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:

IL-2 delivery by engineered mesenchymal stem cells re-invigorates CD8+ T cells to overcome immunotherapy resistance in cancer

Abstract

Immune checkpoint blockade (ICB)-based immunotherapy depends on functional tumour-infiltrating lymphocytes (TILs), but essential cytokines are less understood. Here we uncover an essential role of endogenous IL-2 for ICB responsiveness and the correlation between insufficient IL-2 signalling and T-cell exhaustion as tumours progress. To determine if exogenous IL-2 in the tumour microenvironment can overcome ICB resistance, we engineered mesenchymal stem cells (MSCs) to successfully deliver IL-2 mutein dimer (SIL2-EMSC) to TILs. While MSCs have been used to suppress inflammation, SIL2-EMSCs elicit anti-tumour immunity and overcome ICB resistance without toxicity. Mechanistically, SIL2-EMSCs activate and expand pre-existing CD8+ TILs, sufficient for tumour control and induction of systemic anti-tumour effects. Furthermore, engineered MSCs create synergy of innate and adaptive immunity. The therapeutic benefits of SIL2-EMSCs were also observed in humanized mouse models. Overall, engineered MSCs rejuvenate CD8+ TILs and thus potentiate ICB and chemotherapy.

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: Advanced tumour with insufficient IL-2 limits CD8+ TIL expansion, and IL-2 signalling is essential for ICB responsiveness.
Fig. 2: MSC engineering for inducible IL-2 can generate superior anti-tumour activity and overcome ICB resistance without toxicity.
Fig. 3: SIL2-EMSC treatment activates and expands the pre-existing CD8+ TILs for anti-tumour activity.
Fig. 4: SIL2-EMSCs remodel the TME, which re-invigorates exhausted CD8+ TILs.
Fig. 5: Local production of SIL2 effectively generates more TILs that exit through DLN to control distal tumours.
Fig. 6: Engineered MSCs in combination with β-lap chemotherapy create potential synergy for strong innate and adaptive immunity.
Fig. 7: SIL-EMSC treatment inhibits colon cancer growth in a humanized mouse model.

Similar content being viewed by others

Data availability

scRNA-seq data that support the findings of this study (Fig. 1a–d and Extended Data Fig. 1) can be accessed through the Gene Expression Omnibus under accession code GSE178881. The human SKCM data were derived from the TCGA Research Network: http://cancergenome.nih.gov/. Cumulative survival rate in patients with SKCM and gene correlation were analysed using Tumor Immune Estimation Resource (TIMER; https://cistrome.shinyapps.io/timer/). Source data are provided with this paper, available online for Figs. 17 and Extended Data Figs. 110. All other data that support the findings of this study are available from the corresponding author on reasonable request.

Code availability

The scRNA data were processed using Cell Ranger v.2.1.1 (https://www.10xgenomics.com/) and analysed with the R package Seurat v.3.1.2 (https://satijalab.org/seurat/). The R packages fgesa v.1.16.0 (http://bioconductor.org/packages/release/bioc/html/fgsea.html) and msigdbr v.7.2.1 (https://cran.r-project.org/web/packages/msigdbr/index.html) were used to perform the GSEA.

References

  1. Jiang, Y., Li, Y. & Zhu, B. T-cell exhaustion in the tumor microenvironment. Cell Death Dis. 6, e1792 (2015).

    Article  CAS  Google Scholar 

  2. Baitsch, L., Fuertes-Marraco, S. A., Legat, A., Meyer, C. & Speiser, D. E. The three main stumbling blocks for anticancer T cells. Trends Immunol. 33, 364–372 (2012).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  4. 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  Google Scholar 

  5. Jansen, C. S. et al. An intra-tumoral niche maintains and differentiates stem-like CD8 T cells. Nature 576, 465–470 (2019).

    Article  CAS  Google Scholar 

  6. 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  Google Scholar 

  7. 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 e110 (2019).

    Article  CAS  Google Scholar 

  8. Kurtulus, S. et al. Checkpoint blockade immunotherapy induces dynamic changes in PD-1CD8+ tumor-infiltrating T cells. Immunity 50, 181–194 e186 (2019).

    Article  CAS  Google Scholar 

  9. Jenkins, R. W., Barbie, D. A. & Flaherty, K. T. Mechanisms of resistance to immune checkpoint inhibitors. Br. J. Cancer 118, 9–16 (2018).

    Article  CAS  Google Scholar 

  10. Ayers, M. et al. IFN-γ-related mRNA profile predicts clinical response to PD-1 blockade. J. Clin. Invest. 127, 2930–2940 (2017).

    Article  Google Scholar 

  11. Rosenberg, S. A. IL-2: the first effective immunotherapy for human cancer. J. Immunol. 192, 5451–5458 (2014).

    Article  CAS  Google Scholar 

  12. Schwager, K., Hemmerle, T., Aebischer, D. & Neri, D. The immunocytokine L19-IL2 eradicates cancer when used in combination with CTLA-4 blockade or with L19-TNF. J. Invest. Dermatol. 133, 751–758 (2013).

    Article  CAS  Google Scholar 

  13. Buchbinder, E. I. et al. Therapy with high-dose interleukin-2 (HD IL-2) in metastatic melanoma and renal cell carcinoma following PD1 or PDL1 inhibition. J. Immunother. Cancer 7, 49 (2019).

    Article  Google Scholar 

  14. Pauken, K. E. & Wherry, E. J. Overcoming T cell exhaustion in infection and cancer. Trends Immunol. 36, 265–276 (2015).

    Article  CAS  Google Scholar 

  15. Zhang, L. et al. Lineage tracking reveals dynamic relationships of T cells in colorectal cancer. Nature 564, 268–272 (2018).

    Article  CAS  Google Scholar 

  16. Gubin, M. M. et al. High-dimensional analysis delineates myeloid and lymphoid compartment remodeling during successful immune-checkpoint cancer therapy. Cell 175, 1014–1030 e1019 (2018).

    Article  CAS  Google Scholar 

  17. Khan, O. et al. TOX transcriptionally and epigenetically programs CD8+ T cell exhaustion. Nature 571, 211–218 (2019).

    Article  CAS  Google Scholar 

  18. 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  Google Scholar 

  19. Ren, Z. et al. Selective delivery of low-affinity IL-2 to PD-1+ T cells rejuvenates antitumor immunity with reduced toxicity. J. Clin. Invest. 132, e153604 (2022).

    Article  CAS  Google Scholar 

  20. Sakuishi, K. et al. Targeting Tim-3 and PD-1 pathways to reverse T cell exhaustion and restore anti-tumor immunity. J. Exp. Med. 207, 2187–2194 (2010).

    Article  CAS  Google Scholar 

  21. Sun, Z. et al. A next-generation tumor-targeting IL-2 preferentially promotes tumor-infiltrating CD8+ T-cell response and effective tumor control. Nat. Commun. 10, 3874 (2019).

    Article  CAS  Google Scholar 

  22. Mott, H. R. et al. The solution structure of the F42A mutant of human interleukin 2. J. Mol. Biol. 247, 979–994 (1995).

    Article  CAS  Google Scholar 

  23. Levin, A. M. et al. Exploiting a natural conformational switch to engineer an interleukin-2 ‘superkine’. Nature 484, 529–533 (2012).

    Article  CAS  Google Scholar 

  24. Schlothauer, T. et al. Novel human IgG1 and IgG4 Fc-engineered antibodies with completely abolished immune effector functions. Protein Eng. Des. Sel. 29, 457–466 (2016).

    Article  CAS  Google Scholar 

  25. Liu, L. et al. Rejuvenation of tumour-specific T cells through bispecific antibodies targeting PD-L1 on dendritic cells. Nat. Biomed. Eng. 5, 1261–1273 (2021).

    Article  CAS  Google Scholar 

  26. Hsu, E. J. et al. A cytokine receptor-masked IL2 prodrug selectively activates tumor-infiltrating lymphocytes for potent antitumor therapy. Nat. Commun. 12, 2768 (2021).

    Article  CAS  Google Scholar 

  27. Kidd, S. et al. Direct evidence of mesenchymal stem cell tropism for tumor and wounding microenvironments using in vivo bioluminescent imaging. Stem Cells 27, 2614–2623 (2009).

    Article  CAS  Google Scholar 

  28. Chulpanova, D. S. et al. Application of mesenchymal stem cells for therapeutic agent delivery in anti-tumor treatment. Front. Pharm. 9, 259 (2018).

    Article  Google Scholar 

  29. Zou, W. et al. LIGHT delivery to tumors by mesenchymal stem cells mobilizes an effective antitumor immune response. Cancer Res. 72, 2980–2989 (2012).

    Article  CAS  Google Scholar 

  30. Lv, F. J., Tuan, R. S., Cheung, K. M. & Leung, V. Y. Concise review: the surface markers and identity of human mesenchymal stem cells. Stem Cells 32, 1408–1419 (2014).

    Article  CAS  Google Scholar 

  31. Peister, A. et al. Adult stem cells from bone marrow (MSCs) isolated from different strains of inbred mice vary in surface epitopes, rates of proliferation, and differentiation potential. Blood 103, 1662–1668 (2004).

    Article  CAS  Google Scholar 

  32. van Deursen, J. M. The role of senescent cells in ageing. Nature 509, 439–446 (2014).

    Article  Google Scholar 

  33. Turinetto, V., Vitale, E. & Giachino, C. Senescence in human mesenchymal stem cells: functional changes and implications in stem cell-based therapy. Int. J. Mol. Sci. 17, 1164 (2016).

    Article  Google Scholar 

  34. Bonab, M. M. et al. Aging of mesenchymal stem cell in vitro. BMC Cell Biol. 7, 14 (2006).

    Article  Google Scholar 

  35. Westerman, K. A. & Leboulch, P. Reversible immortalization of mammalian cells mediated by retroviral transfer and site-specific recombination. Proc. Natl Acad. Sci. USA 93, 8971–8976 (1996).

    Article  CAS  Google Scholar 

  36. Iida, Y. et al. Local injection of CCL19-expressing mesenchymal stem cells augments the therapeutic efficacy of anti-PD-L1 antibody by promoting infiltration of immune cells. J. Immunother. Cancer 8, e000582 (2020).

    Article  Google Scholar 

  37. Huang, X. et al. Leveraging an NQO1 bioactivatable drug for tumor-selective use of poly(ADP-ribose) polymerase inhibitors. Cancer Cell 30, 940–952 (2016).

    Article  CAS  Google Scholar 

  38. Li, X. et al. NQO1 targeting prodrug triggers innate sensing to overcome checkpoint blockade resistance. Nat. Commun. 10, 3251 (2019).

    Article  Google Scholar 

  39. Thompson, E. D., Enriquez, H. L., Fu, Y. X. & Engelhard, V. H. Tumor masses support naive T cell infiltration, activation, and differentiation into effectors. J. Exp. Med. 207, 1791–1804 (2010).

    Article  CAS  Google Scholar 

  40. Gupta, P. K. et al. CD39 expression identifies terminally exhausted CD8+ T cells. PLoS Pathog. 11, e1005177 (2015).

    Article  Google Scholar 

  41. Chen, Z. et al. TCF-1-centered transcriptional network drives an effector versus exhausted CD8 T cell-fate decision. Immunity 51, 840–855 e845 (2019).

    Article  CAS  Google Scholar 

  42. Guo, Y. et al. Metabolic reprogramming of terminally exhausted CD8+ T cells by IL-10 enhances anti-tumor immunity. Nat. Immunol. 22, 746–756 (2021).

    Article  CAS  Google Scholar 

  43. Krishna, S. et al. Stem-like CD8 T cells mediate response of adoptive cell immunotherapy against human cancer. Science 370, 1328–1334 (2020).

    Article  CAS  Google Scholar 

  44. Liu, J. et al. Improved efficacy of neoadjuvant compared to adjuvant immunotherapy to eradicate metastatic disease. Cancer Discov. 6, 1382–1399 (2016).

    Article  CAS  Google Scholar 

  45. Forde, P. M. et al. Neoadjuvant PD-1 blockade in resectable lung cancer. N. Engl. J. Med. 378, 1976–1986 (2018).

    Article  CAS  Google Scholar 

  46. Mender, I. et al. Telomere stress potentiates STING-dependent anti-tumor immunity. Cancer Cell 38, 400–411 e406 (2020).

    Article  CAS  Google Scholar 

  47. Arai, S. et al. Infusion of the allogeneic cell line NK-92 in patients with advanced renal cell cancer or melanoma: a phase I trial. Cytotherapy 10, 625–632 (2008).

    Article  CAS  Google Scholar 

  48. Nowakowska, P. et al. Clinical grade manufacturing of genetically modified, CAR-expressing NK-92 cells for the treatment of ErbB2-positive malignancies. Cancer Immunol. Immunother. 67, 25–38 (2018).

    Article  CAS  Google Scholar 

  49. Pink, J. J. et al. NAD(P)H:quinone oxidoreductase activity is the principal determinant of β-lapachone cytotoxicity. J. Biol. Chem. 275, 5416–5424 (2000).

    Article  CAS  Google Scholar 

  50. Bae, J. et al. Phc2 controls hematopoietic stem and progenitor cell mobilization from bone marrow by repressing Vcam1 expression. Nat. Commun. 10, 3496 (2019).

    Article  Google Scholar 

  51. Semenza, G. L. et al. Hypoxia response elements in the aldolase A, enolase 1, and lactate dehydrogenase A gene promoters contain essential binding sites for hypoxia-inducible factor 1. J. Biol. Chem. 271, 32529–32537 (1996).

    Article  CAS  Google Scholar 

  52. Zappasodi, R. et al. CTLA-4 blockade drives loss of Treg stability in glycolysis-low tumours. Nature 591, 652–658 (2021).

    Article  CAS  Google Scholar 

  53. Scrimieri, F. et al. Murine leukemia virus envelope gp70 is a shared biomarker for the high-sensitivity quantification of murine tumor burden. Oncoimmunology 2, e26889 (2013).

    Article  Google Scholar 

  54. Qiao, J. et al. Targeting tumors with IL-10 prevents dendritic cell-mediated CD8+ T cell apoptosis. Cancer Cell 35, 901–915 e904 (2019).

    Article  CAS  Google Scholar 

  55. Liu, L. et al. Concurrent delivery of immune checkpoint blockade modulates T cell dynamics to enhance neoantigen vaccine-generated antitumor immunity. Nat. Cancer 3, 437–452 (2022).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by Cancer Prevention and Research Institute of Texas (CPRIT) grant RR150072 given to Y.-X.F. and the NIH/NCI grant R01-CA240952 given to J.Q. The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript. We thank the Institutional Animal Care and Use Committee Animal Resources Center, and Animal Research Center. SSR#69 plasmid was kindly provided by T. C. He at Chicago University. HIF-1 reporter p2.1 plasmid was kindly provided by W. Luo at UT Southwestern Medical Center. Human cord blood was kindly provided by R. A. Word at Obstetrics and Gynecology Tissue Procurement Facility in UT Southwestern Medical Center, supported by NIH-P01-HD087150. We also thank C. Han, Z. Liu, C. Lu, Y. Liang, X. Cao, C. Dong and B. Moon for providing experiment materials and helpful discussions.

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization, J.B. and Y.-X.F.; methodology, J.B. and Y.-X.F.; investigation, J.B., L.L., C.M., E.H, J.S. and X.W.; writing—original draft, J.B.; writing—review and editing, C.M., E.H., J.Q. and Y.-X.F.; funding acquisition, Y.-X.F. and J.Q.; resources, A.Z., L.L., Z.S., Z.R. and J.Z.; supervision, Y.-X.F.

Corresponding authors

Correspondence to Jian Qiao or Yang-Xin Fu.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Cell Biology thanks Weiyi Peng, George Coukos and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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 Single cell analysis of CD8+ T cells in early and advanced tumor.

a, Schematic workflow of single cell RNA-Seq data generation. b, Feature plot showing the hallmark genes in each single-cell cluster identified by UMAP. c, UMAP plot showing the distribution of CD8+ T cells in the spleen of day 10 (S10), tumor of day 10 (T10), spleen of day 20 (S20), tumor of day 20 (T20). d, Violin plots showing the expression levels of IL-2 related genes across CD8+ T cell clusters. e, the Gene Set Enrichment Analysis (GSEA) was performed between cluster 1 (advanced tumor specific) and cluster 2 (early tumor specific).

Source data

Extended Data Fig. 2 Advanced tumors contain reduced number of CD8 + TILs and predominantly terminally exhausted cells.

a-c, C57BL/6J mice (T10, n = 6; T20, n = 8) were s.c. inoculated with 1 × 106 MC38 cells. 10 days (T10) or 20 days (T20) after tumor inoculation, TILs were analyzed for the frequency of PD1TIM3, PD1 + TIM3, and PD1+TIM3+ subset (a), stem-like exhausted CD8+ T cells (b), terminally exhausted CD8+ T cells (c). d, C57BL/6 J mice (Control, n = 6; anti-PD-L1, n = 5; anti-PD-L1 + anti-IL15, n = 6; anti-PD-L1 + anti-IL2, n = 5; anti-PD-L1 + anti-IL2Rβ, n = 5) were s.c. inoculated with 1 × 106 MC38 cells and i.p. injected with 150 μg of anti-PD-L1 and/or 200 μg anti-IL2Rβ, 200 μg anti-IL2 or 200 μg anti-IL15 on day 7,10 and 13 (black arrow). Tumor growth was measured twice a week. Data are shown as mean ± s.e.m. from two independent experiments. P value was determined by two-tailed unpaired t test (a-c) or two-way ANOVA (d). Source data are available online.

Source data

Extended Data Fig. 3 Survival, biodistribution and SIL2 production of engineered MSC.

a, Functional activity of WTIL2 and SIL2 were assessed using the HEK-BlueTM IL-2 reporter cell assay (n = 2). b,c, CT26 bearing BALB/c mice (n = 4 per group) were treated with SIL2 (20 μg, i.t.) on day 9. Five days after treatment, the concentration of SIL2 in serum (b), tumor and other tissues (c) were determined by hIgG ELISA. d, Body weight of CT26 bearing BALB/c mice (n = 5 per group) treated with SIL2-EMSC (1 × 106, p.t.) or SIL2 (20 μg, i.t.) on day 9 and 12. e, Surface marker expression of MSC by passage number in primary MSC and immortalized MSC (iMSC). f, Proliferation of CFSE-labeled primary MSC and iMSC assessed by flow cytometry at the indicated time points. g, MC38 bearing C57BL/6 mice (n = 3 per group) were p.t. injected with 5 × 105 luciferase expressing iMSCs on day 9. Luciferase signal was analyzed at the indicated time point. h-j, MC38 bearing mice (n = 4 per group) were p.t. injected with 5 × 105 EMSCs on day 9. The frequency of Ki67+GFP+ cells (h), and CD45GFP+ cells (i) in tumor were analyzed at the indicated time points. j, 200 μg anti-CD8 was administered 1 day before EMSC injection. Four days after EMSC injection, CD45GFP+ cells were analyzed in tumor. k, NSG-SGM3 mice were s.c. inoculated with 5 × 105 CT26 tumor cells and 5 × 105 luciferase expressing iMSCs were p.t. treated on day 10. Five days after MSC treatment, organs were extracted and luciferase signal was analyzed. l, Functional activity of SIL2-EMSC culture supernatant was assessed by using HEK-BlueTM IL-2 reporter cell assay (n = 2). Data are shown as mean ± s.e.m. from two independent experiments. P value was determined by two-tailed unpaired t test (b,h,j) or two-way ANOVA (d). Source data are available online.

Source data

Extended Data Fig. 4 Therapeutic effect of IL-2 expressing engineered MSC in vivo.

a, MC38 bearing C57BL/6 mice (Control, n = 7; EMSC (5x), n = 6; SIL2-EMSC (1x), n = 6; SIL2-EMSC (2x), n = 8; SIL2-EMSC, n = 8) were p.t. treated with 1 × 106 EMSC or SIL2-EMSC. Cells were injected every 3 days from day 9 as many times as indicated. b, MC38 bearing C57BL/6 mice (n = 5 per group) were p.t. treated with different number of EMSC or SIL2-EMSC on day 9 and 12 (black arrow). c, B16 bearing C57BL/6 mice (n = 5 per group) p.t. treated with 1 × 106 EMSC or SIL2-EMSC on day 7 and 10 (black arrow). CT26 bearing BALB/c mice (n = 5 per group) p.t. treated with 1 × 106 EMSC or SIL2-EMSC on day 9 and 12 (black arrow). 4T1 bearing BALB/c mice (n = 5 per group) p.t. treated with 1 × 106 EMSC or SIL2-EMSC on day 10 and 13 (black arrow). d,e, Advanced MC38 tumor bearing C57BL/6J mice (EMSC, n = 5; WTIL2-EMSC, n = 6; SIL2-EMSC, n = 6; EMSC + anti-PD-L1, n = 5; WTIL2-EMSC + anti-PD-L1, n = 7; SIL2-EMSC + anti-PD-L1, n = 6) were treated with 100 μg α-PD-L1 (i.p.) in combination with 1 × 106 EMSCs, WTIL2-EMSC or SIL2-EMSCs (p.t.) on day 14 and 17 (black arrow). Tumor growth (d) and survival curve (e) are shown. Tumor growth was measured twice a week. Data are shown as mean ± s.e.m. from two independent experiments. P value was determined by two-way ANOVA (a-d) or log rank test (e). Source data are available online.

Source data

Extended Data Fig. 5 Tumor-targeted production of SIL2 by engineered MSCs prevents systemic toxicity.

a-c, CT26 bearing BALB/c mice (Control, n = 4; SIL2, n = 4; SIL2-EMSC, n = 5) treated with SIL2-EMSC (1 × 106, p.t.) or SIL2 (20 ug, i.t.) on day 9 and 12. Two days after second treatment, serum, lung, and liver were isolated. a, Livers were extracted and fixed in 10% formalin for 7 days, then H&E staining was performed. Representative example of 10x magnification liver staining from each group is shown. b, Serum level of ALT and AST (b) and pulmonary wet weight (c) were examined as described in the Methods. d, CT26 bearing BALB/c mice (Control, n = 5; SIL2, n = 6; SIL2-EMSC, n = 6) treated with SIL2-EMSC (1 × 106, p.t.) or SIL2 (20 ug, i.t.) on day 9 and 12 (black arrow). Tumor growth was measured twice a week. e, CT26 bearing BALB/c mice (n = 5 per group) were treated with SIL2-EMSC (1 × 106, p.t.) or SIL2 (20 ug, i.t.) on day 9. IFNγ and TNFα level in the tumor tissue were determined by CBA at 1, 3, or 5 days after treatment. Data are shown as mean ± s.e.m. from two independent experiments. P value was determined by two-tailed unpaired t test (b,c,e) or two-way ANOVA (d). Source data are available online.

Source data

Extended Data Fig. 6 EMSC and SIL2-EMSC do not affect myeloid cell populations in the TME.

a,b, MC38 bearing mice (Control, n = 4; EMSC, n = 4; SIL2-EMSC, n = 6) were p.t. treated with 1 × 106 EMSCs or SIL2-EMSCs on days 9. a, Two days after treatment, the number of macrophages, dendritic cells, and MDSCs were analyzed. b, Cell surface expression level of CD80 and CD86 on dendritic cells were analyzed. c,d, MC38 bearing mice (n = 5) were p.t. treated with 1 × 106 EMSCs or SIL2-EMSCs on day 9 and 12. Five days after the second treatment, TILs were analyzed for the number of CD4+ T cells (c) and NK cells (d). e, MC38 bearing C57BL/6J mice (n = 5) were p.t. 1 × 106 EMSC, SIL2-EMSC or WTIL2-EMSC on day 9 and 12. Five days after the second treatment, TILs were analyzed for the CD8/Treg ratio. Data are shown as mean ± s.e.m. from two or three independent experiments. P value was determined by two-tailed unpaired t test (a-e). Source data are available online.

Source data

Extended Data Fig. 7 SIL2-EMSC expands and rejuvenates exhausted CD8+ TILs.

a-c, C57BL/6 J mice (n = 6) bearing MC38 were p.t. treated with 1 × 106 EMSCs or SIL2-EMSCs on day 9 and 12. Five days after the last treatment, tumor-infiltrating T cells were analyzed for the fold change of number of PD1TIM3, PD1+TIM3, and PD1+TIM3+ CD44+CD8+ T cell subsets (a) and the frequency of CXCR5+TIM3 subset (b) and CD39+Tcf1 subset (c) in PD1+TOX+CD44+CD8+ T cells in tumor were analyzed. d-f, CT26 bearing BALB/c mice (n = 5) were treated with 1 × 106 EMSC or SIL2-EMSC on day 9 and day 12. Five days after the second treatment, TILs were analyzed for the number of CD8+ T cells (d) and the frequency of PD1TIM3, PD1+TIM3, and PD1+TIM3+ CD8+ T cell subsets (e) and stem-like exhausted CD8+ T cells (f). g-i, C57BL/6J mice (n = 5–6) bearing MC38 were p.t. treated with 1 × 106 EMSCs or SIL2-EMSCs on day 9 and 12. Five days after the last treatment, TILs were analyzed for surface expression level of CD25 and CD122 (g, n = 6 per group), the frequency of Ki67+ cells (h, EMSC, n = 5; SIL2-EMSC, n = 6), and the frequency of active caspase 3+ cells (I, EMSC, n = 5; SIL2-EMSC, n = 6) in PD1TIM3, PD1+TIM3, and PD1+TIM3+ CD8+ T cell subsets. j, k, MC38 bearing C57BL/6J mice (n = 4) were i.t. treated with single high dose (20 μg) or prolonged low dose (5 μg, 4 injections) on day 9. Prolonged low dose group were injected with SIL2 every 12 h. CD8+ TILs were analyzed for the frequency of PD1TIM3, PD1+TIM3, PD1+TIM3+ CD8+ T cell subsets and (j) and stem-like CD8+ T cells (k). l, MC38 bearing C57BL/6J mice (n = 4) were treated with SIL2 (10 μg, i.t.) and/or EMSCs (1 × 106, p.t.) and SIL2-EMSC (1 × 106, p.t.) on day 9 and 12. Five days after treatment, CD8+ TILs were analyzed for the frequency of PD1TIM3, PD1+TIM3, PD1+TIM3+ CD8+ T cell subsets. Data are shown as mean ± s.e.m. from two or three independent experiments. P value was determined by two-tailed unpaired t test (ag,i–l). Source data are available online.

Source data

Extended Data Fig. 8 SIL2-EMSC expands and functionally reinvigorates exhausted CD8+ TILs.

a, CD8+ TILs from MC38 bearing mice (PD1TIM3, n = 3; PD1+TIM3, n = 6; PD1+TIM3+, n = 6) were co-cultured with irradiated MC38 in the presence of WTIL2 or SIL2. Two days later, the frequency of CD8+ TIL subsets were analyzed. b,c, CD8+ TILs from MC38 bearing mice (PD1+TIM3, n = 4; PD1+TIM3+, n = 5) were sorted out and labeled with CFSE. Cells were co-cultured with irradiated MC38 in the presence or absence of SIL2. Three days later, cell proliferation (b) and apoptosis (c) were determined by flow cytometry. d, IFN-γ reporter mice (n = 5 per group) bearing MC38 were p.t. treated with 1 × 106 EMSCs or SIL2-EMSCs on day 9. Two days after treatment, tumor-infiltrating T cells were analyzed to determine the frequency of IFN-γ+ cells in PD1TIM3, PD1+TIM3, and PD1+TIM3+ CD8+ T cell subsets. e, Naive OT-1 splenocytes were activated with OT-1 peptide in the presence of recombinant IL-2. OT-1 peptide primed OT-1 splenocytes were restimulated with dimerized α-CD3 for 2 d. Representative flow plots of the cell surface phenotype after restimulation (left) and PD1+TIM3 or PD1+ TIM3+ CD8+ subsets after sorting (right) are shown. f-h, B16-OVA tumor bearing Rag1 KO mice were adoptively transferred with OTI CD8+ T cells on day 8. Ten days after transfer, PD1+TIM3 or PD1+TIM3+ CD8+ TILs were sorted. B16-OVA tumor bearing C57BL/6 mice (EMSC, n = 5; EMSC + PD1+TIM3, n = 5; EMSC + PD1+TIM3+, n = 5; SIL2-EMSC, n = 6; SIL2-EMSC + PD1+TIM3, n = 5; SIL2-EMSC + PD1+TIM3+, n = 5) were adoptively transferred with 10,000 sorted CD8+ TIL subsets on day 5. EMSC or SIL2-EMSC were treated on day 5 and day 8. f, Experiment scheme. Tumor growth (g) and survival curve (h) are shown. Data are shown as mean ± s.e.m. from two to three independent experiments. P value was determined by two-tailed unpaired t test (a-d) or two-way ANOVA (g) or log rank test (h). Source data are available online.

Source data

Extended Data Fig. 9 SIL2-EMSCs are eliminated by administration of β-lap.

a, NQO1 expression in different cancer cell lines and EMSCs was determined by western blotting assay. Representative example from two independent experiments is shown. b, CT26 bearing Balb/c mice were p.t. injected with 5 × 105 luciferase expressing EMSCs on day 9. Two days after MSC injection, mice were i.t. treated with β-lap (5 mg/kg) every other day for four times. Luciferase signal was analyzed at indicated time points. c, CT26 bearing BALB/c mice (EMSC, n = 6; EMSC + β-lap, n = 5; SIL2-EMSC, n = 8; SIL2-EMSC + β-lap, n = 6) were i.t. treated with 1 × 106 EMSC or SIL2-EMSC on day 9 and 12 (black arrow). Two days after MSC injection, mice were i.t. treated with β-lap (15 mg/kg) every other day for four times. Data are shown as mean ± s.e.m. from two independent experiments. P value was determined by two-way ANOVA (c). Source data are available online.

Source data

Extended Data Fig. 10 SIL-EMSC treatment exhibits antitumor responses on humanized mouse model.

a, Cumulative survival in skin cutaneous melanoma (SKCM) patients according to Tcf7 expression level in TCGA database. b, TCGA database RNA-seq analysis of the correlation between the gene expression of Il2 and Tcf7 in patients with SKCM. c, HCT116 bearing humanized mice (Day 5, n = 5; Day 10, n = 3) were p.t. injected with 5 × 105 EMSCs on day 7. The frequency of CD45GFP+ cells in tumor was analyzed indicated time points. d, Humanized mice (n = 6 per group) were inoculated with 1 × 106 HCT116 cells and p.t. treated with 1 × 106 EMSCs or SIL2-EMSCs on day 7 and 10 (black arrow). Tumor growth was measured twice a week. e,f, HCT116 bearing humanized mice (EMSC, n = 3; SIL2-EMSC, n = 4) were p.t. treated with 1 × 106 EMSCs or SIL2-EMSCs on day 7 and 10. Five days after the last treatment, tumor-infiltrating T cells were analyzed for number of CD8+ T cells (e), and TCF1+ stem-like CD8+ T cells (f). g,h, SIL2-EMSCs irradiated with different doses (0–50 Gy). g, Two days after irradiation, cell viability was determined (n = 4). h, Cells were incubated in 1% (hypoxia) O2 for 24 hours. SIL2 levels from cell culture supernatants were determined by ELISA (n = 3). i, C57BL/6 mice (n = 5) were s.c. inoculated with 1 × 106 MC38 cells and p.t. treated with 1 × 106 EMSCs, SIL2-EMSCs, or SIL2-EMSC irradiated with 20 Gy on days 9 and 12 (indicated by black arrow). Tumor growth was measured twice a week. Data are shown as mean ± s.e.m. from two independent experiments. Statistical analysis of TCGA data were performed by log-rank test (a) or Spearman’s rho correlation test (b). P value was determined by two-way ANOVA (d,i) or two-tailed unpaired t test (e, f). Source data are available online.

Source data

Supplementary information

Supplementary Information

Supplementary Fig. 1.

Reporting Summary

Supplementary Table

Supplementary Table 1. List of differentially expressed genes from scRNA-seq. Supplementary Table 2. List of antibodies used in this study.

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. 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. 5

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. 8

Statistical source data.

Source Data Extended Data Fig. 9

Statistical source data.

Source Data Extended Data Fig. 9

Unprocessed western blots.

Source Data Extended Data Fig. 10

Statistical source data.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Bae, J., Liu, L., Moore, C. et al. IL-2 delivery by engineered mesenchymal stem cells re-invigorates CD8+ T cells to overcome immunotherapy resistance in cancer. Nat Cell Biol 24, 1754–1765 (2022). https://doi.org/10.1038/s41556-022-01024-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-022-01024-5

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing