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
. 2023 Feb 2;14(1):86.
doi: 10.1038/s41467-022-35583-w.

Deletion of SNX9 alleviates CD8 T cell exhaustion for effective cellular cancer immunotherapy

Affiliations

Deletion of SNX9 alleviates CD8 T cell exhaustion for effective cellular cancer immunotherapy

Marcel P Trefny et al. Nat Commun. .

Abstract

Tumor-specific T cells are frequently exhausted by chronic antigenic stimulation. We here report on a human antigen-specific ex vivo model to explore new therapeutic options for T cell immunotherapies. T cells generated with this model resemble tumor-infiltrating exhausted T cells on a phenotypic and transcriptional level. Using a targeted pooled CRISPR-Cas9 screen and individual gene knockout validation experiments, we uncover sorting nexin-9 (SNX9) as a mediator of T cell exhaustion. Upon TCR/CD28 stimulation, deletion of SNX9 in CD8 T cells decreases PLCγ1, Ca2+, and NFATc2-mediated T cell signaling and reduces expression of NR4A1/3 and TOX. SNX9 knockout enhances memory differentiation and IFNγ secretion of adoptively transferred T cells and results in improved anti-tumor efficacy of human chimeric antigen receptor T cells in vivo. Our findings highlight that targeting SNX9 is a strategy to prevent T cell exhaustion and enhance anti-tumor immunity.

PubMed Disclaimer

Conflict of interest statement

H.L., A.Zippelius received research funding from Bristol-Myers Squibb and Novartis. A.Zippelius received consulting/advisor fees from Bristol-Myers Squibb, Merck Sharp & Dohme, Hoffmann–La Roche, NBE Therapeutics, Secarna, ACM Pharma, and Hookipa, and maintains further non-commercial research agreements with Secarna, Hookipa, Roche and Beyondsprings. L.T.J. is a co-founder of, holds equity in and has a sponsored research agreement with Cimeio Therapeutics AG. A.Zippelius and M.P.T. have a pending patent application (PCT/EP2022/077391, “Targeting SNX9 rescues recombinant T cell in adoptive therapy”). The remaining authors declare no competing interests.

Figures

Fig. 1
Fig. 1. Repetitive antigen-specific stimulation ex vivo of T cells results in exhaustion.
a Scheme representing the principle of the exhaustion model. b Representative co-expression of PD-1 and TIM-3 measured by antibody staining and flow cytometry. c PD-1 expression measured by flow cytometry after antibody staining on day 12 after the first stimulation. n = 10 donors from n = 5 experiments except for Teff n = 4 of n = 2 experiments. d Degranulation measured by CD107a surface exposure during re-stimulation of all cells after 13 days of culture. n = 12 donors of n = 6 experiments except for Teff n = 6 of n = 3 experiments. e–f Representative flow cytometry plot and quantification of IFNγ and TNFα production measured by intracellular cytokine staining. n = 14 donors from n = 7 experiments except for Teff n = 8 of n = 4 experiments. g Specific lysis of T2-Luc+ tumor cells pulsed with peptide after 5 hours of co-incubation with T cells measured by luminescence intensity. n = 6 donors for Trest, n = 13 for Tex and n = 8 for Teff. h IFNγ secretion measured by ELISA after 4 days co-culture of Teff or Tex with MDA-MB-231 cells loaded with 100 nM NY-ESO-1-9V peptide in presence of either 10 μg/ml human IgG4 isotype control or anti-PD-1 antibody (Nivolumab). Statistics is a paired 2-way-ANOVA with Holm-Sidak correction with n = 5 donors. i Heatmap of all differentially regulated genes between the four conditions showing row-scaled log-cpms and clustered using k-means (numbers indicated on the left). Selected genes associated with exhaustion or functionality are labeled on the right. n = 4 donors. j Gene set enrichment analysis comparing mRNA expression of the different conditions for the Zheng. et al PanCancer T cell states. Color represents the mean log fold change and the size the -log10 false discovery rate (FDR). c-d; f-g 1-way ANOVA statistics with Holm-Sidak correction, Mean and SD are shown. * p < 0.05, ** p < 0.01, *** p < 0.001, **** p < 0.0001. Source data and exact p-values are provided as a Source Data file.
Fig. 2
Fig. 2. SNX9 is a potential mediator of T cell exhaustion.
a Schematic description of the targeted-pooled CRISPR-Cas9 KO screen. b Mean log2 fold changes for gRNA enrichment in the pooled CRISPR/Cas9 screen. Dots represent the mean enrichment of all 5 guides for each donor biological replicate. c Representative flow cytometry plots and quantification of SNX9 expression measured by antibody staining in flow cytometry of human CD8 T cells of the ex vivo model with the different conditions. Mean and SD are shown. n = 11 donors of n = 3 experiments. Statistics are a 1-way ANOVA with Holm-Sidak correction. d Representative flow cytometry plots of SNX9 vs. PD-1 and TIM-3 expression in tumor-infiltrating CD8 T cells of a non-small cell lung cancer (NSCLC) patient. e TIM-3 expression measured by flow cytometry in SNX9 positive vs. SNX9 negative in intratumoral PD-1+ CD8 T cells of NSCLC patients. Statistics are a two-tailed paired t-test. Mean and SD are shown. n = 10. f Percentage of SNX9+ cells among CD8 T cells for each sample before treatment with ICI. Statistic is a Mann-Whitney test (non-normal distribution). We used 1 log normalized counts as the threshold for SNX9 positivity. Samples with less than ten cells in either population were excluded, resulting in n = 10 nonresponders and n = 9 responders. c, e, f Mean and SD are shown. * p < 0.05, ** p < 0.01, *** p < 0.00 and **** p < 0.0001. Source data and exact p-values are provided as a Source Data file.
Fig. 3
Fig. 3. SNX9 KO improves effector functions of exhausted T cells and dampens the NFAT-NR4A1/3-TOX axis.
a Schematic procedure to generate SNX9 KO T cells. b IFNγ secretion by ELISA of Teff and Tex in response to re-stimulation. n = 10 donors (Teff) and 8 (Tex). c Top row: representative confocal images of Tex co-cultured with T2 tumor cells for actin (n = 6), and LAMP1/Perforin (n = 3). Bottom row: example images of CD28-EGFP (n = 6) or TCRz-EGFP (n = 5) fusion proteins, or anti-CD45 (n = 6) and SNX9 in Teff. All images are provided in the Source Data file. d Nuclear translocation measured by image cytometry of NFATc2 in Teff upon anti-CD3/28 stimulation. n = 7 donors of n = 2 experiments. e Area under the curve (AUC) of Calbryte520-AM Ca2+ flux upon stimulation. n = 8 donor of n = 3 experiments. f RT-qPCR quantifications of mRNA for NR4A1 and NR4A3 in cells on day 6 of the Tex culture. n = 7 donors of n = 2 experiments. g Antibody staining (delta unstained) of TOX in Tex. n = 8 donors of n = 4 experiments. h Expression of CCR7 measured by flow cytometry antibody staining in Tex. n = 8 of n = 3 experiments. b, e-h Statistics are a two-sided paired t-tests. i Geometric mean CD25 upregulation relative to unstimulated cells for Teff co-cultured with T2 wt or T2 CD80 CD86 KO (T2 KO) cells. 10 μg/ml human IgG1 or 10 μg/ml anti-CTLA4 blocking antibody (ipilimumab) was added. n = 6 donor replicates of n = 2 experiments. j CD25 geometric mean intensity of stimulated T cells with or without SNX9 KO. n = 7 donors of n = 2 experiments. k Delta unstimulated geometric mean of phospho-PLCγ1-Tyr783 fluorescence intensity. n = 4 donors. d, i-k Statistics are paired 2-way ANOVA with Holm-Sidak correction. * p < 0.05, ** p < 0.01, *** p < 0.001 and **** p < 0.0001. Source data and exact p-values are provided as a Source Data file.
Fig. 4
Fig. 4. Snx9 KO improves anti-tumor efficacy and reduces terminal exhaustion in vivo.
a Schematic experimental setup of OTI Snx9 KO generation and transfer to MC38-OVA-bearing mice. b Mean and SEM of MC38-OVA tumor volumes in C57BL/6 in n = 6 mice per condition. Curves are shown until the first mouse per group reaches the humane endpoint. Statistics are pairwise 2-way ANOVAs followed by Bonferroni correction. Representative of n = 3 experiments. c Percentage of PD-1high Tim3+ OTI cells in MC38-OVA tumors in C57BL/6 mice on the indicated timepoints. n = 6 intergenic and n = 4 Snx9 KO. Statistics are 2-way ANOVA with Holm-Sidak correction. d UMAP of single-cell RNA sequencing data from OTI cells isolated from MC38-OVA tumors 13 days post-transfer. Intergenic n = 3405 cells of 5 mice and Snx9 KO n = 3612 cells of 6 mice. e Average expression of selected marker genes (color). Size = percentage of cells with detected expression for each cluster. f Proportions of each cluster in the intergenic versus the Snx9 KO sample (n = 1 from 5–6 pooled mice per condition). g Selected differentially expressed genes between intergenic and OTI Snx9 KO cells per cluster. Size indicates the -log10 adjusted p-value and color the mean log2 fold change. Statistics were derived by Seurat’s FindMarkers function. h Quantification of endogenous CD8 T cells (left) and cDC1s (right) in MC38-OVA tumors 3 days post transfer of OTI cells. Statistics are unpaired t-tests. n = 6 mice per group (i) Serum levels of IFNγ and Il-10 for days 2,8 and 15. Limit of detection (LOD) is indicated for IFNγ. Statistics are unpaired 2-way ANOVA with Holm-Sidak correction. n = 6 mice per group. j Tumor volume (mean and SEM) of NSG mice with subcutaneous MC38-OVA tumors with a transfer of OTI cells at day 12 postinjection. n = 6 mice per OTI condition, n = 4 for untreated. Statistics are pairwise 2-way ANOVAs followed by Bonferroni correction * p < 0.05, ** p < 0.01, *** p < 0.001. c, h-i Mean and SD are shown. Source data and exact p-values are provided as a Source Data file.
Fig. 5
Fig. 5. Deletion of SNX9 improves CAR T cell anti-tumor efficacy.
a Schematic representation of the CAR T cell transfer experiments. Healthy donor human CD8 T cells are stimulated ex vivo and lentivirally transduced with an anti-human-CD19(FMC63vH)-CD28-CD3zeta-T2A-copGFP CAR construct and electroporated with Cas9-crRNA-tracrRNA complexes to generate SNX9 KO cells and intergenic controls. These cells are then transferred to NSG mice with subcutaneous Raji tumors (CD19+). b Tumor volume in mm3 of NSG mice treated 3 days post Raji tumor injection by i.v. transfer of 0.5 Mio human CD8 anti-CD19-28z CAR T cells with or without SNX9 KO (mean and SEM). Statistics are pairwise 2-way ANOVAs followed by Bonferroni correction. n = 8 animals for untreated of n = 2 experiments. n = 7 mice for CART-treated mice of n = 1 experiment. Experiment was replicated with similar results with higher CART numbers. c Survival of the NSG mice in 5b until humane endpoint of 1500mm3 tumor size. Statistics are log-rank Mantel-Cox tests followed by Bonferroni correction. (b and c): n = 8 for untreated, n = 7 for intergenic and SNX9 KO CAR T conditions. d Human cytokines measured by Legendplex (Biolegend) in the serum of Raji-bearing NSG mice treated with anti-CD19-28z CAR T cells with and without SNX9 KO. Statistics are paired-2-way ANOVA with Holm-Sidak correction. n = 6 mice per condition. Mean and SD are shown. e Tumor volume in mm3 (mean and SEM) of NSG mice treated 3 days post Raji tumor injection by i.v. transfer of 1 Mio human CD8+ CD28 KO anti-CD19-BBz CAR T cells with or without SNX9 KO. n = 6 for intergenic and SNX9 KO, n = 8 for untreated. Statistics are 2-way ANOVAs followed by Bonferroni correction. * p < 0.05, ** p < 0.01, *** p < 0.001. Source data and exact p-values are provided as a Source Data file.

References

    1. Philip M, Schietinger A. CD8+ T cell differentiation and dysfunction in cancer. Nat. Rev. Immunol. 2022;22:209–223. doi: 10.1038/s41577-021-00574-3. - DOI - PMC - PubMed
    1. Thommen DS, Schumacher TN. T Cell Dysfunction in Cancer. Cancer Cell. 2018;33:547–562. doi: 10.1016/j.ccell.2018.03.012. - DOI - PMC - PubMed
    1. Baitsch L, et al. Exhaustion of tumor-specific CD8+ T cells in metastases from melanoma patients. J. Clin. Invest. 2011;121:2350–2360. doi: 10.1172/JCI46102. - DOI - PMC - PubMed
    1. Zippelius A, et al. Effector Function of Human Tumor-Specific CD8 T Cells in Melanoma Lesions: A State of Local Functional Tolerance. Cancer Res. 2004;64:2865–2873. doi: 10.1158/0008-5472.CAN-03-3066. - DOI - PubMed
    1. Kim PS, Ahmed R. Features of responding T cells in cancer and chronic infection. Curr. Opin. Immunol. 2010;22:223–230. doi: 10.1016/j.coi.2010.02.005. - DOI - PMC - PubMed

Publication types