Next Article in Journal
Advances in Personalized Oncology
Next Article in Special Issue
Barriers to T Cell Functionality in the Glioblastoma Microenvironment
Previous Article in Journal
Molecular Diagnostic Yield and Safety Profile of Ultrasound-Guided Lung Biopsies: A Cross-Sectional Study
Previous Article in Special Issue
Cancer Bioenergetics and Tumor Microenvironments—Enhancing Chemotherapeutics and Targeting Resistant Niches through Nanosystems
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Unveiling the Dynamic Interplay between Cancer Stem Cells and the Tumor Microenvironment in Melanoma: Implications for Novel Therapeutic Strategies

by
Patrizia Limonta
1,*,
Raffaella Chiaramonte
2 and
Lavinia Casati
2,*
1
Department of Pharmacological and Biomolecular Sciences “R. Paoletti”, Università degli Studi di Milano, 20133 Milan, Italy
2
Department of Health Sciences, Università degli Studi di Milano, 20142 Milan, Italy
*
Authors to whom correspondence should be addressed.
Cancers 2024, 16(16), 2861; https://doi.org/10.3390/cancers16162861
Submission received: 28 June 2024 / Revised: 7 August 2024 / Accepted: 13 August 2024 / Published: 16 August 2024

Abstract

:

Simple Summary

Resistance to standard therapies represents a major challenge in melanoma treatment. This is mainly related to the high heterogeneity of this malignancy due to the presence of different types of cells in the tumor mass. Cancer stem cells (CSCs) are a small subset of cells endowed with self-renewal and tumorigenic capacity and the ability to escape anticancer therapies. Melanoma CSCs can be identified by the expression of biomarkers and intracellular pathways deeply involved in their malignant phenotype. Dissecting the molecular mechanisms underlying CSC biological functions might pave the way for novel therapeutic approaches. Herein, we highlight the intricate bidirectional communication between melanoma CSCs and their surrounding cells in the tumor microenvironment and discuss how this interplay might impact the processes of drug resistance and tumor relapse. We also address the potential benefits of CSC-targeted strategies, in combination with conventional drugs, for improving melanoma treatment strategies.

Abstract

Cutaneous melanoma still represents a significant health burden worldwide, being responsible for the majority of skin cancer deaths. Key advances in therapeutic strategies have significantly improved patient outcomes; however, most patients experience drug resistance and tumor relapse. Cancer stem cells (CSCs) are a small subpopulation of cells in different tumors, including melanoma, endowed with distinctive capacities of self-renewal and differentiation into bulk tumor cells. Melanoma CSCs are characterized by the expression of specific biomarkers and intracellular pathways; moreover, they play a pivotal role in tumor onset, progression and drug resistance. In recent years, great efforts have been made to dissect the molecular mechanisms underlying the protumor activities of melanoma CSCs to provide the basis for novel CSC-targeted therapies. Herein, we highlight the intricate crosstalk between melanoma CSCs and bystander cells in the tumor microenvironment (TME), including immune cells, endothelial cells and cancer-associated fibroblasts (CAFs), and its role in melanoma progression. Specifically, we discuss the peculiar capacities of melanoma CSCs to escape the host immune surveillance, to recruit immunosuppressive cells and to educate immune cells toward an immunosuppressive and protumor phenotype. We also address currently investigated CSC-targeted strategies that could pave the way for new promising therapeutic approaches for melanoma care.

1. Introduction

Malignant melanoma still represents the most serious type of skin cancer worldwide and the deadliest form of cutaneous tumors, accounting for up to 80% of cancer-related deaths among skin cancer patients [1]. Most melanomas are diagnosed in the early stage and can be effectively treated with surgery; on the other hand, late-stage tumors undergoing dissemination and metastasis are associated with a very poor prognosis [2,3].
In recent decades, gene mutation analysis has been increasingly utilized to improve diagnosis, classification, prognosis and risk stratification in different tumors, paving the way for targeted therapeutic interventions [4,5,6,7]. In melanoma, driver mutations of the BRAF and NRAS oncogenes are detected in about 40–50% and 10–20% of patients, respectively, leading to the overactivation of the mitogen-activated protein kinase (MAPK) signaling pathway (RAS/RAF/MEK) together with the parallel PI3K/Akt/mTOR cascade, both deeply involved in the processes of cell growth and survival. Additional driver mutations have been found to involve the neurofibromin 1 (NF1) and the KIT (1–2%) genes [8,9,10,11], while mutations associated with tumor development have been observed in the cyclin-dependent kinase inhibitor 2A (CDKN2A) gene, a well-known cell cycle regulator [12]. These observations led to the development of “targeted therapies” based on BRAF inhibitors (vemurafenib, dabrafenib, encorafenib) or MEK inhibitors (trametinib, cobimetinib, binimetinib), either alone or in combination; however, despite the high response rate and reduced toxicity of the combination treatment compared with the use of single compounds, most patients experience a low survival rate (5 years) and drug resistance within a few months [13,14,15,16,17,18,19].
Immunotherapy has emerged as a novel promising therapeutic strategy to trigger antitumoral immunity. In this context, immune checkpoint inhibitors were developed with the aim to improve the activity of the antitumor immune system. Specifically, the inhibitors of the lymphocyte-activation gene 3 (LAG3) transmembrane protein (relatlimab), the programmed cell death-1 (PD-1) receptor (nivolumab, pembrolizumab), the PD-1 ligand (PD-L1) (atezolizumab) and the cytotoxic T-lymphocyte antigen 4 (CTLA-4) (ipilimumab and tremelimumab) have been approved by the Food and Drug Administration (FDA) and are presently used in clinical practice. These drugs, either alone or in combination, have drastically improved melanoma clinical management; however, some limitations still remain due to their association with serious side effects and to the development of primary and acquired resistance [17,18,19,20,21,22,23,24].
The mechanisms underlying the development of resistance to standard-of-care treatments in melanoma have been extensively investigated in the past few years. In patients undergoing targeted therapy treatments, primary (intrinsic) resistance has been reported to be associated with the aberrant activation (ligand-induced dimerization or oligomerization) of receptor tyrosine kinases (RTKs), which in turn trigger a cascade of phosphorylation and, ultimately, the activation of the RAS/RAF/MEK/ERK and PI3K/Akt/mTOR pathways. Phosphatase and tensin homolog (PTEN) is a protein endowed with a peculiar tumor suppressive activity and the most important negative regulator of the PI3K/Akt oncogenic pathway. The increase in PI3K/Akt/mTOR signaling occurring in drug resistance may also be induced by the deletion or loss-of-function mutations of PTEN.
Acquired resistance, developing in initial responders to therapies, mostly results from the reactivation of the MAPK pathway and is associated with alterations in NRAS, BRAF, MEK and NF1 [25,26,27,28,29,30,31,32,33,34]. The RAF isoforms CRAF and ARAF are also frequently dysregulated, allowing mutated BRAF to be bypassed and leading to the reactivation of ERK [35,36,37]. Moreover, the Cancer Osaka thyroid (COT) kinase has been reported to be overexpressed in drug-resistant melanomas and to induce resistance to BRAF inhibition through the direct activation of the MEK/ERK signaling pathway [38]. Additional mechanisms involved in the development of acquired resistance include amplification or alternative splicing of the BRAF gene, alterations in miRNA expression, cell phenotype plasticity defined by the expression of the microphthalmia-associated transcription factor (MITF), activation of different signaling cascades such as the HIPPO/YAP/TAZ pathway and epigenetic regulation of DNA accessibility [39,40,41,42,43,44,45,46,47,48]. Primary and acquired resistance have also been reported to occur in patients challenged with immune checkpoint inhibitor therapies and found to be associated with different mechanisms: poor immunogenicity, impaired maturation of dendritic cells (DCs), altered expression levels of the immunosuppressive marker PD-L1, insufficient T cell activation, antigen-presenting cell dysfunction, loss-of-function mutations in genes involved in antitumor signals (interferon-γ, IFN-γ), melanoma cell dedifferentiation and phenotypic plasticity [20,23,47,49,50,51].
Melanoma is a tumor characterized by high heterogeneity and transcriptional plasticity [52,53,54]. Two models of carcinogenesis have been proposed to explain this heterogeneity. According to the stochastic model, all the tumor cells are biologically equivalent, with the same tumorigenic ability, and acquire different characteristics depending on intrinsic factors, such as genetic and epigenetic alterations and intracellular signaling pathways, and extrinsic factors from the tumor microenvironment (TME). In the hierarchical model, a tumor originates from a small subpopulation of slow-cycling “cancer stem cells” (CSCs) that reside at the top of the hierarchy. These cells acquire mutations but, like normal stem cells, show self-renewal ability as well as the potential to give rise to different progeny cells responsible for the formation of the tumor bulk population through asymmetric cell division [55,56,57,58]. This hierarchical model has been widely considered the most common model of tumor development. However, the recently identified high level of plasticity of tumor cells reconciles the two models and suggests that they may not be mutually exclusive, based on the observation that differentiated cancer cells can shift to a stem cell phenotype and, conversely, stem cells can be converted again into a non-stem cell state [59,60]. In line with these data, Quintana and coworkers reported that 25% of unselected patient-derived melanoma cells could generate the bulk tumor mass when inoculated in small numbers in nude mice. Since this percentage is higher than the small subpopulation of CSCs located into the tumor bulk, these authors conclude that also non-classical CSCs could generate tumors when inoculated in nude mice [61].
Growing bodies of evidence support that CSCs play key roles in tumor progression, treatment resistance and relapse in different tumors, such as malignant melanoma [34,62,63,64,65,66]. Notably, a reciprocal interplay has been observed between CSCs and their TME, which maintains or even promotes CSC survival and self-renewal, thus participating in tumor promotion, resistance and recurrence [67,68,69,70,71]. Based on these considerations, eradication of CSCs is presently considered a potentially promising strategy to overcome drug resistance in cancer. A deeper understanding of the molecular features and biological functions of CSCs might improve the therapeutical options for aggressive tumors, such as melanoma.
Herein, we provide an overview of the molecular and functional characteristics of melanoma CSCs, such as cell immunophenotypical markers, activated signaling pathways and biological roles in melanoma development and progression; the dynamic interplay between melanoma CSCs and their TME will be specifically discussed to shed light on how this interaction might impact tumor progression, resistance and recurrence. We also address currently investigated CSC-targeted strategies that could improve the therapeutic approaches for melanoma care.

2. Melanoma CSC Markers

The interest in the identification of melanoma CSCs prompted the search for specific markers. In vitro, CSCs can grow when cultured in low-adherence cell culture conditions in the presence of growth factors, such as fibroblast growth factor-2 (FGF-2) and epidermal growth factor (EGF) generating floating spheroid colonies (melanospheres) and express markers of epithelial-to-mesenchymal transition (EMT) associated with a highly invasive behavior; in vivo, the inoculation of CSCs in nude mice at limited dilutions has been widely reported to give rise to newly formed tumors sharing the characteristics of the primary tumor mass [63,65,72,73,74,75,76,77].
In addition, melanoma CSCs may be identified by the expression of specific stemness biomarkers, such as surface markers associated with melanocyte undifferentiated state or with drug resistance (ATP-binding cassette transporters involved in drug efflux out of the cells) and intracellular markers involved in drug metabolism (i.e., those belonging to the aldehyde dehydrogenase -ALDH- family). Embryonic stem cell markers, such as Sox2, Oct4 and Notch, have also been shown to be overexpressed in melanoma CSCs (Figure 1).
However, the specificity of most of these markers for CSCs remains a serious controversial issue. By means of in vitro and in vivo experiments, it has been shown that they are expressed in a large population of melanoma cells, being not limited to the CSC subpopulation, as well as in differentiated non-tumoral cells, such as normal melanocytes [63,77,78,79,80]. Alterations in specific intracellular signaling pathways have also been found to correlate with melanoma cell stemness (Figure 1).

2.1. Surface Markers

CD133 (prominin-1/AC133), a member of the pentaspan transmembrane glycoproteins, is considered a relevant surface marker for the identification of CSCs in different types of solid tumors. Specifically, high levels of CD133 were reported to be present in melanoma tissue biopsies when compared to nevi and to correlate with tumor growth [81,82]. In line with this observation, CD133+ melanoma cells are endowed with a high tumorigenic potential when inoculated in nude mice and display in vitro a peculiar metastatic behavior [82,83,84,85,86]. Moreover, CD133-dependent mechanisms are deeply involved in the development of melanoma resistance to standard therapies [34,64]. Mechanistically, the key role of CD133 in maintenance of stemness properties and drug resistance has been demonstrated to be mediated by the activation of the PI3K/Akt/mTOR intracellular signaling pathway [34,64,87]. However, in contrast to these observations, the suitability of CD133 as an appropriate marker for identifying and selecting melanoma CSCs is still a matter of debate [88,89,90]. Since CD133 expression is not limited to the surface of CSCs, distinct biological functions of this protein have been pointed out in cancer cells [91].
CD271 (p75 neurotrophin receptor), the low-affinity nerve growth factor receptor, is a member of the tumor necrosis factor receptor family: it is well known to be expressed in the neural crest throughout embryonic development. High expression levels of CD271 have also been widely detected in melanoma cells and shown to positively correlate with high tumorigenicity, metastatic behavior and drug resistance [92,93,94,95,96,97,98]. In line with these observations, CD271 has been shown to regulate phenotype switching of melanoma cells from a non-stem to a stem-like state [99,100,101,102]. However, also in this case, the reliability of this surface receptor as a suitable melanoma CSC marker is still controversial [59,63,78,80,103,104].
CD147, a transmembrane glycoprotein belonging to the immunoglobulin superfamily, has been reported to play a key role in melanoma initiation and progression toward its aggressive metastatic stage [105,106,107]. Very recently, Jiang and coworkers demonstrated that melanoma cells expressing high levels of CD147 are endowed with stemness features, such as strong sphere formation ability, migration, invasion and high tumorigenic capacity when inoculated into nude mice. Interestingly, the elevated expression of this marker is associated with the activation of the TGF-β1 (transforming growth factor-beta 1) and Notch1 (neurogenic locus notch homolog protein 1) intracellular signaling pathways [108]. However, as stated by these authors, additional studies are necessary to confirm the potential role of CD147 as a novel marker in melanoma treatments.
CD20 is a cell surface protein normally expressed on B cell surface. CD20 has been reported to be consistently expressed on melanoma CSCs and to positively correlate with stemness-related markers [83]. Moreover, it has also been identified in melanoma patient specimens [109].

2.2. Markers of Drug Resistance

The ATP-binding cassette (ABC) transporters are a superfamily of proteins widely known to facilitate the ATP-driven efflux of drugs across cellular membranes, being deeply involved in drug resistance [110,111]. Based on this, their presence has been evaluated at the plasma membrane level in different tumors, including melanoma, and specifically in their CSC subpopulation [63,84,112,113,114,115,116]. ABCB5 has been proposed as a reliable marker of melanoma aggressiveness, multidrug resistance and stemness [84,117,118,119]; it has also been found to induce peculiar modifications of the cellular metabolism in melanoma-initiating cell lines [120]. Interestingly, ABCB5 exists in two isoforms: ABCG5FL, a full transporter, and ABCB5β, a half transporter whose homodimer does not confer multidrug resistance. Very recently, Gerard et al. identified two novel heterodimers of ABCB5β in melanoma cell lines, ABCB5β/B6 and ABCB5β/B9, endowed with ATPase activity. However, as suggested by these authors, the physiological functions of these dimers in melanoma progression still remain to be investigated [121].
In contrast to these observations, Louphrasitthiphol et al. suggested that ABCB5 unlikely represents a marker of melanoma CSCs since its expression is also associated with melanoma cells exhibiting markers of differentiation. Specifically, MITF, the transcription factor known to sustain the expression of ABCB5, is highly expressed in differentiated melanoma cells and this correlates with high levels of ABCB5 [122]. Moreover, a low MITF/high Axl (a receptor tyrosine kinase) ratio was reported in a subpopulation of slowly proliferating melanoma cells endowed with invasive properties and drug resistance [123]. Thus, the low/high MITF ratio could have an essential role in the switch between dedifferentiated and differentiated melanoma cells. Based on these observations, further studies are needed to confirm the reliability of the MITF/ABCB5 axis as a marker of melanoma cell stemness.
ABCG2 is another ABC drug efflux transporter associated with drug resistance and stemness in different types of tumors, such as melanoma [82,124,125]. ABCG2 was found to be highly expressed, together with other stemness markers, in melanoma tissues and to correlate with the clinical features of patients and a worse prognosis [126]. Moreover, it has been shown that melanoma cells resistant to vemurafenib express high levels of ABCG2 and CD271 [127]. Interestingly, Miranda-Lorenzo and coworkers reported that, in epithelial cancer cells, the subpopulation of cells showing CSC traits is endowed with high autofluorescence levels. Specifically, this autofluorescence was shown to be related to the presence of vitamin B2 (riboflavin) in membrane-located cytoplasmic vesicles coated with ABCG2, supporting the hypothesis that autofluorescence might represent a reliable marker of cancer cell stemness [128]. In line with these observations, in our laboratory, we could demonstrate that melanoma cells endowed with autofluorescence properties and expressing high levels of ABCG2 are able to form tumorspheres, express intracellular stemness markers and generate the bulk tumor mass when inoculated at low dilutions in nude mice [78].

2.3. Intracellular Markers

CSCs can also be identified by the overexpression of markers located at the intracellular level, such as detoxifying enzymes and pluripotency transcription factors.
ALDH are enzymes critical for detoxifying different endogenous and exogenous substrates by oxidizing aldehydes to corresponding carboxylic acids. ALDH1 (ALDH isozyme 1), specifically the isoforms ALDH1A1 and ALDH1A3, have been reported to be highly expressed and even overactivated in melanoma CSCs and to correlate with different biological properties of tumor-initiating cells, such as tumorigenesis and drug resistance [129,130]. In line with these observations, Sarvi et al. demonstrated that, in melanoma-initiating cells, the anticancer compounds 5-nitrofurans could be metabolized by ALDH1A1/1A3 into their bioactive, cytotoxic metabolites, which in turn effectively eradicate the subpopulation of melanoma cells, which expresses high levels of ALDH1 [131]. Pharmacological approaches targeting ALDH isozymes are currently being explored with the aim of specifically targeting and eliminating CSCs in different tumors [132,133].
Accumulating evidence supports that embryonic transcription factors (Sox2, Oct4, Nanog) are present in melanoma CSCs where they work in concert to maintain cell stemness; moreover, their expression levels correlate with cell stemness features, such as pluripotency, tumor-initiating capacity and drug resistance [134,135,136,137,138,139]. Sox2 has been reported to positively regulate the self-renewal of cells in melanoma spheroids as well as of ALDH+ melanoma cells [140]. In melanoma cells, this transcription factor was also shown to promote a CSC-like phenotype associated with resistance to chemotherapy or targeted therapy by transactivating the expression of the CD24 and ABCC1 stem cell markers [141,142]. Similar observations were reported for the Oct4 transcription factor. Specifically, Cordaro and coworkers reported that melanoma cells made resistant to dabrafenib exhibit peculiar stem cell-like traits accompanied by the translocation of Oct4 from the cytoplasm into the nucleus and the increased expression of the stemness marker CD20 [143]. Oct4 expression was also found to correlate with worse prognosis and survival in melanoma tissues, suggesting that it might represent a useful biomarker in melanoma prognosis [144]. Last but not least, Nanog overexpression was observed to boost the metastatic potential of melanoma cells through the upregulation of genes involved in glucose transport and oxidative phosphorylation (OXPHOS) [145] and to promote immune resistance and stem-like properties in immune-refractory tumor cell types, including melanoma cells [146]. In contrast to this evidence, Khoo and colleagues recently reported that Nanog-overexpressing melanoma cells release small extracellular vesicles (EVs) endowed with a striking metastasis-suppressive behavior if compared to EVs derived from the original melanoma cell line [147]. Therefore, further studies are needed to establish the role of the Nanog transcription factor as a pluripotency marker in melanoma.

2.4. Intracellular Signaling Pathways

It is now well established that different developmental signaling pathways, particularly Wnt/β-catenin, Notch, Hedgehog and PI3K/Akt, are deeply involved in cancer onset, progression, development of drug resistance and, specifically, cell stemness [148,149,150,151,152,153,154,155,156]. Altogether, these pathways form a complex network of molecular interactions that cooperate to govern the CSC traits regulating self-renewal, survival, cell fate decisions and differentiation.

2.4.1. Wnt/β-Catenin Signaling

The canonical Wnt pathway, also known as the Wnt/β-catenin pathway, is triggered by the binding of the Wnt proteins to their specific membrane receptors in an autocrine or paracrine way. When Wnt ligands are absent, intracellular β-catenin is first phosphorylated by the intracellular β-catenin destruction complex and subsequently ubiquitinated by β-TrCP to be finally degraded at the proteasomal level. On the other hand, when Wnt proteins are present, they bind to specific membrane receptor complexes (Frizzled and LRP5/6 coreceptors), thus protecting β-catenin from degradation and allowing its translocation, facilitated by the APC/Axin/GSK-3 complex, into the nucleus where it binds to T cell factor/lymphatic enhancer factor (TCF/LEF) to promote the transcription of target genes, such as genes involved in cancer cell proliferation, survival, migration and stemness [157,158,159,160,161,162].
Wnt/β-catenin signaling has been found to play a significant role in regulating melanoma initiation and metastasis formation [163] and mediating the prometastatic activity of the CD133 stemness marker [164]. Recent studies pointed out that this intracellular pathway is also involved in the regulation of the biological and functional traits of melanoma CSCs. Wei and coworkers reported that, in melanoma cells, a downregulation of the ubiquitin–protein enzyme RNF128 correlates with EMT and the acquisition of stemness features through the activation of the Wnt pathway [165]. In line with these data, overexpression of the nucleolar protein NOP14 was found to suppress the biological functions and stemness traits of melanoma stem-like cells through the inactivation of the Wnt/β-catenin signaling [166]. Based on these observations, it has been proposed that targeting the Wnt/β-catenin pathway might represent a promising strategy for the effective eradication of CSCs and, therefore, for an effective treatment of melanoma patients [167,168,169].

2.4.2. Notch Signaling

The Notch signaling pathway plays a crucial role in cell fate determination during embryogenesis; it is also deeply involved in developing, progressing and maintaining CSCs in different tumors [148,155,170,171]. Two groups of transmembrane proteins contribute to the activity of this pathway: Notch receptors, Notch1-4, and Notch ligands, including Jagged (JAG) 1 and 2 and Delta-like (DLL) 1, 2, 3 and 4. Upon the binding of a membrane-bound Notch ligand to a Notch receptor on the surface of a neighboring cell, the receptor undergoes two cleavages: the first occurs in the extracellular domain and is mediated by the ADAM family of proteases, while the second is performed at the level of the intracellular domain by γ-secretase and leads to the release of the Notch intracellular domain (NICD) into the cytoplasm. NICD then translocates into the nucleus to promote the transcription of target genes, such as Hes and c-myc, involved in the mechanisms of tumor progression [148,172,173,174,175,176]. However, the potential role of the Notch pathway in CSCs as a tumor promoter or a tumor suppressor signal is still controversial and seems to be related to the cellular setting [177,178].
Lin and coworkers reported that Notch4 is highly expressed in melanoma CSCs and promotes their metastatic and invasive ability [179]. Moreover, it has been reported that Notch1 increases the expression of the CD133 stemness marker in melanoma cells through the activation of the MAPK pathway.
CD133 controls VEGF and matrix metalloproteinase (MMP) expression, thus promoting tumor growth and angiogenesis [180]. Notch1 was also recently shown to mediate the proangiogenic activity of ALDH1A1-overexpressing melanoma cells [181]. In line with these observations, it has been demonstrated that Notch3 plays a key role in the maintenance of melanoma CSC plasticity; knockdown of Notch3 leads to a downregulation of the stemness markers CD133 and CD271, a depletion of the melanoma CSC subpopulation and an attenuation of the proangiogenic activity [182].

2.4.3. Hedgehog Signaling

The Hedgehog (HH) signaling pathway is widely known to be deeply involved in embryogenesis, tissue homeostasis and regeneration; however, it has also been reported to be dysregulated in different types of cancers, as well as in melanoma [183,184,185]. The major components of this pathway include secreted HH proteins (Sonic, Indina, Desert), their cognate transmembrane receptor Patched (Ptch), the transmembrane protein Smoothened (Smo) and the three effector proteins Gli1-3. In the absence of the HH ligands, Ptch constitutively inhibits the activity of Smo; on the other hand, upon ligand binding, this inhibition is released and activated Smo triggers the nuclear translocation of the transcription factors Gli1-3, which in turn regulate the transcription of their target genes. Specifically, Gli1 and Gli2 are endowed with transcriptional activity while Gli3 mainly acts as a transcriptional repressor in the absence of ligands [186].
The role of HH signaling in the regulation of CSC traits and functions has been evidenced in different types of tumors [148,154,187,188]. Knockdown of Gli1 and Gli2 was found to restore drug sensitivity in vemurafenib-resistant melanoma cells [189]. Santini and coworkers observed that the HH/Gli pathway drives self-renewal and tumor initiation of ALDH+ melanoma CSCs. Moreover, in primary melanoma cells, the activation of this signaling cascade promotes the expression of Sox2, the transcription factor well known for its crucial role in self-renewal and tumorigenicity of melanoma CSCs [140,190]. In line with this observation, Horak et al. recently demonstrated that, in melanoma cells, activation of the HH pathway and Gli proteins directly promotes the expression of Slug, a hallmark factor of EMT as well as of cancer cell reprogramming into a stem-like phenotype [191].

2.4.4. PI3K/Akt Signaling

Dysregulation of the phosphatidylinositol 3-kinase (PI3K)/Akt signaling cascade has been widely observed in various human solid cancers, such as melanoma, and found to correlate with disease development and progression [192,193,194,195,196,197,198].
This pathway is a multistep process that is triggered by the binding of different extracellular ligands to transmembrane tyrosine kinase receptors, leading to the phosphorylation of the membrane-associated kinase PI3K. Once activated, PI3K catalyzes the conversion of the membrane lipid phosphatidylinositol (3,4)-bis-phosphate (PIP2) into PIP3, which, in turn, promotes the recruitment of the serine and threonine kinase Akt to the cell membrane and its subsequent phosphorylation by various kinases. Ultimately, Akt activates the mammalian target of rapamycin mTOR (specifically the mTORC1 complex), regulating the downstream effectors, which are deeply involved in cancer cell proliferation, survival, angiogenesis and drug resistance [199,200,201,202,203].
Accumulating evidence supports that the PI3K/Akt pathway is overactivated and plays a relevant functional role in CSCs [148,204]. In BRAFV600E+ melanoma cells, an aberrant activation of this intracellular pathway has been reported to mediate resistance to BRAF and MEK inhibitors and to support the acquisition of stem-like properties [205,206]. Corrales and coworkers demonstrated that, in melanoma cells, a decrease in sensitivity to MAPK/ERK inhibitors is accompanied by higher activity of the PI3K/Akt pathway, which is particularly evident in mesenchymal-like cells supporting their acquisition of stem-like features [207]. In line with these observations, it was reported that a CD133-dependent activation of PI3K/Akt/mTOR signaling is required for the maintenance of stemness traits and drug resistance in melanoma cells [64,87]. Vasculogenic mimicry (VM), the potential of cancer cells to differentiate into endothelial-like cells directly contributing to the “angiogenic switch” and promoting angiogenesis, was first observed in melanoma CSCs [180,208,209]. Notably, it has been reported that the PI3K/Akt pathway plays a pivotal role in the VM ability of melanoma CSCs [210,211].
Together, these observations strongly support that targeting the PI3K/Akt/mTOR pathway could represent a successful strategy to overcome therapy resistance while specifically suppressing CSCs and tumor angiogenesis in melanoma.

3. Cell-to-Cell Communication between Melanoma CSCs and the Tumor Microenvironment

A growing body of evidence strongly supports that a deep interplay exists between cancer cells and their surrounding TME. The TME encompasses different types of cells, such as immune cells, CAFs, endothelial cells, keratinocytes and adipocytes; other factors present in the tumor niche include extracellular matrix components, hypoxia and signaling molecules. Cancer cells, particularly melanoma cells, release molecular messages to surrounding cells to promote a tumor-sustaining microenvironment. On the other hand, the cancer microenvironment promotes tumor growth, progression, invasiveness, angiogenesis and drug resistance [69,212,213,214,215,216]. The molecular messages involved in this bidirectional communication involve growth factors, chemokines and cytokines, as well as EVs carrying a specific molecular cargo of mRNAs, miRNAs, DNA, proteins and lipids. Cells secrete EVs into the extracellular space to release their molecular cargo to near and far cells, thus affecting their biological features [217,218,219,220,221,222]. Notably, a complicated crosstalk also exists between CSCs and their surrounding cells and plays a pivotal role in CSC self-renewal, maintenance and resistance to therapies [69,70,71,223,224,225]. Herein, we specifically address the interplay between melanoma CSCs and their surrounding cells in the TME and its role as a potential target in melanoma therapies.

3.1. Melanoma CSC–Immune Cell Interplay

Based on the remarkable improvements in immunotherapy for cutaneous melanoma patients, the interplay between melanoma cells and infiltrated immune cells has recently gained increasing attention [49,212,214,226,227,228,229]. As discussed above, primary and acquired resistance frequently occurs in patients challenged with immune checkpoint inhibitor therapies and accumulating evidence has recently highlighted the involvement of CSCs in this undesired event. Specifically, melanoma CSCs have been widely shown to possess a striking ability to escape host immune surveillance (immune evasion) and, concurrently, to eliminate antitumor immune cells or to selectively recruit immune cells endowed with peculiar immunosuppressive and tumor-promoting features [63,230,231]. A strict correlation has been observed between melanoma CSCs and tumor-infiltrating immune cells [232,233] (Figure 2).

3.1.1. Interactions with Adaptive Immune Cells

T lymphocytes are the main component of the adaptive immune system, characterized by the ability to gain functional/effector abilities according to the immunological context.
The key role of tumor-associated antigens (TAAs) in eliciting melanoma antitumor immunity is well recognized. These TAAs include melanoma antigen recognized by T cells (MART-1), gp100, tyrosinase, tyrosine-related protein 1/2 (TRP1/2) but also cancer–testis antigens, such as NY-ESO1, MAGE-A3 and MAGE-A4. The immunogenicity of these antigens is supported by the observation that their cytotoxic T cell-mediated targeting results in a significant decrease in tumor burden in mice and melanoma patients [234,235,236]. However, an inadequate adaptive immune system response is frequent in melanoma, suggesting the presence of a subpopulation of dedifferentiated tumor cells similar to CSCs characterized by a low immunogenic profile and an immunosuppressive behavior deeply associated with tumor growth and progression [226,230]. Consistently, it has been reported that ABCB5+ and CD271+ melanoma cells express very low levels of the differentiation marker MART-1 as well as of cancer–testis antigens [92,237,238]. Landsberg and coworkers demonstrated that immunotherapy-resistant melanoma cells xenografted in nude mice models undergo a dedifferentiation process characterized by the concurrent gain of the stemness marker nerve growth factor receptor (NGFR) and loss of TAAs [239]. Similar results were also reported in in vitro and in clinical studies [240,241]. Interestingly, Huang et al. reported that inhibiting the phosphorylation (i.e., activation) of eukaryotic translation initiation factor 4E (eIF4E) in a PTEN KO transgenic melanoma mouse model prevents tumor cell dedifferentiation, resulting in increased expression of TAAs (MART-1 and gp100), thus facilitating their recognition and eradication by cytotoxic T cells. Moreover, phopsho-eIF4E levels were found to correlate negatively with MART-1 expression in melanoma biopsies [242]. Taken together, these data collectively support that differentiated melanoma cells expressing TAAs can be specifically eradicated by cytotoxic T cells; on the other hand, the loss of expression of these antigens in dedifferentiated CSCs is strictly associated with the activation of immune escape. However, it must be underlined that some contradictory results have been reported [239,243] and they have been explained by the possible involvement of different types of TAAs in the T cell-induced eradication of melanoma CSCs [228,244].
It is now well accepted that, in spite of a correct expression of immunogenic antigens, the immune evasion ability of CSCs can be associated with defective antigen presentation or processing. In addition to the reported decreased expression of TAA, it has been recently shown that melanoma CSCs express low levels of molecules involved in antigen presentation, such as major T cell histocompatibility (MHC) class I proteins [237,238]. So, even if TAAs are normally expressed, the absence of functional costimulatory MHC class I proteins hinders cytotoxic T cell activation, thus contributing to immune evasion. Similarly, an impairment of T cell activity can be triggered by the presence of a negative costimulation. For instance, negative modulators of T cell functions, such as PD-L1, PD-1 and CD86, have been reported to be highly expressed in melanoma CSCs, mediating their immune escape behavior. Thus, the low immunogenic features of melanoma CSCs are strictly correlated to a repression of differentiation antigens and costimulatory antigen presentation molecules as well as to an overactivation of negative costimulatory machinery.
In recent years, accumulated evidence has revealed that different melanoma CSC markers are also directly involved in the mechanisms of tumor cell resistance to T cell cytotoxicity. Tripartite motif-containing 28 (TRIM28) is a well-known marker of cancer cell stemness being deeply involved in the maintenance of stem cell renewal [245]. Czerwinska and coworkers reported that melanoma tumors expressing high levels of TRIM28 are significantly depleted with infiltrating cytotoxic T cells, as well as B cells and helper T cells, and suggest that this molecule might be considered as a good predictor of a “stemness high/immune low” melanoma phenotype [246]. It has been shown that melanoma cells resistant to MART-1 T cells express high levels of the stemness marker NGFR. Knockdown of this receptor sensitizes melanoma cells to MART-1 T cells, supporting its specific role in melanoma CSC resistance to T cell cytotoxic activity [241]. Kim and coworkers reported that Nanog triggers the upregulation of the tubule-associated protein 1 light chain 3 beta (LC3), which, in turn, activates the EGFR/Akt signaling, thus rendering melanoma CSCs resistant to T cell recruitment and cytotoxicity [146]. Sox2, another very well-known marker of melanoma stemness, has also been described as a player in the mechanisms of immune resistance. Specifically, Wu et al. recently demonstrated that Sox2 is able to promote the expression of immunosuppressive genes, such as PD-L1 and IDO1, favoring melanoma CSC-mediated depletion of T cell cytotoxic activity; in line with this observation, epigenetic suppression of Sox2 was observed to synergize with anti-PD-1-based immunotherapy in melanoma mouse models [247]. Finally, T cell inhibition may be directly triggered by tumor cell-secreted cytokines, including IL-4, IL-10 and TGF-β; it has been reported that ABCB5+ melanoma cells express and release in the TME high levels of TGF-β-related factors [84].

3.1.2. Interactions with Innate Immune Cells

Natural Killer (NK) cells, a subpopulation of large granular lymphocytes, are effector lymphocytes of the innate immune system that provide host defense against tumors. They directly interact with tumor cells, as well as CSCs, or indirectly with other cells to regulate tumor growth within the TME [248,249]. Different mechanisms have been observed to be activated in melanoma CSCs to escape from cytotoxicity elicited by NK cells. Melanoma cells undergoing EMT were shown to express high levels of the NK-protective HLA-I protein on their surface and to downregulate the expression of various NK cell-activating receptors [250]. The integrin beta-like protein 1 (ITGBL1) is a factor highly expressed in melanoma CSCs; secreted ITGBL1 protein was found to impair NK cell cytotoxicity [251]. It is well known that the β-adrenergic signaling pathway is endowed with a protumor activity through the β3-adrenoreceptor (β3-AR) and blocking β3-AR activation protects against the development of different types of tumors. Calvani and coworkers reported that the pharmacological inhibition of β3-AR reduces the expression of stemness markers in melanoma cells and this effect is associated with an increased recruitment of NK cells. This evidence supports a key role of this receptor as a stemness marker in melanoma cells being deeply involved in tumor immune escape mechanisms [252]. More recently, Lehmann et al. reported that the immune surveillance exerted by NK cells is bypassed by melanoma cells expressing high levels of the NGFR stemness marker. Specifically, by using in vitro and in vivo experimental models, these authors could observe that NGFR protects tumor cells from NK cell-mediated clearance by downregulating NK cell-activating ligands and lipid reprogramming [253]. In cutaneous melanoma, infiltrated NK cells are functionally impaired. A dysregulation of the activating NKG2D receptor expressed in NK cells and involved in tumor recognition has been highlighted as a factor underlying their defective behavior. This receptor signaling is crucial in the regulation of NK cell cytotoxic activity and cytokine production. It has been demonstrated that very low levels of NKG2D can be detected in NK cells from melanoma tissues enriched with the stem cell marker CD133+ [254].
Immunosuppressive cells, such as tumor-associated neutrophils (TANs), tumor-associated macrophages (TAMs), myeloid-derived suppressor cells (MDSCs), DCs and adoptive immune cells like the regulatory T cells (Tregs), are the cellular components of the TME hindering the protective immunosurveillance of neoplasia, impairing the antitumor immune responses and promoting tumor growth and progression [255].
TANs are a type of white blood cells, representing the most abundant immune cells in the TME. They are involved in the early phases of inflammation and represent the first line of defense of the innate immune response, being endowed with an immunostimulatory effect. However, cancer cells release different types of cytokines, such as TGF-β, IL-6 and IL-8, that promote TAN polarization from an antitumor (N1) to an immunosuppressive and protumor phenotype (N2) [256,257]. Specifically, N1 cells are characterized by slow proliferation and by the release of high levels of tumor necrosis factor-alpha (TNF-α). In contrast, N2 cells are endowed with immunosuppressive and protumor activities favoring the release of NETs (neutrophil extracellular traps, a network of extracellular strings of DNA), ROS (reactive oxygen species), MMP-9, IL-8 and IL-6 [257,258]. Recently, Modestino and colleagues reported that melanoma cells induce chemotaxis as well as the activation and release of NETs from polymorphonuclear neutrophils; moreover, high levels of MMP-9, IL-8, NET, granulocytes and monocyte colony-stimulating factors were reported in patients with advanced melanoma when compared to healthy controls [259]. In line with these data, a high presence of TANs was observed in melanoma patients and found to correlate with a low therapeutic outcome [260]. Interestingly, we recently demonstrated that melanoma CSCs release soluble factors, such as TGF-β, IL-6 and IL-8, able to promote neutrophil recruitment and their switch toward the N2 phenotype, mediated by the activation of the ERK, p38 and STAT3 signaling pathway and the concurrent overexpression of CXCR2 and NF-κB. Moreover, the conditioned medium from melanoma CSCs promoted ROS production, cytokines and MMP-9 secretion and NET release from neutrophils. Notably, we could also observe that CSC-activated neutrophils could endow melanoma cells with peculiar stemness traits (i.e., ABCG2 expression and melanosphere formation) [261]. These results strongly support the existence of a bidirectional interplay between CSCs and neutrophils in melanoma.
Macrophages are cells of the innate immune system that derive from the differentiation of monocytes in tissues. Like neutrophils, two opposite TAM polarization states have been described, named M1 and M2 [262]. The former is characterized by the production of proinflammatory cytokines such as TNF-α, IL-1β, IL-6 and IL-12 and promotes an immune response against the tumor. The latter secrete anti-inflammatory cytokines, such as IL-10, CCL18 and TGF-β, and favors an immunosuppressive environment supporting tumor growth and progression [263,264]. M2 cells were shown to promote melanoma cell proliferation, invasion, metastasis and angiogenesis and to be deeply involved in the development of drug resistance [265,266,267,268,269]. Moreover, in different types of cancers, TAMs have been reported to be recruited and educated toward the M2 phenotype by the CSC subpopulations through the release of chemokines and interleukins; on the other hand, M2 cells establish a TME niche favorable for CSC survival and maintenance of stemness properties [270,271]. These data support a strict bidirectional crosstalk between CSCs and macrophages in the TME. Glucosylceramide synthase (GCS) is an enzyme well known to play an important role in sphingolipid biosynthesis, which is necessary for the maintenance of melanoma CSCs. Ghosh and coworkers demonstrated that tumor hypoxia induces GCS expression in melanoma cells, correlated with a significant increase in the melanoma CSC subpopulation. Mechanistically, hypoxia induces the release of TGF-β from TAMs and Tregs (the specialized subpopulation of T cells that act to suppress immune response), which, in turn, promotes GCS expression in melanoma cells, thus favoring the expansion of CSCs. Moreover, siRNA-based silencing of GCS abrogates the immunosuppressive ability of TAMs and Tregs. Together, these data support the notion that a bidirectional communication exists between melanoma CSCs and tumor-infiltrating immune cells and plays a pivotal role in CSC maintenance and tumorigenicity [272].
MDSCs are a heterogeneous group of myeloid cells derived from hematopoietic cells residing in the bone marrow and characterized by a peculiar immunosuppressive activity [273]. Specifically, they are endowed with the ability to impair both adaptive and innate immune responses in different pathological conditions, including cancer, facilitating immune evasion [274,275,276,277,278,279,280,281]. Moreover, they play a crucial role in promoting EMT and boosting CSC stemness. On the other hand, CSCs attract MDSCs, favoring their infiltration, expansion and immunosuppressive abilities [282]. In breast cancer, MDSCs have been shown to play an important role in escaping the immune response as well as in promoting the tumorigenic and metastatic capacity and the stemness features of cancer cells through the activation of the STAT/Notch signaling pathway [283,284]. MDSCs were also reported to promote immune evasion in melanoma cells by inhibiting T cell activity and macrophage-mediated phagocytosis [227,285,286] and to confer stemness properties to tumor cells by activating the IL-6/STAT3 signaling pathway [287]. Notably, in preclinical studies, Shidal and colleagues demonstrated that CD133+ melanoma CSCs in the B16F10 melanoma murine model are associated with the recruitment of high amounts of immunosuppressive cells, such as MDSCs but also M2 macrophages and Treg cells, during tumor formation. Mechanistically, these authors showed that a low expression of miR-92 in melanoma CSCs is associated with the activation of the integrin α/TGF-β axis to promote intratumoral immunosuppression and enhance tumorigenesis [288].

3.2. Melanoma CSC–Endothelial Cell Interplay

The process of angiogenesis, the generation of new capillary blood vessels from pre-existing ones, is now considered a hallmark of cancers, including melanoma [289,290]. New vessel formation is necessary to provide oxygen and critical nutrients for tumor growth and facilitate metastatic dissemination [291]. Several growth factors signaling pathways are involved in angiogenesis. In particular, the vascular endothelial growth factor (VEGF) family (VEGF-A, VEGFB, VEGF-C, VEGF-D, PlGF) and their receptors, VEGFR-1, VEGFR-2 and VEGFR-3, have been consistently reported to be present in melanoma tissues. Activation of this VEGF/VEGFR axis, through the engagement of different intracellular pathways (i.e., MAPK/ERK, PI3K/ERK, PKC and FAK), facilitates melanoma cell proliferation, migration, survival and permeability [292].
It is now well recognized that melanoma CSCs, particularly those located at the tumor margins, express and release several factors deeply involved in the angiogenesis process in their surrounding microenvironment. By setting up a tri-layered melanoma model composed of CSCs, fibroblasts, mesenchymal stem cells and endothelial cells, Lopez de Andrés and coworkers recently observed that embedded cells show high proliferation accompanied by an early onset of vascularization [293]. ABCG2+ and CD133+ melanoma cells were reported to secrete proangiogenic factors such as VEGF and its receptor VEGFR-2, Tie2, angiopoietin and MMP-2/-9 [82,180]. In melanoma cells expressing high levels of CD271, integrin α5β1, through its downstream ERK pathway, regulates the proangiogenic secretome, promoting vessels formation [294]. Similarly, ALDH1A1 overexpression in melanoma cells was found to correlate with an upregulated secretion of proangiogenic factors promoting angiogenic traits in surrounding endothelial cells through the activation of the intracellular Notch signaling cascade [181]. The NOP14 nucleolar protein is known to be involved in the regulation of melanoma pathogenesis. Li and colleagues reported that NOP14 suppresses the stemness and functions of melanoma CSCs through the inactivation of the Wnt/β-catenin intracellular signaling; specifically, its overexpression significantly impairs proliferation, migration and the proangiogenic potential of these cells [166].
VM, the ability of tumor cells to acquire endothelial cell morphology and function, is a process that was first reported to occur in melanoma [295]. In particular, accumulating evidence indicates that melanoma CSCs, characterized by a high level of plasticity, are able to develop an endothelial-like phenotype for the generation of new tumor-derived vessels, giving rise to an alternative microcirculation that is independent of endothelial cells [295,296,297]. The PI3K/Akt signaling pathway has been shown to be deeply involved in this “angiogenic switch” that allows tumor dissemination and metastasis [211]. Moreover, vascular endothelial cadherin (VE-cadherin), a cell–cell adhesion protein commonly expressed by endothelial cells, has been found to be overexpressed in different types of tumors where it contributes to disease progression/aggressiveness and to the acquisition of the VM phenotype [298]. ABCB5+ melanoma cells were reported to express endothelial/proangiogenic molecules as well as specific VM markers, such as VEGF and its receptors, Tie2 and VE-cadherin [299]. Moreover, CD133+ and ABCG2+ melanoma stem cells were shown to overexpress proangiogenic factors and to be characterized by tube-formation capacity [82,180]. Consistently with these observations, CD271+ melanoma stem cells were found to be endowed with VM through activation of the VEGFR/PKC pathway [209]. Syndecan-1, a cell surface heparan sulfate proteoglycan, has been shown to play an important role in the VM process of melanoma CSCs [300]; moreover, Shih et al. recently demonstrated that the heparan sulfate binding angiopoietin-like 4 (ANGPTL4) protein, highly expressed in melanoma CSCs, is deeply involved in the tube-forming ability of these cells in vitro [301]. These data were further confirmed by in vivo studies demonstrating that CD133+ melanoma cells xenografted into nude mice promote tumor growth associated with a significant angiogenesis process [302]. Interestingly, using a two-dimensional (2D) melanoma–endothelium co-culture model, Hsu and colleagues could demonstrate that endothelial cells, through the activation of the Notch3 signaling, facilitate melanoma CSC plasticity by inducing the expression of stemness markers, and promote their VM capacity [182]. Together, these data support that a strict two-way communication between CSCs and endothelial cells is present in melanoma TME and plays a crucial role in regulating the angiogenic process (Figure 3).

3.3. Melanoma CSC-CAF Interplay

CAFs are an important component of the tumor niche, being deeply involved in the promotion of tumor growth and progression, suppressing antitumor immunity and acquiring resistance to cancer therapies [303,304,305,306]. Specifically, a close interaction has been shown between tumor cells and neighboring CAFs in melanoma. CCN2 is a protein belonging to the cellular communication network (CCN) family; high levels of CCN2 have been observed in normal tissue repair and pathologies, including cancers, while it is not expressed by dermal fibroblasts. In human melanoma biopsies, CCN2 expression has been found to negatively correlate with patient survival. Tsang and coworkers reported that tumor cells recruit dermal fibroblasts, fostering their expression of the stem cell marker Sox2 and activation into a CAF phenotype in a CCN2-dependent manner [307]. Eph receptors are the largest family of RTKs and are different from other RTKs by binding to cell-bound ligands (ephrins), being deeply involved in cell–cell junctions and in the regulation of cell adhesion and survival. In particular, the EphA3 isoform has been shown to be overexpressed in melanoma-associated CAFs, while its downregulation is associated with the inhibition of tumor growth and progression [308]. miR-214 expression has been found to be significantly upregulated in malignant melanomas and to act as a prometastatic miRNA through the modulation of a complex molecular pathway including transcription factors, adhesion molecules and the anti-metastatic miR-148b [309]. Orso and colleagues reported that miR-214 is highly expressed in CAFs located in melanoma tissues. Treatment of cancer cells with conditioned medium or EVs derived from miR-214-enriched CAFs promotes melanoma cell migratory and invasive behavior. Interestingly, melanoma cells are able to induce miR-214 production in stromal CAFs, which is then released via EVs and subsequently uptaken by cancer cells. These data support that a bidirectional interplay exists between melanoma cells and CAFs in the TME and plays an important role in coordinating tumor metastatic dissemination [310].
It is now well accepted that tumor-associated CAFs are also deeply involved in CSC renewal, maintenance of stemness traits and drug resistance [311,312]. By means of in vitro and in vivo studies, Kinugasa et al. demonstrated that CAFs expressing the stemness marker CD44 sustain the stemness features of melanoma CSCs via a direct interaction and promote cancer cell drug resistance [313]. More recently, it has been reported that the intracellular Notch1 signaling pathway in CAFs dictates the plasticity of human metastatic melanoma cells. Specifically, Notch1-overexpressing CAFs were found to induce a sustained suppression of melanoma cell stemness; conversely, Notch1-silenced CAFs significantly increased the stemness properties of melanoma CSCs by upregulating the expression of the stemness markers Sox2, Oct4 and Nanog [314] (Figure 3).
Overall, melanoma CSCs and neighboring CAFs in the tumor stroma display a pathological interplay; however, further studies are needed to elucidate the underlying molecular and cellular mechanisms.

4. Cell-to-Cell Communication between CSCs and Non-CSCs in Melanoma

Tumors are generally characterized by a high degree of heterogeneity and plasticity containing cells with different malignant potentials; accumulating evidence supports that the communication between these cells plays an important role in tumor progression and drug resistance [315,316,317,318]. In different tumors, it has been shown that CSCs can endow bystander cells of the bulk population with self-renewal and stemness properties through the horizontal transfer of specific molecular signals [319,320,321,322]. In melanoma, highly metastatic CSCs release exosomes carrying high levels of specific miRNAs, including miR-1268a and miR-4535. Once internalized by target melanoma cells, CSC-derived exosomes deliver both miR-1268a and miR-4535, which in turn foster the invasive behavior and metastatic colonization capability of these cells through the inhibition of the autophagic pathway [323,324]. In line with these observations, it has been demonstrated that EVs derived from melanoma CSCs package and deliver miR-592 to differentiated melanoma cells. Uptaken miR-592 inhibits the expression of its target gene protein tyrosine phosphatase non-receptor type 7 (PTPN7), leading to the activation of the MAPK/ERK signaling pathway and, ultimately, to the promotion of melanoma cell metastatic ability [325]. Interestingly, long non-coding RNAs (lncRNAs) have also been found to be frequently carried by exosomes mediating the communication between the different cell subpopulations of the tumor mass. Chen and coworkers recently reported that the lncRNA Gm33149 is encapsulated into exosomes originating from melanoma CSCs and is then transferred to parental melanoma cells. Mechanistically, upon being uptaken by non-CSCs, Gm33149 competitively binds to miR-5623-3p, thereby activating the Wnt pathway and fostering their acquisition of a high metastatic behavior [326]. So far, studies addressing the horizontal transfer of stemness traits from CSCs to non-stem cells within the melanoma mass are still missing.
Overall, these findings support that exosomal molecular cargos are deeply implicated in the horizontal crosstalk between the different melanoma cell subtypes, being responsible for the transfer of tumor aggressiveness. These cargos not only represent potential biomarkers of tumor progression, but may also provide valuable support for the development of novel therapeutic strategies leading to an improvement in melanoma patient outcome (Figure 3).

5. Targeting CSCs in Melanoma

Chemoresistance to conventional anticancer treatments and immune evasion properties are widely recognized as distinctive properties of CSCs. Thus, targeting the mechanisms operating in CSCs, in combination with standard treatments aimed at eradicating the bulk tumor cell populations, might open new avenues for cancer treatment strategies [327,328,329]. Notably, therapeutic approaches directed against surface and intratumoral targets, such as markers of drug resistance and intracellular pathways in the context of melanoma CSCs, have been recently explored (Table 1 and Table 2).
CD271 is a key molecular marker of melanoma CSCs, and its targeting might represent an effective therapeutic strategy for melanoma patients. Morita and coworkers developed a humanized anti-CD271 monoclonal antibody (mAb) and found that it exerts a cytotoxic activity against CD271+ cell lines. Moreover, in vivo studies showed that this antibody significantly inhibits tumor growth in human melanoma cell xenograft models by depleting the CD271+ cell subpopulation [101]. In line with these observations, it has been shown that mAb-mediated blockade of CD47, a well-known marker of melanoma metastasis and immune evasion, coupled with a CD271 cytotoxic antibody linked to the toxin saporin (CD271S, anti-CD271 mAb-saporin), significantly impairs tumor metastatic behavior in melanoma patient-derived xenografts [94]. In contrast, Saltari et al. reported that activation of the CD271 intracellular domain by a short β-amyloid-derived peptide (Aβ(25-35)), in combination with chemotherapy or targeted therapy, significantly decreases melanoma metastasis in a zebrafish xenograft model as well as tumor growth in mice [330]. Thus, further studies are needed to confirm the reliability of CD271 as an effective target to overcome melanoma progression and drug resistance. In this context, the relevant roles of CD271 in the central nervous system must be considered; appropriate delivery methods need to be developed to specifically target CD271 expressed in the CSC population.
Therapeutic approaches aimed at targeting the CSC marker CD133 have been developed and explored through in vitro and in vivo studies. Rappa and colleagues demonstrated that monoclonal antibodies directed against two distinct epitopes of this marker exert a significant and dose-dependent antitumor activity in melanoma cells. Moreover, the downregulation of CD133, by means of short hairpin (sh) RNAs, markedly reduces in vitro and in vivo the metastatic ability of these cells [331]. Photochemical internalization (PCI) is a laser-controlled technology utilized to promote endosomal escape and specific intracellular delivery of hydrophilic anticancer drugs. AC133-saporin is an immunotoxin complex consisting of a CD133 mAb linked to saporin (AC133-saporin). Bostad and coworkers took advantage of the PCI technology by employing a photosensitizer that induces endosomal disruption and subsequent endosomal escape of internalized anticancer drugs, to investigate the potential of AC133-saporin for targeting the CD133+ population of melanoma cells. These authors observed that treating melanoma cells with PCI/AC133-saporin significantly reduces tumor growth in vitro and in vivo, suggesting this approach as a novel rational strategy for eradicating CD133+ melanoma CSCs [332].
Therapeutic approaches have also been developed to specifically target the melanoma CSC marker CD20. In a chemotherapy-refractory melanoma patient with metastatic disease, intra-lesional injections of the monoclonal anti-CD20 antibody rituximab, together with dacarbazine treatment, induced regression of treated metastasis. Notably, rituximab treatment decreased the number of CD20+ positive cells in metastatic lesions and no treatment-related toxicity could be observed [333]. Winkler and coworkers reported results from a clinical study carried out in seven metastatic melanoma patients receiving rituximab injections for 1 year. All patients had previously experienced different antitumor approaches. Treatment was found to be well tolerated although two patients experienced infusion reactions (grade 2) during the first infusion, but there were no more serious side effects. Five patients showed stable disease, while two patients had progressive disease. Median progression-free survival was 6.3 months, median overall survival was 14.7 months and one patient was still alive 18 months after the start of treatment. These authors suggested that the beneficial toxicity profile of rituximab supports that its combination with other standard systemic treatments (i.e., PD-1 antibodies) might increase therapeutic efficacy and should be further investigated in clinical studies involving large patient cohorts [334]. In line with this suggestion, it has been demonstrated that targeting CD20+ melanoma cells with rituximab significantly potentiates the BRAF inhibitor (vemurafenib)-mediated cell killing [335]. Recently, different therapeutic strategies involving CD20 antibodies were developed. In this context, immunoliposomes conjugated to CD20 antibodies and loaded with the standard anticancer compound vincristin (VCR-Lip-CD20) were reported to significantly decrease the ability of melanoma cells to form melanospheres in vitro and tumor growth in melanoma xenograft mice [336]. Aptamers are short synthetic single-stranded RNA/DNA oligonucleotides that specifically bind to different cell surface molecular targets such as proteins; they have different advantages over antibodies, being highly specific, non-immunogenic and relatively small in size, thus enabling a better drug penetration in tumor cells [337,338]. It was shown that salinomycin-loaded lipid–polymer nanoparticles bound to anti-CD20 aptamers (CD20-SA-NPs), upon being uptaken into CD20+ melanoma cells, exert an important cytotoxic activity against these cells while impairing their ability to form melanospheres [339]. In line with these data, Chen and colleagues reported that anti-CD20 aptamer-modified exosomes loaded with the standard chemotherapy drug adriamycin (ACEXO) efficiently accumulate in melanoma cells and reduce the number of melanospheres; importantly, these results were also confirmed in preclinical studies performed in melanoma tumor-bearing mice [340].
By means of in vitro and in vivo studies, it has been pointed out that lipid nanoparticles coated with hyaluronic acid (HA), a well-known ligand of CD44, and loaded with the taxane chemotherapy drug paclitaxel (PTX-loaded HA-SLNs) suppress the subpopulation of CD44+ melanoma cells [341]. Similarly, albumin nanoparticles functionalized with HA to specifically target CD44-overexpressing cells and filled with all-trans retinoic acid (HA-eNPs/ATRA) were found to suppress the tumorigenicity as well as the metastatic potential of CD44-enriched melanoma cells [342]. RG7356 is a recombinant anti-CD44 immunoglobulin G1 humanized monoclonal antibody. The efficacy of RG7356 has been investigated by Menke-van der Houven van Oordt and colleagues in a first-in-human phase I trial in patients with locally advanced or metastatic cancers expressing high levels of CD44, including melanoma. Patients unresponsive to previous treatments were intravenously treated with escalating doses of the compound. The treatment was well tolerated, showing only minor adverse effects. However, the clinical efficacy was low, with only 21% of patients experiencing a disease stabilization of a median of 12 weeks. Based on these results, these authors suggested that additional investigations of the efficacy of RG7356, possibly in combination regimens, should be carried out [343].
Specific compounds have also been developed to target drug resistance markers. In particular, monoclonal antibodies directed at ABCB5 were reported to reverse melanoma cell resistance to doxorubicin [84]; however, further investigations are warranted to confirm these data. In our laboratory, we demonstrated that the natural compound δ-tocotrienol decreases the ability of melanoma cells to form melanospheres and promotes the disaggregation of melanospheres, suppressing the expression of the ABCG2 marker [78].
The intracellular stemness marker ALDH is another potential target for novel cancer therapies; different inhibitors of multi-isoform ALDH (specifically ALDH1A1, ALDHA2, ALDH3A1) have been developed and their biological activity has been evaluated in different types of tumor cells, including melanoma cells [132]. Interestingly, it has been shown that ALDH1 can induce the activation of nifuroxazide (a member of the 5-nitrofurans family); of interest, activated nifuroxazide targets ALDH1+ melanoma cells, leading to the suppression of their stemness features [131]. A whole-cell melanoma vaccine genetically modified toward a CSC phenotype characterized by high levels of ALDH1 (AGI-101H) was applied in advanced melanoma patients, with resected and non-resected tumors, and combined with surgery in patients with recurring metastases. AGI-101H treatment was associated with a significant increased patient survival (11–19 years) and found to trigger the generation of cytotoxic CD8+ T cells specific for ALDH1A1 [344]. More recently, Liao and coworkers developed a dual ALDH1A1+ALDH1A3 peptide–dendritic cell vaccine and found that it induces suppression of tumor growth by eliciting a peculiar T cell immunity that specifically targets ALDH+ melanoma cells [133].
Last but not least, different natural and synthetic compounds, such as triphenylmetane gentian violet, lunasin, retinoic acid and chelerythrine chloride, have also been shown to specifically target the melanoma CSC subpopulation by regulating the expression/activity of the embryonic transcription factors Oct4, Sox2 and Nanog [345,346,347,348].
Inhibition of the intracellular signaling pathways involved in the promotion of cancer cell stemness is also considered a potential therapeutic approach for cancer patients.
Several anticancer compounds were found to exert their activity by targeting the Wnt/β-catenin pathway in CSCs and some of them are in clinical trials for the treatment of patients with Wnt-driven tumors [149]. Pimozide is an antipsychotic drug approved by the FDA in 2011. Accumulated evidence supports that this compound is endowed with peculiar cytotoxic properties on cancer cells, including melanoma cells, by targeting the CSC subpopulation [169,349]. Specifically, it has been reported that pimozide inhibits the signal transducer and activator of the transcription (STAT-3 and 5) pathway, which is overactivated in CSCs, thus significantly impairing CSC features. Interestingly, recently accumulated evidence supports that this compound is endowed with peculiar cytotoxic properties on cancer cells, including melanoma cells, by specifically targeting the CSC subpopulation [169]. A novel anti-CSCs drug (35b), derived from the natural chemical scaffold of symplostatin 4 (a natural anti-malaria depsipeptide), was reported to exhibit a significant inhibitory effect on melanoma growth by reducing CSC traits, such as melanosphere formation and ALDH expression, by impairing the Wnt/β-catenin signaling pathway [168]. Moreover, the natural compound morin (3,5,7,20,40-pentahydroxyflavone), a natural polyphenol isolated from members of the Moraceae family, has been reported to inhibit the growth of many types of cancer cells, including melanoma CSCs. Mechanistically, it has been demonstrated that morin significantly inhibits the growth as well as the stemness features (self-renewal and sphere-formation capacity) of melanoma cells overexpressing CD133, by increasing the expression of miR-216a while hampering its downstream target Wnt3 [350].
Targeting the Notch signaling pathway has been proposed as an effective strategy to eradicate the CSC subpopulation in different tumors [351,352]. The γ-secretase inhibitors RO4929097 and honokiol were reported to reduce the proliferation and the ability to acquire stemness features of melanoma cells [353,354]. However, Keyghobadi et al. demonstrated that only short-term inhibition of the γ-secretase/NICD axis, using the γ-secretase inhibitor DAPT (N-[N-(3,5-difluorophenacetyl)-L-alanyl]-Sphenylglycine t-butyl ester), is associated with reduced proliferation and stemness traits in melanoma cells; notably, an opposite effect was found after a long-term treatment of the cells with the Notch pathway inhibitor [355]. Thus, further studies are needed to confirm the role of the Notch signaling pathway as an effective molecular target to suppress the CSC subpopulation and to reverse drug resistance in melanoma.
The potential of targeting the HH/Gli pathway with anticancer agents endowed with a specific activity against CSCs has been recently explored in melanoma [351]. Specifically, Smo inhibitors (vismodegib and sonidegib) have been developed and clinically investigated; however, their use was limited due to the occurrence of resistance and serious side effects and to a low selectivity against CSCs [356,357]. Novel and more potent Smo inhibitors, based on acylguanidine scaffolds, have been recently developed and investigated for their potential anticancer activity. Pietrobono and coworkers demonstrated that, in melanoma cells, these compounds specifically target the Smo protein, thus leading to a decreased expression of Gli1, and significantly impair the self-renewal of the CSC subpopulation [358]. Ornidazole is an antibiotic used for the treatment of many bacterial and parasitic infections and was recently reported to suppress CD133+ melanoma CSCs by inhibiting the HH signaling pathway and hindering tumor growth in mouse models [359]. Together, these studies provide the basis for novel, HH/Gli-targeted therapeutic approaches for the eradication of CSCs in melanoma.
So far, different compounds have been developed and reported to impair drug resistance in cancer cells endowed with stem-like traits, including melanoma CSCs, through the suppression of the PI3K/Akt/mTOR signaling cascade. Some of these inhibitors have been approved by the FDA and are presently being investigated in preclinical studies as well as in clinical trials [360,361,362,363,364]. Recently, it has been demonstrated that evodiamine, a novel alkaloid isolated from the fruit of tetradium, significantly inhibits the growth of vemurafenib-resistant melanoma cells by inhibiting the PI3K/Akt signaling pathway through the suppression of IRS4 (insulin receptor substrate 4) [364]. The anaplastic lymphoma kinase (ALK) transcript has been shown to be overexpressed in melanoma cells and to induce CSC-like properties by promoting sphere formation and upregulating the expression of stem cell markers [365]; TAE684, a pharmacological inhibitor of ALK, in combination with a PI3K/mTOR inhibitor, reverts the stemness properties of these cells [360]. Itraconazole, a commonly used antifungal compound, has been observed to impair the growth and colony-formation ability of melanoma cells by hampering the functions of the PI3K/mTOR, HH and Wnt intracellular signaling pathways [361]. Corrales and colleagues described that the PI3K/Akt pathway is overactivated in melanoma cells made resistant to MAPK/ERK inhibitors (vemurafenib or trametinib) and is associated with increasing cell quiescence and migratory properties; notably, treatment of these cells with the dual PI3K/mTOR inhibitor dactolisib markedly reverts their stem-like properties [207]. Thus, impairing the activity of the PI3K/Akt/mTOR intracellular signaling cascade might represent a promising strategy for advancing CSC-targeted therapies for melanoma treatment.
Table 1. Therapeutic agents targeting melanoma CSC markers.
Table 1. Therapeutic agents targeting melanoma CSC markers.
MarkersAgentsActionsReferences
CD271Anti-CD271 mAbDepletion of CD271+ cells and suppression of tumor growth (in vitro and in vivo
studies)
[101]
CD271S + anti-CD47 mAbInhibition of tumor metastasis in patient-derived xenografts[94]
Aβ(25–35) + chemotherapy or targeted therapyActivation of CD271 intracellular domain; induction of apoptosis and reduced tumor volume and metastasis; drug resistance overcome[330]
CD133Anti-CD133 mAbCytotoxic effects[331]
CD133 shRNAReduced metastatic behavior (in vitro and in vivo studies)[331]
AC133-saporinDecreased cell proliferation and colony-forming ability[332]
CD20RituximabRegression of tumor growth in patients[333]
Rituximab + anti-PD-1 mAbBeneficial toxicity profile[334]
Rituximab + BRAF inhibitorPotentiation of vemurafenib-mediated cell killing[335]
VCR-Lip-CD20Decreased ability of melanosphere formation[336]
CD20-SA-NPsCytotoxic activity and decreased melanosphere formation[339]
ACEXODecreased melanosphere formation and reduced tumor growth in nude mice[340]
CD44PTX-loaded HA-SLNsSuppression of CD44+ cells[341]
HA-eNPs/ATRADecreased tumorigenicity and metastatic potential[342]
RG7356 (anti-CD44 mAb)Good toleration, low clinical efficacy in patients[343]
ABCB5Anti-ABCB5 mAbReversion of drug resistance and increased sensitivity to standard treatments[84]
ABCG2δ-TocotrienolDecreased ability of melanosphere formation, promotion of melanosphere disaggregation[78]
ALDHNifuroxazideSuppression of ALDH+ stemness features[131]
AGI-101HInduction of cytotoxic T cells specific for ALDH1A1[344]
ALDH1A1 + ALDH1A3 dual peptide–dendritic cell vaccineInduction of T cell immunity against ALDH+ cells[133]
Stemness-associated transcription factors (Sox2, Nanog, Oct4)Triphenylmethane gentian violet, lunasin, retinoid acid, chelerythrine chlorideDecreased CSC self-renewal, reduced colony formation and tumor growth in mice, CSC differentiation, inhibition of sphere formation[345,346,347,348]
Table 2. Therapeutic agents targeting melanoma CSC signaling pathways.
Table 2. Therapeutic agents targeting melanoma CSC signaling pathways.
Signaling PathwaysAgentsActionsReferences
Wnt/β-cateninPimozideInhibition of CSC features through the downregulation of the STAT-3 and 5 pathway[169]
35bReduced melanosphere formation and ALDH expression[168]
MorinInhibition of cell proliferation, self-renewal and melanosphere formation[350]
NotchRO4929097, honokiolDecreased melanoma cell proliferation and ability to acquire stemness features[353,354]
Hedgehog/GliAcylguanidine derivativesInhibition of self-renewal properties[358]
OrnidazoleInduced DNA damage in CD133+ melanoma cells and suppression of tumor growth in vivo[359]
PI3K/Akt/mTOREvodiamine Inhibition of the growth of vemurafenib-resistant cells through the suppression of IRS4[364]
TAE684 + PI3K/mTOR inhibitorReversion of stemness properties[360]
ItraconazoleDecreased colony formation ability[361]
DactolisibReversion of stemness properties in targeted therapy-resistant cells[207]

6. Conclusions and Future Directions

Cancer cell heterogeneity and plasticity represent a peculiar feature of most solid tumors, including melanoma. In this context, CSCs are a subpopulation of slowly replicating cells endowed with the capacities of cell renewal, proliferation and differentiation, giving rise to the bulk tumor mass; they are also deeply involved in the processes of tumor progression, drug resistance and relapse. Thus, elucidating their molecular and biological features might pave the way for developing novel therapeutic strategies for melanoma patients. From the molecular point of view, melanoma CSCs are defined by the presence of specific surface markers, drug efflux transporters, intracellular markers, pluripotency-associated transcription factors and activated signaling pathways. During the last few years, significant efforts have been made to dissect the molecular mechanisms underlying the biological functions of melanoma CSCs, particularly their role in the development of drug resistance. Accumulated evidence has pointed out an intricate interplay between CSCs and their neighboring cells in the TME. Specifically, it has been reported that melanoma CSCs possess remarkable immune evasion capacities and the ability to selectively recruit and activate immune cells endowed with both immunosuppressive and protumor properties. Moreover, melanoma CSCs release biofactors that improve neoangiogenesis and can differentiate into endothelial-like cells, thus favoring new vessel formation (VM); on the other hand, endothelial cells were reported to sustain CSC plasticity and promote their VM capacity. Notably, detrimental bidirectional crosstalk has also been detected between melanoma CSCs and CAFs. Finally, CSCs were found to horizontally transfer their stemness and aggressive features to bystander non-CSCs within the melanoma tumor mass. Together, these observations support that a precise eradication of CSCs, in a balanced combination with conventionally used therapies, might represent a new promising strategy for the management of melanoma patients, specifically in the advanced and drug-resistant stage. Different compounds targeting CSC molecular and biological traits have been developed and their efficacy in CSC killing has been reported.
However, some challenges remain to be addressed. Firstly, a thorough clarification of the mechanisms involved in the reciprocal CSC-TME crosstalk is still incomplete. Moreover, most of the results so far reported on the activity of CSC-targeting agents derive from in vitro and preclinical studies. Thus, clinical trials confirming these promising observations are urgently needed.

Author Contributions

Conceptualization, P.L. and L.C.; investigation, P.L., R.C. and L.C.; writing—original draft preparation, P.L.; formal analysis, R.C.; writing—review and editing, P.L., R.C. and L.C.; supervision, P.L. and L.C.; figures creation, L.C. and P.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by MIUR: Department of Excellence (Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”, Università degli Studi di Milano, Milan, Italy), by Linea 2 University of Milan (Department of Health Sciences), by PRIN2022C5RHRT and PRIN P2022RSWWF funded by Next Generation EU and MUR, by Associazione Italiana Ricerca sul Cancro, Investigator Grant to RC (20614). Figures are created with https://BioRender.com (License WZ26XH5VU7).

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Siegel, R.L.; Giaquinto, A.N.; Jemal, A. Cancer statistics, 2024. CA Cancer J. Clin. 2024, 74, 12–49. [Google Scholar] [CrossRef] [PubMed]
  2. Di Raimondo, C.; Lozzi, F.; Di Domenico, P.P.; Campione, E.; Bianchi, L. The Diagnosis and Management of Cutaneous Metastases from Melanoma. Int. J. Mol. Sci. 2023, 24, 14535. [Google Scholar] [CrossRef] [PubMed]
  3. Long, G.V.; Swetter, S.M.; Menzies, A.M.; Gershenwald, J.E.; Scolyer, R.A. Cutaneous melanoma. Lancet 2023, 402, 485–502. [Google Scholar] [CrossRef] [PubMed]
  4. Benacka, R.; Szaboova, D.; Gulasova, Z.; Hertelyova, Z.; Radonak, J. Classic and New Markers in Diagnostics and Classification of Breast Cancer. Cancers 2022, 14, 5444. [Google Scholar] [CrossRef] [PubMed]
  5. Timar, J.; Ladanyi, A. Molecular Pathology of Skin Melanoma: Epidemiology, Differential Diagnostics, Prognosis and Therapy Prediction. Int. J. Mol. Sci. 2022, 23, 5384. [Google Scholar] [CrossRef] [PubMed]
  6. Acuna-Ruiz, A.; Carrasco-Lopez, C.; Santisteban, P. Genomic and epigenomic profile of thyroid cancer. Best Pract. Res. Clin. Endocrinol. Metab. 2023, 37, 101656. [Google Scholar] [CrossRef] [PubMed]
  7. Duffield, A.S.; Mullighan, C.G.; Borowitz, M.J. International Consensus Classification of acute lymphoblastic leukemia/lymphoma. Virchows Arch. 2023, 482, 11–26. [Google Scholar] [CrossRef] [PubMed]
  8. Shain, A.H.; Bastian, B.C. From melanocytes to melanomas. Nat. Rev. Cancer 2016, 16, 345–358. [Google Scholar] [CrossRef] [PubMed]
  9. Garbe, C.; Amaral, T.; Peris, K.; Hauschild, A.; Arenberger, P.; Bastholt, L.; Bataille, V.; Del Marmol, V.; Dreno, B.; Fargnoli, M.C.; et al. European consensus-based interdisciplinary guideline for melanoma. Part 1: Diagnostics—Update 2019. Eur. J. Cancer 2020, 126, 141–158. [Google Scholar] [CrossRef]
  10. Scatena, C.; Murtas, D.; Tomei, S. Cutaneous Melanoma Classification: The Importance of High-Throughput Genomic Technologies. Front. Oncol. 2021, 11, 635488. [Google Scholar] [CrossRef] [PubMed]
  11. van Not, O.J.; Blokx, W.A.M.; van den Eertwegh, A.J.M.; de Meza, M.M.; Haanen, J.B.; Blank, C.U.; Aarts, M.J.B.; van den Berkmortel, F.; de Groot, J.W.B.; Hospers, G.A.P.; et al. BRAF and NRAS Mutation Status and Response to Checkpoint Inhibition in Advanced Melanoma. JCO Precis. Oncol. 2022, 6, e2200018. [Google Scholar] [CrossRef] [PubMed]
  12. Shain, A.H.; Joseph, N.M.; Yu, R.; Benhamida, J.; Liu, S.; Prow, T.; Ruben, B.; North, J.; Pincus, L.; Yeh, I.; et al. Genomic and Transcriptomic Analysis Reveals Incremental Disruption of Key Signaling Pathways during Melanoma Evolution. Cancer Cell 2018, 34, 45–55.e4. [Google Scholar] [CrossRef] [PubMed]
  13. Eroglu, Z.; Ribas, A. Combination therapy with BRAF and MEK inhibitors for melanoma: Latest evidence and place in therapy. Ther. Adv. Med. Oncol. 2016, 8, 48–56. [Google Scholar] [CrossRef] [PubMed]
  14. Simeone, E.; Grimaldi, A.M.; Festino, L.; Vanella, V.; Palla, M.; Ascierto, P.A. Combination Treatment of Patients with BRAF-Mutant Melanoma: A New Standard of Care. BioDrugs 2017, 31, 51–61. [Google Scholar] [CrossRef] [PubMed]
  15. Ascierto, P.A.; Dreno, B.; Larkin, J.; Ribas, A.; Liszkay, G.; Maio, M.; Mandala, 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, 5225–5235. [Google Scholar] [CrossRef] [PubMed]
  16. Ascierto, P.A.; Mandala, M.; Ferrucci, P.F.; Guidoboni, M.; Rutkowski, P.; Ferraresi, V.; Arance, A.; Guida, M.; Maiello, E.; Gogas, H.; et al. Sequencing of Ipilimumab Plus Nivolumab and Encorafenib Plus Binimetinib for Untreated BRAF-Mutated Metastatic Melanoma (SECOMBIT): A Randomized, Three-Arm, Open-Label Phase II Trial. J. Clin. Oncol. 2023, 41, 212–221. [Google Scholar] [CrossRef] [PubMed]
  17. de Oliveira Filho, R.S.; de Oliveira, D.A.; Nisimoto, M.M.; Marti, L.C. A Review of Advanced Cutaneous Melanoma Therapies and Their Mechanisms, from Immunotherapies to Lysine Histone Methyl Transferase Inhibitors. Cancers 2023, 15, 5751. [Google Scholar] [CrossRef] [PubMed]
  18. Boutros, A.; Croce, E.; Ferrari, M.; Gili, R.; Massaro, G.; Marconcini, R.; Arecco, L.; Tanda, E.T.; Spagnolo, F. The treatment of advanced melanoma: Current approaches and new challenges. Crit. Rev. Oncol. Hematol. 2024, 196, 104276. [Google Scholar] [CrossRef] [PubMed]
  19. Zhang, J.; Joshua, A.M.; Li, Y.; O’Meara, C.H.; Morris, M.J.; Khachigian, L.M. Targeted therapy, immunotherapy, and small molecules and peptidomimetics as emerging immunoregulatory agents for melanoma. Cancer Lett. 2024, 586, 216633. [Google Scholar] [CrossRef] [PubMed]
  20. Gide, T.N.; Wilmott, J.S.; Scolyer, R.A.; Long, G.V. Primary and Acquired Resistance to Immune Checkpoint Inhibitors in Metastatic Melanoma. Clin Cancer Res. 2018, 24, 1260–1270. [Google Scholar] [CrossRef] [PubMed]
  21. Fares, C.M.; Van Allen, E.M.; Drake, C.G.; Allison, J.P.; Hu-Lieskovan, S. Mechanisms of Resistance to Immune Checkpoint Blockade: Why Does Checkpoint Inhibitor Immunotherapy Not Work for All Patients? Am. Soc. Clin. Oncol. Educ. Book 2019, 39, 147–164. [Google Scholar] [CrossRef] [PubMed]
  22. Bai, R.; Chen, N.; Li, L.; Du, N.; Bai, L.; Lv, Z.; Tian, H.; Cui, J. Mechanisms of Cancer Resistance to Immunotherapy. Front. Oncol. 2020, 10, 1290. [Google Scholar] [CrossRef] [PubMed]
  23. Shah, V.; Panchal, V.; Shah, A.; Vyas, B.; Agrawal, S.; Bharadwaj, S. Immune checkpoint inhibitors in metastatic melanoma therapy (Review). Med. Int. 2024, 4, 13. [Google Scholar] [CrossRef] [PubMed]
  24. Splendiani, E.; Besharat, Z.M.; Covre, A.; Maio, M.; Di Giacomo, A.M.; Ferretti, E. Immunotherapy in melanoma: Can we predict response to treatment with circulating biomarkers? Pharmacol. Ther. 2024, 256, 108613. [Google Scholar] [CrossRef] [PubMed]
  25. Chappell, W.H.; Steelman, L.S.; Long, J.M.; Kempf, R.C.; Abrams, S.L.; Franklin, R.A.; Basecke, J.; Stivala, F.; Donia, M.; Fagone, P.; et al. Ras/Raf/MEK/ERK and PI3K/PTEN/Akt/mTOR inhibitors: Rationale and importance to inhibiting these pathways in human health. Oncotarget 2011, 2, 135–164. [Google Scholar] [CrossRef] [PubMed]
  26. Yadav, V.; Zhang, X.; Liu, J.; Estrem, S.; Li, S.; Gong, X.Q.; Buchanan, S.; Henry, J.R.; Starling, J.J.; Peng, S.B. Reactivation of mitogen-activated protein kinase (MAPK) pathway by FGF receptor 3 (FGFR3)/Ras mediates resistance to vemurafenib in human B-RAF V600E mutant melanoma. J. Biol. Chem. 2012, 287, 28087–28098. [Google Scholar] [CrossRef] [PubMed]
  27. Caporali, S.; Alvino, E.; Lacal, P.M.; Levati, L.; Giurato, G.; Memoli, D.; Caprini, E.; Antonini Cappellini, G.C.; D’Atri, S. Targeting the PI3K/AKT/mTOR pathway overcomes the stimulating effect of dabrafenib on the invasive behavior of melanoma cells with acquired resistance to the BRAF inhibitor. Int. J. Oncol. 2016, 49, 1164–1174. [Google Scholar] [CrossRef] [PubMed]
  28. Catalanotti, F.; Cheng, D.T.; Shoushtari, A.N.; Johnson, D.B.; Panageas, K.S.; Momtaz, P.; Higham, C.; Won, H.H.; Harding, J.J.; Merghoub, T.; et al. PTEN Loss-of-Function Alterations Are Associated With Intrinsic Resistance to BRAF Inhibitors in Metastatic Melanoma. JCO Precis. Oncol. 2017, 1, PO.16.00054. [Google Scholar] [CrossRef]
  29. Kozar, I.; Margue, C.; Rothengatter, S.; Haan, C.; Kreis, S. Many ways to resistance: How melanoma cells evade targeted therapies. Biochim. Biophys. Acta Rev. Cancer 2019, 1871, 313–322. [Google Scholar] [CrossRef]
  30. Czarnecka, A.M.; Bartnik, E.; Fiedorowicz, M.; Rutkowski, P. Targeted Therapy in Melanoma and Mechanisms of Resistance. Int. J. Mol. Sci. 2020, 21, 4576. [Google Scholar] [CrossRef]
  31. Proietti, I.; Skroza, N.; Bernardini, N.; Tolino, E.; Balduzzi, V.; Marchesiello, A.; Michelini, S.; Volpe, S.; Mambrin, A.; Mangino, G.; et al. Mechanisms of Acquired BRAF Inhibitor Resistance in Melanoma: A Systematic Review. Cancers 2020, 12, 2801. [Google Scholar] [CrossRef] [PubMed]
  32. Savoia, P.; Zavattaro, E.; Cremona, O. Clinical Implications of Acquired BRAF Inhibitors Resistance in Melanoma. Int. J. Mol. Sci. 2020, 21, 9730. [Google Scholar] [CrossRef]
  33. Poulikakos, P.I.; Sullivan, R.J.; Yaeger, R. Molecular Pathways and Mechanisms of BRAF in Cancer Therapy. Clin. Cancer Res. 2022, 28, 4618–4628. [Google Scholar] [CrossRef]
  34. Al Hmada, Y.; Brodell, R.T.; Kharouf, N.; Flanagan, T.W.; Alamodi, A.A.; Hassan, S.Y.; Shalaby, H.; Hassan, S.L.; Haikel, Y.; Megahed, M.; et al. Mechanisms of Melanoma Progression and Treatment Resistance: Role of Cancer Stem-like Cells. Cancers 2024, 16, 470. [Google Scholar] [CrossRef] [PubMed]
  35. Montagut, C.; Sharma, S.V.; Shioda, T.; McDermott, U.; Ulman, M.; Ulkus, L.E.; Dias-Santagata, D.; Stubbs, H.; Lee, D.Y.; Singh, A.; et al. Elevated CRAF as a potential mechanism of acquired resistance to BRAF inhibition in melanoma. Cancer Res. 2008, 68, 4853–4861. [Google Scholar] [CrossRef] [PubMed]
  36. Zhang, F.; Tang, X.; Fan, S.; Liu, X.; Sun, J.; Ju, C.; Liang, Y.; Liu, R.; Zhou, R.; Yu, B.; et al. Targeting the p300/NONO axis sensitizes melanoma cells to BRAF inhibitors. Oncogene 2021, 40, 4137–4150. [Google Scholar] [CrossRef]
  37. Pastwinska, J.; Karas, K.; Karwaciak, I.; Ratajewski, M. Targeting EGFR in melanoma—The sea of possibilities to overcome drug resistance. Biochim. Biophys. Acta Rev. Cancer 2022, 1877, 188754. [Google Scholar] [CrossRef]
  38. Sharma, V.; Young, L.; Cavadas, M.; Owen, K. Reproducibility Project: Cancer Biology. Registered Report: COT drives resistance to RAF inhibition through MAP kinase pathway reactivation. eLife 2016, 5, e11414. [Google Scholar] [CrossRef]
  39. Thompson, B.J. YAP/TAZ: Drivers of Tumor Growth, Metastasis, and Resistance to Therapy. Bioessays 2020, 42, e1900162. [Google Scholar] [CrossRef]
  40. Wessely, A.; Steeb, T.; Berking, C.; Heppt, M.V. How Neural Crest Transcription Factors Contribute to Melanoma Heterogeneity, Cellular Plasticity, and Treatment Resistance. Int. J. Mol. Sci. 2021, 22, 5761. [Google Scholar] [CrossRef]
  41. Bokharaie, H.; Kolch, W.; Krstic, A. Analysis of Alternative mRNA Splicing in Vemurafenib-Resistant Melanoma Cells. Biomolecules 2022, 12, 993. [Google Scholar] [CrossRef] [PubMed]
  42. Karami Fath, M.; Azargoonjahromi, A.; Soofi, A.; Almasi, F.; Hosseinzadeh, S.; Khalili, S.; Sheikhi, K.; Ferdousmakan, S.; Owrangi, S.; Fahimi, M.; et al. Current understanding of epigenetics role in melanoma treatment and resistance. Cancer Cell Int. 2022, 22, 313. [Google Scholar] [CrossRef] [PubMed]
  43. Rubanov, A.; Berico, P.; Hernando, E. Epigenetic Mechanisms Underlying Melanoma Resistance to Immune and Targeted Therapies. Cancers 2022, 14, 5858. [Google Scholar] [CrossRef] [PubMed]
  44. Shaw, P.; Raymond, G.; Tzou, K.S.; Baxi, S.; Mani, R.R.; Kumar Govind, S.; Chandramoorthy, H.C.; Sivanandy, P.; Rajagopal, M.; Samiappan, S.; et al. Molecular Investigation of miRNA Biomarkers as Chemoresistance Regulators in Melanoma: A Protocol for Systematic Review and Meta-Analysis. Genes 2022, 13, 115. [Google Scholar] [CrossRef] [PubMed]
  45. Song, K.; Minami, J.K.; Huang, A.; Dehkordi, S.R.; Lomeli, S.H.; Luebeck, J.; Goodman, M.H.; Moriceau, G.; Krijgsman, O.; Dharanipragada, P.; et al. Plasticity of Extrachromosomal and Intrachromosomal BRAF Amplifications in Overcoming Targeted Therapy Dosage Challenges. Cancer Discov. 2022, 12, 1046–1069. [Google Scholar] [CrossRef] [PubMed]
  46. 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, 317. [Google Scholar] [CrossRef] [PubMed]
  47. Hossain, S.M.; Eccles, M.R. Phenotype Switching and the Melanoma Microenvironment; Impact on Immunotherapy and Drug Resistance. Int. J. Mol. Sci. 2023, 24, 1601. [Google Scholar] [CrossRef] [PubMed]
  48. Xie, Q.; Zhang, R.; Liu, D.; Yang, J.; Hu, Q.; Shan, C.; Li, X. Neuropilin-2 Inhibits Drug Resistance and Progression of Melanoma Involving the MiR-331-3p Regulated Cascade. Curr. Mol. Pharmacol. 2023, 16, 787–799. [Google Scholar] [CrossRef] [PubMed]
  49. Marzagalli, M.; Ebelt, N.D.; Manuel, E.R. Unraveling the crosstalk between melanoma and immune cells in the tumor microenvironment. Semin. Cancer Biol. 2019, 59, 236–250. [Google Scholar] [CrossRef] [PubMed]
  50. Ziogas, D.C.; Theocharopoulos, C.; Koutouratsas, T.; Haanen, J.; Gogas, H. Mechanisms of resistance to immune checkpoint inhibitors in melanoma: What we have to overcome? Cancer Treat. Rev. 2023, 113, 102499. [Google Scholar] [CrossRef] [PubMed]
  51. Sun, Q.; Hong, Z.; Zhang, C.; Wang, L.; Han, Z.; Ma, D. Immune checkpoint therapy for solid tumours: Clinical dilemmas and future trends. Signal Transduct. Target. Ther. 2023, 8, 320. [Google Scholar] [CrossRef]
  52. Karras, P.; Bordeu, I.; Pozniak, J.; Nowosad, A.; Pazzi, C.; Van Raemdonck, N.; Landeloos, E.; Van Herck, Y.; Pedri, D.; Bervoets, G.; et al. A cellular hierarchy in melanoma uncouples growth and metastasis. Nature 2022, 610, 190–198. [Google Scholar] [CrossRef]
  53. Pagliuca, C.; Di Leo, L.; De Zio, D. New Insights into the Phenotype Switching of Melanoma. Cancers 2022, 14, 6118. [Google Scholar] [CrossRef]
  54. Centeno, P.P.; Pavet, V.; Marais, R. The journey from melanocytes to melanoma. Nat. Rev. Cancer 2023, 23, 372–390. [Google Scholar] [CrossRef]
  55. Magee, J.A.; Piskounova, E.; Morrison, S.J. Cancer stem cells: Impact, heterogeneity, and uncertainty. Cancer Cell 2012, 21, 283–296. [Google Scholar] [CrossRef]
  56. Cabrera, M.C.; Hollingsworth, R.E.; Hurt, E.M. Cancer stem cell plasticity and tumor hierarchy. World J. Stem Cells 2015, 7, 27–36. [Google Scholar] [CrossRef]
  57. Melzer, C.; von der Ohe, J.; Lehnert, H.; Ungefroren, H.; Hass, R. Cancer stem cell niche models and contribution by mesenchymal stroma/stem cells. Mol. Cancer 2017, 16, 28. [Google Scholar] [CrossRef]
  58. La Porta, C.A.M.; Zapperi, S. Complexity in cancer stem cells and tumor evolution: Toward precision medicine. Semin. Cancer Biol. 2017, 44, 3–9. [Google Scholar] [CrossRef]
  59. Quintana, E.; Shackleton, M.; Foster, H.R.; Fullen, D.R.; Sabel, M.S.; Johnson, T.M.; Morrison, S.J. Phenotypic heterogeneity among tumorigenic melanoma cells from patients that is reversible and not hierarchically organized. Cancer Cell 2010, 18, 510–523. [Google Scholar] [CrossRef]
  60. Friedmann-Morvinski, D.; Verma, I.M. Dedifferentiation and reprogramming: Origins of cancer stem cells. EMBO Rep. 2014, 15, 244–253. [Google Scholar] [CrossRef]
  61. Quintana, E.; Shackleton, M.; Sabel, M.S.; Fullen, D.R.; Johnson, T.M.; Morrison, S.J. Efficient tumour formation by single human melanoma cells. Nature 2008, 456, 593–598. [Google Scholar] [CrossRef] [PubMed]
  62. El-Khattouti, A.; Sheehan, N.T.; Monico, J.; Drummond, H.A.; Haikel, Y.; Brodell, R.T.; Megahed, M.; Hassan, M. CD133(+) melanoma subpopulation acquired resistance to caffeic acid phenethyl ester-induced apoptosis is attributed to the elevated expression of ABCB5: Significance for melanoma treatment. Cancer Lett. 2015, 357, 83–104. [Google Scholar] [CrossRef] [PubMed]
  63. Marzagalli, M.; Raimondi, M.; Fontana, F.; Montagnani Marelli, M.; Moretti, R.M.; Limonta, P. Cellular and molecular biology of cancer stem cells in melanoma: Possible therapeutic implications. Semin. Cancer Biol. 2019, 59, 221–235. [Google Scholar] [CrossRef]
  64. Jamal, S.M.E.; Alamodi, A.; Wahl, R.U.; Grada, Z.; Shareef, M.A.; Hassan, S.Y.; Murad, F.; Hassan, S.L.; Santourlidis, S.; Gomez, C.R.; et al. Melanoma stem cell maintenance and chemo-resistance are mediated by CD133 signal to PI3K-dependent pathways. Oncogene 2020, 39, 5468–5478. [Google Scholar] [CrossRef] [PubMed]
  65. Marzagalli, M.; Fontana, F.; Raimondi, M.; Limonta, P. Cancer Stem Cells-Key Players in Tumor Relapse. Cancers 2021, 13, 376. [Google Scholar] [CrossRef] [PubMed]
  66. Dhanyamraju, P.K.; Schell, T.D.; Amin, S.; Robertson, G.P. Drug-Tolerant Persister Cells in Cancer Therapy Resistance. Cancer Res. 2022, 82, 2503–2514. [Google Scholar] [CrossRef] [PubMed]
  67. Andreucci, E.; Peppicelli, S.; Ruzzolini, J.; Bianchini, F.; Biagioni, A.; Papucci, L.; Magnelli, L.; Mazzanti, B.; Stecca, B.; Calorini, L. The acidic tumor microenvironment drives a stem-like phenotype in melanoma cells. J. Mol. Med. 2020, 98, 1431–1446. [Google Scholar] [CrossRef] [PubMed]
  68. Kyriakou, G.; Melachrinou, M. Cancer stem cells, epigenetics, tumor microenvironment and future therapeutics in cutaneous malignant melanoma: A review. Future Oncol. 2020, 16, 1549–1567. [Google Scholar] [CrossRef] [PubMed]
  69. Li, Y.R.; Fang, Y.; Lyu, Z.; Zhu, Y.; Yang, L. Exploring the dynamic interplay between cancer stem cells and the tumor microenvironment: Implications for novel therapeutic strategies. J. Transl. Med. 2023, 21, 686. [Google Scholar] [CrossRef] [PubMed]
  70. Borlongan, M.C.; Saha, D.; Wang, H. Tumor Microenvironment: A Niche for Cancer Stem Cell Immunotherapy. Stem Cell Rev. Rep. 2024, 20, 3–24. [Google Scholar] [CrossRef] [PubMed]
  71. Guo, Q.; Zhou, Y.; Xie, T.; Yuan, Y.; Li, H.; Shi, W.; Zheng, L.; Li, X.; Zhang, W. Tumor microenvironment of cancer stem cells: Perspectives on cancer stem cell targeting. Genes Dis. 2024, 11, 101043. [Google Scholar] [CrossRef] [PubMed]
  72. Stecca, B.; Santini, R.; Pandolfi, S.; Penachioni, J.Y. Culture and isolation of melanoma-initiating cells. Curr. Protoc. Stem Cell Biol. 2013, 3, 3–6. [Google Scholar] [CrossRef] [PubMed]
  73. Kozovska, Z.; Gabrisova, V.; Kucerova, L. Malignant melanoma: Diagnosis, treatment and cancer stem cells. Neoplasma 2016, 63, 510–517. [Google Scholar] [CrossRef] [PubMed]
  74. Sette, G.; Fecchi, K.; Salvati, V.; Lotti, F.; Pilozzi, E.; Duranti, E.; Biffoni, M.; Pagliuca, A.; Martinetti, D.; Memeo, L.; et al. Mek inhibition results in marked antitumor activity against metastatic melanoma patient-derived melanospheres and in melanosphere-generated xenografts. J. Exp. Clin. Cancer Res. 2013, 32, 91. [Google Scholar] [CrossRef] [PubMed]
  75. Parmiani, G. Melanoma Cancer Stem Cells: Markers and Functions. Cancers 2016, 8, 34. [Google Scholar] [CrossRef] [PubMed]
  76. Abbaszadegan, M.R.; Bagheri, V.; Razavi, M.S.; Momtazi, A.A.; Sahebkar, A.; Gholamin, M. Isolation, identification, and characterization of cancer stem cells: A review. J. Cell. Physiol. 2017, 232, 2008–2018. [Google Scholar] [CrossRef] [PubMed]
  77. Brinckerhoff, C.E. Cancer Stem Cells (CSCs) in melanoma: There’s smoke, but is there fire? J. Cell. Physiol. 2017, 232, 2674–2678. [Google Scholar] [CrossRef] [PubMed]
  78. Marzagalli, M.; Moretti, R.M.; Messi, E.; Marelli, M.M.; Fontana, F.; Anastasia, A.; Bani, M.R.; Beretta, G.; Limonta, P. Targeting melanoma stem cells with the Vitamin E derivative delta-tocotrienol. Sci. Rep. 2018, 8, 587. [Google Scholar] [CrossRef] [PubMed]
  79. Kusienicka, A.; Bukowska-Strakova, K.; Ciesla, M.; Nowak, W.N.; Bronisz-Budzynska, I.; Seretny, A.; Zukowska, M.; Jez, M.; Krutyholowa, R.; Taha, H.; et al. Heme Oxygenase-1 Has a Greater Effect on Melanoma Stem Cell Properties Than the Expression of Melanoma-Initiating Cell Markers. Int. J. Mol. Sci. 2022, 23, 3596. [Google Scholar] [CrossRef] [PubMed]
  80. Speigl, L.; Janssen, N.; Weide, B.; Sinnberg, T.; Pawelec, G.; Shipp, C. Putative Cancer Stem Cell Markers are Frequently Expressed by Melanoma Cells in Vitro and in Situ but are also Present in Benign Differentiated Cells. Front. Biosci. (Landmark Ed.) 2023, 28, 193. [Google Scholar] [CrossRef] [PubMed]
  81. Klein, W.M.; Wu, B.P.; Zhao, S.; Wu, H.; Klein-Szanto, A.J.; Tahan, S.R. Increased expression of stem cell markers in malignant melanoma. Mod. Pathol. 2007, 20, 102–107. [Google Scholar] [CrossRef] [PubMed]
  82. Monzani, E.; Facchetti, F.; Galmozzi, E.; Corsini, E.; Benetti, A.; Cavazzin, C.; Gritti, A.; Piccinini, A.; Porro, D.; Santinami, M.; et al. Melanoma contains CD133 and ABCG2 positive cells with enhanced tumourigenic potential. Eur. J. Cancer 2007, 43, 935–946. [Google Scholar] [CrossRef] [PubMed]
  83. Fang, D.; Nguyen, T.K.; Leishear, K.; Finko, R.; Kulp, A.N.; Hotz, S.; Van Belle, P.A.; Xu, X.; Elder, D.E.; Herlyn, M. A tumorigenic subpopulation with stem cell properties in melanomas. Cancer Res. 2005, 65, 9328–9337. [Google Scholar] [CrossRef] [PubMed]
  84. Schatton, T.; Murphy, G.F.; Frank, N.Y.; Yamaura, K.; Waaga-Gasser, A.M.; Gasser, M.; Zhan, Q.; Jordan, S.; Duncan, L.M.; Weishaupt, C.; et al. Identification of cells initiating human melanomas. Nature 2008, 451, 345–349. [Google Scholar] [CrossRef] [PubMed]
  85. Gonzalez-Herrero, I.; Romero-Camarero, I.; Canueto, J.; Cardenoso-Alvarez, E.; Fernandez-Lopez, E.; Perez-Losada, J.; Sanchez-Garcia, I.; Roman-Curto, C. CD133+ cell content correlates with tumour growth in melanomas from skin with chronic sun-induced damage. Br. J. Dermatol. 2013, 169, 830–837. [Google Scholar] [CrossRef] [PubMed]
  86. El-Khattouti, A.; Selimovic, D.; Haikel, Y.; Megahed, M.; Gomez, C.R.; Hassan, M. Identification and analysis of CD133(+) melanoma stem-like cells conferring resistance to taxol: An insight into the mechanisms of their resistance and response. Cancer Lett. 2014, 343, 123–133. [Google Scholar] [CrossRef] [PubMed]
  87. Kharouf, N.; Flanagan, T.W.; Alamodi, A.A.; Al Hmada, Y.; Hassan, S.Y.; Shalaby, H.; Santourlidis, S.; Hassan, S.L.; Haikel, Y.; Megahed, M.; et al. CD133-Dependent Activation of Phosphoinositide 3-Kinase/AKT/Mammalian Target of Rapamycin Signaling in Melanoma Progression and Drug Resistance. Cells 2024, 13, 240. [Google Scholar] [CrossRef]
  88. Madjd, Z.; Erfani, E.; Gheytanchi, E.; Moradi-Lakeh, M.; Shariftabrizi, A.; Asadi-Lari, M. Expression of CD133 cancer stem cell marker in melanoma: A systematic review and meta-analysis. Int. J. Biol. Markers 2016, 31, e118–e125. [Google Scholar] [CrossRef] [PubMed]
  89. Kloskowski, T.; Jarzabkowska, J.; Jundzill, A.; Balcerczyk, D.; Buhl, M.; Szeliski, K.; Bodnar, M.; Marszalek, A.; Drewa, G.; Drewa, T.; et al. CD133 Antigen as a Potential Marker of Melanoma Stem Cells: In Vitro and In Vivo Studies. Stem Cells Int. 2020, 2020, 8810476. [Google Scholar] [CrossRef] [PubMed]
  90. Korn, P.; Kampmann, A.; Spalthoff, S.; Jehn, P.; Tavassol, F.; Lentge, F.; Gellrich, N.C.; Zimmerer, R. Suitability of CD133 as a Marker for Cancer Stem Cells in Melanoma. Asian Pac. J. Cancer Prev. 2021, 22, 1591–1597. [Google Scholar] [CrossRef] [PubMed]
  91. Moreno-Londono, A.P.; Robles-Flores, M. Functional Roles of CD133: More than Stemness Associated Factor Regulated by the Microenvironment. Stem Cell Rev. Rep. 2024, 20, 25–51. [Google Scholar] [CrossRef] [PubMed]
  92. Boiko, A.D.; Razorenova, O.V.; van de Rijn, M.; Swetter, S.M.; Johnson, D.L.; Ly, D.P.; Butler, P.D.; Yang, G.P.; Joshua, B.; Kaplan, M.J.; et al. Human melanoma-initiating cells express neural crest nerve growth factor receptor CD271. Nature 2010, 466, 133–137. [Google Scholar] [CrossRef] [PubMed]
  93. Civenni, G.; Walter, A.; Kobert, N.; Mihic-Probst, D.; Zipser, M.; Belloni, B.; Seifert, B.; Moch, H.; Dummer, R.; van den Broek, M.; et al. Human CD271-positive melanoma stem cells associated with metastasis establish tumor heterogeneity and long-term growth. Cancer Res. 2011, 71, 3098–3109. [Google Scholar] [CrossRef] [PubMed]
  94. Ngo, M.; Han, A.; Lakatos, A.; Sahoo, D.; Hachey, S.J.; Weiskopf, K.; Beck, A.H.; Weissman, I.L.; Boiko, A.D. Antibody Therapy Targeting CD47 and CD271 Effectively Suppresses Melanoma Metastasis in Patient-Derived Xenografts. Cell Rep. 2016, 16, 1701–1716. [Google Scholar] [CrossRef] [PubMed]
  95. Radke, J.; Rossner, F.; Redmer, T. CD271 determines migratory properties of melanoma cells. Sci. Rep. 2017, 7, 9834. [Google Scholar] [CrossRef] [PubMed]
  96. Redmer, T.; Walz, I.; Klinger, B.; Khouja, S.; Welte, Y.; Schafer, R.; Regenbrecht, C. The role of the cancer stem cell marker CD271 in DNA damage response and drug resistance of melanoma cells. Oncogenesis 2017, 6, e291. [Google Scholar] [CrossRef] [PubMed]
  97. Nielsen, P.S.; Riber-Hansen, R.; Steiniche, T. Immunohistochemical CD271 expression correlates with melanoma progress in a case-control study. Pathology 2018, 50, 402–410. [Google Scholar] [CrossRef] [PubMed]
  98. Beretti, F.; Gatti, M.; Zavatti, M.; Bassoli, S.; Pellacani, G.; Maraldi, T. Reactive Oxygen Species Regulation of Chemoresistance and Metastatic Capacity of Melanoma: Role of the Cancer Stem Cell Marker CD271. Biomedicines 2023, 11, 1229. [Google Scholar] [CrossRef] [PubMed]
  99. Redmer, T.; Welte, Y.; Behrens, D.; Fichtner, I.; Przybilla, D.; Wruck, W.; Yaspo, M.L.; Lehrach, H.; Schafer, R.; Regenbrecht, C.R. The nerve growth factor receptor CD271 is crucial to maintain tumorigenicity and stem-like properties of melanoma cells. PLoS ONE 2014, 9, e92596. [Google Scholar] [CrossRef] [PubMed]
  100. Restivo, G.; Diener, J.; Cheng, P.F.; Kiowski, G.; Bonalli, M.; Biedermann, T.; Reichmann, E.; Levesque, M.P.; Dummer, R.; Sommer, L. low neurotrophin receptor CD271 regulates phenotype switching in melanoma. Nat. Commun. 2017, 8, 1988. [Google Scholar] [CrossRef] [PubMed]
  101. Morita, S.; Mochizuki, M.; Wada, K.; Shibuya, R.; Nakamura, M.; Yamaguchi, K.; Yamazaki, T.; Imai, T.; Asada, Y.; Matsuura, K.; et al. Humanized anti-CD271 monoclonal antibody exerts an anti-tumor effect by depleting cancer stem cells. Cancer Lett. 2019, 461, 144–152. [Google Scholar] [CrossRef] [PubMed]
  102. Vidal, A.; Redmer, T. Decoding the Role of CD271 in Melanoma. Cancers 2020, 12, 2460. [Google Scholar] [CrossRef] [PubMed]
  103. Cheli, Y.; Bonnazi, V.F.; Jacquel, A.; Allegra, M.; De Donatis, G.M.; Bahadoran, P.; Bertolotto, C.; Ballotti, R. CD271 is an imperfect marker for melanoma initiating cells. Oncotarget 2014, 5, 5272–5283. [Google Scholar] [CrossRef] [PubMed]
  104. Boyle, S.E.; Fedele, C.G.; Corbin, V.; Wybacz, E.; Szeto, P.; Lewin, J.; Young, R.J.; Wong, A.; Fuller, R.; Spillane, J.; et al. CD271 Expression on Patient Melanoma Cells Is Unstable and Unlinked to Tumorigenicity. Cancer Res. 2016, 76, 3965–3977. [Google Scholar] [CrossRef] [PubMed]
  105. Caudron, A.; Battistella, M.; Feugeas, J.P.; Pages, C.; Basset-Seguin, N.; Mazouz Dorval, S.; Funck Brentano, E.; Sadoux, A.; Podgorniak, M.P.; Menashi, S.; et al. EMMPRIN/CD147 is an independent prognostic biomarker in cutaneous melanoma. Exp. Dermatol. 2016, 25, 618–622. [Google Scholar] [CrossRef] [PubMed]
  106. Hu, X.; Su, J.; Zhou, Y.; Xie, X.; Peng, C.; Yuan, Z.; Chen, X. Repressing CD147 is a novel therapeutic strategy for malignant melanoma. Oncotarget 2017, 8, 25806–25813. [Google Scholar] [CrossRef] [PubMed]
  107. Lu, L.; Zhang, J.; Gan, P.; Wu, L.; Zhang, X.; Peng, C.; Zhou, J.; Chen, X.; Su, J. Novel Functions of CD147 in the Mitochondria Exacerbates Melanoma Metastasis. Int. J. Biol. Sci. 2021, 17, 285–297. [Google Scholar] [CrossRef] [PubMed]
  108. Jiang, Y.; Liang, R.; Li, L.; Guan, J. Studies on the effect and mechanism of CD147 on melanoma stem cells. Allergol. Immunopathol. 2024, 52, 71–78. [Google Scholar] [CrossRef] [PubMed]
  109. Schmidt, P.; Kopecky, C.; Hombach, A.; Zigrino, P.; Mauch, C.; Abken, H. Eradication of melanomas by targeted elimination of a minor subset of tumor cells. Proc. Natl. Acad. Sci. USA 2011, 108, 2474–2479. [Google Scholar] [CrossRef] [PubMed]
  110. Dean, M.; Annilo, T. Evolution of the ATP-binding cassette (ABC) transporter superfamily in vertebrates. Annu. Rev. Genom. Hum. Genet. 2005, 6, 123–142. [Google Scholar] [CrossRef] [PubMed]
  111. Locher, K.P. Mechanistic diversity in ATP-binding cassette (ABC) transporters. Nat. Struct. Mol. Biol. 2016, 23, 487–493. [Google Scholar] [CrossRef] [PubMed]
  112. Wu, C.P.; Ambudkar, S.V. The pharmacological impact of ATP-binding cassette drug transporters on vemurafenib-based therapy. Acta Pharm. Sin. B 2014, 4, 105–111. [Google Scholar] [CrossRef] [PubMed]
  113. Cho, Y.; Kim, Y.K. Cancer Stem Cells as a Potential Target to Overcome Multidrug Resistance. Front. Oncol. 2020, 10, 764. [Google Scholar] [CrossRef]
  114. Muriithi, W.; Macharia, L.W.; Heming, C.P.; Echevarria, J.L.; Nyachieo, A.; Filho, P.N.; Neto, V.M. ABC transporters and the hallmarks of cancer: Roles in cancer aggressiveness beyond multidrug resistance. Cancer Biol. Med. 2020, 17, 253–269. [Google Scholar] [CrossRef]
  115. Nobili, S.; Lapucci, A.; Landini, I.; Coronnello, M.; Roviello, G.; Mini, E. Role of ATP-binding cassette transporters in cancer initiation and progression. Semin. Cancer Biol. 2020, 60, 72–95. [Google Scholar] [CrossRef]
  116. Sargazi, Z.; Yazdani, Y.; Tahavvori, A.; Youshanlouei, H.R.; Alivirdiloo, V.; Beilankouhi, E.A.V.; Valilo, M. NFR2/ABC transporter axis in drug resistance of breast cancer cells. Mol. Biol. Rep. 2023, 50, 5407–5414. [Google Scholar] [CrossRef]
  117. Wilson, B.J.; Saab, K.R.; Ma, J.; Schatton, T.; Putz, P.; Zhan, Q.; Murphy, G.F.; Gasser, M.; Waaga-Gasser, A.M.; Frank, N.Y.; et al. ABCB5 maintains melanoma-initiating cells through a proinflammatory cytokine signaling circuit. Cancer Res. 2014, 74, 4196–4207. [Google Scholar] [CrossRef] [PubMed]
  118. Wang, S.; Tang, L.; Lin, J.; Shen, Z.; Yao, Y.; Wang, W.; Tao, S.; Gu, C.; Ma, J.; Xie, Y.; et al. ABCB5 promotes melanoma metastasis through enhancing NF-kappaB p65 protein stability. Biochem. Biophys. Res. Commun. 2017, 492, 18–26. [Google Scholar] [CrossRef]
  119. Xiao, J.; Egger, M.E.; McMasters, K.M.; Hao, H. Differential expression of ABCB5 in BRAF inhibitor-resistant melanoma cell lines. BMC Cancer 2018, 18, 675. [Google Scholar] [CrossRef]
  120. Lutz, N.W.; Banerjee, P.; Wilson, B.J.; Ma, J.; Cozzone, P.J.; Frank, M.H. Expression of Cell-Surface Marker ABCB5 Causes Characteristic Modifications of Glucose, Amino Acid and Phospholipid Metabolism in the G3361 Melanoma-Initiating Cell Line. PLoS ONE 2016, 11, e0161803. [Google Scholar] [CrossRef]
  121. Gerard, L.; Duvivier, L.; Fourrez, M.; Salazar, P.; Sprimont, L.; Xia, D.; Ambudkar, S.V.; Gottesman, M.M.; Gillet, J.P. Identification of two novel heterodimeric ABC transporters in melanoma: ABCB5beta/B6 and ABCB5beta/B9. J. Biol. Chem. 2024, 300, 105594. [Google Scholar] [CrossRef] [PubMed]
  122. Louphrasitthiphol, P.; Chauhan, J.; Goding, C.R. ABCB5 is activated by MITF and beta-catenin and is associated with melanoma differentiation. Pigment Cell Melanoma Res. 2020, 33, 112–118. [Google Scholar] [CrossRef]
  123. Muller, J.; Krijgsman, O.; Tsoi, J.; Robert, L.; Hugo, W.; Song, C.; Kong, X.; Possik, P.A.; Cornelissen-Steijger, P.D.; Geukes Foppen, M.H.; et al. Low MITF/AXL ratio predicts early resistance to multiple targeted drugs in melanoma. Nat. Commun. 2014, 5, 5712. [Google Scholar] [CrossRef]
  124. Stacy, A.E.; Jansson, P.J.; Richardson, D.R. Molecular pharmacology of ABCG2 and its role in chemoresistance. Mol. Pharmacol. 2013, 84, 655–669. [Google Scholar] [CrossRef]
  125. Sarkadi, B.; Homolya, L.; Hegedus, T. The ABCG2/BCRP transporter and its variants—From structure to pathology. FEBS Lett. 2020, 594, 4012–4034. [Google Scholar] [CrossRef]
  126. Speigl, L.; Janssen, N.; Weide, B.; Pawelec, G.; Shipp, C. Prognostic impact of the putative cancer stem cell markers ABCG2, CD133, ALDH1A1 and CD44V7/8 in metastatic melanoma. Br. J. Dermatol. 2017, 177, 1447–1449. [Google Scholar] [CrossRef]
  127. Zubrilov, I.; Sagi-Assif, O.; Izraely, S.; Meshel, T.; Ben-Menahem, S.; Ginat, R.; Pasmanik-Chor, M.; Nahmias, C.; Couraud, P.O.; Hoon, D.S.; et al. Vemurafenib resistance selects for highly malignant brain and lung-metastasizing melanoma cells. Cancer Lett. 2015, 361, 86–96. [Google Scholar] [CrossRef] [PubMed]
  128. Miranda-Lorenzo, I.; Dorado, J.; Lonardo, E.; Alcala, S.; Serrano, A.G.; Clausell-Tormos, J.; Cioffi, M.; Megias, D.; Zagorac, S.; Balic, A.; et al. Intracellular autofluorescence: A biomarker for epithelial cancer stem cells. Nat. Methods 2014, 11, 1161–1169. [Google Scholar] [CrossRef]
  129. Luo, Y.; Nguyen, N.; Fujita, M. Isolation of human melanoma stem cells using ALDH as a marker. Curr. Protoc. Stem Cell Biol. 2013, 26, 3–8. [Google Scholar] [CrossRef]
  130. Zhang, S.; Yang, Z.; Qi, F. Aldehyde dehydrogenase-positive melanoma stem cells in tumorigenesis, drug resistance and anti-neoplastic immunotherapy. Mol. Biol. Rep. 2020, 47, 1435–1443. [Google Scholar] [CrossRef] [PubMed]
  131. Sarvi, S.; Crispin, R.; Lu, Y.; Zeng, L.; Hurley, T.D.; Houston, D.R.; von Kriegsheim, A.; Chen, C.H.; Mochly-Rosen, D.; Ranzani, M.; et al. ALDH1 Bio-activates Nifuroxazide to Eradicate ALDH(High) Melanoma-Initiating Cells. Cell Chem. Biol. 2018, 25, 1456–1469.e6. [Google Scholar] [CrossRef] [PubMed]
  132. Dinavahi, S.S.; Gowda, R.; Bazewicz, C.G.; Battu, M.B.; Lin, J.M.; Chitren, R.J.; Pandey, M.K.; Amin, S.; Robertson, G.P.; Gowda, K. Design, synthesis characterization and biological evaluation of novel multi-isoform ALDH inhibitors as potential anticancer agents. Eur. J. Med. Chem. 2020, 187, 111962. [Google Scholar] [CrossRef] [PubMed]
  133. Liao, F.; Zhang, J.; Hu, Y.; Najafabadi, A.H.; Moon, J.J.; Wicha, M.S.; Kaspo, B.; Whitfield, J.; Chang, A.E.; Li, Q. Efficacy of an ALDH peptide-based dendritic cell vaccine targeting cancer stem cells. Cancer Immunol. Immunother. 2022, 71, 1959–1973. [Google Scholar] [CrossRef] [PubMed]
  134. Boyer, L.A.; Lee, T.I.; Cole, M.F.; Johnstone, S.E.; Levine, S.S.; Zucker, J.P.; Guenther, M.G.; Kumar, R.M.; Murray, H.L.; Jenner, R.G.; et al. Core transcriptional regulatory circuitry in human embryonic stem cells. Cell 2005, 122, 947–956. [Google Scholar] [CrossRef] [PubMed]
  135. Kaufhold, S.; Garban, H.; Bonavida, B. Yin Yang 1 is associated with cancer stem cell transcription factors (SOX2, OCT4, BMI1) and clinical implication. J. Exp. Clin. Cancer Res. 2016, 35, 84. [Google Scholar] [CrossRef]
  136. Wang, Y.; Mou, Y.; Zhang, H.; Wang, X.; Li, R.; Cheng, Z.; Liu, X. Reprogramming Factors Remodel Melanoma Cell Phenotype by Changing Stat3 Expression. Int. J. Med. Sci. 2017, 14, 1402–1409. [Google Scholar] [CrossRef] [PubMed]
  137. Ravindran Menon, D.; Luo, Y.; Arcaroli, J.J.; Liu, S.; KrishnanKutty, L.N.; Osborne, D.G.; Li, Y.; Samson, J.M.; Bagby, S.; Tan, A.C.; et al. CDK1 Interacts with Sox2 and Promotes Tumor Initiation in Human Melanoma. Cancer Res. 2018, 78, 6561–6574. [Google Scholar] [CrossRef]
  138. Wang, Y.; Sun, H.J.; Li, R.G.; Wang, X.M.; Cheng, Z.Q.; Lou, N. Reprogramming factors induce proliferation and inhibit apoptosis of melanoma cells by changing the expression of particular genes. Mol. Med. Rep. 2019, 19, 967–973. [Google Scholar] [CrossRef] [PubMed]
  139. Wickremesekera, A.C.; Brasch, H.D.; Lee, V.M.; Davis, P.F.; Woon, K.; Johnson, R.; Tan, S.T.; Itinteang, T. Expression of cancer stem cell markers in metastatic melanoma to the brain. J. Clin. Neurosci. 2019, 60, 112–116. [Google Scholar] [CrossRef] [PubMed]
  140. Santini, R.; Pietrobono, S.; Pandolfi, S.; Montagnani, V.; D’Amico, M.; Penachioni, J.Y.; Vinci, M.C.; Borgognoni, L.; Stecca, B. SOX2 regulates self-renewal and tumorigenicity of human melanoma-initiating cells. Oncogene 2014, 33, 4697–4708. [Google Scholar] [CrossRef] [PubMed]
  141. Huser, L.; Sachindra, S.; Granados, K.; Federico, A.; Larribere, L.; Novak, D.; Umansky, V.; Altevogt, P.; Utikal, J. SOX2-mediated upregulation of CD24 promotes adaptive resistance toward targeted therapy in melanoma. Int. J. Cancer 2018, 143, 3131–3142. [Google Scholar] [CrossRef] [PubMed]
  142. Si, X.; Gao, Z.; Xu, F.; Zheng, Y. SOX2 upregulates side population cells and enhances their chemoresistant ability by transactivating ABCC1 expression contributing to intrinsic resistance to paclitaxel in melanoma. Mol. Carcinog. 2020, 59, 257–264. [Google Scholar] [CrossRef] [PubMed]
  143. Cordaro, F.G.; De Presbiteris, A.L.; Camerlingo, R.; Mozzillo, N.; Pirozzi, G.; Cavalcanti, E.; Manca, A.; Palmieri, G.; Cossu, A.; Ciliberto, G.; et al. Phenotype characterization of human melanoma cells resistant to dabrafenib. Oncol. Rep. 2017, 38, 2741–2751. [Google Scholar] [CrossRef] [PubMed]
  144. Silva, C.T.X.; Saddi, V.A.; Silva, K.; Sugita, D.M.; Guillo, L.A. Expression of the cancer stem cell marker OCT4 is associated with worse prognosis and survival in cutaneous melanoma. Melanoma Res. 2021, 31, 439–448. [Google Scholar] [CrossRef] [PubMed]
  145. Saito, M.; Kishi, R.; Sasai, T.; Hatakenaka, T.; Matsuki, N.; Minagawa, S. Effect of Nanog overexpression on the metastatic potential of a mouse melanoma cell line B16-BL6. Mol. Cell. Biochem. 2021, 476, 2651–2661. [Google Scholar] [CrossRef] [PubMed]
  146. Kim, S.; Cho, H.; Hong, S.O.; Oh, S.J.; Lee, H.J.; Cho, E.; Woo, S.R.; Song, J.S.; Chung, J.Y.; Son, S.W.; et al. LC3B upregulation by NANOG promotes immune resistance and stem-like property through hyperactivation of EGFR signaling in immune-refractory tumor cells. Autophagy 2021, 17, 1978–1997. [Google Scholar] [CrossRef] [PubMed]
  147. Khoo, C.S.M.; Henmi, T.; Saito, M. Comparative Study of Metastasis Suppression Effects of Extracellular Vesicles Derived from Anaplastic Cell Lines, Nanog-Overexpressing Melanoma, and Induced Pluripotent Stem Cells. Int. J. Mol. Sci. 2023, 24, 17206. [Google Scholar] [CrossRef] [PubMed]
  148. Matsui, W.H. Cancer stem cell signaling pathways. Medicine 2016, 95, S8–S19. [Google Scholar] [CrossRef] [PubMed]
  149. Katoh, M. Canonical and non-canonical WNT signaling in cancer stem cells and their niches: Cellular heterogeneity, omics reprogramming, targeted therapy and tumor plasticity (Review). Int. J. Oncol. 2017, 51, 1357–1369. [Google Scholar] [CrossRef] [PubMed]
  150. Nusse, R.; Clevers, H. Wnt/beta-Catenin Signaling, Disease, and Emerging Therapeutic Modalities. Cell 2017, 169, 985–999. [Google Scholar] [CrossRef] [PubMed]
  151. Valcourt, D.M.; Dang, M.N.; Wang, J.; Day, E.S. Nanoparticles for Manipulation of the Developmental Wnt, Hedgehog, and Notch Signaling Pathways in Cancer. Ann. Biomed. Eng. 2020, 48, 1864–1884. [Google Scholar] [CrossRef]
  152. Yang, L.; Shi, P.; Zhao, G.; Xu, J.; Peng, W.; Zhang, J.; Zhang, G.; Wang, X.; Dong, Z.; Chen, F.; et al. Targeting cancer stem cell pathways for cancer therapy. Signal Transduct. Target Ther. 2020, 5, 8. [Google Scholar] [CrossRef]
  153. Bugter, J.M.; Fenderico, N.; Maurice, M.M. Mutations and mechanisms of WNT pathway tumour suppressors in cancer. Nat. Rev. Cancer 2021, 21, 5–21. [Google Scholar] [CrossRef] [PubMed]
  154. Eid, R.A.; Alaa Edeen, M.; Shedid, E.M.; Kamal, A.S.S.; Warda, M.M.; Mamdouh, F.; Khedr, S.A.; Soltan, M.A.; Jeon, H.W.; Zaki, M.S.A.; et al. Targeting Cancer Stem Cells as the Key Driver of Carcinogenesis and Therapeutic Resistance. Int. J. Mol. Sci. 2023, 24, 1786. [Google Scholar] [CrossRef]
  155. Joshi, G.; Basu, A. Epigenetic control of cell signalling in cancer stem cells. Int. Rev. Cell Mol. Biol. 2024, 383, 67–88. [Google Scholar] [CrossRef] [PubMed]
  156. Giannandrea, D.; Platonova, N.; Colombo, M.; Mazzola, M.; Citro, V.; Adami, R.; Maltoni, F.; Ancona, S.; Dolo, V.; Giusti, I.; et al. Extracellular vesicles mediate the communication between multiple myeloma and bone marrow microenvironment in a NOTCH dependent way. Haematologica 2022, 107, 2183–2194. [Google Scholar] [CrossRef]
  157. Kim, Y.M.; Kahn, M. The role of the Wnt signaling pathway in cancer stem cells: Prospects for drug development. Res. Rep. Biochem. 2014, 4, 1–12. [Google Scholar] [CrossRef] [PubMed]
  158. Duchartre, Y.; Kim, Y.M.; Kahn, M. The Wnt signaling pathway in cancer. Crit. Rev. Oncol. Hematol. 2016, 99, 141–149. [Google Scholar] [CrossRef]
  159. Zhang, Y.; Wang, X. Targeting the Wnt/beta-catenin signaling pathway in cancer. J. Hematol. Oncol. 2020, 13, 165. [Google Scholar] [CrossRef]
  160. Hiremath, I.S.; Goel, A.; Warrier, S.; Kumar, A.P.; Sethi, G.; Garg, M. The multidimensional role of the Wnt/beta-catenin signaling pathway in human malignancies. J. Cell. Physiol. 2022, 237, 199–238. [Google Scholar] [CrossRef]
  161. Katoh, M.; Katoh, M. WNT signaling and cancer stemness. Essays Biochem. 2022, 66, 319–331. [Google Scholar] [CrossRef] [PubMed]
  162. Yu, M.; Qin, K.; Fan, J.; Zhao, G.; Zhao, P.; Zeng, W.; Chen, C.; Wang, A.; Wang, Y.; Zhong, J.; et al. The evolving roles of Wnt signaling in stem cell proliferation and differentiation, the development of human diseases, and therapeutic opportunities. Genes Dis. 2024, 11, 101026. [Google Scholar] [CrossRef] [PubMed]
  163. Tiwary, S.; Xu, L. FRIZZLED7 Is Required for Tumor Initiation and Metastatic Growth of Melanoma Cells. PLoS ONE 2016, 11, e0147638. [Google Scholar] [CrossRef] [PubMed]
  164. Rappa, G.; Mercapide, J.; Anzanello, F.; Le, T.T.; Johlfs, M.G.; Fiscus, R.R.; Wilsch-Brauninger, M.; Corbeil, D.; Lorico, A. Wnt interaction and extracellular release of prominin-1/CD133 in human malignant melanoma cells. Exp. Cell Res. 2013, 319, 810–819. [Google Scholar] [CrossRef] [PubMed]
  165. Wei, C.Y.; Zhu, M.X.; Yang, Y.W.; Zhang, P.F.; Yang, X.; Peng, R.; Gao, C.; Lu, J.C.; Wang, L.; Deng, X.Y.; et al. Downregulation of RNF128 activates Wnt/beta-catenin signaling to induce cellular EMT and stemness via CD44 and CTTN ubiquitination in melanoma. J. Hematol. Oncol. 2019, 12, 21. [Google Scholar] [CrossRef] [PubMed]
  166. Li, J.; Fang, R.; Wu, J.; Si, Y.; Bai, J.; Wang, Q. The NOP14 nucleolar protein suppresses the function and stemness of melanoma stem-like cells through Wnt/beta-catenin signaling inactivation. Bioengineered 2022, 13, 7648–7658. [Google Scholar] [CrossRef]
  167. Sellerio, A.L.; Ciusani, E.; Ben-Moshe, N.B.; Coco, S.; Piccinini, A.; Myers, C.R.; Sethna, J.P.; Giampietro, C.; Zapperi, S.; La Porta, C.A. Overshoot during phenotypic switching of cancer cell populations. Sci. Rep. 2015, 5, 15464. [Google Scholar] [CrossRef] [PubMed]
  168. Liu, S.; Gao, X.; Zhang, L.; Qin, S.; Wei, M.; Liu, N.; Zhao, R.; Li, B.; Meng, Y.; Lin, G.; et al. A novel Anti-Cancer Stem Cells compound optimized from the natural symplostatin 4 scaffold inhibits Wnt/beta-catenin signaling pathway. Eur. J. Med. Chem. 2018, 156, 21–42. [Google Scholar] [CrossRef] [PubMed]
  169. Goncalves, J.M.; Silva, C.A.B.; Rivero, E.R.C.; Cordeiro, M.M.R. Inhibition of cancer stem cells promoted by Pimozide. Clin. Exp. Pharmacol. Physiol. 2019, 46, 116–125. [Google Scholar] [CrossRef] [PubMed]
  170. Javadi, A.; Keighobadi, F.; Nekoukar, V.; Ebrahimi, M. Finite-Set Model Predictive Control of Melanoma Cancer Treatment Using Signaling Pathway Inhibitor of Cancer Stem Cell. IEEE/ACM Trans Comput. Biol. Bioinform. 2021, 18, 1504–1511. [Google Scholar] [CrossRef]
  171. Xia, R.; Xu, M.; Yang, J.; Ma, X. The role of Hedgehog and Notch signaling pathway in cancer. Mol. Biomed. 2022, 3, 44. [Google Scholar] [CrossRef] [PubMed]
  172. Iso, T.; Kedes, L.; Hamamori, Y. HES and HERP families: Multiple effectors of the Notch signaling pathway. J. Cell. Physiol. 2003, 194, 237–255. [Google Scholar] [CrossRef] [PubMed]
  173. Palomero, T.; Lim, W.K.; Odom, D.T.; Sulis, M.L.; Real, P.J.; Margolin, A.; Barnes, K.C.; O’Neil, J.; Neuberg, D.; Weng, A.P.; et al. NOTCH1 directly regulates c-MYC and activates a feed-forward-loop transcriptional network promoting leukemic cell growth. Proc. Natl. Acad. Sci. USA 2006, 103, 18261–18266. [Google Scholar] [CrossRef] [PubMed]
  174. Takebe, N.; Miele, L.; Harris, P.J.; Jeong, W.; Bando, H.; Kahn, M.; Yang, S.X.; Ivy, S.P. Targeting Notch, Hedgehog, and Wnt pathways in cancer stem cells: Clinical update. Nat. Rev. Clin. Oncol. 2015, 12, 445–464. [Google Scholar] [CrossRef] [PubMed]
  175. Jin, Y.; Wang, M.; Hu, H.; Huang, Q.; Chen, Y.; Wang, G. Overcoming stemness and chemoresistance in colorectal cancer through miR-195-5p-modulated inhibition of notch signaling. Int. J. Biol. Macromol. 2018, 117, 445–453. [Google Scholar] [CrossRef] [PubMed]
  176. Ferreira, A.; Aster, J.C. Notch signaling in cancer: Complexity and challenges on the path to clinical translation. Semin. Cancer Biol. 2022, 85, 95–106. [Google Scholar] [CrossRef] [PubMed]
  177. Ranganathan, P.; Weaver, K.L.; Capobianco, A.J. Notch signalling in solid tumours: A little bit of everything but not all the time. Nat. Rev. Cancer 2011, 11, 338–351. [Google Scholar] [CrossRef] [PubMed]
  178. Janghorban, M.; Xin, L.; Rosen, J.M.; Zhang, X.H. Notch Signaling as a Regulator of the Tumor Immune Response: To Target or Not To Target? Front. Immunol. 2018, 9, 1649. [Google Scholar] [CrossRef] [PubMed]
  179. Lin, X.; Sun, B.; Zhu, D.; Zhao, X.; Sun, R.; Zhang, Y.; Zhang, D.; Dong, X.; Gu, Q.; Li, Y.; et al. Notch4+ cancer stem-like cells promote the metastatic and invasive ability of melanoma. Cancer Sci. 2016, 107, 1079–1091. [Google Scholar] [CrossRef] [PubMed]
  180. Kumar, D.; Kumar, S.; Gorain, M.; Tomar, D.; Patil, H.S.; Radharani, N.N.V.; Kumar, T.V.S.; Patil, T.V.; Thulasiram, H.V.; Kundu, G.C. Notch1-MAPK Signaling Axis Regulates CD133(+) Cancer Stem Cell-Mediated Melanoma Growth and Angiogenesis. J. Investig. Dermatol. 2016, 136, 2462–2474. [Google Scholar] [CrossRef] [PubMed]
  181. Ciccone, V.; Terzuoli, E.; Ristori, E.; Filippelli, A.; Ziche, M.; Morbidelli, L.; Donnini, S. ALDH1A1 overexpression in melanoma cells promotes tumor angiogenesis by activating the IL-8/Notch signaling cascade. Int. J. Mol. Med. 2022, 50, 99. [Google Scholar] [CrossRef] [PubMed]
  182. Hsu, M.Y.; Yang, M.H.; Schnegg, C.I.; Hwang, S.; Ryu, B.; Alani, R.M. Notch3 signaling-mediated melanoma-endothelial crosstalk regulates melanoma stem-like cell homeostasis and niche morphogenesis. Lab. Investig. 2017, 97, 725–736. [Google Scholar] [CrossRef] [PubMed]
  183. Stecca, B.; Mas, C.; Clement, V.; Zbinden, M.; Correa, R.; Piguet, V.; Beermann, F.; Ruiz i Altaba, A. Melanomas require HEDGEHOG-GLI signaling regulated by interactions between GLI1 and the RAS-MEK/AKT pathways. Proc. Natl. Acad. Sci. USA 2007, 104, 5895–5900. [Google Scholar] [CrossRef] [PubMed]
  184. Jiang, J.; Hui, C.C. Hedgehog signaling in development and cancer. Dev. Cell 2008, 15, 801–812. [Google Scholar] [CrossRef] [PubMed]
  185. Jing, J.; Wu, Z.; Wang, J.; Luo, G.; Lin, H.; Fan, Y.; Zhou, C. Hedgehog signaling in tissue homeostasis, cancers, and targeted therapies. Signal Transduct. Target. Ther. 2023, 8, 315. [Google Scholar] [CrossRef] [PubMed]
  186. Bhateja, P.; Cherian, M.; Majumder, S.; Ramaswamy, B. The Hedgehog Signaling Pathway: A Viable Target in Breast Cancer? Cancers 2019, 11, 1126. [Google Scholar] [CrossRef] [PubMed]
  187. Cochrane, C.R.; Szczepny, A.; Watkins, D.N.; Cain, J.E. Hedgehog Signaling in the Maintenance of Cancer Stem Cells. Cancers 2015, 7, 1554–1585. [Google Scholar] [CrossRef] [PubMed]
  188. Jeng, K.S.; Chang, C.F.; Lin, S.S. Sonic Hedgehog Signaling in Organogenesis, Tumors, and Tumor Microenvironments. Int. J. Mol. Sci. 2020, 21, 758. [Google Scholar] [CrossRef] [PubMed]
  189. Faiao-Flores, F.; Alves-Fernandes, D.K.; Pennacchi, P.C.; Sandri, S.; Vicente, A.L.; Scapulatempo-Neto, C.; Vazquez, V.L.; Reis, R.M.; Chauhan, J.; Goding, C.R.; et al. Targeting the hedgehog transcription factors GLI1 and GLI2 restores sensitivity to vemurafenib-resistant human melanoma cells. Oncogene 2017, 36, 1849–1861. [Google Scholar] [CrossRef] [PubMed]
  190. Santini, R.; Vinci, M.C.; Pandolfi, S.; Penachioni, J.Y.; Montagnani, V.; Olivito, B.; Gattai, R.; Pimpinelli, N.; Gerlini, G.; Borgognoni, L.; et al. Hedgehog-GLI signaling drives self-renewal and tumorigenicity of human melanoma-initiating cells. Stem Cells 2012, 30, 1808–1818. [Google Scholar] [CrossRef] [PubMed]
  191. Horak, P.; Kreisingerova, K.; Reda, J.; Ondrusova, L.; Balko, J.; Vachtenheim, J., Jr.; Zakova, P.; Vachtenheim, J. The Hedgehog/GLI signaling pathway activates transcription of Slug (Snail2) in melanoma cells. Oncol. Rep. 2023, 49, 1–12. [Google Scholar] [CrossRef] [PubMed]
  192. Davies, M.A. The role of the PI3K-AKT pathway in melanoma. Cancer J. 2012, 18, 142–147. [Google Scholar] [CrossRef] [PubMed]
  193. Kakadia, S.; Yarlagadda, N.; Awad, R.; Kundranda, M.; Niu, J.; Naraev, B.; Mina, L.; Dragovich, T.; Gimbel, M.; Mahmoud, F. Mechanisms of resistance to BRAF and MEK inhibitors and clinical update of US Food and Drug Administration-approved targeted therapy in advanced melanoma. Onco Targets Ther. 2018, 11, 7095–7107. [Google Scholar] [CrossRef] [PubMed]
  194. Chamcheu, J.C.; Roy, T.; Uddin, M.B.; Banang-Mbeumi, S.; Chamcheu, R.N.; Walker, A.L.; Liu, Y.Y.; Huang, S. Role and Therapeutic Targeting of the PI3K/Akt/mTOR Signaling Pathway in Skin Cancer: A Review of Current Status and Future Trends on Natural and Synthetic Agents Therapy. Cells 2019, 8, 803. [Google Scholar] [CrossRef] [PubMed]
  195. Olbryt, M. Potential Biomarkers of Skin Melanoma Resistance to Targeted Therapy-Present State and Perspectives. Cancers 2022, 14, 2315. [Google Scholar] [CrossRef] [PubMed]
  196. Bou Antoun, N.; Chioni, A.M. Dysregulated Signalling Pathways Driving Anticancer Drug Resistance. Int. J. Mol. Sci. 2023, 24, 12222. [Google Scholar] [CrossRef] [PubMed]
  197. Florent, L.; Saby, C.; Slimano, F.; Morjani, H. BRAF V600-Mutated Metastatic Melanoma and Targeted Therapy Resistance: An Update of the Current Knowledge. Cancers 2023, 15, 2607. [Google Scholar] [CrossRef] [PubMed]
  198. Khorasani, A.B.S.; Hafezi, N.; Sanaei, M.J.; Jafari-Raddani, F.; Pourbagheri-Sigaroodi, A.; Bashash, D. The PI3K/AKT/mTOR signaling pathway in breast cancer: Review of clinical trials and latest advances. Cell Biochem. Funct. 2024, 42, e3998. [Google Scholar] [CrossRef] [PubMed]
  199. Hemmings, B.A.; Restuccia, D.F. PI3K-PKB/Akt pathway. Cold Spring Harb. Perspect. Biol. 2012, 4, a011189. [Google Scholar] [CrossRef] [PubMed]
  200. Thorpe, L.M.; Yuzugullu, H.; Zhao, J.J. PI3K in cancer: Divergent roles of isoforms, modes of activation and therapeutic targeting. Nat. Rev. Cancer 2015, 15, 7–24. [Google Scholar] [CrossRef] [PubMed]
  201. Afify, S.M.; Oo, A.K.K.; Hassan, G.; Seno, A.; Seno, M. How can we turn the PI3K/AKT/mTOR pathway down? Insights into inhibition and treatment of cancer. Expert Rev. Anticancer Ther. 2021, 21, 605–619. [Google Scholar] [CrossRef]
  202. He, Y.; Sun, M.M.; Zhang, G.G.; Yang, J.; Chen, K.S.; Xu, W.W.; Li, B. Targeting PI3K/Akt signal transduction for cancer therapy. Signal Transduct. Target. Ther. 2021, 6, 425. [Google Scholar] [CrossRef] [PubMed]
  203. Glaviano, A.; Foo, A.S.C.; Lam, H.Y.; Yap, K.C.H.; Jacot, W.; Jones, R.H.; Eng, H.; Nair, M.G.; Makvandi, P.; Geoerger, B.; et al. PI3K/AKT/mTOR signaling transduction pathway and targeted therapies in cancer. Mol. Cancer 2023, 22, 138. [Google Scholar] [CrossRef] [PubMed]
  204. Matsuda, S.; Ichimura, M.; Ogino, M.; Nakano, N.; Minami, A.; Murai, T.; Kitagishi, Y. Effective PI3K modulators for improved therapy against malignant tumors and for neuroprotection of brain damage after tumor therapy (Review). Int. J. Oncol. 2016, 49, 1785–1790. [Google Scholar] [CrossRef] [PubMed]
  205. Radic, M.; Vlasic, I.; Jazvinscak Jembrek, M.; Horvat, A.; Tadijan, A.; Sabol, M.; Duzevic, M.; Herak Bosnar, M.; Slade, N. Characterization of Vemurafenib-Resistant Melanoma Cell Lines Reveals Novel Hallmarks of Targeted Therapy Resistance. Int. J. Mol. Sci. 2022, 23, 9910. [Google Scholar] [CrossRef] [PubMed]
  206. Tovari, J.; Vari-Mezo, D.; Surguta, S.E.; Ladanyi, A.; Kigyos, A.; Cserepes, M. Evolving Acquired Vemurafenib Resistance in a BRAF V600E Mutant Melanoma PDTX Model to Reveal New Potential Targets. Cells 2023, 12, 1919. [Google Scholar] [CrossRef] [PubMed]
  207. Corrales, E.; Levit-Zerdoun, E.; Metzger, P.; Mertes, R.; Lehmann, A.; Munch, J.; Lemke, S.; Kowar, S.; Boerries, M. PI3K/AKT signaling allows for MAPK/ERK pathway independency mediating dedifferentiation-driven treatment resistance in melanoma. Cell Commun. Signal. 2022, 20, 187. [Google Scholar] [CrossRef] [PubMed]
  208. Maniotis, A.J.; Folberg, R.; Hess, A.; Seftor, E.A.; Gardner, L.M.; Pe’er, J.; Trent, J.M.; Meltzer, P.S.; Hendrix, M.J. Vascular channel formation by human melanoma cells in vivo and in vitro: Vasculogenic mimicry. Am. J. Pathol. 1999, 155, 739–752. [Google Scholar] [CrossRef] [PubMed]
  209. Valyi-Nagy, K.; Kormos, B.; Ali, M.; Shukla, D.; Valyi-Nagy, T. Stem cell marker CD271 is expressed by vasculogenic mimicry-forming uveal melanoma cells in three-dimensional cultures. Mol. Vis. 2012, 18, 588–592. [Google Scholar] [PubMed]
  210. Hess, A.R.; Margaryan, N.V.; Seftor, E.A.; Hendrix, M.J. Deciphering the signaling events that promote melanoma tumor cell vasculogenic mimicry and their link to embryonic vasculogenesis: Role of the Eph receptors. Dev. Dyn. 2007, 236, 3283–3296. [Google Scholar] [CrossRef] [PubMed]
  211. Murai, T.; Matsuda, S. Targeting the PI3K-Akt-mTOR signaling pathway involved in vasculogenic mimicry promoted by cancer stem cells. Am. J. Cancer Res. 2023, 13, 5039–5046. [Google Scholar]
  212. Simiczyjew, A.; Dratkiewicz, E.; Mazurkiewicz, J.; Zietek, M.; Matkowski, R.; Nowak, D. The Influence of Tumor Microenvironment on Immune Escape of Melanoma. Int. J. Mol. Sci. 2020, 21, 8359. [Google Scholar] [CrossRef] [PubMed]
  213. Mazurkiewicz, J.; Simiczyjew, A.; Dratkiewicz, E.; Zietek, M.; Matkowski, R.; Nowak, D. Stromal Cells Present in the Melanoma Niche Affect Tumor Invasiveness and Its Resistance to Therapy. Int. J. Mol. Sci. 2021, 22, 529. [Google Scholar] [CrossRef]
  214. Amalinei, C.; Grigoras, A.; Lozneanu, L.; Caruntu, I.D.; Giusca, S.E.; Balan, R.A. The Interplay between Tumour Microenvironment Components in Malignant Melanoma. Medicina 2022, 58, 365. [Google Scholar] [CrossRef] [PubMed]
  215. Fontana, F.; Anselmi, M.; Limonta, P. Adipocytes reprogram prostate cancer stem cell machinery. J. Cell Commun. Signal. 2023, 17, 915–924. [Google Scholar] [CrossRef]
  216. Zhao, Y.; Shen, M.; Wu, L.; Yang, H.; Yao, Y.; Yang, Q.; Du, J.; Liu, L.; Li, Y.; Bai, Y. Stromal cells in the tumor microenvironment: Accomplices of tumor progression? Cell Death Dis. 2023, 14, 587. [Google Scholar] [CrossRef]
  217. Pretti, M.A.M.; Bernardes, S.S.; da Cruz, J.G.V.; Boroni, M.; Possik, P.A. Extracellular vesicle-mediated crosstalk between melanoma and the immune system: Impact on tumor progression and therapy response. J. Leukoc. Biol. 2020, 108, 1101–1115. [Google Scholar] [CrossRef]
  218. Benjamin-Davalos, S.; Koroleva, M.; Allen, C.L.; Ernstoff, M.S.; Shu, S. Co-Isolation of Cytokines and Exosomes: Implications for Immunomodulation Studies. Front. Immunol. 2021, 12, 638111. [Google Scholar] [CrossRef]
  219. Boussadia, Z.; Gambardella, A.R.; Mattei, F.; Parolini, I. Acidic and Hypoxic Microenvironment in Melanoma: Impact of Tumour Exosomes on Disease Progression. Cells 2021, 10, 3311. [Google Scholar] [CrossRef] [PubMed]
  220. Fontana, F.; Anselmi, M.; Carollo, E.; Sartori, P.; Procacci, P.; Carter, D.; Limonta, P. Adipocyte-Derived Extracellular Vesicles Promote Prostate Cancer Cell Aggressiveness by Enabling Multiple Phenotypic and Metabolic Changes. Cells 2022, 11, 2388. [Google Scholar] [CrossRef]
  221. Musi, A.; Bongiovanni, L. Extracellular Vesicles in Cancer Drug Resistance: Implications on Melanoma Therapy. Cancers 2023, 15, 1074. [Google Scholar] [CrossRef] [PubMed]
  222. Zhou, X.; Jia, Y.; Mao, C.; Liu, S. Small extracellular vesicles: Non-negligible vesicles in tumor progression, diagnosis, and therapy. Cancer Lett. 2024, 580, 216481. [Google Scholar] [CrossRef] [PubMed]
  223. Guha, A.; Goswami, K.K.; Sultana, J.; Ganguly, N.; Choudhury, P.R.; Chakravarti, M.; Bhuniya, A.; Sarkar, A.; Bera, S.; Dhar, S.; et al. Cancer stem cell-immune cell crosstalk in breast tumor microenvironment: A determinant of therapeutic facet. Front. Immunol. 2023, 14, 1245421. [Google Scholar] [CrossRef] [PubMed]
  224. Zaarour, R.F.; Ribeiro, M.; Azzarone, B.; Kapoor, S.; Chouaib, S. Tumor microenvironment-induced tumor cell plasticity: Relationship with hypoxic stress and impact on tumor resistance. Front. Oncol. 2023, 13, 1222575. [Google Scholar] [CrossRef]
  225. Zhao, Q.; Zong, H.; Zhu, P.; Su, C.; Tang, W.; Chen, Z.; Jin, S. Crosstalk between colorectal CSCs and immune cells in tumorigenesis, and strategies for targeting colorectal CSCs. Exp. Hematol. Oncol. 2024, 13, 6. [Google Scholar] [CrossRef] [PubMed]
  226. Passarelli, A.; Mannavola, F.; Stucci, L.S.; Tucci, M.; Silvestris, F. Immune system and melanoma biology: A balance between immunosurveillance and immune escape. Oncotarget 2017, 8, 106132–106142. [Google Scholar] [CrossRef] [PubMed]
  227. Fruntealata, R.F.; Marius, M.; Boboc, I.K.S.; Mitran, S.I.; Ciurea, M.E.; Stoica, G.A. Mechanisms of Altered Immune Response in Skin Melanoma. Curr. Health Sci. J. 2023, 49, 297–311. [Google Scholar] [CrossRef] [PubMed]
  228. Benboubker, V.; Boivin, F.; Dalle, S.; Caramel, J. Cancer Cell Phenotype Plasticity as a Driver of Immune Escape in Melanoma. Front. Immunol. 2022, 13, 873116. [Google Scholar] [CrossRef] [PubMed]
  229. Habib, S.; Osborn, G.; Willsmore, Z.; Chew, M.W.; Jakubow, S.; Fitzpatrick, A.; Wu, Y.; Sinha, K.; Lloyd-Hughes, H.; Geh, J.L.C.; et al. Tumor associated macrophages as key contributors and targets in current and future therapies for melanoma. Expert Rev. Clin. Immunol. 2024, 20, 895–911. [Google Scholar] [CrossRef]
  230. Maccalli, C.; Volonte, A.; Cimminiello, C.; Parmiani, G. Immunology of cancer stem cells in solid tumours. A review. Eur. J. Cancer 2014, 50, 649–655. [Google Scholar] [CrossRef]
  231. Shirley, C.A.; Chhabra, G.; Amiri, D.; Chang, H.; Ahmad, N. Immune escape and metastasis mechanisms in melanoma: Breaking down the dichotomy. Front. Immunol. 2024, 15, 1336023. [Google Scholar] [CrossRef] [PubMed]
  232. Zhang, D.; Tang, D.G.; Rycaj, K. Cancer stem cells: Regulation programs, immunological properties and immunotherapy. Semin. Cancer Biol. 2018, 52, 94–106. [Google Scholar] [CrossRef] [PubMed]
  233. Wang, X.; Wan, Q.; Jin, L.; Liu, C.; Liu, C.; Cheng, Y.; Wang, Z. The Integrative Analysis Identifies Three Cancer Subtypes and Stemness Features in Cutaneous Melanoma. Front. Mol. Biosci. 2020, 7, 598725. [Google Scholar] [CrossRef] [PubMed]
  234. Cormier, J.N.; Salgaller, M.L.; Prevette, T.; Barracchini, K.C.; Rivoltini, L.; Restifo, N.P.; Rosenberg, S.A.; Marincola, F.M. Enhancement of cellular immunity in melanoma patients immunized with a peptide from MART-1/Melan A. Cancer J. Sci. Am. 1997, 3, 37–44. [Google Scholar] [PubMed]
  235. Reynolds, S.R.; Celis, E.; Sette, A.; Oratz, R.; Shapiro, R.L.; Johnston, D.; Fotino, M.; Bystryn, J.C. HLA-independent heterogeneity of CD8+ T cell responses to MAGE-3, Melan-A/MART-1, gp100, tyrosinase, MC1R, and TRP-2 in vaccine-treated melanoma patients. J. Immunol. 1998, 161, 6970–6976. [Google Scholar] [CrossRef] [PubMed]
  236. Kawakami, Y.; Dang, N.; Wang, X.; Tupesis, J.; Robbins, P.F.; Wang, R.F.; Wunderlich, J.R.; Yannelli, J.R.; Rosenberg, S.A. Recognition of shared melanoma antigens in association with major HLA-A alleles by tumor infiltrating T lymphocytes from 123 patients with melanoma. J. Immunother. 2000, 23, 17–27. [Google Scholar] [CrossRef] [PubMed]
  237. Schatton, T.; Frank, M.H. Antitumor immunity and cancer stem cells. Ann. N. Y. Acad. Sci. 2009, 1176, 154–169. [Google Scholar] [CrossRef] [PubMed]
  238. Schatton, T.; Schutte, U.; Frank, N.Y.; Zhan, Q.; Hoerning, A.; Robles, S.C.; Zhou, J.; Hodi, F.S.; Spagnoli, G.C.; Murphy, G.F.; et al. Modulation of T cell activation by malignant melanoma initiating cells. Cancer Res. 2010, 70, 697–708. [Google Scholar] [CrossRef] [PubMed]
  239. Landsberg, J.; Kohlmeyer, J.; Renn, M.; Bald, T.; Rogava, M.; Cron, M.; Fatho, M.; Lennerz, V.; Wolfel, T.; Holzel, M.; et al. Melanomas resist T cell therapy through inflammation-induced reversible dedifferentiation. Nature 2012, 490, 412–416. [Google Scholar] [CrossRef] [PubMed]
  240. Mehta, A.; Kim, Y.J.; Robert, L.; Tsoi, J.; Comin-Anduix, B.; Berent-Maoz, B.; Cochran, A.J.; Economou, J.S.; Tumeh, P.C.; Puig-Saus, C.; et al. Immunotherapy Resistance by Inflammation-Induced Dedifferentiation. Cancer Discov. 2018, 8, 935–943. [Google Scholar] [CrossRef]
  241. Boshuizen, J.; Vredevoogd, D.W.; Krijgsman, O.; Ligtenberg, M.A.; Blankenstein, S.; de Bruijn, B.; Frederick, D.T.; Kenski, J.C.N.; Parren, M.; Bruggemann, M.; et al. Reversal of pre-existing NGFR-driven tumor and immune therapy resistance. Nat. Commun. 2020, 11, 3946. [Google Scholar] [CrossRef] [PubMed]
  242. Huang, F.; Goncalves, C.; Bartish, M.; Remy-Sarrazin, J.; Issa, M.E.; Cordeiro, B.; Guo, Q.; Emond, A.; Attias, M.; Yang, W.; et al. Inhibiting the MNK1/2-eIF4E axis impairs melanoma phenotype switching and potentiates antitumor immune responses. J. Clin. Investig. 2021, 131, e140752. [Google Scholar] [CrossRef] [PubMed]
  243. Gedye, C.; Quirk, J.; Browning, J.; Svobodova, S.; John, T.; Sluka, P.; Dunbar, P.R.; Corbeil, D.; Cebon, J.; Davis, I.D. Cancer/testis antigens can be immunological targets in clonogenic CD133+ melanoma cells. Cancer Immunol. Immunother. 2009, 58, 1635–1646. [Google Scholar] [CrossRef] [PubMed]
  244. Schumacher, T.N.; Schreiber, R.D. Neoantigens in cancer immunotherapy. Science 2015, 348, 69–74. [Google Scholar] [CrossRef] [PubMed]
  245. Klimczak, M.; Czerwinska, P.; Mazurek, S.; Sozanska, B.; Biecek, P.; Mackiewicz, A.; Wiznerowicz, M. TRIM28 epigenetic corepressor is indispensable for stable induced pluripotent stem cell formation. Stem Cell Res. 2017, 23, 163–172. [Google Scholar] [CrossRef] [PubMed]
  246. Czerwinska, P.; Jaworska, A.M.; Wlodarczyk, N.A.; Mackiewicz, A.A. Melanoma Stem Cell-Like Phenotype and Significant Suppression of Immune Response within a Tumor Are Regulated by TRIM28 Protein. Cancers 2020, 12, 2998. [Google Scholar] [CrossRef]
  247. Wu, R.; Wang, C.; Li, Z.; Xiao, J.; Li, C.; Wang, X.; Kong, P.; Cao, J.; Huang, F.; Li, Z.; et al. SOX2 promotes resistance of melanoma with PD-L1 high expression to T cell-mediated cytotoxicity that can be reversed by SAHA. J. Immunother. Cancer. 2020, 8, e001037. [Google Scholar] [CrossRef] [PubMed]
  248. Abel, A.M.; Yang, C.; Thakar, M.S.; Malarkannan, S. Natural Killer Cells: Development, Maturation, and Clinical Utilization. Front. Immunol. 2018, 9, 1869. [Google Scholar] [CrossRef]
  249. Melaiu, O.; Lucarini, V.; Cifaldi, L.; Fruci, D. Influence of the Tumor Microenvironment on NK Cell Function in Solid Tumors. Front. Immunol. 2019, 10, 3038. [Google Scholar] [CrossRef]
  250. Huergo-Zapico, L.; Parodi, M.; Cantoni, C.; Lavarello, C.; Fernandez-Martinez, J.L.; Petretto, A.; DeAndres-Galiana, E.J.; Balsamo, M.; Lopez-Soto, A.; Pietra, G.; et al. NK-cell Editing Mediates Epithelial-to-Mesenchymal Transition via Phenotypic and Proteomic Changes in Melanoma Cell Lines. Cancer Res. 2018, 78, 3913–3925. [Google Scholar] [CrossRef]
  251. Cheli, Y.; Tulic, M.K.; El Hachem, N.; Nottet, N.; Jacquel, A.; Gesson, M.; Strub, T.; Bille, K.; Picard-Gauci, A.; Montaudie, H.; et al. ITGBL1 is a new immunomodulator that favors development of melanoma tumors by inhibiting natural killer cells cytotoxicity. Mol. Cancer 2021, 20, 12. [Google Scholar] [CrossRef] [PubMed]
  252. Calvani, M.; Dabraio, A.; Bruno, G.; De Gregorio, V.; Coronnello, M.; Bogani, C.; Ciullini, S.; Marca, G.; Vignoli, M.; Chiarugi, P.; et al. beta3-Adrenoreceptor Blockade Reduces Hypoxic Myeloid Leukemic Cells Survival and Chemoresistance. Int. J. Mol. Sci. 2020, 21, 4210. [Google Scholar] [CrossRef] [PubMed]
  253. Lehmann, J.; Caduff, N.; Krzywinska, E.; Stierli, S.; Salas-Bastos, A.; Loos, B.; Levesque, M.P.; Dummer, R.; Stockmann, C.; Munz, C.; et al. Escape from NK cell tumor surveillance by NGFR-induced lipid remodeling in melanoma. Sci. Adv. 2023, 9, eadc8825. [Google Scholar] [CrossRef] [PubMed]
  254. Touboul, R.; Zaravinos, A.; Bonavida, B. Defective Natural Killer Cells in Melanoma: Role of NKG2D in Pathogenesis and Immunotherapy. Crit. Rev. Immunol. 2021, 41, 45–76. [Google Scholar] [CrossRef] [PubMed]
  255. Tie, Y.; Tang, F.; Wei, Y.Q.; Wei, X.W. Immunosuppressive cells in cancer: Mechanisms and potential therapeutic targets. J. Hematol. Oncol. 2022, 15, 61. [Google Scholar] [CrossRef] [PubMed]
  256. Lecot, P.; Sarabi, M.; Pereira Abrantes, M.; Mussard, J.; Koenderman, L.; Caux, C.; Bendriss-Vermare, N.; Michallet, M.C. Neutrophil Heterogeneity in Cancer: From Biology to Therapies. Front. Immunol. 2019, 10, 2155. [Google Scholar] [CrossRef] [PubMed]
  257. Mukaida, N.; Sasaki, S.I.; Baba, T. Two-Faced Roles of Tumor-Associated Neutrophils in Cancer Development and Progression. Int. J. Mol. Sci. 2020, 21, 3457. [Google Scholar] [CrossRef] [PubMed]
  258. Wang, X.; Qiu, L.; Li, Z.; Wang, X.Y.; Yi, H. Understanding the Multifaceted Role of Neutrophils in Cancer and Autoimmune Diseases. Front. Immunol. 2018, 9, 2456. [Google Scholar] [CrossRef] [PubMed]
  259. Modestino, L.; Cristinziano, L.; Trocchia, M.; Ventrici, A.; Capone, M.; Madonna, G.; Loffredo, S.; Ferrara, A.L.; Romanelli, M.; Simeone, E.; et al. Melanoma-derived soluble mediators modulate neutrophil biological properties and the release of neutrophil extracellular traps. Cancer Immunol. Immunother. 2023, 72, 3363–3376. [Google Scholar] [CrossRef] [PubMed]
  260. Ferrucci, P.F.; Ascierto, P.A.; Pigozzo, J.; Del Vecchio, M.; Maio, M.; Antonini Cappellini, G.C.; Guidoboni, M.; Queirolo, P.; Savoia, P.; Mandala, M.; et al. Baseline neutrophils and derived neutrophil-to-lymphocyte ratio: Prognostic relevance in metastatic melanoma patients receiving ipilimumab. Ann. Oncol. 2016, 27, 732–738. [Google Scholar] [CrossRef] [PubMed]
  261. Anselmi, M.; Fontana, F.; Marzagalli, M.; Gagliano, N.; Sommariva, M.; Limonta, P. Melanoma Stem Cells Educate Neutrophils to Support Cancer Progression. Cancers 2022, 14, 3391. [Google Scholar] [CrossRef] [PubMed]
  262. Murray, P.J. Macrophage Polarization. Annu. Rev. Physiol. 2017, 79, 541–566. [Google Scholar] [CrossRef] [PubMed]
  263. Yunna, C.; Mengru, H.; Lei, W.; Weidong, C. Macrophage M1/M2 polarization. Eur. J. Pharmacol. 2020, 877, 173090. [Google Scholar] [CrossRef] [PubMed]
  264. Gao, J.; Liang, Y.; Wang, L. Shaping Polarization Of Tumor-Associated Macrophages In Cancer Immunotherapy. Front. Immunol. 2022, 13, 888713. [Google Scholar] [CrossRef] [PubMed]
  265. Su, X.; Esser, A.K.; Amend, S.R.; Xiang, J.; Xu, Y.; Ross, M.H.; Fox, G.C.; Kobayashi, T.; Steri, V.; Roomp, K.; et al. Antagonizing Integrin beta3 Increases Immunosuppression in Cancer. Cancer Res. 2016, 76, 3484–3495. [Google Scholar] [CrossRef] [PubMed]
  266. Yamada, K.; Uchiyama, A.; Uehara, A.; Perera, B.; Ogino, S.; Yokoyama, Y.; Takeuchi, Y.; Udey, M.C.; Ishikawa, O.; Motegi, S. MFG-E8 Drives Melanoma Growth by Stimulating Mesenchymal Stromal Cell-Induced Angiogenesis and M2 Polarization of Tumor-Associated Macrophages. Cancer Res. 2016, 76, 4283–4292. [Google Scholar] [CrossRef] [PubMed]
  267. Tian, Y.; Guo, Y.; Zhu, P.; Zhang, D.; Liu, S.; Tang, M.; Wang, Y.; Jin, Z.; Li, D.; Yan, D.; et al. TRIM59 loss in M2 macrophages promotes melanoma migration and invasion by upregulating MMP-9 and Madcam1. Aging 2019, 11, 8623–8641. [Google Scholar] [CrossRef] [PubMed]
  268. Han, N.; Baghdadi, M.; Ishikawa, K.; Endo, H.; Kobayashi, T.; Wada, H.; Imafuku, K.; Hata, H.; Seino, K.I. Enhanced IL-34 expression in Nivolumab-resistant metastatic melanoma. Inflamm. Regen. 2018, 38, 3. [Google Scholar] [CrossRef] [PubMed]
  269. Zhou, Q.; Fang, T.; Wei, S.; Chai, S.; Yang, H.; Tao, M.; Cao, Y. Macrophages in melanoma: A double-edged sword and targeted therapy strategies (Review). Exp. Ther. Med. 2022, 24, 640. [Google Scholar] [CrossRef] [PubMed]
  270. Luo, S.; Yang, G.; Ye, P.; Cao, N.; Chi, X.; Yang, W.H.; Yan, X. Macrophages Are a Double-Edged Sword: Molecular Crosstalk between Tumor-Associated Macrophages and Cancer Stem Cells. Biomolecules 2022, 12, 850. [Google Scholar] [CrossRef]
  271. Basak, U.; Sarkar, T.; Mukherjee, S.; Chakraborty, S.; Dutta, A.; Dutta, S.; Nayak, D.; Kaushik, S.; Das, T.; Sa, G. Tumor-associated macrophages: An effective player of the tumor microenvironment. Front. Immunol. 2023, 14, 1295257. [Google Scholar] [CrossRef] [PubMed]
  272. Ghosh, S.; Juin, S.K.; Bhattacharyya Majumdar, S.; Majumdar, S. Crucial role of glucosylceramide synthase in the regulation of stem cell-like cancer cells in B16F10 murine melanoma. Mol. Carcinog. 2021, 60, 840–858. [Google Scholar] [CrossRef]
  273. Hegde, S.; Leader, A.M.; Merad, M. MDSC: Markers, development, states, and unaddressed complexity. Immunity 2021, 54, 875–884. [Google Scholar] [CrossRef]
  274. Kamran, N.; Li, Y.; Sierra, M.; Alghamri, M.S.; Kadiyala, P.; Appelman, H.D.; Edwards, M.; Lowenstein, P.R.; Castro, M.G. Melanoma induced immunosuppression is mediated by hematopoietic dysregulation. Oncoimmunology 2018, 7, e1408750. [Google Scholar] [CrossRef] [PubMed]
  275. Berckmans, Y.; Hoffert, Y.; Vankerckhoven, A.; Dreesen, E.; Coosemans, A. Drug Repurposing for Targeting Myeloid-Derived Suppressor-Cell-Generated Immunosuppression in Ovarian Cancer: A Literature Review of Potential Candidates. Pharmaceutics 2023, 15, 1792. [Google Scholar] [CrossRef] [PubMed]
  276. He, Z.N.; Zhang, C.Y.; Zhao, Y.W.; He, S.L.; Li, Y.; Shi, B.L.; Hu, J.Q.; Qi, R.Z.; Hua, B.J. Regulation of T cells by myeloid-derived suppressor cells: Emerging immunosuppressor in lung cancer. Discov. Oncol. 2023, 14, 185. [Google Scholar] [CrossRef] [PubMed]
  277. Johnson, B. Targeting Myeloid-Derived Suppressor Cell Trafficking as a Novel Immunotherapeutic Approach in Microsatellite Stable Colorectal Cancer. Cancers 2023, 15, 5484. [Google Scholar] [CrossRef] [PubMed]
  278. Arshad, J.; Rao, A.; Repp, M.L.; Rao, R.; Wu, C.; Merchant, J.L. Myeloid-Derived Suppressor Cells: Therapeutic Target for Gastrointestinal Cancers. Int. J. Mol. Sci. 2024, 25, 2985. [Google Scholar] [CrossRef] [PubMed]
  279. Frederico, S.C.; Sharma, N.; Darling, C.; Taori, S.; Dubinsky, A.C.; Zhang, X.; Raphael, I.; Kohanbash, G. Myeloid cells as potential targets for immunotherapy in pediatric gliomas. Front. Pediatr. 2024, 12, 1346493. [Google Scholar] [CrossRef] [PubMed]
  280. Lasser, S.A.; Ozbay Kurt, F.G.; Arkhypov, I.; Utikal, J.; Umansky, V. Myeloid-derived suppressor cells in cancer and cancer therapy. Nat. Rev. Clin. Oncol. 2024, 21, 147–164. [Google Scholar] [CrossRef] [PubMed]
  281. Lu, J.; Luo, Y.; Rao, D.; Wang, T.; Lei, Z.; Chen, X.; Zhang, B.; Li, Y.; Liu, B.; Xia, L.; et al. Myeloid-derived suppressor cells in cancer: Therapeutic targets to overcome tumor immune evasion. Exp. Hematol Oncol. 2024, 13, 39. [Google Scholar] [CrossRef] [PubMed]
  282. Ding, G.; Yu, H.; Jin, J.; Qiao, X.; Ma, J.; Zhang, T.; Cheng, X. Reciprocal relationship between cancer stem cells and myeloid-derived suppressor cells: Implications for tumor progression and therapeutic strategies. Future Oncol. 2024, 20, 215–228. [Google Scholar] [CrossRef] [PubMed]
  283. Jiang, M.; Chen, J.; Zhang, W.; Zhang, R.; Ye, Y.; Liu, P.; Yu, W.; Wei, F.; Ren, X.; Yu, J. Interleukin-6 Trans-Signalin Pathway Promotes Immunosuppressive Myeloid-Derived Suppressor Cells via Suppression of Suppressor of Cytokine Signaling 3 in Breast Cancer. Front. Immunol. 2017, 8, 1840. [Google Scholar] [CrossRef] [PubMed]
  284. Ouzounova, M.; Lee, E.; Piranlioglu, R.; El Andaloussi, A.; Kolhe, R.; Demirci, M.F.; Marasco, D.; Asm, I.; Chadli, A.; Hassan, K.A.; et al. Monocytic and granulocytic myeloid derived suppressor cells differentially regulate spatiotemporal tumour plasticity during metastatic cascade. Nat. Commun. 2017, 8, 14979. [Google Scholar] [CrossRef] [PubMed]
  285. De Cicco, P.; Ercolano, G.; Ianaro, A. The New Era of Cancer Immunotherapy: Targeting Myeloid-Derived Suppressor Cells to Overcome Immune Evasion. Front. Immunol. 2020, 11, 1680. [Google Scholar] [CrossRef] [PubMed]
  286. Banuelos, A.; Zhang, A.; Berouti, H.; Baez, M.; Yilmaz, L.; Georgeos, N.; Marjon, K.D.; Miyanishi, M.; Weissman, I.L. CXCR2 inhibition in G-MDSCs enhances CD47 blockade for melanoma tumor cell clearance. Proc. Natl. Acad. Sci. USA 2024, 121, e2318534121. [Google Scholar] [CrossRef] [PubMed]
  287. Xu, Z.; Li, L.; Qian, Y.; Song, Y.; Qin, L.; Duan, Y.; Wang, M.; Li, P.; Jiang, B.; Ma, C.; et al. Upregulation of IL-6 in CUL4B-deficient myeloid-derived suppressive cells increases the aggressiveness of cancer cells. Oncogene 2019, 38, 5860–5872. [Google Scholar] [CrossRef] [PubMed]
  288. Shidal, C.; Singh, N.P.; Nagarkatti, P.; Nagarkatti, M. MicroRNA-92 Expression in CD133(+) Melanoma Stem Cells Regulates Immunosuppression in the Tumor Microenvironment via Integrin-Dependent Activation of TGFbeta. Cancer Res. 2019, 79, 3622–3635. [Google Scholar] [CrossRef] [PubMed]
  289. Kerbel, R.S. Tumor angiogenesis. N. Engl. J. Med. 2008, 358, 2039–2049. [Google Scholar] [CrossRef] [PubMed]
  290. Wu, Z.; Bian, Y.; Chu, T.; Wang, Y.; Man, S.; Song, Y.; Wang, Z. The role of angiogenesis in melanoma: Clinical treatments and future expectations. Front. Pharmacol. 2022, 13, 1028647. [Google Scholar] [CrossRef] [PubMed]
  291. Choi, H.; Moon, A. Crosstalk between cancer cells and endothelial cells: Implications for tumor progression and intervention. Arch. Pharm. Res. 2018, 41, 711–724. [Google Scholar] [CrossRef] [PubMed]
  292. Malekan, M.; Haass, N.K.; Rokni, G.R.; Gholizadeh, N.; Ebrahimzadeh, M.A.; Kazeminejad, A. VEGF/VEGFR axis and its signaling in melanoma: Current knowledge toward therapeutic targeting agents and future perspectives. Life Sci. 2024, 345, 122563. [Google Scholar] [CrossRef] [PubMed]
  293. Lopez de Andres, J.; Ruiz-Toranzo, M.; Antich, C.; Chocarro-Wrona, C.; Lopez-Ruiz, E.; Jimenez, G.; Marchal, J.A. Biofabrication of a tri-layered 3D-bioprinted CSC-based malignant melanoma model for personalized cancer treatment. Biofabrication 2023, 15, 035016. [Google Scholar] [CrossRef] [PubMed]
  294. Lee, J.; Abdeen, A.A.; Hedhli, J.; Wycislo, K.L.; Dobrucka, I.T.; Fan, T.M.; Dobrucki, L.W.; Kilian, K.A. Melanoma topology reveals a stem-like phenotype that promotes angiogenesis. Sci. Adv. 2017, 3, e1701350. [Google Scholar] [CrossRef] [PubMed]
  295. Hendrix, M.J.; Seftor, E.A.; Hess, A.R.; Seftor, R.E. Vasculogenic mimicry and tumour-cell plasticity: Lessons from melanoma. Nat. Rev. Cancer 2003, 3, 411–421. [Google Scholar] [CrossRef] [PubMed]
  296. Spiliopoulos, K.; Peschos, D.; Batistatou, A.; Ntountas, I.; Agnantis, N.; Kitsos, G. Vasculogenic mimicry: Lessons from melanocytic tumors. In Vivo 2015, 29, 309–317. [Google Scholar] [PubMed]
  297. Hendrix, M.J.; Seftor, E.A.; Seftor, R.E.; Chao, J.T.; Chien, D.S.; Chu, Y.W. Tumor cell vascular mimicry: Novel targeting opportunity in melanoma. Pharmacol. Ther. 2016, 159, 83–92. [Google Scholar] [CrossRef] [PubMed]
  298. Fernandez-Cortes, M.; Delgado-Bellido, D.; Oliver, F.J. Vasculogenic Mimicry: Become an Endothelial Cell “But Not So Much”. Front. Oncol. 2019, 9, 803. [Google Scholar] [CrossRef] [PubMed]
  299. Frank, N.Y.; Schatton, T.; Kim, S.; Zhan, Q.; Wilson, B.J.; Ma, J.; Saab, K.R.; Osherov, V.; Widlund, H.R.; Gasser, M.; et al. VEGFR-1 expressed by malignant melanoma-initiating cells is required for tumor growth. Cancer Res. 2011, 71, 1474–1485. [Google Scholar] [CrossRef] [PubMed]
  300. Orecchia, P.; Conte, R.; Balza, E.; Pietra, G.; Mingari, M.C.; Carnemolla, B. Targeting Syndecan-1, a molecule implicated in the process of vasculogenic mimicry, enhances the therapeutic efficacy of the L19-IL2 immunocytokine in human melanoma xenografts. Oncotarget 2015, 6, 37426–37442. [Google Scholar] [CrossRef] [PubMed]
  301. Shih, C.Y.; Cheng, Y.C.; Hsieh, C.; Tseng, T.; Jiang, S.; Lee, S.C. Drug-selected population in melanoma A2058 cells as melanoma stem-like cells retained angiogenic features—The potential roles of heparan-sulfate binding ANGPTL4 protein. Aging 2020, 12, 22700–22718. [Google Scholar] [CrossRef]
  302. Zimmerer, R.M.; Matthiesen, P.; Kreher, F.; Kampmann, A.; Spalthoff, S.; Jehn, P.; Bittermann, G.; Gellrich, N.C.; Tavassol, F. Putative CD133+ melanoma cancer stem cells induce initial angiogenesis in vivo. Microvasc. Res. 2016, 104, 46–54. [Google Scholar] [CrossRef]
  303. Nissen, N.I.; Karsdal, M.; Willumsen, N. Collagens and Cancer associated fibroblasts in the reactive stroma and its relation to Cancer biology. J. Exp. Clin. Cancer Res. 2019, 38, 115. [Google Scholar] [CrossRef]
  304. Glabman, R.A.; Choyke, P.L.; Sato, N. Cancer-Associated Fibroblasts: Tumorigenicity and Targeting for Cancer Therapy. Cancers 2022, 14, 3906. [Google Scholar] [CrossRef] [PubMed]
  305. Mazurkiewicz, J.; Simiczyjew, A.; Dratkiewicz, E.; Pietraszek-Gremplewicz, K.; Majkowski, M.; Kot, M.; Zietek, M.; Matkowski, R.; Nowak, D. Melanoma cells with diverse invasive potential differentially induce the activation of normal human fibroblasts. Cell Commun. Signal. 2022, 20, 63. [Google Scholar] [CrossRef] [PubMed]
  306. Zhang, H.; Yue, X.; Chen, Z.; Liu, C.; Wu, W.; Zhang, N.; Liu, Z.; Yang, L.; Jiang, Q.; Cheng, Q.; et al. Define cancer-associated fibroblasts (CAFs) in the tumor microenvironment: New opportunities in cancer immunotherapy and advances in clinical trials. Mol. Cancer 2023, 22, 159. [Google Scholar] [CrossRef]
  307. Tsang, M.; Quesnel, K.; Vincent, K.; Hutchenreuther, J.; Postovit, L.M.; Leask, A. Insights into Fibroblast Plasticity: Cellular Communication Network 2 Is Required for Activation of Cancer-Associated Fibroblasts in a Murine Model of Melanoma. Am. J. Pathol. 2020, 190, 206–221. [Google Scholar] [CrossRef]
  308. Vail, M.E.; Farnsworth, R.H.; Hii, L.; Allen, S.; Arora, S.; Anderson, R.L.; Dickins, R.A.; Orimo, A.; Wu, S.Z.; Swarbrick, A.; et al. Inhibition of EphA3 Expression in Tumour Stromal Cells Suppresses Tumour Growth and Progression. Cancers 2023, 15, 4646. [Google Scholar] [CrossRef]
  309. Penna, E.; Orso, F.; Cimino, D.; Vercellino, I.; Grassi, E.; Quaglino, E.; Turco, E.; Taverna, D. miR-214 coordinates melanoma progression by upregulating ALCAM through TFAP2 and miR-148b downmodulation. Cancer Res. 2013, 73, 4098–4111. [Google Scholar] [CrossRef]
  310. Orso, F.; Virga, F.; Dettori, D.; Dalmasso, A.; Paradzik, M.; Savino, A.; Pomatto, M.A.C.; Quirico, L.; Cucinelli, S.; Coco, M.; et al. Stroma-derived miR-214 coordinates tumor dissemination. J. Exp. Clin. Cancer Res. 2023, 42, 20. [Google Scholar] [CrossRef] [PubMed]
  311. Aramini, B.; Masciale, V.; Arienti, C.; Dominici, M.; Stella, F.; Martinelli, G.; Fabbri, F. Cancer Stem Cells (CSCs), Circulating Tumor Cells (CTCs) and Their Interplay with Cancer Associated Fibroblasts (CAFs): A New World of Targets and Treatments. Cancers 2022, 14, 2408. [Google Scholar] [CrossRef] [PubMed]
  312. Zeng, Z.; Fu, M.; Hu, Y.; Wei, Y.; Wei, X.; Luo, M. Regulation and signaling pathways in cancer stem cells: Implications for targeted therapy for cancer. Mol. Cancer 2023, 22, 172. [Google Scholar] [CrossRef] [PubMed]
  313. Kinugasa, Y.; Matsui, T.; Takakura, N. CD44 expressed on cancer-associated fibroblasts is a functional molecule supporting the stemness and drug resistance of malignant cancer cells in the tumor microenvironment. Stem Cells 2014, 32, 145–156. [Google Scholar] [CrossRef] [PubMed]
  314. Du, Y.; Shao, H.; Moller, M.; Prokupets, R.; Tse, Y.T.; Liu, Z.J. Intracellular Notch1 Signaling in Cancer-Associated Fibroblasts Dictates the Plasticity and Stemness of Melanoma Stem/Initiating Cells. Stem Cells 2019, 37, 865–875. [Google Scholar] [CrossRef] [PubMed]
  315. Trejo-Becerril, C.; Perez-Cardenas, E.; Taja-Chayeb, L.; Anker, P.; Herrera-Goepfert, R.; Medina-Velazquez, L.A.; Hidalgo-Miranda, A.; Perez-Montiel, D.; Chavez-Blanco, A.; Cruz-Velazquez, J.; et al. Cancer progression mediated by horizontal gene transfer in an in vivo model. PLoS ONE 2012, 7, e52754. [Google Scholar] [CrossRef] [PubMed]
  316. Desir, S.; Wong, P.; Turbyville, T.; Chen, D.; Shetty, M.; Clark, C.; Zhai, E.; Romin, Y.; Manova-Todorova, K.; Starr, T.K.; et al. Intercellular Transfer of Oncogenic KRAS via Tunneling Nanotubes Introduces Intracellular Mutational Heterogeneity in Colon Cancer Cells. Cancers 2019, 11, 892. [Google Scholar] [CrossRef] [PubMed]
  317. Xavier, C.P.R.; Belisario, D.C.; Rebelo, R.; Assaraf, Y.G.; Giovannetti, E.; Kopecka, J.; Vasconcelos, M.H. The role of extracellular vesicles in the transfer of drug resistance competences to cancer cells. Drug Resist. Updates 2022, 62, 100833. [Google Scholar] [CrossRef] [PubMed]
  318. Wang, R.; Zhong, H.; Wang, C.; Huang, X.; Huang, A.; Du, N.; Wang, D.; Sun, Q.; He, M. Tumor malignancy by genetic transfer between cells forming cell-in-cell structures. Cell Death Dis. 2023, 14, 195. [Google Scholar] [CrossRef] [PubMed]
  319. Tang, D.; Xu, X.; Ying, J.; Xie, T.; Cao, G. Transfer of metastatic traits via miR-200c in extracellular vesicles derived from colorectal cancer stem cells is inhibited by atractylenolide I. Clin. Transl. Med. 2020, 10, e139. [Google Scholar] [CrossRef] [PubMed]
  320. Yang, Z.; Zhao, N.; Cui, J.; Wu, H.; Xiong, J.; Peng, T. Exosomes derived from cancer stem cells of gemcitabine-resistant pancreatic cancer cells enhance drug resistance by delivering miR-210. Cell. Oncol. 2020, 43, 123–136. [Google Scholar] [CrossRef] [PubMed]
  321. Bao, Z.; Zhang, N.; Niu, W.; Mu, M.; Zhang, X.; Hu, S.; Niu, C. Exosomal miR-155-5p derived from glioma stem-like cells promotes mesenchymal transition via targeting ACOT12. Cell Death Dis. 2022, 13, 725. [Google Scholar] [CrossRef] [PubMed]
  322. Han, T.; Chen, L.; Li, K.; Hu, Q.; Zhang, Y.; You, X.; Han, L.; Chen, T.; Li, K. Significant CircRNAs in liver cancer stem cell exosomes: Mediator of malignant propagation in liver cancer? Mol. Cancer 2023, 22, 197. [Google Scholar] [CrossRef] [PubMed]
  323. Li, X.; Liu, D.; Chen, H.; Zeng, B.; Zhao, Q.; Zhang, Y.; Chen, Y.; Wang, J.; Xing, H.R. Melanoma stem cells promote metastasis via exosomal miR-1268a inactivation of autophagy. Biol. Res. 2022, 55, 29. [Google Scholar] [CrossRef] [PubMed]
  324. Liu, D.; Li, X.; Zeng, B.; Zhao, Q.; Chen, H.; Zhang, Y.; Chen, Y.; Wang, J.; Xing, H.R. Exosomal microRNA-4535 of Melanoma Stem Cells Promotes Metastasis by Inhibiting Autophagy Pathway. Stem Cell Rev. Rep. 2023, 19, 155–169. [Google Scholar] [CrossRef] [PubMed]
  325. Zhang, Y.; Chen, Y.; Shi, L.; Li, J.; Wan, W.; Li, B.; Liu, D.; Li, X.; Chen, Y.; Xiang, M.; et al. Extracellular vesicles microRNA-592 of melanoma stem cells promotes metastasis through activation of MAPK/ERK signaling pathway by targeting PTPN7 in non-stemness melanoma cells. Cell Death Discov. 2022, 8, 428. [Google Scholar] [CrossRef] [PubMed]
  326. Chen, Y.; Zhang, Y.H.; Li, J.; Shi, L.; Xie, J.C.; Han, X.; Chen, Y.T.; Xiang, M.; Li, B.W.; Xing, H.R.; et al. Novel lncRNA Gm33149 modulates metastatic heterogeneity in melanoma by regulating the miR-5623-3p/Wnt axis via exosomal transfer. Cancer Gene Ther. 2024, 31, 364–375. [Google Scholar] [CrossRef] [PubMed]
  327. Turdo, A.; Veschi, V.; Gaggianesi, M.; Chinnici, A.; Bianca, P.; Todaro, M.; Stassi, G. Meeting the Challenge of Targeting Cancer Stem Cells. Front. Cell Dev. Biol. 2019, 7, 16. [Google Scholar] [CrossRef] [PubMed]
  328. Mai, Y.; Su, J.; Yang, C.; Xia, C.; Fu, L. The strategies to cure cancer patients by eradicating cancer stem-like cells. Mol. Cancer 2023, 22, 171. [Google Scholar] [CrossRef] [PubMed]
  329. Chen, Y.C.; Gowda, K.; Amin, S.; Schell, T.D.; Sharma, A.K.; Robertson, G.P. Pharmacological agents targeting drug-tolerant persister cells in cancer. Pharmacol. Res. 2024, 203, 107163. [Google Scholar] [CrossRef] [PubMed]
  330. Saltari, A.; Dzung, A.; Quadri, M.; Tiso, N.; Facchinello, N.; Hernandez-Barranco, A.; Garcia-Silva, S.; Nogues, L.; Stoffel, C.I.; Cheng, P.F.; et al. Specific Activation of the CD271 Intracellular Domain in Combination with Chemotherapy or Targeted Therapy Inhibits Melanoma Progression. Cancer Res. 2021, 81, 6044–6057. [Google Scholar] [CrossRef]
  331. Rappa, G.; Fodstad, O.; Lorico, A. The stem cell-associated antigen CD133 (Prominin-1) is a molecular therapeutic target for metastatic melanoma. Stem Cells 2008, 26, 3008–3017. [Google Scholar] [CrossRef] [PubMed]
  332. Bostad, M.; Olsen, C.E.; Peng, Q.; Berg, K.; Hogset, A.; Selbo, P.K. Light-controlled endosomal escape of the novel CD133-targeting immunotoxin AC133-saporin by photochemical internalization—A minimally invasive cancer stem cell-targeting strategy. J. Control. Release 2015, 206, 37–48. [Google Scholar] [CrossRef] [PubMed]
  333. Schlaak, M.; Schmidt, P.; Bangard, C.; Kurschat, P.; Mauch, C.; Abken, H. Regression of metastatic melanoma in a patient by antibody targeting of cancer stem cells. Oncotarget 2012, 3, 22–30. [Google Scholar] [CrossRef] [PubMed]
  334. Winkler, J.K.; Schiller, M.; Bender, C.; Enk, A.H.; Hassel, J.C. Rituximab as a therapeutic option for patients with advanced melanoma. Cancer Immunol. Immunother. 2018, 67, 917–924. [Google Scholar] [CrossRef] [PubMed]
  335. Mukhtar, A.B.; Morgan, H.J.; Gibbs, A.; Davies, G.E.; Lovatt, C.; Patel, G.K. Targeting CD20-expressing malignant melanoma cells augments BRAF inhibitor killing. Br. J. Dermatol. 2024, 190, 729–739. [Google Scholar] [CrossRef] [PubMed]
  336. Song, H.; Su, X.; Yang, K.; Niu, F.; Li, J.; Song, J.; Chen, H.; Li, B.; Li, W.; Qian, W.; et al. CD20 Antibody-Conjugated Immunoliposomes for Targeted Chemotherapy of Melanoma Cancer Initiating Cells. J. Biomed. Nanotechnol. 2015, 11, 1927–1946. [Google Scholar] [CrossRef] [PubMed]
  337. Shigdar, S.; Ward, A.C.; De, A.; Yang, C.J.; Wei, M.; Duan, W. Clinical applications of aptamers and nucleic acid therapeutics in haematological malignancies. Br. J. Haematol. 2011, 155, 3–13. [Google Scholar] [CrossRef] [PubMed]
  338. Schmohl, J.U.; Vallera, D.A. CD133, Selectively Targeting the Root of Cancer. Toxins 2016, 8, 165. [Google Scholar] [CrossRef] [PubMed]
  339. Zeng, Y.B.; Yu, Z.C.; He, Y.N.; Zhang, T.; Du, L.B.; Dong, Y.M.; Chen, H.W.; Zhang, Y.Y.; Wang, W.Q. Salinomycin-loaded lipid-polymer nanoparticles with anti-CD20 aptamers selectively suppress human CD20+ melanoma stem cells. Acta Pharmacol. Sin. 2018, 39, 261–274. [Google Scholar] [CrossRef] [PubMed]
  340. Chen, H.; Jiang, Y.; Li, X. Adriamycin-loaded exosome with anti-CD20 aptamers selectively suppresses human CD20+ melanoma stem cells. Skin Res. Technol. 2023, 29, e13259. [Google Scholar] [CrossRef] [PubMed]
  341. Shen, H.; Shi, S.; Zhang, Z.; Gong, T.; Sun, X. Coating Solid Lipid Nanoparticles with Hyaluronic Acid Enhances Antitumor Activity against Melanoma Stem-like Cells. Theranostics 2015, 5, 755–771. [Google Scholar] [CrossRef]
  342. Li, Y.; Shi, S.; Ming, Y.; Wang, L.; Li, C.; Luo, M.; Li, Z.; Li, B.; Chen, J. Specific cancer stem cell-therapy by albumin nanoparticles functionalized with CD44-mediated targeting. J. Nanobiotechnol. 2018, 16, 99. [Google Scholar] [CrossRef]
  343. Menke-van der Houven van Oordt, C.W.; Gomez-Roca, C.; van Herpen, C.; Coveler, A.L.; Mahalingam, D.; Verheul, H.M.; van der Graaf, W.T.; Christen, R.; Ruttinger, D.; Weigand, S.; et al. First-in-human phase I clinical trial of RG7356, an anti-CD44 humanized antibody, in patients with advanced, CD44-expressing solid tumors. Oncotarget 2016, 7, 80046–80058. [Google Scholar] [CrossRef] [PubMed]
  344. Kwiatkowska-Borowczyk, E.; Czerwinska, P.; Mackiewicz, J.; Gryska, K.; Kazimierczak, U.; Tomela, K.; Przybyla, A.; Kozlowska, A.K.; Galus, L.; Kwinta, L.; et al. Whole cell melanoma vaccine genetically modified to stem cells like phenotype generates specific immune responses to ALDH1A1 and long-term survival in advanced melanoma patients. Oncoimmunology 2018, 7, e1509821. [Google Scholar] [CrossRef]
  345. Pietrobono, S.; Morandi, A.; Gagliardi, S.; Gerlini, G.; Borgognoni, L.; Chiarugi, P.; Arbiser, J.L.; Stecca, B. Down-Regulation of SOX2 Underlies the Inhibitory Effects of the Triphenylmethane Gentian Violet on Melanoma Cell Self-Renewal and Survival. J. Investig. Dermatol. 2016, 136, 2059–2069. [Google Scholar] [CrossRef]
  346. Shidal, C.; Al-Rayyan, N.; Yaddanapudi, K.; Davis, K.R. Lunasin is a novel therapeutic agent for targeting melanoma cancer stem cells. Oncotarget 2016, 7, 84128–84141. [Google Scholar] [CrossRef]
  347. Wang, Y.B.; Sun, B.; Han, B. Retinoic acid increases the anticancer effect of paclitaxel by inducing differentiation of cancer stem cells in melanoma. Pharmazie 2018, 73, 729–732. [Google Scholar] [CrossRef] [PubMed]
  348. Li, H.; He, M.; Zhao, P.; Liu, P.; Chen, W.; Xu, X. Chelerythrine Chloride Inhibits Stemness of Melanoma Cancer Stem-Like Cells (CSCs) Potentially via Inducing Reactive Oxygen Species and Causing Mitochondria Dysfunction. Comput. Math. Methods Med. 2022, 2022, 4000733. [Google Scholar] [CrossRef] [PubMed]
  349. Jia, H.; Ren, W.; Feng, Y.; Wei, T.; Guo, M.; Guo, J.; Zhao, J.; Song, X.; Wang, M.; Zhao, T.; et al. The enhanced antitumour response of pimozide combined with the IDO inhibitor LMT in melanoma. Int. J. Oncol. 2018, 53, 949–960. [Google Scholar] [CrossRef]
  350. Hu, J.; Guo, X.; Yang, L. Morin inhibits proliferation and self-renewal of CD133(+) melanoma cells by upregulating miR-216a. J. Pharmacol. Sci. 2018, 136, 114–120. [Google Scholar] [CrossRef]
  351. Du, F.Y.; Zhou, Q.F.; Sun, W.J.; Chen, G.L. Targeting cancer stem cells in drug discovery: Current state and future perspectives. World J. Stem Cells. 2019, 11, 398–420. [Google Scholar] [CrossRef] [PubMed]
  352. Feng, M.; Santhanam, R.K.; Xing, H.; Zhou, M.; Jia, H. Inhibition of gamma-secretase/Notch pathway as a potential therapy for reversing cancer drug resistance. Biochem. Pharmacol. 2024, 220, 115991. [Google Scholar] [CrossRef] [PubMed]
  353. Huynh, C.; Poliseno, L.; Segura, M.F.; Medicherla, R.; Haimovic, A.; Menendez, S.; Shang, S.; Pavlick, A.; Shao, Y.; Darvishian, F.; et al. The novel gamma secretase inhibitor RO4929097 reduces the tumor initiating potential of melanoma. PLoS ONE 2011, 6, e25264. [Google Scholar] [CrossRef]
  354. Kaushik, G.; Venugopal, A.; Ramamoorthy, P.; Standing, D.; Subramaniam, D.; Umar, S.; Jensen, R.A.; Anant, S.; Mammen, J.M. Honokiol inhibits melanoma stem cells by targeting notch signaling. Mol. Carcinog. 2015, 54, 1710–1721. [Google Scholar] [CrossRef] [PubMed]
  355. Keyghobadi, F.; Mehdipour, M.; Nekoukar, V.; Firouzi, J.; Kheimeh, A.; Nobakht Lahrood, F.; Azimian Zavareh, V.; Azimi, M.; Mohammadi, M.; Sodeifi, N.; et al. Long-Term Inhibition of Notch in A-375 Melanoma Cells Enhances Tumor Growth Through the Enhancement of AXIN1, CSNK2A3, and CEBPA2 as Intermediate Genes in Wnt and Notch Pathways. Front. Oncol. 2020, 10, 531. [Google Scholar] [CrossRef] [PubMed]
  356. Minami, H.; Ando, Y.; Ma, B.B.; Hsiang Lee, J.; Momota, H.; Fujiwara, Y.; Li, L.; Fukino, K.; Ito, K.; Tajima, T.; et al. Phase I, multicenter, open-label, dose-escalation study of sonidegib in Asian patients with advanced solid tumors. Cancer Sci. 2016, 107, 1477–1483. [Google Scholar] [CrossRef]
  357. Li, W.; Yang, H.; Li, X.; Han, L.; Xu, N.; Shi, A. Signaling pathway inhibitors target breast cancer stem cells in triple-negative breast cancer. Oncol. Rep. 2019, 41, 437–446. [Google Scholar] [CrossRef] [PubMed]
  358. Pietrobono, S.; Santini, R.; Gagliardi, S.; Dapporto, F.; Colecchia, D.; Chiariello, M.; Leone, C.; Valoti, M.; Manetti, F.; Petricci, E.; et al. Targeted inhibition of Hedgehog-GLI signaling by novel acylguanidine derivatives inhibits melanoma cell growth by inducing replication stress and mitotic catastrophe. Cell Death Dis. 2018, 9, 142. [Google Scholar] [CrossRef] [PubMed]
  359. Evyapan, G.; Luleyap, U.; Kaplan, H.M.; Kara, I.O. Ornidazole suppresses CD133+ melanoma stem cells via inhibiting hedgehog signaling pathway and inducing multiple death pathways in a mouse model. Croat. Med. J. 2022, 63, 461–474. [Google Scholar] [CrossRef] [PubMed]
  360. Jokinen, E.; Laurila, N.; Koivunen, P.; Koivunen, J.P. Combining targeted drugs to overcome and prevent resistance of solid cancers with some stem-like cell features. Oncotarget 2014, 5, 9295–9307. [Google Scholar] [CrossRef] [PubMed]
  361. Liang, G.; Liu, M.; Wang, Q.; Shen, Y.; Mei, H.; Li, D.; Liu, W. Itraconazole exerts its anti-melanoma effect by suppressing Hedgehog, Wnt, and PI3K/mTOR signaling pathways. Oncotarget 2017, 8, 28510–28525. [Google Scholar] [CrossRef] [PubMed]
  362. Tolcher, A.W.; Kurzrock, R.; Valero, V.; Gonzalez, R.; Heist, R.S.; Tan, A.R.; Means-Powell, J.; Werner, T.L.; Becerra, C.; Wang, C.; et al. Phase I dose-escalation trial of the oral AKT inhibitor uprosertib in combination with the oral MEK1/MEK2 inhibitor trametinib in patients with solid tumors. Cancer Chemother. Pharmacol. 2020, 85, 673–683. [Google Scholar] [CrossRef] [PubMed]
  363. Karami Fath, M.; Ebrahimi, M.; Nourbakhsh, E.; Zia Hazara, A.; Mirzaei, A.; Shafieyari, S.; Salehi, A.; Hoseinzadeh, M.; Payandeh, Z.; Barati, G. PI3K/Akt/mTOR signaling pathway in cancer stem cells. Pathol. Res. Pract. 2022, 237, 154010. [Google Scholar] [CrossRef] [PubMed]
  364. Guo, X.; Huang, S.; Zhang, Y.; Wang, H.; Li, L.; Ran, J.; Chen, D.; Li, X.; Li, J. Evodiamine inhibits growth of vemurafenib drug-resistant melanoma via suppressing IRS4/PI3K/AKT signaling pathway. J. Nat. Med. 2024, 78, 342–354. [Google Scholar] [CrossRef] [PubMed]
  365. Xu, B.S.; Chen, H.Y.; Que, Y.; Xiao, W.; Zeng, M.S.; Zhang, X. ALK(ATI) interacts with c-Myc and promotes cancer stem cell-like properties in sarcoma. Oncogene 2020, 39, 151–163. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic representation of stemness biomarkers and intracellular pathways in melanoma CSCs. ABC, ATP-binding cassette transporter.
Figure 1. Schematic representation of stemness biomarkers and intracellular pathways in melanoma CSCs. ABC, ATP-binding cassette transporter.
Cancers 16 02861 g001
Figure 2. Schematic overview of the cell-to-cell bidirectional communication between melanoma CSCs and immune cells within the TME. Melanoma CSCs decrease immune cell recruitment while escaping their cytotoxicity (T cells, NK cells), promote the conversion of TANs and TAMs cells into a protumoral phenotype (N2 and M2, respectively) and recruit MDSCs immunosuppressive cells. Conversely, immune cells promote the induction of stemness traits and cell expansion in CSCs. TME, tumor microenvironment; MDSCs, myeloid-derived suppressor cells.
Figure 2. Schematic overview of the cell-to-cell bidirectional communication between melanoma CSCs and immune cells within the TME. Melanoma CSCs decrease immune cell recruitment while escaping their cytotoxicity (T cells, NK cells), promote the conversion of TANs and TAMs cells into a protumoral phenotype (N2 and M2, respectively) and recruit MDSCs immunosuppressive cells. Conversely, immune cells promote the induction of stemness traits and cell expansion in CSCs. TME, tumor microenvironment; MDSCs, myeloid-derived suppressor cells.
Cancers 16 02861 g002
Figure 3. Schematic overview of the cell-to-cell bidirectional communication between melanoma CSCs and stromal cells (endothelial cells, CAFs) within the TME, and between melanoma CSCs and non-stem cancer cells within the tumor bulk. Melanoma CSCs trigger the activation of stromal fibroblasts into CAFs and promote neoangiogenesis by inducing endothelial cell proliferation and new blood vessel formation and acquiring an endothelial phenotype (vasculogenic mimicry, VM). Conversely, CAFs and endothelial cells promote the expression and maintenance of stemness traits in melanoma CSCs. Melanoma CSCs endow non-stem cancer cells in the tumor bulk population with high metastatic properties. CAFs, cancer-associated fibroblasts; TME, tumor microenvironment.
Figure 3. Schematic overview of the cell-to-cell bidirectional communication between melanoma CSCs and stromal cells (endothelial cells, CAFs) within the TME, and between melanoma CSCs and non-stem cancer cells within the tumor bulk. Melanoma CSCs trigger the activation of stromal fibroblasts into CAFs and promote neoangiogenesis by inducing endothelial cell proliferation and new blood vessel formation and acquiring an endothelial phenotype (vasculogenic mimicry, VM). Conversely, CAFs and endothelial cells promote the expression and maintenance of stemness traits in melanoma CSCs. Melanoma CSCs endow non-stem cancer cells in the tumor bulk population with high metastatic properties. CAFs, cancer-associated fibroblasts; TME, tumor microenvironment.
Cancers 16 02861 g003
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Limonta, P.; Chiaramonte, R.; Casati, L. Unveiling the Dynamic Interplay between Cancer Stem Cells and the Tumor Microenvironment in Melanoma: Implications for Novel Therapeutic Strategies. Cancers 2024, 16, 2861. https://doi.org/10.3390/cancers16162861

AMA Style

Limonta P, Chiaramonte R, Casati L. Unveiling the Dynamic Interplay between Cancer Stem Cells and the Tumor Microenvironment in Melanoma: Implications for Novel Therapeutic Strategies. Cancers. 2024; 16(16):2861. https://doi.org/10.3390/cancers16162861

Chicago/Turabian Style

Limonta, Patrizia, Raffaella Chiaramonte, and Lavinia Casati. 2024. "Unveiling the Dynamic Interplay between Cancer Stem Cells and the Tumor Microenvironment in Melanoma: Implications for Novel Therapeutic Strategies" Cancers 16, no. 16: 2861. https://doi.org/10.3390/cancers16162861

APA Style

Limonta, P., Chiaramonte, R., & Casati, L. (2024). Unveiling the Dynamic Interplay between Cancer Stem Cells and the Tumor Microenvironment in Melanoma: Implications for Novel Therapeutic Strategies. Cancers, 16(16), 2861. https://doi.org/10.3390/cancers16162861

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop