1. Introduction
Glioblastoma multiforme (GBM) is the most prevalent and deadly primary brain tumor in adults. It is classified into three molecular subtypes: proneural, classical (CL), and mesenchymal (MES) [
1]. The MES subtype is the most aggressive, while CL has an intermediate prognosis. Standard treatment includes surgical resection, radiotherapy, and temozolomide (TMZ) [
2]. Since 2017, the addition of Tumor Treating Fields (TTFields), which deliver alternating electrical fields to the skull, has improved progression-free and overall survival [
3,
4], independent of MGMT methylation status [
5]. Still, median survival remains about 21 months, largely due to tumor heterogeneity and resistance to radiotherapy [
4]. A major contributor to treatment failure is the presence of GBM stem cells (GSCs), self-renewing, highly tumorigenic cells with strong radioresistance, linked to enhanced DNA damage repair and plasticity [
6,
7,
8]. Our prior studies have highlighted the critical role of FGFR1 in mediating radioresistance in GBM. In a clinical trial combining tipifarnib with radiotherapy, we showed that high FGFR1 expression correlated with worse overall and progression-free survival [
9]. In vitro studies have demonstrated that targeting FGFR1 reduces GBM radioresistance. In differentiated GBM cells, combining FGFR1 inhibition with radiation has been shown to introduce centrosome overduplication, mitotic catastrophe, and a decrease in HIF-1α levels, thereby enhancing radiosensitivity [
10]. While HIF-1α is a key mediator of hypoxic adaptation, it also independently promotes radioresistance through the transcriptional regulation of survival pathways [
11,
12]. In vivo, FGFR1 inhibition delays the growth of irradiated tumor xenografts, a process linked to reduced HIF-1α levels, without affecting blood vessel integrity [
10]. In GSCs, FGFR1 is also crucial in radioresistance, with its inhibition significantly enhancing radiosensitivity. This radiosensitization is associated with downregulated expression of FOXM1, a gene involved in both chemoresistance and radioresistance. The FGFR1/FOXM1 pathway is also implicated in the mesenchymal transition, as silencing
FGFR1 or
FOXM1 in GSCs downregulates genes like
GLI2,
TWIST1, and
ZEB1, which are involved in this process. This inhibition also significantly reduces GSC migration, indicating that the FGFR1/FOXM1 pathway, which governs GSC radioresistance, is also a key driver of mesenchymal transition. Taken together, these results, along with previously reported data in differentiated cells, clearly established that the FGFR1-FOXM1-dependent pathway in GSCs plays a pivotal role in GBM treatment resistance [
13]. In clinical samples comparing primary and recurrent GBM, varying expression levels of targets such as ALK, PDGFRβ, PDGFRA, MET, FGFR1, FGFR2, and FGFR3 have been observed, indicating significant heterogeneity throughout the disease’s progression. Notably, FGFR1 is the only target that maintains consistent expression in tissue samples from GBM patients who have undergone radiotherapy and TMZ treatment [
14]. Recent cluster interaction and spatial transcriptomic analyses have uncovered a critical role for stem-like glioblastoma cells in shaping the tumor microenvironment. These cells secrete specific chemokines that recruit early myeloid-derived suppressor cells (E-MDSCs), which in turn release growth factors that can fuel tumor progression. Among the signaling pathways implicated, the FGF11–FGFR1 axis has emerged as a potential key driver of glioblastoma growth and resistance [
15]. Furthermore, an interaction between FGFR1 and integrin α6 has been identified, demonstrating that α6-integrin contributes to GSC proliferation and stemness by regulating FGFR1 and FOXM1 expression via the ZEB1/YAP1 transcriptional complex [
16]. This α6-integrin–FGFR1 crosstalk underscores the importance of the FGFR1 axis in sustaining glioblastoma stem cell properties and supports its relevance as a therapeutic target in GBM. Pemigatinib, also known as INCB054828, distinguishes itself from earlier FGFR inhibitors through its high potency and selectivity for FGFR1–3, with reported IC
50 values of approximately 0.4 nM for FGFR1, 0.5 nM for FGFR2, and 1.0 nM for FGFR3, while showing markedly weaker activity against FGFR4 (IC
50 ≈ 30 nM). In a broad kinase selectivity screen including more than 100 kinases, pemigatinib exhibited over 100-fold selectivity for FGFR1–3 relative to the vast majority of other targets, with only a few non-FGFR kinases with IC
50 values below 1000 nM, including VEGFR-2 (KDR; IC
50 ≈ 190 nM) and c-Kit (IC
50 ≈ 270 nM) [
17]. These values highlight its ultra-low nanomolar biochemical potency and strong target specificity. In our study, however, higher concentrations were required to elicit functional effects in glioblastoma stem cells, likely reflecting the intrinsic resistance mechanisms and reduced drug permeability characteristic of GSCs compared to conventional cancer cell lines. It works by blocking FGFR autophosphorylation, disrupting signal transduction and the subsequent activation of downstream cellular signaling pathways. What sets pemigatinib apart from earlier FGFR inhibitors is its heightened selectivity for members of the FGFR family.
Its effectiveness and pharmacological properties have allowed it to demonstrate antitumor activity at lower doses in preclinical settings. Pemigatinib is approved for use as a standalone treatment in adult patients with locally advanced or metastatic cholangiocarcinoma that exhibits FGFR2 fusion or rearrangement, particularly in cases where patients have relapsed or are unresponsive after at least one prior systemic therapy. However, its clinical efficacy can be compromised by the development of acquired resistance, primarily through secondary mutations in the kinase domain of FGFR2 [
18]. Numerous clinical trials are currently underway exploring the use of pemigatinib in various cancers, including bladder cancer, nonmuscle invasive bladder cancer, recurrent urothelial carcinoma (NCT03914794), pancreatic cancer (NCT05216120), urothelial cancer (NCT04294277), gastrointestinal cancer (NCT05651672), non-small cell lung cancer (NCT05210946), breast cancer (NCT05560334), and, most recently, recurrent GBM and other primary central nervous system tumors with an activating FGFR1-3 mutation or fusion/rearrangement (NCT05267106).
Based on all the previous preclinical and clinical data, the main objective of our study is to evaluate the radiosensitizing potential of pemigatinib, which specifically targets FGFR1, for differentiated glioblastoma cells and GSCs, when used in conjunction with radiotherapy both in vitro and in vivo.
2. Materials and Methods
2.1. Cell Culture
This study utilized two types of GBM cells: differentiated cells and stem cells. Differentiated human GBM cells, U87MG (RRID:CVCL_0022) (MGMT methylated) and LN-18 (RRID:CVCL_0392) (MGMT unmethylated) from the American Type Culture Collection (Rockville, MD, USA), were cultured according to methods outlined in a previous publication [
10].
Primary stem cell lines, named GC1 and GC2, were derived from GBM biopsy specimens obtained from the Neurosurgery Department of Toulouse University Hospital as part of the clinical study STEMRI, led by PI Pr E. Cohen-Jonathan Moyal. This study received approval from the Human Research Ethics Committee (Ethics Code 12TETE01, ID-RCB No. 2012-A00585-38, Approval Date: 7 May 2012), and the results have recently been published [
19]. Written informed consent was secured from all the participating patients. The World Health Organization classified all the tumors involved as GBM. The characteristics of GC1 and GC2 as stem cells have previously been detailed by our group [
13]. The cells were routinely tested for mycoplasma using MycoAlert (Lonza, Basel, Switzerland). GC1 and GC2 were previously characterized as glioblastoma stem cell lines [
13] and cryopreserved at early passages (P2–P3). To preserve their stem-like characteristics, all the experiments in this study were conducted using cells between passages 3 and 10 after initial derivation from patient tumor tissue.
2.2. Reagents
Pemigatinib (Pemazyre®, INCB054828) was obtained from Incyte and dissolved in DMSO (in vitro) or in 5% DMAC, 50 mM citrate buffer, and 0.5% methyl cellulose (in vivo). MG132 was diluted in ethanol and used at 20 µM. All the reagents, except pemigatinib, were purchased from Sigma-Aldrich (Sigma, Saint-Quentin Fallavier, France).
2.3. Cell Viability
GC1 and GC2 cells (150,000/well, 24-well plates) were treated with pemigatinib for 24–48 h at 125, 250, and 500 nM. The cell number was determined post-treatment, after the dissociation of neurospheres, using a Bio-Rad counter (Biorad, Hercules, CA, USA). U87 and LN18 cells (10,000/well, 96-well plates) were treated with pemigatinib at 1, 2, and 3 μM, and viability was assessed via the MTT assay (Abcam, Cambridge, UK). For combination experiments with temozolomide (100 μM) in differentiated GBM cells, 1 μM pemigatinib was chosen as the lowest concentration that produced a detectable biological effect in these cells, enabling the assessment of potential interaction with TMZ while minimizing off-target effects.
2.4. siRNA Transfection
Neurospheres were dissociated and plated at 500,000 cells in 5 mL of culture medium 24 h before transfection. Transfection was carried out using Lipofectamine RNAimax (Invitrogen, Waltham, MA, USA) according to the manufacturer’s instructions. siRNAs targeting FGFR1 (SI03094637), S100A4(1) (SI00709667), S100A4(8) (SI04227916), and a scramble control siRNA (siscr) were acquired from Qiagen (Qiagen, Venlo, The Netherlands).
2.5. Quantitative Real-Time PCR
For the quantitative analysis of gene expression for the human FGFR1, FOXM1, S100A4, and glyceraldehyde-3-phosphate dehydrogenase (GAPDH) genes, total RNA was extracted using the RNeasy RNA isolation kit (Qiagen), reverse-transcribed using the PrimeScript RT Reagent Kit (TAKARA, Kusatsu, Japan), and amplified using SsoFast EvaGreen Supermix (Bio-Rad, Marnes-la-Coquette, France). Gene expression was normalized to GAPDH using StepOne+ (Applied Biosystems, Waltham, MA, USA).
2.6. Western Blot
Proteins were extracted in 50 mM Tris-HCl (pH 7.5), 0.1% Triton X-100, 5 mM EDTA, and protease inhibitors. SDS-PAGE was performed using Mini-PROTEAN TGX Stain-Free Gels (Bio-Rad) followed by transfer to nitrocellulose membranes (Trans-Blot Turbo Transfer Packs, Bio-Rad) using the Trans-Blot Turbo Transfer system (Bio-Rad). The membranes were incubated with the following primary antibodies: anti-FGFR1 (D8E4) XP rabbit (1:1000 dilution; Cat# 9740, Cell Signaling Technology, Danvers, MA, USA), anti-FRS2 (EPR14724) (1:1000 dilution; abcam, Cambridge, UK), anti-actin clone C4 (1:20,000 dilution; Cat# MAB1501, Millipore, Molsheim, France), and anti-phospho-FRS2-alpha (Tyr196) (1:1000 dilution; Cell Signaling Technology Cat# 3864). Signals were visualized with ECL kits (ECL RevelBlot Plus and ECL RevelBlot Intense, Ozyme, Saint-Cyr-l’école, France) and imaged with Chemi-Doc (Bio-Rad).
2.7. Flow Cytometry
GSCs treated with 500 nM pemigatinib for 48 h were fixed and permeabilized (Cytofix/Cytoperm, BD Biosciences, Franklin Lakes, NJ, USA). The cells were then washed and incubated for 30 min at 4 °C in PBS supplemented with 10% FBS to block nonspecific binding. Subsequently, the cells were incubated for 1 h at 4 °C in the dark with an S100A4-PE conjugated primary antibody (NBP2-54580APC, NOVUS Biologicals, Cambridge, UK) or a mouse IgG1 kappa isotype control (17-4714-42, Invitrogen).
Data were acquired using a MACSQuant10 (Miltenyi Biotec, Bergisch Gladbach, Germany) and analyzed in FlowJo
TM v10.9 Software (BD Life Sciences, Franklin Lakes, NJ, USA). The S100A4 levels were quantified as the specific fluorescence index (SFI) = (Geomean antibody − Geomean IgG control)/Geomean IgG control [
20].
2.8. Clonogenic Assay
U87 and LN18 cells (500/well, 6-well plates) were treated with irradiation, TMZ, and/or pemigatinib. After one week, colonies were stained with crystal violet. The survival fraction was calculated using the following formula: Survival fraction = (colonies counted/(cells seeded × plating efficiency)) × 100. Here, plating efficiency (PE) is the seeding efficiency, defined as the ratio of the number of colonies formed to the number of cells initially seeded.
Primary neurospheres treated with pemigatinib or siRNA targeting S100A4 or vehicle were seeded in 96-well plates at a concentration of 500 cells/well, with 12 wells per condition. The following day, the cells were irradiated or not irradiated with X-rays ranging from 2 to 6 Gy (SmART+ irradiator, Precision X-ray Inc., Madison, CT, USA). One week post irradiation, the number of neurospheres composed of more than 20 cells was assessed. The surviving fraction was determined according to the PE concentration under nonirradiated conditions. PE is defined as follows: PE = number of spheres/number of seeded cells × 100. The sphere size was analyzed using Fiji (ImageJ version 1.53q, Java 1.8.0_172, 64-bit).
2.9. RNA Sequencing
For transcriptomic analysis, the GC1 and GC2 glioblastoma stem cell lines were seeded at 2 × 106 cells in 5 mL of medium in 25 cm3 flasks and treated with either 250 nM pemigatinib or DMSO control for 48 h (three independent biological replicates per condition per cell line; n = 6 total per condition). RNA was extracted using the RNeasy kit (Qiagen) and quality verified (RQN > 8) prior to library preparation with the Illumina Stranded Total RNA Prep protocol, including rRNA depletion, strand-specific library construction, and paired-end sequencing. The library quality and concentration were assessed using the Qubit™ dsDNA BR Assay (Thermo Fisher Scientific, Waltham, MA, USA), High Sensitivity NGS Fragment Analysis Kit (Agilent Technologies, Santa Clara, CA, USA), and KAPA Library Quantification Kit for Illumina® (Roche, Basel, Switzerland). Sequencing was performed on an Illumina NextSeq 550 (San Diego, CA, USA) with a High Output flow cell, and the raw reads were demultiplexed with bcl2fastq v2.20.0.422. The transcriptomic results represent an integrated differential expression analysis across both models, averaged to highlight changes conserved between GSC lines. The RNA-seq data have been deposited in the NCBI SRA under accession PRJNA1320628.
2.10. Fusion and Mutation Determination
Reference and alignment. Reads were aligned to GRCh38 using STAR v2.7.x in two-pass mode with default gene model guidance (GTF: Ensembl release v98). Soft clipping and chimeric junction output were enabled for fusion discovery.
Fusion calling. Gene fusions were called with Arriba v2.x (using the recommended blacklist and database bundles) [
21] and STAR-Fusion v1.x (with the CTAT resource library built on GRCh38; Ensembl v98) [
22]. The minimal evidence requirements followed the tool defaults (e.g., Arriba: ≥1 junction read; STAR-Fusion: junction + spanning support). The results were filtered to remove read-through events, paralog artifacts, mitochondrial fusions, and panel-of-normal hits. High-confidence fusions were defined as (i) detected by both callers or (ii) single-caller calls with strong supporting evidence and manual confirmation in IGV. Under these criteria, no high-confidence FGFR1/2/3 fusions were detected in GC1 or GC2.
Variant calling from RNA-seq. Putative variants in FGFR1–3 were identified from RNA-seq using GATK HaplotypeCaller (gatk-4.2.0.0, GRCh38v98,
https://gatk.broadinstitute.org/hc/en-us/articles/360037225632-HaplotypeCaller (accessed on 1 September 2025)) following RNA-seq best practices: SplitNCigarReads, base quality recalibration, and variant calling restricted to the coding regions of canonical transcripts (Ensembl v98). Variants were filtered with hard thresholds (e.g., low depth/quality or strand bias) and annotated using VEP (SIFT/PolyPhen) with cross-reference to ClinVar, COSMIC, and CIViC. Variants were categorized as pathogenic/likely pathogenic, benign/likely benign, or VUS. Consistent with the results, only VUS (and known SNPs) were found in FGFR1–3; no activating kinase-domain mutations were identified. Selected loci were reviewed visually in IGV to confirm read support and rule out mapping artifacts. This analysis was conducted using Alamut Visual software (version 2.11-0; Interactive Biosoftware, Rouen, France).
2.11. Bioinformatic Analysis
QC and pre-processing. The FASTQ quality was assessed with FastQC and summarized with MultiQC (per-base quality, adaptor content, GC distribution). The post-alignment QC included the mapping rate, multimapping, rRNA content, gene body coverage, strandedness, and duplication estimates (Picard).
Quantification and DE. Gene-level counts were generated with featureCounts (union exon model; stranded = reverse for the Illumina stranded protocol). Differential expression was assessed in R/DESeq2 with the design~cell_line + treatment to estimate the treatment effect while accounting for GC1/GC2 differences. Multiple testing was controlled by the Benjamini–Hochberg procedure; unless noted, genes with adjusted p < 0.05 and |log2FC| > 1 were considered significant. The log2FC values represent line-averaged effects when regulation was concordant across GC1 and GC2.
Pathway and enrichment analyses. The mRNA expression levels of FGFR receptors and S100A4 in GBM tissue compared to normal tissue were evaluated using Gene Expression Profiling Interactive Analysis (GEPIA,
http://gepia.cancer-pku.cn/ (accessed on 1 September 2025)), a web-based tool that provides fast and customizable analyses based on TCGA and GTEx data. For this analysis, the log
2 fold change (log
2FC) cutoff was set at 1, and the
p-value cutoff was set at 0.01. The gene expression values are presented as log
2(TPM + 1), where TPM denotes transcripts per million.
A volcano plot was generated to visualize the differential gene expression in GSCs treated with or without pemigatinib, using RNA-seq data and the Srplot tool (Scientific and Research plot tool,
http://www.bioinformatics.com.cn/SRplot (accessed on 1 September 2025)). This free online platform was also used to visualize the principal pathways affected in GSCs treated with pemigatinib. Impacted and downregulated genes were identified through Gene Ontology (GO) analysis using the web-based portal Metascape (
https://metascape.org (accessed on 1 September 2025)).
2.12. In Vivo Experiments
Six-to-eight-week-old female nude mice were used in accordance with a protocol (APAFIS# 24969-2020040210408334 v4; approval date: 11 June 2020) established by the Institutional Animal Care and Ethic Committee (UMS006 CEEA-122; President: Nicolas Cenac). The NMRI nu/nu mice (Janvier Labs, Genest Saint Isle, France) were orthotopically implanted with 250,000 GC1 cells that had been previously transduced with a vector containing the luciferase gene, GFP gene, and geneticin resistance gene (#LVP403, Amsbio, Abingdon, UK), under approval #2621 for the contained use of genetically modified organisms for research purposes. Tumor progression was monitored weekly using IVIS bioluminescence imaging. Before the start of treatment (day 23), tumor monitoring using IVIS imaging was performed to identify the mice in which tumor cell implantation successfully led to tumor formation. Each group initially included 20 mice. Mice that did not develop tumors were excluded from the study based on the IVIS imaging results. Specifically, 7 mice were excluded from both the control group and the pemigatinib-treated group, 3 mice from the irradiated group, and 6 mice from the group receiving both pemigatinib and irradiation. To ensure comparability between experimental groups, only mice with equivalent tumor sizes were included in each group. Beginning 23 days after xenografting, the mice were administered pemigatinib (0.5 mg/kg) by gavage 5 days a week for the duration of the experiment, until 36 days post-xenografting. The mice received localized irradiation at a dose of 5 Gy (SmART+ irradiator, Precision X-ray Inc., Madison, CT, USA). They were sacrificed as soon as clinical symptoms appeared. To ensure consistency, all animals were treated and measured in the same order by the same experimenters. In addition, different experimenters were responsible for gavage administration and tumor monitoring to reduce the potential bias associated with individual handling or measurement.
2.13. Statistical Analysis
Data are presented as mean ± SD. Comparisons used Student’s t-test or one-way ANOVA. Kaplan–Meier survival was analyzed using the log-rank test (GraphPad Prism version 10.1.2, GraphPad Software, Boston, MA, USA).
4. Discussion
Despite the significant improvement in patient survival achieved with the introduction of TTFields therapy combined with temozolomide and radiotherapy as a standard treatment [
25], GBM remains aggressive and prone to recurrence, emphasizing the need for new therapeutic strategies. In this study, we evaluated pemigatinib, a selective FGFR inhibitor, for its radiosensitizing and antiproliferative effects in GBM. Based on GEPIA and prior data, FGFR1 emerged as the most overexpressed FGFR in GBM and was consistently detected in all the models tested. This led us to focus on the consequences of FGFR1 inhibition. Our previous research demonstrated that FGFR1 inhibition in both differentiated cells and GSCs induced a radiosensitizing effect [
10,
13]. Additionally, we demonstrated that pemigatinib effectively inhibited FGFR1 signaling by reducing FRS2 phosphorylation and promoting FGFR1 degradation via the proteasome pathway. While short-term viability assays in differentiated GBM cells showed minimal effects, likely due to cell adhesion-mediated resistance via focal adhesion kinase [
26,
27], clonogenic assays revealed a pronounced reduction in colony formation particularly in the MGMT-unmethylated LN18 line. In this TMZ-resistant context, pemigatinib alone reduced colony formation by 50%, and its combination with TMZ and radiation further enhanced radiosensitivity. Although we did not formally assess drug synergy, using 1 μM pemigatinib, the lowest active concentration in differentiated cells, enabled us to test whether modest FGFR1 inhibition could influence the TMZ response, particularly in MGMT-unmethylated models, suggesting therapeutic potential in patients with limited response to alkylating agents. Importantly, this therapeutic effect occurred despite the absence of FGFR gene fusions or known activating mutations in the LN18, suggesting that functional FGFR1 pathway activity, rather than mutational status alone, may be sufficient to confer sensitivity to FGFR inhibition. This highlights a potential shift in biomarker strategies, moving from genomic profiling toward assessing pathway dependency, broadening the scope of patients who may benefit from FGFR-targeted therapy. In GSCs, pemigatinib exhibited potent antitumor activity, significantly reducing neurosphere number and size in a dose-dependent manner and inducing significant cell death at nanomolar concentrations. While differentiated GC1 and GC2 cells could theoretically serve as more direct comparators, our preliminary experiments showed that FGFR1 expression was not significantly altered by pemigatinib at concentrations effective in the stem cell state (125–500 nM). As shown in
Supplementary Figure S2, FGFR1 protein levels remained unchanged in differentiated GC1 and GC2 cells, suggesting that FGFR1 pathway activity is reduced or less accessible in the differentiated state. Radiosensitization by pemigatinib was found to be context-dependent: GC1 cells, which were highly sensitive to pemigatinib monotherapy, showed limited additive benefit when pemigatinib was combined with irradiation. In contrast, GC2 cells, which had moderate single-agent sensitivity, displayed synergistic radiosensitization, particularly at higher radiation doses, as reflected in the reduced sphere size and survival fraction. The pronounced antiproliferative effect of pemigatinib appears to be mediated through the disruption of cell cycle regulation, as supported by our transcriptomic analysis. Other studies have also suggested that pemigatinib-induced G1 arrest is a common cytostatic mechanism across various FGFR expression patterns and tumor types [
28], targeting pathways such as PI3K/AKT and RAF/MEK/ERK which are involved in cell growth, migration, proliferation, and metabolism [
29,
30].
Importantly, both GC1 and GC2 were found to be MGMT-unmethylated, a feature associated with poor response to standard therapy. The responsiveness of these cells to pemigatinib reinforces its potential as an alternative or complementary strategy in TMZ-refractory GBM.
Transcriptomic profiling further revealed that pemigatinib treatment downregulated genes involved in the mitotic cell cycle, DNA repair, and radiation response, providing mechanistic support for its radiosensitizing effects. Among the most significantly downregulated genes were
FGFR1,
FOXM1, and
S100A4, each associated with GSC proliferation, mesenchymal transition, and resistance to therapy. S100A4, in particular, is a small calcium-binding protein that can act both extracellularly and intracellularly, affecting a variety of multiple biological processes depending on its binding partners. S100A4 is predominantly located in the cytoplasm and, to a lesser extent, in the nucleus. It interacts directly with proteins such as p53, annexin 2, and myosin IIA heavy chain, thereby enhancing apoptosis, cell migration, and angiogenesis [
31,
32]. S100A4 has been identified as an upregulator of ZEB1 and SNAIL2 and is implicated in mesenchymal transition in GSCs [
24]. Furthermore, we demonstrated that FGFR1 and ZEB1 influence GBM cell proliferation and stemness [
16]. These findings suggest a link between FGFR1, S100A4, and ZEB1, all associated with GSC proliferation, mesenchymal transition, and resistance to therapy. Our functional studies confirmed that S100A4 knockdown sensitized both GC1 and GC2 cells to irradiation, reinforcing its role as a key mediator of radioresistance. The inhibition of S100A4 by pemigatinib suggests that FGFR1 signaling modulates stemness and mesenchymal traits in GBM through downstream effectors such as S100A4 and ZEB1.
Together, these results suggest that pemigatinib can exert radiosensitizing effects at least in part through the suppression of stemness and mesenchymal programs driven by FGFR1 and its downstream effectors FOXM1 and S100A4. These mechanistic insights indicate that FGFR inhibition may complement existing therapeutic strategies in GBM, although further studies will be needed to confirm this in additional models. Importantly, this effect was observed despite the absence of FGFR gene fusions or known activating mutations in our models, suggesting that functional FGFR1 pathway activity, rather than mutational status alone, may contribute to sensitivity to FGFR inhibition. This raises the possibility of a shift in biomarker strategies, moving from purely genomic profiling toward assessing pathway dependency, thereby potentially broadening the scope of patients who could benefit from FGFR-targeted therapy. In addition to biomarker-driven selection, receptor crosstalk may also shape the response to FGFR inhibition. Although pemigatinib inhibits FGFR1–3, we centered our analyses on FGFR1 because protein-level profiling in patient-derived GC1/GC2 [
33] showed robust FGFR1, no detectable FGFR2, and comparatively low FGFR3/FGFR4. Given the extensive RTK crosstalk in GBM, EGFR amplification/variants (e.g., EGFRvIII) may provide bypass signaling that limits the impact of FGFR1 blockade. Consistent with this, the feedback activation of EGFR upon FGFR inhibition and improved activity with dual FGFR–EGFR blockade have been reported [
34], and receptor co-regulation at clathrin-coated sites supports direct EGFR–FGFR interplay at the plasma membrane [
35].
An additional consideration is that FGFR3 expression was found to be higher in normal brain tissue compared to GBM (
Figure 1). Since pemigatinib also targets FGFR3, potential effects on healthy tissue must be taken into account. Two factors may mitigate this concern. First, although pemigatinib is a potent inhibitor of FGFR1–3, it displays markedly reduced activity against the majority of non-FGFR kinases (>100-fold selectivity), thereby limiting broad off-target kinase inhibition. Second, while the blood–brain barrier (BBB) is frequently disrupted within GBM lesions, allowing drug penetration into tumor areas, it remains largely intact in surrounding non-tumoral regions. This differential permeability is likely to restrict drug access to FGFR3-expressing normal parenchyma. Consistent with this, clinical data from trials such as FIGHT-202 have reported primarily on-target adverse effects, including hyperphosphatemia due to altered phosphate handling, along with mucosal toxicities, nail and skin changes, and ocular events. These effects are manageable and largely reversible with supportive measures such as phosphate binders, local care, and dose adjustments [
36,
37]. Neurological adverse events have not been a prominent feature, suggesting limited CNS toxicity.
Another important issue is the possibility of acquired resistance to pemigatinib. In other cancers such as cholangiocarcinoma, resistance has been attributed to secondary mutations in the FGFR kinase domain or activation of alternative signaling pathways. Although such mechanisms have not yet been described in GBM, the intrinsic plasticity of glioblastoma stem cells and the frequent activation of bypass pathways (e.g., EGFR, MET, PDGFR) suggest that resistance could also emerge in this context. In light of our findings showing context-dependent radiosensitization, further studies will be needed to determine whether combining pemigatinib with radiotherapy or temozolomide could help prevent or delay resistance in a subset of GBM. Clinical evaluation will ultimately be required to clarify the efficacy and feasibility of such strategies.
Although a recent review [
38] concluded that pemigatinib did not demonstrate efficacy in glioblastoma based on available clinical trials, it is important to note that this study primarily evaluated monotherapy, often in patients with previously treated recurrent GBM or other primary CNS tumors harboring FGFR1–3 mutations or fusions/rearrangements. Moreover, the molecular characterization of FGFR1–3 alterations has not been systematically compared between primary and recurrent tumors, which may differ significantly due to treatment-induced genetic and phenotypic changes. The ongoing phase II trial assessing pemigatinib (NCT05267106) also underscores the evolving landscape. Given the intrinsic complexity of GBM biology, the more aggressive and therapy-resistant nature of recurrent tumors, and the limited efficacy of monotherapies in this context, current conclusions may be premature and not definitive. Our findings suggest that FGFR1 inhibition, particularly in combination with radiotherapy, holds promise as a strategy for overcoming resistance mechanisms and improving therapeutic outcomes.
These findings are consistent with studies in other cancers, including triple-negative breast cancer, where the FGF2/FGFR1 transduction pathway induces the upregulation and secretion of S100A4 [
39], with extracellular S100A4 released by tumor or stromal cells acting in an autocrine or paracrine manner by binding to its RAGE receptor, thus promoting cell migration, invasion, and angiogenesis [
40,
41]. Additionally, exosomal S100A4 plays a key role in hepatocellular carcinoma metastasis, by activating STAT3 phosphorylation and upregulating osteopontin expression [
42], as well as inducing immunosuppression and non-small cell lung cancer development through STAT3 activation [
43].
One limitation is the use of a limited number of primary GSC models. While GC1 and GC2 reflect clinically relevant MGMT-unmethylated GBM subtypes, additional studies across a broader panel of primary GSCs and patient-derived xenografts are needed to confirm the generalizability of these findings and to better define the subset of GBM patients most likely to benefit from FGFR1-targeted radiosensitization. Additionally, we did not include non-malignant CNS cells (e.g., human astrocytes or neural progenitor/neuronal models), which would allow the estimation of a therapeutic index. Clinical experience with pemigatinib indicates predominantly on-target, manageable toxicities without prominent neurological adverse events [
36,
37], but head-to-head assays in normal CNS models will be important to define selectivity. Although our study employed patient-derived glioblastoma stem cells and relied on functional assays to assess treatment impact, future studies incorporating flow cytometry-based analysis of stemness markers (e.g., Olig2, Sox2, Nestin) will be important to further characterize phenotypic shifts induced by FGFR inhibition and radiation.
Importantly, both GC1 and GC2 GSCs were MGMT-unmethylated and resistant to TMZ, yet still responded to pemigatinib. These findings suggest that FGFR inhibition may provide therapeutic benefit in a patient subset for whom standard-of-care alkylating agents are ineffective. Furthermore, neither GC1 nor GC2 harbored FGFR gene fusions or activating mutations typically associated with FGFR-targeted therapy in other cancers, though multiple variants of unknown significance (VUS) were present. This suggests that high FGFR1 expression and functional pathway activity may serve as a broader biomarker of FGFR dependency, independent of genomic alterations.
Finally, our in vivo data using an orthotopic GC1 xenograft model showed that both pemigatinib and radiation monotherapy significantly extended survival compared to controls. Although the combination did not statistically outperform either monotherapy, it achieved the longest median overall survival (177.7 days). This outcome, while not formally synergistic, is clinically meaningful, especially given the limited options for MGMT-unmethylated GBM. However, given the lack of statistical significance, we recognize that further studies are required to confirm the clinical relevance of this combined approach. The in vivo experiments were designed to assess therapeutic efficacy, and the observed survival benefit is interpreted in this context only, without inference for CSC dynamics.
Taken together, our results suggest that the combination of pemigatinib with radiotherapy, potentially alongside TMZ, may offer therapeutic benefit for patients with newly diagnosed GBM, particularly those with MGMT-unmethylated tumors and no detectable FGFR fusions or mutations. However, further preclinical and clinical studies are needed to confirm the efficacy of this approach.