1. Introduction
Cutaneous melanoma is an aggressive cancer whose incidence is increasing constantly all over the world. Although surgical resection is an effective treatment for early-stage melanoma, natural prognosis of patients with stage IV disease is extremely poor, with a median survival rate of less than 1 year and a 5-year survival rate of less than 10% [
1].
In recent years, a remarkable advance in the therapy of metastatic melanoma has been achieved with the development of BRAF and MEK inhibitors (BRAFi, MEKi), approved for BRAF-mutant tumors, and monoclonal antibodies targeting T-lymphocyte-associated antigen 4 and programmed cell-death protein 1, approved for both BRAF-mutant and BRAF wild-type melanomas.
Monotherapy with BRAFi and MEKi yields objective response rates (ORR) of 50–60% and of 20–30%, respectively, and significantly prolongs progression-free survival (PFS) and overall survival (OS) with respect to dacarbazine chemotherapy [
2,
3]. Better results are achieved with the combination of BRAFi + MEKi—nowadays the standard-of-care of BRAF targeted approach—with ORR of 65–75% and PFS and OS rates of 15–22% and 30–38%, respectively, at 5 years [
4].
Although BRAFi and the combination of BRAFi + MEKi induce undoubtedly high ORR, a non-negligible percentage of patients shows primary resistance. Moreover, long-term efficacy of therapy is limited by the development of drug resistance in nearly all patients [
5,
6]. Novel therapeutic approaches able to improve response to BRAFi and MEKi and to mitigate or overcome acquired resistance, as well as biomarkers able to better predict patients’ response to therapy and survival outcomes, are therefore urgently needed.
MicroRNAs (miRNAs) are a class of small non-coding RNAs that negatively regulate gene expression at the post-transcriptional level by base paring to—usually—the 3′untraslated region (UTR) of target mRNAs [
7]. They are involved in numerous cellular processes, including apoptosis, proliferation, differentiation, and metabolism [
7]. Each miRNA can target numerous transcripts, whereas different miRNAs can converge on a single mRNA [
7]. Therefore, miRNAs can regulate a large fraction of protein-coding genes.
Aberrant expression of miRNAs has been demonstrated in a variety of human cancers, where miRNAs can operate as oncogenes or tumor suppressor genes [
8,
9]. In the clinical setting, tumor miRNA signatures have been correlated with patients’ outcome and response to therapy in several malignancies [
10,
11,
12]. miRNAs appear, therefore, to have both diagnostic and prognostic significance and to potentially constitute novel targets and therapeutic agents for cancer treatment [
12,
13,
14]. Tumor-derived miRNAs can also be detected in plasma/serum of cancer patients—associated with proteins, lipoproteins or included in extracellular vesicles (EVs)—and accumulating experimental evidence points out that circulating cell-free miRNAs (cf-miRNAs) can provide a non-invasive strategy for cancer diagnosis and prognosis, and for predicting and/or monitoring patients’ responses to therapy [
15,
16].
In melanoma, aberrant expression of miRNAs has been widely documented, and shown to affect several well-known pathways involved in the regulation of cell proliferation, invasiveness and survival, as well as to contribute to drug resistance [
17,
18,
19,
20,
21,
22,
23,
24,
25,
26,
27,
28,
29]. Moreover, the potential value of single cf-miRNAs or cf-miRNA signatures as biomarkers for melanoma diagnosis, staging, risk of recurrence and survival prognosis has been highlighted by numerous studies [
26,
28,
30,
31]. On the other hand, to the best of our knowledge, the role of cf-miRNA as biomarkers of patients’ responses to targeted therapy has been addressed by a very limited number of investigations [
21,
32].
In the present study, we used next-generation sequencing (NGS) for profiling plasma cf-miRNAs at baseline and at progression in melanoma patients treated with BRAFi monotherapy or the combination of BRAFi + MEKi to identify cf-miRNAs associated with response to therapy. Selected cf-miRNAs were then subjected to validation by real-time quantitative RT-PCR and investigated for their potential value as biomarkers predictive of patients’ clinical outcomes.
4. Discussion
Circulating cf-miRNAs are emerging as valuable non-invasive biomarkers for cancer diagnosis and assessment of patients’ prognosis and response to therapy [
15,
16,
26,
28]. In this regard, most of the studies performed so far on circulating cf-miRNAs in melanoma have been focused on their role as potential diagnostic and prognostic biomarkers in treatment-naïve patients [
30,
31], while few investigations have been performed to assess the association of cf-miRNA expression patterns with clinical outcomes in patients subjected to immunotherapy [
48,
49] or targeted therapy [
21,
32].
In the present study, using an NGS approach to profile cf-miRNAs in a cohort of 33 patients, we identified a set of cf-miRNAs potentially linked to resistance to therapy with BRAFi and MEKi, as they were DE at baseline between patients who had responded to treatment and patients who had not, and/or at disease progression with respect to baseline, in patients who had initially achieved an objective clinical response. Among those cf-miRNAs, baseline levels of a more restricted set also showed a good ability in discriminating Rs and NRs and/or a significant association with the duration of response to treatment, suggesting their potential utility in the clinical setting. We considered miR-1246, miR-92b-3p, and miR485-3p interesting for further studies, and therefore determined their plasma level at T0 and TP by qRT-PCR in the initial cohort of 33 patients and in an additional 24 patients. The results of qRT-PCR assays were consistent with small RNA-seq data analysis for miR-1246 and miR-485-3p, but not for miR-92b-3p, which therefore was not further investigated.
A large body of experimental evidence indicates that miR-1246 acts as an oncomiR in most, although not all, types of cancer, and that its expression in the tumor tissue and/or in patients’ plasma/serum can have a diagnostic and prognostic value [
50]. Indeed, with respect to normal adjacent tissue, overexpression of miR-1246 has been described in breast, colorectal, lung and ovarian cancer, as well as in hepatocellular and oral squamous cell carcinoma [
50,
51]. Moreover, increased levels of miR-1246 have been detected in plasma/serum of patients with those types of cancer as compared with healthy controls [
50]. In patients with colorectal, lung, esophageal and breast cancer and hepatocellular and oral squamous cell carcinoma, worse disease-free survival and/or OS were also found to be associated with high miR-1246 expression in the tumor tissue and/or in plasma/serum [
50,
51]. An elevated level of circulating miR-1246 was also demonstrated to be a marker of therapy resistance in colorectal and breast cancer [
52,
53]. On the other hand, decreased expression of miR-1246 has been found in prostate [
54] and renal cell carcinoma tissues [
55], suggesting a tumor suppressor function of miR-1246 in these types of neoplasias. Notably, both upregulation [
56] and downregulation [
57] of miR-1246 have been reported in cervical carcinoma specimens.
Numerous functional studies have been performed in cell lines derived from different types of tumors to assess the impact of miR-1246 overexpression or silencing on proliferation, survival, invasiveness and chemoresistance, and to identify the most relevant targets involved in the observed effects. For instance, regarding the oncogenic function of miR-1246, Wang et al. [
58] demonstrated that this miRNA promoted proliferation and invasiveness of colorectal cancer (CRC) cell lines and protected them from apoptosis through negative regulation of the
CCNG2 gene, which encodes cycling G2, an atypical cyclin that induces cell cycle arrest and is frequently downregulated in tumors. miR-1246-mediated downregulation of cycling G2 expression was also demonstrated to increase the stemness-like properties and chemoresistance of pancreatic and oral cancer cells [
59,
60], as well as to enhanced proliferation, invasiveness and drug resistance of breast cancer cells [
61]. In vitro studies addressing the role of miR-1246 in the crosstalk between cancer cells and the tumor microenviroment demonstrated that this miRNA can shuttle between CRC cells and fibroblasts, promoting migration of the tumor cells—via activation of the Wnt/b-catenin signaling pathway—and transdifferentiation of fibroblasts into cancer-associated fibroblasts that support tumor progression [
62]. Similarly, Cooks et al. [
63] demonstrated that CRC cells with specific gain of function mutations in p53 can reprogram neighboring macrophages into a tumor-promoting state by releasing miR-1246-enriched exosomes. Several other target genes of miR-1246 involved in its oncogenic activity have been experimentally identified, including, among others,
GSK3B (Glycogen Synthase Kinase 3 Beta) in lung cancer cells,
SPRED2 (Sprouty Related EVH1 Domain Containing 2) in CRC cells,
CADM1 (Cell Adhesion Molecule 1) in hepatocellular carcinoma cells [
50],
NFE2L3 (NFE2-like bZIP transcription factor 3) in breast cancer cells [
51].
A limited number of studies have also reported inhibitory effects of miR-1246 on tumor cell growth and/or invasiveness. In renal carcinoma cell lines, miR-1246 was shown to target
CXCR4 (C-X-C Motif chemokine receptor 4) leading to impairment of proliferation and migration [
55], while in prostate cancer cells,
CDH2 (Cadherin 2) and
VIM (Vimentin) were identified as direct miR-1246 target genes involved in its suppression of epithelial to mesenchimal transition (EMT) [
54]. Controversial results have instead been reported in cervical cancer cells. Indeed, Chen and collaborators demonstrated that miR-1246 suppressed the
THBS2 (Thrombospondin 2) gene and positively modulated cell proliferation, migration and invasion [
64], whereas inhibitory effects of miR-1246 on cervical cancer cell invasiveness were reported by Yang et al. [
57].
Compared to miR-1246, fewer investigations have addressed the role of miR-485-3p in cancer, but, with few exceptions [
65,
66,
67], it appears to function as a tumor-suppressor miRNA. Its expression in tumor tissue was found to be downregulated in prostate cancer [
68,
69], glioblastoma [
70], breast and colorectal cancer [
71,
72,
73,
74], osteosarcoma [
75] and renal carcinoma [
76]. Interestingly, in some of those studies, the reduced expression of miR-485-3p was linked to upregulation of a long non-coding RNA (LncRNA) [
75] or a circular RNA [
72,
74,
76] able to sponge the miRNA, thus leading to enhanced expression of target genes promoting tumor cell proliferation, survival and invasiveness. In the circulation, lower levels of miR-485-3p were detected in serum and serum exosome of glioblastoma patients as compared with healthy subjects [
77,
78]. Moreover, low pre-surgery levels of miR-485-3p were associated with shorter PFS and OS in glioma patients receiving radiotherapy plus chemotherapy after surgery [
77].
As for miR-1246, functional studies have been performed in cancer cell lines to identify the molecular mechanisms involved in the biological activity of miR-485-3p. Several targets of this miRNA have been experimentally validated, and their downregulation has been implicated in the inhibitory effects exerted by the miRNA on tumor cell proliferation, survival and invasiveness. For instance, in prostate cancer cells, miR-485-3p was shown to inhibit proliferation, migration and invasion by targeting the
TGFBR2 (Transforming Growth Factor Beta Receptor 2) gene, a key regulator of the TGF-β signaling pathway [
69], while in glioblastoma cells, the inhibitory effects of miR-485-3p on proliferation and migration were linked to downregulation of the
RNF135 (Ring Finger Protein 135) gene and impairment of the ERK1/2 signaling pathway [
70]. In breast cancer cells, miR-485-3p was found to suppress migration, invasion and mitochondrial respiration by inhibiting the expression of the
PGC-1A gene, which encodes a transcriptional coactivator that regulates genes involved in energy metabolism [
71]. Other target genes whose negative modulation by miR-485-3p has been shown to impair tumor growth and metastasis include, among others,
ZEB1 (Zinc Finger E-box Binding Homeobox 1) and
BIRC5 (Baculoviral IAP Repeat Containing 5) in breast cancer cells [
72,
79],
MELK (Maternal Embryonic Leucine Zipper Kinase) and
JAK2 (Janus Kinase 2) in CRC cells [
74,
80],
MET and
AKT3, in osteosarcoma cells [
75].
An oncogenic function of miR-485-3p has also been reported. Specifically, miR-485-3p was found to be upregulated in hepatocellular carcinoma, and to support tumor cell proliferation and survival in vitro and tumor growth and metastasis in mice by targeting the
MAT1A gene, which encodes the α1 catalytic subunit of methionine adenosyltransferase [
65]. In hepatocellular carcinoma, miR-485-3p was shown to also promote proliferation and invasiveness by downregulation of
NTRK3, which codes for the neurotrophic tyrosine kinase receptor type 3 [
67]. Moreover, suppression of this target gene was demonstrated to underlie the tumor-promoting activity of miR-485-3p in gastric cancer [
81].
Few data are presently available regarding the expression and function of miR-1246 and miR-485-3p in melanoma. Armand-Labit et al. [
82] reported that miR-1246 plasma levels were significantly higher in metastatic melanoma patients than in healthy controls, and confirmed the expression of this miRNA in melanoma metastases. Furthermore, the authors showed that plasma levels of miR-1246 in combination with those of miR-185 could differentiate patients from healthy controls with elevated accuracy. Increased levels of miR-1246 were also observed by Torii et al. [
41] in serum EVs derived from melanoma patients as compared with those isolated from healthy controls. Interestingly, the authors demonstrated that miR-1246 contained in EVs derived from a highly metastatic melanoma cell line, as well as miR-1246 mimics, were able to increase resistance to 5-fluorouracil in endothelial cells. Enhanced expression of miR-1246 in melanoma specimens with respect to normal tissue was recently reported by Yu et al. [
39], who in addition demonstrated that miR-1246 promoted melanoma cell proliferation, survival and invasiveness by targeting
FOXA2, a gene with a tumor-suppressor function in several types of cancer, including melanoma [
83,
84,
85,
86]. Finally, miR-1246 was found to be upregulated in melanoma cell lines resistant to the BRAFi PLX4720 [
40]. Regarding miR-485-3p, there is only a very recent study by Huo et al. [
47], who reported over-expression of the LncRNA MIR155HG and downregulation of miR-485-3p in melanoma specimens, and demonstrated that the LncRNA acts as a molecular sponge of miR-485-3p, leading to increased expression of its target gene
PSIP1. This gene is upregulated in several cancers and involved in tumor aggressiveness and chemoresistance [
87,
88,
89,
90]. Overall, those studies suggest that in melanoma, miR-1246 and miR-485-3p can have an oncogenic and a tumor-suppressor function, respectively.
In the present investigation, we show for the first time that baseline plasma levels of miR-1246 and miR-485-3p could represent valuable and non-invasive biomarkers to predict clinical response and prognosis in melanoma patients treated with BRAFi and MEKi. Indeed, miR-1246 and miR-485-3p baseline (i.e., T0) plasma levels were significantly higher and lower, respectively, in the group of NRs as compared with the group of Rs, and a trend toward an increase in miR-1246 and a decrease in miR-485-3p was observed in the latter group of patients at the development of secondary resistance (i.e., TP). Both baseline miR-1246 levels and the miR-1246/miR-485-3p ratio displayed a good ability to discriminate between Rs and NRs. Moreover, Kaplan–Meier curves showed that either miR-1246 plasma levels ≥ 8.637 or miR-485-3p plasma levels < 0.013 were associated with poorer PFS and OS, while univariate Cox regression analysis evidenced a more than two-fold higher risk of short-term progression and mortality for the two groups of patients showing those cf-miRNA levels. Accordingly, when the different combinations of miR-1246 and miR-485-3p were considered, the group of patients with favourable levels of both cf-miRNAs showed the best PFS and OS. Notably, the miR-1246/miR-485-3p ratio also appeared to be a useful prognostic parameter for PFS and OS. Indeed, an miR-1246/miR-485-3p ratio ≥ 345.493 was associated with significantly shorter PFS and OS and higher risk of progression and mortality. Altogether, our results are in agreement with previous studies that suggest an oncogenic and tumor-suppressor function in melanoma for miR-1246 and miR-485-3p, respectively.
Our findings also appear to be of clinical relevance. Indeed, both targeted therapy and immunotherapy with immune checkpoint inhibitors are possible therapeutic options for patients with BRAF-mutant melanoma, and the availability of novel biomarkers able to predict patients’ responses to therapy and to estimate their expectancy of PFS and OS may potentially help clinicians choose the optimal therapeutic protocol. The potential clinical utility of assessing baseline plasma levels of miR-1246 and miR-485-3p is also strengthened by the finding that miR-485-3p and the miR-1246/miR-485-3p ratio remained independently associated with PFS after adjusting for sex and sLDH in multivariate analysis. After adjusting for sex and sLDH, patients with a miR-1246/miR-485-3p ratio ≥ 345.493 showed a two-fold increased risk of mortality, but the statistical significance was not reached, most probably for the limited size of our cohort of patients. On the other hand, the stratified analysis by sLDH performed for miR-1246 levels did not evidence any significant association between miR-1246 and PFS or OS for patients with either normal or elevated sLDH, further highlighting that the simultaneous determination of miR-1246 and miR-485-3p can provide a better prognostication of patients’ outcomes.
LDH, which catalyzes the conversion of pyruvate to lactate, is a key enzyme of the glycolytic pathway and plays a central role in the Warburg effect, namely the elevated aerobic glycolysis that is considered a hallmark of cancer [
91]. Accordingly, numerous studies in different types of cancer have demonstrated the importance of LDH in tumor growth and malignant behavior [
91]. LDH can be released into blood stream upon cell damage, and in cancer patients sLDH is considered a biomarker linked to tumor burden and aggressiveness [
91,
92]. Elevated sLDH represents an independent predictor of poor outcome in patients with stage IV melanoma, and presently, sLDH is the only circulating biomarker incorporated by the AJCC in melanoma staging [
1]. High levels of sLDH are also a predictor of inferior OS and PFS in melanoma patients receiving BRAFi with or without MEKi [
93,
94]. These latter findings were confirmed in our study, which also evidenced an interaction effect between miR-1246 and sLDH for PFS and OS, most likely because plasma levels of this miRNA can be an expression of tumor burden and biological aggressiveness, as reported for sLDH. It is, however, noteworthy that although baseline sLDH is a strong biomarker of PSF and OS in patients receiving targeted therapy, in contrast to circulating miR-1246 and the miR-1246/miR-485-3p ratio, it does not appear useful for predicting patient response to treatment. In this regard, the analysis of pooled data derived from three phase 3 clinical trials of dabrafenib + trametinib in melanoma patients evidenced that, although inferior with respect to patients with normal sLDH levels, the response rate was also elevated (about 50%) in patients with high sLDH levels [
95]. Similarly, a response rate of 63% was reported by a large multicentric study in melanoma patients with elevated sLDH treated with targeted therapy [
96]. Consistent with these findings, we did not find any association between baseline sLDH and response to therapy in our cohort of melanoma patients.
A previous study by Svedman et al. [
32] evaluated the levels of 372 miRNAs in plasma EVs isolated before and during treatment from melanoma patients receiving therapy with BRAFi alone or in combination with MEKi. Twenty patients who had achieved PR or SD constituted the group of “disease control”, whereas eight patients progressing on therapy without any previous response constituted the group of NRs. The authors observed that increased levels of let-7g-5p during the course of treatment were associated with better disease control, and that patients displaying high levels of miR-497-5p during therapy had a longer PFS; but the authors did not find any association between pre-treatment EV miRNA levels and response to therapy. Several differences exist between the study of Svedman and collaborators and our investigation that could explain why the former did not identify pre-treatment miRNA levels associated with patients’ response to therapy. We performed cf-miRNA profiling using RNA extracted from total plasma samples, and therefore, in addition to miRNAs included in EVs, those associated with proteins and lipoproteins were also detected, increasing the possibility of identifying a clinically relevant cf-miRNA. Notably, miR-1246 was not present in the 372-miRNA set analyzed by Svedman and collaborators. Furthermore, we classified patients as R and NRs, including in the former group those who had achieved CR and PR, and in the latter group those who had shown SD or PD as their best clinical response, according to RECIST 1.1 criteria. This different classification of patients may have also favoured the identification of cf-miRNAs baseline levels associated with patient response to therapy.
The potential of two cf-miRNAs, namely miR-199b-5p and miR-4488, as biomarkers of melanoma patients’ resistance to targeted therapy was also investigated by Fattore et al. [
21] in a cohort of 25 patients treated with vemurafenib or the combination of vemurafenib + cobimetinib or encorafenib + binimetinib. Consistent with their findings in melanoma cell lines sensitive or resistant to targeted therapy, and in melanoma specimens collected before the start of treatment and at progression, the authors found that miR-199b-5p and miR-4488 plasma levels were downregulated and upregulated, respectively, at disease progression as compared to baseline, and showed a good ability to discriminate pre-treatment samples from progression samples. According to small RNA-seq data analysis, these two miRNAs were not DE at TP vs. T0 in our cohort of Rs, but it is possible that this result depends on the filtering procedures and statistical analyses applied to small RNA-seq row data. Moreover, in contrast with the study of Fattore and collaborators, most of our patients were subjected to dabrafenib or dabrafenib + trametinib, and it cannot be excluded that the changes occurring in cf-miRNA expression patterns from pre-treatment to progression may be related to the type of BRAFi and MEKi received from the patients.
Interestingly, in a number of studies [
97,
98,
99], miR-1246 in serum, plasma or exosomes was shown to be derived from the processing of the RNU2-1 transcript, a small nuclear RNA, and not from the
MIR1246 gene through to the canonical pathway of miRNA biogenesis. Functionality of miR-1246 derived from RNU2-1 transcript was confirmed [
99], and therefore, independently of its origin, miR-1246 remains an important miRNA involved in cancer. Future studies are required to ascertain whether circulating miR-1246 in our cohort of patients derives from the processing of RNU2-1 transcript.
We are aware that our study has some limitations. Treatment with BRAFi and MEKi in our cohort of melanoma patients was eterogenous, and it can not be excluded that this could have affected survival outcomes. Moreover, the study was retrospective, and included a relatively small number of patients. Although our results appear of clinical relevance, they need to be interpreted cautiously and validated in a larger prospective study.