Previous Article in Journal
Three-Year Outcomes of Neoadjuvant Chemoimmunotherapy vs. Neoadjuvant Chemoradiotherapy in Resectable Esophageal Cancer: A Multicenter Retrospective Study
Previous Article in Special Issue
Therapeutic Potential of CAR-CIK Cells in Acute Leukemia Relapsed Post Allogeneic Stem Cell Transplantation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Targeting the MAPK Pathway in Brain Tumors: Mechanisms and Therapeutic Opportunities

by
Dimitrios Vrachas
1,†,
Elisavet Kosma
1,†,
Angeliki-Ioanna Giannopoulou
1,
Angeliki Margoni
1,
Antonios N. Gargalionis
2,
Elias A. El-Habr
1,3,
Christina Piperi
1 and
Christos Adamopoulos
1,4,*
1
Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
2
Laboratory of Clinical Biochemistry, Medical School, ‘Attikon’ University General Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece
3
Sorbonne Université, CNRS, INSERM, Institut de Biologie Paris Seine, Center for Neuroscience at Sorbonne Université, 75005 Paris, France
4
Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Cancers 2026, 18(1), 156; https://doi.org/10.3390/cancers18010156 (registering DOI)
Submission received: 29 November 2025 / Revised: 25 December 2025 / Accepted: 31 December 2025 / Published: 2 January 2026
(This article belongs to the Special Issue Insights from the Editorial Board Member)

Simple Summary

Brain tumors remain among the most difficult cancers to treat, largely because of their biological complexity and the limited ability of many drugs to reach the brain. A major molecular pathway that drives the growth of many brain tumors is the MAPK signaling pathway. In this review, we explain how alterations in this pathway contribute to tumor development in both children and adults, and we summarize current and emerging therapies that specifically target this pathway. We also discuss the main challenges that limit treatment success, including drug resistance, tumor diversity, and the protective blood–brain barrier. By integrating recent advances in molecular biology with therapeutic strategies, this work aims to guide future research and improve precision treatment approaches for patients with brain tumors.

Abstract

Central nervous system (CNS) tumors consist of a diverse set of malignancies that remain clinically challenging due to their biological complexity, high morbidity, and limited responsiveness to current therapies. A growing body of genomic evidence has revealed that dysregulation of the mitogen-activated protein kinase (MAPK) signaling pathway is a recurrent and unifying characteristic across many pediatric and adult CNS tumor entities. Alterations affecting upstream receptor tyrosine kinases (RTKs), RAS GTPases, RAF kinases, and other associated regulators contribute to MAPK signaling pathway hyperactivation, shaping tumor behavior, therapy response and resistance. These aberrations ranging from hotspot mutations such as BRAF V600E and oncogenic fusions like BRAF–KIAA1549 are particularly enriched in gliomas and glioneuronal tumors, highlighting MAPK signaling as a key oncogenic driver. The expanding availability of molecularly targeted compounds, including selective inhibitors of RAF, MEK and ERK, has begun to transform treatment approaches for specific molecular subtypes. However, the clinical benefit of MAPK-directed therapies is frequently limited by restricted blood–brain barrier (BBB) penetration, intratumoral heterogeneity, parallel pathway reactivation, and an immunosuppressive tumor microenvironment (TME). In this review, we synthesize current knowledge on MAPK pathway alterations in CNS tumors and evaluate the therapeutic landscape of MAPK inhibition, with emphasis on approved agents, emerging compounds, combination strategies, and novel drug-delivery technologies. We also discuss mechanisms that undermine treatment efficacy and highlight future directions aimed at integrating MAPK-targeted therapy into precision-based management of brain tumors.

1. Introduction

Central nervous system (CNS) tumors represent a heterogeneous group of both malignant and benign entities, characterized by varying clinical behavior, histological, and molecular traits. In 2020, CNS tumors accounted for 1.6% of all cancer cases globally, while in 2022, 321,624 new cases were estimated, corresponding to an age-standardized incidence rate (ASIR) of 3.5 per 100,000 people [1]. Despite their relatively low incidence rate, they impose a major disease burden due to their disproportionately high mortality rates, especially among children [2,3]. The average lifespan for adults with glioblastoma, the most aggressive type of brain tumor, is approximately 2 years [3]. A study utilizing the global burden of disease (GBD) database predicted that the total number of cases will gradually increase by 2040, even though the mortality rates in certain populations may decrease slightly [4]. To date, the current established treatment approaches include surgical resection, radiotherapy, and chemotherapy [5]. CNS tumors are characterized largely by intratumoral heterogeneity which can be defined as the coexistence of genetically, epigenetically, transcriptionally, and phenotypically distinct cell subpopulations within the same tumor mass. This molecular heterogeneity of these tumors, their anatomical location, in conjunction with the protective role of the blood–brain barrier (BBB) and intrinsic or acquired drug resistance, leads to limited treatment efficacy and poor clinical patient outcomes. Importantly, children and young adults who survive by receiving the standard of care experience long-term complications that largely affect their quality of life [3]. Moreover, recent molecular profiling studies have revealed that pediatric and adult gliomas constitute fundamentally distinct biological entities, driven by different oncogenic alterations and signaling dependencies [6,7]. Consequently, there is a pressing need for more targeted and personalized therapeutic strategies.
The mitogen-activated protein kinase (MAPK) signaling pathway plays a fundamental role in cell physiology by regulating cell cycle, proliferation, survival, differentiation, apoptosis, and is implicated in various other developmental processes [8]. Core components of the MAPK pathway are serine/threonine-specific protein kinases, the mitogen-activated protein kinases (MAPKs), that transduce intracellular signals through sequential protein phosphorylation and activation events. Among them, the rapidly accelerated fibrosarcoma (RAF), the mitogen-activated protein kinase/extracellular signal-regulated kinase (MEK), and the extracellular signal-regulated kinase (ERK) form the RAF-MEK-ERK signaling axis, the central and most thoroughly studied MAPK pathway [8,9] (Figure 1).
The MAPK pathway is usually activated upon binding of a ligand, such as a growth factor, to a receptor tyrosine kinase (RTK), leading to its activation and the following recruitment, intracellularly, of adaptor proteins/regulators, which in turn activate the membrane-anchored small guanosine triphosphatase (GTPase), rat sarcoma virus oncogene (RAS) [10] (Figure 1). Afterward, the active GTP-bound RAS recruits at the membrane and activates RAF through a complex process of dimerization and phosphorylation events [11]. RAF then phosphorylates and activates its substrate MEK, which consecutively phosphorylates and activates ERK [12,13] (Figure 1). Finally, activated ERK phosphorylates its targets, usually transcription factors or co-activators in the nucleus, thereby regulating the expression of several genes [8,14].
RAS-RAF-MEK-ERK signaling axis deregulation, primarily due to its aberrant activation, is a key driver of carcinogenesis [15,16,17,18]. Mutations in key effectors of the pathway, most frequently in RAS and RAF, have been identified in a wide variety of cancers, including melanomas, lung, colorectal, and ovarian cancers [18,19,20].
Consequently, therapeutic efforts that target MAPK pathway components have led to the approval by the Food and Drug Administration (FDA) of several small-molecule inhibitors, while other alternative targeting approaches are under pre-clinical investigation and development, including gene silencing, proteolysis-targeting chimeras (PROTACs), and bispecific antibodies [8,21,22,23].
Notably, MAPK pathway component alterations have also been detected in primary brain tumors. The most common alterations are the gene fusion between the BRAF and KIAA1549 genes and the BRAF V600E mutation, both of which are most prevalent in pediatric compared to adult tumors [24].
Beyond these, diverse genetic alterations involving RTKs, RAS, RAF kinases and pathway regulators lead to the aberrant activation of the MAPK pathway and highlight its central role in the pathogenesis of CNS tumors [8,25].
In this review, we provide a comprehensive overview of MAPK pathway alterations in CNS malignancies, with emphasis on pediatric and adult gliomas, glioneuronal tumors, and ependymomas. We summarize current strategies for MAPK pathway inhibition, including BRAF, MEK, and ERK inhibitors, and discuss how these approaches are being integrated into clinical management [26,27]. Furthermore, we address the major therapeutic challenges that limit efficacy, including restricted BBB penetration, tumor heterogeneity and resistance mechanisms, and the immunosuppressive tumor microenvironment [28,29]. Finally, we highlight emerging treatment concepts and combinatorial approaches that hold promise and shape future perspectives for MAPK-targeted therapy in brain tumors.

2. Central Nervous System (CNS) Tumors

Central nervous system (CNS) tumors can be classified as either primary, originating from cell types within the brain and spinal cord, or metastatic, arising from tumors that develop in distal organs, most commonly the lung and breast, and spread to the brain through the bloodstream or the lymphatic system [30,31,32]. Primary CNS neoplasms depict a heterogeneous group, consisting of gliomas, glioneuronal, neuronal, and ependymal tumors [33]. Although relatively rare, tumors of the brain and other parts of the central nervous system contribute substantially to morbidity and mortality across all age groups. The frequency of these neoplasms is higher in children aged up to 5 years old, with most being malignant gliomas, germ-cell, and embryonal tumors. In adults, malignant CNS tumors, especially gliomas, are among the leading causes of death [31,34].
The basic criteria for the characterization of these entities have traditionally depended on histological, immunohistochemical, and cytological observations, often linked to their likeness to an alleged cell type of origin. The recognition of CNS tumors solely based on morphological features began to hinder the diagnosis of several subgroups and, hence, proper treatment. Eventually, as described in the 5th edition of the World Health Organization (WHO) CNS tumor classification (CNS5, 2021), this obstacle was overcome by incorporating molecular and genetic alterations into the diagnostic criteria. According to the current classification, six families of both benign and malignant tumors have emerged, comprising adult-type diffuse gliomas, pediatric-type diffuse low-grade gliomas (DLGG), pediatric-type diffuse high-grade gliomas (DHGG), circumscribed astrocytic gliomas, glioneuronal/neuronal tumors, and ependymal tumors [33,35].
The implementation of molecular assays in their diagnosis, such as DNA/RNA sequencing, genome-wide methylation profiling, quantitative PCR (qPCR), and FISH, has revealed a broad spectrum of genetic alterations in CNS tumors, encompassing point mutations, insertions and deletions, copy number changes, and gene rearrangements [36]. The genes that are more frequently affected are vital to cellular homeostasis. For instance, alterations in genes encoding for the phosphoinositide 3-kinase (PI3K), epidermal growth factor receptor (EGFR), V-Raf murine sarcoma viral oncogene homolog B (BRAF), platelet-derived growth factor receptor α (PDGFRA), and Met tyrosine-protein kinase (MET) lead to defective receptor tyrosine kinase signaling. The regulation of the cell cycle is also affected by mutations in the p53, retinoblastoma susceptibility (RB1), cyclin-dependent kinase 4 (CDK4), cyclin-dependent kinase inhibitor 2A and 2B (CDKN2A and CDKN2B), and v-myb avian myeloblastosis viral oncogene homolog (MYB) genes. Moreover, genetic changes in telomerase reverse transcriptase (TERT) and α-thalassemia intellectual disability X-linked (ATRX) genes affect the preservation of telomere integrity. Additionally, modifications in histone variants H3.1 and H3.3, predominantly the substitutions K27M and G34V/R, are implicated in abnormal chromatin arrangement and epigenetic regulation of gene expression [35,36,37,38]. Cell metabolism is also affected through the production of oncometabolites, such as 2-hydroxyglutarate (2-HG), which arises from mutations in the isocitrate dehydrogenase (IDH) gene [39]. The mutations in IDH1 and IDH2 often co-exist with concurrent deletion of chromosome arms 1p and 19q (1p/19q codeletion) and TERT alterations [40].
The treatment of CNS tumors continues to pose difficulties in both pediatric and adult age groups. More specifically, several parameters need to be considered in terms of tumor cell origin, location, genetic background, microenvironment and effective drug delivery. The standard clinical strategies, so far, involve surgery, radiation, and chemotherapy [30,41,42]. Over the last years, more targeted therapies, incorporating inhibitors, chimeric antigen receptor-T (CAR-T) cells, and vaccines, among others, have emerged. Regarding glioblastoma multiforme (GBM), the most frequent and aggressive form of glioma, immunotherapy may prove to be a promising treatment option [43]. Nonetheless, brain tumors still represent one of the main causes of cancer-related mortality, while survivors face a high risk of chronic health conditions, thus underscoring the pressing need for new treatment modalities [42,44].

2.1. MAPK Pathway Alterations in CNS Tumors

Aberrant activation of the MAPK pathway is a hallmark of several CNS tumors, frequently driven by genetic alterations such as point mutations, gene fusions, amplifications, or overexpression, most commonly involving RTKs, RAS, RAF, and regulators of the pathway such as SHP2 and NF1 [22,45,46] (Figure 2).

2.1.1. Receptor Tyrosine Kinase (RTK) Alterations

RTKs are critical oncogenic drivers in gliomas, affected by diverse mechanisms, which include point mutations, gene amplifications, and chromosomal rearrangements that create fusion oncoproteins (Figure 2). These aberrations commonly confer ligand-independent kinase activation, leading to persistent MAPK pathway signaling [47]. For instance, EGFR alterations, most notably gene amplification and the EGFRvIII deletion variant, are prevalent in GBM and drive aggressive proliferation and therapeutic resistance [48]. Similarly, PDGFR-A amplification is characteristic of the proneural GBM subtype, while MET amplification and PDGFR overexpression also contribute to glioma pathogenesis [49,50] (Figure 2). Importantly, in pediatric gliomas, gene fusions involving anaplastic lymphoma kinase (ALK), ROS proto-oncogene 1 (ROS1), neurotrophic tyrosine receptor kinase (NTRK2) and MET define a distinct, hemispheric high-grade subgroup with intermediate prognosis [51]. Fibroblast growth factor receptor (FGFR) gene family alterations, including FGFR3–transforming acidic coiled-coil containing protein 3 (TACC3) fusions, constitute actionable drivers in a subset (3–5%) of GBM, producing fusion proteins that promote oncogenesis [52].
In pilocytic astrocytomas (PAs), fusions of the NTRK2 gene, which encode for the tropomyosin receptor kinase B (TrkB) with either the transcriptional repressor nucleus Accumbens-associated protein 2 (NACC2), NACC2-NTRK2, or the pre-mRNA alternative splicing regulator Quaking homolog KH domain containing RNA binding (QKI), QKI-NTRK2, can induce MAPK pathway hyperactivity in a ligand-independent manner [53]. Additionally, in PA patients, the activating substitutions N546K and K656E in the FGFR1 gene have been linked to elevated phosphorylated/activated ERK levels [54]. In a small number of infant-type hemispheric gliomas (IHGs), gene fusions involving ALK, NTRK1, and ROS1 were detected without co-occurrence [54,55]. In the same study, a case of PA bore a fusion of breakpoint cluster region (BCR) and NTRK2 genes (BCR-NTRK2), while a patient with pleomorphic xanthoastrocytoma (PXA) harbored the tropomyosin 3 (TPM3)-NTRK1 fusion. Moreover, two cases of gangliogliomas exhibited the fusion FGFR3-TACC3 [54,55].
The localization of NTRK fusions varies depending on the fusion partner. Thus, when the 5′ fusion partner encodes a membrane or extracellular protein, such as the protein fusions of TrkA with the proteoglycan brevican (BCAN), BCAN–NTRK1, and the cell surface protein neurofascin (NFASC), NFASC–NTRK1, the fusion protein is membrane-associated (Type II) (Figure 2). Otherwise, when the fusion partners are cytosolic or nuclear, such as TPM3, QKI, Ets variant transcription factor 6 (ETV6), localization is cytoplasmic (Type I) (Figure 2). Yet all retain the NTRK kinase domain and drive constitutive MAPK signaling [56]. In GBM, Golgi-associated PDZ and coiled-coil motif-containing protein (GOPC)–ROS1 fusion proteins exhibit isoform-specific subcellular localization with the long form localizing to the Golgi, while the short form is cytoplasmic [57].

2.1.2. RAS Alterations

All three RAS small GTPases, Kirsten rat sarcoma viral oncogene homolog (KRAS), neuroblastoma rat sarcoma viral oncogene homolog (NRAS) and Harvey rat sarcoma viral oncogene homolog (HRAS), function as molecular switches that alternate from their inactive GDP-bound state to their active GTP-bound state (Figure 1). Oncogenic RAS mutations, typically missense substitutions at hotspot codons 12, 13, or 61, largely impair intrinsic GTP hydrolysis and lock RAS in its active form, resulting in constitutive downstream signaling [58] (Figure 2). While RAS mutations are among the most common oncogenic drivers in many cancers, they are relatively rare in gliomas. Nevertheless, accumulating evidence indicates that RAS alterations can contribute to gliomagenesis across different subtypes, either as point mutations or gene copy number gains, often cooperating with other oncogenic events to sustain tumor growth and progression [59].
In two distinct cases of PXA, a KRAS mutation at codon 61 was detected, which codes for glutamine in position 61 of KRAS protein [60]. More specifically, in one case, glutamine was substituted by lysine (Q61K), while in the other, it was replaced by histidine (Q61H). Interestingly, these two cases were the first in which a KRAS mutation was detected in PXA patients [60]. Despite its rarity, a case of ganglioma with an NRAS mutation was reported among a heterogeneous group of 30 patients with infantile CNS tumors [54,55]. Oncogenic missense mutations in KRAS and NRAS were present in 8 patients with IDH-mutant astrocytomas, including G12A/D/V, G13D, D33E, A146T, and K117N substitutions. In the same cohort, 3 tumor samples reported a high increase in KRAS gene copies [25,54,55].

2.1.3. MAPK Pathway Regulators Alterations

Beyond RTKs and RAS itself, several intracellular modulators of MAPK signaling are altered in CNS tumors. The most relevant include neurofibromin (NF1) and the associated sprouty-related EVH1 domain-containing protein 1 (SPRED1) and leucine zipper-like transcription regulator 1 (LZTR1), and Src homology region 2 domain-containing phosphatase-2 (SHP2). NF1 acts as a tumor suppressor by accelerating RAS GTP hydrolysis, a process facilitated by SPRED1, which recruits NF1 to the plasma membrane [24,61]. In PXAs, NF1 mutations were reported in 3 of 13 cases, including two missense and one truncating variant [58]. In IDH-mutant astrocytomas, NF1 alterations occurred in 17 of 27 cases, while LZTR1 mutations (nonsense, frameshift, splice site, or missense) were also observed, consistent with loss of its role in targeting RAS proteins, among others, for ubiquitin-mediated degradation [62]. Inactivating SPRED1 lesions, including biallelic deletion and frameshift changes, have also been described in this tumor type [24].
On the other hand, SHP2 is a positive effector that promotes MAPK pathway activation, as it functions as an adaptor protein and phosphatase downstream of multiple RTKs (Figure 1). Gain-of-function mutations, particularly E69K and E76A, enhance its activity, facilitating sustained RAS/MAPK signaling in PA [54,63] (Figure 2).

2.1.4. RAF Alterations

Within the RAF family of serine/threonine kinases, BRAF is the predominant oncogenic driver in cancers, including gliomas, followed less frequently by CRAF and only rarely by ARAF. RAF proteins under physiological conditions signal as BRAF homodimers or BRAF-CRAF heterodimers, constituting the most common and biologically relevant signaling forms [64,65] (Figure 1). Oncogenic BRAF alterations, through gene fusions or hotspot mutations, result in sustained constitutive downstream MAPK activation.
The most prevalent mutation in pilocytic astrocytomas is a genomic rearrangement that leads to the fusion of the KIAA1549 and BRAF genes. The structural rearrangement involves the duplication of a DNA segment between the KIAA1549 5′-end and BRAF 3′-end genes in the 7q34 chromosomal region, spanning approximately 2 Mb. From this event, five different in-frame variants have been identified: KIAA1549ex16-BRAFex9, KIAA1549ex15-BRAFex9, KIAA1549ex19-BRAFex9, KIAA1549ex16-BRAFex11, and KIAA1549ex18-BRAFex10. All the resulting chimeric proteins exhibit constitutive activation, as they all lack the N-terminal domain responsible for the autoregulation of BRAF, due to substitution from KIAA1549. At the same time, they maintain the kinase domain of BRAF [66,67,68,69]. Interestingly, in a small cohort of PAs several other fusion partners have been identified for BRAF, including family with sequence similarity 131 member B (FAM131B), ring finger protein 130 (RNF130), chloride voltage-gated channel 6 (CLCN6), makorin ring finger protein 1 (MKRN1), guanine nucleotide-binding protein subunit alpha-11 (GNA11), quaking homolog KH domain RNA binding protein (QKI), fizzy and cell division cycle 20 related 1 (FZR1), microtubule actin crosslinking factor 1 (MACF1), and general transcription factor II-I (GTF2I). Although biologically and functionally disparate, these fusion partners render domains that converge on the same outcome: hyperactivation of BRAF and its downstream signaling [24,54,70,71].
Regarding BRAF mutations, the second most common alteration detected in PAs is a substitution of valine in position 600 by glutamic acid, which results in BRAF V600E the most frequent BRAF mutation in human cancers [69,72,73]. This point mutation disrupts the regulatory conformation of BRAF, resulting in loss of its N-terminal autoinhibitory domain and conferring monomeric kinase activity with hyperactivation of the MAPK pathway (Figure 2) [72,73]. BRAF V600E is observed across several glioma subtypes. In a study by Zou et al., who evaluated mutations in a cohort of 13 PXA patients using next-generation sequencing, the BRAF V600E mutation was present in 38% of the cases [60]. In a comprehensive analysis of 30 infantile (<12 months old) CNS tumors, 7/10 cases of desmoplastic infantile ganglioglioma (DIG) harbored alterations in BRAF (5 mutations, 1 duplication and 1 fusion), 1/2 cases of PXA carried the CAP-Gly domain containing linker protein 2 (CLIP2)-BRAF fusion, 1/2 cases of PA had the KIAA1549-BRAF fusion, and a single case of DLGG was BRAF V600E-mutant [55]. Tumors from 3 young-adult patients with IDG-mutant astrocytomas possessed a rare in-frame protein tyrosine phosphatase receptor type Z1 (PTPRZ1)-BRAF gene fusion and two class III BRAF mutations, the substitutions G464E and D594G [25]. G464E affects the kinase domain of BRAF, producing a kinase-impaired protein that requires RAS activation, whereas D594G affects the activation segment of BRAF, resulting in a kinase-dead protein, both relying on upstream RAS/RTK activity [74,75].
In the context of chimeric proteins, the gene encoding CRAF protein (CRAF or RAF1) has been observed to fuse either with the nuclear transcription factor 1A (NF1A) or SLIT-ROBO Rho GTPase activating protein 3 (SRGAP3) gene in some rare PA case reports at chromosomal regions 1q31.3 and 3p25, respectively. The end-product of both translocations is an oncoprotein that augments constitutive MAPK pathway activation [68,76].
Alterations are not limited to BRAF. In the study of Tauziède-Espariat et al., 2/30 infantile patients with tumors characterized as DIGs carried CRAF fusions, particularly one of these cases presented with the protein kinase cAMP-dependent type II regulatory subunit a (PRKAR2A)-RAF1 fusion [55]. CRAF fusions have been described in rare PAs, involving the nuclear transcription factor 1A (NF1A) or SLIT-ROBO Rho GTPase activating protein 3 (SRGAP3) as fusion partners [68,76]. In infantile DIGs, CRAF fusions have also been identified, including a protein kinase cAMP-dependent type II regulatory subunit α (PRKAR2A)-CRAF chimera [55]. Additional RAF1 fusions, contributing to constitutive MAPK activity, have been documented across gliomas and other tumor types [77].

3. RAS/MAPK Pathway Inhibitors in CNS Tumors

The high frequency of activating mutations and other genetic alterations in the RAS/RAF/MEK/ERK signaling axis and its subsequent hyperactivation has highlighted their association with cancer development and progression [78]. Consequently, components of this pathway have become promising therapeutic targets through their selective inactivation by small-molecule inhibitors. In addition, alternative medicinal chemistry strategies with the development of PROTACs or molecular glues have emerged. All these targeting efforts have been directed towards CNS tumors as well [79].

3.1. RAF Inhibitors

The high frequency of the BRAF V600E mutation, accounting for 95% of all BRAF mutations, and the increased kinase activity of the BRAF V600E oncoprotein made it an ideal pharmacological target for small-molecule inhibitors. This led to the development of the first- and eventually the more selective second-generation RAF inhibitors targeting the mutant-BRAF kinase [72,73]. The increased selectivity for the monomeric mutated BRAF versus the dimeric wild-type BRAF is the basis of the high therapeutic index of the second-generation RAF inhibitors [74]. These discoveries resulted in the FDA’s approval of vemurafenib in 2011 and dabrafenib in 2013, as single agents, for the treatment of metastatic BRAF V600E-mutant melanoma [72,73]. Since then, combination therapies using the RAF inhibitors vemurafenib, dabrafenib and encorafenib, along with the MEK inhibitors trametinib, cobimetinib and binimetinib or the EGFR inhibitor cetuximab, have gained FDA approvals in subsequent years for other types of cancer, harboring the BRAF V600E mutation [72,73,80]. However, the effectiveness of these monomer-selective RAF inhibitors is often hindered by the development of adaptive resistance, primarily mediated by the formation of RAF dimers, through MAPK-pathway reactivation because of the relief of negative feedback or via secondary genetic alterations [72,73,74]. To overcome the dimer-forming resistance mechanisms, next-generation RAF inhibitors that target the dimeric form of RAF have been developed [72,73,74]. Recently, one such inhibitor, tovorafenib, was clinically approved for treating pediatric patients with low-grade glioma (LGG) carrying genetic alterations in the BRAF gene [26].

3.1.1. Vemurafenib

Vemurafenib is a selective BRAF V600E inhibitor that competes with ATP binding, thus preventing downstream MEK activation. It exhibits limited penetration across the BBB, which restricts its efficacy in primary brain tumors (Table 1) [81]. Initially approved for metastatic melanoma, vemurafenib has shown partial efficacy in BRAF-mutant gliomas in case series and small trials [82,83,84]. Responses tend to be short-lived due to the development of adaptive resistance and insufficient CNS concentrations. Common adverse effects include rash, joint pain, fatigue and paradoxical activation of wild-type BRAF leading to secondary malignancies like squamous cell carcinoma [82,83,84].

3.1.2. Dabrafenib

Dabrafenib is another selective BRAF V600E inhibitor with superior BBB penetration and a more favorable safety profile compared to vemurafenib [85]. Clinical trials have demonstrated that dabrafenib is effective in pediatric patients with BRAF-mutant LGGs, leading to tumor regression and improved progression-free survival [86]. On 16 March 2023, dabrafenib combined with the MEK inhibitor trametinib gained FDA approval for pediatric BRAF V600E-mutant LGGs (Table 1) [86]. This synergistic regimen shows improved tolerability, with fewer secondary skin malignancies when used in combination therapy. Patients may exhibit pyrexia, fatigue, skin rash and arthralgia [87].

3.1.3. Encorafenib

Encorafenib is a newer first-generation BRAF V600E inhibitor developed to reduce paradoxical activation and enhance the duration of response [88]. While it is primarily used in melanoma and colorectal cancer, preclinical studies are investigating its use in brain tumors [89]. Although encorafenib features a longer dissociation half-life from BRAF V600E and potentially better pharmacodynamic suppression of MAPK signaling, its efficacy in CNS tumors may be limited by its lower BBB penetration (Table 1) [90].

3.1.4. NST-628

NST-628 is a non-degrading molecular glue that binds to both RAF and MEK proteins, stabilizing their complex in a way that prevents MEK phosphorylation by RAF, effectively blocking downstream signaling [91]. This mode of action avoids resistance mechanisms common in traditional kinase inhibitors. NST-628 inhibits all RAF isoforms (ARAF, BRAF, CRAF) and works across multiple RAS- and RAF-mutant cancers, including those resistant to existing therapies. Unlike many inhibitors, NST-628 is brain-penetrant, making it potentially effective against CNS tumors (Table 1). The compound induces long-lasting suppression of the MAPK pathway in both in vitro and, also, in vivo models, including mouse xenografts and organoids derived from human tumors. As a result, due to its broad activity, resistance-evasion capacity, and brain penetration, NST-628 shows promise for treating a wide range of RAS- and RAF-driven CNS cancers, including those with KRAS, NRAS, or BRAF mutations [91].

3.2. MEK Inhibitors

Selective MEK inhibitors have been developed to effectively block the MAPK pathway activation, especially after its reactivation due to the relieved negative feedback mechanisms following BRAF inhibitor therapy [72,73,74]. Thus, combinatorial targeting of MEK inhibitors (trametinib, cobimetinib, binimetinib) with RAF inhibitors (vemurafenib, dabrafenib, encorafenib) has been FDA-approved, from 2014 to 2018, for patients with metastatic melanoma, non-small cell lung cancer (NSCLC), and anaplastic thyroid cancer carrying the BRAF V600E mutation [72,73,92,93,94]. In 2021, the FDA approved the MEK inhibitor selumetinib for pediatric patients with neurofibromatosis type 1, a genetic disorder in which NF1 loss predisposes to peripheral nerve sheath tumors and other cancers (Table 1) [95,96]. Most MEK inhibitors disrupt the formation of the RAF-MEK complex, inhibiting MEK phosphorylation and activation [97].

3.2.1. Selumetinib

Selumetinib is an allosteric MEK inhibitor that prevents ERK activation and has demonstrated significant efficacy in NF1-associated pLGGs [98], as well as in non-NF1-associated pLGGs [99], including disease stabilization and, in some cases, prolonged disease control (Table 1). Furthermore, it is an orphan drug designation for NF1-altered gliomas [100]. Generally, it is well tolerated in children, but adverse effects, like gastrointestinal symptoms, skin rash, rare cardiomyopathy and ocular toxicity may arise [98,99,100]. Ongoing clinical trials comparing selumetinib with conventional chemotherapy in both NF1-associated and non-NF1 pLGG will further clarify its therapeutic value and long-term safety. Notably, emerging evidence indicates that a subset of patients can maintain durable progression-free survival (PFS) even after treatment cessation, underscoring the potential of MEK inhibition as a promising disease control strategy [99,100].

3.2.2. Trametinib

Trametinib is a potent selective allosteric MEK inhibitor, often used in combination with dabrafenib [92,93,94,97,101]. Approved in combination for BRAF V600E-mutant tumors, trametinib enhances efficacy and reduces adverse effects such as secondary skin cancers [92,93,94,101,102]. Current results from an ongoing clinical trial demonstrate significant clinical benefit to the majority of both pLGG and plexiform neurofibromas (PNs) patients, including measurable responses and prolonged PFS (Table 1) [102]. Common adverse effects are diarrhea, skin rash, fatigue, and hypertension [102].

3.2.3. Binimetinib and Cobimetinib

The MEK inhibitors binimetinib and cobimetinib have been tested primarily in non-CNS malignancies but are currently under investigation in gliomas [89]. Ongoing trials are evaluating their BBB permeability and potential in combination with BRAF and mTOR inhibitors, supported by favorable pharmacokinetics and CNS bioavailability [103]. Binimetinib is currently under clinical investigation in brain tumors, including high-grade glioma (HGG) [89]. Cobimetinib provided efficacy when tested in combination with vemurafenib in a refractory case of BRAF V600E-mutated ganglioglioma [104]. Its role in neuro-oncology, however, remains to be fully defined. Given their pharmacologic profiles, both agents represent promising candidates for rational combination strategies targeting multiple signaling pathways in gliomas.

3.2.4. Mirdametinib

Mirdametinib is an orally bioavailable MEK inhibitor that has recently achieved its first regulatory approval in the United States for the treatment of symptomatic, inoperable NF1-associated PNs in both adult and pediatric patients (Table 1) [105]. Beyond neurofibromatosis type 1, ongoing trials are evaluating its efficacy in pLGGs and other RAS/MAPK-driven tumors, providing a rationale for its potential application in primary brain tumors [105,106]. With established clinical activity in NF1-associated CNS tumors and a favorable oral dosing profile, mirdametinib represents a promising candidate for expanding MEK-directed strategies in neuro-oncology [105,106].

3.3. ERK Inhibitors

Although mutations in ERK proteins are rare, selective ERK inhibitors are under preclinical development, given that ERK is the terminal kinase of the RAS/RAF/MEK/ERK signaling cascade, seeking a more durable inhibitory response [107]. These agents are particularly promising in tumors that develop resistance to BRAF and/or MEK inhibitors.

Ulixertinib (BVD-523)

Ulixertinib is an oral, ATP-competitive ERK inhibitor that has demonstrated preclinical efficacy in various tumor models, including those resistant to BRAF and MEK inhibitors. Phase I clinical trials have shown acceptable tolerability and preliminary antitumor activity in patients with advanced solid tumors harboring MAPK pathway alterations. In gliomas, its ability to cross the BBB and suppress ERK-driven transcription makes it a promising candidate, although its application is under exploration (Table 1) [108]. Elevated liver enzymes, diarrhea and fatigue are the main observed side effects of this drug [108].

3.4. SHP2 Protein Inhibitors

Alongside direct targeting of the RAS/RAF/MEK/ERK axis components, selective inhibitors have been developed against the SHP2 phosphatase (e.g., TNO155 and RMC-4630), which block upstream activation of RAS by inhibiting the GRB2-SOS1 interaction [109] (Figure 1). Specifically, a study has shown that SHP2 inhibition, using the SHP2 inhibitor SHP099, could efficiently reduce RAS-GTP loading, block RAS-mediated RAF/MEK/ERK signaling and abrogate tumor growth in NF1-malignant peripheral nerve sheath tumors (MPNSTs) (Table 1) [110]. Furthermore, combining SHP2 inhibition treatment with hydroxychloroquine (HQ), a pharmacological inhibitor of autophagy, showed enhanced effectiveness in mouse and human NF1-MPNST models [110]. Additionally, Sang and colleagues examined the efficacy of SHP099 in GBM with activated PDGFR-A. SHP099 exhibited antitumor activity either as a single agent or in combination with temozolomide (TMZ) and provided significant survival benefits for GBM tumor xenograft-bearing animals [111].

3.5. Combinatorial Therapies

Combined inhibition of multiple MAPK pathway components enhances treatment efficacy and reduces the risk of resistance or overcomes the already developed adaptive resistance [72,74,97,112,113]. BRAF plus MEK inhibitor is the most validated combination, especially in GBM and in pediatric LGG [114,115]. It delays resistance, lowers toxicity, and provides better PFS compared to monotherapy. Furthermore, ongoing trials are exploring a multi-combinatorial strategy of BRAF, MEK, and AKT inhibitors [116,117]. Lastly, MAPK inhibitors could combine with immunotherapy, given that MAPK inhibition may increase immune recognition, making combination with immune checkpoint inhibitors (ICIs) a promising therapeutic avenue [118,119].

3.6. Clinical Application and Efficacy

3.6.1. Pediatric Low-Grade Glioma (LGG)

Pediatric LGGs are the most frequent pediatric brain tumors and are characterized by indolent growth but can cause significant morbidity. Molecular profiling has revealed that most of these tumors harbor MAPK pathway alterations. The combination of dabrafenib and trametinib has demonstrated remarkable efficacy in pediatric LGGs with BRAF V600E mutations [101,114,115]. In clinical trials, response rates exceeded 70%, and the combination was associated with PFS and tolerable side effects [114,115]. Selumetinib has also shown clinical benefit in NF1-associated pediatric LGGs [98]. Results from Phase II trials indicated that selumetinib led to tumor shrinkage and visual improvement in children with optic pathway gliomas [99,100].

3.6.2. High-Grade Glioma (HGG)

In HGG, the MAPK pathway is often only one of many dysregulated networks, and monotherapy with BRAF or MEK inhibitors has generally been less effective [120]. However, in select cases, such as BRAF V600E-positive GBM, targeted therapies have resulted in durable responses [101,121]. Combination therapy is currently under active investigation in clinical trials, including regimens that pair MAPK inhibitors with other targeted or immunotherapeutic agents [122,123]. Specifically, Arbour and colleagues reported an 18-year-old female with a grade III PXA treated upfront with dabrafenib and trametinib [122]. Also, Fusco et al. describe a similar case of a 19-year-old male patient with grade III PXA, who achieved durable PFS with BRAF and MEK inhibitors combination [123].

3.6.3. Ganglioglioma

Gangliogliomas are usually low-grade brain tumors containing both neuronal and glial elements, most often occurring in children and young adults. A large proportion of these tumors harbor activating mutations in the MAPK signaling pathway, particularly BRAF V600E, which renders them responsive to MEK inhibitors [124]. Nonetheless, some gangliogliomas lack identifiable MAPK pathway alterations and therefore have not traditionally been considered candidates for MEK-targeted therapy. Interestingly, a recent report described a young adult patient with ganglioglioma who did not carry MAPK pathway mutations but achieved a marked and durable response to the MEK inhibitor trametinib [125].

3.6.4. Medulloblastoma

Medulloblastoma is a common malignant pediatric brain tumor. While existing treatments can be effective, they often cause significant long-term side effects [126]. A major clinical challenge is resistance to therapy and recurrence, often driven by tumor stem-like cells [127]. The protein BMI1, a known regulator of stem cell renewal and tumorigenesis, is overexpressed in medulloblastoma and supports tumor growth [128]. A study investigates whether targeting BMI1, alone or in combination with MAPK/ERK pathway inhibitors, could be an effective treatment strategy against medulloblastoma [129]. The study used the PD325901, a MEK inhibitor that blocks ERK phosphorylation, as the MAPK/ERK pathway inhibitor in combination with BMI1 inhibition to evaluate synergistic effects on medulloblastoma cells [129].

3.7. Current and Ongoing Clinical Trials

Several ongoing clinical trials assess the MAPK inhibition in different CNS tumor types (Table 2). Selectively, some of them include the evaluation of the dabrafenib plus trametinib combination in pLGGs [114], which paved the way for the FDA approval of this combination for treatment, the investigation of the effectiveness of selumetinib in NF1-associated gliomas [96], and the study of the role of tovorafenib in relapsed pLGG with BRAF alterations (FIREFLY-1/NCT04775485) [130]. These studies are refining indications, dosing and combinations, and will help define future standard-of-care approaches.

4. Therapeutic Challenges of Targeting the MAPK Pathway in Brain Tumors

4.1. Blood–Brain Barrier (BBB) and Drug Delivery Limitations

The therapeutic management of intracranial tumors such as gliomas, meningiomas, pituitary adenomas and craniopharyngiomas is limited by the presence of both the BBB and the blood–tumor barrier (BTB). BBB’s fundamental role through its high selectivity is to maintain cerebral homeostasis, but at the same time, it restricts the entry of many pharmacological agents, especially large or hydrophilic molecules [131,132,133]. In contrast, the BTB, which arises from abnormal tumor-induced angiogenesis, displays heterogeneous permeability [134]. This results in uneven intratumoral drug distribution, particularly in aggressive tumors such as GBM and craniopharyngiomas. These anatomical and physiological characteristics affect the uniform delivery and eventually the efficacy of systemically administered drugs [28,135]. Thus, effective brain tumor treatment requires the development of compounds that both target oncogenic signaling pathways and, at the same time, achieve adequate penetration through the BBB. However, even small, lipophilic molecules can fail to accumulate sufficiently in the CNS due to active efflux mechanisms mediated by ATP-binding cassette (ABC) transporters, which include the P-glycoprotein (P-gp/ABCB1) and breast cancer resistance protein (BCRP/ABCG2) [136,137]. Such transporters are located at the BBB/BTB interface and within tumor cells and contribute to chemoresistance by actively extruding therapeutic agents from the brain parenchyma [138]. Importantly, ABC transporter expression is heterogeneous across tumor subtypes and can be upregulated in response to treatment. For instance, exposure to doxorubicin has been demonstrated to induce the expression of multiple transporters, such as P-gp, BCRP, MRP-1, -2, -3, and -6, in gliomas, further compounding resistance [139].
Many RTK inhibitors, including erlotinib, gefitinib, and afatinib, are known substrates of P-gp and BCRP, restricting their CNS bioavailability [140,141,142]. Certain compounds, such as sunitinib and sorafenib [143], and third-generation EGFR inhibitors, such as osimertinib, rociletinib, and HM61713, demonstrate improved BBB penetration and activity against resistance mutations like EGFR T790M challenges persist [142]. Similar pharmacokinetic barriers are encountered with RAF inhibitors, like vemurafenib and dabrafenib, and MEK inhibitors like trametinib, cobimetinib, binimetinib, selumetinib and pimasertib, many of which are subject to efflux via P-gp and BCRP. Other MEK inhibitors such as PD0325901 and E6201 have shown favorable BBB permeability in preclinical studies [144,145]. Furthermore, newer-generation RAF inhibitors, including dabrafenib, encorafenib, and the molecular glue, dual RAF-MEK inhibitor NST-628, have demonstrated enhanced CNS distribution, thus improving their therapeutic usage in intracranial malignancies [87,89,91]. Compounds that target KRAS G12C, like sotorasib and adagrasib, and ERK, like ulixertinib, are in active clinical evaluation. However, their pharmacokinetic properties and association with efflux transporters are not yet defined [108,146,147]. Several innovative drug delivery strategies are currently being explored, including nanoparticles, focused ultrasound-mediated BBB disruption and convection-enhanced delivery [28].

4.2. Tumor Heterogeneity and Resistance Mechanisms in MAPK Pathway-Targeted Therapies

Genetic alterations in the MAPK signaling pathway vary across different brain tumor types, affecting disease progression and therapeutic response. Hyperactivating mutations such as BRAF V600E and BRAF–KIAA1549 fusions are frequently observed in pediatric and adult low-grade gliomas, such as PAs, gangliogliomas, and PXAs [54,148]. Mutations in genes including ROS1, ALK, NF1, KRAS, MEK, and CRAF have been identified across glioma subtypes, highlighting the necessity for molecular classification beyond traditional histopathology [35,149,150]. Additionally, fusions involving FGFR and NTRK family genes and fusions and amplifications in ALK, ROS1, and MET, have been detected predominantly in pediatric brain tumors [51,151,152]. Mutations in PIK3CA and AKT1 are frequent in meningiomas, whereas activating mutations affecting the RAS/RAF/MEK/ERK, PI3K/AKT, and Wnt pathways have been described in pituitary adenomas [153,154]. Apart from primary CNS tumors, genetic alterations within the MAPK pathway, such as RTK and BRAF mutations, are often observed in brain metastases originating from lung, breast, and melanoma primary cancers [155]. Despite the considerable therapeutic advances of the MAPK pathway-directed targeted therapies, their clinical success is frequently limited by the development of drug resistance [22,156]. These resistance mechanisms often include compensatory activation of parallel signaling cascades like PI3K/AKT/mTOR pathway [72,157,158]. Furthermore, reactivation and/or hyperactivation of the MAPK pathway through the relief of negative feedback loops upon treatment with MAPK inhibitors, fosters epigenetic reprogramming by inducing expression of key transcription factors associated with cellular stemness and mesenchymal transition. This process involves chromatin remodeling, enhancer reconfiguration, and rewiring of the transcription factor network, including the transcription factors SOX2, OLIG2, STAT3, KLF4, and NOTCH, promoting therapeutic evasion and adaptive resistance [72,73,74,157,158,159,160].

4.3. Tumor Microenvironment (TME)

The TME functions as a dynamic ecological system that actively shapes tumor evolution, therapeutic response, and resistance through complex and reciprocal interactions between tumor cells and their surrounding stromal and immune compartments [161]. Genetic and epigenetic alterations influence the transcriptional and secretory programs of cancer cells, thereby reprogramming the TME, which in turn contributes to the emergence of resistance to MAPK pathway inhibitors. Certain oncogenic mutations in both the RAS/MAPK and PI3K/AKT signaling pathways can support an immunosuppressive TME [162,163]. Moreover, in GBM, elevated levels of phosphorylated ERK have been associated with an increased TME infiltration by M2-type tumor-associated macrophages (TAMs) [164]. This altered microenvironment corresponds to “cold tumors,” characterized by minimal infiltration of immune cells and poor response to immunotherapies [165]. Moreover, sustained MAPK pathway activation can induce a senescence-associated secretory phenotype (SASP) that further modifies the TME, promoting the secretion of cytokines, chemokines and growth factors that enhance cancer cell viability and foster therapeutic resistance [166]. In BRAF V600E-mutant HGG, dual BRAF and MEK inhibition affects glioma plasticity, promoting an immunomodulatory phenotype with elevated PD-L1 expression and improving the synergy with ICIs [167]. Furthermore, TME can drive resistance upon combinatorial BRAF inhibitor and ICI treatment in brain metastatic acral melanoma [168]. A comprehensive understanding of the effects of the MAPK pathway inhibition in TME, along with tumor cells, will allow the rational design of novel therapeutic strategies suitable for brain tumors [29].

4.4. Toxicities Associated with MAPK Pathway Inhibitors

Targeted inhibition of the MAPK signaling cascade has significantly improved clinical outcomes in subsets of brain tumor patients. However, it is often associated with adverse effects and toxicities that affect treatment tolerability and limit long-term utilization [169,170,171,172,173,174,175,176,177]. BRAF inhibitors such as dabrafenib and vemurafenib are most commonly associated with cutaneous toxicities, including follicular or acneiform eruptions, xerosis, fatigue, and photosensitivity. While most dermatologic side effects are mild and manageable, treatment discontinuation may be required in cases of severe toxicity [169,170,171,172]. MEK inhibitors, including trametinib and selumetinib, also frequently cause dermatologic adverse events (rash, xerosis, paronychia). In addition, systemic toxicities such as fatigue and cardiovascular complications, including hypertension and bradycardia, have been reported. Given the risk of cardiotoxicity, especially in pediatric patients with CNS tumors, routine cardiac monitoring is strongly recommended [173,174]. Trk inhibitors, such as larotrectinib and entrectinib, target aberrant activation of Trk receptors resulting from NTRK gene fusions. Although generally well tolerated, these compounds have been linked to a spectrum of adverse effects, including gastrointestinal symptoms such as nausea, vomiting, diarrhea, hepatotoxicity, peripheral edema, cutaneous rashes, cardiac dysfunction, and neurological effects including dizziness, headache, and peripheral neuropathy [152,175,176,177].

5. Conclusions

The therapeutic targeting of the MAPK signaling pathway has clinical benefits in various CNS tumors, particularly in pLGGs that express the BRAF V600E mutation [113]. Monotherapy with BRAF or MEK inhibitors is often associated with drug resistance and substantial toxicities, while combination strategies, especially dual inhibition of RAF and MEK, have demonstrated superior efficacy [97,170]. For instance, the dabrafenib–trametinib combination has received FDA approval for BRAF V600E-mutant low-grade gliomas [154,178]. Drug resistance is frequently caused by reactivation of the RAF/MEK/ERK axis and alternative escape mechanisms from MAPK inhibition such as enhanced PI3K/AKT/mTOR signaling [79]. As a result, current efforts in preclinical models and early-phase clinical trials (e.g., NCT02023905, NCT02133183) focus on multi-targeted strategies, combining MAPK pathway inhibitors with inhibitors targeting parallel signaling pathways such as PI3K/AKT/mTOR. Resistance to MAPK inhibition may also arise through mechanisms that bypass the main signaling pathway, including activating mutations in PI3KC, AKT-mediated feedback loops, PTEN loss, mTOR upregulation, and autophagy-associated survival responses [22]. Therapeutic approaches focus on modulating apoptosis, disrupting tumor-associated metabolic reprogramming, regulating autophagy, and inhibiting phenotypic plasticity to enhance treatment efficacy [22,179,180,181]. Novel strategies aiming to reverse the immunosuppressive TME [182]. In this context, current approaches include the assessment of CAR T cells targeting tumor-associated antigens such as IL-13Rα2 and EGFRvIII, as well as inhibitors of PD-1/PD-L1 axis [183,184]. Consistently, PD-L1 overexpression in GBM has been associated with poor clinical outcomes [184,185,186]. Blockade of PD-1/PD-L1 interaction aims to suppress the PD-1-mediated inhibitory signaling, restore cytotoxic T-cell function and enhance anti-tumor immunity [187]. Current clinical trials assessing combinatorial immunotherapies involving ICIs, CAR T cells, and anti-angiogenic drugs (e.g., bevacizumab), showing promising preliminary results [28,188,189]. Combination approaches involving immunotherapies with MAPK pathway inhibition are being evaluated mostly to overcome resistance mechanisms [189,190]. Cytokine therapies are being explored for their potential to reactivate immune function within the TME. Such therapies have shown promise in augmenting immune responses without significant toxicities [188,191,192]. Moreover, inhibitors focused on metabolic reprogramming at the IDH1/2-mutant gliomas are under investigation [193]. Possible synergies of all these strategies with MAPK pathway inhibition in certain contexts could be proven highly beneficial, providing both sustained tumor suppression and enhanced TME immunomodulation.
Despite meaningful progress, the clinical management of high-grade brain tumors, such as GBM, continues to be hindered by tumor heterogeneity, adaptive resistance mechanisms, and the restrictive nature of the BBB. Moving forward, multimodal therapeutic strategies that address both the tumor and its surrounding microenvironment, along with personalized molecular profiling, will be critical for improving survival outcomes. The future of MAPK-targeted therapy in CNS tumors lies in precision medicine, with treatment paradigms tailored to each patient’s unique molecular and immunological landscape.

Author Contributions

Conceptualization, A.N.G., C.P. and C.A.; investigation, D.V., E.K., A.-I.G., A.M. and C.A.; writing—original draft preparation, D.V., E.K. and A.-I.G.; writing—review and editing, A.M., A.N.G., E.A.E.-H., C.P. and C.A.; supervision, C.A. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

No new data were created or analyzed in this study.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
2-HG2-hydroxyglutarate
ABCATP-binding cassette
AKTprotein kinase B (PKB)
ALKanaplastic lymphoma kinase
ARAFA-Raf proto-oncogene
ASIRage-standardized incidence rate
ATRXα-thalassemia intellectual disability X-linked
BBBblood–brain barrier
BCANbrevican
BCRbreakpoint cluster region
BCRPbreast cancer resistance protein
BRAFv-Raf murine sarcoma viral oncogene homolog B
BTBblood–tumor barrier
CARglioblastoma multiforme
CDK4cyclin-dependent kinase 4
CDKN2A/Bcyclin-dependent kinase inhibitor 2A/2B
CLCN6CLCN6 chloride voltage-gated channel
CLIP2CAP-Gly domain containing linker protein 2
CNScentral nervous system
CRAFproto-oncogene c-Raf
DHGGdiffuse high-grade glioma
DIGdesmoplastic infantile ganglioglioma
DLGGdiffuse low-grade glioma
EGFRepidermal growth factor receptor
ERKextracellular signal-regulated kinase
ETV6Ets variant transcription factor 6
FAM131Bfamily with sequence similarity 131 member B
FDAfood and drug administration
FGFRfibroblast growth factor receptor
FZR1fizzy and cell division cycle 20 related 1
GBDglobal burden of disease
GBMglioblastoma multiforme
GNA11guanine nucleotide-binding protein subunit alpha 11
GOPCGolgi-associated PDZ and coiled-coil motif-containing protein
GTF2Igeneral transcription factor II-I
GTPaseguanosine triphosphatase
HGGhigh-grade glioma
HQhydroxychloroquine
HRASHarvey rat sarcoma viral oncogene homolog
ICIimmune checkpoint inhibitor
IDHisocitrate dehydrogenase
IHGinfant-type hemispheric glioma
IL-13Rα2interleukine-13Rα2
KLF4Krüppel-like factor 4
KRASKirsten rat sarcoma viral oncogene homolog
LGGlow-grade glioma
LZTR1leucine zipper-like transcription regulator 1
MACF1microtubule actin crosslinking factor 1
MAPKmitogen-activated protein kinase
MEKmitogen-activated protein kinase extracellular signal-regulated kinase
METmet tyrosine-protein kinase
MKRN1makorin ring finger protein 1
MPNSTmalignant peripheral nerve sheath tumor
MRP multidrug resistance-associated protein
MYBv-myb avian myeloblastosis viral oncogene homolog
NACC2nucleus Accumbens-associated protein 2
NF1neurofibromin
NF1Anuclear transcription factor 1A (NF1A)
NFASCneurofascin
NOTCHneurogenic locus notch homolog
NRAS neuroblastoma rat sarcoma viral oncogene homolog
NTRKneurotrophic tyrosine receptor kinase
OLIG2oligodendrocyte transcription factor 2
PApilocytic astrocytoma
PD-1 programmed cell death-1
PDGFR-Aplatelet-derived growth factor receptor A
PD-L1programmed death-ligand 1
PFSprogression-free survival
P-gpp-glycoprotein
PI3Kphosphoinositide 3 kinase
PIK3CAphosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha
PNplexiform neurofibroma
PRKAR2Aprotein kinase cAMP-dependent type II regulatory subunit α
PROTACproteolysis-targeting chimera
PTPRZ1protein tyrosine phosphatase receptor type Z1
PXApleomorphic xanthoastrocytoma
QKIquaking homolog KH domain containing RNA binding
RAFrapidly accelerated fibrosarcoma
RASrat sarcoma virus oncogene
RB1retinoblastoma susceptibility gene
RNF130ring finger protein 130
ROS1ROS proto-oncogene 1
RTKreceptor tyrosine kinase
SASPsenescence-associated secretory phenotype
SHP2Src homology region 2 domain-containing phosphatase-2
SOX2SRY-box transcription factor 2
SPRED1sprouty-related EVH1 domain-containing protein 1
SRGAP3rho GTPase activating protein 3
STAT3signal transducer and activator of transcription 3
TACC3transforming acidic coiled-coil containing protein 3
TERTtelomerase reverse transcriptase
TMEtumor microenvironment
TMZtemozolomide
TPM3tropomyosin 3
TrkBtropomyosin receptor kinase B
WHOworld health organization

References

  1. Kim, S.; Son, Y.; Oh, J.; Kim, S.; Jang, W.; Lee, S.; Smith, L.; Pizzol, D.; Lee, J.; Lee, H.; et al. Global burden of brain and central nervous system cancer in 185 countries, and projections up to 2050: A population-based systematic analysis of GLOBOCAN 2022. J. Neurooncol. 2025, 175, 673–685. [Google Scholar] [CrossRef] [PubMed]
  2. Huang, J.; Chan, S.C.; Lok, V.; Zhang, L.; Lin, X.; Lucero-Prisno, D.E.; Xu, W.; Zheng, Z.-J.; Elcarte, E.; Withers, M.; et al. Disease burden, risk factors, and trends of primary central nervous system (CNS) cancer: A global study of registries data. Neuro Oncol. 2023, 25, 995–1005. [Google Scholar] [CrossRef] [PubMed]
  3. Aldape, K.; Brindle, K.M.; Chesler, L.; Chopra, R.; Gajjar, A.; Gilbert, M.R.; Gottardo, N.; Gutmann, D.H.; Hargrave, D.; Holland, E.C.; et al. Challenges to curing primary brain tumours. Nat. Rev. Clin. Oncol. 2019, 16, 509–520. [Google Scholar] [CrossRef] [PubMed]
  4. Zhao, X.; He, M.; Yang, R.; Geng, N.; Zhu, X.; Tang, N. The global, regional, and national brain and central nervous system cancer burden and trends from 1990 to 2021: An analysis based on the Global Burden of Disease Study 2021. Front. Neurol. 2025, 16, 1574614. [Google Scholar] [CrossRef]
  5. Angom, R.S.; Nakka, N.M.R.; Bhattacharya, S. Advances in glioblastoma therapy: An update on current approaches. Brain Sci. 2023, 13, 1536. [Google Scholar] [CrossRef]
  6. Wood, M.D.; Beadling, C.; Neff, T.; Moore, S.; Harrington, C.A.; Baird, L.; Corless, C. Molecular profiling of pre- and post-treatment pediatric high-grade astrocytomas reveals acquired increased tumor mutation burden in a subset of recurrences. Acta Neuropathol. Commun. 2023, 11, 143. [Google Scholar] [CrossRef]
  7. He, C.; Xu, K.; Zhu, X.; Dunphy, P.S.; Gudenas, B.; Lin, W.; Twarog, N.; Hover, L.D.; Kwon, C.H.; Kasper, L.H.; et al. Patient-derived models recapitulate heterogeneity of molecular signatures and drug response in pediatric high-grade glioma. Nat. Commun. 2021, 12, 4089. [Google Scholar] [CrossRef]
  8. Ullah, R.; Yin, Q.; Snell, A.H.; Wan, L.; 8. RAF-MEK-ERK pathway in cancer evolution and treatment. Semin. Cancer Biol. 2022, 85, 123–154. [Google Scholar] [CrossRef]
  9. Samatar, A.A.; Poulikakos, P.I. Targeting RAS-ERK signalling in cancer: Promises and challenges. Nat. Rev. Drug Discov. 2014, 13, 928–942. [Google Scholar] [CrossRef]
  10. Margolis, B.; Skolnik, E.Y. Activation of Ras by receptor tyrosine kinases. J. Am. Soc. Nephrol. 1994, 5, 1288–1299. [Google Scholar] [CrossRef]
  11. Marais, R.; Light, Y.; Paterson, H.F.; Marshall, C.J. Ras recruits Raf-1 to the plasma membrane for activation by tyrosine phosphorylation. EMBO J. 1995, 14, 3136–3145. [Google Scholar] [CrossRef] [PubMed]
  12. Crews, C.M.; Alessandrini, A.; Erikson, R.L. The primary structure of MEK, a protein kinase that phosphorylates the ERK gene product. Science 1992, 258, 478–480. [Google Scholar] [CrossRef] [PubMed]
  13. Jang, D.M.; Boxer, K.; Ha, B.H.; Tkacik, E.; Levitz, T.; Rawson, S.; Metivier, R.J.; Schmoker, A.; Jeon, H.; Eck, M.J.; et al. Cryo-EM structures of CRAF/MEK1/14-3-3 complexes in autoinhibited and open-monomer states reveal features of RAF regulation. Nat. Commun. 2025, 16, 8150. [Google Scholar] [CrossRef] [PubMed]
  14. Ma, Y.; Nicolet, J. Specificity models in MAPK cascade signaling. FEBS Open Bio 2023, 13, 1177–1192. [Google Scholar] [CrossRef]
  15. Wan, P.T.C.; Garnett, M.J.; Roe, S.M.; Lee, S.; Niculescu-Duvaz, D.; Good, V.M.; Jones, C.M.; Marshall, C.J.; Springer, C.J.; Barford, D.; et al. Mechanism of activation of the RAF-ERK signaling pathway by oncogenic mutations of B-RAF. Cell 2004, 116, 855–867. [Google Scholar] [CrossRef]
  16. 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]
  17. Hingorani, S.R.; Wang, L.; Multani, A.S.; Combs, C.; Deramaudt, T.B.; Hruban, R.H.; Rustgi, A.K.; Chang, S.; Tuveson, D.A. Trp53R172H and KrasG12D cooperate to promote chromosomal instability and widely metastatic pancreatic ductal adenocacinoma in mice. Cancer Cell 2005, 7, 469–483. [Google Scholar] [CrossRef]
  18. Sanchez-Vega, F.; Mina, M.; Armenia, J.; Chatila, W.K.; Luna, A.; La, K.C.; Dimitriadoy, S.; Liu, D.L.; Kantheti, H.S.; Saghafinia, S.; et al. Oncogenic Signaling Pathways in The Cancer Genome Atlas. Cell 2018, 173, 321–337.e10. [Google Scholar] [CrossRef]
  19. Jackson, E.L.; Willis, N.; Mercer, K.; Bronson, R.T.; Crowley, D.; Montoya, R.; Jacks, T.; Tuveson, D.A. Analysis of lung tumor initiation and progression using conditional expression of oncogenic K-ras. Genes Dev. 2001, 15, 3243–3248. [Google Scholar] [CrossRef]
  20. Cancer Genome Atlas Network. Genomic classification of cutaneous melanoma. Cell 2015, 161, 1681–1696. [Google Scholar] [CrossRef]
  21. Lee, S.; Rauch, J.; Kolch, W. Targeting MAPK signaling in cancer: Mechanisms of drug resistance and sensitivity. Int. J. Mol. Sci. 2020, 21, 1102. [Google Scholar] [CrossRef] [PubMed]
  22. Bahar, M.E.; Kim, H.J.; Kim, D.R. Targeting the RAS/RAF/MAPK pathway for cancer therapy: From mechanism to clinical studies. Signal Transduct. Target Ther. 2023, 8, 455. [Google Scholar] [CrossRef] [PubMed]
  23. Brazel, D.; Nagasaka, M. The development of amivantamab for the treatment of non-small cell lung cancer. Respir. Res. 2023, 24, 256. [Google Scholar] [CrossRef] [PubMed]
  24. Salles, D.; Santino, S.F.; Ribeiro, D.A.; Malinverni, A.C.M.; Stávale, J.N. The involvement of the MAPK pathway in pilocytic astrocytomas. Pathol. Res. Pract. 2022, 232, 153821. [Google Scholar] [CrossRef]
  25. Rodriguez Almaraz, E.; Guerra, G.A.; Al-Adli, N.N.; Young, J.S.; Dada, A.; Quintana, D.; Taylor, J.W.; Oberheim-Bush, N.A.; Clarke, J.L.; Butowski, N.A.; et al. Longitudinal profiling of IDH-mutant astrocytomas reveals acquired RAS-MAPK pathway mutations associated with inferior survival. Neurooncol. Adv. 2025, 7, vdaf024. [Google Scholar] [CrossRef]
  26. Zhang, T.; Ma, X.; Wang, Y.; Liu, H.; Chen, J.; Zhao, L.; Huang, Q.; Li, S.; Fang, Y.; Zhou, Z.; et al. Type II RAF inhibitor tovorafenib for the treatment of pediatric low-grade glioma. Expert Rev. Clin. Pharmacol. 2024, 17, 999–1008. [Google Scholar] [CrossRef]
  27. Li, F.; Bondra, K.M.; Wang, H.; Kurmashev, D.; Mukherjee, B.; Kanji, S.; Habib, A.A.; Chen, Y.; Zheng, S.; Burma, S.; et al. Dual inhibition of MAPK and TORC1 signaling retards development of radiation resistance in pediatric BRAFV600E glioma models. Neuro Oncol. 2025, 27, 1787–1800. [Google Scholar] [CrossRef]
  28. Beylerli, O.; Gareev, I.; Musaev, E.; Roumiantsev, S.; Chekhonin, V.; Ahmad, A.; Chao, Y.; Yang, G. New approaches to targeted drug therapy of intracranial tumors. Cell Death Discov. 2025, 11, 111. [Google Scholar] [CrossRef]
  29. Sigaud, R.; Brummer, T.; Kocher, D.; Milde, T.; Selt, F. MOST wanted: Navigating the MAPK-OIS-SASP-tumor microenvironment axis in primary pediatric low-grade glioma and preclinical models. Childs Nerv. Syst. 2024, 40, 3209–3221. [Google Scholar] [CrossRef]
  30. Sarkar, S.; DeYoung, T.; Ressler, H.; Chandler, W. Brain tumors: Development, drug resistance, and sensitization—An epigenetic approach. Epigenetics 2023, 18, 2237761. [Google Scholar] [CrossRef]
  31. Kuksis, M.; Gao, Y.; Tran, W.; Hoey, C.; Kiss, A.; Komorowski, A.S.; Dhaliwal, A.J.; Sahgal, A.; Das, S.; Chan, K.K.; et al. The incidence of brain metastases among patients with metastatic breast cancer: A systematic review and meta-analysis. Neuro Oncol. 2021, 23, 894–904. [Google Scholar] [CrossRef] [PubMed]
  32. Gillespie, C.S.; Mustafa, M.A.; Richardson, G.E.; Alam, A.M.; Lee, K.S.; Hughes, D.M.; Escriu, C.; Zakaria, R. Genomic alterations and the incidence of brain metastases in advanced and metastatic NSCLC: A systematic review and meta-analysis. J. Thorac. Oncol. 2023, 18, 1703–1713. [Google Scholar] [CrossRef] [PubMed]
  33. Sahm, F.; Brandner, S.; Bertero, L.; Capper, D.; French, P.J.; Figarella-Branger, D.; Giangaspero, F.; Haberler, C.; Hegi, M.E.; Kristensen, B.W.; et al. Molecular diagnostic tools for the World Health Organization (WHO) 2021 classification of gliomas, glioneuronal and neuronal tumors; an EANO guideline. Neuro Oncol. 2023, 25, 1731–1749. [Google Scholar] [CrossRef] [PubMed]
  34. Malbari, F. Pediatric neuro-oncology. Neurol. Clin. 2021, 39, 829–845. [Google Scholar] [CrossRef]
  35. Bale, T.A.; Rosenblum, M.K. The 2021 WHO classification of tumors of the central nervous system: An update on pediatric low-grade gliomas and glioneuronal tumors. Brain Pathol. 2022, 32, e13060. [Google Scholar] [CrossRef]
  36. Gritsch, S.; Batchelor, T.T.; Gonzalez Castro, L.N.G. Diagnostic, therapeutic, and prognostic implications of the 2021 World Health Organization classification of tumors of the central nervous system. Cancer 2022, 128, 47–58. [Google Scholar] [CrossRef]
  37. Ocasio, J.K.; Budd, K.M.; Roach, J.T.; Andrews, J.M.; Baker, S.J. Oncohistones and disrupted development in pediatric-type diffuse high-grade glioma. Cancer Metastasis Rev. 2023, 42, 367–388. [Google Scholar] [CrossRef]
  38. Liu, X.; McEachron, T.A.; Schwartzentruber, J.; Wu, G. Histone H3 mutations in pediatric brain tumors. Cold Spring Harb. Perspect. Biol. 2014, 6, a018689. [Google Scholar] [CrossRef]
  39. Bernstock, J.D.; Kang, K.-D.; Klinger, N.V.; Olsen, H.E.; Gary, S.; Totsch, S.K.; Ghajar-Rahimi, G.; Segar, D.; Thompson, E.M.; Darley-Usmar, V.; et al. Targeting oncometabolism to maximize immunotherapy in brain tumors. Oncogene 2022, 41, 2663–2671. [Google Scholar] [CrossRef]
  40. Eckel-Passow, J.E.; Lachance, D.H.; Molinaro, A.M.; Walsh, K.M.; Decker, P.A.; Sicotte, H.; Pekmezci, M.; Rice, T.; Kosel, M.L.; Smirnov, I.V.; et al. Glioma groups based on 1p/19q, IDH, and TERT promoter mutations in tumors. N. Engl. J. Med. 2015, 372, 2499–2508. [Google Scholar] [CrossRef]
  41. Patel, J.P.; Spiller, S.E.; Barker, E.D. Drug penetration in pediatric brain tumors: Challenges and opportunities. Pediatr. Blood Cancer 2021, 68, e28983. [Google Scholar] [CrossRef] [PubMed]
  42. Aryal, M.; Porter, T. Emerging therapeutic strategies for brain tumors. Neuromolecular Med. 2022, 24, 23–34. [Google Scholar] [CrossRef] [PubMed]
  43. Liu, Y.; Zhou, F.; Ali, H.; Lathia, J.D.; Chen, P. Immunotherapy for glioblastoma: Current state, challenges, and future perspectives. Cell. Mol. Immunol. 2024, 21, 1354–1375. [Google Scholar] [CrossRef] [PubMed]
  44. Khabibov, M.; Garifullin, A.; Boumber, Y.; Khaddour, K.; Fernandez, M.; Khamitov, F.; Khalikova, L.; Kuznetsova, N.; Kit, O.; Kharin, L. Signaling pathways and therapeutic approaches in glioblastoma multiforme. Int. J. Oncol. 2022, 60, 69. [Google Scholar] [CrossRef]
  45. Wang, J.; Pollard, K.; Allen, A.N.; Tomar, T.; Pijnenburg, D.; Yao, Z.; Rodriguez, F.J.; Pratilas, C.A. Combined inhibition of SHP2 and MEK is effective in models of NF1-deficient malignant peripheral nerve sheath tumors. Cancer Res. 2020, 80, 5367–5379. [Google Scholar] [CrossRef]
  46. Cai, J.; Jacob, S.; Kurupi, R.; Dalton, K.M.; Coon, C.; Greninger, P.; Egan, R.K.; Stein, G.T.; Murchie, E.; McClanaghan, J.; et al. High-risk neuroblastoma with NF1 loss of function is targetable using SHP2 inhibition. Cell Rep. 2022, 40, 111095. [Google Scholar] [CrossRef]
  47. Qin, A.; Musket, A.; Musich, P.R.; Schweitzer, J.B.; Xie, Q. Receptor tyrosine kinases as druggable targets in glioblastoma: Do signaling pathways matter? Neurooncol. Adv. 2021, 3, vdab133. [Google Scholar] [CrossRef]
  48. Baez-Rodriguez, S.M.; Ciubotaru, G.V.; Onose, G.; Sevastre, A.-S.; Sfredel, V.; Danoiu, S.; Dricu, A.; Tataranu, L.G. An overview of EGFR mechanisms and their implications in targeted therapies for glioblastoma. Int. J. Mol. Sci. 2023, 24, 11110. [Google Scholar] [CrossRef]
  49. Verhaak, R.G.W.; Hoadley, K.A.; Purdom, E.; Wang, V.; Qi, Y.; Wilkerson, M.D.; Miller, C.R.; Ding, L.; Golub, T.; Mesirov, J.P.; et al. Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell 2010, 17, 98–110. [Google Scholar] [CrossRef]
  50. Al-Ghabkari, A.; Huang, B.; Park, M. Aberrant MET receptor tyrosine kinase signaling in glioblastoma: Targeted therapy and future directions. Cells 2024, 13, 218. [Google Scholar] [CrossRef]
  51. Guerreiro Stucklin, A.S.; Ryall, S.; Fukuoka, K.; Zapotocky, M.; Lassaletta, A.; Li, C.; Bridge, T.; Kim, B.; Arnoldo, A.; Kowalski, P.E.; et al. Alterations in ALK/ROS1/NTRK/MET drive a group of infantile hemispheric gliomas. Nat. Commun. 2019, 10, 4343. [Google Scholar] [CrossRef] [PubMed]
  52. Picca, A.; Sansone, G.; Santonocito, O.S.; Mazzanti, C.M.; Sanson, M.; Di Stefano, A.L. Diffuse gliomas with FGFR3-TACC3 fusions: Oncogenic mechanisms, hallmarks, and therapeutic perspectives. Cancers 2023, 15, 5555. [Google Scholar] [CrossRef] [PubMed]
  53. Gambella, A.; Senetta, R.; Collemi, G.; Vallero, S.G.; Monticelli, M.; Cofano, F.; Zeppa, P.; Garbossa, D.; Pellerino, A.; Rudà, R.; et al. NTRK fusions in central nervous system tumors: A rare, but worthy target. Int. J. Mol. Sci. 2020, 21, 753. [Google Scholar] [CrossRef] [PubMed]
  54. Jones, D.T.W.; Hutter, B.; Jäger, N.; Korshunov, A.; Kool, M.; Warnatz, H.J.; Zichner, T.; Lambert, S.R.; Ryzhova, M.; Quang, D.A.K.; et al. Recurrent somatic alterations of FGFR1 and NTRK2 in pilocytic astrocytoma. Nat. Genet. 2013, 45, 927–932. [Google Scholar] [CrossRef]
  55. Tauziède-Espariat, A.; Beccaria, K.; Dangouloff-Ros, V.; Sievers, P.; Meurgey, A.; Pissaloux, D.; Appay, R.; Saffroy, R.; Grill, J.; Mariet, C.; et al. A comprehensive analysis of infantile central nervous system tumors to improve distinctive criteria for infant-type hemispheric glioma versus desmoplastic infantile ganglioglioma/astrocytoma. Brain Pathol. 2023, 33, e13182. [Google Scholar] [CrossRef]
  56. Zhu, T.; Xie, J.; He, H.; Li, H.; Tang, X.; Wang, S.; Li, Z.; Tian, Y.; Li, L.; Zhu, J.; et al. Phase separation underlies signaling activation of oncogenic NTRK fusions. Proc. Natl. Acad. Sci. USA 2023, 120, e2219589120. [Google Scholar] [CrossRef]
  57. Drilon, A.; Jenkins, C.; Iyer, S.; Schoenfeld, A.; Keddy, C.; Davare, M.A. ROS1-dependent cancers: Biology, diagnostics and therapeutics. Nat. Rev. Clin. Oncol. 2021, 18, 35–55. [Google Scholar] [CrossRef]
  58. Cancer Genome Atlas Research Network; Brat, D.J.; Verhaak, R.G.W.; Aldape, K.D.; Yung, W.K.; Salama, S.R.; Cooper, L.A.; Rheinbay, E.; Miller, C.R.; Vitucci, M.; et al. Comprehensive, integrative genomic analysis of diffuse lower-grade gliomas. N. Engl. J. Med. 2015, 372, 2481–2498. [Google Scholar] [CrossRef]
  59. The Cancer Genome Atlas Research Network. Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature 2008, 455, 1061–1068. [Google Scholar] [CrossRef]
  60. Zou, H.; Duan, Y.; Wei, D.; Zhang, Y.; Dai, J.; Li, J.; Li, X.; Zhou, J.; Liu, Z.; Jin, Z.; et al. Molecular features of pleomorphic xanthoastrocytoma. Hum. Pathol. 2019, 86, 38–48. [Google Scholar] [CrossRef]
  61. Yan, W.; Markegard, E.; Dharmaiah, S.; Urisman, A.; Drew, M.; Esposito, D.; Scheffzek, K.; Nissley, D.V.; McCormick, F.; Simanshu, D.K. Structural insights into the SPRED1-neurofibromin-KRAS complex and disruption of SPRED1-neurofibromin interaction by oncogenic EGFR. Cell Rep. 2020, 32, 107909. [Google Scholar] [CrossRef] [PubMed]
  62. Steklov, M.; Pandolfi, S.; Baietti, M.F.; Batiuk, A.; Carai, P.; Najm, P.; Zhang, M.; Jang, H.; Renzi, F.; Cai, Y.; et al. Mutations in LZTR1 drive human disease by dysregulating RAS ubiquitination. Science 2018, 362, 1177–1182. [Google Scholar] [CrossRef] [PubMed]
  63. Zhao, J.; Chen, C.; Zhang, L.; Jiang, J.; Shen, G.; Yu, R. A Hg(2+)-mediated label-free fluorescent sensing strategy based on G-quadruplex formation for selective detection of glutathione and cysteine. Analyst 2013, 138, 1713–1718. [Google Scholar] [CrossRef] [PubMed]
  64. Siebel, A.; Cubillos-Rojas, M.; Santos, R.C.; Schneider, T.; Bonan, C.D.; Bartrons, R.; Ventura, F.; de Oliveira, J.R.; Rosa, J.L. Contribution of S6K1/MAPK signaling pathways in the response to oxidative stress: Activation of RSK and MSK by hydrogen peroxide. PLoS ONE 2013, 8, e75523. [Google Scholar] [CrossRef]
  65. Ng, P.K.S.; Li, J.; Jeong, K.J.; Shao, S.; Chen, H.; Tsang, Y.H.; Sengupta, S.; Wang, Z.; Bhavana, V.H.; Tran, R.; et al. Systematic functional annotation of somatic mutations in cancer. Cancer Cell 2018, 33, 450–462.e10. [Google Scholar] [CrossRef]
  66. Jones, D.T.W.; Kocialkowski, S.; Liu, L.; Pearson, D.M.; Bäcklund, L.M.; Ichimura, K.; Collins, V.P. Tandem duplication producing a novel oncogenic BRAF fusion gene defines the majority of pilocytic astrocytomas. Cancer Res. 2008, 68, 8673–8677. [Google Scholar] [CrossRef]
  67. Sievert, A.J.; Jackson, E.M.; Gai, X.; Hakonarson, H.; Judkins, A.R.; Resnick, A.C.; Sutton, L.N.; Storm, P.B.; Shaikh, T.H.; Biegel, J.A. Duplication of 7q34 in pediatric low-grade astrocytomas detected by high-density SNP-based arrays results in a novel BRAF fusion gene. Brain Pathol. 2009, 19, 449–458. [Google Scholar] [CrossRef]
  68. Forshew, T.; Tatevossian, R.G.; Lawson, A.R.J.; Ma, J.; Neale, G.; Ogunkolade, B.W.; Jones, T.A.; Aarum, J.; Dalton, J.; Bailey, S.; et al. Activation of the ERK/MAPK pathway: A signature genetic defect in posterior fossa pilocytic astrocytomas. J. Pathol. 2009, 218, 172–181. [Google Scholar] [CrossRef]
  69. Kurani, H.; Gurav, M.; Shetty, O.; Chinnaswamy, G.; Moiyadi, A.; Gupta, T.; Jalali, R.; Epari, S. Pilocytic astrocytomas: BRAFV600E and BRAF fusion expression patterns in pediatric and adult age groups. Childs Nerv. Syst. 2019, 35, 1525–1536. [Google Scholar] [CrossRef]
  70. Zhang, J.; Wu, G.; Miller, C.P.; Tatevossian, R.G.; Dalton, J.D.; Tang, B.; Orisme, W.; Punchihewa, C.; Parker, M.; Qaddoumi, I.; et al. Whole-genome sequencing identifies genetic alterations in pediatric low-grade gliomas. Nat. Genet. 2013, 45, 602–612. [Google Scholar] [CrossRef]
  71. Tomić, T.T.; Olausson, J.; Wilzén, A.; Sabel, M.; Truvé, K.; Sjögren, H.; Dósa, S.; Tisell, M.; Lannering, B.; Enlund, F.; et al. A new GTF2I-BRAF fusion mediating MAPK pathway activation in pilocytic astrocytoma. PLoS ONE 2017, 12, e0175638. [Google Scholar] [CrossRef] [PubMed]
  72. Hanrahan, A.J.; Solit, D.B. BRAF—A tumour-agnostic drug target with lineage-specific dependencies. Nat. Rev. Clin. Oncol. 2024, 21, 224–247. [Google Scholar] [CrossRef] [PubMed]
  73. Poulikakos, P.I.; Sullivan, R.J.; Yaeger, R. Molecular pathways and mechanisms of BRAF in cancer therapy. Clin. Cancer Res. 2022, 28, 4618–4628. [Google Scholar] [CrossRef] [PubMed]
  74. Karoulia, Z.; Wu, Y.; Ahmed, T.A.; Xin, Q.; Bollard, J.; Krepler, C.; Wu, X.; Zhang, C.; Bollag, G.; Herlyn, M.; et al. An Integrated Model of RAF Inhibitor Action Predicts Inhibitor Activity against Oncogenic BRAF Signaling. Cancer Cell 2016, 30, 501–503, Erratum in Cancer Cell 2016, 30, 485–498. https://doi.org/10.1016/j.ccell.2016.06.024. [Google Scholar] [CrossRef]
  75. Cope, N.J.; Novak, B.; Liu, Z.; Cavallo, M.; Gunderwala, A.Y.; Connolly, M.; Wang, Z. Analyses of the Oncogenic BRAF D594G Variant Reveal a Kinase-Independent Function of BRAF in Activating MAPK Signaling. J. Biol. Chem. 2020, 295, 2407–2420. [Google Scholar] [CrossRef]
  76. Yde, C.W.; Sehested, A.; Mateu-Regué, À.; Østrup, O.; Scheie, D.; Nysom, K.; Nielsen, F.C.; Rossing, M. A new NFIA:RAF1 fusion activating the MAPK pathway in pilocytic astrocytoma. Cancer Genet. 2016, 209, 440–444. [Google Scholar] [CrossRef]
  77. Jain, P.; Fierst, T.M.; Han, H.J.; Smith, T.E.; Vakil, A.; Storm, P.B.; Resnick, A.C.; Waanders, A.J. CRAF gene fusions in pediatric low-grade gliomas define a distinct drug response based on dimerization profiles. Oncogene 2017, 36, 6348–6358. [Google Scholar] [CrossRef]
  78. Degirmenci, U.; Wang, M.; Hu, J. Targeting aberrant RAS/RAF/MEK/ERK signaling for cancer therapy. Cells 2020, 9, 198. [Google Scholar] [CrossRef]
  79. Yakubov, R.; Kaloti, R.; Persaud, P.; McCracken, A.; Zadeh, G.; Bunda, S. It’s all downstream from here: RTK/Raf/MEK/ERK pathway resistance mechanisms in glioblastoma. J. Neurooncol. 2025, 172, 327–345. [Google Scholar] [CrossRef]
  80. Hicks, H.M.; McKenna, L.R.; Espinoza, V.L.; Pozdeyev, N.; Pike, L.A.; Sams, S.B.; LaBarbera, D.; Reigan, P.; Raeburn, C.D.; Schweppe, R.E.; et al. Inhibition of BRAF and ERK1/2 has synergistic effects on thyroid cancer growth in vitro and in vivo. Mol. Carcinog. 2021, 60, 201–212. [Google Scholar] [CrossRef]
  81. Hyman, D.M.; Puzanov, I.; Subbiah, V.; Faris, J.E.; Chau, I.; Blay, J.-Y.; Wolf, J.; Raje, N.; Diamond, E.L.; Hollebecque, A.; et al. Vemurafenib in multiple nonmelanoma cancers with BRAF V600 mutations. N. Engl. J. Med. 2015, 373, 726–736. [Google Scholar] [CrossRef] [PubMed]
  82. Kaley, T.; Touat, M.; Subbiah, V.; Hollebecque, A.; Rodon, J.; Lockhart, A.C.; Keedy, V.; Bielle, F.; Hofheinz, R.D.; Joly, F.; et al. BRAF Inhibition in BRAFV600-Mutant Gliomas: Results from the VE-BASKET Study. J. Clin. Oncol. 2018, 36, 3477–3484. [Google Scholar] [CrossRef] [PubMed]
  83. Robinson, G.W.; Orr, B.A.; Gajjar, A. Complete Clinical Regression of a BRAF V600E-Mutant Pediatric Glioblastoma Multiforme after BRAF Inhibitor Therapy. BMC Cancer 2014, 14, 258. [Google Scholar] [CrossRef] [PubMed]
  84. Nicolaides, T.; Nazemi, K.J.; Crawford, J.; Kilburn, L.; Minturn, J.; Gajjar, A.; Gauvain, K.; Leary, S.; Dhall, G.; Aboian, M.; et al. Phase I Study of Vemurafenib in Children with Recurrent or Progressive BRAFV600E Mutant Brain Tumors: Pacific Pediatric Neuro-Oncology Consortium Study (PNOC-002). Oncotarget 2020, 11, 1942–1952. [Google Scholar] [CrossRef]
  85. Vaidhyanathan, S.; Mittapalli, R.K.; Sarkaria, J.N.; Elmquist, W.F. Factors Influencing the CNS Distribution of a Novel MEK-1/2 Inhibitor: Implications for Combination Therapy for Melanoma Brain Metastases. Drug Metab. Dispos. 2014, 42, 1292–1300. [Google Scholar] [CrossRef]
  86. Barbato, M.I.; Nashed, J.; Bradford, D.; Ren, Y.; Khasar, S.; Miller, C.P.; Zolnik, B.S.; Zhao, H.; Li, Y.; Bi, Y.; et al. U.S. FDA approval summary: Dabrafenib in combination with trametinib for BRAF V600E mutation-positive low-grade glioma. Clin. Cancer Res. 2024, 30, 1123–1130. [Google Scholar] [CrossRef]
  87. Brown, N.F.; Carter, T.; Kitchen, N.; Mulholland, P. Dabrafenib and trametinib in BRAFV600E mutated glioma. CNS Oncol. 2017, 6, 291–296. [Google Scholar] [CrossRef]
  88. Delord, J.P.; Robert, C.; Nyakas, M.; McArthur, G.A.; Kudchakar, R.; Mahipal, A.; Yamada, Y.; Sullivan, R.; Arance, A.; Kefford, R.F.; et al. Phase I Dose-Escalation and -Expansion Study of the BRAF Inhibitor Encorafenib (LGX818) in Metastatic BRAF-Mutant Melanoma. Clin. Cancer Res. 2017, 23, 5339–5348. [Google Scholar] [CrossRef]
  89. Schreck, K.C.; Strowd, R.E.; Nabors, L.B.; Ellingson, B.M.; Chang, M.; Tan, S.K.; Abdullaev, Z.; Turakulov, R.; Aldape, K.; Danda, N.; et al. Response rate and molecular correlates to encorafenib and binimetinib in BRAF-V600E–mutant high-grade glioma. Clin. Cancer Res. 2024, 30, 2048–2056. [Google Scholar] [CrossRef]
  90. Wang, J.; Gan, C.; Sparidans, R.W.; Wagenaar, E.; van Hoppe, S.; Beijnen, J.H.; Schinkel, A.H. P-Glycoprotein (MDR1/ABCB1) and Breast Cancer Resistance Protein (BCRP/ABCG2) Affect Brain Accumulation and Intestinal Disposition of Encorafenib in Mice. Pharmacol. Res. 2018, 129, 414–423. [Google Scholar] [CrossRef]
  91. Ryan, M.B.; Quade, B.; Schenk, N.; Fang, Z.; Zingg, M.; Cohen, S.E.; Swalm, B.M.; Li, C.; Özen, A.; Ye, C.; et al. The pan-RAF-MEK nondegrading molecular glue NST-628 is a potent and brain-penetrant inhibitor of the RAS-MAPK pathway with activity across diverse RAS- and RAF-driven cancers. Cancer Discov. 2024, 14, 1190–1205. [Google Scholar] [CrossRef] [PubMed]
  92. 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] [PubMed]
  93. Planchard, D.; Smit, E.F.; Groen, H.J.M.; Mazieres, J.; Besse, B.; Helland, Å.; Giannone, V.; D’Amelio, A.M., Jr.; Zhang, P.; Mookerjee, B.; et al. Dabrafenib plus Trametinib in Patients with Previously Untreated BRAFV600E-Mutant Metastatic Non-Small-Cell Lung Cancer: An Open-Label, Phase 2 Trial. Lancet Oncol. 2017, 18, 1307–1316. [Google Scholar] [CrossRef] [PubMed]
  94. Subbiah, V.; Kreitman, R.J.; Wainberg, Z.A.; Cho, J.Y.; Schellens, J.H.M.; Soria, J.C.; Wen, P.Y.; Zielinski, C.; Cabanillas, M.E.; Urbanowitz, G.; et al. Dabrafenib and Trametinib Treatment in Patients with Locally Advanced or Metastatic BRAF V600-Mutant Anaplastic Thyroid Cancer. J. Clin. Oncol. 2018, 36, 7–13. [Google Scholar] [CrossRef]
  95. Casey, D.; Demko, S.; Sinha, A.; Mishra-Kalyani, P.S.; Shen, Y.L.; Khasar, S.; Goheer, M.A.; Helms, W.S.; Pan, L.; Xu, Y.; et al. FDA approval summary: Selumetinib for plexiform neurofibroma. Clin. Cancer Res. 2021, 27, 4142–4146. [Google Scholar] [CrossRef]
  96. Pan, Y.; Hysinger, J.D.; Barron, T.; Schindler, N.F.; Cobb, O.; Guo, X.; Yalçın, B.; Anastasaki, C.; Mulinyawe, S.B.; Ponnuswami, A.; et al. NF1 mutation drives neuronal activity-dependent initiation of optic glioma. Nature 2021, 594, 277–282. [Google Scholar] [CrossRef]
  97. Adamopoulos, C.; Ahmed, T.A.; Tucker, M.R.; Ung, P.M.U.; Xiao, M.; Karoulia, Z.; Amabile, A.; Wu, X.; Aaronson, S.A.; Ang, C.; et al. Exploiting allosteric properties of RAF and MEK inhibitors to target therapy-resistant tumors driven by oncogenic BRAF signaling. Cancer Discov. 2021, 11, 1716–1735. [Google Scholar] [CrossRef]
  98. Fangusaro, J.; Onar-Thomas, A.; Poussaint, T.Y.; Wu, S.; Ligon, A.H.; Lindeman, N.; Banerjee, A.; Packer, R.J.; Kilburn, L.B.; Goldman, S.; et al. Selumetinib in paediatric patients with BRAF-aberrant or neurofibromatosis type 1-associated recurrent, refractory, or progressive low-grade glioma: A multicentre, phase 2 trial. Lancet Oncol. 2019, 20, 1011–1022. [Google Scholar] [CrossRef]
  99. Fangusaro, J.; Onar-Thomas, A.; Poussaint, T.Y.; Wu, S.; Ligon, A.H.; Lindeman, N.; Campagne, O.; Banerjee, A.; Gururangan, S.; Kilburn, L.B.; et al. A phase II trial of selumetinib in children with recurrent optic pathway and hypothalamic low-grade glioma without NF1: A Pediatric Brain Tumor Consortium study. Neuro Oncol. 2021, 23, 1777–1788. [Google Scholar] [CrossRef]
  100. Fangusaro, J.; Onar-Thomas, A.; Poussaint, T.Y.; Lensing, S.; Ligon, A.H.; Lindeman, N.; Banerjee, A.; Kilburn, L.B.; Lenzen, A.; Pillay-Smiley, N.; et al. A Phase 2 PBTC Study of Selumetinib for Recurrent/Progressive Pediatric Low-Grade Glioma: Strata 2, 5, and 6 with Long-Term Outcomes on Strata 1, 3, and 4. Neuro Oncol. 2025, 27, 2415–2428. [Google Scholar] [CrossRef]
  101. Wen, P.Y.; Stein, A.; van den Bent, M.; De Greve, J.; Wick, A.; de Vos, F.Y.F.L.; von Bubnoff, N.; van Linde, M.E.; Lai, A.; Prager, G.W.; et al. Dabrafenib plus trametinib in patients with BRAFV600E-mutant low-grade and high-grade glioma (ROAR): A multicenter, open-label, single-arm, phase 2, basket trial. Lancet Oncol. 2022, 23, 53–64. [Google Scholar] [CrossRef] [PubMed]
  102. Perreault, S.; Larouche, V.; Tabori, U.; Hawkin, C.; Lippé, S.; Ellezam, B.; Décarie, J.C.; Théoret, Y.; Métras, M.É.; Sultan, S.; et al. A phase 2 study of trametinib for patients with pediatric glioma or plexiform neurofibroma with refractory tumor and activation of the MAPK/ERK pathway: TRAM-01. BMC Cancer 2019, 19, 1250. [Google Scholar] [CrossRef] [PubMed]
  103. Wilcox, J.A.; Boire, A.A. Leveraging Molecular and Immune-Based Therapies in Leptomeningeal Metastases. CNS Drugs 2023, 37, 45–67. [Google Scholar] [CrossRef]
  104. Touat, M.; Smith, K.A.; Freese, K.; Poussaint, T.Y.; Wallace, D.W.; Parker, N.R.; Qaddoumi, I.; Stewart, C.F.; Fangusaro, J.; Patel, K.S.; et al. Vemurafenib and cobimetinib overcome resistance to vemurafenib in BRAF-mutant ganglioglioma. Neurology 2018, 91, 523–525. [Google Scholar] [CrossRef] [PubMed]
  105. Hoy, S.M. Mirdametinib: First Approval. Drugs 2025, 85, 977–984. [Google Scholar] [CrossRef]
  106. Moertel, C.L.; Hirbe, A.C.; Shuhaiber, H.H.; Bielamowicz, K.; Sidhu, A.; Viskochil, D.; Weber, M.D.; Lokku, A.; Smith, L.M.; Foreman, N.K.; et al. ReNeu: A pivotal, phase IIb trial of mirdametinib in adults and children with symptomatic neurofibromatosis 1-associated plexiform neurofibroma. J. Clin. Oncol. 2025, 43, JCO2401034. [Google Scholar] [CrossRef]
  107. Jaiswal, B.S.; Durinck, S.; Stawiski, E.W.; Yin, J.; Wang, W.; Lin, E.; Moffat, J.; Martin, S.E.; Modrusan, Z.; Seshagiri, S.; et al. ERK Mutations and Amplification Confer Resistance to ERK-Inhibitor Therapy. Clin. Cancer Res. 2018, 24, 4044–4055. [Google Scholar] [CrossRef]
  108. Sigaud, R.; Rösch, L.; Gatzweiler, C.; Benzel, J.; von Soosten, L.; Peterziel, H.; Selt, F.; Najafi, S.; Ayhan, S.; Gerloff, X.F.; et al. The first-in-class ERK inhibitor ulixertinib shows promising activity in mitogen-activated protein kinase (MAPK)-driven pediatric low-grade glioma models. Neuro Oncol. 2023, 25, 566–579. [Google Scholar] [CrossRef]
  109. Ahmed, T.A.; Adamopoulos, C.; Karoulia, Z.; Wu, X.; Sachidanandam, R.; Aaronson, S.A.; Poulikakos, P.I. SHP2 Drives Adaptive Resistance to ERK Signaling Inhibition in Molecularly Defined Subsets of ERK-Dependent Tumors. Cell Rep. 2019, 26, 65–78.e5. [Google Scholar] [CrossRef]
  110. Sait, S.F.; Tang, K.H.; Angus, S.P.; Brown, R.; Sun, D.; Xie, X.; Iltis, C.; Lien, M.; Socci, N.D.; Bale, T.A.; et al. Hydroxychloroquine prevents resistance and potentiates the antitumor effect of SHP2 inhibition in NF1-associated malignant peripheral nerve sheath tumors. Proc. Natl. Acad. Sci. USA 2025, 122, e2407745121. [Google Scholar] [CrossRef]
  111. Sang, Y.; Hou, Y.; Cheng, R.; Zheng, L.; Xu, M.; Zhang, X.; Yang, N.; Zhang, S.; Zhang, C.; Zhang, Y. Targeting PDGFRα-Activated Glioblastoma through Specific Inhibition of SHP2-Mediated Signaling. Neuro Oncol. 2019, 21, 1423–1435. [Google Scholar] [CrossRef] [PubMed]
  112. Grossauer, S.; Koeck, K.; Murphy, N.E.; Meyers, I.D.; Daynac, M.; Truffaux, N.; Truong, A.Y.; Nicolaides, T.P.; McMahon, M.; Berger, M.S.; et al. Concurrent MEK targeted therapy prevents MAPK pathway reactivation during BRAFV600E targeted inhibition in a novel syngeneic murine glioma model. Oncotarget 2016, 7, 75839–75853. [Google Scholar] [CrossRef] [PubMed]
  113. O’Hare, P.; Cooney, T.; de Blank, P.; Gutmann, D.H.; Kieran, M.; Milde, T.; Fangusaro, J.; Fisher, M.J.; Avula, S.; Packer, R.; et al. Resistance, rebound, and recurrence regrowth patterns in pediatric low-grade glioma treated by MAPK inhibition: A modified Delphi approach to build international consensus-based definitions-International Pediatric Low-Grade Glioma Coalition. Neuro Oncol. 2024, 26, 1357–1366. [Google Scholar] [CrossRef] [PubMed]
  114. Bouffet, E.; Hansford, J.R.; Garrè, M.L.; Hara, J.; Plant-Fox, A.; Aerts, I.; Locatelli, F.; van der Lugt, J.; Papusha, L.; Sahm, F.; et al. Dabrafenib plus trametinib in pediatric glioma with BRAF V600 mutations. N. Engl. J. Med. 2023, 389, 1108–1120. [Google Scholar] [CrossRef] [PubMed]
  115. Lei, J.; Liu, Y.; Fan, Y. The effects of dabrafenib and/or trametinib treatment in Braf V600-mutant glioma: A systematic review and meta-analysis. Neurosurg Rev. 2024, 47, 458. [Google Scholar] [CrossRef]
  116. Algazi, A.P.; Moon, J.; Lao, C.D.; Chmielowski, B.; Kendra, K.L.; Lewis, K.D.; Gonzalez, R.; Kim, K.; Godwin, J.E.; Curti, B.D.; et al. A Phase 1 Study of Triple-Targeted Therapy with BRAF, MEK, and AKT Inhibitors for Patients with BRAF-Mutated Cancers. Cancer 2024, 130, 1784–1796. [Google Scholar] [CrossRef]
  117. McNeill, R.S.; Canoutas, D.A.; Stuhlmiller, T.J.; Dhruv, H.D.; Irvin, D.M.; Bash, R.E.; Angus, S.P.; Herring, L.E.; Simon, J.M.; Skinner, K.R.; et al. Combination Therapy with Potent PI3K and MAPK Inhibitors Overcomes Adaptive Kinome Resistance to Single Agents in Preclinical Models of Glioblastoma. Neuro Oncol. 2017, 19, 1469–1480. [Google Scholar] [CrossRef]
  118. Das, A.; Fernandez, N.R.; Levine, A.; Bianchi, V.; Stengs, L.K.; Chung, J.; Negm, L.; Dimayacyac, J.R.; Chang, Y.; Nobre, L.; et al. Combined immunotherapy improves outcome for replication-repair-deficient high-grade glioma failing anti–PD-1 monotherapy: A report from the International RRD Consortium. Cancer Discov. 2024, 14, 258–273. [Google Scholar] [CrossRef]
  119. Frederick, D.T.; Piris, A.; Cogdill, A.P.; Cooper, Z.A.; Lezcano, C.; Ferrone, C.R.; Mitra, D.; Boni, A.; Newton, L.P.; Liu, C.; et al. BRAF Inhibition Is Associated with Enhanced Melanoma Antigen Expression and a More Favorable Tumor Microenvironment in Patients with Metastatic Melanoma. Clin. Cancer Res. 2013, 19, 1225–1231. [Google Scholar] [CrossRef]
  120. Capogiri, M.; De Micheli, A.J.; Lassaletta, A.; Muñoz, D.P.; Coppé, J.P.; Mueller, S.; Guerreiro Stucklin, A.S. Response and Resistance to BRAFV600E Inhibition in Gliomas: Roadblocks Ahead? Front. Oncol. 2023, 12, 1074726. [Google Scholar] [CrossRef]
  121. Woo, P.Y.M.; Lam, T.-C.; Pu, J.K.S.; Li, L.-F.; Leung, R.C.Y.; Ho, J.M.K.; Zhung, J.T.F.; Wong, B.; Chan, T.S.K.; Loong, H.H.F.; et al. Regression of BRAF V600E mutant adult glioblastoma after primary combined BRAF–MEK inhibitor targeted therapy: A report of two cases. Oncotarget 2019, 10, 3818–3826. [Google Scholar] [CrossRef] [PubMed]
  122. Arbour, G.; Ellezam, B.; Weil, A.G.; Cayrol, R.; Vanan, M.I.; Coltin, H.; Larouche, V.; Erker, C.; Jabado, N.; Perreault, S.; et al. Upfront BRAF/MEK inhibitors for treatment of high-grade glioma: A case report and review of the literature. Neuro Oncol. Adv. 2022, 4, vdac174. [Google Scholar] [CrossRef] [PubMed]
  123. Fusco, M.J.; Piña, Y.; Macaulay, R.J.; Sahebjam, S.; Forsyth, P.A.; Peguero, E.; Walko, C.M. Durable progression-free survival with the use of BRAF and MEK inhibitors in four cases with BRAF V600E-mutated gliomas. Cancer Control 2021, 28, 10732748211040013. [Google Scholar] [CrossRef] [PubMed]
  124. Pekmezci, M.; Villanueva-Meyer, J.E.; Goode, B.; Van Ziffle, J.; Onodera, C.; Grenert, J.P.; Bastian, B.C.; Chamyan, G.; Maher, O.M.; Khatib, Z.; et al. The Genetic Landscape of Ganglioglioma. Acta Neuropathol. Commun. 2018, 6, 47. [Google Scholar] [CrossRef]
  125. Chatterjee, D.; Garg, C.; Singla, N.; Radotra, B.D. Desmoplastic Non-Infantile Astrocytoma/Ganglioglioma: Rare Low-Grade Tumor with Frequent BRAF V600E Mutation. Hum. Pathol. 2018, 80, 186–191. [Google Scholar] [CrossRef]
  126. Pollack, I.; Agnihotri, S.; Broniscer, A. Childhood Brain Tumours: Current Management, Biological Insights and Future Directions. J. Neurosurg. Pediatr. 2019, 23, 261–273. [Google Scholar] [CrossRef]
  127. Manoranjan, B.; Venugopal, C.; McFarlane, N.; Doble, B.W.; Dunn, S.E.; Scheinemann, K.; Singh, S.K. Medulloblastoma Stem Cells: Where Development and Cancer Cross Pathways. Pediatr. Res. 2012, 71, 516–522. [Google Scholar] [CrossRef]
  128. Merve, A.; Dubuc, A.M.; Zhang, X.; Remke, M.; Baxter, P.A.; Li, X.N.; Taylor, M.D.; Marino, S. Polycomb Group Gene BMI1 Controls Invasion of Medulloblastoma Cells and Inhibits BMP-Regulated Cell Adhesion. Acta Neuropathol. Commun. 2014, 2, 10. [Google Scholar] [CrossRef]
  129. Badodi, S.; Pomella, N.; Lim, Y.M.; Brandner, S.; Morrison, G.; Pollard, S.M.; Zhang, X.; Zabet, N.R.; Marino, S. Combination of BMI1 and MAPK/ERK Inhibitors Is Effective in Medulloblastoma. Neuro Oncol. 2022, 24, 1273–1285. [Google Scholar] [CrossRef]
  130. Kilburn, L.B.; Khuong-Quang, D.-A.; Hansford, J.R.; Landi, D.; van der Lugt, J.; Leary, S.E.S.; Driever, P.H.; Bailey, S.; Perreault, S.; McCowage, G.; et al. The type II RAF inhibitor tovorafenib in relapsed/refractory pediatric low-grade glioma: The phase 2 FIREFLY-1 trial. Nat. Med. 2024, 30, 207–217. [Google Scholar] [CrossRef]
  131. Pardridge, W.M. The Blood–Brain Barrier: Bottleneck in Brain Drug Development. NeuroRx 2005, 2, 3–14. [Google Scholar] [CrossRef]
  132. Abbott, N.J.; Rönnbäck, L.; Hansson, E. Astrocyte–Endothelial Interactions at the Blood–Brain Barrier. Nat. Rev. Neurosci. 2006, 7, 41–53. [Google Scholar] [CrossRef] [PubMed]
  133. Arvanitis, C.D.; Ferraro, G.B.; Jain, R.K. The Blood–Brain Barrier and Blood–Tumour Barrier in Brain Tumours and Metastases. Nat. Rev. Cancer 2020, 20, 26–41. [Google Scholar] [CrossRef] [PubMed]
  134. Groothuis, D.R. The Blood–Brain and Blood–Tumor Barriers: A Review of Strategies for Increasing Drug Delivery. Neuro Oncol. 2000, 2, 45–59. [Google Scholar] [CrossRef] [PubMed]
  135. van Tellingen, O.; Yetkin-Arik, B.; de Gooijer, M.C.; Wesseling, P.; Wurdinger, T.; de Vries, H.E. Overcoming the Blood–Brain Tumor Barrier for Effective Glioblastoma Treatment. Drug Resist. Updat. 2015, 19, 1–12. [Google Scholar] [CrossRef]
  136. Mittapalli, R.K.; Vaidhyanathan, S.; Sane, R.; Elmquist, W.F. Impact of P-Glycoprotein (ABCB1) and Breast Cancer Resistance Protein (ABCG2) on the Brain Distribution of a Novel BRAF Inhibitor: Vemurafenib (PLX4032). J. Pharmacol. Exp. Ther. 2012, 342, 33–40. [Google Scholar] [CrossRef]
  137. Osswald, M.; Blaes, J.; Liao, Y.; Solecki, G.; Gömmel, M.; Berghoff, A.S.; Salphati, L.; Wallin, J.J.; Phillips, H.S.; Wick, W.; et al. Impact of Blood–Brain Barrier Integrity on Tumor Growth and Therapy Response in Brain Metastases. Clin. Cancer Res. 2016, 22, 6078–6087. [Google Scholar] [CrossRef]
  138. Stacy, A.E.; Jansson, P.J.; Richardson, D.R. Molecular Pharmacology of ABCG2 and Its Role in Chemoresistance. Mol. Pharmacol. 2013, 84, 655–669. [Google Scholar] [CrossRef]
  139. Gomez-Zepeda, D.; Taghi, M.; Scherrmann, J.M.; Decleves, X.; Menet, M.C. ABC Transporters at the Blood–Brain Interfaces, Their Study Models, and Drug Delivery Implications in Gliomas. Pharmaceutics 2019, 12, 20. [Google Scholar] [CrossRef]
  140. Agarwal, S.; Sane, R.; Gallardo, J.L.; Ohlfest, J.R.; Elmquist, W.F. Distribution of Gefitinib to the Brain Is Limited by P-Glycoprotein (ABCB1) and Breast Cancer Resistance Protein (ABCG2)-Mediated Active Efflux. J. Pharmacol. Exp. Ther. 2010, 334, 147–155. [Google Scholar] [CrossRef]
  141. de Vries, N.A.; Buckle, T.; Zhao, J.; Beijnen, J.H.; Schellens, J.H.; van Tellingen, O. Restricted Brain Penetration of the Tyrosine Kinase Inhibitor Erlotinib Due to the Drug Transporters P-Gp and BCRP. Investig. New Drugs 2012, 30, 443–449. [Google Scholar] [CrossRef] [PubMed]
  142. Batra, U.; Lokeshwar, N.; Gupta, S.; Shirsath, P. Role of Epidermal Growth Factor Receptor-Tyrosine Kinase Inhibitors in the Management of Central Nervous System Metastases in EGFR Mutation-Positive Non-Small Cell Lung Cancer Patients. Indian J. Cancer 2017, 54, S37–S44. [Google Scholar] [CrossRef] [PubMed]
  143. Hu, S.; Chen, Z.; Franke, R.; Orwick, S.; Zhao, M.; Rudek, M.A.; Sparreboom, A.; Baker, S.D. Interaction of the Multikinase Inhibitors Sorafenib and Sunitinib with Solute Carriers and ATP-Binding Cassette Transporters. Clin. Cancer Res. 2009, 15, 6062–6069. [Google Scholar] [CrossRef] [PubMed]
  144. de Gooijer, M.C.; Zhang, P.; Weijer, R.; Buil, L.C.M.; Beijnen, J.H.; van Tellingen, O. The Impact of P-Glycoprotein and Breast Cancer Resistance Protein on the Brain Pharmacokinetics and Pharmacodynamics of a Panel of MEK Inhibitors. Int. J. Cancer 2018, 142, 381–391. [Google Scholar] [CrossRef] [PubMed]
  145. Gampa, G.; Kim, M.; Cook-Rostie, N.; Laramy, J.K.; Sarkaria, J.N.; Paradiso, L.; DePalatis, L.; Elmquist, W.F. Brain Distribution of a Novel MEK Inhibitor E6201: Implications in the Treatment of Melanoma Brain Metastases. Drug Metab. Dispos. 2018, 46, 658–666. [Google Scholar] [CrossRef]
  146. Sabari, J.K.; Velcheti, V.; Shimizu, K.; Strickland, M.R.; Heist, R.S.; Singh, M.; Nayyar, N.; Giobbie-Hurder, A.; Digumarthy, S.R.; Gainor, J.F.; et al. Activity of Adagrasib (MRTX849) in Brain Metastases: Preclinical Models and Clinical Data from Patients with KRASG12C-Mutant Non-Small Cell Lung Cancer. Clin. Cancer Res. 2022, 28, 3318–3328. [Google Scholar] [CrossRef]
  147. Rijmers, J.; Retmana, I.A.; Bui, V.; Arguedas, D.; Lebre, M.C.; Sparidans, R.W.; Beijnen, J.H.; Schinkel, A.H. ABCB1 Attenuates Brain Exposure to the KRASG12C Inhibitor Opnurasib Whereas Binding to Mouse Carboxylesterase 1c Influences Its Plasma Exposure. Biomed. Pharmacother. 2024, 175, 116720. [Google Scholar] [CrossRef]
  148. Horbinski, C. To BRAF or Not to BRAF: Is That Even a Question Anymore? J. Neuropathol. Exp. Neurol. 2013, 72, 2–7. [Google Scholar] [CrossRef]
  149. Ryall, S.; Zapotocky, M.; Fukuoka, K.; Nobre, L.; Guerreiro Stucklin, A.; Bennett, J.; Siddaway, R.; Li, C.; Pajovic, S.; Arnoldo, A.; et al. Integrated Molecular and Clinical Analysis of 1,000 Pediatric Low-Grade Gliomas. Cancer Cell 2020, 37, 569–583.e5. [Google Scholar] [CrossRef]
  150. Lhermitte, B.; Wolf, T.; Chenard, M.P.; Coca, A.; Todeschi, J.; Proust, F.; Hirsch, E.; Schott, R.; Noel, G.; Guerin, E.; et al. Molecular Heterogeneity in BRAF-Mutant Gliomas: Diagnostic, Prognostic, and Therapeutic Implications. Cancers 2023, 15, 1268. [Google Scholar] [CrossRef]
  151. Lazo De La Vega, L.; Comeau, H.; Sallan, S.; Al-Ibraheemi, A.; Gupta, H.; Li, Y.Y.; Tsai, H.K.; Kang, W.; Ward, A.; Church, A.J.; et al. Rare FGFR Oncogenic Alterations in Sequenced Pediatric Solid and Brain Tumors Suggest FGFR Is a Relevant Molecular Target in Childhood Cancer. JCO Precis. Oncol. 2022, 6, e2200390. [Google Scholar] [CrossRef] [PubMed]
  152. Kim, E.E.; Park, C.K.; Kim, S.K.; Phi, J.H.; Paek, S.H.; Choi, J.Y.; Kang, H.J.; Lee, J.H.; Won, J.K.; Yun, H.; et al. NTRK-Fused Central Nervous System Tumours: Clinicopathological and Genetic Insights and Response to TRK Inhibitors. Acta Neuropathol. Commun. 2024, 12, 118. [Google Scholar] [CrossRef] [PubMed]
  153. Pereira, B.J.A.; Oba-Shinjo, S.M.; de Almeida, A.N.; Marie, S.K.N. Molecular Alterations in Meningiomas: Literature Review. Clin. Neurol. Neurosurg. 2019, 176, 89–96. [Google Scholar] [CrossRef] [PubMed]
  154. Vamvoukaki, R.; Chrysoulaki, M.; Betsi, G.; Xekouki, P. Pituitary Tumorigenesis—Implications for Management. Medicina 2023, 59, 812. [Google Scholar] [CrossRef]
  155. Han, C.H.; Brastianos, P.K. Genetic Characterization of Brain Metastases in the Era of Targeted Therapy. Front. Oncol. 2017, 7, 230. [Google Scholar] [CrossRef]
  156. Muniz, T.P.; Mason, W.P. BRAF Mutations in CNS Tumors—Prognostic Markers and Therapeutic Targets. CNS Drugs 2023, 37, 587–598. [Google Scholar] [CrossRef]
  157. Zhang, Z.; Richmond, A.; Yan, C. Immunomodulatory Properties of PI3K/AKT/mTOR and MAPK/MEK/ERK Inhibition Augment Response to Immune Checkpoint Blockade in Melanoma and Triple-Negative Breast Cancer. Int. J. Mol. Sci. 2022, 23, 7353. [Google Scholar] [CrossRef]
  158. Bergholz, J.S.; Zhao, J.J. How Compensatory Mechanisms and Adaptive Rewiring Have Shaped Our Understanding of Therapeutic Resistance in Cancer. Cancer Res. 2021, 81, 6074–6077. [Google Scholar] [CrossRef]
  159. Riddick, G.; Kotliarova, S.; Rodriguez, V.; Kim, H.S.; Linkous, A.; Storaska, A.J.; Ahn, S.; Walling, J.; Belova, G.; Fine, H.A. A Core Regulatory Circuit in Glioblastoma Stem Cells Links MAPK Activation to a Transcriptional Program of Neural Stem Cell Identity. Sci. Rep. 2017, 7, 43605. [Google Scholar] [CrossRef]
  160. Zaman, A.; Wu, W.; Bivona, T.G. Targeting Oncogenic BRAF: Past, Present, and Future. Cancers 2019, 11, 1197. [Google Scholar] [CrossRef]
  161. Luo, W. Nasopharyngeal carcinoma ecology theory: Cancer as multidimensional spatiotemporal “unity of ecology and evolution” pathological ecosystem. Theranostics 2023, 13, 1607–1631. [Google Scholar] [CrossRef] [PubMed]
  162. Peng, W.; Chen, J.Q.; Liu, C.; Malu, S.; Creasy, C.; Tetzlaff, M.T.; Xu, C.; McKenzie, J.A.; Zhang, C.; Liang, X.; et al. Loss of PTEN Promotes Resistance to T Cell-Mediated Immunotherapy. Cancer Discov. 2016, 6, 202–216. [Google Scholar] [CrossRef] [PubMed]
  163. Coelho, M.A.; de Carné Trécesson, S.; Rana, S.; Zecchin, D.; Moore, C.; Molina-Arcas, M.; East, P.; Spencer-Dene, B.; Nye, E.; Barnouin, K.; et al. Oncogenic RAS Signaling Promotes Tumor Immunoresistance by Stabilizing PD-L1 mRNA. Immunity 2017, 47, 1083–1099.e6. [Google Scholar] [CrossRef] [PubMed]
  164. Lailler, C.; Louandre, C.; Morisse, M.C.; Lhossein, T.; Godin, C.; Lottin, M.; Constans, J.M.; Chauffert, B.; Galmiche, A.; Saidak, Z. ERK1/2 Signaling Regulates the Immune Microenvironment and Macrophage Recruitment in Glioblastoma. Biosci. Rep. 2019, 39, BSR20191433. [Google Scholar] [CrossRef]
  165. Lin, H.; Liu, C.; Hu, A.; Zhang, D.; Yang, H.; Mao, Y. Understanding the immunosuppressive microenvironment of glioma: Mechanistic insights and clinical perspectives. J. Hematol. Oncol. 2024, 17, 31. [Google Scholar] [CrossRef]
  166. Sigaud, R.; Albert, T.K.; Hess, C.; Hielscher, T.; Winkler, N.; Kocher, D.; Walter, C.; Münter, D.; Selt, F.; Usta, D.; et al. MAPK inhibitor sensitivity scores predict sensitivity driven by the immune infiltration in pediatric low-grade gliomas. Nat. Commun. 2023, 14, 4533. [Google Scholar] [CrossRef]
  167. Xing, Y.L.; Panovska, D.; Park, J.W.; Grossauer, S.; Koeck, K.; Bui, B.; Nasajpour, E.; Nirschl, J.J.; Feng, Z.P.; Cheung, P.; et al. BRAF/MEK Inhibition Induces Cell State Transitions Boosting Immune Checkpoint Sensitivity in BRAFV600E-Mutant Glioma. Cell Rep. Med. 2025, 6, 102183. [Google Scholar] [CrossRef]
  168. Lee, R.J.; Khandelwal, G.; Baenke, F.; Cannistraci, A.; Macleod, K.; Mundra, P.; Ashton, G.; Mandal, A.; Viros, A.; Gremel, G.; et al. Brain Microenvironment-Driven Resistance to Immune and Targeted Therapies in Acral Melanoma. ESMO Open 2020, 5, e000707. [Google Scholar] [CrossRef]
  169. Rizzo, D.; Ruggiero, A.; Amato, M.; Maurizi, P.; Riccardi, R. BRAF and MEK inhibitors in pediatric glioma: New therapeutic strategies, new toxicities. Expert Opin. Drug Metab. Toxicol. 2016, 12, 1397–1405. [Google Scholar] [CrossRef]
  170. Bernstein, A.; Mrowczynski, O.D.; Greene, A.; Ryan, S.; Chung, C.; Zacharia, B.E.; Glantz, M. Dual BRAF/MEK inhibitor therapy in BRAF V600E-mutated primary brain tumors: A case series showing dramatic clinical and radiographic responses and a reduction in cutaneous toxicity. J. Neurosurg. 2019, 133, 1704–1709. [Google Scholar] [CrossRef]
  171. Song, H.; Zhong, C.S.; Kieran, M.W.; Chi, S.N.; Wright, K.D.; Huang, J.T. Cutaneous reactions to targeted therapies in children with CNS tumors: A cross-sectional study. Pediatr. Blood Cancer 2019, 66, e27682. [Google Scholar] [CrossRef] [PubMed]
  172. Berzero, G.; Bellu, L.; Baldini, C.; Ducray, F.; Guyon, D.; Eoli, M.; Silvani, A.; Dehais, C.; Idbaih, A.; Younan, N.; et al. Sustained Tumor Control with MAPK Inhibition in BRAF V600-Mutant Adult Glial and Glioneuronal Tumors. Neurology 2021, 97, e673–e683. [Google Scholar] [CrossRef] [PubMed]
  173. Selt, F.; van Tilburg, C.M.; Bison, B.; Sievers, P.; Harting, I.; Ecker, J.; Pajtler, K.W.; Sahm, F.; Bahr, A.; Simon, M.; et al. Response to trametinib treatment in progressive pediatric low-grade glioma patients. J. Neurooncol. 2020, 149, 499–510. [Google Scholar] [CrossRef] [PubMed]
  174. Robison, N.J.; Su, J.A.; Fang, M.J.; Malvar, J.; Menteer, J. Cardiac function in children and young adults treated with MEK inhibitors: A retrospective cohort study. Pediatr. Cardiol. 2022, 43, 1223–1228. [Google Scholar] [CrossRef]
  175. Desai, A.V.; Robinson, G.W.; Gauvain, K.; Basu, E.M.; Macy, M.E.; Maese, L.; Whipple, N.S.; Sabnis, A.J.; Foster, J.H.; Shusterman, S.; et al. Entrectinib in children and young adults with solid or primary CNS tumors harboring NTRK, ROS1, or ALK aberrations (STARTRK-NG). Neuro Oncol. 2022, 24, 1776–1789. [Google Scholar] [CrossRef]
  176. Doz, F.; van Tilburg, C.M.; Geoerger, B.; Højgaard, M.; Øra, I.; Boni, V.; Capra, M.; Chisholm, J.; Chung, H.C.; DuBois, S.G.; et al. Efficacy and safety of larotrectinib in TRK fusion-positive primary central nervous system tumors. Neuro Oncol. 2022, 24, 997–1007. [Google Scholar] [CrossRef]
  177. Desai, A.V.; Bagchi, A.; Armstrong, A.E.; van Tilburg, C.M.; Basu, E.M.; Robinson, G.W.; Wang, H.; Casanova, M.; André, N.; Campbell-Hewson, Q.; et al. Efficacy and safety of entrectinib in children with extracranial solid or central nervous system (CNS) tumours harbouring NTRK or ROS1 fusions. Eur. J. Cancer 2025, 220, 115308. [Google Scholar] [CrossRef]
  178. Felistia, Y.; Wen, P.Y. Molecular profiling and targeted therapies in gliomas. Curr. Neurol. Neurosci. Rep. 2023, 23, 627–636. [Google Scholar] [CrossRef]
  179. Hua, H.; Kong, Q.; Zhang, H.; Wang, J.; Luo, T.; Jiang, Y. Targeting mTOR for cancer therapy. J. Hematol. Oncol. 2019, 12, 71. [Google Scholar] [CrossRef]
  180. Sundstrøm, T.; Prestegarden, L.; Azuaje, F.; Aasen, S.N.; Røsland, G.V.; Varughese, J.K.; Bahador, M.; Bernatz, S.; Braun, Y.; Harter, P.N.; et al. Inhibition of mitochondrial respiration prevents BRAF-mutant melanoma brain metastasis. Acta Neuropathol. Commun. 2019, 7, 55. [Google Scholar] [CrossRef]
  181. Dogan, E.; Kara, H.G.; Kosova, B.; Cetintas, V.B. Targeting Apoptosis to Overcome Chemotherapy Resistance. In Metastasis; Sergi, C.M., Ed.; Exon Publications: Brisbane, Australia, 2022. [Google Scholar] [CrossRef]
  182. Medikonda, R.; Dunn, G.; Rahman, M.; Fecci, P.; Lim, M. A review of glioblastoma immunotherapy. J. Neurooncol. 2021, 151, 41–53. [Google Scholar] [CrossRef] [PubMed]
  183. Sadowski, K.; Jażdżewska, A.; Kozłowski, J.; Zacny, A.; Lorenc, T.; Olejarz, W. Revolutionizing glioblastoma treatment: A comprehensive overview of modern therapeutic approaches. Int. J. Mol. Sci. 2024, 25, 5774. [Google Scholar] [CrossRef] [PubMed]
  184. Berghoff, A.S.; Kiesel, B.; Widhalm, G.; Rajky, O.; Ricken, G.; Wöhrer, A.; Hackl, M.; Birner, P.; Hainfellner, J.A.; Preusser, M. Programmed death ligand 1 expression and tumor-infiltrating lymphocytes in glioblastoma. Neuro Oncol. 2015, 17, 1064–1075. [Google Scholar] [CrossRef] [PubMed]
  185. Jiang, X.; Wang, J.; Deng, X.; Xiong, F.; Ge, J.; Xiang, B.; Wu, X.; Ma, J.; Zhou, M.; Li, X.; et al. Role of the tumor microenvironment in PD-L1/PD-1-mediated tumor immune escape. Mol. Cancer 2019, 18, 1. [Google Scholar] [CrossRef]
  186. Xue, S.; Song, G.; Yu, J. The Prognostic Significance of PD-L1 Expression in Patients with Glioma: A Meta-Analysis. Sci. Rep. 2017, 7, 4231. [Google Scholar] [CrossRef]
  187. Yang, X.; Zeng, Y.; Tan, Q.; Huang, Z.; Jia, J.; Jiang, G. Efficacy of PD-1/PD-L1 inhibitors versus chemotherapy in lung cancer with brain metastases: A systematic review and meta-analysis. J. Immunol. Res. 2022, 2022, 4518898. [Google Scholar] [CrossRef]
  188. Liang, T.; Song, Y.; Gu, L.; Wang, Y.; Ma, W. Insight into the progress in CAR-T cell therapy and combination with other therapies for glioblastoma. Int. J. Gen. Med. 2023, 16, 4121–4141. [Google Scholar] [CrossRef]
  189. Rizwani, F.; Patil, P.; Jain, K. Unlocking glioblastoma: Breakthroughs in molecular mechanisms and next-generation therapies. Med. Oncol. 2025, 42, 28. [Google Scholar] [CrossRef]
  190. Tomaszewski, W.; Sanchez-Perez, L.; Gajewski, T.F.; Sampson, J.H. Brain Tumor Microenvironment and Host State: Implications for Immunotherapy. Clin. Cancer Res. 2019, 25, 4202–4210. [Google Scholar] [CrossRef]
  191. Yang, K.; Wu, Z.; Zhang, H.; Zhang, N.; Wu, W.; Wang, Z.; Dai, Z.; Zhang, X.; Zhang, L.; Peng, Y.; et al. Glioma targeted therapy: Insight into future of molecular approaches. Mol. Cancer 2022, 21, 39. [Google Scholar] [CrossRef]
  192. Mowforth, O.D.; Brannigan, J.; El Khoury, M.; Sarathi, C.I.P.; Bestwick, H.; Bhatti, F.; Mair, R. Personalised therapeutic approaches to glioblastoma: A systematic review. Front. Med. 2023, 10, 116104. [Google Scholar] [CrossRef]
  193. Mojica, C.V.; Gutierrez, K.M.E.; Mason, W.P. Advances in IDH-mutant glioma management: IDH inhibitors, clinical implications of INDIGO trial, and future perspectives. Future Oncol. 2025, 21, 2089–2099. [Google Scholar] [CrossRef]
Figure 1. Schematic overview of the MAPK/ERK signaling cascade. Ligand binding to receptor RTKs, including EGFR, FGFR, PDGFR, ALK, ROS1, Trk, MET, and KIT, induces activation of RAS (KRAS, NRAS, or HRAS) through the recruitment of adaptor proteins such as SHP2, GAB1, GRB2, and SOS1. Active GTP-bound RAS, in turn, recruits and activates RAF kinases through dimerization and phosphorylation, forming active BRAF homodimers or BRAF/CRAF heterodimers. RAF then phosphorylates and activates MEK, which consecutively phosphorylates and activates the terminal kinase ERK. Activated ERK translocates to the nucleus, where it phosphorylates transcription factors and co-activators that regulate gene expression. RTK, receptor tyrosine kinase; EGFR, epidermal growth factor receptor; FGFR, fibroblast growth factor receptor; PDGFR, platelet-derived growth factor receptor; ALK, anaplastic lymphoma kinase; ROS1, ROS proto-oncogene 1; Trk, tropomyosin receptor kinase; MET, mesenchymal–epithelial transition factor; KIT, kit proto-oncogene receptor tyrosine kinase; RAS, rat sarcoma viral oncogene; KRAS, Kirsten rat sarcoma viral oncogene homolog; NRAS, neuroblastoma rat sarcoma viral oncogene homolog; HRAS, Harvey rat sarcoma viral oncogene homolog; SHP2, Src homology region 2 domain-containing phosphatase-2; GAB1, GRB2-associated-binding protein 1; GRB2, growth factor receptor-bound protein 2; SOS1, Son of sevenless homolog 1; RAF, rapidly accelerated fibrosarcoma; BRAF, V-Raf murine sarcoma viral oncogene homolog B; CRAF, proto-oncogene c-Raf; MEK, mitogen-activated protein kinase; ERK, extracellular signal-regulated kinase; GTP, guanosine triphosphate; GDP, guanosine diphosphate. Created in BioRender. Adamopoulos, C. (2025) https://BioRender.com/haba8wa (Assessed on 7 September 2025).
Figure 1. Schematic overview of the MAPK/ERK signaling cascade. Ligand binding to receptor RTKs, including EGFR, FGFR, PDGFR, ALK, ROS1, Trk, MET, and KIT, induces activation of RAS (KRAS, NRAS, or HRAS) through the recruitment of adaptor proteins such as SHP2, GAB1, GRB2, and SOS1. Active GTP-bound RAS, in turn, recruits and activates RAF kinases through dimerization and phosphorylation, forming active BRAF homodimers or BRAF/CRAF heterodimers. RAF then phosphorylates and activates MEK, which consecutively phosphorylates and activates the terminal kinase ERK. Activated ERK translocates to the nucleus, where it phosphorylates transcription factors and co-activators that regulate gene expression. RTK, receptor tyrosine kinase; EGFR, epidermal growth factor receptor; FGFR, fibroblast growth factor receptor; PDGFR, platelet-derived growth factor receptor; ALK, anaplastic lymphoma kinase; ROS1, ROS proto-oncogene 1; Trk, tropomyosin receptor kinase; MET, mesenchymal–epithelial transition factor; KIT, kit proto-oncogene receptor tyrosine kinase; RAS, rat sarcoma viral oncogene; KRAS, Kirsten rat sarcoma viral oncogene homolog; NRAS, neuroblastoma rat sarcoma viral oncogene homolog; HRAS, Harvey rat sarcoma viral oncogene homolog; SHP2, Src homology region 2 domain-containing phosphatase-2; GAB1, GRB2-associated-binding protein 1; GRB2, growth factor receptor-bound protein 2; SOS1, Son of sevenless homolog 1; RAF, rapidly accelerated fibrosarcoma; BRAF, V-Raf murine sarcoma viral oncogene homolog B; CRAF, proto-oncogene c-Raf; MEK, mitogen-activated protein kinase; ERK, extracellular signal-regulated kinase; GTP, guanosine triphosphate; GDP, guanosine diphosphate. Created in BioRender. Adamopoulos, C. (2025) https://BioRender.com/haba8wa (Assessed on 7 September 2025).
Cancers 18 00156 g001
Figure 2. Representative alterations in the MAPK signaling cascade in central nervous system (CNS) tumors. Distinct genetic mechanisms drive aberrant signaling at multiple levels: (i) RTK amplifications, mutations, and fusions (e.g., EGFR, PDGFRA, FGFR, MET, ALK, ROS1, NTRK); (ii) activating RAS mutations in KRAS or NRAS, that impair GTP hydrolysis; (iii) oncogenic RAF alterations, most notably in BRAF, which include fusions such as BRAF-KIAA1549 leading to constitutive activation, hotspot substitutions like BRAF V600E conferring to the oncoprotein strong monomeric kinase activity, and class III mutations (e.g., BRAF G464E, D594G) that produce kinase-impaired proteins dependent on upstream RAS or RTK signaling; and (iv) alterations in negative or positive regulators of MAPK signaling, such as NF1 LOF mutations and activating SHP2 mutations. Collectively, these lesions converge on hyperactivation of the RAF–MEK–ERK axis, driving proliferation, survival, and therapeutic resistance in CNS tumors. Solid red arrows depict activation and dashed arrow depicts deactivation. EGFR, epidermal growth factor receptor; PDGFRA, platelet-derived growth factor receptor α; FGFR, fibroblast growth factor receptor; MET, mesenchymal–epithelial transition factor; ALK, anaplastic lymphoma kinase; ROS1, ROS proto-oncogene 1; NTRK, neurotrophic tyrosine receptor kinase; RAS, rat sarcoma viral oncogene; KRAS, Kirsten rat sarcoma viral oncogene homolog; NRAS, neuroblastoma rat sarcoma viral oncogene homolog, Harvey rat sarcoma viral oncogene homolog; RAF, rapidly accelerated fibrosarcoma; BRAF, V-Raf murine sarcoma viral oncogene homolog B; CRAF, proto-oncogene c-Raf; NF1, neurofibromin; SHP2, Src homology region 2 domain-containing phosphatase-2; MEK, mitogen-activated protein kinase; ERK, extracellular signal-regulated kinase; LOF loss-of-function; GTP, guanosine triphosphate; GDP, guanosine diphosphate. Created in BioRender. Adamopoulos, C. (2025) https://BioRender.com/krxcp45 (Assessed on 7 September 2025).
Figure 2. Representative alterations in the MAPK signaling cascade in central nervous system (CNS) tumors. Distinct genetic mechanisms drive aberrant signaling at multiple levels: (i) RTK amplifications, mutations, and fusions (e.g., EGFR, PDGFRA, FGFR, MET, ALK, ROS1, NTRK); (ii) activating RAS mutations in KRAS or NRAS, that impair GTP hydrolysis; (iii) oncogenic RAF alterations, most notably in BRAF, which include fusions such as BRAF-KIAA1549 leading to constitutive activation, hotspot substitutions like BRAF V600E conferring to the oncoprotein strong monomeric kinase activity, and class III mutations (e.g., BRAF G464E, D594G) that produce kinase-impaired proteins dependent on upstream RAS or RTK signaling; and (iv) alterations in negative or positive regulators of MAPK signaling, such as NF1 LOF mutations and activating SHP2 mutations. Collectively, these lesions converge on hyperactivation of the RAF–MEK–ERK axis, driving proliferation, survival, and therapeutic resistance in CNS tumors. Solid red arrows depict activation and dashed arrow depicts deactivation. EGFR, epidermal growth factor receptor; PDGFRA, platelet-derived growth factor receptor α; FGFR, fibroblast growth factor receptor; MET, mesenchymal–epithelial transition factor; ALK, anaplastic lymphoma kinase; ROS1, ROS proto-oncogene 1; NTRK, neurotrophic tyrosine receptor kinase; RAS, rat sarcoma viral oncogene; KRAS, Kirsten rat sarcoma viral oncogene homolog; NRAS, neuroblastoma rat sarcoma viral oncogene homolog, Harvey rat sarcoma viral oncogene homolog; RAF, rapidly accelerated fibrosarcoma; BRAF, V-Raf murine sarcoma viral oncogene homolog B; CRAF, proto-oncogene c-Raf; NF1, neurofibromin; SHP2, Src homology region 2 domain-containing phosphatase-2; MEK, mitogen-activated protein kinase; ERK, extracellular signal-regulated kinase; LOF loss-of-function; GTP, guanosine triphosphate; GDP, guanosine diphosphate. Created in BioRender. Adamopoulos, C. (2025) https://BioRender.com/krxcp45 (Assessed on 7 September 2025).
Cancers 18 00156 g002
Table 1. MAPK pathway inhibitors in central nervous system (CNS) tumors: molecular targets, tumor indications, and representative clinical outcomes. LGG, low-grade glioma; HGG, high-grade glioma; PXA, pleomorphic xanthoastrocytoma; GBM, glioblastoma multiforme; NF1, neurofibromin; OP, optic pathway; NF1-MPNST, NF1-associated malignant peripheral nerve sheath tumor; PN, plexiform neurofibroma; ORR, overall response rate; PFS, progression-free survival; FDA, Food and Drug Administration; BBB, blood–brain barrier; CNS, central nervous system; TMZ, temozolomide.
Table 1. MAPK pathway inhibitors in central nervous system (CNS) tumors: molecular targets, tumor indications, and representative clinical outcomes. LGG, low-grade glioma; HGG, high-grade glioma; PXA, pleomorphic xanthoastrocytoma; GBM, glioblastoma multiforme; NF1, neurofibromin; OP, optic pathway; NF1-MPNST, NF1-associated malignant peripheral nerve sheath tumor; PN, plexiform neurofibroma; ORR, overall response rate; PFS, progression-free survival; FDA, Food and Drug Administration; BBB, blood–brain barrier; CNS, central nervous system; TMZ, temozolomide.
Drug/StrategyMolecular Target(s)CNS Tumor Type(s)Key Clinical FindingsSelected Trials
Dabrafenib + TrametinibBRAF V600E + MEK1/2Pediatric LGG,
HGG, PXA, GBM
ORR > 70% in BRAF V600E pLGG; significant PFS improvement;
FDA-approved for pLGG (2023)
NCT07110246,
NCT03919071
VemurafenibBRAF V600EGlioma, PXAPartial responses;
limited durability due to resistance;
modest BBB penetration
NCT01748149
Encorafenib
(+ MEK inhibitors)
BRAF V600EGlioma (investigational)Improved pharmacodynamics vs. vemurafenib; CNS efficacy under studyNCT03973918
SelumetinibMEK1/2NF1-associated pLGG,
OP glioma
Tumor shrinkage and visual improvement; durable disease control;
FDA-approved for NF1 tumors
NCT01089101,
NCT03871257
TrametinibMEK1/2pLGG, NF1 tumors,
PXA
Clinical benefit in pLGG and PNs;
enhanced efficacy with dabrafenib
NCT03363217
MirdametinibMEK1/2NF1 tumors, pLGGRecently FDA-approved for
NF1-associated PN; promising CNS activity
NCT04923126
TovorafenibRAFRelapsed pLGG with BRAF alterationsHigh response rate;
effective in BRAF-fusion tumors;
FDA-approved 2024
FIREFLY-1/NCT04775485
NST-628RAF-MEK
molecular glue
RAS/RAF-mutant
gliomas
Potent, brain-penetrant MAPK suppression; preclinical efficacyPreclinical
UlixertinibERK1/2Advanced glioma (investigational)Activity in BRAF/MEK-resistant tumors; BBB penetrationNCT01781429
SHP2 inhibitors (TNO155, RMC-4630)SHP2GBM,
NF1-MPNST
Suppress upstream RAS activation;
synergy with TMZ *
NCT03114319
* Chemotherapy drug (alkylating agent).
Table 2. Current and ongoing clinical trial assessing certain MAPK inhibitors, as single agents or in combinations, for central nervous system (CNS) tumors. LGG, low-grade glioma; HGG, high-grade glioma; HQ, hydroxychloroquine; NF1, neurofibromin; PN, plexiform neurofibroma; OP/HG, optic pathway/hypothalamic glioma; PA, pilocytic astrocytoma.
Table 2. Current and ongoing clinical trial assessing certain MAPK inhibitors, as single agents or in combinations, for central nervous system (CNS) tumors. LGG, low-grade glioma; HGG, high-grade glioma; HQ, hydroxychloroquine; NF1, neurofibromin; PN, plexiform neurofibroma; OP/HG, optic pathway/hypothalamic glioma; PA, pilocytic astrocytoma.
Agent (Target)Tumor TypeAgeStudy Name/
Clinical Trial ID
Stage
Dabrafenib * and trametinib ±BRAF V600-mutant pLGG12 months–25 years oldNCT07110246Phase II
Dabrafenib * and trametinibSeveral CNS tumors1–99 years oldNCT03975829Phase IV
Dabrafenib, trametinib ± and nivolumab BRAF-altered pediatric glioma1–26 years oldNCT06712875Phase I/II
Dabrafenib * and trametinib ±HGG (among other cancer types)18–100 years oldNCT03340506Phase IV
Dabrafenib * and trametinib ±HGG3–25 years oldNCT03919071Phase II
Dabrafenib, trametinib ± and HQ LGG or HGG with BRAF aberration
LGG with NF1
1–30 years oldNCT04201457Phase I/II
Mirdametinib ±LGG2–24 years oldNCT04923126Phase I/II
Mirdametinib ±LGG, activation of MAPK2–24 years oldNCT06666348Phase I/II
Selumetinib ±Recurrent/refractory LGG, OP/HG glioma, NF1, PA3–21 years oldNCT01089101Phase I/II
Selumetinib ±Progressive LGG2–25 years oldNCT04576117Phase III
Selumetinib ±NF1, LGG2–21 years oldNCT03871257Phase III
Selumetinib ±LGG2–21 years oldNCT04166409Phase III
Trametinib ±LGG1 month–25 years oldNCT05180825Phase II
Trametinib ±LGG, NF1, PN, activation of the MAPK/ERK pathway1–25 years oldNCT03363217Phase II
Trametinib ± and everolimus LGG, HGG1–25 years oldNCT04485559Phase I
Tovorafenib *Relapsed/refractory LGG with BRAF alterations6 months–25 years oldFIREFLY-1/NCT04775485Phase II
* RAF inhibitor, ± MEK inhibitor, anti-programmed cell death-1 (PD1) monoclonal antibody, autophagy inhibitor, mammalian target of rapamycin (mTOR) inhibitor.
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

Vrachas, D.; Kosma, E.; Giannopoulou, A.-I.; Margoni, A.; Gargalionis, A.N.; El-Habr, E.A.; Piperi, C.; Adamopoulos, C. Targeting the MAPK Pathway in Brain Tumors: Mechanisms and Therapeutic Opportunities. Cancers 2026, 18, 156. https://doi.org/10.3390/cancers18010156

AMA Style

Vrachas D, Kosma E, Giannopoulou A-I, Margoni A, Gargalionis AN, El-Habr EA, Piperi C, Adamopoulos C. Targeting the MAPK Pathway in Brain Tumors: Mechanisms and Therapeutic Opportunities. Cancers. 2026; 18(1):156. https://doi.org/10.3390/cancers18010156

Chicago/Turabian Style

Vrachas, Dimitrios, Elisavet Kosma, Angeliki-Ioanna Giannopoulou, Angeliki Margoni, Antonios N. Gargalionis, Elias A. El-Habr, Christina Piperi, and Christos Adamopoulos. 2026. "Targeting the MAPK Pathway in Brain Tumors: Mechanisms and Therapeutic Opportunities" Cancers 18, no. 1: 156. https://doi.org/10.3390/cancers18010156

APA Style

Vrachas, D., Kosma, E., Giannopoulou, A.-I., Margoni, A., Gargalionis, A. N., El-Habr, E. A., Piperi, C., & Adamopoulos, C. (2026). Targeting the MAPK Pathway in Brain Tumors: Mechanisms and Therapeutic Opportunities. Cancers, 18(1), 156. https://doi.org/10.3390/cancers18010156

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

Article Metrics

Article metric data becomes available approximately 24 hours after publication online.
Back to TopTop