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:

Bioinstructive implantable scaffolds for rapid in vivo manufacture and release of CAR-T cells

Abstract

Despite their clinical success, chimeric antigen receptor (CAR)-T cell therapies for B cell malignancies are limited by lengthy, costly and labor-intensive ex vivo manufacturing procedures that might lead to cell products with heterogeneous composition. Here we describe an implantable Multifunctional Alginate Scaffold for T Cell Engineering and Release (MASTER) that streamlines in vivo CAR-T cell manufacturing and reduces processing time to a single day. When seeded with human peripheral blood mononuclear cells and CD19-encoding retroviral particles, MASTER provides the appropriate interface for viral vector-mediated gene transfer and, after subcutaneous implantation, mediates the release of functional CAR-T cells in mice. We further demonstrate that in vivo-generated CAR-T cells enter the bloodstream and control distal tumor growth in a mouse xenograft model of lymphoma, showing greater persistence than conventional CAR-T cells. MASTER promises to transform CAR-T cell therapy by fast-tracking manufacture and potentially reducing the complexity and resources needed for provision of this type of therapy.

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
Fig. 2: MASTER promotes activation and retrovirus-mediated transduction of primary human T cells.
Fig. 3: MASTER-mediated gene transfer generates highly functional CAR-T cells.
Fig. 4: Subcutaneously implanted MASTER generates and releases fully functional CAR-T cells in a xenograft model of lymphoma.
Fig. 5: Subcutaneously implanted MASTER outperforms intravenously administered CAR-T cells in a rechallenge model of lymphoma.
Fig. 6: MASTER and conventional CAR-T cells exhibit equal anti-tumor efficacy against established tumor in vivo.

Similar content being viewed by others

Data availability

The authors declare that all data supporting the findings of this study are available within the paper and its extended data and supplementary information files. Source data are provided with this paper.

References

  1. Lee, D. W. et al. T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: a phase 1 dose-escalation trial. Lancet 385, 517–528 (2015).

    Article  CAS  PubMed  Google Scholar 

  2. 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 

  3. Enblad, G. et al. A phase I/IIa trial using CD19-targeted third-generation CAR T cells for lymphoma and leukemia. Clin. Cancer Res. 24, 6185–6194 (2018).

    Article  CAS  PubMed  Google Scholar 

  4. Ramos, C. A., Savoldo, B. & Dotti, G. CD19-CAR trials. Cancer J. 20, 112–118 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Prasad, V. Tisagenlecleucel—the first approved CAR-T-cell therapy: implications for payers and policy makers. Nat. Rev. Clin. Oncol. 15, 11–12 (2018).

    Article  PubMed  Google Scholar 

  6. Sharma, P., King, G. T., Shinde, S. S., Purev, E. & Jimeno, A. Axicabtagene ciloleucel for the treatment of relapsed/refractory B-cell non-Hodgkin’s lymphomas. Drugs Today 54, 187–198 (2018).

    Article  CAS  Google Scholar 

  7. Tang, J., Hubbard-Lucey, V. M., Pearce, L., O’Donnell-Tormey, J. & Shalabi, A. The global landscape of cancer cell therapy. Nat. Rev. Drug Discov. 17, 465–466 (2018).

    Article  CAS  PubMed  Google Scholar 

  8. Bach, P. B. National coverage analysis of CAR-T therapies—policy, evidence, and payment. N. Engl. J. Med. 379, 1396–1398 (2018).

    Article  PubMed  Google Scholar 

  9. Hernandez, I. Analysis determines true cost for CAR T-cell therapy. Healio https://www.healio.com/news/hematology-oncology/20180426/analysis-determines-true-cost-for-car-tcell-therapy (2018).

  10. Leyfman, Y. Chimeric antigen receptors: unleashing a new age of anti-cancer therapy. Cancer Cell Int. 18, 182 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Caffrey, M. With approval of CAR T-cell therapy comes the next challenge: payer coverage. Am. J. Manag. Care 24, SP35–SP36 (2018).

    Google Scholar 

  12. Jackson, H. J., Rafiq, S. & Brentjens, R. J. Driving CAR T-cells forward. Nat. Rev. Clin. Oncol. 13, 370–383 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Shah, N. N. & Fry, T. J. Mechanisms of resistance to CAR T cell therapy. Nat. Rev. Clin. Oncol. 16, 372–385 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. 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 

  15. Maude, S. L. et al. Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. N. Engl. J. Med. 378, 439–448 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Ghassemi, S. et al. Reducing ex vivo culture improves the antileukemic activity of chimeric antigen receptor (CAR) T cells. Cancer Immunol. Res. 6, 1100–1109 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Xu, Y. & Dotti, G. Selection bias: maintaining less-differentiated T cells for adoptive immunotherapy. J. Clin, Invest. 126, 35–37 (2016).

    Article  Google Scholar 

  18. Gattinoni, L., Klebanoff, C. A. & Restifo, N. P. Paths to stemness: building the ultimate antitumour T cell. Nat. Rev. Cancer 12, 671–684 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Mock, U. et al. Automated manufacturing of chimeric antigen receptor T cells for adoptive immunotherapy using CliniMACS prodigy. Cytotherapy 18, 1002–1011 (2016).

    Article  CAS  PubMed  Google Scholar 

  20. Lu, T. L. et al. A rapid cell expansion process for production of engineered autologous CAR-T cell therapies. Hum. Gene Ther. Methods 27, 209–218 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Depil, S., Duchateau, P., Grupp, S. A., Mufti, G. & Poirot, L. ‘Off-the-shelf’ allogeneic CAR T cells: development and challenges. Nat. Rev. Drug Discov. 19, 185–199 (2020).

  22. Kim, D. W. & Cho, J.-Y. Recent advances in allogeneic CAR-T cells. Biomolecules 10, 263 (2020).

  23. 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  PubMed  PubMed Central  Google Scholar 

  24. Agarwal, S., Weidner, T., Thalheimer, F. B. & Buchholz, C. J. In vivo generated human CAR T cells eradicate tumor cells. Oncoimmunology 8, e1671761 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Dautzenberg, I. J. C., Rabelink, M. J. W. E. & Hoeben, R. C. The stability of envelope-pseudotyped lentiviral vectors. Gene Ther. 28, 89–104 (2021).

    Article  CAS  PubMed  Google Scholar 

  26. Rosenblum, D., Joshi, N., Tao, W., Karp, J. M. & Peer, D. Progress and challenges towards targeted delivery of cancer therapeutics. Nat. Commun. 9, 1410 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  27. Lee, K. Y. & Mooney, D. J. Alginate: properties and biomedical applications. Prog. Polym. Sci. 37, 106–126 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Andersen, T., Auk-Emblem, P. & Dornish, M. 3D cell culture in alginate hydrogels. Microarrays (Basel) 4, 133–161 (2015).

    Article  CAS  Google Scholar 

  29. Hwang, C. M. et al. Fabrication of three-dimensional porous cell-laden hydrogel for tissue engineering. Biofabrication 2, 035003 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  30. Agarwalla, P. et al. Scaffold‐mediated static transduction of T cells for CAR‐T cell therapy. Adv. Healthc. Mater. 9, e2000275 (2020).

  31. Savina, I. N., Ingavle, G. C., Cundy, A. B. & Mikhalovsky, S. V. A simple method for the production of large volume 3D macroporous hydrogels for advanced biotechnological, medical and environmental applications. Sci. Rep. 6, 21154 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Shapiro, L. & Cohen, S. Novel alginate sponges for cell culture and transplantation. Biomaterials 18, 583–590 (1997).

    Article  CAS  PubMed  Google Scholar 

  33. Brudno, Y. et al. In vivo targeting through click chemistry. ChemMedChem 10, 617–620 (2015).

    Article  CAS  PubMed  Google Scholar 

  34. 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 

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

    Article  CAS  PubMed  Google Scholar 

  36. Moody, C. T., Palvai, S. & Brudno, Y. Click cross-linking improves retention and targeting of refillable alginate depots. Acta Biomater. 112, 112–121 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Savoldo, B. et al. CD28 costimulation improves expansion and persistence of chimeric antigen receptor-modified T cells in lymphoma patients. J. Clin. Invest. 121, 1822–1826 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Stock, S. et al. Influence of retronectin-mediated T-cell activation on expansion and phenotype of CD19-specific chimeric antigen receptor T cells. Hum. Gene Ther. 29, 1167–1182 (2018).

    Article  CAS  PubMed  Google Scholar 

  39. Hori, Y., Winans, A. M. & Irvine, D. J. Modular injectable matrices based on alginate solution/microsphere mixtures that gel in situ and co-deliver immunomodulatory factors. Acta Biomater. 5, 969–982 (2009).

    Article  CAS  PubMed  Google Scholar 

  40. Diaconu, I. et al. Inducible caspase-9 selectively modulates the toxicities of CD19-specific chimeric antigen receptor-modified T cells. Mol. Ther. 25, 580–592 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Xu, Y. et al. Closely related T-memory stem cells correlate with in vivo expansion of CAR.CD19-T cells and are preserved by IL-7 and IL-15. Blood 123, 3750–3759 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Ramos, C. A. et al. Clinical and immunological responses after CD30-specific chimeric antigen receptor-redirected lymphocytes. J. Clin. Invest. 127, 3462–3471 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  43. Ramos, C. A. et al. Clinical responses with T lymphocytes targeting malignancy-associated κ light chains. J. Clin. Invest. 126, 2588–2596 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Vu, H. N., Ramsey, J. D. & Pack, D. W. Engineering of a stable retroviral gene delivery vector by directed evolution. Mol. Ther 16, 308–314 (2008).

    Article  CAS  PubMed  Google Scholar 

  45. Tay, R. E., Richardson, E. K. & Toh, H. C. Revisiting the role of CD4+ T cells in cancer immunotherapy—new insights into old paradigms. Cancer Gene Ther. 28, 5–17 (2021).

    Article  CAS  PubMed  Google Scholar 

  46. Wen, H. et al. Preclinical safety evaluation of chimeric antigen receptor-modified T cells against CD19 in NSG mice. Ann. Transl. Med. 7, 735 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Zhao, Z. et al. Structural design of engineered costimulation determines tumor rejection kinetics and persistence of CAR T cells. Cancer Cell 28, 415–428 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. McLellan, A. D. & Ali Hosseini Rad, S. M. Chimeric antigen receptor T cell persistence and memory cell formation. Immunol. Cell Biol. 97, 664–674 (2019).

    Article  CAS  PubMed  Google Scholar 

  49. Sterner, R. C. & Sterner, R. M. CAR-T cell therapy: current limitations and potential strategies. Blood Cancer J. 11, 69 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  50. 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 

  51. Liu, L. et al. Enhanced CAR-T activity against established tumors by polarizing human T cells to secrete interleukin-9. Nat. Commun. 11, 5902 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Gostick, J. et al. OpenPNM: a pore network modeling package. Comput. Sci. Eng. 18, 60–74 (2016).

    Article  CAS  Google Scholar 

  53. Vera, J. et al. T lymphocytes redirected against the κ light chain of human immunoglobulin efficiently kill mature B lymphocyte-derived malignant cells. Blood 108, 3890–3897 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Xu, Y. et al. Glycolysis determines dichotomous regulation of T cell subsets in hypoxia. J. Clin. Invest. 126, 2678–2688 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  55. Ramos, C. A. et al. Anti-CD30 CAR-T cell therapy in relapsed and refractory Hodgkin lymphoma. J. Clin. Oncol. 38, 3794–3804 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by the North Carolina Biotechnology Center Flash Grant 2019-FLG-3812; by the National Center for Advancing Translational Sciences and the National Institutes of Health through grant awards R37-CA260223, UL1-TR002489, R01-CA193140, R21-CA229938, T32-CA196589 and R25-NS094093; and by start-up funds provided by the University of North Carolina at Chapel Hill, North Carolina State University at Raleigh, the Lineberger Cancer Center and the Ross M. Lampe Endowed Chair. We thank the North Carolina State University College of Veterinary Medicine staff for proper care of animals used in experiments and valuable resources on training. We also thank the North Carolina State University flow cytometry core and J. Mohammed for training and guidance on flow cytometry analysis. We are grateful to T. Xianming for assistance with power analysis and to C. Clifford for evaluating histology samples. SEM and X-ray CT images were taken at the Analytical Instrumentation Facility at North Carolina State University, which is supported by the State of North Carolina and the National Science Foundation (award ECCS-1542015). The Analytical Instrumentation Facility is a member of the North Carolina Research Triangle Nanotechnology Network, a site in the National Nanotechnology Coordinated Infrastructure. The authors acknowledge the use of the Cellular and Molecular Imaging Facility at North Carolina State University, which is supported by the State of North Carolina and the National Science Foundation. Schematics were created with BioRender.

Author information

Authors and Affiliations

Authors

Contributions

P.A. conceived of the study, designed and performed experiments, analyzed data and wrote the paper. Y.B. conceived of the study, analyzed data and wrote the paper. E.A.O., S.A., K.F. and A.J. prepared experimental materials and performed experiments. F.S.L. and G.D. contributed to the design of experiments and to writing and editing the paper. All authors discussed the results and implications and commented on the manuscript at all stages.

Corresponding author

Correspondence to Yevgeny Brudno.

Ethics declarations

Competing interests

P.A., G.D. and Y.B. are inventors on patents related to the use of biomaterials for generation of CAR-T cell therapeutics. Y.B. receives an industry-sponsored research grant related to CAR-T cell therapeutic technology (unrelated to this work). G.D. is a paid consultant for Bellicum Pharmaceuticals, Tessa Therapeutics and Catamaran. The other authors declare no competing interests.

Peer review

Peer review information

Nature Biotechnology thanks Jeffrey Hubbell, Prasad Adusumilli and the other, anonymous, reviewer(s) 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 Quantitative characterization of MASTER scaffold structure.

A) Scanned volume of MASTER (left) with a colored plane indicating the cross-section seen on the right. In the cross sections a brighter value indicates a higher density (scaffolds) and a darker value a lower density (air porosity). B) Relative frequency of pores of different dimensions. C) The aspect ratio of the pores showing most of the pores has an oblong shape. An aspect ratio of 1 corresponds to a sphere and close to 0 corresponds to a flat plane or stick. D) The surface area as a function of volume plotted. The total surface area inside of MASTER is roughly 810 mm2 E) Connectivity of sample showing most of the pores are connected to 0-3 other pores with a very few pores (around 6%) have more connections than 9.

Source data

Extended Data Fig. 2 Confocal images of GFP-expressing T cells within MASTER scaffold.

3D confocal micrograph showing distribution of GFP + T cells in AF647 labeled MASTER at 10X (A) and 40X (B) magnification. This experiment was repeated twice independently with similar results.

Source data

Extended Data Fig. 3 IL-2 loaded onto MASTER released in a sustained manner over five days in vitro and retained its bioactivity.

(A) Cumulative release of IL-2 from MASTER as quantified by ELISA assay. Data represent mean ± SD of three independent samples (B) Bioactivity of IL2 released at 24 hours as assessed by proliferation of CFSE stained T cells.

Source data

Extended Data Fig. 4 IL-2 promotes lymphocyte proliferation, but not transduction efficiency of MASTER.

(A) MASTER and MASTER without IL-2 was seeded with PBMCs and virus and number of cells were counted 5 days post transduction. *p < 0.05, two tailed unpaired t test, (B) CAR.19 expression in T cells 72 h post transduction. *p < 0.05, two-tailed unpaired t test. Data represent median of n = 3 biologically independent samples.

Source data

Extended Data Fig. 5 MASTER functions as an efficient T cell-release system.

A) Schematic of in vitro release study. B) Percent of cells released from scaffold. Data in represent mean ± SD of n = 3 independent samples.

Source data

Extended Data Fig. 6 Biocompatibility of MASTER and its components.

Representative images of H&E-stained sections of five major organs and implantation site four weeks after subcutaneous implant of MASTER, MASTER + mouse PBMCs + GFP-encoding gamma retrovirus and untreated controls in C57Bl6/J immunocompetent mice. Data is representative of three biologically independent animals.

Extended Data Fig. 7 MASTER loaded with PBMCs and retrovirus does not transduce host cells.

A) In vitro transwell model mimicking the in vivo system. B) GFP expression in fibroblast cells seeded on the bottom of transwell plate. Data represent mean ± SEM of n = 3 biologically independent samples.

Source data

Extended Data Fig. 8 Characterization of host cells infiltrating MASTER.

A) Timeline of experiment B) Representative FACS plot showing efficient engraftment of human PBMCs (Hu-CD45 + CD3+) in blood. C-D) Different subsets of mouse and human cells that infiltrated MASTER. Cells were gated on live cells. Data in d-e represent mean ± SEM and median of three biologically independent samples. E) Phenotype of the engrafted human T cells that infiltrated into the scaffold. Cells were gated on human-CD45 + CD3 + cells. Data represent mean ± SEM of four biologically independent samples F) FACS plot showing no GFP expression in cells infiltrating MASTER, in blood and in the skin surrounding the scaffold.

Source data

Extended Data Fig. 9 Similar numbers of exhausted cells in blood of mice with conventional and MASTER-generated CAR-T cells.

Immunophenotypic composition of CAR-T cells in blood of mice treated with conventionally expanded CAR-T cells i.v. (A) or MASTER (B) at day 12 and 22 post tumor inoculation. Data in a represent mean ± SD of six experimental replicates. Data in b represent mean ± SEM of five experimental replicates.

Source data

Extended Data Fig. 10 Subcutaneously implanted MASTER exhibited better control of tumor growth and extended survival compared under stressed dose conditions.

A) Timeline of the study B) In vivo tumor bioluminescence imaging (BLI) of NSG mice (n = 6) treated with MASTER, conventional CAR-T cells, or control non-transduced (NT) cells. Mice were treated with 0.5 ×106 (left), 0.25 ×106 (middle), or 0.125 ×106 (right) CAR T cells. PBMCs seeded onto MASTER were normalized to transduction efficiency and both groups (MASTER and CAR T, i.v) were treated with equivalent number of CAR T cells. C-E) Survival of mice shown as Kaplan- Meier curves. **p < 0.01; ***p < 0.001 Log-rank (Mantel-Cox) test, Gehan-Breslow-Wilcoxon test.

Source data

Supplementary information

Supplementary Information

Supplementary Figs. 1–9.

Reporting Summary.

Supplementary Data 1

Source Data for Supplementary Figures.

Source data

Source Data Fig. 2

Statistical Source Data.

Source Data Fig. 3

Statistical Source Data.

Source Data Fig. 4

Statistical Source Data.

Source Data Fig. 5

Statistical Source Data.

Source Data Fig. 6

Statistical Source Data.

Source Data Extended Data Fig. 1

Statistical Source Data.

Source Data Extended Data Fig. 2

Full-resolution confocal image ×10.

Source Data Extended Data Fig. 2

Full-resolution confocal image ×40.

Source Data Extended Data Fig. 3

Statistical Source Data.

Source Data Extended Data Fig. 4

Statistical Source Data.

Source Data Extended Data Fig. 5

Statistical Source Data.

Source Data Extended Data Fig. 7

Statistical Source Data.

Source Data Extended Data Fig. 8

Statistical Source Data.

Source Data Extended Data Fig. 9

Statistical Source Data.

Source Data Extended Data Fig. 10

Statistical Source Data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Agarwalla, P., Ogunnaike, E.A., Ahn, S. et al. Bioinstructive implantable scaffolds for rapid in vivo manufacture and release of CAR-T cells. Nat Biotechnol 40, 1250–1258 (2022). https://doi.org/10.1038/s41587-022-01245-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41587-022-01245-x

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing