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
. 2020 Nov 24;4(22):5702-5715.
doi: 10.1182/bloodadvances.2020002229.

Combining T-cell-specific activation and in vivo gene delivery through CD3-targeted lentiviral vectors

Affiliations

Combining T-cell-specific activation and in vivo gene delivery through CD3-targeted lentiviral vectors

Annika M Frank et al. Blood Adv. .

Abstract

Genetic modification of T lymphocytes is a key issue in research and therapy. Conventional lentiviral vectors (LVs) are neither selective for T cells nor do they modify resting or minimally stimulated cells, which is crucial for applications, such as efficient in vivo modification of T lymphocytes. Here, we introduce novel CD3-targeted LVs (CD3-LVs) capable of genetically modifying human T lymphocytes without prior activation. For CD3 attachment, agonistic CD3-specific single-chain variable fragments were chosen. Activation, proliferation, and expansion mediated by CD3-LVs were less rapid compared with conventional antibody-mediated activation owing to lack of T-cell receptor costimulation. CD3-LVs delivered genes not only selectively into T cells but also under nonactivating conditions, clearly outperforming the benchmark vector vesicular stomatitis-LV glycoproteins under these conditions. Remarkably, CD3-LVs were properly active in gene delivery even when added to whole human blood in absence of any further stimuli. Upon administration of CD3-LV into NSG mice transplanted with human peripheral blood mononuclear cells, efficient and exclusive transduction of CD3+ T cells in all analyzed organs was achieved. Finally, the most promising CD3-LV successfully delivered a CD19-specific chimeric antigen receptor (CAR) into T lymphocytes in vivo in humanized NSG mice. Generation of CAR T cells was accompanied by elimination of human CD19+ cells from blood. Taken together, the data strongly support implementation of T-cell-activating properties within T-cell-targeted vector particles. These particles may be ideally suited for T-cell-specific in vivo gene delivery.

PubMed Disclaimer

Conflict of interest statement

Conflict-of-interest disclosure: E.V., I.C.S., and C.J.B are listed as inventors on patents on receptor-targeted LVs that have been licensed to the patent commercializing agencies ipal GmbH and Pulsalys. The remaining authors declare no competing financial interests.

Figures

None
Graphical abstract
Figure 1.
Figure 1.
CD3-LVs transduce T cells independently of activation or cytokine treatment. (A-B) Primary human PBMCs isolated from different blood donations were activated with αCD3 and αCD28 antibodies for 3 days in the presence of IL-2 before they were incubated with CD3-LVs or VSV-LV. Green fluorescent protein (GFP) expression was determined 6 days later by flow cytometry. (A) Representative dot plots show GFP expression in CD4+ and CD4 populations gated for viable cells. (B) Scatter bar diagrams summarize the percentages of GFP+ cells as mean ± standard deviation (SD) of 3 independent experiments with 2-3 donors and 2-3 technical replicates. (C-D) Freshly isolated PBMCs were cultured overnight in presence of IL-2 or IL-7/IL-15, or in absence of cytokines, before they were transduced with CD3-LVs or VSV-LV. GFP expression was determined 6 days later by flow cytometry. (C) Scatter bar diagrams show the percentage of GFP+ cells in all viable single cells. (D) Representative dot plots show GFP expression in the CD8+ and CD8 populations gated for viable cells. Data are mean ± SD from 1 experiment with n = 3 technical replicates; biological replicates from additional experiments with IL-2- and IL-7/IL-15-stimulated cells are shown in supplemental Figure 6C-F. **P < .01, ***P < .001, ****P < .0001 by 2-way analysis of variance (ANOVA) with Dunnett's correction (comparison with VSV-LV for each condition), or unpaired Student t test.
Figure 2.
Figure 2.
CD3-LVs activate cytokine-cultured T cells and induce cytokine production and proliferation. PBMCs isolated from adult blood were cultured overnight in the presence of IL-2 only and then transduced with the indicated CD3-LVs or with VSV-LV. Controls were left untransduced (ut) or activated with αCD3/αCD28 antibodies (1 µg/mL αCD3, 3 µg/ml αCD28) until first medium exchange. (A-B) Expression of the activation markers CD69 (A) and CD25 (B) on all viable cells was followed for 6 days by flow cytometry. The number of activation marker positive cells are shown. N = 3 donors, mean ± standard error of the mean. *P < .05, **P < .01, ***P < .001, ****P < .0001 by 2-way ANOVA with Dunnett's correction. (C-E) One day postincubation with vector particles or recombinant antibodies, cytokines secreted into the cell culture supernatant were quantified. Scatter bar diagrams show the concentration of interferon-γ (C), tumor necrosis factor-α (D), and IL-2 (E) for each condition. Mean ± SD from 1 experiment with n = 3 triplicates each are shown. (F) PBMCs were stained with CellTrace Violet (CTV) before transduction ± costimulation to follow cell proliferation over time. Histograms show the fluorescence of CTV at day 5 posttransduction. Data are representative of 3 different donors. (G) Bar diagrams display T-cell expansion after 6 days compared with day 0 ± costimulation. N = 3 donors, mean ± SD. ns, nonsignificant; *P < .05 by 1-way ANOVA with Dunnett's correction. (H) Bar diagrams show percentages of GFP+ cells gated from all viable single cells ± costimulation at day 6 posttransduction. N = 3 donors. Mean ± SD. ns, nonsignificant by 2-way ANOVA with Sidak's correction.
Figure 3.
Figure 3.
Endocytosis of the TCR/CD3 complex upon transduction. (A-B) Downmodulation of CD3 (A) and TCR (B) on PBMC stimulated only with IL-2 and transduced with CD3-LVs (in absence of Vectofusin-1), VSV-LV, or left untransduced but activated (αCD3/αCD28). The mean fluorescence intensities measured were normalized to those of untreated nonactivated cells. N = 3 donors, mean ± standard error of the mean. *P < .05, **P < .01, ***P < .001 by 2-way ANOVA with Dunnett's correction. (C-D) Inhibition of endocytosis with Bafilomycin A1 (Bfla1) (C) or NH4Cl (D) increases transduction efficiency on Jurkat cells with CD3-LVs. Mean ± SD of 3-4 independent experiments (E), or 1 experiment with 3 technical replicates (D). *P < .05, **P < .01, ****P < .0001 by unpaired Student t test. (E) Transduction of primary PBMC cultured in IL-2 by CD3-LVs upon treatment with Bfla1. N = 2 technical replicates from 1 donor, mean ± SD.
Figure 4.
Figure 4.
CD3-LVs transduce human T cells in whole blood. CD3-LVs and VSV-LV were tested for their ability to transduce T cells in whole blood of healthy donors in absence of additional stimuli and transduction enhancer. (A) Experimental procedure. (B) Representative dot plots show the percentages of GFP+ cells gated from all viable single cells at day 5 posttransduction in isolated PBMC. (C-D) Bar diagrams show the percentages of GFP+ cells of all viable single cells (C) and in CD4+ and CD8+ cell subsets (D) at day 5 posttranduction. *P < .05, **P < .01 by mixed-effect analysis with Sidak's correction. Mean ± SD of 3 independent experiments with 5 different donors. *P < .05, **P < .01 by mixed-effect analysis with Dunnett's correction.
Figure 5.
Figure 5.
TR66-LV mediates efficient in vivo gene delivery into all T-cell subpopulations. (A) Experimental outline: NSG mice were transplanted with αCD3/αCD28-activated human PBMC by intraperitoneal (IP) injection followed by vector administration 1 day later and analyzed for transgene expression by flow cytometry 7 days after vector application. (B) Representative dot plots show transduced T cells gated from all viable single CD3+ cells harvested from the peritoneal cavity at 7 days after vector injection. (C-E) Scatter plots summarize the percentages of GFP+ cells gated from all viable single CD3+ cells for peritoneum (C), blood (D), and spleen (E). (F-H) Respective scatter plots of GFP+ cells gated from CD4+ and CD8+ T cells isolated from peritoneum (F), blood (G), and spleen (H) at the day of analysis. N = 3 mice per group, mean ± SD. **P < .01, ***P < .001, ****P < .0001 by 1-way analysis of variance with Sidak's or Dunnett's correction.
Figure 6.
Figure 6.
Selectivity of TR66-LV in vivo. (A) Strategy for backgating of GFP+ cells generated in PBMC-NSG mice of Figure 5. All viable single GFP+ cells were assessed for expression of human CD45, CD3, and CD19. (B-D) Scatter plots for the percentages of GFP+ cells in the indicated fractions of peritoneum (B), blood (C), and spleen (D). Data are mean ± SD from n = 3 animals per group. ****P < .0001 by 1-way ANOVA with Sidak's correction.
Figure 7.
Figure 7.
CD3-targeted LVs mediate functional CAR T-cell generation in vivo. For in vivo CAR delivery, huNSG mice were subcutaneously (SC) injected with human IL-7 or phosphate-buffered saline 4 days and 1 day before IV injection of TR66.opt-LV harboring the CD19-CAR or vehicle as control. (A) Experimental outline. (B) Representative plots show CAR+ cells in CD8+ and CD8 T cells at day 26 postinjection of vector. (C) Line diagrams show CAR+ cells in CD3+ cells in the peripheral blood of individual mice in group 1 (IL-7), group 2 (TR66.opt-LV), and group 3 (TR66.opt-LV + IL-7). *,#P < .05, **,##P < .01, ***,###P < .001 comparing group 3 to group 1 (*) and group 2 (#) by 2-way ANOVA with Tukey's correction. Arrows point to tendencies for CAR T-cell detection in group 2, which were not statistically significant. (D) Scatter dot plots compare the percentages of CAR+ cells within the CD3+CD45+ T-cell compartment, CD45 mouse cells and CD3CD45+ non-T cells. (E-F) B-cell levels were determined in peripheral blood of the animals to assess CD19-CAR T-cell functionality. (E) Representative dot plots show CD19+CD20+ B cells at day 26 postinjection. (F) Line diagrams summarize relative levels of CD19+ cells compared with day 0 in the peripheral blood of individual mice in group 1 (IL-7), group 2 (TR66.opt-LV), and group 3 (TR66.opt-LV + IL-7). All data from n = 5-6 mice per group. *P < .05, **P < .01, ***P < .001 comparing groups 2 and 3 to group 1 by 2-way ANOVA with Turkey's correction.

References

    1. Maude SL, Laetsch TW, Buechner J, et al. . Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. N Engl J Med. 2018;378(5):439-448. - PMC - PubMed
    1. Neelapu SS, Locke FL, Bartlett NL, et al. . Axicabtagene ciloleucel CAR T-cell therapy in refractory large B-cell lymphoma. N Engl J Med. 2017;377(26):2531-2544. - PMC - PubMed
    1. Hartmann J, Schüßler-Lenz M, Bondanza A, Buchholz CJ. Clinical development of CAR T cells-challenges and opportunities in translating innovative treatment concepts. EMBO Mol Med. 2017;9(9):1183-1197. - PMC - PubMed
    1. June CH, O’Connor RS, Kawalekar OU, Ghassemi S, Milone MC. CAR T cell immunotherapy for human cancer. Science. 2018;359(6382):1361-1365. - PubMed
    1. June CH, Sadelain M. Chimeric antigen receptor therapy. N Engl J Med. 2018;379(1):64-73. - PMC - PubMed

Publication types