Efficacy and safety of CD19 CAR T constructed with a new anti-CD19 chimeric antigen receptor in relapsed or refractory acute lymphoblastic leukemia

Runxia Gu, Fang Liu, Dehui Zou, Yingxi Xu, Yang Lu, Bingcheng Liu, Wei Liu, Xiaojuan Chen, Kaiqi Liu, Ye Guo, Xiaoyuan Gong, Rui Lv, Xia Chen, Chunlin Zhou, Mengjun Zhong, Huijun Wang, Hui Wei, Yingchang Mi, Lugui Qiu, Lulu Lv, Min Wang, Ying Wang, Xiaofan Zhu, Jianxiang Wang, Runxia Gu, Fang Liu, Dehui Zou, Yingxi Xu, Yang Lu, Bingcheng Liu, Wei Liu, Xiaojuan Chen, Kaiqi Liu, Ye Guo, Xiaoyuan Gong, Rui Lv, Xia Chen, Chunlin Zhou, Mengjun Zhong, Huijun Wang, Hui Wei, Yingchang Mi, Lugui Qiu, Lulu Lv, Min Wang, Ying Wang, Xiaofan Zhu, Jianxiang Wang

Abstract

Background: Recent evidence suggests that resistance to CD19 chimeric antigen receptor (CAR)-modified T cell therapy may be due to the presence of CD19 isoforms that lose binding to the single-chain variable fragment (scFv) in current use. As such, further investigation of CARs recognize different epitopes of CD19 antigen may be necessary.

Methods: We generated a new CD19 CAR T (HI19α-4-1BB-ζ CAR T, or CNCT19) that includes an scFv that interacts with an epitope of the human CD19 antigen that can be distinguished from that recognized by the current FMC63 clone. A pilot study was undertaken to assess the safety and feasibility of CNCT19-based therapy in both pediatric and adult patients with relapsed/refractory acute lymphoblastic leukemia (R/R B-ALL).

Results: Data from our study suggested that 90% of the 20 patients treated with infusions of CNCT19 cells reached complete remission or complete remission with incomplete count recovery (CR/CRi) within 28 days. The CR/CRi rate was 82% when we took into account the fully enrolled 22 patients in an intention-to-treat analysis. Of note, extramedullary leukemia disease of two relapsed patients disappeared completely after CNCT19 cell infusion. After a median follow-up of 10.09 months (range, 0.49-24.02 months), the median overall survival and relapse-free survival for the 20 patients treated with CNCT19 cells was 12.91 months (95% confidence interval [CI], 7.74-18.08 months) and 6.93 months (95% CI, 3.13-10.73 months), respectively. Differences with respect to immune profiles associated with a long-term response following CAR T cell therapy were also addressed. Our results revealed that a relatively low percentage of CD8+ naïve T cells was an independent factor associated with a shorter period of relapse-free survival (p = 0.012, 95% CI, 0.017-0.601).

Conclusions: The results presented in this study indicate that CNCT19 cells have potent anti-leukemic activities in patients with R/R B-ALL. Furthermore, our findings suggest that the percentage of CD8+ naïve T cells may be a useful biomarker to predict the long-term prognosis for patients undergoing CAR T cell therapy.

Trial registration: ClinicalTrials.gov : NCT02975687; registered 29 November, 2016. https://ichgcp.net/clinical-trials-registry/NCT02975687.

Keywords: Acute lymphoblastic leukemia; Chimeric antigen receptor-modified T cell; HI19α; Single-chain variable fragment.

Conflict of interest statement

Jianxiang Wang received grant from Celgene. The other authors declare that they have no conflict of interest.

Figures

Fig. 1
Fig. 1
FMC63 and HI19α had different binding epitopes on CD19. a Homology models of hCD19 ECD (left), FMC63 scFv (middle), and HI19α scFv (right). b Docking mode of FMC63 and hCD19 ECD. Green, hCD19 ECD epitopes; purple, FMC63 epitopes. c Docking mode of HI19α and hCD19 ECD. Green, hCD19 ECD epitopes; purple, HI19α epitopes. d Sequence of hCD19 ECD. Gray background, predicted binding amino acid residues on hCD19 ECD with scFvs. Upper panel, interaction with HI19α; lower panel, interaction with FMC63. Red color, shared antigen epitopes; yellow color, key epitopes; green color, both shared antigen epitopes and key epitopes. e Partial interaction modes showed the non-bond interaction between scFvs and hCD19 ECD. Upper panel, HI19α scFv and hCD19 ECD; lower panel, FMC63 scFv and hCD19 ECD. Green, hCD19 ECD epitopes; purple, scFvs epitopes. f Flow cytometry analysis of the proportion of CD19+ Nalm-6 cells stained with indicated concentrations of the antibody. Left panel, HI19α antibody; right panel, FMC63 antibody. g Representative flow cytometry analysis showing the proportion of CD19+ cells on Nalm-6 cells stained with 0.017 pM HI19α and 0.042 pM FMC63
Fig. 2
Fig. 2
Responses of all 20 R/R B-ALL patients to CNCT19 CAR T cell therapy. In this single-center, open-label, prospective clinical trial, a total of 20 patients diagnosed with resistant or refractory CD19+ B cell acute lymphoblastic leukemia received an infusion of CNCT19 CAR T cells. Responses over time of each patient are presented as a swimmer plot
Fig. 3
Fig. 3
Long-term survival. ab Overall survival (a) and relapse-free survival (b) of the 20 R/R B-ALL patients treated with the CNCT19 CAR T cell infusion. Overall survival (c) and relapse-free survival (d) of patients who underwent hematopoietic stem cell transplantation. Overall survival (e) or relapse-free survival (f) of both pediatric and adult patients
Fig. 4
Fig. 4
The expansion kinetics of CAR T cells and different T cell subsets after CD19 CAR T cell infusion. a The expansion and persistence of CNCT19 CAR T cells after infusion. b The expansion and percentage of CD4+ or CD8+ T cells in peripheral blood after CAR infusion. cd The phenotype of CD8+ T cells (c) or CD4+ T cells (d) in peripheral blood after CNCT19 CAR T cell infusion (TN, naïve T cells; TCM, central memory T cells; TEM, effective memory T cells; and TE, effector T cells)
Fig. 5
Fig. 5
The correlation between TN cell percentage and long-term survival. a, b Comparison of CD8+ TN cells (a) or CD4+ TN cell (b) percentage in peripheral blood at different time points after CAR T cell infusion between long-term response and relapsed patients (TN, naïve T cells). CD8+ or CD4+ TN cells percentage were ranked and divided into four quartiles, from lowest (Q1) to highest (Q4). c, d Relapse-free survival (c) or overall survival (d) difference between patients with low CD8+ TN cells percentage (Q1) and those with high CD8+ TN cells percentage (Q2–4). e, f Relapse-free survival (e) or overall survival (f) difference between patients with low CD4+ TN cells percentage (Q1) and high CD4+ TN cells percentage (Q2–4)

References

    1. Grupp SA, Kalos M, Barrett D, Aplenc R, Porter DL, Rheingold SR, et al. Chimeric antigen receptor-modified T cells for acute lymphoid leukemia. N Engl J Med. 2013;368(16):1509–1518.
    1. Lee DW, Kochenderfer JN, Stetler-Stevenson M, Cui YK, Delbrook C, Feldman SA, 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(9967):517–528.
    1. Maude SL, Frey N, Shaw PA, Aplenc R, Barrett DM, Bunin NJ, et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med. 2014;371(16):1507–1517.
    1. Brudno JN, Somerville RPT, Shi V, Rose JJ, Halverson DC, Fowler DH, 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(10):1112–1121.
    1. Davila ML, Riviere I, Wang X, Bartido S, Park J, Curran K, et al. Efficacy and toxicity management of 19-28z CAR T cell therapy in B cell acute lymphoblastic leukemia. Sci Transl Med. 2014;6(224):224ra225.
    1. Savoldo B, Ramos CA, Liu E, Mims MP, Keating MJ, Carrum G, et al. CD28 costimulation improves expansion and persistence of chimeric antigen receptor-modified T cells in lymphoma patients. J Clin Invest. 2011;121(5):1822–1826.
    1. Sommermeyer D, Hudecek M, Kosasih PL, Gogishvili T, Maloney DG, Turtle CJ, et al. Chimeric antigen receptor-modified T cells derived from defined CD8+ and CD4+ subsets confer superior antitumor reactivity in vivo. Leukemia. 2016;30(2):492–500.
    1. Ying Z, Huang XF, Xiang X, Liu Y, Kang X, Song Y, et al. A safe and potent anti-CD19 CAR T cell therapy. Nat Med. 2019;25(6):947–953.
    1. Sotillo E, Barrett DM, Black KL, Bagashev A, Oldridge D, Wu G, et al. Convergence of acquired mutations and alternative splicing of CD19 enables resistance to CART-19 immunotherapy. Cancer Discov. 2015;5(12):1282–1295.
    1. Fischer J, Paret C, El Malki K, Alt F, Wingerter A, Neu MA, et al. CD19 isoforms enabling resistance to CART-19 immunotherapy are expressed in B-ALL patients at initial diagnosis. J Immunother. 2017;40(5):187–195.
    1. An N, Tao Z, Li S, Xing H, Tang K, Tian Z, et al. Construction of a new anti-CD19 chimeric antigen receptor and the anti-leukemia function study of the transduced T cells. Oncotarget. 2016;7(9):10638–10649.
    1. Park JH, Riviere I, Gonen M, Wang X, Senechal B, Curran KJ, et al. Long-term follow-up of CD19 CAR therapy in acute lymphoblastic leukemia. N Engl J Med. 2018;378(5):449–459.
    1. Hinrichs CS, Borman ZA, Gattinoni L, Yu ZY, Burns WR, Huang JP, et al. Human effector CD8(+) T cells derived from naive rather than memory subsets possess superior traits for adoptive immunotherapy. Blood. 2011;117(3):808–814.
    1. Xu Y, Zhang M, Ramos CA, Durett A, Liu E, Dakhova O, 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. 2014;123(24):3750–3759.
    1. Kalos M, Levine BL, Porter DL, Katz S, Grupp SA, Bagg A, 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. Klebanoff CA, Gattinoni L, Restifo NP. Sorting through subsets: which T-cell populations mediate highly effective adoptive immunotherapy? J Immunother. 2012;35(9):651–660.
    1. Sabatino M, Hu J, Sommariva M, Gautam S, Fellowes V, Hocker JD, et al. Generation of clinical-grade CD19-specific CAR-modified CD8+ memory stem cells for the treatment of human B-cell malignancies. Blood. 2016;128(4):519–528.
    1. Louis CU, Savoldo B, Dotti G, Pule M, Yvon E, Myers GD, et al. Antitumor activity and long-term fate of chimeric antigen receptor-positive T cells in patients with neuroblastoma. Blood. 2011;118(23):6050–6056.
    1. Busch DH, Frassle SP, Sommermeyer D, Buchholz VR, Riddell SR. Role of memory T cell subsets for adoptive immunotherapy. Semin Immunol. 2016;28(1):28–34.
    1. Wu F, Zhang W, Shao H, Bo H, Shen H, Li J, et al. Human effector T cells derived from central memory cells rather than CD8(+)T cells modified by tumor-specific TCR gene transfer possess superior traits for adoptive immunotherapy. Cancer Lett. 2013;339(2):195–207.
    1. Hoffmann JM, Schubert ML, Wang L, Huckelhoven A, Sellner L, Stock S, et al. Differences in expansion potential of naive chimeric antigen receptor T cells from healthy donors and untreated chronic lymphocytic leukemia patients. Front Immunol. 2018;8.
    1. Teplyakov A, Obmolova G, Luo J, Gilliland GL. Crystal structure of B-cell co-receptor CD19 in complex with antibody B43 reveals an unexpected fold. Proteins. 2018;86(5):495–500.
    1. Li S, Tao Z, Xu Y, Liu J, An N, Wang Y, et al. CD33-specific chimeric antigen receptor T cells with different co-stimulators showed potent anti-leukemia efficacy and different phenotype. Hum Gene Ther. 2018;29(5):626–639.
    1. Lee DW, Gardner R, Porter DL, Louis CU, Ahmed N, Jensen M, et al. Current concepts in the diagnosis and management of cytokine release syndrome. Blood. 2014;124(2):188–195.
    1. Ghorashian S, Kramer AM, Onuoha S, Wright G, Bartram J, Richardson R, et al. Enhanced CAR T cell expansion and prolonged persistence in pediatric patients with ALL treated with a low-affinity CD19 CAR. Nat Med. 2019;25:1408–1414.
    1. Gardner RA, Finney O, Annesley C, Brakke H, Summers C, Leger K, et al. Intent-to-treat leukemia remission by CD19 CAR T cells of defined formulation and dose in children and young adults. Blood. 2017;129(25):3322–3331.
    1. Locke FL, Ghobadi A, Jacobson CA, Miklos DB, Lekakis LJ, Oluwole OO, 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(1):31–42.
    1. Brentjens RJ, Davila ML, Riviere I, Park J, Wang X, Cowell LG, et al. CD19-targeted T cells rapidly induce molecular remissions in adults with chemotherapy-refractory acute lymphoblastic leukemia. Sci Transl Med. 2013;5(177):177ra138.
    1. Turtle CJ, Hanafi LA, Berger C, Gooley TA, Cherian S, Hudecek M, 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. Maude SL, Laetsch TW, Buechner J, Rives S, Boyer M, Bittencourt H, et al. Tisagenlecleucel in Children and Young Adults with B-Cell Lymphoblastic Leukemia. N Engl J Med. 2018;378(5):439–448.
    1. Fry TJ, Shah NN, Orentas RJ, Stetler-Stevenson M, Yuan CM, Ramakrishnaet S, et al. CD22-targeted CAR T cells induce remission in B-ALL that is naive or resistant to CD19-targeted CAR immunotherapy. Nat Med. 2018;24(1):20–28.
    1. Zhao Y, Liu ZF, Wang X, Wu HT, Zhang JP, Yang JF, et al. Treatment with humanized selective CD19 CAR-T cells shows efficacy in highly treated B-ALL patients who have relapsed after receiving murine-based CD19 CAR-T therapies. Clin Cancer Res. 2019;25(18):5595–5607.
    1. Maus MV, Haas AR, Beatty GL, Albelda SM, Levine BL, Liu XJ, et al. T cells expressing chimeric antigen receptors can cause anaphylaxis in humans. Cancer Immunol Res. 2013;1(1):26–31.
    1. Neelapu SS. CAR-T efficacy: is conditioning the key? Blood. 2019;133(17):1799–1800.
    1. Lowe KL, Mackall CL, Norry E, Amado R, Jakobsen BK, Binder G. Fludarabine and neurotoxicity in engineered T-cell therapy. Gene Ther. 2018;25:176–191.
    1. Santomasso BD, Park JH, Salloum D, Riviere I, Flynn J, Mead E, et al. Clinical and biological correlates of neurotoxicity associated with CAR T-cell therapy in patients with B-cell acute lymphoblastic leukemia. Cancer Discov. 2018;8(8):958–971.
    1. Gust J, Hay KA, Hanafi LA, Li D, Myerson D, Gonzalez-Cuyar LF. Endothelial activation and blood-brain barrier disruption in neurotoxicity after adoptive immunotherapy with CD19 CAR-T cells. Cancer Discov. 2017;7(12):1404–1419.
    1. Yu S, Yi M, Qin S, Wu KM. Next generation chimeric antigen receptor T cells: safety strategies to overcome toxicity. Mol Cancer. 2019;18(1):125.
    1. Gattinoni L, Klebanoff CA, Palmer DC, Wrzesinski C, Kerstann K, Yu Z, et al. Acquisition of full effector function in vitro paradoxically impairs the in vivo antitumor efficacy of adoptively transferred CD8+ T cells. J Clin Invest. 2005;115(6):1616–1626.
    1. Golubovskaya V, Wu L. Different subsets of T cells, memory, effector functions, and CAR-T immunotherapy. Cancers (Basel). 2016;8(3).
    1. McLellan AD, Ali Hosseini Rad SM. Chimeric antigen receptor T cell persistence and memory cell formation. Immunol Cell Biol. 2019;97(7):664–674.
    1. Gattinoni L, Klebanoff CA, Restifo NP. Paths to stemness: building the ultimate antitumour T cell. Nat Rev Cancer. 2012;12(10):671–684.
    1. Hinrichs CS, Borman ZA, Cassard L, Gattinoni L, Spolski R, Yu Z, et al. Adoptively transferred effector cells derived from naive rather than central memory CD8+ T cells mediate superior antitumor immunity. Proc Natl Acad Sci U S A. 2009;106(41):17469–17474.
    1. Berger C, Jensen MC, Lansdorp PM, Gough M, Elliott C, Riddell SR. Adoptive transfer of effector CD8+ T cells derived from central memory cells establishes persistent T cell memory in primates. J Clin Invest. 2008;118(1):294–305.
    1. Gargett T, Brown MP. Different cytokine and stimulation conditions influence the expansion and immune phenotype of third-generation chimeric antigen receptor T cells specific for tumor antigen GD2. Cytotherapy. 2015;17(4):487–495.

Source: PubMed

3
Abonnere