Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2018 Jul 12;3(13):e99442.
doi: 10.1172/jci.insight.99442.

TCRαβ/CD3 disruption enables CD3-specific antileukemic T cell immunotherapy

Affiliations

TCRαβ/CD3 disruption enables CD3-specific antileukemic T cell immunotherapy

Jane Rasaiyaah et al. JCI Insight. .

Abstract

T cells engineered to express chimeric antigen receptors (CARs) against B cell antigens are being investigated as cellular immunotherapies. Similar approaches designed to target T cell malignancies have been hampered by the critical issue of T-on-T cytotoxicity, whereby fratricide or self-destruction of healthy T cells prohibits cell product manufacture. To date, there have been no reports of T cells engineered to target the definitive T cell marker, CD3 (3CAR). Recent improvements in gene editing now provide access to efficient disruption of such molecules on T cells, and this has provided a route to generation of 3CAR, CD3-specific CAR T cells. T cells were transduced with a lentiviral vector incorporating an anti-CD3ε CAR derived from OKT3, either before or after TALEN-mediated disruption of the endogenous TCRαβ/CD3 complex. Only transduction after disrupting assembly of TCRαβ/CD3 yielded viable 3CAR T cells, and these cultures were found to undergo self-enrichment for 3CAR+TCR-CD3- T cells without any further processing. Specific cytotoxicity against CD3ε was demonstrated against primary T cells and against childhood T cell acute lymphoblastic leukemia (T-ALL). 3CAR T cells mediated potent antileukemic effects in a human/murine chimeric model, supporting the application of cellular immunotherapy strategies against T cell malignancies. 3CAR provides a bridging strategy to achieve T cell eradication and leukemic remission ahead of conditioned allogeneic stem cell transplantation.

Keywords: Cell Biology; Gene therapy; Immunology; T cells.

PubMed Disclaimer

Conflict of interest statement

Conflict of interest: IP filed for 3CAR N409687GB (JR, CG, UM, and WQ). Research funding received for unrelated projects from Miltenyi (UM and WQ), Autolus, Cellectis, and Servier (all to WQ). WQ has founder equity Autolus, Orchard Therapeutics.

Figures

Figure 1
Figure 1. Successful generation of 3CAR requires disruption of CD3ε expression prior to lentiviral 3CAR transduction.
(A) Vector configuration showing codon optimized single-chain variable fragment (scFv) derived from OKT3 with CD8 stalk and 41BB/CD3ζ activation domains, all under the control of an internal human phosphoglycerate kinase (hPGK) promoter in a third-generation self-inactivating (SIN) lentivirus with HIV-1–derived reverse response element (RRE), central polypurine tract (cPPT), and mutated woodchuck hepatitis virus posttranscriptional regulatory element (WPRE). Human cytomegalovirus (CMV); Repeat region (R); Unique 5’ region (U5); Psi (Ψ); and modified unique 3’ region (ΔU3). (B) Percentage of CAR+ cells with and without TRAC-KO prior to lentivirus (LV). Data are presented as mean ± SEM. n = 3 donors. (C) Schema of event precedence required for successful 3CAR T cell production. CD3/CD28 activation with TransAct for 48 hours was followed by electroporation of mRNA encoding TRAC-specific TALENs ahead of LV transduction by 72 hours. (D) Summary of CD3+ cells following TRAC-KO (n = 3 donors), mean ± SEM. ***P < 0.0005, by unpaired, 2-tailed Student’s t test. (E) Representative flow cytometry plots of day 7 cells. In cultures with 3CAR expression, there were no surviving CD3+ cells compared with 20% residual expression in the absence of 3CAR transduction (n = 3 donors). (F) Representative flow cytometry plots showing 3CAR T cells retained CD4 and CD8, but not TCRαβ, expression; n = 3 donors. (G) For comparison, control transductions with LV CAR19 mediated 65% transduction but required further processing by column-mediated TCRαβ depletion to yield TRAC-KO CAR19+ T cells; n = 3 donors. (H) Summary of CAR+ cells following TRAC-KO (n = 3 donors); mean ± SEM. Two-tailed Student’s t test.
Figure 2
Figure 2. 3CAR T cell expansion and self-enrichment.
(A) Primary T cells were activated, and following TALEN TRAC mRNA exposure, 2 × 106 T cells were transduced and expanded in a G-Rex flask over 17 days; n = 2 donors. (B) Representative expression of CD3/TCR in TRAC-KO untransduced (top panel) and 3CAR T cells (lower panel) on day 7 and 13, confirming the absence of CD3/TCRαβ cells after 3CAR lentiviral expression; n = 3 donors. (C) Tracking indels by decomposition PCR (TIDE-PCR) confirmed 62.9% of alleles harbored molecular signatures of nonhomologous end joining across the TRAC locus (n = 2). (D) Representative flow cytometry plots at end of manufacture for CD20 and CD56 expression and exhaustion markers PD-1, LAG-3, and TIM-3 (gated on CAR+ and CAR populations); n = 3 donors. (E) The percentage of memory T cell markers on expanded day 17 cells; n = 3 donors, mean ± SEM.
Figure 3
Figure 3. 3CAR T cells mediate potent killing of CD3+ cells in vitro.
(A) TCR+ and TCR Jurkat target cells were stained for TCR expression prior to coculture experiments. (B) 51Cr-labeled CD3+TCR+ (white symbols) or CD3TCR (black symbols) Jurkat leukemia cells were cocultured with either 3CAR T cells (solid lines) or untransduced controls (dotted lines); (n = 3) from 3 donors. (C) Upper panel, 3CAR T cells were cocultured with either GFP+CD3+TCR+ or GPP+CD3TCR Jurkat leukemia cells and, in the lower panel, cocultured with either CFSE-loaded primary CD3+ or CD3 cells at the effector target ratio of 1:1 for 24 hours. Representative flow cytometry plots of gated on GFP+ or CFSE+ cells at the end of coculture demonstrate target-specific killing. (D) Summary of 3CAR cytotoxicity as in C where data are presented as mean ± SEM; n = 3 per group. ***P < 0.0005, by unpaired, 2-tailed Student’s t test.
Figure 4
Figure 4. 3CAR T cell functionality against healthy donor PBMC.
(A) 51Cr-labeled peripheral blood mononuclear cell (PBMC) in autologous (dotted lines) or allogenic (solid lines) coculture. PBMC were cultured with either 3CAR T cells (white symbols) or untransduced controls (black symbols) (n = 3). (B) 3CAR, CAR19, or untransduced T cells were cocultured with CSFE-loaded healthy donor PBMC at the effector target ratio of 1:1 for 24 hours. Top panel shows representative frequency of gated CSFE+ target cells at the end of coculture. Flow cytometry plots below show representative frequency of surface antigen markers CD3, CD19, and CD7 on gated CSFE+ tumor cells, displaying the specificity in mixed cell populations; n = 3 donors. (C) Cytokine production by 3CAR T cells when cocultured with healthy donor PBMC target cells were comparable with untransduced cells; n = 3 donors. (D) Mixed lymphocyte reactions of 3CAR or untransduced T cells cultured with irradiated (*) allogeneic PBMC or CD3 cells. 3H-thymidine proliferation responses are consistent with specific targeting of CD3 rather than alloreactive proliferation); mean ± SEM, 4 experimental replicates (n = 2 donors). #P < 0.05, by unpaired, 2-tailed Student’s t test. Corrected counters per minute, CCPM.
Figure 5
Figure 5. 3CAR T cells specifically target CD3 in childhood T-ALL patient cells.
(A) Representative flow cytometry plots gated on CSFE+ T-ALL tumor cells. Lower panel shows representative frequency of surface antigen CD3 and CD19 of gated CSFE+ tumor cells. (B) Percentage of CD3+ (n = 6) or CD19+ (n = 4) cells as in A from T-ALL donors. *P < 0.05, by unpaired, 2-tailed Student’s t test. (C) 3CAR (lower panels) or untransduced T cells (upper panels) were cocultured with CSFE-loaded T-ALL patient donors at the effector target ratio of 1:1 for 24 hours. Flow cytometry plots show the frequency of CD3 and CD34 cells gated on CSFE+ T-ALL tumor cells. Four of 6 patient samples were positive for CD34 expression.
Figure 6
Figure 6. Antileukemic responses by 3CAR effector T cells against CD3+TCR+ GFP/luciferase human leukemia in immunodeficient mice.
(A) Serial bioluminescence imaging (BLI) of NSG mice (representative images from each cohort) following i.p. administration of D-luciferin substrate showing elimination of CD3+ leukemia by 3CAR T cells but not by untransduced T cells. (B) Kinetics of systemic leukemia progression in mice following administration PBS (n = 2), untransduced T cells (n = 4), or 3CAR (n = 5) effectors. Error bars represent median with interquartile range. Linear regression analysis showed significance between 3CAR vs. untransduced (****P < 0.0001) and 3CAR vs. PBS (****P < 0.0001) groups. (C) Average radiance values at termination (day 18) indicated significant difference in disease burden between untransduced and 3CAR effector–injected groups (*P = 0.0159 by Mann-Whitney U test). Error bars represent median with interquartile range. (D) Upper panel, representative flow cytometry plots of T cells (gated on hCD45+ cells) in BM. Middle panel, plots of leukemic T cell (CD3+TCR+GFP+ Jurkat targets) after effector challenge (gated on hCD45+CD2+ cells). Lower panel, plots of CAR T cells population (gated on hCD45+CD2+GFP expression). (E) Proportion of GFP+ Jurkat leukemia or GFP effector T cells out of CD45+CD2+ population in BM of untransduced or 3CAR-injected mice. Red marking highlights an animal that exhibited selective outgrowth of GFP+CD3 Jurkat cells after 3CAR therapy.

References

    1. Goldberg JM, et al. Childhood T-cell acute lymphoblastic leukemia: the Dana-Farber Cancer Institute acute lymphoblastic leukemia consortium experience. J Clin Oncol. 2003;21(19):3616–3622. doi: 10.1200/JCO.2003.10.116. - DOI - PubMed
    1. Rivière I, Sadelain M. Chimeric Antigen Receptors: A Cell and Gene Therapy Perspective. Mol Ther. 2017;25(5):1117–1124. doi: 10.1016/j.ymthe.2017.03.034. - DOI - PMC - PubMed
    1. Sadelain M, Brentjens R, Rivière I. The basic principles of chimeric antigen receptor design. Cancer Discov. 2013;3(4):388–398. doi: 10.1158/2159-8290.CD-12-0548. - DOI - PMC - PubMed
    1. Poirot L, et al. Multiplex Genome-Edited T-cell Manufacturing Platform for “Off-the-Shelf” Adoptive T-cell Immunotherapies. Cancer Res. 2015;75(18):3853–3864. doi: 10.1158/0008-5472.CAN-14-3321. - DOI - PubMed
    1. Qasim W, et al. Molecular remission of infant B-ALL after infusion of universal TALEN gene-edited CAR T cells. Sci Transl Med. 2017;9(374):eaaj2013. doi: 10.1126/scitranslmed.aaj2013. - DOI - PubMed

Publication types