Glioblastoma (GBM) is an immunologically “cold” tumor that does not respond to current immunotherapy. Here, we demonstrate a fundamental role for the α-isoform of the catalytic subunit of protein phosphatase-2A (PP2Ac) in regulating glioma immunogenicity. Genetic ablation of PP2Ac in glioma cells enhanced double-stranded DNA (dsDNA) production and cGAS–type I IFN signaling, MHC-I expression, and tumor mutational burden. In coculture experiments, PP2Ac deficiency in glioma cells promoted dendritic cell (DC) cross-presentation and clonal expansion of CD8+ T cells. In vivo, PP2Ac depletion sensitized tumors to immune-checkpoint blockade and radiotherapy treatment. Single-cell analysis demonstrated that PP2Ac deficiency increased CD8+ T-cell, natural killer cell, and DC accumulation and reduced immunosuppressive tumor-associated macrophages. Furthermore, loss of PP2Ac increased IFN signaling in myeloid and tumor cells and reduced expression of a tumor gene signature associated with worse patient survival in The Cancer Genome Atlas. Collectively, this study establishes a novel role for PP2Ac in inhibiting dsDNA–cGAS–STING signaling to suppress antitumor immunity in glioma.

Significance:

PP2Ac deficiency promotes cGAS–STING signaling in glioma to induce a tumor-suppressive immune microenvironment, highlighting PP2Ac as a potential therapeutic target to enhance tumor immunogenicity and improve response to immunotherapy.

Despite ongoing improvements in surgery, radiation, and chemotherapy, GBM has a 5-year survival less than 5% and a median survival less than 15 months (1). More effective therapies are needed. Immune-checkpoint blockade, including anti-PD1/PD-L1 and anti-CTLA4, has been approved for multiple cancers but has not demonstrated clinical efficacy in GBM (2, 3). Low immunogenicity of GBM with minimal MHC-I expression and paucity of T-cell infiltration are major barriers for effective immune-checkpoint blockade (4–7).

PP2A is composed of a catalytic (C), regulatory (B), and scaffolding (A) subunit. PP2A is a major protein phosphatase that accounts for 50% to 70% of the total serine/threonine phosphatase activity in eukaryotic cells to counterbalance the regulatory effects of kinases in modulating signaling pathways that underlie normal physiology and pathobiology of cancer and other diseases (8, 9). We and others have demonstrated that inhibiting PP2Ac in tumor cells can impair DNA damage response (10–12). However, prior studies using immunocompromised models failed to address if PP2Ac inhibition alters antitumor immunity. Our group was the first to report that pharmacologic inhibition of the PP2Ac enhances the efficacy of immune-checkpoint blockade in multiple PD-1–resistant mouse tumor models (13, 14). However, given the ubiquity of PP2Ac expression in many cell types and the involvement of PP2Ac in many cellular pathways, the mechanisms of how PP2Ac regulates antitumor immunity are unclear.

cGAS is a critical sensor for cytosolic double-strand DNA (dsDNA) to activate innate immunity through STING–type I IFN signaling (15, 16). cGAS binding to dsDNA in tumors cells leads to the formation of 2′,3′-cyclic GMP-AMP (cGAMP). cGAMP can activate STING in tumor cells or can be exported as an “immunotrasmitter” to activate STING in dendritic cells (DC) and tumor-associated myeloid cells (17, 18). STING activation induces phosphorylation of interferon regulatory factor 3 (IRF3) to promote type I IFN production and antitumor immune responses (19). Here, using SB28, a glioma model with low tumor mutational burden (TMB), MHC-I expression, and resistance to immune-checkpoint blockade (20), we found that PP2Ac deficiency can unleash a CD8+ T cell–dependent antitumor immune response and sensitizes tumor to checkpoint immunotherapy. Mechanistically, glioma-specific PP2Ac deficiency promotes the accumulation of cytosolic dsDNA and cGAMP production, which in turn stimulate IFN signaling in both tumor and immune cells to remodel the immunosuppressive tumor microenvironment. Our results add to accumulating evidence that PP2Ac inhibition represents a promising novel strategy for antitumor immunotherapy. However, this study is the first to establish a role for PP2Ac in regulating cytosolic dsDNA production to mediate STING signaling activation.

Cell lines

Mouse glioma cell line SB28 (RRID: CVCL_A5ED) was kindly provided by Dr. Hideho Okada of University of California, San Francisco (San Francisco, CA). Mouse GL261 (RRID: CVCL_Y003) glioma cell line was kindly provided by Dr. Zhengping Zhuang of the NIH. Human SF8628 (RRID:CVCL_IT46) diffuse intrinsic pontine glioma (DIPG) cell line was purchased from Millipore Sigma. Human medulloblastoma cell line D425 (RRID:CVCL_1275) was provided by Dr. Samuel Cheshier of the University of Utah (Salt Lake City, UT). All cell lines were regularly examined (every 6–12 months) for Mycoplasma contamination (IDEXX, Clongen Laboratories, or Invivogen), last tested March 2023. SB28 and SF8628 are maintained in DMEM with 10% FBS. GL261 cells are maintained in RPMI-1640 with 10% FBS. D425 cells were plated in a neural stem cell expansion medium consisting of Neurobasal−A (Invitrogen), B27 (Invitrogen), human-bFGF (20 ng/mL; Shenandoah Biotech), human-EGF (20 ng/mL; Shenandoah Biotech), human recombinant LIF (Shenandoah Biotech), and heparin (10 ng/mL). All cells were maintained at 37°C under 5% CO2. Cells were passaged for not longer than 3 weeks from thawing to collection for experiments.

Animals

All animal work was approved by the Institutional Animal Care and Use Committee at the University of Texas at Austin and University of California San Francisco. Mice of both sexes, between the ages of 6 and 10 weeks of age, were used for the study. C57BL/6 (RRID:MGI:2159769) were obtained from The Jackson Laboratory. All mice were maintained under pathogen-free conditions.

In vivo experiments

For orthotopic brain tumor models, 8- to 10-week-old C57BL/6 mice (male and female in equal numbers) were used for intracranial studies. Cell lines were suspended in DMEM for inoculation. Mice were anesthetized with isoflurane, and tumor cells were injected orthotopically in 3 μL. Using a stereotactic frame, a burr hole was formed on the skull via 0.7-mm drill bit 1.5 mm laterally to the right and 1.5 mm rostrally from the lambda, and a noncoring needle (Hamilton 7804-04, 26s gauge) was used to inject the cells at a depth of 3 mm into the brain from the burr hole. Skin incision was sutured. Mice were then monitored daily. Survival endpoint was defined as weight loss greater than 20% relative to baseline, body conditioned score less than 2, or presence of focal neurologic deficits. For radiotherapy, 4 days after implantation, mice head was focally irradiated with an X-ray radiator (MultiRad 350, Precision), with 4 Gy daily for 2 consecutive days (4 × 4 Gy). Lead shield was applied to cover the mice except the cranium to achieve local radiation to the brain.

Flow cytometry analysis

Cells were trypsinized and washed with FACS buffer (PBS, 2% FBS, 1 mmol/L EDTA). Surface staining was performed by adding the surface antibodies to the cell suspension in 100 μL FACS buffer. After incubating for 30 minutes, cells were washed with FACS buffer and analyzed using the Cytek Aurora cytometer and analyzed using SpectroFlo (Cytek Bioscience) and FlowJo software (RRID:SCR_008520). For intracellular phospho-staining, cells were washed and resuspended in 4% formaldehyde for 15 minutes and then in ice for 10 minutes with chilled 100% methanol. Cells were then stained for phospho-protein.

DC–SB28-OVA coculture

Bone marrow–derived dendritic cells (BMDC) were derived as previously described (21). Briefly, mouse bone marrow cells were isolated from C57BL/6 mice and cultured in complete RPMI-1640 media with 50 μmol/L of β-mercaptoethanol (BME) and recombinant murine GM-CSF (20 ng/mL; PeproTech) for 8 days. 50K BMDC were plated with 250K of either WT or PP2A KO SB28 OVA cells in a 12-well plate. After 24 hours of incubation, the DCs were collected for flow cytometry analysis.

Cell-cycle analysis

The Click-iT EdU Alexa Fluor 647 Flow Cytometry Assay Kit (Thermo Scientific, C10424) was used per the manufacturer's instruction. EdU was incubated with cells for 2 hours prior to analysis by flow cytometry. DNA content was counterstained using FxCycle Violet (Thermo Scientific, F10347).

Tumor proliferation assay

Electrical impedance was measured every 30 minutes over 72 hours and normalized to the starting value (normalized cell index). For the data analysis, the time-points from when the signal is stable (after cell seeding or drug treatment) to 72 hours was used on the xCELLigence RTCA Software Pro (ACEA Biosciences Inc.).

T-cell proliferation assay

BMDC was differentiated in the same manner as above. Splenocytes from OT-I mouse were harvested and CD8+ T cells were isolated using column-based magnetic cell isolation (Miltenyi Biotec). CD8+ cells were stained with CellTrace Violet (CTV; Thermo Scientific) before being cultured with BMDC and either WT or PP2AKO-OVA tumor at a ratio of 5:1:1 (CD8+:BMDC:tumor). After 48 hours, suspension cells were collected for flow cytometry analysis.

T-cell intracellular staining assay

Splenocytes from OT-I mouse were harvested and CD8+ T cells were isolated using column-based magnetic cell isolation (Miltenyi Biotec). A total of 2 × 106 CD8+ cells were then plated with IL2 (50 ng/mL) and 25 μL of CD3/CD28 Dynabeads (Gibco). After 3 days, CD8 cells were plated with the WT or PP2AKO-OVA SB28 tumors at a ratio of 1:1, 1:5, and 1:10 (tumor: CD8). After 1 hour, brefeldin A (Invitrogen) was added and after another 5 more hours, the cells underwent surface staining for CD8+ and were then washed and resuspended in IC Fixation Buffer (Thermo Scientific) at room temperature for 20 minutes and then washed with Perm/Wash buffer (Thermo Scientific) before being stained for IFNγ and collected for flow cytometry analysis.

HMGB assay

The 50K of SB28 or GL261 cells were plated in a 96-well plate in 100 μL of the medium. After 24 hours, the supernatant was collected. For LB-100 treatment, cells were first treated with a dose titration of LB-100 and 3 hours later supernatant was collected. HMGB in the supernatant was quantified using the Lumit HMGB1 Human/Mouse Immunoassay kit (Promega, W6110) per the manufacturer's instruction.

PP2A phosphatase assay

The PP2A Phosphatase Assay Kit (Millipore) was used according to the manufacturer's instructions. Briefly, using the same amount of starting protein lysate for each condition, PP2A was immunoprecipitated using Anti-PP2A, C subunit (clone 1D6, Millipore) and protein A agarose slurry. The slurry was then washed with TBS before a standard amount of threonine phosphopeptide, a substrate of PP2A, was added to the mixture. Phosphate was released as a product of the reaction. The absolute amount of phosphate released was quantified with a malachite green solution, which was used as a measure of PP2A activity. Experiments were performed in triplicate, and the data are presented as a percentage mean of relative PP2A activity compared with control ± SEM.

Real-time PCR

Total RNA was extracted using the PureLink RNA Mini Kit (Invitrogen, 12183025) according to the manufacturer's instructions. cDNA synthesis was performed with 0.5 to 1 μg of total RNA using the High-Capacity cDNA Reverse Transcription Kit (Invitrogen, 4374967). mRNA levels were measured with gene-specific primers using the SYBR Green PCR Master Mix (Bio-Rad, 1725120). The results were normalized to GAPDH or OAZ in human or mouse samples, respectively. The primers used are shown in Supplementary Table S1.

Immunofluorescence staining

For CD8 and CALR visualization in mice tissue, SB28 tumors were collected at survival endpoint. Frozen tissue sections on slides were set at room temperature, then fixed with 4% paraformaldehyde followed by washing with PBS ×2, and then permeabilized with PBST (0.25% TritonX-100) and blocked with 10% normal goat serum/PBST for 30 minutes at room temperature. Samples were then incubated with primary antibodies at 4°C overnight in a humid box. After washing the samples with PBST for 5 × 10 minutes, secondary antibodies in PBST were added to the sample and incubated for 1 hour. 4,6-diamidino-2-phenylindole (DAPI; 300 nmol/L) was used to stain nuclei. Samples were covered with a coverslip with mounting medium and sealed with nail polish and then subjected for imaging using a confocal microscope (Zeiss). CD8+ cells were quantified by counting the number of CD8 within each field of view. CALR expression was quantified by the mean fluorescent intensity of the red channel of the region of interest. Analysis was performed on 3 independent samples per group (n = 3) and 3 regions of interest per sample.

Immune blotting and cell-surface protein detection

For immunoblot analysis, whole-cell lysates were prepared in RIPA lysis buffer (Thermo Scientific, 89900) containing Halt Protease Inhibitor Cocktail (Thermo Scientific, 78429). The protein concentrations were determined by BCA Protein Assay Kits (Pierce, 23227). A total of 20 to 30 μg protein samples were mixed with 4× Laemmli buffer (Bio-Rad, 1610747) and denatured at 95°C for 10 minutes. The sample was separated by SDS-PAGE and transferred to a nitrocellulose membrane (Bio-Rad, 1704270). Membranes were blocked with 3.5% BSA (Fisher, BP1600) and incubated with primary antibodies overnight at 4°C, followed by HRP-conjugated secondary antibodies for 2 hours at room temperature. Signal was detected using Clarity Western ECL (Bio-Rad, 1705061) and captured using the ChemiDoc Imaging System (Bio-Rad). Antibodies used are shown in Supplementary Table S2.

Quantification and statistical analysis

No statistical methods were used to predetermine the sample size. For cell-based experiments, biological triplicates were performed in each single experiment in general, unless otherwise stated. Animal experiments were performed in C57BL/6 mice. Animals were randomized into different groups after tumor cell inoculation; at least 9 to 10 mice were used for each group, unless otherwise indicated. Survival functions were estimated by the Kaplan–Meier methods and compared using the log-rank test or Wilcoxon rank tests as indicated. Two-tailed t tests were used to compare treatment versus control groups. ANOVA models were used to compare continuous outcomes across multiple experimental groups, unless otherwise indicated in each figure legend. Statistical analysis was performed using GraphPad Prism8 software (GraphPad Software, Inc.; RRID:SCR_002798).

Data and materials availability

Data including total RNA-seq, single-cell RNA-seq are deposited at NCBI GEO GSE213309. Whole-exome sequencing has been deposited to SRA and released under access SUB12057901 (PRJNA973640). Codes, QC results, and supplementary files can be found at Figshare (DOI: 10.6084/m9.figshare.21094390). Publicly available data generated by others were used by the authors including “Merged Cohort of LGG and GBM” (TCGA, Cell 2016, https://www.cbioportal.org/study/summary?id=lgggbm_tcga_pub). All other raw data generated in this study are available upon request from the corresponding author.

PP2Ac deficiency in glioma cells promotes type I IFN signaling and MHC-I expression

We previously showed that pharmacologic inhibition of PP2Ac synergizes with PD1 blockade in the GL261 murine glioma model (13). However, the underlying mechanism or cell type responsible for the PP2Ac-mediated effect on antitumor immunity is unclear. To investigate the effect of PP2Ac deficiency in glioma cells on antitumor immune response, we performed genetic KO of the predominant alpha-isoform of PP2Ac (Ppp2ca) by CRISPR in murine SB28. A clone with confirmed protein-level knockout by Western blot and frameshift mutations in both alleles of Ppp2ca by whole-exome sequencing (WES) was selected for further experiments (Supplementary Fig. S1A and S1B). Compared with GL261, SB28 more consistently recapitulate the immune-silent nature of human glioma with low mutational burden, minimal MHC-I expression, and resistance to immune-checkpoint blockade (20). We obtained the global gene-expression profile of WT and PP2AcKO cultured SB28 cells using RNA-seq. Gene ontology (GO) pathway analysis using differentially upregulated genes (log2FC > 0.5, FDR < 0.01) in PP2AcKO SB28 demonstrated that the top enriched biological pathways are related to type I IFN signaling and MHC-I antigen processing/presentation (Fig. 1A). Gene set enrichment analysis (GSEA) confirmed that type I IFN response pathway was enriched in PP2AcKO SB28 (Fig. 1B). Additionally, we confirmed the transcription of IFNβ and IFN response-related genes to be highly upregulated in PP2AcKO SB28 using RT-PCR (Fig. 1C). Phosphorylation of IRF3, which controls type I IFN transcription, and phosphorylation of STAT1, which is the direct downstream signaling of IFNα/β receptor (IFNAR) were both enhanced in PP2AcKO (Fig. 1D; Supplementary Fig. S1C). Given type I IFN has been shown to inhibit glioma proliferation (22), we examined if PP2Ac deficiency will affect tumor proliferation using the xCELLigence platform that provides continuous real-time measurement of cellular viability. PP2AcKO SB28 grew at a significantly lower rate than WT (Fig. 1E). We then performed cell-cycle analysis that showed PP2AcKO cells have decreased S and increased G0 and G2–M phases (Fig. 1F), suggesting that PP2AcKO cells have enhanced cell-cycle arrest. We then asked if increased cell death or apoptosis also contributed to decreased viability. We found no significant change in the frequency of an apoptosis marker, Annexin V (Fig. 1G). Given the enhanced IFN signaling, a known marker of immunogenic cell death (ICD), we asked if PP2AcKO tumors had an increase in other markers of ICD (23). Calreticulin (CALR) when exposed on plasma surface facilitates phagocytosis by DCs and in turn antigen uptake and presentation (23). CALR expression was significantly enhanced in PP2AcKO SB28 (Fig. 1H and I). Another ICD marker, high-mobility group box 1(HMGB1), when secreted, activates innate immune cells via TLR4-MYD88 signaling (23). We quantified HMGB1 level in tumor-conditioned medium and found a marked elevation of HMGB1 secretion by PP2AcKO cells (Fig. 1J). Downregulation of MHC-I in cancer cells is a common mechanism of immune evasion (24) and type I IFN is known to positively regulate MHC-I expression (25). Consistently, transcription of genes encoding an essential component of MHC-I (β2m), MHC loading machinery (Erap1, Tap 1, and Tap2), and transcriptional coactivator of MHC-I (NLRC5) was upregulated in PP2AcKO SB28 (Fig. 1K). The surface presentation of MHC-I was also consistently enhanced in PP2AcKO (Fig. 1L). We confirm the generalizability of this observation by generating CRISPR KO of PP2Ac in GL261 murine glioma, SF8628 human DIPG, and D425 human medulloblastoma cell lines, all of which demonstrated increased MHC-I expression (Fig. 1M; Supplementary Fig. S1D). Next, we asked if the observed increase in MHC-I expression in PP2AcKO cells is dependent on enhanced IFN signaling. Treatment of PP2AcKO SB28 with anti-IFNAR, which antagonizes type I IFN receptor, IFNAR (Fig. 1N) or ruxolitnib, a janus kinase (JAK) inhibitor (Supplementary Fig. S1E) that blocks JAK/STAT1 activity downstream of IFNAR, reduced MHC-I expression to level close to WT. In addition, the enhanced expression of IFN-related genes in PP2Ac-deficient tumors was also reversed by anti-IFNAR treatment (Fig. 1O). To confirm these mechanistic findings are not limited to SB28, we showed that PP2A deficiency in GL261 similarly resulted in decreased cellular proliferation (Supplementary Fig. S2A), increased pIRF3 and pSTAT1 (Supplementary Fig. S2B), enhanced IFN-stimulated genes (Supplementary Fig. S2C) that is dependent on IFNAR signaling (Supplementary Fig. S2D).

Figure 1.

PP2Ac deficiency in glioma cells promotes type I IFN signaling, ICD, and MHC-I expression. A, WT and PP2AcKO SB28 transcriptome profiles were analyzed by RNA-seq of (n = 3 per group). GO pathway enrichment analysis showing the top 10 enriched pathways ranked with highest fold enrichment using differentially upregulated genes in PP2AcKO compared with WT SB28 (Log2FC > 0.5, FDR < 0.01). *, P value of individual pathway. P value adjustment was performed using Bonferroni correction. B, GSEA plot for type I IFN signature among PP2AcKO vs. WT SB28. NES, normalized enrichment score. C, WT and PP2AcKO SB28 expression of IFN-related genes was measured via RT-qPCR. D, WT and PP2AcKO SB28 protein expression of pSTAT1/STAT1 and pIRF3/IRF3 was analyzed by immunoblotting. Vinculin (vin) was used as housekeeping control. E, Time course of xCELLigence assay showing the normalized cell index over 30 hours of the assay for WT and PP2AcKO SB28. Each time point measurement was from n = 4 biological replicates. F, Cell-cycle analysis of WT and PP2AcKO SB28 cells. Representative plots of DNA vs. EdU content. EdU positivity was used to identify S-phase, whereas DNA content was used to distinguish G0/1 and G2/M phase. *, level of significance for the difference between WT and PP2AcKO. G, The expression of apoptosis marker, Annexin V, in WT and PP2AcKO SB28 was determined by flow cytometry. H and I, Expression of ICD marker CALR. In WT and PP2AcKO SB28 was determined by flow cytometry (H) and immunofluorescence staining (I) for CALR (red) and nucleus [4,6-diamidino-2-phenylindole (DAPI), blue]. Scale bar, 20 μm. J, Conditioned medium was collected from WT and PP2AcKO SB28 cultures after 3 hours. Secreted HMGB levels were quantified using ELISA. K, WT and PP2AcKO SB28 expression of MHC-I components and antigen-presenting machinery genes was measured via RT-qPCR. L, WT and PP2AcKO SB28 surface expression of MHC-I was determined by flow cytometry. M, Surface expression of MHC-I in WT and PP2AcKO GL261 (murine glioma), SF8628 (human DIPG), and D425 (human medulloblastoma) was determined by flow cytometry. PP2Ac protein expression was analyzed by immunoblotting to confirm downregulation of protein expression. N, WT and PP2AcKO SB28 were treated with isotype control or anti-IFNAR for 72 hours. Surface expression of MHC-I was then determined by flow cytometry. O, WT and PP2AcKO SB28 were treated with isotype control or anti-IFNAR for 72 hours. Expression of IFN-related genes was measured via RT-qPCR. Heat maps reflect log2FC of the indicated gene relative to isotype-treated WT SB28. Data for C–O are from at least two independent experiments with similar results. Error bars, SEM. P values were calculated by unpaired t test. **, P < 0.01; ***, P < 0.001; ****, P < 0.0001; ns, nonsignificant. MFI, mean fluorescent intensity.

Figure 1.

PP2Ac deficiency in glioma cells promotes type I IFN signaling, ICD, and MHC-I expression. A, WT and PP2AcKO SB28 transcriptome profiles were analyzed by RNA-seq of (n = 3 per group). GO pathway enrichment analysis showing the top 10 enriched pathways ranked with highest fold enrichment using differentially upregulated genes in PP2AcKO compared with WT SB28 (Log2FC > 0.5, FDR < 0.01). *, P value of individual pathway. P value adjustment was performed using Bonferroni correction. B, GSEA plot for type I IFN signature among PP2AcKO vs. WT SB28. NES, normalized enrichment score. C, WT and PP2AcKO SB28 expression of IFN-related genes was measured via RT-qPCR. D, WT and PP2AcKO SB28 protein expression of pSTAT1/STAT1 and pIRF3/IRF3 was analyzed by immunoblotting. Vinculin (vin) was used as housekeeping control. E, Time course of xCELLigence assay showing the normalized cell index over 30 hours of the assay for WT and PP2AcKO SB28. Each time point measurement was from n = 4 biological replicates. F, Cell-cycle analysis of WT and PP2AcKO SB28 cells. Representative plots of DNA vs. EdU content. EdU positivity was used to identify S-phase, whereas DNA content was used to distinguish G0/1 and G2/M phase. *, level of significance for the difference between WT and PP2AcKO. G, The expression of apoptosis marker, Annexin V, in WT and PP2AcKO SB28 was determined by flow cytometry. H and I, Expression of ICD marker CALR. In WT and PP2AcKO SB28 was determined by flow cytometry (H) and immunofluorescence staining (I) for CALR (red) and nucleus [4,6-diamidino-2-phenylindole (DAPI), blue]. Scale bar, 20 μm. J, Conditioned medium was collected from WT and PP2AcKO SB28 cultures after 3 hours. Secreted HMGB levels were quantified using ELISA. K, WT and PP2AcKO SB28 expression of MHC-I components and antigen-presenting machinery genes was measured via RT-qPCR. L, WT and PP2AcKO SB28 surface expression of MHC-I was determined by flow cytometry. M, Surface expression of MHC-I in WT and PP2AcKO GL261 (murine glioma), SF8628 (human DIPG), and D425 (human medulloblastoma) was determined by flow cytometry. PP2Ac protein expression was analyzed by immunoblotting to confirm downregulation of protein expression. N, WT and PP2AcKO SB28 were treated with isotype control or anti-IFNAR for 72 hours. Surface expression of MHC-I was then determined by flow cytometry. O, WT and PP2AcKO SB28 were treated with isotype control or anti-IFNAR for 72 hours. Expression of IFN-related genes was measured via RT-qPCR. Heat maps reflect log2FC of the indicated gene relative to isotype-treated WT SB28. Data for C–O are from at least two independent experiments with similar results. Error bars, SEM. P values were calculated by unpaired t test. **, P < 0.01; ***, P < 0.001; ****, P < 0.0001; ns, nonsignificant. MFI, mean fluorescent intensity.

Close modal

Pharmacologic inhibition of PP2Ac promotes ICD and IFN signaling

LB-100, a small-molecule inhibitor of PP2Ac, is currently under clinical investigation (NCT04560972 and NCT03027388). We have previously demonstrated in the GL261 orthotopic model that LB-100 synergized with anti–PD1 blockade (13). However, the ubiquity of PP2Ac expression precluded the understanding of what cell type(s) is responsible for the immunogenic effect of LB-100. Our data suggest that tumor-intrinsic PP2Ac deficiency is sufficient to promote IFN signaling in vitro. To show the translation relevance of this effect, we asked if LB-100 can reproduce the phenotypes observed in PP2AcKO glioma cells. First, we confirmed the dose-dependent inhibition of PP2Ac activity in WT SB28 by LB-100 relative to PP2AcKO cells (Fig. 2A). The observed “residual” PP2Ac activity in PP2AcKO SB28 is likely attributed to the persistence of the beta isoform of the PP2Ac (Ppp2cb) that is not distinguished by the immunoprecipitation technique used for the assay. Using the xCELLigence platform, we found that WT SB28 was sensitive to LB-100 treatment, resulting in a dose-dependent decrease in cell viability (Fig. 2B and C) within several hours of drug treatment. We also found that LB-100 induced ICD in a dose-dependent fashion, with increased expression of CALR (Fig. 2D) and secretion of HMGB (Fig. 2E). Similarly, the phosphorylation of STAT1 (Fig. 2F), transcription of IFN-stimulated genes (Fig. 2G), and surface expression of MHC-I (Fig. 2H) were all enhanced by LB-100. We confirmed these are similarly observed in GL261 cells (Supplementary Fig. S2D–S2G).

Figure 2.

Pharmacologic inhibition of PP2Ac, using LB-100, in glioma promotes type I IFN signaling, ICD, and MHC-I expression. A, LB-100–treated WT SB28 with indicated concentrations for 3 hours and PP2AcKO SB28 were assessed for PP2A activity, measured using phosphatase assay, and expressed as ratio relative to untreated WT SB28. B, Time course of xCELLigence assay showing the normalized cell index following LB-100 treatment in WT SB28 cells at indicated concentrations. Each time point measurement was from n = 4 biological replicates. C, The relative normalized cell index 3 hours after LB-100 treatment at indicated concentrations compared with untreated WT SB28. D, LB-100–treated WT SB28 with indicated concentrations for 3 hours and PP2AcKO SB28 were assessed for surface CALR expression by flow cytometry. E, Conditioned medium from LB-100–treated WT SB28 with indicated concentrations for 3 hours and PP2AcKO SB28 were assessed for HMGB secretion using ELISA. F, LB-100–treated WT SB28 with indicated concentrations for 72 hours and PP2AcKO SB28 were assessed for pSTAT1 expression by flow cytometry. G, LB-100–treated WT SB28 with indicated concentrations for 72 hours and PP2AcKO SB28. Expression of IFN-related genes was measured via RT-qPCR. Heat maps reflect log2FC of the indicated gene relative to isotype-treated WT SB28. H, LB-100–treated WT SB28 with indicated concentrations for 72 hours and PP2AcKO SB28 were assessed for MHC-I expression by flow cytometry. All data from at least two independent experiments with similar results. Error bars, SEM. MFI, mean fluorescent intensity.

Figure 2.

Pharmacologic inhibition of PP2Ac, using LB-100, in glioma promotes type I IFN signaling, ICD, and MHC-I expression. A, LB-100–treated WT SB28 with indicated concentrations for 3 hours and PP2AcKO SB28 were assessed for PP2A activity, measured using phosphatase assay, and expressed as ratio relative to untreated WT SB28. B, Time course of xCELLigence assay showing the normalized cell index following LB-100 treatment in WT SB28 cells at indicated concentrations. Each time point measurement was from n = 4 biological replicates. C, The relative normalized cell index 3 hours after LB-100 treatment at indicated concentrations compared with untreated WT SB28. D, LB-100–treated WT SB28 with indicated concentrations for 3 hours and PP2AcKO SB28 were assessed for surface CALR expression by flow cytometry. E, Conditioned medium from LB-100–treated WT SB28 with indicated concentrations for 3 hours and PP2AcKO SB28 were assessed for HMGB secretion using ELISA. F, LB-100–treated WT SB28 with indicated concentrations for 72 hours and PP2AcKO SB28 were assessed for pSTAT1 expression by flow cytometry. G, LB-100–treated WT SB28 with indicated concentrations for 72 hours and PP2AcKO SB28. Expression of IFN-related genes was measured via RT-qPCR. Heat maps reflect log2FC of the indicated gene relative to isotype-treated WT SB28. H, LB-100–treated WT SB28 with indicated concentrations for 72 hours and PP2AcKO SB28 were assessed for MHC-I expression by flow cytometry. All data from at least two independent experiments with similar results. Error bars, SEM. MFI, mean fluorescent intensity.

Close modal

PP2Ac deficiency in glioma cells promote tumor-associated antigen expression, TAA-specific CD8+ T-cell cytotoxicity, and DC cross-presentation

Type I IFN is known to promote DC activation (26, 27). Given the increased expression of IFN-stimulated genes in PP2AcKO glioma cells, we asked if they are more efficient in activating DC. We generated WT and PP2AcKO SB28 cells expressing ovalbumin (OVA) and cocultured WT or PP2cKO SB28 cells with BMDCs. BMDCs cocultured with PP2AcKO SB28 compared with WT SB28 have increased expression of activation markers, including MHC-II, MHC-I, and CD86 (Fig. 3A). Moreover, they also had increased expression of OVA-H2Kb (SIINFEKL; Fig. 3A), which is an OVA peptide fragment presented on MHC-I of DC after phagocytosis of the tumor. Expression of SIINFEKL on DC is an indication of cross-presentation, suggesting that tumor PP2Ac deficiency can promote DC activation of tumor-associated antigen (TAA)–specific CD8+ T cells. Expression of activation markers was similarly enhanced in BMDC cocultured with non-OVA expressing SB28 or GL261 glioma cells (Supplementary Fig. S3). Next, we asked if this enhanced activation of DC is cell-contact dependent by performing coculture in a transwell insert. We similarly found an increase in the activation of DC cells (Fig. 3B), suggesting soluble factors secreted by tumor cells contributed to the promotion of DC activation.

Figure 3.

PP2A deficiency in glioma cells promotes the expression of TAA, TAA-specific CD8+ T-cell cytotoxicity, and DC activation. A, BMDC was cocultured with WT or PP2AcKO SB28-OVA, with a ratio of 1:5 for 24 hours. Surface expressions of SIINFEKL, CD86, MHC-I, and MHC-II were then determined by flow cytometry. B, BMDC was cocultured with WT or PP2AcKO SB28 using a transwell insert with a ratio of 1:5 for 24 hours. Surface expressions of CD86, MHC-I, and MHC-II were then determined by flow cytometry. C, WT and PP2AcKO SB28-OVA expression of OVA-H2Kb (SIINFEKL) was determined by flow cytometry. D–F, CD8 T cells were isolated from splenocytes of OT-I mice and then activated by CD3/CD28 beads and IL2 (50 ng/mL) for 72 hours. Activated T cells were then cocultured with WT or PP2AcKO SB28-OVA cells using three different effector:target ratios (E). After 24 hours, the percentage of dead tumor cells (CD45DAPI+) was quantified via flow cytometry and cocultured with WT and PP2AcKO SB28-Ova cells with an E:T ratio of 1:1 for 6 hours in the presence of brefeldin A (5 μg/mL) for the last 5 hours (F). Intracellular staining was then performed and the percentages of IFNγ+ CD8+ T cells were quantified using flow cytometry. G, BMDC was cocultured with WT or PP2AcKO SB28 and naïve OT-I CD8 T cells at a ratio of (1:5:1). CD8 T cells were prestained with CTV. Forty-eight hours later, the proliferation of CD8 T cells was quantified by analyzing the dilution of CTV using flow cytometry. Division index was calculated using FlowJo software. Representative FACS plots of CTV on CD8+ OT-I T cells. Data are from one experiment representative of at least two independent experiments with similar results. Error bars, SEM. P values were calculated by an unpaired t test. **, P < 0.01; ***, P < 0.001; ****, P < 0.0001. (A and B, Created with BioRender.com.)

Figure 3.

PP2A deficiency in glioma cells promotes the expression of TAA, TAA-specific CD8+ T-cell cytotoxicity, and DC activation. A, BMDC was cocultured with WT or PP2AcKO SB28-OVA, with a ratio of 1:5 for 24 hours. Surface expressions of SIINFEKL, CD86, MHC-I, and MHC-II were then determined by flow cytometry. B, BMDC was cocultured with WT or PP2AcKO SB28 using a transwell insert with a ratio of 1:5 for 24 hours. Surface expressions of CD86, MHC-I, and MHC-II were then determined by flow cytometry. C, WT and PP2AcKO SB28-OVA expression of OVA-H2Kb (SIINFEKL) was determined by flow cytometry. D–F, CD8 T cells were isolated from splenocytes of OT-I mice and then activated by CD3/CD28 beads and IL2 (50 ng/mL) for 72 hours. Activated T cells were then cocultured with WT or PP2AcKO SB28-OVA cells using three different effector:target ratios (E). After 24 hours, the percentage of dead tumor cells (CD45DAPI+) was quantified via flow cytometry and cocultured with WT and PP2AcKO SB28-Ova cells with an E:T ratio of 1:1 for 6 hours in the presence of brefeldin A (5 μg/mL) for the last 5 hours (F). Intracellular staining was then performed and the percentages of IFNγ+ CD8+ T cells were quantified using flow cytometry. G, BMDC was cocultured with WT or PP2AcKO SB28 and naïve OT-I CD8 T cells at a ratio of (1:5:1). CD8 T cells were prestained with CTV. Forty-eight hours later, the proliferation of CD8 T cells was quantified by analyzing the dilution of CTV using flow cytometry. Division index was calculated using FlowJo software. Representative FACS plots of CTV on CD8+ OT-I T cells. Data are from one experiment representative of at least two independent experiments with similar results. Error bars, SEM. P values were calculated by an unpaired t test. **, P < 0.01; ***, P < 0.001; ****, P < 0.0001. (A and B, Created with BioRender.com.)

Close modal

Next, we investigated whether enhanced MHC-I expression in PP2Ac-deficient glioma affects tumor killing by TAA-specific CD8+ T cells. We first confirmed that PP2AcKO SB28 cells have increased expression of OVA-H2Kb (SIINFEKL; Fig. 3C). Then, we tested if PP2Ac deficiency-mediated enhanced MHC-I expression is functionally relevant for TAA-specific CD8+ T-cell cytotoxicity by incubating WT or PP2AcKO SB28-OVA cells with OT-I T cells preactivated by CD3/CD28 beads (Fig. 3D). We found a significant increase in OT-I T cells mediated killing against PP2AcKO SB28-OVA cells (Fig. 3E) across a range of effector:target (E:T) ratios. The percentage of IFNγ producing OT-I T cells was also significantly enhanced when OT-I T cells were cocultured with PP2AcKO SB28 (Fig. 3F), suggesting that PP2AcKO SB28 cells were more susceptible to TAA-specific T-cell cytotoxicity. Given the enhanced activation of BMDC primed by PP2Ac-deficient tumors (25, 26), we tested the ability of tumor-primed BMDC to activate naïve CD8+ OT-I T cells by coculturing BMDC, WT or PP2AcKOSB28-OVA tumors and naïve CD8+ OT-I T cells labeled with CTV dye. BMDC primed by PP2AcKO tumor significantly enhanced OT-I CD8+ cell proliferation (Fig. 3G). Collectively, these data show that PP2Ac deficiency in glioma cells enhances TAA presentation on MHC-I, promotes DC maturation and cross-presentation of TAA-specific antigen to activate naïve CD8+ T cells, and increases susceptibility to TAA-specific T cell–mediated killing.

PP2Ac deficiency enhances survival of glioma tumor-bearing mice and sensitizes checkpoint therapy

We next examined the effect of PP2Ac deficiency in glioma cells in vivo by orthotopically implanting WT or PP2AcKO tumors in C57BL/6 mice. Survival was significantly prolonged in mice bearing PP2AcKO compared with WT SB28 in both SB28 and GL261 (Fig. 4A and B). We first examined the degree of CD8+ T-cell infiltration in the tumor microenvironment. At the survival endpoint, SB28 tumors were stained for CD8 T cells by immunofluorescence. We found a significant increase in tumor-infiltrating CD8+ T cells in PP2AcKO tumors (Fig. 4C). Given the increased abundance of tumor-infiltrating CD8+ T cells, we asked if they are required or contributed to the prolonged survival of PP2AcKO glioma bearing mice. We performed systemic CD8+ T-cell depletion by treating mice with isotype or anti-CD8 depleting antibody prior to tumor implantation and throughout the study. CD8 depletion significantly diminished, but did not completely abolish, the survival benefit of PP2Ac deficiency, suggesting that CD8-mediated adaptive immune response contributed to but is not completely responsible for the beneficial effects of PP2Ac deficiency (Fig. 4D). As we have previously shown that tumor-intrinsic type I IFN signaling promoted MHC-I expression and other IFN-stimulated genes, we tested if PP2Ac deficiency-mediated enhanced survival is dependent on IFN signaling by systemic blockade of IFNAR. Abrogation of type I IFN signaling completely reversed the survival benefit of PP2Ac deficiency (Fig. 4E). Given we observed a significant increase in ICD in vitro (Fig. 1HJ), we asked if this is similarly observed in vivo. Tumors harvested from mice in Fig. 4E at the survival endpoint were stained for CALR by immunofluorescence. We found a significant increase in CALR expression in PP2AcKO tumors (Fig. 4F), which is abolished in mice treated with anti-IFNAR (Fig. 4F). This suggests that PP2Ac deficiency promoted ICD in a type I IFN-dependent manner. Next, we asked if PP2Ac deficiency sensitizes mice to checkpoint blockade. We treated tumor-bearing mice with isotype or combined anti-CTLA4/PD1 blockade. Although dual checkpoint blockade marginally increased the median survival of mice bearing WT SB28, it dramatically prolonged survival of mice bearing PP2AcKO SB28. In addition, the rate of mice with complete response (CR) was strikingly increased from 10% to 80% (Fig. 4G). We then asked if CR mice from PP2AcKO tumors carry immunologic memory against WT and PP2AcKO SB28 tumors by rechallenging CR mice with WT and PP2AcKO SB28 tumors. We simultaneously implanted WT and PP2AcKO SB28 cells in the left and right flank, respectively (Fig. 4H). We found that CR mice had protective immunity against PP2AcKO SB28 and partial immunity against WT SB28. PP2AcKO SB28 was unable to grow in cured mice, whereas WT SB28 grew to smaller sizes in CR mice than in naïve mice (Fig. 4H; Supplementary Fig. S4). These data indicate that PP2Ac-deficient glioma cured by checkpoint blockade can elicit adaptive memory antitumor immunity. However, the fact that memory immunity against WT tumors was incomplete suggests that WT tumors can evade immunosurveillance despite shared neoantigens with PP2AcKO tumors.

Figure 4.

PP2Ac deficiency enhances the survival of glioma tumor-bearing mice and sensitizes checkpoint therapy. A, C57BL/6 mice were inoculated with 3 × 104 WT or PP2AcKO SB28 cells in the brain (n = 18–19). Cumulative survival over time. B, C57BL/6 mice were inoculated with 5 × 104 WT or PP2AcKO GL261 cells in the brain (n = 9–10). Cumulative survival over time. C, At the survival endpoint, WT or PP2AcKO SB28 tumors were harvested, and histologic analysis was performed staining for CD8 (red) and nucleus (DAPI, blue). Scale bar, 10 μm. Nine regions from two tumor samples were used for quantification. D, C57BL/6 mice were treated with anti-CD8 (clone 2.43) depletion antibody or isotype control (rat IgG2B). Mice were given 250 μg i.p. on days −3, −2, −1 and then inoculated with 3 × 104 WT or PP2AKO SB28 cells in the brain (day 0). Depleting antibody or isotype was then given 2×/week until endpoint. (n = 9–10). Cumulative survival over time. E, C57BL/6 mice were inoculated with 3×104 WT or PP2AcKO SB28 cells in the brain. On day 0, mice were randomized to treatment with isotype (mouse IgG1) or anti-IFNAR (MAR1-5A3) blocking antibody (200 μg/mouse). Treatment was continued 2×/week until the endpoint (n = 8–10). Cumulative survival over time. F, At the survival endpoint, WT or PP2AcKO SB28 tumors treated with isotype or anti-IFNAR were harvested, and histologic analysis was performed staining for CALR (red) and nucleus (DAPI, blue). Scale bar, 20 μm. Three regions from each sample and three tumor samples were used for quantification. Mean fluorescent intensity (MFI) of the red channel was used to quantify CALR staining. G, C57BL/6 mice were inoculated with 3 × 104 WT or PP2AcKO SB28 cells in the brain. At day 5, mice were randomized to treatment with isotype (hamster IgG + rat IgG2a) or anti-PD1 (RMP1-14) + anti-CTLA4 (9H10) blocking antibody (250 μg/mouse, 2×/week for 5 weeks). Cumulative survival over time. H, C57BL/6 mice inoculated with PP2AcKO SB28 tumors and cured by treatment with anti-PD1 + anti-CTLA4 were rechallenged by subcutaneous injection of 6×105 WT and PP2AcKO SB28 tumors in the left and right flank, respectively. Naïve C57BL/6 mice were used as control. Tumor volume of both WT and PP2AcKO SB28 was measured 20 days after implantation in naïve and cured mice. Tumors were harvested and photographed. No tumor was grossly noted in cured mice rechallenged with PP2AcKO SB28 tumors. Wilcoxon rank tests were used for survival analysis. Error bars, SEM. P values were calculated by unpaired t test for C and one-way ANOVA with Tukey multiple comparison test for F and H. *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001. Data are from two experiments and combined in A. Data are representative of one independent experiment (B–H).

Figure 4.

PP2Ac deficiency enhances the survival of glioma tumor-bearing mice and sensitizes checkpoint therapy. A, C57BL/6 mice were inoculated with 3 × 104 WT or PP2AcKO SB28 cells in the brain (n = 18–19). Cumulative survival over time. B, C57BL/6 mice were inoculated with 5 × 104 WT or PP2AcKO GL261 cells in the brain (n = 9–10). Cumulative survival over time. C, At the survival endpoint, WT or PP2AcKO SB28 tumors were harvested, and histologic analysis was performed staining for CD8 (red) and nucleus (DAPI, blue). Scale bar, 10 μm. Nine regions from two tumor samples were used for quantification. D, C57BL/6 mice were treated with anti-CD8 (clone 2.43) depletion antibody or isotype control (rat IgG2B). Mice were given 250 μg i.p. on days −3, −2, −1 and then inoculated with 3 × 104 WT or PP2AKO SB28 cells in the brain (day 0). Depleting antibody or isotype was then given 2×/week until endpoint. (n = 9–10). Cumulative survival over time. E, C57BL/6 mice were inoculated with 3×104 WT or PP2AcKO SB28 cells in the brain. On day 0, mice were randomized to treatment with isotype (mouse IgG1) or anti-IFNAR (MAR1-5A3) blocking antibody (200 μg/mouse). Treatment was continued 2×/week until the endpoint (n = 8–10). Cumulative survival over time. F, At the survival endpoint, WT or PP2AcKO SB28 tumors treated with isotype or anti-IFNAR were harvested, and histologic analysis was performed staining for CALR (red) and nucleus (DAPI, blue). Scale bar, 20 μm. Three regions from each sample and three tumor samples were used for quantification. Mean fluorescent intensity (MFI) of the red channel was used to quantify CALR staining. G, C57BL/6 mice were inoculated with 3 × 104 WT or PP2AcKO SB28 cells in the brain. At day 5, mice were randomized to treatment with isotype (hamster IgG + rat IgG2a) or anti-PD1 (RMP1-14) + anti-CTLA4 (9H10) blocking antibody (250 μg/mouse, 2×/week for 5 weeks). Cumulative survival over time. H, C57BL/6 mice inoculated with PP2AcKO SB28 tumors and cured by treatment with anti-PD1 + anti-CTLA4 were rechallenged by subcutaneous injection of 6×105 WT and PP2AcKO SB28 tumors in the left and right flank, respectively. Naïve C57BL/6 mice were used as control. Tumor volume of both WT and PP2AcKO SB28 was measured 20 days after implantation in naïve and cured mice. Tumors were harvested and photographed. No tumor was grossly noted in cured mice rechallenged with PP2AcKO SB28 tumors. Wilcoxon rank tests were used for survival analysis. Error bars, SEM. P values were calculated by unpaired t test for C and one-way ANOVA with Tukey multiple comparison test for F and H. *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001. Data are from two experiments and combined in A. Data are representative of one independent experiment (B–H).

Close modal

PP2Ac deficiency increases TMB and promotes cytoplasmic DNA accumulation and cGAS–STING activation

PP2A is known to play a role in modulating DNA damage repair (DDR; refs. 11, 12, 28, 29). A recent study demonstrated that inactivation of a PP2A scaffolding subunit promotes microsatellite instability in colon cancer (30). We asked if increased antigenicity by enhanced neoantigen production contributed to increased immunogenicity in PP2AcKO SB28 by analyzing the mutational landscape of WT and PP2AcKO SB28 using WES. We identified a significant increase in TMB attributed to new missense mutations in PP2AcKO cells (Fig. 5A). Frequency of frameshift and inframe deletion was unchanged (Supplementary Fig. S5A). In addition, we ran algorithms to predict peptide binding to MHC-I or MHC-II to estimate potential neoepitope formation and found a significant increase in neoepitopes for MHC-I and MHC-II in PP2AcKO SB28 (Fig. 5B; Supplementary Fig. S5B). These data suggest that PP2Ac deficiency, likely through its impact on DDR, promotes TMB and in turn neoantigen formation to enhance tumor antigenicity. We confirmed that PP2Ac-deficient glioma cells harbor increased unrepaired dsDNA in the nucleus by quantifying phosphorylated γH2AX (H2AX-P) in SB28 (Fig. 5C and D) and GL261 cells (Supplementary Fig. S6A and S6B). However, our in vitro data using the OVA-OTI model showed that PP2Ac deficiency could enhance DC priming and OVA-specific T-cell cytotoxicity, suggesting that tumor immunogenicity is enhanced not solely by the promotion of neoantigen formation. Our group and others have previously shown that PP2Ac is critical in regulating G2–M transition, and PP2A inhibition in combination with DNA-damaging treatment (such as chemotherapy or radiation) can promote aberrant entry into mitosis, resulting in cell death by mitotic catastrophe (10–12, 28, 31, 32). It has also been shown that mitotic progression following DNA damage can promote micronuclei formation to induce cGAS–STING–IFN activation (33). We, therefore, hypothesize that PP2Ac deficiency can promote activation of cGAS–STING–type I IFN by promoting the accumulation of cytosolic dsDNA. We measured the level of dsDNA in the cytoplasmic fraction of WT and PP2AcKO SB28, GL261, and D425 cells and found a significant increase in dsDNA accumulation in PP2AcKO tumor cells (Fig. 5E). We confirmed this using immunofluorescence staining of dsDNA in SB28 (Fig. 5F) and GL261 (Supplementary Fig. S6C). Next, we asked if enhanced cytosolic accumulation of dsDNA translates into increased production of cGAMP, which can be exported by tumor cells as “immunotransmitter” and be taken up by host antigen-presenting cells, such as macrophages and DCs, to activate STING to produce type I IFN (17, 18). We measured cGAMP in whole-cell lysate of WT and PP2AcKO SB28, GL261, and D425 cells and found cGAMP production to be enhanced in PP2Ac-deficient tumors (Fig. 5G). This is consistent with our coculture experiment that showed PP2Ac deficiency could promote DC activation in a contact-independent fashion (Fig. 3B), as cGAMP can be exported by tumor to activate immune cells. To determine if the increase in IFN signaling in PP2AcKO tumors is dependent on cGAS, we generated PP2Ac/cGAS double KO (dKO) SB28 cells. We found the promotion of IFN signaling in PP2AcKO SB28 as assessed by protein expression of pIRF3 and pSTAT1 was markedly reduced in dKO SB28 compared with PP2AKO SB28 cells (Fig. 5H). We also found the host of overexpressed IFN-simulated genes in PP2AcKO to be significantly decreased in dKO SB28 (Fig. 5I). These data suggest that PP2Ac deficiency promotes type I IFN signaling in a cGAS-dependent fashion.

Figure 5.

PP2Ac deficiency promotes cytoplasmic DNA accumulation and cGAS–STING activation. A, WES was performed on WT and PP2AcKO SB28 cells. The total number of nonsynonymous somatic mutations (left) and missense mutations (right) was quantified. B, Number of predicted neoepitopes for MHC-I and MHC-II binding. C and D, Unrepaired dsDNA breaks in the nucleus of WT and PP2AcKO SB28 cells were assessed by immunofluorescence staining (C) for H2AX-P (red) and nucleus (DAPI; blue). Fifteen cells were randomly chosen for quantification (scale bar, 10 μm) and immunoblotting (D) for protein expression of H2AX-P in whole-cell lysate. Cyclophilin B (cyc) was used as housekeeping control. E, Cytoplasmic extract was collected from WT or PP2AKO SB28, GL261, and D425 cells. dsDNA normalized to 106 live cells was quantified. F, WT and PP2AcKO SB28 cells underwent immunofluorescence staining for dsDNA(red) and nucleus (DAPI; blue). Scale bar, 20 μm. G, Whole-cell lysate was collected from WT or PP2AKO SB28, GL261, and D425 cells, and cGAMP level was measured with ELISA. H, WT, PP2AcKO, cGASKO, and PP2AcKO cGASKO dKO SB28 protein expression of pSTAT1/STAT1 and pIRF3/IRF3 was analyzed by immunoblotting. Cyclophilin B (cyc) was used as housekeeping control. I, WT, PP2AcKO, cGASKO, and PP2AcKO cGASKO dKO SB28 expression of IFN-related genes, MHC-I components, and antigen-presenting machinery genes was measured via RT-qPCR. P values indicate comparison between cGASKO and PP2Ac+cGAS dKO. J, WT and PP2AcKO SB28 cells were radiated with the indicated doses. Twenty-four hours later, the cytoplasmic extract was collected, and dsDNA/106 live cells were measured. K, C57BL/6 mice were inoculated with 3×104 WT or PP2AcKO SB28 cells in the brain. At day 4, mice were randomized to sham or focal brain radiation (4 Gy on days 4 and 5 for a total dose of 8 Gy; n = 9–10). Cumulative survival over time. Wilcoxon rank tests were used for survival analysis. Error bars, SEM. P values were calculated by an unpaired t test for A, C, E, G, and J and by unpaired one-way ANOVA with Tukey multiple comparison test for I. Data are representative of at least two independent experiments with similar results for C–J. Data are representative of one experiment in K. **, P < 0.01; ***, P < 0.001; ****, P < 0.0001.

Figure 5.

PP2Ac deficiency promotes cytoplasmic DNA accumulation and cGAS–STING activation. A, WES was performed on WT and PP2AcKO SB28 cells. The total number of nonsynonymous somatic mutations (left) and missense mutations (right) was quantified. B, Number of predicted neoepitopes for MHC-I and MHC-II binding. C and D, Unrepaired dsDNA breaks in the nucleus of WT and PP2AcKO SB28 cells were assessed by immunofluorescence staining (C) for H2AX-P (red) and nucleus (DAPI; blue). Fifteen cells were randomly chosen for quantification (scale bar, 10 μm) and immunoblotting (D) for protein expression of H2AX-P in whole-cell lysate. Cyclophilin B (cyc) was used as housekeeping control. E, Cytoplasmic extract was collected from WT or PP2AKO SB28, GL261, and D425 cells. dsDNA normalized to 106 live cells was quantified. F, WT and PP2AcKO SB28 cells underwent immunofluorescence staining for dsDNA(red) and nucleus (DAPI; blue). Scale bar, 20 μm. G, Whole-cell lysate was collected from WT or PP2AKO SB28, GL261, and D425 cells, and cGAMP level was measured with ELISA. H, WT, PP2AcKO, cGASKO, and PP2AcKO cGASKO dKO SB28 protein expression of pSTAT1/STAT1 and pIRF3/IRF3 was analyzed by immunoblotting. Cyclophilin B (cyc) was used as housekeeping control. I, WT, PP2AcKO, cGASKO, and PP2AcKO cGASKO dKO SB28 expression of IFN-related genes, MHC-I components, and antigen-presenting machinery genes was measured via RT-qPCR. P values indicate comparison between cGASKO and PP2Ac+cGAS dKO. J, WT and PP2AcKO SB28 cells were radiated with the indicated doses. Twenty-four hours later, the cytoplasmic extract was collected, and dsDNA/106 live cells were measured. K, C57BL/6 mice were inoculated with 3×104 WT or PP2AcKO SB28 cells in the brain. At day 4, mice were randomized to sham or focal brain radiation (4 Gy on days 4 and 5 for a total dose of 8 Gy; n = 9–10). Cumulative survival over time. Wilcoxon rank tests were used for survival analysis. Error bars, SEM. P values were calculated by an unpaired t test for A, C, E, G, and J and by unpaired one-way ANOVA with Tukey multiple comparison test for I. Data are representative of at least two independent experiments with similar results for C–J. Data are representative of one experiment in K. **, P < 0.01; ***, P < 0.001; ****, P < 0.0001.

Close modal

Previous studies demonstrated that radiation can promote antitumor immunity through activation of cGAS–STING signaling by enhancing dsDNA production (34). We therefore asked if PP2Ac deficiency can promote radiation induced dsDNA accumulation and further enhance radiation-induced antitumor immunity. We radiated WT or PP2AcKO SB28 with increasing dosage (0–20 Gy) and found that radiation complemented the effect of PP2Ac deficiency in promoting dsDNA production (Fig. 5J). To test this effect in vivo, we orthotopically implanted WT or PP2AcKO SB28 and administered focal brain radiation on post-implant day 4 (4 Gy on days 4 and 5 for a total dose of 8 Gy). PP2AcKO SB28 tumors were significantly more sensitive to radiation, achieving a 50% durable response compared with 0% in WT (Fig. 5K). These data suggest that PP2Ac deficiency in tumor cells can synergize with radiotherapy.

scRNA-seq analysis reveals glioma-specific PP2Ac deficiency remodeled immune compartment of the tumor microenvironment

To explore how glioma PP2Ac deficiency reshapes the tumor microenvironment in vivo, we performed scRNA-seq of WT and PP2AcKO SB28 murine glioma after orthotopic implantation. Tumors were harvested on post-implant day 18 and disaggregated. Tissues from 3 mice were pooled for each condition and the whole-cell content was analyzed by scRNA-seq. Unsupervised clustering and uniform manifold approximation and projection (UMAP) analyses were performed on 15,448 cells (Fig. 6A). Immune cells were segregated by the expression of CD45+ and tumor cells by expression of GFP. We utilized the Seurat package to perform fine clustering (Supplementary Fig. S7 and Supplementary Data S1). PP2Ac deficiency in glioma cells distinctly remodeled both the tumor and immune compartments (Fig. 6B). We first focused on the effect of glioma PP2Ac deficiency on immune cells. We utilized gene expression of canonical markers to classify CD8+ T cells (CD3+ CD8b1); CD4+ T cells (CD3+ CD4+); natural killer (NK) cells (NKG7+CD3); macrophages (TAM; CD11b+CD11c); DCs (CD11b+CD11c+; Supplementary Fig. S8A). The DC population was further subclassified into conventional type 1 DC cDC1 (CD11c+, CD103+), which is critical for antigen cross-presentation. We found the frequency of CD8 T cells, NK cells, and cDC1 cells increased 3.8- to 5.5-fold in PP2AcKO tumors (Fig. 6C; Supplementary Fig. S8B). The infiltration of T cells, NK and cDC1s in human cancers has each been associated with improved prognosis and response to immunotherapy (35–37). Consistent with PP2Ac-deficient tumors being more sensitive to checkpoint blockade, we found an increase in expression of PD1 in T cells and PD-L1 in tumor and myeloid cells of PP2AcKO tumors (Supplementary Fig. S8C; ref. 38).

Figure 6.

scRNA-seq analysis reveals tumor-specific PP2Ac deficiency remodeled immune compartment of the tumor microenvironment. C57BL/6 mice were inoculated with 3 × 104 WT or PP2AcKO SB28 cells in the brain. On day 18 after implantation, tumors were harvested and three mice per group were pooled and analyzed by scRNA-seq. A, UMAP analyses were performed on 15,448 cells. Expression of GFP and CD45 identified tumor and immune cells, respectively. B, Glioma-specific PP2AcKO significantly remodeled both tumor and immune compartments. C, Canonical markers were then used to identify major immune populations. The frequencies of NK (NKG7+, CD3), CD8 (CD8+, CD3+), and cDC1 (CD11c+, CD103+) cells among all sequenced cells were compared between WT and PP2AcKO tumors. D, Volcano plot showing DEGs [−log10 (adjusted P) > 20, log2(FC) > 0.5 or < −0.5; red] in TAMs between WT and PP2AcKO tumors. E, GO pathway enrichment analysis was performed using upregulated DEGs in TAMs and DCs from PP2AcKO versus WT SB28. The top 7 enriched biological processes for upregulated DEGs ranked by fold enrichment are shown. *, P value of individual pathway. P value adjustment was performed using Bonferroni correction). F, UMAP plot of TAMs and DC comparing expression levels of IRF7 (IFN-response element), H2-Aa (MHC class I), and CD206 (marker of “M2”-like immunosuppressive TAMs) between WT and PP2AcKO SB28 tumors. G, UMAP plots showing the expression of key signature genes that are differential between the various TAM subsets. H, Overview of TAM subsets: 16, M-MDSC–derived macrophage; 11, classic monocyte; 14 and 13, microglia; 0, IFN BMDM. I, The fold change in frequency of the 5 TAMs clusters between WT and PP2AcKO SB28 tumors. J, Heat map of normalized enrichment score from GSEA identifying pathway enrichment in each TAM cluster.

Figure 6.

scRNA-seq analysis reveals tumor-specific PP2Ac deficiency remodeled immune compartment of the tumor microenvironment. C57BL/6 mice were inoculated with 3 × 104 WT or PP2AcKO SB28 cells in the brain. On day 18 after implantation, tumors were harvested and three mice per group were pooled and analyzed by scRNA-seq. A, UMAP analyses were performed on 15,448 cells. Expression of GFP and CD45 identified tumor and immune cells, respectively. B, Glioma-specific PP2AcKO significantly remodeled both tumor and immune compartments. C, Canonical markers were then used to identify major immune populations. The frequencies of NK (NKG7+, CD3), CD8 (CD8+, CD3+), and cDC1 (CD11c+, CD103+) cells among all sequenced cells were compared between WT and PP2AcKO tumors. D, Volcano plot showing DEGs [−log10 (adjusted P) > 20, log2(FC) > 0.5 or < −0.5; red] in TAMs between WT and PP2AcKO tumors. E, GO pathway enrichment analysis was performed using upregulated DEGs in TAMs and DCs from PP2AcKO versus WT SB28. The top 7 enriched biological processes for upregulated DEGs ranked by fold enrichment are shown. *, P value of individual pathway. P value adjustment was performed using Bonferroni correction). F, UMAP plot of TAMs and DC comparing expression levels of IRF7 (IFN-response element), H2-Aa (MHC class I), and CD206 (marker of “M2”-like immunosuppressive TAMs) between WT and PP2AcKO SB28 tumors. G, UMAP plots showing the expression of key signature genes that are differential between the various TAM subsets. H, Overview of TAM subsets: 16, M-MDSC–derived macrophage; 11, classic monocyte; 14 and 13, microglia; 0, IFN BMDM. I, The fold change in frequency of the 5 TAMs clusters between WT and PP2AcKO SB28 tumors. J, Heat map of normalized enrichment score from GSEA identifying pathway enrichment in each TAM cluster.

Close modal

Next, we examined the impact of glioma PP2Ac deficiency on myeloid cells, which are the predominant immune cells. We identified the differentially expressed genes (DEG; −log10(adjusted P) > 20, log2(FC) > 0.5 or log2(FC) < −0.5) in myeloid cell clusters (TAMs + DC: clusters 0, 11, 12, 13, 14, 16) between WT and PP2AcKO tumors (Fig. 6D). Consistent with our in vitro finding of PP2Ac deficiency enhancing tumor cGAMP production, which can activate STING-IFN signaling in myeloid cells, we found the top upregulated DEGs to be known IFN response genes (ISG15, IRF7, STAT1, CXCL19, RSAD2, and Mx1). In addition, components of MHC-I (B2m, H2-Q1, H2-Q7, and H2-Q6), MHC-I loading machinery (TAP1 and TAPBP), MHC-II (H2-DMa and H2-Aa), and CD74—an invariant MHC-II chain implicated in trafficking of MHC-II proteins—were all highly enriched (Fig. 6D). An unbiased GO pathway analysis demonstrated the top enriched biological pathways of myeloid DEGs in PP2AcKO tumors were related to type I IFN signaling and MHC-II antigen processing/presentation (Fig. 6E and F). On the other hand, among downregulated DEGS in myeloid cells of PP2AcKO tumors were Mrc1(CD206) and Arg1 (Fig. 6D and F), markers frequently associated with anti-inflammatory, immunosuppressive macrophages (39–41). Next, we further characterized the heterogenous TAMs populations. Seurat analysis identified 5 TAMs clusters (0, 11, 13, 14, 16). We examined the expression of known myeloid markers among these clusters (Fig. 6G and H). Cluster 11 expressed markers of classic bone marrow–derived monocytes (Chil3+), whereas cluster 0 had low Chil3 expression and high expression of mature macrophage markers such as C1Q. Clusters 13 and 14 expressed TMEM119, indicating they are likely resident microglia. Cluster 16 exclusively expressed S100A9, which is a marker of immunosuppressive monocytic myeloid-derived suppressor cells (M-MDSC)-derived macrophages (42). We calculated the fold change in cluster frequencies between WT and PP2AcKO tumors and found that clusters 0 and 11 were enriched in PP2AcKO tumors, whereas clusters 13, 14, and 16 were enriched in PP2AcWT tumors (Fig. 6I). To delineate the functional significance of these clusters, we performed GSEA using cluster-defining gene lists (Supplementary Data S1) and compared the relative expression of selected pathways across the five clusters (Fig. 6J). Clusters 0 and 11, which were enriched in PP2AcKO tumors, had increased type I/II IFN signature in addition to NFkB activation. This is consistent with increased STING activation as STING is known to stimulate both IFN and NFkB downstream signaling (43). They also expressed high antigen processing/presentation activity. Clusters 13, 14, 16, which were enriched in WT tumors, all had suppressed IFN signature. Cluster 14 was notable for a distinctly high oxidative phosphorylation signature, which is associated with immunosuppressive “M2”-like macrophages (44). S100A9-positive M-MDSC–derived macrophages (cluster 16) are associated with shorter survival in patients with head and neck cancer and poor response to PD-1 antibody treatment in patients with metastatic melanoma (42). In summary, our data demonstrated that PP2Ac deficiency in glioma cells significantly remodels the immune tumor microenvironment. There was a significant increase in IFN-activated macrophages with enhanced markers of activation and antigen presentation. There was also a decrease in immunosuppressive macrophages. cDC1 DC infiltration was also enhanced, which presumably led to an influx of cytotoxic CD8 T cells and NK cells. These observations are consistent with PP2Ac deficiency-mediated dsDNA accumulation and cGAMP production that can activate STING signaling in antigen-presenting cells (TAMs and DC).

scRNA-seq analysis reveals glioma-specific PP2Ac deficiency remodeled tumor compartment

Finally, we examined the impact of glioma PP2Ac deficiency on GFP+ SB28 cells. Seurat analysis identified 9 clusters (1, 2, 3, 4, 5, 6, 7, 8, 17). We analyzed the DEGs [−log10(adjusted P) > 20, log2(FC) > 0.5 or log2(FC) < −0.5] between WT and PP2AcKO tumors and found the top overexpressed genes in PP2AcKO glioma cells were IFN response elements (ISG15, STAT1, CXCL10, and IRF7) including components of MHC-I (B2m and H2-D1) and MHC-I loading machinery (TAP1 and TAPBP; Fig. 7A). GO pathway analysis using upregulated DEGs in glioma cells of PP2AcKO tumor demonstrated that the top enriched biological pathways were predominately related to type I IFN signaling (Fig. 7B and C). We calculated fold change in cluster frequencies between WT and PP2AcKO tumors and found that clusters 17 and 1 were enriched in PP2AcKO, whereas clusters 7, 8, and 5 were enriched in WT tumors (Fig. 7D). GSEA was performed to compare the relative expression of gene signatures of selected pathway across the 5 clusters (Fig. 7E). Cluster 17, which was markedly enriched in PP2AcKO tumors (by >30-fold), was defined by high type I/II IFN signature. Clusters 5, 7, and 8, which were enriched in WT tumors, had enhanced signature of oxidative phosphorylation. In addition, cluster 8 had increased expression stem cell makers, whereas clusters 5 and 7 had increased MYC target expression. To establish if the downregulated DEG (log2FC < −0.5, FDR < 0.01) signature in GFP+ tumor clusters (1, 2, 3, 4, 5, 6, 7, 8, 17) in PP2AcKO tumors (PP2ADN; Supplementary Table S3) has clinical significance in human samples, we obtained bulk RNA-seq data set from The Cancer Genome Atlas (TCGA) with combined low-grade glioma (LGG) and glioblastoma (GBM). We calculated the average normalized expression of PP2ADN genes and found its expression to be significantly higher in GBM compared with LGG (Fig. 7F). Survival is also worse for patients with higher PP2ADN expressions in the combined GBM/LGG cohort (Fig. 7G). When grading is stratified, PP2ADN expressions continue to significantly predict worse survival in LGG patients (Supplementary Fig. S9A), whereas there is a trend toward worse survival in GBM patients (P = 0.08) with limited sample size (Supplementary Fig. S9B). In summary, the scRNA-seq data demonstrated that glioma PP2Ac deficiency resulted in a marked increase in IFN activation and MHC-I expression in an immune-silent glioma model. In addition, the gene signature downregulated in PP2Ac-deficient glioma cells correlates with higher grading and worse prognosis in human clinical data. Therefore, this study strongly argues for PP2A inhibition as an appealing strategy for novel treatment of GBM.

Figure 7.

scRNA-seq analysis reveals tumor-specific PP2Ac deficiency remodeled tumor compartment of the tumor microenvironment. A, Volcano plot showing DEGs [−log10(adjusted P) > 20, log2(FC) > 0.5 or < −0.5; red] in GFP+ SB28 cells between WT and PP2AcKO tumors. Pseudovalue 1e−300 was added to adjusted P to avoid 0 before log-transformation. B, GO pathway enrichment analysis performed using upregulated DEGs in glioma cells from PP2AcKO vs. WT SB28 tumors. The top 7 enriched biological processes for upregulated DEGs ranked by fold enrichment are shown. *, P value of individual pathway. P value adjustment was performed using Bonferroni correction. C, UMAP plot of glioma cells comparing expression levels of ISG15 (IFN-response element), H2-D1 (MHC class I), and TAP1 (MHC-I loading machinery) between WT and PP2AcKO SB28 tumors. D, The fold change in frequency of glioma clusters between WT and PP2AcKO SB28 tumors. E, Heat map of normalized enrichment score from GSEA identifying pathway enrichment in each tumor cluster with altered frequency between WT and PP2AcKO SB28. F, Average expression level of PP2ADN gene signature (downregulated DEGs in PP2AcKO glioma cells from A) is higher in high-grade glioma (n = 171) compared with low-grade glioma (n = 530) from TCGA data set. P value was calculated by unpaired t test. ****, P < 0.0001. G, Average expression level of PP2ADN gene signature is associated with worse survival in patients with glioma (LGG + GBM) using TCGA bulk RNA-seq data set. Q4 represents patients with highest quartile of average expression and Q1 with the lowest quartile of average expression. Mantel–Cox log-rank tests were used for survival analysis. ****, P < 0.0001.

Figure 7.

scRNA-seq analysis reveals tumor-specific PP2Ac deficiency remodeled tumor compartment of the tumor microenvironment. A, Volcano plot showing DEGs [−log10(adjusted P) > 20, log2(FC) > 0.5 or < −0.5; red] in GFP+ SB28 cells between WT and PP2AcKO tumors. Pseudovalue 1e−300 was added to adjusted P to avoid 0 before log-transformation. B, GO pathway enrichment analysis performed using upregulated DEGs in glioma cells from PP2AcKO vs. WT SB28 tumors. The top 7 enriched biological processes for upregulated DEGs ranked by fold enrichment are shown. *, P value of individual pathway. P value adjustment was performed using Bonferroni correction. C, UMAP plot of glioma cells comparing expression levels of ISG15 (IFN-response element), H2-D1 (MHC class I), and TAP1 (MHC-I loading machinery) between WT and PP2AcKO SB28 tumors. D, The fold change in frequency of glioma clusters between WT and PP2AcKO SB28 tumors. E, Heat map of normalized enrichment score from GSEA identifying pathway enrichment in each tumor cluster with altered frequency between WT and PP2AcKO SB28. F, Average expression level of PP2ADN gene signature (downregulated DEGs in PP2AcKO glioma cells from A) is higher in high-grade glioma (n = 171) compared with low-grade glioma (n = 530) from TCGA data set. P value was calculated by unpaired t test. ****, P < 0.0001. G, Average expression level of PP2ADN gene signature is associated with worse survival in patients with glioma (LGG + GBM) using TCGA bulk RNA-seq data set. Q4 represents patients with highest quartile of average expression and Q1 with the lowest quartile of average expression. Mantel–Cox log-rank tests were used for survival analysis. ****, P < 0.0001.

Close modal

Our group previously reported that LB-100, a small-molecule inhibitor of PP2Ac currently in phase Ib and II clinical trials (NCT03027388 and NCT04560972), can synergize with anti-PD1 checkpoint blockade in preclinical models of non-CNS (14) and CNS tumors (13). However, as PP2Ac is ubiquitously expressed in many cell types, from tumor to immune cells, our previous work using LB-100 failed to delineate what cell type is responsible for the effect of enhanced tumor immunogenicity. In addition, LB-100 also has an antagonistic effect against another phosphatase family, PPP5C, thereby raising the question of whether PP2Ac is the target responsible for promoting tumor immunogenicity (45). In this study, we found that tumor-specific PP2Ac deficiency can promote tumor immunogenicity and sensitize checkpoint blockade in vivo using a nonimmunogenic glioma model SB28. Mechanistically, we found that PP2Ac deficiency promotes ICD, dsDNA accumulation, cGAMP production, and TMB. scRNA-seq demonstrated that glioma-specific PP2AcKO can remodel the immune microenvironment by enhancing the infiltration of IFN-activated TAMs. Tumor infiltration of cross-presenting cDC1 cells, NK cells, and cytotoxic CD8 T cells are all concomitantly increased. These data suggest that the downregulation of PP2Ac in tumor cells can promote antitumor immunity through enhanced cGAS–STING–IFN signaling.

PP2Ac is known to be required in various steps of DNA damage response (46). A recent study showed that the inactivation of a scaffolding subunit of PP2A, PPP2r1a, can induce microsatellite instability by increasing phosphorylation of retinoblastoma protein and histone deacetylase 2, which trigger neoantigen production and in turn promote immunogenicity of tumors in non-CNS cancer models (30). Consistently, we found an increase in TMB and predicted neoepitope bound to MHC-I and MHC-II in PP2AcKO glioma. However, we demonstrate in this study that PP2Ac inactivation can enhance cGAS–STING–IFN signaling in both tumor and immune cells, providing a complementary mechanistic rationale to inhibit PP2Ac to enhance antitumor immunity. The connection between tumor PP2Ac and cGAS–STING–IFN stimulation has not been reported and is the principal novelty and significance of the current study. The precise mechanism of how PP2Ac deficiency leads to the accumulation of cytosolic dsDNA remains to be elucidated. In macrophages, PP2A has also been implicated in the dephosphorylation of IRF3 as a deactivation mechanism that contributes to the termination of IRF3-type I IFN signaling in response to Toll-like receptor and STING (47, 48). Whether this is a relevant regulatory mechanism in tumor cells, specifically in response to cGAS-STING activation, has not been explored. Therefore, beyond the increase in cytosolic dsDNA, we cannot rule out the possibility that PP2Ac deficiency can contribute to enhanced IFN signaling through downstream regulation of the STING–IFN pathway.

Our in vitro data demonstrated that PP2Ac deficiency in tumor cells promoted MHC-I expression in an IFNAR and JAK–STAT-dependent manner, suggesting that KO of PP2Ac promotes autocrine activation of tumor IFN signaling. Systemic IFNAR blockade in vivo abolished the survival benefit of tumor PP2Ac deficiency. Although activation of the IFNAR-JAK/STAT pathway induces expression of interferon-stimulated genes and acutely promotes the efficacy of cancer immunotherapies (25–27), it is also recognized that chronic tumor IFNAR-JAK/STAT stimulation could be counterproductive (49, 50). The role of type I IFN in the GBM microenvironment is controversial as both beneficial (51, 52) and detrimental effects (53, 54) of IFN stimulation had been suggested. A recent study in melanoma showed that the impact of type I IFN signaling on antitumor immunity depends on the balance between immune and tumor type I IFN stimulation (49). Type I IFN signaling in immune cells is beneficial, whereas tumor type I IFN stimulation can have opposing effects. On one hand, it carries the benefit of promoting MHC-I expression, on the other, it induces “adaptive resistance” mechanisms such as upregulation of PD-L1 or IDO that can be immunosuppressive (50). Our scRNA-seq data suggest that in PP2Ac-deficient tumor, IFN signaling is enhanced in both immune and tumor cells. It is possible that in the context of a highly immune-silent tumor such as glioma with low baseline MHC-I expression, the benefits of IFN-induced MHC-I expression outweigh the detriment of adaptive resistance.

Not only is PP2Ac expressed in many cell types, but it is also implicated in many signaling pathways. The diverse function of PP2A holoenzyme is dictated by the heterogenous regulatory B subunits. Therefore, there is reservation in systemically inhibiting the conserved PP2Ac subunit for fear of eliciting unacceptable side effects or antagonizing specific PP2A complexes that function as tumor suppressors (55). In fact, some groups advocate for the development of allosteric PP2A activators of specific PP2A holoenzyme (56) involved in positively regulating tumor-suppressor pathways. Our study is significant in providing the rationale that, at least in the context of glioma, even though inhibition of PP2Ac in tumor cells may perturb various pathways, on balance it results in a favorable therapeutic outcome of increased immunogenicity and sensitivity to checkpoint blockade. It is also possible that PP2Ac deficiency can have a nonimmune mediated, tumor-intrinsic effect on glioma cells. Our in vivo CD8 depletion studies demonstrated a residual benefit of PP2Ac deficiency with CD8 depletion in the absence of checkpoint blockade, which can be attributed to other immune cells (NK, CD4, and DC) or a direct effect on tumor growth. Our scRNA-seq data showed that the downregulated gene signature in PP2Ac-deficient glioma cells is correlated with higher histologic grade of glioma and predicts worse survival in patients, which suggests that PP2Ac deficiency has the potential to directly affect tumor growth independent of its effect on immunogenicity. The translational significance of this study is the implication that if systemic pharmacologic inhibition (i.e., with LB-100) cannot achieve sufficient PP2A inhibition in glioma, either due to the blood–brain barrier or systemic intolerance, local delivery of LB-100, such as via convection-enhanced delivery can be a viable alternative strategy.

I. Mondal reports grants from NIH and Department of Defense (DOD) during the conduct of the study. O. Das reports grants from NIH and the DOD during the conduct of the study. Z. Meng reports grants from NIH/NIGMS, the Elsa U. Pardee Foundation, and NIH/NINDS during the conduct of the study and from the Stanley J. Glaser Foundation outside the submitted work. R.O. Lu reports grants from the NIH and DOD during the conduct of the study; in addition, R.O. Lu has a patent for oxabicycloheptanes for modulation of immune response issued and with royalties paid. W.S. Ho reports grants from NIH/NINDS, Rally Foundation, and Matthew Larson Pediatric Brain Tumor Foundation during the conduct of the study; in addition, W.S. Ho has a patent for oxabicycloheptanes for modulation of immune response issued and with royalties paid. No disclosures were reported by the other authors.

I. Mondal: Data curation, formal analysis, writing–review and editing. O. Das: Data curation, formal analysis, writing–review and editing. R. Sun: Data curation. J. Gao: Data curation. B. Yu: Formal analysis. A. Diaz: Supervision. J. Behnan: Data curation. A. Dubey: Data curation. Z. Meng: Writing–review and editing. E. Eskandar: Writing–review and editing. B. Xu: Software, formal analysis, supervision, visualization, methodology, writing–review and editing. R.O. Lu: Conceptualization, data curation, formal analysis, supervision, funding acquisition, methodology, writing–original draft, writing–review and editing. W.S. Ho: Conceptualization, data curation, formal analysis, supervision, funding acquisition, investigation, methodology, writing–original draft, writing–review and editing.

This study was supported by Department of Defense PRCRP W81XWH-20-1-0428 (R.O. Lu), NIH R01NS126501 (R.O. Lu), NIH R01NS131545 (W.S. Ho), and NIH R35GM142504 (Z. Meng). RNA-seq and scRNA-seq were performed by the Genomic Sequencing and Analysis Facility at UT Austin, Center for Biomedical Research Support. RRID#: SCR_021713.

The publication costs of this article were defrayed in part by the payment of publication fees. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/).

1.
Tamimi
AF
,
Juweid
M
.
Epidemiology and outcome of glioblastoma
.
Glioblastoma
2017
.
Available from:
http://www.ncbi.nlm.nih.gov/pubmed/29251870.
2.
Reardon
DA
,
Brandes
AA
,
Omuro
A
,
Mulholland
P
,
Lim
M
,
Wick
A
, et al
.
Effect of nivolumab vs bevacizumab in patients with recurrent glioblastoma: the CheckMate 143 phase 3 randomized clinical trial
.
JAMA Oncol
2020
;
6
:
1003
10
.
3.
Lim
M
,
Weller
M
,
Idbaih
A
,
Steinbach
J
,
Finocchiaro
G
,
Raval
RR
, et al
.
Phase 3 trial of chemoradiotherapy with temozolomide plus nivolumab or placebo for newly diagnosed glioblastoma with methylated MGMT promoter
.
Neuro Oncol
2022
;
24
:
1935
49
.
4.
Yeung
JT
,
Hamilton
RL
,
Ohnishi
K
,
Ikeura
M
,
Potter
DM
,
Nikiforova
MN
, et al
.
LOH in the HLA class I region at 6p21 is associated with shorter survival in newly diagnosed adult glioblastoma
.
Clin Cancer Res
2013
;
19
:
1816
26
.
5.
Wei
J
,
Barr
J
,
Kong
L-Y
,
Wang
Y
,
Wu
A
,
Sharma
AK
, et al
.
Glioblastoma cancer-initiating cells inhibit T-cell proliferation and effector responses by the signal transducers and activators of transcription 3 pathway
.
Mol Cancer Ther
2010
;
9
:
67
78
.
6.
Alexandrov
LB
,
Nik-Zainal
S
,
Wedge
DC
,
Aparicio
SAJR
,
Behjati
S
,
Biankin
Av
, et al
.
Signatures of mutational processes in human cancer
.
Nature
2013
;
500
:
415
21
.
7.
Jackson
CM
,
Choi
J
,
Lim
M
.
Mechanisms of immunotherapy resistance: lessons from glioblastoma
.
Nat Immunol
2019
;
20
:
1100
9
.
8.
Westermarck
J
.
Targeted therapies don't work for a reason; the neglected tumor suppressor phosphatase PP2A strikes back
.
FEBS J
2018
;
285
:
4139
45
.
9.
Sangodkar
J
,
Farrington
CC
,
McClinch
K
,
Galsky
MD
,
Kastrinsky
DB
,
Narla
G
.
All roads lead to PP2A: exploiting the therapeutic potential of this phosphatase
.
FEBS J
2016
;
283
:
1004
24
.
10.
Ho
WS
,
Sizdahkhani
S
,
Hao
S
,
Song
H
,
Seldomridge
A
,
Tandle
A
, et al
.
LB-100, a novel protein phosphatase 2A (PP2A) inhibitor, sensitizes malignant meningioma cells to the therapeutic effects of radiation
.
Cancer Lett
2018
;
415
:
217
26
.
11.
Lu
J
,
Kovach
JS
,
Johnson
F
,
Chiang
J
,
Hodes
R
,
Lonser
R
, et al
.
Inhibition of serine/threonine phosphatase PP2A enhances cancer chemotherapy by blocking DNA damage-induced defense mechanisms
.
Proc Natl Acad Sci U S A
2009
;
106
:
11697
702
.
12.
Wei
D
,
Parsels
LA
,
Karnak
D
,
Davis
MA
,
Parsels
JD
,
Marsh
AC
, et al
.
Inhibition of protein phosphatase 2A radiosensitizes pancreatic cancers by modulating CDC25C/CDK1 and homologous recombination repair
.
Clin Cancer Res
2013
;
19
:
4422
32
.
13.
Maggio
D
,
Ho
WS
,
Breese
R
,
Walbridge
S
,
Wang
H
,
Cui
J
, et al
.
Inhibition of protein phosphatase-2A with LB-100 enhances antitumor immunity against glioblastoma
.
J Neurooncol
2020
;
148
:
231
44
.
14.
Ho
WS
,
Wang
H
,
Maggio
D
,
Kovach
JS
,
Zhang
Q
,
Song
Q
, et al
.
Pharmacologic inhibition of protein phosphatase-2A achieves durable immune-mediated antitumor activity when combined with PD-1 blockade
.
Nat Commun
2018
;
9
:
2126
.
15.
Woo
SR
,
Fuertes
MB
,
Corrales
L
,
Spranger
S
,
Furdyna
MJ
,
Leung
MYK
, et al
.
STING-dependent cytosolic DNA sensing mediates innate immune recognition of immunogenic tumors
.
Immunity
2014
;
41
:
830
42
.
16.
Sun
L
,
Wu
J
,
Du
F
,
Chen
X
,
Chen
ZJ
.
Cyclic GMP-AMP synthase is a cytosolic DNA sensor that activates the type I interferon pathway
.
Science
2013
;
339
:
786
91
.
17.
Li
J
,
Duran
MA
,
Dhanota
N
,
Chatila
WK
,
Bettigole
SE
,
Kwon
J
, et al
.
Metastasis and immune evasion from extracellular cGAMP hydrolysis
.
Cancer Discov
2021
;
11
:
1212
27
.
18.
Carozza
JA
,
Böhnert
V
,
Nguyen
KC
,
Skariah
G
,
Shaw
KE
,
Brown
JA
, et al
.
Extracellular cGAMP is a cancer cell-produced immunotransmitter involved in radiation-induced anti-cancer immunity
.
Nat Cancer
2020
;
1
:
184
96
.
19.
Yum
S
,
Li
M
,
Fang
Y
,
Chen
ZJ
.
TBK1 recruitment to STING activates both IRF3 and NF-κB that mediate immune defense against tumors and viral infections
.
Proc Natl Acad Sci
2021
;
118
:
e2100225118
.
20.
Genoud
V
,
Marinari
E
,
Nikolaev
SI
,
Castle
JC
,
Bukur
V
,
Dietrich
P-Y
, et al
.
Responsiveness to anti-PD-1 and anti-CTLA-4 immune checkpoint blockade in SB28 and GL261 mouse glioma models
.
Oncoimmunology
2018
;
7
:
e1501137
.
21.
Roney
K
.
Bone marrow-derived dendritic cells
.
Methods Mol Biol
2013
;
1031
:
71
6
.
22.
Chen
K-S
,
Reinshagen
C
,
van Schaik
TA
,
Rossignoli
F
,
Borges
P
,
Mendonca
NC
, et al
.
Bifunctional cancer cell-based vaccine concomitantly drives direct tumor killing and antitumor immunity
.
Sci Transl Med
2023
;
15
:
eabo4778
.
23.
Fucikova
J
,
Kepp
O
,
Kasikova
L
,
Petroni
G
,
Yamazaki
T
,
Liu
P
, et al
.
Detection of immunogenic cell death and its relevance for cancer therapy
.
Cell Death Dis
2020
;
11
:
1013
.
24.
Dhatchinamoorthy
K
,
Colbert
JD
,
Rock
KL
.
Cancer immune evasion through loss of MHC class I antigen presentation
.
Front Immunol
2021
;
12
:
636568
.
25.
Dighe
AS
,
Richards
E
,
Old
LJ
,
Schreiber
RD
.
Enhanced in vivo growth and resistance to rejection of tumor cells expressing dominant negative IFNγ receptors
.
Immunity
1994
;
1
:
447
56
.
26.
Fuertes
MB
,
Kacha
AK
,
Kline
J
,
Woo
S-R
,
Kranz
DM
,
Murphy
KM
, et al
.
Host type I IFN signals are required for antitumor CD8+ T cell responses through CD8α+ dendritic cells
.
J Exp Med
2011
;
208
:
2005
16
.
27.
Diamond
MS
,
Kinder
M
,
Matsushita
H
,
Mashayekhi
M
,
Dunn
GP
,
Archambault
JM
, et al
.
Type I interferon is selectively required by dendritic cells for immune rejection of tumors
.
J Exp Med
2011
;
208
:
1989
2003
.
28.
Hong
CS
,
Ho
W
,
Zhang
C
,
Yang
C
,
Elder
JB
,
Zhuang
Z
.
LB100, a small molecule inhibitor of PP2A with potent chemo- and radio-sensitizing potential
.
Cancer Biol Ther
2015
;
16
:
821
33
.
29.
Gordon
IK
,
Lu
J
,
Graves
CA
,
Huntoon
K
,
Frerich
JM
,
Hanson
RH
, et al
.
Protein phosphatase 2A inhibition with LB100 enhances radiation-induced mitotic catastrophe and tumor growth delay in glioblastoma
.
Mol Cancer Ther
2015
;
14
:
1540
7
.
30.
Yen
YT
,
Chien
M
,
Wu
PY
,
Ho
CC
,
HC
te
,
Huang
KCY
, et al
.
Protein phosphatase 2A inactivation induces microsatellite instability, neoantigen production and immune response
.
Nat Commun
2021
;
12
:
7297
.
31.
Ho
WS
,
Feldman
MJ
,
Maric
D
,
Amable
L
,
Hall
MD
,
Feldman
GM
, et al
.
PP2A inhibition with LB100 enhances cisplatin cytotoxicity and overcomes cisplatin resistance in medulloblastoma cells
.
Oncotarget
2016
;
7
:
12447
63
.
32.
Lv
P
,
Wang
Y
,
Ma
J
,
Wang
Z
,
Li
JL
,
Hong
CS
, et al
.
Inhibition of protein phosphatase 2A with a small molecule LB100 radiosensitizes nasopharyngeal carcinoma xenografts by inducing mitotic catastrophe and blocking DNA damage repair
.
Oncotarget
2014
;
5
:
7512
24
.
33.
Harding
SM
,
Benci
JL
,
Irianto
J
,
Discher
DE
,
Minn
AJ
,
Greenberg
RA
.
Mitotic progression following DNA damage enables pattern recognition within micronuclei
.
Nature
2017
;
548
:
466
70
.
34.
Deng
L
,
Liang
H
,
Xu
M
,
Yang
X
,
Burnette
B
,
Arina
A
, et al
.
STING-dependent cytosolic DNA sensing promotes radiation-induced type I interferon-dependent antitumor immunity in immunogenic tumors
.
Immunity
2014
;
41
:
843
52
.
35.
Cance
JC
,
Crozat
K
,
Dalod
M
,
Mattiuz
R
.
Are conventional type 1 dendritic cells critical for protective antitumor immunity and how?
Front Immunol
2019
;
10
:
9
.
36.
Li
F
,
Li
C
,
Cai
X
,
Xie
Z
,
Zhou
L
,
Cheng
B
, et al
.
The association between CD8+ tumor-infiltrating lymphocytes and the clinical outcome of cancer immunotherapy: a systematic review and meta-analysis
.
EClinicalMedicine
2021
;
41
:
101134
.
37.
Nersesian
S
,
Schwartz
SL
,
Grantham
SR
,
MacLean
LK
,
Lee
SN
,
Pugh-Toole
M
, et al
.
NK cell infiltration is associated with improved overall survival in solid cancers: a systematic review and meta-analysis
.
Transl Oncol
2021
;
14
:
100930
.
38.
Lu
S
,
Stein
JE
,
Rimm
DL
,
Wang
DW
,
Bell
JM
,
Johnson
DB
, et al
.
Comparison of biomarker modalities for predicting response to PD-1/PD-L1 checkpoint blockade: a systematic review and meta-analysis
.
JAMA Oncol
2019
;
5
:
1195
204
.
39.
Murray
PJ
,
Allen
JE
,
Biswas
SK
,
Fisher
EA
,
Gilroy
DW
,
Goerdt
S
, et al
.
Macrophage activation and polarization: nomenclature and experimental guidelines
.
Immunity
2014
;
41
:
14
20
.
40.
Biswas
SK
,
Mantovani
A
.
Macrophage plasticity and interaction with lymphocyte subsets: cancer as a paradigm
.
Nat Immunol
2010
;
11
:
889
96
.
41.
Qian
BZ
,
Pollard
JW
.
Macrophage diversity enhances tumor progression and metastasis
.
Cell
2010
;
141
;
39
51
.
42.
Kwak
T
,
Wang
F
,
Deng
H
,
Condamine
T
,
Kumar
V
,
Perego
M
, et al
.
Distinct populations of immune-suppressive macrophages differentiate from monocytic myeloid-derived suppressor cells in cancer
.
Cell Rep
2020
;
33
:
108571
.
43.
Li
T
,
Chen
ZJ
.
The cGAS-cGAMP-STI NG pathway connects DNA damage to inflammation, senescence, and cancer
.
J Exp Med
2018
;
215
:
1287
99
.
44.
Zhang
Q
,
Wang
J
,
Yadav
DK
,
Bai
X
,
Liang
T
.
Glucose metabolism: the metabolic signature of tumor-associated macrophage
.
Front Immunol
2021
;
12
:
702580
.
45.
D'Arcy
BM
,
Swingle
MR
,
Papke
CM
,
Abney
KA
,
Bouska
ES
,
Prakash
A
, et al
.
The antitumor drug LB-100 is a catalytic inhibitor of protein phosphatase 2A (PPP2CA) and 5 (PPP5C) coordinating with the active-site catalytic metals in PPP5C
.
Mol Cancer Ther
2019
;
18
:
556
66
.
46.
Ramos
F
,
Villoria
MT
,
Alonso-Rodríguez
E
,
Clemente-Blanco
A
.
Role of protein phosphatases PP1, PP2A, PP4 and Cdc14 in the DNA damage response
.
Cell Stress
2019
;
3
:
70
85
.
47.
Long
L
,
Deng
Y
,
Yao
F
,
Guan
D
,
Feng
Y
,
Jiang
H
, et al
.
Recruitment of phosphatase PP2A by RACK1 adaptor protein deactivates transcription factor IRF3 and limits Type I interferon signaling
.
Immunity
2014
;
40
:
515
29
.
48.
Ho
WS
,
Mondal
I
,
Xu
B
,
Das
O
,
Sun
RC
,
Chiou
P
, et al
.
PP2Ac/STRN4 negatively regulates STING-Type I interferon signaling in tumor-associated macrophages
.
J Clin Invest
2023
;
133
:
e162139
.
49.
Benci
JL
,
Johnson
LR
,
Choa
R
,
Xu
Y
,
Qiu
J
,
Zhou
Z
, et al
.
Opposing functions of interferon coordinate adaptive and innate immune responses to cancer immune checkpoint blockade
.
Cell
2019
;
178
:
933
48
.
50.
Benci
JL
,
Xu
B
,
Qiu
Y
,
Wu
TJ
,
Dada
H
,
Twyman-Saint Victor
C
, et al
.
Tumor interferon signaling regulates a multigenic resistance program to immune checkpoint blockade
.
Cell
2016
;
167
:
1540
54
.
51.
Du
Z
,
Cai
C
,
Sims
M
,
Boop
FA
,
Davidoff
AM
,
Pfeffer
LM
.
The effects of type I interferon on glioblastoma cancer stem cells
.
Biochem Biophys Res Commun
2017
;
491
:
343
8
.
52.
Ohkuri
T
,
Ghosh
A
,
Kosaka
A
,
Zhu
J
,
Ikeura
M
,
David
M
, et al
.
STING contributes to antiglioma immunity via triggering type I IFN signals in the tumor microenvironment
.
Cancer Immunol Res
2014
;
2
:
1199
208
.
53.
Silginer
M
,
Nagy
S
,
Happold
C
,
Schneider
H
,
Weller
M
,
Roth
P
.
Autocrine activation of the IFN signaling pathway may promote immune escape in glioblastoma
.
Neuro Oncol
2017
;
19
:
1338
49
.
54.
Khan
S
,
Mahalingam
R
,
Sen
S
,
Martinez-Ledesma
E
,
Khan
A
,
Gandy
K
, et al
.
Intrinsic interferon signaling regulates the cell death and mesenchymal phenotype of glioblastoma stem cells
.
Cancers (Basel)
2021
;
13
:
5284
.
55.
Vainonen
JP
,
Momeny
M
,
Westermarck
J
.
Druggable cancer phosphatases
.
Sci Transl Med
2021
;
13
:
eabe2967
.
56.
Leonard
D
,
Huang
W
,
Izadmehr
S
,
O'Connor
CM
,
Wiredja
DD
,
Wang
Z
, et al
.
Selective PP2A enhancement through biased heterotrimer stabilization
.
Cell
2020
;
181
:
688
701
.