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:

AAV-mediated delivery of a Sleeping Beauty transposon and an mRNA-encoded transposase for the engineering of therapeutic immune cells

Abstract

Engineering cells for adoptive therapy requires overcoming limitations in cell viability and, in the efficiency of transgene delivery, the duration of transgene expression and the stability of genomic integration. Here we report a gene-delivery system consisting of a Sleeping Beauty (SB) transposase encoded into a messenger RNA delivered by an adeno-associated virus (AAV) encoding an SB transposon that includes the desired transgene, for mediating the permanent integration of the transgene. Compared with lentiviral vectors and with the electroporation of plasmids of transposon DNA or minicircle DNA, the gene-delivery system, which we named MAJESTIC (for ‘mRNA AAV–SB joint engineering of stable therapeutic immune cells’), offers prolonged transgene expression, as well as higher transgene expression, therapeutic-cell yield and cell viability. MAJESTIC can deliver chimeric antigen receptors (CARs) into T cells (which we show lead to strong anti-tumour activity in vivo) and also transduce natural killer cells, myeloid cells and induced pluripotent stem cells with bi-specific CARs, kill-switch CARs and synthetic T-cell receptors.

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: Development of MAJESTIC, a composite mRNA:AAV-SB system for highly efficient generation of therapeutic immune cells.
Fig. 2: Comparison between MAJESTIC and conventional CAR T-cell generation approaches.
Fig. 3: Application of MAJESTIC for delivery of different therapeutic transgenes into T human cells.
Fig. 4: Comparison of MC transposon and MAJESTIC methods.
Fig. 5: Genomic insertion and safe harbour preference of MC transposon and MAJESTIC methods.
Fig. 6: CAR-NK generation via MAJESTIC.
Fig. 7: CAR-monocyte and CAR-MA generation via MAJESTIC.

Similar content being viewed by others

Data availability

The main data supporting the results in this study are available within the paper and its supplementary information. The raw and processed genome-sequence data from the splinkerette experiments are available from the NIH Sequence Read Archive/Gene Expression Omnibus under the accession number GSE220202 (token mtqrayyonvwfzmd). The raw and analysed datasets generated during the study are available from the corresponding author on reasonable request. Source data are provided with this paper.

Code availability

The scripts used to process the insertion site-mapping data are available at https://github.com/stanleyzlam/SB-CAR.

References

  1. Hayes, C. Cellular immunotherapies for cancer. Ir. J. Med. Sci. 190, 41–57 (2021).

    Article  PubMed  Google Scholar 

  2. Laskowski, T. & Rezvani, K. Adoptive cell therapy: living drugs against cancer. J. Exp. Med. 217, e20200377 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. 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  ADS  CAS  PubMed  Google Scholar 

  4. 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 

  5. Upadhaya, S. et al. The clinical pipeline for cancer cell therapies. Nat. Rev. Drug Discov. 20, 503–504 (2021).

    Article  CAS  PubMed  Google Scholar 

  6. Verdegaal, E. M. et al. Neoantigen landscape dynamics during human melanoma–T cell interactions. Nature 536, 91–95 (2016).

    Article  ADS  CAS  PubMed  Google Scholar 

  7. Johnson, L. A. et al. Gene therapy with human and mouse T-cell receptors mediates cancer regression and targets normal tissues expressing cognate antigen. Blood 114, 535–546 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Thomas, R. et al. NY-ESO-1 based immunotherapy of cancer: current perspectives. Front. Immunol. 9, 947 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  9. Chu, J. et al. CS1-specific chimeric antigen receptor (CAR)-engineered natural killer cells enhance in vitro and in vivo antitumor activity against human multiple myeloma. Leukemia 28, 917–927 (2014).

    Article  CAS  PubMed  Google Scholar 

  10. Genssler, S. et al. Dual targeting of glioblastoma with chimeric antigen receptor-engineered natural killer cells overcomes heterogeneity of target antigen expression and enhances antitumor activity and survival. Oncoimmunology 5, e1119354 (2016).

    Article  PubMed  Google Scholar 

  11. Zhang, Q. et al. Synergistic effects of cabozantinib and EGFR-specific CAR-NK-92 cells in renal cell carcinoma. J. Immunol. Res. 2017, 6915912 (2017).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  12. Kruschinski, A. et al. Engineering antigen-specific primary human NK cells against HER-2 positive carcinomas. Proc. Natl Acad. Sci. USA 105, 17481–17486 (2008).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  13. Liu, E. et al. Cord blood NK cells engineered to express IL-15 and a CD19-targeted CAR show long-term persistence and potent antitumor activity. Leukemia 32, 520–531 (2018).

    Article  CAS  PubMed  Google Scholar 

  14. Chu, Y. et al. Targeting CD20+ aggressive B-cell non-Hodgkin lymphoma by anti-CD20 CAR mRNA-modified expanded natural killer cells in vitro and in NSG mice. Cancer Immunol. Res. 3, 333–344 (2015).

    Article  CAS  PubMed  Google Scholar 

  15. Li, Y., Hermanson, D. L., Moriarity, B. S. & Kaufman, D. S. Human iPSC-derived natural killer cells engineered with chimeric antigen receptors enhance anti-tumor activity. Cell Stem Cell 23, 181–192 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Klichinsky, M. et al. Human chimeric antigen receptor macrophages for cancer immunotherapy. Nat. Biotechnol. 38, 947–953 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Zhang, L. et al. Pluripotent stem cell-derived CAR-macrophage cells with antigen-dependent anti-cancer cell functions. J. Hematol. Oncol. 13, 153 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Themeli, M. et al. Generation of tumor-targeted human T lymphocytes from induced pluripotent stem cells for cancer therapy. Nat. Biotechnol. 31, 928–933 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Di Stasi, A. et al. Inducible apoptosis as a safety switch for adoptive cell therapy. N. Engl. J. Med. 365, 1673–1683 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Shah, N. N. et al. Bispecific anti-CD20, anti-CD19 CAR T cells for relapsed B cell malignancies: a phase 1 dose escalation and expansion trial. Nat. Med. 26, 1569–1575 (2020).

    Article  CAS  PubMed  Google Scholar 

  21. Morgan, R. A. & Boyerinas, B. Genetic modification of T cells. Biomedicines 4, 9 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Ellis, J. Silencing and variegation of gammaretrovirus and lentivirus vectors. Hum. Gene Ther. 16, 1241–1246 (2005).

    Article  CAS  PubMed  Google Scholar 

  23. Ciuffi, A. Mechanisms governing lentivirus integration site selection. Curr. Gene Ther. 8, 419–429 (2008).

    Article  CAS  PubMed  Google Scholar 

  24. Hacein-Bey-Abina, S. et al. LMO2-associated clonal T cell proliferation in two patients after gene therapy for SCID-X1. Science 302, 415–419 (2003).

    Article  ADS  CAS  PubMed  Google Scholar 

  25. Hacein-Bey-Abina, S. et al. Insertional oncogenesis in 4 patients after retrovirus-mediated gene therapy of SCID-X1. J. Clin. Invest. 118, 3132–3142 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Kustikova, O. et al. Clonal dominance of hematopoietic stem cells triggered by retroviral gene marking. Science 308, 1171–1174 (2005).

    Article  ADS  CAS  PubMed  Google Scholar 

  27. Zhou, S. et al. Evaluating the safety of retroviral vectors based on insertional oncogene activation and blocked differentiation in cultured thymocytes. Mol. Ther. 24, 1090–1099 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Naso, M. F., Tomkowicz, B., Perry, W. L. 3rd & Strohl, W. R. Adeno-associated virus (AAV) as a vector for gene therapy. BioDrugs 31, 317–334 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  30. Dai, X. et al. One-step generation of modular CAR-T cells with AAV-Cpf1. Nat. Methods 16, 247–254 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Nahmad, A. D. et al. Frequent aneuploidy in primary human T cells after CRISPR-Cas9 cleavage. Nat. Biotechnol. 40, 1807–1813 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Papathanasiou, S. et al. Whole chromosome loss and genomic instability in mouse embryos after CRISPR-Cas9 genome editing. Nat. Commun. 12, 5855 (2021).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  33. Zuccaro, M. V. et al. Allele-specific chromosome removal after Cas9 cleavage in human embryos. Cell 183, 1650–1664.e15 (2020).

    Article  CAS  PubMed  Google Scholar 

  34. Wilson, M. H., Coates, C. J. & George, A. L.Jr PiggyBac transposon-mediated gene transfer in human cells. Mol. Ther. 15, 139–145 (2007).

    Article  CAS  PubMed  Google Scholar 

  35. Doherty, J. E. et al. Hyperactive piggyBac gene transfer in human cells and in vivo. Hum. Gene Ther. 23, 311–320 (2012).

    Article  CAS  PubMed  Google Scholar 

  36. Kebriaei, P., Izsvak, Z., Narayanavari, S. A., Singh, H. & Ivics, Z. Gene therapy with the Sleeping Beauty transposon system. Trends Genet. 33, 852–870 (2017).

    Article  CAS  PubMed  Google Scholar 

  37. Sebastian-Martin, A., Barrioluengo, V. & Menendez-Arias, L. Transcriptional inaccuracy threshold attenuates differences in RNA-dependent DNA synthesis fidelity between retroviral reverse transcriptases. Sci. Rep. 8, 627 (2018).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  38. Monjezi, R. et al. Enhanced CAR T-cell engineering using non-viral Sleeping Beauty transposition from minicircle vectors. Leukemia 31, 186–194 (2017).

    Article  CAS  PubMed  Google Scholar 

  39. Singh, H. et al. Manufacture of clinical-grade CD19-specific T cells stably expressing chimeric antigen receptor using Sleeping Beauty system and artificial antigen presenting cells. PLoS ONE 8, e64138 (2013).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  40. Magnani, C. F. et al. Sleeping Beauty-engineered CAR T cells achieve antileukemic activity without severe toxicities. J. Clin. Invest. 130, 6021–6033 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Chicaybam, L. et al. CAR T cells generated using Sleeping Beauty transposon vectors and expanded with an EBV-transformed lymphoblastoid cell line display antitumor activity in vitro and in vivo. Hum. Gene Ther. 30, 511–522 (2019).

    Article  CAS  PubMed  Google Scholar 

  42. Geurts, A. M. et al. Gene mutations and genomic rearrangements in the mouse as a result of transposon mobilization from chromosomal concatemers. PLoS Genet. 2, e156 (2006).

    Article  PubMed  PubMed Central  Google Scholar 

  43. Liu, M. A. A comparison of plasmid DNA and mRNA as vaccine technologies. Vaccines 7, 37 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Ye, L. et al. In vivo CRISPR screening in CD8 T cells with AAV–Sleeping Beauty hybrid vectors identifies membrane targets for improving immunotherapy for glioblastoma. Nat. Biotechnol. 37, 1302–1313 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Mates, L. et al. Molecular evolution of a novel hyperactive Sleeping Beauty transposase enables robust stable gene transfer in vertebrates. Nat. Genet. 41, 753–761 (2009).

    Article  CAS  PubMed  Google Scholar 

  46. Francois, A. et al. Accurate titration of infectious AAV particles requires measurement of biologically active vector genomes and suitable controls. Mol. Ther. Methods Clin. Dev. 10, 223–236 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Ghorashian, S. et al. Enhanced CAR T cell expansion and prolonged persistence in pediatric patients with ALL treated with a low-affinity CD19 CAR. Nat. Med. 25, 1408–1414 (2019).

    Article  CAS  PubMed  Google Scholar 

  48. Chicaybam, L. et al. Transposon-mediated generation of CAR-T cells shows efficient anti B-cell leukemia response after ex vivo expansion. Gene Ther. 27, 85–95 (2020).

    Article  CAS  PubMed  Google Scholar 

  49. Xia, X., Zhang, Y., Zieth, C. R. & Zhang, S. C. Transgenes delivered by lentiviral vector are suppressed in human embryonic stem cells in a promoter-dependent manner. Stem. Cells Dev. 16, 167–176 (2007).

    Article  CAS  PubMed  Google Scholar 

  50. Kolacsek, O. et al. Reliable transgene-independent method for determining Sleeping Beauty transposon copy numbers. Mob. DNA 2, 5 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Schneider, D. et al. A tandem CD19/CD20 CAR lentiviral vector drives on-target and off-target antigen modulation in leukemia cell lines. J. Immunother. Cancer 5, 42 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  52. Straathof, K. C. et al. An inducible caspase 9 safety switch for T-cell therapy. Blood 105, 4247–4254 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Sadelain, M., Papapetrou, E. P. & Bushman, F. D. Safe harbours for the integration of new DNA in the human genome. Nat. Rev. Cancer 12, 51–58 (2011).

    Article  PubMed  Google Scholar 

  54. Querques, I. et al. A highly soluble Sleeping Beauty transposase improves control of gene insertion. Nat. Biotechnol. 37, 1502–1512 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Roth, S. L., Malani, N. & Bushman, F. D. Gammaretroviral integration into nucleosomal target DNA in vivo. J. Virol. 85, 7393–7401 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Hou, A. J., Chen, L. C. & Chen, Y. Y. Navigating CAR-T cells through the solid-tumour microenvironment. Nat. Rev. Drug Discov. 20, 531–550 (2021).

    Article  CAS  PubMed  Google Scholar 

  57. Dolgin, E. Cancer-eating immune cells kitted out with CARs. Nat. Biotechnol. 38, 509–511 (2020).

    Article  CAS  PubMed  Google Scholar 

  58. Marofi, F. et al. CAR-NK cell: a new paradigm in tumor immunotherapy. Front. Oncol. 11, 673276 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Bailey, S. R. & Maus, M. V. Gene editing for immune cell therapies. Nat. Biotechnol. 37, 1425–1434 (2019).

    Article  CAS  PubMed  Google Scholar 

  60. Tang, X. et al. First-in-man clinical trial of CAR NK-92 cells: safety test of CD33-CAR NK-92 cells in patients with relapsed and refractory acute myeloid leukemia. Am. J. Cancer Res. 8, 1083–1089 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Auwerx, J. The human leukemia cell line, THP-1: a multifacetted model for the study of monocyte–macrophage differentiation. Experientia 47, 22–31 (1991).

    Article  CAS  PubMed  Google Scholar 

  62. Enache, O. M. et al. Cas9 activates the p53 pathway and selects for p53-inactivating mutations. Nat. Genet. 52, 662–668 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Miskey, C. et al. Engineered Sleeping Beauty transposase redirects transposon integration away from genes. Nucleic Acids Res. 50, 2807–2825 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Roth, T. L. et al. Reprogramming human T cell function and specificity with non-viral genome targeting. Nature 559, 405–409 (2018).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  65. Fu, Y. et al. High-frequency off-target mutagenesis induced by CRISPR-Cas nucleases in human cells. Nat. Biotechnol. 31, 822–826 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Anderson, K. R. et al. CRISPR off-target analysis in genetically engineered rats and mice. Nat. Methods 15, 512–514 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Frock, R. L. et al. Genome-wide detection of DNA double-stranded breaks induced by engineered nucleases. Nat. Biotechnol. 33, 179–186 (2015).

    Article  CAS  PubMed  Google Scholar 

  68. Tsai, S. Q. et al. GUIDE-seq enables genome-wide profiling of off-target cleavage by CRISPR-Cas nucleases. Nat. Biotechnol. 33, 187–197 (2015).

    Article  CAS  PubMed  Google Scholar 

  69. Cameron, P. et al. Mapping the genomic landscape of CRISPR-Cas9 cleavage. Nat. Methods 14, 600–606 (2017).

    Article  CAS  PubMed  Google Scholar 

  70. Tsai, S. Q. et al. CIRCLE-seq: a highly sensitive in vitro screen for genome-wide CRISPR-Cas9 nuclease off-targets. Nat. Methods 14, 607–614 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Takata, M. et al. Homologous recombination and non-homologous end-joining pathways of DNA double-strand break repair have overlapping roles in the maintenance of chromosomal integrity in vertebrate cells. EMBO J. 17, 5497–5508 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Zhang, W. et al. Hybrid adeno-associated viral vectors utilizing transposase-mediated somatic integration for stable transgene expression in human cells. PLoS ONE 8, e76771 (2013).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  73. Clauss, J. et al. Efficient non-viral T-cell engineering by Sleeping Beauty minicircles diminishing DNA toxicity and miRNAs silencing the endogenous T-cell receptors. Hum. Gene Ther. 29, 569–584 (2018).

    Article  CAS  PubMed  Google Scholar 

  74. Jin, Z. et al. The hyperactive Sleeping Beauty transposase SB100X improves the genetic modification of T cells to express a chimeric antigen receptor. Gene Ther. 18, 849–856 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Kovac, A. et al. RNA-guided retargeting of Sleeping Beauty transposition in human cells. eLife 9, e53868 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  76. Muther, N., Noske, N. & Ehrhardt, A. Viral hybrid vectors for somatic integration—are they the better solution? Viruses 1, 1295–1324 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  77. Balciunas, D. et al. Harnessing a high cargo-capacity transposon for genetic applications in vertebrates. PLoS Genet. 2, e169 (2006).

    Article  PubMed  PubMed Central  Google Scholar 

  78. Ye, L. et al. A genome-scale gain-of-function CRISPR screen in CD8 T cells identifies proline metabolism as a means to enhance CAR-T therapy. Cell Metab. 34, 595–614.e14 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Friedrich, M. J. et al. Genome-wide transposon screening and quantitative insertion site sequencing for cancer gene discovery in mice. Nat. Protoc. 12, 289–309 (2017).

    Article  CAS  PubMed  Google Scholar 

  80. Bushnell, B. 9th Annual Genomics of Energy & Environment Meeting (Walnut Creek, 2014).

  81. Martin, M. Cutadapt removes adapter sequences from high-throughput sequencing reads. EMBnet J. 17, 1 (2011).

  82. Kim, D., Paggi, J. M., Park, C., Bennett, C. & Salzberg, S. L. Graph-based genome alignment and genotyping with HISAT2 and HISAT-genotype. Nat. Biotechnol. 37, 907–915 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Li, H. et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics 25, 2078–2079 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  84. Quinlan, A. R. & Hall, I. M. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics 26, 841–842 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Lawrence, M. et al. Software for computing and annotating genomic ranges. PLoS Comput. Biol. 9, e1003118 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Akalin, A., Franke, V., Vlahovicek, K., Mason, C. E. & Schubeler, D. Genomation: a toolkit to summarize, annotate and visualize genomic intervals. Bioinformatics 31, 1127–1129 (2015).

    Article  CAS  PubMed  Google Scholar 

  87. Yin, T., Cook, D. & Lawrence, M. ggbio: an R package for extending the grammar of graphics for genomic data. Genome Biol. 13, R77 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank all members of the Chen Laboratory and of various entities at the University of Yale for discussions. We thank various Yale core facilities for technical support. In particular, we thank K. Tang and P. Renauer for technical assistance on Illumina sequencing and data analysis. We also thank C. Miskey for sharing processed sequence files from ref. 54. S.C. is supported by a Yale SBI/Genetics Startup Fund and by grants from NIH/NCI/NIDA (DP2CA238295, R01CA231112, R33CA225498, RF1DA048811), DoD (W81XWH-20-1-0072, W81XWH-21-10514), Alliance for Cancer Gene Therapy, Sontag Foundation (DSA), Pershing Square Sohn Cancer Research Alliance, Yale Cancer Center Pilot Award, Dexter Lu Gift, Ludwig Family Foundation, and Chenevert Family Foundation. S.Z.L. is supported by Yale College Fellowships.

Author information

Authors and Affiliations

Authors

Contributions

S.C. conceived the study. L. Ye designed the experiments with assistance from S.Z.L. L. Ye performed most experiments, with assistance from S.Z.L., L.P., K.S., Q.L., Y.Z. and P.C. L. Yang, K.S. and Y.Z. performed revision experiments with technical supervision by L.P. S.Z.L. and K.S. analysed next-generation sequencing data. L. Ye, S.Z.L., L. Yang and S.C. prepared the paper, with inputs from all authors. S.C. secured funding and supervised the work.

Corresponding author

Correspondence to Sidi Chen.

Ethics declarations

Competing interests

A patent related to this study was filed by Yale University (inventors: S.C., L.Y. and S.Z.L.) and licensed to Cellinfinity Bio, a Yale biotech start-up founded by S.C. S.C. is also a (co)founder of EvolveImmune Tx, Chen Consulting, Chen Tech and NumericGlobal, all unrelated to this study. The other authors declare no competing interests.

Peer review

Peer review information

Nature Biomedical Engineering thanks Harjeet Singh and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available.

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 MAJESTIC CAR generation and optimization.

a, Schematic of AAV-SB-CD22.CAR, AAV-SB-BCMA.CAR, and SB100X constructs and key procedures: mRNA in vitro transcription, AAV production, mRNA electroporation, flow cytometry, and kill assay. b, Representative flow cytometry plots of AAV-SB-BCMA.CAR CD4 (left) and CD8 (right) T cells show the percentage of CAR-expressing cells. CD4 cells are defined as CD3+ and CD8 cells. c, Quantification of CD22.CAR T cell ratio of (Fig. 1b, c). d, Quantification of BCMA.CAR T ratio in human CD4 and CD8 T cells. In this figure, for optimization of conditions, each assay was done with one donor with three technical replicates. Donor 2 T cells were used in this figure.

Source data

Extended Data Fig. 2 Timepoint optimization, SB100X transposase mRNA titration, and MAJESTIC CAR T yield.

a, Quantification of CD22.CAR T cell ratio for CD4 T cells that were transduced with AAV-SB-CD22.CAR virus at various time points. b, Quantification of CD22.CAR T cell ratio for CD8 T cells that were transduced with AAV-SB-CD22.CAR virus at various time points. c-d, Representative flow cytometry plots of CD22.CAR T cells produced via AAV-SB and a titrated serial of SB100X mRNA. (c) CD8 T cells. (d) CD4 T cells. e, Quantification of the cell viability of CD8 T cells. f, Quantification of CAR T cells. g, Flow cytometry plots of CAR T cell ratios before and after sorting. h, CD22.CAR T cell yield quantification (yield = total viable cell count x CAR-positive percentage). Cells were split into 3 technical replicates after electroporation. Yield is calculated for each technical replicate separately. i, CAR+ T cell generation efficiency (CAR+%) of MAJESTIC using Neon and Maxcyte approaches. In this figure, for optimization of conditions, each assay was done with one donor with three technical replicates. Donor 2 and donor 0286 T cells were used in this figure.

Source data

Extended Data Fig. 3 Vector-copy-number quantification, immune-marker profiling, and functionality testing of MAJESTIC-produced CD8 CAR T cells.

a-c, Vector copy number (VCN) quantification of MAJESTIC-manufactured CAR-T cells. Purified CAR T cells were collected for DNA extraction after three weeks of mRNA electroporation and viral transduction. (a) left arm probe, (b) right arm probe, (c) left panel: left arm probe, right panel: right arm probe. d, SB100X transposase excision efficiency evaluation. Left panel: left arm probe, right panel: right arm probe. e, Cytolysis analysis of NAML6-GL (NAML6 with GFP and luciferase reporters) cancer cells that were co-cultured with Lenti-CD22.CAR and AAV-SB-CD22.CAR T cells. CAR-Ts were seeded at various effector:target (E:T) ratios, and luciferase imaging was performed at two time points (16h and 40h). f, Cytolysis analysis of MM.1R-GL (MM.1R with GFP and luciferase reporters) cancer cells that were co-cultured with Lenti-BCMA.CAR and AAV-SB-BCMA.CAR T cells. CAR-Ts were seeded at various effector : target (E:T) ratios, and luciferase imaging was performed at two time points (16h and 40h). g, Exhaustion and memory marker expression in CD22-CAR and HER2-CAR T cells before and post transfection. Unpaired t tests were performed to evaluate statistical significance. h, Bioluminescent density of NSG mice that were injected with NALM6-GL cancer cells and with CD22-CAR therapy (n = 7 mice per group). i, Quantification of total luminescence for (h). n = 7 mice. Two-way ANOVA with multiple comparisons tests was performed to evaluate statistical significance. j, Survival curve of NALM6-GL-induced leukemia-bearing NSG mice that treated with PBS, untreated CD8 T cells, and AAV-SB-CD22.CAR T cells. Log-rank (Mantel-Cox) tests were performed to evaluate statistical significance. Donor 0007, 4003, 5003, 003C, 0286 T cells were used in this figure. Significance notes: ns - not significant; * p < 0.05, ** p < 0.01, *** p < 0.001, **** p < 0.0001.

Source data

Extended Data Fig. 4 Generation of various therapeutic immune cells by MAJESTIC.

a, Quantification of the HER2.CAR T cell viability. b, Quantification of HER2.CAR-positive CD8 T cells. c, Representative flow-cytometry plots of EGFRvIII.CAR-NK92 cells were produced via either the SB system or lentiviral transduction. d, Quantification of (c). e, Quantification of CD19.20.CAR T cells. f, Flow cytometry detection of CD19 and CD20 expression in NALM6-GL cells. g, Cytolysis analysis of NALM6-GL cancer cells that were co-cultured with lenti-CD19.20.CAR and AAV-SB-CD19.20.CAR T cells. Donor 2 and donor VP2 T cells were used in this figure.

Source data

Extended Data Fig. 5 Generation of various therapeutic immune cells by MAJESTIC.

a, Representative flow cytometry plots of NY-ESO-1 T cells. b, Quantification of (a). c, Quantification of the CD22.CAR.iCasp T cell viability. d, Representative flow cytometry plots of CD22.CAR.iCasp9 T cells. e, Quantification of (d). f, Quantification of the CD22.CAR.iCasp9 T cells post antigen-specific cancer cells stimulation. In this figure, each assay has three technical replicates, donor 601c and donor 02 T cells were used in this figure.

Source data

Extended Data Fig. 6 Gene-delivery efficiency comparison of MC-SB/SB100X mRNA with the MAJESTIC system.

a, Flow-cytometry histogram overlays and bar plots of cell viability post-electroporation as measured with 7-AAD staining. b, Flow-cytometry data of CD22.CAR T cells from human primary CD3 T cells produced by plasmid transposon plasmid, transposon MC, transposon MC with mRNA-transposase, and MAJESTIC. c, Quantification of (b). d, Yield calculations (yield = CAR% * total viable cell count). All conditions started with an equal amount of primary T cells per replicate. Three CAR% replicates were averaged and then multiplied by the average of 2 cell count replicates. Left panel, total T cell count; Right panel, total CAR+ T cell count. e, Quantification of flow-cytometry data of CD22.CAR T cells from four human PBMCs (same data as Fig. 4e–f, plotted in dot-whisker plots).

Source data

Supplementary information

Supplementary Information

Supplementary Fig. 1 and Table 1.

Reporting Summary

Peer Review File

Supplementary Data 1

List of key oligos.

Supplementary Data 2

Statistical data for Supplementary Fig. 1.

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

Ye, L., Lam, S.Z., Yang, L. et al. AAV-mediated delivery of a Sleeping Beauty transposon and an mRNA-encoded transposase for the engineering of therapeutic immune cells. Nat. Biomed. Eng 8, 132–148 (2024). https://doi.org/10.1038/s41551-023-01058-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41551-023-01058-6

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