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:

‘Off-the-shelf’ allogeneic CAR T cells: development and challenges

Abstract

Autologous chimeric antigen receptor (CAR) T cells have changed the therapeutic landscape in haematological malignancies. Nevertheless, the use of allogeneic CAR T cells from donors has many potential advantages over autologous approaches, such as the immediate availability of cryopreserved batches for patient treatment, possible standardization of the CAR-T cell product, time for multiple cell modifications, redosing or combination of CAR T cells directed against different targets, and decreased cost using an industrialized process. However, allogeneic CAR T cells may cause life-threatening graft-versus-host disease and may be rapidly eliminated by the host immune system. The development of next-generation allogeneic CAR T cells to address these issues is an active area of research. In this Review, we analyse the different sources of T cells for optimal allogeneic CAR-T cell therapy and describe the different technological approaches, mainly based on gene editing, to produce allogeneic CAR T cells with limited potential for graft-versus-host disease. These improved allogeneic CAR-T cell products will pave the way for further breakthroughs in the treatment of cancer.

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: Manufacturing of allogeneic CAR T cells.
Fig. 2: Persistence of CAR T cells and tumour evolution.
Fig. 3: Examples of gene editing strategies to optimize CAR-T cell functions.

Similar content being viewed by others

References

  1. Carpenito, C. et al. Control of large, established tumor xenografts with genetically retargeted human T cells containing CD28 and CD137 domains. Proc. Natl Acad. Sci. USA 106, 3360–3365 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  2. June, C. H., O’Connor, R. S., Kawalekar, O. U., Ghassemi, S. & Milone, M. C. CAR T cell immunotherapy for human cancer. Science 359, 1361–1365 (2018).

    Article  CAS  PubMed  Google Scholar 

  3. Maude, S. L. et al. Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. N. Engl. J. Med. 378, 439–448 (2018). This article reports the results from a phase II study that showed an overall response rate of 81% with the CD19 CAR-T cell therapy tisagenlecleucel in paediatric and young adult ALL.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. 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). This article reports the results from a phase II study that showed an objective response rate of 82% with the CD19 CAR-T cell therapy axicabtagene ciloleucel in refractory large B cell lymphoma.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Köhl, U., Arsenieva, S., Holzinger, A. & Abken, H. CAR T cells in trials: recent achievements and challenges that remain in the production of modified T cells for clinical applications. Hum. Gene Ther. 29, 559–568 (2018).

    Article  CAS  PubMed  Google Scholar 

  6. Thommen, D. S. & Schumacher, T. N. T cell dysfunction in cancer. Cancer Cell 33, 547–562 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Salmikangas, P., Kinsella, N. & Chamberlain, P. Chimeric antigen receptor T-cells (CAR T-cells) for cancer immunotherapy - moving target for industry? Pharm. Res. 35, 152 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Lin, J. K. et al. Cost effectiveness of chimeric antigen receptor T-cell therapy in multiply relapsed or refractory adult large B-cell lymphoma. J. Clin. Oncol. 37, 2105–2119 (2019).

    Article  CAS  PubMed  Google Scholar 

  9. Eapen, M. et al. Effect of graft source on unrelated donor haemopoietic stem-cell transplantation in adults with acute leukaemia: a retrospective analysis. Lancet Oncol. 11, 653–660 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  10. Kwoczek, J. et al. Cord blood-derived T cells allow the generation of a more naïve tumor-reactive cytotoxic T-cell phenotype. Transfusion 58, 88–99 (2018).

    Article  CAS  PubMed  Google Scholar 

  11. Kadereit, S. et al. Reduced NFAT1 protein expression in human umbilical cord blood T lymphocytes. Blood 94, 3101–3107 (1999).

    Article  CAS  PubMed  Google Scholar 

  12. Kang, L. et al. Characterization and ex vivo expansion of human placenta-derived natural killer cells for cancer immunotherapy. Front. Immunol. 4, 101 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Juch, H., Blaschitz, A., Dohr, G. & Hutter, H. HLA class I expression in the human placenta. Wien. Med. Wochenschr. 162, 196–200 (2012).

    Article  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Butler, C. L., Valenzuela, N. M., Thomas, K. A. & Reed, E. F. Not all antibodies are created equal: factors that influence antibody mediated rejection. J. Immunol. Res. 2017, 7903471 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Ciurea, S. O. et al. The European Society for Blood and Marrow Transplantation (EBMT) consensus guidelines for the detection and treatment of donor-specific anti-HLA antibodies (DSA) in haploidentical hematopoietic cell transplantation. Bone Marrow Transpl. 53, 521–534 (2018).

    Article  CAS  Google Scholar 

  18. Frame, J. N. et al. T cell depletion of human bone marrow. Comparison of campath-1 plus complement, anti-T cell ricin A chain immunotoxin, and soybean agglutinin alone or in combination with sheep erythrocytes or immunomagnetic beads. Transplantation 47, 984–988 (1989).

    Article  CAS  PubMed  Google Scholar 

  19. Champlin, R. E. et al. T-cell depletion of bone marrow transplants for leukemia from donors other than HLA-identical siblings: advantage of T-cell antibodies with narrow specificities. Blood 95, 3996–4003 (2000).

    CAS  PubMed  Google Scholar 

  20. Prentice, H. G. OKT3 incubation of donor marrow for prophylaxis of acute graft-vs.-host disease (GvHD) in allogeneic bone marrow transplantation. J. Clin. Immunol. 2, 148S–153S (1982).

    CAS  PubMed  Google Scholar 

  21. Rådestad, E. et al. Alpha/beta T-cell depleted grafts as an immunological booster to treat graft failure after hematopoietic stem cell transplantation with HLA-matched related and unrelated donors. J. Immunol. Res. 2014, 578741 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Abdelhakim, H., Abdel-Azim, H. & Saad, A. Role of αβ T cell depletion in prevention of graft versus host disease. Biomedicines 5, 35 (2017).

    Article  CAS  PubMed Central  Google Scholar 

  23. Felix, N. J. & Allen, P. M. Specificity of T-cell alloreactivity. Nat. Rev. Immunol. 7, 942–953 (2007).

    Article  CAS  PubMed  Google Scholar 

  24. Zeiser, R. & Blazar, B. R. Acute graft-versus-host disease - biologic process, prevention, and therapy. N. Engl. J. Med. 377, 2167–2179 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Baker, M. B., Altman, N. H., Podack, E. R. & Levy, R. B. The role of cell-mediated cytotoxicity in acute GVHD after MHC-matched allogeneic bone marrow transplantation in mice. J. Exp. Med. 183, 2645–2656 (1996).

    Article  CAS  PubMed  Google Scholar 

  26. Brudno, J. N. et al. Allogeneic T cells that express an anti-CD19 chimeric antigen receptor induce remissions of B-cell malignancies that progress after allogeneic hematopoietic stem-cell transplantation without causing graft-versus-host disease. J. Clin. Oncol. 34, 1112–1121 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Kochenderfer, J. N. et al. Donor-derived CD19-targeted T cells cause regression of malignancy persisting after allogeneic hematopoietic stem cell transplantation. Blood 122, 4129–4139 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Cruz, C. R. Y. et al. Infusion of donor-derived CD19-redirected virus-specific T cells for B-cell malignancies relapsed after allogeneic stem cell transplant: a phase 1 study. Blood 122, 2965–2973 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Leen, A. M. et al. Multicenter study of banked third-party virus-specific T cells to treat severe viral infections after hematopoietic stem cell transplantation. Blood 121, 5113–5123 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Melenhorst, J. J. et al. Allogeneic virus-specific T cells with HLA alloreactivity do not produce GVHD in human subjects. Blood 116, 4700–4702 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Fanning, S. L. et al. Unraveling graft-versus-host disease and graft-versus-leukemia responses using TCR Vβ spectratype analysis in a murine bone marrow transplantation model. J. Immunol. 190, 447–457 (2013).

    Article  CAS  PubMed  Google Scholar 

  32. Fuji, S., Kapp, M. & Einsele, H. Alloreactivity of virus-specific T cells: possible implication of graft-versus-host disease and graft-versus-leukemia effects. Front. Immunol. 4, 330 (2013).

    PubMed  PubMed Central  Google Scholar 

  33. O’Reilly, R. J., Prockop, S., Hasan, A. N., Koehne, G. & Doubrovina, E. Virus-specific T-cell banks for ‘off the shelf’ adoptive therapy of refractory infections. Bone Marrow Transpl. 51, 1163–1172 (2016).

    Article  CAS  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  35. Han, J. et al. CAR-engineered NK cells targeting wild-type EGFR and EGFRvIII enhance killing of glioblastoma and patient-derived glioblastoma stem cells. Sci. Rep. 5, 11483 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Becker, P. S. A. et al. Selection and expansion of natural killer cells for NK cell-based immunotherapy. Cancer Immunol. Immunother. 65, 477–484 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Tonn, T. et al. Treatment of patients with advanced cancer with the natural killer cell line NK-92. Cytotherapy 15, 1563–1570 (2013).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Exley, M. et al. CD1d structure and regulation on human thymocytes, peripheral blood T cells, B cells and monocytes. Immunology 100, 37–47 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Nickoloff, B. J., Wrone-Smith, T., Bonish, B. & Porcelli, S. A. Response of murine and normal human skin to injection of allogeneic blood-derived psoriatic immunocytes: detection of T cells expressing receptors typically present on natural killer cells, including CD94, CD158, and CD161. Arch. Dermatol. 135, 546–552 (1999).

    CAS  PubMed  Google Scholar 

  42. Chaidos, A. et al. Graft invariant natural killer T-cell dose predicts risk of acute graft-versus-host disease in allogeneic hematopoietic stem cell transplantation. Blood 119, 5030–5036 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Leveson-Gower, D. B. et al. Low doses of natural killer T cells provide protection from acute graft-versus-host disease via an IL-4-dependent mechanism. Blood 117, 3220–3229 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Rubio, M.-T. et al. Pre-transplant donor CD4− invariant NKT cell expansion capacity predicts the occurrence of acute graft-versus-host disease. Leukemia 31, 903–912 (2017).

    Article  CAS  PubMed  Google Scholar 

  45. Schneidawind, D. et al. CD4+ invariant natural killer T cells protect from murine GVHD lethality through expansion of donor CD4+CD25+FoxP3+ regulatory T cells. Blood 124, 3320–3328 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Rotolo, A. et al. Enhanced anti-lymphoma activity of CAR19-iNKT cells underpinned by dual CD19 and CD1d targeting. Cancer Cell 34, 596–610.e11 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Kato, Y., Tanaka, Y., Miyagawa, F., Yamashita, S. & Minato, N. Targeting of tumor cells for human gammadelta T cells by nonpeptide antigens. J. Immunol. 167, 5092–5098 (2001).

    Article  CAS  PubMed  Google Scholar 

  48. Nagamine, I., Yamaguchi, Y., Ohara, M., Ikeda, T. & Okada, M. Induction of gamma delta T cells using zoledronate plus interleukin-2 in patients with metastatic cancer. Hiroshima J. Med. Sci. 58, 37–44 (2009).

    CAS  PubMed  Google Scholar 

  49. Thompson, K. et al. Activation of γδ T cells by bisphosphonates. Adv. Exp. Med. Biol. 658, 11–20 (2010).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  51. Choulika, A., Perrin, A., Dujon, B. & Nicolas, J. F. Induction of homologous recombination in mammalian chromosomes by using the I-SceI system of Saccharomyces cerevisiae. Mol. Cell Biol. 15, 1968–1973 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Rouet, P., Smih, F. & Jasin, M. Introduction of double-strand breaks into the genome of mouse cells by expression of a rare-cutting endonuclease. Mol. Cell Biol. 14, 8096–8106 (1994).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Mehta, A. & Haber, J. E. Sources of DNA double-strand breaks and models of recombinational DNA repair. Cold Spring Harb. Perspect. Biol. 6, a016428 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Chiruvella, K. K., Liang, Z. & Wilson, T. E. Repair of double-strand breaks by end joining. Cold Spring Harb. Perspect. Biol. 5, a012757 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Torikai, H. et al. A foundation for universal T-cell based immunotherapy: T cells engineered to express a CD19-specific chimeric-antigen-receptor and eliminate expression of endogenous TCR. Blood 119, 5697–5705 (2012). This is a seminal study describing the development of allogeneic universal CAR T cells based on the elimination of the TCR by gene editing.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Bridgeman, J. S. et al. The optimal antigen response of chimeric antigen receptors harboring the CD3zeta transmembrane domain is dependent upon incorporation of the receptor into the endogenous TCR/CD3 complex. J. Immunol. 184, 6938–6949 (2010).

    Article  CAS  PubMed  Google Scholar 

  57. Poirot, L. et al. Multiplex genome-edited T-cell manufacturing platform for “off-the-shelf” adoptive T-cell immunotherapies. Cancer Res. 75, 3853–3864 (2015). This article demonstrates that allogeneic CAR T cells can be rendered resistant to a lymphodepletion regimen containing alemtuzumab by CD52 disruption.

    Article  CAS  PubMed  Google Scholar 

  58. Osborn, M. J. et al. Evaluation of TCR gene editing achieved by TALENs, CRISPR/Cas9, and megaTAL nucleases. Mol. Ther. 24, 570–581 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. MacLeod, D. T. et al. Integration of a CD19 CAR into the TCR alpha chain locus streamlines production of allogeneic gene-edited CAR T cells. Mol. Ther. 25, 949–961 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Ren, J. et al. A versatile system for rapid multiplex genome-edited CAR T cell generation. Oncotarget 8, 17002–17011 (2017).

    PubMed  PubMed Central  Google Scholar 

  61. Brunet, E. & Jasin, M. Induction of chromosomal translocations with CRISPR-Cas9 and other nucleases: understanding the repair mechanisms that give rise to translocations. Adv. Exp. Med. Biol. 1044, 15–25 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Rasaiyaah, J., Georgiadis, C., Preece, R., Mock, U. & Qasim, W. TCRαβ/CD3 disruption enables CD3-specific antileukemic T cell immunotherapy. JCI Insight 3, (2018).

  64. Eyquem, J. et al. Targeting a CAR to the TRAC locus with CRISPR/Cas9 enhances tumour rejection. Nature 543, 113–117 (2017). This study shows the advantages of targeting the CAR directly to the TRAC locus.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. 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). This article reports the first clinical results obtained with allogeneic gene-edited CAR T cells.

    Article  PubMed  Google Scholar 

  67. Jain, N. et al. UCART19, an allogeneic anti-CD19 CAR T-cell product, in high risk adult patients with CD19+ relapsed/refractory B-cell acute lymphoblastic leukemia: preliminary results of phase I CALM study (Poster). EHA https://learningcenter.ehaweb.org/eha/2018/stockholm/214674/reuben.benjamin.ucart19.an.allogeneic.anti-cd19.car.t-cell.product.in.high.html?f=media=3*c_id=214674*listing=3*browseby=8 (2018).

  68. Dudley, M. E. et al. Cancer regression and autoimmunity in patients after clonal repopulation with antitumor lymphocytes. Science 298, 850–854 (2002). This seminal study shows that a lymphodepleting chemotherapy allows efficient expansion of the administered T cells in patients, which is associated with clinical efficacy.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Tchao, N. K. & Turka, L. A. Lymphodepletion and homeostatic proliferation: implications for transplantation. Am. J. Transpl. 12, 1079–1090 (2012).

    Article  CAS  Google Scholar 

  70. Sandau, M. M., Winstead, C. J. & Jameson, S. C. IL-15 is required for sustained lymphopenia-driven proliferation and accumulation of CD8 T cells. J. Immunol. 179, 120–125 (2007).

    Article  CAS  PubMed  Google Scholar 

  71. Fry, T. J. et al. A potential role for interleukin-7 in T-cell homeostasis. Blood 97, 2983–2990 (2001).

    Article  CAS  PubMed  Google Scholar 

  72. Muranski, P. et al. Increased intensity lymphodepletion and adoptive immunotherapy–how far can we go? Nat. Clin. Pract. Oncol. 3, 668–681 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Maus, M. V. & June, C. H. Making better chimeric antigen receptors for adoptive T-cell therapy. Clin. Cancer Res. 22, 1875–1884 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Kochenderfer, J. N. & Rosenberg, S. A. Treating B-cell cancer with T cells expressing anti-CD19 chimeric antigen receptors. Nat. Rev. Clin. Oncol. 10, 267–276 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Yao, X. et al. Levels of peripheral CD4+FoxP3+ regulatory T cells are negatively associated with clinical response to adoptive immunotherapy of human cancer. Blood 119, 5688–5696 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Rapoport, A. P. et al. Restoration of immunity in lymphopenic individuals with cancer by vaccination and adoptive T-cell transfer. Nat. Med. 11, 1230–1237 (2005).

    Article  CAS  PubMed  Google Scholar 

  77. Rapoport, A. P. et al. Rapid immune recovery and graft-versus-host disease-like engraftment syndrome following adoptive transfer of costimulated autologous T cells. Clin. Cancer Res. 15, 4499–4507 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Lowe, K. L. et al. Fludarabine and neurotoxicity in engineered T-cell therapy. Gene Ther. 25, 176–191 (2018).

    Article  CAS  PubMed  Google Scholar 

  79. 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 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Finney, O. C. et al. CD19 CAR T cell product and disease attributes predict leukemia remission durability. J. Clin. Invest. 129, 2123–2132 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  82. Majzner, R. G. & Mackall, C. L. Clinical lessons learned from the first leg of the CAR T cell journey. Nat. Med. 25, 1341–1355 (2019).

    Article  CAS  PubMed  Google Scholar 

  83. Yang, Y. et al. TCR engagement negatively affects CD8 but not CD4 CAR T cell expansion and leukemic clearance. Sci. Transl Med. 9, eaag1209 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

  86. Lytle, N. K., Barber, A. G. & Reya, T. Stem cell fate in cancer growth, progression and therapy resistance. Nat. Rev. Cancer 18, 669–680 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Opelz, G. & Döhler, B. Effect of human leukocyte antigen compatibility on kidney graft survival: comparative analysis of two decades. Transplantation 84, 137–143 (2007).

    Article  CAS  PubMed  Google Scholar 

  88. Johnson, R. J. et al. Factors influencing outcome after deceased heart beating donor kidney transplantation in the united kingdom: an evidence base for a new national kidney allocation policy. Transplantation 89, 379–386 (2010).

    Article  PubMed  Google Scholar 

  89. Kurtzberg, J. et al. Results of the Cord Blood Transplantation Study (COBLT): clinical outcomes of unrelated donor umbilical cord blood transplantation in pediatric patients with hematologic malignancies. Blood 112, 4318–4327 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Taylor, C. J., Peacock, S., Chaudhry, A. N., Bradley, J. A. & Bolton, E. M. Generating an iPSC bank for HLA-matched tissue transplantation based on known donor and recipient HLA types. Cell Stem Cell 11, 147–152 (2012).

    Article  CAS  PubMed  Google Scholar 

  91. Valton, J. et al. A multidrug-resistant engineered CAR T cell for allogeneic combination immunotherapy. Mol. Ther. 23, 1507–1518 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Wang, D., Quan, Y., Yan, Q., Morales, J. E. & Wetsel, R. A. Targeted disruption of the β2-microglobulin gene minimizes the immunogenicity of human embryonic stem cells. Stem Cell Transl. Med. 4, 1234–1245 (2015). Demonstration of the elimination of MHC class I molecule expression by disruption of the gene encoding β 2-microglobulin.

    Article  CAS  Google Scholar 

  93. Mancusi, A. et al. Haploidentical hematopoietic transplantation from KIR ligand-mismatched donors with activating KIRs reduces nonrelapse mortality. Blood 125, 3173–3182 (2015).

    Article  CAS  PubMed  Google Scholar 

  94. Ichise, H. et al. NK cell alloreactivity against KIR-ligand-mismatched HLA-haploidentical tissue derived from HLA haplotype-homozygous iPSCs. Stem Cell Rep. 9, 853–867 (2017).

    Article  CAS  Google Scholar 

  95. Baier, C. et al. Natural killer cells modulation in hematological malignancies. Front. Immunol. 4, 459 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Costello, R. T. et al. Defective expression and function of natural killer cell-triggering receptors in patients with acute myeloid leukemia. Blood 99, 3661–3667 (2002).

    Article  CAS  PubMed  Google Scholar 

  97. Fauriat, C. et al. Deficient expression of NCR in NK cells from acute myeloid leukemia: Evolution during leukemia treatment and impact of leukemia cells in NCRdull phenotype induction. Blood 109, 323–330 (2007).

    Article  CAS  PubMed  Google Scholar 

  98. Gornalusse, G. G. et al. HLA-E-expressing pluripotent stem cells escape allogeneic responses and lysis by NK cells. Nat. Biotechnol. 35, 765–772 (2017). This study shows that ‘missing-self’ response can be prevented by expression of HLA-E molecules.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Jandus, C. et al. Interactions between siglec-7/9 receptors and ligands influence NK cell-dependent tumor immunosurveillance. J. Clin. Invest. 124, 1810–1820 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Holling, T. M., van der Stoep, N., Quinten, E. & van den Elsen, P. J. Activated human T cells accomplish MHC class II expression through T cell-specific occupation of class II transactivator promoter III. J. Immunol. 168, 763–770 (2002).

    Article  CAS  PubMed  Google Scholar 

  101. Krawczyk, M. et al. Long distance control of MHC class II expression by multiple distal enhancers regulated by regulatory factor X complex and CIITA. J. Immunol. 173, 6200–6210 (2004).

    Article  CAS  PubMed  Google Scholar 

  102. Berger, C. et al. Adoptive transfer of effector CD8+ T cells derived from central memory cells establishes persistent T cell memory in primates. J. Clin. Invest. 118, 294–305 (2008).

    Article  CAS  PubMed  Google Scholar 

  103. Gattinoni, L. et al. A human memory T cell subset with stem cell-like properties. Nat. Med. 17, 1290–1297 (2011). This article is the first to describe T SCM cells.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Sommermeyer, D. et al. Chimeric antigen receptor-modified T cells derived from defined CD8+ and CD4+ subsets confer superior antitumor reactivity in vivo. Leukemia 30, 492–500 (2016).

    Article  CAS  PubMed  Google Scholar 

  105. Xu, Y. et al. Closely related T-memory stem cells correlate with in vivo expansion of CAR.CD19-T cells and are preserved by IL-7 and IL-15. Blood 123, 3750–3759 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. 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 article reports that higher frequency of CD27 +PD-1 CD8 + CAR T cells predicts therapeutic response of CD19 CAR-T cell therapy in chronic lymphocytic leukaemia.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Louis, C. U. et al. Antitumor activity and long-term fate of chimeric antigen receptor-positive T cells in patients with neuroblastoma. Blood 118, 6050–6056 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Busch, D. H., Fräßle, S. P., Sommermeyer, D., Buchholz, V. R. & Riddell, S. R. Role of memory T cell subsets for adoptive immunotherapy. Semin. Immunol. 28, 28–34 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Blaeschke, F. et al. Induction of a central memory and stem cell memory phenotype in functionally active CD4+ and CD8+ CAR T cells produced in an automated good manufacturing practice system for the treatment of CD19+ acute lymphoblastic leukemia. Cancer Immunol. Immunother. 67, 1053–1066 (2018).

    Article  CAS  PubMed  Google Scholar 

  110. Geiger, R. et al. L-Arginine modulates T cell metabolism and enhances survival and anti-tumor activity. Cell 167, 829–842.e13 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

  112. Salter, A. I., Pont, M. J. & Riddell, S. R. Chimeric antigen receptor-modified T cells: CD19 and the road beyond. Blood 131, 2621–2629 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Ehninger, A. et al. Distribution and levels of cell surface expression of CD33 and CD123 in acute myeloid leukemia. Blood Cancer J. 4, e218 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Cruz, N. M. et al. Selection and characterization of antibody clones are critical for accurate flow cytometry-based monitoring of CD123 in acute myeloid leukemia. Leuk. Lymphoma 59, 978–982 (2018).

    Article  CAS  PubMed  Google Scholar 

  115. Taussig, D. C. et al. Hematopoietic stem cells express multiple myeloid markers: implications for the origin and targeted therapy of acute myeloid leukemia. Blood 106, 4086–4092 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Malaer, J. D. & Mathew, P. A. CS1 (SLAMF7, CD319) is an effective immunotherapeutic target for multiple myeloma. Am. J. Cancer Res. 7, 1637–1641 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  117. Hemmati, P. G. et al. Predictive significance of the European LeukemiaNet classification of genetic aberrations in patients with acute myeloid leukaemia undergoing allogeneic stem cell transplantation. Eur. J. Haematol. 98, 160–168 (2017).

    Article  PubMed  Google Scholar 

  118. 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).

    Article  CAS  PubMed  Google Scholar 

  119. Anguille, S. et al. Dendritic cell vaccination as postremission treatment to prevent or delay relapse in acute myeloid leukemia. Blood 130, 1713–1721 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Mirzaei, H. R., Rodriguez, A., Shepphird, J., Brown, C. E. & Badie, B. Chimeric antigen receptors T cell therapy in solid tumor: challenges and clinical applications. Front. Immunol. 8, 1850 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Zhang, E., Gu, J. & Xu, H. Prospects for chimeric antigen receptor-modified T cell therapy for solid tumors. Mol. Cancer 17, 7 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Castellarin, M., Watanabe, K., June, C. H., Kloss, C. C. & Posey, A. D. Driving cars to the clinic for solid tumors. Gene Ther. 25, 165–175 (2018).

    Article  CAS  PubMed  Google Scholar 

  123. D’Aloia, M. M., Zizzari, I. G., Sacchetti, B., Pierelli, L. & Alimandi, M. CAR-T cells: the long and winding road to solid tumors. Cell Death Dis. 9, 282 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Jackson, H. J., Rafiq, S. & Brentjens, R. J. Driving CAR T-cells forward. Nat. Rev. Clin. Oncol. 13, 370–383 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Jin, C. et al. Allogeneic lymphocyte-licensed DCs expand T cells with improved antitumor activity and resistance to oxidative stress and immunosuppressive factors. Mol. Ther. Methods Clin. Dev. 1, 14001 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Ligtenberg, M. A. et al. Coexpressed Catalase protects chimeric antigen receptor–redirected T cells as well as bystander cells from oxidative stress–induced loss of antitumor activity. J. Immunol. 196, 759–766 (2016).

    Article  CAS  PubMed  Google Scholar 

  127. Juillerat, A. et al. An oxygen sensitive self-decision making engineered CAR T-cell. Sci. Rep. 7, 39833 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. 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).

    Article  PubMed  PubMed Central  Google Scholar 

  129. Liu, X. et al. A chimeric switch-receptor targeting PD1 augments the efficacy of second-generation CAR T cells in advanced solid tumors. Cancer Res. 76, 1578–1590 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Mariathasan, S. et al. TGFβ attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells. Nature 554, 544–548 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Sukumaran, S. et al. Enhancing the potency and specificity of engineered T cells for cancer treatment. Cancer Discov. 8, 972–987 (2018). This study describes the approach of combining receptors to activate CAR T cells exclusively at the tumour site.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Peng, W. et al. Transduction of tumor-specific T cells with CXCR2 chemokine receptor improves migration to tumor and antitumor immune responses. Clin. Cancer Res. 16, 5458–5468 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Moon, E. K. et al. Expression of a functional CCR2 receptor enhances tumor localization and tumor eradication by retargeted human T cells expressing a mesothelin-specific chimeric antibody receptor. Clin. Cancer Res. 17, 4719–4730 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Craddock, J. A. et al. Enhanced tumor trafficking of GD2 chimeric antigen receptor T cells by expression of the chemokine receptor CCR2b. J. Immunother. 33, 780–788 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Caruana, I. et al. Heparanase promotes tumor infiltration and antitumor activity of CAR-redirected T lymphocytes. Nat. Med. 21, 524–529 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Zhang, L. et al. Improving adoptive T cell therapy by targeting and controlling IL-12 expression to the tumor environment. Mol. Ther. 19, 751–759 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Krenciute, G. et al. Transgenic expression of IL15 improves antiglioma activity of IL13Rα2-CAR T cells but results in antigen loss variants. Cancer Immunol. Res. 5, 571–581 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Kim, Y. G., Cha, J. & Chandrasegaran, S. Hybrid restriction enzymes: zinc finger fusions to Fok I cleavage domain. Proc. Natl Acad. Sci. USA 93, 1156–1160 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Urnov, F. D., Rebar, E. J., Holmes, M. C., Zhang, H. S. & Gregory, P. D. Genome editing with engineered zinc finger nucleases. Nat. Rev. Genet. 11, 636–646 (2010).

    Article  CAS  PubMed  Google Scholar 

  143. Porteus, M. H. & Carroll, D. Gene targeting using zinc finger nucleases. Nat. Biotechnol. 23, 967–973 (2005).

    Article  CAS  PubMed  Google Scholar 

  144. Cermak, T. et al. Efficient design and assembly of custom TALEN and other TAL effector-based constructs for DNA targeting. Nucleic Acids Res. 39, e82 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Moscou, M. J. & Bogdanove, A. J. A simple cipher governs DNA recognition by TAL effectors. Science 326, 1501–1501 (2009).

    Article  CAS  PubMed  Google Scholar 

  146. Boch, J. et al. Breaking the code of DNA binding specificity of TAL-type III effectors. Science 326, 1509–1512 (2009).

    Article  CAS  PubMed  Google Scholar 

  147. Doyle, E. L. et al. TAL effector specificity for base 0 of the DNA target is altered in a complex, effector- and assay-dependent manner by substitutions for the tryptophan in cryptic repeat -1. PLOS ONE 8, e82120 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Beurdeley, M. et al. Compact designer TALENs for efficient genome engineering. Nat. Commun. 4, 1762 (2013).

    Article  CAS  PubMed  Google Scholar 

  149. Boissel, S. et al. megaTALs: a rare-cleaving nuclease architecture for therapeutic genome engineering. Nucleic Acids Res. 42, 2591–2601 (2014).

    Article  CAS  PubMed  Google Scholar 

  150. Zaslavskiy, M., Bertonati, C., Duchateau, P., Duclert, A. & Silva, G. H. Efficient design of meganucleases using a machine learning approach. BMC Bioinformatics 15, 191 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Werther, R. et al. Crystallographic analyses illustrate significant plasticity and efficient recoding of meganuclease target specificity. Nucleic Acids Res. 45, 8621–8634 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Lee, C. M., Cradick, T. J., Fine, E. J. & Bao, G. Nuclease target site selection for maximizing on-target activity and minimizing off-target effects in genome editing. Mol. Ther. 24, 475–487 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Cong, L. et al. Multiplex genome engineering using CRISPR/Cas systems. Science 339, 819–823 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Hussain, W. et al. CRISPR/Cas system: a game changing genome editing technology, to treat human genetic diseases. Gene 685, 70–75 (2018).

    Article  CAS  PubMed  Google Scholar 

  156. Mali, P. et al. RNA-guided human genome engineering via Cas9. Science 339, 823–826 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Li, T. et al. CRISPR-Cpf1-mediated genome editing and gene regulation in human cells. Biotechnol. Adv. 37, 21–27 (2019).

    Article  CAS  PubMed  Google Scholar 

  158. van den Broek, T., Borghans, J. A. M. & van Wijk, F. The full spectrum of human naive T cells. Nat. Rev. Immunol. 18, 363–373 (2018).

    Article  CAS  PubMed  Google Scholar 

  159. Sallusto, F., Geginat, J. & Lanzavecchia, A. Central memory and effector memory T cell subsets: function, generation, and maintenance. Annu. Rev. Immunol. 22, 745–763 (2004).

    Article  CAS  PubMed  Google Scholar 

  160. Zhang, Q. & Lakkis, F. G. Memory T cell migration. Front. Immunol. 6, 504 (2015).

    PubMed  PubMed Central  Google Scholar 

  161. Valbon, S. F., Condotta, S. A. & Richer, M. J. Regulation of effector and memory CD8+ T cell function by inflammatory cytokines. Cytokine 82, 16–23 (2016).

    Article  CAS  PubMed  Google Scholar 

  162. Koch, S. et al. Multiparameter flow cytometric analysis of CD4 and CD8 T cell subsets in young and old people. Immun. Ageing 5, 6 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Ahmed, R. et al. Human stem cell-like memory T cells are maintained in a state of dynamic flux. Cell Rep. 17, 2811–2818 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  164. Cieri, N. et al. IL-7 and IL-15 instruct the generation of human memory stem T cells from naive precursors. Blood 121, 573–584 (2013).

    Article  CAS  PubMed  Google Scholar 

  165. Amsen, D., van Gisbergen, K. P. J. M., Hombrink, P. & van Lier, R. A. W. Tissue-resident memory T cells at the center of immunity to solid tumors. Nat. Immunol. 19, 538–546 (2018).

    Article  CAS  PubMed  Google Scholar 

  166. Gebhardt, T., Palendira, U., Tscharke, D. C. & Bedoui, S. Tissue-resident memory T cells in tissue homeostasis, persistent infection, and cancer surveillance. Immunol. Rev. 283, 54–76 (2018).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors thank C. Delenda (Cellectis) and V. Alcazer (Centre Léon Bérard) for their help in preparing tables and figures and D. Sourdive (Cellectis) for his critical comments on process and manufacturing and his help in establishing Fig. 1.

Author information

Authors and Affiliations

Authors

Contributions

S.D. contributed to all aspects of the article. P.D and L.P. researched data, provided substantial contribution to the content and wrote the article. S.A.G. contributed substantially to discussion of the content, wrote the article and edited and reviewed the manuscript before submission. G.M. and S.D. edited and reviewed the article before submission.

Corresponding author

Correspondence to S. Depil.

Ethics declarations

Competing interests

S.D. has been an employee of Cellectis and has served as a consultant for or on the scientific advisory boards of Servier, Celyad, PDC*line Pharma, Erytech, AstraZeneca, Elsalys and Netris Pharma. L.P. and P.D. are employees of Cellectis. S.A.G. has received research and/or clinical trial support from Novartis, Servier and Kite and has served as a consultant for or on study steering committees or scientific advisory boards of Novartis, Cellectis, Adaptimmune, Eureka, TCR2, Juno, GlaxoSmithKline, Vertex, Cure Genetics, Humanigen and Roche.

Additional information

Publisher’s note

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

Glossary

Leukapheresis

Procedure through which white blood cells are separated and collected from the blood with an advanced centrifuge while red blood cells and other blood components are returned into the circulation.

HLA disparities

Human leukocyte antigen (HLA) incompatibility between the donor and the recipient. HLA disparities are associated with higher risk of graft failure, delayed immune reconstitution and graft-versus-host disease. New protocols allow donors mismatched for up to six alleles (haploidentical donors) to be used in haematopoietic stem cell transplantation without detrimental graft-versus-host disease.

Antigen-naive

Not previously exposed to foreign antigens.

Extravillous cytotrophoblast cells

The cells of the outermost layer of the fetal component of the placenta.

Syncytiotrophoblast

The epithelial covering of the vascular embryonic placental villi, which invades the wall of the uterus to establish nutrient circulation between the embryo and the mother.

Alloimmunization

Formation of antibodies against non-self antigens (here human leukocyte antigen molecules of the donor).

Stem cell transplantation

(SCT). For allogeneic SCT, haematopoietic stem cells are taken from the bone marrow, peripheral blood or umbilical cord blood of a healthy donor matched for human leukocyte antigen alleles. For haploidentical transplant, a healthy first-degree relative — a parent, sibling or child — serves as a donor, who needs to be only a 50% match to the recipient.

Spectratyping

Technique that measures T cell receptor repertoire diversity.

Multiplex gene editing

Gene editing technology that targets multiple regions in a genome.

Non-myeloablative and lymphodepletive chemotherapy

A chemotherapy regimen that does not destroy all the cells of the bone marrow but specifically induces destruction of the lymphocytes.

Killer cell immunoglobulin-like receptors

(KIRs). These receptors on natural killer (NK) cells recognize groups of for human leukocyte antigen class I alleles. The interaction between a KIR and a class I allele inhibits reactivity of the NK cell. The absence of recognition of the appropriate KIR ligand on  a mismatched cell triggers NK cell reactivity.

Immunomagnetic separation

A technique for separating cells by means of their antigens bound to antibodies coating microscopic paramagnetic beads, which can then be separated by magnetic attraction.

PiggyBac transposition

The piggyBac transposon is a movable genetic element that efficiently transposes between vectors and chromosomes through a ‘cut-and-paste’ mechanism.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Depil, S., Duchateau, P., Grupp, S.A. et al. ‘Off-the-shelf’ allogeneic CAR T cells: development and challenges. Nat Rev Drug Discov 19, 185–199 (2020). https://doi.org/10.1038/s41573-019-0051-2

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41573-019-0051-2

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer