Log in

Subcutaneous biodegradable scaffolds for restimulating the antitumour activity of pre-administered CAR-T cells

  • Article
  • Published:

From Nature Biomedical Engineering

View current issue Submit your manuscript

Abstract

The efficacy of adoptive T-cell therapies based on chimaeric antigen receptors (CARs) is limited by the poor proliferation and persistence of the engineered T cells. Here we show that a subcutaneously injected biodegradable scaffold that facilitates the infiltration and egress of specific T-cell subpopulations, which forms a microenvironment mimicking features of physiological T-cell activation, enhances the antitumour activity of pre-administered CAR-T cells. CAR-T-cell expansion, differentiation and cytotoxicity were driven by the scaffold’s incorporation of co-stimulatory bound ligands and soluble molecules, and depended on the types of co-stimulatory molecules and the context in which they were presented. In mice with aggressive lymphoma, a single, local injection of the scaffold following non-curative CAR-T-cell dosing led to more persistent memory-like T cells and extended animal survival. Injectable biomaterials with optimized ligand presentation may boost the therapeutic performance of CAR-T-cell therapies.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Subscribe and save

Springer+ Basic
EUR 32.99 /Month
  • Get 10 units per month
  • Download Article/Chapter or Ebook
  • 1 Unit = 1 Article or 1 Chapter
  • Cancel anytime
Subscribe now

Buy Now

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1: TES enable CAR-T-cell restimulation in vitro.
Fig. 2: Subcutaneously injected TES are biodegradable and remain functional.
Fig. 3: Injected TES facilitate T-cell infiltration, activation and egress.
Fig. 4: A single local injection of TES enhances systemic antitumour activity in an aggressive liquid tumour model.

Similar content being viewed by others

Data availability

The data supporting the findings in this study are available within the paper and its Supplementary Information. All data generated in this study are available from the corresponding author on reasonable request. Source data are provided with this paper.

References

  1. Levine, B. L., Miskin, J., Wonnacott, K. & Keir, C. Global manufacturing of CAR T cell therapy. Mol. Ther. Methods Clin. Dev. 4, 92–101 (2017).

    Article  CAS  PubMed  Google Scholar 

  2. Majzner, R. G. & Mackall, C. L. Clinical lessons learned from the first leg of the CAR T cell journey. Nat. Med. 25, 1341–1355 (2019).

    Article  CAS  PubMed  Google Scholar 

  3. Hernandez-Lopez, R. A. et al. T cell circuits that sense antigen density with an ultrasensitive threshold. Science 371, 1166–1171 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. James, J. R. Tuning ITAM multiplicity on T cell receptors can control potency and selectivity to ligand density. Sci. Signal. 11, eaan1088 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  5. 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 

  6. Weber, E. W. et al. Transient ‘rest’ restores functionality in exhausted CAR-T cells via epigenetic remodeling. Science 372, eaba1786 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Good, Z. et al. Proliferation tracing with single-cell mass cytometry optimizes generation of stem cell memory-like T cells. Nat. Biotechnol. 37, 259–266 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Lynn, R. C. et al. c-Jun overexpression in CAR T cells induces exhaustion resistance. Nature 576, 293–300 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. 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 

  10. Rafiq, S., Hackett, C. S. & Brentjens, R. J. Engineering strategies to overcome the current roadblocks in CAR T cell therapy. Nat. Rev. Clin. Oncol. 17, 147–167 (2020).

    Article  PubMed  Google Scholar 

  11. Eyquem, J. et al. Targeting a CAR to the TRAC locus with CRISPR/Cas9 enhances tumour rejection. Nature 543, 113–117 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Ma, L. et al. Enhanced CAR–T cell activity against solid tumors by vaccine boosting through the chimeric receptor. Science 365, 162–168 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Reinhard, K. et al. An RNA vaccine drives expansion and efficacy of claudin-CAR-T cells against solid tumors. Science 367, 446–453 (2020).

  14. Satthaporn, S. et al. Dendritic cells are dysfunctional in patients with operable breast cancer. Cancer Immunol. Immunother. 53, 510–518 (2004).

    Article  PubMed  PubMed Central  Google Scholar 

  15. Bandola-Simon, J. & Roche, P. A. Dysfunction of antigen processing and presentation by dendritic cells in cancer. Mol. Immunol. 113, 31–37 (2019).

    Article  CAS  PubMed  Google Scholar 

  16. 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 

  17. Coon, M. E., Stephan, S. B., Gupta, V., Kealey, C. P. & Stephan, M. T. Nitinol thin films functionalized with CAR-T cells for the treatment of solid tumours. Nat. Biomed. Eng. 4, 195–206 (2020).

    Article  CAS  PubMed  Google Scholar 

  18. 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  PubMed  PubMed Central  Google Scholar 

  19. Zhang, D. K. Y., Cheung, A. S. & Mooney, D. J. Activation and expansion of human T cells using artificial antigen-presenting cell scaffolds. Nat. Protoc. https://doi.org/10.1038/s41596-019-0249-0 (2020).

  20. Kim, J. et al. Injectable, spontaneously assembling, inorganic scaffolds modulate immune cells in vivo and increase vaccine efficacy. Nat. Biotechnol. 33, 61–69 (2014).

    Google Scholar 

  21. Li, A. W. et al. A facile approach to enhance antigen response for personalized cancer vaccination. Nat. Mater. 17, 528 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Dellacherie, M. O. et al. Single-shot mesoporous silica rods scaffold for induction of humoral responses against small antigens. Adv. Funct. Mater. 30, 2002448 (2020).

    Article  CAS  Google Scholar 

  23. Deng, Q. et al. Characteristics of anti-CD19 CAR T cell infusion products associated with efficacy and toxicity in patients with large B cell lymphomas. Nat. Med. https://doi.org/10.1038/s41591-020-1061-7 (2020).

  24. Li, W. A. et al. The effect of surface modification of mesoporous silica micro-rod scaffold on immune cell activation and infiltration. Biomaterials 83, 249–256 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Brentjens, R. J. et al. Safety and persistence of adoptively transferred autologous CD19-targeted T cells in patients with relapsed or chemotherapy refractory B-cell leukemias. Blood 118, 4817–4828 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Agarwalla, P. et al. Bioinstructive implantable scaffolds for rapid in vivo manufacture and release of CAR-T cells. Nat. Biotechnol. 40, 1250–1258 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Zhang, D. K. Y. et al. Enhancing CAR-T cell functionality in a patient-specific manner. Nat. Commun. 14, 506 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Cruz, C. R. Y. et al. Infusion of donor-derived CD19-redirected virus-specific T cells for B-cell malignancies relapsed after allogeneic stem cell transplant: a phase 1 study. Blood 122, 2965–2973 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Sockolosky, J. T. et al. Selective targeting of engineered T cells using orthogonal IL-2 cytokine-receptor complexes. Science 359, 1037–1042 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Bauer, C. A. et al. Dynamic Treg interactions with intratumoral APCs promote local CTL dysfunction. J. Clin. Invest. 124, 2425–2440 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Chen, M.-L. et al. Regulatory T cells suppress tumor-specific CD8 T cell cytotoxicity through TGF-β signals in vivo. Proc. Natl Acad. Sci. USA 102, 419–424 (2005).

    Article  CAS  PubMed  Google Scholar 

  32. Rosenberg, S. A. et al. Treatment of 283 consecutive patients with metastatic melanoma or renal cell cancer using high-dose bolus interleukin 2. JAMA 271, 907–913 (1994).

    Article  CAS  PubMed  Google Scholar 

  33. Raeber, M. E., Sahin, D., Karakus, U. & Boyman, O. A systematic review of interleukin-2-based immunotherapies in clinical trials for cancer and autoimmune diseases. EBioMedicine 90, 104539 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Dutcher, J. P. et al. High dose interleukin-2 (Aldesleukin) - expert consensus on best management practices-2014. J. Immunother. Cancer 2, 26 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  35. Grosskopf, A. K. et al. Delivery of CAR-T cells in a transient injectable stimulatory hydrogel niche improves treatment of solid tumors. Sci. Adv. 8, eabn8264 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank A. Najibi for critical reading of the manuscript and D. Neuberg for statistical guidance; A. Cheung, O. Ali, Z. Good, N. Cerri, E. Smith, C. Louvet, C. Wu, M. Pezzone, E. Doherty, H. Ijaz, C. Johnson, D. White, M. Perez, E. Zigon, G. Cuneo and T. Ferrante for many important technical and scientific discussions. This work was supported by the Wyss Institute at Harvard University (D.J.M.), the Food and Drug Administration (grant number 5R01FD006589), and the National Institutes of Health under grant numbers R01 EB015498, R01 CA276459, and U01 CA214369. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health. D.K.Y.Z. acknowledges support from the Canadian Institutes of Health Research (CIHR). J.M.B. acknowledges support from the National Cancer Institute (grant number K00CA234959). Material characterization was performed at the Center for Nanoscale Systems (CNS) at Harvard University, which is supported by the National Science Foundation (grant number 1541979).

Author information

Authors and Affiliations

Authors

Contributions

D.K.Y.Z., J.M.B. and D.J.M. conceived the project. D.K.Y.Z., J.M.B., K.A.-B., Y.L., Y.B., I.d.L., M.C.S. and R.T. performed experiments. D.K.Y.Z. and J.M.B. analysed data and generated figures. D.K.Y.Z., J.M.B. and D.J.M. wrote the paper. All authors commented on the paper.

Corresponding author

Correspondence to David J. Mooney.

Ethics declarations

Competing interests

D.J.M. is an inventor in patent applications Harvard University has filed in relation to the TES technology, which has been licensed to Lyell Immunopharma (US Patent App. 18/072,449). D.J.M. has stock in Lyell Immunopharma. D.J.M, D.K.Y.Z. and J.M.B. are listed as inventors in a patent application based on the findings of this study (WO 2023/147185). The other authors declare no competing interests.

Peer review

Peer review information

Nature Biomedical Engineering thanks **aoyuan Chen, Zhen Gu and Jonathan Schneck for their contribution to the peer review of this work.

Additional information

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

Extended data

Extended Data Fig. 1 TES dictates T-cell proliferation kinetics in vivo.

(a) Naïve, unstimulated, primary human T cells were labeled with CFSE and co-delivered subcutaneously with TES presenting αCD3/αCD28/IL-2 or without any ligands (labeled ‘blank’). T-cell proliferation as indicated by CFSE dilution of cells isolated from TES-containing explants at 5 days post injection. (b,c) Analysis of TES’ T-cell boosting capacity in different tumor burden contexts. Tumor-bearing animals (inoculated with 5 × 105 Raji-luc cells) were treated with either a sub-curative dose of 5 × 105 CAR-T cells or a curative dose of 1 × 106 CAR-T cells. TES were injected subcutaneously 5 days following CAR-T cell dosing, and circulating T cells in the blood were subsequently analyzed. (b) Frequency of CD3+ T cells in circulating blood. (c) Number of CD3+ T cells in the blood following subcutaneous scaffold injection under sub-curative CAR-T dosing conditions. (d, e) The capacity of injected material scaffolds to boost T-cell proliferation was evaluated in a xenograft lymphoma model. Animals were inoculated with 5 × 105 Raji-luc cells and treated 4 days afterwards with a curative dose of 1 × 106 CD19 CAR-T cells. (d) Scaffolds were injected into mice with baseline CAR-T, and human T cells were detected in αCD3/αCD28 presenting TES, while very few human T cells were detected in non-ligand-presenting ‘blank’ TES. Left: representative FACS plots showing enrichment of CD3+ T cells. (e) Expression of CD25 among T cells in day 14-explanted TES. Left: representative FACS histoplots. Data represents n = 3-4 mice per condition and mean ± s.e.m. and is representative of at least two experimental replicates. Comparisons in (d, e) were measured using unpaired Student’s T-tests with Welch’s correction for unequal s.d. Data in (b) represents mean ± s.e.m. of n = 5–7 mice.

Source data

Supplementary information

Supplementary Information

Supplementary figures and table.

Reporting Summary

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

Zhang, D.K.Y., Brockman, J.M., Adu-Berchie, K. et al. Subcutaneous biodegradable scaffolds for restimulating the antitumour activity of pre-administered CAR-T cells. Nat. Biomed. Eng (2024). https://doi.org/10.1038/s41551-024-01216-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41551-024-01216-4

  • Springer Nature Limited

Navigation