key: cord-0830515-blx0ybgg authors: El-Hachem, Nehme; Eid, Edward; Nemer, Georges; Dbaibo, Ghassan; Abbas, Ossama; Rubeiz, Nelly; Zeineldine, Salah; Matar, Ghassan M.; Bikorimana, Jean-Pierre; Shammaa, Riam; Haibe-Kains, Benjamin; Kurban, Mazen; Rafei, Moutih title: Integrative transcriptome analyses empower the anti-COVID-19 drug arsenal date: 2020-10-19 journal: iScience DOI: 10.1016/j.isci.2020.101697 sha: b7e6d92cfdd523cd7b3db51ba83b871eefb20628 doc_id: 830515 cord_uid: blx0ybgg The beginning of the twenty-first century has been marked by three distinct waves of zoonotic coronavirus outbreaks into the human population. The COVID-19 (Coronavirus disease 2019) pandemic is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and emerged as a global threat endangering the livelihoods of millions worldwide. Currently, and despite collaborative efforts, diverse therapeutic strategies from ongoing clinical trials are still debated. To address the need for such an immediate call of action, we leveraged the largest dataset of drug-induced transcriptomic perturbations, public SARS-CoV-2 transcriptomic datasets, and expression profiles from normal lung transcriptomes. Most importantly, our unbiased systems biology approach prioritized more than 50 repurposable drug candidates (e.g., Corticosteroids, Janus kinase and Bruton kinase inhibitors). Further clinical investigation of these FDA approved candidates as monotherapy or in combination with an antiviral regimen (e.g., Remdesivir) could lead to promising outcomes in COVID-19 patients. m human (high and collapsed resolution he estimated cell populations from mouse timates) (Fig. 2f, Supplementary Fig. 1 ). We populations (1, 3, and 4); and was slightly less correlated with the collapsed hepatocyte population estimate (r = 0.64, p-value = 1.015 × 10 −21 ) (Fig. 2f, g) . This indicates that the low-resolution patients and on the other to the heavy-travel profile characteristic of most populations in the current era of globalization. COVID-19 can clinically manifest on a spectrum ranging from mild non-specific flu-like symptoms to near-fatal Acute Respiratory Distress Syndrome (ARDS) leading to inflammation, pneumonia, acute lung injury (ALI), and sepsis especially in the elderly and persons with underlying comorbidities (Chen et al., 2020) , (Wang et al., no date; Guan et al., 2020) . The current understanding of COVID-19 pathogenesis, etiology, and clinical features has been first extrapolated from knowledge gathered from other similar zoonotic infections associated with upper respiratory illness, such as the severe acute respiratory syndrome coronavirus (SARS-CoV) and the Middle East respiratory syndrome coronavirus (MERS-CoV) (Menter et al., 2020) , (Rota, 2003) , (Gu and Korteweg, 2007; Ng et al., 2016) . It is now established that SARS-CoV-2 specifically recognizes the human angiotensin-converting enzyme 2 protein (ACE2) as its main target on the cell surface (Gheblawi et al., 2020) , (Hoffmann, Kleine-Weber, et al., 2020) . Following virus entry, the cell triggers the physiological response to the infection through several defense arms that are also thought to be implicated in the pathogenesis of the infection Thus, characterizing the downstream transcriptional hallmarks of COVID-19 is crucial to illuminating the mechanisms of this deadly respiratory infection and to guide potent therapeutic countermeasures. However, with the urgency of the current pandemic situation, developing new drugs capable of combating COVID-19 is deemed difficult. To that end, drug repurposing represents a rapid approach to expediting the process of finding a therapeutic candidate (Harrison, 2020) (Gordon et al., 2020; Harrison, 2020) . Cases of drug repurposing have been serendipitous rather than data-driven discoveries and this extrapolates to COVID-19 research. The hydroxychloroquine-azithromycin combination has been proposed as a panacea for COVID-19 but has not been backed by a proven scientific rationale. Despite initial fanfare generated from favorable outcomes noted in an open-label, non-randomized clinical trial (Gautret et al., 2020) , the azithromycin-hydroxychloroquine combination fell short of expectations in later trials with therapeutic inefficacy overshadowed by cardiovascular side effects associated with its use (Bessière et al., 2020; Geleris et al., 2020; Rosenberg et al., 2020; Tang et al., J o u r n a l P r e -p r o o f 2020). This setback highlights the dire need for an unbiased methodology to further identify drug-target associations that can be efficiently translated to the clinic. Motivated by both an unmatched global pandemic and a lack of plausible drug candidates to treat COVID-19, we have conducted a unique data-driven approach To fill the unmet need for effective COVID-10 treatments, we leveraged the Library of In order to address pharmacological gaps in SARS-CoV-2 research, it was of paramount importance to understand the molecular mechanisms underlying the SARS-CoV-2 infection. To this end, we have collected public SARS-CoV-2 transcriptome-wide datasets and normal lung transcriptomics from GTEx. Subsequently, we conducted a differential expression analysis to compare SARS-CoV-2 conditions with their respective control groups. Next, all genes from the different comparisons were merged by the shrunken log2 fold change values (no-cutoff) to yield a full matrix corresponding to 7433 genes x 7 contrasts. These are referred to as SARS2_BALF_WUHAN1-2, SARS2_LUNG, SARS2_NHBE, SARS2_A549_ACE2, SARS2_Calu3 and For each of these seven contrasts, the fraction of significant up-and down-regulated genes (|Log2FoldChange| > 1 and p-adjusted-value < 0.05) and the corresponding overlap between contrasts is given in Supplementary Table S2 ) and genes were ranked accordingly. The ranked lists of genes across all SARS2 contrasts as well as genes ranked by their correlation coefficient from ACE2_GTEX were investigated at the pathway level using gene set enrichment analysis (GSEA). We assessed enrichment against a collection of hallmark genesets and a curated set of genes associated with SARS_CoV infection from the literature (See methods section and Table S3 ). To assess the similarity between settings, we first computed all pairwise correlations of pathway enrichment scores from all GSEA analysis ( Figure 2A ). Interestingly, both SARS2_A549_ACE2_RUXO and ACE2_GTEX were strongly concordant (rho= 0.63, p J o u r n a l P r e -p r o o f < 0.01). This suggests that ruxolitinib treatment in ACE2-expressing A549 cells and ACE2 baseline expression in the lungs engage a similar set of molecular pathways. Indeed, similarly to ACE2_GTEX, Ruxolitinib-induced pathways were highly anticorrelated with those from all the SARS2 settings (Fig 2A) . Although SARS2_LUNG was correlated with BALF settings, this correlation was not significant (p-value > 0.05, Fig 2A) suggesting a functional diversity in response to SARS-CoV-2. We further investigated the enriched hallmarks/pathways in both the normal lung and SARS-CoV-2 infected in vivo and in vitro settings. By keeping all gene sets with a normalized enrichment score |NES| greater or equal to 1 and a p-adjusted-value threshold of 5%, our analysis identified a highly conserved hallmark process, the TNF Alpha (Tumor necrosis factor-alpha) signaling via NF-κB. This hallmark was significantly upregulated by the SARS-CoV-2 infection but downregulated in both ruxolitinib-treated and ACE2_GTEX groups ( Figure 2B ). Moreover, to investigate the pathogenic cellular response of SARS-CoV-2, we proceeded as follow: (1) (2) were negatively scoring genesets in ACE2_GTEX and positively enriched in all SARS2 settings but the WUHAN_BALF samples ( Figure 2B ). Across all settings, the resulting genes were assigned either the shrunken log2 fold change from their corresponding DEG analysis or the correlation value for ACE2_GTEX ( Figure 2C ). From this collection of relevant genes of innate immunity, we identified a set of conserved upregulated genes in SARS2 settings (group1, Figure 2C ). These same upregulated genes were downregulated in ruxolitinib-treated cells and highly anticorrelated with reference to ACE2_GTEX. Most of these genes (e.g. In the previous section, we highlighted molecular vulnerabilities providing a rationale for targeting hyperinflammatory response in COVID-19. Although ruxolitinib provided a proof of concept that abrogating inflammation is an appealing therapeutic strategy, we conducted a full data-driven computational approach to identify potential drug J o u r n a l P r e -p r o o f candidates that, in addition to ruxolitinib, could perturb the pathological mechanism induced by the new SARS-CoV. To this end, we have interrogated the L1000, the largest repository of chemical-induced gene perturbation profiles. The curation and prefiltering of the L1000 dataset used in this study are described in the methods section. We first validated our approach on ruxolitinib signature in SARS-CoV2 infected cells (SARS2_A549_ACE2_RUXO). Our pipeline could accurately and unbiasedly identify Ruxolitinib from L1000 as the second-best matching hit out of 4,487 chemical compounds (cosine score= 0.18, p-value from 10,000 random permutations = 2e-04). To further ascertain that our approach can identify not only ruxolitinib but its drug target class, we performed a drug set enrichment analysis. In this setting, a drug set is a target gene/protein that is associated with at least 3 drugs. We first ranked drugs by the cosine value and ran the enrichment analysis against 295 target genes (see method section for drug-target preparation and database). With respect to SARS2_A549_ACE2_RUXO, we could confirm that indeed JAK2 inhibitors were significantly overrepresented at the top of the ranked list of cosine scores (ES= 0.825, adjusted p-value=0.08) ( Figure 3C ). Next, we investigated drug-target classes that display a negative enrichment score (their drug members likely reverse the phenotype) against a computed consensus cosine score which is the median value of the cosine spanning 4,487 drugs across all SARS2 settings. We postulate that the consensus score would emphasize drug classes J o u r n a l P r e -p r o o f that target generic processes induced by SARS-CoV-2 across diverse settings and infectivity phases/severity levels. Furthermore, drug-target enrichment analyses were conducted separately for each of the SARS2 settings (excluding RUXO group). ( Figure 3A ). Importantly, when considering an enrichment drug score (ES) less or equal to -0.5 and adjusted-p-value < 0.25 ( Figure 3A ), our consensus signature exhibited the strongest enrichment for JAK2 inhibitors (filgotinib, baricitinib, fostamatinib), Bruton kinase-BTK inhibitors (GDC-0834, ibrutinib, dasatinib; Figure 3D ), CACNA1C blockers ( Given the important role of ACE2 in both SARS-CoV-2 infectivity and normal lung physiology, we next sought to identify drugs that could counteract the SARS-CoV-2 invasion by restoring the baseline co-expression network of ACE2 in the healthy lung. In this particular analysis and similar to SARS2_A549_ACE2_RUXO, a positive enrichment score is the preferred outcome ( Figure 3B) . A drug-target class with an enrichment score equal or greater than 0.5 is considered a putative candidate to maintain a normal ACE2 expression whereas a drug-target with a negative enrichment score is likely to be beneficial for the virus. Figure 3E ). We are aware that only a limited fraction of the drugs is assigned to a target class (1030 out of 4487 drugs; 23%). As such, all results from our analyses will be made publicly available to accelerate the search for COVID-19 treatments. To understand the mechanism by which the candidate drug candidates block the molecular events underlying COVID-19, we focused on significant drug-target lists associated with the previously described consensus score. This involved 25 drug-target families and an ensemble of 106 drugs (Supplementary Figure S2) . Given the small number of genes from the L1000 dataset (727 out of the 978 landmark genes are shared with our data), we opted out to choose gene set enrichment analysis but instead, we conducted a row-wise Welch's t-test to identify the landmark genes that show significant changes between the 106 drugs and the SARS2 groups. This analysis yielded a set of 124 genes that displayed extreme values from the two-tailed t-test (False discovery rate < 5%, 73 genes with positive t-stat values genes, 51 genes with negative t-stat values)( Figure 4A ). When performing a hypergeometric test, separately for the negatively and positively modulated genes considering the set of significant genes, we showed that several down-regulated processes were enriched for Cytokine Signaling, Jak-STAT signaling, In this study, we focus on (i) defining aspects of molecular pathogenesis with regards to SARS-CoV-2 and (ii) validating potential therapeutic targets using integrative datadriven computational analyses. This was carried on large transcriptomic datasets encompassing both in vitro and in vivo SARS-CoV-2 infected samples as well as normal human lung biopsy samples in order to provide a global solid basis to validate our results. GSEA was initially carried out on all of the above datasets to ascertain the molecular pathways involved in SARS-CoV-2 pathogenesis. TNF alpha signaling via NFkB was found to be a highly conserved hallmark process across all SARS-CoV-2 models. This pathogenesis. It has been reported that serum levels of TNFα along with many other pro-inflammatory cytokines and chemokines were higher in SARS-CoV-2 ICU patients relative to their non-ICU counterparts (Siu et al., 2019; Huang et al., 2020) . TNFα plays an instrumental role in orchestrating the clinical outcomes seen in severe SARS-CoV and SARS-CoV-2 infections (reviewed in (Tay et al., 2020) ). Besides its role in the ominous cytokine storm, TNF-α strongly induces hyaluronan-synthase-2 (HAS2) in pulmonary epithelial and fibroblasts potentially leading to ARDS (Xu et al., 2020) . the acute lung injury characteristic of severe SARS-CoV and possibly SARS-CoV-2 (Kuba et al., 2005) , (Hoffmann, Kleine-Weber, et al., 2020) . In one study, it was shown that SARS-CoV S protein downregulates ACE2 by upregulating TNFα production which in turn acts in an autocrine fashion to induce the TNF-α-converting enzyme (TACE)dependent shedding of the ACE2 ectodomain (Haga et al., 2008) . Very recently, Feldman and colleagues (Robinson et al., 2020) severe SARS-CoV-2 infection (Channappanavar et al., 2017; Bi et al., 2020; Stelzig et al., 2020) . Furthermore, estrogen (E2) was found to decrease ACE2 expression in vitro J o u r n a l P r e -p r o o f cell models and thus potentially regulate SARS-CoV-2 infection (Bi et al., 2020; Stelzig et al., 2020) . Interferon-related genes such as STAT1 and CXCL10 as well as inflammatory response and IL-6-JAK-STAT pathway genes were significantly upregulated in the lung biopsy samples and minimally expressed in BALF tissue samples. The latter result possibly stems from the impressive arsenal of anti-IFN molecular tricks that beta coronaviruses have evolved to evade the impending immune response. The SARS-CoV nsp1 protein has demonstrated the ability to interfere with IFN-signaling pathways by decreasing the levels of phosphorylated STAT1 (Wathelet et al., 2007) while MERS-CoV membrane protein (M protein) and SARS-CoV ORF3b and -6 were found to prevent nuclear IRF3 translocation and subsequent interferonstimulated genes expression (Kopecky-Bromberg et al., 2007; Yang et al., 2013) . A testament to its pathogenic prowess, SARS-CoV-2 was found to trigger significantly lower levels of IFNs than SARS-CoV while boasting a superior infection and replication rate in the lungs (Chu et al., 2020) . In case of progression into severe SARS-CoV-2, the immune milieu adopts the transcriptomic profile of the lung sample dataset. Unabated viral replication eventually induces the delayed release of IFN-α/β and the subsequent influx of inflammatory macrophages. Consequently, proinflammatory cytokines (TNFalpha, IL-6, and IL1-β) are released to devastating effect. Inevitably, the aforementioned "cytokine storm" hinders viral clearance via T cell apoptosis and damages pulmonary microvascular and alveolar epithelial cell barriers via endothelial and epithelial cell apoptosis. The resultant vascular leakage and alveolar edema induce ARDS and lung damage (Ye, Wang and Mao, 2020) . Interestingly and in stark contrast to the other in vitro SARS-CoV-2 datasets, the NHBE SARS-CoV-2 set failed to register any tangible hallmark-associated gene expression. This could be explained by the cell tropism of SARS-CoV-2 in the human lungs that favors type I pneumocytes, type II pneumocytes, and alveolar macrophages over bronchial epithelia (Chu et al., 2020) . Alternatively, NHBE might be innately capable of clearing the virus. Elucidation of the exact cause might be addressed in future cellular studies. To identify potential repurposing candidates, we mined the L1000 connectivity map for Other enriched targets interfere with viral entry. During SARS-CoV and SARS-CoV-2 entry, cell surface-associated transmembrane protease serine 2 (TMPRSS2) cleaves the S protein to permit viral and cellular membrane fusion (Glowacka et al., 2011) . Nafamostat (RACONA trial; NCT04352400) and to a lesser extent, camostat (NCT04353284), both enriched in our analysis, have been shown to prevent SARS-CoV-2 entry by inhibiting TMPRSS2 (Glowacka et al., 2011; Hoffmann, Kleine-Weber, et al., 2020; Hoffmann, Schroeder, et al., 2020) . Clathrin-mediated endocytosis then ensues with regulation afforded by numb-associated kinases (NAK), such as AP2associated protein kinase 1 (AAK1) and cyclin G-associated kinase (GAK) (Sorrell et al., J o u r n a l P r e -p r o o f 2016). In addition to the JAK-STAT blockade discussed above, the JAK inhibitor baricitinib might also disrupt viral entry via AAK1 and GAK inhibition (Richardson et al., 2020) . Interestingly, chloroquine and hydroxychloroquine were not significantly enriched in our drug analysis. This pertinent negative finding is in line with the failure of hydroxychloroquine to demonstrate clinical efficacy. In effect, this finding lends credence to our data-driven analysis as it outperformed extrapolated biological validation of chloroquine and hydroxychloroquine, albeit on non-SARS-CoV-2 cell models. Finally, and given the limited set of genes profiled in LINCS L1000, we investigated whether the observed drug mechanism of action is associated with molecular events et al., 2006 , 2009 Lokugamage et al., 2012) . This results in the shutoff mechanism whereby viral RNA is preferentially transcribed and translated over host mRNA. Moreover, the massive metabolic suppression observed concur with recent findings from proteomic and metabolomic characterization from a severe case of COVID-19 (Shen et al., no date) . In conclusion, through a rigorous data-driven computational analysis, this study validates aspects of a complex pathogenetic framework for SARS-CoV-2 infection; however, targeted biological validation is still a requisite for a comprehensive With that in mind and as time is of the essence, the notion of combining drug candidates with different mechanisms of actions might provide a synergistic effect while simultaneously decreasing the risk of associated adverse drug reactions and expediting the process of testing each therapeutic candidate based on the severity of COVID-19 symptoms. The study does not provide experimental validation were significantly downregulated/upregulated in response to top candidate drugs (A) Welch's t-test was performed between gene levels induced by 106 selected drugs from L1000 and SARS-CoV-2 expression settings. Heatmap illustrating the expression level of 124 genes that significantly separate the two groups (p-adjusted value from the t-test of less than 0.05). (B) Heatplot for core pathways in which gene members were significantly downregulated in response to L1000 drugs. (C) Heatplot for core pathways in which gene members were significantly upregulated in response to L1000 drugs, in contrast to SARS2 settings (p-value < 0.05). Summary of public datasets used in the analytical pipeline J o u r n a l P r e -p r o o f IDO1 BCL2A1 CXCL10 IL1A RSAD2 IFIT2 MX2 IFIT1 IFIT3 OAS2 MX1 ISG15 IFITM1 TNFSF10 IFIH1 DDX58 IFITM3 IFITM2 CEACAM1 OAS1 OAS3 EIF2AK2 IFI27 EGFR CCL2 CEBPD BIRC3 ISG20 EDN1 NFAT5 ABCA1 NFE2L2 SMAD3 BCL3 IRF2 NAMPT RIPK2 PELI1 CFLAR TLR2 CD55 B2M ELF1 CASP4 STAT4 TLR1 LYN TLR4 IFNAR2 CDK6 CDKN1A AHR TRAF3 CREB1 EIF4E TNIP1 PTPN2 PANX1 XBP1 IL18 F2RL1 IL1R2 SLAMF7 CFH HIF1A IL1R1 PTAFR IRS2 PTGER4 CEBPB BIRC2 CASP8 TRAFD1 RBCK1 IFNGR1 MOV10 IFNGR2 SDC4 AXL IRF5 C5AR1 PTPN6 HLA−B TYK2 HLA−C TGFB1 CBL HMOX1 CASP7 RELA PLA2G4A IL13RA1 CTSB TNFRSF1A IRF3 CD40 STAT6 CASP9 STAT5A MAP2K3 IL4R SMAD7 HBEGF CSF1 IL2RG IL1B ZC3H12A STAT1 UBE2L6 TRIM21 DDX60 STAT2 DHX58 PLSCR1 SAMHD1 TRIM5 CASP1 PML JAK2 PLAUR TRIM25 MYD88 GBP2 TRIM14 C1R ADAR TXNIP IRF7 CD274 NLRC5 BST2 SERPING1 UBA7 CXCL2 EGR1 TNFAIP3 NFKBIA ATF3 NR4A3 TRAF1 CCL5 PTGS2 EREG SOCS3 NFIL3 NFKBIE REL IL23A NFKB1 NFKB2 TANK DDIT3 VEGFA RCAN1 TBK1 ATF4 TRAF6 MYC ICAM1 MAP3K8 RELB RASGRP1 IRF1 NOD2 DUSP1 JUN SPHK1 FOS SARS2_A549_ACE2 SARS2_Calu3 SARS2_LUNG SARS2_NHBE SARS2_BALF_WUHAN_1 SARS2_BALF_WUHAN_2 SARS2_A549_ACE2_RUXO group1 ZNF586 CDK7 RALA PMAIP1 TLK2 DNAJB6 FRS2 TRIB1 CCL2 TNFRSF21 ARL4C UBE2L6 IKBKB CSNK2A2 SPRED2 WDR7 SHB TFAP2A SOCS2 ADAT1 KIAA0753 DUSP11 PAFAH1B1 TRIM13 PTPRF TES ATP11B DNMT3A EDN1 GNA15 CASP10 IFNAR1 NFE2L2 SUPV3L1 CAMSAP2 STX4 CRKL EXT1 NVL NIT1 SNX13 LAP3 FBXL12 UBE2J1 HSPA4 PIK3CA CD40 RAB21 CAST PIP4K2B ELAVL1 YKT6 HDAC2 GNAI2 DCTD VAPB PGM1 NNT SYPL1 MLEC LBR ST7 CLTC TM9SF2 LIG1 NUP62 MSH6 NUP133 ETFB PFKL MRPS16 CDCA4 PSMD10 ADH5 WDR61 DAG1 HK1 HSD17B10 SMARCD2 SLC5A6 PSMG1 ANXA7 GLOD4 CRTAP TRAPPC3 NUDCD3 AKR7A2 TSPAN4 PPIE COPS7A BLMH IARS2 PRPF4 STMN1 NUP88 C2CD5 USP22 NUP85 XPO7 ZW10 COASY XPNPEP1 SORBS3 HERC6 TMEM109 RRP1B MCM3 RPA1 ICMT SACM1L UBE3C MPZL1 RPN1 SCARB1 LRPAP1 ITGB5 VAT1 INTS3 ARHGAP1 CAT HMG20B CLSTN1 Enriched for up-regulated genes in drugs vs. SARS-CoV2 • Drug enrichment identified BTK inhibitors as repurposable candidates for COVID- • Identified drug candidates warrant investigation in models of SARS-CoV-2 infection J o u r n a l P r e -p r o o f Assessment of QT Intervals in a Case Series of Patients With Coronavirus Disease 2019 (COVID-19) Infection Treated With Hydroxychloroquine Alone or in Combination With Azithromycin in an Intensive Care Unit Epidemiology and transmission of COVID-19 in 391 cases and 1286 of their close contacts in Shenzhen, China: a retrospective cohort study Imbalanced Host Response to SARS-CoV-2 Drives Development of COVID-19 Sex-Based Differences in Susceptibility to Severe Acute Respiratory Syndrome Coronavirus Infection Epidemiological and clinical characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: a descriptive study Comparative replication and immune activation profiles of SARS-CoV-2 and SARS-CoV in human lungs: an ex vivo study with implications for the pathogenesis of COVID-19', Clinical infectious diseases: an official publication of the Infectious Diseases Society of America Reproducible RNA-seq analysis using recount2 Coronavirus disease Inhibition of NF-κB-mediated inflammation in severe acute respiratory syndrome coronavirus-infected mice increases survival Repurposing FDA-Approved Drugs for COVID-19 Using a Data-Driven Approach Hydroxychloroquine and azithromycin as a treatment of COVID-19: results of an open-label non-randomized clinical trial Observational Study of Hydroxychloroquine in Hospitalized Patients with Covid-19 Angiotensin-Converting Enzyme 2: SARS-CoV-2 Receptor and Regulator of the Renin-Angiotensin System: Celebrating the 20th Anniversary of the Discovery of ACE2 Complex Immune Dysregulation in COVID-19 Patients with Severe Respiratory Failure Evidence that TMPRSS2 activates the severe acute respiratory syndrome coronavirus spike protein for membrane fusion and reduces viral control by the humoral immune response A SARS-CoV-2 protein interaction map reveals targets for drug repurposing' The Genotype-Tissue Expression (GTEx) project', Nature genetics Comorbidity and its impact on 1590 patients with COVID-19 in China: a nationwide analysis Pathology and Pathogenesis of Severe Acute Respiratory Syndrome Modulation of TNF-alpha-converting enzyme by the spike protein of SARS-CoV and ACE2 induces TNF-alpha production and facilitates viral entry Coronavirus puts drug repurposing on the fast track Nafamostat Mesylate Blocks Activation of SARS-CoV-2: New Treatment Option for COVID-19 SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor Clinical features of patients infected with 2019 novel coronavirus in Wuhan Severe acute respiratory syndrome coronavirus open reading frame (ORF) 3b, ORF 6, and nucleocapsid proteins function as interferon antagonists A crucial role of angiotensin converting enzyme 2 (ACE2) in SARS coronavirus-induced lung injury Geneshot: search engine for ranking genes from arbitrary text queries Similarity measures, author cocitation analysis, and information theory The Molecular Signatures Database Hallmark Gene Set Collection Discovery of potential drugs for COVID-19 based on the connectivity map Severe acute respiratory syndrome coronavirus protein nsp1 is a novel eukaryotic translation inhibitor that represses multiple steps of translation initiation Differential analysis of count data--the DESeq2 package Post-mortem examination of COVID19 patients reveals diffuse alveolar damage with severe capillary congestion and variegated findings of lungs and other organs suggesting vascular dysfunction Obesity and disease severity magnify disturbed microbiomeimmune interactions in asthma patients' Clinicopathologic, Immunohistochemical, and Ultrastructural Findings of a Fatal Case of Middle East Respiratory Syndrome Coronavirus Infection in the United Arab Emirates A Bayesian approach to accurate and robust signature detection on LINCS L1000 data Baricitinib as potential treatment for 2019-nCoV acute respiratory disease Accumulating evidence suggests anti-TNF therapy needs to be given trial priority in COVID-19 treatment Association of Treatment With Hydroxychloroquine or Azithromycin With In-Hospital Mortality in Patients With COVID-19 in New York State Characterization of a Novel Coronavirus Associated with Severe Acute J o u r n a l P r e -p r o o f Respiratory Syndrome Proteomic and Metabolomic Characterization of COVID-19 Patient Sera Severe acute respiratory syndrome coronavirus ORF3a protein activates the NLRP3 inflammasome by promoting TRAF3-dependent ubiquitination of ASC Family-wide Structural Analysis of Human Numb-Associated Protein Kinases The IUPHAR/BPS Guide to PHARMACOLOGY in 2016: towards curated quantitative interactions between 1300 protein targets and 6000 ligands Estrogen regulates the expression of SARS-CoV-2 receptor ACE2 in differentiated airway epithelial cells Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles A Next Generation Connectivity Map: L1000 Platform And The First 1,000,000 Profiles Hydroxychloroquine in patients with mainly mild to moderate coronavirus disease 2019: open label, randomised controlled trial The trinity of COVID-19: immunity, inflammation and intervention Clinical Characteristics of Patients with Severe Pneumonia Caused by the 2019 Novel Coronavirus in Wuhan Severe acute respiratory syndrome coronavirus evades antiviral signaling: role of nsp1 and rational design of an attenuated strain WikiPathways : An Experiment in the Community-based Curation of Biology DrugBank: a knowledgebase for drugs, drug actions and drug targets SARS and MERS: recent insights into emerging coronaviruses Pathological findings of COVID-19 associated with acute respiratory distress syndrome The structural and accessory proteins M, ORF 4a, ORF 4b, and ORF 5 of Middle East respiratory syndrome coronavirus (MERS-CoV) are potent interferon antagonists The pathogenesis and treatment of the `Cytokine Storm' in COVID-19 Nucleocapsid protein of SARS-CoV activates interleukin-6 expression through cellular transcription factor NF-kappaB Network-based drug repurposing for novel coronavirus 2019-nCoV/SARS-CoV-2', Cell discovery The authors would like to thank all researchers who shared their data publicly and made this project possible. This work is partially supported by a Discovery Grant from the National Sciences and Engineering Research Council of Canada (RGPIN/06101-2014) and an operating grant from the Cancer Research Society (OG24054). M.R holds a Fonds de la Recherche en Santé du Québec Junior II Award.All authors declare no conflict of interest R.S. is founder of IntelliStem Technologies Inc., Toronto, ON, Canada. The other authors have no competing interests. Processed data will be available upon request. A shiny app has been made available for public use