Skip to main content

Circulating MicroRNAs: functional biomarkers for melanoma prognosis and treatment

Abstract

MicroRNAs (miRNAs) hold significant promise as circulating cancer biomarkers and unlike many other molecular markers, they can provide valuable insights that extend beyond tumour biology. The expression of circulating miRNAs may parallel the cellular composition and dynamic activity within the tumour microenvironment and reveal systemic immune responses. The functional complexity of miRNAs—where a single miRNA can regulate multiple messenger RNAs (mRNAs) to fine tune fundamental processes, and a single mRNA can be targeted by multiple miRNAs—underscores their broad significance and impact. However, this complexity poses significant challenges for translating miRNA research into clinical practice. In melanoma, specific miRNA signatures have shown notable diagnostic, prognostic and predictive value, with lineage-specific and immune-related miRNAs frequently identified as valuable markers. In this review, we explore the role of circulating miRNAs as potential biomarkers in melanoma, and highlight the current status and advances required to translate miRNA research into therapeutic opportunities.

Introduction

Melanoma is an aggressive malignancy that arises from melanocytes, specialised pigment-producing cells derived from the neural crest. While melanocytes are primarily found in the skin, they are also present in the eyes, ears and mucosal membranes, and melanoma can develop in any of these locations [1]. Cutaneous melanoma accounts for over 90% of all melanoma diagnoses, while mucosal and uveal melanomas are rare, and comprise less than 5% of cases. The global incidence of cutaneous melanoma continues to rise, particularly in fair-skinned populations [2, 3] – worldwide melanoma incidence in 2019 was six times higher than 40 years prior [4], with an estimated 325,000 cases globally in 2020 [2]. Since 2011, significant advancements in treatment, including US Food and Drug Administration (FDA)-approved molecular targeted therapies (e.g. V-Raf Murine Sarcoma Viral Oncogene Homolog (BRAF) and mitogen-activated protein kinase kinase (MEK) inhibitors), as well as immune checkpoint inhibitors (e.g. programmed cell death-1 (PD-1) and cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) inhibitors), have contributed to a 17.9% reduction in melanoma mortality rates between 2013 and 2016 [5, 6]. The clinical benefits of these therapies are limited, however, by the development of innate and acquired resistance and the incidence of severe treatment-related adverse events. Approximately 70–90% of patients with advanced melanoma experience disease progression despite treatment with these therapies [7, 8], and nearly 60% of melanoma patients receiving combination anti-CTLA-4 and anti-PD-1 therapy experience grade 3 or 4 adverse events [8].

Further improvements in melanoma outcomes are expected through earlier screening and correct diagnosis [9, 10], alternative combination therapies such as relatlimab (Lymphocyte-activation gene 3 (LAG3) inhibitor) with nivolumab (PD-1 inhibitor) rather than nivolumab alone [11], and optimised treatment timing such as perioperative and response-driven adjuvant therapy for patients with high-risk disease [12, 13]. However, these advancements underscore the urgent need to identify biomarkers to monitor treatment response, detect resistance, and identify post-surgical recurrence. miRNAs are key regulators of cancer biology, playing crucial roles in tumour initiation, progression, and metastasis [14]. The expression of miRNAs is dysregulated in many cancers, including melanoma [15] and miRNAs are emerging as promising biomarkers for disease diagnosis and prognosis. In this review, we examine the role of circulating miRNAs in melanoma, focusing on their diagnostic, prognostic, and predictive potential. Additionally, we evaluate the therapeutic potential of miRNAs, exploring their role in enhancing treatment strategies for melanoma.

MicroRNAs: biosynthesis and functional roles

miRNAs are single-stranded ribonucleic acids (RNAs), typically 21–25 nucleotides in length [16, 17]. Most miRNAs are derived from longer non-coding or intronic primary transcripts, which are capped and polyadenylated. A smaller number of miRNAs are derived from exonic or intergenic deoxyribonucleic acid (DNA) regions [18, 19]. miRNAs are transcribed in the nucleus and exported to the cytoplasm, where they are cleaved into mature miRNAs by the Dicer endonuclease enzyme in complex with RNA binding proteins (Fig. 1) [20]. Comprehensive details on miRNA biogenesis are provided in several recent reviews [21, 22].

Fig. 1
figure 1

Canonical miRNA biosynthesis and miRNA function. Following transcription by RNA polymerase II/III (RNA pol II/III), primary miRNAs (pri-miRNAs) are cleaved at the base of a hairpin structure by the nuclear DROSHA RNase III to yield ~ 60–70 nucleotide precursor miRNAs (pre-miRNAs). Pre-miRNAs are exported to the cytoplasm via an Exportin-5 receptor-dependent process where they undergo further cleavage by the Dicer RNase III bound to RNA-binding proteins, such as TRBP. The resulting miRNA duplex associates with an AGO protein, where one miRNA strand (the guide strand) is selected to form the functional miRISC. The miRNA may originate from the 5’ end of the pre-miRNA or the 3’ end of the pre-miRNA giving rise to the -5p or -3p miRNAs, respectively. The unbound strand of the miRNA duplex is ejected from the AGO protein and degraded. Once formed, miRISC binds to target mRNAs and inhibits translation by competing with eIF4F recognition of the 7-methyl-G (m7G) cap, inhibiting cap-dependent translation initiation and destabilizing the target mRNA through the recruitment of de-adenylation complexes, which remove the stabilizing 3’ poly(A) tail

The 2024 Nobel Prize in Medicine was awarded to US scientists Victor Ambros and Gary Ruvkun for the discovery of miRNAs and their pivotal role in post-transcriptional regulation of gene expression. miRNAs bind to complementary sequences in the 3’ untranslated region (3’ UTR) or the 5’ UTR of target mRNAs to regulate the expression of approximately 60% of human genes [23]. This miRNA-mRNA interaction promotes the degradation or direct translational suppression of the target mRNAs [24,25,26] (Fig. 1). According to the miRNA repository (miRBase registry V22.1), there are currently 2656 mature human miRNA species [27]. A single miRNA can regulate multiple mRNA targets, with its binding specificity determined by the complementarity between the miRNA seed region (bases 2–8 of the miRNA [28]) and the 3’ UTR of target mRNAs [29, 30]. For example, miR-34 interacts with and downregulates the expression of several cell cycle regulatory genes, including cyclin-dependent kinase 4 (CDK4) [31], CDK6, cyclin D1 [32], and E2F transcription factor 3 [33]. Conversely, a single mRNA can be targeted by multiple miRNAs, leading to complex regulation of gene expression. For instance, over 15 miRNAs have been predicted to bind with high probability to the 3’ UTR of microphthalmia-associated transcription factor (MITF), a key transcriptional regulator of melanocyte development and function (Fig. 2).

Fig. 2
figure 2

miRNA regulators of the MITF gene. Multiple miRNAs can bind to identical and overlapping regions within the 3’ UTR of the target human MITF mRNA. miRNAs with 8mer or 7mer seed sites are highlighted in red and blue, respectively. Data are derived from TargetScanHuman (release 7.2) [181]. The hg19 chromosome coordinates of the MITF mRNA and the miRNA binding sites are shown

miRNAs regulate many biological processes, including cell development, growth, differentiation, metabolism, and homeostasis, both under normal physiological conditions and in various diseases [34, 35]. Generally, oncogenic miRNAs or oncomiRs promote cancer development and are overexpressed in cancer, whereas tumour suppressor miRNAs may promote apoptosis or cell cycle arrest and are under expressed in cancer [36]. Importantly, this classification lacks consistency and may not be informative, as miRNAs can act as either tumour-promoting or tumour-suppressive (e.g. miR-125 [37]), a distinction most likely dictated by the transcriptional milieu of the cancer.

miRNAs are also released into extracellular fluids, and circulating miRNAs have been detected in plasma, serum [38, 39], cerebrospinal fluid [40], and urine [41]. In cancer, both viable and dying tumour cells actively secrete miRNA to produce circulating miRNAs [39, 42]. Most circulating miRNAs are stabilised due to their association with proteins, such as the miRNA-inducing silencing complex (miRISC) effector protein Argonaute 2 (AGO2), nucleophosmin, high-density lipoproteins [43, 44] or extracellular vesicles such as exosomes, microvesicles and apoptotic bodies [45]. The lineage, differentiation and tissue-specificity of miRNAs make them valuable disease biomarkers, while their stability enhances their utility as reliable liquid biopsy biomarkers.

Circulating MiRNAs as diagnostic biomarkers in melanoma

The current standard for melanoma diagnosis involves the histopathological assessment of an excision biopsy, interpreted in conjunction with clinical information such as patient age, lesion site, and lesion history [46]. Despite the standardisation of diagnostic criteria, the diagnosis of melanomas can be challenging. A histopathological review of over 3300 melanocytic lesions demonstrated substantial diagnostic variation, with 24% of cases receiving equivocal diagnostic results between dermatopathologists [47]. Diagnostic ambiguity was associated with variations in treatment [47], and heightened diagnostic scrutiny has led to an increased rate of misdiagnosing benign melanocytic lesions as melanoma [4]. Consequently, clinicians are evaluating and implementing adjunct diagnostic tools, including immunohistochemistry stains (e.g. PReferentially expressed Antigen in MElanoma or PRAME [48]), gene expression signatures [49], genomic analysis [50] and digital pathology workflows [51] to support the diagnosis of challenging cases.

A recent systematic review [52] identified nine independent studies [53,54,55,56,57,58,59,60,61] investigating the diagnostic utility of circulating miRNAs in melanoma. All studies used a melanoma-healthy control design and employed reverse transcription-quantitative polymerase chain reaction (RT-qPCR) to measure the expression of individual or panels of circulating miRNAs. Across these studies, 82 distinct miRNAs were detected in blood, plasma or serum, with individual diagnostic sensitivity ranging from 0.71 to 0.97 and specificity from 0.62 to 0.98. Notably, despite minimal overlap in miRNAs investigated across the nine studies, a pooled analysis of these 82 miRNAs demonstrated a diagnostic sensitivity of 0.89, specificity of 0.85, and an area under the curve (AUC) of 0.93 [52]. Importantly, several miRNAs that distinguished melanoma from healthy controls also discriminated melanoma from other cancers. For instance, loss of serum miR-29c distinguished melanoma from metastatic colon and renal cancer [62]. miRNA cancer specificity also likely reflects lineage specific differences as some miRNAs, such as miR-211-5p, are located within an intron of TPRM1, which is a target gene of MITF [63].

The circulating MEL38 miRNA signature emerges as a notable diagnostic panel for melanoma with significant validation data. MEL38 miRNAs were identified as differentially expressed between individuals with or without cutaneous melanoma by whole miRNA profiling [53]. This signature consists of skin-cell derived miRNAs and 22/38 miRNAs have been previously identified in melanoma [53]. The MEL38 miRNA signature is enriched for pathways related to melanogenesis, T cell activation and mitogen-activated protein kinase (MAPK) activation [53]. The diagnostic utility of MEL38 has been independently validated in peripheral blood [53], in formalin-fixed paraffin-embedded tissue biopsies [64], and recently, in 582 plasma samples derived from melanoma patients or control individuals with non-melanoma skin conditions [54]. In the latter study, using a threshold of 5.5, MEL38 achieved a 93% true-positive rate (sensitivity) and a 98% true-negative rate (specificity) for the presence of invasive melanoma [54]. MEL38 was associated with melanoma, irrespective of tumour thickness and melanoma type (superficial, nodal or amelanotic) [53]. Importantly, despite being designed as a diagnostic signature, MEL38 also showed prognostic value as a continuous predictor of melanoma-specific survival (MSS) [54]. Although circulating miRNAs hold potential as an adjunct to pathological assessment in diagnosing challenging lesions, their clinical implementation remains limited. In fact, we identified only one miRNA-based diagnostic tool currently in use in oncology. The ThyGeNEXT oncogene panel, combined with the ThyraMIR v2 miRNA panel, is applied to improve the diagnostic accuracy of cytologically indeterminate thyroid nodules, with a reported sensitivity of 96% and specificity of 99% [65].

Overall, miRNAs have yet to demonstrate consistent and reliable diagnostic utility in broader clinical settings, and their application remains largely exploratory. Advancing the diagnostic potential of miRNA signatures will require large-scale studies involving representative cohorts, comprehensive clinical annotation and standardised experimental workflows. These studies should also integrate multi-omics data, including genetic, clinicopathological, and imaging information, to enhance the robustness and clinical utility of miRNA-based diagnostic markers.

Circulating MiRNAs as prognostic biomarkers in melanoma

Immune and targeted therapies have revolutionised the treatment of patients with advanced (stage IV) melanoma and these therapies are being used in the earlier-stage setting, including in patients with stage II (node-negative) [66, 67] and stage III melanoma (disease has spread to regional lymph nodes [13, 68, 69]). The outcomes of these patients are heterogenous, with substantial risks of recurrence. For instance, 27.2% of patients with stage II melanoma will recur within 5 years [70] and the 5-year MSS rates for Stage IIB (87%), IIC (82%) and IIIB (83%) are comparable [71]. There is, therefore, significant interest in developing reliable prognostic biomarkers, across all stages of melanoma, to guide treatment-decision making. Several commercially available gene expression profiles have been developed to stratify melanoma patients according to risk of recurrence. These include the DecisionDx-melanoma 31-gene expression profiling (GEP) assay (Castle Biosciences, USA) [72, 73], the Merlin 8-GEP test (Skyline Dx, Netherlands) [74] and the MelaGenix 11-GEP test (NeraCare, GmbH) [75].

Numerous studies have also investigated the association of circulating miRNAs with melanoma patient outcomes, including overall survival (OS), disease free survival (DFS), MSS and the development of brain or distant metastases (Table 1). These investigations typically focus on a narrow set of miRNAs, and we have identified over 50 reported prognostic circulating miRNAs in melanoma from various stages; most have not been validated and only a few have been identified across multiple studies (Table 1).

Table 1 Prognostic Circulating MiRNA biomarkers in cutaneous melanoma

An intriguing finding from melanoma prognostic studies is the link between immune-modulatory miRNAs (im-miRNAs) and patient outcomes. For instance, miR-150-5p is predominantly derived from tumour infiltrating lymphocytes [90] and has been shown to be released into the circulation upon T cell activation [91]. Predictably, low miR-150-5p expression in primary melanoma tissue correlated with low intratumoural CD45+ immune cells [90]. Low tissue expression of miR-150 was also associated with poor survival outcomes in stage III and IV melanoma patients [83]. In the circulation, serum miR-150 expression shows an inconsistent association with melanoma prognosis, although two out of three studies report lower serum miR-150 levels associated with poorer outcomes in early-stage and advanced melanoma [76, 83] (Table 2). miR-15b (targets programmed cell death ligand 1 (PD-L1) [92]), and miR-23a (targets transcription factor B lymphocyte-induced maturation protein-1 [93]), activate cytotoxic CD8+ T cells and lower serum levels of these immune regulators are linked to poor outcomes in melanoma patients [77, 80]. Finally, miR-221, a commonly identified prognostic melanoma marker, inhibits Janus kinase/Signal transducers and activators of transcription (JAK/STAT) signalling by targeting protein coding genes such as SOCS3 and IRF2 [94], and modulates T helper 17 cell responses [95]; overexpression of this miRNA in serum is consistently associated with worse OS in melanoma patients [81, 86, 87].

These findings underscore the value and potential limitations of miRNAs. Their pleiotropic roles result in markers that provides a dynamic fingerprint that may reflect tumour-specific features, local immune responses and systemic immunity alongside patient outcomes. However, these extensive effects can also complicate the derivation of specific miRNA signatures with high prognostic specificity.

Circulating MiRNAs in melanoma treatment response and resistance

The development of therapies targeting BRAF and MEK and immune checkpoint inhibitors have dramatically improved the outcomes of patients with advanced and high-risk, stage III melanoma. Targeted therapies are an option for patients with BRAFV600-mutant melanoma, which represents about 40% of metastatic melanoma [96]. Approximately 70% of these patients will respond to combination BRAF/MEK inhibitors but acquired treatment resistance is common, and few patients will achieve durable responses. The 5-year progression-free survival (PFS) and OS rates for patients with advanced BRAF-mutant melanoma receiving combination BRAF/MEK inhibitors range from 14–25% and 31–35%, respectively [97,98,99]. Conversely, immune checkpoint inhibitors, including ipilimumab, an antibody inhibitor of CTLA-4, and PD-1 inhibitors, nivolumab and pembrolizumab, demonstrate long-term efficacy. In the phase 3 CheckMate 067 clinical trial, 5-year PFS and OS rates were 36% and 52%, respectively, in patients with advanced melanoma receiving combination nivolumab and ipilimumab [100]. The combination of nivolumab with relatlimab also shows durable responses with 5-year PFS and OS rates of 28% and 49%, respectively [11].

Despite the effectiveness of current molecular and immune-based therapies, more than 50% of melanoma patients will exhibit innate resistance or develop acquired resistance to therapy [7, 101, 102]. Many promising new treatments are being tested, including tumour-infiltrating lymphocytes (2024 FDA-approved Lifileucel [103]), mRNA and multi-peptide vaccines (e.g. phase 2 KEYNOTE-942 and Mel44 clinical trials [104, 105]), and bi-specific fusion proteins (e.g. PD-1/interleukin-2 bispecific antibody fusion protein IBI363 [106]). Ongoing efforts are focussed on identifying reliable biomarkers that can accurately predict and monitor treatment response.

The miRNAome undergoes large changes in response to BRAF inhibitor treatment and during the acquisition of resistance in melanoma cell lines. Although, the majority of miRNA changes are cell line dependent, it is evident that common pathways, including the MAPK cascade, survival networks and melanoma differentiation are deregulated during BRAF inhibitor resistance [107, 108], and miRNAs that regulate these pathways may function as potential biomarkers of treatment response and resistance. For example, miR-4443 (targets multiple survival pathways [109]), miR-199b-5p (targets vascular endothelial growth factor A (VEGFA) and hypoxia inducible factor 1 subunit alpha (HIF1α) [107]) and miR-4488 (targets differentiation marker nestin [110]) in plasma samples of melanoma patients discriminated BRAF/MEK inhibitor responders from non-responders with an AUC of 0.894 [107]. Melanoma dedifferentiation, characterized by MITF downregulation [111], leads to a slow-cycling cell state with increased resistance to BRAF/MEK and immune checkpoint inhibitors [112,113,114]. miRNAs targeting MITF (Fig. 2) are upregulated in BRAF inhibitor resistant melanoma cell models [108], but the relationship between MITF-targeting miRNAs in the circulation and BRAF inhibitor resistance remains to be investigated.

As reviewed elsewhere [115], im-miRNAs can regulate immune checkpoint expression, antigen processing/presentation and interferon gamma (IFNγ) signalling, and these miRNAs can additionally function as predictive biomarkers of immune checkpoint inhibitor therapy. For instance, a circulating miRNA signature (miR-146a, miR-155, miR-125b, miR-100, let-7e, miR-125a, miR-146b, and miR-99b) identified in extracellular vesicles released by melanoma tumours was associated with increased myeloid-derived suppressor cells and resistance to ipilimumab and nivolumab therapy [116].

Another study reported that patients with high plasma levels of miR-155-5p, miR-320a and miR-424-5p prior to first-line anti-PD-1 therapy had longer PFS and OS. This study also confirmed the immune regulatory role of miR-155-5p, with plasma miR-155-5p levels correlating with PD1+ CD4+ T cells [117]. Additionally, miR-28 (targets PD-1, T-cell immunoglobulin and mucin domain 3 and B- and T-lymphocyte attenuator [118]) and miR-17-5p (targets PD-L1 [119]), suppress expression of immune checkpoint molecules, thus modulating anti-tumour immune functions, and serum levels of miR-17-5p inversely correlated with tumour PD-L1 expression in patients with metastatic melanoma [119]. In a cohort of 33 patients, serum levels of miR-16-5p, miR-17-5p, miR-451a and miR-20a-5p were higher in melanoma anti-PD-1 responders (n = 10) compared to non-responders (n = 23), and elevated serum levels of miR-1972 and miR-4502 were associated with non-response to anti-PD-1 [120]. Multivariate logistic regression analysis indicated miR-16-5p, miR-17-5p and miR-20a-5p to be independent predictors of response to PD-1 inhibitor therapy [120]. These data suggest that miRNAs predictive of PD-1 inhibitor response may contribute to patient outcomes by modulating the immunosuppressive PD-1/PD-L1 axis, and more than 100 miRNAs have been implicated in the regulation of immune checkpoints, including PD-1, PD-L1 and CTLA-4 (reviewed in [121]). Collectively, these data indicate that miRNAs can act as predictive biomarkers, but also actively modulate treatment response and resistance. As a result, miRNAs are emerging as promising therapeutic targets.

Therapeutic potential of MiRNAs in melanoma

Given the critical roles that miRNAs play in melanoma progression and therapy response, targeting miRNAs has emerged as a compelling therapeutic strategy. Several miRNAs have been reported to regulate the MAPK pathway in melanoma (Fig. 3), and manipulating the expression of these miRNAs can alter melanoma sensitivity to BRAF/MEK inhibitors. For instance, miR-7 suppresses epidermal growth factor receptor (EGFR), insulin like growth factor 1 receptor (IGF-1R) and RAF proto-oncogene serine/threonine-protein kinase (CRAF) expression, consequently inhibiting MAPK and phosphatidylinositol 3-kinase/protein kinase B signalling, and transfection of melanoma cells with miR-7 mimics resensitised cells to the BRAF inhibitor vemurafenib [122]. Similarly, miR-579-3p was shown to target BRAF and restoration of miR-579-3p expression sensitised melanoma cells to BRAF and MEK inhibitors [123], suggesting that combining miR-579-3p mimics with existing targeted treatments could be beneficial. miRNAs can also modulate anti-tumour activity during immune checkpoint inhibitor therapy. For example, miR-155 promotes the activity of effector CD8+ T cells and its deficiency in T cells can dampen T cell activation by inhibiting IFNγ production; miR-155 upregulation in peripheral blood correlates with increased CD8+ T cell infiltration and improved tumour control [124], whilst miR-155 deficiency enhanced tumour growth in a B16F10 melanoma mouse model [125]. This enhanced tumour growth, however, can be attenuated by the administration of immune checkpoint inhibitors (anti-PD-1, anti-PD-L1 and anti-CTLA-4 combination), suggesting that combining miR-155 overexpression with immune checkpoint blockade may augment anti-tumour responses [125]. miR-146a has also been reported to suppress Signal transducer and activator of transcription 1 (STAT1) signalling and IFNγ production in T cells, and combination of miR-146a inhibition with anti-PD-1 improved survival in a melanoma mouse model compared to anti-PD-1 alone [126].

Fig. 3
figure 3

miRNAs modulating components of the MAPK pathway in melanoma. Several miRNAs have been shown to regulate the MAPK pathway in melanoma by binding to the 3’ or 5’ UTR of various MAPK pathway components. These include miR-7 (targets EGFR, IGF-1R and CRAF [122]), miR-199b-5p (targets VEGFA and HIF1α [107]), miR-204-5p (targets VEGFA [182]), miR-126-3p (targets VEGFA and BRAF [182]), miR-579-3p (targets BRAF [123]), miR-524-5p (targets BRAF and ERK [183]), and miR-876-3p (targets ERK2 [184])

The expression of miRNAs can be modulated, either by silencing or restoring their levels, to influence melanoma proliferation, progression, and survival. Antisense oligonucleotides, known as antagomiRs or anti-miRs, silence specific miRNAs by preventing their binding to target mRNAs (Fig. 4), and these can have broad functional effects. For example, transfection of melanoma cell lines with a miR-211 antagomiR derepressed insulin like growth factor 2 receptor (IGF-2R), nuclear factor of activated T cells 5 (NFAT5) and transforming growth factor beta receptor 2 (TGFBR2) expression, and increased melanoma cell invasion and migration [127]. In contrast, inhibition of the pro-metastatic miR-182 with antisense oligonucleotides reduced cell viability and suppressed migration and invasion of melanoma cell lines, likely by restoring the expression of miR-182 targets FOXO3 and MITF [128]. In another study, treatment of mice with anti-miR-182 reduced the number and size of melanoma liver metastases compared to control treated mice [129], further supporting miR-182 as a therapeutic target. Inhibition of miRNAs can also be achieved using the CRISPR/Cas system, but like other gene editing technologies, this approach is limited by off-target effects and the need for selective and efficient delivery to target cells (reviewed in [130]).

Fig. 4
figure 4

Strategies and delivery systems for miRNA-based therapies. miRNA constructs can be designed to either inhibit (antagomiRs, BlockmiRs, miRNA masking, miRNA sponges) or overexpress (miRNA mimics) target miRNAs. To maximise stability of miRNAs and minimise off-target effects, miRNA constructs can be packaged within viral or non-viral delivery systems for transport to specific tumour sites

BlockmiRs are modified oligonucleotides that inhibit miRNA activity by specifically binding to the 3’ UTR of the target mRNA and sterically hindering the miRNA-mRNA interaction. Indeed, miR-27a interaction with CDH5 (encodes Cadherin 5/VE-Cadherin) is abolished by BlockmiR CD5-2, and combination treatment of CD5-2 with anti-PD-1 significantly reduced tumour burden compared to CD5-2 or anti-PD-1 alone in a liver cancer mouse model [131]. Other inhibitory methods include small molecule inhibitors that interfere with miRNA transcription, miRNA sponges that serve as decoys with multiple binding sites to prevent miRNA-mRNA interactions, and miRNA masking, which uses sequences complementary to the miRNA binding site on the mRNA to form duplexes that abrogate miRNA effects (reviewed in [132], Fig. 4). Interestingly, endogenous RNA molecules that act as miRNA sponges, sequestering miRNAs away from their targets, have been described [133]. For example, TYRP1 mRNA binds miR-16, and prevents the suppression of miR-16 targets, including RAB17, which promotes melanoma proliferation. Masking of miR-16 binding sites on the TYRP1 mRNA with small antisense oligonucleotides increased miR-16 activity, decreased RAB17 mRNA and reduced tumour growth in a xenograft melanoma mouse model [134].

Conversely, miRNA expression and effects on mRNA targets can be restored using double-stranded oligonucleotides referred to as miRNA mimics or mimetics (Fig. 4). For instance, transfection of melanoma cells with a miR-26a mimic decreased cell proliferation by inducing cell cycle arrest, and inhibited cell migration and invasion; transfection of the mouse B16F10 melanoma cells with the miR-26 mimic inhibited tumour growth in vivo [135]. Overall, therapeutic strategies that inhibit or restore miRNA expression have demonstrated efficacy in modulating melanoma growth and progression in preclinical models.

A major challenge in the therapeutic targeting of miRNAs is achieving delivery to precise locations, such as tumour regions and metastatic sites, whilst reducing nonspecific cellular uptake and consequent toxicity. Because miRNAs can be rapidly degraded by nucleases and have poor and nonspecific cellular uptake, several modifications and delivery strategies have been developed and tested in vivo to circumvent these challenges [136]. These approaches must be well tolerated, safe and specific, and designed to minimize off-target effects and can include chemical modifications, organic, inorganic, and polymer-based non-viral delivery methods (e.g. liposomes, nanoparticles, peptide polymers, extracellular vesicles or exosomes) and viral vectors (e.g. lentiviral, retroviral, and adenoviral, Fig. 4). For example, antisense oligonucleotides can be modified with locked nucleic acids (LNAs), RNA nucleotides with a modified ribose moiety, to offer higher thermal stability, resistance to nuclease degradation and higher specificity and affinity [137]. Administration of an LNA-anti-miR-21 reduced miR-21 expression in tumours and inhibited tumour growth in a B16F10 melanoma mouse model [137] and a LNA-modified antisense oligonucleotides (LNA-miR-221 inhibitor) demonstrated excellent safety profile and anti-tumour activity in a Phase I clinical trial [138]. Likewise, the incorporation of mesyl phosphoramidate groups in miRNA-targeting antisense oligonucleotides has been shown to increase oligonucleotide hybridisation, stability and efficacy, when compared to equivalent phosphorothioate antisense oligonucleotides [139]. In a melanoma mouse model, mesyl phosphoramidate-modified anti-miRs targeting miR-21, miR-17 and miR-155 showed limited toxicity, including minimal dystrophy and necrosis in the liver parenchyma [140]. Moreover, combined targeting of all three miRNAs with the mesyl phosphoramidate modified antisense oligonucleotides was more effective compared to targeting single miRNA [140], demonstrating the improved efficacy and safety of targeting multiple miRNAs.

miRNAs packaged in vector systems have also been tested in melanoma. Polyethylenimine, a cationic polymer, can interact electrostatically with oligonucleotides and can protect small RNA molecules from degradation whilst increasing cellular uptake [141], and polyethylenimine-complexed anti-miR-150 and anti-miR-638 significantly inhibited tumour growth and metastasis in melanoma xenograft mouse models [142]. In addition to silencing miRNA expression with anti-miRs, vector systems can be utilised to deliver miRNAs to restore expression; delivery of lipid nanoparticles encapsulating miR-199-5p and miR-204-5p to xenograft mouse models enhanced BRAF/MEK inhibitor therapy with minimal adverse events [143]. Similarly, delivery of miR-21-3p encapsulated in gold nanoparticles increased tumour-specific cellular uptake and synergised with anti-PD-1 treatment to decrease tumour growth in a melanoma mouse model [144]. Overall, these chemical modifications and delivery systems are advancing miRNA-based therapies towards clinical application, by enhancing their stability and therapeutic efficacy while reducing toxicity and off-target effects.

Despite the improvements in miRNA targeting and delivery, to date, only a few clinical trials have investigated miRNA-based therapeutics in cancer (Table 2). In melanoma-focussed miRNA-based clinical studies, research has centred on evaluating miRNAs for their diagnostic and prognostic potential, although none have yet advanced to routine clinical application. In the therapeutic setting, some notable candidates have emerged, including miR-34 [145], miR-16 [146], miR-155 [147], and miR-10b [148], but among these, only MRX34, a miR-34 mimic encapsulated in liposomal nanoparticles, has been evaluated in a Phase I trial in patients with melanoma, liver cancer, small cell lung cancer, lymphoma, multiple myeloma, and renal cell carcinoma (NCT01829971). Results indicated that three out of 85 patients (3.5%) experienced prolonged partial responses, while 16 patients (19%) had stable disease; however, the trial was terminated due to unexpected immune-mediated adverse events resulting in deaths of four patients [145]. Other miRNA therapeutics have been evaluated in clinical trials for other cancer types and are summarised in Table 2.

Collectively, the clinical trials summarised in Table 2 underscore the low specificity and off-target effects of miRNA-based therapies, which may contribute to the occurrence of severe immune-related adverse events. Of note, many of the therapeutic trials were terminated or withdrawn, and only one (NCT06260774) has progressed to Phase II for patients with confirmed diagnoses of relapsed advanced or locally advanced solid tumours with no available standard therapy. The initial phase 0 data with a single patient receiving an injection of TTX-MC138 (nanoparticles loaded with miR-10b antagomiR) demonstrated uptake to bone, lung and liver metastases and selective retention of the drug in tumour tissue [148]. The Phase II trial is currently recruiting. This limited progress suggests minimal clinical benefit to date, raising concerns about the future viability of miRNA-based therapeutics [149]. Importantly, in the context of melanoma, the goal of miRNA-based therapies is not to replace current effective treatments but to complement them, potentially in combination with BRAF/MEK inhibitors or immune checkpoint inhibitors to enhance drug efficacy, prolong the durability of responses, or overcome resistance to these therapies.

Table 2 miRNA-based diagnostics and therapeutics in cancer clinical trials

Challenges and recommendations for circulating MiRNA research and analyses

As previously noted, the identification of circulating miRNA biomarkers across various independent studies has shown limited consistency, with conflicting findings regarding their diagnostic, prognostic, and predictive value. Numerous reports underscore the lack of standardisation in both pre-analytical and analytical steps for profiling circulating miRNAs [153,154,155,156,157,158]. As discussed in these reports, discrepancies likely arise from differences in miRNA detection methods (e.g. RT-qPCR, NanoString, microarray, whole or targeted miRNA sequencing), the quality and type of biospecimens analysed (fresh, archival, whole blood, plasma or serum), and the heterogeneity of the patient and control cohorts, including variations in subtype, stage and treatment history. Moreover, the low abundance of secreted miRNAs, and differences in extraction methods, quality control, normalisation of miRNA data and variable statistical evaluation methods contribute to the limited consistency and hinder the broader implementation of circulating miRNA biomarkers.

To address the lack of standardization, the National Cancer Institute (NCI) recently published a best practice guide for blood collection and processing, specifically for circulating miRNA research [159]. Table 3 summarises key analytical and pre-analytical considerations for miRNA research. It is important to note that this lack of standardisation extends beyond circulating miRNA research to other circulating biomarkers, including cell-free DNA and proteins [160].

Table 3 MiRNA pre-analytical and analytical guidelines

Conclusions

Despite significant advancements in miRNA sequencing technologies and the inherent advantages of miRNAs—such as their stability and low detection costs—research focused on melanoma-specific miRNAs has not translated into clinical impact. To date, only one miRNA-based diagnostic signature (ThyraMIR v2 used in combination with ThyGeNEXT oncogene panel) has been approved to provide risk stratification for thyroid nodules, and only one miRNA-based therapy (MRX34) has progressed to a Phase I clinical trial in melanoma (NCT01829971), and this trial was closed early due to severe immune-mediated adverse events [145]. The intricate regulatory functions of miRNAs remain challenging to fully understand, but they also underpin the significant prognostic and mechanistic roles of miRNAs.

As miRNA research evolves, the unique technical challenges, including selection of robust internal controls and understanding the influence of biological and technical factors, will be resolved. Only then can miRNAs be integrated into multi-omics frameworks that support personalized treatment approaches. Additionally, conducting discovery and validation studies in larger, well-annotated patient cohorts, along with a focus on miRNA panels as diagnostic and prognostic biomarkers, will accelerate the miRNA field. Certainly, in melanoma, miRNAs show the greatest promise as diagnostic and prognostic tools, and continued efforts in clinical trials are essential to assess the specificity of miRNA-based biomarkers. However, miRNA-based therapies face significant hurdles, including the need to mitigate off-target effects and improve therapeutic efficacy, and future research should also explore their synergistic effects with existing treatments, such as BRAF/MEK inhibitors and immune checkpoint inhibitors. A deeper understanding of miRNAs and their interactions will be essential to realise their potential as biomarkers and therapeutic targets, ultimately enhancing the care and outcomes of cancer patients.

Data availability

No datasets were generated or analysed during the current study.

Abbreviations

AGO:

Argonaute

AUC:

Area under curve

BRAF:

V-Raf Murine Sarcoma Viral Oncogene Homolog

CDK:

Cyclin-dependent kinases

CRAF:

RAF proto-oncogene serine/threonine-protein kinase

CTLA-4:

Cytotoxic T lymphocyte-associated antigen-4

DFS:

Disease-free survival

DNA:

Deoxyribonucleic acid

EGFR:

Epidermal growth factor receptor

eIF4F:

Eukaryotic initiation factor 4 F

FDA:

Food and Drug Administration

GEP:

Gene expression profiling

HIF1α:

Hypoxia inducible factor 1 subunit alpha

IFNγ:

Interferon gamma

IGF1R:

Insulin like growth factor 1 receptor

IGF2R:

Insulin like growth factor 2 receptor

im-miRNA:

Immune-modulatory miRNA

LAG3:

Lymphocyte-Activation Gene 3

LNA:

Locked Nucleic Acids

m7G:

7-methyl-G

MAPK:

Mitogen-activated protein kinase

MEK:

Mitogen-activated protein kinase kinase

MicroRNA:

Micro ribonucleic acid

miRISC:

miRNA-induced silencing complex

MITF:

Microphthalmia-associated transcription factor

mRNA:

Messenger RNA

MSS:

Melanoma-specific survival

NFAT5:

Nuclear factor of activated T-Cells 5

OS:

Overall survival

PD-1:

Programmed cell death-1

PD-L1:

Programmed cell death ligand 1

PFS:

Progression free survival

RNA:

Ribonucleic acid

RT-qPCR:

Reverse transcription-quantitative polymerase chain reaction

STAT1:

Signal transducer and activator of transcription 1

TGFBR2:

Transforming growth factor beta receptor 2

UTR:

Untranslated region

VEGFA:

Vascular endothelial growth factor A

References

  1. Schadendorf D, Fisher DE, Garbe C, Gershenwald JE, Grob JJ, Halpern A, Herlyn M, Marchetti MA, McArthur G, Ribas A, et al. Melanoma Nat Rev Dis Primers. 2015;1:15003.

    Article  PubMed  Google Scholar 

  2. Arnold M, Singh D, Laversanne M, Vignat J, Vaccarella S, Meheus F, Cust AE, de Vries E, Whiteman DC, Bray F. Global burden of cutaneous melanoma in 2020 and projections to 2040. JAMA Dermatol. 2022;158(5):495–503.

    Article  PubMed  PubMed Central  Google Scholar 

  3. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, Bray F. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209–49.

    Article  PubMed  Google Scholar 

  4. Welch HG, Mazer BL, Adamson AS. The rapid rise in cutaneous melanoma diagnoses. N Engl J Med. 2021;384(1):72–9.

    Article  PubMed  Google Scholar 

  5. Berk-Krauss J, Stein JA, Weber J, Polsky D, Geller AC. New systematic therapies and trends in cutaneous melanoma deaths among US Whites, 1986–2016. Am J Public Health. 2020;110(5):731–3.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Reinhart JP, Campbell EH, Proffer SL, Crum OM, Todd A, Gibson LE, Brewer JD, Demer AM. Incidence and mortality trends of primary cutaneous melanoma: A 50-year Rochester epidemiologic project study. JAAD Int. 2024;16:144–54.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Long GV, Weber JS, Infante JR, Kim KB, Daud A, Gonzalez R, Sosman JA, Hamid O, Schuchter L, Cebon J, et al. Overall survival and durable responses in patients with BRAF V600-Mutant metastatic melanoma receiving Dabrafenib combined with Trametinib. J Clin Oncol. 2016;34(8):871–8.

    Article  CAS  PubMed  Google Scholar 

  8. Wolchok JD, Chiarion-Sileni V, Rutkowski P, Cowey CL, Schadendorf D, Wagstaff J, Queirolo P, Dummer R, Butler MO, Hill AG et al. Final, 10-Year outcomes with nivolumab plus ipilimumab in advanced melanoma. N Engl J Med 2024.

  9. Matsumoto M, Wack S, Weinstock MA, Geller A, Wang H, Solano FX, Kirkwood JM, Ferris LK. Five-Year outcomes of a melanoma screening initiative in a large health care system. JAMA Dermatol. 2022;158(5):504–12.

    Article  PubMed  PubMed Central  Google Scholar 

  10. Whiteman DC, Olsen CM, MacGregor S, Law MH, Thompson B, Dusingize JC, Green AC, Neale RE, Pandeya N. Study FtQ: the effect of screening on melanoma incidence and biopsy rates*. Br J Dermatol. 2022;187(4):515–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Tawbi HA, Schadendorf D, Lipson EJ, Ascierto PA, Matamala L, Castillo Gutiérrez E, Rutkowski P, Gogas HJ, Lao CD, De Menezes JJ, et al. Relatlimab and nivolumab versus nivolumab in untreated advanced melanoma. N Engl J Med. 2022;386(1):24–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Patel SP, Othus M, Chen Y, Wright GP Jr., Yost KJ, Hyngstrom JR, Hu-Lieskovan S, Lao CD, Fecher LA, Truong TG, et al. Neoadjuvant-Adjuvant or Adjuvant-Only pembrolizumab in advanced melanoma. N Engl J Med. 2023;388(9):813–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Blank CU, Lucas MW, Scolyer RA, van de Wiel BA, Menzies AM, Lopez-Yurda M, Hoeijmakers LL, Saw RPM, Lijnsvelt JM, Maher NG, et al. Neoadjuvant nivolumab and ipilimumab in resectable stage III melanoma. N Engl J Med. 2024;391(18):1696–708.

    Article  CAS  PubMed  Google Scholar 

  14. Peng Y, Croce CM. The role of MicroRNAs in human cancer. Signal Transduct Target Ther. 2016;1:15004.

    Article  PubMed  PubMed Central  Google Scholar 

  15. Zhang L, Huang J, Yang N, Greshock J, Megraw MS, Giannakakis A, Liang S, Naylor TL, Barchetti A, Ward MR, et al. MicroRNAs exhibit high frequency genomic alterations in human cancer. Proc Natl Acad Sci U S A. 2006;103(24):9136–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Lagos-Quintana M, Rauhut R, Lendeckel W, Tuschl T. Identification of novel genes coding for small expressed RNAs. Science. 2001;294(5543):853–8.

    Article  CAS  PubMed  Google Scholar 

  17. Lau NC, Lim LP, Weinstein EG, Bartel DP. An abundant class of tiny RNAs with probable regulatory roles in caenorhabditis elegans. Science. 2001;294(5543):858–62.

    Article  CAS  PubMed  Google Scholar 

  18. Lee LW, Zhang S, Etheridge A, Ma L, Martin D, Galas D, Wang K. Complexity of the MicroRNA repertoire revealed by next-generation sequencing. RNA. 2010;16(11):2170–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Lee Y, Kim M, Han J, Yeom KH, Lee S, Baek SH, Kim VN. MicroRNA genes are transcribed by RNA polymerase II. Embo J. 2004;23(20):4051–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Lee HY, Zhou K, Smith AM, Noland CL, Doudna JA. Differential roles of human Dicer-binding proteins TRBP and PACT in small RNA processing. Nucleic Acids Res. 2013;41(13):6568–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Shang R, Lee S, Senavirathne G, Lai EC. MicroRNAs in action: biogenesis, function and regulation. Nat Rev Genet. 2023;24(12):816–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. O’Brien J, Hayder H, Zayed Y, Peng C. Overview of MicroRNA biogenesis, mechanisms of actions, and circulation. Front Endocrinol (Lausanne). 2018;9:402.

    Article  PubMed  Google Scholar 

  23. Liu B, Li J, Cairns MJ. Identifying MiRNAs, targets and functions. Brief Bioinform. 2012;15(1):1–19.

    Article  PubMed  PubMed Central  Google Scholar 

  24. Liu J, Carmell MA, Rivas FV, Marsden CG, Thomson JM, Song JJ, Hammond SM, Joshua-Tor L, Hannon GJ. Argonaute2 is the catalytic engine of mammalian RNAi. Science. 2004;305(5689):1437–41.

    Article  CAS  PubMed  Google Scholar 

  25. Meister G, Landthaler M, Patkaniowska A, Dorsett Y, Teng G, Tuschl T. Human Argonaute2 mediates RNA cleavage targeted by MiRNAs and SiRNAs. Mol Cell. 2004;15(2):185–97.

    Article  CAS  PubMed  Google Scholar 

  26. Djuranovic S, Nahvi A, Green R. miRNA-mediated gene Silencing by translational repression followed by mRNA deadenylation and decay. Science. 2012;336(6078):237–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Kozomara A, Birgaoanu M, Griffiths-Jones S. MiRBase: from MicroRNA sequences to function. Nucleic Acids Res. 2019;47(D1):D155–62.

    Article  CAS  PubMed  Google Scholar 

  28. Lewis BP, Shih Ih, Jones-Rhoades MW, Bartel DP, Burge CB. Prediction of mammalian MicroRNA targets. Cell. 2003;115(7):787–98.

    Article  CAS  PubMed  Google Scholar 

  29. Brennecke J, Stark A, Russell RB, Cohen SM. Principles of microRNA-target recognition. PLoS Biol. 2005;3(3):e85.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Bartel DP. MicroRNAs: target recognition and regulatory functions. Cell. 2009;136(2):215–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Feng H, Ge F, Du L, Zhang Z, Liu D. MiR-34b-3p represses cell proliferation, cell cycle progression and cell apoptosis in non-small-cell lung cancer (NSCLC) by targeting CDK4. J Cell Mol Med. 2019;23(8):5282–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Sun F, Fu H, Liu Q, Tie Y, Zhu J, Xing R, Sun Z, Zheng X. Downregulation of CCND1 and CDK6 by miR-34a induces cell cycle arrest. FEBS Lett. 2008;582(10):1564–8.

    Article  CAS  PubMed  Google Scholar 

  33. Xiang W, Lin H, Wang Q, Chen W, Liu Z, Chen H, Zhang H, Chen W. miR–34a suppresses proliferation and induces apoptosis of human lens epithelial cells by targeting E2F3. Mol Med Rep. 2016;14(6):5049–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Stefani G, Slack FJ. Small non-coding RNAs in animal development. Nat Rev Mol Cell Biol. 2008;9(3):219–30.

    Article  CAS  PubMed  Google Scholar 

  35. Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 2004;116(2):281–97.

    Article  CAS  PubMed  Google Scholar 

  36. Zhang B, Pan X, Cobb GP, Anderson TA. MicroRNAs as oncogenes and tumor suppressors. Dev Biol. 2007;302(1):1–12.

    Article  CAS  PubMed  Google Scholar 

  37. Sun Y-M, Lin K-Y, Chen Y-Q. Diverse functions of miR-125 family in different cell contexts. J Hematol Oncol. 2013;6(1):6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Chen X, Ba Y, Ma L, Cai X, Yin Y, Wang K, Guo J, Zhang Y, Chen J, Guo X, et al. Characterization of MicroRNAs in serum: a novel class of biomarkers for diagnosis of cancer and other diseases. Cell Res. 2008;18(10):997–1006.

    Article  CAS  PubMed  Google Scholar 

  39. Mitchell PS, Parkin RK, Kroh EM, Fritz BR, Wyman SK, Pogosova-Agadjanyan EL, Peterson A, Noteboom J, O’Briant KC, Allen A, et al. Circulating MicroRNAs as stable blood-based markers for cancer detection. Proc Natl Acad Sci U S A. 2008;105(30):10513–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Teplyuk NM, Mollenhauer B, Gabriely G, Giese A, Kim E, Smolsky M, Kim RY, Saria MG, Pastorino S, Kesari S, et al. MicroRNAs in cerebrospinal fluid identify glioblastoma and metastatic brain cancers and reflect disease activity. Neuro Oncol. 2012;14(6):689–700.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Juracek J, Slaby O. Urinary MicroRNAs as emerging class of noninvasive biomarkers. Methods Mol Biol. 2020;2115:221–47.

    Article  CAS  PubMed  Google Scholar 

  42. Schwarzenbach H, Hoon DS, Pantel K. Cell-free nucleic acids as biomarkers in cancer patients. Nat Rev Cancer. 2011;11(6):426–37.

    Article  CAS  PubMed  Google Scholar 

  43. Arroyo JD, Chevillet JR, Kroh EM, Ruf IK, Pritchard CC, Gibson DF, Mitchell PS, Bennett CF, Pogosova-Agadjanyan EL, Stirewalt DL, et al. Argonaute2 complexes carry a population of Circulating MicroRNAs independent of vesicles in human plasma. Proc Natl Acad Sci U S A. 2011;108(12):5003–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Geekiyanage H, Rayatpisheh S, Wohlschlegel JA, Brown R Jr., Ambros V. Extracellular MicroRNAs in human circulation are associated with MiRISC complexes that are accessible to anti-AGO2 antibody and can bind target mimic oligonucleotides. Proc Natl Acad Sci U S A. 2020;117(39):24213–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Chevillet JR, Kang Q, Ruf IK, Briggs HA, Vojtech LN, Hughes SM, Cheng HH, Arroyo JD, Meredith EK, Gallichotte EN, et al. Quantitative and stoichiometric analysis of the MicroRNA content of exosomes. Proc Natl Acad Sci U S A. 2014;111(41):14888–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Scolyer RA, Soyer HP, Kelly JW, James C, McLean CA, Coventry BJ, Ferguson PM, Rawson RV, Mar VJ, de Menezes SL, et al. Improving diagnostic accuracy for suspicious melanocytic skin lesions: new Australian melanoma clinical practice guidelines stress the importance of clinician/pathologist communication. Aust J Gen Pract. 2019;48(6):357–62.

    Article  PubMed  Google Scholar 

  47. Hosler GA, Goldberg MS, Estrada SI, O’Neil B, Amin SM, Plaza JA. Diagnostic discordance among histopathological reviewers of melanocytic lesions. J Cutan Pathol. 2024;51(8):624–33.

    Article  PubMed  Google Scholar 

  48. Lezcano C, Jungbluth AA, Busam KJ. Comparison of immunohistochemistry for PRAME with cytogenetic test results in the evaluation of challenging melanocytic tumors. Am J Surg Pathol. 2020;44(7):893–900.

    Article  PubMed  PubMed Central  Google Scholar 

  49. Clarke LE, Mabey B, Flake Ii DD, Meek S, Cassarino DS, Duncan LM, High WA, Napekoski KM, Prieto VG, Tetzlaff MT, et al. Clinical validity of a gene expression signature in diagnostically uncertain neoplasms. Per Med. 2020;17(5):361–71.

    Article  CAS  PubMed  Google Scholar 

  50. Andea AA. Molecular testing in melanoma for the surgical pathologist. Pathology. 2023;55(2):245–57.

    Article  CAS  PubMed  Google Scholar 

  51. Maher NG, Danaei Mehr H, Cong C, Adegoke NA, Vergara IA, Liu S, Scolyer RA. Weakly supervised deep learning image analysis can differentiate melanoma from Naevi on haematoxylin and eosin-stained histopathology slides. J Eur Acad Dermatol Venereol. 2024;38(12):2250–8.

    Article  CAS  PubMed  Google Scholar 

  52. Jones N, Nonaka T. Circulating MiRNAs as biomarkers for the diagnosis in patients with melanoma: systematic review and meta-analysis. Front Genet. 2024;15:1339357.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Van Laar R, Lincoln M, Van Laar B. Development and validation of a plasma-based melanoma biomarker suitable for clinical use. Br J Cancer. 2018;118(6):857–66.

    Article  PubMed  PubMed Central  Google Scholar 

  54. Van Laar R, Latif B, King S, Love C, Taubenheim N, Kalansooriya E, Wang W, Saad M, Winship I, Azzi A, et al. Validation of a MicroRNA liquid biopsy assay for diagnosis and risk stratification of invasive cutaneous melanoma. Br J Dermatol. 2023;189(3):292–301.

    Article  PubMed  Google Scholar 

  55. Leidinger P, Keller A, Borries A, Reichrath J, Rass K, Jager SU, Lenhof HP, Meese E. High-throughput MiRNA profiling of human melanoma blood samples. BMC Cancer. 2010;10:262.

    Article  PubMed  PubMed Central  Google Scholar 

  56. Stark MS, Klein K, Weide B, Haydu LE, Pflugfelder A, Tang YH, Palmer JM, Whiteman DC, Scolyer RA, Mann GJ et al. The Prognostic and Predictive Value of Melanoma-related MicroRNAs Using Tissue and Serum: A MicroRNA Expression Analysis. EBioMedicine. 2015;2(7):671–680.

  57. Bai M, Zhang H, Si L, Yu N, Zeng A, Zhao R. Upregulation of serum miR-10b is associated with poor prognosis in patients with melanoma. J Cancer. 2017;8(13):2487–91.

    Article  PubMed  PubMed Central  Google Scholar 

  58. Armand-Labit V, Meyer N, Casanova A, Bonnabau H, Platzer V, Tournier E, Sansas B, Verdun S, Thouvenot B, Hilselberger B, et al. Identification of a Circulating MicroRNA profile as a biomarker of metastatic cutaneous melanoma. Acta Derm Venereol. 2016;96(1):29–34.

    Article  CAS  PubMed  Google Scholar 

  59. Fogli S, Polini B, Carpi S, Pardini B, Naccarati A, Dubbini N, Lanza M, Breschi MC, Romanini A, Nieri P. Identification of plasma MicroRNAs as new potential biomarkers with high diagnostic power in human cutaneous melanoma. Tumour Biol. 2017;39(5):1010428317701646.

    Article  PubMed  Google Scholar 

  60. Tengda L, Shuping L, Mingli G, Jie G, Yun L, Weiwei Z, Anmei D. Serum Exosomal MicroRNAs as potent Circulating biomarkers for melanoma. Melanoma Res. 2018;28(4):295–303.

    Article  PubMed  Google Scholar 

  61. Mo H, Guan J, Yuan ZC, Lin X, Wu ZJ, Liu B, He JL. Expression and predictive value of miR-489 and miR-21 in melanoma metastasis. World J Clin Cases. 2019;7(19):2930–41.

    Article  PubMed  PubMed Central  Google Scholar 

  62. Greenberg E, Besser MJ, Ben-Ami E, Shapira-Frommer R, Itzhaki O, Zikich D, Levy D, Kubi A, Eyal E, Onn A, et al. A comparative analysis of total serum MiRNA profiles identifies novel signature that is highly indicative of metastatic melanoma: a pilot study. Biomarkers. 2013;18(6):502–8.

    Article  CAS  PubMed  Google Scholar 

  63. Margue C, Philippidou D, Reinsbach SE, Schmitt M, Behrmann I, Kreis S. New target genes of MITF-induced microRNA-211 contribute to melanoma cell invasion. PLoS ONE. 2013;8(9):e73473.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Laar RV, King S, McCoy R, Saad M, Fereday S, Winship I, Uzzell C, Landgren A. Translation of a Circulating MiRNA signature of melanoma into a solid tissue assay to improve diagnostic accuracy and precision. Biomark Med. 2021;15(13):1111–22.

    Article  CAS  PubMed  Google Scholar 

  65. Verma T, Marshall C, Dantey KE, Thompson DV, Banizs A, Finkelstein SD, DelTondo J. The role of the thygenext oncogene panel used in combination with the expanded MiRNA panel ThyraMIRv2 in indeterminate thyroid nodules: A large, blinded, real-world, observational study. Cancer Cytopathol. 2024;132(9):556–63.

    Article  CAS  PubMed  Google Scholar 

  66. Kirkwood JM, Del Vecchio M, Weber J, Hoeller C, Grob JJ, Mohr P, Loquai C, Dutriaux C, Chiarion-Sileni V, Mackiewicz J et al. Adjuvant nivolumab in resected stage IIB/C melanoma: primary results from the randomized, phase 3 checkmate 76K trial. Nat Med 2023.

  67. Luke JJ, Rutkowski P, Queirolo P, Del Vecchio M, Mackiewicz J, Chiarion-Sileni V, de la Cruz Merino L, Khattak MA, Schadendorf D, Long GV, et al. Pembrolizumab versus placebo as adjuvant therapy in completely resected stage IIB or IIC melanoma (KEYNOTE-716): a randomised, double-blind, phase 3 trial. Lancet. 2022;399(10336):1718–29.

    Article  CAS  PubMed  Google Scholar 

  68. Long GV, Hauschild A, Santinami M, Kirkwood JM, Atkinson V, Mandala M, Merelli B, Sileni VC, Nyakas M, Haydon A et al. Final results for adjuvant Dabrafenib plus Trametinib in stage III melanoma. N Engl J Med 2024.

  69. Versluis JM, Menzies AM, Sikorska K, Rozeman EA, Saw RPM, van Houdt WJ, Eriksson H, Klop WMC, Ch’ng S, van Thienen JV et al. Survival update of neoadjuvant ipilimumab plus nivolumab in macroscopic stage III melanoma in the opacin and opacin-neo trials. Ann Oncol 2023.

  70. Bleicher J, Swords DS, Mali ME, McGuire L, Pahlkotter MK, Asare EA, Bowles TL, Hyngstrom JR. Recurrence patterns in patients with stage II melanoma: the evolving role of routine imaging for surveillance. J Surg Oncol. 2020;122(8):1770–7.

    Article  PubMed  Google Scholar 

  71. Gershenwald JE, Scolyer RA. Melanoma staging: American joint committee on cancer (AJCC) 8th edition and beyond. Ann Surg Oncol. 2018;25(8):2105–10.

    Article  PubMed  Google Scholar 

  72. Zager JS, Gastman BR, Leachman S, Gonzalez RC, Fleming MD, Ferris LK, Ho J, Miller AR, Cook RW, Covington KR, et al. Performance of a prognostic 31-gene expression profile in an independent cohort of 523 cutaneous melanoma patients. BMC Cancer. 2018;18(1):130.

    Article  PubMed  PubMed Central  Google Scholar 

  73. Gastman BR, Gerami P, Kurley SJ, Cook RW, Leachman S, Vetto JT. Identification of patients at risk of metastasis using a prognostic 31-gene expression profile in subpopulations of melanoma patients with favorable outcomes by standard criteria. J Am Acad Dermatol. 2019;80(1):149–e157144.

    Article  CAS  PubMed  Google Scholar 

  74. Stassen R, Mulder E, Veringa A, Mooyaart A, Dwarkasing J, Tempel D, Hage JAvd, Lendfers S, Aarts MJB, Verhoef C, et al. Use of Merlin assay to identify patients with a low-risk for SN metastasis in a prospective multicenter Dutch study of a primary melanoma gene-signature (CP-GEP model) to predict Sentinel node status during COVID-19. J Clin Oncol. 2022;40(16suppl):9571–9571.

    Article  Google Scholar 

  75. Marchetti MA, Coit DG, Dusza SW, Yu A, McLean L, Hu Y, Nanda JK, Matsoukas K, Mancebo SE, Bartlett EK. Performance of gene expression profile tests for prognosis in patients with localized cutaneous melanoma: A systematic review and Meta-analysis. JAMA Dermatology. 2020;156(9):953–62.

    Article  PubMed  Google Scholar 

  76. Fleming NH, Zhong J, da Silva IP, Vega-Saenz de Miera E, Brady B, Han SW, Hanniford D, Wang J, Shapiro RL, Hernando E, et al. Serum-based MiRNAs in the prediction and detection of recurrence in melanoma patients. Cancer. 2015;121(1):51–9.

    Article  CAS  PubMed  Google Scholar 

  77. Friedman EB, Shang S, de Miera EV, Fog JU, Teilum MW, Ma MW, Berman RS, Shapiro RL, Pavlick AC, Hernando E, et al. Serum MicroRNAs as biomarkers for recurrence in melanoma. J Transl Med. 2012;10:155.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Guo S, Guo W, Li S, Dai W, Zhang N, Zhao T, Wang H, Ma J, Yi X, Ge R, et al. Serum miR-16: A potential biomarker for predicting melanoma prognosis. J Invest Dermatol. 2016;136(5):985–93.

    Article  CAS  PubMed  Google Scholar 

  79. Saldanha G, Potter L, Shendge P, Osborne J, Nicholson S, Yii N, Varma S, Aslam MI, Elshaw S, Papadogeorgakis E, et al. Plasma microRNA-21 is associated with tumor burden in cutaneous melanoma. J Invest Dermatol. 2013;133(5):1381–4.

    Article  CAS  PubMed  Google Scholar 

  80. Guo W, Wang H, Yang Y, Guo S, Zhang W, Liu Y, Yi X, Ma J, Zhao T, Liu L, et al. Down-regulated miR-23a contributes to the metastasis of cutaneous melanoma by promoting autophagy. Theranostics. 2017;7(8):2231–49.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Bagheri MS, Polivka J, Treskova I, Houfkova K, Knizkova T, Woznica V, Fikrle T, Pivovarcikova K, Svaton M, Shetti D, et al. Preoperative plasma MiRNA levels predict prognosis in Early-stage malignant melanoma. Anticancer Res. 2023;43(2):695–706.

    Article  CAS  PubMed  Google Scholar 

  82. Ono S, Lam S, Nagahara M, Hoon DS. Circulating MicroRNA biomarkers as liquid biopsy for cancer patients: pros and cons of current assays. J Clin Med. 2015;4(10):1890–907.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Tembe V, Schramm SJ, Stark MS, Patrick E, Jayaswal V, Tang YH, Barbour A, Hayward NK, Thompson JF, Scolyer RA, et al. MicroRNA and mRNA expression profiling in metastatic melanoma reveal associations with BRAF mutation and patient prognosis. Pigment Cell Melanoma Res. 2015;28(3):254–66.

    Article  CAS  PubMed  Google Scholar 

  84. Tian R, Liu T, Qiao L, Gao M, Li J. Decreased serum microRNA-206 level predicts unfavorable prognosis in patients with melanoma. Int J Clin Exp Pathol. 2015;8(3):3097–103.

    PubMed  PubMed Central  Google Scholar 

  85. Ono S, Oyama T, Lam S, Chong K, Foshag LJ, Hoon DS. A direct plasma assay of Circulating microRNA-210 of hypoxia can identify early systemic metastasis recurrence in melanoma patients. Oncotarget. 2015;6(9):7053–64.

    Article  PubMed  PubMed Central  Google Scholar 

  86. Li P, He QY, Luo CQ, Qian LY. Circulating miR-221 expression level and prognosis of cutaneous malignant melanoma. Med Sci Monit. 2014;20:2472–7.

    Article  PubMed  PubMed Central  Google Scholar 

  87. Kanemaru H, Fukushima S, Yamashita J, Honda N, Oyama R, Kakimoto A, Masuguchi S, Ishihara T, Inoue Y, Jinnin M, et al. The Circulating microRNA-221 level in patients with malignant melanoma as a new tumor marker. J Dermatol Sci. 2011;61(3):187–93.

    Article  CAS  PubMed  Google Scholar 

  88. Love CG, Coombs L, Van Laar R. RNA-seq validation of MicroRNA expression signatures for precision melanoma diagnosis and prognostic stratification. BMC Med Genomics. 2024;17(1):256.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Bustos MA, Tran KD, Rahimzadeh N, Gross R, Lin SY, Shoji Y, Murakami T, Boley CL, Tran LT, Cole H et al. Integrated assessment of Circulating Cell-Free MicroRNA signatures in plasma of patients with melanoma brain metastasis. Cancers (Basel) 2020, 12(6).

  90. Hanniford D, Zhong J, Koetz L, Gaziel-Sovran A, Lackaye DJ, Shang S, Pavlick A, Shapiro R, Berman R, Darvishian F, et al. A miRNA-Based signature detected in primary melanoma tissue predicts development of brain metastasis. Clin Cancer Res. 2015;21(21):4903–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. de Candia P, Torri A, Gorletta T, Fedeli M, Bulgheroni E, Cheroni C, Marabita F, Crosti M, Moro M, Pariani E, et al. Intracellular modulation, extracellular disposal and serum increase of MiR-150 mark lymphocyte activation. PLoS ONE. 2013;8(9):e75348.

    Article  PubMed  PubMed Central  Google Scholar 

  92. Pathania AS, Prathipati P, Olwenyi OA, Chava S, Smith OV, Gupta SC, Chaturvedi NK, Byrareddy SN, Coulter DW, Challagundla KB. miR-15a and miR-15b modulate natural killer and CD8(+)T-cell activation and anti-tumor immune response by targeting PD-L1 in neuroblastoma. Mol Ther Oncolytics. 2022;25:308–29.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Lin R, Chen L, Chen G, Hu C, Jiang S, Sevilla J, Wan Y, Sampson JH, Zhu B, Li QJ. Targeting miR-23a in CD8 + cytotoxic T lymphocytes prevents tumor-dependent immunosuppression. J Clin Invest. 2014;124(12):5352–67.

    Article  PubMed  PubMed Central  Google Scholar 

  94. Kneitz B, Krebs M, Kalogirou C, Schubert M, Joniau S, van Poppel H, Lerut E, Kneitz S, Scholz CJ, Ströbel P, et al. Survival in patients with high-risk prostate cancer is predicted by miR-221, which regulates proliferation, apoptosis, and invasion of prostate cancer cells by inhibiting IRF2 and SOCS3. Cancer Res. 2014;74(9):2591–603.

    Article  CAS  PubMed  Google Scholar 

  95. Mikami Y, Philips RL, Sciumè G, Petermann F, Meylan F, Nagashima H, Yao C, Davis FP, Brooks SR, Sun HW, et al. MicroRNA-221 and– 222 modulate intestinal inflammatory Th17 cell response as negative feedback regulators downstream of interleukin-23. Immunity. 2021;54(3):514–e525516.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Genomic Classification of Cutaneous Melanoma. Cell. 2015;161(7):1681–96.

    Article  Google Scholar 

  97. Dummer R, Flaherty KT, Robert C, Arance A, de Groot JWB, Garbe C, Gogas HJ, Gutzmer R, Krajsová I, Liszkay G, et al. COLUMBUS 5-Year update: A randomized, Open-Label, phase III trial of Encorafenib plus binimetinib versus Vemurafenib or Encorafenib in patients with BRAF V600-Mutant melanoma. J Clin Oncol. 2022;40(36):4178–88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Robert C, Grob JJ, Stroyakovskiy D, Karaszewska B, Hauschild A, Levchenko E, Chiarion Sileni V, Schachter J, Garbe C, Bondarenko I, et al. Five-Year outcomes with Dabrafenib plus Trametinib in metastatic melanoma. N Engl J Med. 2019;381(7):626–36.

    Article  CAS  PubMed  Google Scholar 

  99. Ascierto PA, Dréno B, Larkin J, Ribas A, Liszkay G, Maio M, Mandalà M, Demidov L, Stroyakovskiy D, Thomas L, et al. 5-Year outcomes with Cobimetinib plus Vemurafenib in BRAFV600 Mutation-Positive advanced melanoma: extended Follow-up of the cobrim study. Clin Cancer Res. 2021;27(19):5225–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Larkin J, Chiarion-Sileni V, Gonzalez R, Grob JJ, Rutkowski P, Lao CD, Cowey CL, Schadendorf D, Wagstaff J, Dummer R, et al. Five-Year survival with combined nivolumab and ipilimumab in advanced melanoma. N Engl J Med. 2019;381(16):1535–46.

    Article  CAS  PubMed  Google Scholar 

  101. Wolchok JD, Chiarion-Sileni V, Gonzalez R, Grob JJ, Rutkowski P, Lao CD, Cowey CL, Schadendorf D, Wagstaff J, Dummer R, et al. Long-Term outcomes with nivolumab plus ipilimumab or nivolumab alone versus ipilimumab in patients with advanced melanoma. J Clin Oncol. 2022;40(2):127–37.

    Article  CAS  PubMed  Google Scholar 

  102. Long GV, Eroglu Z, Infante J, Patel S, Daud A, Johnson DB, Gonzalez R, Kefford R, Hamid O, Schuchter L, et al. Long-Term outcomes in patients with BRAF V600-Mutant metastatic melanoma who received Dabrafenib combined with Trametinib. J Clin Oncol. 2018;36(7):667–73.

    Article  CAS  PubMed  Google Scholar 

  103. Julve M, Lythgoe MP, Larkin J, Furness AJS. Lifileucel: the first cellular therapy approved for solid tumours. Trends Cancer. 2024;10(6):475–7.

    Article  CAS  PubMed  Google Scholar 

  104. Weber JS, Carlino MS, Khattak A, Meniawy T, Ansstas G, Taylor MH, Kim KB, McKean M, Long GV, Sullivan RJ, et al. Individualised neoantigen therapy mRNA-4157 (V940) plus pembrolizumab versus pembrolizumab monotherapy in resected melanoma (KEYNOTE-942): a randomised, phase 2b study. Lancet. 2024;403(10427):632–44.

    Article  CAS  PubMed  Google Scholar 

  105. Ninmer EK, Zhu H, Chianese-Bullock KA, von Mehren M, Haas NB, Ross MI, Dengel LT, Slingluff CL Jr. Multipeptide vaccines for melanoma in the adjuvant setting: long-term survival outcomes and post-hoc analysis of a randomized phase II trial. Nat Commun. 2024;15(1):2570.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Chen Y, Lin J, Wang H, Zhang W, Liang X, Cui J, Sun Y, Fang M, Pan Y, Chu Q, et al. First-in-class PD-1/IL-2 bispecific antibody fusion protein IBI363 in patients with advanced melanoma: safety and efficacy results from a phase I study. J Clin Oncol. 2024;42(16suppl):9562–9562.

    Article  Google Scholar 

  107. Fattore L, Ruggiero CF, Pisanu ME, Liguoro D, Cerri A, Costantini S, Capone F, Acunzo M, Romano G, Nigita G, et al. Reprogramming MiRNAs global expression orchestrates development of drug resistance in BRAF mutated melanoma. Cell Death Differ. 2019;26(7):1267–82.

    Article  CAS  PubMed  Google Scholar 

  108. Kozar I, Cesi G, Margue C, Philippidou D, Kreis S. Impact of BRAF kinase inhibitors on the mirnomes and transcriptomes of melanoma cells. Biochim Biophys Acta Gen Subj. 2017;1861(11 Pt B):2980–92.

    Article  CAS  PubMed  Google Scholar 

  109. Mao Y, Shen J, Wu Y, Wenjing R, Zhu F, Duan S. Aberrant expression of microRNA-4443 (miR-4443) in human diseases. Bioengineered. 2022;13(6):14770–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Castaldo V, Minopoli M, Di Modugno F, Sacconi A, Liguoro D, Frigerio R, Ortolano A, Di Martile M, Gesualdi L, Madonna G, et al. Upregulated expression of miR-4443 and miR-4488 in drug resistant melanomas promotes migratory and invasive phenotypes through downregulation of intermediate filament Nestin. J Exp Clin Cancer Res. 2023;42(1):317.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Hoek KS, Eichhoff OM, Schlegel NC, Döbbeling U, Kobert N, Schaerer L, Hemmi S, Dummer R. In vivo switching of human melanoma cells between proliferative and invasive States. Cancer Res. 2008;68(3):650–6.

    Article  CAS  PubMed  Google Scholar 

  112. Muller J, Krijgsman O, Tsoi J, Robert L, Hugo W, Song C, Kong X, Possik PA, Cornelissen-Steijger PD, Geukes Foppen MH, et al. Low MITF/AXL ratio predicts early resistance to multiple targeted drugs in melanoma. Nat Commun. 2014;5:5712.

    Article  PubMed  Google Scholar 

  113. Landsberg J, Kohlmeyer J, Renn M, Bald T, Rogava M, Cron M, Fatho M, Lennerz V, Wölfel T, Hölzel M, et al. Melanomas resist T-cell therapy through inflammation-induced reversible dedifferentiation. Nature. 2012;490(7420):412–6.

    Article  CAS  PubMed  Google Scholar 

  114. Konieczkowski DJ, Johannessen CM, Abudayyeh O, Kim JW, Cooper ZA, Piris A, Frederick DT, Barzily-Rokni M, Straussman R, Haq R, et al. A melanoma cell state distinction influences sensitivity to MAPK pathway inhibitors. Cancer Discov. 2014;4(7):816–27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Eichmüller SB, Osen W, Mandelboim O, Seliger B. Immune modulatory MicroRNAs involved in tumor attack and tumor immune escape. J Natl Cancer Inst 2017, 109(10).

  116. Huber V, Vallacchi V, Fleming V, Hu X, Cova A, Dugo M, Shahaj E, Sulsenti R, Vergani E, Filipazzi P, et al. Tumor-derived MicroRNAs induce myeloid suppressor cells and predict immunotherapy resistance in melanoma. J Clin Invest. 2018;128(12):5505–16.

    Article  PubMed  PubMed Central  Google Scholar 

  117. Durante G, Comito F, Naddeo M, De Matteis S, Bonafè M, Dika E, Ardizzoni A, Ferracin M. Abstract 5160: evaluation of Circulating MicroRNAs as predictive and prognostic biomarkers for advanced melanoma patients. Cancer Res. 2024;84(6Supplement):5160–5160.

    Article  Google Scholar 

  118. Li Q, Johnston N, Zheng X, Wang H, Zhang X, Gao D, Min W. miR-28 modulates exhaustive differentiation of T cells through Silencing programmed cell death-1 and regulating cytokine secretion. Oncotarget. 2016;7(33):53735–50.

    Article  PubMed  PubMed Central  Google Scholar 

  119. Audrito V, Serra S, Stingi A, Orso F, Gaudino F, Bologna C, Neri F, Garaffo G, Nassini R, Baroni G, et al. PD-L1 up-regulation in melanoma increases disease aggressiveness and is mediated through miR-17-5p. Oncotarget. 2017;8(9):15894–911.

    Article  PubMed  PubMed Central  Google Scholar 

  120. 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(1):77–9.

    Article  CAS  PubMed  Google Scholar 

  121. Kipkeeva F, Muzaffarova T, Korotaeva A, Mansorunov D, Apanovich P, Nikulin M, Malikhova O, Stilidi I, Karpukhin A. The features of immune checkpoint gene regulation by MicroRNA in cancer. Int J Mol Sci 2022, 23(16).

  122. Sun X, Li J, Sun Y, Zhang Y, Dong L, Shen C, Yang L, Yang M, Li Y, Shen G, et al. miR-7 reverses the resistance to BRAFi in melanoma by targeting EGFR/IGF-1R/CRAF and inhibiting the MAPK and PI3K/AKT signaling pathways. Oncotarget. 2016;7(33):53558–70.

    Article  PubMed  PubMed Central  Google Scholar 

  123. Fattore L, Mancini R, Acunzo M, Romano G, Laganà A, Pisanu ME, Malpicci D, Madonna G, Mallardo D, Capone M, et al. miR-579-3p controls melanoma progression and resistance to target therapy. Proc Natl Acad Sci U S A. 2016;113(34):E5005–5013.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Martinez-Usatorre A, Sempere LF, Carmona SJ, Carretero-Iglesia L, Monnot G, Speiser DE, Rufer N, Donda A, Zehn D, Jandus C, et al. MicroRNA-155 expression is enhanced by T-cell receptor stimulation strength and correlates with improved tumor control in melanoma. Cancer Immunol Res. 2019;7(6):1013–24.

    Article  CAS  PubMed  Google Scholar 

  125. Huffaker TB, Lee SH, Tang WW, Wallace JA, Alexander M, Runtsch MC, Larsen DK, Thompson J, Ramstead AG, Voth WP, et al. Antitumor immunity is defective in T cell-specific microRNA-155-deficient mice and is rescued by immune checkpoint Blockade. J Biol Chem. 2017;292(45):18530–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Mastroianni J, Stickel N, Andrlova H, Hanke K, Melchinger W, Duquesne S, Schmidt D, Falk M, Andrieux G, Pfeifer D, et al. miR-146a controls immune response in the melanoma microenvironment. Cancer Res. 2019;79(1):183–95.

    Article  CAS  PubMed  Google Scholar 

  127. Levy C, Khaled M, Iliopoulos D, Janas MM, Schubert S, Pinner S, Chen PH, Li S, Fletcher AL, Yokoyama S, et al. Intronic miR-211 assumes the tumor suppressive function of its host gene in melanoma. Mol Cell. 2010;40(5):841–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Segura MF, Hanniford D, Menendez S, Reavie L, Zou X, Alvarez-Diaz S, Zakrzewski J, Blochin E, Rose A, Bogunovic D, et al. Aberrant miR-182 expression promotes melanoma metastasis by repressing FOXO3 and microphthalmia-associated transcription factor. Proc Natl Acad Sci U S A. 2009;106(6):1814–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Huynh C, Segura MF, Gaziel-Sovran A, Menendez S, Darvishian F, Chiriboga L, Levin B, Meruelo D, Osman I, Zavadil J, et al. Efficient in vivo MicroRNA targeting of liver metastasis. Oncogene. 2011;30(12):1481–8.

    Article  CAS  PubMed  Google Scholar 

  130. Hussen BM, Rasul MF, Abdullah SR, Hidayat HJ, Faraj GSH, Ali FA, Salihi A, Baniahmad A, Ghafouri-Fard S, Rahman M, et al. Targeting MiRNA by CRISPR/Cas in cancer: advantages and challenges. Mil Med Res. 2023;10(1):32.

    CAS  PubMed  PubMed Central  Google Scholar 

  131. Liu K, Chen J, Zhao Y, Boland J, Ting KK, Lockwood G, McKenzie C, Kench J, Vadas MA, Gamble JR, et al. Novel miRNA-based drug CD5-2 reduces liver tumor growth in diethylnitrosamine-treated mice by normalizing tumor vasculature and altering immune infiltrate. Front Immunol. 2023;14:1245708.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Lima JF, Cerqueira L, Figueiredo C, Oliveira C, Azevedo NF. Anti-miRNA oligonucleotides: A comprehensive guide for design. RNA Biol. 2018;15(3):338–52.

    Article  PubMed  PubMed Central  Google Scholar 

  133. Thomson DW, Dinger ME. Endogenous MicroRNA sponges: evidence and controversy. Nat Rev Genet. 2016;17(5):272–83.

    Article  CAS  PubMed  Google Scholar 

  134. Gilot D, Migault M, Bachelot L, Journe F, Rogiers A, Donnou-Fournet E, Mogha A, Mouchet N, Pinel-Marie ML, Mari B, et al. A non-coding function of TYRP1 mRNA promotes melanoma growth. Nat Cell Biol. 2017;19(11):1348–57.

    Article  CAS  PubMed  Google Scholar 

  135. Qian H, Yang C, Yang Y. MicroRNA-26a inhibits the growth and invasiveness of malignant melanoma and directly targets on MITF gene. Cell Death Discov. 2017;3:17028.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Chen Y, Gao DY, Huang L. In vivo delivery of MiRNAs for cancer therapy: challenges and strategies. Adv Drug Deliv Rev. 2015;81:128–41.

    Article  CAS  PubMed  Google Scholar 

  137. Javanmard SH, Vaseghi G, Ghasemi A, Rafiee L, Ferns GA, Esfahani HN, Nedaeinia R. Therapeutic Inhibition of microRNA-21 (miR-21) using locked-nucleic acid (LNA)-anti-miR and its effects on the biological behaviors of melanoma cancer cells in preclinical studies. Cancer Cell Int. 2020;20:384.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Tassone P, Di Martino MT, Arbitrio M, Fiorillo L, Staropoli N, Ciliberto D, Cordua A, Scionti F, Bertucci B, Salvino A, et al. Safety and activity of the first-in-class locked nucleic acid (LNA) miR-221 selective inhibitor in refractory advanced cancer patients: a first-in-human, phase 1, open-label, dose-escalation study. J Hematol Oncol. 2023;16(1):68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Patutina OA, Gaponova Miroshnichenko SK, Sen’kova AV, Savin IA, Gladkikh DV, Burakova EA, Fokina AA, Maslov MA, Shmendel EV, Wood MJA, et al. Mesyl phosphoramidate backbone modified antisense oligonucleotides targeting miR-21 with enhanced in vivo therapeutic potency. Proc Natl Acad Sci U S A. 2020;117(51):32370–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Gaponova S, Patutina O, Sen’kova A, Burakova E, Savin I, Markov A, Shmendel E, Maslov M, Stetsenko D, Vlassov V et al. Single Shot vs. Cocktail: A Comparison of Mono- and Combinative Application of miRNA-Targeted Mesyl Oligonucleotides for Efficient Antitumor Therapy. Cancers (Basel). 2022;14(18).

  141. Thomas M, Lange-Grunweller K, Dayyoub E, Bakowsky U, Weirauch U, Aigner A, Hartmann RK, Grunweller A. PEI-complexed LNA antiseeds as MiRNA inhibitors. RNA Biol. 2012;9(8):1088–98.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Kunz M, Brandl M, Bhattacharya A, Nobereit-Siegel L, Ewe A, Weirauch U, Hering D, Reinert A, Kalwa H, Guzman J, et al. Nanoparticle-complexed AntimiRs for inhibiting tumor growth and metastasis in prostate carcinoma and melanoma. J Nanobiotechnol. 2020;18(1):173.

    Article  CAS  Google Scholar 

  143. Fattore L, Cafaro G, Di Martile M, Campani V, Sacconi A, Liguoro D, Marra E, Bruschini S, Stoppoloni D, Cirombella R, et al. Oncosuppressive MiRNAs loaded in lipid nanoparticles potentiate targeted therapies in BRAF-mutant melanoma by inhibiting core escape pathways of resistance. Oncogene. 2023;42(4):293–307.

    Article  CAS  PubMed  Google Scholar 

  144. Guo W, Wu Z, Chen J, Guo S, You W, Wang S, Ma J, Wang H, Wang X, Wang H et al. Nanoparticle delivery of miR-21-3p sensitizes melanoma to anti-PD-1 immunotherapy by promoting ferroptosis. J Immunother Cancer 2022, 10(6).

  145. Hong DS, Kang YK, Borad M, Sachdev J, Ejadi S, Lim HY, Brenner AJ, Park K, Lee JL, Kim TY, et al. Phase 1 study of MRX34, a liposomal miR-34a mimic, in patients with advanced solid tumours. Br J Cancer. 2020;122(11):1630–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. van Zandwijk N, Pavlakis N, Kao SC, Linton A, Boyer MJ, Clarke S, Huynh Y, Chrzanowska A, Fulham MJ, Bailey DL, et al. Safety and activity of microRNA-loaded minicells in patients with recurrent malignant pleural mesothelioma: a first-in-man, phase 1, open-label, dose-escalation study. Lancet Oncol. 2017;18(10):1386–96.

    Article  PubMed  Google Scholar 

  147. Querfeld C, Foss FM, Kim YH, Pinter-Brown L, William BM, Porcu P, Pacheco T, Haverkos BM, DeSimone J, Guitart J, et al. Phase 1 trial of Cobomarsen, an inhibitor of Mir-155, in cutaneous T cell lymphoma. Blood. 2018;132(Supplement 1):2903–2903.

    Article  Google Scholar 

  148. Medarova Z, Robertson N, Ghosh S, Duggan S, Varkaris A. Initial clinical experience with the first-in-class anti-metastasis therapeutic TTX-MC138. J Clin Oncol. 2024;42(16suppl):e15072–15072.

    Article  Google Scholar 

  149. What will it. Take to get MiRNA therapies to market? Nat Biotechnol. 2024;42(11):1623–4.

    Article  Google Scholar 

  150. Sand M, Gambichler T, Sand D, Altmeyer P, Stuecker M, Bechara FG. Immunohistochemical expression patterns of the microRNA-processing enzyme Dicer in cutaneous malignant melanomas, benign melanocytic nevi and dysplastic melanocytic nevi. Eur J Dermatol. 2011;21(1):18–21.

    Article  PubMed  Google Scholar 

  151. Sand M, Skrygan M, Georgas D, Sand D, Gambichler T, Altmeyer P, Bechara FG. The MiRNA machinery in primary cutaneous malignant melanoma, cutaneous malignant melanoma metastases and benign melanocytic nevi. Cell Tissue Res. 2012;350(1):119–26.

    Article  CAS  PubMed  Google Scholar 

  152. Sand M, Skrygan M, Sand D, Georgas D, Gambichler T, Hahn SA, Altmeyer P, Bechara FG. Comparative microarray analysis of MicroRNA expression profiles in primary cutaneous malignant melanoma, cutaneous malignant melanoma metastases, and benign melanocytic nevi. Cell Tissue Res. 2013;351(1):85–98.

    Article  CAS  PubMed  Google Scholar 

  153. Babayan A, Neumann MHD, Herdean A, Shaffer JM, Janning M, Kobus F, Loges S, Di Pasquale F, Kubista M, Schlumpberger M et al. Multicenter evaluation of independent High-Throughput and RT-qPCR technologies for the development of analytical workflows for Circulating MiRNA analysis. Cancers (Basel) 2020, 12(5).

  154. Foye C, Yan IK, David W, Shukla N, Habboush Y, Chase L, Ryland K, Kesari V, Patel T. Comparison of MiRNA quantitation by Nanostring in serum and plasma samples. PLoS ONE. 2017;12(12):e0189165.

    Article  PubMed  PubMed Central  Google Scholar 

  155. Androvic P, Benesova S, Rohlova E, Kubista M, Valihrach L. Small RNA-Sequencing for analysis of Circulating MiRNAs: benchmark study. J Mol Diagn. 2022;24(4):386–94.

    Article  CAS  PubMed  Google Scholar 

  156. Satake E, Krolewski B, Kobayashi H, Md Dom ZI, Ricca J, Wilson JM, Hoon DS, Duffin KL, Pezzolesi MG, Krolewski AS. Preanalytical considerations in quantifying Circulating MiRNAs that predict end-stage kidney disease in diabetes. JCI Insight 2024, 9(12).

  157. Chan SF, Cheng H, Goh KK, Zou R. Preanalytic methodological considerations and sample quality control of Circulating MiRNAs. J Mol Diagn. 2023;25(7):438–53.

    Article  CAS  PubMed  Google Scholar 

  158. Zendjabil M. Preanalytical, analytical and postanalytical considerations in Circulating MicroRNAs measurement. Biochem Med (Zagreb). 2024;34(2):020501.

    Article  PubMed  Google Scholar 

  159. Greytak SR, Engel KB, Hoon DSB, Elias KM, Lockwood CM, Guan P, Moore HM. Evidence-based procedures to improve the reliability of Circulating MiRNA biomarker assays. Clin Chem Lab Med. 2024;62(1):60–6.

    Article  CAS  PubMed  Google Scholar 

  160. Felekkis K, Papaneophytou C. The Circulating biomarkers league: combining MiRNAs with Cell-Free DNAs and proteins. Int J Mol Sci 2024, 25(6).

  161. Grasedieck S, Schöler N, Bommer M, Niess JH, Tumani H, Rouhi A, Bloehdorn J, Liebisch P, Mertens D, Döhner H, et al. Impact of serum storage conditions on MicroRNA stability. Leukemia. 2012;26(11):2414–6.

    Article  CAS  PubMed  Google Scholar 

  162. Sourvinou IS, Markou A, Lianidou ES. Quantification of Circulating MiRNAs in plasma: effect of preanalytical and analytical parameters on their isolation and stability. J Mol Diagn. 2013;15(6):827–34.

    Article  CAS  PubMed  Google Scholar 

  163. Mompeon A, Ortega-Paz L, Vidal-Gomez X, Costa TJ, Perez-Cremades D, Garcia-Blas S, Brugaletta S, Sanchis J, Sabate M, Novella S, et al. Disparate MiRNA expression in serum and plasma of patients with acute myocardial infarction: a systematic and paired comparative analysis. Sci Rep. 2020;10(1):5373.

    Article  PubMed  PubMed Central  Google Scholar 

  164. Blondal T, Jensby Nielsen S, Baker A, Andreasen D, Mouritzen P, Wrang Teilum M, Dahlsveen IK. Assessing sample and MiRNA profile quality in serum and plasma or other biofluids. Methods. 2013;59(1):S1–6.

    Article  CAS  PubMed  Google Scholar 

  165. Kirschner MB, Edelman JJ, Kao SC, Vallely MP, van Zandwijk N, Reid G. The impact of hemolysis on Cell-Free MicroRNA biomarkers. Front Genet. 2013;4:94.

    CAS  PubMed  PubMed Central  Google Scholar 

  166. Kirschner MB, Kao SC, Edelman JJ, Armstrong NJ, Vallely MP, van Zandwijk N, Reid G. Haemolysis during sample Preparation alters MicroRNA content of plasma. PLoS ONE. 2011;6(9):e24145.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. Pizzamiglio S, Zanutto S, Ciniselli CM, Belfiore A, Bottelli S, Gariboldi M, Verderio P. A methodological procedure for evaluating the impact of hemolysis on Circulating MicroRNAs. Oncol Lett. 2017;13(1):315–20.

    Article  CAS  PubMed  Google Scholar 

  168. Shah JS, Soon PS, Marsh DJ. Comparison of methodologies to detect low levels of hemolysis in serum for accurate assessment of serum MicroRNAs. PLoS ONE. 2016;11(4):e0153200.

    Article  PubMed  PubMed Central  Google Scholar 

  169. Smith MD, Leemaqz SY, Jankovic-Karasoulos T, McAninch D, McCullough D, Breen J, Roberts CT, Pillman KA. Haemolysis detection in MicroRNA-Seq from clinical plasma samples. Genes (Basel) 2022, 13(7).

  170. Ainsztein AM, Brooks PJ, Dugan VG, Ganguly A, Guo M, Howcroft TK, Kelley CA, Kuo LS, Labosky PA, Lenzi R, et al. The NIH extracellular RNA communication consortium. J Extracell Vesicles. 2015;4:27493.

    Article  PubMed  Google Scholar 

  171. Liu Y, Min Z, Mo J, Ju Z, Chen J, Liang W, Zhang L, Li H, Chan GC, Wei Y, et al. ExomiRHub: A comprehensive database for hosting and analyzing human disease-related extracellular MicroRNA transcriptomics data. Comput Struct Biotechnol J. 2024;23:3104–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Mestdagh P, Hartmann N, Baeriswyl L, Andreasen D, Bernard N, Chen C, Cheo D, D’Andrade P, DeMayo M, Dennis L, et al. Evaluation of quantitative MiRNA expression platforms in the MicroRNA quality control (miRQC) study. Nat Methods. 2014;11(8):809–15.

    Article  CAS  PubMed  Google Scholar 

  173. Margue C, Reinsbach S, Philippidou D, Beaume N, Walters C, Schneider JG, Nashan D, Behrmann I, Kreis S. Comparison of a healthy mirnome with melanoma patient mirnomes: are MicroRNAs suitable serum biomarkers for cancer? Oncotarget. 2015;6(14):12110–27.

    Article  PubMed  PubMed Central  Google Scholar 

  174. Ferracin M, Lupini L, Salamon I, Saccenti E, Zanzi MV, Rocchi A, Da Ros L, Zagatti B, Musa G, Bassi C, et al. Absolute quantification of cell-free MicroRNAs in cancer patients. Oncotarget. 2015;6(16):14545–55.

    Article  PubMed  PubMed Central  Google Scholar 

  175. Ambros V, Bartel B, Bartel DP, Burge CB, Carrington JC, Chen X, Dreyfuss G, Eddy SR, Griffiths-Jones S, Marshall M, et al. A uniform system for MicroRNA annotation. RNA. 2003;9(3):277–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  176. Griffiths-Jones S, Grocock RJ, van Dongen S, Bateman A, Enright AJ. MiRBase: MicroRNA sequences, targets and gene nomenclature. Nucleic Acids Res. 2006;34(Database issue):D140–144.

    Article  CAS  PubMed  Google Scholar 

  177. Andersen CL, Jensen JL, Orntoft TF. Normalization of real-time quantitative reverse transcription-PCR data: a model-based variance Estimation approach to identify genes suited for normalization, applied to bladder and colon cancer data sets. Cancer Res. 2004;64(15):5245–50.

    Article  CAS  PubMed  Google Scholar 

  178. Vandesompele J, De Preter K, Pattyn F, Poppe B, Van Roy N, De Paepe A, Speleman F. Accurate normalization of real-time quantitative RT-PCR data by geometric averaging of multiple internal control genes. Genome Biol. 2002;3(7):RESEARCH0034.

    Article  PubMed  PubMed Central  Google Scholar 

  179. Pfaffl MW, Tichopad A, Prgomet C, Neuvians TP. Determination of stable housekeeping genes, differentially regulated target genes and sample integrity: BestKeeper–Excel-based tool using pair-wise correlations. Biotechnol Lett. 2004;26(6):509–15.

    Article  CAS  PubMed  Google Scholar 

  180. Pang Y, Wang C, Lu L, Wang C, Sun Z, Xiao R. Dual-SERS biosensor for one-step detection of MicroRNAs in exosome and residual plasma of blood samples for diagnosing pancreatic cancer. Biosens Bioelectron. 2019;130:204–13.

    Article  CAS  PubMed  Google Scholar 

  181. Agarwal V, Bell GW, Nam JW, Bartel DP. Predicting effective MicroRNA target sites in mammalian mRNAs. Elife 2015, 4.

  182. Caporali S, Amaro A, Levati L, Alvino E, Lacal PM, Mastroeni S, Ruffini F, Bonmassar L, Antonini Cappellini GC, Felli N, et al. miR-126-3p down-regulation contributes to Dabrafenib acquired resistance in melanoma by up-regulating ADAM9 and VEGF-A. J Exp Clin Cancer Res. 2019;38(1):272.

    Article  PubMed  PubMed Central  Google Scholar 

  183. Liu SM, Lu J, Lee HC, Chung FH, Ma N. miR-524-5p suppresses the growth of oncogenic BRAF melanoma by targeting BRAF and ERK2. Oncotarget. 2014;5(19):9444–59.

    Article  PubMed  PubMed Central  Google Scholar 

  184. Bezrookove V, Khan I, Bhattacharjee A, Fan J, Jones R, Sharma A, Nosrati M, Desprez PY, Salomonis N, Shi Y, et al. miR-876-3p is a tumor suppressor on 9p21 that is inactivated in melanoma and targets ERK. J Transl Med. 2024;22(1):758.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Funding

This work was supported by Macquarie University, Melanoma Institute Australia, the CLEARbridge Foundation and the National Health Medical Research Council of Australia project grants (NHMRC; grant 2012860, 2028055) and Medical Research Future Fund grant (MRFF 2009057). Biorender (https://www.biorender.com) was used to generate the figures.

Author information

Authors and Affiliations

Authors

Contributions

SYL, SB, RJD and HR designed the review, wrote, edited and revised the manuscript. SYL and HR prepared the figures. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Helen Rizos.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lim, S.Y., Boyd, S.C., Diefenbach, R.J. et al. Circulating MicroRNAs: functional biomarkers for melanoma prognosis and treatment. Mol Cancer 24, 99 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12943-025-02298-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12943-025-02298-7

Keywords