PD-1 blockade restores helper activity of tumor-infiltrating, exhausted PD-1hiCD39+ CD4 T cells

Camille-Charlotte Balança, Anna Salvioni, Clara-Maria Scarlata, Marie Michelas, Carlos Martinez-Gomez, Carlos Gomez-Roca, Victor Sarradin, Marie Tosolini, Carine Valle, Frédéric Pont, Gwénaël Ferron, Laurence Gladieff, Sébastien Vergez, Agnès Dupret-Bories, Eliane Mery, Philippe Rochaix, Jean-Jacques Fournié, Jean-Pierre Delord, Christel Devaud, Alejandra Martinez, Maha Ayyoub, Camille-Charlotte Balança, Anna Salvioni, Clara-Maria Scarlata, Marie Michelas, Carlos Martinez-Gomez, Carlos Gomez-Roca, Victor Sarradin, Marie Tosolini, Carine Valle, Frédéric Pont, Gwénaël Ferron, Laurence Gladieff, Sébastien Vergez, Agnès Dupret-Bories, Eliane Mery, Philippe Rochaix, Jean-Jacques Fournié, Jean-Pierre Delord, Christel Devaud, Alejandra Martinez, Maha Ayyoub

Abstract

Tumor antigen-specific CD4 T cells accumulate at tumor sites, evoking their involvement in antitumor effector functions in situ. Contrary to CD8 cytotoxic T lymphocyte exhaustion, that of CD4 T cells remains poorly appreciated. Here, using phenotypic, transcriptomic, and functional approaches, we characterized CD4 T cell exhaustion in patients with head and neck, cervical, and ovarian cancer. We identified a CD4 tumor-infiltrating lymphocyte (TIL) population, defined by high PD-1 and CD39 expression, which contained high proportions of cytokine-producing cells, although the quantity of cytokines produced by these cells was low, evoking an exhausted state. Terminal exhaustion of CD4 TILs was instated regardless of TIM-3 expression, suggesting divergence with CD8 T cell exhaustion. scRNA-Seq and further phenotypic analyses uncovered similarities with the CD8 T cell exhaustion program. In particular, PD-1hiCD39+ CD4 TILs expressed the exhaustion transcription factor TOX and the chemokine CXCL13 and were tumor antigen specific. In vitro, PD-1 blockade enhanced CD4 TIL activation, as evidenced by increased CD154 expression and cytokine secretion, leading to improved dendritic cell maturation and consequently higher tumor-specific CD8 T cell proliferation. Our data identify exhausted CD4 TILs as players in responsiveness to immune checkpoint blockade.

Keywords: Cancer immunotherapy; Immunology; T cells.

Conflict of interest statement

Conflict of interest: CGR received a research grant from Bristol-Myers Squibb; received speakers’ bureau honoraria from Bristol-Myers Squibb, Hoffmann-La Roche, and Pierre Fabre; and is a consultant/advisory board member for Bristol-Myers Squibb. JPD received speakers’ bureau honoraria from Roche, Merck Sharp & Dohme, Bristol-Myers Squibb, and AstraZeneca. PR received research support from Roche Diagnostic/Ventana and MSD. MA received a research grant from Roche/Genentech (imCORE), received speakers’ bureau honoraria from AstraZeneca and Bristol-Myers Squibb, and is a consultant/advisory board member for AstraZeneca.

Figures

Figure 1. CD39 expression in CD4 +…
Figure 1. CD39 expression in CD4+ Tconv TILs.
Isolated CD4+ and CD8+ TILs were stained ex vivo and analyzed by flow cytometry. (A) Dot plot shows PD-1 versus CD39 expression in cells gated on memory CD4 Tconvs as shown in Supplemental Figure 1A. Proportions of CD39+ cells (center, n = 21) and correlation between the proportion of PD-1+ and CD39+ (right, n = 19) in CD4 Tconvs. (B) Dot plot shows PD-1 expression and gates defining PD-1neg, PD-1int, and PD-1hi cells within CD4 Tconvs. Proportions of PD-1neg, PD-1int, and PD-1hi cells in CD4 Tconvs (n = 21). (C and D) Histogram plots in C show TIGIT, CD39, CTLA-4, and TIM-3 expression in PD-1neg, PD-1int, and PD-1hi CD4 Tconvs and proportions are summarized in D (n = 21). (E) Dot plot shows TIM-3 versus CD39 expression in PD-1hi CD4 Tconvs. Proportions of TIM-3–CD39–, TIM-3+CD39–, TIM-3–CD39+, and TIM-3+CD39+ cells among PD-1hi CD4 Tconvs (n = 21) and PD-1hi CD8+ TILs (n = 10). Data are presented as mean ± SD. **P < 0.01; ***P < 0.001; ****P < 0.0001. Pearson’s correlation (A), 2-tailed paired t test or Wilcoxon (D), and 2-tailed unpaired t test (E) were used to compare variables. Tconvs, conventional FOXP3- CD4 T cells; TILs, tumor-infiltrating lymphocytes.
Figure 2. PD-1 hi CD39 + tumor-infiltrating…
Figure 2. PD-1hiCD39+ tumor-infiltrating CD4 Tconvs are functionally exhausted.
(A and B) Isolated CD4+ TILs were stimulated in vitro with PMA/ionomycin and stained and analyzed by flow cytometry. (A) Top left dot plot shows PD-1 versus CD39 expression in CD4 Tconvs. IFN-γ versus TNF-α expression is shown in the indicated CD4 Tconv populations. Proportions of cytokine+ (IFN-γ and/or TNF-α; bottom left) and TNF-α+IFN-γ–, TNF-α–IFN-γ+, and TNF-α+IFN-γ+ cells in PD-1–CD39–, PD-1loCD39–, PD-1hiCD39–, and PD-1hiCD39+ subsets and according to TIM-3 expression (n = 8). (B) Dot plots show CD39 versus IFN-γ or TNF-α in CD4 Tconvs. Numbers in dot plots correspond to MFI of cytokine staining. MFI of IFN-γ and TNF-α staining in IFN-γ+ and TNF-α+ cells, respectively, are summarized for Tconv subpopulations defined as in A (n = 8). Samples in A and B are shown according to the proportion of TIM-3+ cells among PD-1hiCD39+ CD4 Tconv (<50% TIM-3+, square; ≥50% TIM-3+, triangle; unknown, round) (C) CD4 Tconv TIL subsets PD-1–CD39– (gray), PD-1loCD39– (green), PD-1hiCD39– (blue), and PD-1hiCD39+ (red) were sorted, expanded in vitro for 10 days, and restimulated or not with PMA/ionomycin overnight. TNF-α and IFN-γ secretion was quantified by CBA in the supernatant (n = 5). (D) Isolated CD4+ TILs were stained ex vivo and analyzed by flow cytometry. Proportions of TOX+ cells in each Tconv subset are summarized (n = 10). Data are presented as mean ± SD. **P < 0.01; ***P < 0.001; ****P < 0.0001. P values were determined using the Wilcoxon (A and B) and 2-tailed paired t tests (D). Bonferroni’s correction was applied to account for multiple testing in D and significance level was adjusted accordingly (*P < 0.0083; **P < 0.00166; ***P < 0.000166). Tconvs, conventional FOXP3- CD4 T cells; CBA, cytometric bead array; TILs, tumor-infiltrating lymphocytes.
Figure 3. scRNA-Seq of tumor-infiltrating CD4 Tconvs.
Figure 3. scRNA-Seq of tumor-infiltrating CD4 Tconvs.
CD45+ cells isolated ex vivo from 4 head and neck cancer specimens were subjected to scRNA-Seq. (A) t-SNE plot of 2060 CD4 Tconvs color-coded by their associated cluster. (B) Results of differential expression analysis of scRNA-Seq data from ENTPD1+ versus ENTPD1– CD4 Tconvs are shown in a volcano plot. Genes significantly upregulated in ENTPD1+ cells are shown in red and those significantly downregulated are shown in orange. (C) Violin plots showing the expression of CTLA4, HAVCR2, and LAG3 in ENTPD1– and ENTPD1+ cells (left) and t-SNE plots color-coded by levels of expression (gray to red) of ENTPD1, CTLA4, HAVCR2, and LAG3 (right). (D) t-SNE plots showing analysis of TOX exhaustion gene sets (left,ref. , and right, ref. 12) using Single-Cell Signature Explorer Viewer. Tconvs, conventional FOXP3- CD4 T cells; TILs, tumor-infiltrating lymphocytes.
Figure 4. PD-1 hi CD39 + CD4…
Figure 4. PD-1hiCD39+ CD4 Tconv TILs encompass tumor Ag–specific cells and respond to PD-1 blockade by enhancing DC-mediated CD8 T cell proliferation.
(AC) Ex vivo isolated CD4+ TILs from one OC NY-ESO-1–seropositive patient were FACS-sorted into PD-1–CD39–, PD-1hiCD39–, and PD-1hiCD39+ CD4 Tconv (CD3+CD4+CD25-CD127+) subsets and cloned. (A and B) Clonal populations were stained and analyzed by flow cytometry. (A) PD-1 versus CD39 expression in clones representative of the 3 sorted Tconv populations. (B) Proportions of PD-1+ and CD39+ cells are summarized for all clones derived from PD-1–CD39– (n = 54), PD-1hiCD39– (n = 17), and PD-1hiCD39+ (n = 22) subsets. (C) IFN-γ concentration in the supernatant was quantified by ELISA for each clone stimulated with NY-ESO-1 peptide pool (fold increase over unstimulated condition) (n as in B). (DF) Ex vivo CD4+ TILs (± anti–PD-1 mAbs pretreatment) were cocultured with iDCs in the presence or absence of PHA. (D) TNF-α, IFN-γ, IL-2, and IL-12 concentrations were quantified by CBA in the 24-hour supernatants (n = 3). (E) Histogram plots show CD154 expression in CD4 Tconvs after 6-hours stimulation. Proportions of CD154+ cells are summarized (n = 5). (F) Histogram plots show CD86 expression in DCs in day 2 cultures. MFI of CD86 staining are summarized (n = 4). (G) CD4+ TILs from OC NY-ESO-1–seropositive patients (± anti–PD-1 mAbs pretreatment); autologous iDCs and circulating CD8 T cells were cocultured in the presence of NY-ESO-1 peptides, stained with MHC-I/NY-ESO-1 peptide multimers on day 10, and analyzed by flow cytometry. Examples of dot plots show multimer staining and CD8 expression and proportions of multimer+CD8+ cells are summarized (n = 5). A 2-tailed paired t test was used to compare variables (E and F). Bonferroni’s correction was applied to account for multiple testing (E and F) and significance level was adjusted accordingly (*P < 0.016; **P < 0.0032). (E and F). Tconvs, conventional FOXP3- CD4 T cells; TILs, tumor-infiltrating lymphocytes; OC, ovarian cancer; iDCs, immature DCs; CBA, cytometric bead array.

References

    1. Ayyoub M, et al. An immunodominant SSX-2-derived epitope recognized by CD4+ T cells in association with HLA-DR. J Clin Invest. 2004;113(8):1225–1233. doi: 10.1172/JCI20667.
    1. Hamaï A, et al. Human T(H)17 immune cells specific for the tumor antigen MAGE-A3 convert to IFN-γ-secreting cells as they differentiate into effector T cells in vivo. Cancer Res. 2012;72(5):1059–1063. doi: 10.1158/0008-5472.CAN-11-3432.
    1. Borst J, et al. CD4+ T cell help in cancer immunology and immunotherapy. Nat Rev Immunol. 2018;18(10):635–647. doi: 10.1038/s41577-018-0044-0.
    1. Kim H-J, Cantor H. CD4 T-cell subsets and tumor immunity: the helpful and the not-so-helpful. Cancer Immunol Res. 2014;2(2):91–98. doi: 10.1158/2326-6066.CIR-13-0216.
    1. Ayyoub M, et al. CD4+ T effectors specific for the tumor antigen NY-ESO-1 are highly enriched at ovarian cancer sites and coexist with, but are distinct from, tumor-associated Treg. Cancer Immunol Res. 2013;1(5):303–308. doi: 10.1158/2326-6066.CIR-13-0062-T.
    1. Alspach E, et al. MHC-II neoantigens shape tumour immunity and response to immunotherapy. Nature. 2019;574(7780):696–701. doi: 10.1038/s41586-019-1671-8.
    1. Balança C-C, et al. Dual relief of T-lymphocyte proliferation and effector function underlies response to PD-1 blockade in epithelial malignancies. Cancer Immunol Res. 2020;8(7):869–882. doi: 10.1158/2326-6066.CIR-19-0855.
    1. Duhen T, et al. Co-expression of CD39 and CD103 identifies tumor-reactive CD8 T cells in human solid tumors. Nat Commun. 2018;9(1):2724. doi: 10.1038/s41467-018-05072-0.
    1. Thommen DS, et al. A transcriptionally and functionally distinct PD-1+ CD8+ T cell pool with predictive potential in non-small-cell lung cancer treated with PD-1 blockade. Nat Med. 2018;24(7):994–1004. doi: 10.1038/s41591-018-0057-z.
    1. Sade-Feldman M, et al. Defining T cell states associated with response to checkpoint immunotherapy in melanoma. Cell. 2018;175(4):998–1013. doi: 10.1016/j.cell.2018.10.038.
    1. Wherry EJ. T cell exhaustion. Nat Immunol. 2011;12(6):492–499. doi: 10.1038/ni.2035.
    1. Khan O, et al. TOX transcriptionally and epigenetically programs CD8+ T cell exhaustion. Nature. 2019;571(7764):211–218. doi: 10.1038/s41586-019-1325-x.
    1. Scott AC, et al. TOX is a critical regulator of tumour-specific T cell differentiation. Nature. 2019;571(7764):270–274. doi: 10.1038/s41586-019-1324-y.
    1. Pont F, et al. Single-Cell Signature Explorer for comprehensive visualization of single cell signatures across scRNA-seq datasets. Nucleic Acids Res. 2019;47(21):e133. doi: 10.1093/nar/gkz601.
    1. Alfei F, et al. TOX reinforces the phenotype and longevity of exhausted T cells in chronic viral infection. Nature. 2019;571(7764):265–269. doi: 10.1038/s41586-019-1326-9.
    1. Gourdin N, et al. Autocrine adenosine regulates tumor polyfunctional CD73+CD4+ effector T cells devoid of immune checkpoints. Cancer Res. 2018;78(13):3604–3618.
    1. McLane LM, et al. CD8 T cell exhaustion during chronic viral infection and cancer. Annu Rev Immunol. 2019;37:457–495. doi: 10.1146/annurev-immunol-041015-055318.
    1. Bengsch B, et al. Epigenomic-guided mass cytometry profiling reveals disease-specific features of exhausted CD8 T cells. Immunity. 2018;48(5):1029–1045. doi: 10.1016/j.immuni.2018.04.026.
    1. Yoshitomi H, et al. Human Sox4 facilitates the development of CXCL13-producing helper T cells in inflammatory environments. Nat Commun. 2018;9(1):3762. doi: 10.1038/s41467-018-06187-0.
    1. Workel HH, et al. A transcriptionally distinct CXCL13+CD103+CD8+ T-cell population is associated with B-cell recruitment and neoantigen load in human cancer. Cancer Immunol Res. 2019;7(5):784–796. doi: 10.1158/2326-6066.CIR-18-0517.
    1. Djenidi F, et al. CD8+CD103+ tumor-infiltrating lymphocytes are tumor-specific tissue-resident memory T cells and a prognostic factor for survival in lung cancer patients. J Immunol. 2015;194(7):3475–3486. doi: 10.4049/jimmunol.1402711.
    1. Skon CN, et al. Transcriptional downregulation of S1pr1 is required for the establishment of resident memory CD8+ T cells. Nat Immunol. 2013;14(12):1285–1293. doi: 10.1038/ni.2745.
    1. Pallett LJ, et al. IL-2 high tissue-resident T cells in the human liver: sentinels for hepatotropic infection. J Exp Med. 2017;214(6):1567–1580. doi: 10.1084/jem.20162115.
    1. Kortekaas KE, et al. CD39 identifies the CD4+ tumor-specific T-cell population in human cancer. Cancer Immunol Res. 2020;8(10):1311–1321. doi: 10.1158/2326-6066.CIR-20-0270.
    1. Miller BC, et al. Subsets of exhausted CD8+ T cells differentially mediate tumor control and respond to checkpoint blockade. Nat Immunol. 2019;20(3):326–336. doi: 10.1038/s41590-019-0312-6.
    1. Siddiqui I, et al. Intratumoral Tcf1+PD-1+CD8+ T Cells with stem-like properties promote tumor control in response to vaccination and checkpoint blockade immunotherapy. Immunity. 2019;50(1):195–211. doi: 10.1016/j.immuni.2018.12.021.
    1. Kamphorst AO, et al. Proliferation of PD-1+ CD8 T cells in peripheral blood after PD-1-targeted therapy in lung cancer patients. Proc Natl Acad Sci U S A. 2017;114(19):4993–4998. doi: 10.1073/pnas.1705327114.
    1. Garris CS, et al. Successful anti-PD-1 cancer immunotherapy requires T cell-dendritic cell crosstalk involving the cytokines IFN-γ and IL-12. Immunity. 2018;49(6):1148–1161. doi: 10.1016/j.immuni.2018.09.024.
    1. Perrot I, et al. Blocking antibodies targeting the CD39/CD73 immunosuppressive pathway unleash immune responses in combination cancer therapies. Cell Rep. 2019;27(8):2411–2425. doi: 10.1016/j.celrep.2019.04.091.
    1. Stoeckius, et al. Simultaneous epitope and transcriptome measurement in single cells. Nat Methods. 2017;14(9):865–868. doi: 10.1038/nmeth.4380.
    1. Stuart T, et al. Comprehensive integration of single-cell data. Cell. 2019;177(7):1888–1902. doi: 10.1016/j.cell.2019.05.031.
    1. Pont F, et al. Single-cell virtual cytometer allows user-friendly and versatile analysis and visualization of multimodal single cell RNAseq datasets. NAR Genomics and Bioinformatics. 2020;2(2))
    1. Saelens W, et al. A comparison of single-cell trajectory inference methods. Nat Biotechnol. 2019;37(5):547–554. doi: 10.1038/s41587-019-0071-9.
    1. Street K, et al. Slingshot: cell lineage and pseudotime inference for single-cell transcriptomics. BMC Genomics. 2018;19(1):477. doi: 10.1186/s12864-018-4772-0.

Source: PubMed

Подписаться