A novel antibody-TCR (AbTCR) platform combines Fab-based antigen recognition with gamma/delta-TCR signaling to facilitate T-cell cytotoxicity with low cytokine release

Yiyang Xu, Zhiyuan Yang, Lucas H Horan, Pengbo Zhang, Lianxing Liu, Bryan Zimdahl, Shon Green, Jingwei Lu, Javier F Morales, David M Barrett, Stephan A Grupp, Vivien W Chan, Hong Liu, Cheng Liu, Yiyang Xu, Zhiyuan Yang, Lucas H Horan, Pengbo Zhang, Lianxing Liu, Bryan Zimdahl, Shon Green, Jingwei Lu, Javier F Morales, David M Barrett, Stephan A Grupp, Vivien W Chan, Hong Liu, Cheng Liu

Abstract

The clinical use of genetically modified T-cell therapies has led to unprecedented response rates in leukemia and lymphoma patients treated with anti-CD19 chimeric antigen receptor (CAR)-T. Despite this clinical success, FDA-approved T-cell therapies are currently limited to B-cell malignancies, and challenges remain with managing cytokine-related toxicities. We have designed a novel antibody-T-cell receptor (AbTCR) platform where we combined the Fab domain of an antibody with the γ and δ chains of the TCR as the effector domain. We demonstrate the ability of anti-CD19-AbTCR-T cells to trigger antigen-specific cytokine production, degranulation, and killing of CD19-positive cancer cells in vitro and in xenograft mouse models. By using the same anti-CD19 binding moiety on an AbTCR compared to a CAR platform, we demonstrate that AbTCR activates cytotoxic T-cell responses with a similar dose-response as CD28/CD3ζ CAR, yet does so with less cytokine release and results in T cells with a less exhausted phenotype. Moreover, in comparative studies with the clinically validated CD137 (4-1BB)-based CAR, CTL019, our anti-CD19-AbTCR shows less cytokine release and comparable tumor inhibition in a patient-derived xenograft leukemia model.

Conflict of interest statement

Y.X., Z.Y., L.H.H., P.Z., L.L., B.Z., S.G., J.L., J.F.M., V.W.C., H.L., and C.L. are employees of and have equity ownership and/or stock options in Eureka Therapeutics, Inc. S.A.G. is a scientific advisor to Eureka Therapeutics with stock options in the company. D.M.B. is a consultant to Eureka Therapeutics. Other associations: S.A.G. has received research and/or clinical trial support from Novartis, Servier and Kite. He consults for, has participated in ad boards, or serves on study steering committees or scientific advisory boards for the following companies: Novartis, Adaptimmune, TCR2, Juno, GlaxoSmithKline, Cellectis, Vertex, Roche and Janssen. He is listed on a patent related to toxicity management in cell therapy managed by University of Pennsylvania and CHOP policies.

Figures

Fig. 1. The molecular design, expression, and…
Fig. 1. The molecular design, expression, and cellular characterization of our antibody-TCR.
a Schematic of our antibody-TCR (AbTCR) platform compared to a TCR and a second-generation CAR platform. b ET190L1-AbTCR is expressed on the surface of primary T cells following lentiviral transduction. Anti-human Fab antibody was used to detect AbTCR expression. Representative flow cytometry plots and images (40×) of T cells stained with anti-human Fab antibody are shown. c AbTCR expression stabilizes the cell surface presentation of CD3ɛ on TCRβ negative J.RT3-T3.5 cells. d The CD3 complex co-immunoprecipitates with the AbTCR. Cell lysates and anti-FLAG immunoprecipitates from primary T cells expressing a FLAG-tagged ET190L1-AbTCR (or untransduced; Mock) were subjected to western blot analysis for CD3δ, ε, γ, and ζ chains
Fig. 2. AbTCR-T cells have a less…
Fig. 2. AbTCR-T cells have a less exhausted surface phenotype and a higher percentage of naïve and stem cell memory T cells compared to CAR-T cells.
a ET190L1-AbTCR T cells (AbTCR) and ET190L1-CAR-T cells (CAR) were cultured and the number of cells determined at the indicated time points. be Flow cytometry analysis of ET190L1-AbTCR-T cells and ET190L1-CAR-T cells at day 10 of in vitro expansion prior to antigen stimulation (b) Proportions of CD4/CD8 within receptor+ cells. c Frequency of naïve (CCR7+ CD45RA+), central memory (CM; CCR7+ CD45RA−), effector memory (EM; CCR7- CD45RA-) and effector (E; CCR7- CD45RA+) T cells within CD8+ receptor+ cells. d Frequency of stem cell memory (SCM; CCR7+ CD45RO- CD95+ CD122+) T cells within CD8+ receptor+ cells. e Expression of T cell differentiation markers CD28, CCR7, and granzyme B (GranB). f Expression of T cell exhaustion markers PD-1, LAG-3, and TIM-3. **P< 0.01
Fig. 3. AbTCR-T cells mediate antigen-specific responses…
Fig. 3. AbTCR-T cells mediate antigen-specific responses in vitro.
a Expression of activation markers CD69 and CD25 on ET190L1-AbTCR-T cells (AbTCR) and ET190L1-CAR-T cells (CAR) following a 16 h co-incubation with Raji or Raji K/O cells. Raji K/O: Raji cells in which CD19 was knocked out using CRISPR technology. Gated on CD3+ receptor+ cells. b ET190L1-AbTCR-T cells selectively degranulate in the presence of CD19+ target cells. Representative flow cytometry plot showing CD107a expression on ET190L1-AbTCR incubated with target cells for 4 h at an E:T of 1:1. Degranulation in AbTCR+ CD8+ T cells is shown. c ET190L1-AbTCR-T cells selectively kill CD19+ tumor cells and are comparable to ET190L1-CAR-T cells. T cells were incubated with target cells for 16 h at an E:T ratio of 2:1. Cytotoxicity was measured by LDH release assay (n= 3 technical replicates). Error bars, SEM. d ET190L1-AbTCR-T cells and ET190L1-CAR-T cells proliferate at similar rates upon antigen stimulation in vitro. T cells were labeled with CFSE and co-cultured with CD19+ Raji cells and the degree of T cell proliferation was assessed by flow cytometry. e ET190L1-AbTCR CD4+ T cells express lower levels of exhaustion markers following co-incubation with CD19+ tumor cells
Fig. 4. AbTCR-T cells release less CRS-related…
Fig. 4. AbTCR-T cells release less CRS-related cytokines than CAR-T cells, including monocyte-lineage-derived IL-6.
a ET190L1-AbTCR-T cells (AbTCR) release less cytokines than ET190L1-CAR-T cells (CAR) during in vitro killing assays. T cells were incubated with target cells for 16 h at an E:T ratio of 5:1. Secreted cytokine levels in the supernatant were measured. Data shown are representative of three independent experiments. b Cytokine expression after trans-well co-culture. T cells, targets (CD19+ NALM-6 cells) were placed in plate wells, and monocyte-lineage cells (APCs) were place in transwell inserts. Supernatants were collected after 18 h of co-cultures
Fig. 5. Robust tumor rejection and reduced…
Fig. 5. Robust tumor rejection and reduced cytokine release with ET190L1-AbTCR-T cells in pre-clinical xenograft tumor models.
a ET190L1-AbTCR-T cell therapy shows efficacy in a Raji lymphoma model. Bioluminescent images (left panel) and total flux (right panel) over time of Raji-luc implanted mice intravenously administered with 5 × 106 (1) un-transduced donor-matched T cells (Mock), (2) ET190L1-CAR-T cells (CAR), or (3) ET190L1-AbTCR-T cells (AbTCR). Doses were based on number of receptor-positive cells; n= 6–8 mice/group. b ET190L1-AbTCR treated mice resist Raji lymphoma and NALM-6 B-ALL tumor re-challenge. ET190L1-AbTCR and ET190L1-CAR-treated mice that showed no detectable Raji lymphoma were re-challenged with Raji cells (left panel) or NALM-6 cells (right panel). As controls, Raji-naïve mice were implanted with Raji cells (left panel) or NALM-6 cells (right panel) following an injection of Mock T cells; n= 2–3 mice/group. c ET190L1-AbTCR shows anti-tumor activity in a PDX model of CD80/86 negative primary B-ALL (CHP105R1). Total flux overtime of CHP105R1-bearing mice treated with either un-transduced T cells (Mock), CTL019 T cells, or ET190L1-AbTCR T cells; n = 5 mice/group. d ET190L1-AbTCR-T cells release significantly less cytokines than ET190L1-CAR-T cells in vivo. Serum cytokine levels (pg/ml) collected from Raji-bearing mice 24 h after T cell dosing. IL-2, IL-10, IFN-γ and TNF-α levels are shown. e ET190L1-AbTCR-T cells express significantly less PD-1 levels than ET190L1-CAR-T cells in vivo. PD-1 expression levels (mean fluorescent intensity; MFI) on CAR+ CD4+ and AbTCR+ CD4+ T cells at select times from Raji-bearing mice after T cell infusion. *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001.

References

    1. Oluwole OO, Davila ML. At The Bedside: Clinical review of chimeric antigen receptor (CAR) T cell therapy for B cell malignancies. J. Leukoc. Biol. 2016;100:1265–1272. doi: 10.1189/jlb.5BT1115-524R.
    1. Maude SL, et al. Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. N. Engl. J. Med. 2018;378:439–448. doi: 10.1056/NEJMoa1709866.
    1. Gross G, Gorochov G, Waks T, Eshhar Z. Generation of effector T cells expressing chimeric T cell receptor with antibody type-specificity. Transplant. Proc. 1989;21:127–130.
    1. Eshhar Z, Waks T, Gross G, Schindler DG. Specific activation and targeting of cytotoxic lymphocytes through chimeric single chains consisting of antibody-binding domains and the gamma or zeta subunits of the immunoglobulin and T-cell receptors. Proc. Natl. Acad. Sci. USA. 1993;90:720–724. doi: 10.1073/pnas.90.2.720.
    1. Shankaran V, et al. IFNgamma and lymphocytes prevent primary tumour development and shape tumour immunogenicity. Nature. 2001;410:1107–1111. doi: 10.1038/35074122.
    1. Zou C, et al. gammadelta T cells in cancer immunotherapy. Oncotarget. 2017;8:8900–8909.
    1. Guy CS, Vignali DA. Organization of proximal signal initiation at the TCR:CD3 complex. Immunol. Rev. 2009;232:7–21. doi: 10.1111/j.1600-065X.2009.00843.x.
    1. Johnson LA, June CH. Driving gene-engineered T cell immunotherapy of cancer. Cell Res. 2017;27:38–58. doi: 10.1038/cr.2016.154.
    1. Liu H, et al. Targeting alpha-fetoprotein (AFP)-MHC complex with CAR T-cell therapy for liver cancer. Clin. Cancer Res. 2017;23:478–488. doi: 10.1158/1078-0432.CCR-16-1203.
    1. Harris DT, et al. Comparison of T cell activities mediated by human TCRs and CARs that use the same recognition domains. J. Immunol. 2018;200:1088–1100. doi: 10.4049/jimmunol.1700236.
    1. Oren R, et al. Functional comparison of engineered T cells carrying a native TCR versus TCR-like antibody-based chimeric antigen receptors indicates affinity/avidity thresholds. J. Immunol. 2014;193:5733–5743. doi: 10.4049/jimmunol.1301769.
    1. Davenport AJ, et al. Chimeric antigen receptor T cells form nonclassical and potent immune synapses driving rapid cytotoxicity. Proc. Natl. Acad. Sci. U.S.A. 2018;115:E2068–E2076. doi: 10.1073/pnas.1716266115.
    1. Koning F, Maloy WL, Cohen D, Coligan JE. Independent association of T cell receptor beta and gamma chains with CD3 in the same cell. J. Exp. Med. 1987;166:595–600. doi: 10.1084/jem.166.2.595.
    1. Saito T, et al. Surface expression of only gamma delta and/or alpha beta T cell receptor heterodimers by cells with four (alpha, beta, gamma, delta) functional receptor chains. J. Exp. Med. 1988;168:1003–1020. doi: 10.1084/jem.168.3.1003.
    1. Ren J, et al. Multiplex genome editing to generate universal CAR T cells resistant to PD1 inhibition. Clin. Cancer Res. 2017;23:2255–2266. doi: 10.1158/1078-0432.CCR-16-1300.
    1. Ren J, et al. A versatile system for rapid multiplex genome-edited CAR T cell generation. Oncotarget. 2017;8:17002–17011.
    1. Singh N, Perazzelli J, Grupp SA, Barrett DM. Early memory phenotypes drive T cell proliferation in patients with pediatric malignancies. Sci. Transl. Med. 2016;8:320ra323.
    1. Fraietta JA, et al. Biomarkers of response to anti-CD19 chimeric antigen receptor (CAR) T-cell therapy in patients with chronic lymphocytic leukemia. Blood. 2016;128:57–57.
    1. Mahnke YD, Brodie TM, Sallusto F, Roederer M, Lugli E. The who’s who of T-cell differentiation: human memory T-cell subsets. Eur. J. Immunol. 2013;43:2797–2809. doi: 10.1002/eji.201343751.
    1. Long AH, et al. 4-1BB costimulation ameliorates T cell exhaustion induced by tonic signaling of chimeric antigen receptors. Nat. Med. 2015;21:581–590. doi: 10.1038/nm.3838.
    1. Gomes-Silva D, et al. Tonic 4-1BB costimulation in chimeric antigen receptors impedes T cell survival and is vector-dependent. Cell Rep. 2017;21:17–26. doi: 10.1016/j.celrep.2017.09.015.
    1. Kalos M, 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;3:95ra73. doi: 10.1126/scitranslmed.3002842.
    1. Kalos M, June CH. Adoptive T cell transfer for cancer immunotherapy in the era of synthetic biology. Immunity. 2013;39:49–60. doi: 10.1016/j.immuni.2013.07.002.
    1. Teachey DT, et al. Identification of predictive biomarkers for cytokine release syndrome after chimeric antigen receptor T-cell therapy for acute lymphoblastic leukemia. Cancer Discov. 2016;6:664–679. doi: 10.1158/-16-0040.
    1. Grupp SA, et al. Chimeric antigen receptor-modified T cells for acute lymphoid leukemia. N. Engl. J. Med. 2013;368:1509–1518. doi: 10.1056/NEJMoa1215134.
    1. Davila ML, et al. Efficacy and toxicity management of 19-28z CAR T cell therapy in B cell acute lymphoblastic leukemia. Sci. Transl. Med. 2014;6:224ra225. doi: 10.1126/scitranslmed.3008226.
    1. Lee DW, 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. doi: 10.1016/S0140-6736(14)61403-3.
    1. Singh N, et al. Monocyte lineage-derived IL-6 does not affect chimeric antigen receptor T-cell function. Cytotherapy. 2017;19:867–880. doi: 10.1016/j.jcyt.2017.04.001.
    1. Grupp SA. Advances in T-cell therapy for ALL. Best. Pract. Res. Clin. Haematol. 2014;27:222–228. doi: 10.1016/j.beha.2014.10.014.
    1. Nicolas L, et al. Human gammadelta T cells express a higher TCR/CD3 complex density than alphabeta T cells. Clin. Immunol. 2001;98:358–363. doi: 10.1006/clim.2000.4978.
    1. Rothlisberger D, Honegger A, Pluckthun A. Domain interactions in the Fab fragment: a comparative evaluation of the single-chain Fv and Fab format engineered with variable domains of different stability. J. Mol. Biol. 2005;347:773–789. doi: 10.1016/j.jmb.2005.01.053.
    1. Brentjens RJ, et al. Genetically targeted T cells eradicate systemic acute lymphoblastic leukemia xenografts. Clin. Cancer Res. 2007;13:5426–5435. doi: 10.1158/1078-0432.CCR-07-0674.
    1. Milone MC, et al. Chimeric receptors containing CD137 signal transduction domains mediate enhanced survival of T cells and increased antileukemic efficacy in vivo. Mol. Ther. 2009;17:1453–1464. doi: 10.1038/mt.2009.83.
    1. Klapper JA, et al. Single-pass, closed-system rapid expansion of lymphocyte cultures for adoptive cell therapy. J. Immunol. Methods. 2009;345:90–99. doi: 10.1016/j.jim.2009.04.009.
    1. Gardner RA, et al. Intent-to-treat leukemia remission by CD19 CAR T cells of defined formulation and dose in children and young adults. Blood. 2017;129:3322–3331. doi: 10.1182/blood-2016-10-748772.
    1. Barrett DM, et al. Relation of clinical culture method to T-cell memory status and efficacy in xenograft models of adoptive immunotherapy. Cytotherapy. 2014;16:619–630. doi: 10.1016/j.jcyt.2013.10.013.
    1. Gust J, et al. Endothelial activation and blood-brain barrier disruption in neurotoxicity after adoptive immunotherapy with CD19 CAR-T cells. Cancer Discov. 2017;7:1404–1419. doi: 10.1158/-17-0698.
    1. Lee DW, et al. Current concepts in the diagnosis and management of cytokine release syndrome. Blood. 2014;124:188–195. doi: 10.1182/blood-2014-05-552729.
    1. Maude SL, Barrett D, Teachey DT, Grupp SA. Managing cytokine release syndrome associated with novel T cell-engaging therapies. Cancer J. 2014;20:119–122. doi: 10.1097/PPO.0000000000000035.
    1. Jinek M, et al. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science. 2012;337:816–821. doi: 10.1126/science.1225829.
    1. Ran FA, et al. Genome engineering using the CRISPR-Cas9 system. Nat. Protoc. 2013;8:2281–2308. doi: 10.1038/nprot.2013.143.
    1. Sotillo E, et al. Convergence of acquired mutations and alternative splicing of CD19 enables resistance to CART-19 immunotherapy. Cancer Discov. 2015;5:1282–1295. doi: 10.1158/-15-1020.
    1. Barrett DM, et al. Noninvasive bioluminescent imaging of primary patient acute lymphoblastic leukemia: a strategy for preclinical modeling. Blood. 2011;118:e112–e117. doi: 10.1182/blood-2011-04-346528.

Source: PubMed

3
S'abonner