Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused a global pandemic, resulting millions of infections and deaths with few effective interventions available. Here, we demonstrate that SARS-CoV-2 evades interferon (IFN) activation in respiratory epithelial cells, resulting in a delayed response in bystander cells. Since pretreatment with IFNs can block viral infection, we reasoned that pharmacological activation of innate immune pathways could control SARS-CoV-2 infection. To identify potent antiviral innate immune agonists, we screened a panel of 75 microbial ligands that activate diverse signaling pathways and identified cyclic dinucleotides (CDNs), canonical STING agonists, as antiviral. Since CDNs have poor bioavailability, we tested the small molecule STING agonist diABZI, and found that it potently inhibits SARS-CoV-2 infection of diverse strains including variants of concern (B.1.351) by transiently stimulating IFN signaling. Importantly, diABZI restricts viral replication in primary human bronchial epithelial cells and in mice in vivo. Our study provides evidence that activation of STING may represent a promising therapeutic strategy to control SARS-CoV-2.
INTRODUCTION
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019 (COVID-19), is a novel coronavirus that emerged in Wuhan, China in December 2019 (1). As a highly transmissible and pathogenic virus, SARS-CoV-2 rapidly spread around the world, causing a pandemic that poses a significant threat to global public health. As of January 2021, SARS-CoV-2 has infected more than 100 million people and resulted in more than 2 million deaths worldwide (2). Therefore, the development of effective antivirals is urgently needed for controlling SARS-CoV-2 infection and disease.
SARS-CoV-2 initially targets respiratory epithelial cells (1, 3). As the first line of defense against infection, the host innate immune system within the respiratory tract recognizes viral pathogens by detecting their pathogen-associated molecular patterns (PAMPs) through host-encoded pattern recognition receptors (PRRs) (4). Viral recognition by PRRs activates downstream signaling cascades, leading to the induction of effectors including type I and type III interferons (IFNs) (5). IFNs subsequently bind to their specific receptors in an autocrine and paracrine manner to activate JAK/STAT signaling and up-regulate the expression of hundreds of interferon-stimulated genes (ISGs), conferring an antiviral state to the host cells. Therefore, activation of IFN and other innate immune pathways can potentially block infection. Indeed, there are clinical trials underway to treat COVID-19 with IFNs with mixed success (6, 7)
We set out to characterize the innate response to SARS-CoV-2 and found that there was delayed activation due to immune evasion. This suggested that there may be additional innate immune agonists that could block SARS-CoV-2 infection, and perhaps small molecules with drug-like properties would have advantages over the protein IFN. To identify antiviral innate immune agonists to block viral infection, we performed high throughput screening and identified endogenous STING agonists, cyclic dinucleotides (CDNs), as antiviral against SARS-CoV-2. STING activation leads to the induction of IFNs as well as inflammatory NF-κB signaling and can inhibit diverse viral infections (8). While CDNs have low potency and make poor drugs, potent small molecule STING agonists have been developed including diABZI (9). We found that diABZI was active against SARS-CoV-2 in primary human respiratory epithelial cells and in vivo in two different mouse models of infection. We also found that we could treat infection therapeutically against diverse strains of SARS-CoV-2 suggesting that this STING agonist may serve as a novel therapeutic strategy to combat COVID-19.
RESULTS
SARS-CoV-2 induces immune activation at late time points in human respiratory epithelial cells
Although replication of SARS-CoV-2 in the respiratory tract stimulates innate immunity, the responses are attenuated compared to those observed after infection with other respiratory viruses (1, 10). Indeed, using RNAseq, we found that human lung epithelial Calu-3 cells show few transcriptional changes at 24 hours post-infection (hpi), whereas at 48 hpi 988 genes are induced (adjusted p 1, avg reads > 20 across all samples). Gene ontology (GO) enrichment analysis reveals that genes associated with IFN and antiviral defense are induced at the 48 hours time point (Fig. 1A and S1A). We validated the induction of type I (IFNβ) and III (IFNλ1) IFNs, as well as ISGs (e.g., IFIT1, TRIM22, and MX2) during infection by RT-qPCR (Fig. 1B to F). In addition, we validated that proinflammatory cytokines and chemokines (e.g., IL-6, CXCL10 and TNFα) are induced in SARS-CoV-2-infected Calu-3 cells by RT-qPCR (Fig. 1G to I). We also observed that SARS-CoV-2 induced type I IFNs, type III IFNs and ISG expression in primary normal human bronchial epithelial (NHBE) cells at late time points post infection (Fig. 1J to L).
(A) Calu-3 cells were mock-infected or infected with SARS-CoV-2 for 24 or 48 hours in 3 independent experiments. Total RNA was extracted and subjected to RNAseq analysis. Heat map shows the genes with greater than 1 log2 fold change, an adjusted P value less than 0.05, and average read number greater than 20. (B to I) Calu-3 cells were either uninfected (Uninf.) or infected with SARS-CoV-2 (MOI of 0.5) for 24, 36, or 48 hours. mRNA expression of IFNβ (B), IFNλ1 (C), IFIT1 (D), TRIM22 (E), MX2 (F), IL-6 (G), CXCL10 (H), and TNFα (I) was examined by RT-qPCR. Gene expression (gene/18S) was normalized to uninfected cells. (J to L) NHBE cells were either uninfected (Uninf.) or infected with SARS-CoV-2 (MOI of 0.5) for 24, 48 or 72 hours. mRNA expression of IFNβ (J), IFNλ1 (K) and IFIT1 (L) was quantified by RT-qPCR. Gene expression (gene/18S) was normalized to uninfected cells. (M) Calu-3 cells were uninfected (Uninf.) or infected with SARS-CoV-2 (MOI of 0.5) for the indicated time points. Cells were lysed, and the protein expression level was determined by immunoblotting using indicated antibodies. Representative blots of 3 independent experiments are shown. (N and O) Calu-3 cells were uninfected or infected with SARS-CoV-2 (MOI of 0.5) for 48 hours. Cells were fixed and stained with antibodies against IRF3 (N) or p-TBK1 (O). Representative images of 3 independent experiments are shown. Scale bar: 20 μm. (P and Q) Calu-3 cells were uninfected (Uninf.) or infected with SARS-CoV-2 (MOI of 0.5) for 24 hours. The uninfected or infected cells were then treated with Sendai virus (SeV) for 8 hours (P) or poly(I:C) for 6 hours (Q). The levels of IFNβ mRNA were quantified by RT-qPCR. (R) Calu-3 cells were uninfected (Uninf.) or infected with SARS-CoV-2 (MOI of 0.5) for 24 hours. The uninfected or infected cells were treated with IFNβ for 8 hours. The expression of IFIT1 were analyzed by RT-qPCR. (S) Calu-3 cells were untreated or pretreated with 10 ng/mL of IFNβ or IFNλ1 for 1 hour and inoculated with SARS-CoV-2 (MOI of 0.2) for 48 hours. Viral replication was quantified by RT-qPCR. For all graphs, shown is the mean ± SEM for 3 independent experiments. The significance was calculated using one-way ANOVA and is indicated by (*P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001; ns, not significant).
Since induction of ISGs was apparent at 48 but not 24 hpi in Calu-3 cells, we hypothesized there might be early evasion of canonical innate immune pathways. To evaluate this, we examined the activation status of diverse PRR and innate signaling pathways in Calu-3 cells. We monitored the canonical cytoplasmic viral RNA sensing pathway including the RIG-I/MDA5 receptors (RLRs), the adapter MAVS, and downstream signaling molecules TBK1 and IRF3 (Fig. 1M). We found that SARS-CoV-2 infection has a modest stimulatory effect on the expression of RIG-I, MDA5 and MAVS, but phosphorylation of TBK1 and IRF3 is more strongly induced, but again only at late time points after infection (Fig. 1M). Consistent with late activation of IRF3, the interferon-stimulated gene (ISG) TRIM22 is also induced at 48 hpi (Fig. 1M).
We also examined the expression of the DNA sensor cGAS that produces cyclic dinucleotides that activate STING (11–13). Although the cGAS-STING pathway has a key role in immune recognition of DNA viruses, it also has been found to play roles in RNA virus infections (14). SARS-CoV-2 infection leads to mild activation of STING as measured by phosphorylated STING at later stages of infection, but no change in cGAS was observed (Fig. 1M). Furthermore, STING does not have constitutive antiviral activity in Calu-3 cells, as treatment with the STING covalent antagonist H-151 did not affect infection as measured by microscopy or RT-qPCR (Fig. S1C and D) (15).
Inflammatory NF-κB signaling also was induced at 48 hpi, as measured by increased phospho-p65, which is consistent with the up-regulation of canonical targets, including TNFα (Fig. 1I and M). Taken together, these data suggest that SARS-CoV-2 infection elicits a delayed immune response in respiratory epithelial cells.
SARS-CoV-2 evades IFN activation but can be controlled by IFN treatment
To further explore how SARS-CoV-2 infection impacts innate immune signaling we monitored SARS-CoV-2-dependent activation of IRF3 and TBK1 at the single cell level using confocal microscopy. We observed virus-induced activation and nuclear accumulation of IRF3 at 48 hpi, but this was principally in uninfected bystander cells (Fig. 1N). Similarly, the up-regulation of phosphorylated TBK1 at 48 hpi occurred primarily in uninfected cells (Fig. 1O). To determine if this delayed response was due to delayed replication kinetics, we also monitored viral infection using an antibody to the viral replication intermediate dsRNA, which is also a potent immune stimulus, at an early time point (24 hpi). We detected robust viral dsRNA accumulation without apparent IRF3 nuclear translocation, further suggesting virus evades early immune detection (Fig. S1B). We confirmed that bystander activation of innate signaling was not specific to Calu-3 cells. We also observed virus-induced IRF3 translocation of bystander cells in the human respiratory cell line A549 that ectopically expresses ACE2 (Fig. S1E).
The lack of induction of ISGs early in infection could be due to viral antagonism of innate signaling pathways (16). We tested this hypothesis by inducing innate signaling using canonical ligands during SARS-CoV-2 infection. We included two stimuli that are known to induce IFNβ through RLR signaling: Sendai virus (SeV), and transfected poly(I:C). If SARS-CoV-2 antagonizes RLR signaling then we expect that the SeV or poly(I:C) induced IFN induction will be reduced in SARS-CoV-2-infected cells. Therefore, Calu-3 cells were either uninfected or infected with SARS-CoV-2 for 24 hours and then either co-infected with SeV (Fig. 1P) or transfected with poly(I:C) (Fig. 1Q). We observed similar levels of IFNβ induction by SeV or poly(I:C) in uninfected and co-infected cells; thus, SARS-CoV-2 does not antagonize RLR-dependent IFNβ mRNA induction. We also tested whether SARS-CoV-2 antagonizes IFN signaling and downstream ISG production. For these studies we treated cells with IFNβ and monitored IFNβ-induced up-regulation of ISGs IFIT1 and TRIM22. We found that the IFNβ-induced levels of ISG mRNAs were similar in uninfected and SARS-CoV-2-infected cells (Fig. 1R and S1F). These data suggest that SARS-CoV-2 is evading but not antagonizing PRR detection.
Next, we monitored this evasion using confocal microscopy which allowed us to assess the activation status of IRF3 in infected cells and uninfected bystander cells. We found that IRF3 nuclear translocation induced by SeV occurs in both uninfected and SARS-CoV-2-infected cells (Fig. S1G). This demonstrates that SeV-induced IRF3 activation is not antagonized by SARS-CoV-2. Again, we confirmed that this was not specific for Calu-3 cells as we observed that SeV induced IRF3 translocation in SARS-CoV-2 infected A549-ACE2 cells (Fig. S1H). Altogether, these data suggest that SARS-CoV-2 evades early recognition by canonical innate immune pathways.
Since SARS-CoV-2 was evading recognition, this suggested that treatment with interferons would induce antiviral gene expression and block SARS-CoV-2 infection. Whereas virtually all cells respond to type I IFNs, epithelial cells also respond to type III IFNs (17, 18). Therefore, we tested whether pretreatment of Calu-3 cells with different concentrations of type I IFNβ, or type III IFNλ1 would impact SARS-CoV-2 infection. We monitored both the percentage of infected cells and cell viability at 48 hpi using automated microscopy. Consistent with other studies (19, 20), IFNβ potently blocked SARS-CoV-2 infection at low concentration with an EC50 < 0.03 ng/mL and no apparent toxicity (Fig. S1, I and K). IFNλ1 is less active than IFNβ, and never fully blocked infection, even at high concentrations (Fig. S1, J and K). We confirmed the stronger antiviral activity of IFNβ than IFNλ1 by RT-qPCR (Fig. 1S). Since proteins such as IFNs are difficult to produce at scale for therapeutic use, we set out to identify additional drug-like molecules that could activate innate antiviral immunity.
High throughput screening identified STING agonists as antivirals against SARS-CoV-2
Indeed, we reasoned that additional innate immune agonists or pathogen-associated molecular patterns (PAMPs) could be used to suppress viral replication in respiratory cells. We screened a library of 75 agonists targeting diverse PRR sensing pathways in Calu-3 cells. These agonists include Toll-like receptor (TLR) agonists, RIG-I-like receptor (RLR) agonists, NOD-like receptor agonists, C-type lectin-like receptor agonists and cyclic dinucleotides (CDNs) (Table S1). We examined their effects on viral infection using automated microscopy and identified nine candidates that suppressed infection more than 10-fold (Fig. S2A). This included two CDNs (2’2′-cGAMP and 2’3′-cGAMP), four TLR7 agonists (imiquimod, gardiquimod, loxoribine and adilipoline) and HMW poly(I:C) (Fig. 2A, S2A, and Table S1). We performed dose-response studies with eight of the nine agonists and found that only the CDNs that activate STING (2’2′-cGAMP and 2’3′-cGAMP) and HMW poly(I:C) were active (Fig. 2B, S2A to I). We confirmed the potency of the canonical STING ligand produced by cGAS 2’3′-cGAMP using RT-qPCR, which showed a greater than 100-fold reduction in viral RNA levels (Fig. 2C).
(A) Calu-3 cells were treated with 75 innate immune agonists targeting different PRR sensing pathways for 2 hours, followed by inoculation with SARS-CoV-2 (MOI of 0.5). At 48 hpi, cells were fixed and stained for dsRNA and nuclei, followed by imaging with an automated microscope. Z-scores were calculated to quantify viral infection relative to vehicle controls. The two CDNs are labeled in red. (B) Dose-response analysis of Calu-3 cells treated with 2’,3′-cGAMP at the indicated concentrations and infected with SARS-CoV-2 (MOI of 0.5) for 48 hours. Viral infection and cell viability were quantified and normalized to vehicle control. (C) Untreated or 2’,3′-cGAMP-treated (100 μg/mL) Calu-3 cells were inoculated with SARS-CoV-2 (MOI of 0.2) for 48 hours. Viral RNA was monitored by RT-qPCR. Shown is the mean ± SEM for 3 independent experiments. Significance was calculated using an unpaired, two-tailed Student’s t test (*P < 0.05). (D) Calu-3 cells treated with serial dilutions of diABZI were infected with SARS-CoV-2. At 48 hpi, cells were stained with antibody against dsRNA and nuclei, and imaged. Percentage of infection and cell viability relative to vehicle control are shown. EC50, CC50 and SI were quantified. (E) Calu-3 cells were pretreated with DMSO or 10 μM diABZI for 1 hour, followed by inoculation with SARS-CoV-2 (MOI of 0.2) for 48 hours. Viral RNA was quantified using RT-qPCR. Shown is the mean ± SEM for 3 independent experiments. Significance was calculated using an unpaired, two-tailed Student’s t test (**P < 0.01). (F) Calu-3 cells were uninfected (Uninf.) or infected with SARS-CoV-2 (MOI of 0.5) for 24 hours. The uninfected or infected cells were treated with diABZI for 6 hours. The induction of IFIT1 were analyzed by RT-qPCR. Shown is the mean ± SEM for 3 independent experiments. Significance was calculated using one-way ANOVA (*P < 0.05; ns, not significant). (G) Calu-3 cells were treated either with DMSO or 10 μM diABZI for 2, 4, or 6 hours. Immunoblotting was performed to assess the protein expression levels with the indicated antibodies. Representative blots of 3 independent experiments are shown. (H and I) Calu-3 cells were treated with DMSO or 10 μM diABZI for 0.5 hours. Cells were fixed and stained with antibodies against p-STING (H) or IRF3 (I). Images shown are representative of 3 independent experiments. Scale bar: 20 μm.
CDNs are negatively charged molecules that do not readily accumulate in the cell cytoplasm; therefore, CDNs are less potent in activating STING than newly developed small molecule agonists (21). Because of increased potency and drug-like properties, we next tested the activity of a non-nucleotide STING agonist diABZI on SARS-CoV-2 infection (Fig. S2J) (9). The dose-response analysis showed a dramatic inhibition of SARS-CoV-2 infection in diABZI-treated cells with a nanomolar EC50 and low toxicity (Fig. 2D). diABZI treatment led to a ~1,000-fold reduction in viral RNA levels as measured by RT-qPCR (Fig. 2E). We hypothesized that diABZI inhibits infection post-entry and indeed found that when diABZI was added to Calu-3 cells either 1 hour prior to infection (T = -1 hour) or 3 hours post infection (T = 3 hours) which is post entry, we observed a similar level of antiviral activity (Fig. S2K). Since we found that STING activation blocks infection, we also tested if SARS-CoV-2 antagonizes STING signaling. Compared to diABZI-treated uninfected cells, diABZI-dependent up-regulation of IFIT1 and IFNλ1 was similar in uninfected and SARS-CoV-2-infected cells (Fig. 2F and S2L). Therefore, SARS-CoV-2 does not antagonize STING signaling.
We verified that treatment with diABZI results in activation of STING in respiratory epithelial cells. A 2-hours treatment with diABZI results in rapid accumulation of a higher molecular weight species consistent with activation and phosphorylation, which we confirmed by using antibodies that recognize phosphorylated forms of STING (Fig. 2G). Phosphorylation of STING (Ser366) leads to both activation and subsequent degradation (22). Indeed, we observed the reduced expression of phosphorylated STING at 4 and 6 hours after diABZI treatment along with reductions in the higher molecular weight species and overall levels of STING over time (Fig. 2G). STING is known to activate TBK1, IRF3, and NF-κB signaling. Indeed, we detected increased phosphorylation of TBK1 and IRF3 at 2 hours post treatment that wanes over time closely mirroring the activation status of STING. Moreover, we found modest increases in NF-κB signaling as measured by phospho-p65 at 2 hours (Fig. 2G). We confirmed activation of STING by diABZI using confocal microscopy. STING activation and phosphorylation led to perinuclear accumulation and we observed phosphorylation and accumulation at perinuclear foci 30 min after diABZI treatment (Fig. 2H) (23). In addition, we observed diABZI-dependent activation of IRF3 as measured by its nuclear accumulation (Fig. 2I). Collectively, these data suggest diABZI activates the STING-TBK1-IRF3 signaling pathway in human respiratory Calu-3 epithelial cells.
STING activation potently and transiently induces innate signaling pathways
Activation of STING and its downstream signaling pathways induce antiviral gene programs that control infection (24). To determine the genes induced by diABZI, we performed RNAseq on Calu-3 cells treated for 6 or 12 hours. diABZI treatment leads to the robust activation of 403 genes, of which more than 146 are associated with antiviral IFN pathways (Fig. 3A and S3A). Indeed, we observed a remarkable induction of type I and III IFNs and ISGs after diABZI treatment (Fig. 3, B to H). Many classical ISGs including genes encoding OAS family proteins (OAS1, OAS2, OAS3, OASL) that are known human susceptibility/resistance loci are up-regulated in diABZI-treated cells (Fig. 3, I to K and S3B) (25, 26). diABZI treatment also causes a mild induction of NF-κB target gene TNFα (Fig. S3C).
(A) Calu-3 cells were treated with DMSO or 10 μM diABZI for 6 or 12 hours in two independent experiments. Total RNA was isolated and prepared for RNAseq analysis. Heatmap shows the differentially expressed genes with greater than 1 log2 fold change, an adjusted P value less than 0.05, and average read number greater than 20. (B to K) Calu-3 cells were treated with DMSO or 10 μM diABZI for the indicated time points. Total RNA was extracted and the induction of IFNβ (B), IFNλ1 (C), IFIT1 (D), TRIM22 (E), MX2 (F), IL6 (G), CXCL10 (H), OAS1 (I), OAS2 (J), and OAS3 (K) was determined using RT-qPCR. Shown is the mean ± SEM for 3 independent experiments. Asterisks indicate statistical significance as compared with DMSO-treated cells. (L to N) Calu-3 cells were incubated with 10 μM diABZI, 10 ng/mL IFNβ or IFNλ1 for the indicated time points. RNA was extracted and the expression of IFIT1 (L) or TRIM22 (M) mRNA was analyzed by RT-qPCR. Gene expression at 0 hour was set to 1. Shown is the mean ± SEM for 3 independent experiments. (N) Expression of IFIT1 and TRIM22 proteins was examined by Western blotting compared to tubulin control. Representative blots were shown. For all graphs, significance was calculated using one-way ANOVA (*P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001).
Moreover, we compared the kinetics of gene expression between IFNs and diABZI. Both diABZI and IFNβ induce ISGs (IFIT1, TRIM22, MX2) with maximal activation within 6 or 12 hours of treatment (Fig. 3, L and M, and S3D). However, IFNβ-induced ISGs expression is more sustained, whereas diABZI transiently up-regulates ISGs, followed by a more rapid decline. We confirmed the distinct kinetics of IFIT1 and TRIM22 by immunoblot where again both diABZI and IFNβ induce protein accumulation but the activation by diABZI is more transient (Fig. 3N). We also monitored expression of two NF-κB gene targets (CXCL10 and TNFα) and found transient activation by diABZI and IFNβ (Fig. S3, E and F). Consistent with its lack of antiviral activity, IFNλ1 has a minimal effect on the induction of ISGs or NF-κB targets that were profiled (Fig. 3, L and M, and S3, D to F).
STING activation blocks SARS-CoV-2 in human primary respiratory cells and in vivo
We next examined the antiviral activity of diABZI in primary normal human bronchial epithelial (NHBE) cells. NHBE cells were cultured either in submersion culture (NHBE-Submersion) or in an air-liquid interface (NHBE-ALI). Treatment with diABZI in NHBE-Submersion restricts SARS-CoV-2 replication, with a similar magnitude as remdesivir, an inhibitor of SARS-CoV-2 RNA replication (Fig. 4A). In the air-liquid interface culture system, which more closely mimics the human respiratory tract, both type I and III IFNs inhibit viral infection, although type III IFN is less active (Fig. 4D). Furthermore, treatment with diABZI blocked infection (~1,000-fold), similar to type I interferon treatment in these ALI cultures (Fig. 4, B to D, and S4).
(A) Primary normal human bronchial epithelial (NHBE) cells cultured in submersion culture (NHBE-Submersion) were treated with diABZI (10 μM) or remdesivir (10 μM), followed by infection of SARS-CoV-2 (MOI of 0.2). Viral RNA was quantified 72 hpi using RT-qPCR. Graphs show the mean ± SEM compared to DMSO-treated cells for 3 independent experiments. Significance was calculated using one-way ANOVA and is indicated by ***P < 0.001 as compared with DMSO-treated cells. (B and C) NHBE cells cultured in an air-liquid interface (NHBE-ALI) were mock-treated, or treated with DMSO or diABZI (10 μM) and infected with SARS-CoV-2 (MOI of 0.2). 72 hpi cells were then fixed and stained with SARS-CoV-2 Spike and nucleocapsid antibodies. Confocal microscopy was used to image the whole insert. Representative images are shown in (B) and quantified in (C) for two independent experiments. Significance was calculated using one-way ANOVA and is indicated by ***P < 0.001 as compared with DMSO control. (D) NHBE-ALI cells were pretreated with diABZI (10 μM), IFNβ (10 ng/mL) or IFNλ1 (10 ng/mL) and challenged with SARS-CoV-2 (MOI=0.2). RT-qPCR was used to quantify the level of viral RNA 72 hpi. Graphs show the mean ± SEM compared to DMSO-treated cells for 4 independent experiments. Significance was calculated using one-way ANOVA (****P < 0.0001) as compared with DMSO-treated cells. (E to G) Wild type C57BL/6 mice were treated with vehicle (PBS) or 10μg diABZI intranasally. At 6 or 12 hours post-treatment, lung tissues were collected and homogenized, followed by RNA extraction and RNAseq analysis. (E) Heatmap shows gene expression profile upon PBS or diABZI treatment of genes with a log2 fold change greater than 1 in at least one condition, an adjusted P value less than 0.05, and average read number greater than 20. (F and G) The expression of mouse Ifit1 was confirmed by RT-qPCR at 6 hours (F) or 12 hours (G) post-treatment (n = 5, Mann-Whitney test, is indicated by *P < 0.05; **P < 0.01). (H to K) K18-hACE2-transgenic mice were treated with vehicle control or 10μg diABZI via intranasal route 1 day prior (D-1) to infection and the day of infection (D0). Mice were inoculated via intranasal route with 103 PFU of SARS-CoV-2. (H) Body weight change of vehicle- or diABZI-treated mice was monitored daily (two independent experiments; from Day -1 to Day 3, n = 12 for Vehicle, n = 13 for diABZI; from Day 4 to Day 7, n = 8 for both Vehicle and diABZI; two-way ANOVA; P values: ***P = 0.0003 and ****P < 0.0001). At 3 and 7 dpi, viral burden in the lung tissues was determined by RT-qPCR for viral RNA (I and J) (n = 4 for Vehicle, n = 5 for diABZI; Mann-Whitney test; P value: *P = 0.0159 (I); n = 8; Mann-Whitney test; P value: ***P = 0.0002 (J)) (K) Viral load in the nasal wash was examined by RT-qPCR at the indicated time points (Day 3, n = 4 for Vehicle, n = 5 for diABZI; Day 7, n = 6 for Vehicle, n = 8 for diABZI; Mann-Whitney test; P values: **P = 0.0027 and *P = 0.0159). (L and M) K18-hACE2-transgenic mice were treated with vehicle control or 10 μg diABZI via intranasal route 1 day prior (D-1) to infection and the day of infection (D0) or 6 hours prior to infection (-6 hours). Survival rates (L) and body weight changes (M) of vehicle- or diABZI-treated mice were monitored daily (from Day -1 to Day 7, n = 8 for Vehicle (D-1/D0); from Day -1 to Day 8, n = 8 for Vehicle (-6h); from Day -1 to Day 14, n = 9 for diABZI (D-1/D0); from Day -1 to Day 14, n = 8 for diABZI (-6h). Significance in (L) was calculated using Log-rank (Mantel-Cox) test. P values: ***P = 0.0005; ****P < 0.0001. (N) K18-hACE2-transgenic mice were treated with vehicle control or indicated dose of diABZI at D0 and D-1 via intranasal route, followed by viral infection. Body weight change was monitored daily (n = 5 for both Vehicle and diABZI).
To further explore the utility of STING agonists in vivo, we evaluated whether intranasal treatment of mice with diABZI induces an antiviral gene expression program. RNAseq analysis revealed a rapid activation of IFN signaling pathways in the lung after a single intranasal administration (Fig. 4, E to G, and S5, A to M). By comparing orthologs of genes induced by diABZI in Calu-3 human respiratory epithelial cells and mouse lungs, we found striking overlap suggesting a robust and common gene expression program across models (Fig. S6, A to D).
We next set out to determine if diABZI treatment in vivo was protective. K18 hACE2-transgenic mice express the human angiotensin-converting enzyme 2 (ACE2), a host cellular receptor for SARS-CoV-2 entry, and support high levels of SARS-CoV-2 infection in lung tissues, resulting in intense innate and adaptive immune cell infiltration and respiratory dysfunction (27). Using the K18-hACE2 transgenic mice, we evaluated the effect of diABZI on viral infection in vivo. Mice were pretreated with diABZI via intranasal route at day (D)-1, and given a second dose at the time of inoculation (D0) of SARS-CoV-2. At day 7 post-infection (dpi), whereas vehicle-treated mice had lost 25% of their body weight, diABZI-treated mice show much less weight loss (Fig. 4H). We did observe an early phase of weight loss (1 and 2 dpi) in diABZI-treated mice, which could reflect the induction of innate immune responses. Viral RNA levels in the lungs and nasal washes were reduced at 3 and 7 dpi (Fig. 4, I to K). We also quantified cytokine and chemokine proteins in mouse lung tissue at day 3 post infection comparing vehicle and diABZI treatment groups. As expected from our profiling studies, IFNs are induced by diABZI treatment as well as cytokines and chemokines involved in activation and recruitment of neutrophils (Fig. S5N). And there is specificity as a number of other cytokines and chemokines remain unchanged (Fig. S5O). These data further support that diABZI stimulates IFN for protective immunity.
Using the K18-hACE2 transgenic mice, we also evaluated whether diABZI can protect the animals from lethality and whether we could mitigate the weight loss observed upon diABZI treatment by reducing the dosing schedule to a single intranasal delivery of diABZI. Mice were pretreated with diABZI via intranasal route at day (D)-1, and given a second dose at the time of inoculation (D0) of SARS-CoV-2 or treated with one single dose at 6 hours prior to infection (-6 hours). Mice treated with diABZI with either regimen were protected from lethality (Fig. 4L). Moreover, animals treated only once had reduced weight loss early, and completely recovered at later time points (Fig. 4M). Since a single dose of 10 μg was protective, we also tested if we could reduce the dose of diABZI. We found that treatment with a single dose of 10 μg, 3 μg or 1 μg of diABZI delivered intranasally was protective from virus-induced weight loss and led to long-term survival (Fig. 4N). We also monitored lung histology comparing vehicle-treated mice to mice treated with a single 3 μg dose of diABZI delivered at the time of infection (Fig. S5P). Vehicle-treated infected lungs showed that immune cells, including neutrophils and mononuclear cells, infiltrated and accumulated throughout the lung in alveolar and interstitial locations with alveolar wall thickening. In contrast, mice treated with diABZI showed a marked attenuation of the inflammatory response in the lung with very few confined spots of cellular infiltration. Therefore, diABZI treatment decreases viral replication and lung inflammation.