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:

Recent advances and discoveries in the mechanisms and functions of CAR T cells

Abstract

This Review discusses the major advances and changes made over the past 3 years to our understanding of chimeric antigen receptor (CAR) T cell efficacy and safety. Recently, the field has gained insight into how various molecular modules of the CAR influence signalling and function. We report on mechanisms of toxicity and resistance as well as novel engineering and pharmaceutical interventions to overcome these challenges. Looking forward, we discuss new targets and indications for CAR T cell therapy expected to reach the clinic in the next 1–2 years. We also consider some new studies that have implications for the future of CAR T cell therapies, including changes to manufacturing, allogeneic products and drug-regulatable CAR T cells.

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: Schematic of a basic second-generation CAR T cell.
Fig. 2: Current molecular understanding and therapeutic intervention of CAR T cell-induced cytokine release syndrome and neurotoxicity.
Fig. 3: Antigen-dependent and antigen-independent resistance.
Fig. 4: CAR T cell subsets with increased efficacy and universal CARs with interchangeable targets.

Similar content being viewed by others

References

  1. Stancovski, I. et al. Targeting of T lymphocytes to Neu/HER2-expressing cells using chimeric single chain Fv receptors. J. Immunol. 151, 6577–6582 (1993).

    CAS  PubMed  Google Scholar 

  2. Eshhar, Z. et al. The T-body approach: potential for cancer immunotherapy. Springer Semin. Immunopathol. 18, 199–209 (1996).

    CAS  PubMed  Google Scholar 

  3. Hwu, P. et al. Lysis of ovarian cancer cells by human lymphocytes redirected with a chimeric gene composed of an antibody variable region and the Fc receptor γ chain. J. Exp. Med. 178, 361–366 (1993).

    CAS  PubMed  Google Scholar 

  4. Hwu, P. et al. In vivo antitumor activity of T cells redirected with chimeric antibody/T-cell receptor genes. Cancer Res. 55, 3369–3373 (1995).

    CAS  PubMed  Google Scholar 

  5. Kershaw, M. H. et al. A phase I study on adoptive immunotherapy using gene-modified T cells for ovarian cancer. Clin. Cancer Res. 12, 6106–6115 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Krause, A. et al. Antigen-dependent CD28 signaling selectively enhances survival and proliferation in genetically modified activated human primary T lymphocytes. J. Exp. Med. 188, 619–626 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Vandenberghe, P. et al. Antibody and B7/BB1-mediated ligation of the CD28 receptor induces tyrosine phosphorylation in human T cells. J. Exp. Med. 175, 951–960 (1992).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Brentjens, R. J. et al. Safety and persistence of adoptively transferred autologous CD19-targeted T cells in patients with relapsed or chemotherapy refractory B-cell leukemias. Blood 118, 4817–4828 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Kalos, M. et al. T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia. Sci. Transl Med. 3, 95ra73 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Brentjens, R. J. et al. CD19-targeted T cells rapidly induce molecular remissions in adults with chemotherapy-refractory acute lymphoblastic leukemia. Sci. Transl Med. 5, 177ra138 (2013).

    Google Scholar 

  12. Mullard, A. FDA approves first CAR T therapy. Nat. Rev. Drug Discov. 16, 669–669 (2017).

    PubMed  Google Scholar 

  13. Whitlow, M. et al. An improved linker for single-chain Fv with reduced aggregation and enhanced proteolytic stability. Protein Eng. 6, 989–995 (1993).

    CAS  PubMed  Google Scholar 

  14. Lee, L. et al. An APRIL-based chimeric antigen receptor for dual targeting of BCMA and TACI in multiple myeloma. Blood 131, 746–758 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Rajabzadeh, A., Rahbarizadeh, F., Ahmadvand, D., Kabir Salmani, M. & Hamidieh, A. A. A VHH-based anti-MUC1 chimeric antigen receptor for specific retargeting of human primary T cells to MUC1-positive cancer cells. Cell J. 22, 502–513 (2021).

    PubMed  Google Scholar 

  16. Balakrishnan, A. et al. Multispecific targeting with synthetic ankyrin repeat motif chimeric antigen receptors. Clin. Cancer Res. 25, 7506–7516 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Brudno, J. N. et al. Safety and feasibility of anti-CD19 CAR T cells with fully human binding domains in patients with B-cell lymphoma. Nat. Med. 26, 270–280 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Bridgeman, J. et al. The optimal antigen response of chimeric antigen receptors harboring the CD3 transmembrane domain is dependent upon incorporation of the receptor into the endogenous TCR/CD3 complex. J. Immunol. 184, 6938–6949 (2010).

    CAS  PubMed  Google Scholar 

  19. Schmidts, A. et al. Rational design of a trimeric APRIL-based CAR-binding domain enables efficient targeting of multiple myeloma. Blood Adv. 3, 3248–3260 (2019).

    PubMed  PubMed Central  Google Scholar 

  20. 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).

    CAS  PubMed  Google Scholar 

  21. Imai, C. et al. Chimeric receptors with 4-1BB signaling capacity provoke potent cytotoxicity against acute lymphoblastic leukemia. Leukemia 18, 676–684 (2004).

    CAS  PubMed  Google Scholar 

  22. Pulè, M. A. et al. A chimeric T cell antigen receptor that augments cytokine release and supports clonal expansion of primary human T cells. Mol. Ther. 12, 933–941 (2005).

    PubMed  Google Scholar 

  23. Song, D.-G. et al. CD27 costimulation augments the survival and antitumor activity of redirected human T cells in vivo. Blood 119, 696–706 (2012).

    CAS  PubMed  Google Scholar 

  24. Guedan, S. et al. ICOS-based chimeric antigen receptors program bipolar TH17/TH1 cells. Blood 124, 1070–1080 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Feucht, J. et al. Calibration of CAR activation potential directs alternative T cell fates and therapeutic potency. Nat. Med. 25, 82–88 (2019).

    CAS  PubMed  Google Scholar 

  26. Yeku, O. O., Brentjens, R. J. & Armored, C. A. R. T-cells: utilizing cytokines and pro-inflammatory ligands to enhance CAR T-cell anti-tumour efficacy. Biochem. Soc. Trans. 44, 412–418 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Chmielewski, M. & Abken, H. CAR T cells transform to trucks: chimeric antigen receptor–redirected T cells engineered to deliver inducible IL-12 modulate the tumour stroma to combat cancer. Cancer Immunol. Immunother. 61, 1269–1277 (2012).

    CAS  PubMed  Google Scholar 

  28. Ramello, M. C. et al. An immunoproteomic approach to characterize the CAR interactome and signalosome. Sci. Signal. 12, eaap9777 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Singh, N. et al. Impaired death receptor signaling in leukemia causes antigen-independent resistance by inducing CAR T-cell dysfunction. Cancer Discov. 10, 552–567 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Dufva, O. et al. Integrated drug profiling and CRISPR screening identify essential pathways for CAR T-cell cytotoxicity. Blood 135, 597–609 (2020).

    PubMed  PubMed Central  Google Scholar 

  31. Benmebarek, M. R. et al. Killing mechanisms of chimeric antigen receptor (CAR) T cells. Int. J. Mol. Sci. 20, 1283 (2019).

    CAS  PubMed Central  Google Scholar 

  32. Guedan, S. et al. Single residue in CD28-costimulated CAR-T cells limits long-term persistence and antitumor durability. J. Clin. Invest. 130, 3087–3097 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Salter, A. I. et al. Phosphoproteomic analysis of chimeric antigen receptor signaling reveals kinetic and quantitative differences that affect cell function. Sci. Signal. 11, eaat6753 (2018). This paper shows that CD28-based and 4-1BB-based CAR T cells utilize similar signalling molecules upon activation. However, CD28-based CAR T cells have a much larger magnitude of phosphorylation that may contribute to activation-induced cell death and early exhaustion compared with the persistence of 4-1BB-based CAR T cells, which have a more memory-like phenotype.

    PubMed  PubMed Central  Google Scholar 

  34. Turtle, C. J. et al. Immunotherapy of non-Hodgkin’s lymphoma with a defined ratio of CD8+ and CD4+ CD19-specific chimeric antigen receptor-modified T cells. Sci. Transl Med. 8, 355ra116 (2016).

    PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  36. Levine, B. L. et al. Effects of CD28 costimulation on long-term proliferation of CD4+ T cells in the absence of exogenous feeder cells. J. Immunol. 159, 5921–5930 (1997).

    CAS  PubMed  Google Scholar 

  37. Maus, M. V. et al. Ex vivo expansion of polyclonal and antigen-specific cytotoxic T lymphocytes by artificial APCs expressing ligands for the T-cell receptor, CD28 and 4-1BB. Nat. Biotechnol. 20, 143–148 (2002).

    CAS  PubMed  Google Scholar 

  38. Rubinstein, M. P. et al. IL-7 and IL-15 differentially regulate CD8+ T-cell subsets during contraction of the immune response. Blood 112, 3704–3712 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Gong, W. et al. Comparison of IL-2 vs IL-7/IL-15 for the generation of NY-ESO-1-specific T cells. Cancer Immunol. Immunother. 68, 1195–1209 (2019).

    CAS  PubMed  Google Scholar 

  40. Alizadeh, D. et al. IL15 enhances CAR-T cell antitumor activity by reducing mTORC1 activity and preserving their stem cell memory phenotype. Cancer Immunol. Res. 7, 759–772 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Mitchell, R. S. et al. Retroviral DNA integration: ASLV, HIV, and MLV show distinct target site preferences. PLoS Biol. 2, E234 (2004).

    PubMed  PubMed Central  Google Scholar 

  42. Schröder, A. R. W. et al. HIV-1 integration in the human genome favors active genes and local hotspots. Cell 110, 521–529 (2002).

    PubMed  Google Scholar 

  43. Kebriaei, P. et al. Phase I trials using sleeping beauty to generate CD19-specific CAR T cells. J. Clin. Invest. 126, 3363–3376 (2016).

    PubMed  PubMed Central  Google Scholar 

  44. Benjamin, R. et al. 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 (2018).

    Google Scholar 

  45. Stadtmauer, E. A. et al. CRISPR-engineered T cells in patients with refractory cancer. Science 367, eaba7365 (2020).

    CAS  PubMed  Google Scholar 

  46. Fraietta, J. A. et al. Determinants of response and resistance to CD19 chimeric antigen receptor (CAR) T cell therapy of chronic lymphocytic leukemia. Nat. Med. 24, 563–571 (2018). This paper shows that CAR T cell products with a less exhausted phenotype have more favourable clinical outcomes.

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Fraietta, J. A. et al. Disruption of TET2 promotes the therapeutic efficacy of CD19-targeted T cells. Nature 558, 307–312 (2018). This paper shows that a single CAR T cell is capable of creating a long-lasting, durable antitumour response.

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Shifrut, E. et al. Genome-wide CRISPR screens in primary human T cells reveal key regulators of immune function. Cell 175, 1958–1971.e15 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Nobles, C. L. et al. CD19-targeting CAR T cell immunotherapy outcomes correlate with genomic modification by vector integration. J. Clin. Invest. 130, 673–685 (2020).

    CAS  PubMed  Google Scholar 

  50. van Bruggen, J. A. C. et al. Chronic lymphocytic leukemia cells impair mitochondrial fitness in CD8+ T cells and impede CAR T-cell efficacy. Blood 134, 44–58 (2019).

    PubMed  PubMed Central  Google Scholar 

  51. Garfall, A. L. et al. T-cell phenotypes associated with effective CAR T-cell therapy in postinduction vs relapsed multiple myeloma. Blood Adv. 3, 2812–2815 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Neelapu, S. S. et al. Axicabtagene ciloleucel CAR T-cell therapy in refractory large B-cell lymphoma. N. Engl. J. Med. 377, 2531–2544 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  54. van der Stegen, S. J. C., Hamieh, M. & Sadelain, M. The pharmacology of second-generation chimeric antigen receptors. Nat. Rev. Drug Discov. 14, 499–509 (2015).

    PubMed  PubMed Central  Google Scholar 

  55. Porter, D. L. et al. Chimeric antigen receptor T cells persist and induce sustained remissions in relapsed refractory chronic lymphocytic leukemia. Sci. Transl Med. 7, 303ra139 (2015).

    PubMed  PubMed Central  Google Scholar 

  56. Kawalekar, O. U. et al. Distinct signaling of coreceptors regulates specific metabolism pathways and impacts memory development in CAR T cells. Immunity 44, 712 (2016).

    CAS  PubMed  Google Scholar 

  57. Zhang, H. et al. 4-1BB is superior to CD28 costimulation for generating CD8+ cytotoxic lymphocytes for adoptive immunotherapy. J. Immunol. 179, 4910–4918 (2007).

    CAS  PubMed  Google Scholar 

  58. Philipson, B. I. et al. 4-1BB costimulation promotes CAR T cell survival through noncanonical NF-κB signaling. Sci. Signal. 13, eaay8248 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Li, G. et al. 4-1BB enhancement of CAR T function requires NF-κB and TRAFs. JCI Insight 3, e121322 (2018).

    PubMed Central  Google Scholar 

  60. Boroughs, A. C. et al. A distinct transcriptional program in human CAR T cells bearing the 4-1BB signaling domain revealed by scRNA-seq. Mol. Ther. 28, 2577–2592 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Li, W. et al. Chimeric antigen receptor designed to prevent ubiquitination and downregulation showed durable antitumor efficacy. Immunity 53, 456–470.e6 (2020).

    CAS  PubMed  Google Scholar 

  62. Boroughs, A. C. et al. Chimeric antigen receptor costimulation domains modulate human regulatory T cell function. JCI Insight 4, e126194 (2019).

    PubMed Central  Google Scholar 

  63. Guedan, S. et al. Enhancing CAR T cell persistence through ICOS and 4-1BB costimulation. JCI Insight 3, e96976 (2018).

    PubMed Central  Google Scholar 

  64. Youngblood, B., Davis, C. W. & Ahmed, R. Making memories that last a lifetime: heritable functions of self-renewing memory CD8 T cells. Int. Immunol. 22, 797–803 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Wherry, E. J. & Kurachi, M. Molecular and cellular insights into T cell exhaustion. Nat. Rev. Immunol. 15, 486–499 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Walker, A. J. et al. Tumor antigen and receptor densities regulate efficacy of a chimeric antigen receptor targeting anaplastic lymphoma kinase. Mol. Ther. 25, 2189–2201 (2017). This paper shows that antigen density on target cells plays a large role in CAR T cell efficacy.

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Singh, N. et al. Single chain variable fragment linker length regulates CAR biology and T cell efficacy. Blood 134, 247–247 (2019). This paper shows that linker length alone can affect CAR T cell efficacy.

    Google Scholar 

  68. Qin, H. et al. Preclinical development of bivalent chimeric antigen receptors targeting both CD19 and CD22. Mol. Ther. Oncolytics 11, 127–137 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Majzner, R. G. et al. Tuning the antigen density requirement for CAR T cell activity. Cancer Discov. 10, 702–723 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Stoiber, S. et al. Limitations in the design of chimeric antigen receptors for cancer therapy. Cells 8, 472 (2019).

    CAS  PubMed Central  Google Scholar 

  71. Alabanza, L. et al. Function of novel anti-CD19 chimeric antigen receptors with human variable regions is affected by hinge and transmembrane domains. Mol. Ther. 25, 2452–2465 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Hudecek, M. et al. Receptor affinity and extracellular domain modifications affect tumor recognition by ROR1-specific chimeric antigen receptor T cells. Clin. Cancer Res. 19, 3153–3164 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Hudecek, M. et al. The nonsignaling extracellular spacer domain of chimeric antigen receptors is decisive for in vivo antitumor activity. Cancer Immunol. Res. 3, 125–135 (2015).

    CAS  PubMed  Google Scholar 

  75. US National Library of Medicine. ClinicalTrials.gov https://ClinicalTrials.gov/show/NCT03620058 (2018).

  76. US National Library of Medicine. ClinicalTrials.gov https://ClinicalTrials.gov/show/NCT02650414 (2016).

  77. Guest, R. D. et al. The role of extracellular spacer regions in the optimal design of chimeric immune receptors: evaluation of four different scfvs and antigens. J. Immunother. 28, 203–211 (2005).

    CAS  PubMed  Google Scholar 

  78. James, S. E. et al. Antigen sensitivity of CD22-specific chimeric TCR is modulated by target epitope distance from the cell membrane. J. Immunol. 180, 7028–7038 (2008).

    CAS  PubMed  Google Scholar 

  79. Xu, Y. et al. A novel antibody–TCR (AbTCR) platform combines Fab-based antigen recognition with γ/δ-TCR signaling to facilitate T-cell cytotoxicity with low cytokine release. Cell Discov. 4, 62 (2018).

    PubMed  PubMed Central  Google Scholar 

  80. Wu, W. et al. Multiple signaling roles of CD3ε and its application in CAR-T cell therapy. Cell 182, 855–871.e23 (2020).

    CAS  PubMed  Google Scholar 

  81. Hartl, F. A. et al. Noncanonical binding of Lck to CD3ε promotes TCR signaling and CAR function. Nat. Immunol. 21, 902–913 (2020).

    CAS  PubMed  Google Scholar 

  82. Neelapu, S. S. et al. Chimeric antigen receptor T-cell therapy — assessment and management of toxicities. Nat. Rev. Clin. Oncol. 15, 47–62 (2018).

    CAS  PubMed  Google Scholar 

  83. Hay, K. A. et al. Kinetics and biomarkers of severe cytokine release syndrome after CD19 chimeric antigen receptor-modified T-cell therapy. Blood 130, 2295–2306 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  84. Gust, J., Taraseviciute, A. & Turtle, C. J. Neurotoxicity associated with CD19-targeted CAR-T cell therapies. CNS Drugs 32, 1091–1101 (2018).

    PubMed  PubMed Central  Google Scholar 

  85. Riddell, S. R. Adrenaline fuels a cytokine storm. Nature 564, 194–196 (2018).

    CAS  PubMed  Google Scholar 

  86. Staedtke, V. et al. Disruption of a self-amplifying catecholamine loop reduces cytokine release syndrome. Nature 564, 273–277 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  87. Gust, J. et al. Endothelial activation and blood–brain barrier disruption in neurotoxicity after adoptive immunotherapy with CD19 CAR-T cells. Cancer Discov. 7, 1404–1419 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  88. Mueller, K. T. et al. Clinical pharmacology of tisagenlecleucel in B-cell acute lymphoblastic leukemia. Clin. Cancer Res. 24, 6175–6184 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  89. Santomasso, B. D. et al. Clinical and biological correlates of neurotoxicity associated with CAR T-cell therapy in patients with B-cell acute lymphoblastic leukemia. Cancer Discov. 8, 958–971 (2018). This paper shows that neurotoxicity is correlated with levels of pro-inflammatory cytokines in the CSF.

    CAS  PubMed  PubMed Central  Google Scholar 

  90. Parker, K. R. et al. Single-cell analyses identify brain mural cells expressing CD19 as potential off-tumor targets for CAR-T immunotherapies. Cell 183, 126–142.e17 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Giavridis, T. et al. CAR T cell-induced cytokine release syndrome is mediated by macrophages and abated by IL-1 blockade. Nat. Med. 24, 731–738 (2018). This paper shows that interfering with IL-1 signalling can have a large affect, mitigating CRS and neurotoxicity.

    CAS  PubMed  PubMed Central  Google Scholar 

  92. Taraseviciute, A. et al. Chimeric antigen receptor T cell-mediated neurotoxicity in nonhuman primates. Cancer Discov. 8, 750–763 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  93. Pennell, C. A. et al. Human CD19-targeted mouse T cells induce B cell aplasia and toxicity in human CD19 transgenic mice. Mol. Ther. 26, 1423–1434 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  94. Norelli, M. et al. Monocyte-derived IL-1 and IL-6 are differentially required for cytokine-release syndrome and neurotoxicity due to CAR T cells. Nat. Med. 24, 739–748 (2018).

    CAS  PubMed  Google Scholar 

  95. Sterner, R. M. et al. GM-CSF inhibition reduces cytokine release syndrome and neuroinflammation but enhances CAR-T cell function in xenografts. Blood 133, 697–709 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  96. US National Library of Medicine. ClinicalTrials.gov https://ClinicalTrials.gov/show/NCT04150913 (2019).

  97. Mestermann, K. et al. The tyrosine kinase inhibitor dasatinib acts as a pharmacologic on/off switch for CAR T cells. Sci. Transl Med. 11, eaau5907 (2019).

    PubMed  PubMed Central  Google Scholar 

  98. Casucci, M. et al. Extracellular NGFR spacers allow efficient tracking and enrichment of fully functional CAR-T cells co-expressing a suicide gene. Front. Immunol. 9, 507 (2018).

    PubMed  PubMed Central  Google Scholar 

  99. Weber, E. W. et al. Pharmacologic control of CAR-T cell function using dasatinib. Blood Adv. 3, 711–717 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  100. Orlando, E. J. et al. Genetic mechanisms of target antigen loss in CAR19 therapy of acute lymphoblastic leukemia. Nat. Med. 24, 1504–1506 (2018). This paper shows that mutations and selective pressure can lead to loss of target antigen on tumours, leading to antigen-negative relapse.

    CAS  PubMed  Google Scholar 

  101. 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). This paper shows that tumour cell-based contamination during the manufacturing process can lead to relapse due to accidental transduction with a CAR, masking the cell surface antigen.

    CAS  PubMed  PubMed Central  Google Scholar 

  102. Hamieh, M. et al. CAR T cell trogocytosis and cooperative killing regulate tumour antigen escape. Nature 568, 112–116 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  103. Maude, S. L. et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N. Engl. J. Med. 371, 1507–1517 (2014).

    PubMed  PubMed Central  Google Scholar 

  104. Bagley, S. J., Desai, A. S., Linette, G. P., June, C. H. & O’Rourke, D. M. CAR T-cell therapy for glioblastoma: recent clinical advances and future challenges. Neuro-oncology 20, 1429–1438 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Zhang, W.-y et al. Treatment of CD20-directed chimeric antigen receptor-modified T cells in patients with relapsed or refractory B-cell non-Hodgkin lymphoma: an early phase IIa trial report. Signal. Transduct. Target. Ther. 1, 16002 (2016).

    PubMed  PubMed Central  Google Scholar 

  106. Fry, T. J. et al. CD22-targeted CAR T cells induce remission in B-ALL that is naive or resistant to CD19-targeted CAR immunotherapy. Nat. Med. 24, 20–28 (2018).

    CAS  PubMed  Google Scholar 

  107. Kuhn, N. F. et al. CD40 ligand-modified chimeric antigen receptor T cells enhance antitumor function by eliciting an endogenous antitumor response. Cancer Cell 35, 473–488.e6 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  108. Pont, M. J. et al. γ-Secretase inhibition increases efficacy of BCMA-specific chimeric antigen receptor T cells in multiple myeloma. Blood 134, 1585–1597 (2019).

    PubMed  PubMed Central  Google Scholar 

  109. Ramakrishna, S. et al. Modulation of target antigen density improves CAR T-cell functionality and persistence. Clin. Cancer Res. 25, 5329–5341 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  110. Liu, X. et al. Affinity-tuned ErbB2 or EGFR chimeric antigen receptor T cells exhibit an increased therapeutic index against tumors in mice. Cancer Res. 75, 3596–3607 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  111. Sharpe, A. H. & Pauken, K. E. The diverse functions of the PD1 inhibitory pathway. Nat. Rev. Immunol. 18, 153–167 (2018).

    CAS  PubMed  Google Scholar 

  112. Topalian, S. L., Drake, C. G. & Pardoll, D. M. Targeting the PD-1/B7-H1(PD-L1) pathway to activate anti-tumor immunity. Curr. Opin. Immunol. 24, 207–212 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  113. Topalian, S. L. et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N. Engl. J. Med. 366, 2443–2454 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  114. Cherkassky, L. et al. Human CAR T cells with cell-intrinsic PD-1 checkpoint blockade resist tumor-mediated inhibition. J. Clin. Invest. 126, 3130–3144 (2016). This paper shows that exhausted CAR T cells may be revived through checkpoint blockade.

    PubMed  PubMed Central  Google Scholar 

  115. Chong, E. A. et al. Sequential anti-CD19 directed chimeric antigen receptor modified T-cell therapy (CART19) and PD-1 blockade with pembrolizumab in patients with relapsed or refractory B-cell non-Hodgkin lymphomas. Blood 132, 4198 (2018).

    Google Scholar 

  116. Hirayama, A. V. et al. Efficacy and toxicity of JCAR014 in combination with durvalumab for the treatment of patients with relapsed/refractory aggressive B-cell non-Hodgkin lymphoma. Blood 132, 1680 (2018).

    Google Scholar 

  117. Jacobson, C. A. et al. End of phase 1 results from ZUMA-6: axicabtagene ciloleucel (axi-cel) in combination with atezolizumab for the treatment of patients with refractory diffuse large B cell lymphoma. Biol. Blood Marrow Transplant. 25, S173 (2019).

    Google Scholar 

  118. US National Library of Medicine. ClinicalTrials.gov https://ClinicalTrials.gov/show/NCT03310619 (2017).

  119. Chong, E. A. et al. PD-1 blockade modulates chimeric antigen receptor (CAR)-modified T cells: refueling the CAR. Blood 129, 1039–1041 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  120. Suarez, E. R. et al. Chimeric antigen receptor T cells secreting anti-PD-L1 antibodies more effectively regress renal cell carcinoma in a humanized mouse model. Oncotarget 7, 34341–34355 (2016).

    PubMed  PubMed Central  Google Scholar 

  121. Rafiq, S. et al. Targeted delivery of a PD-1-blocking scFv by CAR-T cells enhances anti-tumor efficacy in vivo. Nat. Biotechnol. 36, 847–856 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  122. Postow, M. A., Sidlow, R. & Hellmann, M. D. Immune-related adverse events associated with immune checkpoint blockade. N. Engl. J. Med. 378, 158–168 (2018).

    CAS  PubMed  Google Scholar 

  123. 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).

    PubMed  PubMed Central  Google Scholar 

  124. Shum, T. et al. Constitutive signaling from an engineered IL7 receptor promotes durable tumor elimination by tumor-redirected T cells. Cancer Discov. 7, 1238–1247 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  125. Lynn, R. C. et al. c-Jun overexpression in CAR T cells induces exhaustion resistance. Nature 576, 293–300 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  126. Adachi, K. et al. IL-7 and CCL19 expression in CAR-T cells improves immune cell infiltration and CAR-T cell survival in the tumor. Nat. Biotechnol. 36, 346–351 (2018).

    CAS  PubMed  Google Scholar 

  127. Yeku, O. O. et al. T cells enhance antitumor efficacy and overcome the tumor microenvironment. Sci. Rep. 7, 10541 (2017).

    PubMed  PubMed Central  Google Scholar 

  128. Avanzi, M. P. et al. Engineered tumor-targeted T cells mediate enhanced anti-tumor efficacy both directly and through activation of the endogenous immune system. Cell Rep. 23, 2130–2141 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  129. Ma, X. et al. Interleukin-23 engineering improves CAR T cell function in solid tumors. Nat. Biotechnol. 38, 448–459 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  130. Kloss, C. C. et al. Dominant-negative TGF-β receptor enhances PSMA-targeted human CAR T cell proliferation and augments prostate cancer eradication. Mol. Ther. 26, 1855–1866 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  131. Narayan, V. et al. A phase I clinical trial of PSMA-directed/TGFβ-insensitive CAR-T cells in metastatic castration-resistant prostate cancer. J. Clin. Oncol. 38, TPS269 (2020).

    Google Scholar 

  132. 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).

    PubMed Central  Google Scholar 

  133. O’Rourke, D. M. et al. A single dose of peripherally infused EGFRvIII-directed CAR T cells mediates antigen loss and induces adaptive resistance in patients with recurrent glioblastoma. Sci. Transl Med. 9, eaaa0984 (2017).

    PubMed  PubMed Central  Google Scholar 

  134. Choi, B. D. et al. CAR-T cells secreting BiTEs circumvent antigen escape without detectable toxicity. Nat. Biotechnol. 37, 1049–1058 (2019).

    CAS  PubMed  Google Scholar 

  135. Wing, A. et al. Improving CART-cell therapy of solid tumors with oncolytic virus-driven production of a bispecific T-cell engager. Cancer Immunol. Res. 6, 605–616 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  136. Ma, L. et al. Enhanced CAR-T cell activity against solid tumors by vaccine boosting through the chimeric receptor. Science 365, 162–168 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  137. Raje, N. et al. Anti-BCMA CAR T-cell therapy bb2121 in relapsed or refractory multiple myeloma. N. Engl. J. Med. 380, 1726–1737 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  138. Cohen, A. D. et al. B cell maturation antigen-specific CAR T cells are clinically active in multiple myeloma. J. Clin. Invest. 129, 2210–2221 (2019).

    PubMed  PubMed Central  Google Scholar 

  139. Ali, S. A. et al. T cells expressing an anti-B-cell maturation antigen chimeric antigen receptor cause remissions of multiple myeloma. Blood 128, 1688–1700 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  140. Ma, T., Shi, J. & Liu, H. Chimeric antigen receptor T cell targeting B cell maturation antigen immunotherapy is promising for multiple myeloma. Ann. Hematol. 98, 813–822 (2019).

    PubMed  PubMed Central  Google Scholar 

  141. US National Library of Medicine. ClinicalTrials.gov https://ClinicalTrials.gov/show/NCT03287804 (2017).

  142. Smith, E. L. et al. GPRC5D is a target for the immunotherapy of multiple myeloma with rationally designed CAR T cells. Sci. Transl Med. 11, eaau7746 (2019).

    PubMed  PubMed Central  Google Scholar 

  143. Gagelmann, N. et al. Development of CAR-T cell therapies for multiple myeloma. Leukemia 34, 2317–2332 (2020).

    CAS  PubMed  Google Scholar 

  144. US National Library of Medicine. ClinicalTrials.gov https://ClinicalTrials.gov/show/NCT02982941 (2016).

  145. Desantes, K. et al. A phase 1, open-label, dose escalation study of enoblituzumab (MGA271) in pediatric patients with B7-H3-expressing relapsed or refractory solid tumors. J. Clin. Oncol. 35, TPS2596 (2017).

    Google Scholar 

  146. US National Library of Medicine. ClinicalTrials.gov https://ClinicalTrials.gov/show/NCT02381314 (2015).

  147. Urba, W. et al. A phase I, open-label, dose escalation study of MGA271 in combination with ipilimumab in patients with B7-H3-expressing melanoma, squamous cell cancer of the head and neck or non-small cell lung cancer. J. Immunother. Cancer 3, P176 (2015).

    PubMed Central  Google Scholar 

  148. US National Library of Medicine. ClinicalTrials.gov https://ClinicalTrials.gov/show/NCT03406949 (2018).

  149. Shankar, S. et al. A phase 1, open label, dose escalation study of MGD009, a humanized B7-H3 × CD3 DART protein, in combination with MGA012, an anti-PD-1 antibody, in patients with relapsed or refractory B7-H3-expressing tumors. J. Clin. Oncol. 36, TPS2601 (2018).

    Google Scholar 

  150. US National Library of Medicine. ClinicalTrials.gov https://ClinicalTrials.gov/show/NCT02628535 (2015).

  151. Tolcher, A. W. et al. Phase 1, first-in-human, open label, dose escalation study of MGD009, a humanized B7-H3 × CD3 dual-affinity re-targeting (DART) protein in patients with B7-H3-expressing neoplasms or B7-H3 expressing tumor vasculature. J. Clin. Oncol. 34, TPS3105 (2016).

    Google Scholar 

  152. US National Library of Medicine. ClinicalTrials.gov https://ClinicalTrials.gov/show/NCT03729596 (2018).

  153. Scribner, J. A. et al. Preclinical development of MGC018, a duocarmycin-based antibody-drug conjugate targeting B7-H3 for solid cancer [abstract 820]. American Association for Cancer Research Annual Meeting, 2018. (American Association for Cancer Research, 2018).

  154. Majzner, R. G. et al. CAR T cells targeting B7-H3, a pan-cancer antigen, demonstrate potent preclinical activity against pediatric solid tumors and brain tumors. Clin. Cancer Res. 25, 2560–2574 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  155. He, Y. et al. Multiple cancer-specific antigens are targeted by a chimeric antigen receptor on a single cancer cell. JCI Insight 4, e135306 (2019).

    PubMed Central  Google Scholar 

  156. Morello, A., Sadelain, M. & Adusumilli, P. S. Mesothelin-targeted CARs: driving T cells to solid tumors. Cancer Discov. 6, 133–146 (2016).

    CAS  PubMed  Google Scholar 

  157. Adusumilli P. S. et al. A phase I clinical trial of malignant pleural disease treated with regionally delivered autologous mesothelin-targeted CAR-T cells [abstract CT036]. 2019 AACR Annual Meeting (American Association for Cancer Research, 2019).

  158. Haas, A. R. et al. Phase I study of lentiviral-transduced chimeric antigen receptor-modified T cells recognizing mesothelin in advanced solid cancers. Mol. Ther. 27, 1919–1929 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  159. US National Library of Medicine. ClinicalTrials.gov https://ClinicalTrials.gov/show/NCT03907852 (2019).

  160. Baeuerle, P. A. et al. Synthetic TRuC receptors engaging the complete T cell receptor for potent anti-tumor response. Nat. Commun. 10, 2087 (2019).

    PubMed  PubMed Central  Google Scholar 

  161. Brown, C. E. et al. Bioactivity and safety of IL13Rα2-redirected chimeric antigen receptor CD8+ T cells in patients with recurrent glioblastoma. Clin. Cancer Res. 21, 4062–4072 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  162. 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).

    PubMed  PubMed Central  Google Scholar 

  163. Wang, D.-r. et al. Chlorotoxin-directed CAR T cells for specific and effective targeting of glioblastoma. Sci. Transl Med. 12, eaaw2672 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  164. Richards, R. M., Sotillo, E. & Majzner, R. G. CAR T cell therapy for neuroblastoma. Front. Immunol. 9, 2380 (2018).

    PubMed  PubMed Central  Google Scholar 

  165. Mount, C. W. et al. Potent antitumor efficacy of anti-GD2 CAR T cells in H3-K27M+ diffuse midline gliomas. Nat. Med. 24, 572–579 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  166. US National Library of Medicine. ClinicalTrials.gov https://ClinicalTrials.gov/show/NCT03294954 (2017).

  167. 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).

    PubMed  Google Scholar 

  168. Bosse, K. R. et al. Identification of GPC2 as an oncoprotein and candidate immunotherapeutic target in high-risk neuroblastoma. Cancer Cell 32, 295–309.e12 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  169. Macdonald, G. Cell and Gene Therapy https://www.pmlive.com/pharma_intelligence/Cell_and_gene_therapy_1278537?SQ_DESIGN_NAME=2 (PMLiVE, 2019).

  170. US National Library of Medicine. ClinicalTrials.gov https://ClinicalTrials.gov/show/NCT00968760 (2009).

  171. US National Library of Medicine. ClinicalTrials.gov https://ClinicalTrials.gov/show/NCT01497184 (2011).

  172. US National Library of Medicine. ClinicalTrials.gov https://ClinicalTrials.gov/show/NCT03389035 (2018).

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

    CAS  PubMed  PubMed Central  Google Scholar 

  174. Barnett, B. E. et al. piggyBacTM-produced CAR-T cells exhibit stem-cell memory phenotype. Blood 128, 2167 (2016).

    Google Scholar 

  175. Poseida Therapeutics. Pipeline https://poseida.com/pipeline/ (2020).

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

    CAS  PubMed  PubMed Central  Google Scholar 

  177. Schober, K. et al. Orthotopic replacement of T-cell receptor α- and β-chains with preservation of near-physiological T-cell function. Nat. Biomed. Eng. 3, 974–984 (2019).

    PubMed  Google Scholar 

  178. Silva, D. A. et al. De novo design of potent and selective mimics of IL-2 and IL-15. Nature 565, 186–191 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  179. US National Library of Medicine. ClinicalTrials.gov https://ClinicalTrials.gov/show/NCT01318317 (2011).

  180. US National Library of Medicine. ClinicalTrials.gov https://ClinicalTrials.gov/show/NCT01815749 (2013).

  181. Wang, X. et al. Phase 1 studies of central memory-derived CD19 CAR T-cell therapy following autologous HSCT in patients with B-cell NHL. Blood 127, 2980–2990 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  182. Murray, C., Pao, E., Jann, A., Park, D. E. & Di Carlo, D. Continuous and quantitative purification of T-cell subsets for cell therapy manufacturing using magnetic ratcheting cytometry. SLAS Technol. 23, 326–337 (2018).

    CAS  PubMed  Google Scholar 

  183. Bailey, S. R. et al. Human CD26high T cells elicit tumor immunity against multiple malignancies via enhanced migration and persistence. Nat. Commun. 8, 1961 (2017).

    PubMed  PubMed Central  Google Scholar 

  184. Deniger, D. C. et al. Bispecific T-cells expressing polyclonal repertoire of endogenous γδ T-cell receptors and introduced CD19-specific chimeric antigen receptor. Mol. Ther. 21, 638–647 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  185. Capsomidis, A. et al. Chimeric antigen receptor-engineered human γδ T cells: enhanced cytotoxicity with retention of cross presentation. Mol. Ther. 26, 354–365 (2018).

    CAS  PubMed  Google Scholar 

  186. Gentles, A. J. et al. The prognostic landscape of genes and infiltrating immune cells across human cancers. Nat. Med. 21, 938–945 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  187. Cho, J. H., Collins, J. J. & Wong, W. W. Universal chimeric antigen receptors for multiplexed and logical control of T cell responses. Cell 173, 1426–1438.e11 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  188. Lohmueller, J. J., Ham, J. D., Kvorjak, M. & Finn, O. J. mSA2 affinity-enhanced biotin-binding CAR T cells for universal tumor targeting. OncoImmunology 7, e1368604 (2018).

    Google Scholar 

  189. Ma, J. S. Y. et al. Versatile strategy for controlling the specificity and activity of engineered T cells. Proc. Natl Acad. Sci. USA 113, E450–E458 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  190. Cartellieri, M. et al. Switching CAR T cells on and off: a novel modular platform for retargeting of T cells to AML blasts. Blood Cancer J. 6, e458 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  191. 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).

    CAS  PubMed  Google Scholar 

  192. Qasim, W. et al. Molecular remission of infant B-ALL after infusion of universal TALEN gene-edited CAR T cells. Sci. Transl Med. 9, eaaj2013 (2017).

    PubMed  Google Scholar 

  193. US National Library of Medicine. ClinicalTrials.gov https://ClinicalTrials.gov/show/NCT02746952 (2016).

  194. US National Library of Medicine. ClinicalTrials.gov https://ClinicalTrials.gov/show/NCT02808442 (2016).

  195. Ren, J. et al. Multiplex genome editing to generate universal CAR T cells resistant to PD1 inhibition. Clin. Cancer Res. 23, 2255–2266 (2017).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  197. Liu, E. et al. Use of CAR-transduced natural killer cells in CD19-positive lymphoid tumors. N. Engl. J. Med. 382, 545–553 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  198. Daher, M. et al. Targeting a cytokine checkpoint enhances the fitness of armored cord blood CAR-NK cells. Blood https://doi.org/10.1182/blood.2020007748 (2020).

    Article  PubMed  Google Scholar 

  199. US National Library of Medicine. ClinicalTrials.gov https://ClinicalTrials.gov/show/NCT02435849 (2015).

  200. Locke, F. L. et al. Long-term safety and activity of axicabtagene ciloleucel in refractory large B-cell lymphoma (ZUMA-1): a single-arm, multicentre, phase 1–2 trial. Lancet Oncol. 20, 31–42 (2019).

    CAS  PubMed  Google Scholar 

  201. US National Library of Medicine. ClinicalTrials.gov https://ClinicalTrials.gov/show/NCT02348216 (2015).

  202. Schuster, S. J. et al. Chimeric antigen receptor T cells in refractory B-cell lymphomas. N. Engl. J. Med. 377, 2545–2554 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  203. US National Library of Medicine. ClinicalTrials.gov https://ClinicalTrials.gov/show/NCT02030834 (2014).

  204. Wang, M. et al. KTE-X19 CAR T-cell therapy in relapsed or refractory mantle-cell lymphoma. N. Engl. J. Med. 382, 1331–1342 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  205. US National Library of Medicine. ClinicalTrials.gov https://ClinicalTrials.gov/show/NCT02601313 (2015).

  206. Park, J. H. et al. Long-term follow-up of CD19 CAR therapy in acute lymphoblastic leukemia. N. Engl. J. Med. 378, 449–459 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  207. US National Library of Medicine. ClinicalTrials.gov https://ClinicalTrials.gov/show/NCT01044069 (2010).

  208. US National Library of Medicine. ClinicalTrials.gov https://ClinicalTrials.gov/show/NCT02315612 (2014).

  209. US National Library of Medicine. ClinicalTrials.gov https://ClinicalTrials.gov/show/NCT02658929 (2016).

  210. US National Library of Medicine. ClinicalTrials.gov https://ClinicalTrials.gov/show/NCT02546167 (2015).

  211. Iliopoulou, E. G. et al. A phase I trial of adoptive transfer of allogeneic natural killer cells in patients with advanced non-small cell lung cancer. Cancer Immunol. Immunother. 59, 1781–1789 (2010).

    PubMed  Google Scholar 

  212. Mehta, R. S. & Rezvani, K. Chimeric antigen receptor expressing natural killer cells for the immunotherapy of cancer. Front. Immunol. 9, 283 (2018).

    PubMed  PubMed Central  Google Scholar 

  213. Maluski, M. et al. Chimeric antigen receptor-induced BCL11B suppression propagates NK-like cell development. J. Clin. Invest. 129, 5108–5122 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  215. Noyan, F. et al. Prevention of allograft rejection by use of regulatory T cells with an MHC-specific chimeric antigen receptor. Am. J. Transpl. 17, 917–930 (2017).

    CAS  Google Scholar 

  216. Boardman, D. A. et al. Expression of a chimeric antigen receptor specific for donor HLA class I enhances the potency of human regulatory T cells in preventing human skin transplant rejection. Am. J. Transpl. 17, 931–943 (2017).

    CAS  Google Scholar 

  217. Blat, D., Zigmond, E., Alteber, Z., Waks, T. & Eshhar, Z. Suppression of murine colitis and its associated cancer by carcinoembryonic antigen-specific regulatory T cells. Mol. Ther. 22, 1018–1028 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  218. Fritsche, E., Volk, H.-D., Reinke, P. & Abou-El-Enein, M. Toward an optimized process for clinical manufacturing of CAR-Treg cell therapy. Trends Biotechnol. 38, 1099–1112 (2020).

    PubMed  Google Scholar 

  219. Dawson, N. A. J. et al. Systematic testing and specificity mapping of alloantigen-specific chimeric antigen receptors in regulatory T cells. JCI Insight 4, e123672 (2019).

    PubMed Central  Google Scholar 

  220. European Medicines Agency EU Clinical Trials Register https://www.clinicaltrialsregister.eu/ctr-search/trial/2019-001730-34/NL (2019).

Download references

Acknowledgements

M.V.M. is funded by the National Institutes of Health (NIH) (R01 CA238268, R01 CA252940), the Leukemia and Lymphoma Society, Stand Up to Cancer and the Damon Runyon Cancer Research Foundation. R.C.L. has been funded by NIH T32 GM007306 and is currently funded by NIH T32 AI007529.

Author information

Authors and Affiliations

Authors

Contributions

M.V.M. and R.C.L. researched the data for the article and selected the content, and otherwise contributed equally to writing the article and to review and/or editing of the manuscript before submission.

Corresponding author

Correspondence to Marcela V. Maus.

Ethics declarations

Competing interests

M.V.M. and R.C.L. have intellectual property on certain chimeric antigen receptor (CAR) T cells and antibodies (not yet licensed). M.V.M. receives consulting income from several industry sponsors that market CAR T cell therapies, serves on several scientific advisory boards and has equity in TCR2 and Century Therapeutics.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Glossary

Camelid antibodies

Antibodies generated from Camelidae mammals, which have two identical heavy chains and, compared with typical antibodies, are much smaller (15 kDa compared with 150 kDa) and lack a light chain.

Graft-versus-host disease

(GvHD). A condition that can occur after allogeneic transplant owing to donor cells recognizing the host as foreign, resulting in donor cell attack of the host body.

Leukapheresis

A procedure in which white blood cells are separated from the blood and the remaining cells are returned to the circulation.

Artificial antigen-presenting cells

(Artificial APCs). Synthetic versions of APCs that activate immune cells; in the context of chimeric antigen receptor (CAR) T cells, artificial APCs are engineered with T cell receptor (TCR) stimulation and co-stimulatory molecules to expand T cells ex vivo.

Transcription activator-like effector nucleases

(TALENs). DNA-binding domains fused to non-specific DNA-cleaving nucleases to target a specific sequence for gene alteration.

Hypomorphic mutation

An altered gene resulting in lower expression and/or activity of the gene product.

Tonic signalling

Ligand-independent constitutive signalling of a chimeric antigen receptor (CAR).

Activation-induced cell death

Programmed cell death caused by repeated stimulation of T cells that serves as a negative regulator of activation.

Maximum tolerated dose

The highest dose of treatment that does not cause intolerable side effects.

Suicide switches

Genetically encoded molecules included in a chimeric antigen receptor (CAR) vector that can be targeted to induce CAR T cell death.

Trogocytosis

A process where lymphocytes extract ligands from antigen-presenting cells and express them on their own surface.

Bispecific T cell engager

(BiTE). An artificial bispecific antibody made up of two single-chain variable fragments (scFvs) — one that recognizes a specific antigen and the other that binds CD3 on T cells, eliciting an activation response.

Amphiphilic

A description of a molecule containing both hydrophobic and hydrophilic regions.

Transposons

Genetic segments that can be translocated in the genome from one location to another by a DNA transposase.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

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). https://doi.org/10.1038/s41568-020-00323-z

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41568-020-00323-z

This article is cited by

Search

Quick links

Nature Briefing

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

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