Previous Article in Journal
Mimicking the LOX-Related Autosomal Recessive Congenital Ichthyosis Skin Disease Using a CRISPR-Cas9 System and Unravelling 12S-LOX Function in the Skin
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Molecular and Genetic Markers for Malignant Melanoma: Implications for Prognosis and Therapy

1
School of Medicine, New York Medical College, Valhalla, NY 10595, USA
2
Dermatology Department, NYC Health + Hospital/Metropolitan, New York, NY 10029, USA
3
Dermatology Department, NYC Health + Hospital/South Brooklyn Health, Brooklyn, NY 11235, USA
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Co-senior Authors.
Dermatopathology 2025, 12(3), 31; https://doi.org/10.3390/dermatopathology12030031
Submission received: 28 July 2025 / Revised: 9 September 2025 / Accepted: 11 September 2025 / Published: 12 September 2025

Abstract

Despite therapeutic advancements, malignant melanoma remains a leading cause of skin cancer-related mortality, with incidence continuing to rise globally. Traditional prognostic tools offer important clinical guidance but fail to capture the biological heterogeneity of melanoma or reliably predict responses to emerging therapies. In this review, we summarize recent advances in prognostic and predictive molecular biomarkers reported over the past five years. We discuss immunohistochemical and tissue-based markers, circulating biomarkers, microRNAs, and gene expression profiles that enhance risk stratification and inform surveillance strategies. We also review immune-related markers that may predict response to immune-checkpoint inhibitor therapy. Lastly, we highlight investigational biomarkers—including gene signatures, epigenomic alterations, and microbiome influences—that are shaping the future landscape. Together, these advances reflect a shift toward precision oncology in melanoma, with the integration of biomarker-driven strategies offering the potential to personalize treatment and improve patient outcomes.

1. Introduction

Malignant melanoma remains a leading cause of cancer-related deaths, and its incidence continues to increase worldwide [1]. In 2020, there were an estimated 325,000 new cases of melanoma and 57,000 melanoma-related deaths globally; these numbers are projected to rise to 510,000 cases and 96,000 deaths by 2040 [2]. Although melanoma remains a significant public health concern, mortality rates have gradually declined since the introduction of immune-checkpoint inhibitors (ICIs) [3]. However, because ICIs are effective only in a subset of patients and carry significant toxicity and cost, the identification of biomarkers that predict treatment response is essential to guide therapy and optimize outcomes [4].
Several melanoma staging algorithms are used to determine prognosis and guide treatment decisions, with the American Joint Committee on Cancer (AJCC) tumor/node/metastasis (TNM) system and National Comprehensive Cancer Network (NCCN) guidelines among the most widely adopted [5,6]. Prognostic factors in melanoma include age, ethnicity, primary location, mitotic rate, ulceration, Breslow depth, sentinel lymph node (SLN) status, and the presence of metastasis [5,7]. While these clinicopathological variables provide important information, they do not adequately account for the underlying biological heterogeneity of melanoma or predict response to modern therapies such as immunotherapy and targeted agents [8]. In recent years, molecular and immunologic advances have led to the development and validation of assays with promising potential to improve risk stratification, better predict treatment response, and provide earlier detection of disease recurrence. This narrative review summarizes advances over the past five years in molecular and genetic biomarkers with prognostic relevance in cutaneous malignant melanoma.
A literature search of PubMed and Google Scholar was completed in May 2025. The search was limited to peer-reviewed studies published within the last five years (May 2020 to May 2025). The following search terms were used: “melanoma” AND (biomarkers OR “prognostic biomarker” OR “gene expression” OR immunohistochemistry OR “risk stratification”) AND (survival OR recurrence OR prognosis). The search was supplemented by a manual review of pertinent references. Excluded from the search were case reports, case series, editorials, commentaries, abstracts, poster presentations, and non-English publications.

2. Immunohistochemical and Tissue-Based Prognostic Markers

2.1. Preferentially Expressed Antigen in Melanoma (PRAME; Gene Symbol PRAME)

Preferentially Expressed Antigen in Melanoma (PRAME) is a tumor-associated antigen that was first identified in a patient with metastatic cutaneous melanoma and belongs to the cancer-testis antigen family [9]. It has since emerged as a potential prognostic biomarker in melanoma (Table 1), with several studies reporting that high PRAME expression (typically defined as ≥50% positive tumor cells as PRAME-positive, though individual study cutoffs may vary). Additionally, some studies use H-score systems that incorporate immunohistochemical staining intensity, which is associated with a worse prognosis and reduced overall survival (OS) [9,10,11]. In a study of 123 cases of cutaneous melanoma, Asato et al. reported that both the extent and intensity of PRAME expression were individually associated with decreased OS (p = 0.0267 and p = 0.043, respectively) [10]. PRAME has also been reported to be highly expressed in uveal melanomas and its presence correlated with an increased risk of metastasis, particularly to the liver [11]. In metastatic melanoma, PRAME expression serves as a useful marker to distinguish true lymph node metastases from benign nodal melanocytic deposits [12]. It has also shown value in acral melanoma, where it improves diagnostic accuracy by helping differentiate malignant tumors from benign acral melanocytic proliferations that can closely mimic melanoma [13].

2.2. Ki-67

Ki-67 is a well-established marker of cellular proliferation that has been identified in many cancer types [74]. A 2021 meta-analysis by Liu et al. evaluated the prognostic role of Ki-67 in 929 patients across 10 melanoma studies and found that its overexpression, typically defined as ≥20% proliferation index considered a “high” cutoff (most studies had cutoffs ranging from 10 to 30%, with most studies deriving these thresholds post hoc), was associated with worsened OS (HR = 2.92; 95% CI, 2.17–3.91, p < 0.0001) [14]. However, the authors reported no apparent correlation between Ki-67 overexpression and overall progression-free survival (PFS) (HR = 0.999; 95% CI, 0.958–1.041; p = 0.958; I2 = 21.80%; p = 0.258) or relapse-free survival (RFS) (HR = 1.14; 95% CI, 0.42–3.11; p = 0.993; I2 = 85.00%, p = 0.01) [14]. Similarly, Asato et al. found that Ki-67 expression was significantly associated with worse OS (p = 0.005) and sentinel lymph node (SLN) status (p = 0.0403) [10]. Additionally, Du et al. reported that patients with acral melanoma in the Ki-67 high expression group had significantly shorter median melanoma-specific survival than those in the low Ki-67 expression group (p < 0.001) [15].

2.3. Activating Molecule in Beclin1-Regulated Autophagy (AMBLor)

Activating molecule in Beclin1-regulated autophagy (AMBRA1 gene), in combination with loricrin (termed “AMBLor” together), has been recently validated as a prognostic biomarker of nonulcerated AJCC stage I/II melanomas [16]. AMBLor is a combined epidermal immunohistochemical marker panel that assesses loss of AMBRA1, an autophagy-regulatory protein, with loricrin, a marker of terminal keratinocyte differentiation. In a large multivariate study by Ewen et al., absence of AMBLor expression in nonulcerated AJCC stage I–II melanoma was associated with an increased risk of recurrence (HR = 3.49; p = 0.007). Also, AMBLor positivity was strongly associated with favorable outcomes, with a negative predictive value of 96.5% for recurrence [16]. These findings suggest that AMBLor may serve as a valuable tool for identifying patients with early-stage melanoma who are at low risk of recurrence.

2.4. Y-Box Binding Protein 1 (YB-1)

Y-box binding protein (YB-1) is a multifunctional transcription and translational factor involved in cell proliferation, stress response, and oncogenesis [17]. It serves as a marker of epithelial-to-mesenchymal transition, a process that, despite melanoma’s neural crest origin, has been documented in cutaneous melanoma [17]. Elevated YB-1 expression is significantly upregulated in primary melanomas and is associated with worse survival outcomes. Experimental data suggests that YB-1 promotes melanoma progression and metastasis [17].

2.5. Microphthalmia Transcription Factor (MITF) and Transient Receptor Potential Melastatin-1 (TRPM1)

Microphthalmia transcription factor (MITF) plays a critical role in the development and function of melanocytes [18]. In melanoma, MITF protein functions as a transcription factor involved in proliferation and differentiation. However, MITF’s function in melanoma varies across specimens, where in advanced melanoma MITF is typically downregulated, but has been amplified in select cases [19]. Transient receptor potential melastatin-1 (TRPM1) is a calcium-permeable ion channel originally identified as a tumor suppressor gene and downregulated in metastatic melanoma [20]. It is transcriptionally regulated by MITF, and its decreased expression correlates with tumor aggressiveness [20,21].

Summary

Immunohistochemistry-based biomarkers including PRAME, Ki-67, AMBLor, YB-1, TRPM1, and MITF have been investigated as prognostic markers in cutaneous melanoma. Evidence is strongest for PRAME and Ki-67, with elevated expression linked to poorer OS and higher SLN positivity. AMBLor positivity has been associated with reduced recurrence risk in early-stage melanoma, while data for YB-1, TRPM1, and MITF continue to emerge. Currently, none of these biomarkers are included in NCCN or AJCC guidelines, though PRAME is increasingly adopted as a diagnostic adjunct.

3. Circulating Biomarkers and Liquid Biopsies

3.1. Lactate Dehydrogenase (LDH)

Lactate dehydrogenase (LDH) is a key metabolic enzyme upregulated in many cancers that contributes to tumor growth and metastatic progression. LDH is incorporated into the TNM (tumor/node/metastasis) staging of melanoma, where elevated expression is associated with worse prognosis [22]. In a study of 121 melanoma patients, LDH showed 92.2% specificity and 41.9% sensitivity for identifying regional or distant metastases (stage III–IV disease) [23].
Serum LDH is also an independent prognostic factor for survival and is included in the AJCC classification for stage IV melanoma [22,24]. LDH has also been evaluated as a biomarker for melanoma patients receiving anti-programmed death-1/programmed death-ligand 1 (PD-1/PD-L1) therapy, where elevated baseline LDH levels are associated with reduced OS (HR = 2.44; 95% CI, 1.95–3.04; p < 0.001) and shorter PFS (HR = 1.61; 95% CI, 1.34–1.92; p < 0.001) [25].

3.2. Serum S100 Calcium-Binding Protein B (S100B)

Serum S100 calcium-binding protein B (S100B) plays a key role in cytoskeletal integrity, regulating the cell cycle, and promoting apoptosis. It is one of the most studied biomarkers, released into the bloodstream by melanoma cells upon cellular damage or death [26]. Elevated S100B levels have been linked to worse outcomes in melanoma [27]. Notably, S100B is among the serum protein biomarkers with the most robust clinical evidence supporting its role in melanoma surveillance, particularly for monitoring recurrence during follow-up [26].
S100B levels correlate closely with tumor burden, with higher concentrations observed in advanced compared to early-stage disease [27]. A recent meta-analysis of melanoma follow-up studies found that S100B surpassed LDH in detecting recurrence, demonstrating a higher area under the curve (AUC) and greater sensitivity while maintaining high specificity [27].
Serum S100B has also shown utility as an early indicator of treatment response in melanoma patients receiving immunotherapy. In a study by Wagner et al., baseline S100B levels strongly correlated with overall survival in patients treated with anti–PD-1 agents [28].
Despite strong supporting data, S100B has not been incorporated into the AJCC staging system. LDH remains the only serum biomarker formally recognized. This reflects practical and historical considerations rather than biological inferiority. LDH is globally available, inexpensive, and standardized, whereas S100B has been regionally adopted and lacks widespread assay standardization, limiting its broader implementation [26].

3.3. Melanoma-Inhibiting Activity (MIA) Protein

Melanoma inhibitory activity (MIA) protein plays a role in melanoma progression by promoting metastasis and contributing to tumor-associated immunosuppression. It is considered a melanoma-specific biomarker because it is absent in normal melanocytes and benign melanocytic nevi but strongly expressed in malignant melanoma cells [29]. Elevated serum MIA levels have been associated with advanced disease stage, increased tumor burden, and worse overall prognosis [29].
In recent studies, MIA has also been evaluated as a dynamic marker for treatment monitoring, particularly in the context of BRAF-mutant melanoma [30]. In patients with BRAF V600 mutations undergoing targeted therapy with BRAF inhibitors (iBRAF), MIA levels, alongside S100B and LDH, were shown to significantly decline within the first month of treatment [30]. These reductions correlated with clinical response and PFS, suggesting that early decreases in MIA may serve as indicators of therapeutic efficacy.
Although MIA is less commonly used than S100B or LDH in clinical practice, it provides greater specificity for melanoma and may have added value when combined with other biomarkers. Further studies are needed to validate its role in routine surveillance and to determine its utility in predicting long-term outcomes across different therapeutic settings.

3.4. Liquid Biopsy Biomarkers (Cell Free DNA (cfDNA), Cell Free RNA (cfRNA), Circulating Tumor DNA (ctDNA))

Liquid biopsy approaches such as circulating cell-free DNA (cfDNA), cell-free RNA (cfRNA), and particularly circulating tumor DNA (ctDNA) are being investigated as prognostic and predictive biomarkers in melanoma.
ctDNA represents the fraction of cfDNA derived from tumor cells and is typically more fragmented than cfDNA of non-tumor origin. In practice, ctDNA is commonly measured by droplet digital PCR or next-generation sequencing (NGS), with results reported as mutant allele fraction (normal limit of detection ~0.1–0.5%) or copies/mL of plasma. It has emerged as a clinically relevant blood-based biomarker across multiple solid tumors, including melanoma [31,32]. In a multicenter study, ctDNA was detectable in 43.8% of patients with stage III melanoma, where its presence was significantly associated with recurrence at distant or multiple sites (p < 0.05) [33]. Palacios-Diaz et al. reported that ctDNA detection in plasma was significantly associated with disease progression (p = 0.011), overall mortality (p < 0.001), and melanoma-specific death (p < 0.001) [33]. Aoude et al. similarly demonstrated that patients with stage IV melanoma and low concentrations of ctDNA (<10 ng/mL) had better disease-specific survival and PFS [36]. Low ctDNA levels have demonstrated prognostic value in melanoma, correlating with improved distant metastasis-free survival (DMFS) during treatment and more favorable overall outcomes. Eroglu et al. reported that melanoma patients receiving adjuvant therapy who became ctDNA-positive, or remained persistently positive, had significantly worse DMFS compared with ctDNA-negative patients 3.5 months after surgery (HR = 34.54; p < 0.001) [37]. In sum, detectable ctDNA at baseline, increasing ctDNA levels, or reduced clearance of ctDNA during therapy correlate with worse outcomes. However, pre-analytic factors including time to plasma separation (>2 h) or the use of non-stabilizing blood tubes can diminish ctDNA yield.
Circulating cfDNA comprises extracellular DNA fragments released mainly from apoptotic and necrotic cells, but also through active processes such as neutrophil extracellular trap formation; erythroid precursors may contribute under certain conditions [32,34]. One study by Váraljai et al. analyzed cfDNA concentrations among patients with metastatic melanoma, compared to healthy controls, and reported that higher baseline cfDNA concentrations were significantly associated with higher AJCC stage, presence of metastases and shorter OS [35]. Cell-free RNA (cfRNA), particularly circulating microRNAs (miRNAs), is emerging as a promising non-invasive biomarker in melanoma by enabling pan-tumor monitoring and treatment response surveillance. Distinct plasma miRNA patterns can differentiate melanoma patients from healthy individuals with high sensitivity and specificity [35]. Specific circulating miRNAs have been shown to carry prognostic significance, with elevated miR-221 correlating with thicker primary tumors, metastasis and worse survival outcomes. Additional specific messenger RNAs (mRNAs) such as KPNA2, DTL, BACE2, and DTYMK have been utilized as a panel for liquid biopsy in melanoma detection [38]. Low baseline cfRNA levels were associated with significantly longer PFS (p = 0.0349) and improved OS (p = 0.0393) [38]. Other miRNAs can reflect treatment response or recurrence likelihood. Taken together, cfDNA and cfRNA biomarkers show promise for enabling earlier detection of melanoma, informing prognosis and allowing dynamic, non-invasive monitoring of therapeutic response.

Summary

Among circulating biomarkers, LDH has the strongest evidence base and remains the only serum marker incorporated into AJCC staging for melanoma, supported by consistent findings from large multicenter studies. By comparison, S100B and MIA show moderate-quality evidence and in some studies may outperform LDH for recurrence detection, but their clinical adoption has been limited by lack of assay standardization and regional variability in use. Emerging liquid biopsy approaches, including ctDNA, cfDNA, and cfRNA, provide additional prognostic information, with ctDNA in particular associated with early relapse, shorter PFS, and worse OS. Notably, ctDNA has demonstrated prognostic value independent of AJCC stage.

4. MicroRNA

4.1. miR-200a-3p

miR-200a-3p has demonstrated both diagnostic and prognostic relevance. Prodan et al. reported that elevated circulating levels of miR-200a-3p were associated with reduced OS in patients with advanced-stage melanoma [39]. In addition, higher expression levels of miR-200a-3p were associated with increased risk of mortality (HR = 2.28; p = 0.001). In vulvar melanoma, miR-200a-3p expression was significantly lower than in primary cutaneous melanoma (p = 0.0008) [75].

4.2. miR-335-5p

Elevated circulating levels of miR-335-5p, have been linked to worse OS in melanoma, potentially through regulation of metastasis-associated pathways. In tumor tissue, miR-335 expression is reduced relative to adjacent non-tumor tissue, yet within melanoma samples higher expression correlates with greater invasive depth, lymph node metastasis, and advanced stage, underscoring its prognostic significance [39,76]. Furthermore, patients with high expression levels had significantly shorter median survival times, compared to patients with low expression [39]. Collectively, these findings support miR-335-5p as a marker of poor prognosis in cutaneous melanoma.

4.3. miR-451a

In contrast to the aforementioned microRNAs, miR-451a is considered a favorable prognostic and diagnostic biomarker [39,77]. Higher expression of miR-451a was associated with a lower risk of a higher Breslow index [77]. miR-451a has also been found to correlate with patient responsiveness to anti-PD1 therapies in patients with advanced melanoma [78].

4.4. miR-29b-3p

Elevated levels of miR-29b-3p have also been associated with better survival outcomes and correlate with improved response to ICI therapy (OR = 2.35, 95% CI: 1.15–4.79) [39]. Prodan et al. showed downregulation (fold change = 0.92) of miR-29b-3p in melanoma [77]. Lower expression was also strongly associated with greater tumor thickness, with reduced miR-29b-3p linked to a higher Breslow index (OR = 2.51).

4.5. miR-424

Xu et al. reported that elevated miR-424 expression was associated with more aggressive melanoma subtypes and correlated with reduced disease-free survival (DFS) and OS, indicating its role as a marker of poor prognosis [40]. Furthermore, miR-424 expression was correlated with tumor thickness (p = 0.031), metastasis (p = 0.010), tumor stage (p = 0.005) and ulceration (p < 0.001) [40]. Du et al. supported these findings demonstrating that miR-424-5p was highly expressed in melanoma cells and their exosomes [79].

Summary

Evidence for miRNAs as melanoma biomarkers is emerging but remains limited. Elevated miR-200a-3p, miR-335-5p, and miR-424 have been associated with worse OS, whereas higher levels of miR-451a and miR-29b-3p correlate with improved survival and enhanced response to ICIs. Most findings are based on single-center, retrospective studies that did not control for AJCC staging variables, making it unclear whether these associations are independent of established prognostic factors. At present, miRNAs remain investigational, and larger prospective studies are required to define their role in clinical decision-making.

5. Genomic and Transcriptomic

5.1. Gene Expression Profile (GEP) Analysis

Gene expression profiling (GEP) evaluates multiple mRNAs in melanoma tissue, using methods such as microarray analysis, mRNA sequencing, or reverse transcription PCR (RT-PCR), to predict metastasis and recurrence risk and potentially guide treatment and surveillance decisions [80,81].
11-Gene GEP: This qRT-PCR–based assay generates molecular scores for prognostic stratification [41]. In 245 patients with AJCC stage II cutaneous melanoma, high scores predicted significantly worse outcomes than low scores, including melanoma-specific survival (p = 0.018), DMFS (p = 0.005) and RFS (p = 0.009) [41]. The 11-GEP score remained an independent prognostic factor even when accounting for tumor thickness and age (HR = 1.21, and 1.05, respectively.) [41]. Despite prospective validation, clinical adoption is minimal due to limited cohort sizes and unclear impact on management decisions.
31-Gene GEP: Evaluated across 17 Surveillance, Epidemiology and End Results (SEER) registries (n = 4687), this assay stratifies patients into three risk categories: Class 1A was defined as low risk, class 1B/2A was intermediate risk, and class 2B was considered high risk. Bailey et al. found that class 2B patients experienced significantly worse OS (HR = 2.39), compared to Class 1A [81]. This model offers the largest real-world validation and broader clinical uptake but questions remain regarding its impact on clinical decision-making.
CP-GEP: This hybrid model integrates clinicopathologic features with gene expression to improve risk stratification in stage I/IIA melanoma [42,82]. It identifies high-risk patients for relapse while recognizing those with <5% SLN metastasis risk who may avoid biopsy [42,82,83]. Eggermont et al.’s model demonstrated high negative predictive value (90.5%, 95% CI: 77.9–96.2%) and superior relapse-free survival stratification (HR = 2.98; p < 0.0001) [42]. While this assay shows promise, it is still investigational and has limited clinical adoption.

Summary

GEP assays can provide risk stratification beyond traditional AJCC criteria. The 11-gene model shows prognostic value but has minimal uptake. The 31-gene model offers the largest validation cohort and broader adoption but unclear management impact. The CP-GEP model uniquely integrates molecular and clinical data for SLN decision-making but requires further validation. Key debates remain concerning clinical utility, cost-effectiveness, and which model best guides management decisions.

5.2. Telomerase Activation Through Telomerase Reverse Transcriptase (TERT) Promoter Mutation

Telomerase activation via telomerase reverse transcriptase (TERT) is a fundamental driver of carcinogenesis. In melanoma, TERT promoter mutations are associated with worse clinical outcomes. A meta-analysis of more than 2500 cases demonstrated that TERT promoter-mutations had significantly reduced OS compared with TERT-wild type (HR = 1.43; 95% CI, 1.05–1.95) [43]. The adverse prognostic effect was even stronger in melanoma-specific survival and in subgroup analyses of non-acral cutaneous melanoma [43]. The oncogenic role of TERT is further supported by findings in uveal melanoma, where activating promoter mutations have been identified in primary tumor samples, reinforcing its importance in melanoma pathogenesis through telomerase reactivation and cellular immortality [84].

5.3. Tumor Mutational Burden (TMB)

Tumor mutational burden (TMB) refers to the total number of somatic mutations within a tumor’s genome and has been shown to correlate with response to immunotherapy [85]. In 2022, Ning et al. published a meta-analysis evaluating the association between TMB and ICI outcomes in melanoma. They reported that high TMB was significantly associated with improved OS (HR = 0.49; 95% CI, 0.33–0.73; p = 0.001) and PFS (HR = 0.47; 95% CI, 0.33–0.68; p < 0.001) compared with low TMB [44]. These findings suggest that TMB may serve as a predictive biomarker for response to PD-1/PD-L1 and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) blockade, with higher TMB indicating a greater likelihood of benefit. Real-world data further support this association, showing that high TMB was independently predictive of progression-free survival and OS following first-line ICI therapy, reinforcing its potential role in guiding treatment decisions [45].

5.4. BRAF Mutation

The BRAF mutation, specifically the single point mutation V600E, is one of the most frequently mutated oncogenes in melanoma [46]. Pathogenic gene variants in the BRAF gene occur in about 50% of patients with melanoma [46]. This constitutively activating mutation drives tumor cell proliferation, survival, invasion, and metastasis [46]. Assessment of BRAF mutational status is now a standard component of the diagnostic workup for advanced melanoma [47]. BRAF-mutated melanomas are generally more aggressive in stage IV disease and are associated with reduced survival [86]. Moreover, patients with localized BRAF-mutant melanoma have demonstrated poorer DFS compared with those harboring wild-type tumors (HR = 2.2; 95% CI, 1.1–4.3) independent of AJCC staging [87].

5.5. Neuroblastoma RAS (NRAS) Mutation

NRAS mutations, present in approximately 15–25% of melanomas, define a subset of tumors that are generally more aggressive and associated with poorer outcomes compared to NRAS wild-type disease [48,49]. They are most frequently observed in nodular melanoma [49].
NRAS mutations have been linked to a higher frequency of nodal relapse (p = 0.013) and development of metastatic disease (p = 0.021) [48]. However, NRAS status does not appear to predict treatment efficacy, as studies have shown no significant differences in progression-free or OS among NRAS-mutant versus wild-type patients treated with anti–PD-1–based ICIs [50].

5.6. Cyclin-Dependent Kinase Inhibitor 2A (CDKN2A) Deletions/Mutations

Loss of the tumor suppressor gene, cyclin-dependent kinase inhibitor 2A (CDKN2A), is a frequent driver of melanoma progression. Inactivation may occur through somatic mutations, deletions, epigenetic alterations or inherited variants [51,52]. Patients with germline CDKN2A mutations tend to develop multiple primary melanomas (p < 0.001) and present, on average, 15 years earlier than those with sporadic disease (p < 0.001). OS and RFS do not appear significantly different between germline mutation carriers and non-carriers [53]. However, van Doorn et al. reported that familial melanoma patients with CDKN2A mutations had significantly worse melanoma-specific survival compared with wild-type cases, but this difference observed only in those not undergoing regular dermatologic surveillance [54].

5.7. Tissue Epigenetic Biomarkers (DNA Methylation)

DNA methylation is an important epigenetic mechanism with emerging utility as a biomarker in cutaneous melanoma. A 2024 study identified a three-CpG methylation signature significantly associated with OS in cutaneous melanoma, which was validated in two independent cohorts and remained prognostic across clinical subgroups [55]. Similarly, Huo et al. developed a novel prognostic biomarker based on relative methylation orderings of eight pairs of loci, stratifying patients into high- and low-risk groups [56]. The ‘low-risk’ (hypomethylated) group demonstrated greater CD8+ T-cell infiltration and higher tumor mutational burden (p < 0.05), correlating with improved treatment responses compared to the high-risk (hypermethylated) group [56]. Furthermore, global DNA methylation levels assessed by 5-methylcytosine staining were linked to melanoma aggressiveness in a recent study by Meevassana et al. [57]. Low 5-methylcytosine expression was significantly associated with greater Breslow thickness (comparing levels 2 and 4; p = 0.046), presence of ulceration (p = 0.039), and poorer OS (p = 0.027), suggesting that hypomethylation may be linked to more aggressive disease and worse outcomes [57].

5.8. Long Noncoding RNAs (lncRNA)

A 2024 meta-analysis by Masrour et al. reported that long noncoding RNAs (lncRNAs) may serve as prognostic biomarkers in melanoma, as altered lncRNA expression significantly associated with OS (HR = 2.72; p < 0.0001) [58]. Several lncRNA-based expression signatures have since been developed to improve risk stratification and correlate with clinical outcomes. Guo et al. identified immune- and ferroptosis-related lncRNA panels that independently predicted survival and reflected tumor immune and metastatic characteristics, suggesting that patients with higher expression of their five-gene lncRNA signature might benefit from ICI therapy [59]. Similarly, Zhou et al. (2021) developed a 15-lncRNA signature that predicted prolonged survival in advanced melanoma patients treated with anti–PD-1 immunotherapy [60]. However, because few lncRNAs appear consistently across studies, further validation is needed to establish the prognostic significance of individual lncRNAs.

Summary

Gene expression profile (GEP) assays provide additional prognostic information beyond Breslow thickness, ulceration and SLN status, particularly in patients with stage I–II melanoma where recurrence risk is often underestimated. The 11-gene model has demonstrated prognostic value but has seen limited uptake in clinical practice. By contrast, the CP-GEP model integrates gene expression with age and Breslow thickness to refine SLN biopsy decision-making, though broader validation is still required before incorporation into guidelines. Mutational biomarkers also offer important insights; TERT promoter mutations and CDKN2A alterations have been associated with aggressive tumor features and shorter recurrence-free duration, but standardized clinical integration remains lacking. Tumor mutational burden (TMB) has emerged as an investigational biomarker predicting immune checkpoint inhibitor (ICI) therapy responsiveness, with higher TMB linked to improved PFS and OS. Of the genomic/transcriptomic biomarkers discussed, only BRAF V600E/K mutations are routinely tested in practice, as they directly guide eligibility for BRAF ± MEK inhibitors independent of AJCC staging. NRAS mutations correlate with higher recurrence risk and poorer OS, though no targeted therapies are currently approved. Importantly, TERT and CDKN2A mutations have not consistently demonstrated independence from established AJCC prognostic factors.
Beyond genomic mutations and gene expression profiles, epigenetic biomarkers and long noncoding RNAs represent an emerging area of interest. Aberrant DNA methylation patterns at promoter regions and other epigenetic modifications have been associated with tumor progression, immune evasion, and adverse survival outcomes in melanoma. Similarly, dysregulation of lncRNAs has been linked to tumor invasiveness, metastatic potential, and resistance to systemic therapy. While these findings are promising, studies remain limited to small cohorts, and external validation is required before these biomarkers can influence clinical management.

6. Immune-Related Markers

6.1. Programmed Death-Ligand 1 (PD-L1)

Programmed death-ligand 1 (PD-L1) expression has been associated with improved outcomes in melanoma patients treated with anti-PDL-1 therapy such as pembrolizumab [88]. In advanced melanoma, higher response rates have been observed in patients with PD-L1–positive tumors, although thresholds for positivity (commonly ≥1% or ≥5% PD-L1–positive cells) vary across studies and assays, limiting comparability. PD-L1 expression, particularly when co-occurring with BRAF V600 mutations, has also been linked to longer overall survival following ICI therapy [61]. In a real-world analysis, Ellebaek et al. demonstrated that PD-L1 expression (threshold defined as >1%, assay clone and platform not specified) was an independent positive prognostic biomarker for melanoma-specific survival in patients receiving ICI therapy (HR = 0.66; 95% CI 0.52–0.83, p < 0.001) [62].

6.2. Lymphocyte Activation Gene-3 (LAG-3)

Lymphocyte activation gene-3 (LAG-3) is an inhibitory receptor expressed on T cells that has emerged as a potential predictive marker of responsiveness to ICI therapy, particularly PD-1 blockade. In one study of 11 melanoma patients who had completed anti–PD-1–based treatment, LAG-3 expression was detected in 81% of cases [64]. Responders to ICIs demonstrated significantly higher proportions of LAG-3+ cells compared with non-responders (p = 0.0210), and patients with ≥1% LAG-3+ tumor cells had significantly longer PFS than those with <1% LAG-3 (p = 0.0037) [64]. Similarly, Armura et al. identified LAG-3 as a prognostic biomarker, reporting that elevated LAG-3 expression correlated with poor prognosis in ocular melanoma (p < 0.0001) [65]. These apparently inconsistent findings likely reflect differences in melanoma subtype, treatment context, scoring methodology, and clinical endpoints. In cutaneous melanoma treated with anti–PD-1, LAG-3 positivity aligns with ICI responsiveness and longer PFS, whereas in ocular melanoma, high LAG-3 expression is instead associated with adverse outcomes.

6.3. T Cell Immunoglobulin and Mucin Domain-Containing Protein 3 (TIM-3)

T cell immunoglobulin and mucin domain-containing protein 3 (TIM-3) is an immune-checkpoint protein that has recently been investigated as a potential biomarker in melanoma. Machiraju et al. analyzed tumor samples from 90 patients with advanced cutaneous melanoma and found that increased filtration with TIM-3+ T cells in pretreatment metastases was associated with significantly shorter PFS under anti–PD-1 therapy (p = 0.019) [66]. In contrast, Wiecken et al. reported that high TIM-3 expression correlated with significantly longer overall OS in the anti-PD-1 treatment group (p = 0.007) [67]. TIM-3 has also been studied as a circulating biomarker: Navani et al. observed that TIM-3 expression on CD4+ T cells was significantly higher in non-responders compared to responders (27.8% vs. 13.4%, p = 0.01), suggesting it may serve as a marker of treatment failure [68]. As with LAG-3, these inconsistent findings likely reflect differences in where TIM-3 expression is measured (tumor tissue vs. circulating T cells), the treatment context, and the clinical endpoints analyzed. Overall, high TIM-3+ infiltration in pretreatment metastases has been linked to shorter PFS with anti–PD-1, whereas other cohorts report an association with longer OS.

6.4. Cytotoxic T-Lymphocyte Antigen-4 (CTLA-4)

Cytotoxic lymphocyte antigen-4 (CTLA-4) is an immune-checkpoint receptor that regulates T-cell activity and has been evaluated in the context of cutaneous melanoma. In a retrospective study of 432, germline variants of CTLA-4, such as the c.-1577 AA genotype, were identified as independent predictors of worsened event-free survival at 60 months (52.2% vs. 71.1%, p = 0.02 [69]. With the advent of anti–CTLA-4 therapies such as ipilimumab, attention has turned to CTLA-4 expression as a potential predictor of treatment response. Mastracci et al. and Pistillo et al. reported that high CTLA-4 expression on tumor-infiltrating lymphocytes and tumor cells was associated with improved clinical response to ipilimumab [70,71].

6.5. Melanocortin-1 Receptor (MC1R) Variants

Germline variants in the melanocortin-1 receptor (MC1R) gene, which affect pigmentation and UV susceptibility, have been studied in melanoma. In a large cohort, MC1R status was not associated with OS in general; however, male patients carrying an MC1R variant showed a significant survival disadvantage compared with those with wild-type MC1R [72]. Beyond germline associations, Su et al. reported a stepwise increase in MC1R protein expression across melanoma progression, with higher levels in deeper primary lesions (p < 0.0001) and in ulcerated tumors (p = 0.0008) [73]. Elevated MC1R protein expression correlated with worse OS in both primary (p = 0.0031) and metastatic melanoma (p = 0.0034) [73]. MC1R variants may also impact treatment outcomes: metastatic melanoma patients with concurrent BRAF V600 mutations and MC1R variants experienced lower response rates and shorter PFS on BRAF inhibitor therapy [89].

Summary

Immune-checkpoint biomarkers in melanoma show variable levels of evidence and prognostic relevance. Among them, PD-L1 has the most consistent data, with tumor proportion scores ≥1% predicting improved response to anti–PD-1 and anti–PD-L1 therapies across multiple large studies. PD-L1 retains predictive value independent of AJCC stage, and its immunohistochemistry is routinely used in clinical practice to guide treatment selection. By contrast, findings for LAG-3 and TIM-3 remain inconsistent, reflecting differences in tumor subtype, treatment context, and study design. Elevated CTLA-4 expression has been linked to enhanced immune responses, though survival correlations are mixed. MC1R germline variants increase melanoma susceptibility, but their prognostic utility in the context of ICI therapy is limited. Overall, PD-L1 is the only immune-checkpoint biomarker with established clinical value, while others remain investigational and require further validation.

7. Therapy Associated and Investigational Biomarkers

In addition to prognostic markers, several biomarkers in melanoma are tied to therapy selection, particularly in the context of targeted and immunotherapies. The most clinically established is the BRAF V600E/K mutation which serves as a prerequisite for BRAF and MEK inhibitor therapy. BRAF inhibitors are small molecule drugs that inhibit mutant BRAF kinase in the MAPK pathway, causing tumor regression in melanoma [90]. BRAF inhibitors may be given in conjunction with MEK inhibitors which improve and prolong responses of the therapy [91]. As summarized in Table 2, this approach exemplifies the utility of molecular biomarkers in guiding FDA-approved therapy decisions.
ICIs such as anti-PD-1 antibodies are broadly effective across melanoma subtypes regardless of BRAF mutational status and have substantially improved survival [90,92,93]. Additionally, anti-CTLA-4 antibody was the first approved immune-checkpoint inhibitor in melanoma and may be used alone or in combination with anti-PD-1 antibodies [90,92,95]. In select subtypes such as acral and mucosal melanomas, KIT mutations can guide off-label use of KIT inhibitors [90,95,98]. In addition, while not required for treatment selection, PD-L1 expression and tumor mutational burden are under investigation as predictors of response to immunotherapy responsiveness [90]. Oncolytic virus therapy (T-VEC) also provides benefits for patients with injectable, unresectable lesions [90,92]. Together, these examples reflect the evolving landscape of biomarker-driven therapy in melanoma (Table 2).
Beyond these validated markers, several investigational biomarkers are under active clinical or translational study for their potential to improve prognostic stratification and predict treatment outcomes. These include immune-checkpoint molecules such as LAG-3, which may inform responsiveness to dual checkpoint blockade; ctDNA, which can indicate minimal residual disease or early relapse; and TILs, which correlate with ICI responsiveness [63,99,100,101,102]. Additional emerging areas include gut microbiota composition, which may modulate ICI efficacy; tumor gene expression signatures (e.g., IFN-γ or MYC/E2F pathways); and epigenomic or proteomic markers that reflect immune resistance or tumor aggression [63,100,103,104,105,106,107]. Table 3 highlights several of these investigational biomarkers, including immune-checkpoint molecules like LAG-3, ctDNA, and tumor gene expression profiles that may inform immunotherapy response. Although promising, none are yet validated for routine clinical use, and further prospective studies are required to establish their clinical utility.

8. Limitations and Future Directions

This review is limited by several methodological challenges. Many included studies were retrospective and single-center, creating heterogeneity in assay platforms and cut-off values that compromises cross-study comparability. Furthermore, endpoint definitions varied across studies, and many analyses failed to adequately adjust for established AJCC prognostic factors such as Breslow depth, ulceration, and sentinel lymph node status, potentially leading to overestimation or underestimation of biomarker effects. Publication bias favoring positive findings represents an additional concern.
Future research should prioritize prospective, multi-center studies employing standardized assay platforms and pre-specified cut-off values. Decision impact trials are critically needed to determine whether incorporating these biomarkers into existing AJCC staging frameworks would meaningfully alter clinical management and improve patient outcomes. Additionally, evaluation of external validity must address practical considerations, including cost-effectiveness, accessibility, and health equity, to ensure that promising biomarkers can be successfully translated into routine clinical practice across diverse healthcare settings.

9. Conclusions

In the last five years, there have been significant advances in molecular and clinical assays that have expanded the repertoire of prognostic biomarkers for malignant melanoma. Emerging literature surrounding tissue-based markers appears promising for better risk stratification in early-stage disease, while microRNAs and gene expression sequences may aid in prognostic assessment. ctDNA may be beneficial in determining relapse risk, though further studies are needed. Finally, immune-related markers may help predict tumor response to ICI therapy, which can dramatically improve OS in patients with advanced melanoma. These recent developments underscore the importance of biomarker-driven approaches to diagnosis and prognosis, aimed at enhancing personalized care and promoting better outcomes for patients with malignant melanoma.

Author Contributions

Conceptualization, B.F., B.S. and S.M.; methodology, S.M. and L.F.; software, L.F. and A.S.; validation, S.M., L.F. and A.S.; formal analysis, L.F., R.H., M.F.A., A.S. and S.M.; investigation, L.F., R.H., M.F.A., A.S. and S.M.; resources, L.F., R.H., M.F.A., A.S. and S.M.; data curation, L.F., R.H., M.F.A., A.S. and S.M.; writing—original draft preparation, L.F., R.H., M.F.A. and A.S.; writing—review and editing, L.F., A.S. and S.M.; visualization, R.H.; supervision, B.F., B.S. and S.M.; project administration, B.F., B.S. and S.M.; funding acquisition, N/A. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not Applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study.

Conflicts of Interest

The authors have no conflict of interest to declare.

References

  1. Abbas, O.; Miller, D.D.; Bhawan, J. Cutaneous Malignant Melanoma: Update on Diagnostic and Prognostic Biomarkers. Am. J. Dermatopathol. 2014, 36, 363. [Google Scholar] [CrossRef] [PubMed]
  2. Arnold, M.; Singh, D.; Laversanne, M.; Vignat, J.; Vaccarella, S.; Meheus, F.; Cust, A.E.; de Vries, E.; Whiteman, D.C.; Bray, F. Global Burden of Cutaneous Melanoma in 2020 and Projections to 2040. JAMA Dermatol. 2022, 158, 495–503. [Google Scholar] [CrossRef]
  3. Kahlon, N.; Doddi, S.; Yousif, R.; Najib, S.; Sheikh, T.; Abuhelwa, Z.; Burmeister, C.; Hamouda, D.M. Melanoma Treatments and Mortality Rate Trends in the US, 1975 to 2019. JAMA Netw. Open 2022, 5, e2245269. [Google Scholar] [CrossRef] [PubMed]
  4. Pilard, C.; Ancion, M.; Delvenne, P.; Jerusalem, G.; Hubert, P.; Herfs, M. Cancer immunotherapy: It’s time to better predict patients’ response. Br. J. Cancer 2021, 125, 927–938. [Google Scholar] [CrossRef] [PubMed]
  5. Keung, E.Z.; Gershenwald, J.E. The eighth edition American Joint Committee on Cancer (AJCC) melanoma staging system: Implications for melanoma treatment and care. Expert Rev. Anticancer Ther. 2018, 18, 775–784. [Google Scholar] [CrossRef]
  6. Swetter, S.M.; Johnson, D.; Albertini, M.R.; Barker, C.A.; Bateni, S.; Baumgartner, J.; Bhatia, S.; Bichakjian, C.; Boland, G.; Chandra, S.; et al. NCCN Guidelines® Insights: Melanoma: Cutaneous, Version 2.2024. J. Natl. Compr. Cancer Netw. JNCCN 2024, 22, 290–298. [Google Scholar] [CrossRef]
  7. Rozeman, E.A.; Dekker, T.J.A.; Haanen, J.B.A.G.; Blank, C.U. Advanced Melanoma: Current Treatment Options, Biomarkers, and Future Perspectives. Am. J. Clin. Dermatol. 2018, 19, 303–317. [Google Scholar] [CrossRef]
  8. Safai, B.; Wu, A.G.; Hamby, C.V. Prognostic Biomarkers in Melanoma: Tailoring Treatments to the Patient. J. Clin. Aesthetic Dermatol. 2021, 14, 44–48. [Google Scholar]
  9. Lezcano, C.; Jungbluth, A.A.; Nehal, K.S.; Hollmann, T.J.; Busam, K.J. PRAME Expression in Melanocytic Tumors. Am. J. Surg. Pathol. 2018, 42, 1456–1465. [Google Scholar] [CrossRef]
  10. Asato, M.A.; Neto, F.A.M.M.; Moraes, M.P.d.T.; Ocanha-Xavier, J.P.; Takita, L.C.; Fung, M.A.; Marques, M.E.A.; Xavier-Júnior, J.C.C. The Utility of PRAME and Ki-67 as Prognostic Markers for Cutaneous Melanoma. Am. J. Dermatopathol. 2025, 47, 9–16. [Google Scholar] [CrossRef]
  11. Broggi, G.; Failla, M.; Russo, A.; Longo, A.; Palicelli, A.; Zanelli, M.; Lombardo, C.; Loreto, C.; Merolla, F.; Di Crescenzo, R.M.; et al. Immunohistochemical expression of PRAME is a marker of poor prognosis in uveal melanoma: A clinico-pathologic and immunohistochemical study on a series of 85 cases. Pathol. Res. Pract. 2023, 247, 154543. [Google Scholar] [CrossRef]
  12. Lezcano, C.; Pulitzer, M.; Moy, A.P.; Hollmann, T.J.; Jungbluth, A.A.; Busam, K.J. Immunohistochemistry for PRAME in the distinction of nodal nevi from metastatic melanoma. Am. J. Surg. Pathol. 2020, 44, 503–508. [Google Scholar] [CrossRef]
  13. Cassalia, F.; Danese, A.; Tudurachi, I.; Federico, S.; Zambello, A.; Guidotti, A.; Franceschin, L.; Bolzon, A.; Naldi, L.; Fortina, A.B. PRAME Updated: Diagnostic, Prognostic, and Therapeutic Role in Skin Cancer. Int. J. Mol. Sci. 2024, 25, 1582. [Google Scholar] [CrossRef]
  14. Liu, Q.; Peng, Z.; Shen, L.; Shen, L. Prognostic and Clinicopathological Value of Ki-67 in Melanoma: A Meta-Analysis. Front. Oncol. 2021, 11, 737760. [Google Scholar] [CrossRef]
  15. Du, Y.; Li, C.; Mao, L.; Wei, X.; Bai, X.; Chi, Z.; Cui, C.; Sheng, X.; Lian, B.; Tang, B.; et al. A nomogram incorporating Ki67 to predict survival of acral melanoma. J. Cancer Res. Clin. Oncol. 2023, 149, 13077–13085. [Google Scholar] [CrossRef]
  16. Ewen, T.; Husain, A.; Stefanos, N.; Barrett, P.; Jones, C.; Ness, T.; Long, A.; Horswell, S.; Bosomworth, H.; Lowenstein, J.; et al. Validation of epidermal AMBRA1 and loricrin (AMBLor) as a prognostic biomarker for nonulcerated American Joint Committee on Cancer stage I/II cutaneous melanoma. Br. J. Dermatol. 2024, 190, 549–558. [Google Scholar] [CrossRef] [PubMed]
  17. Kosnopfel, C.; Sinnberg, T.; Sauer, B.; Busch, C.; Niessner, H.; Schmitt, A.; Forchhammer, S.; Grimmel, C.; Mertens, P.R.; Hailfinger, S.; et al. YB-1 Expression and Phosphorylation Regulate Tumorigenicity and Invasiveness in Melanoma by Influencing EMT. Mol. Cancer Res. MCR 2018, 16, 1149–1160. [Google Scholar] [CrossRef] [PubMed]
  18. Levy, C.; Khaled, M.; Fisher, D.E. MITF: Master regulator of melanocyte development and melanoma oncogene. Trends Mol. Med. 2006, 12, 406–414. [Google Scholar] [CrossRef] [PubMed]
  19. Hartman, M.L.; Czyz, M. Pro-Survival Role of MITF in Melanoma. J. Investig. Dermatol. 2015, 135, 352–358. [Google Scholar] [CrossRef]
  20. Hammock, L.; Cohen, C.; Carlson, G.; Murray, D.; Ross, J.S.; Sheehan, C.; Nazir, T.M.; Carlson, J.A. Chromogenic in situ hybridization analysis of melastatin mRNA expression in melanomas from American Joint Committee on Cancer stage I and II patients with recurrent melanoma. J. Cutan. Pathol. 2006, 33, 599–607. [Google Scholar] [CrossRef]
  21. Carlson, J.A.; Ross, J.S.; Slominski, A.; Linette, G.; Mysliborski, J.; Hill, J.; Mihm, M. Molecular diagnostics in melanoma. J. Am. Acad. Dermatol. 2005, 52, 743–775. [Google Scholar] [CrossRef] [PubMed]
  22. Gershenwald, J.E.; Scolyer, R.A.; Hess, K.R.; Sondak, V.K.; Long, G.V.; Ross, M.I.; Lazar, A.J.; Faries, M.B.; Kirkwood, J.M.; McArthur, G.A.; et al. Melanoma Staging: Evidence-Based Changes in the American Joint Committee on Cancer Eighth Edition Cancer Staging Manual. CA Cancer J. Clin. 2017, 67, 472–492. [Google Scholar] [CrossRef]
  23. Henry, L.; Fabre, C.; Guiraud, I.; Bastide, S.; Fabbro-Peray, P.; Martinez, J.; Lavabre-Bertrand, T.; Meunier, L.; Stoebner, P. Clinical use of p-proteasome in discriminating metastatic melanoma patients: Comparative study with LDH, MIA and S100B protein. Int. J. Cancer 2013, 133, 142–148. [Google Scholar] [CrossRef]
  24. Diem, S.; Kasenda, B.; Spain, L.; Martin-Liberal, J.; Marconcini, R.; Gore, M.; Larkin, J. Serum lactate dehydrogenase as an early marker for outcome in patients treated with anti-PD-1 therapy in metastatic melanoma. Br. J. Cancer 2016, 114, 256–261. [Google Scholar] [CrossRef]
  25. Xu, J.; Zhao, J.; Wang, J.; Sun, C.; Zhu, X. Prognostic value of lactate dehydrogenase for melanoma patients receiving anti-PD-1/PD-L1 therapy. Medicine 2021, 100, e25318. [Google Scholar] [CrossRef]
  26. 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]
  27. Janka, E.A.; Várvölgyi, T.; Sipos, Z.; Soós, A.; Hegyi, P.; Kiss, S.; Dembrovszky, F.; Csupor, D.; Kéringer, P.; Pécsi, D.; et al. Predictive Performance of Serum S100B Versus LDH in Melanoma Patients: A Systematic Review and Meta-Analysis. Front. Oncol. 2021, 11, 772165. [Google Scholar] [CrossRef]
  28. Wagner, N.B.; Forschner, A.; Leiter, U.; Garbe, C.; Eigentler, T.K. S100B and LDH as early prognostic markers for response and overall survival in melanoma patients treated with anti-PD-1 or combined anti-PD-1 plus anti-CTLA-4 antibodies. Br. J. Cancer 2018, 119, 339–346. [Google Scholar] [CrossRef] [PubMed]
  29. Revythis, A.; Shah, S.; Kutka, M.; Moschetta, M.; Ozturk, M.A.; Pappas-Gogos, G.; Ioannidou, E.; Sheriff, M.; Rassy, E.; Boussios, S. Unraveling the Wide Spectrum of Melanoma Biomarkers. Diagnostics 2021, 11, 1341. [Google Scholar] [CrossRef]
  30. Sanmamed, M.F.; Fernández-Landázuri, S.; Rodríguez, C.; Lozano, M.D.; Echeveste, J.I.; Gracia, J.L.P.; Alegre, E.; Carranza, O.; Zubiri, L.; Martín-Algarra, S.; et al. Relevance of MIA and S100 serum tumor markers to monitor BRAF inhibitor therapy in metastatic melanoma patients. Clin. Chim. Acta Int. J. Clin. Chem. 2014, 429, 168–174. [Google Scholar] [CrossRef]
  31. Marchisio, S.; Ricci, A.A.; Roccuzzo, G.; Bongiovanni, E.; Ortolan, E.; Bertero, L.; Berrino, E.; Pala, V.; Ponti, R.; Fava, P.; et al. Monitoring circulating tumor DNA liquid biopsy in stage III BRAF-mutant melanoma patients undergoing adjuvant treatment. J. Transl. Med. 2024, 22, 1074. [Google Scholar] [CrossRef]
  32. Dao, J.; Conway, P.J.; Subramani, B.; Meyyappan, D.; Russell, S.; Mahadevan, D. Using cfDNA and ctDNA as Oncologic Markers: A Path to Clinical Validation. Int. J. Mol. Sci. 2023, 24, 13219. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  33. Palacios-Diaz, R.D.; de Unamuno-Bustos, B.; Carratalá-García, A.; Pérez-Simó, G.; Moreno-Ramírez, D.; Ferrándiz, L.; Almazán-Fernández, F.M.; Boada, A.; Loidi-Pascual, L.; Palanca-Suela, S.; et al. Circulating tumor DNA predicts tumor progression and poor survival in patients with stage III melanoma. Melanoma Res. 2025, 35, 259–267. [Google Scholar] [CrossRef] [PubMed]
  34. Knuever, J.; Weiss, J.; Persa, O.-D.; Kreuzer, K.; Mauch, C.; Hallek, M.; Schlaak, M. The use of circulating cell-free tumor DNA in routine diagnostics of metastatic melanoma patients. Sci. Rep. 2020, 10, 4940. [Google Scholar] [CrossRef]
  35. Váraljai, R.; Elouali, S.; Lueong, S.; Wistuba-Hamprecht, K.; Seremet, T.; Siveke, J.; Becker, J.; Sucker, A.; Paschen, A.; Horn, P.; et al. The predictive and prognostic significance of cell-free DNA concentration in melanoma. J. Eur. Acad. Dermatol. Venereol. JEADV 2021, 35, 387–395. [Google Scholar] [CrossRef] [PubMed]
  36. Aoude, L.G.; Brosda, S.; Ng, J.; Lonie, J.M.; Belle, C.J.; Patel, K.; Koufariotis, L.T.; Wood, S.; Atkinson, V.; Smithers, B.M.; et al. Circulating Tumor DNA: A Promising Biomarker for Predicting Recurrence in Patients with BRAF-Negative Melanoma. J. Mol. Diagn. 2023, 25, 771–781. [Google Scholar] [CrossRef]
  37. Eroglu, Z.; Krinshpun, S.; Kalashnikova, E.; Sudhaman, S.; Topcu, T.O.; Nichols, M.; Martin, J.; Bui, K.M.; Palsuledesai, C.C.; Malhotra, M.; et al. Circulating tumor DNA-based molecular residual disease detection for treatment monitoring in advanced melanoma patients. Cancer 2023, 129, 1723–1734. [Google Scholar] [CrossRef] [PubMed]
  38. Albrecht, L.J.; Höwner, A.; Griewank, K.; Lueong, S.S.; von Neuhoff, N.; Horn, P.A.; Sucker, A.; Paschen, A.; Livingstone, E.; Ugurel, S.; et al. Circulating cell-free messenger RNA enables non-invasive pan-tumour monitoring of melanoma therapy independent of the mutational genotype. Clin. Transl. Med. 2022, 12, e1090. [Google Scholar] [CrossRef]
  39. Prodan, M.; Dema, A.; Nataras, B.R.; Seclaman, E.; Bloanca, V.; Crainiceanu, Z.; Deak, I.E.; Virzob, C.R.B.; Toma, A.-O.; Fericean, R.M. Prognostic Significance of miRNA Subtypes in Melanoma: A Survival Analysis and Correlation with Treatment Response Across Patient Stages. Biomedicines 2024, 12, 2809. [Google Scholar] [CrossRef]
  40. Xu, S.-J.; Xu, W.-J.; Zeng, Z.; Zhang, M.; Zhang, D.-Y. MiR-424 Functions as Potential Diagnostic and Prognostic Biomarker in Melanoma. Clin. Lab. 2020, 66, 1207–1213. [Google Scholar] [CrossRef]
  41. Amaral, T.M.; Hoffmann, M.-C.; Sinnberg, T.; Niessner, H.; Sülberg, H.; Eigentler, T.K.; Garbe, C. Clinical validation of a prognostic 11-gene expression profiling score in prospectively collected FFPE tissue of patients with AJCC v8 stage II cutaneous melanoma. Eur. J. Cancer Oxf. Engl. 1990 2020, 125, 38–45. [Google Scholar] [CrossRef]
  42. Eggermont, A.M.; Bellomo, D.; Arias-Mejias, S.M.; Quattrocchi, E.; Sominidi-Damodaran, S.; Bridges, A.G.; Lehman, J.S.; Hieken, T.J.; Jakub, J.W.; Murphree, D.H.; et al. Identification of stage I/IIA melanoma patients at high risk for disease relapse using a clinicopathologic and gene expression model. Eur. J. Cancer Oxf. Engl. 1990 2020, 140, 11–18. [Google Scholar] [CrossRef]
  43. Gandini, S.; Zanna, I.; De Angelis, S.; Palli, D.; Raimondi, S.; Ribero, S.; Masala, G.; Suppa, M.; Bellerba, F.; Corso, F.; et al. TERT promoter mutations and melanoma survival: A comprehensive literature review and meta-analysis. Crit. Rev. Oncol. Hematol. 2021, 160, 103288. [Google Scholar] [CrossRef]
  44. Ning, B.; Liu, Y.; Wang, M.; Li, Y.; Xu, T.; Wei, Y. The Predictive Value of Tumor Mutation Burden on Clinical Efficacy of Immune Checkpoint Inhibitors in Melanoma: A Systematic Review and Meta-Analysis. Front. Pharmacol. 2022, 13, 748674. [Google Scholar] [CrossRef]
  45. Andrews, M.C.; Li, G.; Graf, R.P.; Fisher, V.A.; Mitchell, J.; Aboosaiedi, A.; O’ROurke, H.; Shackleton, M.; Iddawela, M.; Oxnard, G.R.; et al. Predictive Impact of Tumor Mutational Burden on Real-World Outcomes of First-Line Immune Checkpoint Inhibition in Metastatic Melanoma. JCO Precis. Oncol. 2024, 8, e2300640. [Google Scholar] [CrossRef] [PubMed]
  46. 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]
  47. 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] [PubMed]
  48. Heppt, M.V.; Siepmann, T.; Engel, J.; Schubert-Fritschle, G.; Eckel, R.; Mirlach, L.; Kirchner, T.; Jung, A.; Gesierich, A.; Ruzicka, T.; et al. Prognostic significance of BRAF and NRAS mutations in melanoma: A German study from routine care. BMC Cancer 2017, 17, 536. [Google Scholar] [CrossRef]
  49. 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]
  50. Zaremba, A.; Mohr, P.; Gutzmer, R.; Meier, F.; Pföhler, C.; Weichenthal, M.; Terheyden, P.; Forschner, A.; Leiter, U.; Ulrich, J.; et al. Immune checkpoint inhibition in patients with NRAS mutated and NRAS wild type melanoma: A multicenter Dermatologic Cooperative Oncology Group study on 637 patients from the prospective skin cancer registry ADOREG. Eur. J. Cancer 2023, 188, 140–151. [Google Scholar] [CrossRef]
  51. Danishevich, A.; Bilyalov, A.; Nikolaev, S.; Khalikov, N.; Isaeva, D.; Levina, Y.; Makarova, M.; Nemtsova, M.; Chernevskiy, D.; Sagaydak, O.; et al. CDKN2A Gene Mutations: Implications for Hereditary Cancer Syndromes. Biomedicines 2023, 11, 3343. [Google Scholar] [CrossRef]
  52. Smith-Sørensen, B.; Hovig, E. CDKN2A (p16INK4A) somatic and germline mutations. Hum. Mutat. 1996, 7, 294–303. [Google Scholar] [CrossRef]
  53. Ipenburg, N.A.; El Sharouni, M.A.; van Doorn, R.; van Diest, P.J.; van Leerdam, M.E.; van der Rhee, J.I.; Goeman, J.; Kukutsch, N.A. Lack of association between CDKN2A germline mutations and survival in patients with melanoma: A retrospective cohort study. J. Am. Acad. Dermatol. 2022, 87, 479–482. [Google Scholar] [CrossRef] [PubMed]
  54. van Doorn, R. Surveillance, CDKN2A and survival of familial melanoma. J. Eur. Acad. Dermatol. Venereol. 2023, 37, 218–219. [Google Scholar] [CrossRef]
  55. Lin, Z.; Yang, L. Identification of a CpG-based signature coupled with gene expression as prognostic indicators for melanoma: A preliminary study. Sci. Rep. 2024, 14, 5302. [Google Scholar] [CrossRef]
  56. Huo, Y.; Gao, Y.; Ruan, J.; Wang, L.; Li, H.; Hong, G. A qualitative prognostic biomarker for melanoma based on the relative methylation orderings of CpG loci. Epigenetics 2025, 20, 2487316. [Google Scholar] [CrossRef]
  57. Meevassana, J.; Varophas, S.; Prabsattru, P.; Kamolratanakul, S.; Ruangritchankul, K.; Kitkumthorn, N. 5-Methylcytosine immunohistochemistry for predicting cutaneous melanoma prognosis. Sci. Rep. 2024, 14, 7554. [Google Scholar] [CrossRef]
  58. Masrour, M.; Khanmohammadi, S.; Fallahtafti, P.; Hashemi, S.M.; Rezaei, N. Long non-coding RNA as a potential diagnostic and prognostic biomarker in melanoma: A systematic review and meta-analysis. J. Cell. Mol. Med. 2024, 28, e18109. [Google Scholar] [CrossRef]
  59. Guo, S.; Chen, J.; Yi, X.; Lu, Z.; Guo, W. Identification and validation of ferroptosis-related lncRNA signature as a prognostic model for skin cutaneous melanoma. Front. Immunol. 2022, 13, 985051. [Google Scholar] [CrossRef]
  60. Zhou, J.-G.; Liang, B.; Liu, J.-G.; Jin, S.-H.; He, S.-S.; Frey, B.; Gu, N.; Fietkau, R.; Hecht, M.; Ma, H.; et al. Identification of 15 lncRNAs Signature for Predicting Survival Benefit of Advanced Melanoma Patients Treated with Anti-PD-1 Monotherapy. Cells 2021, 10, 977. [Google Scholar] [CrossRef]
  61. Sorroche, B.P.; Teixeira, R.d.J.; Pereira, C.A.D.; Santana, I.V.V.; Vujanovic, L.; Vazquez, V.d.L.; Arantes, L.M.R.B. PD-L1 Tumor Expression as a Predictive Biomarker of Immune Checkpoint Inhibitors’ Response and Survival in Advanced Melanoma Patients in Brazil. Diagnostics 2023, 13, 1041. [Google Scholar] [CrossRef] [PubMed]
  62. Ellebaek, E.; Khan, S.; Bastholt, L.; Schmidt, H.; Haslund, C.A.; Donia, M.; Svane, I.M. PD-L1 is a biomarker of real-world clinical outcomes for anti-CTLA-4 plus anti-PD-1 or anti-PD-1 monotherapy in metastatic melanoma. Eur. J. Cancer 2024, 198, 113476. [Google Scholar] [CrossRef]
  63. Reschke, R.; Enk, A.H.; Hassel, J.C. Prognostic Biomarkers in Evolving Melanoma Immunotherapy. Am. J. Clin. Dermatol. 2025, 26, 213–223. [Google Scholar] [CrossRef]
  64. Gide, T.N.; Paver, E.C.; Yaseen, Z.; Maher, N.; Adegoke, N.; Menzies, A.M.; da Silva, I.P.; Wilmott, J.S.; Long, G.V.; Scolyer, R.A. Lag-3 expression and clinical outcomes in metastatic melanoma patients treated with combination anti-lag-3 + anti-PD-1-based immunotherapies. Oncoimmunology 2023, 12, 2261248. [Google Scholar] [CrossRef]
  65. Arimura, K.; Hiroshima, K.; Nagashima, Y.; Nakazawa, T.; Ogihara, A.; Orimo, M.; Sato, Y.; Katsura, H.; Kanzaki, M.; Kondo, M.; et al. LAG3 is an independent prognostic biomarker and potential target for immune checkpoint inhibitors in malignant pleural mesothelioma: A retrospective study. BMC Cancer 2023, 23, 1206. [Google Scholar] [CrossRef]
  66. Machiraju, D.; Wiecken, M.; Lang, N.; Hülsmeyer, I.; Roth, J.; Schank, T.E.; Eurich, R.; Halama, N.; Enk, A.; Hassel, J.C. Soluble immune checkpoints and T-cell subsets in blood as biomarkers for resistance to immunotherapy in melanoma patients. Oncoimmunology 2021, 10, 1926762. [Google Scholar] [CrossRef]
  67. Wiecken, M.; Machiraju, D.; Chakraborty, S.; Mayr, E.-M.; Lenoir, B.; Eurich, R.; Richter, J.; Pfarr, N.; Halama, N.; Hassel, J.C. The immune checkpoint LAG-3 is expressed by melanoma cells and correlates with clinical progression of the melanoma. Oncoimmunology 2025, 14, 2430066. [Google Scholar] [CrossRef]
  68. Navani, V.; Graves, M.C.; Marchett, G.C.; Mandaliya, H.; Bowden, N.A.; van der Westhuizen, A. Overall survival in metastatic melanoma correlates with pembrolizumab exposure and T cell exhaustion markers. Pharmacol. Res. Perspect. 2021, 9, e00808. [Google Scholar] [CrossRef] [PubMed]
  69. Ferreira, A.M.C.; Carron, J.; Gomez, G.V.B.; Vazquez, V.L.; Serrano, S.V.; Lourenço, G.J.; Lima, C.S.P. Association of Cytotoxic T-Lymphocyte Antigen-4 (CTLA-4) Genetic Variants with Risk and Outcome of Cutaneous Melanoma. Int. J. Mol. Sci. 2024, 25, 12327. [Google Scholar] [CrossRef]
  70. Pistillo, M.P.; Carosio, R.; Grillo, F.; Fontana, V.; Mastracci, L.; Morabito, A.; Banelli, B.; Tanda, E.; Cecchi, F.; Dozin, B.; et al. Phenotypic characterization of tumor CTLA-4 expression in melanoma tissues and its possible role in clinical response to Ipilimumab. Clin. Immunol. 2020, 215, 108428. [Google Scholar] [CrossRef] [PubMed]
  71. Mastracci, L.; Fontana, V.; Queirolo, P.; Carosio, R.; Grillo, F.; Morabito, A.; Banelli, B.; Tanda, E.; Boutros, A.; Dozin, B.; et al. Response to ipilimumab therapy in metastatic melanoma patients: Potential relevance of CTLA-4+ tumor infiltrating lymphocytes and their in situ localization. Cancer Immunol. Immunother. CII 2020, 69, 653–662. [Google Scholar] [CrossRef]
  72. Dębniak, T.; Baszuk, P.; Rowińska, K.; Feherpataky, M.; Boer, M.; Kiedrowicz, M.; Marchlewicz, M.; Cybulski, C.; Jakubowska, A.; Huzarski, T.; et al. MC1R Common Variants and the 10-Year Survival after Melanoma in Polish Population. Clin. Oncol. 2024, 9, 2061. [Google Scholar]
  73. Su, D.G.; Djureinovic, D.; Schoenfeld, D.; Marquez-Nostra, B.; Olino, K.; Jilaveanu, L.; Kluger, H. Melanocortin 1 receptor (MC1R) expression as a marker of progression in melanoma. JCO Precis. Oncol. 2024, 8, e2300702. [Google Scholar] [CrossRef]
  74. Sun, X.; Kaufman, P.D. Ki-67: More than a proliferation marker. Chromosoma 2018, 127, 175–186. [Google Scholar] [CrossRef] [PubMed]
  75. DiVincenzo, M.J.; Angell, C.D.; Suarez-Kelly, L.P.; Ren, C.; Barricklow, Z.; Moufawad, M.; Fadda, P.; Yu, L.; Backes, F.J.; Ring, K.; et al. Expression of microRNAs and their target genes in melanomas originating from gynecologic sites. PLoS ONE 2023, 18, e0285804. [Google Scholar] [CrossRef] [PubMed]
  76. Cheng, Y.; Shen, P. miR-335 Acts as a Tumor Suppressor and Enhances Ionizing Radiation-Induced Tumor Regression by Targeting ROCK1. Front. Oncol. 2020, 10, 278. [Google Scholar] [CrossRef] [PubMed]
  77. Prodan, M.; Costescu, S.; Elagez, A.; Laitin, S.M.D.; Bloanca, V.; Crainiceanu, Z.; Seclaman, E.; Toma, A.-O.; Fericean, R.M.; Puenea, G.; et al. Molecular Markers in Melanoma Progression: A Study on the Expression of miRNA Gene Subtypes in Tumoral vs. Benign Nevi. Curr. Oncol. (Tor. Ont.) 2024, 31, 2881–2894. [Google Scholar] [CrossRef]
  78. Nakahara, S.; Fukushima, S.; Okada, E.; Morinaga, J.; Kubo, Y.; Tokuzumi, A.; Matsumoto, S.; Tsuruta-Kadohisa, M.; Kimura, T.; Kuriyama, H.; et al. MicroRNAs that predict the effectiveness of anti-PD-1 therapies in patients with advanced melanoma. J. Dermatol. Sci. 2020, 97, 77–79. [Google Scholar] [CrossRef]
  79. Du, J.; Zhang, Q.; Zhang, J.; Maihemuti, M.; He, H.; Jiang, R. Melanoma cell line-derived exosomal miR-424-5p: A key promoter of angiogenesis through LATS2 interaction. Oncol. Res. 2025, 33, 357–367. [Google Scholar] [CrossRef]
  80. Segundo-Val, I.S.; Sanz-Lozano, C.S. Introduction to the Gene Expression Analysis. Methods Mol. Biol. 2016, 1434, 29–43. [Google Scholar]
  81. Bailey, C.N.; Martin, B.J.; Petkov, V.I.; Schussler, N.C.; Stevens, J.L.; Bentler, S.; Cress, R.D.; Doherty, J.A.; Durbin, E.B.; Gomez, S.L.; et al. 31-Gene Expression Profile Testing in Cutaneous Melanoma and Survival Outcomes in a Population-Based Analysis: A SEER Collaboration. JCO Precis. Oncol. 2023, 7, e2300044. [Google Scholar] [CrossRef]
  82. Sun, J.; Karasaki, K.M.; Farma, J.M. The Use of Gene Expression Profiling and Biomarkers in Melanoma Diagnosis and Predicting Recurrence: Implications for Surveillance and Treatment. Cancers 2024, 16, 583. [Google Scholar] [CrossRef]
  83. Mulder, E.; Dwarkasing, J.; Tempel, D.; van der Spek, A.; Bosman, L.; Verver, D.; Mooyaart, A.; van der Veldt, A.; Verhoef, C.; Nijsten, T.; et al. Validation of a clinicopathological and gene expression profile model for sentinel lymph node metastasis in primary cutaneous melanoma. Br. J. Dermatol. 2021, 184, 944–951. [Google Scholar] [CrossRef]
  84. Perzia, B.M.; Heng, J.S.; Sinard, J.; Lim, R. Telomerase gene promoter (TERT) mutation in recurrent uveal melanoma after secondary enucleation revealed by targeted next-generation sequencing. Investig. Ophthalmol. Vis. Sci. 2022, 63, 2350-A0019. [Google Scholar]
  85. Budczies, J.; Kazdal, D.; Menzel, M.; Beck, S.; Kluck, K.; Altbürger, C.; Schwab, C.; Allgäuer, M.; Ahadova, A.; Kloor, M.; et al. Tumour mutational burden: Clinical utility, challenges and emerging improvements. Nat. Rev. Clin. Oncol. 2024, 21, 725–742. [Google Scholar] [CrossRef]
  86. Czarnecka, A.M.; Bartnik, E.; Fiedorowicz, M.; Rutkowski, P. Targeted Therapy in Melanoma and Mechanisms of Resistance. Int. J. Mol. Sci. 2020, 21, 4576. [Google Scholar] [CrossRef]
  87. Nagore, E.; Requena, C.; Traves, V.; Guillen, C.; Hayward, N.K.; Whiteman, D.C.; Hacker, E. Prognostic value of BRAF mutations in localized cutaneous melanoma. J. Am. Acad. Dermatol. 2014, 70, 858–862.e2. [Google Scholar] [CrossRef]
  88. Rizzetto, G.; De Simoni, E.; Molinelli, E.; Offidani, A.; Simonetti, O. Efficacy of Pembrolizumab in Advanced Melanoma: A Narrative Review. Int. J. Mol. Sci. 2023, 24, 12383. [Google Scholar] [CrossRef]
  89. Guida, M.; Strippoli, S.; Ferretta, A.; Bartolomeo, N.; Porcelli, L.; Maida, I.; Azzariti, A.; Tommasi, S.; Grieco, C.; Guida, S.; et al. Detrimental effects of melanocortin-1 receptor (MC1R) variants on the clinical outcomes of BRAF V600 metastatic melanoma patients treated with BRAF inhibitors. Pigment Cell Melanoma Res. 2016, 29, 679–687. [Google Scholar] [CrossRef] [PubMed]
  90. Curti, B.D.; Faries, M.B. Recent Advances in the Treatment of Melanoma. N. Engl. J. Med. 2021, 384, 2229–2240. [Google Scholar] [CrossRef] [PubMed]
  91. Poulikakos, P.I.; Sullivan, R.J.; Yaeger, R. Molecular Pathways and Mechanisms of BRAF in Cancer Therapy. Clin. Cancer Res. 2022, 28, 4618–4628. [Google Scholar] [CrossRef]
  92. Maher, N.G.; Vergara, I.A.; Long, G.V.; Scolyer, R.A. Prognostic and predictive biomarkers in melanoma. Pathology 2024, 56, 259–273. [Google Scholar] [CrossRef]
  93. Teixido, C.; Castillo, P.; Martinez-Vila, C.; Arance, A.; Alos, L. Molecular Markers and Targets in Melanoma. Cells 2021, 10, 2320. [Google Scholar] [CrossRef]
  94. Szabó, I.L.; Emri, G.; Ladányi, A.; Tímár, J. Clinical Applications of the Molecular Landscape of Melanoma: Integration of Research into Diagnostic and Therapeutic Strategies. Cancers 2025, 17, 1422. [Google Scholar] [CrossRef] [PubMed]
  95. Jia, D.-D.; Li, T. Comprehensive insights on genetic alterations and immunotherapy prognosis in Chinese melanoma patients. Sci. Rep. 2024, 14, 16607. [Google Scholar] [CrossRef] [PubMed]
  96. Kashani-Sabet, M.; Leachman, S.A.; Stein, J.A.; Arbiser, J.L.; Berry, E.G.; Celebi, J.T.; Curiel-Lewandrowski, C.; Ferris, L.K.; Grant-Kels, J.M.; Grossman, D.; et al. Early Detection and Prognostic Assessment of Cutaneous Melanoma: Consensus on Optimal Practice and the Role of Gene Expression Profile Testing. JAMA Dermatol. 2023, 159, 545–553. [Google Scholar] [CrossRef]
  97. Tonella, L.; Pala, V.; Ponti, R.; Rubatto, M.; Gallo, G.; Mastorino, L.; Avallone, G.; Merli, M.; Agostini, A.; Fava, P.; et al. Prognostic and Predictive Biomarkers in Stage III Melanoma: Current Insights and Clinical Implications. Int. J. Mol. Sci. 2021, 22, 4561. [Google Scholar] [CrossRef] [PubMed]
  98. Puglisi, R.; Bellenghi, M.; Pontecorvi, G.; Pallante, G.; Carè, A.; Mattia, G. Biomarkers for Diagnosis, Prognosis and Response to Immunotherapy in Melanoma. Cancers 2021, 13, 2875. [Google Scholar] [CrossRef]
  99. Roccuzzo, G.; Sarda, C.; Pala, V.; Ribero, S.; Quaglino, P. Prognostic biomarkers in melanoma: A 2023 update from clinical trials in different therapeutic scenarios. Expert Rev. Mol. Diagn. 2024, 24, 379–392. [Google Scholar] [CrossRef]
  100. Hossain, S.M.; Carpenter, C.; Eccles, M.R. Genomic and Epigenomic Biomarkers of Immune Checkpoint Immunotherapy Response in Melanoma: Current and Future Perspectives. Int. J. Mol. Sci. 2024, 25, 7252. [Google Scholar] [CrossRef]
  101. Adeuyan, O.; Gordon, E.R.; Kenchappa, D.; Bracero, Y.; Singh, A.; Espinoza, G.; Geskin, L.J.; Saenger, Y.M. An update on methods for detection of prognostic and predictive biomarkers in melanoma. Front. Cell Dev. Biol. 2023, 11, 1290696. [Google Scholar] [CrossRef]
  102. He, R.; Lu, J.; Feng, J.; Lu, Z.; Shen, K.; Xu, K.; Luo, H.; Yang, G.; Chi, H.; Huang, S. Advancing immunotherapy for melanoma: The critical role of single-cell analysis in identifying predictive biomarkers. Front. Immunol. 2024, 15, 1435187. [Google Scholar] [CrossRef]
  103. Tímár, J.; Ladányi, A. Molecular Pathology of Skin Melanoma: Epidemiology, Differential Diagnostics, Prognosis and Therapy Prediction. Int. J. Mol. Sci. 2022, 23, 5384. [Google Scholar] [CrossRef] [PubMed]
  104. Svedman, F.C.; Das, I.; Tuominen, R.; Ramqvist, E.D.; Höiom, V.; Brage, S.E. Proliferation and Immune Response Gene Signatures Associated with Clinical Outcome to Immunotherapy and Targeted Therapy in Metastatic Cutaneous Malignant Melanoma. Cancers 2022, 14, 3587. [Google Scholar] [CrossRef] [PubMed]
  105. Pilla, L.; Alberti, A.; Di Mauro, P.; Gemelli, M.; Cogliati, V.; Cazzaniga, M.E.; Bidoli, P.; Maccalli, C. Molecular and Immune Biomarkers for Cutaneous Melanoma: Current Status and Future Prospects. Cancers 2020, 12, 3456. [Google Scholar] [CrossRef] [PubMed]
  106. Seyhan, A.A.; Carini, C. Insights and Strategies of Melanoma Immunotherapy: Predictive Biomarkers of Response and Resistance and Strategies to Improve Response Rates. Int. J. Mol. Sci. 2022, 24, 41. [Google Scholar] [CrossRef]
  107. He, Y.; Wang, X. Identifying biomarkers associated with immunotherapy response in melanoma by multi-omics analysis. Comput. Biol. Med. 2023, 167, 107591. [Google Scholar] [CrossRef]
Table 1. Summary of Updated Diagnostic and Prognostic Biomarkers in Malignant Melanoma. Abbreviations: OS: overall survival, SLN: sentinel lymph node, PFS: progression free survival, DFS: disease-free survival, ICI: immune-checkpoint inhibitor.
Table 1. Summary of Updated Diagnostic and Prognostic Biomarkers in Malignant Melanoma. Abbreviations: OS: overall survival, SLN: sentinel lymph node, PFS: progression free survival, DFS: disease-free survival, ICI: immune-checkpoint inhibitor.
BiomarkerGeneAssociationIndependence
from AJCC
Covariates
Clinical
Readiness
Immunohistochemical and Tissue-Based Prognostic Markers
PRAME [10,11,12,13]PRAMEElevated PRAME protein expression is linked to worse overall survival (OS).UnclearValidated/used
Ki-67 [10,14,15]MKi67Elevated Ki67 predicts worse OS and higher sentinel lymph node (SLN) positivity.YesPromising
AMBRA1 and loricin 1 [16]AMBRA1/LORElevated AMBL or was associated with low risk of recurrence.YesPromising
Y-box binding protein 1 [17] YB-1Elevated YB-1 expression is associated with epithelial-to-mesenchymal transition and poor survival in melanoma.UnclearInvestigational
Microphthalmia transcription factor [18] MITFActs as a transcription factor in melanoma; high expression promotes proliferation; low expression is linked to invasion and therapy resistance.UnclearInvestigational
Transient receptor potential melastatin-1 [19,20,21]TRPM1Downregulated in metastatic melanoma; loss of TRPM1 is associated with increased invasiveness and poor prognosis.UnclearInvestigational
Circulating Biomarkers and Liquid Biopsies
Lactate Dehydrogenase (LDH) [22,23,24,25] LDHA/LDHBElevated LDH expression is linked with worse OS and shorter progression-free survival (PFS).YesValidated/used
S100B [26,27,28]S100Elevated S100B is linked to worse outcomes and advanced stages.UnclearValidated/used
MIA [29,30]MIAElevated MIA is linked with advanced disease stage, worse tumor burden, and worse overall prognosis.UnclearValidated/used
Circulating Tumor DNA [31,32,33,34,35]ctDNAPlasma ctDNA detection was associated with an overall worse prognosis and mortality.
Patients negative for BRAF mutation-positive ctDNA also had better responses to the MEK inhibitor (trametinib) and the BRAF inhibitor (dabrafenib).
Low ctDNA concentrations are associated with better disease-specific survival and PFS, as opposed to higher concentrations.
YesPromising
Cell free DNA (CfDNA) and Cell free RNA (CfRNA) [34,35,36,37,38]CfDNA presence correlates with higher tumor burden and advanced stage. Elevated CfRNA is linked with shorter PFS.NoInvestigational/promising
MicroRNA
miR-200a-3p [39]miR-200a-3pElevated levels were significantly associated with poorer OS.UnclearInvestigational
miR-335-5p [39]miR-335-5pElevated levels were significantly associated with poorer OS.UnclearInvestigational
miR-451a [39]miR-451aElevated levels correlated with improved survival rates.UnclearInvestigational
miR-29b-3p [39]miR-29b-3pPatients with increased expression showed better survival outcomes.UnclearInvestigational
miR-424 [40]miR-424Elevated expression has shown decreased OS and DFS compared to those with low expression.UnclearInvestigational
Genomic and Transcriptomic Biomarkers
11-gene GEP [41]“High-risk” GEP score had significant differences in melanoma-specific survival, distant metastasis-free survival, and RFS compared to “low-risk”.UnclearPromising
CP-GEP [42]
The CP-GEP model combines Breslow thickness and patient age, with the expression of eight genes in the primary tumor.
High-risk CP-GEP had considerably worse five-year RFS than the low-risk patient group.UnclearPromising
TERT Promoter Mutation [43]TERTTERT-mutated melanoma patients had a significantly worse OS.UnclearPromising
Tumor Mutational Burden (TMB) [44,45]High TMB predicts OS after first-line ICIs and PFS.Unclear
BRAF [46,47]BRAFThe BRAF V600E mutation promotes tumor proliferation and is associated with more aggressive disease, brain metastasis, and shorter survival in patients with advanced melanoma.YesValidated/Used
NRAS [48,49,50]NRASNRAS mutations are associated with aggressive tumor behavior, increased risk of nodal relapse and metastasis, and poorer outcomes.UnclearPromising
CDKN2A [51,52,53,54]CDKN2ALoss or mutation of CDKN2A is linked to earlier melanoma onset, multiple primary melanomas, and worse melanoma-specific survival in unscreened patients.UnclearValidated/used
Tissue Epigenetic Biomarkers (DNA methylation) [55,56,57]Global hypomethylation and specific CpG signatures are associated with tumor aggressiveness and poor survival.UnclearPromising
Long noncoding RNAs (lncRNA) [58,59,60]Altered lncRNA expression is significantly associated with OS; specific lncRNA panels may predict prognosis and ICI therapy response.UnclearInvestigational
Immune-related Markers
PD-L1 [61,62,63]CD274PD-L1 expression is correlated with improved OS.YesValidated/used
LAG-3 [64,65]LAG3LAG-3 was associated with poor prognosis.UnclearPromising
TIM-3 [66,67,68]HAVCR2High TIM-3 protein expression on tumor-infiltrating lymphocytes is associated with resistance to anti–PD-1 therapy and variable survival outcomes.UnclearPromising
CTLA-4 [69,70,71]CTLA-4CTLA-4 expression is associated with worsened event-free survival. Higher tumor/TIL expression may predict a better response to ipilimumab (anti-CTLA-4).UnclearPromising
MC1R Variants [72,73]MC1RHigher MC1R expression was associated with worse OS in primary and metastatic melanomas.UnclearInvestigational
Table 2. FDA-approved Therapies for Malignant Melanoma and The Role of Associated Molecular or Genetic Biomarkers in Treatment Selection and Outcome Prediction. Abbreviations: OS: overall survival, ORR: overall response rate, PFS: progression-free survival).
Table 2. FDA-approved Therapies for Malignant Melanoma and The Role of Associated Molecular or Genetic Biomarkers in Treatment Selection and Outcome Prediction. Abbreviations: OS: overall survival, ORR: overall response rate, PFS: progression-free survival).
FDA-Approved TherapyMolecular/Genetic BiomarkerRole of Biomarker in Therapy SelectionPredictive Value for Treatment Outcome
BRAF inhibitors (vemurafenib, dabrafenib, encorafenib) [90,92,93,94]BRAF V600E/K mutationRequired for therapy selection; only patients with BRAF V600 mutations are eligibleStrong predictor of response; high ORR and PFS improvement with BRAF/MEK inhibitors
MEK inhibitors (trametinib, cobimetinib, binimetinib) (used in combination with BRAF inhibitors) [90,92,93]BRAF V600E/K mutationUsed only in combination with BRAF inhibitors in BRAF-mutant melanomaCombination improves progression-free survival (PFS) and overall survival (OS) vs. BRAF inhibitor alone
Anti–PD-1 antibodies (pembrolizumab, nivolumab) [90,92,93,95,96,97]None required; BRAF status not requiredUsed in all advanced melanoma regardless of mutation statusPredictive biomarkers (PD-L1, tumor mutational burden, TMB) not required for selection; benefit seen across subgroups
Anti–CTLA-4 antibody (ipilimumab) [90,92,95]None requiredUsed in all advanced melanoma regardless of mutation statusNo validated predictive biomarker; used alone or in combination with anti–PD-1
Oncolytic virus (Talimogene laherparepvec, T-VEC) [90,92]None requiredUsed for injectable, unresectable cutaneous, subcutaneous, and nodal lesionsNo validated predictive biomarker; best for limited disease burden
KIT inhibitors (imatinib, off-label use) [95,98]KIT mutation (rare, acral/mucosal)Considered in KIT-mutant melanoma (not FDA-approved for melanoma)KIT mutation predicts response in select cases
PD-L1 inhibitors (atezolizumab, in combination) [90]None requiredUsed in combination with BRAF/MEK inhibitors in BRAF-mutant melanomaNo validated predictive biomarker; PD-L1 expression not required
Table 3. Emerging Investigational Biomarkers Under Study for Prognostic and Predictive Use in Malignant Melanoma. Abbreviations: ICI: immune-checkpoint inhibitor).
Table 3. Emerging Investigational Biomarkers Under Study for Prognostic and Predictive Use in Malignant Melanoma. Abbreviations: ICI: immune-checkpoint inhibitor).
Biomarker/SignatureInvestigational StatusPotential Clinical Impact
LAG-3 expression [63,100]Early-phase clinical/translational studies; not standardMay predict response/resistance to immune-checkpoint inhibitors (ICIs)
Circulating tumor DNA (ctDNA) [63,99]Prospective validation ongoing; translational researchEarly detection of relapse; dynamic monitoring of treatment response
Immune cell phenotyping (TILs, peripheral blood) [63,102]Under investigation with single-cell and spatial profilingMay stratify prognosis and predict ICI response; not yet validated
Gut microbiota composition [63,100]Exploratory clinical studiesPotentially modulates ICI efficacy; not yet actionable
Gene expression signatures (e.g., allograft rejection, IFN-γ, inflammatory response, E2F/MYC downregulation) [99,103,104]Identified in transcriptomic studies; validation requiredMay predict benefit from ICI or targeted therapy; not yet standard
Mast cell and dendritic cell activation [107]Multi-omics and translational studiesNegative correlation with ICI response; potential for risk stratification
Oncogenic pathway enrichment (JAK-STAT, RAS, MAPK, HIF-1, PI3K-Akt, VEGF) [107]Multi-omics studies; not in clinical useMay predict ICI response or resistance; further validation needed
MicroRNA and protein expression profiles [105,107]Identified in omics studies; not validatedPotential predictive/prognostic markers for ICI response
Intratumor heterogeneity (ITH) [103,107]Genomic/transcriptomic studiesMay predict resistance to immunotherapy; not yet clinically actionable
Epigenomic signatures (e.g., DNA methylation, chromatin modifiers) [100,106]Early translational researchMay influence ICI response; under investigation
Tumor-associated antibodies [102]Single-cell and serological studiesPotential for non-invasive prediction of response; not validated
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

Fleshner, L.; Sayegh, A.; Atak, M.F.; Hirani, R.; Farabi, B.; Safai, B.; Marmon, S. Molecular and Genetic Markers for Malignant Melanoma: Implications for Prognosis and Therapy. Dermatopathology 2025, 12, 31. https://doi.org/10.3390/dermatopathology12030031

AMA Style

Fleshner L, Sayegh A, Atak MF, Hirani R, Farabi B, Safai B, Marmon S. Molecular and Genetic Markers for Malignant Melanoma: Implications for Prognosis and Therapy. Dermatopathology. 2025; 12(3):31. https://doi.org/10.3390/dermatopathology12030031

Chicago/Turabian Style

Fleshner, Lauren, Alyssa Sayegh, Mehmet Fatih Atak, Rahim Hirani, Banu Farabi, Bijan Safai, and Shoshana Marmon. 2025. "Molecular and Genetic Markers for Malignant Melanoma: Implications for Prognosis and Therapy" Dermatopathology 12, no. 3: 31. https://doi.org/10.3390/dermatopathology12030031

APA Style

Fleshner, L., Sayegh, A., Atak, M. F., Hirani, R., Farabi, B., Safai, B., & Marmon, S. (2025). Molecular and Genetic Markers for Malignant Melanoma: Implications for Prognosis and Therapy. Dermatopathology, 12(3), 31. https://doi.org/10.3390/dermatopathology12030031

Article Metrics

Back to TopTop