Next Article in Journal
Plasma and Serum LC-MS Lipidomic Fingerprints of Bipolar Disorder and Schizophrenia
Previous Article in Journal
Circadian-Tuned Peptide Drug/Gene Co-Delivery Nanocomplexes to Enhance Glioblastoma Targeting and Transfection
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Fibroblasts in Melanoma Development: From Tumor Microenvironment Remodeling to Pre-Metastatic Niche Formation

1
Department of Psychology and Health Sciences, Telematic University Pegaso, 80143 Naples, Italy
2
Department of Medicine and Health Sciences “Vincenzo Tiberio”, University of Molise, 86100 Campobasso, Italy
3
Department of Mental and Physical Health and Preventive Medicine, Section of Human Anatomy, University of Campania “Luigi Vanvitelli”, 80131 Naples, Italy
4
Departmental Faculty of Medicine and Surgery, UniCamillus-Saint Camillus International University of Health Sciences, 00131 Rome, Italy
5
Department of Medical, Human Movement and Well-Being Sciences, University of Naples Parthenope, 80133 Naples, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2025, 26(13), 6132; https://doi.org/10.3390/ijms26136132
Submission received: 27 May 2025 / Revised: 19 June 2025 / Accepted: 24 June 2025 / Published: 26 June 2025

Abstract

Melanoma is the most aggressive form of skin cancer, and despite significant therapeutic advances over the past decade, a substantial number of patients still progress to a fatal outcome. The initiation and progression of melanoma are strongly influenced by interactions between melanoma cells and other components of the tumor microenvironment (TME). In this review, we focus on the interplay between fibroblasts resident in the tumor microenvironment and tumor cells. In particular, we examine the molecular mechanisms through which melanoma cells induce the transformation of resident fibroblasts into their active form, known as cancer-associated fibroblasts (CAFs). We also explore the role of CAFs in shaping the melanoma microenvironment (MME) and in organizing the pre-metastatic niche, a specialized microenvironment that forms in distant organs or tissues to support the survival and expansion of metastatic melanoma cells. Finally, we discuss emerging therapeutic strategies aimed at disrupting the interactions between CAFs, melanoma cells, and other components of the tumor microenvironment to improve treatment outcomes.

Graphical Abstract

1. Introduction

It is widely recognized that melanomagenesis can result from alterations in the internal mechanisms of cancer cells, such as mutations in specific genes (BRAF, CDKN2A, NRAS, and TP53) or epigenetic alterations, including the deregulation of non-coding RNAs such as microRNAs (miRNAs) and long non-coding RNAs (lncRNAs) [1,2,3].
The development of targeted therapies for melanoma has arisen from the identification of these genetic mutations, and such therapies have shown remarkable clinical success [4,5]. In modern clinical settings, therapeutic approaches frequently involve targeting the mitogen-activated protein kinase (MAPK) signaling cascade. Drugs designed to inhibit BRAF (such as encorafenib, dabrafenib, and vemurafenib) and MEK (including trametinib, binimetinib, and cobimetinib) are widely utilized, either as monotherapy or in combination regimens [4,5].
However, many patients initially respond to these therapies, but after approximately six to eight months, develop resistance mechanisms, limiting the long-term efficacy of these treatments [6]. Over the past ten years, the treatment of melanoma has been revolutionized by the introduction of immunotherapies, particularly immune checkpoint blockade. Since their initial FDA approval in 2011, checkpoint inhibitors have become central to melanoma treatment strategies. These monoclonal antibodies inhibit key immune-regulatory molecules, including PD-1 (e.g., pembrolizumab, nivolumab), PD-L1 (e.g., atezolizumab), CTLA-4 (e.g., ipilimumab), and LAG-3 (e.g., relatlimab). Used either as monotherapy or in combination, these agents have demonstrated remarkable success in clinical trials involving advanced melanoma cases [7,8]. Although melanoma therapy has been greatly improved by the use of immune checkpoint inhibitors, not all patients respond to them, showing primary or secondary resistance [9,10].
For this reason, there is a need to identify new target molecules and molecular mechanisms that regulate tumor development, progression, and drug resistance, enabling more effective and long-lasting treatment strategies [11,12,13,14].
In this context, recent studies have highlighted complex interactions between melanoma cells and the TME, showing that they play a very important role in supporting disease progression [15]. The TME is composed of cancer cells, immune cells, stromal cells, and the extracellular matrix (ECM), all of which interact through soluble molecules released by both cancerous and non-cancerous cells [16]. The genesis of the TME is driven by stromal alterations induced by tumor development and is shaped by the dynamic balance between pro-tumorigenic and antitumor cells. These cellular populations interact to establish the specific characteristics of the TME [17,18,19].
Cells within the melanoma microenvironment (MME) produce molecules that, in a paracrine manner, affect tumor growth by degrading the extracellular matrix, enhancing melanoma angiogenesis, and increasing tumor cell invasiveness [20].
In the MME, melanoma cells lose their responsiveness to keratinocyte regulation because E-cadherin expression is replaced by N-cadherin. The extracellular domain of N-cadherin facilitates interactions with other cells that express N-cadherin, allowing melanoma cells to communicate and bind to fibroblasts [21].
Fibroblasts within the MME become activated through interactions with melanoma cells and differentiate into CAFs. These CAFs, in turn, promote tumor cell proliferation, enhance invasiveness and migratory capacity, and influence the patient’s response to therapy and prognosis [20,22,23,24,25].
In this review, we examine the role of reciprocal interactions between melanoma cells and fibroblasts in regulating tumor growth and progression. Furthermore, we discuss the potential of targeting these interactions as a promising therapeutic strategy.

2. Crosstalk Between Fibroblasts and Melanoma Cells in the Melanoma Microenvironment (MME)

In the MME, the reprogramming of fibroblasts to CAFs by tumor cells plays a pivotal role in tumor promotion and progression, influencing several aspects of melanoma biology. CAFs acquire a specific pro-tumorigenic phenotype, characterized by enhanced contractility, ECM production, and secretion of cytokines and chemokines [26].
Among these, CXC motif chemokine ligand 12 (CXCL12), also known as stromal-derived factor-1) is notably enriched. CXCL12 binds to chemokine receptor type 4 (CXCR4 receptor) expressed on melanoma cells, promoting chemotaxis, survival, and homing to pre-metastatic niches such as the lungs and lymph nodes [27,28,29,30]. The functional role of the CXCL12/CXCR4 axis has been validated in preclinical models, where CXCR4 inhibition reduced both invasion and metastatic dissemination [31]. Additionally, CXCL12 mediates the recruitment of macrophages and endothelial progenitor cells, contributing to immune evasion and neovascularization [32,33,34,35] (Figure 1).
Beyond the production of chemokines, CAFs represent a crucial source of transforming growth factor beta (TGF-β), a cytokine that profoundly impacts melanoma progression. Within the primary tumor microenvironment, TGF-β acts on melanoma cells to induce changes similar to epithelial-to-mesenchymal transition (EMT), characterized by reduced cell adhesion, increased motility, and elevated expression of matrix metalloproteinases (MMPs). These alterations facilitate the remodeling of the extracellular matrix and support local tumor invasion [36].
Furthermore, TGF-β plays a significant role in modulating immune responses by promoting regulatory T cell accumulation and hindering the infiltration of cytotoxic T lymphocytes into the tumor core, thereby establishing a protective physical and immunological barrier around the tumor [37,38,39] (Figure 1).
Moreover, activated CAFs release hepatocyte growth factor (HGF), vascular endothelial growth factor (VEGF), and fibroblast growth factor 2 (FGF2), which promote angiogenesis and activate survival pathways in melanoma cells [40,41,42,43]. In BRAF-mutant melanomas, CAF-derived HGF has been implicated in resistance to BRAF inhibitors by reactivating the MAPK signaling pathway, thereby allowing melanoma cells to evade therapeutic pressure [24,44,45] (Figure 1).
The reciprocal communication between melanoma cells and fibroblasts is not only mediated by soluble factors but also by extracellular vesicles (EVs), particularly exosomes that deliver regulatory microRNAs (miRNAs), proteins, and metabolites [46]. These interactions contribute to the remodeling of the ECM, induce immune evasion and angiogenesis, and improve pre-metastatic niche formation with a sophisticated mechanism of stromal reprogramming [47].
Dror et al. demonstrated that miR-211 enclosed in melanosomes is transferred to dermal fibroblasts, where it downregulates the insulin-like growth factor 2 receptor (IGF2R), leading to the activation of the MAPK pathway [48]. This, in turn, promotes fibroblast proliferation, metabolic reprogramming, and the secretion of cytokines such as IL-1β, IL-6, and IL-8, thereby creating a pro-inflammatory microenvironment that precedes a visible tumor expansion [48,49].

3. Cancer-Associated Fibroblasts (CAFs) Activate Extracellular Matrix (ECM) Remodeling

CAFs are responsible for the extensive production and remodeling of the ECM (Figure 1). Specifically, during melanoma progression, activated fibroblasts secrete abundant structural ECM proteins, such as collagens (especially type I collagen), fibronectin, laminin, and various proteoglycans accumulating around the tumors and leading to the formation of a denser extracellular matrix, rich in collagen fibers or, in some cases, a high proteoglycan content stroma [50]. The deposition of collagen and fibronectin driven by CAFs provides a scaffold that not only enhances melanoma cell adhesion and migration but also increases tissue stiffness, which can promote the invasive behavior of tumor cells by activating a specific mechano-signaling pathway [50]. By building up a permissive fibrotic scaffold and carving out routes for dissemination, CAFs fundamentally reshape the melanoma microenvironment to support tumor invasion and metastasis. Melanoma cells are indeed highly sensitive to ECM composition, stiffness, and alignment, as they directly interact with the ECM in the tumor’s microenvironment via cell surface receptors, secreted factors, or enzymes [51,52,53]. Furthermore, PDGF released by melanoma cells has been shown to stimulate nearby fibroblasts to produce collagen, fibronectin, and laminin, illustrating how the crosstalk between tumor cells and the stromal microenvironment can prompt the remodeling of the ECM [54,55]. As a matter of fact, clinical melanoma specimens often exhibit activated fibroblasts co-expressing ECM proteins (e.g., collagen, fibronectin) and myofibroblastic markers (α-SMA), highlighting the direct role of CAFs in reshaping the tumor stromal microenvironment [56].
Additionally, to facilitate melanoma invasion, CAFs actively remodel and degrade the ECM, secreting high levels of MMPs that digest structural barriers and support tumor growth [50]. Secretion of MMPs by CAFs not only destroys physical barriers but also disengages pro-tumoral signals embedded in the ECM, amplifying melanoma progression [57]. Specific proteases like MMP-2, MMP-9, MMP-13, and MT1-MMP are often upregulated in the melanoma stromal microenvironment, where they degrade type I collagen fibers and basement membrane components like collagen IV, elastin, and laminin, operating a targeted ECM proteolysis that smooths the way for melanoma cells to invade surrounding tissue [58,59]. Moreover, MMPs activity releases bioactive molecules that are physiologically sequestered within the ECM, such as growth factors and angiogenic factors [60]. For instance, MMP-mediated collagen degradation can free VEGF and FGF bound to the matrix, thus accelerating new blood vessel formation, and, consequently, tumor invasion [61]. High MMP expression in melanoma lesions correlates with advanced disease and poorer patient survival [62], highlighting the pathological importance of ECM remodeling driven by CAFs.

4. Fibroblasts in the Pre-Metastatic Niche

The early stages of the metastatic process are characterized by a hostile microenvironment for the engraftment of tumor cells in distant organs. Fibroblasts in distant organs (e.g., in the lungs, liver, or bone) can be reprogrammed by circulating melanoma cells to create a favorable environment for metastatic growth. Indeed, the characterization of melanoma-derived exosomes has demonstrated their function in promoting metastatic progression [63] by reprogramming stromal cells and promoting the formation of an inflammatory metastatic niche that promotes tumor development and suppresses adaptive immunity [64].
Distinctly, at distant metastatic sites, melanoma-derived TGF-β stimulates resident fibroblasts to produce extracellular matrix components such as collagen and tenascin-C, which contribute to the formation of a pre-metastatic niche that supports subsequent tumor colonization [50].
In turn, the reprogramming of stromal cells induces a modification of the microenvironment that begins to favor the colonization of the organ and metastatic growth [65,66]. The fibroblasts present in secondary sites remodel the extracellular matrix in the pre-metastatic niche, acting as a true guide for the tumor cells; they create paths through the action of forces and proteases [67]. Force-induced matrix remodeling is activated by α3 and α5 integrins and by activation of the Rho pathway in fibroblasts [67]. Furthermore, infiltrating tumor cells, in turn, stimulate fibroblasts of secondary target organs to express periostin, a component of the stromal extracellular matrix that promotes colonization by tumor cells [68]. Moreover, fibroblasts contribute to the formation of new blood vessels, a crucial process for metastatic growth. The VEGF and FGF secreted by fibroblasts can induce vascularization at the metastatic site, providing nutrients and oxygen to the growing melanoma metastases [69,70,71]. EVs secreted by melanoma cells contain several miRNAs, including miR-155. By regulating the suppressor of cytokine signaling type 1 (SOCS1), miR-155 activates the Janus kinase 2/STAT3 signaling pathway, promoting the expression of pro-angiogenic factors (VEGFα, FGF2, and MMPs) in CAFs [72].
Similar to what occurs in the primary tumor microenvironment, fibroblasts located at metastatic sites also contribute to shaping an immunosuppressive milieu that facilitates melanoma’s immune evasion. In these pre-metastatic niches, fibroblasts may secrete TGF-β, which suppresses the activation of immune effector cells such as T lymphocytes and natural killer (NK) cells, thereby enabling melanoma cells to evade immune-mediated destruction [73].

5. Therapeutic Implications

Given the significant role of fibroblasts in the development of the primary tumor and the metastatic niche in melanoma progression, targeting CAFs or ECM components and secreted products may represent a promising strategy to disrupt the tumor microenvironment and enhance therapy [72]. Table 1 summarizes the potential therapeutic targets and the studies conducted to investigate them.

5.1. Inhibition of CXCR4

Further, in melanoma cells, CXCR4 is expressed on fibroblasts and CD133+ melanoma subpopulations. This signaling axis regulates tumor angiogenesis and immune cell trafficking within the TME [74,75,76], with evidence showing CXCL12’s chemotactic action on CD133+ cells [77]. The inhibition of CXCR4 has also been linked to suppressed melanoma growth and enhanced CD8+ T cell infiltration in in vivo models [27,78,79]. In a phase Ib clinical trial involving patients with advanced metastatic melanoma, Andtbacka et al. reported that treatment with the CXCR4 inhibitor mavorixafor for three weeks enhanced the recruitment of CD8+ T cells into melanoma lesions. Immunohistochemical analysis of patient biopsy samples revealed a marked increase in CD8+ T cells at the interface between tumor and adjacent normal tissue, compared to controls [80]. Collectively, these findings indicate that mavorixafor facilitates immune cell infiltration into melanoma metastatic sites. The enhanced infiltration of CD8+ T cells into the TME has been associated with reduced metastatic potential and improved therapeutic outcomes in melanoma [81,82,83,84].

5.2. Inhibition of Fibroblast Activation Protein (FAP)

Fibroblast activation protein (FAP) is a transmembrane serine protease classified within the dipeptidyl peptidase (DPP) family. It is predominantly expressed by CAFs and is detected on stromal fibroblasts of all melanocytic tumors, including benign, premalignant, and malignant lesions [16,85]. The marked upregulation of FAP is widely regarded as a defining marker of CAFs and has been associated with poor prognosis across multiple cancer types [86,87,88]. Capaccione et al. evaluated a novel therapeutic approach for melanoma, based on the targeting of fibro-blast activation protein (FAP) with the radionuclide 177Lu-FAPI-04. To this end, B16F10 murine models of melanoma were treated with 177Lu-FAPI-04 alone or in combination with checkpoint inhibitor immunotherapy (anti-murine CTLA-4 and anti-murine PD-1). Notably, both monotherapy with 177Lu-FAPI-04 and combined radioligand-immunotherapy induced tumor regression, with the combined approach showing increased tumor cell apoptosis and reduced proliferative indices. Flow cytometric analysis revealed distinct, tumor-specific changes in tumor-associated macrophages in the combination therapy group, differing from the effects seen with either treatment alone. These findings highlight the potential of 177Lu-FAPI-04 as an effective therapeutic strategy for melanoma, working at least in part through the induction of apoptosis and modulation of the tumor immune microenvironment. Further translational studies are warranted to evaluate the clinical potential of this combination regimen. Collectively, these findings support the therapeutic utility of 177Lu-FAPI-04 as a monotherapy and its potential to enhance the efficacy of immunotherapy through complementary mechanisms involving direct cytotoxicity and remodeling of the tumor immune microenvironment [89]. In a phase Ib clinical study, Munoz-Cosuelo et al. evaluated the combination of fibroblast activation protein interleukin-2 variant (FAP-IL2v), a novel immunocytokine engineered to address the limitations of wild-type IL-2, with the checkpoint inhibitor pembrolizumab in patients with advanced or metastatic melanoma, including both checkpoint inhibitor (CPI)-naïve and CPI-experienced individuals. The combination therapy demonstrated limited antitumor efficacy in patients who had previously progressed on CPI treatment, indicating that FAP-IL2v alone is insufficient to overcome resistance or lack of responsiveness to checkpoint inhibition [90]. Talabostat is an orally bioavailable inhibitor of dipeptidyl peptidases with immunostimulatory properties that demonstrated antitumor activity in a mouse model of melanoma [91]. Nevertheless, results from the Eager et al. phase II clinical trial showed that the combination of talabostat and cisplatin in patients with metastatic melanoma did not improve anticancer activity in comparison to standard of care therapies [92].

5.3. Inhibition of TGF-β

Beyond its well-known immunosuppressive and pro-invasive functions, TGF-β exhibits additional context-dependent roles during melanoma progression. While initially acting as a growth inhibitor in normal melanocytes and early-stage lesions, its signaling undergoes a functional switch in malignancy, becoming a critical driver of tumor progression [54]. In this pathological setting, melanoma cells exploit TGF-β signaling to suppress the production of key immunomodulatory cytokines, such as TNF-α, VEPH1, and INF-γ, and to alter regulatory pathways including Notch1, IL-6, and ERK/MAPK, ultimately promoting an immunologically inert microenvironment [93,94,95].
Moreover, excessive TGF-β expression hampers interleukin-12-mediated stimulation of CD4+, CD8+ T cells, and NK cells, impairing both lymphocyte proliferation and their cytotoxic activity, thereby contributing to immune escape [96,97].
Due to this dual role in promoting tumor progression and subverting immune responses, therapeutic strategies aimed at neutralizing TGF-β signaling have gained significant attention. Preclinical studies in melanoma models, such as those employing the TGF-β receptor inhibitor SB-505124 in combination with IL-12-based immunotherapy, have shown promising results in enhancing antitumor efficacy and prolonging survival [96,98]. Among the emerging agents, Fresolimumab (GC1008), a monoclonal antibody targeting all isoforms of TGF-β, represents a potential candidate for clinical application. In a phase I clinical trial, Morris et al. assessed the safety and antitumor activity of GC1008 in patients with advanced malignant melanoma. The study found that repeated dosing was generally well tolerated and showed preliminary signs of antitumor efficacy. These findings support continued investigation of GC1008 as both a monotherapy and in combination with other anticancer agents [99].
Table 1. Potential therapeutic targets in melanoma microenvironment-directed therapy.
Table 1. Potential therapeutic targets in melanoma microenvironment-directed therapy.
TargetsInhibitory MoleculesDrug ClassPhase of Clinical StudiesClinical TrialCurrent Status of the Clinical TrialRefs.
CXCR4CXCR4 antagonist X4-136Small moleculesPreclinical [73]
CXCR4 antagonist MavorixaforClinical phase INCT02823405Not yet completed, completion in 2029[80]
FAP177Lu-FAPI-04 (radionuclide)Fibroblast Activation Protein (FAP) inhibitorsPreclinical [85]
Simlukafusp alfa, RO6874281 (FAP-IL2v)Engineered immunocytokineClinical phase INCT02627274Completed. Tolerability acceptable; common adverse events included fatigue, asthenia, and drug-induced liver injury[90]
TalabostatSmall moleculesClinical phase IINCT00083239Completed, limited efficacy as monotherapy, highlighting the need for further studies, potentially in combination with other agents[92]
TGF-βSB-505124 + IL-12Small molecule+ encoding adenoviral vectorPreclinical [96,98]
FresolimumabHuman monoclonal antibodiesClinical phase INCT00356460Completed. Well-tolerated with manageable adverse events; limited efficacy data but supports further TGF-β research[99]
MMPsBatimastatAngiogenesis inhibitorsPreclinical [100,101]
MarimastatSmall moleculesClinical phase IINCT00004248Completed, limited clinical benefit and significant side effects, leading to discontinuation of the trial for melanoma[102]

5.4. Inhibition of MMPs

Matrix metalloproteinases (MMPs) have been extensively studied for their role in melanoma progression, particularly in facilitating extracellular matrix degradation, tumor invasion, and metastasis. This has led to the development of MMP inhibitors (MMPIs) such as Batimastat and Marimastat. Batimastat, a broad-spectrum MMPI, demonstrated efficacy in preclinical models by inhibiting tumor growth and angiogenesis. However, its clinical application was limited due to poor solubility and low oral bioavailability, necessitating intraperitoneal administration, which posed practical challenges and led to adverse effects like peritonitis and liver dysfunction. [100,101].
Marimastat, developed as an orally bioavailable analog, showed promise in early-phase clinical trials. Nevertheless, its non-selective inhibition of MMPs and related enzymes such as ADAMs resulted in significant side effects, including musculoskeletal pain, inflammation, and fibrosis. These adverse events, coupled with a lack of substantial survival benefits, led to the discontinuation of Marimastat’s development. However, in a phase II trial involving patients with metastatic melanoma, Marimastat demonstrated only modest therapeutic efficacy [100,101,102].
The challenges faced by MMPIs underscore the complexity of targeting proteolytic enzymes in cancer therapy. MMPs not only facilitate tumor progression but also play roles in normal physiological processes, such as tissue remodeling and the release of anti-angiogenic factors. Inhibiting these enzymes indiscriminately can disrupt these processes, leading to unintended consequences [101].
Beyond MMPs, other proteolytic enzymes like cathepsins and urokinase-type plasminogen activator (uPA) have been implicated in melanoma progression. Cathepsins B, K, and L, as well as uPA and its receptor uPAR, are upregulated in melanoma and stromal cells, contributing to tumor invasiveness. While these enzymes present potential therapeutic targets, none have yet advanced to clinical trials specifically for melanoma [103,104,105].
In conclusion, while proteolytic enzymes remain attractive targets in melanoma therapy, the development of effective and safe inhibitors requires a nuanced understanding of their diverse roles in both tumor biology and normal physiology.

5.5. Additional TME-Related Targets in Melanoma Treatment

In addition to CAFs, ECM components, and previously discussed targets such as CXCR4, FAP, TGF-β, and MMPs, several other TME-associated molecules and pathways have emerged as relevant to melanoma progression and therapy resistance. These include the vitamin D receptor (VDR), platelet-derived growth factor receptor (PDGFR), fibroblast growth factor receptor (FGFR), angiotensin receptor signaling, JAK-STAT signaling, the Hedgehog (Hh) pathway, and hyaluronic acid.
The VDR, expressed in both melanoma cells and stromal fibroblasts, has been implicated in regulating cell proliferation, differentiation, and immune modulation. Its activation may suppress melanoma progression and enhance immune surveillance [106,107].
Angiotensin receptor signaling, particularly via the angiotensin II type 1 receptor (AT1R), has been linked to immunosuppression and fibrosis within the TME. Targeting this axis may improve vascular normalization and drug delivery [108,109,110,111]. Similarly, the JAK-STAT pathway plays a role in stromal inflammation and immune evasion, and its pharmacological blockade may restore antitumor immunity [112].
The Hedgehog pathway, involved in tissue patterning and fibroblast behavior, is aberrantly activated in various tumors and supports CAF expansion and ECM production [113]. Finally, hyaluronic acid, a major ECM component, influences melanoma cell motility and immune exclusion. Strategies to degrade or block its accumulation are under investigation to improve TME accessibility and therapy penetration [114,115].
Together, these stromal-related targets represent promising avenues for combinatorial strategies aimed at disrupting TME-mediated resistance mechanisms in melanoma.

6. Conclusions and Discussion

This review explores the role of interactions between fibroblasts and melanoma cells within the tumor microenvironment (TME) in driving tumor initiation, progression, and the formation of the pre-metastatic niche. Previous studies have demonstrated that genetic and epigenetic alterations in melanoma cells are key oncogenic drivers, enabling the development of targeted therapies that are still used in the treatment of melanoma [4,5]. However, responses to these therapies are often not durable, highlighting the need for continued research to achieve long-lasting treatment outcomes.
In recent years, growing evidence has indicated that the interaction between tumor cells, the stroma, and the extracellular matrix is crucial for the genesis and progression of melanoma. A dynamic and not yet fully understood balance exists within the TME, where stromal cells, tumor cells, and the extracellular matrix influence one another through complex feedback loops [14,15,16,17].
Within this environment, fibroblasts can transition from an inactive to an active state, becoming CAFs. These CAFs, in turn, stimulate tumor cells and contribute to melanoma progression [20,22,23,24,25]. Biomedical research is increasingly focusing on the TME to identify potential therapies that target the multiple interactions occurring within it.
The ultimate goal of this review is to highlight how the complex interactions between fibroblasts and tumor cells can play a crucial role in clinical outcomes. A deeper understanding of the alterations in tumor cells and their crosstalk with other components of the TME, particularly fibroblasts, may offer new targets for therapeutic intervention. CAFs have emerged as promising targets for anticancer therapies; however, their clinical application is challenged by several intrinsic complexities. A primary difficulty arises from their heterogeneous cellular origins, which vary depending on the tissue and tumor microenvironment. CAFs exhibit substantial phenotypic and functional heterogeneity within the TME, where they originate from various sources, including tissue-resident fibroblasts, mesenchymal stem cells, endothelial cells, and pericytes. Advances in single-cell RNA sequencing and proteomic profiling have refined the classification of CAF subsets, revealing transcriptionally distinct populations that vary according to tumor context and evolve dynamically during disease progression [116].
Among the primary CAF subtypes are myofibroblastic CAFs (myCAFs), which are implicated in extracellular matrix (ECM) remodeling and collagen deposition, processes that contribute to tissue stiffness, tumor growth, and metastatic dissemination; inflammatory or immune-modulatory CAFs (iCAFs), which influence immune cell recruitment, suppress antitumor immunity, and support tumor cell survival and proliferation; and antigen-presenting CAFs (apCAFs), a less-characterized group thought to engage with T lymphocytes, potentially shaping local immune responses [116] (Table 2).
These diverse subsets underscore the dualistic and sometimes opposing roles of CAFs in modulating tumor architecture and immune dynamics. Their complexity necessitates a nuanced understanding when designing targeted therapies, as different CAF populations may either support or hinder therapeutic efficacy [116].
Despite advances in identifying CAF-associated markers, no single biomarker demonstrates absolute specificity. Currently, CAFs are defined by the lack of epithelial, endothelial, and hematopoietic markers, while expressing mesenchymal markers such as vimentin, alpha-smooth muscle actin (α-SMA), fibroblast activation protein (FAP), and platelet-derived growth factor receptor alpha (PDGFRα), typically without oncogenic mutations [117,118]. Due to this complexity, accurate identification of CAFs necessitates the combined use of multiple markers adapted to the specific tumor context.
However, molecular data alone are not sufficient; interpreting the functional roles of each CAF subset in relation to tumor cells, immune infiltrates, and other stromal components is equally important. The classification of CAF subtypes emphasizes not only their phenotypic variability but also their ability to adapt functionally to changes within the tumor microenvironment. In particular, the subset known as inflammatory CAFs (iCAFs) has been shown to foster an immunosuppressive environment through the expression of immune checkpoint ligands, including PD-L1 and PD-L2, which inhibit T cell activity without requiring direct cell contact. Additionally, factors secreted by CAFs, such as CXCL2, CXCL5, or miRNAs delivered via extracellular vesicles, can stimulate PD-L1 expression in tumor cells, further limiting the effectiveness of immune responses. Notably, certain CAF populations have also been reported to display antigen-presenting features and to trigger antigen-specific cytotoxic T cell death, highlighting the extent to which CAF heterogeneity impacts immune regulation and resistance to immune checkpoint therapies [119].
Clarifying the relationships among CAF subsets, whether they function as independent lineages, occupy overlapping states, or follow a hierarchical structure, is crucial. Mapping these interactions and spatial arrangements within the tumor microenvironment could enable the development of more precise therapeutic strategies aimed at targeting only the CAF populations that actively contribute to tumor progression, while preserving or even enhancing those that may exert regulatory or antitumoral effects.

Author Contributions

Conceptualization, M.L.M.; methodology, I.B., R.P. and M.L.M.; writing—original draft preparation, I.B., R.P., A.M., E.D.G. and M.L.M.; writing—review and editing, I.B., R.P., A.M., E.D.G., S.B., C.S. and M.L.M.; supervision M.L.M. Critical revision of the work and final version approval S.P. and M.L.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

No new data were created or analyzed in this study.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

CAFcancer-associated fibroblast
TMEtumor microenvironment
MMEmelanoma microenvironment
CXCR4chemokine receptor type 4
CXCL12CXC motif chemokine ligand 12
SDF1stromal-derived factor-1
TGF-βtransforming growth factor beta
EMTepithelial-mesenchymal transition
HGFhepatocyte growth factor
VEGFvascular endothelial growth factor
FGF2fibroblast growth factor 2
IGF2Rinsulin-like growth factor 2 receptor
MAPKmitogen-activated protein kinase
EVextracellular vesicle
miRNAMicro-RNA
ILinterleukin
FAPFibroblast activation protein
DPPdipeptidyl peptidase
IFN-γinterferon-gamma
TNF-αtumor necrosis factor-alpha
VEPH1Ventricular Zone Expressed PH Domain Containing 1
SMAD4Mothers against decapentaplegic homolog 4
SKIsubtilisin/kexin isozyme
myCAFsMyofibroblastic CAFs
iCAFsinflammatory CAFs
apCAFsAntigen-presenting CAFs
VDRvitamin D receptor
PDGFRplatelet-derived growth factor receptor
FGFRfibroblast growth factor receptor
HhHedgehog
AT1Rangiotensin II type 1 receptor

References

  1. The Cancer Genome Atlas Network. Genomic Classification of Cutaneous Melanoma. Cell 2015, 161, 1681–1696. [Google Scholar] [CrossRef] [PubMed]
  2. Hayward, N.K.; Wilmott, J.S.; Waddell, N.; Johansson, P.A.; Field, M.A.; Nones, K.; Patch, A.M.; Kakavand, H.; Alexandrov, L.B.; Burke, H.; et al. Whole-genome landscapes of major melanoma subtypes. Nature 2017, 545, 175–180. [Google Scholar] [CrossRef] [PubMed]
  3. Randic, T.; Kozar, I.; Margue, C.; Utikal, J.; Kreis, S. NRAS mutant melanoma: Towards better therapies. Cancer Treat. Rev. 2021, 99, 102238. [Google Scholar] [CrossRef] [PubMed]
  4. 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]
  5. 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]
  6. Emery, C.M.; Vijayendran, K.G.; Zipser, M.C.; Sawyer, A.M.; Niu, L.; Kim, J.J.; Hatton, C.; Chopra, R.; Oberholzer, P.A.; Karpova, M.B.; et al. MEK1 mutations confer resistance to MEK and B-RAF inhibition. Proc. Natl. Acad. Sci. USA 2009, 106, 20411–20416. [Google Scholar] [CrossRef]
  7. Lugowska, I.; Teterycz, P.; Rutkowski, P. Immunotherapy of melanoma. Contemp. Oncol. 2018, 22, 61–67. [Google Scholar] [CrossRef]
  8. Ascierto, P.A.; Lipson, E.J.; Dummer, R.; Larkin, J.; Long, G.V.; Sanborn, R.E.; Chiarion-Sileni, V.; Dréno, B.; Dalle, S.; Schadendorf, D.; et al. Nivolumab and Relatlimab in Patients With Advanced Melanoma That Had Progressed on Anti-Programmed Death-1/Programmed Death Ligand 1 Therapy: Results From the Phase I/IIa RELATIVITY-020 Trial. J. Clin. Oncol. 2023, 41, 2724–2735. [Google Scholar] [CrossRef]
  9. Trocchia, M.; Ventrici, A.; Modestino, L.; Cristinziano, L.; Ferrara, A.L.; Palestra, F.; Loffredo, S.; Capone, M.; Madonna, G.; Romanelli, M.; et al. Innate Immune Cells in Melanoma: Implications for Immunotherapy. Int. J. Mol. Sci. 2024, 25, 8523. [Google Scholar] [CrossRef]
  10. 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]
  11. Maietta, I.; Viscusi, E.; Laudati, S.; Iannaci, G.; D’Antonio, A.; Melillo, R.M.; Motti, M.L.; De Falco, V. Targeting the p90RSK/MDM2/p53 Pathway Is Effective in Blocking Tumors with Oncogenic Up-Regulation of the MAPK Pathway Such as Melanoma and Lung Cancer. Cells 2024, 13, 1546. [Google Scholar] [CrossRef] [PubMed]
  12. Fratangelo, F.; Camerlingo, R.; Carriero, M.V.; Pirozzi, G.; Palmieri, G.; Gentilcore, G.; Ragone, C.; Minopoli, M.; Ascierto, P.A.; Motti, M.L. Effect of ABT-888 on the apoptosis, motility and invasiveness of BRAFi-resistant melanoma cells. Int. J. Oncol. 2018, 53, 1149–1159. [Google Scholar] [CrossRef] [PubMed]
  13. Motti, M.L.; Minopoli, M.; Di Carluccio, G.; Ascierto, P.A.; Carriero, M.V. MicroRNAs as Key Players in Melanoma Cell Resistance to MAPK and Immune Checkpoint Inhibitors. Int. J. Mol. Sci. 2020, 21, 4544. [Google Scholar] [CrossRef]
  14. Orlandella, F.M.; Arcone, R.; Luciano, N.; Salvatore, G.; Motti, M.L. Novel Biological Strategies for Melanoma Therapy: A Focus on lncRNAs and Their Targeting. Cancers 2025, 17, 1273. [Google Scholar] [CrossRef] [PubMed]
  15. Wang, Q.; Shao, X.; Zhang, Y.; Zhu, M.; Wang, F.X.C.; Mu, J.; Li, J.; Yao, H.; Chen, K. Role of tumor microenvironment in cancer progression and therapeutic strategy. Cancer Med. 2023, 12, 11149–11165. [Google Scholar] [CrossRef]
  16. Cao, Z.; Quazi, S.; Arora, S.; Osellame, L.D.; Burvenich, I.J.; Janes, P.W.; Scott, A.M. Cancer-associated fibroblasts as therapeutic targets for cancer: Advances, challenges, and future prospects. J. Biomed. Sci. 2025, 32, 7. [Google Scholar] [CrossRef]
  17. Chen, X.; Song, E. The theory of tumor ecosystem. Cancer Commun. 2022, 42, 587–608. [Google Scholar] [CrossRef]
  18. Luo, W. Nasopharyngeal carcinoma ecology theory: Cancer as multidimensional spatiotemporal “unity of ecology and evolution” pathological ecosystem. Theranostics 2023, 13, 1607–1631. [Google Scholar] [CrossRef]
  19. Ingangi, V.; Minopoli, M.; Ragone, C.; Motti, M.L.; Carriero, M.V. Role of Microenvironment on the Fate of Disseminating Cancer Stem Cells. Front. Oncol. 2019, 9, 82. [Google Scholar] [CrossRef]
  20. Shiga, K.; Hara, M.; Nagasaki, T.; Sato, T.; Takahashi, H.; Takeyama, H. Cancer-Associated Fibroblasts: Their Characteristics and Their Roles in Tumor Growth. Cancers 2015, 7, 2443–2458. [Google Scholar] [CrossRef]
  21. Hsu, M.Y.; Meier, F.; Herlyn, M. Melanoma development and progression: A conspiracy between tumor and host. Differ. Res. Biol. Divers. 2002, 70, 522–536. [Google Scholar] [CrossRef] [PubMed]
  22. Yuan, Y.; Jiang, Y.C.; Sun, C.K.; Chen, Q.M. Role of the tumor microenvironment in tumor progression and the clinical applications (Review). Oncol. Rep. 2016, 35, 2499–2515. [Google Scholar] [CrossRef]
  23. Jobe, N.P.; Rösel, D.; Dvořánková, B.; Kodet, O.; Lacina, L.; Mateu, R.; Smetana, K.; Brábek, J. Simultaneous blocking of IL-6 and IL-8 is sufficient to fully inhibit CAF-induced human melanoma cell invasiveness. Histochem. Cell Biol. 2016, 146, 205–217. [Google Scholar] [CrossRef] [PubMed]
  24. Straussman, R.; Morikawa, T.; Shee, K.; Barzily-Rokni, M.; Qian, Z.R.; Du, J.; Davis, A.; Mongare, M.M.; Gould, J.; Frederick, D.T.; et al. Tumour micro-environment elicits innate resistance to RAF inhibitors through HGF secretion. Nature 2012, 487, 500–504. [Google Scholar] [CrossRef]
  25. Capparelli, C.; Rosenbaum, S.; Berger, A.C.; Aplin, A.E. Fibroblast-derived neuregulin 1 promotes compensatory ErbB3 receptor signaling in mutant BRAF melanoma. J. Biol. Chem. 2015, 290, 24267–24277. [Google Scholar] [CrossRef]
  26. Caligiuri, G.; Tuveson, D.A. Activated fibroblasts in cancer: Perspectives and challenges. Cancer Cell 2023, 41, 434–449. [Google Scholar] [CrossRef]
  27. Feig, C.; Jones, J.O.; Kraman, M.; Wells, R.J.; Deonarine, A.; Chan, D.S.; Connell, C.M.; Roberts, E.W.; Zhao, Q.; Caballero, O.L.; et al. Targeting CXCL12 from FAP-expressing carcinoma-associated fibroblasts synergizes with anti-PD-L1 immunotherapy in pancreatic cancer. Proc. Natl. Acad. Sci. USA 2013, 110, 20212–20217. [Google Scholar] [CrossRef]
  28. Sun, X.; Cheng, G.; Hao, M.; Zheng, J.; Zhou, X.; Zhang, J.; Taichman, R.S.; Pienta, K.J.; Wang, J. CXCL12/CXCR4/CXCR7 chemokine axis and cancer progression. Cancer Metastasis Rev. 2010, 29, 709–722. [Google Scholar] [CrossRef]
  29. Anastasiadou, D.P.; Quesnel, A.; Duran, C.L.; Filippou, P.S.; Karagiannis, G.S. An emerging paradigm of CXCL12 involvement in the metastatic cascade. Cytokine Growth Factor Rev. 2024, 75, 12–30. [Google Scholar] [CrossRef]
  30. Wendel, C.; Hemping-Bovenkerk, A.; Krasnyanska, J.; Mees, S.T.; Kochetkova, M.; Stoeppeler, S.; Haier, J. CXCR4/CXCL12 Participate in Extravasation of Metastasizing Breast Cancer Cells within the Liver in a Rat Model. PLoS ONE 2012, 7, e30046. [Google Scholar] [CrossRef]
  31. Ieranò, C.; D’Alterio, C.; Giarra, S.; Napolitano, M.; Rea, G.; Portella, L.; Santagata, A.; Trotta, A.M.; Barbieri, A.; Campani, V.; et al. CXCL12 loaded-dermal filler captures CXCR4 expressing melanoma circulating tumor cells. Cell Death Dis. 2019, 10, 562. [Google Scholar] [CrossRef] [PubMed]
  32. Orimo, A.; Gupta, P.B.; Sgroi, D.C.; Arenzana-Seisdedos, F.; Delaunay, T.; Naeem, R.; Carey, V.J.; Richardson, A.L.; Weinberg, R.A. Stromal fibroblasts present in invasive human breast carcinomas promote tumor growth and angiogenesis through elevated SDF-1/CXCL12 secretion. Cell 2005, 121, 335–348. [Google Scholar] [CrossRef] [PubMed]
  33. Fukumura, D.; Xavier, R.; Sugiura, T.; Chen, Y.; Park, E.C.; Lu, N.; Selig, M.; Nielsen, G.; Taksir, T.; Jain, R.K.; et al. Tumor induction of VEGF promoter activity in stromal cells. Cell 1998, 94, 715–725. [Google Scholar] [CrossRef]
  34. Okabe, H.; Beppu, T.; Hayashi, H.; Ishiko, T.; Masuda, T.; Otao, R.; Horlad, H.; Jono, H.; Ueda, M.; Shinriki, S.; et al. Hepatic stellate cells accelerate the malignant behavior of cholangiocarcinoma cells. Ann. Surg. Oncol. 2011, 18, 1175–1184. [Google Scholar] [CrossRef]
  35. Guo, X.; Oshima, H.; Kitmura, T.; Taketo, M.M.; Oshima, M. Stromal fibroblasts activated by tumor cells promote angiogenesis in mouse gastric cancer. J. Biol. Chem. 2008, 283, 19864–19871. [Google Scholar] [CrossRef]
  36. Hao, Y.; Baker, D.; Ten Dijke, P. TGF-β-Mediated Epithelial-Mesenchymal Transition and Cancer Metastasis. Int. J. Mol. Sci. 2019, 20, 2767. [Google Scholar] [CrossRef]
  37. Ozdamar, B.; Bose, R.; Barrios-Rodiles, M.; Wang, H.R.; Zhang, Y.; Wrana, J.L. Regulation of the polarity protein Par6 by TGFbeta receptors controls epithelial cell plasticity. Science 2005, 307, 1603–1609. [Google Scholar] [CrossRef]
  38. Zhang, Y.; Alexander, P.B.; Wang, X.F. TGF-β Family Signaling in the Control of Cell Proliferation and Survival. Cold Spring Harb. Perspect. Biol. 2017, 9, a022145. [Google Scholar] [CrossRef]
  39. Yang, L.; Pang, Y.; Moses, H.L. TGF-beta and immune cells: An important regulatory axis in the tumor microenvironment and progression. Trends Immunol. 2010, 31, 220–227. [Google Scholar] [CrossRef]
  40. Zalpoor, H.; Aziziyan, F.; Liaghat, M.; Bakhtiyari, M.; Akbari, A.; Nabi-Afjadi, M.; Forghaniesfidvajani, R.; Rezaei, N. The roles of metabolic profiles and intracellular signaling pathways of tumor microenvironment cells in angiogenesis of solid tumors. Cell Commun. Signal. 2022, 20, 186. [Google Scholar] [CrossRef]
  41. de Visser, K.E.; Joyce, J.A. The evolving tumor microenvironment: From cancer initiation to metastatic outgrowth. Cancer Cell 2023, 41, 374–403. [Google Scholar] [CrossRef] [PubMed]
  42. Zhao, Z.; Li, T.; Sun, L.; Yuan, Y.; Zhu, Y. Potential mechanisms of cancer-associated fibroblasts in therapeutic resistance. Biomed. Pharmacother. 2023, 166, 115425. [Google Scholar] [CrossRef] [PubMed]
  43. Perna, A.; Sellitto, C.; Komici, K.; Hay, E.; Rocca, A.; De Blasiis, P.; Lucariello, A.; Moccia, F.; Guerra, G. Transient Receptor Potential (TRP) Channels in Tumor Vascularization. Int. J. Mol. Sci. 2022, 23, 14253. [Google Scholar] [CrossRef] [PubMed]
  44. Knauf, J.A.; Luckett, K.A.; Chen, K.Y.; Voza, F.; Socci, N.D.; Ghossein, R.; Fagin, J.A. Hgf/Met activation mediates resistance to BRAF inhibition in murine anaplastic thyroid cancers. J. Clin. Investig. 2018, 128, 4086–4097. [Google Scholar] [CrossRef]
  45. Xia, Y.; Huang, C.; Zhong, M.; Zhong, H.; Ruan, R.; Xiong, J.; Yao, Y.; Zhou, J.; Deng, J. Targeting HGF/c-MET signaling to regulate the tumor microenvironment: Implications for counteracting tumor immune evasion. Cell Commun. Signal. 2025, 23, 46. [Google Scholar] [CrossRef]
  46. Maia, J.; Caja, S.; Strano Moraes, M.C.; Couto, N.; Costa-Silva, B. Exosome-Based Cell-Cell Communication in the Tumor Microenvironment. Front. Cell Dev. Biol. 2018, 6, 18. [Google Scholar] [CrossRef]
  47. Matsumoto, A.; Takahashi, Y.; Nishikawa, M.; Sano, K.; Morishita, M.; Charoenviriyakul, C.; Saji, H.; Takakura, Y. Accelerated growth of B16BL6 tumor in mice through efficient uptake of their own exosomes by B16BL6 cells. Cancer Sci. 2017, 108, 1803–1810. [Google Scholar] [CrossRef]
  48. Dror, S.; Sander, L.; Schwartz, H.; Sheinboim, D.; Barzilai, A.; Dishon, Y.; Apcher, S.; Golan, T.; Greenberger, S.; Barshack, I.; et al. Melanoma miRNA trafficking controls tumour primary niche formation. Nat. Cell Biol. 2016, 18, 1006–1017. [Google Scholar] [CrossRef]
  49. Avagliano, A.; Granato, G.; Ruocco, M.R.; Romano, V.; Belviso, I.; Carfora, A.; Montagnani, S.; Arcucci, A. Metabolic Reprogramming of Cancer Associated Fibroblasts: The Slavery of Stromal Fibroblasts. BioMed Res. Int. 2018, 2018, 6075403. [Google Scholar] [CrossRef]
  50. Villanueva, J.; Herlyn, M. Melanoma and the tumor microenvironment. Curr. Oncol. Rep. 2008, 10, 439–446. [Google Scholar] [CrossRef]
  51. Nazemi, M.; Rainero, E. Cross-Talk Between the Tumor Microenvironment, Extracellular Matrix, and Cell Metabolism in Cancer. Front. Oncol. 2020, 10, 239. [Google Scholar] [CrossRef] [PubMed]
  52. Ju, R.J.; Stehbens, S.J.; Haass, N.K. The Role of Melanoma Cell-Stroma Interaction in Cell Motility, Invasion, and Metastasis. Front. Med. 2018, 5, 307. [Google Scholar] [CrossRef] [PubMed]
  53. Ahmadzadeh, H.; Webster, M.R.; Behera, R.; Jimenez Valencia, A.M.; Wirtz, D.; Weeraratna, A.T.; Shenoy, V.B. Modeling the two-way feedback between contractility and matrix realignment reveals a nonlinear mode of cancer cell invasion. Proc. Natl. Acad. Sci. USA 2017, 114, E1617–E1626. [Google Scholar] [CrossRef] [PubMed]
  54. Romano, V.; Belviso, I.; Venuta, A.; Ruocco, M.R.; Masone, S.; Aliotta, F.; Fiume, G.; Montagnani, S.; Avagliano, A.; Arcucci, A. Influence of Tumor Microenvironment and Fibroblast Population Plasticity on Melanoma Growth, Therapy Resistance and Immunoescape. Int. J. Mol. Sci. 2021, 22, 5283. [Google Scholar] [CrossRef]
  55. Hynes, R.O.; Naba, A. Overview of the matrisome--an inventory of extracellular matrix constituents and functions. Cold Spring Harb. Perspect. Biol. 2012, 4, a004903. [Google Scholar] [CrossRef]
  56. Papaccio, F.; Kovacs, D.; Bellei, B.; Caputo, S.; Migliano, E.; Cota, C.; Picardo, M. Profiling Cancer-Associated Fibroblasts in Melanoma. Int. J. Mol. Sci. 2021, 22, 7255. [Google Scholar] [CrossRef]
  57. Mahabeleshwar, G.H.; Byzova, T.V. Angiogenesis in melanoma. Semin. Oncol. 2007, 34, 555–565. [Google Scholar] [CrossRef]
  58. Pasco, S.; Brassart, B.; Ramont, L.; Maquart, F.X.; Monboisse, J.C. Control of melanoma cell invasion by type IV collagen. Cancer Detect. Prev. 2005, 29, 260–266. [Google Scholar] [CrossRef]
  59. Kramer, R.H.; Marks, N. Identification of integrin collagen receptors on human melanoma cells. J. Biol. Chem. 1989, 264, 4684–4688. [Google Scholar] [CrossRef]
  60. Mott, J.D.; Werb, Z. Regulation of matrix biology by matrix metalloproteinases. Curr. Opin. Cell Biol. 2004, 16, 558–564. [Google Scholar] [CrossRef]
  61. Wang, F.T.; Sun, W.; Zhang, J.T.; Fan, Y.Z. Cancer-associated fibroblast regulation of tumor neo-angiogenesis as a therapeutic target in cancer. Oncol. Lett. 2019, 17, 3055–3065. [Google Scholar] [CrossRef] [PubMed]
  62. Moogk, D.; da Silva, I.P.; Ma, M.W.; Friedman, E.B.; de Miera, E.V.; Darvishian, F.; Scanlon, P.; Perez-Garcia, A.; Pavlick, A.C.; Bhardwaj, N.; et al. Melanoma expression of matrix metalloproteinase-23 is associated with blunted tumor immunity and poor responses to immunotherapy. J. Transl. Med. 2014, 12, 342. [Google Scholar] [CrossRef] [PubMed]
  63. Lazar, I.; Clement, E.; Ducoux-Petit, M.; Denat, L.; Soldan, V.; Dauvillier, S.; Balor, S.; Burlet-Schiltz, O.; Larue, L.; Muller, C.; et al. Proteome characterization of melanoma exosomes reveals a specific signature for metastatic cell lines. Pigment Cell Melanoma Res. 2015, 28, 464–475. [Google Scholar] [CrossRef] [PubMed]
  64. Gener Lahav, T.; Adler, O.; Zait, Y.; Shani, O.; Amer, M.; Doron, H.; Abramovitz, L.; Yofe, I.; Cohen, N.; Erez, N. Melanoma-derived extracellular vesicles instigate proinflammatory signaling in the metastatic microenvironment. Int. J. Cancer 2019, 145, 2521–2534. [Google Scholar] [CrossRef]
  65. Hanahan, D.; Coussens, L.M. Accessories to the crime: Functions of cells recruited to the tumor microenvironment. Cancer Cell 2012, 21, 309–322. [Google Scholar] [CrossRef]
  66. Obenauf, A.C.; Massagué, J. Surviving at a Distance: Organ-Specific Metastasis. Trends Cancer 2015, 1, 76–91. [Google Scholar] [CrossRef]
  67. Gaggioli, C.; Hooper, S.; Hidalgo-Carcedo, C.; Grosse, R.; Marshall, J.F.; Harrington, K.; Sahai, E. Fibroblast-led collective invasion of carcinoma cells with differing roles for RhoGTPases in leading and following cells. Nat. Cell Biol. 2007, 9, 1392–1400. [Google Scholar] [CrossRef]
  68. Malanchi, I.; Santamaria-Martínez, A.; Susanto, E.; Peng, H.; Lehr, H.A.; Delaloye, J.F.; Huelsken, J. Interactions between cancer stem cells and their niche govern metastatic colonization. Nature 2011, 481, 85–89. [Google Scholar] [CrossRef]
  69. Wilczak, M.; Surman, M.; PrzybyłO, M. Melanoma-derived extracellular vesicles transfer proangiogenic factors. Oncol. Res. 2025, 33, 245–262. [Google Scholar] [CrossRef]
  70. Hutchenreuther, J.; Vincent, K.; Norley, C.; Racanelli, M.; Gruber, S.B.; Johnson, T.M.; Fullen, D.R.; Raskin, L.; Perbal, B.; Holdsworth, D.W.; et al. Activation of cancer-associated fibroblasts is required for tumor neovascularization in a murine model of melanoma. Matrix Biol. 2018, 74, 52–61. [Google Scholar] [CrossRef]
  71. De Palma, M.; Biziato, D.; Petrova, T.V. Microenvironmental regulation of tumour angiogenesis. Nat. Rev. Cancer 2017, 17, 457–474. [Google Scholar] [CrossRef] [PubMed]
  72. Zhou, X.; Yan, T.; Huang, C.; Xu, Z.; Wang, L.; Jiang, E.; Wang, H.; Chen, Y.; Liu, K.; Shao, Z.; et al. Melanoma cell-secreted exosomal miR-155-5p induce proangiogenic switch of cancer-associated fibroblasts via SOCS1/JAK2/STAT3 signaling pathway. J. Exp. Clin. Cancer Res. 2018, 37, 242. [Google Scholar] [CrossRef] [PubMed]
  73. Forsthuber, A.; Aschenbrenner, B.; Korosec, A.; Jacob, T.; Annusver, K.; Krajic, N.; Kholodniuk, D.; Frech, S.; Zhu, S.; Purkhauser, K.; et al. Cancer-associated fibroblast subtypes modulate the tumor-immune microenvironment and are associated with skin cancer malignancy. Nat. Commun. 2024, 15, 9678. [Google Scholar] [CrossRef]
  74. Scala, S.; Giuliano, P.; Ascierto, P.A.; Ieranò, C.; Franco, R.; Napolitano, M.; Ottaiano, A.; Lombardi, M.L.; Luongo, M.; Simeone, E.; et al. Human melanoma metastases express functional CXCR4. Clin. Cancer Res. 2006, 12, 2427–2433. [Google Scholar] [CrossRef]
  75. Toyozawa, S.; Kaminaka, C.; Furukawa, F.; Nakamura, Y.; Matsunaka, H.; Yamamoto, Y. Chemokine receptor CXCR4 is a novel marker for the progression of cutaneous malignant melanomas. Acta Histochem. Et Cytochem. 2012, 45, 293–299. [Google Scholar] [CrossRef]
  76. Duda, D.G.; Kozin, S.V.; Kirkpatrick, N.D.; Xu, L.; Fukumura, D.; Jain, R.K. CXCL12 (SDF1alpha)-CXCR4/CXCR7 pathway inhibition: An emerging sensitizer for anticancer therapies? Clin. Cancer Res. 2011, 17, 2074–2080. [Google Scholar] [CrossRef]
  77. Kim, M.; Koh, Y.J.; Kim, K.E.; Koh, B.I.; Nam, D.H.; Alitalo, K.; Kim, I.; Koh, G.Y. CXCR4 signaling regulates metastasis of chemoresistant melanoma cells by a lymphatic metastatic niche. Cancer Res. 2010, 70, 10411–10421. [Google Scholar] [CrossRef]
  78. Righi, E.; Kashiwagi, S.; Yuan, J.; Santosuosso, M.; Leblanc, P.; Ingraham, R.; Forbes, B.; Edelblute, B.; Collette, B.; Xing, D.; et al. CXCL12/CXCR4 blockade induces multimodal antitumor effects that prolong survival in an immunocompetent mouse model of ovarian cancer. Cancer Res. 2011, 71, 5522–5534. [Google Scholar] [CrossRef]
  79. Saxena, R.; Wang, Y.; Mier, J.W. CXCR4 inhibition modulates the tumor microenvironment and retards the growth of B16-OVA melanoma and Renca tumors. Melanoma Res. 2020, 30, 14–25. [Google Scholar] [CrossRef]
  80. Andtbacka, R.H.I.; Wang, Y.; Pierce, R.H.; Campbell, J.S.; Yushak, M.; Milhem, M.; Ross, M.; Niland, K.; Arbeit, R.D.; Parasuraman, S.; et al. Mavorixafor, an Orally Bioavailable CXCR4 Antagonist, Increases Immune Cell Infiltration and Inflammatory Status of Tumor Microenvironment in Patients with Melanoma. Cancer Res. Commun. 2022, 2, 904–913. [Google Scholar] [CrossRef]
  81. Erdag, G.; Schaefer, J.T.; Smolkin, M.E.; Deacon, D.H.; Shea, S.M.; Dengel, L.T.; Patterson, J.W.; Slingluff, C.L., Jr. Immunotype and immunohistologic characteristics of tumor-infiltrating immune cells are associated with clinical outcome in metastatic melanoma. Cancer Res. 2012, 72, 1070–1080. [Google Scholar] [CrossRef] [PubMed]
  82. Galon, J.; Angell, H.K.; Bedognetti, D.; Marincola, F.M. The continuum of cancer immunosurveillance: Prognostic, predictive, and mechanistic signatures. Immunity 2013, 39, 11–26. [Google Scholar] [CrossRef] [PubMed]
  83. Pagès, F.; Berger, A.; Camus, M.; Sanchez-Cabo, F.; Costes, A.; Molidor, R.; Mlecnik, B.; Kirilovsky, A.; Nilsson, M.; Damotte, D.; et al. Effector memory T cells, early metastasis, and survival in colorectal cancer. N. Engl. J. Med. 2005, 353, 2654–2666. [Google Scholar] [CrossRef] [PubMed]
  84. Zaretsky, J.M.; Garcia-Diaz, A.; Shin, D.S.; Escuin-Ordinas, H.; Hugo, W.; Hu-Lieskovan, S.; Torrejon, D.Y.; Abril-Rodriguez, G.; Sandoval, S.; Barthly, L.; et al. Mutations Associated with Acquired Resistance to PD-1 Blockade in Melanoma. N. Engl. J. Med. 2016, 375, 819–829. [Google Scholar] [CrossRef]
  85. Crane, J.N.; Graham, D.S.; Mona, C.E.; Nelson, S.D.; Samiei, A.; Dawson, D.W.; Dry, S.M.; Masri, M.G.; Crompton, J.G.; Benz, M.R.; et al. Fibroblast Activation Protein Expression in Sarcomas. Sarcoma 2023, 2023, 2480493. [Google Scholar] [CrossRef]
  86. Lyu, Z.; Li, Y.; Zhu, D.; Wu, S.; Hu, F.; Zhang, Y.; Li, Y.; Hou, T. Fibroblast Activation Protein-Alpha is a Prognostic Biomarker Associated With Ferroptosis in Stomach Adenocarcinoma. Front. Cell Dev. Biol. 2022, 10, 859999. [Google Scholar] [CrossRef]
  87. Barrett, R.L.; Puré, E. Cancer-associated fibroblasts and their influence on tumor immunity and immunotherapy. eLife 2020, 9, e57243. [Google Scholar] [CrossRef]
  88. Jorgenson, L.C.; Torbenson, M.S.; Halfdanarson, T.R.; Kankeu Fonkoua, L.A.; Tran, N.H.; Roberts, L.R.; Smoot, R.L.; Goenka, A.H.; Thompson, S.M. Immunohistochemical basis for FAP as a candidate theranostic target across a broad range of cholangiocarcinoma subtypes. Front. Nucl. Med. 2024, 4, 1480471. [Google Scholar] [CrossRef]
  89. Capaccione, K.M.; Doubrovin, M.; Braumuller, B.; Leibowitz, D.; Bhatt, N.; Momen-Heravi, F.; Molotkov, A.; Kissner, M.; Goldner, K.; Soffing, M.; et al. Evaluating the Combined Anticancer Response of Checkpoint Inhibitor Immunotherapy and FAP-Targeted Molecular Radiotherapy in Murine Models of Melanoma and Lung Cancer. Cancers 2022, 14, 4575. [Google Scholar] [CrossRef]
  90. Munoz-Couselo, E.; Soria Rivas, A.; Sandhu, S.; Long, G.V.; Sanmamed, M.F.; Spreafico, A.; Buchbinder, E.; Sznol, M.; Prenen, H.; Fedenko, A.; et al. Phase Ib Study of Immunocytokine Simlukafusp Alfa (FAP-IL2v) Combined with Pembrolizumab for Treatment of Advanced and/or Metastatic Melanoma. Cancer Res. Commun. 2025, 5, 358–368. [Google Scholar] [CrossRef]
  91. Cunningham, C.C. Talabostat. Expert Opin. Investig. Drugs 2007, 16, 1459–1465. [Google Scholar] [CrossRef] [PubMed]
  92. Eager, R.M.; Cunningham, C.C.; Senzer, N.N.; Stephenson, J., Jr.; Anthony, S.P.; O’Day, S.J.; Frenette, G.; Pavlick, A.C.; Jones, B.; Uprichard, M.; et al. Phase II assessment of talabostat and cisplatin in second-line stage IV melanoma. BMC Cancer 2009, 9, 263. [Google Scholar] [CrossRef]
  93. Wang, Y.; Chu, J.; Yi, P.; Dong, W.; Saultz, J.; Wang, Y.; Wang, H.; Scoville, S.; Zhang, J.; Wu, L.C.; et al. SMAD4 promotes TGF-β-independent NK cell homeostasis and maturation and antitumor immunity. J. Clin. Investig. 2018, 128, 5123–5136. [Google Scholar] [CrossRef]
  94. Lauden, L.; Siewiera, J.; Boukouaci, W.; Ramgolam, K.; Mourah, S.; Lebbe, C.; Charron, D.; Aoudjit, F.; Jabrane-Ferrat, N.; Al-Daccak, R. TGF-β-induced (TGFBI) protein in melanoma: A signature of high metastatic potential. J. Investig. Dermatol. 2014, 134, 1675–1685. [Google Scholar] [CrossRef]
  95. Jenkins, M.H.; Croteau, W.; Mullins, D.W.; Brinckerhoff, C.E. The BRAF(V600E) inhibitor, PLX4032, increases type I collagen synthesis in melanoma cells. Matrix Biol. 2015, 48, 66–77. [Google Scholar] [CrossRef]
  96. Jiang, J.; Zhang, Y.; Peng, K.; Wang, Q.; Hong, X.; Li, H.; Fan, G.; Zhang, Z.; Gong, T.; Sun, X. Combined delivery of a TGF-β inhibitor and an adenoviral vector expressing interleukin-12 potentiates cancer immunotherapy. Acta Biomater. 2017, 61, 114–123. [Google Scholar] [CrossRef]
  97. Wilson, E.B.; El-Jawhari, J.J.; Neilson, A.L.; Hall, G.D.; Melcher, A.A.; Meade, J.L.; Cook, G.P. Human tumour immune evasion via TGF-β blocks NK cell activation but not survival allowing therapeutic restoration of anti-tumour activity. PLoS ONE 2011, 6, e22842. [Google Scholar] [CrossRef]
  98. DaCosta Byfield, S.; Major, C.; Laping, N.J.; Roberts, A.B. SB-505124 is a selective inhibitor of transforming growth factor-beta type I receptors ALK4, ALK5, and ALK7. Mol. Pharmacol. 2004, 65, 744–752. [Google Scholar] [CrossRef]
  99. Morris, J.C.; Tan, A.R.; Olencki, T.E.; Shapiro, G.I.; Dezube, B.J.; Reiss, M.; Hsu, F.J.; Berzofsky, J.A.; Lawrence, D.P. Phase I study of GC1008 (fresolimumab): A human anti-transforming growth factor-beta (TGFβ) monoclonal antibody in patients with advanced malignant melanoma or renal cell carcinoma. PLoS ONE 2014, 9, e90353. [Google Scholar] [CrossRef]
  100. Szczygielski, O.; Dąbrowska, E.; Niemyjska, S.; Przylipiak, A.; Zajkowska, M. Targeting Matrix Metalloproteinases and Their Inhibitors in Melanoma. Int. J. Mol. Sci. 2024, 25, 13558. [Google Scholar] [CrossRef]
  101. Winer, A.; Adams, S.; Mignatti, P. Matrix Metalloproteinase Inhibitors in Cancer Therapy: Turning Past Failures Into Future Successes. Mol. Cancer Ther. 2018, 17, 1147–1155. [Google Scholar] [CrossRef] [PubMed]
  102. Quirt, I.; Bodurth, A.; Lohmann, R.; Rusthoven, J.; Belanger, K.; Young, V.; Wainman, N.; Stewar, W.; Eisenhauer, E. Phase II study of marimastat (BB-2516) in malignant melanoma: A clinical and tumor biopsy study of the National Cancer Institute of Canada Clinical Trials Group. Investig. N. Drugs 2002, 20, 431–437. [Google Scholar] [CrossRef] [PubMed]
  103. Ragone, C.; Minopoli, M.; Ingangi, V.; Botti, G.; Fratangelo, F.; Pessi, A.; Stoppelli, M.P.; Ascierto, P.A.; Ciliberto, G.; Motti, M.L.; et al. Targeting the cross-talk between Urokinase receptor and Formyl peptide receptor type 1 to prevent invasion and trans-endothelial migration of melanoma cells. J. Exp. Clin. Cancer Res. 2017, 36, 180. [Google Scholar] [CrossRef] [PubMed]
  104. Fröhlich, E. Proteases in cutaneous malignant melanoma: Relevance as biomarker and therapeutic target. Cell. Mol. Life Sci. 2010, 67, 3947–3960. [Google Scholar] [CrossRef]
  105. Marconi, C.; Bianchini, F.; Mannini, A.; Mugnai, G.; Ruggieri, S.; Calorini, L. Tumoral and macrophage uPAR and MMP-9 contribute to the invasiveness of B16 murine melanoma cells. Clin. Exp. Metastasis 2008, 25, 225–231. [Google Scholar] [CrossRef]
  106. Muralidhar, S.; Filia, A.; Nsengimana, J.; Poźniak, J.; O’Shea, S.J.; Diaz, J.M.; Harland, M.; Randerson-Moor, J.A.; Reichrath, J.; Laye, J.P.; et al. Vitamin D–VDR Signaling Inhibits Wnt/β-Catenin–Mediated Melanoma Progression and Promotes Antitumor Immunity. Cancer Res. 2019, 79, 5986–5998. [Google Scholar] [CrossRef]
  107. Wei, J.; Zhan, J.; Ji, H.; Xu, Y.; Xu, Q.; Zhu, X.; Liu, Y. Fibroblast Upregulation of Vitamin D Receptor Represents a Self-Protective Response to Limit Fibroblast Proliferation and Activation during Pulmonary Fibrosis. Antioxidants 2023, 12, 1634. [Google Scholar] [CrossRef]
  108. Otake, A.H.; Mattar, A.L.; Freitas, H.C.; Machado, C.M.; Nonogaki, S.; Fujihara, C.K.; Zatz, R.; Chammas, R. Inhibition of angiotensin II receptor 1 limits tumor-associated angiogenesis and attenuates growth of murine melanoma. Cancer Chemother. Pharmacol. 2010, 66, 79–87. [Google Scholar] [CrossRef]
  109. Catarata, M.J.; Ribeiro, R.; Oliveira, M.J.; Robalo Cordeiro, C.; Medeiros, R. Renin-Angiotensin System in Lung Tumor and Microenvironment Interactions. Cancers 2020, 12, 1457. [Google Scholar] [CrossRef]
  110. Pinter, M.; Jain, R.K. Targeting the renin-angiotensin system to improve cancer treatment: Implications for immunotherapy. Sci. Transl. Med. 2017, 9, eaan5616. [Google Scholar] [CrossRef]
  111. Egami, K.; Murohara, T.; Shimada, T.; Sasaki, K.; Shintani, S.; Sugaya, T.; Ishii, M.; Akagi, T.; Ikeda, H.; Matsuishi, T.; et al. Role of host angiotensin II type 1 receptor in tumor angiogenesis and growth. J. Clin. Investig. 2003, 112, 67–75. [Google Scholar] [CrossRef] [PubMed]
  112. Bournazou, E.; Bromberg, J. Targeting the tumor microenvironment: JAK-STAT3 signaling. Jak-Stat 2013, 2, e23828. [Google Scholar] [CrossRef] [PubMed]
  113. Wang, J.; Cui, B.; Li, X.; Zhao, X.; Huang, T.; Ding, X. The emerging roles of Hedgehog signaling in tumor immune microenvironment. Front. Oncol. 2023, 13, 1171418. [Google Scholar] [CrossRef] [PubMed]
  114. Goebeler, M.; Kaufmann, D.; Bröcker, E.B.; Klein, C.E. Migration of highly aggressive melanoma cells on hyaluronic acid is associated with functional changes, increased turnover and shedding of CD44 receptors. J. Cell Sci. 1996, 109 Pt 7, 1957–1964. [Google Scholar] [CrossRef]
  115. Mayasin, Y.P.; Osinnikova, M.N.; Kharisova, C.B.; Kitaeva, K.V.; Filin, I.Y.; Gorodilova, A.V.; Kutovoi, G.I.; Solovyeva, V.V.; Golubev, A.I.; Rizvanov, A.A. Extracellular Matrix as a Target in Melanoma Therapy: From Hypothesis to Clinical Trials. Cells 2024, 13, 1917. [Google Scholar] [CrossRef]
  116. Hsu, C.Y.; Saleh, R.O.; Mohammed, J.S.; Mansuri, N.; Rekha, M.M.; Kundlas, M.; Anand, A.; Sahoo, S.; Zwamel, A.H.; Hulail, H.M. The dynamic interplay between melanoma cells and CAFs: Implications drug resistance and immune evasion and possible therapeutics. Exp. Cell Res. 2025, 449, 114581. [Google Scholar] [CrossRef]
  117. Nurmik, M.; Ullmann, P.; Rodriguez, F.; Haan, S.; Letellier, E. In search of definitions: Cancer-associated fibroblasts and their markers. Int. J. Cancer 2020, 146, 895–905. [Google Scholar] [CrossRef]
  118. Ping, Q.; Yan, R.; Cheng, X.; Wang, W.; Zhong, Y.; Hou, Z.; Shi, Y.; Wang, C.; Li, R. Cancer-associated fibroblasts: Overview, progress, challenges, and directions. Cancer Gene Ther. 2021, 28, 984–999. [Google Scholar] [CrossRef]
  119. Feng, B.; Wu, J.; Shen, B.; Jiang, F.; Feng, J. Cancer-associated fibroblasts and resistance to anticancer therapies: Status, mechanisms, and countermeasures. Cancer Cell Int. 2022, 22, 166. [Google Scholar] [CrossRef]
Figure 1. Key roles of CAFs in melanoma progression. Schematic representation of the key roles of fibroblasts activated by melanoma-derived signals in promoting tumor development and progression. Metalloproitenase (MMP), extracellular matrix (ECM), CXC motif chemokine ligand 12 (CXCL12), hepatocyte growth factor (HGF), fibroblast growth factor (FGF), vascular endothelial growth factor (VEGF), platelet-derived growth factor (PDGF), Epithelial- mesenchymal transition (EMT), transforming growth factor beta (TGF-β).
Figure 1. Key roles of CAFs in melanoma progression. Schematic representation of the key roles of fibroblasts activated by melanoma-derived signals in promoting tumor development and progression. Metalloproitenase (MMP), extracellular matrix (ECM), CXC motif chemokine ligand 12 (CXCL12), hepatocyte growth factor (HGF), fibroblast growth factor (FGF), vascular endothelial growth factor (VEGF), platelet-derived growth factor (PDGF), Epithelial- mesenchymal transition (EMT), transforming growth factor beta (TGF-β).
Ijms 26 06132 g001
Table 2. CAFs comprise heterogeneous subtypes with distinct origins and functions within the TME. Among the main categories are myofibroblastic CAFs (myCAFs), inflammatory CAFs (iCAFs), and antigen-presenting CAFs (apCAFs).
Table 2. CAFs comprise heterogeneous subtypes with distinct origins and functions within the TME. Among the main categories are myofibroblastic CAFs (myCAFs), inflammatory CAFs (iCAFs), and antigen-presenting CAFs (apCAFs).
CAFs SubtypesFunctions in the TME
Myofibroblastic CAFs (myCAFs)involved in extracellular matrix remodeling and collagen deposition, contributing to increased tissue stiffness, tumor progression, and metastasis
Inflammatory CAFs (iCAFs)secrete cytokines and chemokines that modulate the immune response, promoting immune evasion, cancer cell survival, and proliferation
Antigen-presenting CAFs (apCAFs)characterized by the expression of MHC class II molecules, they may interact with T cells, although their immunological role is still being clarified
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

Belviso, I.; Pastore, R.; Mileo, A.; Del Genio, E.; Boccia, S.; Palermi, S.; Sellitto, C.; Motti, M.L. The Role of Fibroblasts in Melanoma Development: From Tumor Microenvironment Remodeling to Pre-Metastatic Niche Formation. Int. J. Mol. Sci. 2025, 26, 6132. https://doi.org/10.3390/ijms26136132

AMA Style

Belviso I, Pastore R, Mileo A, Del Genio E, Boccia S, Palermi S, Sellitto C, Motti ML. The Role of Fibroblasts in Melanoma Development: From Tumor Microenvironment Remodeling to Pre-Metastatic Niche Formation. International Journal of Molecular Sciences. 2025; 26(13):6132. https://doi.org/10.3390/ijms26136132

Chicago/Turabian Style

Belviso, Immacolata, Raffaele Pastore, Aldo Mileo, Emiliano Del Genio, Stefania Boccia, Stefano Palermi, Carmine Sellitto, and Maria Letizia Motti. 2025. "The Role of Fibroblasts in Melanoma Development: From Tumor Microenvironment Remodeling to Pre-Metastatic Niche Formation" International Journal of Molecular Sciences 26, no. 13: 6132. https://doi.org/10.3390/ijms26136132

APA Style

Belviso, I., Pastore, R., Mileo, A., Del Genio, E., Boccia, S., Palermi, S., Sellitto, C., & Motti, M. L. (2025). The Role of Fibroblasts in Melanoma Development: From Tumor Microenvironment Remodeling to Pre-Metastatic Niche Formation. International Journal of Molecular Sciences, 26(13), 6132. https://doi.org/10.3390/ijms26136132

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