Introduction

Gliomas, the most common brain tumors in children and adults, are classified by the World Health Organization (WHO) as low- (grades 1 and 2) or high- (grades 3 and 4) grade tumors. HGG, like glioblastoma, predominate in the adult population and differ from LGGs by their rapid growth, microvascular proliferation, necrosis, and poor prognosis1. In contrast, LGGs, including pilocytic astrocytoma (PA; grade 1), account for the majority of pediatric brain tumors and are associated with good overall survival rates2. The neoplastic cells in these gliomas are embedded within a microenvironment rich in immune cells, including tumor-associated monocytes (macrophages, microglia) and lymphoid cells3. Within this stromal cell compartment, T cells (conventional CD4+ T cells, regulatory CD4+ T cells, and CD8+ cytotoxic T lymphocytes) constitute 0–5% of the total tumor cellular content4,5. Typically, the CD8+ T cells in these tumors are thought to function as “effector” (cytotoxic) T cells, which can recognize antigens expressed or shed by the neoplastic cells (“tumor neoantigens”). When activated, these effector T cells limit glioma growth by killing the tumor cells6,7. Considerable efforts have focused on harnessing their anti-tumoral capabilities for therapeutic purposes by reactivating them from an exhausted (non-reactive) state. Exhausted T cells are characterized by increased expression of cytotoxic T-lymphocyte-associated protein 4 (CTLA4), T cell immunoglobulin and mucin-domain containing-3 (TIM3), ectonucleoside triphosphate diphosphohydrolase 1 (CD39), T cell immunoreceptor with Ig and ITIM domains (TIGIT), lymphocyte activating 3 (LAG3), and programmed cell death protein-1 (PD1). The latter antigen serves as the target for many immune checkpoint therapies that release exhausted T cells from their non-reactive state and allows them to kill malignant cancer tumor cells8,9.

Unfortunately, ICI therapy has not proven efficacious for malignant brain tumors, like glioblastoma10. This outcome could reflect a relative paucity of canonical CD8+ exhausted T cells in these “ICI therapy non-responsive” tumors11. For this reason, we leveraged single cell RNA sequencing data from three independent sets of pilocytic astrocytoma (PA)12,13, as well as several adult and pediatric HGG datasets14,15,16, to reveal an enrichment of PD1+ and TIGIT+ CD8+ exhausted T cells in PAs relative to their malignant pediatric and adult HGG counterparts.

To explore the contribution of CD8+ T cells to LGG growth in an immunologically intact mouse system in vivo, we used the only currently available authenticated preclinical mouse models of pediatric LGG – those that recapitulate the genetics of the neurofibromatosis type 1 (NF1) cancer predisposition syndrome, in which children develop LGGs of the optic nerve and chiasm17,18. Similarly, Nf1-mutant mice develop optic nerve and chiasm LGGs (optic pathway gliomas; OPG) in a highly penetrant and reproducible manner by 12 weeks of age19. In these Nf1-OPG mice, CD8+ T cells20 are stimulated by midkine produced by Nf1-mutant neurons21. This results in the elaboration of Ccl4 by CD8+ T cells to induce tumor-associated monocyte (TAM) expression of Ccl5, a mitogen required for Nf1-OPG growth22.

Herein, we demonstrate that Ccl4-producing PD1+ and TIGIT+ CD8+ exhausted T cells are restricted to the tumors of Nf1-OPG mice. In these tumors, CD8+ exhausted T cell-mediated tumor growth is mediated by both Ccl4 TAM activation and a second TGFβ-dependent effect on tumor cell proliferation. In this regard, α-PD1 ICI treatment reduces TGFβ-mediated tumor cell growth, while combined α-PD1/α-TIGIT ICI therapy abrogates Ccl4-controlled TAM production of Ccl5. Similar to Ccl5 or TAM targeting, neutralizing α-TGFβ antibodies reduce Nf1-OPG growth in vivo. Additionally, we elucidate a second T cell chemoattraction circuit involving TAM production of Ccl12 and Cxcl13, which is abrogated by ICI treatment. Taken together, these findings establish unexpected roles for this specialized population of T lymphocytes in brain tumor biology relevant to immune targeted therapies.

Results

CD8+ exhausted T cells are enriched in human and mouse low-grade glioma relative to high-grade gliomas

Clinical trials using ICI therapy have shown efficacy for some malignant tumors, including melanomas, non-small-cell lung carcinomas (NSCLC), and breast cancer23,24. In contrast, this immune-targeted approach has not been successful for the treatment of glioblastoma9. Leveraging published single cell RNA-sequencing (scRNAseq) datasets from human pediatric LGG12,13 (pLGG, PA) (grade 1 glioma; pilocytic astrocytoma [PA], GSE222850 and dbGAP phs001854.v1.p1), pediatric HGG16 (pHGG) (grade 4 diffuse midline glioma [DMG], GSE102130), and adult HGG (aHGG) (grade 4 glioblastoma14,15 [GBM], Annotare [ArrayExpress] accession # E-MTAB-9435 and GSE138794), we found that PAs harbor greater T cell content (11.88% T cells, pediatric LGG; 0.18% T cells, adult HGG; 0.09% T cells, pediatric HGG) (Fig. 1A, B), increased CD8A gene expression (Fig. 1C, D), and higher expression of classic exhaustion markers25 (TIGIT, PDCD1, LAG3, and CTLA4; Fig. 1E), than HGGs. To study CD8+ exhausted T cells in human PAs, we performed scRNAseq of four sporadic and one NF1-associated tumors from the Pediatric Brain Tumor Repository at St. Louis Children’s Hospital. Unsupervised clustering identified multiple cell types, including endothelial cells, TAM, neurons, T cells, and tumor cells (Fig. 1F, G, Supplementary Fig. 1A, D). Within the CD8+ T cell cluster, a subset of expressed genes and pathways associated with T cell exhaustion (PDCD1, TIGIT, and LAG3; Fig. 1H–J, Supplementary Fig. 1B, C, E, F, O), regardless of NF1 disease status (Supplementary Fig. 1A–F). Moreover, these T cells also expressed cytokines and effector molecules typically associated with a canonical exhausted T cell phenotype26. This CD8+ exhausted T cell population was also found in two additional independently generated human PA datasets from Boston Children’s Hospital and KiTZ (Heidelberg University; Supplementary Fig. 1G–N).

Fig. 1: CD8+ exhausted T cells are present in human pilocytic astrocytomas.
figure 1

A UMAP visualization of T cells from human pLGG ([pilocytic astrocytoma, PA] PA set 1 and PA set 2 datasets; WHO grade 1 PA tumors) (n = 2 datasets, 11 tumors, GSE222850 and dbGAP phs001854.v1.p1), human pediatric HGG (pHGG [diffuse midline glioma, DMG] datasets; WHO grade 4 tumors) (n = 1 dataset, 5 tumors, GSE102130), and human adult HGG ([glioblastoma, GBM], aHGG1 and aHGG2 datasets; WHO grade 4 tumors) (n = 2 datasets, 7 tumors, Annotare [ArrayExpress] accession # E-MTAB-9435 and GSE138794) scRNAseq datasets. B Charts showing the percentage of T cells from human PA (PA set 1 and PA set 2 datasets; WHO grade 1 PA tumors) (n = 2 datasets, 11 tumors), human pediatric HGG (pHGG [diffuse midline glioma, DMG] datasets; WHO grade 4 tumors) (n = 1 dataset, 5 tumors), and human adult HGG ([glioblastoma, GBM], aHGG1 and aHGG2 datasets; WHO grade 4 tumors) (n = 2 datasets, 7 tumors) scRNAseq datasets. C CD8A expression in human PA (n = 2 datasets), pediatric HGG (n = 1 dataset) and adult HGG samples (n = 2 datasets). Each dot is an individual cell. D Table showing the P value, false discovery rate (FDR), and fold change of CD8A expression in human PA, pediatric and adult HGGs. Differential analyses were performed using gene specific analysis (GSA). E Dot plot showing the genes used to identify CD8+ exhausted T cells in human PA (n = 2) and HGG datasets (pediatric, n = 1; adult, n = 2). F UMAP visualization of scRNAseq data from five human PA specimens (PA1 [4463 cells], PA2 [4829 cells], PA3 [10404 cells], PA4 [7210 cells], PA5 [4636 cells]) based on tumor sample. G UMAP visualization of scRNAseq data from five human PA specimens (PA1, PA2, PA3, PA4, PA5) based on cell type. H UMAP visualization of scRNAseq data from five human PA specimens (PA1, PA2, PA3, PA4, PA5) color-coded by gene expression (CD8A, PDCD1, TIGIT, LAG3, TOX, CTLA4). I Dot plot showing the genes used to identify CD8+ exhausted T cells in human PA specimens. J Top 20 transcripts in CD8A+ PDCD1+ T cells relative to other T cells calculated with DESeq2 and filtered P values ≤ 0.05, false discovery rates ≤0.1, and log fold changes greater or equal to ±2. Differential analyses were performed using gene specific analysis (GSA).

To confirm the existence of CD8+ exhausted T cells in mouse LGGs, scRNAseq was performed on optic nerves from two independent cohorts of Nf1-OPG mice (Fig. 2A, Supplementary Fig. 2A). Similar to their human counterparts, we identified a CD8+ exhausted T cell population expressing Pdcd1, Tigit, Tox, and Lag3 in both cohorts, as well as cytokines, effector molecules, and pathways associated with exhaustion (Fig. 2B, C, Supplementary Fig. 2B, C, 2E, F). T cell receptor (TCR) sequencing revealed that the highest frequency clonotype corresponded to this CD8+ exhausted population (Fig. 2D, Supplementary Fig. 2D). Further analysis of the exhausted T cell populations in the mouse datasets revealed that the CD8+ exhausted T cell cluster comprised T cells expressing terminal exhaustion markers (terminally exhausted T cells, TEX; Pdcd1+ Havcr2+ Tcf7-), CD39/Entpd127, and progenitor exhaustion markers (progenitor exhausted T cells, PEX, Pdcd1+ Havcr2- Tcf7+) (Supplementary Fig. 2G)28. Taken together, these findings identify a unique population of CD8+ exhausted T cells in both human and mouse LGGs.

Fig. 2: CD8+ exhausted T cells are present in the optic nerve of Nf1-OPG mice.
figure 2

A t-SNE visualization of scRNAseq data of optic nerves collected from 12-week-old Nf1-OPG mice, based on cell type; n = 10 pooled mice. Microglia and monocytes constitute tumor associated monocytes (TAM). B t-SNE visualization of the T cell populations from 12-week-old Nf1-OPG mouse optic nerves, color-coded by T cell type. NOS, not otherwise specified. C Dot plot showing the genes used to identify CD8+ exhausted T cells in 12-week-old Nf1-OPG mouse optic nerves. D Stacked bar plot representing the barcode frequency of the top clones in the different T cell populations in 12-week-old Nf1-OPG mouse optic nerves. The graph is color coded to show the 1–10 clones and 11–10,000 clones.

CD8+ exhausted T cells are enriched in the optic nerves of tumor-bearing mice

Next, we sought to define the tissue compartments in which CD8+ exhausted T cells reside in Nf1-OPG mice (Fig. 3A). While CD8+ exhausted T cells were not detected in peripheral blood mononuclear cells (PBMCs) from Nf1-OPG mice by scRNAseq (Fig. 3B, C), a CD8+ exhausted-like population with low level (Tigit, Tox, Pdcd1) or no (Lag3, Cst7, Ctla4, Tnfrsf9, Cd27, Cd28) expression of exhaustion markers was detected in the dura (Fig. 3D, E). Following gating on live single cells, the PD1+ TIGIT+ CD8+ exhausted population was not found in WT or Nf1+/− mice (Supplementary Fig. 3A–E) or in the PBMCs and deep cervical lymph nodes of 12-week-old Nf1-OPG mice (Fig. 3F, I, L). Although a CD8+/PD1+ population was detected in both the dura and optic nerves of 12-week-old Nf1-OPG mice (29.65% CD8+/PD1+ T cells in the optic nerve; 16.1% CD8+/PD1+ T cells in the dura), a PD1/TIGIT double-positive CD8+ exhausted T cell population was mainly found in the optic nerve (7.87% CD8+/PD1+/TIGIT+ T cells in the optic nerve; 2.75% CD8+/PD1+/TIGIT+ T cells in the dura, 0.17% CD8+/PD1+/TIGIT+ T cells in the PBMCs, 0.03% CD8+/PD1+/TIGIT+ T cells in the dCLN) (Fig. 3G–H, 3J–L, Supplementary Fig. 3G–I). Importantly, CD8+ exhausted T cell populations expressing TEX and PEX markers were only found in the optic nerves of tumor-bearing mice, and not in the meningeal dura or PBMC scRNAseq datasets. CD39 expression was also restricted to the optic nerve TEX population and Entpd1 (CD39) was not present in either the meningeal dura or PBMCs (Supplementary Fig. 3F, 3I). Similar to mouse LGG, a PD1+/TIGIT+ CD8+ exhausted T cell population was also detected in human LGG (PA) tumors using flow cytometry (Fig. 3M, Supplementary Fig. 3J–K).

Fig. 3: PD1+ TIGIT+ CD8+ exhausted T cells are enriched in the tumors from Nf1-OPG mice.
figure 3

A Schematic representation of T cell circulation from the blood through the dural meninges (dura), optic nerve (ON), and draining deep cervical lymph nodes (dCLN). Created in BioRender. Chatterjee, J. (2023) BioRender.com/f03c341. B t-SNE visualization of scRNAseq data from 12-week-old Nf1-OPG mice peripheral blood mononuclear cells (PBMCs), based on cell type; n = 10 pooled mice. C t-SNE visualization of scRNAseq data from 12-week-old Nf1-OPG PBMCs, color-coded by gene expression (Cd8a, Pdcd1, Tigit, Lag3). D t-SNE visualization of the T cell populations from 12-week-old Nf1-OPG mouse meningeal dura, color-coded by T cell type, n = 10 pooled mice. E Dot plot showing the genes used to identify CD8+ exhausted T cells in the 12-week-old Nf1-OPG mouse meningeal dura scRNAseq dataset. Representative gating strategy for flow cytometry of 12-week-old PMBC T cells (n = 9 mice) (F), meningeal dura (n = 9 mice) (G), optic nerve (n = 4, 3 pooled optic nerves per sample) (H), and deep cervical lymph nodes dura (n = 9 mice) (I). J Dot plot showing the genes used to identify CD8+ exhausted T cells in 12-week-old Nf1-OPG mouse optic nerves (corresponds to the dataset shown in Fig. 2; n = 10 pooled mice), meningeal dura (corresponds to the dataset shown in (D); n = 10 pooled mice), and PBMC (corresponds to the dataset shown in (B); n = 10 pooled mice) scRNAseq datasets. K Graph representing the percentages of CD8+/PD1+ and CD8+/TIGIT+ cells in the optic nerves (ON) and dura of 12-week-old Nf1-OPG mice (ON, n = 4, 3 pooled optic nerves per sample; dura, n = 9 mice). Data are represented as mean ± SD. A one-way ANOVA test was performed followed by a Dunnett’s post-test correction. Exact P values are indicated. L Graph comparing the percentages of CD8+/PD1+/TIGIT+ cells in the PBMCs, dura, optic nerve (ON), and dCLN, of 12-week-old Nf1-OPG mice (PBMCs, n = 8 mice; dura, n = 9 mice; ON, n = 4, 3 pooled optic nerves per sample; dCLN, n = 8 mice). Data are shown as mean ± SD. A one-way ANOVA test was performed followed by a Holm-Šídák post-test correction. Exact P values are included. M Representative gating strategy for flow cytometry of one representative human PA1 (n = 1; corresponds to PA1 sample in Fig. 1F).

CD8+ exhausted T cells express Ccl4

Relevant to the role of T cells in Nf1-OPG tumors, we previously demonstrated that CD8+ T cells produce Ccl4 to induce TAM expression of Ccl5, a mitogen required for tumor cell growth (Fig. 4A)22,29, such that CD8+ T cell antibody-mediated depletion abrogates Nf1-OPG growth in vivo22. To establish the importance of this discovered CD8+ exhausted T cell population to Nf1-OPG growth, we performed a series of complementary experiments: First, by RT-qPCR, we showed that Ccl4 was highly expressed in the optic nerves (tumor site) of 12-week-old Nf1-OPG mice relative to non-tumor-containing brain regions (cerebellum, brainstem, cortex, hippocampus) or tissues where T cells circulate (dura, deep cervical lymph nodes, superficial cervical lymph nodes) (Fig. 4B). Second, using scRNAseq, we demonstrated that Ccl4 expression was enriched in CD8+ exhausted T cells from the optic nerves relative to other tissues (Fig. 4C), other optic nerve T cell populations (fold change = 2.67, P = 2.63E–6, Fig. 4D; fold change = 5.90, P = 3.52E–18, Supplementary Fig. 4A; fold change = 1.81, P = 6.71E–15, Supplementary Fig. 4B), CD4+ Treg cells (fold change = 4.01, P = 2.06E–3, Supplementary Fig. 4C; fold change = 7.31, P = 4.09E–15, Supplementary Fig. 4D), PBMC T cells (fold change = 8.40, P = 5.95E–31, Fig. 4E), and dural T cells (fold change = 2.52, P = 3.8E–31, Fig. 4F). Additionally, the Ccl4-expressing T cells in the optic nerves of Nf1-OPG mice show an enrichment in genes and pathways related to cytokine activity and chemokine binding (Supplementary Fig. 4G, H). While TAM in the optic nerve also express Ccl4, monocyte Ccl4 expression in non-tumor bearing (Nf1+/− and WT) is similar to that seen in tumor bearing (Nf1-OPG) mice (Supplementary Fig. 4E, F). Moreover, Ccl4 and Entpd1 (CD39) expression were also enriched in the TEX compared to non-TEX (Ccl4 fold change = 151.27, P = 3.25E–9; Entpd1 (CD39) fold change = 3.86, P = 5.56E–17, Fig. 4G) and PEX T cell populations within Nf1-OPG mouse optic nerves (Ccl4 fold change = 26.74, P = 5.97E–6; Entpd1 (CD39) fold change = 7.41, P = 1.99E–11, Supplementary Fig. 4I).

Fig. 4: CD8+ exhausted T cells express Ccl4.
figure 4

A Schematic representation of the optic glioma immune circuit. Created in BioRender. Chatterjee, J. (2024) BioRender.com/e28x983. B Ccl4 expression in the dura, optic nerve (ON), cerebellum (CBL), brainstem (BSTM), cortex (CTX), hippocampus (HC), deep cervical lymph nodes (dCLN), and superficial cervical lymph nodes (CLN) of 12-week-old Nf1-OPG mice, following normalization to the optic nerve. Data are represented as mean ± SD. (dura, n = 6 mice; ON, n = 3, 2 pooled optic nerves per sample; CBL, n = 6 mice; BSTM, n = 5 mice; CTX, n = 5 mice; HC, n = 6 mice; dCLN, n = 6 mice; CLN, n = 6 mice). A one-way ANOVA test was performed followed by a Holm-Šídák post-test correction. Exact P values are included. C Dot plot showing Ccl4 expression from 12-week-old Nf1-OPG mouse optic nerves (corresponds to the dataset shown in Fig. 2; n = 10 pooled mice), meningeal dura (corresponds to the dataset shown in Fig. 3D; n = 10 pooled mice), and PBMC (corresponds to the dataset shown in Fig. 3B; n = 10 pooled mice) scRNAseq datasets. D Volcano plot showing fold change and P value comparing CD8+ exhausted T cells and the other T cell populations in the optic nerves of 12-week-old Nf1-OPG mice (corresponds to the dataset shown in Fig. 2; n = 10 pooled mice). Upregulated genes in red, downregulated genes in blue. Differential analyses were performed using gene specific analysis (GSA). E Volcano plot showing fold change and P value comparing CD8+ exhausted T cells in the optic nerves (corresponds to the dataset shown in Fig. 2; n = 10 pooled mice) and PMBC T cells of 12-week-old Nf1-OPG mice (corresponds to the dataset shown in Fig. 3B; n = 10 pooled mice). Upregulated genes in red, downregulated genes in blue. Differential analyses were performed using gene specific analysis (GSA). F Volcano plot showing fold change and P value comparing CD8+ exhausted T cells in the optic nerves (corresponds to the dataset shown in Fig. 2, Supplementary Fig. 2; n = 10 pooled mice in each dataset) and T cells from the dura of 12-week-old Nf1-OPG mice (corresponds to the dataset shown in Fig. 3D; n = 10 pooled mice). Upregulated genes in red, downregulated genes in blue. Differential analyses were performed using gene specific analysis (GSA). G Volcano plot showing fold change and P value comparing CD8+ TEX in the optic nerves (corresponds to the dataset shown in Fig. 2; n = 10 pooled mice, and to the dataset shown in Supplementary Fig. 2; n = 10 pooled mice) and CD8+ non-TEX T cells of 12-week-old Nf1-OPG mice (corresponds to the dataset shown in Fig. 2; n = 10 pooled mice, and to the dataset shown in Supplementary Fig. 2; n = 10 pooled mice). Upregulated genes in red, downregulated genes in blue. Differential analyses were performed using gene specific analysis (GSA). H Graph showing levels of secreted Ccl4 in the TCM by ELISA of unstimulated and CD8+ exhausted T cells (α-CD3+α-CD28 antibody stimulated) with and without FK506 treatment (none, n = 4; α-CD3+α-CD28, n = 4; α-CD3+α-CD28 + FK506, n = 4). A one-way ANOVA test was performed followed by a Holm-Šídák post-test correction. Data are represented as mean ± SD. Exact P values are indicated.

Third, since we previously showed that CD8+ T cell Ccl4 expression is dependent on NFAT activity22, we stimulated CD8+ T cells in vitro with α-CD3+α-CD28 antibodies to generate CD8+ “exhausted” T cells (exhausted). Following exhaustion, CD8+ T cells express more Ccl4 relative to unstimulated CD8+ T cells (non-exhausted) (fold change = 8.8, P = < 0.0001, Fig. 4H). Moreover, following treatment with the FK506 calcineurin inhibitor, Ccl4 production was reduced (79% decrease, P = < 0.0001, Fig. 4H), confirming that exhaustion-mediated production of Ccl4 is NFAT1-dependent. In addition, Nfatc2 expression was enriched in Ccl4-expressing TEX compared to CD8+ non-TEX (fold change = 24.13, P = 1.25E–11, Fig. 4G) or PEX (fold change = 28.89, P = 8.73E–8, Supplementary Fig. 4J) T cells.

Fourth, scRNA-seq analysis revealed that CD8+ T cell exhaustion increases during Nf1-OPG tumor evolution. No CD8+ exhausted T cell cluster was identified at 6 weeks of age at a stage before obvious tumors are found (Fig. 5A, B), but was detected in mice with optic gliomas at both 12 (Fig. 2A–D; Supplementary Fig. 2A–D) and 24 weeks (Fig. 5C, D) of age (Fig. 5E), paralleling changes in tumor proliferation (%Ki67+ cells) and Ccl4/Ccl5 expression (Fig. 5F, H–K) during tumor evolution. However, there was no difference in CD8+ T cell numbers or the percentage of CD8+ exhausted T cells between 12- and 24-week-old tumor-bearing mice (Fig. 5F, G).

Fig. 5: Presence of CD8+ exhausted T cells mirrors optic glioma growth and Ccl4/Ccl5 expression.
figure 5

A t-SNE visualization of scRNAseq data from optic nerves of 6-week-old Nf1-OPG mice, based on cell type, n = 10 pooled mice. B t-SNE visualization of the T cell populations from 6-week-old Nf1-OPG mice, color-coded by T cell type. Created in BioRender. Chatterjee, J. (2023) BioRender.com/p15a720. C t-SNE visualization of scRNAseq data from optic nerves of 24-week-old Nf1-OPG mice, based on cell type, n = 10 pooled mice. Microglia and monocytes constitute tumor associated monocytes (TAM). D t-SNE visualization of the T cell populations from 24-week-old Nf1-OPG mice, color-coded by T cell type. Created in BioRender. Chatterjee, J. (2023) BioRender.com/p15a720. E Dot plot showing genes used to identify CD8+ exhausted T cells in 6- (corresponds to the dataset shown in Fig. 5A; n = 10 pooled mice), 12- (corresponds to the datasets shown in Fig. 2 and Supplementary Fig. 2; n = 10 pooled mice in each dataset), and 24-week-old Nf1-OPG mouse optic nerves (corresponds to the dataset shown in (C); n = 10 pooled mice). F Graph showing changes in the percentage of CD8+ exhausted T cells (blue) in 6-week-old (corresponds to the dataset shown in (A); n = 10 pooled mice), 12-week-old (corresponds to the datasets shown in Fig. 2 and Supplementary Fig. 2; n = 10 pooled mice in each dataset), and 24-week-old (corresponds to the dataset shown in (C); n = 10 pooled mice) Nf1-OPG mice and proliferating (Ki67+ cells; red) tumor cells over time (6 weeks, n = 5 mice; 12 weeks, n = 4 mice; 24 weeks, n = 4 mice). Data are represented as mean ± SD. G Quantification CD8+ T cells in the optic nerves of Nf1-OPG mice at 12 and 24 weeks of age (12 weeks, n = 5 mice; 24 weeks, n = 5 mice). Data are represented as mean ± SD. To evaluate statistical differences, a two-tailed non-parametric Mann–Whitney test was performed. Exact P values are indicated. ns, not significant. H Ccl4 RNA expression in the optic nerves of 6-week-old, 12-week-old, and 24-week-old Nf1-OPG mice. Data are represented relative to the 12-week-old group (6-week-old mice, n = 6, 2 pooled optic nerves per sample; 12-week-old mice, n = 4, 2 pooled optic nerves per sample; 24-week-old mice, n = 4, 2 pooled optic nerves per sample). Data are represented as mean ± SD. A one-way ANOVA test was performed followed by a Dunnett’s post-test correction. Exact P values are indicated. ns, not significant. I Dot plot showing Ccl4 expression in T cells from optic nerves of 6-week-old (6 W; corresponds to the dataset shown in (A); n = 10 pooled mice), 12-week-old (12 W) (corresponds to the datasets shown in Fig. 2 and Supplementary Fig. 2; n = 10 pooled mice in each dataset), and 24-week-old (24 W) (corresponds to the dataset shown in (C); n = 10 pooled mice) Nf1-OPG mice. J Ccl5 RNA expression in the optic nerves of 6-week-old, 12-week-old, and 24-week-old Nf1-OPG mice. Data are represented relative to the 12-week-old group (6-week-old mice, n = 3, 2 pooled optic nerves per sample; 12-week-old mice, n = 5, 2 pooled optic nerves per sample; 24-week-old mice, n = 4, 2 pooled optic nerves per sample). Data are represented as mean ± SD. A one-way ANOVA test was performed followed by a Dunnett’s post-test correction. Exact P values are indicated. ns, not significant. K Dot plot showing Ccl5 expression in TAM from the optic nerves of 6-week-old (6 W; corresponds to the dataset shown in (A); n = 10 pooled mice), 12-week-old (12 W) (corresponds to the datasets shown in Fig. 2 and Supplementary Fig. 2; n = 10 pooled mice in each dataset), and 24-week-old (24 W) (corresponds to the dataset shown in (C); n = 10 pooled mice) Nf1-OPG mice.

Fifth, we analyzed two additional Nf1-mutant mouse strains, one that also forms optic gliomas by 12 weeks of age (Nf1Arg681XC)30,31 and one that does not develop tumors (Nf1Arg1809CC)21. Consistent with a key role for CD8+ exhausted T cells in Nf1-OPG pathogenesis, PD1+/TIGIT+ CD8+ T cells were only detected by flow cytometry in Nf1Arg681XC, but not Nf1Arg1809CC, mice (Supplementary Fig. 5A–C). Taken together, the presence of PD1+/TIGIT+/CD39+ CD8+ exhausted T cells exclusively in the optic nerves of tumor-bearing mice, their absence in non-tumor-bearing mice, and their NFAT1-dependent Ccl4 expression establish CD8+ exhausted T cells expressing terminal exhaustion markers as the most likely lymphocyte population within the immune axis that regulates Nf1-OPG growth.

Anti-PD1+/TIGIT+ ICI treatment reduces CD8+ T cell infiltration by decreasing TAM-mediated Ccl12 and Cxcl13 chemoattraction

To determine whether CD8+ exhausted T cells are necessary for Nf1-OPG formation, 12-week-old Nf1-OPG mice with established tumors were treated with α-PD1 or α-TIGIT antibodies or both (200 µg/dose, twice per week) (Fig. 6A). All treatments reduced CD8+ T cell content in the tumors (Fig. 6B), and the number of circulating CD8+ T cells in the dura (Supplementary Fig. 6A). Previous studies from our group demonstrated that Ccl2, produced by Nf1-OPG tumor cells, can attract T cells into the tumor20,22,32. However, Ccl2 was only attenuated following combined α-PD1 and α-TIGIT antibody treatment (75% decrease, P = 0.0240) (Fig. 6C), which does not explain the reduction of CD8+ T cells in the optic nerves of Nf1-OPG mice treated with either α-PD1 or α-TIGIT antibodies.

Fig. 6: Anti-PD1+/TIGIT+ ICI treatment reduces CD8+ T cell infiltration by decreasing TAM-mediated Ccl12 and Cxcl13 chemoattraction.
figure 6

A Schematic representation of the anti-PD1 (α-PD1), anti-TIGIT (α-TIGIT), and combined (α-PD1 + TIGIT) antibody treatments. Nf1-OPG mice were treated (200 µg/dose/ i.p., twice per week) from 12 to 16 weeks of age, and tissues were analyzed at 16 weeks. The control group was injected with anti-IgG isotype control antibodies. Created in BioRender. Chatterjee, J. (2024) BioRender.com/r98n914. B Immunohistochemistry and quantification CD8+ T cells in the entire optic nerve (IgG, n = 10 mice; α-TIGIT, n = 10 mice; α-PD1, n = 6 mice; α-PD1 + TIGIT, n = 5 mice). Data are represented as mean ± SD. A one-way ANOVA test was performed followed by a Dunnett’s post-test correction. Exact P values are indicated (α-TIGIT, P = 0.0054; α-PD1, P = 0.0003; α-PD1 + TIGIT, P = 0.0032). Scale bar, 200 µm. C Ccl2 RNA expression in the optic nerves of Nf1-OPG mice treated with IgG, α-TIGIT, α-PD1 or a combination of α-PD1 and α-TIGIT antibodies. Data are represented relative to the IgG control group (IgG, n = 5, 2 pooled optic nerves per sample; α-TIGIT n = 5, 2 pooled optic nerves per sample; α-PD1, n = 4, 2 pooled optic nerves per sample; α-PD1 + TIGIT, n = 4, 2 pooled optic nerves per sample). Data are represented as mean ± SD. A one-way ANOVA test was performed followed by a Dunnett’s post-test correction. Exact P values are indicated. D Volcano plot showing fold change and P value comparing Nf1-OPG TAM to Nf1+/ and WT monocytes in the optic nerves of 12-week-old mice. Upregulated genes in red, downregulated genes in blue. Differential analyses were performed using gene specific analysis (GSA). E Ccr2, Cxcr3, and Cxcr5 expression in T cell populations from 12-week-old Nf1-OPG mouse optic nerves, color-coded by T cell type. F Cxcl9, Ccl12, and Cxcl13 RNA expression in the optic nerves of 12-week-old WT and Nf1-OPG mice. Data are represented relative to the WT group (Cxcl9; WT n = 3, 2 pooled optic nerves per sample; Nf1-OPG n = 4, 2 pooled optic nerves per sample; Ccl12; WT n = 4, 2 pooled optic nerves per sample; Nf1-OPG n = 4, 2 pooled optic nerves per sample; Cxcl13; WT n = 4, 2 pooled optic nerves per sample; Nf1-OPG n = 4, 2 pooled optic nerves per sample). Data are represented as mean ± SD. To evaluate statistical differences, a two-tailed non-parametric Mann–Whitney test was performed. Exact P values are indicated. ns, not significant. G Graph showing the percentage of migrated Nf1+/- CD8+ T cells treated with medium (Control) (n = 5), Ccl12 (n = 6), or Cxcl13 (n = 5). Data are represented as mean ± SD. A one-way ANOVA test was performed followed by a Dunnett’s post-test correction. Exact P values are indicated. Created in BioRender. Chatterjee, J. (2024) BioRender.com/r98n914. H Ccl12 RNA expression in the optic nerves of Nf1-OPG mice treated with IgG, α-TIGIT, α-PD1 or a combination of α-PD1 and α-TIGIT antibodies. Data are represented relative to the IgG control group (IgG, n = 5, 2 pooled optic nerves per sample; α-TIGIT n = 5, 2 pooled optic nerves per sample; α-PD1, n = 4, 2 pooled optic nerves per sample; α-PD1 + TIGIT, n = 3, 2 pooled optic nerves per sample). Data are represented as mean ± SD. A one-way ANOVA test was performed followed by a Tukey’s post-test correction. Exact P values are indicated (α-TIGIT, P = 0.0012; α-PD1, P = 0.0187; α-PD1 + TIGIT, P = 0.0013). I Cxcl13 RNA expression in the optic nerves of Nf1-OPG mice treated with IgG, α-TIGIT, α-PD1 or a combination of α-PD1 and α-TIGIT antibodies. Data are represented relative to the IgG control group (IgG, n = 4, 2 pooled optic nerves per sample; α-TIGIT n = 4, 2 pooled optic nerves per sample; α-PD1, n = 4, 2 pooled optic nerves per sample; α-PD1 + TIGIT, n = 4, 2 pooled optic nerves per sample). Data are represented as mean ± SD. A one-way ANOVA test was performed followed by a Tukey’s post test correction. Exact P values are indicated (α-TIGIT, P = 0.0003; α-PD1, P = 0.0007; α-PD1 + TIGIT, P = 0.0001).

To elucidate the mechanism responsible for the reduction in CD8+ T cell numbers, we first leveraged data from scRNAseq data from 12-week-old Nf1-OPG, Nf1+/−, and WT mouse optic nerves to identify potential chemokines that might attract T cells into the tumor. Using this approach, there were no additional chemoattractants, besides Ccl2, expressed by Nf1-OPG tumor cells. However, we identified three potential chemokines expressed by TAM in the optic nerves of Nf1-OPG mice relative to Nf1+/− and WT optic nerve monocytes (Ccl12, Cxcl9, and Cxcl13) (Fig. 6D). Importantly, the receptors for each of these chemokines/cytokines (Ccl12:Ccr2, Cxcl9:Cxcr3, Cxcl13:Cxcr5) were expressed in CD8+ exhausted T cells (Fig. 6E). Second, we performed RT-qPCR analyses using a separate cohort of Nf1-OPG and WT optic nerve samples to demonstrate that only Ccl12 and Cxcl13, but not Cxcl9, expression was increased in Nf1-OPG relative to WT optic nerves (Fig. 6F). Third, using a modified Boyden chamber migration assay, we found that Ccl12 and Cxcl13 increased CD8+ T cell motility (Fig. 6G). Fourth, to assess Ccl12 and Cxcl13 expression in the optic nerves of Nf1-OPG mice treated with α-PD1, α-TIGIT and α-PD1 + TIGIT antibody therapies, we performed RT-qPCR and confirmed decreased Ccl12 (72% decrease, α-TIGIT; 53% decrease, α-PD1; 82% decrease, α-PD1 + TIGIT) and Cxcl13 (63% decrease, α-TIGIT; 65% decrease, α-PD1; 79% decrease, α-PD1 + TIGIT) expression relative to IgG-treated controls (Fig. 6H–I). Taken together, these results establish that CD8+ T cells are attracted by both tumor cells (Ccl2) and TAM (Ccl12 and Cxcl13).

Anti-PD1+/TIGIT+ ICI treatment reduces tumor cell proliferation through two distinct mechanisms

To determine whether Ccl4-expressing CD8+ exhausted T cells support Nf1-OPG growth, we assessed the percentage of proliferating cells in the optic nerves of mice treated with α-PD1 or α-TIGIT antibodies or both. Ki67 was used as a marker of tumor cell proliferation, as the proliferating cells are glioma cells33, rather than CD3+ T cells or Tmem119+ TAM as previously shown32 or endothelial cells (Supplementary Fig. 6B, C). While α-TIGIT antibody treatment did not reduce Nf1-OPG proliferation, α-PD1 or combined α-PD1 and α-TIGIT antibody treatment decreased tumor proliferation (%Ki67+ cells) (Fig. 7A). As observed in other preclinical experiments32,34, optic nerve volumes calculated using Eq.(1) and TAM content (%Iba1+ cells) in the optic nerve were not affected by any of the treatments (Supplementary Fig. 6D, E). In this regard, optic nerve volumes and TAM content are similar in Nf1+/− and Nf1-OPG mice, where they reflect the impact of a heterozygous Nf1 mutation, rather than features of an optic glioma in mice35.

Fig. 7: Anti-PD1+/TIGIT+ ICI treatment reduces tumor cell proliferation through two distinct mechanisms.
figure 7

A Immunohistochemistry and quantification of proliferating (%Ki67+) cells in the optic nerves of Nf1-OPG mice (IgG, n = 9 mice; α-TIGIT, n = 10 mice; α-PD1, n = 9 mice; α-PD1 + TIGIT, n = 6 mice). Data are represented as mean ± SD. A one-way ANOVA test was performed followed by a Dunnett’s post-test correction. Exact P values are indicated (α-PD1, P < 0.0001; α-PD1 + TIGIT, P = 0.001). Scale bar, 200 µm. B, C Ccl4 and Ccl5 RNA expression in the optic nerves of Nf1-OPG mice treated with IgG, α-TIGIT, α-PD1 or a combination of α-PD1 and α-TIGIT antibodies. Data are represented relative to the IgG control group (IgG, n = 5, 2 pooled optic nerves per sample; α-TIGIT n = 5, 2 pooled optic nerves per sample; α-PD1, n = 4, 2 pooled optic nerves per sample; α-PD1 + TIGIT, n = 4, 2 pooled optic nerves per sample). Data are represented as mean ± SD. A one-way ANOVA test was performed followed by a Dunnett’s post test correction. Exact P values are indicated. D Immunocytochemistry and quantification of proliferating (%Ki67+) optic glioma tumor cells (o-GTCs) treated with TCM from unstimulated (none) and exhausted (α-CD3+α-CD28 antibody stimulated) CD8+ T cells (Control, n = 8; TCM none, n = 4; TCM α-CD3+α-CD28, n = 4). Data are represented as mean ± SD. A one-way ANOVA test was performed followed by a Dunnett’s post-test correction. Exact P values are indicated. Scale bar, 200 µm. E Volcano plot showing fold change and P value comparing CD8+ PD1+ T cells (corresponds to the dataset shown in Fig. 2; n = 10 pooled mice) and in CD8+ PD1- T cells (corresponds to the dataset shown in Fig. 2; n = 10 pooled mice) the optic nerve of 12-week-old Nf1-OPG mice. Upregulated genes in red, downregulated genes in blue. Differential analyses were performed using gene specific analysis (GSA). F Graph showing levels of secreted TGFβ in the TCM of unstimulated (none) and exhausted (α-CD3+α-CD28 antibody stimulated) CD8+ T cells by ELISA (none, n = 4; α-CD3+α-CD28, n = 4). Data are represented as mean ± SD. To evaluate statistical differences, a two-tailed non-parametric Mann–Whitney test was performed. Exact P values are indicated. G Tgfb1 RNA expression in the optic nerves of Nf1-OPG mice treated with IgG, α-TIGIT, α-PD1 or a combination of α-PD1 and α-TIGIT antibodies. Data are represented relative to the IgG control group (IgG, n = 5, 2 pooled optic nerves per sample; α-TIGIT n = 5, 2 pooled optic nerves per sample; α-PD1, n = 4, 2 pooled optic nerves per sample; α-PD1 + TIGIT, n = 4, 2 pooled optic nerves per sample). Data are represented as mean ± SD. A one-way ANOVA test was performed followed by a Holm-Šídák post-test correction. Exact P values are indicated. H Schematic representation of the anti-TGFβ treatment (α-TGFβ). Nf1-OPG mice were treated (200 µg/dose, i.p., twice per week) from 12 to 16 weeks of age, tissues were analyzed at 16 weeks. The control group was injected with anti-IgG isotype control antibodies. Created in BioRender. Chatterjee, J. (2024) BioRender.com/z17k582. I Immunohistochemistry and quantification of proliferating (%Ki67+) cells in the optic nerves of Nf1-OPG mice (IgG, n = 5 mice; α-TGFβ, n = 4 mice). Data are represented as mean ± SD. To evaluate statistical differences, a two-tailed non-parametric Mann–Whitney test was performed. Exact P values are indicated. Scale bar, 200 µm. J Immunohistochemistry and quantification of CD8+ T cells in the entire optic nerve of Nf1-OPG mice (IgG, n = 5 mice; α-TGFβ, n = 4 mice). Data are represented as mean ± SD. To evaluate statistical differences, a two-tailed non-parametric Mann–Whitney test was performed. Exact P values are indicated. Scale bar, 200 µm. K Schematic representation of the direct and indirect mechanisms by which CD8+ exhausted T cells regulate Nf1-OPG growth. Created in BioRender. Chatterjee, J. (2024) BioRender.com/z17k582.

Since children with NF1-OPG experience vision loss as a result of tumor-induced retinal ganglion cell (RGC) content and axon thickness36,37, we assessed the number of retinal ganglion cells (RGCs) and the retinal nerve fiber layer (RNFL) thickness in Nf1-OPG mice following ICI treatment, as previously reported37. While there was no improvement in RGC content, the RNFL thickness was restored to wild type levels following combined α-PD1 + TIGIT antibody treatment (Supplementary Fig. 6F).

Since CTLA4 is expressed at higher levels than TIGIT in human PA CD8+ exhausted T cells, we also treated Nf1-OPG mice with α-CTLA4 antibodies (Fig. 1E, I). Following α-CTLA4 antibody treatment (Supplementary Fig. 7A), we observed a 90% decrease in tumor proliferation, as well as a reduction in the number of CD8+ T cells and optic nerve volume (Supplementary Fig. 7B-D). In contrast, the percentage of Iba1+ cells was not changed (Supplementary Fig. 7E).

As the CD8+ exhausted T cells express Ccl4, which is necessary for immune support of Nf1-OPG growth, we assayed Ccl4 mRNA levels by RT-qPCR in the optic nerve. Reduced Ccl4 expression was only observed after combination antibody therapy (75% decrease, P = 0.0077) (Fig. 7B). Since T cell Ccl4 induces TAM expression of Ccl5 in Nf1-OPG mice, we measured Ccl5 levels. Similar to Ccl4, Ccl5 expression was only reduced following combination α-PD1/α-TIGIT antibody therapy (80% decrease, P = 0.0179) (Fig. 7C). These results demonstrate that combination α-PD1/α-TIGIT treatment reduces tumor cell proliferation by abrogating Ccl4/Ccl5 stromal support but does not explain the tumor suppressive effect of α-PD1 antibody therapy alone.

To identify this PD1-mediated Ccl4-independent CD8+ T cell-dependent mechanism, we hypothesized that CD8+ T cells might produce a growth factor that directly regulates tumor cell growth. First, using CD8+ T cell conditioned medium (TCM) from exhausted CD8+ T cells (α-CD3 and α-CD28 antibody stimulated), we found that TCM from these CD8+ exhausted T cells (α-CD3 and α-CD28 antibody treated) increased optic glioma tumor cell (o-GTC) proliferation (%Ki67+ cells) by two-fold relative to non-exhausted (unstimulated) CD8+ TCM in vitro (Fig. 7D). Second, we leveraged our Nf1-OPG optic glioma scRNAseq data to identify a potential soluble factor enriched in CD8+ PD1+ T cells relative to CD8+ PD1neg T cells. We found that Tgfb1 expression was increased (fold change = 2.03, P = 3.89E–9) in CD8+ PD1+ T cells relative to other CD8+ PD1neg T cell populations (Fig. 7E). scRNAseq of optic nerves from 6, 12, and 24-week-old Nf1-OPG mice (Supplementary Fig. 8A) revealed higher expression levels of Tgfb1 at 12 and 24 weeks of age relative to 6 weeks of age, coinciding with the infiltration of CD8+ T cells in these tumors. Third, we demonstrated that TGFβ expression is increased in stimulated CD8+ T cells (α-CD3+α-CD28 antibody treatment) compared to unstimulated CD8+ T cells using RT-qPCR (fold change = 2.3, P = 0.0286) and ELISA (fold change = 4.9, P = 0.0286) (Supplementary Fig. 8B, Fig. 7F). Fourth, the addition of TGFβ at the concentration detected in the TCM of CD8+ T cells following α-CD3+α-CD28 antibody treatment in vitro increased optic glioma tumor cell (o-GTC) proliferation (fold change = 1.62, P = 0.0043, Supplementary Fig. 8C). Fifth, Tgfb1 expression was only reduced (57% reduction) in Nf1-OPG mice treated with α-PD1, but not α-TIGIT, antibodies alone (Fig. 7G). Sixth, to confirm that TGFβ is a stromal mitogen critical for Nf1-OPG growth, Nf1-OPG mice were treated with α-TGFβ neutralizing antibodies from 12 to 16 weeks of age (Fig. 7H). Following α-TGFβ antibody treatment in vivo, optic glioma proliferation (Ki67+ cells) and CD8+ T cell numbers were reduced and the RNFL was increased to wild type levels (Fig. 7I, J, Supplementary Fig. 8D–F). As before, no changes were observed in optic nerve volume, TAM content, or RGC number (Supplementary Fig. 8D-E). Taken together, these data reveal a CD8+ PD1+ T cell-TGFβ-dependent mechanism additionally important for governing Nf1-OPG tumor growth (Fig. 7K).

In contrast to previous studies in which ICI therapy results in increased CD8+ T cell-mediated tumor cell death (killing)38,39, we observed no increase in tumor apoptosis (Supplementary Fig. 9A), ferroptosis (Supplementary Fig. 9B), or expression of genes associated with T cell-mediated cytotoxicity (Supplementary Fig. 9C). Additionally, there was also no increase in o-GTC death following the addition of TCM from exhausted CD8+ T cells (stimulated with α-CD3 and α-CD28 antibodies) in vitro (Supplementary Fig. 9D).

Discussion

While lymphoid cells constitute a minor population of non-neoplastic cells within the brain tumor microenvironment, studies over the past decade have revealed important contributions for T cells in tumor maintenance and progression. Combining several converging and complementary strategies, we identify a specialized population of CD8+ exhausted T cells enriched in human LGGs relative to their malignant counterparts, which, in experimental LGG mouse models, is critical for stromal support of LGG growth. These observations raise several important points relevant to T cell control of brain tumor biology.

First, while there are numerous reports of CD8+ exhausted T cells in malignant tumors, few studies have focused on low-grade tumors40. While CD8+ T cells are found in benign schwannomas, PD1+TIM3+ CD8+ exhausted T cells were enriched only in malignant schwannomas41. Similarly, high-grade pancreatic cancers are highly infiltrated by CD8+ exhausted T cells, whereas their low-grade counterparts harbor a high ratio of activated CD8+ effector T cells42. In contrast, we demonstrate that CD8+ exhausted T cells are enriched in pediatric LGGs relative to both pediatric and adult HGGs. It should be noted that, unlike HGGs, PAs in children are typically chronic disorders with limited mortality and tend to behave in an indolent manner43. To this end, the majority of NF1-associated PAs (OPGs) grow during early childhood, with rare examples of spontaneous tumor regression reported44. For these reasons, we postulate that this unique population of CD8+ exhausted T cells functions in a unique manner in the setting of childhood LGG.

Second, the CD8+ exhausted T cells do not appear to function as cytotoxic effectors or inert bystanders, as described for some malignant cancers25,45. In this regard, α-PD1, α-TIGIT, or combined α-PD1/α-TIGIT ICI therapy did not result in tumor cell death by apoptosis or ferroptosis, nor was there increased expression of effector CD8+ T cell cytotoxic genes typically induced following successful PD1 treatment of malignant cancers46,47,48. Additionally, the PD1+/TIGIT+/CD39+ population of CD8+ T cells was restricted to the tumor in Nf1-OPG mice, where specific cellular interactions allow for their enrichment and/or expansion. In this regard, there was a higher expression of PD1 (CD274) and TIGIT (Pvr and Nectin2) ligands in the optic nerve relative to the meningeal dura by scRNAseq and RT-qPCR (Supplementary Fig. 10A, B).

Third, to exclude the possibility that the anti-tumor effects observed following α-PD1 or α-PD1/TIGIT antibody treatments reflected non-specific targeting of multiple cell populations, we examined expression of classical exhaustion genes in the scRNAseq datasets (Supplementary Fig. 6G, H). We found that TAM have low Pdcd1 (PD1) and no Tigit expression, NK cells express Tigit, but not Pdcd1, and CD4+ Tregs express both Pdcd1 and Tigit. While all three of these stromal cell types have been shown to regulate tumor growth49,50,51 in the context of Nf1-OPG tumors, neither TAM (Supplementary Fig. 4F), NK cells (Supplementary Fig. 4K) or CD4+ Tregs (Supplementary Fig. 4C, D) were responsible for observed increase in Ccl4 expression. In addition, antibody-mediated NK cell depletion had no effect on Nf1-OPG proliferation (Supplementary Fig. 11A–C). For these reasons, the tumor-suppressing effect of α-PD1/TIGIT antibody treatment is most likely due to the targeting of CD8+ exhausted T cells.

Fourth, in addition to Ccl4-mediated TAM Ccl5 production, we elucidated a second CD8+ exhausted T cell-mediated Nf1-OPG growth regulatory mechanism. While prior reports have shown that TGFβ can induce tumor cell proliferation and migration in other cancers52,53, this growth factor can also create an immunosuppressive environment causing CD8+ T cell anergy and suppression of cytotoxic function54. In contrast, we demonstrate, that the CD8+ exhausted T cells are active participants in tumor growth. As such, CD8+ exhausted T cells also produce TGFβ, which is sufficient to increase o-GTC growth in vitro, and α-PD1/TIGIT antibody treatment reduces Tgfb1 expression. Moreover, α-TGFβ antibody treatment attenuates Nf1-OPG growth in vivo, establishing that this cytokine is necessary for tumor proliferation. Relevant to future therapeutic application, the encouraging successes observed with the use of TGFβ inhibitors in combination with standard therapies for malignant glioma (HGG) and multiple myeloma55,56 provides support for the use of TGFβ inhibitors, including TGFβ-directed small molecule inhibitors, chimeric monoclonal antibodies, ligand traps, and vaccines57, as adjuvant therapies to improve ICI responses.

Fifth, in addition to discovering another growth regulatory mechanism, we identified other chemokines responsible for CD8+ T cell tumor infiltration. As such, we previously showed that Ccl2 was produced by optic glioma tumor cells, which attracts CD8+ T cells in a Ccr4-dependent manner32. Using an unbiased approach, we identified two chemoattractants, Ccl12 and Cxcl13, that are responsible for the recruitment of CD8+ T cells. While the mechanism for TAM production of Cxcl13 and Ccl12 in the Nf1-OPG model is unknown, we are actively investigating the role of cytokine/chemokines and other factors in TAM Cxcl13/Ccl12 regulation. Although Ccl12 is not expressed in humans, CXCL13 has been shown to mediate T cell attraction in other human cancers58,59. In this regard, CXCL13 is produced by macrophages in some cancers60 and was originally described as a B cell chemoattractant61. Moreover, it has been postulated to represent a potential marker to predict the response to ICI therapy62. Future studies will be required to determine whether CXCL13 could serve as a target for LGG therapy or a marker of disease progression.

Taken together, we report a new role for CD8+ exhausted T cells in LGGs where they function as part of two distinct immune paracrine stromal circuits that support tumor growth. Further investigation of this unique CD8+ T cell population may reveal additional molecular vulnerabilities that could lead to adjuvant immune-directed treatments for low-grade brain tumors in children.

Methods

All research is compliant with the relevant ethical bodies. All experiments were performed in compliance with active Animal Studies Committee protocols at the Washington University School of Medicine.

Mice

All experiments were performed according to the active Animal Studies Committee protocols at the Washington University School of Medicine (Washington University in St. Louis Institutional Animal Care and Use Committee). According to the ethics committee, any animals with compromised motion/eating habits or an unhealthy appearance are euthanized. No animals were euthanized due to their tumor burden or as a result of the treatments performed in this study. Mice harboring (1) a neomycin cassette insertion in exon 31 (Nf1 ±) with GFAP-Cre-mediated Nf1 loss in neuroglial progenitors (Nf1flox/neo; GFAP-Cre, Nf1-OPG mice)17, (2) the patient-derived germline Nf1 c.2041 C > T mutation30,31 with GFAP-Cre-mediated Nf1 loss in neuroglial progenitors (Nf1flox/R681X; GFAP-Cre, Nf1681C mice), and (3) the patient-derived germline Nf1 c.5425 C > T mutation21 with GFAP-Cre-mediated Nf1 loss in neuroglial progenitors (Nf1flox/R1809C; GFAP-Cre; Nf11809Cmice) were used. Nf1+/−, WT or Nf1flox/flox mice were employed as controls. Mice of both sexes were randomly assigned to all experimental groups. All mice used in these experiments were maintained on a strict C57BL/6 background and were housed in a specific pathogen-free barrier facility with controlled temperature (21 °C), humidity (55%), light-dark cycles (12:12 h) and ad libitum access to food and water. Mice were euthanasied by a barbiturate overdose using the Fatal Plus Pentobarbital solution at a dose of 86.6 mg/ml.

Optic nerve volume analysis

Dissected optic nerves were photographed using a Leica S9D Flexcam C3 microscope. The images were analyzed using ImageJ. Measurements at the chiasm 0 μm (D0), and at 150 μm (D150), 300 μm (D300), and 450 μm (D450) anterior to the chiasm were taken to generate optic nerve volume estimates, as previously reported19. The volumes for regions 1, 2, and 3 at the three 150 μm high-truncated cones were merged using the diameter (D0, D150, D300, and D450) values from each optic nerve measurement. Total optic nerve volumes were calculated according to this formula:

$$V1=\frac{1}{12}\pi ({D}_{0}^{2}+{D}_{0}{D}_{150}+{D}_{150}^{2})$$
(1)

In vivo mouse treatments

α-PD1 (BE0146; 200 μg/dose, twice per week), α-TIGIT (BE0274; 200 μg/dose, twice per week), combined α-PD1 and α-TIGIT (200 μg + 200 μg/dose, twice per week), α-CTLA4 (BE0131; 100 μg/dose, twice per week), α-TGFβ (BE0057; 200 μg/dose, twice per week), α-Nk1.1 (BE0036; 200 μg/dose, twice per week), or control IgG antibodies were administered to 12-week-old Nf1-OPG mice twice weekly i.p. for four consecutive weeks63,64,65,66,67. All monoclonal antibodies and their respective control IgG antibodies are listed in Supplementary Table 1.

Immunohistochemistry and immunofluorescence

Mice were euthanized and transcardially perfused with Ringer’s solution, followed by 4% PFA. Optic nerves were harvested and embedded in paraffin, while eyes were collected and embedded in OCT. Serial 4-μm-thick paraffin sections or 10-μm-thick retinal cryosections were immunostained with the primary antibodies listed in Supplementary Table 1. Development was performed using the Vectastain ABC kit (Vector Laboratories, PK4000) and appropriate biotinylated secondary antibodies (Vector Laboratories) or Alexa-fluor-conjugated secondary antibodies (Supplementary Table 1)22. Slides were imaged with a Leica ICC50W microscope using LAS EZ software, or with a Leica DFC3000G fluorescent microscope using LAS X software, or with a Thunder Imager 3D assay using LAS AF Lite 3.2.0 software. Skullcaps were harvested, and meningeal duras were peeled and stained as previously described68. Four sites in the superior sagittal sinus were imaged using a Zeiss LSM 880 confocal microscope (Washington University Center for Cellular Imaging).

RNA extraction and real-time RT-qPCR

RNA was isolated from optic nerves using the NucleoSpin RNA Plus kit (Takara-740984.205) according to the manufacturer’s instructions. For each experiment, two mice were used per sample. Total RNA was reverse transcribed into cDNA using the High-Capacity cDNA Reverse Transcription Kit (Applied Biosystems, 4368814). Real-time quantitative PCR (RT-qPCR) was performed by TaqMan gene expression (Alox1, Bap1, Ccl4, Ccl5, Ccl2, Ccl12, Cxcl9, Cxcl13, Cd274, Gapdh, Gpx4, Gzmb, Gzmk, Ifng, Nectin2, Pdcdlg2, Prf1, Pvr, Slc7a11; Supplementary Table 2) using the TaqMan Fast Advanced Master Mix (Applied Biosystems). All reactions were performed using the QuantStudio 3 (Applied Biosystems) system. ΔΔCT values were calculated using Gapdh as an internal control.

Flow cytometry

Dissociated frozen tumor single cells from de-identified human PA samples (clinical information in Supplemental Table 3) were acquired under an approved Human Studies Protocol as from the St. Louis Children’s Hospital Pediatric Tumor Bank. The frozen dissociated tumor single cells were thawed and resuspended in DMEM with 10% FBS. Blood, deep cervical lymph nodes, meninges, and optic nerve single cell isolations were generated from 12-week-old WT, Nf1+/−, Nf1-OPG, Nf1816C mice and Nf11809C mice were performed as previously described68. Red blood cells (RBC) were lysed with ACK lysis buffer (Quality Biological) and aspirated from the cell pellet. Once single cell suspensions were obtained, they were resuspended in fluorescence activated cell sorting (FACS) buffer on ice and incubated with the Zombie NIR Fixable Viability Kit (BioLegend, 423105). Samples were resuspended in FACS buffer with CD16/32 (FC block; BioLegend, 101302) or Human TruStain FcX (Fc Receptor Blocking Solution; BioLegend, 422301) for 15 min and fluorescently conjugated antibodies (Supplementary Table 1) on ice were added for 20 min. Samples were washed in FACS buffer, run on a Cytek Aurora spectral flow cytometer (Cytek) (Siteman Flow Cytometry Core), and analyzed using FlowJo software (Tree Star) (Supplementary Fig. 12).

scRNA sequencing and analysis

Dissociated cells from de-identified human PA samples (four female sporadic PAs and one female NF1-PA; clinical information in Supplemental Table 3) previously banked in the St. Louis Children’s Hospital Tumor Repository and obtained following appropriate patient consent under an approved Human Studies Protocol, were used for these experiments. The frozen dissociated tumor single cells were thawed and resuspended in DMEM. The cells were then stained with 7-Aminoactinomycin D (7-AAD) (Fisher Scientific, A1310) for sorting using the MoFlo (Siteman Flow Cytometry Core at the Washington University in St Louis). Live cells were collected and submitted for single cell RNA sequencing (scRNAseq).

Nf1-OPG optic nerves and meningeal dura specimens were collected separately from ten 12-week-old mice and pooled to prepare a single-cell suspension. Optic nerves were collected and dissociated using the neural dissociation kit (Miltenyi Biotec, 130-095-942) followed by debris (Miltenyi Biotec, 130-107-677) and myelin (Miltenyi Biotec, 130-096-733) removal. Single cell suspensions from meningeal dural tissue were prepared as described above, stained with 7-AAD, and sorted using MoFlo. Live cells were collected and sent for scRNA sequencing with TCR V(D)J enrichment.

Single-cell data processing, filtering, cohort data integration, and dimension reduction

For all samples, raw sequencing data was processed using the 10X Genomics Cell Ranger pipeline (version 6.1.1) to generate gene count matrices, and then aligned to the mm10-2020-A (mouse) or GRCh38 (human). All single cell RNA sequencing data were analyzed using Partek Flow software (version 10.0.23). Cell quality was evaluated by the number of reads, detected gene counts, and percentages of mitochondrial reads, followed by CPM (counts per million) normalization. To account for library chemistry and other possible unknown differences between experiments, cells were integrated using the Seurat3 integration method to produce batch-corrected data. PCA analysis followed by graph-based clustering using Louvain algorithm and t-SNE/ UMAP generation employing Euclidean distance were conducted on the first 20 principal components of the integrated expression data. To filter T cells from the datasets, clusters containing increased expression of CD3 genes were selected. Differential analyses were performed using gene specific analysis (GSA), and the results were filtered for genes with P values < 0.05, and fold changes either less than or equal to −2 or greater than or equal to 2. Pathway analyses were performed using DAVID Bioinformatics platform, and the significant pathways were selected by FDR < 0.1. Gene expression and TCR V(D)J data were overlaid by R scRepertoire package.

T cell isolation

Nf1+/− mouse spleens were homogenized into single cell suspensions by digestion in MACS buffer (Miltenyi Biotec, 130-091-222) with 3% BSA (Miltenyi Biotec, 130-091-376). Erythrocytes were lysed using RBC lysis buffer (eBioscience, 00433357). CD8+ T cells were isolated by selection using a CD8a T cell isolation kit (Miltenyi Biotec, 130-104-075). T cells were maintained at 2.5 × 106 cells/ml in RPMI-1640 medium supplemented with 10% FBS and 1% P/S. To mimic T cell exhaustion in vitro, CD8+ T cells were chronically stimulated every other day with α-mouse CD3 (Fisher Scientific, 16-0031-85) and α-mouse CD28 (Fisher Scientific, 16-0281-82)69. The NFAT1 inhibitor FK-506 (F4679) was purchased from Sigma-Aldrich.

ELISA

T cell conditioned media (TCM) from non-stimulated, exhausted (α-CD3+α-CD28 antibody stimulated), and exhausted (α-CD3+α-CD28 antibody stimulated) + FK-506 (10 μM) T cells were collected. Ccl4 and TGFβ in the TCM were detected using the mouse CCL4/MIP-1 beta Quantikine ELISA Kit (R&D Systems, MMB00) or the Mouse TGFβ ELISA Kit (Abcam, ab119557) according to the manufacturer’s instructions. Data from all of these colorimetric assays were collected on an Infinite M Nano (Tecan) microplate reader and analyzed using Magellan Pro 7.4 (Tecan) software.

Migration assays

5 × 105 T cells were added to the upper chamber of a transwell chamber (Corning, 6.5 mm insert, 3.0 μm polycarbonate membrane, CLS3398-2EA) with 200 μl serum free PRIM 1640 media. 500 μl of chemoattractant media (Ccl12 [25 ng/ml, 428-P5-025- R&D systems32] and Cxcl13 [1 µg/ml, 470-BC-025- R&D systems]70) was added to the lower chamber, and the number of T cells in the lower chambers counted 6 h later.

Optic glioma tumor cell (o-GTC) proliferation and apoptosis assays

104 o-GTCs were plated into 12-well plates coated with fibronectin (10 μg/ml) in complete NSC medium. O-GTCs were incubated with complete NSC medium, TCM from non-stimulated CD8+ T cells, and TCM from exhausted (α-CD3+α-CD28 antibody stimulated) CD8+ T cells for 48 h. Cells were fixed in 4% paraformaldehyde (PFA), and proliferation and apoptosis quantitated using the primary antibodies listed in Supplementary Table 1. Cells were labeled with secondary Alexa-fluor-conjugated secondary antibodies (Supplementary Table 1). The cells were imaged with a Leica DFC3000G fluorescent microscope using LAS X software.

Statistical analysis

Data were analyzed using GraphPad Prism software (GraphPad Prism 9.3.1). To determine differences between two groups, a two-tailed Mann–Whitney test was used, whereas multiple comparisons were analyzed using a one-way analysis of variance (ANOVA) with Dunnett’s, Tukey’s, Holm-Šídák multiple comparisons test. Statistical significance was set at P ≤ 0.05. All experiments were independently repeated at least three times. Numbers (n) are noted for each individual analysis.

Reporting summary

Further information on research design is available in the Nature Portfolio Reporting Summary linked to this article.