EZH1 iPSC CAR T
EZH1 iPSC CAR T
EZH1 iPSC CAR T
Correspondence
[email protected]
In brief
Jing et al. combine a stroma-free
differentiation system with EZH1-
repression-mediated epigenetic
reprogramming to generate
developmentally mature iPSC-derived
CAR T cells with enhanced antitumor
activities.
Highlights
d Stroma-free differentiation of iPSCs into T cells expressing
diverse TCRs
Article
EZH1 repression generates mature iPSC-derived
CAR T cells with enhanced antitumor activity
Ran Jing,1,2 Irene Scarfo,3,4 Mohamad Ali Najia,1,2,5,6 Edroaldo Lummertz da Rocha,7 Areum Han,1,2 Michael Sanborn,1
Trevor Bingham,1 Caroline Kubaczka,1,2 Deepak K. Jha,1,2 Marcelo Falchetti,8 Thorsten M. Schlaeger,1,9
Trista E. North,1,9,10 Marcela V. Maus,3,4 and George Q. Daley1,2,9,10,11,*
1Stem Cell Program, Boston Children’s Hospital, Boston, MA 02115, USA
2Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
3Cellular Immunotherapy Program, Massachusetts General Hospital Cancer Center, Charlestown, MA 02114, USA
4Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
5Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
6Harvard-MIT Health Sciences & Technology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
7Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianópolis, SC 88040-900, Brazil
8Graduate Program of Pharmacology, Center for Biological Sciences, Federal University of Santa Catarina, Florianópolis, SC 88040-900,
Brazil
9Division of Hematology/Oncology, Boston Children’s Hospital and Dana Farber Cancer Institute, Boston, MA 02115, USA
10Developmental and Regenerative Biology Program, Harvard Medical School, Boston, MA 02115, USA
11Lead contact
*Correspondence: [email protected]
https://doi.org/10.1016/j.stem.2022.06.014
SUMMARY
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 repres-
sion facilitates in vitro differentiation and maturation of T cells from iPSCs. Coupling a stroma-free T cell dif-
ferentiation 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 TCRab 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 adop-
tive cell therapy.
Cell Stem Cell 29, 1181–1196, August 4, 2022 ª 2022 Elsevier Inc. 1181
ll
Article
A B
D E F
stroma-free production of fully functional, developmentally sion of endogenous or transgenic TCR has been shown to
mature iPSC-T cells are needed to realize a broad range of produce T cells with innate-like features (Terrence et al., 2000;
iPSC-based adoptive T cell therapies. Baldwin et al., 2005; Egawa et al., 2008; Zhao et al., 2007).
Recent studies have revealed key roles for epigenetic regula- Though it has been shown that co-culture with OP9-DL4 cells
tors during definitive hematopoiesis and lymphoid development. yields iPSC-T cells with a broad TCR repertoire (Chang et al.,
Our lab identified EZH1, a component of polycomb repressive 2014), whether a stroma-free system can fully recapitulate
complex 2 (PRC2), as a critical negative regulator of definitive normal T cell development and support developmental matura-
lymphoid commitment during embryonic hematopoietic devel- tion and random TCR rearrangement in vitro remains unclear.
opment (Vo et al., 2018). In this study, we tested the hypothesis To answer this question, we sought to produce highly differenti-
that EZH1 repression might lead to more faithful in vitro recapit- ated T cells from non-T cell-derived iPSCs while avoiding animal
ulation of T cell differentiation and developmental maturation stromal cells and serum. The differentiation procedure is illus-
from iPSCs. Below, we show that iPSC-T cells derived in a trated in Figure 1A. In the first step, human erythroblast-derived
stroma-free, serum-free system following EZH1 knockdown iPSCs (cell line 1157) were induced to form embryoid bodies to
(EZ-T cells) display a molecular signature that more closely ap- generate hemogenic endothelial (HE) cells with hematopoietic
proximates peripheral blood ab T cells than innate-like gd potential using a previously reported protocol (Ditadi et al.,
T cells and that upon activation are capable of giving rise to 2015). The CD34+ HE cells were collected and seeded as a mono-
effector cytotoxic T cells and T cell subsets that exhibit a mem- layer on tissue culture dishes coated with Notch Delta-like Ligand
ory-like phenotype. EZ-T cells expressing anti-CD19 CARs 4 (DLL4) and VCAM-1 (Shukla et al., 2017). To initiate T cell spec-
showed more robust tumor-killing activity and cytokine secretion ification, HE cells were cultured in SFEMII media supplemented
compared with control CAR-loaded iPSC-T cells generated with a cocktail of cytokines (SCF, FLT3, IL-7, IL-3, TPO). During
without EZH1 repression. Injection of CAR-loaded EZ-T cells this time period, HE cells underwent endothelial-to-hematopoiet-
into B cell lymphoma-bearing mice led to superior persistence ic transition (EHT), giving rise to floating cells that contain
and more efficient tumor clearance than control iPSC-derived CD5+CD7+ T cell progenitors (proTs). On day 14, the floating he-
CAR T cells. Thus, a combination of EZH1 repression and matopoietic cells were collected and replated on new DLL4
stroma-free T cell differentiation allows efficient production of coated plates, followed by 3 weeks of culture with FLT3 and
mature iPSC-T cells with enhanced functionality. Such an IL-7. The proT cells continued to expand and passed through a
approach is compatible with commercial-scale production for transient CD3-CD4+ immature CD4 single positive (ISP) stage
CAR T cell-based immunotherapy. before activating the expression of CD8 to form CD4+CD8+ dou-
ble positive (DP) cells (Figures 1B and 1C). After 5 weeks of differ-
RESULTS entiation, CD3+TCRab+ T cells were detected. Further culture of
these cells in the presence of anti-CD3/CD28 antibodies and IL-
A serum-free, stroma-free system allows efficient 15 facilitated the induction of single positive (SP) cells. At week 6,
differentiation of iPSCs into T cells expressing a majority of cells expressed CD3, with the population consisting
diverse TCRs of both DP and CD4/CD8 SP T cells (Figure 1D).
Existing in vitro T cell differentiation protocols largely rely on en- To further evaluate the yield and efficiency of our stroma-free
gineered mouse stromal cells expressing Notch ligands, such protocol, we differentiated iPSCs into T cells using both the
as OP9-DL1/DL4 or MS5-DL1 cells, to provide the continuous stroma-free method and a previously published OP9-DL1 co-
Notch signaling needed for T cell development (Holmes and Zú- culture system (Themeli et al., 2013). The stroma-free protocol
€cker, 2009; Timmermans et al., 2009; Seet et al., 2017).
ñiga-Pflu resulted in a significant increase in CD3+ T cell production (Fig-
Previous studies have shown that immobilized Notch ligands ure 1E). Compared with OP9-DL1 co-culture, the stroma-free
support the induction of primary CD34+ hematopoietic stem or system also led to more efficient T cell-specific differentiation,
progenitor cells (HSPCs) into T cell lineages (Huijskens et al., indicated by an increased proportion of T cells and concomitant
2014; Shukla et al., 2017). Recently, a stroma-free differentiation reductions of non-lymphoid cell frequencies (Figures 1F and
protocol was reported to generate T cells from TCR-transduced S1A). To determine whether the stroma-free differentiation was
iPSCs or antigen-specific cytotoxic T cell (CTL)-derived iPSCs accompanied by normal TCR rearrangement, we extracted
(Iriguchi et al., 2021). These iPSCs carry pre-rearranged TCRs genomic DNA from stroma-free iPSC-T cells and sequenced
and display different T cell differentiation kinetics than their the complementarity-determining region 3 (CDR3) of the TCRb
wild-type counterparts (Themeli et al., 2013). Premature expres- locus to profile the TCR repertoire. ImmunoSEQ analysis
identified tens of thousands of unique rearrangements with a phenotypic and molecular analyses to characterize the nature of
high degree of diversity in the usage of Vb family genes, demon- T cells produced following EZH1 knockdown. It has been sug-
strating the random recombination of CDR3 in the iPSC-T cells gested that T cell-derived iPSCs (T-iPSCs) bearing rearranged
(Figure 1G). Taken together, these data establish a stroma-free endogenous TCR genes result in the premature expression of
system that faithfully recapitulates normal T cell development TCR receptors during in vitro differentiation (Themeli et al.,
to produce iPSC-T cells with a highly diverse TCR repertoire. 2013), whereas T cell differentiation from non-T cell-derived-
PSCs displays similar kinetics to cord blood (CB) CD34+ cells,
EZH1 repression facilitates in vitro T cell differentiation with both gradually upregulating surface TCR/CD3 expression
from iPSCs only after the DP stage (Seet et al., 2017) (Figure 1C).
Previous studies have shown that EZH1 acts as a key regulator After differentiation, EZ-T cells displayed a significant increase
of hematopoietic multipotency, and repression of EZH1 function in CD3+TCRab+ and decrease in CD3+TCRgd+ T cells
promotes lymphoid potential during mouse and zebrafish em- (Figures 2F, 2G, S2E, and S2F), indicating that EZH1 knockdown
bryonic development (Vo et al., 2018; Soto et al., 2021). To deter- promotes differentiation toward ab T cell fate rather than gd
mine whether the inhibition of EZH1 would facilitate in vitro T cell T cells. EZH1 knockdown also resulted in a decrease in CD1a,
differentiation from iPSCs, we performed shRNA-mediated which is expressed in thymocytes but not mature peripheral
EZH1 knockdown during T cell specification from iPSC-derived blood T cells, further demonstrating that EZ-T cells exhibit a
CD34+ HE cells (Figures S1B and S1C). EZH1-knockdown HE more mature T cell phenotype (Figures 2H and S2G). Unlike
cells were then induced to differentiate into EZ-T cells via the mature, conventional cytotoxic T cells that express the CD8ab
stroma-free system and compared with control iPSC-SF-T heterodimer, innate-like gd T cells or intestinal intraepithelial lym-
cells derived as above (Figure 2A). EZH1 knockdown produced phocytes (IELs) express CD8 as a CD8aa homodimer, which has
a 2-fold increase in live cells after 2 weeks of differentiation been considered a less robust co-receptor and may even sup-
(Figure 2B) and after 6 weeks resulted in both a higher proportion press TCR activation (Bosselut et al., 2000; Holler and Kranz,
of T cells among CD45+ hematopoietic cells and an increased 2003; Cheroutre and Lambolez, 2008). Studies using OP9-DL1
absolute number of T cells generated from each starting stroma have yielded CD8aa T cells (Themeli et al., 2013; Maeda
CD34+ cell, suggesting enhanced T cell differentiation specificity et al., 2016), while recent protocols using 3D artificial thymic or-
and efficiency (Figures 2C, 2D, and S1D). Similar results were ganoids or a Notch ligand-based feeder-free system have
obtained in multiple iPSC lines that were derived from distinct yielded iPSC-T cells that express CD8ab (Montel-Hagen et al.,
donors using different reprogramming strategies, indicating 2019; Iriguchi et al., 2021). Similarly, our stroma-free differentia-
that the EHZ1-knockdown-mediated enhancement of T cell dif- tion protocol supports the production of CD8ab T cells, with
ferentiation was not cell line restricted (Figure S1E). As an alter- EZH1 knockdown promoting higher yields of CD8ab T cells
native to the shRNA-mediated EZH1 knockdown, we engineered and barely detectable quantities of innate-like CD8aa T cells
a doxycycline-inducible CRISPR interference (CRISPRi) (Figures 2I and S2H). Collectively, these analyses revealed that
construct into iPSC-derived CD34+ HE cells to transcriptionally repression of EZH1 promotes efficient in vitro differentiation of
repress EZH1 expression (Figure S2A). Interestingly, CRISPRi- iPSC cells into mature SP T cells.
mediated EZH1 knockdown during T cell specification (weeks
0–2) led to a significant increase in the production of CD3+ EZ-T cells display a molecular signature similar to that
T cells, while induction of EZH1-CRISPRi after the formation of of peripheral blood TCRab T cells
proT cells (weeks 2–5) failed to promote T cell differentiation (Fig- To assess the extent to which our iPSC-T cells resemble their
ure S2B). In control cells, the expression of EZH1 significantly in vivo counterparts, we evaluated the fidelity of stroma-free
increased during specification of HE into proT cells and was in vitro T cell differentiation via CellNet, a network biology plat-
downregulated after the proT stage (Figure S2C). EZH2, a homo- form used to analyze the faithfulness of cell fate conversions
log of EZH1 that likewise acts as a catalytic subunit of the PRC2 and the similarity between in vitro-derived cell types and gold-
complex, was highly expressed at the later stages of T cell differ- standard native-tissue equivalents (Cahan et al., 2014; Radley
entiation in both control and EZH1 shRNA-treated cells (Fig- et al., 2017). Analysis of RNA-seq gene expression profiles by
ure S2D). Such an observation is in agreement with previous CellNet revealed a high degree of similarity between iPSC-
findings that EZH1, but not EZH2, functions in HE cells to inhibit derived T cells and donor-derived T cells isolated from peripheral
lymphoid lineage commitment (Vo et al., 2018) and explains why blood mononuclear cells (PBMC), and a clear discrimination
the inhibition of EZH1 had no impact on later stages of T cell dif- from less-differentiated multipotent HSPCs (Figures 3A and
ferentiation. After 6 weeks of differentiation, the control iPSC-SF- S3A). We next sought a more refined analysis and therefore per-
T cells contained a substantial proportion of DP T cells, whereas formed RNA-seq to compare the gene expression profile of EZ-T
the EZ-T cells consisted predominantly of CD8 or CD4 SP cells cells with PBMC-derived TCRab T cells, PBMC-TCRgd T cells,
(Figure 2E). In contrast to the limited production of SP T cells and PBMC-NK cells, as well as T cells generated from CB
when differentiated on OP9-DL1 stroma, (La Motte-Mohs CD34+ HSPCs or iPSCs in the absence of EZH1 knockdown
et al., 2005; Montel-Hagen and Crooks, 2019), our data demon- via in vitro differentiation on OP9-DL1 stroma or our stroma-
strate that a combination of stroma-free differentiation with free system. Examining the expression of T cell signature genes,
EZH1-knockdown supports more efficient induction of SP cells. we found that iPSC-T cells differentiated via our stroma-free pro-
As previous studies have shown that iPSC-derived T cells tend tocol without EZH1 knockdown (PSC-SF-T) were more similar to
to resemble innate-like gd T cells (Nishimura et al., 2013; Viz- CB-HSPC-derived T cells than were iPSC-T cells differentiated
cardo et al., 2013; Themeli et al., 2013), we performed immuno- via OP9-DL1 stroma (Figure 3B). In contrast, the EZ-T cells
A B
C D E
F G
H I
were most similar to PBMC TCRab T cells. Furthermore, we that was most similar to ab T cells (Figure 3C). To investigate the
compared the transcriptional signature of PBMC-derived TCRab molecular mechanism underlying the mature phenotype of EZ-T
T with that of TCRgd T cells and identified a list of genes that can cells, we performed gene set enrichment analysis (GSEA) on the
be used to distinguish ab T from gd T cells. Expression levels of most significantly upregulated genes in EZ-T cells compared
these genes were determined across all cell types, and the result with control iPSC-SF-T cells (lacking EZH1 knockdown), and
again showed that EZ-T cells exhibited a gene expression profile observed that these genes were highly enriched in biological
A B C
D E
processes directly associated with T cell differentiation or func- pressed TRAC, TRBC2, CD2, and CD7, and showed a greater
tion (Figures 3D and 3E). To access whether the differences iden- downregulation of residue ‘‘innate’’ genes (TRDC, KLRB1) (Fig-
tified by bulk RNA-seq were due to changes in cell type compo- ure S3B). Collectively, EZH1 repression during in vitro iPSC dif-
sitions, we performed single-cell RNA sequencing (scRNA-seq) ferentiation promotes the production of ab T cells that display
analysis on control iPSC-SF-T cells and EZ-T cells and a more mature phenotype.
compared gene expression profiles for the sub-populations of We next performed immunosequencing to determine the TCR
TCRab T cells (TRAC+TRDC-). A relatively small number of genes repertoire of EZ-T cells and observed a high degree of TCRb
were differentially expressed between control iPSC-SF ab T cells diversity with no preferential Vb gene usage (Figure 4A), suggest-
and those with EZH1 knockdown (Table S1). Interestingly, ing that EZH1-knockdown did not cause significant
TCRab T cells with EZH1 knockdown more abundantly ex- clonal expansion of individual T cells with specific TCR
B C
rearrangements. Notably, EZ-T cells displayed longer CDR3 re- from iPSCs have previously been shown to display a naive
gions than iPSC-SF-T cells without EZH1 knockdown (Fig- phenotype (Seet et al., 2017; Iriguchi et al., 2021), but which
ure 4B). iPSC-derived T cells tend to have shorter CDR3 regions T cell subsets can be derived from naive iPSC-T cells is still
than mature peripheral blood T cells, likely due to lower expres- largely unknown. To answer this question, we used scRNA-seq
sion levels of terminal deoxynucleotidyl transferase (TdT), the to profile the distinct changes and characterize the T cell subsets
enzyme responsible for random nucleotide insertion during that arise during T cell differentiation and activation. Upon the
VDJ recombination, which is encoded by the DNTT gene (Mon- completion of EZ-T cell differentiation, we sorted CD45+ he-
tel-Hagen et al., 2019). To test this, we determined the expres- matopoietic cells before and after T cell activation and generated
sion levels of TdT/DNTT in both iPSC-derived HE cells and 11,131 single-cell transcriptomes. We identified eight clusters
week-4 DP T cells harboring control or EZH1 shRNA. As ex- across all samples, with the majority of cells manifesting a
pected, TdT/DNTT was only expressed in DP T cells and more T cell fate (CD5+) (Figures 5A and 5B). Consistent with previous
abundantly expressed in cells with EZH1-knockdown (Fig- immunophenotypic analyses, cells were predominantly CD8
ure 4C). These data suggest that EZH1-knockdown during SP cells, with lower numbers of CD4 SP and DP T cells (Fig-
iPSC differentiation promotes T cell maturation, with the result- ure 5B). Further analyses on the signature gene expression
ing EZ-T cells exhibiting molecular signatures that most closely profiles revealed only small proportions of innate NK-like
resemble peripheral blood TCRab T cells. (CD56+CD5-KLRB1+) or gd T cells (TRDC+TRGC1+). For
the mature T cell compartment, we identified two naive-like
EZ-T cell subsets display effector and memory-like T cell clusters (CCR7+SELL+IL2RAlowLEF1high) distinguished
phenotypes by their cycling status (Willinger et al., 2006), two effector-
After exiting the thymus, newly produced naive T cells give rise to like clusters (CCR7-SELL-GZMBhighGZMAhighNKG7+), and
effector and memory subsets, which are characterized by notably, a memory-like T cell cluster that expresses low levels
distinct phenotypic and functional features and cumulatively of cytotoxicity-related genes (GZMB, NKG7) and highly
shape T cell immunity (Kumar et al., 2018). T cells generated expresses genes that have been linked to memory T cell
A B
F G
Figure 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.
identities (CCR7+SELL+IL7R+CD2highCCL5highFAShighEOMEShigh) et al., 2019) (Figure S5A). Almost all the CD45+ cells derived
(Sanders et al., 1988; Huster et al., 2004; Intlekofer et al., 2005; via EZ-T cell differentiation overlapped with peripheral blood
Marçais et al., 2006). We further validated the presence of a T cells; cells that represent early HSPCs or other non-lymphoid
memory-like T cell subset by detecting the expression of lineages were barely detectable (Figure S5B). Consistent with
CD45RA, CD45RO, and CCR7 (Figure S4A). Moreover, it is previous analysis, following activation, a subset of EZ-T cells
known that cytotoxic T cells can express NK cell genes, emerged as CD8 central memory T cells (Figure S5B). A cross
including inhibitory NK cell receptors that raise the threshold of comparison between iPSC-derived T cells with their in vivo coun-
TCR stimulation and dampen T cell responses (Vivier and An- terparts also indicates that the memory-like CD8 cluster in EZ-T
fossi, 2004). Among the inhibitory NK cell receptors, KLRB1 is cells closely correlates with peripheral blood central memory
expressed by tumor infiltrating effector T cells for several types CD8 T cells (Figure S5C). Taken together, iPSC-derived EZ-T
of human cancer, negatively regulating their antitumor activity cells recapitulate the differentiation of naive T cells that give
(Mathewson et al., 2021). A more recent study further identified rise to effector cells and T cell subsets that exhibit a memory-
KLRB1, together with other NK cell receptors, as a hallmark of like phenotype.
exhausted, dysfunctional CAR T cells (Good et al., 2021). In
our cell populations, the memory-like T cell cluster expressed CAR T cells generated from EZ-T cells exhibit enhanced
lower levels of NK cell genes, including KLRB1, which further dis- antitumor activity
tinguishes these cells from the terminally differentiated effector- Having shown that EZ-T cells display molecular features similar
like T cell clusters (Figure 5C). In addition to the expression of to those of mature peripheral blood TCRab T cells, we next per-
major marker genes, GSEA analysis indicated a gene expression formed functional characterizations of effector cell properties.
profile similar to that of memory T cells rather than naive or Compared to iPSC-OP9-T cells or iPSC-SF-T cells, EZ-T cells
effector T cells (Figure 5D). None of the clusters showed sub- showed more robust upregulation of CD69 in response to
stantial expression of inhibitory receptors or regulatory T cell PMA/ionomycin treatment (Figure 6A). Consistently, EZ-T cells
markers (Figure S4B). expressed higher levels of CD107a than control iPSC-SF-T cells
We next sought to capture the compositional changes in im- upon PMA/ionomycin stimulation; the degranulation efficiency
mune cell clusters during T cell expansion. Compared with unac- was comparable to peripheral blood-derived T cells (Figure 6B).
tivated cells, T cell expansion resulted in enrichment of mature In light of the EZ-T cells’ superior activation/degranulation effi-
T cell populations and a concomitant reduction of immature ciency, we next explored whether EZ-T cells could be used to
DP T cells and innate-like cells (NK-like cells, gd T-like cells). generate CAR T cells with enhanced cytotoxic effector functions.
Similarly, naive-like T cells were predominantly present in unac- We transduced control iPSC-OP9-T cells, iPSC-SF-T cells, EZ-T
tivated samples and significantly reduced upon activation. cells, and donor-derived peripheral blood T cells with anti-CD19
Importantly, T cells displaying a memory-like phenotype were CARs containing a 4-1BB costimulatory domain, and co-
exclusively detected in activated cells after extended expansion, cultured with two different types of CD19+ lymphoma cells to
suggesting the occurrence of cell fate conversions from naive- compare their cytotoxicity profiles. CD19 CAR EZ-T cells caused
like cells into more differentiated subsets (Figures 5E and 5F). more efficient specific target cytolysis against both Jeko-1 and
To examine this, we performed gene regulatory network (GRN) OCI-Ly1 cells than CAR T cells derived from control iPSC-OP-
analysis using CellRouter, aiming to reconstruct the trajectories 9 and iPSC-SF-T cells and displayed a specific killing capacity
of cell-state transitions from naive to memory-like T cells (Lum- comparable to PBMC-T cells (Figures 6C and 6D). Cytotoxicity
mertz da Rocha et al., 2018). We identified a series of key tran- assays were also performed using presorted iPSC-SF TCRab
scriptional networks governing the changes in T cell composition T cells with or without EZH1 knockdown. Similarly, TCRab
and found multiple top-ranked transcriptional regulators, T cells with EZH1 knockdown elicited more efficient killing of
including BATF, LYAR, LITAF, IRF4, and RUNX3, which were target tumor cells (Figure S6A). Moreover, co-culture with tumor
previously known to drive the development of long-lived memory cells triggered EZ-T cells to secrete higher levels of cytokines
T cells (Figure 5G) (Seo et al., 2021; Chen et al., 2020; Mackay that are essential for T cell antitumor responses, including IL-2,
et al., 2013; Harberts et al., 2021; Wang et al., 2018b). Addition- interferon-g (IFN-g), and tumor necrosis factor a (TNFa)
ally, we performed single-cell regulatory network inference and (Figures 6E–6G). Both control SF-T and EZ-T cells produced
clustering (SCENIC) analysis to map regulons enriched in each lower levels of cytokines than PBMC-T cells. Because iPSC-
annotated cell type (Aibar et al., 2017) (Figure S4C). Consistent derived T cells are predominantly CD8+ cytotoxic cells, whereas
with the CellRouter analysis, SCENIC identified a similar set of the PBMC-T cells include a large proportion of CD4+ cells, this
regulons associated with the memory-like subset. In particular, observation is consistent with the higher cytokine production ca-
regulatory networks that have been linked to the generation pacity/profile of CD4 T cells (Pfizenmaier et al., 1984; Ngai et al.,
and homeostasis of memory T cells, such as BATF and IRF9 2007). Collectively, these data indicate that EZ-T cells exhibit
regulons, were exclusively enriched in the memory-like cluster enhanced cytotoxic and cytokine-producing effector functions
(Kurachi et al., 2014; Martinet et al., 2015; Seo et al., 2021) (Fig- against tumor cells in vitro.
ure S4D). Finally, we compared EZ-T cells with a range of he- To further evaluate the efficacy of EZ-T cell-derived CAR
matopoietic cells by mapping our scRNA-seq data on a publicly T cells, we established a xenograft mouse model by intrave-
available reference dataset that includes hematopoietic stem nously injecting luciferase-expressing diffuse large B cell lym-
cells (HSCs), lineage-restricted blood progenitors, and terminally phoma (DLBCL) cells (OCI-Ly1) into immunodeficient non-obese
differentiated lymphoid/erythroid/myeloid cells collected from diabetic-SCID IL2Rgammanull (NSG) mice. These animals were
healthy bone marrow and peripheral blood samples (Granja then infused with PBS, CD19 CAR iPSC-SF-T cells, CD19 CAR
A B
C D
E F G
EZ-T cells, or PBMC-derived CD19 CAR T cells, and subjected cause abnormal expansion of EZ-T cells, we administrated
to weekly bioluminescence imaging (BLI) to assess tumor both control and EZ-T cells into healthy mice and monitored
burden (Figure 7A). Although CAR T cells generated from the presence of CAR T cells over a long time period. Without
iPSC-SF-T cells suppressed tumor growth, they failed to eradi- simultaneous tumor engraftment, CAR T cells were barely
cate tumor cells in any animal after 7 weeks. Notably, CAR detectable in peripheral blood after 5 weeks, and these animals
EZ-T cells displayed significant improvement of efficacy and remained healthy and were free of human T cells after 20 weeks
were capable of eradicating tumor cells and caused complete (Figure S7B). In summary, when engineered with anti-CD19
remissions in most animals (Figure 7B). Consistent with CARs, EZ-T cells elicit superior antitumor effects relative to con-
improved tumor clearance, more CAR EZ-T cells were detected trol iPSC-SF-T cells lacking EZH1 knockdown, both in vitro and
in peripheral blood than control CAR iPSC-SF-T cells 3 weeks af- in vivo.
ter injection (Figure 7C), and most circulating EZ-T cells ex-
pressed abTCR and not gd TCR (Figure S6B), suggesting that DISCUSSION
the enhanced persistence of EZ-T cells is largely due to the
enrichment of TCRab T cells. As a result, though some mice The identification of signaling pathways that are essential for
showed persistent BLI signal intensity, the injection of CAR T cell development has led to the design of protocols that allow
EZ-T cells produced comparable survival rates to PBMC CAR the generation of T cells in vitro from human pluripotent stem
T cell treatment for the 7-week duration of the experiment cells (Holmes and Zúñiga-Pflu€cker, 2009; Timmermans et al.,
(Figures 7D and S7A). To exclude that EZH1 repression may 2009; Seet et al., 2017). However, past in vitro differentiation
C
D
approaches have been plagued by dependency on mouse stro- similar to T cells differentiated from CB CD34+ HSPCs. More-
mal cells as well as deficiencies in the terminal stages of T cell over, by avoiding the cumbersome co-culture with mouse feeder
maturation, thereby limiting their clinical application in adoptive cells, the stroma-free protocol minimizes batch-to-batch varia-
immunotherapy. The generation of definitive (adult-type) HSCs tion that can confound phenotypic characterization. Leveraging
with lymphoid potential and differentiation of functional T cells this stroma-free differentiation platform, we further demonstrate
from iPSCs has proven difficult, as in vitro differentiation of iPSCs that repression of EZH1 expression during lymphoid specifica-
tends to default into the production of embryonic cell types (Dou- tion facilitates T cell differentiation from human iPSCs and leads
latov et al., 2013; Sugimura et al., 2017). Past studies have to robust generation of developmentally mature iPSC-derived
demonstrated that iPSC-derived T cells resemble innate-like T cells. Importantly, while iPSC-SF-T cells exhibit some innate-
gd T cells and are not as robustly functional as primary ab like phenotypes that have been previously reported in OP9-
T cells, again suggesting that our ability to recapitulate the mech- stroma dependent iPSC differentiation systems (Themeli et al.,
anisms underlying commitment and progression of T cell devel- 2013), EZ-T cells display molecular signatures resembling pe-
opment is incomplete. Screening of critical epigenetic modifying ripheral blood TCRab T cells. These results are in agreement
enzymes during the generation of HSPCs discovered that EZH1 with the hypothesis that EZH1 knockdown produces definitive
plays a central role in regulating multipotency and lymphoid po- progenitors that preferentially support adult-like lymphopoiesis
tential in embryonic blood progenitors (Vo et al., 2018). As a rather than the generation of immature primitive embryonic
component of PRC2, EZH1 modulates chromatin accessibility lymphoid cells. Such a mechanism has also been supported
by mediating histone H3 lys27 trimethylation (H3K27me3) by recent findings that hPSC-derived CD34+ progenitors with a
(Shen et al., 2008). During embryonic hematopoiesis, EZH1 re- primary phenotype, defined by restricted hematopoietic poten-
presses the transcription of genes associated with definitive tial, produce fetal-like NK cells via in vitro differentiation (Dege
hematopoietic fates, and EZH1 deficiency in genetically engi- et al., 2020). In contrast, the multipotent CD34+ progenitors
neered mice promotes the precocious emergence of bona fide with full lymphoid potential are capable of giving rise to adult-
HSC and lymphoid progenitors (Vo et al., 2018). A recent study like NK cells that are functionally distinct from their fetal counter-
in the zebrafish model further showed that EZH1 suppresses parts (Dege et al., 2020).
HSPC formation by regulating HE commitment. Specifically, Another focus of our study was to test the impact of EZH1
EZH1 enhances Notch signaling to facilitate arterial gene expres- repression on the functional properties of iPSC-derived T cells
sion at the expense of HE specification and HSPC development. and to evaluate the potential of using EZ-T cells for adoptive
As a result, the knockdown of EZH1 unlocks definitive hemato- T cell immunotherapy. We engineered iPSC-derived T cells
poiesis and leads to the enhanced production of multipotent with anti-CD19 CARs and assessed their antitumor capacities.
HSPCs with lymphoid potential (Soto et al., 2021). Compared with CAR T cells generated from iPSC-SF-T cells,
In light of the repressive function of EZH1 in lymphopoiesis, we CD19-CAR EZ-T cells exhibit superior antitumor activity,
investigated the impact of EZH1 knockdown during in vitro T cell measured by tumor cell killing and cytokine production against
differentiation. Although 3D thymic-like culture systems have different types of CD19+ tumor cells in vitro and elicit more effi-
been established to mimic mouse or human T cell development cient tumor clearance in a xenograft mouse model. Notably,
and yield iPSC-derived T cells that display a mature phenotype, scRNA-seq analysis revealed that EZ-T cells, after activation,
these methods rely on engineered stromal cells or mouse- give rise to a subset of T cells that express relatively low levels
derived fetal thymic cells (Vizcardo et al., 2018; Montel-Hagen of cytotoxicity genes and high levels of memory T cell signature
et al., 2019; Wang et al., 2022). Therefore, we developed a genes. Such an observation is consistent with previous findings
stroma-free system that supports efficient T cell differentiation that in vitro T cell stimulation allows faster effector/memory
without using mouse-derived feeder cells. Such a strategy based commitment than physiological T cell differentiation/expansion
on immobilized Notch ligands has recently been employed to (Li and Kurlander, 2010; McLellan and Ali Hosseini Rad, 2019).
induce the iPSC-derived CD34+ HSPCs to differentiate into Trajectory analyses based on the activity of GRNs also identified
CD3+TCRab+CD8ab T cells that are immunophenotypically transcriptional regulators that drive the conversion of naive-like
similar to our iPSC-SF-T cells (Iriguchi et al., 2021; Trotman- EZ-T cells into memory-like cells. Among these networks are
Grant et al., 2021). However, whether such stroma-free differen- BATF, IRF4/9, and RUNX3, all of which are known to promote
tiation could support normal TCR rearrangement remained un- T cell longevity or prevent exhaustion (Martinet et al., 2015;
clear. Here, we show that the stroma-free system can faithfully Huber et al., 2017; Wang et al., 2018b; Istaces et al., 2019;
recapitulate T cell development by differentiating non-T cell- Seo et al., 2021). This is of particular interest as a growing
derived iPSCs (without pre-rearranged TCRs) into T cells with a body of evidence has shown that the enrichment of memory
high degree of TCR diversity. Compared with iPSC-OP9-T cells, T cells rather than terminally differentiated effector cells corre-
iPSC-SF-T cells exhibit a marginally more mature phenotype, lates with superior T cell persistence and improved clinical
Figure 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/s).
(C) Numbers of CAR T cells per 100 mL 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).
iPSCs are more amenable to genetic manipulations and could B Quantitative Real-Time PCR analysis
B Flow cytometry
facilitate the engineering of ‘‘armored’’ CAR T cells that can
secrete specific cytokines or checkpoint inhibitor antibodies to B Bulk RNA-seq and data analysis
and are currently being tested in multiple clinical trials (Basar d QUANTIFICATION AND STATISTICAL ANALYSIS
et al., 2020; Liu et al., 2021). Compared with T cells, NK cell cyto-
SUPPLEMENTAL INFORMATION
toxicity is not constrained by MHC recognition and could target
tumor cells that are resistant to T cell killing. Moreover, NK cells Supplemental information can be found online at https://doi.org/10.1016/j.
are less prone to GVHD and CAR-associated toxicity (Ruggeri stem.2022.06.014.
et al., 2002; Liu et al., 2020). Multiple studies have successfully
generated iPSC-derived NK cells that elicit antitumor activities ACKNOWLEDGMENTS
(Knorr et al., 2013; Zeng et al., 2017; Li et al., 2018; Woan
We thank the Boston Children’s Hospital Flow Cytometry Core and the hESC
et al., 2021). How EZH1-mediated regulation of lymphoid
Core Facility. We also thank John Atwater, Merve Akyol, and Tamer Onder for
commitment might affect NK cell differentiation from iPSCs re- assistance with experiments and advice. This study was supported by grants
mains an open question. from NIH NHLBI U01HL134812 (G.Q.D.), NIH NIDDK 2U01DK104218 (G.Q.D.),
NIH NHLBI 5U24HL134763 subaward 1701192 (R.J.), the Emerson Collective
Limitations of the study Cancer Research Fund (G.Q.D.), and philanthropic support from the Manton
Although our data suggest that EZ-T cells show comparable Foundation.
efficacy to donor-derived CAR T cells, there are limitations
AUTHOR CONTRIBUTIONS
to be addressed in future studies. First, iPSC-derived EZ-T
cells produced by existing methods predominantly consist R.J. and G.Q.D. conceived the project and designed the experiments. R.J.
of CD8 cytotoxic T cells. Therefore, new strategies are needed performed iPSC and iPSC-T cell studies. R.J. and I.S. performed CAR T cell
functional assays. M.N., E.L.d.R., and M.F. analyzed scRNA-seq data. A.H.
to enable the efficient production of mature CD4 SP T cells, as
and M.N. analyzed bulk RNA-seq data. T.B. and M.N. generated CRISPRi
a more balanced ratio of cytotoxic versus helper T cells has
iPSC lines. R.J. and M.S. performed studies in the xenograft mouse model.
been associated with improved therapeutic responses (Som- C.K. and D.J. participated in experimental optimization and data interpreta-
mermeyer et al., 2016; Wang et al., 2018a). Second, our cur- tion. T.M.S., T.E.N., M.V.M., and G.Q.D. participated in project planning, su-
rent methods for repressing EZH1 activity during T cell differ- pervised research, and interpreted data. R.J. and G.Q.D. wrote the manuscript
entiation employ viral vectors that integrate into host DNA. with input from all co-authors.
Though EZH1/2 dual inhibitors have been shown to promote
DECLARATION OF INTERESTS
NK cell development (Damele et al., 2021), small molecule
treatment fails to phenocopy shRNA-mediated EZH1 knock- R.J., G.Q.D., and Boston Children’s Hospital hold intellectual property and
down during iPSC-T cell differentiation. This is consistent receive consulting fees and/or hold equity interest relevant to the generation
with previous findings that EZH1/EZH2 double knockdown 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.
had no effects on T cell potential (Vo et al., 2018). Given the
lack of EZH1-specific inhibitors that do not repress EZH2
Received: February 2, 2022
function, the development of non-integrating gene knockdown Revised: May 31, 2022
strategies will be needed to facilitate translation and clinical Accepted: June 29, 2022
applications of EZ-T cells. Published: August 4, 2022
Klebanoff, C.A., Gattinoni, L., Torabi-Parizi, P., Kerstann, K., Cardones, A.R., McLellan, A.D., and Ali Hosseini Rad, S.M. (2019). Chimeric antigen receptor
Finkelstein, S.E., Palmer, D.C., Antony, P.A., Hwang, S.T., Rosenberg, S.A., T cell persistence and memory cell formation. Immunol. Cell Biol. 97, 664–674.
et al. (2005). Central memory self/tumor-reactive CD8+ T cells confer superior Montel-Hagen, A., and Crooks, G.M. (2019). From pluripotent stem cells to
antitumor immunity compared with effector memory T cells. Proc. Natl. Acad. T cells. Exp. Hematol. 71, 24–31.
Sci. USA 102, 9571–9576.
Montel-Hagen, A., Seet, C.S., Li, S., Chick, B., Zhu, Y., Chang, P., Tsai, S.,
Knorr, D.A., Ni, Z., Hermanson, D., Hexum, M.K., Bendzick, L., Cooper, L.J., Sun, V., Lopez, S., Chen, H.C., et al. (2019). Organoid-induced differentiation
Lee, D.A., and Kaufman, D.S. (2013). Clinical-scale derivation of natural killer of conventional T cells from human pluripotent stem cells. Cell Stem Cell 24,
cells from human pluripotent stem cells for cancer therapy. Stem Cells 376–389.e8.
Transl. Med. 2, 274–283.
Ngai, P., McCormick, S., Small, C., Zhang, X., Zganiacz, A., Aoki, N., and Xing,
Kumar, B.V., Connors, T.J., and Farber, D.L. (2018). Human T cell develop- Z. (2007). Gamma interferon responses of CD4 and CD8 T-cell subsets are
ment, localization, and function throughout life. Immunity 48, 202–213. quantitatively different and independent of each other during pulmonary
Kurachi, M., Barnitz, R.A., Yosef, N., Odorizzi, P.M., DiIorio, M.A., Lemieux, Mycobacterium bovis BCG infection. Infect. Immun. 75, 2244–2252.
M.E., Yates, K., Godec, J., Klatt, M.G., Regev, A., et al. (2014). The transcrip- Nishimura, T., Kaneko, S., Kawana-Tachikawa, A., Tajima, Y., Goto, H., Zhu,
tion factor BATF operates as an essential differentiation checkpoint in early D., Nakayama-Hosoya, K., Iriguchi, S., Uemura, Y., Shimizu, T., et al. (2013).
effector CD8+ T cells. Nat. Immunol. 15, 373–383. Generation of rejuvenated antigen-specific T cells by reprogramming to plurip-
€cker, J.C. (2005). Induction of
La Motte-Mohs, R.N., Herer, E., and Zúñiga-Pflu otency and redifferentiation. Cell Stem Cell 12, 114–126.
T-cell development from human cord blood hematopoietic stem cells by delta- Pegram, H.J., Purdon, T.J., van Leeuwen, D.G., Curran, K.J., Giralt, S.A.,
like 1 in vitro. Blood 105, 1431–1439. Barker, J.N., and Brentjens, R.J. (2015). IL-12-secreting CD19-targeted cord
Li, Y., Hermanson, D.L., Moriarity, B.S., and Kaufman, D.S. (2018). Human blood-derived T cells for the immunotherapy of B-cell acute lymphoblastic leu-
iPSC-derived natural killer cells engineered with chimeric antigen receptors kemia. Leukemia 29, 415–422.
enhance anti-tumor activity. Cell Stem Cell 23, 181–192.e5. €ubener, W., Krönke, M., Röllinghoff, M., and
Pfizenmaier, K., Scheurich, P., Da
Li, Y., and Kurlander, R.J. (2010). Comparison of anti-CD3 and anti-CD28- Wagner, H. (1984). Quantitative representation of all T cells committed to
coated beads with soluble anti-CD3 for expanding human T cells: differing develop into cytotoxic effector cells and/or interleukin 2 activity-producing
impact on CD8 T cell phenotype and responsiveness to restimulation. helper cells within murine T lymphocyte subsets. Eur. J. Immunol. 14, 33–39.
J. Transl. Med. 8, 104. Radley, A.H., Schwab, R.M., Tan, Y., Kim, J., Lo, E.K.W., and Cahan, P. (2017).
Liu, E., Marin, D., Banerjee, P., Macapinlac, H.A., Thompson, P., Basar, R., Assessment of engineered cells using CellNet and RNA-seq. Nat. Protoc. 12,
Nassif Kerbauy, L., Overman, B., Thall, P., Kaplan, M., et al. (2020). Use of 1089–1102.
CAR-transduced natural killer cells in CD19-positive lymphoid tumors. Rafiq, S., Yeku, O.O., Jackson, H.J., Purdon, T.J., van Leeuwen, D.G., Drakes,
N. Engl. J. Med. 382, 545–553. D.J., Song, M., Miele, M.M., Li, Z., Wang, P., et al. (2018). Targeted delivery of a
Liu, S., Galat, V., Galat, Y., Lee, Y.K.A., Wainwright, D., and Wu, J. (2021). NK PD-1-blocking scFv by CAR-T cells enhances anti-tumor efficacy in vivo. Nat.
cell-based cancer immunotherapy: from basic biology to clinical development. Biotechnol. 36, 847–856.
J. Hematol. Oncol. 14, 7. Ruggeri, L., Capanni, M., Urbani, E., Perruccio, K., Shlomchik, W.D., Tosti, A.,
Lummertz da Rocha, E., Kubaczka, C., Sugden, W.W., Najia, M.A., Jing, R., Posati, S., Rogaia, D., Frassoni, F., Aversa, F., et al. (2002). Effectiveness of
Markel, A., LeBlanc, Z.C., Dos Santos Peixoto, R., Falchetti, M., Collins, J.J., donor natural killer cell alloreactivity in mismatched hematopoietic transplants.
et al. (2022). CellComm infers cellular crosstalk that drives haematopoietic Science 295, 2097–2100.
stem and progenitor cell development. Nat. Cell Biol. 24, 579–589. Sanders, M.E., Makgoba, M.W., Sharrow, S.O., Stephany, D., Springer, T.A.,
Lummertz da Rocha, E., Rowe, R.G., Lundin, V., Malleshaiah, M., Jha, D.K., Young, H.A., and Shaw, S. (1988). Human memory T lymphocytes express
Rambo, C.R., Li, H., North, T.E., Collins, J.J., and Daley, G.Q. (2018). increased levels of three cell adhesion molecules (LFA-3, CD2, and LFA-1)
Reconstruction of complex single-cell trajectories using CellRouter. Nat. and three other molecules (UCHL1, CDw29, and Pgp-1) and have enhanced
Commun. 9, 892. IFN-gamma production. J. Immunol. 140, 1401–1407.
Mackay, L.K., Rahimpour, A., Ma, J.Z., Collins, N., Stock, A.T., Hafon, M.L., Sanson, K.R., Hanna, R.E., Hegde, M., Donovan, K.F., Strand, C., Sullender,
Vega-Ramos, J., Lauzurica, P., Mueller, S.N., Stefanovic, T., et al. (2013). M.E., Vaimberg, E.W., Goodale, A., Root, D.E., Piccioni, F., and Doench,
The developmental pathway for CD103(+)CD8+ tissue-resident memory J.G. (2018). Optimized libraries for CRISPR-Cas9 genetic screens with multiple
T cells of skin. Nat. Immunol. 14, 1294–1301. modalities. Nat. Commun. 9, 5416.
MacLeod, D.T., Antony, J., Martin, A.J., Moser, R.J., Hekele, A., Wetzel, K.J., Scarfò, I., Ormhøj, M., Frigault, M.J., Castano, A.P., Lorrey, S., Bouffard, A.A.,
Brown, A.E., Triggiano, M.A., Hux, J.A., Pham, C.D., et al. (2017). Integration of van Scoyk, A., Rodig, S.J., Shay, A.J., Aster, J.C., et al. (2018). Anti-CD37
a CD19 CAR into the TCR alpha chain locus streamlines production of alloge- chimeric antigen receptor T cells are active against B- and T-cell lymphomas.
neic gene-edited CAR T cells. Mol. Ther. 25, 949–961. Blood 132, 1495–1506.
Maeda, T., Nagano, S., Ichise, H., Kataoka, K., Yamada, D., Ogawa, S., Seet, C.S., He, C., Bethune, M.T., Li, S., Chick, B., Gschweng, E.H., Zhu, Y.,
Koseki, H., Kitawaki, T., Kadowaki, N., Takaori-Kondo, A., et al. (2016). Kim, K., Kohn, D.B., Baltimore, D., et al. (2017). Generation of mature T cells
Regeneration of CD8ab T cells from T-cell-derived iPSC imparts potent tumor from human hematopoietic stem and progenitor cells in artificial thymic orga-
antigen-specific cytotoxicity. Cancer Res. 76, 6839–6850. noids. Nat. Methods 14, 521–530.
Marçais, A., Coupet, C.A., Walzer, T., Tomkowiak, M., Ghittoni, R., and Marvel, Seo, H., González-Avalos, E., Zhang, W., Ramchandani, P., Yang, C., Lio, C.J.,
J. (2006). Cell-autonomous CCL5 transcription by memory CD8 T cells is regu- Rao, A., and Hogan, P.G. (2021). BATF and IRF4 cooperate to counter exhaus-
lated by IL-4. J. Immunol. 177, 4451–4457. tion in tumor-infiltrating CAR T cells. Nat. Immunol. 22, 983–995.
STAR+METHODS
Continued
REAGENT or RESOURCE SOURCE IDENTIFIER
FLT3 R&D Systems Cat#308-FK-250
TPO Peprotech Cat#300-18
IL-3 Peprotech Cat#200-03
rhDLL1/DLL4-Fc Invitrogen Cat#A42510
IL-7 R&D Systems Cat#207-IL-010
IL-15 STEMCELL Technologies Cat#78031
ImmunoCult human CD3/CD28 T cell STEMCELL Technologies Cat#10971
activator
VivoGlo Luciferin Promega Cat#P1041
Critical commercial assays
Embryoid Body Dissociation Kit Miltenyi Biotec Cat#130-096-348
CD34 MicroBead Kit Miltenyi Biotec Cat#130-046-702
LEGENDplex Hu Th1/Th2 Panel BioLegend Cat#741030
Bright-Glo Luciferase Assay System Promega Cat#E2610
Maxima cDNA Sythesis Kit Thermo Fisher Scientific Cat#FERK1641
RNeasy Plus Micro Kit QIAGEN Cat# 74034
Power SYBR Green PCR Master Mix Thermo Fisher Scientific Cat# 4367659
DNeasy Blood and Tissue Kit Qiagen Cat# 69504
Human TCRB ImmunoSEQ Assay Adaptive Biotechnologies N/A
Deposited data
Raw and processed RNA-seq data This paper GEO: GSE195667
Experimental models: Cell lines
Human 1157.2 iPSCs Daley lab N/A
Human 273 iPSCs BCH Stem Cell Core N/A
Human 1381.3 iPSCs BCH Stem Cell Core N/A
Jeko-1 cells Maus lab N/A
OCI-Ly1 cells Daley lab N/A
Experimental models: Organisms/strains
NOD-scid IL2Rgammanull (NSG) mice Jackson Laboratories Cat# 005557
Oligonucleotides
EZH1 CRISPRi gRNA: GGTGAGTGAG- Broad Institute Addgene#92385
TAAACAAGCC
DNTT forward primer: ORIGENE Cat#HP233227
5’-CAGAGCGTTCCTCATGGAGCTG-3’
DNTT reverse primer: ORIGENE Cat#HP233227
5’-GTGCTTGAAGCCACTCCAGAAC-3’
EZH1 forward primer: Integrated DNA Technologies N/A
5’-CACCACATAGTCAGTGCTTCCTG-3’
EZH1 reverse primer: Integrated DNA Technologies N/A
5’-AGTCTGACAGCGAGAGTTAGCC-3’
EZH2 forward primer: 5’- ORIGENE Cat#HP207764
GACCTCTGTCTTACTTGTGGAGC-3’
EZH2 reverse primer: 5’- ORIGENE Cat#HP207764
CGTCAGATGGTGCCAGCAATAG-3’
GAPDH forward primer: Integrated DNA Technologies N/A
5’-ACCCAGAAGACTGTGGATGG-3’
GAPDH reverse primer: Integrated DNA Technologies N/A
5’-TTCAGCTCAGGGATGACCTT-3’
Recombinant DNA
CROPseq-EZH1gRNA-Zeo plasmid This paper N/A
PB03-NDI-dCAS9-KRAB plasmid This paper N/A
(Continued on next page)
Continued
REAGENT or RESOURCE SOURCE IDENTIFIER
Software and algorithms
immunoSEQ Analyzer 3.0 Adaptive Biotechnologies https://www.immunoseq.com/analyzer/
R R https://www.r-project.org/
CellRouter Lummertz da Rocha et al. (2018) https://github.com/edroaldo/cellrouter
R package FUSCA Lummertz da Rocha et al. (2022) https://github.com/edroaldo/fusca
Symphony Kang et al. (2021) https://github.com/immunogenomics/
symphony
GraphPad Prism v8 Graphpad Software, Inc https://www.graphpad.com/
FlowJo 10.8.0 BD Bioscience https://www.flowjo.com/
GSEA Broad Institute https://www.gsea-msigdb.org/gsea/
index.jsp
Aura imaging software Spectral Instruments Imaging https://spectralinvivo.com/aura-imaging-
software/
BioRender BioRender https://biorender.com/
RESOURCE AVAILABILITY
Lead contact
Further information and requests for resources and reagents should be directed to and will be fulfilled by the lead contact, George Q.
Daley ([email protected]).
Materials availability
Plasmids/reagents generated in this study will be made available upon request.
Cell line
Human 1157.2 iPSCs established by the Stem Cell Core at Boston Children’s Hospital were maintained on Matrigel matrix (Corning,
354277) using Stemflex media (Gibco, A3349401) in 5% CO2 at 37 C.
METHOD DETAILS
collected for the generation CD34+ HE cells using a previous described protocol (Ditadi et al., 2015). Briefly, iPSC colonies were
scraped and transferred to ultra-low attachment plates and cultured with aggregation media contains BMP4 (10ng/ml, day 0-2),
bFGF (5ng/ml, day 1-2), CHIR99021 (3mM, day 2), and SB431542 (6mM, day2) to allow EB formation. From day 3, EBs were cultured
with StemPro-34 media (Gibco, 10639011) supplemented with VEGF (15ng/ml, day3-8), bFGF (5ng/ml, day3-8), SCF (50ng/ml, day
6-8), EPO (day6-8), IL-6 (day6-8), IL-11 (day6-8), and IGF-1 (day6-8). After 8 days of culture, EBs were dissociated using EB Disso-
ciation Kit (Miltenyi, 130096348), and HE cells were isolated by magnetic-activated cell sorting (MSCS) using the human CD34 Mi-
crobead Kit (Miltenyi, 130046702).
Immunoblot
Whole cell lysates were collected using NP-40 lysis buffer (Invitrogen, FNN0021) with protease and phosphatase inhibitor cocktail
(Thermo Scientific, 78443). 50mg total protein was separated by SDS–PAGE using Any kDTM Mini-protean TGX precast polyacryl-
amide gels (BioRad, 4569033), and transferred to PVDF membranes using the Trans-Blot Cell Transfer System (BioRad). Membranes
were incubated overnight with antibodies against TBP (Cell Signaling, 8515, 1:1000), EZH1 (Abcam, ab64850, 1:1000) at 4 C, fol-
lowed by incubation with horseradish peroxidase-conjugated secondary antibodies (1:2000) for 1 hour at room temperature. Chem-
iluminescence was detected using SuperSignal West Pico Plus Chemiluminescent substrate (Thermo Scientific, 34579).
Flow cytometry
Cells were stained with PI/DAPI (BD Biosciences, RUO) and antibodies at 1:100 dilution in PBS with 2% FBS for 30 min at room
temperature in the dark. BD LSRII and BD FACSAria II were used for flow cytometry analysis and cells sorting. Compensation
was performed by automated compensation with anti-mouse Igk and negative beads (BD Biosciences) using the BD FACSDiva soft-
ware. The following human antibodies were used: CD45RA-BV510 (BD Biosciences, H100), CD45RO-PE (BD Biosciences, UCHL1),
CCR7-APC (BD Biosciences, 2-L1-A), CD3-PE/Cy7 (Biolegend, SK7), CD8a-BV421 (BD Biosciences, RPA-T8), TCRab-APC (Bio-
legend, IP26), TCRgd-PE (Biolegend, B1), CD7-PE (BD Biosciences, M-T701), CD5-BV510 (BD Biosciences, UCHT2), CD4-PE/
Cy5 (Biolegend, RPA-T4), CD8b-APC(Miltenyi, REA715), CD45-APC/Cy7 (BD Biosciences, 2D1), CD279-BV421 (BD Biosciences,
EH12.1), CD366-APC (BD Biosciences, F38-2E2), CD223-PE (BD Biosciences, T47-530), CD107a-PE (Biolegend, LAMP-1), CD69
APC (Biolegend, FN50), CD33-APC (Biolegend, WM53).
supplemented with 20 IU/ml rhIL-2 beginning on day 0 of culture and were maintained at a constant cell concentration of 0.5 x106/mL
by counting every 2-3 days. On day 10 cells were de-beaded (when using Dynabeads) and used for assays. For cytotoxicity assays,
control PBMC or iPSC-T cells were co-cultured with luciferase-expressing Jeko-1 or OCI-Ly1 tumor cells at the indicated ratios for 18
hours. Luciferase activity was measured with a Synergy Neo2 luminescence microplate reader (Biotek). Percentage of specific lysis
was calculated as (total RLU / target cells only RLU) x100. Cell-free supernatants were collected for cytokine release assay. Levels of
cytokines were measured using a LEGENDplex Multiplex Assay Kit (Biolegend, 741030) following manufacture’s instructions.
The significance of the difference between control and experimental results generated from in vitro assays was determined by two
tailed student’s T test, and P < 0.05 was considered statistically significant. For in vivo experiments, survival results were presented in
a Kaplan-Meier survival plot and compared by log-rank (Mantel-Cox) test. Statistic parameters in each experiment (value of n, SEM,
or SD) are described in the figure legends.