstenblith, M.R. Ultraviolet Radiation and Melanoma. Semin. Cutan. Med. Surg. 2011, 30, 222–228. [Google Scholar] [CrossRef] [PubMed]
  79. Mo, X.; Preston, S.; Zaidi, M.R. Macroenvironment-Gene-Microenvironment Interactions in Ultraviolet Radiation-Induced Melanomagenesis. Adv. Cancer Res. 2019, 144, 1–54. [Google Scholar]
  80. Davies, H.; Bignell, G.R.; Cox, C.; Stephens, P.; Edkins, S.; Clegg, S.; Teague, J.; Woffendin, H.; Garnett, M.J.; Bottomley, W.; et al. Mutations of the BRAF Gene in Human Cancer. Nature 2002, 417, 949–954. [Google Scholar] [CrossRef]
  81. Wan, P.T.C.; Garnett, M.J.; Roe, S.M.; Lee, S.; Niculescu-Duvaz, D.; Good, V.M.; Project, C.G.; Jones, C.M.; Marshall, C.J.; Springer, C.J.; et al. Mechanism of Activation of the RAF-ERK Signaling Pathway by Oncogenic Mutations of B-RAF. Cell 2004, 116, 855–867. [Google Scholar] [CrossRef]
  82. Garnett, M.J.; Marais, R. Guilty as Charged: B-RAF Is a Human Oncogene. Cancer Cell 2004, 6, 313–319. [Google Scholar] [CrossRef]
  83. 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] [PubMed]
  84. Bos, J.L. Ras Oncogenes in Human Cancer: A Review. Cancer Res. 1989, 49, 4682–4689. [Google Scholar]
  85. Burd, C.E.; Liu, W.; Huynh, M.V.; Waqas, M.A.; Gillahan, J.E.; Clark, K.S.; Fu, K.; Martin, B.L.; Jeck, W.R.; Souroullas, G.P.; et al. Mutation-Specific RAS Oncogenicity Explains NRAS Codon 61 Selection in Melanoma. Cancer Discov. 2014, 4, 1418–1429. [Google Scholar] [CrossRef]
  86. Wang, H.; Zhang, H.; Chen, Y.; Wang, H.; Tian, Y.; Yi, X.; Shi, Q.; Zhao, T.; Zhang, B.; Gao, T.; et al. Targeting Wnt/β-Catenin Signaling Exacerbates Ferroptosis and Increases the Efficacy of Melanoma Immunotherapy via the Regulation of MITF. Cells 2022, 11, 3580. [Google Scholar] [CrossRef] [PubMed]
  87. Dahl, C.; Guldberg, P. The Genome and Epigenome of Malignant Melanoma. Acta Pathol. Microbiol. Immunol. Scand. 2007, 115, 1161–1176. [Google Scholar] [CrossRef] [PubMed]
  88. Chin, L.; Garraway, L.A.; Fisher, D.E. Malignant Melanoma: Genetics and Therapeutics in the Genomic Era. Genes Dev. 2006, 20, 2149–2182. [Google Scholar] [CrossRef]
  89. Natali, P.G.; Nicotra, M.R.; Di Renzo, M.F.; Prat, M.; Bigotti, A.; Cavaliere, R.; Comoglio, P.M. Expression of the c-Met/HGF Receptor in Human Melanocytic Neoplasms: Demonstration of the Relationship to Malignant Melanoma Tumour Progression. Br. J. Cancer 1993, 68, 746–750. [Google Scholar] [CrossRef]
  90. Curtin, J.A.; Busam, K.; Pinkel, D.; Bastian, B.C. Somatic Activation of KIT in Distinct Subtypes of Melanoma. J. Clin. Oncol. 2006, 24, 4340–4346. [Google Scholar] [CrossRef]
  91. Stark, M.; Hayward, N. Genome-Wide Loss of Heterozygosity and Copy Number Analysis in Melanoma Using High-Density Single-Nucleotide Polymorphism Arrays. Cancer Res. 2007, 67, 2632–2642. [Google Scholar] [CrossRef]
  92. Nazarian, R.; Shi, H.; Wang, Q.; Kong, X.; Koya, R.C.; Lee, H.; Chen, Z.; Lee, M.K.; Attar, N.; Sazegar, H.; et al. Melanomas Acquire Resistance to B-RAF(V600E) Inhibition by RTK or N-RAS Upregulation. Nature 2010, 468, 973–977. [Google Scholar] [CrossRef]
  93. Kawakami, A.; Fisher, D.E. The Master Role of Microphthalmia-Associated Transcription Factor in Melanocyte and Melanoma Biology. Lab. Investig. 2017, 97, 649–656. [Google Scholar] [CrossRef]
  94. Garraway, L.A.; Widlund, H.R.; Rubin, M.A.; Getz, G.; Berger, A.J.; Ramaswamy, S.; Beroukhim, R.; Milner, D.A.; Granter, S.R.; Du, J.; et al. Integrative Genomic Analyses Identify MITF as a Lineage Survival Oncogene Amplified in Malignant Melanoma. Nature 2005, 436, 117–122. [Google Scholar] [CrossRef] [PubMed]
  95. Cheli, Y.; Ohanna, M.; Ballotti, R.; Bertolotto, C. Fifteen-Year Quest for Microphthalmia-Associated Transcription Factor Target Genes. Pigment. Cell Melanoma Res. 2010, 23, 27–40. [Google Scholar] [CrossRef]
  96. Yokoyama, S.; Woods, S.L.; Boyle, G.M.; Aoude, L.G.; MacGregor, S.; Zismann, V.; Gartside, M.; Cust, A.E.; Haq, R.; Harland, M.; et al. A Novel Recurrent Mutation in MITF Predisposes to Familial and Sporadic Melanoma. Nature 2011, 480, 99–103. [Google Scholar] [CrossRef]
  97. Lin, X.; Sun, R.; Zhao, X.; Zhu, D.; Zhao, X.; Gu, Q.; Dong, X.; Zhang, D.; Zhang, Y.; Li, Y.; et al. C-myc Overexpression Drives Melanoma Metastasis by Promoting Vasculogenic Mimicry via c-myc/Snail/Bax Signaling. J. Mol. Med. 2017, 95, 53–67. [Google Scholar] [CrossRef] [PubMed]
  98. Vance, K.W.; Carreira, S.; Brosch, G.; Goding, C.M. Tbx2 Is Overexpressed and Plays an Important Role in Maintaining Proliferation and Suppression of Senescence in Melanomas. Cancer Res. 2005, 65, 2260–2268. [Google Scholar] [CrossRef] [PubMed]
  99. Jönsson, G.; Dahl, C.; Staaf, J.; Sandberg, T.; Bendahl, P.O.; Ringnér, M.; Guldberg, P.; Borg, Å. Genomic Profiling of Malignant Melanoma Using Tiling-Resolution ArrayCGH. Oncogene 2007, 26, 4738–4748. [Google Scholar] [CrossRef]
  100. Macerola, E.; Loggini, B.; Giannini, R.; Garavello, G.; Giordano, M.; Proietti, A.; Niccoli, C.; Basolo, F.; Fontanini, G. Coexistence of TERT Promoter and BRAF Mutations in Cutaneous Melanoma Is Associated with More Clinicopathological Features of Aggressiveness. Virchows Arch. 2015, 467, 177–184. [Google Scholar] [CrossRef]
  101. Heidenreich, B.; Nagore, E.; Rachakonda, P.S.; Garcia-Casado, Z.; Requena, C.; Traves, V.; Becker, J.; Soufir, N.; Hemminki, K.; Kumar, R. Telomerase Reverse Transcriptase Promoter Mutations in Primary Cutaneous Melanoma. Nat. Commun. 2014, 5, 3401. [Google Scholar] [CrossRef]
  102. Vicente, A.L.S.A.; Novoloaca, A.; Cahais, V.; Awada, Z.; Cuenin, C.; Spitz, N.; Carvalho, A.L.; Evangelista, A.F.; Crovador, C.S.; Reis, R.M.; et al. Cutaneous and Acral Melanoma Cross-OMICs Reveals Prognostic Cancer Drivers Associated with Pathobiology and Ultraviolet Exposure. Nat. Commun. 2022, 13, 4115. [Google Scholar] [CrossRef]
  103. Trucco, L.D.; Mundra, P.A.; Hogan, K.; Garcia-Martinez, P.; Viros, A.; Mandal, A.K.; Macagno, N.; Gaudy-Marqueste, C.; Allan, D.; Baenke, F.; et al. Ultraviolet Radiation–Induced DNA Damage Is Prognostic for Outcome in Melanoma. Nat. Med. 2019, 25, 221–224. [Google Scholar] [CrossRef]
  104. Zhang, S.; Xiao, X.; Yi, Y.; Wang, X.; Zhu, L.; Shen, Y.; Lin, D.; Wu, C. Tumor Initiation and Early Tumorigenesis: Molecular Mechanisms and Interventional Targets. Signal Transduct. Target. Ther. 2024, 9, 149. [Google Scholar] [CrossRef] [PubMed]
  105. Shain, A.H.; Yeh, I.; Kovalyshyn, I.; Sriharan, A.; Talevich, E.; Gagnon, A.; Dummer, R.; North, J.; Pincus, L.; Ruben, B.; et al. The Genetic Evolution of Melanoma from Precursor Lesions. N. Engl. J Med. 2015, 373, 1926–1936. [Google Scholar] [CrossRef] [PubMed]
  106. Tang, J.; Fewings, E.; Chang, D.; Zeng, H.; Liu, S.; Jorapur, A.; Belote, R.L.; McNeal, A.S.; Tan, T.M.; Yeh, I.; et al. The Genomic Landscapes of Individual Melanocytes from Human Skin. Nature 2020, 586, 600–605. [Google Scholar] [CrossRef] [PubMed]
  107. Belote, R.L.; Le, D.; Maynard, A.; Lang, U.E.; Sinclair, A.; Lohman, B.K.; Planells-Palop, V.; Baskin, L.; Tward, A.D.; Darmanis, S.; et al. Human Melanocyte Development and Melanoma Dedifferentiation at Single-Cell Resolution. Nat. Cell Biol. 2021, 23, 1035–1047. [Google Scholar] [CrossRef]
  108. Viros, A.; Sanchez-Laorden, B.; Pedersen, M.; Furney, S.J.; Rae, J.; Hogan, K.; Ejiama, S.; Girotti, M.R.; Cook, M.; Dhomen, N.; et al. Ultraviolet Radiation Accelerates BRAF-Driven Melanomagenesis by Targeting TP53. Nature 2014, 511, 478–482. [Google Scholar] [CrossRef]
  109. Reichrath, J.; Rass, K. Ultraviolet Damage, DNA Repair and Vitamin D in Nonmelanoma Skin Cancer and in Malignant Melanoma: An Update. Adv. Exp. Med. Biol. 2014, 810, 208–233. [Google Scholar]
  110. Barón, A.; Zaba, B.; Green, J.; Jackson, A.C.J.; Rees, J.; Slominski, A. Mechanism of UV-Related Carcinogenesis and Its Contribution to Nevi/Melanoma. Expert Rev. Dermatol. 2007, 2, 451–469. [Google Scholar] [CrossRef]
  111. Inamdar, G.S.; Madhunapantula, S.V.; Robertson, G.P. Targeting the MAPK Pathway in Melanoma: Why Some Approaches Succeed and Others Fail. Biochem. Pharmacol. 2010, 80, 624–637. [Google Scholar] [CrossRef]
  112. Guo, W.; Xu, T.; Lee, J.J.; Murphy, G.F.; Lian, C.G. Epigenetic Markers in Melanoma. Melanoma Manag. 2015, 2, 367–382. [Google Scholar] [CrossRef]
  113. Zhang, M.; Wang, L.; Zhong, D. Photolyase: Dynamics and Electron-Transfer Mechanisms of DNA Repair. Arch. Biochem. Biophys. 2017, 632, 158–174. [Google Scholar] [CrossRef] [PubMed]
  114. Funasaka, Y.; Chakraborty, A.K.; Hayashi, Y.; Komoto, M.; Ohashi, A.; Nagahama, M.; Inoue, Y.; Pawelek, J.; Ichihashi, M. Modulation of Melanocyte-Stimulating Hormone Receptor Expression on Normal Human Melanocytes: Evidence for a Regulatory Role of Ultraviolet B, Interleukin-1α, Interleukin-1β, Endothelin-1 and Tumor Necrosis Factor-α. Br. J. Dermatol. 1998, 139, 216–224. [Google Scholar] [CrossRef] [PubMed]
  115. Kadekaro, A.L.; Kavanagh, R.; Kanto, H.; Terzieva, S.; Hauser, J.; Kobayashi, N.; Schwemberger, S.; Cornelius, J.; Babcock, G.; Shertzer, H.G.; et al. α-Melanocortin and Endothelin-1 Activate Antiapoptotic Pathways and Reduce DNA Damage in Human Melanocytes. Cancer Res. 2005, 65, 4292–4299. [Google Scholar] [CrossRef]
  116. Schiller, M.; Brzoska, T.; Böhm, M.; Metze, D.; Scholzen, T.E.; Rougier, A.; Luger, T.A. Solar-Simulated Ultraviolet Radiation-Induced Upregulation of the Melanocortin-1 Receptor, Proopiomelanocortin, and α-Melanocyte-Stimulating Hormone in Human Epidermis In Vivo. J. Investig. Dermatol. 2004, 122, 468–476. [Google Scholar] [CrossRef] [PubMed]
  117. Im, S.; Moro, O.; Peng, F.; Medrano, E.E.; Cornelius, J.; Babcock, G.; Nordlund, J.J.; Abdel-Malek, Z.A. Activation of the Cyclic AMP Pathway by α-Melanotropin Mediates the Response of Human Melanocytes to Ultraviolet B Radiation. Cancer Res. 1998, 58, 47–54. [Google Scholar]
  118. Manganelli, M.; Guida, S.; Ferretta, A.; Pellacani, G.; Porcelli, L.; Azzariti, A.; Guida, G. Behind the Scene: Exploiting MC1R in Skin Cancer Risk and Prevention. Genes 2021, 12, 1093. [Google Scholar] [CrossRef]
  119. Dong, L.; Wen, J.; Pier, E.; Zhang, X.; Zhang, B.; Dong, F.; Ziegler, N.; Mysz, M.; Armenta, R.; Cui, R. Melanocyte-Stimulating Hormone Directly Enhances UV-Induced DNA Repair in Keratinocytes by a Xeroderma Pigmentosum Group A-Dependent Mechanism. Cancer Res. 2010, 70, 3547–3556. [Google Scholar] [CrossRef]
  120. Budden, T.; Bowden, N.A. The Role of Altered Nucleotide Excision Repair and UVB-Induced DNA Damage in Melanomagenesis. Int. J. Mol. Sci. 2013, 14, 1132–1151. [Google Scholar] [CrossRef]
  121. Ouhtit, A.; Gupta, I.; Gaur, R.L.; Fernando, A.; Abd El-Azim, A.O.; Eid, A.; Becker, T.M. Deregulation of Cell Growth and Apoptosis in UV-Induced Melanomagenesis. Front. Biosci. Elit. 2020, 12, 223–236. [Google Scholar] [CrossRef]
  122. Sarkar, S.; Gaddameedhi, S. Solar Ultraviolet-Induced DNA Damage Response: Melanocytes’ Story in Transformation to Environmental Melanomagenesis. Environ. Mol. Mutagen. 2020, 61, 736–751. [Google Scholar] [CrossRef]
  123. Kao, H.I.; Henricksen, L.A.; Liu, Y.; Bambara, R.A. Cleavage Specificity of Saccharomyces cerevisiae Flap Endonuclease 1 Suggests a Double-Flap Structure as the Cellular Substrate. J. Biol. Chem. 2002, 277, 14379–14389. [Google Scholar] [CrossRef] [PubMed]
  124. Cappelli, E.; Taylor, R.; Cevasco, M.; Abbondandolo, A.; Caldecott, K.; Frosina, G. Involvement of XRCC1 and DNA Ligase III Gene Products in DNA Base Excision Repair. J. Biol. Chem. 1997, 272, 23970–23975. [Google Scholar] [CrossRef]
  125. Pascucci, B.; Stucki, M.; Jónsson, Z.O.; Dogliotti, E.; Hübscher, U. Long Patch Base Excision Repair with Purified Human Proteins. DNA Ligase I as Patch Size Mediator for DNA Polymerases δ and ε. J. Biol. Chem. 1999, 274, 33696–33702. [Google Scholar] [CrossRef] [PubMed]
  126. Limpose, K.L.; Corbett, A.H.; Doetsch, P.W. BERing the Burden of Damage: Pathway Crosstalk and Posttranslational Modification of Base Excision Repair Proteins Regulate DNA Damage Management. DNA Repair 2017, 56, 51–64. [Google Scholar] [CrossRef]
  127. Cui, R.; Widlund, H.R.; Feige, E.; Lin, J.Y.; Wilensky, D.L.; Igras, V.E.; D’Orazio, J.; Fung, C.Y.; Schanbacher, C.F.; Granter, S.R.; et al. Central Role of p53 in the Suntan Response and Pathologic Hyperpigmentation. Cell 2007, 128, 853–864. [Google Scholar] [CrossRef] [PubMed]
  128. Murphy, G.; Young, A.R.; Wulf, H.C.; Kulms, D.; Schwarz, T. The molecular determinants of sunburn cell formation. Exp. Dermatol. 2001, 10, 155–160. [Google Scholar] [CrossRef]
  129. Bashir, M.M.; Sharma, M.R.; Werth, V.P. UVB and proinflammatory cytokines synergistically activate TNF-α production in keratinocytes through enhanced gene transcription. J. Investig. Dermatol. 2009, 129, 994–1001. [Google Scholar] [CrossRef]
  130. Kim, I.Y.; He, Y.Y. Ultraviolet radiation-induced non-melanoma skin cancer: Regulation of DNA damage repair and inflammation. Genes Dis. 2014, 1, 188–198. [Google Scholar] [CrossRef]
  131. Johnson, K.E.; Wulff, B.C.; Oberyszyn, T.M.; Wilgus, T.A. Ultraviolet light exposure stimulates HMGB1 release by keratinocytes. Arch. Dermatol. Res. 2013, 305, 805–815. [Google Scholar] [CrossRef]
  132. Yang, Q.; Guo, N.; Zhou, Y.; Chen, J.; Wei, Q.; Han, M. The role of tumor-associated macrophages (TAMs) in tumor progression and relevant advance in targeted therapy. Acta Pharm. Sin. B 2020, 10, 2156–2170. [Google Scholar] [CrossRef]
  133. Rana, S.; Byrne, S.N.; MacDonald, L.J.; Chan, C.Y.Y.; Halliday, G.M. Ultraviolet B suppresses immunity by inhibiting effector and memory T cells. Am. J. Pathol. 2008, 172, 993–1004. [Google Scholar] [CrossRef] [PubMed]
  134. Aberer, W.; Schuler, G.; Stingl, G.; Hönigsmann, H.; Wolff, K. Ultraviolet light depletes surface markers of Langerhans cells. J. Investig. Dermatol. 1981, 76, 202–210. [Google Scholar] [CrossRef] [PubMed]
  135. Stingl, L.A.; Sauder, D.N.; Iijima, M.; Wolff, K.; Pehamberger, H.; Stingl, G. Mechanism of UV-B-induced impairment of the antigen-presenting capacity of murine epidermal cells. J. Immunol. 1983, 130. [Google Scholar] [CrossRef]
  136. Vink, A.A.; Shreedhar, V.; Roza, L.; Krutmann, J.; Kripke, M.L. Cellular target of UVB-induced DNA damage resulting in local suppression of contact hypersensitivity. J. Photochem. Photobiol. B Biol. 1998, 44, 107–111. [Google Scholar] [CrossRef]
  137. Brenner, M.; Hearing, V.J. The protective role of melanin against UV damage in human skin. Photochem. Photobiol. 2008, 84, 539–549. [Google Scholar] [CrossRef] [PubMed]
  138. Sung, W.W.; Chang, C.H. Nevi, dysplastic nevi, and melanoma: Molecular and immune mechanisms involving the progression. Tzu Chi Med. J. 2022, 34, 1–7. [Google Scholar] [CrossRef]
  139. Fromme, J.C.; Banerjee, A.; Verdine, G.L. DNA Glycosylase Recognition and Catalysis. Curr. Opin. Struct. Biol. 2004, 14, 43–49. [Google Scholar] [CrossRef]
  140. Beard, W.A.; Prasad, R.; Wilson, S.H. Activities and Mechanism of DNA Polymerase β. Methods Enzymol. 2006, 408, 91–107. [Google Scholar]
  141. Paller, C.J.; Tukachinsky, H.; Maertens, A.; Decker, B.; Sampson, J.R.; Cheadle, J.P.; Antonarakis, E.S. Pan-Cancer Interrogation of MUTYH Variants Reveals Biallelic Inactivation and Defective Base Excision Repair Across a Spectrum of Solid Tumors. JCO Precis. Oncol. 2024, 8, e2300251. [Google Scholar] [CrossRef]
  142. Garces, F.; Pearl, L.H.; Oliver, A.W. The Structural Basis for Substrate Recognition by Mammalian Polynucleotide Kinase 3′ Phosphatase. Mol. Cell 2011, 44, 385–396. [Google Scholar] [CrossRef]
  143. Wang, Z. Translesion Synthesis by the UmuC Family of DNA Polymerases. Mutat. Res. DNA Repair 2001, 486, 59–70. [Google Scholar] [CrossRef]
  144. Yoon, J.H.; Prakash, L.; Prakash, S. Error-Free Replicative Bypass of (6–4) Photoproducts by DNA Polymerase ζ in Mouse and Human Cells. Genes Dev. 2010, 24, 123–128. [Google Scholar] [CrossRef]
  145. Taylor, J.S. New Structural and Mechanistic Insight into the A-Rule and the Instructional and Non-Instructional Behavior of DNA Photoproducts and Other Lesions. Mutat. Res. 2002, 510, 55–70. [Google Scholar] [CrossRef]
  146. Tanaka, M.; Takahara, M.; Nukina, K.; Hayashi, A.; Sakai, W.; Sugasawa, K.; Shiomi, Y.; Nishitani, H. Mismatch Repair Proteins Recruited to Ultraviolet Light-Damaged Sites Lead to Degradation of Licensing Factor Cdt1 in the G1 Phase. Cell Cycle 2017, 16, 673–684. [Google Scholar] [CrossRef] [PubMed]
  147. Russo, T.; Piccolo, V.; Moscarella, E.; Tschandl, P.; Kittler, H.; Paoli, J.; Lallas, A.; Braun, R.P.; Thomas, L.; Soyer, H.P.; et al. Indications for Digital Monitoring of Patients with Multiple Nevi: Recommendations from the International Dermoscopy Society. Dermatol. Pract. Concept. 2022, 12, e2022182. [Google Scholar] [CrossRef] [PubMed]
  148. Djavid, A.R.; Stonesifer, C.; Fullerton, B.T.; Wang, S.W.; Tartaro, M.A.; Kwinta, B.D.; Grimes, J.M.; Geskin, L.J.; Saenger, Y.M. Etiologies of Melanoma Development and Prevention Measures: A Review of the Current Evidence. Cancers 2021, 13, 4914. [Google Scholar] [CrossRef]
  149. Zhu, G.A.; Raber, I.; Sakshuwong, S.; Li, S.; Li, A.S.; Tan, C.; Chang, A.L.S. Estimation of Individual Cumulative Ultraviolet Exposure Using a Geographically-AdjustedGeographically Adjusted, Openly-AccessibleOpenly Accessible Tool. BMC Dermatol. 2016, 16, 1. [Google Scholar] [CrossRef] [PubMed]
  150. Valenzuela, F.I.; Hohnadel, M. Dermatoscopic Characteristics of Melanoma Versus Benign Lesions and Nonmelanoma Cancers; StatPearls Publishing LLC: Treasure Island, FL, USA, 2024. [Google Scholar]
  151. Argenziano, G.; Catricalà, C.; Ardigo, M.; Buccini, P.; De Simone, P.; Eibenschutz, L.; Ferrari, A.; Mariani, G.; Silipo, V.; Sperduti, I.; et al. Seven-Point Checklist of Dermoscopy Revisited. Br. J. Dermatol. 2011, 164, 785–790. [Google Scholar] [CrossRef]
  152. Sanna, A.; Harbst, K.; Johansson, I.; Christensen, G.; Lauss, M.; Mitra, S.; Rosengren, F.; Häkkinen, J.; Vallon-Christersson, J.; Olsson, H.; et al. Tumor Genetic Heterogeneity Analysis of Chronic Sun-Damaged Melanoma. Pigment. Cell Melanoma Res. 2020, 33, 480–489. [Google Scholar] [CrossRef]
  153. Bobos, M. Histopathologic Classification and Prognostic Factors of Melanoma: A 2021 Update. Ital. J. Dermatol. Venerol. 2021, 156, 300–321. [Google Scholar] [CrossRef]
  154. Ferrara, G.; Argenziano, G. The WHO 2018 Classification of Cutaneous Melanocytic Neoplasms: Suggestions from Routine Practice. Front. Oncol. 2021, 11, 675296. [Google Scholar] [CrossRef] [PubMed]
  155. Nebhan, C.A.; Johnson, D.B. Predictive Biomarkers of Response to Immune Checkpoint Inhibitors in Melanoma. Expert Rev. Anticancer. Ther. 2020, 20, 137–145. [Google Scholar] [CrossRef] [PubMed]
  156. Koizumi, S.; Inozume, T.; Nakamura, Y. Current Surgical Management for Melanoma. J. Dermatol. 2024, 51, 312–323. [Google Scholar] [CrossRef]
  157. Johnston, L.; Starkey, S.; Mukovozov, I.; Robertson, L.; Petrella, T.; Alhusayen, R. Surveillance After a Previous Cutaneous Melanoma Diagnosis: A Scoping Review of Melanoma Follow-Up Guidelines. J. Cutan. Med. Surg. 2023, 27, 516–525. [Google Scholar] [CrossRef]
  158. Dennis, L.K. Cumulative Sun Exposure and Melanoma in a Population-Based Case–Control Study: Does Sun Sensitivity Matter? Cancers 2022, 14, 1008. [Google Scholar] [CrossRef]
  159. Singh, N.; Dunlop, K.L.A.; Woolley, N.; Wills Vashishtha, T.; Damian, D.L.; Vuong, K.; Cust, A.E.; Smit, A.K. A Review of Skin Cancer Primary Prevention Activities in Primary Care Settings. Public Health Res. Pract. 2024, 34, 34012401. [Google Scholar] [CrossRef]
  160. Johr, R.H. Dermoscopy: Alternative Melanocytic Algorithms—The ABCD Rule of Dermatoscopy, Menzies Scoring Method, and 7-Point Checklist. Clin. Dermatol. 2002, 20, 240–247. [Google Scholar] [CrossRef] [PubMed]
  161. Carrera, C.; Marchetti, M.A.; Dusza, S.W.; Argenziano, G.; Braun, R.P.; Halpern, A.C.; Jaimes, N.; Kittler, H.J.; Malvehy, J.; Menzies, S.W.; et al. Validity and Reliability of Dermoscopic Criteria Used to Differentiate Nevi From Melanoma: A Web-Based International Dermoscopy Society Study. JAMA Dermatol. 2016, 152, 798–806. [Google Scholar] [CrossRef]
  162. Pellacani, G.; Farnetani, F.; Ciardo, S.; Chester, J.; Kaleci, S.; Mazzoni, L.; Bassoli, S.; Casari, A.; Pampena, R.; Mirra, M.; et al. Effect of Reflectance Confocal Microscopy for Suspect Lesions on Diagnostic Accuracy in Melanoma: A Randomized Clinical Trial. JAMA Dermatol. 2022, 158, 754–761. [Google Scholar] [CrossRef]
  163. Konopinski, J.C.; Danialan, R.; Torres-Cabala, C.A.; Tetzlaff, M.T.; Nagarajan, P.; Curry, J.L.; Ivan, D.; Prieto, V.G.; Aung, P.P. Melanoma Coexisting with Solar Elastosis: A Potential Pitfall in the Differential Diagnosis Between Nevus and Melanoma. Hum. Pathol. 2019, 84, 270–274. [Google Scholar] [CrossRef]
  164. Moscarella, E.; Guitera, P.; Scolyer, R.A.; Rocha, L.; Thomas, L.; Ronchi, A.; Scharf, C.; Brancaccio, G.; Argenziano, G. Junctional Nevus and Early Melanoma on Sun-Damaged Skin of the Head/Neck: A Clinico-Pathologic Challenge. Dermatol. Pract. Concept. 2023, 13, e2023122. [Google Scholar] [CrossRef] [PubMed]
  165. Heinlein, L.; Maron, R.C.; Hekler, A.; Haggenmüller, S.; Wies, C.; Utikal, J.S.; Meier, F.; Hobelsberger, S.; Gellrich, F.F.; Sergon, M.; et al. Prospective Multicenter Study Using Artificial Intelligence to Improve Dermoscopic Melanoma Diagnosis in Patient Care. Commun. Med. 2024, 4, 177. [Google Scholar] [CrossRef]
  166. Prkačin, I.; Mokos, M.; Ferara, N.; Šitum, M. Melanoma’s New Frontier: Exploring the Latest Advances in Blood-Based Biomarkers for Melanoma. Cancers 2024, 16, 4219. [Google Scholar] [CrossRef] [PubMed]
  167. Cadet, J.; Douki, T.; Ravanat, J.L. Oxidatively generated damage to cellular DNA by UVB and UVA radiation. Photochem. Photobiol. 2015, 91, 140–155. [Google Scholar] [CrossRef] [PubMed]
  168. Fisher, G.J.; Kang, S.; Varani, J.; Bata-Csorgo, Z.; Wan, Y.; Datta, S.; Voorhees, J.J. Mechanisms of photoaging and chronological skin aging. Arch. Dermatol. 2002, 138, 1462–1470. [Google Scholar] [CrossRef]
  169. Haenssle, H.A.; Fink, C.; Schneiderbauer, R.; Toberer, F.; Buhl, T.; Blum, A.; Kalloo, A.; Ben Hadj Hassen, A.; Thomas, L.; Enk, A.; et al. Man against Machine: Diagnostic performance of a deep learning convolutional neural network for dermoscopic melanoma recognition in comparison to 58 dermatologists. Ann. Oncol. 2018, 29, 1836–1842. [Google Scholar] [CrossRef]
Figure 1. The image illustrates the three main DNA repair mechanisms: (A) NER, (B) BER, and (C) TLS. XPC: Xeroderma Pigmentosum Complementation group C; HR23B: Homologous Recombination 23B; UV-DOB: UV-Damage DNA Binding; XPF-ERCC1: Xeroderma Pigmentosum Complementation group F-Excision Repair Cross-Complementation group 1; XPB: Xeroderma Pigmentosum Complementation group B; XPG: Xeroderma Pigmentosum Complementation group G; DNAPol: DNA Polymerase; PCNA: Proliferating Cell Nuclear Antigen; RNAPII: RNA Polymerase II; CSB: Cockayne Syndrome group B-protein; XPD: Xeroderma Pigmentosum Complementation group D; APE1: Apurinic/Apyrimidinic Endonuclease 1; Pol-β: DNA Polymerase β; XRCC1: X-Ray Repair Cross-Complementing protein 1; FEN1: Flap Endonuclease 1; Pol δ/ε: DNA Polymerases δ and ε; Ub: ubiquitin.
Figure 1. The image illustrates the three main DNA repair mechanisms: (A) NER, (B) BER, and (C) TLS. XPC: Xeroderma Pigmentosum Complementation group C; HR23B: Homologous Recombination 23B; UV-DOB: UV-Damage DNA Binding; XPF-ERCC1: Xeroderma Pigmentosum Complementation group F-Excision Repair Cross-Complementation group 1; XPB: Xeroderma Pigmentosum Complementation group B; XPG: Xeroderma Pigmentosum Complementation group G; DNAPol: DNA Polymerase; PCNA: Proliferating Cell Nuclear Antigen; RNAPII: RNA Polymerase II; CSB: Cockayne Syndrome group B-protein; XPD: Xeroderma Pigmentosum Complementation group D; APE1: Apurinic/Apyrimidinic Endonuclease 1; Pol-β: DNA Polymerase β; XRCC1: X-Ray Repair Cross-Complementing protein 1; FEN1: Flap Endonuclease 1; Pol δ/ε: DNA Polymerases δ and ε; Ub: ubiquitin.
Cancers 17 01784 g001
Table 1. The table describes the main genetic factors that predispose to the development of melanoma.
Table 1. The table describes the main genetic factors that predispose to the development of melanoma.
CategoryGene/PathwayMutation TypeEffectMelanoma RiskReference
PigmentationMC1RVariants (especially red hair color—RHC-associated)Alters pigmentation, UV sensitivity, DNA repairIncreased (doubles risk with CDKN2A mutations)[33,50,51]
ASIPVariantsAlters melanocortin signalingSlightly increased[50]
TYRVariantsAlters melanin synthesisSlightly increased[50]
Tumor SuppressorCDKN2A (p16/INK4a, p14/ARF)Germline mutations, Loss of functionDeregulates cell cycle, impairs senescence, affects oxidative stressHigh (67% lifetime risk with heterozygous loss)[52,53,54,55,56,57,58,59,60,61,62,63,64]
RBGermline mutationsReduces tumor suppressor activityIncreased[65,66]
PTENInactivating mutations, deletions, epigenetic silencingActivates AKT signalingIncreased[67,68]
NF1Loss-of-function mutationsHyperactivates NRAS, MAPK, and PI3K/AKT pathwaysIncreased (especially with sun damage)[69,70]
Signaling Pathway (MAPK)BRAFV600E (most common), V600R, V600DConstitutive activation of kinase activity, activates MEK/ERKHighly increased[71,72,73,74,75,76]
NRASQ61R, Q61K (most common)Activates MAPK pathwayIncreased[77,78]
RAS (KRAS, HRAS, NRAS)MutationsActivates RAF kinasesIncreased[76]
Signaling Pathway (WNT)CTNNB1MutationsStabilizes β-catenin, activates transcriptionIncreased (2–23% of cases)[79,80]
Receptor Tyrosine Kinase (RTK)EGFRGene copy number gains, point mutationsDysregulationIncreased[81]
HGF/METGene copy number gains, point mutationsDysregulationIncreased[82]
KITL576P mutationActivates KIT signalingIncreased (small number of melanomas)[83]
PTPRDDeletionsLoss of phosphatase activityIncreased[84]
PDGFR, IGFRUpregulationIncreased signalingIncreased[85]
Transcription FactorMITFAmplification,
E318K mutation
Drives melanocytic lineage, survival, growth, differentiationIncreased[86,87,88,89]
MYCOverexpressionEnhances melanoma progressionIncreased[90]
TBX2AmplificationRepresses p14ARF and p21CIP1Increased[91,92]
TERTPromoter mutations (C→T)Creates ETS transcription factor binding sitesIncreased proliferation[93,94]
Epigenetic FactorsMultiple genesDifferential methylation (UV exposure signature)May drive melanoma developmentIncreased[95,96]
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

Manganelli, M.; Stabile, G.; Scharf, C.; Podo Brunetti, A.; Paolino, G.; Giuffrida, R.; Bigotto, G.D.; Damiano, G.; Mercuri, S.R.; Sallustio, F.; et al. Skin Photodamage and Melanomagenesis: A Comprehensive Review. Cancers 2025, 17, 1784. https://doi.org/10.3390/cancers17111784

AMA Style

Manganelli M, Stabile G, Scharf C, Podo Brunetti A, Paolino G, Giuffrida R, Bigotto GD, Damiano G, Mercuri SR, Sallustio F, et al. Skin Photodamage and Melanomagenesis: A Comprehensive Review. Cancers. 2025; 17(11):1784. https://doi.org/10.3390/cancers17111784

Chicago/Turabian Style

Manganelli, Michele, Giorgio Stabile, Camila Scharf, Antonio Podo Brunetti, Giovanni Paolino, Roberta Giuffrida, Gianmarco Diego Bigotto, Giuseppe Damiano, Santo Raffaele Mercuri, Fabio Sallustio, and et al. 2025. "Skin Photodamage and Melanomagenesis: A Comprehensive Review" Cancers 17, no. 11: 1784. https://doi.org/10.3390/cancers17111784

APA Style

Manganelli, M., Stabile, G., Scharf, C., Podo Brunetti, A., Paolino, G., Giuffrida, R., Bigotto, G. D., Damiano, G., Mercuri, S. R., Sallustio, F., Mangano, E., Bordoni, R., De Nardi, P., Guida, G., Foti, C., Argenziano, G., Longo, C., Pellacani, G., Rizzo, N., ... Rongioletti, F. (2025). Skin Photodamage and Melanomagenesis: A Comprehensive Review. Cancers, 17(11), 1784. https://doi.org/10.3390/cancers17111784

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