Recent studies have shown that stem cell memory T (TSCM) cell-like properties are important for successful adoptive immunotherapy by the chimeric antigen receptor–engineered-T (CAR-T) cells. We previously reported that both human and murine-activated T cells are converted into stem cell memory-like T (iTSCM) cells by coculture with stromal OP9 cells expressing the NOTCH ligand. However, the mechanism of NOTCH-mediated iTSCM reprogramming remains to be elucidated. Here, we report that the NOTCH/OP9 system efficiently converted conventional human CAR-T cells into TSCM-like CAR-T, “CAR-iTSCM” cells, and that mitochondrial metabolic reprogramming played a key role in this conversion. NOTCH signaling promoted mitochondrial biogenesis and fatty acid synthesis during iTSCM formation, which are essential for the properties of iTSCM cells. Forkhead box M1 (FOXM1) was identified as a downstream target of NOTCH, which was responsible for these metabolic changes and the subsequent iTSCM differentiation. Like NOTCH-induced CAR-iTSCM cells, FOXM1-induced CAR-iTSCM cells possessed superior antitumor potential compared with conventional CAR-T cells. We propose that NOTCH- or FOXM1-driven CAR-iTSCM formation is an effective strategy for improving cancer immunotherapy.

Significance:

Manipulation of signaling and metabolic pathways important for directing production of stem cell memory–like T cells may enable development of improved CAR-T cells.

Chimeric antigen receptor–engineered T cells (CAR-T) transfer has been developed as a potent therapeutic approach for hematologic cancers (1–3). A recent clinical trial revealed that CAR-T therapy for leukemia achieved high rates of short-time complete remission, but was not able to sustain 1-year relapse-free survival in a proportion of patients (4). To manufacture CAR-T cells, T-cell receptors (TCR) and cytokines continuously stimulate T cells, resulting in T-cell exhaustion and dysfunction (5). These dysfunctional or exhausted CAR-T cells are easily expired and elevate the risk of relapse (6). It has been reported that CAR-T cells from completely responding patients with chronic lymphocytic leukemia (CLL) were enriched in memory-related genes (7) and sustained remission was associated with an elevated frequency of stem cell–like memory T (TSCM) phenotypes of CAR-T cells in patients (7, 8).

To improve the efficacy of CAR-T cells, various methods have been tried for tuning T-cell differentiation and memory formation (9–12). Less-differentiated memory phenotypes such as central memory T (TCM) and TSCM phenotypes govern high proliferative capacity, long-term survival, and in vivo durability (13, 14). Especially, TSCM cells provide long-term antitumor response and are considered to be fit for CAR-T therapy (15).

In general, differentiated T cells use glycolysis for proliferation in response to antigens, whereas memory T cells prefer to use fatty acid oxidation (FAO)–dependent oxidative phosphorylation (OXPHOS; refs. 16, 17), and OXPHOS-mediated ATP production contributes to a favorable and durable antitumor response in tumor microenvironments (18–20). Peroxisome proliferator activated receptor gamma coactivator 1α (PGC1A)–driven mitochondrial biogenesis and OXPHOS support a strong antitumor effect (21). Furthermore, a strong link between FAO-dependent OXPHOS and memory T-cell development has been reported. For example, cell-intrinsic lipolysis following intrinsic fatty acid synthesis is required for memory formation (17). Mitochondrial biogenesis and morphological changes by CD28 signaling promote memory T-cell generation (22), and selective agents modifying mitochondrial metabolism conserve less-differentiated phenotypes of T cells that enhances the antitumor effect (23–25).

Although various in vitro methods for TSCM generation have been reported, most of them generate TSCM cells from naïve T cells (12, 14). We previously reported that the coculture with NOTCH ligand-expressing feeder cells converts fully activated or more differentiated memory cells into induced TSCM-like (iTSCM) cells (26, 27). These “functional” iTSCM cells expanded more efficiently in response to antigen re-stimulation than any other T-cell subsets. iTSCM cells also showed a long-lived and self-renewing potential, were resistant to cell-cycle arrest and apoptosis after TCR stimulation, and exhibited a superior antitumor ability in comparison with naïve and other memory T-cell subsets. However, why NOTCH signaling redirects fully activated cells toward TSCM-like cells remains unclear. Here, we show that NOTCH and its downstream gene, forkhead box M1 (FOXM1) facilitate mitochondria biogenesis, fatty acid synthesis and OXPHOS, resulting in iTSCM formation. CAR-iTSCM induction by FOXM1 gene overexpression enhanced antitumor effects compared with conventional CAR-T. We also propose that FOXM1-mediated metabolic fitness can provide a novel CAR-T strategy.

Cell lines

Authenticated and Mycoplasma-tested OP9 (RCB1124), NALM6 (RCB1933), and K562 (RCB0027) cells were purchased from RIKEN Bio Resource Center (Tsukuba, Ibaraki, Japan). We cloned human Delta-like 1 (hDLL1) and produced hDLL1 expression vector with lentiviral vector CS-II Human Delta-like 1–expressing-OP9 cells (OP9-hDLL1) were cultured in alpha-minimum essential medium (α-MEM; Thermo Fisher Scientific) supplemented with 20% FBS and 1% penicillin/streptomycin. Lymphoblastoid cell lines (LCL) were established from peripheral blood mononuclear cells (PBMC) provided from three healthy donors, as described previously (28). Authentication and Mycoplasma testing of LCLs were not conducted. All cell lines were frozen down at early passages (<7) and used in the experiments within 5 passages after thawing.

Human T-cell culture

Human PBMCs were prepared by specific gravity centrifugal methods from peripheral blood. Peripheral blood was provided by three healthy donors who were EBV-seropositive [VCA-IgG (+), EBNA (+)]. We prepared CD8+ T cells from PBMCs with depletion of non-CD8α+ T cells using a human CD8+ T-cell isolation kit (#130-096-495, Miltenyi Biotec). To induce EBV-specific CD8+ T cells, CD8+ T cells were labeled with carboxyfluorescein diacetate succinimidyl ester (CFSE) or Cell Trace Far Red and were subsequently cocultured with 40 Gy-irradiated autologous LCLs for 7 days. To induce anti-CD3/CD28 Abs-activated CD8+ T cells, CD8+ T cells were stimulated with Dynabeads Human T-Activator CD3/CD28. This study was approved by the Ethics Committee of Keio University (Tokyo, Japan; approved number 20120039).

Human iTSCM induction by OP9-hDLL1 cell coculture

We previously described a method for human iTSCM generation (26). In brief, to activate Notch signaling, activated T cells were cocultured with OP9-hDLL1 cells. Human T cells and OP9-hDLL1 cells were cocultured with human IL7 (PeproTech; 10 ng/mL) in α-MEM for 11 days.

Plasmid construction and retroviral transduction

Human Notch1 intracellular domain (NICD) was synthesized, and human FOXM1 gene was PCR amplified from the human cDNA library. The cDNA was subcloned into pMEI-5 vector containing the MCS-IRES-Venus or the MCS-P2A sequence. A method for generating the CAR expression vector was described previously (29). Retroviral transduction was performed, as described previously (30). Briefly, human CD8+ T cells were activated by anti-CD3/28 microbeads starting on day 0. Twenty-four hours after activation, retroviral solution was added, and the cells were centrifuged at 2,500 rpm. for 2 hours at 35°C. Eighteen hours after spin infection, the cells were transferred into 10% AB serum containing fresh RPMI1640 media to remove retrovirus.

Extracellular acidification rate, oxygen consumption rate, and ATP measurement

Measurement of the extracellular acidification rate (ECAR) and the oxygen consumption rate (OCR) was performed using an XF24 Extracellular Flux Analyzer (Seahorse Bioscience). Isolated naïve, TEM, TCM, iTSCM, and both the NICD- and FOXM1ΔN-transduced T cells were resuspended in the Seahorse XF base medium and plated (5 × 105 cells per well) into Seahorse 24-well plates coated with Cell-Tak (20 μg/mL, Corning). The ECAR and OCR were measured with a Mitostress set (Seahorse Bioscience). Oligomycin inhibits ATP synthase (complex V), which decreases electron flow through the electron transport chain (ETC), resulting in a reduction of the OCR. This decrease in OCR is linked to cellular ATP production. FCCP is an uncoupler that collapses the proton gradient and disrupts the mitochondrial membrane potential. As a result, electron flow through the ETC is uninhibited, and oxygen consumption by complex IV reaches the maximum. The third injection is a mixture of rotenone and antimycin A, a complex I inhibitor and a complex III inhibitor, respectively. The treatment completely shuts down mitochondrial respiration. Oligomycin (1.5 μmol/L), carbonyl cyanide-p-trifluoromethoxyphenylhydrazone (FCCP; 1.5 μmol/L), and the mixture of antimycin A/rotenone (1 μmol/L each) and etomoxir (200 μmol/L) were added at the indicated time points. The OCR/ECAR ratio was calculated as the basal OCR value per basal ECAR value. We measure the amount of intracellular ATP in T cells using an ATP Bioluminescence Assay Kit (Roche) following the manufacturer's instructions.

Metabolome analysis

Before capillary electrophoresis-mass spectrometry (CE-MS)-based metabolome analysis was performed, human CD8+ T cells were harvested and washed with Dulbecco PBS. These cells were washed in 10% mannitol (Wako) and then plunged into methanol that contained internal standards of methionine sulfone for cations and 2-morpholinoethanesulfonic acid for anions (300 μmol/L each). Cells were collected for CE-MS using an Agilent capillary electrophoresis system equipped with an air pressure pump, an Agilent 1100 series mass selective detector mass spectrometer, and an Agilent 1100 series isocratic high-performance liquid-chromatography pump with a G1603A adaptor kit and a G1607A sprayer kit (Agilent Technologies) as described previously (31). Values (%) were corrected for the amount of total protein.

Electroporation

The recombinant SpCas9 protein bearing a nuclear localization was prepared as described previously (32). To prepare SpCas9/sgRNA RNP complexes, 30 μg of crRNA and 30 μg of tracRNA were hybridized for 5 minutes at 95°C. The 60 μg of hybridized crRNA/tracRNA complex and 30 μg of purified recombinant SpCas9 protein were preincubated with a P2 Primary Cell 4D-NucleofectorTM X Kit S (P2 buffer; Lonza) for 15 minutes at room temperature. Human CD8+ T cells were activated by anti-CD3/28 microbeads starting on day 0. Forty-eight hours after activation, CD8+ T cells were resuspended in P2 buffer (Lonza).The cell suspensions were then mixed with P2 buffer containing Cas9 RNPs, to a final volume of 20 μL. The 20 μL of cell suspension was electroporated by a 4D-Nucleofector X unit (program code: EH100, Lonza). After electroporation, the cells were cultured in serum-free RPMI1640 medium for 3 hours and replated in RPMI1640 medium with 10% AB serum without CD3/28 microbeads. Twenty-four hours after electroporation, T cells were restimulated by anti-CD3/28 microbeads. The cells were cultured over 4 days until further experiments.

Flow cytometry and fluorescent cell imaging

Antibody dilution factors are 1:100 for antibodies purchased from BioLegend or eBioscience. To measure mitochondria mass, cells were stained with 100 nmol/L MitoTracker Green (Thermo Fisher Scientific) or MitoTracker Far Red (Thermo Fisher Scientific) for 15 minutes in RPMI1640 medium with 10% AB serum then washed twice before flow cytometry. To measure glucose uptake ability, T cells were incubated with 100 μmol/L 2-NBDG (Invitrogen) for 2 hours in RPMI1640 medium with 10% AB serum then washed twice before flow cytometry. To measure neutral lipids, cells were stained with 500 ng/mL Bodipy 493/503 (Thermo Fisher Scientific) for 10 minutes in PBS then washed twice before flow cytometry or fluorescent microscopy cell imaging. To identify fatty acid uptake ability, cells were stained with 1 μmol/L Bodipy FL C16 (Thermo Fisher Scientific) in PBS with 20 μmol/L fatty acid-free BSA then quenched by adding ice-cold PBS with 2% FBS and washed twice before flow cytometry. For PGC1A and CPT1A staining, cells were fixed and permeabilized with 4% paraformaldehyde and ice-cold 90% methanol and incubated with antibodies for 1 hour at room temperature. We performed flow cytometry acquisition on a FACS Canto II cytometer (BD Biosciences) and analyzed the data using FlowJo software (Tree Star). We sorted mouse and human T cells using a FACS Aria II cell sorter (BD Biosciences). We performed fluorescent cell imaging using a BZ-X8000 fluorescence microscope (Keyence).

Analysis of T-cell proliferation ability

A method for the generation of EBV-specific T cells was described previously (26). These EBV-specific CD8+ T cells were CFSE labeled (5 × 104), then stimulated with 1.25 × 104 autologous LCL or 5 × 104 Dynabeads Human T-Activator CD3/CD28 microbeads for 60 hours in normal media with or without 10% AB serum.

Flow-FISH

To detect comparative telomere length in T cells, we performed flow cytometry using FISH and a fluorescein-conjugated PNA probe (Dako).

Quantitative real-time PCR

Total RNA was extracted using RNAiso Plus (#Z6012, Takara Bio) or ReliaPrep RNA Miniprep Systems (#4368814, Promega) and subjected to reverse transcription using a High Capacity Complementary DNA (cDNA) Synthesis Kit (Thermo Fisher Scientific). PCR analysis was performed using an iCycler iQ multicolor Real-Time PCR Detection System (Bio-Rad) and SsoFast EvaGreen Supermix (Bio-Rad). All primer sets yielded a single product of the correct size. Relative expression levels were normalized to 18SrRNA.

Global gene analysis

The expression levels of the Affymetrix Human Gene 1.0 ST Array data were calculated from CEL files using GeneSpring GX software 14.9 (Agilent). Low or no expressed genes were removed and genes were selected by one-way ANOVA with Tukey test (P < 0.05, iTSCM vs. TCM in Fig. 1 and S3, iTSCM vs. activated in Fig. 6 and Supplementary Fig. S10), and picked over 2-fold expression in iTSCM compared with that in TCM or that in activated, respectively. Clustering analysis, principal component analysis and gene ontology analysis were performed using GeneSpring GX software 14.9. Red and blue colors indicate increased and decreased expression of genes, respectively.

Figure 1.

Enhanced OXPHOS in iTSCM cells. A, Hierarchical clustering of “Oxidative phosphorylation” genes in human-naïve T cells, TEM, TCM, and iTSCM cells. Each EBV-specific CD8+ T-cell population is defined by surface markers as follows: naïve T cells, CD8α+CFSECD45RA+CCR7+; TEM, CD8α+CFSE+CD45RACCR7; TCM, CD8α+CFSE+CD45RACCR7+; and iTSCM, CD8α+CD45RA+CCR7+ as described previously (26). B–E, OCR and ECAR measurement of naïve and EBV-specific T cells (n = 5-7). F, Flow cytometry of mitochondrial mass and PGC1A expression (n = 3). G, ATP measurement in naïve and EBV-specific T cells by luminescence assay (n = 4). H, Effects of 2-DG or rotenone on recall responses to EBV. CFSE-labeled EBV-specific T cells (5 × 104) were cocultured with autologous LCL for 60 hours with DW, DMSO, 2 mmol/L 2-DG, or 500 nmol/L rotenone. The percentages of the CFSE-diluted cells are shown in histograms (top). The line graphs show the recovered cell number of divided T cells with inhibitor compared with that without inhibitor (bottom; n = 4). *, P < 0.05; **, P < 0.01; n.s., not significant (one-way ANOVA). Data are representative of at least two independent experiments. Error bars, SEM.

Figure 1.

Enhanced OXPHOS in iTSCM cells. A, Hierarchical clustering of “Oxidative phosphorylation” genes in human-naïve T cells, TEM, TCM, and iTSCM cells. Each EBV-specific CD8+ T-cell population is defined by surface markers as follows: naïve T cells, CD8α+CFSECD45RA+CCR7+; TEM, CD8α+CFSE+CD45RACCR7; TCM, CD8α+CFSE+CD45RACCR7+; and iTSCM, CD8α+CD45RA+CCR7+ as described previously (26). B–E, OCR and ECAR measurement of naïve and EBV-specific T cells (n = 5-7). F, Flow cytometry of mitochondrial mass and PGC1A expression (n = 3). G, ATP measurement in naïve and EBV-specific T cells by luminescence assay (n = 4). H, Effects of 2-DG or rotenone on recall responses to EBV. CFSE-labeled EBV-specific T cells (5 × 104) were cocultured with autologous LCL for 60 hours with DW, DMSO, 2 mmol/L 2-DG, or 500 nmol/L rotenone. The percentages of the CFSE-diluted cells are shown in histograms (top). The line graphs show the recovered cell number of divided T cells with inhibitor compared with that without inhibitor (bottom; n = 4). *, P < 0.05; **, P < 0.01; n.s., not significant (one-way ANOVA). Data are representative of at least two independent experiments. Error bars, SEM.

Close modal

Adoptive T-cell transfer for humanized leukemic mice

All experiments using mice were approved by the Institutional Animal Care and Use Committee (IACUC; approval number 08004) of Keio University (Tokyo, Japan), and performed according to IACUC guidelines. The tumor transplantation and therapeutic model was performed according to the procedure reported by Sabatino and colleagues (14). NALM6 (1 × 106) cells were intravascularly inoculated into NOD-SCIDIl2rγnull (NSG) mice 7 days before CAR-T transfer to establish therapeutic models. CAR-T cells (5 × 105) were adoptively transferred into tumor-bearing mice. Animals were closely monitored for signs of graft-versus-host disease and other toxicity as evidenced by loss of fur, diarrhea, or conjunctivitis, as well as for leukemia-related hind-limb paralysis. When mice reached the humane endpoint in the tumor model, they were sacrificed, as approved by the Animal Ethics Committee of Keio University (Tokyo, Japan).

Study approval

This study was approved by the Institutional Review Board of Keio University School of Medicine (approval number: 20120039), and conducted in compliance with the Declaration of Helsinki. Written informed consent was obtained from all individuals.

Statistical analysis

Statistical analysis was performed using Student t test, one-way ANOVA with the Tukey–Kramer method, two-way ANOVA with Tukey–Kramer method and the Kaplan–Meier method, using GraphPad Prism version 7 software (GraphPad Software). The variance among groups was estimated using the F test, and P values <0.05 were considered statistically significant. All data are presented as the mean ± SEM. Mice were randomly assigned to experimental groups. The investigators were not blinded to allocation during experiments and outcome assessment.

Data availability

The data that support the findings of this study are available within the Supplementary Information files or from the corresponding author upon reasonable request. Microarray data that support the findings of this study have been deposited in GEO with the primary accession codes GSE23321, GSE93211, and GSE136765.

iTSCM cell proliferation is dependent on OXPHOS

We previously reported that the coculture of EBV-specific T cells with OP9-hDLL1 cells induced EBV-specific iTSCM cells and these iTSCM cells exhibited a strong in vivo antitumor effect (26, 27). Gene set enrichment analysis (GSEA) indicated that highly expressed genes in iTSCM cells were enriched in gene sets of “Myc targets,” “G2–M checkpoint,” and “E2F targets” that were supposed to be related to the higher proliferation potential of iTSCM cells, and “Oxidative Phosphorylation.” Recent studies suggested a strong correlation between OXPHOS and memory phenotypes (16, 17). Therefore, we investigated the mechanism of the enhanced antitumor activity of iTSCM cells by focusing on T-cell metabolism.

To confirm the difference between EBV-specific T cells before and after coculturing with OP9-hDLL1 cells, genes that were differentially expressed between TCM and iTSCM were extracted and the genes related to OXPHOS were significantly upregulated in iTSCM cells (Fig. 1A). Enhanced OXPHOS has been shown to be a hallmark for a strong antitumor effect of endogenous antitumor T cells or CAR-T cells (21, 22, 33). Thus, we compared the metabolic status by using a Seahorse Flux analyzer among naïve T cells and Epstein Barr virus (EBV)-specific TEM, TCM, and iTSCM cells (26, 27). iTSCM cells possessed higher basal and maximal respiration capacity compared with TEM and TCM cells (Fig. 1B). The ECAR and OCR of iTSCM cells were significantly increased compared with those of naïve T cells, TEM, and TCM cells (Fig. 1C and D). The OCR/ECAR ratio of naïve T cells and iTSCM cells was higher than that of TEM cells and TCM cells (Fig. 1E). CE-MS analysis also revealed that the total amount of glycolytic metabolites significantly increased in TCM cells, whereas TCA cycle–related metabolites did not change among TCM and iTSCM cells (Supplementary Fig. S1A–S1C). The ratio of TCA cycle/glycolysis-related metabolite was increased by the coculture with OP9-hDLL1 cells (Supplementary Fig. S1C). In addition, mitochondrial amounts, PGC1A expression, ATP content, and the NADH/NAD ratio were all increased in iTSCM cells compared with those in the other cells (Fig. 1F and G; Supplementary Fig. S1D). These data indicate that iTSCM cells preferred OXPHOS to glycolysis compared with other T-cell subsets.

We have shown that iTSCM cells exhibited stronger proliferative ability than other effector/memory T-cell subsets (26). To determine the fuel for this rapid proliferation of iTSCM cells during the recall response, we examined the proliferative ability in the presence of the glycolysis inhibitor, 2-deoxy-d-glucose (2-DG) or the mitochondrial ETC inhibitor, rotenone. Glycolysis inhibition suppressed TEM and TCM proliferation more strongly than iTSCM cells, whereas rotenone inhibited the iTSCM recall response more efficiently than TEM and TCM cells (Fig. 1H). In addition, iTSCM cells showed lower glucose uptake capacity compared with other T-cell subsets (Supplementary Fig. S1E). These results confirmed that the proliferation of iTSCM depends on OXPHOS rather than glycolysis.

FAO-dependent OXPHOS in iTSCM cells

Activated T cells prefer glycolysis, OXPHOS, and glutaminolysis for the rapid production of ATP, whereas memory T cells preferentially engage de novo fatty acid synthesis and fatty acid consumption by OXPHOS (34). Glycolysis-derived pyruvate is also consumed as the energy source of mitochondria. To validate the energy dependency of iTSCM induction, we first examined the effects of metformin, which disrupts OXPHOS and promotes glycolysis, on iTSCM induction. Expectedly, metformin interfered with iTSCM induction (Supplementary Fig. S2A). Exogenous glutamine addition did not promote iTSCM induction and did not rescue the effect of metformin (Supplementary Fig. S2B). The pyruvate-enriched condition slightly enhanced iTSCM induction, but this condition did not rescue the effect of metformin on iTSCM induction (Supplementary Fig. S2C). These data are consistent with our observation that OXPHOS rather than glycolysis plays an important role in iTSCM phenotypes and function. Furthermore, these data also suggest that iTSCM cells do not prefer to consume both pyruvate and glutamine as fuel of OXPHOS.

To identify the contribution of fatty acid metabolism to iTSCM phenotypes, we next examined fatty acid uptake, storage, and synthesis. iTSCM cells showed low fatty acid uptake ability but contained larger amounts of intracellular neutral lipid droplets (Fig. 2AC). The expression of fatty acid synthesis–related genes that regulate the entire fatty acid synthesis pathway from acetyl-CoA also increased and fatty acid transporters to mitochondria, l-carnitine, and carnitine palmitoyltransferase 1A (CPT1A) were highly expressed in iTSCM cells (Fig. 2DF; ref. 35). The OCR in the lipid-depleted state was drastically increased after loading the mitochondrial uncoupler, FCCP (Fig. 2G), indicating an extremely high mitochondrial reserve capacity in iTSCM cells. The CPT1A inhibitor, etomoxir, rapidly reduced OCR in lipid-depleted medium (Fig. 2G). Lipid depletion significantly inhibited the recall response of TEM and TCM, but the recall response of iTSCM cells did not change (Fig. 2H). Thus, these data indicate that iTSCM cells use FAO from largely stored neutral lipids for OXPHOS in response to antigen stimulation.

Figure 2.

Enhanced fatty acid synthesis in iTSCM cells. A, Fatty acid uptake by naïve and EBV-specific T cells (n = 3). B and C, Flow cytometry (B) and fluorescent staining (C) of intracellular neutral lipid in naïve and EBV-specific T cells. Neutral lipid was stained with BODIPY 493/503 (green) and DNA was visualized with Hoechst 33342 (blue). Number of intracellular lipid droplets is indicated. D, Gene expression of fatty acid synthesis–associated genes in naïve and EBV-specific T cells (n = 3). E, Quantitative measurement of l-carnitine in TCM and iTSCM cells by CE/MS-MS. F, Flow cytometry of CPT1A expression in naïve and EBV-specific T cells (n = 3). G, OCR measurement of iTSCM cells in the lipid-depleted or control medium, with or without the administration of etomoxir (n = 5 for Control + DMSO and Control + Etomoxir; n = 9 for Lipid-depleted + DMSO; n = 10 for Lipid-depleted + Etomoxir). H, Recall responses to EBV under the lipid-depleted condition. EBV-specific T cells (5 × 104) were cocultured with autologous LCL for 60 hours in control or lipid-depleted medium. The cell number of divided T cells in the lipid-depleted condition compared with that in the control is shown (n = 3). **, P < 0.01; Student t tests (E) or one-way ANOVA (A–D, F, and H). Data are representative of at least two independent experiments. Error bars, SEM.

Figure 2.

Enhanced fatty acid synthesis in iTSCM cells. A, Fatty acid uptake by naïve and EBV-specific T cells (n = 3). B and C, Flow cytometry (B) and fluorescent staining (C) of intracellular neutral lipid in naïve and EBV-specific T cells. Neutral lipid was stained with BODIPY 493/503 (green) and DNA was visualized with Hoechst 33342 (blue). Number of intracellular lipid droplets is indicated. D, Gene expression of fatty acid synthesis–associated genes in naïve and EBV-specific T cells (n = 3). E, Quantitative measurement of l-carnitine in TCM and iTSCM cells by CE/MS-MS. F, Flow cytometry of CPT1A expression in naïve and EBV-specific T cells (n = 3). G, OCR measurement of iTSCM cells in the lipid-depleted or control medium, with or without the administration of etomoxir (n = 5 for Control + DMSO and Control + Etomoxir; n = 9 for Lipid-depleted + DMSO; n = 10 for Lipid-depleted + Etomoxir). H, Recall responses to EBV under the lipid-depleted condition. EBV-specific T cells (5 × 104) were cocultured with autologous LCL for 60 hours in control or lipid-depleted medium. The cell number of divided T cells in the lipid-depleted condition compared with that in the control is shown (n = 3). **, P < 0.01; Student t tests (E) or one-way ANOVA (A–D, F, and H). Data are representative of at least two independent experiments. Error bars, SEM.

Close modal

TSCM-like CAR-T cells exhibit potent antitumor responses

To apply the method for iTSCM generation to CAR-T therapy, we next produced anti-CD19 CAR–expressing iTSCM cells by the coculturing of CAR-T cells with OP9-hDLL1 cells. As shown in Fig. 3A, to be engineered with CARs, T cells were first primed by anti-CD3/CD28 antibodies and then transduced by retrovirus carrying anti-CD19 CARs at 24 hours after activation. The CAR construct is shown in Supplementary Fig. S3A. Five days after retroviral transduction, CAR-expressing T cells were sorted out and expanded by further TCR stimulation for 6 to 8 days. These “CAR-T” cells were cocultured with OP9-hDLL1 cells for 11 days and converted into iTSCM cells, designated as “CAR-iTSCM” cells (Fig. 3A). Empty vector- and CAR-transduced T cells displayed CD45RA TCM and TEM phenotypes, whereas the coculture with OP9-hDLL1 cells efficiently converted these empty vector- and CAR-transduced T cells into CD45RA+CCR7+ TSCM phenotypes (Fig. 3B).

Figure 3.

Antitumor potential of CAR-iTSCM cells. A, Scheme of the experimental design for CAR-T and CAR-iTSCM generation. Human CD8α+ T cells were activated with anti-CD3/CD28 antibody-conjugated microbeads (anti-CD3/28 microbeads; T cells/beads ratio is 1:1) for 6 days as the “Prime” step. Retroviral transduction with anti-CD19 CAR was performed 24 hours after activation. Six days after T-cell activation, CAR-transduced T cells were sorted and cocultured with irradiated human CD19-expressing K562 cells for 6 to 8 days as the “Expansion” step. To induce CAR-iTSCM cells, the activated CAR-T cells were FACS-sorted by CD8α+CD45RACAR+ and cocultured with OP9-hDLL1 cells in the presence of hIL7 for 11 days. B, CD45RA/CCR7 expression on empty vector or CAR-transduced human CD8α+ T cells on the day before (day 0) or after (day 11) the OP9-hDLL1 coculture. Representative data of three independent experiments are shown. C, Surface marker expression on human CAR-T cells or CAR-iTSCM. The number on each panel represents mean fluorescent intensity. Gray shaded histograms represent the fluorescent minus one controls. Representative data of three independent experiments are shown. D, Early memory-associated gene profile in CAR-T or CAR-iTSCM cells (n = 3). E and F, Recall responses and activation-induced cell death by CAR stimulation (n = 3). Each T-cell population (5 × 104) was cocultured with CD19+ NALM6 cells for 60 hours. The bar graphs show the fold increase of recovered T cells (E) and the percentage of apoptotic CD8α+ cells (F). G, Schematic for CAR-T or CAR-iTSCM therapeutic strategy for human B-ALL model mice. NSG mice were intravenously inoculated with NALM6 cells. CAR-T or CAR-iTSCM cells were adoptively transferred into NSG mice 7 days after NALM6 inoculation. H, Number of peripheral NALM6 cells detected by flow cytometry on 4, 7, and 10 days after T-cell infusion (n = 5 for no transfer; n = 6 for CAR-T; n = 7 for CAR-iTSCM). Actual mean numbers per 1 mL peripheral blood on days 4, 7, and 10 were 250; 6,400; 7,300 (no transfer group), 12, 710; 2,800 (CAR-T group), and 54, 69, 90 (CAR-iTSCM group), respectively. I and J, The number of human CD3ϵ+CD8α+ cells in the spleens 4 days after T-cell infusion (n = 4 for no transfer; n = 6 for CAR-T; n = 5 for CAR-iTSCM; I). Each phenotype of naïve/TSCM, TCM, TEM, and TEMRA is determined as CD45RA+CCR7+, CD45RACCR7+, CD45RACCR7, and CD45RA+CCR7, respectively. Mean ± SEM of each subset is shown in J. K, Survival rates of NALM6-bearing mice (n = 6 for no transfer; n = 7 for CAR-T; n = 8 for CAR-iTSCM). *, P < 0.05; **, P < 0.01; n.s., not significant; Student t tests (DF), one-way ANOVA (H and I), the Kaplan–Meier method (K). Data are representative of at least two independent experiments. Error bars, SEM.

Figure 3.

Antitumor potential of CAR-iTSCM cells. A, Scheme of the experimental design for CAR-T and CAR-iTSCM generation. Human CD8α+ T cells were activated with anti-CD3/CD28 antibody-conjugated microbeads (anti-CD3/28 microbeads; T cells/beads ratio is 1:1) for 6 days as the “Prime” step. Retroviral transduction with anti-CD19 CAR was performed 24 hours after activation. Six days after T-cell activation, CAR-transduced T cells were sorted and cocultured with irradiated human CD19-expressing K562 cells for 6 to 8 days as the “Expansion” step. To induce CAR-iTSCM cells, the activated CAR-T cells were FACS-sorted by CD8α+CD45RACAR+ and cocultured with OP9-hDLL1 cells in the presence of hIL7 for 11 days. B, CD45RA/CCR7 expression on empty vector or CAR-transduced human CD8α+ T cells on the day before (day 0) or after (day 11) the OP9-hDLL1 coculture. Representative data of three independent experiments are shown. C, Surface marker expression on human CAR-T cells or CAR-iTSCM. The number on each panel represents mean fluorescent intensity. Gray shaded histograms represent the fluorescent minus one controls. Representative data of three independent experiments are shown. D, Early memory-associated gene profile in CAR-T or CAR-iTSCM cells (n = 3). E and F, Recall responses and activation-induced cell death by CAR stimulation (n = 3). Each T-cell population (5 × 104) was cocultured with CD19+ NALM6 cells for 60 hours. The bar graphs show the fold increase of recovered T cells (E) and the percentage of apoptotic CD8α+ cells (F). G, Schematic for CAR-T or CAR-iTSCM therapeutic strategy for human B-ALL model mice. NSG mice were intravenously inoculated with NALM6 cells. CAR-T or CAR-iTSCM cells were adoptively transferred into NSG mice 7 days after NALM6 inoculation. H, Number of peripheral NALM6 cells detected by flow cytometry on 4, 7, and 10 days after T-cell infusion (n = 5 for no transfer; n = 6 for CAR-T; n = 7 for CAR-iTSCM). Actual mean numbers per 1 mL peripheral blood on days 4, 7, and 10 were 250; 6,400; 7,300 (no transfer group), 12, 710; 2,800 (CAR-T group), and 54, 69, 90 (CAR-iTSCM group), respectively. I and J, The number of human CD3ϵ+CD8α+ cells in the spleens 4 days after T-cell infusion (n = 4 for no transfer; n = 6 for CAR-T; n = 5 for CAR-iTSCM; I). Each phenotype of naïve/TSCM, TCM, TEM, and TEMRA is determined as CD45RA+CCR7+, CD45RACCR7+, CD45RACCR7, and CD45RA+CCR7, respectively. Mean ± SEM of each subset is shown in J. K, Survival rates of NALM6-bearing mice (n = 6 for no transfer; n = 7 for CAR-T; n = 8 for CAR-iTSCM). *, P < 0.05; **, P < 0.01; n.s., not significant; Student t tests (DF), one-way ANOVA (H and I), the Kaplan–Meier method (K). Data are representative of at least two independent experiments. Error bars, SEM.

Close modal

Next, we examined the differences in the T-cell phenotypes between CAR-T and CAR-iTSCM cells. Expression of early memory-associated molecules CD27, CD28, and CD62L was increased in CAR-iTSCM cells but the effector markers, CD25, CD95, and PD-1 were reduced (Fig. 3C). Early memory-associated genes, EOMES, BCL6, TCF7, LEF1, KLF2, and BACH2 were also upregulated in CAR-iTSCM cells compared with those in CAR-T cells (Fig. 3D). The CAR-iTSCM phenotype potentiated recall responses to CD19-positive leukemia, and significantly suppressed activation-induced cell death (AICD; Fig. 3E and F).

To verify the antitumor effect of CAR-iTSCM cells in vivo, CAR-T or CAR-iTSCM cells were transferred into mice carrying leukemic (NALM6) cells. NALM6 cells were transferred into NSG mice, and CAR-T or CAR-iTSCM infusion was performed 7 days later (Fig. 3G). CAR-T and CAR-iTSCM infusion significantly reduced the number of NALM6 cells in the peripheral blood, whereas infused T cells were retained in both the peripheral blood and the spleen (Fig. 3H; Supplementary Fig. S3B). These data indicate an expansion of CAR-iTSCM cells in the peripheral blood on day 4, and a decrease thereafter due to the eradication of leukemic cells. It is of note that 7 to 10 days after T-cell injection, leukemia cells almost completely disappeared in mice infused with CAR-iTSCM cells, whereas remained in mice infused with CAR-T cells (Fig. 3H). Furthermore, a larger number of CD8+ T cells with the naïve/TSCM phenotype (CD45RA+CCR7+) was detected in the spleen of CAR-iTSCM-injected mice (Fig. 3I and J) and the survival of these mice was prolonged (Fig. 3K). These data indicate that CAR-iTSCM cells have much higher antitumor potential than conventional CAR-T cells.

NOTCH signaling contributes to TSCM-like induction with mitochondrial biogenesis and telomere elongation

Next, we investigated how NOTCH contributes to iTSCM formation. Both NOTCH1 and NOTCH2 but not NOTCH3 were highly expressed on human activated T cells and progressively downregulated during the OP9-hDLL1 coculture (Supplementary Fig. S4A). To define the effect of NOTCH signaling, the NICD region was expressed in activated T cells by a retrovirus vector (Supplementary Fig. S4B). Forced expression of NICD upregulated NOTCH target gene expression (Supplementary Fig. S4C). However, we failed to induce iTSCM cells only by NICD transduction due to cell death during long-term culture. It has been reported that OP9-hDLL1 culture conditioned medium (CM) with NOTCH signaling can induce mature T cells from T-cell progenitors (36). Thus, we tried to culture NICD-transduced T cells in CM of OP9-hDLL1 cells (Fig. 4A). NICD-transduction and the culture in CM supported long-term cell proliferation for 11 days, and converted activated T cells into CD45RA+CCR7+ cells at a similar efficiency as iTSCM cells induced by OP9-hDLL1 cells (Fig. 4B, CM). No additional effects of NICD transduction were not observed when NICD-transduced cells were cocultured with OP9-hDLL1 feeder cells (FC; Fig. 4B). NICD transduction also increased the mitochondrial mass and PGC1A expression (Fig. 4C) and neutral lipid storage (Fig. 4D) and upregulated the gene expression of fatty acid synthesis–related enzymes (Fig. 4E).

Figure 4.

NOTCH signaling facilitates TSCM-like cell induction. A, Scheme of the experimental design for NICD overexpression in T cells. Human CD8α+ T cells were activated with anti-CD3/28 microbeads for 6 days (Prime). Retroviral transduction with NICD was performed 24 hours after activation. Six days after T-cell activation, NICD-transduced T cells were sorted out as Venus+ cells and reactivated with anti-CD3/28 microbeads (Expansion). TCR-stimulated T cells were FACS-sorted by CD8α+CD45RA and cultured either on “OP9-hDLL1 feeder cells (FC)” or in “OP9-hDLL1 CM” in the presence of human IL7 for 11 days (TSCM-like induction). B and C, CD45RA/CCR7 expression on empty vector or NICD-transduced CD8α+ T cells on the day before or after the TSCM-like induction. B, The percentage of CD45RA+CCR7+ cells the day before or after the TSCM-like induction. C, Flow cytometry of mitochondrial mass (left) and PGC1A expression (right) of empty vector or NICD-transduced cells cultured on FC or with CM. D, Flow cytometry of or fluorescent staining of intracellular neutral lipid in empty vector or NICD-transduced cells cultured on FC or in CM. E, Gene expression of fatty acid synthesis-associated genes in empty vector or NICD-transduced cells. *, P < 0.05; **, P < 0.01; n.s., not significant; Student t tests (E), two-way ANOVA (BD). Sample size of all experiments is n = 3. Data are representative of at least two independent experiments. Error bars, SEM.

Figure 4.

NOTCH signaling facilitates TSCM-like cell induction. A, Scheme of the experimental design for NICD overexpression in T cells. Human CD8α+ T cells were activated with anti-CD3/28 microbeads for 6 days (Prime). Retroviral transduction with NICD was performed 24 hours after activation. Six days after T-cell activation, NICD-transduced T cells were sorted out as Venus+ cells and reactivated with anti-CD3/28 microbeads (Expansion). TCR-stimulated T cells were FACS-sorted by CD8α+CD45RA and cultured either on “OP9-hDLL1 feeder cells (FC)” or in “OP9-hDLL1 CM” in the presence of human IL7 for 11 days (TSCM-like induction). B and C, CD45RA/CCR7 expression on empty vector or NICD-transduced CD8α+ T cells on the day before or after the TSCM-like induction. B, The percentage of CD45RA+CCR7+ cells the day before or after the TSCM-like induction. C, Flow cytometry of mitochondrial mass (left) and PGC1A expression (right) of empty vector or NICD-transduced cells cultured on FC or with CM. D, Flow cytometry of or fluorescent staining of intracellular neutral lipid in empty vector or NICD-transduced cells cultured on FC or in CM. E, Gene expression of fatty acid synthesis-associated genes in empty vector or NICD-transduced cells. *, P < 0.05; **, P < 0.01; n.s., not significant; Student t tests (E), two-way ANOVA (BD). Sample size of all experiments is n = 3. Data are representative of at least two independent experiments. Error bars, SEM.

Close modal

TSCM cells have been shown to have a long-life due to high telomerase activity (37). Telomerase component genes were highly expressed by NICD overexpression (Supplementary Fig. S4D). Consistently, a significant increase in telomere length was observed in iTSCM cells compared with other memory T cells (Supplementary Fig. S4E). These data suggest that, in addition to intracellular neutral lipid storage, telomere elongation is a mechanism responsible for the long-life of iTSCM cells.

These data indicate that NOTCH signaling is necessary for mitochondrial biogenesis, fatty acid synthesis, and telomere elongation to acquire TSCM phenotypes.

Next, to validate the transcriptional activity of NICD, we generated various deletion mutants of NICD and examined the ability of NICD mutants for iTSCM induction. The ankyrin (ANK) domain of NOTCH is an integral part of the transcription complex, whereas the RBP-Jκ–associated molecule (RAM) domain may trigger allosteric changes in the structure needed for the derepression of transcription (38). The transactivation (TAD) domain may be important for LEF-1 activation (39). Induction of various NOTCH target genes was severely impaired by deletion of the ANK domain, but not the TAD domain, and moderately affected by the RAM domain deletion (Supplementary Fig. S5A). Consistently, TSCM induction potential was destroyed by ANK domain deletion but not so severely by TAD or RAM domain deletion (Supplementary Fig. S5B). These data clearly demonstrate that transcriptional activation by NICD is necessary for iTSCM induction.

Transcription factor FOXM1 underlies NOTCH-mediated iTSCM formation

Next, to identify a key transcription factor regulated by NOTCH signaling, we have searched iTSCM-specific genes into pathways listed in a molecular signature database. Gene set enrichment analysis revealed that several cell-cycle–regulating pathways were enriched as iTSCM-specific phenotypes (Fig. 5A). In particular, we noticed the forkhead box protein M1 (FOXM1), which is well known to regulate cell-cycle progression, G2–M checkpoint and other cell-cycle–related genes (Fig. 5A). FOXM1 has been reported to regulate not only the cell cycle but also stemness, mitochondrial function and redox network in various tumor cells (40–43). We confirmed a high expression of FOXM1 in iTSCM cells (Fig. 5B). NICD overexpression increased FOXMl expression in T cells (Fig. 5C; Supplementary Fig. S5C), and we confirmed that the deletion of RAM, ANK and TAD domain of NICD decreased FOXM1 expression, which indicates that NICD-induced transcription plays an essential role in the induction of FOXM1 (Supplementary Fig. S5D).

Figure 5.

NOTCH signaling induces TSCM phenotype via FOXM1 signaling. A, Gene enrichment analysis of iTSCM-specific genes that were highly expressed in iTSCM cells compared with TCM cells. Each bar represents an enriched pathway that was extracted as P < 0.05 by using GeneSpring and the number of selected genes in each pathway. Black bars, cell-cycle and E2F-associated pathways; red bar, the FOXM1 pathway. B, FOXM1 expression in naïve and EBV-specific T cells. C, FOXM1 expression in empty vector- or FOXM1ΔN-transduced T cells on Day 0 (left) and on Day 11 (right). D and E, CD45RA, CCR7, and CD27 expression on empty vector- or N-terminal–deleted FOXM1 (FOXM1ΔN) gene-transduced T cells the day before or after the TSCM-like induction. FOXM1ΔN-transduced activated T cells were subsequently cultured on FC or in CM as described in Fig. 4A. F, CD45RA and CCR7 expression on human CD8α+ T cells after the OP9-hDLL1 coculture with FOXM1 inhibitor, thiostrepton or DMSO. G, Effect of thiostrepton on CD45RA/CCR7 expression in empty vector-transduced T cells after the culture on FC (black) or FOXM1ΔN-transduced T cells after the culture in CM (red). **, P <0.01; n.s., not significant; Student t tests (C), one-way ANOVA (B, F, and G), two-way ANOVA (C and E). Sample size of all experiments is n = 3. Data are representative of at least two independent experiments. Error bars, SEM.

Figure 5.

NOTCH signaling induces TSCM phenotype via FOXM1 signaling. A, Gene enrichment analysis of iTSCM-specific genes that were highly expressed in iTSCM cells compared with TCM cells. Each bar represents an enriched pathway that was extracted as P < 0.05 by using GeneSpring and the number of selected genes in each pathway. Black bars, cell-cycle and E2F-associated pathways; red bar, the FOXM1 pathway. B, FOXM1 expression in naïve and EBV-specific T cells. C, FOXM1 expression in empty vector- or FOXM1ΔN-transduced T cells on Day 0 (left) and on Day 11 (right). D and E, CD45RA, CCR7, and CD27 expression on empty vector- or N-terminal–deleted FOXM1 (FOXM1ΔN) gene-transduced T cells the day before or after the TSCM-like induction. FOXM1ΔN-transduced activated T cells were subsequently cultured on FC or in CM as described in Fig. 4A. F, CD45RA and CCR7 expression on human CD8α+ T cells after the OP9-hDLL1 coculture with FOXM1 inhibitor, thiostrepton or DMSO. G, Effect of thiostrepton on CD45RA/CCR7 expression in empty vector-transduced T cells after the culture on FC (black) or FOXM1ΔN-transduced T cells after the culture in CM (red). **, P <0.01; n.s., not significant; Student t tests (C), one-way ANOVA (B, F, and G), two-way ANOVA (C and E). Sample size of all experiments is n = 3. Data are representative of at least two independent experiments. Error bars, SEM.

Close modal

To examine the roles of FOXM1 in iTSCM phenotypes, we transduced a constitutive active form of N-terminal truncated FOXM1 (FOXM1ΔN; ref. 44) in T cells (Supplementary Fig. S6A) and examined the function by using the same culture method shown in Fig. 4A. FOXM1ΔN-transduction plus OP9-hDLL1-CM conferred TSCM phenotypes to activated T cells-like coculture with OP9-hDLL1-FC (Fig. 5D and E). FOXM1-transduced cells expressed CD27 molecules at high levels (Fig. 5D and E), whereas FOXM1 inhibitor, thiostrepton treatment, and FOXM1 knockout suppressed the induction of TSCM phenotype (CD45RA+CCR7+; Fig. 5F; Supplementary Fig. S6B) and NICD-induced iTSCM formation was cancelled by thiostrepton treatment (Fig. 5G). In addition, the FOXM1 promoter contains two RBP-J–binding sites where NICD binds, and this region showed transcriptionally positive chromatin modification such as H3K27 acetylation (Supplementary Fig. S6C). Importance of the RBP-J–binding sites of the FOXM1 promoter is also supported by the requirement of the RAM (RBP-J–associated module) domain for induction of FOXM1 expression (see Supplementary Fig. S5D). These results suggest that FOXM1 is a direct downstream gene of Notch signaling and positively regulates the induction of TSCM-like phenotypes.

Antitumor potential of FOXM1ΔN-overexpressed TSCM-like cells

We confirmed the enhanced antitumor potentials of TSCM-like cells induced by FOXM1ΔN overexpression using the CAR-T system. Normal CAR or FOXM1ΔN tandemly jointed CAR (FOXM1ΔN-CAR) constructs were retrovirally transduced (Supplementary Fig. S7A) and these cells were cultured on OP9-hDLL1-FC or with OP9-hDLL1-CM (Fig. 6A). These CAR-transduced T cells were transferred to NALM6-bearing NSG mice (Fig. 6B). Twelve days after CAR-T transfer, few NALM6 cells were detected in CAR-iTSCM- and CAR-TSCM-like (FOXM1ΔN)-transferred mice, whereas leukemic cells remained in the other CAR-T–transferred mice (Fig. 6C). Both CAR-iTSCM- and CAR-TSCM-like (FOXM1ΔN) cells prolonged the survival of NALM6-bearing mice compared with the other groups (Fig. 6D). These data indicate that TSCM-like cells induced by FOXM1ΔN and CM have similar antitumor potential to iTSCM cells induced by coculture with OP9-hDLL1-FC, suggesting that FOXM1 is a key factor for generating TSCM-like CAR-T cells.

Figure 6.

FOXM1 overexpression enhances antitumor activity and iTSCM formation by metabolic reprogramming. A, Induction of CAR-iTSCM-like cells by gene transduction of FOXM1ΔN. CAR-iTSCM cells were induced as shown in Fig. 1A. CAR-iTSCM–like cells were generated by transducing activated T cells with CAR-FOXM1ΔN retrovirus. The fraction of CD45RA+CCR7+ cells was determined by flow cytometry. B, Scheme of the experimental design for CAR-iTSCM therapy for NSG mice carrying NALM6 cells. C, Number of peripheral NALM6 cells on 12 days after T-cell infusion [n = 5 for no transfer; n = 8 for CAR-T (Empty); n = 4 for CAR-iTSCM (Empty); n = 10 for CAR-T (FOXM1ΔN); n = 3 for CAR-TSCM-like (FOXM1ΔN)]. NALM6 cells in peripheral blood were detected as hCD19+ cells by flow cytometry. D, Survival rates of NALM6-bearing mice [n = 8 for no transfer; n = 8 for CAR-T (Empty); n = 6 for CAR-iTSCM (Empty); n = 10 for CAR-T (FOXM1ΔN); n = 5 for CAR-iTSCM (FOXM1ΔN)]. E, Flow cytometry of mitochondrial mass and PGC1A expression of the T cells after the OP9-hDLL1 coculture with thiostrepton or DMSO (n = 3). F, OCR and ECAR measurement of the T cells after the OP9-hDLL1 coculture with thiostrepton or DMSO (n = 9 for DMSO; n = 5 for thiostrepton). G, Flow cytometry of the mitochondrial mass of scramble gRNA–transduced T cells (negative control) or FOXM1 knockout T cells cultured on FC (n = 3). H, Flow cytometry of CD45RA/CCR7 expression on empty vector–transduced T cells after the culture on FC, or NICD- or FOXM1ΔN-transduced T cells after the culture with CM with rotenone or DMSO (n = 3). I, PC analysis of activated, NICD-induced TSCM-like, FOXM1ΔN-induced TSCM-like, iTSCM, and naïve T cells from GSE23321 and GSE136765 using the 130 genes highly expressed in all datasets (n = 3). *, P < 0.05; **, P < 0.01; n.s., not significant; Student t tests (E–G), one-way ANOVA (H), the Kaplan–Meier method (D). Data are representative of at least two independent experiments. Error bars, SEM.

Figure 6.

FOXM1 overexpression enhances antitumor activity and iTSCM formation by metabolic reprogramming. A, Induction of CAR-iTSCM-like cells by gene transduction of FOXM1ΔN. CAR-iTSCM cells were induced as shown in Fig. 1A. CAR-iTSCM–like cells were generated by transducing activated T cells with CAR-FOXM1ΔN retrovirus. The fraction of CD45RA+CCR7+ cells was determined by flow cytometry. B, Scheme of the experimental design for CAR-iTSCM therapy for NSG mice carrying NALM6 cells. C, Number of peripheral NALM6 cells on 12 days after T-cell infusion [n = 5 for no transfer; n = 8 for CAR-T (Empty); n = 4 for CAR-iTSCM (Empty); n = 10 for CAR-T (FOXM1ΔN); n = 3 for CAR-TSCM-like (FOXM1ΔN)]. NALM6 cells in peripheral blood were detected as hCD19+ cells by flow cytometry. D, Survival rates of NALM6-bearing mice [n = 8 for no transfer; n = 8 for CAR-T (Empty); n = 6 for CAR-iTSCM (Empty); n = 10 for CAR-T (FOXM1ΔN); n = 5 for CAR-iTSCM (FOXM1ΔN)]. E, Flow cytometry of mitochondrial mass and PGC1A expression of the T cells after the OP9-hDLL1 coculture with thiostrepton or DMSO (n = 3). F, OCR and ECAR measurement of the T cells after the OP9-hDLL1 coculture with thiostrepton or DMSO (n = 9 for DMSO; n = 5 for thiostrepton). G, Flow cytometry of the mitochondrial mass of scramble gRNA–transduced T cells (negative control) or FOXM1 knockout T cells cultured on FC (n = 3). H, Flow cytometry of CD45RA/CCR7 expression on empty vector–transduced T cells after the culture on FC, or NICD- or FOXM1ΔN-transduced T cells after the culture with CM with rotenone or DMSO (n = 3). I, PC analysis of activated, NICD-induced TSCM-like, FOXM1ΔN-induced TSCM-like, iTSCM, and naïve T cells from GSE23321 and GSE136765 using the 130 genes highly expressed in all datasets (n = 3). *, P < 0.05; **, P < 0.01; n.s., not significant; Student t tests (E–G), one-way ANOVA (H), the Kaplan–Meier method (D). Data are representative of at least two independent experiments. Error bars, SEM.

Close modal

FOXM1 induces TSCM-like formation through mitochondrial biogenesis

Next, we investigated the role of FOXM1 in the mitochondrial changes of iTSCM cells. Treatment with a FOXM1 inhibitor, thiostrepton, during coculture on OP9-hDLL1-FC decreased mitochondrial mass, PGC1A expression, and OCR, but increased ECAR (Fig. 6E and F). FOXM1 deficiency also decreased mitochondrial mass (Fig. 6G), indicating that FOXM1 is required for mitochondrial changes. Overexpression of FOXM1ΔN with OP9-hDLL1-CM increased mitochondrial mass and PGC1A expression to similar levels in iTSCM cells induced by OP9-hDLL1-FC (Supplementary Fig. S7B). Furthermore, the increase in mitochondrial biogenesis induced by overexpression of NICD was cancelled by FOXM1 inhibition (Supplementary Fig. S7C). ETC inhibitor, rotenone, also partly inhibited iTSCM formation either by OP9-hDLL1 coculture or by NICD or FOXM1ΔN overexpression (Fig. 6H). In contrast, CPT1A inhibition had little effect on FOXM1-driven iTSCM induction (Supplementary Fig. S7D). Thus, although NOTCH and FOXMl tightly regulate both the mitochondrial biogenesis and iTSCM phenotypes, CPT1A and FAO themselves may not be involved in iTSCM surface phenotypes but are important for high proliferation ability of iTSCM cells in glucose-low conditions.

NICD- and FOXM1-transduced TSCM-like cells are close to iTSCM cells induced by OP9-hDLL1 cells

To further confirm the relationship among activated T cells, gene expression profiles were analyzed among naïve T cells, iTSCM cells induced by OP9-DLL1-FC, and iTSCM-like cells induced by NICD or FOXM1. Principal component (PC) analysis and clustering analysis revealed that NICD- and FOXM1ΔN-induced TSCM-like cells were most closely related to iTSCM cells compared with naïve T cells and activated T cells (Fig. 6I; Supplementary Fig. S8A). OXPHOS and glycolysis profiles of both NICD- and FOXM1ΔN-induced TSCM-like cells highly resembled iTSCM cells (Supplementary S8B and S8C). These data confirmed that both NOTCH and FOXM1 promote the transcriptional program that induce iTSCM cells.

We have reported that the NOTCH signaling induces TSCM-like, “iTSCM” cells from fully activated T cells. Unlike other TSCM-generating methods that induce TSCM-like cells from naïve T cells, our two-step culture is more suitable for generating CAR-iTSCM cells, because T-cell activation is necessary for the transduction of the CAR gene. We also found that the gene sets of OXPHOS were highly enriched in iTSCM cells. Therefore, we focused on OXPHOS because Myc targets, G2–M checkpoint and E2F targets appear to be related to a higher proliferation capacity of iTSCM cells. Here, we demonstrated that the coculture with NOTCH ligand-expressing feeder cells, or the forced expression of NICD or FOXM1ΔN conferred TSCM-like properties, including OXPHOS preference, and potentiate the antitumor ability of CAR-T cells. Our data also indicated that mitochondrial biogenesis and fatty acid synthesis are important for iTSCM induction and function.

The potent antitumor activity of iTSCM cells appears to be associated with not only early memory phenotypes but also FAO-dependent OXPHOS of iTSCM cells. Fraietta and colleagues reported that CAR-T cells from complete-responding patients with CLL were enriched in early memory–related genes with reduced glycolysis signatures (7). TCR stimulation with Wnt signaling from naïve T cells sustains the TSCM property and leads to strong antitumor response in CAR-T therapy (13). Less-differentiated CAR-T induction was obtained by inhibiting the Akt–mTOR pathway, enhancing FOXO1 activity and potentiating mitochondrial function (23–25, 45–47). In addition, IL21 in combination with rapamycin has been reported to enhance generation of tumor antigen-specific TSCM-like cells (48), and IL21 enhances TCM formation in a FAO-dependent manner (49). Conversely, chronic CD4+ T cells are irreversibly dependent on OXPHOS and FAO to produce ATP for IFNγ synthesis, but PD-1 blockade unlocks the irreversible metabolism and reprograms to functional T cells (50). These reports suggest that the metabolic fitness regulates T-cell differentiation, memory formation, and exhaustion, and therefore maintains the antitumor ability of T cells.

Here, we identified FOXM1 as a key regulator for iTSCM induction as a downstream target of NOTCH signaling. FOXM1 overexpression can mostly substitute for the effects of NOTCH signaling on CAR-iTSCM properties, including OXPHOS preference, telomere elongation, and potent antitumor ability. FOXM1 has been characterized as an important regulating factor for cancer stemness, chemo-resistance, mitochondrial function, and redox pathways in tumor cells (40–43). We propose that FOXM1 is also an important factor for the generation of iTSCM cells. Therefore, the induction of FOXM1 is a potent method for conferring TSCM-like properties to CAR-T cells. In this sense, it is of interest that FOXM1 is also upregulated in TSCM-like cells induced by the Wnt signaling. Our data suggest that NOTCH1 and FOXM1 are sufficient for the induction of TSCM-cell surface phenotypes and the induction of OXPHOS-dominant ATP synthesis; however, FAO-dependent OXPHOS is not sufficient for induction of the entire iTSCM program. The precise molecular mechanism of FAO and OXPHOS-dependent ATP synthesis in iTSCM cells remains to be clarified.

W. Tomisato is an employee at Daiichi Sankyo Co., Ltd. No potential conflicts of interest were disclosed by the other authors.

Conception and design: T. Kondo, W. Tomisato, A. Yoshimura

Development of methodology: T. Kondo

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): T. Kondo, M. Ando, T. Srirat, S. Mise-Omata, T. Hishiki

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): T. Kondo, M. Ando, N. Nagai, T. Srirat, M. Ohmura

Writing, review, and/or revision of the manuscript: T. Kondo, B. Liu, S. Chikuma

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): T. Kondo, M. Ando, M. Ikeda, H. Nishimasu, O. Nureki, N. Hayakawa, R. Uchibori, K. Ozawa

Study supervision: T. Kondo, A. Yoshimura

We thank Yoshiko Noguchi, Yasuko Hirata, Yukiko Tokifuji, Mika Inoue, Eiji Sugihara, Hideyuki Saya (Keio University) for their technical assistance and Taeka Hayakawa and Yutaka Kawakami (Keio University) for discussions. This work was supported by JSPS KAKENHI (S) JP17H06175, Challenging Research (P) JP18H05376, and AMED-CREST JP19gm1110009, TaNeDS grant program (Daiichi Sankyo Co., Ltd.), the Takeda Science Foundation, the Uehara Memorial Foundation, the Mochida Memorial Foundation for Medical and Pharmaceutical Research, Bristol–Myers Squibb Research grant, the Kanae Foundation, the SENSHIN Medical Research Foundation and the Keio Gijuku Academic Developmental Funds.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
June
CH
,
O'Connor
RS
,
Kawalekar
OU
,
Ghassemi
S
,
Milone
MC
. 
CAR T-cell immunotherapy for human cancer
.
Science
2018
;
359
:
1361
5
.
2.
Dunbar
CE
,
High
KA
,
Joung
JK
,
Kohn
DB
,
Ozawa
K
,
Sadelain
M
. 
Gene therapy comes of age
.
Science
2018
;
359
.
DOI: 10.1126/science.aan4672.
3.
June
CH
,
Sadelain
M
. 
Chimeric antigen receptor therapy
.
N Engl J Med
2018
;
379
:
64
73
.
4.
Maude
SL
,
Laetsch
TW
,
Buechner
J
,
Rives
S
,
Boyer
M
,
Bittencourt
H
, et al
Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia
.
N Engl J Med
2018
;
378
:
439
48
.
5.
Levine
BL
,
Miskin
J
,
Wonnacott
K
,
Keir
C
. 
Global manufacturing of CAR T cell therapy
.
Mol Ther Methods Clin Dev
2017
;
4
:
92
101
.
6.
Louis
CU
,
Savoldo
B
,
Dotti
G
,
Pule
M
,
Yvon
E
,
Myers
GD
, et al
Antitumor activity and long-term fate of chimeric antigen receptor-positive T cells in patients with neuroblastoma
.
Blood
2011
;
118
:
6050
6
.
7.
Fraietta
JA
,
Lacey
SF
,
Orlando
EJ
,
Pruteanu-Malinici
I
,
Gohil
M
,
Lundh
S
, et al
Determinants of response and resistance to CD19 chimeric antigen receptor (CAR) T-cell therapy of chronic lymphocytic leukemia
.
Nat Med
2018
;
24
:
563
71
.
8.
Xu
Y
,
Zhang
M
,
Ramos
CA
,
Durett
A
,
Liu
E
,
Dakhova
O
, et al
Closely related T-memory stem cells correlate with in vivo expansion of CAR.CD19-T cells and are preserved by IL-7 and IL-15
.
Blood
2014
;
123
:
3750
9
.
9.
Kagoya
Y
,
Nakatsugawa
M
,
Ochi
T
,
Cen
Y
,
Guo
T
,
Anczurowski
M
, et al
Transient stimulation expands superior antitumor T cells for adoptive therapy
.
JCI Insight
2017
;
2
:
e89580
.
10.
Kagoya
Y
,
Nakatsugawa
M
,
Yamashita
Y
,
Ochi
T
,
Guo
T
,
Anczurowski
M
, et al
BET bromodomain inhibition enhances T-cell persistence and function in adoptive immunotherapy models
.
J Clin Invest
2016
;
126
:
3479
94
.
11.
Kagoya
Y
,
Tanaka
S
,
Guo
T
,
Anczurowski
M
,
Wang
CH
,
Saso
K
, et al
A novel chimeric antigen receptor containing a JAK-STAT signaling domain mediates superior antitumor effects
.
Nat Med
2018
;
24
:
352
9
.
12.
Alvarez-Fernandez
C
,
Escriba-Garcia
L
,
Vidal
S
,
Sierra
J
,
Briones
J
. 
A short CD3/CD28 costimulation combined with IL-21 enhance the generation of human memory stem T cells for adoptive immunotherapy
.
J Transl Med
2016
;
14
:
214
.
13.
Gattinoni
L
,
Lugli
E
,
Ji
Y
,
Pos
Z
,
Paulos
CM
,
Quigley
MF
, et al
A human memory T-cell subset with stem cell-like properties
.
Nat Med
2011
;
17
:
1290
7
.
14.
Sabatino
M
,
Hu
J
,
Sommariva
M
,
Gautam
S
,
Fellowes
V
,
Hocker
JD
, et al
Generation of clinical-grade CD19-specific CAR-modified CD8+ memory stem cells for the treatment of human B-cell malignancies
.
Blood
2016
;
128
:
519
28
.
15.
Gattinoni
L
,
Speiser
DE
,
Lichterfeld
M
,
Bonini
C
. 
T memory stem cells in health and disease
.
Nat Med
2017
;
23
:
18
27
.
16.
O'Sullivan
D
,
van der Windt
GJ
,
Huang
SC
,
Curtis
JD
,
Chang
CH
,
Buck
MD
, et al
Memory CD8(+) T cells use cell-intrinsic lipolysis to support the metabolic programming necessary for development
.
Immunity
2014
;
41
:
75
88
.
17.
Cui
G
,
Staron
MM
,
Gray
SM
,
Ho
PC
,
Amezquita
RA
,
Wu
J
, et al
IL-7-induced glycerol transport and TAG synthesis promotes memory CD8+ T-cell longevity
.
Cell
2015
;
161
:
750
61
.
18.
Ho
PC
,
Kaech
SM
. 
Reenergizing T-cell anti-tumor immunity by harnessing immunometabolic checkpoints and machineries
.
Curr Opin Immunol
2017
;
46
:
38
44
.
19.
Kishton
RJ
,
Sukumar
M
,
Restifo
NP
. 
Metabolic regulation of T-cell longevity and function in tumor immunotherapy
.
Cell Metab
2017
;
26
:
94
109
.
20.
Chang
CH
,
Pearce
EL
. 
Emerging concepts of T-cell metabolism as a target of immunotherapy
.
Nat Immunol
2016
;
17
:
364
8
.
21.
Scharping
NE
,
Menk
AV
,
Moreci
RS
,
Whetstone
RD
,
Dadey
RE
,
Watkins
SC
, et al
The tumor microenvironment represses T-cell mitochondrial biogenesis to drive intratumoral T-cell metabolic insufficiency and dysfunction
.
Immunity
2016
;
45
:
701
3
.
22.
Klein Geltink
RI
,
O'Sullivan
D
,
Corrado
M
,
Bremser
A
,
Buck
MD
,
Buescher
JM
, et al
Mitochondrial priming by CD28
.
Cell
2017
;
171
:
385
97
.
23.
Klebanoff
CA
,
Crompton
JG
,
Leonardi
AJ
,
Yamamoto
TN
,
Chandran
SS
,
Eil
RL
, et al
Inhibition of AKT signaling uncouples T-cell differentiation from expansion for receptor-engineered adoptive immunotherapy
.
JCI Insight
2017
;
2
.
DOI: 10.1172/jci.insight.95103
.
24.
Pilipow
K
,
Scamardella
E
,
Puccio
S
,
Gautam
S
,
De Paoli
F
,
Mazza
EM
, et al
Antioxidant metabolism regulates CD8+ T memory stem cell formation and antitumor immunity
.
JCI Insight
2018
;
3
.
DOI: 10.1172/jci.insight.122299
.
25.
Mousset
CM
,
Hobo
W
,
Ji
Y
,
Fredrix
H
,
De Giorgi
V
,
Allison
RD
, et al
Ex vivo AKT-inhibition facilitates generation of polyfunctional stem cell memory-like CD8(+) T cells for adoptive immunotherapy
.
Oncoimmunology
2018
;
7
:
e1488565
.
26.
Kondo
T
,
Imura
Y
,
Chikuma
S
,
Hibino
S
,
Omata-Mise
S
,
Ando
M
, et al
Generation and application of human induced-stem cell memory T cells for adoptive immunotherapy
.
Cancer Sci
2018
;
109
:
2130
40
.
27.
Kondo
T
,
Morita
R
,
Okuzono
Y
,
Nakatsukasa
H
,
Sekiya
T
,
Chikuma
S
, et al
Notch-mediated conversion of activated T cells into stem cell memory-like T cells for adoptive immunotherapy
.
Nat Commun
2017
;
8
:
15338
.
28.
Hui-Yuen
J
,
McAllister
S
,
Koganti
S
,
Hill
E
,
Bhaduri-McIntosh
S
. 
Establishment of Epstein-Barr virus growth-transformed lymphoblastoid cell lines
.
J Vis Exp
2011
;(57):
3321
.
DOI: 10.3791/3321
.
29.
Tsukahara
T
,
Ohmine
K
,
Yamamoto
C
,
Uchibori
R
,
Ido
H
,
Teruya
T
, et al
CD19 target-engineered T-cells accumulate at tumor lesions in human B-cell lymphoma xenograft mouse models
.
Biochem Biophys Res Commun
2013
;
438
:
84
9
.
30.
Sekiya
T
,
Kashiwagi
I
,
Inoue
N
,
Morita
R
,
Hori
S
,
Waldmann
H
, et al
The nuclear orphan receptor Nr4a2 induces Foxp3 and regulates differentiation of CD4+ T cells
.
Nat Commun
2011
;
2
:
269
.
31.
Yamamoto
T
,
Takano
N
,
Ishiwata
K
,
Ohmura
M
,
Nagahata
Y
,
Matsuura
T
, et al
Reduced methylation of PFKFB3 in cancer cells shunts glucose towards the pentose phosphate pathway
.
Nat Commun
2014
;
5
:
3480
.
32.
Nishimasu
H
,
Ran
FA
,
Hsu
PD
,
Konermann
S
,
Shehata
SI
,
Dohmae
N
, et al
Crystal structure of Cas9 in complex with guide RNA and target DNA
.
Cell
2014
;
156
:
935
49
.
33.
Kawalekar
OU
,
O'Connor
RS
,
Fraietta
JA
,
Guo
L
,
McGettigan
SE
,
Posey
AD
 Jr
, et al
Distinct signaling of coreceptors regulates specific metabolism pathways and impacts memory development in CAR T Cells
.
Immunity
2016
;
44
:
380
90
.
34.
Zhang
L
,
Romero
P
. 
Metabolic control of CD8(+) T-cell fate decisions and antitumor immunity
.
Trends Mol Med
2018
;
24
:
30
48
.
35.
Qian
X
,
Yang
Z
,
Mao
E
,
Chen
E
. 
Regulation of fatty acid synthesis in immune cells
.
Scand J Immunol
2018
;
88
:
e12713
.
36.
Fernandez
I
,
Ooi
TP
,
Roy
K
. 
Generation of functional, antigen-specific CD8+ human T cells from cord blood stem cells using exogenous Notch and tetramer-TCR signaling
.
Stem Cells
2014
;
32
:
93
104
.
37.
Ahmed
R
,
Roger
L
,
Costa Del Amo
P
,
Miners
KL
,
Jones
RE
,
Boelen
L
, et al
Human stem cell-like memory T cells are maintained in a state of dynamic flux
.
Cell Rep
2016
;
17
:
2811
8
.
38.
Barrick
D
,
Kopan
R
. 
The Notch transcription activation complex makes its move
.
Cell
2006
;
124
:
883
5
.
39.
Ross
DA
,
Kadesch
T
. 
The notch intracellular domain can function as a coactivator for LEF-1
.
Mol Cell Biol
2001
;
21
:
7537
44
.
40.
Chiu
WT
,
Huang
YF
,
Tsai
HY
,
Chen
CC
,
Chang
CH
,
Huang
SC
, et al
FOXM1 confers to epithelial–mesenchymal transition, stemness and chemoresistance in epithelial ovarian carcinoma cells
.
Oncotarget
2015
;
6
:
2349
65
.
41.
Lee
Y
,
Kim
KH
,
Kim
DG
,
Cho
HJ
,
Kim
Y
,
Rheey
J
, et al
FoxM1 promotes stemness and radio-resistance of glioblastoma by regulating the master stem cell regulator sox2
.
PLoS ONE
2015
;
10
:
e0137703
.
42.
Song
IS
,
Jeong
YJ
,
Han
J
. 
Mitochondrial metabolism in cancer stem cells: a therapeutic target for colon cancer
.
BMB Rep
2015
;
48
:
539
40
.
43.
Song
IS
,
Jeong
YJ
,
Jeong
SH
,
Heo
HJ
,
Kim
HK
,
Bae
KB
, et al
FOXM1-induced PRX3 regulates stemness and survival of colon cancer cells via maintenance of mitochondrial function
.
Gastroenterology
2015
;
149
:
1006
16
.
44.
Laoukili
J
,
Alvarez
M
,
Meijer
LA
,
Stahl
M
,
Mohammed
S
,
Kleij
L
, et al
Activation of FoxM1 during G2 requires cyclin A/Cdk-dependent relief of autorepression by the FoxM1 N-terminal domain
.
Mol Cell Biol
2008
;
28
:
3076
87
.
45.
Sukumar
M
,
Liu
J
,
Ji
Y
,
Subramanian
M
,
Crompton
JG
,
Yu
Z
, et al
Inhibiting glycolytic metabolism enhances CD8+ T-cell memory and antitumor function
.
J Clin Invest
2013
;
123
:
4479
88
.
46.
Crompton
JG
,
Sukumar
M
,
Roychoudhuri
R
,
Clever
D
,
Gros
A
,
Eil
RL
, et al
Akt inhibition enhances expansion of potent tumor-specific lymphocytes with memory cell characteristics
.
Cancer Res
2015
;
75
:
296
305
.
47.
Scholz
G
,
Jandus
C
,
Zhang
L
,
Grandclement
C
,
Lopez-Mejia
IC
,
Soneson
C
, et al
Modulation of mTOR signalling triggers the formation of stem cell-like memory T cells
.
EBioMedicine
2016
;
4
:
50
61
.
48.
Jeza
VT
,
Li
X
,
Chen
J
,
Liang
Z
,
Aggrey
AO
,
Wu
X
. 
IL-21 augments rapamycin in expansion of alpha fetoprotein antigen specific stem-cell-like memory T cells in vitro
.
The Pan African medical journal
2017
;
27
:
163
.
49.
Loschinski
R
,
Bottcher
M
,
Stoll
A
,
Bruns
H
,
Mackensen
A
,
Mougiakakos
D
. 
IL-21 modulates memory and exhaustion phenotype of T-cells in a fatty acid oxidation-dependent manner
.
Oncotarget
2018
;
9
:
13125
38
.
50.
Bettonville
M
,
d'Aria
S
,
Weatherly
K
,
Porporato
PE
,
Zhang
J
,
Bousbata
S
, et al
Long-term antigen exposure irreversibly modifies metabolic requirements for T-cell function
.
Elife
2018
;
7
.
DOI: 10.7554/eLife.30938
.