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.

  • Article
  • Published:

Safety and feasibility of anti-CD19 CAR T cells with fully human binding domains in patients with B-cell lymphoma

An Author Correction to this article was published on 14 April 2020

This article has been updated

Abstract

Anti-CD19 chimeric antigen receptor (CAR)-expressing T cells are an effective treatment for B-cell lymphoma, but often cause neurologic toxicity. We treated 20 patients with B-cell lymphoma on a phase I, first-in-human clinical trial of T cells expressing the new anti-CD19 CAR Hu19-CD828Z (NCT02659943). The primary objective was to assess safety and feasibility of Hu19-CD828Z T-cell therapy. Secondary objectives included assessments of blood levels of CAR T cells, anti-lymphoma activity, second infusions and immunogenicity. All objectives were met. Fifty-five percent of patients who received Hu19-CD828Z T cells obtained complete remission. Hu19-CD828Z T cells had clinical anti-lymphoma activity similar to that of T cells expressing FMC63-28Z, an anti-CD19 CAR tested previously by our group, which contains murine binding domains and is used in axicabtagene ciloleucel. However, severe neurologic toxicity occurred in only 5% of patients who received Hu19-CD828Z T cells, whereas 50% of patients who received FMC63-28Z T cells experienced this degree of toxicity (Pā€‰=ā€‰0.0017). T cells expressing Hu19-CD828Z released lower levels of cytokines than T cells expressing FMC63-28Z. Lower levels of cytokines were detected in blood from patients who received Hu19-CD828Z T cells than in blood from those who received FMC63-28Z T cells, which could explain the lower level of neurologic toxicity associated with Hu19-CD828Z. Levels of cytokines released by CAR-expressing T cells particularly depended on the hinge and transmembrane domains included in the CAR design.

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: Hu19-CD828Z CAR T cells have substantial anti-lymphoma activity.
Fig. 2: CAR T-cell characteristics and persistence.
Fig. 3: Cytokine production by infused CAR T cells, and blood cytokine levels.
Fig. 4: Comparison of CAR designs.
Fig. 5: Functional comparison of CARs.

Similar content being viewed by others

Data availability

All requests for raw and analyzed data and materials are promptly reviewed by the National Cancer Institute Technology Transfer Center to verify whether the request is subject to any intellectual property or confidentiality obligations. Patient-related data not included in the paper were generated as part of clinical trials and may be subject to patient confidentiality. Any data and materials that can be shared will be released via a material transfer agreement. All other data that support the findings of this study will be provided by the corresponding author upon reasonable request when possible. Raw data for all Figs. 1ā€“5 and Extended Data Fig. 3 are in the submitted Source Data Excel file.

CAR sequences were all submitted to GenBank.

GenBank accession number for LSIN-Hu19-CD828Z: MN698642

GenBank accession number for LSIN-FMC63-CD828Z: MN702884

GenBank accession number for LSIN-Hu19-28Z: MN702882

GenBank accession number for MSGV-Hu19-CD828Z: MN702883

GenBank accession number for MSGV-FMC63-28Z: HM852952.1

Change history

References

  1. Turtle, C. J. et al. Immunotherapy of non-Hodgkinā€™s lymphoma with a defined ratio of CD8+ and CD4+ CD19-specific chimeric antigen receptor-modified T cells. Sci. Transl. Med 8, 355ra116 (2016).

    ArticleĀ  PubMedĀ  PubMed CentralĀ  CASĀ  Google ScholarĀ 

  2. Kochenderfer, J. N. et al. Eradication of B-lineage cells and regression of lymphoma in a subject treated with autologous T cells genetically engineered to recognize CD19. Blood 116, 4099ā€“4102 (2010).

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  3. Kochenderfer, J. N. et al. B-cell depletion and remissions of malignancy along with cytokine-associated toxicity in a clinical trial of anti-CD19 chimeric-antigen-receptor-transduced T cells. Blood 119, 2709ā€“2720 (2012).

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  4. Kochenderfer, J. N. et al. Chemotherapy-refractory diffuse large B-cell lymphoma and indolent B-cell malignancies can be effectively treated with autologous T cells expressing an anti-CD19 chimeric antigen receptor. J. Clin. Oncol. 33, 540ā€“549 (2015).

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  5. Kochenderfer, J. N. et al. Lymphoma remissions caused by anti-CD19 chimeric antigen receptor T cells are associated with high serum interleukin-15 levels. J. Clin. Oncol. 35, 1803ā€“1813 (2017).

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  6. Kochenderfer, J. N. et al. Long-duration complete remissions of diffuse large B cell lymphoma after anti-CD19 chimeric antigen receptor T cell therapy. Mol. Ther. 25, 2245ā€“2253 (2017).

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

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

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

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

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  9. Brudno, J. N. & Kochenderfer, J. N. Chimeric antigen receptor T-cell therapies for lymphoma. Nat. Rev. Clin. Oncol. 15, 31ā€“46 (2018).

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  10. Savoldo, B. et al. CD28 costimulation improves expansion and persistence of chimeric antigen receptor-modified T cells in lymphoma subjects. J. Clin. Invest. 121, 1822ā€“1826 (2011).

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  11. Schuster, S. J. et al. Tisagenlecleucel in adult relapsed or refractory diffuse large B-cell lymphoma. N. Engl. J. Med. 380, 45ā€“56 (2019).

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  12. Sadelain, M. CAR therapy: the CD19 paradigm. J. Clin. Invest. 125, 3392ā€“3400 (2015).

    ArticleĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  13. Johnson, L. A. & June, C. H. Driving gene-engineered T cell immunotherapy of cancer. Cell Res. 27, 38ā€“58 (2017).

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  14. Ramos, C. A., Heslop, H. E. & Brenner, M. K. CAR-T cell therapy for lymphoma. Annu. Rev. Med. 67, 165ā€“183 (2016).

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  15. Ramos, C. A. et al. In vivo fate and activity of second- versus third-generation CD19-specific CAR-T cells in B cell non-Hodgkinā€™s lymphomas. Mol. Ther. 26, 2727ā€“2737 (2018).

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  16. 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 1ā€“2 trial. Lancet. Oncol. 20, 31ā€“42 (2019).

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  17. Brudno, J. N. & Kochenderfer, J. N. Recent advances in CAR T-cell toxicity: mechanisms, manifestations and management. Blood. Rev. 34, 45ā€“55 (2019).

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  18. Brudno, J. N. & Kochenderfer, J. N. Toxicities of chimeric antigen receptor T cells: recognition and management. Blood 127, 3321ā€“3330 (2016).

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

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

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  20. Hirayama, A. V. & Turtle, C. J. Toxicities of CD19 CAR-T cell immunotherapy. Am. J. Hematol. 94, S42ā€“S49 (2019).

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  21. Teachey, D. T. et al. Identification of predictive biomarkers for cytokine release syndrome after chimeric antigen receptor T-cell therapy for acute lymphoblastic leukemia. Cancer Discov 6, 664ā€“679 (2016).

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  22. Singh, N. et al. Monocyte lineageā€“derived IL-6 does not affect chimeric antigen receptor T-cell function. Cytotherapy. 19, 867ā€“880 (2017).

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  23. Santomasso, B. D. et al. Clinical and biological correlates of neurotoxicity associated with car t-cell therapy in subjects with B-cell acute lymphoblastic leukemia. Cancer Discov 8, 958ā€“971 (2018).

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  24. Gust, J. et al. Endothelial activation and bloodā€“brain barrier disruption in neurotoxicity after adoptive immunotherapy with CD19 CAR-T cells. Cancer Discov 7, 1404ā€“1419 (2017).

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  25. Karschnia, P. et al. Clinical presentation, management, and biomarkers of neurotoxicity after adoptive immunotherapy with CAR T cells. Blood 133, 2212ā€“2221 (2019).

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  26. Gust, J. et al. Glial injury in neurotoxicity after pediatric CD19-directed chimeric antigen receptor T cell therapy. Ann. Neurol. 86, 42ā€“54 (2019).

    CASĀ  PubMedĀ  Google ScholarĀ 

  27. Taraseviciute, A. et al. Chimeric antigen receptor T cellā€“mediated neurotoxicity in nonhuman primates. Cancer Discov 8, 750ā€“763 (2018).

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  28. 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, 2452ā€“2465 (2017).

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  29. Kochenderfer, J. N. et al. Construction and preclinical evaluation of an anti-CD19 chimeric antigen receptor. J. Immunother. 32, 689ā€“702 (2009).

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  30. Hughes, M. S. et al. Transfer of a TCR gene derived from a subject with a marked antitumor response conveys highly active T-cell effector functions. Hum. Gene Ther. 16, 457ā€“472 (2005).

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  31. Goff, S. L. et al. Randomized, prospective evaluation comparing intensity of lymphodepletion before adoptive transfer of tumor-infiltrating lymphocytes for subjects with metastatic melanoma. J. Clin. Oncol. 34, 2389ā€“2397 (2016).

    ArticleĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  32. Kochenderfer, J. N., Yu, Z., Frasheri, D., Restifo, N. P. & Rosenberg, S. A. Adoptive transfer of syngeneic T cells transduced with a chimeric antigen receptor that recognizes murine CD19 can eradicate lymphoma and normal B cells. Blood 116, 3875ā€“3886 (2010).

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  33. Gattinoni, L. et al. Removal of homeostatic cytokine sinks by lymphodepletion enhances the efficacy of adoptively transferred tumor-specific CD8+ T cells. J. Exp. Med. 202, 907ā€“912 (2005).

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  34. North, R. J. Cyclophosphamide-facilitated adoptive immunotherapy of an established tumor depends on elimination of tumor-induced suppressor T cells. J. Exp. Med. 155, 1063ā€“1074 (1982).

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  35. Sievers, S. A., Kelley, K. A., Astrow, S. H., Bot, A. & Wiltzius, J. J. W. Design and development of anti-linker antibodies for the detection and characterization of CAR T cells. Proceedings: AACR Annual Meeting, abstr. 1204 (2019).

  36. 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, 1112ā€“1121 (2016).

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  37. Brudno, J. N. et al. T cells genetically modified to express an anti-B-Cell maturation antigen chimeric antigen receptor cause remissions of poor-prognosis relapsed multiple myeloma. J. Clin. Oncol. 36, 2267ā€“2280 (2018).

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  38. Cheson, B. D. et al. Revised response criteria for malignant lymphoma. J. Clin. Oncol. 25, 579ā€“586 (2007).

    ArticleĀ  PubMedĀ  Google ScholarĀ 

  39. Lee, D. W. et al. ASTCT consensus grading for cytokine release syndrome and neurologic toxicity associated with immune effector cells. Biol. Blood. Marrow Transplant. 25, 625ā€“638 (2019).

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  40. National Cancer Institute Common Terminology Criteria for Adverse Events v3.0 https://www.eortc.be/services/doc/ctc/ctcaev3.pdf (2006).

  41. Sallusto, F., Lenig, D., Fƶrster, R., Lipp, M. & Lanzavecchia, A. Two subsets of memory T lymphocytes with distinct homing potentials and effector functions. Nature 401, 708ā€“712 (1999).

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  42. Meierhoff, G., Ott, P. A., Lehmann, P. V. & Schloot, N. C. Cytokine detection by ELISPOT: relevance for immunological studies in type 1 diabetes. Diabetes Metab. Res. Rev. 18, 367ā€“380 (2002).

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  43. De Angelo, D. et al. Clinical Outcomes for a phase II, single arm, multicenter trial of JCAR015 in adult B-ALL (ROCKET Study). SITC 2017 Annual Meeting Abstract Book, abstr. P217 (2017).

  44. Brentjens, R. J. et al. Genetically targeted T cells eradicate systemic acute lymphoblastic leukemia xenografts. Clin. Cancer Res. 13, 5426ā€“5435 (2007).

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  45. Ying, Z. et al. A safe and potent anti-CD19 CAR T cell therapy. Nat. Med. 25, 947ā€“953 (2019).

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

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

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  47. Ghorashian, S. et al. Enhanced CAR T cell expansion and prolonged persistence in pediatric subjects with ALL treated with a low-affinity CD19 CAR. Nat. Med. 25, 1408ā€“1414 (2019).

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  48. Zheng, Z., Chinnasamy, N. & Morgan, R. A. Protein L: a novel reagent for the detection of chimeric antigen receptor (CAR) expression by flow cytometry. J. Transl. Med. 10, 29 (2012).

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  49. Feldman, S. A. et al. Rapid production of clinical-grade gammaretroviral vectors in expanded surface roller bottles using a ā€œmodifiedā€ step-filtration process for clearance of packaging cells. Hum. Gene Ther. 22, 107ā€“115 (2011).

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  50. Warren, E. H. et al. Therapy of relapsed leukemia after allogeneic hematopoietic cell transplantation with T cells specific for minor histocompatibility antigens. Blood 115, 3869ā€“3878 (2010).

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  51. Morgan, R. A. et al. Case report of a serious adverse event following the administration of t cells transduced with a chimeric antigen receptor recognizing ERBB2. Mol. Ther. 18, 843ā€“851 (2010).

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  52. Cachau, R. E., Erickson, J. W. & Villar, H. O. Novel procedure for structure refinement in homology modeling and its application to the human class mu glutathione s-transferases. Protein Eng. Des. Sel. 7, 831ā€“839 (1994).

    ArticleĀ  CASĀ  Google ScholarĀ 

  53. Yokoyama, S. et al. A novel pathway of LPS uptake through syndecan-1 leading to pyroptotic cell death. eLife 7, e37854 (2018).

    ArticleĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  54. Marrink, S. J. et al. Computational modeling of realistic cell membranes. Chem. Rev. 119, 6184ā€“6226 (2019).

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  55. Bello, M. & Correa-Basurto, J. Energetic and flexibility properties captured by long molecular dynamics simulations of a membrane-embedded pMHCII-TCR complex. Mol. Biosyst. 12, 1350ā€“1366 (2016).

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

Download references

Acknowledgements

The clinical LSIN-Hu19-CD828Z vector was produced and gene-therapy monitoring was performed with assistance from the NHLBI-funded National Gene Vector Biorepository at Indiana University. We thank the following clinical units at the NIH clinical center for patient care: Experimental Transplantation and Immunotherapy Branch Clinical Service, the 3 Northeast Nursing Unit and the Department of Critical Care Medicine and the Intensive Care Unit staff. Funding for this work was from National Cancer Institute Intramural funding and Kite, a Gilead Company.

Author information

Authors and Affiliations

Authors

Contributions

J.N.K. designed the trial and supervised all activities. J.N.B. and J.N.K. wrote the paper. J.N.B., N.L., D.V., J.J.R., Y.-w.S., J.R., A.X., A.B., N.S., L.M., R.C., P.Y., B.H., D.F.S., S.A.R. and J.N.K. analyzed data. J.N.B., L.M., R.E., M.R., R.D., B.H., R.P., R.E.G. and J.N.K. provided patient care. N.L., D.V., Y-W.S., J.J.R., R.C. and P.Y. conducted experiments. All authors reviewed, edited and approved the manuscript.

Corresponding author

Correspondence to James N. Kochenderfer.

Ethics declarations

Competing interests

This work was supported by intramural funding of the Center for Cancer Research, NCI, NIH. In addition, the NCI has cooperative research and development agreements with Kite Pharma, a Gilead Company that supports development of anti-CD19 CAR T-cell therapies, and both J.N.K. and S.A.R. are NCI principal investigators of these research agreements. J.K. has a patent application for the Hu19-CD828Z CAR and has received royalty payments from Kite, a Gilead Company. A.B., J.R., A.X. and N.S. are all employees of Kite, a Gilead Company.

Additional information

Peer review information Saheli Sadanand was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

Publisherā€™s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 CONSORT.

Consort diagram of the Hu19-CD828Z clinical trial.

Extended Data Fig. 2 Hu19-CD828Z neurologic toxicities.

All Grade 4, 3, and 2 neurologic adverse events within the first month after CAR T-cell infusion are listed. Grading by National Cancer Institute Common Terminology Criteria for Adverse Events Version 3; all adverse events listed under ā€œNeurologicā€ are included except syncope. Syncope was not included because it was associated with cytokine-release syndrome and hypotension. The highest grade of each adverse event experienced by each patient is listed. For example, if a patient had both Grade 2 and Grade 3 tremor at different times, tremor is only listed under Grade 3.

Extended Data Fig. 3 FMC63-28Z neurologic toxicities.

All Grade 4, 3, and 2 neurologic adverse events within the first month after CAR T-cell infusion are listed. Grading by National Cancer Institute Common Terminology Criteria for Adverse Events Version 3; all adverse events listed under ā€œNeurologicā€ are included except syncope. Syncope was not included because it was associated with hypotension from cytokine-release syndrome. The highest grade of each adverse event experienced by each patient is listed. For example, if a patient had both Grade 2 and Grade 3 confusion at different times, confusion is only listed under Grade 3.

Extended Data Fig. 4 Peak serum protein levels.

For all proteins, all 22 patients on the trial of FMC63-28Z T cells and all 20 patients on the trial of Hu19-CD828Z T cells were compared. Proteins were measured in serum samples by LuminexĀ® assay between day 2 and 14 after CAR T-cell infusion. Statistics were by 2-tailed Mann-Whitney test.

Extended Data Fig. 5 Patient 3 immunologic protein levels.

Patient 3 was the only patient with Grade 3 or 4 neurologic toxicity on the Hu19-CD828Z trial. Peak serum levels of 9 immunological proteins are shown for patient 3. Peak levels were determined between day 2 and day 14 after CAR T-cell infusion. These 9 proteins are shown because they were found to be prominently different between the Hu19-CD828Z and FMC63-28Z clinical trials (Fig. 3). Proteins were measured by LuminexĀ® assay. MCP-1, monocyte chemotactic protein-1; IL, interleukin; TNF-alpha, tumor necrosis factor-alpha; MIP-1-alpha, macrophage inflammatory protein-1-alpha; IFN-gamma, interferon-gamma. The red bars indicate the median protein levels for all 20 patients that received Hu19-CD828Z CAR T cells.

Source data

Extended Data Fig. 6 Serum proteins areas under the curves.

For all proteins, all 22 patients on the trial of FMC63-28Z T cells and all 20 patients on the trial of Hu19-CD828Z T cells were compared. Proteins were measured in serum samples by LuminexĀ® assay from days 2 to 14 after CAR T-cell infusion. Area under the curve (AUC) was calculated by trapezoidal method. Statistics were by 2-tailed Mann-Whitney test.

Extended Data Fig. 7 Structural Models of CARs.

Top row: schematic representations of Hu19-CD828Z (left) and FMC63-28Z (right) CAR models are shown; scFv in blue; hinge in green; transmembrane domain in yellow; intracellular domain in red. The membrane position during molecular dynamics simulations is shown in grey. Bottom row: conformational flexibility for each corresponding CAR depicted as superimposed carbon-alpha traces for a set of 50 representative conformations observed during a 50 nanosecond molecular dynamics trajectory. The differences in flexibility originate in the very different structure and dynamic behavior of the corresponding hinge regions during the dynamics simulations. Transmembrane and scFv domains are affected by the hinge properties and display very different behaviors as well. A quantitative analysis of the molecular dynamics trajectories reveals that these behaviors affect the scFv mobility (assessed as molecular diffusibility) and the proper formation of a transmembrane dimer evaluated by the helix-helix occluded surface. All models assume a dimeric structure anchored by disulfide bonds. In short, Hu19-CD828Z exhibited less conformational flexibility than FMC63-28Z.

Extended Data Fig. 8 Anti-CAR immune responses measured by ELISPOT.

*Positive anti-CAR response was defined as 3x or greater increase in spot number from pretreatment to post-CAR T-cell infusion, and post-treatment spot number must have been 3x or more than the spot number of the media control. #Bin A contained peptides from the signal sequences, scFv linker, and hinge regions. Bin B contained peptides from the scFv light chain. Bin C contained peptides from the scFv heavy chain. Bin D contained peptides from transmembrane and intracellular domains. ^The increase in spots was the number of spots/400,000 total input PBMC at the positive time-point minus the number of spots/400,000 total input PBMC before CAR T-cell treatment.

Extended Data Fig. 9 Blood CAR T-cell levels and anti-CAR immune responses.

Anti-CAR T-cell responses were assessed by ELISPOT analysis of PBMC before CAR T-cell treatment and at time-points within 6 weeks after CAR T-cell infusion as summarized in Extended Data Fig. 8. CAR+ cell levels in the blood were assessed by quantitative PCR. The top row shows peak blood CAR+ cell levels with results divided into patients with or without anti-CAR responses by ELISPOT. (a) Hu19-CD828Z (b) FMC63-28Z. The bottom row shows blood CAR+ cell levels 1-month after CAR T-cell infusion with results divided into patients with or without anti-CAR responses detected by ELISPOT: (c) Hu19-CD828Z, (d) FMC63-28Z. No statistically significant differences in blood CAR+ cell levels were found between patients with or without anti-CAR responses. All patients with adequate cell samples for both ELISPOT and qPCR are included. P values by Mann-Whitney test are shown on the plots; significance was defined as P<0.05. Of the 4 comparisons, the FMC63-28Z 1-month comparison was closest to statistical significance with P=0.061. Each symbol represents an individual patient. The number of unique patients analyzed were as follows: Hu19-CD828Z Peak, n=18; FMC63-28Z Peak, n=18; Hu19-CD828Z 1 month, n=18; FMC63-28Z 1 month n=13.

Source data

Supplementary information

Supplementary Information

Supplementary Figs. 1ā€“7, Supplementary Tables 1ā€“7 and Clinical Trial Protocols

Reporting Summary

Source data

Source Data Fig. 1

Statistical source data

Source Data Fig. 2

Statistical source data

Source Data Fig. 3

Statistical source data

Source Data Fig. 4

Statistical source data

Source Data Fig. 5

Statistical source data

Source Data Extended Data Fig. 5

Statistical source data

Source Data Extended Data Fig. 9

Statistical source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Brudno, J.N., Lam, N., Vanasse, D. 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, 270ā€“280 (2020). https://doi.org/10.1038/s41591-019-0737-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41591-019-0737-3

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter ā€” top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research