Next Article in Journal
Correlation Between Body Plethysmography and Impulse Oscillometry Across Obstructive and Restrictive Lung Diseases: Evidence from an Adult Pilot Study Cohort
Previous Article in Journal
Evaluating GPT-5 for Melanoma Detection Using Dermoscopic Images
Previous Article in Special Issue
Myxoid Glomus Tumors Showing CD34 Expression: A Series of Eight Cases
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Toward Personalized Response Monitoring in Melanoma Patients Treated with Immunotherapy and Target Therapy

1
Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum, University of Bologna, 40138 Bologna, Italy
2
Oncologic Dermatology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
3
Oncology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
4
Plastic Surgery, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
5
Department of Medical Science, Dermatology Clinic, University of Turin, 10126 Turin, Italy
6
Dermatology Unit, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milano, Italy
7
Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milano, Italy
*
Author to whom correspondence should be addressed.
Diagnostics 2025, 15(23), 3054; https://doi.org/10.3390/diagnostics15233054 (registering DOI)
Submission received: 5 November 2025 / Revised: 25 November 2025 / Accepted: 28 November 2025 / Published: 29 November 2025

Abstract

Background/Objectives: Immunotherapy and targeted therapy have revolutionized the treatment of advanced cutaneous melanoma. However, predicting individual response and managing resistance remain major challenges. This narrative review aims to evaluate the prognostic and predictive value of treatment-related adverse events (TRAEs) and circulating biomarkers—including lactate dehydrogenase (LDH), circulating tumor DNA (ctDNA), and microRNAs (miRNAs)—in anticipating therapeutic outcomes and personalizing treatment strategies. Methods: A comprehensive literature search was conducted across PubMed, Scopus, and Web of Science for studies published between January 2010 and September 2025. Eligible studies included clinical trials, observational cohorts, and translational research evaluating biomarkers or toxicity profiles in melanoma patients receiving immune checkpoint inhibitors or BRAF/MEK inhibitors. Emphasis was placed on dynamic indicators of treatment efficacy and integrative modeling approaches. Results: Evidence indicates that the emergence of low-to-moderate grade TRAEs—especially immune-related events like vitiligo, thyroiditis, and rash—is positively associated with response to immunotherapy. Similarly, pyrexia and dermatologic toxicities may correlate with outcomes under BRAF/MEK inhibition. ctDNA clearance within 6–12 weeks of therapy strongly predicts durable response and precedes radiologic changes. Specific miRNAs (e.g., miR-21-5p, miR-146a-5p) demonstrate dynamic modulation during treatment and may signal response or resistance. Interferon-driven gene expression profiles further stratify tumors into “hot” or “cold” immune phenotypes, refining predictive accuracy. Conclusions: Integrative models combining TRAEs, ctDNA, miRNA signatures, and interferon-related gene expression offer a multi-dimensional framework for early, individualized response monitoring. Prospective validation, harmonization of assays, and incorporation into adaptive clinical workflows are key to translating these insights into personalized melanoma care.

1. Introduction

Cutaneous melanoma remains one of the most aggressive forms of skin cancer, accounting for the majority of skin cancer-related deaths. In the United States, cutaneous melanoma comprises over 90% of melanoma cases and is associated with thousands of deaths annually, with mortality rates far exceeding those of nonmelanoma skin cancers, which rarely result in death [1,2,3,4,5,6,7,8,9,10]. In recent years, the therapeutic landscape has been transformed by the advent of immune checkpoint inhibitors (ICIs) targeting PD-1/PD-L1 and CTLA-4, as well as targeted therapies against BRAF and MEK in patients harboring BRAF mutations [11,12,13,14]. Despite these advances, not all patients respond equally, and durable benefit is limited to a subset. As a result, there is a pressing need for reliable predictive biomarkers to guide treatment decisions and personalize care [15]. Traditional predictors such as baseline tumor burden, lactate dehydrogenase (LDH) levels, and performance status offer limited granularity. Recently, attention has turned to on-treatment indicators, including the emergence of treatment-related adverse events (TRAEs), which may reflect immune system activation or off-target effects associated with efficacy. In parallel, circulating biomarkers such as ctDNA and microRNAs (miRNAs) are being explored as minimally invasive tools to monitor treatment response in real time. This review evaluates the potential of both toxicity profiles and circulating biomarkers to serve as early predictors of response and long-term outcomes in melanoma patients.

2. Materials and Methods

This narrative review was conducted to comprehensively synthesize current evidence on clinical, molecular, and immunologic biomarkers for treatment response prediction and monitoring in patients with cutaneous melanoma treated with ICIs and targeted therapies. A comprehensive literature search was performed across PubMed/MEDLINE, Scopus, and Web of Science databases for studies published between January 2010 and September 2025. The search strategy combined both Medical Subject Headings (MeSH) and free-text terms related to the main concepts of the review: “melanoma,” “immune checkpoint inhibitors,” “targeted therapy,” “immune-related adverse events,” “BRAF/MEK inhibitors,” “circulating tumor DNA,” “microRNA,” “liquid biopsy,” “interferon signature,” “immune-inflamed tumor,” “predictive biomarkers,” and “treatment response.” Eligible studies included original research articles (prospective or retrospective), randomized controlled trials, meta-analyses, and significant mechanistic or translational studies investigating biomarkers of treatment response, resistance, or toxicity in melanoma. Exclusion criteria included case reports, editorials, non-peer-reviewed preprints, and animal-only studies unless they provided mechanistic insights directly supporting clinical findings. Only studies in English were considered. Reference lists of key reviews and seminal articles were manually screened to identify additional relevant publications. Studies were critically appraised for sample size, methodology, reproducibility, and translational impact. Preference was given to large multicenter studies, prospective biomarker validation cohorts, and high-impact mechanistic investigations (Figure 1).

3. Results

3.1. Treatment-Related Adverse Events (TRAEs) and Their Association with Therapeutic Efficacy

The development of TRAEs has emerged as a potential surrogate marker of therapeutic efficacy in melanoma, reflecting on-target biological activity and systemic immune engagement. Among these, immune-related adverse events (irAEs) associated with immune checkpoint inhibitors (ICIs) and targeted therapy–related adverse events (ttAEs) from MAPK pathway inhibition represent two mechanistically distinct but clinically informative phenomena. IrAEs arise from nonspecific activation of the immune system due to PD-1/PD-L1 or CTLA-4 blockade. Numerous retrospective and prospective studies have demonstrated a positive correlation between the occurrence of irAEs and improved clinical outcomes, including higher objective response rates, longer progression-free survival (PFS), and overall survival (OS) [16,17,18,19,20]. In a retrospective cohort of 49 melanoma patients treated with anti-PD-1 monotherapy, Wu et al. found that patients experiencing grade 1–2 irAEs achieved longer median progression-free survival (4.6 vs. 2.5 months; HR 0.52) and a trend toward improved overall survival (15.2 vs. 5.7 months; HR 0.50) compared with those without irAEs [18]. A larger meta-analysis in metastatic melanoma (n = 1474) reported that patients with any irAEs had superior overall survival (median 15.24 vs. lower) compared to those without; the authors estimated a substantial survival advantage in the irAE+ subgroup [20]. More recently, a pan-tumor prospective study further noted a positive correlation between irAEs and improved survival metrics, pointing to the reproducibility of the association beyond melanoma alone [21]. Cutaneous manifestations—such as vitiligo, rash, or pruritus—are among the most frequent and are particularly relevant in melanoma due to antigenic overlap between melanoma cells and normal melanocytes [22,23,24,25,26,27]. The appearance of vitiligo during anti-PD-1 or combination immunotherapy has been consistently associated with robust and durable responses, suggesting a shared immunological mechanism of melanocyte destruction [28,29,30]. Similarly, endocrine irAEs such as thyroiditis or hypophysitis, and gastrointestinal or hepatic toxicities, have been linked to favorable outcomes when adequately managed [31,32,33,34]. Importantly, the timing and grade of irAEs also appear relevant: early-onset, low-to-moderate toxicities may signal effective immune activation, whereas severe or multi-organ irAEs can necessitate treatment discontinuation, potentially offsetting clinical benefit [35,36]. Despite this association, the predictive value of irAEs is not absolute, and their absence does not preclude response. The interplay between immune activation, host genetics (e.g., HLA polymorphisms, cytokine gene variants), and gut microbiota composition may further modulate both efficacy and toxicity [37,38,39].
In parallel, ttAEs observed with BRAF and MEK inhibitors—including pyrexia, rash, arthralgia, photosensitivity, and laboratory abnormalities such as elevated liver enzymes—have also shown potential predictive implications. Pyrexia is among the most common toxicities in patients treated with dabrafenib plus trametinib, affecting up to 59% of patients (all grades) [40,41]. Some analyses suggest that the occurrence of pyrexia may correlate with better outcomes [42,43,44,45,46]. Atkinson et al. reported improved responses in patients experiencing pyrexia under BRAFi/MEKi regimens, suggesting pyrexia might act as a pharmacodynamic biomarker [47]. Earlier ASCO results posited that pyrexia was associated with more durable responses under combination BRAF/MEK therapy. Similarly, Schaefer et al. devised a pyrexia score algorithm combining C-reactive protein (CRP), LDH, leukocyte, and platelet counts; they showed that this score differentiated patients who developed pyrexia, and that systemic biomarker shifts (e.g., increase in CRP, fall in leukocytes/platelets) correlated with the inflammatory response induced by therapy [48,49]. However, the relationship is not uniform: in a study of patients with metastatic BRAF V600E/K melanoma, Menzies et al. observed that while pyrexia was frequent and recurrent under dabrafenib + trametinib, it was not statistically associated with baseline characteristics or drug efficacy, cautioning against overinterpretation [40].
Beyond pyrexia, rash and photosensitivity—common dermatologic toxicities—have been analogized in other kinase inhibitor settings (e.g., EGFR inhibitors) as correlates of adequate target inhibition, though direct evidence in melanoma/triose MAPK contexts remains limited [50,51]. Maculopapular rashes typically appear within the first few weeks of therapy and may be pruritic or desquamative. In a pooled analysis, their development was associated with better disease control and, in some cohorts, longer PFS [52,53]. Photosensitivity reactions, more commonly linked to vemurafenib, can significantly impact patient quality of life and adherence. These cutaneous reactions may represent a marker of on-target inhibition in skin tissues, paralleling drug activity in the tumor microenvironment. Optimal supportive care, including sun protection and topical corticosteroids, is essential to maintain treatment adherence [54]. Unlike irAEs, these toxicities arise from on-target inhibition of the MAPK pathway in normal tissues. Nevertheless, ttAEs can also compromise adherence and quality of life, highlighting the importance of proactive management and dose optimization to sustain long-term benefit [55,56].
The biological distinction between immune-driven and kinase-driven toxicities underscores the heterogeneity of TRAE mechanisms—yet both may act as dynamic indicators of drug–target interaction and patient immune or metabolic responsiveness. Collectively, evidence suggests that the emergence, timing, and severity of TRAEs—whether immune- or targeted-therapy-related—provide valuable, noninvasive clues to therapeutic efficacy. Integrating these clinical observations with molecular and serologic biomarkers (e.g., ctDNA kinetics, cytokine signatures, and microRNA profiles) could enhance early stratification of responders versus non-responders. Prospective, standardized analyses are required to validate TRAEs as functional biomarkers and to define how they can be integrated into predictive algorithms guiding individualized melanoma therapy.

3.2. Circulating Biomarkers

Circulating biomarkers—including conventional serologic markers (LDH, S100B), circulating tumor DNA (ctDNA), and small non-coding RNAs such as microRNAs (miRNAs)—offer complementary information that can anticipate therapeutic efficacy or resistance long before radiologic evidence becomes apparent. These biomarkers reflect different layers of tumor and host interaction: metabolic burden, genomic evolution, and immune modulation, respectively [57].

3.2.1. Serum LDH and S100B

Serum lactate dehydrogenase (LDH) remains one of the most established and clinically implemented biomarkers in melanoma [58,59]. Elevated baseline LDH is incorporated into the AJCC 8th edition staging system as an indicator of poor prognosis, correlating with increased tumor burden, hypoxia-driven glycolytic reprogramming, and aggressive disease biology [9,60]. Multiple studies and meta-analyses have confirmed that elevated LDH predicts shorter PFS and OS in patients treated with both ICIs and BRAF/MEK inhibitors [61,62]. However, the utility of LDH as a dynamic biomarker remains limited due to its lack of specificity and delayed kinetics. Fluctuations may reflect necrosis or inflammation rather than true disease progression [63]. Similarly, S100B, a calcium-binding protein expressed by melanocytes and glial cells, has been validated as a sensitive marker for disease activity and relapse [64,65,66]. Rising S100B during therapy correlates with disease progression, whereas sustained decreases are associated with response. Nonetheless, S100B’s sensitivity for early response detection remains modest, particularly in patients with low tumor burden or isolated organ metastases (e.g., CNS or subcutaneous sites) [67,68,69].

3.2.2. Circulating Tumor DNA (ctDNA)

The emergence of ctDNA as a real-time, quantitative measure of tumor burden has profoundly changed melanoma monitoring [70]. ctDNA, comprising short DNA fragments released from apoptotic or necrotic tumor cells, enables both quantification of tumor load and molecular profiling of resistance mechanisms [71,72]. The detection and dynamics of ctDNA during therapy have shown remarkable correlation with clinical outcomes. Several studies have demonstrated that early ctDNA clearance predicts durable response to immune checkpoint inhibition [73,74,75]. Lee et al. (Nat Med 2020) reported that patients achieving undetectable ctDNA within 12 weeks of anti-PD-1 therapy had significantly longer OS and PFS compared to those with persistent ctDNA [76]. Similarly, Seremet et al. found that a ≥90% decrease in ctDNA at week 6 predicted radiologic response with high specificity, outperforming LDH and radiographic criteria [77]. Under targeted therapy, the dynamics are even more pronounced [75,78,79]. Conversely, ctDNA reappearance often heralds progression weeks before clinical relapse, highlighting its potential as an early resistance marker [80]. Technological advances, such as ultrasensitive digital PCR and next-generation sequencing (NGS) platforms (e.g., CAPP-Seq, BEAMing, and ddPCR), have enabled detection of ctDNA at variant allele frequencies below 0.1% [81]. This sensitivity allows not only monitoring of treatment response but also identification of emerging resistance mutations, such as NRAS or MEK1/2 alterations during BRAF/MEK inhibition, or PTEN loss and β2-microglobulin mutations under ICI therapy [82]. Furthermore, recent multi-omic approaches integrating ctDNA with circulating tumor cells (CTCs) and exosomal RNA signatures provide complementary insights into tumor heterogeneity and immune escape dynamics. Despite these advances, standardization of ctDNA assays remains a major challenge. Differences in pre-analytical handling, sequencing depth, and threshold definitions limit inter-study comparability. Harmonization efforts—such as those proposed by the European Liquid Biopsy Society (ELBS)—are ongoing to establish reference standards and facilitate ctDNA incorporation into clinical trials and real-world response monitoring. These initiatives aim to define pre-analytical and analytical workflows (e.g., blood collection, processing, sequencing), improve inter-laboratory comparability through ring trials, and build consensus on clinical validity and utility endpoints to support ctDNA use as a surrogate biomarker of treatment response [83,84]. The Association for Molecular Pathology and the College of American Pathologists have issued joint consensus recommendations emphasizing the need to report key pre-analytical variables, assay performance metrics, and validation procedures to ensure clinical utility and reproducibility of ctDNA assays [85].

3.2.3. Circulating MicroRNAs (miRNAs)

Among novel circulating biomarkers, miRNAs have gained increasing attention for their potential to mirror both tumor-intrinsic biology and host immune dynamics [10]. These small, non-coding RNAs regulate post-transcriptional gene expression and are remarkably stable in serum, plasma, and extracellular vesicles. In melanoma, deregulated miRNA profiles are associated with tumor progression, metastatic potential, and immune modulation [86,87,88,89,90,91]. Several circulating miRNAs have been implicated as predictors of response to ICIs and targeted therapies [92,93]. For example, elevated baseline levels of miR-21-5p and miR-146a-5p—two key regulators of NF-κB-dependent inflammatory signaling—have been associated with adverse prognosis and resistance to immunotherapy [87]. Dynamic downregulation of these miRNAs during treatment correlates with favorable response, suggesting they reflect tumor-immune equilibrium. Conversely, upregulation of miR-155-5p and miR-34a has been linked to enhanced T-cell activation and longer PFS under anti-PD-1 therapy [94]. Other studies have shown that exosomal miRNAs such as miR-125b or miR-31-5p may predict early progression, while miR-16 and miR-223 correlate with immune-related toxicity, hinting at shared regulatory networks between efficacy and irAEs [95,96]. The integration of exosomal miRNA profiling offers a promising step forward: exosomes protect miRNAs from degradation and originate from both tumor and immune cells, making them reflective of tumor–host crosstalk. In a 2023 study, De Martino et al. identified a three-miRNA plasma signature (namely hsa-miR-200c-3p, hsa-miR-144-3p and hsa-miR-221-3p) that were differentially expressed in plasma-derived exosomes from melanoma patients and controls [97]. Despite these promising data, translational application is still hampered by technical variability (sample processing, normalization, platform heterogeneity) and lack of universally accepted reference miRNAs. Harmonized pipelines and validation in large, prospective cohorts are essential to move these signatures from discovery to clinical use. An emerging frontier involves the integration of multiple circulating biomarkers into composite predictive frameworks. Combined analysis of ctDNA kinetics, LDH levels, and specific miRNA expression patterns improves sensitivity and specificity for early response detection compared to any single biomarker. Similarly, machine-learning algorithms incorporating ctDNA, cytokine panels, and immune cell profiling have been proposed to generate personalized “response probability scores,” opening the way for adaptive treatment strategies.

3.3. Hot and Cold Tumors: Interferon Signatures and Immune-Inflamed Phenotypes

Melanomas (and more broadly solid tumors) can be broadly stratified into “hot” (immune-inflamed/T cell-infiltrated) and “cold” (immune-excluded or immune-desert) phenotypes, which differ fundamentally in their immune microenvironment, transcriptional programs, and responsiveness to immune checkpoint blockade [98]. Key to this stratification is the presence or absence of interferon (IFN)-driven gene expression signatures that reflect active immune engagement within the tumor microenvironment (TME). A seminal work by Ayers et al. introduced a T cell-inflamed gene expression profile (GEP) centered on IFN-γ-responsive genes (e.g., CXCL9, CXCL10, IDO1, STAT1, GZMB) that correlated with response to anti-PD-1 therapy across multiple tumor types, including melanoma [99]. Their analysis showed that baseline expression of this IFN-γ signature predicted clinical benefit better than PD-L1 immunohistochemistry alone, and the signature was subsequently refined and validated in larger cohorts. Subsequent studies in melanoma and pan-cancer cohorts have reinforced the centrality of IFN signaling in distinguishing “hot” from “cold” tumors. Grasso et al. (2020) demonstrated that conserved IFN-γ signaling and T-cell infiltration were among the strongest correlates of clinical response to checkpoint inhibitors, further validating the relevance of IFN signatures [100]. The multi-omic profiling of checkpoint-treated melanoma by Newell et al. confirmed that “hot” tumors are characterized by elevated expression of IFN-γ-responsive genes, abundant CD8+ T cells, and PD-L1 upregulation, while “cold” tumors show suppression of these immune modules [101]. More recently, the Dual Role of Interferon-Gamma in Melanoma was reviewed by Wawrzyniak & Hartman, who emphasize that IFN-γ is not a purely beneficial mediator: although its presence is often favorable (driving antigen presentation, T-cell recruitment, and cytotoxicity), chronic or dysregulated IFN signaling may contribute to immune exhaustion or upregulation of resistance pathways (for instance JAK/STAT pathway defects) that blunt responsiveness to therapy [102]. Likewise, Brockwell et al. reviewed how type I interferons (IFN-α/β) act upstream to prime the innate immune milieu and support the transition to a T cell-inflamed phenotype, thereby promoting responses to checkpoint inhibitors [103]. Beyond static baseline classification, dynamic changes in IFN signature—and shifts from “cold” to “hot” over time—have prognostic significance [102,104]. For instance, the DONIMI trial used a baseline IFN-γ signature to stratify melanoma patients for neoadjuvant immunotherapy, demonstrating that patients with high IFN-γ scores derived benefit from PD-1 monotherapy, whereas low-score tumors might require intensified therapy [105]. In that trial, tumors that converted from IFN-γ-low to -high early under therapy showed a higher rate of pathologic response, indicating that temporal induction of IFN signaling may act as a biomarker of treatment engagement. Another layer of nuance comes from spatially informed gene signatures. A recent study partitioned gene expression by the spatial compartments defined by CD68+ macrophages, CD45+ leukocytes, and tumor cells and found that the strongest predictive signals of response came from genes enriched in IFN and immune pathways within these immune niches, underscoring the importance of local IFN–immune interactions rather than bulk tumor averages [106]. Furthermore, a recent study proposed an ITRGM signature to stratify melanoma patients into immune-hot vs. immune-cold categories, showing that high signature scores correlated with increased immune cell infiltration, tumor mutational burden, and better immunotherapy response across multiple cohorts [107]. Mechanistically, Holzgruber et al. showed that type I interferon signaling in melanoma cells can induce PD-1 expression via the IFNAR–JAK/STAT axis, which has implications for how tumor-intrinsic IFN pathways modulate checkpoint receptor expression and response to therapy [108]. This highlights that both tumor-cell intrinsic and immune-compartment IFN signaling intersect to define the immune phenotype of melanoma, with potential feedback loops influencing ICI sensitivity or resistance.
Collectively, the literature supports a refined view of the hot vs. cold tumor paradigm (Figure 2):
-
Hot tumors: enriched in IFN-γ-driven transcripts, T cell infiltration, chemokine expression, antigen presentation machinery, and adaptive immune activation—generally more responsive to ICIs.
-
Cold tumors: deficient in IFN signature, with limited immune infiltrate, exclusion of effector T cells, and immunosuppressive barriers—often refractory to monotherapy.
-
Dynamic conversion: Some tumors initially cold may “heat up” under therapy, with induced IFN signaling and immune infiltration signaling favorable biological response.
-
Dualistic roles: IFN signatures can also be modulated by resistance mechanisms (e.g., JAK mutations, chronic IFN exposure, induction of inhibitory pathways), complicating their predictive utility.
Incorporating baseline IFN signatures, spatially mapped immune-IFN modules, and dynamic IFN induction into predictive models may therefore deepen our ability to stratify patients, tailor immunotherapy combinations, and monitor early immune activation in melanoma.

4. Discussion

The assessment of treatment response in melanoma is increasingly driven by integrative models that combine clinical, molecular, and immune-derived data, moving beyond single-parameter predictors. Recent studies demonstrate that multidimensional approaches—incorporating imaging techniques, toxicity profiles, circulating biomarkers (such as cell-free DNA, RNA, proteins, and immune cell subsets), and tumor immune signatures—enable more accurate, real-time, and personalized prediction of response to ICIs [109,110,111,112,113,114,115,116]. Clinical models that utilize routine parameters (e.g., performance status, metastatic sites, serum lactate dehydrogenase, neutrophil–lymphocyte ratio) have shown moderate predictive value for response and survival in metastatic melanoma treated with anti-PD-1 ± ipilimumab, with validated nomograms available for clinical decision support [117,118]. Molecular profiling, including genomic, transcriptomic, and immune signatures from tumor tissue, further refines prediction, identifying features such as tumor mutational burden, antigen presentation machinery, and immune cell infiltration as key determinants of response [110,111,119,120,121,122]. Immune-derived data, particularly the characterization of the tumor microenvironment (TME) and peripheral immune cell populations (e.g., circulating PD-1+ CD4+ effector memory T cells), have emerged as independent predictors of progression-free survival and response to ICIs [112,113]. Integrative models that combine these data types outperform single biomarkers and facilitate patient stratification, therapy selection, and monitoring for resistance or toxicity [121,123,124]. This comprehensive framework supports precision medicine in melanoma, allowing clinicians to tailor therapies and optimize outcomes by leveraging multidimensional, real-time data (Table 1).
The findings summarized in this review support a growing paradigm shift in melanoma care—from static, baseline predictors to dynamic, integrative monitoring strategies. Across the included studies, several key themes emerged.
First, the consistent association between low-to-moderate grade TRAEs (particularly vitiligo, thyroiditis, and rash) and treatment response across retrospective and prospective cohorts reinforces their role as on-treatment indicators of immune engagement [125,126,127,128,129,130]. However, most studies were observational and variably reported toxicity grading, warranting caution in interpretation and highlighting the need for standardized toxicity-response models.
Second, ctDNA dynamics—particularly early clearance—demonstrated strong and re-producible correlation with progression-free and overall survival. Several studies used orthogonal validation with radiologic and clinical endpoints, enhancing the robustness of ctDNA as a biomarker. Nevertheless, technical heterogeneity in ctDNA platforms (e.g., ddPCR vs. NGS) remains a major barrier to widespread clinical adoption [78,131,132,133,134,135].
Third, circulating miRNA signatures offered nuanced insights into tumor-immune crosstalk, though the current evidence base is limited by small sample sizes and variability in profiling techniques. Validation in larger, prospective cohorts is essential [136,137,138].
Lastly, interferon-related gene expression profiling (IFN-GEP) emerged as a powerful predictor of ICI response, stratifying tumors into hot vs. cold phenotypes. Its clinical relevance has been demonstrated in both retrospective analyses and prospective stratification trials (e.g., DONIMI). However, the dual role of IFN signaling in both activation and resistance underscores the need for context-aware interpretation [100,102,139,140,141].
Taken together, these findings support a framework in which toxicity patterns, liquid biopsy metrics (ctDNA, miRNAs), and transcriptomic signatures (IFN-GEP) are integrated into early, individualized response monitoring algorithms. Future research should focus on harmonizing assay methodologies, validating composite biomarkers across treatment modalities, and translating these insights into adaptive clinical workflows that can truly personalize melanoma care.
Despite the promising findings reported across studies, several limitations must be acknowledged. First, the underlying evidence is largely based on retrospective analyses, early-phase prospective cohorts, or biomarker sub-studies with limited sample sizes and heterogeneous patient populations. This heterogeneity, along with variability in treatment regimens, follow-up intervals, and outcome definitions, may limit the generalizability of results. Additionally, potential publication bias cannot be excluded, as negative studies are less frequently reported. Second, our review is narrative in nature: while supported by a structured search strategy, we did not perform a formal risk-of-bias assessment nor a meta-analytic synthesis of effect sizes. These limitations should be considered when interpreting the clinical relevance of candidate biomarkers.
Future research should prioritize the prospective validation of promising on-treatment biomarkers such as low-grade immune-related adverse events (e.g., vitiligo, thyroiditis), ctDNA dynamics, and miRNA expression profiles. Integration of these biomarkers into stratified clinical trial designs may help define composite predictors of response. Large-scale, multi-center studies with harmonized methodologies and real-time sampling will be essential to confirm their predictive utility. Additionally, longitudinal multi-omics profiling may offer insights into the temporal evolution of treatment response and resistance mechanisms.
The convergence of clinical, molecular, and immune-derived data is redefining how treatment response in melanoma is assessed. Rather than relying on single-parameter predictors, integrative models that combine toxicity profiles, circulating biomarkers, and tumor immune signatures provide a multidimensional framework for real-time and personalized response prediction.

5. Conclusions

The integration of clinical, molecular, and immune-derived biomarkers is reshaping how therapeutic efficacy is monitored and predicted in melanoma. TRAEs, particularly immune-related toxicities, provide valuable real-time insight into immune activation; while circulating biomarkers such as ctDNA and miRNAs offer dynamic and minimally invasive indicators of tumor burden and biological response. In parallel, interferon-driven gene expression profiles and immune-inflamed (“hot”) tumor signatures have emerged as robust determinants of checkpoint inhibitor sensitivity, bridging the gap between tumor-intrinsic features and host immune competence. Collectively, these complementary dimensions—clinical, serological, and transcriptomic—form the foundation for integrative predictive models capable of anticipating treatment outcomes with unprecedented precision (Figure 3). Although not the primary focus of this review, it is important to acknowledge that standard imaging modalities such as PET/CT and MRI remain integral components of treatment monitoring algorithms. These imaging tools are already embedded in current follow-up protocols and are routinely employed to assess disease burden, detect progression, or confirm radiological responses. For this reason, they were not explicitly discussed in detail here, as our goal was to emphasize emerging, complementary predictors that may enhance or personalize treatment monitoring. Nonetheless, the proposed approach is meant to be integrative, and imaging findings should be interpreted alongside toxicity profiles and molecular biomarkers, in line with established guidelines.
While the algorithm presented here is primarily tailored to melanoma, its conceptual framework—based on dynamic, multimodal predictors such as immune-related toxicities, liquid biopsy dynamics, and transcriptomic signatures—may be broadly applicable across other malignancies [130,132]. For example, immune-related thyroid dysfunction has been correlated with treatment response in non-small cell lung cancer (NSCLC), while early clearance of ctDNA has shown predictive value in colorectal and breast cancers. Several tumor types are now being investigated in prospective trials using composite biomarkers to guide immunotherapy or targeted therapy response. These analogies underscore the potential for a cross-disciplinary approach to personalized response monitoring, and suggest that similar biomarker- and toxicity-based models could be adapted and validated beyond the melanoma setting.
The convergence of these multi-omic indicators within artificial intelligence-based frameworks will likely enable early identification of responders, optimization of therapeutic intensity, and avoidance of unnecessary toxicity. Ultimately, advancing toward personalized response monitoring in melanoma will depend on large-scale prospective validation, standardization of biomarker assays, and incorporation of longitudinal data into adaptive clinical workflows. Such multidimensional and dynamic approaches promise to transform melanoma management from reactive to proactive care, aligning therapeutic decisions with each patient’s unique biological and immune landscape (Table 1).

Author Contributions

Conception and design: F.V. and E.D.; Acquisition of data: F.V., A.G., E.M., B.S., C.B., A.M.A., L.V., E.M.C., B.M., P.V.M., D.T., S.R., M.A.; Analysis and interpretation of data: F.V. and E.D.; Drafting of the manuscript: F.V. and E.D.; Critical revision of the manuscript for important intellectual content: F.V. and E.D. 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 declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
trAEstreatment-related adverse events
irAEsimmune-related adverse events
ICIsimmune checkpoint inhibitors
ttAEsTargeted therapy-related adverse events
LDHlactate dehydrogenase
miRNAsmicroRNAs
PFSprogression-free survival
OSOverall survival
ctDNAcirculating tumor DNA
NGSnext-generation sequencing
ELBSEuropean Liquid Biopsy Society
GEPgene expression profile
TMEtumor microenvironment

References

  1. Tasdogan, A.; Sullivan, R.J.; Katalinic, A.; Lebbe, C.; Whitaker, D.; Puig, S.; van de Poll-Franse, L.V.; Massi, D.; Schadendorf, D. Cutaneous melanoma. Nat. Rev. Dis. Primer 2025, 11, 23. [Google Scholar] [CrossRef]
  2. US Preventive Services Task Force; Grossman, D.C.; Curry, S.J.; Owens, D.K.; Barry, M.J.; Caughey, A.B.; Davidson, K.W.; Doubeni, C.A.; Epling, J.W.; Kemper, A.R.; et al. Behavioral Counseling to Prevent Skin Cancer: US Preventive Services Task Force Recommendation Statement. JAMA 2018, 319, 1134–1142. [Google Scholar] [CrossRef] [PubMed]
  3. Joshi, U.M.; Kashani-Sabet, M.; Kirkwood, J.M. Cutaneous Melanoma: A Review. JAMA 2025. [Google Scholar] [CrossRef] [PubMed]
  4. 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]
  5. De Giorgi, V.; Venturi, F.; Silvestri, F.; Trane, L.; Savarese, I.; Scarfì, F.; Cencetti, F.; Pecenco, S.; Tramontana, M.; Maio, V.; et al. Atypical Spitz tumours: An epidemiological, clinical and dermoscopic multicentre study with 16 years of follow-up. Clin. Exp. Dermatol. 2022, 47, 1464–1471. [Google Scholar] [CrossRef] [PubMed]
  6. Dika, E.; Chessa, M.A.; Veronesi, G.; Ravaioli, G.M.; Fanti, P.A.; Ribero, S.; Tripepi, G.; Gurioli, C.; Traniello Gradassi, A.; Lambertini, M.; et al. A single institute’s experience on melanoma prognosis: A long-term follow-up. G. Ital. Dermatol. Venereol. 2018, 153, 326–332. [Google Scholar] [CrossRef]
  7. Siegel, R.L.; Kratzer, T.B.; Giaquinto, A.N.; Sung, H.; Jemal, A. Cancer statistics, 2025. CA Cancer J. Clin. 2025, 75, 10–45. [Google Scholar] [CrossRef]
  8. De Giorgi, V.; Magnaterra, E.; Zuccaro, B.; Magi, S.; Magliulo, M.; Medri, M.; Mazzoni, L.; Venturi, F.; Silvestri, F.; Tomassini, G.M.; et al. Is Pediatric Melanoma Really That Different from Adult Melanoma? A Multicenter Epidemiological, Clinical and Dermoscopic Study. Cancers 2023, 15, 1835. [Google Scholar] [CrossRef]
  9. Broseghini, E.; Veronesi, G.; Gardini, A.; Venturi, F.; Scotti, B.; Vespi, L.; Marchese, P.V.; Melotti, B.; Comito, F.; Corti, B.; et al. Defining high-risk patients: Beyond the 8the AJCC melanoma staging system. Arch. Dermatol. Res. 2024, 317, 78. [Google Scholar] [CrossRef]
  10. Venturi, F.; Magnaterra, E.; Scotti, B.; Ferracin, M.; Dika, E. Predictive Factors for Sentinel Lymph Node Positivity in Melanoma Patients-The Role of Liquid Biopsy, MicroRNA and Gene Expression Profile Panels. Cancers 2025, 17, 1281. [Google Scholar] [CrossRef]
  11. Dika, E.; Veronesi, G.; Altimari, A.; Riefolo, M.; Ravaioli, G.M.; Piraccini, B.M.; Lambertini, M.; Campione, E.; Gruppioni, E.; Fiorentino, M.; et al. BRAF, KIT, and NRAS Mutations of Acral Melanoma in White Patients. Am. J. Clin. Pathol. 2020, 153, 664–671. [Google Scholar] [CrossRef]
  12. Amaral, T.; Ottaviano, M.; Arance, A.; Blank, C.; Chiarion-Sileni, V.; Donia, M.; Dummer, R.; Garbe, C.; Gershenwald, J.E.; Gogas, H.; et al. Cutaneous melanoma: ESMO Clinical Practice Guideline for diagnosis, treatment and follow-up. Ann. Oncol. 2025, 36, 10–30. [Google Scholar] [CrossRef]
  13. Garbe, C.; Amaral, T.; Peris, K.; Hauschild, A.; Arenberger, P.; Basset-Seguin, N.; Bastholt, L.; Bataille, V.; Del Marmol, V.; Dréno, B.; et al. European consensus-based interdisciplinary guideline for melanoma. Part 2: Treatment—Update 2022. Eur. J. Cancer 2022, 170, 256–284. [Google Scholar] [CrossRef] [PubMed]
  14. Scarfì, F.; Patrizi, A.; Veronesi, G.; Lambertini, M.; Tartari, F.; Mussi, M.; Melotti, B.; Dika, E. The role of topical imiquimod in melanoma cutaneous metastases: A critical review of the literature. Dermatol. Ther. 2020, 33, e14165. [Google Scholar] [CrossRef] [PubMed]
  15. Gogas, H.; Eggermont, A.M.M.; Hauschild, A.; Hersey, P.; Mohr, P.; Schadendorf, D.; Spatz, A.; Dummer, R. Biomarkers in melanoma. Ann. Oncol. 2009, 20 (Suppl. S6), vi8–vi13. [Google Scholar] [CrossRef] [PubMed]
  16. Lozzi, F.; Di Raimondo, C.; Lanna, C.; Diluvio, L.; Mazzilli, S.; Garofalo, V.; Dika, E.; Dellambra, E.; Coniglione, F.; Bianchi, L.; et al. Latest Evidence Regarding the Effects of Photosensitive Drugs on the Skin: Pathogenetic Mechanisms and Clinical Manifestations. Pharmaceutics 2020, 12, 1104. [Google Scholar] [CrossRef]
  17. Indini, A.; Di Guardo, L.; Cimminiello, C.; Prisciandaro, M.; Randon, G.; De Braud, F.; Del Vecchio, M. Immune-related adverse events correlate with improved survival in patients undergoing anti-PD1 immunotherapy for metastatic melanoma. J. Cancer Res. Clin. Oncol. 2019, 145, 511–521. [Google Scholar] [CrossRef]
  18. Wu, C.-E.; Yang, C.-K.; Peng, M.-T.; Huang, P.-W.; Chang, C.-F.; Yeh, K.-Y.; Chen, C.-B.; Wang, C.-L.; Hsu, C.-W.; Chen, I.-W.; et al. The association between immune-related adverse events and survival outcomes in Asian patients with advanced melanoma receiving anti-PD-1 antibodies. BMC Cancer 2020, 20, 1018. [Google Scholar] [CrossRef] [PubMed]
  19. Larkin, J.; Minor, D.; D’Angelo, S.; Neyns, B.; Smylie, M.; Miller, W.H.; Gutzmer, R.; Linette, G.; Chmielowski, B.; Lao, C.D.; et al. Overall Survival in Patients with Advanced Melanoma Who Received Nivolumab Versus Investigator’s Choice Chemotherapy in CheckMate 037: A Randomized, Controlled, Open-Label Phase III Trial. J. Clin. Oncol. 2018, 36, 383–390. [Google Scholar] [CrossRef]
  20. Hussaini, S.; Chehade, R.; Boldt, R.G.; Raphael, J.; Blanchette, P.; Maleki Vareki, S.; Fernandes, R. Association between immune-related side effects and efficacy and benefit of immune checkpoint inhibitors—A systematic review and meta-analysis. Cancer Treat. Rev. 2021, 92, 102134. [Google Scholar] [CrossRef]
  21. Quandt, Z.; Lucas, A.; Liang, S.I.; Yang, E.; Stone, S.; Fadlullah, M.Z.H.; Bayless, N.L.; Marr, S.S.; Thompson, M.A.; Padron, L.J.; et al. Associations between immune checkpoint inhibitor response, immune-related adverse events, and steroid use in RADIOHEAD: A prospective pan-tumor cohort study. J. Immunother. Cancer 2025, 13, e011545. [Google Scholar] [CrossRef] [PubMed]
  22. Dika, E.; Patrizi, A.; Ribero, S.; Fanti, P.A.; Starace, M.; Melotti, B.; Sperandi, F.; Piraccini, B.M. Hair and nail adverse events during treatment with targeted therapies for metastatic melanoma. Eur. J. Dermatol. 2016, 26, 232–239. [Google Scholar] [CrossRef]
  23. Venturi, F.; Veronesi, G.; Scotti, B.; Dika, E. Cutaneous Toxicities of Advanced Treatment for Cutaneous Melanoma: A Prospective Study from a Single-Center Institution. Cancers 2024, 16, 3679. [Google Scholar] [CrossRef]
  24. Burzi, L.; Alessandrini, A.M.; Quaglino, P.; Piraccini, B.M.; Dika, E.; Ribero, S. Cutaneous Events Associated with Immunotherapy of Melanoma: A Review. J. Clin. Med. 2021, 10, 3047. [Google Scholar] [CrossRef]
  25. Scarfì, F.; Melotti, B.; Veronesi, G.; Ravaioli, G.M.; Baraldi, C.; Lambertini, M.; Patrizi, A.; Dika, E. Sweet syndrome in metastatic melanoma during treatment with dabrafenib and trametinib. Australas. J. Dermatol. 2019, 60, e242–e243. [Google Scholar] [CrossRef]
  26. Dika, E.; Lambertini, M.; Gouveia, B.; Mussi, M.; Marcelli, E.; Campione, E.; Gurioli, C.; Melotti, B.; Alessandrini, A.; Ribero, S. Oral Manifestations in Melanoma Patients Treated with Target or Immunomodulatory Therapies. J. Clin. Med. 2021, 10, 1283. [Google Scholar] [CrossRef]
  27. De Giorgi, V.; Colombo, J.; Trane, L.; Silvestri, F.; Venturi, F.; Zuccaro, B.; Doni, L.; Stanganelli, I.; Covarelli, P. Cutaneous immune-related adverse events and photodamaged skin in patients with metastatic melanoma: Could nicotinamide be useful? Clin. Exp. Dermatol. 2022, 47, 1558–1560. [Google Scholar] [CrossRef]
  28. Nakamura, Y.; Tanaka, R.; Asami, Y.; Teramoto, Y.; Imamura, T.; Sato, S.; Maruyama, H.; Fujisawa, Y.; Matsuya, T.; Fujimoto, M.; et al. Correlation between vitiligo occurrence and clinical benefit in advanced melanoma patients treated with nivolumab: A multi-institutional retrospective study. J. Dermatol. 2017, 44, 117–122. [Google Scholar] [CrossRef]
  29. Teulings, H.-E.; Limpens, J.; Jansen, S.N.; Zwinderman, A.H.; Reitsma, J.B.; Spuls, P.I.; Luiten, R.M. Vitiligo-like depigmentation in patients with stage III-IV melanoma receiving immunotherapy and its association with survival: A systematic review and meta-analysis. J. Clin. Oncol. 2015, 33, 773–781. [Google Scholar] [CrossRef] [PubMed]
  30. Matsuya, T.; Nakamura, Y.; Matsushita, S.; Tanaka, R.; Teramoto, Y.; Asami, Y.; Uehara, J.; Aoki, M.; Yamamura, K.; Nakamura, Y.; et al. Vitiligo expansion and extent correlate with durable response in anti-programmed death 1 antibody treatment for advanced melanoma: A multi-institutional retrospective study. J. Dermatol. 2020, 47, 629–635. [Google Scholar] [CrossRef] [PubMed]
  31. Faje, A.T.; Sullivan, R.; Lawrence, D.; Tritos, N.A.; Fadden, R.; Klibanski, A.; Nachtigall, L. Ipilimumab-induced hypophysitis: A detailed longitudinal analysis in a large cohort of patients with metastatic melanoma. J. Clin. Endocrinol. Metab. 2014, 99, 4078–4085. [Google Scholar] [CrossRef] [PubMed]
  32. Mao, L.; Ding, Y.; Bai, X.; Sheng, X.; Dai, J.; Chi, Z.; Cui, C.; Kong, Y.; Fan, Y.; Xu, Y.; et al. Overall Survival of Patients With Unresectable or Metastatic BRAF V600-Mutant Acral/Cutaneous Melanoma Administered Dabrafenib Plus Trametinib: Long-Term Follow-Up of a Multicenter, Single-Arm Phase IIa Trial. Front. Oncol. 2021, 11, 720044. [Google Scholar] [CrossRef]
  33. Bai, X.; Dai, J.; Li, C.; Cui, C.; Mao, L.; Wei, X.; Sheng, X.; Chi, Z.; Yan, X.; Tang, B.; et al. Risk Models for Advanced Melanoma Patients Under Anti-PD-1 Monotherapy-Ad hoc Analyses of Pooled Data from Two Clinical Trials. Front. Oncol. 2021, 11, 639085. [Google Scholar] [CrossRef]
  34. Chamorro-Pareja, N.; Faje, A.T.; Miller, K.K. The Risk of Adrenal Insufficiency after Treatment with Relatlimab in Combination with Nivolumab is Higher than Expected. J. Clin. Endocrinol. Metab. 2025, 110, e3827–e3831. [Google Scholar] [CrossRef]
  35. Rozeman, E.A.; Hoefsmit, E.P.; Reijers, I.L.M.; Saw, R.P.M.; Versluis, J.M.; Krijgsman, O.; Dimitriadis, P.; Sikorska, K.; van de Wiel, B.A.; Eriksson, H.; et al. Survival and biomarker analyses from the OpACIN-neo and OpACIN neoadjuvant immunotherapy trials in stage III melanoma. Nat. Med. 2021, 27, 256–263. [Google Scholar] [CrossRef]
  36. Das, S.; Johnson, D.B. Immune-related adverse events and anti-tumor efficacy of immune checkpoint inhibitors. J. Immunother. Cancer 2019, 7, 306. [Google Scholar] [CrossRef]
  37. Sivan, A.; Corrales, L.; Hubert, N.; Williams, J.B.; Aquino-Michaels, K.; Earley, Z.M.; Benyamin, F.W.; Lei, Y.M.; Jabri, B.; Alegre, M.-L.; et al. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science 2015, 350, 1084–1089. [Google Scholar] [CrossRef]
  38. Fan, Y.; Xie, W.; Huang, H.; Wang, Y.; Li, G.; Geng, Y.; Hao, Y.; Zhang, Z. Association of Immune Related Adverse Events with Efficacy of Immune Checkpoint Inhibitors and Overall Survival in Cancers: A Systemic Review and Meta-analysis. Front. Oncol. 2021, 11, 633032. [Google Scholar] [CrossRef] [PubMed]
  39. Lima Ferreira, J.; Costa, C.; Marques, B.; Castro, S.; Victor, M.; Oliveira, J.; Santos, A.P.; Sampaio, I.L.; Duarte, H.; Marques, A.P.; et al. Improved survival in patients with thyroid function test abnormalities secondary to immune-checkpoint inhibitors. Cancer Immunol. Immunother. CII 2021, 70, 299–309. [Google Scholar] [CrossRef] [PubMed]
  40. Menzies, A.M.; Ashworth, M.T.; Swann, S.; Kefford, R.F.; Flaherty, K.; Weber, J.; Infante, J.R.; Kim, K.B.; Gonzalez, R.; Hamid, O.; et al. Characteristics of pyrexia in BRAFV600E/K metastatic melanoma patients treated with combined dabrafenib and trametinib in a phase I/II clinical trial. Ann. Oncol. 2015, 26, 415–421. [Google Scholar] [CrossRef] [PubMed]
  41. Schadendorf, D.; Robert, C.; Dummer, R.; Flaherty, K.T.; Tawbi, H.A.; Menzies, A.M.; Banerjee, H.; Lau, M.; Long, G.V. Pyrexia in patients treated with dabrafenib plus trametinib across clinical trials in BRAF-mutant cancers. Eur. J. Cancer Oxf. Engl. 2021, 153, 234–241. [Google Scholar] [CrossRef]
  42. Garbe, C.; Keim, U.; Suciu, S.; Amaral, T.; Eigentler, T.K.; Gesierich, A.; Hauschild, A.; Heinzerling, L.; Kiecker, F.; Schadendorf, D.; et al. Prognosis of Patients With Stage III Melanoma According to American Joint Committee on Cancer Version 8: A Reassessment on the Basis of 3 Independent Stage III Melanoma Cohorts. J. Clin. Oncol. 2020, 38, 2543–2551. [Google Scholar] [CrossRef] [PubMed]
  43. Dummer, R.; Ascierto, P.A.; Gogas, H.J.; Arance, A.; Mandala, M.; Liszkay, G.; Garbe, C.; Schadendorf, D.; Krajsova, I.; Gutzmer, R.; et al. Encorafenib plus binimetinib versus vemurafenib or encorafenib in patients with BRAF-mutant melanoma (COLUMBUS): A multicentre, open-label, randomised phase 3 trial. Lancet Oncol. 2018, 19, 603–615. [Google Scholar] [CrossRef]
  44. Heinzerling, L.; Eigentler, T.K.; Fluck, M.; Hassel, J.C.; Heller-Schenck, D.; Leipe, J.; Pauschinger, M.; Vogel, A.; Zimmer, L.; Gutzmer, R. Tolerability of BRAF/MEK inhibitor combinations: Adverse event evaluation and management. ESMO Open 2019, 4, e000491. [Google Scholar] [CrossRef]
  45. Schadendorf, D.; Dummer, R.; Flaherty, K.T.; Robert, C.; Arance, A.; de Groot, J.W.B.; Garbe, C.; Gogas, H.J.; Gutzmer, R.; Krajsová, I.; et al. COLUMBUS 7-year update: A randomized, open-label, phase III trial of encorafenib plus binimetinib versus vemurafenib or encorafenib in patients with BRAF V600E/K-mutant melanoma. Eur. J. Cancer. 2024, 204, 114073. [Google Scholar] [CrossRef]
  46. Gogas, H.J.; Flaherty, K.T.; Dummer, R.; Ascierto, P.A.; Arance, A.; Mandala, M.; Liszkay, G.; Garbe, C.; Schadendorf, D.; Krajsova, I.; et al. Adverse events associated with encorafenib plus binimetinib in the COLUMBUS study: Incidence, course and management. Eur. J. Cancer. 2019, 119, 97–106. [Google Scholar] [CrossRef]
  47. Atkinson, V.; Robert, C.; Grob, J.J.; Gogas, H.; Dutriaux, C.; Demidov, L.; Gupta, A.; Menzies, A.M.; Ryll, B.; Miranda, F.; et al. Improved pyrexia-related outcomes associated with an adapted pyrexia adverse event management algorithm in patients treated with adjuvant dabrafenib plus trametinib: Primary results of COMBI-APlus. Eur. J. Cancer. 2022, 163, 79–87. [Google Scholar] [CrossRef]
  48. Schaefer, H.; Rübben, A.; Esser, A.; Araujo, A.; Persa, O.-D.; Leijs, M. A distinct four-value blood signature of pyrexia under combination therapy of malignant melanoma with dabrafenib and trametinib evidenced by an algorithm-defined pyrexia score. PLoS ONE 2022, 17, e0273478. [Google Scholar] [CrossRef]
  49. De Giorgi, V.; Scarfì, F.; Silvestri, F.; Maida, P.; Venturi, F.; Trane, L.; Gori, A. Genital piercing: A warning for the risk of vulvar lichen sclerosus. Dermatol. Ther. 2021, 34, e14703. [Google Scholar] [CrossRef] [PubMed]
  50. Dummer, R.; Tsao, H.; Robert, C. How cutaneous eruptions help to understand the mode of action of kinase inhibitors. Br. J. Dermatol. 2012, 167, 965–967. [Google Scholar] [CrossRef] [PubMed]
  51. Abdullah, S.E.; Haigentz, M.; Piperdi, B. Dermatologic Toxicities from Monoclonal Antibodies and Tyrosine Kinase Inhibitors against EGFR: Pathophysiology and Management. Chemother. Res. Pract. 2012, 2012, 351210. [Google Scholar] [CrossRef]
  52. Lacouture, M.E.; Duvic, M.; Hauschild, A.; Prieto, V.G.; Robert, C.; Schadendorf, D.; Kim, C.C.; McCormack, C.J.; Myskowski, P.L.; Spleiss, O.; et al. Analysis of dermatologic events in vemurafenib-treated patients with melanoma. Oncologist 2013, 18, 314–322. [Google Scholar] [CrossRef] [PubMed]
  53. Vanneste, L.; Wolter, P.; Van den Oord, J.J.; Stas, M.; Garmyn, M. Cutaneous adverse effects of BRAF inhibitors in metastatic malignant melanoma, a prospective study in 20 patients. J. Eur. Acad. Dermatol. Venereol. 2015, 29, 61–68. [Google Scholar] [CrossRef] [PubMed]
  54. Chen, P.; Chen, F.; Zhou, B. Systematic review and meta-analysis of prevalence of dermatological toxicities associated with vemurafenib treatment in patients with melanoma. Clin. Exp. Dermatol. 2019, 44, 243–251. [Google Scholar] [CrossRef]
  55. Robert, C.; Karaszewska, B.; Schachter, J.; Rutkowski, P.; Mackiewicz, A.; Stroiakovski, D.; Lichinitser, M.; Dummer, R.; Grange, F.; Mortier, L.; et al. Improved overall survival in melanoma with combined dabrafenib and trametinib. N. Engl. J. Med. 2015, 372, 30–39. [Google Scholar] [CrossRef] [PubMed]
  56. Lacouture, M.E.; Mitchell, E.P.; Piperdi, B.; Pillai, M.V.; Shearer, H.; Iannotti, N.; Xu, F.; Yassine, M. Skin toxicity evaluation protocol with panitumumab (STEPP), a phase II, open-label, randomized trial evaluating the impact of a pre-Emptive Skin treatment regimen on skin toxicities and quality of life in patients with metastatic colorectal cancer. J. Clin. Oncol. 2010, 28, 1351–1357. [Google Scholar] [CrossRef]
  57. Várvölgyi, T.; Janka, E.A.; Szász, I.; Koroknai, V.; Toka-Farkas, T.; Szabó, I.L.; Ványai, B.; Szegedi, A.; Emri, G.; Balázs, M. Combining Biomarkers for the Diagnosis of Metastatic Melanoma. J. Clin. Med. 2024, 13, 174. [Google Scholar] [CrossRef]
  58. Martens, A.; Wistuba-Hamprecht, K.; Foppen, M.G.; Yuan, J.; Postow, M.A.; Wong, P.; Romano, E.; Khammari, A.; Dreno, B.; Capone, M.; et al. Baseline peripheral blood biomarkers associated with clinical outcome of advanced melanoma patients treated with ipilimumab. Clin. Cancer Res. 2016, 22, 2908–2918. [Google Scholar] [CrossRef]
  59. Palmer, S.R.; Erickson, L.A.; Ichetovkin, I.; Knauer, D.J.; Markovic, S.N. Circulating Serologic and Molecular Biomarkers in Malignant Melanoma. Mayo Clin. Proc. 2011, 86, 981–990. [Google Scholar] [CrossRef]
  60. 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]
  61. van Wilpe, S.; Tolmeijer, S.H.; de Vries, I.J.M.; Koornstra, R.H.T.; Mehra, N. LDH Isotyping for Checkpoint Inhibitor Response Prediction in Patients with Metastatic Melanoma. Immuno 2021, 1, 67–77. [Google Scholar] [CrossRef]
  62. Liang, X.; Zhou, S.; Xiao, Z. Prognostic value of lactate dehydrogenase in patients with uveal melanoma treated with immune checkpoint inhibition. Aging 2023, 15, 8770–8781. [Google Scholar] [CrossRef] [PubMed]
  63. Jurisic, V.; Radenkovic, S.; Konjevic, G. The Actual Role of LDH as Tumor Marker, Biochemical and Clinical Aspects. Adv. Exp. Med. Biol. 2015, 867, 115–124. [Google Scholar] [CrossRef]
  64. Nikolin, B.; Djan, I.; Trifunovic, J.; Dugandzija, T.; Novkovic, D.; Djan, V.; Vucinic, N. MIA, S100 and LDH as important predictors of overall survival of patients with stage IIb and IIc melanoma. J. BUON Off. J. Balk. Union. Oncol. 2016, 21, 691–697. [Google Scholar]
  65. Gassenmaier, M.; Lenders, M.M.; Forschner, A.; Leiter, U.; Weide, B.; Garbe, C.; Eigentler, T.K.; Wagner, N.B. Serum S100B and LDH at Baseline and During Therapy Predict the Outcome of Metastatic Melanoma Patients Treated with BRAF Inhibitors. Target. Oncol. 2021, 16, 197–205. [Google Scholar] [CrossRef]
  66. 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]
  67. De Giorgi, V.; Maida, P.; Salvati, L.; Scarfì, F.; Trane, L.; Gori, A.; Silvestri, F.; Venturi, F.; Covarelli, P. Trauma and foreign bodies may favour the onset of melanoma metastases. Clin. Exp. Dermatol. 2020, 45, 619–621. [Google Scholar] [CrossRef]
  68. Kaskel, P.; Berking, C.; Sander, S.; Volkenandt, M.; Peter, R.U.; Krähn, G. S-100 protein in peripheral blood: A marker for melanoma metastases: A prospective 2-center study of 570 patients with melanoma. J. Am. Acad. Dermatol. 1999, 41, 962–969. [Google Scholar] [CrossRef]
  69. Hauschild, A.; Engel, G.; Brenner, W.; Gläser, R.; Mönig, H.; Henze, E.; Christophers, E. S100B protein detection in serum is a significant prognostic factor in metastatic melanoma. Oncology 1999, 56, 338–344. [Google Scholar] [CrossRef] [PubMed]
  70. Lipson, E.J.; Velculescu, V.E.; Pritchard, T.S.; Sausen, M.; Pardoll, D.M.; Topalian, S.L.; Diaz, L.A. Circulating tumor DNA analysis as a real-time method for monitoring tumor burden in melanoma patients undergoing treatment with immune checkpoint blockade. J. Immunother. Cancer 2014, 2, 42. [Google Scholar] [CrossRef] [PubMed]
  71. Maloberti, T.; De Leo, A.; Coluccelli, S.; Sanza, V.; Gruppioni, E.; Altimari, A.; Comito, F.; Melotti, B.; Marchese, P.V.; Dika, E.; et al. Molecular Characterization of Advanced-Stage Melanomas in Clinical Practice Using a Laboratory-Developed Next-Generation Sequencing Panel. Diagnostics 2024, 14, 800. [Google Scholar] [CrossRef]
  72. Wen, X.; Pu, H.; Liu, Q.; Guo, Z.; Luo, D. Circulating Tumor DNA—A Novel Biomarker of Tumor Progression and Its Favorable Detection Techniques. Cancers 2022, 14, 6025. [Google Scholar] [CrossRef] [PubMed]
  73. 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] [PubMed]
  74. Marsavela, G.; McEvoy, A.C.; Pereira, M.R.; Reid, A.L.; Al-Ogaili, Z.; Warburton, L.; Khattak, M.A.; Abed, A.; Meniawy, T.M.; Millward, M.; et al. Detection of clinical progression through plasma ctDNA in metastatic melanoma patients: A comparison to radiological progression. Br. J. Cancer 2022, 126, 401–408. [Google Scholar] [CrossRef]
  75. Di Nardo, L.; Del Regno, L.; Di Stefani, A.; Mannino, M.; Fossati, B.; Catapano, S.; Quattrini, L.; Pellegrini, C.; Cortellini, A.; Parisi, A.; et al. The dynamics of circulating tumour DNA (ctDNA) during treatment reflects tumour response in advanced melanoma patients. Exp. Dermatol. 2023, 32, 1785–1793. [Google Scholar] [CrossRef] [PubMed]
  76. Lee, J.H.; Long, G.V.; Boyd, S.; Lo, S.; Menzies, A.M.; Tembe, V.; Guminski, A.; Jakrot, V.; Scolyer, R.A.; Mann, G.J.; et al. Circulating tumour DNA predicts response to anti-PD1 antibodies in metastatic melanoma. Ann. Oncol. 2017, 28, 1130–1136. [Google Scholar] [CrossRef]
  77. Seremet, T.; Jansen, Y.; Planken, S.; Njimi, H.; Delaunoy, M.; El Housni, H.; Awada, G.; Schwarze, J.K.; Keyaerts, M.; Everaert, H.; et al. Undetectable circulating tumor DNA (ctDNA) levels correlate with favorable outcome in metastatic melanoma patients treated with anti-PD1 therapy. J. Transl. Med. 2019, 17, 303. [Google Scholar] [CrossRef]
  78. Syeda, M.M.; Long, G.V.; Garrett, J.; Atkinson, V.; Santinami, M.; Schadendorf, D.; Hauschild, A.; Millward, M.; Mandala, M.; Chiarion-Sileni, V.; et al. Clinical validation of droplet digital PCR assays in detecting BRAFV600-mutant circulating tumour DNA as a prognostic biomarker in patients with resected stage III melanoma receiving adjuvant therapy (COMBI-AD): A biomarker analysis from a double-blind, randomised phase 3 trial. Lancet Oncol. 2025, 26, 641–653. [Google Scholar] [CrossRef]
  79. Warburton, L.; Meniawy, T.M.; Calapre, L.; Pereira, M.; McEvoy, A.; Ziman, M.; Gray, E.S.; Millward, M. Stopping targeted therapy for complete responders in advanced BRAF mutant melanoma. Sci. Rep. 2020, 10, 18878. [Google Scholar] [CrossRef]
  80. McEvoy, A.C.; Pereira, M.R.; Reid, A.; Pearce, R.; Cowell, L.; Al-Ogaili, Z.; Khattak, M.A.; Millward, M.; Meniawy, T.M.; Gray, E.S.; et al. Monitoring melanoma recurrence with circulating tumor DNA: A proof of concept from three case studies. Oncotarget 2019, 10, 113–122. [Google Scholar] [CrossRef]
  81. Diefenbach, R.J.; Lee, J.H.; Rizos, H. Monitoring Melanoma Using Circulating Free DNA. Am. J. Clin. Dermatol. 2019, 20, 1–12. [Google Scholar] [CrossRef] [PubMed]
  82. Broseghini, E.; Venturi, F.; Veronesi, G.; Scotti, B.; Migliori, M.; Marini, D.; Ricci, C.; Casadei, R.; Ferracin, M.; Dika, E. Exploring the Common Mutational Landscape in Cutaneous Melanoma and Pancreatic Cancer. Pigment. Cell Melanoma Res. 2025, 38, e13210. [Google Scholar] [CrossRef] [PubMed]
  83. van der Leest, P.; Schuuring, E. Critical Factors in the Analytical Work Flow of Circulating Tumor DNA-Based Molecular Profiling. Clin. Chem. 2024, 70, 220–233. [Google Scholar] [CrossRef] [PubMed]
  84. Ntzifa, A.; Lianidou, E. Pre-analytical conditions and implementation of quality control steps in liquid biopsy analysis. Crit. Rev. Clin. Lab. Sci. 2023, 60, 573–594. [Google Scholar] [CrossRef] [PubMed]
  85. Lockwood, C.M.; Borsu, L.; Cankovic, M.; Earle, J.S.L.; Gocke, C.D.; Hameed, M.; Jordan, D.; Lopategui, J.R.; Pullambhatla, M.; Reuther, J.; et al. Recommendations for Cell-Free DNA Assay Validations: A Joint Consensus Recommendation of the Association for Molecular Pathology and College of American Pathologists. J. Mol. Diagn. 2023, 25, 876–897. [Google Scholar] [CrossRef]
  86. Broseghini, E.; Dika, E.; Londin, E.; Ferracin, M. MicroRNA Isoforms Contribution to Melanoma Pathogenesis. Non-Coding RNA 2021, 7, 63. [Google Scholar] [CrossRef]
  87. Naddeo, M.; Broseghini, E.; Venturi, F.; Vaccari, S.; Corti, B.; Lambertini, M.; Ricci, C.; Fontana, B.; Durante, G.; Pariali, M.; et al. Association of miR-146a-5p and miR-21-5p with Prognostic Features in Melanomas. Cancers 2024, 16, 1688. [Google Scholar] [CrossRef]
  88. Dika, E.; Riefolo, M.; Porcellini, E.; Broseghini, E.; Ribero, S.; Senetta, R.; Osella-Abate, S.; Scarfì, F.; Lambertini, M.; Veronesi, G.; et al. Defining the Prognostic Role of MicroRNAs in Cutaneous Melanoma. J. Investig. Dermatol. 2020, 140, 2260–2267. [Google Scholar] [CrossRef]
  89. Durante, G.; Broseghini, E.; Comito, F.; Naddeo, M.; Milani, M.; Salamon, I.; Campione, E.; Dika, E.; Ferracin, M. Circulating microRNA biomarkers in melanoma and non-melanoma skin cancer. Expert Rev. Mol. Diagn. 2022, 22, 305–318. [Google Scholar] [CrossRef]
  90. Durante, G.; Comito, F.; Lambertini, M.; Broseghini, E.; Dika, E.; Ferracin, M. Non-coding RNA dysregulation in skin cancers. Essays Biochem. 2021, 65, 641–655. [Google Scholar] [CrossRef]
  91. Dika, E.; Broseghini, E.; Porcellini, E.; Lambertini, M.; Riefolo, M.; Durante, G.; Loher, P.; Roncarati, R.; Bassi, C.; Misciali, C.; et al. Unraveling the role of microRNA/isomiR network in multiple primary melanoma pathogenesis. Cell Death Dis. 2021, 12, 473. [Google Scholar] [CrossRef] [PubMed]
  92. Dong, L.; Tian, X.; Zhao, Y.; Tu, H.; Wong, A.; Yang, Y. The Roles of MiRNAs (MicroRNAs) in Melanoma Immunotherapy. Int. J. Mol. Sci. 2022, 23, 14775. [Google Scholar] [CrossRef]
  93. Fattore, L.; Costantini, S.; Malpicci, D.; Ruggiero, C.F.; Ascierto, P.A.; Croce, C.M.; Mancini, R.; Ciliberto, G. MicroRNAs in melanoma development and resistance to target therapy. Oncotarget 2017, 8, 22262–22278. [Google Scholar] [CrossRef]
  94. García-Giménez, J.L.; Saadi, W.; Ortega, A.L.; Lahoz, A.; Suay, G.; Carretero, J.; Pereda, J.; Fatmi, A.; Pallardó, F.V.; Mena-Molla, S. miRNAs Related to Immune Checkpoint Inhibitor Response: A Systematic Review. Int. J. Mol. Sci. 2024, 25, 1737. [Google Scholar] [CrossRef]
  95. Sánchez-Sendra, B.; González-Muñoz, J.F.; Pérez-Debén, S.; Monteagudo, C. The Prognostic Value of miR-125b, miR-200c and miR-205 in Primary Cutaneous Malignant Melanoma Is Independent of BRAF Mutational Status. Cancers 2022, 14, 1532. [Google Scholar] [CrossRef] [PubMed]
  96. Ye, Q.; Li, Z.; Li, Y.; Li, Y.; Zhang, Y.; Gui, R.; Cui, Y.; Zhang, Q.; Qian, L.; Xiong, Y.; et al. Exosome-Derived microRNA: Implications in Melanoma Progression, Diagnosis and Treatment. Cancers 2022, 15, 80. [Google Scholar] [CrossRef] [PubMed]
  97. De Martino, E.; Gandin, I.; Azzalini, E.; Massone, C.; Pizzichetta, M.A.; Giulioni, E.; Javor, S.; Pinzani, C.; Conforti, C.; Zalaudek, I.; et al. A group of three miRNAs can act as candidate circulating biomarkers in liquid biopsies from melanoma patients. Front. Med. 2023, 10, 1180799. [Google Scholar] [CrossRef]
  98. Cristescu, R.; Mogg, R.; Ayers, M.; Albright, A.; Murphy, E.; Yearley, J.; Sher, X.; Liu, X.Q.; Lu, H.; Nebozhyn, M.; et al. Pan-tumor genomic biomarkers for PD-1 checkpoint blockade–based immunotherapy. Science 2018, 362, eaar3593. [Google Scholar] [CrossRef]
  99. Ayers, M.; Lunceford, J.; Nebozhyn, M.; Murphy, E.; Loboda, A.; Kaufman, D.R.; Albright, A.; Cheng, J.D.; Kang, S.P.; Shankaran, V.; et al. IFN-γ-related mRNA profile predicts clinical response to PD-1 blockade. J. Clin. Investig. 2017, 127, 2930–2940. [Google Scholar] [CrossRef]
  100. Grasso, C.S.; Tsoi, J.; Onyshchenko, M.; Abril-Rodriguez, G.; Ross-Macdonald, P.; Wind-Rotolo, M.; Champhekar, A.; Medina, E.; Torrejon, D.Y.; Shin, D.S.; et al. Conserved Interferon-γ Signaling Drives Clinical Response to Immune Checkpoint Blockade Therapy in Melanoma. Cancer Cell 2020, 38, 500–515.e3. [Google Scholar] [CrossRef]
  101. Newell, F.; Pires da Silva, I.; Johansson, P.A.; Menzies, A.M.; Wilmott, J.S.; Addala, V.; Carlino, M.S.; Rizos, H.; Nones, K.; Edwards, J.J.; et al. Multiomic profiling of checkpoint inhibitor-treated melanoma: Identifying predictors of response and resistance, and markers of biological discordance. Cancer Cell 2022, 40, 88–102.e7. [Google Scholar] [CrossRef]
  102. Wawrzyniak, P.; Hartman, M.L. Dual role of interferon-gamma in the response of melanoma patients to immunotherapy with immune checkpoint inhibitors. Mol. Cancer 2025, 24, 89. [Google Scholar] [CrossRef]
  103. Brockwell, N.K.; Parker, B.S. Tumor inherent interferons: Impact on immune reactivity and immunotherapy. Cytokine 2019, 118, 42–47. [Google Scholar] [CrossRef]
  104. Versluis, J.M.; Blankenstein, S.A.; Dimitriadis, P.; Wilmott, J.S.; Elens, R.; Blokx, W.A.M.; van Houdt, W.; Menzies, A.M.; Schrage, Y.M.; Wouters, M.W.J.M.; et al. Interferon-gamma signature as prognostic and predictive marker in macroscopic stage III melanoma. J. Immunother. Cancer 2024, 12, e008125. [Google Scholar] [CrossRef]
  105. Blank, C.U.; Reijers, I.L.M.; Versluis, J.M.; Menzies, A.M.; Dimitriadis, P.; Wouters, M.W.; Saw, R.P.M.; Klop, W.M.C.; Pennington, T.E.; Bosch, L.J.W.; et al. LBA39 Personalized combination of neoadjuvant domatinostat, nivolumab (NIVO) and ipilimumab (IPI) in stage IIIB-D melanoma patients (pts) stratified according to the interferon-gamma signature (IFN-γ sign): The DONIMI study. Ann. Oncol. 2021, 32, S1315. [Google Scholar] [CrossRef]
  106. Aung, T.N.; Warrell, J.; Martinez-Morilla, S.; Gavrielatou, N.; Vathiotis, I.; Yaghoobi, V.; Kluger, H.M.; Gerstein, M.; Rimm, D.L. Spatially Informed Gene Signatures for Response to Immunotherapy in Melanoma. Clin. Cancer Res. 2024, 30, 3520–3532. [Google Scholar] [CrossRef]
  107. Deng, Z.; Liu, J.; Yu, Y.V.; Jin, Y.N. Machine learning-based identification of an immunotherapy-related signature to enhance outcomes and immunotherapy responses in melanoma. Front. Immunol. 2024, 15, 1451103. [Google Scholar] [CrossRef] [PubMed]
  108. Type I Interferon Signaling Induces Melanoma Cell-Intrinsic PD-1 and Its Inhibition Antagonizes Immune Checkpoint Blockade|Nature Communications. Available online: https://www.nature.com/articles/s41467-024-51496-2 (accessed on 13 October 2025).
  109. Zhai, H.; Dika, E.; Goldovsky, M.; Maibach, H.I. Tape-stripping method in man: Comparison of evaporimetric methods. Skin. Res. Technol. 2007, 13, 207–210. [Google Scholar] [CrossRef] [PubMed]
  110. Liu, D.; Schilling, B.; Liu, D.; Sucker, A.; Livingstone, E.; Jerby-Arnon, L.; Zimmer, L.; Gutzmer, R.; Satzger, I.; Loquai, C.; et al. Integrative molecular and clinical modeling of clinical outcomes to PD1 blockade in patients with metastatic melanoma. Nat. Med. 2019, 25, 1916–1927. [Google Scholar] [CrossRef]
  111. Gide, T.N.; Mao, Y.; Scolyer, R.A.; Long, G.V.; Wilmott, J.S. Tissue-Based Profiling Techniques to Achieve Precision Medicine in Cancer: Opportunities and Challenges in Melanoma. Clin. Cancer Res. 2024, 30, 5270–5280. [Google Scholar] [CrossRef] [PubMed]
  112. Molina-García, M.; Rojas-Lechuga, M.J.; Torres Moral, T.; Crespí-Payeras, F.; Bagué, J.; Mateu, J.; Paschalidis, N.; de Souza, V.G.; Podlipnik, S.; Carrera, C.; et al. Integrative Molecular and Immune Profiling in Advanced Unresectable Melanoma: Tumor Microenvironment and Peripheral PD-1+ CD4+ Effector Memory T-Cells as Potential Markers of Response to Immune Checkpoint Inhibitor Therapy. Cancers 2025, 17, 2022. [Google Scholar] [CrossRef]
  113. Splendiani, E.; Besharat, Z.M.; Covre, A.; Maio, M.; Di Giacomo, A.M.; Ferretti, E. Immunotherapy in melanoma: Can we predict response to treatment with circulating biomarkers? Pharmacol. Ther. 2024, 256, 108613. [Google Scholar] [CrossRef]
  114. De Giorgi, V.; Scarfì, F.; Gori, A.; Silvestri, F.; Trane, L.; Maida, P.; Venturi, F.; Covarelli, P. Short-term teledermoscopic monitoring of atypical melanocytic lesions in the early diagnosis of melanoma: Utility more apparent than real. J. Eur. Acad. Dermatol. Venereol. 2020, 34, e398–e399. [Google Scholar] [CrossRef]
  115. Cinotti, E.; Veronesi, G.; Labeille, B.; Cambazard, F.; Piraccini, B.M.; Dika, E.; Perrot, J.L.; Rubegni, P. Imaging technique for the diagnosis of onychomatricoma. J. Eur. Acad. Dermatol. Venereol. 2018, 32, 1874–1878. [Google Scholar] [CrossRef] [PubMed]
  116. De Giorgi, V.; Silvestri, F.; Cecchi, G.; Venturi, F.; Zuccaro, B.; Perillo, G.; Cosso, F.; Maio, V.; Simi, S.; Antonini, P.; et al. Dermoscopy as a Tool for Identifying Potentially Metastatic Thin Melanoma: A Clinical-Dermoscopic and Histopathological Case-Control Study. Cancers 2024, 16, 1394. [Google Scholar] [CrossRef]
  117. van Duin, I.A.J.; Verheijden, R.J.; van Diest, P.J.; Blokx, W.A.M.; El-Sharouni, M.-A.; Verhoeff, J.J.C.; Leiner, T.; van den Eertwegh, A.J.M.; de Groot, J.W.B.; van Not, O.J.; et al. A prediction model for response to immune checkpoint inhibition in advanced melanoma. Int. J. Cancer 2024, 154, 1760–1771. [Google Scholar] [CrossRef] [PubMed]
  118. Pires da Silva, I.; Ahmed, T.; McQuade, J.L.; Nebhan, C.A.; Park, J.J.; Versluis, J.M.; Serra-Bellver, P.; Khan, Y.; Slattery, T.; Oberoi, H.K.; et al. Clinical Models to Define Response and Survival with Anti-PD-1 Antibodies Alone or Combined With Ipilimumab in Metastatic Melanoma. J. Clin. Oncol. 2022, 40, 1068–1080. [Google Scholar] [CrossRef] [PubMed]
  119. 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]
  120. Abbott, C.W.; Boyle, S.M.; Pyke, R.M.; McDaniel, L.D.; Levy, E.; Navarro, F.C.P.; Mellacheruvu, D.; Zhang, S.V.; Tan, M.; Santiago, R.; et al. Prediction of Immunotherapy Response in Melanoma through Combined Modeling of Neoantigen Burden and Immune-Related Resistance Mechanisms. Clin. Cancer Res. 2021, 27, 4265–4276. [Google Scholar] [CrossRef]
  121. 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]
  122. Ahmed, Y.B.; Al-Bzour, A.N.; Ababneh, O.E.; Abushukair, H.M.; Saeed, A. Genomic and Transcriptomic Predictors of Response to Immune Checkpoint Inhibitors in Melanoma Patients: A Machine Learning Approach. Cancers 2022, 14, 5605. [Google Scholar] [CrossRef]
  123. Li, C.; Lin, X.; Wang, J.; Zhou, Q.; Feng, F.; Xu, J. Multi-omics-based subtyping of melanoma suggests distinct immune and targeted therapy strategies. Front. Immunol. 2025, 16, 1601243. [Google Scholar] [CrossRef]
  124. Tarhini, A.A.; Obermayer, A.; Lee, S.J.; LaFramboise, W.A.; Hodi, F.S.; Karunamurthy, A.D.; Eljilany, I.; Chen, D.-T.; Hwu, P.; El Naqa, I.M.; et al. Integrative Immune Signature of Complementary Circulating and Tumoral Biomarkers Maximizes the Predictive Power of Adjuvant Immunotherapeutic Benefits in High-risk Melanoma. Clin. Cancer Res. 2025, 31, 3249–3258. [Google Scholar] [CrossRef]
  125. Muir, C.A.; Clifton-Bligh, R.J.; Long, G.V.; Scolyer, R.A.; Lo, S.N.; Carlino, M.S.; Tsang, V.H.M.; Menzies, A.M. Thyroid Immune-related Adverse Events Following Immune Checkpoint Inhibitor Treatment. J. Clin. Endocrinol. Metab. 2021, 106, e3704–e3713. [Google Scholar] [CrossRef]
  126. Asher, N.; Bar-Hai, N.; Ben-Betzalel, G.; Stoff, R.; Grynberg, S.; Schachter, J.; Frommer-Shapira, R. Exploring the clinical significance of specific immune-related adverse events in melanoma patients undergoing immune checkpoint inhibitor therapy. Melanoma Res. 2024, 34, 439–449. [Google Scholar] [CrossRef] [PubMed]
  127. Tang, B.; Yu, Y.; Wan, J.; Yu, C.; Sun, Y.; Yu, X.; Liu, R.; Huang, H.; Du, Y.; Hu, W.; et al. Immune checkpoint inhibitor-induced vitiligo: A large-scale real world pharmacovigilance study. Int. J. Cancer 2025. [Google Scholar] [CrossRef] [PubMed]
  128. Geisler, A.N.; Phillips, G.S.; Barrios, D.M.; Wu, J.; Leung, D.Y.M.; Moy, A.P.; Kern, J.A.; Lacouture, M.E. Immune checkpoint inhibitor-related dermatologic adverse events. J. Am. Acad. Dermatol. 2020, 83, 1255–1268. [Google Scholar] [CrossRef] [PubMed]
  129. Wang, Y.; Zhou, S.; Yang, F.; Qi, X.; Wang, X.; Guan, X.; Shen, C.; Duma, N.; Vera Aguilera, J.; Chintakuntlawar, A.; et al. Treatment-Related Adverse Events of PD-1 and PD-L1 Inhibitors in Clinical Trials: A Systematic Review and Meta-analysis. JAMA Oncol. 2019, 5, 1008–1019. [Google Scholar] [CrossRef]
  130. Schneider, B.J.; Naidoo, J.; Santomasso, B.D.; Lacchetti, C.; Adkins, S.; Anadkat, M.; Atkins, M.B.; Brassil, K.J.; Caterino, J.M.; Chau, I.; et al. Management of Immune-Related Adverse Events in Patients Treated With Immune Checkpoint Inhibitor Therapy: ASCO Guideline Update. J. Clin. Oncol. 2021, 39, 4073–4126. [Google Scholar] [CrossRef]
  131. Malla, M.; Loree, J.M.; Kasi, P.M.; Parikh, A.R. Using Circulating Tumor DNA in Colorectal Cancer: Current and Evolving Practices. J. Clin. Oncol. 2022, 40, 2846–2857. [Google Scholar] [CrossRef]
  132. Chidharla, A.; Rapoport, E.; Agarwal, K.; Madala, S.; Linares, B.; Sun, W.; Chakrabarti, S.; Kasi, A. Circulating Tumor DNA as a Minimal Residual Disease Assessment and Recurrence Risk in Patients Undergoing Curative-Intent Resection with or without Adjuvant Chemotherapy in Colorectal Cancer: A Systematic Review and Meta-Analysis. Int. J. Mol. Sci. 2023, 24, 10230. [Google Scholar] [CrossRef]
  133. Eroglu, Z.; Krinshpun, S.; Kalashnikova, E.; Sudhaman, S.; Ozturk Topcu, T.; 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]
  134. Gouda, M.A.; Huang, H.J.; Piha-Paul, S.A.; Call, S.G.; Karp, D.D.; Fu, S.; Naing, A.; Subbiah, V.; Pant, S.; Dustin, D.J.; et al. Longitudinal Monitoring of Circulating Tumor DNA to Predict Treatment Outcomes in Advanced Cancers. JCO Precis. Oncol. 2022, 6, e2100512. [Google Scholar] [CrossRef]
  135. Dang, D.K.; Park, B.H. Circulating tumor DNA: Current challenges for clinical utility. J. Clin. Investig. 2022, 132, e154941. [Google Scholar] [CrossRef]
  136. Tang, W.W.; Battistone, B.; Bauer, K.M.; Weis, A.M.; Barba, C.; Fadlullah, M.Z.H.; Ghazaryan, A.; Tran, V.B.; Lee, S.-H.; Agir, Z.B.; et al. A microRNA-regulated transcriptional state defines intratumoral CD8+ T cells that respond to immunotherapy. Cell Rep. 2025, 44, 115301. [Google Scholar] [CrossRef] [PubMed]
  137. Lim, S.Y.; Boyd, S.C.; Diefenbach, R.J.; Rizos, H. Circulating MicroRNAs: Functional biomarkers for melanoma prognosis and treatment. Mol. Cancer 2025, 24, 99. [Google Scholar] [CrossRef] [PubMed]
  138. Liu, Z.; Lu, T.; Wang, Y.; Jiao, D.; Li, Z.; Wang, L.; Liu, L.; Guo, C.; Zhao, Y.; Han, X. Establishment and experimental validation of an immune miRNA signature for assessing prognosis and immune landscape of patients with colorectal cancer. J. Cell. Mol. Med. 2021, 25, 6874–6886. [Google Scholar] [CrossRef] [PubMed]
  139. Horowitch, B.; Lee, D.Y.; Ding, M.; Martinez-Morilla, S.; Aung, T.N.; Ouerghi, F.; Wang, X.; Wei, W.; Damsky, W.; Sznol, M.; et al. Subsets of IFN Signaling Predict Response to Immune Checkpoint Blockade in Patients with Melanoma. Clin. Cancer Res. 2023, 29, 2908–2918. [Google Scholar] [CrossRef] [PubMed]
  140. Reijers, I.L.M.; Rao, D.; Versluis, J.M.; Menzies, A.M.; Dimitriadis, P.; Wouters, M.W.; Spillane, A.J.; Klop, W.M.C.; Broeks, A.; Bosch, L.J.W.; et al. IFN-γ signature enables selection of neoadjuvant treatment in patients with stage III melanoma. J. Exp. Med. 2023, 220, e20221952. [Google Scholar] [CrossRef]
  141. Gibney, G.T.; Weiner, L.M.; Atkins, M.B. Predictive biomarkers for checkpoint inhibitor-based immunotherapy. Lancet Oncol. 2016, 17, e542–e551. [Google Scholar] [CrossRef]
Figure 1. Flow diagram summarizing the identification, screening, eligibility, and inclusion process for studies analyzed. A comprehensive search was conducted across PubMed, Scopus, and Web of Science from January 2010 to September 2025. A total of 897 records were initially identified, of which 587 remained after duplicate removal. Following title and abstract screening, 195 full-text articles were evaluated for eligibility. Studies were excluded if they were non-relevant, duplicates, preclinical, or lacked data on treatment-related adverse events, circulating biomarkers, or immune transcriptomic signatures. Finally, 97 studies meeting the inclusion criteria were included in the qualitative synthesis.
Figure 1. Flow diagram summarizing the identification, screening, eligibility, and inclusion process for studies analyzed. A comprehensive search was conducted across PubMed, Scopus, and Web of Science from January 2010 to September 2025. A total of 897 records were initially identified, of which 587 remained after duplicate removal. Following title and abstract screening, 195 full-text articles were evaluated for eligibility. Studies were excluded if they were non-relevant, duplicates, preclinical, or lacked data on treatment-related adverse events, circulating biomarkers, or immune transcriptomic signatures. Finally, 97 studies meeting the inclusion criteria were included in the qualitative synthesis.
Diagnostics 15 03054 g001
Figure 2. Representation of the immunologic spectrum in melanoma.
Figure 2. Representation of the immunologic spectrum in melanoma.
Diagnostics 15 03054 g002
Figure 3. Algorithm for Personalized Response Monitoring in Melanoma. This dynamic, multi-parametric algorithm integrates clinical toxicity patterns, liquid-biopsy biomarkers, and tumor immune signatures into a unified precision-response monitoring framework. It supports early, individualized therapeutic adaptation aimed at maximizing efficacy while minimizing toxicity.
Figure 3. Algorithm for Personalized Response Monitoring in Melanoma. This dynamic, multi-parametric algorithm integrates clinical toxicity patterns, liquid-biopsy biomarkers, and tumor immune signatures into a unified precision-response monitoring framework. It supports early, individualized therapeutic adaptation aimed at maximizing efficacy while minimizing toxicity.
Diagnostics 15 03054 g003
Table 1. Stepwise algorithm summarizing the proposed clinical workflow.
Table 1. Stepwise algorithm summarizing the proposed clinical workflow.
StepProcessKey Indicators/ToolsInterpretation/Clinical Action
Baseline profilingComprehensive clinical and molecular characterization before therapy initiation.LDH, S100B, baseline ctDNA, miRNA panel (miR-21-5p, miR-146a-5p), PD-L1 IHC, IFN-γ gene-expression profile (GEP), tumor mutational burden (TMB).Stratify tumors as “hot” (IFN-high, inflamed) or “cold” (immune-desert). Define initial risk and expected treatment sensitivity.
Early on-treatment assessment (weeks 4–8)Initial biological and clinical response evaluation.Onset and grade of TRAEs (irAEs or ttAEs), repeat ctDNA quantification, miRNA modulation, early imaging if available.Early ctDNA clearance ± low-grade irAEs favorable immune activation; persistent ctDNA ± no toxicity consider intensification or switch.
Dynamic monitoring (weeks 8–16)Continuous assessment of therapeutic efficacy and resistance emergence.Longitudinal ctDNA kinetics, serial miRNA profiles, cytokine panels, serial IFN-γ GEP measurements.Declining ctDNA + induced IFN signature sustained response; rising ctDNA + loss of IFN signature emerging resistance.
Integrative modelingMultivariate data integration to quantify individualized response probability.Machine-learning model combining TRAEs, ctDNA, miRNA, LDH, and IFN-γ GEP.Generates a Response Probability Score (RPS) categorized as high/intermediate/low.
Adaptive therapeutic decisionAdjust treatment intensity based on integrated signal.Compare RPS trajectory with toxicity profile and radiologic data.High RPS: continue current regimen. Low RPS + no irAEs: escalate (add CTLA-4 blockade or switch). Intermediate RPS: maintain with close follow-up.
Validation and feedback loopContinuous model refinement and prospective data accrual.Real-world registry data, digital pathology, radiomics integration.Calibrate thresholds, improve predictive accuracy, and enable AI-assisted decision support for future patients.
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

Venturi, F.; Magnaterra, E.; Gualandi, A.; Scotti, B.; Baraldi, C.; Alessandrini, A.M.; Veneziano, L.; Cama, E.M.; Melotti, B.; Marchese, P.V.; et al. Toward Personalized Response Monitoring in Melanoma Patients Treated with Immunotherapy and Target Therapy. Diagnostics 2025, 15, 3054. https://doi.org/10.3390/diagnostics15233054

AMA Style

Venturi F, Magnaterra E, Gualandi A, Scotti B, Baraldi C, Alessandrini AM, Veneziano L, Cama EM, Melotti B, Marchese PV, et al. Toward Personalized Response Monitoring in Melanoma Patients Treated with Immunotherapy and Target Therapy. Diagnostics. 2025; 15(23):3054. https://doi.org/10.3390/diagnostics15233054

Chicago/Turabian Style

Venturi, Federico, Elisabetta Magnaterra, Alberto Gualandi, Biagio Scotti, Carlotta Baraldi, Aurora Maria Alessandrini, Leonardo Veneziano, Elena Maria Cama, Barbara Melotti, Paola Valeria Marchese, and et al. 2025. "Toward Personalized Response Monitoring in Melanoma Patients Treated with Immunotherapy and Target Therapy" Diagnostics 15, no. 23: 3054. https://doi.org/10.3390/diagnostics15233054

APA Style

Venturi, F., Magnaterra, E., Gualandi, A., Scotti, B., Baraldi, C., Alessandrini, A. M., Veneziano, L., Cama, E. M., Melotti, B., Marchese, P. V., Tassone, D., Ribero, S., Ardigò, M., & Dika, E. (2025). Toward Personalized Response Monitoring in Melanoma Patients Treated with Immunotherapy and Target Therapy. Diagnostics, 15(23), 3054. https://doi.org/10.3390/diagnostics15233054

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