Reovirus inhibits the growth of murine colorectal cancer cell lines.
We investigated the cytotoxic effect of reovirus against CT26, CMT93, and MC38 using cell viability assay. After treatment with reovirus for three days, proliferation was significantly decreased in all cell lines at various degrees (Fig. 1A). Next, we examined the expression of a cytosolic DNA sensor, cGAS and STING, in murine colorectal cancer cell lines including CT26, CMT93, and MC38. Western blot analysis demonstrated that cGAS was expressed in CT26 and MC38. STING showed varied expression in all cell lines examined (Fig. 1B). A STING agonist, ADU-S100, had no significant additive effect on cytotoxicity of the reovirus against all cell lines we examined in vitro (supplemental Fig. 1).
A STING agonist suppresses the inhibitory effect of reovirus against IFN responses.
We used CT26 cells to investigate the activation of the STING signaling pathway responsible for type I IFN regulation in CT26 cells. We examined the activation of STING signaling stimulated by reovirus alone or in combination with ADU-S100 by evaluating the phosphorylation of IRF-3, downstream factor of STING signaling, using western blot analysis. No activation of STING signals was detected in CT26 cells treated with reovirus. In contrast, STING agonist, ADU-S100 significantly induced the phosphorylation of IRF3; however, this effect was attenuated when ADU-S100 was administered with the reovirus (Fig. 2A, B). We performed immunofluorescence staining to examine the translocation of IRF3 in CT26 cells treated with reovirus. One hour after treatment with ADU-S100, IRF3 localized into the nucleus confirming the western blot analysis. Although reovirus alone did not induce the translocation of IRF3 into the nucleus, IRF3 was detected in the nucleus of cells treated with reovirus combined with ADU-S100 (Fig. 2C). Quantitative RT-PCR revealed that CT26 cells produced IFN-β in a concentration-dependent manner in response to ADU-S100 (supplemental Fig. 2). Reovirus significantly suppressed the expression of IFN-β enhanced by ADU-S100 in CT26 cells (Fig. 2D). STING agonist also suppresses the inhibitory effect of reovirus against IFN responses in MC 38 cells (supplemental Fig. 3). These results indicate the suppressive effect of reovirus against IFN-β production was partially canceled by a STING agonist in vitro.
Induction of immunogenic cell death in CT26 cells by reovirus
We measured the concentration of ATP in culture media of CT26 cells treated with reovirus and the HGMB-1 released from CT26 cells after exposure to reovirus to evaluate the immunogenic cell death induced by reovirus in CT26 cells. The concentration of extracellular ATP in culture media of CT26 treated with reovirus was significantly higher than that in culture media of CT26 without reovirus treatment (Fig. 2D). A higher concentration of HGMB-1 was detected in the culture media congaing CT26 treated reovirus (Fig. 2E). These results demonstrate that reovirus induces immunogenic cell death in CT26.
Efficacy of reovirus in combination with a STING agonist on the growth of CT26 cells in vivo.
To investigate whether reovirus and ADU-S100 synergistically induced anti-tumor efficacy against colorectal cancer (CRC) in immunogenic settings, we used a syngeneic murine model of CRC. To analyze the anti-tumor immune response, CT26 cells were subcutaneously injected into BALB/c mice. The tumor size was measured, and the tumor volume was calculated after the intratumoral (IT) injection of reovirus, ADU-S100, or reovirus and ADU-S100. Reovirus IT injection significantly inhibited tumor growth compared with controls. IT treatment with ADU-S100 also inhibited tumor growth, and the combination of reovirus and ADU-S100 demonstrated the strongest anti-tumor activity, and ADU-S100 showed the significant additive effect on anti-tumor activity of the reovirus (Fig. 3A). Additionally, the combination treatment significantly improved the survival of tumor-bearing mice compared with controls (supplemental Fig. 4). In the combination treatment group, four of eight mice achieved complete regression of the tumor. Histopathological analyses (supplemental data 5) of extracted tumors from mice three days after IT injection of anticancer agents revealed the number of Ki-67-positive cells was significantly decreased in the implanted tumor treated with the IT injection of reovirus, either alone or in combination with ADU-S100, compared with controls (Fig. 3B). Furthermore, the number of cleaved caspase-3-positive cells was increased in the combination reovirus and ADU-S100 group compared with controls (Fig. 3C). We also detected a significant increase in the number of granzyme B positive cells in tumors treated with the combination treatment compared with controls (Fig. 3D). These results suggest that reovirus synergizes with ADU-S100 to promote an anti-tumor effect in tumor-bearing mice and that the efficacy of the combination treatment might be derived by enhanced anti-tumor immunity.
Combined reovirus and ADU-S100 control tumor growth in a dual flank tumor model.
To investigate the role of immune responses in primary tumor growth inhibition, we performed a dual flank study using an immunocompetent syngeneic model. In the dual flank syngeneic mouse tumor model, CT26 cells were implanted to both sides of a mouse and then one tumor was injected with reovirus, ADU-S100, or reovirus and ADU-S100, while the other tumor remained untreated. The combination reovirus and ADU-S100 treatment induced the significant regression of the treated tumor and the untreated distant tumor, suggesting a systemic immune response was activated by the combination treatment in a dual flunk tumor model (Fig. 4A). In contrast, IT injection of reovirus or ADU-S100 could not significantly delayed tumor growth in the untreated distal tumor. The combination treatment also significantly prolonged the survival of dual tumor-bearing mice (Fig. 4B). Furthermore, we found the survival of tumor-bearing mice was determined by the growth of the untreated tumor in this model. These results demonstrate the abscopal effect mediated by tumor immunity induced by the focal IT injection of reovirus in combination with ADU-S100, and that control of the untreated tumor is essential for achieving long-term survival.
Microenvironment of tumors treated with reovirus and a STING agonist.
To investigate whether the STING agonist restricted the propagation of reovirus in vivo, we performed immunofluorescence staining of reovirus protein in tumor samples extracted from mice. Three days after reovirus IT injection, reovirus protein was detected in the reovirus IT injected tumors but not in untreated distal tumor grafts (Fig. 5A). In addition to that, in tumors treated with the IT injection of reovirus followed by ADU-S100, the number of reovirus protein positive cells were almost same as those in tumors injected with reovirus alone (Fig. 5B). These results indicate that pretreatment with a STING agonist did not restrict the propagation of reovirus in the tumor despite the activation of STING signaling, and that reovirus infection had not spread to the untreated distal tumor within 3 days after reovirus IT injection. Immunohistochemical staining demonstrated that combination reovirus and ADU-S100 treatment induced significant apoptotic cell death (Fig. 5C, D). Enhanced granzyme B staining and upregulation of IFN-β mRNA expression in untreated tumors of the combination group indicated the combination IT injection boosted cytotoxic immune responses (Fig. 5E, F). These results suggest the combination reovirus and ADU-S100 treatment enhanced antitumoral immune responses via type I IFNs without reovirus elimination.
To obtain an overview of the immune cell profile within the tumor microenvironment with each treatment, the expression of cancer immunology-related genes of the tumors was analyzed using the NanoString nCounter PanCancer Immune Profiling. Gene clusters involved in immune cell functions, including adaptive immune cells, T cell function, and interferon responses, were found to be upregulated in the distal tumors of mice treated with reovirus combined with ADU-S100 compared to those in control (Fig. 6A). In contrast, a remarkable upregulation of cluster genes related to immune functions could not be detected in the distal tumors in the mice with reovirus or ADU-S100 single treatment. We also identified tumor-infiltrating lymphocytes using the NanoString nCounter data analysis and found that cytotoxic cells and CD8 cells scores were upregulated in the distal tumor in mice treated with the combination of reovirus and ADU-S100 compared to the distal tumor in mice treated with reovirus or ADU-S100 alone (Fig. 6B). These results imply that the enhanced anti-tumor activity in the tumor with a combination treatment reovirus and ADU-S100 was induced by accumulation of cytotoxic immune cells within tumor microenvironment.
To examine the profile of immune cells in the tumor microenvironment that might promote enhanced anti-tumor immunity in the combination group, we performed immunohistochemical staining of tumors treated with reovirus alone or in combination with ADU-S100. The proportions of M1 and M2 tumor-associated macrophages (TAMs) infiltrated in tumors were evaluated by determining M1 marker inducible nitric oxide synthesis (iNOS) and the M2 marker arginase-I with immunohistochemical staining. The expression of iNOS was not significantly upregulated in the tumor tissue of mice treated with the reovirus alone or in combination with ADU-S100 (Fig. 7A). The expression of arginase I was significantly downregulated in all treatment groups as compared with controls, and we detect the significant difference in the expression of arginase I between the mice treated with the reovirus alone and the mice treated with reovirus in combination with ADU-S100. (Fig. 7B). From these results, the polarization of TAMs seems to be partially involved in the anti-tumor immunity enhanced by the combination treatment. To further verify the immunotherapeutic effect of the T cell infiltration in tumor tissues of mice receiving the combination treatment, we also performed the tumor-infiltrating lymphocytes profiling in the tumor tissues of mice with the treatment. The number of CD8+ T cells was significantly increased in the treated and untreated tumors in all treatment groups compared with controls (Fig. 7D). By contrast, decreased numbers of CD4+ T cells were observed in the treated and untreated tumors of all treated groups (Fig. 7C). Next, we investigated the profile of immune cells in the tumor microenvironment using fluorescence-activated cell sorting (FACS). Tumor samples were collected and dissociated 3 days after IT injection of reovirus with or without pretreatment with ADU-S100, and subjected to FACS analysis. Significantly lower CD4:CD8 ratios of T cells were observed in the treated and untreated tumors from CT26 tumor-bearing mice treated with the combination reovirus and ADU-S100 IT injection compared with untreated controls, whereas no significant differences in the CD4:CD8 ratio were observed between the control and other treatment groups (Fig. 7E). We detect a significant reduction in the ratio of Foxp3+CD45+CD4+CD3+ regulatory T cells in tumors treated with reovirus alone or the combination reovirus and AUD-S100 compared to the control (Fig. 7F). Taken together, local treatment with reovirus in combination with a STING agonist increased the levels of infiltrating CD8+ T cells in the treated tumor and untreated distal tumor. The significant tumor regression and survival benefit observed in combination treated mice were mediated by an inflamed tumor microenvironment associated with the increased numbers of infiltrating cytotoxic T cells.