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:

Engineering CAR-T cells to activate small-molecule drugs in situ

Abstract

Chimeric antigen receptor (CAR)-T cells represent a major breakthrough in cancer therapy, wherein a patient’s own T cells are engineered to recognize a tumor antigen, resulting in activation of a local cytotoxic immune response. However, CAR-T cell therapies are currently limited to the treatment of B cell cancers and their effectiveness is hindered by resistance from antigen-negative tumor cells, immunosuppression in the tumor microenvironment, eventual exhaustion of T cell immunologic functions and frequent severe toxicities. To overcome these problems, we have developed a novel class of CAR-T cells engineered to express an enzyme that activates a systemically administered small-molecule prodrug in situ at a tumor site. We show that these synthetic enzyme-armed killer (SEAKER) cells exhibit enhanced anticancer activity with small-molecule prodrugs, both in vitro and in vivo in mouse tumor models. This modular platform enables combined targeting of cellular and small-molecule therapies to treat cancers and potentially a variety of other 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: Modular prodrug designs for use with SEAKER cells.
Fig. 2: In vitro validation of prodrug activation by CPG2- and β-Lac-expressing HEK293T cells.
Fig. 3: Construction and characterization of CPG2- and β-Lac-expressing SEAKER cells.
Fig. 4: In vitro validation of prodrug activation by SEAKER cells and antigen-negative cell killing.
Fig. 5: Enhanced in vivo efficacy of prodrug–SEAKER cell combinations in mouse i.p. xenografts.
Fig. 6: Enhanced in vivo efficacy of Ceph-AMS–β-Lac SEAKER cell combinations in mouse subcutaneous xenografts.

Similar content being viewed by others

Data availability

The authors declare that the data supporting the findings of this study are available within the article and its Supplementary Information files. Any raw data not provided therein are available from the corresponding authors upon reasonable request.

References

  1. Feldman, S. A., Assadipour, Y., Kriley, I., Goff, S. L. & Rosenberg, S. A. Adoptive cell therapy—tumor-infiltrating lymphocytes, T-cell receptors, and chimeric antigen receptors. Semin. Oncol. 42, 626–639 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  2. Leon, E., Ranganathan, R. & Savoldo, B. Adoptive T cell therapy: boosting the immune system to fight cancer. Semin. Immunol. 49, 101437 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Beyar-Katz, O. & Gill, S. Advances in chimeric antigen receptor T cells. Curr. Opin. Hematol. 27, 368–377 (2020).

    Article  CAS  PubMed  Google Scholar 

  5. 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 

  6. Sermer, D. & Brentjens, R. CAR T-cell therapy: full speed ahead. Hematol. Oncol. 37, 95–100 (2019).

    Article  CAS  PubMed  Google Scholar 

  7. Sotillo, E. et al. Convergence of acquired mutations and alternative splicing of CD19 enables resistance to CART-19 immunotherapy. Cancer Discov. 5, 1282–1295 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Maude, S. L. et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N. Engl. J. Med. 371, 1507–1517 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  9. Majzner, R. G. & Mackall, C. L. Tumor antigen escape from CAR T-cell therapy. Cancer Discov. 8, 1219–1226 (2018).

    Article  CAS  PubMed  Google Scholar 

  10. 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 

  11. Kasakovski, D., Xu, L. & Li, Y. T cell senescence and CAR-T cell exhaustion in hematological malignancies. J. Hematol. Oncol. 11, 91 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  12. Neelapu, S. S. et al. Chimeric antigen receptor T-cell therapy: assessment and management of toxicities. Nat. Rev. Clin. Oncol. 15, 47–62 (2018).

    Article  CAS  PubMed  Google Scholar 

  13. Dunbar, C. E. et al. Gene therapy comes of age. Science 359, eaan4672 (2018).

    Article  PubMed  Google Scholar 

  14. Lim, W. A. & June, C. H. The principles of engineering immune cells to treat cancer. Cell 168, 724–740 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Rafiq, S. et al. Targeted delivery of a PD-1-blocking scFv by CAR-T cells enhances anti-tumor efficacy in vivo. Nat. Biotechnol. 36, 847–856 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Yeku, O. O., Purdon, T. J., Koneru, M., Spriggs, D. & Brentjens, R. J. Armored CAR T cells enhance antitumor efficacy and overcome the tumor microenvironment. Sci. Rep. 7, 10541 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  17. Avanzi, M. P. et al. Engineered tumor-targeted T cells mediate enhanced anti-tumor efficacy both directly and through activation of the endogenous immune system. Cell Rep. 23, 2130–2141 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Boice, M. et al. Loss of the HVEM tumor suppressor in lymphoma and restoration by modified CAR-T cells. Cell 167, 405–418 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. 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 

  20. Brudno, J. N. & Kochenderfer, J. N. Recent advances in CAR T-cell toxicity: mechanisms, manifestations and management. Blood Rev. 34, 45–55 (2019).

    Article  CAS  PubMed  Google Scholar 

  21. Lambert, J. M. & Berkenblit, A. Antibody-drug conjugates for cancer treatment. Annu. Rev. Med. 69, 191–207 (2018).

    Article  CAS  PubMed  Google Scholar 

  22. Sharma, S. K. & Bagshawe, K. D. Translating antibody directed enzyme prodrug therapy (ADEPT) and prospects for combination. Expert Opin. Biol. Ther. 17, 1–13 (2017).

    Article  CAS  PubMed  Google Scholar 

  23. Nemunaitis, J. et al. Pilot trial of genetically modified, attenuated Salmonella expressing the E. coli cytosine deaminase gene in refractory cancer patients. Cancer Gene Ther. 10, 737–744 (2003).

    Article  CAS  PubMed  Google Scholar 

  24. Sherwood, R. F., Melton, R. G., Alwan, S. M. & Hughes, P. Purification and properties of carboxypeptidase G2 from Pseudomonas sp. strain RS-16. Use of a novel triazine dye affinity method. Eur. J. Biochem. 148, 447–453 (1985).

    Article  CAS  PubMed  Google Scholar 

  25. Fleming, P. C., Goldner, M. & Glass, D. G. Observations on the nature, distribution, and significance of cephalosporinase. Lancet 1, 1399–1401 (1963).

    Article  CAS  PubMed  Google Scholar 

  26. Jaffe, J. J., McCormack, J. J. & Meymerian, E. Trypanocidal properties of 5′-O-sulfamoyladenosine, a close structural analog of nucleocidin. Exp. Parasitol. 28, 535–543 (1970).

    Article  CAS  PubMed  Google Scholar 

  27. Rengaraju, S. et al. 5′-O-Sulfamoyladenosine (defluoronucleocidin) from a Streptomyces. Meiji Seika Kenkyu Nenpo 25, 49–55 (1986).

    Google Scholar 

  28. Francis, R. J. et al. A Phase I trial of antibody directed enzyme prodrug therapy (ADEPT) in patients with advanced colorectal carcinoma or other CEA producing tumours. Br. J. Cancer 87, 600–607 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Springer, C. J. et al. Optimization of alkylating agent prodrugs derived from phenol and aniline mustards: a new clinical candidate prodrug (ZD2767) for antibody-directed enzyme prodrug therapy (ADEPT). J. Med. Chem. 38, 5051–5065 (1995).

    Article  CAS  PubMed  Google Scholar 

  30. Yoo, B. et al. Ultrasmall dual-modality silica nanoparticle drug conjugates: design, synthesis, and characterization. Bioorg. Med. Chem. 23, 7119–7130 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Harding, F. A. et al. A beta-lactamase with reduced immunogenicity for the targeted delivery of chemotherapeutics using antibody-directed enzyme prodrug therapy. Mol. Cancer Ther. 4, 1791–1800 (2005).

    Article  CAS  PubMed  Google Scholar 

  32. Marais, R. et al. A cell surface tethered enzyme improves efficiency in gene-directed enzyme prodrug therapy. Nat. Biotechnol. 15, 1373–1377 (1997).

    Article  CAS  PubMed  Google Scholar 

  33. 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 

  34. Liu, Z. et al. Systematic comparison of 2A peptides for cloning multi-genes in a polycistronic vector. Sci. Rep. 7, 2193 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Levy, C. C. & Goldman, P. The enzymatic hydrolysis of methotrexate and folic acid. J. Biol. Chem. 242, 2933–2938 (1967).

    Article  CAS  PubMed  Google Scholar 

  36. Bulychev, A. & Mobashery, S. Class C beta-lactamases operate at the diffusion limit for turnover of their preferred cephalosporin substrates. Antimicrob. Agents Chemother. 43, 1743–1746 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. 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 

  38. Gurung, N., Ray, S., Bose, S. & Rai, V. A broader view: microbial enzymes and their relevance in industries, medicine, and beyond. BioMed. Res. Int. 2013, 329121 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Mayer, A. et al. Modifying an immunogenic epitope on a therapeutic protein: a step towards an improved system for antibody-directed enzyme prodrug therapy (ADEPT). Br. J. Cancer 90, 2402–2410 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Liu, S. et al. Corticosteroids do not influence the efficacy and kinetics of CAR-T cells for B-cell acute lymphoblastic leukemia. Blood Cancer J. 10, 15 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Bagshawe, K. D. & Sharma, S. K. Cyclosporine delays host immune response to antibody enzyme conjugate in ADEPT. in Proc. International Conference on New Trends in Clinical and Experimental Immunosuppression, Geneva (1996).

  42. Smith, G. K. et al. Toward antibody-directed enzyme prodrug therapy with the T268G mutant of human carboxypeptidase A1 and novel in vivo stable prodrugs of methotrexate. J. Biol. Chem. 272, 15804–15816 (1997).

    Article  CAS  PubMed  Google Scholar 

  43. Oosterhoff, D. et al. Secreted and tumour targeted human carboxylesterase for activation of irinotecan. Br. J. Cancer 87, 659–664 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Chen, K. C. et al. Membrane-localized activation of glucuronide prodrugs by beta-glucuronidase enzymes. Cancer Gene Ther. 14, 187–200 (2007).

    Article  CAS  PubMed  Google Scholar 

  45. Peraro, L. et al. Incorporation of bacterial immunoevasins to protect cell therapies from host antibody-mediated immune rejection Mol. Ther. https://doi.org/10.1016/j.ymthe.2021.06.022 (2021).

  46. Porter, D. L., Levine, B. L., Kalos, M., Bagg, A. & June, C. H. Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N. Engl. J. Med. 365, 725–733 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Gardner, T. J. et al. Targeted cellular micropharmacies: cells engineered for localized drug delivery. Cancers 12, 2175 (2020).

    Article  CAS  PubMed Central  Google Scholar 

  48. Quintarelli, C. et al. Co-expression of cytokine and suicide genes to enhance the activity and safety of tumor-specific cytotoxic T lymphocytes. Blood 110, 2793–2802 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Garber, K. Driving T-cell immunotherapy to solid tumors. Nat. Biotechnol. 36, 215–219 (2018).

    Article  CAS  PubMed  Google Scholar 

  50. Riviere, I., Brose, K. & Mulligan, R. C. Effects of retroviral vector design on expression of human adenosine deaminase in murine bone marrow transplant recipients engrafted with genetically modified cells. Proc. Natl Acad. Sci. USA 92, 6733–6737 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Sanjana, N. E., Shalem, O. & Zhang, F. Improved vectors and genome-wide libraries for CRISPR screening. Nat. Methods 11, 783–784 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank E. de Stanchina and C. Hagen (MSK Antitumor Assessment Core Facility) for assistance with mouse toxicology studies, G. Sukenick and R. Wang (MSK Analytical NMR Core Facility) for expert nuclear magnetic resonance and mass spectral support, J. Fraser Glickman and C. Adura Alcaino (Rockefeller High-Throughput and Spectroscopy Resource Center) for assistance with SPE–MS experiments, G. Chiosis and S. Sharma (MSK) for assistance with LC–MS/MS experiments and B. Yoo (MSK) for helpful discussions on the synthesis of APdMG-Glu. Financial support from the NIH (grant nos. P01 CA023766 to D.A.S. and D.S.T., R01 CA55349 and R35 CA241894 to D.A.S., R01 AI118224 to D.S.T. and CCSG P30 CA008748 to C.B. Thompson), the Tudor Fund (to D.A.S.), the Lymphoma Fund (to D.A.S.), the Commonwealth Foundation and MSK Center for Experimental Therapeutics (to D.A.S. and D.S.T.) is gratefully acknowledged.

Author information

Authors and Affiliations

Authors

Contributions

T.J.G., C.M.B. and D.A.S. conceived the in vitro and in vivo experiments with technical advice from D.W. and R.J.B. T.J.G. and C.M.B. executed the in vitro and in vivo experiments with assistance from K.G.K., M.M.D., A.Y.C. and G.M. J.P.L., N.K., B.C.C. and D.S.T. conceived the chemical, biochemical and ADME-PK experiments. J.P.L., B.C.C. and K.M.N. carried out the chemical syntheses. J.P.L., N.K. and B.C.C. carried out the biochemical assays. T.J.G., J.P.L., C.M.B., D.S.T. and D.A.S. wrote the paper with input from all coauthors.

Corresponding authors

Correspondence to Derek S. Tan or David A. Scheinberg.

Ethics declarations

Competing interests

D.A.S., D.S.T. and R.J.B. are consultants for, have equity in and have sponsored research agreements with CoImmune, which has licensed technology described in this paper from MSK. D.A.S. has equity in or is a consultant for: Actinium Pharmaceuticals, Arvinas, Eureka Therapeutics, Iovance Biotherapeutics, OncoPep, Pfizer, Repretoire and Sellas. D.S.T. has been a consultant and/or paid speaker for Eli Lilly, Elsevier, Emerson Collective, Merck, National Institutes of Health, Venenum Biodesign, the Research Center for Molecular Medicine of the Austrian Academy of Sciences and the Institute for Research in Biomedicine, Barcelona. R.J.B. is a cofounder and receives royalties from Juno Therapeutics/Celgene. MSK has filed for patent protection behalf of T.J.G., J.P.L., D.S.T. and D.A.S. for inventions described in this paper. The remaining authors declare no competing interests.

Additional information

Peer review information Nature Chemical Biology thanks Yuan Gao 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

Extended Data Fig. 1 Synthesis of AMS-Glu prodrug (2).

DMA = N,N-dimethylacetamide; DMF = N,N-dimethylformamide; TBAF = tetrabutylammonium fluoride; TBS = t-butyldimethylsilyl.

Extended Data Fig. 2 Synthesis of Ceph-AMS prodrug (3).

(a) Synthesis of protected AMS precursor 16. (b) Synthesis of Ceph-AMS (3). pyr = pyridine; TFA = trifluoroacetic acid; THF = tetrahydrofuran; TMS = trimethylsilyl.

Extended Data Fig. 3 Complete bioluminescent imaging data for in vivo efficacy in mouse intraperitoneal Raji tumor xenografts.

Mice treated with (a) CPG2-19BBz SEAKER cells and AMS-Glu (2) (50 mg/kg, ip, bid, days 2-7 post-CAR engraftment, 12 doses total, gray band) or (b) β-Lac-19BBz SEAKER cells and Ceph-AMS (3) (4 mg/kg, ip, bid, days 2–3 post-CAR engraftment, 3 doses total, gray band). Raw BLI is plotted on log scale; AUC is plotted on split linear scale. (mean ± s.d. of n = 5 mice per group; experiment was repeated with similar results.) Representative images are shown in Fig. 5d and e, respectively, of the manuscript.

Extended Data Fig. 4 Complete bioluminescent imaging data for in vivo efficacy against antigen-negative cells in intraperitoneal heterogeneous tumor xenografts.

(a) Bioluminescent imaging (BLI) and quantification of CD19+ Nalm6 cells expressing mCherry and Gaussia luciferase (Nalm6-mCherry/gLuc [CD19+]) in untreated mice or mice receiving β-Lac-19BBz SEAKER cells plus or minus 3 injections of Ceph-AMS (3: 4 mg/kg, ip, bid). Images taken at day 20 post tumor engraftment; one mouse in the group treated with SEAKER cells and prodrug showed tumor clearance but died after day 14 and is omitted. Complete radiance data is shown in the right two panels, with raw BLI plotted on log scale and AUC plotted on a linear scale. (Mean ± s.d. of n = 4 (untreated, SEAKER + prodrug) or n = 5 (SEAKER alone) mice per group; Student’s two-tailed t-test: ns = not significant; *p < 0.05; experiment was performed once). (b) BLI and quantification of CD19 Nalm6 cells expressing eGFP and firefly luciferase (Nalm6-eGFP/fLuc [CD19]) in untreated mice or mice receiving β-Lac-19BBz SEAKER cells plus or minus 3 injections of Ceph-AMS (3: 4 mg/kg, ip, bid). Images taken at day 17 post tumor engraftment; one mouse in the group treated with SEAKER cells and prodrug died after day 14 and is omitted. The left two panels also appear in Fig. 5g,h of the manuscript. Complete radiance data is shown in the right two panels, with raw BLI plotted on log scale and AUC plotted on a linear scale. (Mean ± s.d. of n = 4 (untreated, SEAKER + prodrug) or n = 5 (SEAKER alone) mice per group; Student’s two-tailed t-test: ns = not significant; *p < 0.05; experiment was performed once).

Extended Data Fig. 5 Persistence of β-Lac enzyme activity and Ceph-AMS prodrug activation in mouse intraperitoneal Raji tumor xenografts after SEAKER cell exhaustion in vivo.

(a) Experimental scheme for rescue of relapsed Raji xenograft by treatment with Ceph-AMS (3: 4 mg/kg, ip, bid, days 4–5, 3 doses total, then days 22–23 (gray bar), 3 doses total) after exhaustion of β-Lac-19BBz SEAKER cells. (b) Quantitation of tumor bioluminescence before and after second dosing period (median with s.d. of n = 4 mice per group; experiment was performed once). (c) Persistence of β-Lac enzyme activity in β-Lac-19BBz SEAKER cells extracted from two of the mice from the experiment in panel a at day 30 (day 28 post administration), in comparison to standard 19BBz CAR-T cell controls (flow cytometry analysis: β-Lac substrate = CCF2-AM).

Extended Data Fig. 6 Construction and characterization of β-Lac-expressing murine SEAKER cells.

(a) SEAKER construct encoding secreted β-Lac and a murine CAR: β-Lac-MUC28z (β-Lac/α-MUC16/CD28/CD3ζ). LTR = long terminal, Ψ = psi packaging element, FLAG = FLAG epitope tag (pink), P2A = 2A self cleaving peptide, α-MUC16 scFv = MUC-16-specific mouse-derived single chain variable fragment, myc = Myc epitope tag (brown), mCD28 = mouse CD28 costimulatory domain (green), mCD3ζ = mouse CD3 zeta chain (red). (b) Flow cytometry analysis of α-MUC16 CAR expression in retrovirally-transduced primary mouse T cells (fluorescently (phycoerythrin, PE) labeled anti-idiotype antibody; representative data from 5 independent experiments). (c) Trans-cytotoxicity of supernatant fluid (sn) from β-Lac-MUC28z SEAKER cells with or without Ceph-AMS (3: 107 nM) against mouse EL4 lymphoma cells, compared to prodrug alone and parent drug AMS (1: 500 nM) (24 h, CellTiter-Glo assay; mean ± s.d. of n = 3 technical replicates; Student’s two-tailed t-test: ns = not significant, ***p < 0.001; experiment was perfomed once).

Extended Data Fig. 7 Assessment of SEAKER cell immunogenicity in an immunocompetent mouse model.

Assessment of SEAKER cell immunogenicity in an immunocompetent mouse model. (a) Experimental scheme to assess immunogenicity of β-Lac-MUC28z SEAKER cells in a syngeneic intraperitoneal ID8 tumor model. Sera were collected on Days 21, 24, 28, and 31 and tested for anti-β-Lac antibodies in panel b. In a separate experiment, ascites were recovered on Day 28 by peritoneal lavage and tested for the presence of SEAKER cells in panels c,d and β-Lac enzyme activity in panel e. (b) Detection anti-β-Lac antibodies in sera over 10 days following SEAKER cell engraftment (Days 21–31) (median with lines representing each individual mouse of n = 12; experiment was performed once). (c,d) Flow cytometry analysis of SEAKER cell (myc+) persistence among T cells (CD3+) and (e) nitrocefin cleavage-based quantitation of β-Lac enzyme activity in ascites recovered 7 days after SEAKER cell engraftment (Day 28) (representative data shown from n = 2 mice per group; on average, 25% of T cells were SEAKER-positive; experiment was performed once). SSC = side scatter. (f) In a third experiment, mice were treated as in panel a, but without cyclophosphamide pretreatment to maximize the antibody response, then sera were recovered 5 days after SEAKER cell engraftment (Day 26) and analyzed for anti-β-Lac antibodies (n = 1 mouse in untreated group; n = 5 mice in treated group; mean ± s.d. of n = 3 technical replicates from each mouse; experiment was performed once). Sera from the 4 mice showing anti-β-Lac antibodies were used for the ex vivo enzyme activity experiment in panel g. (g) Nitrocefin cleavage-based quantitation of enzyme activity of recombinant β-Lac treated with sera from untreated or SEAKER-treated mice from panel f (n = 2 mice in untreated group; mean ± s.d. of n = 4 mice in treated group; experiment was performed once).

Supplementary information

Supplementary Information

Supplementary Tables 1–3, Figs 1–15 and Note (chemistry).

Reporting Summary

Supplementary Data

Original western blots from Supplementary Figs. 5, 6 and 8; representative ancestry gate for flow cytometry analysis.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gardner, T.J., Lee, J.P., Bourne, C.M. et al. Engineering CAR-T cells to activate small-molecule drugs in situ. Nat Chem Biol 18, 216–225 (2022). https://doi.org/10.1038/s41589-021-00932-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41589-021-00932-1

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