Strategies to reprogram the tumor microenvironment are being explored to improve cancer immunotherapy. In one approach, we have targeted dendritic cells (DC) to improve their function with adjuvant vector cells (aAVC) that are engineered from NKT ligand-loaded CD1d+ allogeneic cells transfected with tumor antigen mRNAs. Here, we report the finding that this approach also programs local immune responses by establishing tertiary lymphoid structures (TLS), which include expanded antigen-specific CD8+ T-cell clones, mobilized DCs, and normalized tumor vasculature. aAVC therapy also expanded specific Vβ-expressing antitumor T-cell clones, leading to the formation of long-term memory T cells. When combined with PD-1 blockade, aAVC infusion triggered regression of poorly immunogenic tumor cells that did not respond to PD-1 blockade alone, as well as expansion of antigen-specific CD8+ T-cell clones in the tumor. The findings of this study help to inform a next-generation platform for the generation of efficacious cancer vaccines. Cancer Res; 76(13); 3756–66. ©2016 AACR.

Cancer immunotherapy has recently enjoyed a renaissance as a result of new and more effective approaches (1). For example, adoptive T-cell transfer with tumor antigen–specific TCR or chimeric antigen receptor (CAR) gene-transferred T cells has shown antitumor effects on some cancers (2, 3) Also, blocking immune checkpoints using mAbs to CTL–associated protein-4 (CTLA-4) or programmed cell death protein-1 (PD-1) has demonstrated clinical efficacy (4). Another promising approach involves active immunization using a cancer vaccine, and in this context the function of dendritic cells (DC) appears to be very important (5, 6). The next breakthroughs for its use as an effective antitumor immunotherapy would be the induction of memory T cells and the modulation of tumor microenvironment in the light of systemic and local immune responses. Once established, memory T cells could quickly respond to emerging cancer cells in the recurrence and metastasis. The persistence of functional memory T cells was desired for immunotherapies. In addition, therapeutic options to overcome the barriers of tumor microenvironment and to support T-cell infiltration and function in tumors are required. For this perspective, organized lymphoid aggregates found locally at tumor sites, termed tertiary lymphoid structures (TLS), should be a major focus that may lead effector T cells to recruit to tumor cells (7, 8).

For the success of immunotherapy, it has been recently argued that combining multiple therapies should be beneficial, because different types of immune responses may prevent tumor cells from immune escape. In this point, a strategy linking innate and adaptive immune responses is attractive and promising, for example, they can attack MHC+ or MHC tumor cells. NKT cells have bipotential capacity and can suppress or activate immune responses. Once activated by their ligands, they can closely contact DCs and mature them systemically in lung, liver, spleen, and bone marrow. DCs thus licensed by activated NKT cells via both CD40L on NKT cells and their simultaneous production of inflammatory cytokines, for example, IFNγ and TNFα (9). Then, the DCs can induce adaptive immunity in infection and tumor models (10–13). We and others have shown that coadministration of NKT cell ligand and antigen can induce adaptive immunity (10, 14, 15).

On the basis of these evidences, we previously illustrated the concept of a unique approach of using NKT ligand–loaded CD1d+ allogeneic cells transfected with tumor antigen mRNA as artificial adjuvant vector cells (aAVC; refs. 16, 17). The first generation of aAVCs demonstrated certain of its immunologic features inducing both innate and adaptive immunity in the lymphoid organs through antigen-captured DCs in situ. To develop the aAVC therapy, we assumed that the optimal amount of target protein delivered to in vivo DC would affect the magnitude of the ensuing immune responses in the tumor. By the new method, aAVCs can produce a suitable amount of protein for protein-based clinical immunotherapy and be developed as a current type of aAVC. Here we assessed the antitumor effects and further analyzed the cellular mechanism against tumor in terms of local and systemic immune responses. This study not only demonstrates the efficacy of our approach, but will also help to identify the key components needed for successful future therapeutics.

Reagents

Human IL2 was purchased from Shionogi & Co., LTD. Human recombinant IL7 and IL15 were purchased from Peprotech, Inc. The clinical grade of α-GalCer was provided from Regimmune. OVA257–264 peptide (SIINFEKL) and WT1235–243 (CMTWNQMNL) were obtained from Toray Research Center, Inc. The antibodies used in this study were purchased and described in Supplementary Table S1. OVA-tetramer was purchased from MBL. WT1-tetramer (HLA-A24) and HIV-tetramer (HLA-A24) were produced by our group. A FACSCalibur or FACSCanto II instrument and CELLQuest, Diva (BD Biosciences), and FlowJo (Tree Star) software were used for the analysis. Anti-PD-1 mAb (RPMI-14) was produced as described previously (18). Recombinant human WT1 was synthesized in RIKEN.

Mice and cell lines

B16 and HEK293 cell lines were purchased from ATCC. NIH3T3 and WEHI3B cell lines were obtained from the RIKEN Cell Bank. MO4 (19) and J558 (20) cell lines were received from Dr. R.M. Steinman (The Rockefeller University, New York, NY) and has been routinely tested for OVA expression or for IgA and H2-Kd expression by our hands, respectively. The J558-WT1 cell line was established by transfection of a human WT1 cDNA expression vector into J558. All cell lines were tested according to the manufacturer's protocol and proved to be mycoplasma free (Mycoplasma Detection Kit; Minerva Biolabs). C57BL/6 or BALB/c mice were purchased from CLEA Japan. Ly5.1 congenic OT-1 mice were generated by cross/backcross breeding of OT-1 with B6. Ly5.1 mice and screening for the presence of Vα2 and Ly5.1 and absence of Ly5.2 by flow cytometry. OT-1 TCR transgenic mice, CD11c-DTR/GFP mice (21), and XCR1-DTR-venus mice (22) and other all the mice were maintained under specific pathogen-free conditions and studied in compliance with our institutional guidelines.

Cell preparation

Human PBMCs were isolated from healthy volunteers. All studies were approved by the RIKEN institutional review board. Murine bone marrow–derived DCs were generated in the presence of GM-CSF for 6 days as described previously (23). The preparation of aAVCs using HEK293 or NIH3T3 was performed as described previously (17). Briefly, cells resuspended in OptiMEM and RNA were transferred to a cuvette and then the cell suspension was pulsed in ECM 830 Square Wave Electroporation System (Harvard Apparatus). Pulse condition was a single 500 V, 3 ms square pulse. Immediately after electroporation, the cells were transferred to culture medium and cultured in the presence of 500 ng/mL of α-GalCer. The protein expression of transfected cells was analyzed by ELISA (ITEA) for OVA, flow cytometry for CD1d, and Western blot analysis for WT1 or TRP-2 protein as described previously (17).

Immunologic analyses

Antigen-specific IFNγ-secreting cells were performed by ELISPOT assay as described previously (24). The cytotoxic activity of NKT cell line was analyzed using LDH assay kit according to the manufacturer's instructions (Takara Bio Company). Expression of immune response–related molecules in DCs and tumors was analyzed by quantitative PCR assay. In the analysis of splenic DCs and tumor, the total RNA was isolated using RNeasy Mini Kit (Qiagen) and cDNA was synthesized by ReverTra Ace (Toyobo) according to the manufacturer's instructions. Tumor-associated DCs were sorted and directly subjected to cDNA synthesis using a CellsDirect One-Step qRT-PCR Kit (Invitrogen) with a mixture of pooled gene-specific primers (Supplementary Table S2). Synthesized cDNA was appropriately diluted with water and used as template for subsequent quantitative PCR as described previously (25).

Statistical analysis

The P values were calculated with the two-side Student t test or the Mann–Whitney U test. The log-rank test was used for survival calculations. P < 0.05 was considered statistically significant.

Generation of cytotoxic T cells and their infiltration into tumor sites by administration of α-GalCer-loaded, CD1d, and antigen mRNA–cotransfected cells

We previously established aAVCs, which are α-GalCer-loaded and antigen mRNA-transfected, CD1d-expressing allogeneic cells (NIH3T3 or HEK293 cells for mice and humans, respectively; ref. 17). In the current study, using a developed method by optimizing several points of the protocol to increase protein production (Supplementary Fig. S1A), we succeeded in the production of target antigen protein by aAVC at levels about 100 times higher than with the previous method. As shown in Supplementary Fig. S1B–S1D, we verified that aAVC-OVA cells, which were loaded with α-GalCer and cotransfected with OVA and CD1d mRNA, highly expressed the CD1d molecule and produced abundant OVA protein, and could directly stimulate NKT cells, but not OT-1 T cells in vitro (17). In the initial study, we assessed antitumor effects in a therapeutic model. We administered MO4 cells (OVA-expressing B16 melanoma) and, after verifying that large tumors (tumor size: 441.7 ± 176.8 mm3) were established, mice were treated with aAVC-OVA. As shown in Fig. 1A, untreated tumors grew larger; however, they stopped growing after the aAVC-OVA treatment and the center of tumor mass became necrotic. Given this striking immunotherapeutic outcome, we began to analyze the antitumor effects in more detail. This initial study thus demonstrated that treatment with aAVC-OVA increased the frequency of tumor-infiltrating T cell, especially CD8+ T cells in spleen and the tumor (Fig. 1B and Supplementary Fig. S2).

Figure 1.

Infiltration of cytotoxic T cells into tumor sites after immunization with artificial adjuvant vector cells. A, C57BL/6 mice were injected with 5 × 105 MO4 cells subcutaneously and then treated with or without 5 × 105 aAVC-OVA at day 12. Tumor growth was monitored at indicated time points by measuring three perpendicular diameters as described previously (ref. 17; bottom). Images are representative of pre (d12)- and post-aAVC-OVA treatment (d22; top); mean ± SEM, n = 16/group; ***, P < 0.001. B, C57BL/6 mice were injected with 2 × 105 MO4 cells subcutaneously and then treated with or without 5 × 105 aAVC-OVA at day 7. The infiltration of CD8+ T cells into the tumor was analyzed by IHC. Scale bar, 100 μm. Left, CD8+ T-cell numbers/mm2 are shown; right, mean ± SEM, n = 6; ***, P < 0.001. C, activation of STAT transcription factors was evaluated by intracellular staining of the phosphorylated active forms of CD8a+ and CD8a subsets of DCs in the spleen by Phospho Flow at 5 hours after immunization with aAVCs. (n = 4). D, the cross-presentation activity of OVA by in vivo DC was tested. WT mice that had been transferred with CFSE-labeled OT-1 cells were administered aAVC-OVA. Cell division was analyzed 3 days later. In some studies, CD11c-DTR and XCR1-DTR mice were used together with DT as recipients. (n = 4). E, expression of each mRNA in purified CD8a+ DCs was determined by quantitative real-time PCR using the primer sets shown in Supplementary Table S2 and is depicted as the number of transcripts per one copy of the housekeeping gene HPRT (mean ± SEM, n = 5); *, P < 0.05; **, P < 0.01, 0 hour vs. 6 or 12 hours.

Figure 1.

Infiltration of cytotoxic T cells into tumor sites after immunization with artificial adjuvant vector cells. A, C57BL/6 mice were injected with 5 × 105 MO4 cells subcutaneously and then treated with or without 5 × 105 aAVC-OVA at day 12. Tumor growth was monitored at indicated time points by measuring three perpendicular diameters as described previously (ref. 17; bottom). Images are representative of pre (d12)- and post-aAVC-OVA treatment (d22; top); mean ± SEM, n = 16/group; ***, P < 0.001. B, C57BL/6 mice were injected with 2 × 105 MO4 cells subcutaneously and then treated with or without 5 × 105 aAVC-OVA at day 7. The infiltration of CD8+ T cells into the tumor was analyzed by IHC. Scale bar, 100 μm. Left, CD8+ T-cell numbers/mm2 are shown; right, mean ± SEM, n = 6; ***, P < 0.001. C, activation of STAT transcription factors was evaluated by intracellular staining of the phosphorylated active forms of CD8a+ and CD8a subsets of DCs in the spleen by Phospho Flow at 5 hours after immunization with aAVCs. (n = 4). D, the cross-presentation activity of OVA by in vivo DC was tested. WT mice that had been transferred with CFSE-labeled OT-1 cells were administered aAVC-OVA. Cell division was analyzed 3 days later. In some studies, CD11c-DTR and XCR1-DTR mice were used together with DT as recipients. (n = 4). E, expression of each mRNA in purified CD8a+ DCs was determined by quantitative real-time PCR using the primer sets shown in Supplementary Table S2 and is depicted as the number of transcripts per one copy of the housekeeping gene HPRT (mean ± SEM, n = 5); *, P < 0.05; **, P < 0.01, 0 hour vs. 6 or 12 hours.

Close modal

Prominent function of XCR1+ DCs after administration of aAVC-OVA

We and others previously demonstrated that in vivo DCs stimulated by activated NKT cells produce IL12 and certain chemokines for inducing adaptive immunity (9, 10, 15, 24). In the current study, we analyzed the DCs in vivo in more detail after administration of aAVC. Both CD8a+ and CD8a subsets of DCs in the spleen highly expressed CD40, CD80, and CD86 16 hours after aAVC treatment, whereas 40 hours later, CD8a+ DCs also highly expressed crucial molecules for the generation of memory T-cell response, such as CD70, IL15Ra, and 4-1BBL (Supplementary Fig. S3A). In addition, phosphorylation of both STAT1, the signal transducer for type I and II IFNs, and STAT5, the signal transducer for IL2, IL7, IL15, and GM-CSF, was observed in both subsets of DCs 5 hours later (Fig. 1C). DCs could also be seen to have taken up debris of aAVC and to undergo maturation after capturing aAVC (Supplementary Fig. S3B). XCR1+ DCs is the relevant of CD8a+ subset of CD11c+ splenic DCs and are responsible for cross-presentation of exogenous antigen (22). We confirmed that XCR1 is exclusively expressed among CD8a+CD11c+MHCII+ DCs in the spleen and CD8a+ CD11bCD11c+MHCII+ DCs (resident DCs) and CD103+CD11bCD11c+MHCII+ DCs (migratory DCs) in lymph node (Supplementary Fig. S4A and S4C). In fact, XCR1+ DCs are specifically depleted by diphtheria toxin (DT) treatment (Supplementary Fig. S4B and S4D). To test the cross-presenting activity of the DCs in aAVC-immunized mice, we used WT, CD11c-DTR, and XCR1-DTR mice. These mice were transferred with CFSE-labeled OT-1 T cells and then injected with aAVC-OVA on the following day. Three days later, the OT-1 cells showed significant proliferation in WT mice, but not in CD11c-DTR or XCR-DTR mice treated with DT (Fig. 1D). These findings indicated that the CD8a+ DCs that had engulfed aAVC-OVA were activated and able to cross-present OVA peptide to CTLs. Next, we sorted the CD8a+ DCs and evaluated their expression of chemokines, cytokines, and genes involved in antigen processing, molecules that would play a key role in antigen presentation. We detected significantly elevated expression of CCL17, CCL22, IL12a, IL15, IL27, TAP1, TAP2, and PA28, a component of the immunoproteasome (Fig. 1E). Taken together, aAVC therapy can deliver tumor antigens to in vivo DC.

The TLS were formed by aAVC

Because the aAVC therapy induced tumor regression, we next focused on the tumor microenvironment. The expression of CXCL9, CXCL10, and CXCL11 were upregulated in tumors of aAVC-treated, but not untreated mice, as assessed by real-time PCR (Fig. 2A). Interestingly, CXCL10 was apparently expressed on DCs in tumor site (Fig. 2B). We verified it at protein level by IHC (Fig. 2C). Next, to focus on the expression of CXCR3 that are receptors for CXCL10, we detected downregulation of CXCR3 on OVA-tetramer+CD8+ T cells in tumor but not in spleen (Fig. 2D), suggesting that the activation of this receptor. We further analyzed CD8+ T cells and DCs and observed that many CD8+ T cells were located close to the CD31+ tumor vessels along with CD11c+ DCs in the aAVC-injected mice but not in untreated mice (Figs. 1B and 2E). In addition, the blood vessels in the tumors of the treated mice were CD31+VCAM-1+ICAM-1+ (Fig. 2F), while those in the untreated mice did not express these markers. It has been reported that ICAM-1 and VCAM-1 expression is essential for normalization of tumor vasculature, an effect should be important for vaccine efficacy (26, 27). Thus, the closely aggregated structures composed of CD11c+ DCs and antigen-specific CD8+ T cells around the CD31+VCAM-1+ICAM-1+ vessels seen in tumor sites resembled previously described TLS.

Figure 2.

Analyses of TLS formed in the tumor after immunization with aAVC. C57BL/6 mice were injected with 2 × 105 MO4 cells subcutaneously and then treated with or without 5 × 105 aAVC-OVA at day 7. A–C, expression of CXCL9, 10, and 11 in the tumor site (A) and in DCs (CD11c+ ClassII+F4/80 cells) sorted from the tumors at day 8 (B) was assessed by real-time PCR (mean ± SEM, n = 6; *, P < 0.05; Student t test, **, P < 0.005). C, the expression of CXCL10 at the protein level was assessed by IHC (n = 6). Scale bar, 100 μm. D, CXCR3 expression on OVA-tetramer+CD8+ T cells were assessed in the spleen (top) and TIL (bottom) at day 14 by flow cytometry. (n = 6). E, at day 14–17, CD8+ T cells, vessels, and CD11c+ DCs in the tumors were assessed by immunofluorescent staining using CD8 (red), CD31 (blue), and CD11c (green). CD11c+ cell numbers/mm2 are shown (mean ± SEM, n = 6). Scale bar, 100 μm. F, the phenotype of CD31+ vessels in tumor site was also analyzed using CD31 (green), VCAM-1 or ICAM-1 (red), and DAPI (n = 6). Scale bar, 100 μm.

Figure 2.

Analyses of TLS formed in the tumor after immunization with aAVC. C57BL/6 mice were injected with 2 × 105 MO4 cells subcutaneously and then treated with or without 5 × 105 aAVC-OVA at day 7. A–C, expression of CXCL9, 10, and 11 in the tumor site (A) and in DCs (CD11c+ ClassII+F4/80 cells) sorted from the tumors at day 8 (B) was assessed by real-time PCR (mean ± SEM, n = 6; *, P < 0.05; Student t test, **, P < 0.005). C, the expression of CXCL10 at the protein level was assessed by IHC (n = 6). Scale bar, 100 μm. D, CXCR3 expression on OVA-tetramer+CD8+ T cells were assessed in the spleen (top) and TIL (bottom) at day 14 by flow cytometry. (n = 6). E, at day 14–17, CD8+ T cells, vessels, and CD11c+ DCs in the tumors were assessed by immunofluorescent staining using CD8 (red), CD31 (blue), and CD11c (green). CD11c+ cell numbers/mm2 are shown (mean ± SEM, n = 6). Scale bar, 100 μm. F, the phenotype of CD31+ vessels in tumor site was also analyzed using CD31 (green), VCAM-1 or ICAM-1 (red), and DAPI (n = 6). Scale bar, 100 μm.

Close modal

TCRVβ repertoire of tumor-infiltrating CD8+ T cells after treatment with aAVC

We analyzed antigen-specific CD8+ T cells in spleen and tumor sites in tumor-bearing aAVC-treated or untreated mice. As shown in Fig. 3A and B, the frequency of OVA-specific CD8+ T cells in the tumor was higher than in the spleen (30% vs. 7% respectively) in aAVC-OVA–treated mice. The OVA-tetramer+CD8+ T cells in the tumor from aAVC-OVA–treated mice were not only increased in number but also had better function, as measured by expression of IFNγ and CD107 as a degranulation marker in response to OVA antigen (Fig. 3C). These results suggest that the CTLs were either primed by the DCs in lymphoid tissues and subsequently recruited into the tumor, or by the tumor-associated DCs in situ. To better understand the nature of the T-cell response to tumor antigen (OVA), we assessed the TCR repertoire of the OVA-tetramer+CD8+ T cells of spleen and tumor-infiltrating lymphocytes (TIL) from three tumor-bearing, aAVC-OVA–treated mice (Fig. 3D; Supplementary Table S3). On the basis of the results of deep sequencing, the TCRβ CDR3 repertoire in each of three groups of mice was different, even though all of the cells analyzed were OVA-tetramer+CD8+ T. For example, the sequence of CDR3 in cls-1 in spleen 1 was different from those of cls-1 in spleen 2 or 3 (Supplementary Table S3). On the other hand, when we compared the TCRβ CDR3 repertoire in the spleen and tumor in the same mouse, it was coincident (Supplementary Table S3). Thus, these TCR repertoire analyses indicate that the dominant OVA-tetramer+CD8+ T clones that expanded were different in each mouse, but were the same in spleen and tumor of individual mice. CTLs could first be primed by DCs in lymphoid tissues and then subsequently recruited to the tumor sites and further expanded by tumor-associated DCs.

Figure 3.

Analyses of TCR repertoire of cytotoxic T cells in the spleen and tumor. MO4 melanoma-bearing mice were treated with or without aAVC-OVA at day7. At day 14, the frequency and function and TCR repertoire of OVA-specific T cells were assessed. A and B, the frequency of Ag-specific CD8+ T cells in the spleen and tumor (gating on CD45+CD3+) in the untreated or aAVC-OVA–treated, tumor-bearing mice was analyzed using OVA-tetramer (mean ± SEM, n = 5–7). **, P < 0.01. C, the function of antigen-specific OVA-tetramer+CD8+ T cells in the tumor site from untreated and aAVC-OVA–treated mice was assessed by restimulation with OVA257–264 peptide and staining with IFNγ-APC and CD107a–FITC gating on CD45.1+CD8+ cells (n = 5). D, the T-cell repertoire of OVA-tetramer+CD8+ T cells in spleen and TIL was assessed in tumor-bearing, aAVC-OVA–treated mice. OVA-tetramer+CD8+ T cells in spleen and tumor site in individual mice (n = 3) were sorted and analyzed. The left and right panels indicated the frequencies of top 5 cluster reads (csl-1–5) of TCRβ in spleen (left) and TIL (right), respectively. The CDR3 sequences of each cluster are shown in Supplementary Table S3.

Figure 3.

Analyses of TCR repertoire of cytotoxic T cells in the spleen and tumor. MO4 melanoma-bearing mice were treated with or without aAVC-OVA at day7. At day 14, the frequency and function and TCR repertoire of OVA-specific T cells were assessed. A and B, the frequency of Ag-specific CD8+ T cells in the spleen and tumor (gating on CD45+CD3+) in the untreated or aAVC-OVA–treated, tumor-bearing mice was analyzed using OVA-tetramer (mean ± SEM, n = 5–7). **, P < 0.01. C, the function of antigen-specific OVA-tetramer+CD8+ T cells in the tumor site from untreated and aAVC-OVA–treated mice was assessed by restimulation with OVA257–264 peptide and staining with IFNγ-APC and CD107a–FITC gating on CD45.1+CD8+ cells (n = 5). D, the T-cell repertoire of OVA-tetramer+CD8+ T cells in spleen and TIL was assessed in tumor-bearing, aAVC-OVA–treated mice. OVA-tetramer+CD8+ T cells in spleen and tumor site in individual mice (n = 3) were sorted and analyzed. The left and right panels indicated the frequencies of top 5 cluster reads (csl-1–5) of TCRβ in spleen (left) and TIL (right), respectively. The CDR3 sequences of each cluster are shown in Supplementary Table S3.

Close modal

Potent synergistic antitumor effect by the combination therapy of aAVC with anti-PD-1 mAb

It has been noted in several studies that immunosuppressive molecules are often expressed on TIL (28) and inhibit their function. When TILs in untreated tumor-bearing mice were examined, we detected very few OVA-tetramer+CD8+ T cells, too few to analyze, but the OVA-tetramer CD8+ T cells expressed relatively high levels of PD-1, and Lag3 (Fig. 4A, bottom). In contrast, OVA-tetramer+CD8+ T cells in aAVC-OVA–treated mice were plentiful and they expressed low levels of PD-1 and Lag3 (Fig. 4A upper).

Figure 4.

Synergistic immune responses by vaccination of aAVC together with anti-PD-1 mAb in tumor-bearing mice. A, the expression of immune suppressive molecules (PD-1, Lag3, and TIM3) on OVA-tetramer+CD8+ T or tetramer CD8+ T cells among the TIL was assessed by flow cytometry. (n = 5/group). B, as in Fig. 1B, but mice were treated with aAVC-OVA (5 × 105 cells/mouse) or anti-PD-1 mAb (250 μg/mouse) or a combination of the two at day 12. Tumor size was evaluated at the indicated time points in each group (mean ± SEM, n = 5–10). **, P < 0.01. C, as in B, but mice were treated with low dose aAVC-OVA (1 × 104 cells/mouse) with or without anti-PD-1 mAb at day 12. Tumor size was evaluated at the indicated time points in each group (mean ± SEM, n = 10–12). ***, P < 0.005. D, the frequency of OVA-tetramer+CD8+ T cells in spleen and TIL. Mice inoculated with 2 × 105 MO4 were treated with aAVC-OVA or anti-PD-1 mAb or a combination of the two at day 7, then the frequency of OVA-tetramer+CD8+ T cells in spleen and TIL was analyzed at day 20–21 (mean ± SEM, n = 5/group). *P < 0.05 (two-tailed Student t test).

Figure 4.

Synergistic immune responses by vaccination of aAVC together with anti-PD-1 mAb in tumor-bearing mice. A, the expression of immune suppressive molecules (PD-1, Lag3, and TIM3) on OVA-tetramer+CD8+ T or tetramer CD8+ T cells among the TIL was assessed by flow cytometry. (n = 5/group). B, as in Fig. 1B, but mice were treated with aAVC-OVA (5 × 105 cells/mouse) or anti-PD-1 mAb (250 μg/mouse) or a combination of the two at day 12. Tumor size was evaluated at the indicated time points in each group (mean ± SEM, n = 5–10). **, P < 0.01. C, as in B, but mice were treated with low dose aAVC-OVA (1 × 104 cells/mouse) with or without anti-PD-1 mAb at day 12. Tumor size was evaluated at the indicated time points in each group (mean ± SEM, n = 10–12). ***, P < 0.005. D, the frequency of OVA-tetramer+CD8+ T cells in spleen and TIL. Mice inoculated with 2 × 105 MO4 were treated with aAVC-OVA or anti-PD-1 mAb or a combination of the two at day 7, then the frequency of OVA-tetramer+CD8+ T cells in spleen and TIL was analyzed at day 20–21 (mean ± SEM, n = 5/group). *P < 0.05 (two-tailed Student t test).

Close modal

Immunotherapy with anti-PD-1 or PD-L1 mAb has shown clinical efficacy with some tumor types (29). We therefore assessed whether there would be any synergistic effect with combined aAVC-OVA and anti-PD-1 mAb therapy but none was observed (Fig. 4B), possibly because the aAVC-OVA therapy was already so effective. We thus reduced the number of aAVC-OVA to 1/50, that is, 1 × 104 cells/mouse, thinking that the antitumor efficacy of the combination of aAVC-OVA and anti-PD-1 mAb would be more apparent under this suboptimal condition. Indeed, T-cell responses in these vaccinated mice were decreased by about 75% (the frequency of OVA-tetramer+CD8+ T cells in total spleen was % 2.12 ± 0.61 with the normal aAVC-OVA dose vs. % 0.61 ± 0.10 with the lower dose; Supplementary Fig. S5). Although the antitumor effect with aAVC therapy alone was also weakened, we detected a potent synergistic effect with the aAVC-OVA plus anti-PD-1 mAb combination (Fig. 4C). There was no increase in the number of T cells after administration of the anti-PD-1 mAb alone; however, the combination of anti-PD-1 mAb and aAVC-OVA therapy led to an increased frequency of OVA-tetramer+CD8+ T cells in the tumor as well as in spleen (Fig. 4D). Therefore, this combination therapy would show a therapeutic advantage.

Prime and boosting effect on memory T cells

Memory T cells are readily generated after an acute infection; however, it has been difficult to generate memory T cells against cancer cell–associated antigens. If any, most of the studies have had some success by using the transfer of transgenic tumor-specific T cells, but not under physiologic conditions. Therefore, we assessed the induction of memory T cells following aAVC-OVA therapy, without transferring transgenic T cells. As shown in Fig. 5A (top), we detected memory T cells systemically, not only in lymphoid tissues but also in non-lymphoid tissues one year after immunization with aAVC-OVA. Further analysis showed that CD44hiCD62Lhi central memory T cells (TCM) are predominant in lymphoid tissues, that is, spleen and lymph nodes, whereas in the lung the TCM and CD44hiCD62Llo T effector memory (TEM) cells were equivalent. On the other hand, TEM cells were predominant in the bone marrow (Fig. 5A lower) and liver (not shown). We then assessed the function of the memory T cells in the spleen and bone marrow. They produced IFNγ and TNFα after a stimulation with OVA peptide (Fig. 5B and C), suggesting that multifunctional memory T cells were induced by aAVC-OVA therapy.

Figure 5.

The prime and boost effect of memory T cells by aAVC vaccine. A, C57BL/6 mice were immunized with aAVC-OVA. Twelve months later, the frequency (top) and phenotype (bottom) of OVA-tetramer+CD8+ T cells were evaluated in the indicated organs/tissue. The expression of CD44 and CD62L is shown in flow cytometric contour plots gated on OVA-tetramer+CD8+ T cells. Numbers are shown as means ± SEM (n = 9). B and C, the function of the memory T cells was assessed by stimulating with antigen for production of IFNγ and TNFα 12 months after immunization. Spleen cells or BMMNCs were stimulated for 6 hours with OVA257–264 peptide, and then stained for cytokine production using the combination of TNFα-PE and IFNγ-APC gating on CD8+CD44hi T cells (mean ± SEM; n = 3). D and E, mice that had been vaccinated with aAVC-OVA 6 months previously were boosted with aAVC-OVA (homologous boost) or OVA peptide-pulsed DCs (heterologous boost) and the frequency (D) and cell number (E) of OVA-tetramer+CD8+ T cells were analyzed 5 days later (mean ± SEM, n = 4–5). *, P < 0.05. BM, bone marrow; LN, lymph node.

Figure 5.

The prime and boost effect of memory T cells by aAVC vaccine. A, C57BL/6 mice were immunized with aAVC-OVA. Twelve months later, the frequency (top) and phenotype (bottom) of OVA-tetramer+CD8+ T cells were evaluated in the indicated organs/tissue. The expression of CD44 and CD62L is shown in flow cytometric contour plots gated on OVA-tetramer+CD8+ T cells. Numbers are shown as means ± SEM (n = 9). B and C, the function of the memory T cells was assessed by stimulating with antigen for production of IFNγ and TNFα 12 months after immunization. Spleen cells or BMMNCs were stimulated for 6 hours with OVA257–264 peptide, and then stained for cytokine production using the combination of TNFα-PE and IFNγ-APC gating on CD8+CD44hi T cells (mean ± SEM; n = 3). D and E, mice that had been vaccinated with aAVC-OVA 6 months previously were boosted with aAVC-OVA (homologous boost) or OVA peptide-pulsed DCs (heterologous boost) and the frequency (D) and cell number (E) of OVA-tetramer+CD8+ T cells were analyzed 5 days later (mean ± SEM, n = 4–5). *, P < 0.05. BM, bone marrow; LN, lymph node.

Close modal

Next, we examined whether the long-lived memory T cells could be boosted. If the response to aAVC-OVA was predominantly mediated by allogeneic T cells reacting with NIH3T3 cells, then it likely could not be boosted. However, when we administered aAVC-OVA to mice that had been vaccinated with aAVC-OVA as a homologous prime-boost strategy 6 months previously, a strong secondary response was elicited (Fig. 5D, middle and E). Moreover, peptide-pulsed DCs could also induce a robust T-cell response as a heterologous prime-boost setting (Fig. 5D lower and E). Thus, apparently memory T cells were induced by aAVC-OVA therapy.

Therapeutic effects against melanoma by administration of aAVC-TRP-2

OVA-expressing tumor cells have been a very useful model to study tumor immunity, however, OVA is an artificial tumor antigen. To evaluate the T-cell response to an authentic tumor antigen, we established aAVC expressing TRP-2 (aAVC-TRP-2), a melanoma tumor antigen, instead of OVA, and then assessed the antitumor and T-cell responses. We analyzed the T-cell response by IFNγ ELISPOT assay 7 days after administration of aAVC-TRP-2. As shown in Fig. 6A, the TRP-2–specific T-cell response was characterized by antigen-specific IFNγ production. We then assessed the antitumor response in a therapeutic model. B16 melanoma cells were allowed to grow subcutaneously for 7 days and then the mice were treated with aAVC-TRP-2 (Fig. 6B). Compared with untreated mice, the aAVC-TRP-2–treated mice showed significant protection against the melanoma.

Figure 6.

Antigen-specific T-cell responses and the antitumor effects were also induced in other mouse tumor models. A, C57BL/6 mice were immunized with 5 × 105 hTRP-2 tumor antigen-expressing aAVC (aAVC-TRP-2) and a week later the TRP-2–specific T-cell response was assessed by ELISPOT analysis of IFNγ secretion. CD8+ T cells (5 × 105 cells/well) from spleens of untreated or treated mice were cocultured with hTRP-2 mRNA–transfected bone marrow–derived DCs (5 × 104 cells/well) for 36 hours (n = 5). *, P < 0.05. B, C57BL/6 mice were injected with 1 × 105 B16 melanoma cells subcutaneously and then treated with aAVC-TRP-2 7 days later. Tumor size was evaluated at the indicated time points. Images are representative of control (left) and aAVC-OVA–treated mice (right; n = 10–14 per group). ***, P < 0.001. C, BALB/c mice were immunized with 5 × 105 WT1-expressing aAVC (aAVC-WT1) and a week later the WT1 specific T-cell response was assessed by IFNγ ELISPOT (mean ± SEM, n = 8). *, P < 0.01. D, mice were inoculated with 1 × 106 J558 (left) or WT1-expressing J558 (middle) or WEHI 3B cells (right), treated with aAVC-WT1 at day 7, and then evaluated for survival (n = 15–30/group). ***, P < 0.001 (log-rank test). E, three to 6 months later, the surviving mice from (D, left) were rechallenged with J558-WT1 cells or irrelevant WEHI 3B cells and evaluated for their survival (n = 9–10/group). ***, P < 0.001 (log-rank test).

Figure 6.

Antigen-specific T-cell responses and the antitumor effects were also induced in other mouse tumor models. A, C57BL/6 mice were immunized with 5 × 105 hTRP-2 tumor antigen-expressing aAVC (aAVC-TRP-2) and a week later the TRP-2–specific T-cell response was assessed by ELISPOT analysis of IFNγ secretion. CD8+ T cells (5 × 105 cells/well) from spleens of untreated or treated mice were cocultured with hTRP-2 mRNA–transfected bone marrow–derived DCs (5 × 104 cells/well) for 36 hours (n = 5). *, P < 0.05. B, C57BL/6 mice were injected with 1 × 105 B16 melanoma cells subcutaneously and then treated with aAVC-TRP-2 7 days later. Tumor size was evaluated at the indicated time points. Images are representative of control (left) and aAVC-OVA–treated mice (right; n = 10–14 per group). ***, P < 0.001. C, BALB/c mice were immunized with 5 × 105 WT1-expressing aAVC (aAVC-WT1) and a week later the WT1 specific T-cell response was assessed by IFNγ ELISPOT (mean ± SEM, n = 8). *, P < 0.01. D, mice were inoculated with 1 × 106 J558 (left) or WT1-expressing J558 (middle) or WEHI 3B cells (right), treated with aAVC-WT1 at day 7, and then evaluated for survival (n = 15–30/group). ***, P < 0.001 (log-rank test). E, three to 6 months later, the surviving mice from (D, left) were rechallenged with J558-WT1 cells or irrelevant WEHI 3B cells and evaluated for their survival (n = 9–10/group). ***, P < 0.001 (log-rank test).

Close modal

Therapeutic effects targeting the WT1 antigen by administration of aAVC-WT1

Wilms' tumor gene WT1 encodes a protein that has been reported to be a tumor antigen in a variety of malignancies (30, 31). To evaluate whether WT1 mRNA could be used in our vaccine models, we established α-GalCer-loaded, mCD1d mRNA, and WT1 mRNA cotransfected NIH3T3 cells, denoted mouse aAVC-WT1. As aAVC-WT1 vaccine efficacy, we confirmed WT1-specific T-cell responses by IFNγ ELISPOT assay 7 days after administration of aAVC-WT1 (Fig. 6C). Next, we injected mice with a WT1-expressing plasmacytoma cell line (J558-WT1) and then treated them with aAVC-WT1 at day 7. In the control untreated group, all of the mice died within 30 days after inoculation with tumor cells. In contrast, 75% of the aAVC-WT1–treated mice survived more than 3 months (Fig. 6D, middle). However, aAVC-WT1 therapy did not protect mice from the parental J558 or the WEHI3B leukemia cell line (Fig. 6D, left and right), demonstrating specificity. These results clearly indicated that the WT1-specific antitumor effect was generated by aAVC-WT1 treatment. When the surviving mice were rechallenged with the J558-WT1 cells more than 4 months later, all of them were protected (Fig. 6E), but all of them died when injected with irrelevant WEHI3B cells, demonstrating the specificity of the protection (Fig. 6E). The data suggest that aAVC therapy can induce a tumor antigen–specific memory response.

Next, we asked whether human NKT cells can respond to human aAVC-WT1. For this purpose, we established α-GalCer–loaded, hCD1d mRNA and WT1 mRNA–cotransfected HEK293 cells as human aAVC (denoted human aAVC-WT1). At first, we verified that CD1d and WT1 are expressed by HEK293 cells using flow cytometry and Western blot analysis, respectively (Fig. 7A and B). Human aAVC-WT1 contained 178.2 ± 41.3 μg WT1 protein/1 × 106 cells. Human NKT cells recognized and killed α-GalCer–loaded, CD1d-expressing HEK293 but not unloaded CD1d-HEK293 (Fig. 7C). Then, we examined whether NKT and T-cell responses could be induced after coculturing PBMCs and human aAVC-WT1 loaded with or without α-GalCer. As shown in Fig. 7D, NKT cells expressed IFNγ after a 6-hour culture with human aAVC-WT1, whereas they did not produce IFNγ in response to unmanipulated HEK293 cells (Fig. 7D). Also, when we cultured HLA-A24+ PBMCs derived from healthy volunteers with human aAVC-WT1, WT1-tetramer+ T cells were generated, but no T cells specific for the control HIV peptide were observed (Fig. 7E). The human aAVC-WT1 were more effective at generating WT1-tetramer+ CD8+ T cells than culture of PBMCs in the presence of WTI peptide and IL2. Some human DCs and monocytes as DC precursor express HLA-DR and/or CD14 (32). To confirm the importance of DCs in this system, we depleted CD14+ and DR+ cells before the coculture, and no WT1-tetramer+ CD8+ T cells were generated (Fig. 7F). Thus, aAVC-WT1 can generate human WT1-specific T cells, and this process is dependent on antigen-presenting cells, mainly DCs.

Figure 7.

Preclinical study targeting WT1 antigen by treatment with human aAVC-WT1 in vitro. Human artificial adjuvant vector cells expressing WT1 (aAVC-WT1) were established using HEK293 by loading with α-GalCer, and coelectroporation with WT1 and CD1d mRNA. Eight hours after electroporation, CD1d expression and WT1 protein production were assessed by flow cytometry (A) and Western Blotting analysis (B), respectively. C, NKT cell killing activity against CD1d-HEK293 or α-GalCer–loaded CD1d-HEK293 (CD1d-HEK293/Gal) was examined. Target cells (1 × 104 cell/well) were cultured with effector cells at the indicated effector:target (E:T) ratio for 12 hours (n = 4). D, for the primary NKT cell response, 1 × 105 human PBMCs were cultured alone or cocultured with 30 Gy–irradiated 1 × 104 aAVC-WTI or CD1d-HEK293 in the presence of GolgiPlug for 6 hours. Intracellular IFNγ staining was performed. Data are representative of 7 healthy volunteers (HV) and gating on CD3+6B11+ cells (n = 7). E, to assess the T-cell response, HLA-A24+PBMCs were cultured with irradiated aAVC-WTI or WT1 peptide in the presence of IL2 for 7 days. WT1-specific CD8+ T cells were evaluated using HLA-A24/WT1-tetramer-PE/CD8-APC compared with HIV-tetramer-PE/CD8-APC as a negative control. Flow cytometry data (i) are representative of five HVs and the frequencies of WT1-tetramer+CD8+ T cells were summarized (ii; n = 5). F, as in E, but before coculturing, CD14+ cells and HLA-DR+ cells were depleted with magnetic beads. One week later, WT1-specific CD8+ T cells were evaluated WT1-tet-PE/CD8-APC compared with HIV-tetramer-PE/CD8-APC as a negative control (n = 3).

Figure 7.

Preclinical study targeting WT1 antigen by treatment with human aAVC-WT1 in vitro. Human artificial adjuvant vector cells expressing WT1 (aAVC-WT1) were established using HEK293 by loading with α-GalCer, and coelectroporation with WT1 and CD1d mRNA. Eight hours after electroporation, CD1d expression and WT1 protein production were assessed by flow cytometry (A) and Western Blotting analysis (B), respectively. C, NKT cell killing activity against CD1d-HEK293 or α-GalCer–loaded CD1d-HEK293 (CD1d-HEK293/Gal) was examined. Target cells (1 × 104 cell/well) were cultured with effector cells at the indicated effector:target (E:T) ratio for 12 hours (n = 4). D, for the primary NKT cell response, 1 × 105 human PBMCs were cultured alone or cocultured with 30 Gy–irradiated 1 × 104 aAVC-WTI or CD1d-HEK293 in the presence of GolgiPlug for 6 hours. Intracellular IFNγ staining was performed. Data are representative of 7 healthy volunteers (HV) and gating on CD3+6B11+ cells (n = 7). E, to assess the T-cell response, HLA-A24+PBMCs were cultured with irradiated aAVC-WTI or WT1 peptide in the presence of IL2 for 7 days. WT1-specific CD8+ T cells were evaluated using HLA-A24/WT1-tetramer-PE/CD8-APC compared with HIV-tetramer-PE/CD8-APC as a negative control. Flow cytometry data (i) are representative of five HVs and the frequencies of WT1-tetramer+CD8+ T cells were summarized (ii; n = 5). F, as in E, but before coculturing, CD14+ cells and HLA-DR+ cells were depleted with magnetic beads. One week later, WT1-specific CD8+ T cells were evaluated WT1-tet-PE/CD8-APC compared with HIV-tetramer-PE/CD8-APC as a negative control (n = 3).

Close modal

In some types of human cancer, including melanoma (33), colorectal cancer (34), breast cancer (35) and lung cancer (36), a correlation between a low density of CD8+ T cells in the tumor and poor prognosis has been reported. These cases where there is minimal T-cell infiltration are termed non-T-cell–inflamed tumors (37) and therefore inefficient T-cell migration into the tumor appears to be a major limitation of cancer immunotherapy (27). We showed how aAVC can show better T-cell–mediated antitumor efficacy at the tumor microenvironment in an aggressive tumor model. As a mechanistic summary, in vivo DC activation can induce the formation of TLS in which DCs recruited from lymphoid tissues may induce the formation of ICAM-1- and VCAM-1–expressing blood vessels, resulting in normalization of the vasculature within the tumor. Subsequently, CXCR3-expressing CTLs are efficiently recruited to CXCR3 ligand-expressing DCs, resulting in the aggregation of CTLs in the tumor. We directly showed such a mechanism for aAVC therapy in this study. There are some fragmentary clinical reports describing a correlation between the existence of tumor-associated TLS in patient samples and clinical prognosis (8). Coexistence of effector T cells and a high density of mature DCs in TLS is correlated with longer term survival in lung cancer patients (38). The immunologic results obtained in murine studies with our vaccine were similar to those in the patients with a good clinical prognosis.

PD-1 blockade is a promising therapy because it can have a beneficial effect on approximately 30% of cancer patients (39); however, some types of cancer patients are refractory to this treatment. There are some reports accounting for the differences between the two patient groups, such as PD-L1 expression on tumors, IFNγ production, and preexisting T cells in the tumor sites (37, 39). Preexisting PD-1hiCD8+ T cells in the tumor site have been known to be dysfunctional, but PD-1loCD8+ T cells are capable of producing IFNγ and TNFα, that is functional (39, 40). In addition, PD-1loCD8+ T cells can easily be stimulated by anti-PD-1 mAb (40). Recently, many strategies have been explored that might be combined with the PD1 blockade therapy. In fact, we showed here that the MO4 melanoma was resistant to anti-PD-1 mAb alone, but that a combination therapy with low dose of aAVC therapy and anti-PD-1 mAb was effective. Infiltrating PD-1loCD8+ T cells in the tumor that were generated by aAVC treatment (Fig. 4A) is likely one reason for this successful outcome. Such an increase of antigen-specific T cells in the tumor by synergistic combination therapy might be useful for patients who are resistant to PD-1 blockade therapy.

Generation of memory T cells is one hallmark of vaccine success that may possibly control the long-term antitumor immune response. Criteria and quality of ideal T-cell memory following vaccination has been defined in many studies, typically in viral infection models, and include: (i) long-term maintenance, (ii) recall response to reencounter with the same pathogen, and (iii) multifunctionality, for example, in terms of cytokine production (41–43). In this study, we verified that the memory T cells induced by the aAVC vaccine completely met the above criteria for memory T cells.

Altogether, our findings with the current aAVC vaccine emphasize a novel role for DC activation in situ that shapes both local (i.e., within the tumor) and systemic (i.e., memory T cells) immune responses due to the linkage of innate and adaptive immunity. Further analysis of these local and systemic immune responses may give us important clues about how the antitumor immunologic network operates and should be helpful for developing the next generation of immunotherapy.

No potential conflicts of interest were disclosed.

Conception and design: K. Shimizu, S.-I. Fujii

Development of methodology: K. Shimizu, M. Asakura, S.-I. Fujii

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): K. Shimizu, S. Yamasaki, J. Shinga, Y. Sato, O. Ohara, H. Yagita, S.-I. Fujii

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): K. Shimizu, T. Watanabe, O. Ohara, M. Asakura, S.-I. Fujii

Writing, review, and/or revision of the manuscript: K. Shimizu, H. Yagita, S.-I. Fujii

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): K. Kuzushima, Y. Komuro

Study supervision: S.-I. Fujii

The authors thank Drs. P.D. Burrows (University of Alabama) and I. Mellman (Genentech) for critical reading of the manuscript and Ms. C. Oikawa, Ms. M. Sakurai, and Mr. M. Kawamura (RIKEN, IMS) for providing technical assistance.

This work is supported by JSPS KAKENHI grants (K. Shimizu and S. Fujii) and the Japan Agency for Medical Research and Development (Translational Research Network Program; S. Fujii).

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.
Chen
DS
,
Mellman
I
. 
Oncology meets immunology: the cancer-immunity cycle
.
Immunity
2013
;
39
:
1
10
.
2.
Robbins
PF
,
Morgan
RA
,
Feldman
SA
,
Yang
JC
,
Sherry
RM
,
Dudley
ME
, et al
Tumor regression in patients with metastatic synovial cell sarcoma and melanoma using genetically engineered lymphocytes reactive with NY-ESO-1
.
J Clin Oncol
2011
;
29
:
917
24
.
3.
Maus
MV
,
Fraietta
JA
,
Levine
BL
,
Kalos
M
,
Zhao
Y
,
June
CH
. 
Adoptive immunotherapy for cancer or viruses
.
Annu Rev Immunol
2014
;
32
:
189
225
.
4.
Postow
MA
,
Callahan
MK
,
Wolchok
JD
. 
Immune checkpoint blockade in cancer therapy
.
J Clin Oncol
2015
;
33
:
1974
82
.
5.
Steinman
RM
. 
Decisions about dendritic cells: past, present, and future
.
Annu Rev Immunol
2012
;
30
:
1
22
.
6.
Palucka
K
,
Banchereau
J
. 
Dendritic-cell-based therapeutic cancer vaccines
.
Immunity
2013
;
39
:
38
48
.
7.
Gajewski
TF
,
Schreiber
H
,
Fu
YX
. 
Innate and adaptive immune cells in the tumor microenvironment
.
Nat Immunol
2013
;
14
:
1014
22
.
8.
Dieu-Nosjean
MC
,
Goc
J
,
Giraldo
NA
,
Sautes-Fridman
C
,
Fridman
WH
. 
Tertiary lymphoid structures in cancer and beyond
.
Trends Immunol
2014
;
35
:
571
80
.
9.
Fujii
S
,
Liu
K
,
Smith
C
,
Bonito
AJ
,
Steinman
RM
. 
The linkage of innate to adaptive immunity via maturing dendritic cells in vivo requires CD40 ligation in addition to antigen presentation and CD80/86 costimulation
.
J Exp Med
2004
;
199
:
1607
18
.
10.
Fujii
S
,
Shimizu
K
,
Smith
C
,
Bonifaz
L
,
Steinman
RM
. 
Activation of natural killer T cells by α-galactosylceramide rapidly induces the full maturation of dendritic cells in vivo and thereby acts as an adjuvant for combined CD4 and CD8 T cell immunity to a co-administered protein
.
J Exp Med
2003
;
198
:
267
79
.
11.
Fujii
S
,
Shimizu
K
,
Hemmi
H
,
Steinman
RM
. 
Innate Vα14+ natural killer T cells mature dendritic cells, leading to strong adaptive immunity
.
Immunol Rev
2007
;
220
:
183
98
.
12.
Fujii
S
,
Shimizu
K
,
Okamoto
Y
,
Kunii
N
,
Nakayama
T
,
Motohashi
S
, et al
NKT cells as an ideal anti-tumor immunotherapeutic
.
Front Immunol
2013
;
4
:
409
.
13.
Faveeuw
C
,
Trottein
F
. 
Optimization of natural killer T cell-mediated immunotherapy in cancer using cell-based and nanovector vaccines
.
Cancer Res
2014
;
74
:
1632
8
.
14.
Gonzalez-Aseguinolaza
G
,
Van Kaer
L
,
Bergmann
CC
,
Wilson
JM
,
Schmieg
J
,
Kronenberg
M
, et al
Natural killer T cell ligand α-galactosylceramide enhances protective immunity induced by malaria vaccines
.
J Exp Med
2002
;
195
:
617
24
.
15.
Hermans
IF
,
Silk
JD
,
Gileadi
U
,
Salio
M
,
Mathew
B
,
Ritter
G
, et al
NKT cells enhance CD4+ and CD8+ T cell responses to soluble antigen in vivo through direct interaction with dendritic cells
.
J Immunol
2003
;
171
:
5140
47
.
16.
Fujii
S
,
Goto
A
,
Shimizu
K
. 
Antigen mRNA-transfected, allogeneic fibroblasts loaded with NKT-cell ligand confer antitumor immunity
.
Blood
2009
;
113
:
4262
72
.
17.
Shimizu
K
,
Mizuno
T
,
Shinga
J
,
Asakura
M
,
Kakimi
K
,
Ishii
Y
, et al
Vaccination with antigen-transfected, NKT cell ligand-loaded, human cells elicits robust in situ immune responses by dendritic cells
.
Cancer Res
2013
;
73
:
62
73
.
18.
Agata
Y
,
Kawasaki
A
,
Nishimura
H
,
Ishida
Y
,
Tsubata
T
,
Yagita
H
, et al
Expression of the PD-1 antigen on the surface of stimulated mouse T and B lymphocytes
.
Int Immunol
1996
;
8
:
765
72
.
19.
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
.
20.
Guilloux
Y
,
Bai
XF
,
Liu
X
,
Zheng
P
,
Liu
Y
. 
Optimal induction of effector but not memory antitumor cytotoxic T lymphocytes involves direct antigen presentation by the tumor cells
.
Cancer Res
2001
;
61
:
1107
12
.
21.
Jung
S
,
Unutmaz
D
,
Wong
P
,
Sano
G
,
De los Santos
K
,
Sparwasser
T
, et al
In vivo depletion of CD11c+ dendritic cells abrogates priming of CD8+ T cells by exogenous cell-associated antigens
.
Immunity
2002
;
17
:
211
20
.
22.
Yamazaki
C
,
Sugiyama
M
,
Ohta
T
,
Hemmi
H
,
Hamada
E
,
Sasaki
I
, et al
Critical roles of a dendritic cell subset expressing a chemokine receptor, XCR1
.
J Immunol
2013
;
190
:
6071
82
.
23.
Fujii
S
,
Shimizu
K
,
Kronenberg
M
,
Steinman
RM
. 
Prolonged interferon-γ producing NKT response induced with α-galactosylceramide-loaded dendritic cells
.
Nat Immunol
2002
;
3
:
867
74
.
24.
Shimizu
K
,
Kurosawa
Y
,
Taniguchi
M
,
Steinman
RM
,
Fujii
S
. 
Cross-presentation of glycolipid from tumor cells loaded with α-galactosylceramide leads to potent and long-lived T cell mediated immunity via dendritic cells
.
J Exp Med
2007
;
204
:
2641
53
.
25.
Shimizu
K
,
Sato
Y
,
Shinga
J
,
Watanabe
T
,
Endo
T
,
Asakura
M
, et al
KLRG+ invariant natural killer T cells are long-lived effectors
.
Proc Natl Acad Sci U S A
2014
;
111
:
12474
9
.
26.
Klug
F
,
Prakash
H
,
Huber
PE
,
Seibel
T
,
Bender
N
,
Halama
N
, et al
Low-dose irradiation programs macrophage differentiation to an iNOS(+)/M1 phenotype that orchestrates effective T cell immunotherapy
.
Cancer Cell
2013
;
24
:
589
602
.
27.
Oelkrug
C
,
Ramage
JM
. 
Enhancement of T cell recruitment and infiltration into tumours
.
Clin Exp Immunol
2014
;
178
:
1
8
.
28.
Gros
A
,
Robbins
PF
,
Yao
X
,
Li
YF
,
Turcotte
S
,
Tran
E
, et al
PD-1 identifies the patient-specific CD8(+) tumor-reactive repertoire infiltrating human tumors
.
J Clin Invest
2014
;
124
:
2246
59
.
29.
Taube
JM
,
Klein
A
,
Brahmer
JR
,
Xu
H
,
Pan
X
,
Kim
JH
, et al
Association of PD-1, PD-1 ligands, and other features of the tumor immune microenvironment with response to anti-PD-1 therapy
.
Clin Cancer Res
2014
;
20
:
5064
74
.
30.
Cheever
MA
,
Allison
JP
,
Ferris
AS
,
Finn
OJ
,
Hastings
BM
,
Hecht
TT
, et al
The prioritization of cancer antigens: a national cancer institute pilot project for the acceleration of translational research
.
Clin Cancer Res
2009
;
15
:
5323
37
.
31.
Huff
V
. 
Wilms' tumours: about tumour suppressor genes, an oncogene and a chameleon gene
.
Nat Rev Cancer
2011
;
11
:
111
21
.
32.
Schlitzer
A
,
Ginhoux
F
. 
Organization of the mouse and human DC network
.
Curr Opin Immunol
2014
;
26
:
90
9
.
33.
Azimi
F
,
Scolyer
RA
,
Rumcheva
P
,
Moncrieff
M
,
Murali
R
,
McCarthy
SW
, et al
Tumor-infiltrating lymphocyte grade is an independent predictor of sentinel lymph node status and survival in patients with cutaneous melanoma
.
J Clin Oncol
2012
;
30
:
2678
83
.
34.
Pages
F
,
Berger
A
,
Camus
M
,
Sanchez-Cabo
F
,
Costes
A
,
Molidor
R
, et al
Effector memory T cells, early metastasis, and survival in colorectal cancer
.
N Engl J Med
2005
;
353
:
2654
66
.
35.
Menard
S
,
Tomasic
G
,
Casalini
P
,
Balsari
A
,
Pilotti
S
,
Cascinelli
N
, et al
Lymphoid infiltration as a prognostic variable for early-onset breast carcinomas
.
Clin Cancer Res
1997
;
3
:
817
9
.
36.
Hiraoka
K
,
Miyamoto
M
,
Cho
Y
,
Suzuoki
M
,
Oshikiri
T
,
Nakakubo
Y
, et al
Concurrent infiltration by CD8+ T cells and CD4+ T cells is a favourable prognostic factor in non-small-cell lung carcinoma
.
Br J Cancer
2006
;
94
:
275
80
.
37.
Woo
SR
,
Corrales
L
,
Gajewski
TF
. 
The STING pathway and the T cell-inflamed tumor microenvironment
.
Trends Immunol
2015
;
36
:
250
6
.
38.
Goc
J
,
Germain
C
,
Vo-Bourgais
TK
,
Lupo
A
,
Klein
C
,
Knockaert
S
, et al
Dendritic cells in tumor-associated tertiary lymphoid structures signal a Th1 cytotoxic immune contexture and license the positive prognostic value of infiltrating CD8+ T cells
.
Cancer Res
2014
;
74
:
705
15
.
39.
Chen
L
,
Han
X
. 
Anti-PD-1/PD-L1 therapy of human cancer: past, present, and future
.
J Clin Invest
2015
;
125
:
3384
91
.
40.
Ngiow
SF
,
Young
A
,
Jacquelot
N
,
Yamazaki
T
,
Enot
D
,
Zitvogel
L
, et al
A threshold level of intratumor CD8+ T-cell PD1 expression dictates therapeutic response to anti-PD1
.
Cancer Res
2015
;
75
:
3800
11
.
41.
Seder
RA
,
Darrah
PA
,
Roederer
M
. 
T-cell quality in memory and protection: implications for vaccine design
.
Nat Rev Immunol
2008
;
8
:
247
58
.
42.
Sallusto
F
,
Lanzavecchia
A
,
Araki
K
,
Ahmed
R
. 
From vaccines to memory and back
.
Immunity
2010
;
33
:
451
63
.
43.
Pulendran
B
,
Ahmed
R
. 
Immunological mechanisms of vaccination
.
Nat Immunol
2011
;
12
:
509
17
.