Pooled safety analysis of tisagenlecleucel in children and young adults with B cell acute lymphoblastic leukemia

John E Levine, Stephan A Grupp, Michael A Pulsipher, Andrew C Dietz, Susana Rives, G Douglas Myers, Keith J August, Michael R Verneris, Jochen Buechner, Theodore W Laetsch, Henrique Bittencourt, Andre Baruchel, Michael W Boyer, Barbara De Moerloose, Muna Qayed, Stella M Davies, Christine L Phillips, Timothy A Driscoll, Peter Bader, Krysta Schlis, Patricia A Wood, Rajen Mody, Lan Yi, Mimi Leung, Lamis K Eldjerou, Carl H June, Shannon L Maude, John E Levine, Stephan A Grupp, Michael A Pulsipher, Andrew C Dietz, Susana Rives, G Douglas Myers, Keith J August, Michael R Verneris, Jochen Buechner, Theodore W Laetsch, Henrique Bittencourt, Andre Baruchel, Michael W Boyer, Barbara De Moerloose, Muna Qayed, Stella M Davies, Christine L Phillips, Timothy A Driscoll, Peter Bader, Krysta Schlis, Patricia A Wood, Rajen Mody, Lan Yi, Mimi Leung, Lamis K Eldjerou, Carl H June, Shannon L Maude

Abstract

Background: Tisagenlecleucel, an anti-CD19 chimeric antigen receptor T cell therapy, has demonstrated efficacy in children and young adults with relapsed/refractory B cell acute lymphoblastic leukemia (B-ALL) in two multicenter phase 2 trials (ClinicalTrials.gov, NCT02435849 (ELIANA) and NCT02228096 (ENSIGN)), leading to commercialization of tisagenlecleucel for the treatment of patients up to age 25 years with B-ALL that is refractory or in second or greater relapse.

Methods: A pooled analysis of 137 patients from these trials (ELIANA: n=79; ENSIGN: n=58) was performed to provide a comprehensive safety profile for tisagenlecleucel.

Results: Grade 3/4 tisagenlecleucel-related adverse events (AEs) were reported in 77% of patients. Specific AEs of interest that occurred ≤8 weeks postinfusion included cytokine-release syndrome (CRS; 79% (grade 4: 22%)), infections (42%; grade 3/4: 19%), prolonged (not resolved by day 28) cytopenias (40%; grade 3/4: 34%), neurologic events (36%; grade 3: 10%; no grade 4 events), and tumor lysis syndrome (4%; all grade 3). Treatment for CRS included tocilizumab (40%) and corticosteroids (23%). The frequency of neurologic events increased with CRS severity (p<0.001). Median time to resolution of grade 3/4 cytopenias to grade ≤2 was 2.0 (95% CI 1.87 to 2.23) months for neutropenia, 2.4 (95% CI 1.97 to 3.68) months for lymphopenia, 2.0 (95% CI 1.87 to 2.27) months for leukopenia, 1.9 (95% CI 1.74 to 2.10) months for thrombocytopenia, and 1.0 (95% CI 0.95 to 1.87) month for anemia. All patients who achieved complete remission (CR)/CR with incomplete hematologic recovery experienced B cell aplasia; however, as nearly all responders also received immunoglobulin replacement, few grade 3/4 infections occurred >1 year postinfusion.

Conclusions: This pooled analysis provides a detailed safety profile for tisagenlecleucel during the course of clinical trials, and AE management guidance, with a longer follow-up duration compared with previous reports.

Keywords: chimeric antigen; hematologic neoplasms; pediatrics; receptors.

Conflict of interest statement

Competing interests: JEL has received research and/or clinical trial support from Incyte, Kamada, Mesoblast, and Biogen and has participated in consulting, study steering committees, or scientific/clinical advisory boards for Novartis, bluebird bio, Incyte, Ironwood, Mesoblast, Omeros, Oncoimmune, Talaris, and X4 Pharmaceuticals. SAG has received research and/or clinical trial support from Novartis, Servier, and Kite and has participated in consulting, study steering committees, or scientific/clinical advisory boards for Novartis, Cellectis, Adaptimmune, Eureka, TCR2, Juno, GlaxoSmithKline, Vertex, Cure Genetics, Humanigen, and Roche. MAP has participated in steering committees for the ENSIGN and ELIANA trials for Novartis, advisory boards, and educational activities for Novartis. ACD is a current employee of bluebird bio; bluebird bio has had no support or oversight of this research or manuscript. SR has received clinical trial support from Novartis, Servier, and Celgene and has participated in consulting, study steering committees, or scientific/clinical advisory boards for Novartis, Servier, Celgene, Cellectis, Kite/Bristol-Myers Squibb, JazzPharma, and Amgen. GDM has received payment and honoraria as a consultant to Novartis Pharma and for serving on the speaker’s bureau for Kymriah. KJA has participated in a speaker bureau and received travel accommodations and expenses from Novartis. MRV has participated in advisory boards for Novartis, Fate Therapeutics, and B-Mogen and has stock options from Fate Therapeutics and B-Mogen. JB has participated in study steering committees, advisory boards, and educational activities for Novartis, and advisory boards for Kite and Janssen. TWL has consulted for Novartis, Cellectis, Loxo Oncology, Eli Lilly, and Bayer; has received research funding from Novartis, Pfizer, and Bayer; and is currently affiliated with the Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA and the Division of Oncology, Center for Childhood Cancer Research and Cancer Immunotherapy Program, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA. HB has participated in consulting for Novartis and Jazz Pharmaceuticals and his department has received financial reimbursement for participation in tisagenlecleucel trials for Novartis. AB has received research and/or clinical trial support from Novartis, Servier, and Kite and participated in consulting, study steering committees, or scientific/clinical advisory boards for Novartis, Servier, Celgene, Jazz Pharma, AstraZeneca, Janssen, and Amgen. MWB has participated in advisory boards for Novartis and Thunder Biotech. BDM has received a travel grant from Jazz Pharma; her department has received financial reimbursement and compensation for participation in CTL019 trials for Novartis, and consulting for Novartis. MQ has received support for service on clinical advisory boards for Novartis and Bristol-Myers Squibb. SMD has received research support from Alexion Pharmaceuticals and served in a consulting role for Novartis. CLP has received support for service on clinical advisory board for Novartis.TAD has no conflicts to declare. PB has received institutional research grants from medac, Riemser, and Neovii and institutional compensation for advisory activity and speakers bureau from Miltenyi, Amgen, Novartis, Servier, medac, and Riemser. KS has no conflicts to declare. PAW was an employee of Novartis Pharmaceuticals Corporation, East Hanover, New Jersey, USA. RM has no conflicts to declare. LY is an employee of Novartis Pharmaceuticals Corporation, East Hanover, New Jersey, USA. ML is an employee of Novartis Pharmaceuticals Corporation, East Hanover, New Jersey, USA. LKE is an employee of Novartis Pharmaceuticals Corporation, East Hanover, New Jersey, USA. CHJ has received research support from Novartis and Tmunity Therapeutics. SLM has received clinical trial support from Novartis and has served in a consulting role, on advisory boards, or on study steering committees for Novartis, Kite, and Wugen.

© Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
Time of onset of CRS and neurologic events among patients who experienced both events. CRS, cytokine-release syndrome.
Figure 2
Figure 2
Kaplan-Meier analysis of time to resolution of prolonged grade 3/4 (A) neutropenia,* (B) lymphopenia,† (C) leukopenia,‡ and (D) thrombocytopenia§ to grade 2 or better in patients with response (CR/CRi) and indicated cytopenia at day 28 after tisagenlecleucel infusion. *CTCAE grading, neutrophils/mm3: grade 1: <LLN to 1500; grade 2: 1000–<1500; grade 3: 500–<1000; grade 4: <500. †CTCAE grading, lymphocytes/mm3: grade 1: <LLN to 800; grade 2: 500–<800; grade 3: 200–<500; grade 4: <200. ‡CTCAE grading, WBC/mm3: grade 1: <LLN to 3000; grade 2: 2000–<3000; grade 3: 1000–<2000; grade 4: <1000. §CTCAE grading, platelets/mm3: grade 1: <LLN to 75,000; grade 2: 50,000–<75,000; grade 3: 25,000–<50,000; grade 4: <25,000. CR, complete remission; CRi, complete remission with incomplete hematologic recovery; CTCAE, Common Terminology Criteria for Adverse Events; LLN, lower limit of normal; NE, not estimable; WBC, white blood cell.

References

    1. Kymriah (tisagenlecleucel) . Full prescribing information. East Hanover, NJ: Novartis Pharmaceuticals Corporation; 2018.
    1. Kymriah (tisagenlecleucel) . Summary of product characteristics. Dublin, Ireland: Novartis Europharm Limited; 2018.
    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, Laetsch TW, Buechner J, et al. . Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. N Engl J Med 2018;378:439–48. 10.1056/NEJMoa1709866
    1. Schuster SJ, Svoboda J, Chong EA, et al. . Chimeric antigen receptor T cells in refractory B-cell lymphomas. N Engl J Med 2017;377:2545–54. 10.1056/NEJMoa1708566
    1. Schuster SJ, Bishop MR, Tam CS, et al. . Tisagenlecleucel in adult relapsed or refractory diffuse large B-cell lymphoma. N Engl J Med 2019;380:45–56. 10.1056/NEJMoa1804980
    1. Locke FL, Ghobadi A, Jacobson CA, 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 2019;20:31–42. 10.1016/S1470-2045(18)30864-7
    1. Wang M, Munoz J, Goy A, et al. . KTE-X19 CAR T-cell therapy in relapsed or refractory mantle-cell lymphoma. N Engl J Med 2020;382:1331–42. 10.1056/NEJMoa1914347
    1. Shah BD, Bishop MR, Oluwole OO, et al. . End of phase I results of ZUMA-3, a phase 1/2 study of KTE-X19, anti-CD19 chimeric antigen receptor (CAR) T cell therapy, in adult patients (pts) with relapsed/refractory (R/R) acute lymphoblastic leukemia (ALL). J Clin Oncol 2019;37:7006. 10.1200/JCO.2019.37.15_suppl.7006
    1. Wayne A, Huynh V, Hijiya N. ZUMA-4 phase 1: KTE-x19, an anti-CD19 CAR T cell therapy, in children and adolescents with R/R B-ALL. Pediatric Blood and Cancer 2019;66:S23–4.
    1. Jacobson CA, Chavez JC, Sehgal AR, et al. . Interim analysis of zuma-5: a phase 2 study of axicabtagene ciloleucel (AXI-cel) in patients with relapsed/refractory indolent non-Hodgkin lymphoma. HemaSphere 2020;4:105.
    1. Abramson J, Palomba ML, Gordon L. Safety and efficacy results from transcend NHL 001, a multicenter phase 1 study of lisocabtagene maraleucel (Liso-cel) in relapsed/refractory (R/R) large B-cell lymphoma (LBCL). Oncolo Res Treatment 2020;43:215.
    1. Raje N, Berdeja J, Lin Y, et al. . Anti-BCMA CAR T-cell therapy bb2121 in relapsed or refractory multiple myeloma. N Engl J Med 2019;380:1726–37. 10.1056/NEJMoa1817226
    1. Berdeja JG, Alsina M, Shah ND, et al. . Updated results from an ongoing phase 1 clinical study of bb21217 anti-Bcma CAR T cell therapy. Blood 2019;134:927. 10.1182/blood-2019-126660
    1. San Miguel J, Shah N, Oriol A, et al. . Idecabtagene vicleucel (IDE-CEL; BB2121), a BCMA-targeted CAR T cell therapy, in patients with relapsed and refractory multiple myeloma: initial karmma results. HemaSphere 2020;4:61–2.
    1. Curran KJ, Margossian SP, Kernan NA, et al. . Toxicity and response after CD19-specific CAR T-cell therapy in pediatric/young adult relapsed/refractory B-ALL. Blood 2019;134:2361–8. 10.1182/blood.2019001641
    1. Shah NN, Highfill SL, Shalabi H, et al. . CD4/CD8 T-cell selection affects chimeric antigen receptor (CAR) T-cell potency and toxicity: updated results from a phase I anti-CD22 CAR T-cell trial. J Clin Oncol 2020;38:1938–50. 10.1200/JCO.19.03279
    1. Teachey DT, Bishop MR, Maloney DG, et al. . Toxicity management after chimeric antigen receptor T cell therapy: one size does not fit 'ALL'. Nat Rev Clin Oncol 2018;15:218. 10.1038/nrclinonc.2018.19
    1. Neelapu SS, Tummala S, Kebriaei P, et al. . Chimeric antigen receptor T-cell therapy - assessment and management of toxicities. Nat Rev Clin Oncol 2018;15:47–62. 10.1038/nrclinonc.2017.148
    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. JCO 2016;34:3011. 10.1200/JCO.2016.34.15_suppl.3011
    1. Porter DL, Hwang W-T, Frey NV, et al. . Chimeric antigen receptor T cells persist and induce sustained remissions in relapsed refractory chronic lymphocytic leukemia. Sci Transl Med 2015;7:303ra139. 10.1126/scitranslmed.aac5415
    1. Porter D, Frey N, Wood PA, et al. . Grading of cytokine release syndrome associated with the CAR T cell therapy tisagenlecleucel. J Hematol Oncol 2018;11:35. 10.1186/s13045-018-0571-y
    1. Maude SL, Pulsipher MA, Boyer MW, et al. . Efficacy and safety of CTL019 in the first US phase II multicenter trial in pediatric relapsed/refractory acute lymphoblastic leukemia: results of an interim analysis. Blood 2016;128:2801. 10.1182/blood.V128.22.2801.2801
    1. Maude SL, Grupp SA, Mody R, et al. . An updated analysis of tisagenlecleucel in pediatric/ young adult patients with relapsed/refractory (R/R) B-cell acute lymphoblastic leukemia (B-ALL) in a US multicenter clinical trial (ENSIGN). HemaSphere 2018;2:41.
    1. Grupp SA, Maude SL, Rives S, et al. . Updated analysis of the efficacy and safety of Tisagenlecleucel in pediatric and young adult patients with relapsed/refractory (r/r) acute lymphoblastic leukemia. Blood 2018;132:895. 10.1182/blood-2018-99-112599
    1. Tyagarajan S, Schmitt D, Acker C, et al. . Autologous cryopreserved leukapheresis cellular material for chimeric antigen receptor-T cell manufacture. Cytotherapy 2019;21:1198–205. 10.1016/j.jcyt.2019.10.005
    1. Tyagarajan S, Spencer T, Smith J. Optimizing CAR-T cell manufacturing processes during pivotal clinical trials. Mol Ther Methods Clin Dev 2020;16:136–44. 10.1016/j.omtm.2019.11.018
    1. US National Institutes of Health . Study of efficacy and safety of CTL019 in pediatric ALL patients (NCT02228096). Available: [Accessed 29 Oct 2018].
    1. US Department of Health and Human Services . Common terminology criteria for adverse events (CTCAE) version 4.03. Available: [Accessed 29 Oct 2018].
    1. Jacobsohn DA, Vogelsang GB. Acute graft versus host disease. Orphanet J Rare Dis 2007;2:35. 10.1186/1750-1172-2-35
    1. Blood and Marrow Transplant Clinical Trials Network . Definitions of chronic GVHD. Available: [Accessed 29 Oct 2018].
    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. Lee DW, Santomasso BD, Locke FL, et al. . ASTCT consensus grading for cytokine release syndrome and neurologic toxicity associated with immune effector cells. Biol Blood Marrow Transplant 2019;25:625–38. 10.1016/j.bbmt.2018.12.758
    1. Schuster SJ, Maziarz RT, Rusch ES, et al. . Grading and management of cytokine release syndrome in patients treated with tisagenlecleucel in the JULIET trial. Blood Adv 2020;4:1432–9. 10.1182/bloodadvances.2019001304
    1. Hay KA, Hanafi L-A, Li D, et al. . Kinetics and biomarkers of severe cytokine release syndrome after CD19 chimeric antigen receptor-modified T-cell therapy. Blood 2017;130:2295–306. 10.1182/blood-2017-06-793141
    1. Tedesco VE, Mohan C. Biomarkers for predicting cytokine release syndrome following CD19-Targeted CAR T cell therapy. J Immunol 2021;206:1561–8. 10.4049/jimmunol.2001249
    1. Porter DL, Levine BL, Kalos M, et al. . Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N Engl J Med 2011;365:725–33. 10.1056/NEJMoa1103849
    1. Grupp SA, Kalos M, Barrett D, et al. . Chimeric antigen receptor-modified T cells for acute lymphoid leukemia. N Engl J Med 2013;368:1509–18. 10.1056/NEJMoa1215134
    1. Gofshteyn JS, Shaw PA, Teachey DT, et al. . Neurotoxicity after CTL019 in a pediatric and young adult cohort. Ann Neurol 2018;84:537–46. 10.1002/ana.25315
    1. Topp M, Van Meerten T, Houot R, et al. . Earlier steroid use with axicabtagene ciloleucel (axi-cel) in patients with relapsed/refractory large B cell lymphoma. Blood 2019;134:243. 10.1182/blood-2019-126081
    1. Strati P, Ahmed S, Kebriaei P, et al. . Clinical efficacy of anakinra to mitigate CAR T-cell therapy-associated toxicity in large B-cell lymphoma. Blood Adv 2020;4:3123–7. 10.1182/bloodadvances.2020002328
    1. Jamal FA, Khaled SK. The cardiovascular complications of chimeric antigen receptor T cell therapy. Curr Hematol Malig Rep 2020;15:130–2. 10.1007/s11899-020-00567-4
    1. Ghosh AK, Chen DH, Guha A. CAR T cell therapy–related cardiovascular outcomes and management: systemic disease or direct cardiotoxicity? JACC CardioOncol 2020;2:97–109. 10.1016/j.jaccao.2020.02.011
    1. Cordeiro A, Bezerra ED, Hirayama AV, et al. . Late events after treatment with CD19-targeted chimeric antigen receptor modified T cells. Biol Blood Marrow Transplant 2020;26:26–33. 10.1016/j.bbmt.2019.08.003
    1. Gill S, Carney D, Ritchie D, et al. . The frequency, manifestations, and duration of prolonged cytopenias after first-line fludarabine combination chemotherapy. Ann Oncol 2010;21:331–4. 10.1093/annonc/mdp297
    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. Jiang H, Liu L, Guo T, et al. . Improving the safety of CAR-T cell therapy by controlling CRS-related coagulopathy. Ann Hematol 2019;98:1721–32. 10.1007/s00277-019-03685-z
    1. Wang Y, Qi K, Cheng H, et al. . Coagulation disorders after chimeric antigen receptor T cell therapy: analysis of 100 patients with relapsed and refractory hematologic malignancies. Biol Blood Marrow Transplant 2020;26:865–75. 10.1016/j.bbmt.2019.11.027
    1. Buechner J, Grupp SA, Hiramatsu H, et al. . Practical guidelines for monitoring and management of coagulopathy following tisagenlecleucel CAR T-cell therapy. Blood Adv 2021;5:593–601. 10.1182/bloodadvances.2020002757

Source: PubMed

3
S'abonner