Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

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

Https

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

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2025 Jul 3;16(1):6123.
doi: 10.1038/s41467-025-59021-9.

Tumor site-directed A1R expression enhances CAR T cell function and improves efficacy against solid tumors

Affiliations

Tumor site-directed A1R expression enhances CAR T cell function and improves efficacy against solid tumors

Kevin Sek et al. Nat Commun. .

Abstract

The efficacy of Chimeric Antigen Receptor T cells against solid tumors is limited by immunosuppressive factors in the tumor microenvironment including adenosine, which suppresses Chimeric Antigen Receptor T cells through activation of the A2A receptor. To overcome this, Chimeric Antigen Receptor T cells are engineered to express A1 receptor, a receptor that signals inversely to A2A receptor. Using murine and human Chimeric Antigen Receptor T cells, constitutive A1 receptor overexpression significantly enhances Chimeric Antigen Receptor T cell effector function albeit at the expense of Chimeric Antigen Receptor T cell persistence. Through a CRISPR/Cas9 homology directed repair "knock-in" approach we demonstrate that Chimeric Antigen Receptor T cells engineered to express A1 receptor in a tumor-localized manner, enhances anti-tumor therapeutic efficacy. This is dependent on the transcription factor IRF8 and is transcriptionally unique when compared to A2A receptor deletion. This data provides a novel approach for enhancing Chimeric Antigen Receptor T cell efficacy in solid tumors and provides proof of principle for site-directed expression of factors that promote effector T cell differentiation.

PubMed Disclaimer

Conflict of interest statement

Competing interests: P.A.B. declares the following conflicts; Research funding: Gilead, Bristol Myers Squibb. P.K.D. declares the following conflicts: research funding from Myeloid Therapeutics, Prescient Therapeutics and Bristol-Myers-Squibb. K.S. declares the following conflicts: Research funding: Prescient Therapeutics. I.A.P. declares the following conflicts; Research funding: AstraZeneca, Bristol Myers Squibb. A.B. declares the following conflicts: Founder of INSiGENe Pty Ltd, which is related to this work. Co-founder and director of Respiradigm Pty Ltd that is unrelated to this work. All other authors declare no conflict of interest.

Figures

Fig. 1
Fig. 1. A1R expression enhances anti-tumor functionality and effector differentiation of mouse and human CAR T cells.
A Schematic of mouse or human adenosine receptors, A1R and A3R, tagged with mCherry/NGFR expression markers. B qRT-PCR cycle threshold normalized against Rpl32, n = 3 independent experiments. C cAMP response to ADO of mouse CAR T cells primed with FSK (1 μM), n = 2 independent experiments. D Cytokine production in mouse anti-HER2 CAR T cells after coculture with E0771-HER2 or 24JK-HER2 tumor cell lines for 16 h. E Cytokine production by human anti-Lewis Y CAR T cells after coculture with OVCAR-3 or MCF-7 tumor cell lines for 16 h in the presence or absence of an A1R antagonist (DPCPX, 100 nM). F Cytokine production by mouse anti-HER2 CAR T cells after serial coculture with E0771-HER2 tumors every 24 h. G FACS plots and quantification of CAR T cells after 72-hour serial coculture. H GSEA plot of pathways from the mSigDB database based on RNA-seq of tumor stimulated A1R or A3R CAR T cells versus Control. IK Human anti-Lewis Y A1R CAR T cells stimulated with OVCAR-3 tumor cells for 16 h in the presence or absence of an A1R inhibitor (100 nM), DPCPX. I FACS plots, JK marker expression in CD8+/CD4+ CAR T cells. L Differentially expressed genes of OVCAR-3 stimulated human anti-Lewis Y CAR T cells. M GSEA of PID and KEGG datasets of stimulated hA1R CAR T cells versus control. N EnrichR plots of differentially expressed genes in both mouse and human A1R CAR T cells versus control for positively enriched genesets. P values were determined using the Fisher’s exact test/hypergeometric test based on the enrichR package. ****p < 0.0001, ***p < 0.001, **p < 0.01, *p < 0.05, mean ± SD (BE, G, J, K). One-way ANOVA or (F) two-way ANOVA. D, F, G, H All mouse data is representative of at least 3 independent experiments, with the bulk RNA-seq experiment performed with 2 technical replicates. E, IL All human data is representative of at least 3 independent experiments. Human bulk RNA-seq was performed with 3 technical replicates. H, M GSEA P-value estimation is based on an adaptive multi-level split Monte-Carlo scheme based off the fgsea package.
Fig. 2
Fig. 2. Constitutive A1R expression drives terminal differentiation of CAR T cells.
C57BL6-HER2 transgenic mice bearing orthotopic E0771-HER2 tumors were pre-treated with 4 Gy of total body irradiation prior to adoptive transfer of 2 doses of 10e6 anti-HER2 CAR T cells. 5 doses of 50,000 IU of IL-2 were injected i.p every 24 h. A Tumor growth curve, representative of 2 independent experiments with n = 4-6 mice per group. B CAR T cell counts in spleen and tumors from n = 5-6 mice except for non-treated group (n = 2 mice). Data represented as the mean ± SEM (A, B). C Quantification of %TSCM (CD45RO-CD45RA+CD27+CD62L+) in CD4+ and CD8+ NGFR+ human anti-Lewis Y CAR T cells shown as pooled data of means ± SEM from n = 4 independent healthy donors. D CD69 and LAG3 expression in human CAR T cells cultured with or without A1R antagonist DPCPX (1 μM) in CD8+/CD4+ CAR T cells prior to tumor stimulation, representative of n = 4 donors. E Volcano plot of DEGs for unstimulated CAR T cells. F GSEA of DEGs enriched in A1R CAR T cells using the KEGG and Sade-Feldman single-cell RNA-seq datasets (GSE120575). G Correlation of significant DEGs between tumor stimulated and unstimulated hA1R vs Ctrl CAR T cells. H CD8+ T cell meta-clusters from integrated atlas of single cell RNA-seq experiments across 21 tumor types and cells from 316 donors. Derived A1R signature genes up or down-regulated are overlayed onto meta-clusters, I GSEA of up or down-regulated genes from each meta-cluster for unstimulated hA1R CAR T cells vs control. J Quantification of gene expression in each meta-cluster GSEA for select core-enriched genes from the A1R signature as defined by ref. . K Heatmap of significant DEGs from the A1R signature upregulated or downregulated with a logFC≥1 or logFC≤1 respectively between tumor stimulated and unstimulated hA1R vs Ctrl CAR T cells. ****p < 0.0001, ***p < 0.001, **p < 0.01, *p < 0.05. C paired two-sided t-test (B) One-way ANOVA or (A) two-way ANOVA. Human bulk RNA-seq was performed on a single donor with 3 technical replicates. F, I GSEA P-value estimation is based on an adaptive multi-level split Monte-Carlo scheme based off the fgsea package.
Fig. 3
Fig. 3. CAR T cells engineered to express A1R under the control of the NR4A2 promoter exhibit enhanced CAR T cell function without loss of memory precursors.
A Cytokine secretion of anti-Lewis Y CAR T cells that were pre-conditioned in culture with DPCPX (100 nM) prior to co-culture with OVCAR-3 tumors, representative with mean ± SD of triplicate cultures. B CRISPR/HDR template with homology arms for the NR4A2 gene, to knock-in payload transgene under the control of the NR4A2 promoter. C RNA-seq counts per million (CPM) of ADORA1 gene after serial coculture with OVCAR-3 tumors, represented as the mean ± SD (D) Cytokine secretion and E intracellular cytokine staining in CAR T cells after serial coculture with OVCAR-3 tumors, represented as the mean ± SD of triplicate cultures. F CD69 and LAG3 expression in CD8+/CD4+ CAR T cells without tumor stimulation. G Quantification of %TSCM (CD45RO-CD45RA+CD27+CD62L+) in CD4+/CD8+ human anti-Lewis Y CAR T cells shown as pooled data of means ± SEM from 3 donors. H Expression of markers following coculture with OVCAR-3 tumor cells, represented as the mean ± SD of triplicate cultures. I Cytokine secretion by anti-HER2 CAR T cells against MCF-7 breast cancer after serial coculture, represented as the mean ± SD of triplicate cultures. JK Differential accessibility peaks from ATAC-sequencing in J unstimulated or (K) OVCAR-3 tumor stimulated human CAR T cells. L HOMER motif analyses of enriched motifs in NR4A2/hA1R CAR T cells. M PCA plot for each round of coculture. N Quantificaiton of DEGs with each round of stimulation. O Magnitude of up or downregulated DEGs from the previously identified A1R signature in NR4A2/hA1R CAR T cells. Box-plot defined by box (1st and 3rd interquartile range), median-line, whiskers extending to points within 1.5x interquartile range. PQ GSEA of gene-sets from mSigDB Hallmarks, KEGG and Sade-feldman gene signatures (GSE120575) for P hA1R vs Control or (Q) hA1R vs NR4A2KO CAR T cell groups after first stimulation with tumor cells. ****p < 0.0001, ***p < 0.001, **p < 0.01, *p < 0.05. A, F, G, K, O One-way ANOVA or (D, E, H, I, L, M, N) two-way ANOVA. C, JQ Human bulk ATAC-seq and RNA-seq was performed in independent experiments, 3 technical replicates each.
Fig. 4
Fig. 4. Tumor site specific expression of hA1R enhances CAR T cell therapeutic efficacy against solid tumors in vivo.
NSG mice were injected in the fourth mammary fat pad or sub-cutaneously with 1.25 × 106 HER-2Low MDA-MB231 breast or 5 × 106 LewisY+ OVCAR-3 ovarian tumor cells respectively. Once tumors were established (15-20 mm2), mice were then irradiated (1 Gy) and treated with one dose of 5 × 106 anti-HER2 or 15 × 106 anti-Lewis Y CAR T cells, respectively. Mice were supplemented with 50,000 IU of IL-2 on days 0-4 post treatment. A Tumor growth, data shown as means ± SEM of n = 5–6 mice per group (B) Tumor growth, data shown as means ± SEM of n = 7 mice per group and C individual tumor growth curves. D Waterfall plot of % change in tumor size on Day 28 post therapy. EG Counts of CD8+ and CD4+ FLAG+ CAR T cells in E tumor and spleen at (F) D17 post therapy and G D28 post therapy. H Quantification of %TSCM CAR T cells in spleen D17 post therapy. BH Data represented as the mean ± SEM of n = 6 mice per group. I TSNE plots and unbiased clustering of tumor infiltrating CD8+FLAG+ CAR T cells analyzed by flow cytometry at day 17 post therapy. J Heatmap and K quantification of marker expression sorted by unbiased clustering in tumor at D17 post therapy. Box-plot defined by box (interquartile range, 1st and 3rd quartiles), median-line, whiskers extending to SD. L Quantification of memory subsets of CD8+ FLAG+ TPEX (PD-1INTTCF-1+CD62L+/−), TEFF (PD-1INTTCF-1) and TEX (PD-1HITCF-1) in tumor and spleen at D17 post therapy. ****p < 0.0001, ***p < 0.001, **p < 0.01, *p < 0.05. EH, L one-way anova or (A, B, K) two-way ANOVA. A, B Statistics shown are NR4A2/hA1R versus NR4A2KO groups. Data representative of 2 (a, n = 5–6 mice per group) or 3 independent experiments (B, n = 7 mice per group).
Fig. 5
Fig. 5. Enhanced effector function of A1R expressing CAR T cells drives IRF8 dependent distinct transcriptional pathways compared to A2AR deletion.
Bulk RNA-seq was performed on anti-Lewis Y CAR T cells stimulated for 72 h with OVCAR-3 tumors in the presence or absence of pan-adenosine receptor agonist, NECA (10 μM). A PCA plot, (B) DEGs for of CAR T cells stimulated with tumors in the absence of NECA (top). Venn Diagram showing overlap of up or down-regulated DEGs between NECA vs no NECA treated CAR T cells (bottom). C GSEA of A1R gene signature for A2ARKO and NR4A2/hA1R CAR T cells versus NR4A2KO controls. D Network dendrogram and gene-coexpression modules identified using WGCNA of human CAR T cells after serial coculture with OVCAR-3 tumors over 72 h. E Eigengene score of individual groups at each round of stimulation for the red module, two-way ANOVA, n = 3 technical replicates per group, data represented as the mean ± SD. F Module-trait relationship of key gene signatures against modules eigengene scores. G Heatmap of 221 genes in the red module after 1 round of coculture. H GSEA of the genes identified from the red module using EnrichR. P values were determined using the Fisher’s exact test/ hypergeometric test based off the enrichR package. I Ranked connectivity scores for transcription factors within the red module (J) Network plot of IRF8 and 1st neighbors in the red module. K CPM counts of IRF8 at each stimulation timepoint, represented by mean ± SD of triplicates. LN IRF8 knockout NR4A2/hA1R CAR T cells were generated using CRISPR/Cas9 editing and stimulated with OVCAR-3 tumor cells overnight. L Cytokine secretion, M, N activation (CD69+CD62L-) and memory (CD62L+CD69-) phenotype. LN Data shown as means ± SD of triplicate cultures and representative of 2 independent experiments with 2 healthy donors. ****p < 0.0001, ***p < 0.001, **p < 0.01, *p < 0.05. G, K two-way ANOVA or (L, N) one-way ANOVA. Human bulk RNA-seq was performed on a single donor with 3 technical replicates. C GSEA P-value estimation is based on an adaptive multi-level split Monte-Carlo scheme based off the fgsea package.

References

    1. Melenhorst, J. J. et al. Decade-long leukaemia remissions with persistence of CD4(+) CAR T cells. Nature602, 503–509 (2022). - PMC - PubMed
    1. Fraietta, J. A. et al. Determinants of response and resistance to CD19 chimeric antigen receptor (CAR) T cell therapy of chronic lymphocytic leukemia. Nat. Med.24, 563–571 (2018). - PMC - PubMed
    1. Kershaw, M. H., Westwood, J. A. & Darcy, P. K. Gene-engineered T cells for cancer therapy. Nat. Rev. Cancer13, 525–541 (2013). - PubMed
    1. Joyce, J. A. & Fearon, D. T. T cell exclusion, immune privilege, and the tumor microenvironment. Science348, 74–80 (2015). - PubMed
    1. Bansal, R. & Reshef, R. Revving the CAR - Combination strategies to enhance CAR T cell effectiveness. Blood Rev.45, 100695 (2021). - PubMed

MeSH terms

Substances

LinkOut - more resources