The use of dendritic cells (DC) to prime tumor-associated antigen-specific T-cell responses provides a promising approach to cancer immunotherapy. Embryonic stem cells (ESC) and induced pluripotent stem cells (iPSC) can differentiate into functional DCs, thus providing an unlimited source of DCs. However, the previously established methods of generating practical volumes of DCs from pluripotent stem cells (PSC) require a large number of PSCs at the start of the differentiation culture. In this study, we generated mouse proliferating myeloid cells (pMC) as a source of antigen-presenting cells (APC) using lentivirus-mediated transduction of the c-Myc gene into mouse PSC-derived myeloid cells. The pMCs could propagate almost indefinitely in a cytokine-dependent manner, while retaining their potential to differentiate into functional APCs. After treatment with IL4 plus GM-CSF, the pMCs showed impaired proliferation and differentiated into immature DC-like cells (pMC-DC) expressing low levels of major histocompatibility complex (MHC)-I, MHC-II, CD40, CD80, and CD86. In addition, exposure to maturation stimuli induced the production of TNFα and IL12p70, and enhanced the expression of MHC-II, CD40, and CD86, which is thus suggestive of typical DC maturation. Similar to bone marrow–derived DCs, they stimulated a primary mixed lymphocyte reaction. Furthermore, the in vivo transfer of pMC-DCs pulsed with H-2Kb-restricted OVA257-264 peptide primed OVA-specific cytotoxic T cells and elicited protection in mice against challenge with OVA-expressing melanoma. Overall, myeloid cells exhibiting cytokine-dependent proliferation and DC-like differentiation may be used to address issues associated with the preparation of DCs. Cancer Immunol Res; 3(6); 668–77. ©2015 AACR.

Dendritic cells (DC) are commonly used as biologic therapy for cancer because of their physiologic roles in initiating and modulating the host immune response (1, 2). In many cases, monocytes obtained from patients by apheresis have been used as a source of DCs (3, 4). However, the number of monocytes obtained from peripheral blood, their ability to differentiate into DCs, and the quality of the resulting DCs vary among patients. Therefore, it can be difficult to stably generate a sufficient number of high-quality DCs for use in cancer therapy. In addition, the requirement to prepare DCs separately for each patient prevents the broader application of this strategy.

Pluripotent stem cells (PSC), such as embryonic stem cells (ESC) and the recently developed induced PSCs (iPSC), have the potential to propagate indefinitely and can differentiate into various somatic cell types (5–7). Previous studies have established methods of generating DCs from PSCs, and demonstrated the utility of PSC-derived DCs in cancer immunotherapy (8–25). However, generating a large number of DCs from ESCs or iPSCs has required a scaling-up of the initial volume of undifferentiated PSCs. In addition, these methods are too laborious for practical application in the clinical setting.

We recently developed a simple and efficient method for obtaining a large number of functional antigen-presenting cells (APC) from human iPSCs (26, 27). By introducing the c-Myc gene, along with antisenescence factors, such as Bmi-1 or Ezh2, the myeloid cells generated from iPSCs could proliferate almost indefinitely while retaining the capacity to differentiate into potent APCs. In addition, cells derived from transporter associated with antigen processing 2 (TAP2)–deficient iPSCs downregulated the expression of endogenous human leukocyte antigen (HLA)-I and avoided recognition by alloreactive CD8+ T cells, and cells expressing exogenously introduced HLA-I genes stimulated tumor-associated antigen-specific CD8+ T cells in vitro (26). Genetic modification of macrophages derived from the proliferating myeloid cells (pMC) to express an anti-HER2 antibody or interferon (IFN) could also exert therapeutic effects against peritoneally disseminated gastric and pancreatic cancer in xenograft models (27). However, their in vivo therapeutic potential against cancer and in vivo safety have not been examined in syngeneic mouse models. In addition, whether pMCs can be generated from mouse PSCs remains unclear.

In this study, we generated pMCs from mouse ESCs and iPSCs, and showed their potential to differentiate into functional APCs. This new method enabled us to obtain a large number of APCs from a small number of undifferentiated PSCs. Moreover, once the pMCs were established, it was possible to supply APCs in a few days using the pMCs as a cell source. Using a model antigen, we demonstrated that vaccination with APCs was safe and feasible for cancer immunotherapy, without any induction of autoimmunity or leukemia in vivo.

Mice

C57BL/6 and BALB/c mice were purchased from Japan SLC, Inc. or CLEA Japan, Inc., and were maintained under specific pathogen-free conditions. All animal experiments were performed with approval from the Animal Experiment Committee of the Aichi Cancer Center.

Cells

The mouse ESC line, B6-ES, derived from C57BL/6 blastocysts (28), and the mouse iPSC lines, 2A-4F-100 and 2A-4F-136 (29), derived from C57BL/6 embryonic fibroblasts, were maintained as described previously (28, 29). The M-CSF–defective bone marrow–derived stromal cell line, OP9 (30), was maintained in α-minimum essential medium (α-MEM; Life Technologies) supplemented with 20% fetal bovine serum (FBS), and the cells were seeded onto gelatin-coated dishes before being used as feeder cells. MO4 (31), a C57BL/6-derived B16 melanoma cell line expressing OVA, luciferase-transduced MO4 (MO4-Luc), and the EL-4 thymoma cell line, were maintained in RPMI-1640 medium (Sigma-Aldrich) supplemented with 10% FBS.

Generation of recombinant lentivirus

A lentivirus vector, CSII-EF, and the plasmids used for lentiviral vector packaging, pCMV-VSV-G-RSV-Rev and pCAG-HIVgp, were kindly provided by Dr. H. Miyoshi (RIKEN BioResource Center; Tsukuba, Japan). CSII-EF containing a human c-Myc cDNA insert was used as described previously (32). Plasmid constructs were introduced into 293T cells using lipofection (Lipofectamine 2000; Life Technologies). Three days later, recombinant lentiviruses were recovered from the culture supernatant using a Lenti-X Concentrator (Clontech).

Generation of pMCs from mouse ESCs or iPSCs

ESCs or iPSCs were induced to differentiate into myeloid cells according to an established procedure (Fig. 1; refs. 9, 11–14, 18, 19). Briefly, the ESCs or iPSCs were cultured on feeder layers of OP9 cells for 6 to 7 days in α-MEM supplemented with 20% FBS. The mesodermally differentiated cells were then harvested, reseeded onto fresh OP9 cell layers, and cultured in α-MEM supplemented with 20% FBS, 20 ng/mL GM-CSF, and 50 μmol/L 2-ME. On day 13 to 14, floating cells were recovered by pipetting. These cells were considered to be ESC- or iPSC-derived myeloid cells (ES-MCs or iPS-MCs, respectively). The cells were infected with lentivirus vectors expressing the c-Myc gene in the presence of 8 ng/mL polybrene (Sigma-Aldrich), and were cultured in α-MEM supplemented with 20% FBS, 30 ng/mL GM-CSF, and 30 ng/mL M-CSF. After 5 to 6 days, proliferating cells appeared and were considered to be ESC- or iPSC-derived pMCs (ES-pMC or iPS-pMC, respectively). To induce the differentiation of these cells into DC-like cells (pMC-DC), they were cultured in RPMI-1640 supplemented with 20% FBS in the presence of 20 ng/mL IL4 plus 30 ng/mL GM-CSF for 3 days.

Figure 1.

Schematic illustration showing the generation of pMCs from mouse ESCs and iPSCs. A, the protocol for the generation of pMCs. The culture medium, supplements in the medium, and feeder cells are indicated. B, the fold-increase in the number of differentiated cells from PSCs at each stage. pMC (with c-Myc transduction on days 13–14), MC, myeloid cell (without c-Myc transduction). C, phase contrast images of the cells in the stages of each differentiation. Day 0, PSCs (iPSC 2A-4F-100) on primary embryonic fibroblasts. Day 7, mesodermally differentiated cells on OP9 feeder layers. Day 14, myeloid differentiated cells (myeloid cells) on OP9 feeder layers. Day 24, pMCs generated by transduction of the c-Myc gene. Day 27, pMCs treated with IL4 and GM-CSF for 3 days. Detailed information is provided in the legend for Supplementary Fig. S1.

Figure 1.

Schematic illustration showing the generation of pMCs from mouse ESCs and iPSCs. A, the protocol for the generation of pMCs. The culture medium, supplements in the medium, and feeder cells are indicated. B, the fold-increase in the number of differentiated cells from PSCs at each stage. pMC (with c-Myc transduction on days 13–14), MC, myeloid cell (without c-Myc transduction). C, phase contrast images of the cells in the stages of each differentiation. Day 0, PSCs (iPSC 2A-4F-100) on primary embryonic fibroblasts. Day 7, mesodermally differentiated cells on OP9 feeder layers. Day 14, myeloid differentiated cells (myeloid cells) on OP9 feeder layers. Day 24, pMCs generated by transduction of the c-Myc gene. Day 27, pMCs treated with IL4 and GM-CSF for 3 days. Detailed information is provided in the legend for Supplementary Fig. S1.

Close modal

Flow cytometry and microscopy

Cell samples were treated with a Fc-receptor blocking reagent (Miltenyi Biotec) for 10 minutes, stained with the fluorochrome-conjugated monoclonal antibody (mAb; Supplementary Materials and Methods) for 20 minutes, and washed three times with phosphate-buffered saline/2% FBS. The stained cell samples were analyzed on a FACSCalibur flow cytometer, and the data were analyzed using the BD CellQuest Pro software program (BD Biosciences) or FlowJo software program (TreeStar Inc.). For the morphologic analysis, cytospin specimens were stained with May-Grünwald/Giemsa (Merck).

Cell proliferation

Cell proliferation was evaluated by the direct enumeration of cells, the standard 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, and the [3H]-thymidine incorporation assay (Supplementary Materials and Methods).

Cytokine production

Cytokine levels in the culture supernatants were evaluated using enzyme-linked immunosorbent assays (ELISA; mouse TNFα and IL12p70: Affymetrix eBioscience).

Analysis of the in vivo priming of antigen-specific T cells by pMC-derived DC-like cells

ES-pMC- or iPS-pMC–derived DC-like cells (pMC-DC) were loaded with vehicle or 10 μmol/L OVA257-264 peptide for 3 hours, and were injected i.p. into C57BL/6 mice (1.0 × 105 cells/mouse) twice at 7-day intervals. In one experiment, the pMC-DCs were heat-killed by incubation at 70°C for 20 minutes or were irradiated (45 Gy) before injection. Seven days after the second injection, spleen cells were isolated from the mice and pooled for each group of 3 mice. After hemolysis, the spleen cells were cultured in 24-well culture plates (2.5 × 106/well) in RPMI-1640 supplemented with 10% horse serum, 2-ME (50 μmol/L), IL2 (10 U/mL), and OVA257–264 peptide (0.1 μmol/L). After 5 days, the cells were recovered, and the OVA-specific cytotoxic T lymphocyte (CTL) activity was analyzed using the 51Cr-release assay using EL-4 thymoma cells or MO4 melanoma cells as targets. The percentage of specific lysis was calculated as: 100 × (experimental release − spontaneous release)/(maximal release − spontaneous release). The spontaneous release and maximal release were determined in the presence of medium alone and with 1% Triton X-100, respectively.

Tumor challenge experiments

The OVA257–264 peptide (10 μmol/L)- or vehicle-loaded pMC-DCs were injected i.p. into C57BL/6 mice (1.0 × 105 cells/mouse) twice at 7-day intervals. Seven days after the second transfer, 2.0 × 105 luciferase-transduced MO4 cells were inoculated s.c. into the shaved flanks. The mice were monitored for tumor growth and survival; tumor size was measured at 5-day intervals until the mice either died or were euthanized because of tumor progression.

In vivo bioluminescent imaging

Tumor-bearing mice were injected with 200-μL d-luciferin (15 mg/mL, VivoGlo Luciferin; Promega) under 2% inhaled isoflurane anesthesia, and bioluminescence images were obtained using the IVIS Lumina II instrument with the Living Image software version 3.2 (Xenogen).

Statistical analysis

The Prism version 6.0e software program (GraphPad Software) was used for all of the statistical analyses. To compare multiple experimental groups, a one-way ANOVA with the Bonferroni post hoc test was used to assess statistical significance. For statistical analysis of the Kaplan–Meier survival curves, a log-rank (Mantel–Cox) test was used to calculate P values. P values < 0.05 were considered to denote statistical significance, and are indicated in figures by asterisks (*, P < 0.05; **, P < 0.01).

Generation of pMCs from mouse PSCs

We previously established a method for generating human myelomonocytic cells with the capacity for proliferation using the simultaneous transduction of c-Myc together with Bmi-1 or Ezh2 into human iPSC-derived myeloid cells (26, 27). In that preliminary study, mouse ES-MCs were transduced with several genes to identify those that induced proliferation. On the basis of these results, we found that mouse ES-MCs could proliferate after lentivirus-mediated transduction of c-Myc alone (data not shown). We then attempted to induce the proliferation of mouse ES-MCs and iPS-MCs through the transduction of c-Myc alone, and successfully stimulated their differentiation into DC-like APCs. The procedure for the generation of proliferating ES- or iPS-MCs (ES- or iPS-pMCs) and the subsequent differentiation of these cells into DC-like cells (pMC-DC) are shown in Fig. 1A. ESCs or iPSCs (day 0) were seeded on the OP9 feeder cell layers. After 6 to 7 days, the mesodermally differentiated cells (day 6–7) were transferred onto newly prepared OP9 cell layers in the presence of GM-CSF. On days 13 to 14, the differentiated cells (myeloid cells) were transduced with c-Myc and cultured in the presence of M-CSF and GM-CSF. After about 5 to 6 days, pmCs with protrusions appeared and propagated continuously (Supplementary Fig. S1A). The pMCs were treated with GM-CSF and IL4 (days 23–24). After 3 days of culture, the pMCs differentiated into DC-like cells (pMC-DC; days 26–27). The fold-increase in the number of differentiated cells from PSCs at each stage and the phase contrast micrographs of the cells at each differentiation stage are shown in Fig. 1B and C, respectively. The number of pMC-DCs generated via the c-Myc transduction of myeloid cells ranged from 8,000 to 40,000 times that obtained from undifferentiated stem cells. This total was estimated to be at least 160 times more than that of the MC-DCs (without c-Myc transduction).

Surface phenotype of PSC-derived cells in the stages of MCs and pMCs

The myeloid cells (days 13–14) were a heterogeneous population, almost 90% of which was characterized to be CD11b+ (Fig. 2A). In this stage, CD11b+Gr-1high, CD11c+Gr-1low, and DEC205lowGr-1+/− cells were present as minor populations. The pMCs (days 23–24) obtained by transduction of the c-Myc gene showed the upregulation of both CD11c and DEC205, and decreases in the Gr-1high population (Fig. 2B). The pMC-DCs (days 26–27) obtained after 3 days of culture in the presence of GM-CSF and IL4 showed an enhanced expression of CD11c and a downregulation of Gr-1 (Fig. 2B). In contrast to the BM-DCs, which were composed of a heterogeneous population (Fig. 2C), the pMC-DCs (GM-CSF/IL4-treated pMCs) were largely composed of a relatively homogeneous population, thus indicating that they were CD11b+/CD11c+/DEC205+/Gr-1low DC-like cells (Supplementary Fig. S1B).

Figure 2.

Characterization of mouse PSC-derived cells in the stages of myeloid cells and pMCs. Representative flow cytometry profiles of iPSC-derived myeloid cells (without c-Myc transduction), iPSC-derived pMCs (with c-Myc transduction), and BM-DCs. CD11b, CD11c, DEC205, and Gr-1 expression was analyzed using flow cytometry. GM-CSF–induced BM-DCs and GM-CSF/IL4–treated BM-DCs served as references. A, iPSC (2A-4F-100)–derived myeloid cells (days 13–14) differentiated on OP9 feeders in the presence of GM-CSF. B, top, iPSC (2A-4F-100)–derived pMCs cultured in the presence of GM-CSF and M-CSF (days 23–24). Bottom, iPSC (2A-4F-100)–derived pMCs cultured with IL4 and GM-CSF (days 26–27). C, top, GM-CSF-BM-DCs. Bottom, GM-CSF/IL4-BM-DCs. To obtain GM-CSF/IL4-BM-DCs, GM-CSF–induced BM-DCs were cultured for another 2 days in the additional presence of IL4.

Figure 2.

Characterization of mouse PSC-derived cells in the stages of myeloid cells and pMCs. Representative flow cytometry profiles of iPSC-derived myeloid cells (without c-Myc transduction), iPSC-derived pMCs (with c-Myc transduction), and BM-DCs. CD11b, CD11c, DEC205, and Gr-1 expression was analyzed using flow cytometry. GM-CSF–induced BM-DCs and GM-CSF/IL4–treated BM-DCs served as references. A, iPSC (2A-4F-100)–derived myeloid cells (days 13–14) differentiated on OP9 feeders in the presence of GM-CSF. B, top, iPSC (2A-4F-100)–derived pMCs cultured in the presence of GM-CSF and M-CSF (days 23–24). Bottom, iPSC (2A-4F-100)–derived pMCs cultured with IL4 and GM-CSF (days 26–27). C, top, GM-CSF-BM-DCs. Bottom, GM-CSF/IL4-BM-DCs. To obtain GM-CSF/IL4-BM-DCs, GM-CSF–induced BM-DCs were cultured for another 2 days in the additional presence of IL4.

Close modal

Cytokine-dependent proliferation of mouse ES- and iPS-pMCs

In the maintenance culture medium containing M-CSF and GM-CSF, mouse ES- and iPS-pMCs proliferated for more than 3 months. The doubling times of the ES-pMCs and iPS-pMCs after 15 days of c-Myc transduction were 16.73 and 17.60 hours, respectively (Fig. 3A). The doubling times of the iPS-pMCs cultured for 1, 2, and 3 months were 18.74, 17.71, and 20.06 hours, respectively (Fig. 3B). In addition, there were no significant changes in the phenotypes of these cultured pMCs (Supplementary Fig. S1C).

Figure 3.

GM-CSF–dependent proliferation of mouse ES- or iPS-pMCs. A, ESC (left)- or iPSC (2A-4F-100; right)–derived pMCs were cultured in the presence of M-CSF and GM-CSF on 24-well plates (5 × 104 cells/well), and the cells were enumerated at the indicated time points. B, proliferation of iPS-pMCs cultured for 1, 2, or 3 months. The cells were enumerated as in A. C, ES- (left) or iPS- (right) pMCs were seeded in 96-well culture plates (2 × 103 cells/well) in the presence of the indicated cytokines, and the proliferation was determined at each time point using a standard MTT assay. D, ES-pMCs were cultured in 96-well culture plates (2.0 × 103 cells/well) in the presence of graded concentrations of the indicated cytokines for 4 days, and 37 kBq/well [3H]-methylthymidine was added to the culture for the last 16 hours. The incorporated radioactivity was measured. n.s., not statistically significant. The data represent the mean ± SD of triplicate cultures. n.s., not statistically significant.

Figure 3.

GM-CSF–dependent proliferation of mouse ES- or iPS-pMCs. A, ESC (left)- or iPSC (2A-4F-100; right)–derived pMCs were cultured in the presence of M-CSF and GM-CSF on 24-well plates (5 × 104 cells/well), and the cells were enumerated at the indicated time points. B, proliferation of iPS-pMCs cultured for 1, 2, or 3 months. The cells were enumerated as in A. C, ES- (left) or iPS- (right) pMCs were seeded in 96-well culture plates (2 × 103 cells/well) in the presence of the indicated cytokines, and the proliferation was determined at each time point using a standard MTT assay. D, ES-pMCs were cultured in 96-well culture plates (2.0 × 103 cells/well) in the presence of graded concentrations of the indicated cytokines for 4 days, and 37 kBq/well [3H]-methylthymidine was added to the culture for the last 16 hours. The incorporated radioactivity was measured. n.s., not statistically significant. The data represent the mean ± SD of triplicate cultures. n.s., not statistically significant.

Close modal

To increase our understanding of the proliferation of these cells, we examined their proliferation in the presence or absence of each cytokine or with combinations of cytokines (Fig. 3C). The results showed that proliferation was dependent on GM-CSF, but not M-CSF. The additional presence of M-CSF had no effect on the proliferation of these cells. The cells proliferated in a GM-CSF concentration–dependent manner, and the maximal proliferation was induced by 10 ng/mL GM-CSF (Fig. 3D).

Differentiation of mouse ES- and iPS-pMCs into DC-like cells

In the maintenance culture medium containing M-CSF and GM-CSF, mouse ES- and iPS-pMCs proliferated and expressed major histocompatibility complex (MHC)-I, MHC-II, CD80, and CD86 (Fig. 4A). However, CD40 was expressed at a low level. Three days after treatment with IL4 plus GM-CSF, the proliferation of pMCs was impaired (Supplementary Fig. S2). In the additional presence of lipopolysaccharide (LPS), which is known to be an inducer of DC maturation, the pMCs showed enhanced expression of MHC-II, CD40, and CD86, and their proliferation was further impaired (Fig. 4B and Supplementary Fig. S2). The iPSC (2A-4F-100)–derived pMCs showed a tendency to have a substantially lower expression of MHC-II. This tendency was also observed when the cells were stimulated with LPS.

Figure 4.

ES- or iPS-pMCs can differentiate into DC-like cells. A, the surface phenotypes of mouse BM-DCs, ES-pMCs, or iPS-pMCs under different culture conditions; BM-DCs were cultured in the presence of 20 ng/mL GM-CSF alone (top), GM-CSF plus 20 ng/mL IL4 (middle), or GM-CSF, IL4, and 1 μg/mL LPS (bottom) for 3 days. ES- or iPS-pMCs (clone 2A-4F-100- or 2A-4F-136–derived iPSCs) were cultured in the presence of 30 ng/mL GM-CSF plus 30 ng/mL M-CSF (top each), GM-CSF alone (top middle each), GM-CSF plus 20 ng/mLIL4 (bottom middle each), or GM-CSF, IL4, and 1 μg/mL LPS (bottom panels each) for 3 days, and the expression levels of MHC-I (H-2Kb/H-2Db), MHC-II (I-A/E), CD40, CD80, and CD86 were analyzed using flow cytometry. Staining histograms of the indicated surface molecules (open histograms) and isotype-matched controls (filled histograms) are shown. B, BM-DCs, ES-pMCs, and iPS (2A-4F-100)-pMCs were treated as described in A, and the expression levels of MHC-II, CD40, and CD86 were analyzed. The bar graph indicates the mean fluorescence intensity (MFI) of triplicate cultures. *, P < 0.05; **, P < 0.01. C, ES- or iPS (2A-4F-100)-pMC-DCs were stimulated with 1 μg/mLLPS or 10 μg/mLOK432 for 24 hours. The TNFα and IL12p70 levels in the culture supernatants were evaluated by ELISA. The medium control served as the reference for the other culture conditions. D, T cells (1.5 × 105 cells/well) from unprimed female BALB/c mice were cultured with graded numbers of stimulator cells (BM-DCs, ES-pMC-DCs) and cultured for 6 days. A concentration of 37 kBq/well [3H]-methylthymidine was added to the culture for the last 16 hours. The incorporated radioactivity was measured. The data represent the mean ± SD of triplicate cultures.

Figure 4.

ES- or iPS-pMCs can differentiate into DC-like cells. A, the surface phenotypes of mouse BM-DCs, ES-pMCs, or iPS-pMCs under different culture conditions; BM-DCs were cultured in the presence of 20 ng/mL GM-CSF alone (top), GM-CSF plus 20 ng/mL IL4 (middle), or GM-CSF, IL4, and 1 μg/mL LPS (bottom) for 3 days. ES- or iPS-pMCs (clone 2A-4F-100- or 2A-4F-136–derived iPSCs) were cultured in the presence of 30 ng/mL GM-CSF plus 30 ng/mL M-CSF (top each), GM-CSF alone (top middle each), GM-CSF plus 20 ng/mLIL4 (bottom middle each), or GM-CSF, IL4, and 1 μg/mL LPS (bottom panels each) for 3 days, and the expression levels of MHC-I (H-2Kb/H-2Db), MHC-II (I-A/E), CD40, CD80, and CD86 were analyzed using flow cytometry. Staining histograms of the indicated surface molecules (open histograms) and isotype-matched controls (filled histograms) are shown. B, BM-DCs, ES-pMCs, and iPS (2A-4F-100)-pMCs were treated as described in A, and the expression levels of MHC-II, CD40, and CD86 were analyzed. The bar graph indicates the mean fluorescence intensity (MFI) of triplicate cultures. *, P < 0.05; **, P < 0.01. C, ES- or iPS (2A-4F-100)-pMC-DCs were stimulated with 1 μg/mLLPS or 10 μg/mLOK432 for 24 hours. The TNFα and IL12p70 levels in the culture supernatants were evaluated by ELISA. The medium control served as the reference for the other culture conditions. D, T cells (1.5 × 105 cells/well) from unprimed female BALB/c mice were cultured with graded numbers of stimulator cells (BM-DCs, ES-pMC-DCs) and cultured for 6 days. A concentration of 37 kBq/well [3H]-methylthymidine was added to the culture for the last 16 hours. The incorporated radioactivity was measured. The data represent the mean ± SD of triplicate cultures.

Close modal

To determine whether the lower expression of MHC-II is a property common to all iPSC-derived pMC lines, we examined the expression of surface antigens in other pMCs derived from a different iPSC clone (2A-4F-136). The levels of MHC-II expression of 2A-4F-136–derived iPS-pMCs were generally low, but were higher than those from 2A-4F-100–derived iPS-pMCs (Fig. 4A). Therefore, low expression of MHC-II may be a common property, but levels of expression may differ depending on the parental iPSC clone. LPS or OK432 (penicillin-killed Streptococcus pyogenes) stimulation induced the production of TNFα and IL12p70 in the IL4/GM-CSF–treated pMCs, similar to that observed in BM-DCs (Fig. 4C).

To determine the capacity of the cells to activate naïve T cells, primary MLR assays were performed. LPS-treated pMC-DCs stimulated proliferative responses in allogeneic T cells, but the responses were lower than those induced by LPS-treated BM-DCs (Fig. 4D). These findings collectively indicate that pMCs treated with IL4 plus GM-CSF exhibited some features of DCs.

In vivo priming of OVA-specific CTLs by adoptive transfer of OVA peptide-loaded pMC-DCs

To determine whether pMC-DCs could prime antigen-specific T-cell responses in vivo, OVA257–264 peptide- or vehicle-loaded pMC-DCs were i.p. transferred into syngeneic C57BL/6 mice twice at 7-day intervals. Spleen cells were isolated 7 days after the second transfer and were cultured in vitro in the presence of the OVA257–264 peptide. After 5 days, the cells were recovered and assayed for their capacity to kill OVA257–264 peptide-loaded EL-4 thymoma cells or OVA-expressing melanoma cells (MO4; Fig. 5). The CTLs primed in vivo by OVA257–264 peptide-loaded pMC-DCs or OVA257–264 peptide-loaded BM-DCs killed OVA peptide-loaded EL-4 cells, but not vehicle-loaded EL-4 cells (Fig. 5A–C). In contrast, the CTLs primed by vehicle-loaded pMC-DCs killed neither the OVA peptide-loaded nor the vehicle-loaded EL-4 cells. No differences in the capacity for antigen-specific CTL priming were observed among the DCs derived from pMCs cultured for 1, 2, or 3 months (Fig. 5D). In addition, the CTLs primed in vivo by OVA257–264 peptide-loaded pMC-DCs killed OVA-expressing melanoma, but the CTLs primed by vehicle-loaded pMC-DCs did not (Fig. 5E). Moreover, the CTLs primed in vivo by OVA257–264 peptide-loaded, irradiated pMC-DCs killed OVA-expressing melanoma. In contrast, OVA257–264 peptide-loaded, heat-killed pMC-DCs did not result in the priming of OVA-specific CTLs (Fig. 5E). It is conceivable that heat-killed pMC-DCs may fail to induce immunity due to their loss of an adjuvant effect, whereas irradiated pMC-DCs survived for a certain period in vivo and were able to prime antigen-specific CTLs. These results suggest that peptide antigen-loaded pMC-DCs could prime the antigen-specific CTL response in vivo, and their function was maintained for at least 3 months.

Figure 5.

In vivo priming of antigen-specific CTLs by ES- or iPS-pMC-DCs. Vehicle- or OVA257–264 peptide-loaded DCs were injected i.p. into 3 C57BL/6 mice twice at 7-day intervals. BM-DCs (A), ES-pMC-DCs (B), iPSC (2A-4F-100)-pMC-DCs (1.0 × 105 cells/mouse each; C), and iPSC (clone 2A-4F-100)-pMC-DCs (D) cultured for 1, 2, or 3 months. E, irradiated iPS-pMC-DCs or heat-killed iPS-pMC-DCs were used. Seven days after the second transfer, isolated spleen cells were pooled for each group and were cultured in the presence of OVA257–264 peptide. After 5 days, the cells were recovered and assayed for their ability to kill vehicle- or OVA257–264 peptide-loaded thymoma cells (EL-4; A–D), or OVA protein-expressing melanoma cells (MO4; E).

Figure 5.

In vivo priming of antigen-specific CTLs by ES- or iPS-pMC-DCs. Vehicle- or OVA257–264 peptide-loaded DCs were injected i.p. into 3 C57BL/6 mice twice at 7-day intervals. BM-DCs (A), ES-pMC-DCs (B), iPSC (2A-4F-100)-pMC-DCs (1.0 × 105 cells/mouse each; C), and iPSC (clone 2A-4F-100)-pMC-DCs (D) cultured for 1, 2, or 3 months. E, irradiated iPS-pMC-DCs or heat-killed iPS-pMC-DCs were used. Seven days after the second transfer, isolated spleen cells were pooled for each group and were cultured in the presence of OVA257–264 peptide. After 5 days, the cells were recovered and assayed for their ability to kill vehicle- or OVA257–264 peptide-loaded thymoma cells (EL-4; A–D), or OVA protein-expressing melanoma cells (MO4; E).

Close modal

Induction of protective immunity against OVA-expressing melanoma by OVA peptide-loaded pMC-DCs

To determine whether CTLs primed by pMC-DCs adoptively transferred into syngeneic C57BL/6 mice could protect against a subsequent challenge with OVA-expressing tumor cells, OVA257–264 peptide- or vehicle-loaded pMC-DCs were i.p. transferred into mice twice at 7-day intervals. Seven days after the second transfer, the mice were inoculated s.c. with MO4 (Fig. 6 and Supplementary Fig. S3). Treatment with OVA257–264 peptide-loaded ES- or iPS-pMC-DCs inhibited the growth of the inoculated MO4 for 1 month (Fig. 6A–C and Supplementary Fig. S3A–S3C) and significantly prolonged the survival of the mice compared with the vehicle-loaded pMC-DC treatment (Fig. 6D and Supplementary Fig. S3D). These observations indicate that peptide antigen–loaded pMC-DCs induced antigen-specific protective immunity against melanoma in vivo.

Figure 6.

Protection against s.c. inoculated OVA-expressing melanoma by OVA-peptide loaded ES-pMC-DCs. Vehicle- or OVA257–264 peptide-loaded BM-DCs or ES-pMC-DCs (1.0 × 105 cells/mouse each) were injected i.p. into C57BL/6 mice twice at 7-day intervals. Seven days after the second transfer, luciferase-transduced MO4 cells (2.0 × 105 cells/mouse) were inoculated s.c. into the shaved flank. A, tumor growth was monitored at the indicated time points using bioluminescence imaging (n = 3 per group). The bioluminescence images of representative mice from the individual groups are shown. B, total flux (photon/s) is shown as the mean ± SD (n = 3 mice per group). The control mice were treated with saline. C, tumor size (mm2: length × width) was measured at 5-day intervals (n = 7 per group). D, survival rate (n = 7 per group). **, P < 0.01.

Figure 6.

Protection against s.c. inoculated OVA-expressing melanoma by OVA-peptide loaded ES-pMC-DCs. Vehicle- or OVA257–264 peptide-loaded BM-DCs or ES-pMC-DCs (1.0 × 105 cells/mouse each) were injected i.p. into C57BL/6 mice twice at 7-day intervals. Seven days after the second transfer, luciferase-transduced MO4 cells (2.0 × 105 cells/mouse) were inoculated s.c. into the shaved flank. A, tumor growth was monitored at the indicated time points using bioluminescence imaging (n = 3 per group). The bioluminescence images of representative mice from the individual groups are shown. B, total flux (photon/s) is shown as the mean ± SD (n = 3 mice per group). The control mice were treated with saline. C, tumor size (mm2: length × width) was measured at 5-day intervals (n = 7 per group). D, survival rate (n = 7 per group). **, P < 0.01.

Close modal

In vivo safety of pMC-DC treatment

We next explored whether the mice were adversely affected by pMC-DC i.p. treatment. Two groups of mice treated with 2.0 × 105 or 1.0 × 107 iPS-pMC-DCs were monitored. No tumor formation or immune-related toxicities were observed for 3 months, even at a dose that was 100-fold higher than the doses used in the immunization protocols (Supplementary Fig. S4). Collectively, these observations indicate that peptide antigen-loaded pMC-DCs exhibited antigen-specific antitumor reactivity and were safe in vivo.

Mouse and human ESCs or iPSCs are capable of differentiating into functional DCs, thus providing an unlimited novel source of DCs for cancer immunotherapy (9, 17, 19). However, the process of inducing differentiation was relatively complicated, and generating practical volumes of DCs from ESCs or iPSCs has required a large number of PSCs at the start of the differentiation culture. Therefore, this process has not been suitable for the rapid mass production of cell preparations. To address this issue, we recently established a method for generating human pMCs (the precursors of DC-like cells) by transduction of c-Myc, together with antisenescence factors, such as Bmi-1 and Ezh2, into human iPSC-derived myeloid cells (26). To evaluate the possible application of these cells and their safety, we required an in vivo model not requiring xenotransplantation, which we developed for this study. Using this model, we demonstrated that pMCs could be generated from mouse ESCs or iPSCs, and that the cells could differentiate into functional APCs to induce antitumor immune responses in vivo without inducing autoimmunity or leukemia.

Mouse ESC-derived myeloid cells could acquire the capacity to proliferate in a cytokine-dependent manner after lentivirus-mediated transduction of c-Myc alone. Cell proliferation could also be induced in mouse iPSC-derived myeloid cells using the same procedure (Figs. 1 and 3). Because both the ESC- and iPSC-derived pMCs were of C57BL/6 origin, it is possible that these observations were unique to this strain. However, we made similar observations for the 129/Sv strain (data not shown). On the basis of these results, it appears that mouse PSC-derived myeloid cells acquire the capacity to proliferate continuously through the increased expression of c-Myc alone, irrespective of the mouse strain.

The c-Myc protein is a transcription factor that induces cell proliferation and cell senescence (33). Other than c-Myc, antisenescence genes, such as Bmi-1, Mdm2, and Ezh2, may be indispensable for the continuous proliferation of human ESC- or iPSC-derived myeloid cells (34–36). However, antisenescence factors were not required for acquisition of the capacity for long-term proliferation in the mouse PSC-derived myeloid cells. In addition, the increased expression of Bmi-1 had no effect on the cell proliferation, morphology, or surface markers in mouse ES- or iPS-pMCs (data not shown). This may be explained by differences between species, but the precise mechanism remains unclear.

When the medium was supplemented with GM-CSF with or without M-CSF, the mouse ES- and iPS-pMCs proliferated indefinitely. In the additional presence of IL4, the proliferation was impaired, and the cells functioned as APCs. LPS exposure induced the production of TNFα and IL12p70, and further enhanced the expression of antigen-presenting molecules and costimulatory molecules (Fig. 4). These findings were similar to the results of DC maturation. However, the T-cell stimulatory activity of ES- or iPS-pMC-DCs was slightly inferior to that of BM-DCs, based on their lower expression of T cell–stimulating molecules and the decreased production of cytokines (Fig. 4). In particular, iPSC-derived pMCs showed a tendency to have substantially lower expression of MHC-II than BM-DCs or ESC-derived pMCs (Fig. 4A). At present, the mechanisms by which iPSC-derived pMCs have downregulated MHC-II expression are not clear. The expression levels of MHC-II in iPS-pMC-DCs varied depending on the iPSC lines used as the source, and the epigenetic memory of iPSCs might affect the expression of MHC-II (37). In any case, despite the relatively lower levels of MHC-II expression, the iPS-pMC-DCs could prime the antigen-specific T cells in vivo (Fig. 5).

We successfully stimulated OVA257–264 peptide-specific CTLs in vivo by administration of OVA257–264 peptide-prepulsed ES- or iPS-pMC-DCs, leading to protection against OVA-expressing melanoma (Fig. 6 and Supplementary Fig. S3). DCs transduced with the entire tumor antigen gene expressed diverse T-cell epitopes via different MHC molecules and efficiently stimulated diverse antigen-specific T cells (38). Therefore, vaccination with pMC-DCs expressing the entire tumor antigen may be more effective for inducing antigen-specific CTLs, which could elicit potent antitumor responses. Moreover, pMC-DCs expressing multiple tumor antigens may exert a higher therapeutic effect (39–41). In addition to manipulation of the tumor antigens, modification of pMC-DCs through the transduction of immunostimulatory molecules (such as IL21) may improve the cellular immune responses (25, 42). To this end, genetic modification of pMC-DCs can be performed by introducing exogenous genes into pMCs via lentivirus transduction, followed by subsequent induction of pMC-DCs.

The human iPS-pMCs proliferated in an M-CSF–dependent manner (26), whereas the pMCs established from mouse ESCs or iPSCs in this study proliferated in a GM-CSF–dependent manner (Fig. 3C and D). There is a good concordance between the human and mouse pMCs in terms of their cytokine-dependent proliferation. In either case, the long-term proliferating capacity of pMCs may lead to the development of leukemia after administration to a patient. However, proliferation was induced by cytokine concentrations higher (>10 ng/mL) than the physiologic levels. The observation that the administration of a large amount of pMC-DCs (1.0 × 107) did not induce the onset of leukemia in this study is suggestive of their in vivo safety (Supplementary Fig. S4). Although pMCs may not proliferate in the absence of GM-CSF, we can further enhance their safety by irradiating such pMCs before they are administered as therapy.

Despite the observed in vivo safety of the treatment, there are several challenges to the possible clinical application of this technique in humans. The lentivirus-mediated delivery of the c-Myc gene permanently integrates the transgene into the genome, potentially altering the genomic features. In addition, the oncogenic c-Myc gene can lead to the development of certain cancers. In this regard, a genome-integrating but excisable system (43), and transient expression of c-Myc with Sendai-virus (44), adenoviral vectors (45), or nonviral expression [such as transduction of the c-Myc protein (46) and the use of modified mRNA (47)] may provide safer pMCs in the future. In addition, using other genes that promote the proliferation of pMCs may secure their safety.

Construction of an extensive bank of iPSCs, including the most common HLA haplotypes, is currently in progress. With this resource it will be possible to generate pMCs from the cells in the iPSC bank. When the iPS-pMC bank is established, functional APCs can be supplied within a short period, which will permit broader clinical applications of these cells. Considering the use of pMC-DCs in an allogeneic or semi-allogeneic setting as a cancer vaccine, one potential hurdle is that the alloreactive CTLs may eliminate pMC-DCs, thereby compromising their efficacy. To avoid such issues, the genes associated with the cell-surface expression of HLA-I molecules, such as TAP or β2-microglobulin, can be disrupted. Alternatively, recipient-matched HLA-I heavy chain or the β2-microglobulin–linked form of the HLA-I heavy chain can be transduced into the TAP- or β2-microglobulin gene–deficient iPS-pMCs to induce suitable CTL responses (18).

iPSC-derived DCs may resolve the problems associated with the number and quality of DCs derived from patients with cancer. However, considering the cost, labor, and preparation time, iPSC-derived DC vaccine therapy as personalized medicine is not yet suitable as a widely applicable form of cancer immunotherapy. In contrast, production of iPS-pMCs is easily scalable to rapidly provide an unlimited number of functional APCs, which would benefit patients requiring multiple vaccine doses. The iPSC-derived proliferating APCs with the capacity to stimulate tumor antigen–specific T-cell responses and with good in vivo safety profiles may provide a novel vaccine strategy for clinical cancer therapy, although further investigation is required to improve their effectiveness and to further confirm their safety.

No potential conflicts of interest were disclosed.

Conception and design: N. Hirosawa, Y. Sakamoto, Y. Uemura

Development of methodology: R. Zhang, T.-Y. Liu, S. Senju, M. Haruta, M. Suzuki

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): R. Zhang, T.-Y. Liu, M. Tatsumi, N. Ueda, H. Maki, H. Nakanishi, Y. Akatsuka

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): R. Zhang, T.-Y. Liu, N. Hirosawa, Y. Sakamoto, K. Kuzushima, Y. Uemura

Writing, review, and/or revision of the manuscript: R. Zhang, T.-Y. Liu, S. Senju, N. Hirosawa, Y. Akatsuka, Y. Sakamoto, Y. Uemura

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): R. Nakatsuka, Y. Matsuoka, Y. Sasaki, S. Tsuzuki, R. Araki, M. Abe, Y. Sonoda, K. Kuzushima

Study supervision: N. Hirosawa, Y. Sakamoto, Y. Nishimura, Y. Uemura

pCSII-EF, pCMV-VSV-G-RSV-Rev, and pCAG-HIVgp constructs were kindly provided by Dr. H. Miyoshi (RIKEN BioResource Center).

This study was supported by grants from the Japan Science and Technology Agency (JST), a Kansai Medical University Internal grant C (9), the Osaka Cancer Research Foundation (9), the Takamatsu Cancer Research Foundation (9), the Aichi Cancer Research Foundation (12, 13), the Nagono Medical Foundation (12, 13), the Daiwa Securities Health Foundation (13), the Pancreas Research Foundation of Japan (13), and the Foundation for Promotion of Cancer Research in Japan (13). R. Zhang, M. Suzuki, and Y. Uemura were supported, in part, by Grants-in-Aid 25861253, 23791850, or 23592022, respectively, from the Ministry of Education, Culture, Sports, Science and Technology (MEXT) of Japan. T.-Y. Liu was supported by the National Natural Science Foundation of China (81101882). K. Kuzushima was supported by the Takeda Science Foundation (12–14).

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.
Eksioglu
EA
,
Eisen
S
,
Reddy
V
. 
Dendritic cells as therapeutic agents against cancer
.
Front Biosci (Landmark Ed)
2010
;
15
:
321
47
.
2.
Radford
KJ
,
Tullett
KM
,
Lahoud
MH
. 
Dendritic cells and cancer immunotherapy
.
Curr Opin Immunol
2014
;
27C
:
26
32
.
3.
Higano
CS
,
Schellhammer
PF
,
Small
EJ
,
Burch
PA
,
Nemunaitis
J
,
Yuh
L
, et al
Integrated data from 2 randomized, double-blind, placebo-controlled, phase 3 trials of active cellular immunotherapy with sipuleucel-T in advanced prostate cancer
.
Cancer
2009
;
115
:
3670
9
.
4.
Schuler
G
. 
Dendritic cells in cancer immunotherapy
.
Eur J Immunol
2010
;
40
:
2123
30
.
5.
Takahashi
K
,
Yamanaka
S
. 
Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors
.
Cell
2006
;
126
:
663
76
.
6.
Takahashi
K
,
Tanabe
K
,
Ohnuki
M
,
Narita
M
,
Ichisaka
T
,
Tomoda
K
, et al
Induction of pluripotent stem cells from adult human fibroblasts by defined factors
.
Cell
2007
;
131
:
861
72
.
7.
Lorenzo
IM
,
Fleischer
A
,
Bachiller
D
. 
Generation of mouse and human induced pluripotent stem cells (iPSC) from primary somatic cells
.
Stem Cell Rev
2013
;
9
:
435
50
.
8.
Fairchild
PJ
,
Brook
FA
,
Gardner
RL
,
Graca
L
,
Strong
V
,
Tone
Y
, et al
Directed differentiation of dendritic cells from mouse embryonic stem cells
.
Curr Biol
2000
;
10
:
1515
8
.
9.
Senju
S
,
Hirata
S
,
Matsuyoshi
H
,
Masuda
M
,
Uemura
Y
,
Araki
K
, et al
Generation and genetic modification of dendritic cells derived from mouse embryonic stem cells
.
Blood
2003
;
101
:
3501
8
.
10.
Zhan
X
,
Dravid
G
,
Ye
Z
,
Hammond
H
,
Shamblott
M
,
Gearhart
J
, et al
Functional antigen-presenting leucocytes derived from human embryonic stem cells in vitro
.
Lancet
2004
;
364
:
163
71
.
11.
Matsuyoshi
H
,
Senju
S
,
Hirata
S
,
Yoshitake
Y
,
Uemura
Y
,
Nishimura
Y
. 
Enhanced priming of antigen-specific CTLs in vivo by embryonic stem cell-derived dendritic cells expressing chemokine along with antigenic protein: application to antitumor vaccination
.
J Immunol
2004
;
172
:
776
86
.
12.
Fukuma
D
,
Matsuyoshi
H
,
Hirata
S
,
Kurisaki
A
,
Motomura
Y
,
Yoshitake
Y
, et al
Cancer prevention with semi-allogeneic ES cell-derived dendritic cells
.
Biochem Biophys Res Commun
2005
;
335
:
5
13
.
13.
Matsuyoshi
H
,
Hirata
S
,
Yoshitake
Y
,
Motomura
Y
,
Fukuma
D
,
Kurisaki
A
, et al
Therapeutic effect of alpha-galactosylceramide-loaded dendritic cells genetically engineered to express SLC/CCL21 along with tumor antigen against peritoneally disseminated tumor cells
.
Cancer Sci
2005
;
96
:
889
96
.
14.
Motomura
Y
,
Senju
S
,
Nakatsura
T
,
Matsuyoshi
H
,
Hirata
S
,
Monji
M
, et al
Embryonic stem cell-derived dendritic cells expressing glypican-3, a recently identified oncofetal antigen, induce protective immunity against highly metastatic mouse melanoma, B16-F10
.
Cancer Res
2006
;
66
:
2414
22
.
15.
Slukvin
II
,
Vodyanik
MA
,
Thomson
JA
,
Gumenyuk
ME
,
Choi
KD
. 
Directed differentiation of human embryonic stem cells into functional dendritic cells through the myeloid pathway
.
J Immunol
2006
;
176
:
2924
32
.
16.
Su
Z
,
Frye
C
,
Bae
KM
,
Kelley
V
,
Vieweg
J
. 
Differentiation of human embryonic stem cells into immunostimulatory dendritic cells under feeder-free culture conditions
.
Clin Cancer Res
2008
;
14
:
6207
17
.
17.
Senju
S
,
Suemori
H
,
Zembutsu
H
,
Uemura
Y
,
Hirata
S
,
Fukuma
D
, et al
Genetically manipulated human embryonic stem cell-derived dendritic cells with immune regulatory function
.
Stem Cells
2007
;
25
:
2720
9
.
18.
Matsunaga
Y
,
Fukuma
D
,
Hirata
S
,
Fukushima
S
,
Haruta
M
,
Ikeda
T
, et al
Activation of antigen-specific cytotoxic T lymphocytes by beta 2-microglobulin or TAP1 gene disruption and the introduction of recipient-matched MHC class I gene in allogeneic embryonic stem cell-derived dendritic cells
.
J Immunol
2008
;
181
:
6635
43
.
19.
Senju
S
,
Haruta
M
,
Matsunaga
Y
,
Fukushima
S
,
Ikeda
T
,
Takahashi
K
, et al
Characterization of dendritic cells and macrophages generated by directed differentiation from mouse induced pluripotent stem cells
.
Stem Cells
2009
;
27
:
1021
31
.
20.
Tseng
SY
,
Nishimoto
KP
,
Silk
KM
,
Majumdar
AS
,
Dawes
GN
,
Waldmann
H
, et al
Generation of immunogenic dendritic cells from human embryonic stem cells without serum and feeder cells
.
Regen Med
2009
;
4
:
513
26
.
21.
Senju
S
,
Hirata
S
,
Motomura
Y
,
Fukuma
D
,
Matsunaga
Y
,
Fukushima
S
, et al
Pluripotent stem cells as source of dendritic cells for immune therapy
.
Int J Hematol
2010
;
91
:
392
400
.
22.
Senju
S
,
Matsunaga
Y
,
Fukushima
S
,
Hirata
S
,
Matsuyoshi
H
,
Nishimura
Y
. 
Pluripotent stem cell-derived dendritic cells for immunotherapy
.
Front Biosci (Elite Ed)
2010
;
2
:
1520
7
.
23.
Senju
S
,
Matsunaga
Y
,
Fukushima
S
,
Hirata
S
,
Motomura
Y
,
Fukuma
D
, et al
Immunotherapy with pluripotent stem cell-derived dendritic cells
.
Semin Immunopathol
2011
;
33
:
603
12
.
24.
Silk
KM
,
Silk
JD
,
Ichiryu
N
,
Davies
TJ
,
Nolan
KF
,
Leishman
AJ
, et al
Cross-presentation of tumour antigens by human induced pluripotent stem cell-derived CD141(+)XCR1 +dendritic cells
.
Gene Ther
2012
;
19
:
1035
40
.
25.
Iwamoto
H
,
Ojima
T
,
Hayata
K
,
Katsuda
M
,
Miyazawa
M
,
Iida
T
, et al
Antitumor immune response of dendritic cells (DCs) expressing tumor-associated antigens derived from induced pluripotent stem cells: in comparison to bone marrow-derived DCs
.
Int J Cancer
2014
;
134
:
332
41
.
26.
Haruta
M
,
Tomita
Y
,
Yuno
A
,
Matsumura
K
,
Ikeda
T
,
Takamatsu
K
, et al
TAP-deficient human iPS cell-derived myeloid cell lines as unlimited cell source for dendritic cell-like antigen-presenting cells
.
Gene Ther
2013
;
20
:
504
13
.
27.
Koba
C
,
Haruta
M
,
Matsunaga
Y
,
Matsumura
K
,
Haga
E
,
Sasaki
Y
, et al
Therapeutic effect of human iPS-cell-derived myeloid cells expressing IFN-beta against peritoneally disseminated cancer in xenograft models
.
PLoS ONE
2013
;
8
:
e67567
.
28.
Fukushima
S
,
Hirata
S
,
Motomura
Y
,
Fukuma
D
,
Matsunaga
Y
,
Ikuta
Y
, et al
Multiple antigen-targeted immunotherapy with alpha-galactosylceramide-loaded and genetically engineered dendritic cells derived from embryonic stem cells
.
J Immunother
2009
;
32
:
219
31
.
29.
Araki
R
,
Uda
M
,
Hoki
Y
,
Sunayama
M
,
Nakamura
M
,
Ando
S
, et al
Negligible immunogenicity of terminally differentiated cells derived from induced pluripotent or embryonic stem cells
.
Nature
2013
;
494
:
100
4
.
30.
Nakano
T
,
Kodama
H
,
Honjo
T
. 
In vitro development of primitive and definitive erythrocytes from different precursors
.
Science
1996
;
272
:
722
4
.
31.
Falo
LD
 Jr
,
Kovacsovics-Bankowski
M
,
Thompson
K
,
Rock
KL
. 
Targeting antigen into the phagocytic pathway in vivo induces protective tumour immunity
.
Nat Med
1995
;
1
:
649
53
.
32.
Haruta
M
,
Tomita
Y
,
Imamura
Y
,
Matsumura
K
,
Ikeda
T
,
Takamatsu
K
, et al
Generation of a large number of functional dendritic cells from human monocytes expanded by forced expression of cMYC plus BMI1
.
Hum Immunol
2013
;
74
:
1400
8
.
33.
Lee
S
,
Schmitt
CA
,
Reimann
M
. 
The Myc/macrophage tango: oncogene-induced senescence, Myc style
.
Semin Cancer Biol
2011
;
21
:
377
84
.
34.
Jacobs
JJ
,
Kieboom
K
,
Marino
S
,
DePinho
RA
,
van Lohuizen
M
. 
The oncogene and Polycomb-group gene bmi-1 regulates cell proliferation and senescence through the ink4a locus
.
Nature
1999
;
397
:
164
8
.
35.
Bracken
AP
,
Kleine-Kohlbrecher
D
,
Dietrich
N
,
Pasini
D
,
Gargiulo
G
,
Beekman
C
, et al
The Polycomb group proteins bind throughout the INK4A-ARF locus and are disassociated in senescent cells
.
Genes Dev
2007
;
21
:
525
30
.
36.
Kamminga
LM
,
Bystrykh
LV
,
de Boer
A
,
Houwer
S
,
Douma
J
,
Weersing
E
, et al
The Polycomb group gene Ezh2 prevents hematopoietic stem cell exhaustion
.
Blood
2006
;
107
:
2170
9
.
37.
Hu
Q
,
Friedrich
AM
,
Johnson
LV
,
Clegg
DO
. 
Memory in induced pluripotent stem cells: reprogrammed human retinal-pigmented epithelial cells show tendency for spontaneous redifferentiation
.
Stem Cells
2010
;
28
:
1981
91
.
38.
Perez-Diez
A
,
Butterfield
LH
,
Li
L
,
Chakraborty
NG
,
Economou
JS
,
Mukherji
B
. 
Generation of CD8+ and CD4+ T-cell response to dendritic cells genetically engineered to express the MART-1/Melan-A gene
.
Cancer Res
1998
;
58
:
5305
9
.
39.
Wang
B
,
He
J
,
Liu
C
,
Chang
LJ
. 
An effective cancer vaccine modality: lentiviral modification of dendritic cells expressing multiple cancer-specific antigens
.
Vaccine
2006
;
24
:
3477
89
.
40.
Kavanagh
B
,
Ko
A
,
Venook
A
,
Margolin
K
,
Zeh
H
,
Lotze
M
, et al
Vaccination of metastatic colorectal cancer patients with matured dendritic cells loaded with multiple major histocompatibility complex class I peptides
.
J Immunother
2007
;
30
:
762
72
.
41.
Blalock
LT
,
Landsberg
J
,
Messmer
M
,
Shi
J
,
Pardee
AD
,
Haskell
R
, et al
Human dendritic cells adenovirally-engineered to express three defined tumor antigens promote broad adaptive and innate immunity
.
Oncoimmunology
2012
;
1
:
287
357
.
42.
Aravindaram
K
,
Wang
PH
,
Yin
SY
,
Yang
NS
. 
Tumor-associated antigen/IL-21-transduced dendritic cell vaccines enhance immunity and inhibit immunosuppressive cells in metastatic melanoma
.
Gene Ther
2014
;
21
:
457
67
.
43.
Sommer
CA
,
Sommer
AG
,
Longmire
TA
,
Christodoulou
C
,
Thomas
DD
,
Gostissa
M
, et al
Excision of reprogramming transgenes improves the differentiation potential of iPS cells generated with a single excisable vector
.
Stem Cells
2010
;
28
:
64
74
.
44.
Ban
H
,
Nishishita
N
,
Fusaki
N
,
Tabata
T
,
Saeki
K
,
Shikamura
M
, et al
Efficient generation of transgene-free human induced pluripotent stem cells (iPSCs) by temperature-sensitive Sendai virus vectors
.
Proc Natl Acad Sci U S A
2011
;
108
:
14234
9
.
45.
Stadtfeld
M
,
Nagaya
M
,
Utikal
J
,
Weir
G
,
Hochedlinger
K
. 
Induced pluripotent stem cells generated without viral integration
.
Science
2008
;
322
:
945
9
.
46.
Kim
D
,
Kim
CH
,
Moon
JI
,
Chung
YG
,
Chang
MY
,
Han
BS
, et al
Generation of human induced pluripotent stem cells by direct delivery of reprogramming proteins
.
Cell Stem Cell
2009
;
4
:
472
6
.
47.
Warren
L
,
Manos
PD
,
Ahfeldt
T
,
Loh
YH
,
Li
H
,
Lau
F
, et al
Highly efficient reprogramming to pluripotency and directed differentiation of human cells with synthetic modified mRNA
.
Cell Stem Cell
2010
;
7
:
618
30
.