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
. 2018 May;32(5):1157-1167.
doi: 10.1038/s41375-017-0008-6. Epub 2018 Feb 2.

PI3K orchestration of the in vivo persistence of chimeric antigen receptor-modified T cells

Affiliations

PI3K orchestration of the in vivo persistence of chimeric antigen receptor-modified T cells

Wenting Zheng et al. Leukemia. 2018 May.

Abstract

In vivo persistence of chimeric antigen receptor (CAR)-modified T cells correlates with therapeutic efficacy, yet CAR-specific factors that support persistence are not well resolved. Using a CD33-specific CAR in an acute myeloid leukemia (AML) model, we show how CAR expression alters T cell differentiation in a ligand independent manner. Ex vivo expanded CAR-T cells demonstrated decreased naïve and stem memory populations and increased effector subsets relative to vector-transduced control cells. This was associated with reduced in vivo persistence. Decreased persistence was not due to specificity or tumor presence, but to pre-transfer tonic signaling through the CAR CD3ζ ITAMs. We identified activation of the PI3K pathway in CD33 CAR-T cells as responsible. Treatment with a PI3K inhibitor modulated the differentiation program of CAR-T cells, preserved a less differentiated state without affecting T cell expansion, and improved in vivo persistence and reduced tumor burden. These results resolve mechanisms by which tonic signaling of CAR-T cells modulates their fate, and identifies a novel pharmacologic approach to enhance the durability of CAR-T cells for immunotherapy.

PubMed Disclaimer

Conflict of interest statement

The authors have no conflicts of interest to declare.

Figures

Figure 1
Figure 1. CD33 and CD19 CAR-T cells control AML growth
Mice were injected with MOLM-13-CD19 tumor cells and treated with CD33 CAR, CD19 CAR, or control T cells. Mice were monitored for survival and tumor burden, or organs were harvested 18 days after transfer. (A) Kaplan-Meier survival analysis. (B) Xenogen images and bioluminescent signal intensities (C) MOLM-13-CD19 tumor cell numbers *** p<0.001, **** p<0.0001.
Figure 2
Figure 2. Poor CAR-T cell survival is tumor independent
(A) Mice were injected with MOLM-13-CD19 tumor cells and treated with CD33 CAR, CD19 CAR or control T cells. Organs were harvested 5 days after transfer. Total numbers of CD3+CD8+ CAR or control T cells are shown. (B–C) CD33 CAR-T cells (GFP) and control T cells (RFP) were co-transferred at ratios of 0.9:1–1.1:1 into mice with or without tumor. Organs were harvested 5 days after transfer. (B) Numbers of CD8+ CAR and control T cells with and without tumor. (C) Ratio of CD8+ control T cells to CD33 CAR-T cells with and without tumor, normalized to input ratio. * p<0.05, ** p<0.01, *** p<0.001, **** p<0.0001.
Figure 3
Figure 3. CAR-T cells exhibit increased effector differentiation
(A) Composition of TN, TCM, TEM and TEFF CD8+ T cell subsets in CD33 CAR and control T cells after ex vivo activation. (B) Percent of TSCM cells after ex vivo activation. (C) Methylation analysis of genomic DNA CpG sites within the IFNγ promotor. Naïve CD8+CD45RA+CD45ROCCR7+CD95 T cells were sorted from donor samples, transduced with CD33 CAR or control vector, and assessed 9 days after ex vivo activation. Each line represents an individual clone. Bar graphs show % CpG methylation at each site of the locus in CD33 CAR or control T cells. * p<0.05, ** p<0.01, **** p<0.0001.
Figure 4
Figure 4. Altered CAR-T cell differentiation due to CAR CD3ζ ITAM signaling
(A) Composition of CD8+ TN, TCM, TEM, and TEFF subsets in CD33 41BB, CD33 ITAM, and CD33 41BB ITAM T cells relative to CD33 CAR and control T cells 12 days after activation. (B) Activation and exhaustion marker expression on CD8+ CD33 CAR and CD33-ITAM T cells 12 days after activation. (C) MOLM-13-CD19 cells were incubated with CD33 CAR, CD33 CAR ITAM mutant, or control T cells as indicated for 24–48h. Tumor cells were quantified by flow cytometry and normalized to cultures without added T cells. * p<0.05, *** p<0.001, **** p<0.0001.
Figure 5
Figure 5. Differentiation status of CAR-T cells influences in vivo persistence
(A–B) CD33 CAR and control T cells were mixed 1:1 and transferred with or without MOLM-13-CD19 tumor cells into mice. Cell numbers were determined 5 days after transfer. (A) Proportions of and (B) Numbers of CD8+ TN, TCM, TEM and TEFF subsets. (C) CD8+CCR7+CD45RA+ TN and CD8+CCR7CD45RA TEM cells were sorted from activated CD33 CAR or control T cells and transferred individually into mice. Cell numbers were normalized to the CD33 CAR TEM group. * p<0.05, ** p<0.01, *** p<0.001, **** p<0.0001.
Figure 6
Figure 6. CD33 CAR-T cells exhibit a distinct transcriptional profile
(A) Differentially expressed genes from PI3K/AKT and glycolysis pathways identified by Ingenuity Pathway Analysis (IPA) of CD8+ CD33 CAR and control T cells 12 days after ex vivo activation. (B) Phospho-flow staining of pS6 and p4EBP-1 in CD33 CAR and control T cells. CD33 CAR-T cells were stimulated with PMA/Ionomycin as a positive control. ** p<0.01, **** p<0.0001.
Figure 7
Figure 7. PI3K inhibition restrains aberrant CAR-T cell differentiation ex vivo
CD33 CAR or control T cells were stimulated for 5 days, treated with inhibitor for 4 days, and analyzed by flow cytometry. PI3K inhibitors LY294002 (LY) and IC87114 (IC), mTOR inhibitors rapamycin (RAPA) and PP242, AKT inhibitor API-2, and glycolysis inhibitor DCA were used as indicated. (A) Percentages of TN, TCM, TEM and TEFF subsets of CD8+ T cells. (B) Numbers of TN, TCM, TEM and TEFF subsets of CD8+ T cells normalized to the number of untreated CD33 CAR-T cells. (C) Relative numbers of total CD8+ T cells after 4 days of inhibitor treatment, normalized to untreated CAR-T cells. * p<0.05, ** p<0.01, *** p<0.001, **** p<0.0001.
Figure 8
Figure 8. PI3K inhibition during ex vivo expansion improves CAR-T cell persistence and reduces tumor burden in vivo
Mice were injected with MOLM-13-CD19 tumor cells and either untreated or LY-treated CD33 CAR-T cells. Organs were harvested 14 days after transfer. (A) Total CD8+ T cell numbers (top) and total MOLM-13-CD19 tumor cell numbers (bottom). (B) Proportions of CD8+ TN, TCM, TEM and TEFF subsets. (C) Kaplan-Meier survival analysis of mice with MOLM-13-CD19 tumors treated with CD33 CAR, CD33 CAR + LY treatment, or vector-transduced control T cells. ** p<0.01, *** p<0.001, **** p<0.0001.

References

    1. Srivastava S, Riddell SR. Engineering CAR-T cells: Design concepts. Trends in immunology. 2015 Aug;36(8):494–502. - PMC - PubMed
    1. Kalos M, June CH. Adoptive T cell transfer for cancer immunotherapy in the era of synthetic biology. Immunity. 2013 Jul 25;39(1):49–60. - PMC - PubMed
    1. Park JH, Geyer MB, Brentjens RJ. CD19-targeted CAR T-cell therapeutics for hematologic malignancies: interpreting clinical outcomes to date. Blood. 2016 Jun 30;127(26):3312–3320. - PMC - PubMed
    1. Casucci M, Nicolis di Robilant B, Falcone L, Camisa B, Norelli M, Genovese P, et al. CD44v6-targeted T cells mediate potent antitumor effects against acute myeloid leukemia and multiple myeloma. Blood. 2013 Nov 14;122(20):3461–3472. - PubMed
    1. Pizzitola I, Anjos-Afonso F, Rouault-Pierre K, Lassailly F, Tettamanti S, Spinelli O, et al. Chimeric antigen receptors against CD33/CD123 antigens efficiently target primary acute myeloid leukemia cells in vivo. Leukemia. 2014 Aug;28(8):1596–1605. - PubMed

Publication types

MeSH terms

Substances