Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Metabolic reprogramming of terminally exhausted CD8+ T cells by IL-10 enhances anti-tumor immunity

Abstract

T cell exhaustion presents one of the major hurdles to cancer immunotherapy. Among exhausted CD8+ tumor-infiltrating lymphocytes, the terminally exhausted subset contributes directly to tumor cell killing owing to its cytotoxic effector function. However, this subset does not respond to immune checkpoint blockades and is difficult to be reinvigorated with restored proliferative capacity. Here, we show that a half-life-extended interleukin-10–Fc fusion protein directly and potently enhanced expansion and effector function of terminally exhausted CD8+ tumor-infiltrating lymphocytes by promoting oxidative phosphorylation, a process that was independent of the progenitor exhausted T cells. Interleukin-10–Fc was a safe and highly efficient metabolic intervention that synergized with adoptive T cell transfer immunotherapy, leading to eradication of established solid tumors and durable cures in the majority of treated mice. These findings show that metabolic reprogramming by upregulating mitochondrial pyruvate carrier-dependent oxidative phosphorylation can revitalize terminally exhausted T cells and enhance the response to cancer immunotherapy.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: IL-10–Fc expands terminally exhausted CD8+ TILs.
Fig. 2: IL-10–Fc expands CD8+ T cells through IL-10R and enhances their effector function.
Fig. 3: IL-10–Fc expands antigen-specific terminally exhausted CD8+ T cells in a progenitor exhausted cell–independent manner.
Fig. 4: IL-10–Fc reprograms CD8+ T cell metabolism by promoting OXPHOS.
Fig. 5: IL-10–Fc potentiates ACT therapies to eradicate established tumors in multiple mouse models with durable protection.
Fig. 6: IL-10–Fc upregulates OXPHOS and expression of genes encoding effector function of terminally exhausted CD8+ TILs.
Fig. 7: IL-10–Fc promotes T cell OXPHOS and anti-tumor immunity in an MPC-dependent manner.

Similar content being viewed by others

Data availability

Gene sets in the MSigDB database (C2 and C7) were used for gene set enrichment analysis. All data generated and supporting the findings of this study are available within the paper. The RNA-seq data for tumor-infiltrating lymphocytes are available in the Gene Expression Omnibus database under accession code GSE168990. Source data are provided with this paper. Additional information and materials will be made available upon reasonable request.

References

  1. Chen, D. S. & Mellman, I. Elements of cancer immunity and the cancer–immune set point. Nature 541, 321–330 (2017).

    Article  CAS  PubMed  Google Scholar 

  2. Robert, C. et al. Pembrolizumab versus ipilimumab in advanced melanoma. N. Engl. J. Med. 372, 2521–2532 (2015).

    Article  CAS  PubMed  Google Scholar 

  3. Page, D. B., Postow, M. A., Callahan, M. K., Allison, J. P. & Wolchok, J. D. Immune modulation in cancer with antibodies. Annu. Rev. Med. 65, 185–202 (2014).

    Article  CAS  PubMed  Google Scholar 

  4. Sharma, P., Hu-Lieskovan, S., Wargo, J. A. & Ribas, A. Primary, adaptive, and acquired resistance to cancer immunotherapy. Cell 168, 707–723 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. McLane, L. M., Abdel-Hakeem, M. S. & Wherry, E. J. CD8 T cell exhaustion during chronic viral infection and cancer. Annu. Rev. Immunol. 37, 457–495 (2019).

    Article  CAS  PubMed  Google Scholar 

  6. Thommen, D. S. & Schumacher, T. N. T cell dysfunction in cancer. Cancer Cell 33, 547–562 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Chen, J. et al. NR4A transcription factors limit CAR T cell function in solid tumours. Nature 567, 530–534 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Miller, B. C. et al. Subsets of exhausted CD8+ T cells differentially mediate tumor control and respond to checkpoint blockade. Nat. Immunol. 20, 326–336 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. 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 50, 195–211.e10 (2019).

    Article  CAS  PubMed  Google Scholar 

  10. Kurtulus, S. et al. Checkpoint blockade immunotherapy induces dynamic changes in PD-1 CD8+ tumor-infiltrating T cells. Immunity 50, 181–194.e6 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. LaFleur, M. W. et al. PTPN2 regulates the generation of exhausted CD8+ T cell subpopulations and restrains tumor immunity. Nat. Immunol. 20, 1335–1347 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Paley, M. A. et al. Progenitor and terminal subsets of CD8+ T cells cooperate to contain chronic viral infection. Science 338, 1220–1225 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. He, R. et al. Follicular CXCR5-expressing CD8+ T cells curtail chronic viral infection. Nature 537, 412–416 (2016).

    Article  CAS  PubMed  Google Scholar 

  14. Im, S. J. et al. Defining CD8+ T cells that provide the proliferative burst after PD-1 therapy. Nature 537, 417–421 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Franco, F., Jaccard, A., Romero, P., Yu, Y. R. & Ho, P. C. Metabolic and epigenetic regulation of T-cell exhaustion. Nat. Metab. 2, 1001–1012 (2020).

    Article  CAS  PubMed  Google Scholar 

  16. Zhang, L. & Romero, P. Metabolic control of CD8+ T cell fate decisions and antitumor immunity. Trends Mol. Med. 24, 30–48 (2018).

    Article  CAS  PubMed  Google Scholar 

  17. Bengsch, B. et al. Bioenergetic insufficiencies due to metabolic alterations regulated by the inhibitory receptor PD-1 are an early driver of CD8+ T cell exhaustion. Immunity 45, 358–373 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Scharping, N. E. et al. Mitochondrial stress induced by continuous stimulation under hypoxia rapidly drives T cell exhaustion. Nat. Immunol. 22, 205–215 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Yu, Y. R. et al. Disturbed mitochondrial dynamics in CD8+ TILs reinforce T cell exhaustion. Nat. Immunol. 21, 1540–1551 (2020).

    Article  CAS  PubMed  Google Scholar 

  20. Vardhana, S. A. et al. Impaired mitochondrial oxidative phosphorylation limits the self-renewal of T cells exposed to persistent antigen. Nat. Immunol. 21, 1022–1033 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Fujii, S. I., Shimizu, K., Shimizu, T. & Lotze, M. T. Interleukin-10 promotes the maintenance of antitumor CD8+ T-cell effector function in situ. Blood 98, 2143–2151 (2001).

    Article  CAS  PubMed  Google Scholar 

  22. Mumm, J. B. et al. IL-10 elicits IFNγ-dependent tumor immune surveillance. Cancer Cell 20, 781–796 (2011).

    Article  CAS  PubMed  Google Scholar 

  23. Tanikawa, T. et al. Interleukin-10 ablation promotes tumor development, growth, and metastasis. Cancer Res. 72, 420–429 (2012).

    Article  CAS  PubMed  Google Scholar 

  24. Naing, A. et al. PEGylated IL-10 (pegilodecakin) induces systemic immune activation, CD8+ T cell invigoration and polyclonal T cell expansion in cancer patients. Cancer Cell 34, 775–791.e3 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Ip, W. K. E., Hoshi, N., Shouval, D. S., Snapper, S. & Medzhitov, R. Anti-inflammatory effect of IL-10 mediated by metabolic reprogramming of macrophages. Science 356, 513–519 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Tan, J. C., Indelicato, S. R., Narula, S. K., Zavodny, P. J. & Chou, C. C. Characterization of interleukin-10 receptors on human and mouse cells. J. Biol. Chem. 268, 21053–21059 (1993).

    Article  CAS  PubMed  Google Scholar 

  27. Wang, J., Saffold, S., Krauss, J., Chen, W. & Cao, X. Eliciting T cell immunity against poorly immunogenic tumors by immunization with dendritic cell-tumor fusion vaccines. J. Immunol. 161, 5516–5524 (1998).

    CAS  PubMed  Google Scholar 

  28. Lechner, M. G. et al. Immunogenicity of murine solid tumor models as a defining feature of in vivo behavior and response to immunotherapy. J. Immunother. 36, 477–489 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Alfei, F. et al. TOX reinforces the phenotype and longevity of exhausted T cells in chronic viral infection. Nature 571, 265–269 (2019).

    Article  CAS  PubMed  Google Scholar 

  30. Moynihan, K. D. et al. Eradication of large established tumors in mice by combination immunotherapy that engages innate and adaptive immune responses. Nat. Med. 22, 1402–1410 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Pai, C. C. S. et al. Clonal deletion of tumor-specific T cells by interferon-γ confers therapeutic resistance to combination immune checkpoint blockade. Immunity 50, 477–492.e8 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. June, C. H., O’Connor, R. S., Kawalekar, O. U., Ghassemi, S. & Milone, M. C. CAR T cell immunotherapy for human cancer. Science 359, 1361–1365 (2018).

    Article  CAS  PubMed  Google Scholar 

  33. Santos, J. M. et al. Adenovirus coding for interleukin-2 and tumor necrosis factor alpha replaces lymphodepleting chemotherapy in adoptive T cell therapy. Mol. Ther. 26, 2243–2254 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Klapper, J. A. et al. High-dose interleukin-2 for the treatment of metastatic renal cell carcinoma: a retrospective analysis of response and survival in patients treated in the Surgery Branch at the National Cancer Institute between 1986 and 2006. Cancer 113, 293–301 (2008).

    Article  CAS  PubMed  Google Scholar 

  35. Floros, T. & Tarhini, A. A. Anticancer cytokines: biology and clinical effects of interferon-α2, interleukin (IL)-2, IL-15, IL-21, and IL-12. Semin. Oncol. 42, 539–548 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Buck, M. D. D. et al. Mitochondrial dynamics controls T cell fate through metabolic programming. Cell 166, 63–76 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Herzig, S. et al. Identification and functional expression of the mitochondrial pyruvate carrier. Science 336, 93–96 (2012).

    Article  CAS  Google Scholar 

  38. Gray, L. R. et al. Hepatic mitochondrial pyruvate carrier 1 is required for efficient regulation of gluconeogenesis and whole-body glucose homeostasis. Cell Metab. 22, 669–681 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Grenell, A. et al. Loss of MPC1 reprograms retinal metabolism to impair visual function. Proc. Natl Acad. Sci. USA 116, 3530–3535 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Wegrzyn, J. et al. Function of mitochondrial Stat3 in cellular respiration. Science 323, 793–797 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Hamaidi, I. et al. Sirt2 inhibition enhances metabolic fitness and effector functions of tumor-reactive T cells. Cell Metab. 32, 420–436 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Lim, A. R., Rathmell, W. K. & Rathmell, J. C. The tumor microenvironment as a metabolic barrier to effector T cells and immunotherapy. Elife 9, e55185 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  43. Li, X. et al. Navigating metabolic pathways to enhance antitumour immunity and immunotherapy. Nat. Rev. Clin. Oncol. 16, 425–441 (2019).

    Article  CAS  PubMed  Google Scholar 

  44. Chapman, N. M., Boothby, M. R. & Chi, H. Metabolic coordination of T cell quiescence and activation. Nat. Rev. Immunol. 20, 55–70 (2020).

    Article  CAS  PubMed  Google Scholar 

  45. Qiao, J. et al. Targeting tumors with IL-10 prevents dendritic cell-mediated CD8+ T cell apoptosis. Cancer Cell 35, 901–915.e4 (2019).

    Article  CAS  PubMed  Google Scholar 

  46. Naing, A. et al. Pegilodecakin combined with pembrolizumab or nivolumab for patients with advanced solid tumours (IVY): a multicentre, multicohort, open-label, phase 1b trial. Lancet Oncol. 20, 1544–1555 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Platt, R. J. et al. CRISPR-Cas9 knockin mice for genome editing and cancer modeling. Cell 159, 440–455 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Vanderperre, B. et al. Embryonic lethality of mitochondrial pyruvate carrier 1 deficient mouse can be rescued by a ketogenic diet. PLoS Genet. 12, e1006056 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  49. Cheng, W. C. et al. Uncoupling protein 2 reprograms the tumor microenvironment to support the anti-tumor immune cycle. Nat. Immunol. 20, 206–217 (2019).

    Article  CAS  PubMed  Google Scholar 

  50. Tschumi, B. O. et al. CART cells are prone to Fas- and DR5-mediated cell death. J. Immunother. Cancer 6, 71 (2018).

  51. Guo, Y. et al. Purification and characterization of human IL-10/Fc fusion protein expressed in Pichia pastoris. Protein Expr. Purif. 83, 152–156 (2012).

    Article  CAS  PubMed  Google Scholar 

  52. Armour, K. L., Clark, M. R., Hadley, A. G. & Williamson, L. M. Recombinant human IgG molecules lacking Fcγ receptor I binding and monocyte triggering activities. Eur. J. Immunol. 29, 2613–2624 (1999).

    Article  CAS  PubMed  Google Scholar 

  53. Steele, A. W., Nickerson, P. W., Steurer, W., Steiger, J. & Strom, T. B. Administration of noncytolytic IL-10/Fc in murine models of lipopolysaccharide-induced septic shock and allogeneic islet transplantation. J. Immunol. 154, 5590–5600 (1995).

    PubMed  Google Scholar 

  54. Doench, J. G. et al. Optimized sgRNA design to maximize activity and minimize off-target effects of CRISPR-Cas9. Nat. Biotechnol. 34, 184–191 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Ma, L. et al. Enhanced CAR-T cell activity against solid tumors by vaccine boosting through the chimeric receptor. Science 365, 162–168 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Michelet, X. et al. Metabolic reprogramming of natural killer cells in obesity limits antitumor responses. Nat. Immunol. 19, 1330–1340 (2018).

    Article  CAS  PubMed  Google Scholar 

  57. Picelli, S. et al. Full-length RNA-seq from single cells using Smart-seq2. Nat. Protoc. 9, 171–181 (2014).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank D. Trono and B.E. Correia for providing plasmids of Delta 8.9 and pVSV-G; J.-C. Martinou (University of Geneva) for providing Mpc1fl/fl mice; and A. Donda (University of Lausanne) for the technical support on human CAR T cells. We acknowledge the EPFL Center of PhenoGenomics, Flow Cytometry Core Facility and Protein Expression Core Facility for technical assistance. This work was supported in part by the Swiss National Science Foundation (SNSF project grant no. 315230_173243), the ISREC Foundation with a donation from the Biltema Foundation, the Swiss Cancer League (grant no. KFS-4600-08-2018), the European Research Council under the ERC grant agreement MechanoIMM (grant no. 805337), the Kristian Gerhard Jebsen Foundation, Fondation Pierre Mercier pour la science, an Anna Fuller Fund grant and the EPFL (L.T.). P.-C.H. was supported in part by the Swiss Institute for Experimental Cancer Research (ISREC grant no. 26075483), SNSF project grants (grant nos. 31003A_163204 and 31003A_182470), the Cancer Research Institute Lloyd J. Old STAR award and the European Research Council Starting Grant (grant no. 802773-MitoGuide). P.R. was supported in part by grants from the SNSF (grant nos. 310030_182735 and 310030E-164187). W.H. was supported in part by the Swiss Cancer League (grant no. KFS-4407-02-2018) and the SNSF (grant no. 310030B_179570). W.X. was supported in part by the Strategic Priority Research Program of the Chinese Academy of Sciences (grant no. XDB29030000) and the Ministry of Science and Technology of China (grant no. 2016YFC1303503). M.G. was supported by the Chinese Scholarship Council (grant no. 201808320453).

Author information

Authors and Affiliations

Authors

Contributions

Y.G., Y.-Q.X. and L.T. conceived the study. Y.G., Y.-Q.X., L.T. and P.-C.H. designed the experiments. Y.G., Y.-Q.X., M.G., Y.Z., F.F., M.W., I.S., A.B., H.W., H.Y., B.F., X.X., C.M.S., B.T., A.C., Y.W., W.L., W.X., W.H. and P.R. performed the experiments. Y.G., Y.-Q.X., L.T. and P.-C.H. analyzed the data. Y.G., Y.-Q.X. and L.T. wrote the manuscript. All authors edited the manuscript.

Corresponding authors

Correspondence to Ping-Chih Ho or Li Tang.

Ethics declarations

Competing interests

Y.G., L.T. and Y.-Q.X. are inventors on the patent (International Publication Number WO 2021053134) related to the technology described in this manuscript. The remaining authors declare no competing interests.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 IL-10–Fc promotes tumor infiltration of T cells but shows less effects on other immune cells.

a, Schematic diagram of the production and purification of IL-10–Fc. b, Representative size-exclusion chromatographic traces. Peak 3 (P3) was collected and analyzed. c, Representative sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) analysis of purified IL-10–Fc. DTT, dithiothreitol. Results are one representative of three independent experiments. d, Splenocytes from PMEL mice were cultured with human gp10025–33 (hgp100) peptide (1 μM) and IL-2 (10 ng ml−1) for 3 d in the presence of IL-10–Fc or recombinant IL-10 at a series of concentrations. Shown are fold changes of CD8+ T cell counts (normalized by that in PBS control) at each concentration of cytokine. e-i, The experimental setting was the same as described in Fig. 1. Data are one representative of three or four independent experiments. e, Counts of CD45.2+ tumor infiltrating lymphocytes (TILs) and CD3+ TILs in B16F10 tumors. Shown are pooled data of two independent experiments (n = 10 independent animals). f, Representative immunofluorescence images of samples from each group. Green, CD3; blue, DAPI. Scale bar: 100 μm. Results are one representative of two independent experiments. g, Counts of various immune cell subsets in B16F10 tumors. For endogenous CD8+ T cells, PMEL CD8+ T cells, and total CD4+ T cells, shown are pooled data of two independent experiments (n = 10 independent animals). For other immune cells, data are representative of three independent experiments (n = 5 independent animals). NK: natural killer; DCs: dendritic cells; TAMs: tumor associated macrophages; MDSCs: myeloid-derived suppressive cells. h, Mean fluorescence intensity (MFI) of maturation markers on CD11b+CD11c+ tumor infiltrating DCs (n = 5 independent animals). i, Frequencies of M1 phenotype (CD206MHC-II+) among CD11b+F4/80+ TAMs (n = 5 independent animals). All data represent the mean ± s.e.m. and are analyzed by two-sided Student’s t-test or one-way ANOVA and Tukey’s test; NS, not significant (P > 0.05).

Source data

Extended Data Fig. 2 Tumor infiltrating PD-1+TIM-3+CD8+ T cells are PD-1+TCF-1TIM-3+ terminally exhausted CD8+ T cells, which respond to IL-10–Fc through IL-10 receptor.

a-d, The experimental setting was the same as described in Fig. 1. a-c, Data are representative of two independent experiments (n = 4 independent animals). a, Representative flow cytometry plots showing the frequencies of progenitor exhausted (TCF-1+TIM-3) and terminally exhausted (TCF-1TIM-3+) CD8+ TILs among total CD44+PD-1+CD8+ TILs. b,c, Frequencies and counts of three subpopulations of CD8+ TILs based on TCF-1/TIM-3 four-quadrant gating as shown in a. d, Frequencies of IFNγ+TNFα+ polyfunctional and IL-2+ T cells among PD-1+TIM-3CD8+ TILs or PD-1+TIM-3+CD8+ TILs from untreated tumors (n = 5 independent animals). Memory PMEL CD8+ T cells (n = 5 independent animals) in spleens from cured mice (schedule shown in Fig. 5c) were stimulated in the same way as positive controls. Data are one representative of two independent experiments. e-g, Activated CD8+ T cells from CRISPR-Cas9 KI P14 T cell receptor (TCR) transgenic mice were transfected with control gRNA or IL-10 receptor-α (IL-10Rα) allele specific gRNA to generate wild type control P14 T cells (Ctrl) or IL-10Rα-KO P14 T cells (IL-10Rα-KO), respectively. e, MFI of IL-10Rα expression. Data are one representative of two independent experiments (n = 6 independent samples). f, Ctrl P14 or IL-10Rα-KO P14 CD8+ T cells were restimulated by dimerized anti-CD3 (α-CD3) antibody in the presence or absence of IL-10–Fc and cultured for 2 d. Cell counts were analyzed by flow cytometry. Shown are relative T cell counts treated with IL-10–Fc versus that with PBS control. Data are one representative of two independent experiments (n = 3 independent samples). g, Experimental timeline of Fig. 2b. All data represent the mean ± s.e.m. and are analyzed by two-sided Student’s t-test or one-way ANOVA and Tukey’s test; NS, not significant (P > 0.05).

Source data

Extended Data Fig. 3 IL-10–Fc expands terminally exhausted CD8+ T cells in a progenitor exhausted cell-independent manner.

a,b, Thy1.2+ C57BL/6 mice were sublethally lymphodepleted and received adoptive transfer of Thy1.1+ naïve PMEL CD8+ T cells. Mice were inoculated with B16F10 tumor cells and were treated with adoptive transfer of Thy1.2+ activated PMEL CD8+ T cells. Subset [1] (PD-1+TIM-3) and subset [2] (PD-1+TIM-3+) were sorted from pooled Thy1.1+CD8+ TILs (n = 10 independent animals). Sorted subsets (5 × 104 for each) were transferred separately into Thy1.2+ C57BL/6 recipient mice bearing B16F10 tumors followed by administration of IL-10–Fc or PBS control. On day 13, mice were killed (n = 5 independent animals for groups receiving subset [1]; n = 3 independent animals for groups receiving subset [2]). a, Experimental timeline. b, Counts of tumor infiltrating Thy1.1+ donor T cells. c-e, Thy1.2+ C57BL/6 mice bearing B16F10 tumors received adoptive transfer of activated PMEL Thy1.1+CD8+ T cells and were killed. TILs were pooled from 3 ~ 6 mice and two subsets of Thy1.2+ endogenous CD8+ T cells were sorted. Sorted subsets were cultured separately ex vivo in the presence of dimerized α-CD3 and IL-2 with or without IL-10–Fc for 3 d. Data are one representative of two independent experiments (n = 3 independent samples). c, Experimental timeline. d, Fold change of T cell counts after 3-d ex vivo culture (n = 3 independent samples). e, Frequencies of PD-1+TIM-3+ terminally exhausted CD8+ T cells after 3-d ex vivo culture (n = 3 independent samples). f, Stacked bar graphs show the putative origins of PD-1+TIM-3+ cells in IL-10–Fc-treated tumors based on experimental results in d and e. g, The experimental setting was the same as that described in Fig. 1. MFI of active caspase-3 among three subpopulations (n = 4 independent animals). Data are one representative of two independent experiments. h,i, The experimental setting was the same as that shown in Fig. 3e. (n = 4 independent animals). h, Frequencies of Tcf7DTR-GFP+ P14 CD8+ T cells among all CD8+ TILs before (day 15) and after (day 17) the DT treatment. i, Representative flow cytometry plots. All data represent the mean ± s.e.m. and are analyzed by two-sided Student’s t-test; NS, not significant (P > 0.05).

Source data

Extended Data Fig. 4 IL-10–Fc shows minimal effects on T cell glycolysis.

Primed PMEL CD8+ T cells in resting phase (day 7) were co-cultured with B16F10 tumor cells at a ratio of 1 to 1 in the presence or absence of IL-10–Fc. After a 2-d co-culture, CD8+ T cells were isolated for Seahorse assay and flow cytometry analyses, respectively. Data are representative of three independent experiments (n = 3 independent samples). a, Real-time analysis of extracellular acidification rate (ECAR) of PMEL CD8+ T cells from the co-culture assay. b, Basal and maximal ECAR levels of PMEL CD8+ T cells. All data represent the mean ± s.e.m. and are analyzed by one-way ANOVA and Tukey’s test; NS, not significant (P > 0.05).

Source data

Extended Data Fig. 5 Ex vivo generated PD-1+TIM-3+CD8+ T cells by restimulation exhibit exhaustion phenotypes similarly as terminally exhausted CD8+ TILs.

a, Dimerized α-CD3 antibody was produced by mixing rat α-CD3 antibody with goat α-rat IgG Fc antibody at the mole ratio of 2:1. Primed PMEL CD8+ T cells in resting phase (day 7) were restimulated by dimerized α-CD3 at indicated concentrations and cultured in complete RPMI 1640 medium supplemented with IL-2 (10 ng ml−1) for 2 d. Shown are representative flow cytometry plots and frequencies of PD-1+TIM-3+CD8+ T cells among total PMEL CD8+ T cells. Data are representative of three independent experiments (n = 3 independent samples). b, Activated PMEL CD8+ T cells in resting phase were restimulated by dimerized α-CD3 antibody and IL-2 (10 ng ml−1) for 2 d. Freshly isolated naïve PMEL CD8+ T cells from splenocytes and restimulated PMEL CD8+ T cells were compared for the expression of PD-1, TIM-3 surface markers and TCF-1 transcription factor.

Extended Data Fig. 6 IL-10–Fc enhances OXPHOS and proliferation of human CD8+ T cells as well as their killing efficiency of target cells.

a-c, Human HER2 CAR-T cells were co-cultured with SKOV3-HER2 (a) or ME275-HER2 (b,c) tumor cells in vitro at a ratio of 1 to 1 in the presence or absence of IL-10–Fc. After a 2-d co-culture, counts of tumor cells and CD8+ T cells were measured by flow cytometry. CD8+ T cells from the co-culture were further isolated for Seahorse assay (a). Data are one representative of two independent experiments (n = 3 independent samples). a, Average basal OCR of the CD8+ HER2 CAR-T cells. b, Relative counts of PD-1+LAG-3+CD8+ CAR-T cells treated with IL-10–Fc versus that with PBS. c, Percentage of target cell killing by CAR-T cells. d-f, Activated human CD8+ T cells in resting phase were restimulated by coated α-CD3 antibody (0.05 μg mL−1) for 2 d. PD-1+LAG-3+CD8+ T cells were sorted for a Seahorse assay. d, Average basal OCR of PD-1+LAG-3+CD8+ T cells. e, Relative counts of PD-1+LAG-3+CD8+ T cells treated with IL-10–Fc versus that with PBS. f, MFI of IL-10Rα expression on three different subpopulations. Data are one representative of two independent experiments (n = 3 independent samples). All data represent the mean ± s.e.m. and are analyzed by one-way ANOVA and Tukey’s test or two-sided Student’s t-test; NS, not significant (P > 0.05).

Source data

Extended Data Fig. 7 IL-10–Fc potentiates adoptive T cell transfer therapies to eradicate established solid tumors without overt toxicity.

a, Shown are individual tumor growth curves of the therapy experiments in Fig. 5a. Indicated are numbers of long-term surviving mice among the total number of mice in the group. b-c, The experimental setting was the same as that described in Fig. 1. b, Ratio of counts of CD8+ T cells to regulatory T cells (Tregs) in tumors in different treatment groups. c, Frequencies of granzyme B+IFNγ+TNFα+ polyfunctional CD8+ T cells among total CD8+ TILs. Data are one representative of three independent experiments (n = 5 independent animals). d-h, Thy1.2+ C57BL/6 mice were inoculated s.c. with B16F10 melanoma cells (5 × 105) on day 0, and received i.v. adoptive transfer of activated Thy1.1+ PMEL CD8+ T cells (5 × 106) on day 6, followed by p.t. administration of IL-10–Fc (20 µg) or PBS control every 4 days until day 18. d, Experimental timeline. e-h, Shown are average tumor growth curves (e), survival curves (f), and individual tumor growth curves (g, h) of each treatment group. Indicated are numbers of long-term surviving mice out of the total numbers in the group. Data are representative of two independent experiments (n = 10 independent animals). i-k, The experimental setting was the same as that described in Fig. 1. On day 14, mice were killed and serum samples were collected for analysis. Shown are relative body weight (i), serum alanine transaminase (ALT, j) and aspartate transaminase (AST, k) levels of the treated mice. Shown are pooled data of two independent experiments (n = 10 independent animals). All data represent the mean ± s.e.m. and are analyzed by one-way ANOVA and Tukey’s test; NS, not significant (P > 0.05).

Source data

Extended Data Fig. 8 IL-10–Fc synergizes with immune checkpoint blockade therapy to eradicate established tumors.

BALB/c mice were inoculated subcutaneously with CT26 colon adenocarcinoma cells (3 × 105) and received p.t. administration of IL-10–Fc (20 µg) or PBS control every other day until day 14, together with p.t. administration of α-PD-1 (RMP1-14, 100 µg) every 3 days until day 12. Data are one representative of three independent experiments (n = 5 or 10 independent animals). a, Experimental timeline. b, Shown are survival curves of each treatment group. Indicated are numbers of long-term surviving mice among the total number of mice in the group. c, Shown are individual tumor growth curves. d, Cured mice from treatment group of combination of α-PD-1 and IL-10–Fc were re-challenged subcutaneously with CT26 (3 × 105) cells at day 90 post primary inoculation. Naïve wild type mice were inoculated with the same number of tumor cells as controls. Shown are survival curves and numbers of long-term surviving mice against the re-challenges. All data represent the mean ± s.e.m. and are analyzed by Log-rank test for survival curves; NS, not significant (P > 0.05).

Extended Data Fig. 9 IL-10–Fc upregulates OXPHOS and effector function related pathways in terminally exhausted CD8+ TILs in vivo.

The experimental setting for RNA-sequencing (RNA-seq) was shown in Fig. 6a. a, Heatmap of top 100 differentially expressed genes (IL-10–Fc versus PBS) were generated using z-scores derived from log2 (fold change). RNA-seq datasets were generated from two independent animals from PBS treatment group and four independent animals from IL-10–Fc treatment group. b, Volcano plot of differentially expressed genes (IL-10–Fc versus PBS). Transcripts with a false discovery rate (FDR) value < 0.05 and log2 (fold change) > 1 are highlighted in red; transcripts with an FDR value < 0.05 and log2 (fold change) < -1 are highlighted in blue. c, Canonical pathway analysis was performed by using QIAGEN Ingenuity Pathway Analysis (IPA) based on differentially expressed genes (DEGs) from RNA-seq dataset. Enrichment Z-score is generated based on hypergeometric distribution, where the negative logarithm of the significance level (P value) is obtained by Fisher’s exact test at the right tail. Shown are top 15 pathways that were significantly upregulated by IL-10–Fc treatment (-log (P value) > 6). Numbers on the right of each bar indicates the significance -log (P value) of each pathway. d, Bean plot of log2 (fold change) of genes involved in oxidative phosphorylation pathway shown in c.

Extended Data Fig. 10 IL-10–Fc promotes mitochondrial fitness and function of terminally exhausted CD8+ TILs in vivo in an MPC dependent manner.

a-c, CD45.1+CD45.2+ mice were inoculated subcutaneously with B16F10-OVA tumor cells (5 × 105) and received i.v. adoptive co-transfer of activated CD45.1+ WT OT-I CD8+ T cells and CD45.2+ MPC1-KO OT-I CD8+ T cells (1:1, 5 × 106 for each) on day 6 followed by p.t. administration of IL-10–Fc (20 µg) or PBS control every other day until day 12. On day 13, mice were killed and tumors were processed and analyzed by flow cytometry. Data are one representative of two independent experiments (n = 7 independent animals). a, b, Relative MFI of MitoTracker Green FM (MitoGreen) (a) and MitoTracker Deep Red FM (MitoDeepRed) (b) of WT or MPC1-KO PD-1+TIM-3+CD8+ OT-I TILs treated with IL-10–Fc versus that with PBS control. c, Frequencies of MitoSOX+CD8+ T cells among total WT or MPC1-KO PD-1+TIM-3+CD8+ OT-I TILs. d, f, Activated PMEL CD8+ T cells were starved overnight and restimulated by dimerized α-CD3 antibody in the presence or absence of IL-10–Fc for overnight (d) or indicated time (f). Proteins from total cell lysates were separated by SDS-PAGE and MPC1 (d) or pSTAT3 (Tyr705) (f) were detected by Western blot. Results are one representative of three independent experiments. e, Transcript expression of Mpc1, Mpc1-ps, and Mpc2. Data are extracted from analyses of RNA-seq. All data represent the mean ± s.e.m. and are analyzed by two-sided Student’s t-test; NS, not significant (P > 0.05).

Source data

Supplementary information

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 1

Unprocessed SDS–PAGE.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 10

Statistical source data.

Source Data Extended Data Fig. 10

Unprocessed western blots.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Guo, Y., Xie, YQ., Gao, M. et al. Metabolic reprogramming of terminally exhausted CD8+ T cells by IL-10 enhances anti-tumor immunity. Nat Immunol 22, 746–756 (2021). https://doi.org/10.1038/s41590-021-00940-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-021-00940-2

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer