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.

Enhancing T cell therapy through TCR-signaling-responsive nanoparticle drug delivery

Abstract

Adoptive cell therapy (ACT) with antigen-specific T cells has shown remarkable clinical success; however, approaches to safely and effectively augment T cell function, especially in solid tumors, remain of great interest. Here we describe a strategy to 'backpack' large quantities of supporting protein drugs on T cells by using protein nanogels (NGs) that selectively release these cargos in response to T cell receptor activation. We designed cell surface–conjugated NGs that responded to an increase in T cell surface reduction potential after antigen recognition and limited drug release to sites of antigen encounter, such as the tumor microenvironment. By using NGs that carried an interleukin-15 super-agonist complex, we demonstrated that, relative to systemic administration of free cytokines, NG delivery selectively expanded T cells 16-fold in tumors and allowed at least eightfold higher doses of cytokine to be administered without toxicity. The improved therapeutic window enabled substantially increased tumor clearance by mouse T cell and human chimeric antigen receptor (CAR)-T cell therapy in vivo.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Figure 1: Synthesis and characterization of TCR-signaling-responsive protein nanogels.
Figure 2: Nanogel anchoring to CD45 promotes prolonged cell surface retention.
Figure 3: IL-15Sa-NG backpacks promote T cell expansion in vitro.
Figure 4: IL-15Sa-NGs promote specific expansion of adoptively transferred T cells in tumors.
Figure 5: IL-15Sa-NG backpacks increase the therapeutic window for adjuvant cytokine delivery during ACT.
Figure 6: TCR signaling–responsive NG backpacks improve T cell therapies.

References

  1. Rosenberg, S.A. & Restifo, N.P. Adoptive cell transfer as personalized immunotherapy for human cancer. Science 348, 62–68 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Gill, S. & June, C.H. Going viral: chimeric antigen receptor T cell therapy for hematological malignancies. Immunol. Rev. 263, 68–89 (2015).

    Article  CAS  PubMed  Google Scholar 

  3. Corrigan-Curay, J. et al. T cell immunotherapy: looking forward. Mol. Ther. 22, 1564–1574 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Johnson, L.A. et al. Rational development and characterization of humanized anti–EGFR variant III chimeric antigen receptor T cells for glioblastoma. Sci. Transl. Med. 7, 275ra22 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Klebanoff, C.A. et al. IL-15 enhances the in vivo antitumor activity of tumor-reactive CD8+ T cells. Proc. Natl. Acad. Sci. USA 101, 1969–1974 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Wallace, A. et al. Transforming growth factor–β receptor blockade augments the effectiveness of adoptive T cell therapy of established solid cancers. Clin. Cancer Res. 14, 3966–3974 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Conlon, K.C. et al. Redistribution, hyperproliferation, activation of natural killer cells and CD8 T cells, and cytokine production during first-in-human clinical trial of recombinant human interleukin-15 in patients with cancer. J. Clin. Oncol. 33, 74–82 (2015).

    Article  CAS  PubMed  Google Scholar 

  8. Leonard, J.P. et al. Effects of single-dose interleukin-12 exposure on interleukin-12-associated toxicity and interferon-γ production. Blood 90, 2541–2548 (1997).

    CAS  PubMed  Google Scholar 

  9. Zhang, L. et al. Tumor-infiltrating lymphocytes genetically engineered with an inducible gene encoding interleukin-12 for the immunotherapy of metastatic melanoma. Clin. Cancer Res. 21, 2278–2288 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Huang, B. et al. Active targeting of chemotherapy to disseminated tumors using nanoparticle-carrying T cells. Sci. Transl. Med. 7, 291ra94 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  11. Stephan, M.T., Moon, J.J., Um, S.H., Bershteyn, A. & Irvine, D.J. Therapeutic cell engineering with surface-conjugated synthetic nanoparticles. Nat. Med. 16, 1035–1041 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Stephan, M.T., Stephan, S.B., Bak, P., Chen, J. & Irvine, D.J. Synapse-directed delivery of immunomodulators using T cell–conjugated nanoparticles. Biomaterials 33, 5776–5787 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Ghezzi, P., Bonetto, V. & Fratelli, M. Thiol-disulfide balance: from the concept of oxidative stress to that of redox regulation. Antioxid. Redox Signal. 7, 964–972 (2005).

    Article  CAS  PubMed  Google Scholar 

  14. Lawrence, D.A., Song, R. & Weber, P. Surface thiols of human lymphocytes and their changes after in vitro and in vivo activation. J. Leukoc. Biol. 60, 611–618 (1996).

    Article  CAS  PubMed  Google Scholar 

  15. Berridge, M.V. & Tan, A.S. Trans-plasma membrane electron transport: a cellular assay for NADH- and NADPH-oxidase based on extracellular, superoxide-mediated reduction of the sulfonated tetrazolium salt WST-1. Protoplasma 205, 74–82 (1998).

    Article  CAS  Google Scholar 

  16. Berridge, M.V. & Tan, A.S. Cell-surface NAD(P)H-oxidase: relationship to trans-plasma membrane NADH-oxidoreductase and a potential source of circulating NADH-oxidase. Antioxid. Redox Signal. 2, 277–288 (2000).

    Article  CAS  PubMed  Google Scholar 

  17. Riber, C.F., Smith, A.A. & Zelikin, A.N. Self-immolative linkers literally bridge disulfide chemistry and the realm of thiol-free drugs. Adv. Healthc. Mater. 4, 1887–1890 (2015).

    Article  CAS  PubMed  Google Scholar 

  18. Jones, L.R. et al. Releasable luciferin–transporter conjugates: tools for the real-time analysis of cellular uptake and release. J. Am. Chem. Soc. 128, 6526–6527 (2006).

    Article  CAS  PubMed  Google Scholar 

  19. Xu, J. et al. Rendering protein-based particles transiently insoluble for therapeutic applications. J. Am. Chem. Soc. 134, 8774–8777 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Zhu, X. et al. Novel human interleukin-15 agonists. J. Immunol. 183, 3598–3607 (2009).

    Article  CAS  PubMed  Google Scholar 

  21. Rhode, P.R. et al. Comparison of the superagonist complex, ALT-803, to IL-15 as cancer immunotherapeutics in animal models. Cancer Immunol. Res. 4, 49–60 (2016).

    Article  CAS  PubMed  Google Scholar 

  22. Yu, A., Olosz, F., Choi, C.Y. & Malek, T.R. Efficient internalization of IL-2 depends on the distal portion of the cytoplasmic tail of the IL-2R common γ-chain and a lymphoid cell environment. J. Immunol. 165, 2556–2562 (2000).

    Article  CAS  PubMed  Google Scholar 

  23. Overwijk, W.W. et al. Tumor regression and autoimmunity after reversal of a functionally tolerant state of self-reactive CD8+ T cells. J. Exp. Med. 198, 569–580 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. 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  CAS  Google Scholar 

  25. Maus, M.V. et al. Adoptive immunotherapy for cancer or viruses. Annu. Rev. Immunol. 32, 189–225 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Guo, Y. et al. IL-15 superagonist-mediated immunotoxicity: role of NK cells and IFN-γ. J. Immunol. 195, 2353–2364 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Patel, S.K., Zhang, Y., Pollock, J.A. & Janjic, J.M. Cyclooxgenase-2 inhibiting perfluoropoly (ethylene glycol) ether theranostic nanoemulsions–in vitro study. PLoS One 8, e55802 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Pradhan, D. & Morrow, J. The spectrin–ankyrin skeleton controls CD45 surface display and interleukin-2 production. Immunity 17, 303–315 (2002).

    Article  CAS  PubMed  Google Scholar 

  29. Chang, V.T. et al. Initiation of T cell signaling by CD45 segregation at 'close contacts'. Nat. Immunol. 17, 574–582 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Johnson, K.G., Bromley, S.K., Dustin, M.L. & Thomas, M.L. A supramolecular basis for CD45 tyrosine phosphatase regulation in sustained T cell activation. Proc. Natl. Acad. Sci. USA 97, 10138–10143 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Wang, L. et al. Nanoclusters self-assembled from conformation-stabilized influenza M2e as broadly cross-protective influenza vaccines. Nanomedicine (Lond.) 10, 473–482 (2014).

    Article  CAS  Google Scholar 

  32. Scott, E.A. et al. Protein adsorption and cell adhesion on nanoscale bioactive coatings formed from poly(ethylene glycol) and albumin microgels. Biomaterials 29, 4481–4493 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Tan, H. et al. PEG-urokinase nanogels with enhanced stability and controllable bioactivity. Soft Matter 8, 2644–2650 (2012).

    Article  CAS  Google Scholar 

  34. Lin, X. et al. Oxidative stress in malignant melanoma enhances tumor necrosis factor–α secretion of tumor-associated macrophages that promote cancer cell invasion. Antioxid. Redox Signal. 19, 1337–1355 (2013).

    Article  CAS  PubMed  Google Scholar 

  35. Koshy, S.T., Ferrante, T.C., Lewin, S.A. & Mooney, D.J. Injectable, porous and cell-responsive gelatin cryogels. Biomaterials 35, 2477–2487 (2014).

    Article  CAS  PubMed  Google Scholar 

  36. Singh, N. et al. Bioresponsive mesoporous silica nanoparticles for triggered drug release. J. Am. Chem. Soc. 133, 19582–19585 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Au, K.M. et al. Folate-targeted pH-responsive calcium zoledronate nanoscale metal-organic frameworks: turning a bone anti-resorptive agent into an anticancer therapeutic. Biomaterials 82, 178–193 (2016).

    Article  CAS  PubMed  Google Scholar 

  38. Ling, D. et al. pH-sensitive nanoformulated triptolide as a targeted therapeutic strategy for hepatocellular carcinoma. ACS Nano 8, 8027–8039 (2014).

    Article  CAS  PubMed  Google Scholar 

  39. Yang, Y. et al. Polymer nanoparticles modified with photo- and pH-dual-responsive polypeptides for enhanced and targeted cancer therapy. Mol. Pharm. 13, 1508–1519 (2016).

    Article  CAS  PubMed  Google Scholar 

  40. Chmielewski, M., Kopecky, C., Hombach, A.A. & Abken, H. IL-12 release by engineered T cells expressing chimeric antigen receptors can effectively muster an antigen-independent macrophage response on tumor cells that have shut down tumor antigen expression. Cancer Res. 71, 5697–5706 (2011).

    Article  CAS  PubMed  Google Scholar 

  41. Chinnasamy, D. et al. Local delivery of interleukin-12 using T cells targeting VEGF receptor 2 eradicates multiple vascularized tumors in mice. Clin. Cancer Res. 18, 1672–1683 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Pegram, H.J. et al. Tumor-targeted T cells modified to secrete IL-12 eradicate systemic tumors without need for prior conditioning. Blood 119, 4133–4141 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Weinstein-Marom, H. et al. Membrane-attached cytokines expressed by mRNA electroporation act as potent T cell adjuvants. J. Immunother. 39, 60–70 (2016).

    Article  CAS  PubMed  Google Scholar 

  44. Desnoyers, L.R. et al. Tumor-specific activation of an EGFR-targeting probody enhances therapeutic index. Sci. Transl. Med. 5, 207ra144 (2013).

    Article  PubMed  CAS  Google Scholar 

  45. Wu, C.-Y., Roybal, K.T., Puchner, E.M., Onuffer, J. & Lim, W.A. Remote control of therapeutic T cells through a small-molecule-gated chimeric receptor. Science 350, aab4077 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  46. Fedorov, V.D., Themeli, M. & Sadelain, M. PD-1- and CTLA-4-based inhibitory chimeric antigen receptors (iCARs) divert off-target immunotherapy responses. Sci. Transl. Med. 5, 215ra172 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. Kloss, C.C., Condomines, M., Cartellieri, M., Bachmann, M. & Sadelain, M. Combinatorial antigen recognition with balanced signaling promotes selective tumor eradication by engineered T cells. Nat. Biotechnol. 31, 71–75 (2013).

    Article  CAS  PubMed  Google Scholar 

  48. Morgan, R.A. et al. Case report of a serious adverse event following the administration of T cells transduced with a chimeric antigen receptor recognizing ERBB2. Mol. Ther. 18, 843–851 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Han, K.P. et al. IL-15:IL-15 receptor–α superagonist complex: high-level co-expression in recombinant mammalian cells, purification and characterization. Cytokine 56, 804–810 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Zheng, Y. et al. In vivo targeting of adoptively transferred T cells with antibody- and cytokine-conjugated liposomes. J. Control. Release 172, 426–435 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank K.D. Wittrup (MIT) for the gift of the engineered IL-2-Fc constructs and the Koch Institute Swanson Biotechnology Center for technical support on flow cytometry, IVIS imaging and MALDI mass spectrometry. This work was supported in part by the Ragon Institute of MGH, MIT and Harvard (D.J.I.), the Melanoma Research Alliance (award 306833; D.J.I.), the NIH (Koch Institute Support (core) grant P30-CA14051 from the National Cancer Institute and CA172164; D.J.I.) and the Koch Institute Marble Center for Cancer Nanomedicine (D.J.I.). L.T. was funded by a Cancer Research Institute (CRI) Irvington Postdoctoral Fellowship, and Y.Z. was supported by a National Science fellowship from the Agency for Science, Technology and Research, Singapore. L.T. and Y.-Q.X. were supported by the ISREC Foundation with a donation from the Biltema Foundation and Swiss National Science Foundation (project grant 315230_173243). M.V.M. was supported by NIH grant CA K08166039. D.J.I. is an investigator of the Howard Hughes Medical Institute.

Author information

Authors and Affiliations

Authors

Contributions

L.T., Y.Z., M.B.M. and D.J.I. designed the in vitro and syngeneic mouse experiments; H.C.W. and E.K.J. provided ALT-803; L.T., Y.Z., D.J.I., A.P.C., S.B.K. and M.V.M. designed the studies with the humanized mice; L.T., Y.Z., L.M., M.B.M., Y.-Q.X., N.L., A.P.C. and S.B.K. performed the experiments; L.T., Y.Z., M.B.M. and D.J.I. analyzed the data and wrote the manuscript; and all authors edited the manuscript.

Corresponding authors

Correspondence to Li Tang or Darrell J Irvine.

Ethics declarations

Competing interests

D.J.I., L.T., and Y.Z. are inventors on licensed patents related to the technology described in this manuscript. D.J.I. is a co-founder of Torque Therapeutics, which licensed patents related to this technology.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–17, Supplementary Tables 1–3, and Supplementary Scheme 1 (PDF 3051 kb)

Life Sciences Reporting Summary (PDF 142 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Tang, L., Zheng, Y., Melo, M. et al. Enhancing T cell therapy through TCR-signaling-responsive nanoparticle drug delivery. Nat Biotechnol 36, 707–716 (2018). https://doi.org/10.1038/nbt.4181

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nbt.4181

This article is cited by

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