COMPOUNDS FOR THE TREATMENT OF A DISEASE OR DISORDER, METHODS FOR IDENTIFYING SAID COMPOUNDS

Abstract
Disclosed herein are methods of identifying a compound for treating or preventing an infection with an infectious microbe, such as a coronavirus, in a subject in need thereof. Also disclosed herein are compounds and compositions identified by said methods, and methods of use thereof.
Description
BACKGROUND

Covid-19 (coronavirus disease-2019) caused by SARS-CoV-2 (severe acute respiratory syndrome coronavirus (CoV) type 2 virus) has led to over 1.3 million deaths as of mid-November, 2020, due to its high contagiousness and therefore rapid spread. There is an urgent need to develop new therapeutics against Covid-19.


While efforts to target viral proteins are underway, an alternative strategy is to pursue host-targeted therapies. The host cell response is essential to enabling viral entry, endosomal escape, translation, replication, assembly, and release. Host cells are also naturally armed with antiviral programs, which, if properly induced, can constrain the in vivo viral spread within a canonical 4-7 day period, upon sufficient adaptive immunity development.


There is a need for the identification and development of drugs that interact with cell host proteins involved in viral infection and immune response to viral infection. There is a need for the identification and development of drugs that interact with cell host proteins involved in SARS-CoV-2 infection and that protect from SARS-CoV-2 hyperinflammation. There is a need for the identification and development of drugs that inhibit SARS-CoV-2 entry into cells.


The compounds, compositions, and methods disclosed herein address these and other needs.


SUMMARY

In accordance with the purposes of the disclosed materials and methods, as embodied and broadly described herein, the disclosed subject matter, in one aspect, relates to compounds, compositions, and methods of identifying, making, and using compounds and compositions.


For example, disclosed herein are methods of identifying a compound for treating or preventing an infection with an infectious microbe in a subject in need thereof, the methods comprising:

    • a) obtaining transcriptomic data from cells infected with the infectious microbe,
    • b) identifying differentially expressed genes (DEGs),
    • c) characterizing host-targeted antimicrobial or anticytokine signature,
    • d) identifying compounds that stimulate the anti-microbial or -cytokine signature,
    • e) evaluating known and predicted targets of compounds identified in step d),
    • f) constructing an infection host response protein-protein interaction (PPI) network and modules,
    • g) prioritizing compounds based on network proximity analysis,
    • h) clustering of prioritized compounds associated with selected disease modules,
    • i) selecting representative compounds from each cluster for in vitro assays, and
    • j) analyzing the results of steps a-i to thereby identify the compound for treating or preventing the infection.


In some examples, the infectious microbe can comprise a coronavirus.


Also disclosed herein are methods of identifying a compound for treating or preventing a coronavirus infection in a subject in need thereof, the method comprising:

    • a) obtaining transcriptomic data from coronavirus infected cells,
    • b) identifying differentially expressed genes (DEGs),
    • c) characterizing host-targeted antiviral or anticytokine signature,
    • d) identifying compounds that stimulate the anti-viral or -cytokine signature,
    • e) evaluating known and predicted targets of compounds identified in step d),
    • f) constructing a coronavirus infection host response protein-protein interaction (PPI) network and modules,
    • g) prioritizing compounds based on network proximity analysis,
    • h) clustering of prioritized compounds associated with selected disease modules,
    • i) selecting representative compounds from each cluster for in vitro assays, and
    • j) analyzing the results of steps a-i to thereby identify the compound for treating or preventing the infection.


Also disclosed herein are compositions comprising the compound identified by any of the methods disclosed herein. Also disclosed herein are methods of treating a disease or disorder in a subject in need thereof, the method comprising administering to the subject a composition comprising a therapeutically effective amount of the composition comprising the compound identified by any of the methods disclosed herein.


Also disclosed herein are methods of treating or preventing a coronavirus infection in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a composition comprising a compound selected from the group consisting of: imipramine, salmeterol, hexylresorcinol, brompheniramine, ezetimibe, temsirolimus, linsitinib, torin-1, rottlerin, semaxanib, ipratropium, AS-605240, mefenamic acid, JNJ16259685, QL-XII-47, midodrine, olanzapine, trifluoperazine, fluphenazine, azelastine, chlorphenamine, clarithromycin, saracatinib, JAK3-Inhibitor-II, AZD-8055, CGP-60474, hexamethylene, loperamide, nifedipine, liothyronine, atorvastatin, triptolide, pirfenidone, isoliquiritigenin, rucaparib, berbamine, darinaparsin, taurodeoxycholic acid; derivatives thereof; and combinations thereof.


Also disclosed herein are methods of treating or preventing a coronavirus infection in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a composition comprising an antiviral compound and an anti-hyperinflammatory compound.


Also disclosed herein are pharmaceutical compositions for the treatment of a coronavirus infection in a subject in need thereof, wherein the pharmaceutical composition comprises a pharmaceutically acceptable excipient and a therapeutically effective amount of a composition comprising a compound selected from the group consisting of: imipramine, salmeterol, hexylresorcinol, brompheniramine, ezetimibe, temsirolimus, linsitinib, torin-1, rottlerin, semaxanib, ipratropium, AS-605240, mefenamic acid, JNJ16259685, QL-XII-47, midodrine, olanzapine, trifluoperazine, fluphenazine, azelastine, chlorphenamine, clarithromycin, saracatinib, JAK3-Inhibitor-II, AZD-8055, CGP-60474, hexamethylene, loperamide, nifedipine, liothyronine, atorvastatin, triptolide, pirfenidone, isoliquiritigenin, rucaparib, berbamine, darinaparsin, taurodeoxycholic acid; derivatives thereof; and combinations thereof


Also disclosed herein are pharmaceutical compositions for the treatment of coronavirus comprising a pharmaceutically acceptable excipient and a therapeutically effective amount of a composition comprising an antiviral compound and an anti-hyperinflammatory compound


Additional advantages of the disclosed devices and methods will be set forth in part in the description which follows, and in part will be obvious from the description. The advantages of the disclosed devices and methods will be realized and attained by means of the elements and combinations particularly pointed out in the appended claims. It is to be understood that both the foregoing general description and the following detailed description are exemplary and explanatory only and are not restrictive of the disclosed devices and methods, as claimed.


The details of one or more embodiments of the invention are set forth in the accompanying drawings and the description below. Other features, objects, and advantages of the invention will be apparent from the description and drawings, and from the claims.





DESCRIPTION OF FIGURES

The accompanying figures, which are incorporated in and constitute a part of this specification, illustrate several aspects of the disclosure, and together with the description, serve to explain the principles of the disclosure.



FIG. 1. Workflow of the quantitative systems pharmacology approach for selecting compounds for experimental evaluation. Panel A: The RNA-seq data from SARS-CoV-2 infected A549 cells (Blanco-Melo D et al. bioRxiv, 2020, 10.1101/2020.03.24.004655) and ACE2-overexpressing A549 cells were used as input (Blanco-Melo D et al. Cell, 2020, 181, 1036-1045.e1039). Panel B: Up- and down-regulated differentially expressed genes (DEGs) were identified from these data using Wald test with false-discovery rate (FDR) default upper value of 0.05. Panel C: The antiviral gene signature (top) and anti-cytokine gene signature (bottom) were identified upon manual curation of GO enrichment results corresponding to the DEGs, using the QuickGO hierarchical annotation (Binns D et al. Bioinformatics 2009, 25, 3045-3046) (see FIG. 2-FIG. 5 for details). Panel D: Two sets of compounds or repurposable drugs that best reproduced the antiviral and anti-cytokine signatures were extracted from Cmap (Lamb J et al. Science, 2006, 313, 1929-1935; Subramanian A et al. Cell, 2017, 171, 1437-1452.e1417). Panel E: Known and predicted targets of these compounds were identified using QuartataWeb (Li H et al. Bioinformatics, 2020, 36, 3935-3937). Panel F: A host response network composed of four modules related to SARS-CoV-2 infection (called disease modules) was constructed. Panel G: The target of the compounds identified in Panel E and the disease modules in Panel F were subjected to network proximity analysis (Guney E et al. Nat Commun. 2016, 7, 10331) using BioSNAP lung PPI network, to prioritize 25 repurposable or investigational drugs for each module. This step has been performed for antiviral compounds only. Panel H and Panel I: The compounds were clustered based on the interaction patterns with their targets, using QuartataWeb. Representatives from each cluster (Panel H) and additional compounds identified by manual curation were selected for experimental testing (Panel I).



FIG. 2. Illustration of the 4-step pipeline for identifying the intrinsic antiviral signature in A549 cells 24 h after SARS-CoV-2 infection: (1) Identification of 100 upregulated and 20 downregulated genes; (2) GO enrichment analysis for up- and downregulated genes, respectively. The hierarchy of enriched GO terms was generated using QuickGO; (3) Classification of pro- or antiviral GO terms. Upregulated GO terms are classified as either proviral, antiviral, or ambiguous. Downregulated GO terms are all considered as anti-viral; (4) Gene selection for antiviral signature from the classified GO terms. Genes were included if they were antiviral or unknown.



FIG. 3. GO enrichment of up (left) and down (right) regulated genes. GO terms were filtered by size and overlapping genes as described in Materials and Methods. A total of 17 upregulated (Biological Process) and 13 downregulated (Cellular Component) genes are illustrated. P-values were derived from Fisher's one-tailed test and adjusted by Benjamini-Hochberg for multiple test correction.



FIG. 4. Change in the expression levels of 36 genes defining the host-targeted antiviral signature. Log2 fold change at 24-h post-SARS-CoV-2 infection from A549 cells are shown.



FIG. 5. Change in the expression levels of 17 genes defining anti-cytokine signature; log2 fold change at 24 h post-high SARS-CoV-2 infection from A549-ACE2 cells are shown.



FIG. 6. Identification of candidate compounds/drugs, their prioritization and final selection of a small set for experimental tests, illustrated for Dataset 1 (related to FIG. 1). The flow diagram depicts the number of compounds/drugs extracted at various stages, indicated by the Panels A-I (on the left) consistent with FIG. 1 panels A-I. The original analysis of transcriptomics data from A549 cells leads to 36 DEGs, whose antiviral signature screened against Cmap database identifies 263 candidate compounds. Comparison with Excelra DB shows those (10 of them) already listed therein. Of these 263 compounds, 168 have target information available and/or predictable in QuartataWeb (Li H et al. Bioinformatics, 2020, 36, 3935-3937)—an interface that utilizes as input DrugBank and STITCH database. Two different paths are then followed, for the respective subsets of 168 and 95 compounds. In the former case, the targets of these 168 compounds are subjected to network proximity analysis with respect to four disease modules in SARS-CoV-2-host interactome, using BioSNAP human lung PPI network; this analysis yields 64 compounds, which, upon clustering (using QuartataWeb) to select representatives, are reduced to 13 high-priority compounds. The latter set of 95 compounds are manually analyzed to select two compounds, leading to a total set of 15 high priority compounds that have been further investigated in experiments. The diagram depicts the protocol for antiviral compounds. In the case of anti-cytokine compounds, the same schema without panel G is adopted.



FIG. 7. Host cell proteins targeted by potential antiviral (Dataset 1) compounds/drugs, rank-ordered by their promiscuity. Promiscuity refers to the number of predicted compounds/drugs (also called chemicals) that target the protein. FIG. 7 lists the top 100 targets corresponding to Dataset 1 compounds/drugs. The ordinate lists the proteins, and the horizontal bars (abscissa) show the corresponding number of compounds.



FIG. 8. Host cell proteins targeted by potential anti-cytokine (Dataset 2) compounds/drugs, rank-ordered by their promiscuity. Promiscuity refers to the number of predicted compounds/drugs (also called chemicals) that target the protein. FIG. 8 lists the top 100 targets corresponding to Dataset 2 compounds/drugs. The ordinate lists the proteins, and the horizontal bars (abscissa) show the corresponding number of compounds.



FIG. 9. Prioritized compounds proposed to have potential antiviral activities and their involvement in different modules in the viral-host PPI network. 25 compounds/drugs were identified for each of the four modules, resulting in a total of 64 distinct repurposable drugs or investigational compounds, some participating in multiple modules. The entries in the heat map display the ranking, color-coded from red (highest) to blue (lowest). The ranking was based on the proximity of their targets to proteins belonging to the modules.



FIG. 10. Distribution of the same compounds/drugs in the four studied modules. Compounds belonging to selected intersections and to the viral entry module are listed. Those colored red have been experimentally tested. See the complete list in FIG. 9 and Table 9.



FIG. 11. Interaction-pattern-based clustering of top-ranking compounds from four modules. Results for 64 compounds (or chemicals) identified to yield closest proximity to four selected modules. The compounds are clustered based on their interaction patterns with their targets listed in QuartataWeb. 12 main clusters (clusters 1-8, 10, 11, 13 and 14, from left to right, delimited by yellow squares) contain two or more compounds each; 16 chemicals do not belong to any cluster. From each of cluster, up to two chemicals were selected based on their side effects and MOA.



FIG. 12. Interaction-pattern-based clustering of chemicals targeting immune response. Clustering of 163 chemicals proposed to modulate the immune response, based on anti-cytokine signature gene derived from infected A546-Ace2 cells. The chemicals are clustered based on their interaction patterns reported in DrugBank or STITCH. 20 main clusters were distinguish (marked by yellow squares) which contain two or more chemicals, and 35 additional chemicals that do not form clusters.



FIG. 13. Structure of ten chemicals tested for SARS-CoV-2 infection inhibitory activity in vitro. Structures of salmeterol, rottlerin, temsirolimus, torin-1, ezetimibe, brompheniramine, imipramine, linsitinib, hexylresorcinol, and semaxanib, selected for in vitro assays.



FIG. 14. Representative fluorescence images of Mock, SARS-CoV-2 infected (Control), and Salmeterol-treated wells analyzed with the Multiwavelength Cell Scoring application in MetaXpress. Grayscales of the images were adjusted to enable direct comparison of the relative levels of fluorescence among the treatments: Segmentation images show how cells were segmented and identified as spike positive. Purple, nuclei; cyan, spike. Scale bar, 100 μm.



FIG. 15. Suppression of SARS-CoV-2 infection by identified compounds. Vero-E6 cells were pretreated with compounds (salmeterol, rottlerin, temsirolimus, torin-1, or ezetimibe) for 1 h prior to SARS-CoV-2 inoculation. 48-h post-infection cells were fixed and labeled for SARS-CoV-2 S protein. Images are representative of five imaging fields in triplicate wells. Scale bar, 100 μm.



FIG. 16. Violin plots of Vero-E6 cells labeled for Spike protein. The Multiwavelength Cell Scoring algorithm in MetaXpress was used to determine the integrated fluorescent signal in individual cells as a measure of the amount of Spike protein within each cell. The plots show the population distribution of the integrated signal for all of the treatments. The Boxes in the plot show the interquartile range (IQR) with the top and bottom edges marking the 75th and 25th percentiles, respectively. The horizontal line in the box is the median value, and the whiskers are defined to be 1.5 IQR. The ordinate is a log scale. The effect of the treatment is assessed quantitatively by changes in the median signal level, and qualitatively by observing changes in the modes. The dashed line is 3 standard deviations above the mean signal in the Mock samples and is used as a cutoff to quantify the number of cells that are positive or negative for the Spike signal. The statistics table below the plots shows the number of cells counted in each treatment group and the median of the population.



FIG. 17. Pie charts showing the effect of treatment on preventing infection of Vero-E6 cells. The number of cells above and below the cutoffs for being positive for Spike were counted and the percent cells in each category were determined. All analyses were done in Tibco Spotfire.



FIG. 18. Dose-response curve for Nafamostat in the syncytia assay. Data are the aggregate of 8 independent biological repeats; where errors are shown they represent SD from matching concentrations in at least three experiments.



FIG. 19. Dose-response curve for Linsitinib in the syncytia assay. Data are the aggregate of 8 independent biological repeats; where errors are shown they represent SD from matching concentrations in at least three experiments.



FIG. 20. Dose-response curve for Hexylresorcinol in the syncytia assay. Data are the aggregate of 8 independent biological repeats; where errors are shown they represent SD from matching concentrations in at least three experiments.



FIG. 21. Dose-response curves for dec-RVKR-CMK in the syncytia assay. Data are the aggregate of 8 independent biological repeats; where errors are shown they represent SD from matching concentrations in at least three experiments.



FIG. 22. Dose-response curve for Bromopheniramine in the syncytia assay. Data are the aggregate of 8 independent biological repeats; where errors are shown they represent SD from matching concentrations in at least three experiments.



FIG. 23. Dose-response curve for Salmeterol in the syncytia assay. Data are the aggregate of 8 independent biological repeats; where errors are shown they represent SD from matching concentrations in at least three experiments.



FIG. 24. Quantification of syncytia formation in HEK293 cells treated with dec-RVKR-CMK relative to nafamostat. Numbers indicate p-values obtained by one-way ANOVA (non-matched, unpaired) with Dunnett's multiple comparisons test in Graph Pad Prism (v7.00) compared with vehicle control (dotted line). No p-value, p>0.05. Bars and errors represent the means±SD from multiple independent biological repeats, each performed in quadruplicate. No error bars for n=1.



FIG. 25. Quantification of syncytia formation in HEK293 cells treated with brompheniramine relative to nafamostat. Numbers indicate p-values obtained by one-way ANOVA (non-matched, unpaired) with Dunnett's multiple comparisons test in Graph Pad Prism (v7.00) compared with vehicle control (dotted line). No p-value, p>0.05. Bars and errors represent the means±SD from multiple independent biological repeats, each performed in quadruplicate. No error bars for n=1.



FIG. 26. Quantification of syncytia formation in HEK293 cells treated with hexylresorcinol relative to nafamostat. Numbers indicate p-values obtained by one-way ANOVA (non-matched, unpaired) with Dunnett's multiple comparisons test in Graph Pad Prism (v7.00) compared with vehicle control (dotted line). No p-value, p>0.05. Bars and errors represent the means±SD from multiple independent biological repeats, each performed in quadruplicate. No error bars for n=1.



FIG. 27. Quantification of syncytia formation in HEK293 cells treated with imipramine relative to nafamostat. Numbers indicate p-values obtained by one-way ANOVA (non-matched, unpaired) with Dunnett's multiple comparisons test in Graph Pad Prism (v7.00) compared with vehicle control (dotted line). No p-value, p>0.05. Bars and errors represent the means±SD from multiple independent biological repeats, each performed in quadruplicate. No error bars for n=1.



FIG. 28. Quantification of syncytia formation in HEK293 cells treated with linsitinib relative to nafamostat. Numbers indicate p-values obtained by one-way ANOVA (non-matched, unpaired) with Dunnett's multiple comparisons test in Graph Pad Prism (v7.00) compared with vehicle control (dotted line). No p-value, p>0.05. Bars and errors represent the means±SD from multiple independent biological repeats, each performed in quadruplicate. No error bars for n=1.



FIG. 29. Quantification of syncytia formation in HEK293 cells treated with semaxanib relative to nafamostat. Numbers indicate p-values obtained by one-way ANOVA (non-matched, unpaired) with Dunnett's multiple comparisons test in Graph Pad Prism (v7.00) compared with vehicle control (dotted line). No p-value, p>0.05. Bars and errors represent the means±SD from multiple independent biological repeats, each performed in quadruplicate. No error bars for n=1.



FIG. 30. Quantification of syncytia formation in HEK293 cells treated with ezetimibe relative to nafamostat. Numbers indicate p-values obtained by one-way ANOVA (non-matched, unpaired) with Dunnett's multiple comparisons test in Graph Pad Prism (v7.00) compared with vehicle control (dotted line). No p-value, p>0.05. Bars and errors represent the means±SD from multiple independent biological repeats, each performed in quadruplicate. No error bars for n=1.



FIG. 31. Quantification of syncytia formation in HEK293 cells treated with salmeterol relative to nafamostat. Numbers indicate p-values obtained by one-way ANOVA (non-matched, unpaired) with Dunnett's multiple comparisons test in Graph Pad Prism (v7.00) compared with vehicle control (dotted line). No p-value, p>0.05. Bars and errors represent the means±SD from multiple independent biological repeats, each performed in quadruplicate. No error bars for n=1.



FIG. 32. Representative images illustrating syncytia phenotype and compound activity in HEK293 cells. No spike, donor cells expressing GFP only. Upper panel, raw fluorescence micrograph; lower panel, image with CNT overlay. GFP positive objects that met the criteria for syncytia are colored purple; cellular aggregates that are not syncytia are shown in yellow. Scale bar, 100 μm.



FIG. 33. Representative images illustrating syncytia phenotype and compound activity in HEK293 cells treated with 100 μM DMSO. Upper panel, raw fluorescence micrograph; lower panel, image with CNT overlay. GFP positive objects that met the criteria for syncytia are colored purple; cellular aggregates that are not syncytia are shown in yellow. Scale bar, 100 μm.



FIG. 34. Representative images illustrating syncytia phenotype and compound activity in HEK293 cells treated with nafamostat (5.5 μM). Upper panel, raw fluorescence micrograph; lower panel, image with CNT overlay. GFP positive objects that met the criteria for syncytia are colored purple; cellular aggregates that are not syncytia are shown in yellow. Scale bar, 100 μm.



FIG. 35. Representative images illustrating syncytia phenotype and compound activity in HEK293 cells treated with 100 μM dec-RVKR-CMK. Upper panel, raw fluorescence micrograph; lower panel, image with CNT overlay. GFP positive objects that met the criteria for syncytia are colored purple; cellular aggregates that are not syncytia are shown in yellow. Scale bar, 100 μm.



FIG. 36. Representative images illustrating syncytia phenotype and compound activity in HEK293 cells treated with 100 μM brompheniramine. Upper panel, raw fluorescence micrograph; lower panel, image with CNT overlay. GFP positive objects that met the criteria for syncytia are colored purple; cellular aggregates that are not syncytia are shown in yellow. Scale bar, 100 μm.



FIG. 37. Representative images illustrating syncytia phenotype and compound activity in HEK293 cells treated with 100 μM hexylresorcinol. Upper panel, raw fluorescence micrograph; lower panel, image with CNT overlay. GFP positive objects that met the criteria for syncytia are colored purple; cellular aggregates that are not syncytia are shown in yellow. Scale bar, 100 μm.



FIG. 38. Representative images illustrating syncytia phenotype and compound activity in HEK293 cells treated with 100 μM imipramine. Upper panel, raw fluorescence micrograph; lower panel, image with CNT overlay. GFP positive objects that met the criteria for syncytia are colored purple; cellular aggregates that are not syncytia are shown in yellow. Scale bar, 100 μm.



FIG. 39. Representative images illustrating syncytia phenotype and compound activity in HEK293 cells treated with linsitinib (25 μM). Upper panel, raw fluorescence micrograph; lower panel, image with CNT overlay. GFP positive objects that met the criteria for syncytia are colored purple; cellular aggregates that are not syncytia are shown in yellow. Scale bar, 100 μm.



FIG. 40. Representative images illustrating syncytia phenotype and compound activity in HEK293 cells treated with semaxanib (50 μM). Upper panel, raw fluorescence micrograph; lower panel, image with CNT overlay. GFP positive objects that met the criteria for syncytia are colored purple; cellular aggregates that are not syncytia are shown in yellow. Scale bar, 100 μm.



FIG. 41. Representative images illustrating syncytia phenotype and compound activity in HEK293 cells treated with 100 μM ezetimibe. Upper panel, raw fluorescence micrograph; lower panel, image with CNT overlay. GFP positive objects that met the criteria for syncytia are colored purple; cellular aggregates that are not syncytia are shown in yellow. Scale bar, 100 μm.



FIG. 42. Representative images illustrating syncytia phenotype and compound activity in HEK293 cells treated with 100 μM salmeterol. Upper panel, raw fluorescence micrograph; lower panel, image with CNT overlay. GFP positive objects that met the criteria for syncytia are colored purple; cellular aggregates that are not syncytia are shown in yellow. Scale bar, 100 μm. No spike, donor cells expressing GFP only.



FIG. 43. Quantification of syncytia formation in Calu-3 cells treated with dec-RVKR-CMK relative to nafamostat. Numbers indicate p-values obtained by one-way ANOVA (non-matched, unpaired) with Dunnett's multiple comparisons test in Graph Pad Prism (v7.00) compared with vehicle control (dotted line). No p-value, p>0.05. Bars and errors represent the means±SD from multiple independent biological repeats, each performed in quadruplicate. No error bars, n=1.



FIG. 44. Quantification of syncytia formation in Calu-3 cells treated with brompheniramine relative to nafamostat. Numbers indicate p-values obtained by one-way ANOVA (non-matched, unpaired) with Dunnett's multiple comparisons test in Graph Pad Prism (v7.00) compared with vehicle control (dotted line). No p-value, p>0.05. Bars and errors represent the means±SD from multiple independent biological repeats, each performed in quadruplicate. No error bars, n=1.



FIG. 45. Quantification of syncytia formation in Calu-3 cells treated with hexylresorcinol relative to nafamostat. Numbers indicate p-values obtained by one-way ANOVA (non-matched, unpaired) with Dunnett's multiple comparisons test in Graph Pad Prism (v7.00) compared with vehicle control (dotted line). No p-value, p>0.05. Bars and errors represent the means±SD from multiple independent biological repeats, each performed in quadruplicate. No error bars, n=1.



FIG. 46. Quantification of syncytia formation in Calu-3 cells treated with imipramine relative to nafamostat. Numbers indicate p-values obtained by one-way ANOVA (non-matched, unpaired) with Dunnett's multiple comparisons test in Graph Pad Prism (v7.00) compared with vehicle control (dotted line). No p-value, p>0.05. Bars and errors represent the means±SD from multiple independent biological repeats, each performed in quadruplicate. No error bars, n=1.



FIG. 47. Quantification of syncytia formation in Calu-3 cells treated with linsitinib relative to nafamostat. Numbers indicate p-values obtained by one-way ANOVA (non-matched, unpaired) with Dunnett's multiple comparisons test in Graph Pad Prism (v7.00) compared with vehicle control (dotted line). No p-value, p>0.05. Bars and errors represent the means±SD from multiple independent biological repeats, each performed in quadruplicate. No error bars, n=1.



FIG. 48. Quantification of syncytia formation in Calu-3 cells treated with semaxanib relative to nafamostat. Numbers indicate p-values obtained by one-way ANOVA (non-matched, unpaired) with Dunnett's multiple comparisons test in Graph Pad Prism (v7.00) compared with vehicle control (dotted line). No p-value, p>0.05. Bars and errors represent the means±SD from multiple independent biological repeats, each performed in quadruplicate. No error bars, n=1.



FIG. 49. Quantification of syncytia formation in Calu-3 cells treated with ezetimibe relative to nafamostat. Numbers indicate p-values obtained by one-way ANOVA (non-matched, unpaired) with Dunnett's multiple comparisons test in Graph Pad Prism (v7.00) compared with vehicle control (dotted line). No p-value, p>0.05. Bars and errors represent the means±SD from multiple independent biological repeats, each performed in quadruplicate. No error bars, n=1.



FIG. 50. Quantification of syncytia formation in Calu-3 cells treated with salmeterol relative to nafamostat. Numbers indicate p-values obtained by one-way ANOVA (non-matched, unpaired) with Dunnett's multiple comparisons test in Graph Pad Prism (v7.00) compared with vehicle control (dotted line). No p-value, p>0.05. Bars and errors represent the means±SD from multiple independent biological repeats, each performed in quadruplicate. No error bars, n=1.



FIG. 51. Representative images illustrating syncytia phenotype and compound activity in Calu-3 cells. No spike, donor cells expressing GFP only. Upper panel, raw fluorescence micrograph; lower panel, image with CNT overlay. GFP positive objects that met the criteria for syncytia are colored purple; cellular aggregates that are not syncytia are shown in yellow. Scale bar, 100 μm.



FIG. 52. Representative images illustrating syncytia phenotype and compound activity in Calu-3 cells treated with 100 μM DMSO. Upper panel, raw fluorescence micrograph; lower panel, images with CNT overlay. GFP positive objects that met the criteria for syncytia are colored purple; cellular aggregates that are not syncytia are shown in yellow. Scale bar, 100 μm.



FIG. 53. Representative images illustrating syncytia phenotype and compound activity in Calu-3 cells treated with nafamostat (5.5 μM). Upper panel, raw fluorescence micrograph; lower panel, image with CNT overlay. GFP positive objects that met the criteria for syncytia are colored purple; cellular aggregates that are not syncytia are shown in yellow. Scale bar, 100 μm.



FIG. 54. Representative images illustrating syncytia phenotype and compound activity in Calu-3 cells treated with 100 μM dec-RVKR-CMK. Upper panel, raw fluorescence micrograph; lower panel, image with CNT overlay. GFP positive objects that met the criteria for syncytia are coloredpurp/e; cellular aggregates that are not syncytia are shown in yellow. Scale bar, 100 μm.



FIG. 55. Representative images illustrating syncytia phenotype and compound activity in Calu-3 cells treated with 100 μM brompheniramine. Upper panel, raw fluorescence micrograph; lower panel, image with CNT overlay. GFP positive objects that met the criteria for syncytia are colored purple; cellular aggregates that are not syncytia are shown in yellow. Scale bar, 100 μm.



FIG. 56. Representative images illustrating syncytia phenotype and compound activity in Calu-3 cells treated with 100 μM hexylresorcinol. Upper panel, raw fluorescence micrograph; lower panel, image with CNT overlay. GFP positive objects that met the criteria for syncytia are colored purple; cellular aggregates that are not syncytia are shown in yellow. Scale bar, 100 μm.



FIG. 57. Representative images illustrating syncytia phenotype and compound activity in Calu-3 cells treated with 100 μM imipramine. Upper panel, raw fluorescence micrograph; lower panel, image with CNT overlay. GFP positive objects that met the criteria for syncytia are colored purple; cellular aggregates that are not syncytia are shown in yellow. Scale bar, 100 μm.



FIG. 58. Representative images illustrating syncytia phenotype and compound activity in Calu-3 cells treated with linsitinib (25 μM). Upper panel, raw fluorescence micrograph; lower panel, image with CNT overlay. GFP positive objects that met the criteria for syncytia are colored purple; cellular aggregates that are not syncytia are shown in yellow. Scale bar, 100 μm.



FIG. 59. Representative images illustrating syncytia phenotype and compound activity in Calu-3 cells treated with semaxanib (50 μM). Upper panel, raw fluorescence micrograph; lower panel, image with CNT overlay. GFP positive objects that met the criteria for syncytia are colored purple; cellular aggregates that are not syncytia are shown in yellow. Scale bar, 100 μm.



FIG. 60. Representative images illustrating syncytia phenotype and compound activity in Calu-3 cells treated with 100 μM ezetimibe. Upper panel, raw fluorescence micrograph; lower panel, image with CNT overlay. GFP positive objects that met the criteria for syncytia are colored purple; cellular aggregates that are not syncytia are shown in yellow. Scale bar, 100 μm.



FIG. 61. Representative images illustrating syncytia phenotype and compound activity in Calu-3 cells treated with 100 μM salmeterol. Upper panel, raw fluorescence micrograph; lower panel, image with CNT overlay. GFP positive objects that met the criteria for syncytia are colored purple; cellular aggregates that are not syncytia are shown in yellow. Scale bar, 100 μm.



FIG. 62. The 36 genes of antiviral signature expression in A549 cells (same as FIG. 4).



FIG. 63. The 36 genes of antiviral signature expression in A549-ACE2 cells.



FIG. 64. The 17 genes of anti-inflammatory signature expression in A549-ACE2 cells (same as FIG. 5).



FIG. 65. The 17 genes of anti-inflammatory signature expression in A549 cells.



FIG. 66. Schematic representation of various stages of SARS-CoV-2 infection: viral entry, endosomal maturation, replication, translation, and accompanying cell signaling and regulation or immune responses, described in the main text. Mainly, SARS-CoV-2 spike binds the host receptor ACE2 (Hoffmann M et al. Cell, 2020, 181, 271-280) complexed with the amino acid transporter B0 AT1 (Yan R et al. Science, 2020, 367, 1444-1448). Proteolytic cleavages (e.g., by TMPRSS2) are essential to viral entry, including spike priming and membrane fusion, or lysosomal escape after endocytosis. PlKfyve is the main enzyme synthesizing PI(3,5)P2 in early endosome (de Lartigue J et al. Traffic, 2009, 10, 883-893), and PI(3,5)P2 regulates early-to-late endosome events. TPC2 is a major downstream effector of PI(3,5)P2 (Li P et al. Trends Biochem Sci. 2019, 44, 110-124). Dominant pathways in four modules involved in SARS-CoV-2 infection are listed in the upper right boxes (see also Table 7). The diagram also shows selected drugs that have been identified and experimentally validated to inhibit or reduce SARS-2-CoV-2 infection (mainly viral entry) in highlighted in boxes (with red fonts).



FIG. 67. Subnet of PPIs between host cell proteins implicated in SARS-CoV-2 infection and those targeted by selected compounds. The sandy brown nodes and edges represent the proteins and interactions in the SARS-CoV-2 host response network; and in the background (transparent light blue nodes and edges) is the lung tissue-specific protein interactome. The relative size of each protein node is consistent with its degree (number of connections) in the PPI network. Thirteen compounds were identified as candidate repurposable or investigational drugs for host-targeted antiviral therapy (based on Dataset 1) and their connections to targets in host response network (as reported in DrugBank or STITCH) are shown by color-coded labels and connectors. Magenta nodes represent the compounds that predominantly inhibit viral entry; light green and red represent those against viral translation, replication, and immune response; and cyan nodes represent multifunctional compounds.



FIG. 68. Chemical structures of selected drugs displayed in FIG. 66 and FIG. 67 targeting various components of the viral-host interactome; see all tested drugs in FIG. 13.





DETAILED DESCRIPTION

The materials, compounds, compositions, and methods described herein may be understood more readily by reference to the following detailed description of specific aspects of the disclosed subject matter and the Examples included therein.


Before the present materials, compounds, compositions, and methods are disclosed and described, it is to be understood that the aspects described below are not limited to specific synthetic methods or specific reagents, as such may, of course, vary. It is also to be understood that the terminology used herein is for the purpose of describing particular aspects only and is not intended to be limiting.


Also, throughout this specification, various publications are referenced. The disclosures of these publications in their entireties are hereby incorporated by reference into this application in order to more fully describe the state of the art to which the disclosed matter pertains. The references disclosed are also individually and specifically incorporated by reference herein for the material contained in them that is discussed in the sentence in which the reference is relied upon.


General Definitions

In this specification and in the claims that follow, reference will be made to a number of terms, which shall be defined to have the following meanings:


Throughout the specification and claims the word “comprise” and other forms of the word, such as “comprising” and “comprises,” means including but not limited to, and is not intended to exclude, for example, other additives, components, integers, or steps.


As used in the description and the appended claims, the singular forms “a,” “an,” and “the” include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to “a composition” includes mixtures of two or more such compositions, reference to “an analog” includes mixtures of two or more such analogs, and the like.


“Optional” or “optionally” means that the subsequently described event or circumstance can or cannot occur, and that the description includes instances where the event or circumstance occurs and instances where it does not.


Ranges can be expressed herein as from “about” one particular value, and/or to “about” another particular value. By “about” is meant within 5% of the value, e.g., within 4, 3, 2, or 1% of the value. When such a range is expressed, another aspect includes from the one particular value and/or to the other particular value. Similarly, when values are expressed as approximations, by use of the antecedent “about,” it will be understood that the particular value forms another aspect. It will be further understood that the endpoints of each of the ranges are significant both in relation to the other endpoint, and independently of the other endpoint.


“Exemplary” means “an example of” and is not intended to convey an indication of a preferred or ideal embodiment. “Such as” is not used in a restrictive sense, but for explanatory purposes.


Values can be expressed herein as an “average” value. “Average” generally refers to the statistical mean value.


By “substantially” is meant within 5%, e.g., within 4%, 3%, 2%, or 1%.


It is understood that throughout this specification the identifiers “first” and “second” are used solely to aid the reader in distinguishing the various components, features, or steps of the disclosed subject matter. The identifiers “first” and “second” are not intended to imply any particular order, amount, preference, or importance to the components or steps modified by these terms.


References in the specification and concluding claims to parts by weight of a particular element or component in a composition denotes the weight relationship between the element or component and any other elements or components in the composition or article for which a part by weight is expressed. Thus, in a compound containing 2 parts by weight of component X and 5 parts by weight component Y, X and Y are present at a weight ratio of 2:5, and are present in such ratio regardless of whether additional components are contained in the compound.


A weight percent (wt. %) of a component, unless specifically stated to the contrary, is based on the total weight of the formulation or composition in which the component is included.


The term “or combinations thereof” as used herein refers to all permutations and combinations of the listed items preceding the term. For example, “A, B, C, or combinations thereof” is intended to include at least one of: A, B, C, AB, AC, BC, or ABC, and if order is important in a particular context, also BA, CA, CB, CBA, BCA, ACB, BAC, or CAB. Continuing with this example, expressly included are combinations that contain repeats of one or more item or term, such as BB, AAA, AB, BBC, AAABCCCC, CBBAAA, CABABB, and so forth. The skilled artisan will understand that typically there is no limit on the number of items or terms in any combination, unless otherwise apparent from the context.


As used herein, by a “subject” is meant an individual. Thus, the “subject” can include domesticated animals (e.g., cats, dogs, etc.), livestock (e.g., cattle, horses, pigs, sheep, goats, etc.), laboratory animals (e.g., mouse, rabbit, rat, guinea pig, etc.), and birds. “Subject” can also include a mammal, such as a primate or a human. Thus, the subject can be a human or veterinary patient. The term “patient” refers to a subject under the treatment of a clinician, e.g., physician.


As used herein, microbes include, for example, bacteria, fungi, viruses, protozoa, etc.


As used herein, antimicrobials include, for example, antibacterials, antifungals, and antivirals. As used herein, “antimicrobial” refers to the ability to treat or control (e.g., reduce, prevent, treat, or eliminate) the growth of a microbe at any concentration. Similarly, the terms “antibacterial,” “antifungal,” and “antiviral” refer to the ability to treat or control the growth of bacteria, fungi, and viruses at any concentration, respectively.


The term “inhibit” refers to a decrease in an activity, response, condition, disease, or other biological parameter. This can include but is not limited to the complete ablation of the activity, response, condition, or disease. This can also include, for example, a 10% reduction in the activity, response, condition, or disease as compared to the native or control level. Thus, the reduction can be a 10, 20, 30, 40, 50, 60, 70, 80, 90, 100%, or any amount of reduction in between as compared to native or control levels.


By “reduce” or other forms of the word, such as “reducing” or “reduction,” is meant lowering of an event or characteristic (e.g., microbe population/infection). Similarly, “increase” or other forms of the word, such as “increasing” or “increase,” is meant raising of an event or characteristic. It is understood that in both cases this is typically in relation to some standard or expected value, in other words it is relative, but that it is not always necessary for the standard or relative value to be referred to. For example, “reduces tumor growth” means decreasing the amount of tumor cells relative to a standard or a control. For example, “reducing microbial infection” means reducing the spread of a microbial infection relative to a standard or a control.


By “prevent” or other forms of the word, such as “preventing” or “prevention,” is meant to stop a particular event or characteristic, to stabilize or delay the development or progression of a particular event or characteristic, or to minimize the chances that a particular event or characteristic will occur. “Prevent” does not require comparison to a control as it is typically more absolute than, for example, “reduce.” As used herein, something could be reduced but not prevented, but something that is reduced could also be prevented. Likewise, something could be prevented but not reduced, but something that is prevented could also be reduced. It is understood that where reduce or prevent are used, unless specifically indicated otherwise, the use of the other word is also expressly disclosed. For example, the terms “prevent” or “suppress” can refer to a treatment that forestalls or slows the onset of a disease or condition or reduced the severity of the disease or condition. Thus, if a treatment can treat a disease in a subject having symptoms of the disease, it can also prevent or suppress that disease in a subject who has yet to suffer some or all of the symptoms.


As used herein, “treat” or other forms of the word, such as “treated” or “treatment” refers to obtaining beneficial or desired clinical results. Beneficial or desired clinical results include, but are not limited to, any one or more of: alleviating of one or more symptoms (such as viral spread), diminishing the extent of cancer or viral infection, stabilizing (i.e., not worsening) state of disease, preventing or delaying spread of the viral infection, delaying occurrence or recurrence of disease, delaying or slowing of disease progression, ameliorating the disease state, and remission (whether partial or total). For example, “treat” or other forms of the word, such as “treated” or “treatment,” can refer to administration of a composition or performing a method in order to reduce, prevent, inhibit, or eliminate a particular characteristic or event (e.g., microbe growth or survival). The term “control” is used synonymously with the term “treat.”


The term “therapeutically effective amount” refers to the amount of the composition used is of sufficient quantity to ameliorate one or more causes or symptoms of a disease or disorder. Such amelioration only requires a reduction or alteration, not necessarily elimination. In reference to viral infections, an effective amount comprises an amount sufficient to cure, palliate, ameliorate, stabilize, reverse, prevent, slow or delay the progression of the disease, pathological condition, or disorder. In some embodiments, an effective amount is an amount sufficient to delay development or infection. In some embodiments, an effective amount is an amount sufficient to prevent or delay occurrence and/or recurrence. An effective amount can be administered in one or more doses. In the case of a viral infection, the effective amount of the drug or composition may: cure viral infections, palliate or ameliorate symptoms associated with viral infections, stabilize to some extent and preferably stop viral replication, prevent viral infections or the onset of complications associated with viral infections, slow or delay the progression of viral replication.


The term “pharmaceutically acceptable” refers to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problems or complications commensurate with a reasonable benefit/risk ratio.


“Pharmaceutically acceptable salt” refers to a salt that is pharmaceutically acceptable and has the desired pharmacological properties. Such salts include those that may be formed where acidic protons present in the compounds are capable of reacting with inorganic or organic bases. Suitable inorganic salts include those formed with the alkali metals, e.g., sodium, potassium, magnesium, calcium, and aluminum. Suitable organic salts include those formed with organic bases such as the amine bases, e.g., ethanolamine, diethanolamine, triethanolamine, tromethamine, N-methylglucamine, and the like. Such salts also include acid addition salts formed with inorganic acids (e.g., hydrochloric and hydrobromic acids) and organic acids (e.g., acetic acid, citric acid, maleic acid, and the alkane- and arene-sulfonic acids such as methanesulfonic acid and benzenesulfonic acid). When two acidic groups are present, a pharmaceutically acceptable salt may be a mono-acid-mono-salt or a di-salt; similarly, where there are more than two acidic groups present, some or all of such groups can be converted into salts.


“Pharmaceutically acceptable excipient” refers to an excipient that is conventionally useful in preparing a pharmaceutical composition that is generally safe, non-toxic, and desirable, and includes excipients that are acceptable for veterinary use as well as for human pharmaceutical use. Such excipients can be solid, liquid, semisolid, or, in the case of an aerosol composition, gaseous.


A “pharmaceutically acceptable carrier” is a carrier, such as a solvent, suspending agent or vehicle, for delivering the disclosed compounds to the patient. The carrier can be liquid or solid and is selected with the planned manner of administration in mind. Liposomes are also a pharmaceutical carrier. As used herein, “carrier” includes any and all solvents, dispersion media, vehicles, coatings, diluents, antibacterial and antifungal agents, isotonic and absorption delaying agents, buffers, carrier solutions, suspensions, colloids, and the like. The use of such media and agents for pharmaceutical active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions is contemplated.


As used herein, the term “delivery” encompasses both local and systemic delivery. As used herein, the term “nucleic acid,” in its broadest sense, refers to any compound and/or substance that is or can be incorporated into a polynucleotide chain. In some embodiments, a nucleic acid is a compound and/or substance that is or can be incorporated into a polynucleotide chain via a phosphodiester linkage. In some embodiments, “nucleic acid” refers to individual nucleic acid residues (e.g., nucleotides and/or nucleosides). In some embodiments, “nucleic acid” refers to a polynucleotide chain comprising individual nucleic acid residues. In some embodiments, “nucleic acid” encompasses RNA as well as single and/or double-stranded DNA and/or cDNA. Furthermore, the terms “nucleic acid,” “DNA,” “RNA,” and/or similar terms include nucleic acid analogs, i.e., analogs having other than a phosphodiester backbone.


Methods and Compositions


Disclosed herein are methods of identifying a compound for treating, preventing, or ameliorating a disease or disorder in a subject in need thereof. Also disclosed herein are methods for of treating, preventing, or ameliorating a disease or disorder in a subject in need thereof, the methods comprising administering to the subject a therapeutically effect amount of a compound, such as a compound identified by the methods disclosed herein, or a therapeutically effective amount of a composition (such as a pharmaceutical composition) comprising said compound.


For example, the compounds and compositions described herein or pharmaceutically acceptable salts thereof are useful for treating a disease or disorder in humans, e.g., pediatric and geriatric populations, and in animals, e.g., veterinary applications. The disclosed methods can optionally include identifying a patient who is or may be in need of treatment of a disease or disorder.


In some examples, the disease or disorder comprises an infection, such as with an infectious microbe (e.g., bacteria, virus, fungi, protozoa, etc.). In some examples, the disease or disorder comprises an infection with a coronavirus.


Examples of viruses include both DNA viruses and RNA viruses. Exemplary viruses can belong to the following non-exclusive list of families Adenoviridae, Arenaviridae, Astroviridae, Baculoviridae, Barnaviridae, Betaherpesvirinae, Birnaviridae, Bromoviridae, Bunyaviridae, Caliciviridae, Chordopoxvirinae, Circoviridae, Comoviridae, Coronaviridae, Cystoviridae, Corticoviridae, Entomopoxvirinae, Filoviridae, Flaviviridae, Fuselloviridae, Geminiviridae, Hepadnaviridae, Herpesviridae, Gammaherpesvirinae, Inoviridae, Iridoviridae, Leviviridae, Lipothrixviridae, Microviridae, Myoviridae, Nodaviridae, Orthomyxoviridae, Papovaviridae, Paramyxoviridae, Paramyxovirinae, Partitiviridae, Parvoviridae, Phycodnaviridae, Picornaviridae, Plasmaviridae, Pneumovirinae, Podoviridae, Polydnaviridae, Potyviridae, Poxviridae, Reoviridae, Retroviridae, Rhabdoviridae, Sequiviridae, Siphoviridae, Tectiviridae, Tetraviridae, Togaviridae, Tombusviridae, and Totiviridae.


Specific examples of viruses include, but are not limited to, Mastadenovirus, Adenovirus, Human adenovirus 2, Aviadenovirus, African swine fever virus, classical swine fever virus, arenavirus, Lymphocytic choriomeningitis virus, Ippy virus, Lassa virus, Arterivirus, Human astrovirus 1, Nucleopolyhedrovirus, Autographa californica nucleopolyhedrovirus, Granulovirus, Plodia interpunctella granulovirus, Badnavirus, Commelina yellow mottle virus, Rice tungro bacilliform, Barnavirus, Mushroom bacilliform virus, Aquabirnavirus, Infectious pancreatic necrosis virus, Avibirnavirus, Infectious bursal disease virus, Entomobirnavirus, Drosophila X virus, Alfamovirus, Alfalfa mosaic virus, Ilarvirus, Ilarvirus Subgroups 1-10, Tobacco streak virus, Bromovirus, Brome mosaic virus, Cucumovirus, Cucumber mosaic virus, Bhanja virus Group, Kaisodi virus, Mapputta virus, Okola virus, Resistencia virus, Upolu virus, Yogue virus, Bunyavirus, Anopheles A virus, Anopheles B virus, Bakau virus, Bunyamwera virus, Bwamba virus, C virus, California encephalitis virus, Capim virus, Gamboa virus, Guama virus, Koongol virus, Minatitlan virus, Nyando virus, Olifantsvlei virus, Patois virus, Simbu virus, Tete virus, Turlock virus, Hantavirus, Hantaan virus, Nairovirus, Crimean-Congo hemorrhagic fever virus, Dera Ghazi Khan virus, Hughes virus, Nairobi sheep disease virus, Qalyub virus, Sakhalin virus, Thiafora virus, Crimean-congo hemorrhagic fever virus, Phlebovirus, Sandfly fever virus, Bujaru complex, Candiru complex, Chilibre complex, Frijoles complex, Punta Toro complex, Rift Valley fever complex, Salehabad complex, Sandfly fever Sicilian virus, Uukuniemi virus, Uukuniemi virus, Tospovirus, Tomato spotted wilt virus, Calicivirus, Vesicular exanthema of swine virus, Capillovirus, Apple stem grooving virus, Carlavirus, Carnation latent virus, Caulimovirus, Cauliflower mosaic virus, Circovirus, Chicken anemia virus, Closterovirus, Beet yellows virus, Comovirus, Cowpea mosaic virus, Fabavirus, Broad bean wilt virus 1, Nepovirus, Tobacco ringspot virus, Coronavirus, Avian infectious bronchitis virus, Bovine coronavirus, Canine coronavirus, Feline infectious peritonitis virus, Human coronavirus 299E, Human coronavirus OC43, Murine hepatitis virus, Porcine epidemic diarrhea virus, Porcine hemagglutinating encephalomyelitis virus, Porcine transmissible gastroenteritis virus, porcine reproductive and respiratory syndrome virus, Rat coronavirus, Turkey coronavirus, Rabbit coronavirus, Torovirus, Berne virus, Breda virus, Corticovirus, Alteromonas phage PM2, Pseudomonas Phage phi6, Deltavirus, Hepatitis delta virus, Hepatitis D virus, Hepatitis E virus, Dianthovirus, Carnation ringspot virus, Red clover necrotic mosaic virus, Sweet clover necrotic mosaic virus, Enamovirus, Pea enation mosaic virus, Filovirus, Marburg virus, Ebola virus, Ebola virus Zaire, Flavivirus, Yellow fever virus, Tick-borne encephalitis virus, Rio Bravo Group, Japanese encephalitis, Tyuleniy Group, Ntaya Group, Uganda S Group, Dengue Group, Modoc Group, Pestivirus, Bovine diarrhea virus, Hepatitis C virus, Furovirus, Soil-borne wheat mosaic virus, Beet necrotic yellow vein virus, Fusellovirus, Sulfobolus virus 1, Subgroup I, II, and III geminivirus, Maize streak virus, Beet curly top virus, Bean golden mosaic virus, Orthohepadnavirus, Hepatitis B virus, Avihepadnavirus, Alphaherpesvirinae, Simplexvirus, Human herpesvirus 1, Herpes Simplex virus-1, Herpes Simplex virus-2, Varicellovirus, Varicella-Zoster virus, Epstein-Barr virus, Human herpesvirus 3, Cytomegalovirus, Human herpesvirus 5, Muromegalovirus, Mouse cytomegalovirus 1, Roseolovirus, Human herpesvirus 6, Lymphocryptovirus, Human herpesvirus 4, Rhadinovirus, Ateline herpesvirus 2, Hordeivirus, Barley stripe mosaic virus, Hypoviridae, Hypovirus, Cryphonectria hypovirus 1-EP713, Idaeovirus, Raspberry bushy dwarf virus, Inovirus, Coliphage fd, Plectrovirus, Acholeplasma phage L51, Iridovirus, Chilo iridescent virus, Chloriridovirus, Mosquito iridescent virus, Ranavirus, Frog virus 3, Lymphocystivirus, Lymphocystis disease virus flounder isolate, Goldfish virus 1, Levivirus, Enterobacteria phage MS2, Allolevirus, Enterobacteria phage Qbeta, Lipothrixvirus, Thermoproteus virus 1, Luteovirus, Barley yellow dwarf virus, Machlomovirus, Maize chlorotic mottle virus, Marafivirus, Maize rayado fino virus, Microvirus, Coliphage phiX174, Spiromicrovirus, Spiroplasma phage 4, Bdellomicrovirus, Bdellovibrio phage MAC 1, Chlamydiamicrovirus, Chlamydia phage 1, T4-like phages, coliphage T4, Necrovirus, Tobacco necrosis virus, Nodavirus, Nodamura virus, Influenzavirus A, B and C, Thogoto virus, Polyomavirus, Murine polyomavirus, Papillomavirus, Rabbit (Shope) Papillomavirus, Paramyxovirus, Human parainfluenza virus 1, Morbillivirus, Measles virus, Rubulavirus, Mumps virus, Pneumovirus, Human respiratory syncytial virus, Partitivirus, Gaeumannomyces graminis virus 019/6-A, Chrysovirus, Penicillium chrysogenum virus, Alphacryptovirus, White clover cryptic viruses 1 and 2, Betacryptovirus, Parvovirinae, Parvovirus, Minute mice virus, Erythrovirus, B19 virus, Dependovirus, Adeno-associated virus 1, Densovirinae, Densovirus, Junonia coenia densovirus, Iteravirus, Bombyx mori virus, Contravirus, Aedes aegypti densovirus, Phycodnavirus, 1-Paramecium bursaria Chlorella NC64A virus group, Paramecium bursaria chlorella virus 1, 2-Paramecium bursaria Chlorella Pbi virus, 3-Hydra viridis Chlorella virus, Enterovirus, Poliovirus, Human poliovirus 1, Rhinovirus, Human rhinovirus 1A, Hepatovirus, Human hepatitis A virus, Cardiovirus, Encephalomyocarditis virus, Aphthovirus, Foot-and-mouth disease virus, Plasmavirus, Acholeplasma phage L2, Podovirus, Coliphage T7, Ichnovirus, Campoletis sonorensis virus, Bracovirus, Cotesia melanoscela virus, Potexvirus, Potato virus X, Potyvirus, Potato virus Y, Rymovirus, Ryegrass mosaic virus, Bymovirus, Barley yellow mosaic virus, Orthopoxvirus, Vaccinia virus, Parapoxvirus, Orf virus, Avipoxvirus, Fowlpox virus, Capripoxvirus, Sheep pox virus, Leporipoxvirus, Myxoma virus, Suipoxvirus, Swinepox virus, Molluscipoxvirus, Molluscum contagiosum virus, Yatapoxvirus, Yaba monkey tumor virus, Entomopoxviruses A, B, and C, Melolontha melolontha entomopoxvirus, Amsacta moorei entomopoxvirus, Chironomus luridus entomopoxvirus, Orthoreovirus, Mammalian orthoreoviruses, reovirus 3, Avian orthoreoviruses, Orbivirus, African horse sickness viruses 1, Bluetongue viruses 1, Changuinola virus, Corriparta virus, Epizootic hemarrhogic disease virus 1, Equine encephalosis virus, Eubenangee virus group, Lebombo virus, Orungo virus, Palyam virus, Umatilla virus, Wallal virus, Warrego virus, Kemerovo virus, Rotavirus, Groups A-F rotaviruses, Simian rotavirus SA11, Coltivirus, Colorado tick fever virus, Aquareovirus, Groups A-E aquareoviruses, Golden shiner virus, Cypovirus, Cypovirus types 1-12, Bombyx mori cypovirus 1, Fijivirus, Fijivirus groups 1-3, Fiji disease virus, Fijivirus groups 2-3, Phytoreovirus, Wound tumor virus, Oryzavirus, Rice ragged stunt, Mammalian type B retroviruses, Mouse mammary tumor virus, Mammalian type C retroviruses, Murine Leukemia Virus, Reptilian type C oncovirus, Viper retrovirus, Reticuloendotheliosis virus, Avian type C retroviruses, Avian leukosis virus, Type D Retroviruses, Mason-Pfizer monkey virus, BLV-HTLV retroviruses, Bovine leukemia virus, Lentivirus, Bovine lentivirus, Bovine immunodeficiency virus, Equine lentivirus, Equine infectious anemia virus, Feline lentivirus, Feline immunodeficiency virus, Canine immunodeficiency virus Ovine/caprine lentivirus, Caprine arthritis encephalitis virus, Visna/maedi virus, Primate lentivirus group, Human immunodeficiency virus 1, Human immunodeficiency virus 2, Human immunodeficiency virus 3, Simian immunodeficiency virus, Spumavirus, Human spuma virus, Vesiculovirus, Vesicular stomatitis virus, Vesicular stomatitis Indiana virus, Lyssavirus, Rabies virus, Ephemerovirus, Bovine ephemeral fever virus, Cytorhabdovirus, Lettuce necrotic yellows virus, Nucleorhabdovirus, Potato yellow dwarf virus, Rhizidiovirus, Rhizidiomyces virus, Sequivirus, Parsnip yellow fleck virus, Waikavirus, Rice tungro spherical virus, Lambda-like phages, Coliphage lambda, Sobemovirus, Southern bean mosaic virus, Tectivirus, Enterobacteria phage PRD1, Tenuivirus, Rice stripe virus, Nudaurelia capensis beta-like viruses, Nudaurelia beta virus, Nudaurelia capensis omega-like viruses, Nudaurelia omega virus, Tobamovirus, Tobacco mosaic virus (vulgare strain; ssp. NC82 strain), Tobravirus, Tobacco rattle virus, Alphavirus, Sindbis virus, Rubivirus, Rubella virus, Tombusvirus, Tomato bushy stunt, virus, Carmovirus, Carnation mottle virus, Turnip crinkle virus, Totivirus, Saccharomyces cerevisiae virus, Giardiavirus, Giardia lamblia virus, Leishmaniavirus, Leishmania brasiliensis virus 1-1, Trichovirus, Apple chlorotic leaf spot virus, Tymovirus, Turnip yellow mosaic virus, Umbravirus, Carrot mottle virus, Variola virus, Coxsackie virus, Dengue virus, Rous sarcoma virus, Zika virus, Lassa fever virus, Eastern Equine Encephalitis virus, Venezuelan equine encephalitis virus, Western equine encephalitis virus, St. Louis Encephalitis virus, Murray Valley fever virus, West Nile virus, Human T-cell Leukemia virus type-1, echovirus, norovirus, and feline calicivirus (FCV).


In some examples, the virus can comprise an influenza virus, a coronavirus, or a combination thereof. Examples of influenza viruses include, but are not limited to, Influenzavirus A (including the H1N1, H2N2, H3N2, H5N1, H7N7, H1N2, H9N2, H7N2, H7N3, H10N7, H7N9, and H6N1 serotypes), Influenzavirus B, Influenzavirus C, and Influenzavirus D. Examples of coronaviruses include, but are not limited to, avian coronavirus (IBV), porcine epidemic diarrhea virus (PEDV), porcine respiratory coronavirus (PRCV), porcine reproductive and respiratory syndrome (PRRS) virus, transmissible gastroenteritis virus (TGEV), feline coronavirus (FCoV), feline infectious peritonitis virus (FIPV), feline enteric coronavirus (FECV), canine coronavirus (CCoV), rabbit coronavirus (RaCoV), mouse hepatitis virus (MHV), rat coronavirus (RCoV), sialodacryadenitis virus of rats (SDAV), bovine coronavirus (BCoV), bovine enterovirus (BEV), porcine coronavirus HKU15 (PorCoV HKU15), Porcine epidemic diarrhea virus (PEDV), porcine hemagglutinating encephalomyelitis virus (HEV), turkey bluecomb coronavirus (TCoV), human coronavirus (HCoV)-229E, HCoV-OC43, HCoV-HKU1, HCoV-NL63, Severe Acute Respiratory Syndrome (SARS)-Coronavirus (CoV)(SARS-CoV), Severe Acute Respiratory Syndrome (SARS)-Coronavirus (CoV)-2 (SARS-CoV-2), and middle east respiratory syndrome (MERS) coronavirus (CoV) (MERS-CoV). In some examples, the virus can comprise Severe Acute Respiratory Syndrome (SARS)-Coronavirus (CoV)-2 (SARS-CoV-2).


Specific examples of bacteria include, but are not limited to, Mycobacterium tuberculosis, Mycobacterium bovis, Mycobacterium bovis strain BCG, BCG substrains, Mycobacterium avium, Mycobacterium intracellular, Mycobacterium africanum, Mycobacterium kansasii, Mycobacterium marinum, Mycobacterium ulcerans, Mycobacterium avium subspecies paratuberculosis, Nocardia asteroides, other Nocardia species, Legionella pneumophila, other Legionella species, Acetinobacter baumanii, Salmonella typhi, Salmonella enterica, Salmonella Typhimurium, other Salmonella species, Shigella boydii, Shigella dysenteriae, Shigella sonnei, Shigella flexneri, other Shigella species, Yersinia pestis, Pasteurella haemolytica, Pasteurella multocida, other Pasteurella species, Actinobacillus pleuropneumoniae, Listeria monocytogenes, Listeria ivanovii, Brucella abortus, Brucella suis, Brucella melitensis, other Brucella species, Cowdria ruminantium, Borrelia burgdorferi, Bordetella avium, Bordetella pertussis, Bordetella bronchiseptica, Bordetella trematum, Bordetella hinzii, Bordetella pteri, Bordetella parapertussis, Bordetella ansorpii, other Bordetella species, Burkholderia mallei, Burkholderia psuedomallei, Burkholderia cepacian, Chlamydia pneumoniae, Chlamydia trachomatis, Chlamydia psittaci, Coxiella burnetii, rickettsia, rickettsia prowazekii, rickettsia typhi, other Rickettsial species, Ehrlichia species, Staphylococcus aureus, Staphylococcus epidermidis, Streptococcus pneumoniae, Streptococcus pyogenes, Streptococcus agalactiae, Streptococcus uberis, Escherichia coli, Vibrio cholerae, Vibrio parahaemolyticus, Campylobacter species, Neiserria meningitidis, Neiserria gonorrhea, Pseudomonas aeruginosa, other Pseudomonas species, Haemophilus influenzae, Haemophilus ducreyi, other Hemophilus species, Clostridium tetani, Clostridium difficile, Clostridium botulinum, Clostridium perfringens, other Clostridium species, Yersinia enterolitica, Yersinia pestis, other Yersinia species, Mycoplasma species, Bacillus anthracis, Bacillus abortus, other Bacillus species, Corynebacterium diptheriae, Corynebacterium bovis, Francisella tularensis, Chlamydophila psittaci, Campylocavter jejuni, Enterobacter aerogenes, Klebsiella pneumoniae, Klebsiella oxytoca, Proteus spp., Serratia marcescens, Trueperella pyogenes, and Vibria vulnificus.


Specific examples of fungi include, but are not limited to, Candida albicans, Cryptococcus neoformans, Histoplama capsulatum, Aspergillus niger, Aspergillus oryzae, Aspergillus fumigatus, Coccidiodes immitis, Paracoccidioides brasiliensis, Blastomyces dermitidis, Pneumocystis carinii, Penicillium marneffi, Alternaria alternate, coccidioides immitits, Fusarium oxysporum, Geotrichum candidum, and Histoplasma capsulatum.


Specific examples of parasites include, but are not limited to, Toxoplasma gondii, Plasmodium falciparum, Plasmodium vivax, Plasmodium malariae, other Plasmodium species, Entamoeba histolytica, Naegleria fowleri, Rhinosporidium seeberi, Giardia lamblia, Enterobius vermicularis, Enterobius gregorii, Ascaris lumbricoides, Ancylostoma duodenale, Necator americanus, Cryptosporidium spp., Trypanosoma brucei, Trypanosoma cruzi, Leishmania major, other Leishmania species, Diphyllobothrium latum, Hymenolepis nana, Hymenolepis diminuta, Echinococcus granulosus, Echinococcus multilocularis, Echinococcus vogeli, Echinococcus oligarthrus, Diphyllobothrium latum, Clonorchis sinensis; Clonorchis viverrini, Fasciola hepatica, Fasciola gigantica, Dicrocoelium dendriticum, Fasciolopsis buski, Metagonimus yokogawai, Opisthorchis viverrini, Opisthorchis felineus, Clonorchis sinensis, Trichomonas vaginalis, Acanthamoeba species, Schistosoma intercalatum, Schistosoma haematobium, Schistosoma japonicum, Schistosoma mansoni, other Schistosoma species, Trichobilharzia regenti, Trichinella spiralis, Trichinella britovi, Trichinella nelsoni, Trichinella nativa, and Entamoeba histolytica.


For example, disclosed herein are methods of identifying a compound for treating or preventing an infection with an infectious microbe in a subject in need thereof, the methods comprising:

    • a) obtaining transcriptomic data from cells infected with the infectious microbe,
    • b) identifying differentially expressed genes (DEGs),
    • c) characterizing host-targeted antimicrobial or anti-cytokine signature,
    • d) identifying compounds that stimulate the anti-microbial or -cytokine signature,
    • e) evaluating known and predicted targets of compounds identified in step d),
    • f) constructing an infection host response protein-protein interaction (PPI) network and modules,
    • g) prioritizing compounds based on network proximity analysis,
    • h) clustering of prioritized compounds associated with selected disease modules,
    • i) selecting representative compounds from each cluster for in vitro assays, and
    • j) analyzing the results of steps a-i to thereby identify the compound for treating or preventing the infection.


In some examples, the infectious microbe comprises a coronavirus.


Also disclosed herein are methods of identifying a compound for treating or preventing a coronavirus infection in a subject in need thereof, the methods comprising:

    • a) obtaining transcriptomic data from coronavirus infected cells,
    • b) identifying differentially expressed genes (DEGs),
    • c) characterizing host-targeted antiviral or anti-cytokine signature,
    • d) identifying compounds that stimulate the anti-viral or -cytokine signature,
    • e) evaluating known and predicted targets of compounds identified in step d),
    • f) constructing a coronavirus infection host response protein-protein interaction (PPI) network and modules,
    • g) prioritizing compounds based on network proximity analysis,
    • h) clustering of prioritized compounds associated with selected disease modules,
    • i) selecting representative compounds from each cluster for in vitro assays, and
    • j) analyzing the results of steps a-i to thereby identify the compound for treating or preventing the infection.


In some examples, the coronavirus comprises human coronavirus, SARS-CoV, SARS-CoV-2, or MERS-CoV.


The infected cells can, for example, comprise infected A549 cells, ACE2-overexpressing A549 cells, or a combination thereof.


In some examples, the differentially expressed genes are identified using Wald test with false-discovery rate (FDR) default upper value of 0.05.


In some examples, the host-targeted antimicrobial, antiviral, and/or anti-cytokine signature is/are characterized using manual curation of gene ontology (GO) enrichment results corresponding to the DEGs.


In some examples, the compounds that stimulate the antimicrobial, antiviral, and/or anti-cytokine signature are identified using Cmap.


In some examples, the known and predicted targets of compounds are evaluated using QuartataWeb.


The compounds can, for example, be prioritized based on network proximity analysis using the lung PPI network in BioSNAP.


In some examples, the in vitro assays can comprise viral inhibition or cell fusion (syncytia) assays.


In some examples, the methods can further comprise considering additional criteria such as drug development status, side effects, mechanism of action (MOA), and antiviral activities of the prioritized compounds in order to identify the compound for treating the infection.


Also disclosed herein are methods of treating or preventing a coronavirus infection in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a composition comprising a compound selected from the group consisting of: imipramine, salmeterol, hexylresorcinol, brompheniramine, ezetimibe, temsirolimus, linsitinib, torn-1, rottlerin, semaxanib, ipratropium, AS-605240, mefenamic acid, JNJ16259685, QL-XII-47, midodrine, olanzapine, trifluoperazine, fluphenazine, azelastine, chlorphenamine, clarithromycin, saracatinib, JAK3-Inhibitor-II, AZD-8055, CGP-60474, hexamethylene, loperamide, nifedipine, liothyronine, atorvastatin, triptolide, pirfenidone, isoliquiritigenin, rucaparib, berbamine, darinaparsin, taurodeoxycholic acid; derivatives thereof; and combinations thereof. In some examples, the composition comprises an antiviral compound, an anti-hyperinflammatory compound, or a combination thereof. In some examples, the coronavirus comprises human coronavirus, SARS-CoV, SARS-CoV-2, or MERS-CoV.


Also disclosed herein are methods of treating or preventing a coronavirus infection in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a composition comprising an antiviral compound and an anti-hyperinflammatory compound. In some examples, the coronavirus comprises human coronavirus, SARS-CoV, SARS-CoV-2, or MERS-CoV.


In some examples, the antiviral compound inhibits cell fusion or viral entry. In some examples, the antiviral compound comprises a histamine receptor antagonist, an acetylcholine receptor antagonist, a norepinephrine and serotonin reuptake inhibitor, an autophagy enhancer, a mTOR inhibitor, a PI3K inhibitor, an IGF-1- and insulin receptor inhibitor, a TB K1 activator through ARF1, an adrenergic receptor agonist, a VEGFR inhibitor, a local anesthetic, a cyclooxygenase inhibitor, a glutamate receptor antagonist, a Niemann-Pick Cl-like 1 protein antagonist, a cholesterol inhibitor, a cytoplasmic tyrosine protein kinase BMX inhibitor, a MAPK and protein kinase inhibitor, or a combination thereof. In some examples, the antiviral compound comprises: imipramine, salmeterol, hexylresorcinol, brompheniramine, ezetimibe, temsirolimus, linsitinib, torin-1, rottlerin, semaxanib, ipratropium, AS-605240, mefenamic acid, JNJ16259685, QL-XII-47; derivatives thereof; or a combination thereof. In some examples, the antiviral compound comprises: salmeterol, rottlerin, imipramine, linsitinib, hexylresorcinol, ezetimibe, brompheniramine; derivatives thereof; or a combination thereof. In some examples, the antiviral compound comprises salmeterol, linisitinib, imipramine, derivatives thereof, or a combination thereof. In some examples, the antiviral compound comprises salmeterol, linisitinib, imipramine, fluvoxamine, or a combination thereof. In some examples, the antiviral compound comprises an IGF-1R and/or insulin receptor inhibitor, such as linsitinib. In some examples, the antiviral compound comprises an adrenergic receptor agonist, such as salmeterol.


In some examples, the anti-hyperinflammatory compound comprises an adrenergic receptor agonist, a dopamine receptor antagonist, an autophagy enhancer, an autophagy dual modulator, a histamine receptor antagonist, a bacterial 50S ribosomal subunit inhibitor, an autophagy inhibitor, a SRC inhibitor, a JAK inhibitor, a mTOR inhibitor, a CDK inhibitor, a sodium/hydrogen antiport inhibitor, an opioid receptor agonist, a calcium channel blocker, a thyroid hormone stimulant, a HMGCR inhibitor, a RNA polymerase inhibitor, a TGFβ receptor inhibitor, an anti-fibrotic, a guanylate cyclase activator, a PARP inhibitor, a calmodulin antagonist, an apoptosis stimulant, a bile acid, or a combination thereof. In some examples, the anti-hyperinflammatory compound comprises midodrine, olanzapine, trifluoperazine, fluphenazine, azelastine, chlorphenamine, clarithromycin, saracatinib, JAK3-Inhibitor-II, AZD-8055, CGP-60474, hexamethylene, loperamide, nifedipine, liothyronine, atorvastatin, triptolide, pirfenidone, isoliquiritigenin, rucaparib, berbamine, darinaparsin, taurodeoxycholic acid, derivatives thereof, or a combination thereof. In some examples, the anti-hyperinflammatory compound elevates IFN signaling and/or suppresses cytokine pathways. In some examples, the anti-hyperinflammatory compound elevates IFN signaling and suppresses cytokine pathways.


In some examples, the composition comprises salmeterol, linsitinib, impramine, derivatives thereof, or a combination thereof, optionally in combination with one or more additional agents.


In some examples, the composition comprises salmeterol in combination with one or more additional agents. In some examples, the composition comprises salmeterol in combination with an RNA-dependent RNA polymerase inhibitor, a 3CL protease inhibitor, or a combination thereof. In some examples, the composition comprises salmeterol in combination with molnupiravir, paxlovid, or a combination thereof. In some examples, the composition comprises salmeterol, molnupiravir, and paxlovid.


In some examples, wherein the composition comprises linsitinib in combination with one or more additional agents.


In some examples, the composition comprises impramine or a derivative thereof in combination with one or more additional agents.


Also disclosed herein are pharmaceutical compositions comprising any of the compositions and/or compounds disclosed herein.


For example, also disclosed herein are pharmaceutical compositions comprising any of the compounds disclosed herein (e.g., a compound identified by any of the methods disclosed herein) and one more additional agents.


Also disclosed herein are compositions comprising the compound identified by any of the methods disclosed herein. In some examples, the composition further comprises a pharmaceutically acceptable excipient.


Also disclosed herein are pharmaceutical compositions comprises a pharmaceutically acceptable excipient and a therapeutically effective amount of any of the compositions disclosed herein.


In some examples, the compositions can further comprise one or more additional agents.


In some examples, the compound comprises imipramine, salmeterol, hexylresorcinol, brompheniramine, ezetimibe, temsirolimus, linsitinib, torin-1, rottlerin, semaxanib, ipratropium, AS-605240, mefenamic acid, JNJ16259685, QL-XII-47, midodrine, olanzapine, trifluoperazine, fluphenazine, azelastine, chlorphenamine, clarithromycin, saracatinib, JAK3-Inhibitor-II, AZD-8055, CGP-60474, hexamethylene, loperamide, nifedipine, liothyronine, atorvastatin, triptolide, pirfenidone, isoliquiritigenin, rucaparib, berbamine, darinaparsin, taurodeoxycholic acid; derivatives thereof; or a combination thereof. In some examples, the compound comprises imipramine, salmeterol, hexylresorcinol, brompheniramine, ezetimibe, temsirolimus, linsitinib, torin-1, rottlerin, semaxanib, ipratropium, AS-605240, mefenamic acid, JNJ16259685, QL-XII-47; derivatives thereof; or a combination thereof. In some examples, the compound comprises salmeterol, rottlerin, imipramine, linsitinib, hexylresorcinol, ezetimibe, brompheniramine; derivatives thereof; or a combination thereof. In some examples, the compound comprises salmeterol, linisitinib, imipramine, derivatives thereof, or a combination thereof. In some examples, the compound comprises salmeterol, linisitinib, imipramine, fluvoxamine, or a combination thereof.


Also disclosed herein are pharmaceutical compositions for the treatment of a coronavirus infection in a subject in need thereof, wherein the pharmaceutical composition comprises a pharmaceutically acceptable excipient and a therapeutically effective amount of a composition comprising a compound selected from the group consisting of: imipramine, salmeterol, hexylresorcinol, brompheniramine, ezetimibe, temsirolimus, linsitinib, torin-1, rottlerin, semaxanib, ipratropium, AS-605240, mefenamic acid, JNJ16259685, QL-XII-47, midodrine, olanzapine, trifluoperazine, fluphenazine, azelastine, chlorphenamine, clarithromycin, saracatinib, JAK3-Inhibitor-II, AZD-8055, CGP-60474, hexamethylene, loperamide, nifedipine, liothyronine, atorvastatin, triptolide, pirfenidone, isoliquiritigenin, rucaparib, berbamine, darinaparsin, taurodeoxycholic acid; derivatives thereof; and combinations thereof. In some examples, the composition comprises an antiviral compound, an anti-hyperinflammatory compound, or a combination thereof.


Also disclosed herein are pharmaceutical compositions for the treatment of coronavirus comprising a pharmaceutically acceptable excipient and a therapeutically effective amount of a composition comprising an antiviral compound and an anti-hyperinflammatory compound.


In some examples, the antiviral compound inhibits cell fusion or viral entry. In some examples, the antiviral compound comprises a histamine receptor antagonist, an acetylcholine receptor antagonist, a norepinephrine and serotonin reuptake inhibitor, an autophagy enhancer, a mTOR inhibitor, a PI3K inhibitor, an IGF-1- and insulin receptor inhibitor, a TB K1 activator through ARF1, an adrenergic receptor agonist, a VEGFR inhibitor, a local anesthetic, a cyclooxygenase inhibitor, a glutamate receptor antagonist, a Niemann-Pick Cl-like 1 protein antagonist, a cholesterol inhibitor, a cytoplasmic tyrosine protein kinase BMX inhibitor, a MAPK and protein kinase inhibitor, or a combination thereof. In some examples, the antiviral compound comprises: imipramine, salmeterol, hexylresorcinol, brompheniramine, ezetimibe, temsirolimus, linsitinib, torin-1, rottlerin, semaxanib, ipratropium, AS-605240, mefenamic acid, JNJ16259685, QL-XII-47; derivatives thereof; or a combination thereof. In some examples, the antiviral compound comprises: salmeterol, rottlerin, imipramine, linsitinib, hexylresorcinol, ezetimibe, brompheniramine; derivatives thereof; or a combination thereof. In some examples, the antiviral compound comprises salmeterol, linisitinib, imipramine, derivatives thereof, or a combination thereof. In some examples, the antiviral compound comprises salmeterol, linisitinib, imipramine, fluvoxamine, or a combination thereof. In some examples, the antiviral compound comprises an IGF-1R and/or insulin receptor inhibitor, such as linsitinib. In some examples, the antiviral compound comprises an adrenergic receptor agonist, such as salmeterol.


In some examples, the anti-hyperinflammatory compound comprises an adrenergic receptor agonist, a dopamine receptor antagonist, an autophagy enhancer, an autophagy dual modulator, a histamine receptor antagonist, a bacterial 50S ribosomal subunit inhibitor, an autophagy inhibitor, a SRC inhibitor, a JAK inhibitor, a mTOR inhibitor, a CDK inhibitor, a sodium/hydrogen antiport inhibitor, an opioid receptor agonist, a calcium channel blocker, a thyroid hormone stimulant, a HMGCR inhibitor, a RNA polymerase inhibitor, a TGFβ receptor inhibitor, an anti-fibrotic, a guanylate cyclase activator, a PARP inhibitor, a calmodulin antagonist, an apoptosis stimulant, a bile acid, or a combination thereof. In some examples, the anti-hyperinflammatory compound comprises midodrine, olanzapine, trifluoperazine, fluphenazine, azelastine, chlorphenamine, clarithromycin, saracatinib, JAK3-Inhibitor-II, AZD-8055, CGP-60474, hexamethylene, loperamide, nifedipine, liothyronine, atorvastatin, triptolide, pirfenidone, isoliquiritigenin, rucaparib, berbamine, darinaparsin, taurodeoxycholic acid, derivatives thereof, or a combination thereof. In some examples, the anti-hyperinflammatory compound elevates IFN signaling and/or suppresses cytokine pathways. In some examples, the anti-hyperinflammatory compound elevates IFN signaling and suppresses cytokine pathways.


In some examples, the composition comprises salmeterol, linsitinib, impramine, derivatives thereof, or a combination thereof, optionally in combination with one or more additional agents.


In some examples, the composition comprises salmeterol in combination with one or more additional agents. In some examples, the composition comprises salmeterol in combination with an RNA-dependent RNA polymerase inhibitor, a 3CL protease inhibitor, or a combination thereof. In some examples, the composition comprises salmeterol in combination with molnupiravir, paxlovid, or a combination thereof. In some examples, the composition comprises salmeterol, molnupiravir, and paxlovid.


In some examples, wherein the composition comprises linsitinib in combination with one or more additional agents.


In some examples, the composition comprises impramine or a derivative thereof in combination with one or more additional agents.


In some examples, the compositions can further comprise one or more additional agents.


Also disclosed herein are methods of treating a disease or disorder in a subject in need thereof comprising administering a therapeutically effective amount of any of the compositions (e.g., pharmaceutical compositions) as disclosed herein. In some examples, the disease or disorder comprises an infection, such as with an infectious microbe (e.g., bacteria, virus, fungi, protozoa, etc.). In some examples, the disease or disorder comprises an infection with a coronavirus. In some examples, the coronavirus comprises human coronavirus, SARS-CoV, SARS-CoV-2, or MERS-CoV.


The methods of treatment of the disease or disorder described herein can further include treatment with one or more additional agents. The one or more additional agents and the compounds as described herein can be administered in any order, including simultaneous administration, as well as temporally spaced order of up to several days apart. The methods can also include more than a single administration of the one or more additional agents and/or the compounds or compositions as described herein. The administration of the one or more additional agents and the compounds or compositions as described herein can be by the same or different routes. When treating with one or more additional agents, the compounds or compositions as described herein can be combined into a pharmaceutical composition that includes the one or more additional agents.


The one or more additional agents can, for example, comprise an anti-inflammatory agent, an antimicrobial agent, or a combination thereof. As used herein, antimicrobials include, for example, antibacterials, antifungals, and antivirals. Accordingly, in some examples, the methods can further include treatment with one or more additional antiviral agents, anti-inflammatory agents, or a combination thereof.


Examples of antimicrobial agents include, but are not limited to, alexidine, asphodelin A, atromentin, auranthine, austrocortilutein, austrocortirubin, azerizin, chlorbisan, chloroxine, cidex, cinoxacin, citreorosein, copper usnate, cupiennin, curvularin, DBNPA, dehydrocurvularin, desoxyfructo-serotonin, dichloroisocyanuric acid, elaiomycin, holtfreter's solution, malettinin, naphthomycin, neutrolin, niphimycin, nitrocefin, oxadiazoles, paenibacterin, proclin, ritiometan, ritipenem, silicone quaternary amine, stylisin, taurolidine, tirandamycin, trichloroisocyanuric acid, triclocarban, and combinations thereof.


Examples of antibacterials include, but are not limited to, acetoxycycloheximide, aciduliprofundum, actaplanin, actinorhodin, alazopeptin, albomycin, allicin, allistatin, allyl isothiocyanate, ambazone, aminocoumarin, aminoglycosides, 4-aminosalicylic acid, ampicillin, ansamycin, anthramycin, antimycin A, aphidicolin, aplasmomycin, archaeocin, arenicin, arsphenamine, arylomycin A2, ascofuranone, aspergillic acid, avenanthramide, avibactam, azelaic acid, bafilomycin, bambermycin, beauvericin, benzoyl peroxide, blasticidin S, bottromycin, brilacidin, caprazamycin, carbomycin, cathelicidin, cephalosporins, ceragenin, chartreusin, chromomycin A3, citromycin, clindamycin, clofazimine, clofoctol, clorobiocin, coprinol, coumermycin A1, cyclic lipopeptides, cycloheximide, cycloserine, dalfopristin, dapsone, daptomycin, debromomarinone, 17-dimethylaminoethylamino-17-demethoxygeldanamycin, echinomycin, endiandric acid C, enediyne, enviomycin, eravacycline, erythromycin, esperamicin, etamycin, ethambutol, ethionamide, (6S)-6-fluoroshikimic acid, fosfomycin, fosmidomycin, friulimicin, furazolidone, furonazide, fusidic acid, geldanamycin, gentamycin, gepotidacin, glycyciclines, glycyrrhizol, gramicidin S, guanacastepene A, hachimycin, halocyamine, hedamycin, helquinoline, herbimycin, hexamethylenetetramine, hitachimycin, hydramacin-1, isoniazid, kanamycin, katanosin, kedarcidin, kendomycin, kettapeptin, kidamycin, lactivicin, lactocillin, landomycin, landomycinone, lasalocid, lenapenem, leptomycin, lincosamides, linopristin, lipiarmycins, macbecin, macrolides, macromomycin B, maduropeptin, mannopeptimycin glycopeptide, marinone, meclocycline, melafix, methylenomycin A, methylenomycin B, monensin, moromycin, mupirocin, mycosubtilin, myriocin, myxopyronin, naphthomycin A, narasin, neocarzinostatin, neopluramycin, neosalvarsan, neothramycin, netropsin, nifuroxazide, nifurquinazol, nigericin, nitrofural, nitrofurantoin, nocathiacin I, novobiocin, omadacycline, oxacephem, oxazolidinones, penicillins, peptaibol, phytoalexin, plantazolicin, platensimycin, plectasin, pluramycin A, polymixins, polyoxins, pristinamycin, pristinamycin IA, promin, prothionamide, pulvinone, puromycin, pyocyanase, pyocyanin, pyrenocine, questiomycin A, quinolones, quinupristin, ramoplanin, raphanin, resistome, reuterin, rifalazil, rifamycins, ristocetin, roseophilin, salinomycin, salinosporamide A, saptomycin, saquayamycin, seraticin, sideromycin, sodium sulfacetamide, solasulfone, solithromycin, sparassol, spectinomycin, staurosporine, streptazolin, streptogramin, streptogramin B, streptolydigin, streptonigrin, styelin A, sulfonamides, surfactin, surotomycin, tachyplesin, taksta, tanespimycin, telavancin, tetracyclines, thioacetazone, thiocarlide, thiolutin, thiostrepton, tobramycin, trichostatin A, triclosan, trimethoprim, trimethoprim, tunicamycin, tyrocidine, urauchimycin, validamycin, viridicatumtoxin B, vulgamycin, xanthomycin A, xibornol, amikacin, amoxicillin, ampicillin, atovaquone, azithromycin, aztreonam, bacitracin, carbenicillin, cefadroxil, cefazolin, cefdinir, cefditoren, cefepime, cefiderocol, cefoperazone, cefotetan, cefoxitin, cefotaxime, cefpodoxime, cefprozil, ceftaroline, ceftazidime, ceftibuten, ceftizoxime, ceftriaxone, chloramphenicol, colistimethate, cefuroxime, cephalexin, cephradine, cilastatin, cinoxacin, ciprofloxacin, clarithromycin, clindamycin, dalbavancin, dalfopristin, daptomycin, demeclocycline, dicloxacillin, doripenem, doxycycline, eravacycline, ertapenem, erythromycin, fidaxomicin, fosfomycin, gatifloxacin, gemifloxacin, gentamicin, imipenem, lefamulin, lincomycin, linezolid, lomefloxacin, loracarbef, meropenem, metronidazole, minocycline, moxifloxacin, nafcillin, nalidixic acid, neomycin, norfloxacin, ofloxacin, omadacycline, oritavancin, oxacillin, oxytetracycline, paromomycin, penicillin, pentamidine, piperacillin, plazomicin, quinupristin, rifaximin, sarecycline, secnidazole, sparfloxacin, spectinomycin, sulfamethoxazole, sulfisoxazole, tedizolid, telavancin, telithromycin, ticarcillin, tigecycline, tobramycin, trimethoprim, trovafloxacin, vancomycin, and combinations thereof.


Examples of antifungals include, but are not limited to, abafungin, acibenzolar, acibenzolar-S-methyl, acrisorcin, allicin, aminocandin, amorolfine, amphotericin B, anidulafungin, azoxystrobin, bacillomycin, Bacillus pumilus, barium borate, benomyl, binapacryl, boric acid, bromine monochloride, bromochlorosalicylanilide, bupirimate, butenafine, candicidin, caprylic acid, captafol, captan, carbendazim, caspofungin, cerulenin, chloranil, chlormidazole, chlorophetanol, chlorothalonil, chloroxylenol, chromated copper arsenate, ciclopirox, cilofungin, cinnamaldehyde, clioquinol, copper(I) cyanide, copper(II) arsenate, cruentaren, cycloheximide, davicil, dehydroacetic acid, dicarboximide fungicides, dichlofluanid, dimazole, diphenylamine, echinocandin, echinocandin B, epoxiconazole, ethonam, falcarindiol, falcarinol, famoxadone, fenamidone, fenarimol, fenpropimorph, fentin acetate, fenticlor, filipin, fluazinam, fluopicolide, flusilazole, fluxapyroxad, fuberidazole, griseofulvin, halicylindramide, haloprogin, hamycin, hexachlorobenzene, hexachlorocyclohexa-2,5-dien-1-one, 5-hydroxy-2(5H)-furanone, iprodione, lime sulfur, mancozeb, maneb, melafix, metalaxyl, metam sodium, methylisothiazolone, methylparaben, micafungin, miltefosine, monosodium methyl arsenate, mycobacillin, myclobutanil, natamycin, beta-nitrostyrene, nystatin, paclobutrazol, papulacandin B, parietin, pecilocin, pencycuron, pentamidine, pentachloronitrobenzene, pentachlorophenol, perimycin, 2-phenylphenol, polyene antimycotic, propamocarb, propiconazole, pterulone, ptilomycalin A, pyrazophos, pyrimethanil, pyrrolnitrin, selenium disulfide, sparassol, strobilurin, sulbentine, tavaborole, tebuconazole, terbinafine, theonellamide F, thymol, tiabendazole, ticlatone, tolciclate, tolnaftate, triadimefon, triamiphos, tribromometacresol, 2,4,6-tribromophenol, tributyltin oxide, triclocarban, triclosan, tridemorph, trimetrexate, undecylenic acid, validamycin, venturicidin, vinclozolin, vinyldithiin, vusion, xanthene, zinc borate, zinc pyrithione, zineb, ziram, voriconazole, itraconazole, posaconazole, fluconazole, ketoconazole, clotrimazole, isavuconazonium, miconazole, caspofungin, anidulafungin, micafungin, griseofulvin, terbinafine, flucytosine, terbinafine, nystatin, amphotericin b., and combinations thereof.


Examples of antivirals include, but are not limited to, afovirsen, alisporivir, angustific acid, angustifodilactone, alovudine, beclabuvir, 2,3-bis(acetylmercaptomethyl)quinoxaline, brincidofovir, dasabuvir, docosanol, fialuridine, ibacitabine, imiquimod, inosine, inosine pranobex, interferon, metisazone, miltefosine, neokadsuranin, neotripterifordin, ombitasvir, oragen, oseltamivir, pegylated interferon, podophyllotoxin, radalbuvir, semapimod, tecovirimat, telbivudine, theaflavin, tilorone, triptofordin C-2, variecolol, ZMapp, abacavir, acyclovir, adefovir, amantadine, amprenavir, atazanavir, balavir, baloxavir marboxil, boceprevir, cidofovir, cobicistat, daclatasvir, darunavir, delavirdine, didanosine, docasanol, dolutegravir, doravirine, ecoliever, edoxudine, efavirenz, elvitegravir, emtricitabine, enfuvirtide, entecavir, etravirine, famciclovir, fomivirsen, fosamprenavir, forscarnet, fosnonet, famciclovir, favipravir, fomivirsen, foscavir, ganciclovir, ibacitabine, idoxuridine, indinavir, inosine, inosine pranobex, interferon type I, interferon type II, interferon type III, lamivudine, letermovir, lopinavir, loviride, maraviroc, methisazone, moroxydine, nelfinavir, nevirapine, nitazoxanide, oseltamivir, peginterferon alfa-2a, peginterferon alfa-2b, penciclovir, peramivir, pleconaril, podophyllotoxin, pyramidine, raltegravir, remdesevir, ribavirin, rilpivirine, rimantadine, rintatolimod, ritonavir, saquinavir, simeprevir, sofosbuvir, stavudine, tarabivirin, telaprevir, telbivudine, tenofovir alafenamide, tenofovir disoproxil, tenofovir, tipranavir, trifluridine, trizivir, tromantadine, umifenovir, valaciclovir, valganciclovir, vidarabine, zalcitabine, zanamivir, zidovudine. and combinations thereof.


Examples of suitable immunotherapeutic agents include, but are not limited to, alemtuzumab, cetuximab (ERBITUX), gemtuzumab, iodine 131 tositumomab, rituximab, trastuzamab (HERCEPTIN), and combinations thereof.


In some examples, the one or more additional agents can comprise an anti-inflammatory agent, such as steroidal and/or non-steroidal anti-inflammatory agents. Examples of steroidal anti-inflammatory agents include, but are not limited to, hydrocortisone, dexamethasone, prednisolone, prednisone, triamcinolone, methylprednisolone, budesonide, betamethasone, cortisone, and deflazacort. Examples of non-steroidal anti-inflammatory drugs include acetaminophen, aspirin, ibuprofen, naproxen, Celebrex, ketoprofen, tolmetin, etodolac, fenoprofen, flurbiprofen, diclofenac, piroxicam, indomethacin, sulindax, meloxicam, nabumetone, oxaprozin, mefenamic acid, and diflunisal.


In some examples, the one or more additional agents comprises a nucleic acid. Particular nucleic acid examples include, but are not limited to, oligonucleotides, miRNA, saRNA, shRNA, siRNA, DNA, RNA, mRNA, cDNA, double stranded nucleic acid, single stranded nucleic acid, and so forth. In some examples, the nucleic acid encodes a protein or peptide, e.g. for therapeutic use.


In some examples, the one or more additional agents can comprise an RNA-dependent RNA polymerase inhibitor, a 3CL protease inhibitor, or a combination thereof.


In some examples, the one or more additional agents comprises molnupiravir, paxlovid, or a combination thereof.


In some examples, the one or more additional agents can comprise an antiviral agent(s) selected from the group consisting of abacavir, acyclovir, adefovir, amantadine, amprenavir, ampligen, arbidol, atazanavir, atripla, balapiravir, BCX4430/Galidesivir, boceprevir, cidofovir, combivir, daclatasvir, darunavir, dasabuvir, delavirdine, didanosine, docosanol, edoxudine, efavirenz, emtricitabine, enfuvirtide, entecavir, famciclovir, favipiravir, fomivirsen, fosamprenavir, foscarnet, fosfonet, ganciclovir, GS-5734/remdesivir, ibacitabine, imunovir, idoxuridine, imiquimod, indinavir, inosine, interferon type III, interferon type II, interferon type I, lamivudine, ledipasvir, lopinavir, loviride, maraviroc, moroxydine, methisazone, nelfinavir, nevirapine, nexavir, NITD008, ombitasvir, oseltamivir, paritaprevir, peginterferon alfa-2a, penciclovir, peramivir, pleconaril, podophyllotoxin, raltegravir, ribavirin, rimantadine, ritonavir, pyramidine, saquinavir, simeprevir, sofosbuvir, stavudine, telaprevir, telbivudine, tenofovir, tenofovir disoproxil, Tenofovir Exalidex, tipranavir, trifluridine, trizivir, tromantadine, truvada, valaciclovir, valganciclovir, vicriviroc, vidarabine, viramidine zalcitabine, zanamivir, zidovudine, and combinations thereof.


Effective amounts of a compound or composition described herein for treating a mammalian subject can, in some examples, be 1 microgram (μg) per kilogram (kg) of body weight of the subject per day (μg/kg/day) or more (e.g., 5 μg/kg/day or more, 10 μg/kg/day or more, 15 μg/kg/day or more, 20 μg/kg/day or more, 25 μg/kg/day or more, 30 μg/kg/day or more, 35 μg/kg/day or more, 40 μg/kg/day or more, 45 μg/kg/day or more, 50 μg/kg/day or more, 60 μg/kg/day or more, 70 μg/kg/day or more, 80 μg/kg/day or more, 90 μg/kg/day or more, 100 μg/kg/day or more, 125 μg/kg/day or more, 150 μg/kg/day or more, 175 μg/kg/day or more, 200 μg/kg/day or more, 225 μg/kg/day or more, 250 μg/kg/day or more, 300 μg/kg/day or more, 350 μg/kg/day or more, 400 μg/kg/day or more, 450 μg/kg/day or more, 500 μg/kg/day or more, 600 μg/kg/day or more, 700 μg/kg/day or more, 800 μg/kg/day or more, 900 μg/kg/day or more, 1 milligram (mg) per kilogram (kg) of body weight of the subject per day (mg/kg/day) or more, 5 mg/kg/day or more, 10 mg/kg/day or more, 15 mg/kg/day or more, 20 mg/kg/day or more, 25 mg/kg/day or more, 30 mg/kg/day or more, 35 mg/kg/day or more, 40 mg/kg/day or more, 45 mg/kg/day or more, 50 mg/kg/day or more, 60 mg/kg/day or more, 70 mg/kg/day or more, 80 mg/kg/day or more, 90 mg/kg/day or more, 100 mg/kg/day or more, 125 mg/kg/day or more, 150 mg/kg/day or more, 175 mg/kg/day or more, 200 mg/kg/day or more, 225 mg/kg/day or more, 250 mg/kg/day or more, 300 mg/kg/day or more, 350 mg/kg/day or more, 400 mg/kg/day or more, 450 mg/kg/day or more, 500 mg/kg/day or more, 600 mg/kg/day or more, 700 mg/kg/day or more, 800 mg/kg/day or more, or 900 mg/kg/day or more). In some examples, effective amounts of a compound or composition described herein for treating a mammalian subject can be 1000 milligrams (mg) per kilogram (kg) of body weight of the subject per day (mg/kg/day) or less (e.g., 900 mg/kg/day or less, 800 mg/kg/day or less, 700 mg/kg/day or less, 600 mg/kg/day or less, 500 mg/kg/day or less, 450 mg/kg/day or less, 400 mg/kg/day or less, 350 mg/kg/day or less, 300 mg/kg/day or less, 250 mg/kg/day or less, 225 mg/kg/day or less, 200 mg/kg/day or less, 175 mg/kg/day or less, 150 mg/kg/day or less, 125 mg/kg/day or less, 100 mg/kg/day or less, 90 mg/kg/day or less, 80 mg/kg/day or less, 70 mg/kg/day or less, 60 mg/kg/day or less, 50 mg/kg/day or less, 45 mg/kg/day or less, 40 mg/kg/day or less, 35 mg/kg/day or less, 30 mg/kg/day or less, 25 mg/kg/day or less, 20 mg/kg/day or less, 15 mg/kg/day or less, 10 mg/kg/day or less, 5 mg/kg/day or less, 1 mg/kg/day or less, 900 microgram (μg) per kilogram (kg) of body weight of the subject per day (μg/kg/day) or less, 800 μg/kg/day or less, 700 μg/kg/day or less, 600 μg/kg/day or less, 500 μg/kg/day or less, 450 μg/kg/day or less, 400 μg/kg/day or less, 350 μg/kg/day or less, 300 μg/kg/day or less, 250 μg/kg/day or less, 225 μg/kg/day or less, 200 μg/kg/day or less, 175 μg/kg/day or less, 150 μg/kg/day or less, 125 μg/kg/day or less, 100 μg/kg/day or less, 90 μg/kg/day or less, 80 μg/kg/day or less, 70 μg/kg/day or less, 60 μg/kg/day or less, 50 μg/kg/day or less, 45 μg/kg/day or less, 40 μg/kg/day or less, 35 μg/kg/day or less, 30 μg/kg/day or less, 25 μg/kg/day or less, 20 μg/kg/day or less, 15 μg/kg/day or less, 10 μg/kg/day or less, or 5 μg/kg/day or less).


Effective amounts of a compound or composition described herein for treating a mammalian subject can range from any of the minimum values described above to any of the maximum values described above. For example, effective amounts of a compound or composition described herein for treating a mammalian subject can include from 1 microgram (μg) per kilogram (kg) of body weight of the subject per day (mg/kg/day) to 1000 mg per kg of body weight of the subject per day (e.g., from 1 mg/kg/day to 1 mg/kg/day, from 1 mg/kg/day to 1000 mg/kg/day, from 1 μg/kg/day to 100 μg/kg/day, from 100 μg/kg/day to 1 mg/kg/day, from 1 mg/kg/day to 100 mg/kg/day, from 100 mg/kg/day to 1000 mg/kg/day, from 5 μg/kg/day to 1000 mg/kg/day, from 1 μg/kg/day to 900 mg/kg/day, from 5 μg/kg/day to 900 mg/kg/day, from 500 mg/kg/day to 500 mg/kg/day, from 1 to 100 mg/kg/day, or from 10 to 100 mg/kg/day). The doses can be acute or chronic. A broad range of disclosed composition dosages are believed to be both safe and effective.


It is understood, however, that the specific dose level for any particular subject will depend upon a variety of factors. Such factors include the age, body weight, general health, sex, and diet of the subject. Other factors include the time and route of administration, rate of excretion, drug combination, and the type and severity of the particular disease or disorder.


The methods, compounds, and compositions as described herein are useful for both prophylactic and therapeutic treatment. As used herein the term treating or treatment includes prevention; delay in onset; diminution, eradication, or delay in exacerbation of signs or symptoms after onset; and prevention of relapse. For prophylactic use, a therapeutically effective amount of the compounds or compositions as described herein are administered to a subject prior to onset (e.g., before obvious signs of the disease or disorder), during early onset (e.g., upon initial signs and symptoms of the disease or disorder), or after an established development of the disease or disorder. Prophylactic administration can occur for several days to years prior to the manifestation of symptoms of a disease or disorder. Therapeutic treatment involves administering to a subject a therapeutically effective amount of the compound or composition as described herein after the disease or disorder is diagnosed.


In certain embodiments, it is desirable to target a nanoparticle using a targeting moiety that is specific to a cell type and/or tissue type. In some embodiments, a nanoparticle may be targeted to a particular cell, tissue, and/or organ using a targeting moiety. Exemplary non-limiting targeting moieties include ligands, cell surface receptors, glycoproteins, vitamins (e.g., riboflavin) and antibodies (e.g., full-length antibodies, antibody fragments (e.g., Fv fragments, single chain Fv (scFv) fragments, Fab′ fragments, or F(ab′)2 fragments), single domain antibodies, camelid antibodies and fragments thereof, human antibodies and fragments thereof, monoclonal antibodies, and multispecific antibodies (e.g., bispecific antibodies)). In some embodiments, the targeting moiety may be a polypeptide. The targeting moiety may include the entire polypeptide (e.g., peptide or protein) or fragments thereof. A targeting moiety is typically positioned on the outer surface of the nanoparticle in such a manner that the targeting moiety is available for interaction with the target, for example, a cell surface receptor. A variety of different targeting moieties and methods are known and available in the art, including those described, e.g., in Sapra et al., Prog. Lipid Res. 42(5):439-62, 2003 and Abra et al., J. Liposome Res. 12:1-3, 2002.


The targeting moiety can target any known cell type, including, but not limited to, hepatocytes, colon cells, epithelial cells, hematopoietic cells, epithelial cells, endothelial cells, lung cells, bone cells, stem cells, mesenchymal cells, neural cells, cardiac cells, adipocytes, vascular smooth muscle cells, cardiomyocytes, skeletal muscle cells, beta cells, pituitary cells, synovial lining cells, ovarian cells, testicular cells, fibroblasts, B cells, T cells, reticulocytes, leukocytes, granulocytes, and tumor cells (including primary tumor cells and metastatic tumor cells).


In some examples, the pharmaceutical composition is administered to a subject. In some examples, the subject is a mammal. In some examples, the mammal is a primate. In some examples, the mammal is a human. In some examples, the human is a patient.


In some examples, the disclosed compositions comprise the disclosed compounds (including pharmaceutically acceptable salt(s) thereof) as an active ingredient, a pharmaceutically acceptable carrier, and, optionally, other therapeutic ingredients or adjuvants. The instant compositions include those suitable for oral, rectal, topical, and parenteral (including subcutaneous, intramuscular, and intravenous) administration, although the most suitable route in any given case will depend on the particular host, and nature and severity of the conditions for which the active ingredient is being administered. The compositions can be conveniently presented in unit dosage form and prepared by any of the methods well known in the art of pharmacy.


Methods of Making


Also disclosed herein are methods of making any of the compounds or compositions disclosed herein.


The compounds described herein can be prepared in a variety of ways known to one skilled in the art of organic synthesis or variations thereon as appreciated by those skilled in the art. The compounds described herein can be prepared from readily available starting materials. Optimum reaction conditions can vary with the particular reactants or solvents used, but such conditions can be determined by one skilled in the art.


Variations on the compounds described herein include the addition, subtraction, or movement of the various constituents as described for each compound. Similarly, when one or more chiral centers are present in a molecule, the chirality of the molecule can be changed. Additionally, compound synthesis can involve the protection and deprotection of various chemical groups. The use of protection and deprotection, and the selection of appropriate protecting groups can be determined by one skilled in the art. The chemistry of protecting groups can be found, for example, in Wuts and Greene, Protective Groups in Organic Synthesis, 4th Ed., Wiley & Sons, 2006, which is incorporated herein by reference in its entirety.


The starting materials and reagents used in preparing the disclosed compounds and compositions are either available from commercial suppliers such as Katchem (Prague, Czech Republic), Aldrich Chemical Co., (Milwaukee, WI), Acros Organics (Morris Plains, NJ), Fisher Scientific (Pittsburgh, PA), Sigma (St. Louis, MO), Pfizer (New York, NY), GlaxoSmithKline (Raleigh, NC), Merck (Whitehouse Station, NJ), Johnson & Johnson (New Brunswick, NJ), Aventis (Bridgewater, NJ), AstraZeneca (Wilmington, DE), Novartis (Basel, Switzerland), Wyeth (Madison, NJ), Bristol-Myers-Squibb (New York, NY), Roche (Basel, Switzerland), Lilly (Indianapolis, IN), Abbott (Abbott Park, Ill.), Schering Plough (Kenilworth, NJ), or Boehringer Ingelheim (Ingelheim, Germany), or are prepared by methods known to those skilled in the art following procedures set forth in references such as Fieser and Fieser's Reagents for Organic Synthesis, Volumes 1-17 (John Wiley and Sons, 1991); Rodd's Chemistry of Carbon Compounds, Volumes 1-5 and Supplementals (Elsevier Science Publishers, 1989); Organic Reactions, Volumes 1-40 (John Wiley and Sons, 1991); March's Advanced Organic Chemistry, (John Wiley and Sons, 4th Edition); and Larock's Comprehensive Organic Transformations (VCH Publishers Inc., 1989). Other materials, such as the pharmaceutical excipients disclosed herein can be obtained from commercial sources.


Reactions to produce the compounds described herein can be carried out in solvents, which can be selected by one of skill in the art of organic synthesis. Solvents can be substantially nonreactive with the starting materials (reactants), the intermediates, or products under the conditions at which the reactions are carried out, i.e., temperature and pressure. Reactions can be carried out in one solvent or a mixture of more than one solvent. Product or intermediate formation can be monitored according to any suitable method known in the art. For example, product formation can be monitored by spectroscopic means, such as nuclear magnetic resonance spectroscopy (e.g., 1H or 13C) infrared spectroscopy, spectrophotometry (e.g., UV-visible), or mass spectrometry, or by chromatography such as high performance liquid chromatography (HPLC) or thin layer chromatography.


Compositions, Formulations, Methods of Administration, and Kits


In vivo application of the disclosed compounds, and compositions containing them, can be accomplished by any suitable method and technique presently or prospectively known to those skilled in the art. For example, the disclosed compounds can be formulated in a physiologically- or pharmaceutically-acceptable form and administered by any suitable route known in the art including, for example, oral, nasal, rectal, topical, and parenteral routes of administration. As used herein, the term parenteral includes subcutaneous, intradermal, intravenous, intramuscular, intraperitoneal, and intrasternal administration, such as by injection. Administration of the disclosed compounds or compositions can be a single administration, or at continuous or distinct intervals as can be readily determined by a person skilled in the art.


The compounds disclosed herein, and compositions comprising them, can also be administered utilizing liposome technology, slow release capsules, implantable pumps, and biodegradable containers. These delivery methods can, advantageously, provide a uniform dosage over an extended period of time. The compounds can also be administered in their salt derivative forms or crystalline forms.


The compounds disclosed herein can be formulated according to known methods for preparing pharmaceutically acceptable compositions. Formulations are described in detail in a number of sources which are well known and readily available to those skilled in the art. For example, Remington's Pharmaceutical Science by E. W. Martin (1995) describes formulations that can be used in connection with the disclosed methods. In general, the compounds disclosed herein can be formulated such that an effective amount of the compound is combined with a suitable excipient in order to facilitate effective administration of the compound. The compositions used can also be in a variety of forms. These include, for example, solid, semisolid, and liquid dosage forms, such as tablets, pills, powders, liquid solutions or suspension, suppositories, injectable and infusible solutions, and sprays. The preferred form depends on the intended mode of administration and application. The compositions can also include conventional pharmaceutically-acceptable carriers and diluents which are known to those skilled in the art.


Examples of carriers or diluents for use with the compounds include ethanol, dimethyl sulfoxide, glycerol, alumina, starch, saline, and equivalent carriers and diluents. To provide for the administration of such dosages for the desired application, compositions disclosed herein can comprise between about 0.1% and 100% by weight of the total of one or more of the subject compounds based on the weight of the total composition including carrier or diluent.


The pharmaceutical carrier employed can be, for example, a solid, liquid, or gas. Examples of solid carriers include lactose, terra alba, sucrose, talc, gelatin, agar, pectin, acacia, magnesium stearate, and stearic acid. Examples of liquid carriers are sugar syrup, peanut oil, olive oil, and water. Examples of gaseous carriers include carbon dioxide and nitrogen.


Formulations suitable for administration include, for example, aqueous sterile injection solutions, which can contain antioxidants, buffers, bacteriostats, and solutes that render the formulation isotonic with the blood of the intended recipient; and aqueous and nonaqueous sterile suspensions, which can include suspending agents and thickening agents. The formulations can be presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and can be stored in a freeze dried (lyophilized) condition requiring only the condition of the sterile liquid carrier, for example, water for injections, prior to use. Extemporaneous injection solutions and suspensions can be prepared from sterile powder, granules, tablets, etc. It should be understood that in addition to the excipients particularly mentioned above, the compositions disclosed herein can include other agents conventional in the art having regard to the type of formulation in question.


Compounds disclosed herein, and compositions comprising them, can be delivered to a cell either through direct contact with the cell or via a carrier means. Carrier means for delivering compounds and compositions to cells are known in the art.


For the treatment of oncological disorders, the compounds or compositions disclosed herein can be administered to a patient in need of treatment in combination with other antitumor or anticancer substances and/or with radiation and/or photodynamic therapy and/or with surgical treatment to remove a tumor. These other substances or treatments can be given at the same as or at different times from the compounds or compositions disclosed herein. For example, the compounds or compositions disclosed herein can be used in combination with mitotic inhibitors such as taxol or vinblastine, alkylating agents such as cyclophosamide or ifosfamide, antimetabolites such as 5-fluorouracil or hydroxyurea, DNA intercalators such as adriamycin or bleomycin, topoisomerase inhibitors such as etopo side or camptothecin, antiangiogenic agents such as angiostatin, antiestrogens such as tamoxifen, and/or other anti-cancer drugs or antibodies, such as, for example, GLEEVEC (Novartis Pharmaceuticals Corporation) and HERCEPTIN (Genentech, Inc.), respectively, or an immunotherapeutic such as ipilimumab and bortezomib.


In certain examples, compounds and compositions disclosed herein can be locally administered at one or more anatomical sites, such as sites of unwanted cell growth (such as a tumor site or benign skin growth, e.g., injected or topically applied to the tumor or skin growth), optionally in combination with a pharmaceutically acceptable carrier such as an inert diluent. Compounds and compositions disclosed herein can be systemically administered, such as intravenously or orally, optionally in combination with a pharmaceutically acceptable carrier such as an inert diluent, or an assimilable edible carrier for oral delivery. They can be enclosed in hard or soft shell gelatin capsules, can be compressed into tablets, or can be incorporated directly with the food of the patient's diet. For oral therapeutic administration, the active compound can be combined with one or more excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, aerosol sprays, and the like.


The tablets, troches, pills, capsules, and the like can also contain the following: binders such as gum tragacanth, acacia, corn starch or gelatin; diluents such as dicalcium phosphate; a disintegrating agent such as corn starch, potato starch, alginic acid and the like; a lubricant such as magnesium stearate; and a sweetening agent such as sucrose, fructose, lactose or aspartame or a flavoring agent such as peppermint, oil of wintergreen, or cherry flavoring can be added. When the unit dosage form is a capsule, it can contain, in addition to materials of the above type, a liquid carrier, such as a vegetable oil or a polyethylene glycol. Various other materials can be present as coatings or to otherwise modify the physical form of the solid unit dosage form. For instance, tablets, pills, or capsules can be coated with gelatin, wax, shellac, or sugar and the like. A syrup or elixir can contain the active compound, sucrose or fructose as a sweetening agent, methyl and propylparabens as preservatives, a dye and flavoring such as cherry or orange flavor. Of course, any material used in preparing any unit dosage form should be pharmaceutically acceptable and substantially non-toxic in the amounts employed. In addition, the active compound can be incorporated into sustained-release preparations and devices.


Compounds and compositions disclosed herein, including pharmaceutically acceptable salts thereof, can be administered intravenously, intramuscularly, or intraperitoneally by infusion or injection. Solutions of the active agent or its salts can be prepared in water, optionally mixed with a nontoxic surfactant. Dispersions can also be prepared in glycerol, liquid polyethylene glycols, triacetin, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations can contain a preservative to prevent the growth of microorganisms.


The pharmaceutical dosage forms suitable for injection or infusion can include sterile aqueous solutions or dispersions or sterile powders comprising the active ingredient, which are adapted for the extemporaneous preparation of sterile injectable or infusible solutions or dispersions, optionally encapsulated in liposomes. The ultimate dosage form should be sterile, fluid and stable under the conditions of manufacture and storage. The liquid carrier or vehicle can be a solvent or liquid dispersion medium comprising, for example, water, ethanol, a polyol (for example, glycerol, propylene glycol, liquid polyethylene glycols, and the like), vegetable oils, nontoxic glyceryl esters, and suitable mixtures thereof. The proper fluidity can be maintained, for example, by the formation of liposomes, by the maintenance of the required particle size in the case of dispersions or by the use of surfactants. Optionally, the prevention of the action of microorganisms can be brought about by various other antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars, buffers or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by the inclusion of agents that delay absorption, for example, aluminum monostearate and gelatin.


Pharmaceutical compositions disclosed herein suitable for injectable use include sterile aqueous solutions or dispersions. Furthermore, the compositions can be in the form of sterile powders for the extemporaneous preparation of such sterile injectable solutions or dispersions. In some examples, the final injectable form can be sterile and can be effectively fluid for easy syringability. In some examples, the pharmaceutical compositions can be stable under the conditions of manufacture and storage; thus, they can be preserved against the contaminating action of microorganisms such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (e.g., glycerol, propylene glycol and liquid polyethylene glycol), vegetable oils, and suitable mixtures thereof.


Sterile injectable solutions are prepared by incorporating a compound and/or agent disclosed herein in the required amount in the appropriate solvent with various other ingredients enumerated above, as required, followed by filter sterilization. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum drying and the freeze drying techniques, which yield a powder of the active ingredient plus any additional desired ingredient present in the previously sterile-filtered solutions.


Pharmaceutical compositions disclosed herein can be in a form suitable for topical use such as, for example, an aerosol, cream, ointment, lotion, dusting powder, mouth washes, gargles, solution, tincture, and the like. In some examples, the compositions can be in a form suitable for use in transdermal devices. In some examples, it will be desirable to administer them topically to the skin as compositions, in combination with a dermatologically acceptable carrier, which can be a solid or a liquid. Compounds and agents and compositions disclosed herein can be applied topically to a subject's skin. These formulations can be prepared, utilizing any of the compounds disclosed herein or pharmaceutically acceptable salts thereof, via conventional processing methods.


Useful solid carriers include finely divided solids such as talc, clay, microcrystalline cellulose, silica, alumina and the like. Useful liquid carriers include water, alcohols or glycols or water-alcohol/glycol blends, in which the compounds can be dissolved or dispersed at effective levels, optionally with the aid of non-toxic surfactants. Adjuvants such as fragrances and additional antimicrobial agents can be added to optimize the properties for a given use. The resultant liquid compositions can be applied from absorbent pads, used to impregnate bandages and other dressings, or sprayed onto the affected area using pump-type or aerosol sprayers, for example.


Thickeners such as synthetic polymers, fatty acids, fatty acid salts and esters, fatty alcohols, modified celluloses or modified mineral materials can also be employed with liquid carriers to form spreadable pastes, gels, ointments, soaps, and the like, for application directly to the skin of the user.


Pharmaceutical compositions disclosed herein can be in a form suitable for rectal administration wherein the carrier is a solid. In some examples, the mixture forms unit dose suppositories. Suitable carriers include cocoa butter and other materials commonly used in the art. The suppositories can be conveniently formed by first admixing the composition with the softened or melted carriers) followed by chilling and shaping in molds.


In some examples, the pharmaceutical compositions disclosed herein can further comprise a propellant. Examples of propellants include, but are not limited to, compressed air, ethanol, nitrogen, carbon dioxide, nitrous oxide, hydrofluoroalkanes (HFA), 1,1,1,2,-tetrafluoroethane, 1,1,1,2,3,3,3-heptafluoropropane, and combinations thereof.


For administration by inhalation, the compounds or compositions can be delivered in the form of an aerosol spray from pressured container or dispenser which contains a suitable propellant or a nebulizer.


Also disclosed herein are pressurized containers comprising any of the compounds or compositions (e.g., pharmaceutical compositions) disclosed herein. Examples of containers include, but are not limited to, manual pump sprays, inhalers (e.g., meter-dosed inhalers, dry powder inhalers, etc.), and nebulizers (e.g., vibrating mesh nebulizers, jet nebulizers, ultrasonic wave nebulizers, etc.).


Systemic administration can also be by transmucosal or transdermal means. For transmucosal or transdermal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art, and include, for example, for transmucosal administration, detergents, bile salts, and fusidic acid derivatives. Transmucosal administration can be accomplished through the use of nasal sprays or suppositories. For transdermal administration, the active compounds are formulated into ointments, salves, gels, or creams as generally known in the art.


In addition to the aforementioned carrier ingredients, the pharmaceutical formulations described above can include, as appropriate, one or more additional carrier ingredients such as diluents, buffers, flavoring agents, binders, surface-active agents, thickeners, lubricants, preservatives (including anti-oxidants) and the like. Furthermore, other adjuvants can be included to render the formulation isotonic with the blood of the intended recipient. Compositions containing any of the compounds disclosed herein, and/or pharmaceutically acceptable salts thereof, can also be prepared in powder or liquid concentrate form.


Useful dosages of the compounds and agents and pharmaceutical compositions disclosed herein can be determined by comparing their in vitro activity, and in vivo activity in animal models. Methods for the extrapolation of effective dosages in mice, and other animals, to humans are known to the art.


The dosage ranges for the administration of the compositions are those large enough to produce the desired effect in which the symptoms or disorder are affected. The dosage should not be so large as to cause adverse side effects, such as unwanted cross-reactions, anaphylactic reactions, and the like. Generally, the dosage will vary with the age, condition, sex and extent of the disease in the patient and can be determined by one of skill in the art. The dosage can be adjusted by the individual physician in the event of any counterindications. Dosage can vary, and can be administered in one or more dose administrations daily, for one or several days.


Also disclosed are kits that comprise a compound disclosed herein in one or more containers. The disclosed kits can optionally include pharmaceutically acceptable carriers and/or diluents. In one embodiment, a kit includes one or more other components, adjuncts, or adjuvants as described herein. In one embodiment, a kit includes instructions or packaging materials that describe how to administer a compound or composition of the kit. Containers of the kit can be of any suitable material, e.g., glass, plastic, metal, etc., and of any suitable size, shape, or configuration. In one embodiment, a compound and/or agent disclosed herein is provided in the kit as a solid, such as a tablet, pill, or powder form. In another embodiment, a compound and/or agent disclosed herein is provided in the kit as a liquid or solution. In one embodiment, the kit comprises an ampoule or syringe containing a compound and/or agent disclosed herein in liquid or solution form.


In some examples, the kit further comprises at least one agent, wherein the compound and the agent are co-formulated.


In some examples, the compound and the agent are co-packaged.


The kits can also comprise compounds and/or products co-packaged, co-formulated, and/or co-delivered with other components. For example, a drug manufacturer, a drug reseller, a physician, a compounding shop, or a pharmacist can provide a kit comprising a disclosed compound and/or product and another component for delivery to a patient.


It is contemplated that the disclosed kits can be used in connection with the disclosed methods of making, the disclosed methods of using, and/or the disclosed compositions.


A number of embodiments of the invention have been described. Nevertheless, it will be understood that various modifications may be made without departing from the spirit and scope of the invention. Accordingly, other embodiments are within the scope of the following claims.


The examples below are intended to further illustrate certain aspects of the systems and methods described herein, and are not intended to limit the scope of the claims.


EXAMPLES

The following examples are set forth below to illustrate the methods and results according to the disclosed subject matter. These examples are not intended to be inclusive of all aspects of the subject matter disclosed herein, but rather to illustrate representative methods and results. These examples are not intended to exclude equivalents and variations of the present invention which are apparent to one skilled in the art.


Efforts have been made to ensure accuracy with respect to numbers (e.g., amounts, temperature, etc.) but some errors and deviations should be accounted for. Unless indicated otherwise, parts are parts by weight, temperature is in ° C. or is at ambient temperature, and pressure is at or near atmospheric. There are numerous variations and combinations of measurement conditions, e.g., component concentrations, temperatures, pressures and other measurement ranges and conditions that can be used to optimize the described process.


Example 1—a Systems-Level Study Reveals Host-Targeted Repurposable Drugs Against SARS-CoV-2 Infection

Abstract. Understanding the mechanism of SARS-CoV-2 infection and identifying potential therapeutics are global imperatives. Using a quantitative systems pharmacology approach, a set of repurposable and investigational drugs were identified as potential therapeutics against COVID-19. These were deduced from the gene expression signature of SARS-CoV-2-infected A549 cells screened against Connectivity Map and prioritized by network proximity analysis with respect to disease modules in the viral-host interactome. Immuno-modulating compounds aiming at suppressing hyperinflammatory responses in severe COVID-19 patients were also identified based on the transcriptome of ACE2-overexpressing A549 cells. Experiments with Vero-E6 cells infected by SARS-CoV-2, as well as independent syncytia formation assays for probing ACE2/SARS-CoV-2 spike protein-mediated cell fusion using HEK293T and Calu-3 cells, showed that several predicted compounds had inhibitory activities. Among them, salmeterol, rottlerin, and mTOR inhibitors exhibited antiviral activities in Vero-E6 cells; imipramine, linsitinib, hexylresorcinol, ezetimibe, and brompheniramine impaired viral entry. These findings provide new paths for broadening the repertoire of compounds pursued as therapeutics against COVID-19.


Introduction. Coronavirus disease-2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus (CoV) type 2 virus (SARS-CoV-2) has led to over 3 million deaths as of April 2021, and there is an urgent need to better understand the mechanisms of infection and the host cell response and to develop new therapeutics. Identification of repurposable drugs became a widespread approach for addressing current pharmacological challenges, including those faced by the current pandemic. Many compounds under clinical trials against SARS-CoV-2 are potentially repurposable drugs that target viral proteins (Esposito S et al. Infez Med. 2020, 28, 198-211; Tu Y-F et al. Int J Mot Sci. 2020, 21, 2657). While such efforts are worth pursuing, an alternative strategy is to discover host-targeted therapies. The focus herein is on the identification of repurposable compounds that modulate host cell responses, using a comprehensive, mechanism unbiased, and highly integrated systems-level approach.


The current quantitative systems pharmacology approach leverages recent progress in the field in an integrated computational/experimental framework (Stern A M et al. J Biomol Screen, 2016, 21, 521-534): One is the rigorous evaluation of the differentially expressed genes (DEGs) in SARS-CoV-2-infected cells, and the use of these DEG patterns for extracting from the Connectivity Map (CMap) database (Lamb J et al. Science, 2006, 313, 1929-1935; Subramanian A et al. Cell, 2017, 171, 1437-1452.e1417) candidate compounds/drugs that would reverse the infected cells' transcriptional program. Recent study showed, for example, the success of a CMap-based drug signature refinement approach for improving drug repositioning predictions (Iorio F et al. PLoS One, 2015, 10, e0139446). Herein, the transcriptome data from SARS-CoV-2-infected A549 (human adenocarcinomic alveolar basal epithelial) cells (Blanco-Melo D et al. bioRxiv, 2020, 10.1101/2020.03.24.004655) from lung tissue, as well as those of A549 cells overexpressing the host cell receptor angiotensin-converting enzyme 2 (ACE2) (Blanco-Melo D et al. Cell, 2020, 181, 1036-1045.e1039), were used. The latter ensures high multiplicity of infection and allows for observing the DEGs under severe infection.


Another important advance is the characterization of virus-host cell interactome for SARS-CoV-2 (Gordon D E et al. Nature, 2020, 583, 459-468) and knowledge of cell-specific protein-protein interaction (PPI) networks. These data, combined with network-based proximity analysis (Guney E et al. Nat Commun. 2016, 7, 10331), can help quantify the extent of interaction between the targets of each compound and the host cell proteins participating in the interactome with the virus. For example, Zhou et al. recently proposed 16 repurposable drugs using a network proximity analysis between drug targets in the human PPIs and host cell proteins associated with four human CoVs (SARS-CoV, MERS-CoV, HCoV-229E, and HCoV-NL63), the mouse MHV, and avian IBV, but not SARS-CoV-2 (Zhou Y et al. Cell Discov. 2020, 6, 14).


There is also access to increasingly larger databases on protein-target interactions and target-pathway mappings and interfaces, such as QuartataWeb webserver (Li H et al. Bioinformatics, 2020, 36, 3935-3937), that permit one to identify and/or predict drug-target associations and to bridge targets to cellular pathways completing chemical-target-pathway mappings.


Herein, the identification of 15 compounds is reported, including repurposable and investigational drugs, that are proposed to act against SARS-CoV-2 upon targeting the host cell machinery. In vitro assays conducted in Vero-E6 cells, HEK293T cells, and Calu-3 lung cancer cells for 10 of these prioritized compounds—six repurposable FDA-approved drugs (imipramine, salmeterol, hexylresorcinol, brompheniramine, ezetimibe, and temsirolimus) and four under development (linsitinib, torin-1, rottlerin, semaxanib)—demonstrated that several of them inhibited SARS-CoV-2 viral entry in a dose-dependent manner, with linsitinib being particularly effective. Additionally, 23 compounds are proposed for possible anti-hyperinflammatory (adjuvant) actions. These findings expand the repertoire of drugs/compounds that could be repurposed/developed for possible COVID-19 treatment.


Results


Overall workflow. FIG. 1 schematically describes the computational workflow adopted in the present study. As input, the RNA-seq data from SARS-CoV-2-infected A549 cells (Blanco-Melo D et al. bioRxiv, 2020, 10.1101/2020.03.24.004655) (referred to as Dataset 1), and those from SARS-CoV-2-infected A549 cells overexpressing ACE2 were used (shortly designated as A549-ACE2 cells) (Blanco-Melo D et al. Cell, 2020, 181, 1036-1045.e1039; tenOever B R et al. GSE147507, 2020, https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi ?acc=GSE147507; Blanco-Melo D et al. bioRxiv, 2020, 10.1101/2020.03.24.004655) (referred to as Dataset 2). The corresponding DEGs were analyzed to construct antiviral and immuno-modulating (anti-inflammatory) gene signatures respectively, which were then used to predict optimal compounds/drugs that match those signatures using CMap (FIG. 1, panels A-D). Of note, the simple signature reversal approach, as utilized in many CMap studies and a recent study of SARS-CoV-2 (Duarte R R R et al. ChemRxiv, 2020, https://doi.org/10.26434/chemrxiv. 12148764. v12148761) is not applicable here, because part of the infection-induced signature promotes viral life cycle while another part reflects antiviral responses which should be promoted rather than suppressed. To address this point, 36 DEGs were selected from Dataset 1 and 17 DEGs were selected from Dataset 2, whose actions should be either reversed or promoted by CMap-deduced drugs/compounds, depending on their role in the host proteome, as will be presented in the next subsection.


Following the identification of the compounds or repurposable drugs expected to reverse the SARS-CoV-2 pathogenic (and not the host cell immunoprotective) effects (FIG. 1, panel D), a subset was prioritized following the network proximity analysis introduced by Guney et al. (Guney E et al. Nat Commun. 2016, 7, 10331) (FIG. 1, panels E-G). To this aim, the SARS-CoV-2-host interactome (Gordon D E et al. Nature, 2020, 583, 459-468) and the lung PPI network in the BioSNAP dataset were used (Zitnik M et al. BioSNAP datasets: Stanford biomedical network dataset collection. 2018, http://snapstanfordedu/biodata) (FIG. 1, panel F). First, four disease modules—viral entry, viral replication and translation, cell signaling and regulation, and immune response modules—were identified in the viral—host interactome; and then, the “distance” of each compound from each disease module was evaluated based on the proximity of the compounds' targets to the proteins belonging to the module using the lung PPI network in BioSNAP (FIG. 1, panel G).


The compounds “closest” to each module, called the prioritized compounds, were then analyzed and clustered based on their interaction patterns with targets using QuartataWeb (Li H et al. Bioinformatics, 2020, 36, 3935-3937), to select representatives from each cluster (FIG. 1, panel H). Additional criteria, such as drug development status, side effects, mechanism of action (MOA), and antiviral activities from databases and/or literature, were considered in making the final selections from among the cluster representatives for experimental tests and possible validation (FIG. 1, panel I). More specifics on the successive steps and outputs are provided below.


Antiviral and anti-inflammatory signatures derived from post-SARS-CoV-2 infection transcriptomics. 120 DEGs composed of 100 upregulated and 20 downregulated genes were identified by DESeq2 analysis (Love M I et al. Genome Biol. 2014, 15, 550) of the transcriptome of SARS-CoV-2-infected A549 cells (Dataset 1), using false-discovery rate (FDR) default upper value of 0.05 (FIG. 2 and Table 1).



FIG. 2-FIG. 5 show the antiviral and anti-cytokine signature derived from the post-SARS-CoV-2-infection transcriptome the respective A549 and A549-ACE2.


Gene Ontology (GO) (Ashburner M et al. Nat Genet. 2000, 25, 25-29; UniProt Consortium. Nucleic Acids Res. 2019, 47, D506-D515) enrichment analysis of the 100 upregulated genes showed that they were mainly involved in viral life cycle and some in early defensive immune responses mediated by interferons (IFNs; FIG. 3). Such early responses include viral translation inhibition, RNA degradation, RNA editing, or nitric oxide synthesis (Samuel C E. Clin Microbiol Rev. 2001, 14, 778-809). Nevertheless, the induction of interferon types I and III was relatively more “muted” in SARS-CoV-2-infected A549 cells compared to those of other respiratory viruses such as influenza A and respiratory syncytial virus (Blanco-Melo D et al. Cell, 2020, 181, 1036-1045.e1039).


As to downregulated genes, they mainly comprised vesicle-related structures or endosomal events, including autophagosome formation for autophagic elimination of the virus (Kudchodkar S B et al. Rev Med Virol. 2009, 19, 359-378). Promoting autophagy showed potential in reducing MERS infection (Gassen N C et al. Nat Commun. 2019, 10, 5770) and thus down-regulation of this process might contribute to viral escape. In CMap applications to diabetes (Zhang M et al. PLoS One, 2015, 10, e0126082) and obesity (Liu J et al. Cell, 2015, 161, 999-1011), compounds that reverse the gene signature induced by the disease were selected. However, in SARS-CoV-2 infection, it is important to promote the adaptive immune response mediated by IFNs at early stage rather than blindly reversing the complete gene signature. Therefore, after overrepresentation analysis, and evaluation of the GO annotations associated with these genes as described in the Materials and Methods, 36 genes were selected to be upregulated (FIG. 4). These genes include (i) 26 genes upregulated in SARS-CoV-2 infected A549 cells, which are associated with viral defense and should be upregulated for antiviral activity, and (ii) 10 genes downregulated in A549 cells, associated with endocytic or vesicular processes, which should be reverted. Table 2 lists the corresponding gene products/proteins (left two columns). Table 3 provides information on their GO biological processes.


The A549-ACE2 cells (Dataset 2) repeatedly exhibited a more pronounced cytokine upregulation, along with IFN response insufficiency, compared to A549 cells. Based on this observation, immune-modulating therapies have been suggested (Blanco-Melo D et al. Cell, 2020, 181, 1036-1045.e1039). The most strongly upregulated 17 genes were selected (log2 fold change of 3.5 or higher; see Materials and Methods), toward identifying compounds that would suppress the excessive inflammatory cytokine response in severe COVID-19 patients. This led to the anti-inflammatory (or anti-cytokine) signature shown in FIG. 5 composed of 17 genes to be downregulated (Table 2 right two columns). Table 4 list the corresponding proteins and their GO annotations.









TABLE 1







120 differentially expressed genes (DEGs) in


SARS-CoV-2-infected A549 cells.












log2





fold



Gene name
Protein name
change
Padjusted










Upregulated 100 genes in SARS-CoV-2-infected A549 cells










MX1
IFN-induced
5.20
1.16E−93



GTP-binding protein Mx1
4.59
6.80E−04


IFI44
IFN-induced protein 44 (Fragment)




IFIT1
IFN-induced protein with
4.43
6.18E−96



tetratricopeptide repeats 1





(IFIT-1) (IFN-induced 56 kDa





protein) (IFI-56K) (P56)




IFI6
IFNα-inducible protein 6
4.27
1.01E−145


OAS2
2′-5′-oligoadenylate synthase 2
4.25
1.49E−06


IFITM1
IFN induced transmembrane
4.22
3.12E−05



protein 1 (9-27), isoform





CRA_a




ISG15
Ubiquitin-like protein ISG15
3.80
6.21E−85



(Fragment)




IFI27
IFNα-inducible protein 27,
3.54
8.09E−14



mitochondrial (Fragment)




IRF7
IFN regulatory factor 7,
3.13
5.69E−43



isoform CRA_a




PTPRE
Receptor-type tyrosine-protein
3.02
3.36E−10



phosphatase epsilon





(Protein-tyrosine phosphatase





epsilon) (R-PTP-epsilon)





(EC 3.1.3.48)




OASL
2′-5′-oligoadenylate
2.53
8.17E−07



synthase-like protein (Fragment)




DDX60
ATP-dependent RNA helicase DDX60
2.42
1.43E−25


CMPK2
Mitochondrial cytidine
2.23
7.66E−03



monophosphate (UMP-CMP)





kinase 2 (Fragment)




PARP9
Protein mono-ADP-ribosyltransferase
2.13
4.32E−43



PARP9 (Fragment)




IRF9
IFN regulatory factor 9
2.11
6.73E−42


IFIT3
IFN-induced protein with
2.07
1.88E−21



tetratricopeptide repeats 3





(IFIT-3) (CIG49) (ISG-60)





(IFN-induced 60 kDa protein)





(IFI-60K) (Retinoic acid-induced





gene G protein) (P60)





(RIG-G)




SAMD9L
Sterile α motif domain-
1.97
5.51E−06



containing protein 9-like





(Fragment)




DDX58
Antiviral innate immune
1.88
3.16E−19



response receptor RIG-I




IFIH1
IFN-induced helicase C
1.88
4.01E−15



domain-containing protein 1




PARP10
Poly [ADP-ribose] polymerase
1.83
9.35E−07



(PARP) (EC 2.4.2 .-)




SAMD9
Sterile a motif domain-
1.79
1.76E−07



containing protein 9 (Fragment)




TRIM34
Tripartite motif-containing
1.71
2.07E−04



protein 34 (IFN-responsive





finger protein 1) (RING





finger protein 21)




HERC6
Probable E3 ubiquitin-protein
1.52
9.14E−14



ligase HERC6




REC8
Meiotic recombination protein
1.51
1.81E−04



REC8 homolog (Fragment)




OAS1
2′-5′ oligoadenylate synthetase
1.51
2.69E−33



1 p49 isoform (Fragment)




DTX3L
E3 ubiquitin-protein ligase
1.46
1.44E−21



DTX3L (EC 2.3.2.27) (B-





lymphoma-and BAL-associated





protein) (Protein deltex-3-





like) (RING-type E3





ubiquitin transferase DTX3L;





Rhysin-2)




HELZ2
Helicase with zinc finger
1.38
3.78E−17



domain 2 (ATP-dependent





helicase PRIC285)





(transcriptional coactivator) (PPAR-α-





interacting complex protein





285) (PPAR-γ DNA-binding





domain-interacting protein 1)





(PDIP1) (Peroxisomal





proliferator-activated receptor





A-interacting complex 285





kDa protein




EIF2AK2
eIF2AK2 protein
1.36
3.59E−12


STAT1
Signal transducer and activator
1.35
1.90E−28



of transcription 1





(Fragment)




OAS3
2′-5′-oligoadenylate
1.35
3.29E−26



synthetase 3, 100 kDa, isoform





CRA_a




IFI16
Γ-IFN-inducible protein 16
1.35
4.30E−03


PLSCR1
Phospholipid scramblase (Fragment)
1.27
4.77E−13


IFIT2
IFN-induced protein with
1.25
1.68E−02



tetratricopeptide repeats 2




SP110
Sp110 nuclear body
1.23
6.32E−08



protein (Fragment)




CCL20
C-C motif chemokine 20 (Fragment)
1.20
3.03E−06


FGG
Fibrinogen y chain
1.19
5.53E−03


DDX60L
Putative ATP-dependent
1.13
1.68E−05



RNA helicase DDX60 (EC





3.6.4.13)




CFB
Complement factor B (Fragment)
1.12
7.26E−08


TRIM14
Tripartite motif-containing
1.09
3.97E−11



14, isoform CRA_c




IFIT5
IFN-induced protein with
1.09
3.90E−07



tetratricopeptide repeats 5





(IFIT-5) (Retinoic acid-





and IFN-inducible 58 kDa





protein) (P58)




SAMHD1
Deoxynucleoside triphosphate
1.08
5.81E−12



triphosphohydrolase





SAMHD1




PHF11
PHD finger protein 11 (cDNA
1.07
2.63E−03



FLJ56933, highly similar to





Homo sapiens PHD finger





protein 11 (PHF11), transcript





variant 1, mRNA)




IFI35
IFN-induced 35 kDa protein
1.06
1.54E−08



(IFP 35) (Ifi-35)




LAP3
Cytosol aminopeptidase (Fragment)
1.03
2.82E−13


CXCL5
C-X-C motif chemokine 5
0.96
3.86E−08



(ENA-78(1-78)) (Epithelial-





derived neutrophil-activating





protein 78) (Small-inducible





cytokine B5) [Cleaved into:





ENA-78(8-78); ENA-78(9-78)]




PARP14
Protein mono-ADP-
0.96
4.02E−08



ribosyltransferase PARP14




HERC5
E3 ISG15—protein ligase
0.94
3.70E−05



HERC5 (HECT and RLD





domain- containing E3





ubiquitin protein ligase 5)





(Fragment)




SP100
Nuclear autoantigen Sp-100
0.94
1.23E−07



(Fragment)




BCL2A1
Bcl-2-related protein A1
0.93
1.15E−02



(Bcl-2-like protein 5) (Bcl2-L-5)





(Hemopoietic-specific early





response protein) (Protein





BFL-1) (Protein GRS)




IFITM3
IFN-induced transmembrane protein 3
0.91
1.75E−03


GBP3
Guanylate-binding protein 3
0.89
1.26E−02


USP18
Ubl carboxyl-terminal
0.86
1.21E−04



hydrolase 18 (EC 3.4.19.-) (43 kDa





ISG15-specific protease)





(hUBP43) (ISG15-specific-





processing protease)





(Ubl thioesterase 18)




CP
CP protein (Ceruloplasmin
0.86
8.59E−04



(Ferroxidase)) (Ceruloplasmin





(Ferroxidase), isoform CRA_a)




CFH
Complement factor H
0.85
1.15E−02


PARP12
Poly (ADP-ribose) polymerase
0.82
9.48E−05



family, member 12,





isoform CRA_b (Zinc





finger CCCH type domain





containing 1)




STEAP1
Six transmembrane epithelial
0.81
9.43E−03



antigen of the prostate 1,





isoform CRA_a




EHF
ETS homologous factor (Fragment)
0.81
1.60E−02


PTGS2
Prostaglandin-endoperoxidase
0.80
1.47E−03



synthase 2 (EC 1.14.99.1)





(Fragment)




C1R
Complement C1r subcomponent
0.80
4.43E−07


SAT1
Diamine acetyltransferase 1
0.78
1.89E−06


BIVM-
BIVM-ERCC5 readthrough
0.76
3.05E−02


ERCC5
(Fragment)




C19orf66
Shiftless antiviral inhibitor
0.76
1.03E−03



of ribosomal frameshifting





protein (SFL) (SHFL)





(IFN-regulated antiviral protein)





(IRAV) (Repressor of yield





of DENV protein) (RyDEN)




SNAP25
Synaptosomal-associated
0.76
2.62E−03



protein 25 (Fragment)




CXCL8
Multifunctional fusion
0.75
1.17E−06



protein [Includes: Interleukin-8





(IL-8) (C-X-C motif chemokine 8)




PDK4
Protein-serine/threonine
0.75
1.29E−02



kinase (EC 2.7.11.-)




PNPT1
Polyribonucleotide
0.74
9.53E−04



nucleotidyltransferase 1, mitochondrial




MMD
Monocyte to macrophage
0.72
2.08E−02



differentiation factor




APOL6
Apolipoprotein L, 6
0.71
3.42E−02



(Apolipoprotein L6) (cDNA





FLJ38562 fis, clone





HCHON2004002, similar to





Apolipoprotein-L6)




C1S
Complement C1s subcomponent
0.71
1.43E−06


CXCL2
C-X-C motif chemokine
0.71
2.37E−04


UBE2L6
Ubiquitin/ISG15-conjugating
0.71
3.07E−04



enzyme E2 L6 (Fragment)




NUCB2
Nesfatin 1 (Fragment)
0.70
3.56E−02


APOL1
Apolipoprotein L, 1, isoform CRA_b
0.70
5.96E−03


PLA2G4A
Cytosolic phospholipase
0.70
1.93E−02



A2 (cPLA2) (Phospholipase A2





group IVA)




PAPPA-
Protein PAPPAS
0.69
2.25E−02


AS1
(DIPLA1 antisense RNA 1)




TYMP
Thymidine phosphorylase isoform 2
0.66
1.33E−02


FGA
Fibrinogen α chain
0.66
9.95E−04


PTPN12
Tyrosine-protein phosphatase
0.66
1.56E−02



non-receptor type 12




FILIP1
Filamin A interacting protein
0.66
2.08E−02



1, isoform CRA_c (Filamin-





A-interacting protein 1)




ESF1
ESF1 homolog
0.64
3.36E−02


NCOA7
Nuclear receptor coactivator 7,
0.63
2.58E−03



isoform CRA_c




CXCL3
C-X-C motif chemokine 3
0.62
1.61E−02



(GRO-γ(1-73)) (Growth-





regulated protein γ) (GRO-γ)





(Macrophage inflammatory





protein 2-β) (MIP2-β)





[Cleaved into: GRO-γ(5-73)]




B2M
B-2-microglobulin
0.61
1.17E−02


TDRD7
Tudor domain-containing
0.61
3.92E−03



protein 7 (PCTAIRE2-binding





protein) (Tudor repeat





associator with PCTAIRE-2) (Trap)




C3
Complement C3 (Fragment)
0.61
1.47E−03


PSIP1
Alternative protein PSIP1
0.60
5.96E−03


KTN1
Kinectin 1 (Kinesin receptor),
0.59
3.36E−02



isoform CRA_a (Kinectin 1





(Kinesin receptor), isoform CRA_b)




CXCL1
Growth-regulated a protein
0.59
2.00E−03



(C-X-C motif chemokine 1)





(GRO-α(1-73)) (Melanoma





growth stimulatory activity)





(MGSA) (Neutrophil-





activating protein 3) (NAP-3)





[Cleaved into: GRO-α(4-73);





GRO-α(5-73); GRO-α(6-





73)]




TCIM
Transcriptional and immune
0.58
5.53E−03



response regulator (Thyroid





cancer protein 1) (TC-1)




ADAR
Double-stranded RNA-specific
0.57
1.92E−05



adenosine deaminase





(fragment)




DKK1
Dickkopf-like protein 1
0.57
1.24E−03


KYNU
Kynureninase (Fragment)
0.57
2.98E−02


AREG
Amphiregulin
0.57
2.09E−03


LGALS3BP
Lectin galactoside-binding
0.55
1.25E−02



soluble 3 binding protein





isoform 1 (CRA_d) (Fragment)




CDH1
Cadherin-1
0.55
2.88E−03


STAT2
Signal transducer and activator
0.55
7.66E−03



of transcription 2




HIF1A
Hypoxia-inducible factor 1,
0.54
3.82E−02



a subunit (Basic helix-loop-





helix transcription factor),





isoform CRA_a




TRIM25
E3 ubiquitin/ISG15 ligase TRIM25
0.53
7.01E−04


HSP90B1
Endoplasmin
0.48
3.44E−02


EDN1
Endothelin-1 (Preproendothelin-1)
0.45
2.50E−02



(PPET1) [Cleaved into:





Endothelin-1 (ET-1); Big endothelin-1]









Downregulated 20 genes in SARS-CoV-2-infected A549 cells










SQSTM1
Sequestosome-1
−0.46
2.50E−02


NPTX1
Neuronal pentraxin-1 (NP1)
−0.51
2.25E−02



(Neuronal pentraxin I) (NP-I)




AHNAK2
Protein AHNAK2
−0.51
3.19E−03


NEU1
Sialidase-1
−0.52
1.24E−02


WDR81
WD repeat-containing protein 81
−0.53
2.50E−02


FAM102A
Protein FAM102A
−0.57
2.34E−03



(Early estrogen-induced gene 1





protein)




DANCR
Uncharacterized protein
−0.59
1.84E−02



DANCR (Anti-differentiation





ncRNA protein) (Small





nucleolar RNA host gene protein





13)




NT5DC2
5′-nucleotidase domain-
−0.59
1.23E−02



containing protein 2 (Fragment)




MAP3K14
Mitogen-activated protein
−0.59
1.44E−02



kinase kinase kinase 14





(Fragment)




KLHL21
Kelch-like protein 21
−0.61
5.58E−03


LOC284454
/(ncRNA)
−0.64
3.92E−02


SYNE1
Nesprin-1
−0.68
2.34E−03


COL1A1
Collagen, type I, α 1, isoform CRA_a
−0.73
1.05E−03


OSGIN1
Oxidative stress-induced
−0.74
6.80E−04



growth inhibitor 1 (Fragment)




RAP1GAP
Rap1 GTPase-activating protein 1
−0.74
6.04E−05


NPIPB5
Nuclear pore complex-
−0.83
1.06E−02



interacting protein family member





B5




UAP1L1
UDP-N-acetylhexosamine
−0.84
4.55E−04



pyrophosphorylase-like protein





1




NUPR1
Nuclear protein 1
−0.99
5.96E−03


NECAB2
N-terminal EF-hand calcium-
−1.12
2.50E−02



binding protein 2 (Fragment)




KRT4
Keratin, type II cytoskeletal 4
−1.23
1.84E−04
















TABLE 2







Antiviral and anti-inflammatory signature genes derived from


SARS-CoV-2-infected cells.








Antiviral signature
Anti-inflammatory signature


(based on A549 Cells)
(A549-ACE2 cells)










Genea
Proteinb
Genec
Proteinb











To-be-upregulated
To-be-downregulated










IFI6
IFNα-inducible protein 6
EGR1
Early growth





response





protein


IRF7
IFN regulatory factor
IFNB1
Interferon beta



7, isoform CRA_a




DDX60
ATP-dependent RNA
CXCL2
C-X-C motif



helicase DDX60

chemokine


PARP9
Protein mono-ADP-
NFKBIA
NFκB inhibitor α



ribosyltransferase





PARP9




IRF9
IFN regulatory factor 9
SELE
E-selectin


IFIT3
IFN-induced protein
IL8
Interleukin-8



with tetratricopeptide





repeats 3 (Retinoic





acid-induced gene G





protein) (RIG-G)




DDX58
Antiviral innate immune
IRF7
IFN regulatory



response receptor

factor 7



RIG-I




IFIH1
IFN-induced helicase
IFITM1
IFN-induced



C domain-containing

transmembrane



protein 1

protein 1


TRIM34
Tripartite motif-containing
NFIL3
Nuclear factor



protein 34 (IFN-

interleukin-



responsive finger

3-regulated



protein 1) (RING finger

prot



protein 21)




DTX3L
E3 ubiquitin-protein
TNF
Tumor necrosis



ligase DTX3L (EC

factor



2.3.2.27)

α


STAT1
Signal transducer
CXCL3
C-X-C motif



and activator of

chemokine 3



transcription 1




IFIT2
IFN-induced protein
SOCS1
Suppressor



with tetratricopeptide

of cytokine



repeats 2

signaling 1


CCL20
C-C motif chemokine
CD274
Programmed



20 (Fragment)

cell death





1 ligand 1


TRIM14
Tripartite motif-
IL20RB
Interleukin-20



containing 14, isoform

receptor



CRA_c

subunit β


SAMHD1
Deoxynucleoside
CCL20
C-C motif



triphosphate

chemokine



triphosphohydrolase

20


IFI35
IFN-induced 35 kDa protein
CCR6
C-C chemokine



(IFP 35) (Ifi-35)

receptor type 6


PARP14
Protein mono-ADP-
PARP14
HLA-F Human



ribosyltransferase

leukocyte





antigen F


HERC5
E3 ISG15—protein





ligase HERC5





(Fragment)




SP100
Nuclear autoantigen





Sp-100 (Fragment)




GBP3
Guanylate-binding protein 3




USP18
Ubl carboxyl-terminal





hydrolase 18




B2M
β2-microglobulin




KYNU
Kynureninase (Fragment)




STAT2
Signal transducer





and activator of





transcription 2




TRIM25
E3 ubiquitin/ISG15





ligase TRIM25




EDN1
Endothelin-1





(Preproendothelin-1) (PPET1)











Gened
Proteinb










To-be-upregulated








SQSTM1
Sequestosome-1


AHNAK2
Protein AHNAK2


NPTX1
Neuronal pentraxin-1



(NP1)


NEU1
Sialidase-1


WDR81
WD repeat-containing



protein 81


KLHL21
Kelch-like protein 21


SYNE1
Nesprin-1


COL1A1
Collagen, type I, α1,



isoform CRA_a


RAP1GAP
Rap1 GTPase-



activating protein 1


KRT4
Keratin, type II



cytoskeletal 4





ªGenes observed to be upregulated in the transcriptome of A549 cells (Dataset 1).



bProtein: gene product from UniProt Consortium (UniProt Consortium. Nucleic Acids Res. 2019, 47, D506-D515).




cGenes observed to be upregulated in the transcriptome of ACE2-overexpressing A549 cells (Dataset 2); All genes are ordered by log2 fold change in descending order. See Table 3 and Table 4 for the log2 fold change values and associated GO biological processes or cellular components. See also FIG. 4 and FIG. 5 for the respective log2 fold change profiles observed in SARS-CoV-2-infected-A549 and SARS-CoV-2-infected-A549-ACE2 cells.




dGenes observed to be downregulated in the transcriptome of A549 cells (Dataset 1).














TABLE 3







Properties of the 36 DEGs that define the antiviral gene signature


derived from the transciptome of SARS-CoV-2-infected A549 cells.














Gene

log2 fold

GO



Index
name
Protein name
Change
Database
Annotation
GO number










Upregulated Genes That Should Be Upregulated By Small Molecules













1
IFI6
IFNα-inducible
4.27
GO:BP
type I IFN
GO:0060337




protein 6


signaling pathway







defense response
GO:0051607







to virus







cellular response
GO:0071357







to type I IFN







response to type I IFN
GO:0034340


2
IRF7
IFN regulatory
3.13
GO:BP
cellular response
GO:0071346




factor 7, isoform


to IFN-γ




CRA_a


type I IFN
GO:0060337







signaling pathway







cellular response
GO:0071357







to type I IFN







response to type I IFN
GO:0034340







response to IFN-γ
GO:0034341







defense response
GO:0051607







to virus







regulation of response
GO:0060759







to cytokine stimulus







IFN-γ-mediated
GO:0060333







signaling pathway


3
DDX60
ATP-dependent
2.42
GO:BP
defense response
GO:0051607




RNA helicase DDX60


to virus


4
PARP9
Protein mono-ADP-
2.13
GO:BP
regulation of response
GO:0060759




ribosyltransferase


to cytokine stimulus




PARP9 (Fragment)


cellular response
GO:0071346







to IFN-γ







IFN-γ-mediated
GO:0060333







signaling pathway







response to IFN-γ
GO:0034341







defense response
GO:0051607







to virus


5
IRF9
IFN regulatory
2.11
GO:BP
response to type I IFN
GO:0034340




factor 9


cellular response
GO:0071346







to IFN-γ







response to IFN-γ
GO:0034341







IFN-γ-mediated
GO:0060333







signaling pathway







cellular response
GO:0071357







to type I IFN







type I IFN
GO:0060337







signaling pathway







defense response
GO:0051607







to virus


6
IFIT3
IFN-induced
2.07
GO:BP
cellular response
GO:0071357




protein with


to type I IFN




tetratricopeptide


defense response
GO:0051607




repeats 3


to virus







type I IFN
GO:0060337







signaling pathway







response to type I IFN
GO:0034340


7
DDX58
Antiviral innate
1.88
GO:BP
defense response
GO:0051607




immune response


to virus




receptor RIG-I


regulation of response
GO:0060759







to cytokine stimulus


8
IFIH1
IFN-induced
1.88
GO:BP
defense response
GO:0051607




helicase C domain-


to virus




containing protein 1


regulation of response
GO:0060759







to cytokine stimulus


9
TRIM34
Tripartite motif-
1.71
GO:BP
defense response
GO:0051607




containing protein


to virus




34 (IFN-responsive


response to IFN-γ
GO:0034341




finger protein 1)


IFN-γ-mediated
GO:0060333




(RING finger


signaling pathway




protein 21)


cellular response
GO:0071346







to IFN-γ


10
DTX3L
E3 ubiquitin-protein
1.46
GO:BP
defense response
GO:0051607




ligase DTX3L


to virus


11
STAT1
Signal transducer
1.35
GO:BP
regulation of response
GO:0060759




and activator of


to cytokine stimulus




transcription 1


type I IFN
GO:0060337




(Fragment)


signaling pathway







defense response
GO:0051607







to virus







cellular response
GO:0071357







to type I IFN







negative regulation
GO:0048525







of viral process







response to type I IFN
GO:0034340







IFN-γ-mediated
GO:0060333







signaling pathway







response to IFN-γ
GO:0034341







regulation of symbiosis,
GO:0043903







encompassing mutualism







through parasitism







negative regulation of
GO:0043901







multi-organism process







cellular response
GO:0071346







to IFN-γ







regulation of
GO:0050792







viral process


12
IFIT2
IFN-induced
1.25
GO:BP
cellular response
GO:0071357




protein with


to type I IFN




tetratricopeptide


defense response
GO:0051607




repeats 2


to virus







type I IFN
GO:0060337







signaling pathway







response to type I IFN
GO:0034340


13
CCL20
C-C motif
1.2
GO:BP
response to IFN-γ
GO:0034341




chemokine 20


cellular response
GO:0071346




(Fragment)


to IFN-γ


14
TRIM14
Tripartite motif-
1.09
GO:BP
regulation of
GO:0050792




containing 14,


viral process




isoform CRA_c


regulation of symbiosis,
GO:0043903







encompassing mutualism







through parasitism







negative regulation of
GO:0043901







multi-organism process







negative regulation
GO:0048525







of viral process


15
SAMHD1
Deoxynucleoside
1.08
GO:BP
cellular response
GO:0071357




triphosphate


to type I IFN




triphosphohydrolase


negative regulation of
GO:0043901




SAMHD1


multi-organism process







regulation of response
GO:0060759







to cytokine stimulus







response to type I IFN
GO:0034340







defense response
GO:0051607







to virus







type I IFN
GO:0060337







signaling pathway


16
IFI35
IFN-induced 35
1.06
GO:BP
type I IFN
GO:0060337




kDa protein (IFP


signaling pathway




35) (Ifi- 35)


cellular response
GO:0071357







to type I IFN







response to type I IFN
GO:0034340


17
PARP14
Protein mono-ADP-
0.96
GO:BP
regulation of response
GO:0060759




ribosyltransferase


to cytokine stimulus




PARP14


IFN-γ-mediated
GO:0060333







signaling pathway







cellular response
GO:0071346







to IFN-γ







response to IFN-γ
GO:0034341







negative regulation of
GO:0043901







multi-organism process


18
HERC5
E3 ISG15-protein
0.94
GO:BP
defense response
GO:0051607




ligase HERC5


to virus




(Fragment)


19
SP100
Nuclear autoantigen
0.94
GO:BP
cellular response
GO:0071357




Sp-100 (Fragment)


to type I IFN







type I IFN
GO:0060337







signaling pathway







response to type I IFN
GO:0034340







response to IFN-γ
GO:0034341







IFN-γ-mediated
GO:0060333







signaling pathway







cellular response
GO:0071346







to IFN-γ


20
GBP3
Guanylate-binding
0.89
GO:BP
response to IFN-γ
GO:0034341




protein 3


cellular response
GO:0071346







to IFN-γ







defense response
GO:0051607







to virus


21
USP18
Ubl carboxyl-
0.86
GO:BP
type I IFN
GO:0060337




terminal hydrolase


signaling pathway




18


cellular response
GO:0071357







to type I IFN







response to type I IFN
GO:0034340







regulation of response
GO:0060759







to cytokine stimulus


22
B2M
B-2-microglobulin
0.61
GO:BP
response to IFN-γ
GO:0034341







cellular response
GO:0071346







to IFN-γ







IFN-γ-mediated
GO:0060333







signaling pathway


23
KYNU
Kynureninase
0.57
GO:BP
response to IFN-γ
GO:0034341




(Fragment)


24
STAT2
Signal transducer
0.55
GO:BP
defense response
GO:0051607




and activator of


to virus




transcription 2


response to type I IFN
GO:0034340







cellular response
GO:0071357







to type I IFN







type I IFN
GO:0060337







signaling pathway


25
TRIM25
E3 ubiquitin/ISG15
0.53
GO:BP
cellular response
GO:0071346




ligase TRIM25


to IFN-γ







regulation of
GO:1903900







viral life cycle







response to IFN-γ
GO:0034341







negative regulation
GO:1903901







of viral life cycle







IFN-γ-mediated
GO:0060333







signaling pathway







regulation of
GO:0050792







viral process







negative regulation of
GO:0043901







multi-organism process







defense response
GO:0051607







to virus







negative regulation
GO:0048525







of viral process







regulation of symbiosis,
GO:0043903







encompassing mutualism







through parasitism


26
EDN1
Endothelin-1
0.45
GO:BP
regulation of response
GO:0060759




(Preproendothelin-


to cytokine stimulus




1) (PPET1)


response to IFN-γ
GO:0034341







cellular response
GO:0071346







to IFN-γ







Down regulated Genes That Should Be Upregulated By Small Molecules













1
SQSTM1
Sequestosome-1
−0.46
GO:CC
supramolecular complex
GO:0099080







intracellular vesicle
GO:0097708







supramolecular fiber
GO:0099512







supramolecular polymer
GO:0099081







cytoplasmic vesicle
GO:0031410







myofibril
GO:0030016







sarcomere
GO:0030017







contractile fiber
GO:0043292







amphisome
GO:0044753







P-body
GO:0000932







autophagosome
GO:0005776







vesicle
GO:0031982


2
AHNAK2
Protein AHNAK2
−0.51
GO:CC
supramolecular complex
GO:0099080







intracellular vesicle
GO:0097708







cytoplasmic vesicle
GO:0031410







sarcomere
GO:0030017







supramolecular polymer
GO:0099081







supramolecular fiber
GO:0099512







myofibril
GO:0030016







contractile fiber
GO:0043292







vesicle
GO:0031982


3
NPTX1
Neuronal pentraxin-
−0.51
GO:CC
cytoplasmic vesicle
GO:0031410




1 (NP1) (Neuronal


intracellular vesicle
GO:0097708




pentraxin I) (NP-I)


vesicle
GO:0031982


4
NEU1
Sialidase-1
−0.52
GO:CC
cytoplasmic vesicle
GO:0031410







intracellular vesicle
GO:0097708







vesicle
GO:0031982


5
WDR81
WD repeat-
−0.53
GO:CC
cytoplasmic vesicle
GO:0031410




containing protein


vesicle
GO:0031982




81


autophagosome
GO:0005776







intracellular vesicle
GO:0097708


6
KLHL21
Kelch-like protein
−0.61
GO:CC
supramolecular polymer
GO:0099081




21


cytoplasmic vesicle
GO:0031410







intracellular vesicle
GO:0097708







supramolecular complex
GO:0099080







vesicle
GO:0031982







supramolecular fiber
GO:0099512


7
SYNE1
Nesprin-1
−0.68
GO:CC
P-body
GO:0000932







myofibril
GO:0030016







contractile fiber
GO:0043292







supramolecular polymer
GO:0099081







sarcomere
GO:0030017







supramolecular complex
GO:0099080







supramolecular fiber
GO:0099512


8
COL1A1
Collagen, type I, α
−0.73
GO:CC
vesicle
GO:0031982




1, isoform CRA_a


supramolecular fiber
GO:0099512







supramolecular polymer
GO:0099081







cytoplasmic vesicle
GO:0031410







intracellular vesicle
GO:0097708







supramolecular complex
GO:0099080







collagen type I trimer
GO:0005584


9
RAP1GAP
Rap1 GTPase-
−0.74
GO:CC
cytoplasmic vesicle
GO:0031410




activating protein 1


vesicle
GO:0031982







intracellular vesicle
GO:0097708


10
KRT4
Keratin, type II
−1.23
GO:CC
supramolecular fiber
GO:0099512




cytoskeletal 4


supramolecular polymer
GO:0099081







supramolecular complex
GO:0099080





* Showing enrichment result from Gene Ontology Biological Process (GO:BP) and Cellular Component (GO:CC) databases with term size <300 genes and overlap size >10 genes.













TABLE 4







Properties of the 17 DEGs that define the anti-cytokine signature derived


from the transcriptome of SARS-CoV-2-infected A549-ACE2 cells.















Gene

log2 Fold


GO



Index
name
Protein name
Change
Padjusted
Database
Annotation
GO number

















1
EGR1
Early growth
6.62
0.00E+00
GO:BP
cytokine-mediated
GO:0019221




response protein



signaling pathway








cellular response to
GO:0071345








cytokine stimulus


2
IFNB1
Interferon β
5.76
1.15E−28
GO:BP
cytokine-mediated
GO:0019221








signaling pathway








cellular response to
GO:0071345








cytokine stimulus








regulation of response
GO:0032101








to external stimulus








positive regulation of
GO:0002684








immune system process


3
CXCL2
C-X-C motif
5.27
0.00E+00
GO:BP
cytokine-mediated
GO:0019221




chemokine



signaling pathway








cellular response to
GO:0071345








cytokine stimulus


4
NFKBIA
NFκB
5.23
0.00E+00
GO:BP
cytokine-mediated
GO:0019221




inhibitorα



signaling pathway








cellular response to
GO:0071345








cytokine stimulus








regulation of response
GO:0032101








to external stimulus


5
SELE
E-selectin
4.85
1.59E−25
GO:BP
regulation of response
GO:0032101








to external stimulus








positive regulation of
GO:0002684








immune system process


6
IL8
Multifunctional
4.85
0.00E+00
GO:BP
cytokine-mediated
GO:0019221




fusion protein



signaling pathway




[Includes:



cellular response to
GO:0071345




Interleukin-8



cytokine stimulus








regulation of response
GO:0032101








to external stimulus








positive regulation of
GO:0002684








immune system process


7
IRF7
Interferon
4.81
2.69E−97
GO:BP
cytokine-mediated
GO:0019221




regulatory



signaling pathway




factor 7



cellular response to
GO:0071345








cytokine stimulus








regulation of response
GO:0032101








to external stimulus








positive regulation of
GO:0002684








immune system process


8
IFITM1
Interferon
4.58
3.21E−47
GO:BP
cytokine-mediated
GO:0019221




induced



signaling pathway




transmembrane



cellular response to
GO:0071345




protein 1



cytokine stimulus


9
NFIL3
Nuclear factor
4.22
0.00E+00
GO:BP
cellular response to
GO:0071345




interleukin-3-



cytokine stimulus




regulated protein


10
TNF
Tumor necrosis
4.16
3.70E−29
GO:BP
cytokine-mediated
GO:0019221




factor



signaling pathway








cellular response to
GO:0071345








cytokine stimulus








regulation of response
GO:0032101








to external stimulus








positive regulation of
GO:0002684








immune system process


11
CXCL3
C-X-C motif
4.03
0.00E+00
GO:BP
cytokine-mediated
GO:0019221




chemokine 3



signaling pathway








cellular response to
GO:0071345








cytokine stimulus


12
SOCS1
Suppressor of
4.02
1.48E−16
GO:BP
cytokine-mediated
GO:0019221




cytokine



signaling pathway




signaling 1



cellular response to
GO:0071345








cytokine stimulus








regulation of response
GO:0032101








to external stimulus








positive regulation of
GO:0002684








immune system process


13
CD274
Programmed cell
4.01
1.20E−46
GO:BP
positive regulation of
GO:0002684




death 1 ligand 1



immune system process


14
IL20RB
Interleukin-20
3.79
1.15E−34
GO:BP
cytokine-mediated
GO:0019221




receptor



signaling pathway




subunit β



cellular response to
GO:0071345








cytokine stimulus








regulation of response
GO:0032101








to external stimulus


15
CCL20
C-C motif
3.77
0.00E+00
GO:BP
cytokine-mediated
GO:0019221




chemokine 20



signaling pathway








cellular response to
GO:0071345








cytokine stimulus








positive regulation of
GO:0002684








immune system process


16
CCR6
C-C chemokine
3.66
1.60E−16
GO:BP
cytokine-mediated
GO:0019221




receptor type 6



signaling pathway








cellular response to
GO:0071345








cytokine stimulus








regulation of response
GO:0032101








to external stimulus








positive regulation of
GO:0002684








immune system process


17
HLA-F
HLA-F
3.49
3.59E−86
GO:BP
cytokine-mediated
GO:0019221








signaling pathway








cellular response to
GO:0071345








cytokine stimulus








regulation of response
GO:0032101








to external stimulus








positive regulation of
GO:0002684








immune system process





* Gene name: gene symbol of the DEGs. Protein name: protein name of corresponding genes from UniProt (UniProt Consortium. Nucleic Acids Res. 2019, 47, D506-D515) (Genes are ordered by log2 fold change in descending order. see STAR*Methods for the definition of adjusted P. Showing enrichment results from GO Biological Process database with term size < 1,200 genes and overlap size >10 genes. Log2 Fold Change: log2 transformation of the fold change in gene expression level after viral infection; Database: Gene ontology (GO) database used. BP refers to biological process, CC refers to cellular component; Term: name of the gene set in the GO database; GO number: the index of the GO term.


* Genes are ordered by descending log2 Fold Change.






Identification of antiviral and anti-cytokine compounds and corresponding targets. The compounds that best matched the antiviral and anti-cytokine signatures determined above were identified by screening each signature against the CMap database. Briefly, the Touchstone collection of perturbagen signatures from 3,000 compounds on six cell lines was searched to assign a Cmap connectivity score to each compound. The score is based on the similarity between the compound-induced gene signature in Cmap and the query/input signature, repeated separately for the antiviral and anti-cytokine signatures. This led to a set of 263 potentially antiviral compounds, and another of 275 potentially anti-cytokine compounds, using default thresholds in Cmap (see Materials and Methods), listed in Table 5 and Table 6, respectively. The compounds included twelve (chlorpromazine, apicidin, ribavirin, mycophenolate, entacapone, equilin, metformin, mercaptopurine, gemcitabine, mepacrine/quinacrine, daunorubicin, and valproic acid) listed in the COVID-19 drug repurposing database compiled by Excelra (Excelra, 2020 COVID-19 drug repurposing database. https://wwwexcelracom/covid-19-drug-repurposing-database/).


Of these two respective sets, 168 and 163 compounds were annotated in QuartataWeb (Li H et al. Bioinformatics, 2020, 36, 3935-3937), which provided information on the targets of these compounds using DrugBank-all (Wishart D S et al. Nucleic Acids Res. 2018, 46, D1074-D1082) and STITCH-experimental (Szklarczyk D et al. Nucleic Acids Res. 2016, 44, D380-384) data as input. The remaining compounds were “manually” analyzed based on existing literature, as schematically described in the FIG. 6 for Dataset 1. FIG. 7-FIG. 8 shows that the host cell proteins most frequently targeted by the candidate antiviral compounds were adrenergic receptor α1A (gene ADRA1A), serotonin receptor 2A (HTR2A), and histamine H1 receptor (HRH1). Incidentally, ADRA1A and HRH1 were also among the most upregulated genes in SARS-CoV-2 infected A549 cells (Emanuel W et al. bioRxiv, 2020, https://doi.org/10.1101/2020.05.05.079194). Elevated HRH1 can be associated with hyperinflammation (Thurmond R L et al. Nat Rev Drug Discov. 2008, 7, 41-53). Serotonin receptor 2A was maximally targeted by potential anti-cytokine compounds drawing attention to the impact on neurotransmission.









TABLE 5







263 compounds with potential antiviral activity against SARS-CoV-2


infected A549 cells and corresponding CMap scores (all >90).











In-


CMap
Mechanism of


dex
Compound name
CMap ID
score
Action (MOA)














1
Salmeterol
BRD-A01320529
100.00
Adrenergic






receptor agonist


2
Avrainvillamide-
BRD-A70731303
99.99
nucleophosmin



analog-5


inhibitor


3
Terbutaline
BRD-A50157456
99.98
Adrenergic






receptor agonist


4
Oxybutynin
BRD-A65013509
99.98
Acetylcholine






receptor antagonist


5
Leflunomide
BRD-K78692225
99.98
Dihydroorotate






dehydrogenase






inhibitor, PDGFR






receptor inhibitor


6
GS-39783
BRD-K75478907
99.98
GABA receptor






modulator


7
nTZDpa
BRD-K54708045
99.98
PPAR receptor






agonist


8
n-arachidonyl-
BRD-K06024458
99.98
cannabinoid



GABA


receptor agonist


9
Salicin
BRD-K64614248
99.98
Anti-inflammatory


10
SR-27897
BRD-K35629949
99.98
CCK receptor






antagonist


11
Thiotepa
BRD-K09631521
99.98
Cytochrome






P450 inhibitor


12
Brazilin
BRD-A83326220
99.98
Nitric oxide






production inhibitor


13
Stavudine
BRD-K93880783
99.98
DNA directed






DNA polymerase






inhibitor, Reverse






transcriptase






inhibitor


14
Oxiconazole
BRD-K23369905
99.98
Bacterial cell






wall synthesis






inhibitor


15
Mirtazapine
BRD-A64977602
99.98
Adrenergic






receptor antagonist,






Serotonin receptor






antagonist


16
Liothyronine
BRD-K89152108
99.98
Thyroid hormone






stimulant


17
Bergenin
BRD-A15034104
99.98
Interleukin inhibitor


18
Chlorpromazine
BRD-K89997465
99.98
Dopamine receptor






antagonist


19
Fludarabine
BRD-K66788707
99.96
DNA synthesis






inhibitor, DNA






repair enzyme






inhibitor, Purine






antagonist


20
Iodophenpropit
BRD-K51918615
99.96
Histamine receptor






antagonist


21
SA-94315
BRD-K20197062
99.96
Caspase inhibitor


22
L-733060
BRD-K15791587
99.96
Tachykinin






antagonist


23
Tetrahydro-
BRD-A67605442
99.96
Nitric oxide (NO)



biopterin


stimulant, NO






synthase stimulant,






Phenylalanine






4-hydroxylase






stimulant


24
Duloxetine
BRD-K71103788
99.96
Serotonin and






norepinephrine






reuptake inhibitor


25
PSB-36
BRD-A70407468
99.96
Adenosine receptor






antagonist


26
CGP-7930
BRD-K65786282
99.95
GABA receptor






positive allosteric






modulator


27
Ochratoxin-a
BRD-K39944607
99.95
Phenylalanyl






tRNA synthetase






inhibitor


28
Alfacalcidol
BRD-K93433262
99.94
Vitamin D receptor






agonist


29
Molsidomine
BRD-K35531059
99.94
Guanylyl cyclase






activator


30
NGB-2904
BRD-K05181084
99.94
Dopamine receptor






antagonist


31
BH3I-1
BRD-A38913120
99.93
BCL inhibitor


32
Isotretinoin
BRD-K76723084
99.93
Retinoid receptor






agonist


33
Lonidamine
BRD-K96670504
99.92
Glucokinase






inhibitor


34
L-741626
BRD-K05181463
99.91
Dopamine receptor






antagonist


35
PG-9
BRD-A70268693
99.91
Acetylcholine






receptor agonist


36
GSK-1059615
BRD-K06750613
99.90
PI3K inhibitor


37
Cheno-
BRD-K18135438
99.90
11-β-HSD1



deoxycholic-


inhibitor, FXR



acid


agonist


38
Edrophonium
BRD-K81128206
99.89
Acetylcholinesterase






inhibitor


39
Nilotinib
BRD-K81528515
99.89
ABL inhibitor,






BCR-ABL kinase






inhibitor


40
5-nonyloxy-
BRD-K08219523
99.88
Serotonin receptor



tryptamine


agonist


41
Tribenoside
BRD-A60294240
99.88
Anti-inflammatory,






Capillary






stabilizing agent


42
Meclozine
BRD-A50311610
99.85
CAR agonist


43
Azathioprine
BRD-K32821942
99.85
Dehydrogenase






inhibitor


44
HDAC3-selective
BRD-K29313308
99.84
HDAC inhibitor


45
MRS-1845
BRD-A32949107
99.84
Calcium channel






blocker


46
KU-55933
BRD-K25311561
99.84
ATM kinase inhibitor


47
Arecaidine
BRD-K63792901
99.84
Acetylcholine






receptor agonist


48
Cortisone
BRD-A54487287
99.83
Glucocorticoid






receptor agonist


49
Torin-1
BRD-K40175214
99.80
MTOR inhibitor,






PI3K inhibitor


50
ENMD-2076
BRD-K68488863
99.79
FLT3 inhibitor,






VEGFR inhibitor,






Aurora kinase






inhibitor


51
Aspirin
BRD-K11433652
99.79
Cyclooxygenase






inhibitor


52
AS-703026
BRD-K89014967
99.74
MEK inhibitor


53
Razoxane
BRD-K07265709
99.74
Chelating agent,






Topoisomerase






inhibitor


54
Ibuprofen
BRD-A17655518
99.72
Cyclooxygenase






inhibitor, NFkB






pathway inhibitor


55
Demeclocycline
BRD-A75368507
99.71
Bacterial 30S






ribosomal subunit






inhibitor


56
KU-0063794
BRD-K67566344
99.71
MTOR inhibitor


57
Desoxy-
BRD-A75402480
99.69
Mineralocorticoid



corticosterone


receptor agonist


58
Imipramine
BRD-K38436528
99.65
Norepinephrine






and Serotonin






transporter inhibitor


59
AS-605240
BRD-K41895714
99.63
PI3K inhibitor


60
OSI-027
BRD-K94294671
99.62
MTOR inhibitor


61
PD-0325901
BRD-K49865102
99.59
MEK inhibitor,






MAPK inhibitor,






Protein kinase






inhibitor


62
PKCβ-inhibitor
BRD-K89687904
99.59
PKC inhibitor


63
Fostamatinib
BRD-K20285085
99.58
SYK inhibitor


64
Semaxanib
BRD-K63504947
99.57
VEGFR inhibitor


65
SB-216641
BRD-K30867024
99.57
Serotonin receptor






antagonist


66
Rescinnamine
BRD-K52930707
99.53
ACE inhibitor


67
Tolazoline
BRD-K46211610
99.53
Adrenergic receptor






antagonist


68
Dactolisib
BRD-K12184916
99.51
MTOR inhibitor,






PI3K inhibitor,






Protein kinase






inhibitor


69
GSK-1904529A
BRD-K04833372
99.50
IGF-1 inhibitor,






IGF-1R inhibitor,






Insulin receptor






ligand


70
ML-9
BRD-K68402494
99.44
Myosin light chain






kinase inhibitor


71
UNC-0321
BRD-K74236984
99.42
Histone lysine






methyltransferase






inhibitor


72
Pizotifen
BRD-K75958195
99.41
Serotonin receptor






antagonist


73
Pterostilbene
BRD-K92870997
99.40
Cyclooxygenase






inhibitor, PPAR






receptor agonist


74
VER-155008
BRD-K32330832
99.39
HSP inhibitor


75
PIK-90
BRD-K99818283
99.35
PI3K inhibitor


76
Panobinostat
BRD-K02130563
99.34
HDAC inhibitor


77
Oleoyl-
BRD-K66956375
99.31
Cannabinoid



ethanolamide


receptor agonist,






Glucose dependent






insulinotropic






receptor agonist,






Potassium channel






blocker, PPAR






receptor agonist


78
TC-2559
BRD-K67352070
99.30
Acetylcholine






receptor agonist


79
Mosapride
BRD-A39052811
99.26
Serotonin receptor






agonist


80
Dapsone
BRD-K62363391
99.21
Bacterial antifolate


81
BAY-36-7620
BRD-K54704028
99.20
Glutamate receptor






antagonist


82
Emetine
BRD-A25687296
99.20
Protein synthesis






inhibitor


83
Arecaidine
BRD-K23922020
99.19
Acetylcholine






receptor agonist


84
Apicidin
BRD-K64606589
99.18
HDAC inhibitor


85
Niacin
BRD-K61993165
99.16
NAD precursor






with lipid lowering






effect, vitamin B


86
Entinostat
BRD-K77908580
99.16
HDAC inhibitor


87
Elesclomol
BRD-K82135108
99.15
Oxidative stress






inducer


88
BMY-14802
BRD-A15435692
99.14
Sigma receptor






antagonist


89
Proxymetacaine
BRD-K79116891
99.14
Sodium channel






blocker


90
Zamifenacin
BRD-K80451230
99.14
Acetylcholine






receptor antagonist


91
Anandamide
BRD-K78280988
99.12
Cannabinoid






receptor agonist


92
Temsirolimus
BRD-A62025033
99.10
MTOR inhibitor


93
Desmethyl-
BRD-K10042277
99.07
Acetylcholine



clozapine


receptor agonist


94
QL-XII-47
BRD-U86922168
99.03
BTK inhibitor,






Cytoplasmic






tyrosine protein






kinase BMX






inhibitor


95
PI-103
BRD-K67868012
99.03
MTOR inhibitor,






PI3K inhibitor


96
Eugenol
BRD-K32977963
99.01
Androgen receptor






antagonist


97
SKF-81297
BRD-A09828896
98.98
Dopamine receptor






agonist


98
2-aminopurine
BRD-K35128472
98.97
Serine/threonine






kinase inhibitor


99
GBR-12783
BRD-K92015269
98.97
Dopamine uptake






inhibitor


100
Mephenytoin
BRD-A83937277
98.96
Hydantoin






antiepileptic


101
Terfenadine
BRD-A06352418
98.89
Histamine receptor






antagonist


102
KIN001-127
BRD-A29901043
98.89
ITK inhibitor


103
GBR-12935
BRD-K50135270
98.88
Dopamine uptake






inhibitor


104
Flavanone
BRD-A07824748
98.74
11-β-HSD1 inhibitor


105
VU-0366037-2
BRD-K39823328
98.73
Glutamate receptor






modulator


106
Ioxaglic-acid
BRD-K79124250
98.68
Radiopaque medium


107
FR-122047
BRD-K30990140
98.65
Cyclooxygenase






inhibitor


108
WZ-3146
BRD-K73293050
98.61
EGFR inhibitor


109
Droxinostat
BRD-K11558771
98.60
HDAC inhibitor


110
Cimaterol
BRD-A65440446
98.56
Adrenergic






receptor agonist


111
SCH-23390
BRD-K45435259
98.48
Dopamine






receptor antagonist


112
Ribavirin
BRD-A96255180
98.45
Antiviral


113
Mycophenolate-
BRD-K92428153
98.41
Dehydrogenase



mofetil


inhibitor,






Hydroxycarboxylic






acid receptor






agonist,






Immunosuppressant,






Inosine






monophosphate






dehydrogenase






inhibitor, Inositol






monophosphatase






inhibitor


114
Linsitinib
BRD-K08589866
98.37
IGF-1 inhibitor


115
LY-288513
BRD-K24675965
98.28
CCK receptor






antagonist


116
Wiskostatin
BRD-A18579359
98.25
Neural Wiskott-






Aldrich syndrome






protein inhibitor


117
AG-879
BRD-K59469039
98.23
Angiogenesis






inhibitor, Tyrosine






kinase inhibitor,






VEGFR inhibitor


118
BIX-01338
BRD-K26863634
98.21
Histone lysine






methyltransferase






inhibitor


119
Arcyriaflavin-a
BRD-K72726508
98.20
CDK inhibitor


120
AY-9944
BRD-K03642198
98.15
Hedgehog pathway






modulator


121
NU-7026
BRD-K09537769
98.15
DNA dependent






protein kinase






inhibitor, MTOR






inhibitor, PI3K






inhibitor


122
GW-9662
BRD-K93258693
98.08
PPAR receptor






antagonist


123
APHA-
BRD-K74733595
98.07
HDAC inhibitor



compound-8





124
Mefenamic-acid
BRD-K92778217
98.00
Cyclooxygenase






inhibitor


125
Heliomycin
BRD-K64517075
98.00
ATP synthase






inhibitor


126
PP-30
BRD-K30677119
97.98
RAF inhibitor


127
NVP-TAE684
BRD-K50140147
97.96
ALK inhibitor


128
Ropivacaine
BRD-K50938786
97.64
Sodium channel






blocker


129
MK-5108
BRD-K53665955
97.59
Aurora kinase






inhibitor


130
Ciclacillin
BRD-K89046952
97.54
Bacterial cell






wall synthesis






inhibitor


131
Sulfafurazole
BRD-K50859149
97.43
Bacterial antifolate


132
Dephostatin
BRD-K60274257
97.43
Tyrosine phosphatase






inhibitor


133
Entacapone
BRD-K83636919
97.31
Catechol O






methyltransferase






inhibitor


134
Oxfendazole
BRD-A33447119
97.21
Anthelmintic


135
Rottlerin
BRD-K03816923
97.11
MAP kinase






inhibitor, Protein






kinase inhibitor


136
Desipramine
BRD-K60762818
97.10
Tricyclic






antidepressant


137
Perospirone
BRD-K85503079
97.10
Dopamine and






serotonin receptors'






antagonist


138
Pimozide
BRD-K01292756
97.10
Dopamine receptor






antagonist


139
Ceforanide
BRD-K37848908
97.00
Penicillin binding






protein inhibitor


140
Equilin
BRD-K04046242
96.99
Estrogen receptor






agonist


141
SB-590885
BRD-K01253243
96.95
RAF inhibitor


142
LY-2140023
BRD-K49519144
96.93
Glutamate receptor






agonist


143
Glipizide
BRD-K12219985
96.92
Sulfonylurea


144
Moracizine
BRD-K21548250
96.91
Sodium channel






blocker


145
Kavain
BRD-A75455249
96.82
Calcium channel






modulator,






Sodium channel






blocker


146
Wortmannin
BRD-A11678676
96.81
PI3K inhibitor


147
Decitabine
BRD-K79254416
96.67
DNA






methyltransferase






inhibitor


148
Metformin
BRD-K79602928
96.61
Insulin sensitizer


149
Eicosatrienoic-
BRD-K63913457
96.58
Vasodilator



acid





150
Raloxifene
BRD-K63828191
96.58
Estrogen receptor






antagonist,






Selective estrogen






receptor






modulator (SERM)


151
Ezetimibe
BRD-A41519720
96.57
Niemann-Pick






C1-like 1 protein






antagonist,






Cholesterol






inhibitor


152
NBI-27914
BRD-K61177364
96.54
CRF receptor






antagonist


153
RS-67333
BRD-K46142322
96.52
Serotonin receptor






partial agonist


154
BMS-191011
BRD-K95609758
96.46
Potassium channel






activator


155
H-7
BRD-A55756846
96.39
PKA inhibitor


156
VU-0404997-2
BRD-A34208323
96.39
Glutamate receptor






modulator


157
Cisapride
BRD-K06895174
96.23
Serotonin receptor






agonist


158
Y-134
BRD-K94832621
96.20
Estrogen receptor






antagonist


159
Metrizamide
BRD-A45543382
95.98
Radiopaque medium


160
Dydrogesterone
BRD-K68620903
95.94
Progesterone receptor






agonist


161
Altrenogest
BRD-A27554692
95.93
Progestogen hormone


162
Homosalate
BRD-A34751532
95.90
HSP inducer


163
Bosutinib
BRD-K99964838
95.89
ABL inhibitor,






BCR-ABL kinase






inhibitor, SRC






inhibitor


164
Puromycin
BRD-A28970875
95.88
Protein synthesis






inhibitor


165
Methimazole
BRD-K54416256
95.71
Antithyroid


166
ALW-II-38-3
BRD-K68191783
95.60
Ephrin inhibitor


167
SN-38
BRD-A36630025
95.51
Topoisomerase






inhibitor


168
Ipratropium
BRD-A05352148
95.50
Acetylcholine






receptor antagonist


169
TGX-221
BRD-A41692738
95.47
PI3K inhibitor


170
Homo-
BRD-K76674262
95.43
Protein synthesis



harringtonine


inhibitor


171
Metergoline
BRD-A30435184
95.42
Dopamine receptor






agonist,






Serotonin receptor






antagonist


172
WZ-4-145
BRD-U25771771
95.41
EGFR inhibitor


173
Mercaptopurine
BRD-K91601245
95.33
Immunosuppressant,






Protein






synthesis inhibitor,






Purine






antagonist


174
Calmidazolium
BRD-A98283014
95.28
Calcium channel






blocker,






Calmodulin






antagonist


175
Mesna
BRD-
95.21
Antioxidant




M40783228




176
SDZ-205-557
BRD-K15868788
95.20
Serotonin receptor






antagonist


177
Procyclidine
BRD-A31800922
95.16
Acetylcholine






receptor antagonist


178
Amiodarone
BRD-K17561142
95.15
Potassium channel






blocker


179
Midodrine
BRD-A79981887
95.14
Adrenergic receptor






agonist


180
Mepireserpate
BRD-A71765365
95.11
Catecholamine






depleting






sympatholytic


181
SA-792728
BRD-K20755323
95.06
Sphingosine kinase






inhibitor


182
Brompheniramine
BRD-A68723818
94.90
Histamine receptor






antagonist


183
Sumatriptan
BRD-K50938287
94.83
Serotonin receptor






agonist


184
Gemcitabine
BRD-K15108141
94.82
Ribonucleotide






reductase inhibitor


185
JAK3-Inhibitor-II
BRD-K52850071
94.81
JAK inhibitor


186
CHEMBL-
BRD-K59962020
94.79
NFkB pathway



374350


inhibitor


187
Vorinostat
BRD-K81418486
94.70
HDAC inhibitor


188
Dipyridamole
BRD-K86301799
94.48
Phosphodiesterase






inhibitor


189
JNJ-16259685
BRD-K64670467
94.47
Glutamate






receptor antagonist


190
VU-0415374-1
BRD-K83010055
94.46
Glutamate receptor






modulator


191
Pidorubicine
BRD-K04548931
94.42
Topoisomerase






inhibitor


192
KU-C103443N
BRD-A81402010
94.22
CDC inhibitor,






Rho associated






kinase inhibitor


193
Dichlorobenzamil
BRD-K12906962
94.16
Sodium/calcium






exchange inhibitor


194
Mepacrine
BRD-A45889380
94.14
Cytokine production






inhibitor,






NFkB pathway






inhibitor, TP53






activator


195
E-4031
BRD-K41713976
93.96
Potassium






channel blocker


196
Narciclasine
BRD-K06792661
93.94
Coflilin






signaling pathway






activator, LIM






kinase activator,






Rho associated






kinase activator


197
Mesoridazine
BRD-A14395271
93.80
Dopamine






receptor antagonist


198
Tranylcypromine
BRD-A43974575
93.79
Monoamine






oxidase inhibitor


199
Lypressin
BRD-K93331255
93.76
Vasopressin






receptor agonist


200
Reserpine
BRD-K95921201
93.69
Vesicular






monoamine






transporter






inhibitor


201
Abiraterone
BRD-K55301415
93.64
17, 20 lyase






inhibitor, Androgen






biosynthesis






inhibitor,






Cytochrome






P450 inhibitor,






Steroid sulfatase






inhibitor


202
I-OMe-AG-538
BRD-K35377380
93.59
IGF-1 inhibitor


203
Somatostatin
BRD-K14681867
93.48
Somatostatin






receptor agonist


204
Splitomycin
BRD-K27710560
93.37
SIRT inhibitor


205
AM-281
BRD-K59419204
93.34
Cannabinoid






receptor antagonist


206
Sphingosine
BRD-K62959606
93.32
Ceramidase inhibitor


207
Hydroxy-
BRD-A36707673
93.30
LXR agonist



cholesterol





208
TPCA-1
BRD-K51575138
93.29
IKK inhibitor


209
FGIN-1-27
BRD-K09778810
93.26
Inositol






monophosphatase






inhibitor


210
Trichostatin-a
BRD-K68202742
93.06
HDAC inhibitor,






CDK activator,






ID1 inhibitor


211
Hexylresorcinol
BRD-K99946902
92.99
Local anesthetic


212
Epicatechin
BRD-K50660797
92.96
Bacterial DNA






gyrase inhibitor,






Cyclooxygenase






inhibitor, DNA






polymerase inhibitor


213
RS-17053
BRD-K76840893
92.95
Adrenergic






receptor antagonist


214
NSC-663284
BRD-K03109492
92.78
CDC inhibitor


215
L-165041
BRD-K40656405
92.55
PPAR receptor






agonist


216
ML-7
BRD-K93201660
92.46
Myosin light chain






kinase inhibitor


217
Alisertib
BRD-K75295174
92.44
Aurora kinase






inhibitor


218
GR-127935
BRD-K11911061
92.37
Serotonin receptor






antagonist


219
Clobenpropit
BRD-K71430621
92.36
Histamine receptor






antagonist


220
NNC-55-0396
BRD-K78122587
92.31
T-type calcium






channel blocker


221
Barasertib
BRD-K63923597
92.27
Aurora kinase






inhibitor


222
Benidipine
BRD-A35519318
92.25
Calcium channel






blocker


223
Sertraline
BRD-K82036761
92.24
Serotonin






receptor antagonist


224
ZM-447439
BRD-K72703948
92.16
Aurora kinase






inhibitor


225
BIBX-1382
BRD-K70914287
92.15
EGFR inhibitor,






Tyrosine kinase






inhibitor


226
Immethridine
BRD-K49519092
92.15
Histamine receptor






agonist


227
MR-16728
BRD-A30590053
92.15
Acetylcholine






release enhancer,






Acetylcholine






release stimulant


228
Heraclenol
BRD-A77050075
92.11
Vitamin K antagonist


229
NU-7441
BRD-K00337317
92.10
DNA dependent






protein kinase






inhibitor,






P-glycoprotein






inhibitor


230
Hyoscyamine
BRD-K40530731
91.85
Acetylcholine






receptor antagonist


231
m-
BRD-K36965586
91.76
Serotonin receptor



chlorophenyl-


agonist



biguanide





232
Prostaglandin-b2
BRD-K82865713
91.75
cAMP inhibitor


233
BML-ST330
BRD-A77118605
91.59
Phospholipase






inhibitor


234
STO-609
BRD-K52620403
91.57
Calmodulin






antagonist


235
Tyrphostin-
BRD-K14441456
91.50
EGFR inhibitor



AG-556





236
Corynanthine
BRD-K06467078
91.46
Adrenergic receptor






antagonist


237
PD-102807
BRD-A89337244
91.31
Acetylcholine






receptor antagonist


238
Norgestrel
BRD-A50928468
91.25
Progesterone






receptor agonist


239
Telmisartan
BRD-K73999723
91.25
Angiotensin






receptor antagonist


240
BMY-45778
BRD-K84895041
91.22
IP1 prostacyclin






receptor agonist


241
Dihydrosamidin
BRD-K63945320
91.19
Phospholipase






inhibitor, Nitric






oxide production






inhibitor, platelet






activating factor






receptor






antagonist


242
HG-6-64-01
BRD-U37049823
91.09
RAF inhibitor


243
KUC104502N
BRD-K24538644
91.06



244
Formestane
BRD-A31801025
91.00
Aromatase inhibitor


245
BRD-K64835161
BRD-K64835161
90.93



246
M2-PK-activator
BRD-K80672993
90.89



247
Cetraxate
BRD-K48932581
90.81
Mucus protecting






agent


248
Terbinafine
BRD-K68132782
90.76
Fungal squalene






epoxidase






inhibitor


249
Phospho-
BRD-K68873215
90.69
Phosphodiesterase



diesterase-V-


inhibitor



inhibitor-II





250
Ponalrestat
BRD-K68332390
90.68
Aldose reductase






inhibitor


251
Phenytoin
BRD-K55930204
90.67
Hydantoin






antiepileptic


252
Phylloquinone
BRD-A55815733
90.64
Vitamin K, Γ






carboxylase enzyme


253
AZD-8055
BRD-K69932463
90.64
MTOR inhibitor


254
PHA-665752
BRD-K95435023
90.53
c-Met inhibitor


255
PD-184352
BRD-K05104363
90.52
MEK inhibitor


256
RU-28318
BRD-A92585442
90.43
Cytochrome P450






inhibitor


257
Fenoldopam
BRD-A50684349
90.38
Dopamine receptor






agonist


258
Camptothecin
BRD-A30437061
90.32
Topoisomerase






inhibitor


259
Tretinoin
BRD-K06926592
90.31
Retinoid receptor






agonist, Retinoid






receptor ligand


260
Metixene
BRD-A33711280
90.16
Acetylcholine






receptor antagonist


261
Tetra-
BRD-A43940795
90.12
Serotonin release



hydropalmatine


inhibitor


262
YM-976
BRD-K12932420
90.09
Phosphodiesterase






inhibitor


263
Alaproclate
BRD-A14966924
90.03
Serotonin receptor






antagonist
















TABLE 6







275 compounds with CMap scores <−90, which can


potentially elicit anti-cytokine activity against


hyperinflammation in SARS-CoV-2-infected A549-ACE2 cells.











In-
Compound

CMap
Mechanism of


dex
name
CMap ID
score
Action (MOA)














1
n-(3-acetami-
BRD-K61217870
−100
Glutamate



dophenyl)-3-


receptor



chloro-


antagonist



benzamide





2
BI-78D3
BRD-K73982490
 −99.98
JNK inhibitor


3
Xaliproden
BRD-K88358234
 −99.98
Serotonin






receptor agonist


4
Rhamnetin
BRD-K37206356
 −99.98
HDAC inhibitor


5
MR-16728
BRD-A30590053
 −99.98
Acetylcholine






release enhancer






or stimulant


6
Palonosetron
BRD-K08924299
 −99.98
Serotonin






receptor antagonist


7
Clarithromycin
BRD-K49668410
 −99.98
Bacterial 50S






ribosomal subunit






inhibitor


8
SAL-1
BRD-K40213712
 −99.98
Adenosine






receptor antagonist


9
Nimodipine
BRD-A58048407
 −99.98
Calcium channel






blocker


10
Isoliquiritigenin
BRD-K33583600
 −99.98
Guanylate cyclase






activator


11
Cyclazosin
BRD-A37837077
 −99.98
Adrenergic






receptor antagonist


12
Eicosatetray-
BRD-K06080977
 −99.98
Cyclooxygenase



noic-acid


inhibitor,






Lipoxygenase






inhibitor


13
Oxantel
BRD-K66019333
 −99.96
Anthelmintic


14
Nor-
BRD-A11135865
 −99.96
Opioid receptor



binaltorphimine


antagonist


15
PCA-4248
BRD-A29289453
 −99.95
Platelet activating






factor






receptor






antagonist


16
Ketanserin
BRD-K49671696
 −99.95
Serotonin receptor






antagonist


17
Tyrphostin-
BRD-K03670461
 −99.94
EGFR inhibitor



AG-82





18
Azelastine
BRD-A68888262
 −99.94
Histamine receptor






antagonist


19
Diethyl-
BRD-K45330754
 −99.93
Estrogen receptor



stilbestrol


agonist


20
Raltegravir
BRD-K05658747
 −99.93
HIV integrase






inhibitor


21
KI-16425
BRD-A25569250
 −99.93
Lysophosphatidic






acid receptor






antagonist


22
Pyroxamide
BRD-K11663430
 −99.93
HDAC inhibitor


23
Maprotiline
BRD-K03319035
 −99.93
Norepinephrine






reuptake






inhibitor, Tricyclic






antidepressant


24
Reserpic-acid
BRD-K32755366
 −99.91
Norepinephrine






transporter






inhibitor


25
Fostamatinib
BRD-K20285085
 −99.91
SYK inhibitor


26
Y-27632
BRD-K44084986
 −99.91
Rho associated






kinase inhibitor


27
Dexketoprofen
BRD-K43764301
 −99.9
Cyclooxygenase






inhibitor


28
EMF-bca1-60
BRD-K68437527
 −99.9
caspase inhibitor


29
Fluphenazine
BRD-K55127134
 −99.9
Dopamine receptor






antagonist


30
Gabazine
BRD-K93280214
 −99.89
GABA receptor






antagonist


31
α-estradiol
BRD-A60070924
 −99.88
Estrogen receptor






agonist


32
Benzydamine
BRD-K76133116
 −99.88
Membrane integrity






inhibitor,






Prostanoid receptor






antagonist,






Prostanoid receptor






inhibitor


33
Navitoclax
BRD-K82746043
 −99.88
BCL inhibitor


34
Nifurtimox
BRD-A00100033
 −99.86
DNA inhibitor


35
Thenoyltri-
BRD-K00959089
 −99.86
Chelating agent



fluoroacetone





36
NVP-AUY922
BRD-K41859756
 −99.86
HSP inhibitor


37
BRD-
BRD-K64835161
 −99.85
NA



K64835161





38
Atorvastatin
BRD-U88459701
 −99.85
HMGCR (HMG






CoA reductase)






inhibitor


39
Securinine
BRD-A25775766
 −99.82
GABA receptor






antagonist,






TP53 activator


40
Nikkomycin
BRD-A74771556
 −99.82
Chitin inhibitor


41
Zuclopenthixol
BRD-K28761384
 −99.78
Dopamine receptor






antagonist


42
Temozolomide
BRD-K32107296
 −99.78
DNA alkylating






agent


43
HY-11007
BRD-K97056771
 −99.77
BCR-ABL kinase






inhibitor


44
Salsolinol
BRD-K99595596
 −99.74
Monoamine






oxidase inhibitor,






Tyrosine hydroxylase






inhibitor


45
SCH-28080
BRD-K55748775
 −99.73
ATPase inhibitor


46
Retinol
BRD-K13927029
 −99.72
Retinoid receptor






ligand


47
SB-216763
BRD-K59184148
 −99.72
Glycogen synthase






kinase






inhibitor


48
YS-035
BRD-K06208435
 −99.71
Calcium channel






blocker


49
Bisoprolol
BRD-A89175223
 −99.69
Adrenergic receptor






antagonist


50
Carteolol
BRD-A42167015
 −99.62
Adrenergic receptor






antagonist


51
TER-14687
BRD-A33833419
 −99.62
Inhibitor of






translocation of






PKCq in T cells


52
Selegiline
BRD-K86434416
 −99.59
Monoamine






oxidase inhibitor


53
Triptolide
BRD-A13122391
 −99.58
RNA polymerase






inhibitor


54
Lisuride
BRD-K88871508
 −99.57
Dopamine receptor






agonist


55
Topiramate
BRD-K29653726
 −99.57
Carbonic






anhydrase inhibitor,






Glutamate receptor






antagonist,






Kainate receptor






antagonist


56
Berbamine
BRD-K50464341
 −99.55
Calmodulin antagonist


57
Hexamethyl-
BRD-K40990712
 −99.53
Sodium/hydrogen



eneamiloride


antiport






inhibitor


58
MW-STK33-
BRD-K64310881
 −99.52
Potassium channel



3B


activator


59
MLN-4924
BRD-K67844266
 −99.5
Nedd activating






enzyme






inhibitor


60
EHNA
BRD-K27450477
 −99.48
Adenosine






deaminase inhibitor


61
Chlor-
BRD-K59058766
 −99.46
Dopamine receptor



prothixene


antagonist


62
DUP-697
BRD-K06221026
 −99.45
Cyclooxygenase






inhibitor


63
Chlor-
BRD-A04553218
 −99.45
Histamine receptor



phenamine


antagonist


64
NAS-181
BRD-A23683907
 −99.38
Serotonin receptor






antagonist


65
Linsitinib
BRD-K08589866
 −99.37
IGF-1 inhibitor,






insulin






inhibitor, ARF6






and TBK1






activator


66
YC-1
BRD-K60476892
 −99.34
Guanylyl cyclase






activator


67
Olanzapine
BRD-K18895904
 −99.32
Dopamine






receptor/serotonin






receptor antagonist


68
Orantinib
BRD-K91696562
 −99.3
FGFR, VEGFR,






PDGFR






inhibitor


69
Phenelzine
BRD-K87024524
 −99.3
Monoamine






oxidase inhibitor


70
TGX-221
BRD-A41692738
 −99.29
PI3K inhibitor


71
Latrepirdine
BRD-K55703048
 −99.29
Glutamate receptor






antagonist


72
PU-H71
BRD-K36529613
 −99.28
HSP inhibitor


73
Bromocriptine
BRD-A69960130
 −99.24
Dopamine receptor






agonist


74
Syrosingopine
BRD-K14200658
 −99.21
Vesicular monoamine






transporter inhibitor


75
UNC-0321
BRD-K74236984
 −99.21
Histone lysine






methyltransferase






inhibitor


76
BRD-
BRD-A80383043
 −99.19
Glutamate



A80383043


receptor agonist






and/or antagonist


77
Trifluoperazine
BRD-K89732114
 −99.17
Dopamine






receptor antagonist


78
AQ-RA741
BRD-K81729199
 −99.16
Acetylcholine receptor






antagonist


79
Bromfenac
BRD-K47679368
 −99.14
Cyclooxygenase






inhibitor


80
Oxaprozin
BRD-K25394294
 −99.14
Cyclooxygenase






inhibitor


81
CGS-20625
BRD-K68103045
 −99.12
Benzodiazepine






receptor






agonist, GABA






benzodiazepine






site receptor






partial agonist


82
Rucaparib
BRD-K88560311
 −99.11
PARP inhibitor


83
o-3M3FBS
BRD-K46384212
 −99.09
phospholipase






activator


84
L-655240
BRD-K89402695
 −99.07
Thromboxane receptor






antagonist


85
L-750667
BRD-K28806945
 −99.04
Dopamine receptor






antagonist


86
Daunorubicin
BRD-K43389675
 −98.99
RNA synthesis






inhibitor,






Topoisomerase






inhibitor


87
Profenamine
BRD-A16311756
 −98.98
Butyrylcholinesterase






inhibitor,






Cholinergic receptor






antagonist


88
Saracatinib
BRD-K19540840
 −98.97
SRC inhibitor


89
Trazodone
BRD-K70778732
 −98.96
Adrenergic receptor






antagonist,






Serotonin receptor






antagonist,






Serotonin reuptake






inhibitor


90
Valproic-acid
BRD-K41260949
 −98.93
HDAC inhibitor


91
Medetomidine
BRD-A66563878
 −98.93
Adrenergic receptor






agonist


92
Piperine
BRD-K59522102
 −98.92
Monoamine oxidase






inhibitor


93
Pyrazinamide
BRD-K28667793
 −98.83
Fatty acid synthase






inhibitor


94
Fraxidin
BRD-K66944906
 −98.83
Carbonic anhydrase






inhibitor


95
Larixinic-acid
BRD-K40619305
 −98.78
Compound that






interacts with






metal centers


96
Midodrine
BRD-A79981887
 −98.74
Adrenergic receptor






agonist


97
XAV-939
BRD-K12762134
 −98.73
Tankyrase inhibitor


98
AICA-
BRD-A67373739
 −98.7
AMPK activator



ribonucleotide





99
PIK-90
BRD-K99818283
 −98.66
PI3K inhibitor


100
PNU-22394
BRD-K16551401
 −98.63
Serotonin receptor






agonist


101
AY-9944
BRD-K03642198
 −98.62
Hedgehog pathway






modulator


102
Gavestinel
BRD-K49890030
 −98.59
Glutamate receptor






antagonist


103
Foliosidine
BRD-A49734948
 −98.54
Plant alkaloid


104
Naftopidil
BRD-A01787639
 −98.48
Adrenergic receptor






antagonist


105
GDC-0941
BRD-K52911425
 −98.38
PI3K inhibitor


106
Clonidine
BRD-K98530306
 −98.37
Adrenergic receptor






agonist


107
CGP-54626
BRD-A55369275
 −98.35
GABA receptor






antagonist


108
Tosyllysyl-
BRD-K10136726
 −98.32
Chymotrypsin



chloromethyl-


inhibitor



ketone





109
Indatraline
BRD-K01649396
 −98.31
Norepinephrine






transporter






inhibitor


110
9-methyl-
BRD-K14696368
 −98.28
NFkB pathway



5H-6-thia-4,5-


inhibitor



diaza-chrysene-






6,6-dioxide





111
NNC-05-2090
BRD-K85015012
 −98.27
GAT inhibitor,






GABA uptake






inhibitor


112
3-matida
BRD-A87125127
 −98.19
Glutamate receptor






antagonist


113
Phenothiazine
BRD-K59597909
 −98.17
Dopamine receptor






antagonist


114
Piribedil
BRD-K47936004
 −98.16
Dopamine receptor






agonist


115
Mebeverine
BRD-A09467419
 −98.09
Acetylcholine






receptor






antagonist


116
GANT-58
BRD-K64451768
 −98.06
GLI antagonist


117
RITA
BRD-K00317371
 −98.03
MDM inhibitor


118
Pirenperone
BRD-K25224017
 −98.01
Serotonin receptor






antagonist


119
KIN001-244
BRD-K09186807
 −98.01
Phosphoinositide






dependent






kinase inhibitor


120
Ozagrel
BRD-K19525698
 −97.97
Thromboxane






synthase inhibitor


121
PP-30
BRD-K30677119
 −97.96
RAF inhibitor


122
CNQX
BRD-K53545112
 −97.96
Glutamate receptor






antagonist


123
STO-609
BRD-K52620403
 −97.91
Calmodulin antagonist


124
Loperamide
BRD-K61250553
 −97.74
Opioid receptor






agonist


125
Dichloroacetic-
BRD-K13664374
 −97.69
Pyruvate



acid


dehydrogenase kinase






inhibitor


126
PI-103
BRD-K67868012
 −97.66
MTOR inhibitor,






PI3K inhibitor


127
Metoclopramide
BRD-K75641298
 −97.54
Dopamine






receptor and






serotonin receptor






antagonist


128
Spironolactone
BRD-K90027355
 −97.54
Mineralocorticoid






receptor






antagonist


129
Loratadine
BRD-K82795137
 −97.51
Histamine receptor






antagonist


130
Pirfenidone
BRD-K96862998
 −97.45
TGF β receptor






inhibitor


131
ICI-89406
BRD-A03359064
 −97.34
Adrenergic receptor






antagonist


132
Clebopride
BRD-K17294426
 −97.31
Dopamine receptor






antagonist


133
Prostaglandin-
BRD-K04010869
 −97.3
HSP inducer,



a1


NFkB pathway






inhibitor


134
Butylparaben
BRD-K08287586
 −97.27
DNA synthesis






inhibitor


135
Testosterone
BRD-A48720949
 −97.13
androgen receptor






agonist


136
WZ-4002
BRD-K72420232
 −97.11
EGFR inhibitor


137
PLX-4720
BRD-K16478699
 −97.01
RAF inhibitor


138
Darinaparsin
BRD-K35723520
 −97.01
Apoptosis stimulant


139
PTB1
BRD-K16554956
 −96.94
AMPK activator


140
Alosetron
BRD-K46742498
 −96.93
Serotonin receptor






antagonist


141
U-99194
BRD-K70281171
 −96.87
Dopamine receptor






antagonist


142
Otenzepad
BRD-A00520476
 −96.85
Acetylcholine






receptor






antagonist


143
Fursultiamine
BRD-A71157293
 −96.84
Vitamin B


144
Piperacetazine
BRD-K16277217
 −96.79
Dopamine receptor






antagonist


145
SD-169
BRD-K91904471
 −96.77
p38 MAPK inhibitor


146
Liothyronine
BRD-K89152108
 −96.77
Thyroid hormone






stimulant


147
Mepacrine
BRD-A45889380
 −96.77
Cytokine production






inhibitor,






NFkB pathway






inhibitor, TP53






activator


148
Nicotine
BRD-K05395900
 −96.71
Acetylcholine






receptor agonist


149
TG-101348
BRD-K12502280
 −96.66
FLT3 inhibitor,






JAK inhibitor


150
Quinpirole
BRD-A85280935
 −96.54
Dopamine receptor






agonist


151
Mafenide
BRD-K30649484
 −96.53
Carbonic anhydrase






inhibitor


152
Dephostatin
BRD-K60274257
 −96.5
Tyrosine phosphatase






inhibitor


153
Cisapride
BRD-K06895174
 −96.49
Serotonin receptor






agonist


154
Dicyclo-
BRD-K81521265
 −96.48
Epoxide hydolase



hexylurea


inhibitor


155
m-
BRD-K36965586
 −96.47
Serotonin receptor



chlorophenyl-


agonist



biguanide





156
Auraptene
BRD-K85013741
 −96.45
Nitric oxide






production inhibitor


157
Alprenolol
BRD-A00993607
 −96.44
Adrenergic






receptor antagonist


158
TPCA-1
BRD-K51575138
 −96.39
IKK inhibitor


159
Sertraline
BRD-K82036761
 −96.37
Serotonin receptor






antagonist


160
AC-55649
BRD-K93176058
 −96.37
Retinoid receptor






agonist


161
CDK1-5-
BRD-K87932577
 −96.35
CDK inhibitor,



inhibitor


Glycogen






synthase kinase






inhibitor


162
D-64406
BRD-K27665173
 −96.25
PDGFR receptor






inhibitor


163
Fipronil
BRD-A50675702
 −96.24
GABA gated






chloride channel






blocker


164
RO-25-6981
BRD-K51541829
 −96.14
Ionotropic






glutamate receptor






antagonist, Monamine






transporter modulator


165
BIIB021
BRD-K51967704
 −96.11
HSP inhibitor


166
AZD-6482
BRD-K58772419
 −96.1
PI3K inhibitor


167
EMD-386088
BRD-K47659338
 −96.09
Serotonin receptor






agonist


168
CITCO
BRD-K53263234
 −96.07
CAR agonist


169
Exemestane
BRD-A73741725
 −95.97
Aromatase inhibitor


170
GR-206
BRD-K00184207
 −95.95
Aryl hydrocarbon






receptor






ligand


171
Dasatinib
BRD-K49328571
 −95.91
BCR-ABL kinase






inhibitor,






Ephrin inhibitor,






KIT inhibitor,






PDGFR receptor






inhibitor, SRC






inhibitor,






Tyrosine kinase






inhibitor


172
MDM2-
BRD-K84987553
 −95.89
MDM inhibitor



inhibitor





173
Aminomethyl-
BRD-A28318179
 −95.8
Nitric oxide



transferase


synthase inhibitor


174
BRL-52537
BRD-A37347161
 −95.7
Opioid receptor






agonist


175
Amoxapine
BRD-K02265150
 −95.68
Norepinephrine






reuptake






inhibitor


176
RO-08-2750
BRD-K00486786
 −95.68
NGF binding inhibitor


177
Flutamide
BRD-K28307902
 −95.68
Androgen receptor






antagonist


178
DMBI
BRD-K96084870
 −95.63
PDGFR receptor






inhibitor,






VEGFR inhibitor


179
Carmoxirole
BRD-K82484965
 −95.61
Dopamine receptor






agonist


180
Tauro-
BRD-K33572481
 −95.59
Bile acid



deoxycholic-






acid





181
Bupropion
BRD-A05186015
 −95.57
Dopamine uptake






inhibitor


182
Chlor-
BRD-K86595100
 −95.49
Benzodiazepine



diazepoxide


receptor agonist


183
Roscovitine
BRD-K07691486
 −95.37
CDK inhibitor


184
ALW-II-38-3
BRD-K68191783
 −95.33
Ephrin inhibitor


185
Ornidazole
BRD-A42759514
 −95.22
Antiprotozoal


186
Iodophenpropit
BRD-K51918615
 −95.18
Histamine receptor






antagonist


187
Prima-1-met
BRD-K49456190
 −95.18
thioredoxin inhibitor


188
EI-247
BRD-K32710582
 −95.17
IGF-1 inhibitor


189
MK-2206
BRD-K68065987
 −95.16
AKT inhibitor


190
BMS-299897
BRD-K02950022
 −95.14
γ secretase inhibitor


191
Promazine
BRD-K06980535
 −95.11
Dopamine receptor






antagonist


192
CGP-60474
BRD-K79090631
 −95.09
CDK inhibitor


193
PF-04217903
BRD-K73319509
 −95.06
c-Met inhibitor


194
Pantoprazole
BRD-A22380646
 −95.02
ATPase inhibitor


195
Norgestimate
BRD-A04756508
 −94.95
Progesterone receptor






agonist


196
Mead-
BRD-K09764130
 −94.82
Cannabinoid receptor



ethanolamide


agonist


197
CL-82198
BRD-K00675675
 −94.73
Metalloproteinase






inhibitor


198
HLI-373
BRD-K17349619
 −94.73
MDM inhibitor


199
Nifedipine
BRD-K96354014
 −94.71
Calcium channel






blocker


200
Sildenafil
BRD-K50128260
 −94.71
Phosphodiesterase






inhibitor


201
ICI-199441
BRD-K73290745
 −94.63
Opioid receptor






agonist


202
Ipsapirone
BRD-K90574421
 −94.61
Serotonin receptor






agonist


203
Milrinone
BRD-K67080878
 −94.49
Phosphodiesterase






inhibitor


204
Cotinine
BRD-K94144010
 −94.35
Nicotine metabolite


205
Etilefrine
BRD-A09925278
 −94.23
Adrenergic receptor






agonist


206
Thio-
BRD-K08619574
 −94.19
Dopamine receptor



properazine


antagonist


207
Acadesine
BRD-A95696820
 −94.15
AMPK activator


208
Danazol
BRD-A92537424
 −94.09
Estrogen receptor






antagonist,






Progesterone






receptor agonist


209
z-prolyl-p
BRD-K60174629
 −94.04
Prolyl endopeptidase



rolinal


inhibitor


210
Pravastatin
BRD-K60511616
 −94.03
HMGCR inhibitor


211
PP-2
BRD-K95785537
 −94.01
SRC inhibitor


212
BRD-
BRD-K63784565
 −94.01
Topoisomerase



K63784565


inhibitor


213
Geldanamycin
BRD-A19500257
 −93.9
HSP inhibitor


214
Verapamil
BRD-A09533288
 −93.88
Calcium channel






blocker


215
Amylocaine
BRD-A09062839
 −93.70
Local anesthetic


216
Anagrelide
BRD-K62200014
 −93.38
Phosphodiesterase






inhibitor


217
JAK3-
BRD-K52850071
 −93.37
JAK inhibitor



Inhibitor-II





218
Felbamate
BRD-K99107520
 −93.35
Glutamate receptor






antagonist


219
BP-554
BRD-K45479396
 −93.33
Serotonin receptor






agonist


220
Dicycloverine
BRD-K68507560
 −93.3
Acetylcholine receptor






antagonist


221
Nicorandil
BRD-K97752965
 −93.29
Nitric oxide donor,






Potassium






channel activator


222
SCH-442416
BRD-K46469693
 −93.27
Adenosine receptor






antagonist


223
Carpindolol
BRD-A15530910
 −93.19
Adrenergic receptor






antagonist,






serotonin receptor






antagonist


224
VU-0420363-1
BRD-K59633790
 −92.96
SARS coronavirus






3C-like






protease inhibitor


225
Oxybutynin
BRD-A65013509
 −92.9
Acetylcholine receptor






antagonist


226
SA-792541
BRD-K68143200
 −92.8
CDC inhibitor


227
Dipropyl-5ct
BRD-K32645441
 −92.74
Serotonin receptor






agonist


228
Ticlopidine
BRD-K00603606
 −92.72
Purinergic receptor






antagonist


229
SDZ-WAG-
BRD-A31007383
 −92.71
Adenosine receptor



994


agonist


230
Meprylcaine
BRD-K65417056
 −92.69
Local anesthetic


231
Cycloserine
BRD-K87226815
 −92.58
Bacterial cell






wall synthesis






inhibitor


232
KIN001-127
BRD-A29901043
 −92.54
ITK inhibitor


233
Enrofloxacin
BRD-K76534306
 −92.54
Bacterial DNA






gyrase inhibitor


234
Alverine
BRD-K89055274
 −92.52
Muscle relaxant


235
Bepridil
BRD-A91008255
 −92.5
Calcium channel






or L-type Ca++






channel blocker


236
Nefazodone
BRD-K90789829
 −92.39
Adrenergic inhibitor,






Norepinephrine






reuptake






inhibitor, Serotonin






receptor






antagonist,






Serotonin reuptake






inhibitor


237
PSB-11
BRD-K10177585
 −92.33
Adenosine receptor






antagonist


238
Acetyl-
BRD-U01690642
 −92.32
Isoprenylated protein



geranyl-


methylation inhibitor



cysteine





239
Estradiol
BRD-A18917088
 −92.31
Contraceptive






agent, Estrogen






receptor agonist


240
Seco-
BRD-K91733562
 −92.2
Antioxidant



isolariciresinol





241
Prostaglandin
BRD-K09436313
 −92.13
Prostanoid receptor






antagonist


242
Alfuzosin
BRD-A09056319
 −92.11
Adrenergic receptor






antagonist


243
Oxybenzone
BRD-K59037100
 −92.1
Lipase inhibitor


244
KIN001-220
BRD-K53561341
 −91.98
Aurora kinase






inhibitor


245
AZD-8055
BRD-K69932463
 −91.85
MTOR inhibitor


246
Toltrazuril
BRD-K64514229
 −91.83
Antiprotozoal


247
Mepyramine
BRD-K97564742
 −91.79
Histamine receptor






antagonist


248
Edaravone
BRD-K35458079
 −91.74
Nootropic agent


249
FIT
BRD-K17896185
 −91.6
Opioid receptor






agonist


250
Dopamine
BRD-K43887077
 −91.51
Dopamine receptor






agonist


251
Tolterodine
BRD-K54316499
 −91.49
Acetylcholine receptor






antagonist


252
L-BSO
BRD-A47706533
 −91.44
Glutathione transferase






inhibitor


253
Dinoprostone
BRD-K26521938
 −91.34
Prostanoid receptor






agonist


254
GR-144053
BRD-K12120659
 −91.31
Integrin antagonist


255
O-2050
BRD-K02590140
 −91.29
Cannabinoid receptor






antagonist


256
bis-tyrphostin
BRD-K32906660
 −91.25
EGFR inhibitor


257
ITE
BRD-K60298136
 −91.16
Aryl hydrocarbon






receptor






agonist


258
Nevirapine
BRD-K15502390
 −91.15
Reverse transcriptase






inhibitor


259
GR-235
BRD-K26674531
 −91.08
Estrogen receptor






agonist, FXR






antagonist,






Progesterone






receptor agonist


260
Latrunculin-b
BRD-A19248578
 −91.08
Actin polymerization






inhibitor,






Unidentified






pharmacological






activity


261
AR-
BRD-K40892394
 −91.08
Nitric oxide synthase



C133057XX


inhibitor


262
Temefos
BRD-K51805276
 −90.87
Cholinesterase






inhibitor


263
Ilomastat
BRD-K51662849
 −90.81
Matrix






metalloprotease






inhibitor


264
SID-26681509
BRD-K08417745
 −90.75
Cathepsin inhibitor


265
Formestane
BRD-A31801025
 −90.66
Aromatase inhibitor


266
Iproniazid
BRD-K88568253
 −90.6
Monoamine oxidase






inhibitor


267
Buphenine
BRD-A36267905
 −90.46
Adrenergic receptor






agonist


268
Desipramine
BRD-K60762818
 −90.43
Tricyclic






antidepressant


269
Tyrphostin-46
BRD-K60184833
 −90.39
Tyrosine kinase






inhibitor


270
RS-67506
BRD-K50018155
 −90.29
Serotonin






receptor partial






agonist


271
BRD-
BRD-K34437622
 −90.15
Thymidylate



K34437622


synthase inhibitor


272
Rotenonic-acid
BRD-K34330170
 −90.11
Retinoid receptor






antagonist


273
PIK-75
BRD-
 −90.1
DNA protein




M16762496

kinase inhibitor,






PI3K inhibitor


274
Zacopride
BRD-A65615053
 −90.01
Serotonin receptor






antagonist


275
Etifenin
BRD-K63979671
 −90.00
Compound used






in hepatobiliary






scans of the liver









Classification of host proteins implicated in SARS-CoV-2 infection in four modules. A set of 348 SARS-CoV-2-related host cell proteins composed of 332 proteins identified by Gordon et al., plus 16 reportedly involved in SARS-CoV-2 life cycle, were considered (Gordon D E et al. Nature, 2020, 583, 459-468; de Lartigue J et al. Traffic, 2009, 10, 883-893; Li P et al. Trends Biochem Sci. 2019, 44, 110-124; Hoffmann M et al. Cell, 2020, 181, 271-280; Ou X et al. Nat Commun. 2020, 11, 1620).


The 332 host cell proteins were identified by mass spectrometry upon expressing 26 of 29 SARS-CoV-2 proteins (non-structural proteins Nsp1-16, spike [S], envelop [E], membrane [M], nucleocapsid [N], and nine open reading frames [Orfs]), individually in HEK293T cells (Gordon D E et al. Nature, 2020, 583, 459-468). Comparison of the viral-human interactomes for SARS-CoV-2, SARS-CoV, and MERS-CoV (Gordon D E et al. Science 2020, 370, 1181) revealed that 14.7% of the SARS-CoV-2 host proteins were not among those detected in SARS-CoV-1 or MERSCoV interactomes, underscoring the significance of utilizing the viral/host interactome specific to SARS-CoV-2.


The additional 16 proteins are the receptor ACE2, the proteases transmembrane protease serine 2 (TMPRSS2), cathepsin B, and cathepsin L, as well as several cell signaling and regulation proteins (interleukin 6 [IL6] receptor, myeloid differentiation primary response 88 [MyD88], MAP kinase 1, protein kinase B [AKT1], mammalian target of rapamycin [mTOR], nuclear factor of activated T cells cytoplasmic 1 [NFATC1], nuclear factor κB subunit 1 [NFκB1], STAT3, ADAM metallopeptidase domain 17 [ADAM17], phosphatidylinositol 3-kinase catalytic subunit α [PIK3CA], phosphatidylinositol 3-phosphate 5-kinase [PlKfyve], and the two-pore channel 2 [TPC2]).


In order to better assess the involvement of these 348 host cell proteins in different phases of SARS-CoV-2 infection, they were mapped onto their KEGG pathways (243 pathways) and four functional modules (viral entry, viral replication and translation, host cell regulation and signaling, and immune response) were identified based on their KEGG annotations. This led to 27, 45, 27, and 32 proteins in the respective modules (see Table 7 and Table 8). Several proteins were shared between these modules, such that their union contained 103 host proteins. For example, MAPK and PI3KAKT-mTOR signaling pathways regulate CoV replication and translation (Zumla A et al. Nat Rev Drug Discov. 2016, 15, 327-347), in addition to mediating the immune response (Prompetchara E et al. Asian Pac J Allergy Immunol. 2020, 38, 1-9). Some proteins distinguished in a recent CRISPR screen (Daniloski Z et al. Cell 2021, 184, 1-14), including the Ras-related protein Rab-7A (RAB7A), and subunits of the ATPase vacuolar pump (ATP6AP1 and ATP6V1A) and intracellular cholesterol transporter (NPC2) are also noted in Table 7.









TABLE 7







Four modules mediating host cell response during SARS-CoV-2 infection,


corresponding pathways, and proteins.ª









Module

Gene names of the host cell


(# of
KEGG
proteins involved in


proteins)
pathways
the module





Viral Entry
Endocytosis,
SCARB1; ATP6AP1; AP3B1; NPC2; ITGB1;


(27
lysosome
RAB8A; AP2A2; PIKFYVE; RHOA;


proteins)
pathway
RAB10; ACE2; AP2M1; ATP6V1A;




RNF41; CHMP2A; CTSB; WASHC4;




TMPRSS2; RAB7A; GLA;




SPART; CTSL; PPT1; ARF6; RAB5C;




NEU1; TPC2


Viral
DNA
NUP62; ERLEC1; NUP214;


replication
replication,
EIF4E2; RPL36;


&
RNA
LMAN2; EXOSC3; NUP54; WFS1; PRIM2;


translation
transport,
SRP72; SIL1; UPF1;


(45
RNA
SELENOS; POLA1; NUP88;


proteins)
degradation,
OS9; HYOU1; RAE1; RBX1;



protein
EXOSC2; MRPS2; NUP98; PSMD8;



processing
NGLY1; NUP58; ERO1B; EDEM3;



in ER and
MRPS5; PRIM1; NUP210; ELOC;



protein export
SRP54; ELOB; UGGT2; EXOSC5; IMPDH2;




PABPC4; EXOSC8; POLA2; SRP19; SLU7;




CUL2; MOGS; PABPCI


Regulation
Ras signaling,
IL6R; PIK3CA; RAB8A;


and
autophagy,
RALA; MTOR; TBK1;


signaling
AMPK
AKT1; NFKB1; GNG5; EIF4E2; PRKAR2A;


(27
signaling,
RAB2A; MYD88; ATP6V1A;


proteins)
mTOR
COL6A1; RAB14;



signaling,
MAPK1; RHOA; RAB10;



PI3K-AKT
PRKAR2B; ITGB1;



signaling,
GNB1; ARF6; RAB5C; ECSIT; PRKACA;



and insulin
NFATC1



signaling



Immune
Toll-like
MYD88; MAPK1; STAT3;


response
receptor-,
CUL2; HMOX1;


(32
chemokine-,
ELOB; RIPK1; IL17RA;


proteins)
RIG-like
CSNK2B; MTOR;



receptor-,
INHBE; PRKACA; GNB1;



B cell
NLRX1; ERC1;



receptor-,
RHOA; GDF15; TBK1; IL6R;



NF-kB-,
AKT1; CSNK2A2;



TCR-,
GNG5; NFATC1; TBKBP1; PIK3CA;



and HIF-1-
CTSB; BX1; NFKB1; ELOC;



signaling
REIF4E2; PLAT; ARF6



pathways







aSee Table 8 for the full names of the proteins whose gene codes are listed in column 3. Genes corresponding to some key proteins targeted by the proposed compounds/drug and/or mentioned in the text are written in bold face, including: ARF6 (ADP ribosylation factor 6); ATP6AV1A (ATPase H + transporting V1 subunit A); TBK1 (TANK-binding kinase 1); PRKACA (protein kinase CAMP-activated catalytic subunit α, or the catalytic subunit α of protein kinase A (PKA); RAB7A (Ras-related protein Rab-7A; RHOA (recombinant human RhoA); CTSL and CTSB (cathepsin L and B).














TABLE 8







Composition of four modules mediating host cell response during


SARS-CoV-2 infection.









Module (#
Dominant
Host cell proteins (gene names)


of proteins)
pathways/processes
in the module





Viral entry
Endocytosis,
Scavenger receptor class B


(27
lysosome
member 1 (SCARB1); V-


proteins)
pathway
type proton ATPase subunit




S1 (ATP6AP1); AP-3




complex subunit β-1 (AP3B1);




NPC intracellular




cholesterol transporter 2




(NPC2); integrin β-1




(ITGB1); Ras-related protein




Rab-8A (RAB8A); AP-2




complex subunit a-2 (AP2A2);




1-phosphatidylinositol




3-phosphate 5-kinase




(PIKFYVE); transforming




protein RhoA (RHOA);




Ras-related protein Rab-10




(RAB10); angiotensin-




converting enzyme 2 (ACE2);




AP-2 complex subunit mu




(AP2M1); V-type proton




ATPase catalytic subunit A




(ATP6V1A); E3 ubiquitin-




protein ligase NRDP1




(RNF41); charged




multivesicular body protein




2a (CHMP2A); cathepsin




B (CTSB); WASH complex




subunit 4 (WASHC4);




TMPRSS2; Ras-related protein




Rab-7a (RAB7A); α-




galactosidase A (GLA);




spartin (SPART); cathepsin




L1 (CTSL); palmitoyl-protein




thioesterase 1 (PPT1);




ADP-ribosylation factor 6




(ARF6); Ras-related protein




Rab-5C (RAB5C); sialidase-1




(NEU1); two pore Ca2+




channel protein 2 (TPC2)


Viral
DNA
Nuclear pore glycoprotein


replication
replication,
p62 (NUP62); endoplasmic


and
RNA transport,
reticulum lectin 1 (ERLEC1);


translation
RNA
nuclear pore complex


(45
degradation,
protein Nup214 (NUP214);


proteins)
protein
eukaryotic translation



processing
initiation factor 4E type 2



in ER
(EIF4E2); 60S ribosomal



protein export
protein L36 (RPL36); vesicular




integral-membrane




protein VIP36 (LMAN2);




exosome complex




component RRP40 (EXOSC3);




nucleoporin p54




(NUP54); wolframin (WFS1);




DNA primase large




subunit (PRIM2); signal




recognition particle subunit




SRP72 (SRP72); nucleotide




exchange factor SIL1




(SIL1); regulator of nonsense




transcripts 1 (UPF1);




selenoprotein S (SELENOS);




DNA polymerase α




catalytic subunit (POLA1);




nuclear pore complex




protein Nup88 (NUP88); protein




OS-9 (OS9); hypoxia




up-regulated protein 1




(HYOU1); mRNA export factor




(RAE1); E3 ubiquitin-protein




ligase RBX1 (RBX1);




exosome complex component




RRP4 (EXOSC2);




mitochondrial small ribosomal




subunit protein uS2m




(MRPS2); nuclear pore




complex protein Nup98-Nup96




(NUP98); 26S proteasome




non-ATPase regulatory




subunit 8 (PSMD8); peptide-N




(4)-(N-acetyl-β-




glucosaminyl)asparagine




amidase (NGLY1);




nucleoporin p58 (NUP58);




ERO1-like protein β




(ERO1B); ER degradation-




enhancing α-mannosidase-




like protein 3 (EDEM3);




mitochondrial small




ribosomal subunit protein




uS5m (MRPS5); DNA




primase small subunit




(PRIM1); nuclear pore




membrane glycoprotein




210 (NUP210); elongin-C




(ELOC); signal recognition




particle 54 kDa protein




(SRP54); elongin-B (ELOB); UDP-




glucose:glycoprotein




glucosyltransferase 2 (UGGT2);




exosome complex component




RRP46 (EXOSC5);




inosine-5′-monophosphate




dehydrogenase 2




(IMPDH2); polyadenylate-




binding protein 4




(PABPC4); exosome complex




component RRP43




(EXOSC8); DNA polymerase




α subunit B (POLA2);




signal recognition particle




19 kDa protein (SRP19);




pre-mRNA-splicing factor




SLU7 (SLU7); cullin-2




(CUL2); mannosyl-




oligosaccharide glucosidase




(MOGS); polyadenylate-




binding protein 1 (PABPC


Regulation
Ras signaling,
Interleukin-6 receptor subunit


and
autophagy,
α (IL6R); PIK3CA; Ras-


signaling
AMPK
related protein Rab-8A


(27
signaling,
(RAB8A); Ras-related protein


proteins)
mTOR
Ral-A (RALA); mTOR;



signaling, PI3K-
serine/threonine-protein



AKT
kinase TBK1 (TBK1);



signaling and
AKT1; NFkB1; guanine



insulin signaling
nucleotide-binding protein




G (I)/G (S)/G (O) subunit γ-




5 (GNG5); eukaryotic




translation initiation factor 4E




type 2 (EIF4E2); cAMP-




dependent protein kinase type




II-α regulatory subunit




(PRKAR2A); Ras-related




protein Rab-2A (RAB2A);




MYD88; V-type proton




ATPase catalytic subunit A




(ATP6V1A); collagen α-1




(VI) chain (COL6A1);




Ras-related protein Rab-14




(RAB14); MAPK1; RHOA;




RAB10; cAMP-




dependent protein kinase type




II-β regulatory subunit




(PRKAR2B); integrin




β-1 (ITGB1); guanine




nucleotide-binding protein




G (I)/G (S)/G (T) subunit β-




1 (GNB1); ARF6; RAB5C;




evolutionarily conserved




signaling intermediate in




Toll pathway (ECSIT);




CAMP-dependent protein




kinase catalytic subunit α




(PRKACA); NFATC1


Immune
Interferon-, TLR-,
Myeloid differentiation primary


response
chemokine-,
response protein


(32
NFkB-,
MyD88 (MYD88); MAPK1;


proteins)
RIG-like
STAT3; cullin-2 (CUL2);



receptor-,
heme oxygenase 1 (HMOX1);



B-cell
ELOB; receptor-



receptor-, T
interacting serine/threonine-



cell receptor-
protein kinase 1 (RIPK1);



and
interleukin-17 receptor A



HIF-1-signaling
(IL17RA); casein kinase II




subunit β (CSNK2B); serine/




threonine-protein kinase




mTOR (MTOR); inhibin β




E chain (INHBE); cAMP-




dependent protein kinase




catalytic subunit α




(PRKACA); guanine nucleotide-




binding protein G




(I)/G (S)/G (T) subunit β-1




(GNB1); NLR family




member X1 (NLRX1); ELKS/




Rab6-interacting/CAST




family member 1 (ERC1); RHOA;




growth/differentiation




factor 15 (GDF15);




serine/threonine-protein kinase




TBK1 (TBK1); IL6R;




AKT1; casein kinase II




subunit α (CSNK2A2);




guanine nucleotide-binding




protein G (I)/G (S)/G (O)




subunit γ-5 (GNG5); NFATC1;




TANK-binding kinase




1-binding protein 1 (TBKBP1);




PIK3CA; cathepsin B




(CTSB); E3 ubiquitin-protein




ligase RBX1 (RBX1);




NFkB1; ELOC; eukaryotic




translation initiation factor




4E type 2 (EIF4E2); tissue-type




plasminogen activator




(PLAT); ARF6









Prioritization of candidate compounds proposed to have antiviral effects. As a measure of the potential antiviral effect of the compounds deduced from the computational analysis, the proximity of their targets to each disease module were calculated. Specifically, the distance between the targets of each compound and the proteins belonging to each module were evaluated using the lung-specific PPI network from BioSNAP (Zitnik M et al. BioSNAP datasets: Stanford biomedical network dataset collection. 2018, http://snapstanfordedu/biodata) and network proximity analysis (Guney E et al. Nat Commun. 2016, 7, 10331) (see Materials and Methods). Top-ranking 25 compounds were selected for each module (FIG. 9 and Table 9) leading to a set of 64 distinct compounds in the union of four modules (FIG. 10). FIG. 9-FIG. 10 show the identification and classification of prioritized potentially antiviral compounds. Clustering of these based on their interaction patterns with target proteins (using QuartataWeb) led to 12 clusters (FIG. 11 and Table 10) containing 48 of the compounds; the remaining 16 exhibited unique interaction patterns. Up to two representatives were selected from each cluster and further evaluated (manually) with literature-based evidence including their MOAs, side effects, availability, and antiviral evidence if any, to generate a reduced set of 13 high-priority compounds, listed in Table 11. In addition, after manual evaluation of 95 compounds that lack data in DrugBank and STITCH, two investigational compounds, rottlerin, and QL-XII-47, with respective CMap scores of 97.11 and 99.03, were added to the high-priority list (see FIG. 6).


The final set of 15 compounds that are proposed to have antiviral activities (Table 11) contains eight FDA-approved (repurposable) drugs and seven under investigation. Ten of these have been tested in in vitro assays (indicated by asterisks in Table 11; and labeled in red in FIG. 10). FIG. 13 displays the corresponding chemical structures.









TABLE 9







Top-ranking 64 compounds involved in four


disease modules, rank-ordered by the proximity


of the corresponding targets to the disease modules (*)








Viral entry
Viral replication & translation












In-
Compound

In-
Compound



dex
name
z-score
dex
name
z-score





1
SN-38
−2.97E+00
1
Methimazole
−3.93E+00


2


Hexylresorcinol


−1.87E+00
2
Mefenamic acid
−2.62E+00


3
GSK-1904529A
−1.79E+00
3
Fludarabine
−2.50E+00


4


Linsitinib


−1.79E+00
4
TGX-221
−1.87E+00


5
Sphingosine
−1.48E+00
5
Ibuprofen
−1.50E+00


6


Semaxanib


−1.09E+00
6
Razoxane
−1.38E+00


7
Azathioprine
−1.08E+00
7
Somatostatin
−8.50E−01


8


Imipramine


−8.18E−01
8
NU-7441
−8.39E−01


9
KU-55933
−7.96E−01
9
AS-605240
−7.83E−01


10
Mesoridazine
−7.43E−01
10
Leflunomide
−7.80E−01


11
Alisertib
−7.13E−01
11
NU-7026
−6.66E−01






(LY-293646)



12


Salmeterol


−6.89E−01
12
Aspirin
−6.62E−01


13
Terbutaline
−6.89E−01
13
Tolazoline
−4.83E−01


14
NBI-27914
−6.56E−01
14
Clobenpropit
−4.83E−01


15
Desoxy-
−6.45E−01
15
JNJ-16259685
−4.79E−01



corticosterone






16
GR-127935
−6.45E−01
16
VU-0415374-1
−4.79E−01


17
Terfenadine
−4.98E−01
17
NBI-27914
−4.79E−01


18
Dactolisib
−4.57E−01
18
Ponalrestat
−4.72E−01


19
JNJ-16259685
−4.48E−01
19


Brompheniramine


−2.93E−01


20
VU-0415374-1
−4.48E−01
20
Oxybutynin
−2.44E−01


21
SCH-23390
−4.46E−01
21
Ipratropium
−2.44E−01


22


Ezetimibe


−4.42E−01
22
Procyclidine
−2.44E−01


23


Brompheniramine


−4.39E−01
23
Hyoscyamine
−2.44E−01


24
Desipramine
−4.03E−01
24
Metixene
−2.44E−01


25
Oxybutynin
−3.61E−01
25
Rescinnamine
−2.28E−01











Cell signaling & regulation
Immune response












In-
Compound

In-
Compound



dex
name
z-score
dex
name
z-score





1
PKCβ-inhibitor
−4.10E+00
1
Fostamatinib
−5.16E+00


2
Dactolisib
−3.91E+00
2
NVP-TAE684
−4.47E+00


3
Fostamatinib
−3.48E+00
3
PKCβ-inhibitor
−4.37E+00


4
NVP-TAE684
−3.24E+00
4
Bosutinib
−3.59E+00


5
Wortmannin
−3.19E+00
5
Wortmannin
−3.29E+00


6
PDE-V-
−2.83E+00
6
WHI-P154
−3.04E+00



Inhibitor II


(JAK3-







Inhibitor-II)



7
TPCA-1
−2.56E+00
7
TPCA-1
−3.04E+00


8
STO-609
−2.50E+00
8
Dactolisib
−2.88E+00


9
Bosutinib
−2.39E+00
9
NU-7441
−2.77E+00


10
Benidipine
−2.37E+00
10
PDE-V-
−2.68E+00






Inhibitor II



11
Dipyridamole
−2.22E+00
11
Benidipine
−2.32E+00


12
SCH-23390
−2.00E+00
12
Dipyridamole
−2.06E+00


13
PI-103
−1.92E+00
13


Semaxanib


−2.06E+00


14
WHI-P154
−1.88E+00
14
AS-605240
−1.89E+00



(JAK3-







Inhibitor-II)






15
AS-605240
−1.88E+00
15
KU-0063794
−1.87E+00


16
NU-7441
−1.81E+00
16


Temsirolimus


−1.87E+00


17
Alisertib
−1.80E+00
17
AZD-8055
−1.87E+00


18
KU-0063794
−1.79E+00
18
STO-609
−1.84E+00


19
Temsirolimus
−1.79E+00
19
PI-103
−1.81E+00


20
AZD-8055
−1.79E+00
20
Pimozide
−1.79E+00


21
Torin-1
−1.72E+00
21
SCH-23390
−1.70E+00


22
Semaxanib
−1.66E+00
22
Torin-1
−1.69E+00


23
Reserpine
−1.54E+00
23
Reserpine
−1.65E+00


24
Telmisartan
−1.48E+00
24
Leflunomide
−1.58E+00


25
Terfenadine
−1.36E+00
25
Aspirin
−1.40E+00


18
KU-0063794
−1.79E+00
18
STO-609
−1.84E+00





*Bold and italicized compounds/drugs have been experimentally tested. z-score is a measure of proximity, the lower scores indicating closer proximity. 25 compounds with the lowest proximity score are listed for each module. Several compounds participate in multiple modules resulting in 64 distinct compounds in the listed four modules.













TABLE 10







Grouping of 64 potentially antiviral compounds/drugs into clusters


based on their interaction patterns with their targets (*).











Cluster


PubChem
DrugBank


index
Index
Compound name
ID
ID














1
1


Ipratropium


657309
DB00332



2
Terfenadine
5405
DB00342



3


Brompheniramine


6834
DB00835



4
Metixene
4167
DB00340



5
Oxybutynin
4634
DB01062



6
Procyclidine
4919
DB00387



7
Hyoscyamine
154417
DB00424


2
8
Desipramine
2995
DB01151



9


Imipramine


3696
DB00458



10
Mesoridazine
4078
DB00933



11
SCH-23390
5018
NA



12
Pimozide
16362
DB01100



13
Desoxycorticosterone
6166
NA



14
GR-127935
107780
NA


3
15
AZD-8055
25262965
DB12774



16
KU-0063794
16736978
NA



17


Temsirolimus


6918289
DB06287


4
18
Wortmannin
312145
DB08059



19


Torin-1


49836027
NA



20
Dactolisib
11977753
NA



21
PI-103
9884685
NA



22


AS-605240


5289247
DB04769



23
NU-7441
11327430
NA



24
TGX-221
9907093
NA


5
25
GSK-1904529A
25124816
NA



26
Linsitinib
11640390
NA


6
27


Salmeterol


5152
DB00938



28


Terbutaline


5403
DB00871


7
29
STO-609
3467590
NA



30
Bosutinib
5328940
DB06616



31
Fostamatinib
11671467
DB12010



32
NVP-TAE684
16038120
NA



33
PKCβ-inhibitor
6419755
NA



34
WHI-P154 (JAK3-Inhibitor-II)
3795
NA



35


Semaxanib


5329098
NA



36
TPCA-1
9903786
NA


8
37
Tolazoline
5504
DB00797



38
Clobenpropit
2790
NA


9
39
NU-7026 (LY-293646)
9860529
NA


10
40
SN-38
104842
DB05482



41


Hexylresorcinol


3610
DB11254


11
42
Ponalrestat
5278
NA



43
Aspirin
2244
DB00945



44


Mefenamic acid


4044
DB00784



45
Ibuprofen
3672
DB01050


12
46
Alisertib
24771867
DB05220


13
47


JNJ-16259685


11313361
NA



48
VU-0415374-1
46869940
NA


14
49
PDE-V-Inhibitor II
9844109
NA



50
Dipyridamole
3108
DB00975


15
51
Telmisartan
65999
DB00966


16
52
Leflunomide
3899
DB01097


17
53
Methimazole
1349907
DB00763


18
54
Sphingosine
5280335
DB03203


19
55
Somatostatin
16129681
DB09099


20
56
Razoxane
30623
NA


21
57
Fludarabine
657237
DB01073


22
58
Reserpine
5770
DB00206


23
59
Benidipine
656668
DB09231


24
60


Ezetimibe


150311
DB00973


25
61
NBI-27914
176157
NA


26
62
KU-55933
5278396
NA


27
63
Azathioprine
2265
DB00993


28
64
Rescinnamine
5280954
DB01180





*13 compounds bolded and italicized are prioritized after the analysis













TABLE 11





High-priority compounds with potential antiviral effects based on Dataset 1 (*).







Prioritized compounds based on Cmap scores and Network Proximity Ranks












Drug/

Disease

Description/



compoundª
Status
module
Rankb
MOAs
Ref.





Brompheniramine*
FDA-approved
Viral entry
23
Histamine
Gwaltney Jr JM et






receptor
al. Clin Infect Dis.






antagonist
1997, 25, 1188-







1194


Ipratropium
FDA-approved
Viral
21
Acetylcholine
Barnes PJ. Am J




replication

receptor
Med. 2004,




and

antagonist
117(Suppl 12A),




translation


24S-32S


Imipramine*
FDA-approved
Viral entry
8
Norepinephrine
Shchors K et al.






and serotonin
Cancer Cell, 2015,






reuptake
28, 456-471);






inhibitor,
Wichit S et al. Sci






autophagy
Rep. 2017, 7, 3145;






enhancer
Plenge P et al. Nat







Commun. 2020, 11,







1491


Temsirolimus*
FDA-approved
Immune
16
mTOR
Di Benedetto F et al.




response

inhibitor,
Transplantation




Regulation
19
autophagy
2010, 89, 733-738;




and

enhancer
Soliman A et al. Exp




signaling


Clin Transplant,







2013, 11, 408-411;







Bergmann L et al.







Expert Rev







Anticancer Ther.







2014, 14, 9-21;







Kindrachuk J et al.







Antimicrob Agents







Chemother. 2015,







59, 1088-1099


Torin-1*
Investigational
Immune
22
mTOR
Clippinger AJ et al. J




response

inhibitor, PI3K
Virol. 2011, 85,




Regulation
21
inhibitor,
3930-3939);




and

autophagy
Bergmann L et al.




signaling

enhancer
Expert Rev







Anticancer Ther.







2014, 14, 9-21


AS-605240
Investigational
Regulation
15
PI3K inhibitor,
Azzi J et al. Diabetes




and

autophagy
2012, 61, 1509-




signaling

enhancer
1518




Viral
9






replication







and







translation







Immune
14






response





Linsitinib*
Investigational
Viral entry
4
IGF-1-and
Mulvihill MJ et al.






insulin receptor
Future Med Chem.






inhibitor, TBK1
2009, 1, 1153-






activator
1171; Sparrer KMJ






through ARF1
et al. Nat Microbiol.







2017, 2, 1543-1557


Salmeterol*
FDA-approved
Viral entry
12
β2 Adrenergic
Medigeshi GR et al.






receptor
Antimicrob Agents






agonist,
Chemother. 2016,






autophagy
60, 6709-6718






enhancer



Semaxanib*
Investigational
Viral entry
6
VEGFR
O'Donnell A et al.




Regulation
22
inhibitor
Br J Cancer, 2005,




and







signaling







Immune
13

93, 876-883




response





Hexylresorcinol*
FDA-approved
Viral entry
2
Local anesthetic
Wilson CO et al.







Textbook of organic







medicinal and







pharmaceutical







chemistry, 1966, 5th







edn. Philadelphia,







PA: Lippincott


Mefenamic acid
FDA-approved
Viral repl
2
Cyclooxygenase
Rothan HA et al.




and

inhibitor
Antiviral Res. 2016,




translation


127,50-56


JNJ16259685
Investigational
Viral entry
19
Glutamate
Lavreysen H et al.






receptor
Neuropharmacology,






antagonist
2004, 47, 961-972


Ezetimibe*
FDA-approved
Viral entry
22
Niemann-Pick
Osuna-Ramos JF et




Regulation
9
C1-like 1
al. Antiviral Res.




and

protein
2018, 160, 151-164




signaling

antagonist,







cholesterol







inhibitor,







autophagy







enhancer










Additional prioritized compounds (based on Cmap scores and literature)










Drug/compoundª
Status
Description/MOAs
Ref.





QL-XII-47
Investigational
Cytoplasmic tyrosine protein
de Wispelaere M et




kinase BMX inhibitor
al. J Biol Chem.





2020, 295, 1694-





1703


Rottlerin*
Investigational
MAPK and protein kinase
Lama Z et al.




inhibitor, autophagy enhancer
Antiviral Res.





2019, 168, 51-60





ªThose tested in experiments are indicated by asterisks in the first column.



bRank refers to the proximity to the module in the third column, the lower the better.







Prioritization of candidate compounds proposed to have anti-inflammatory effects. A similar interaction pattern-based clustering of the 163 compounds predicted to potentially have anti-cytokine effect (among the high Cmap-scoring 275; see Table 6) led to 20 clusters of two or more compounds based on compound-protein interaction patterns, while 35 compounds were left as singletons (FIG. 12 and Table 12). Nineteen high-priority compounds representative of these clusters in addition to 5 singletons were selected. Furthermore, literature search of the remaining 112 potentially anti-inflammatory compounds for which no target data were available in DrugBank and STITCH, led to three additional candidate compounds. The resulting set of 27 potentially anti-inflammatory/cytokine compounds is presented in Table 13.


Table 13 contains 15 FDA-approved drugs and 12 compounds under investigation. Of note, two of the compounds under investigation (JAK3-Inhibitor-II and AZD-8055; in boldface) also belong to the 64 top-ranking compounds based on Dataset 1; and one, mepacrine/quinacrine, is listed in the Excelra COVID-19 drug repurposing database (https://wwwexcelracom/covid-19-drug-repurposing-database/). Another investigational drug in the list, PCA4248, is a platelet-activating factor (PAF) receptor antagonist (Fernandez-Gallardo S et al. J Pharmacol Exp Ther. 1990, 255, 34-39), and its utility against COVID-19 (e.g., for preventing coagulation or blood clots) is to be explored, as well as those of the two His receptor antagonists azelastine and chlorphenamine, identified here. Recent study draws attention to the possible repurposing of PAF receptor antagonists and His receptor antagonists against hyperinflammation and microthromboses in COVID-19 patients (Demopoulos C et al. BioFactors 2020, 46, 927-933).


Among approved drugs, pirfenidone is known to inhibit furin (Burghardt I et al. Biochem Biophys Res Commun. 2007, 354, 542-547), a human protease involved in the cleavage of the viral spike glycoprotein into S1 and S2 subunits (like TMPRSS2). Spike cleavage is essential to activate the S1 fusion trimer for viral entry. Pirfenidone combined with melatonin has been pointed out to be a promising therapy for reducing cytokine storm in COVID-19 patients (Artigas L et al. PloS One 2020, 15, e0240149). Finally, Table 13 also contains two approved cyclooxygenase inhibitors, oxaprozin and dexketoprofen, known as non-steroidal anti-inflammatory drugs (NSAIDs) (Miller L G. Clin Pharm. 1992, 11, 591-603; Moore R A et al. Clin Pharm. 2008, 8, 11).









TABLE 12







Grouping of 163 potential modulators of hyperinflammatory response


into clusters based on their interaction patterns with their targets (*).











Cluster


PubChem
DrugBank


index
Index
Compound name
ID
ID














1
1
Chlorprothixene
667467
DB01239



2


Olanzapine


4585
DB00334



3
Amoxapine
2170
DB00543



4
Desipramine
2995
DB01151



5


Trifluoperazine


5566
DB00831



6
Maprotiline
4011
DB00934



7
Promazine
4926
DB00420



8
Ipsapirone
56971
NA



9
Trazodone
5533
DB00656



10
Nefazodone
4449
DB01149



11
Zuclopenthixol
5311507
DB01624



12
Cisapride
2769
DB00604



13


Fluphenazine


3372
DB00623



14
Lisuride
28864
DB00589



15
Bromocriptine
31101
DB01200



16
Piribedil
4850
DB12478



17
Thioproperazine
9429
DB01622



18
Quinpirole
54562
NA



19
Pirenperone
4847
NA



20
Ketanserin
3822
DB12465



21
Latrepirdine
197033
DB11725



22
Cyclazosin
132266
NA



23


Midodrine


4195
DB00211



24
Nor-binaltorphimine
5480230
NA



25
EMD-386088
10131112
NA



26
Dopamine
681
DB00988



27


Azelastine


2267
DB00972



28
Loratadine
3957
DB00455



29
Verapamil
2520
DB00661



30
Naftopidil
4418
DB12092



31
Clonidine
2803
DB00575



32
Alfuzosin
2092
DB00346



33
Clebopride
2780
DB13511



34
U-99194
5626
NA


2
35
Mepyramine
4992
DB06691



36


Chlorphenamine


2725
DB01114



37
Dicycloverine
3042
DB00804



38
Oxybutynin
4634
DB01062



39
Otenzepad
107867
NA



40
Tolterodine
443879
DB01036



41
Profenamine
3290
DB00392



42
Xaliproden
NA
DB06393



43
Alverine
3678
DB01616



44
Bupropion
444
DB01156



45
Indatraline
126280
NA



46
Sertraline
68617
DB01104


3
47
Alprenolol
2119
DB00866



48
ICI-89406
123686
NA



49
Bisoprolol
2405
DB00612



50
Carteolol
2583
DB00521


4
51


Clarithromycin


84029
DB01211



52
Bepridil
2351
DB01244


5
53
Fipronil
3352
NA



54
CGS-20625
163844
NA



55
Topiramate
5284627
DB00273



56
Chlordiazepoxide
2712
DB00475


6
57


Saracatinib


10302451
DB11805



58
HY-11007
5311510
NA



59
Dasatinib
3062316
DB01254



60
Fostamatinib
11671467
DB12010



61
TG-101348 (Fedratinib)
16722836
DB12500



62
PLX-4720
24180719
DB06999



63
TPCA-1
9903786
NA



64
DMBI
5353593
NA



65
Orantinib
5329099
DB12072



66
D-64406
5330535
NA



67
WZ-4002
44607530
NA



68
bis-tyrphostin
5329255
NA



69


JAK3-Inhibitor-II


3795
NA


7
70
TGX-221
9907093
NA



71
AZD-6482
44137675
DB14980



72
GDC-0941 (Pictilisib)
17755052
DB11663



73
PI-103
9884685
NA



74


AZD-8055


25262965
DB12774


8
75
Roscovitine (Seliciclib)
160355
DB06195



76


CGP-60474


644215
NA


9
77
Milrinone
4197
DB00235



78
Anagrelide
2182
DB00261


10
79


Hexamethylene


1794
NA



80
FIT
84008
NA



81


Loperamide


3955
DB00836



82
BRL-52537
6603740
NA


11
83
Nicotine
89594
DB00184



84
Zacopride
108182
NA



85
Metoclopramide
4168
DB01233



86
m-chlorophenylbiguanide
1354
NA



87
Palonosetron
6337614
DB00377



88
Alosetron
2099
DB00969


12
89
Flutamide
3397
DB00499



90
Danazol
28417
DB01406



91
Norgestimate
6540478
DB00957



92
Oxybenzone
4632
DB01428



93
α-estradiol
68570
NA



94
Diethylstilbestrol
448537
DB00255



95
Estradiol
5757
DB00783



96
Testosterone
6013
DB00624



97
Ticlopidine
5472
DB00208



98


Nifedipine


4485
DB01115



99
Nimodipine
4497
DB00393



100
Spironolactone
5833
DB00421



101


Liothyronine


5920
DB00279



102
CITCO
9600409
NA


13
103
Piperine
638024
DB12582



104
Phenelzine
3675
DB00780



105
Iproniazid
3748
DB04818



106
Selegiline
26757
DB01037



107
Auraptene
1550607
NA


14
108


Atorvastatin


60823
DB01076



109
Pravastatin
54687
DB00175


15
110
Exemestane
60198
DB00990



111
Formestane
11273
DB08905


16
112
Felbamate
3331
DB00949



113
Cycloserine
6234
DB00260



114
Gavestinel
6450546
DB06741



115
RO-25-6981
6604887
NA


17
116
EHNA
3206
NA



117
SCH-442416
10668061
NA


18
118
Bromfenac
60726
DB00963



119
DUP-697
3177
NA



120


Dexketoprofen


667550
DB09214



121


Oxaprozin


4614
DB00991



122
Phenothiazine
7108
DB11447



123


Triptolide


107985
DB12025



124
Pyrazinamide
1046
DB00339


19
125
SCH-28080
108137
NA



126
Pantoprazole
4679
DB00213


20
127
BIIB021
16736529
DB12359



128
Geldanamycin
5288382
DB02424


21
129
STO-609
3467590
NA


22
130
Mafenide
3998
DB06795


23
131
AR-C133057XX
9797857
DB07002


24
132
Buphenine (Nylidrin)
4567
DB06152


25
133
AICA-ribonucleotide
65110
DB01700


26
134
Valproic acid
3121
DB00313


27
135
Iodophenpropit
3035746
NA


28
136
Retinol (Vitamin A)
445354
DB00162


29
137
Sildenafil
5212
DB00203


30
138
MK-2206
24964624
NA


31
139
KI-16425
10367662
NA


32
140
Dinoprostone
5280360
DB00917


33
141
BI-78D3
2747117
NA


34
142
Y-27632
448042
DB08756


35
143
SB-216763
176158
NA


36
144
Linsitinib
11640390
NA


37
145
PF-04217903
17754438
DB12848


38
146


Mepacrine


237
DB01103


39
147
Dicyclohexylurea
4277
NA


40
148


Pirfenidone


40632
DB04951


41
149
Dichloroacetic acid
6597
DB08809


42
150
MLN-4924 (Pevonedistat)
16720766
DB11759


43
151
Thenoyltrifluoroacetone
5601
DB04795


44
152
Navitoclax
24978538
DB12340


45
153
Daunorubicin
30323
DB00694


46
154
UNC-0321
46901937
NA


47
155


Isoliquiritigenin


638278
DB03285


48
156


PCA-4248


4698
NA


49
157
Pyroxamide
4996
DB12847


50
158


Rucaparib


9931954
DB12332


51
159
Ilomastat
132519
DB02255


52
160
Latrunculin-b
6436219
DB08080


53
161
Nicorandil
47528
DB09220


54
162
Z-prolyl-prolinal
122623
DB03535


55
163
BRD-K63784565
97226
DB12385





* The 24 compounds bolded and italicized are prioritized based on the cluster analysis













TABLE 13







Compounds proposed to help attenuate hyperinflammation based on Dataset 2.










Drug/

Description/



Compound
Status
MOAs
Ref.










Compounds extracted from Cmap and prioritized after QuartataWeb cluster analysis










Midodrine
FDA-approved
Adrenergic receptor
Josset L et al. PLOS One, 2010,




agonist
5, e13169


Olanzapine

Dopamine receptor
Altschuler EL et al. Med




antagonist,
Hypotheses 2020, 141, 109774




autophagy enhancer



Trifluoperazine

Dopamine receptor
Ochiai H et al. Antiviral Res.




antagonist,
1991, 15, 149-160




autophagy dual-





modulator



Fluphenazine

Dopamine receptor
Otreba M et al. Eur J




antagonist,
Pharmacol. 2020, 887, 173553




autophagy enhancer



Azelastine

Dopamine receptor
Konrat R et al. bioRxiv, 2020,




antagonist, His
https://doi.org/10.1101/




receptor antagonist
2020.09.15.296228


Chlorphenamine

Histamine receptor
Xu W et al. Front Microbiol.




antagonist
2018, 9, 2643


Clarithromycin

Bacterial 50S
Yamaya M et al. Eur Respir J.




ribosomal subunit
2012, 40, P4364; Pani A et al.




inhibitor autophagy
Int J Antimicrob Agents, 2020,




inhibitor
56, 106053


Saracatinib
Investigational
SRC inhibitor
Shin JS et al. Viruses, 2018, 10,





283



JAK3-Inhibitor-II


JAK inhibitor
Schwartz DM et al. Nat Rev





Drug Discov. 2017, 17, 843-





862



AZD-8055


mTOR inhibitor,
Jiang Q et al. Cancer Res.




autophagy enhancer
2011, 71, 4074-4084


CGP-60474

CDK inhibitor
He B et al. F1000Research





2020, 9, 609



Mepacrine/


Cytokine
Dermawan JKT et al. Mol



Quinacrine


production
Cancer Ther. 2014, 14, 2203-




inhibitor, NFκB
2214




inhibitor



Hexamethylene

Sodium/hydrogen
Wilson L et al. Virology, 2006,




antiport inhibitor
353, 294-306


Loperamide
FDA-approved
Opioid receptor
Shen L et al. J Virol. 2019, 93,




agonist, autophagy
e00023-e119




enhancer



Nifedipine

Calcium channel
Liu W et al. Life Sci. 2009, 85,




blocker, autophagy
235 -240; Straus MR et al.




enhancer
bioRxiv, 2020,





https://doi.org/10.1101/





2020.07.21.214577


Liothyronine

Thyroid hormone
CN103705497B




stimulant



Atorvastatin

HMGCR inhibitor,
Episcopio D et al. FASEB J.




autophagy enhancer
2019, 33, 9516-9525


Triptolide
Investigational
RNA polymerase
Chaparala S et al. Preprints,




inhibitor, TNF-α
2020, https://doi.org/10.20944/




inhibitor
preprints202009.0459.v1


Pirfenidone
FDA-approved
TGFβ receptor
Ferrara F et al. Eur J Clin




inhibitor, furin
Pharmacol. 2020, 76, 1615-




inhibitor, anti-
1618




fibrotic, autophagy





enhancer



Oxaprozin

Cyclooxygenase
Miller LG. Clin Pharm. 1992,




inhibitor, NSAID
11,591-603




(non-steroidal anti-





inflammatory drug)



Dexketoprofen

Cyclooxygenase
Moore RA et al. Clin Pharm.




inhibitor, NSAID
2008, 8, 11


Isoliquiritigenin
Investigational
Guanylate cyclase
Traboulsi H et al. Antimicrob




activator,
Agents Chemother. 2015, 59,




autophagy enhancer
6317-6327


PCA-4248

Platelet activating
Fernandez-Gallardo S et al. J




factor (PAF)
Pharmacol Exp Ther. 1990,




receptor antagonist
255, 34-39


Rucaparib
FDA-approved
PARP inhibitor,
Guo T et al. Nat Microbiol.




autophagy enhancer
2019, 4, 1872-1884







Compounds extracted from Cmap and prioritized by literature search










Berbamine
Investigational
Calmodulin
Huang L et al. Res Sq. 2020,




antagonist,
https://doi.org/10.21203/




autophagy inhibitor
rs.3.rs-30922/v1


Darinaparsin

Apoptosis stimulant
Chowdhury T et al. Infect





Disord Drug Targets 2020, 21,





608-618


Taurodeoxycholic

Bile acid
Li N et al. Sci Bull (Beijing),


acid


2019, 64: 180-188





The three drugs/compounds in boldface are also predicted as antiviral drugs based on Dataset 1, listed in Table 11.






Testing the SARS-CoV-2 inhibitory properties of prioritized compounds in in vitro assays. First, five compounds (salmeterol, rottlerin, temsirolimus, torin-1, and ezetimibe) were selected from the list of 15 prioritized compounds described in Table 11 for a proof of concept in vitro evaluation of their anti-SARS-CoV-2 potential. FIG. 14-FIG. 17 show the suppression of SARS-CoV-2 infection by identified compounds.


A SARS-CoV-2 infectious cell culture system (FIG. 14 and FIG. 15) where host Vero-E6 cells were pretreated with compounds (salmeterol, rottlerin (R077), temsirolimus, torin-1, or ezetimibe) for 1 h prior to SARS-CoV-2 inoculation was used. After 48-h post-infection, cells were fixed and fluorescently labeled for SARS-CoV-2 S protein and immunofluorescence was performed to assess viral infection (SARS-CoV-2 S protein; FIG. 14 and FIG. 15). Images were analyzed for spike-positive cells using the Multiwavelength Cell Scoring algorithm in MetaXpress. Representative mock and vehicle control images and their segmentation are shown in FIG. 14. Violin plots describing the distribution of the log integrated spike for each cell in the untreated and treated samples are shown in FIG. 16 along with complementary pie charts indicating the percent of cells positive for spike protein (FIG. 17). In the untreated controls, a bimodal distribution of spike-positive cells was evident, indicating the presence of two infected cell populations with one expressing more spike protein per cell than the other (FIG. 16). Salmeterol at 0.1 and 1 μM reduced the median of the spike-expressing population and showed a preferential antiviral effect for the lower spike-expressing subpopulation (FIG. 16). At 10 μM, salmeterol exhibited a greater antiviral effect on the entire population, although some (˜14%) spike-positive cells were evident (FIG. 16). Qualitatively similar results to salmeterol were obtained with rottlerin and the mTOR inhibitors, Temsirolimus, and Torin-1, although dose-limiting toxicity as evidenced by reduced cell count prevented a determination of a more complete antiviral effect on the higher-spike protein-expressing subpopulation in torin-1- and R077-treated cells (FIG. 16). Ezetimibe reduced spike protein-expressing populations only at the highest concentration studied (25 μM), where a reduction in cell numbers was also observed.


Next, cell fusion assays were used as a proxy for ACE2/SARS-CoV-2-mediated viral entry. Prioritized compounds predicted to potentially act as viral entry blockers, i.e., imipramine, brompheniramine, linsitinib, semaxanib, and hexylresorcinol, in addition to salmeterol and ezetimibe from the above set were focused on (see Table 11). The cell fusion assay, first described by Simmons et al. (Simmons G et al. Proc Natl Acad Sci USA, 2004, 101, 4240-4245), detects host-cell-spike interactions on a shorter time scale than the viral infection assay and has been used by several groups to investigate the mechanisms of cell entry of SARS-CoV-1, such as endosomal and protease involvement including TMPRSS2 (Matsuyama S et al. Proc Natl Acad Sci USA 2005, 102, 12543-12547; Matsuyama S et al. J Virol. 2010, 84, 12658-12664). More recently, the assay has also been used to investigate SARS-CoV-2-mediated cell entry (Ou X et al. Nat Commun. 2020, 11, 1620). The assay is based on the principle that susceptible host cells (“acceptors”) fuse with spike-expressing “donor” cells, forming large cell fusion constructs (syncytia), which can be quantified by fluorescence imaging.


This assay was implemented in a high-content, 384-well microplate format using HEK293T cells, which are not susceptible to viral infection unless transfected with ACE2 and TMPRSS2, and Calu-3 lung cancer cells, which possess the replete machinery for spike-mediated viral infection (Hoffmann M et al. Cell, 2020, 181, 271-280). HEK293T cells transfected with ACE2 and TMPRSS2 or native Calu-3 cells were incubated with donor cells co-expressing green fluorescent protein (GFP) and SARS-CoV-2 spike, and syncytia formation monitored by following GFP over time by fluorescence microscopy. After a 4-h incubation, syncytia were quantified by high-content analysis. Cell fusion was dependent on the presence of SARS-CoV-2 spike as donor cells expressing only GFP did not form syncytia.


Quantification of syncytia formation in HEK293 cells is shown in FIG. 24-FIG. 31 (related to FIG. 18-FIG. 23). HEK293 acceptor cells transfected with or without ACE2 and TMPRSS2 were seeded in 384 well plates, pretreated with 7-point gradients of test compounds for 1-2 h, and co-cultured for 4 hours with HEK293 donor cells expressing SARS-CoV-2 spike and GFP, or donor cells expressing GFP only (no spike). Images of GFP-positive objects were acquired on a confocal high-content imager and analyzed for syncytia formation and total GFP as a measure of cytotoxicity, using a CNT algorithm as described in the Methods Section. Representative images illustrating syncytia phenotype and compound activity in HEK293 cells are shown in FIG. 32-FIG. 42. Images are shown at the 100 μM condition except nafamostat (5.5 μM), semaxanib (50 μM), and linsitinib (25 μM). Upper panels, raw fluorescence micrographs; lower panels, images with CNT overlay. GFP positive objects that met the criteria for syncytia are colored purple; cellular aggregates that are not syncytia are shown in yellow.


Quantification of syncytia formation in Calu-3 cells is shown in FIG. 43-FIG. 50. Calu-3 acceptor cells were seeded in 384 well plates, pretreated with compounds for 1-2 h, and co-cultured for 4 hours with HEK293 donor cells expressing SARS-CoV-2 spike and GFP. Images of GFP-positive objects were acquired on a confocal high-content imager and analyzed for syncytia formation using a CNT algorithm as described in the Methods. Representative images illustrating syncytia phenotype and compound activity in Calu-3 cells are shown in FIG. 51-FIG. 61. Images are shown at the 100 μM condition except nafamostat (5.5 μM), semaxanib (50 μM), and linsitinib (25 μM). Upper panels, raw fluorescence micrographs; lower panels, images with CNT overlay. GFP positive objects that met the criteria for syncytia are colored purple; cellular aggregates that are not syncytia are shown in yellow.


In a preliminary screen of seven computationally predicted compounds and two serine protease inhibitor positive controls (dec-RVKR-CMK and nafamostat), pretreatment with dec-RVKR-CMK and nafamostat prevented syncytia formation (FIG. 24-FIG. 42 and FIG. 43-FIG. 61), consistent with the involvement of those enzymes in spike-mediated viral entry (Ozden S et al. J Blot Chem. 2008, 283, 21899-21908; Matsuyama S et al. J Virol. 2018, 92, e00683-18; Hoffmann M et al. Cell, 2020, 181, 271-280). Notably, nafamostat, a potent wide spectrum serine protease inhibitor, has recently been found to inhibit the membrane fusion of SARS-CoV-2 at 15-fold higher efficiency than camostat mesylate (Hoffmann M et al. Antimicrob Agents Chemother. 2020, 64, e00754-00720). Dec-RVKR-CMK inhibits not only the enzymatic activity of furin but also those of cathepsin L, cathepsin B, trypsin, papain, and TMPRSS2 (Matsuyama S et al. J Virol. 2018, 92, e00683-18). With the exception of semaxanib, all predicted compounds/drugs inhibited cell fusion to some extent, although some did so only at high concentrations (FIG. 24-FIG. 42 and FIG. 43-FIG. 61).


Both agents that prevented viral infection in the experiments with Vero-E6 cells (salmeterol and ezetimibe), also had inhibitory activity in the cell fusion assay, although salmeterol was at least two orders of magnitude less potent in the cell fusion assay, and ezetimibe was inactive at the highest concentration tested in the viral infection assay, suggesting that their antiviral activity might not originate from an interference in viral entry, but other effects such as enhancement of autophagy, as discussed below. The most potent agent was the insulin-like growth factor 1 receptor (IGF1R) inhibitor, linsitinib. Inhibitor effects were qualitatively conserved in Calu-3 cells but generally more pronounced in transfected HEK293T cells (FIG. 24-FIG. 42 and FIG. 43-FIG. 61). The one exception was the furin inhibitor dec-RVKR-CMK, which was similarly potent in both cell types but with a seemingly larger maximal magnitude of inhibition in Calu-3 cells, suggesting it inhibited other cellular pathways in addition to viral entry.


Then, full dose-response curves were performed in HEK293 cells with selected compounds (linsitinib, brompheniramine, hexylresorcinol, and salmeterol), together with cytotoxicity assessments to test whether inhibition of syncytia formation could merely be a result of cell loss.


HEK293 acceptor cells transfected with or without ACE2 and TMPRSS2 were seeded in 384-well plates, pretreated with 7-point gradients of compounds for 1-2 h, and co-cultured for 4 h with HEK293 donor cells expressing SARS-CoV-2 spike and GFP, or donor cells expressing GFP only (no spike). Images of GFP-positive objects were acquired on a confocal high-content imager and analyzed for syncytia formation and integrated GFP area (total GFP) as a measure of cytotoxicity, using a CNT algorithm as described in the Materials and Methods.


Nafamostat, dec-RVKR-CMK, linsitinib, and to a lesser extent, brompheniramine, showed dose-responsive inhibition of syncytia formation that did not mirror cell loss (FIG. 18-FIG. 23). For example, linsitinib induced complete inhibition of cell fusion, whereas only partial cell loss was observed with a flattening of its dose-response curve. This quantitative and qualitative difference between the two dose-response curves suggests that the observed cell loss is likely to be an epiphenomenon, and not causing the inhibition of syncytia formation. In contrast, hexylresorcinol and salmeterol showed partial and full responses, respectively, on syncytia formation that were mirrored by cell loss (FIG. 18-FIG. 23). Further studies are required to determine in this assay with these particular drugs if cell loss (i) precedes inhibition of cell fusion thereby representing a nonspecific mechanism for preventing syncytia formation or (ii) is a specific result of inhibition of syncytia formation.


Discussion

Utility of the computational pipeline for identifying repurposable drugs. Presented herein are the results from a computation-driven approach for identifying repurposable drugs or new compounds that comply with the antiviral or anti-cytokine signatures derived from SARSCoV-2-infected cells. The overall analysis was driven by the RNAseq data from SARS-CoV-2-infected A549 cells and A549-ACE2 cells, as well as a SARS-CoV-2-host PPI network, toward gaining a system-level understanding of the key players in the host cell that are involved in SARS-CoV-2 infection and identifying potential modulators of these key players. The extensive study led to 15 potentially antiviral and 23 potentially immune-modulatory compounds (Table 11 and Table 13). The assays conducted to test ten of the proposed antiviral compounds pointed to several repurposable drugs or investigational compounds that could be pursued for lead development against SARS-CoV-2 infection. Among them, salmeterol exhibited particularly strong inhibitory activities in Vero-E6 cells infected by SARS-CoV-2 and linsitinib substantially reduced spike-protein-dependent syncytia formation (viral entry) in engineered HEK293T cells.


Recent studies point to the utility of computational systems pharmacology approaches for identifying repurposable drugs against SARS-CoV-2 (Beck B R et al. Comput Struct Biotechnol J. 2020, 18, 784-790; Gordon D E et al. Nature, 2020, 583, 459-468; Riva L et al. Nature, 2020, 586, 113-119; Singh T U et al. Pharmacol Rep. 2020, 72, 1479-1508; Zhou Y et al. Cell Discov. 2020, 6, 14; Zhou Y et al. PLoS Biol. 2020, 18, e3000970; Zhou Y et al. Lancet Digit Health, 2020, 2, e667-676). Of note is the work of Zhou et al., where repurposable drugs against SARS-CoV-2 were identified by evaluating the proximity of targets of known drugs to human proteins engaged in the human-CoV-host cell interactome (Zhou Y et al. Cell Discov. 2020, 6, 14). This type of network proximity analysis, originally introduced by Guney et al. (Guney E et al. Nat Commun. 2016, 7, 10331), is also used here, but in a different context, mainly for prioritizing the candidate compounds/drugs that have been already identified from the DEG patterns of SARS-CoV-2 infected cells and corresponding Cmap signatures. In contrast, Zhou et al. used gene set enrichment data (from MERS-CoV and SARS-CoV-infected cells) and Cmap gene-drug signatures for validating their predicted drugs (Zhou Y et al. Cell Discov. 2020, 6, 14). Another important component unique to this analysis is the use of the interface QuartataWeb that allows for identifying drug-target associations, and for evaluating and classifying the pathways implicated in the disease modules deduced from the SARS-CoV-2-specific virus-host interactome (Gordon D E et al. Science 2020, 370, 1181; Gordon D E et al. Nature, 2020, 583, 459-468) and assessing the mechanisms of action. QuartataWeb was further used to cluster the selected compounds based on their mechanisms of action and select representatives from each cluster to obtain a sufficiently diverse set for experimental testing. Thus, this study differs from that of Zhou et al. (Zhou Y et al. Cell Discov. 2020, 6, 14) in the overall design of the computational protocol, the types of data used as input, as well as the output analyses for compound selection, prioritization, and validation, while both studies utilize state-of-the-art methods (network proximity analysis) and resources (e.g., Cmap library) at different steps of the workflow.


Unlike influenza A and respiratory syncytial virus, the host immune defensive reactions of SARS-CoV-2 were significantly muted unless ACE2 was overexpressed (Blanco-Melo D et al. bioRxiv, 2020, 10.1101/2020.03.24.004655). FIG. 62-FIG. 65 show a comparison of the behavior of A546 and A546-ACE2 cells vis-à-vis the expression levels of the genes that have been adopted for defining antiviral and anticytokine signatures. Cross-examination of the expression levels of the 17 anticytokine signature genes in A549 cells showed that most of these genes could not be clearly distinguished in those cells, i.e., their upregulation was specific to A549-ACE2 cells (compare FIG. 64 and FIG. 65), whereas the 36 genes that define the antiviral signature exhibited a comparable expression pattern in A549-ACE2 cells (see FIG. 62 and FIG. 63). These observations support the robustness of the antiviral signature on the one hand, and the utility of A549-ACE2 cells for detecting genes implicated in hyperinflammatory responses, on the other.


Potential mechanisms of action of drug candidates. Two types of in vitro assays were performed with ten predicted repurposable or investigational drug candidates most of which are proposed to be implicated in viral entry: linsitinib, imipramine, ezetimibe, hexylresorcinol, brompheniramine, salmeterol, semaxanib, rottlerin, temsirolimus, and torin-1. Viral entry is used here in a broad sense including (i) the fusion between viral and host cell membrane (involving ACE2 and B0 AT1 on the host cell membrane, and facilitated by host cell proteases such as TMPRSS2 and furin) and (ii) endosomal processes mediating the endocytosis of the virus and its release from the vesicles. The latter involves many signaling and regulatory proteins including those activated by the immune response, in addition to proteases such as cathepsins, as schematically depicted in FIG. 66. The two experimental assays were chosen to complement each other: the viral infection assay recapitulates the entire virus infection process, whereas the syncytia assay addresses a specific, defined mechanism in viral entry, namely fusion of the virus with the host cell, which is mediated by interaction of viral spike protein with the host cell receptor (ACE2), and facilitated by host cell proteases.


Below the experimental results for the tested compounds are discussed in the light of their CMap scores, the similarities between their interaction patterns (as indicated by the clusters in FIG. 7-FIG. 8), the involvement of their targets in the host cell PPI network or disease modules (FIG. 9-FIG. 10) with reference to lung-tissue interactome (FIG. 67), and relevant findings from previous work. The discussion begins with compounds/drugs implicated in viral entry, as the focus of current tests (FIG. 68).


Linsitinib. Linsitinib showed the highest inhibitory activity without overt cytotoxicity in the spike-induced syncytia formation assay that specifically measures viral entry. It is interesting to note that its proximity rank to the viral entry module (rank 4) was one of the highest among all tested compounds. Linsitinib is an IGF-1R and insulin receptor inhibitor (Mulvihill M J et al. Future Med Chem. 2009, 1, 1153-1171) currently under investigation for various types of cancer due to its ability to prevent tumor cell proliferation and induce tumor cell apoptosis (Fassnacht M et al. Lancet Oncol. 2015, 16, 426-435). The analysis also indicated that it targets the insulin receptor, which interacts with ADP ribosylation factor 6 (ARF6), a binding partner of SARS-CoV-2 endonuclease nsp15 (Gordon D E et al. Nature, 2020, 583, 459-468). As listed in Table 7, ARF is involved in multiple modules. Notably, the ubiquitination of the ARF domain of TRIM23 is essential for mediating virus-induced autophagy, an antiviral defense mechanism, via activation of TANK-binding kinase 1 (TBK1) (Sparrer K M J et al. Nat Microbiol. 2017, 2, 1543-1557). Therefore, it is proposed that its possible MOA is activation of TBK1 that promotes autophagy (see FIG. 66). While linsitinib was selected as a potential antiviral compound, it was also identified as an anti-inflammatory compound with a very high (−99.37) CMap score (Table 6), in strong support of its selection as a high priority compound. In this context, the EC50 for linsitinib was 25 μM in the cell fusion assay that may not be disparate from the reported Cmax of 5-10 μM in patients (Macaulay V M et al. Clin Cancer Res. 2016, 22, 2897-2907). Since several IGF1/InsR inhibitors are available, this class of compounds is well suited for structure-activity studies. Such a study is particularly relevant, since CMap can implicitly account for structure-dependent non-canonical modes of antiviral activity that can differ among members of a particular drug class.


Imipramine. Imipramine, an FDA-approved tricyclic antidepressant (Gillman P K. Br J Pharmacol. 2007, 151, 737-748), has been also reported to inhibit Chikungunya virus fusion (entry) (Wichit S et al. Sci Rep. 2017, 7, 3145). It was distinguished by a high network proximity ranking (8th) in viral entry module (Table 11). Notably, imipramine is a high-affinity allosteric inhibitor of serotonin transporter (SLC6A4) (Plenge P et al. Nat Commun. 2020, 11, 1491). Importantly, ACE2 is anchored into the host membrane through close association with the amino acid transporter, B0 AT1 (see FIG. 66). B0 AT1 is structurally homologous to serotonin transporter, sharing the LeuT fold typical of this family of sodium-coupled neurotransmitter transporters (Cheng M H et al. Nat Struct Mot Biol. 2019, 26, 545-556). Thus, imipramine is likely to also target B0 AT1, which may impair the ACE2-spike interaction, hence the observed inhibitory effect. In addition, imipramine has been reported to promote autophagy (Shchors K et al. Cancer Cell, 2015, 28, 456-471), and this could be another (indirect) mechanism for alleviating SARS-CoV-2 infection.


Brompheniramine. Brompheniramine is an FDA-approved drug known as a first-generation antihistamine drug, for treating common colds and allergic rhinitis (Simons F E et al. J Allergy Clin Immunol. 1982, 70, 458-464). It shares a similar mode of action with imipramine, also targeting serotonin transporter. In this study, brompheniramine was indicated to be highly related to SARS-CoV-2 entry (ranked 23rd in the viral entry module). Both imipramine and brompheniramine inhibited syncytia formation, consistent with their hypothesized interaction with membrane-anchored ACE2.


Salmeterol. Salmeterol had the highest CMap score for inducing the antiviral signature, and very high (network) proximity to the viral entry module. It is canonically used as a bronchial smooth muscle relaxant in asthma and COPD, as a long-acting 02-adrenergic receptor ((32-AR) agonist. COPD has been shown to be associated with increased expression of ACE2 (Leung J M et al. Eur Respir J. 2020, 55, 2000688), and a recent study on the effects of inhaled corticosteroids (ICS) on the bronchial epithelial cell expression of SARS-CoV-2-related genes in COPD patients demonstrated that a treatment with ICS in combination with salmeterol/fluticasone propionate decreased the expression of ACE2 and ADAM17 (Milne S et al. medRxiv, 2020, https://doi.org/10.1101/2020.08.19.20178368). It is also noted that β2-AR interacts with the PKA catalytic subunit α (Cα; encoded by PRKACA), which promotes autophagy-mediated degradation (Lizaso A et al. Autophagy, 2013, 9, 1228-1243). Salmeterol has been reported to induce autophagy as a potential mechanism of inhibiting Dengue virus in vitro (Medigeshi G R et al. Antimicrob Agents Chemother. 2016, 60, 6709-6718). The observed inhibitory effect in Vero-E6 cells (FIG. 14-FIG. 17), which were not borne out by syncytia formation experiments with either HEK293T or Calu-3 cells, except at high concentration (FIG. 18-FIG. 23), is consistent with activities unrelated to viral entry, such as an innate immune response stimulation or autophagy enhancement.


Ezetimibe. Ezetimibe, an FDA-approved lipid-lowering drug (Kosoglou T et al. Clin Pharmacokinet. 2005, 44, 467-494), has a distinct MOA via the sterol transporter Niemann-Pick C1-Like 1 (Nutescu E A et al. Pharmacotherapy, 2003, 23, 1463-1474). It targets sterol O-acyltransferase 1 (SOAT1) in the ER, which, in turn, interacts with the Ras proteins encoded by RAB5C, RAB2A, and RAB7A, implicated in early-to-late endosomal maturation. These proteins bind SARS-CoV-2 nsp7 (Gordon D E et al. Nature, 2020, 583, 459-468). Loss of RAB7A (see FIG. 66 and FIG. 67) has been shown to reduce viral entry by altering endosomal trafficking and sequestering ACE2 inside cells (Daniloski Z et al. Cell 2021, 184, 1-14). Finally, ezetimibe was also reported to interfere with the entry and replication of Dengue virus (Osuna-Ramos J F et al. Antiviral Res. 2018, 160, 151-164). Herein, ezetimibe inhibited both viral infection and cell fusion. Its lower potency in the cell fusion assay is consistent with multiple mechanisms in addition to the dominant effect on viral entry, as described above.


Hexylresorcinol. Hexylresorcinol ranked 2nd in the viral entry module. It is a FDA-approved over-the-counter product with anesthetic, antiseptic, and anthelmintic properties often used for upper respiratory irritations such as sore throat (Wilson C O et al. Textbook of organic medicinal and pharmaceutical chemistry, 1966, 5th edn. Philadelphia, PA: Lippincott). It has sodium channel blocking effects and interacts with transglutaminase 2, a substrate of two SARS-CoV-2-related host proteins RhoA and PKA Cα. It also showed potential action against respiratory virus parainfluenza type 3 and cytomegalovirus (Shephard A et al. Antiviral Res. 2015, 123, 158-162). Yet, the in vitro cell fusion assay herein suggests that virus-host cell interactions may not be major contributors to its reported antiviral activities.


Rottlerin. Rottlerin (R077), a natural polyphenolic compound, has been reported to inhibit influenza replication as an inhibitor of PKC (Hoffmann H H et al. Antiviral Res. 2008, 80, 124-134), and the translation of rabies virus circle by reducing intracellular ATP contents (Lama Z et al. Antiviral Res. 2019, 168, 51-60). It may have neuroprotective effects by its anti-oxidative and anti-inflammatory action in the central nervous system (Lee T H et al. J Neuroinflammation 2020, 17, 177). Rottlerin inhibited viral infection but dose-limiting toxicity prevented a detailed analysis of viral entry vs. infection.


Temsirolimus and torin-1. Temsirolimus and torin-1 are indicated to inhibit the protein kinase mTOR (Bergmann L et al. Expert Rev Anticancer Ther. 2014, 14, 9-21). The temsirolimus metabolite, sirolimus, as well as mTOR inhibitor rapamycin, are among the 128 approved drugs listed in the Excelra COVID-19 Drug Repurposing Database (https://wwwexcelracom/covid-19-drug-repurposing-database/). The PI3K-AKT-mTOR signaling pathway provides a cross-protective immunity against viral infection, especially against the influenza viruses (Lehrer S. World Acad Sci J. 2020, 2, 1), and has been recognized to regulate the translation and replication of coronaviruses (Zumla A et al. Nat Rev Drug Discov. 2016, 15, 327-347). mTOR inhibitors induce autophagy, which has been attributed to the inhibition of MERS-CoV (Gassen N C et al. Nat Commun. 2019, 10, 5770). Temsirolimus is currently FDA-approved for treating renal cell carcinoma (Miao H et al. J Virol. 2010, 84, 6687-6698). It has been reported to inhibit MERS-CoV infection (Kindrachuk J et al. Antimicrob Agents Chemother. 2015, 59, 1088-1099). Torin-1 inhibits both mTORC1/2 complexes with IC50 values between 2 and 10 nM and therefore was used at 1-10 and 100 nM levels and was toxic at 100 nM. Further studies will be required to determine the relative antiviral effects of these mTOR inhibitors in the context of their intrinsic dose-limiting toxicity.


Semaxanib. Semaxanib a tyrosine kinase inhibitor, under development as a cancer therapeutic (O'Donnell A et al. Br J Cancer, 2005, 93, 876-883), did not exhibit any inhibitory activity, despite its involvement in multiple modules.


Compounds targeting immune response. Immunopathology of COVID-19 is longitudinally dynamic, individually diverse, more unique than other respiratory viral infections, and potentially detrimental when uncontrolled. It features lack of interferon response, lymphopenia, and overwhelming inflammatory activation-especially in severe stages or patients with poor prognosis (Blanco-Melo D et al. Cell, 2020, 181, 1036-1045.e1039; Liu J et al. EBioMedicine, 2020, 55, 102763; Ong E Z et al. Cell Host Microbe 2020, 27, 879-882; Zhou F et al. Lancet, 2020, 395, 1054-1062). Anti-cytokine therapeutics inhibiting IL-1 (NCT04324021, NCT0436281), IL-6 (NCT04320615, NCT04315298), TNF-α (Feldmann M et al. Lancet. 2020, 395, 1407-1409), or the broad-spectrum immune response by glucocorticoids (Lu S et al. Ann Transl Med. 2020, 8, 627) are currently investigated. Stemming from transcriptomic response following infection in A549-ACE2, inducers that both elevate IFN signaling while suppressing cytokine pathways were the aim. The resulting compounds (Table 13), interestingly, included His receptor antagonists and TNFα inhibitors as expected, while also containing candidates such as PAF receptor antagonists, NFκB, SRC, JAK, and mTOR inhibitors, and neurological drugs blocking ion channels or neurotransmitter receptors. These results reveal the complexity of immune transcriptome modulation, involving heterogeneous states of multiple components and their coupled dynamics.


Autophagy enhancement as a possible mechanism to exploit in combination therapies. The present analysis showed that certain autophagy-related vesicle pathways were downregulated, especially in the SARS-CoV-2-infected A549-ACE2 cells, which could be a potential escape mechanism from the immune system, as lysosomal digestion serves as an intrinsic antiviral program. These observations point to the opportunity of discovering drugs that exploit systems-level host response, i.e., stimulate autophagic response while suppressing hyperinflammatory responses. A recurrent pattern in several candidate compounds was indeed their involvement in autophagy enhancement. These include antidepressants as well as compounds repurposed to eliminate aggregates in the central nervous system, lung, or liver, such as trifluoperazine, fluphenazine (Table 13), and others (salmeterol and imipramine) that exhibited inhibitory activity in these experiments. Microglial autophagy has been recently pointed out to be essential for recovery from neuroinflammation (Berglund R et al. Sci Immunol. 2020, 5, eabb5077). In general, the role of autophagy in viral infection remains context-dependent, and both pathogen-destroying or viral-promoting effects have been reported (Maier H J et al. Viruses, 2012, 4, 3440-3451), whereas inducing autophagy has markedly reduced MERS-CoV replication (Gassen N C et al. Nat Commun. 2019, 10, 5770). The effectiveness of selected autophagy enhancers observed here support their further investigation, at least in combination therapy, against COVID-19.


Summary and Conclusions. The compounds prioritized here targeted system-level modules, rather than individual targets. Beyond the urgent need for repurposing, these drugs can also be exploited as mechanistic probes to enhance understanding of SARS-CoV-2 pathogenicity and drug resistance and provide a systems framework for developing combination therapies.


Comparison with earlier work showed that there are only nine compounds (apicidin, daunorubicin, entacapone, loratadine, metformin, mycophenolic acid, ribavirin, verapamil, and valproic acid) shared between the predictions herein and the recently reported 69 repurposable drugs (Gordon D E et al. Nature, 2020, 583, 459-468). Given the little overlap with the drugs currently under clinical trials against SARS-CoV-2, the current findings may help complement the global COVID-19 drug discovery pipeline.


While a systems-level approach was adopted herein, it should also be noted that the viral-host cell interactions that mediate viral entry and endosomal transitions, and on accompanying cell signaling and regulation events and immune response, were focused on, in line with the assays conducted for probing viral entry. Events at the nucleus relevant to viral replication and translation play an equally important role, as evidenced by recent genome-wide CRISPR screens in Vero-E6 cells (Wei J et al. Cell, 2021, 184, 1-16), which identified many proviral genes involved in chromatin regulation, histone modification, or epigenetic regulation. Compounds that target these specific pathways/processes, such as those involving the ubiquitous nuclear protein HMGB1 and the SWI/SNF chromatin remodeling complex (Wei J et al. Cell, 2021, 184, 1-16) or the upregulation of cholesterol biosynthesis (Daniloski Z et al. Cell 2021, 184, 1-14), are yet to be determined.


Materials and Methods


Evaluation of host-targeted antiviral and anti-hyperinflammatory signature from post-SARS-CoV-2 infection transcriptomics. The up- and downregulated gene list of A549 cells (human lung cancer) after 24 h of SARS-CoV-2 infection was obtained from GSE147507, and the corresponding DEGs were acquired from the DESeq2 result from the original publication with FDR adjusted P-value smaller than 0.05. This resulted in 100 upregulated and 20 downregulated genes listed in Table 1. Overrepresentation analysis was performed using gProfiler (Raudvere U et al. Nucleic Acids Res. 2019, 47, W191-W198) with GO database (Carbon S et al. Nucleic Acids Res. 2019, 47, D330-D338) for up- or downregulated genes, respectively, using Benjamini-Hochberg multiple test correction with a threshold of 0.05. Examination of the GO Biological Process (GO-BP) and GO cellular components (GO-CC) data for up- or downregulated genes resulted in 319 GO-BP and 13 GO-CC terms. The number of enriched upregulated terms was reduced by retaining those associated with no more than 300 genes, and not fewer than 10 overlapping genes, resulting in 16 GO terms (see column 6 in Table 3). Downregulated terms were all kept. The enriched GO terms were organized and visualized with quickGO and classified as antiviral, proviral, or ambiguous. Those genes that defined the “antiviral signature” were obtained by merging the up- (innate immune response) or down-(intracellular vesicle) regulated antiviral genes and excluding proviral (viral genome replication) components. Genes classified as proviral or ambiguous were not included in the antiviral signature.


The resulting signature (composed of 36 genes) was used to screen for compounds/drugs in the L1000 database (Subramanian A et al. Cell, 2017, 171, 1437-1452.e1417) which elicit a response that best matches the antiviral signature, reflected by their sufficiently high Cmap connectivity scores, at https://clue.io/query. CMap scores range from −100 to 100, the two limits representing the least and the most similar compound-induced gene signatures, compared to the input antiviral signature. Compounds with top scores (in the suggested default range of 90-100) were selected for further analysis.


For the construction of anti-hyperinflammation signature, cytokine-related events (to be suppressed) were focused on by overlapping the GO cytokine response gene set (GO:0034097) with the upregulated genes (adjusted P-value <0.05) from A549-ACE2-infected cells with high MOI of SARS-CoV-2 (GSE147507). A final candidate set of 17 genes at the 0.05 upper quantile of log2 fold change were selected (see Table 4). This set of 17 genes was used as the upregulated gene input in Cmap screening within the L1000 database, and the 275 compounds with lowest connectivity scores (varying from −90 to −100), showing strongest opposing effect, were selected.


Identification of known compound-target interactions. The compound-target interaction search engine QuartataWeb (Li H et al. Bioinformatics, 2020, 36, 3935-3937), which integrates STITCH (version 5) (Szklarczyk D et al. Nucleic Acids Res. 2016, 44, D380-384) and DrugBank (version 5.1.7) (Wishart D S et al. Nucleic Acids Res. 2018, 46, D1074-D1082), was used to identify targets for compounds obtained from Cmap prediction. Specifically, all compound-target interactions recorded in DrugBank and the compound-target interactions with experimental confidence score no <0.4 in STITCH were integrated for further analysis. As a result, 1,800 known interactions between 168 compounds and 746 targets were retrieved, while no targets were identified for the remaining 95 compounds.


Prioritizing the predicted compounds using their network proximity. The basic idea of network proximity (Guney E et al. Nat Commun. 2016, 7, 10331) is to evaluate the significance of the network distance between a compound and a given disease module in the interactome. The methodology assumes that a compound is effective if it targets proteins within or in the immediate vicinity of a disease module. In this case, the human lung protein-protein interactome was extracted from the Biomedical Network Dataset Collection BioSNAP (Zitnik M et al. BioSNAP datasets: Stanford biomedical network dataset collection. 2018, http://snapstanfordedu/biodata). Five viral-related modules were defined, each containing a set (S) of pre-defined proteins derived from the host proteins implicated in SARS-CoV-2 infection (see the Results). For each compound, the set (T) of targets were determined using QuartataWeb in the human lung PPI network. The proteins in sets S and T were connected via paths of zero or more intermediate protein nodes. Then the distance between these targets and the pre-defined proteins from each viral-related module were evaluated, in the human lung PPI network, as the average shortest distance path between the respective nodes s and t belonging to the sets S and T, as:







d

(

S
,
T

)

=


1


T








t

T





min

s

S




d

(

s
,
t

)








Then, a reference distance distribution was constructed, corresponding to the expected distance between the disease module proteins and a randomly selected groups of proteins in the network, with the same size and degree of distribution as drug targets in the network. This procedure was repeated 1,000 times, and the mean and standard deviation of the reference distance distribution were used to calculate a z-score by converting the observed distance to a normalized distance. Each compound was assigned a z-score with respect to each disease module, a lower z-score meaning that its targets were closer to the disease module, or the compound would be more effective. The z-scores were evaluated using the toolbox package developed by Guney et al. (Guney E et al. Nat Commun. 2016, 7, 10331). Note that the network proximity provides a relative measure, the absolute value of which depends on the disease and application. In the current application to four disease modules, a uniform cutoff for the z-score was not selected. Instead, the top 25 compounds from each module were selected to include a set of compounds with diverse MOAs.


Compound clustering by means of interaction-pattern-based similarities. Top-ranking compounds were clustered by evaluating the similarities between the interaction patterns of these compounds vis-à-vis their known targets compiled in DrugBank and STITCH. Specifically, each compound i was assigned a vector ui, the elements of which were the confidence score for the compound-target interaction (0 if there is no known interaction). Then, the interaction-pattern-based similarities between compound i and j were evaluated by calculating cosine distance between vector ui and vector uj using the similarity metric s=1−(ui·uj)/(|ui||uj|).


In vitro viral inhibition assays. SARS-CoV-2 viral assays were performed in UCLA BSL3 high containment facility. Vero-E6 [VERO C1008 (ATCC #CRL-1586™)] cells were obtained from ATCC and cultured at 37° C. with 5% CO2 in EMEM growth media with 10% fetal bovine serum and 100 units/ml penicillin. SARS-CoV-2 Isolate USA-WA1/2020 was obtained from BEI Resources of National Institute of Allergy and Infectious Diseases (NIAID). Temsirolimus (CAS 162635-04-3), Ezetimibe (CAS 163222-33-1), Salmeterol (CAS 89365-50-4), and Torin-1 (CAS 1222998-36-8) were purchased from Selleckchem. Rottlerin (CAS 82-08-6) was purchased from TOCRIS. Vero-E6 cells were plated in 96-well plates (5×103 cells/well) and pretreated with compounds (in triplicate, at indicated concentrations) for 1 h prior to addition of SARS-CoV-2 (MOI 0.1). After 48-h post-infection (hpi) the cells were fixed with methanol for 30-60 min in −20° C. Cells were washed three times with PBS and permeabilized using blocking buffer (0.3% Triton X-100, 2% BSA, 5% Goat Serum, 5% Donkey Serum in 1×PBS) for 1 h at room temperature.


Subsequently, cells were incubated with anti-SARS-CoV-2 Spike antibody (Sino Biological, 40150-R007, 1:200) at 4° C. overnight. Cells were then washed three times with PBS and incubated with Goat anti-mouse IgG Secondary Antibody, Alexa Fluor 555 (Fisher Scientific PIA32790, 1:1,000) for 1 h at room temperature. Nuclei were stained with DAPI (40,6-Diamidino-2-Phenylindole, Dihydrochloride; Life Technologies) at a dilution of 1:5,000 in PBS for 10 min. Cells were analyzed by fluorescence microscopy. Five images per well were quantified for each condition. The Multiwavelength Cell Scoring module in MetaXpress (Molecular Devices, Sunnyvale, CA) was used to measure the total integrated fluorescence spike signal in each cell. Histograms of the log of the integrated intensities were plotted in Spotfire (Tibco, Palo Alto, CA). A cutoff value of three standard deviations of the total integrated signal from the mock samples was established, above which cells were considered to have a positive spike signal, and thus be infected. The number of infected cells was divided by the total number of cells in each treatment group to determine the percent of infected cells after treatment.


Cell Fusion (Syncytia) Assay


Cell culture. HEK293T cells (ATCC CRL-3216) were maintained at 37° C. in a humidified incubator with a 5% CO2 atmosphere. Cells were cultured in Dulbecco's modified Eagle medium (DMEM, Gibco 11965092) supplemented with 10% fetal bovine serum (FBS, Corning 35010CV), 1% penicillin-streptomycin (Cytiva HyClone SV30010), and 1% L-glutamine (Cytiva HyClone SH3003401). A cell bank of defined passage was established, and cells were propagated for no more than 15 passages in culture. A cell bank of Calu-3 cells (ATCC HTB-55) from cells maintained in DMEM as recommended by ATCC was established at early passage. Because Calu-3 cells grew very slowly in DMEM, for experiments cells were switched to Roswell Park Memorial Institute (RPMI) 1640 (Cytiva HyClone SH30027.01), which provided much better growth conditions. All cell lines were routinely tested for mycoplasma infection and passaged no more than 10 times from ATCC authenticated stocks.


Reagents. Expression plasmids for human ACE2, TMPRSS2, and HA-tagged SARS-CoV-2 spike were a gift from Stefan Pohlmann (Hoffmann M et al. Cell, 2020, 181, 271-280). Dec-RVKR-CMK (furin inhibitor-1) was from EMD Millipore (344930). Imipramine hydrochloride, Salmeterol, and Brompheniramine were from AK Scientific (J10511, K-590, and M-1266, respectively). Hexylresorcinol, Semaxanib (SU-5416), Ezetimibe, and Linsitinib (OSI-906) were from TargetMol (T0314, T2064, T1593, and T6017, respectively).


Transfection of cells for syncytia assay. On the day of experiments, acceptor cells were transfected with mammalian expression plasmids for ACE2 and TMPRSS2 using FuGene6 (Roche) at a 1:3 DNA-to-reagent ratio with 22 ng DNA per well (30 μl) of a 384-well plate. 4,000 cells were plated in collagen-coated microplates (Greiner 781956) and centrifuged at 500 g for 1 min. Donor cells were transfected under the same conditions with expression plasmids for eGFP or eGFP plus SARS-CoV-2 spike protein and plated in T-25 flasks (3 ml). Both donor and acceptor cells were incubated for 3 days at 37° C. Calu-3 cells were left untransfected and seeding density was 8,000 cells/well in RPMI.


Cell treatment for High Content Screening. On the day of co-culture, acceptor cells were pretreated for 1-2 h with vehicle or test agents; compounds were dissolved in DMSO and diluted into complete DMEM to a 3× concentration of the highest desired concentration in the assay. The resulting solutions were serially diluted on a 96-well plate into DMEM containing 3% DMSO. Fifteen microliter of the resulting gradients were transferred to cells using a Biomek 2000 liquid handler (Beckman Coulter) in duplicate to yield quadruplicate measurements for each concentration of test agents. The final concentration of DMSO in the assay was 1%. Each plate contained 80 wells of vehicle controls, 16 wells of mock-transfected acceptor cells, and 16 wells of ACE2/TMPRSS2 transfected acceptor cells incubated with GFP-only expressing donor cells (no spike).


Syncytia assay co-culture, imaging, and analysis. Donor cells were dislodged from their flasks with non-enzymatic cell dissociation buffer (Thermo Fisher 13151014) after two gentle washes with PBS. GFP-positive cells were counted in a hemocytometer. 2,000 GFP-positive cells in 15 μl DMEM were added to acceptor cells, plates centrifuged at 500 g for 1 min, and syncytia formation monitored. After 4 h cells were imaged live in the GFP channel (Ex485/Em525 nm) on a Molecular Devices ImageXpress Ultra or a Perkin Elmer OPERA Phenix Plus High Content Screening (HCS) reader using a 20× objective. Four fields were acquired per well. Images were uploaded to Definiens Developer (Ver 6, Definiens AG, Germany) and analyzed by a custom Cognition Network Technology (CNT) ruleset that separated individual cells, cell aggregates, and syncytia based on size, intensity, and texture of GFP expressing objects. The final parameters used for plotting were the percentage of GFP-positive area covered by syncytia relative to the total area covered by GFP-positive objects, and the total GFP-positive area as a surrogate for cell number. Data were averaged from the four imaging fields and normalized to vehicle-treated controls. Data from multiple independent experiments were pooled and analyzed by one-way ANOVA followed by Dunnett's multiple comparisons test. Dose-response data were fitted to a four-parameter logistic equation in GraphPad Prism (Ver. 7).


Data availability. The data and codes generated during the study are available at: https://github.com/Hannah-Qingya/Covid19_systems-level_analysis. The QuartataWeb server that is online accessible at http://quartata.csb.pitt.edu/ was also used.


Example 2—Approach for the Discovery of Repurposed Drugs and Compounds for Treatment Against SARS-CoV-2 Infection

Disclosed herein are strategies for repurposing existing drugs and identifying new compounds for the treatment of SARS-CoV-2 infection


Summary: Covid-19 (Coronavirus Disease-2019) caused by SARS-CoV-2 (Severe Acute Respiratory Syndrome Coronavirus (CoV) type 2 virus) has led to over 1.3 million deaths as of mid-November 2020, due to its high contagiousness (basic reproductive number (R0) of 2.0-2.5) and therefore rapid spread, compared to the SARS-CoV (R0=1.7-1.9) or the Middle East respiratory syndrome (MERS) (R0=0.7) of the same coronavirus subfamily; and mortality rates in the ranges 4-28%, 3.6-30% and 60-65% have been reported for the three respective CoVs. A large number of vaccine- and drug-candidates under preclinical or clinical trials (191 vaccine candidates and 319 drug candidates) have been reported as of September 2020; with two vaccine trials, Pfizer/BioNTech and Moderna, now showing 95% success rate. Yet, no drugs have been FDA-approved to date, apart from remdesivir, an RNA-dependent RNA polymerase (RdRP) inhibitor that inhibits SARS-CoV and MERS-CoV, as a repurposable drug. Current practices such as the use of corticosteroids, such as dexamethasone, or intravenous immunoglobulin (IVIG) are supportive (for alleviating or preventing hyperinflammatory complications) rather than therapeutic according to the Centers for Disease Control (CDC) guidelines. There is an urgent need to develop new therapeutics against Covid-19.


While efforts to target viral proteins are underway, an alternative strategy is to pursue host-targeted therapies. The host cell response is essential to enabling viral entry, endosomal escape, translation, replication, assembly, and release. Host cells are also naturally armed with antiviral programs, which, if properly induced, can constrain the in vivo viral spread within a canonical 4-7 day period, upon sufficient adaptive immunity development. Herein, the focus was on the identification of compounds that modulate host cell responses, using a comprehensive, mechanism unbiased, and highly integrated systems-level approach. An important component of this type of study is knowledge of networks of protein-protein interactions (PPIs) in the host cell, or disease modules that play a role at various stages of viral infection.


Using transcriptome data from SARS-CoV-2-infected A549 (human adenocarcinomic alveolar basal epithelial) cells from lung tissue, and A549 cells overexpressing the host cell receptor angiotensin-converting enzyme 2 (ACE2), CMap analysis was employed to predict targets involved with SARS-CoV-2 infection, and the QuartataWeb server, developed for compound-target-pathway mapping, was utilized to predict drugs/compounds that would modulate those targets. This is a Quantitative Systems Pharmacology (QSP) approach that considers systems-level effects originating from the promiscuity of drugs and/or the pleiotropy of targets. Rather than a limited drug repurposing effort that merely preserves target class across indications, the focus herein was on a comprehensive unbiased virus-infected cell phenotype (manifested as a DEG signature) that reflects emergent virally driven cellular networks and connects these to drugs that can be repurposed without the need for molecular information.


Using this approach, 38 priority candidate compounds were identified (including repurposable and investigational drugs) that target the host system comprised of 15 compounds with potentially antiviral actions (Table 14) and 23 with possible anti-hyperinflammatory (adjuvant) actions (Table 15). Ten belonging to the first group, including six that are FDA-approved (imipramine, salmeterol, hexylresorcinol, brompheniraamine, ezetimibe, and temsirolimus) and four that are under development (linsitinib, torin-1, rottlerin, semaxanib), have been selected for in vitro assays with different types of cell lines (Vero-E6 cells, HEK293T cells and Calu-3 lung cancer cells). Several of these drug/compounds inhibited SARS-CoV-2 infection in a dose-dependent manner with salmeterol and linsitinib being particularly effective. These findings expand the repertoire of drugs/compounds that could be repurposed/developed for possible Covid-19 treatment as either single drugs or drug combinations.


Overall, a QSP workflow for predicting drugs and compounds that interact cell host proteins involved in viral infection and immune response to viral infection based on transcriptomic profiles was conceived and established (FIG. 1). The workflow was applied and drugs/compounds that interact with cell host proteins involved in SARS-CoV-2 infection (Table 14 and Table 16) and that would elicit anti-cytokine activity to protect from SARS-CoV-2 hyperinflammation were predicted therefrom (Table 15 and Table 17). A subset of the predicted drugs/compounds were experimentally tested and drugs/compounds that inhibit SARS-CoV-2 entry into cells were identified (FIG. 17 and FIG. 24-FIG. 31).


The approach established here is a combination of computational and systems biology analyses for the identification of cellular targets and mechanism associated with viral infection and the identification of compounds and repurposable drugs to modulate infection. The focus on host targets enables a new series of previously unrecognized targets with the potential for inhibition of SARS-CoV-2 infections. This approach can be more efficient than traditional screening approaches for identifying drugs/compounds to move forward into the clinical testing. Compounds were identified herein that were previously unknown to have potential for the inhibition of SARS-CoV-2 infection. Combinations of the predicted drugs may increase the efficacy.









TABLE 14







Prioritized potential anti-viral compounds and repurposable drugs
















Viral



Drug/Compound

Disease
Description/
Entry
Syncytia


Name
Status
module
MOAs
Assay
Assay





Brompheniramine
FDA-approved
Viral entry
Histamine
Not
Active





receptor
Tested






antagonist




Ezetimibe
FDA-approved
Viral entry,
Niemann-Pick
Active
Active




Regulation
C1-like 1 protein






and signaling
antagonist,







cholesterol







inhibitor,







autophagy







enhancer




Hexylresorcinol
FDA-approved
Viral entry
Local anesthetic
Not
Active






Teste



Imipramine
FDA-approved
Viral entry
Norepinephrine
Not
Active





and serotonin
Tested






reuptake







inhibitor,







autophagy







enhancer




Ipratropium
FDA-approved
Viral
Acetylcholine
Not
Not




replication
receptor
Tested
Tested




and
antagonist






translation





Mefenamic acid
FDA-approved
Viral repl and
Cyclooxygenase
Not
Not




translation
inhibitor
Tested
Tested


Salmeterol
FDA-approved
Viral entry
Adrenergic
Active
Active





receptor agonist,







autophagy







enhancer




Temsirolimus
FDA-approved
Immune
mTOR inhibitor,
Active
Not




response,
autophagy

Tested




Regulation
enhancer






and signaling





AS-605240
Investigational
Viral
PI3K inhibitor,
Not
Not




replication
autophagy
Tested
Tested




and
enhancer






translation,







Regulation







and signaling,







Immune







response





JNJ16259685
Investigational
Viral entry
Glutamate
Not
Not





receptor
Tested
Tested





antagonist




Linsitinib
Investigational
Viral entry
IGF-1-and
Not
Active





insulin receptor
Tested






inhibitor, TBK1







activator through







ARF1




QL-XII-47
Investigational
XXX
Cytoplasmic
Not
Not





tyrosine protein
Tested
Tested





kinase BMX







inhibitor




Rottlerin
Investigational
XXX
MAPK and
Active
Not





protein kinase

Tested





inhibitor,







autophagy







enhancer




Semaxanib
Investigational
Viral entry,
VEGFR inhibitor
Not
Active




Regulation

Tested





and signaling,







Immune







response





Torin-1
Investigational
Immune
mTOR inhibitor,
Active
Not




response,
PI3K inhibitor,

Tested




Regulation
autophagy






and
enhancer






signaling
















TABLE 15







Prioritized potential anti-hyperimmune compounds and repurposable drugs.









Drug/Compound




Name
Status
Description/MOAs





Atorvastatin
FDA-approved
HMGCR inhibitor, autophagy enhancer


Azelastine
FDA-approved
Dopamine receptor antagonist


Chlorphenamine
FDA-approved
Histamine receptor antagonist


Clarithromycin
FDA-approved
Bacterial 50S ribosomal subunit inhibitor,




autophagy inhibitor


Fluphenazine
FDA-approved
Dopamine receptor antagonist, autophagy enhancer


Liothyronine
FDA-approved
Thyroid hormone stimulant


Loperamide
FDA-approved
Opioid receptor agonist, autophagy enhancer


Midodrine
FDA-approved
Adrenergic receptor agonist


Nifedipine
FDA-approved
Calcium channel blocker, autophagy enhancer


Olanzapine
FDA-approved
Dopamine receptor antagonist, autophagy enhancer


Pirfenidone
FDA-approved
TGFβ receptor inhibitor, anti-fibrotic, autophagy




enhancer


Rucaparib
FDA-approved
PARP inhibitor, autophagy enhancer


Trifluoperazine
FDA-approved
Dopamine receptor antagonist, autophagy dual-




modulator


AZD-8055
Investigational
mTOR inhibitor, autophagy enhancer


Berbamine
Investigational
Calmodulin antagonist, autophagy inhibitor


CGP-60474
Investigational
CDK inhibitor


Darinaparsin
Investigational
Apoptosis stimulant


Hexamethylene
Investigational
Sodium/hydrogen antiport inhibitor


Isoliquiritigenin
Investigational
Guanylate cyclase activator, autophagy enhancer


JAK3-Inhibitor-II
Investigational
JAK inhibitor


Saracatinib
Investigational
SRC inhibitor


Taurodeoxycholic
Investigational
Bile acid


acid




Triptolide
Investigational
RNA polymerase inhibitor
















TABLE 16







Complete list of drugs and compounds with potential antiviral activity against SARS-


CoV-2-infection.









Drug/Compound




Name
CMap ID
Mechanism of Action





2-aminopurine
BRD-K35128472
Serine/threonine kinase inhibitor


5-nonyloxytryptamine
BRD-K08219523
Serotonin receptor agonist


Abiraterone
BRD-K55301415
17,20 lyase inhibitor, Androgen biosynthesis




inhibitor, Cytochrome P450 inhibitor, Steroid




sulfatase inhibitor


AG-879
BRD-K59469039
Angiogenesis inhibitor, Tyrosine kinase




inhibitor, VEGFR inhibitor


Alaproclate
BRD-A14966924
Serotonin receptor antagonist


Alfacalcidol
BRD-K93433262
Vitamin D receptor agonist


Alisertib
BRD-K75295174
Aurora kinase inhibitor


Altrenogest
BRD-A27554692
Progestogen hormone


ALW-II-38-3
BRD-K68191783
Ephrin inhibitor


AM-281
BRD-K59419204
Cannabinoid receptor antagonist


Amiodarone
BRD-K17561142
Potassium channel blocker


Anandamide
BRD-K78280988
Cannabinoid receptor agonist


APHA-compound-8
BRD-K74733595
HDAC inhibitor


Apicidin
BRD-K64606589
HDAC inhibitor


Arcyriaflavin-a
BRD-K72726508
CDK inhibitor


Arecaidine
BRD-K63792901
Acetylcholine receptor agonist


Arecaidine
BRD-K23922020
Acetylcholine receptor agonist


AS-605240
BRD-K41895714
PI3K inhibitor


AS-703026
BRD-K89014967
MEK inhibitor


Aspirin
BRD-K11433652
Cyclooxygenase inhibitor


Avrainvillamide-
BRD-A70731303
nucleophosmin inhibitor


analog-5




AY-9944
BRD-K03642198
Hedgehog pathway modulator


Azathioprine
BRD-K32821942
Dehydrogenase inhibitor


AZD-8055
BRD-K69932463
MTOR inhibitor


Barasertib
BRD-K63923597
Aurora kinase inhibitor


BAY-36-7620
BRD-K54704028
Glutamate receptor antagonist


Benidipine
BRD-A35519318
Calcium channel blocker


Bergenin
BRD-A15034104
Interleukin inhibitor


BH31-1
BRD-A38913120
BCL inhibitor


BIBX-1382
BRD-K70914287
EGFR inhibitor, Tyrosine kinase inhibitor


BIX-01338
BRD-K26863634
Histone lysine methyltransferase inhibitor


BML-ST330
BRD-A77118605
Phospholipase inhibitor


BMS-191011
BRD-K95609758
Potassium channel activator


BMY-14802
BRD-A15435692
Sigma receptor antagonist


BMY-45778
BRD-K84895041
IP1 prostacyclin receptor agonist


Bosutinib
BRD-K99964838
ABL inhibitor, BCR-ABL kinase inhibitor, SRC




inhibitor


Brazilin
BRD-A83326220
Nitric oxide production inhibitor


BRD-K64835161
BRD-K64835161



Brompheniramine
BRD-A68723818
Histamine receptor antagonist


Calmidazolium
BRD-A98283014
Calcium channel blocker, Calmodulin




antagonist


Camptothecin
BRD-A30437061
Topoisomerase inhibitor


Ceforanide
BRD-K37848908
Penicillin binding protein inhibitor


Cetraxate
BRD-K48932581
Mucus protecting agent


CGP-7930
BRD-K65786282
GABA receptor positive allosteric modulator


CHEMBL-374350
BRD-K59962020
NFKB pathway inhibitor


Chenodeoxycholic-
BRD-K18135438
11-β-HSD1 inhibitor, FXR agonist


acid




Chlorpromazine
BRD-K89997465
Dopamine receptor antagonist


Ciclacillin
BRD-K89046952
Bacterial cell wall synthesis inhibitor


Cimaterol
BRD-A65440446
Adrenergic receptor agonist


Cisapride
BRD-K06895174
Serotonin receptor agonist


Clobenpropit
BRD-K71430621
Histamine receptor antagonist


Cortisone
BRD-A54487287
Glucocorticoid receptor agonist


Corynanthine
BRD-K06467078
Adrenergic receptor antagonist


Dactolisib
BRD-K12184916
MTOR inhibitor, PI3K inhibitor, Protein kinase




inhibitor


Dapsone
BRD-K62363391
Bacterial antifolate


Decitabine
BRD-K79254416
DNA methyltransferase inhibitor


Demeclocycline
BRD-A75368507
Bacterial 30S ribosomal subunit inhibitor


Dephostatin
BRD-K60274257
Tyrosine phosphatase inhibitor


Desipramine
BRD-K60762818
Tricyclic antidepressant


Desmethylclozapine
BRD-K10042277
Acetylcholine receptor agonist


Desoxycorticosterone
BRD-A75402480
Mineralocorticoid receptor agonist


Dichlorobenzamil
BRD-K12906962
Sodium/calcium exchange inhibitor


Dihydrosamidin
BRD-K63945320
Phospholipase inhibitor, Nitric oxide production




inhibitor, platelet activating factor receptor




antagonist


Dipyridamole
BRD-K86301799
Phosphodiesterase inhibitor


Droxinostat
BRD-K11558771
HDAC inhibitor


Duloxetine
BRD-K71103788
Serotonin and norepinephrine reuptake inhibitor


Dydrogesterone
BRD-K68620903
Progesterone receptor agonist


E-4031
BRD-K41713976
Potassium channel blocker


Edrophonium
BRD-K81128206
Acetylcholinesterase inhibitor


Eicosatrienoic-acid
BRD-K63913457
Vasodilator


Elesclomol
BRD-K82135108
Oxidative stress inducer


Emetine
BRD-A25687296
Protein synthesis inhibitor


ENMD-2076
BRD-K68488863
FLT3 inhibitor, VEGFR inhibitor, Aurora




kinase inhibitor


Entacapone
BRD-K83636919
Catechol O methyltransferase inhibitor


Entinostat
BRD-K77908580
HDAC inhibitor


Epicatechin
BRD-K50660797
Bacterial DNA gyrase inhibitor,




Cyclooxygenase inhibitor, DNA polymerase




inhibitor


Equilin
BRD-K04046242
Estrogen receptor agonist


Eugenol
BRD-K32977963
Androgen receptor antagonist


Ezetimibe
BRD-A41519720
Niemann-Pick C1-like 1 protein antagonist,




Cholesterol inhibitor


Fenoldopam
BRD-A50684349
Dopamine receptor agonist


FGIN-1-27
BRD-K09778810
Inositol monophosphatase inhibitor


Flavanone
BRD-A07824748
11-β-HSD1 inhibitor


Fludarabine
BRD-K66788707
DNA synthesis inhibitor, DNA repair enzyme




inhibitor, Purine antagonist


Formestane
BRD-A31801025
Aromatase inhibitor


Fostamatinib
BRD-K20285085
SYK inhibitor


FR-122047
BRD-K30990140
Cyclooxygenase inhibitor


GBR-12783
BRD-K92015269
Dopamine uptake inhibitor


GBR-12935
BRD-K50135270
Dopamine uptake inhibitor


Gemcitabine
BRD-K15108141
Ribonucleotide reductase inhibitor


Glipizide
BRD-K12219985
Sulfonylurea


GR-127935
BRD-K11911061
Serotonin receptor antagonist


GS-39783
BRD-K75478907
GABA receptor modulator


GSK-1059615
BRD-K06750613
PI3K inhibitor


GSK-1904529A
BRD-K04833372
IGF-1 inhibitor, IGF-1R inhibitor, Insulin




receptor ligand


GW-9662
BRD-K93258693
PPAR receptor antagonist


H-7
BRD-A55756846
PKA inhibitor


HDAC3-selective
BRD-K29313308
HDAC inhibitor


Heliomycin
BRD-K64517075
ATP synthase inhibitor


Heraclenol
BRD-A77050075
Vitamin K antagonist


Hexylresorcinol
BRD-K99946902
Local anesthetic


HG-6-64-01
BRD-U37049823
RAF inhibitor


Homoharringtonine
BRD-K76674262
Protein synthesis inhibitor


Homosalate
BRD-A34751532
HSP inducer


Hydroxycholesterol
BRD-A36707673
LXR agonist


Hyoscyamine
BRD-K40530731
Acetylcholine receptor antagonist


Ibuprofen
BRD-A17655518
Cyclooxygenase inhibitor, NFkB pathway




inhibitor


Imipramine
BRD-K38436528
Norepinephrine and Serotonin transporter




inhibitor


Immethridine
BRD-K49519092
Histamine receptor agonist


Iodophenpropit
BRD-K51918615
Histamine receptor antagonist


I-OMe-AG-538
BRD-K35377380
IGF-1 inhibitor


Ioxaglic-acid
BRD-K79124250
Radiopaque medium


Ipratropium
BRD-A05352148
Acetylcholine receptor antagonist


Isotretinoin
BRD-K76723084
Retinoid receptor agonist


JAK3-Inhibitor-II
BRD-K52850071
JAK inhibitor


JNJ-16259685
BRD-K64670467
Glutamate receptor antagonist


Kavain
BRD-A75455249
Calcium channel modulator, Sodium channel




blocker


KIN001-127
BRD-A29901043
ITK inhibitor


KU-0063794
BRD-K67566344
MTOR inhibitor


KU-55933
BRD-K25311561
ATM kinase inhibitor


KU-C103443N
BRD-A81402010
CDC inhibitor, Rho associated kinase inhibitor


KUC104502N
BRD-K24538644



L-165041
BRD-K40656405
PPAR receptor agonist


L-733060
BRD-K15791587
Tachykinin antagonist


L-741626
BRD-K05181463
Dopamine receptor antagonist


Leflunomide
BRD-K78692225
Dihydroorotate dehydrogenase inhibitor,




PDGFR receptor inhibitor


Linsitinib
BRD-K08589866
IGF-1 inhibitor


Liothyronine
BRD-K89152108
Thyroid hormone stimulant


Lonidamine
BRD-K96670504
Glucokinase inhibitor


LY-2140023
BRD-K49519144
Glutamate receptor agonist


LY-288513
BRD-K24675965
CCK receptor antagonist


Lypressin
BRD-K93331255
Vasopressin receptor agonist


M2-PK-activator
BRD-K80672993



m-
BRD-K36965586
Serotonin receptor agonist


chlorophenylbiguanide




Meclozine
BRD-A50311610
CAR agonist


Mefenamic-acid
BRD-K92778217
Cyclooxygenase inhibitor


Mepacrine
BRD-A45889380
Cytokine production inhibitor, NFkB pathway




inhibitor, TP53 activator


Mephenytoin
BRD-A83937277
Hydantoin antiepileptic


Mepireserpate
BRD-A71765365
Catecholamine depleting sympatholytic


Mercaptopurine
BRD-K91601245
Immunosuppressant, Protein synthesis inhibitor,




Purine antagonist


Mesna
BRD-M40783228
Antioxidant


Mesoridazine
BRD-A14395271
Dopamine receptor antagonist


Metergoline
BRD-A30435184
Dopamine receptor agonist, Serotonin receptor




antagonist


Metformin
BRD-K79602928
Insulin sensitizer


Methimazole
BRD-K54416256
Antithyroid


Metixene
BRD-A33711280
Acetylcholine receptor antagonist


Metrizamide
BRD-A45543382
Radiopaque medium


Midodrine
BRD-A79981887
Adrenergic receptor agonist


Mirtazapine
BRD-A64977602
Adrenergic receptor antagonist, Serotonin




receptor antagonist


MK-5108
BRD-K53665955
Aurora kinase inhibitor


ML-7
BRD-K93201660
Myosin light chain kinase inhibitor


ML-9
BRD-K68402494
Myosin light chain kinase inhibitor


Molsidomine
BRD-K35531059
Guanylyl cyclase activator


Moracizine
BRD-K21548250
Sodium channel blocker


Mosapride
BRD-A39052811
Serotonin receptor agonist


MR-16728
BRD-A30590053
Acetylcholine release enhancer, Acetylcholine




release stimulant


MRS-1845
BRD-A32949107
Calcium channel blocker


Mycophenolate-
BRD-K92428153
Dehydrogenase inhibitor, Hydroxycarboxylic


mofetil

acid receptor agonist, Immunosuppressant,




Inosine monophosphate dehydrogenase




inhibitor, Inositol monophosphatase inhibitor


n-arachidonyl-GABA
BRD-K06024458
cannabinoid receptor agonist


Narciclasine
BRD-K06792661
Coflilin signaling pathway activator, LIM




kinase activator, Rho associated kinase activator


NBI-27914
BRD-K61177364
CRF receptor antagonist


NGB-2904
BRD-K05181084
Dopamine receptor antagonist


Niacin
BRD-K61993165
NAD precursor with lipid lowering effect,




vitamin B


Nilotinib
BRD-K81528515
ABL inhibitor, BCR-ABL kinase inhibitor


NNC-55-0396
BRD-K78122587
T-type calcium channel blocker


Norgestrel
BRD-A50928468
Progesterone receptor agonist


NSC-663284
BRD-K03109492
CDC inhibitor


nTZDpa
BRD-K54708045
PPAR receptor agonist


NU-7026
BRD-K09537769
DNA dependent protein kinase inhibitor, MTOR




inhibitor, PI3K inhibitor


NU-7441
BRD-K00337317
DNA dependent protein kinase inhibitor, P-




glycoprotein inhibitor


NVP-TAE684
BRD-K50140147
ALK inhibitor


Ochratoxin-a
BRD-K39944607
Phenylalanyl tRNA synthetase inhibitor


Oleoylethanolamide
BRD-K66956375
Cannabinoid receptor agonist, Glucose




dependent insulinotropic receptor agonist,




Potassium channel blocker, PPAR receptor




agonist


OSI-027
BRD-K94294671
MTOR inhibitor


Oxfendazole
BRD-A33447119
Anthelmintic


Oxiconazole
BRD-K23369905
Bacterial cell wall synthesis inhibitor


Oxybutynin
BRD-A65013509
Acetylcholine receptor antagonist


Panobinostat
BRD-K02130563
HDAC inhibitor


PD-0325901
BRD-K49865102
MEK inhibitor, MAPK inhibitor, Protein kinase




inhibitor


PD-102807
BRD-A89337244
Acetylcholine receptor antagonist


PD-184352
BRD-K05104363
MEK inhibitor


Perospirone
BRD-K85503079
Dopamine and serotonin receptors' antagonist


PG-9
BRD-A70268693
Acetylcholine receptor agonist


PHA-665752
BRD-K95435023
c-Met inhibitor


Phenytoin
BRD-K55930204
Hydantoin antiepileptic


Phosphodiesterase-V-
BRD-K68873215
Phosphodiesterase inhibitor


inhibitor-II




Phylloquinone
BRD-A55815733
Vitamin K, Γ carboxylase enzyme


PI-103
BRD-K67868012
MTOR inhibitor, PI3K inhibitor


Pidorubicine
BRD-K04548931
Topoisomerase inhibitor


PIK-90
BRD-K99818283
PI3K inhibitor


Pimozide
BRD-K01292756
Dopamine receptor antagonist


Pizotifen
BRD-K75958195
Serotonin receptor antagonist


PKCβ-inhibitor
BRD-K89687904
PKC inhibitor


Ponalrestat
BRD-K68332390
Aldose reductase inhibitor


PP-30
BRD-K30677119
RAF inhibitor


Procyclidine
BRD-A31800922
Acetylcholine receptor antagonist


Prostaglandin-b2
BRD-K82865713
CAMP inhibitor


Proxymetacaine
BRD-K79116891
Sodium channel blocker


PSB-36
BRD-A70407468
Adenosine receptor antagonist


Pterostilbene
BRD-K92870997
Cyclooxygenase inhibitor, PPAR receptor




agonist


Puromycin
BRD-A28970875
Protein synthesis inhibitor


QL-XII-47
BRD-U86922168
BTK inhibitor, Cytoplasmic tyrosine protein




kinase BMX inhibitor


Raloxifene
BRD-K63828191
Estrogen receptor antagonist, Selective estrogen




receptor modulator (SERM)


Razoxane
BRD-K07265709
Chelating agent, Topoisomerase inhibitor


Rescinnamine
BRD-K52930707
ACE inhibitor


Reserpine
BRD-K95921201
Vesicular monoamine transporter inhibitor


Ribavirin
BRD-A96255180
Antiviral


Ropivacaine
BRD-K50938786
Sodium channel blocker


Rottlerin
BRD-K03816923
MAP kinase inhibitor, Protein kinase inhibitor


RS-17053
BRD-K76840893
Adrenergic receptor antagonist


RS-67333
BRD-K46142322
Serotonin receptor partial agonist


RU-28318
BRD-A92585442
Cytochrome P450 inhibitor


SA-792728
BRD-K20755323
Sphingosine kinase inhibitor


SA-94315
BRD-K20197062
Caspase inhibitor


Salicin
BRD-K64614248
Anti-inflammatory


Salmeterol
BRD-A01320529
Adrenergic receptor agonist


SB-216641
BRD-K30867024
Serotonin receptor antagonist


SB-590885
BRD-K01253243
RAF inhibitor


SCH-23390
BRD-K45435259
Dopamine receptor antagonist


SDZ-205-557
BRD-K15868788
Serotonin receptor antagonist


Semaxanib
BRD-K63504947
VEGFR inhibitor


Sertraline
BRD-K82036761
Serotonin receptor antagonist


SKF-81297
BRD-A09828896
Dopamine receptor agonist


SN-38
BRD-A36630025
Topoisomerase inhibitor


Somatostatin
BRD-K14681867
Somatostatin receptor agonist


Sphingosine
BRD-K62959606
Ceramidase inhibitor


Splitomycin
BRD-K27710560
SIRT inhibitor


SR-27897
BRD-K35629949
CCK receptor antagonist


Stavudine
BRD-K93880783
DNA directed DNA polymerase inhibitor,




Reverse transcriptase inhibitor


STO-609
BRD-K52620403
Calmodulin antagonist


Sulfafurazole
BRD-K50859149
Bacterial antifolate


Sumatriptan
BRD-K50938287
Serotonin receptor agonist


TC-2559
BRD-K67352070
Acetylcholine receptor agonist


Telmisartan
BRD-K73999723
Angiotensin receptor antagonist


Temsirolimus
BRD-A62025033
MTOR inhibitor


Terbinafine
BRD-K68132782
Fungal squalene epoxidase inhibitor


Terbutaline
BRD-A50157456
Adrenergic receptor agonist


Terfenadine
BRD-A06352418
Histamine receptor antagonist


Tetrahydrobiopterin
BRD-A67605442
Nitric oxide (NO) stimulant, NO synthase




stimulant, Phenylalanine 4-hydroxylase




stimulant


Tetrahydropalmatine
BRD-A43940795
Serotonin release inhibitor


TGX-221
BRD-A41692738
PI3K inhibitor


Thiotepa
BRD-K09631521
Cytochrome P450 inhibitor


Tolazoline
BRD-K46211610
Adrenergic receptor antagonist


Torin-1
BRD-K40175214
MTOR inhibitor, PI3K inhibitor


TPCA-1
BRD-K51575138
IKK inhibitor


Tranylcypromine
BRD-A43974575
Monoamine oxidase inhibitor


Tretinoin
BRD-K06926592
Retinoid receptor agonist, Retinoid receptor




ligand


Tribenoside
BRD-A60294240
Anti-inflammatory, Capillary stabilizing agent


Trichostatin-a
BRD-K68202742
HDAC inhibitor, CDK activator, ID1 inhibitor


Tyrphostin-AG-556
BRD-K14441456
EGFR inhibitor


UNC-0321
BRD-K74236984
Histone lysine methyltransferase inhibitor


VER-155008
BRD-K32330832
HSP inhibitor


Vorinostat
BRD-K81418486
HDAC inhibitor


VU-0366037-2
BRD-K39823328
Glutamate receptor modulator


VU-0404997-2
BRD-A34208323
Glutamate receptor modulator


VU-0415374-1
BRD-K83010055
Glutamate receptor modulator


Wiskostatin
BRD-A18579359
Neural Wiskott-Aldrich syndrome protein




inhibitor


Wortmannin
BRD-A11678676
PI3K inhibitor


WZ-3146
BRD-K73293050
EGFR inhibitor


WZ-4-145
BRD-U25771771
EGFR inhibitor


Y-134
BRD-K94832621
Estrogen receptor antagonist


YM-976
BRD-K12932420
Phosphodiesterase inhibitor


Zamifenacin
BRD-K80451230
Acetylcholine receptor antagonist


ZM-447439
BRD-K72703948
Aurora kinase inhibitor
















TABLE 17







Complete list of drugs and compounds which can potentially elicit anti-cytokine


activity against hyperinflammation in SARS-COV-2-infected cells.









Drug/Compound Name
CMap ID
Mechanism of Action





3-matida
BRD-A87125127
Glutamate receptor antagonist


9-methyl-5H-6-thia-4,5-
BRD-K14696368
NFkB pathway inhibitor


diaza-chrysene-6,6-dioxide




AC-55649
BRD-K93176058
Retinoid receptor agonist


Acadesine
BRD-A95696820
AMPK activator


Acetyl-geranyl-cysteine
BRD-U01690642
Isoprenylated protein methylation




inhibitor


AICA-ribonucleotide
BRD-A67373739
AMPK activator


Alfuzosin
BRD-A09056319
Adrenergic receptor antagonist


Alosetron
BRD-K46742498
Serotonin receptor antagonist


Alprenolol
BRD-A00993607
Adrenergic receptor antagonist


Alverine
BRD-K89055274
Muscle relaxant


ALW-II-38-3
BRD-K68191783
Ephrin inhibitor


Aminomethyltransferase
BRD-A28318179
Nitric oxide synthase inhibitor


Amoxapine
BRD-K02265150
Norepinephrine reuptake inhibitor


Amylocaine
BRD-A09062839
Local anesthetic


Anagrelide
BRD-K62200014
Phosphodiesterase inhibitor


AQ-RA741
BRD-K81729199
Acetylcholine receptor antagonist


AR-C133057XX
BRD-K40892394
Nitric oxide synthase inhibitor


Atorvastatin
BRD-U88459701
HMGCR inhibitor


Auraptene
BRD-K85013741
Nitric oxide production inhibitor


AY-9944
BRD-K03642198
Hedgehog pathway modulator


AZD-6482
BRD-K58772419
PI3K inhibitor


AZD-8055
BRD-K69932463
MTOR inhibitor


Azelastine
BRD-A68888262
Histamine receptor antagonist


Benzydamine
BRD-K76133116
Membrane integrity inhibitor, Prostanoid




receptor antagonist, Prostanoid receptor




inhibitor


Bepridil
BRD-A91008255
Calcium channel or L-type Ca++ channel




blocker


Berbamine
BRD-K50464341
Calmodulin antagonist


BI-78D3
BRD-K73982490
JNK inhibitor


BIIB021
BRD-K51967704
HSP inhibitor


Bisoprolol
BRD-A89175223
Adrenergic receptor antagonist


bis-tyrphostin
BRD-K32906660
EGFR inhibitor


BMS-299897
BRD-K02950022
γ secretase inhibitor


BP-554
BRD-K45479396
Serotonin receptor agonist


BRD-A80383043
BRD-A80383043
Glutamate receptor agonist and/or




antagonist


BRD-K34437622
BRD-K34437622
Thymidylate synthase inhibitor


BRD-K63784565
BRD-K63784565
Topoisomerase inhibitor


BRD-K64835161
BRD-K64835161
NA


BRL-52537
BRD-A37347161
Opioid receptor agonist


Bromfenac
BRD-K47679368
Cyclooxygenase inhibitor


Bromocriptine
BRD-A69960130
Dopamine receptor agonist


Buphenine
BRD-A36267905
Adrenergic receptor agonist


Bupropion
BRD-A05186015
Dopamine uptake inhibitor


Butylparaben
BRD-K08287586
DNA synthesis inhibitor


Carmoxirole
BRD-K82484965
Dopamine receptor agonist


Carpindolol
BRD-A15530910
Adrenergic receptor antagonist, serotonin




receptor antagonist


Carteolol
BRD-A42167015
Adrenergic receptor antagonist


CDK1-5-inhibitor
BRD-K87932577
CDK inhibitor, Glycogen synthase kinase




inhibitor


CGP-54626
BRD-A55369275
GABA receptor antagonist


CGP-60474
BRD-K79090631
CDK inhibitor


CGS-20625
BRD-K68103045
Benzodiazepine receptor agonist, GABA




benzodiazepine site receptor partial




agonist


Chlordiazepoxide
BRD-K86595100
Benzodiazepine receptor agonist


Chlorphenamine
BRD-A04553218
Histamine receptor antagonist


Chlorprothixene
BRD-K59058766
Dopamine receptor antagonist


Cisapride
BRD-K06895174
Serotonin receptor agonist


CITCO
BRD-K53263234
CAR agonist


CL-82198
BRD-K00675675
Metalloproteinase inhibitor


Clarithromycin
BRD-K49668410
Bacterial 50S ribosomal subunit inhibitor


Clebopride
BRD-K17294426
Dopamine receptor antagonist


Clonidine
BRD-K98530306
Adrenergic receptor agonist


CNQX
BRD-K53545112
Glutamate receptor antagonist


Cotinine
BRD-K94144010
Nicotine metabolite


Cyclazosin
BRD-A37837077
Adrenergic receptor antagonist


Cycloserine
BRD-K87226815
Bacterial cell wall synthesis inhibitor


D-64406
BRD-K27665173
PDGFR receptor inhibitor


Danazol
BRD-A92537424
Estrogen receptor antagonist,




Progesterone receptor agonist


Darinaparsin
BRD-K35723520
Apoptosis stimulant


Dasatinib
BRD-K49328571
BCR-ABL kinase inhibitor, Ephrin




inhibitor, KIT inhibitor, PDGFR receptor




inhibitor, SRC inhibitor, Tyrosine kinase




inhibitor


Daunorubicin
BRD-K43389675
RNA synthesis inhibitor, Topoisomerase




inhibitor


Dephostatin
BRD-K60274257
Tyrosine phosphatase inhibitor


Desipramine
BRD-K60762818
Tricyclic antidepressant


Dexketoprofen
BRD-K43764301
Cyclooxygenase inhibitor


Dichloroacetic-acid
BRD-K13664374
Pyruvate dehydrogenase kinase inhibitor


Dicyclohexylurea
BRD-K81521265
Epoxide hydolase inhibitor


Dicycloverine
BRD-K68507560
Acetylcholine receptor antagonist


Diethylstilbestrol
BRD-K45330754
Estrogen receptor agonist


Dinoprostone
BRD-K26521938
Prostanoid receptor agonist


Dipropyl-5ct
BRD-K32645441
Serotonin receptor agonist


DMBI
BRD-K96084870
PDGFR receptor inhibitor, VEGFR




inhibitor


Dopamine
BRD-K43887077
Dopamine receptor agonist


DUP-697
BRD-K06221026
Cyclooxygenase inhibitor


Edaravone
BRD-K35458079
Nootropic agent


EHNA
BRD-K27450477
Adenosine deaminase inhibitor


EI-247
BRD-K32710582
IGF-1 inhibitor


Eicosatetraynoic-acid
BRD-K06080977
Cyclooxygenase inhibitor, Lipoxygenase




inhibitor


EMD-386088
BRD-K47659338
Serotonin receptor agonist


EMF-bca1-60
BRD-K68437527
caspase inhibitor


Enrofloxacin
BRD-K76534306
Bacterial DNA gyrase inhibitor


Estradiol
BRD-A18917088
Contraceptive agent, Estrogen receptor




agonist


Etifenin
BRD-K63979671
Compound used in hepatobiliary scans of




the liver


Etilefrine
BRD-A09925278
Adrenergic receptor agonist


Exemestane
BRD-A73741725
Aromatase inhibitor


Felbamate
BRD-K99107520
Glutamate receptor antagonist


Fipronil
BRD-A50675702
GABA gated chloride channel blocker


FIT
BRD-K17896185
Opioid receptor agonist


Fluphenazine
BRD-K55127134
Dopamine receptor antagonist


Flutamide
BRD-K28307902
Androgen receptor antagonist


Foliosidine
BRD-A49734948
Plant alkaloid


Formestane
BRD-A31801025
Aromatase inhibitor


Fostamatinib
BRD-K20285085
SYK inhibitor


Fraxidin
BRD-K66944906
Carbonic anhydrase inhibitor


Fursultiamine
BRD-A71157293
Vitamin B


Gabazine
BRD-K93280214
GABA receptor antagonist


GANT-58
BRD-K64451768
GLI antagonist


Gavestinel
BRD-K49890030
Glutamate receptor antagonist


GDC-0941
BRD-K52911425
PI3K inhibitor


Geldanamycin
BRD-A19500257
HSP inhibitor


GR-144053
BRD-K12120659
Integrin antagonist


GR-206
BRD-K00184207
Aryl hydrocarbon receptor ligand


GR-235
BRD-K26674531
Estrogen receptor agonist, FXR




antagonist, Progesterone receptor agonist


Hexamethyleneamiloride
BRD-K40990712
Sodium/hydrogen antiport inhibitor


HLI-373
BRD-K17349619
MDM inhibitor


HY-11007
BRD-K97056771
BCR-ABL kinase inhibitor


ICI-199441
BRD-K73290745
Opioid receptor agonist


ICI-89406
BRD-A03359064
Adrenergic receptor antagonist


Ilomastat
BRD-K51662849
Matrix metalloprotease inhibitor


Indatraline
BRD-K01649396
Norepinephrine transporter inhibitor


Iodophenpropit
BRD-K51918615
Histamine receptor antagonist


Iproniazid
BRD-K88568253
Monoamine oxidase inhibitor


Ipsapirone
BRD-K90574421
Serotonin receptor agonist


Isoliquiritigenin
BRD-K33583600
Guanylate cyclase activator


ITE
BRD-K60298136
Aryl hydrocarbon receptor agonist


JAK3-Inhibitor-II
BRD-K52850071
JAK inhibitor


Ketanserin
BRD-K49671696
Serotonin receptor antagonist


KI-16425
BRD-A25569250
Lysophosphatidic acid receptor antagonist


KIN001-127
BRD-A29901043
ITK inhibitor


KIN001-220
BRD-K53561341
Aurora kinase inhibitor


KIN001-244
BRD-K09186807
Phosphoinositide dependent kinase




inhibitor


L-655240
BRD-K89402695
Thromboxane receptor antagonist


L-750667
BRD-K28806945
Dopamine receptor antagonist


Larixinic-acid
BRD-K40619305
Compound that interacts with metal




centers


Latrepirdine
BRD-K55703048
Glutamate receptor antagonist


Latrunculin-b
BRD-A19248578
Actin polymerization inhibitor,




Unidentified pharmacological activity


L-BSO
BRD-A47706533
Glutathione transferase inhibitor


Linsitinib
BRD-K08589866
IGF-1 inhibitor, insulin inhibitor, ARF6




and TBK1 activator


Liothyronine
BRD-K89152108
Thyroid hormone stimulant


Lisuride
BRD-K88871508
Dopamine receptor agonist


Loperamide
BRD-K61250553
Opioid receptor agonist


Loratadine
BRD-K82795137
Histamine receptor antagonist


Mafenide
BRD-K30649484
Carbonic anhydrase inhibitor


Maprotiline
BRD-K03319035
Norepinephrine reuptake inhibitor,




Tricyclic antidepressant


m-chlorophenylbiguanide
BRD-K36965586
Serotonin receptor agonist


MDM2-inhibitor
BRD-K84987553
MDM inhibitor


Mead-ethanolamide
BRD-K09764130
Cannabinoid receptor agonist


Mebeverine
BRD-A09467419
Acetylcholine receptor antagonist


Medetomidine
BRD-A66563878
Adrenergic receptor agonist


Mepacrine
BRD-A45889380
Cytokine production inhibitor, NFkB




pathway inhibitor, TP53 activator


Meprylcaine
BRD-K65417056
Local anesthetic


Mepyramine
BRD-K97564742
Histamine receptor antagonist


Metoclopramide
BRD-K75641298
Dopamine receptor and serotonin receptor




antagonist


Midodrine
BRD-A79981887
Adrenergic receptor agonist


Milrinone
BRD-K67080878
Phosphodiesterase inhibitor


MK-2206
BRD-K68065987
AKT inhibitor


MLN-4924
BRD-K67844266
Nedd activating enzyme inhibitor


MR-16728
BRD-A30590053
Acetylcholine release enhancer or




stimulant


MW-STK33-3B
BRD-K64310881
Potassium channel activator


n-(3-acetamidophenyl)-3-
BRD-K61217870
Glutamate receptor antagonist


chlorobenzamide




Naftopidil
BRD-A01787639
Adrenergic receptor antagonist


NAS-181
BRD-A23683907
Serotonin receptor antagonist


Navitoclax
BRD-K82746043
BCL inhibitor


Nefazodone
BRD-K90789829
Adrenergic inhibitor, Norepinephrine




reuptake inhibitor, Serotonin receptor




antagonist, Serotonin reuptake inhibitor


Nevirapine
BRD-K15502390
Reverse transcriptase inhibitor


Nicorandil
BRD-K97752965
Nitric oxide donor, Potassium channel




activator


Nicotine
BRD-K05395900
Acetylcholine receptor agonist


Nifedipine
BRD-K96354014
Calcium channel blocker


Nifurtimox
BRD-A00100033
DNA inhibitor


Nikkomycin
BRD-A74771556
Chitin inhibitor


Nimodipine
BRD-A58048407
Calcium channel blocker


NNC-05-2090
BRD-K85015012
GAT inhibitor, GABA uptake inhibitor


Nor-binaltorphimine
BRD-A11135865
Opioid receptor antagonist


Norgestimate
BRD-A04756508
Progesterone receptor agonist


NVP-AUY922
BRD-K41859756
HSP inhibitor


O-2050
BRD-K02590140
Cannabinoid receptor antagonist


o-3M3FBS
BRD-K46384212
phospholipase activator


Olanzapine
BRD-K18895904
Dopamine receptor/serotonin receptor




antagonist


Orantinib
BRD-K91696562
FGFR, VEGFR, PDGFR inhibitor


Ornidazole
BRD-A42759514
Antiprotozoal


Otenzepad
BRD-A00520476
Acetylcholine receptor antagonist


Oxantel
BRD-K66019333
Anthelmintic


Oxaprozin
BRD-K25394294
Cyclooxygenase inhibitor


Oxybenzone
BRD-K59037100
Lipase inhibitor


Oxybutynin
BRD-A65013509
Acetylcholine receptor antagonist


Ozagrel
BRD-K19525698
Thromboxane synthase inhibitor


Palonosetron
BRD-K08924299
Serotonin receptor antagonist


Pantoprazole
BRD-A22380646
ATPase inhibitor


PCA-4248
BRD-A29289453
Platelet activating factor receptor




antagonist


PF-04217903
BRD-K73319509
c-Met inhibitor


Phenelzine
BRD-K87024524
Monoamine oxidase inhibitor


Phenothiazine
BRD-K59597909
Dopamine receptor antagonist


PI-103
BRD-K67868012
MTOR inhibitor, PI3K inhibitor


PIK-75
BRD-M16762496
DNA protein kinase inhibitor, PI3K




inhibitor


PIK-90
BRD-K99818283
PI3K inhibitor


Piperacetazine
BRD-K16277217
Dopamine receptor antagonist


Piperine
BRD-K59522102
Monoamine oxidase inhibitor


Pirenperone
BRD-K25224017
Serotonin receptor antagonist


Pirfenidone
BRD-K96862998
TGF β receptor inhibitor


Piribedil
BRD-K47936004
Dopamine receptor agonist


PLX-4720
BRD-K16478699
RAF inhibitor


PNU-22394
BRD-K16551401
Serotonin receptor agonist


PP-2
BRD-K95785537
SRC inhibitor


PP-30
BRD-K30677119
RAF inhibitor


Pravastatin
BRD-K60511616
HMGCR inhibitor


Prima-1-met
BRD-K49456190
thioredoxin inhibitor


Profenamine
BRD-A16311756
Butyrylcholinesterase inhibitor,




Cholinergic receptor antagonist


Promazine
BRD-K06980535
Dopamine receptor antagonist


Prostaglandin
BRD-K09436313
Prostanoid receptor antagonist


Prostaglandin-a1
BRD-K04010869
HSP inducer, NFkB pathway inhibitor


PSB-11
BRD-K10177585
Adenosine receptor antagonist


PTB1
BRD-K16554956
AMPK activator


PU-H71
BRD-K36529613
HSP inhibitor


Pyrazinamide
BRD-K28667793
Fatty acid synthase inhibitor


Pyroxamide
BRD-K11663430
HDAC inhibitor


Quinpirole
BRD-A85280935
Dopamine receptor agonist


Raltegravir
BRD-K05658747
HIV integrase inhibitor


Reserpic-acid
BRD-K32755366
Norepinephrine transporter inhibitor


Retinol
BRD-K13927029
Retinoid receptor ligand


Rhamnetin
BRD-K37206356
HDAC inhibitor


RITA
BRD-K00317371
MDM inhibitor


RO-08-2750
BRD-K00486786
NGF binding inhibitor


RO-25-6981
BRD-K51541829
Ionotropic glutamate receptor antagonist,




Monamine transporter modulator


Roscovitine
BRD-K07691486
CDK inhibitor


Rotenonic-acid
BRD-K34330170
Retinoid receptor antagonist


RS-67506
BRD-K50018155
Serotonin receptor partial agonist


Rucaparib
BRD-K88560311
PARP inhibitor


SA-792541
BRD-K68143200
CDC inhibitor


SAL-1
BRD-K40213712
Adenosine receptor antagonist


Salsolinol
BRD-K99595596
Monoamine oxidase inhibitor, Tyrosine




hydroxylase inhibitor


Saracatinib
BRD-K19540840
SRC inhibitor


SB-216763
BRD-K59184148
Glycogen synthase kinase inhibitor


SCH-28080
BRD-K55748775
ATPase inhibitor


SCH-442416
BRD-K46469693
Adenosine receptor antagonist


SD-169
BRD-K91904471
p38 MAPK inhibitor


SDZ-WAG-994
BRD-A31007383
Adenosine receptor agonist


Secoisolariciresinol
BRD-K91733562
Antioxidant


Securinine
BRD-A25775766
GABA receptor antagonist, TP53




activator


Selegiline
BRD-K86434416
Monoamine oxidase inhibitor


Sertraline
BRD-K82036761
Serotonin receptor antagonist


SID-26681509
BRD-K08417745
Cathepsin inhibitor


Sildenafil
BRD-K50128260
Phosphodiesterase inhibitor


Spironolactone
BRD-K90027355
Mineralocorticoid receptor antagonist


STO-609
BRD-K52620403
Calmodulin antagonist


Syrosingopine
BRD-K14200658
Vesicular monoamine transporter inhibitor


Taurodeoxycholic-acid
BRD-K33572481
Bile acid


Temefos
BRD-K51805276
Cholinesterase inhibitor


Temozolomide
BRD-K32107296
DNA alkylating agent


TER-14687
BRD-A33833419
Inhibitor of translocation of PKCq in T




cells


Testosterone
BRD-A48720949
androgen receptor agonist


TG-101348
BRD-K12502280
FLT3 inhibitor, JAK inhibitor


TGX-221
BRD-A41692738
PI3K inhibitor


Thenoyltrifluoroacetone
BRD-K00959089
Chelating agent


Thioproperazine
BRD-K08619574
Dopamine receptor antagonist


Ticlopidine
BRD-K00603606
Purinergic receptor antagonist


Tolterodine
BRD-K54316499
Acetylcholine receptor antagonist


Toltrazuril
BRD-K64514229
Antiprotozoal


Topiramate
BRD-K29653726
Carbonic anhydrase inhibitor, Glutamate




receptor antagonist, Kainate receptor




antagonist


Tosyllysyl-chloromethyl-
BRD-K10136726
Chymotrypsin inhibitor


ketone




TPCA-1
BRD-K51575138
IKK inhibitor


Trazodone
BRD-K70778732
Adrenergic receptor antagonist, Serotonin




receptor antagonist, Serotonin reuptake




inhibitor


Trifluoperazine
BRD-K89732114
Dopamine receptor antagonist


Triptolide
BRD-A13122391
RNA polymerase inhibitor


Tyrphostin-46
BRD-K60184833
Tyrosine kinase inhibitor


Tyrphostin-AG-82
BRD-K03670461
EGFR inhibitor


U-99194
BRD-K70281171
Dopamine receptor antagonist


UNC-0321
BRD-K74236984
Histone lysine methyltransferase inhibitor


Valproic-acid
BRD-K41260949
HDAC inhibitor


Verapamil
BRD-A09533288
Calcium channel blocker


VU-0420363-1
BRD-K59633790
SARS coronavirus 3C-like protease




inhibitor


WZ-4002
BRD-K72420232
EGFR inhibitor


Xaliproden
BRD-K88358234
Serotonin receptor agonist


XAV-939
BRD-K12762134
Tankyrase inhibitor


Y-27632
BRD-K44084986
Rho associated kinase inhibitor


YC-1
BRD-K60476892
Guanylyl cyclase activator


YS-035
BRD-K06208435
Calcium channel blocker


Zacopride
BRD-A65615053
Serotonin receptor antagonist


z-prolyl-prolinal
BRD-K60174629
Prolyl endopeptidase inhibitor


Zuclopenthixol
BRD-K28761384
Dopamine receptor antagonist


α-estradiol
BRD-A60070924
Estrogen receptor agonist









Example 3

Highly safe approved drugs repurposed for an antiviral indication, whose tissue distribution and mode of action overlap with the tropism of SARS-CoV-2 infection (e.g., airways), have the potential to complement and enhance the efficacy of drugs that are designed to specifically target virus-expressed proteins.


A recent study (Example 1) has shown that the long acting beta2-adrenoreceptor agonist bronchodilator, salmeterol, can block in vitro SARS-CoV-2 replication at clinically relevant concentrations without apparent host cell toxicity. The systems-level analyses are consistent with either salmeterol acting to enhance autophagy as previously suggested for Dengue virus infection (Medigeshi G et al. Antimicrob Agents Chemother. 2016, 60(11), 6709-9718) or alternatively, by acting to stimulate the innate immune response (Example 1).


A second compound, linisitinib (known to be an insulin-like growth factor 1 receptor (IGF1R) inhibitor, currently under investigation, not FDA-approved), also computationally predicted and experimentally verified (Example 1) to be a potent inhibitor of SARS-CoV-2 viral entry in a dose-dependent manner showed the highest inhibitory activity without overt cytotoxicity in spike-induced syncytia formation assays. It is further noted that this compound may have multiple modes of action: it interacts with ADP ribosylation factor 6 (ARF6), a binding partner of SARS-CoV-2 endonuclease nsp15 and it promotes autophagy through activation of TANK-binding kinase 1 (TBK1) mediated by the ubiquitination of the ARF domain TRIM23.


A third compound that deserves special attention among those proposed herein is imipramine, an FDA-approved tricyclic antidepressant known to act as an inhibitor of serotonin transporter (SERT). It is proposed that imipramine targets the amino acid transporter, BOAT1 (that is structurally homologous to SERT) and supports the host cell ACE2 receptor to which the SARS-CoV-2 spike protein binds. Notably, a serotonin transporter inhibitor (fluvoxamine) has recently been reported to decrease Covid-19 deaths by 90% (Sidik S. Nature, 2021, doi: 10.1038/d41586-021-02988-4; Reis G. et al. Lancet, 2022, 10(1), E42-E51). The derivatives of imipramine can likely serve as important antiviral drugs for alleviating, if not curing, Covid-10 effects.


The repurposing of approved (or investigational) drugs predicted to block virus replication-dependent host cell machinery creates both a high barrier to viral induced drug resistance and a low barrier to risk-averse regulatory approval relative to drugs specifically targeting virus-expressed proteins. Importantly, the simultaneous targeting of diverse viral vulnerabilities involving both host cell and viral encoded proteins can result in effective synergistic drug combinations that include salmeterol in combination with either the SARS-CoV-2 RNA-dependent RNA polymerase inhibitor molnupiravir or the 3CL protease inhibitor paxlovid or with both as a triple drug combination. This strategy could potentially be applied to the predicted drugs as indicated herein this application in combination with any drugs in development targeting any essential viral-encoded protein. The same type of combination therapies could be adopted using linsitinib and imipramine-derivatives. Finally, the use of drugs targeting viral proteins essential for evading the innate immune response (nsp16/nsp10) in combination with those, repurposed, that enhance the immune response is a promising strategy, that could be facilitated by utilizing the QuartataWeb interface (Li H et al. Bioinformatics, 2020, 36(12), 3935-3937).


Other advantages which are obvious and which are inherent to the invention will be evident to one skilled in the art. It will be understood that certain features and sub-combinations are of utility and may be employed without reference to other features and sub-combinations. This is contemplated by and is within the scope of the claims. Since many possible embodiments may be made of the invention without departing from the scope thereof, it is to be understood that all matter herein set forth or shown in the accompanying drawings is to be interpreted as illustrative and not in a limiting sense.


The methods of the appended claims are not limited in scope by the specific methods described herein, which are intended as illustrations of a few aspects of the claims and any methods that are functionally equivalent are intended to fall within the scope of the claims. Various modifications of the methods in addition to those shown and described herein are intended to fall within the scope of the appended claims. Further, while only certain representative method steps disclosed herein are specifically described, other combinations of the method steps also are intended to fall within the scope of the appended claims, even if not specifically recited. Thus, a combination of steps, elements, components, or constituents may be explicitly mentioned herein or less, however, other combinations of steps, elements, components, and constituents are included, even though not explicitly stated.

Claims
  • 1. The method of claim 3, wherein: the antiviral compound is selected from the group consisting of: imipramine, salmeterol, hexylresorcinol, brompheniramine, ezetimibe, temsirolimus, linsitinib, torin-1, rottlerin, semaxanib, ipratropium, AS-605240, mefenamic acid, JNJ16259685, QL-XII-47; derivatives thereof; and combinations thereof; andthe anti-hyperinflammatory compound is selected from the group consisting of: midodrine, olanzapine, trifluoperazine, fluphenazine, azelastine, chlorphenamine, clarithromycin, saracatinib, JAK3-Inhibitor-II, AZD-8055, CGP-60474, hexamethylene, loperamide, nifedipine, liothyronine, atorvastatin, triptolide, pirfenidone, isoliquiritigenin, rucaparib, berbamine, darinaparsin, taurodeoxycholic acid; derivatives thereof; and combinations thereof.
  • 2. (canceled)
  • 3. A method of treating or preventing a coronavirus infection in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a composition comprising an antiviral compound and an anti-hyperinflammatory compound.
  • 4. The method of claim 2, wherein the antiviral compound inhibits cell fusion or viral entry.
  • 5. The method of claim 3, wherein the antiviral compound comprises a histamine receptor antagonist, an acetylcholine receptor antagonist, a norepinephrine and serotonin reuptake inhibitor, an autophagy enhancer, a mTOR inhibitor, a PI3K inhibitor, an IGF-1- and insulin receptor inhibitor, a TBK1 activator through ARF1, an adrenergic receptor agonist, a VEGFR inhibitor, a local anesthetic, a cyclooxygenase inhibitor, a glutamate receptor antagonist, a Niemann-Pick Cl-like 1 protein antagonist, a cholesterol inhibitor, a cytoplasmic tyrosine protein kinase BMX inhibitor, a MAPK and protein kinase inhibitor, or a combination thereof.
  • 6. The method of claim 3, wherein the antiviral compound comprises: imipramine, salmeterol, hexylresorcinol, brompheniramine, ezetimibe, temsirolimus, linsitinib, torin-1, rottlerin, semaxanib, ipratropium, AS-605240, mefenamic acid, JNJ16259685, QL-XII-47; derivatives thereof; or a combination thereof.
  • 7. The method of claim 3, wherein the antiviral compound comprises: salmeterol, rottlerin, imipramine, linsitinib, hexylresorcinol, ezetimibe, brompheniramine; derivatives thereof; or a combination thereof.
  • 8. The method of claim 3, wherein the antiviral compound comprises salmeterol, linisitinib, imipramine, fluvoxamine, derivatives thereof, or a combination thereof.
  • 9. (canceled)
  • 10. The method of claim 3, wherein the antiviral compound is selected from an IGF-1R and insulin receptor inhibitor, an adrenergic receptor agonist, or a combination thereof.
  • 11. (canceled)
  • 12. The method of claim 3, wherein the anti-hyperinflammatory compound is selected from an adrenergic receptor agonist, a dopamine receptor antagonist, an autophagy enhancer, an autophagy dual modulator, a histamine receptor antagonist, a bacterial 50S ribosomal subunit inhibitor, an autophagy inhibitor, a SRC inhibitor, a JAK inhibitor, a mTOR inhibitor, a CDK inhibitor, a sodium/hydrogen antiport inhibitor, an opioid receptor agonist, a calcium channel blocker, a thyroid hormone stimulant, a HMGCR inhibitor, a RNA polymerase inhibitor, a TGFβ receptor inhibitor, an anti-fibrotic, a guanylate cyclase activator, a PARP inhibitor, a calmodulin antagonist, an apoptosis stimulant, a bile acid, or a combination thereof.
  • 13. The method of claim 3, wherein the anti-hyperinflammatory compound is selected from midodrine, olanzapine, trifluoperazine, fluphenazine, azelastine, chlorphenamine, clarithromycin, saracatinib, JAK3-Inhibitor-II, AZD-8055, CGP-60474, hexamethylene, loperamide, nifedipine, liothyronine, atorvastatin, triptolide, pirfenidone, isoliquiritigenin, rucaparib, berbamine, darinaparsin, taurodeoxycholic acid, or a combination thereof.
  • 14. The method of claim 3, wherein the anti-hyperinflammatory compound elevates IFN signaling, suppresses cytokine pathways, or a combination thereof.
  • 15. The method of claim 3, wherein the composition comprises salmeterol, linsitinib, impramine, derivatives thereof, or a combination thereof, optionally in combination with one or more additional agents.
  • 16. (canceled)
  • 17. (canceled)
  • 18. The method of claim 3, wherein the composition comprises salmeterol in combination with molnupiravir, paxlovid, or a combination thereof.
  • 19. (canceled)
  • 20. (canceled)
  • 21. (canceled)
  • 22. (canceled)
  • 23. (canceled)
  • 24. (canceled)
  • 25. The pharmaceutical composition of claim 27, wherein: the antiviral compound is selected from the group consisting of: imipramine, salmeterol, hexylresorcinol, brompheniramine, ezetimibe, temsirolimus, linsitinib, torin-1, rottlerin, semaxanib, ipratropium, AS-605240, mefenamic acid, JNJ16259685, QL-XII-47; derivatives thereof; and combinations thereof; andthe anti-hyperinflammatory compound is selected from the group consisting of: midodrine, olanzapine, trifluoperazine, fluphenazine, azelastine, chlorphenamine, clarithromycin, saracatinib, JAK3-Inhibitor-II, AZD-8055, CGP-60474, hexamethylene, loperamide, nifedipine, liothyronine, atorvastatin, triptolide, pirfenidone, isoliquiritigenin, rucaparib, berbamine, darinaparsin, taurodeoxycholic acid; derivatives thereof; and combinations thereof.
  • 26. (canceled)
  • 27. A pharmaceutical composition for the treatment of coronavirus comprising a pharmaceutically acceptable excipient and a therapeutically effective amount of a composition comprising an antiviral compound and an anti-hyperinflammatory compound.
  • 28. The pharmaceutical composition of claim 27, further comprising a propellant.
  • 29. The pharmaceutical composition of claim 28, wherein the propellant comprises compressed air, ethanol, nitrogen, carbon dioxide, nitrous oxide, hydrofluoroalkanes (HFA), 1,1,1,2,-tetrafluoroethane, 1,1,1,2,3,3,3-heptafluoropropane, or a combination thereof.
  • 30. A pressurized container comprising the pharmaceutical composition of claim 27, wherein the pressurized container comprises a manual pump spray, inhaler, meter-dosed inhaler, dry powder inhaler, nebulizer, vibrating mesh nebulizer, jet nebulizer, or ultrasonic wave nebulizer.
  • 31. (canceled)
  • 32. A method of identifying a compound for treating or preventing an infection with an infectious microbe in a subject in need thereof, the method comprising: a) obtaining transcriptomic data from cells infected with the infectious microbe,b) identifying differentially expressed genes (DEGs),c) characterizing host-targeted antimicrobial or anticytokine signature,d) identifying compounds that stimulate the anti-microbial or -cytokine signature,e) evaluating known and predicted targets of compounds identified in step d),f) constructing an infection host response protein-protein interaction (PPI) network and modules,g) prioritizing compounds based on network proximity analysis,h) clustering of prioritized compounds associated with selected disease modules,i) selecting representative compounds from each cluster for in vitro assays, andj) analyzing the results of steps a-i to thereby identify the compound for treating or preventing the infection.
  • 33. The method of claim 32, wherein the infectious microbe comprises a coronavirus.
  • 34-56. (canceled)
CROSS-REFERENCE TO RELATED APPLICATIONS

This application claims the benefit of priority to U.S. Provisional Application No. 63/140,574, filed Jan. 22, 2021, which is hereby incorporated herein by reference in its entirety.

STATEMENT OF GOVERNMENT SUPPORT

This invention was made with government support under grant numbers GM103712; DK119973; and DK117881 awarded by the National Institutes of Health. The government has certain rights in the invention.

PCT Information
Filing Document Filing Date Country Kind
PCT/US2022/013449 1/24/2022 WO
Provisional Applications (1)
Number Date Country
63140574 Jan 2021 US