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
. 2024 Jan 15;15(1):519.
doi: 10.1038/s41467-024-44786-2.

Generation and optimization of off-the-shelf immunotherapeutics targeting TCR-Vβ2+ T cell malignancy

Affiliations

Generation and optimization of off-the-shelf immunotherapeutics targeting TCR-Vβ2+ T cell malignancy

Jingjing Ren et al. Nat Commun. .

Abstract

Current treatments for T cell malignancies encounter issues of disease relapse and off-target toxicity. Using T cell receptor (TCR)Vβ2 as a model, here we demonstrate the rapid generation of an off-the-shelf allogeneic chimeric antigen receptor (CAR)-T platform targeting the clone-specific TCR Vβ chain for malignant T cell killing while limiting normal cell destruction. Healthy donor T cells undergo CRISPR-induced TRAC, B2M and CIITA knockout to eliminate T cell-dependent graft-versus-host and host-versus-graft reactivity. Second generation 4-1BB/CD3zeta CAR containing high affinity humanized anti-Vβ scFv is expressed efficiently on donor T cells via both lentivirus and adeno-associated virus transduction with limited detectable pre-existing immunoreactivity. Our optimized CAR-T cells demonstrate specific and persistent killing of Vβ2+ Jurkat cells and Vβ2+ patient derived malignant T cells, in vitro and in vivo, without affecting normal T cells. In parallel, we generate humanized anti-Vβ2 antibody with enhanced antibody-dependent cellular cytotoxicity (ADCC) by Fc-engineering for NK cell ADCC therapy.

PubMed Disclaimer

Conflict of interest statement

J.R., X.L., J.M.L. and M.G are inventors on patent applications filed by Yale University. Application # 63/484,916. The remaining authors declare no competing interests.

Figures

Fig. 1
Fig. 1. Generation of mCAR-T cells targeting the TCR-Vβ2 chain.
a TRBV usage frequency in CTCL cells from 72 patients seen at the Yale Photopheresis Unit from 2016 to 2022 as determined by anti-Vβ antibody staining (Beckman Coulter IOTest Beta Mark) and flow cytometry. b Distribution of TRBV frequency in total CD4 T cells from a Vβ2+ Sézary Syndrome (SS) patient determined by anti-Vβ antibody set staining and flow cytometry (top) or by paired single cell mRNA/TCR sequencing (bottom,). c Live CTCL cell counts from three Vβ2+ and one Vβ13.2 + CTCL patients after overnight in vitro culture with allogeneic mCAR-Vβ2 T cells with (right) or without (left) knockout (KO) of the endogenous T cell receptor alpha constant (TRAC) region at different effector-to-target (E:T) ratios, as determined by flow cytometry. d, e Live fraction of Vβ2+ CTCL cells or normal Vβ2+ cells (red), Vβ2- normal T cells (blue), and non-T cells (black) from PBMC of two Vβ2+ CTCL patients (d) and healthy control (HC)1 (e) at different E:T ratios after overnight co-culture with allogeneic triple KO (TRAC/B2M/CIITA) mCAR-Vβ2 T cells, as determined by flow cytometry. f Live fraction of each detectable Vβ subtype+ cells from total T cells of HC2 at different E:T ratios after overnight co-culture with allogeneic triple KO mCAR-Vβ2 T cells, as determined by flow cytometry. cf N = 3 replicates of each E:T condition. Data are presented as mean values +/− SEM. *p < 0.05, **p < 0.01 and ****p < 0.0001 by two-way ANOVA. All replicates are independent samples. Source data and exact p-values are provided as a Source Data file.
Fig. 2
Fig. 2. Efficient and specific elimination of Vβ2+ malignant T cells in vivo by allogeneic CAR-Vβ2 T cells.
a Schematic of the NSG mouse model used to assess in vivo Vβ2+ T lymphoma killing by mCAR-Vβ2 T cells. Illustrations were created with BioRender.com and Adobe Illustrator. bd Analysis of NSG bone marrow (BM) 10 days post inoculation of Jurkat-TRBV20-1 cells and subsequent treatment with triple KO CD8 mCAR-Vβ2 T cells vs no-treatment control (NC). b Representative flow cytometry quantified in (c, d). c Vβ2+ Jurkat-TRBV20-1 cell count, and (d) mCAR-Vβ2 T cell count with (red) or without (black) mCAR-Vβ2 treatment. el Analysis of NSG mice carrying CD4+ T cells from a Vβ2+ PTCL patient 1 week post treatment with triple KO CD8 mCAR-Vβ2 T cells vs no-treatment control (NC). e Representative NSG spleen cell flow cytometry quantified in (fh) showing Vβ2+ PTCL cells, Vβ2-negative normal T cells and CD8 mCAR-Vβ2. f Vβ2+ PTCL cell count, (g) Vβ2-negative normal CD4+ T cell count, and (h) mCAR-Vβ2 T cell count in spleen with (red) or without (black) mCAR-Vβ2 treatment. i Representative NSG BM flow cytometry quantified in (jl) showing Vβ2+ PTCL cells, Vβ2-negative normal T cells and CD8 mCAR-Vβ2. j Vβ2+ CTCL cell count (k) Vβ2- normal CD4+ T cell count, and (l) mCAR-Vβ2 T cell count in BM with (red) or without (black) mCAR-Vβ2 treatment. cl N = 3 mice per group. Data are presented as mean values +/− SEM. *p < 0.05 and **p < 0.01 by two-sided t-test. Source data and exact p-values are provided as a Source Data file.
Fig. 3
Fig. 3. Humanization of CAR-Vβ2 T cells with preserved efficacy.
a, b Representative flow cytometry of (a) BM and (b) spleen, showing CD3+ Vβ2+ CTCL cells, CD3+ Vβ2-negative normal T cells and CD3-negative hCAR-Vβ2 T cells 3 days after in vivo hCAR-Vβ2 treatment, nonspecific CAR-CD19 treatment, or no-treatment control (NC). c Vβ2+ CTCL cells (d) Vβ2-negative normal T cells (e) CD8 hCAR-T and (f) CD4 hCAR-T cells in NSG BM 3 days after in vivo allogeneic triple-KO hCAR-Vβ2 (red) or CAR-CD19 (blue) pan-T cell treatment generated from a healthy donor compared to no-treatment control (NC, black). g Vβ2+ CTCL cell counts (h) Vβ2-negative normal T cell counts (i) CD8 CAR-T cell counts and (j) CD4 CAR-T cell count in spleen of the same mice. cj N = 3 mice per group. Data are presented as mean values +/− SEM. *p < 0.05, **p < 0.01 and ***p < 0.001 by one-way ANOVA. Source data and exact p-values are provided as a Source Data file.
Fig. 4
Fig. 4. CRISPR-AAV system for CAR-Vβ2 T cell generation and treatment of Vβ2+ PTCL PDX.
a Adeno-associated virus (AAV) chimeric antigen-receptor (CAR) template structure and the strategy to integrate into the TRAC region using Cas9-TRAC-sgRNA. b Representative flow cytometry showing CD3- purity, CD4 and CD8 population percentages and CAR expression of AAV-dependent allogeneic hCAR-Vβ2 T cells compared to lentiviral-dependent allogeneic CAR-CD19 T cells. c Live PTCL/CTCL cell counts from two Vβ2+ patients and (d) live Jurkat-TRBV20-1 (Vβ2+, left) or Jurkat-TRBV6-2 (Vβ13.2+, right) cell counts after overnight in vitro culture with allogeneic lenti-CAR-CD19 T cells (black) or AAV-hCAR-Vβ2 T cells (red) at different E:T ratios, determined by flow cytometry. ek Total CD4 T cells isolated from a Vβ2+ PTCL patient were adoptively transferred into groups of NSG mice that were then treated with allogeneic triple-KO AAV-hCAR-Vβ2 (red) or lenti-CAR-CD19 (blue) generated from healthy donor pan T cells, compared to no-treatment control (NC, black). Three days post-treatment (e) Vβ2+ CTCL cells (f) Vβ2- normal T cells (g) CD69+ % in CD8 CAR-T cells, and (h) CD69+ % in CD4 CAR-T cells in spleen were quantified by flow cytometry, as were (i) Vβ2+ CTCL cells (j) CD69+ % in CD8 CAR-T cells, and (k) CD69+ % in CD4 CAR-T cells in BM. lo Long-term bioluminescence monitoring (IVIS) of Jurkat-TRBV20-1-lucifer cell-bearing NSG mice, at (ln) ventral or (mo) dorsal position, without treatment (NC, black) or following treatment with CAR-CD19 T cells (blue), lenti-hCAR-Vβ2 T cells (purple) or AAV-hCAR-Vβ2 T cells (red). p, Body weight and (q) survival of the same mice. c, d n = 3 replicates in each group. ***p < 0.001 and ****p < 0.0001 by two-way ANOVA. e-k, n = 3 mice in each group. *p < 0.05, **p < 0.01 and ***p < 0.001 by one-way ANOVA. nq n = 5 mice in each group. n, o *p < 0.05, **p < 0.01, ***p < 0.001 and ****p < 0.0001 by two-way ANOVA. q *p < 0.05 and **p < 0.01 by survival analysis (Kaplan-Meier). cknq Data are presented as mean values +/− SEM. All replicates are independent samples. Source data and exact p-values are provided as a Source Data file.
Fig. 5
Fig. 5. Cytokine optimization for hCAR-Vβ2 T cell expansion in vitro.
a Schematic of the timeline of cytokine optimization and functional analysis. b Live total T cell counts during in vitro expansion of hCAR-Vβ2 T cells from three different donors, determined by trypan blue cell counting. c Jurkat-TRBV20-1 cell killing % and (d) live CD3- hCAR-Vβ2 T cell counts in acute repeated Jurkat-TRBV20-1 killing assay, determined by trypan blue cell counting and flow cytometry. e CD45RA+ CD45RO+ % of CD8 + CD3- hCAR-Vβ2 T cells 4 days post repeated Jurkat-TRBV20-1 cell killing assay, determined by flow cytometry. be N = 3 replicates in each group. Data are presented as mean values +/− SEM. f relative Jurkat-TRBV20-1 killing by chronically stimulated hCAR-Vβ2 T cells from two healthy donors generated in seven cytokine conditions, with each killing round normalized to the first round. g summary of optimized cytokine combinations for allogeneic hCAR-Vβ2 T cell expansion in vitro. Illustrations in Fig. 5a, g were created using Adobe Illustrator. All replicates are independent samples. Source data are provided as a Source Data file.
Fig. 6
Fig. 6. Humanized anti-Vβ2 therapeutic antibody with enhanced ADCC.
a An anti-Vβ2 antibody competition assay, in which 1.25μg/ml mouse anti-Vβ2-FITC antibody was mixed with varying concentrations of humanized anti-Vβ2-PE antibody and used to stain CTCL cells from a Vβ2+ patient, with Vβ2 MFI determined by flow cytometry. b ADCC assay luminescence signal after a 6 h co-culture of patient derived Vβ2+ target CTCL cells and Jurkat-NFAT-luciferase effector cells mixed with either 100 ng/ml mouse anti-Vβ2 antibody (black) or humanized anti-Vβ2 antibody (red). PBMC from Vβ2+ CTCL patient 1 (ce) or from Vβ2+ CTCL patient 2 (fh) were cultured overnight with NK effector cells from a healthy donor at different E:T ratios without antibody addition (black) or with addition of mouse anti-Vβ2 antibody (blue) or humanized anti-Vβ2 antibody (red), then live CTCL cells (cf) Vβ2- normal T cells (dg) and non-T cell PBMC (eh) counts were determined by flow cytometry. ch N = 3 replicates in each group. Data are presented as mean values +/− SEM. ***p < 0.001 and ****p < 0.0001 by two-way ANOVA. All replicates are independent samples. Source data and exact p-values are provided as a Source Data file.

Similar articles

References

    1. Safarzadeh Kozani P, Safarzadeh Kozani P, Rahbarizadeh F. CAR-T cell therapy in T-cell malignancies: is success a low-hanging fruit? Stem Cell Res. Ther. 2021;12:527. doi: 10.1186/s13287-021-02595-0. - DOI - PMC - PubMed
    1. Neelapu SS, et al. Axicabtagene Ciloleucel CAR T-cell therapy in refractory large B-cell Lymphoma. N. Engl. J. Med. 2017;377:2531–2544. doi: 10.1056/NEJMoa1707447. - DOI - PMC - PubMed
    1. Schuster SJ, et al. Chimeric antigen receptor T cells in refractory B-cell lymphomas. N. Engl. J. Med. 2017;377:2545–2554. doi: 10.1056/NEJMoa1708566. - DOI - PMC - PubMed
    1. Ellebrecht CT, et al. Reengineering chimeric antigen receptor T cells for targeted therapy of autoimmune disease. Science. 2016;353:179–184. doi: 10.1126/science.aaf6756. - DOI - PMC - PubMed
    1. Mackensen A, et al. Anti-CD19 CAR T cell therapy for refractory systemic lupus erythematosus. Nat. Med. 2022;28:2124–2132. doi: 10.1038/s41591-022-02017-5. - DOI - PubMed

Substances