We previously reported that systemic administration of mHAdLyp.sT in immunodeficient nude mice resulted in reduced uptake in the liver and spleen, reduced hepatotoxicity and systemic toxicity, and attenuated innate immune response [6]. In this study we used immunocompetent BALB/c mice bearing 4T1 TNBC tumors to determine viral tropism, toxicity, and immune responses by the systemic administration of mHAdLyp.sT. First of all, liver samples collected 48 hours after adenovirus injection were evaluated for viral genomic DNA copy numbers. Unlike the control vectors lacking hexon modification for liver-de-targeted tropism (AdLyp.sT, Ad.sT and Ad(E-).null), mHAdLyp.sT treatment didn’t lead to a significant increase of liver viral genomic DNA copy numbers compared to the buffer group (Fig. 1A). mHAdLyp.sT treatment also did not significantly increase viral uptake in liver on day 25, although a significant increase of liver viral DNA remained in the Ad.sT and Ad(E-).null groups (Fig. 1B). Thus, we concluded that mHAdLyp.sT had reduced liver uptake in immunocompetent mice as well. We further evaluated the tumor viral uptake by qRT-PCR of viral genome and sTGFβRIIFc expression. We observed significant viral genome tumor expression for all of our adenoviruses (AdLyp.sT, mHAd.sT and Ad(E-).null vs buffer: P < 0.05; mHAdLyp.sT and Ad.sT vs buffer: P < 0.01) in day 14 tumor samples (Fig. 1C), although these changes were no longer observed in day 25 tumor samples (Fig. 1D). We also found that mHAdLyp.sT and Ad.sT treatment led to a significant increase in sTGFβRIIFc mRNA expression when compared to the Ad(E-).null treated group (mHAdLyp.sT: P < 0.01; Ad.sT: P < 0.05) in day 14 tumor samples (Fig. 1E). In addition, a significant increase of blood sTGFβRIIFc protein expression was observed in serum samples on day 14 for mHAdLyp.sT, mHAd.sT, and Ad.sT (Fig. 1F, P < 0.05, 0.01, or 0.001 vs Ad(E-).null, respectively). These data suggest that mHAdLyp.sT maintained tumor tropism in 4T1 mouse TNBC tumors but had reduced liver uptakes when administered systemically.
We used mouse sera obtained 48 hours after adenovirus injection to examine the short-term hepatotoxicity, systemic toxicity, and systemic inflammatory responses in this 4T1 syngeneic mouse model. The quantification of serum LDH, ALT, and AST levels suggested that the systemic administration of mHAdLyp.sT and another adenovirus containing Ad5/48 chimeric hexon (mHAd.sT) resulted in no significant systemic and hepatic toxicity (Fig. 2A-C). Also, while the replicating adenovirus with Ad5 hexon (Ad.sT) stimulated a significant increase of both Th2 cytokines (IL-4, IL-6, and IL-10) (Fig. 2D-F, Ad.sT vs buffer: P < 0.05 or 0.01) and Th1 cytokines (IL-12p70, IL-2, TNF-α and IFN-γ) (Fig. 2G-J, Ad.sT vs buffer: P < 0.01 or 0.001), mHAdLyp.sT only elicited the significant increase of IL-12p70 and IFN-γ Th1 cytokines (Fig. 2G and 2J, mHAdLyp.sT vs buffer: P < 0.05). For the other two adenoviruses we tested (AdLyp.sT and mHAd.sT), AdLyp.sT is similar to Ad.sT but mHAd.sT seems less effective in eliciting Th1 cytokine response than mHAdLyp.sT as only a significant change in IL-12p70 was detected (Fig. 2G, mHAd.sT vs buffer: P < 0.05). Severe systemic toxicity and inflammatory responses to adenoviruses upon systemic delivery is a major obstacle for their potential clinical application [13]. Thus, mHAdLyp.sT is more likely to be applicable for future clinical trials as it didn’t induce detectable hepatotoxicity and systemic toxicity but maintained some critical anti-tumor Th1 cytokine response such as IL-12p70 and IFN-γ.
We also tested serum GM-CSF and TGFβ-1 levels on day 14 to examine changes in these immunosurveillance molecules where different expression levels can either promote anti-tumor immune responses (GM-CSF) or lead to immunosuppression (TGFβ-1) [29, 30, 10]. All replicating adenoviruses expressing sTGFβRIIFc almost equally prompted GM-CSF production while hindering immune inhibitory TGFβ-1 secretion (Fig. 2K-L). It is not surprising to see reduced TGFβ-1 levels in mouse sera by these therapeutic adenoviruses since they all express sTGFβRIIFc that can bind TGFβ-1 and neutralize its downstream signaling events via Type II receptors, but the increase of serum GM-CSF levels are also encouraging because many GM-CSF based treatment strategies are currently used in the clinic or hold promise in clinical trials for several cancer types, including breast cancer. Additionally, the expression levels of these biomarkers in sera obtained on day 25 were also analyzed, but the results did not support persistent changes (data not shown).
In tumor-bearing mice, spleen is the major immunomodulatory organ and tumor is where all actions are propelled. Therefore, we analyzed the expression levels of the biomarkers above on both day 14 and day 25 samples of spleen and tumor by qRT-PCR. Only mHAdLyp.sT inhibited TGFβ-1 expression in spleen on both day 14 and day 25 (Fig. 3A, day 14; Fig. 3C, day 25, both the first panel, P < 0.05 or 0.01 vs buffer). We also analyzed the expression of other Th2 cytokines besides TGFβ-1 (IL-4 and IL-6) in spleen and tumor samples, but treatment with mHAdLyp.sT didn’t lead to any significant changes when compared to the buffer group (data not shown). For Th1 cytokines of interest (IL-12, IL-2, TNF-α and IFN-γ), in spleen, we detected increased levels of IL-2 and IFN-γ expression by mHAdLyp.sT treatment on day 25 (Fig. 3C, day 25; IL-2, the second panels; IFN-γ, the fourth panels; both P < 0.01 vs buffer). It is interesting that no significant changes of them were observed in the mHAdLyp.sT treatment group in day 14 spleen samples (Fig. 3A, the second to fourth panels), suggesting mHAdLyp.sT may be more likely to have delayed but persistent systemic immunomodulatory effects in spleen. In the tumor microenvironment, mHAdLyp.sT treated group had increased expression of IL-12 on both day 14 and day 25, and IFN-γ on day 25 (Fig. 3B, day 14; Fig. 3D, day 25; IL-12, the third panels; IFN-γ, the fourth panels; P < 0.05 or 0.01 vs buffer). No significant changes of TNF-α in both spleen and tumor was seen at any of our ending points (data not shown), but the localized stimulation of two critical Th1 cytokines (IL-12 and IFN-γ) in tumors by mHAdLyp.sT suggests that mHAdLyp.sT is able to help prime antitumor immunity directly in the TME, specifically with IL-12, which was elevated by mHAdLyp.sT at both early and late stages of tumor resistance.
Next, we profiled the immune cell population changes in blood, spleen, and tumor samples from both day 14 and day 25 samples. On day 14 of blood samples, mHAdLyp.sT treatment led to a significant increase in the percentage of central memory cells (TCM, CD44+CD62L+) among CD8+ T lymphocytes (Fig. 4A, the right panel, P < 0.01 vs buffer), even though the CD8+ proportion of total T cells remained similar in blood (Fig. 4A, the left panel). In spleen and tumor, we observed significantly elevated percentages of CD8+ T lymphocytes by mHAdLyp.sT (Fig. 4B and 4C, the left panels, P < 0.05 vs buffer). The control adenovirus we used in the flow studies, mHAd.sT, only increased the percentage of CD8+ T cells in spleen (Fig. 4B, the left panel). Importantly, in tumor, mHAdLyp.sT treatment also increased the percentage of CD8+ central memory cells (TCM, CD44+CD62L+) significantly on day 14 (Fig. 4C, the right panel, P < 0.05 vs buffer). Interestingly, in spleen, the percentage of CD8+ effector memory cells (TEM, CD44+CD62L−) was raised significantly by both adenoviruses (Fig. 4B, the right panel, P < 0.05 vs buffer). In mouse models, both CD8+ TCM and TEM cells are vital components of the anti-tumor response [31]. Based on these data mHAdLyp.sT may depend on different microenvironments of specific tissues to help CD8+ T cells acquire distinct anti-tumor memory cell characteristics.
We also conducted flow analysis for various myeloid cell populations on day 14 samples. CD86 is a key target for CTLA-4 immune regulation and is important for T cell activation and survival [32]. In blood, the mHAdLyp.sT treated group had a significant increase in CD86+ dendritic cells (DCs) (CD86+CD11C+) on day 14 (Fig. 5A, the left panel, P < 0.05 vs buffer). The increased percentage of DCs was also observed in spleen by mHAdLyp.sT treatment (Fig. 5B, P < 0.05 vs buffer). For myeloid-derived suppressor cells (MDSCs), both adenoviruses led to a decrease of in granulocytic/polymorphonuclear MDSCs (g-MDSCs, Ly6CintLy6G+CD11b+, Fig. 5A, the second panel, P < 0.01 vs buffer) and an increase in monocytic MDSCs (m-MDSCs, Ly6ChiLy6G−CD11b+, Fig. 5A, the third panel, P < 0.05 vs buffer) in blood. Similar changes in MDSCs subpopulations were also detected in tumor samples (Fig. 5C, the first two panels, P < 0.05 vs buffer). Both g-MDSCs and m-MDSCs are immune suppressive. However, mHAdLyp.sT treatment favors predominantly m-MDSCs, which employs nitric oxide (NO) and immunosuppressive cytokines/molecules such as IL-10, TGFβ-1, and PD-L1 to mediate immune suppression [33]. Since mHAdLyp.sT inhibits the TGFβ signaling pathway, a combination therapy with immune checkpoint inhibitors could more effectively inhibit MDSCs related immune suppression, both systemically and in the TME.
Polarizing macrophages towards a pro-inflammatory and tumor-inhibiting M1 phenotype is considered another important sign of immune-inflamed response by potential immunotherapy agents [34]. Both mHAd.sT and mHAdLyp.sT treatment significantly increased M1 macrophage (CD11C+) percentages among F4/80+CD45+ cells in blood and tumor samples (Fig. 5A, blood, the fourth panel; Fig. 5C, tumor, the third panel, P < 0.01 or P < 0.05 vs buffer). Both adenoviruses also significantly reduced the percentage of M2 macrophages (CD206+) in blood (Fig. 5A, the last panel, P < 0.01 or P < 0.05 vs buffer). Noteworthy, mHAdLyp.sT seems to be more effective in macrophage polarization towards a cancer cell-killing phenotype systemically than mHAd.sT since changes in both M1 and M2 macrophages in blood by mHAdLyp.sT were more significant than those by mHAd.sT, when they were compared to the buffer group (Fig. 5A, the last two panels; mHAdLyp.sT vs buffer, P < 0.01; mHAd.sT vs buffer, P < 0.05).
The same set of immune cell analyses were conducted with the Day 25 samples of blood, spleen and tumor. We did not detect any meaningful differences systemically at this late tumor resistant stage (data not shown). However, an immune inflamed TME seems largely retained in adenovirus treated groups, especially for those with mHAdLyp.sT treatment. Within tumors, the percentage of CD8+ T lymphocytes and IFN-γ producing CD8+ T cells were both significantly increased by adenovirus treatments (Fig. 6A, P < 0.05 vs buffer). Furthermore, only mHAdLyp.sT led to a significant increase of m-MDSCs (Ly6ChiLy6G−CD11b+, Fig. 6B, P < 0.05 vs buffer) and M1 macrophage (CD11C+F4/80+, Fig. 6C, P < 0.05 vs buffer) in day 25 tumors. Taken together, our immune cell analysis data in the tumor-bearing immunocompetent mouse model supports mHAdLyp.sT as a potent primer towards the anti-tumor phenotype that could synergize with other immunomodulators to achieve better therapeutic results.
In the past, we showed that systemic administration of mHAdLyp.sT inhibited bone metastases in a human TNBC cell line (MDA-MB-231) immunodeficient mouse model. Therefore, we applied mHAdLyp.sT alone and together with immune checkpoint inhibitors (ICIs) to test their treatment efficacy in this 4T1 immunocompetent model. Our tumor volume analysis indicated that the combination of mHAdLyp.sT, anti-PD-1 and anti-CTLA-4 antibodies (triple treatment) was the only group that significantly inhibited primary tumor progression when compared to the buffer group using a two-way ANOVA analysis at the end of this experiment (Day 23) (Fig. 7A, P < 0.001 vs buffer). This was further supported by tumor weight analysis because the average tumor weight of the triple treatment group was lowest and was significantly different from two other groups by t tests (Fig. 7B, P < 0.05 vs mHAdLyp.sT and mHAdLyp.sT + anti-CTLA-4). Most importantly, all treatment groups, except for anti-PD-1 alone, were almost equally effective in inhibiting lung metastasis by our H&E staining microscopy analysis (Fig. 7C, P < 0.001 or P < 0.0001 vs buffer; Fig. 7D, representative images of H&E-stained lung sections). Although mHAdLyp.sT didn’t alleviate the growth of the primary tumor, no significant difference in inhibiting lung metastasis between mHAdLyp.sT alone and the triple treatment when comparing them directly by t test was observed. Because mHAdLyp.sT is much safer to be used systemically, currently we are working on new treatment experiments with increased doses of mHAdLyp.sT to enhance its anti-tumor activities in this model.