Skip to main content
Elsevier - PMC COVID-19 Collection logoLink to Elsevier - PMC COVID-19 Collection
. 2021 Jan 29;152:104762. doi: 10.1016/j.micpath.2021.104762

Inhibitory efficiency of potential drugs against SARS-CoV-2 by blocking human angiotensin converting enzyme-2: Virtual screening and molecular dynamics study

Abdul Ashik Khan a, Nabajyoti Baildya b, Tanmoy Dutta c, Narendra Nath Ghosh a,
PMCID: PMC7845504  PMID: 33524563

Abstract

Till date millions of people are infected by SARS-CoV-2 throughout the world, while no potential therapeutics or vaccines are available to combat this deadly virus. Blocking of human angiotensin-converting enzyme 2 (ACE-2) receptor, the binding site of SARS-CoV-2 spike protein, an effective strategy to discover a drug for COVID-19. Herein we have selected 24 anti-bacterial and anti-viral drugs and made a comprehensive analysis by screened them virtually against ACE-2 receptor to find the best blocker by molecular docking and molecular dynamics studies. Analysis of results revealed that, Cefpiramide (CPM) showed the highest binding affinity of −9.1 kcal/mol. Furthermore, MD study for 10 ns and evaluation of parameters like RMSD, RMSF, radius of gyration, solvent accessible surface area analysis confirmed that CPM effectively binds and blocks ACE-2 receptor efficiently.

Keywords: SARS-CoV-2, Molecular dynamics simulation, ACE-2, COVID-19, CPM

Graphical abstract

Image 1

1. Introduction

The outbreak of COVID 19 caused by severe acute respiratory syndrome-related coronavirus (SARS-CoV-2) has thrown a pandemic threat to the humanity of the world [1]. Symptoms like cold, flu and in major cases lung failure or brain failure are shown by the infected patients [2]. This virus has a huge transmission rate, and without developing a suitable therapeutic option, the human lives can't come back in their previous rhythm [3].

Coronaviruses (CoVs) belong to the family of Coronaviride with spike glycoprotein on their outer surface, which is similar to severe acute respiratory syndrome (SARS) and middle east respiratory syndrome (MERS) [4]. SARS-CoV-2 is a large enveloped positive sense RNA virus containing structural and non-structural proteins (nsps), including several accessory proteins [5]. 82% genomic sequence identity of SARS-CoV-2 with SARS-CoV helps us to gather knowledge about the pathogenesis of SARS-CoV-2 [6]. SARS-CoV and SARS-CoV-2, S protein mediated host cell invasion occurred through binding angiotensin converting enzyme-2 (ACE-2), a receptor protein [6,7]. ACE-2 is located at the surface membrane of the host cell. The infection process initiates with the interaction between viral S protein and ACE-2 on the surface of the host cell [8]. According to the analysis of Cryo-EM structure, the binding affinity of S protein (SARS-CoV-2) with ACE-2 is approximately 10–20 times greater than the SARS-CoV S protein [9,10]. So higher contagiousness and transmissibility are reflected for SARS-CoV-2 with respect to SARS-CoV [11,12]. Various attempts have been made to inhibit different proteins and enzymes that are involved in replication process of SARS-CoV-2 viz. hydroxychloroquine inhibits Mpro [13], remdesivir inhibits RdRp [14], Sofosbuvir, Ribavirin inhibit RdRp [15], extract from Azadiractha Indica inhibits PL-pro [16]. Furthermore, to discover therapeutic agents for effective blocking of ACE-2 protein, Chloroquine and hydroxychloroquine are already reported [[17], [18], [51]].

Systematic checking of drug-drug target interaction (DTI) is a standard method of drug repurposing. Various scoring functions (e.g. docking scoring function) are applied for drug repurpose [17].

In this study, we have selected 24 anti-bacterial and anti-viral drugs for virtual screening against ACE2 proteins of human body. Molecular docking study has been done with ACE2 receptor against these drugs. Molecular dynamics simulation was also performed to check the stability of ACE2 with that drugs by different plots like RMSD, RMSF, SASA radius of gyration analysis.

2. Methodology

2.1. Molecular docking studies

The crystal structure of SARS-CoV-2 spike binding site angiotensin converting enzyme-2 (ACE-2) (PDB ID:6M0J) receptor was obtained from protein data bank (http://www.rcsb.org). The structure was then cleaned using Autodock tools by removing heteroatoms and by adding necessary hydrogen atoms. The structures of the 24 drug molecules were obtained from PubChem. Using UCSF Chimera [19] the pdb files of the drugs were created for docking. Only chain-A of ACE-2 receptor was selected for docking with drugs. Autodock Vina [20] package was used for docking between the best binding sites of ACE-2 and drugs.

2.2. Molecular dynamics (MD) simulation studies

10ns MD-simulation was performed with the minimum energy conformer of the ACE-2 and Cefpiramide (CPM) complex using Gromacs (5.1) [20] with CHARMM36-march2019 force field [21]. The TIP3P water model [22] was used for solvation of the complex. Necessary topology and parameter files for the drug (CPM) were generated by using CGenFF server. A cubical box with a buffer dimension 10 × 10 × 10 Å3 was created and adequate number of Na+ ions were added to maintain electro neutrality. After performing energy minimization of the ACE-2-drug complex to 10 kJ mol−1nm−1, a 100 ps NVT equilibration was then performed at 300 K followed by another equilibration NPT for 100 ps, keeping 2fs time step.

Modified Berendsen thermostat was used for the NPT ensemble. Here also the time step was 2 fs? For both NVT and NPT equilibration, cut-offs for electrostatic and van der Waals interactions were kept at 1.0 nm. Long range interactions were calculated using smooth particle mesh Ewald (PME) method [23]. The equilibrated ensembles were finally subjected to MD simulation for 10 ns, with electrostatic and van der Waals cut off as before. PME method was used to calculate long range electrostatic interactions. A modified Berendsen thermostat and a Parinello-Rahman barostat were used with reference temperature and pressure at 300 K and 1 bar respectively. Snapshots of the trajectory were saved every 1 ns for each case.

2.3. Binding free energy calculation

Molecular mechanics Poison-Boltzmann surface area (MM-PBSA) method [24], implemented on Gromacs tool (g_mmpbsa) [25] was used for the calculation of binding free energies. The binding energies were calculated by using the following formulae

ΔGbind = Gw-complex - Gw-protein - Gw-drug (1)
Gw-complex = ⟨EMM⟩ + ⟨Gsol⟩– TS (2)
EMM = Ebonded + Enon-bonded = Ebonded + (EvdW + Eelec) (3)
Gsol = Gpolar + Gnon-polar = Gpolar + (γSASA + b) (4)
Where, Gw-complex is the total free energy of the ACE2 and drug complex, Gw-protein, Gw-drug are the free energies of the protein and drug respectively. EMM is the average MM potential energy including bonding, non-bonding energies, Gsol is the free energy of solvation including polar and non-polar energies. SASA is the solvent accessible surface area, γ is the coefficient of surface tension of solvent and b is the fitting parameter. TS is not considered by g_mmpbsa.

3. Results and discussions

24 potentially active drugs were selected for virtual screening against human angiotensin converting enzyme-2 (ACE-2) receptor. According to the previous studies, all these drugs have either anti-bacterial or anti-viral activities as shown in Table 1 . Among the selected drugs four drugs (Formoterol, Cefpiramide, Mitoxantrone and Tigecycline) are FDA approved. In the present study we have made a comprehensive analysis of the inhibitory activity of these drugs against ACE-2 receptor. Docking scores, summarised in Table 1 clearly indicate the binding efficiency of these drugs with ACE-2 receptor. All the 24 drugs showed binding affinities with ACE-2 receptor and 12 of them showed high binding affinities with a docking score greater than or equals to −7.0 kcal/mol.

Table 1.

Docking score with resource of studied potentially active drugs.

Some potentially active drug for repurposing Pubchem CID MW (g/mol) MF Docking Score (Kcal/mol) Ref
2-Amino-6-chloropurine 5360349 169.57 C5H4ClN5 −5.3 [26]
3-Pyridinemethanol 7510 109.13 C6H7NO −4.5 [27]
Ametantrone 2134 412.5 C22H28N4O4 −7.0 [28]
Arfomoterol (Formoterol) 3083544 344.4 C19H24N2O4 −7.8 [29]
Arildone 41728 368.9 C20H29ClO4 −6.0 [30]
Azanidazole 6436171 246.23 C10H10N6O2 −5.9 [31]
Bometolol 68850 472.5 C25H32N2O7 −7.5 [32]
Cefpiramide 636405 612.6 C25H24N8O7S2 −9.1 [33]
Cletoquine 71826 307.82 C16H22ClN3O −6.4 [34]
Denopamine 5311064 317.4 C18H23NO4 −6.4 [35]
Emiglitate 72004 355.4 C17H25NO7 −7.1 [36]
Flurocitabine 3034016 243.19 C9H10FN3O4 −6.5 [37]
Lasinavir 464372 659.8 C35H53N3O9 −7.8 [38]
Metossamina 688523 211.26 C11H17NO3 −5.7 [39]
Mitoxantrone 4212 444.5 C22H28N4O6 −7.2 [40]
Nifurpirinol 6436061 246.22 C12H10N2O4 −6.9 [41]
Oxiracetam 4626 158.16 C6H10N2O3 −5.1 [42]
Piroxantrone 59916 411.5 C21H25N5O4 −7.1 [43]
Stiripentol 5311454 234.29 C14H18O3 −6.4 [44]
Sulfinalol 44439 377.5 C20H27NO4S −7.6 [45]
Teloxantrone 124644 411.5 C21H25N5O4 −7.1 [46]
Tigecycline 54686904 585.6 C29H39N5O8 −7.5 [47]
Toborinone 60790 384.4 C21H24N2O5 −7.6 [48]
Xamoterol 155774 339.39 C16H25N3O5 −6.5 [49]

Cefpiramide, which showed a broad spectrum antibiotic activity showed the highest docking score against the human ACE-2 receptor of −9.1 kcal/mol. The binding affinities of the studied drugs against ACE-2 are shown in Fig. 1 .

Fig. 1.

Fig. 1

Docking score of different compounds against ACE-2.

Pharmacological analysis of these compounds showed interesting results. ADME toxicity analysis has been performed against these selected compounds.

3.1. ADMET calculations

ADMET (i.e. Absorption, Distribution, Metabolism, Excretion, and Toxicity) profiling of the compounds were performed with the help of pkCSM online server [50]. All the studied compounds have skin permeability ranging from −2.665 to −4.3. Most of the compounds do not inhibit P-glycoprotein I and II. Blood-brain barrier (BBB) permeability values are between −2.083 and +0.087, whereas CNS permeability values appear between −5.4 and −1.632. Most of the drugs do not inhibit CYP1A2, CYP2C19, CYP2C9, CYP2D6, CYP3A4 enzymes and do not interact with renal OCT2 substrate. Along with that, most of the drugs neither show AMES toxicity nor inhibit the hERGI inhibitor. The highest value of LD50 toxicity of these drugs are 3.328 mol/kg. Few of them are hepatotoxic in nature and almost all of them do not create skin sensitization. The highest value of minnow toxicity level of these drugs is 5.424. These values are tabulated in Table 2 .

Table 2.

Toxicity prediction of compounds of ACE2 inhibitor.

Compound AMES toxicity Max. tolerated dose (human) hERG I inhibitor hERG II inhibitor Oral Rat Acute Toxicity (LD50) (mol/kg) Oral Rat Chronic Toxicity (LOAEL)
(log mg/kg_bw/day)
Hepatot-oxicity Skin Sensitis-ation T.Pyriformis toxicity (log ug/L) Minnow toxicity (log mM)
2-Amino-6-chloropurine Yes 0.217 No No 2.318 1.227 No No 0.285 2.308
3-Pyridinemethanol No 0.861 No No 1.949 2.421 No Yes −0.684 2.307
Ametantrone No 0.647 No Yes 2.565 4.466 Yes No 0.285 2.718
Arfomoterol (Formoterol) No 0.144 No Yes 2.725 2.666 Yes No 0.348 1.198
Arildone No 1.029 No Yes 3.328 1.932 No No 1.471 −1.579
Azanidazole Yes 0.482 No No 1.897 1.575 Yes No 0.285 2.141
Bometolol No −0.106 No Yes 2.019 1.757 Yes No 0.327 −0.023
Cefpiramide No 0.774 No No 2.437 3.071 Yes No 0.285 4.32
Cletoquine No 0.436 No Yes 2.717 1.371 Yes No 0.672 2.641
Denopamine No −0.139 No Yes 2.83 2.101 Yes No 0.476 1.298
Emiglitate No 0.927 No No 2.215 3.652 Yes No 0.284 4.156
Flurocitabine No 1.08 No No 2.491 2.357 No No 0.311 3.591
Lasinavir No −0.242 No Yes 2.535 2.631 Yes No 0.285 −0.014
Metossamina No 0.319 No No 2.596 1.267 No No 0.227 0.226
Mitoxantrone No 0.689 No Yes 2.499 2.605 Yes No 0.285 5.057
Nifurpirinol Yes 0.687 No No 2.491 1.987 No No 0.761 1.514
Oxiracetam No 1.29 No No 1.839 1.871 No No −0.52 3.878
Piroxantrone No 0.76 No Yes 2.479 3.902 Yes No 0.285 3.195
Stiripentol No 0.777 No No 1.867 1.965 No No 2.045 0.612
Sulfinalol No 0.262 No Yes 2.69 2.106 Yes No 0.403 0.184
Teloxantrone Yes 0.647 No Yes 2.483 3.489 Yes No 0.285 4.036
Tigecycline No 0.622 No Yes 2.274 3.327 No No 0.285 5.424
Toborinone No −0.294 No Yes 2.883 1.297 Yes No 0.308 1.311
Xamoterol No −0.235 No No 1.536 1.239 Yes No 0.27 5.022

Compounds which showed potential binding affinities with ACE2 are shown in Fig. 2 . The interaction site of the docked structure of the drugs with ACE2 is given in the figure.

Fig. 2.

Fig. 2

Docked structure of ACE2 receptor with few drugs having high binding affinity.

Fig. 3 represents the docked structure of Cefpiramide (CPM) with ACE2. From Fig. 3 it is clear that there is strong binding interaction between the drug and ACE2 receptor due to the formation of H-bonding, electrostatic and van der Waal interactions. The nearest residues are shown in the 2D contour plot (left panel) as well as in the 3D structure is shown in right panel. The H-bonding distances (in angstrom) are given in the 2D structure and the donor and acceptor sites within the docked cavity are given in the right panel 3D structure.

Fig. 3.

Fig. 3

Docked structure of Cefpiramide (CPM) docked ACE2.

Analysing the ADME data and binding energies obtained from docking results we have chosen the drug Cefpiramide (CPM BE = −9.1 kcal/mol) to study the MD-simulation against ACE-2. The RMSD plot of the docked CPM against ACE-2 is shown in Fig. 4 . We found a profound stabilization of the docked structure after 2 ns?as compared to the undocked one.

Fig. 4.

Fig. 4

RMSD plot (a) and RMSF plot for undocked and docked ACE2.

Furthermore, we also note that after 2 ns the RMSD fluctuation of the docked structure is relatively low with respect to the undocked one suggesting during the progress of MD-simulation, the drug moiety interacts strongly within the cavity of the ACE-2. RMSF plot as shown in Fig. 4b reveals that the fluctuations of residues for the docked structure are quite low compared to the undocked one. Radius of gyration (Rg) indicates the compactness of a system. With increasing the value of Rg, the compactness of the system also increases. Rg for the docked and undocked structure is shown in Fig. 5 a. Rg for the docked structure is quite high as compared to the undocked one which confirms that after docking the drug (CPM) is nicely fitted within the cavity of ACE-2. We also analyzed the surface accessible surface area (SASA) plot for undocked and docked ACE-2.

Fig. 5.

Fig. 5

Radius of gyration plot (a) and SASA plot (b) of undocked and CPM docked ACE-2.

Fig. 5b represents the SASA plot of undocked and docked ACE-2. A closer look to Fig. 5b revealed that after 4 ns the docked structure corresponds to the higher SASA value compared to the undocked one suggesting the entry of the drug stabilizes ACE-2 conformation. Fig. 6 represents the sequence analysis of undocked and docked ACE-2. From Fig. 6 it is clear that there is a substantial structural alternation on amino acids in ACE2 before and after docking. Residue numbers from 280 to 381 of ACE2 were mostly affected by the drug CPM. This result is further elevated by the contribution energy with respect to residue number, as shown in Fig. 7 .

Fig. 6.

Fig. 6

Structural alteration of amino acid in undocked and docked ACE-2.

Fig. 7.

Fig. 7

Contribution of residues to binding energy in the docked structure of ACE2.

The binding energy of CPM against ACE2 showed a high value of −79.958 ± 18.653 kJ/mol. As shown in Table 3 all the interaction energies between ACE2 and CPM showed a high value confirming profound conformational changes of ACE2 by CPM.

Table 3.

Different types of interaction energies between ACE2 and CPM.

System Binding Energy (kJ/mol) van der Waal energy (kJ/mol) Electrostatic energy (kJ/mol) Polar solvation energy (kJ/mol) SASA energy (kJ/mol)
ACE2+CPM −79.958 ± 18.653 −172.786 ± 15.261 −59.125 ± 15.418 170.592 ± 24.815 −18.638 ± 1.223

Fig. 8 represents the binding free energy, MM energy, polar solvation and non-polar solvation energy. The binding free energy of polar and non-polar parts of the docked structure with respect to time is shown in Fig. 8 c & d respectively. During MD-simulation non-polar binding free energy (van der Waal interaction) decreased indicating much stronger binding of the drug CPM in the ACE2 cavity. A stronger binding between CPM and ACE2 is indicated by the substantial structural change in ACE2 receptor. Fig. 8 a & b represented the binding energy and MM energy of CPM against ACE2 during MD-simulation. We found the average binding energy of −79.958 kJ/mol and the average MM energy of −231.327 kJ/mol.

Fig. 8.

Fig. 8

Variation of different binding energy components: (a) Binding energy, (b)ΔEMM, (c) ΔGpolar and (d) ΔGnon-polar with time.

Conformational changes during MD-simulation is represented in Fig. 9 . These changes are captured at each nanosecond and revealed that these changes are profound. RMSD plot indicates a significant change in the structure after two ns. Hence it is clear that CPM has a considerable impact on the conformation of ACE-2.

Fig. 9.

Fig. 9

Conformational changes of the docked structure of ACE2 at each nanosecond during MD-simulation [t = (n-1)ns, brown and t = n ns, sky-blue; n = 0–10 ns]

4. Conclusion

In the present work, we have virtually screened 24 potentially active anti-bacterial and anti-viral drugs against SARS-CoV-2 binding receptor, ACE-2. ADMET profiling confirms that these drugs are suitable to use against COVID-19 treatment. The screening results revealed that cefpiramide (CPM) showed a decent binding affinity SARS-CoV-2 human ACE-2 receptor. CPM entry to the cavity of ACE-2 is facilitated by forming H-bonding interactions and electrostatic interactions. Furthermore, MD-simulation of CPM against ACE-2 showed a striking result by stabilizing ACE-2 conformation. The total disruption of ACE-2 sequence indicates that the drug has a significant impact on the receptor. Considerable stabilization and effective blocking of ACE-2 by CPM are confirmed by RMSD, RMSF analysis along with the binding energy calculation. We believe that the drug, CPM can be anticipated as an effective blocker for ACE-2 receptor and showed potential inhibitory activity against SARS-CoV-2.

Data availability

Data is available upon request to the corresponding author.

Declaration of competing interest

The authors declare no conflicting interest in the present work.

References

  • 1.Xu X., Chen P., Wang J., Feng J., Zhou H., Li X., et al. Evolution of the novel coronavirus from the ongoing Wuhan outbreak and modeling of its spike protein for risk of human transmission. Sci. China Life Sci. 2020;63:457–460. doi: 10.1007/s11427-020-1637-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Jain V., Yuan J.-M. Predictive symptoms and comorbidities for severe COVID-19 and intensive care unit admission: a systematic review and meta-analysis. International Journal of Public Health. 2020:1. doi: 10.1007/s00038-020-01390-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Jayaweera M., Perera H., Gunawardana B., Manatunge J. Transmission of COVID-19 virus by droplets and aerosols: a critical review on the unresolved dichotomy. Environ. Res. 2020:109819. doi: 10.1016/j.envres.2020.109819. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Su S., Wong G., Shi W., Liu J., Lai A.C., Zhou J., et al. Epidemiology, genetic recombination, and pathogenesis of coronaviruses. Trends Microbiol. 2016;24:490–502. doi: 10.1016/j.tim.2016.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Indwiani Astuti Y. 2020. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2): an overview of viral structure and host response. Diabetes & metabolic syndrome. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Liu W., Morse J.S., Lalonde T., Xu S. Learning from the past: possible urgent prevention and treatment options for severe acute respiratory infections caused by 2019-nCoV. Chembiochem. 2020;21(5):730–738. doi: 10.1002/cbic.202000047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Chan J.F.-W., Kok K.-H., Zhu Z., Chu H., To K.K.-W., Yuan S., et al. Genomic characterization of the 2019 novel human-pathogenic coronavirus isolated from a patient with atypical pneumonia after visiting Wuhan. Emerg. Microb. Infect. 2020;9:221–236. doi: 10.1080/22221751.2020.1719902. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Hofmann H., Pöhlmann S. Cellular entry of the SARS coronavirus. Trends Microbiol. 2004;12:466–472. doi: 10.1016/j.tim.2004.08.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Lu R., Zhao X., Li J., Niu P., Yang B., Wu H., et al. Genomic characterisation and epidemiology of 2019 novel coronavirus: implications for virus origins and receptor binding. Lancet. 2020;395:565–574. doi: 10.1016/S0140-6736(20)30251-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Wrapp D., Wang N., Corbett K.S., Goldsmith J.A., Hsieh C.-L., Abiona O., et al. Cryo-EM structure of the 2019-nCoV spike in the prefusion conformation. Science. 2020;367:1260–1263. doi: 10.1126/science.abb2507. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Tang B., Bragazzi N.L., Li Q., Tang S., Xiao Y., Wu J. An updated estimation of the risk of transmission of the novel coronavirus (2019-nCov) Infectious disease modelling. 2020;5:248–255. doi: 10.1016/j.idm.2020.02.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Li R., Pei S., Chen B. 2020. Substantial undocumented infection facilitates the rapid dissemination of novel coronavirus (SARS-CoV2) published online ahead of print March 16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Baildya N., Ghosh N.N., Chattopadhyay A.P. Inhibitory activity of hydroxychloroquine on COVID-19 main protease: an insight from MD-simulation studies. J. Mol. Struct. 2020:128595. doi: 10.1016/j.molstruc.2020.128595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Tchesnokov E.P., Feng J.Y., Porter D.P., Götte M.J.V. Mechanism of inhibition of Ebola virus RNA-dependent RNA polymerase by remdesivir. 2019;11:326. doi: 10.3390/v11040326. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Elfiky A.A. Anti-HCV, nucleotide inhibitors, repurposing against COVID-19. Life Sci. 2020:117477. doi: 10.1016/j.lfs.2020.117477. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Baildya N., Khan A.A., Ghosh N.N., Dutta T., Chattopadhyay A.P. Screening of potential drug from Azadiractha Indica (Neem) extracts for SARS-CoV-2: an insight from molecular docking and MD-simulation studies. J. Mol. Struct. 2020:129390. doi: 10.1016/j.molstruc.2020.129390. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Fantini J., Di Scala C., Chahinian H., Yahi N. Structural and molecular modeling studies reveal a new mechanism of action of chloroquine and hydroxychloroquine against SARS-CoV-2 infection. Int. J. Antimicrob. Agents. 2020:105960. doi: 10.1016/j.ijantimicag.2020.105960. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Kruse R.L. Therapeutic strategies in an outbreak scenario to treat the novel coronavirus originating in Wuhan, China. F1000Research. 2020:9. doi: 10.12688/f1000research.22211.2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Pettersen E.F., Goddard T.D., Huang C.C., Couch G.S., Greenblatt D.M., Meng E.C., et al. UCSF Chimera—a visualization system for exploratory research and analysis. J. Comput. Chem. 2004;25:1605–1612. doi: 10.1002/jcc.20084. [DOI] [PubMed] [Google Scholar]
  • 20.Trott O., Olson A.J. AutoDock Vina: improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. J. Comput. Chem. 2010;31:455–461. doi: 10.1002/jcc.21334. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Lee S., Tran A., Allsopp M., Lim J.B. Hénin Jrm, Klauda JB. CHARMM36 united atom chain model for lipids and surfactants. J. Phys. Chem. B. 2014;118:547–556. doi: 10.1021/jp410344g. [DOI] [PubMed] [Google Scholar]
  • 22.Boonstra S., Onck P.R., van der Giessen E. CHARMM TIP3P water model suppresses peptide folding by solvating the unfolded state. J. Phys. Chem. B. 2016;120:3692–3698. doi: 10.1021/acs.jpcb.6b01316. [DOI] [PubMed] [Google Scholar]
  • 23.Abraham M.J., Gready J.E. Optimization of parameters for molecular dynamics simulation using smooth particle-mesh Ewald in GROMACS 4.5. J. Comput. Chem. 2011;32:2031–2040. doi: 10.1002/jcc.21773. [DOI] [PubMed] [Google Scholar]
  • 24.Kumari R., Kumar R., Consortium OSDD. Lynn A. g_mmpbsa A GROMACS tool for high-throughput MM-PBSA calculations. J. Chem. Inf. Model. 2014;54:1951–1962. doi: 10.1021/ci500020m. [DOI] [PubMed] [Google Scholar]
  • 25.Baker N.A., Sept D., Joseph S., Holst M.J., McCammon J.A. Electrostatics of nanosystems: application to microtubules and the ribosome. Proc. Natl. Acad. Sci. Unit. States Am. 2001;98:10037–10041. doi: 10.1073/pnas.181342398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Krasnov V., Vigorov A.Y., Gruzdev D., Levit G., Kravchenko M., Skornyakov S., et al. Tuberculostatic activity of 2-amino-6-chloropurine derivatives. Pharmaceut. Chem. J. 2017;51:769–772. [Google Scholar]
  • 27.Durdagi S., Aksoydan B., Dogan B., Sahin K., Shahraki A., Birgül-İyison N. 2020. Screening of clinically approved and investigation drugs as potential inhibitors of SARS-CoV-2 main protease and spike receptor-binding domain bound with ACE2 COVID19 target proteins: a virtual drug repurposing study. [Google Scholar]
  • 28.Piccart M., Rozencweig M., Abele R., Cumps E., Dodion P., Dupont D., et al. Phase I clinical trial with ametantrone (NSC-287513) Eur. J. Cancer Clin. Oncol. 1981;17:775–779. doi: 10.1016/0014-2964(81)90233-4. [DOI] [PubMed] [Google Scholar]
  • 29.Jenkins C.R., Bateman E.D., Sears M.R., O'Byrne P.M. 2020. What have we learnt about asthma control from trials of budesonide/formoterol as maintenance and reliever? Respirology. [DOI] [PubMed] [Google Scholar]
  • 30.Kim K., Sapienza V., Carp R. Antiviral activity of arildone on deoxyribonucleic acid and ribonucleic acid viruses. Antimicrob. Agents Chemother. 1980;18:276–280. doi: 10.1128/aac.18.2.276. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Marchionni M., Degli Innocenti A., Penna C. Combined systemic and topical treatment of trichomoniasis vaginalis with azanidazol. Clin. Exp. Obstet. Gynecol. 1981;8:18–20. [PubMed] [Google Scholar]
  • 32.Watanabe T., Watanabe Y., Hasegawa Y., Kudo Y., Kawashima K., Sokabe H. Acute and subchronic effects of bometolol on blood pressure in hypertensive rats. J. Pharmacobio-Dyn. 1981;4:505–512. doi: 10.1248/bpb1978.4.505. [DOI] [PubMed] [Google Scholar]
  • 33.Wang H., Yu Y., Xie X., Wang C., Zhang Y., Yuan Y., et al. In-vitro antibacterial activities of cefpiramide and other broad-spectrum antibiotics against 440 clinical isolates in China. J. Infect. Chemother. 2000;6:81–85. doi: 10.1007/pl00012156. [DOI] [PubMed] [Google Scholar]
  • 34.Qu Y., Noe G., Breaud A.R., Vidal M., Clarke W.A., Zahr N., et al. Development and validation of a clinical HPLC method for the quantification of hydroxychloroquine and its metabolites in whole blood. Future science OA. 2015:1. doi: 10.4155/fso.15.24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Denopamine Ishide T. A selective 1-receptor agonist and a new coronary vasodilator. Curr. Med. Res. Opin. 2002;18:407–413. doi: 10.1185/030079902125001119. [DOI] [PubMed] [Google Scholar]
  • 36.Brendel E., Wingender W. Clinical pharmacology of glucosidase inhibitors. Oral Antidiabetics: Springer. 1996:611–632. [Google Scholar]
  • 37.Liss R.H., Charest M.C., Mead J. Comparative ultrastructure of submaxillary salivary glands from mice treated with cytosine arabinoside, cyclocytidine, and anhydro-ara-5-fluorocytidine. Canc. Treat Rep. 1976;60:881–888. [PubMed] [Google Scholar]
  • 38.Organization Wh International nonproprietary names for pharmaceutical substances (INN): recommended INN: list 69. WHO Drug Inf. 2013;27:41–93. [Google Scholar]
  • 39.Li B., Wang X., Rutz B., Wang R., Tamalunas A., Strittmatter F., et al. The STK16 inhibitor STK16-IN-1 inhibits non-adrenergic and non-neurogenic smooth muscle contractions in the human prostate and the human male detrusor. N. Schmied. Arch. Pharmacol. 2019:1–14. doi: 10.1007/s00210-019-01797-x. [DOI] [PubMed] [Google Scholar]
  • 40.Parker C., Waters R., Leighton C., Hancock J., Sutton R., Moorman A.V., et al. Effect of mitoxantrone on outcome of children with first relapse of acute lymphoblastic leukaemia (ALL R3): an open-label randomised trial. Lancet. 2010;376:2009–2017. doi: 10.1016/S0140-6736(10)62002-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Kennedy C., Gill K., Walsh P. The effects of nifurpirinol treatment on the activities of hepatic xenobiotic transforming enzymes in the gulf toadfish, Opsanus beta (Goode and Bean) J. Fish. Dis. 1990;13:525–529. [Google Scholar]
  • 42.Malykh A.G., Sadaie M.R. Piracetam and piracetam-like drugs. Drugs. 2010;70:287–312. doi: 10.2165/11319230-000000000-00000. [DOI] [PubMed] [Google Scholar]
  • 43.Berg S.L., Balis F.M., Godwin K.S., Poplack D.G. Pharmacokinetics, cerebrospinal fluid penetration, and metabolism of piroxantrone in the Rhesus monkey. Invest. N. Drugs. 1993;11:255–261. doi: 10.1007/BF00874424. [DOI] [PubMed] [Google Scholar]
  • 44.Frampton J.E. Stiripentol: a review in dravet syndrome. Drugs. 2019;79:1785–1796. doi: 10.1007/s40265-019-01204-y. [DOI] [PubMed] [Google Scholar]
  • 45.Sybertz E.J., Baum T., Pula K.K., Nelson S., Eynon E., Sabin C. Studies on the mechanism of the acute antihypertensive and vasodilator actions of several β-adrenoceptor antagonists. J. Cardiovasc. Pharmacol. 1982;4:749–758. doi: 10.1097/00005344-198209000-00009. [DOI] [PubMed] [Google Scholar]
  • 46.Leteurtre F., Kohlhagen G., Paull K.D., Pommier Y. Topoisomerase II inhibition and cytotoxicity of the anthrapyrazoles DuP 937 and DuP 941 (Losoxantrone) in the National Cancer Institute preclinical antitumor drug discovery screen. J. Natl. Cancer Inst.: Journal of the National Cancer Institute. 1994;86:1239–1244. doi: 10.1093/jnci/86.16.1239. [DOI] [PubMed] [Google Scholar]
  • 47.Rose W.E., Rybak M.J. Tigecycline: first of a new class of antimicrobial agents. Pharmacotherapy. 2006;26:1099–1110. doi: 10.1592/phco.26.8.1099. [DOI] [PubMed] [Google Scholar]
  • 48.Kageyama K., Mizobe T., Nozuchi S., Hiramatsu N., Nakajima Y., Aoki H. Toborinone and olprinone, phosphodiesterase III inhibitors, inhibit human platelet aggregation due to the inhibition of both calcium release from intracellular stores and calcium entry. J. Anesth. 2004;18:107–112. doi: 10.1007/s00540-004-0228-6. [DOI] [PubMed] [Google Scholar]
  • 49.Marlow H. Xamoterol, a beta 1-adrenoceptor partial agonist: review of the clinical efficacy in heart failure. Br. J. Clin. Pharmacol. 1989;28:23S–30S. doi: 10.1111/j.1365-2125.1989.tb03570.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Pires D.E., Blundell T.L., Ascher D.B. pkCSM: predicting small-molecule pharmacokinetic and toxicity properties using graph-based signatures. J. Med. Chem. 2015;58:4066–4072. doi: 10.1021/acs.jmedchem.5b00104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Baildya N., Ghosh N.N., Chattopadhyay A.P. Inhibitory capacity of Chloroquine against SARS-COV-2 by effective binding with Angiotensin converting enzyme-2 receptor: An insight from molecular docking and MD-simulation studies. J. Mol. Struct. 2021;1230 doi: 10.1016/j.molstruc.2021.129891. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

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

Data Availability Statement

Data is available upon request to the corresponding author.


Articles from Microbial Pathogenesis are provided here courtesy of Elsevier

RESOURCES