Abstract
The clinical potential of current chimeric antigen receptor-engineered T (CAR-T) cell therapy is hampered by its autologous nature that poses considerable challenges in manufacturing, costs and patient selection. This spurs demand for off-the-shelf therapies. Here we introduce an ex vivo feeder-free culture method to differentiate gene-engineered hematopoietic stem and progenitor (HSP) cells into allogeneic invariant natural killer T (AlloNKT) cells and their CAR-armed derivatives (AlloCAR-NKT cells). We include detailed information on lentivirus generation and titration, as well as the five stages of ex vivo culture required to generate AlloCAR-NKT cells, including HSP cell engineering, HSP cell expansion, NKT cell differentiation, NKT cell deep differentiation and NKT cell expansion. In addition, we describe procedures for evaluating the pharmacology, antitumor efficacy and mechanism of action of AlloCAR-NKT cells. It takes ~2 weeks to generate and titrate lentiviruses and ~6 weeks to generate mature AlloCAR-NKT cells. Competence with human stem cell and T cell culture, gene engineering and flow cytometry is required for optimal results.
Key points
-
This protocol describes an ex vivo feeder-free culture method to differentiate genetically engineered hematopoietic stem and progenitor cells into allogeneic CAR-NKT cells for off-the-shelf cancer immunotherapy. In addition, procedures are provided for evaluating their pharmacology, antitumor efficacy and mechanism of action.
-
This approach overcomes challenges in manufacturing, costs and patient selection that hamper the clinical potential of current autologous CAR-T cell therapies.
This is a preview of subscription content, access via your institution
Access options
Access Nature and 54 other Nature Portfolio journals
Get Nature+, our best-value online-access subscription
$29.99 / 30 days
cancel any time
Subscribe to this journal
Receive 12 print issues and online access
$259.00 per year
only $21.58 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to full article PDF
Prices may be subject to local taxes which are calculated during checkout
Similar content being viewed by others
Data availability
All associated data are presented in the protocol paper or Supplementary Information. The genomics data were reanalyzed from the public repository Gene Expression Omnibus database: GSE241996 (scRNA-seq, related to Fig. 5c–e). Additional information and materials will be made available upon reasonable request. Source data are provided with this paper.
References
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).
Sterner, R. C. & Sterner, R. M. CAR-T cell therapy: current limitations and potential strategies. Blood Cancer J. 11, 69 (2021).
Labanieh, L. & Mackall, C. L. CAR immune cells: design principles, resistance and the next generation. Nature 614, 635–648 (2023).
Finck, A. V., Blanchard, T., Roselle, C. P., Golinelli, G. & June, C. H. Engineered cellular immunotherapies in cancer and beyond. Nat. Med. 28, 678–689 (2022).
Benjamin, R. et al. Genome-edited, donor-derived allogeneic anti-CD19 chimeric antigen receptor T cells in paediatric and adult B cell acute lymphoblastic leukaemia: results of two phase 1 studies. Lancet 396, 1885–1894 (2020).
van der Stegen, S. J. C. et al. Generation of T cell-receptor-negative CD8αβ-positive CAR T cells from T cell-derived induced pluripotent stem cells. Nat. Biomed. Eng. 6, 1284–1297 (2022).
Liu, E. et al. Use of CAR-transduced natural killer cells in CD19-positive lymphoid tumors. N. Engl. J. Med. 382, 545–553 (2020).
Heczey, A. et al. Anti-GD2 CAR-NKT cells in patients with relapsed or refractory neuroblastoma: an interim analysis. Nat. Med. 26, 1686–1690 (2020).
Klichinsky, M. et al. Human chimeric antigen receptor macrophages for cancer immunotherapy. Nat. Biotechnol. 38, 947–953 (2020).
Godfrey, D. I., Le Nours, J., Andrews, D. M., Uldrich, A. P. & Rossjohn, J. Unconventional T cell targets for cancer immunotherapy. Immunity 48, 453–473 (2018).
Chandra, S. & Kronenberg, M. Activation and function of iNKT and MAIT cells. Adv. Immunol. 127, 145–201 (2015).
Chaidos, A. et al. Graft invariant natural killer T cell dose predicts risk of acute graft-versus-host disease in allogeneic hematopoietic stem cell transplantation. Blood 119, 5030–5036 (2012).
Exley, M. A. et al. Adoptive transfer of invariant NKT cells as immunotherapy for advanced melanoma: a phase I clinical trial. Clin. Cancer Res. 23, 3510–3519 (2017).
Bae, E. A. et al. Activation of NKT cells in an anti-PD-1-resistant tumor model enhances antitumor immunity by reinvigorating exhausted CD8 T cells. Cancer Res. 78, 5315–5326 (2018).
Varghese, B. et al. Invariant NKT cell-augmented GM-CSF-secreting tumor vaccine is effective in advanced prostate cancer model. Cancer Immunol. Immunother. 71, 2943–2955 (2022).
Lynch, L. et al. Adipose tissue invariant NKT cells protect against diet-induced obesity and metabolic disorder through regulatory cytokine production. Immunity 37, 574–587 (2012).
Brennan, P. J., Brigl, M. & Brenner, M. B. Invariant natural killer T cells: an innate activation scheme linked to diverse effector functions. Nat. Rev. Immunol. 13, 101–117 (2013).
Li, Y.-R. et al. Profiling ovarian cancer tumor and microenvironment during disease progression for cell-based immunotherapy design. iScience 26, 107952 (2023).
Li, Y.-R. et al. Development of allogeneic HSC-engineered iNKT cells for off-the-shelf cancer immunotherapy. Cell Rep. Med. 2, 100449 (2021).
Rotolo, A. et al. Enhanced anti-lymphoma activity of CAR19-iNKT cells underpinned by dual CD19 and CD1d targeting. Cancer Cell 34, 596–610.e11 (2018).
Landoni, E. et al. IL-12 reprograms CAR-expressing natural killer T cells to long-lived Th1-polarized cells with potent antitumor activity. Nat. Commun. 15, 89 (2024).
Xu, X. et al. NKT cells coexpressing a GD2-specific chimeric antigen receptor and IL15 show enhanced in vivo persistence and antitumor activity against neuroblastoma. Clin. Cancer Res. 25, 7126–7138 (2019).
Liu, Y. et al. IL-21-armored B7H3 CAR-iNKT cells exert potent antitumor effects. iScience 27, 108597 (2024).
Heczey, A. et al. Anti-GD2 CAR-NKT cells in relapsed or refractory neuroblastoma: updated phase 1 trial interim results. Nat. Med. 29, 1379–1388 (2023).
Li, Y.-R. et al. Generation of allogeneic CAR-NKT cells from hematopoietic stem and progenitor cells using a clinically guided culture method. Nat. Biotechnol. https://doi.org/10.1038/s41587-024-02226-y (2024).
Li, Y.-R. et al. Engineering allorejection-resistant CAR-NKT cells from hematopoietic stem cells for off-the-shelf cancer immunotherapy. Mol. Ther. 32, 1849–1874 (2024).
Labanieh, L., Majzner, R. G. & Mackall, C. L. Programming CAR-T cells to kill cancer. Nat. Biomed. Eng. 2, 377–391 (2018).
Park, J. H., Geyer, M. B. & Brentjens, R. J. CD19-targeted CAR T-cell therapeutics for hematologic malignancies: interpreting clinical outcomes to date. Blood 127, 3312–3320 (2016).
Teoh, P. J. & Chng, W. J. CAR T-cell therapy in multiple myeloma: more room for improvement. Blood Cancer J. 11, 84 (2021).
Majzner, R. G. et al. GD2-CAR T cell therapy for H3K27M-mutated diffuse midline gliomas. Nature 603, 934–941 (2022).
Jiang, Z. et al. Anti-GPC3-CAR T cells suppress the growth of tumor cells in patient-derived xenografts of hepatocellular carcinoma. Front. Immunol. 7, 690 (2016).
Sarah, C. CAR T cells in glioblastoma. Nat. Rev. Drug Discov. 16, 602 (2017).
Schoutrop, E. et al. Mesothelin-specific CAR T cells target ovarian cancer. Cancer Res. 81, 3022–3035 (2021).
Adusumilli, P. S. et al. A phase I trial of regional mesothelin-targeted CAR T cell therapy in patients with malignant pleural disease, in combination with the anti-PD-1 agent pembrolizumab. Cancer Discov. 11, 2748–2763 (2021).
Tchou, J. et al. Safety and efficacy of intratumoral injections of chimeric antigen receptor (CAR) T cells in metastatic breast cancer. Cancer Immunol. Res. 5, 1152–1161 (2017).
Ko, A. H. et al. Dual targeting of mesothelin and CD19 with chimeric antigen receptor-modified T cells in patients with metastatic pancreatic cancer. Mol. Ther. 28, 2367–2378 (2020).
Hegde, M. et al. Autologous HER2-specific CAR T cells after lymphodepletion for advanced sarcoma: a phase 1 trial. Nat. Cancer https://doi.org/10.1038/s43018-024-00749-6 (2024).
Vitanza, N. A. et al. Locoregional infusion of HER2-specific CAR T cells in children and young adults with recurrent or refractory CNS tumors: an interim analysis. Nat. Med. 27, 1544–1552 (2021).
Ahmed, N. et al. HER2-specific chimeric antigen receptor-modified virus-specific t cells for progressive glioblastoma: a phase 1 dose-escalation trial. JAMA Oncol. 3, 1094–1101 (2017).
Bagley, S. J. et al. Intrathecal bivalent CAR T cells targeting EGFR and IL13Rα2 in recurrent glioblastoma: phase 1 trial interim results. Nat. Med. https://doi.org/10.1038/s41591-024-02893-z (2024).
Narayan, V. et al. PSMA-targeting TGFβ-insensitive armored CAR T cells in metastatic castration-resistant prostate cancer: a phase 1 trial. Nat. Med. 28, 724–734 (2022).
Sauer, T. et al. CD70-specific CAR T cells have potent activity against acute myeloid leukemia without HSC toxicity. Blood 138, 318–330 (2021).
Schett, G., Mackensen, A. & Mougiakakos, D. CAR T-cell therapy in autoimmune diseases. Lancet 402, 2034–2044 (2023).
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).
Mackensen, A. et al. Anti-CD19 CAR T cell therapy for refractory systemic lupus erythematosus. Nat. Med. 28, 2124–2132 (2022).
Müller, F. et al. CD19-targeted CAR T cells in refractory antisynthetase syndrome. Lancet 401, 815–818 (2023).
Fischbach, F. et al. CD19-targeted chimeric antigen receptor T cell therapy in two patients with multiple sclerosis. Med https://doi.org/10.1016/j.medj.2024.03.002 (2024).
Gupta, S. et al. CAR-T cell-mediated B cell depletion in central nervous system autoimmunity. Neurol. Neuroimmunol. Neuroinflamm. 10, e200080 (2023).
Zhang, B. et al. In vitro elimination of autoreactive B cells from rheumatoid arthritis patients by universal chimeric antigen receptor T cells. Ann. Rheum. Dis. 80, 176–184 (2021).
Li, Y. R. et al. Development of off-the-shelf hematopoietic stem cell-engineered invariant natural killer T cells for COVID-19 therapeutic intervention. Stem Cell Res. Ther. 13, 1–15 (2022).
Li, Y.-R. et al. Off-the-shelf third-party HSC-engineered iNKT cells for ameliorating GvHD while preserving GvL effect in the treatment of blood cancers. iScience 25, 104859 (2022).
Larson, R. C. & Maus, M. V. Recent advances and discoveries in the mechanisms and functions of CAR T cells. Nat. Rev. Cancer 21, 145–161 (2021).
Mullard, A. FDA approves fourth CAR-T cell therapy. Nat. Rev. Drug Discov. 20, 166 (2021).
Rafiq, S., Hackett, C. S. & Brentjens, R. J. Engineering strategies to overcome the current roadblocks in CAR T cell therapy. Nat. Rev. Clin. Oncol. 17, 147–167 (2020).
Neelapu, S. S. et al. Chimeric antigen receptor T cell therapy—assessment and management of toxicities. Nat. Rev. Clin. Oncol. 15, 47–62 (2018).
Roddie, C., O’Reilly, M., Dias Alves Pinto, J., Vispute, K. & Lowdell, M. Manufacturing chimeric antigen receptor T cells: issues and challenges. Cytotherapy 21, 327–340 (2019).
Caldwell, K. J., Gottschalk, S. & Talleur, A. C. Allogeneic CAR cell therapy-more than a pipe dream. Front. Immunol. 11, 618427 (2020).
Depil, S., Duchateau, P., Grupp, S. A., Mufti, G. & Poirot, L. Off-the-shelf’ allogeneic CAR T cells: development and challenges. Nat. Rev. Drug Discov. 19, 185–199 (2020).
Eyquem, J. et al. Targeting a CAR to the TRAC locus with CRISPR–Cas9 enhances tumour rejection. Nature 543, 113–117 (2017).
Georgiadis, C. et al. Long terminal repeat CRISPR–CAR-coupled ‘universal’ T cells mediate potent anti-leukemic effects. Mol. Ther. 26, 1215–1227 (2018).
Cichocki, F. et al. Off-the-shelf, multiplexed-engineered iPSC-derived NK cells mediate potent multi-antigen targeting of B cell malignancies with reduced cytotoxicity against healthy B cells. Blood 138, 407 (2021).
Cichocki, F. et al. iPSC-derived NK cells maintain high cytotoxicity and enhance in vivo tumor control in concert with T cells and anti-PD-1 therapy. Sci. Transl. Med. 12, eaaz5618 (2020).
Li, Y.-R. et al. Advancing cell-based cancer immunotherapy through stem cell engineering. Cell Stem Cell https://doi.org/10.1016/j.stem.2023.02.009 (2023).
Torikai, H. et al. A foundation for universal T cell based immunotherapy: T cells engineered to express a CD19-specific chimeric-antigen-receptor and eliminate expression of endogenous TCR. Blood 119, 5697–5705 (2012).
Wang, D., Quan, Y., Yan, Q., Morales, J. E. & Wetsel, R. A. Targeted disruption of the β2-microglobulin gene minimizes the immunogenicity of human embryonic stem cells. Stem Cells Transl. Med. 4, 1234–1245 (2015).
Kagoya, Y. et al. Genetic ablation of HLA Class I, Class II, and the T-cell receptor enables allogeneic T cells to be used for adoptive T-cell therapy. Cancer Immunol. Res. 8, 926–936 (2020).
Stenger, D. et al. Endogenous TCR promotes in vivo persistence of CD19-CAR-T cells compared to a CRISPR–Cas9-mediated TCR knockout CAR. Blood 136, 1407–1418 (2020).
Li, Y.-R., Wilson, M. & Yang, L. Target tumor microenvironment by innate T cells. Front. Immunol. 13, 999549 (2022).
Li, Y.-R., Dunn, Z. S., Zhou, Y., Lee, D. & Yang, L. Development of stem cell-derived immune cells for off-the-shelf cancer immunotherapies. Cells 10, 3497 (2021).
Fang, Y. et al. Graft-versus-host disease modulation by innate T cells. Int. J. Mol. Sci. 24, 4084 (2023).
Qu, G. et al. Comparing mouse and human tissue-resident γδ T cells. Front. Immunol. 13, 891687 (2022).
Bendelac, A., Savage, P. B. & Teyton, L. The biology of NKT cells. Annu. Rev. Immunol. 25, 297–336 (2007).
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.e5 (2018).
Zeng, J., Tang, S. Y., Toh, L. L. & Wang, S. Generation of ‘off-the-shelf’ natural killer cells from peripheral blood cell-derived induced pluripotent stem cells. Stem Cell Rep. 9, 1796–1812 (2017).
Hermanson, D. L. et al. Induced pluripotent stem cell-derived natural killer cells for treatment of ovarian cancer. Stem Cells 34, 93–101 (2016).
Zhou, Y. et al. Engineering induced pluripotent stem cells for cancer immunotherapy. Cancers 14, 2266 (2022).
Frank, C. et al. iPSC-derived NK cells maintain high cytotoxicity and enhance in vivo tumor control in concert with T cells and anti–PD-1 therapy. Sci. Transl. Med. 12, eaaz5618 (2020).
Perez, C., Gruber, I. & Arber, C. Off-the-shelf allogeneic T cell therapies for cancer: opportunities and challenges using naturally occurring ‘universal’ donor T cells. Front. Immunol. 11, 583716 (2020).
Jo, S. et al. Endowing universal CAR T-cell with immune-evasive properties using TALEN-gene editing. Nat. Commun. 13, 3453 (2022).
Chen, Y. et al. Genetic engineering strategies to enhance antitumor reactivity and reduce alloreactivity for allogeneic cell-based cancer therapy. Front. Med. 10, 1135468 (2023).
Gornalusse, G. G. et al. HLA-E-expressing pluripotent stem cells escape allogeneic responses and lysis by NK cells. Nat. Biotechnol. 35, 765–772 (2017).
Hu, X. et al. Abstract LB144: overexpression of CD47 protects hypoimmune CAR T cells from innate immune cell killing. Cancer Res. 81, LB144–LB144 (2021).
Iriguchi, S. et al. A clinically applicable and scalable method to regenerate T-cells from iPSCs for off-the-shelf T-cell immunotherapy. Nat. Commun. 12, 430 (2021).
Montel-Hagen, A. et al. Organoid-induced differentiation of conventional T cells from human pluripotent stem cells. Cell Stem Cell 24, 376–389.e8 (2019).
Wang, D. et al. Glioblastoma-targeted CD4+ CAR T cells mediate superior antitumor activity. JCI Insight 3, e99048 (2018).
Yang, Y. et al. TCR engagement negatively affects CD8 but not CD4 CAR T cell expansion and leukemic clearance. Sci. Transl. Med. 9, eaag1209 (2017).
Sommermeyer, D. et al. Chimeric antigen receptor-modified T cells derived from defined CD8+ and CD4+ subsets confer superior antitumor reactivity in vivo. Leukemia 30, 492–500 (2016).
Tang, N. et al. TGF-β inhibition via CRISPR promotes the long-term efficacy of CAR T cells against solid tumors. JCI Insight 5, e133977 (2020).
Rupp, L. J. et al. CRISPR–Cas9-mediated PD-1 disruption enhances anti-tumor efficacy of human chimeric antigen receptor T cells. Sci. Rep. 7, 737 (2017).
Giuffrida, L. et al. CRISPR–Cas9 mediated deletion of the adenosine A2A receptor enhances CAR T cell efficacy. Nat. Commun. 12, 3236 (2021).
Pavel-Dinu, M. et al. Gene correction for SCID-X1 in long-term hematopoietic stem cells. Nat. Commun. 10, 1634 (2019).
Humbert, O. et al. Therapeutically relevant engraftment of a CRISPR–Cas9-edited HSC-enriched population with HbF reactivation in nonhuman primates. Sci. Transl. Med. 11, eaaw3768 (2019).
Charlesworth, C. T. et al. Priming human repopulating hematopoietic stem and progenitor cells for Cas9/sgRNA gene targeting. Mol. Ther. Nucleic Acids 12, 89–104 (2018).
Li, Y.-R., Zhou, Y., Kramer, A. & Yang, L. Engineering stem cells for cancer immunotherapy. Trends Cancer 7, 1059–1073 (2021).
Zhu, Y. et al. Development of hematopoietic stem cell-engineered invariant natural killer T cell therapy for cancer. Cell Stem Cell 25, 542–557.e9 (2019).
Seet, C. S. et al. Generation of mature T cells from human hematopoietic stem and progenitor cells in artificial thymic organoids. Nat. Methods 14, 521–530 (2017).
Acknowledgements
We thank the UCLA Technology Centre for Genomics and Bioinformatics facility for providing RNA-seq services, the UCLA Center for AIDS Research (CFAR) Virology Core for providing human cells and the UCLA Broad Stem Cell Research Center (BSCRC) Flow Cytometry Core Facility for cell sorting support. This work was supported by a Partnering Opportunity for Discovery Stage Research Projects Award and a Partnering Opportunity for Translational Research Projects Awards from the California Institute for Regenerative Medicine (grant nos. DISC2-11157 and TRAN1-12250, to L.Y.), a Department of Defense Congressionally Directed Medical Research Program (CDMRP) Peer Reviewed Cancer Research Program (PRCRP) Impact Award (grant no. CA200456 to L.Y.), a UCLA BSCRC Innovation Award (to L.Y.) and an Ablon Scholars Award (to L.Y.). Y.-R.L. is a postdoctoral fellow supported by a UCLA MIMG M. John Pickett Post-Doctoral Fellow Award, a California Institute for Regenerative Medicine (CIRM)-BSCRC Postdoctoral Fellowship and a UCLA Sydney Finegold Postdoctoral Award. K.Z. is a predoctoral fellow supported by a CIRM-BSCRC Predoctoral Fellowship. D.L. is a postdoctoral fellow supported by T32 Tumor Immunology Training Grant postdoctoral fellowships (USHHS Ruth L. Kirschstein Institutional National Research Service Award, T32-CA009120). Figures were created with BioRender.com.
Author information
Authors and Affiliations
Contributions
Y.-R.L., K.Z., D.L. and L.Y. designed the experiments, analyzed the data and wrote the manuscript. L.Y. conceived and oversaw the study, with assistance from Y.-R.L., K.Z. and D.L. Y.-R.L., K.Z. and D.L. performed all experiments, with assistance from Y. Zhu, T.H., J.Y., Y. Zhou, Y.F., Z.L., Y.C. and S.S.
Corresponding author
Ethics declarations
Competing interests
Y.-R.L., D.L., J.Y., Y. Zhou and L.Y. are inventors on patents relating to this study filed by UCLA. J.Y. is currently an employee of Appia Bio. Y. Zhou is currently an employee of Amberstone Biosciences. L.Y. is a scientific advisor to AlzChem and Amberstone Biosciences, and a cofounder, stockholder and advisory board member of Appia Bio. Appia Bio licensed some patents relating to this study from UCLA. None of the declared companies contributed to or directed any of the research reported in this article. The other authors declare no competing interests.
Peer review
Peer review information
Nature Protocols thanks Haopeng Wang 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.
Key references
Li, Y.-R. et al. Nat. Biotechnol. (2024): https://doi.org/10.1038/s41587-024-02226-y
Li, Y.-R. et al. Mol. Ther. 32, 1849–1874 (2024): https://doi.org/10.1016/j.ymthe.2024.04.005
Supplementary information
Supplementary Information
Supplementary Fig. 1.
Supplementary Data 1
Source Data for Supplementary Fig. 1.
Source data
Source Data Figs. 4 and 6
Statistical 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.
About this article
Cite this article
Li, YR., Zhou, K., Lee, D. et al. Generating allogeneic CAR-NKT cells for off-the-shelf cancer immunotherapy with genetically engineered HSP cells and feeder-free differentiation culture. Nat Protoc (2025). https://doi.org/10.1038/s41596-024-01077-w
Received:
Accepted:
Published:
DOI: https://doi.org/10.1038/s41596-024-01077-w