Tisagenlecleucel immunogenicity in relapsed/refractory acute lymphoblastic leukemia and diffuse large B-cell lymphoma

Karen Thudium Mueller, Stephan A Grupp, Shannon L Maude, John E Levine, Michael A Pulsipher, Michael W Boyer, Keith J August, G Doug Myers, Constantine S Tam, Ulrich Jaeger, Stephen Ronan Foley, Peter Borchmann, Stephen J Schuster, Edmund K Waller, Rakesh Awasthi, Bernd Potthoff, Andy Warren, Edward R Waldron, Fraser McBlane, Andrea Chassot-Agostinho, Theodore W Laetsch, Karen Thudium Mueller, Stephan A Grupp, Shannon L Maude, John E Levine, Michael A Pulsipher, Michael W Boyer, Keith J August, G Doug Myers, Constantine S Tam, Ulrich Jaeger, Stephen Ronan Foley, Peter Borchmann, Stephen J Schuster, Edmund K Waller, Rakesh Awasthi, Bernd Potthoff, Andy Warren, Edward R Waldron, Fraser McBlane, Andrea Chassot-Agostinho, Theodore W Laetsch

Abstract

Tisagenlecleucel is indicated for pediatric and young adult patients with relapsed/refractory (r/r) B-cell acute lymphoblastic leukemia (B-ALL) and adult patients with r/r diffuse large B-cell lymphoma (DLBCL). The tisagenlecleucel chimeric antigen receptor (CAR) contains a murine single-chain variable fragment domain; we examined the effects of humoral and cellular immune responses to tisagenlecleucel on clinical outcomes using 2 validated assays. Data were pooled from the ELIANA (registered at www.clinicaltrials.gov as #NCT02435849) and ENSIGN (#NCT02228096) trials in r/r B-ALL (N = 143) and the JULIET trial (#NCT02445248) in r/r DLBCL (N = 115). Humoral responses were determined by flow cytometric measurement of anti-murine CAR19 (mCAR19) antibodies in serum. Cellular responses were determined using T-cell production of interferon-γ in response to 2 different pools of mCAR19 peptides. Pretreatment anti-mCAR19 antibodies were detected in 81% of patients with r/r B-ALL and 94% of patients with r/r DLBCL. Posttreatment anti-mCAR19 antibodies were higher than patient-specific baseline in 42% of r/r B-ALL and 9% of r/r DLBCL patients. Pretreatment and posttreatment anti-mCAR19 antibodies did not affect tisagenlecleucel cellular kinetics, including maximum concentration and persistence (r2 < 0.05), clinical response (day-28 response, duration of response, and event-free survival), and safety. T-cell responses were consistent over time, with net responses <1% at baseline and posttreatment time points in a majority of patients and no effect on transgene expansion or persistence or outcomes. Presence of baseline and/or posttreatment anti-mCAR19 antibodies or T-cell responses did not alter the activity of tisagenlecleucel in patients with r/r B-ALL or r/r DLBCL.

© 2021 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.

Figures

Graphical abstract
Graphical abstract
Figure 1.
Figure 1.
Preexisting and posttreatment anti-mCAR19 antibodies vs tisagenlecleucel expansion. Tisagenlecleucel expansion (Cmax) in pediatric and young adult patients with r/r B-ALL by preexisting (baseline) (A) and posttreatment (postinfusion) (C) anti-mCAR19 antibodies. Patients in the ELIANA trial are indicated by blue circles, and patients in the ENSIGN trial are indicated by red triangles. Tisagenlecleucel expansion (Cmax) in adult patients with r/r DLBCL by preexisting (baseline) (B) and posttreatment (postinfusion) (D) anti-mCAR19 antibodies. Patients in the JULIET trial are indicated by blue diamonds.
Figure 2.
Figure 2.
Pretreatment humoral immunogenicity and maximum fold change in anti-mCAR19 antibodies posttreatment by response category. Baseline anti-mCAR19 antibody levels (A-D) and maximum fold change in posttreatment anti-mCAR19 antibody levels (E-H) (by MFI) by response category. CRi, CR with incomplete blood count recovery; NR, no response; PR, partial response; PD, progressive disease; SD, stable disease.
Figure 3.
Figure 3.
Effects of anti-mCAR19 antibodies on duration of response in pediatric and young adult patients with B-ALL. Duration of response among patients who were positive (blue bars) and negative (orange bars) for posttreatment anti-mCAR19 antibodies that increased above baseline levels. Censoring reasons are indicated by the symbols in the legend. Patients in the 90th percentile for anti-mCAR antibody MFI are shown (green circles). HSCT, hematopoietic stem cell transplantation.
Figure 4.
Figure 4.
Effects of posttreatment anti-mCAR19 antibodies on tisagenlecleucel persistence in pediatric and young adult patients with r/r B-ALL. Solid red circles represent patients who were positive for posttreatment anti-mCAR19 antibodies. Solid blue triangles represent patients who were negative for posttreatment anti-mCAR19 antibodies. Data are presented as the arithmetic mean ± standard deviation.
Figure 5.
Figure 5.
Effects of CD4+ T-cell responses to 2 pools of mCAR19 peptides on tisagenlecleucel transgene expansion and patient outcomes in pediatric and young adult patients with r/r B-ALL. Tisagenlecleucel transgene expansion (A-B) and patient outcomes (C-D). Patients from the ELIANA trial are indicated by blue circles, and patients from the ENSIGN trial are indicated by red triangles. Maximum T-cell responses are grouped by response category. Negative response data represent patients whose posttreatment T-cell responses were lower than their baseline responses. CRi, CR with incomplete blood count recovery; NR, no response.
Figure 6.
Figure 6.
Net T-cell responses by scheduled time point. CD4+ and CD8+ net responses to pool-1 peptides in pediatric and young adult patients with r/r B-ALL (A,C) and adult patients with r/r DLBCL (B,D). Patients with sustained CR/CR with incomplete blood count recovery (CRi) or CR/partial response (PR) are indicated by solid color circles and are color-coded by scheduled time point for assessment. Negative net response data represent patients whose posttreatment T-cell responses were lower than the negative control (dimethylsulfoxide DMSO).

References

    1. Mueller KT, Maude SL, Porter DL, et al. . Cellular kinetics of CTL019 in relapsed/refractory B-cell acute lymphoblastic leukemia and chronic lymphocytic leukemia. Blood. 2017;130(21):2317-2325.
    1. Mueller KT, Waldron E, Grupp SA, et al. . Clinical pharmacology of tisagenlecleucel in B-cell acute lymphoblastic leukemia. Clin Cancer Res. 2018;24(24):6175-6184.
    1. van Brummelen EM, Ros W, Wolbink G, Beijnen JH, Schellens JH.. Antidrug antibody formation in oncology: clinical relevance and challenges. Oncologist. 2016;21(10):1260-1268.
    1. Wang J, Lozier J, Johnson G, et al. . Neutralizing antibodies to therapeutic enzymes: considerations for testing, prevention and treatment. Nat Biotechnol. 2008;26(8):901-908.
    1. Wang YM, Wang J, Hon YY, Zhou L, Fang L, Ahn HY.. Evaluating and reporting the immunogenicity impacts for biological products—a clinical pharmacology perspective. AAPS J. 2016;18(2):395-403.
    1. Courtenay-Luck NS, Epenetos AA, Winearls CG, Ritter MA.. Preexisting human anti-murine immunoglobulin reactivity due to polyclonal rheumatoid factors. Cancer Res. 1987;47(16):4520-4525.
    1. van Schie KA, Wolbink GJ, Rispens T.. Cross-reactive and pre-existing antibodies to therapeutic antibodies--Effects on treatment and immunogenicity. MAbs. 2015;7(4):662-671.
    1. Lamers CH, Willemsen R, van Elzakker P, et al. . Immune responses to transgene and retroviral vector in patients treated with ex vivo-engineered T cells. Blood. 2011;117(1):72-82.
    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(8):1453-1464.
    1. KYMRIAH™ (tisagenlecleucel) suspension for intravenous infusion: prescribing information. East Hanover, NJ: Novartis Pharmceuticals Corp.; 2018.
    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(5):439-448.
    1. Schuster SJ, Bishop MR, Tam CS, et al. . JULIET investigators. Tisagenlecleucel in adult relapsed or refractory diffuse large B-cell lymphoma. N Engl J Med. 2019;380(1):45-56.
    1. Kalos M, Levine BL, Porter DL, et al. . T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia. Sci Transl Med. 2011;3(95):95ra73.
    1. Zhang H, Snyder KM, Suhoski MM, et al. . 4-1BB is superior to CD28 costimulation for generating CD8+ cytotoxic lymphocytes for adoptive immunotherapy. J Immunol. 2007;179(7):4910-4918.
    1. Maude SL, Grupp SA, Mody R, et al. . An updated analysis of tisagenlecleucel in pediatric/young adult patients with relapsed/refractory B-cell acute lymphoblastic leukemia (B-ALL) in a US multicenter clinical trial (ENSIGN) [abstract]. Presented at the Congress of the European Hematology Association. 14-17 June 2018. Stockholm, Sweden. Abstract PF174.
    1. Potthoff B, McBlane F, Spindeldreher S, Sickert D.. A cell-based immunogenicity assay to detect antibodies against chimeric antigen receptor expressed by tisagenlecleucel. J Immunol Methods. 2020;476:112692.
    1. Van Winkle P, Burchette R, Kim R, Raghunathan R, Qureshi N.. Prevalence and safety of intravenous immunoglobulin administration during maintenance chemotherapy in children with acute lymphoblastic leukemia in first complete remission: a health maintenance organization perspective. Perm J. 2018;22:17-141.
    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(4):625-638.
    1. Awasthi R, Pacaud L, Waldron E, et al. . Tisagenlecleucel cellular kinetics, dose, and immunogenicity in relation to clinical factors in relapsed/refractory DLBCL. Blood Adv. 2020;4(3):560-572.
    1. Xue L, Fiscella M, Rajadhyaksha M, et al. . Pre-existing biotherapeutic-reactive antibodies: survey results within the American Association of Pharmaceutical Scientists. AAPS J. 2013;15(3):852-855.
    1. Tjandra JJ, Ramadi L, McKenzie IF.. Development of human anti-murine antibody (HAMA) response in patients. Immunol Cell Biol. 1990;68(Pt 6): 367-376.
    1. Meredith RF, Bueschen AJ, Khazaeli MB, et al. . Treatment of metastatic prostate carcinoma with radiolabeled antibody CC49. J Nucl Med. 1994;35(6):1017-1022.
    1. Papadopoulos KP, Isaacs R, Bilic S, et al. . Unexpected hepatotoxicity in a phase I study of TAS266, a novel tetravalent agonistic Nanobody® targeting the DR5 receptor. Cancer Chemother Pharmacol. 2015;75(5):887-895.
    1. Holland MC, Wurthner JU, Morley PJ, et al. . Autoantibodies to variable heavy (VH) chain Ig sequences in humans impact the safety and clinical pharmacology of a VH domain antibody antagonist of TNF-α receptor 1. J Clin Immunol. 2013;33(7):1192-1203.
    1. Terness P, Opelz G.. Natural anti-immunoglobulin autoantibodies: irrelevant by-products or immunoregulatory molecules? Int Arch Allergy Immunol. 1998;115(4):270-277.
    1. Xue L, Rup B.. Evaluation of pre-existing antibody presence as a risk factor for posttreatment anti-drug antibody induction: analysis of human clinical study data for multiple biotherapeutics. AAPS J. 2013;15(3):893-896.
    1. Ling NR, Drysdale P.. Antibodies in human sera to F(ab’)2 fragments of monoclonal and polyclonal IgG. Int Arch Allergy Appl Immunol. 1981;66(4):459-463.
    1. Brezski RJ, Jordan RE.. Cleavage of IgGs by proteases associated with invasive diseases: an evasion tactic against host immunity? MAbs. 2010;2(3):212-220.
    1. Baumgarth N, Tung JW, Herzenberg LA.. Inherent specificities in natural antibodies: a key to immune defense against pathogen invasion. Springer Semin Immunopathol. 2005;26(4):347-362.
    1. Lutz HU, Binder CJ, Kaveri S.. Naturally occurring auto-antibodies in homeostasis and disease. Trends Immunol. 2009;30(1):43-51.
    1. YESCARTA® (axicabtagene ciloleucel) suspension for intravenous infusion. Santa Monica, CA: Kite Pharma Inc.; 2019.
    1. Bivi N, Moore T, Rodgers G, et al. . Investigation of pre-existing reactivity to biotherapeutics can uncover potential immunogenic epitopes and predict immunogenicity risk. MAbs. 2019;11(5):861-869.
    1. Bhoj VG, Arhontoulis D, Wertheim G, et al. . Persistence of long-lived plasma cells and humoral immunity in individuals responding to CD19-directed CAR T-cell therapy. Blood. 2016;128(3):360-370.
    1. Maus MV, Haas AR, Beatty GL, et al. . T cells expressing chimeric antigen receptors can cause anaphylaxis in humans. Cancer Immunol Res. 2013;1(1):26-31.
    1. Maude S, Barrett D, Rheingold S, et al. . Efficacy of humanized CD19-targeted chimeric antigen receptor (CAR)-modified T cells in children with relapsed ALL [abstract]. Presented at the 2016 ASCO Annual Meeting. 3-7 Jun 2016. Chicago, IL. Abstract 3007.
    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(6):2123-2138.
    1. Brudno JN, 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 [published correction appears in Nat Med. 2020;26(5):803]. Nat Med. 2020;26(2):270-280.

Source: PubMed

3
S'abonner