Chimeric Antigen Receptor T Cells Guided by the Single-Chain Fv of a Broadly Neutralizing Antibody Specifically and Effectively Eradicate Virus Reactivated from Latency in CD4+ T Lymphocytes Isolated from HIV-1-Infected Individuals Receiving Suppressive Combined Antiretroviral Therapy

Bingfeng Liu, Fan Zou, Lijuan Lu, Cancan Chen, Dalian He, Xu Zhang, Xiaoping Tang, Chao Liu, Linghua Li, Hui Zhang, Bingfeng Liu, Fan Zou, Lijuan Lu, Cancan Chen, Dalian He, Xu Zhang, Xiaoping Tang, Chao Liu, Linghua Li, Hui Zhang

Abstract

Despite the advent of combined antiretroviral therapy (cART), the persistence of viral reservoirs remains a major barrier to curing human immunodeficiency virus type 1 (HIV-1) infection. Recently, the shock and kill strategy, by which such reservoirs are eradicated following reactivation of latent HIV-1 by latency-reversing agents (LRAs), has been extensively practiced. It is important to reestablish virus-specific and reliable immune surveillance to eradicate the reactivated virus-harboring cells. In this report, we attempted to reach this goal by using newly developed chimeric antigen receptor (CAR)-T cell technology. To generate anti-HIV-1 CAR-T cells, we connected the single-chain variable fragment of the broadly neutralizing HIV-1-specific antibody VRC01 to a third-generation CAR moiety as the extracellular and intracellular domains and subsequently transduced this into primary CD8+ T lymphocytes. We demonstrated that the resulting VC-CAR-T cells induced T cell-mediated cytolysis of cells expressing HIV-1 Env proteins and significantly inhibited HIV-1 rebound after removal of antiviral inhibitors in a viral infectivity model in cell culture that mimics the termination of the cART in the clinic. Importantly, the VC-CAR-T cells also effectively induced the cytolysis of LRA-reactivated HIV-1-infected CD4+ T lymphocytes isolated from infected individuals receiving suppressive cART. Our data demonstrate that the special features of genetically engineered CAR-T cells make them a particularly suitable candidate for therapeutic application in efforts to reach a functional HIV cure.

Importance: The presence of latently infected cells remains a key obstacle to the development of a functional HIV-1 cure. Reactivation of dormant viruses is possible with latency-reversing agents, but the effectiveness of these compounds and the subsequent immune response require optimization if the eradication of HIV-1-infected cells is to be achieved. Here, we describe the use of a chimeric antigen receptor, comprised of T cell activation domains and a broadly neutralizing antibody, VRC01, targeting HIV-1 to treat the infected cells. T cells expressing this construct exerted specific cytotoxic activity against wild-type HIV-1-infected cells, resulting in a dramatic reduction in viral rebound in vitro, and showed persistent effectiveness against reactivated latently infected T lymphocytes from HIV-1 patients receiving combined antiretroviral therapy. The methods used in this study constitute an improvement over existing CD4-based CAR-T technology and offer a promising approach to HIV-1 immunotherapy.

Copyright © 2016, American Society for Microbiology. All Rights Reserved.

Figures

FIG 1
FIG 1
Characterization of VRC01-28BBZ CAR-T cells. (A) Schematic representation of the VRC01-28BBZ CAR (not to scale). (B) Western blotting was performed to detect the expression of VRC01-28BBZ with anti-Flag tag antibody in HEK293T cells by transfecting three pCPPT-IRES-VRC01-28BBZ-mStrawberry plasmids. The HEK293T cells transfected with pCPPT-IRES-mStrawberry empty vector served as a negative control. (C) Immunofluorescence staining was performed to detect VRC01-28BBZ-3 CAR (VC-CAR) expression in HeLa cells with anti-Flag tag antibody. DAPI staining represents the nuclei. (D) Flow cytometry was performed to detect the transduction efficiencies of VC-CAR and empty vector in human CD8+ T cells by detection of mStrawberry fluorescence. Transduced CD8+ T cells were measured by detecting mStrawberry fluorescence and human Fab stained with goat antibody through flow cytometry. Untransduced CD8+ T cells served as a negative control. The CD4-CAR-transduced CD8+ T cells were detected with anti-CD4 antibody through flow cytometry. CD4-negative gating was set on untransduced control cells (data not shown).
FIG 2
FIG 2
Selection of the most efficient CAR moiety. (A) qRT-PCR was performed to characterize the expression of HIV-1 Env and eGFP in Jurkat-based target cells. Jurkat cells served as a negative control. (B) Cytotoxicity assays were performed using the CytoTox nonradioactive cytotoxicity kit, using as targets (T) Jurkat-gp160NL4-3 cells constitutively expressing Env from HIV-1NL4-3. Cocultures were performed for 24 h with T cells expressing the indicated CARs (E, effector cells); the RFP-CD8+ T cells (Mock) served as a negative control. Shown are the cytotoxic effects of VRC01-28BBZ-transduced T cells on target cells at 2:1 and 1:1 (E:T) ratios for 24 h. (C) Characterization of VRC01-28BBZ-transduced effector CD8+ T cells after coculture with Jurkat-gp160NL4-3 or Jurkat-GFP cells at a 4:1 (E:T) ratio for 20 h by IFN-γ ELISpot assay. Effector cells alone served as the negative control (NC). The PHA-stimulated effector cells served as the positive control (PC). These data represent three independent experiments. (D) Cytotoxic effect of VC-CAR-T cells and CD4-CAR-T cells on Jurkat-gp160NL4-3 at the indicated range of E:T ratios (24 h). (E) The CD8+ T cells expressing VC-CAR were challenged with cell-free HIV-1NL4-3 (p24 titer of 200 ng ml−1). Infection was analyzed at the indicated time points by staining for intracellular p24 after gating the CD3+ CD8+ subpopulation. Untransduced CD8+ T cells and CD8+ T cells transduced with empty vector (mock) served as a negative control, and CD4+ T cells (gate CD3+ CD8−) served as a positive control. The percent value in each histogram (left three panels) indicates the p24-positive population. (A, B, and D) Data reflect means ± standard errors of the means (SEM), and P values were calculated using the two-tailed unpaired Student's t test with equal variances (n = 3). *, P < 0.05; **, P < 0.01; ***, P < 0.001. Data shown in panel E represent two independent experiments.
FIG 3
FIG 3
Cytokine release of VC-CAR-T cells after coculture with Env-expressing Jurkat target cells. (A) Characterization of VC-CAR or control transduced effector CD8+ T cells cocultured with Jurkat-gp160NL4-3 or Jurkat-GFP cells at 2:1 and 4:1 (E:T) ratios for 20 h by IFN-γ ELISpot assay. The PHA-stimulated effector cells served as the positive control (PC). Effector cells alone served as the negative control (NC). (B) Summary of IFN-γ ELISpot assays described for panel A. The count number of the y axis was measured as spot-forming cells (SFCs)/104 CD8+ T cells. (C) Characterization of VC-CAR or control transduced effector CD8+ T cells cocultured with Jurkat-gp160BaL cells. (D) Summary of IFN-γ ELISpot assay described for panel C. The count number of the y axis was measured as SFCs/104 CD8+ T cells. (E) Granzyme B production in cocultures of Jurkat-gp160NL4-3 target cells with VC-CAR-transduced effector CD8+ T cells at the indicated E:T ratios for 20 h. The effector cells expressing red fluorescent protein served as negative controls. (F) IL-2 production from triple VC-CAR or control effector CD8+ T cells cocultured with Jurkat-gp160NL4-3 cells for 18 h. (B, E, and F) Data reflects means ± SEM. P values were calculated using the two-tailed unpaired Student's t test with equal variances (n = 3). *, P < 0.05; **, P < 0.01; ***, P < 0.001.
FIG 4
FIG 4
Specific killing of Env-expressing cells by VC-CAR-transduced effector CD8+ T cells. Direct killing of target cell lines was performed using the CytoTox nonradioactive cytotoxicity kit with Jurkat-gp160NL4-3 (A), Jurkat-gp160BaL (B), and Jurkat-GFP (C) cells as targets. Cocultures were performed for 24 h with CD8+ T cells expressing the VC-CAR (E, effector cells); the red fluorescent protein-transduced CD8+ T cells served as a negative control. Data reflect means ± SEM.
FIG 5
FIG 5
Specific killing of HIV-1NL4-3-infected primary CD4+ T cells by VC-CAR-transduced effector CD8+ T cells. (A) Primary CD4+ T cells were infected with HIV-1NL4-3 (p24 titer of 200 ng ml−1). At day 8 postinfection, CD4+ T cells were mixed with autologous VC-CAR or control transduced effector CD8+ T cells at a 1:2 or 1:4 ratio. Seventy-two hours after coculture, specific cytotoxicity was analyzed by flow cytometry for the intracellular staining of Gag+ T cells gated on CD3+ CD8− or CD3+ CD8+ subpopulations, respectively, as indicated at the top. (B) Summary of residual CD3+ CD8− Gag+ T cells from the flow cytometry analysis. (C) VC-CAR or control transduced effector CD8+ T cells were cocultured with HIV-1NL4-3-infected primary CD4+ T cells at a 2:1 (E:T) ratio for 20 h. IFN-γ secretion was analyzed by ELISpot assay. The PHA-stimulated effector cells served as the positive control (PC). Effector cells alone served as the negative control (NC). (D) Specific killing of HIV-1NL4-3-infected primary CD4+ T cells by VC-CAR-transduced effector CD8+ T cells. Cocultures were performed for 24 h. (B and D) Data reflect means ± SEM from triplicates. P values were calculated using the two-tailed unpaired Student's t test with equal variances (n = 3). *, P < 0.05; **, P < 0.01; ***, P < 0.001.
FIG 6
FIG 6
Effective suppression of HIV-1 rebound after the withdrawal of antiviral treatment in vitro. The primary CD4+ T cells were infected with HIV-1NL4-3 (1 ng ml−1 p24). (A) Experimental design. (B) The expression of the activation markers CD25 and HLA-DR on unstimulated and activated CD4+ T cells and CD4+ T cells at day 14 postinfection. The percentage of cells in each quadrant is indicated. (C) After the withdrawal of antiviral drugs, 0.5 × 106 CD4+ T cells were mixed with autologous VC-CAR engineered or control CD8+ T cells at a 1:2 or 1:4 ratio. On day 24, cells were collected and viral RNAs were isolated and amplified by real-time RT-qPCR with primer SK38 and SK39. The cutoff for cell-associated viral RNA is 800 copies ml−1. (D) Every 2 days the cultures were tested for the presence of p24 in the supernatant by ELISA. The dotted line represents the addition of antiviral drugs. (C and D) Data reflect means ± SEM. P values were calculated using the two-tailed unpaired Student's t test with equal variances (n = 3). *, P < 0.05; **, P < 0.01; ***, P < 0.001.
FIG 7
FIG 7
Elimination of reactivated HIV-1 latently infected CD4+ T lymphocytes from HIV-1-infected individuals receiving suppressive cART. (A) Experimental design. (B) On day 0, CD4+ T cells from HIV-1-infected patients were stimulated by PHA-M or by a combination of specific LRAs (SAHA plus bryostatin-1). After 1 day, 1 × 106 CD4+ T cells were mixed with autologous VC-CAR-engineered or control CD8+ T cells at a 1:1 ratio. On day 3, cells were collected and viral RNAs were isolated and amplified by real-time qRT-PCR with primers SK38 and SK39. The cutoff for cell-associated viral RNA is 800 copies ml−1. (C) The cultures were further tested for the presence of p24 with viral outgrowth by ELISA at the indicated time points. (B and C) Data reflect means ± SEM. P values were calculated using the two-tailed unpaired Student's t test with equal variances (n = 3). *, P < 0.05; **, P < 0.01; ***, P < 0.001.

Source: PubMed

3
購読する