Advancing cell-based cancer immunotherapy through stem cell engineering

Cell Stem Cell - Tập 30 - Trang 592-610 - 2023
Yan-Ruide Li1, Zachary Spencer Dunn1,2, Yanqi Yu1, Miao Li1, Pin Wang2,3, Lili Yang1,4,5,6
1Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
2Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA 90089, USA
3Department of Pharmacology and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
4Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
5Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
6Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, 90095, USA

Tài liệu tham khảo

Wilson, 2009, Medicine. A history lesson for stem cells, Science, 324, 727, 10.1126/science.1174935 Singh, 2016, Allogeneic stem cell transplantation: A historical and scientific overview, Cancer Res., 76, 6445, 10.1158/0008-5472.CAN-16-1311 Dunn, 2004, The immunobiology of cancer immunosurveillance and immunoediting, Immunity, 21, 137, 10.1016/j.immuni.2004.07.017 Rosenberg, 2011, Durable complete responses in heavily pretreated patients with metastatic melanoma using T-cell transfer immunotherapy, Clin. Cancer Res., 17, 4550, 10.1158/1078-0432.CCR-11-0116 Radvanyi, 2015, Tumor-infiltrating lymphocyte therapy: addressing prevailing questions, Cancer J., 21, 450, 10.1097/PPO.0000000000000162 June, 2007, Adoptive T cell therapy for cancer in the clinic, J. Clin. Invest., 117, 1466, 10.1172/JCI32446 Kansagra, 2020, Expanding access to chimeric antigen receptor T-cell therapies: challenges and opportunities, Am. Soc. Clin. Oncol. Educ. B., 40, e27, 10.1200/EDBK_279151 Mirzaei, 2017, Chimeric antigen receptors T cell therapy in solid tumor: challenges and clinical applications, Front. Immunol., 8, 1850, 10.3389/fimmu.2017.01850 Roddie, 2019, Manufacturing chimeric antigen receptor T cells: issues and challenges, Cytotherapy, 21, 327, 10.1016/j.jcyt.2018.11.009 Srivastava, 2018, Chimeric antigen receptor T cell therapy: challenges to bench-to-bedside efficacy, J. Immunol., 200, 459, 10.4049/jimmunol.1701155 Xia, 2017, Chimeric-antigen receptor T (CAR-T) cell therapy for solid tumors: challenges and opportunities, Oncotarget, 8, 90521, 10.18632/oncotarget.19361 Lim, 2017, The principles of engineering immune cells to treat cancer, Cell, 168, 724, 10.1016/j.cell.2017.01.016 Mikkilineni, 2017, Chimeric antigen receptor T-cell therapies for multiple myeloma, Blood, 130, 2594, 10.1182/blood-2017-06-793869 Ramos, 2016, CAR-T cell therapy for lymphoma, Annu. Rev. Med., 67, 165, 10.1146/annurev-med-051914-021702 Themeli, 2015, New cell sources for T cell engineering and adoptive immunotherapy, Cell Stem Cell, 16, 357, 10.1016/j.stem.2015.03.011 Ruella, 2018, Induction of resistance to chimeric antigen receptor T cell therapy by transduction of a single leukemic B cell, Nat. Med., 24, 1499, 10.1038/s41591-018-0201-9 Good, 2021, An NK-like CAR T cell transition in CAR T cell dysfunction, Cell, 184, 6081, 10.1016/j.cell.2021.11.016 Deng, 2020, Characteristics of anti-CD19 CAR T cell infusion products associated with efficacy and toxicity in patients with large B cell lymphomas, Nat. Med., 26, 1878, 10.1038/s41591-020-1061-7 Arcangeli, 2022, CAR T-cell manufacturing from naive/stem memory T-lymphocytes enhances antitumor responses while curtailing cytokine release syndrome, J. Clin. Invest., 132, e150807, 10.1172/JCI150807 Fraietta, 2018, Determinants of response and resistance to CD19 chimeric antigen receptor (CAR) T cell therapy of chronic lymphocytic leukemia, Nat. Med., 24, 563, 10.1038/s41591-018-0010-1 Fraietta, 2018, Disruption of TET2 promotes the therapeutic efficacy of CD19-targeted T cells, Nature, 558, 307, 10.1038/s41586-018-0178-z Majzner, 2018, Tumor antigen escape from CAR T-cell therapy, Cancer Discov., 8, 1219, 10.1158/2159-8290.CD-18-0442 Sabatino, 2016, Generation of clinical-grade CD19-specific CAR-modified CD8+ memory stem cells for the treatment of human B-cell malignancies, Blood, 128, 519, 10.1182/blood-2015-11-683847 Turtle, 2016, CD19 CAR-T cells of defined CD4+:CD8+ composition in adult B cell ALL patients, J. Clin. Invest., 126, 2123, 10.1172/JCI85309 Shah, 2018, A Phase 1 study with point-of-care manufacturing of dual targeted, tandem anti-CD19, anti-CD20 chimeric antigen receptor modified T (CAR-T) cells for relapsed, refractory, non-Hodgkin lymphoma, Blood, 132, 4193, 10.1182/blood-2018-99-110194 Benjamin, 2018, Preliminary data on safety, cellular kinetics and anti-leukemic activity of UCART19, an allogeneic anti-CD19 CAR T-cell product, in a pool of adult and pediatric patients with high-risk CD19+ relapsed/refractory B-cell acute lymphoblastic leukemia, Blood, 132, 896, 10.1182/blood-2018-99-111356 Murthy, 2019, Cytokine release syndrome: current perspectives, ImmunoTargets Ther., 8, 43, 10.2147/ITT.S202015 Labanieh, 2018, Programming CAR-T cells to kill cancer, Nat. Biomed. Eng., 2, 377, 10.1038/s41551-018-0235-9 Liu, 2020, Use of CAR-transduced natural killer cells in CD19-positive lymphoid tumors, N. Engl. J. Med., 382, 545, 10.1056/NEJMoa1910607 Petersen, 2018, Improving T-cell expansion and function for adoptive T-cell therapy using ex vivo treatment with PI3Kd inhibitors and VIP antagonists, Blood Adv., 2, 210, 10.1182/bloodadvances.2017011254 Majzner, 2022, GD2-CAR T cell therapy for H3K27M-mutated diffuse midline gliomas, Nature, 603, 934, 10.1038/s41586-022-04489-4 Mount, 2022, Advances in chimeric antigen receptor (CAR) T-cell therapies for the treatment of primary brain tumors, Antibodies (Basel), 11, 31, 10.3390/antib11020031 Qi, 2022, Claudin18.2-specific CAR T cells in gastrointestinal cancers: phase 1 trial interim results, Nat. Med., 28, 1189, 10.1038/s41591-022-01800-8 Depil, 2020, ‘Off-the-shelf’ allogeneic CAR T cells: development and challenges, Nat. Rev. Drug Discov., 19, 185, 10.1038/s41573-019-0051-2 Ruella, 2017, Next-generation chimeric antigen receptor T-cell therapy: going off the shelf, BioDrugs, 31, 473, 10.1007/s40259-017-0247-0 Sommer, 2019, Preclinical evaluation of allogeneic CAR T cells targeting BCMA for the treatment of multiple myeloma, Mol. Ther., 27, 1126, 10.1016/j.ymthe.2019.04.001 Perez, 2020, Off-the-shelf allogeneic T cell therapies for cancer: opportunities and challenges using naturally occurring “universal” donor T cells, Front. Immunol., 11, 583716, 10.3389/fimmu.2020.583716 Ren, 2017, Multiplex genome editing to generate universal CAR T cells resistant to PD1 inhibition, Clin. Cancer Res., 23, 2255, 10.1158/1078-0432.CCR-16-1300 Chaidos, 2012, Graft invariant natural killer T-cell dose predicts risk of acute graft-versus-host disease in allogeneic hematopoietic stem cell transplantation, Blood, 119, 5030, 10.1182/blood-2011-11-389304 Liu, 2018, 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, 10.1038/leu.2017.226 Tugues, 2018, Graft-versus-host disease, but not graft-versus-leukemia immunity, is mediated by GM-CSF-licensed myeloid cells, Sci. Transl. Med., 10, eaat8410, 10.1126/scitranslmed.aat8410 Delfanti, 2022, Adoptive immunotherapy with engineered iNKT cells to target cancer cells and the suppressive microenvironment, Front. Med. (Lausanne), 9, 897750, 10.3389/fmed.2022.897750 Bae, 2019, Roles of NKT cells in cancer immunotherapy, Arch. Pharm. Res., 42, 543, 10.1007/s12272-019-01139-8 Godfrey, 2018, Unconventional T cell targets for cancer immunotherapy, Immunity, 48, 453, 10.1016/j.immuni.2018.03.009 Zhang, 2020, Targeting natural killer cells for tumor immunotherapy, Front. Immunol., 11, 60, 10.3389/fimmu.2020.00060 Li, 2022, Mucosal-associated invariant T cells for cancer immunotherapy, Mol. Ther., 31, 631, 10.1016/j.ymthe.2022.11.019 Cortés-Selva, 2021, Innate and innate-like cells: the future of chimeric antigen receptor (CAR) cell therapy, Trends Pharmacol. Sci., 42, 45, 10.1016/j.tips.2020.11.004 Bulcha, 2021, Viral vector platforms within the gene therapy landscape, Signal Transduct. Target. Ther., 6, 53, 10.1038/s41392-021-00487-6 Ellis, 2021, Genetic engineering of T cells for immunotherapy, Nat. Rev. Genet., 22, 427, 10.1038/s41576-021-00329-9 Weber, 2020, The emerging landscape of immune cell therapies, Cell, 181, 46, 10.1016/j.cell.2020.03.001 Cascalho, 2005, Basic mechanisms of humoral rejection, Pediatr. Transplant., 9, 9, 10.1111/j.1399-3046.2004.00231.x Montgomery, 2011, Humoral immunity and antibody-mediated rejection in solid organ transplantation, Semin. Immunol., 23, 224, 10.1016/j.smim.2011.08.021 Figueiredo, 2015, A future with less HLA: potential clinical applications of HLA-universal cells, Tissue Antigens, 85, 443, 10.1111/tan.12564 Gornalusse, 2017, HLA-E-expressing pluripotent stem cells escape allogeneic responses and lysis by NK cells, Nat. Biotechnol., 35, 765, 10.1038/nbt.3860 Li, 2021, Development of stem cell-derived immune cells for off-the-shelf cancer immunotherapies, Cells, 10, 10.3390/cells10123497 Li, 2021, Engineering stem cells for cancer immunotherapy, Trends Cancer, 7, 1059, 10.1016/j.trecan.2021.08.004 Zhu, 2018, Concise review: human pluripotent stem cells to produce cell-based cancer immunotherapy, Stem Cells, 36, 134, 10.1002/stem.2754 Vodyanik, 2005, Human embryonic stem cell-derived CD34+ cells: efficient production in the coculture with OP9 stromal cells and analysis of lymphohematopoietic potential, Blood, 105, 617, 10.1182/blood-2004-04-1649 Kaufman, 2001, Hematopoietic colony-forming cells derived from human embryonic stem cells, Proc. Natl. Acad. Sci. USA, 98, 10716, 10.1073/pnas.191362598 Ng, 2008, A protocol describing the use of a recombinant protein-based, animal product-free medium (APEL) for human embryonic stem cell differentiation as spin embryoid bodies, Nat. Protoc., 3, 768, 10.1038/nprot.2008.42 Kennedy, 2012, T lymphocyte potential marks the emergence of definitive hematopoietic progenitors in human pluripotent stem cell differentiation cultures, Cell Rep., 2, 1722, 10.1016/j.celrep.2012.11.003 Kitayama, 2016, Cellular adjuvant properties, direct cytotoxicity of re-differentiated Vα24 invariant NKT-like cells from human induced pluripotent stem cells, Stem Cell Rep., 6, 213, 10.1016/j.stemcr.2016.01.005 Nishimura, 2019, Generation of antigen-specific T cells from human induced pluripotent stem cells, Methods Mol. Biol., 1899, 25, 10.1007/978-1-4939-8938-6_3 Nishimura, 2013, Generation of rejuvenated antigen-specific T cells by reprogramming to pluripotency and redifferentiation, Cell Stem Cell, 12, 114, 10.1016/j.stem.2012.11.002 Vizcardo, 2013, Regeneration of human tumor antigen-specific T cells from iPSCs derived from mature CD8(+) T cells, Cell Stem Cell, 12, 31, 10.1016/j.stem.2012.12.006 Wakao, 2013, Expansion of functional human mucosal-associated invariant T cells via reprogramming to pluripotency and redifferentiation, Cell Stem Cell, 12, 546, 10.1016/j.stem.2013.03.001 Zeng, 2017, Generation of “off-the-shelf” natural killer cells from peripheral blood cell-derived induced pluripotent stem cells, Stem Cell Rep., 9, 1796, 10.1016/j.stemcr.2017.10.020 Brentjens, 2011, Safety and persistence of adoptively transferred autologous CD19-targeted T cells in patients with relapsed or chemotherapy refractory B-cell leukemias, Blood, 118, 4817, 10.1182/blood-2011-04-348540 Brentjens, 2013, CD19-targeted T cells rapidly induce molecular remissions in adults with chemotherapy-refractory acute lymphoblastic leukemia, Sci. Transl. Med., 5, 177ra38, 10.1126/scitranslmed.3005930 Davila, 2014, Efficacy and toxicity management of 19–28z CAR T cell therapy in B cell acute lymphoblastic leukemia, Sci. Transl. Med., 6, 224ra25, 10.1126/scitranslmed.3008226 Kochenderfer, 2012, B-cell depletion and remissions of malignancy along with cytokine-associated toxicity in a clinical trial of anti-CD19 chimeric-antigen-receptor-transduced T cells, Blood, 119, 2709, 10.1182/blood-2011-10-384388 Robbins, 2011, Tumor regression in patients with metastatic synovial cell sarcoma and melanoma using genetically engineered lymphocytes reactive with NY-ESO-1, J. Clin. Oncol., 29, 917, 10.1200/JCO.2010.32.2537 Rapoport, 2015, NY-ESO-1–specific TCR–engineered T cells mediate sustained antigen-specific antitumor effects in myeloma, Nat. Med., 21, 914, 10.1038/nm.3910 Zhang, 2022, TCR engineered T cells for solid tumor immunotherapy, Exp. Hematol. Oncol., 11, 38, 10.1186/s40164-022-00291-0 Themeli, 2013, Generation of tumor-targeted human T lymphocytes from induced pluripotent stem cells for cancer therapy, Nat. Biotechnol., 31, 928, 10.1038/nbt.2678 Abramson, 2020, Lisocabtagene maraleucel for patients with relapsed or refractory large B-cell lymphomas (Transcend NHL 001): a multicentre seamless design study, Lancet, 396, 839, 10.1016/S0140-6736(20)31366-0 Kharfan-Dabaja, 2022, Lisocabtagene maraleucel in relapsed or refractory diffuse large B cell lymphoma: what is the evidence?, Hematol. Oncol. Stem Cell Ther., 15, 168 Li, 2021, Development of allogeneic HSC-engineered iNKT cells for off-the-shelf cancer immunotherapy. Cell reports, Cell Rep. Med., 2, 100449, 10.1016/j.xcrm.2021.100449 Seet, 2017, Generation of mature T cells from human hematopoietic stem and progenitor cells in artificial thymic organoids, Nat. Methods, 14, 521, 10.1038/nmeth.4237 Montel-Hagen, 2019, Organoid-induced differentiation of conventional T cells from human pluripotent stem cells, Cell Stem Cell, 24, 376, 10.1016/j.stem.2018.12.011 Smith, 2015, In vitro T-cell generation from adult, embryonic, and induced pluripotent stem cells: many roads to one destination, Stem Cells, 33, 3174, 10.1002/stem.2115 Robey, 1996, An activated form of Notch influences the choice between CD4 and CD8 T cell lineages, Cell, 87, 483, 10.1016/S0092-8674(00)81368-9 Robey, 1999, Regulation of T cell fate by Notch, Annu. Rev. Immunol., 17, 283, 10.1146/annurev.immunol.17.1.283 Yasutomo, 2000, The duration of antigen receptor signalling determines CD4+ versus CD8+ T-cell lineage fate, Nature, 404, 506, 10.1038/35006664 Li, 2018, Human iPSC-derived natural killer cells engineered with chimeric antigen receptors enhance anti-tumor activity, Cell Stem Cell, 23, 181, 10.1016/j.stem.2018.06.002 Goldenson, 2020, Umbilical cord blood and iPSC-derived natural killer cells demonstrate key differences in cytotoxic activity and KIR profiles, Front. Immunol., 11, 561553, 10.3389/fimmu.2020.561553 Hermanson, 2016, Induced pluripotent stem cell-derived natural killer cells for treatment of ovarian cancer, Stem Cells, 34, 93, 10.1002/stem.2230 Rezvani, 2017, Engineering natural killer cells for cancer immunotherapy, Mol. Ther., 25, 1769, 10.1016/j.ymthe.2017.06.012 Cany, 2015, Combined IL-15 and IL-12 drives the generation of CD34+-derived natural killer cells with superior maturation and alloreactivity potential following adoptive transfer, Oncoimmunology, 4, e1017701, 10.1080/2162402X.2015.1017701 Cichocki, 2020, 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, 10.1126/scitranslmed.aaz5618 Fang, 2017, NK cell-based immunotherapy for cancer, Semin. Immunol., 31, 37, 10.1016/j.smim.2017.07.009 Tang, 2018, 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 Woll, 2005, Human embryonic stem cell-derived NK cells acquire functional receptors and cytolytic activity, J. Immunol., 175, 5095, 10.4049/jimmunol.175.8.5095 Zhu, 2019, An improved method to produce clinical-scale natural killer cells from human pluripotent stem cells, Methods Mol. Biol., 2048, 107, 10.1007/978-1-4939-9728-2_12 Zhu, 2020, Metabolic reprograming via deletion of CISH in human iPSC-derived NK cells promotes in vivo persistence and enhances anti-tumor activity, Cell Stem Cell, 27, 224, 10.1016/j.stem.2020.05.008 Abel, 2018, Natural killer cells: development, maturation, and clinical utilization, Front. Immunol., 9, 1869, 10.3389/fimmu.2018.01869 Liu, 2019, Role of NKG2D and its ligands in cancer immunotherapy, Am. J. Cancer Res., 9, 2064 Ruggeri, 2007, Donor natural killer cell allorecognition of missing self in haploidentical hematopoietic transplantation for acute myeloid leukemia: challenging its predictive value, Blood, 110, 433, 10.1182/blood-2006-07-038687 Hu, 2019, Cancer immunotherapy based on natural killer cells: current progress and new opportunities, Front. Immunol., 10, 1205, 10.3389/fimmu.2019.01205 Mehta, 2018, Chimeric antigen receptor expressing natural killer cells for the immunotherapy of cancer, Front. Immunol., 9, 283, 10.3389/fimmu.2018.00283 Zhu, 2020, Pluripotent stem cell-derived NK cells with high-affinity noncleavable CD16a mediate improved antitumor activity, Blood, 135, 399, 10.1182/blood.2019000621 Alfarra, 2020, Targeting NK cell inhibitory receptors for precision multiple myeloma immunotherapy, Front. Immunol., 11, 575609, 10.3389/fimmu.2020.575609 Fujii, 2013, NKT cells as an ideal anti-tumor immunotherapeutic, Front. Immunol., 4, 1, 10.3389/fimmu.2013.00409 Li, 2022, Development of off – the – shelf hematopoietic stem cell – engineered invariant natural killer T cells for COVID - 19 therapeutic intervention, Stem Cell Res. Ther., 13, 112, 10.1186/s13287-022-02787-2 Zhu, 2019, Development of hematopoietic stem cell-engineered invariant natural killer T cell therapy for cancer, Cell Stem Cell, 25, 542, 10.1016/j.stem.2019.08.004 Smith, 2015, Genetic engineering of hematopoietic stem cells to generate invariant natural killer T cells, Proc. Natl. Acad. Sci. USA, 112, 1523, 10.1073/pnas.1424877112 Wakao, 2020, Reprogramming of MAIT cells to pluripotency and redifferentiation, Methods Mol. Biol., 2098, 237, 10.1007/978-1-0716-0207-2_16 Wakao, 2013, Toward the realization of cell therapy: the advent of MAIT cells from iPSCs, Cell Cycle, 12, 2341, 10.4161/cc.25706 Zhou, 2021, Methods for studying mouse and human invariant natural killer T cells, Methods Mol. Biol., 2388, 35, 10.1007/978-1-0716-1775-5_4 Hansen, 2018, Efficient production of erythroid, megakaryocytic and myeloid cells, using single cell-derived iPSC colony differentiation, Stem Cell Res., 29, 232, 10.1016/j.scr.2018.04.016 Horton, 2020, Induced pluripotent stem cells reprogrammed from primary dendritic cells provide an abundant source of immunostimulatory dendritic cells for use in immunotherapy, Stem Cells, 38, 67, 10.1002/stem.3095 Sachamitr, 2018, Directed differentiation of human induced pluripotent stem cells into dendritic cells displaying tolerogenic properties and resembling the CD141(+) subset, Front. Immunol., 8, 1935, 10.3389/fimmu.2017.01935 Zhang, 2020, Pluripotent stem cell-derived CAR-macrophage cells with antigen-dependent anti-cancer cell functions, J. Hematol. Oncol., 13, 153, 10.1186/s13045-020-00983-2 Kitchen, 2009, Engineering antigen-specific T cells from genetically modified human hematopoietic stem cells in immunodeficient mice, PLoS One, 4, e8208, 10.1371/journal.pone.0008208 Lan, 2006, Reconstitution of a functional human immune system in immunodeficient mice through combined human fetal thymus/liver and CD34+ cell transplantation, Blood, 108, 487, 10.1182/blood-2005-11-4388 Melkus, 2006, Humanized mice mount specific adaptive and innate immune responses to EBV and TSST-1, Nat. Med., 12, 1316, 10.1038/nm1431 Kim, 2007, A novel culture technique for human embryonic stem cells using porous membranes, Stem Cells, 25, 2601, 10.1634/stemcells.2006-0814 Yu, 2015, Feeder Cell Sources and Feeder-Free Methods for Human iPS Cell Culture BT – Interface Oral Health Science 2014, 145 Lu, 2010, Derivation and long-term culture of human parthenogenetic embryonic stem cells using human foreskin feeders, J. Assist. Reprod. Genet., 27, 285, 10.1007/s10815-010-9408-5 Park, 2011, Human feeder cells can support the undifferentiated growth of human and mouse embryonic stem cells using their own basic fibroblast growth factors, Stem Cells Dev., 20, 1901, 10.1089/scd.2010.0496 Unger, 2009, Immortalized human skin fibroblast feeder cells support growth and maintenance of both human embryonic and induced pluripotent stem cells, Hum. Reprod., 24, 2567, 10.1093/humrep/dep232 Pekkanen-Mattila, 2012, The effect of human and mouse fibroblast feeder cells on cardiac differentiation of human pluripotent stem cells, Stem Cells Int., 2012, 875059, 10.1155/2012/875059 Huijskens, 2014, Technical advance: ascorbic acid induces development of double-positive T cells from human hematopoietic stem cells in the absence of stromal cells, J. Leukoc. Biol., 96, 1165, 10.1189/jlb.1TA0214-121RR Shukla, 2017, Progenitor T-cell differentiation from hematopoietic stem cells using Delta-like-4 and VCAM-1, Nat. Methods, 14, 531, 10.1038/nmeth.4258 Iriguchi, 2021, A clinically applicable and scalable method to regenerate T-cells from iPSCs for off-the-shelf T-cell immunotherapy, Nat. Commun., 12, 430, 10.1038/s41467-020-20658-3 Johnson, 2006, Gene transfer of tumor-reactive TCR confers both high avidity and tumor reactivity to nonreactive peripheral blood mononuclear cells and tumor-infiltrating lymphocytes, J. Immunol., 177, 6548, 10.4049/jimmunol.177.9.6548 Cohen, 2005, Recognition of fresh human tumor by human peripheral blood lymphocytes transduced with a bicistronic retroviral vector encoding a murine anti-p53 TCR, J. Immunol., 175, 5799, 10.4049/jimmunol.175.9.5799 Parkhurst, 2009, Characterization of genetically modified T-cell receptors that recognize the CEA:691–699 peptide in the context of HLA-A2.1 on human colorectal cancer cells, Clin. Cancer Res., 15, 169, 10.1158/1078-0432.CCR-08-1638 Li, 2005, Directed evolution of human T-cell receptors with picomolar affinities by phage display, Nat. Biotechnol., 23, 349, 10.1038/nbt1070 Varela-Rohena, 2008, Control of HIV-1 immune escape by CD8 T cells expressing enhanced T-cell receptor, Nat. Med., 14, 1390, 10.1038/nm.1779 Adair, 2017, Hematopoietic stem cell approaches to cancer, Hematol. Oncol. Clin. North Am., 31, 897, 10.1016/j.hoc.2017.06.012 Milone, 2018, Clinical use of lentiviral vectors, Leukemia, 32, 1529, 10.1038/s41375-018-0106-0 Yang, 2005, Long-term in vivo provision of antigen-specific T cell immunity by programming hematopoietic stem cells, Proc. Natl. Acad. Sci. USA, 102, 4518, 10.1073/pnas.0500600102 Yang, 2002, Generation of functional antigen-specific T cells in defined genetic backgrounds by retrovirus-mediated expression of TCR cDNAS in hematopoietic precursor cells, Proc. Natl. Acad. Sci. USA, 99, 6204, 10.1073/pnas.092154599 Basar, 2020, Next-generation cell therapies: the emerging role of CAR-NK cells, Blood Adv., 4, 5868, 10.1182/bloodadvances.2020002547 Qasim, 2017, Molecular remission of infant B-ALL after infusion of universal TALEN gene-edited CAR T cells, Sci. Transl. Med., 9, 1, 10.1126/scitranslmed.aaj2013 Webber, 2019, Highly efficient multiplex human T cell engineering without double-strand breaks using Cas9 base editors, Nat. Commun., 10, 5222, 10.1038/s41467-019-13007-6 Zhao, 2018, Universal CARs, universal T cells, and universal CAR T cells, J. Hematol. Oncol., 11, 132, 10.1186/s13045-018-0677-2 Torikai, 2013, Toward eliminating HLA class I expression to generate universal cells from allogeneic donors, Blood, 122, 1341, 10.1182/blood-2013-03-478255 Xu, 2019, Mechanisms of relapse after CD19 CAR T-cell therapy for acute lymphoblastic leukemia and its prevention and treatment strategies, Front. Immunol., 10, 2664, 10.3389/fimmu.2019.02664 Zhang, 2017, CRISPR-Cas9 mediated LAG-3 disruption in CAR-T cells, Front. Med., 11, 554, 10.1007/s11684-017-0543-6 Beane, 2015, Clinical scale zinc finger nuclease-mediated gene editing of PD-1 in tumor infiltrating lymphocytes for the treatment of metastatic melanoma, Mol. Ther., 23, 1380, 10.1038/mt.2015.71 Jang, 2019, Development of immunocompatible pluripotent stem cells via CRISPR-based human leukocyte antigen engineering, Exp. Mol. Med., 51, 1, 10.1038/s12276-019-0352-x Jung, 2018, CRISPR/Cas9-mediated knockout of DGK improves antitumor activities of human T cells, Cancer Res., 78, 4692, 10.1158/0008-5472.CAN-18-0030 Liu, 2017, CRISPR-Cas9-mediated multiplex gene editing in CAR-T cells, Cell Res., 27, 154, 10.1038/cr.2016.142 Provasi, 2012, Editing T cell specificity towards leukemia by zinc finger nucleases and lentiviral gene transfer, Nat. Med., 18, 807, 10.1038/nm.2700 Rupp, 2017, CRISPR/Cas9-mediated PD-1 disruption enhances anti-tumor efficacy of human chimeric antigen receptor T cells, Sci. Rep., 7, 737, 10.1038/s41598-017-00462-8 Shi, 2017, CRISPR knock out CTLA-4 enhances the anti-tumor activity of cytotoxic T lymphocytes, Gene, 636, 36, 10.1016/j.gene.2017.09.010 Thongsin, 2022, CRISPR/Cas9 ribonucleoprotein complex-mediated efficient B2M knockout in human induced pluripotent stem cells (iPSCs), Methods Mol. Biol., 2454, 607, 10.1007/7651_2021_352 Eyquem, 2017, Targeting a CAR to the TRAC locus with CRISPR/Cas9 enhances tumour rejection, Nature, 543, 113, 10.1038/nature21405 MacLeod, 2017, Integration of a CD19 CAR into the TCR alpha chain locus streamlines production of allogeneic gene-edited CAR T cells, Mol. Ther., 25, 949, 10.1016/j.ymthe.2017.02.005 Joung, 2017, Genome-scale CRISPR-Cas9 knockout and transcriptional activation screening, Nat. Protoc., 12, 828, 10.1038/nprot.2017.016 Joung, 2022, CRISPR activation screen identifies BCL-2 proteins and B3GNT2 as drivers of cancer resistance to T cell-mediated cytotoxicity, Nat. Commun., 13, 1606, 10.1038/s41467-022-29205-8 Larson, 2022, CAR T cell killing requires the IFNγR pathway in solid but not liquid tumours, Nature, 604, 563, 10.1038/s41586-022-04585-5 Sanson, 2018, Optimized libraries for CRISPR-Cas9 genetic screens with multiple modalities, Nat. Commun., 9, 5416, 10.1038/s41467-018-07901-8 Shifrut, 2018, Genome-wide CRISPR screens in primary human T cells reveal key regulators of immune function, Cell, 175, 1958, 10.1016/j.cell.2018.10.024 Wang, 2021, Crispr screening of car T cells and cancer stem cells reveals critical dependencies for cell-based therapies, Cancer Discov., 11, 1192, 10.1158/2159-8290.CD-20-1243 Zhang, 2020, Engineering T Cells Using CRISPR/Cas9 for Cancer Therapy, 419 Fujisaki, 2009, Expansion of highly cytotoxic human natural killer cells for cancer cell therapy, Cancer Res., 69, 4010, 10.1158/0008-5472.CAN-08-3712 Heczey, 2020, Anti-GD2 CAR-NKT cells in patients with relapsed or refractory neuroblastoma: an interim analysis, Nat. Med., 26, 1686, 10.1038/s41591-020-1074-2 Liu, 2012, IL-15 protects NKT cells from inhibition by tumor-associated macrophages and enhances antimetastatic activity, J. Clin. Invest., 122, 2221, 10.1172/JCI59535 Xu, 2019, 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, 10.1158/1078-0432.CCR-19-0421 Riteau, 2001, HLA-G1 co-expression boosts the HLA class I-mediated NK lysis inhibition, Int. Immunol., 13, 193, 10.1093/intimm/13.2.193 Lee, 1998, HLA-E is a major ligand for the natural killer inhibitory receptor CD94/NKG2A, Proc. Natl. Acad. Sci. USA, 95, 5199, 10.1073/pnas.95.9.5199 Benjamin, 2020, 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, 10.1016/S0140-6736(20)32334-5 Bedford, 2018, Considering cell therapy product “good manufacturing practice” status, Front. Med. (Lausanne), 5, 118, 10.3389/fmed.2018.00118 Giancola, 2012, Cell therapy: cGMP facilities and manufacturing, Muscles Ligaments Tendons J., 2, 243 Carpenter, 2015, Concise review: making and using clinically compliant pluripotent stem cell lines, Stem Cells Transl. Med., 4, 381, 10.5966/sctm.2014-0202 Carpenter, 2009, Developing safe therapies from human pluripotent stem cells, Nat. Biotechnol., 27, 606, 10.1038/nbt0709-606 Panchision, 2013, Meeting report: using stem cells for biological and therapeutics discovery in mental illness, April 2012, Stem Cells Transl. Med., 2, 217, 10.5966/sctm.2012-0149 Kleitman, 2013, Pluripotent stem cells in translation: a Food and Drug Administration-National Institutes of Health collaboration, Stem Cells Transl. Med., 2, 483, 10.5966/sctm.2013-0042 Frey-Vasconcells, 2012, Translation of stem cell research: points to consider in designing preclinical animal studies, Stem Cells Transl. Med., 1, 353, 10.5966/sctm.2012-0018 Brudno, 2019, Recent advances in CAR T-cell toxicity: mechanisms, manifestations and management, Blood Rev., 34, 45, 10.1016/j.blre.2018.11.002 Zhou, 2022, Engineering induced pluripotent stem cells for cancer immunotherapy, Cancers, 14, 2266, 10.3390/cancers14092266 Furukawa, 2022, Advances in allogeneic cancer cell therapy and future perspectives on “Off-the-shelf” T cell therapy using iPSC technology and gene editing, Cells, 11, 269, 10.3390/cells11020269 Yamanaka, 2020, Pluripotent stem cell-based cell therapy-promise and challenges, Cell Stem Cell, 27, 523, 10.1016/j.stem.2020.09.014 Ben-David, 2018, Genetic and transcriptional evolution alters cancer cell line drug response, Nature, 560, 325, 10.1038/s41586-018-0409-3 Sato, 2019, Tumorigenicity assessment of cell therapy products: the need for global consensus and points to consider, Cytotherapy, 21, 1095, 10.1016/j.jcyt.2019.10.001 Peruzzi, 2013, Karyotype asymmetry: again, how to measure and what to measure?, Comp. Cytogenet., 7, 1, 10.3897/compcytogen.v7i1.4431 Seki, 2010, Generation of induced pluripotent stem cells from human terminally differentiated circulating T cells, Cell Stem Cell, 7, 11, 10.1016/j.stem.2010.06.003 Okita, 2007, Generation of germline-competent induced pluripotent stem cells, Nature, 448, 313, 10.1038/nature05934 Li, 2000, A cytoplasmic RNA vector derived from nontransmissible Sendai virus with efficient gene transfer and expression, J. Virol., 74, 6564, 10.1128/JVI.74.14.6564-6569.2000 Yu, 2009, Human induced pluripotent stem cells free of vector and transgene sequences, Science, 324, 797, 10.1126/science.1172482 Mashima, 2021, Improved safety of induced pluripotent stem cell-derived antigen-presenting cell-based cancer immunotherapy, Mol. Ther. Methods Clin. Dev., 21, 171, 10.1016/j.omtm.2021.03.002 Karantalis, 2015, Allogeneic cell therapy: a new paradigm in therapeutics, Circ. Res., 116, 12, 10.1161/CIRCRESAHA.114.305495 Appelbaum, 2001, Haematopoietic cell transplantation as immunotherapy, Nature, 411, 385, 10.1038/35077251 Hill, 2021, Current concepts and advances in graft-versus-host disease immunology, Annu. Rev. Immunol., 39, 19, 10.1146/annurev-immunol-102119-073227 Martinez-Cibrian, 2021, Graft-versus-host disease prophylaxis: pathophysiology-based review on current approaches and future directions, Blood Rev., 48, 100792, 10.1016/j.blre.2020.100792 Penack, 2020, Prophylaxis and management of graft versus host disease after stem-cell transplantation for haematological malignancies: updated consensus recommendations of the European Society for Blood and Marrow Transplantation, Lancet Haematol., 7, e157, 10.1016/S2352-3026(19)30256-X Sabry, 2020, Killers at the crossroads: the use of innate immune cells in adoptive cellular therapy of cancer, Stem Cells Transl. Med., 9, 974, 10.1002/sctm.19-0423 Lan, 2003, Host conditioning with total lymphoid irradiation and antithymocyte globulin prevents graft-versus-host disease: the role of CD1-reactive natural killer T cells, Biol. Blood Marrow Transplant. J. Am. Soc. Blood Marrow Transplant., 9, 355, 10.1016/S1083-8791(03)00108-3 Yamasaki, 2003, Influence of transplanted dose of CD56+ cells on development of graft-versus-host disease in patients receiving G-CSF-mobilized peripheral blood progenitor cells from HLA-identical sibling donors, Bone Marrow Transplant., 32, 505, 10.1038/sj.bmt.1704165 Fishman, 2017, Infection in organ transplantation, Am. J. Transplant., 17, 856, 10.1111/ajt.14208 Holt, 2017, Overview of immunosuppressive therapy in solid organ transplantation, Anesthesiol. Clin., 35, 365, 10.1016/j.anclin.2017.04.001 van Rood, 2008, Eleven million donors in Bone Marrow Donors Worldwide! Time for reassessment?, Bone Marrow Transplant., 41, 1, 10.1038/sj.bmt.1705866 Hurley, 2021, Naming HLA diversity: a review of HLA nomenclature, Hum. Immunol., 82, 457, 10.1016/j.humimm.2020.03.005 Taylor, 2005, Banking on human embryonic stem cells: estimating the number of donor cell lines needed for HLA matching, Lancet, 366, 2019, 10.1016/S0140-6736(05)67813-0 Lanza, 2019, Engineering universal cells that evade immune detection, Nat. Rev. Immunol., 19, 723, 10.1038/s41577-019-0200-1 Ordikhani, 2020, Macrophages in organ transplantation, Front. Immunol., 11, 582939, 10.3389/fimmu.2020.582939 Deuse, 2019, Hypoimmunogenic derivatives of induced pluripotent stem cells evade immune rejection in fully immunocompetent allogeneic recipients, Nat. Biotechnol., 37, 252, 10.1038/s41587-019-0016-3 Wang, 2021, Generation of hypoimmunogenic T cells from genetically engineered allogeneic human induced pluripotent stem cells, Nat. Biomed. Eng., 5, 429, 10.1038/s41551-021-00730-z Holstein, 2020, CAR T-cell therapy in hematologic malignancies: A voyage in progress, Clin. Pharmacol. Ther., 107, 112, 10.1002/cpt.1674 Han, 2021, Current progress in CAR-T cell therapy for hematological malignancies, J. Cancer, 12, 326, 10.7150/jca.48976 Shah, 2019, Multi targeted CAR-T cell therapies for B-cell malignancies, Front. Oncol., 9, 146, 10.3389/fonc.2019.00146 Newick, 2017, CAR T cell therapy for solid tumors, Annu. Rev. Med., 68, 139, 10.1146/annurev-med-062315-120245 Liu, 2017, Driving better and safer HER2-specific CARs for cancer therapy, Oncotarget, 8, 62730, 10.18632/oncotarget.17528 Navai, 2019, Abstract LB-147: Administration of HER2-CAR T cells after lymphodepletion safely improves T cell expansion and induces clinical responses in patients with advanced sarcomas, Cancer Res., 79, LB-147, 10.1158/1538-7445.AM2019-LB-147 Marofi, 2021, CAR T cells in solid tumors: challenges and opportunities, Stem Cell Res. Ther., 12, 81, 10.1186/s13287-020-02128-1 Cordoba, 2021, CAR T cells with dual targeting of CD19 and CD22 in pediatric and young adult patients with relapsed or refractory B cell acute lymphoblastic leukemia: a phase 1 trial, Nat. Med., 27, 1797, 10.1038/s41591-021-01497-1 van der Schans, 2020, Dual targeting to overcome current challenges in multiple myeloma CAR T-cell treatment, Front. Oncol., 10, 1362, 10.3389/fonc.2020.01362 Hirabayashi, 2021, Dual-targeting CAR-T cells with optimal co-stimulation and metabolic fitness enhance antitumor activity and prevent escape in solid tumors, Nat. Cancer, 2, 904, 10.1038/s43018-021-00244-2 Zah, 2020, Systematically optimized BCMA/CS1 bispecific CAR-T cells robustly control heterogeneous multiple myeloma, Nat. Commun., 11, 2283, 10.1038/s41467-020-16160-5 Roybal, 2016, Engineering T cells with customized therapeutic response programs using synthetic notch receptors, Cell, 167, 419, 10.1016/j.cell.2016.09.011 Srivastava, 2019, Logic-gated ROR1 chimeric antigen receptor expression rescues T cell-mediated toxicity to normal tissues and enables selective tumor targeting, Cancer Cell, 35, 489, 10.1016/j.ccell.2019.02.003 Choe, 2021, SynNotch-CAR T cells overcome challenges of specificity, heterogeneity, and persistence in treating glioblastoma, Sci. Transl. Med., 13, eabe7378, 10.1126/scitranslmed.abe7378 Salter, 2021, Comparative analysis of TCR and CAR signaling informs CAR designs with superior antigen sensitivity and in vivo function, Sci. Signal., 14, eabe2606, 10.1126/scisignal.abe2606 Mansilla-Soto, 2022, HLA-independent T cell receptors for targeting tumors with low antigen density, Nat. Med., 28, 345, 10.1038/s41591-021-01621-1 Dixon, 2021, Engineering anti-tumor monoclonal antibodies and Fc receptors to enhance ADCC by human NK cells, Cancers, 13, 312, 10.3390/cancers13020312 Jing, 2015, Identification of an ADAM17 cleavage region in human CD16 (FcγRIII) and the engineering of a non-cleavable version of the receptor in NK cells, PLoS One, 10, e0121788, 10.1371/journal.pone.0121788 Koene, 1997, Fc gammaRIIIa-158V/F polymorphism influences the binding of IgG by natural killer cell Fc gammaRIIIa, independently of the Fc gammaRIIIa-48L/R/H phenotype, Blood, 90, 1109, 10.1182/blood.V90.3.1109 Saito, 2016, Reprogramming of melanoma tumor-infiltrating lymphocytes to induced pluripotent stem cells, Stem Cells Int., 2016, 8394960, 10.1155/2016/8394960 Woo, 2015, Innate immune recognition of cancer, Annu. Rev. Immunol., 33, 445, 10.1146/annurev-immunol-032414-112043 Deng, 2022, Gamma delta (γδ) T cells in cancer immunotherapy; where it comes from, where it will go?, Eur. J. Pharmacol., 919, 174803, 10.1016/j.ejphar.2022.174803 Zeng, 2019, Derivation of mimetic γδ T cells endowed with cancer recognition receptors from reprogrammed γδ T cell, PLoS One, 14, e0216815, 10.1371/journal.pone.0216815 Chan, 2021, Cellular networks controlling T cell persistence in adoptive cell therapy, Nat. Rev. Immunol., 21, 769, 10.1038/s41577-021-00539-6 McLellan, 2019, Chimeric antigen receptor T cell persistence and memory cell formation, Immunol. Cell Biol., 97, 664, 10.1111/imcb.12254 Yeku, 2016, Armored CAR T-cells: utilizing cytokines and pro-inflammatory ligands to enhance CAR T-cell anti-tumour efficacy, Biochem. Soc. Trans., 44, 412, 10.1042/BST20150291 Ohteki, 2002, Critical role for IL-15 in innate immunity, Curr. Mol. Med., 2, 371, 10.2174/1566524023362519 Makkouk, 2021, Off-the-shelf Vδ1 gamma Delta T cells engineered with glypican-3 (GPC-3)-specific chimeric antigen receptor (CAR) and soluble IL-15 display robust antitumor efficacy against hepatocellular carcinoma, J. Immunother. Cancer, 9, e003441, 10.1136/jitc-2021-003441 Allen, 2021, A bicistronic vector expressing CD16 and a membrane bound IL-15 construct in iPSC derived NK cells increased cytotoxicity and persistence, Blood, 138, 4809, 10.1182/blood-2021-153258 Christodoulou, 2021, Engineered interleukin-15 autocrine signaling invigorates anti-CD123 CAR-NK cells, Blood, 138, 2806, 10.1182/blood-2021-146609 Gerew, 2021, Deletion of CISH and TGFβR2 in iPSC-derived NK cells promotes high cytotoxicity and enhances in vivo tumor killing, Blood, 138, 2780, 10.1182/blood-2021-150731 Lynn, 2019, C-Jun overexpression in CAR T cells induces exhaustion resistance, Nature, 576, 293, 10.1038/s41586-019-1805-z Seo, 2021, BATF and IRF4 cooperate to counter exhaustion in tumor-infiltrating CAR T cells, Nat. Immunol., 22, 983, 10.1038/s41590-021-00964-8 Koyanagi-Aoi, 2013, Differentiation-defective phenotypes revealed by large-scale analyses of human pluripotent stem cells, Proc. Natl. Acad. Sci. USA, 110, 20569, 10.1073/pnas.1319061110 Osafune, 2008, Marked differences in differentiation propensity among human embryonic stem cell lines, Nat. Biotechnol., 26, 313, 10.1038/nbt1383 Yamanaka, 2012, Induced pluripotent stem cells: past, present, and future, Cell Stem Cell, 10, 678, 10.1016/j.stem.2012.05.005 Nishizawa, 2016, Epigenetic variation between human induced pluripotent stem cell lines is an indicator of differentiation capacity, Cell Stem Cell, 19, 341, 10.1016/j.stem.2016.06.019 Theunissen, 2014, Systematic identification of culture conditions for induction and maintenance of naive human pluripotency, Cell Stem Cell, 15, 471, 10.1016/j.stem.2014.07.002 Takashima, 2014, Resetting transcription factor control circuitry toward ground-state pluripotency in human, Cell, 158, 1254, 10.1016/j.cell.2014.08.029 Di Stefano, 2018, Reduced MEK inhibition preserves genomic stability in naive human embryonic stem cells, Nat. Methods, 15, 732, 10.1038/s41592-018-0104-1 Theunissen, 2016, Molecular criteria for defining the naive human pluripotent state, Cell Stem Cell, 19, 502, 10.1016/j.stem.2016.06.011 Imai, 2005, Genetic modification of primary natural killer cells overcomes inhibitory signals and induces specific killing of leukemic cells, Blood, 106, 376, 10.1182/blood-2004-12-4797 Ueda, 2020, Non-clinical efficacy, safety and stable clinical cell processing of induced pluripotent stem cell-derived anti-glypican-3 chimeric antigen receptor-expressing natural killer/innate lymphoid cells, Cancer Sci., 111, 1478, 10.1111/cas.14374 Hong, 2020, 380 Preliminary results of an ongoing phase I trial of FT500, a first-in-class, off-the-shelf, induced pluripotent stem cell (iPSC) derived natural killer (NK) cell therapy in advanced solid tumors, J. Immunother. Cancer, 8, A231.2 Cichocki, 2021, 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, 10.1182/blood-2021-148654 Hsu, 2021, An Alternative Cell Therapy for Cancers: Induced Pluripotent Stem Cell (iPSC)-Derived Natural Killer Cells, Biomedicines, 9, 10.3390/biomedicines9101323 Luevano, 2012, Generation of natural killer cells from hematopoietic stem cells in vitro for immunotherapy, Cell. Mol. Immunol., 9, 310, 10.1038/cmi.2012.17 Shah, 2017, Phase I study of cord blood-derived natural killer cells combined with autologous stem cell transplantation in multiple myeloma, Br. J. Haematol., 177, 457, 10.1111/bjh.14570 Vivier, 2012, Targeting natural killer cells and natural killer T cells in cancer, Nat. Rev. Immunol., 12, 239, 10.1038/nri3174 Ando, 2015, A safeguard system for induced pluripotent stem cell-derived rejuvenated T cell therapy, Stem Cell Rep., 5, 597, 10.1016/j.stemcr.2015.07.011 Minagawa, 2018, Enhancing T cell receptor stability in rejuvenated iPSC-derived T cells improves their use in cancer immunotherapy, Cell Stem Cell, 23, 850, 10.1016/j.stem.2018.10.005 Sadeqi Nezhad, 2021, Induced pluripotent stem cells (iPSCs) provide a potentially unlimited T cell source for CAR-T cell development and off-the-shelf products, Pharm. Res., 38, 931, 10.1007/s11095-021-03067-z Wang, 2022, 3D-organoid culture supports differentiation of human CAR+ iPSCs into highly functional CAR T cells, Cell Stem Cell, 29, 515, 10.1016/j.stem.2022.02.009 Jing, 2022, EZH1 repression generates mature iPSC-derived CAR T cells with enhanced antitumor activity, Cell Stem Cell, 29, 1181, 10.1016/j.stem.2022.06.014 Montoya, 2007, Characterization of human invariant natural killer T subsets in health and disease using a novel invariant natural killer T cell-clonotypic monoclonal antibody, 6B11, Immunology, 122, 1, 10.1111/j.1365-2567.2007.02647.x Li, 2022, 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, 10.1016/j.isci.2022.104859 Li, 2022, Targeting immunosuppressive tumor-associated macrophages using innate T cells for enhanced antitumor reactivity, Cancers, 14, 2749, 10.3390/cancers14112749 Metelitsa, 2011, Anti-tumor potential of type-I NKT cells against CD1d-positive and CD1d-negative tumors in humans, Clin. Immunol., 140, 119, 10.1016/j.clim.2010.10.005 Neff Newitt, 2022, The incredible story of Emily Whitehead & CAR T-cell therapy, Oncol. Times, 44, 1.19 Johansson, 2022, Multi-omics characterization of a human stem cell-based model of cardiac hypertrophy, Life (Basel), 12 Brooks, 2022, Functional genomics and the future of iPSCs in disease modeling, Stem Cell Rep., 17, 1033, 10.1016/j.stemcr.2022.03.019 Bayat Mokhtari, 2017, Combination therapy in combating cancer, Oncotarget, 8, 38022, 10.18632/oncotarget.16723