Next Article in Journal
Beyond Lipids and Platelets: A Review of Anti-Inflammatory Strategies in Secondary Prevention of Acute Coronary Syndromes
Previous Article in Journal
Psychometric Properties of the Five-Item Ultrashort Oral Health Impact Profile (OHIP5) in the Serbian Cultural Environment: A Cross-Sectional Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Pathway-Specific Therapeutic Modulation of Melanoma: Small-Molecule Inhibition of BRAF–MEK and KIT Signaling in Contemporary Precision Oncology with a Special Focus on Vemurafenib, Trametinib, and Imatinib

1
Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Medical University of Gdańsk, 80-416 Gdańsk, Poland
2
Department of Nursing and Medical Rescue, Institute of Health Sciences, Pomeranian University in Słupsk, 76-200 Słupsk, Poland
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2025, 14(22), 7906; https://doi.org/10.3390/jcm14227906
Submission received: 8 October 2025 / Revised: 3 November 2025 / Accepted: 6 November 2025 / Published: 7 November 2025
(This article belongs to the Special Issue Clinical Advances in the Management of Melanoma)

Abstract

Melanoma is an aggressive form of skin cancer marked by unique genetic alterations that promote tumor growth and resistance to therapy. Advances in targeted therapy have markedly improved clinical outcomes by selectively inhibiting key oncogenic pathways. This review focuses on three clinically relevant agents—vemurafenib, trametinib, and imatinib—analyzing their mechanisms of action, clinical applications, efficacy, and limitations. Vemurafenib, a selective BRAFV600E inhibitor, significantly extends progression-free and overall survival in BRAF-mutant melanoma but is limited by acquired resistance and frequent cutaneous toxicities. Trametinib, a MEK1/2 inhibitor, acts downstream in the MAPK pathway and is typically combined with BRAF inhibitors to enhance efficacy and delay resistance. Imatinib, targeting c-KIT and PDGFR mutations, demonstrates therapeutic benefit primarily in acral and mucosal melanoma subtypes, though with lower response rates than BRAF-directed therapies. Adverse events associated with these drugs are generally manageable with appropriate monitoring. Despite substantial advances, secondary mutations and reactivation of oncogenic signaling remain major challenges. This narrative review integrates data from clinical, preclinical, and real-world studies to update the current understanding of targeted therapies in cutaneous melanoma and highlight ongoing research aimed at overcoming resistance and optimizing personalized treatment strategies.

1. Introduction

Cutaneous melanoma is a severe and rapidly progressing skin malignancy of melanocytic origin and represents the most lethal form of skin cancer. According to recent epidemiologic data, its global incidence continues to rise, with more than 320,000 new cases and approximately 57,000 deaths reported annually. Major risk factors include ultraviolet (UV) radiation exposure, fair skin phenotype, genetic predisposition, and the presence of dysplastic nevi. Despite significant therapeutic progress, advanced melanoma remains associated with high morbidity and mortality due to its strong metastatic potential and frequent development of therapeutic resistance [1,2,3].
The efficacy of approved small-molecule inhibitors in melanoma is primarily determined by the presence of actionable mutations—most notably BRAF V600 and activating KIT alterations—rather than by the tumor’s anatomical site alone. BRAF + MEK inhibitors demonstrate the greatest clinical benefit in BRAF V600–mutant cutaneous melanomas, achieving high response rates (~65–75%) and significant improvements in survival outcomes. Acral and mucosal melanomas, although less frequently harboring BRAF V600 mutations, can also respond to these inhibitors when such mutations are present. In contrast, KIT-mutant acral and mucosal melanomas derive modest but meaningful benefit from KIT inhibitors (e.g., imatinib, nilotinib), particularly when activating mutations occur in sensitive regions such as exon 11, though responses are typically less durable than those achieved with BRAF + MEK therapy. Uveal melanoma, characterized by GNAQ, GNA11, CYSLTR2, or PLCB4 mutations, remains largely refractory to both BRAF- and KIT-directed therapies and requires alternative, non-MAPK-targeted strategies. Overall, inhibitor performance in melanoma is driven by molecular genotype and influenced by subtype, with BRAF/MEK inhibitors offering the strongest benefit in BRAF V600–mutant cutaneous disease and KIT inhibitors providing a limited but targeted option for select non-cutaneous subtypes [4].
The molecular characterization of cutaneous melanoma has revealed key driver mutations that activate oncogenic signaling pathways, most notably the mitogen-activated protein kinase (MAPK) cascade. Approximately 40–60% of cutaneous melanomas harbor BRAF mutations—predominantly the V600E variant—which constitutively activates MAPK signaling, promoting tumor proliferation and survival [5,6,7,8,9,10,11]. This breakthrough enabled the development of small-molecule inhibitors that selectively target mutant BRAF, fundamentally reshaping the therapeutic landscape of melanoma. The advent of these agents marked a paradigm shift from nonspecific cytotoxic chemotherapy to mechanism-based, precision therapies guided by molecular profiling. Initially, clinical investigations centered on the selective inhibitors vemurafenib, trametinib, and imatinib; however, the therapeutic repertoire has since broadened considerably, with several additional small-molecule inhibitors now receiving regulatory approval for the treatment of melanoma.
Among these, the BRAF inhibitors—vemurafenib (Zelboraf), dabrafenib (Tafinlar), and encorafenib (Braftovi)—have shown remarkable efficacy in patients harboring BRAF V600E/K mutations, producing rapid tumor regression and high initial response rates [12,13,14]. However, these benefits are often short-lived, as acquired resistance commonly develops within months of treatment initiation. Preclinically, BRAF inhibitors (vemurafenib, dabrafenib, encorafenib) suppress ERK phosphorylation in BRAF V600-mutant models but differ in pharmacokinetics and paradoxical MAPK activation [15]. Clinically, dabrafenib and encorafenib have improved tolerability and lower cutaneous toxicity than vemurafenib [16].
Mechanisms of resistance include secondary mutations in NRAS or MEK1/2, BRAF amplification, alternative splicing, receptor tyrosine kinase (RTK) upregulation (e.g., epidermal growth factor receptor [EGFR] and platelet-derived growth factor receptor [PDGFR]), and compensatory activation of the phosphoinositide 3-kinase (PI3K)/AKT/mammalian target of the rapamycin (mTOR) pathway [17,18,19,20].
To counteract these resistance mechanisms, combination therapy with BRAF and MEK inhibitors has become the standard of care. Regimens such as dabrafenib plus trametinib and vemurafenib plus cobimetinib have demonstrated improvements in progression-free survival (PFS) and overall survival (OS), typically extending median PFS to 12–15 months while reducing paradoxical MAPK activation and adverse cutaneous events such as squamous cell carcinoma [21,22,23,24].
Although other signaling elements such as the KIT oncogene, the tumor microenvironment (TME), and immune checkpoint inhibitors (ICIs) play relevant roles in melanoma biology, their further discussion is presented in the subsequent section—“Cutaneous Melanoma Treatment and Mechanisms of Action”—to maintain focus in the introduction on the central role of BRAF-targeted therapy.
The introduction of small-molecule inhibitors has fundamentally shifted melanoma management from nonspecific cytotoxic chemotherapy toward mechanism-based, precision treatments guided by molecular profiling. Continued research is focused on understanding resistance mechanisms, optimizing combination regimens, and identifying predictive biomarkers to achieve more durable responses [25,26,27,28,29,30,31,32,33,34,35,36,37,38].
For the present synthesis, a narrative literature review was conducted using the PubMed and Scopus databases. The search strategy included the terms “vemurafenib,” “trametinib,” and “imatinib,” in combination with “targeted therapy” and “melanoma.” Peer-reviewed studies published between 2010 and 2025 were considered, with emphasis on clinical, translational, and mechanistic investigations. Articles were selected based on relevance, quality, and their contribution to understanding the therapeutic efficacy, mechanisms of action, and resistance patterns of these agents. Both preclinical and clinical studies were included when they provided mechanistic insights or informed therapeutic strategies. By applying these criteria, this review provides a balanced synthesis of current knowledge, highlighting both the clinical benefits and limitations of vemurafenib, trametinib, and imatinib in the management of cutaneous melanoma.

2. Cutaneous Melanoma Treatment and Mechanisms of Action

Targeted therapies have transformed the management of cutaneous melanoma by selectively inhibiting oncogenic signaling pathways. BRAF inhibitors, including vemurafenib, dabrafenib, and encorafenib, act by selectively binding the ATP-binding pocket of mutant BRAF (predominantly V600E). This suppresses constitutive MAPK pathway signaling, inhibits tumor proliferation, and induces apoptosis [11,12,13,14,15,16]. Clinical trials of vemurafenib demonstrated rapid tumor regression with overall response rates (ORR) of 50–60%, and improvements in progression-free survival (PFS) and overall survival (OS) in advanced BRAF-mutant melanoma [38,39,40]. Dabrafenib and encorafenib have shown similar efficacy, with encorafenib frequently combined with the MEK inhibitor binimetinib to enhance response and delay resistance [14,15,16].
The MEK inhibitors trametinib (Mekinist), cobimetinib (Cotellic), and binimetinib (Mektovi) thus play an essential role in combination strategies aimed at enhancing the durability of the response and mitigating toxicity. MEK inhibitors, such as trametinib and cobimetinib, act downstream of BRAF to inhibit MEK1/2, preventing ERK phosphorylation and cell proliferation [41,42,43]. While MEK inhibitor monotherapy provides modest benefits, combination therapy with BRAF inhibitors significantly improves PFS and OS. It also reduces paradoxical MAPK pathway activation, which lowers the incidence of BRAF inhibitor–associated cutaneous squamous cell carcinoma [44].
MEK inhibitors show modest single-agent activity but enhance responses when combined with BRAF inhibitors. Dabrafenib + trametinib, vemurafenib + cobimetinib, and encorafenib + binimetinib yield median overall survival >30 months. Real-world data support trial outcomes but are influenced by LDH levels, brain metastases, and performance status [15,16,45].
Despite these advances, resistance remains a major clinical challenge. Tumors can acquire secondary mutations in NRAS or MEK1/2, amplify BRAF, or undergo alternative splicing [46,47]. Upregulation of receptor tyrosine kinases (RTKs), including EGFR and PDGFR, further contributes to pathway reactivation [46,47,48]. Compensatory signaling through PI3K/AKT/mTOR also drives resistance [49,50,51]. In addition, non-genetic adaptations—such as tumor cell plasticity, epigenetic modifications, and TME interactions—allow melanoma cells to escape therapy and support survival under MAPK inhibition [51,52,53,54,55].
To address resistance, rational combination and sequencing strategies are being developed. Dual or triple inhibition of the MAPK pathway, incorporation of ERK inhibitors, and co-targeting of PI3K/AKT or CDK4/6 pathways have shown preclinical efficacy and early clinical promise [56,57,58]. MEK inhibitors combined with CDK4/6 inhibitors can suppress compensatory cell cycle progression, while PI3K/AKT pathway blockade addresses parallel survival signaling. Integration with ICIs is also under investigation. MAPK inhibitors modulate tumor immunogenicity, enhancing antigen presentation and T-cell infiltration, potentially improving the efficacy of ICIs [59,60,61,62]. Biomarker-guided sequencing of these combinations is critical to optimize patient outcomes and limit toxicity [63,64].
In addition to BRAF and MEK mutations, alterations in the KIT oncogene define a distinct molecular subset of melanoma, particularly mucosal and acral types. For these patients, KIT inhibitors such as imatinib (Gleevec) and nilotinib (Tasigna) offer clinically meaningful responses, underscoring the importance of genotype-directed therapy.
Melanoma patients benefit from the off-label use of KIT inhibitors, imatinib and nilotinib, only after stratification based on their KIT mutation status. KIT amplifications do not confer sensitivity to KIT inhibitors. Growing evidence suggest that depending on the melanoma subtype, KIT can act as an oncogene or a tumor suppressor [4].
KIT mutations, present in ~3–8% of melanomas, define a therapeutically relevant subset, especially in acral, mucosal, and chronically sun-damaged subtypes [23,65,66]. Clinical benefit is mutation-dependent: activating KIT point mutations (e.g., L576P, K642E) respond to imatinib or nilotinib, whereas KIT amplifications alone do not. Molecular testing is essential. KIT biology is context-dependent, acting as an oncogene in some subtypes and as a tumor suppressor in others, with implications for therapy selection [67].
KIT inhibitors provide therapeutic options for mucosal, acral, and chronically sun-damaged melanomas with activating KIT mutations in exons 11 and 13. KIT inhibitors show variable responses (16–30%) dependent on genotype, with nilotinib sometimes active post-imatinib [65,66]. Tyrosine kinase inhibitors (TKIs) such as imatinib, sunitinib, and nilotinib achieve objective response rates of 30–50%, though median PFS is often only ~4 months [63,64]. Response depends on specific KIT mutations rather than amplification alone, highlighting the need for comprehensive molecular profiling [68]. Resistance mechanisms include MAPK reactivation, PI3K-AKT pathway compensation, and phenotypic dedifferentiation. Emerging biomarkers, including ctDNA monitoring, may guide therapy sequencing [17,69]. Combination approaches with ICIs or other targeted agents are under active study to improve durability [29,30,70].
A comparison of approved BRAF, MEK, and KIT inhibitors used in the treatment of melanoma is presented in Table 1.
Adverse events vary by drug class. BRAF inhibitors commonly cause arthralgia, rash, fatigue, and photosensitivity [38,39,40]. MEK inhibitors are associated with diarrhea, hypertension, cardiomyopathy, and ocular toxicity [41,42,43,44]. KIT inhibitors frequently induce cytopenias, edema, and hepatotoxicity [63,64]. Early recognition and management of these adverse events are essential for maintaining therapy adherence and quality of life.
BRAF, MEK, and KIT inhibitors illustrate the promise of precision medicine in cutaneous melanoma. They improve ORR, PFS, and OS in molecularly stratified patients, yet resistance and incomplete durability of response remain challenges [31,32,33]. Ongoing research aims to refine combination regimens, develop next-generation inhibitors, and incorporate biomarker-driven strategies to overcome these barriers and optimize long-term outcomes.
In addition to BRAF, MEK, and KIT inhibitors, several other therapeutic modalities are integral to the current management of melanoma. ICIs targeting programmed death-1 (PD-1) (nivolumab, pembrolizumab) and cytotoxic T-lymphocyte–associated protein 4 (CTLA-4) (ipilimumab) constitute the cornerstone of first-line treatment for advanced, unresectable, or metastatic melanoma, providing durable responses and significant survival benefits compared to chemotherapy or monotherapy approaches. Combination ICI therapy with nivolumab and ipilimumab is also approved for selected patients, offering higher response rates but with increased immune-related toxicity [71,72,73].
Oncolytic viral therapy, particularly talimogene laherparepvec (T-VEC), is utilized for unresectable stage IIIB–IVM1a melanoma. T-VEC selectively replicates within tumor cells and expresses granulocyte–macrophage colony-stimulating factor (GM-CSF), enhancing systemic antitumor immunity. It may be administered alone or in combination with ICIs, with ongoing trials demonstrating synergistic activity [74,75].
Prior to the introduction of immunotherapy and targeted agents, cytotoxic chemotherapy (e.g., dacarbazine, temozolomide) was the standard of care for advanced melanoma; however, due to limited efficacy and high toxicity, it is now reserved for refractory or palliative cases [76]. Radiotherapy remains important in the adjuvant or palliative setting, particularly for brain or soft tissue metastases, and may enhance immune activation when combined with checkpoint inhibitors [77].
Surgical resection remains the mainstay of early-stage melanoma management (stages I–II and some stage III), aiming for curative intent. For resectable stage III–IV or high-risk melanoma, adjuvant or neoadjuvant immunotherapy (nivolumab, pembrolizumab) or targeted therapy (dabrafenib plus trametinib in BRAF-mutant cases) are now recommended to reduce recurrence risk and improve long-term survival [78].
Within this therapeutic landscape, BRAF, MEK, and KIT inhibitors are primarily used in patients with BRAF V600– or KIT-mutated melanoma, either as first-line targeted approaches or following immunotherapy failure, depending on molecular profile, tumor burden, and clinical course. These treatment strategies collectively illustrate the evolution of melanoma therapy, summarized in Table 2, which provides an overview of melanoma treatment strategies, classified according to therapeutic modality and summarizes the most severe and frequent adverse events, including their severity.

3. Vemurafenib—BRAF Inhibitor

Vemurafenib is a small-molecule, ATP-competitive kinase inhibitor developed through fragment-based drug discovery and designed to selectively target BRAFV600E/K mutations, which occur in approximately 40–60% of cutaneous melanomas [79,80]. These gain-of-function mutations cause constitutive activation of the mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) signaling pathway, independent of upstream RAS activity, thereby driving uncontrolled proliferation, enhanced survival, and resistance to apoptosis.
Figure 1 illustrates the chemical structure of vemurafenib.
The therapeutic effect of vemurafenib arises from its selective binding to mutant BRAF monomers, inhibiting downstream phosphorylation of MEK and ERK. This suppression of oncogenic MAPK signaling induces G1-phase cell cycle arrest, promotes apoptosis, and leads to marked tumor regression in melanomas harboring BRAFV600E/K alterations [81,82,83,84].
Figure 2 illustrates the mechanism of action of vemurafenib.
Preclinical studies, employing both in vitro cellular assays and in vivo xenograft models, demonstrated that vemurafenib exhibits high specificity and potent antitumor activity against melanoma cells carrying the BRAFV600E mutation. These findings provided strong mechanistic and pharmacological rationale for its rapid progression into clinical development. Early-phase clinical trials, initiated in 2008, confirmed the preclinical evidence, showing substantial tumor regression and ORRs ranging from 50% to 80% in patients with advanced BRAF-mutant melanoma. Further validation came from the pivotal BRIM-3 randomized, multicenter phase III trial, which compared vemurafenib with dacarbazine in previously untreated patients with metastatic BRAF-mutant melanoma. The trial was terminated early following the demonstration of a significant survival benefit in the vemurafenib group. Specifically, vemurafenib reduced the risk of death by 63% compared to dacarbazine (hazard ratio [HR] = 0.37; p < 0.0001) and significantly improved both PFS and OS. These results led to its accelerated approval by the U.S. Food and Drug Administration (FDA) in 2011 and the European Medicines Agency (EMA) in 2012 [86,87].
Beyond melanoma, the clinical application of vemurafenib expanded in 2017 with its approval for Erdheim–Chester disease (ECD), a rare non-Langerhans cell histiocytosis characterized by BRAFV600E mutations. In this setting, vemurafenib has produced durable clinical responses, radiographic tumor regression, and substantial symptom relief, highlighting the oncogenic relevance of BRAFV600E across diverse malignancies [88,89].
Table 3 summarizes the distribution and clinical significance of RAF mutations across different cancer types.
In routine clinical practice, vemurafenib has significantly reshaped treatment expectations for patients with advanced BRAF-mutant melanoma. Compared with dacarbazine, it doubled median PFS from 1.6 to approximately 5.3 months and achieved ORRs of 50–60%, often inducing rapid and profound tumor regression [91,92,93]. However, the durability of these responses is limited, with most patients experiencing disease progression within six to eight months.
Mechanisms of resistance to vemurafenib are diverse and multifactorial. Reactivation of the MAPK pathway is the most frequent, often mediated by secondary NRAS mutations that promote RAF dimerization, bypassing the monomer-selective inhibition of vemurafenib. Alternative BRAF splicing, particularly the p61BRAF isoform, facilitates constitutive dimerization and drug evasion, while BRAF amplification can elevate protein levels beyond the inhibitory threshold. Upregulation of RTKs such as PDGFRβ, IGF-1R, and EGFR, and activation of compensatory survival signaling via the PI3K–AKT pathway—particularly in the context of PTEN loss—further contribute to resistance. The tumor microenvironment also plays a pivotal role; stromal fibroblasts and tumor-associated macrophages can secrete hepatocyte growth factor, activating c-MET signaling and reinforcing resistance phenotypes [94,95,96,97].
Vemurafenib is generally well tolerated, with common adverse events including arthralgia, rash, photosensitivity, and fatigue, which are typically reversible with supportive care or dose adjustment. More serious but less frequent toxicities include hepatotoxicity, QT interval prolongation, and bradyarrhythmias, which warrant biochemical and electrocardiographic monitoring. Additional adverse events include gastrointestinal disturbances, musculoskeletal pain, secondary cutaneous neoplasms with an incidence of approximately 12%, and rare complications such as uveitis and cytochrome P450–mediated drug interactions [98,99,100]. Toxic epidermal necrolysis has been reported in combination with cobimetinib [101]. A clinically significant adverse event is the development of cutaneous squamous cell carcinomas and keratoacanthomas, observed in 20–30% of treated patients. These low-grade lesions, usually managed by surgical excision, arise from paradoxical MAPK activation in RAS-mutant, BRAF–wild-type keratinocytes, representing an on-target, off-tumor effect of vemurafenib [102,103].
To overcome resistance and enhance response durability, dual inhibition of the MAPK pathway with vemurafenib and MEK inhibitors such as cobimetinib or trametinib has demonstrated superior outcomes. These regimens prolong median PFS to approximately 12 months, compared with around seven months for monotherapy, and reduce paradoxical cutaneous toxicities by suppressing MEK-driven signaling in non-malignant cells [22,104]. Building on this, triplet regimens incorporating BRAF and MEK inhibitors with ICIs are under investigation. The IMspire150 trial showed that the addition of atezolizumab (anti–PD-L1) to vemurafenib and cobimetinib significantly improved PFS and overall survival in advanced BRAF-mutant melanoma, supporting the hypothesis that targeted therapies can enhance neoantigen release and improve immune checkpoint blockade efficacy [105,106,107,108,109].
Emerging strategies aim to delay or overcome resistance through inhibitors of the PI3K, AKT, and mTOR pathways to block compensatory survival signals, histone deacetylase inhibitors to reverse transcriptional reprogramming, and HSP90 inhibitors to degrade mutant BRAF and other oncogenic proteins. Immunomodulatory agents are being explored to remodel the tumor microenvironment, enhance antigen presentation, and facilitate T-cell infiltration. Additional targets include integrins and TGF-β signaling components such as ITGA5, ITGB3, PAI1, and p21, which may provide new avenues for overcoming resistance and improving therapeutic outcomes [110,111,112,113,114].
Future directions focus on optimizing vemurafenib therapy through intermittent dosing to reduce selective pressure and toxicity, advanced drug delivery systems such as nanoparticles and liposomes to improve pharmacokinetics and minimize off-target effects, and real-time biomarker monitoring using circulating tumor DNA, phosphoproteomics, and single-cell RNA sequencing to enable adaptive treatment strategies [115,116,117]. Vemurafenib has redefined the therapeutic landscape for BRAF-mutant melanoma and exemplifies the clinical potential of mutation-targeted drug development. Its success has catalyzed the adoption of molecularly guided therapeutic strategies across oncology. Continued innovation through rational drug combinations, integration of dynamic biomarkers, and development of next-generation therapeutic platforms holds promise for achieving durable disease control, reducing toxicity, and moving closer to long-term remission in this historically aggressive malignancy [118,119,120,121,122,123,124,125,126,127,128,129,130,131].

4. Trametinib—MEK Inhibitor

Trametinib is a selective, reversible, allosteric inhibitor of mitogen-activated protein kinase kinases 1 and 2 (MEK1/2), key downstream components of the RAS–RAF–MEK–ERK signaling cascade, also known as the mitogen-activated protein kinase (MAPK) pathway. Dysregulation of this pathway is a hallmark of multiple malignancies, with particular pathogenic importance in cutaneous melanoma. The MAPK cascade regulates critical cellular processes such as proliferation, differentiation, survival, and migration. In melanoma, activating BRAF mutations—most notably BRAFV600E and BRAFV600K—drive constitutive MEK activation and sustained ERK phosphorylation, leading to aberrant oncogenic signaling independent of upstream receptor stimulation or external mitogenic cues [132,133].
Figure 3 illustrates the chemical structure of trametinib.
Mechanistically, trametinib binds to MEK1/2 in a non-competitive, allosteric manner adjacent to the ATP-binding site, stabilizing the kinase in its inactive conformation and preventing phosphorylation by upstream RAF kinases. This inhibition results in potent suppression of ERK phosphorylation (pERK), downregulation of proliferation- and survival-associated gene expression, G1-phase cell cycle arrest, and induction of apoptosis in sensitive tumor cells [134,135]. The mechanism is particularly relevant in BRAFV600-mutant cancers, where persistent MAPK pathway activation sustains oncogenic growth.
Figure 4 illustrates the mechanism of action of trametinib.
Trametinib became the first MEK inhibitor to achieve regulatory approval, marking a significant milestone in targeted oncology. In May 2013, the U.S. Food and Drug Administration (FDA) approved trametinib monotherapy for patients with unresectable or metastatic melanoma harboring BRAFV600E or BRAFV600K mutations, based on phase III trials demonstrating improved PFS and OS compared with conventional chemotherapy such as dacarbazine [94,137,138]. Unlike BRAF inhibitors, trametinib does not cause paradoxical MAPK activation in RAS-mutant keratinocytes, reducing the risk of secondary cutaneous neoplasms.
Despite its initial clinical efficacy, resistance to trametinib monotherapy frequently develops within four to six months. The mechanisms underlying resistance include activation of compensatory survival pathways such as PI3K/AKT/mTOR, secondary MEK1 mutations (e.g., MEK1P124L/Q) that reduce drug binding affinity, and upregulation of RTKs including PDGFRβ and IGF-1R, which reactivate MAPK signaling through bypass mechanisms [94,139].
To overcome resistance and improve treatment outcomes, trametinib is often combined with the BRAF inhibitor dabrafenib. This dual blockade of the MAPK pathway provides synergistic inhibition, limits feedback reactivation, and delays the emergence of resistance. The pivotal COMBI-d and COMBI-v phase III trials demonstrated significantly improved outcomes with the combination compared with dabrafenib monotherapy, including median PFS of 11.0 versus 8.8 months, overall survival of 25.1 versus 18.7 months, and objective response rates of 67% versus 51% [140,141,142,143,144,145,146,147,148,149,150,151]. Subsequent studies confirmed these findings across diverse populations, including Chinese patients with resected stage III disease [152] and pediatric and adolescent cohorts [153]. These data established the dabrafenib–trametinib combination as the standard of care for advanced BRAF-mutant melanoma and stimulated further exploration of dual MAPK inhibition in other malignancies [154,155,156,157].
The safety profile of trametinib reflects its mechanism of MEK inhibition in non-malignant tissues. Common adverse events include acneiform dermatitis, diarrhea, peripheral edema, and hypertension, consistent with effects on epithelial and vascular signaling [158,159,160]. More serious but less frequent toxicities include cardiomyopathy, manifesting as decreased left ventricular ejection fraction, and ocular events such as central serous retinopathy and retinal vein occlusion, necessitating cardiovascular and ophthalmologic monitoring [161,162]. Importantly, trametinib does not induce cutaneous squamous cell carcinomas—a toxicity observed with BRAF inhibitors—owing to its upstream inhibition that prevents paradoxical ERK activation in RAS-mutant keratinocytes [41].
In efforts to enhance therapeutic durability and immune engagement, rational triplet regimens have been developed. Clinical trials combining dabrafenib, trametinib, and ICIs—including spartalizumab (anti–PD-1) and atezolizumab (anti–PD-L1)—have shown encouraging results. Proposed mechanisms of synergy involve increased T-cell infiltration, suppression of regulatory T cells and myeloid-derived suppressor cells, and improved antigen presentation within the tumor microenvironment [163,164,165]. The IMspire150 phase III trial, using a similar triplet of vemurafenib, cobimetinib, and atezolizumab, further supports the therapeutic potential of MEK inhibitor–based triplet combinations [166].
Beyond melanoma, trametinib has demonstrated clinical efficacy across multiple BRAFV600-mutant malignancies, including non-small cell lung cancer, anaplastic thyroid carcinoma, and histiocytic disorders such as Erdheim–Chester disease and Langerhans cell histiocytosis [167,168]. Ongoing research is exploring its combination with inhibitors of compensatory signaling pathways, including PI3K, HDAC, and HSP90 inhibitors, to overcome resistance and broaden its therapeutic scope. Basket trials are also assessing trametinib in genetically defined malignancies harboring MAPK pathway alterations [169,170,171,172,173,174,175,176,177].
Preclinical and translational research has highlighted trametinib’s immunomodulatory properties. MEK inhibition can remodel the tumor microenvironment by suppressing pro-tumorigenic cytokine production, enhancing cytotoxic T-cell function, and reducing immunosuppressive cell populations. These immune-modifying effects may potentiate responses to immune checkpoint blockade, positioning trametinib as a potential immune-sensitizing agent in refractory cancers [178,179,180]. Clinical trials are underway to evaluate its integration into combination regimens for checkpoint inhibitor–resistant malignancies [15,181,182,183].
Overall, trametinib represents a cornerstone of targeted therapy for MAPK-driven cancers, particularly in combination with dabrafenib for BRAF-mutant melanoma. Its incorporation into multidrug regimens exemplifies a mechanism-based therapeutic strategy designed to enhance treatment depth, delay resistance, and improve immune responsiveness. As precision oncology continues to evolve toward more personalized and durable treatment paradigms, trametinib remains a key agent in the expanding landscape of cancer therapeutics.

5. Imatinib—C-KIT Inhibitor

Imatinib mesylate is a first-in-class, small-molecule TKI that selectively targets the ATP-binding sites of several oncogenic kinases, most notably BCR-ABL, c-KIT (CD117), and platelet-derived growth factor receptors (PDGFR) α and β. These kinases are critical regulators of intracellular signaling pathways controlling cellular proliferation, differentiation, survival, and apoptosis [184,185,186].
Figure 5 illustrates the molecular structure of imatinib.
Mechanistically, imatinib binds competitively to the ATP-binding pocket of these kinases in their inactive conformation, preventing autophosphorylation and subsequent phosphorylation of downstream substrates. This blockade effectively disrupts aberrant signal transduction pathways responsible for malignant transformation and uncontrolled cellular growth. The high target selectivity conferred by this mechanism minimizes off-target interactions commonly associated with conventional cytotoxic chemotherapies, thereby contributing to its favorable therapeutic index [187,188,189].
The development of imatinib mesylate marked a paradigm shift in oncology, originating from its ability to selectively inhibit the constitutively active BCR-ABL fusion protein—an aberrant tyrosine kinase generated by the t(9;22)(q34;q11) translocation known as the Philadelphia chromosome. This cytogenetic abnormality defines chronic myeloid leukemia (CML) and drives unchecked proliferation and impaired apoptosis in hematopoietic progenitor cells [190,191,192]. Preclinical studies demonstrated that imatinib induces apoptosis and G1-phase cell cycle arrest in BCR-ABL–expressing cells, providing strong rationale for clinical translation [184]. Following these findings, early 1990s research led by Druker and colleagues developed the first potent and specific BCR-ABL inhibitor, rapidly advancing it into clinical trials. Early-phase studies showed unprecedented hematologic and cytogenetic responses in interferon-refractory CML, leading to accelerated approval in 2001 and ushering in the modern era of precision oncology [193,194,195].
Approved in 2001, imatinib revolutionized the treatment of CML, transforming a once-fatal disease into a chronic, manageable condition characterized by dramatic improvements in progression-free and overall survival. Beyond CML, imatinib demonstrated substantial clinical efficacy in gastrointestinal stromal tumors (GIST), which frequently harbor activating mutations in c-KIT or PDGFRA that lead to ligand-independent receptor activation and oncogenic signaling. Imatinib effectively inhibits these mutant kinases, resulting in tumor regression and prolonged survival in patients with advanced or metastatic GIST [196,197,198,199]. These successes established imatinib as a prototype molecularly targeted agent and validated the strategy of exploiting discrete oncogenic dependencies across tumor types [200,201,202,203].
Imatinib is FDA-approved for a broad range of malignancies driven by dysregulated kinase activity, including Philadelphia chromosome–positive CML and acute lymphoblastic leukemia (ALL), KIT-mutant GIST, myelodysplastic/myeloproliferative disorders with PDGFR rearrangements, dermatofibrosarcoma protuberans, hypereosinophilic syndrome (HES), chronic eosinophilic leukemia, and aggressive systemic mastocytosis lacking the KIT D816V mutation. Off-label uses include chordomas, desmoid tumors, advanced KIT-mutant melanoma, and post–allogeneic transplant relapse in CML [204,205,206,207].
In melanoma, imatinib has shown modest but clinically meaningful efficacy in specific biologically distinct subtypes characterized by KIT mutations or amplifications, including acral, mucosal, and chronically sun-damaged melanomas [208,209,210]. While the overall response rates are lower than in CML or GIST, partial responses and durable disease stabilization have been observed in selected patients, underscoring the importance of molecular stratification [26,65]. Clinical studies specifically evaluating KIT-mutant acral, mucosal, uveal, and sun-damaged melanomas have reported heterogeneous yet encouraging results [211,212,213,214,215,216].
Despite its transformative success, resistance to imatinib frequently emerges, particularly in advanced stages of disease. Mechanisms of resistance include point mutations within the BCR-ABL kinase domain—most notably the T315I mutation—gene amplification, and activation of alternative survival pathways [217,218,219,220]. To overcome these limitations, second- and third-generation TKIs such as dasatinib, nilotinib, bosutinib, and ponatinib were developed, providing enhanced potency and broader activity against resistant BCR-ABL variants [221,222,223,224].
Imatinib exhibits a favorable safety profile that supports long-term use. The most common adverse events are mild and include fluid retention, nausea, myalgia, fatigue, and low-grade myelosuppression [225,226]. Severe but rare toxicities include hepatotoxicity, cardiotoxicity, and prolonged cytopenias, necessitating regular monitoring [226,227]. The overall tolerability of imatinib contributes to its sustained use in chronic disease management such as CML [228].
Emerging research continues to expand the therapeutic applications of imatinib through advanced molecular profiling, identifying novel tumors with imatinib-sensitive kinase alterations. In melanoma, early intervention in KIT-mutant acral or mucosal subtypes appears to yield superior outcomes [26,229]. Combination strategies incorporating imatinib with immune checkpoint inhibitors or other targeted agents are actively being investigated to suppress compensatory pathways, overcome resistance, and enhance therapeutic durability [230,231,232,233,234,235,236,237,238,239]. Parallel developments include next-generation TKIs, allosteric inhibitors, and targeted protein degraders designed to inhibit or eliminate oncogenic kinases more effectively [218,240,241].
Imatinib’s discovery and clinical success remain a landmark in oncology, exemplifying the transformative impact of rational drug design and molecularly guided therapy on patient outcomes [242,243,244]. In melanoma, it continues to serve as a valuable therapeutic option for patients with KIT mutations or amplifications, with ongoing research focused on refining patient selection, optimizing combination regimens, and integrating imatinib into evolving immunotherapy and precision oncology frameworks [55,65].
Figure 6 illustrates molecular mechanism underlying imatinib activity.

6. Comparative Analysis of Vemurafenib, Trametinib, and Imatinib in Cutaneous Melanoma

Targeted therapies have markedly improved outcomes for patients with BRAF- or KIT-mutant cutaneous melanoma, yet differences in mechanism, efficacy, and toxicity necessitate a comparative evaluation of vemurafenib, trametinib, and imatinib. Vemurafenib is a small-molecule, ATP-competitive inhibitor that selectively targets BRAFV600E/K mutations, present in 40–60% of cutaneous melanomas, thereby blocking constitutive activation of the MAPK/ERK pathway and inducing G1-phase arrest, apoptosis, and tumor regression [65,66,67,68,69,70]. Preclinical studies demonstrated potent, mutation-specific antitumor activity, providing the rationale for rapid clinical development. Early-phase trials initiated in 2008 showed ORRs of 50–80% in advanced BRAF-mutant melanoma. The pivotal BRIM-3 phase III trial confirmed a median PFS of 5.3 months versus 1.6 months with dacarbazine and a median OS of 13.6 months versus 9.7 months, leading to FDA approval in 2011 and EMA approval in 2012 [86,87]. Combination therapy with MEK inhibitors such as trametinib or cobimetinib further improved outcomes, extending median PFS to approximately 12 months and increasing ORRs to ~70% while reducing paradoxical MAPK-driven toxicities [22,104]. Triplet regimens including ICIs, such as atezolizumab, are under investigation and show promise for enhancing neoantigen-mediated immune responses [106,107,108,109]. Common adverse events for vemurafenib include arthralgia, rash, photosensitivity, fatigue, gastrointestinal disturbances, hepatotoxicity, and QT prolongation, with more serious but less frequent events including cutaneous squamous cell carcinoma or keratoacanthomas (~20–30%) [97,98,99,100,101,102]. Resistance mechanisms are multifactorial and include MAPK reactivation via secondary NRAS or MEK mutations, BRAF amplification or alternative splicing, upregulation of RTKs (PDGFRβ, IGF-1R, EGFR), compensatory PI3K–AKT signaling, and tumor microenvironment interactions [110,111,112,113,114].
Trametinib is a selective MEK1/2 inhibitor acting downstream of BRAF to prevent ERK phosphorylation and inhibit cellular proliferation. Monotherapy trials reported ORRs of 20–25%, median PFS of 4–5 months, and median OS of approximately 11 months [54,55,56]. However, when combined with BRAF inhibitors, trametinib significantly improves ORRs to 60–70%, median PFS to 11–12 months, and OS up to 25 months, demonstrating superior durability of response compared with monotherapy [22,104]. Adverse events include rash, diarrhea, fatigue, peripheral edema, hypertension, cardiomyopathy, and retinopathy, with rare cases of interstitial lung disease [41,42,43]. Resistance to trametinib commonly arises through MEK or ERK mutations, receptor tyrosine kinase upregulation, or bypass signaling, and current investigational strategies include ERK inhibitors, PI3K/AKT pathway antagonists, CDK4/6 inhibitors, and combination with ICIs to enhance anti-tumor immunity [110,111,112,113,114].
Imatinib, a tyrosine kinase inhibitor targeting KIT mutations, is primarily used in acral, mucosal, or chronically sun-damaged melanomas harboring activating KIT alterations, particularly in exons 11 and 13. Preclinical studies demonstrated potent inhibition of KIT-mutant melanoma cells. Phase II clinical trials showed ORRs of 30–50%, median PFS of approximately 3.5–4 months, and median OS around 12 months [25,26,27,28]. Responses are largely limited to patients with specific KIT mutations, with resistance frequently developing within months. Adverse effects are generally mild to moderate, including edema, nausea, diarrhea, fatigue, and rash, while severe toxicities such as myelosuppression, hepatotoxicity, and cardiotoxicity have been reported [63,64]. Combination strategies with ICIs or chemotherapy are under investigation to enhance efficacy and overcome resistance [29,30].
Comparing these therapies, BRAF-targeted combination therapy (vemurafenib plus trametinib) demonstrates the highest clinical efficacy, with OS up to 25 months, PFS approximately 12 months, and ORRs of 60–70%. Vemurafenib monotherapy offers moderate efficacy with OS of ~13.6 months, PFS of ~5.3 months, and ORRs of 50–60%, while imatinib shows intermediate ORRs (30–50%) and lower durability, primarily in KIT-mutant melanoma. Resistance remains a challenge across all agents, driven by genetic and non-genetic mechanisms, necessitating combination therapy, careful monitoring, and biomarker-guided patient selection. Toxicity profiles differ: vemurafenib is associated with cutaneous events and photosensitivity, trametinib with rash and cardiotoxicity, and imatinib with fluid retention and cytopenias. Integration of ICIs with BRAF/MEK inhibitors is emerging as a strategy to enhance durability and achieve long-term disease control [105,106,107,108,109].
In conclusion, vemurafenib/trametinib combination therapy provides the most effective treatment for BRAF-mutant melanoma, while vemurafenib monotherapy and imatinib remain important options for selected patient subsets. Future research focusing on resistance mechanisms, rational combination strategies, and biomarker-driven personalization is expected to further improve outcomes and advance the precision management of cutaneous melanoma.
Table 4 presents a comparative analysis of the mechanisms of action of vemurafenib, trametinib, and imatinib, along with their combination strategies and associated clinical biomarkers and Table 5 illustrates a comparison of adverse effects of vemurafenib, trametinib, and imatinib.

7. Future Directions and Clinical Relevance

The therapeutic landscape of cutaneous melanoma has been revolutionized by targeted agents such as vemurafenib, trametinib, and imatinib, which have substantially improved outcomes for patients with advanced disease. Despite these advances, significant clinical challenges persist, including the emergence of drug resistance, limited durability of responses, intra- and inter-tumoral heterogeneity, and the need for individualized treatment strategies. Consequently, current research focuses on refining therapeutic approaches, overcoming resistance mechanisms, and integrating targeted therapies with complementary treatment modalities to achieve durable disease control and improved long-term survival.
One of the most pressing challenges in targeted melanoma therapy is the development of resistance to BRAF, MEK, and KIT inhibitors. Resistance arises through various mechanisms, including secondary mutations, activation of compensatory signaling pathways such as PI3K/AKT/mTOR, or phenotypic switching toward a mesenchymal-like state that promotes therapeutic escape [247]. For example, reactivation of the MAPK pathway downstream of BRAF via MEK or ERK mutations has been documented in patients progressing on BRAF/MEK inhibitors, underscoring the adaptive plasticity of melanoma cells [235]. Novel approaches to circumvent resistance are under investigation, including dual or triple blockade of the MAPK pathway, co-targeting of parallel signaling networks, and inhibition of RTKs that drive adaptive survival responses [248,249]. These strategies aim to suppress compensatory mechanisms, extend response duration, and prevent or delay resistance development.
Combination therapy is another critical area of ongoing investigation. The rationale for combining targeted therapies with immunotherapies stems from their complementary mechanisms of action: targeted agents induce rapid tumor regression and modulate the tumor microenvironment, while immune checkpoint inhibitors (ICIs) provide durable immune surveillance and control. Clinical trials combining BRAF/MEK inhibitors with PD-1 or PD-L1 blockade have shown improved progression-free survival and durable responses compared with monotherapies [250,251]. Innovative strategies integrating targeted therapy with adoptive T-cell transfer, oncolytic virus therapy, or personalized cancer vaccines are also being explored, aiming to enhance antitumor immunity and achieve more robust clinical outcomes [252,253]. These multimodal approaches represent a paradigm shift in melanoma management, moving from sequential monotherapies to integrated treatment models that exploit synergistic mechanisms.
Precision oncology is increasingly guiding clinical decision-making, enabling individualized therapy selection based on molecular profiling. Advances in next-generation sequencing (NGS), liquid biopsy technologies, and circulating tumor DNA (ctDNA) analysis now allow for real-time monitoring of tumor evolution and treatment response [254]. These tools facilitate early detection of resistance-associated mutations, inform dynamic treatment adjustments, and enable adaptive therapy before clinical progression [255]. Moreover, multi-omics integration—including transcriptomic and proteomic profiling—is expected to uncover predictive biomarkers of therapeutic response and resistance, thereby refining patient stratification and optimizing treatment selection [256].
The development of next-generation inhibitors remains a key focus of translational research. Novel pan-RAF inhibitors, ERK inhibitors, and allosteric MEK inhibitors are being investigated to overcome resistance to first-generation agents and broaden activity against resistant clones [257]. Similarly, new KIT inhibitors with improved potency and selectivity are under evaluation for KIT-mutant melanoma, a rare but challenging subtype with limited treatment options [258,259]. These compounds may become important components of rational combination regimens or serve as salvage therapies following disease progression on standard treatments.
As the therapeutic landscape evolves, real-world application of these strategies will require careful consideration of toxicity management, treatment sequencing, and cost-effectiveness. The integration of molecular diagnostics into routine clinical workflows demands robust bioinformatics infrastructure and multidisciplinary collaboration among oncologists, pathologists, geneticists, and data scientists. Ensuring equitable access to advanced diagnostics and therapies remains a critical global priority, particularly in resource-limited healthcare systems [260].
The next decade of melanoma therapy is poised to witness the convergence of targeted therapy, immunotherapy, and precision medicine, ushering in a new era of personalized oncologic care. The future relevance of targeted agents such as vemurafenib, trametinib, and imatinib will likely be defined not as standalone treatments but as integral components of multifaceted, patient-specific strategies. Liquid biopsy-guided adaptive therapy, multi-omics-driven biomarker discovery, and the integration of artificial intelligence for predictive modeling are expected to further refine treatment selection and monitoring [254,256]. Emerging approaches—including personalized neoantigen vaccines, adoptive T-cell therapies, and epigenetic modulators—offer additional avenues to enhance the efficacy of targeted interventions [252,255]. As these innovations mature, the overarching goal will shift from merely extending survival to achieving durable remission and, ultimately, functional cures in select patient populations.
Collectively, these developments underscore a rapidly evolving therapeutic landscape in cutaneous melanoma—one defined by innovation, integration, and personalization. The continued refinement of targeted therapies, in conjunction with advances in molecular diagnostics and immunotherapy, promises to extend the boundaries of clinical benefit. As the field moves toward adaptive, data-driven treatment paradigms, the emphasis is shifting from short-term tumor control to sustained remission and improved quality of life. The following conclusion synthesizes the key insights from this review and highlights their clinical relevance, emphasizing how ongoing translational progress is reshaping both current practice and future research directions in melanoma management.
Table 6 summarizes novel melanoma therapies, classified based on their mechanisms of action and therapeutic strategies.

8. Conclusions

The advent of targeted therapies such as vemurafenib, trametinib, and imatinib has transformed the management of cutaneous melanoma, enabling precision treatment guided by molecular profiling. These agents exemplify the success of genotype-driven therapy, demonstrating substantial improvements in survival and quality of life for patients with BRAF or c-KIT mutations. However, their long-term efficacy remains constrained by acquired resistance and cumulative toxicity, emphasizing the need for ongoing molecular and clinical investigation.
Future research should prioritize elucidating resistance mechanisms, refining predictive biomarkers, and developing next-generation inhibitors with improved selectivity and safety. Integrating targeted therapies with immune checkpoint inhibitors and other novel modalities holds promise for enhancing the durability of the response and overcoming therapeutic escape.
Sustained collaboration across translational research, drug development, and clinical practice is essential to extend treatment durability and achieve more personalized, durable outcomes for patients with cutaneous melanoma.

Author Contributions

Conceptualization, T.B. and P.K.; writing—original draft preparation, P.K.; writing—review and editing, P.K. and T.B.; visualization P.K.; supervision, T.B. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
ADPAdenosine Diphosphate
AEsAdverse Events
AKTProtein Kinase B
ALLAcute Lymphoblastic Leukemia
ARAndrogen Receptor
ATPAdenosine Triphosphate
A-RAFA-Raf Proto-Oncogene, Serine/Threonine Kinase
B-RAFB-Raf Proto-Oncogene, Serine/Threonine Kinase
BCR-ABLBreakpoint Cluster Region-Abelson Fusion Gene
BRAFv-Raf Murine Sarcoma Viral Oncogene Homolog B1
C-RAFC-Raf Proto-Oncogene, Serine/Threonine Kinase
CAR-TChimeric Antigen Receptor T-cell
CCND1Cyclin D1
CDK4/6Cyclin-Dependent Kinases 4 and 6
CMLChronic Myeloid Leukemia
CTLA-4Cytotoxic T-Lymphocyte–Associated Protein 4
DFSPDermatofibrosarcoma Protuberans
ECGElectrocardiogram
ERKExtracellular Signal-Regulated Kinase
FISHFluorescence In Situ Hybridization
GAPGTPase-Activating Protein
GDPGuanosine Diphosphate
GEFGuanine Nucleotide Exchange Factor
GISTGastrointestinal Stromal Tumor
GM-CSFGranulocyte-Macrophage Colony-Stimulating Factor
GTPGuanosine Triphosphate
HCQHydroxychloroquine
HESHypereosinophilic Syndrome
HLAHuman Leukocyte Antigen
HSV-1Herpes Simplex Virus Type 1
ICIImmune Checkpoint Inhibitor
IFN-γInterferon Gamma
ILDInterstitial lung disease
JAKJanus Kinase
JAK/STATJanus Kinase/Signal Transducer and Activator of Transcription
KITStem Cell Factor Receptor/Receptor Tyrosine Kinase
LAG-3Lymphocyte-Activation Gene 3
LDHLactate Dehydrogenase
MAP2K1/2Mitogen-Activated Protein Kinase Kinase 1 and 2
MAPKMitogen-Activated Protein Kinase
MEKMitogen-Activated Protein Kinase Kinase
MHCMajor Histocompatibility Complex
mTORMechanistic Target of Rapamycin
NGSNext-Generation Sequencing
NK cellNatural Killer Cell
NRASNeuroblastoma RAS Viral Oncogene Homolog
ORRObjective response rate
OSOverall survival
PPhosphate
PCRPolymerase Chain Reaction
PD-1Programmed Cell Death Protein 1
PDGFPlatelet-Derived Growth Factor
PDGFRAPlatelet-Derived Growth Factor Receptor Alpha
PDGFRPlatelet-Derived Growth Factor Receptor
PD-L1Programmed Death-Ligand 1
PFSProgression-free survival
Ph+Philadelphia Chromosome–Positive
PI3KPhosphoinositide 3-Kinase
QTQT Interval
RAFRapidly Accelerated Fibrosarcoma
RASRat Sarcoma Virus Oncogene
RTRadiotherapy
RTKReceptor Tyrosine Kinase
SCCSquamous cell carcinoma.
SCFStem Cell Factor
SLNBSentinel Lymph Node Biopsy
STATSignal Transducer and Activator of Transcription
TERTTelomerase Reverse Transcriptase
TILTumor-Infiltrating Lymphocyte
TKITyrosine Kinase Inhibitor
TMBTumor Mutational Burden
TregRegulatory T Cell
TYP1Tyrosinase-Related Protein 1
V600E/KValine to Glutamic Acid/Lysine Substitution at Position 600
WTWild Type

References

  1. Caraban, B.M.; Aschie, M.; Deacu, M.; Cozaru, G.C.; Pundiche, M.B.; Orasanu, C.I.; Voda, R.I. A Narrative Review of Current Knowledge on Cutaneous Melanoma. Clin. Pract. 2024, 14, 214–241. [Google Scholar] [CrossRef] [PubMed]
  2. Guo, W.; Wang, H.; Li, C. Signal Pathways of Melanoma and Targeted Therapy. Signal Transduct. Target. Ther. 2021, 6, 424. [Google Scholar] [CrossRef]
  3. Alsadiq, S.; Kartolo, A.; McWhirter, E.; Hopman, W.; Baetz, T. Efficacy and Safety of Adjuvant Systemic Therapies in Trial Non-Eligible Resected Stages III and IV Melanoma Patients. Melanoma Manag. 2025, 12, 2461963. [Google Scholar] [CrossRef]
  4. Eddy, K.; Chen, S. Overcoming Immune Evasion in Melanoma. Int. J. Mol. Sci. 2020, 21, 8984. [Google Scholar] [CrossRef]
  5. Tawbi, H.; Nimmagadda, N. Targeted Therapy in Melanoma. Biologics 2009, 3, 475–484. [Google Scholar] [CrossRef] [PubMed]
  6. Cerchia, C.; Lavecchia, A. Small Molecule Drugs and Targeted Therapy for Melanoma: Current Strategies and Future Directions. Curr. Med. Chem. 2017, 24, 2312–2344. [Google Scholar] [CrossRef]
  7. Kwong, A.; Sanlorenzo, M.; Rappersberger, K.; Vujic, I. Update on Advanced Melanoma Treatments: Small Molecule Targeted Therapy, Immunotherapy, and Future Combination Therapies. Wien. Med. Wochenschr. 2019, 169, 314–322. [Google Scholar] [CrossRef]
  8. Sun, J.; Carr, M.J.; Khushalani, N.I. Principles of Targeted Therapy for Melanoma. Surg. Clin. N. Am. 2020, 100, 175–188. [Google Scholar] [CrossRef]
  9. Davies, H.; Bignell, G.R.; Cox, C.; Stephens, P.; Edkins, S.; Clegg, S.; Teague, J.; Woffendin, H.; Garnett, M.J.; Bottomley, W.; et al. Mutations of the BRAF gene in human cancer. Nature 2002, 417, 949–954. [Google Scholar] [CrossRef] [PubMed]
  10. Lim, S.Y.; Menzies, A.M.; Rizos, H. Mechanisms and strategies to overcome resistance to molecularly targeted therapy for melanoma. Cancer 2017, 123, 2118–2129. [Google Scholar] [CrossRef]
  11. LoRusso, P.M.; Schalper, K.; Sosman, J. Targeted Therapy and Immunotherapy: Emerging Biomarkers in Metastatic Melanoma. Pigment Cell Melanoma Res. 2020, 33, 390–402. [Google Scholar] [CrossRef] [PubMed]
  12. Chapman, P.B.; Hauschild, A.; Robert, C.; Haanen, J.B.; Ascierto, P.; Larkin, J.; Dummer, R.; Garbe, C.; Testori, A.; Maio, M.; et al. Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N. Engl. J. Med. 2011, 364, 2507–2516. [Google Scholar] [CrossRef]
  13. Flaherty, K.T.; Robert, C.; Hersey, P.; Nathan, P.; Garbe, C.; Milhem, M.; Demidov, L.V.; Hassel, J.C.; Rutkowski, P.; Mohr, P.; et al. Improved Survival with MEK Inhibition in BRAF-Mutated Melanoma. N. Engl. J. Med. 2012, 367, 107–114. [Google Scholar] [CrossRef]
  14. Long, G.V.; Flaherty, K.T.; Stroyakovskiy, D.; Gogas, H.; Levchenko, E.; de Braud, F.; Larkin, J.; Garbe, C.; Jouary, T.; Hauschild, A.; et al. Dabrafenib plus Trametinib versus Dabrafenib Monotherapy in Patients with Metastatic BRAF V600E/K-Mutant Melanoma: Long-Term Survival and Safety Analysis of a Phase 3 Study. Ann. Oncol. 2017, 28, 1631–1639. [Google Scholar] [CrossRef]
  15. Dummer, R.; Ascierto, P.A.; Gogas, H.J.; Arance, A.; Mandala, M.; Liszkay, G.; Garbe, C.; Schadendorf, D.; Krajsova, I.; Gutzmer, R.; et al. Encorafenib plus binimetinib versus vemurafenib or encorafenib in patients with BRAF-mutant melanoma (COLUMBUS): A multicentre, open-label, randomised phase 3 trial. Lancet Oncol. 2018, 19, 603–615. [Google Scholar] [CrossRef]
  16. Robert, C.; Grob, J.J.; Stroyakovskiy, D.; Karaszewska, B.; Hauschild, A.; Levchenko, E.; Chiarion Sileni, V.; Schachter, J.; Garbe, C.; Bondarenko, I.; et al. Five-year outcomes with dabrafenib plus trametinib in metastatic melanoma. N. Engl. J. Med. 2019, 381, 626–636. [Google Scholar] [CrossRef] [PubMed]
  17. Luke, J.J.; Flaherty, K.T.; Ribas, A.; Long, G.V. Targeted agents and immunotherapies: Optimizing outcomes in melanoma. Nat. Rev. Clin. Oncol. 2017, 14, 463–482. [Google Scholar] [CrossRef] [PubMed]
  18. Nazarian, R.; Shi, H.; Wang, Q.; Kong, X.; Koya, R.C.; Lee, H.; Chen, Z.; Lee, M.K.; Attar, N.; Sazegar, H.; et al. Melanomas acquire resistance to B-RAF(V600E) inhibition by RTK or N-RAS upregulation. Nature 2010, 468, 973–977. [Google Scholar] [CrossRef]
  19. Villanueva, J.; Vultur, A.; Herlyn, M. Resistance to BRAF inhibitors: Unraveling mechanisms and future treatment options. Cancer Res. 2011, 71, 7137–7140. [Google Scholar] [CrossRef]
  20. Lito, P.; Pratilas, C.A.; Joseph, E.W.; Tadi, M.; Halilovic, E.; Zubrowski, M.; Huang, A.; Wong, W.L.; Callahan, M.K.; Merghoub, T.; et al. Relief of Profound Feedback Inhibition of Mitogenic Signaling by RAF Inhibitors Attenuates Their Activity in BRAFV600E Melanomas. Cancer Cell 2012, 22, 668–682. [Google Scholar] [CrossRef]
  21. Yu, Q.; Xie, J.; Li, J.; Lu, Y.; Liao, L. Clinical Outcomes of BRAF plus MEK Inhibition in Melanoma: A Meta-Analysis and Systematic Review. Cancer Med. 2019, 8, 5414–5424. [Google Scholar] [CrossRef]
  22. Larkin, J.; Ascierto, P.A.; Dréno, B.; Atkinson, V.; Liszkay, G.; Maio, M.; Mandalá, M.; Demidov, L.V.; Stroyakovskiy, D.; Thomas, L.; et al. Combined vemurafenib and cobimetinib in BRAF-mutant melanoma. N. Engl. J. Med. 2014, 371, 1867–1876. [Google Scholar] [CrossRef]
  23. Ascierto, P.A.; McArthur, G.A.; Dréno, B.; Atkinson, V.; Liszkay, G.; Di Giacomo, A.M.; Mandalà, M.; Demidov, L.; Stroyakovskiy, D.; Thomas, L.; et al. Cobimetinib Combined with Vemurafenib in Advanced BRAF(V600)-Mutant Melanoma (coBRIM): Updated Efficacy Results from a Randomised, Double-Blind, Phase 3 Trial. Lancet Oncol. 2016, 17, 1248–1260. [Google Scholar] [CrossRef]
  24. Long, G.V.; Eroglu, Z.; Infante, J.; Patel, S.; Daud, A.; Johnson, D.B.; Gonzalez, R.; Kefford, R.; Hamid, O.; Schuchter, L.; et al. Long-Term Outcomes in Patients with BRAF V600-Mutant Metastatic Melanoma Who Received Dabrafenib Combined with Trametinib. J. Clin. Oncol. 2018, 36, 667–673. [Google Scholar] [CrossRef]
  25. Sabbah, M.; Najem, A.; Krayem, M.; Awada, A.; Journe, F.; Ghanem, G.E. RTK Inhibitors in Melanoma: From Bench to Bedside. Cancers 2021, 13, 1685. [Google Scholar] [CrossRef] [PubMed]
  26. Hodi, F.S.; Corless, C.L.; Giobbie-Hurder, A.; Fletcher, J.A.; Zhu, M.; Marino-Enriquez, A.; Friedlander, P.; Gonzalez, R.; Weber, J.S.; Gajewski, T.F.; et al. Imatinib for Melanomas Harboring Mutationally Activated or Amplified KIT Arising on Mucosal, Acral, and Chronically Sun-Damaged Skin. J. Clin. Oncol. 2013, 31, 3182–3190. [Google Scholar] [CrossRef]
  27. Carvajal, R.D.; Antonescu, C.R.; Wolchok, J.D.; Chapman, P.B.; Roman, R.A.; Teitcher, J.; Panageas, K.S.; Busam, K.J.; Chmielowski, B.; Lutzky, J.; et al. KIT as a Therapeutic Target in Metastatic Melanoma. JAMA 2011, 305, 2327–2334. [Google Scholar] [CrossRef]
  28. Jung, S.; Armstrong, E.; Wei, A.Z.; Ye, F.; Lee, A.; Carlino, M.S.; Sullivan, R.J.; Carvajal, R.D.; Shoushtari, A.N.; Johnson, D.B. Clinical and Genomic Correlates of Imatinib Response in Melanomas with KIT Alterations. Br. J. Cancer 2022, 127, 1726–1732. [Google Scholar] [CrossRef] [PubMed]
  29. Joshi, U.M.; Hundal, J.; Mata, J.R.; Schollenberger, M.D.; Warrier, G.; Luke, J.J.; Lipson, E.J.; Funchain, P. Beyond Checkpoint Inhibition: Keeping Therapeutic Options Open. Am. Soc. Clin. Oncol. Educ. Book 2025, 45, e473856. [Google Scholar] [CrossRef] [PubMed]
  30. Hirai, I.; Tanese, K.; Fukuda, K.; Fusumae, T.; Nakamura, Y.; Sato, Y.; Amagai, M.; Funakoshi, T. Imatinib Mesylate in Combination with Pembrolizumab in Patients with Advanced KIT-Mutant Melanoma Following Progression on Standard Therapy: A Phase I/II Trial and Study Protocol. Medicine 2021, 100, e27832. [Google Scholar] [CrossRef]
  31. Buchbinder, E.I.; Cohen, J.V.; Tarantino, G.; Lian, C.G.; Liu, D.; Haq, R.; Hodi, F.S.; Lawrence, D.P.; Giobbie-Hurder, A.; Knoerzer, D.; et al. A Phase II Study of ERK Inhibition by Ulixertinib (BVD-523) in Metastatic Uveal Melanoma. Cancer Res. Commun. 2024, 4, 1321–1327. [Google Scholar] [CrossRef] [PubMed]
  32. Najem, A.; Krayem, M.; Perdrix, A.; Kerger, J.; Awada, A.; Journe, F.; Ghanem, G. New Drug Combination Strategies in Melanoma: Current Status and Future Directions. Anticancer Res. 2017, 37, 5941–5953. [Google Scholar] [CrossRef]
  33. Rager, T.; Eckburg, A.; Patel, M.; Qiu, R.; Gantiwala, S.; Dovalovsky, K.; Fan, K.; Lam, K.; Roesler, C.; Rastogi, A.; et al. Treatment of Metastatic Melanoma with a Combination of Immunotherapies and Molecularly Targeted Therapies. Cancers 2022, 14, 3779. [Google Scholar] [CrossRef]
  34. Devlin, O.; Oladipo, O. Sequencing of Targeted Treatment and Immunotherapy in Advanced BRAF-Mutant Melanoma. J. Clin. Oncol. 2023, 41, 2295–2296. [Google Scholar] [CrossRef] [PubMed]
  35. Elkabets, M.; Vora, S.; Juric, D.; Morse, N.; Mino-Kenudson, M.; Muranen, T.; Tao, J.; Campos, A.D.; Rodon, J.; Ibrahim, Y.H.; et al. mTORC1 inhibition is required for sensitivity to PI3K inhibitors in PIK3CA-mutant breast cancer. Sci. Transl. Med. 2013, 5, 196ra99. [Google Scholar] [CrossRef]
  36. Okimoto, R.A.; Breitenbuecher, F.; Olivas, V.R.; Wu, W.; Gini, B.; Hofree, M.; Asthana, S.; Hrustanovic, G.; Flanagan, J.; Tulpule, A.; et al. Inactivation of Capicua Drives Cancer Metastasis. Nat. Genet. 2017, 49, 87–96. [Google Scholar] [CrossRef]
  37. O’Donnell, J.S.; Teng, M.W.L.; Smyth, M.J. Tumor immunoevasion through loss of interferon gamma pathway components. Nat. Rev. Clin. Oncol. 2019, 16, 442–456. [Google Scholar]
  38. Spranger, S.; Gajewski, T.F. Impact of oncogenic pathways on evasion of antitumor immune responses. Nat. Rev. Cancer 2018, 18, 139–147. [Google Scholar] [CrossRef]
  39. Ribas, A.; Lawrence, D.; Atkinson, V.; Agarwal, S.; Miller, W.H., Jr.; Carlino, M.S.; Fisher, R.; Long, G.V.; Hodi, F.S.; Tsoi, J.; et al. Combined BRAF and MEK Inhibition with PD-1 Blockade Immunotherapy in BRAF-Mutant Melanoma. Nat. Med. 2019, 25, 936–940. [Google Scholar] [CrossRef]
  40. Hu-Lieskovan, S.; Mok, S.; Homet Moreno, B.; Tsoi, J.; Robert, L.; Goedert, L.; Pinheiro, E.M.; Koya, R.C.; Graeber, T.G.; Comin-Anduix, B.; et al. Improved Antitumor Activity of Immunotherapy with BRAF and MEK Inhibitors in BRAF(V600E) Melanoma. Sci. Transl. Med. 2015, 7, 279ra41. [Google Scholar] [CrossRef] [PubMed]
  41. Frederick, D.T.; Piris, A.; Cogdill, A.P.; Cooper, Z.A.; Lezcano, C.; Ferrone, C.R.; Mitra, D.; Boni, A.; Newton, L.; Liu, C.; et al. BRAF inhibition is associated with enhanced melanoma antigen expression and a more favorable tumor microenvironment. Clin. Cancer Res. 2013, 19, 1225–1231. [Google Scholar] [CrossRef] [PubMed]
  42. Cooper, Z.A.; Juneja, V.R.; Sage, P.T.; Frederick, D.T.; Piris, A.; Mitra, D.; Lo, J.A.; Hodi, F.S.; Freeman, G.J.; Bosenberg, M.W. Response to BRAF Inhibition in Melanoma Is Enhanced When Combined with Immune Checkpoint Blockade. Cancer Immunol. Res. 2014, 2, 643–654. [Google Scholar] [CrossRef] [PubMed]
  43. Dummer, R.; Lebbé, C.; Atkinson, V.; Mandalà, M.; Nathan, P.D.; Arance, A.; Richtig, E.; Yamazaki, N.; Robert, C.; Schadendorf, D.; et al. Combined PD-1, BRAF and MEK Inhibition in Advanced BRAF-Mutant Melanoma: Safety Run-In and Biomarker Cohorts of COMBI-i. Nat. Med. 2020, 26, 1557–1563. [Google Scholar] [CrossRef]
  44. Richmond, C.S.; Vallatharasu, Y.; Deviley, J.A.; Vos, C.R.; Parsons, B.M.; Kenny, P.A. Sequential Treatment Failures in Response to BRAF/MEK and Immune Checkpoint Inhibitors Mediated by MAP2K2 and B2M Mutations in Melanoma. Exp. Mol. Pathol. 2019, 110, 104260. [Google Scholar] [CrossRef]
  45. Stege, H.; Haist, M.; Schultheis, M.; Fleischer, M.I.; Mohr, P.; Meier, F.; Schadendorf, D.; Ugurel, S.; Livingstone, E.; Zimmer, L.; et al. Discontinuation of BRAF/MEK-directed targeted therapy after complete remission of metastatic melanoma—A retrospective multicenter ADOReg study. Cancers 2021, 13, 2312. [Google Scholar] [CrossRef]
  46. Arozarena, I.; Wellbrock, C. Overcoming Resistance to BRAF Inhibitors. Ann. Transl. Med. 2017, 5, 387. [Google Scholar] [CrossRef]
  47. Morris, E.J.; Jha, S.; Restaino, C.R.; Dayananth, P.; Zhu, H.; Cooper, A.; Carr, D.; Deng, Y.; Jin, W.; Black, S.; et al. Discovery of a Novel ERK Inhibitor with Activity in Models of Acquired Resistance to BRAF and MEK Inhibitors. Cancer Discov. 2013, 3, 742–750. [Google Scholar] [CrossRef]
  48. Hung, C.; Nguyen, T.T.T.; Poulikakos, P.I.; Polsky, D. Recent Developments in Targeting the Cell Cycle in Melanoma. Cancers 2025, 17, 1291. [Google Scholar] [CrossRef]
  49. Lelliott, E.J.; Sheppard, K.E.; McArthur, G.A. Harnessing the Immunotherapeutic Potential of CDK4/6 Inhibitors in Melanoma: Is Timing Everything? NPJ Precis. Oncol. 2022, 6, 26. [Google Scholar] [CrossRef] [PubMed]
  50. Garutti, M.; Targato, G.; Buriolla, S.; Palmero, L.; Minisini, A.M.; Puglisi, F. CDK4/6 Inhibitors in Melanoma: A Comprehensive Review. Cells 2021, 10, 1334. [Google Scholar] [CrossRef]
  51. Posch, C.; Vujic, I.; Monshi, B.; Sanlorenzo, M.; Weihsengruber, F.; Rappersberger, K.; Ortiz-Urda, S. Searching for the Chokehold of NRAS Mutant Melanoma. J. Investig. Dermatol. 2016, 136, 1330–1336. [Google Scholar] [CrossRef] [PubMed]
  52. Alobaidi, R.; Islam, N.; Olkey, T.; Haribabu, Y.; Shamo, M.; Sykora, P.; Simbulan-Rosenthal, C.M.; Rosenthal, D.S. Combinational Inhibition of MEK and AKT Synergistically Induces Melanoma Stem Cell Apoptosis and Blocks NRAS Tumor Growth. Cells 2025, 14, 248. [Google Scholar] [CrossRef]
  53. Kado, S.; Komine, M. Recent Advances in Molecular Research and Treatment for Melanoma in Asian Populations. Int. J. Mol. Sci. 2025, 26, 5370. [Google Scholar] [CrossRef]
  54. Cancer Genome Atlas Network. Genomic classification of cutaneous melanoma. Cell 2015, 161, 1681–1696. [Google Scholar] [CrossRef] [PubMed]
  55. Vergara, I.A.; Wilmott, J.S.; Long, G.V.; Scolyer, R.A. Genetic drivers of non-cutaneous melanomas: Challenges and opportunities in a heterogeneous landscape. Exp. Dermatol. 2022, 31, 13–30. [Google Scholar] [CrossRef]
  56. Norain, A.; Dadachova, E. Targeted Radionuclide Therapy of Melanoma. Semin. Nucl. Med. 2016, 46, 250–259. [Google Scholar] [CrossRef] [PubMed]
  57. Namikawa, K.; Yamazaki, N. Targeted Therapy and Immunotherapy for Melanoma in Japan. Curr. Treat. Options Oncol. 2019, 20, 7. [Google Scholar] [CrossRef]
  58. Ferrucci, P.F.; Lens, M.; Cocorocchio, E. Combined BRAF-Targeted Therapy with Immunotherapy in BRAF-Mutated Advanced Melanoma Patients. Curr. Oncol. Rep. 2021, 23, 138. [Google Scholar] [CrossRef]
  59. McKenna, S.; García-Gutiérrez, L. Resistance to Targeted Therapy and RASSF1A Loss in Melanoma: What Are We Missing? Int. J. Mol. Sci. 2021, 22, 5115. [Google Scholar] [CrossRef]
  60. Jia, Z.; Zhang, Y.; Cao, L.; Wang, J.; Liang, H. Research Hotspots and Trends of Immunotherapy and Melanoma: A Bibliometric Analysis during 2014–2024. Hum. Vaccines Immunother. 2025, 21, 2464379. [Google Scholar] [CrossRef]
  61. Noman, M.Z.; Szpakowska, M.; Xiao, M.; Gao, R.; Van Moer, K.; Kumar, A.; Ollert, M.; Berchem, G.; Chevigné, A.; Janji, B. Targeting the Atypical Chemokine Receptor 2 (Ackr2) Improves the Benefit of Anti-PD-1 Immunotherapy in Melanoma Mouse Model. Oncoimmunology 2025, 14, 2494426. [Google Scholar] [CrossRef]
  62. Paul, S.; Kaya, M.; Johnsson, O.; Grauers Wiktorin, H.; Törnell, A.; Arabpour, M.; Hellstrand, K.; Martner, A. Targeting Murine Metastatic Cancers with Cholera Toxin A1-Adjuvanted Peptide Vaccines. Hum. Vaccines Immunother. 2025, 21, 2455240. [Google Scholar] [CrossRef]
  63. Shang, Y.; Zhao, H. Research Progress of Chinese Medicinal Monomers in the Process of Melanoma Occurrence. Pharm. Biol. 2025, 63, 53–67. [Google Scholar] [CrossRef] [PubMed]
  64. Chang, H.W.; Frey, G.; Wang, J.; Liu, H.; Xing, C.; Chen, J.; Boyle, W.J.; Short, J.M. Preclinical Development of Ozuriftamab Vedotin (BA3021), a Novel ROR2-Specific Conditionally Active Biologic Antibody-Drug Conjugate. MAbs 2025, 17, 2490078. [Google Scholar] [CrossRef] [PubMed]
  65. Guo, J.; Si, L.; Kong, Y.; Flaherty, K.T.; Xu, X.; Zhu, Y.; Shi, X.; Yu, X.; Xiao, L.; Ma, X.; et al. Phase II, open-label, single-arm trial of imatinib mesylate in patients with metastatic melanoma harboring c-KIT mutation or amplification. J. Clin. Oncol. 2011, 29, 2904–2909. [Google Scholar] [CrossRef]
  66. Beadling, C.; Jacobson-Dunlop, E.; Hodi, F.S.; Le, C.; Warrick, A.; Patterson, J.; Town, A.; Duvic, M.; Haluska, F.G. KIT gene mutations and copy number in melanoma subtypes. Clin. Cancer Res. 2008, 14, 6821–6828. [Google Scholar] [CrossRef]
  67. Grimaldi, A.M.; Simeone, E.; Festino, L.; Vanella, V.; Palla, M.; Ascierto, P.A. Novel mechanisms and therapeutic approaches in melanoma: Targeting the MAPK pathway. Discov. Med. 2015, 19, 455–461. [Google Scholar]
  68. 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]
  69. Menzies, A.M.; Haydu, L.E.; Carlino, M.S.; Azer, M.W.; Carr, P.J.; Kefford, R.F.; Long, G.V. Inter- and intra-patient heterogeneity of response and progression to targeted therapy in metastatic melanoma. PLoS ONE 2014, 9, e85004. [Google Scholar] [CrossRef]
  70. Wang, X.; Ma, S.; Zhu, S.; Zhu, L.; Guo, W. Advances in Immunotherapy and Targeted Therapy of Malignant Melanoma. Biomedicines 2025, 13, 225. [Google Scholar] [CrossRef]
  71. Larkin, J.; Chiarion-Sileni, V.; Gonzalez, R.; Grob, J.J.; Rutkowski, P.; Lao, C.D.; Cowey, C.L.; Schadendorf, D.; Wagstaff, J.; Dummer, R.; et al. Five-Year Survival with Combined Nivolumab and Ipilimumab in Advanced Melanoma. N. Engl. J. Med. 2019, 381, 1535–1546. [Google Scholar] [CrossRef]
  72. Robert, C.; Ribas, A.; Schachter, J.; Long, G.V.; Arance, A.; Grob, J.J.; Mortier, L.; Daud, A.; Carlino, M.S.; McNeil, C.; et al. Pembrolizumab versus Ipilimumab in Advanced Melanoma. N. Engl. J. Med. 2015, 372, 2521–2532. [Google Scholar] [CrossRef]
  73. Carlino, M.S.; Larkin, J.; Long, G.V. Immune Checkpoint Inhibitors in Melanoma. Lancet 2021, 398, 1002–1014. [Google Scholar] [CrossRef]
  74. Andtbacka, R.H.I.; Kaufman, H.L.; Collichio, F.; Amatruda, T.; Senzer, N.; Chesney, J.; Delman, K.A.; Spitler, L.E.; Puzanov, I.; Agarwala, S.S.; et al. Talimogene Laherparepvec Improves Durable Response Rate in Patients with Advanced Melanoma. J. Clin. Oncol. 2015, 33, 2780–2788. [Google Scholar] [CrossRef]
  75. Robert, C.; Gastman, B.; Gogas, H.; Rutkowski, P.; Long, G.V.; Chaney, M.F.; Joshi, H.; Lin, Y.L.; Snyder, W.; Chesney, J.A. Open-Label, Phase II Study of Talimogene Laherparepvec plus Pembrolizumab for the Treatment of Advanced Melanoma That Progressed on Prior Anti-PD-1 Therapy: MASTERKEY-115. Eur. J. Cancer 2024, 207, 114120. [Google Scholar] [CrossRef]
  76. Garbe, C.; Amaral, T.; Peris, K.; Hauschild, A.; Arenberger, P.; Basset-Seguin, N.; Bastholt, L.; Bataille, V.; Brochez, L.; Del Marmol, V.; et al. European Consensus-Based Interdisciplinary Guideline for Melanoma. Part 1: Diagnostics—Update 2024. Eur. J. Cancer 2025, 215, 115152. [Google Scholar] [CrossRef] [PubMed]
  77. Tagliaferri, L.; Lancellotta, V.; Fionda, B.; Mangoni, M.; Casà, C.; Di Stefani, A.; Pagliara, M.M.; D’Aviero, A.; Schinzari, G.; Chiesa, S.; et al. Immunotherapy and Radiotherapy in Melanoma: A Multidisciplinary Comprehensive Review. Hum. Vaccines Immunother. 2022, 18, 1903827. [Google Scholar] [CrossRef] [PubMed]
  78. Long, G.V.; Hauschild, A.; Santinami, M.; Kirkwood, J.M.; Atkinson, V.; Mandala, M.; Merelli, B.; Sileni, V.C.; Nyakas, M.; Haydon, A.; et al. Final Results for Adjuvant Dabrafenib plus Trametinib in Stage III Melanoma. N. Engl. J. Med. 2024, 391, 1709–1720. [Google Scholar] [CrossRef]
  79. Sharma, A.; Shah, S.R.; Illum, H.; Dowell, J. Vemurafenib: Targeted Inhibition of Mutated BRAF for Treatment of Advanced Melanoma and Its Potential in Other Malignancies. Drugs 2012, 72, 2207–2222. [Google Scholar] [CrossRef] [PubMed]
  80. Shaw, H.M.; Nathan, P.D. Vemurafenib in Melanoma. Expert Rev. Anticancer Ther. 2013, 13, 513–522. [Google Scholar] [CrossRef]
  81. Bollag, G.; Tsai, J.; Zhang, J.; Zhang, C.; Ibrahim, P.; Nolop, K.; Hirth, P. Vemurafenib: The First Drug Approved for BRAF-Mutant Cancer. Nat. Rev. Drug Discov. 2012, 11, 873–886. [Google Scholar] [CrossRef]
  82. Tsai, J.; Lee, J.T.; Wang, W.; Zhang, J.; Cho, H.; Mamo, S.; Bremer, R.; Gillette, S.; Kong, J.; Haass, N.K.; et al. Discovery of a Selective Inhibitor of Oncogenic B-Raf Kinase with Potent Antimelanoma Activity. Proc. Natl. Acad. Sci. USA 2008, 105, 3041–3046. [Google Scholar] [CrossRef]
  83. Heidorn, S.J.; Milagre, C.; Whittaker, S.; Nourry, A.; Niculescu-Duvaz, D.; Dhomen, N.; Marais, R. Kinase-Dead BRAF and Oncogenic RAS Cooperate to Drive Tumor Progression through CRAF. Cell 2010, 140, 209–221. [Google Scholar] [CrossRef]
  84. Daugaard, N.D.; Tholstrup, R.; Tornby, J.R.; Bendixen, S.M.; Larsen, F.T.; De Zio, D.; Barnkob, M.B.; Ravnskjaer, K.; Brewer, J.R. Characterization of Human Melanoma Skin Cancer Models: A Step towards Model-Based Melanoma Research. Acta Biomater. 2025, 191, 308–324. [Google Scholar] [CrossRef]
  85. Wanchoo, R.; Jhaveri, K.D.; Deray, G.; Launay-Vacher, V. Renal Effects of BRAF Inhibitors: A Systematic Review by the Cancer and the Kidney International Network. Clin. Kidney J. 2016, 9, 245–251. [Google Scholar] [CrossRef]
  86. Flaherty, K.T.; Yasothan, U.; Kirkpatrick, P. Vemurafenib. Nat. Rev. Drug Discov. 2011, 10, 811–812. [Google Scholar] [CrossRef]
  87. Sosman, J.A.; Kim, K.B.; Schuchter, L.; Gonzalez, R.; Pavlick, A.C.; Weber, J.S.; McArthur, G.A.; Hutson, T.E.; Moschos, S.J.; Flaherty, K.T.; et al. Survival in BRAF V600–Mutant Advanced Melanoma Treated with Vemurafenib. N. Engl. J. Med. 2012, 366, 707–714. [Google Scholar] [CrossRef] [PubMed]
  88. Haroche, J.; Cohen-Aubart, F.; Emile, J.F.; Arnaud, L.; Maksud, P.; Charlotte, F.; Cluzel, P.; Aouba, A.; Hermine, O.; Amoura, Z. Dramatic Efficacy of Vemurafenib in Both Multisystemic and Refractory Erdheim–Chester Disease and Langerhans Cell Histiocytosis Harboring the BRAF V600E Mutation. Blood 2013, 121, 1495–1500. [Google Scholar] [CrossRef]
  89. Diamond, E.L.; Subbiah, V.; Lockhart, A.C.; Blay, J.Y.; Puzanov, I.; Chau, I.; Wolf, J.; Raje, N.; Van de Velde, H.; Thappa, R.; et al. Vemurafenib for BRAF V600–Mutant Erdheim–Chester Disease and Langerhans Cell Histiocytosis: Analysis of Data from the Histology-Independent, Phase 2 VE-BASKET Study. JAMA Oncol. 2018, 4, 384–388. [Google Scholar] [CrossRef] [PubMed]
  90. Anaya, Y.A.; Bracho, R.P.; Chauhan, S.C.; Tripathi, M.K.; Bandyopadhyay, D. Small Molecule B-RAF Inhibitors as Anti-Cancer Therapeutics: Advances in Discovery, Development, and Mechanistic Insights. Int. J. Mol. Sci. 2025, 26, 2676. [Google Scholar] [CrossRef] [PubMed]
  91. Hauschild, A.; Grob, J.J.; Demidov, L.V.; Jouary, T.; Gutzmer, R.; Millward, M.; Rutkowski, P.; Blank, C.U.; Miller, W.H.; Kaempgen, E.; et al. Dabrafenib in BRAF-Mutated Metastatic Melanoma: A Multicentre, Open-Label, Phase 3 Randomised Controlled Trial. Lancet 2012, 380, 358–365. [Google Scholar] [CrossRef]
  92. McArthur, G.A.; Chapman, P.B.; Robert, C.; Larkin, J.; Haanen, J.B.; Dummer, R.; Ascierto, P.A.; Garbe, C.; Testori, A.; Maio, M.; et al. Safety and Efficacy of Vemurafenib in BRAF(V600E) and BRAF(V600K) Mutation-Positive Melanoma (BRIM-3): Extended Follow-Up of a Phase 3, Randomised, Open-Label Study. Lancet Oncol. 2014, 15, 323–332. [Google Scholar] [CrossRef] [PubMed]
  93. Long, G.V.; Stroyakovskiy, D.; Gogas, H.; Levchenko, E.; De Braud, F.; Larkin, J.; Garbe, C.; Jouary, T.; Hauschild, A.; Grob, J.J.; et al. Dabrafenib and Trametinib versus Dabrafenib and Placebo for Val600 BRAF-Mutant Melanoma: A Multicentre, Double-Blind, Phase 3 Randomised Controlled Trial. Lancet 2015, 386, 444–451. [Google Scholar] [CrossRef]
  94. Poulikakos, P.I.; Persaud, Y.; Janakiraman, M.; Kong, X.; Ng, C.; Moriceau, G.; Shi, H.; Atefi, M.; Titz, B.; Gabay, M.T.; et al. RAF Inhibitor Resistance Is Mediated by Dimerization of Aberrantly Spliced BRAF(V600E). Nature 2011, 480, 387–390. [Google Scholar] [CrossRef] [PubMed]
  95. Shi, H.; Hugo, W.; Kong, X.; Hong, A.; Koya, R.C.; Moriceau, G.; Chodon, T.; Guo, R.; Johnson, D.B.; Dahlman, K.B.; et al. Acquired Resistance and Clonal Evolution in Melanoma during BRAF Inhibitor Therapy. Cancer Discov. 2014, 4, 80–93. [Google Scholar] [CrossRef]
  96. 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] [PubMed]
  97. Titz, B.; Lomova, A.; Le, A.; Hugo, W.; Ten Hoeve, J.; Shi, H.; Moriceau, G.; Song, C.; Hong, A.; Johnson, D.B.; et al. JUN Dependency in Distinct Early and Late BRAF Inhibitor Resistance States of Melanoma. Cell Discov. 2016, 2, 16028. [Google Scholar] [CrossRef]
  98. Lacouture, M.E.; Duvic, M.; Hauschild, A.; Prieto, V.G.; Robert, C.; Schadendorf, D.; Kim, C.C.; McCormack, C.J.; Myskowski, P.L.; Spleiss, O.; et al. Analysis of Dermatologic Events in Vemurafenib-Treated Patients with Melanoma. Oncologist 2013, 18, 314–322. [Google Scholar] [CrossRef]
  99. Anforth, R.M.; Blumetti, T.C.; Kefford, R.F.; Sharma, R.; Scolyer, R.A.; Kossard, S.; Long, G.V.; Fernandez-Peñas, P. Cutaneous Manifestations of Dabrafenib (GSK2118436): A Selective Inhibitor of Mutant BRAF in Patients with Metastatic Melanoma. Br. J. Dermatol. 2012, 167, 1153–1160. [Google Scholar] [CrossRef]
  100. Jordan, E.J.; Kelly, C.M. Vemurafenib for the Treatment of Melanoma. Expert Opin. Pharmacother. 2012, 13, 2533–2543. [Google Scholar] [CrossRef]
  101. Poduje, S.; Brozić, J.M.; Prkačin, I.; Delaš Aždajić, M.; Goren, A. Vemurafenib and Cobimetinib-Induced Toxic Epidermal Necrolysis in a Patient with Metastatic Melanoma. Dermatol. Ther. 2020, 33, e13174. [Google Scholar] [CrossRef] [PubMed]
  102. Su, F.; Viros, A.; Milagre, C.; Trunzer, K.; Bollag, G.; Spleiss, O.; Reis-Filho, J.S.; Kong, X.; Koya, R.C.; Flaherty, K.T.; et al. RAS Mutations in Cutaneous Squamous-Cell Carcinomas in Patients Treated with BRAF Inhibitors. N. Engl. J. Med. 2012, 366, 207–215. [Google Scholar] [CrossRef]
  103. Oberholzer, P.A.; Kee, D.; Dziunycz, P.; Sucker, A.; Kamsukom, N.; Jones, R.; Prena, J.; Huber, C.; Goldinger, S.M.; Dornbierer, J.; et al. RAS Mutations Are Associated with the Development of Cutaneous Squamous Cell Tumors in Patients Treated with RAF Inhibitors. J. Clin. Oncol. 2012, 30, 316–321. [Google Scholar] [CrossRef]
  104. Robert, C.; Karaszewska, B.; Schachter, J.; Rutkowski, P.; Mackiewicz, A.; Stroiakovski, D.; Lichinitser, M.; Dummer, R.; Grange, F.; Mortier, L.; et al. Improved Overall Survival in Melanoma with Combined Dabrafenib and Trametinib. N. Engl. J. Med. 2015, 372, 30–39. [Google Scholar] [CrossRef]
  105. Wu, J.; Ding, Q.; Zhang, Q.; Chen, Q.; Wen, X.; Ding, Y.; Li, J.; Chen, Z.; Zhang, T.; Wang, J.; et al. Addition of Anti-PD-1 Immunotherapy to BRAF Inhibitor-Based Targeted Therapy Improves Real-World Survival and Delays Brain Metastases in Patients with BRAFV600-Mutant Advanced Melanoma: A Multicenter Cohort Study. MedComm 2025, 6, e70102. [Google Scholar] [CrossRef]
  106. Gutzmer, R.; Stroyakovskiy, D.; Gogas, H.; Robert, C.; Larkin, J.; Nyakas, M.; Dutriaux, C.; Garbe, C.; de la Cruz-Merino, L.; Schachter, J.; et al. Atezolizumab, Vemurafenib, and Cobimetinib as First-Line Treatment for Unresectable Advanced BRAF(V600) Mutation-Positive Melanoma (IMspire150): Primary Analysis of the Randomised, Double-Blind, Placebo-Controlled, Phase 3 Trial. Lancet 2020, 395, 1835–1844. [Google Scholar] [CrossRef]
  107. Dugan, M.M.; Perez, M.C.; Karapetyan, L.; Zager, J.S. Combination Atezolizumab, Cobimetinib, and Vemurafenib as a Treatment Option in BRAF V600 Mutation-Positive Melanoma: Patient Selection and Perspectives. Cancer Manag. Res. 2024, 16, 933–939. [Google Scholar] [CrossRef] [PubMed]
  108. Livingstone, E.; Gogas, H.J.; Kandolf, L.; Meier, F.; Eigentler, T.K.; Ziemer, M.; Terheyden, P.; Gesierich, A.; Herbst, R.A.; Kähler, K.C.; et al. Early Switch from Run-In with Targeted to Immunotherapy in Advanced BRAFV600-Positive Melanoma: Final Results of the Randomised Phase II ImmunoCobiVem Trial. ESMO Open 2025, 10, 105053. [Google Scholar] [CrossRef] [PubMed]
  109. Block, M.S.; Nelson, G.D.; Chen, J.; Johnson, S.; Yang, L.; Flotte, T.J.; Grewal, E.P.; McWilliams, R.R.; Kottschade, L.A.; Domingo-Musibay, E.; et al. Neoadjuvant Cobimetinib and Atezolizumab with or without Vemurafenib for Stage III Melanoma: Outcomes and the Impact of the Microbiome from the NeoACTIVATE Trial. J. Immunother. Cancer 2025, 13, e011706. [Google Scholar] [CrossRef]
  110. Boz Er, A.B.; Sheldrake, H.M.; Sutherland, M. Overcoming Vemurafenib Resistance in Metastatic Melanoma: Targeting Integrins to Improve Treatment Efficacy. Int. J. Mol. Sci. 2024, 25, 7946. [Google Scholar] [CrossRef]
  111. Galluzzi, L.; Humeau, J.; Buqué, A.; Zitvogel, L.; Kroemer, G. Immunostimulation with Chemotherapy in the Era of Immune Checkpoint Inhibitors. Nat. Rev. Clin. Oncol. 2020, 17, 725–741. [Google Scholar] [CrossRef] [PubMed]
  112. Ruscetti, M.; Dadashian, E.L.; Guo, W.; Quach, B.; Mulholland, D.J.; Park, J.W.; Tran, L.M.; Kobayashi, N.; Bianchi-Frias, D.; Xing, Y.; et al. HDAC Inhibition Impedes Epithelial-Mesenchymal Plasticity and Suppresses Metastatic, Castration-Resistant Prostate Cancer. Oncogene 2016, 35, 3781–3795. [Google Scholar] [CrossRef]
  113. Acquaviva, J.; Smith, D.L.; Jimenez, J.P.; Zhang, C.; Sequeira, M.; He, S.; Sang, J.; Bates, R.C.; Proia, D.A. Overcoming Acquired BRAF Inhibitor Resistance in Melanoma via Targeted Inhibition of HSP90 with Ganetespib. Mol. Cancer Ther. 2014, 13, 353–363. [Google Scholar] [CrossRef] [PubMed]
  114. Biersack, B.; Nitzsche, B.; Höpfner, M. Histone Deacetylases in the Regulation of Cell Death and Survival Mechanisms in Resistant BRAF-Mutant Cancers. Cancer Drug Resist. 2025, 8, 6. [Google Scholar] [CrossRef]
  115. Das Thakur, M.; Salangsang, F.; Landman, A.S.; Sellers, W.R.; Pryer, N.K.; Levesque, M.P.; Dummer, R.; McMahon, M. Modelling Vemurafenib Resistance in Melanoma Reveals a Strategy to Forestall Drug Resistance. Nature 2013, 494, 251–255. [Google Scholar] [CrossRef]
  116. Wang, M.; Yu, F.; Zhang, Y. Present and Future of Cancer Nano-Immunotherapy: Opportunities, Obstacles and Challenges. Mol. Cancer 2025, 24, 26. [Google Scholar] [CrossRef] [PubMed]
  117. Shebrain, A.; Idris, O.A.; Jawad, A.; Zhang, T.; Xing, Y. Advancements and Challenges in Personalized Therapy for BRAF-Mutant Melanoma: A Comprehensive Review. J. Clin. Med. 2024, 13, 5409. [Google Scholar] [CrossRef]
  118. Holderfield, M.; Deuker, M.M.; McCormick, F.; McMahon, M. Targeting RAF Kinases for Cancer Therapy: BRAF-Mutated Melanoma and Beyond. Nat. Rev. Cancer 2014, 14, 455–467. [Google Scholar] [CrossRef]
  119. Karoulia, Z.; Gavathiotis, E.; Poulikakos, P.I. New Perspectives for Targeting RAF Kinase in Human Cancer. Nat. Rev. Cancer 2017, 17, 676–691. [Google Scholar] [CrossRef]
  120. Kim, G.; McKee, A.E.; Ning, Y.M.; Hazarika, M.; Theoret, M.; Johnson, J.R.; Xu, Q.C.; Tang, S.; Sridhara, R.; Jiang, X.; et al. FDA Approval Summary: Vemurafenib for Treatment of Unresectable or Metastatic Melanoma with the BRAFV600E Mutation. Clin. Cancer Res. 2014, 20, 4994–5000. [Google Scholar] [CrossRef]
  121. Carlino, M.S.; Fung, C.; Shahheydari, H.; Todd, J.R.; Boyd, S.C.; Irvine, M.; Nagrial, A.; Wilmott, J.S.; Kefford, R.F.; Long, G.V.; et al. Preexisting MEK1P124 Mutations Diminish Response to BRAF Inhibitors in Melanoma. Clin. Cancer Res. 2015, 21, 98–105. [Google Scholar] [CrossRef] [PubMed]
  122. Prado-Souza, L.F.L.D.; Ferraz, L.S.; Citrangulo, T.T., Jr.; Ribeiro, C.A.J.; Amaral, D.T.D.; Arruda, D.C.; Oliveira, É.A.; Chammas, R.; Maria-Engler, S.S.; Rodrigues, T. Exploiting Paradoxical Activation of Oncogenic MAPK Signaling by Targeting Mitochondria to Sensitize NRAS Mutant-Melanoma to Vemurafenib. Int. J. Mol. Sci. 2025, 26, 2675. [Google Scholar] [CrossRef] [PubMed]
  123. Suzuki, Y.; Usuki, S.; Nishizawa, M.; Tanaka, N.; Suhara, Y.; Yajima, I. Acyclic Retinoid Overcomes Vemurafenib Resistance in Melanoma Cells via Dual Inhibition of MAPK and PI3K/AKT/mTOR Pathways. Anticancer Res. 2025, 45, 2265–2278. [Google Scholar] [CrossRef]
  124. van der Hiel, B.; de Wit-van der Veen, B.J.; van den Eertwegh, A.J.M.; Vogel, W.V.; Stokkel, M.P.M.; Lopez-Yurda, M.; Boellaard, R.; Kapiteijn, E.W.; Hospers, G.A.P.; Aarts, M.J.B.; et al. Metabolic Parameters on Baseline and Early [18F]FDG PET/CT as a Predictive Biomarker for Resistance to BRAF/MEK Inhibition in Advanced Cutaneous BRAFV600-Mutated Melanoma. EJNMMI Res. 2025, 15, 60. [Google Scholar] [CrossRef]
  125. Foda, B.M.; Baker, A.E.; Joachimiak, Ł.; Mazur, M.; Neubig, R.R. Mechanistic Insights into Rho/MRTF Inhibition-Induced Apoptotic Events and Prevention of Drug Resistance in Melanoma: Implications for the Involvement of Pirin. Front. Pharmacol. 2025, 16, 1505000. [Google Scholar] [CrossRef]
  126. Kim, J.; Brunetti, B.; Kumar, A.; Mangla, A.; Honda, K.; Yoshida, A. Inhibition of Glutaminase Elicits Senolysis in Therapy-Induced Senescent Melanoma Cells. Cell Death Dis. 2024, 15, 902. [Google Scholar] [CrossRef] [PubMed]
  127. Zhang, R.; Wang, Z.; Wang, H.; Li, L.; Dong, L.; Ding, L.; Li, Q.; Zhu, L.; Zhang, T.; Zhu, Y.; et al. CTHRC1 Is Associated with BRAF(V600E) Mutation and Correlates with Prognosis, Immune Cell Infiltration, and Drug Resistance in Colon Cancer, Thyroid Cancer, and Melanoma. Biomol. Biomed. 2024, 25, 42–61. [Google Scholar] [CrossRef]
  128. Li, Y.; Feng, Y.; Chen, D. Interfering Nuclear Protein Laminb1 Induces DNA Damage and Reduces Vemurafenib Resistance in Melanoma Cells In Vitro. Cancers 2024, 16, 4060. [Google Scholar] [CrossRef]
  129. Shalem, O.; Sanjana, N.E.; Hartenian, E.; Shi, X.; Scott, D.A.; Mikkelson, T.; Heckl, D.; Ebert, B.L.; Root, D.E.; Doench, J.G.; et al. Genome-Scale CRISPR-Cas9 Knockout Screening in Human Cells. Science 2014, 343, 84–87. [Google Scholar] [CrossRef]
  130. Wang, X.; Zhong, F.; Chen, T.; Wang, H.; Wang, W.; Jin, H.; Li, C.; Guo, X.; Liu, Y.; Zhang, Y.; et al. Cholesterol Neutralized Vemurafenib Treatment by Promoting Melanoma Stem-Like Cells via Its Metabolite 27-Hydroxycholesterol. Cell. Mol. Life Sci. 2024, 81, 226. [Google Scholar] [CrossRef]
  131. Park, B.S.; Jeon, H.; Kim, Y.; Kwon, H.; Choi, G.E.; Chi, S.G.; Park, H.M.; Lee, H.; Kim, T. Polyamine and EIF5A Hypusination Downstream of c-Myc Confers Targeted Therapy Resistance in BRAF Mutant Melanoma. Mol. Cancer 2024, 23, 136. [Google Scholar] [CrossRef]
  132. Flaherty, K.T.; Infante, J.R.; Daud, A.; Gonzalez, R.; Kefford, R.F.; Sosman, J.; Hamid, O.; Schuchter, L.; Cebon, J.; Ibrahim, N.; et al. Combined BRAF and MEK inhibition in melanoma with BRAF V600 mutations. N. Engl. J. Med. 2012, 367, 1694–1703. [Google Scholar] [CrossRef] [PubMed]
  133. Yao, Z.; Torres, N.M.; Tao, A.; Gao, Y.; Luo, L.; Li, Q.; de Stanchina, E.; Abdel-Wahab, O.; Solit, D.B.; Poulikakos, P.I.; et al. BRAF mutants evade ERK-dependent feedback by different mechanisms that determine their sensitivity to pharmacologic inhibition. Cancer Cell 2015, 28, 370–383. [Google Scholar] [CrossRef]
  134. Owen, K.L.; Brockwell, N.K.; Parker, B.S. JAK-STAT Signaling: A Double-Edged Sword of Immune Regulation and Cancer Progression. Cancers 2019, 11, 2002. [Google Scholar] [CrossRef] [PubMed]
  135. U.S. Food and Drug Administration. Drug Approval Package Mekinist (Trametinib). Available online: https://www.accessdata.fda.gov/drugsatfda_docs/nda/2013/204114Orig1s000TOC.cfm (accessed on 24 June 2025).
  136. Foth, M.; McMahon, M. Autophagy Inhibition in BRAF-Driven Cancers. Cancers 2021, 13, 3498. [Google Scholar] [CrossRef]
  137. Hoffner, B.; Benchich, K. Trametinib: A Targeted Therapy in Metastatic Melanoma. J. Adv. Pract. Oncol. 2018, 9, 741–745. [Google Scholar]
  138. Sullivan, R.J.; Flaherty, K.T. Resistance to BRAF-targeted therapy in melanoma. Eur. J. Cancer 2013, 49, 1297–1304. [Google Scholar] [CrossRef] [PubMed]
  139. Wagle, N.; Emery, C.; Berger, M.F.; Davis, M.J.; Sawyer, A.; Pochanard, P.; Kehoe, S.M.; Johannessen, C.M.; MacConaill, L.E.; Hahn, W.C.; et al. Dissecting therapeutic resistance to RAF inhibition in melanoma by tumor genomic profiling. J. Clin. Oncol. 2011, 29, 3085–3096. [Google Scholar] [CrossRef] [PubMed]
  140. Khunger, A.; Khunger, M.; Velcheti, V. Dabrafenib in Combination with Trametinib in the Treatment of Patients with BRAF V600-Positive Advanced or Metastatic Non-Small Cell Lung Cancer: Clinical Evidence and Experience. Ther. Adv. Respir. Dis. 2018, 12, 1753466618767611. [Google Scholar] [CrossRef] [PubMed]
  141. Quaglino, P.; Fava, P.; Tonella, L.; Rubatto, M.; Ribero, S.; Fierro, M.T. Treatment of Advanced Metastatic Melanoma. Dermatol. Pract. Concept 2021, 11 (Suppl. S1), e2021164S. [Google Scholar] [CrossRef]
  142. Gouda, M.A.; Subbiah, V. Expanding the Benefit: Dabrafenib/Trametinib as Tissue-Agnostic Therapy for BRAF V600E-Positive Adult and Pediatric Solid Tumors. Am. Soc. Clin. Oncol. Educ. Book 2023, 43, e404770. [Google Scholar] [CrossRef]
  143. Mubarak, O.; Middleton, G.W. The Impact of the Immunological Context on Outcomes of Solid Cancer Patients Treated with Genotype-Matched Targeted Therapies: A Comprehensive Review. Ann. Oncol. 2025, 36, 737–748. [Google Scholar] [CrossRef] [PubMed]
  144. Li, F.; Shen, F. Metastatic Pancreatic Cancer with Activating BRAF V600E Mutations: A Case Report. World J. Clin. Cases 2025, 13, 101665. [Google Scholar] [CrossRef]
  145. Cheng, Z.; Wang, C.; Cai, Y. Primary Rectal Malignant Melanoma with Schistosomiasis. Intractable Rare Dis. Res. 2025, 14, 145–147. [Google Scholar] [CrossRef]
  146. Weber, J.; Haque, W.; Markovic, S.N.; Salama, A.K.S.; Mehmi, I.; Sullivan, R.J.; Najjar, Y.G.; van Akkooi, A.C.J.; Menzies, A.M.; Long, G.V.; et al. Relapse-Free Survival with Adjuvant Dabrafenib/Trametinib Therapy after Relapse on a Prior Adjuvant CPI in BRAF V600-Mutated Stage III/IV Melanoma. Oncologist 2025, 30, oyae289. [Google Scholar] [CrossRef]
  147. Gorry, C.; McCullagh, L.; O’Donnell, H.; Barrett, S.; Schmitz, S.; Barry, M.; Curtin, K.; Beausang, E.; Barry, R.; Coyne, I. Neoadjuvant Treatment for Stage III and IV Cutaneous Melanoma. Cochrane Database Syst. Rev. 2023, 1, CD012974. [Google Scholar] [CrossRef]
  148. Keser, M.; Atmaca, H. Let-7a MicroRNA Modulates Caspase-3-Dependent Apoptosis in Melanoma Cells Treated with Dabrafenib and Trametinib Combination. Ir. J. Med. Sci. 2025, 194, 797–805. [Google Scholar] [CrossRef]
  149. Long, G.V.; Hauschild, A.; Santinami, M.; Atkinson, V.; Mandalà, M.; Chiarion-Sileni, V.; Larkin, J.; Nyakas, M.; Dutriaux, C.; Haydon, A.; et al. Adjuvant dabrafenib plus trametinib in stage III BRAF-mutated melanoma. N. Engl. J. Med. 2017, 377, 1813–1823. [Google Scholar] [CrossRef]
  150. Davies, M.A.; Saiag, P.; Robert, C.; Grob, J.J.; Flaherty, K.T.; Arance, A.M.; Demidov, L.V.; Garbe, C.; Chiarion-Sileni, V.; Rutkowski, P.; et al. Dabrafenib plus trametinib in patients with BRAF-mutant melanoma brain metastases (COMBI-MB): A multicentre, multicohort, open-label, phase 2 trial. Lancet Oncol. 2017, 18, 863–873. [Google Scholar] [CrossRef] [PubMed]
  151. Wilmott, J.S.; Tawbi, H.; Engh, J.A.; Amankulor, N.M.; Shivalingam, B.; Banerjee, H.; Vergara, I.A.; Lee, H.; Johansson, P.A.; Ferguson, P.M.; et al. Clinical Features Associated with Outcomes and Biomarker Analysis of Dabrafenib plus Trametinib Treatment in Patients with BRAF-Mutant Melanoma Brain Metastases. Clin. Cancer Res. 2023, 29, 521–531. [Google Scholar] [CrossRef] [PubMed]
  152. Jia, D.D.; Xu, Y.; Ren, Z.W.; Li, L.Q.; Zhang, L.; Li, Y.; Lou, L.S.; Yao, W.T.; Liu, Z.; Li, X.A.; et al. Is One Year Enough? Extended Adjuvant Dabrafenib Plus Trametinib for Chinese Patients with Resected Stage III Melanoma. J. Dermatol. 2025, 52, 1146–1151. [Google Scholar] [CrossRef] [PubMed]
  153. Chiaravalli, S.; Indini, A.; Del Vecchio, M.; Casanova, M.; Massimino, M.; Bergamaschi, L.; Ferrari, A. Dabrafenib and Trametinib for the Treatment of Pediatric and Adolescent Melanoma: Single Center Experience Data from Italian Compassionate Use. Pediatr. Blood Cancer 2025, 72, e31644. [Google Scholar] [CrossRef] [PubMed]
  154. Pélouard, F.; Chedru-Legros, V.; Nganoa, C.; Dompmartin, A.; L’Orphelin, J.M. Optimizing Targeted Therapy for Metastatic Melanoma: A Combination of Encorafenib and Trametinib beyond Standard Protocols. Dermatol. Rep. 2025. [Google Scholar] [CrossRef]
  155. Zhang, Y.; Zhang, X.; Shao, W.; Gao, J.; Xiang, M.; Wang, Y.; Liu, M.; Zhang, W.; Liang, X. MEK Inhibitors for the Treatment of Immunotherapy-Resistant, AGK-BRAF Fusion Advanced Acral Melanoma: A Case Report and Literature Review. J. Cancer Res. Clin. Oncol. 2025, 151, 133. [Google Scholar] [CrossRef]
  156. Wiesweg, M.; Alaffas, A.; Rasokat, A.; Saalfeld, F.C.; Rost, M.; Assmann, C.; Herster, F.; Hilbrandt, M.; Griesinger, F.; Kron, A.; et al. Treatment Sequences in BRAF-V600-Mutated NSCLC: First-Line Targeted Therapy Versus First-Line (Chemo-) Immunotherapy. J. Thorac. Oncol. 2025, 20, 1328–1335. [Google Scholar] [CrossRef]
  157. Goethe, E.A.; Srinivasan, S.; Kumar, S.; Prabhu, S.S.; Gubbiotti, M.A.; Ferguson, S.D. High-Grade Astrocytoma with Piloid Features: A Single-Institution Case Series and Literature Review. Acta Neuropathol. Commun. 2025, 13, 82. [Google Scholar] [CrossRef]
  158. Kim, K.B.; Kefford, R.; Pavlick, A.C.; Infante, J.R.; Ribas, A.; Sosman, J.A.; Fecher, L.A.; Millward, M.; McArthur, G.A.; Hwu, P.; et al. Phase II Study of the MEK1/MEK2 Inhibitor Trametinib in Patients with Metastatic BRAF-Mutant Cutaneous Melanoma Previously Treated with or without a BRAF Inhibitor. J. Clin. Oncol. 2013, 31, 482–489. [Google Scholar] [CrossRef]
  159. Livingstone, E.; Zimmer, L.; Vaubel, J.; Schadendorf, D. BRAF, MEK and KIT Inhibitors for Melanoma: Adverse Events and Their Management. Chin. Clin. Oncol. 2014, 3, 29. [Google Scholar] [CrossRef]
  160. 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]
  161. Grimaldi, A.M.; Simeone, E.; Festino, L.; Vanella, V.; Strudel, M.; Ascierto, P.A. MEK Inhibitors in the Treatment of Metastatic Melanoma and Solid Tumors. Am. J. Clin. Dermatol. 2017, 18, 745–754. [Google Scholar] [CrossRef]
  162. Wilmott, J.S.; Long, G.V.; Howle, J.R.; Haydu, L.E.; Sharma, R.; Thompson, J.F.; Kefford, R.F.; Hersey, P.; Scolyer, R.A. Selective BRAF inhibitors induce marked T-cell infiltration into human metastatic melanoma. Clin. Cancer Res. 2012, 18, 1386–1394. [Google Scholar] [CrossRef]
  163. Sullivan, R.J.; Hamid, O.; Gonzalez, R.; Infante, J.R.; Patel, S.P.; Kudchadkar, R.R.; Sznol, M.; Kluger, H.; Maio, M.; Mitchell, T.C.; et al. Atezolizumab plus cobimetinib and vemurafenib in BRAF-mutated melanoma patients. Nat. Med. 2019, 25, 929–935. [Google Scholar] [CrossRef]
  164. Planchard, D.; Besse, B.; Groen, H.J.M.; Souquet, P.J.; Quoix, E.; Baik, C.S.; Barlesi, F.; Kim, T.M.; Mazieres, J.; Novello, S.; et al. Dabrafenib plus trametinib in patients with previously untreated BRAF(V600E)-mutant metastatic non-small cell lung cancer: An open-label, phase 2 trial. Lancet Oncol. 2017, 18, 1307–1316. [Google Scholar] [CrossRef]
  165. Subbiah, V.; Kreitman, R.J.; Wainberg, Z.A.; Cho, B.C.; Schellens, J.H.M.; Soria, J.C.; Gainor, J.F.; Vansteenkiste, J.F.; Wolf, J.; Seto, T.; et al. Dabrafenib and trametinib treatment in patients with locally advanced or metastatic BRAF V600E-mutant anaplastic thyroid cancer. J. Clin. Oncol. 2018, 36, 7–13. [Google Scholar] [CrossRef]
  166. Ebert, P.J.R.; Cheung, J.; Yang, Y.; McNamara, E.; Hong, R.; Moskalenko, M.; Gould, S.E.; Maecker, H.; Irving, B.A.; Kim, J.M.; et al. MAP kinase inhibition promotes T cell and anti-tumor activity in combination with PD-L1 checkpoint blockade. Immunity 2016, 44, 609–621. [Google Scholar] [CrossRef] [PubMed]
  167. Boni, A.; Cogdill, A.P.; Dang, P.; Udayakumar, D.; Njauw, C.N.; Sloss, C.M.; Ferrone, C.R.; Flaherty, K.T.; Lawrence, D.P.; Fisher, D.E.; et al. Selective BRAF(V600E) inhibition enhances T-cell recognition of melanoma without affecting lymphocyte function. Cancer Res. 2010, 70, 5213–5219. [Google Scholar] [CrossRef]
  168. Sumimoto, H.; Imabayashi, F.; Iwata, T.; Kawakami, Y. The BRAF–MAPK signaling pathway is essential for cancer-immune evasion in human melanoma cells. J. Exp. Med. 2006, 203, 1651–1656. [Google Scholar] [CrossRef] [PubMed]
  169. Choi, H.Y.; Chang, J.E. Targeted Therapy for Cancers: From Ongoing Clinical Trials to FDA-Approved Drugs. Int. J. Mol. Sci. 2023, 24, 13618. [Google Scholar] [CrossRef] [PubMed]
  170. Corcoran, R.B.; Atreya, C.E.; Falchook, G.S.; Kwak, E.L.; Ryan, D.P.; Bendell, J.C.; Hamid, O.; Messersmith, W.A.; Daud, A.; Kurzrock, R.; et al. Combined BRAF and MEK Inhibition With Dabrafenib and Trametinib in BRAF V600-Mutant Colorectal Cancer. J. Clin. Oncol. 2015, 33, 4023–4031. [Google Scholar] [CrossRef]
  171. Roberts, S.K.; Galgadas, I.; Clarke, D.T.; Zanetti-Domingues, L.C.; Gervasio, F.L.; Martin-Fernandez, M.L. Targeting Mutant EGFR in Non-Small Cell Lung Cancer in the Context of Cell Adaptation and Resistance. Drug Discov. Today 2025, 30, 104407. [Google Scholar] [CrossRef]
  172. Liu, S.; Zou, Q.; Chen, J.P.; Yao, X.; Guan, P.; Liang, W.; Deng, P.; Lai, X.; Yin, J.; Chen, J.; et al. Targeting Enhancer Reprogramming to Mitigate MEK Inhibitor Resistance in Preclinical Models of Advanced Ovarian Cancer. J. Clin. Investig. 2021, 131, e145035. [Google Scholar] [CrossRef] [PubMed]
  173. Sigaud, R.; Stefanski, A.; Selt, F.; Kocher, D.; Usta, D.; Picard, D.; Büdenbender, I.; Remke, M.; Pfister, S.M.; Jones, D.T.W.; et al. Multi-Omics Dissection of MAPK-Driven Senescence Unveils Therapeutic Vulnerabilities in KIAA1549::BRAF-Fusion Pediatric Low-Grade Glioma Models. Signal Transduct. Target. Ther. 2025, 10, 197. [Google Scholar] [CrossRef]
  174. Yang, S.; Zhang, S.; Wang, J.; Zhang, N.; Wang, J.; Jiang, M.; Meng, D.; Xie, J.; Chang, X.; Cheng, H.; et al. Case Report: Successful Use of MEK Inhibitors as an Adjuvant Approach in the Treatment of Pediatric MAP4-RAF1 Fusion-Positive Solid Tumor. NPJ Precis. Oncol. 2025, 9, 201. [Google Scholar] [CrossRef]
  175. Hernando-Calvo, A.; Kurzrock, R.; Gonzalez, N.S.; Magidi, S.; Bresson, C.; Wunder, F.; Pretelli, G.; Casado, A.M.; El-Deiry, W.S. Case Report WIN-MTB-2023001 WIN International Molecular Tumor Board: A 62-Year-Old Male with Metastatic Colorectal Cancer with 5 Prior Lines of Treatment. Oncotarget 2025, 16, 456–466. [Google Scholar] [CrossRef]
  176. Hartman, M.L.; Sztiller-Sikorska, M.; Czyz, M. Synergistic Activity of S63845 and Parthenolide to Overcome Acquired Resistance to MEK1/2 Inhibitor in Melanoma Cells: Mechanisms and Therapeutic Potential. Biomed. Pharmacother. 2025, 188, 118183. [Google Scholar] [CrossRef]
  177. Gu, Q.; Wang, K.; Lu, T.; Xiao, Y.; Wu, Y.; Zhou, H.; Zhou, K. Single-Cell and Spatial Transcriptome Analyses Reveal MAZ+ NPC-like Clusters as Key Role Contributing to Glioma Recurrence and Drug Resistance. J. Transl. Med. 2025, 23, 657. [Google Scholar] [CrossRef] [PubMed]
  178. Gilmartin, A.G.; Bleam, M.R.; Groy, A.; Moss, K.G.; Stewart, A.E.; Dicker, D.T.; Hollis, A.; McMahon, G.; LoRusso, P.M.; Owens, T.D.; et al. GSK1120212 (JTP-74057) is an inhibitor of MEK activity and activation with favorable pharmacokinetic properties for sustained in vivo pathway inhibition. Clin. Cancer Res. 2011, 17, 989–1000. [Google Scholar] [CrossRef]
  179. Yaeger, R.; Corcoran, R.B. Targeting alterations in the RAF–MEK pathway. Cancer Discov. 2019, 9, 329–341. [Google Scholar] [CrossRef] [PubMed]
  180. Ahronian, L.G.; Sennott, E.M.; Van Allen, E.M.; Wagle, N.; Whittaker, S.; Montesion, M.; Ducar, M.; Margolis, C.A.; Greenberg, M.; Godfrey, J.T.; et al. Clinical acquired resistance to RAF inhibitor combinations in BRAF-mutant colorectal cancer through MAPK pathway alterations. Cancer Discov. 2015, 5, 358–367. [Google Scholar] [CrossRef]
  181. King, J.W.; Nathan, P.D. Role of the MEK Inhibitor Trametinib in the Treatment of Metastatic Melanoma. Future Oncol. 2014, 10, 1559–1570. [Google Scholar] [CrossRef]
  182. Barrow, C.; Browning, J.; MacGregor, D.; Davis, I.D.; Sturrock, S.; Jungbluth, A.A.; Cebon, J. Tumor antigen expression in melanoma varies according to antigen and stage. Clin. Cancer Res. 2006, 12, 764–771. [Google Scholar] [CrossRef]
  183. Teixidó, C.; Castillo, P.; Martinez-Vila, C.; Arance, A.; Alós, L. Molecular Markers and Targets in Melanoma. Cells 2021, 10, 2320. [Google Scholar] [CrossRef]
  184. Druker, B.J.; Talpaz, M.; Resta, D.J.; Peng, B.; Buchdunger, E.; Ford, J.M.; Lydon, N.B.; Kantarjian, H.; Capdeville, R.; Ohno-Jones, S.; et al. Efficacy and safety of a specific inhibitor of the BCR-ABL tyrosine kinase in chronic myeloid leukemia. N. Engl. J. Med. 2001, 344, 1031–1037. [Google Scholar] [CrossRef]
  185. Heinrich, M.C.; Corless, C.L.; Duensing, A.; McGreevey, L.; Chen, C.J.; Joseph, N.; Singer, S.; Griffith, D.J.; Haley, A.; Town, A.; et al. PDGFRA activating mutations in gastrointestinal stromal tumors. Science 2003, 299, 708–710. [Google Scholar] [CrossRef] [PubMed]
  186. Carofiglio, F.; Lopalco, A.; Lopedota, A.; Cutrignelli, A.; Nicolotti, O.; Denora, N.; Stefanachi, A.; Leonetti, F. Bcr-Abl Tyrosine Kinase Inhibitors in the Treatment of Pediatric CML. Int. J. Mol. Sci. 2020, 21, 4469. [Google Scholar] [CrossRef] [PubMed]
  187. Buchdunger, E.; Cioffi, C.L.; Law, N.; Stover, D.; Ohno-Jones, S.; Druker, B.J.; Lydon, N.B. Abl protein-tyrosine kinase inhibitor STI571 inhibits in vitro signal transduction mediated by c-kit and platelet-derived growth factor receptors. J. Pharmacol. Exp. Ther. 2000, 295, 139–145. [Google Scholar] [CrossRef] [PubMed]
  188. Weisberg, E.; Manley, P.W.; Breitenstein, W.; Bruggen, J.; Cowan-Jacob, S.W.; Ray, A.; Huntly, B.J.P.; Fabbro, D.; Fendrich, G.; Hall-Meyers, E.; et al. Characterization of AMN107, a selective inhibitor of native and mutant Bcr-Abl. Cancer Cell 2005, 7, 129–141. [Google Scholar] [CrossRef]
  189. Mittal, B.; Tulsyan, S.; Kumar, S.; Mittal, R.D.; Agarwal, G. Cytochrome P450 in Cancer Susceptibility and Treatment. Adv. Clin. Chem. 2015, 71, 77–139. [Google Scholar] [CrossRef]
  190. Rowley, J.D. A new consistent chromosomal abnormality in chronic myelogenous leukemia identified by quinacrine fluorescence and Giemsa staining. Nature 1973, 243, 290–293. [Google Scholar] [CrossRef]
  191. Nowell, P.C. The Minute Chromosome (Phl) in Chronic Granulocytic Leukemia. Blut 1962, 8, 65–66. [Google Scholar] [CrossRef] [PubMed]
  192. Warfvinge, R.; Geironson Ulfsson, L.; Dhapola, P.; Safi, F.; Sommarin, M.; Soneji, S.; Hjorth-Hansen, H.; Mustjoki, S.; Richter, J.; Thakur, R.K.; et al. Single-Cell Multiomics Analysis of Chronic Myeloid Leukemia Links Cellular Heterogeneity to Therapy Response. eLife 2024, 12, RP92074. [Google Scholar] [CrossRef]
  193. O’Brien, S.G.; Guilhot, F.; Larson, R.A.; Gathmann, I.; Baccarani, M.; Cervantes, F.; Cornelissen, J.J.; Fischer, T.; Hochhaus, A.; Hughes, T.; et al. Imatinib compared with interferon and low-dose cytarabine for newly diagnosed chronic-phase chronic myeloid leukemia. N. Engl. J. Med. 2003, 348, 994–1004. [Google Scholar] [CrossRef]
  194. Jackson, S.E.; Chester, J.D. Personalised cancer medicine. Int. J. Cancer 2015, 137, 262–266. [Google Scholar] [CrossRef] [PubMed]
  195. Kantarjian, H.; Giles, F.; Bhalla, K.; O’Brien, S.; Cortes, J.; Rios, M.B.; Shan, J.; Thomas, D.; Wierda, W.; Faderl, S.; et al. Nilotinib in imatinib-resistant CML and Philadelphia chromosome-positive acute lymphoblastic leukemia. N. Engl. J. Med. 2006, 354, 2542–2551. [Google Scholar] [CrossRef]
  196. Demetri, G.D.; von Mehren, M.; Blanke, C.D.; Van den Abbeele, A.D.; Eisenberg, B.; Roberts, P.J.; Heinrich, M.C.; Tuveson, D.A.; Singer, S.; Janicek, M.; et al. Efficacy and safety of imatinib mesylate in advanced gastrointestinal stromal tumors. N. Engl. J. Med. 2002, 347, 472–480. [Google Scholar] [CrossRef]
  197. Verweij, J.; Casali, P.G.; Zalcberg, J.; Le Cesne, A.; Reichardt, P.; Blay, J.Y.; Issels, R.; van Oosterom, A.T.; Hogendoorn, P.C.; Dimitrijevic, S.; et al. Progression-free survival in gastrointestinal stromal tumours with high-dose imatinib: Randomised trial. Lancet 2004, 364, 1127–1134. [Google Scholar] [CrossRef] [PubMed]
  198. Bello, D.M.; Dematteo, R.P.; Ariyan, C.E. The GIST of targeted therapy for malignant melanoma. Ann. Surg. Oncol. 2014, 21, 2059–2067. [Google Scholar] [CrossRef]
  199. Ben Ami, E.; Demetri, G.D. A Safety Evaluation of Imatinib Mesylate in the Treatment of Gastrointestinal Stromal Tumor. Expert Opin. Drug Saf. 2016, 15, 571–578. [Google Scholar] [CrossRef] [PubMed]
  200. Johnson, D.B.; Sosman, J.A. Update on the targeted therapy of melanoma. Curr. Treat. Options Oncol. 2013, 14, 280–292. [Google Scholar] [CrossRef]
  201. Bellouard, M.; Donadieu, J.; Thiebot, P.; Giroux Leprieur, E.; Saiag, P.; Etting, I.; Dugues, P.; Abe, E.; Alvarez, J.C.; Larabi, I.A. Validation of liquid chromatography coupled with tandem mass spectrometry for the determination of 12 tyrosine kinase inhibitors (TKIs) and their application to therapeutic drug monitoring in adult and pediatric populations. Pharmaceutics 2023, 16, 5. [Google Scholar] [CrossRef]
  202. Wang, X.; Zhou, Z.; Li, D.; Zhang, B.; Zong, X.; Liang, X.; Li, Z. The Molecular Mechanisms of Imatinib Treatment on Acute Lung Injury in Septic Mice through Proteomic Technology. J. Immunol. Res. 2025, 2025, 4526375. [Google Scholar] [CrossRef]
  203. Li, Q.; Liu, T.; Lv, K.; Liao, F.; Wang, J.; Tu, Y.; Chen, Q. Malaria: Past, Present, and Future. Signal Transduct. Target. Ther. 2025, 10, 188. [Google Scholar] [CrossRef]
  204. Roskoski, R., Jr. Cost in the United States of FDA-approved small molecule protein kinase inhibitors used in the treatment of neoplastic and non-neoplastic diseases. Pharmacol. Res. 2024, 199, 107036. [Google Scholar] [CrossRef]
  205. Sposito, M.; Belluomini, L.; Pontolillo, L.; Tregnago, D.; Trestini, I.; Insolda, J.; Avancini, A.; Milella, M.; Bria, E.; Carbognin, L.; et al. Adjuvant targeted therapy in solid cancers: Pioneers and new glories. J. Pers. Med. 2023, 13, 1427. [Google Scholar] [CrossRef] [PubMed]
  206. Bouraqqadi, O.; Elloudi, S.; Hammas, N.; Boubekri, N.; Mernissi, F. Rapidly Evolving Cervical Dermatofibrosarcoma Protuberans with Deep Muscle Infiltration: A Rare and Aggressive Presentation. Cureus 2025, 17, e84596. [Google Scholar] [CrossRef]
  207. Meisenheimer, J.; Usovich, M.; Rypka, K.J.; Liu, L.; Warshaw, E.; Gravely, A.; Lynch, J.; Westanmo, A.D.; Goldfarb, N. Variations in Anatomic Locations of Squamous Cell Carcinoma Stratified by Histologic Risk: A Single-Center Retrospective Study. Int. J. Dermatol. 2025. [Google Scholar] [CrossRef]
  208. Curtin, J.A.; Busam, K.; Pinkel, D.; Bastian, B.C. Somatic activation of KIT in distinct subtypes of melanoma. J. Clin. Oncol. 2006, 24, 4340–4346. [Google Scholar] [CrossRef] [PubMed]
  209. Delyon, J.; Lebbe, C.; Dumaz, N. Targeted therapies in melanoma beyond BRAF: Targeting NRAS-mutated and KIT-mutated melanoma. Curr. Opin. Oncol. 2020, 32, 79–84. [Google Scholar] [CrossRef]
  210. Berger, M.; Richtig, G.; Kashofer, K.; Aigelsreiter, A.; Richtig, E. The window of opportunities for targeted therapy in BRAFwt/NRASwt/KITwt melanoma: Biology and clinical implications of fusion proteins and other mutations. G. Ital. Dermatol. Venereol. 2018, 153, 349–360. [Google Scholar] [CrossRef] [PubMed]
  211. Spencer, K.R.; Mehnert, J.M. Mucosal Melanoma: Epidemiology, Biology and Treatment. Cancer Treat. Res. 2016, 167, 295–320. [Google Scholar] [CrossRef]
  212. Wei, X.; Mao, L.; Chi, Z.; Sheng, X.; Cui, C.; Kong, Y.; Dai, J.; Wang, X.; Li, S.; Tang, B.; et al. Efficacy evaluation of imatinib for the treatment of melanoma: Evidence from a retrospective study. Oncol. Res. 2019, 27, 495–501. [Google Scholar] [CrossRef]
  213. Spagnolo, F.; Caltabiano, G.; Queirolo, P. Uveal melanoma. Cancer Treat. Rev. 2012, 38, 549–553. [Google Scholar] [CrossRef]
  214. Dummer, R.; Rozati, S.; Eggmann, N.; Rinderknecht, J.; Goldinger, S.M. From chemotherapy to targeted treatment. Ann. Oncol. 2012, 23 (Suppl. 10), x101–x103. [Google Scholar] [CrossRef]
  215. Johnson, D.B.; Pollack, M.H.; Sosman, J.A. Emerging targeted therapies for melanoma. Expert Opin. Emerg. Drugs 2016, 21, 195–207. [Google Scholar] [CrossRef]
  216. Stoff, R.; Markovic, S.N.; McWilliams, R.R.; Kottschade, L.A.; Montane, H.N.; Dimou, A.; Dudek, A.Z.; Tan, W.; Dronca, R.S.; Seetharam, M.; et al. Real-world evidence on efficacy and toxicity of targeted therapy in older melanoma patients treated in a tertiary-hospital setting. Melanoma Res. 2024, 34, 510–518. [Google Scholar] [CrossRef] [PubMed]
  217. Gorre, M.E.; Mohammed, M.; Ellwood, K.; Hsu, N.; Paquette, R.; Rao, P.N.; Sawyers, C.L. Clinical resistance to STI-571 cancer therapy caused by BCR-ABL gene mutation or amplification. Science 2001, 293, 876–880. [Google Scholar] [CrossRef]
  218. Shah, N.P.; Nicoll, J.M.; Nagar, B.; Gorre, M.E.; Paquette, R.; Kuriyan, J.; Sawyers, C.L. Multiple BCR-ABL kinase domain mutations confer polyclonal resistance to the tyrosine kinase inhibitor imatinib (STI571) in chronic phase and blast crisis chronic myeloid leukemia. Cancer Cell 2002, 2, 117–125. [Google Scholar] [CrossRef]
  219. Tsubaki, M.; Obana, T.; Matsuo, T.; Komori, R.; Takeda, T.; Koumoto, Y.; Usami, H.; Nagai, N.; Nishida, S. Overexpression of miR-29a and miR-29b Is Involved in Imatinib Resistance via Abrogated NF1 Expression and Increased ERK1/2 Activation in Chronic Myeloid Leukemia Cells. Med. Oncol. 2025, 42, 268. [Google Scholar] [CrossRef] [PubMed]
  220. Ghadyani Nejhad, L.; Sohani, M.; Ghandforoush, N.A.; Nikbakht, M.; Mohammadi, S.; Vaezi, M.; Rostami, S.; Chahardouli, B. Non-Coding RNAs: Emerging Contributors to Chemoresistance in Chronic Myeloid Leukemia. Leuk. Res. Rep. 2025, 23, 100513. [Google Scholar] [CrossRef]
  221. Kantarjian, H.; Sawyers, C.L.; Hochhaus, A.; Guilhot, F.; Schiffer, C.A.; Gambacorti-Passerini, C.; Niederwieser, D.; Rousselot, P.; Ottmann, O.G.; Pasquini, R.; et al. Hematologic and cytogenetic responses to imatinib mesylate in chronic myelogenous leukemia. N. Engl. J. Med. 2002, 346, 645–652. [Google Scholar] [CrossRef] [PubMed]
  222. Cortes, J.E.; Kim, D.W.; Pinilla-Ibarz, J.; le Coutre, P.; Paquette, R.; Chuah, C.; Nicolini, F.E.; Apperley, J.F.; Khoury, H.J.; Talpaz, M.; et al. Ponatinib efficacy and safety in Philadelphia chromosome-positive leukemia: Final results of the phase 2 PACE trial. Blood 2018, 132, 393–404. [Google Scholar] [CrossRef] [PubMed]
  223. Zhou, S.; Abdihamid, O.; Tan, F.; Zhou, H.; Liu, H.; Li, Z.; Xiao, S.; Li, B. KIT Mutations and Expression: Current Knowledge and New Insights for Overcoming IM Resistance in GIST. Cell Commun. Signal. 2024, 22, 153. [Google Scholar] [CrossRef]
  224. Liu, X.; Yu, J.; Li, Y.; Shi, H.; Jiao, X.; Liu, X.; Guo, D.; Li, Z.; Tian, Y.; Dai, F.; et al. Deciphering the Tumor Immune Microenvironment of Imatinib-Resistance in Advanced Gastrointestinal Stromal Tumors at Single-Cell Resolution. Cell Death Dis. 2024, 15, 190. [Google Scholar] [CrossRef]
  225. Gambacorti-Passerini, C.; Antolini, L.; Mahon, F.X.; Guilhot, J.; Deininger, M.; Niederwieser, D.; Soverini, S.; Hochhaus, A.; Hughes, T.P.; Clark, R.E.; et al. Multicenter independent assessment of outcomes in chronic myeloid leukemia patients treated with imatinib. J. Natl. Cancer Inst. 2011, 103, 553–561. [Google Scholar] [CrossRef]
  226. Kantarjian, H.; Shah, N.; Hochhaus, A.; Cortes, J.; Shah, N.P.; Tanaka, C.; Giles, F.; Boque, C.; O’Brien, S.; Jorgensen, J.; et al. Dasatinib or imatinib in newly diagnosed chronic-phase chronic myeloid leukemia: 2-Year follow-up from a randomized phase 3 trial (DASISION). Blood 2012, 119, 1123–1129. [Google Scholar] [CrossRef] [PubMed]
  227. Steegmann, J.L.; Baccarani, M.; Breccia, M.; Casado, L.F.; Garcia-Gutierrez, V.; Gratwohl, A.; Huguet, F.; le Coutre, P.; Mahon, F.X.; Nicolini, F.E.; et al. European LeukemiaNet recommendations for the management and avoidance of adverse events of treatment in chronic myeloid leukemia. Leukemia 2016, 30, 1648–1671. [Google Scholar] [CrossRef] [PubMed]
  228. Quintás-Cardama, A.; Cortes, J. Chronic myeloid leukemia: Diagnosis and treatment. Mayo Clin. Proc. 2006, 81, 973–988. [Google Scholar] [CrossRef]
  229. Hughes, T.P.; Saglio, G.; Kantarjian, H.M.; Guilhot, F.; Niederwieser, D.; Rosti, G.; Nakaseko, C.; De Souza, C.A.; Kalaycio, M.E.; Meier, S.; et al. Early Molecular Response Predicts Outcomes in Patients with Chronic Myeloid Leukemia in Chronic Phase Treated with Frontline Nilotinib or Imatinib. Blood 2014, 123, 1353–1360. [Google Scholar] [CrossRef]
  230. D’Angelo, S.P.; Shoushtari, A.N.; Keohan, M.L.; Dickson, M.A.; Gounder, M.M.; Chi, P.; Loo, J.K.; Gaffney, L.; Schneider, L.; Patel, Z.; et al. Combined KIT and CTLA-4 Blockade in Patients with Refractory GIST and Other Advanced Sarcomas: A Phase Ib Study of Dasatinib plus Ipilimumab. Clin. Cancer Res. 2017, 23, 2972–2980. [Google Scholar] [CrossRef]
  231. Soria, J.C.; Mauguen, A.; Reck, M.; Hofman, P.; Osório, A.; Lena, H.; Jackman, D.M.; Sequist, L.V.; Mahé, M.; Riely, G.J.; et al. Osimertinib in untreated EGFR-mutated advanced non–small-cell lung cancer. N. Engl. J. Med. 2018, 378, 113–125. [Google Scholar] [CrossRef]
  232. Yao, Z.; Lu, L.; Xu, Q.; Hua, S.; Wang, H.; Jiang, H. Integrated analysis reveals COL4A3 as a novel diagnostic and therapeutic target in UV-related skin cutaneous melanoma. Clin. Cosmet. Investig. Dermatol. 2024, 17, 1429–1446. [Google Scholar] [CrossRef]
  233. Pham, T.M.; Ahmed, M.; Lai, T.H.; Bahar, M.E.; Hwang, J.S.; Maulidi, R.F.; Ngo, Q.N.; Kim, D.R. Regulation of cell cycle progression through RB phosphorylation by nilotinib and AT-9283 in human melanoma A375P cells. Int. J. Mol. Sci. 2024, 25, 2956. [Google Scholar] [CrossRef] [PubMed]
  234. Han, X.; Ge, P.; Liu, S.; Yang, D.; Zhang, J.; Wang, X.; Liang, W. Efficacy and Safety of Bevacizumab in Patients with Malignant Melanoma: A Systematic Review and PRISMA-Compliant Meta-Analysis of Randomized Controlled Trials and Non-Comparative Clinical Studies. Front. Pharmacol. 2023, 14, 1163805. [Google Scholar] [CrossRef]
  235. Peters, J.; Bollaert, E.; Cloos, A.S.; Claus, M.; Essaghir, A.; Lenglez, S.; Saussoy, P.; Dachy, G.; Autin, P.; Demoulin, J.B.; et al. MiR-92a-1-5p Contributes to Cellular Proliferation and Survival in Chronic Myeloid Leukemia and Its Inhibition Enhances Imatinib Efficacy. Noncoding RNA Res. 2025, 14, 14–24. [Google Scholar] [CrossRef] [PubMed]
  236. Ishida, T.; Takahashi, T.; Kurokawa, Y.; Nishida, T.; Hirota, S.; Serada, S.; Fujimoto, M.; Naka, T.; Teranishi, R.; Saito, T.; et al. Targeted Therapy for Drug-Tolerant Persister Cells after Imatinib Treatment for Gastrointestinal Stromal Tumours. Br. J. Cancer 2021, 125, 1511–1522. [Google Scholar] [CrossRef]
  237. Kantarjian, H.M.; Hughes, T.P.; Larson, R.A.; Kim, D.W.; Issaragrisil, S.; le Coutre, P.; Etienne, G.; Boquimpani, C.; Pasquini, R.; Clark, R.E.; et al. Long-Term Outcomes with Frontline Nilotinib versus Imatinib in Newly Diagnosed Chronic Myeloid Leukemia in Chronic Phase: ENESTnd 10-Year Analysis. Leukemia 2021, 35, 440–453. [Google Scholar] [CrossRef]
  238. Liu, W.Z.; Du, Y.Q.; Shen, Q.; Tao, K.X.; Zhang, P. Ripretinib for the Treatment of Advanced, Imatinib-Resistant Gastrointestinal Stromal Tumors. J. Dig. Dis. 2024, 25, 559–563. [Google Scholar] [CrossRef]
  239. Sun, X.; Zhang, Q.; Lin, X.; Shu, P.; Gao, X.; Shen, K. Imatinib Induces Ferroptosis in Gastrointestinal Stromal Tumors by Promoting STUB1-Mediated GPX4 Ubiquitination. Cell Death Dis. 2023, 14, 839. [Google Scholar] [CrossRef]
  240. Szucs, Z.; Thway, K.; Fisher, C.; Bulusu, R.; Constantinidou, A.; Benson, C.; van der Graaf, W.T.; Jones, R.L. Promising Novel Therapeutic Approaches in the Management of Gastrointestinal Stromal Tumors. Future Oncol. 2017, 13, 185–194. [Google Scholar] [CrossRef] [PubMed]
  241. O’Hare, T.; Eide, C.A.; Deininger, M.W.N. BCR-ABL kinase domain mutations, drug resistance, and the road to a cure for chronic myeloid leukemia. Blood 2007, 110, 2242–2249. [Google Scholar] [CrossRef]
  242. Sawyers, C.L. Targeted cancer therapy. Nature 2004, 432, 294–297. [Google Scholar] [CrossRef]
  243. Druker, B.J. Translation of the Philadelphia chromosome into therapy for CML. Blood 2008, 112, 4808–4817. [Google Scholar] [CrossRef]
  244. Cohen, P.; Cross, D.; Jänne, P.A. Kinase drug discovery 20 years after imatinib: Progress and future directions. Nat. Rev. Drug Discov. 2021, 20, 551–569. [Google Scholar] [CrossRef] [PubMed]
  245. Lee, N.; Kim, K.H.; Lee, S.-C. Oral chemotherapeutic agents in current use. J. Korean Med. Assoc. 2011, 54, 1191–1198. [Google Scholar] [CrossRef]
  246. Amouei, A.; Daeian, N.; Khezrnia, S.S.; Mansouri, A.; Hadjibabaie, M. Imatinib efficacy, safety and resistance in Iranian patients with chronic myeloid leukemia: A review of literature. Int. J. Hematol. Oncol. Stem Cell Res. 2021, 15, 114–131. [Google Scholar] [CrossRef] [PubMed]
  247. Zhong, J.; Yan, W.; Wang, C.; Liu, W.; Lin, X.; Zou, Z.; Sun, W.; Chen, Y. BRAF Inhibitor Resistance in Melanoma: Mechanisms and Alternative Therapeutic Strategies. Curr. Treat. Options Oncol. 2022, 23, 1503–1521. [Google Scholar] [CrossRef]
  248. Luebker, S.A.; Koepsell, S.A. Diverse Mechanisms of BRAF Inhibitor Resistance in Melanoma Identified in Clinical and Preclinical Studies. Front. Oncol. 2019, 9, 268. [Google Scholar] [CrossRef]
  249. Paton, E.L.; Turner, J.A.; Schlaepfer, I.R. Overcoming Resistance to Therapies Targeting the MAPK Pathway in BRAF-Mutated Tumours. J. Oncol. 2020, 2020, 1079827. [Google Scholar] [CrossRef]
  250. Lelliott, E.J.; McArthur, G.A.; Oliaro, J.; Sheppard, K.E. Immunomodulatory Effects of BRAF, MEK, and CDK4/6 Inhibitors: Implications for Combining Targeted Therapy and Immune Checkpoint Blockade for the Treatment of Melanoma. Front. Immunol. 2021, 12, 661737. [Google Scholar] [CrossRef]
  251. Idris, O.A.; Shebrain, A.; Jawad, A.; Pacione, S.C.; Haj, D.; Bzizi, H.; Ahmedfiqi, Y.O.; Saadaie Jahromi, B.; Deleon, N.; Zhang, T.; et al. Breaking Barriers: Pembrolizumab’s Role in Overcoming Targeted Therapy Resistance in BRAF-Mutant Melanoma. Clin. Surg. Oncol. 2025, 4, 100095. [Google Scholar] [CrossRef]
  252. Buchanan, T.; Amouzegar, A.; Luke, J.J. Next-Generation Immunotherapy Approaches in Melanoma. Curr. Oncol. Rep. 2021, 23, 116. [Google Scholar] [CrossRef]
  253. Zoroddu, S.; Bagella, L. Next-Generation mRNA Vaccines in Melanoma: Advances in Delivery and Combination Strategies. Cells 2025, 14, 1476. [Google Scholar] [CrossRef]
  254. Slusher, N.; Jones, N.; Nonaka, T. Liquid Biopsy for Diagnostic and Prognostic Evaluation of Melanoma. Front. Cell Dev. Biol. 2024, 12, 1420360. [Google Scholar] [CrossRef] [PubMed]
  255. Connor, C.; Carr, Q.L.; Sweazy, A.; McMasters, K.; Hao, H. Clinical Approaches for the Management of Skin Cancer: A Review of Current Progress in Diagnosis, Treatment, and Prognosis for Patients with Melanoma. Cancers 2025, 17, 707. [Google Scholar] [CrossRef]
  256. Song, Y.; Bi, Z.; Liu, Y.; Qin, F.; Wei, Y.; Wei, X. Targeting RAS-RAF-MEK-ERK Signaling Pathway in Human Cancer: Current Status in Clinical Trials. Genes Dis. 2022, 10, 76–88. [Google Scholar] [CrossRef] [PubMed]
  257. 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]
  258. Huang, C.-Y.; Huang, W.-K.; Yeh, K.-Y.; Chang, J.W.-C.; Lin, Y.-C.; Chou, W.-C. Integrating Comprehensive Genomic Profiling in the Management of Oncology Patients: Applications and Challenges in Taiwan. Biomed. J. 2025, 48, 100851. [Google Scholar] [CrossRef]
  259. Nero, C.; Duranti, S.; Giacomini, F.; Minucci, A.; Giacò, L.; Piermattei, A.; Genuardi, M.; Pasciuto, T.; Urbani, A.; Daniele, G.; et al. Integrating a Comprehensive Cancer Genome Profiling into Clinical Practice: A Blueprint in an Italian Referral Center. J. Pers. Med. 2022, 12, 1746. [Google Scholar] [CrossRef] [PubMed]
  260. Shih, Y.T.; Pan, I.W.; Teich, N. Global Challenges in Access to and Implementation of Precision Oncology: The Health Care Manager and Health Economist Perspective. ASCO Educ. Book 2022, 42, 429–437. [Google Scholar] [CrossRef]
Figure 1. Structural formula of vemurafenib.
Figure 1. Structural formula of vemurafenib.
Jcm 14 07906 g001
Figure 2. The mechanism of vemurafenib according to [85] involves targeting the MAPK/ERK signaling pathway, specifically inhibiting the BRAF V600E mutation—where valine (V) at position 600 is replaced by glutamic acid (E)—which causes continuous activation of the BRAF protein and promotes cancer growth; where: ERK—Extracellular Signal-Regulated Kinase, a protein kinase activated by MEK that controls cell division, survival, and differentiation; MEK—Mitogen-Activated Protein Kinase Kinase, an enzyme that activates ERK by phosphorylation and acts downstream of RAS and BRAF in the signaling pathway; RAS—a family of proteins (e.g., KRAS, NRAS, HRAS) that regulate cell growth and survival by transmitting signals from outside the cell to the nucleus; mutations can lead to cancer.
Figure 2. The mechanism of vemurafenib according to [85] involves targeting the MAPK/ERK signaling pathway, specifically inhibiting the BRAF V600E mutation—where valine (V) at position 600 is replaced by glutamic acid (E)—which causes continuous activation of the BRAF protein and promotes cancer growth; where: ERK—Extracellular Signal-Regulated Kinase, a protein kinase activated by MEK that controls cell division, survival, and differentiation; MEK—Mitogen-Activated Protein Kinase Kinase, an enzyme that activates ERK by phosphorylation and acts downstream of RAS and BRAF in the signaling pathway; RAS—a family of proteins (e.g., KRAS, NRAS, HRAS) that regulate cell growth and survival by transmitting signals from outside the cell to the nucleus; mutations can lead to cancer.
Jcm 14 07906 g002
Figure 3. Structural formula of trametinib.
Figure 3. Structural formula of trametinib.
Jcm 14 07906 g003
Figure 4. Mechanism of trametinib according to [136] involves inhibition of the BRAF–MEK1/2–ERK1/2 signaling pathway, which is activated when RAS, a small GTPase, switches from an inactive GDP-bound form to an active GTP-bound form, initiating a phosphorylation cascade wherein mutated BRAF (e.g., V600E) activates MEK, which subsequently phosphorylates and activates ERK, ultimately promoting metabolic reprogramming, cell growth, proliferation, and differentiation; where: BRAF—a protein kinase; MEK—Mitogen-Activated Protein Kinase Kinase; ERK—Extracellular Signal-Regulated Kinase; RAS—a small GTPase; GAP—GTPase-activating protein; GEF—Guanine nucleotide exchange factor; GDP—guanosine diphosphate; GTP—guanosine triphosphate.
Figure 4. Mechanism of trametinib according to [136] involves inhibition of the BRAF–MEK1/2–ERK1/2 signaling pathway, which is activated when RAS, a small GTPase, switches from an inactive GDP-bound form to an active GTP-bound form, initiating a phosphorylation cascade wherein mutated BRAF (e.g., V600E) activates MEK, which subsequently phosphorylates and activates ERK, ultimately promoting metabolic reprogramming, cell growth, proliferation, and differentiation; where: BRAF—a protein kinase; MEK—Mitogen-Activated Protein Kinase Kinase; ERK—Extracellular Signal-Regulated Kinase; RAS—a small GTPase; GAP—GTPase-activating protein; GEF—Guanine nucleotide exchange factor; GDP—guanosine diphosphate; GTP—guanosine triphosphate.
Jcm 14 07906 g004
Figure 5. Structural formula of imatinib.
Figure 5. Structural formula of imatinib.
Jcm 14 07906 g005
Figure 6. The mechanism of imatinib according to [245] involves inhibition of the BCR-ABL fusion protein, a constitutively active tyrosine kinase, by occupying its ATP-binding site; this prevents substrate phosphorylation, disrupts downstream effector interactions, interrupts the signal transduction pathway, and ultimately blocks oncogenic signaling, halts uncontrolled cell proliferation, and induces apoptosis, where: ATP—adenosine triphosphate; P—phosphate.
Figure 6. The mechanism of imatinib according to [245] involves inhibition of the BCR-ABL fusion protein, a constitutively active tyrosine kinase, by occupying its ATP-binding site; this prevents substrate phosphorylation, disrupts downstream effector interactions, interrupts the signal transduction pathway, and ultimately blocks oncogenic signaling, halts uncontrolled cell proliferation, and induces apoptosis, where: ATP—adenosine triphosphate; P—phosphate.
Jcm 14 07906 g006
Table 1. Approved BRAF, MEK, and KIT inhibitors for melanoma according to [15,16,17,23,45]. All abbreviations employed are defined in the text in the Abbreviations section.
Table 1. Approved BRAF, MEK, and KIT inhibitors for melanoma according to [15,16,17,23,45]. All abbreviations employed are defined in the text in the Abbreviations section.
Class/
Agent
Key TargetsPivotal TrialsORR (%)Median PFS (mo)Median OS (mo)Common AEsDistinct Features/
Resistance Notes
Vemurafenib (Zelboraf)BRAF V600E/KBRIM-3486.913.6Photosensitivity, arthralgiaFirst-in-class; higher cutaneous toxicity; paradoxical MAPK activation
Dabrafenib (Tafinlar)BRAF V600E/KBREAK-3519.318.2Pyrexia, fatigueLower rash/photosensitivity; improved tolerability
Encorafenib (Braftovi)BRAF V600E/KCOLUMBUS6014.933.6Arthralgia, fatigueLongest half-life; least paradoxical activation
Trametinib (Mekinist)MEK1/2METRIC224.815.6Rash, diarrheaAlone or combined with dabrafenib
Cobimetinib (Cotellic)MEK1coBRIM— (combo)12.3 22.3 Diarrhea, photosensitivityUsed with vemurafenib
Binimetinib (Mektovi)MEK1/2COLUMBUS— (combo)14.9 33.6 Fatigue, nauseaUsed with encorafenib
Imatinib (Gleevec)KIT (L576P, K642E)Phase II16–293–712–18Edema, nauseaBenefit limited to activating mutations; not amplifications
Nilotinib (Tasigna)KIT (L576P, K642E)Phase II20–263–6Fatigue, cytopeniaMay retain efficacy post-imatinib; off-label use
Table 2. Melanoma Treatment Strategies—overview by modality according to [14,72,74], where: BRAF—v-Raf murine sarcoma viral oncogene homolog B; CTLA-4—Cytotoxic T-lymphocyte–associated protein 4; HSV—Herpes simplex virus; IFN-γ—Interferon gamma; LDH—Lactate dehydrogenase; MAPK—Mitogen-activated protein kinase; MEK—MAPK/ERK kinase; MHC—Major histocompatibility complex; NGS—Next-generation sequencing; NRAS—Neuroblastoma RAS viral oncogene homolog; PD-1—Programmed cell death protein 1; PD-L1—Programmed death-ligand 1; PCR—Polymerase chain reaction; QT—QT interval, RTK—Receptor tyrosine kinase; SLNB—Sentinel lymph node biopsy; TME—Tumor microenvironment; TMB—Tumor mutational burden.
Table 2. Melanoma Treatment Strategies—overview by modality according to [14,72,74], where: BRAF—v-Raf murine sarcoma viral oncogene homolog B; CTLA-4—Cytotoxic T-lymphocyte–associated protein 4; HSV—Herpes simplex virus; IFN-γ—Interferon gamma; LDH—Lactate dehydrogenase; MAPK—Mitogen-activated protein kinase; MEK—MAPK/ERK kinase; MHC—Major histocompatibility complex; NGS—Next-generation sequencing; NRAS—Neuroblastoma RAS viral oncogene homolog; PD-1—Programmed cell death protein 1; PD-L1—Programmed death-ligand 1; PCR—Polymerase chain reaction; QT—QT interval, RTK—Receptor tyrosine kinase; SLNB—Sentinel lymph node biopsy; TME—Tumor microenvironment; TMB—Tumor mutational burden.
Treatment TypeDrug/
Class
Molecular Target/
Mechanism
IndicationsBiomarkersCombination StrategiesCommon Resistance MechanismsKey Side Effects
(Including Severe/Fatal)
Targeted TherapyVemurafenib, DabrafenibBRAF V600E/K mutation,
inhibits MAPK pathway at BRAF
BRAF V600+ advanced or metastatic melanomaBRAF V600 mutation (PCR/NGS)With MEK inhibitors (e.g., Trametinib, Cobimetinib)BRAF amplification, NRAS mutation, MAPK reactivationRash, arthralgia, QT prolongation (potentially fatal arrhythmia), photosensitivity, cutaneous squamous cell carcinoma (severe)
Trametinib, CobimetinibMEK1/2 inhibition,
downstream of BRAF
BRAF V600+ (always combined with BRAF inhibitors)BRAF mutationWith BRAF inhibitors to avoid resistanceMEK/ERK mutations, RTK upregulationDiarrhea, fatigue, cardiac toxicity (heart failure, rare fatal), retinopathy, interstitial lung disease (rare severe)
KIT inhibitors (Imatinib, Nilotinib)KIT mutations or amplifications (rare subtype of melanoma)Acral, mucosal, or chronically sun-damaged melanomaKIT exon 11/13 mutationRarely used; sometimes with immunotherapy or chemotherapySecondary KIT mutationsEdema, cytopenias, nausea, hepatotoxicity, cardiac toxicity (rare fatal)
ImmunotherapyAnti–PD-1 (Nivolumab, Pembrolizumab)Blocks PD-1 receptor,
restores T-cell activity
Metastatic or unresectable melanoma; adjuvant settingPD-L1 expression (optional), TMB, IFN-γ signatureWith CTLA-4 inhibitors, targeted therapy, or chemoLoss of MHC expression, JAK/STAT mutations, T cell exhaustionImmune-related AEs: colitis, hepatitis, pneumonitis (can be severe/fatal)
Anti–CTLA-4 (Ipilimumab)Blocks CTLA-4,
enhances T-cell priming and activation
Metastatic or refractory melanomaNone specificWith PD-1 inhibitors (dual checkpoint blockade)Severe immune evasion, Treg expansionColitis, dermatitis, hypophysitis, enteritis (severe/fatal in rare cases)
Oncolytic TherapyTalimogene laherparepvec (T-VEC)Modified HSV-1 virus that replicates in tumors and expresses GM-CSFUnresectable stage IIIB–IV melanoma (locoregional)HSV-seronegative status (relative)With PD-1 inhibitors (under investigation)Immunosuppressive tumor microenvironmentFlu-like symptoms, injection site pain (generally mild; rare systemic viral spread possible)
ChemotherapyDacarbazine, TemozolomideAlkylating agents causing DNA damagePreviously standard in advanced melanomaNone requiredRarely used; now replaced by immunotherapy/targeted agentsHigh toxicity, low response ratesMyelosuppression, nausea, fatigue (potentially severe infections)
RadiotherapyStereotactic, whole-brain, adjuvant RTDNA damage, p53-dependent apoptosisBrain metastases, palliationNone requiredWith checkpoint inhibitors (for synergy)Radioresistance (via DNA repair enzymes)Cognitive impairment, fatigue, dermatitis (rare severe necrosis)
SurgeryWide local excision, lymph node dissectionCurative in early-stage melanomaStage I–II and some stage IIITumor thickness, ulceration, SLNB resultsAdjuvant immunotherapy or radiotherapy in high-risk casesN/AWound complications, lymphedema, infection
Adjuvant/NeoadjuvantImmunotherapy or targeted therapySame as above—used to reduce recurrence risk or downstage tumorsStage III–IV resectable or high-risk patientsBRAF status, LDH, PD-L1 (optional)Nivolumab, Dabrafenib/Trametinib as standard optionsSimilar to primary therapiesDependent on regimen used (includes severe immune or cardiac events for targeted/immunotherapy)
PD-1/PD-L1 therapies are now first-line in most metastatic or unresectable melanomas. BRAF/MEK inhibitors are preferred for rapid tumor burden reduction in BRAF-mutated patients. Combination immunotherapy (PD-1 + CTLA-4) improves response but increases immune toxicity. T-VEC is limited to superficial lesions and is not systemic. Chemotherapy is largely obsolete unless other options fail or are contraindicated.
Table 3. RAF isoforms and mutations with associated cancer types and clinical relevance types according to [90]. All abbreviations employed are defined in the text in the Abbreviations section.
Table 3. RAF isoforms and mutations with associated cancer types and clinical relevance types according to [90]. All abbreviations employed are defined in the text in the Abbreviations section.
RAF Isoform/MutationCancer Type(s)Clinical Relevance
B-RAF V600EMelanoma, Colorectal, Thyroid (Papillary), Ovarian, NSCLCHighly oncogenic; Targetable with B-RAF and MEK inhibitors; In colorectal cancer, associated with poor prognosis and resistance to B-RAF inhibitor monotherapy
B-RAF Non-V600 Mutations (general)MelanomaTargetable with B-RAF and MEK inhibitors
B-RAF F595LMelanoma, Colorectal, Thyroid (Papillary), NSCLCWeak/intermediate kinase activity; sometimes respond better to MEK inhibitors rather than B-RAF inhibitors
B-RAF L597QMelanoma, Colorectal, Thyroid (Papillary), NSCLCWeak/intermediate kinase activity; sometimes respond better to MEK inhibitors rather than B-RAF inhibitors
B-RAF G469AMelanoma, Colorectal, Thyroid (Papillary), NSCLCWeak/intermediate kinase activity; sometimes respond better to MEK inhibitors rather than B-RAF inhibitors
B-RAF V600KHairy Cell LeukemiaHighly sensitive to B-RAF inhibition
C-RAF F133LMelanoma, ColorectalMay respond to B-RAF inhibitors
C-RAF S257LMelanoma, ColorectalMay respond to B-RAF inhibitors
A-RAF Rare MutationsColorectal, Ovarian, Lung, Pancreatic, GliomasLimited therapeutic targeting due to complexity of RAF dimerization; limited treatment options
Table 4. Comparison of vemurafenib, trametinib, and imatinib according to [12,132,196,246]. All abbreviations employed are defined in the text in the Abbreviations section.
Table 4. Comparison of vemurafenib, trametinib, and imatinib according to [12,132,196,246]. All abbreviations employed are defined in the text in the Abbreviations section.
FeatureVemurafenibTrametinibImatinib
Drug ClassBRAF inhibitor (ATP-competitive)MEK1/2 inhibitor (non-ATP competitive)Tyrosine kinase inhibitor (TKI)
Primary Molecular Target(s)Mutant BRAF V600EMEK1 and MEK2 (MAP2K1/2) downstream of BRAFBCR-ABL (CML, Ph+ ALL)
KIT (GIST)
PDGFRα/β (DFSP, HES)
Primary Signaling PathwayMAPK/ERK pathway (RAS–RAF–MEK–ERK)MAPK/ERK pathway (inhibits downstream of RAF, at MEK level)BCR-ABL
STAT/JAK
KIT/PDGFR
PI3K/AKT/mTOR
MAPK/ERK
Upstream ActivatorsRAS– Receptor tyrosine kinases (RTKs)BRAF (WT or mutant)
RAS mutations
RTKs
BCR-ABL fusion gene
SCF for KIT
PDGF for PDGFR
Direct ActionInhibits aberrant BRAF V600E kinase activity to block downstream MEK/ERK signalingInhibits MEK1/2 phosphorylation to prevent ERK activationInhibits phosphorylation of BCR-ABL, KIT, and PDGFR, blocking their downstream signal transduction
Downstream Effects↓ MEK1/2 and ERK1/2 phosphorylation
↓ cyclin D1
↓ proliferation
↑ apoptosis
↓ ERK phosphorylation
↓ proliferation
↑ apoptosis
↓ PI3K/AKT, MAPK, and JAK/STAT signaling
↓ proliferation
↑ apoptosis
Cellular ConsequencesG1-phase arrest
Decreased cell survival and angiogenesis
G1-phase arrest
Reduced tumor growth
Suppression of malignant hematopoietic cell growth
Long-term disease control in CML and GIST
Resistance MechanismsBRAF amplification
NRAS mutation
MAPK pathway reactivation
PI3K activation
MEK amplification
ERK mutations
Bypass via PI3K/AKT,
RTKs
BCR-ABL mutations (e.g., T315I)
Overexpression
KIT exon 17 mutations
Activation of bypass kinases
Combination StrategiesWith MEK inhibitors (e.g., trametinib)
With PI3K/mTOR inhibitors
Autophagy inhibitors (HCQ)
With BRAF inhibitors
With CDK4/6 inhibitors
Immunotherapy
2nd/3rd gen TKIs (dasatinib, ponatinib)
Allosteric BCR-ABL inhibitors (asciminib)
Epigenetic agents
Clinical BiomarkersBRAF V600E mutationBRAF V600E/K, NRAS (trial/experimental)BCR-ABL, KIT, PDGFRA mutations (PCR, FISH, NGS)
Median OS (months)13.6 (monotherapy);
up to 25 (with MEK inhibitor)
~11 (monotherapy);
up to 25 (combo)
~12
Median PFS (months)5.3 (monotherapy);
~12 (combo)
4–5 (mono);
11–12 (combo)
3.5–4
ORR (%)50–60 (mono);
~70 (combo)
20–25 (mono);
60–70 (combo)
30–50
Table 5. Adverse effects of vemurafenib, trametinib, and imatinib with frequency and severity [38,39,40,41,63], where—frequency categories: Very common (>30%), Common (10–30%), Uncommon (1–10%), Rare (<1%);—severity grading: Mild—manageable without dose change; Moderate—may require dose adjustment/supportive therapy; Severe/Fatal—life-threatening, may require therapy discontinuation. All abbreviations employed are defined in the text in the Abbreviations section.
Table 5. Adverse effects of vemurafenib, trametinib, and imatinib with frequency and severity [38,39,40,41,63], where—frequency categories: Very common (>30%), Common (10–30%), Uncommon (1–10%), Rare (<1%);—severity grading: Mild—manageable without dose change; Moderate—may require dose adjustment/supportive therapy; Severe/Fatal—life-threatening, may require therapy discontinuation. All abbreviations employed are defined in the text in the Abbreviations section.
Adverse EffectVemurafenibTrametinibImatinib
ArthralgiaVery common (>30%)—Mild–Moderate
RashVery common (>30%)—Mild–ModerateVery common (>30%)—Mild–ModerateCommon (10–30%)—Mild–Moderate
PhotosensitivityVery common (>30%)—Mild–Moderate
FatigueCommon (10–30%)—Mild–ModerateVery common (>30%)—Mild–ModerateCommon (10–30%)—Mild–Moderate
DiarrheaVery common (>30%)—Mild–ModerateCommon (10–30%)—Mild–Moderate
NauseaCommon (10–30%)—MildCommon (10–30%)—MildVery common (>30%)—Mild
VomitingCommon (10–30%)—Mild–Moderate
PruritusCommon (10–30%)—Mild–Moderate
AlopeciaCommon (10–30%)—Mild
Edema/Periorbital edemaCommon (10–30%)—Mild–ModerateVery common (>30%)—Mild–Moderate
HypertensionCommon (10–30%)—Moderate–Severe
Cutaneous squamous cell carcinoma/KeratoacanthomaCommon (10–30%)—Severe
Cardiotoxicity (heart failure, QT prolongation)Uncommon (1–10%)—Severe/Potentially fatalUncommon (1–10%)—SevereRare (<1%)—Severe/Potentially fatal
QT prolongationUncommon (1–10%)—Severe/Potentially fatalRare (<1%)—Severe/Potentially fatal
HepatotoxicityUncommon (1–10%)—SevereUncommon (1–10%)—Severe/Potentially fatal
Cardiomyopathy/decreased ejection fractionUncommon (1–10%)—SevereRare (<1%)—Severe/Potentially fatal
Retinopathy/Retinal vein occlusionUncommon (1–10%)—Severe/Vision-threatening
Interstitial lung disease/PneumonitisRare (<1%)—Severe/Potentially fatal
Myelosuppression (neutropenia, thrombocytopenia)Common (10–30%)—Severe
Muscle crampsCommon (10–30%)—Mild–Moderate
Table 6. Novel melanoma therapies categorized by mode of action and strategy according to [53]. All abbreviations employed are defined in the text in the Abbreviations section.
Table 6. Novel melanoma therapies categorized by mode of action and strategy according to [53]. All abbreviations employed are defined in the text in the Abbreviations section.
Mechanism of ActionTreatment Approach
Identifies predictive markers (PD-L1, TMB, IFN-γ, LAG-3, TILs, HLA types, gut microbiota) to personalize therapy and improve treatment outcomes.Biomarker Discovery and Validation
Employs genetically engineered T cells (e.g., TYP1-targeted) to recognize and kill melanoma cells; currently in preclinical and early clinical stages.CAR-T Cell Therapy Targeting Melanoma Antigens
Enhances antitumor responses by integrating ICIs with radiotherapy, oncolytic viruses, vaccines, or TILs to overcome immune resistance mechanisms.Combination Immunotherapy Approaches
Androgen receptor inhibition may boost NK cell activity and improve ICI effectiveness; currently under preclinical evaluation.Natural Killer (NK) Cell-Based Immunotherapy
Suppresses melanoma cell growth by inhibiting mutations in key driver genes (BRAF, KIT, NRAS, TERT, CCND1); TERT and CCND1 are common in acral melanoma.Targeted Inhibition of Oncogenic Signaling
Utilizes expanded TILs (e.g., lifileucel) to directly attack tumor cells; effective after ICI and BRAF/MEK inhibitor failure, though limited by cost and toxicity.Tumor-Infiltrating Lymphocyte (TIL) Therapy
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

Kawczak, P.; Bączek, T. Pathway-Specific Therapeutic Modulation of Melanoma: Small-Molecule Inhibition of BRAF–MEK and KIT Signaling in Contemporary Precision Oncology with a Special Focus on Vemurafenib, Trametinib, and Imatinib. J. Clin. Med. 2025, 14, 7906. https://doi.org/10.3390/jcm14227906

AMA Style

Kawczak P, Bączek T. Pathway-Specific Therapeutic Modulation of Melanoma: Small-Molecule Inhibition of BRAF–MEK and KIT Signaling in Contemporary Precision Oncology with a Special Focus on Vemurafenib, Trametinib, and Imatinib. Journal of Clinical Medicine. 2025; 14(22):7906. https://doi.org/10.3390/jcm14227906

Chicago/Turabian Style

Kawczak, Piotr, and Tomasz Bączek. 2025. "Pathway-Specific Therapeutic Modulation of Melanoma: Small-Molecule Inhibition of BRAF–MEK and KIT Signaling in Contemporary Precision Oncology with a Special Focus on Vemurafenib, Trametinib, and Imatinib" Journal of Clinical Medicine 14, no. 22: 7906. https://doi.org/10.3390/jcm14227906

APA Style

Kawczak, P., & Bączek, T. (2025). Pathway-Specific Therapeutic Modulation of Melanoma: Small-Molecule Inhibition of BRAF–MEK and KIT Signaling in Contemporary Precision Oncology with a Special Focus on Vemurafenib, Trametinib, and Imatinib. Journal of Clinical Medicine, 14(22), 7906. https://doi.org/10.3390/jcm14227906

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