Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2021 May;9(5):e002432.
doi: 10.1136/jitc-2021-002432.

In situ delivery of iPSC-derived dendritic cells with local radiotherapy generates systemic antitumor immunity and potentiates PD-L1 blockade in preclinical poorly immunogenic tumor models

Affiliations

In situ delivery of iPSC-derived dendritic cells with local radiotherapy generates systemic antitumor immunity and potentiates PD-L1 blockade in preclinical poorly immunogenic tumor models

Takaaki Oba et al. J Immunother Cancer. 2021 May.

Abstract

Background: Dendritic cells (DCs) are a promising therapeutic target in cancer immunotherapy given their ability to prime antigen-specific T cells, and initiate antitumor immune response. A major obstacle for DC-based immunotherapy is the difficulty to obtain a sufficient number of functional DCs. Theoretically, this limitation can be overcome by using induced pluripotent stem cells (iPSCs); however, therapeutic strategies to engage iPSC-derived DCs (iPSC-DCs) into cancer immunotherapy remain to be elucidated. Accumulating evidence showing that induction of tumor-residing DCs enhances immunomodulatory effect of radiotherapy (RT) prompted us to investigate antitumor efficacy of combining intratumoral administration of iPSC-DCs with local RT.

Methods: Mouse iPSCs were differentiated to iPSC-DCs on OP9 stromal cells expressing the notch ligand delta-like 1 in the presence of granulocyte macrophage colony-stimulating factor. Phenotype and the capacities of iPSC-DCs to traffic tumor-draining lymph nodes (TdLNs) and prime antigen-specific T cells were evaluated by flow cytometry and imaging flow cytometry. Antitumor efficacy of intratumoral injection of iPSC-DCs and RT was tested in syngeneic orthotopic mouse tumor models resistant to anti-PD-1 ligand 1 (PD-L1) therapy.

Results: Mouse iPSC-DCs phenotypically resembled conventional type 2 DCs, and had a capacity to promote activation, proliferation and effector differentiation of antigen-specific CD8+ T cells in the presence of the cognate antigen in vitro. Combination of in situ administration of iPSC-DCs and RT facilitated the priming of tumor-specific CD8+ T cells, and synergistically delayed the growth of not only the treated tumor but also the distant non-irradiated tumors. Mechanistically, RT enhanced trafficking of intratumorally injected iPSC-DCs to the TdLN, upregulated CD40 expression, and increased the frequency of DC/CD8+ T cell aggregates. Phenotypic analysis of tumor-infiltrating CD8+ T cells and myeloid cells revealed an increase of stem-like Slamf6+ TIM3- CD8+ T cells and PD-L1 expression in tumor-associated macrophages and DCs. Consequently, combined therapy rendered poorly immunogenic tumors responsive to anti-PD-L1 therapy along with the development of tumor-specific immunological memory.

Conclusions: Our findings illustrate the translational potential of iPSC-DCs, and identify the therapeutic efficacy of a combinatorial platform to engage them for overcoming resistance to anti-PD-L1 therapy in poorly immunogenic tumors.

Keywords: adaptive immunity; dendritic cells; programmed cell death 1 receptor; radioimmunotherapy; vaccination.

PubMed Disclaimer

Conflict of interest statement

Competing interests: AO was a co-founder of Tactiva Therapeutics and receives research support from AstraZeneca and Tessaro.

Figures

Figure 1
Figure 1
Generation of dendritic cells (DCs) from mouse induced pluripotent stem cells (iPSCs). (A) Schematic illustration showing the generation of DCs from iPSCs. (B) Phenotypic analysis of iPSC-derived cells on day 26, and bone marrow-derived cells on day 10 differentiated in the presence of GM-CSF in vitro. (C) Representative flow cytometric plots showing IRF4 and IRF8 expression of CD11c+ MHC class II+ iPSC-derived DCs (iPSC-DCs). (D) Phenotypic analysis of CD11c+ MHC class II+ cells. Representative histogram showing CD24, DEC205, CD80, CD11b, B220, CD8α, XCR1 and F4/80 expressions on CD11c+ MHC class II+ 2A-4F-118 iPSC-DCs, 2A-4F-136 iPSC-DCs and bone marrow-derived DCs (BM-DCs) (red). Isotype-matched controls are shown in blue. Number denotes per cent positive cells for each marker. Data shown are representative of three independent experiments. FMT, fluorescence minus two; GM-CSF, granulocyte macrophage colony-stimulating factor; IRF4/8, interferon regulatory factor-4 and 8; MHC, Major Histocompatibility Complex; OP9-DL-1, OP9 cells expressing a notch ligand, delta-like-1; pHEMA, poly 2-hydroxyethyl methacrylate.
Figure 2
Figure 2
iPSC-derived DCs (iPSC-DCs) activate antigen-specific CD8+ T cells in vitro. (A, B) Pmel-1 T cells (CD90.1+ CD8+) were isolated from splenocytes by CD8α positive selection, labeled with CFSE, and co-cultured with iPSC-DCs in the presence of influenza nucleoprotein (NP) epitope NP366–374 or hgp10025–33 and IL-2 (60 IU/mL). Two days later, cells were harvested for flow cytometric analysis. (A) Representative histogram showing CFSE dilution in Pmel-1 T cells. (B) Representative flow cytometric plots showing CD62L, CD25, and PD-1 expression in Pmel-1 T cells. Numbers denote per cent dividing CD62L, CD25+, or PD-1+ cells. (C) Activated Pmel-1 T cells from the experiment (A, B) were co-cultured with hgp10025–33 or NP366–374 in the presence of antigen-presenting cells (splenocytes from C57BL/6 mice) for another 2 days. Representative flow cytometric plots show intracellular production of IFNγ, TNFα, and IL-2 in Pmel-1 T cells activated by iPSC-DCs. Numbers denote per cent positive cells. Data shown are representative of two independent experiments. CFSE, carboxyfluorescein succinimidyl ester; IFNγ, interferon gamma; IL-2, interleukin 2; PD-1, programmed cell death protein 1; TNFα, tumor necrosis factor alpha.
Figure 3
Figure 3
Synergistic antitumor efficacy of in situ iPSC-DC administration and local radiotherapy (RT) against poorly immunogenic tumors. (A, B) Tumor volume curves (mean) and survival curves in AT-3 (n=7) (A) and B16 (n=6–7) (B) tumor-bearing mice in different treatment as indicated. NT, non treatment. Individual tumor volume curves are shown in online supplemental figure 1. NS not significant, *p<0.05, **p<0.01, ***p<0.001 by a log-rank test. Data shown are representative of two independent experiments. Mean±SEM. iPSC-DCs, induced pluripotent stem cell-derived dendritic cells; i.t., intratumorally.
Figure 4
Figure 4
RT augments trafficking of intratumorally injected iPSC-DCs to the TdLN, upregulates CD40 expression, and increases the frequency of DC/CD8+ T cell aggregates. (A) Experimental set-up. Mice bearing AT-3 or AT-3-GFP tumors had an intratumoral iPSC-DC injection with or without local RT (9 Gy). Gating strategy for identifying fluorescently (CTO: CellTracker Orange)-labeled iPSC-DCs by flow cytometry and imaging flow cytometry is shown in online supplemental figure 4 and online supplemental figure 5, respectively. FACS, fluorescence-activated cell sorting. (B) Frequency of iPSC-DCs in the tumor and TdLN in different treatment groups as indicated (n=6). (C) Representative flow cytometric plots showing annexin V and near-IR expression of iPSC-DCs in the tumor and TdLN. The data panel shows the frequency of annexin V+ iPSC-DCs in the tumor and TdLN (n=3–6). Data with total iPSC-DCs >10 from the experiment (B) were evaluated. (D–G) Representative dot plots and frequencies of GFP+ iPSC-DCs as single cells or engaged in cell aggregates (D), CD40+ CD11c+ cells in iPSC-DCs (E), GFP+ iPSC-DC in direct contact with CD8+ T-cell (F), and GFP+ iPSC-DC-XCR1+ DC in direct contact with CD8+ T-cell (G) analyzed by imaging flow cytometry. Representative images are shown in online supplemental figure 6. Each dot represents biologically independent mice (B–G). One-way ANOVA with Tukey’s multiple comparisons (C) and two-tailed unpaired t-test (B, D–G). Mean±SEM. ANOVA, analysis of variance; GFP, green fluorescent protein; iPSC-DCs, induced pluripotent stem cell-derived dendritic cells; IR, infrared; i.t., intratumorally; RT, radiotherapy; TdLN, tumor-draining lymph node.
Figure 5
Figure 5
A combination of in situ iPSC-DC injection and RT increases stem-like progenitor exhausted CD8+ T cells and PD-L1 expression in myeloid cells in the tumor. (A) Experimental set-up. (B) Phenotypic analysis of CD8+ T cells among CD45+ cells in AT-3 tumors in different treatment groups as indicated. Numbers denote per cent Slamf6+ TIM3 cells. Representative flow cytometric plots showing expression of Slamf6 and TIM3 in CD8+ TILs. The data panel shows the frequency of Slamf6+ TIM3 cells in CD8+ TILs (n=7). (C) Phenotypic characterization of Slamf6+ TIM3 and Slamf6 TIM3+ CD8+ TILs (n=6). (D) PD-L1 expression (MFI: median fluorescence intensity) of Ly6c CD11c+ class II+ F4/80hi CD24− tumor-associated macrophages (TAMs) (upper) and Ly6c CD11c+ class II+ F4/80lo CD24+ DCs (lower) in AT-3 tumors (n=7). Gating strategy for identifying TAMs and DCs is shown in online supplemental figure 3. NS not significant, *p<0.05, **p<0.01, ***p<0.001, ****p<0.0001 by one-way ANOVA with Tukey’s multiple comparisons (B, D) and two-tailed paired t-test (C). Each dot represents biologically independent mice (B, D). Data shown are representative of two independent experiments. Mean±SEM. ANOVA, analysis of variance; iPSC-DC, induced pluripotent stem cell-derived dendritic cells; i.t., intratumorally; PD-1, programmed cell death protein 1; PD-L1, PD-1 ligand 1; RT, radiotherapy; TILs, tumor-infiltrating lymphocytes; TIM3, T cell immunoglobulin and mucin domain-containing protein 3.
Figure 6
Figure 6
In situ injection of iPSC-DCs with local RT increases antigen-specific CD8+ T cell infiltrates in the tumor. (A) Experimental set-up. (B) Representative flow cytometric plots showing OT-I T cells (CD90.1+ CD8+) in CD45+ cells in tumors. Data panels show numbers (/g) (tumor) of OT-I T cells (n=3). (C) Representative flow cytometric plots showing expression of Slamf6 and TIM3 in OT-I (CD90.1+) and endogenous (CD90.2+) CD8+ TILs. Numbers denote per cent Slamf6+ TIM3 T cells. (D) Tumor growth curves (mean) (left) and tumor weight (right) of B16-OVA tumor-bearing mice in different treatment as indicated. (n=5). (E) Numbers (/g) (tumor) of OT-I T cells (n=5). Each dot represents biologically independent mice (B, D–F). NS not significant, *p<0.05, **p<0.01, ***p<0.001, one-way ANOVA with Tukey’s multiple comparisons (B, D, E) and two-tailed paired (C) and unpaired (F) t-test. Mean±SEM. ANOVA, analysis of variance; BM-DCs, bone marrow-derived dendritic cells; iPSC-DC, induced pluripotent stem cell-derived dendritic cells; i.t., intratumorally; OVA, ovalbumin; RT, radiotherapy; TILs, tumor-infiltrating lymphocytes; TIM3, T cell immunoglobulin and mucin domain-containing protein 3.
Figure 7
Figure 7
Combining in situ administration of iPSC-DC with RT attenuates growth of distant untreated tumors.(A) Experimental set-up. (B) Frequency of peripheral blood effector memory CD44+ CD62L CD8+ T cells (Tem) in different treatment groups as indicated (n=6–7). (C) Tumor growth curves (mean) (left) and tumor weight (right) of distant untreated tumors in bilateral AT-3 tumor-bearing mice in different treatment as indicated (n=6–7). Individual tumor volume curves are shown in online supplemental figure 8. (D, E). Frequency of CD8+ T cells among CD45+ cells (D) and CD8+/CD11b+ cell ratio (E) in untreated distant AT-3 tumors in different treatment groups as indicated (n=6–7). (F) Representative images of immunohistochemistry for CD8 in untreated distant AT-3 tumors. Scale bars, 100 µm. Data panels show mean numbers of CD8+ cells per each high-power field (HPF) within five different areas. Each dot represents biologically independent mice (B–E). NS not significant, *p<0.05, **p<0.01, ***p<0.001, ****p<0.0001 by one-way ANOVA with Tukey’s multiple comparisons. Data shown are representative of two independent experiments. Mean±SEM. ANOVA, analysis of variance; iPSC-DC, induced pluripotent stem cell-derived dendritic cells; i.t., intratumorally; RT, radiotherapy; TILs, tumor-infiltrating lymphocytes.
Figure 8
Figure 8
Combined therapy of in situ iPSC-DC injection and RT renders tumors responsive to anti-PD-L1 therapy, has potential to eradicate poorly T cell-inflamed tumors, and establishes immunological memory. (A) Experimental set-up. Mice bearing AT-3 tumors in the left fourth mammary gland were treated with intratumoral iPSC-DC injections, RT, and anti-PD-L1 antibody (αPD-L1 Ab) or isotype Ab. (B) Tumor growth curves (individual) in AT-3 tumor-bearing mice in different treatment groups as indicated (n=5–9). (C) Survival curves in AT-3 tumor-bearing mice in different treatment as indicated. (D) Naïve mice and surviving mice from the experiment (B, C) were rechallenged with AT-3 and MC38 in the right flank at day 123 (AT-3) and on back at day 127 (B16), respectively. NS not significant, **p<0.01, ***p<0.001, ****p=0.0001 by a log-rank test (C). Mean±SEM. Data shown are representative of two independent experiments. i.p., intraperitoneally; iPSC-DC, induced pluripotent stem cell-derived dendritic cells; i.t., intratumorally; PD-L1, PD-1 ligand 1; RT, radiotherapy.

Similar articles

Cited by

References

    1. Schuler G, Schuler-Thurner B, Steinman RM. The use of dendritic cells in cancer immunotherapy. Curr Opin Immunol 2003;15:138–47. 10.1016/s0952-7915(03)00015-3 - DOI - PubMed
    1. Palucka K, Banchereau J. Cancer immunotherapy via dendritic cells. Nat Rev Cancer 2012;12:265–77. 10.1038/nrc3258 - DOI - PMC - PubMed
    1. Wculek SK, Cueto FJ, Mujal AM, et al. . Dendritic cells in cancer immunology and immunotherapy. Nat Rev Immunol 2020;20:7–24. 10.1038/s41577-019-0210-z - DOI - PubMed
    1. Anguille S, Smits EL, Lion E, et al. . Clinical use of dendritic cells for cancer therapy. Lancet Oncol 2014;15:e257–67. 10.1016/S1470-2045(13)70585-0 - DOI - PubMed
    1. Saxena M, Balan S, Roudko V, et al. . Towards superior dendritic-cell vaccines for cancer therapy. Nat Biomed Eng 2018;2:341–6. 10.1038/s41551-018-0250-x - DOI - PMC - PubMed

Publication types

MeSH terms