With the rapid global spread of SARS-CoV-2, we have become acutely aware of the inadequacies of our ability to respond to viral epidemics. Although disrupting the viral life cycle is critical for limiting viral spread and disease, it has proven challenging to develop targeted and selective therapeutics. Synthetic lethality offers a promising but largely unexploited strategy against infectious viral disease; as viruses infect cells, they abnormally alter the cell state, unwittingly exposing new vulnerabilities in the infected cell. Therefore, we propose that effective therapies can be developed to selectively target the virally reconfigured host cell networks that accompany altered cellular states to cripple the host cell that has been converted into a virus factory, thus disrupting the viral life cycle.

Introduction

Infectious viruses continue to threaten our way of life. For example, every year, seasonal influenza causes 3,000,000 to 5,000,000 cases of severe disease and 290,000 to 650,000 deaths globally (World Health Organization, 2020). Dengue, an enveloped, positive-sense RNA flavivirus, is endemic in more than 100 countries and infects more than 390,000,000 people per year (Bhatt et al., 2013; Messina et al., 2019). In the past five years alone, the RNA viruses Ebola and Zika have caused several widespread epidemics. And at the time of writing, we are in the midst of a devastating global pandemic caused by the novel severe acute respiratory syndrome coronavirus (SARS-CoV)-2. The changing world mediated by the interconnected nature of our societies, coupled with environmental changes and human encroachment into natural ecosystems, only serve to magnify the threat.

Most epidemics over the past decade have been caused by RNA viruses, which include the coronaviruses (e.g., SARS-CoV-1, Middle East Respiratory Syndrome [MERS], and SARS-CoV-2), filoviruses (e.g., Ebola), flaviviruses (e.g., dengue, Zika, yellow fever, and West Nile), orthomyxoviruses (e.g., influenza), and paramyxoviruses (e.g., measles and mumps; Table 1; Heaton, 2019). The remarkable evolvability of RNA viruses contributes to their success as infectious agents and their frequent ability to cross species barriers (Osterhaus, 2001; Carrasco-Hernandez et al., 2017). RNA viruses are prone to recombination and, in some cases such as influenza, reassortment of gene segments, whereby cells infected with two different viral strains can generate a hybrid strain with novel features (Van Poelvoorde et al., 2020). The generation of hybrid strains, coupled with rapid mutational adaptation to new hosts, enables cross-species transmission of lethal variants into naive populations. SARS-CoV-1, MERS, and perhaps SARS-CoV-2 transitioned from bats, which are reservoirs for thousands of coronavirus strains, to humans through an intermediary host, recombining with an endogenous strain of coronavirus of the intermediary host organism (Graham et al., 2013; Menachery et al., 2017; Cui et al., 2019; Boni et al., 2020; Lam et al., 2020; Liu et al., 2020b; Tse et al., 2020; Ye et al., 2020). Influenza is particularly adept at generating new strains and zoonotic transfer by reassortment of its segmented genome, as demonstrated by the flu pandemics of 1918–1919, 1957–1958, and 2009–2011. The avian and swine variants of hemagglutinin and neuraminidase in particular are associated with severe disease (Table 1; Castro et al., 2020; Van Poelvoorde et al., 2020). Most RNA viruses also lack a proofreading replicative polymerase, leading to increased mutation rates that can drive diversity and the emergence of new serotypes (Bell et al., 2019; Durham et al., 2019) and drug resistance. Interestingly, coronaviruses have an alternative error-correction mechanism encoded in nonstructural protein 14, which may help explain the relatively low mutation rate of SARS-CoV-2 (Denison et al., 2011; Ferron et al., 2018; Wang et al., 2020a).

The diversity, adaptability, and evolutionary dynamics of RNA viruses are subject to selective pressures associated with ecosystem changes, stability, infection, transmission, and host susceptibility (Pontremoli et al., 2016) and can present formidable challenges for vaccine development and the utility of broadly effective drugs. Despite decades of effort, there is still no universal vaccine against influenza. The vaccine to seasonal influenza must be updated annually to stay current with the antigenic drift of the virus (Castro et al., 2020). For dengue, achieving vaccine efficacy has been hampered by the presence of four different serotypes. Dengue is notorious for using host immunity to its advantage in a process termed antibody-dependent enhancement (Halstead and O’Rourke, 1977; Peiris and Porterfield, 1979) in which antibodies to one serotype can bind weakly to a different serotype to mediate widespread viral uptake through Fc receptors, raising the risk of severe disease including hemorrhagic fever, shock syndrome, and death (Pierson and Diamond, 2020). This effect can also occur between different flavivirus species, as shown for enhancement of Zika virus infections by prior dengue virus infection in some circumstances (Rodriguez-Barraquer et al., 2019; Whitehead and Pierson, 2019). While concerns about similar antibody-dependent enhancement effects have been raised for SARS-CoV-1 (Jaume et al., 2011; Wang et al., 2016) and SARS-CoV-2 (Eroshenko et al., 2020), perhaps more serious are concerns of the variable magnitude, durability, and protective capacity of the immune response. For seasonal coronaviruses, and possibly SARS-CoV-2, the nature of immunity can be short-lived and insufficient to prevent new infection (Alshukairi et al., 2016; Hamre and Beem, 1972; Isaacs et al., 1983; Long et al., 2020; Mo et al., 2006). Collectively, these uncertainties underscore the need for new approaches to rapidly discover and deploy new antiviral small molecule therapeutics.

Virus-directed therapeutics

Antiviral drugs complement vaccines, and have the potential advantage of rapid deployment during an outbreak. Most current antivirals directly inhibit virus-encoded enzymes that are essential for infection and/or replication. The largest class of antiviral drugs is nucleoside analogues and nonnucleoside inhibitors that target viral replicative polymerases, whether RNA-dependent RNA polymerases or reverse transcriptases (De Clercq, 2009; Hoofnagle, 2012). Remdesivir is a nucleoside triphosphate analogue that has broad-spectrum activities against RNA viruses such as Ebola, MERS, and SARS-CoV-1 in cell culture and animal models. It has recently been shown to shorten the recovery time of hospitalized COVID-19 patients (Beigel et al., 2020; Ferner and Aronson, 2020; Li and De Clercq, 2020; Scavone et al., 2020). Protease inhibitors form a second major category and prevent the processing of viral polyproteins by virus-encoded proteases (Hoofnagle, 2012; Agbowuro et al., 2018). Although there are no clinically approved inhibitors of coronavirus-encoded proteases, pre-clinical inhibitors active against the main protease of SARS-CoV-1 nonstructural protein 5 (nsp5, also called Mpro or 3CLpro) have been reported (Yang et al., 2005). Other viral enzymatic functions that have been successfully targeted include integrases, ion channels, and cap-dependent endonuclease activities (De Clercq, 2009; Hayden et al., 2018; Hoofnagle, 2012; Ison et al., 2020). Non-enzymatic functions can also be interdicted, for example by binding viral entry factors or disrupting RNA-based activities that can affect viral RNA replication and virion assembly (Liu et al., 2015).

Drug discovery in the wake of SARS-CoV-2 includes screening small molecule libraries (Riva et al., 2020; Zhou et al., 2020), machine learning–aided computational drug design approaches (Wang, 2020), and/or concerted medicinal chemistry efforts against various virus-encoded targets (Ghosh et al., 2020). Such targets include the nsp5 protease (Dai et al., 2020; Freitas et al., 2020; Jin et al., 2020a,b; Ma et al., 2020; Rut et al., 2020; Ton et al., 2020; Zhang et al., 2020), RNA-dependent RNA polymerase (Hillen et al., 2020; Wang et al., 2020b; Yin et al., 2020), exo- and endoribonucleases (Kim et al., 2020), and RNA cap nucleotide methyltransferases (Viswanathan et al., 2020). The interaction between spike and angiotensin converting enzyme 2 (ACE2) is a top priority nonenzymatic target (Lan et al., 2020; Monteil et al., 2020; Walls et al., 2020; Yan et al., 2020). Candidate molecules for therapeutic and/or prophylactic use include decoy fragments of ACE2 (Monteil et al., 2020), convalescent sera (Casadevall and Pirofski, 2020; Casadevall et al., 2020), monoclonal antibodies cloned from COVID-19 patients (Liu et al., 2020a; Pinto et al., 2020; Robbiani et al., 2020; Rogers et al., 2020; Seydoux et al., 2020) and nanobodies, single-domain antibody fragments derived from the variable heavy domain region of an IgG subclass of camelid antibodies (Fridy et al., 2014; Huo et al., 2020; Wrapp et al., 2020; Wu et al., 2020). Discovery of inhibitors of SARS-CoV-2–encoded targets is enabled by a wealth of x-ray crystallography and cryo-EM structural data that has become available since January 2020 with a protein structure determined forover half of the SARS-CoV-2 proteins or one of their constituent domains (Ghosh et al., 2020; Zhou et al., 2020).

Most antiviral drugs are developed in a virus-specific fashion and tend to be only marginally effective as monotherapies, in part because it is difficult to fully inhibit viral replication in patients due to dose-limiting toxicity (Jefferson et al., 2014) and in part because of the rapid emergence of resistance (Van Poelvoorde et al., 2020). An effective strategy to overcome these hurdles is to combine multiple drugs against different viral targets, with highly active antiretroviral combination therapies against human immunodeficiency virus (HIV) and hepatitis C virus being successful examples (Hofmann et al., 2009). Unfortunately, the antiviral drug combinations can only be explored after long-term development against multiple targets encoded by a particular pathogen. The difficulties inherent to antiviral drug development are underscored by a stark statistic: while there are currently 214 RNA viruses known to infect humans, U.S. Food and Drug Administration–approved antiviral therapeutics exist for only eight of these pathogens (Chaudhuri et al., 2018; Heaton, 2019; Woolhouse and Brierley, 2018).

Identifying host cell dependencies

As obligate pathogens, viruses require the host machinery for replication and propagation. Viruses exploit various host cell functions, including natural host receptors, endocytic machinery, organellar compartments, primary metabolism, RNA and protein synthesis, protein homeostasis, membrane biogenesis, and the endomembrane secretory system, among other functions. Viruses may also interdict innate antiviral responses, block host cell proliferation, and suppress cell death. Given the reliance on a myriad of host processes, host-based targets dramatically increase the potential search space for antiviral compounds.

Host genes in virally infected cells on which the virus is dependent are termed host-dependency factors and can be revealed as host–virus genetic interactions. Systematic loss-of-function genetic screens identify two main classes of genes that affect viral replication. The first class contains host genes that a virus needs to initiate and complete its life cycle, termed proviral genes because loss of function renders the host cell resistant to infection. The second class encompasses genes that have antiviral activity because loss of function sensitizes cells to the effects of infection. Antiviral gene function may be direct, such as for innate immunity genes, or may indirectly buffer and suppress the adverse effects of infection, for instance ER chaperones that help the host cell cope with the massive secretory flux imposed during virion biogenesis (Table 2). Loss of direct-acting antiviral genes increases viral replication and enhances infectivity, whereas loss of indirect-acting genes increases susceptibility to host cell death and may limit infectivity. These indirect antiviral functions manifest as synthetic lethality, as will be discussed.

Early RNAi screens hinted at the promise of genetic approaches to understand host dependencies (Krishnan et al., 2008) but were plagued by high false-positive and false-negative rates (Chung et al., 2014; Hart et al., 2014; Mohr et al., 2010). With the advent of genome-wide CRISPR knockout screens using complex pooled gRNA libraries that efficiently target every human gene, system-level interrogation of virus–host cell dependencies has the potential to define broad and virus-specific host genetic dependencies (Hart et al., 2015; Wang et al., 2015).

To date, most genome-wide CRISPR screens have been performed in a positive selection format that allows recovery only of gene knockouts that prevent the cytopathic effect and cell death caused by viral infection (Table 2). Phenotypic screens for noncytolytic viruses that employ sorting strategies to enrich for factors that enhance or inhibit persistence have also been performed (Puschnik et al., 2017). While screens revealed some common host pathways that mediate infection, many differences between viruses are evident. In addition, CRISPR screens with the same virus in different cell lines reveal cell type–specific dependencies (Table 2; Li et al., 2019; Savidis et al., 2016). Initial results from a genome-wide CRISPR screen for pro- and antiviral host genes for SARS-CoV-2 identified components of the TGF-β signaling pathway, the switch/sucrose nonfermenting chromatin remodeling complex, histone demethylases, ACE2, and the cathepsin L protease as proviral (Table 2; Wei et al., 2020). Antiviral host genes include components of the histone H3.3 chaperone complex, nucleosome remodeling factor complex, transcription factor IIH complex, and small nuclear ribonucleoprotein chaperone complex (Table 2; Wei et al., 2020). Further systematic identification of host–coronavirus genetic interactions in relevant human cell types will almost certainly yield a plethora of new candidate antiviral targets for SARS-CoV-2.

Host-directed antivirals

Several host-based antiviral therapeutics have been developed that modulate the host immune response (Kaufmann et al., 2018), and in doing so aim to dampen virus-induced pathology and buy the immune system time to mount a strong antiviral response. For example, imiquimod activates the toll-like receptor TLR7 during human papilloma virus infection to activate the host innate immune response through production of the cytokines IFN-α, interleukins 1 and 6, and TNF-α (Bilu and Sauder, 2003; Kaufmann et al., 2018). In another example, the retinoic acid derivative acitretin stimulates the cytosolic pattern recognition receptor, RIG-I, in cells with reactivated latent HIV, thereby inducing an IFN response, triggering apoptosis and depletion of the viral reservoir (Kaufmann et al., 2018; Li et al., 2016). Dexamethasone, a glucocorticoid receptor agonist with anti-inflammatory and immunosuppressant effects, reduced mortality for COVID-19 patients receiving invasive mechanical ventilation or oxygen support (Horby et al., 2020). Potential host-directed, immune-modulating antivirals that may be repurposed against SARS-CoV-2 also include acitretin, the C-C chemokine receptor type 1 antagonist MLN-3897, and apilimod (Gordon et al., 2020a; Riva et al., 2020). However, nonspecific immune modulators can lead to unintended side effects. A key to solving this problem may lie in targeting attributes of the viral-infected cell that are not shared with the uninfected cell and exploiting these features for therapeutic intervention.

Virus-induced vulnerability

As viruses infect cells, they execute control programs and hijack the host machinery to serve the viruses’ goal of producing thousands of virions. They do so by using a relatively small number of precise control elements to subvert host cell functions (Ravindran et al., 2019). With a compact but highly adaptable toolset at their disposal, viruses continuously evolve to redirect the function of hundreds of host molecules to optimize viral fitness, thus rewiring the host cell. These radical changes create a novel genetic architecture that is specific to the virus-infected state (Gulbahce et al., 2012).

Rewiring cellular functions by virus–host protein–protein interactions involves co-opting so-called “driver” or “control” nodes that have greater influence on the host network function (Liu et al., 2011; Ravindran et al., 2019). These interactions can inhibit existing functions, redirect host proteins to other locations, or generate new targets and functionalities, which may lead to other downstream network effects. Because viruses depend on these host functions usurped by protein–protein interactions, targeting the interfaces between virus and host proteins directly has been proposed as a mechanism to inhibit the viral life cycle (Basler et al., 2019; Carpp et al., 2014; Gordon et al., 2020a; Heaton, 2019; Luo et al., 2016). However, we propose that the susceptible state induced by these interactions may be best targeted by chemically exploiting the genetic concept of synthetic lethality.

Synthetic lethality

Genetic interactions, typically observed by the synthetic combination of loss of function alleles in two genes, expose the functional organization of a cell and are broadly classified as either positive or negative (Costanzo et al., 2019). Negative genetic interactions reveals functional dependencies within cells and result when the combination of loss-of-function alleles in the same cell leads to a greater than multiplicative phenotype, the extreme form of which is synthetic lethality (Hartwell et al., 1997; Costanzo et al., 2019). Synthetic suppression, a type of positive genetic interaction, occurs when the combination results in a less severe phenotype than either of the single mutant alleles. Cell viability or growth is commonly used as a readout for genetic interactions, but any quantitative phenotype can be used to detect genetic interactions. The nonlinear interaction of two mutant alleles that results in synthetic lethality often reflects the genetic buffering that is inherent to biological systems: loss or reduction in functionality of either gene is not strongly deleterious due to genetic redundancy, but loss of both functions causes inviability (Fig. 1). Systematic genetic screens in the budding yeast Saccharomyces cerevisiae have revealed nearly 1,000,000 binary synthetic lethal and synthetic suppressive relationships, which far exceeds the approximately 1,000 single essential genes in this organism (Costanzo et al., 2019). At the molecular level, synthetic lethality can reflect the operation of two parallel pathways that perform the same function, the redundant contribution of nonessential subunits to essential protein complexes, or damage prevention–response relationships (Bader et al., 2003). For example, in yeast, the essential nuclear pore complex can often survive the removal of one but usually not two nonessential subunits (Fabre and Hurt, 1997; Kim et al., 2018).

The phenomenon of synthetic lethality can be exploited to target specific disease states. For example, synthetic lethal drug–gene mutation interactions have been identified that exploit the genetic vulnerabilities of cancer cells (Fig. 1; Ashworth et al., 2011; Hartwell et al., 1997; Kaelin, 2005; Mendes-Pereira et al., 2009; Turner et al., 2008; Wiltshire et al., 2010). In a paradigm-shifting example, cells bearing mutations in the breast cancer type 1 susceptibility gene BRCA1 were found to rely on an alternative poly-ADP ribose polymerase pathway for homologous recombination, and hence to be exquisitely sensitive to poly-ADP ribose polymerase inhibitors (Lord and Ashworth, 2017). Numerous other examples of genetic vulnerabilities have been identified in cancer and other disease states (Huang et al., 2020; Mair et al., 2019), and this concept is readily extended to infectious disease (Mast et al., 2014; Tyers and Wright, 2019).

Viruses are dependent on the redundancies present within the host cell that allow it to survive for enough time to produce new virions despite infection. These redundancies may be targeted using the principle of synthetic lethality (Fig. 1). Applying the principle of synthetic lethality to viral infection requires identifying the viral-induced vulnerabilities and then targeting the redundancy and buffering capacity of cells in the infected state, thus killing or crippling both the host cell and the virus. Systems biology approaches, such as comprehensive identification and characterization of host–pathogen interactions, offer an unprecedented opportunity to exploit host vulnerabilities after sensitization by the virus (Eckhardt et al., 2020). With comprehensive functional system maps (Krogan et al., 2015; Mast et al., 2014), it may be possible to identify drug targets whose modulation kills or otherwise disrupts infected cells selectively during viral infection. The existing protein–protein interaction network (PPI) and drug repurposing data already provide ample resources for screening for potential synthetic lethal drug and gene candidates (Gordon et al., 2020a; Zhou et al., 2020). Below we discuss strategies to uncover viral-induced vulnerabilities and how to exploit these vulnerabilities for antiviral therapeutics.

Exploiting known virus biology

The accumulated wealth of information on functional interaction of viruses with host cells provides a rich source of candidate host processes that might be targeted in a synthetic lethal strategy. Many of the genes required for these processes are not strictly essential in the host cell and thus present viable therapeutic windows, particularly for chemical inhibitors that have passed safety criteria in human clinical trials for other indications. A nonexhaustive list of examples follows.

Virus-induced proteolysis

Many viruses target host proteins for degradation either directly or indirectly by usurping the host’s degradation machinery. Enteroviruses are known to target host cell proteins with viral proteases (Laitinen et al., 2016; Lamphear et al., 1993; Lloyd, 2016). As an example, coxsackievirus 2A, a viral-encoded protease, cleaves translation initiation factor 4G and the poly-A binding protein (Lamphear et al., 1993). The depletion of these two proteins in virus-infected cells results in shutting down cap-dependent translation by preventing engagement of the messenger ribonucleoprotein to the small ribosomal subunit (Laitinen et al., 2016; Lloyd, 2016). Because enteroviruses use cap-independent translation, the translation machinery shifts to viral transcripts. In another example, coxsackievirus B3, viral-encoded protease, cleaves dystrophin, a protein whose disruption leads to cardiomyopathies, demonstrating the resulting phenotypic consequences of viral proteolysis (Badorff et al., 2000). SARS-CoV-2 nsp5 interacts with histone deacetylase 2 and tRNA methyltransferase 1, which have putative nsp5-specific cleavage sites (Gordon et al., 2020a). Degradation of histone deacetylase 2 might suppress host inflammatory and immune signaling responses (Comalada et al., 2010; Guise et al., 2013; Roger et al., 2011), while loss of tRNA methyltransferase 1 may impair host–protein translation and redox homeostasis (Dewe et al., 2017). Loss of either protein would sensitize SARS-CoV-2–infected cells to drugs targeting their synthetic lethal partners.

In addition, the ubiquitin proteasome system is exploited by some viruses to deplete host proteins by proteolysis in a more indirect fashion (Isaacson and Ploegh, 2009), as exemplified by degradation of p53 upon human papilloma virus infection (Laitinen et al., 2016). Numerous other host cell proteins are targeted for proteolysis upon viral infection (Laitinen et al., 2016), effectively rewiring the host cell network as functional proteins are depleted or attenuated. For example, the SARS-CoV-2 papain-like PLpro cysteine protease nonstructural protein (nsp)3 has deubiquitinase and de-(IFN-stimulated gene 15)-ylase activity in vitro, suggesting potential activity in further altering cell proteostasis (Freitas et al., 2020). Experiments in yeast and other model systems demonstrate that reduced gene expression (and consequent reduced functional protein levels) renders the cell more vulnerable to targeted therapeutic treatment (Andrusiak et al., 2012; Cokol et al., 2011). In the case of viral-induced proteolysis that reduces host protein function below a critical level, synthetic lethal dependencies may be targeted to cripple the host cell and disrupt the viral life cycle.

Virus-induced adaptive network states

Studies in bacteria treated with antimicrobials and in cancer cells treated with chemotherapeutics have demonstrated that all cells elicit protective transcriptional responses to better tolerate cytotoxic drug effects (Huang et al., 2020; Niepel et al., 2017). Secondary drugs targeting new vulnerabilities within active regulatory and metabolic networks of the tolerant state can potentiate the cytotoxic effect of the primary drug (Cokol et al., 2018; Ma et al., 2019; Peterson et al., 2016; Plaisier et al., 2016). Similarly, we posit that new vulnerabilities in the regulatory and metabolic networks of a virus-infected cell can be targeted with drugs that will have no cytotoxicity to uninfected cells. Common transcriptional responses of virally infected cells include innate immune responses as well as cellular stress responses such as the unfolded protein response, which may suggest specific targets for host-directed antivirals that can be designed to either selectively disable infected cells or support antiviral activity of the innate cellular, and perhaps the coupled adaptive immune, response to infection (Banerjee et al., 2020; Ferreira et al., 2020; Netea et al., 2020).

Virus-induced protein–protein interactions

Viruses use their limited proteomes to bind to host cell proteins, altering PPIs (Basler et al., 2019; Lum and Cristea, 2016). Each viral protein can interact with numerous host cell proteins. The large repertoire of host-interacting proteins is enhanced by the presence of short linear motifs in numerous viral proteins that mediate interactions (Davey et al., 2011). These interactions can have many effects, from controlling signaling and host responses to viral infection, to reorganizing structures such as the secretory system to enable viral replication. Drugs that target these host proteins of the PPIs have been shown to disrupt the viral life cycle (Carpp et al., 2014), most notably with viral entry inhibitors that prevent interaction between host cell surface receptors and viral coat proteins (Kaufmann et al., 2018). However, these druggable targets are limited in number and often essential in humans (Heaton, 2019).

Exploiting the synthetic lethal concept with PPI networks may be an effective host-based antiviral strategy. As an example, consider the effect of a virus-dependent PPI usurping the normal function of a host protein. Once recruited, this host protein may become less able to perform its normal function, compromising that function. This recruited protein may be considered a viral-induced “hypomorph” due to its reduced, but not abolished, functionality. Consequently, the cell becomes more dependent on proteins with which the viral-induced hypomorph functionally interacts or with proteins that perform overlapping functions. This dependence sensitizes the infected cell to drugs that target these protein partners of the viral-induced hypomorph. Typically, these partner proteins are numerous (Carpp et al., 2014; Luo et al., 2016; Shah et al., 2018). Studies in yeast show that the average gene participates in ∼100 negative and ∼65 positive interactions (Costanzo et al., 2016), each one of which, in the case of viral-induced hypomorphs, potentially presents a new host-based drug target. As with other viral-induced vulnerabilities, these targets are only exposed as a result of a viral infection, and such drugs should therefore have little effect on uninfected cells. It should also be possible to identify or predict “synthetic lethal” partners of the viral-induced hypomorph based on CRISPR screen, PPI, and genetic network and emerging human synthetic lethal data (Benstead-Hume et al., 2019).

Virus-induced targeting of common pathways

Analysis of PPI data from viral infection models suggest that many viruses target common proteins, networks, or processes (de Chassey et al., 2014; Meyniel-Schicklin et al., 2012; Pfefferle et al., 2011). Viral proteins often interact with “hubs” in PPI networks (Heaton, 2019; Ravindran et al., 2019; Shah et al., 2018). There is a correlation between the number of protein interactions a protein has and its synthetic lethal partners (Costanzo et al., 2016), suggesting that common druggable targets should be present in these networks. Different viruses also target the same host pathways via targeting different protein members of the pathway. Borrowing from what has been learned from yeast, genes in the same pathway have correlated genetic interactions, suggesting common targets can be identified. However, direct translation from yeast remains challenging due to the added complexities of multicellularity and genetic diversity in humans.

For example, the host translational machinery is required by viruses to synthesize viral proteins at the expense of host proteins. Many viruses target the signaling proteins that regulate host protein synthesis to ease ribosome demand by the host, in turn allowing enhanced virus protein production. Enveloped viruses must all take advantage of the ER translocon and chaperone machinery, by redirecting host lipid synthesis and glycosylation machineries to become properly enveloped with functional and mature surface proteins. Several members of the flaviviruses target the ER stress response to ensure the production of new virions in specialized subdomains of the ER (Shah et al., 2018; Zhang et al., 2016). They also target autophagy, with both responses contributing to lengthening the lifespan of the infected cell, while minimizing detection by host immune surveillance and clearance and redirecting membrane resources in support of viral production. During replication, viruses retarget host proteins to replication complexes to assist in chaperoned folding, packaging, and assembly of new virions (Coyaud et al., 2018; Zhang et al., 2016). In the case of membrane-bound virions, a reconfiguration of the host secretory pathway occurs to support virion trafficking and budding from the cell. Such large-scale morphological changes may rely on the buffering capacity of cells and reveal specific vulnerabilities that could be targeted by synthetic lethal approaches. Furthermore, redistribution of critical host factors may contribute to a virus-induced state that could be selectively targeted by synthetic lethality, such as the movement of the endosomal sorting complex required for transport (ESCRT)-III to sites of flavivirus replication in the ER (Tabata et al., 2016) that leads to a reduced capacity of ESCRT-III function elsewhere in the cell. Thus, commonly reconfigured networks could be harnessed for developing broad-spectrum therapeutics against many different viruses by targeting conserved synthetic lethal partners of common interactors and processes.

Results from CRISPR screens also support the notion of broad-spectrum viral targets (Li et al., 2020; Savidis et al., 2016; Shah et al., 2018; Zhang et al., 2016). A comparison of the top 100 highest scoring hits in studies performed to date reveals that some genes are important not only for closely related viruses but also for unrelated clades. Among the highest scoring vulnerabilities are multiple subunits of the chaperone ER-membrane protein complex (EMC) that are essential for West Nile, dengue, Zika, rhino, influenza A, and hepatitis B viral infections (Barrows et al., 2019; Ngo et al., 2019). The EMC is a highly conserved transmembrane domain complex and loss of the EMC results in accumulation of misfolded membrane proteins (Jonikas et al., 2009). The EMC physically interacts with components of the ER-associated protein degradation (ERAD) pathway, which is also often essential for virus replication, as exemplified requirements for the ubiquitin conjugating enzymes UBE2J1 and UBE2G2 in Epstein-Barr, dengue, and West Nile virus replication. Other common host factor dependency pathways include components of the oligosaccharyltransferase complex (OST; Harada et al., 2019) and the vacuolar ATPase (Ho et al., 2017; Perreira et al., 2015). The dependence of these different viruses on these common processes suggests the existence of common druggable targets on which different viruses may be directly or indirectly dependent through viral-induced vulnerability.

From host dependency factor genes to antiviral drugs

Pharmacological inhibition of host dependency factors identified in CRISPR screens should phenocopy resistant to infection and/or replication, and in instances where preexisting drugs are available, present immediate opportunities for drug repurposing. Several examples support this strategy. In a screen with Zaire Ebola virus (EBOV), knockout of the N-acetylglucosamine-1-phosphate transferase GNPTAB impaired infection, likely due to loss of cathepsin B activity, a known EBOV entry factor (Flint et al., 2019). GNPTAB activity requires proteolytic processing by the subtilisin kexin isozyme-1/site-1 protease, and correspondingly, inhibition of ubtilisin kexin isozyme-1/site-1 protease with the small molecule PF-429242 also impaired EBOV infection (Flint et al., 2019). Similarly, loss of poly(A) RNA polymerase associated domain containing proteins 5 and 7, two subunits of the Trf4/5–Air1/2–Mtr4 polyadenylation complex, impairs hepatitis A replication and phenocopied the inhibitor RG7834 (Mueller et al., 2018, 2019). The histone demethylase inhibitor JIB-04 was able to phenocopy the loss of myc-induced nuclear antigen 53 in a model of HIV infection and latency, and it exhibited synergy with other HIV latency–reversing agents (Huang et al., 2019). This new therapeutic strategy may help purge HIV-1 viral reservoirs. Small molecule inhibitors of proviral targets identified for SARS-CoV-2 block virus-induced cell death and viral replication in cell culture. These compounds include the switch/sucrose nonfermenting–related, matrix-associated, actin-dependent regulator of chromatin, subfamily A, member 4 (SMARCA4) inhibitor PFI3, and SIS2, an inhibitor of the mothers against the decapentaplegic homolog 4 (SMAD4)/TGF-β pathway (Wei et al., 2020). Finally, even in the absence of known inhibitors, antiviral gene–drug interactions can validate new targets. For example, a host dependency factor identified in an influenza A screen, the mRNA cap methyltransferase 1, exhibits strong and highly specific synergy with the U.S. Food and Drug Administration–approved influenza A endonuclease inhibitor baloxavir (Li et al., 2020). Building on these successes, further CRISPR-based screens against SARS-CoV-2 and many other viruses, combined with synthetic lethal concepts, should enable new antiviral drugs.

The path forward

The development of therapeutics for targeting viral-induced vulnerabilities based on synthetic lethality presents an exciting opportunity to identify therapeutics that are specific for virally infected cells. Unlike viral targets, host targets are not subjected to powerful mutational selection for resistance and may inhibit a spectrum of viral pathogens that rely on common host systems. Importantly, complete ablation of the host target may not be required to achieve a dramatic negative effect on the virus-infected cell. Less potent drugs that are still effective should be a lot easier to develop and may also have fewer side effects in uninfected cells. Furthermore, combining synergistic drugs at what would otherwise be subtherapeutic levels may be an effective antiviral strategy (Tyers and Wright, 2019). Moreover, approved drugs or advanced preclinical candidates discovered through efforts originally aimed at other indications may be rapidly repurposed as antivirals.

CRISPR-based genetic profiles for all major human viral pathogens will help identify viral-induced vulnerabilities and guide the development of inhibitors that are broadly effective against sets of related viruses as front-line interventions. However, as demonstrated in the development of synthetic lethal cancer therapeutics, care with respect to genetic diversity and the differences between cell culture and more physiologically relevant disease environments remains an important consideration (Huang et al., 2020). Interactions dependent on a particular genetic background can become irrelevant in different genetic backgrounds, cell lines, or model systems. Beyond screening, systems-based models that capture the network dynamics of the viral life cycle, infection, and host responses can help to predict and prioritize synthetic lethal interactions among host genes, drug combinations, and their potential synergy or other complex interactions that will be robust to genetic and physiological idiosyncrasies. Exploration of drug combinations based on the concept of genetic interactions is already underway (Eckhardt et al., 2020; Gordon et al., 2020b) and, as demonstrated by synthetic lethal cancer therapeutics (Huang et al., 2020), holds promise as an effective solution. In addition to targeting infected cells, host-based antivirals can target the subsequent immune response that is often associated with pathology. Therapeutics should target the early stages of infection to promote cell death, limit viral release, minimize dysregulation of the host inflammatory response and host tissue damage, and ideally promote immune surveillance and clearance of infected cells. Of course, as with any pharmacological agent, side effects that arise by on-or-off target effects must be balanced with efficacy.

Quantitative multiscale models that address the multiscale nature of infection will guide the development of therapeutics that are effective and minimize unintended negative impacts on the host. Computing such models is difficult, in part because of the limited amount, relevance, and quality of the data. However, integrative modeling methods may help us address some of these challenges (Calhoun et al., 2018; Rout and Sali, 2019). Moreover, emerging data on virus-mediated network remodeling, including host–viral protein interactions, and global phosphorylation dynamics in SARS-CoV-2 infections (Bouhaddou et al., 2020; Gordon et al., 2020a), host responses to infection and viral-induced morphological changes, genetic interactions, and host dependency factors augers well for the rapid development and deployment of novel therapeutic strategies to combat new and old viruses that plague humankind (Benstead-Hume et al., 2019).

Conclusions

By increasing the diversity of targets and explicitly addressing the challenge of drug specificity, synthetic lethal approaches that target viral-induced vulnerabilities of the host cell can help us respond to the pressing challenge of outbreaks caused by viruses. Identifying solutions to infectious disease through the familiar prism of genetic interactions has the potential to illuminate numerous new drug targets, rationally repurpose existing drugs, and define mechanisms of action for drugs discovered through unbiased screening approaches. By working now to identify common host functions used by many viruses and to develop synthetic lethal drug approaches based on these targets, we may be able to more rapidly and comprehensively counter future global outbreaks.

The authors gratefully acknowledge support from the National Institutes of Health: U19 AI100627 (A. Aderem), R01 AI032972 (A. Aderem), U19 AI135976 (A. Aderem, J.D. Aitchison, N.S. Baliga), U19 AI111276 (J.D. Aitchison), P41 GM109824 (J.D. Aitchison, M.P. Rout, A. Sali, B.T. Chait), R01 GM112108 (J.D. Aitchison, M.P. Rout), R01 AI141953 (N.S. Baliga, J.D. Aitchison), R01 AI128215 (N.S. Baliga), GM 109824 (B.T. Chait), R01 CA228351 (M.P. Rout), U19AI135990 (A. Sali), R01GM083960 (A. Sali), P01 AI138398-S1 (C.M. Rice), U19 AI111825 (C.M. Rice), R01 AI091707 (C.M. Rice), R21 AI151344 (A. Kaushansky), and R01 GM101183 (A. Kaushansky); the Canadian Institutes for Health Research: FDN-167277 (M. Tyers); The G. Harold and Leila Y. Mathers Charitable Foundation (J.D. Aitchison, M.P. Rout, B.T. Chait, C.M. Rice); and a George Mason University Fast Grant (C.M. Rice).

The authors declare no competing financial interests.

Author contributions: J.D. Aitchison, F.D. Mast, and A.T. Navare wrote the original draft of the manuscript. F.D. Mast, A.T. Navare, A.M van der Sloot, J. Coulombe-Huntinton, M.P. Rout, N.S. Baliga, A. Kaushansky, B.T. Chait, A. Aderem, C.M. Rice, A. Sali, and J.D. Aitchison contributed ideas and examples to expand and define the scope of applying the synthetic lethal concept to viral infections. All authors commented on and edited the manuscript.

Agbowuro
,
Ayodeji A
,
Wilhelmina M
Huston
,
Allan B
Gamble
, and
Joel D A
Tyndall
.
2018
.
Proteases and protease inhibitors in infectious diseases
.
Med Res Rev
.
38
(
4
):
1295
1331
.
[PubMed]
.
Alshukairi
,
A.N.
,
I.
Khalid
,
W.A.
Ahmed
,
A.M.
Dada
,
D.T.
Bayumi
,
L.S.
Malic
,
S.
Althawadi
,
K.
Ignacio
,
H.S.
Alsalmi
,
H.M.
Al-Abdely
, et al
.
2016
.
Antibody Response and Disease Severity in Healthcare Worker MERS Survivors
.
Emerg. Infect. Dis
.
22
:
1113
1115
.
Andrusiak
,
K.
,
J.S.
Piotrowski
, and
C.
Boone
.
2012
.
Chemical-genomic profiling: systematic analysis of the cellular targets of bioactive molecules
.
Bioorg. Med. Chem
.
20
:
1952
1960
.
Ashworth
,
A.
,
C.J.
Lord
, and
J.S.
Reis-Filho
.
2011
.
Genetic interactions in cancer progression and treatment
.
Cell
.
145
:
30
38
.
Bader
,
G.D.
,
A.
Heilbut
,
B.
Andrews
,
M.
Tyers
,
T.
Hughes
, and
C.
Boone
.
2003
.
Functional genomics and proteomics: charting a multidimensional map of the yeast cell
.
Trends Cell Biol
.
13
:
344
356
.
Badorff
,
C.
,
N.
Berkely
,
S.
Mehrotra
,
J.W.
Talhouk
,
R.E.
Rhoads
, and
K.U.
Knowlton
.
2000
.
Enteroviral protease 2A directly cleaves dystrophin and is inhibited by a dystrophin-based substrate analogue
.
J. Biol. Chem
.
275
:
11191
11197
.
Banerjee
,
A.
,
S.J.
Czinn
,
R.J.
Reiter
, and
T.G.
Blanchard
.
2020
.
Crosstalk between endoplasmic reticulum stress and anti-viral activities: A novel therapeutic target for COVID-19
.
Life Sci
.
255
. 117842.
Barrows
,
N.J.
,
Y.
Anglero-Rodriguez
,
B.
Kim
,
S.F.
Jamison
,
C.
Le Sommer
,
C.E.
McGee
,
J.L.
Pearson
,
G.
Dimopoulos
,
M.
Ascano
,
S.S.
Bradrick
, et al
.
2019
.
Dual roles for the ER membrane protein complex in flavivirus infection: viral entry and protein biogenesis
.
Sci. Rep
.
9
:
9711
.
Basler
,
C.F.
,
N.J.
Krogan
,
D.W.
Leung
, and
G.K.
Amarasinghe
.
2019
.
Virus and host interactions critical for filoviral RNA synthesis as therapeutic targets
.
Antiviral Res
.
162
:
90
100
.
Beigel
,
J.H.
,
K.M.
Tomashek
,
L.E.
Dodd
,
A.K.
Mehta
,
B.S.
Zingman
,
A.C.
Kalil
,
E.
Hohmann
,
H.Y.
Chu
,
A.
Luetkemeyer
,
S.
Kline
, et al;
ACTT-1 Study Group Members
.
2020
.
Remdesivir for the Treatment of Covid-19 - Preliminary Report
.
N. Engl. J. Med
.
Bell
,
S.M.
,
L.
Katzelnick
, and
T.
Bedford
.
2019
.
Dengue genetic divergence generates within-serotype antigenic variation, but serotypes dominate evolutionary dynamics
.
eLife
.
8
. e42496.
Benstead-Hume
,
G.
,
X.
Chen
,
S.R.
Hopkins
,
K.A.
Lane
,
J.A.
Downs
, and
F.M.G.
Pearl
.
2019
.
Predicting synthetic lethal interactions using conserved patterns in protein interaction networks
.
PLOS Comput. Biol
.
15
. e1006888.
Bhatt
,
S.
,
P.W.
Gething
,
O.J.
Brady
,
J.P.
Messina
,
A.W.
Farlow
,
C.L.
Moyes
,
J.M.
Drake
,
J.S.
Brownstein
,
A.G.
Hoen
,
O.
Sankoh
, et al
.
2013
.
The global distribution and burden of dengue
.
Nature
.
496
:
504
507
.
Bilu
,
D.
, and
D.N.
Sauder
.
2003
.
Imiquimod: modes of action
.
Br. J. Dermatol
.
149
(
Suppl 66
):
5
8
.
Boni
,
Maciej F
,
Philippe
Lemey
,
Xiaowei
Jiang
,
Tommy Tsan-Yuk
Lam
,
Blair W
Perry
,
Todd A
Castoe
,
Andrew
Rambaut
, and
David L
Robertson
.
2020
.
Evolutionary origins of the SARS-CoV-2 sarbecovirus lineage responsible for the COVID-19 pandemic
.
Nat Microbiol
.
[PubMed]
.
Bouhaddou
,
M.
,
D.
Memon
,
B.
Meyer
,
K.M.
White
,
V.V.
Rezelj
,
M.
Correa Marrero
,
B.J.
Polacco
,
J.E.
Melnyk
,
S.
Ulferts
,
R.M.
Kaake
, et al
.
2020
.
The Global Phosphorylation Landscape of SARS-CoV-2 Infection
.
Cell
.
In press
. S0092-8674(20)30811-4.
Calhoun
,
S.
,
M.
Korczynska
,
D.J.
Wichelecki
,
B.
San Francisco
,
S.
Zhao
,
D.A.
Rodionov
,
M.W.
Vetting
,
N.F.
Al-Obaidi
,
H.
Lin
,
M.J.
O’Meara
, et al
.
2018
.
Prediction of enzymatic pathways by integrative pathway mapping
.
eLife
.
7
. e31097.
Carpp
,
L.N.
,
R.S.
Rogers
,
R.L.
Moritz
, and
J.D.
Aitchison
.
2014
.
Quantitative proteomic analysis of host-virus interactions reveals a role for Golgi brefeldin A resistance factor 1 (GBF1) in dengue infection
.
Mol. Cell. Proteomics
.
13
:
2836
2854
.
Carrasco-Hernandez
,
R.
,
R.
Jácome
,
Y.
López Vidal
, and
S.
Ponce de León
.
2017
.
Are RNA Viruses Candidate Agents for the Next Global Pandemic? A Review
.
ILAR J
.
58
:
343
358
.
Casadevall
,
Arturo
, and
Liise-Anne
Pirofski
.
2020
.
The convalescent sera option for containing COVID-19
.
J. Clin. Invest
.
130
(
4
):
1545
1548
.
[PubMed]
.
Casadevall
,
Arturo
,
Michael J
Joyner
, and
Liise-Anne
Pirofski
.
2020
.
A Randomized Trial of Convalescent Plasma for COVID-19-Potentially Hopeful Signals
.
JAMA
.
[PubMed]
.
Castro
,
L.A.
,
T.
Bedford
, and
L.
Ancel Meyers
.
2020
.
Early prediction of antigenic transitions for influenza A/H3N2
.
PLOS Comput. Biol
.
16
. e1007683.
Chaudhuri
,
S.
,
J.A.
Symons
, and
J.
Deval
.
2018
.
Innovation and trends in the development and approval of antiviral medicines: 1987-2017 and beyond
.
Antiviral Res
.
155
:
76
88
.
Chung
,
H.Y.
,
M.
Gu
,
E.
Buehler
,
M.R.
MacDonald
, and
C.M.
Rice
.
2014
.
Seed sequence-matched controls reveal limitations of small interfering RNA knockdown in functional and structural studies of hepatitis C virus NS5A-MOBKL1B interaction
.
J. Virol
.
88
:
11022
11033
.
Cokol
,
M.
,
H.N.
Chua
,
M.
Tasan
,
B.
Mutlu
,
Z.B.
Weinstein
,
Y.
Suzuki
,
M.E.
Nergiz
,
M.
Costanzo
,
A.
Baryshnikova
,
G.
Giaever
, et al
.
2011
.
Systematic exploration of synergistic drug pairs
.
Mol. Syst. Biol
.
7
:
544
.
Cokol
,
M.
,
C.
Li
, and
S.
Chandrasekaran
.
2018
.
Chemogenomic model identifies synergistic drug combinations robust to the pathogen microenvironment
.
PLOS Comput. Biol
.
14
. e1006677.
Comalada
,
M.
,
N.
Serrat
, and
J.
Xaus
.
2010
.
Cell death, it always matters
.
J. Leukoc. Biol
.
88
:
1063
1064, NaN–1066
.
Costanzo
,
M.
,
B.
VanderSluis
,
E.N.
Koch
,
A.
Baryshnikova
,
C.
Pons
,
G.
Tan
,
W.
Wang
,
M.
Usaj
,
J.
Hanchard
,
S.D.
Lee
, et al
.
2016
.
A global genetic interaction network maps a wiring diagram of cellular function
.
Science
.
353
. aaf1420.
Costanzo
,
M.
,
E.
Kuzmin
,
J.
van Leeuwen
,
B.
Mair
,
J.
Moffat
,
C.
Boone
, and
B.
Andrews
.
2019
.
Global Genetic Networks and the Genotype-to-Phenotype Relationship
.
Cell
.
177
:
85
100
.
Coyaud
,
E.
,
C.
Ranadheera
,
D.
Cheng
,
J.
Gonçalves
,
B.J.A.
Dyakov
,
E.M.N.
Laurent
,
J.
St-Germain
,
L.
Pelletier
,
A.C.
Gingras
,
J.H.
Brumell
, et al
.
2018
.
Global Interactomics Uncovers Extensive Organellar Targeting by Zika Virus
.
Mol. Cell. Proteomics
.
17
:
2242
2255
.
Cui
,
J.
,
F.
Li
, and
Z.L.
Shi
.
2019
.
Origin and evolution of pathogenic coronaviruses
.
Nat. Rev. Microbiol
.
17
:
181
192
.
Dai
,
W.
,
B.
Zhang
,
X.M.
Jiang
,
H.
Su
,
J.
Li
,
Y.
Zhao
,
X.
Xie
,
Z.
Jin
,
J.
Peng
,
F.
Liu
, et al
.
2020
.
Structure-based design of antiviral drug candidates targeting the SARS-CoV-2 main protease
.
Science
.
368
:
1331
1335
.
Davey
,
N.E.
,
G.
Travé
, and
T.J.
Gibson
.
2011
.
How viruses hijack cell regulation
.
Trends Biochem. Sci
.
36
:
159
169
.
de Chassey
,
B.
,
L.
Meyniel-Schicklin
,
J.
Vonderscher
,
P.
André
, and
V.
Lotteau
.
2014
.
Virus-host interactomics: new insights and opportunities for antiviral drug discovery
.
Genome Med
.
6
:
115
.
De Clercq
,
E.
.
2009
.
Anti-HIV drugs: 25 compounds approved within 25 years after the discovery of HIV
.
Int. J. Antimicrob. Agents
.
33
:
307
320
.
Denison
,
M.R.
,
R.L.
Graham
,
E.F.
Donaldson
,
L.D.
Eckerle
, and
R.S.
Baric
.
2011
.
Coronaviruses: an RNA proofreading machine regulates replication fidelity and diversity
.
RNA Biol
.
8
:
270
279
.
Dewe
,
J.M.
,
B.L.
Fuller
,
J.M.
Lentini
,
S.M.
Kellner
, and
D.
Fu
.
2017
.
TRMT1-Catalyzed tRNA Modifications Are Required for Redox Homeostasis To Ensure Proper Cellular Proliferation and Oxidative Stress Survival
.
Mol. Cell. Biol
.
37
. e00214-17.
Diep
,
J.
,
Y.S.
Ooi
,
A.W.
Wilkinson
,
C.E.
Peters
,
E.
Foy
,
J.R.
Johnson
,
J.
Zengel
,
S.
Ding
,
K.F.
Weng
,
O.
Laufman
, et al
.
2019
.
Enterovirus pathogenesis requires the host methyltransferase SETD3
.
Nat. Microbiol
.
4
:
2523
2537
.
Durham
,
N.D.
,
A.
Agrawal
,
E.
Waltari
,
D.
Croote
,
F.
Zanini
,
M.
Fouch
,
E.
Davidson
,
O.
Smith
,
E.
Carabajal
,
J.E.
Pak
, et al
.
2019
.
Broadly neutralizing human antibodies against dengue virus identified by single B cell transcriptomics
.
eLife
.
8
. e52384.
Eckhardt
,
M.
,
J.F.
Hultquist
,
R.M.
Kaake
,
R.
Hüttenhain
, and
N.J.
Krogan
.
2020
.
A systems approach to infectious disease
.
Nat. Rev. Genet
.
21
:
339
354
.
Eroshenko
,
N.
,
T.
Gill
,
M.K.
Keaveney
,
G.M.
Church
,
J.M.
Trevejo
, and
H.
Rajaniemi
.
2020
.
Implications of antibody-dependent enhancement of infection for SARS-CoV-2 countermeasures
.
Nat. Biotechnol
.
38
:
789
791
.
Fabre
,
E.
, and
E.
Hurt
.
1997
.
Yeast genetics to dissect the nuclear pore complex and nucleocytoplasmic trafficking
.
Annu. Rev. Genet
.
31
:
277
313
.
Ferner
,
R.E.
, and
J.K.
Aronson
.
2020
.
Remdesivir in covid-19
.
BMJ
.
369
:
m1610
.
Ferreira
,
A.R.
,
B.
Ramos
,
A.
Nunes
, and
D.
Ribeiro
.
2020
.
Hepatitis C Virus: Evading the Intracellular Innate Immunity
.
J. Clin. Med
.
9
:
790
.
Ferron
,
F.
,
L.
Subissi
,
A.T.
Silveira De Morais
,
N.T.T.
Le
,
M.
Sevajol
,
L.
Gluais
,
E.
Decroly
,
C.
Vonrhein
,
G.
Bricogne
,
B.
Canard
, et al
.
2018
.
Structural and molecular basis of mismatch correction and ribavirin excision from coronavirus RNA
.
Proc. Natl. Acad. Sci. USA
.
115
:
E162
E171
.
Flint
,
M.
,
P.
Chatterjee
,
D.L.
Lin
,
L.K.
McMullan
,
P.
Shrivastava-Ranjan
,
É.
Bergeron
,
M.K.
Lo
,
S.R.
Welch
,
S.T.
Nichol
,
A.W.
Tai
, et al
.
2019
.
A genome-wide CRISPR screen identifies N-acetylglucosamine-1-phosphate transferase as a potential antiviral target for Ebola virus
.
Nat. Commun
.
10
:
285
.
Freitas
,
B.T.
,
I.A.
Durie
,
J.
Murray
,
J.E.
Longo
,
H.C.
Miller
,
D.
Crich
,
R.J.
Hogan
,
R.A.
Tripp
, and
S.D.
Pegan
.
2020
.
Characterization and Noncovalent Inhibition of the Deubiquitinase and deISGylase Activity of SARS-CoV-2 Papain-Like Protease
.
ACS Infect. Dis.
doi:.
Fridy
,
P.C.
,
Y.
Li
,
S.
Keegan
,
M.K.
Thompson
,
I.
Nudelman
,
J.F.
Scheid
,
M.
Oeffinger
,
M.C.
Nussenzweig
,
D.
Fenyö
,
B.T.
Chait
, et al
.
2014
.
A robust pipeline for rapid production of versatile nanobody repertoires
.
Nat. Methods
.
11
:
1253
1260
.
Ghosh
,
A.K.
,
M.
Brindisi
,
D.
Shahabi
,
M.E.
Chapman
, and
A.D.
Mesecar
.
2020
.
Drug Development and Medicinal Chemistry Efforts toward SARS-Coronavirus and Covid-19 Therapeutics
.
ChemMedChem
.
15
:
907
932
.
Gordon
,
D.E.
,
G.M.
Jang
,
M.
Bouhaddou
,
J.
Xu
,
K.
Obernier
,
K.M.
White
,
M.J.
O’Meara
,
V.V.
Rezelj
,
J.Z.
Guo
,
D.L.
Swaney
, et al
.
2020
a
.
A SARS-CoV-2 protein interaction map reveals targets for drug repurposing
.
Nature.
583
:
459
468
.
Gordon
,
D.E.
,
A.
Watson
,
A.
Roguev
,
S.
Zheng
,
G.M.
Jang
,
J.
Kane
,
J.
Xu
,
J.Z.
Guo
,
E.
Stevenson
,
D.L.
Swaney
, et al
.
2020
b
.
A Quantitative Genetic Interaction Map of HIV Infection
.
Mol. Cell.
78
:
197
209.e7
.
Graham
,
R.L.
,
E.F.
Donaldson
, and
R.S.
Baric
.
2013
.
A decade after SARS: strategies for controlling emerging coronaviruses
.
Nat. Rev. Microbiol
.
11
:
836
848
.
Guise
,
A.J.
,
H.G.
Budayeva
,
B.A.
Diner
, and
I.M.
Cristea
.
2013
.
Histone deacetylases in herpesvirus replication and virus-stimulated host defense
.
Viruses
.
5
:
1607
1632
.
Gulbahce
,
N.
,
H.
Yan
,
A.
Dricot
,
M.
Padi
,
D.
Byrdsong
,
R.
Franchi
,
D.S.
Lee
,
O.
Rozenblatt-Rosen
,
J.C.
Mar
,
M.A.
Calderwood
, et al
.
2012
.
Viral perturbations of host networks reflect disease etiology
.
PLOS Comput. Biol
.
8
. e1002531.
Haga
,
K.
,
A.
Fujimoto
,
R.
Takai-Todaka
,
M.
Miki
,
Y.H.
Doan
,
K.
Murakami
,
M.
Yokoyama
,
K.
Murata
,
A.
Nakanishi
, and
K.
Katayama
.
2016
.
Functional receptor molecules CD300lf and CD300ld within the CD300 family enable murine noroviruses to infect cells
.
Proc. Natl. Acad. Sci. USA
.
113
:
E6248
E6255
.
Halstead
,
S.B.
, and
E.J.
O’Rourke
.
1977
.
Dengue viruses and mononuclear phagocytes. I. Infection enhancement by non-neutralizing antibody
.
J. Exp. Med
.
146
:
201
217
.
Hamre
,
D.
, and
M.
Beem
.
1972
.
Virologic studies of acute respiratory disease in young adults. V. Coronavirus 229E infections during six years of surveillance
.
Am. J. Epidemiol
.
96
:
94
106
.
Han
,
J.
,
J.T.
Perez
,
C.
Chen
,
Y.
Li
,
A.
Benitez
,
M.
Kandasamy
,
Y.
Lee
,
J.
Andrade
,
B.
tenOever
, and
B.
Manicassamy
.
2018
.
Genome-wide CRISPR/Cas9 Screen Identifies Host Factors Essential for Influenza Virus Replication
.
Cell Rep
.
23
:
596
607
.
Harada
,
Y.
,
Y.
Ohkawa
,
Y.
Kizuka
, and
N.
Taniguchi
.
2019
.
Oligosaccharyltransferase: A Gatekeeper of Health and Tumor Progression
.
Int. J. Mol. Sci
.
20
:
6074
.
Hart
,
T.
,
K.R.
Brown
,
F.
Sircoulomb
,
R.
Rottapel
, and
J.
Moffat
.
2014
.
Measuring error rates in genomic perturbation screens: gold standards for human functional genomics
.
Mol. Syst. Biol
.
10
:
733
.
Hart
,
T.
,
M.
Chandrashekhar
,
M.
Aregger
,
Z.
Steinhart
,
K.R.
Brown
,
G.
MacLeod
,
M.
Mis
,
M.
Zimmermann
,
A.
Fradet-Turcotte
,
S.
Sun
, et al
.
2015
.
High-Resolution CRISPR Screens Reveal Fitness Genes and Genotype-Specific Cancer Liabilities
.
Cell
.
163
:
1515
1526
.
Hartwell
,
L.H.
,
P.
Szankasi
,
C.J.
Roberts
,
A.W.
Murray
, and
S.H.
Friend
.
1997
.
Integrating genetic approaches into the discovery of anticancer drugs
.
Science
.
278
:
1064
1068
.
Hayden
,
F.G.
,
N.
Sugaya
,
N.
Hirotsu
,
N.
Lee
,
M.D.
de Jong
,
A.C.
Hurt
,
T.
Ishida
,
H.
Sekino
,
K.
Yamada
,
S.
Portsmouth
, et al;
Baloxavir Marboxil Investigators Group
.
2018
.
Baloxavir Marboxil for Uncomplicated Influenza in Adults and Adolescents
.
N. Engl. J. Med
.
379
:
913
923
.
Heaton
,
S.M.
.
2019
.
Harnessing host-virus evolution in antiviral therapy and immunotherapy
.
Clin. Transl. Immunology
.
8
. e1067.
Hillen
,
H.S.
,
G.
Kokic
,
L.
Farnung
,
C.
Dienemann
,
D.
Tegunov
, and
P.
Cramer
.
2020
.
Structure of replicating SARS-CoV-2 polymerase
.
Nature
.
Ho
,
M.R.
,
T.T.
Tsai
,
C.L.
Chen
,
M.K.
Jhan
,
C.C.
Tsai
,
Y.C.
Lee
,
C.H.
Chen
, and
C.F.
Lin
.
2017
.
Blockade of dengue virus infection and viral cytotoxicity in neuronal cells in vitro and in vivo by targeting endocytic pathways
.
Sci. Rep
.
7
:
6910
.
Hofmann
,
W.P.
,
V.
Soriano
, and
S.
Zeuzem
.
2009
.
Antiviral combination therapy for treatment of chronic hepatitis B, hepatitis C, and human immunodeficiency virus infection
.
Handb. Exp. Pharmacol
.
189
:
321
346
.
Hoofnagle
,
J.H.
2012
. Antiviral Agents. In LiverTox: Clinical and Research Information on Drug-Induced Liver Injury [Internet]. Bethesda (MD): National Institute of Diabetes and Digestive and Kidney Diseases; 2012-2018 Jun 21.
Huang
,
H.
,
W.
Kong
,
M.
Jean
,
G.
Fiches
,
D.
Zhou
,
T.
Hayashi
,
J.
Que
,
N.
Santoso
, and
J.
Zhu
.
2019
.
A CRISPR/Cas9 screen identifies the histone demethylase MINA53 as a novel HIV-1 latency-promoting gene (LPG)
.
Nucleic Acids Res
.
47
:
7333
7347
.
Huang
,
A.
,
L.A.
Garraway
,
A.
Ashworth
, and
B.
Weber
.
2020
.
Synthetic lethality as an engine for cancer drug target discovery
.
Nat. Rev. Drug Discov
.
19
:
23
38
.
Huo
,
J.
,
A.
Le Bas
,
R.R.
Ruza
,
H.M.E.
Duyvesteyn
,
H.
Mikolajek
,
T.
Malinauskas
,
T.K.
Tan
,
P.
Rijal
,
M.
Dumoux
,
P.N.
Ward
, et al
.
2020
.
Neutralizing nanobodies bind SARS-CoV-2 spike RBD and block interaction with ACE2
.
Nat. Struct. Mol. Biol
.
In press
.
Hyrina
,
A.
,
C.
Jones
,
D.
Chen
,
S.
Clarkson
,
N.
Cochran
,
P.
Feucht
,
G.
Hoffman
,
A.
Lindeman
,
C.
Russ
,
F.
Sigoillot
, et al
.
2019
.
A Genome-wide CRISPR Screen Identifies ZCCHC14 as a Host Factor Required for Hepatitis B Surface Antigen Production
.
Cell Rep
.
29
:
2970
2978.e6
.
Isaacs
,
D.
,
D.
Flowers
,
J.R.
Clarke
,
H.B.
Valman
, and
M.R.
MacNaughton
.
1983
.
Epidemiology of coronavirus respiratory infections
.
Arch. Dis. Child
.
58
:
500
503
.
Isaacson
,
M.K.
, and
H.L.
Ploegh
.
2009
.
Ubiquitination, ubiquitin-like modifiers, and deubiquitination in viral infection
.
Cell Host Microbe
.
5
:
559
570
.
Ison
,
M.G.
,
S.
Portsmouth
,
Y.
Yoshida
,
T.
Shishido
,
M.
Mitchener
,
K.
Tsuchiya
,
T.
Uehara
, and
F.G.
Hayden
.
2020
.
Early treatment with baloxavir marboxil in high-risk adolescent and adult outpatients with uncomplicated influenza (CAPSTONE-2): a randomised, placebo-controlled, phase 3 trial
.
Lancet Infect. Dis
.
Jaume
,
M.
,
M.S.
Yip
,
C.Y.
Cheung
,
H.L.
Leung
,
P.H.
Li
,
F.
Kien
,
I.
Dutry
,
B.
Callendret
,
N.
Escriou
,
R.
Altmeyer
, et al
.
2011
.
Anti-severe acute respiratory syndrome coronavirus spike antibodies trigger infection of human immune cells via a pH- and cysteine protease-independent FcγR pathway
.
J. Virol
.
85
:
10582
10597
.
Jefferson
,
T.
,
M.
Jones
,
P.
Doshi
,
E.A.
Spencer
,
I.
Onakpoya
, and
C.J.
Heneghan
.
2014
.
Oseltamivir for influenza in adults and children: systematic review of clinical study reports and summary of regulatory comments
.
BMJ
.
348
:
g2545
.
Jin
,
Z.
,
X.
Du
,
Y.
Xu
,
Y.
Deng
,
M.
Liu
,
Y.
Zhao
,
B.
Zhang
,
X.
Li
,
L.
Zhang
,
C.
Peng
, et al
.
2020
a
.
Structure of Mpro from SARS-CoV-2 and discovery of its inhibitors
.
Nature.
582
:
289
293
.
Jin
,
Z.
,
Y.
Zhao
,
Y.
Sun
,
B.
Zhang
,
H.
Wang
,
Y.
Wu
,
Y.
Zhu
,
C.
Zhu
,
T.
Hu
,
X.
Du
, et al
.
2020
b
.
Structural basis for the inhibition of SARS-CoV-2 main protease by antineoplastic drug carmofur
.
Nat. Struct. Mol. Biol.
27
:
529
532
.
Jonikas
,
M.C.
,
S.R.
Collins
,
V.
Denic
,
E.
Oh
,
E.M.
Quan
,
V.
Schmid
,
J.
Weibezahn
,
B.
Schwappach
,
P.
Walter
,
J.S.
Weissman
, et al
.
2009
.
Comprehensive characterization of genes required for protein folding in the endoplasmic reticulum
.
Science
.
323
:
1693
1697
.
Kaelin
,
W.G.
, Jr.
.
2005
.
The concept of synthetic lethality in the context of anticancer therapy
.
Nat. Rev. Cancer
.
5
:
689
698
.
Kaufmann
,
S.H.E.
,
A.
Dorhoi
,
R.S.
Hotchkiss
, and
R.
Bartenschlager
.
2018
.
Host-directed therapies for bacterial and viral infections
.
Nat. Rev. Drug Discov
.
17
:
35
56
.
Kim
,
S.J.
,
J.
Fernandez-Martinez
,
I.
Nudelman
,
Y.
Shi
,
W.
Zhang
,
B.
Raveh
,
T.
Herricks
,
B.D.
Slaughter
,
J.A.
Hogan
,
P.
Upla
, et al
.
2018
.
Integrative structure and functional anatomy of a nuclear pore complex
.
Nature
.
555
:
475
482
.
Kim
,
Y.
,
R.
Jedrzejczak
,
N.I.
Maltseva
,
M.
Wilamowski
,
M.
Endres
,
A.
Godzik
,
K.
Michalska
, and
A.
Joachimiak
.
2020
.
Crystal structure of Nsp15 endoribonuclease NendoU from SARS-CoV-2
.
Protein Sci
.
29
:
1596
1605
.
Krishnan
,
M.N.
,
A.
Ng
,
B.
Sukumaran
,
F.D.
Gilfoy
,
P.D.
Uchil
,
H.
Sultana
,
A.L.
Brass
,
R.
Adametz
,
M.
Tsui
,
F.
Qian
, et al
.
2008
.
RNA interference screen for human genes associated with West Nile virus infection
.
Nature
.
455
:
242
245
.
Krogan
,
N.J.
,
S.
Lippman
,
D.A.
Agard
,
A.
Ashworth
, and
T.
Ideker
.
2015
.
The cancer cell map initiative: defining the hallmark networks of cancer
.
Mol. Cell
.
58
:
690
698
.
Kulsuptrakul
,
J.
,
R.
Wang
,
N.L.
Meyers
,
M.
Ott
, and
A.S.
Puschnik
.
2020
.
UFMylation and TRAMP-like complexes are required for hepatitis A virus pathogenesis
.
bioRxiv
.
Laitinen
,
O.H.
,
E.
Svedin
,
S.
Kapell
,
A.
Nurminen
,
V.P.
Hytönen
, and
M.
Flodström-Tullberg
.
2016
.
Enteroviral proteases: structure, host interactions and pathogenicity
.
Rev. Med. Virol
.
26
:
251
267
.
Lam
,
T.T.
,
N.
Jia
,
Y.W.
Zhang
,
M.H.
Shum
,
J.F.
Jiang
,
H.C.
Zhu
,
Y.G.
Tong
,
Y.X.
Shi
,
X.B.
Ni
,
Y.S.
Liao
, et al
.
2020
.
Identifying SARS-CoV-2-related coronaviruses in Malayan pangolins
.
Nature
.
583
:
282
285
.
Lamphear
,
B.J.
,
R.
Yan
,
F.
Yang
,
D.
Waters
,
H.D.
Liebig
,
H.
Klump
,
E.
Kuechler
,
T.
Skern
, and
R.E.
Rhoads
.
1993
.
Mapping the cleavage site in protein synthesis initiation factor eIF-4 γ of the 2A proteases from human Coxsackievirus and rhinovirus
.
J. Biol. Chem
.
268
:
19200
19203
.
Lan
,
J.
,
J.
Ge
,
J.
Yu
,
S.
Shan
,
H.
Zhou
,
S.
Fan
,
Q.
Zhang
,
X.
Shi
,
Q.
Wang
,
L.
Zhang
, et al
.
2020
.
Structure of the SARS-CoV-2 spike receptor-binding domain bound to the ACE2 receptor
.
Nature
.
581
:
215
220
.
Li
,
G.
, and
E.
De Clercq
.
2020
.
Therapeutic options for the 2019 novel coronavirus (2019-nCoV)
.
Nat. Rev. Drug Discov
.
19
:
149
150
.
Li
,
P.
,
P.
Kaiser
,
H.W.
Lampiris
,
P.
Kim
,
S.A.
Yukl
,
D.V.
Havlir
,
W.C.
Greene
, and
J.K.
Wong
.
2016
.
Stimulating the RIG-I pathway to kill cells in the latent HIV reservoir following viral reactivation
.
Nat. Med
.
22
:
807
811
.
Li
,
Y.
,
J.
Muffat
,
A.
Omer Javed
,
H.R.
Keys
,
T.
Lungjangwa
,
I.
Bosch
,
M.
Khan
,
M.C.
Virgilio
,
L.
Gehrke
,
D.M.
Sabatini
, et al
.
2019
.
Genome-wide CRISPR screen for Zika virus resistance in human neural cells
.
Proc. Natl. Acad. Sci. USA
.
116
:
9527
9532
.
Li
,
B.
,
S.M.
Clohisey
,
B.S.
Chia
,
B.
Wang
,
A.
Cui
,
T.
Eisenhaure
,
L.D.
Schweitzer
,
P.
Hoover
,
N.J.
Parkinson
,
A.
Nachshon
, et al
.
2020
.
Genome-wide CRISPR screen identifies host dependency factors for influenza A virus infection
.
Nat. Commun
.
11
:
164
.
Lin
,
D.L.
,
N.A.
Cherepanova
,
L.
Bozzacco
,
M.R.
MacDonald
,
R.
Gilmore
, and
A.W.
Tai
.
2017
.
Dengue Virus Hijacks a Noncanonical Oxidoreductase Function of a Cellular Oligosaccharyltransferase Complex
.
MBio
.
8
. e00939-e17.
Liu
,
Y.Y.
,
J.J.
Slotine
, and
A.L.
Barabási
.
2011
.
Controllability of complex networks
.
Nature
.
473
:
167
173
.
Liu
,
D.
,
J.
Ji
,
T.P.
Ndongwe
,
E.
Michailidis
,
C.M.
Rice
,
R.
Ralston
, and
S.G.
Sarafianos
.
2015
.
Fast hepatitis C virus RNA elimination and NS5A redistribution by NS5A inhibitors studied by a multiplex assay approach
.
Antimicrob. Agents Chemother
.
59
:
3482
3492
.
Liu
,
L.
,
P.
Wang
,
M.S.
Nair
,
J.
Yu
,
M.
Rapp
,
Q.
Wang
,
Y.
Luo
,
J.F.
Chan
,
V.
Sahi
,
A.
Figueroa
, et al
.
2020
a
.
Potent neutralizing antibodies directed to multiple epitopes on SARS-CoV-2 spike
.
Nature.
Liu
,
P.
,
J.Z.
Jiang
,
X.F.
Wan
,
Y.
Hua
,
L.
Li
,
J.
Zhou
,
X.
Wang
,
F.
Hou
,
J.
Chen
,
J.
Zou
, and
J.
Chen
.
2020
b
.
Are pangolins the intermediate host of the 2019 novel coronavirus (SARS-CoV-2)?
PLoS Pathog.
16
:e1008421.
Lloyd
,
R.E.
.
2016
.
Enterovirus Control of Translation and RNA Granule Stress Responses
.
Viruses
.
8
:
93
.
Long
,
Q.X.
,
X.J.
Tang
,
Q.L.
Shi
,
Q.
Li
,
H.J.
Deng
,
J.
Yuan
,
J.L.
Hu
,
W.
Xu
,
Y.
Zhang
,
F.J.
Lv
, et al
.
2020
.
Clinical and immunological assessment of asymptomatic SARS-CoV-2 infections
.
Nat. Med
.
Lord
,
C.J.
, and
A.
Ashworth
.
2017
.
PARP inhibitors: Synthetic lethality in the clinic
.
Science
.
355
:
1152
1158
.
Lum
,
K.K.
, and
I.M.
Cristea
.
2016
.
Proteomic approaches to uncovering virus-host protein interactions during the progression of viral infection
.
Expert Rev. Proteomics
.
13
:
325
340
.
Luo
,
Y.
,
E.Y.
Jacobs
,
T.M.
Greco
,
K.D.
Mohammed
,
T.
Tong
,
S.
Keegan
,
J.M.
Binley
,
I.M.
Cristea
,
D.
Fenyö
,
M.P.
Rout
, et al
.
2016
.
HIV-host interactome revealed directly from infected cells
.
Nat. Microbiol
.
1
:
16068
.
Ma
,
H.
,
Y.
Dang
,
Y.
Wu
,
G.
Jia
,
E.
Anaya
,
J.
Zhang
,
S.
Abraham
,
J.G.
Choi
,
G.
Shi
,
L.
Qi
, et al
.
2015
.
A CRISPR-Based Screen Identifies Genes Essential for West-Nile-Virus-Induced Cell Death
.
Cell Rep
.
12
:
673
683
.
Ma
,
Y.
,
M.J.
Walsh
,
K.
Bernhardt
,
C.W.
Ashbaugh
,
S.J.
Trudeau
,
I.Y.
Ashbaugh
,
S.
Jiang
,
C.
Jiang
,
B.
Zhao
,
D.E.
Root
, et al
.
2017
.
CRISPR/Cas9 Screens Reveal Epstein-Barr Virus-Transformed B Cell Host Dependency Factors
.
Cell Host Microbe
.
21
:
580
591.e7
.
Ma
,
S.
,
S.
Jaipalli
,
J.
Larkins-Ford
,
J.
Lohmiller
,
B.B.
Aldridge
,
D.R.
Sherman
, and
S.
Chandrasekaran
.
2019
.
Transcriptomic Signatures Predict Regulators of Drug Synergy and Clinical Regimen Efficacy against Tuberculosis
.
MBio
.
10
. e02627-e19.
Ma
,
C.
,
M.D.
Sacco
,
B.
Hurst
,
J.A.
Townsend
,
Y.
Hu
,
T.
Szeto
,
X.
Zhang
,
B.
Tarbet
,
M.T.
Marty
,
Y.
Chen
, et al
.
2020
.
Boceprevir, GC-376, and calpain inhibitors II, XII inhibit SARS-CoV-2 viral replication by targeting the viral main protease
.
Cell Res
.
Mair
,
B.
,
J.
Moffat
,
C.
Boone
, and
B.J.
Andrews
.
2019
.
Genetic interaction networks in cancer cells
.
Curr. Opin. Genet. Dev
.
54
:
64
72
.
Marceau
,
C.D.
,
A.S.
Puschnik
,
K.
Majzoub
,
Y.S.
Ooi
,
S.M.
Brewer
,
G.
Fuchs
,
K.
Swaminathan
,
M.A.
Mata
,
J.E.
Elias
,
P.
Sarnow
, et al
.
2016
.
Genetic dissection of Flaviviridae host factors through genome-scale CRISPR screens
.
Nature
.
535
:
159
163
.
Mast
,
F.D.
,
A.V.
Ratushny
, and
J.D.
Aitchison
.
2014
.
Systems cell biology
.
J. Cell Biol
.
206
:
695
706
.
Menachery
,
V.D.
,
R.L.
Graham
, and
R.S.
Baric
.
2017
.
Jumping species-a mechanism for coronavirus persistence and survival
.
Curr. Opin. Virol
.
23
:
1
7
.
Mendes-Pereira
,
A.M.
,
S.A.
Martin
,
R.
Brough
,
A.
McCarthy
,
J.R.
Taylor
,
J.S.
Kim
,
T.
Waldman
,
C.J.
Lord
, and
A.
Ashworth
.
2009
.
Synthetic lethal targeting of PTEN mutant cells with PARP inhibitors
.
EMBO Mol. Med
.
1
:
315
322
.
Messina
,
J.P.
,
O.J.
Brady
,
N.
Golding
,
M.U.G.
Kraemer
,
G.R.W.
Wint
,
S.E.
Ray
,
D.M.
Pigott
,
F.M.
Shearer
,
K.
Johnson
,
L.
Earl
, et al
.
2019
.
The current and future global distribution and population at risk of dengue
.
Nat. Microbiol
.
4
:
1508
1515
.
Meyniel-Schicklin
,
L.
,
B.
de Chassey
,
P.
André
, and
V.
Lotteau
.
2012
. Viruses and interactomes in translation. Mol. Cell Proteomics. 11:M111.014738. .
Mo
,
H.
,
G.
Zeng
,
X.
Ren
,
H.
Li
,
C.
Ke
,
Y.
Tan
,
C.
Cai
,
K.
Lai
,
R.
Chen
,
M.
Chan-Yeung
, et al
.
2006
.
Longitudinal profile of antibodies against SARS-coronavirus in SARS patients and their clinical significance
.
Respirology
.
11
:
49
53
.
Mohr
,
S.
,
C.
Bakal
, and
N.
Perrimon
.
2010
.
Genomic screening with RNAi: results and challenges
.
Annu. Rev. Biochem
.
79
:
37
64
.
Monteil
,
Vanessa
,
Hyesoo
Kwon
,
Patricia
Prado
,
Astrid
Hagelkrüys
,
Reiner A
Wimmer
,
Martin
Stahl
,
Alexandra
Leopoldi
,
Elena
Garreta
,
Carmen
Hurtado Del Pozo
,
Felipe
Prosper
, et al
.
2020
.
Inhibition of SARS-CoV-2 Infections in Engineered Human Tissues Using Clinical-Grade Soluble Human ACE2
.
Cell
.
181
(
4
):
905
913.e7
.
[PubMed]
.
Mueller
,
H.
,
S.
Wildum
,
S.
Luangsay
,
J.
Walther
,
A.
Lopez
,
P.
Tropberger
,
G.
Ottaviani
,
W.
Lu
,
N.J.
Parrott
,
J.D.
Zhang
, et al
.
2018
.
A novel orally available small molecule that inhibits hepatitis B virus expression
.
J. Hepatol
.
68
:
412
420
.
Mueller
,
H.
,
A.
Lopez
,
P.
Tropberger
,
S.
Wildum
,
J.
Schmaler
,
L.
Pedersen
,
X.
Han
,
Y.
Wang
,
S.
Ottosen
,
S.
Yang
, et al
.
2019
.
PAPD5/7 Are Host Factors That Are Required for Hepatitis B Virus RNA Stabilization
.
Hepatology
.
69
:
1398
1411
.
Netea
,
M.G.
,
E.J.
Giamarellos-Bourboulis
,
J.
Domínguez-Andrés
,
N.
Curtis
,
R.
van Crevel
,
F.L.
van de Veerdonk
, and
M.
Bonten
.
2020
.
Trained Immunity: a Tool for Reducing Susceptibility to and the Severity of SARS-CoV-2 Infection
.
Cell
.
181
:
969
977
.
Ngo
,
A.M.
,
M.J.
Shurtleff
,
K.D.
Popova
,
J.
Kulsuptrakul
,
J.S.
Weissman
, and
A.S.
Puschnik
.
2019
.
The ER membrane protein complex is required to ensure correct topology and stable expression of flavivirus polyproteins
.
eLife
.
8
. e48469.
Niepel
,
M.
,
M.
Hafner
,
Q.
Duan
,
Z.
Wang
,
E.O.
Paull
,
M.
Chung
,
X.
Lu
,
J.M.
Stuart
,
T.R.
Golub
,
A.
Subramanian
, et al
.
2017
.
Common and cell-type specific responses to anti-cancer drugs revealed by high throughput transcript profiling
.
Nat. Commun
.
8
:
1186
.
Orchard
,
R.C.
,
C.B.
Wilen
,
J.G.
Doench
,
M.T.
Baldridge
,
B.T.
McCune
,
Y.C.
Lee
,
S.
Lee
,
S.M.
Pruett-Miller
,
C.A.
Nelson
,
D.H.
Fremont
, et al
.
2016
.
Discovery of a proteinaceous cellular receptor for a norovirus
.
Science
.
353
:
933
936
.
Osterhaus
,
A.
.
2001
.
Catastrophes after crossing species barriers
.
Philos. Trans. R. Soc. Lond. B Biol. Sci
.
356
:
791
793
.
Park
,
R.J.
,
T.
Wang
,
D.
Koundakjian
,
J.F.
Hultquist
,
P.
Lamothe-Molina
,
B.
Monel
,
K.
Schumann
,
H.
Yu
,
K.M.
Krupzcak
,
W.
Garcia-Beltran
, et al
.
2017
.
A genome-wide CRISPR screen identifies a restricted set of HIV host dependency factors
.
Nat. Genet
.
49
:
193
203
.
Peiris
,
J.S.
, and
J.S.
Porterfield
.
1979
.
Antibody-mediated enhancement of Flavivirus replication in macrophage-like cell lines
.
Nature
.
282
:
509
511
.
Perreira
,
J.M.
,
A.M.
Aker
,
G.
Savidis
,
C.R.
Chin
,
W.M.
McDougall
,
J.M.
Portmann
,
P.
Meraner
,
M.C.
Smith
,
M.
Rahman
,
R.E.
Baker
, et al
.
2015
.
RNASEK Is a V-ATPase-Associated Factor Required for Endocytosis and the Replication of Rhinovirus, Influenza A Virus, and Dengue Virus
.
Cell Rep
.
12
:
850
863
.
Peterson
,
E.J.R.
,
S.
Ma
,
D.R.
Sherman
, and
N.S.
Baliga
.
2016
.
Network analysis identifies Rv0324 and Rv0880 as regulators of bedaquiline tolerance in Mycobacterium tuberculosis
.
Nat. Microbiol
.
1
:
16078
.
Pfefferle
,
S.
,
J.
Schöpf
,
M.
Kögl
,
C.C.
Friedel
,
M.A.
Müller
,
J.
Carbajo-Lozoya
,
T.
Stellberger
,
E.
von Dall’Armi
,
P.
Herzog
,
S.
Kallies
, et al
.
2011
.
The SARS-coronavirus-host interactome: identification of cyclophilins as target for pan-coronavirus inhibitors
.
PLoS Pathog
.
7
. e1002331.
Pierson
,
T.C.
, and
M.S.
Diamond
.
2020
.
The continued threat of emerging flaviviruses
.
Nat. Microbiol
.
5
:
796
812
.
Pinto
,
D.
,
Y.J.
Park
,
M.
Beltramello
,
A.C.
Walls
,
M.A.
Tortorici
,
S.
Bianchi
,
S.
Jaconi
,
K.
Culap
,
F.
Zatta
,
A.
De Marco
, et al
.
2020
.
Cross-neutralization of SARS-CoV-2 by a human monoclonal SARS-CoV antibody
.
Nature
.
583
:
290
295
.
Plaisier
,
C.L.
,
S.
O’Brien
,
B.
Bernard
,
S.
Reynolds
,
Z.
Simon
,
C.M.
Toledo
,
Y.
Ding
,
D.J.
Reiss
,
P.J.
Paddison
, and
N.S.
Baliga
.
2016
.
Causal Mechanistic Regulatory Network for Glioblastoma Deciphered Using Systems Genetics Network Analysis
.
Cell Syst
.
3
:
172
186
.
Pontremoli
,
C.
,
D.
Forni
,
R.
Cagliani
,
G.
Filippi
,
L.
De Gioia
,
U.
Pozzoli
,
M.
Clerici
, and
M.
Sironi
.
2016
.
Positive Selection Drives Evolution at the Host-Filovirus Interaction Surface
.
Mol. Biol. Evol
.
33
:
2836
2847
.
Puschnik
,
A.S.
,
K.
Majzoub
,
Y.S.
Ooi
, and
J.E.
Carette
.
2017
.
A CRISPR toolbox to study virus-host interactions
.
Nat. Rev. Microbiol
.
15
:
351
364
.
Ravindran
,
V.
,
J.C.
Nacher
,
T.
Akutsu
,
M.
Ishitsuka
,
A.
Osadcenco
,
V.
Sunitha
,
G.
Bagler
,
J.M.
Schwartz
, and
D.L.
Robertson
.
2019
.
Network controllability analysis of intracellular signalling reveals viruses are actively controlling molecular systems
.
Sci. Rep
.
9
:
2066
.
Horby
,
P.
,
W.S.
Lim
,
J.R.
Emberson
,
M.
Mafham
,
J.L.
Bell
,
L.
Linsell
,
N.
Staplin
,
C.
Brightling
,
A.
Ustianowski
,
E.
Elmahi
, et al;
RECOVERY Collaborative Group
.
2020
.
Dexamethasone in Hospitalized Patients with Covid-19 - Preliminary Report
.
N. Engl. J. Med
.
Richardson
,
R.B.
,
M.B.
Ohlson
,
J.L.
Eitson
,
A.
Kumar
,
M.B.
McDougal
,
I.N.
Boys
,
K.B.
Mar
,
P.C.
De La Cruz-Rivera
,
C.
Douglas
,
G.
Konopka
, et al
.
2018
.
A CRISPR screen identifies IFI6 as an ER-resident interferon effector that blocks flavivirus replication
.
Nat. Microbiol
.
3
:
1214
1223
.
Riva
,
L.
,
S.
Yuan
,
X.
Yin
,
L.
Martin-Sancho
,
N.
Matsunaga
,
L.
Pache
,
S.
Burgstaller-Muehlbacher
,
P.D.
De Jesus
,
P.
Teriete
,
M.V.
Hull
, et al
.
2020
.
Discovery of SARS-CoV-2 antiviral drugs through large-scale compound repurposing
.
Nature
.
Robbiani
,
Davide F
,
Christian
Gaebler
,
Frauke
Muecksch
,
Julio C C
Lorenzi
,
Zijun
Wang
,
Alice
Cho
,
Marianna
Agudelo
,
Christopher O
Barnes
,
Anna
Gazumyan
,
Shlomo
Finkin
, et al
.
2020
.
Convergent antibody responses to SARS-CoV-2 in convalescent individuals
.
Nature
.
[PubMed]
.
Rodriguez-Barraquer
,
I.
,
F.
Costa
,
E.J.M.
Nascimento
,
N.
Nery
,
P.M.S.
Castanha
,
G.A.
Sacramento
,
J.
Cruz
,
M.
Carvalho
,
D.
De Olivera
,
J.E.
Hagan
, et al
.
2019
.
Impact of preexisting dengue immunity on Zika virus emergence in a dengue endemic region
.
Science
.
363
:
607
610
.
Roger
,
T.
,
J.
Lugrin
,
D.
Le Roy
,
G.
Goy
,
M.
Mombelli
,
T.
Koessler
,
X.C.
Ding
,
A.L.
Chanson
,
M.K.
Reymond
,
I.
Miconnet
, et al
.
2011
.
Histone deacetylase inhibitors impair innate immune responses to Toll-like receptor agonists and to infection
.
Blood
.
117
:
1205
1217
.
Rogers
,
Thomas F
,
Fangzhu
Zhao
,
Deli
Huang
,
Nathan
Beutler
,
Alison
Burns
,
Wan-Ting
He
,
Oliver
Limbo
,
Chloe
Smith
,
Ge
Song
,
Jordan
Woehl
, et al
.
2020
.
Isolation of potent SARS-CoV-2 neutralizing antibodies and protection from disease in a small animal model
.
Science
.
[PubMed]
.
Rout
,
M.P.
, and
A.
Sali
.
2019
.
Principles for Integrative Structural Biology Studies
.
Cell
.
177
:
1384
1403
.
Rut
,
W.
,
Z.
Lv
,
M.
Zmudzinski
,
S.
Patchett
,
D.
Nayak
,
S.J.
Snipas
,
F.
El Oualid
,
T.T.
Huang
,
M.
Bekes
,
M.
Drag
, et al
.
2020
.
Activity profiling and structures of inhibitor-bound SARS-CoV-2-PLpro protease provides a framework for anti-COVID-19 drug design
.
bioRxiv
.
Savidis
,
G.
,
W.M.
McDougall
,
P.
Meraner
,
J.M.
Perreira
,
J.M.
Portmann
,
G.
Trincucci
,
S.P.
John
,
A.M.
Aker
,
N.
Renzette
,
D.R.
Robbins
, et al
.
2016
.
Identification of Zika Virus and Dengue Virus Dependency Factors using Functional Genomics
.
Cell Rep
.
16
:
232
246
.
Scavone
,
C.
,
S.
Brusco
,
M.
Bertini
,
L.
Sportiello
,
C.
Rafaniello
,
A.
Zoccoli
,
L.
Berrino
,
G.
Racagni
,
F.
Rossi
, and
A.
Capuano
.
2020
. Current pharmacological treatments for COVID-19: What’s next? Br. J. Pharmacol.:bph.15072. https://doi.org/
Seydoux
,
E.
,
L.J.
Homad
,
A.J.
MacCamy
,
K.R.
Parks
,
N.K.
Hurlburt
,
M.F.
Jennewein
,
N.R.
Akins
,
A.B.
Stuart
,
Y.H.
Wan
,
J.
Feng
, et al
.
2020
.
Analysis of a SARS-CoV-2-Infected Individual Reveals Development of Potent Neutralizing Antibodies with Limited Somatic Mutation
.
Immunity
.
53
:
98
105.e5
.
Shah
,
P.S.
,
N.
Link
,
G.M.
Jang
,
P.P.
Sharp
,
T.
Zhu
,
D.L.
Swaney
,
J.R.
Johnson
,
J.
Von Dollen
,
H.R.
Ramage
,
L.
Satkamp
, et al
.
2018
.
Comparative Flavivirus-Host Protein Interaction Mapping Reveals Mechanisms of Dengue and Zika Virus Pathogenesis
.
Cell
.
175
:
1931
1945.e18
.
Tabata
,
K.
,
M.
Arimoto
,
M.
Arakawa
,
A.
Nara
,
K.
Saito
,
H.
Omori
,
A.
Arai
,
T.
Ishikawa
,
E.
Konishi
,
R.
Suzuki
, et al
.
2016
.
Unique Requirement for ESCRT Factors in Flavivirus Particle Formation on the Endoplasmic Reticulum
.
Cell Rep
.
16
:
2339
2347
.
Ton
,
A.T.
,
F.
Gentile
,
M.
Hsing
,
F.
Ban
, and
A.
Cherkasov
.
2020
.
Rapid Identification of Potential Inhibitors of SARS-CoV-2 Main Protease by Deep Docking of 1.3 Billion Compounds
.
Mol. Inform
.
Tse
,
L.V.
,
R.M.
Meganck
,
R.L.
Graham
, and
R.S.
Baric
.
2020
.
The Current and Future State of Vaccines, Antivirals and Gene Therapies Against Emerging Coronaviruses
.
Front. Microbiol
.
11
:
658
.
Turner
,
N.C.
,
C.J.
Lord
,
E.
Iorns
,
R.
Brough
,
S.
Swift
,
R.
Elliott
,
S.
Rayter
,
A.N.
Tutt
, and
A.
Ashworth
.
2008
.
A synthetic lethal siRNA screen identifying genes mediating sensitivity to a PARP inhibitor
.
EMBO J
.
27
:
1368
1377
.
Tyers
,
M.
, and
G.D.
Wright
.
2019
.
Drug combinations: a strategy to extend the life of antibiotics in the 21st century
.
Nat. Rev. Microbiol
.
17
:
141
155
.
Van Poelvoorde
,
L.A.E.
,
X.
Saelens
,
I.
Thomas
, and
N.H.
Roosens
.
2020
.
Next-Generation Sequencing: An Eye-Opener for the Surveillance of Antiviral Resistance in Influenza
.
Trends Biotechnol
.
38
:
360
367
.
Viswanathan
,
T.
,
S.
Arya
,
S.H.
Chan
,
S.
Qi
,
N.
Dai
,
A.
Misra
,
J.G.
Park
,
F.
Oladunni
,
D.
Kovalskyy
,
R.A.
Hromas
, et al
.
2020
.
Structural basis of RNA cap modification by SARS-CoV-2
.
Nat. Commun
.
11
:
3718
.
Walls
,
A.C.
,
Y.J.
Park
,
M.A.
Tortorici
,
A.
Wall
,
A.T.
McGuire
, and
D.
Veesler
.
2020
.
Structure, Function, and Antigenicity of the SARS-CoV-2 Spike Glycoprotein
.
Cell
.
181
:
281
292.e6
.
Wang
,
J.
.
2020
.
Fast Identification of Possible Drug Treatment of Coronavirus Disease-19 (COVID-19) through Computational Drug Repurposing Study
.
J. Chem. Inf. Model
.
60
:
3277
3286
.
Wang
,
T.
,
K.
Birsoy
,
N.W.
Hughes
,
K.M.
Krupczak
,
Y.
Post
,
J.J.
Wei
,
E.S.
Lander
, and
D.M.
Sabatini
.
2015
.
Identification and characterization of essential genes in the human genome
.
Science
.
350
:
1096
1101
.
Wang
,
Q.
,
L.
Zhang
,
K.
Kuwahara
,
L.
Li
,
Z.
Liu
,
T.
Li
,
H.
Zhu
,
J.
Liu
,
Y.
Xu
,
J.
Xie
, et al
.
2016
.
Immunodominant SARS Coronavirus Epitopes in Humans Elicited both Enhancing and Neutralizing Effects on Infection in Non-human Primates
.
ACS Infect. Dis
.
2
:
361
376
.
Wang
,
C.
,
Z.
Liu
,
Z.
Chen
,
X.
Huang
,
M.
Xu
,
T.
He
, and
Z.
Zhang
.
2020
a
.
The establishment of reference sequence for SARS-CoV-2 and variation analysis
.
J. Med. Virol.
92
:
667
674
.
Wang
,
Q.
,
J.
Wu
,
H.
Wang
,
Y.
Gao
,
Q.
Liu
,
A.
Mu
,
W.
Ji
,
L.
Yan
,
Y.
Zhu
,
C.
Zhu
, et al
.
2020
b
.
Structural Basis for RNA Replication by the SARS-CoV-2 Polymerase
.
Cell.
182
:
417
428.e13
.
Wei
,
J.
,
M.M.
Alfajaro
,
R.E.
Hanna
,
P.C.
DeWeirdt
,
M.S.
Strine
,
W.J.
Lu-Culligan
,
S.-M.
Zhang
,
V.R.
Graziano
,
C.O.
Schmitz
,
J.S.
Chen
, et al
.
2020
.
Genome-wide CRISPR screen reveals host genes that regulate SARS-CoV-2 infection
.
bioRxiv
.
Whitehead
,
S.S.
, and
T.C.
Pierson
.
2019
.
Effects of dengue immunity on Zika virus infection
.
Nature
.
567
:
467
468
.
Wiltshire
,
T.D.
,
C.A.
Lovejoy
,
T.
Wang
,
F.
Xia
,
M.J.
O’Connor
, and
D.
Cortez
.
2010
.
Sensitivity to poly(ADP-ribose) polymerase (PARP) inhibition identifies ubiquitin-specific peptidase 11 (USP11) as a regulator of DNA double-strand break repair
.
J. Biol. Chem
.
285
:
14565
14571
.
Woolhouse
,
M.E.J.
, and
L.
Brierley
.
2018
.
Epidemiological characteristics of human-infective RNA viruses
.
Sci. Data
.
5
. 180017.
World Health Organization
. Influenza (Seasonal).
2020
. Available at: https://www.who.int/en/news-room/fact-sheets/detail/influenza-(seasonal) (accessed August 7, 2020)
Wrapp
,
D.
,
D.
De Vlieger
,
K.S.
Corbett
,
G.M.
Torres
,
N.
Wang
,
W.
Van Breedam
,
K.
Roose
,
L.
van Schie
,
M.
Hoffmann
,
S.
Pöhlmann
, et al;
VIB-CMB COVID-19 Response Team
.
2020
.
Structural Basis for Potent Neutralization of Betacoronaviruses by Single-Domain Camelid Antibodies
.
Cell
.
181
:
1004
1015.e15
.
Wu
,
Y.
,
C.
Li
,
S.
Xia
,
X.
Tian
,
Y.
Kong
,
Z.
Wang
,
C.
Gu
,
R.
Zhang
,
C.
Tu
,
Y.
Xie
, et al
.
2020
.
Identification of Human Single-Domain Antibodies against SARS-CoV-2
.
Cell Host Microbe
.
27
:
891
898.e5
.
Yan
,
R.
,
Y.
Zhang
,
Y.
Li
,
L.
Xia
,
Y.
Guo
, and
Q.
Zhou
.
2020
.
Structural basis for the recognition of SARS-CoV-2 by full-length human ACE2
.
Science
.
367
:
1444
1448
.
Yang
,
H.
,
W.
Xie
,
X.
Xue
,
K.
Yang
,
J.
Ma
,
W.
Liang
,
Q.
Zhao
,
Z.
Zhou
,
D.
Pei
,
J.
Ziebuhr
, et al
.
2005
.
Design of wide-spectrum inhibitors targeting coronavirus main proteases
.
PLoS Biol
.
3
. e324.
Ye
,
Z.W.
,
S.
Yuan
,
K.S.
Yuen
,
S.Y.
Fung
,
C.P.
Chan
, and
D.Y.
Jin
.
2020
.
Zoonotic origins of human coronaviruses
.
Int. J. Biol. Sci
.
16
:
1686
1697
.
Yin
,
W.
,
C.
Mao
,
X.
Luan
,
D.D.
Shen
,
Q.
Shen
,
H.
Su
,
X.
Wang
,
F.
Zhou
,
W.
Zhao
,
M.
Gao
, et al
.
2020
.
Structural basis for inhibition of the RNA-dependent RNA polymerase from SARS-CoV-2 by remdesivir
.
Science
.
368
:
1499
1504
.
Zhang
,
R.
,
J.J.
Miner
,
M.J.
Gorman
,
K.
Rausch
,
H.
Ramage
,
J.P.
White
,
A.
Zuiani
,
P.
Zhang
,
E.
Fernandez
,
Q.
Zhang
, et al
.
2016
.
A CRISPR screen defines a signal peptide processing pathway required by flaviviruses
.
Nature
.
535
:
164
168
.
Zhang
,
L.
,
D.
Lin
,
X.
Sun
,
U.
Curth
,
C.
Drosten
,
L.
Sauerhering
,
S.
Becker
,
K.
Rox
, and
R.
Hilgenfeld
.
2020
.
Crystal structure of SARS-CoV-2 main protease provides a basis for design of improved α-ketoamide inhibitors
.
Science
.
368
:
409
412
.
Zhou
,
Y.
,
Y.
Hou
,
J.
Shen
,
Y.
Huang
,
W.
Martin
, and
F.
Cheng
.
2020
.
Network-based drug repurposing for novel coronavirus 2019-nCoV/SARS-CoV-2
.
Cell Discov
.
6
:
14
.

Author notes

*

F.D. Mast and A. Navare contributed equally to this paper.

This article is distributed under the terms of an Attribution–Noncommercial–Share Alike–No Mirror Sites license for the first six months after the publication date (see http://www.rupress.org/terms/). After six months it is available under a Creative Commons License (Attribution–Noncommercial–Share Alike 4.0 International license, as described at https://creativecommons.org/licenses/by-nc-sa/4.0/).