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:

Senolytic CAR T cells reverse senescence-associated pathologies

An Author Correction to this article was published on 21 February 2024

This article has been updated

Abstract

Cellular senescence is characterized by stable cell-cycle arrest and a secretory program that modulates the tissue microenvironment1,2. Physiologically, senescence serves as a tumour-suppressive mechanism that prevents the expansion of premalignant cells3,4 and has a beneficial role in wound-healing responses5,6. Pathologically, the aberrant accumulation of senescent cells generates an inflammatory milieu that leads to chronic tissue damage and contributes to diseases such as liver and lung fibrosis, atherosclerosis, diabetes and osteoarthritis1,7. Accordingly, eliminating senescent cells from damaged tissues in mice ameliorates the symptoms of these pathologies and even promotes longevity1,2,8,9,10. Here we test the therapeutic concept that chimeric antigen receptor (CAR) T cells that target senescent cells can be effective senolytic agents. We identify the urokinase-type plasminogen activator receptor (uPAR)11 as a cell-surface protein that is broadly induced during senescence and show that uPAR-specific CAR T cells efficiently ablate senescent cells in vitro and in vivo. CAR T cells that target uPAR extend the survival of mice with lung adenocarcinoma that are treated with a senescence-inducing combination of drugs, and restore tissue homeostasis in mice in which liver fibrosis is induced chemically or by diet. These results establish the therapeutic potential of senolytic CAR T cells for senescence-associated diseases.

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: uPAR is a cell-surface and secreted biomarker of senescence.
Fig. 2: uPAR CAR T cells are bona fide senolytic agents.
Fig. 3: Senolytic CAR T cells show therapeutic efficacy in CCl4-induced liver fibrosis.
Fig. 4: Senolytic CAR T cells are therapeutic in NASH-induced liver fibrosis.

Similar content being viewed by others

Data availability

The RNA-seq data have been deposited in the Gene Expression Omnibus under the accession number GSE145642. Source data are provided with this paper. All other data supporting the findings of this study will be made available upon reasonable request to the corresponding authors. Source data

Change history

References

  1. He, S. & Sharpless, N. E. Senescence in health and disease. Cell 169, 1000–1011 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Sharpless, N. E. & Sherr, C. J. Forging a signature of in vivo senescence. Nat. Rev. Cancer 15, 397–408 (2015).

    Article  CAS  PubMed  Google Scholar 

  3. Serrano, M., Lin, A. W., McCurrach, M. E., Beach, D. & Lowe, S. W. Oncogenic ras provokes premature cell senescence associated with accumulation of p53 and p16INK4a. Cell 88, 593–602 (1997).

    Article  CAS  PubMed  Google Scholar 

  4. Kang, T. W. et al. Senescence surveillance of pre-malignant hepatocytes limits liver cancer development. Nature 479, 547–551 (2011).

    Article  ADS  CAS  PubMed  Google Scholar 

  5. Demaria, M. et al. An essential role for senescent cells in optimal wound healing through secretion of PDGF-AA. Dev. Cell 31, 722–733 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Krizhanovsky, V. et al. Senescence of activated stellate cells limits liver fibrosis. Cell 134, 657–667 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Collado, M., Blasco, M. A. & Serrano, M. Cellular senescence in cancer and aging. Cell 130, 223–233 (2007).

    Article  CAS  PubMed  Google Scholar 

  8. Baker, D. J. et al. Clearance of p16Ink4a-positive senescent cells delays ageing-associated disorders. Nature 479, 232–236 (2011).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  9. Baar, M. P. et al. Targeted apoptosis of senescent cells restores tissue homeostasis in response to chemotoxicity and aging. Cell 169, 132–147 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Childs, B. G. et al. Senescent intimal foam cells are deleterious at all stages of atherosclerosis. Science 354, 472–477 (2016).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  11. Smith, H. W. & Marshall, C. J. Regulation of cell signalling by uPAR. Nat. Rev. Mol. Cell Biol. 11, 23–36 (2010).

    Article  CAS  PubMed  Google Scholar 

  12. Kirkland, J. L. & Tchkonia, T. Cellular senescence: a translational perspective. EBioMedicine 21, 21–28 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Xu, M. et al. Senolytics improve physical function and increase lifespan in old age. Nat. Med. 24, 1246–1256 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Sadelain, M., Rivière, I. & Riddell, S. Therapeutic T cell engineering. Nature 545, 423–431 (2017).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  15. Park, J. H. et al. Long-term follow-up of CD19 CAR therapy in acute lymphoblastic leukemia. N. Engl. J. Med. 378, 449–459 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Aghajanian, H. et al. Targeting cardiac fibrosis with engineered T cells. Nature (2019).

  17. Du, H. et al. Antitumor responses in the absence of toxicity in solid tumors by targeting B7-H3 via chimeric antigen receptor T cells. Cancer Cell 35, 221–237 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Pellegatta, S. et al. Constitutive and TNFα-inducible expression of chondroitin sulfate proteoglycan 4 in glioblastoma and neurospheres: implications for CAR-T cell therapy. Sci. Transl. Med. 10, eaao2731 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Ruscetti, M. et al. NK cell-mediated cytotoxicity contributes to tumor control by a cytostatic drug combination. Science 362, 1416–1422 (2018).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  20. Perna, F. et al. Integrating proteomics and transcriptomics for systematic combinatorial chimeric antigen receptor therapy of AML. Cancer Cell 32, 506–519 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Tasdemir, N. et al. BRD4 connects enhancer remodeling to senescence immune surveillance. Cancer Discov. 6, 612–629 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Simon, D. I. et al. Mac-1 (CD11b/CD18) and the urokinase receptor (CD87) form a functional unit on monocytic cells. Blood 88, 3185–3194 (1996).

    Article  CAS  PubMed  Google Scholar 

  23. Bugge, T. H. et al. The receptor for urokinase-type plasminogen activator is not essential for mouse development or fertility. J. Biol. Chem. 270, 16886–16894 (1995).

    Article  CAS  PubMed  Google Scholar 

  24. Coppé, J. P. et al. Senescence-associated secretory phenotypes reveal cell-nonautonomous functions of oncogenic RAS and the p53 tumor suppressor. PLoS Biol. 6, 2853–2868 (2008).

    Article  PubMed  Google Scholar 

  25. Hayek, S. S. et al. Soluble urokinase receptor and chronic kidney disease. N. Engl. J. Med. 373, 1916–1925 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Belcher, C., Fawthrop, F., Bunning, R. & Doherty, M. Plasminogen activators and their inhibitors in synovial fluids from normal, osteoarthritis, and rheumatoid arthritis knees. Ann. Rheum. Dis. 55, 230–236 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Guthoff, M. et al. Soluble urokinase receptor (suPAR) predicts microalbuminuria in patients at risk for type 2 diabetes mellitus. Sci. Rep. 7, 40627 (2017).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  28. Schuliga, M. et al. The fibrogenic actions of lung fibroblast-derived urokinase: a potential drug target in IPF. Sci. Rep. 7, 41770 (2017).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  29. Brentjens, R. J. et al. Eradication of systemic B-cell tumors by genetically targeted human T lymphocytes co-stimulated by CD80 and interleukin-15. Nat. Med. 9, 279–286 (2003).

    Article  CAS  PubMed  Google Scholar 

  30. Wang, C. et al. Inducing and exploiting vulnerabilities for the treatment of liver cancer. Nature 574, 268–272 (2019).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  31. Schnabl, B., Purbeck, C. A., Choi, Y. H., Hagedorn, C. H. & Brenner, D. Replicative senescence of activated human hepatic stellate cells is accompanied by a pronounced inflammatory but less fibrogenic phenotype. Hepatology 37, 653–664 (2003).

    Article  CAS  PubMed  Google Scholar 

  32. Puche, J. E. et al. A novel murine model to deplete hepatic stellate cells uncovers their role in amplifying liver damage in mice. Hepatology 57, 339–350 (2013).

    Article  PubMed  Google Scholar 

  33. Kuhn, N. F. et al. CD40 ligand-modified chimeric antigen receptor T cells enhance antitumor function by eliciting an endogenous antitumor response. Cancer Cell 35, 473–488 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Dobrenkov, K. et al. Monitoring the efficacy of adoptively transferred prostate cancer-targeted human T lymphocytes with PET and bioluminescence imaging. J. Nucl. Med. 49, 1162–1170 (2008).

    Article  PubMed  Google Scholar 

  35. Giavridis, T. et al. CAR T cell-induced cytokine release syndrome is mediated by macrophages and abated by IL-1 blockade. Nat. Med. 24, 731–738 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Norelli, M. et al. Monocyte-derived IL-1 and IL-6 are differentially required for cytokine-release syndrome and neurotoxicity due to CAR T cells. Nat. Med. 24, 739–748 (2018).

    Article  CAS  PubMed  Google Scholar 

  37. Feucht, J. et al. Calibration of CAR activation potential directs alternative T cell fates and therapeutic potency. Nat. Med. 25, 82–88 (2019).

    Article  CAS  PubMed  Google Scholar 

  38. Brunt, E. M. et al. Nonalcoholic fatty liver disease. Nat. Rev. Dis. Primers 1, 15080 (2015).

    Article  PubMed  Google Scholar 

  39. Wang, L. et al. Basing on uPAR-binding fragment to design chimeric antigen receptors triggers antitumor efficacy against uPAR expressing ovarian cancer cells. Biomed. Pharmacother. 117, 109173 (2019).

    Article  CAS  PubMed  Google Scholar 

  40. Paszkiewicz, P. J. et al. Targeted antibody-mediated depletion of murine CD19 CAR T cells permanently reverses B cell aplasia. J. Clin. Invest. 126, 4262–4272 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  41. Gargett, T. & Brown, M. P. The inducible caspase-9 suicide gene system as a “safety switch” to limit on-target, off-tumor toxicities of chimeric antigen receptor T cells. Front. Pharmacol. 5, 235 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  42. Anderson, K. G., Stromnes, I. M. & Greenberg, P. D. Obstacles posed by the tumor microenvironment to T cell activity: a case for synergistic therapies. Cancer Cell 31, 311–325 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Lujambio, A. et al. Non-cell-autonomous tumor suppression by p53. Cell 153, 449–460 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Bolger, A. M., Lohse, M. & Usadel, B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 30, 2114–2120 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    Article  CAS  PubMed  Google Scholar 

  46. Liao, Y., Smyth, G. K. & Shi, W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics 30, 923–930 (2014).

    Article  CAS  PubMed  Google Scholar 

  47. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  48. Chen, E. Y. et al. Enrichr: interactive and collaborative HTML5 gene list enrichment analysis tool. BMC Bioinformatics 14, 128 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  49. Livshits, G. et al. Arid1a restrains Kras-dependent changes in acinar cell identity. eLife 7, e35216 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  50. Zhu, C. et al. Hepatocyte Notch activation induces liver fibrosis in nonalcoholic steatohepatitis. Sci. Transl. Med. 10, eaat0344 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Wang, X. et al. Hepatocyte TAZ/WWTR1 promotes inflammation and fibrosis in nonalcoholic steatohepatitis. Cell Metab. 24, 848–862 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Fujii, M. et al. A murine model for non-alcoholic steatohepatitis showing evidence of association between diabetes and hepatocellular carcinoma. Med. Mol. Morphol. 46, 141–152 (2013).

    Article  CAS  PubMed  Google Scholar 

  53. Davila, M. L., Kloss, C. C., Gunset, G. & Sadelain, M. CD19 CAR-targeted T cells induce long-term remission and B cell aplasia in an immunocompetent mouse model of B cell acute lymphoblastic leukemia. PLoS ONE 8, e61338 (2013).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  54. Maher, J., Brentjens, R. J., Gunset, G., Rivière, I. & Sadelain, M. Human T-lymphocyte cytotoxicity and proliferation directed by a single chimeric TCRζ/CD28 receptor. Nat. Biotechnol. 20, 70–75 (2002).

    Article  CAS  PubMed  Google Scholar 

  55. Brentjens, R. J. et al. Genetically targeted T cells eradicate systemic acute lymphoblastic leukemia xenografts. Clin. Cancer Res. 13, 5426–5435 (2007).

    Article  CAS  PubMed  Google Scholar 

  56. Hagani, A. B., Rivière, I., Tan, C., Krause, A. & Sadelain, M. Activation conditions determine susceptibility of murine primary T-lymphocytes to retroviral infection. J. Gene Med. 1, 341–351 (1999).

    Article  CAS  PubMed  Google Scholar 

  57. Santos, E. B. et al. Sensitive in vivo imaging of T cells using a membrane-bound Gaussia princeps luciferase. Nat. Med. 15, 338–344 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Van der Schueren, B. et al. Low cytochrome oxidase 4I1 links mitochondrial dysfunction to obesity and type 2 diabetes in humans and mice. Int. J. Obes. 39, 1254–1263 (2015).

    Article  Google Scholar 

Download references

Acknowledgements

We thank A. Lujambio and R. Brody and the Biorepository and Pathology Core at Icahn School of Medicine at Mount Sinai, and G. Askan and O. Basturk at the Department of Pathology at MSKCC, for tissue samples; L. Zender and H. Chen for sharing plasmids; N. Salgado, H. Chen, T. Baslan, S. Tian, A. Wuest, W. Luan and G. Gunset for technical assistance; and C. J. Sherr, E. de Stanchina, N. Kuhn, A. Dobrin, M. L. Sjöstrand and other members of the Lowe and Sadelain laboratories for insightful discussions. This work was supported by a grant from the National Institute of Aging (AG065396) to S.W.L., the Pasteur-Weizmann/Servier award to M.S. and a Memorial Sloan Kettering Cancer Center support grant (P30 CA008748) to both S.W.L. and M.S. laboratories. S.L.F. was supported by a grant from the National Institute of Diabetes and Digestive and Kidney Diseases (R01DK56621), a grant from the Department of Defense (CA150272) and the P30 grant (CA165979); C.A. was supported by a postgraduate fellowship from La Caixa foundation and is the recipient of the Harold E. Varmus graduate student fellowship from the Gerstner Sloan Kettering graduate school; J.F. was supported by the Care-for-Rare Foundation and the German Research Foundation (DFG); J.L. was supported by a fellowship from the DFG and a Shulamit Katzman Endowed Postdoctoral Research Fellowship; J.F. and J.L. are part of the Experimental Medicine Program at the University of Tuebingen; D.A.-C. was supported by a postdoctoral fellowship from Fundación Ramón Areces; J.A.B. was supported by the Grayer postgraduate fellowship and the Geoffrey Been graduate student fellowship from the Gerstner Sloan Kettering graduate school; and A.K. was supported by a grant from the National Cancer Institute (U54 0D020355-01). S.W.L. is the Geoffrey Been Chair of Cancer Biology and a Howard Hughes Medical Institute Investigator. We thank the following MSKCC core facilities for support: SKI flow cytometry core facility, animal facility, antitumor assessment core, laboratory for comparative biology, bioinformatics core and integrated genomics operation core.

Author information

Authors and Affiliations

Authors

Contributions

C.A., J.F. and J.L. conceived the project, designed, performed and analysed experiments and wrote the paper with assistance from all authors. Y.-J.H. analysed RNA-seq data. C.Z., D.A.-C., J.M.-S., J.A.B., X.L., A.K., S.H. and T.G. performed and analysed experiments. A.P. performed histopathological toxicity analysis. S.L.F. provided human liver samples, analysed data and reviewed the manuscript. E.P. provided human carotid endarterectomy samples and reviewed the manuscript. V.P. helped with T cell imaging studies. M.S. and S.W.L. conceived the project, supervised experiments and wrote the paper. All authors read and approved of the paper.

Corresponding authors

Correspondence to Michel Sadelain or Scott W. Lowe.

Ethics declarations

Competing interests

A patent application (PCT/US2020/016290; filed on 02/01/2020) has been submitted based in part on results presented in this manuscript on the use of CAR T cells that target uPAR as senolytic agents. C.A., J.F., J.L., M.S. and S.W.L. are listed as the inventors. J.F. and M.S. hold other unrelated patents on CAR technologies.S.W.L. is an advisor for and has equity in the following biotechnology companies: ORIC Pharmaceuticals, Faeth Therapeutics, Blueprint Medicines, Geras Bio, Mirimus Inc., PMV Pharmaceuticals and Constellation Pharmaceuticals.

Additional information

Peer review information Nature thanks Jesus Gil, Stephen Gottschalk and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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

Extended data figures and tables

Extended Data Fig. 1 Genes encoding surface molecules that are commonly upregulated in senescence.

a, Heat map of genes upregulated in therapy-induced senescence (TIS), oncogene-induced senescence (OIS) or replication-induced senescence (RIS) in HSCs. b, Venn diagram showing the number of common genes upregulated in the three datasets in a. c, Fold change (log2(expression in senescent cells/expression in non-senescent cells)) of the eight commonly upregulated genes in the three different datasets in a. d, Combined enrichment score of significantly enriched gene sets among the eight commonly upregulated genes in senescence. ECM, extracellular matrix; GPI, glycosylphosphatidylinositol. e, Heat map showing the expression profile of uPAR (PLAUR) in human vital tissues (as determined by the Human Proteome Map) compared to the expression profiles of other targets of CAR T cells in clinical trials. NK cells, natural killer cells. f, Immunohistochemical staining of mouse uPAR (m.uPAR) in vital tissues of C57BL/6J mice. Representative results of n = 2 independent experiments. g, Reads per kilobase (RPKM) of PLAUR mRNA in proliferating, quiescent (induced by serum starvation) or senescent (triggered by overexpression of HRASG12V) human IMR-90 fibroblasts. Results of one independent experiment with n = 3 replicates for proliferating, quiescent and senescent conditions. Data are mean ± s.e.m.; two-tailed unpaired Student’s t-test

Source Data

Extended Data Fig. 2 uPAR is a cell-surface and secreted biomarker of senescence.

a, b, qPCR of SASP-associated gene expression in senescent versus proliferating mouse KP tumour cells (a) or human primary melanocytes (b) and representative SA-β-gal staining; a.u. arbitrary units. c, d, Co-immunofluorescence staining and quantifications of uPAR (red) and Ki-67 (green) (c) or uPAR (red) and IL-6 (green) (d). e, Immunohistochemical staining of uPAR or phosphorylated ERK (P-ERK) in serial sections of mouse livers six days after transfection by HTVI with a plasmid encoding NrasG12V. Representative results of two independent experiments (n = 3 mice per group). fi, Mice expressing endogenous KrasG12D in pancreatic epithelial cells were treated with caerulein (Cr) and euthanized 21 weeks afterwards when they had developed pancreatic intraepithelial neoplasias. Age-matched C;RIK mice (expressing wild-type Kras) injected with PBS were used as controls. f, Co-immunofluorescence staining of KATE (red) and uPAR (green). Representative results of two independent experiments (n = 3 mice per group). g, Levels of suPAR in the mice in f. Representative results of two independent experiments (n = 2 mice per group). h, Co-immunofluorescence staining and quantification of uPAR (red) and Ki-67 (green). Representative results of two independent experiments (n = 3 mice per group). i, Representative SA-β-gal staining. Representative results of one independent experiment (n = 3 mice per group). jm, Mice were treated with either vehicle or CCl4 twice weekly for six weeks to induce liver fibrosis. j, Fold change in serum levels of suPAR. Representative results of two independent experiments (vehicle, n = 4; CCl4, n = 9 mice per group). Two-tailed unpaired Student’s t-test. k, Co-immunofluorescence staining and quantification of uPAR (red) and Ki-67 (green). Representative results of two independent experiments (n = 2 mice per group). l, Co-immunofluorescence staining and quantification of uPAR (red) and IL-6 (green). Representative results of two independent experiments (n = 3 mice per group). m, Representative SA-β-gal staining. Representative results of two independent experiments (n = 3 mice per group). Data are mean ± s.e.m. (c, d, h, j, l).

Source Data

Extended Data Fig. 3 uPAR is a marker of senescence in senescence-associated human pathologies.

a, Left, immunohistochemical expression of human uPAR (h.uPAR) and SA-β-gal in human samples of hepatitis-induced liver fibrosis (n = 7 patients). Right, co-immunofluorescence staining and quantification of uPAR (red) and p16 (green) or uPAR (red) and IL-6 (green) in human samples of hepatitis-induced liver fibrosis (n = 3). b, Left, immunohistochemical expression of uPAR and SA-β-gal in human samples from patients with eradicated hepatitis C virus (HCV) and residual liver fibrosis (n = 7 patients). Right, co-immunofluorescence staining and quantification of uPAR (red) and p16 (green) or uPAR (red) and IL-6 (green) in human samples of HCV-induced liver fibrosis (n = 3). Data are mean ± s.e.m. (a, b). c, Immunohistochemical staining of uPAR in human carotid endarterectomy samples (n = 5 patients). d, Immunohistochemical staining of uPAR in human pancreas bearing pancreatic intraepithelial neoplasia (PanIN) compared to normal pancreas controls (n = 3 patients).

Source Data

Extended Data Fig. 4 m.uPAR-h.28z CAR T cells selectively target uPAR-positive cells.

a, Construct maps encoding human m.uPAR-h.28z and h.19-h.28z CARs or mouse m.uPAR-m.28z and m.19-m.28z CARs. b, Flow cytometry analysis showing the expression levels of CAR and LNGFR in m.uPAR-h.28z and h.19-h.28z CAR T cells compared to untransduced T cells. Representative results of n = 4 independent experiments. c, Flow cytometry analysis of mouse uPAR and human CD19 expression on wild-type NALM6 cells and NALM6-m.uPAR cells. Representative results of n = 3 independent experiments. d, Cytotoxic activity of m.uPAR-h.28z, h.19-h.28z and untransduced T cells as determined by 4-h calcein assay with firefly luciferase (FFL)-expressing NALM6 wild-type or NALM6-m.uPAR cells as targets. Representative results of n = 3 independent experiments performed in triplicate. Data are mean ± s.e.m. e, Granzyme B (GrB) and IFNγ expression of CD4+ and CD8+ m.uPAR-h.28z CAR T cells 18 h after co-culture with wild-type NALM6, NALM6-m.uPAR or senescent KP cells as determined by intracellular cytokine staining. Results of n = 1 independent experiment (no target and NALM6 WT, n = 2; NALM6-m.uPAR and KP senescent, n = 3 replicates). Data are mean ± s.e.m. f, Experimental layout for Fig. 2c–i. Mice were injected with a plasmid encoding NrasG12V-GFP-luciferase and treated with 0.5 × 106 m.uPAR-h.28z CAR T cells or untransduced T cells 10 days after injection. Mice were euthanized 15 days after CAR administration and livers were used for further analysis. Images were created with BioRender.com. g, Flow cytometry analysis of mouse uPAR and human CD19 expression on wild-type Eμ-ALL01 cells and Eμ-ALL01-m.uPAR cells. Representative results of n = 3 independent experiments. h, Flow cytometry staining of Myc-tag and mouse uPAR on m.uPAR-m.28z CAR T cells, m.19-m.28z CAR T cells and untransduced T cells as compared to FMO control. Representative results of n = 2 independent experiments.

Source Data

Extended Data Fig. 5 Senolytic CAR T cells target senescent cells in a KrasG12D-driven model of lung cancer.

a, Experimental layout. C57BL/6N mice were intravenously injected with 10,000 KrasG12D;p53−/− cells. Treatment with combined MEK inhibitor (1 mg per kg body weight) and CDK4/6 inhibitors (100 mg per kg body weight) was started seven days later, followed by adoptive transfer of 2 × 106 CD45.1+ T cells (m.uPAR-m.28z CAR T cells, m.19-m.28z CAR T cells or untransduced T cells) one week later. A subset of mice received a second infusion of 1 × 106 m.uPAR-m.28z CAR T cells, m.19-m.28z CAR T cells or untransduced T cells seven days after the first injection of T cells. The images of the mouse, tumour cells and CAR T cells were created with BioRender.com. Cp, cyclophosphamide. b, Kaplan–Meier curve showing survival of mice (one-sided log-rank (Mantel–Cox) test). Results of two independent experiments (UT, n = 16; m.19-m.28z, n = 14; m.uPAR-m.28z, n = 18; UT reinjection, n = 6; m.19-m.28z reinjection, n = 7; m.uPAR-m.28z reinjection, n = 7 mice). c, d, Weight (c) and temperature (d) measured 24 h before and at different time points after CAR T cell infusion. P values (ns, not significant) refer to the comparison between untransduced and m.uPAR-m.28z injected mice at 48 h (weight, P = 0.9329; temperature, P = 0.1534). Results of one independent experiment (UT, n = 5; m.19-m.28z, n = 5; m.uPAR-m.28z, n = 8; UT reinjection, n = 5; m.19-m.28z reinjection, n = 7; m.uPAR-m.28z reinjection, n = 7 mice). e, Cell counts of CD45.1+ T cells and expression of the activation markers CD25 and CD69 (UT, n = 4; m.19-m.28z, n = 5; m.uPAR-m.28z, n = 5 mice) on CD45.1+ T cells in the lungs of mice seven days after administration of m.uPAR-m.28z CAR T cells, m.19-m.28z CAR T cells or untransduced T cells. f, Representative SA-β-gal staining and quantification in the lungs of mice seven days after treatment with m.uPAR-m.28z CAR T cells compared to mice that were treated with m.19-m.28z CAR T cells or untransduced T cells (n = 3 mice per group). Data are mean ± s.e.m.; two-tailed unpaired Student’s t-test. (cf).

Source Data

Extended Data Fig. 6 Senolytic CAR T cells show therapeutic activity in CCl4-induced liver fibrosis.

a, Layout for experiments performed using the CCl4-induced liver fibrosis model: C57BL/6N mice received intraperitoneal infusions of CCl4 twice weekly for six weeks and were intravenously infused with 0.5–1 × 106 (Fig. 3) or 2–3 × 106 (ci) mouse m.uPAR-m.28z CAR T cells, m.19-m.28z CAR T cells or untransduced T cells 16–24 h after administration of cyclophosphamide (200 mg kg−1). Mice were euthanized 20 days after CAR T cell infusion to assess liver fibrosis. Images were created with BioRender.com. b, Expression of GFP-tagged click beetle red (CBR) luciferase and Myc-tag in m.uPAR-m.28z and m.19-m.28z CAR T cells that were used for T cell imaging experiments (Fig. 3g, h) compared to control T cells. Representative results of n = 2 independent experiments. c, Sirius red and SA-β-gal staining and quantifications in livers from treated mice (n = 6 mice per group). d, Co-immunofluorescence of uPAR (red) and SMA (green) or Myc-tag (red) and SMA (green) in the livers of treated mice. e, Fold change difference in serum levels of suPAR 20 days after compared to 1 day before (day −1) injection of CAR T cells (UT, n = 18; m.19-m.28z, n = 6; m.uPAR-m.28z, n = 17 mice). f, g, Levels of serum ALT (f) and AST (g) 20 days after CAR treatment (UT, n = 10; m.19-m.28z, n = 8; m.uPAR-m.28z, n = 10 mice). h, Co-immunofluorescence staining of desmin (red) and Ki-67 (green) in the livers of mice 15, 20 and 77 days after treatment with CAR T cells. CCl4 treatment was stopped 20 days after T cell infusion (n = 3 mice per group). i, Mice were treated with CCl4 for 10 weeks. Sirius red staining in the livers of mice before (day −1) and 20 days after T cell administration (UT, n = 4; m.uPAR-m.28z, n = 2 mice). Representative results of n = 2 independent experiments (ci). Data are mean ± s.e.m.; two-tailed unpaired Student’s t-test (c, eg).

Source Data

Extended Data Fig. 7 Safety profile of m.uPAR-m.28z CAR T cells at therapeutic doses of T cells.

ae, C57BL/6N mice received intraperitoneal infusions of CCl4 twice weekly for six weeks and were intravenously injected with 0.5–1 × 106 m.uPAR-m.28z CAR T cells, 1 × 106 m.19-m.28z CAR T cells or untransduced T cells 16 h after administration of cyclophosphamide (200 mg kg−1). Mice were euthanized 20 days after T cell administration to assess potential toxicities and lung histopathology. a, Kaplan–Meier curve showing survival of mice after treatment with m.uPAR-m.28z CAR T cells (n = 16 mice), m.19-m.28z CAR T cells (n = 6 mice) or untransduced T cells (n = 6 mice). b, c, Weight (b) and temperature (c) of mice measured before and at different time points after CAR T cell infusion (UT and m.19-m.28z, n = 6; m.uPAR-m.28z, n = 7 mice). The P value in b refers to differences in weight at 48 h. d, e, Representative H&E staining of lungs (d) and complete blood counts (e) of treated mice 20 days after T cell infusion (UT and m.19-m.28z, n = 3 or 4; m.uPAR-m.28z, n = 4 mice). An increased accumulation of macrophages was observed in the immunodeficient setting. Representative results of n = 1 independent experiment (ae). Data are mean ± s.e.m. (b, c, e); two-tailed unpaired Student’s t-test (b, e).

Source Data

Extended Data Fig. 8 Safety profile of m.uPAR-m.28z CAR T cells at supratherapeutic doses of T cells.

C57BL/6N mice received intraperitoneal infusions of CCl4 twice weekly for six weeks followed by intravenous infusion of 2–3 × 106 m.uPAR-m.28z CAR T cells or untransduced T cells 16–24 h after administration of cyclophosphamide (200 mg kg−1). A subset of mice (as specified in the figure) received additional treatment with IL-6R-blocking antibodies (IL6Ri) and the IL-1R antagonist anakinra (IL1Ri), starting 24 h before T cell infusion and continuing daily until 6 days after T cell infusion. Mice were euthanized 12 weeks after CAR infusion to assess potential toxicities. a, Kaplan–Meier curve showing survival of mice after injection of CAR T cells (UT, n = 19; UT + IL6Ri/IL1Ri, n = 7; m.uPAR-m.28z, n = 30; m.uPAR-m.28z + IL6Ri/IL1Ri, n = 19 mice). b, c, Temperature (b) and weight (c) of treated mice (UT, n = 7; UT + IL6Ri/IL1Ri, n = 8; m.uPAR-m.28z, n = 11; m.uPAR-m.28z + IL6Ri/IL1Ri, n = 10 mice). d, Weight of mice 120 h after infusion with either m.uPAR-m.28z or m.uPAR-m.28z CAR T cells and additional treatment with IL6Ri and IL1Ri (m.uPAR-m.28z, n = 11; m.uPAR-m.28z + IL6Ri/IL1Ri, n = 10 mice). e, Serum levels of IL-6, GM-CSF, G-CSF and IFNγ in mice that were treated with either m.uPAR-m.28z or untransduced T cells 72 h or 20 days after T cell infusion (UT, n = 5; m.uPAR-m.28z, n = 4 mice at 72 h; n = 5 mice at 20 days). f, g, Number of adoptively transferred CD45.1+ T cells (f) and number of macrophages, uPAR+ and iNOS+ macrophages (g) in the lungs of mice that were treated with m.uPAR-m.28z CAR T cells, m.19-m.28z CAR T cells or untransduced T cells alone or in combination with treatment with IL6Ri and ILR1Ri three days after T cell infusion (n = 4 mice per group). h, i, Number of macrophages (h) and uPAR+ macrophages (i) in the lungs, liver, bone marrow (BM) and spleen of untreated mice or mice treated with either m.uPAR-m.28z CAR T cells or untransduced T cells 12 weeks after T cell infusion (n = 3 mice per group). Representative results of n = 3 independent experiments (ad) or n = 1 independent experiment (ei). All data are mean ± s.e.m.; two-tailed unpaired Student’s t-test (d, e).

Source Data

Extended Data Fig. 9 Therapeutic intervention with IL-6R and IL-1R inhibitors does not decrease the therapeutic efficacy of senolytic CAR T cells in CCl4-induced liver fibrosis.

a, Experimental layout. C57BL/6N mice received intraperitoneal infusions of CCl4 twice weekly for six weeks and were intravenously infused with 2–3 × 106 m.uPAR-m.28z CAR T cells or untransduced T cells 24 h after administration of cyclophosphamide (200 mg kg−1). IL-6R-blocking antibodies (IL6Ri) and anakinra (ILRi) were first administered 24 h before T cell infusion followed by daily (IL6Ri) or twice daily (IL1Ri) injections for the first six days until treatment was stopped. Mice were euthanized 20 days after T cell infusion to assess liver fibrosis. Images were created with BioRender.com. b, Fold change difference in serum levels of suPAR 20 days after compared to 1 day before (day −1) CAR T cell treatment (UT, n = 4; UT + IL6Ri/IL1Ri, n = 8; m.uPAR, n = 5; m.uPAR + IL6Ri/IL1Ri, n = 8 mice). c, d, Levels of serum ALT (c) and AST (d) in treated mice 20 days after T cell infusion (UT, n = 3; UT + IL6Ri/IL1Ri, n = 5; m.uPAR-m.28z, n = 5 (ALT) and n = 3 (AST); m.uPAR-m.28z + IL6Ri/IL1Ri, n = 5 mice). e, Representative levels of fibrosis evaluated by Sirius red staining and SA-β-gal staining in livers from treated mice and quantification of liver fibrosis and SA-β-gal+ cells in the respective livers 20 days after treatment (UT, n = 4; UT + IL6Ri/IL1Ri, n = 4; m.uPAR-m.28z, n = 4; m.uPAR-m.28z + IL6Ri/IL1Ri, n = 5 mice). f, Co-immunofluorescence staining of uPAR (red) and SMA (green) or Myc-tag (red) and SMA (green) in the livers of treated mice. Representative results of n = 1 independent experiment (bf). Data are mean ± s.e.m.; two-tailed unpaired Student’s t-test (be).

Source Data

Extended Data Fig. 10 Safety profile of senolytic CAR T cells at therapeutic doses in a mouse model of NASH-induced liver fibrosis.

a, Immunohistochemical expression of uPAR in samples from the ‘STAM’ model52,58 (n = 3 mice). b, Experimental layout for experiments performed using the model of diet-induced NASH (Fig. 4, this figure). C57BL/6N mice were treated with a chow or a NASH-inducing diet50 for three months, followed by intravenous infusion with 0.5 × 106 m.uPAR-m.28z CAR T cells or untransduced T cells 16 h after administration of cyclophosphamide (200 mg kg−1). Mice were euthanized 20 days after CAR infusion to assess liver fibrosis. Images were created with BioRender.com. c, Kaplan–Meier curve showing survival of mice after treatment with either m.uPAR-m.28z CAR T cells or untransduced T cells (m.uPAR-m.28z, n = 16; UT, n = 10 mice). d, e, Weight (d) and temperature (e) of mice 24 h before and at different time points after T cell infusion (m.uPAR-m.28z, n = 11; UT, n = 9 mice). Data are mean ± s.e.m. f, Representative H&E staining of the lungs of treated mice (m.uPAR-m.28z, n = 6; UT, n = 4 mice). Representative results of n = 2 independent experiments (cf).

Source Data

Extended Data Fig. 11 Gating strategies, summary and potential applications of senolytic CAR T cells.

a, b, Representative flow cytometry staining of m.uPAR-h.28z CAR T cells (a) or untransduced T cells (b) obtained from the livers of mice that had undergone HTVI (as depicted in Fig. 2). Representative results of one independent experiment (n = 4 mice per group). c, Summary of the key points of our findings. uPAR-28z CAR T cells (red) infiltrate fibrotic livers that contain senescent cells (blue) and efficiently eliminate them, leading to fibrosis resolution and improved liver function. The therapeutic action of senolytic uPAR-28z CAR T cells might be extended to other senescence-associated diseases such as atherosclerosis, diabetes or osteoarthritis. Images were created with BioRender.com.

Supplementary information

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

Amor, C., Feucht, J., Leibold, J. et al. Senolytic CAR T cells reverse senescence-associated pathologies. Nature 583, 127–132 (2020). https://doi.org/10.1038/s41586-020-2403-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-020-2403-9

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research