Graphical abstract graphic file with name fx1.jpg [78]Open in a new tab Highlights * • BRAFi+MEKi triggers glioma cell state transitions that foster immune evasion * • BRAFi+MEKi activates interferon response and simultaneously suppresses T cells * • High PD-L1 expression in BRAF-mutant GBM provides a criterion for anti-PD-1 therapy * • Concurrent BRAF/MEK and checkpoint inhibition enhance anti-tumor immunity and survival __________________________________________________________________ Xing et al. report that combined BRAF and MEK inhibition (BRAFi+MEKi) in BRAF^V600E-mutant high-grade glioma shifts tumor cell states and upregulates PD-L1, via Galectin-3 secretion, contributing to T cell suppression. Concurrent but not sequential immune checkpoint inhibition in mice overcomes these tumor-intrinsic adaptations, highlighting its translational promise for glioma therapy. Introduction Brain tumors cause the most years of life lost among cancers and are the leading cause of cancer-related death in children.[79]^1^,[80]^2^,[81]^3^,[82]^4 Unlike other cancers, targeted therapies for brain tumors remain limited, with notable exceptions including BRAF (v-raf murine sarcoma viral oncogene homolog B1) and IDH (isocitrate dehydrogenase) inhibitors now clinically used for BRAF- and IDH-mutant gliomas, respectively.[83]^5^,[84]^6 BRAF, a kinase in the mitogen-activated protein kinase (MAPK) pathway, links growth factor signals to transcriptional changes, thereby regulating cell proliferation, survival, and differentiation.[85]^7 It is the most mutated kinase in cancer, with the V600E mutation comprising 90% of BRAF alterations.[86]^8 BRAF^V600E occurs in 7% of pediatric and 4% of adult primary brain tumors, particularly epithelioid glioblastoma (GBM), pleomorphic xanthoastrocytoma (PXA), and ganglioglioma.[87]^9^,[88]^10^,[89]^11 In pediatric low-grade gliomas (pLGGs), BRAF^V600E confers poor outcomes with conventional therapies and increases the risk of malignant progression to high-grade gliomas (HGGs), especially with CDKN2A deletion.[90]^10^,[91]^12^,[92]^13^,[93]^14^,[94]^15 The BRAF^V600E mutation results in constitutive MAPK activation and loss of feedback inhibition, promoting oncogenic transformation.[95]^7^,[96]^16^,[97]^17 ATP-competitive BRAF monomer inhibitors (BRAFi)—vemurafenib, dabrafenib, and encorafenib—effective in melanoma,[98]^18^,[99]^19^,[100]^20^,[101]^21^,[102]^22 were repurposed for patients with BRAF^V600E-mutant glioma alongside broader BRAF^V600E screening.[103]^7 However, BRAFi monotherapy shows limited efficacy due to multiple adaptive resistance mechanisms, including MAPK pathway reactivation.[104]^7 Sustained MAPK suppression correlates with clinical benefit,[105]^23 leading to strong preclinical and clinical support for combining BRAF with mitogen-activated protein kinase kinase (MEK) inhibitors (MEKi).[106]^24^,[107]^25^,[108]^26 Combined BRAF and MEK inhibition (BRAFi+MEKi) outperforms chemoradiation in LGG and shows encouraging response rates (∼47% in pLGG, ∼54% in adult LGG, and ∼33% in adult HGG) with reduced toxicity compared to monotherapy.[109]^27^,[110]^28^,[111]^29 In 2022, dabrafenib plus the allosteric MEKi trametinib received histology-agnostic, accelerated Food and Drug Administration approval for patients with unresectable or metastatic BRAF^V600E/K-mutant solid tumors who have progressed following prior treatment and lack satisfactory alternatives.[112]^30^,[113]^31 In 2023, approval expanded to pediatric patients with BRAF^V600E-mutant LGG who require systemic therapy.[114]^6^,[115]^32 Despite promising objective response rates (ORRs), most BRAF^V600E-mutant gliomas escape BRAFi+MEKi, with residual tumor driving rebound, progression, resistance, and unbridled mortality.[116]^7^,[117]^27^,[118]^28^,[119]^33^,[120]^34^,[121]^35^,[1 22]^36^,[123]^37^,[124]^38^,[125]^39 Resistance mechanisms, well characterized in melanoma, remain understudied in gliomas. Nevertheless, both tumor types exhibit intrinsic, acquired, and adaptive resistance to BRAFi, with shared resistance mechanisms including MAPK reactivation[126]^7^,[127]^33^,[128]^36^,[129]^40 and MAPK-independent mechanisms such as activation of the PI3K/AKT/mTOR pathway.[130]^26^,[131]^33^,[132]^40^,[133]^41 Unlike melanoma, BRAF^V600E-mutant HGGs exhibit a higher intrinsic (primary) resistance often due to concomitant genetic alterations,[134]^37 like CDKN2A deletion and PI3K-mTOR mutations.[135]^42^,[136]^43 High intra-tumoral heterogeneity contributes to this intrinsic resistance, with polo-like-kinase 1 (PLK1)-high subpopulation characterized by prolonged G2-M phase and increased asymmetric cell division, driving recurrence.[137]^44 Hence, preclinical studies have explored combining BRAF^V600E inhibitors with PLK1, MEK, mammalian target of rapamycin (mTOR), or epidermal growth factor receptor (EGFR) inhibitors,[138]^40^,[139]^44^,[140]^45^,[141]^46^,[142]^47^,[143]^48 although only a few combinations have entered clinical trials ([144]NCT04201457 and [145]NCT03919071). Intrinsic resistance could also be attributed to the highly immunosuppressive glioma tumor microenvironment (TME), making immunotherapy extremely challenging, in contrast to melanoma’s immunologically active TME characterized by robust T cell infiltration.[146]^49^,[147]^50^,[148]^51^,[149]^52 Programmed cell death protein-1 (PD-1) and cytotoxic T-lymphocyte antigen-4 (CTLA-4) immune checkpoints, exploited by tumors to evade immunity, offer therapeutic targets for immune checkpoint inhibitors (ICIs)[150]^49^,[151]^50^,[152]^51 and were effective in BRAF^V600E-mutant metastatic melanoma.[153]^52^,[154]^53 However, ICIs have demonstrated limited efficacy in HGGs[155]^54^,[156]^55 althoughencouraging responses in replication-repair-deficient HGG and evidence of immune synergism with PD-1 blockade and MEKi (trametinib) in a small number of patients have been reported.[157]^56 Moreover, ERK1/2 phosphorylation predicts survival following anti-PD-1 immunotherapy in recurrent GBM.[158]^57 Considering the variability in BRAFi+MEKi responses across tumors,[159]^37 and the distinct intra- and inter-tumoral heterogeneity and TME of BRAF^V600E-mutant HGGs, their treatment adaptations require deeper investigation.[160]^58^,[161]^59^,[162]^60 Evidence suggests that therapies tap into tumor plasticity, forcing cells to transition to distinct cell states that enable survival and resistance of solid tumors.[163]^61 Understanding BRAFi+MEKi-induced adaptations is critical yet largely unexplored in gliomas due to limited patient cohorts; therefore, mechanisms of cell state transitions in response to treatment in BRAF^V600E-mutant glioma remain unknown. Preclinical models are essential tools to investigate potential mechanisms of glioma cell adaptations and plasticity in BRAF^V600E-mutant HGGs. In this study, we investigated changes in HGG cell states following BRAFi+MEKi treatment and uncovered cell plasticity as a potential adaptive resistance mechanism of BRAF^V600E-mutant HGG. BRAFi+MEKi heightened cell state transitions along glial differentiation trajectories, giving rise to astrocyte (AC)- and oligodendrocyte (OL)-like states. PD-L1 upregulation in OL-like cells linked cell state transitions to immune evasion, possibly orchestrated by Galectin-3. BRAFi+MEKi-induced adaptive transcriptional changes included upregulation of interferon (IFN)-γ response- and antigen presentation-associated gene signatures, suggesting enhanced T cell-mediated anti-tumor immunity and potentially linking cytotoxicity to therapy response. Therapy-resistant murine tumors exhibited diminished cytotoxicity and impaired antigen presentation to T cells, indicative of immune evasion. This effect can be reversed by concurrently combining BRAFi+MEKi with ICI treatment, leading to significantly improved survival in a T cell-dependent manner. PD-L1 expression was consistently elevated in BRAF-mutant versus BRAF-wild-type GBM patient cohorts, suggesting a pervasive immune evasion mechanism. Collectively, our parallel investigations of patient tumor samples, high-fidelity murine models, and patient-derived cell lines offer insights into BRAFi+MEKi modulation of tumor cell states and the tumor immune microenvironment and may hold promise for a rational strategy combining BRAFi+MEKi with ICIs to improve outcomes in BRAF^V600E-mutant HGGs. Results BRAF^V600E-mutant HGGs undergo cell state transitions along glial differentiation trajectories following extensive treatment in patients Cancer treatment induces cellular adaptations via non-genetic mechanisms to drive resistance. Adaptive responses to BRAFi+MEKi in HGG are poorly understood. To investigate potential cellular adaptations to BRAFi+MEKi treatment, we analyzed the transcriptomes of paired BRAF^V600E-mutant HGG from patients before dabrafenib + trametinib treatment and at recurrence (Figure 1A) using gene ontology (GO) analysis of bulk RNA sequencing (RNA-seq). Magnetic resonance imaging (MRI) confirmed recurrence (Figure 1B), and immunofluorescence (IF) and immunohistochemistry (IHC) were conducted for validation ([164]Figures 1E-F and [165]S1; [166]Table S1). GO analyses showed upregulation of genes engaged in gliogenesis, synapse organization, axonogenesis, and cell-cell adhesion among other pathways after BRAFi+MEKi treatment ([167]Figures 1C and [168]S1D). Figure 1. [169]Figure 1 [170]Open in a new tab Patient BRAF^V600E-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 [171]Figure S1 and [172]Table S1. We further investigated pathways associated with gliogenesis for specific cell states and found that they are significantly enriched for markers for neural stem/progenitor cell (NSPC) (PROM1/CD133), pre-oligodendrocyte progenitor cell (pre-OPCs) (ASCL1, HES6, EGFR, and DLL1), AC (GFAP and AQP4), myelin-forming OL (PROM1/CD133, MBP, and PLP1),[173]^62 and—albeit at lower rates—mature OL (ASPA, CNP, and CLDN11) states ([174]Figures 1D, [175]S1D, and S1E). Increased PROM1/CD133+ expression aligns with previous findings showing a CD133+ BRAF inhibitor-resistant cell population in BRAF^V600E-mutant GBM.[176]^44 PROM1/CD133 and NESTIN expression—typically associated with a stemness state in GBM—were not co-regulated since NESTIN transcript ([177]Figure 1D) and protein levels ([178]Figure 1F) were unchanged after BRAFi+MEKi. Further IF validation confirmed the upregulation of selected glial markers at the protein level, showing increased GFAP, MBP, and OLIG2 expression in post-treatment samples ([179]Figure 1E). In conclusion, heavily treated BRAF^V600E-mutant HGG retain NSPC markers and transition to AC-, pre-OPC-, and OL-like states, the latter reminiscent of pre-myelinating and mature OL-like cells. Orthotopic BRAF^V600E-mutant mouse models show intra-tumoral cellular heterogeneity To address small patient cohort limitations, particularly for BRAFi+MEKi treatment, we generated two BRAF^600E-mutant glioma models (BRAF-M34 and RCAS-BRAF), with distinct co-mutations on the C57BL/6 genetic background, inducing gliomas at 54 and 70 days post-injection, respectively ([180]Figures 2A, 2F, [181]S2A, and S2B; [182]Table S2, STAR Methods). We subsequently isolated cells from these endogenous BRAF-M34 and RCAS-BRAF gliomas and re-injected dissociated cells into C57BL/6 mouse brains to generate large orthotopic masses with high tumor take rate (BRAF-M34: 95%; RCAS-BRAF: 90%) ([183]Figure 2A; [184]Table S2), as visualized by MRI ([185]Figures 2B and 2G). Histopathologic analyses of orthotopic masses revealed higher grade features, including high nuclear pleomorphism, necrosis, and microvascular proliferation, with subtle model-specific differences ([186]Figures 2C, 2H, [187]S2C, and S2I; [188]Table S2, STAR Methods). IF revealed high intra-tumoral heterogeneity in BRAF-M34 and RCAS-BRAF orthotopic glioma tested alongside BRAF-2341, which we previously generated on the FVB/N background.[189]^26 Expression of GFAP, OLIG2, NESTIN, PROM1/CD133, and OPC markers PDGFRα and NG2 in single tumor entities was detected ([190]Figures 2D, 2E, 2I, [191]S2C–S2H, and S2J; [192]Table S2). Glial and neuronal markers are no longer lineage restricted as in the normal brain but are frequently co-expressed in orthotopic gliomas, consistent with human HGGs ([193]Figure 2I).[194]^63 Reactive ACs (GFAP+) and glioma-associated oligodendrocyte progenitor cells (OPCs) (PDGFRα/NG2+) and OLs (nuclear OLIG2/PROM1+)[195]^62^,[196]^64^,[197]^65^,[198]^66 identified by lack of GFP and CRE expression, were found in all three models as part of the TME ([199]Figure 2D, middle panel, [200]S2C, S2D, and S2H). PROM1/CD133 was co-expressed with NESTIN, potentially marking a stemness state ([201]Figure 2E), and with OLIG2, which encompasses highly proliferative OPC-like states in addition to the non-proliferative, mature OLs ([202]Figures S2F, S2H, and S2J) and, when cytoplasmic, also ACs ([203]Figure S2J).[204]^65 Staining with proliferation marker Ki67 revealed that PROM1/CD133-positive cells are less proliferative than OLIG2-single-positive cells, suggesting that PROM1/CD133 differentiate into OL-like cells.[205]^62 Our two murine models (BRAF-M34 and RCAS-BRAF) recapitulate features of human BRAF^V600E-mutant gliomas,[206]^10^,[207]^66^,[208]^67 including intra-tumoral cellular heterogeneity, with model-specific differences. These models provide robust preclinical tools, facilitating immunological studies due to their intact immune system and cross-model comparisons owing to their identical C57BL/6 genetic background. Figure 2. [209]Figure 2 [210]Open in a new tab Orthotopic BRAF^V600E-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 BRAF^V600E 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 [211]Figure S2 and [212]Table S2. BRAFi+MEKi heightenscell state transitions along glial differentiation trajectories in BRAF^V600E-mutant gliomas Our analyses of heavily treated BRAF^V600E-mutant HGG patient materials strongly suggested that BRAFi+MEKi treatment boosts cell states associated with glial differentiation ([213]Figures 1D and [214]S1D). To investigate these changes in additional samples, we assessed whether BRAFi+MEKi boosts similar cell state changes in murine BRAF^V600E-mutant gliomas. Moreover, we used single-cell RNA sequencing (scRNA-seq) of tumors in addition to bulk RNA-seq to analyze these changes on a more granular level. Established (BRAF-2341) and newly generated (RCAS-BRAF) mouse models were treated with dabrafenib + trametinib for up to 14 days, and tumors were analyzed by bulk RNA-seq or scRNA-seq and then flow cytometry (FC) and IF for validation ([215]Figures 3A and 3C). In BRAF-2341 cells, isolated by flow cytometry (FC) sorting for A2B5,[216]^67 BRAFi+MEKi increased the expression of transcripts associated with myelin-forming OL- (Plp1, Mbp, Cnp, Prom1/CD133, and Cldn11), mature OL- (Aspa), and AC-like states (Gfap and Clu) ([217]Figures 3B and [218]S1E). We confirmed that BRAFi+MEKi upregulated transcripts associated with AC- and OL-like states in two patient-derived BRAF^V600E-mutant glioma cell lines (aGBM5 and STN-49), albeit with quantitative differences in certain transcripts ([219]Figures S3A and S3B), consistent with their selected upregulation in patient tumors ([220]Figures 1D and 1E). Additionally, BRAFi+MEKi boosted the expression of transcripts associated with myelin-forming OL- (Mbp, Plp1, Pllp, and Prom1) and AC-like states (Gfap and Aqp4) in the RCAS-BRAF model, as analyzed by scRNA-seq and validated by IF ([221]Figures 3C–3E, [222]S3F, and S3G). GO analysis revealed significant upregulation of transcripts involved in gliogenesis ([223]Figure 3E), while IF confirmed upregulation of GFAP and MBP proteins, associated with glial differentiation (AC and OL) ([224]Figures 3F and 3G). Markers for NSPC- (Nes), pre-OPC- (Dll1, Egfr, and Hes6), and OPC-like (Pdgfrα and Cspg4) states were downregulated by BRAFi+MEKi in BRAF-2341 cells ([225]Figures 3B and [226]S1E), suggesting that NSPC-, pre-OPC-, and OPC-like cells are sensitive to short-term 3-day treatment. Decreased expression of transcripts for NSPC- and OPC-like states (Nes and Cspg4) was also detected, whereas some markers for pre-OPC-like states (Ascl1, Hes6, and Egfr) were unchanged in the RCAS-BRAF tumors ([227]Figure 3D), and the OPC-like state marker PDGFRα protein was reduced, as shown by IF ([228]Figure 3G). Figure 3. [229]Figure 3 [230]Open in a new tab BRAFi+MEKi treatment promotes glial differentiation in BRAF^V600E-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 BRAF^V600E 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 [231]Figure S3. In glioblastoma, NESTIN and PROM1/CD133 are markers associated with a stemness state that contributes to chemoradiation resistance and drives tumor recurrence.[232]^68^,[233]^69 We therefore investigated the effects of BRAFi+MEKi treatment on NESTIN and CD133/PROM1 cells in mice. Double-positive cell states decreased, as indicated by FC and IF, consistent with downregulated Nestin transcript with BRAFi+MEKi ([234]Figures S3C–S3E). The abundance of CD133/PROM1+ single-positive cells, however, increased while their overall abundance remained unchanged ([235]Figures S3C and S3D). This suggests that BRAFi+MEKi reduces NESTIN+ but not CD133/PROM1+ cell states; the latter appears to be intrinsically resistant to treatment, aligning with increased CD133/PROM1 transcript level in patient’s recurrent tumors after treatment ([236]Figure 1D) and prior reports of CD133+ cells involved in BRAFi resistance.[237]^44 Lastly, we tested BRAFi+MEKi effects on tumor cell states in vitro by treating murine (BRAF-M34 and RCAS-BRAF) and human (STN-49 and aGBM5) glioma cell lines with BRAFi+MEKi for 13 days. STN-49, specifically generated for this study, was derived from patient 49 before treatment ([238]Figures 3H and [239]S3H, [240]STAR Methods). Tumor cells were subsequently analyzed by immunocytochemistry (ICC) for markers for OL-like (MBP), pan-OPC/OL-like (OLIG2), and NSPC/CSC-like states (NESTIN). BRAFi+MEKi reduced tumor cell viability in all cell lines and increased MBP+ and OLIG2+ cell abundance ([241]Figures 3I, 3J, [242]S3I, and S3J), consistent with a treatment-induced boost in glial differentiation states (AC- and OL-like) observed in vivo. NESTIN+ cells, abundant at baseline (vehicle treatment) in all four cell lines, were unchanged with 13-day BRAFi+MEKi treatment in murine cells, which is further corroborated by findings from the RCAS-BRAF model ([243]Figure 3F) and patients ([244]Figures 1D and 1F). These data indicated that cells with NSPC/CSC-like states initially decreased with treatment ([245]Figures 3B, 3D, and [246]S3C–S3E) and were reinstated with treatment over time ([247]Figures 3I, 3J, [248]S3I, and S3J). Notably, mRNA and protein levels were not concurrent in the RCAS-BRAF tumors, and model-specific differences are evident as Nestin+ cells are more abundant in untreated RCAS-BRAF tumors ([249]Figure 2I) compared to BRAF-2341 and BRAF-M34 tumors ([250]Figures 2D and 2E; [251]Table S2). These findings demonstrate that BRAFi+MEKi treatment temporarily suppresses NSPCs and reduces cells in pre-OPC and OPC-like states, while consistently heightening transitions toward glial cell differentiation states reminiscent of myelin-forming OL and AC in BRAF^V600E-mutant gliomas. BRAFi+MEKi boosts IFN response pathway gene expression signature in BRAF^V600E-mutant HGG BRAF-altered glioma exhibits higher CD8^+ T cell infiltration and major histocompatibility complex MHC class I expression, when compared to BRAF-wild-type glioma,[252]^63^,[253]^64 suggestive of a robust anti-tumor immune response that could be exploited therapeutically in combination with BRAFi+MEKi. We therefore investigated whether BRAFi+MEKi alters immunomodulatory programs in BRAF^V600E-mutant glioma cells, using transcriptome analyses and quantitative reverse-transcription PCR (RT-qPCR) for validation. We first analyzed the whole transcriptomes of BRAF^V600E-mutant patient-derived (STN-49 and aGBM5) and murine (BRAF-2341) glioma cell lines for differential gene expression and Reactome pathway analysis. We observed that 48-h BRAFi+MEKi treatment upregulated IFN-γ receptor-associated signature with 67 transcripts across human and mouse transcriptomes ([254]Figures 4A, 4B, [255]S4A–S4C, and S4F). Transcripts for MHC genes were included in this signature, along with other inflammation-related transcripts. RT-qPCR confirmed upregulation of selected MHC class I/II genes (HLA-A, HLA-B, HLA-DRA, and CIITA) in patient-derived and murine cell lines ([256]Figures S4D, S4E, and S4G; [257]Table S3). These data suggested that BRAFi+MEKi alters the immunomodulatory activity of HGG cells, heightening antigen presentation by MHC class I/II molecules and potentially boosting anti-tumor immune responses. Figure 4. [258]Figure 4 [259]Open in a new tab 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: BRAF^V600E-mutated tumor cell negative for PD-L1 (arrow). PD-L1-positive non-tumor cell (arrowhead). BRAFi+MEKi panel: differentiated BRAF^V600E-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 BRAF^V600E 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 BRAF^V600E and/or PD-L1 (right) were measured and compared between groups. ∗p < 0.05 for BRAF^V600E+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 BRAF^V600E-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 [260]Figure S4. BRAFi+MEKi induces glial differentiation cell state transitionsand a PD-L1-expressing immunomodulatory subpopulation IFN-γ signaling induces the PD-1 immune checkpoint pathway, which is frequently hijacked by cancer cells to circumvent anti-tumor immune responses and is an important immunotherapy target.[261]^70 We therefore investigated BRAFi+MEKi effects on PD-1 signaling through Reactome pathway analysis of whole transcriptomes, validated by ICC and IF. PD-1 signaling was one of the pathways significantly altered by BRAFi+MEKi treatment, in human and murine BRAF^V600E-mutant glioma cell line transcriptomes ([262]Figures 4A, [263]S4H, and [264]S5A). PD-1 signaling components, including T cell receptor complex protein CD247, and the PD-1 ligand PD-L1 (CD274), were upregulated, as shown by gene network analysis ([265]Figure S5B). Next, we rigorously tested whether PD-L1 protein and gene expression aligned, using immunostainings. BRAFi+MEKi indeed increased PD-L1 protein expression in BRAF^V600E-mutant HGG cells in vitro ([266]Figure 4C), and in vivo ([267]Figures 4D–4F), consistent with the upregulation of PD-L1 transcript. We also found upregulated PD-L1 expression at protein and transcriptomic levels in BRAF^V600E glioma patients post-BRAFi+MEKi treatment ([268]Figures 4G–4I). PD-L1 is also expressed in murine BRAF-2341 gliomas, accounting for 83% of NESTIN+ cells ([269]Figure S4I). Immunomodulatory roles for disease-associated OL-lineage cells have been reported, aside from their function in myelination.[270]^71^,[271]^72^,[272]^73^,[273]^74 We then investigated if OL-like cell states enriched in response to BRAFi+MEKi express the immunomodulatory PD-L1, by assessing the co-expression of PD-L1 with OL markers PLP1, MBP, and OLIG2, by immunostaining. BRAFi+MEKi significantly upregulated PLP1 expression in BRAF-M34 glioma (BRAF^V600E+PLP1^+) after 12 days of treatment ([274]Figures 4D–4F), consistent with an increase in OL-like cell states in the patient tumor ([275]Figure 3I). The vast majority (85.92%) of these OL-like tumor cells expressed PD-L1 ([276]Figure 4F, left panel), suggesting that treatment induced immunomodulatory function in OL-like glioma cells. Interestingly, 40.28% of PLP1^+ BRAF^V600-negative cells were immunoreactive for PD-L1 even before treatment (vs. 25.87% after treatment), indicating that these non-malignant OLs, also called glioma-associated OLs, might be hijacked by glioma cells to promote immunosuppression. Co-expression of MBP/Olig2 and PD-L1 was confirmed in vitro to complement our in vivo results ([277]Figure S4J). Positive fold changes for co-expression in murine and human cell lines are variable, suggesting model-specific differences ([278]Figure S4K, left panel). Lastly, co-expression of pan-OL marker OLIG2 and PD-L1 in BRAF^V600E -mutant patient tumors revealed a 10-fold increase in OLIG2/PD-L1 double-positive cells after BRAFi+MEKi treatment ([279]Figures 4G and 4H) In conclusion, our data provide compelling evidence that BRAFi+MEKi treatment induces a transition to differentiating glia cell states, with a subpopulation of OL-like glioma cells acquiring immunomodulatory properties via PD-L1 expression, establishing a direct mechanistic link between cell state transitions and immune evasion. PD-L1 expression is elevated in BRAF-mutant gliomas Our data demonstrated that BRAFi+MEKi upregulated PD-L1 expression, a mechanism for T cell suppression and a key criterion for patient enrollment in PD-1 inhibition therapy.[280]^75 Therefore, we evaluated PD-L1 expression in a large cohort of patients with BRAF-mutant glioma, including those with BRAF^V600E. First, we evaluated the association between PD-L1 expression and BRAF mutation status in a large cohort of adult glioblastoma (IDH-wild-type) specimens (n = 3,126), undergoing different characterization analyses ([281]STAR Methods). PD-L1 expression by IHC was significantly higher in BRAF-mutant (54%) than in BRAF-wild-type glioblastoma (17%, p < 0.0001) ([282]Figure 5A). This was orthogonally validated using whole-transcriptome sequencing data, showing increased PD-L1 (CD274) gene expression in BRAF-mutant (n = 17) versus BRAF-wild-type (n = 1,022) glioblastoma (p = 0.045) ([283]Figure 5B). We found that PD-L1 and BRAFV600E showed a high degree of co-expression by IHC ([284]Figure 5C), further corroborating PD-L1 expression in BRAF^V600E-mutant glioma cells. We also showed that PD-L1 is expressed in various BRAF^V600E-mutant glioma types, including ganglioglioma, PXA, and malignant astrocytoma/glioma, albeit with a varied frequency score, as determined by IHC ([285]Figure S5C; [286]Table S4). Figure 5. [287]Figure 5 [288]Open in a new tab 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 BRAF^V600E-mutant HGG. Scale bars: 250 μm. See also [289]Table S4. Our findings demonstrate that BRAF mutations, including the BRAF^V600E variant, are associated with elevated PD-L1 expression in GBM and other glioma subtypes. This correlation suggests that patients with BRAF-mutant gliomas may harbor suppressed T cells that are reactivatable through anti-PD-1 therapy, potentially eliciting a robust anti-tumor immune response and driving a significant survival benefit. BRAFi+MEKi treatment enhances tumor-infiltrating T cells The efficacy of T cell checkpoint therapy is significantly influenced by the presence of tumor-infiltrating T cells.[290]^76^,[291]^77 Since BRAF-altered glioma exhibits robust CD8^+ T cell infiltration,[292]^11^,[293]^78^,[294]^79 and our data showed that they express PD-L1 ([295]Figures 4C, 4D, 4F, and 4H), we closely assessed tumor-infiltrating T cells in BRAF^V600E-mutant murine glioma with and without BRAFi+MEKi treatment, using high-dimensional single-cell mass cytometry (CyTOF), scRNA-seq, and validation by IF ([296]Figure 6A, [297]STAR Methods). We identified robust infiltration of CD4^+ and CD8^+ effector T cells (T[eff]), and CD4+FoxP3+ regulatory T cells (T[reg]) within the tumor mass, as confirmed by CyTOF ([298]Figures 6B and [299]S6A–S6D), scRNA-seq ([300]Figures 6G, 6K and 6L), and IF ([301]Figures 6H-J, 6M, [302]S5D, and [303]S6G). The presence of tumor-infiltrating T cells in murine BRAF^V600E-mutant gliomas is consistent with a heightened frequency of CD3^+CD8^+ T cells in human BRAF-altered glioma compared to BRAF-wild-type astrocytoma.[304]^78^,[305]^79 BRAFi+MEKi further elevated the frequency of CD4^+ and CD8^+ T cells in all three models ([306]Figures 6B and 6C, 6H–6J, 6M, and S6G), consistent with upregulation of the antigen presentation machinery ([307]Figures 4B and [308]S4B–S4G) and suggestive of anti-tumor immunity heightened by treatment. Figure 6. [309]Figure 6 [310]Open in a new tab 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 [311]Tables S5 and [312]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 [313]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 [314]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 [315]Figures S5 and [316]S6 and [317]Tables S5–[318]S7. Combination of BRAFi+MEKi with ICI enhances survival outcomes in a T cell-dependent manner Our findings provided a strong rationale for combining BRAFi+MEKi with PD-1/PD-L1 ICI to boost T cell activation and improve survival. To explore this, we assessed the effects of BRAFi+MEKi combined with ICIs on the T cell compartment in our three mouse models ([319]Figure 6A). Mice bearing orthotopic BRAF^V600E-mutant gliomas were treated with dabrafenib and trametinib (BRAFi+MEKi) and/or anti-PD-L1 antibody. Since PD-1/PD-L1 inhibitors alone have shown no survival benefit in HGGs,[320]^54^,[321]^55 we also included CTLA-4 blockade (α-PD-L1+α-CTLA-4; referred to as ICI) ([322]Figures 6A and [323]S5E). Concurrent, quadruple treatment (BRAFi+MEKi+ICI) significantly improved survival over BRAFi+MEKi or ICI alone in all three models ([324]Figures 6D–6F; [325]Tables S5 and [326]S6). This survival benefit is associated with T cell reactivation, as shown by reduced frequencies of PD-1- and CTLA-4-expressing CD4+/CD8+ T cells ([327]Figure 6C) and T[reg] cells ([328]Figures S6A–S6D). To assess model-specific differences for BRAF-M34 and RCAS-BRAF further, we analyzed the frequency of CD4^+ and CD8^+ T cells co-expressing inhibitory (PD-L1/PD-1) and activation (CD69) markers. BRAFi+MEKi increased PD-L1+ and PD-1+ T cell frequencies ([329]Figures 6H–6J, [330]S6F, and S6H), suggesting that the PD-L1/PD-1 checkpoints become activated to escape BRAFi+MEKi-induced T cell activation. Only PD-1+ T cells in the RCAS-BRAF model showed the opposite trend, most likely undergoing apoptosis after treatment ([331]Figures 6G and [332]S6H). We observed differences in T cell inactivity across the models. RCAS-BRAF showed the highest T cell suppression, while BRAF-2341 had the least ([333]Figure S6F). This variability is expected and consistent with reported model-specific differences and differences in the immune responses, even in genetically identical mice ([334]Figures 6D–6F). Interestingly, both BRAF-M34 and RCAS-BRAF models did not respond to ICI alone, suggesting their T cells were unresponsive to activation ([335]Figures 6D, 6F, and 6H). However, their T cells were responsive to BRAFi+MEKi alone, with the BRAF-M34 model showing significantly higher CD4^+CD69^+ and CD8^+CD69^+ T cells, indicative of early T cell activation, compared to the RCAS-BRAF model ([336]Figure 6M). Furthermore, our in vitro data showed that BRAF-M34 glioma cells with OL-like states have less immunosuppressive effect on T cells ([337]Figure S4K). The survival benefits seen with therapeutic antibodies, either alone or in combination with BRAFi+MEKi, were completely abolished in athymic mice lacking T cells. Similar results were observed in BRAF-M34 mice that are depleted of T cells, confirming T cell-dependent anti-tumor efficacy ([338]Figure S6E; [339]Tables S5 and [340]S6). Although T cells are not strictly required for the anti-tumor effects of BRAFi+MEKi, increased T cell activation and anti-tumor immunity may contribute to the treatment’s effectiveness, particularly in the BRAF-M34 model. Lastly, we tested whether sequential therapy optimizes BRAFi+MEKi responses, similar to what has been reported for BRAF^V600E-mutant melanoma patients.[341]^80 ICI 1^st showed a trend for improved survival over BRAFi+MEKi 1^st, which provided no survival benefit over BRAFi+MEKi treatment alone. Importantly, concurrent quadruple treatment provided the greatest survival benefit and is superior to any sequential approach ([342]Figures 6D and 6E; [343]Table S7). These findings suggest that patients with BRAF^V600E-mutant gliomas are most likely to benefit from a combination treatment regimen integrating BRAFi+MEKi and ICIs concurrently. Synergistic effect of BRAFi+MEKi with ICI on T cell reactivation Our data demonstrated that quadruple (BRAFi+MEKi+ICI) treatment overcomes resistance observed with dual therapies (BRAFi+MEKi and ICI alone) in the RCAS-BRAF model ([344]Figure 6F). To gain insights into potential differences in T cell dynamics with dual and quadruple treatments, we analyzed CD4+ and CD8+ T cell and tumor clusters in scRNA-seq data from the RCAS-BRAF model when mice reached their disease-related endpoint ([345]Figure 6A). BRAFi+MEKi moderately upregulated selected suppression markers on T cells, including Cd274 (PD-L1), and robustly upregulated others, such as Cd276, Eomes, Socs, Fas, and Fasl in both CD4^+ and CD8^+ T cell clusters ([346]Figure 6G). Foxp3 was upregulated in CD4^+ T cells post-treatment, indicating the expansion of T[reg] cells. BRAFi+MEKi downregulated activation markers in CD4^+ T cells, including Gzmk, Gzmb, IL2ra, and others, while upregulating immunosuppressive markers, including Cd27, Cd7, Tnfrsf18, and Tnfrsf4 in CD8^+ T cells ([347]Figure 6K), suggesting the presence of dysfunctional CD8^+ T cells after BRAFi+MEKi treatment.[348]^81 We also found that treatment reduced expression of MHC class I and II genes in CD4^+ and CD8^+ T cells ([349]Figure 6L). MHC class I gene expression was diminished in tumor cells and tumor-associated macrophages/microglia ([350]Figures S6J and S6K). This downregulation was suggestive of suppressed antigen presentation, potentially enabling immune evasion and BRAFi+MEKi resistance at the survival endpoint. ICI alone upregulated T[reg] expansion markers, including Foxp3 and Btla ([351]Figures 6G and 6K). Moreover, increased expression of inhibitory markers like Ctla-4, Lag3, and Pdcd1 ([352]Figures 6G and 6K) in CD4^+ and CD8^+ T cells, and increased MHC and Ifng expression, indicated activation balanced with suppression of T cell activity ([353]Figures 6K and 6L). ICI also caused excessive pro-inflammatory cytokine production in CD4^+ T cells and tumor cells, resembling a “cytokine storm” ([354]Figure S6I). In contrast, quadruple treatment induced expression of genes linked to T cell activation and effector functions, including Itgae, Gzmk, Cd28, Tbx21, Itga1, Cd69, and Tnfrsf9 ([355]Figure 6K). Importantly, MHC class I/II gene expression was restored for effective antigen presentation without causing excessive cytokine production ([356]Figures 6L and [357]S6I). MHC class II gene upregulation in CD8^+ T cells is associated with memory T cell differentiation ([358]Figures 6L and [359]S6D).[360]^82 This may contribute to a more durable anti-tumor immune response. Our findings indicate that BRAFi+MEKi resistance is attributed to T[eff]cell suppression, T[reg] expansion, and impaired antigen presentation, which collectively could lead to immune evasion in the RCAS-BRAF model. While ICI activates T cells, it also induces severe cytokine release syndrome (cytokine storm) due to excessive cytokine production. In contrast, quadruple treatment effectively ensures full activation of CD4^+ and CD8^+ T cells, which could be pivotal for overcoming therapy resistance and conferring a significant survival benefit. BRAFi+MEKi-induced Galectin-3 secretion links glial differentiation to PD-L1 upregulation To investigate the mechanism for cell state transitions and PD-L1 expression by BRAFi+MEKi, we assessed the secretome of BRAF^V600E-mutant glioma cells in vitro using a high-throughput enzyme-linked immunosorbent assay (nELISA). STN-49 and aGBM5 cell lines were treated with BRAFi+MEKi over 13 days, mimicking the duration of in vivo treatments ([361]Figure 7A, [362]STAR Methods). Molecular factors measured by nELISA that were either upregulated or below the detectable threshold post-treatment are presented in a heatmap ([363]Figure S7A). BRAFi+MEKi significantly elevates the secretion of the carbohydrate-binding lectin family protein Galectin-3[364]^83 ([365]Figure 7B), with levels peaking at day 13, coincidently with increased glial differentiation ([366]Figures S3I and S3J). RNA-seq confirmed upregulation of the Galectin-3-encoding gene LGALS3 with 48 h treatment of BRAFi+MEKi ([367]Figures 7C and 7D). In addition to Galectin-3, other factors interleukin (IL)-23 and IL-32 were also upregulated post-treatment, albeit the timing of upregulation varied between STN-49 and aGBM5 cell lines ([368]Figures S7B and S7C). This indicates that cytotoxic response rates to BRAFi+MEKi could be cell line specific. Figure 7. [369]Figure 7 [370]Open in a new tab 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 [371]Figure S7. Next, we investigated a potential direct role for Galectin-3 in inducing glial differentiation cell state transitions and PD-L1 expression in STN-49 and aGBM5 cell lines ([372]Figure 7A). Adding recombinant Galectin-3 to cells in vitro efficiently enhanced both glial differentiation cell states and PD-L1 expression in STN-49 cells ([373]Figure 7E), whereas aGBM5 exhibited only an upward trend in PD-L1 expression in differentiated glial populations ([374]Figure 7F). These findings suggest that Galectin-3 secretion from glioma cells potentially provides a key link between tumor differentiation and PD-L1 upregulation after BRAFi+MEKi treatment. Discussion BRAF and MEK inhibitors have revolutionized treatment for BRAF^V600E-mutant cancers, including gliomas, where targeted therapies are otherwise limited. Nevertheless, treatment challenges persist, particularly in glioma patients who experience tumor rebound after treatment cessation and in patients who develop therapy resistance.[375]^84 Resistance mechanisms can be categorized as primary (intrinsic), adaptive, and acquired. While intrinsic and acquired resistance mechanisms to BRAFi and BRAFi+MEKi are well described in several solid tumors, like melanoma, the specific mechanisms underlying resistance in glioma remain poorly understood.[376]^33^,[377]^85 Our study addresses this gap, focusing on glioma lineage-specific resistance. Compared to melanoma, the higher intrinsic resistance to BRAFi+MEKi of BRAF^V600E-mutant HGG underscores the importance of cancer lineage-specific studies. The high rates for intrinsic resistance in HGG may partially stem from the high intra-tumoral heterogeneity compared to LGG.[378]^44^,[379]^67 Our data confirm that BRAF^V600E-mutant HGG express gene programs of both neuronal and glial lineages, suggesting diverse differentiation states within tumors. We found that BRAFi+MEKi treatment induces significant shifts in glioma cell state, favoring more differentiated phenotypes at the expense of stem- and progenitor-like states. Our in vitro data support this observation, showing that surviving cells adapt to treatment by acquiring a more differentiated state,[380]^86 which is associated with upregulated immunomodulatory PD-L1 expression. This phenotypic heterogeneity and cell plasticity may underlie the high intrinsic resistance and rapid adaptation to treatment observed in BRAF^V600E-mutant HGGs. Indeed, similar mechanisms are emerging as key features glioblastomas, where tumor cells transition among four major cellular states.[381]^63 Lineage-specific responses to BRAFi+MEKi were also observed. CD133/PROM1, a marker for both CSCs and differentiating OLs,[382]^62^,[383]^87 remains stable or increases with BRAFi+MEKi; whereas NESTIN+ cells decrease after short-term BRAFi+MEKi but persist with prolonged exposure, suggesting distinct CSC populations with unclear roles in tumor recurrence. Notably, differentiated glioma cells have been shown to accelerate CSC-driven tumor growth,[384]^88 implying that BRAFi+MEKi-induced differentiation could paradoxically support tumor persistence by maintaining a CSC reservoir. Traditionally, therapy resistance in glioma is associated with cell de-differentiation and the acquisition of stem cell-like states. In contrast, we demonstrate that BRAFi+MEKi induces a shift toward a more differentiated, immunosuppressive state, which may be a unique adaptation and resistance mechanism in glioma. Analysis of paired pre-/post-treatment glioma samples identified 13 putative resistance-associated alterations, including mutations in ERRF1, BAP1, ANKHD1, and MAP2K1.[385]^33 Despite a small patient cohort size (4 out of 15 with BRAFi+MEKi), only about half of the cases harbored de novo mutations, suggesting that non-genetic adaptive resistance mechanisms, such as tumor cell plasticity and cell state transitions described in our studies, may play an important role in resistance in BRAF^V600E-mutant glioma. Our work shows that BRAFi+MEKi induces cellular plasticity, which may serve as a therapeutic vulnerability. Further studies are needed to determine whether the adaptive mechanisms also extend to non-BRAF^V600E-mutant and BRAF-wild-type gliomas. Our data reveal that BRAFi+MEKi may induce adaptive resistance via increased Galectin-3 secretion from glioma cells. Galectin-3 promotes cell survival and proliferation[386]^89 and induces MAPK reactivation via K-Ras-Raf-ERK1/2 signaling,[387]^89 facilitating glioma cell differentiation while simultaneously upregulating PD-L1 as an adaptive response to BRAFi+MEKi.[388]^90^,[389]^91^,[390]^92 It is not well understood what drives cancer cell state transitions in glioma, although they are a well-recognized adaptive mechanism enabling immune evasion, therapy resistance, and metastasis in many cancer types,[391]^93 influencing both plasticity and the TME. We found that BRAFi+MEKi-induced Galectin-3 secretion promotes glioma cell differentiation into AC- and OL-like states, mirroring its role in normal OL differentiation with increased MBP expression.[392]^94^,[393]^95 This differentiation is associated with increased PD-L1 expression, fostering an immunosuppressive TME. Given the antigen-presenting capabilities of ACs and OLs,[394]^71^,[395]^72^,[396]^96^,[397]^97 these cell state transitions may actively shape immune responses in glioma. Gliomas are considered immune inactive due to their low mutational burden,[398]^98 but BRAF^V600E-mutant gliomas are more immune rich, with elevated CD8^+ T cell infiltration, MHC class I expression,[399]^78 and PD-L1 expression (shown here), suggesting intrinsic ICI sensitivity. However, clinical trials with PD-1 and/or CTLA-4 inhibitors show limited efficacy in glioblastoma. PD-1 and CTLA-4 blockade are effective as second-line treatment after BRAFi+MEKi in BRAF^V600E-mutant metastatic melanoma,[400]^51^,[401]^52 providing a strong rationale for exploring BRAFi+MEKi+ICI combinations in glioma. Our study demonstrates that this concurrent quadruple therapy significantly improves survival compared to BRAFi+MEKi or ICI alone across all preclinical models, by preserving antigen presentation, enhancing T cell responses, and conferring a significant survival advantage. While clinical trials of BRAFi+MEKi+ICI in BRAF^V600E gliomas are lacking, melanoma studies provide insights. Patients previously treated with BRAFi+MEKi showed similar outcomes when subsequently treated with PD-1 or PD-1+ipilimumab, though the combination treatment had higher toxicity.[402]^52 Subsequent BRAFi+MEKi treatment after anti-PD-1 monotherapy doubled objective response rates compared to anti-CTLA-4 treatment alone.[403]^99 This is consistent with our findings showing that sequential ICI followed by BRAFi+MEKi offers benefit, although less than concurrent quadruple treatment. Additionally, BRAF-wild-type melanoma patients responded better to anti-PD-1+MEKi, while BRAF-mutant patients had lower ORR due to acquired resistance to treatment.[404]^100 Toxicity remains a concern, as BRAFi+MEKi causes fever, leukopenia, and hyponatremia, while ICIs induce immune-related adverse effects.[405]^27^,[406]^101 The principal reason for stopping sequential treatment (ICI 1^st, BRAFi+MEKi 2^nd) was disease progression rather than adverse effects, which is promising and could support quadruple treatment in glioma. Dose reductions for BRAFi+MEKi could be considered for patients on quadruple treatment when tumors respond.[407]^53 Quadruple treatment may increase toxicity, but pediatric patients - enriched for BRAF-mutant gliomas - may tolerate it better than adults, and dose reductions or sequential treatment combined with Galectin-3 inhibitors could help balance efficacy and safety. Clinical data in metastatic melanoma and glioma highlight the necessity for further preclinical studies to refine treatment timing and minimize toxicity in HGG. In our preclinical models, BRAFi+MEKi could synergize with ICI by increasing T cell activity and antigen presentation, thus enhancing tumor intrinsic sensitivity. We hypothesize that sequential treatment (BRAFi+MEKi 1^st, ICI 2^nd) is less effective because it allows time for tumor cells to suppress T cell activity to reduce ICI efficacy, potentially through Galectin-3 secretion[408]^102 ([409]Figure S7D). Future studies will address whether Galectin-3 inhibition could further improve therapeutic outcomes by simultaneously targeting adaptive resistance and immunosuppressive TME. In summary, our study underscores the necessity of implementing concurrent quadruple therapy to generate more durable responses, thereby mitigating resistance mechanisms in BRAF^V600E-mutant HGG and GBM. Incorporating Galectin-3 inhibitors into existing treatment regimens for these gliomas also offers a promising strategy to enhance therapeutic efficacy while managing toxicity, thereby improving the overall quality of life for patients. Limitations of the study Due to the rarity of matched pre- and post-treatment BRAF^V600E-mutant glioma patient samples, we generated two patient-derived cell lines and two immunocompetent mouse models/murine cell lines that effectively recapitulate key disease features.[410]^103 Despite expected inter-individual variability in treatment response within models,[411]^104^,[412]^105^,[413]^106 concurrent quadruple treatment consistently improved survival across all models, except for limited response in the RCAS-BRAF model, potentially caused by a higher PD-L1+ T cell proportion. Future studies are required to elucidate resistance mechanisms in this model. Additionally, future studies should fully characterize BRAFi+MEKi-induced cytokine alterations and their effects on glioma cells, and on CD4/CD8 T cell activation. Lastly, variability in drug responses across different patient-derived cell lines may reflect intrinsic differences in tumor cell types or Galectin-3 receptor expression, highlighting the need for single-cell analyses and expanded biological replicates or patient-derived cell lines. Resource availability Lead contact Requests for further information, resources, and reagents should be directed to and will be fulfilled by the lead contact: Claudia K. Petritsch, cpetri@stanford.edu and clpetritsch39@gmail.com. Materials availability Murine BRAF-M34 and RCAS-BRAF cell lines, and patient-derived cell line (STN-49) generated for this study, will be made available upon request and require a materials transfer agreement according to Stanford guidelines. Data and code availability * • Data supporting the findings of this study are available in the article and its supplementary files. Bulk RNA-seq data have been deposited in NCBI’s Gene Expression Omnibus (GEO) under accession number [414]GSE287972 . scRNA-seq data have been deposited in GEO and are accessible through the accession number [415]GSE288791. * • All original code for analyzing bulk and single-cell RNA-seq data is indicated in the [416]key resources table. * • Any additional information required to reanalyze the data reported in this paper is available from the [417]lead contact upon request. Acknowledgments