Third-generation CD28/4-1BB chimeric antigen receptor T cells for chemotherapy relapsed or refractory acute lymphoblastic leukaemia: a non-randomised, open-label phase I trial protocol

Xiao-Yi Tang, Yao Sun, Ang Zhang, Guo-Liang Hu, Wei Cao, Dan-Hong Wang, Bin Zhang, Hu Chen, Xiao-Yi Tang, Yao Sun, Ang Zhang, Guo-Liang Hu, Wei Cao, Dan-Hong Wang, Bin Zhang, Hu Chen

Abstract

Introduction: There is no curative treatment available for patients with chemotherapy relapsed or refractory CD19+ B cells-derived acute lymphoblastic leukaemia (r/r B-ALL). Although CD19-targeting second-generation (2nd-G) chimeric antigen receptor (CAR)-modified T cells carrying CD28 or 4-1BB domains have demonstrated potency in patients with advanced B-ALL, these 2 signalling domains endow CAR-T cells with different and complementary functional properties. Preclinical results have shown that third-generation (3rd-G) CAR-T cells combining 4-1BB and CD28 signalling domains have superior activation and proliferation capacity compared with 2nd-G CAR-T cells carrying CD28 domain. The aim of the current study is therefore to investigate the safety and efficacy of 3rd-G CAR-T cells in adults with r/r B-ALL.

Methods and analysis: This study is a phase I clinical trial for patients with r/r B-ALL to test the safety and preliminary efficacy of 3rd-G CAR-T cells. Before receiving lymphodepleting conditioning regimen, the peripheral blood mononuclear cells from eligible patients will be leukapheresed, and the T cells will be purified, activated, transduced and expanded ex vivo. On day 6 in the protocol, a single dose of 1 million CAR-T cells per kg will be administrated intravenously. The phenotypes of infused CAR-T cells, copy number of CAR transgene and plasma cytokines will be assayed for 2 years after CAR-T infusion using flow cytometry, real-time quantitative PCR and cytometric bead array, respectively. Moreover, several predictive plasma cytokines including interferon-γ, interleukin (IL)-6, IL-8, Soluble Interleukin (sIL)-2R-α, solubleglycoprotein (sgp)130, sIL-6R, Monocyte chemoattractant protein (MCP1), Macrophage inflammatory protein (MIP1)-α, MIP1-β and Granulocyte-macrophage colony-stimulating factor (GM-CSF), which are highly associated with severe cytokine release syndrome (CRS), will be used to forecast CRS to allow doing earlier intervention, and CRS will be managed based on a revised CRS grading system. In addition, patients with grade 3 or 4 neurotoxicities or persistent B-cell aplasia will be treated with dexamethasone (10 mg intravenously every 6 hours) or IgG, respectively. Descriptive and analytical analyses will be performed.

Ethics and dissemination: Ethical approval for the study was granted on 10 July 2014 (YLJS-2014-7-10). Written informed consent will be taken from all participants. The results of the study will be reported, through peer-reviewed journals, conference presentations and an internal organisational report.

Trial registration number: NCT02186860.

Keywords: IMMUNOLOGY; Third-generation; acute lymphoblastic leukemia; chimeric antigen receptor.

Conflict of interest statement

Conflicts of Interest: None declared.

Published by the BMJ Publishing Group Limited. For permission to use (where not already granted under a licence) please go to http://www.bmj.com/company/products-services/rights-and-licensing/.

Figures

Figure 1
Figure 1
The general study schema. CAR, chimeric antigen receptor; PBMC, peripheral blood mononuclear cell.
Figure 2
Figure 2
Anti-CD19 3rd-G CAR structure including FMC63 single-chain variable fragment (scFv), CD8α hinge and transmembrane, CD28 and 4-1BB signalling domains, and CD3ζ.e. 3rd-G, third-generation; CAR, chimeric antigen receptor.
Figure 3
Figure 3
CRS treatment algorithm. CRS, cytokine release syndrome.
Figure 4
Figure 4
Neurotoxicities treatment algorithm. CRS, cytokine release syndrome; IV Q6h, intravenously every 6 hours.

References

    1. Pui CH, Robison LL, Look AT. Acute lymphoblastic leukaemia. Lancet 2008;371:1030–43. 10.1016/S0140-6736(08)60457-2
    1. Kantoff PW, Higano CS, Shore ND et al. . Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med 2010;363:411–22. 10.1056/NEJMoa1001294
    1. Palucka K, Banchereau J. Cancer immunotherapy via dendritic cells. Nat Rev Cancer 2012;12:265–77. 10.1038/nrc3258
    1. Hodi FS, O'Day SJ, McDermott DF et al. . Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med 2010;363:711–23. 10.1056/NEJMoa1003466
    1. Robert C, Long GV, Brady B et al. . Nivolumab in previously untreated melanoma without BRAF mutation. N Engl J Med 2015;372:320–30. 10.1056/NEJMoa1412082
    1. Garon EB, Rizvi NA, Hui R et al. . Pembrolizumab for the treatment of non-small-cell lung cancer. N Engl J Med 2015;372:2018–28. 10.1056/NEJMoa1501824
    1. Rosenberg JE, Hoffman-Censits J, Powles T et al. . Atezolizumab in patients with locally advanced and metastatic urothelial carcinoma who have progressed following treatment with platinum-based chemotherapy: a single-arm, multicentre, phase 2 trial. Lancet 2016;387:1909–20. 10.1016/S0140-6736(16)00561-4
    1. June CH. Adoptive T cell therapy for cancer in the clinic. J Clin Invest 2007;117:1466–76. 10.1172/JCI32446
    1. Keppler SJ, Rosenits K, Koegl T et al. . Signal 3 cytokines as modulators of primary immune responses during infections: the interplay of type I IFN and IL-12 in CD8 T cell responses. PLoS ONE 2012;7:e40865 10.1371/journal.pone.0040865
    1. Imai C, Mihara K, Andreansky M et al. . Chimeric receptors with 4-1BB signaling capacity provoke potent cytotoxicity against acute lymphoblastic leukemia. Leukemia 2004;18:676–84. 10.1038/sj.leu.2403302
    1. Kowolik CM, Topp MS, Gonzalez S et al. . CD28 costimulation provided through a CD19-specific chimeric antigen receptor enhances in vivo persistence and antitumor efficacy of adoptively transferred T cells. Cancer Res 2006;66:10995–1004. 10.1158/0008-5472.CAN-06-0160
    1. Gross G, Waks T, Eshhar Z. Expression of immunoglobulin-T-cell receptor chimeric molecules as functional receptors with antibody-type specificity. Proc Natl Acad Sci USA 1989;86:10024–8. 10.1073/pnas.86.24.10024
    1. Finney HM, Akbar AN, Lawson AD. Activation of resting human primary T cells with chimeric receptors: costimulation from CD28, inducible costimulator, CD134, and CD137 in series with signals from the TCR zeta chain. J Immunol 2004;172:104–13. 10.4049/jimmunol.172.1.104
    1. Milone MC, Fish JD, Carpenito C et al. . Chimeric receptors containing CD137 signal transduction domains mediate enhanced survival of T cells and increased antileukemic efficacy in vivo. Mol Ther 2009;17:1453–64. 10.1038/mt.2009.83
    1. Gardner RA, Finney O, Smithers H et al. . Prolonged functional persistence of CD19CAR t cell products of defined CD4:CD8 composition and transgene expression determines durability of MRD-negative ALL remission. J Clin Oncol 2016;34(Suppl); abstr 3048.
    1. Brentjens RJ, Riviere I, Park JH et al. . Safety and persistence of adoptively transferred autologous CD19-targeted T cells in patients with relapsed or chemotherapy refractory B-cell leukemias. Blood 2011;118:4817–28. 10.1182/blood-2011-04-348540
    1. Brentjens RJ, Davila ML, Riviere I et al. . CD19-targeted T cells rapidly induce molecular remissions in adults with chemotherapy-refractory acute lymphoblastic leukemia. Sci Transl Med 2013;5:177ra38 10.1126/scitranslmed.3005930
    1. Davila ML, Riviere I, Wang X et al. . Efficacy and toxicity management of 19-28z CAR T cell therapy in B cell acute lymphoblastic leukemia. Sci Transl Med 2014;6:224ra25 10.1126/scitranslmed.3008226
    1. Park JH, Riviere I, Wang X et al. . Implications of minimal residual disease negative complete remission (MRD-CR) and allogeneic stem cell transplant on safety and clinical outcome of CD19-targeted 19-28z CAR modified T cells in adult patients with relapsed, refractory B-cell ALL. Blood 2015;126:682.
    1. Lee DW, Kochenderfer JN, Stetler-Stevenson M et al. . T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: a phase 1 dose-escalation trial. Lancet 2015;385:517–28. 10.1016/S0140-6736(14)61403-3
    1. Lee DW, Stetler-Stevenson M, Yuan CM et al. . Safety and response of incorporating CD19 chimeric antigen receptor T cell therapy in typical salvage regimens for children and young adults with acute lymphoblastic leukemia. Blood 2015;126:684.
    1. Brudno JN, Somerville RP, Shi V 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 2016;34:1112–21. 10.1200/JCO.2015.64.5929
    1. Brudno JN, Somerville R, Shi V et al. . Allogeneic T-cells expressing an anti-CD19 chimeric antigen receptor cause remissions of B-cell malignancies after allogeneic hematopoietic stem cell transplantation without causing graft-versus-host disease. Blood 2015;126:99.
    1. Maude SL, Frey N, Shaw PA et al. . Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med 2014;371:1507–17. 10.1056/NEJMoa1407222
    1. Maude SL, Teachey DT, Rheingold SR et al. . Sustained remissions with CD19-specific chimeric antigen receptor (CAR)-modified T cells in children with relapsed/refractory ALL. J Clin Oncol 2016;34(Suppl); abstr 3011.
    1. Maude SL, Barrett DM, Rheingold SR et al. . Efficacy of humanized CD19-targeted chimeric antigen receptor (CAR)-modified T cells in children with relapsed ALL. J Clin Oncol 2016;34(Suppl); abstr 3007.
    1. Cruz CR, Micklethwaite KP, Savoldo B 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 2013;122:2965–73. 10.1182/blood-2013-06-506741
    1. Dai H, Zhang W, Li X et al. . Tolerance and efficacy of autologous or donor-derived T cells expressing CD19 chimeric antigen receptors in adult B-ALL with extramedullary leukemia. Oncoimmunology 2015;4:e1027469 10.1080/2162402X.2015.1027469
    1. Turtle CJ, Hanafi LA, Berger C et al. . CD19 CAR-T cells of defined CD4+:CD8+ composition in adult B cell ALL patients. J Clin Invest 2016;126:2123–38. 10.1172/JCI85309
    1. Turtle CJ, Hanafi LA, Berger C et al. . Rate of durable complete response in ALL, NHL, and CLL after immunotherapy with optimized lymphodepletion and defined composition CD19 CAR-T cells. J Clin Oncol 2016;34(Suppl); abstr 102.
    1. Kebriaei P, Singh H, Huls MH et al. . Phase I trials using Sleeping Beauty to generate CD19-specific CAR T cells. J Clin Invest 2016;126:3363–76. 10.1172/JCI86721
    1. Hu Y, Sun J, Wu Z et al. . Predominant cerebral cytokine release syndrome in CD19-directed chimeric antigen receptor-modified T cell therapy. J Hematol Oncol 2016;9:70 10.1186/s13045-016-0299-5
    1. Dong L, Chang L, Gao Z et al. . Chimeric antigen receptor 4SCAR19-modified T cells in acute lymphoid leukemia: a phase II multi-center clinical trial in China. Blood 2015;126:3774.
    1. van der Stegen SJ, Hamieh M, Sadelain M. The pharmacology of second-generation chimeric antigen receptors. Nat Rev Drug Discov 2015;14:499–509. 10.1038/nrd4597
    1. Karlsson H, Svensson E, Gigg C et al. . Evaluation of intracellular signaling downstream chimeric antigen receptors. PLoS ONE 2015;10:e0144787 10.1371/journal.pone.0144787
    1. Lee DW, Gardner R, Porter DL et al. . Current concepts in the diagnosis and management of cytokine release syndrome. Blood 2014;124:188–95. 10.1182/blood-2014-05-552729
    1. Brudno JN, Kochenderfer JN. Toxicities of chimeric antigen receptor T cells: recognition and management. Blood 2016;127:3321–30. 10.1182/blood-2016-04-703751
    1. Frey NV, Luger SM. How I treat adults with relapsed or refractory Philadelphia chromosome-negative acute lymphoblastic leukemia. Blood 2015;126:589–96. 10.1182/blood-2014-09-551937
    1. Haso W, Lee DW, Shah NN et al. . Anti-CD22-chimeric antigen receptors targeting B-cell precursor acute lymphoblastic leukemia. Blood 2013;121:1165–74. 10.1182/blood-2012-06-438002
    1. Kochenderfer JN, Rosenberg SA. Treating B-cell cancer with T cells expressing anti-CD19 chimeric antigen receptors. Nat Rev Clin Oncol 2013;10:267–76. 10.1038/nrclinonc.2013.46
    1. Kalaitsidou M, Kueberuwa G, Schutt A et al. . CAR T-cell therapy: toxicity and the relevance of preclinical models. Immunotherapy 2015;7:487–97. 10.2217/imt.14.123
    1. Teachey DT, Lacey SF, Shaw PA et al. . Identification of predictive biomarkers for cytokine release syndrome after chimeric antigen receptor t-cell therapy for acute lymphoblastic leukemia. Cancer Discov 2016;6:664–79. 10.1158/-16-0040
    1. Bai Y, Kan S, Zhou S et al. . Enhancement of the in vivo persistence and antitumor efficacy of CD19 chimeric antigen receptor T cells through the delivery of modified TERT mRNA. Cell Discov 2015;1:15040 10.1038/celldisc.2015.40

Source: PubMed

3
Sottoscrivi