Oncolytic reovirus, which possesses 10 segments of dsRNA genome, mediates antitumor effects via not only virus replication in a tumor cell–specific manner, but also activation of antitumor immunity; however, the mechanism(s) of reovirus-induced activation of antitumor immunity have not been fully elucidated. Recent studies have demonstrated that overcoming an immunosuppressive environment in tumor-bearing hosts is important to achieve efficient activation of antitumor immunity. Among the various types of cells involved in immunosuppression, it has been revealed that myeloid-derived suppressor cells (MDSCs) are significantly increased in tumor-bearing hosts and play crucial roles in the immunosuppression in tumor-bearing hosts. In this study, we examined whether reovirus inhibits the immunosuppressive activity of MDSCs, resulting in efficient activation of immune cells after in vivo administration. The results showed that splenic MDSCs recovered from PBS-treated tumor-bearing mice significantly suppressed the Ag-specific proliferation of CD8+ T cells. In contrast, the suppressive activity of MDSCs on T cell proliferation was significantly reduced after reovirus administration. Reovirus also inhibited the immunosuppressive activity of MDSCs in IFN-β promoter stimulator-1 knockout (KO) mice and in wild-type mice. In contrast, the immunosuppressive activity of MDSCs in TLR-3 KO mice was not significantly altered by reovirus treatment. The activation levels of CD4+ and CD8+ T cells were significantly lower in TLR3 KO mice than in wild-type mice after reovirus administration. These results indicate that reovirus inhibits the immunosuppressive activity of MDSCs in a TLR3, but not IFN-β promoter stimulator-1, signaling-dependent manner.

Reovirus is a nonenveloped virus that contains 10 segments of dsRNA genome (1). Reovirus has been actively studied as an oncolytic virus that specifically replicates in tumor cells, but not normal cells, leading to efficient tumor cell killing (2). Many clinical studies, including phase III trials, have been ongoing internationally to clarify the effects of reovirus against various types of tumors (3, 4), based on the efficient tumor-killing efficiency, superior safety profile, and other promising properties of reovirus as an oncolytic virus. Moreover, reovirus either used clinically or encountered by common infection rarely induces severe symptoms in adults (2). In addition to these promising properties of reovirus, it has become clear that reovirus-mediated activation of antitumor immunity contributes to the antitumor activity of reovirus (e.g., facilitation of Ag presentation by dendritic cells [DCs]) (5, 6); however, the mechanism of the reovirus-mediated activation of antitumor immunity has not been fully understood.

Activation of antitumor immunity is known to be crucial for tumor regression (7, 8), and thus cancer immunotherapy has been garnering attention for more than a decade (9, 10). Several clinical studies of cancer immunotherapies, including T cell–based immunotherapies (11) and cancer vaccines (12), have been carried out in humans. However, the expected antitumor effects were obtained in only a limited number of clinical studies (13, 14), probably because antitumor immunity is significantly suppressed in cancer patients via various mechanisms (14), such as programmed death-1–mediated and CTLA-4–mediated signaling (15). The blockade of immune checkpoint molecules by using anti–programmed death-1 and anti–CTLA-4 Abs has provided a significant breakthrough in cancer immunotherapy (16). These findings indicate that overriding the immunosuppressive environment in cancer patients is important for effective tumor regression via the activation of antitumor immunity.

Among several known immunosuppressive mechanisms, myeloid-derived suppressor cells (MDSCs) have recently been demonstrated to play crucial roles in the immunosuppression in cancer patients (17). MDSCs are an immature and heterogeneous population that exert immunosuppressive functions via various mechanisms (18, 19). For example, MDSCs produce NO and reactive oxygen species, leading to suppression of T cell proliferation and induction of regulatory T cells (Tregs) (19, 20). Mouse MDSCs are Gr-1+/CD11b+, whereas human MDSCs lack HLA d-related and express either or both CD11b and CD33 (21). Previous studies demonstrated that a decrease in immunosuppressive MDSCs leads to the activation of host immunity, and that several chemotherapy drugs, including fluorouracil and gemcitabine, reduced the numbers of MDSCs in tumor-bearing hosts (22, 23). In addition, immunostimulatory molecules, including CpG oligodeoxynucleotide and polyinosinic-polycytidylic acid (polyI:C), which are ligands of TLRs and/or retinoic acid–inducible gene-I (RIG-I)–like receptors, inhibit the immunosuppressive functions of MDSCs and/or induce differentiation of MDSCs into immune-activating cells, including macrophages (2426). These data suggest that MDSCs are a promising target for cancer immunotherapy, and that anticancer reagents capable of both directly killing tumor cells and also inhibiting the immunosuppressive functions of MDSCs exhibit superior antitumor effects via both mechanisms.

In this study, we hypothesized that reovirus inhibits the immunosuppressive functions of MDSCs after i.v. administration into tumor-bearing hosts, and this effect contributes to the reovirus-mediated tumor regression. We found that the suppressive activity of MDSCs on T cell proliferation was significantly reduced after i.v. injection of reovirus into tumor-bearing mice. The reduction in the immunosuppressive activity of MDSCs by reovirus was observed in IFN-β promoter stimulator-1 (IPS-1)-knockout (KO) mice, but not in TLR3 KO mice. These results indicate that reovirus inhibits the immunosuppressive functions of MDSCs via the TLR3 signaling pathway, followed by activation of antitumor immunity.

C57BL/6J mice were obtained from Nippon SLC (Hamamatsu, Japan). IPS-1 KO mice of a C57BL/6 background were kindly provided by K.J. Ishii (National Institute of Biomedical Innovation, Health and Nutrition, Osaka, Japan) and S. Akira (Osaka University, Osaka, Japan). TLR3 KO mice of a C57BL/6 background were obtained from OrientalBioService (Kyoto, Japan). OT-I transgenic C57BL/6 mice were purchased from Jackson Laboratories (Bar Harbor, ME). GFP transgenic mice were kindly provided by M. Ikawa (Osaka University, Osaka, Japan) (27). All mice were used at 5–7 wk of age. All animal experimental procedures used in this study were performed in accordance with the institutional guidelines for animal experiments at Osaka University. L929 cells (a mouse fibroblast cell line) were maintained with MEM Joklik’s modified (56449C; SAFC Biosciences, Lenexa, KS) supplemented with 5% FBS (Thermo Fisher Scientific, Grand Island, NY). EL4 (a mouse lymphoma cell line) and B16 (a mouse melanoma cell line) cells were maintained with RPMI 1640 medium (R8758-500ML; Sigma-Aldrich, St. Louis, MO) containing 10% FBS. All mediums described earlier were supplemented with streptomycin (100 μg/ml), penicillin (100 U/ml) (Nacalai Tesque, Kyoto, Japan), and GlutaMAX (35050-061; Thermo Fisher Scientific). All cells were routinely monitored for changes in morphology and growth rate, and tested for mycoplasma. Mammalian reovirus type 3 Dearing was amplified in L929 cells and purified by CsCl density gradient centrifugation as previously described (28). Viral titers were determined by a plaque assay using L929 cells. UV-irradiated reovirus (UV-Reo) was prepared by exposing live virus to UV light for 15 min. Loss of infection ability of reovirus was checked by a plaque assay using L929 cells. Information about Abs used in this study are available upon request.

B16 and EL4 cells were seeded at a density of 1 × 104 cells per well in a 96-well plate. On the following day, cells were infected with reovirus for 48 h. Cell viability was evaluated by an AlamarBlue assay.

EL4 tumor cells (2 × 105 cells per mouse) were s.c. transplanted into mice. On day 7, reovirus (2 × 107 PFUs per mouse) and UV-Reo (3 × 108 PFUs per mouse) were i.v. administered to the tumor-bearing mice via the tail vein. The tumor volume was calculated using the following formula: tumor volume (mm3) = 1/2 × (width)2 × (length).

Mice were s.c. transplanted with EL4 cells (2 × 105 cells per mouse) or B16 cells (5 × 105 cells per mouse). Two to three weeks after transplantation, reovirus was i.v. administered at a dose of 3 × 108 PFUs per mouse into tumor-bearing mice. Two days after administration, MDSCs were isolated from the spleen using mouse/human CD11b MicroBeads (130-049-601; Miltenyi, Bergisch Gladbach, Germany) and used for the subsequent experiments, including T cell proliferation assay, as described later.

Splenocytes recovered from OT-I transgenic mice were stained with 50 μM CFSE (341-07401; Dojindo, Kumamoto, Japan). CFSE-stained splenocytes were seeded at a density of 1 × 105 cells per well in a 96-well plate and were stimulated with 20 ng/ml H-2Kb OVA peptide (TS-5001-P; MBL, Nagoya, Japan) in the presence or absence of MDSCs. Three days after coculture, CFSE levels in splenic CD8+ T cells were determined by using flow cytometry (MACS Quant; Miltenyi) to evaluate the proliferation of CD8+ T cells.

Mouse MDSCs were differentiated from bone marrow cells as previously described (29). In brief, mouse bone marrow cells were flushed out from the femur and tibia of mice, after which 2 × 106 cells were cultured in RPMI 1640 medium supplemented with 10% FBS, 100 U/ml penicillin G, 100 μg/ml streptomycin, GlutaMAX, and 40 ng/ml GM-CSF (315-03; PeproTech, Rocky Hill, NJ) for 4 d. Cells were gently collected and inoculated with reovirus or UV-Reo at a concentration equivalent to a multiplicity of infection (MOI) of 5. Twenty-four hours later, the cells were collected, washed, and transferred at a dose of 4 × 106 cells per mouse via tail vein injection to EL4 tumor-bearing mice that received tumor cell transplantation 7–8 d before MDSC transfer. The tumor volume was calculated as described earlier. To evaluate the distribution of MDSCs in the tumor and spleen after transfer in tumor-bearing mice, we differentiated MDSCs from bone marrow cells of GFP-transgenic mice (GFP-MDSCs). GFP-MDSCs were treated with reovirus and transferred to EL4 tumor-bearing mice as described earlier. Spleen and tumor were recovered 24 h after transfer, followed by flow cytometric analysis.

Total RNA was extracted from cells using ISOGEN (319-90211; Nippon Gene, Tokyo, Japan). After the treatment with RNase-free DNase I (M0303L; New England Biolabs, Ipswich, MA), cDNA was synthesized from 1 μg of total RNA using a Superscript VILO cDNA synthesis kit (11754250; Invitrogen). Quantitative PCR analysis was carried out using THUNDERBIRD qPCR Mix (QPS-101; TOYOBO, Osaka, Japan) and StepOnePlus Real-Time PCR systems (Life Technologies). The mRNA levels of the indicated genes were normalized by GAPDH mRNA levels. Sequences of the primers are available upon request.

Cells were washed with cold PBS and lysed with radioimmunoprecipitation assay buffer (89901; Thermo Fisher Scientific, Waltham, MA) containing phosphatase and protease inhibitor mixture (P8340; Sigma-Aldrich) and DL-DTT solution (646563-10X; Sigma-Aldrich). The cell lysates were analyzed by SDS-PAGE. The proteins were electrotransferred onto a polyvinylidene difluoride membrane. Western blot analysis were carried out by using anti–α-tubulin mAb (clone B-7, sc-5286; Santa Cruz Biotechnology, Santa Cruz, CA), anti-reovirus σ3 mAb (clone 4F2; Developmental Studies Hybridoma Bank, Iowa City, IA), and anti–β-actin mAb (clone AC-15, A5441; Sigma-Aldrich) as a primary Ab and Chemi-Lumi One Super (02230; Nacalai Tesque). Images were captured using an LAS-3000 system (Fujifilm, Tokyo, Japan).

Statistical significance was determined using Student t test. Data are presented as the mean ± SD.

To evaluate whether reovirus induced the activation of immune cells, including NK cells and T cells, after i.v. administration into tumor-bearing mice, we measured CD69 expression levels on NK cells and T cells 2 d after i.v. administration of reovirus. CD69 is well known as an activation marker of immune cells (30). We used EL4 and B16 cells for the formation of s.c. tumors in this study because Gr-1+/CD11b+ cells, which included MDSCs, were efficiently increased in the spleen of EL4 and B16 tumor-bearing mice (see later), although EL4 and B16 cells were less susceptible to reovirus infection in vitro than the reovirus-susceptible tumor cells we previously reported (31). The finding that reovirus had only small cytotoxic effects on these cells was partly due to the relatively low expression levels of the primary receptor for reovirus, junctional adhesion molecule-A (JAM-A) (32), on EL4 and B16 cells (Supplemental Fig. 1). The expression levels of CD69 on the cell surface were significantly upregulated not only on NK cells and T cells in the spleen (Fig. 1A, 1B), but also on CD8+ T cells in the tumors of reovirus-treated mice (Fig. 1C, 1D). The average numbers of CD4+/CD69+ T cells in the tumors were also increased by reovirus administration, although not to a statistically significant degree. The numbers of CD69+ NK cells in the tumors were comparable between reovirus-treated and PBS-treated tumor-bearing mice at this time point (2 d after administration). In addition, the numbers of Tregs (CD25+/Foxp3+ cells) in the spleen were reduced by >40% compared with those in PBS-treated mice on day 8 after reovirus administration (Fig. 1E, 1F). Splenic CD8+ T cells expressing CD107a, which is a degranulation maker and is used as a surface marker of activated T cells (33), increased by ∼2-fold on day 8 after reovirus administration (Fig. 1E). UV-Reo also significantly activated NK and T cells in the spleen after i.v. administration (Fig. 1G). Probably because of reovirus-mediated activation of immune cells, not only live reovirus but also UV-Reo significantly suppressed the s.c. EL4 tumor growth after systemic administration, although 15-fold higher amounts of UV-Reo were required to suppress the tumor growth at levels similar to live reovirus (Fig. 1H). These results indicate that antitumor immune cells are significantly activated after reovirus administration in tumor-bearing mice, leading to the suppression of tumor growth.

FIGURE 1.

Reovirus-mediated activation of immune cells in the spleen and tumors of tumor-bearing mice. EL4 tumor cells were s.c. transplanted into mice. Reovirus was i.v. administered on day 7 at a dose of 3 × 108 PFU per mouse. (AD) The expression levels of CD69 on NK cells, CD4+ T cells, and CD8+ T cells in the spleen (A and B) and tumor (C and D). CD69 expression levels were analyzed by flow cytometry 2 d after reovirus administration. Representative dot plots of CD69 expression on NK cells, CD4+ T cells, and CD8+ T cells in the spleen (B) and tumor (D). (E) Frequencies of Foxp3+ cells in CD4+ T cells (Tregs) and of CD107a+ cells in CD8+ T cells (activated CTL) in the spleen. Flow cytometric analysis was performed at 1 wk after reovirus administration. Representative dot plots of Foxp3 expression on CD4+ cells and CD107a expression on CD8+ T cells in the spleen (F). (G) CD69 expression levels on splenic NK cells and T cells following i.v. administration of UV-Reo in tumor-bearing mice. EL4 tumor cells were s.c. transplanted into mice. On day 7, UV-Reo (3 × 108 PFU/mouse) was systemically administered into the tumor-bearing mice. The expression levels of CD69 on NK cells and T cells were analyzed by flow cytometry 2 d after administration. The data represent the mean ± SD (n = 4). *p < 0.05, **p < 0.01, ***p < 0.001 compared with the PBS-treated mice. (H) Comparison of the tumor regression activity of reovirus and UV-Reo in EL4 tumor-bearing mice. Reovirus (2 × 107 PFU per mouse) and UV-Reo (3 × 108 PFU per mouse) were systemically administered to tumor-bearing mice 7 d after transplantation of EL4 cells. The tumor volume was measured following reovirus administration. The data represent the mean ± SD (n = 3–6). *p < 0.05, **p < 0.01, comparing the mice injected with PBS versus reovirus. #p < 0.05, ##p < 0.01 comparing the mice injected with PBS versus UV-Reo.

FIGURE 1.

Reovirus-mediated activation of immune cells in the spleen and tumors of tumor-bearing mice. EL4 tumor cells were s.c. transplanted into mice. Reovirus was i.v. administered on day 7 at a dose of 3 × 108 PFU per mouse. (AD) The expression levels of CD69 on NK cells, CD4+ T cells, and CD8+ T cells in the spleen (A and B) and tumor (C and D). CD69 expression levels were analyzed by flow cytometry 2 d after reovirus administration. Representative dot plots of CD69 expression on NK cells, CD4+ T cells, and CD8+ T cells in the spleen (B) and tumor (D). (E) Frequencies of Foxp3+ cells in CD4+ T cells (Tregs) and of CD107a+ cells in CD8+ T cells (activated CTL) in the spleen. Flow cytometric analysis was performed at 1 wk after reovirus administration. Representative dot plots of Foxp3 expression on CD4+ cells and CD107a expression on CD8+ T cells in the spleen (F). (G) CD69 expression levels on splenic NK cells and T cells following i.v. administration of UV-Reo in tumor-bearing mice. EL4 tumor cells were s.c. transplanted into mice. On day 7, UV-Reo (3 × 108 PFU/mouse) was systemically administered into the tumor-bearing mice. The expression levels of CD69 on NK cells and T cells were analyzed by flow cytometry 2 d after administration. The data represent the mean ± SD (n = 4). *p < 0.05, **p < 0.01, ***p < 0.001 compared with the PBS-treated mice. (H) Comparison of the tumor regression activity of reovirus and UV-Reo in EL4 tumor-bearing mice. Reovirus (2 × 107 PFU per mouse) and UV-Reo (3 × 108 PFU per mouse) were systemically administered to tumor-bearing mice 7 d after transplantation of EL4 cells. The tumor volume was measured following reovirus administration. The data represent the mean ± SD (n = 3–6). *p < 0.05, **p < 0.01, comparing the mice injected with PBS versus reovirus. #p < 0.05, ##p < 0.01 comparing the mice injected with PBS versus UV-Reo.

Close modal

To examine whether reovirus administration resulted in a reduction in the numbers of MDSCs in tumor-bearing mice, we determined the frequencies of Gr-1+/CD11b+ cells in the spleen. Compared with naive mice, the percentages of Gr-1+/CD11b+ cells in the spleen increased by >4-fold in the EL4 tumor-bearing mice (Fig. 2A, 2B). i.v. administration of reovirus at this dose (3 × 108 PFUs per mouse) did not alter the percentages of Gr-1+/CD11b+ cells in the spleen, compared with those in PBS-treated mice, although the flow cytometric patterns of Gr-1 and CD11b expression on the splenocytes were slightly different between PBS- and reovirus-injected mice. We further examined the frequencies of granulocytic MDSCs (Ly6Cmid/Ly6G+) and monocytic MDSCs (Ly6Chigh/Ly6G) in the spleen after reovirus administration. Similar levels of granulocytic MDSCs were found in the spleen of reovirus-treated and PBS-treated mice, whereas the percentages of monocytic MDSCs were significantly lower in the spleen of reovirus-treated mice than in those of PBS-treated mice (Fig. 2C, 2D).

FIGURE 2.

Reduction in the immunosuppressive activity of MDSCs following reovirus administration into tumor-bearing mice. (A and B) The percentages of Gr-1+/CD11b+ cells in the spleen of tumor-bearing mice following reovirus administration are shown by a dot plot (A) and graph (B). The data represent the mean ± SD. **p < 0.01, ***p < 0.001 (n = 4). (C and D) The percentages of Ly6Clow/Ly6Ghigh granulocytic MDSCs and Ly6Chigh/Ly6G monocytic MDSCs in the spleen following reovirus administration are shown by a dot plot (C) and graph (D). Reovirus was i.v. injected into tumor-bearing mice at a dose of 3 × 108 PFUs per mouse. Forty-eight hours later, splenocytes were harvested and stained with anti–Gr-1, CD11b, Ly6C, and Ly6G Abs. (E) Suppressive activities of splenic MDSCs on CD8+ T cell proliferation. Reovirus was i.v. administered at a dose of 3 × 108 PFUs per mouse. Splenic Gr-1+/CD11b+ cells (MDSCs) were cocultured with CFSE-stained splenocytes of OT-I mice. Three days after coculture, the proliferation of CD8+ T cells was evaluated by measuring the CFSE intensity in CD8+ T cells using flow cytometry. The data represent the mean ± SD. **p < 0.01, ***p < 0.001 compared with OVA-stimulated splenocytes without MDSCs (n = 4).

FIGURE 2.

Reduction in the immunosuppressive activity of MDSCs following reovirus administration into tumor-bearing mice. (A and B) The percentages of Gr-1+/CD11b+ cells in the spleen of tumor-bearing mice following reovirus administration are shown by a dot plot (A) and graph (B). The data represent the mean ± SD. **p < 0.01, ***p < 0.001 (n = 4). (C and D) The percentages of Ly6Clow/Ly6Ghigh granulocytic MDSCs and Ly6Chigh/Ly6G monocytic MDSCs in the spleen following reovirus administration are shown by a dot plot (C) and graph (D). Reovirus was i.v. injected into tumor-bearing mice at a dose of 3 × 108 PFUs per mouse. Forty-eight hours later, splenocytes were harvested and stained with anti–Gr-1, CD11b, Ly6C, and Ly6G Abs. (E) Suppressive activities of splenic MDSCs on CD8+ T cell proliferation. Reovirus was i.v. administered at a dose of 3 × 108 PFUs per mouse. Splenic Gr-1+/CD11b+ cells (MDSCs) were cocultured with CFSE-stained splenocytes of OT-I mice. Three days after coculture, the proliferation of CD8+ T cells was evaluated by measuring the CFSE intensity in CD8+ T cells using flow cytometry. The data represent the mean ± SD. **p < 0.01, ***p < 0.001 compared with OVA-stimulated splenocytes without MDSCs (n = 4).

Close modal

Next, we evaluated the immunosuppressive activity of MDSCs recovered from reovirus-treated mice by T cell proliferation assay. T cell proliferation assay is the most commonly used assay for evaluation of the immunosuppressive activity of MDSCs (34, 35). Almost all of the CD11b+ cells (>90%) that we recovered from the spleen by a magnetic sorting expressed Gr-1 (data not shown); therefore, the CD11b+ cells recovered from the spleen are designated as Gr-1+/CD11b+ cells hereafter. As shown in Fig. 2E, MDSCs from PBS-treated mice significantly suppressed the proliferation of CD8+ T cells in a manner dependent on the ratios of MDSCs. Gr-1+/CD11b+ cells from tumor-free mice did not suppress T cell proliferation (data not shown). In contrast, T cell proliferation was not significantly suppressed by MDSCs from reovirus-treated tumor-bearing mice. A reduction in the suppressive activities against T cell proliferation was also found in MDSCs of UV-Reo–treated mice (Supplemental Fig. 2A). Similar results were obtained in MDSCs isolated from B16 tumor-bearing mice (Supplemental Fig. 2B). These results indicate that i.v. administration of reovirus results in inhibition in the immunosuppressive activity of MDSCs in tumor-bearing mice.

Next, to examine whether reovirus was taken up by MDSCs after i.v. administration, we examined whether reovirus genomic RNA was present in the MDSCs recovered from the spleen of reovirus-treated tumor-bearing mice by RT-PCR analysis. More than 90% of MDSCs in the spleen expressed JAM-A, which is an infection receptor for reovirus (36) (Fig. 3A). Reovirus genome was detected in the splenic MDSCs from reovirus-treated mice, but not PBS-treated mice (Fig. 3B). These data suggested that reovirus was internalized into MDSCs in the spleen after i.v. administration. To examine whether reovirus was replicated in MDSCs, we examined outer capsid protein σ3 levels in the splenic MDSCs by Western blotting 48 h after i.v. administration of reovirus and UV-Reo. Detectable levels of the σ3 protein expression were not found in MDSCs of any of the mice (Fig. 3C). In addition, the numbers of viral plaques were comparable between 24 and 48 h after inoculation with reovirus in MDSCs that were induced from mouse bone marrow cells in vitro by cultivation in the presence of GM-CSF (in vitro–generated MDSCs) (29) (Fig. 3D). These data indicate that neither detectable levels of virus protein expression nor progeny virus production occur in MDSCs after inoculation with reovirus.

FIGURE 3.

Reovirus was taken up into MDSCs after i.v. administration. (A) JAM-A expression on splenic MDSCs in tumor-bearing mice. JAM-A expression levels on splenic MDSCs were determined using flow cytometry. (B) Uptake of reovirus in the splenic MDSCs after i.v. administration. Total RNA was extracted from splenic MDSCs following reovirus administration. RT-PCR analysis was performed to detect reovirus L1 genome in the splenic MDSCs. (C) Western blotting analysis of reovirus σ3 protein expression in the splenic MDSCs following reovirus administration. Reovirus and UV-Reo were administered to tumor-bearing mice at a dose of 3 × 108 PFUs per mouse. Splenic MDSCs were recovered 2 d after administration, followed by Western blotting analysis. (D) Progeny virus production in MDSCs following reovirus treatment. In vitro–generated MDSCs were incubated with reovirus and UV-Reo at an MOI of 5 for 24 h. MDSCs were recovered, washed, and subjected to plaque-forming assay. The data represent the mean ± SD (n = 3). N.D., not detected.

FIGURE 3.

Reovirus was taken up into MDSCs after i.v. administration. (A) JAM-A expression on splenic MDSCs in tumor-bearing mice. JAM-A expression levels on splenic MDSCs were determined using flow cytometry. (B) Uptake of reovirus in the splenic MDSCs after i.v. administration. Total RNA was extracted from splenic MDSCs following reovirus administration. RT-PCR analysis was performed to detect reovirus L1 genome in the splenic MDSCs. (C) Western blotting analysis of reovirus σ3 protein expression in the splenic MDSCs following reovirus administration. Reovirus and UV-Reo were administered to tumor-bearing mice at a dose of 3 × 108 PFUs per mouse. Splenic MDSCs were recovered 2 d after administration, followed by Western blotting analysis. (D) Progeny virus production in MDSCs following reovirus treatment. In vitro–generated MDSCs were incubated with reovirus and UV-Reo at an MOI of 5 for 24 h. MDSCs were recovered, washed, and subjected to plaque-forming assay. The data represent the mean ± SD (n = 3). N.D., not detected.

Close modal

Next, in vitro–generated MDSCs were treated with reovirus to examine whether reovirus directly inhibited the immunosuppressive activity of MDSCs after internalization in MDSCs. After a 4-d cultivation in the presence of GM-CSF, >85% of bone marrow cells exhibited high-level expressions of Gr-1 and CD11b (Fig. 4A). Nontreated in vitro–generated MDSCs significantly suppressed the CD8+ T cell proliferation (Fig. 4B), indicating that MDSCs were successfully differentiated from bone marrow cells. Treatment of MDSCs with reovirus led to a significant restoration of CD8+ T cell proliferation. The immunosuppressive activity of MDSCs on CD8+ T cell proliferation was also completely diminished by UV-Reo. Treatment with reovirus did not appear to alter the expression profiles of Gr-1 or CD11b on in vitro–generated MDSCs (Fig. 4C). Reovirus did not greatly reduce the viabilities of MDSCs after inoculation (Fig. 4D). Taken together, these data indicate that reovirus is directly taken up by MDSCs after i.v. administration, leading to a reduction in the suppressive activity of MDSCs independently of virus replication.

FIGURE 4.

UV-inactivated reovirus-treated MDSCs suppressed the s.c. tumor growth in tumor-bearing mice. (A) Expression profiles of CD11b and Gr-1 on in vitro–generated MDSCs. Expression levels of CD11b and Gr-1 were measured before (left) and after (right) a 4-d cultivation in the presence of GM-CSF. (B) Suppressive activities of in vitro–generated MDSCs on CD8+ T cell proliferation. In vitro–generated MDSCs were treated with reovirus and UV-Reo at an MOI of 5, followed by coculture with CFSE-stained OT-1 splenocytes. Three days after coculture, the proliferation of CD8+ T cells was evaluated using flow cytometry. The data represent the mean ± SD. *p < 0.05, **p < 0.01 compared with OVA-stimulated splenocytes without MDSCs (n = 4). (C) Expression profiles of CD11b and Gr-1 on in vitro–generated MDSCs following treatment with reovirus and UV-Reo. (D) Viabilities of in vitro–generated MDSCs following treatment of reovirus and UV-Reo. In vitro–generated MDSCs were incubated with reovirus and UV-Reo at an MOI of 5 for 24 h. Viabilities of MDSCs were determined by 7-AAD staining. The data represent the mean ± SD (n = 4). (E) Tumor growth following transfer of in vitro–generated MDSCs. In vitro–generated MDSCs were incubated with reovirus and UV-Reo at an MOI of 5 for 4 d. After washing, MDSCs were i.v. injected into tumor-bearing mice 7 d after tumor cell transplantation. The data represent the mean ± SD (n = 4–7). *p < 0.05, **p < 0.01 comparing the mice transferred with mock-MDSCs versus reovirus or UV-Reo-treated MDSCs. #p < 0.05 comparing the mice injected with PBS versus mock-MDSCs.

FIGURE 4.

UV-inactivated reovirus-treated MDSCs suppressed the s.c. tumor growth in tumor-bearing mice. (A) Expression profiles of CD11b and Gr-1 on in vitro–generated MDSCs. Expression levels of CD11b and Gr-1 were measured before (left) and after (right) a 4-d cultivation in the presence of GM-CSF. (B) Suppressive activities of in vitro–generated MDSCs on CD8+ T cell proliferation. In vitro–generated MDSCs were treated with reovirus and UV-Reo at an MOI of 5, followed by coculture with CFSE-stained OT-1 splenocytes. Three days after coculture, the proliferation of CD8+ T cells was evaluated using flow cytometry. The data represent the mean ± SD. *p < 0.05, **p < 0.01 compared with OVA-stimulated splenocytes without MDSCs (n = 4). (C) Expression profiles of CD11b and Gr-1 on in vitro–generated MDSCs following treatment with reovirus and UV-Reo. (D) Viabilities of in vitro–generated MDSCs following treatment of reovirus and UV-Reo. In vitro–generated MDSCs were incubated with reovirus and UV-Reo at an MOI of 5 for 24 h. Viabilities of MDSCs were determined by 7-AAD staining. The data represent the mean ± SD (n = 4). (E) Tumor growth following transfer of in vitro–generated MDSCs. In vitro–generated MDSCs were incubated with reovirus and UV-Reo at an MOI of 5 for 4 d. After washing, MDSCs were i.v. injected into tumor-bearing mice 7 d after tumor cell transplantation. The data represent the mean ± SD (n = 4–7). *p < 0.05, **p < 0.01 comparing the mice transferred with mock-MDSCs versus reovirus or UV-Reo-treated MDSCs. #p < 0.05 comparing the mice injected with PBS versus mock-MDSCs.

Close modal

Previous studies demonstrated that immunostimulatory molecules, including CpG oligodeoxynucleotide and polyI:C, which are ligands of TLRs and/or retinoic acid–inducible gene-I (RIG-I)–like receptors, induce differentiation of MDSCs into immune-activating cells (25, 26). In order to examine whether reovirus treatment induced differentiation of MDSCs into immune-activating cells, the expression levels of immune activation markers, including CD86 and MHC class II (MHC II), on MDSCs were examined after reovirus treatment. The expression levels of CD86 and MHC II were significantly elevated on the surface of splenic MDSCs after treatment with reovirus (see below and Supplemental Fig. 3A). In addition, reovirus treatment largely induced type I IFN expression in the splenic MDSCs and in vitro–generated MDSCs (Fig. 6, Supplemental Fig. 3B). Next, to examine whether reovirus-treated MDSCs exhibited antitumor effects, in vitro–generated MDSCs were incubated with reovirus, followed by transfer into tumor-bearing mice. The tumor growth showed a tendency to be promoted in the mice receiving nontreated MDSCs (Fig. 4E). In contrast, transfer of reovirus-treated MDSCs significantly suppressed the tumor growth compared with transfer of mock-treated MDSCs. The growth of s.c. tumors was also significantly reduced by transfer of UV-Reo–treated MDSCs. We did not find significant differences in the tumor growth between the reovirus- and UV-Reo-treated groups. These results suggest that reovirus directly inhibits the immunosuppressive activity of MDSCs and induces differentiation of MDSCs into immune-activating cells after internalization, irrespectively of virus replication. Reovirus-mediated differentiation of MDSCs into immune-activating cells would contribute to a decline in tumor growth.

Next, to examine the biodistribution of in vitro–generated MDSCs after transfer to tumor-bearing mice via tail vein injection, we generated GFP-MDSCs. GFP-MDSCs were treated with reovirus and then transferred into tumor-bearing mice. Flow cytometric analysis demonstrated that similar levels of GFP-MDSCs were detected in the spleen of mice receiving reovirus-treated and mock-treated GFP-MDSCs (Supplemental Fig. 4A). In contrast, the percentages of reovirus-treated GFP-MDSCs in the tumors were almost 3-fold higher than those of mock-treated GFP-MDSCs (Supplemental Fig. 4B). Expression levels of CCR5 and CX3CR1, which have been shown to be involved in the monocytic migration (37, 38), on in vitro–generated MDSCs were upregulated by reovirus treatment (Supplemental Fig. 4C). These results might partly explain the efficient accumulation of reovirus-treated GFP-MDSCs in the tumor. These data indicate that reovirus treatment enhances the tumor accumulation of in vitro–generated MDSCs after i.v. transfer. In addition to the differences in the immune properties between reovirus-treated and mock-treated MDSCs, the differences in the tumor growth after MDSC transfer might be partly due to the differences in the biodistribution of reovirus-treated and mock-treated MDSCs after transfer to tumor-bearing mice.

To examine the mechanisms of reovirus-mediated inhibition of the immunosuppressive activity of MDSCs, we focused on the reovirus dsRNA genome. dsRNA is known to efficiently activate innate immunity via recognition by pattern recognition receptors and to work as an immunogenic adjuvant (39). Reovirus genome has been demonstrated to be recognized by RIG-I, melanoma differentiation–associated protein 5 (MDA5), and TLR3, leading to innate immune responses (3941). IPS-1 KO mice and TLR3 KO mice were administered reovirus to examine the involvement of innate immune responses in reovirus-induced inhibition of the immunosuppressive activity of MDSCs. IPS-1 is an adaptor protein that mediates RIG-I– and MDA5-mediated type I IFN induction (42). Both splenic MDSCs and in vitro–generated MDSCs expressed TLR3 on the endosomal membrane (Fig. 5A). Expressions of RIG-I, MDA5, and IPS-1 were detected in the MDSCs by real-time RT-PCR analysis (Fig. 5B). The mRNA levels of RIG-I, MDA5, and TLR3 were upregulated in MDSCs after reovirus administration, although the expression level of IPS-1 was not significantly altered.

FIGURE 5.

The immunosuppressive activity of MDSCs was reduced in a TLR3-dependent manner following reovirus infection. (A) TLR3 expression levels on splenic MDSCs and in vitro–generated MDSCs. (B) mRNA levels of RIG-I, MDA5, IPS-1, and TLR3 in splenic MDSCs following reovirus administration in tumor-bearing mice. Total RNA was extracted from MDSCs recovered from the spleen of reovirus-treated mice. Quantitative RT-PCR analysis was performed to measure RIG-I, MDA5, IPS-1, and TLR3 mRNA levels. The data were normalized by those in PBS-treated mice. The data represent the mean ± SD (n = 4). (C and D) Suppressive activities of splenic MDSCs recovered from IPS-1 KO mice (C) and TLR3 KO mice (D) following reovirus administration. CFSE-stained OT-I splenocytes were cocultured with splenic MDSCs recovered from WT, IPS-1 KO, and TLR3 KO mice. The data represent the mean ± SD (n = 4). *p < 0.05, **p < 0.01 compared with splenocytes cocultured with MDSCs recovered from PBS-treated mice. (E) Tumor growth following transfer of in vitro–generated WT and TLR3 KO MDSCs. In vitro–generated MDSCs were incubated with UV-Reo at an MOI of 5 for 24 h. After washing, MDSCs were i.v. injected into tumor-bearing mice. The data represent the mean ± SD (n = 5). **p < 0.01 compared with the mice transferred with UV-Reo–treated MDSCs.

FIGURE 5.

The immunosuppressive activity of MDSCs was reduced in a TLR3-dependent manner following reovirus infection. (A) TLR3 expression levels on splenic MDSCs and in vitro–generated MDSCs. (B) mRNA levels of RIG-I, MDA5, IPS-1, and TLR3 in splenic MDSCs following reovirus administration in tumor-bearing mice. Total RNA was extracted from MDSCs recovered from the spleen of reovirus-treated mice. Quantitative RT-PCR analysis was performed to measure RIG-I, MDA5, IPS-1, and TLR3 mRNA levels. The data were normalized by those in PBS-treated mice. The data represent the mean ± SD (n = 4). (C and D) Suppressive activities of splenic MDSCs recovered from IPS-1 KO mice (C) and TLR3 KO mice (D) following reovirus administration. CFSE-stained OT-I splenocytes were cocultured with splenic MDSCs recovered from WT, IPS-1 KO, and TLR3 KO mice. The data represent the mean ± SD (n = 4). *p < 0.05, **p < 0.01 compared with splenocytes cocultured with MDSCs recovered from PBS-treated mice. (E) Tumor growth following transfer of in vitro–generated WT and TLR3 KO MDSCs. In vitro–generated MDSCs were incubated with UV-Reo at an MOI of 5 for 24 h. After washing, MDSCs were i.v. injected into tumor-bearing mice. The data represent the mean ± SD (n = 5). **p < 0.01 compared with the mice transferred with UV-Reo–treated MDSCs.

Close modal

Next, we examined the immunosuppressive activity of MDSCs recovered from IPS-1 KO and TLR3 KO mice after reovirus administration. Splenic MDSCs recovered from PBS-injected IPS-1 KO and TLR3 KO mice significantly suppressed T cell proliferation at levels similar to those from wild-type (WT) mice (Fig. 5C, 5D). As observed for the MDSCs of WT mice, the suppressive activity of MDSCs of IPS-1 KO mice on CD8+ T cell proliferation was ablated by reovirus administration (Fig. 5C). In contrast, administration of reovirus did not alter the suppressive activity of MDSCs of TLR3 KO mice on CD8+ T cell proliferation (Fig. 5D), indicating that TLR3 was involved in reovirus-induced inhibition of immunosuppressive activity of MDSCs. We further performed adoptive MDSC transfer experiments to examine the role of TLR3 on reovirus-induced inhibition of the immunosuppressive activity of MDSCs. UV-Reo–treated MDSCs differentiated from TLR3 KO bone marrow cells did not suppress the tumor growth after transfer into tumor-bearing WT mice, whereas tumor growth was significantly slowed by transfer of UV-Reo–treated MDSCs differentiated from WT bone marrow cells (Fig. 5E), as shown in Fig. 4E. The results described earlier indicate that the reovirus dsRNA genome is recognized by TLR3 in MDSCs and that TLR3 signaling, not IPS-1–dependent signaling, is largely involved in the reovirus-mediated inhibition of immunosuppressive functions of MDSCs.

To further examine the mechanism of TLR3-dependent suppression of the immunosuppressive activity of MDSCs by reovirus, we first examined the expression levels of type I IFNs, IL-6, and IL-12β in in vitro–generated MDSCs after treatment with reovirus (Fig. 6). Reovirus significantly upregulated the mRNA levels of all of the cytokines examined in in vitro–generated MDSCs. In particular, the expression levels of IFN-α and IFN-β were largely increased by reovirus. The mRNA levels of type I IFNs were significantly (∼2-fold) lower in the TLR3 KO MDSCs, compared with WT MDSCs, although there were no significant differences in the mRNA levels of IL-6 or IL-12β between WT MDSCs and TLR3 KO MDSCs after reovirus treatment. Expression levels of all of the cytokines were greatly lower in IPS-1 KO MDSCs than in WT MDSCs. When comparing IPS-1 KO MDSCs with TLR3 KO MDSCs, the cytokine mRNA levels in IPS-1 KO MDSCs were significantly lower than those in TLR3 KO MDSCs. These data indicate that the reovirus genome is recognized by both TLR3 and RIG-I and/or MDA5 in MDSCs, although RIG-I and/or MDA5 play greater roles in reovirus-mediated cytokine induction in MDSCs compared with TLR3.

FIGURE 6.

Cytokine expression levels in in vitro–generated MDSCs following reovirus treatment. Total RNA was extracted from in vitro–generated MDSCs of WT, TLR3 KO, and IPS-1 KO mice following a 24-h incubation with reovirus at an MOI of 5, followed by quantitative RT-PCR analysis. The data represent the mean ± SD (n = 4). ***p < 0.001 compared with the PBS-treated group.

FIGURE 6.

Cytokine expression levels in in vitro–generated MDSCs following reovirus treatment. Total RNA was extracted from in vitro–generated MDSCs of WT, TLR3 KO, and IPS-1 KO mice following a 24-h incubation with reovirus at an MOI of 5, followed by quantitative RT-PCR analysis. The data represent the mean ± SD (n = 4). ***p < 0.001 compared with the PBS-treated group.

Close modal

Next, the expression levels of several cytokines in the splenic MDSCs were evaluated after systemic administration of reovirus. mRNA levels of several cytokines, including type I IFNs, were significantly upregulated in the splenic MDSCs of tumor-bearing WT mice 48 h after i.v. administration of reovirus (Supplemental Fig. 3B). IPS-1 KO MDSCs exhibited a significant reduction in the mRNA levels of type I IFNs and IL-10, compared with WT MDSCs, after reovirus administration. In contrast, the mRNA levels of the cytokines examined were comparable between TLR3 KO MDSCs and WT MDSCs after reovirus administration, although the mRNA levels of type I IFNs in in vitro–generated MDSCs of TLR3 KO mice were significantly lower than those of WT mice following reovirus treatment. This is probably because cytokines and/or soluble factors secreted from other cells in a TLR3-independent manner induced type I IFN expression in splenic MDSCs following reovirus administration. We also determined the mRNA levels of cellular factors that have been reported to be involved in the immunosuppressive activity of MDSCs. mRNA levels of inducible NO synthase (iNOS) and arginase-I tended to be elevated in WT MDSCs following reovirus administration (Supplemental Fig. 3C). The mRNA levels of iNOS and arginase-I were significantly lower in IPS-1 KO MDSCs, compared with WT MDSCs, following reovirus treatment, whereas the levels of iNOS and arginase-I mRNAs were comparable between the WT and TLR3 KO MDSCs. There were no apparent differences in the expression levels of the other genes, including IL-1β or cyclooxygenase 2, between WT MDSCs and IPS-1 or TLR3 KO MDSCs recovered from tumor-bearing mice following reovirus administration. Next, to examine whether reovirus induced differentiation of MDSCs to immunostimulatory cells in a TLR3-dependent manner, the expression levels of CD86 and MHC II, which are immune activation markers, on the MDSCs were analyzed following reovirus injection. Significant upregulation of CD86 and MHC II expression was observed on the WT, IPS-1 KO, and TLR3 KO MDSCs (Supplemental Fig. 3A). The DC marker CD11c was also upregulated on MDSCs in all types of mice receiving reovirus, although there was no statistically significant difference between PBS- and reovirus-infected WT mice when WT and TLR3 KO MDSCs were directly compared. The macrophage differentiation marker F4/80 was not significantly increased on MDSCs after reovirus administration. Comparable levels of these surface markers were found between the WT and TLR3 KO MDSCs, whereas the expression levels of CD86 and MHC II on IPS-1 KO MDSCs were lower than those on WT MDSCs following reovirus treatment. These results suggest that an unknown mechanism dependent on TLR3 signaling is involved in reovirus-mediated suppression of the immunosuppressive activity of MDSCs.

Previous studies have demonstrated that reovirus activated antitumor immunity after i.v. administration into tumor-bearing host, resulting in tumor regression (913). To examine whether TLR3 was involved in the reovirus-mediated activation of immune cells, we assessed the activation status of immune cells, including NK cells and T cells, on the basis of CD69 expression levels. Reovirus induced significant upregulation of CD69 expression on NK cells, CD4+ T cells, and CD8+ T cells of WT mice in the spleen as previously shown; in contrast, the CD69 expression levels on these immune cells in TLR3 KO mice were lower than those in WT mice following the reovirus administration (Fig. 7A, 7B). Moreover, UV-Reo was i.v. injected into tumor-bearing WT and TLR3 KO mice to evaluate the antitumor effects of reovirus-mediated immune activation in a TLR3-dependent manner. UV-Reo was used to remove the contribution of reovirus-mediated lysis of tumor cells to tumor regression in this experiment. UV-Reo significantly suppressed tumor progression in WT mice, but not in TLR3 KO mice (Fig. 7C). These results suggest that reovirus activates the immune cells, at least partly in a TLR3-dependent manner, which contributes to tumor regression.

FIGURE 7.

UV-Reo suppressed the tumor growth in WT mice, but not in TLR3 KO mice. (A and B) CD69 expression levels on the splenic immune cells following reovirus administration. CD69 expression levels were analyzed by flow cytometry 2 d after i.v. administration of reovirus in tumor-bearing mice. Representative dot plots of CD69 expression were shown in (B). Data represent the mean ± SD (n = 4). **p < 0.01, ***p < 0.001 compared with PBS-treated mice. #p < 0.05, ##p < 0.01 comparing reovirus-injected WT mice versus TLR3 KO mice. (C) Tumor growth in WT and TLR3 KO mice following i.v. administration of UV-Reo. Tumor volume was measured following i.v. administration of UV-Reo at a dose of 3 × 108 PFUs per mouse in the WT or TLR3 KO tumor-bearing mice. *p < 0.05, **p < 0.01 compared with PBS-treated mice.

FIGURE 7.

UV-Reo suppressed the tumor growth in WT mice, but not in TLR3 KO mice. (A and B) CD69 expression levels on the splenic immune cells following reovirus administration. CD69 expression levels were analyzed by flow cytometry 2 d after i.v. administration of reovirus in tumor-bearing mice. Representative dot plots of CD69 expression were shown in (B). Data represent the mean ± SD (n = 4). **p < 0.01, ***p < 0.001 compared with PBS-treated mice. #p < 0.05, ##p < 0.01 comparing reovirus-injected WT mice versus TLR3 KO mice. (C) Tumor growth in WT and TLR3 KO mice following i.v. administration of UV-Reo. Tumor volume was measured following i.v. administration of UV-Reo at a dose of 3 × 108 PFUs per mouse in the WT or TLR3 KO tumor-bearing mice. *p < 0.05, **p < 0.01 compared with PBS-treated mice.

Close modal

MDSCs are a crucial target for cancer therapy due to their strong induction of immunosuppressive environments via multiple mechanisms, including suppression of CTL activities and induction of Tregs (17). Various approaches have been used to inhibit the immunosuppressive activities of MDSCs and to induce death of MDSCs. The numbers of Gr-1+/CD11b+ cells, a cell population that includes MDSCs, have been dramatically reduced in tumor-bearing hosts by the administration of gemcitabine and 5-FU (22, 23). In another approach, administration of paclitaxel and CpG oligodeoxynucleotides has been shown to induce differentiation of MDSCs into mature DCs and macrophages, respectively (25, 43). Thus, in addition to exerting direct cytotoxic effects, these anticancer agents also inhibit MDSCs, which further contributes to their efficient antitumor effects.

In a similar manner, the data in this study indicate that reovirus efficiently inhibits the immunosuppressive activities of MDSCs after administration in tumor-bearing mice. Two previous studies have reported the effects of reovirus treatments on the frequencies and immunosuppressive functions of MDSCs following administration in tumor-bearing mice (44, 45). Clements et al. (45) reported that the numbers of Gr-1+/CD11b+ cells in the tumor increased 3 d after i.p. administration of reovirus. They also demonstrated that the mRNA levels of inflammatory cytokines, including IL-6 and TNF-α, which had been reported to be involved in induction of immunosuppressive activities of MDSCs (4648), were significantly upregulated in reovirus-treated mice, compared with control mice, although immunosuppressive activities of MDSCs were not examined in that study. We also found that the mRNA levels of these inflammatory cytokines were significantly elevated in the MDSCs of reovirus-treated mice (Supplemental Fig. 3); however, we did not observe significant increases in the frequency of MDSCs (Gr-1+/CD11b+ cells) in the spleen of tumor-bearing mice, compared with PBS-treated mice, following i.v. administration (Fig. 2A, 2B). In addition, the i.v. administration of reovirus resulted in a significant reduction in the suppressive activities of MDSCs against T cell proliferation (Fig. 2E). These differences in the effects of reovirus on MDSCs between our present study and the previous reports may have been attributable to the different administration routes or the different tumor models, because the characteristics of MDSCs have been shown to differ by tumor types (19). This suggests that we should pay attention to the type of MDSCs in cancer patients, even though the characteristics and classification of MDSC types remain to be clarified.

This study demonstrated that the reovirus dsRNA genome was recognized by TLR3 after internalization of reovirus into MDSCs, leading to inhibition of the immunosuppressive activity of MDSCs. In addition, UV-Reo–treated TLR3 KO MDSCs did not suppress the tumor growth following transfer into the tumor-bearing mice. However, UV-Reo–treated WT MDSCs did significantly delay the tumor growth. These results indicate that TLR3 plays a major role in the reovirus-mediated reduction of the immunosuppressive activity of MDSCs. TLR3 is mainly localized on the endosomes (49). We consider that the reovirus dsRNA genome is recognized by TLR3 on the endosomal membrane following endocytosis of reovirus in MDSCs after i.v. administration. We showed that in vitro–generated TLR3 KO MDSCs expressed lower levels of type I IFN mRNAs than in vitro–generated WT MDSCs following incubation with reovirus, and that there was no apparent reduction in the viability of MDSCs following incubation with reovirus (Figs. 4D, 6). In addition, the previous reports showed that the tumor and virus Ags, including the virus genome, were released into the tumor microenvironment because of virus-infected tumor cell death following oncolytic virus administration (50, 51). Therefore, MDSCs not only took up reovirus, but also might take up the reovirus dsRNA genome released from dying tumor cells and/or reovirus-infected cells following reovirus infection, leading to recognition of the reovirus dsRNA genome by TLR3 in the endosomes.

In contrast, reovirus-mediated inhibition of MDSCs was found in IPS-1 KO mice, similarly to WT mice, indicating that neither RIG-I nor MDA5 is involved in the reovirus-mediated inhibition of MDSCs. Several groups, including ours, reported that the reovirus dsRNA genome is recognized by RIG-I and/or MDA5 postinfection, leading to activation of innate immunity (41, 52, 53). In this study, the mRNA levels of several cytokines, including type I IFNs, were significantly lower in IPS-1 KO MDSCs than in WT MDSCs following reovirus injection (Supplemental Fig. 3). Rather, the mRNA levels of IFN-β were more largely reduced by IPS-1 KO than by TLR3 KO. These data indicate that the RIG-I/MDA5-IPS-1 pathway plays a greater role in reovirus-induced innate immunity in MDSCs than the TLR3 pathway.

Nonetheless, at the present time it remains unclear why the signaling pathway of TLR3, not RIG-I or MDA5, is involved in the reduction in the immunosuppressive activity of MDSCs by reovirus, although both the RIG-I/MDA5 and TLR3 signaling pathways activate IRF3 and NF-κB. An unknown signaling pathway specific for TLR3 might play an important role in the reovirus-mediated suppression of the immunosuppressive activity of MDSCs. The TLR3 signaling pathway would be important for the inhibition of immunosuppressive cells or differentiation of bone marrow–derived immune cells. Shime et al. (54) showed that tumor-supportive macrophages were converted to tumoricidal macrophages via TLR3 signaling following treatment with a dsRNA analog, polyI:C, which is a TLR3 ligand. However, we found that the cell surface markers, including a DC marker CD11c, on splenic Gr-1+/CD11b+ cells of WT mice were not altered by TLR3-dependent signaling following reovirus injection (Supplemental Fig. 3A). The expression profiles of several cytokines and enzymes that are known to be involved in the immunosuppressive activity of MDSCs were comparable between the WT and TLR3 KO MDSCs following reovirus injection. Further experiments are now ongoing to elucidate the mechanisms of reovirus-mediated inhibition of MDSCs via TLR3.

Mouse MDSCs are divided into the two subsets, granulocytic MDSCs and monocytic MDSCs. Monocytic MDSCs have a greater potential for immunosuppressive activity than granulocytic MDSCs (20). In this study, similar levels of granulocytic MDSCs were observed in the spleen of reovirus-treated and PBS-treated mice (Fig. 2C, 2D). In contrast, the percentages of monocytic MDSCs in the spleen were significantly decreased following reovirus administration. The decrease of monocytic MDSCs might partly explain the reovirus-mediated reduction in the suppressive activity of MDSCs.

iNOS and arginase-I are well-known as the important molecules for the immunosuppressive activities of MDSCs; however, reovirus treatment significantly elevated the mRNA levels of iNOS and arginase-I in the splenic MDSCs, although reovirus reduced the immunosuppressive activity of MDSCs. Previous studies reported that iNOS and arginase-I were upregulated by stimulation of innate immunity (55, 56). Reovirus strongly activates innate immunity, resulting in significant upregulation of iNOS and arginase-I. Unknown factors involved in the immunosuppressive activities of MDSCs would be downregulated by reovirus treatment.

We mainly examined the effects of reovirus on splenic MDSCs after i.v. administration in tumor-bearing mice. It remains unclear whether immunosuppressive activities of tumor-infiltrating MDSCs were downregulated following reovirus administration, although tumor-infiltrating NK and T cells were activated (Fig. 1C, 1D). Tumor microenvironment factors, including hypoxia and inflammation, contribute to the immunosuppressive functions of MDSCs (57). Previous studies reported that monocytic MDSCs were more prominent than granulocytic MDSCs in the tumor (58, 59) and that tumor-infiltrating MDSCs have more potent immune suppressive activity than splenic MDSCs (60, 61). In contrast, spleen functions as a reservoir of myeloid cells, including MDSCs (62, 63). Cortez-Retamozo et al. (63) demonstrated that splenic MDSCs physically relocated from the spleen to the tumor. Reovirus-mediated inhibition in the immunosuppressive activity of splenic MDSCs would contribute to the activation of immune cells in the tumor and tumor regression.

Several oncolytic viruses, including vaccinia virus (VV), vesicular stomatitis virus (VSV), HSV, and adenovirus (Ad), which are widely studied as oncolytic viruses, have been reported to increase or decrease the numbers of Gr-1+/CD11b+ cells (MDSCs) after injection in tumor-bearing hosts (6467); however, there have been few studies reporting the functions of MDSCs after administration of these oncolytic viruses. In this study, we revealed that oncolytic reovirus did not appear to increase the numbers of Gr-1+/CD11b+ cells in the spleen after administration, and that the immunosuppressive activity of MDSCs recovered from reovirus-treated mice was significantly suppressed in a TLR3-dependent manner, indicating that TLR3 signaling is crucial for the reduction in the immunosuppressive activity of MDSCs. Reovirus possesses a dsRNA genome, whereas the genomes of VV, HSV, and Ad are dsDNAs. The VSV genome consists of ssRNA. TLR3 signaling would not be efficiently activated by VV, VSV, HSV, or Ad. Reovirus, which has a dsRNA genome, would be a unique oncolytic virus that can efficiently inhibit the immunosuppressive functions of MDSCs.

In summary, we demonstrated that reovirus inhibits the immunosuppressive activity of MDSCs in a TLR3-dependent manner, but not an RIG-I/IPS-1–dependent manner, after systemic administration in tumor-bearing mice. This study indicates that reovirus not only kills cancer cells directly, but also overcomes the immunosuppressive environment in tumor-bearing hosts, leading to efficient activation of antitumor immune responses. Reovirus shows effective antitumor effects through the various mechanisms described earlier and could become a promising antitumor agent.

We thank Sayuri Okamoto and Eri Hosoyamada (Osaka University, Osaka, Japan) for their help, and Dr. Naoki Okada and Kento Fujiwara (Osaka University, Osaka, Japan) for their technical assistance. Reovirus was kindly provided by Dr. Akira Nishizono and Dr. Tsuyoshi Eto (Oita University, Oita, Japan).

This work was supported by a Grant-in-Aid for Scientific Research (B) (to F.S.) from the Ministry of Education, Culture, Sports, Sciences, and Technology of Japan, the Yokoyama Foundation for Clinical Pharmacology (to F.S.), and the SEI Group CSR Foundation (to M.T.). Y.K. is a Research Fellow of the Japan Society for the Promotion of Science.

The online version of this article contains supplemental material.

Abbreviations used in this article:

Ad

adenovirus

DC

dendritic cell

GFP-MDSC

MDSC differentiated from bone marrow cells of GFP-transgenic mouse

iNOS

inducible NO synthase

IPS-1

IFN-β promoter stimulator-1

JAM-A

junctional adhesion molecule-A

MDA5

melanoma differentiation–associated protein 5

MDSC

myeloid-derived suppressor cell

MHC II

MHC class II

MOI

multiplicity of infection

polyI:C

polyinosinic-polycytidylic acid

RIG-I

retinoic acid–inducible gene-I

Treg

regulatory T cell

UV-Reo

UV-irradiated reovirus

VSV

vesicular stomatitis virus

VV

vaccinia virus

WT

wild-type.

1
Gomatos
,
P. J.
,
I.
Tamm
.
1963
.
The secondary structure of reovirus Rna.
Proc. Natl. Acad. Sci. USA
49
:
707
714
.
2
Kelly
,
K.
,
S.
Nawrocki
,
A.
Mita
,
M.
Coffey
,
F. J.
Giles
,
M.
Mita
.
2009
.
Reovirus-based therapy for cancer.
Expert Opin. Biol. Ther.
9
:
817
830
.
3
Galanis
,
E.
,
S. N.
Markovic
,
V. J.
Suman
,
G. J.
Nuovo
,
R. G.
Vile
,
T. J.
Kottke
,
W. K.
Nevala
,
M. A.
Thompson
,
J. E.
Lewis
,
K. M.
Rumilla
, et al
.
2012
.
Phase II trial of intravenous administration of Reolysin(®) (Reovirus Serotype-3-dearing Strain) in patients with metastatic melanoma.
Mol. Ther.
20
:
1998
2003
.
4
Kyula
,
J. N.
,
V.
Roulstone
,
E. M.
Karapanagiotou
,
A. A.
Melcher
,
K. J.
Harrington
.
2012
.
Oncolytic reovirus type 3 (Dearing) as a novel therapy in head and neck cancer.
Expert Opin. Biol. Ther.
12
:
1669
1678
.
5
Errington
,
F.
,
L.
Steele
,
R.
Prestwich
,
K. J.
Harrington
,
H. S.
Pandha
,
L.
Vidal
,
J.
de Bono
,
P.
Selby
,
M.
Coffey
,
R.
Vile
,
A.
Melcher
.
2008
.
Reovirus activates human dendritic cells to promote innate antitumor immunity.
J. Immunol.
180
:
6018
6026
.
6
Gujar
,
S. A.
,
P.
Marcato
,
D.
Pan
,
P. W.
Lee
.
2010
.
Reovirus virotherapy overrides tumor antigen presentation evasion and promotes protective antitumor immunity.
Mol. Cancer Ther.
9
:
2924
2933
.
7
Vesely
,
M. D.
,
M. H.
Kershaw
,
R. D.
Schreiber
,
M. J.
Smyth
.
2011
.
Natural innate and adaptive immunity to cancer.
Annu. Rev. Immunol.
29
:
235
271
.
8
Lichty
,
B. D.
,
C. J.
Breitbach
,
D. F.
Stojdl
,
J. C.
Bell
.
2014
.
Going viral with cancer immunotherapy.
Nat. Rev. Cancer
14
:
559
567
.
9
Makkouk
,
A.
,
G. J.
Weiner
.
2015
.
Cancer immunotherapy and breaking immune tolerance: new approaches to an old challenge.
Cancer Res.
75
:
5
10
.
10
Duong
,
C. P.
,
C. S.
Yong
,
M. H.
Kershaw
,
C. Y.
Slaney
,
P. K.
Darcy
.
2015
.
Cancer immunotherapy utilizing gene-modified T cells: From the bench to the clinic.
Mol. Immunol.
67
(
2
Pt A
):
46
57
.
11
Rosenberg
,
S. A.
,
N. P.
Restifo
.
2015
.
Adoptive cell transfer as personalized immunotherapy for human cancer.
Science
348
:
62
68
.
12
Wang
,
R. F.
1999
.
Human tumor antigens: implications for cancer vaccine development.
J. Mol. Med.
77
:
640
655
.
13
Kissick
,
H. T.
,
M. G.
Sanda
.
2015
.
The role of active vaccination in cancer immunotherapy: lessons from clinical trials.
Curr. Opin. Immunol.
35
:
15
22
.
14
Ghirelli
,
C.
,
T.
Hagemann
.
2013
.
Targeting immunosuppression for cancer therapy.
J. Clin. Invest.
123
:
2355
2357
.
15
Sharma
,
P.
,
J. P.
Allison
.
2015
.
The future of immune checkpoint therapy.
Science
348
:
56
61
.
16
Postow
,
M. A.
,
M. K.
Callahan
,
J. D.
Wolchok
.
2015
.
Immune checkpoint blockade in cancer therapy.
J. Clin. Oncol.
33
:
1974
1982
.
17
Gabrilovich
,
D. I.
,
S.
Nagaraj
.
2009
.
Myeloid-derived suppressor cells as regulators of the immune system.
Nat. Rev. Immunol.
9
:
162
174
.
18
Ostrand-Rosenberg
,
S.
,
P.
Sinha
.
2009
.
Myeloid-derived suppressor cells: linking inflammation and cancer.
J. Immunol.
182
:
4499
4506
.
19
Youn
,
J. I.
,
S.
Nagaraj
,
M.
Collazo
,
D. I.
Gabrilovich
.
2008
.
Subsets of myeloid-derived suppressor cells in tumor-bearing mice.
J. Immunol.
181
:
5791
5802
.
20
Movahedi
,
K.
,
M.
Guilliams
,
J.
Van den Bossche
,
R.
Van den Bergh
,
C.
Gysemans
,
A.
Beschin
,
P.
De Baetselier
,
J. A.
Van Ginderachter
.
2008
.
Identification of discrete tumor-induced myeloid-derived suppressor cell subpopulations with distinct T cell-suppressive activity.
Blood
111
:
4233
4244
.
21
Talmadge
,
J. E.
,
D. I.
Gabrilovich
.
2013
.
History of myeloid-derived suppressor cells.
Nat. Rev. Cancer
13
:
739
752
.
22
Vincent
,
J.
,
G.
Mignot
,
F.
Chalmin
,
S.
Ladoire
,
M.
Bruchard
,
A.
Chevriaux
,
F.
Martin
,
L.
Apetoh
,
C.
Rébé
,
F.
Ghiringhelli
.
2010
.
5-Fluorouracil selectively kills tumor-associated myeloid-derived suppressor cells resulting in enhanced T cell-dependent antitumor immunity.
Cancer Res.
70
:
3052
3061
.
23
Suzuki
,
E.
,
V.
Kapoor
,
A. S.
Jassar
,
L. R.
Kaiser
,
S. M.
Albelda
.
2005
.
Gemcitabine selectively eliminates splenic Gr-1+/CD11b+ myeloid suppressor cells in tumor-bearing animals and enhances antitumor immune activity.
Clin. Cancer Res.
11
:
6713
6721
.
24
Zoglmeier
,
C.
,
H.
Bauer
,
D.
Nörenberg
,
G.
Wedekind
,
P.
Bittner
,
N.
Sandholzer
,
M.
Rapp
,
D.
Anz
,
S.
Endres
,
C.
Bourquin
.
2011
.
CpG blocks immunosuppression by myeloid-derived suppressor cells in tumor-bearing mice.
Clin. Cancer Res.
17
:
1765
1775
.
25
Shirota
,
Y.
,
H.
Shirota
,
D. M.
Klinman
.
2012
.
Intratumoral injection of CpG oligonucleotides induces the differentiation and reduces the immunosuppressive activity of myeloid-derived suppressor cells.
J. Immunol.
188
:
1592
1599
.
26
Shime
,
H.
,
A.
Kojima
,
A.
Maruyama
,
Y.
Saito
,
H.
Oshiumi
,
M.
Matsumoto
,
T.
Seya
.
2014
.
Myeloid-derived suppressor cells confer tumor-suppressive functions on natural killer cells via polyinosinic:polycytidylic acid treatment in mouse tumor models.
J. Innate Immun.
6
:
293
305
.
27
Okabe
,
M.
,
M.
Ikawa
,
K.
Kominami
,
T.
Nakanishi
,
Y.
Nishimune
.
1997
.
‘Green mice’ as a source of ubiquitous green cells.
FEBS Lett.
407
:
313
319
.
28
Berard
,
A.
,
K. M.
Coombs
.
2009
.
Mammalian reoviruses: propagation, quantification, and storage.
Curr. Protoc. Microbiol.
DOI: 10.1002/9780471729259.mc15c01s14.
29
Rössner
,
S.
,
C.
Voigtländer
,
C.
Wiethe
,
J.
Hänig
,
C.
Seifarth
,
M. B.
Lutz
.
2005
.
Myeloid dendritic cell precursors generated from bone marrow suppress T cell responses via cell contact and nitric oxide production in vitro.
Eur. J. Immunol.
35
:
3533
3544
.
30
Testi
,
R.
,
J. H.
Phillips
,
L. L.
Lanier
.
1989
.
T cell activation via Leu-23 (CD69).
J. Immunol.
143
:
1123
1128
.
31
Terasawa
,
Y.
,
T.
Hotani
,
Y.
Katayama
,
M.
Tachibana
,
H.
Mizuguchi
,
F.
Sakurai
.
2015
.
Activity levels of cathepsins B and L in tumor cells are a biomarker for efficacy of reovirus-mediated tumor cell killing.
Cancer Gene Ther.
22
:
188
197
.
32
Campbell
,
J. A.
,
P.
Schelling
,
J. D.
Wetzel
,
E. M.
Johnson
,
J. C.
Forrest
,
G. A.
Wilson
,
M.
Aurrand-Lions
,
B. A.
Imhof
,
T.
Stehle
,
T. S.
Dermody
.
2005
.
Junctional adhesion molecule a serves as a receptor for prototype and field-isolate strains of mammalian reovirus.
J. Virol.
79
:
7967
7978
.
33
Betts
,
M. R.
,
J. M.
Brenchley
,
D. A.
Price
,
S. C.
De Rosa
,
D. C.
Douek
,
M.
Roederer
,
R. A.
Koup
.
2003
.
Sensitive and viable identification of antigen-specific CD8+ T cells by a flow cytometric assay for degranulation.
J. Immunol. Methods
281
:
65
78
.
34
Watanabe
,
S.
,
K.
Deguchi
,
R.
Zheng
,
H.
Tamai
,
L. X.
Wang
,
P. A.
Cohen
,
S.
Shu
.
2008
.
Tumor-induced CD11b+Gr-1+ myeloid cells suppress T cell sensitization in tumor-draining lymph nodes.
J. Immunol.
181
:
3291
3300
.
35
Pan
,
P. Y.
,
G.
Ma
,
K. J.
Weber
,
J.
Ozao-Choy
,
G.
Wang
,
B.
Yin
,
C. M.
Divino
,
S. H.
Chen
.
2010
.
Immune stimulatory receptor CD40 is required for T-cell suppression and T regulatory cell activation mediated by myeloid-derived suppressor cells in cancer.
Cancer Res.
70
:
99
108
.
36
Barton
,
E. S.
,
J. C.
Forrest
,
J. L.
Connolly
,
J. D.
Chappell
,
Y.
Liu
,
F. J.
Schnell
,
A.
Nusrat
,
C. A.
Parkos
,
T. S.
Dermody
.
2001
.
Junction adhesion molecule is a receptor for reovirus.
Cell
104
:
441
451
.
37
Geissmann
,
F.
,
S.
Jung
,
D. R.
Littman
.
2003
.
Blood monocytes consist of two principal subsets with distinct migratory properties.
Immunity
19
:
71
82
.
38
Tacke
,
F.
,
D.
Alvarez
,
T. J.
Kaplan
,
C.
Jakubzick
,
R.
Spanbroek
,
J.
Llodra
,
A.
Garin
,
J.
Liu
,
M.
Mack
,
N.
van Rooijen
, et al
.
2007
.
Monocyte subsets differentially employ CCR2, CCR5, and CX3CR1 to accumulate within atherosclerotic plaques.
J. Clin. Invest.
117
:
185
194
.
39
Alexopoulou
,
L.
,
A. C.
Holt
,
R.
Medzhitov
,
R. A.
Flavell
.
2001
.
Recognition of double-stranded RNA and activation of NF-kappaB by Toll-like receptor 3.
Nature
413
:
732
738
.
40
Goubau
,
D.
,
M.
Schlee
,
S.
Deddouche
,
A. J.
Pruijssers
,
T.
Zillinger
,
M.
Goldeck
,
C.
Schuberth
,
A. G.
Van der Veen
,
T.
Fujimura
,
J.
Rehwinkel
, et al
.
2014
.
Antiviral immunity via RIG-I-mediated recognition of RNA bearing 5′-diphosphates.
Nature
514
:
372
375
.
41
Kato
,
H.
,
O.
Takeuchi
,
E.
Mikamo-Satoh
,
R.
Hirai
,
T.
Kawai
,
K.
Matsushita
,
A.
Hiiragi
,
T. S.
Dermody
,
T.
Fujita
,
S.
Akira
.
2008
.
Length-dependent recognition of double-stranded ribonucleic acids by retinoic acid-inducible gene-I and melanoma differentiation-associated gene 5.
J. Exp. Med.
205
:
1601
1610
.
42
Kawai
,
T.
,
K.
Takahashi
,
S.
Sato
,
C.
Coban
,
H.
Kumar
,
H.
Kato
,
K. J.
Ishii
,
O.
Takeuchi
,
S.
Akira
.
2005
.
IPS-1, an adaptor triggering RIG-I- and Mda5-mediated type I interferon induction.
Nat. Immunol.
6
:
981
988
.
43
Michels
,
T.
,
G. V.
Shurin
,
H.
Naiditch
,
A.
Sevko
,
V.
Umansky
,
M. R.
Shurin
.
2012
.
Paclitaxel promotes differentiation of myeloid-derived suppressor cells into dendritic cells in vitro in a TLR4-independent manner.
J. Immunotoxicol.
9
:
292
300
.
44
Gujar
,
S. A.
,
D.
Clements
,
R.
Dielschneider
,
E.
Helson
,
P.
Marcato
,
P. W. K.
Lee
.
2014
.
Gemcitabine enhances the efficacy of reovirus-based oncotherapy through anti-tumour immunological mechanisms.
Br. J. Cancer
110
:
83
93
.
45
Clements
,
D. R.
,
A. M.
Sterea
,
Y.
Kim
,
E.
Helson
,
C. A.
Dean
,
A.
Nunokawa
,
K. M.
Coyle
,
T.
Sharif
,
P.
Marcato
,
S. A.
Gujar
,
P. W.
Lee
.
2015
.
Newly recruited CD11b+, GR-1+, Ly6C(high) myeloid cells augment tumor-associated immunosuppression immediately following the therapeutic administration of oncolytic reovirus.
J. Immunol.
194
:
4397
4412
.
46
Bunt
,
S. K.
,
L.
Yang
,
P.
Sinha
,
V. K.
Clements
,
J.
Leips
,
S.
Ostrand-Rosenberg
.
2007
.
Reduced inflammation in the tumor microenvironment delays the accumulation of myeloid-derived suppressor cells and limits tumor progression.
Cancer Res.
67
:
10019
10026
.
47
Sumida
,
K.
,
D.
Wakita
,
Y.
Narita
,
K.
Masuko
,
S.
Terada
,
K.
Watanabe
,
T.
Satoh
,
H.
Kitamura
,
T.
Nishimura
.
2012
.
Anti-IL-6 receptor mAb eliminates myeloid-derived suppressor cells and inhibits tumor growth by enhancing T-cell responses.
Eur. J. Immunol.
42
:
2060
2072
.
48
Zhao
,
X.
,
L.
Rong
,
X.
Zhao
,
X.
Li
,
X.
Liu
,
J.
Deng
,
H.
Wu
,
X.
Xu
,
U.
Erben
,
P.
Wu
, et al
.
2012
.
TNF signaling drives myeloid-derived suppressor cell accumulation.
J. Clin. Invest.
122
:
4094
4104
.
49
Matsumoto
,
M.
,
K.
Funami
,
M.
Tanabe
,
H.
Oshiumi
,
M.
Shingai
,
Y.
Seto
,
A.
Yamamoto
,
T.
Seya
.
2003
.
Subcellular localization of Toll-like receptor 3 in human dendritic cells.
J. Immunol.
171
:
3154
3162
.
50
Schulz
,
O.
,
S. S.
Diebold
,
M.
Chen
,
T. I.
Näslund
,
M. A.
Nolte
,
L.
Alexopoulou
,
Y. T.
Azuma
,
R. A.
Flavell
,
P.
Liljeström
,
C.
Reis e Sousa
.
2005
.
Toll-like receptor 3 promotes cross-priming to virus-infected cells.
Nature
433
:
887
892
.
51
Walker
,
J. R.
,
K. G.
McGeagh
,
P.
Sundaresan
,
T. J.
Jorgensen
,
S. D.
Rabkin
,
R. L.
Martuza
.
1999
.
Local and systemic therapy of human prostate adenocarcinoma with the conditionally replicating herpes simplex virus vector G207.
Hum. Gene Ther.
10
:
2237
2243
.
52
Katayama
,
Y.
,
Y.
Terasawa
,
M.
Tachibana
,
H.
Mizuguchi
,
F.
Sakurai
.
2015
.
Proteolytic disassembly of viral outer capsid proteins is crucial for reovirus-mediated type-I interferon induction in both reovirus-susceptible and reovirus-refractory tumor cells.
BioMed Res. Int.
2015
:
468457
.
53
Loo
,
Y. M.
,
J.
Fornek
,
N.
Crochet
,
G.
Bajwa
,
O.
Perwitasari
,
L.
Martinez-Sobrido
,
S.
Akira
,
M. A.
Gill
,
A.
García-Sastre
,
M. G.
Katze
,
M.
Gale
Jr
2008
.
Distinct RIG-I and MDA5 signaling by RNA viruses in innate immunity
.
J. Virol.
82
:
335
345
.
54
Shime
,
H.
,
M.
Matsumoto
,
H.
Oshiumi
,
S.
Tanaka
,
A.
Nakane
,
Y.
Iwakura
,
H.
Tahara
,
N.
Inoue
,
T.
Seya
.
2012
.
Toll-like receptor 3 signaling converts tumor-supporting myeloid cells to tumoricidal effectors.
Proc. Natl. Acad. Sci. USA
109
:
2066
2071
.
55
Akaike
,
T.
,
H.
Maeda
.
2000
.
Nitric oxide and virus infection.
Immunology
101
:
300
308
.
56
Munder
,
M.
2009
.
Arginase: an emerging key player in the mammalian immune system.
Br. J. Pharmacol.
158
:
638
651
.
57
Di Mitri
,
D.
,
A.
Toso
,
A.
Alimonti
.
2015
.
Molecular pathways: targeting tumor-infiltrating myeloid-derived suppressor cells for cancer therapy.
Clin. Cancer Res.
21
:
3108
3112
.
58
Hossain
,
F.
,
A. A.
Al-Khami
,
D.
Wyczechowska
,
C.
Hernandez
,
L.
Zheng
,
K.
Reiss
,
L. D.
Valle
,
J.
Trillo-Tinoco
,
T.
Maj
,
W.
Zou
, et al
.
2015
.
Inhibition of fatty acid oxidation modulates immunosuppressive functions of myeloid-derived suppressor cells and enhances cancer therapies.
Cancer Immunol. Res.
3
:
1236
1247
.
59
Haverkamp
,
J. M.
,
S. A.
Crist
,
B. D.
Elzey
,
C.
Cimen
,
T. L.
Ratliff
.
2011
.
In vivo suppressive function of myeloid-derived suppressor cells is limited to the inflammatory site.
Eur. J. Immunol.
41
:
749
759
.
60
Maenhout
,
S. K.
,
S.
Van Lint
,
P. U.
Emeagi
,
K.
Thielemans
,
J. L.
Aerts
.
2014
.
Enhanced suppressive capacity of tumor-infiltrating myeloid-derived suppressor cells compared with their peripheral counterparts.
Int. J. Cancer
134
:
1077
1090
.
61
Corzo
,
C. A.
,
T.
Condamine
,
L.
Lu
,
M. J.
Cotter
,
J. I.
Youn
,
P.
Cheng
,
H. I.
Cho
,
E.
Celis
,
D. G.
Quiceno
,
T.
Padhya
, et al
.
2010
.
HIF-1α regulates function and differentiation of myeloid-derived suppressor cells in the tumor microenvironment.
J. Exp. Med.
207
:
2439
2453
.
62
Younos
,
I. H.
,
A. J.
Dafferner
,
D.
Gulen
,
H. C.
Britton
,
J. E.
Talmadge
.
2012
.
Tumor regulation of myeloid-derived suppressor cell proliferation and trafficking.
Int. Immunopharmacol.
13
:
245
256
.
63
Cortez-Retamozo
,
V.
,
M.
Etzrodt
,
A.
Newton
,
P. J.
Rauch
,
A.
Chudnovskiy
,
C.
Berger
,
R. J.
Ryan
,
Y.
Iwamoto
,
B.
Marinelli
,
R.
Gorbatov
, et al
.
2012
.
Origins of tumor-associated macrophages and neutrophils.
Proc. Natl. Acad. Sci. USA
109
:
2491
2496
.
64
Condamine
,
T.
,
D. I.
Gabrilovich
.
2011
.
Molecular mechanisms regulating myeloid-derived suppressor cell differentiation and function.
Trends Immunol.
32
:
19
25
.
65
Gil
,
M.
,
M.
Bieniasz
,
M.
Seshadri
,
D.
Fisher
,
M. J.
Ciesielski
,
Y.
Chen
,
R. K.
Pandey
,
D.
Kozbor
.
2011
.
Photodynamic therapy augments the efficacy of oncolytic vaccinia virus against primary and metastatic tumours in mice.
Br. J. Cancer
105
:
1512
1521
.
66
Kaufman
,
H. L.
,
D. W.
Kim
,
G.
DeRaffele
,
J.
Mitcham
,
R. S.
Coffin
,
S.
Kim-Schulze
.
2010
.
Local and distant immunity induced by intralesional vaccination with an oncolytic herpes virus encoding GM-CSF in patients with stage IIIc and IV melanoma.
Ann. Surg. Oncol.
17
:
718
730
.
67
Cerullo
,
V.
,
I.
Diaconu
,
V.
Romano
,
M.
Hirvinen
,
M.
Ugolini
,
S.
Escutenaire
,
S. L.
Holm
,
A.
Kipar
,
A.
Kanerva
,
A.
Hemminki
.
2012
.
An oncolytic adenovirus enhanced for toll-like receptor 9 stimulation increases antitumor immune responses and tumor clearance.
Mol. Ther.
20
:
2076
2086
.

The authors have no financial conflicts of interest.

Supplementary data