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
Clinical Trial
. 2016 Jun;24(6):1135-1149.
doi: 10.1038/mt.2016.63. Epub 2016 Mar 29.

GD2-specific CAR T Cells Undergo Potent Activation and Deletion Following Antigen Encounter but can be Protected From Activation-induced Cell Death by PD-1 Blockade

Affiliations
Clinical Trial

GD2-specific CAR T Cells Undergo Potent Activation and Deletion Following Antigen Encounter but can be Protected From Activation-induced Cell Death by PD-1 Blockade

Tessa Gargett et al. Mol Ther. 2016 Jun.

Abstract

Chimeric antigen receptor (CAR) T cells have shown great promise in the treatment of hematologic malignancies but more variable results in the treatment of solid tumors and the persistence and expansion of CAR T cells within patients has been identified as a key correlate of antitumor efficacy. Lack of immunological "space", functional exhaustion, and deletion have all been proposed as mechanisms that hamper CAR T-cell persistence. Here we describe the events following activation of third-generation CAR T cells specific for GD2. CAR T cells had highly potent immediate effector functions without evidence of functional exhaustion in vitro, although reduced cytokine production reversible by PD-1 blockade was observed after longer-term culture. Significant activation-induced cell death (AICD) of CAR T cells was observed after repeated antigen stimulation, and PD-1 blockade enhanced both CAR T-cell survival and promoted killing of PD-L1(+) tumor cell lines. Finally, we assessed CAR T-cell persistence in patients enrolled in the CARPETS phase 1 clinical trial of GD2-specific CAR T cells in the treatment of metastatic melanoma. Together, these data suggest that deletion also occurs in vivo and that PD-1-targeted combination therapy approaches may be useful to augment CAR T-cell efficacy and persistence in patients.

PubMed Disclaimer

Figures

Figure 1
Figure 1
Markers of activation and exhaustion on GD2-iCAR and paired nontransduced T cells in vitro. Patient peripheral blood mononuclear cells were activated, transduced with GD2-iCAR retroviral vector and expanded for 14 days before cryopreservation. Chimeric antigen receptor (CAR) T cells and nontransduced (NT) T-cell controls were manufactured from four patients with metastatic melanoma. Cells were then thawed and restimulated. (a) CD25 and CD69 expression during culture. (b) PD-1 expression during culture. (c) LAG-3 expression during culture. (d) Difference in PD-1 expression after restimulation with 1A7 or -anti-CD3/CD28 antibodies. Representative data are shown for patient 101. All samples were analyzed in duplicate, with graphs displaying mean ± standard error of the mean. The gating strategy is shown in Supplementary Figure S1.
Figure 2
Figure 2
Functional capacity of GD2-iCAR T cells in vitro with or without PD-1 blockade. Thawed chimeric antigen receptor (CAR) T cells were stimulated via CD3/CD28 or CAR in the absence of exogenous cytokine and in the absence (white) or presence (gray) of 20 µg/ml anti-PD-1 blocking antibody. Supernatants were collected after 3 days (day 3 plain bars) stimulation or 3 days after a restimulation on day 7 (Day 10, striped bars) and diluted 1/10 prior to analysis for cytokine secretion. (a) IFNγ. (b) TNFα. (c) IL-2. (d) IL-10. (e) IL-4. (f) IL-6. (g) CAR T-cell killing of GD2+ PD-L1- (M238, circles) and PD-L1 + (M238R, squares) melanoma cell lines ± 20 µg/ml anti-PD-1. Cells were thawed, rested overnight and then cultured with 51Cr-labeled tumor lines for 6 hours. (h) CAR T-cell killing of melanoma cell lines on day 7 after 3 days of stimulation. Cells were thawed, rested overnight and then stimulated with anti-CAR for 3 days and rested for 4 days before culture with 51Cr-labeled tumor lines for 6 hours. Flow samples were analyzed in duplicate and 51Cr assays in triplicate. Representative data are shown for patient 101, and graph display mean ± standard error of the mean. Cytokine secretion for nontransduced controls are shown in Supplementary Figure S2.
Figure 3
Figure 3
Comparison of downstream signaling events following chimeric antigen receptor (CAR) or CD3/CD28 stimulation. Calcium flux after stimulation of thawed CAR T cells with primary (1°) antibodies specific for (a) the CAR (1A7) or (b) the CD3 (OKT3) receptors, with or without cross-linking by antimouse IgG secondary (2°) antibodies. (c) Ionomycin and isotype control and a secondary antibody were used as positive and negative controls, respectively. Anti-CD28 and anti-OX40 antibodies were also used in conjunction with anti-CD3 but did not induce markedly different calcium flux compared to anti-CD3 alone (data not shown). CAR T cells, nontransduced (NT) T cells and peripheral blood mononuclear cells (PBMC) were stimulated via CAR or CD3/CD28 receptors and analyzed for signaling molecule phosphorylation. (d) and (e) pERK+ T cells at 6 and 48 hours. (f) and (g) pAKT+ T cells 12 and 24 hours. (h) pSTAT5+ T cells at 24 hours. (i) pSTAT6+ T cells at 12 hours. (j) and (k) active caspase 3 in T cells at 12 and 24 hours. CAR T cells and NT T cells and PBMC were tested in duplicate. Representative data are shown for patient 102, and graphs display mean ± standard error of the mean. The gating strategy is shown in Supplementary Figure S3.
Figure 4
Figure 4
Activation-induced cell death (AICD) after repeated antigen stimulation of GD2-iCAR T cells with GD2+ tumor cells. Thawed chimeric antigen receptor (CAR) T cells were passaged daily onto tumor cells, and cells were collected at days 1, 2, and 3 for analysis of activation (CD25 and CD69) and cell death (Annexin V and DAPI) by flow cytometry. SK-Mel cells provide a GD2- control and nontransduced (NT) T cells provided controls lacking CAR expression. Anti-PD-1was included in some cultures. CAR T-cell activation (a) and viability (b) after repeated stimulation with tumor cell lines. (c) CAR T-cell death after repeated stimulation by GD2hi neuroblastoma cell lines (LAN-1), with or without anti-PD-1. (d) CAR T-cell killing of GD2hi PD-L1- neuroblastoma cells (LAN-1), with or without anti-PD-1. (e) CART-cell killing of GD2+PD-L1+ melanoma cells (M238R), with or without anti-PD-1. Repetitive antigen stress-test assays were performed in duplicate. Representative data are shown for patient 101, and graphs display mean ± standard error of the mean. The gating strategy is shown in Supplementary Figure S4.
Figure 5
Figure 5
PD-1/PD-L1-dependent activation-induced cell death (AICD) after activation of GD2-iCAR T cells. (a) PD-L1 expression on thawed chimeric antigen receptor (CAR) T cells after stimulation in vitro. (b) Immune phenotype of CAR PD-L1+ and PD-L1- T cells at day 3 poststimulation. Immune phenotype defined as: Naive (CD45RA+CCR7+), effector memory (Tem; ––CD45RACCR7), central memory (Tcm; CD45RACCR7+), effector (CD45RA+CCR7), activated (CD25+/CD69+), proliferating (CFSE low). (c) CAR T-cell viability after repetitive stimulation with LAN-1. (d) GD2+PD-L1- M238 tumor cell viability and (e) GD2++PD-L1- LAN-1 tumor cell viability and (f) GD2+PD-L1+ M238R tumor cell viability after stress-test assay with PD-L1 blockade (anti-PD-L1). Repetitive antigen stress-test assays and flow cytometry analyses were performed in duplicate. Representative data are shown for patient 101, and graphs display mean ± stanadard error of the mean. The gating strategy is shown in Supplementary Figure S5.
Figure 6
Figure 6
Chimeric antigen receptor (CAR) cross-linking also induces activation-induced cell death. (a) Viability, (b) activation, (c) PD-1, and (d) PD-L1 expression after CAR cross-linking with 1A7 and secondary antibody (2°) of thawed CAR T cells and nontransduced (NT) controls. (e) GD2-iCAR T-cell AICD in the presence of PD-1-, PD-L1-, or FAS-blocking antibodies after CAR cross-linking. Cross-linking assays and flow cytometry analyses were performed in duplicate. Representative data are shown for patient 101, and graphs display mean ± stanadard error of the mean. The gating strategy is shown in Supplementary Figure S5.
Figure 7
Figure 7
Chimeric antigen receptor (CAR) T-cell persistence and immune phenotype in patients receiving third-generation GD2-specific CAR T cells for the treatment of V600 BRAF-mutant metastatic melanoma. (a) Flow cytometry using 1A7 to detect GD2-iCAR-expressing T cells in peripheral blood. (b) Quantitative polymerase chain reaction (QPCR) to detect GD2-iCAR transgene-positive cells in peripheral blood. (c) Patient 102 serum cytokine levels. (d) Patient 102 CD4 and CD8 CAR T-cell subsets for peripheral blood mononuclear cells, the GD2-iCAR T-cell product and circulating GD2-iCAR T cells detected from day 7 postadministration. (e) Patient 102 CD8+ T-cell immune phenotype and (f) PD1/PD-L1 expression on peripheral blood normal T cells and 1A7+ GD2-iCAR T cells. Experiments were performed in duplicate (flow cytometry) or triplicate (QPCR), with graphs displaying mean ± standard error of the mean. Gating strategy and data for Patients 101, 201 and 203 are shown in Supplementary Figure S6.

References

    1. Porter, DL, Levine, BL, Kalos, M, Bagg, A and June, CH (2011). Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N Engl J Med 365: 725–733. - PMC - PubMed
    1. Grupp, SA, Kalos, M, Barrett, D, Aplenc, R, Porter, DL, Rheingold, SR et al. (2013). Chimeric antigen receptor-modified T cells for acute lymphoid leukemia. N Engl J Med 368: 1509–1518. - PMC - PubMed
    1. Kershaw, MH, Westwood, JA, Parker, LL, Wang, G, Eshhar, Z, Mavroukakis, SA et al. (2006). A phase I study on adoptive immunotherapy using gene-modified T cells for ovarian cancer. Clin Cancer Res 12(20 Pt 1): 6106–6115. - PMC - PubMed
    1. Louis, CU, Savoldo, B, Dotti, G, Pule, M, Yvon, E, Myers, GD et al. (2011). Antitumor activity and long-term fate of chimeric antigen receptor-positive T cells in patients with neuroblastoma. Blood 118: 6050–6056. - PMC - PubMed
    1. Lamers, CH, Sleijfer, S, van Steenbergen, S, van Elzakker, P, van Krimpen, B, Groot, C et al. (2013). Treatment of metastatic renal cell carcinoma with CAIX CAR-engineered T cells: clinical evaluation and management of on-target toxicity. Mol Ther 21: 904–912. - PMC - PubMed

Publication types