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.

  • Review Article
  • Published:

In vivo chimeric antigen receptor (CAR)-T cell therapy

Abstract

Chimeric antigen receptor (CAR)-T cell therapy has transformed the outcomes of patients with haematological malignancies, yet its use is limited by labour-intensive manufacturing, constrained production capacity and variable clinical performance. In vivo CAR-T cell engineering, in which CAR-T cells are generated directly inside the patient’s body, seeks to overcome these challenges by eliminating the need for ex vivo cell processing and complex logistics, as well as improve clinical performance. Recent advances in virology, RNA medicines and nanotechnology have catalysed a radical overhaul of this approach, which uses targeted delivery systems such as lentiviral vectors and lipid nanoparticles to introduce CAR-encoding genetic material into endogenous T cells. Early clinical studies have shown efficient transduction, sustained CAR expression and initial signs of antitumour activity, establishing proof of concept. This Review explores the underlying technologies — including RNA delivered by lipid nanoparticles and engineered viral vectors — and discusses how they are being adapted to develop more broadly applicable, scalable, safe and effective CAR-T cell therapies. By removing the need for ex vivo manipulation and chemotherapeutic conditioning, this strategy could enable the wider application of CAR-T cell therapies not just to blood cancers but to autoimmune diseases for which ex vivo CAR-T cell therapies have shown strong promise, such as systemic lupus erythematosus.

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  : Timeline: major discoveries and milestones leading to clinical translation of in vivo CAR technologies.
Fig. 2: Major in vivo CAR platforms with a focus on the engineering vector and payloads.
Fig. 3: Major in vivo CAR platforms in development, and their mechanism of action.
Fig. 4: Top-line perspective of development of in vivo CAR therapies.
Fig. 5: In vivo engineering platform features inform on target product profile characteristics and therapeutic applicability.
Fig. 6: The rapidly evolving, in vivo CAR therapy ecosystem.

Similar content being viewed by others

References

  1. Leavy, O. The birth of monoclonal antibodies. Nat. Immunol. 17, 13 (2016).

    Article  Google Scholar 

  2. Dumontet, C., Reichert, J. M., Senter, P. D., Lambert, J. M. & Beck, A. Antibody–drug conjugates come of age in oncology. Nat. Rev. Drug. Discov. 22, 641–661 (2023).

    Article  PubMed  CAS  Google Scholar 

  3. Rosenberg, S. A. IL-2: the first effective immunotherapy for human cancer. J. Immunol. 192, 5451–5458 (2014).

    Article  PubMed  CAS  Google Scholar 

  4. Baeuerle, P. A. & Reinhardt, C. Bispecific T-cell engaging antibodies for cancer therapy. Cancer Res. 69, 4941–4944 (2009).

    Article  PubMed  CAS  Google Scholar 

  5. Wolff, J. A. et al. Direct gene transfer into mouse muscle in vivo. Science 247, 1465–1468 (1990).

    Article  PubMed  CAS  Google Scholar 

  6. Pardi, N., Hogan, M. J., Porter, F. W. & Weissman, D. mRNA vaccines—a new era in vaccinology. Nat. Rev. Drug. Discov. 17, 261–279 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  8. Hinrichs, C. & Rosenberg, S. A. Exploiting the curative potential of adoptive T-cell therapy for cancer. Immunol. Rev. 257, 56–71 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. Cappell, K. M. & Kochenderfer, J. N. Long-term outcomes following CAR T cell therapy: what we know so far. Nat. Rev. Clin. Oncol. 20, 359–371 (2023).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  10. Melenhorst, J. J. et al. Decade-long leukaemia remissions with persistence of CD4+ CAR T cells. Nature 602, 503–509 (2022).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  11. US Food and Drug Administration. KYMRIAH. FDA https://www.fda.gov/vaccines-blood-biologics/cellular-gene-therapy-products/kymriah (2017). This label corresponds to the first CAR-T cell product approved.

  12. US Food and Drug Administration. YESCARTA. FDA https://www.fda.gov/vaccines-blood-biologics/cellular-gene-therapy-products/yescarta (2017).

  13. US Food and Drug Administration. TECARTUS. FDA https://www.fda.gov/vaccines-blood-biologics/cellular-gene-therapy-products/tecartus (2020).

  14. US Food and Drug Administration. BREYANZI. FDA https://www.fda.gov/vaccines-blood-biologics/cellular-gene-therapy-products/breyanzi-lisocabtagene-maraleucel (2021).

  15. US Food and Drug Administration. ABECMA. FDA https://www.fda.gov/vaccines-blood-biologics/abecma-idecabtagene-vicleucel (2021).

  16. US Food and Drug Administration. CARVYKTI. FDA https://www.fda.gov/vaccines-blood-biologics/cellular-gene-therapy-products/carvykti (2022).

  17. US Food and Drug Administration. AUCATZYL. FDA https://www.fda.gov/vaccines-blood-biologics/aucatzyl (2024).

  18. Joy, R., Phair, K., O’Hara, R. & Brady, D. Recent advances and current challenges in CAR-T cell therapy. Biotechnol. Lett. 46, 115–126 (2024).

    Article  PubMed  CAS  Google Scholar 

  19. Lonez, C. & Breman, E. Allogeneic CAR-T therapy technologies: has the promise been met? Cells 13, 146 (2024).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Wang, X. et al. Allogeneic CD19-targeted CAR-T therapy in patients with severe myositis and systemic sclerosis. Cell 187, 4890–4904.e9 (2024).

    Article  PubMed  CAS  Google Scholar 

  21. Mougakiakos, D. Allogeneic CAR T cells for autoimmune diseases: a glimpse into the future. Signal. Transduct. Target. Ther. 9, 276 (2024).

    Article  Google Scholar 

  22. Aghajanian, H. et al. Targeting cardiac fibrosis with engineered T cells. Nature 573, 430–433 (2019). This work is the first preclinical proof of concept with CAR-T cells in a fibrotic disease model.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  23. Kansal, R. et al. Sustained B cell depletion by CD19-targeted CAR T cells is a highly effective treatment for murine lupus. Sci. Transl. Med. 11, eaav1648 (2019). This work is the first preclinical proof of concept with CAR-T cells in an autoimmune disease model.

    Article  PubMed  PubMed Central  Google Scholar 

  24. Müller, F. et al. CD19 CAR T-cell therapy in autoimmune disease—a case series with follow up. N. Engl. J. Med. 390, 687–700 (2024).

    Article  PubMed  Google Scholar 

  25. Mougiakakos, D. et al. CD19-targeted CAR T cells in refractory systemic lupus erythematosus. N. Engl. J. Med. 385, 567–569 (2021). This work is the first clinical proof of concept with CAR-T cells in patients with autoimmune disease.

    Article  PubMed  Google Scholar 

  26. Müller, F. et al. CD19-targeted CAR T cells in refractory antisynthetase syndrome. Lancet 401, 815–818 (2023).

    Article  PubMed  Google Scholar 

  27. Haghikia, A. et al. Anti-CD19 CAR T cells for refractory myasthenia gravis. Lancet Neurol. 22, 1104–1105 (2023).

    Article  PubMed  CAS  Google Scholar 

  28. Pecher, A. C. et al. CD19-targeting CAR T cells for myositis and interstitial lung disease associated with antisynthetase syndrome. JAMA 329, 2154–2162 (2023).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  29. Zhang, W. et al. Treatment of systemic lupus erythematosus using BCMA-CD19 compound CAR. Stem Cell Rev. Rep. 17, 2120–2123 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  30. Granit, V. et al. Safety and clinical activity of autologous RNA chimeric antigen receptor T-cell therapy in myasthenia gravis (MG-001): a prospective, multicentre, open-label, non-randomised phase 1b/2a study. Lancet Neurol. 22, 578–590 (2023).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  31. Fischbach, F. et al. CD19-targeted chimeric antigen receptor T cell therapy in two patients with multiple sclerosis. Med 5, 550–558.e2 (2024).

    Article  PubMed  CAS  Google Scholar 

  32. Oliver-Caldes, A. First report of CART treatment in AL amyloidosis and relapsed/refractory multiple myeloma. J. Immunother. Cancer 9, e003783 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Korell, F. et al. First third-generation CAR T cell application targeting CD19 for the treatment of systemic IgM AL amyloidosis with underlying marginal zone lymphoma. Biomark. Res. 14, 11–91 (2023).

    Google Scholar 

  34. Qin, C. et al. Anti-BCMA CAR T-cell therapy CT103A in relapsed or refractory AQP4-IgG seropositive neuromyelitis optica spectrum disorders: phase 1 trial interim results. Signal. Transduct. Target. Ther. 8, 5 (2023).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Tobias, A. & Grego, A. Hematopoietic stem cell transplantation and cellular therapies for autoimmune diseases: overview and future considerations from the Autoimmune Diseases Working Party (ADWP) of the European Society for Blood and Marrow Transplantation (EBMT). Bone Marrow Transplant. 57, 1055–1062 (2022).

    Article  Google Scholar 

  36. Muraro, P. A. et al. Autologous haematopoietic stem cell transplantation for treatment of multiple sclerosis. Nat. Rev. Neurol. 13, 391–405 (2017). This review discusses clinical evidence of immune reset by a radical procedure — high-dose chemotherapy followed by rescue with autologous stem cell transplantation — thus supporting similar approaches with the aim to elicit durable, drug-free responses in autoimmunity.

    Article  PubMed  CAS  Google Scholar 

  37. Brudno, J. N. & Kochenderfer, J. N. Recent advances in CAR T-cell toxicity: mechanisms, manifestations and management. Blood Rev. Mar. 34, 45–55 (2018).

    Article  Google Scholar 

  38. Lee, D. W. ASTCT consensus grading for cytokine release syndrome and neurologic toxicity associated with immune effector cells. Biol. Blood Marrow Transplant. 25, 625–638 (2019).

    Article  PubMed  CAS  Google Scholar 

  39. Santomasso, B. D. et al. Management of immune-related adverse events in patients treated with chimeric antigen receptor T-cell therapy: ASCO guideline. J. Clin. Oncol. 39, 3978–3992 (2021).

    Article  PubMed  CAS  Google Scholar 

  40. Hines, M. R. et al. Immune effector cell-associated hemophagocytic lymphohistiocytosis-like syndrome. Transplant. Cell. Ther. 29, 438.e1–438.e16 (2023).

    Article  PubMed  Google Scholar 

  41. Mikhael, J., Fowler, J. & Shah, N. Chimeric antigen receptor T-cell therapies: barriers and solutions to access. JCO Oncol. Pract. 18, 800–807 (2022). This review describes major hurdles to successfully deploying ex vivo engineered CAR-T cells.

    Article  PubMed  Google Scholar 

  42. 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 

  43. Borgert, R. Improving outcomes and mitigating costs associated with CAR T-cell therapy. Am. J. Manag. Care 27, S253–S261 (2021).

    Article  PubMed  Google Scholar 

  44. Lickefett, B. Lymphodepletion—an essential but undervalued part of the chimeric antigen receptor T-cell therapy cycle. Front. Immunol. 14, 1303935 (2023).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  45. Bot, A. et al. Cyclophosphamide and fludarabine conditioning chemotherapy induces a key homeostatic cytokine profile in patients prior to CAR T cell therapy. Blood 126, 4426 (2015).

    Article  Google Scholar 

  46. Warkentin, P. I. Voluntary accreditation of cellular therapies: foundation for the accreditation of cellular therapy (FACT). Cytotherapy 5, 299–305 (2003).

    Article  PubMed  CAS  Google Scholar 

  47. Locke, F. L. et al. Tumor burden, inflammation, and product attributes determine outcomes of axicabtagene ciloleucel in large B-cell lymphoma. Blood Adv. 4, 4898–4911 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  48. Scholler, N. et al. Tumor immune contexture is a determinant of anti-CD19 CAR T cell efficacy in large B cell lymphoma. Nat. Med. 28, 1872–1882 (2022).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  49. Locke, F. L. et al. Impact of tumor microenvironment on efficacy of anti-CD19 CAR T cell therapy or chemotherapy and transplant in large B cell lymphoma. Nat. Med. 30, 507–518 (2024).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  50. Sotillo, E. et al. Convergence of acquired mutations and alternative splicing of CD19 enables resistance to CART-19 immunotherapy. Cancer Discov. 5, 1282–1295 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  51. Plaks, V. et al. CD19 target evasion as a mechanism of relapse in large B-cell lymphoma treated with axicabtagene ciloleucel. Blood 138, 1081–1085 (2021).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  52. Da Via, M. C. et al. Homozygous BCMA gene deletion in response to anti-BCMA CAR T cells in a patient with multiple myeloma. Nat. Med. 27, 616–619 (2021).

    Article  PubMed  Google Scholar 

  53. Smith, T. T. et al. In situ programming of leukaemia-specific T cells using synthetic DNA nanocarriers. Nat. Nanotechnol. 12, 813–820 (2017). This work is the first preclinical report of in vivo cell engineering with non-viral payload delivered via a nanoformulation.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  54. Pfeiffer, A. et al. In vivo generation of human CD19-CAR T cells results in B-cell depletion and signs of cytokine release syndrome. EMBO Mol. Med. 10, e9158 (2018). This work is the first preclinical report of in vivo CAR-T cell engineering utilizing a viral vector-based approach.

    Article  PubMed  PubMed Central  Google Scholar 

  55. Zhou, Q. et al. T-cell receptor gene transfer exclusively to human CD8+ cells enhances tumor cell killing. Blood 120, 4334–4342 (2012).

    Article  PubMed  CAS  Google Scholar 

  56. Mullin, R. Gilead to acquire Kite Pharma for $11.9 billion. Chem. Eng. N. 95, 15 (2017).

    Google Scholar 

  57. Themeli, M., Rivière, I. & Sadelain, M. New cell sources for T cell engineering and adoptive immunotherapy. Cell Stem Cell 16, 357–366 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  58. Jaeger, A. J. CAR-T in the courts. Genet. Eng. Biotechnol. N. 37, 31–33 (2017).

    Article  Google Scholar 

  59. Sanofi. Sanofi R&D Day. sanofi.com https://www.sanofi.com/assets/dotcom/content-app/events/investor-presentation/2023/r-and-d-day-2023/Presentation.pdf (2023).

  60. AstraZeneca to acquire EsoBiotec to advance cell therapy ambition. Astra Zeneca (17 March 2025); https://www.astrazeneca.com/media-centre/press-releases/2025/astrazeneca-to-acquire-esobiotec.html.

  61. Mullard, A. In vivo CAR T cells gain traction, with AbbVie’s US$2.1 billion acquisition of Capstan therapeutics. Nature https://www.nature.com/articles/d41573-025-00131-w (2025).

  62. Klebanoff, K. A., Khong, H. T., Antony, P. A., Palmer, D. C. & Restifo, N. P. Sinks, suppressors and antigen presenters: how lymphodepletion enhances T cell-mediated tumor immunotherapy. Trends Immunol. 26, 111–117 (2005).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  63. Anliker, B. et al. Specific gene transfer to neurons, endothelial cells and hematopoietic progenitors with lentiviral vectors. Nat. Methods 7, 929–935 (2010).

    Article  PubMed  CAS  Google Scholar 

  64. Funke, S. et al. Targeted cell entry of lentiviral vectors. Mol. Ther. 16, 1427–1436 (2008).

    Article  PubMed  CAS  Google Scholar 

  65. Bender, R. R. et al. Receptor-targeted Nipah virus glycoproteins improve cell-type selective gene delivery and reveal a preference for membrane-proximal cell attachment. PLoS Pathog. 12, e1005641 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  66. Charitidis, F. T. et al. CAR gene delivery by T-cell targeted lentiviral vectors is enhanced by rapamycin induced reduction of antiviral mechanisms. Adv. Sci. 10, e2302992 (2023).

    Article  Google Scholar 

  67. Dobson, C. S. et al. Antigen identification and high-throughput interaction mapping by reprogramming viral entry. Nat. Methods 19, 449–460 (2022).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  68. Strebinger, D. et al. Cell type-specific delivery by modular envelope design. Nat. Commun. 14, 5141 (2023).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  69. Frank, A. M. et al. CD8-specific designed ankyrin repeat proteins improve selective gene delivery into human and primate T lymphocytes. Hum. Gene Ther. 31, 679–691 (2020).

    Article  PubMed  CAS  Google Scholar 

  70. 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 

  71. Agarwal, S. et al. In vivo generation of CAR T cells selectively in human CD4+ lymphocytes. Mol. Ther. 28, 1783–1794 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  72. Frank, A. M. et al. Combining T-cell-specific activation and in vivo gene delivery through CD3-targeted lentiviral vectors. Blood Adv. 4, 5702–5715 (2020).

    PubMed  PubMed Central  CAS  Google Scholar 

  73. Huckaby, J. T. et al. Bispecific binder redirected lentiviral vector enables in vivo engineering of CAR-T cells. J. Immunother. Cancer 9, e002737 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  74. Braun, A. H., Frank, A. M., Ho, N. & Buchholz, C. J. Dasatinib is a potent enhancer for CAR T cell generation by CD3-targeted lentiviral vectors. Mol. Ther. Methods Clin. Dev. 28, 90–98 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  75. Kapitza, L. et al. CD62L as target receptor for specific gene delivery into less differentiated human T lymphocytes. Front. Immunol. 14, 1183698 (2023).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  76. Ho, N. et al. In vivo generation of CAR T cells in the presence of human myeloid cells. Mol. Ther. Methods Clin. Dev. 26, 144–156 (2022).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  77. Krug, A. et al. In vivo CAR T cell therapy against angioimmunoblastic T cell lymphoma. J. Exp. Clin. Cancer Res. 43, 262 (2024).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  78. Cunningham, J. et al. In vivo delivery of a CD20 CAR using a CD8-targeted fusosome in southern pig-tail macaques (M. nemestrina) results in B cell depletion. Blood 138, 2769 (2021).

    Article  Google Scholar 

  79. Mullard, A. In vivo CAR T cells move into clinical trials. Nat. Rev. Drug. Discov. 23, 727–730 (2024). This news feature reports the translation of in vivo CAR technology to the clinic.

    Article  PubMed  CAS  Google Scholar 

  80. Nicolai, C. J. et al. In vivo CAR T-cell generation in nonhuman primates using lentiviral vectors displaying a multidomain fusion ligand. Blood 144, 977–987 (2024). This work is the first peer-reviewed publication describing preclinical evidence of activity of in vivo CAR therapy in NHPs.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  81. Andorko, J. I. et al. Targeted in vivo generation of CAR T and NK cells utilizing an engineered lentiviral vector platform. Blood 142, 763 (2023).

    Article  Google Scholar 

  82. Andorko, J. I. et al. Targeted in vivo delivery of genetic medicines utilizing an engineered lentiviral vector platform results in CAR T and NK cell generation. Molecular Therapy https://doi.org/10.1016/j.ymthe.2025.06.036 (2025).

  83. Russell, R. M. et al. Rational design of a detargeted vesiculovirus fusogen to enable targeted in vivo gene delivery. In: 27th American Society of Gene & Cell Therapy (ASGCT) Annual Meeting https://doi.org/10.1016/j.ymthe.2024.04.020 (2024).

  84. Mullen, K. F. et al. In vivo generation of both CAR T cells and CAR NK cells using a CD7 targeted lentiviral vector. In: 27th American Society of Gene & Cell Therapy (ASGCT) Annual Meeting https://doi.org/10.1016/j.ymthe.2024.04.020 (2024).

  85. Grinstein, J. D. First patient dosed with in vivo gene therapy for CAR T. Inside Precision Medicine https://www.insideprecisionmedicine.com/topics/precision-medicine/first-patient-dosed-with-in-vivo-gene-therapy-for-car-t/ (2024).

  86. Kite to acquire Interius BioTherapeutics to advance in vivo platform [press release]. Kite (21 August 2025); https://www.kitepharma.com/news/press-releases/2025/8/kite-to-acquire-interius-biotherapeutics-to-advance-in-vivo-platform.

  87. Michels, K. R. et al. UB-VV100, a novel platform for in vivo engineering of therapeutic anti-CD19 CAR T cells, shows effective T cell transduction, B cell depletion, and tumor control in a humanized murine model. Umoja Biopharma https://www.umoja-biopharma.com/wp-content/uploads/2021/05/Umoja-Biopharma-ASGCT-FINAL.pdf (2021).

  88. Nicolai, C. et al. VivoVec: a novel lentiviral-based in vivo CAR T cell generation platform with viral particle surface engineering incorporating T cell activating and co-stimulatory ligands. Umoja Biopharma https://www.umoja-biopharma.com/wp-content/uploads/2022/11/CN-ASGCT-Poster-Umoja1.pdf (2022).

  89. Michels, K. R. et al. Preclinical proof of concept for VivoVec, a lentiviral-based platform for in vivo CAR T-cell engineering. J. Immunother. Cancer 2023 11, e006292 (2023).

    Article  Google Scholar 

  90. AbbVie and Umoja Biopharma announce strategic collaboration to develop novel in-situ CAR-T cell therapies. Umoja Biopharma https://www.umoja-biopharma.com/news/abbvie-and-umoja-biopharma-announce-strategic-collaboration-to-develop-novel-in-situ-car-t-cell-therapies/ (2024).

  91. Beitz, L. et al. In vivo generation of B cell depleting CAR T cell therapies for treatment of autoimmune diseases. In: ACR Convergence Annual Meeting https://acrabstracts.org/abstract/in-vivo-generation-of-b-cell-depleting-car-t-cell-therapies-for-treatment-of-autoimmune-diseases/ (2024).

  92. Teoh, J. et al. Development of a surface engineered lentiviral vector for in vivo generation of CD22-directed CAR T cells. In: SITC Annual Meeting https://jitc.bmj.com/content/12/Suppl_2/A1267.info (2024).

  93. Friesen, T. et al. Preclinical development of UB-VV400, a surface engineered lentiviral vector for the in vivo generation of CD22-directed CAR T cells. In: ASGCT Annual Meeting Abstract 795 https://www.sciencedirect.com/journal/molecular-therapy/vol/33/issue/4/suppl/S1 (2025).

  94. IASO BIO announces new development partnership with umoja biopharma to develop ex vivo and in vivo cell and gene therapies. Umoja Biopharma https://www.umoja-biopharma.com/news/iaso-bio-announces-new-development-partnership-with-umoja-biopharma-to-develop-ex-vivo-and-in-vivo-cell-and-gene-therapies/ (2024).

  95. Chen, X.-F. et al. In-vivo CAR-T cells produced by specific virus in refractory B cell lymphoma. In: SITC Annual Meeting https://doi.org/10.1136/jitc-2024-SITC2024.1482 (2024).

  96. Chen, X. et al. Lentiviral particle-mediated generation of in-vivo CAR-T cells for treating refractory B-cell lymphoma. In: ISCT Annual Meeting https://www.isct-cytotherapy.org/article/S1465-3249%2825%2900518-3/abstract (2025).

  97. Galvis, L. et al. ENaBL, a novel lentiviral vector platform for in vivo engineering of therapeutic anti-BCMA CAR T cells, shows effective T cell transduction and tumor control in a humanized murine model. In: ASGCT Annual Meeting Abstract 793 https://www.sciencedirect.com/journal/molecular-therapy/vol/33/issue/4/suppl/S1 (2025).

  98. Weerens, L. et al. Development of novel T cell-specific synthetic promoters for safer and more effective in vivo CAR-T cell therapies. In: ASGCT Annual Meeting Abstract 868 https://www.sciencedirect.com/journal/molecular-therapy/vol/33/issue/4/suppl/S1 (2025).

  99. Doix, B. et al. Engineering T cell-specific lentiviral vectors for safe and effective in vivo CAR-T therapy. In: ASGCT Annual Meeting Abstract 879 https://www.sciencedirect.com/journal/molecular-therapy/vol/33/issue/4/suppl/S1 (2025).

  100. EsoBiotec doses first patient in investigator initiated trial of in vivo BCMA CAR-T candidate ESO-T01 for multiple myeloma [press release]. EsoBiotec https://www.esobiotec.com/esobiotec-doses-first-patient-in-investigator-initiated-trial-of-in-vivo-bcma-car-t-candidate-eso-t01-for-multiple-myeloma (2025).

  101. Xu, J. et al. In-vivo B-cell maturation antigen CAR T-cell therapy for relapsed or refractory multiple myeloma. Lancet 406, 228–231 (2025). This is the first clinical proof of concept published in a peer-reviewed journal describing the efficacy and safety of an in vivo CAR technology.

    Article  PubMed  CAS  Google Scholar 

  102. Contrastano, S. G. et al. Potent in vivo transduction by iGPS particles generates CAR T cells with durable anti-tumor activity. Kelonia https://keloniatx.com/wp-content/uploads/2023/05/2023-ASGCT-Kelonia-Presentation.vFINAL.pdf (2023).

  103. Beura, E. T. et al. T cell-specific in vivo transduction with preclinical candidate KLN-1010 generates BCMA directed CAR T cells with potent anti-multiple myeloma activity. Cancer Res. https://doi.org/10.1158/1538-7445.AM2024-48 (2024).

  104. Bender, R. R. et al. Receptor-targeted Nipah virus glycoproteins improve cell-type selective gene delivery and reveal a preference for membrane-proximal cell attachment. PLoS Pathog. 12, 1–28 (2016).

    Article  Google Scholar 

  105. Nakamura, T. et al. Rescue and propagation of fully retargeted oncolytic measles viruses. Nat. Biotech. 23, 209–214 (2005).

    Article  CAS  Google Scholar 

  106. Cordes, N. et al. Anti-CD19 CARs displayed at the surface of lentiviral vector particles promote transduction of target-expressing cells. Mol. Ther. Methods Clin. Dev. 21, 42–53 (2021).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  107. Ruella, M. et al. Induction of resistance to chimeric antigen receptor T cell therapy by transduction of a single leukemic B cell. Nat. Med. 24, 1499–1503 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  108. Amatya, S. et al. Enabling potent T cell delivery and mitigating key off-target concerns of in vivo CAR T lentiviral vectors with the targeted paramyxovirus fusogen system. Blood 144, 2047 (2024).

    Article  Google Scholar 

  109. Cruite, P. et al. Retargeted “fusosomes” for in vivo delivery to T cells. Mol. Ther. 30, 1081 (2022).

    Google Scholar 

  110. Chavan, H. et al. In vitro and in vivo specificity and biodistribution of a novel CD8-targeted fusosome. Blood 142, 3630 (2023).

    Article  Google Scholar 

  111. Trudeau, K. et al. Advancements in manufacturing of CD8- targeted fusosomes enhance transduction of resting T cells in vitro and in vivo. Mol. Ther. 31, 760 (2023).

    Google Scholar 

  112. Chokshi, C. et al. In vivo engineering of hematopoietic stem cells with virus-like particles to generate multi-lineage CAR immune cell therapy for cancer. In: ASGCT annual meeting Abstract 1783 https://www.sciencedirect.com/journal/molecular-therapy/vol/33/issue/4/suppl/S1 (2025).

  113. Vyriad announces strategic collaboration with Novartis to develop in vivo CAR-T cell therapies [press release]. PR Newswire https://www.prnewswire.com/news-releases/vyriad-announces-strategic-collaboration-with-novartis-to-develop-in-vivo-car-t-cell-therapies-302310717.html (2024).

  114. In vivo delivery of CD3-directed CD19-CAR lentivectors leads to the generation of CAR-T and NK-like (CAR-TaNK) cells capable of complete ablation of B cells in the blood, bone marrow, and tissue in NSG-SGM3 CD34+ humanized mice. In: AACR Annual Meeting https://exumabio.com/wp-content/uploads/2023/04/AACR-Poster-2023-Final-2.pdf (2023).

  115. Coillard, A. et al. A novel polymeric gene delivery platform to precisely target and reprogram cells in vivo for haematological malignancies & beyond. In: ASGCT Annual Meeting Abstract 1213 https://www.sciencedirect.com/journal/molecular-therapy/vol/33/issue/4/suppl/S1 (2025).

  116. Jaudoin, C. et al. Lentiviral vector engineering with polymer surface modification for car-T cell therapy. In: ASGCT Annual Meeting Abstract 1214 https://www.sciencedirect.com/journal/molecular-therapy/vol/33/issue/4/suppl/S1 (2025).

  117. Parayath, N. N., Stephan, S. B., Koehne, A. L., Nelson, P. S. & Stephan, M. T. In vitro-transcribed antigen receptor mRNA nanocarriers for transient expression in circulating T cells in vivo. Nat. Commun. 11, 6080 (2020). This seminal preclinical study demonstrates in vivo CAR engineering using mRNA formulated in nanocarriers.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  118. Cullis, P. R. & Hope, M. G. Lipid nanoparticle systems for enabling gene therapies. Mol. Ther. 25, 1467–1475 (2017). This review describes the discovery and development of current-generation LNPs comprising ionizable lipids and facilitating the design of first-in-class RNA nanomedicines.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  119. Kularatne, R. N. The future of tissue-targeted lipid nanoparticle-mediated nucleic acid delivery. Pharmaceuticals 15, 897 (2022).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  120. Park, M. D., Silvin, A., Ginhoux, F. & Merad, M. Macrophages in health and disease. Cell 185, 4259–4279 (2022).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  121. Wang, Y. et al. In vivo programming of myeloid cells by mRNA mediated delivery of novel FcA fusion receptor activates anti-tumor immunity. J. Immunother. Cancer https://doi.org/10.1136/jitc-2022-SITC2022.1237 (2022).

  122. Myeloid Therapeutics enters into agreement with Acuitas Therapeutics for lipid nanoparticle (LNP) system, enabling the first-ever delivery of mRNA-encoded CARs directly to humans [press release]. Myeloid https://myeloidtx.com/myeloid-therapeutics-enters-into-agreement-with-acuitas-therapeutics-for-lipid-nanoparticle-system/ (2022).

  123. Argueta, S. et al. In vivo programmed myeloid cells expressing novel chimeric antigen receptors show potent anti-tumor activity in preclinical solid tumor models. Front. Immunol. https://doi.org/10.3389/fimmu.2024.1501365 (2024).

  124. Argueta, S. et al. Preclinical development of MT-303, a novel LNP-formulated GPC3-specific CAR mRNA, for in vivo programming of monocytes to treat hepatocellular carcinoma. J. Immunother. Cancer https://doi.org/10.1136/jitc-2024-SITC2024.1125 (2024).

  125. Myeloid Therapeutics initiates patient dosing with MT-302, a novel TROP2-targeting RNA CAR, in phase 1 study for advanced or metastatic epithelial tumors [press release]. Myeloid https://myeloidtx.com/myeloid-therapeutics-initiates-patient-dosing-with-mt-302-a-novel-trop2-targeting-rna-car-in-phase-1-study-for-advanced-or-metastatic-epithelial-tumors/ (2023).

  126. Myeloid Therapeutics initiates patient dosing with MT-303, a novel GPC3 targeting RNA CAR, in phase 1 study for advanced hepatocellular carcinoma (HCC) [press release]. Myeloid https://myeloidtx.com/myeloid-therapeutics-initiates-patient-dosing-with-mt-303-a-novel-gpc3-targeting-rna-car-in-phase-1-study-for-advanced-hepatocellular-carcinoma-hcc/ (2024).

  127. Myeloid therapeutics to present at 43rd Annual JP Morgan Healthcare Conference [press release]. Myeloid https://myeloidtx.com/myeloid-therapeutics-to-present-at-43rd-annual-jp-morgan-healthcare-conference/ (2025).

  128. Wang, Y. et al. Novel innate immune receptor-based chimeric antigen receptors for in vivo programming of myeloid cells show potent anti-tumor activity in preclinical solid tumor models. In: ASGCT Annual Meeting Abstract 1727 https://www.sciencedirect.com/journal/molecular-therapy/vol/33/issue/4/suppl/S1 (2025).

  129. Parhiz, H., Atochina-Vasserman, E. N. & Weissman, D. mRNA-based therapeutics: looking beyond COVID-19 vaccines. Lancet 403, 1192–1204 (2024).

    Article  PubMed  CAS  Google Scholar 

  130. Karikó, K. et al. Incorporation of pseudouridine into mRNA yields superior nonimmunogenic vector with increased translational capacity and biological stability. Molecular Therapy 16, 1833–1840 (2008). This report describes the seminal discovery that incorporation of synthetic nucleotides enables the utilization of RNA.

    Article  PubMed  Google Scholar 

  131. Cullis, P. R. & Felgner, P. L. The 60-year evolution of lipid nanoparticles for nucleic acid delivery. Nat. Rev. Drug. Discov. 23, 709–722 (2024).

    Article  PubMed  CAS  Google Scholar 

  132. Rurik, J. G. CAR T cells produced in vivo to treat cardiac injury. Science 375, 91–96 (2022). This is the first preclinical report of in vivo CAR-T cell therapy in a non-oncologic indication.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  133. Hunter et al. A novel product candidate (CPTX2309) for in vivo mRNA engineering of anti-CD19 CAR T cells utilizing novel CD8-targeted lipid nanoparticles. In: American College of Rheumatology Annual Meeting (2024).

  134. Adams, G. et al. In vivo engineering of CAR T cells using a novel targeted LNP-mRNA technology. J. Immunother. Cancer https://doi.org/10.1136/jitc-2023-SITC2023.1202 (2023).

  135. Zhang, Y. et al. Effective engineering of CD8+ T cells from autoimmune disease patients utilizing a CD8-targeted lipid nanoparticle encoding an anti-CD19 CAR mRNA(CPTX2309). In: American College of Rheumatology Annual Meeting (2024).

  136. Franz, A. et al. Profound B Cell depletion and repopulation with predominantly naïve B cells in non-human primates achieved through a novel in vivo CD8-targeted lipid nanoparticle mRNA CAR. In: American College of Rheumatology Annual Meeting (2024).

  137. Hunter, T. et al. In vivo CAR T cell generation for cancer and autoimmune disease. Science 388, 1311–1317 (2025). This work is the first preclinical report in NHPs demonstrating proof of concept with respect to in vivo CAR-T cell generation utilizing a non-viral platform.

    Article  PubMed  CAS  Google Scholar 

  138. Aghajanian, H. et al. A two-infusion regimen with a novel in vivo non-viral chimeric antigen receptor (CAR) achieves up to 90% CD8+ T cell engineering and tissue depletion of target cells in non-human primates (NHPs). In: ASGCT Annual Meeting Abstract 258 https://www.sciencedirect.com/journal/molecular-therapy/vol/33/issue/4/suppl/S1 (2025).

  139. Breda, L. et al. In vivo hematopoietic stem cell modification by mRNA delivery. Science 381, 436–443 (2023).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  140. Immorna Biotherapeutics announces first clinical administration of JCXH-213, a breakthrough mRNA-based in vivo CAR therapy [press release]. Pipeline Review https://pipelinereview.com/immorna-biotherapeutics-announces-first-clinical-administration-of-jcxh-213-a-breakthrough-mrna-based-in-vivo-car-therapy/ (2025).

  141. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/study/NCT06801119 (2025).

  142. Xiao, T. et al. An vivo CAR T producer (HN2301) for autoimmune disease enables B cell depletion in NHP tissues and performs therapeutic effect in an SLE mouse model. In: ASGCT Annual Meeting Abstract 794 https://www.sciencedirect.com/journal/molecular-therapy/vol/33/issue/4/suppl/S1 (2025).

  143. Wang, Q. et al. In vivo CD19 CAR T-cell therapy for refractory systemic lupus erythematosus. N. Engl. J. Med. https://doi.org/10.1056/NEJMc2509522 (2025).

  144. Wesselhoeft, R. A., Kowalski, P. S. & Anderson, D. G. Engineering circular RNA for potent and stable translation in eukaryotic cells. Nat. Commun. 9, 2629 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  145. Wesselhoeft, R. A. et al. RNA circularization diminishes immunogenicity and can extend translation duration in vivo. Mol. Cell 74, 508–520.e4 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  146. Becker, A. M. et al. In situ CAR therapy using oRNATM lipid nanoparticles regresses tumors in mice. In: SITC Annual Meeting https://www.ornatx.com/wp-content/uploads/2022/11/Orna_SITC-2022.pdf (2022).

  147. Orna Therapeutics announces strategic acquisition of ReNAgade therapeutics to solidify leadership in circular RNA therapies. Orna https://www.ornatx.com/orna-therapeutics-announces-strategic-acquisition-of-renagade-therapeutics-to-solidify-leadership-in-circular-rna-therapies/ (2024).

  148. Crabtree, J. et al. In vivo panCAR-mediated depletion of B cells in non-human primates using a circular (oRNA®) anti-CD20 CAR. In: ASH Annual Meeting https://ash.confex.com/ash/2024/webprogram/Paper210256.html (2024).

  149. Beaulieu, A. et al. In vivo transient programming of T cells with Sail’s targeted nanoparticles encapsulating eRNATM-encoded hCD19 CAR achieves deep depletion of B cells in blood and lymphoid tissues in a B-cell repopulating humanized mouse model. In: ASGCT Annual Meeting Abstract 774 https://www.sciencedirect.com/journal/molecular-therapy/vol/33/issue/4/suppl/S1 (2025).

  150. Sharma, K. et al. Long-acting circular RNA CAR drives potent and durable tumor elimination invivo. In: ASGCT Annual Meeting Abstract 781 https://www.sciencedirect.com/journal/molecular-therapy/vol/33/issue/4/suppl/S1 (2025).

  151. Pan, X. et al. In vivo generation of anti-CD19 CAR T cells utilizing circular RNA encapsulated in targeted lipid nanoparticles. In: ASGCT Annual Meeting Abstract 1762 https://www.sciencedirect.com/journal/molecular-therapy/vol/33/issue/4/suppl/S1 (2025).

  152. Lemgart, V. et al. In vitro and in vivo activity of a lipid nanoparticle system for the in vivo generation of CAR T cells. In: ASH Annual Meeting https://doi.org/10.1182/blood-2024-207566 (2024).

  153. Kamoun, W. S. et al. Antitumour activity and tolerability of an EphA2-targeted nanotherapeutic in multiple mouse models. Nat. Biomed. Eng. 3, 264–280 (2019).

    Article  PubMed  CAS  Google Scholar 

  154. Lemgart, V. et al. CD19 CAR T cells generated in vivo by T cell targeted lipid nanoparticles demonstrate robust and durable B cell depletion in non-human primates. In: ASGCT Annual Meeting Abstract 260 https://www.sciencedirect.com/journal/molecular-therapy/vol/33/issue/4/suppl/S1 (2025).

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  156. Pierini, S. et al. Chimeric antigen receptor macrophages (CAR-M) sensitize solid tumors to anti-PD1 immunotherapy. J. Immunother. Cancer https://doi.org/10.1136/jitc-2022-SITC2022.0371 (2022).

  157. Gabitova, L. et al. Pre-clinical development of a CAR monocyte platform for cancer immunotherapy. J. Immunother. Cancer https://doi.org/10.1136/jitc-2022-SITC2022.0318 (2022).

  158. Carisma Therapeutics corporate presentation November [corporate presentation]. Carisma https://ir.carismatx.com/static-files/42a456e9-9007-4574-8aaa-e7c8db3ac852 (2024).

  159. Abdou, Y. et al. PO2-04-08 translational insights from a phase 1, first-in-human (FIH) clinical trial of the anti-HER2 CAR macrophage CT-0508 in participants with HER2 positive metastatic breast cancer and other HER2 overexpressing solid tumors. In: SABCS (2023).

  160. Moderna and Carisma establish collaboration to develop in vivo engineered chimeric antigen receptor monocytes (CAR-M) for oncology [press release]. Mpderna https://investors.modernatx.com/news/news-details/2022/Moderna-and-Carisma-Establish-Collaboration-to-Develop-in-vivo-Engineered-Chimeric-Antigen-Receptor-Monocytes-CAR-M-for-Oncology/default.aspx (2022).

  161. Varghese, B. et al. In vivo CAR-M: redirecting endogenous myeloid cells with mRNA for cancer immunotherapy. J. Immunother. Cancer https://doi.org/10.1136/jitc-2023-SITC2023.1514 (2023).

  162. Bagashev, A. et al. Pre-clinical efficacy of a novel anti-GPC3 in vivo CAR-M for hepatocellular carcinoma. J. Immunother. Cancer https://doi.org/10.1136/jitc-2024-SITC2024.0329 (2024).

  163. Carisma and Moderna expand collaboration to develop two in vivo CAR-M therapies for autoimmune diseases [press release]. Carisma https://ir.carismatx.com/news-releases/news-release-details/carisma-and-moderna-expand-collaboration-develop-two-vivo-car-m (2024).

  164. Carisma Therapeutics provides corporate updates [press release]. Carisma https://ir.carismatx.com/news-releases/news-release-details/carisma-therapeutics-provides-corporate-updates#:~:Text=The%20Company%20will%20assess%20a,value%20of%20all%20our%20assets (2025).

  165. Yan, F. et al. Improved T cell delivery and expression of mRNA using CD8-targeted lipid nanoparticles with Athebody® designed ankyrin repeat proteins (DARPins). In: ASGCT Annual Meeting Abstract 667 https://www.sciencedirect.com/journal/molecular-therapy/vol/33/issue/4/suppl/S1 (2025).

  166. Fang, J. et al. Mobilize LNPs: a novel platform for in vivo targeted delivery of RNA therapeutics to T cells. In: ASGCT Annual Meeting Abstract 483 https://www.sciencedirect.com/journal/molecular-therapy/vol/33/issue/4/suppl/S1 (2025).

  167. Hashiba, K. et al. Optimizing VHH ligand density in T cell-targeted LNPs: an inverted U-shaped relationship for effective in vivo CAR T cell generation. In: ASGCT Annual Meeting Abstract 665 https://www.sciencedirect.com/journal/molecular-therapy/vol/33/issue/4/suppl/S1 (2025).

  168. Khairallah, C. et al. In: ASGCT Annual Meeting Abstract 770 https://www.sciencedirect.com/journal/molecular-therapy/vol/33/issue/4/suppl/S1 (2025).

  169. Sun, J. et al. In vivo CAR-T therapy powered by novel CLAMP technology for T cell targeted mRNA delivery. In: ASGCT Annual Meeting Abstract 1282 https://www.sciencedirect.com/journal/molecular-therapy/vol/33/issue/4/suppl/S1 (2025).

  170. Magee, M. S. et al. Deploying an RNA-based gene writer system and lipid nanoparticle (LNP) delivery to generate functional chimeric antigen receptor (CAR) T cells with in vitro and in vivo anti-tumor activity. Blood 142, 2073 (2023).

    Article  Google Scholar 

  171. Rodriguez, J. et al. Targeted lipid nanoparticle delivery of an RNA gene writer in vivo enables generation of CAR-T cells in a humanized mouse model. Blood 144, 4799 (2024).

    Article  Google Scholar 

  172. De Iaco, A. et al. In: ASGCT Annual Meeting Abstract 153 https://www.sciencedirect.com/journal/molecular-therapy/vol/33/issue/4/suppl/S1 (2025).

  173. Monte, M. et al. In vivo RNA delivery to T cells and hematopoietic stem cells in humanized mice and non-human primates using targeting lipid nanoparticles. In: ASGCT Annual Meeting Abstract 668 https://www.sciencedirect.com/journal/molecular-therapy/vol/33/issue/4/suppl/S1 (2025).

  174. Wang, Z. et al. Efficient and effective in vivo CAR T generation using recombinase-based, precise, high-capacity, off-the-shelf, LNP-based in vivo genomic engineering. In: ASGCT Annual Meeting Abstract 797 https://www.sciencedirect.com/journal/molecular-therapy/vol/33/issue/4/suppl/S1 (2025).

  175. Petazzi, P. et al. Precise gene writing system for CAR-T cell therapy generation. In: ASGCT Annual Meeting Abstract 1140 https://www.sciencedirect.com/journal/molecular-therapy/vol/33/issue/4/suppl/S1 (2025).

  176. Bimbo, J. et al. Novel non-viral DNA-based gene therapy vector for CAR T engineering in vivo. In: ASGCT Annual Meeting Abstract 213 https://www.sciencedirect.com/journal/molecular-therapy/vol/33/issue/4/suppl/S1 (2025).

  177. Urits, I. et al. A review of patisiran (ONPATTRO®) for the treatment of polyneuropathy in people with hereditary transthyretin amyloidosis. Neurol. Ther. 9, 301–315 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  178. Patil, H. et al. CAR-T cell therapy in rheumatic diseases: a review article. Clin Rheumatol. https://doi.org/10.1007/s10067-025-07451-7 (2025).

  179. Mueller, F. et al. Comparison of the safety profiles of CD19-targeting CAR T-cell therapy in patients with SLE and B-cell lymphoma. Blood https://doi.org/10.1182/blood.2025028375 (2025).

  180. Pecher, A. C. et al. CD19-targeting CAR T cell therapy in five patients with systemic sclerosis unsuitable for autologous stem cell transplantation. EULAR Rheumatologyhttps://doi.org/10.1016/j.ero.2025.03.005 (2025).

  181. Hagen, M. et al. Local immune effector cell-associated toxicity syndrome in CAR T-cell treated patients with autoimmune disease: an observational study. Lancet Rheumatol. https://doi.org/10.1016/S2665-9913(25)00091-8 (2025).

  182. Gust, J. BCMA-CAR T-cell treatment-associated parkinsonism. Blood 142, 1181–1183 (2023).

    Article  PubMed  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  184. Hu, J. & Dunbar, C. E. T-cell lymphomas in recipients of CAR-T cells: assessing risks and causalities. Blood 144, 2473–2481 (2024).

    Article  PubMed  CAS  Google Scholar 

  185. Hamilton, M. P. et al. Risk of second tumors and T-cell lymphoma after CAR T-cell therapy. N. Engl. J. Med. 390, 2047–2060 (2024).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  186. Ruella, M. & June, C. H. CAR T-cell resistance to oncogenic transformation. Blood Cancer Discov. 5, 229–233 (2024).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  187. Fraietta, J. A. et al. Disruption of TET2 promotes the therapeutic efficacy of CD19-targeted T-cells. Nature 558, 307–312 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  188. US Department of Health and Human Services, Food and Drug Administration, Center for Biologics Evaluation and Research. Guidance for industry: long term follow-up after administration of human gene therapy products. FDA https://www.fda.gov/media/113768/download (2020).

  189. Szebeni, J., Simberg, D., González-Fernández, A., Barenholz, Y. & Dobrovolskaia, M. A. Roadmap and strategy for overcoming infusion reactions to nanomedicines. Nat. Nanotechnol. 13, 1100–1108 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  190. Uehata, T. & Takeuchi, O. RNA recognition and immunity—innate immune sensing and its posttranscriptional regulation mechanisms. Cells 9, 1701 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  191. Hosseini-Kharat, M., Bremmell, K. E. & Prestidge, C. A. Why do lipid nanoparticles target the liver? Understanding of biodistribution and liver-specific tropism. Mol. Ther. Methods Clin. Dev. https://doi.org/10.1016/j.omtm.2025.101436 (2025).

  192. Szebeni, J. et al. Applying lessons learned from nanomedicines to understand rare hypersensitivity reactions to mRNA-based SARS-CoV-2 vaccines. Nat. Nanotechnol. 17, 337–346 (2022).

    Article  PubMed  CAS  Google Scholar 

  193. FDA Center for Biologics Evaluation and Research (CBER) Office of Therapeutic Products (OTP). Guidance document: considerations for the development of chimeric antigen receptor (CAR) T cell products. FDA https://www.fda.gov/regulatory-information/search-fda-guidance-documents/considerations-development-chimeric-antigen-receptor-car-t-cell-products (2024).

  194. US Department of Health and Human Services, Food and Drug Administration, Center for Drug Evaluation and Research (CDER), Center for Biologics Evaluation and Research (CBER), Center for Devices and Radiological Health (CDRH), Office of Good Clinical Practice (OGCP), Office of Regulatory Affairs (ORA). Guidance for industry: oversight of clinical investigations—a risk-based approach to monitoring. FDA https://www.fda.gov/media/116754/download (2013).

  195. Rejeski, K. et al. Immune effector cell-associated haematotoxicity after CAR T-cell therapy: from mechanism to management. Lancet Haematol. 11, e459–e470 (2024).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  196. Neelapu, S. S. et al. Axicabtagene ciloleucel as first-line therapy in high-risk large B-cell lymphoma: the phase 2 ZUMA-12 trial. Nat. Med. 28, 735–742 (2022).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  197. Swan, D., Madduri, D. & Hocking, J. CAR-T cell therapy in multiple myeloma: current status and future challenges. Blood Cancer J. 14, 206 (2024).

    Article  PubMed  PubMed Central  Google Scholar 

  198. Costa, L. J. MRD accelerating myeloma drug development. Blood 144, 345–347 (2024).

    Article  PubMed  CAS  Google Scholar 

  199. Amor, C. et al. Senolytic CAR T cells reverse senescence-associated pathologies. Nature 583, 127–132 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  200. Fuchs, E. J. Transplantation tolerance: from theory to clinic. Immunol. Rev. 258, 64–79 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  201. Su, Y., Yuan, C. & Shi, M. Editorial: screening and verification of new targets for CAR-T immunotherapy in cancer. Front. Immunol. 14, 1189773 (2023).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  202. Lee, J. et al. Antigen-specific B cell depletion for precision therapy of mucosal pemphigus vulgaris. J. Clin. Invest. 130, 6317–6324 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  203. Reincke, S. M. et al. Chimeric autoantibody receptor T cells deplete NMDA receptor-specific B cells. Cell 186, 5084–5097.e18 (2023).

    Article  PubMed  CAS  Google Scholar 

  204. Yang, J. et al. Dual-targeted CAR T-cell immunotherapies for hematological malignancies: latest updates from the 2023 ASH annual meeting. Exp. Hematol. Oncol. 13, 25 (2024).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  205. Ramishetti, S. et al. Systemic gene silencing in primary T lymphocytes using targeted lipid nanoparticles. ACS Nano 9, 6706–6716 (2015). This article provides evidence for the first time of in vivo delivery to a T cell subset using targeted nanomedicine.

    Article  PubMed  CAS  Google Scholar 

  206. Inamdar, V. V., Hao, S., Stephan, S. B. & Stephan, M. T. Biomaterial-based scaffolds for direct in situ programming of tumor-infiltrating T lymphocytes. J. Control. Rel. 370, 310–317 (2024).

    Article  CAS  Google Scholar 

  207. Nyberg, W. A. et al. An evolved AAV variant enables efficient genetic engineering of murine T cells. Cell 186, 446–460.e19 (2023).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  208. Hale, M. et al. Homology-directed recombination for enhanced engineering of chimeric antigen receptor T cells. Mol. Ther. Methods Clin. Dev. 4, 192–203 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  209. Demircan, M. B. et al. T-cell specific in vivo gene delivery with DART-AAVs targeted to CD8. Mol. Ther. 32, 3470–3484 (2024).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  210. Theuerkauf, S. A. et al. AAV vectors displaying bispecific DARPins enable dual-control targeted gene delivery. Biomaterials 303, 122399 (2023).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  211. Chen, H. et al. In vivo CAR-T cell therapy using modified AAV capsids targeting human T cells. In: ASGCT Annual Meeting Abstract 1739 https://www.sciencedirect.com/journal/molecular-therapy/vol/33/issue/4/suppl/S1 (2025).

  212. Decker, C. et al. In vivo VLP-based engineering generates multi-lineage CAR-immune cells that mediate potent and durable anti-tumor activity. In: SITC Annual Meeting https://ensoma.com/wp-content/uploads/SITC-2024-HER2-poster-final.pdf (2024).

  213. Tessera Therapeutics to present new data across preclinical programs for its gene writing™ and delivery platforms at the American Ssociety of Gene and Cell Therapy 28th Annual Meeting [press release]. Tessera https://www.tesseratherapeutics.com/news/tessera-therapeutics-to-present-new-data-across-preclinical-programs-for-its-gene-writing-and-delivery-platforms-at-the-american-society-of-gene-and-cell-therapy-28th-annual-meeting (2025).

  214. Li, Y.-R., Zhu, Y., Halladay, T. & Yang, L. In vivo CAR engineering for immunotherapy. Nat. Rev. Immunol. https://doi.org/10.1038/s41577-025-01174-1 (2025).

    Article  PubMed  Google Scholar 

  215. Lu, L., Xie, M., Yang, B., Zhao, W. B. & Cao, J. Enhancing the safety of CAR-T cell therapy: synthetic genetic switch for spatiotemporal control. Sci. Adv. 10, eadj6251 (2024).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  216. Dannenfelser, R. et al. Discriminatory power of combinatorial antigen recognition in cancer T cell therapies. Cell Syst. 11, 215–228.e5 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  217. Parrett, B. J. et al. Reducing off-target expression of mRNA therapeutics and vaccines in the liver with microRNA binding sites. Mol. Ther. Methods Clin. Dev. 33, 1101402 (2025).

    Article  Google Scholar 

  218. Xue, L. et al. High-throughput barcoding of nanoparticles identifies cationic, degradable lipid-like materials for mRNA delivery to the lungs in female preclinical models. Nat. Commun. 15, 1884 (2024).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  219. Rhym, L. H. et al. Peptide-encoding mRNA barcodes for the high-throughput in vivo screening of libraries of lipid nanoparticles for mRNA delivery. Nat. Biomed. Eng. 7, 901–910 (2023).

    Article  PubMed  CAS  Google Scholar 

  220. Da Silva Sanchez, A. J. et al. Universal barcoding predicts in vivo ApoE-independent lipid nanoparticle delivery. Nano Lett. 22, 4822–4830 (2022).

    Article  PubMed  CAS  Google Scholar 

  221. Das, K. P. & Chandra, J. Nanoparticles and convergence of artificial intelligence for targeted drug delivery for cancer therapy: current progress and challenges. Front. Med. Technol. 4, 1067144 (2023).

    Article  PubMed  PubMed Central  Google Scholar 

  222. Zhou, J. et al. Lipid nanoparticles produce chimeric antigen receptor T cells with interleukin-6 knockdown in vivo. J. Controlled Rel. 350, 298–307 (2022).

    Article  CAS  Google Scholar 

  223. Kath, J. et al. PASTA: CRISPR knock-in paired with integrase for simple, precise, and efficient large transgene insertion in T cells. In: ASGCT Annual Meeting Abstract 649 https://www.sciencedirect.com/journal/molecular-therapy/vol/33/issue/4/suppl/S1 (2025).

  224. Amabile, A. et al. Bispecific antibody targeting of lipid nanoparticles. Preprint at bioRxiv https://doi.org/10.1101/2024.12.20.629467 (2024).

  225. Kim, M. et al. Development of in vivo CAR T cell therapy using nanoparticle and RNA-based gene delivery systems. In: ASGCT Annual Meeting Abstract 796 https://www.sciencedirect.com/journal/molecular-therapy/vol/33/issue/4/suppl/S1 (2025).

  226. Divita, G. et al. In vivo mRNA CAR-T cell engineering utilizing a novel CD3/CD5 peptide based targeted nanoparticles. In: ASGCT Annual Meeting Abstract 1283 https://www.sciencedirect.com/journal/molecular-therapy/vol/33/issue/4/suppl/S1 (2025).

  227. Lee, C. et al. In vivo CAR delivery as supercoiled DNA with lymphatic tropism polyasparagine nanoparticles. In: ASGCT Annual Meeting Abstract 699 https://www.sciencedirect.com/journal/molecular-therapy/vol/33/issue/4/suppl/S1 (2025).

  228. Lian, H. et al. Cytokine therapy combined with nanomaterials participates in cancer immunotherapy. Pharmaceutics 14, 2606 (2022).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  229. Huang, C. et al. Lipid nanoparticle delivery system for mRNA encoding B7H3-redirected bispecific antibody displays potent antitumor effects on malignant tumors. Adv. Sci. https://doi.org/10.1002/advs.202205532 (2022).

  230. Hamilton, A. G. et al. Ionizable lipid nanoparticles with integrated immune checkpoint inhibition for mRNA CAR T cell engineering. Adv. Healthc. Mater. 12, e2301515 (2023).

    Article  PubMed  Google Scholar 

  231. Danko, I. & Wolff, J. A. Direct gene transfer into muscle. Vaccine 12, 1499–1502 (1994).

    Article  PubMed  CAS  Google Scholar 

  232. University Of Pittsburgh Medical Center. University of Pittsburgh Medical Center performs first in vivo gene therapy for hemophilia A. Science Daily https://www.sciencedaily.com/releases/1999/06/990604081001.htm (1999).

  233. Kuwana, Y. et al. Expression of chimeric receptor composed of immunoglobulin-derived V regions and T-cell receptor-derived c regions. Biochem. Biophys. Res. Commun. 149, 960–968 (1987). This work is the first report of design and in vitro activity of a construct comparable with current CARs.

    Article  PubMed  CAS  Google Scholar 

  234. Gross, G., Waks, T. & Eshhar, Z. Expression of immunoglobulin-T-cell receptor chimeric molecules as functional receptors with antibody-type specificity. Proc. Natl Acad. Sci. USA 86, 10024–10028 (1989).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  235. Eshhar, Z., Waks, T., Gross, G. & Schindler, D. G. Specific activation and targeting of cytotoxic lymphocytes through chimeric single chains consisting of antibody-binding domains and the γ or ζ subunits of the immunoglobulin and T-cell receptors. Proc. Natl Acad. Sci. USA 90, 720–724 (1993).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  236. Finney, H. M., Lawson, A. D., Bebbington, C. R. & Weir, A. N. Chimeric receptors providing both primary and costimulatory signaling in T cells from a single gene product. J. Immunol. 161, 2791–2797 (1998).

    Article  PubMed  CAS  Google Scholar 

  237. Maher, J., Brentjens, R. J., Gunset, G., Rivière, I. & Sadelain, M. Human T-lymphocyte cytotoxicity and proliferation directed by a single chimeric TCRζ/CD28 receptor. Nat. Biotechnol. 20, 70–75 (2002).

    Article  PubMed  CAS  Google Scholar 

  238. Kochenderfer, J. N. Eradication of B-lineage cells and regression of lymphoma in a patient treated with autologous T cells genetically engineered to recognize CD19. Blood 116, 4099–4102 (2010). This article reports the first evidence of objective clinical response afforded by CAR-T cell intervention in a patient with B cell lymphoma.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  239. Porter, D. L., Levine, B. L., Kalos, M., Bagg, A. & June, C. J. Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N. Engl. J. Med. 365, 725–733 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  240. Umoja Biopharma announces FDA clearance of IND application for UB-VV111, a CD19 directed in situ CAR T for hematologic malignancies press release. Umoja Biopharma https://www.umoja-biopharma.com/news/umoja-biopharma-announces-fda-clearance-of-ind-application-for-ub-vv111-a-cd19-directed-in-situ-car-t-for-hematologic-malignancies/ (2024).

  241. Papahadjopoulos, D., Vail, W. J., Jacobson, K. & Poste, G. Cochleate lipid cylinders: formation by fusion of unilamellar lipid vesicles. Biochim. Biophys. Acta 394, 483–491 (1975).

    Article  PubMed  CAS  Google Scholar 

  242. Dimitriadis, G. J. Translation of rabbit globin mRNA introduced by liposomes into mouse lymphocytes. Nature 274, 923–924 (1978).

    Article  PubMed  CAS  Google Scholar 

  243. Malone, R. W., Felgner, P. L. & Verma, I. M. Cationic liposome-mediated RNA transfection. Proc. Natl Acad. Sci. USA 86, 6077–6081 (1989). This seminal report describes the use of mRNA to directly transfect human, rat, mouse, Xenopus and Drosophila cells.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  244. Conry, R. M. et al. Characterization of a messenger RNA polynucleotide vaccine vector. Cancer Res. 55, 1397–1400 (1995).

    PubMed  CAS  Google Scholar 

  245. Hafez, I. M., Ansell, S. & Cullis, P. R. Tunable pH-sensitive liposomes composed of mixtures of cationic and anionic lipids. Biophysical J. 79, 1438–1446 (2000).

    Article  CAS  Google Scholar 

  246. Karikó, K., Buckstein, M., Ni, H. & Weissman, D. Suppression of RNA recognition by Toll-like receptors: the impact of nucleoside modification and the evolutionary origin of RNA. Immunity 23, 165–175 (2005).

    Article  PubMed  Google Scholar 

  247. Barrett, D. M. et al. Treatment of advanced leukemia in mice with mRNA engineered T cells. Hum. Gene Ther. 22, 1575–1586 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  248. US Food and Drug Administration. COMIRNATY FDA approval. FDA https://www.fda.gov/media/151710/download?attachment (2021).

  249. US Food and Drug Administration. SPIKEVAX FDA approval. FDA https://www.fda.gov/media/155815/download?attachment (2022).

  250. Uslu, U. & June, C. H. Beyond the blood: expanding CAR T cell therapy to solid tumors. Nat. Biotechnol. https://doi.org/10.1038/s41587-024-02446-2 (2024).

  251. Aghajanian, H., Rurik, J. G. & Epstein, J. A. CAR-based therapies: opportunities for immuno-medicine beyond cancer. Nat. Metab. 4, 163–169 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  252. Wat, J. & Barmettler, S. Hypogammaglobulinemia after chimeric antigen receptor (CAR) T-cell therapy: characteristics, management, and future directions. J. Allergy Clin. Immunol. Pract. 10, 460–466 (2022).

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgements

The authors acknowledge the contribution of P. Johnson and D. Fontana to the section ‘Viral vectors’. The authors apologize to those authors whose work was not cited directly owing to space limitations.

Author information

Authors and Affiliations

Authors

Contributions

J.I.A., A.B., A.S., K.F., L.G., R.H., M.K., F.N., J.V.S., A.J.S. and K.T. generated the sections ‘Viral vectors’ and ‘RNA-based platforms’. A.B. conceived the article layout. M.T.S., C.J.B., D.W. and C.H.J. contributed to the Abstract, Introduction and Outlook. All authors reviewed and edited the manuscript before submission. C.J.B.’s contribution to the manuscript represents his own perspective and not the official view of the Paul-Erlich Institute.

Corresponding author

Correspondence to Adrian Bot.

Ethics declarations

Competing interests

J.I.A. is a paid employee of Interius BioTherapeutics, Inc., holds equity in the company, and is an inventor on patents issued and/or pending related to this work. A.B. is a shareholder and employee of Capstan Therapeutics, a company developing in vivo chimeric antigen receptor (CAR) therapies, and inventor or co-inventor on multiple relevant patents in the CAR and related fields. C.J.B. is a co-inventor on patents covering T cell-targeted lentiviral vectors. K.F. is a shareholder and member of the Board of Directors of Kelonia Therapeutics, and is also an inventor on patent applications assigned to Kelonia. L.G. is a shareholder and employee of Moderna, and a shareholder of Tessera. A.S. is a shareholder and employee of Umoja Therapeutics, a company developing in vivo CAR therapies. R.H. is a shareholder and employee of Myeloid Therapeutics, a company developing in vivo CAR therapies. C.H.J. is an inventor of multiple CAR patents and shareholder of several companies developing CAR products. M.K. is a founder, employee and shareholder of Carisma Therapeutics; a founder, shareholder and board director of Chymal Therapeutics; and an inventor of multiple patents related to CAR macrophages and monocytes (CAR-M) that have been licensed by Carisma Therapeutics. F.N. is a shareholder and employee of Orna Therapeutics, a company developing in vivo CAR therapies. A.J.S. is a shareholder and current employee of Sanofi. J.V.S. is an employee and shareholder in Mirai Bio, and a consultant and shareholder in Sana Biotechnology. M.T.S. is co-founder of and has received stock options from Persistence Therapeutics (Jupiter Bioventures); and has IP Licensing with Sanofi, Juno Therapeutics (now Bristol Myers Squibb) and Jupiter Bioventures. K.T. is a shareholder and employee of Sana Therapeutics, a company developing in vivo CAR and cell gene therapies. D.W. has filed patent applications based on some aspects of this work; those interests were fully disclosed to the University of Pennsylvania, with an approved plan in place for managing any potential conflicts arising from licensing of these patents.

Peer review

Peer review information

Nature Reviews Drug Discovery thanks Melgious Ang 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.

Glossary

Circular RNA

A single-stranded RNA molecule that forms a continuous loop. Although naturally a type of non-coding RNA that is found in many species, circular RNA can be adapted for cell engineering.

Designed ankyrin repeat protein

(DARPin). A genetically engineered antibody mimetic protein typically exhibiting highly specific and high-affinity target protein binding, derived from natural ankyrin repeat proteins.

Fusogen

A protein on the surface of a virus that helps the virus enter a host cell by fusing the viral membrane with the host cell membrane. This process releases the genetic material of the virus into the host cell’s cytoplasm.

Hypogammaglobulinaemia

A condition characterized by abnormally low levels of immunoglobulins in the blood, making individuals more susceptible to infections.

Internal ribosomal entry site

A specific RNA sequence that allows ribosomes to bind to mRNA and initiate protein synthesis at an internal location, bypassing the typical cap-dependent initiation process.

Ionizable lipids

Lipid molecules that can change their charge depending on the pH level of their environment. They are a key component of lipid nanoparticles (LNPs), which are used to deliver RNA therapeutics.

Lymphodepletion conditioning

A treatment that prepares the body for chimeric antigen receptor (CAR)-T cell therapy or adoptive cell transfer in general. It involves using chemotherapy drugs (most frequently cyclophosphamide and fludarabine) to deplete endogenous immune cells.

Minimal residual disease

A term used for a small number of cancer cells that remain in the body after treatment. Minimal residual disease can occur in blood cancers such as leukaemia and lymphoma, as well as solid tumours.

N 1-Methylpseudouridine

A chemical compound found in tRNA and mRNA vaccines. Although a natural component of archaea, it is presently utilized in biochemistry and molecular biology.

Nipah virus (NiV) glycoproteins

NiV uses two key glycoproteins on its surface: the attachment glycoprotein (G) and the fusion glycoprotein (F), which are crucial for viral entry into host cells. The G protein facilitates attachment to the host cell, whereas the F protein triggers membrane fusion, allowing the virus to enter the cell.

PiggyBac transposon system

A genetic engineering tool used to introduce and integrate DNA sequences into a genome, often in a stable and reproducible manner. It utilizes a transposase enzyme to ‘cut and paste’ a transposon (a DNA sequence) into a new location in the genome.

Pseudotype

A virus particle that has been engineered to display a foreign viral envelope protein on its surface.

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

Bot, A., Scharenberg, A., Friedman, K. et al. In vivo chimeric antigen receptor (CAR)-T cell therapy. Nat Rev Drug Discov (2025). https://doi.org/10.1038/s41573-025-01291-5

Download citation

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41573-025-01291-5

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