Skip to main content

This is a preprint.

It has not yet been peer reviewed by a journal.

The National Library of Medicine is running a pilot to include preprints that result from research funded by NIH in PMC and PubMed.

bioRxiv logoLink to bioRxiv
[Preprint]. 2020 May 17:2020.05.12.091256. [Version 1] doi: 10.1101/2020.05.12.091256

The Host Cell ViroCheckpoint: Identification and Pharmacologic Targeting of Novel Mechanistic Determinants of Coronavirus-Mediated Hijacked Cell States

Pasquale Laise 1,2, Gideon Bosker 1, Xiaoyun Sun 1, Yao Shen 1, Eugene F Douglass 2, Charles Karan 2, Ronald B Realubit 2, Sergey Pampou 2, Andrea Califano 2,3,4,5,6, Mariano J Alvarez 1,2
PMCID: PMC7263489  PMID: 32511361

Abstract

Most antiviral agents are designed to target virus-specific proteins and mechanisms rather than the host cell proteins that are critically dysregulated following virus-mediated reprogramming of the host cell transcriptional state. To overcome these limitations, we propose that elucidation and pharmacologic targeting of host cell Master Regulator proteins—whose aberrant activities govern the reprogramed state of coronavirus-infected cells—presents unique opportunities to develop novel mechanism-based therapeutic approaches to antiviral therapy, either as monotherapy or as a complement to established treatments. Specifically, we propose that a small module of host cell Master Regulator proteins (ViroCheckpoint) is hijacked by the virus to support its efficient replication and release. Conventional methodologies are not well suited to elucidate these potentially targetable proteins. By using the VIPER network-based algorithm, we successfully interrogated 12h, 24h, and 48h signatures from Calu-3 lung adenocarcinoma cells infected with SARS-CoV, to elucidate the time-dependent reprogramming of host cells and associated Master Regulator proteins. We used the NYS CLIA-certified Darwin OncoTreat algorithm, with an existing database of RNASeq profiles following cell perturbation with 133 FDA-approved and 195 late-stage experimental compounds, to identify drugs capable of virtually abrogating the virus-induced Master Regulator signature. This approach to drug prioritization and repurposing can be trivially extended to other viral pathogens, including SARS-CoV-2, as soon as the relevant infection signature becomes available.

Keywords: Coronavirus, Regulatory networks, Master regulator, Anti-viral drugs

Introduction

SARS-CoV is an enveloped, positive-sense, single-stranded RNA virus of the genera Betacoronavirus introduced into the human population from an animal reservoir and culminating in a lethal epidemic in 2002–03, affecting 8,098 individuals, 774 of whom died (9.6%)(1). The virus shares 79% genome sequence identity with SARS-CoV-2, which is responsible for the current COVID-19 pandemic(2). SARS-CoV can generate a rapid inflammatory cascade eventually leading to pneumonia or severe acute respiratory syndrome (SARS), characterized by diffuse alveolar damage, extensive disruption of epithelial cells and accumulation of reactive macrophages(3). Similar to SARS-CoV-2, SARS-CoV spike protein S binds to angiotensin converting enzyme 2 (ACE2), which is widely expressed on the cell membrane of oral, lung, and nasal mucosa epithelial cells, arterial smooth muscle and venous endothelial cells, as well of other organs, including stomach, small intestine, colon, skin, lymph nodes, spleen, liver, kidney, and brain(4). Supportive care—including prevention of Acute Respiratory Distress Syndrome (ARDS), multi-organ failure, and secondary infections—remains the foundational approach for managing serious infections caused by coronaviruses, although preliminary analysis of a recently-reported, prospective, randomized, placebo-controlled trial, suggests that patients receiving remdesivir recovered faster than those receiving placebo(57). Despite early optimism and approval on May 1st, 2020 of remdesivir for emergency use in hospitalized patients with COVID-19, no other specific antiviral treatment has been proven to be effective in randomized, placebo-controlled trials(5, 6). Consequently, there remains a formidable unmet need to identify pharmacologic treatments, alone or in combination—directly targeting either viral mechanisms and/or host cell factors—that significantly inhibit viral replication and, by extension, minimize progression of target organ failure associated with COVID-19.

Current efforts focusing on antiviral drug discovery can be summarized as belonging to two broad strategies: (a) disrupting the synthesis and assembly of viral proteins or (b) targeting host proteins and mechanisms required by the viral replication cycle. The first strategy has yielded drugs targeting (i) viral proteases, required for processing of the virus large replicase polyprotein 1a, producing non-structural proteins involved in viral transcription and replication(5, 8); (ii) RNA-dependent RNA-polymerase, using guanosine and adenosine analogs, as well as acyclovir derivatives; (iii) virus helicases; (iv) viral spike proteins, with antibodies, peptide decoys and carbohydrate-binding agents; and (v) structural proteins such as those maintaining ion channel activity of CoV E protein and RNA-binding affinity of CoV N protein(5, 6, 9, 10). Although virus-targeting approaches have the advantage of being specific, and, therefore, generally offer acceptable toxicity profiles, targeting viral products typically restricts the applicability of antiviral agents to only one, or only a few, closely related virus species. Moreover, due to the high mutation rate of viral genomes, such drugs are prone to rapid virus adaptation by resistant strain selection(11, 12). Considering the time required to develop new pharmacologic agents, this strategy has proven unsuitable to address new viral epidemics and pandemics in real time.

In contrast, targeting host cell proteins, especially at an early stage when viral hijacking of host mechanisms may still be reversible, may have more universal and longer term value because the same host factors may be required by multiple, potentially unrelated viral species and because host target proteins mutate far less rapidly than viral proteins, thereby limiting emergence of drug resistance(13). Unfortunately, pharmacologic targeting of host factors is more commonly associated with toxicity, thereby limiting clinical application of many drugs identified as potential anti-viral agents in vitro, for instance, with anti-CoV drugs EC50 markedly exceeding their maximum tolerated serum concentration (Cmax)(5). Despite these translational challenges, current approaches to target host proteins are primarily based on either boosting innate anti-viral immune response, in particular interferon response, or targeting proteins and processes mediating viral infection, such as ACE2 receptors(14), cell surface and endosomal proteases(15), and clathrin mediated endocytosis(16). Moreover, broad availability of high-throughput screening approaches has allowed the purposing and repurposing of drugs based on their effect on virus replication(1619), leading to identification of several anti-coronavirus candidates, such as chloroquine, tamoxifen, dasatinib and lopinavir, among others(16, 19). Yet, this approach is limited by the idiosyncratic nature of the in vitro models used in antiviral screens and by drug concentrations that may not be achievable in patients(5).

More recently, systems biology approaches, including temporal kinome analysis(20) and proteomics(2124), have also been used to identify protein kinases—and associated pathways—modulated in response to virus infection, as well as to generate virus-host protein-protein interactomes (PPI). These methods also present an opportunity to develop and test host-targeting therapeutic approaches that apply functional genomics to the “infected system as a whole.”(24) The output of these predictions can be used to direct drug repurposing efforts(2123) and to design more focused in vitro screens, with models that better recapitulate disease pathophysiology, such as primary cells, organoids or 3D organ-on-chip systems(25).

Coronaviruses have been shown to extensively hijack the cellular machinery of host cells they infect; as one example, this class of viruses induces arrest in S phase, allowing them to benefit from physiological alterations they induce in host cells that enhance their reproductive rate(26). As shown for other physiologic(2729) and pathologic cell states—among them, cancer(3034), neurodegeneration(35, 36), and diabetes(29)—we propose that such transcriptionally “locked” states are established by the virus and maintained by a handful of Master Regulator (MR) proteins, organized within a highly auto-regulated protein module, or checkpoint (see Califano & Alvarez(30) for a recent perspective). For simplicity, in a viral infection context, we will call such modules “ViroCheckpoints.” Accordingly, we propose that aberrant, virus-mediated activation of a ViroCheckpoint is ultimately responsible for creating a transcriptionally “locked” cellular context that is primed for viral replication and release. We thus propose ViroCheckpoint activity reversal as a potentially valuable therapeutic strategy for pharmacologic intervention.

Here we show that time-dependent, SARS-CoV-mediated ViroCheckpoints—and the specific MR proteins of which they are comprised—can be effectively elucidated by network-based analysis using the Virtual Inference of Protein activity by Enriched Regulon (VIPER) algorithm(37). More importantly, once the MR protein identity is available, drugs can be effectively and reproducibly prioritized based on their ability to invert the activity of ViroCheckpoint MR proteins, using the OncoTreat algorithm(34), a NYS CLIA-certified algorithm that is used routinely on cancer patients at Columbia University.(38)

Accurate identification of virus-dependent MR proteins permits deployment of the same OncoTreat-based methodological approach for mechanism-based repurposing or development of new drugs with potential anti-viral activity. To avoid confusion, we will use the term “ViroTreat” to indicate the virus-specific version of OncoTreat. Specifically, ViroTreat uses the full repertoire of virus-induced MR proteins in the ViroCheckpoint as a reporter assay to identify drugs capable of reversing its activity(34), thereby preventing emergence of or abrogating the virus-mediated transcriptional locked state. While limited by the availability of data on SARS-CoV-2, including of infection in an appropriate pathophysiologic cell context, we provide proof of concept that this approach can be applied to prioritizing FDA-approved and late-stage investigational drugs representing potential antiviral agents for SARS-CoV based on infection in cancer-related lung epithelial cells.

Results

Elucidating MRs of SARS-CoV infection in lung epithelial cells.

To identify candidate MR proteins that mechanistically regulate the host cell gene expression signature induced by SARS-CoV infection (i.e. the SARS-CoV ViroCheckpoint), we applied the VIPER algorithm to a previously-published, microarray-based gene expression signature of a Calu-3 lung adenocarcinoma cell clone expressing elevated ACE2 levels, compared to the parental line, at 12h, 24h, and 48h following infection with SARS-CoV at MOI = 0.1(39). A total of 6,054 regulatory proteins were considered in the analysis, including 1,793 transcription factors (TFs), 656 co-transcription factors (co-TFs), and 3,755 signaling proteins (SP).

Similar to a highly-multiplexed gene reporter assay, VIPER measures the activity of an individual protein based on the enrichment of its positively regulated and repressed targets in genes that are over- and under-expressed in a specific cell state, compared to a control(37). We have shown that VIPER can accurately measure the activity of >70% of regulatory proteins and, as a result, the algorithm has been used to elucidate MRs of both pathologic(3133, 35, 36, 40, 41) and physiologic cell states(2729) that have been experimentally validated. Moreover, VIPER-inferred protein activity has been shown to provide a better biomarker of cell phenotype than the original transcriptional profile(30, 34, 42, 43); and, importantly, is a better reporter for validating clinically relevant drug sensitivity(44). Accordingly, VIPER requires a differential expression signature for each sample to be analyzed and a regulatory model comprising the transcriptional targets of each regulatory protein. For the former, we computed a differential gene expression signature for each SARS-CoV infected sample, by comparing it to three 12h mock control replicates. For the latter, we leveraged a transcriptional regulatory model (interactome) generated by ARACNe(45) analysis of 517 samples in the lung adenocarcinoma cohort of The Cancer Genome Atlas (TCGA)(37). Use of a cancer-related interactome is well justified as we have shown that protein transcriptional targets are highly conserved between cancer and normal cells(28).

The analysis revealed n = 236 proteins, whose activity was significantly affected by SARS-CoV infection in at least one time point (p < 105, Bonferroni Corrected (BC), see Supplementary Table 1). Examination of the top 10 activated MR proteins at each of the evaluated time-points (Fig. 1a) revealed the presence of canonical cell-cycle regulators, including (a) cyclins (CCNA2), and other proteins involved in G1/S transition(46) (E2F8 and UHRF1); (b) S-phase proteins, such as topoisomerases (TOP2A(47)) and other factors involved in S-phase cell cycle arrest(48) (CHEK1, GTSE1); (c) mitotic checkpoint proteins(49) (BUB1B, KIF11 and NDC80); and (d) proteins involved in nucleotide synthesis (GMPS). These showed significant activation as early as 12h after SARS-CoV infection. In contrast, established innate immune response proteins were also found among the top activated MRs, including IFN-induced factors(50) (MX1, IRF9 and IFI27) but their activation became most evident only at the latest time point (48h). Interestingly, some proteins previously identified as key tumor MRs were strongly activated, such as FOXM1 and CENPF(33, 51), although this may be a byproduct of the cancer related nature of the Calu-3 cells used in the infection assays.

Fig. 1.

Fig. 1.

SARS-CoV-induced ViroCheckpoint in Calu-3 lung adenocarcinoma cells. (a) Heatmap showing the VIPER-inferred protein activity, expressed as normalized enrichment score (NES), for the top 10 most activated and the top 10 most inactivated proteins in response to SARS-CoV infection for each of the three time points. (b) Heatmap showing the similarity between the SARS-CoV induced protein activity signatures, expressed as Pearson’s correlation coefficient.

We then systematically evaluated whether viral infection could affect host proteins known to be involved in SARS-CoV host-pathogen protein-protein interactions (PPI). We based this analysis on a set of 36 proteins previously identified by high-throughput yeast-2-hybrid screen and validated by luciferase assays(23). Of the 36, 12 were represented among our set of 6,054 regulons and could thus be assessed for enrichment in SARS-CoV-induced differentially active proteins. Despite the low statistical power of a test based on only 12 proteins, enrichment was statistically significant for the 12h activity signature (p < 0.01, Supplementary Fig. 1a). Enrichment was borderline non-significant at 24h (p = 0.08), and not significant at 48h (Supplementary Figs. 1b and c).

To increase the test’s sensitivity, we leveraged a larger set of proteins identified as PPI for 26 of the 29 proteins coded by the closely related SARS-CoV-2 virus, as identified by mass-spec analysis of pull-down assays(21). Of 332 host proteins identified by that analysis, 89 were represented among those analyzed by VIPER. Confirming the prior results, enrichment was highly significant (p12h < 105 by 2-tail aREA test(37); p24h < 0.01 and p48h < 0.001 by 1-tail aREA test, see Supplementary Fig. 1g, k and l, respectively). Interestingly, while enrichment was significant at all three time points, (p < 0.01, 1-tail aREA test, Supplementary Fig. 1jl), several of the human SARS-CoV-2 PPIs activated at 12h became inactivated at later time points (Supplementary Fig. 1hi).

Correlation analysis showed a gradual shift in protein-activity signatures from 12h to 48h after infection (Fig. 1b), suggesting dynamic activation and inactivation of a diverse repertoire of genetic programs by virus-host interaction and thus dynamic transition across multiple, time-dependent ViroCheckpoints. To gain insight into the biological programs most profoundly affected by SARS-CoV infection, we performed Gene-Set Enrichment Analysis (GSEA)(52) of a set of 50 biologically-relevant hallmark gene-sets from MSigDB(53) in differentially active, infection-mediated proteins (Fig. 2). The analysis identified four time-dependent program classes including: (a) cell cycle programs, consistently up-regulated at all three time points; (b) immune-related programs, associated with interferon response, inflammatory response, TNF-α, and IL-6/JACK/STAT3 signaling, which were progressively upregulated over time; (c) DNA repair pathways and (d) PI3K/AKT/mTOR programs more strongly activated at 12h (Fig. 2).

Fig. 2.

Fig. 2.

Biological programs activated by SARS-CoV infection. (a) Hallmark gene-sets from MSigDB significantly enriched (FDR < 0.05) in proteins activated at 12h, 24h and 48h after SARS-CoV infection. The bars indicate the GSEA-estimated Normalized Enrichment Score (NES). Pathways and processes related to cell cycle progression and cell proliferation, DNA-repair, mTOR, IFN-α and inflammation are indicated by blue, yellow, purple, red and green arrows, respectively. (b) GSEA plots showing the enrichment of E2F-targets, IFN-α-response and IL6/JAK/STAT pathway hallmark gene-sets on the differential activity of 6,054 regulatory proteins at 12h, 24h and 48h after SARS-CoV infection. The x-axis shows the regulatory proteins sorted from the most inactivated (left), to the most activated (right) in response to viral infection. The y-axis shows the enrichment score estimated by GSEA. The blue vertical lines indicate the proteins annotated as part of each of the analyzed biological programs/pathways.

Consistent with the multifarious effects that coronaviruses are known to exert through their complex, synchronized modulations of cell cycle progression, interferon antagonism, interleukin 6 and 8 induction, and host protein synthesis(26), these findings disclose a time-dependency, with early vs. late activation of protein signatures each linked to a distinct set of biofunctional hallmarks resulting from a virus-governed reconfiguration of the host cell’s regulatory state, with alterations in cell cycle during the initial post-infection phase, followed by a phase characterized by ignition of pro-inflammatory cytokine signaling pathways.

ViroTreat analysis of SARS-CoV infected cells identifies novel therapeutic targets for drug repurposing.

We have previously developed and validated a systematic approach (OncoTreat) for identifying drugs and compounds capable of reversing the aberrant activity of all Tumor Checkpoint MRs, representing mechanistic determinants of cell state, on a patient by patient basis(34). As a direct result of the high reproducibility demonstrated by VIPER,(37) the test has been certified by the NYS-CLIA laboratory and is available in the United States from the Columbia University Laboratory of Personalized Genomic Medicine(38); and, in China, from the Xiamen Encheng Group Ltd.

OncoTreat is used routinely to assess potential therapy for cancer patients who are progressing on standard of care, as part of the Columbia Precision Oncology Initiative(54). Despite the fact that it was originally developed for deployment and drug prioritization in the setting of precision oncology, the OncoTreat methodology is fully generalizable and can be applied to any state transition and any drug collection, including transitions related to and induced by viral infection. To avoid confusion, we will use the term ViroTreat to refer to the algorithm when used to identify antiviral drugs (see description in Fig. 3).

Fig. 3.

Fig. 3.

ViroTreat diagram. ViroTreat requires two components: (A) a context-specific ViroTreata context-specific drug Mechanism of Action (MoA) database, which is generated by perturbing an appropriate cell model with therapeutically relevant drug concentrations, followed by VIPER analysis of the drug-induced gene expression signatures and identification of the top most differentially active proteins, both activated and inactivated in response to the drug; and (B) the specific virus-induced protein activity signature—where the most differentially active proteins constitute the ViroCheckpoint—dissected by VIPER analysis of a gene expression signature, obtained by comparing an infected tissue or relevant model with non-infected mock controls. ViroTreat then predicts the effect of the drugs on the ViroCheckpoint by matching their MoA with the virus-induced protein activity signature, and quantifies the inverse enrichment using the aREA algorithm. The diagram shows 3 drugs, where only drug B, by activating the host proteins that are being inactivated during virus infection, and inactivating the proteins that are being activated by the virus infection, effectively acts by inverting the ViroCheckpoint activity pattern; and, therefore, would be prioritized as a host cell-targeted antiviral therapeutic option.

ViroTreat requires a tissue-matched drug perturbation database. For this analysis, we had previously generated a collection of RNASeq profiles of NCI-H1793 lung adenocarcinoma cells, at 24h following treatment with a repertoire of 133 FDA approved and 195 late-stage (Phase 2 and 3) drugs—primarily used in or developed for the oncology setting—at their highest subtoxic concentration (48h IC20) or maximum serum concentration (Cmax), whichever is lower. RNASeq data was generated using a fully automated, 96-well based microfluidic technology called PLATE-Seq(55) (Supplementary Table 2). Selection of the NCI-H1793 cell line as an adequate model for the analysis was based on the significant overlap of SARS-CoV infection MR proteins with proteins differentially activated in this cell line (p < 1028, 1038, and 1024 at 12h, 24h and 48h after infection, by 1-tail aREA test; see Supplementary Fig. 2). In addition, the main rationale for these assays is the elucidation of protein-level MoA of a drug repertoire and MoA is generally well-recapitulated in lineage matched cells(56).

Using this predictive model, ViroTreat prioritized 44 FDA-approved drugs and 49 investigational compounds in oncology, based on their ability to significantly invert the ViroCheckpoint protein activity signature, at one or more of the 3 evaluated time-points following infection (p < 1010, BC; see Supplementary Table 3). Based on this analysis, two FDA-approved drugs—the CDK inhibitor palbociclib and the MEK inhibitor trametinib—and 4 investigational compounds, including three MAP kinase and one AKT/CHEK1 inhibitors, were able to significantly invert the ViroCheckpoint activity at all three time-points (p < 1010, BC, Fig. 4a). In addition, six FDA-approved drugs and seven investigational compounds demonstrated the capacity to invert the ViroCheckpoint protein activity pattern at the two earliest time points (12h and 24h, p < 1010, BC, Fig. 4a); while two FDA-approved drugs—the ALK and EGFR inhibitors brigatinib and osimertinib—and five investigational compounds were predicted to significantly invert the MR signature identified at later time points (24h and 48h, p < 1010, BC, Fig. 4a).

Fig. 4.

Fig. 4.

Top drugs and compounds identified by ViroTreat. (a) Table of FDA-approved drugs and investigational compounds identified by ViroTreat as significantly inverting the pattern of activity of the SARS-CoV induced checkpoint (p < 1010, BC) for at least one of the three analyzed time points, and being simultaneously significant (p < 105, BC) for at least another time point. The drugs and compounds were organized in blocks according to the biological role or pathway membership of their primary target protein. For each block, the drugs and compounds significant for each time point (p < 1010, BC), were sorted by their ViroTreat significant level for 12h, followed by 24h and 48h. FDA-approved drugs were reported prior to investigational compounds. The table also shows the concentration used to perturb NCI-H1793 cells, the ViroTreat significance level, as −log10(p-value), BC, indicated by the green heatmap, and the primary target for each of the significant drugs and compounds. (b–d) GSEA plots showing the enrichment of the top 25 proteins most activated (red vertical lines), and the top 25 proteins most inactivated (blue vertical lines), in NCI-H1793 cells in response to selinexor perturbation, on the protein activity signatures induced by SARS-CoV infection of Calu-3 cells (x-axis) for 12h (b), 24h (c) and 48h (d). NES and p-value, estimated by 2-tail aREA test, are indicated on top of each plot.

Consistent with the pathways enrichment analysis (Fig. 2), several drug families were enriched among the top ViroTreat predictions, including MAP kinases, PI3K/AKT/mTOR, CDK and other cell cycle-related drugs; HDAC and bromodomain protein inhibitors; proteasome and HSP90 inhibitors; and NF-κB and JAK inhibitors (Fig. 4a).

Of special clinical relevance in the context of the COVID-19 pandemic, ViroTreat independently identified the Selective Inhibitor of Nuclear Export (SINE) drug selinexor—FDA-approved for the treatment of relapsed or refractory multiple myeloma—as an extremely potent inverter of SARS-CoV induced ViroCheckpoint activity, in particular, at 12h and 24h time points after infection (p12h < 1016 and p24h < 1019, BC, Fig. 4).

Discussion

ViroTreat presents an application of the extensively validated OncoTreat algorithm for targeting MR proteins driving virus-mediated, reprogrammed cell states induced by viral hijacking of the host cell regulatory machinery. It also provides proof-of-concept of the ability to rapidly prioritize drugs capable of abrogating the reprogrammed, transcriptionally-locked state induced by viral infection, responsible for creating an environment permissive to viral replication and release. Our analysis identified 44 FDA-approved and 49 investigational agents capable of virtually abrogating the MR signature—the ViroCheckpoint protein activity pattern—induced by SARS-CoV infection.

Consistent with the observation that coronaviruses interfere with cell cycle progression to benefit from the physiology of host cells arrested in S phase(26), we show SARS-CoV infection-induced activation of MRs involved in cell cycle progression and DNA repair pathways. Notably, it has been reported previously that coronaviruses inhibit the pRb tumor suppressor protein, inducing infected cell to progress rapidly from G1 and to arrest the host cell in S phase(57). SARS-CoV further favors host cell arrest in S phase by inhibiting CDK4 and CDK6 kinase activity(58). We also observed activation of PI3K/AKT/mTOR pathway proteins, suggesting that SARS-CoV—similar to other viruses(59), including +ssRNA viruses like chikungunya(60), hepatitis C(61), west nile(62) and dengue(63), as well as other RNA respiratory viruses like influenza(64) and the respiratory syncytial virus(65)—might subvert mTOR pathway activity. Indeed, temporal kinome analysis of human hepatocytes infected with MERS-CoV had previously revealed changes in MAPK and PI3K/AKT/mTOR pathways(20). Finally, we observed activation of proteins involved in innate immunity, including interferon response and pro-inflammatory pathways, which have been also previously described for coronaviruses(26).

While formal experimental validation is still required, there are several positive indications this approach may be effective. Specifically, drugs for SARS-CoV most highly prioritized by ViroTreat were highly consistent, at least based on their primary target proteins, with biological programs and pathways known to be modulated by coronavirus infection(26, 66). Notably, in this regard, cell cycle progression/proliferation, PI3K/AKT/mTOR, innate immunity and inflammation are well represented among the primary target proteins for those pharmacologic agents strongly predicted by ViroTreat to possess host cell-targeted, antiviral effects.

A literature search revealed that many of the oncology drugs and compounds identified by ViroTreat have been considered previously for their potential antiviral effects. For instance, the MAPK inhibitor trametinib, one of the top ViroTreat hits for SARS-CoV, was shown to inhibit MERS-CoV replication in vitro(5, 20), as well as influenza A virus both in vitro and in vivo(67). Similarly, everolimus, an mTOR inhibitor identified by ViroTreat, has also been shown to inhibit MERS-CoV(5, 20) and cytomegalovirus(68) replication in vitro, as well as to reduce incidence of cytomegalovirus infections following kidney transplant(69). Among tyrosine kinase inhibitors identified by ViroTreat, dasatinib was previously described to inhibit MERS-CoV(5, 19) and HIV-1(70) replication in vitro; while erlotinib was shown to inhibit dengue(71), hepatitis C(72) and ebola(73) replication. The HSP90 inhibitors SNX-2112 and luminespib, as well as the sarco/endoplasmatic reticulum Ca2+ ATPase inhibitor thapsigargin, all identified by ViroTreat as inverters of the SARS- CoV induced checkpoint, have been shown to inhibit herpes simplex(74), chikungunya(75), foot and mouth disease virus(76), respiratory syncytial virus(77), rhinovirus(78) and hepatitis A virus replication(79).

Finally, ViroTreat independently identified the SINE drug molecule selinexor—an FDA-approved agent for the treatment of relapsed or refractory multiple myeloma—as an extremely potent inverter of SARS-CoV-induced ViroCheckpoint activity. Selinexor is a potent and highly-specific inhibitor of XPO1 activity, which leads to nuclear retention of its cargo proteins containing leucine rich Nuclear Export Signals. Based on experimental studies performed by Karyopharm Therapeutics Inc., low Selinexor concentrations (leq 100 nM) inhibited viral replication by 90% in green monkey kidney Vero cells infected with SARS-CoV-2(80). As a result of these observations and data, which are consistent with the ViroTreat prioritization of selinexor we report in this study, a randomized, placebo-controlled Phase 2 clinical study (NCT04355676 and NCT04349098), evaluating low dose oral selinexor in hospitalized patients with severe COVID-19 has been initiated and is currently enrolling patients, with results anticipated to be reported by August 31st, 2020(80).

This analysis has several limitations that partially restrict its value as proof of concept. Specifically, infection was conducted in a cancer cell line, rather than in a more physiologically relevant context, such as in primary bronchial or alveolar epithelial cells. In addition, drug perturbations were also performed in a cancer cell line context, thus potentially introducing undesired confounding factors, even though use of mock controls for the infection, and vehicle control for the drug perturbations, from the same cancer cell line should have eliminated most of the cancer-related bias and cell line idiosyncrasies. As a result, extrapolation of this approach to the clinic may be limited by the following assumptions: (a) that the host cell regulatory checkpoint hijacked by the virus is conserved between the Calu-3 adenocarcinoma cell line and the normal alveolar or bronchial epithelial cells in vivo; and (2) that the drugs’ and compounds’ MoA is conserved between the NCI-H1793 lung adenocarcinoma cells and normal lung epithelial cells in vivo. Moreover, while for the generation of the perturbational data and the context-specific MoA database we used subtoxic drug concentrations that, in most cases, were well below the maximum tolerated dose for all drugs and compounds, the relevant pharmacologic concentration for their deployment as antiviral therapy may be much lower than the original recommended concentration for their use as anti-cancer drugs.

Further research is necessary to benchmark the ViroTreat approach. Specifically, better reporters of SARS-CoV infection should be established, ideally directly from nasopharyngeal swabs or bronchial lavage of SARS-CoV patients. More relevant to the current pandemic, such samples are starting to emerge from COVID-19 patients and may lead to elucidation of critical entry points for COVID-19 therapeutic intervention. Similarly, drug profiles should be generated in a more physiologic context, including primary airway epithelial cells. It is also important to establish whether virus-induced transcriptional lock states are similar across all cell and tissue contexts infected by the virus, or whether the hijacked states are cell context-specific. Finally, appropriate environments for in vitro and in vivo validation of prioritized drugs should be developed(56).

To our knowledge, this is the first time a virus-induced MR module (i.e., the ViroCheckpoint) is proposed as a pharmacological target to abrogate the virus’s ability to hijack the cellular machinery of host cells, a strategy that coronaviruses are known to employ to prime the host cell environment so it is amenable to viral replication and release(26). In addition, ViroTreat represents a unique method for the systematic and quantitative prioritization of mechanism-based, host-directed drugs capable of abrogating this critical, and previously unaddressed component of viral infection. If effectively validated, this approach presents several advantages: First, ViroTreat is tailored to target the entire repertoire of host proteins hijacked by the virus to create a permissive environment, rather than a single host or viral protein. As such, we anticipate drugs identified by ViroTreat to have more universal applications, including being effective against a broader viral repertoire, while also being more effective at eluding virus adaptation mechanisms arising from rapid mutation under drug selection stress. Indeed, drug-mediated reprogramming of host cell to a transcriptional state that confers resistance against coronavirus-induced reprogramming presents the opportunity to identify drugs that are potentially effective for a broader class of viruses, as long as they share similar pathobiological strategies for host cell takeover. Second, the ViroTreat analysis can be performed expeditiously—as soon as the ViroCheckpoint signature of a novel virus becomes available. Therefore, this methodology is especially well-suited to the urgency characteristic of epidemics and pandemics.

Developing effective treatments for respiratory tract infections—i.e., those that reduce such hard end points as hospitalization, need for mechanical ventilation, and mortality—exclusively through direct viral targeting has been historically challenging. Drugs identified specifically for host cell-targeting have the potential therapeutic advantage of acting in a mechanistically complementary—even synergistic—way with readily available antivirals, thereby suggesting roadmaps for developing and testing combination regimens that may mitigate viral replication by acting upon the infected system as a whole. Such multi-mechanistic pharmacologic approaches targeting both the virus and host cell proteins that are critically dysregulated as a result of viral infection may be required to optimize clinical outcomes, especially in challenging and vulnerable patients exposed to lethal pathogens with high virulence and viral load.

Methods

Cell lines.

NCI-H1793 cells were obtained from ATCC (CRL-5896), mycoplasm tested and maintained in DMEM:F12 medium supplemented with 5 μg/ml insulin, 10 μg/ml transferrin, 30 nM sodium selenite, 10 nM -estradiol, 4.5 mM L-glutamine and 5% fetal bovine serum. Cells were grown in a humidified incubator at 37°C and 5% CO2.

Lung epithelium context-specific drug mechanism of action database.

The drug-perturbation dataset was generated as follows. First, the ED20 for each of the 133 FDA-approved drugs and 195 investigational compounds in oncology was estimated in NCI-H1793 cells by performing 10-point dose-response curves in triplicate, using total ATP content as read-out. Briefly, 2,000 cells per well were plated in 384-well plates. Small-molecule compounds were added with a 96-well pin-tool head 12h after cell plating. Viable cells were quantified 48h later by ATP assay (CellTiterGlo, Promega). Relative cell viability was computed using matched DMSO control wells as reference. ED20 was estimated by fitting a four-parameter sigmoid model to the titration results. NCI-H1793 cells, plated in 384-well plates, were then perturbed with a library of 328 FDA-approved drugs and small-molecule compounds at their corresponding ED20 concentration. Cells were lysed at 24h after small-molecule compound perturbation and the transcriptome was profiled by PLATE-Seq(55). RNA-Seq reads were mapped to the human reference genome assembly 38 using the STAR aligner(81). Expression data were then normalized by equivariance transformation, based on the negative binomial distribution with the DESeq R-system package (Bioconductor(82)). At least two replicates for each condition were obtained. Differential gene expression signatures were computed by comparing each condition with plate-matched vehicle control samples using a moderated Student’s t-test as implemented in the limma package from Bioconductor(83). Individual gene expression signatures were then transformed into protein activity signatures with the VIPER algorithm(37), based on the a lung adenocarcinoma context-specific regulatory network available from the aracne.networks package from Bioconductor.

Computational analysis.

Enrichment of gene-sets for biological hallmarks was performed using Gene Set Enrichment Analysis(52) with the Molecular Signatures Database MSigDB v7.1(53). Enrichment analysis for virus-interacting host proteins (PPI) on SARS-CoV induced protein activity signatures, as well as the OncoMatch(56) analysis to assess the conservation of the virus-induced MR protein activity on NCI-H1793 lung adenocarcinoma cells were performed with the aREA algorithm(37).

ViroTreat analysis.

ViroTreat was performed by computing the enrichment of the top/bottom 50 most differentially active proteins in response to drug perturbation—the context-specific mechanism of action—on the virus-induced protein activity signature using the aREA algorithm(37). P-values for significantly negative enrichment were estimated using 1-tail aREA analysis, and multiple hypothesis testing was controlled by the Bonferroni’s correction.

Code availability.

All the code used in this work is freely available for research purposes. VIPER and aREA algorithms are part of the “viper” R-system’s package available from Bioconductor. The lung adenocarcinoma context-specific interactome is available as part of the “aracne.networks” R-system’s package from Bioconductor.

Supplementary Material

1

ACKNOWLEDGEMENTS

We thank Christopher Walker for reviewing selinexor data accuracy and Tatiana Alvarez for original artwork. This research was supported by the following NIH grants to Andrea Califano: R35 CA197745 (Outstanding Investigator Award); U01 CA217858 (Cancer Target Discovery and Development); S10 OD012351 and S10 OD021764 (Shared Instrument Grants).

Footnotes

Competing Financial Interests Statement. P.L. is Director of Single-Cell Systems Biology at DarwinHealth, Inc., a company that has licensed some of the algorithms used in this manuscript from Columbia University. G.B. is founder, CEO and equity holder of DarwinHealth, Inc. X.S. is Senior Computational Biologist at DarwinHealth, Inc. A.C. is founder, equity holder, consultant, and director of DarwinHealth Inc. M.J.A. is CSO and equity holder of DarwinHealth, Inc. Columbia University is also an equity holder in DarwinHealth Inc.

Bibliography

  • 1.Peiris Joseph S.M., Yuen Kwok Y., Osterhaus Albert D.M.E., and Stöhr Klaus. The Severe Acute Respiratory Syndrome. New England Journal of Medicine, 349(25):2431–2441, December 2003. ISSN 0028–4793. doi: 10.1056/NEJMra032498 [DOI] [PubMed] [Google Scholar]
  • 2.Zhu Na, Zhang Dingyu, Wang Wenling, Li Xingwang, Yang Bo, Song Jingdong, Zhao Xiang, Huang Baoying, Shi Weifeng, Lu Roujian, Niu Peihua, Zhan Faxian, Ma Xuejun, Wang Dayan, Xu Wenbo, Wu Guizhen, Gao George F., Tan Wenjie, and China Novel Coronavirus Investigating and Research Team. A Novel Coronavirus from Patients with Pneumonia in China, 2019. The New England journal of medicine, 382(8):727–733, 2020. ISSN 1533–4406. doi: 10.1056/NEJMoa2001017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Nicholls John M, Poon Leo LM, Lee Kam C, Ng Wai F, Lai Sik T, Leung Chung Y, Chu Chung M, Hui Pak K, Mak Kong L, Lim Wilna, Yan Kin W, Chan Kwok H, Tsang Ngai C, Guan Yi, Yuen Kwok Y, and Peiris JS Malik. Lung pathology of fatal severe acute respiratory syndrome. The Lancet, 361(9371):1773–1778, May 2003. ISSN 01406736. doi: 10.1016/S0140-6736(03)13413-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Li Wenhui, Zhang Chengsheng, Sui Jianhua, Kuhn Jens H, Moore Michael J, Luo Shiwen, Wong Swee-Kee, Huang I-Chueh, Xu Keming, Vasilieva Natalya, Murakami Akikazu, He Yaqing, Marasco Wayne A, Guan Yi, Choe Hyeryun, and Farzan Michael. Receptor and viral determinants of SARS-coronavirus adaptation to human ACE2. The EMBO Journal, 24(8): 1634–1643, April 2005. ISSN 0261–4189. doi: 10.1038/sj.emboj.7600640 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Zumla Alimuddin, Chan Jasper F W, Azhar Esam I., Hui David S C, and Yuen Kwok-Yung. Coronaviruses - drug discovery and therapeutic options. Nature reviews. Drug discovery, 15(5):327–47, May 2016. ISSN 1474–1784. doi: 10.1038/nrd.2015.37 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Sanders James M, Monogue Marguerite L, Jodlowski Tomasz Z, and Cutrell James B. Pharmacologic Treatments for Coronavirus Disease 2019 (COVID-19): A Review. JAMA, April 2020. ISSN 1538–3598. doi: 10.1001/jama.2020.6019 [DOI] [PubMed] [Google Scholar]
  • 7.NIAID. NIH Clinical Trial Shows Remdesivir Accelerates Recovery from Advanced COVID-19, 2020.
  • 8.Báez-Santos Yahira M., St. John Sarah E., and Mesecar Andrew D.. The SARS-coronavirus papain-like protease: Structure, function and inhibition by designed antiviral compounds. Antiviral Research, 115:21–38, March 2015. ISSN 01663542. doi: 10.1016/j.antiviral.2014.12.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Li Guangdi and De Clercq Erik. Therapeutic options for the 2019 novel coronavirus (2019-nCoV). Nature reviews. Drug discovery, 19(3):149–150, 2020. ISSN 1474–1784. doi: 10.1038/d41573-020-00016-0 [DOI] [PubMed] [Google Scholar]
  • 10.Adedeji Adeyemi O. and Sarafianos Stefan G.. Antiviral drugs specific for coronaviruses in preclinical development. Current Opinion in Virology, 8:45–53, 2014. ISSN 18796265. doi: 10.1016/j.coviro.2014.06.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Dharan Nila J.. Infections With Oseltamivir-Resistant Influenza A(H1N1) Virus in the United States. JAMA, 301(10):1034, March 2009. ISSN 0098–7484. doi: 10.1001/jama.2009.294 [DOI] [PubMed] [Google Scholar]
  • 12.Lee Nelson and Hurt Aeron C.. Neuraminidase inhibitor resistance in influenza. Current Opinion in Infectious Diseases, 31(6):520–526, December 2018. ISSN 0951–7375. doi: 10.1097/QCO.0000000000000498 [DOI] [PubMed] [Google Scholar]
  • 13.van de Wakker Simonides I., Fischer Marcel J E, and Oosting Ronald S.. New drug-strategies to tackle viral-host interactions for the treatment of influenza virus infections. European journal of pharmacology, 809:178–190, August 2017. ISSN 1879–0712. doi: 10.1016/j.ejphar.2017.05.038 [DOI] [PubMed] [Google Scholar]
  • 14.Han Dong P., Penn-Nicholson Adam, and Cho Michael W.. Identification of critical determinants on ACE2 for SARS-CoV entry and development of a potent entry inhibitor. Virology, 350(1):15–25, June 2006. ISSN 00426822. doi: 10.1016/j.virol.2006.01.029 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Zhou Yanchen, Vedantham Punitha, Lu Kai, Agudelo Juliet, Carrion Ricardo, Nunneley Jerritt W., Barnard Dale, Pöhlmann Stefan, McKerrow James H., Renslo Adam R., and Simmons Graham. Protease inhibitors targeting coronavirus and filovirus entry. Antiviral Research, 116:76–84, April 2015. ISSN 01663542. doi: 10.1016/j.antiviral.2015.01.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.de Wilde Adriaan H, Jochmans Dirk, Posthuma Clara C., Zevenhoven-Dobbe Jessika C., van Nieuwkoop Stefan, Bestebroer Theo M., van den Hoogen Bernadette G, Neyts Johan, and Snijder Eric J.. Screening of an FDA-approved compound library identifies four small-molecule inhibitors of Middle East respiratory syndrome coronavirus replication in cell culture. Antimicrobial agents and chemotherapy, 58(8):4875–84, August 2014. ISSN 1098–6596. doi: 10.1128/AAC.03011-14 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Xu Miao, Lee Emily M., Wen Zhexing, Cheng Yichen, Huang Wei-Kai, Qian Xuyu, Tcw Julia, Kouznetsova Jennifer, Ogden Sarah C., Hammack Christy, Jacob Fadi, Ha Nam Nguyen Misha Itkin, Hanna Catherine, Shinn Paul, Allen Chase, Michael Samuel G., Simeonov Anton, Huang Wenwei, Christian Kimberly M., Goate Alison, Brennand Kristen J., Huang Ruili, Xia Menghang, Ming Guo-Li, Zheng Wei, Song Hongjun, and Tang Hengli. Identification of small-molecule inhibitors of Zika virus infection and induced neural cell death via a drug repurposing screen. Nature medicine, 22(10):1101–1107, October 2016. ISSN 1546–170X. doi: 10.1038/nm.4184 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Johansen Lisa M., Brannan Jennifer M., Delos Sue E., Shoemaker Charles J., Stossel Andrea, Lear Calli, Hoffstrom Benjamin G., Dewald Lisa Evans, Schornberg Kathryn L., Scully Corinne, Lehár Joseph, Hensley Lisa E., White Judith M., and Olinger Gene G.. FDA-approved selective estrogen receptor modulators inhibit Ebola virus infection. Science translational medicine, 5(190):190ra79, June 2013. ISSN 1946–6242. doi: 10.1126/scitranslmed.3005471 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Dyall Julie, Coleman Christopher M., Hart Brit J., Venkataraman Thiagarajan, Holbrook Michael R., Kindrachuk Jason, Johnson Reed F., Olinger Gene G., Jahrling Peter B., Laidlaw Monique, Johansen Lisa M., Lear-Rooney Calli M., Glass Pamela J., Hensley Lisa E., and Frieman Matthew B.. Repurposing of clinically developed drugs for treatment of Middle East respiratory syndrome coronavirus infection. Antimicrobial agents and chemotherapy, 58(8): 4885–93, August 2014. ISSN 1098–6596. doi: 10.1128/AAC.03036-14 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Kindrachuk Jason, Ork Britini, Hart Brit J., Mazur Steven, Holbrook Michael R., Frieman Matthew B., Traynor Dawn, Johnson Reed F., Dyall Julie, Kuhn Jens H., Olinger Gene G., Hensley Lisa E, and Jahrling Peter B. Antiviral potential of ERK/MAPK and PI3K/AKT/mTOR signaling modulation for Middle East respiratory syndrome coronavirus infection as identified by temporal kinome analysis. Antimicrobial agents and chemotherapy, 59(2):1088–99, February 2015. ISSN 1098–6596. doi: 10.1128/AAC.03659-14 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Gordon David E, Jang Gwendolyn M, Bouhaddou Mehdi, Xu Jiewei, Obernier Kirsten, White Kris M., O’Meara Matthew J., Rezelj Veronica V, Guo Jeffrey Z, Swaney Danielle L, Tummino Tia A., Huettenhain Ruth, Kaake Robyn M, Richards Alicia L, Tutuncuoglu Beril, Foussard Helene, Batra Jyoti, Haas Kelsey, Modak Maya, Kim Minkyu, Haas Paige, Polacco Benjamin J., Braberg Hannes, Fabius Jacqueline M, Eckhardt Manon, Soucheray Margaret, Bennett Melanie J, Cakir Merve, McGregor Michael J., Li Qiongyu, Meyer Bjoern, Roesch Ferdinand, Vallet Thomas, Kain Alice Mac, Miorin Lisa, Moreno Elena, Chi Naing Zun Zar, Zhou Yuan, Peng Shiming, Shi Ying, Zhang Ziyang, Shen Wenqi, Kirby Ilsa T, Melnyk James E, Chorba John S., Lou Kevin, Dai Shizhong A., Barrio-Hernandez Inigo, Memon Danish, Hernandez-Armenta Claudia, Lyu Jiankun, Mathy Christopher J. P., Perica Tina, Pilla Kala B., Ganesan Sai J., Saltzberg Daniel J., Rakesh Ramachandran, Liu Xi, Rosenthal Sara B., Calviello Lorenzo, Venkataramanan Srivats, Liboy-Lugo Jose, Lin Yizhu, Huang Xi-Ping, Liu YongFeng, Wankowicz Stephanie A., Bohn Markus, Safari Maliheh, Ugur Fatima S., Koh Cassandra, Savar Nastaran Sadat, Tran Quang Dinh, Shengjuler Djoshkun, Fletcher Sabrina J, O’Neal Michael C., Cai Yiming, Chang Jason C J, Broadhurst David J, Klippsten Saker, Sharp Phillip P, Wenzell Nicole A., Kuzuoglu Duygu, Wang Hao-Yuan, Trenker Raphael, Young Janet M., Cavero Devin A., Hiatt Joseph, Roth Theodore L, Rathore Ujjwal, Subramanian Advait, Noack Julia, Hubert Mathieu, Stroud Robert M., Frankel Alan D., Rosenberg Oren S., Verba Kliment A, Agard David A., Ott Melanie, Emerman Michael, Jura Natalia, von Zastrow Mark, Verdin Eric, Ashworth Alan, Schwartz Olivier, D’Enfert Christophe, Mukherjee Shaeri, Jacobson Matt, Malik Harmit S, Fujimori Danica G, Ideker Trey, Craik Charles S, Floor Stephen N, Fraser James S., Gross John D, Sali Andrej, Roth Bryan L, Ruggero Davide, Taunton Jack, Kortemme Tanja, Beltrao Pedro, Vignuzzi Marco, García-Sastre Adolfo, Shokat Kevan M, Shoichet Brian K., and Krogan Nevan J.. A SARS-CoV-2 protein interaction map reveals targets for drug repurposing. Nature, April 2020. ISSN 0028–0836. doi: 10.1038/s41586-020-2286-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Zhou Yadi, Hou Yuan, Shen Jiayu, Huang Yin, Martin William, and Cheng Feixiong. Network-based drug repurposing for novel coronavirus 2019-nCoV/SARS-CoV-2. Cell discovery, 6:14, 2020. ISSN 2056–5968. doi: 10.1038/s41421-020-0153-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Pfefferle Susanne, Schöpf Julia, Kögl Manfred, Friedel Caroline C., Müller Marcel A., Carbajo-Lozoya Javier, Stellberger Thorsten, von Dall’Armi Ekatarina, Herzog Petra, Kallies Stefan, Niemeyer Daniela, Ditt Vanessa, Kuri Thomas, Züst Roland, Pumpor Ksenia, Hilgenfeld Rolf, Schwarz Frank, Zimmer Ralf, Steffen Imke, Weber Friedemann, Thiel Volker, Herrler Georg, Thiel Heinz-Jürgen, Schwegmann-Wessels Christel, Pöhlmann Stefan, Haas Jürgen, Drosten Christian, and von Brunn Albrecht. The SARS-coronavirus-host interactome: identification of cyclophilins as target for pan-coronavirus inhibitors. PLoS pathogens, 7(10):e1002331, October 2011. ISSN 1553–7374. doi: 10.1371/journal.ppat.1002331 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Mitchell Hugh D., Eisfeld Amie J., Sims Amy C., McDermott Jason E., Matzke Melissa M., Webb-Robertson Bobbi-Jo M, Tilton Susan C., Tchitchek Nicolas, Josset Laurence, Li Chengjun, Ellis Amy L., Chang Jean H., Heegel Robert A., Luna Maria L., Schepmoes Athena A., Shukla Anil K., Metz Thomas O., Neumann Gabriele, Benecke Arndt G., Smith Richard D., Baric Ralph S., Kawaoka Yoshihiro, Katze Michael G., and Waters Katrina M.. A network integration approach to predict conserved regulators related to pathogenicity of influenza and SARS-CoV respiratory viruses. PloS one, 8(7):e69374, 2013. ISSN 1932–6203. doi: 10.1371/journal.pone.0069374 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Stucki Janick D, Nina Hobi, Galimov Artur, Stucki Andreas O., Schneider-Daum Nicole, Lehr Claus-Michael, Huwer Hanno, Frick Manfred, Funke-Chambour Manuela, Geiser Thomas, and Guenat Olivier T.. Medium throughput breathing human primary cell alveolus-on-chip model. Scientific reports, 8(1):14359, December 2018. ISSN 2045–2322. doi: 10.1038/s41598-018-32523-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.de Wilde Adriaan H, Snijder Eric J, Kikkert Marjolein, and van Hemert Martijn J. Host Factors in Coronavirus Replication. Current topics in microbiology and immunology, 419 (October):1–42, 2012. ISSN 0070–217X. doi: 10.1007/82_2017_25 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Kushwaha Ritu, Jagadish Nirmala, Kustagi Manjunath, Tomishima Mark J., Mendiratta Geetu, Bansal Mukesh, Kim Hyunjae R., Sumazin Pavel, Alvarez Mariano J., Lefebvre Celine, Patricia Villagrasa-Gonzalez Agnes Viale, Korkola James E., Houldsworth Jane, Feldman Darren R., Bosl George J., Califano Andrea, and Chaganti R. S. K.. Interrogation of a context-specific transcription factor network identifies novel regulators of pluripotency. Stem cells (Dayton, Ohio), 33(2):367–77, February 2015. ISSN 1549–4918. doi: 10.1002/stem.1870 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Lefebvre Celine, Rajbhandari Presha, Mariano J Alvarez Pradeep Bandaru, Wei Keat Lim Mai Sato, Wang Kai, Sumazin Pavel, Kustagi Manjunath, Brygida C Bisikirska Katia Basso, Beltrao Pedro, Krogan Nevan, Gautier Jean, Dalla-Favera Riccardo, and Califano Andrea. A human B-cell interactome identifies MYB and FOXM1 as master regulators of proliferation in germinal centers. Molecular systems biology, 6(377):377, June 2010. ISSN 1744–4292. doi: 10.1038/msb.2010.31 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Son Jinsook, Ding Hongxu, Accii Domenico, and Califano Andrea. AFF3 and BACH2 are master regulators of metabolic inflexibility, beta/alpha-cell transition, and dedifferentiation in type 2 diabetes. bioRxiv, page 768135, September 2019. doi: 10.1101/768135 [DOI] [Google Scholar]
  • 30.Califano Andrea and Alvarez Mariano J. The recurrent architecture of tumour initiation, progression and drug sensitivity. Nature reviews. Cancer, 17(2):116–130, December 2017. ISSN 1474–1768. doi: 10.1038/nrc.2016.124 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Carro Maria Stella, Lim Wei Keat, Alvarez Mariano Javier, Bollo Robert J, Zhao Xudong, Snyder Evan Y, Sulman Erik P, Anne Sandrine L, Doetsch Fiona, Colman Howard, Lasorella Anna, Aldape Ken, Califano Andrea, and Iavarone Antonio. The transcriptional network for mesenchymal transformation of brain tumours. Nature, 463(7279):318–25, January 2010. ISSN 1476–4687. doi: 10.1038/nature08712 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Rajbhandari Presha, Lopez Gonzalo, Capdevila Claudia, Salvatori Beatrice, Yu Jiyang, Rodriguez-Barrueco Ruth, Martinez Daniel, Yarmarkovich Mark, Weichert-Leahey Nina, Abraham Brian J., Alvarez Mariano J., Iyer Archana, Harenza Jo Lynne, Oldridge Derek, De Preter Katleen, Koster Jan, Asgharzadeh Shahab, Seeger Robert C., Wei Jun S., Khan Javed, Vandesompele Jo, Mestdagh Pieter, Versteeg Rogier, Look A. Thomas, Young Richard A., Iavarone Antonio, Lasorella Anna, Silva Jose M., Maris John M., and Califano Andrea. Cross-Cohort Analysis Identifies a TEAD4-MYCN Positive Feedback Loop as the Core Regulatory Element of High-Risk Neuroblastoma. Cancer discovery, 8(5):582–599, May 2018. ISSN 2159–8290. doi: 10.1158/2159-8290.CD-16-0861 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Aytes Alvaro, Mitrofanova Antonina, Lefebvre Celine, Alvarez Mariano J., Castillo-Martin Mireia, Zheng Tian, Eastham James A., Gopalan Anuradha, Pienta Kenneth J., Shen Michael M., Califano Andrea, and Abate-Shen Cory. Cross-species regulatory network analysis identifies a synergistic interaction between FOXM1 and CENPF that drives prostate cancer malignancy. Cancer cell, 25(5):638–51, May 2014. ISSN 1878–3686. doi: 10.1016/j.ccr.2014.03.017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Alvarez Mariano J, Subramaniam Prem S, Tang Laura H, Grunn Adina, Aburi Mahalaxmi, Rieckhof Gabrielle, Komissarova Elena V, Hagan Elizabeth A, Bodei Lisa, Clemons Paul A, Dela Cruz Filemon S, Dhall Deepti, Diolaiti Daniel, Fraker Douglas A, Ghavami Afshin, Kaemmerer Daniel, Karan Charles, Kidd Mark, Kim Kyoung M, Kim Hee C, Kunju Lakshmi P, Langel Ülo, Li Zhong, Lee Jeeyun, Li Hai, LiVolsi Virginia, Pfragner Roswitha, Rainey Allison R, Realubit Ronald B, Remotti Helen, Regberg Jakob, Roses Robert, Rustgi Anil, Sepulveda Antonia R, Serra Stefano, Shi Chanjuan, Yuan Xiaopu, Barberis Massimo, Bergamaschi Roberto, Chinnaiyan Arul M, Detre Tony, Ezzat Shereen, Frilling Andrea, Hommann Merten, Jaeger Dirk, Kim Michelle K, Knudsen Beatrice S, Kung Andrew L, Leahy Emer, Metz David C, Milsom Jeffrey W, Park Young S, Reidy-Lagunes Diane, Schreiber Stuart, Washington Kay, Wiedenmann Bertram, Modlin Irvin, and Califano Andrea. A precision oncology approach to the pharmacological targeting of mechanistic dependencies in neuroendocrine tumors. Nature genetics, 50(7):979–989, July 2018. ISSN 1546–1718. doi: 10.1038/s41588-018-0138-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Ikiz Burcin, Alvarez Mariano J, Ré Diane B, Le Verche Virginia, Politi Kristin, Lotti Francesco, Phani Sudarshan, Pradhan Radhika, Yu Changhao, Croft Gist F, Jacquier Arnaud, Henderson Christopher E, Califano Andrea, and Przedborski Serge. The Regulatory Machinery of Neurodegeneration in In Vitro Models of Amyotrophic Lateral Sclerosis. Cell reports, 12(2): 335–45, July 2015. ISSN 2211–1247. doi: 10.1016/j.celrep.2015.06.019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Brichta Lars, Shin William, Jackson-Lewis Vernice, Blesa Javier, Yap Ee-Lynn, Walker Zachary, Zhang Jack, Roussarie Jean-Pierre, Alvarez Mariano J, Califano Andrea, Przedborski Serge, and Greengard Paul. Identification of neurodegenerative factors using translatome-regulatory network analysis. Nature neuroscience, 18(9):1325–33, September 2015. ISSN 1546–1726. doi: 10.1038/nn.4070 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Alvarez Mariano J, Shen Yao, Giorgi Federico M, Lachmann Alexander, Ding B Belinda, Ye B Hilda, and Califano Andrea. Functional characterization of somatic mutations in cancer using network-based inference of protein activity. Nature genetics, 48(8):838–47, August 2016. ISSN 1546–1718. doi: 10.1038/ng.3593 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Columbia University. Darwin OncoTarget and OncoTreat, 2018.
  • 39.Yoshikawa Tomoki, Hill Terence E., Yoshikawa Naoko, Popov Vsevolod L., Galindo Cristi L., Garner Harold R., Peters C. J., and Tseng Chien-Te Kent. Dynamic innate immune responses of human bronchial epithelial cells to severe acute respiratory syndrome-associated coronavirus infection. PloS one, 5(1):e8729, January 2010. ISSN 1932–6203. doi: 10.1371/journal.pone.0008729 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Walsh Logan A., Alvarez Mariano J., Sabio Erich Y., Reyngold Marsha, Makarov Vladimir, Mukherjee Suranjit, Lee Ken-Wing, Desrichard Alexis, Turcan Sevin, Dalin Martin G., Rajasekhar Vinagolu K., Chen Shuibing, Vahdat Linda T., Califano Andrea, and Chan Timothy A.. An Integrated Systems Biology Approach Identifies TRIM25 as a Key Determinant of Breast Cancer Metastasis. Cell reports, 20(7):1623–1640, August 2017. ISSN 2211–1247. doi: 10.1016/j.celrep.2017.07.052 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Repunte-Canonigo Vez, Shin William, Vendruscolo Leandro F, Lefebvre Celine, van der Stap Lena, Kawamura Tomoya, Schlosburg Joel E, Alvarez Mariano, Koob George F, Califano Andrea, and Sanna Pietro Paolo. Identifying candidate drivers of alcohol dependence-induced excessive drinking by assembly and interrogation of brain-specific regulatory networks. Genome biology, 16(1):68, 2015. ISSN 1474–760X. doi: 10.1186/s13059-015-0593-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Ding Hongxu, Douglass Eugene F., Sonabend Adam M., Mela Angeliki, Bose Sayantan, Gonzalez Christian, Canoll Peter D., Sims Peter A., Alvarez Mariano J., and Califano Andrea. Quantitative assessment of protein activity in orphan tissues and single cells using the metaVIPER algorithm. Nature communications, 9(1):1471, April 2018. ISSN 2041–1723. doi: 10.1038/s41467-018-03843-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Paull Evan O., Aytes Alvaro, Subramaniam Prem, Giorgi Federico M., Douglass Eugene F., Chu Brennan, Jones Sunny J., Zheng Siyuan, Verhaak Roel, Abate-Shen Cory, Alvarez Mariano J., and Califano Andrea. A Modular Master Regulator Landscape Determines the Impact of Genetic Alterations on the Transcriptional Identity of Cancer Cells. bioRxiv, page 758268, 2019. doi: 10.1101/758268 [DOI] [Google Scholar]
  • 44.Chari Ajai, Vogl Dan T, Gavriatopoulou Maria, Nooka Ajay K, Yee Andrew J, Huff Carol A, Moreau Philippe, Dingli David, Cole Craig, Lonial Sagar, Dimopoulos Meletios, Stewart A Keith, Richter Joshua, Vij Ravi, Tuchman Sascha, Raab Marc S, Weisel Katja C, Delforge Michel, Cornell Robert F, Kaminetzky David, Hoffman James E, Costa Luciano J, Parker Terri L, Levy Moshe, Schreder Martin, Meuleman Nathalie, Frenzel Laurent, Mohty Mohamad, Choquet Sylvain, Schiller Gary, Comenzo Raymond L, Engelhardt Monika, Illmer Thomas, Vlummens Philip, Doyen Chantal, Facon Thierry, Karlin Lionel, Perrot Aurore, Podar Klaus, Kauffman Michael G, Shacham Sharon, Li Lingling, Tang Shijie, Picklesimer Carla, Saint-Martin Jean-Richard, Crochiere Marsha, Chang Hua, Parekh Samir, Landesman Yosef, Shah Jatin, Richardson Paul G, and Jagannath Sundar. Oral Selinexor-Dexamethasone for Triple-Class Refractory Multiple Myeloma. The New England journal of medicine, 381(8):727–738, August 2019. ISSN 1533–4406. doi: 10.1056/NEJMoa1903455 [DOI] [PubMed] [Google Scholar]
  • 45.Basso Katia, Margolin Adam a, Stolovitzky Gustavo, Klein Ulf, Dalla-Favera Riccardo, and Califano Andrea. Reverse engineering of regulatory networks in human B cells. Nature genetics, 37(4):382–90, April 2005. ISSN 1061–4036. doi: 10.1038/ng1532 [DOI] [PubMed] [Google Scholar]
  • 46.Bertoli Cosetta, Skotheim Jan M., and de Bruin Robertus A. M.. Control of cell cycle transcription during G1 and S phases. Nature Reviews Molecular Cell Biology, 14(8):518–528, August 2013. ISSN 1471–0072. doi: 10.1038/nrm3629 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Wang James C.. Cellular roles of DNA topoisomerases: a molecular perspective. Nature Reviews Molecular Cell Biology, 3(6):430–440, June 2002. ISSN 1471–0072. doi: 10.1038/nrm831 [DOI] [PubMed] [Google Scholar]
  • 48.Bartek Jiri and Lukas Jiri. Mammalian G1- and S-phase checkpoints in response to DNA damage. Current Opinion in Cell Biology, 13(6):738–747, December 2001. ISSN 09550674. doi: 10.1016/S0955-0674(00)00280-5 [DOI] [PubMed] [Google Scholar]
  • 49.Löbrich Markus and Jeggo Penny A.. The impact of a negligent G2/M checkpoint on genomic instability and cancer induction. Nature Reviews Cancer, 7(11):861–869, November 2007. ISSN 1474–175X. doi: 10.1038/nrc2248 [DOI] [PubMed] [Google Scholar]
  • 50.Levy David E, Marié Isabelle J, and Durbin Joan E. Induction and function of type I and III interferon in response to viral infection. Current Opinion in Virology, 1(6):476–486, December 2011. ISSN 18796257. doi: 10.1016/j.coviro.2011.11.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Bisikirska Brygida, Bansal Mukesh, Shen Yao, Teruya-Feldstein Julie, Chaganti Raju, and Califano Andrea. Elucidation and Pharmacological Targeting of Novel Molecular Drivers of Follicular Lymphoma Progression. Cancer research, 76(3):664–74, February 2016. ISSN 1538–7445. doi: 10.1158/0008-5472.CAN-15-0828 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Subramanian Aravind, Tamayo Pablo, Mootha Vamsi K, Mukherjee Sayan, Ebert Benjamin L, Gillette Michael a, Paulovich Amanda, Pomeroy Scott L, Golub Todd R, Lander Eric S, and Mesirov Jill P. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proceedings of the National Academy of Sciences of the United States of America, 102(43):15545–50, October 2005. ISSN 0027–8424. doi: 10.1073/pnas.0506580102 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Liberzon Arthur, Birger Chet, Thorvaldsdóttir Helga, Ghandi Mahmoud, Mesirov Jill P., and Tamayo Pablo. The Molecular Signatures Database Hallmark Gene Set Collection. Cell Systems, 1(6):417–425, December 2015. ISSN 24054712. doi: 10.1016/j.cels.2015.12.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Columbia University. Columbia Precision Oncology Initiative, 2018.
  • 55.Bush Erin C., Ray Forest, Alvarez Mariano J., Realubit Ronald, Li Hai, Karan Charles, Califano Andrea, and Sims Peter A.. PLATE-Seq for genome-wide regulatory network analysis of high-throughput screens. Nature communications, 8(1):105, July 2017. ISSN 2041–1723. doi: 10.1038/s41467-017-00136-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Alvarez Mariano J, Yan Pengrong, Alpaugh Mary L, Bowden Michaela, Sicinska Ewa, Zhou Chensheng W, Karan Charles, Realubit Ronald B, Mundi Prabhjot S, Grunn Adina, Jäger Dirk, Chabot John A, Fojo Antonio T, Oberstein Paul E, Hibshoosh Hanina, Milsom Jeffrey W, Kulke Matthew H, Loda Massimo, Chiosis Gabriela, Reidy-Lagunes Diane L, and Califano Andrea. Unbiased Assessment of H-STS cells as high-fidelity models for gastro-enteropancreatic neuroendocrine tumor drug mechanism of action analysis. bioRxiv, page 677435, June 2019. doi: 10.1101/677435 [DOI] [Google Scholar]
  • 57.Bhardwaj Kanchan, Liu Pinghua, Leibowitz Julian L, and Kao C Cheng. The coronavirus endoribonuclease Nsp15 interacts with retinoblastoma tumor suppressor protein. Journal of virology, 86(8):4294–304, April 2012. ISSN 1098–5514. doi: 10.1128/JVI.07012-11 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Surjit Milan, Liu Boping, Chow Vincent T K, and Lal Sunil K. The nucleocapsid protein of severe acute respiratory syndrome-coronavirus inhibits the activity of cyclin-cyclin-dependent kinase complex and blocks S phase progression in mammalian cells. The Journal of biological chemistry, 281(16):10669–81, April 2006. ISSN 0021–9258. doi: 10.1074/jbc.M509233200 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Le Sage Valerie, Cinti Alessandro, Amorim Raquel, and Mouland Andrew J.. Adapting the Stress Response: Viral Subversion of the mTOR Signaling Pathway. Viruses, 8(6), June 2016. ISSN 1999–4915. doi: 10.3390/v8060152 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Thaa Bastian, Biasiotto Roberta, Eng Kai, Neuvonen Maarit, Benjamin Götte Lara Rheinemann, Mutso Margit, Utt Age, Varghese Finny, Balistreri Giuseppe, Merits Andres, Ahola Tero, and McInerney Gerald M. Differential Phosphatidylinositol-3-Kinase-Akt-mTOR Activation by Semliki Forest and Chikungunya Viruses Is Dependent on nsP3 and Connected to Replication Complex Internalization. Journal of virology, 89(22):11420–37, November 2015. ISSN 1098–5514. doi: 10.1128/JVI.01579-15 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Peng Lu, Liang Dongyu, Tong Wenyan, Li Jianhua, and Yuan Zhenghong. Hepatitis C virus NS5A activates the mammalian target of rapamycin (mTOR) pathway, contributing to cell survival by disrupting the interaction between FK506-binding protein 38 (FKBP38) and mTOR. The Journal of biological chemistry, 285(27):20870–81, July 2010. ISSN 1083–351X. doi: 10.1074/jbc.M110.112045 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Shives Katherine D, Beatman Erica L, Chamanian Mastooreh, O’Brien Caitlin, Hobson-Peters Jody, and Beckham J David. West nile virus-induced activation of mammalian target of rapamycin complex 1 supports viral growth and viral protein expression. Journal of virology, 88(16):9458–71, August 2014. ISSN 1098–5514. doi: 10.1128/JVI.01323-14 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Lee Chyan-Jang, Liao Ching-Len, and Lin Yi-Ling. Flavivirus activates phosphatidylinositol 3-kinase signaling to block caspase-dependent apoptotic cell death at the early stage of virus infection. Journal of virology, 79(13):8388–99, July 2005. ISSN 0022–538X. doi: 10.1128/JVI.79.13.8388-8399.2005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Shin Yeun-Kyung, Liu Qiang, Tikoo Suresh K, Babiuk Lorne A, and Zhou Yan. Effect of the phosphatidylinositol 3-kinase/Akt pathway on influenza A virus propagation. The Journal of general virology, 88(Pt 3):942–50, March 2007. ISSN 0022–1317. doi: 10.1099/vir.0.82483-0 [DOI] [PubMed] [Google Scholar]
  • 65.de Souza A P Duarte, de Freitas D Nascimento, Fernandes K E Antuntes, da Cunha M D’Avila, Fernandes J L Antunes, Gassen R Benetti, Fazolo T, Pinto L A, Scotta M, Mattiello R, Pitrez P M, Bonorino C, and Stein R T. Respiratory syncytial virus induces phosphorylation of mTOR at ser2448 in CD8 T cells from nasal washes of infected infants. Clinical and experimental immunology, 183(2):248–57, February 2016. ISSN 1365–2249. doi: 10.1111/cei.12720 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Fung To Sing and Liu Ding Xiang. Human Coronavirus: Host-Pathogen Interaction. Annual review of microbiology, 73(1):529–557, 2019. ISSN 1545–3251. doi: 10.1146/annurev-micro-020518-115759 [DOI] [PubMed] [Google Scholar]
  • 67.Schräder Tobias, Dudek Sabine E., Schreiber André, Ehrhardt Christina, Planz Oliver, and Ludwig Stephan. The clinically approved MEK inhibitor Trametinib efficiently blocks influenza A virus propagation and cytokine expression. Antiviral research, 157(July):80–92, 2018. ISSN 1872–9096. doi: 10.1016/j.antiviral.2018.07.006 [DOI] [PubMed] [Google Scholar]
  • 68.Tan Long, Sato Noriaki, Shiraki Atsuko, Yanagita Motoko, Yoshida Yoshihiro, Takemura Yoshinori, and Shiraki Kimiyasu. Everolimus delayed and suppressed cytomegalovirus DNA synthesis, spread of the infection, and alleviated cytomegalovirus infection. Antiviral research, 162(September 2018):30–38, 2019. ISSN 1872–9096. doi: 10.1016/j.antiviral.2018.12.004 [DOI] [PubMed] [Google Scholar]
  • 69.Malvezzi Paolo, Jouve Thomas, and Rostaing Lionel. Use of Everolimus-based Immuno-suppression to Decrease Cytomegalovirus Infection After Kidney Transplant. Experimental and clinical transplantation : official journal of the Middle East Society for Organ Transplantation, 14(4):361–6, August 2016. ISSN 2146–8427. doi: 10.6002/ect.2015.0292 [DOI] [PubMed] [Google Scholar]
  • 70.Bermejo Mercedes, López-Huertas María Rosa, García-Pérez Javier, Climent Núria, Descours Benjamin, Ambrosioni Juan, Mateos Elena, Rodríguez-Mora Sara, Rus-Bercial Lucía, Benkirane Monsef, Miró José M, Plana Montserrat, Alcamí José, and Coiras Mayte. Dasatinib inhibits HIV-1 replication through the interference of SAMHD1 phosphorylation in CD4+ T cells. Biochemical pharmacology, 106:30–45, April 2016. ISSN 1873–2968. doi: 10.1016/j.bcp.2016.02.002 [DOI] [PubMed] [Google Scholar]
  • 71.Pu Szu-Yuan, Xiao Fei, Schor Stanford, Bekerman Elena, Zanini Fabio, Barouch-Bentov Rina, Nagamine Claude M, and Einav Shirit. Feasibility and biological rationale of repurposing sunitinib and erlotinib for dengue treatment. Antiviral research, 155:67–75, 2018. ISSN 1872–9096. doi: 10.1016/j.antiviral.2018.05.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Neveu Gregory, Ziv-Av Amotz, Barouch-Bentov Rina, Berkerman Elena, Mulholland Jon, and Einav Shirit. AP-2-associated protein kinase 1 and cyclin G-associated kinase regulate hepatitis C virus entry and are potential drug targets. Journal of virology, 89(8):4387–404, April 2015. ISSN 1098–5514. doi: 10.1128/JVI.02705-14 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Bekerman Elena, Neveu Gregory, Shulla Ana, Brannan Jennifer, Pu Szu-Yuan, Wang Stanley, Xiao Fei, Barouch-Bentov Rina, Bakken Russell R., Mateo Roberto, Govero Jennifer, Nagamine Claude M., Diamond Michael S., De Jonghe Steven, Herdewijn Piet, Dye John M., Randall Glenn, and Einav Shirit. Anticancer kinase inhibitors impair intracellular viral trafficking and exert broad-spectrum antiviral effects. The Journal of clinical investigation, 127(4):1338–1352, April 2017. ISSN 1558–8238. doi: 10.1172/JCI89857 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Xiang Yang-Fei, Qian Chui-Wen, Xing Guo-Wen, Hao Jing, Xia Min, and Wang Yi-Fei. Antiherpes simplex virus efficacies of 2-aminobenzamide derivatives as novel HSP90 inhibitors. Bioorganic & medicinal chemistry letters, 22(14):4703–6, July 2012. ISSN 1464–3405. doi: 10.1016/j.bmcl.2012.05.079 [DOI] [PubMed] [Google Scholar]
  • 75.Rathore Abhay P S, Haystead Timothy, Das Pratyush K, Merits Andres, Ng Mah-Lee, and Vasudevan Subhash G. Chikungunya virus nsP3 & nsP4 interacts with HSP-90 to promote virus replication: HSP-90 inhibitors reduce CHIKV infection and inflammation in vivo. Antivi- ral research, 103:7–16, March 2014. ISSN 1872–9096. doi: 10.1016/j.antiviral.2013.12.010 [DOI] [PubMed] [Google Scholar]
  • 76.Newman Joseph, Asfor Amin S, Berryman Stephen, Jackson Terry, Curry Stephen, and Tuthill Tobias J. The Cellular Chaperone Heat Shock Protein 90 Is Required for Foot-and- Mouth Disease Virus Capsid Precursor Processing and Assembly of Capsid Pentamers. Journal of virology, 92(5), 2018. ISSN 1098–5514. doi: 10.1128/JVI.01415-17 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Cui Rui, Wang Yizhuo, Wang Liu, Li Guiming, Lan Ke, Altmeyer Ralf, and Zou Gang. Cyclopiazonic acid, an inhibitor of calcium-dependent ATPases with antiviral activity against human respiratory syncytial virus. Antiviral research, 132:38–45, 2016. ISSN 1872–9096. doi: 10.1016/j.antiviral.2016.05.010 [DOI] [PubMed] [Google Scholar]
  • 78.Schögler Aline, Caliaro Oliver, Brügger Melanie, Esteves Blandina I Oliveira, Nita Izabela, Gazdhar Amiq, Geiser Thomas, and Alves Marco P. Modulation of the unfolded protein response pathway as an antiviral approach in airway epithelial cells. Antiviral research, 162:44–50, 2019. ISSN 1872–9096. doi: 10.1016/j.antiviral.2018.12.007 [DOI] [PubMed] [Google Scholar]
  • 79.Win Nan Nwe, Kanda Tatsuo, Nakamura Masato, Nakamoto Shingo, Okamoto Hiroaki, Yokosuka Osamu, and Shirasawa Hiroshi. Free fatty acids or high-concentration glucose enhances hepatitis A virus replication in association with a reduction in glucose-regulated protein 78 expression. Biochemical and biophysical research communications, 483(1):694–699, 2017. ISSN 1090–2104. doi: 10.1016/j.bbrc.2016.12.080 [DOI] [PubMed] [Google Scholar]
  • 80.Karyopharm Therapeutics Inc. Karyopharm Announces Dosing of First Patient in Randomized Study Evaluating Low Dose Selinexor in Patients with Severe COVID-19, 2020.
  • 81.Dobin Alexander, Davis Carrie A., Schlesinger Felix, Drenkow Jorg, Zaleski Chris, Jha Sonali, Batut Philippe, Chaisson Mark, and Gingeras Thomas R.. STAR: ultrafast universal RNA-seq aligner. Bioinformatics (Oxford, England), 29(1):15–21, January 2013. ISSN 1367–4811. doi: 10.1093/bioinformatics/bts635 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Love Michael I, Huber Wolfgang, and Anders Simon. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome biology, 15(12):550, 2014. ISSN 1474–760X. doi: 10.1186/s13059-014-0550-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Smyth G K. Linear models and empirical Bayes methods for assessing differential expression in microarray experiments. Statistical Applications in Genetics and Molecular Biology, 3(1):Art3, 2004. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

1

Articles from bioRxiv are provided here courtesy of Cold Spring Harbor Laboratory Preprints

RESOURCES