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
. 2022 Aug 4;29(8):1181-1196.e6.
doi: 10.1016/j.stem.2022.06.014.

EZH1 repression generates mature iPSC-derived CAR T cells with enhanced antitumor activity

Affiliations

EZH1 repression generates mature iPSC-derived CAR T cells with enhanced antitumor activity

Ran Jing et al. Cell Stem Cell. .

Erratum in

Abstract

Human induced pluripotent stem cells (iPSCs) provide a potentially unlimited resource for cell therapies, but the derivation of mature cell types remains challenging. The histone methyltransferase EZH1 is a negative regulator of lymphoid potential during embryonic hematopoiesis. Here, we demonstrate that EZH1 repression facilitates in vitro differentiation and maturation of T cells from iPSCs. Coupling a stroma-free T cell differentiation system with EZH1-knockdown-mediated epigenetic reprogramming, we generated iPSC-derived T cells, termed EZ-T cells, which display a highly diverse T cell receptor (TCR) repertoire and mature molecular signatures similar to those of TCRαβ T cells from peripheral blood. Upon activation, EZ-T cells give rise to effector and memory T cell subsets. When transduced with chimeric antigen receptors (CARs), EZ-T cells exhibit potent antitumor activities in vitro and in xenograft models. Epigenetic remodeling via EZH1 repression allows efficient production of developmentally mature T cells from iPSCs for applications in adoptive cell therapy.

Keywords: CAR T cells; EZH1; T cell differentiation; cancer immunotherapy; hematopoietic stem and progenitor cells; pluripotent stem cells.

PubMed Disclaimer

Conflict of interest statement

Declaration of interests R.J., G.Q.D., and Boston Children’s Hospital hold intellectual property and receive consulting fees and/or hold equity interest relevant to the generation of iPSC-derived T cells. T.M.S. receives sponsored research support from Elevate Bio. G.Q.D. is a member of Cell Stem Cell’s advisory board.

Figures

Fig. 1.
Fig. 1.. Stroma-free differentiation of human iPSCs into T cells.
A) Schematic illustration of the stroma-free T cell differentiation. B) Representative images showing day 0 CD34+ HE cells, day 14 proT cells, and day 35 T cells. Scale bar: 200μm. C) Expression of T cell lineage-specific markers during the stroma-free T cell differentiation of iPSCs (gated on CD45+ cells). D) Frequencies of CD4, CD8, and DP T cells in CD3+ cells after 6 weeks of differentiation (n=3, mean ± SEM). E) Numbers of week 6 CD3+ T cells generated via OP9-DL1 (blue) or stroma-free (red) differentiation, normalized to numbers of CD34+ HE cells seeded on day 0 (n=3, mean ± SEM, * P≤0.05). F) Frequencies of B cells (CD19+), NK cells (CD56+), Myeloid cells (CD33+), T cell precursors (CD5+CD3), and T cells (CD3+) in CD45+ hematopoietic cells after 6 weeks of T cell differentiation using the OP9-DL1 (blue) or stroma-free (red) method (n=3, mean ± SEM, * P≤0.05, ** P≤0.01). G) Frequencies of Vβ family genes determined by sequencing of the TCRβ CDR3 region (n=2)
Fig. 2.
Fig. 2.. EZH1 knockdown facilitates in vitro T cell differentiation from iPSCs.
A) Schematic illustration of EZ-T cell generation. B) Numbers of live cells during T cell differentiation using control (blue) or EZH1 knockdown (KD) (red) iPSC-derived HE cells (n=3, mean ± SEM, * P≤0.05). C) Frequencies of CD3+ T cells in CD45+ cells generated from control (blue) or EZH1 KD (red) cells after stroma-free T cell differentiation (n=3, mean ± SEM, *** P≤0.001). D) Numbers of CD3+ T cells generated from control (blue) or EZH1 KD (red) cells, normalized to numbers of CD34+ HE cells seeded on day 0 (n=3, mean ± SEM, ** P≤0.01). E) Frequencies of CD4, CD8, and DP T cells generated from control (blue) or EZH1 KD (red) cells after 6 weeks of stroma-free T cell differentiation (n=3, mean ± SEM, *** P≤0.001). F-H) Expression of CD3 and TCRαβ /TCRγδ/CD1a in control and EZH1 KD cells after stroma-free T cell differentiation, gated on CD45+ cells. I) Expression of CD8α and CD8β in control and EZH1 KD cells after stroma-free T cell differentiation, gated on CD8 T cells.
Fig. 3.
Fig. 3.. EZ-T cells display molecular features of mature TCRαβ T cells.
A) Heatmap showing CellNet analysis of RNA-seq data from iPSC-derived CD34+ HSPCs and iPSC-derived T cells generated via stroma-free protocol, either with EZH1 knockdown (iPSC-EZ-T), or without (iPSC-SF-T). B) Dendrogram representing hierarchical cluster analysis based on expression of TCR pathway genes (BIOCARTA_TCR_PATHWAY, M19784) (n=3). C) Heatmap showing unsupervised clustering analysis based on TCRαβ signature genes (n=3). D) Top GO terms of biological process enriched in iPSC-EZ-T cells vs. iPSC-SF-T cells by GSEA analysis (n=3). E) GSEA enrichment plots showing over-representation of gene sets related to T cell development and functions.
Fig. 4.
Fig. 4.. TCR repertoire analysis of EZ-T cells.
A) Frequencies of Vβ family genes in EZ-T cells determined by sequencing of the TCRβ CDR3 region (n=2). B) TCRβ CDR3 length of iPSC-EZ-T cells (red) compared to control iPSC-SF-T cells without EZH1 knockdown (blue) (n=2). C) Relative expression levels of TdT/DNTT in undifferentiated iPSCs or iPSC-SF-T cells with (red) or without (blue) EZH1 KD (n=3, mean ± SEM, * P≤0.05).
Fig. 5.
Fig. 5.. Single cell RNA-seq analysis identifies memory-like T cell subsets in EZ-T cells after activation.
A) Uniform Manifold Approximation and Projection (UMAP) visualization of all the CD45+ cells generated from EZ-T cell differentiation with and without activation. B) UMAP visualization of the expression of hematopoietic and T cell markers. C) Heatmap showing expression levels of T/NK cell signature genes across all clusters. D) GSEA analysis of the memory-like CD8 cluster showing over-representation of genes enriched in memory T cells but not naive or effector T cells. E) UMAP analysis comparing cell types in CD45+ cells generated from EZ-T cell differentiation before and after activation. F) Proportion of cells in unactivated and activated CD45+ cells generated via EZ-T cell differentiation. G) CellRouter analysis showing transcriptional regulators enriched in the memory-like CD8 T cell cluster.
Fig. 6.
Fig. 6.. EZ-T cells display enhanced effector functions.
A) CD69 expression and B) CD107a degranulation of iPSC-derived T cells and peripheral blood T cells after 6 hours of PMA/ionomycin stimulation, determined by flow cytometry analysis (n=3, mean ± SEM). C) CAR-T cells generated from iPSC-derived T cells or peripheral blood T cells were co-cultured with JeKo1 and D) OCI-Ly1 tumor cell lines at indicated effector to target (E:T) ratios. Bar graph showing percentages of specific cytolysis of target tumor cells (n=3, mean ± SEM). E) Production of IL-2, F) INFγ, and G) TNFα by CD19 CAR T cells generated from iPSC-SF-T, iPSC-EZ-T, and PBMC-T cells cultured in the absence (Unstim) and presence of OCI-Ly1 target tumor cells (n=3, mean ± SEM, **** P≤0.0001).
Fig. 7.
Fig. 7.. CD19 CAR EZ-T cells mediate more robust in vivo tumor clearance
A) Schematic illustration of in vivo CAR T cell functional studies using a DLBCL mouse model. B) Bioluminescent images of tumor xenografts over time and quantification of the tumor burden over time in each animal (n=9 animals from 3 experiments), represented by mean total flux (photons/sec). C) Numbers of CAR T cells per 100μl peripheral blood from each animal, determined by flow cytometry 3 weeks after CAR T cell injection (n=8, *** P≤0.001). D) Kaplan-Meier curve showing percentage survival of untreated animals (black) and animal groups treated with CD19 CAR T cells generated from control iPSC-SF-T (red), iPSC-EZ-T (blue), or PBMC-T cells (yellow) (n=9, ** P≤0.01 by log-rank test).

Comment in

  • T cell development made EZ.
    Lavaert M, Bhandoola A. Lavaert M, et al. Cell Stem Cell. 2022 Aug 4;29(8):1155-1156. doi: 10.1016/j.stem.2022.07.003. Cell Stem Cell. 2022. PMID: 35931025

References

    1. Adachi K, Kano Y, Nagai T, Okuyama N, Sakoda Y, and Tamada K (2018) IL-7 and CCL19 expression in CAR-T cells improves immune cell infiltration and CAR-T cell survival in the tumor. Nat Biotechnol, 36, 346–351. - PubMed
    1. Aibar S, González-Blas CB, Moerman T, Huynh-Thu VA, Imrichova H, Hulselmans G, Rambow F, Marine JC, Geurts P, Aerts J et al. (2017) SCENIC: single-cell regulatory network inference and clustering. Nat Methods, 14, 1083–1086. - PMC - PubMed
    1. Baldwin TA, Sandau MM, Jameson SC, and Hogquist KA (2005) The timing of TCR alpha expression critically influences T cell development and selection. J Exp Med, 202, 111–121. - PMC - PubMed
    1. Basar R, Daher M, and Rezvani K (2020) Next-generation cell therapies: the emerging role of CAR-NK cells. Hematology Am Soc Hematol Educ Program, 2020, 570–578. - PMC - PubMed
    1. Bosselut R, Kubo S, Guinter T, Kopacz JL, Altman JD, Feigenbaum L, and Singer A (2000) Role of CD8beta domains in CD8 coreceptor function: importance for MHC I binding, signaling, and positive selection of CD8+ T cells in the thymus. Immunity, 12, 409–418. - PubMed

Publication types

MeSH terms

Substances