Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2025 Jun 17;6(6):102183.
doi: 10.1016/j.xcrm.2025.102183. Epub 2025 Jun 12.

BRAF/MEK inhibition induces cell state transitions boosting immune checkpoint sensitivity in BRAFV600E-mutant glioma

Affiliations

BRAF/MEK inhibition induces cell state transitions boosting immune checkpoint sensitivity in BRAFV600E-mutant glioma

Yao Lulu Xing et al. Cell Rep Med. .

Abstract

Resistance to v-raf murine sarcoma viral oncogene homolog B1 (BRAF) plus mitogen-activated protein kinase kinase (MEK) inhibition (BRAFi+MEKi) in BRAFV600E-mutant gliomas drives rebound, progression, and high mortality, yet it remains poorly understood. This study addresses the urgent need to develop treatments for BRAFi+MEKi-resistant glioma using preclinical mouse models and patient-derived materials. BRAFi+MEKi reveals glioma plasticity by heightening cell state transitions along glial differentiation trajectories, giving rise to astrocyte- and immunomodulatory oligodendrocyte (OL)-like states. PD-L1 upregulation in OL-like cells links cell state transitions to immune evasion, possibly orchestrated by Galectin-3. BRAFi+MEKi induces interferon response signatures, tumor infiltration, and suppression of T cells. Combining BRAFi+MEKi with immune checkpoint inhibition enhances survival in a T cell-dependent manner, reinvigorates T cells, and outperforms individual or sequential therapies in mice. Elevated PD-L1 expression in BRAF-mutant versus BRAF-wild-type glioblastoma supports the rationale for PD-1 inhibition in patients. These findings underscore the potential of targeting glioma plasticity and highlight combination strategies to overcome therapy resistance in BRAFV600E-mutant high-grade glioma.

Keywords: BRAF V600E; BRAF and MEK inhibitor adaptation; Galectin-3; T cell modulation; cell state transitions; high-grade glioma; immune checkpoint inhibition; programmed death-ligand 1.

PubMed Disclaimer

Conflict of interest statement

Declaration of interests The authors declare no competing interests.

Figures

None
Graphical abstract
Figure 1
Figure 1
Patient BRAFV600E-mutant HGG undergoes glial differentiation after treatment (A) Schematic of longitudinal analyses in two patients. (B) MRI scans of patient 49 showing tumor (left), post-surgical cavity (middle), and recurrence (right) after BRAFi+MEKi. (C) GO analysis showing gliogenesis pathway upregulation at second recurrence after BRAFi+MEKi in patient 12. (D) Scatterplot of glial and progenitor expression changes before and after BRAFi+MEKi in patient 12. (E) Representative IF images for GFAP, MBP, and OLIG2 before and after BRAFi+MEKi and chemoradiation in patient 49. High-magnification images of boxed areas are shown on the right. Scale bars: 180 μm (left) and 140 μm (right). Quantification of GFAP and MBP intensities and OLIG2+ cells in patient 49 (n = 5 tumor-containing images). ∗p < 0.05 and ∗∗p < 0.01 (Student’s t test). Mean ± SEM. (F) Representative H&E- and NESTIN-stained tumor sections before and after BRAFi+MEKi in patient 49. High-magnification images of boxed areas are shown on the right. Scale bars: 150 μm. See also Figure S1 and Table S1.
Figure 2
Figure 2
Orthotopic BRAFV600E-mutant mouse models show tumor heterogeneity (A) Schematic of BRAF-2341 and BRAF-M34 model generation. (B) MRI scans showing tumor mass in BRAF-2341 and BRAF-M34 HGG mice (yellow dashed outlines). (C) Representative low- and high-magnification images of H&E-stained coronal sections of BRAF-2341 and BRAF-M34 HGGs. Scale bars: 80 μm (top) and 60 μm (bottom). (D) Representative IF images of BRAF-2341. GFP and/or CRE expression mark tumor cells. DAPI (cell nuclei). Left: GFP and PROM1/CD133 double-positive tumor cells (white arrowheads). Middle: OLIG2 and PROM1/CD133 double-positive/GFP-negative glioma-associated OLs (white arrowheads). Right: GFP and CRE double-positive tumor cells (white arrowheads); tumor cells co-express GFAP and OLIG2 (yellow arrowheads) or OLIG2 alone (blue arrowhead). Single-channel images of the white-boxed area are shown next to the merged images. Scale bars: 60 μm (left), 45 μm (middle), and 75 μm (right). (E) Representative IF images of BRAF-M34 HGG. Left: CRE, NESTIN, and PROM1/CD133 triple-positive NSPC/CSC-like tumor cell (white arrowheads). CRE and PROM1/CD133 double-positive (NESTIN negative) OL-like tumor cells (yellow arrowheads). Right: NeuN, OLIG2, and GFAP single-positive cells (white arrowheads) in the tumor core featuring hypercellularity. Single-channel images of the white-boxed area are shown next to the merged images. Scale bars: 110 μm (left) and 50 μm (right). (F) Schematic of RCAS (replication-competent avian retrovirus)-induced BRAFV600E TP53-deleted (RCAS-BRAF) HGG and orthotopic, immunocompetent model development. (G) MRI scans showing the tumor infiltrating into the contralateral hemisphere (yellow dashed outlines). (H) Representative image of an H&E-stained coronal section of orthotopic RCAS-BRAF HGG showing hypercellular proliferation of malignant ovoid to spindled cells arranged in fascicles and sheets. Scale bar: 140 μm. (I) Representative IF images of RCAS-BRAF HGG. GFAP-positive astrocyte-like cells, infrequently double-positive for OLIG2 (green arrowhead). An abundance of NESTIN+ cells, frequently double-positive for OLIG2 (white arrowheads), and PDGFRα+ cells (white arrowheads). Single-channel images of the white-boxed area are shown next to the merged images. Scale bars: 90 μm (left) and 115 μm (right). See also Figure S2 and Table S2.
Figure 3
Figure 3
BRAFi+MEKi treatment promotes glial differentiation in BRAFV600E-mutant HGG (A) Schematic of BRAFi+MEKi treatment timeline and analyses (RNA-seq, flow cytometry, [FC[, and immunofluorescence [IF]) of BRAF-2341 HGG. (B) Heatmap from bulk RNA-seq data depicting average expression levels of stem/progenitor cell and glial differentiation-associated transcripts between two replicates in BRAF-2341 HGG after 3-day BRAFi+MEKi treatment. (C) Schematic of 14-day BRAFi+MEKi treatment timeline and analyses (scRNA-seq and IF) of RCAS-BRAF HGG. (D) Heatmap from scRNA-seq data showing transcripts in the RCAS-BRAF tumor cluster. (E) Cnetplot showing enrichment of glial-related transcripts from GO analysis of the scRNA-seq dataset from RCAS-BRAF HGG. (F) Representative IF images of control or BRAFi+MEKi-treated RCAS-BRAF HGGs stained for GFAP, NESTIN, and DAPI. Scale bar: 120 μm. Quantification of marker-positive cells within the tumor field (right) ∗∗p < 0.01 (unpaired t test). n = 4 mice/group, mean ± SEM. (G) Representative IF images of control/BRAFi+MEKi-treated RCAS-BRAF HGGs stained for MBP, PDGFRα, and DAPI. Scale bar: 170 μm. High-magnification images of the white-boxed areas are shown on the right. Quantification of marker-positive cells within the tumor field (right). ∗p < 0.05 (unpaired t test). n = 4 mice/group, mean ± SEM. (H) Schematic of BRAFi+MEKi treatment timeline and analysis of BRAFV600E HGG cell lines in vitro. (I and J) Representative ICC images of murine BRAF-M34 (I) and RCAS-BRAF (J) cell lines treated with control/BRAFi+MEKi and stained for MBP, OLIG2, NESTIN, and DAPI. Scale bars: 40 μm. ∗p < 0.05 (unpaired multiple t tests and Holm-Šídák method). n = 2 independent experiments, mean ± SEM. See also Figure S3.
Figure 4
Figure 4
BRAFi+MEKi treatment induces interferon response pathway signature and glial differentiation, with a PD-L1-expressing immunomodulatory subpopulation (A) Reactome pathway analysis revealed upregulated genes associated with interferon receptor signaling after 48-h BRAFi+MEKi treatment in the STN-49 cells. Numbers on the bar graph indicate the gene count. (B) Heatmap demonstrating expression changes of IFN-γ receptor-related transcripts after 24- and 48-h BRAFi+MEKi treatment to the STN-49 cell line. (C) Representative ICC images of murine and patient-derived cell lines treated with control/BRAFi+MEKi for 13 days. Scale bars: 40 μm. Quantification of (C). ∗p < 0.05 (multiple paired t tests). n = 2 independent experiments, mean ± SEM. (D) Representative IF images of BRAF-M34 HGGs stained for PD-L1, BRAF V600E, PLP1, and DAPI. Control panel: BRAFV600E-mutated tumor cell negative for PD-L1 (arrow). PD-L1-positive non-tumor cell (arrowhead). BRAFi+MEKi panel: differentiated BRAFV600E-mutant glioma cells with PLP1 and PD-L1 co-expression (white arrows). PLP1-negative, PD-L1-positive glioma cells (yellow arrows). PD-L1 only positive cells (arrowheads). Scale bars: 70 μm (left) and 80 μm (right). (E) Representative IF image of BRAFi+MEKi-treated BRAF-M34 HGG stained for PD-L1, Cre, PLP1, and DAPI. Differentiated Cre-positive glioma cell co-expressing PLP1 and PD-L1. Scale bar: 20 μm. (F) Quantification of (D). Proportions of BRAFV600E and PLP1 double-positive differentiated glioma cells with OL-like state positive or negative for PD-L1 (left) and proportions of PLP1-positive cells co-expressing BRAFV600E and/or PD-L1 (right) were measured and compared between groups. ∗p < 0.05 for BRAFV600E+PLP1+PD-L1+ triple-positive cells between treatment groups (two-way ANOVA with Sidak’s multiple comparisons test). n = 3 mice/group, mean ± SEM. (G) Representative IF images of human BRAFV600E-mutant HGG (patient 49) tissue before and after BRAFi+MEKi treatment, stained for PD-L1, Olig2, and DAPI. Scale bars, 180 μm. (H) Quantification of (G). n = 4 images from 2 consecutive sections of patient 49 tissue, ∗p<0.05 (unpaired t test), mean ± SEM. (I) The scatterplot of upregulated CD274 transcript (PD-L1) after BRAFi+MEKi treatment in patient 12. See also Figure S4.
Figure 5
Figure 5
PD-L1 expression is elevated in BRAF-mutant gliomas (A) Bar graph depicting percentage of patients with PD-L1+ tumors in the BRAF mutation/fusion versus BRAF-wild-type (no alterations) groups. n = 59 patients with BRAF mutation/fusion and 3,126 total patients (chi-squared tests). (B) Boxplot representing CD274 (PD-L1) transcripts per million in the BRAF mutation/fusion (n = 17) versus BRAF-wild-type (no alterations; n = 1,022) groups, from whole-transcriptome sequencing (Wilcoxon rank-sum test). (C) Representative IHC images for BRAF V600E and PD-L1 in consecutive sections of BRAFV600E-mutant HGG. Scale bars: 250 μm. See also Table S4.
Figure 6
Figure 6
BRAFi+MEKi enhances T cell infiltration and synergizes with ICI to improve T cell-dependent survival (A) Experimental timeline (green: 12–14 days treatment for CyTOF, IF, and scRNA-seq analyses; pink: continuous treatment for survival analysis) in all three murine models. (B) Frequency of populations of tumor-infiltrating lymphocytes in BRAF-2341 tumors by CyTOF analysis. 3 mice/group and two independent experiments. ∗∗p < 0.01, ∗∗∗p < 0.001 (two-way ANOVA with Sidak’s multiple comparisons test), mean ± SEM. (C) Graphs depicting the frequency of PD-1+CD8+ and PD-1+CD4+ (left) or CTLA-4+CD8+ and CTLA-4+CD4+ (right) T cells normalized to control by CyTOF analysis. 3 mice/group and two independent experiments. ∗p < 0.05, ∗∗p < 0.01, ∗∗∗p < 0.001, ∗∗∗∗p < 0.0001 (two-way ANOVA with Tukey’s multiple comparisons test), mean ± SEM. (D) Kaplan-Meier survival curve of orthotopic BRAF-2341 (left) or BRAF-M34 (right) HGG-bearing mice of all four treatment groups. Treatment initiation at day 30 (BRAF-2341) or day 14 (BRAF-M34) post-injection (black arrow). Data representative of two independent experiments and 5–6 mice/group (BRAF-2341) and 4–5 mice/group (BRAF-M34). p values are indicated in the graph and Tables S5 and S6 when compared to the control group. (E) Kaplan-Meier survival curve in immunocompetent C57Bl/6 mice with orthotopic BRAF-M34 HGG treated with BRAFi+MEKi and ICI, either concurrently (BRAFi+MEKi+ICI) or sequentially. Treatment initiation at day 14 post-injection (black arrow). 4–5 mice/group. p values are indicated in the graph and Table S7 when compared to the concurrent treatment group. (F) Kaplan-Meier survival curve in immunocompetent C57Bl/6 mice with orthotopic RCAS-BRAF HGG treated with BRAFi+MEKi and/or ICI for 14 days. Treatment period from day 22 to day 36 post-injection (shaded area). Data representative of two independent experiments and 7–10 mice/group. p values are indicated in the graph and Table S5 when compared to the control group. (G) Heatmaps of T cell inhibition- and apoptosis-associated transcripts in CD4+ and CD8+ T cells in the RCAS-BRAF model. (H) Densities of CD4+PD-L1+ and CD8+PD-L1+ T cells in BRAF-M34 and RCAS-BRAF (IF analysis). ∗p < 0.05, ∗∗∗p < 0.001, ∗∗∗∗p < 0.0001 (two-way ANOVA with uncorrected Fisher’s LSD). n = 4 mice/group, mean ± SEM. (I and J) Representative IF images of RCAS-BRAF tissue stained for PD-L1, CD4 (I) and CD8 (J). CD4+ and CD8+ cells positive for PD-L1 (arrows); CD4+ and CD8+ cells negative for PD-L1 (arrowheads). Scale bars: (I) 20 μm (top), 15 μm (bottom), (J) 10 μm (top), and 20 μm (bottom). Quantifications of cell populations before versus after treatment. ∗p < 0.05, ∗∗p < 0.01, ∗∗∗p < 0.001, ∗∗∗∗p < 0.0001 (two-way ANOVA with Sidak’s multiple comparisons test). n = 4 mice/group, mean ± SEM. (K) Heatmaps of T cell activation-associated transcripts in CD4+ and CD8+ T cells in the RCAS-BRAF model. (L) Heatmaps of MHC class I (green), II (red), and IFN-γ (black)-related transcripts associated with antigen presentation in CD4+ and CD8+ T cells in the RCAS-BRAF model. (M) Densities of CD4+CD69+ and CD8+CD69+ T cells from BRAF-M34 and RCAS-BRAF (IF analysis). ∗p < 0.05, ∗∗∗p < 0.001, ∗∗∗∗p < 0.0001 (two-way ANOVA with uncorrected Fisher’s LSD). n = 3–4 mice/group, mean ± SEM. See also Figures S5 and S6 and Tables S5–S7.
Figure 7
Figure 7
BRAFi+MEKi-induced Galectin-3 secretion links glial differentiation to PD-L1 upregulation (A) Timelines of in vitro experiments using STN-49 and aGBM5 cell lines. (B) Relative Galectin-3 concentrations measured over time with nELISA in supernatants of patient-derived cell lines treated with control/BRAFi+MEKi (two-way ANOVA with multiple comparisons). Simple linear regression ± SEM. (C and D) Scatterplots representing Galectin-3 expression levels in STN-49 (C) and aGBM5 (D) cell lines treated with DMSO or BRAFi+MEKi at 24 and 48 h (E and F) Representative ICC images of STN-49 (E) and aGBM5 (F) cell lines treated with BSA or recombinant Galectin-3 protein for 72 h and subsequently stained for PD-L1, MBP, or OPALIN, and DAPI. Scale bars: 40 μm. Bar graphs showing cellular quantification (% of total DAPI+ cells). ∗p < 0.05 (two-way ANOVA with Sidak’s multiple comparisons test). ns, not significant. n = 2–3 replicates from one independent experiment, mean ± SEM. See also Figure S7.

Update of

References

    1. Burnet N.G., Jefferies S.J., Benson R.J., Hunt D.P., Treasure F.P. Years of life lost (YLL) from cancer is an important measure of population burden--and should be considered when allocating research funds. Br. J. Cancer. 2005;92:241–245. doi: 10.1038/sj.bjc.6602321. - DOI - PMC - PubMed
    1. Ostrom Q.T., de Blank P.M., Kruchko C., Petersen C.M., Liao P., Finlay J.L., Stearns D.S., Wolff J.E., Wolinsky Y., Letterio J.J., Barnholtz-Sloan J.S. Alex's Lemonade Stand Foundation Infant and Childhood Primary Brain and Central Nervous System Tumors Diagnosed in the United States in 2007-2011. Neuro Oncol. 2015;16:x1–x36. doi: 10.1093/neuonc/nou327. Suppl 10. - DOI - PMC - PubMed
    1. Ostrom Q.T., Patil N., Cioffi G., Waite K., Kruchko C., Barnholtz-Sloan J.S. CBTRUS Statistical Report: Primary Brain and Other Central Nervous System Tumors Diagnosed in the United States in 2013-2017. Neuro Oncol. 2020;22:iv1–iv96. doi: 10.1093/neuonc/noaa200. - DOI - PMC - PubMed
    1. Ostrom Q.T., Patil N., Cioffi G., Waite K., Kruchko C., Barnholtz-Sloan J.S. Corrigendum to: CBTRUS Statistical Report: Primary Brain and Other Central Nervous System Tumors Diagnosed in the United States in 2013-2017. Neuro Oncol. 2022;24:1214. doi: 10.1093/neuonc/noaa269. - DOI - PMC - PubMed
    1. Mullard A. FDA approves IDH1 and IDH2 inhibitor for brain cancer. Nat. Rev. Drug Discov. 2024;23:731. doi: 10.1038/d41573-024-00135-y. - DOI - PubMed

MeSH terms