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:

DNA scaffolds enable efficient and tunable functionalization of biomaterials for immune cell modulation

Abstract

Biomaterials can improve the safety and presentation of therapeutic agents for effective immunotherapy, and a high level of control over surface functionalization is essential for immune cell modulation. Here, we developed biocompatible immune cell-engaging particles (ICEp) that use synthetic short DNA as scaffolds for efficient and tunable protein loading. To improve the safety of chimeric antigen receptor (CAR) T cell therapies, micrometre-sized ICEp were injected intratumorally to present a priming signal for systemically administered AND-gate CAR-T cells. Locally retained ICEp presenting a high density of priming antigens activated CAR T cells, driving local tumour clearance while sparing uninjected tumours in immunodeficient mice. The ratiometric control of costimulatory ligands (anti-CD3 and anti-CD28 antibodies) and the surface presentation of a cytokine (IL-2) on ICEp were shown to substantially impact human primary T cell activation phenotypes. This modular and versatile biomaterial functionalization platform can provide new opportunities for immunotherapies.

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

Fig. 1: Polymeric micro-/nanoparticles with surface DNA scaffolds for protein presentation allow versatile modulation of immune cell therapies.
Fig. 2: DNA scaffolds enable efficient loading of multiple therapeutic proteins on particle surfaces at precisely tunable ratios.
Fig. 3: Compatibility of DNA-scaffolded PLGA particles for in vivo applications.
Fig. 4: Local activation of AND-gate CAR-T cell for tumour killing by intratumoral injection of ICEp presenting a priming antigen.
Fig. 5: ICEp, capable of versatile and precisely controlled modulatory signals, regulates T cell characteristics during ex vivo expansion.

Similar content being viewed by others

Data availability

The original data of the gel electrophoresis images are publicly available at Dryad Digital Repository (https://doi.org/10.7272/Q6CC0XXJ). Any other raw data that support the plots within this paper are available from the authors upon reasonable request.

References

  1. Riley, R. S., June, C. H., Langer, R. & Mitchell, M. J. Delivery technologies for cancer immunotherapy. Nat. Rev. Drug Discov. 18, 175–196 (2019).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  3. Chen, D. S. & Mellman, I. Oncology meets immunology: the cancer-immunity cycle. Immunity 39, 1–10 (2013).

    Article  Google Scholar 

  4. June, C. H. & Sadelain, M. Chimeric antigen receptor therapy. N. Engl. J. Med. 379, 64–73 (2018).

    Article  CAS  Google Scholar 

  5. Gajewski, T. F., Schreiber, H. & Fu, Y. X. Innate and adaptive immune cells in the tumor microenvironment. Nat. Immunol. 14, 1014–1022 (2013).

    Article  CAS  Google Scholar 

  6. Brudno, J. N. & Kochenderfer, J. N. Toxicities of chimeric antigen receptor T cells: recognition and management. Blood 127, 3321–3330 (2016).

    Article  CAS  Google Scholar 

  7. Riches, J. C. et al. T cells from CLL patients exhibit features of T-cell exhaustion but retain capacity for cytokine production. Blood 121, 1612–1621 (2013).

    Article  CAS  Google Scholar 

  8. Gust, J. et al. Endothelial activation and blood–brain barrier disruption in neurotoxicity after adoptive immunotherapy with CD19 CAR-T cells. Cancer Discov. 7, 1404–1419 (2017).

    Article  CAS  Google Scholar 

  9. Maus, M. V. et al. T cells expressing chimeric antigen receptors can cause anaphylaxis in humans. Cancer Immunol. Res. 1, 26–31 (2013).

    Article  CAS  Google Scholar 

  10. Sagiv-Barfi, I. et al. Eradication of spontaneous malignancy by local immunotherapy. Sci. Transl. Med. 10, eaan4488 (2018).

    Article  Google Scholar 

  11. Adachi, K. et al. IL-7 and CCL19 expression in CAR-T cells improves immune cell infiltration and CAR-T cell survival in the tumor. Nat. Biotechnol. 36, 346–351 (2018).

    Article  CAS  Google Scholar 

  12. 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  Google Scholar 

  13. 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  Google Scholar 

  14. Tang, H. et al. Facilitating T cell infiltration in tumor microenvironment overcomes resistance to PD-L1 blockade. Cancer Cell 29, 285–296 (2016).

    Article  CAS  Google Scholar 

  15. Shah, N. J. et al. An injectable bone marrow-like scaffold enhances T cell immunity after hematopoietic stem cell transplantation. Nat. Biotechnol. 37, 293–302 (2019).

    Article  CAS  Google Scholar 

  16. Chen, Q. et al. In situ sprayed bioresponsive immunotherapeutic gel for post-surgical cancer treatment. Nat. Nanotechnol. 14, 89–97 (2019).

    Article  CAS  Google Scholar 

  17. Ye, Y. et al. A melanin-mediated cancer immunotherapy patch. Sci. Immunol. 2, eaan5692 (2017).

    Article  Google Scholar 

  18. Jiang, W. et al. Designing nanomedicine for immuno-oncology. Nat. Biomed. Eng. 1, 0029 (2017).

    Article  CAS  Google Scholar 

  19. Stephan, S. B. et al. Biopolymer implants enhance the efficacy of adoptive T-cell therapy. Nat. Biotechnol. 33, 97–101 (2015).

    Article  CAS  Google Scholar 

  20. Roybal, K. T. et al. Precision tumor recognition by T cells with combinatorial antigen-sensing circuits. Cell 164, 770–779 (2016).

    Article  CAS  Google Scholar 

  21. Brenner, M. J., Cho, J. H., Wong, N. M. L. & Wong, W. W. Synthetic biology: immunotherapy by design. Annu. Rev. Biomed. Eng. 20, 95–118 (2018).

    Article  CAS  Google Scholar 

  22. Yu, C. S., Xi, J. C., Li, M., An, M. & Liu, H. P. Bioconjugate strategies for the induction of antigen-specific tolerance in autoimmune diseases. Bioconjug. Chem. 29, 719–732 (2018).

    Article  CAS  Google Scholar 

  23. Shao, K. et al. Nanoparticle-based immunotherapy for cancer. ACS Nano 9, 16–30 (2015).

    Article  CAS  Google Scholar 

  24. Li, Y. & Kurlander, R. J. Comparison of anti-CD3 and anti-CD28-coated beads with soluble anti-CD3 for expanding human T cells: differing impact on CD8 T cell phenotype and responsiveness to restimulation. J. Transl. Med. 8, 104 (2010).

    Article  Google Scholar 

  25. Slifka, M. K. & Amanna, I. J. Role of multivalency and antigenic threshold in generating protective antibody responses. Front. Immunol. 10, 956 (2019).

    Article  CAS  Google Scholar 

  26. Hartwell, B. L. et al. Multivalent nanomaterials: learning from vaccines and progressing to antigen-specific immunotherapies. J. Pharm. Sci. 104, 346–361 (2015).

    Article  CAS  Google Scholar 

  27. Shaw, A. et al. Binding to nanopatterned antigens is dominated by the spatial tolerance of antibodies. Nat. Nanotechnol. 14, 398–398 (2019).

    Article  CAS  Google Scholar 

  28. Shi, B. Y. et al. Challenges in DNA delivery and recent advances in multifunctional polymeric DNA delivery systems. Biomacromolecules 18, 2231–2246 (2017).

    Article  CAS  Google Scholar 

  29. Sapsford, K. E. et al. Functionalizing nanoparticles with biological molecules: developing chemistries that facilitate nanotechnology. Chem. Rev. 113, 1904–2074 (2013).

    Article  CAS  Google Scholar 

  30. Iyisan, B. & Landfester, K. Modular approach for the design of smart polymeric nanocapsules. Macromol. Rapid Commun. 40, e1800577 (2019).

    Article  Google Scholar 

  31. Huang, X., Lai, Y. F., Braun, G. B. & Reich, N. O. Modularized gold nanocarriers for TAT-mediated delivery of siRNA. Small 13, 1602473 (2017).

    Article  Google Scholar 

  32. Cheung, A. S., Zhang, D. K. Y., Koshy, S. T. & Mooney, D. J. Scaffolds that mimic antigen-presenting cells enable ex vivo expansion of primary T cells. Nat. Biotechnol. 36, 160–169 (2018).

    Article  CAS  Google Scholar 

  33. Zhou, J., Patel, T. R., Fu, M., Bertram, J. P. & Saltzman, W. M. Octa-functional PLGA nanoparticles for targeted and efficient siRNA delivery to tumors. Biomaterials 33, 583–591 (2012).

    Article  CAS  Google Scholar 

  34. Douglas, S. M., Bachelet, I. & Church, G. M. A logic-gated nanorobot for targeted transport of molecular payloads. Science 335, 831–834 (2012).

    Article  CAS  Google Scholar 

  35. Stephanopoulos, N. Hybrid nanostructures from the self-assembly of proteins and DNA. Chem 6, 364–405 (2020).

    Article  CAS  Google Scholar 

  36. Hu, Y. & Niemeyer, C. M. From DNA nanotechnology to material systems engineering. Adv. Mater. 31, e1806294 (2019).

    Article  Google Scholar 

  37. Wang, S. et al. DNA-functionalized metal–organic framework nanoparticles for intracellular delivery of proteins. J. Am. Chem. Soc. 141, 2215–2219 (2019).

    Article  CAS  Google Scholar 

  38. Peterson, A. M. & Heemstra, J. M. Controlling self-assembly of DNA–polymer conjugates for applications in imaging and drug delivery. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 7, 282–297 (2015).

    Article  CAS  Google Scholar 

  39. Wang, Z. G., Li, N., Wang, T. & Ding, B. Surface-guided chemical processes on self-assembled DNA nanostructures. Langmuir 34, 14954–14962 (2018).

    Article  CAS  Google Scholar 

  40. Allahyari, M. & Mohit, E. Peptide/protein vaccine delivery system based on PLGA particles. Hum. Vaccin. Immunother. 12, 806–828 (2016).

    Article  Google Scholar 

  41. Schmid, D. et al. T cell-targeting nanoparticles focus delivery of immunotherapy to improve antitumor immunity. Nat. Commun. 8, 1747 (2017).

    Article  Google Scholar 

  42. Zamecnik, C. R., Lowe, M. M., Patterson, D. M., Rosenblum, M. D. & Desai, T. A. Injectable polymeric cytokine-binding nanowires are effective tissue-specific immunomodulators. ACS Nano 11, 11433–11440 (2017).

    Article  CAS  Google Scholar 

  43. Blanco, E., Shen, H. & Ferrari, M. Principles of nanoparticle design for overcoming biological barriers to drug delivery. Nat. Biotechnol. 33, 941–951 (2015).

    Article  CAS  Google Scholar 

  44. Rodriguez, P. L. et al. Minimal ‘self’ peptides that inhibit phagocytic clearance and enhance delivery of nanoparticles. Science 339, 971–975 (2013).

    Article  CAS  Google Scholar 

  45. Gattinoni, L. et al. A human memory T cell subset with stem cell-like properties. Nat. Med. 17, 1290–1297 (2011).

    Article  CAS  Google Scholar 

  46. Arenas-Ramirez, N. et al. Improved cancer immunotherapy by a CD25-mimobody conferring selectivity to human interleukin-2. Sci. Transl. Med. 8, 367ra166 (2016).

    Article  Google Scholar 

  47. Tang, L. et al. Enhancing T cell therapy through TCR-signaling-responsive nanoparticle drug delivery. Nat. Biotechnol. 36, 707–716 (2018).

    Article  CAS  Google Scholar 

  48. Singha, S. et al. Peptide–MHC-based nanomedicines for autoimmunity function as T-cell receptor microclustering devices. Nat. Nanotechnol. 12, 701–710 (2017).

    Article  CAS  Google Scholar 

  49. Huang, B. et al. Active targeting of chemotherapy to disseminated tumors using nanoparticle-carrying T cells. Sci. Transl. Med. 7, 291ra294 (2015).

    Article  Google Scholar 

  50. Smith, T. T. et al. In situ programming of leukaemia-specific T cells using synthetic DNA nanocarriers. Nat. Nanotechnol. 12, 813–820 (2017).

    Article  CAS  Google Scholar 

  51. Kuai, R., Ochyl, L. J., Bahjat, K. S., Schwendeman, A. & Moon, J. J. Designer vaccine nanodiscs for personalized cancer immunotherapy. Nat. Mater. 16, 489–496 (2017).

    Article  CAS  Google Scholar 

  52. Wang, C., Ye, Y., Hu, Q., Bellotti, A. & Gu, Z. Tailoring biomaterials for cancer immunotherapy: emerging trends and future outlook. Adv. Mater. 29, 1606036 (2017).

    Article  Google Scholar 

Download references

Acknowledgements

Portions of this work were supported by the National Institutes of Health Grants 5T32GM008155 and 1U54CA244438. We thank Z. Gartner for DNA synthesis, S. Habelitz for DLS analysis, C. Hayzelden for SEM imaging, B. Hann for IVIS imaging, K. Shokat for Tecan plate reader and C. Zamecnik and A. Li for helpful discussion. X.H. was supported by a UCSF programme for breakthrough biomedical research (PBBR) postdoctoral independent research grant and a Li foundation fellowship. J.Z.W. was supported by a Genentech Pre-Doctoral Fellowship. R.C. was supported by National Institute of General Medical Sciences (NIGMS) Medical Scientist Training Program no. T32GM007618.

Author information

Authors and Affiliations

Authors

Contributions

X.H., J.Z.W., R.C., K.T.R., W.A.L. and T.A.D. designed the experiments and interpreted the results. X.H., J.Z.W., R.C., Z.L., C.E.B., R.H.-L., I.S., E.G. and W.Y. performed the experiments, and D.M.P. contributed to material designs and synthesis. X.H. analysed the data and drafted the manuscript. X.H., J.Z.W., R.C., I.S., W.A.L. and T.A.D. edited the manuscript.

Corresponding authors

Correspondence to Wendell A. Lim or Tejal A. Desai.

Ethics declarations

Competing interests

T.A.D., W.A.L., X.H., J.Z.W. and R.C. are inventors of pending patents related to the technology described in the manuscript. Z.L., C.E.B., R.H.-L., I.S., E.G., D.M.P., W.Y. and K.T.R. declare no competing interests.

Additional information

Peer review information Nature Nanotechnology thanks the anonymous reviewers 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.

Supplementary information

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

Huang, X., Williams, J.Z., Chang, R. et al. DNA scaffolds enable efficient and tunable functionalization of biomaterials for immune cell modulation. Nat. Nanotechnol. 16, 214–223 (2021). https://doi.org/10.1038/s41565-020-00813-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41565-020-00813-z

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