key: cord-0871643-vvz2xhez authors: Milani, Mario; Donalisio, Manuela; Bonotto, Rafaela Milan; Schneider, Edoardo; Arduino, Irene; Boni, Francesco; Lembo, David; Marcello, Alessandro; Mastrangelo, Eloise title: Combined in silico and in vitro approaches identified the antipsychotic drug lurasidone and the antiviral drug elbasvir as SARS-CoV2 and HCoV-OC43 inhibitors date: 2021-03-10 journal: Antiviral Res DOI: 10.1016/j.antiviral.2021.105055 sha: c5b1e5f17cdc2b608c9ee470eb471c4de0aaa2fb doc_id: 871643 cord_uid: vvz2xhez The current emergency of the novel coronavirus SARS-CoV2 urged the need for broad-spectrum antiviral drugs as the first line of treatment. Coronaviruses are a large family of viruses that already challenged humanity in at least two other previous outbreaks and are likely to be a constant threat for the future. In this work we developed a pipeline based on in silico docking of known drugs on SARS-CoV1 and 2 RNA-dependent RNA polymerase combined with in vitro antiviral assays on both SARS-CoV2 and the common cold human coronavirus HCoV-OC43. Results showed that certain drugs displayed activity for both viruses at a similar inhibitory concentration, while others were specific. In particular, the antipsychotic drug lurasidone and the antiviral drug elbasvir showed promising activity in the low micromolar range against both viruses with good selectivity index. The growth in human and animal population density through urbanization and agricultural development, combined with increased mobility and commercial transportation, land perturbation and climate change, all have an impact on virus emergence and epidemiology. Over the past decades, emerging zoonotic RNA viruses continuously gripped the world's attention, either briefly (like the severe acute respiratory syndrome coronavirus, SARS-CoV1, in 2003) , or continuously. Many RNA virus threats were considered as reemerging including Dengue, Zika, Ebola, and Chikungunya virus, and current consensus predicts that novel and potentially highly pathogenic agents will continue to emerge from the large, genetically variable natural pools present in the environment. CoVs are of particular concern due to high case-fatality rates, lack of therapeutics as well as the ability to seed outbreaks that rapidly cross geographic borders. A large number of highly diverse CoVs have been identified in animal hosts and especially in bat species, where they may have the potential to diffuse in other species including humans (Fan et al., 2019) . CoVs consist of a large and diverse family of viruses that cause multiple respiratory, gastrointestinal and neurologic diseases of varying severity, including the common cold, bronchiolitis, and pneumonia (Weiss & Leibowitz, 2011) . The CoV family is divided into four genera (alpha, beta, gamma, and delta) and thus far human CoV are limited to the alpha (HCoV-229E and HCoV-NL63) and beta genera (HCoV-OC43, HCoV-HKU1); the latter includes SARS-CoV1 and the Middle East respiratory syndrome coronavirus (MERS-CoV). A new previously unknown coronavirus, named SARS-CoV2, was discovered in December 2019 in Wuhan (Hubei province of China) and sequenced by January 2020 (Lu et al., 2020) . SARS-CoV2 is associated with an ongoing outbreak of atypical pneumonia , and was declared as 'Public Health Emergency of International Concern' on January 30 th , 2020 by the World Health Organization (www.who.int). Currently, for the COVID19 outbreak, many known drugs are under clinical investigation (Kupferschmidt, 2020) (Magro, 2020) , following different general principles and mechanisms of action: 1. the control of cytokine storms due to the hyper-reaction of the immune system against the virus (e.g. corticosteroids, (Salton et al., 2020) ); 2. the control of coagulopathy (e.g. heparin) 3. the inhibition of viral RdRp (e.g. prodrugs favipiravir and remdesivir); 4. the inhibition of viral entry (e.g. hydroxychloroquine); 5. the inhibition of the viral main protease (e.g. lopinavir and ritonavir); 5. the inhibition of viral attachment (the viral receptor ACE2 antagonist losartan). Despite their species diversity, CoVs share key genomic elements that are essential for viral replication, suggesting the possibility to design broad spectrum therapeutic agents to address the current epidemic and manage possible future outbreaks. The target considered in this work to identify new inhibitors is the highly conserved RNA dependent RNA polymerase (RdRp), that plays a crucial role in CoV replication cycle, catalyzing the synthesis of new viral RNA (Te Velthuis et al., 2012) . The cryo-EM structure of RdRp of SARS-CoV1 and of SARS-CoV2, bound to nsp7 and nsp8 co-factors, have been recently solved (PDB-id: 6NUR (Kirchdoerfer & Ward, 2019) , and 6M71 , respectively). The two proteins share a sequence identity of 96% (98% conservative substitution) and a structural r.m.s.d. of 0.54 Å (considering 788 Cαs). The exploration of libraries of molecules already in use as human drugs and well characterized in terms of human metabolism might allow the identification of antivirals that could be, in principle, rapidly tested in patients. Accordingly, we chose to analyze in silico the public database of approved/investigational drugs (DrugBank library, https://go.drugbank.com/), targeting a wide region around the active site of SARS-CoV1 RdRp. The computational work allowed the selection of 13 commercially available compounds with predicted high affinity for the protein and favorable solubility properties. J o u r n a l P r e -p r o o f These potential inhibitors (together with suramin, known to inhibit several RNA viruses) have been tested in cell-based assays against SARS-CoV2 and CoV-OC43 (Su et al., 2016) , revealing moderate to high antiviral activities for seven of them. Our results confirm antiviral properties already described for some of the selected compounds, and, more importantly, show new interesting properties for the compounds lurasidone and elbasvir as betacoronavirus inhibitors. In silico docking. The virtual Library of DrugBank (https://go.drugbank.com/) employed for the docking analysis includes FDA-approved drugs as well as experimental drugs going through the FDA approval process. Starting from the 2D sdf structures of the library (7180 molecules), we filtered out all the molecules with Mw ≥ 900 Da (keeping 7025 molecules) and then added explicit hydrogens (at pH 7.4) with the program Open Babel (O'Boyle et al., 2011) . Next, we used the program Molconvert (https://chemaxon.com/) to obtain a low energy 3D conformer for most of the molecules (6996 compounds) that were finally transformed into the autodock4 pdbqt format (adding charges and defining rotational freedom) with the AutoDockTools package (http://mgltools.scripps.edu). The atomic coordinates of SARS-CoV RdRp (PDB-id: 6NUR) bound to NSP7 and NSP8 co-factors, were chosen as docking model for CoV polymerase. Hydrogen atoms and Kollman charges (Singh & Kollman, 1984) were added using the program Python Molecule Viewer 1.5.4 (MGL-tools package http://mgltools.scripps.edu/). The protein model was then used to build a discrete grid within a box of dimensions 22.5x26.3x22.5 Å 3 (program AutoGrid (Goodford, 1985) ) as the explored volume for both the AutoDock4.2 and AutoDock Vina searches. The grid was centered near the side chain of Lys545, to include a wide region around the protein active site. During the computational analysis, the protein was J o u r n a l P r e -p r o o f constrained as rigid, whereas the small molecules were free to move. The in silico screen was divided into two runs: a fast procedure using the program AutoDock Vina (Trott & Olson, 2009 ) for the selection of the best compounds, followed by a more accurate screen using the program AutoDock4.2 (Morris et al., 2009) . The Autodock Vina docking search (energy_range=4; num_modes=4 exhaustiveness=10) produced a ranked list of all compounds, with predicted binding free energy values (ΔG) ranging between -0.9 kcal/mol and -8.9 kcal/mol. The best 118 compounds (~2% of the library, ΔG between -8.9 to -7.6 kcal/mol) were further analyzed using AutoDock4.2 (Morris et al., 2009) , with 80 hybrid GA-LS genetic algorithm runs (ga_num_evals=1750000, ga_pop_size=150). Among the molecules with higher predicted affinity for RdRp (ΔG values varying between -4.67 and -11 .7 kcal/mol), 13 FDA approved drugs were selected, taking into account commercial availability and solubility (as suggested by the theoretical logP values in the DrugBank library) properties, for in vitro assays. To confirm the binding of the 13 selected compounds to the recently released SARS-CoV2 RdRp we performed an additional in silico docking using as model the structure of Gao et al. (PDB-id: 6m71; ) with 300 hybrid GA-LS genetic algorithm runs. Furthermore, since among such drugs were present known inhibitors of viral protease we investigated in silico their binding affinity for CoV main protease (PDB-id: 6LU7; (Jin et al., 2020) ). Briefly, we explored with AutoDock4.2 a region of 15x22.5x22.5 Å 3 (after mutating the active site Cys145 to Ala) centered between the side chains of Asn142 and Gln189 to cover the whole cavity in front of the protease active site. We used the same docking procedure as described for the RdRp. Where possible, half-maximal antiviral effective concentration (EC 50 ) and SD values were calculated by regression analysis using the software GraphPad Prism 5.0 (GraphPad Software, San Diego, CA) by fitting a variable slope-sigmoidal dose-response curve. 2.3.5 Virus yield reduction assay. MRC5 cells were seeded in 24-well plates at a density of 2x10 5 cells/well and grown overnight at 37°C. The next day, infection was performed with HCoV-OC43 at a MOI of 0.02 PFU/cells in the presence of serial dilutions of compound, ranging from 100 to 0.006 μM. Following adsorption at 33°C for 1 h, the virus inoculum was removed and cells were grown in presence of compound. Supernatants were harvested and pooled as appropriate 24 h after infection and cell-free virus infectivity titers were determined by focus reduction assay in MRC5 cell monolayers. The end-point of the assay was the effective concentration of compound that reduced virus yield by 50% (EC 50 ) compared to untreated virus controls. All data were analyzed using GraphPad Prism (GraphPad Software, San Diego, CA). All results are presented as means ± standard deviations. The p value was calculated by comparing between % inhibition of infected-treated samples with % inhibition of control infected nottreated samples and one-tailed Student's T-test was used to compare groups. Significance was reported for p-value < 0.05 (*), <0.01 (**) and <0.001 (***). For the purpose of known drugs repurposing, a total of 6,996 molecules were analyzed from the DrugBank library (https://go.drugbank.com/) to target a wide region (~13,300 Å 3 ) around the active site of SARS-CoV RdRp (PDB-id: 6NUR; (Kirchdoerfer & Ward, 2019) ). The in silico screening was divided into two runs: a fast procedure for the selection of the best 2% of the library (118 compounds), with predicted binding free energy values (ΔG) from -8.9 to -7.6 kcal/mol, followed by a more accurate analysis with AutoDock4.2. In this way we ranked the 118 known drugs based on the lower ΔG value among the 80 poses tested for every compound [between -11.7 kcal/mol (predicted Ki=2.7 nM) and -4.67 kcal/mol (predicted Ki=377.6 μM)]. The list of the first best 60 compounds is reported in supplemental material (Table S1) . From our list a reasonable number of compounds was selected for cell-based assays (Table 1) , taking into account commercial availability and solubility properties. Such compounds were also submitted to an additional screening (with 300 hybrid GA-LS genetic algorithm runs) using the SARS-CoV2 RdRp structures (PDB-id: 6m71) and the scored Ki and number of conformations clustered around the one with lower ΔG are reported on Table 1 . Suramin was added to the list, since it was already known to inhibit several RNA viruses such as flavivirus (Basavannacharya & Vasudevan, 2014) (Albulescu et al., 2017) , norovirus (Mastrangelo et al., 2012) , but also chikungunya and Ebola viruses (Henß et al., 2016) . The best in silico docking pose of lurasidone and elbasvir, in the RdRp active site, is reported in Figure 1A . The protein region explored is located between thumb, fingers and palm domains and would host growing dsRNA during polymerase activity ( Figure 1A) . Such region defines a wide, complex and variable hydrophilic protein surface, and it is therefore able to host very different types of ligands. In Figure 1B Table 1 . Eight of the selected compounds showed predicted binding affinity for the protease lower than 50 nM suggesting that such known drugs could be in principle active against multiple targets. The antiviral activity of the selected compounds was assessed against two pathogenic CoVs strains: SARS-CoV2 and HCoV-OC43. 3.2.1 Antiviral activity against SARS-CoV2. A High Content Assay (HCA) has been developed to test antiviral drugs against SARS-CoV2 in vitro. Since a preliminary characterization showed sub-optimal infection of Huh7 cells, as also reported elsewhere , we decided to engineer Huh7 cells carrying the human ACE2 receptor. The panel of compounds was tested in dose response, and the assay was validated using Hydroxychloroquine as reference compound. Results are reported in Table 2 , Figure 3 and Figure S2 . All 14 compounds were tested from a starting concentration of 100 µM in 2-fold dilutions. 11 compounds showed activity at least in one tested concentration. Lurasidone and elbasvir showed the best outcomes, with EC 50 in the micromolar range and cytotoxicity >1000 µM. (Table 4) . activity of the selected compounds, focus reduction assays were performed on MRC-5 cells, as described in the Materials and Methods section and elsewhere . Results on antiviral activity and cell toxicity are reported in Table 3 , Figure 4 and Figure S3 . Among the tested compounds, alectinib showed the strongest inhibitory activity against HCoV-OC43, with an EC 50 in the low micromolar range (0.6 µM). Lurasidone and elbasvir also exerted high antiviral activity against HCoV-OC43, exhibiting EC 50 in the low micromolar range: 1.1 µM and 1.5 µM, respectively. A moderate antiviral activity was shown by tedizolid, carbenoxolone and suramin, with EC 50 ranging from 11.0 µM to 94.0 µM. The afore-mentioned compounds' antiviral effect was not a consequence of cytotoxicity, since none of the screened compounds significantly reduced cell viability at any concentration used in the antiviral assays (i.e. up to 100 µM), exhibiting CC 50 values higher than 1000 µM. By contrast, the remaining compounds did not exhibit interesting features as anti-HCoV-OC43 molecules, due to either no antiviral activity (cefoperazone, teniposide, irinotecan, natamycin), or low-moderate selectivity index (ponatinib, venetoclax, simeprevir, grazoprevir). In summary, these data showed that alectinib, lurasidone and elbasvir were endowed with strong anti-HCoV-OC43 activity (Figure 4) , with minimal toxicity and selectivity indexes higher than 600. The antiviral activity of alectinib, lurasidone, and elbasvir was further validated by means of virus yield reduction assay. All compounds were found able to effectively reduce HCoV-OC43 yield on MRC-5 cell cultures in a dose-dependent manner (Table 4) . Specifically, an EC 50 of 1.0 μM was calculated for alectinib, 8.3 μM for lurasidone, and 1.3 μM for elbasvir. Table 4 . Effects of selected compounds on SARS-CoV2 and HCoV-OC43 yield reduction and cell viability. Virus titer reduction Log 10 (mean ± SD a ) at highest tested dose (µM) EC 50 b (µM) (mean ± SD) CC 50 c (µM) (mean ± SD) J o u r n a l P r e -p r o o f Grazoprevir 3.9 ± 0.07 (100 µM) 3.3 ± 0.9 118 ± 6 Lurasidone 1.3±0.3 (100 µM) 6.4 ± 3.9 >1000 Elbasvir 0.6 ±0.2 (100 µM) 14.4 ± 7.9 >1000 Alectinib 2.9 ± 0.1 (33 µM) 1.0 ± 0.1 >1000 Lurasidone 6.3 ± 1.5 (100 µM) 8.3 ± 2.0 >1000 Elbasvir 1.4 ± 0.2 (100 µM) 1.3 ± 0.4 >1000 Virus titer reduction is the difference between log 10 values of PFU (SARS-CoV2) or FFU/mL (HCoV-OC43) from infected untreated controls and infected-treated at the highest tested dose. Mean virus titer for untreated control was 9.8 x 10 3 pfu/mL and 3 x 10 6 FFU/mL for SARS-CoV2 and HCoV-OC43, respectively. a Standard deviation. b Half maximal effective concentration obtained by virus yield reduction assay. c Half maximal cytotoxic concentration obtained by Alarma Blue (SARS-CoV2) and MTS (HCoV-OC43) assays. An accurate in silico docking search within a wide region around the SARS-CoV RdRp active site, allowed us to select 13 known drugs from the DrugBank library to be experimentally tested. We added suramin to the list, a well-known compound able to inhibit different RNA viruses (Mastrangelo et al., 2012 ) (De Clercq, 1979 ) (Albulescu et al., 2015) . To test the in vitro antiviral activity of the selected compounds, cell-based assays were established for SARS-CoV2 and HCoV-OC43 using Huh7-hACE2 and MRC-5 cells, respectively. Huh7 cells have already been widely used for screening purposes, being considered suitable model for image processing and being able to support infection of several viruses. Since preliminary analysis and previous published data indicated a limited infection capacity of SARS-CoV2 in Huh7 cells , we engineered the human ACE2 receptor in these cells to increase infection as previously proposed ) (Rothan et al., 2020) (Rut et al., 2021) . Interestingly, a rather high percentage (>60%) of the selected compounds showed some in vitro activity against one or both of the tested CoV This explanation is supported by the predicted high affinity of most of the selected compounds for another unrelated protein, the main protease of SARS-CoV2 (Table 1) . Among the tested compounds lurasidone and elbasvir displayed higher activity and lower cytotoxicity against both SARS-CoV2 and HCoV-OC43 strains. Lurasidone lead to complete inhibition of both strains with EC 50 values in the micromolar range (18 and 1.1 µM, respectively) and favorable selectivity indexes. Lurasidone is an antipsychotic drug for treatment of acute depression and schizophrenia, known to bind with a low nanomolar affinity to Dopamine-2, 5-HT1A, 5-HT2A, and 5-HT7 receptors, and with slightly lower affinity to alpha-2C adrenergic receptors (Greenberg & Citrome, 2017) . Lurasidone was already identified as a potential inhibitor of SARS-CoV2 main protease (Elmezayen et al., 2020) ; (Shamsi et al., 2020) ), and in our in silico analysis it showed good predicted binding affinity for both RdRp and the main protease. Furthermore, in a very recent paper, it has been reported that lurasidone and its derivatives displayed in silico binding affinity against five proteins (Mpro, PLpro, Spro, helicase and RdRp; (Thurakkal et al., 2021) (Massignan et al., 2020) . Since SARS-CoV2 and HCoV-OC43 share a high J o u r n a l P r e -p r o o f level of protein sequence conservation (Vijgen et al., 2005) we hypothesize that mechanisms of action of lurasidone against these viruses might be the same. Elbasvir inhibited SARS-CoV2 and HCoV-OC43 with EC 50 values in the micromolar range (about 23 and 1.5 µM, respectively). Previous in silico studies predicted elbasvir as a high affinity ligand for the RdRp, the papain-like protease and the helicase of SARS-CoV2 (Balasubramaniam & Shmookler Reis, 2020) , whereas our in silico investigation suggested a preferential binding for the main protease (predicted Kd=1.2 nM). In combination with grazoprevir (Zepatier), elbasvir has been shown to increase 25-fold remdesivir's apparent potency in preventing SARS-CoV2 replication (Nguyenla et al., 2020) . Elbasvir is an inhibitor of the HCV NS5A protein that has no homologues in coronaviruses: in light of our and previous work results, it has the potential to inhibit different viral proteins. Alectinib showed no activity against SARS-CoV2 but it is the best compound against HCoV-OC43 (EC 50 =0.6 µM, CC 50 value >1000 µM). In a very recent pre-print paper (Yaron et al., 2020) , the authors assert that alectinib limits SARS-CoV2 replication in Vero E6, A549-hACE2, Calu-3 and primary human pneumocyte cells. The activity was correlated with the lower phosphorylation levels of SRPK1/2, described as a required factor for replication of coronaviruses, including SARS-CoV2. Alectinib inhibits the anaplastic lymphoma kinase (ALK) tyrosine kinase receptor in the nM range (Kinoshita et al., 2012) , binding to the ATP binding site of the protein. Therefore, we can speculate that it might inhibit other host kinases essential for virus replication in lung epithelia and Vero E6 cells, but not in Huh7 cells. Grazoprevir inhibited SARS-CoV2 with an EC 50 around 16 µM (CC 50 value >100 µM). Published computational studies suggested grazoprevir as a potential inhibitor of the nucleocapsid protein or the papain-like protease of SARS-CoV2 (Behera et al., 2020) . Furthermore, in a very recent pre-print report (Bafna et al., 2020) it was shown that grazoprevir, together with simeprevir, synergizes with the viral polymerase inhibitor J o u r n a l P r e -p r o o f remdesivir, increasing its inhibitory activity as much as 10-fold. Grazoprevir is an inhibitor of HCV protease and it is often used for therapy in combination with elbasvir (in the drug named zepatier). Simeprevir, another inhibitor of HCV protease, has been previously shown to inhibit SARS-CoV2 in synergy with remdesivir (Lo et al., 2020) . In our experiments it showed a similar potency against both SARS-CoV2 (EC 50 about 9.3 µM) and HCoV-OC43 (EC 50 about 2.1 µM) but with low SI. From our docking results its effect is likely directed against the protease, but RdRp inhibition cannot be excluded. ABT-199, also known as venetoclax, is a potent selective Bcl2 inhibitor, which induces the apoptosis pathway. An early work showed that Bcl2 expression prevents SARS-CoV induced apoptosis (Bordi et al., 2006) . In addition, previous reports demonstrated that SARS-CoV 7a protein was dependent on Bcl2 to induce apoptosis, suggesting Bcl2 as an important host factor for virus replication and pathogenesis (Tan et al., 2007) . However, despite its good EC 50 (about 6.2 µM) against SARS-CoV2, venetoclax shows high toxicity in the tested cells. Ponatinib, an oral drug for the treatment of chronic myeloid leukemia and Philadelphia chromosome-positive acute lymphoblastic leukemia, was already proposed as SARS-CoV2 inhibitor (Nguyen et al., 2020) (Sauvat et al., 2020) (Gordon et al., 2020) , and it is shown here to inhibit SARS-CoV2 and HCoV-OC43 with a poor selective index. All the other compounds, although some of them have been described in the literature as potential inhibitors of SARS-CoV2 (i.e. teniposide (Kadioglu1 et al., n.d.) and irinotecan (B, 2020), did not show any relevant activity in either of the two viruses tested. J o u r n a l P r e -p r o o f In our work we have: 1. excluded SARS-CoV2 antiviral activity for teniposide (Kadioglu1 et al., n.d.) and irinotecan (B, 2020), selected from previous computational studies; 2. showed the ability of some of the already described anti-SARS-CoV2 compounds to inhibit also coronavirus HCoV-OC43 causing the common cold (suramin, ponatinibalthough with a low SI); and most importantly 3. showed the capability of some of the selected drugs to selectively inhibit HCoV-OC43 (alectinib) or SARS-CoV2 (grazoprevir) or be active against both CoV strains (lurasidone and elbasvir). This antiviral activity was confirmed by virus yield reduction assay for both viruses. Treatment of CoV infections with drugs that could inhibit different viral targets, as predicted for lurasidone and elbasvir, would be an effective way to lower chances of the emergence of drug resistant viral strains. Of note, in previous works it was demonstrated that alectinib (Song et al., 2015) could penetrate the blood-brain barrier (BBB) exerting its activity in the central nervous system (CNS). Since HCoV-OC43, as other coronaviruses, is able to invade the CNS (Dubé et al., 2018) , alectinib might be an interesting candidate for the treatment of HCoV-OC43 persistent infections in the brain. Moreover, the free levels of alectinib found in both plasma and cerebrospinal fluid are similar (Herden & Waller, 2018) and its EC 50 against HCoV-OC43 (0.6 uM) is lower than the maximum level attainable in human serum with daily recommended dosage (676 ng/mL corresponding to 1.4 µM) (Ly et al., 2018) . In conclusion, in this work we showed that lurasidone and elbasvir are not only potential drugs against SARS-CoV2, but that they can also inhibit the infection established by another beta-Coronavirus, HCoV-OC43. Thus, our approach allowed the identification of lead-drugs for further in vitro and clinical investigation to contain the present outbreak. Furthermore, it could contribute to the identification of broad spectrum anti-CoV inhibitors / therapies that would allow for a rapid and effective reaction to future epidemics. Suramin inhibits Zika virus replication by interfering with virus attachment and release of infectious particles Suramin inhibits chikungunya virus replication through multiple mechanisms The AI-Discovered Aetiology of COVID-19 and Rationale of the Irinotecan+Etoposide Combination Therapy for Critically Ill COVID-19 Patients Hepatitis C Virus drugs simeprevir and grazoprevir synergize with remdesivir to suppress SARS-CoV-2 replication in Cell Culture Computational Target-Based Drug Repurposing of Elbasvir, an Antiviral Drug Predicted to Bind Multiple SARS Suramin inhibits helicase activity of NS3 protein of dengue virus in a fluorescence-based high throughput assay format Simultaneous Inhibition of Entry and Replication of Novel Corona Virus by Grazoprevir: A Computational Drug Repurposing Study Bcl-2 inhibits the caspase-dependent apoptosis induced by SARS-CoV without affecting virus replication kinetics Suramin: A potent inhibitor of the reverse transcriptase of RNA tumor viruses Axonal Transport Enables Neuron-to-Neuron Propagation of Human Drug repurposing for coronavirus (COVID-19): in silico screening of known drugs against coronavirus 3CL hydrolase and protease enzymes Bat Coronaviruses in China Structure of the RNA-dependent RNA polymerase from COVID-19 virus A Computational Procedure for Determining Energetically Favorable Binding Sites on Biologically Important Macromolecules A SARS-CoV-2 protein interaction map reveals targets for drug repurposing Pharmacokinetics and Pharmacodynamics of Lurasidone Hydrochloride, a Second-Generation Antipsychotic: A Systematic Review of the Published Literature Suramin is a potent inhibitor of Chikungunya and Ebola virus cell entry Alectinib Structure of Mpro from SARS-CoV-2 and discovery of its inhibitors Identification of novel compounds against three targets of SARS CoV-2 coronavirus by combined virtual screening and supervised machine learning Design and synthesis of a highly selective, orally active and potent anaplastic lymphoma kinase inhibitor (CH5424802) Structure of the SARS-CoV nsp12 polymerase bound to nsp7 and nsp8 co-factors WHO launches global megatrial of the four most promising coronavirus treatments Auto-associative heparin nanoassemblies: A biomimetic platform against the heparan sulfate-dependent viruses HSV-1, HSV-2, HPV-16 and RSV Isolation and Full-Length Genome Characterization of SARS-CoV-2 from COVID-19 Cases in Northern Italy Hydroxychloroquine, a less toxic derivative of chloroquine, is effective in inhibiting SARS-CoV-2 infection in vitro Springer Nature Simeprevir potently suppresses SARS-CoV-2 replication and synergizes with remdesivir. BioRxiv, 2020.05.26.116020 Genomic characterisation and epidemiology of 2019 novel coronavirus: implications for virus origins and receptor binding Alectinib for advanced ALK -positive nonsmall-cell lung cancer COVID-19: Review on latest available drugs and therapies against SARS-CoV-2. Coagulation and inflammation cross-talking The cholesterol metabolite 27-hydroxycholesterol inhibits SARS-CoV-2 and is markedly decreased in COVID-19 patients Antimalarial Artefenomel Inhibits Human SARS-CoV-2 Replication in Cells while Suppressing the Receptor ACE2. ArXiv, 2004.13493 Structure-based inhibition of norovirus RNA-dependent RNA polymerases Software news and updates AutoDock4 and AutoDockTools4: Automated docking with selective receptor flexibility Potentially highly potent drugs for 2019-nCoV Discovery of SARS-CoV-2 antiviral synergy between remdesivir and approved drugs in human lung cells Open Babel: An Open chemical toolbox SARS-coronavirus-2 replication in Vero E6 cells: Replication kinetics, rapid adaptation and cytopathology Repurposing of Miglustat to inhibit the coronavirus Severe Acquired Respiratory Syndrome SARS-CoV-2 The FDAapproved gold drug auranofin inhibits novel coronavirus (SARS-COV-2) replication and attenuates inflammation in human cells SARS-CoV-2 Mpro inhibitors and activity-based probes for patient-sample imaging Suramin inhibits SARS-CoV-2 infection in cell culture by interfering with early steps of the replication cycle Prolonged Low-Dose Methylprednisolone in Patients With Severe COVID-19 Pneumonia Python: A programming language for software integration and development On-target versus off-target effects of drugs inhibiting the replication of SARS-CoV-2 Glecaprevir and Maraviroc are high-affinity inhibitors of SARS-CoV-2 main protease: Possible implication in COVID-19 therapy An approach to computing electrostatic charges for molecules Alectinib: A novel second generation anaplastic lymphoma kinase (ALK) inhibitor for overcoming clinically-acquired resistance Epidemiology, Genetic Recombination, and Pathogenesis of Coronaviruses Induction of Apoptosis by the Severe Acute Respiratory Syndrome Coronavirus 7a Protein Is Dependent on Its Interaction with the Bcl-XL Protein The SARScoronavirus nsp7+nsp8 complex is a unique multimeric RNA polymerase capable of both de novo initiation and primer extension An in-silico study on selected organosulfur compounds as potential drugs for SARS-CoV-2 infection via binding multiple drug targets AutoDock Vina: Improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading Complete Genomic Sequence of Human Coronavirus OC43: Molecular Clock Analysis Suggests a Relatively Recent Zoonotic Coronavirus Transmission Event Ligplot: A program to generate schematic diagrams of protein-ligand interactions Remdesivir and chloroquine effectively inhibit the recently emerged novel coronavirus (2019-nCoV) in vitro Springer Nature Coronavirus pathogenesis The FDA-approved drug Alectinib compromises SARS-CoV-2 nucleocapsid phosphorylation and inhibits viral infection in vitro We would like to thank all the people that contributed to the #FarmaCovid crowd founding initiative and in particular Mrs. Paola Allegretti, who with great commitment has advised many people from Perugia (Italy) to support our research work, and DoveConviene S.r.l. for its substantial contribution..