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 VSports app下载. gov or . mil. Before sharing sensitive information, make sure you’re on a federal government site. .

Https

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

. 2016 Apr;34(4):430-4.
doi: 10.1038/nbt.3461. Epub 2016 Feb 22.

Inclusion of Strep-tag II in design of antigen receptors for T-cell immunotherapy

Affiliations

Inclusion of Strep-tag II in design of antigen receptors for T-cell immunotherapy

Lingfeng Liu et al. Nat Biotechnol. 2016 Apr.

Abstract

Adoptive immunotherapy with genetically engineered T cells has the potential to treat cancer and other diseases. The introduction of Strep-tag II sequences into specific sites in synthetic chimeric antigen receptors or natural T-cell receptors of diverse specificities provides engineered T cells with a marker for identification and rapid purification, a method for tailoring spacer length of chimeric receptors for optimal function, and a functional element for selective antibody-coated, microbead-driven, large-scale expansion. These receptor designs facilitate cGMP manufacturing of pure populations of engineered T cells for adoptive T-cell therapies and enable in vivo tracking and retrieval of transferred cells for downstream research applications. VSports手机版.

PubMed Disclaimer

Conflict of interest statement (VSports注册入口)

Competing Financial Interests Statement

L. L. and S. R V体育安卓版. R. are co-inventors of a patent “Tagged chimeric effector molecules and receptors thereof” filed by Fred Hutchinson Cancer Research Center (PCT/US2014/072007), and licensed to Juno Therapeutics.

S. R. R V体育ios版. holds equity stake in, and is a cofounder of, Juno Therapeutics and is on the advisory board for and consults for Cell Medica.

No potential conflicts of interest were disclosed by the other authors.

Figures

Figure 1
Figure 1. Expression and function of CD19 CARs that contain Strep-tag II
(a) Analysis of CD19 CAR expression. Primary human CD8+ T cells were transduced with epHIV7 lentiviral vectors encoding a CD19-Hi CAR or CD19 CARs with Strep-tag in various extracellular locations. Each CAR contained a 4-1BB/CD3ζ intracellular signaling domain and EGFRt downstream of a T2A element. Transduced cells were sorted for EGFRt+ cells by fluorescence-activated cell sorting (FACS), and purity confirmed by staining with anti-EGFR (grey – top panels). Cell surface expression of the Strep-tag CARs was evaluated by staining with anti-Strep-tag II antibodies (grey - bottom panels). Non-transduced cells (white) served as controls for staining. (b) Cytolytic activity of CD19-Hi and Strep-tag 4-1BBζ CAR-T cells. After sorting for EGFRt expression, CD8+ T cells transduced with each of the CARs were tested for lysis of CD19+ Raji lymphoma and K562 leukemia transduced with CD19 (K562/CD19) or ROR1 (K562/ROR1) at various effector/target (E:T) ratios. (c) IFN-γ and IL2 production by CD19-Hi and Strep-tag 4-1BBζ CAR-T cells 24h after stimulation with K562/CD19 and K562/ROR1. PMA/Ionomycin treated T cells were used as a positive control. The data in a–c is representative of three experiments with CD8+ T cells from different donors. (d) Cohorts of NSG mice were inoculated with 0.5×106 firefly luciferase expressing CD19+ Raji lymphoma cells (Raji-ffluc) via tail vein injection and treated 7 days after tumor inoculation with 2.5×106 CD19 4-1BBζ CAR-T cells with Hi, 1ST, and 3ST spacers, respectively. CAR-T cells were formulated in a CD8:CD4 ratio of 1:1. Tumor progression and distribution were evaluated by serial bioluminescence imaging. (e) Tracking CAR-expressing T cells in vivo by staining with anti-Strep-tag II mAb. Blood obtained from mice 8 days after the T cell infusions was stained with anti-human CD45, CD8, CD4, anti-Strep-tag II and anti-EGFR mAbs, and analyzed by flow cytometry. Expression of Strep-tag II and EGFRt on CD45+ CD8+ and CD45+ CD4+ T cells is shown. (f) Kinetics of expansion and contraction of CD19 CAR-T cells in the blood after adoptive transfer to NSG mice bearing Raji tumors. The mean frequency of CD45+CD8+ EGFRt+ and CD45+CD4+ EGFRt+ human T cells in blood of the mice (n=5) of each group at various times after the T cell infusion is shown. The data in d–f are representative of three experiments. (g) Fold-change in expression of selected cytokine genes in CD19-1ST/4-1BBζ CAR-T cells after infusion to Raji tumor bearing and non-tumor bearing NSG mice. CAR-T cells were sorted 2 days after infusion from blood, bone marrow and spleen after staining with anti-EGFR or anti-Strep-tag mAb. Gene expression was analyzed using a human common cytokine PCR array. The mean fold change values of cytokine genes in the sorted CAR-T cells from NSG/Raji vs sorted CAR-T cells from non-tumor bearing NSG mice were calculated. Samples were run in triplicate and data are presented as the mean fold increase ± SD. (h) Cytokine production by CD8+ T cells expressing CD19-1ST/4-1BBζ CARs 2 days after stimulation with Raji cells in vitro. Supernatants of CAR-T cells co-cultured with CD19+ Raji cells (intra-assay triplicates) for 48 h were pooled together and analyzed using the Luminex Multiplex platform to validate the production of cytokines that were upregulated in CAR-T cells in NSG mice bearing Raji tumors. PMA/Ionomycin treated and non-treated T cells were used as positive and negative control, respectively. The results are representative of biological replicates.
Figure 2
Figure 2. Activation, proliferation and function of Strep-tag CAR-T cells after stimulation with anti-Strep tag II mAb
(a) CD4+ and CD8+ T cells expressing each of the CD19 CARs were sorted for EGFRt expression and stimulated with anti-Strep-tag II or anti-Strep-tag II/CD28 mAb–coated microbeads. After 48 h of stimulation, expression of the CD25 activation marker was determined by flow cytometry. Unstimulated cells (medium) were used as controls. (b) Growth curves of Strep-tag CAR-T cells. FACS sorted EGFRt+ CD19 CAR-T cells (CD8+ and CD4+) were cultured with anti-Strep-tag II or anti-Strep-tag II/CD28 mAb coated microbeads in CTL media containing IL-2 (30–50 U/ml) and IL-15 (2 ng/ml) for 9 days. Aliquots of T cells were removed from the cultures for counting on days 3, 6, and 9 and the fold-increase in cell number determined. The data show the mean fold expansion obtained in three experiments with T cells from different donors. (c) Stimulation of Strep-tag CAR-T cells with anti-Strep-tag II/CD28 beads induces selective outgrowth of transduced cells. CD8+ and CD4+ T cells were transduced with CD19 1ST/4-1BBζ and 1ST/CD28ζ CARs and 10 days later stimulated with anti-Strep-tag II/CD28 microbeads plus IL-2 or cultured with IL-2 alone for 9 additional days. The percentage of Strep-tag II positive cells at day 0 (before stimulation) and day 9 (after stimulation) were measured by flow cytometry. The results are representative of three experiments. (d) Anti-tumor activity of CD19 1ST/4-1BBζ or 1ST/CD28ζ CAR-T cells in Raji-ffluc-bearing NSG mice. 2.5×106 CD19 CAR-T cells expanded with anti-CD3/CD28 beads or with anti-Strep-tag II/CD28 microbeads, and control non-transduced T cells were formulated in a CD8:CD4 ratio of 1:1 and infused into cohorts of NSG mice 7 days after inoculation with 0.5×x106 Raji/ffluc tumor cells. Tumor progression and distribution were evaluated by serial bioluminescence imaging.
Figure 3
Figure 3. Strep-tag CAR-T cells can be enriched and exhibit potent anti-tumor activity in vivo
(a) Enrichment of Strep-tag CAR-T cells containing 1, 2, or 3 Strep-tag II sequences in the spacer region using StrepTactin coated beads on the automated T-CATCH device. Flow cytometric analysis of the frequency of Strep-tag CAR-T cells before and after enrichment using anti-Strep-tag II staining. Data is representative of six experiments using T cells from 3 donors. (b) Yield of Strep-tag CAR-T cells after T-CATCH enrichment. Yield was determined by the absolute numbers of Strep-tag CAR-T cells in the enriched fraction divided by the absolute numbers of Strep-tag CAR-T cells in the starting population. Data is derived from four experiments and expressed as means ± SD. Statistical analysis was performed using the Student’s t test. *P<0.05. (c) Experimental scheme for adoptive transfer of Strep-tag CAR-T cells enriched by StrepTactin selection or using EGFR mAb. CD8+ and CD4+ T cells were stimulated in independent cultures with anti-CD3/CD28 microbeads and transduced with the CD19-3ST/41BBζ CAR. Cultures were established at different times so that T cell administration into tumor bearing mice occurred simultaneously. Anti-CD3/CD28 beads were removed at day 5 in all groups and CAR-T cells were prepared for inoculation into mice either by selection on the T-CATCH at day 8, FACS sorting for EGFRt+ cells on day 10, or FACS sorting of EGFRt+ cells followed by 8 days of culture on irradiated CD19+ LCL cells with IL2 to remove residual bound anti EGFR mAb. (d) Flow cytometric analysis of CD19 CAR expression using Strep-tag II staining of CD8+ and CD4+ CAR-T cells before enrichment, after T-CATCH purification, after EGFR mAb sorting, and after EGFR mAb sorting followed by culture. (e) NSG mice engrafted 7 days earlier with 0.5×106 Raji/ffluc were treated with a total dose of 2.5×106 CAR-T cells selected by T-CATCH, EGFR sorting or EGFR sorting followed by culture and formulated in a CD4:CD8 ratio of 1:1. Tumor progression and distribution were evaluated by serial bioluminescence imaging after injection of luciferin substrate. (f) Persistence of CD19 CAR-T cells in each cohort of NSG/Raji mice. Flow cytometric analysis of CD4+ and CD8+ CAR T cells in the peripheral blood of each group of mice after staining with CD45, CD8, CD4 and EGFR Ab at different time points after T cell infusion. The frequency of CAR-T cells is presented as percentage of live peripheral blood cells. (g) Survival of mice treated with different CAR-T cell products or with non-transduced T cells depicted as Kaplan-Meier curves. The data in d-g are representative of two independent experiments.

References

    1. Brentjens RJ, et al. CD19-targeted T cells rapidly induce molecular remissions in adults with chemotherapy-refractory acute lymphoblastic leukemia. Science translational medicine. 2013;5:177ra138. doi: 10.1126/scitranslmed.3005930. - DOI - PMC - PubMed
    1. Kochenderfer JN, et al. Chemotherapy-refractory diffuse large B-cell lymphoma and indolent B-cell malignancies can be effectively treated with autologous T cells expressing an anti-CD19 chimeric antigen receptor. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2015;33:540–549. doi: 10.1200/jco.2014.56.2025. - DOI - PMC - PubMed
    1. Robbins PF, et al. A pilot trial using lymphocytes genetically engineered with an NY-ESO-1-reactive T-cell receptor: long-term follow-up and correlates with response. Clinical cancer research : an official journal of the American Association for Cancer Research. 2015;21:1019–1027. doi: 10.1158/1078-0432.ccr-14-2708. - DOI - PMC - PubMed
    1. Morgan RA, et al. Cancer regression in patients after transfer of genetically engineered lymphocytes. Science (New York, NY) 2006;314:126–129. doi: 10.1126/science.1129003. - DOI - PMC - PubMed
    1. Maude SL, et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. The New England journal of medicine. 2014;371:1507–1517. doi: 10.1056/NEJMoa1407222. - DOI - PMC - PubMed

Publication types

MeSH terms

Substances