Switching CAR T cells on and off: a novel modular platform for retargeting of T cells to AML blasts

M Cartellieri, A Feldmann, S Koristka, C Arndt, S Loff, A Ehninger, M von Bonin, E P Bejestani, G Ehninger, M P Bachmann, M Cartellieri, A Feldmann, S Koristka, C Arndt, S Loff, A Ehninger, M von Bonin, E P Bejestani, G Ehninger, M P Bachmann

Abstract

The adoptive transfer of CD19-specific chimeric antigen receptor engineered T cells (CAR T cells) resulted in encouraging clinical trials in indolent B-cell malignancies. However, they also show the limitations of this fascinating technology: CAR T cells can lead to even life-threatening off-tumor, on-target side effects if CAR T cells crossreact with healthy tissues. Here, we describe a novel modular universal CAR platform technology termed UniCAR that reduces the risk of on-target side effects by a rapid and reversible control of CAR T-cell reactivity. The UniCAR system consists of two components: (1) a CAR for an inert manipulation of T cells and (2) specific targeting modules (TMs) for redirecting UniCAR T cells in an individualized time- and target-dependent manner. UniCAR T cells can be armed against different tumor targets simply by replacement of the respective TM for (1) targeting more than one antigen simultaneously or subsequently to enhance efficacy and (2) reducing the risk for development of antigen-loss tumor variants under treatment. Here we provide 'proof of concept' for retargeting of UniCAR T cells to CD33- and/or CD123-positive acute myeloid leukemia blasts in vitro and in vivo.

Conflict of interest statement

MPB, AE and GE have filed patents and patent applications related to mAbs directed to CD33, La and the UniCAR platform technology. AE, SL and MC are employed by GEMoaB and CPT, respectively. The other authors declare no conflict of interest.

Figures

Figure 1
Figure 1
UniCAR T cells eradicate AML cells upon redirection with CD33-specific (αCD33 TM) or CD123-specific (αCD123 TM) targeting modules. (a) Schematic representation of T cells engineered with a conventional CAR (left panel, cCAR) or a UniCAR (right panel). For explanation, see text. (b) UniCAR surface expression was detected as described in the Materials and methods section. For all experiments, transduced T cells were sorted to >90% purity to allow comparison between different human donors. (c) Reduction of leukemic cells from three AML cell lines after 24 h of incubation with 2 × 104 engineered human T cells isolated from healthy donors in the presence (+) or absence (−) of 0.1 nM TM in an effector-to-target (e/t) ratio of 1:1. Samples were normalized to a target cell control without any T cells. Engineered T cells expressed either UniCARs containing a dual CD28/CD3-ζ signaling domain (open and closed circles), UniCARs lacking any signaling domain (head up triangle), enhanced green fluorescent protein (EGFP) marker protein (head down triangle) or where not genetically modified (rhombus). (d, e) Experimental set-up was similar to (c) but a lower e/t ratio of 1:5 was chosen. (d) The number of living MOLM-13 target cells was normalized to a control sample without any T cells. (e) T-cell expansion was calculated as the ratio of T cells present in the samples after 120 h (d5) to the number of cells seeded at the start of the experiment (d0). (f) Effective TM concentration required for lysis of MOLM-13 AML cells was determined after 24 h of cultivation with UniCAR T cells. TMs were added at the indicated concentrations. Statistical analysis for (c) was performed using nonparametric one-way analysis of variance (ANOVA; Kruskal–Wallis test) and post hoc Dunn's multiple comparison test. Results are indicated for UniCAR modified T cells plus TMs versus other samples (*P<0.05, **P<0.01, ***P<0.001).
Figure 2
Figure 2
Dual retargeting of UniCAR modified T cells against CD33 and CD123 enhances leukemic cell lysis. (a) Because of its modular nature, the UniCAR technology allows a redirection of UniCAR modified T cells against two antigens simultaneously or consecutively using either two monospecific TMs (TM 1+TM 2, left side) or combined dual-specific TMs (TM1-2, right side). (b) Concentration–response curves for combined CD33- and CD123-specific retargeting of UniCAR T cells. UniCAR T cells were incubated in an effector-to-target (e/t) cell ratio of 1:1 with 51Cr-labeled MOLM-13 (n=4, open squares) and OCI-AML3 (n=2, open rhombus) for 24 h. EC50 values were determined for anti-CD33 (αCD33 TM)+anti-CD123 TM (αCD123 TM): EC50 MOLM-13=70.2 pM, EC50 OCI-AML3=80.2 pM, for anti-CD123-anti-CD33 TM (αCD123-CD33 TM): EC50 MOLM-13=2.9 pM, EC50 OCI-AML3=11.7 pM. (c, d) Human engineered T cells from healthy donors or AML patients were incubated with 5 × 104 CD3-CD19- leukemic cells from AML patients in the presence (+) or absence (−) of anti-CD123-CD33 TM at the indicated concentrations and an e/t ratio of 1:1. T cells expressed either UniCARs containing a dual CD28/CD3-ζ signaling domain (open and closed circles), UniCARs lacking any signaling domain (head up triangle) or enhanced green fluorescent protein (EGFP) marker protein (head down triangle). (c) The number of living target cells after 48 h (left panel) and 120 h (right panel) is shown and compared with control AML samples without adding T cells (open squares). (d) T-cell expansion was calculated as the ratio of T cells present in the samples after 120 h (d5) to the number of cells seeded at the start of the experiment (d0). (e) T cell-specific cytokine secretion was determined from supernatants taken after 48 h from the experiments shown in (c). (f) Engineered T cells from AML patients were incubated with 2 × 104 AML target cells in the presence (+) or absence (−) of 0.5 nM TMs in an e/t ratio of 1:1. Number of living target cells was determined by flow cytometry after 24 h and compared with a control sample with target cells and TM but without T cells. Results from one representative donor are shown. Statistical analysis for (c) was performed using nonparametric one-way analysis of variance (ANOVA; Kruskal–Wallis test) and post hoc Dunn's multiple comparison test. Results are indicated for UniCAR modified T cells plus TMs versus other samples (*P<0.05).
Figure 3
Figure 3
UniCAR engineered T cells delay AML engraftment in a TM-dependent manner in vivo. (a) Pharmacokinetics of the dual-specific anti-CD123/CD33 TM (αCD123-CD33 TM) in peripheral blood of NSG mice upon i.v. injection via the tail vain or i.p. injection. Concentration of TM in blood samples was determined by ELISA. (b, c) Short-term treatment with anti-CD123-CD33 TM enhances the survival of NSG mice in an aggressive AML model. 1 × 106 human T cells engineered to express functional UniCARs (28/ζ+TM, red line, n=5), UniCARs lacking any signaling domain (Stop+TM, blue line, n=5) or expressing only enhanced green fluorescent protein (EGFP) marker protein (vc+TM, green line, n=3) were i.v. injected into NSG mice. After 28 days, 5 × 105 MOLM-13 were transferred into NSG mice via i.v. injection (Tx) and treatment with anti-CD123-CD33 TM (T) was started 5 days later. For this purpose, 250 ng TM per g mouse body weight was injected i.p. twice a day over 2 consecutive days. As additional controls, one group of mice was transplanted only with MOLM-13 and treated with anti-CD123-CD33 TM (w/o+TM, black line, n=4), and another group of mice was transplanted with functional UniCAR T cells and MOLM-13, but not treated with anti-CD123-CD33 TM (28/ζ w/o TM, light red line, n=4). (b) Survival curves of experimental groups and (c) percentage of CD3+ T cells, CD33+ MOLM-13 cells and human CD45+CD3-CD33- cells in the bone marrow of killed mice are shown. (d) To evaluate in vivo toxicity of UniCAR modified T cells, 1 × 106 engineered human T cells were i.v. injected into NSG mice. Body weight of mice was monitored in weekly intervals and expressed as percentage body weight change over time.

References

    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: 177ra38.
    1. Kochenderfer JN, Dudley ME, Feldman SA, Wilson WH, Spaner DE, Maric I 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 2012; 119: 2709–2720.
    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: 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: 1507–1517.
    1. Cartellieri M, Bachmann M, Feldmann A, Bippes C, Stamova S, Wehner R et al. Chimeric antigen receptor-engineered T cells for immunotherapy of cancer. J Biomed Biotechnol 2010; 5: 956304.
    1. Topfer K, Kempe S, Muller N, Schmitz M, Bachmann M, Cartellieri M et al. Tumor evasion from T cell surveillance. J Biomed Biotechnol 2011; 2011: 918471.
    1. Anurathapan U, Chan RC, Hindi HF, Mucharla R, Bajgain P, Hayes BC et al. Kinetics of tumor destruction by chimeric antigen receptor-modified T cells. Mol Ther 2014; 22: 623–633.
    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: 1509–1518.
    1. Lamers CH, Sleijfer S, Vulto AG, Kruit WH, Kliffen M, Debets R et al. Treatment of metastatic renal cell carcinoma with autologous T-lymphocytes genetically retargeted against carbonic anhydrase IX: first clinical experience. J Clin Oncol 2006; 24: e20–e22.
    1. Morgan RA, Yang JC, Kitano M, Dudley ME, Laurencot CM, Rosenberg SA. Case report of a serious adverse event following the administration of T cells transduced with a chimeric antigen receptor recognizing ERBB2. Mol Ther 2010; 18: 843–851.
    1. Ehninger A, Kramer M, Rollig C, Thiede C, Bornhauser M, von Bonin M et al. Distribution and levels of cell surface expression of CD33 and CD123 in acute myeloid leukemia. Blood Cancer J 2014; 4: e218.
    1. Barrett AJ, Le BK. Immunotherapy prospects for acute myeloid leukaemia. Clin Exp Immunol 2010; 161: 223–232.
    1. Walter RB, Appelbaum FR, Estey EH, Bernstein ID. Acute myeloid leukemia stem cells and CD33-targeted immunotherapy. Blood 2012; 119: 6198–6208.
    1. Taussig DC, Pearce DJ, Simpson C, Rohatiner AZ, Lister TA, Kelly G et al. Hematopoietic stem cells express multiple myeloid markers: implications for the origin and targeted therapy of acute myeloid leukemia. Blood 2005; 106: 4086–4092.
    1. Arndt C, Feldmann A, von Bonin M, Cartellieri M, Ewen EM, Koristka S et al. Costimulation improves the killing capability of T cells redirected to tumor cells expressing low levels of CD33: description of a novel modular targeting system. Leukemia 2014; 28: 59–69.
    1. Koristka S, Cartellieri M, Arndt C, Bippes CC, Feldmann A, Michalk I et al. Retargeting of regulatory T cells to surface-inducible autoantigen La/SS-B. J Autoimmun 2013; 42: 105–116.
    1. Cartellieri M, Koristka S, Arndt C, Feldmann A, Stamova S, von Bonin M et al. A novel ex vivo isolation and expansion procedure for chimeric antigen receptor engrafted human T cells. PLoS One 2014; 9: e93745.
    1. Bippes CC, Feldmann A, Stamova S, Cartellieri M, Schwarzer A, Wehner R et al. A novel modular antigen delivery system for immuno targeting of human 6-sulfo LacNAc-positive blood dendritic cells (SlanDCs). PLoS One 2011; 6: e16315.
    1. Stirnnagel K, Luftenegger D, Stange A, Swiersy A, Mullers E, Reh J et al. Analysis of prototype foamy virus particle-host cell interaction with autofluorescent retroviral particles. Retrovirology 2010; 7: 45.
    1. Szymczak AL, Workman CJ, Wang Y, Vignali KM, Dilioglou S, Vanin EF et al. Correction of multi-gene deficiency in vivo using a single 'self-cleaving' 2A peptide-based retroviral vector. Nat Biotechnol 2004; 22: 589–594.
    1. Arndt C, von Bonin M, Cartellieri M, Feldmann A, Koristka S, Michalk I et al. Redirection of T cells with a first fully humanized bispecific CD33-CD3 antibody efficiently eliminates AML blasts without harming hematopoietic stem cells. Leukemia 2013; 27: 964–967.
    1. Tettamanti S, Biondi A, Biagi E, Bonnet D. CD123 AML targeting by chimeric antigen receptors: a novel magic bullet for AML therapeutics? Oncoimmunology 2014; 3: e28835.
    1. Feldmann A, Arndt C, Topfer K, Stamova S, Krone F, Cartellieri M et al. Novel humanized and highly efficient bispecific antibodies mediate killing of prostate stem cell antigen-expressing tumor cells by CD8+ and CD4+ T cells. J Immunol 2012; 189: 3249–3259.
    1. Koristka S, Cartellieri M, Theil A, Feldmann A, Arndt C, Stamova S et al. Retargeting of human regulatory T cells by single-chain bispecific antibodies. J Immunol 2012; 188: 1551–1558.
    1. Bachmann M, Chang S, Bernd A, Mayet W, Meyer zum Buschenfelde KH, Muller WE. Translocation of the nuclear autoantigen La to cell surface: assembly and disassembly with the extracellular matrix. Autoimmunity 1991; 9: 99–107.
    1. von Bonin M, Wermke M, Cosgun KN, Thiede C, Bornhauser M, Wagemaker G et al. In vivo expansion of co-transplanted T cells impacts on tumor re-initiating activity of human acute myeloid leukemia in NSG mice. PLoS One 2013; 8: e60680.
    1. Rowe JM, Lowenberg B. Gemtuzumab ozogamicin in acute myeloid leukemia: a remarkable saga about an active drug. Blood 2013; 121: 4838–4841.
    1. Stamova S, Cartellieri M, Feldmann A, Bippes CC, Bartsch H, Wehner R et al. Simultaneous engagement of the activatory receptors NKG2D and CD3 for retargeting of effector cells to CD33-positive malignant cells. Leukemia 2011; 25: 1053–1056.
    1. Dutour A, Marin V, Pizzitola I, Valsesia-Wittmann S, Lee D, Yvon E et al. In vitro and in vivo antitumor effect of anti-CD33 chimeric receptor-expressing EBV-CTL against CD33 acute myeloid leukemia. Adv Hematol 2012; 2012: 683065.
    1. Laszlo GS, Estey EH, Walter RB. The past and future of CD33 as therapeutic target in acute myeloid leukemia. Blood Rev 2014; 28: 143–153.
    1. Marin V, Pizzitola I, Agostoni V, Attianese GM, Finney H, Lawson A et al. Cytokine-induced killer cells for cell therapy of acute myeloid leukemia: improvement of their immune activity by expression of CD33-specific chimeric receptors. Haematologica 2010; 95: 2144–2152.
    1. Gorczyca W, Sun ZY, Cronin W, Li X, Mau S, Tugulea S. Immunophenotypic pattern of myeloid populations by flow cytometry analysis. Methods Cell Biol 2011; 103: 221–266.
    1. Jordan CT, Upchurch D, Szilvassy SJ, Guzman ML, Howard DS, Pettigrew AL et al. The interleukin-3 receptor alpha chain is a unique marker for human acute myelogenous leukemia stem cells. Leukemia 2000; 14: 1777–1784.
    1. Qiu S, Jia Y, Xing H, Yu T, Yu J, Yu P et al. N-Cadherin and Tie2 positive CD34(+)CD38(-)CD123(+) leukemic stem cell populations can develop acute myeloid leukemia more effectively in NOD/SCID mice. Leuk Res 2014; 38: 632–637.
    1. He SZ, Busfield S, Ritchie DS, Hertzberg MS, Durrant S, Lewis ID et al. A Phase 1 study of the safety, pharmacokinetics and anti-leukemic activity of the anti-CD123 monoclonal antibody CSL360 in relapsed, refractory or high-risk acute myeloid leukemia. Leuk Lymphoma 2015; 56: 1406–1415.
    1. Gill S, Tasian SK, Ruella M, Shestova O, Li Y, Porter DL et al. Preclinical targeting of human acute myeloid leukemia and myeloablation using chimeric antigen receptor-modified T cells. Blood 2014; 123: 2343–2354.
    1. Mardiros A, Dos SC, McDonald T, Brown CE, Wang X, Budde LE et al. T cells expressing CD123-specific chimeric antigen receptors exhibit specific cytolytic effector functions and antitumor effects against human acute myeloid leukemia. Blood 2013; 122: 3138–3148.
    1. Wang QS, Wang Y, Lv HY, Han QW, Fan H, Guo B et al. Treatment of CD33-directed chimeric antigen receptor-modified T cells in one patient with relapsed and refractory acute myeloid leukemia. Mol Ther 2015; 23: 184–191.
    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: 104–113.
    1. Hombach A, Sent D, Schneider C, Heuser C, Koch D, Pohl C et al. T-cell activation by recombinant receptors: CD28 costimulation is required for interleukin 2 secretion and receptor-mediated T-cell proliferation but does not affect receptor-mediated target cell lysis. Cancer Res 2001; 61: 1976–1982.
    1. Maher J, Brentjens RJ, Gunset G, Riviere I, Sadelain M. Human T-lymphocyte cytotoxicity and proliferation directed by a single chimeric TCRzeta /CD28 receptor. Nat Biotechnol 2002; 20: 70–75.
    1. Balkhi MY, Ma Q, Ahmad S, Junghans RP. T cell exhaustion and Interleukin 2 downregulation. Cytokine 2015; 71: 339–347.
    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: 1822–1826.
    1. Scholler J, Brady TL, Binder-Scholl G, Hwang WT, Plesa G, Hege KM et al. Decade-long safety and function of retroviral-modified chimeric antigen receptor T cells. Sci Transl Med 2012; 4, 132ra53.
    1. Semsei I, Tröster H, Bartsch H, Schwemmle M, Igloi GL, Bachmann M. Isolation of rat cDNA clones coding for the autoantigen SS-B/La: detection of species-specific variations. Gene 1993; 126: 265–268.
    1. Yiannaki EE, Tzioufas AG, Bachmann M, Hantoumi J, Tsikaris V, Sakarellos-Daitsiotis M et al. The value of synthetic linear epitope analogues of La/SSB for the detection of autoantibodies to La/SSB; specificity, sensitivity and comparison of methods. Clin Exp Immunol 1998; 112: 152–158.

Source: PubMed

3
Subscribe