ArticlePDF Available

In silico analysis and identification of promising hits against 2019 novel coronavirus 3C-like main protease enzyme

Authors:
  • Chosun University College of Medicine
  • JIS University

Abstract and Figures

The recent outbreak of the 2019 novel coronavirus disease (COVID-19) has been proved as a global threat. No particular drug or vaccine has not yet been discovered which may act specifically against severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) and causes COVID-19. For this highly infectious virus, 3CL-like main protease (3CLpro) plays a key role in the virus life cycle and can be considered as a pivotal drug target. Structure-based virtual screening of DrugBank database resulted in 20 hits against 3CLpro. Atomistic 100 ns molecular dynamics of five top hits and binding energy calculation analyses were performed for main protease-hit complexes. Among the top five hits, Nafarelin and Icatibant affirmed the binding energy (g_MMPBSA) of –712.94 kJ/mol and –851.74 kJ/mol, respectively. Based on binding energy and stability of protein-ligand complex; the present work reports these two drug-like hits against SARS-CoV-2 main protease. Communicated by Ramaswamy H. Sarma
Content may be subject to copyright.
Full Terms & Conditions of access and use can be found at
https://www.tandfonline.com/action/journalInformation?journalCode=tbsd20
Journal of Biomolecular Structure and Dynamics
ISSN: 0739-1102 (Print) 1538-0254 (Online) Journal homepage: https://www.tandfonline.com/loi/tbsd20
In silico analysis and identification of promising
hits against 2019 novel coronavirus 3C-like main
protease enzyme
Shilpa Chatterjee, Arindam Maity, Suchana Chowdhury, Md Ataul Islam, Ravi
K. Muttinini & Debanjan Sen
To cite this article: Shilpa Chatterjee, Arindam Maity, Suchana Chowdhury, Md Ataul Islam, Ravi
K. Muttinini & Debanjan Sen (2020): In silico analysis and identification of promising hits against
2019 novel coronavirus 3C-like main protease enzyme, Journal of Biomolecular Structure and
Dynamics, DOI: 10.1080/07391102.2020.1787228
To link to this article: https://doi.org/10.1080/07391102.2020.1787228
View supplementary material
Published online: 01 Jul 2020.
Submit your article to this journal
View related articles
View Crossmark data
In silico analysis and identification of promising hits against 2019 novel
coronavirus 3C-like main protease enzyme
Shilpa Chatterjee
a
, Arindam Maity
b
, Suchana Chowdhury
c
, Md Ataul Islam
d,e
, Ravi K. Muttinini
f
and
Debanjan Sen
c
a
Department of Biomedical Science, Chosun University, Gwangju, South Korea;
b
School of Pharmaceutical Technology, Adamas University,
Kolkata, India;
c
BCDA College of Pharmaceutical Technology, Hridaypur, Kolkata, India;
d
Division of Pharmacy and Optometry, School of
Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK;
e
School of Health Sciences, University
of Kwazulu-Natal, Durban, South Africa;
f
Immunocure Discovery solution Pvt. Ltd, IKP, Hyderabad, India
Communicated by Ramaswamy H. Sarma
ABSTRACT
The recent outbreak of the 2019 novel coronavirus disease (COVID-19) has been proved as a global
threat. No particular drug or vaccine has not yet been discovered which may act specifically against
severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) and causes COVID-19. For this highly
infectious virus, 3CL-like main protease (3CL
pro
) plays a key role in the virus life cycle and can be con-
sidered as a pivotal drug target. Structure-based virtual screening of DrugBank database resulted in 20
hits against 3CL
pro
. Atomistic 100ns molecular dynamics of five top hits and binding energy calcula-
tion analyses were performed for main protease-hit complexes. Among the top five hits, Nafarelin and
Icatibant affirmed the binding energy (g_MMPBSA) of 712.94 kJ/mol and 851.74 kJ/mol, respectively.
Based on binding energy and stability of protein-ligand complex; the present work reports these two
drug-like hits against SARS-CoV-2 main protease.
ARTICLE HISTORY
Received 27 April 2020
Accepted 16 June 2020
KEYWORDS
COVID-19; cobicistat; drug
hits; icatibant; main
protease enzyme; nafarelin
Introduction
The coronaviruses (CoVs) belong to the Coronaviridae family,
a positive single-stranded largest RNA virus (Tyrrell & Bynoe,
1966). The CoVs possesses of four genera included
Alphacoronavirus, Betacoronavirus, Gammacoronavirus, and
Deltacoronavirus which are responsible for disease associated
with the event of a severe acute respiratory syndrome. In the
past two decades, Middle East respiratory syndrome corona-
virus (MERS-CoV) and severe acute respiratory syndrome cor-
onavirus (SARS-CoV), a type of b-coronavirus infected 1
million humans worldwide (Boopathi et al., 2020). The 2019
novel coronavirus is currently known as SARS-CoV-2 was
identified at Wuhan fish market in China in the year 2019
(Wu et al., 2020), which turned out to be an epidemic and
already an outbreak in almost in every country worldwide
(Hasan et al., 2020). The disease caused by the SARS-CoV is
known as COVID-19 and has infected more than 2.8 million
people around the world (www.worldometers.info/corona-
virus) with 20% death rates. World Health Organization
(WHO) declared this infection as a global threat and entire
world compelled to locked down. All over the world
researchers are working very hard to find suitable drugs or
vaccines for effective management of COVID-19 or to inhibit
its spreading. Li et al. (2020) reports the re-purposing drug
options for COVID-19, and suggested to use antiviral drugs
Favipiravir, Ribavirin, Remdesivir, Galidesivir (Li & De Clercq,
2020) for this purpose. A recent study reports, the genome
of coronavirus possesses a long RNA strand (Chen et al.,
2020) which acts as a messenger RNA when it infects the
host cell, regulate the synthesis of two crucial long polypro-
teins required for viral replication. These polyproteins are
associated with the events of viral replication or transcription
process to produce other viral structural proteins to form
virions and protease enzymes. The formed protease enzymes
are responsible for cutting polyproteins to produce necessary
functional proteins (Li et al., 2020). The 3CL
pro
is the main
protease (Islam et al., 2020) which is encoded by non-struc-
tural protein 5 (NSP5), involved in polyprotein cleavage,
immune regulation and important for viral replication. The
3CL
pro
in SARS-COV-2 has substrate recognition pockets use-
ful for binding with viral polyprotein including the activator
of transcription 2, signal transducer and the NF-jB essential
modulator signalling protein (Perlman & Netland, 2009;
Wang et al., 2016; Yang et al., 2005; Zhu et al., 2017).
Targeting 3CL
pro
is believed to act against the virus by
inhibiting viral maturation and refurbishing the natural
immune response (Lamarre et al., 2003). Designing or identi-
fication of small molecules has a close structural similarity
with the natural substrate of an enzyme, exploit superior
interaction with binding site residues and drug resistance-
related issues can be avoided (Deeks et al., 1997).
CONTACT Debanjan Sen debanjansen48@gmail.com Department of Pharmaceutical Technology, BCDA College of Pharmacy and Technology, 78 Jessore
Road, Hridaypur, Kolkata 700127, India.
Supplemental data for this article can be accessed online at https://doi.org/10.1080/07391102.2020.1787228.
ß2020 Informa UK Limited, trading as Taylor & Francis Group
JOURNAL OF BIOMOLECULAR STRUCTURE AND DYNAMICS
https://doi.org/10.1080/07391102.2020.1787228
Therefore, considering SARS-COV-2 main protease enzyme
as a drug target might be an effective approach to develop
novel therapeutic agents against this uncontrollable SARS-
COV-2 infection. Jin et al. (2020) reported the structure of
SARS-COV-2 main protease enzyme complexed with a pep-
tide-like inhibitor N3. The N3 ligand form covalent bond with
one of the catalytic amino residues CYS145 of the protease
enzyme. Residues like HIS41, PHE140, LEU141, ASN142,
HIS163, MET165, HIS172, GLN189, and GLN192 shows non-
covalent interaction with N3 ligand. These residues can be
considered as the crucial amino acid residues of 3CL
pro
receptor binding site (Das et al., 2020).
It is a well-known fact that drug discovery is a time
demanding and costlier process. Therefore, observing the
worst scenario lately it was resolved to screen the US Food
and Drug Administration (FDA) approved small molecule
database using computational methods (S. A. Khan et al.,
2020). Such an approach accelerates the drug repositioning
process against SARS-CoV-2 main protease enzyme. Drug
repurposing or repositioning (R. J. Khan et al., 2020)isa
drug development approach to find out and re-use of
already approved existing drugs for application in different
new diseases. The process is now becoming a universal strat-
egy mainly for a neglected/RARE disease (Pushpakom et al.,
2019). This strategy offers many advantages including, fewer
clinical trial steps which will minimize the cost and time
(Paul et al., 2010). The advantages of re-purposing the drug
discovery approach can also be used to explore potential
lead molecules to combat the COVID-19 pandemic. In the
current study, structure-based virtual screening of FDA-
approved drug database was carried out against the SARS-
COV-2 main protease. Best molecules obtained based on
binding affinity were further considered for molecular
dynamics (MD) simulation. Finally, the binding free energy
was estimated from the MD simulation trajectories through
molecular mechanics Poisson-Boltzmann surface area (MM-
PBSA) (Kumari et al. 2014). The credential of the work, sub-
stantiated by the finding of two most crucial drug molecules
effective for inhibition of SARS-COV-2 main protease.
Materials and method
Protein preparation
The novel coronavirus main protease enzyme structure com-
plex with a peptide-like ligand (N3) having PDB ID: 6LU7
(resolution: 2.16 Å and r-value:0.235) and consisting of 312
amino acid residues, was retrieved from Protein Data Bank
(www.rcsb.org). The protein structure was prepared using
the Autodock Tools (Morris et al., 2009), a part of MGL Tools
(Forli et al., 2016) molecular visualization interface. The miss-
ing atoms were checked and repaired. A sufficient number
of polar hydrogen atoms and Gasteiger charges were added.
The co-crystalline water molecules and bound ligand N3
were removed. The prepared protein molecule was saved for
molecular docking study. Also, molecular visualization soft-
ware included UCSF chimera (www.cgl.ucsf.edu/chimera),
and VMD (Humphrey et al., 1996) were used for rendering
images. The binding site was determined by confining N3
with the coordinates of 9.231, 11.910 and 69.213 along
X-, Y-, and Z-axis, respectively.
FDA-approved database screening
Structure-based virtual screening of small molecular data-
bases is an important and effective tool to find out promis-
ing molecules for a specific target. A large number of
macromolecular crystal structure availability offers an excel-
lent opportunity to explore potent and safer molecules. To
find out potential anti-COVID compounds the FDA-approved
drug data set was screened through the LEA3D (Douguet
et al., 2005) online software tool. The FDA-approved drug
molecules are already tested and explored small molecular
dataset for their function and effects on humans. The dataset
is consisting of a total of 1930 drug molecules. All molecules
in the database are highly enriched with characteristics being
effective candidates as administered drug and optimize the
search for new therapeutic agents (Douguet, 2018). The
LEA3D is an online tool, used for de novo design and screen-
ing of in-house database or FDA-approved drug molecules.
The prepared protease protein molecule was imported in the
online LEA3D server. The binding site radius and the weight
in the final score were set to 10 Å and 1, respectively. In the
virtual screening option, the FDA-approved drug data set
was selected. After successful completion, a set of hit mole-
cules was retrieved. The server uses a genetic algorithm
based protein-ligand ANT system known as PLANTS docking
program associated with empirical scoring functions (Korb
et al., 2009) for molecular docking, reliable pose prediction
and search speed. The LEA3D program generates conformers
with RDKit (www.) and the maximum number of conformers
was set to 10. The scoring function of PLANTS can be calcu-
lated by employing the following functions (Islam & Pillay,
2020).
fPLANTS ¼fplp þfclash þftors þCsite (1)
where f
plp
is the piecewise linear potential to model steric
complementarity between protein and ligand atoms, f
clash
represents the heavy atom potential to prevent internal lig-
and clashes, the f
tors
is the torsional potential of ligand, C
site
denotes the distance-dependent quadratic potential in order
to calculate the reference point of the ligand.
The structural information (Isomeric SMILES) of co-crystal-
line ligand N3 (ID: PRD_002214) was retrieved from Protein
Data Bank. The structure was prepared by using LEA3D web
server tool (https://chemoinfo.ipmc.cnrs.fr/LEA3D/drawonline.
html). PLANTS docking exercise was conducted to dock the
prepared ligand N3 with SARS-CoV-2 main protease enzyme
(6lu7) in the same binding site. The Ligplotþ(Laskowski &
Swindells, 2011) and Discovery studio visualizer (www.
3dsbiovia.com) were used to explore the ligand-protein
interactions.
Molecular dynamics simulation
To explore the dynamic states of any protein-ligand complex
the all-atoms MD simulation is an important application.
2 S. CHATTERJEE ET AL.
Final proposed molecules complex with the protease were
considered for the all-atom MD simulation study. The study
was conducted in GPU (NVIDIA RTX 2070) accelerated
Gromacs 2019.4 software (Abraham et al., 2015; R. J. Khan
et al., 2020)(www.gromacs.org), running over Linuxmint
64-bit operating system supported by 9th generation Intel i7
9700k processor. Total 100 ns dynamics simulation was con-
ducted with a time step of 2 fs considering constant pressure
of 1 atm and constant temperature of 300K. The SwissParam
tool (Zoete et al., 2011)(www.swissparam.ch), online server
software, was used to generate ligand topology and param-
eter. For the protein molecule, the CHARMM36 (Huang &
Mackerell, 2013) all-atom force field was used. The TIP3P
water model was used for the solvation of the system fol-
lowed. In order to define the system size for simulation, 10 Å
distance from the surface of the center of the protein was
maintained. The system was neutralized by adding an appro-
priate amount of Na
þ
ions or Cl
ions. The steepest descent
algorithm of 25,000 steps was used to equilibrate and min-
imize the system. The cut off was set to 0.9 and 1.4 nm for
long-range interaction of van der Waals and electrostatic,
respectively. Parameters like root-mean-square deviation
(RMSD), root-mean-square fluctuation (RMSF), and radius of
gyration (Rg) were calculated from simulation trajectory.
Grace software (http://plasma-gate.weizmann.ac.il/Grace/)
was used as a plotting program.
Binding free energy calculation
The binding free energy (DG
bind
) (Wahedi et al., 2020) was
calculated by MM-PBSA approach through the g_mmpbsa
script program (Kumari et al., 2014) for the selected mole-
cules in order to investigate relative binding affinity toward
the protease protein. The MD simulation trajectory of last
10 ns was considered for the DG
bind
calculation. The detailed
procedure of DG
bind
is described below.
Equation (1) was used to calculate the DG
bind
.
DGbind ¼GComplexGprotein Gligand (1)
Here, the G
complex
represents the total free energy of pro-
tein and ligand complex. Individual binding energy of pro-
tein and ligand in the solvent can be defined as G
protein
and
G
ligand
, respectively. Separate binding energy of each compo-
nent such as complex (G), protein (G) and ligand (G) is calcu-
lated as per Equation (2).
G¼hEMMiTS þhGsolvationi(2)
The E
MM
describes the average molecular mechanics (MM)
potential energy in a vacuum. The temperature and entropy
are represented by the Tand S, respectively. The free energy
of solvation is denoted by G
solvation
.
Further, the E
MM
is calculated using Equation (3).
EMM ¼Ebonded þEnonbonded (3)
The term, E
bonded
represents the bonded interaction such as
bond length, bond angle and dihedral angle. On the other
hand, the E
nonbonded
explains the nonbonding interactions
including the electrostatic and van der Waals interactions.
The term G
solvation
in Equation (2) is the energy required
to transfer a solute from a vacuum to the solvent. This term
can be calculated by Equation (4)
Gsolvation ¼Gpolar þGnonpolar (4)
The electrostatic and nonelectrostatic contribution to the
solvation free energy is represented by the G
polar
and
G
nonpolar
, respectively.
Results
Analysis of novel coronavirus protease enzyme
structure complex
For a successful structure-based drug design, verifying the
quality of protein structure is a crucial step. Therefore after
collecting crystal structure of novel coronavirus protease
enzyme (PDB ID: 6LU7) from the RCSB Protein Data Bank, the
quality was checked with PROCHECK server (Laskowski et al.,
1993). The obtained Ramachandran plot (Figure S1 in supple-
mentary material) was analysed and it was found that 90.6%
residues are in the favourable region whereas 8.7% residues
lie in the additional allowed region. Only 0.4% of residues lie
in generously allowed regions and disallowed regions. The
above results unambiguously support the quality and reliabil-
ity of the protein molecule and considered for further study.
The crystal structure of protease enzyme which is complex
with covalently bound with a peptide-like ligand (N3), con-
sists of alanine, valine and leucine amino acid residue along
with non-amino acid organic moiety. The ligand N3 form
hydrogen bond with PHE140, GLY143, CYS145, HIS163,
HIS164, GLU166, GLN189 and THR190 amino acid residues
present in the binding pocket of this protease enzyme.
Among these amino acid residues, GLU166 form two hydro-
gen bonds with N3. The amino acid CYS145 forms a covalent
interaction with electrophilic carbon of the ligand (Figure 1).
The ligand-binding site was used to explore the FDA-
approved drug molecules and identifying high-affinity com-
pounds for this binding site using a structure-based drug
design approach. In the purpose of molecular docking, the
PLANTS, an open-source tool was used (Korb et al., 2009).
Validating docking protocol is a crucial step before per-
forming molecular docking based virtual screening and to
determine threshold parameters (Taha et al., 2011). PLANTS
program based docking of ligand N3 yielded 10 docking
poses. All the poses were found to be present inside the
binding pocket. RMSD value less than 2 Å after superimpos-
ing co-crystalline ligand upon the pose of dock ligand infer
the validity of docking protocol (Huang & Zou, 2006). In
order to validate the docking protocol, best docking pose of
ligand N3 were superimposed upon co-crystal structure of
ligand N3 (superimposed co-crystal structure given in supple-
mentary material) and the RMSD was found to be 0.746 Å
along with PLANTS score 115.467. Based upon the RMSD
value of 0.746 Å and PLANT score, docking pose of ligand
N3 was selected. The interactions between protein and
selected docked pose of ligand N3 were analysed.
JOURNAL OF BIOMOLECULAR STRUCTURE AND DYNAMICS 3
Virtual screening
FDA-approved drugs data set was screened by LEA3D server-
based program. For each molecule, the LEA3D program gen-
erates 10 conformers. Based on high negative PLANT score,
the best conformer was selected. After successful completion
of screening, the molecules were considered for further anal-
yses upon the a) threshold PLANTS score (115.467), b) inter-
action with crucial amino acids (Umesh et al., 2020), and c)
reasonable orientation of the molecules inside the pockets.
In agreement with threshold PLANTS score, crucial amino
acid interaction, reasonable orientation, a total of 20 mole-
cules (Table 1) were considered. The PLANTS score of
selected molecules was found to be in the range of 142
to 128.
Among these 20 compounds, top five compounds were
Triptorelin, Nafarelin, Icatibant, Cobicistat and Histrelin
(Figure 2) exhibiting binding score in terms of PLANTS score
142.900, 130.820, 130.710, 128.740, 128.710,
respectively.
Besides binding score, analysis of amino acid residue
interaction depicts, Triptorelin shows hydrogen bonds
(H-bond) with THR45, SER6, TYR54, ASN142, GLY143, GLU166
and GLN189 amino acid residues along with superior hydro-
phobic interactions. Nafarelin form H-bond with THR26,
ASN42, GLY43, SER46, PRO168, GLN189 amino acid residues.
Icatibant exhibits H-bond interactions with PHE140, ASN142,
GLU166, LEU167, PRO168, and GLN189. Cobicistat bind with
the main protease enzyme through one H-bond with GLY143
and by strong hydrophobic interactions with amino acid resi-
dues (residue id 2527, 41, 46, 49, 140142, 145, 164167,
187189). The fifth ranking compound Histrelin exhibits
H-bond interaction with ASN42, THR45, SER 46, GLY143, CYS
145, and ASP187. Other than Cobicistat all four ligands form
an average of 6 H-bonds with 2019-main protease enzyme.
Further analysis of protein-ligand interactions (supplementary
material Figure S2) shows Triptorelin form p-sulfur interaction
with CYS145 and MET49 residues. A p-p-stacking interaction
was found with HIS41, LEU50 residues. Nafarelin exhibits
p-alkyl interaction with MET49, HIS163, and MET165.
Icatibant form p-sulfur interaction with HIS41, MET165. p-sul-
fur interaction was also found in Cobicistat-main protease
enzyme complex and involved amino acid residues are
MET49, 165, GLU166. T-shaped p-pinteraction was observed
with HIS41 residues; however, LEU27, MET49 and CYS145
form p-alkyl interaction. The fifth ligand Histrelin exhibit
p-sulfur interaction with MET165 and p-pstacking with
HIS41, LEU141, HIS163, HIS172. Pharmacological properties of
shorted molecules indicate Triptorelin, Nafarelin and
Icatibant associated with the event of human hormone func-
tion with acceptable pharmacokinetic properties. Icatibant
acts as a bradykinin B2 receptor antagonist (Riedl et al.,
2018). Third molecule Cobicistat along with other antiviral
drugs as a combination is in phase II clinical trial as an anti
2019nCoVD management (Rosa & Santos, 2020), exhibit its
Figure 1. Interaction of ligand N3 with main protease enzyme.
4 S. CHATTERJEE ET AL.
Table 1. List of FDA-approved drugs exhibiting high negative binding with 2019 novel coronavirus protease enzyme.
S. no Name Pharmacological property Score (%) PLANTS score
1 Triptorelin Antineoplastic, synthetic analog of gonadotropin releasing hormone
palliative treatment of advanced prostate cancer. Stimulates the
release of luteinizing hormone
95.27 142.900
2 Nafarelin Treatment of central precocious puberty (CPP) gonadotropin-releasing
hormone agonist
87.2 130.820
3 Icatibant Hereditary angioedema bradykinin B2 receptor antagonist 87.14 130.710
4 Cobicistat Inhibiting cytochrome P450 3A isoforms. Treatment of HIV-1 infection 85.83 128.740
5 Histrelin Gonadotropin releasing hormone (gnrh) agonist 85.81 128.710
6 Goserelin Plant prostate cancer (therapeutic) antineoplastic. Gonadotropin
releasing hormone (gnrh) agonist
84.45 126.670
7 Angiotensin II A vasoconstrictor to increase blood pressure in adults 83.44 125.160
8 Sincalide Various diagnostic imaging, choleretic 82.99 124.480
9 Ceruletide Various spasmogenic effect, diagnostic agents 82.82 124.230
10 Saralasin Cardiovascular antihypertensive angiotensin ii inhibitor 82.58 123.870
11 Carfilzomib Antineoplastic, treatment of multiple myeloma 81.39 122.080
12 Etelcalcetide Treatment of secondary hyperparathyroidism (hpt). Calcium-sensing
receptor (casr) agonist
81.04 121.560
13 Venetoclax Antineoplastic agent. Bcl-2 (b-cell lymphoma 2) inhibitor 80.40 120.600
14 Colfosceril palmitate Lung surfactants 79.69 119.530
15 Latanoprost Antiglaucoma, antihypertensive agents 79.29 119.530
16 Ritonavir Anti-infectives , antiviral, anti-HIV protease inhibitors 77.93 116.900
17 Abarelix Antineoplastic, anti-testosterone agents gonadotropin-releasing
hormone (gnrh) antagonist
77.60 116.400
18 Latanoprostene Antiglaucoma, antihypertensive agents , prostaglandin analog 77.35 116.020
19 Fluphenazine Antipsychotic 77.33 116.000
20 Amphotericin B Fungistatic or fungicidal 77.06 115.590
21 Ligand N3 Co-crystalline ligand 76.46 115.467
Figure 2. Binding site interactions of each ligand with 2019 novel coronavirus main protease enzyme (a. Triptorelin, b. Nafarelin, c. Icatibant, d. Cobicistat,
e. Histrelin).
JOURNAL OF BIOMOLECULAR STRUCTURE AND DYNAMICS 5
pharmacological action by inhibiting cytochrome P450 3A
isoforms and enhance anti-HIV drugs bioavailability.
Triptorelin (DrugBank id: DB06825) potentiate luteinizing
hormone-releasing activity and follicle-stimulating hormone-
releasing activity 13-folds and 21-folds, respectively. It is gen-
erally administered through oral and intravenous route (M
uller
et al., 1997). Chemical structure of Nafarelin (DrugBank id:
DB00666) have 3-(2-naphthyl)-D-alanine substitution which
acts as a potent synthetic agonist of gonadotropin-releasing
hormone generally used as a nasal drop (Garner, 1994).
Synthetic peptidomimetic drug Icatibant having 10 amino acid
residues exhibits bradykinin B2 receptor agonistic activity
(Bachelard et al., 2018) and is generally used for the treatment
of hereditary angioedema with superior bioavailability profile.
Cobicistat is generally used in combination with other anti-
retroviral agents for effective management of HIV-1 infection.
During clinical trials, adverse effect like jaundice was reported
for this molecule. The fifth molecule Histrelin (DrugBank id:
DB06788) acts as a gonadotropin-releasing hormone agonist
(Klein et al., 2020) generally used as an implant. Based on the
above pharmacological analysis we have considered these top
five molecules for molecular dynamics simulation analysis in
order to examine stability with ligand-SARS-COV-2 main prote-
ase enzyme complex.
Molecular dynamics study
All atom molecular dynamics simulation was conducted for
top five hits complexed with SARS-COV-2 main protease.
Dynamic properties of each complex were compared with
the dynamic behaviour of co-crystalline ligand N3 and ligand
free protein. From 100 ns MD simulation trajectory, the RMSD
was calculated and plotted against time (Figure 3).
The average RMSD of the N3, Triptorelin, Nafarelin,
Icatibant and Cobicistatprotein systems was found to be
2, 9.2, ~2.3, 1.4, 2.07 Å, respectively. The ligand free
protein or apo protein exhibits average RMSD value of
2.15 Å. A sharp increase in RMSD (10 Å) was observed
with Triptorelinmain protease enzyme system after 10 ns of
simulation. However, the Triptorelin system exhibited incon-
siderable RMSD fluctuation (4 Å) throughout the simula-
tion. Histrelin system after 2 ns of simulation shows sudden
increase in RMSD (8 Å) and similar pattern (RMSD >)
like Triptorelinprotein system. Notably large RMSD of the
atomic positionsbehaviour were observed for the Triptorelin
and Histrelinprotein system, respectively. Therefore, the
simulation was not continued further for the Histrelin system
(average RMSD 8.45 Å up to 30 ns). The Icatibantprotein
system exhibits the lowest average RMSD 1.4 Å and the
RMSD was found consistent throughout the 100 ns simula-
tion time. Nafarelin (average RMSD 2.3 Å) at 31st ns to
38th ns exhibits rise in RMSD (0.017 Å higher than average
RMSD). At 50th ns to 52th ns time span and 56th58th ns
time span of simulation this system exhibited 0.013 to
0.01 Å higher RMSD than average RMSD value. After 87th
ns to 100 ns the average RMSD value was found to be 1.5 Å.
The RMSD value of Cobicistatprotein system after 95 ns was
found to be 1.45 Å.
Figure 3. Protein backbone RMSD of each ligand-protein complex and ligand free protein.
6 S. CHATTERJEE ET AL.
In order to understand the deviation of each protein,
amino acid residues with respect to the reference position,
the RMSF plot was constructed from 100 ns atomistic
molecular dynamics trajectory which is shown in Figure 4.
The average value of RMSF for N3, Triptorelin, Nafarelin,
Icatibant and Cobicistatprotein complexes were found to be
1.3, 1.5, 1.3, 0.92, 1.4 Å, respectively. The average
RMSF value for ligand free protein was found to be 1.42 Å.
In Nafarelinprotein system, residues in between residue
ID: 210 and residue ID: 290 show slightly high RMSF (1.48 Å).
Residues number 222, 223, and 224 shows average RMSF 2.6
Å in Cobicistatprotein system. Comparatively higher fluctu-
ation can be observed with terminal residues (6.5 Å) of each
system including apo protein. HIS41 and CYS145 residues in
apo-protein exhibits RMSF 0.081 and 0.073 Å, respectively. The
residue HIS41 shows RMSF of 0.12, 0.068, 0.065 Å for N3,
Nafarelin, Icatibantprotein systems, respectively whereas in
Cobicistatprotein system HIS41 exhibits RMSF 0.095 Å. The
CYS145 residue exhibits the RMSF of 0.11, 0.053, 0.048, 0.081 Å
for N3, Nafarelin, Icatibant and Cobicistatprotein systems,
respectively. Icatibant system shows lowest average RMSF
0.92 Å. The radius of gyration (Rg) was calculated from
100 ns trajectory to analyse the compactness and rigidity of
the protein-ligand system (Figure 5).
Cobicistatprotein system after 26.4 ns exhibit little devi-
ation (23.24 to 22.75 Å) and for the rest of the simulation
time it shows average Rg value of 22.36 Å. Ligand free
SARS-COV-2 main protease enzyme exhibit average Rg of
22.6 Å. The average Rg value of N3, Triptorelin, Nafarelin,
Icatibant and Cobicistatprotein systems were found to be
22.5, 22.7, 22.4, 22.42, 23.1 Å, respectively. For a stable com-
plex, it is essential, that the ligand and protein should
maintain acceptable distance throughout the simulation. The
distance between protein and ligand during the simulation
was calculated and it is shown in Figure 6.
The average distance between protein and ligand during the
simulation was calculated from 100ns molecular dynamics tra-
jectory. Average distance 1.7, 1.8, 1.6, 1.6, 2.0 Å was
found for N3, Triptorelin, Nafarelin, Icatibant and Cobicistat
complexed with main protease enzyme, respectively. In order to
study the surface area that is accessible to solvent for each sys-
tem, a solvent accessible surface area (SASA) plot was con-
structed from the trajectory both for protein. (Figure 7).
The SASA information will be helpful to analyse, whether
the ligand retained inside the shallow binding pocket or it
expels out from the binding cavity. The ligand free protein
exhibits average SASA value of 1685.2 Å
2
. The average SASA
values 1540, 1515, 1545, 1555, 1545
2
) were found
for N3, Triptorelin, Nafarelin, Icatibant and Cobicistat protein
complex, respectively. The average SASA values for ligand N3,
Triptorelin, Nafarelin, Icatibant and Cobicistat was found to be
110, 1900, 161, 155, 110
2
), respectively. Analysis of the main
protease-ligand complexes revealed most of the compounds
form H-bond with the binding pocket amino acid residues. In
order to analyse the H-bond interaction property during the
100 ns period of simulation, H-bond interaction plot was con-
structed and it is depicted in Figure 8.
Average 5.5, 2.5, 9.5, 7 and 1.25 numbers of
H-bond was found for ligand N3, Triptorelin, Nafarelin,
Icatibant and Cobicistat complexed with main protease
enzyme, respectively. Percentage frequency of hydrogen
bonds (%HB) calculated from the 100 ns and last 25 ns of
molecular dynamics trajectory was calculated and it is shown
in Figure 9.
Figure 4. RMSF of protein-ligand complexes.
JOURNAL OF BIOMOLECULAR STRUCTURE AND DYNAMICS 7
From the plot it clearly observed that Nafarelin shows
maximum frequency of hydrogen bonding interaction with
main protease enzyme. Icatibant exhibits comparatively less
frequency of hydrogen bond formation during last 25 ns
of simulation.
High negative binding energy often infers the superiority
of protein-ligand complex (Gallicchio et al., 2010). The DG
bind
was calculated for each of five final molecules including N3
towards the main protease enzyme through MM-GBSA
approach. It was observed that the DG
bind
of N3 found to be
599.15 kJ/mol. Moreover, the DG
bind
of Nafarelin, Icatibant
and Cobicistat was found to be 712.94, 851.75, 252.65 kJ/
mol, respectively. A number of parameters calculated from
the MD simulation are given in Table 2.
Discussion
Protease enzymes play a crucial role in peptide bond
hydrolysis. The functionally crucial cysteine and histidine
Figure 5. Radius of gyration (Rg) vs. time.
Figure 6. Distance calculated from protein-ligand complex.
8 S. CHATTERJEE ET AL.
residues (HIS41-CYS145 dyad) (Ul Qamar et al., 2020) are pre-
sent in the catalytic site of this enzyme. PLANTS docking was
performed for co-crystalline ligand N3. The RMSD value of
0.746 Å between co-crystal N3 and best dock pose of N3
clearly infer the validity of the docking protocol used for this
study. In agreement with this, it can be contemplated that
using the same protocol, docking of the other molecules
with this protein and considering the same binding pocket,
can furnish positive docked conformations. Hence, the same
docking protocol was applied for molecular docking based
virtual screening.
The co-crystalline ligand N3 forms hydrogen bond inter-
action with SARS-CoV-2main protease enzyme. Beside
hydrogen bonds, it exhibits hydrophobic interactions. The
analysis revealed that the co-crystalline ligand forms a cova-
lent interaction as well as H-bond interaction with CYS145
amino acid residue and hydrophobic interaction with
HIS41 residue.
Figure 7. Solvent accessible surface area vs. time (ns) plot.
Figure 8. H-bond interactions exhibited by each ligand when complexed with this main protease enzyme.
JOURNAL OF BIOMOLECULAR STRUCTURE AND DYNAMICS 9
Results obtained by computational screening (Table S1,
given in supplementary material), Ritonavir a classical HIV-1
protease inhibitor (EC
50
25 nM), currently under anti-COVID
19 clinical trial (Cao et al., 2020) having PLANTS score and %
score116.40, 77.93 respectively, formed H-bond with
GLN189 (Figure S2 in supplementary material). However, it
shows a strong hydrophobic interaction. The 4th ranked
drug Cobicistat, a cytochrome P450-3A isoform inhibitory
agent which facilitates the anti-retroviral properties of
Atazanavir or Darunavir (Crutchley et al., 2016) exhibiting sat-
isfactory PLANTS score and % score 128.740 and 85.83,
respectively and form H-bond with GLY143 amino acid resi-
due of protein molecules. Rest of the three molecules among
top five molecules namely Triptorelin (rank 1), Nafarelin (rank
2) are either synthetic analogues of gonadotropin-releasing
hormone or gonadotropin-releasing hormone agonists exhib-
iting superior binding with nCoV-main protease enzyme.
Triptorelin form two H-bonds with SER46 exhibiting distance
2.65 and 3.07 Å, respectively. Both N3 ligand and Triptorelin
show identical H-bond interaction with GLY43, GLU166,
GLN189 and identical hydrophobic interaction with THR25,
LEU141, MET165, ARG188 and PRO168. However, Triptorelin
depicts H-bond interaction with ASN142 but these residues
interact with ligand N3 through hydrophobic interaction.
Besides, residue like PHE140, HIS163, and HIS164 show H-
bond with ligand N3 but with Triptore in it forms hydropho-
bic interaction. The above findings provide suitable informa-
tion about Triptorelin that it exhibits binding energy of 142
with the main protease enzyme. However, Nafarelin forms
three H-bonds with ASN142 depicting the length of 2.81,
2.87 and 3.22 Å. A 2.89 Å H-bond was found in between
Nafarelin and GLN189 amino acid residue. GLN189 amino
acid residue also forms 2.89 Å H-bond with co-crystalline lig-
and N3. HIS41 and CYS145 can be considered as crucial resi-
dues for this enzyme function. Ligand N3 shows
hydrophobic interaction with HIS41, similarly Nafarelin
exhibit identical interaction with this residue. Like N3-
CYS145 H-bond interaction, Nafarelin does not show inter-
action with CYS145. However, identical hydrophobic inter-
action with THR 25, LEU141, MET165, ARG188, GLN192
amino acid residues was found. Few residues like PHE140,
HIS163, and THR190 form a hydrogen bond with co-crystal-
line ligand N3, but interact with Nafarelin through hydropho-
bic interaction. Similarly THR26, ASN142, PRO168 form H-
Figure 9. Percentage frequency of hydrogen bonds calculated from molecular dynamics trajectory for each protein-ligand system.
Table 2. Results of molecular dynamics simulation.
Name
Average
RMSD
protein-
ligand (Å)
Average
RMSF (Å)
Average
number of
H-bond
protein-
ligand
Frequency
of hydrogen
bond
(0100 ns)
%
Frequency
of hydrogen
bond
(75100 ns)
%
Average
distance
protein-
ligand (Å)
Average Rg-
protein (Å)
Average
SASA
protein
2
)
Average
SASA
ligand
2
)
Binding
energy
(kJ/mol)
6LU7-LF 2.15 1.42 ND ND 22.6 1685.2 ––
N3 2 1.3 5.5 237 228 1.7 22.5 1540 110 599.15
Triptorelin 9.2 1.5 2.5 139 68 1.8 22.7 1515 190 ND
Nafarelin 2.3 1.3 9.5 284 253 1.6 22.4 1545 161 712.94
Icatibant 1.4 0.92 7 205 100 1.6 22.4 1555 155 851.74
Cobicistat 2.07 1.4 1.25 11.5 19 2.0 23.1 1545 110 252.65
Histrelin
a
8.45 ND ND ND ND ND ND ND ND ND
a
Simulation continues up to 30 ns.
LF ¼ligand free, ND ¼not done, Rg ¼radius of gyration.
10 S. CHATTERJEE ET AL.
bond with Nafarelin; validated by 100 ns molecular dynamics
trajectory and those residues showing hydrophobic inter-
action with co-crystalline ligand N3. ASN142 of the main pro-
tease enzyme forms three H-bonds (length 2.81, 2.87, 3.22 Å
respectively) with this ligand. In addition, Nafarelin exhibits a
mean distance of 0.5 Å from CYS145 residue (Figure S3 in
supplementary material). The above facts infer Nafarelin
exhibit superior binding interaction with the catalytic domain
of 2019 main protease enzyme and exhibiting one identical
H-bond interaction and number of identical hydrophobic
interactions followed by either H-bond or hydrophobic inter-
action with binding site residues. Icatibant shows identical H-
bond interaction with PHE140, GLU 166, GLN 189 and hydro-
phobic intersection THR 24-26, LEU141, ARG188. Residues
like ASN142, PRO168 show H-bond interaction with Icatibant
but showing interacting with N3 ligand through hydrophobic
interaction. Similarly, residues like HIS164 form H-bond with
co-crystalline ligand but exhibiting hydrophobic interaction
with Icatibant. Though there is no H-bond and hydrophobic
interaction with CYS145 and HIS41 was observed, Icatibant
shows p-sulfur interaction with HIS 41, based on this fact this
molecule was considered for further study. Cobicistat exhibits
one identical H-bond interaction and identical hydrophobic
interaction with THR25-26, HIS41, ASN142, MET165, ARG188,
residues of main protease enzyme. On the other hand,
Histrelin forms H-bond with CYS145 like co-crystalline ligand
as well as exhibits identical hydrophobic interactions with
THR 24-26, HIS 41 residues of main protease enzyme.
Residues like PHE140; HIS143 shows hydrophobic interaction
with Histrelin but forms H-bond with a co-crystalline ligand.
Considering its binding interaction with CYS145 and HIS41,
this molecule was considered for molecular dynamics study.
Atomistic 100 ns MD simulation of ligand free protein and
each protein-ligand complexes were conducted. Various
parameters like RMSD, RMSF, Rg, distance of protein and lig-
and, SASA, number of hydrogen bonds, etc. were calculated
from the trajectory of MD simulation.
The RMSD parameter calculated through molecular
dynamics simulation provides brief insight into the structural
conformations of proteins. Overall information about the sta-
bility of protein backbone when bound with a ligand or
small molecule can be analysed by RMSD parameter. Lower
value of RMSD throughout the molecular dynamics simula-
tion suggests the high stability of protein ligand system and
higher RMSD value infer comparatively low stability of the
system. Changes in RMSD towards larger ranges might indi-
cate that the protein-ligand system during dynamics simula-
tion, probably undergoes remarkable or some shorts of
conformational changes. However, RMSD values that
undergo lower ranges are ideally acceptable for protein sys-
tems (Kufareva & Abagyan, 2012).
The average RMSD parameter is less than 3 Å which is
perfectly acceptable for globular protein (Kufareva &
Abagyan, 2012). RMSD plot of protein-ligand complex
depicts, Triptorelin bound protein backbone exhibits incon-
siderable divination of atomistic root mean square position
throughout the simulation time, which suggests probably
due to the presence of high degree of rotatable bond or
structural flexibility this molecule is unable to attain stability
inside the binding pocket, which is assumed to be shallow.
Cobicistat-protein system up to 86 ns exhibit average RMSD
of 2.2 Å and 86th ns to 95 ns it depicts RMSD of 1.35 Å.
After 95 ns, average RMSD for this system was found to be
1.45 Å. Total increase in RMSD from 86th to 95th time
span to 95th to 100th ns was 0.1 Å. For Nafarelinprotein
system exhibit 0.15 Å higher average RMSD than apo protein
RMSD value; however, after 87th ns the average RMSD value
is less than (0.65 Å) apo protein average RMSD value. In
agreement with the above observation, it can be concluded
this variation in RMSD found in Nafarelin and
Cobicistatprotein system does not cause significant struc-
tural changes and achieves equilibration state during
the simulation.
The RMSF parameter explores the individual fraction of
the protein structure that is fluctuating from its mean struc-
ture. Higher degrees of RMSF values infer the protein struc-
ture will attain greater flexibility. On the other hand,
marginal flexibility of the protein-ligand system results in
lower RMSF values (Bhowmick et al., 2020). Each ligand-pro-
tein system other than Triptorelinprotein system exhibits
average RMSF value less than 1.42 Å, which is apo protein
RMSF value. Throughout the 100 ns simulation, the low RMSF
value was found for both HIS41 and CYS145 residues of
Nafarelin and Icatibant systems which indicate these two
ligands exhibit interaction with HIS41 and CYS145 residues.
The terminal residues exhibits highest fluctuation and can be
clearly observed in Figure 8. These residues are located in
the loop region of this protein. The RMSF of the terminal res-
idues of this protein reported by Muralidharan et al., 2020
exhibit almost identical RMSF profile.
The Rg parameter is defined as the mass-weighted root
mean square distance of a set of atoms from their typical
center of mass (Lobanov et al., 2008). Rg parameters provide
information about overall dimensions and quantify changes
of protein structure. Large variation in Rg value indicates
that protein-ligand system was inconsistent throughout the
simulation. Uniform variation of Rg value infers the consist-
ent stability of the protein during simulation (Bhowmick
et al., 2020). From the Rg value (Å) of each frame versus
time (ns) plot (Figure 9), it can be observed that the ligand
free protein was rigid and consistent with an average Rg
value of 22.6 Å. Nafarelin and Icatibant systems oscillated at
22.4 Å, which is 0.1 Å less than the Rg value of co-crystal
bound ligand N3. Moreover, these systems exhibit 0.2 Å less
Rg than the apo protein Rg value and exhibit a similar Rg
profile. The Rg of Triptorelin deviates little bit higher (0.1 Å)
than the ligand free protein. Cobicistat system exhibits 0.5 Å
higher Rg value than apo protein and 0.6 Å higher than co-
crystalline ligand N3. In argument with above observation, it
can be stated Nafarelin and Icatibant system exhibits accept-
able Rg profile. Binding of these two hits with SARS-COV-2
remained stable.
Nafarelin and Icatibant system exhibits alike SASA profile
with ligand N3 and apo-protein. This indicates, these two
ligands exhibiting superior contacts with this enzyme. After
analysing the trajectory it was our assumption and
JOURNAL OF BIOMOLECULAR STRUCTURE AND DYNAMICS 11
interpretation, declination pattern exhibited by the protein
when complexed with ligands probably due to expelling of
water from binding pockets which certainly occupied by the
ligands. This fact can be supported by SASA plot calculated
for the ligands (supplementary material Figure S4) which
depicted all the ligands follows uniformity throughout the
simulation time.
Surface area up to 110 Å
2
and 110 Å
2
depicted by
Cobicistat and ligand N3 suggests that these molecules are
less exposed to solvent. Nafarelin and Icatibant higher (51
Å
2
and 45 Å
2
, respectively) solvent-exposed surface area
than co-crystalline ligand N3 which decreased after 65 ns of
simulation. On the other hand, Triptorelin after 64 ns shows
a slight increase in SASA value. SASA analysis inferred that
Cobicistat, Nafarelin and Icatibant have the properties of
forming a stable complex with this main protease enzyme.
Mean distance of protein and ligand was calculated, which
depicts clearly Nafarelin and Icatibant show 1.7 Å distance
from the whole protein throughout the simulation. H-bond
plot calculated from dynamics trajectory depicts Cobicistat
form comparatively fewer H-bond, whereas Nafarelin and
Icatibant form a higher degree of H-bond interaction with
SARS-COV-2 main protease enzyme in comparison with co-
crystalline ligand and Triptorelin. The percentage frequency
of hydrogen bonds were calculated from 100 ns and last
25 ns (75100 ns) trajectory indicates Nafarelin exhibits 284%
(100 ns) and 253% (last 25 ns) frequency in hydrogen bond
formation with main protease enzyme. Comparing with co-
crystalline ligand hydrogen bond formation pattern, Nafarelin
forms 47% and 25% more hydrogen bonds with main prote-
ase enzyme. Icatibant exhibits 32% and 196% less ability to
from hydrogen bond with SARS-CoV-2-main protease
enzyme when compared with co-crystalline ligands fre-
quency of hydrogen bonding profile. Binding energy calcu-
lated from last 10 ns of molecular dynamics trajectory
exhibits Nafarelin (712.94 kJ/mol) and Icatibant (851.74 kJ/
mol) exhibits superior binding energy in comparison with co-
crystalline ligand (599.15 kJ/mol) and Cobicistat (252.65 kJ/
mol), further supports there superior binding properties with
main protease enzyme.
The above results predicted from protein-ligand complex
generated by PLANTS docking server-based virtual screening
and molecular dynamics analysis highly infer that Nafarelin
and Icatibant can be considered as a reference point to re-pur-
posing existing drugs for efficient management of COVID-19.
Considering molecular dynamics is a statistical dynamics pro-
cess, we performed another 100 ns molecular dynamics simu-
lation for Nafarelin-protease enzyme complex (Figure 10)
using same force field only, supports the reproducibility of the
results, which strongly infer, the highest degree of protein-lig-
and complex stability can be observed with Nafarelin-main
protease enzyme complex.
Moreover, from pharmaceutical point of view, Nafarelin is
used as a nasal drop and attain bioavailability very quickly
with minimal unwanted effects. The RMSD, RMSF, Rg, SASA
and binding energy values unveils that Icatibant shows
acceptable stability with SARS-COV-2 main protease enzyme.
These hits can be a better re-purposing option; however,
there should be proper biological evaluations to be con-
ducted to validate its efficacy of the predicted molecules
against this global threat in terms of 2019-novel corona-
virus infection.
Figure 10. Repetition of molecular dynamics simulation for Nafarelinprotein (main protease enzyme) complex.
12 S. CHATTERJEE ET AL.
Conclusion
New anti-COVID-19 drug poses a worldwide challenge, drug
repositioning may be a quicker and safer approach to rely on
and find new therapy against this dangerous life-threatening
infection. Well-defined structure of 2019 novel coronavirus
main protease enzyme is an excellent reference point to iden-
tify new therapeutics, from already examined and approved
drugs for human use. Results obtained through computational
approach can be considered, as computational screening of
small molecules library and molecular dynamics simulation
techniques are the well-established and validated tools for the
drug discovery process. We performed 100 ns atomistic MD
simulations for top five protein-hit complexes that is
Triptorelin, Nafarelin, Icatibant, Cobicistat, Histrelin-main pro-
tease enzyme (PDB ID: 6LU7) of SARS-CoV-2, with an aim to
establish the binding potential of this FDA approved mole-
cules. Analysis of the results portrays higher stability of the
Nafarelin and Icatibant-enzyme systems and their established
pharmaceutical profiles unveils these hits may show a possible
promising application in anti-COVID-19 therapy. Nafarelin and
Icatibant exhibiting promising binding with SARS-CoV-2 main
protease enzyme can open a new window towards the discov-
ery of effective therapy against COVID-19.
Disclosure statement
Authors declare that there are no conflicts of interest.
ORCID
Md Ataul Islam http://orcid.org/0000-0001-6286-6262
Debanjan Sen http://orcid.org/0000-0001-7600-6781
Data availability statement
Data and materials are available upon request to the correspond-
ing author.
References
Abraham, M. J., Murtola, T., Schulz, R., P
all, S., Smith, J. C., Hess, B., &
Lindah, E. (2015). Gromacs: High performance molecular simulations
through multi-level parallelism from laptops to supercomputers.
SoftwareX,1-2,1925. https://doi.org/10.1016/j.softx.2015.06.001
Bachelard, H., Charest-Morin, X., & Marceau, F. (2018). D-arg0-bradykinin-
Arg-Arg, a latent vasoactive bradykinin B2 receptor agonist metabolic-
ally activated by carboxypeptidases. Frontiers in Pharmacology,9(Mar),
273. https://doi.org/10.3389/fphar.2018.00273
Bhowmick, S., AlFaris, N. A., ALTamimi, J. Z., ALOthman, Z. A., Aldayel,
T. S., Wabaidur, S. M., & Islam, M. A. (2020). Screening and analysis of
bioactive food compounds for modulating the CDK2 protein for cell
cycle arrest: Multi-cheminformatics approaches for anticancer thera-
peutics. Journal of Molecular Structure,1216, 128316. https://doi.org/
10.1016/j.molstruc.2020.128316
Boopathi, S., Poma, A. B., & Kolandaivel, P. (2020). Novel 2019 corona-
virus structure, mechanism of action, antiviral drug promises and rule
out against its treatment. Journal of Biomolecular Structure and
Dynamics,110. https://doi.org/10.1080/07391102.2020.1758788
Cao, B., Wang, Y., Wen, D., Liu, W., Wang, J., Fan, G., Ruan, L., Song, B., Cai,
Y., Wei, M., Li, X., Xia, J., Chen, N., Xiang, J., Yu, T., Bai, T., Xie, X., Zhang,
L., Li, C., Wang, C. (2020). A trial of lopinavir-ritonavir in adults
hospitalized with severe covid-19. The New England Journal of
Medicine, 382(19), 17871799. https://doi.org/10.1056/NEJMoa2001282
Chen L., Liu, H. G., Liu, W., Liu, J., Liu, K., Shang, J., Deng, Y., & Wei, S.
(2020). Analysis of clinical features of 29 patients with 2019 novel cor-
onavirus pneumonia. Zhonghua Jie He Hu Xi Za Zhi, 43. https://doi.
org/10.3760/CMA.J.ISSN.1001-0939.2020.0005
Chen, Y., Liu, Q., & Guo, D. (2020). Emerging coronaviruses: Genome
structure, replication, and pathogenesis. Journal of Medical Virology,
92(4), 418423. https://doi.org/10.1002/jmv.25681
Crutchley, R. D., Guduru, R. C., & Cheng, A. M. (2016). Evaluating the role
of atazanavir/cobicistat and darunavir/cobicistat fixed-dose combina-
tions for the treatment of HIV-1 infection. HIV/AIDS (Auckland, N.Z.),8,
4765. https://doi.org/10.2147/HIV.S99063
Das, S., Sarmah, S., Lyndem, S., & Singha Roy, A. (2020). An investigation
into the identification of potential inhibitors of SARS-CoV-2 main pro-
tease using molecular docking study. Journal of Biomolecular Structure
and Dynamics,111. https://doi.org/10.1080/07391102.2020.1763201
Deeks, S. G., Smith, M., Holodniy, M., & Kahn, J. O. (1997). HIV-1 protease
inhibitors. JAMA,277(2), 145153. https://doi.org/10.1001/jama.1997.
03540260059037
Douguet, D. (2018). Data sets representative of the structures and experi-
mental properties of FDA-approved drugs. ACS Medicinal Chemistry
Letters,9(3), 204209. https://doi.org/10.1021/acsmedchemlett.
7b00462
Douguet, D., Munier-Lehmann, H., Labesse, G., & Pochet, S. (2005).
LEA3D: A computer-aided ligand design for structure-based drug
design. Journal of Medicinal Chemistry,48(7), 24572468. https://doi.
org/10.1021/jm0492296
Forli, S., Huey, R., Pique, M. E., Sanner, M. F., Goodsell, D. S., & Olson,
A. J. (2016). Computational protein-ligand docking and virtual drug
screening with the AutoDock suite. Nature Protocols,11(5), 905919.
https://doi.org/10.1038/nprot.2016.051
Gallicchio, E., Lapelosa, M., & Levy, R. M. (2010). The binding energy dis-
tribution analysis method (BEDAM) for the estimation of protein-lig-
and binding affinities. Journal of Chemical Theory and Computation,
6(9), 29612977. https://doi.org/10.1021/ct1002913
Garner, C. (1994). Uses of GnRH agonists. Journal of Obstetric,
Gynecologic, and Neonatal Nursing,23(7), 563570. https://doi.org/10.
1111/j.1552-6909.1994.tb01922.x
Hasan, A., Paray, B. A., Hussain, A., Qadir, F. A., Attar, F., Aziz, F. M.,
Sharifi, M., Derakhshankhah, H., Rasti, B., Mehrabi, M., Shahpasand, K.,
Saboury, A. A., & Falahati, M. (2020). A review on the cleavage pri-
ming of the spike protein on coronavirus by angiotensin-converting
enzyme-2 and furin. Journal of Biomolecular Structure and Dynamics,
19. https://doi.org/10.1080/07391102.2020.1754293
Huang, J., & Mackerell, A. D. (2013). CHARMM36 all-atom additive protein
force field: Validation based on comparison to NMR data. Journal of
Computational Chemistry,34(25), 21352145. https://doi.org/10.1002/
jcc.23354
Huang, S.-Y., & Zou, X. (2006). Efficient molecular docking of NMR struc-
tures: Application to HIV-1 protease. Protein Science,16(1), 4351.
https://doi.org/10.1110/ps.062501507
Humphrey, W., Dalke, A., & Schulten, K. (1996). VMD: Visual molecular
dynamics. Journal of Molecular Graphics,14(1), 3338. https://doi.org/10.
1016/0263-7855(96)00018-5 https://doi.org/10.1016/0263-7855(96)00018-
5 https://www.worldometers.info/coronavirus/dated 25.04.2020.
Islam, R., Parves, M. R., Paul, A. S., Uddin, N., Rahman, M. S., Mamun,
A. A., Hossain, M. N., Ali, M. A., & Halim, M. A. (2020). A molecular
modeling approach to identify effective antiviral phytochemicals
against the main protease of SARS-CoV-2. Journal of Biomolecular
Structure & Dynamics,112. https://doi.org/10.1080/07391102.2020.
1761883
Islam, M. A., & Pillay, T. S. (2020). Identification of promising anti-DNA
gyrase antibacterial compounds using de novo design, molecular
docking and molecular dynamics studies. Journal of Biomolecular
Structure & Dynamics,38(6), 17981809. https://doi.org/10.1080/
07391102.2019.1617785
Jin, Z., Du, X., Xu, Y., Deng, Y., Liu, M., Zhao, Y., Zhang, B., Li, X., Zhang,
L., Peng, C., Duan, Y., Yu, J., Wang, L., Yang, K., Liu, F., Jiang, R., Yang,
X., You, T., Liu, X., Yang, H. (2020). Structure of Mpro from COVID-
JOURNAL OF BIOMOLECULAR STRUCTURE AND DYNAMICS 13
19 virus and discovery of its inhibitors. Nature,582(7811), 289293.
https://doi.org/10.1038/s41586-020-2223-y
Khan, R. J., Jha, R. K., Amera, G. M., Jain, M., Singh, E., Pathak, A., Singh,
R. P., Muthukumaran, J., & Singh, A. K. (2020). Targeting SARS-CoV-2:
A systematic drug repurposing approach to identify promising inhibi-
tors against 3C-like proteinase and 2-O-ribosemethyltransferase.
Journal of Biomolecular Structure & Dynamics,114. https://doi.org/10.
1080/07391102.2020.1753577
Khan, S. A., Zia, K., Ashraf, S., Uddin, R., & Ul-Haq, Z. (2020). Identification
of chymotrypsin-like protease inhibitors of SARS-CoV-2 via integrated
computational approach. Journal of Biomolecular Structure and
Dynamics,110. https://doi.org/10.1080/07391102.2020.1751298
Klein, K. O., Soliman, A. M., Bonafede, M., Nelson, J. K., & Grubb, E.
(2020). Treatment patterns, health resource utilization and costs
among central precocious puberty patients treated with leuprolide or
histrelin: An examination of the commercial and Medicaid popula-
tions. Journal of Medical Economics,23(4), 407414. https://doi.org/10.
1080/13696998.2019.1697700
Korb, O., St
utzle, T., & Exner, T. E. (2009). Empirical scoring functions for
advanced protein-ligand docking with plants. Journal of Chemical
Information and Modeling,49(1), 8496. https://doi.org/10.1021/
ci800298z
Kufareva, I., & Abagyan, R. (2012). Methods of protein structure compari-
son. Methods in Molecular Biology (Clifton, N.J.),857, 231257. https://
doi.org/10.1007/978-1-61779-588-6_10
Kumari, R., Kumar, R., & Lynn, A, Open Source Drug Discovery
Consortium. (2014). g_mmpbsa-A GROMACS tool for high-throughput
MM-PBSA calculations. Journal of Chemical Information and Modeling,
54(7), 19511962. https://doi.org/10.1021/ci500020m
Lamarre, D., Anderson, P. C., Bailey, M., Beaulieu, P., Bolger, G., Bonneau,
P., B
os, M., Cameron, D. R., Cartier, M., Cordingley, M. G., Faucher, A.-
M., Goudreau, N., Kawai, S. H., Kukolj, G., Lagac
e, L., LaPlante, S. R.,
Narjes, H., Poupart, M.-A., Rancourt, J., Llin
as-Brunet, M. (2003). An
NS3 protease inhibitor with antiviral effects in humans infected with
hepatitis C virus. Nature,426(6963), 186189. https://doi.org/10.1038/
nature02099
Laskowski, R. A., MacArthur, M. W., Moss, D. S., & Thornton, J. M. (1993).
PROCHECK: A program to check the stereochemical quality of protein
structures. Journal of Applied Crystallography,26(2), 283291. https://
doi.org/10.1107/S0021889892009944
Laskowski, R. A., & Swindells, M. B. (2011). LigPlotþ: Multiple ligand-pro-
tein interaction diagrams for drug discovery. Journal of Chemical
Information and Modeling,51(10), 27782786. https://doi.org/10.1021/
ci200227u
Lobanov, M. Y., Bogatyreva, N. S., & Galzitskaya, O. V. (2008). Radius of
gyration as an indicator of protein structure compactness. Molecular
Biology,42(4), 623628. https://doi.org/10.1134/S0026893308040195
Li, G., & De Clercq, E. (2020). Therapeutic options for the 2019 novel cor-
onavirus (2019-nCoV). Nature Reviews Drug Discovery,19(3), 149150.
https://doi.org/10.1038/d41573-020-00016-0
Morris, G. M., Ruth, H., Lindstrom, W., Sanner, M. F., Belew, R. K.,
Goodsell, D. S., & Olson, A. J. (2009). Software news and updates
AutoDock4 and AutoDockTools4: Automated docking with selective
receptor flexibility. Journal of Computational Chemistry,30(16),
27852791. https://doi.org/10.1002/jcc.21256
M
uller, F. O., Terblanch
e, J., Schall, R., Van Zyl Smit, R., Tucker, T., Marais,
K., Groenewoud, G., Porchet, H. C., Weiner, M., & Hawarden, D. (1997).
Pharmacokinetics of triptorelin after intravenous bolus administration
in healthy males and in males with renal or hepatic insufficiency.
British Journal of Clinical Pharmacology,44(4), 335341. https://doi.
org/10.1046/j.1365-2125.1997.t01-1-00592.x
Muralidharan, N., Sakthivel, R., Velmurugan, D., & Gromiha, M. M. (2020).
Computational studies of drug repurposing and synergism of lopina-
vir, oseltamivir and ritonavir binding with SARS-CoV-2 protease
against COVID-19. Journal of Biomolecular Structure and Dynamics,
16. https://doi.org/10.1080/07391102.2020.1752802
Paul, S. M., Mytelka, D. S., Dunwiddie, C. T., Persinger, C. C., Munos, B. H.,
Lindborg, S. R., & Schacht, A. L. (2010). How to improve R&D product-
ivity: the pharmaceutical industrys grand challenge. Nature Reviews
Drug Discovery,9(3), 203214. https://doi.org/10.1038/nrd3078
Perlman, S., & Netland, J. (2009). Coronaviruses post-SARS: Update on
replication and pathogenesis. Nature Reviews Microbiology,7(6),
439450. https://doi.org/10.1038/nrmicro2147
Pushpakom, S., Iorio, F., Eyers, P. A., Escott, K. J., Hopper, S., Wells, A.,
Doig, A., Guilliams, T., Latimer, J., McNamee, C., Norris, A., Sanseau, P.,
Cavalla, D., & Pirmohamed, M. (2019). Drug repurposing: Progress,
challenges and recommendations. Nature Reviews Drug Discovery,
18(1), 4158. https://doi.org/10.1038/nrd.2018.168
Riedl, M. A., Banerji, A., Manning, M. E., Burrell, E., Joshi, N., Patel, D.,
MacHnig, T., Tai, M. H., & Watson, D. J. (2018). Treatment patterns and
healthcare resource utilization among patients with hereditary
angioedema in the United States. Orphanet Journal of Rare Diseases,
13(1), 180. https://doi.org/10.1186/s13023-018-0922-3
Rosa, S. G. V., & Santos, W. C. (2020). Clinical trials on drug repositioning
for COVID-19 treatment. Revista Panamericana de Salud P
ublica,44,1.
https://doi.org/10.26633/RPSP.2020.40
Taha, M. O., Habash, M., Al-Hadidi, Z., Al-Bakri, A., Younis, K., & Sisan, S.
(2011). Docking-based comparative intermolecular contacts analysis
as new 3-D QSAR concept for validating docking studies and in silico
screening: NMT and GP inhibitors as case studies. Journal of Chemical
Information and Modeling,51(3), 647669. https://doi.org/10.1021/
ci100368t
Tyrrell, D. A., & Bynoe, M. L. (1966). Cultivation of viruses from a high
proportion of patients with colds. Lancet (London, England),1(7428),
7677. https://doi.org/10.1016/S0140-6736(66)92364-6 https://doi.org/
10.1016/S0140-6736(66)92364-6
Ul Qamar, M. T., Alqahtani, S. M., Alamri, M. A., & Chen, L.-L. (2020).
Structural basis of SARS-CoV-2 3CLpro and anti-COVID-19 drug discov-
ery from medicinal plants. Journal of Pharmaceutical Analysis.https://
doi.org/10.1016/j.jpha.2020.03.009
Umesh Kundu, D., Selvaraj, C., Singh, S. K., & Dubey, V. K. (2020).
Identification of new anti-nCoV drug chemical compounds from
Indian spices exploiting SARS-CoV-2 main protease as target. Journal
of Biomolecular Structure & Dynamics,19. https://doi.org/10.1080/
07391102.2020.1763202
Wahedi, H. M., Ahmad, S., & Abbasi, S. W. (2020). Stilbene-based natural
compounds as promising drug candidates against COVID-19. Journal
of Biomolecular Structure and Dynamics,110. https://doi.org/10.1080/
07391102.2020.1762743
Wang, D., Fang, L., Shi, Y., Zhang, H., Gao, L., Peng, G., Chen, H., Li, K., &
Xiao, S. (2016). Porcine epidemic diarrhea virus 3C-like protease regu-
lates its interferon antagonism by cleaving NEMO. Journal of Virology,
90(4), 20902101. https://doi.org/10.1128/JVI.02514-15
Wu, F., Zhao, S., Yu, B., Chen, Y. M., Wang, W., Song, Z. G., Hu, Y., Tao,
Z. W., Tian, J. H., Pei, Y. Y., Yuan, M. L., Zhang, Y. L., Dai, F. H., Liu, Y.,
Wang, Q. M., Zheng, J. J., Xu, L., Holmes, E. C., & Zhang, Y. Z. (2020).
A new coronavirus associated with human respiratory disease in
China. Nature,579(7798), 265269. https://doi.org/10.1038/s41586-
020-2008-3
Yang, H., Xie, W., Xue, X., Yang, K., Ma, J., Liang, W., Zhao, Q., Zhou, Z.,
Pei, D., Ziebuhr, J., Hilgenfeld, R., Kwok, Y. Y., Wong, L., Gao, G., Chen,
S., Chen, Z., Ma, D., Bartlam, M., & Rao, Z. (2005). Design of wide-spec-
trum inhibitors targeting coronavirus main proteases. PLoS Biology,
3(10), e324. https://doi.org/10.1371/journal.pbio.0030324
Zhu, X., Wang, D., Zhou, J., Pan, T., Chen, J., Yang, Y., Lv, M., Ye, X., Peng,
G., Fang, L., & Xiao, S. (2017). Porcine deltacoronavirus nsp5 antago-
nizes type I interferon signaling by cleaving STAT2. Virology,500(10),
9195. https://doi.org/10.1128/jvi.00003-17
Zoete, V., Cuendet, M. A., Grosdidier, A., & Michielin, O. (2011).
SwissParam: A fast force field generation tool for small organic mole-
cules. Journal of Computational Chemistry,32(11), 23592368. https://
doi.org/10.1002/jcc.21816
14 S. CHATTERJEE ET AL.
... The charmm36 [41] force field was used to translate the protein for molecular dynamics (MD) simulation. The protein-ligand system was incorporated in to a 10Ǻ × 10Ǻ × 10Ǻ cubic box using the TIP3P water model [42] followed by minimization [23] of the generated system. A total 2 ns each NVT and NPT ensembled equilibration was executed before 3rounds of 100 ns production run [38]. ...
... For a successful structure-based method, validated molecular docking protocol poses great importance. In this work, the re-docking method guided validation protocol depicts ligand superimposition (Supplementary Figure S1) with an RMSD value of ~ 0.87Ǻ [42], and the corresponding docking score was found to be − 7.0 kcal/mol. This value is far less than 2Ǻ [42] and strongly advocates the validity of the docking protocol used in this work. ...
... In this work, the re-docking method guided validation protocol depicts ligand superimposition (Supplementary Figure S1) with an RMSD value of ~ 0.87Ǻ [42], and the corresponding docking score was found to be − 7.0 kcal/mol. This value is far less than 2Ǻ [42] and strongly advocates the validity of the docking protocol used in this work. Employing an identical docking protocol. ...
Article
Full-text available
Cancer is one of the life-threatening diseases and the second leading cause of death in the world. The estrogen receptor can be considered as one of the significant drug targets for cancer. A large number of clinically used anticancer drugs were identified from phytochemicals. Multiple literatures suggested that extracts of Datura sp. significantly inhibit estrogen receptors associated with human cancer. In the present study, all reported natural products present in Datura sp. were subjected to molecular docking against estrogen receptors. The top hits were shortlisted based on binding orientation and docking score and subjected to molecular dynamics simulation to explore the conformational stability followed by binding energy calculation. The ligand [(1S,5R)-8-Methyl-8-Azabicyclo [3.2.1] Octan-3-yl] (2R)-3-Hydroxy-2-Phenylpropanoate depicts highly acceptable MD simulations outcomes and drug-likeness profile. Knowledge-based de novo design and similar ligand screening were executed using the structural information. The designed ligand DL-50 exhibited satisfactory binding, drug-likeness profile, and well-accepted ADMET profile followed by easy synthetic accessibility which further requires experimental validation. Graphical abstract
... This effect has been demonstrated through gene expression analysis at the molecular level, using highly accurate MD simulation of molecules. 39 Our study indicates that specific genes such as JAK-2, ERK-2, MCL-1, NFkB, and STAT-3 are activated following the activation of TPO-R (cMPL) in both hUCMSC and hGMSC. These findings are consistent with previous reports. ...
Article
Full-text available
Plain Language Summary What is the context? ● Chemotherapy, radiation treatment, or immunological disorders can cause a decrease in platelet count (thrombocytopenia) or decrease all blood cell types (pancytopenia) in the bone marrow. This can make it challenging to choose the appropriate cancer treatment plan. ● Eltrombopag (EPAG) is an oral non-peptide thrombopoietin (TPO) mimetic that activates the cMPL receptor in the body. This activation leads to cell differentiation and proliferation, stimulating platelet production and reducing thrombocytopenia. The cMPL receptor is present in liver cells, megakaryocytes, and hematopoietic cells. However, its effects on stem cell proliferation and differentiation are not entirely understood. What is the new? ● This study delves into the molecular interactions and therapeutic applications of EPAG, a small molecule that activates cMPL (TPO-R). ● The study offers a comprehensive analysis of the ligand-receptor complex formation, including an examination of downstream signaling elements. Furthermore, molecular dynamics simulations demonstrate the stability of the ligand when interacting with targeted proteins. ● The research investigates the presence of TPO-R on stem cell-derived endothelial cells, shedding insight into the ability of EPAG TPO-mimetic to promote angiogenesis and vasculature formation. ● The study revealed that EPAG has the potential to protect against UVB-induced radiation damage and stimulate stem cell growth. What is the implications? The study emphasizes the potential of EPAG as a promising option for addressing radiation injury and minimizing the adverse effects of radiotherapy. It could revolutionize treatments not only for thrombocytopenia but also for enhancing the growth of stem cells. Furthermore, the research deepens our understanding of EPAG’s molecular mechanisms, providing valuable insights for developing future drugs and therapeutic approaches for cell therapy to treat radiation damage.
... As reference drug, Remdesivir (-7.1 kcal/mol) lacks the hydrogen bonding with catalytic dyad but demonstrates a useful interaction with PHE-140, LEU-141, SER-144, HIS-163, HIS-164, ASN-142, and hydrophobic interaction as that of the native ligand with missing one or more residue with each other (Figure 1). This result was in coherence with the previously reported literature suggesting PHE-140 and LEU-141 as important interaction residues of 3CLpro [45,51]. ...
Article
Full-text available
In present study, the inhibitory potential of Neolamarckia cadamba phytoconstituents was investigated against SARS-CoV-2 3CL protease (3CL pro) (PDB ID: 6M2N). Molecular docking was analyzed using AutoDock Vina software by setting the grid parameter as X= -33.163, Y= -65.074 and Z= 41.434 with dimensions of the grid box 25 × 25 × 25 Å. Remdesivir was taken as the standard for comparative analysis along with inhibitor 5, 6, 7-trihydroxy-2-phenyl-4H-chromen-4-one. Furthermore, the exploration of 2 D Hydrogen-bond interactions was performed by Biovia Discovery Studio 4.5 program to identify the interactions between an amino acid of target and ligand followed by assessment of physicochemical properties using Lipinski’s rule and Swiss ADME database. The decent bonding scores of secondary metabolites owing to hydrogen bonding with catalytic residues suggest the effectiveness of these phytochemicals towards 3CLpro. The results are further consolidated positively by Lipinski’s rule and Swiss ADME prediction. Thus reasonably, observations with docking studies suggest possibility of phytochemicals from Neolamarckia cadamba to inhibit the 3CLpro and consequently would be explored further as agents for preventing COVID-19.
... Model structures were compared with the models developed by Robetta server (http://robetta.bakerlab. org/) [34] and trRosetta server [35,36]. (https://yanglab.nankai.edu. ...
Article
Klebsiella pneumoniae (Kp) is a common pathogen inducing catheter-related biofilm infections. Developing effective therapy to overcome antimicrobial resistance (AMR) in Kp is a severe therapeutic challenge that must be solved. This study aimed to prepare niosome-encapsulated GENT (Gentamicin) and EDTA (Ethylenediaminetetraacetic acid) (GENT-EDTA/Nio) to evaluate its efficacy toward Kp strains. The thin-film hydration method was used to prepare various formulations of GENT-EDTA/Nio. Formulations were characterized for their physicochemical characteristics. GENT-EDTA/Nio properties were used for optimization with Design-Expert Software. Molecular docking was utilized to determine the antibacterial activity of GENT. The niosomes displayed a controlled drug release and storage stability of at least 60 days at 4 and 25 °C. GENT-EDTA/Nio performance as antimicrobial agents has been evaluated by employing agar well diffusion method, minimum bactericidal concentration (MBC), and minimum inhibitory concentration (MIC) against the Kp bacteria strains. Biofilm formation was investigated after GENT-EDTA/Nio administration through different detection methods, which showed that this formulation reduces biofilm formation. The effect of GENT-EDTA/Nio on the expression of biofilm-related genes (mrkA, ompA, and vzm) was estimated using QRT-PCR. MTT assay was used to evaluate the toxicity effect of niosomal formulations on HFF cells. The present study results indicate that GENT-EDTA/Nio decreases Kp's resistance to antibiotics and increases its antibiotic and anti-biofilm activity and could be helpful as a new approach for drug delivery.
... The effective binding of these compounds to the M pro and their good ADMET profiles is in line with previous studies. [77][78][79][80][81][82] Thus, considering these compounds for further applications is of significance; however, in vitro, and in vivo chronic toxicity studies are pivotal. 52 Previous studies also reported similar findings to our study. ...
Article
Full-text available
Background: COVID-19 is still instigating significant social and economic chaos worldwide; however, there is no approved antiviral drug yet. Here, we used in silico analysis to screen potential SARS-CoV-2 main protease (Mpro) inhibitors extracted from the essential oil of Thymus schimperi which could contribute to the discovery of potent anti-SARS-CoV-2 phytochemicals. Methods: The absorption, distribution, metabolism, excretion, and toxicity (ADMET) profiles of compounds were determined through SwissADME and ProToxII servers. AutoDock tools were used for molecular docking analysis studies, while Chimera, DS studio, and LigPlot were used for post-docking studies. Molecular dynamic simulations were performed for 200 ns under constant pressure. Results: All compounds exhibited a bioavailability score of ≥0.55 entailing that at least 55% of the drugs can be absorbed unchanged. Only five (9%), nine (16%) and two (3.6%) of the compounds showed active hepatotoxicity, carcinogenicity, and immunotoxicity, respectively. Except for flourazophore P, which showed a little mutagenicity, all other compounds did not show mutagenic properties. On the other hand, only pinene beta was found to have a little cytotoxicity. Five compounds demonstrated effective binding to the catalytic dyad of the SARS-CoV-2 Mpro substrate binding pocket, while two of them (geranylisobutanoate and 3-octane) are found to be the best hits that formed hydrogen bonds with Glu166 and Ser144 of SARS-CoV-2 Mpro. Conclusion: Based on our in silico analysis, top hits from Thymus schimperi may serve as potential anti-SARS-CoV-2 compounds. Further in vitro and in vivo studies are recommended to characterize these compounds for clinical applications.
... Its good safety record [17] justifies repeated dosing to stop fluid leaking into the lungs as COVID-19 interstitial pneumonia unfolds. In addition, in silico screenings have consistently identified icatibant among other drugs as a potential SARS-CoV-2 inhibitor [18,19]. Previous clinical studies have yielded promising results [20,21]. ...
Article
Full-text available
Background: We aimed to evaluate icatibant, a competitive antagonist of the bradykinin B2 receptors, for the treatment of inpatients with COVID-19 pneumonia admitted in the early hypoxemic stage. Methods: The randomized, open-label clinical trial of icatibant for COVID-19 pneumonia (ICAT·COVID, registered as NCT04978051 at ClinicalTrials.gov) was conducted in Barcelona. Inpatients requiring supplemental but not high-flow oxygen or mechanical ventilation were allocated (1:1) to treatment with either three 30 mg icatibant doses/day for three consecutive days plus standard care or standard care alone, and followed for up to 28 days after initial discharge. The primary and key secondary outcomes were clinical response on study day 10/discharge and clinical efficacy at 28 days from initial discharge, respectively. Results: Clinical Response occurred in 27 out of 37 patients (73.0%) in the icatibant group and 20 out of 36 patients (55.6%) in the control group (rate difference, 17.42; 95% CI, -4.22 to 39.06; p = 0.115). Clinical efficacy ensued in 37 patients (100.0%) in the icatibant group and 30 patients (83.3%) in the control group (rate difference, 16.67; 95% CI, 4.49 to 28.84; p = 0.011). No patient died in the icatibant group, compared to six patients (16.7%) in the control group (p = 0.011). All patients but one had adverse events, which were evenly distributed between study arms. No patient withdrew because of adverse events. Conclusions: Adding icatibant to standard care was safe and improved both COVID-19 pneumonia and mortality in this proof-of-concept study. A larger, phase 3 trial is warranted to establish the clinical value of this treatment.
Chapter
Full-text available
Severe fever with thrombocytopenia syndrome virus (SFTSV) also known as Dabie bandavirus of the family Phenuiviridae is a negative-strand RNA virus and a tick-borne virus. Replication of SFTSV into systemic circulation and occurrence of viremia cause cytokine storm and T-cell overstimulation. The event of viremia-induced thrombocytopenia causes reduced platelet count and splenic macrophages, followed by endothelial damages and compromised immune system that cause multi-organ damages. Limited options for specific anti-SFTSV drugs pose significant challenges associated with clinical management of SFTSV infection. This book chapter chiefly emphasizes upon the genetic diversity, geographical distribution, pathogenesis associated with various clinical aspects like symptoms, diagnosis, and available clinical management options. In addition, current research linked with anti-SFTSV drug development is comprehensively portrayed in this review.KeywordsSevere Fever with Thrombocytopenia Syndrome virus (SFTSV)Dabie bandavirusHuaiyangshan BanyangvirusPhenuiviridaeClinical symptoms of SFTSV infectionClinical diagnosis of SFTSVSFTS L proteinSFTSV drug targetMolecular DockingMolecular Dynamics
Article
Full-text available
There is a dire need to develop any antiviral therapy for the treatment of SARS-CoV-2. Objective: To investigate the potential therapeutic drug agents from Cymbopogon citratus compounds against the main-protease (Mpro) of SARS-CoV-2. Methods: Initial screening was carried out using molecular docking, dynamic simulation followed by ADMET profiling and Lipinski’s physiochemical parameters for prediction of drug likeliness. MOE/PyRx was used for docking before determining the stability of the best complexes through NAMD/VMD softwares. Moreover, SwissADME and admetSAR web-based tools were used for drug likeliness of the best complexes. Results: Out of total 50 compounds, 11 presented the lowest binding energies which includes tannic acid, isoorientin, swertiajaponin, chlorogenic acid, cymbopogonol, warfarin, citral diethyl acetal, citral acetate, luteolin, kaempferol and cianidanol with binding energies of -8.12, -7.38, -7.33, -6.88, -6.48, -6.32, -6.31, -6.18, -6.18, -6.13 and -6.02, respectively. Current studies show isoorientin, chlorogenic acid and tannic acid as the promising drug agents using RMSD, Hbond, heatmap graphs. Conclusion: Further in-vivo experiments are suggested to ascertain the medicinal use of these potential inhibitors against COVID-19.
Article
Full-text available
The emergence of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) from China in December 2019 led to the coronavirus disorder 2019 pandemic, which has affected tens of millions of humans worldwide. Various in silico research via bio-cheminformatics methods were performed to examine the efficiency of a range of repurposed approved drugs with a new role as anti-SARS-CoV-2 drugs. The current study has been performed to screen the approved drugs in the DrugBank database based on a novel bioinformatics/cheminformatics strategy to repurpose available approved drugs towards introducing them as a possible anti-SARS-CoV-2 drug. As a result, 96 approved drugs with the best docking scores passed through several relevant filters were presented as the candidate drugs with potential novel antiviral activities against the SARS-CoV-2 virus.
Article
Full-text available
The 2019-novel coronavirus (nCoV) has caused a global health crisis by causing coronavirus disease-19 (COVID-19) pandemic in the human population. The unavailability of specific vaccines and anti-viral drug for nCoV, science demands sincere efforts in the field of drug design and discovery for COVID-19. The novel coronavirus main protease (SARS-CoV-2 Mpro) play a crucial role during the disease propagation, and hence SARS-CoV-2 Mpro represents as a drug target for the drug discovery. Herein, we have applied bioinformatics approach for screening of chemical compounds from Indian spices as potent inhibitors of SARS-CoV-2 main protease (PDBID: 6Y84). The structure files of Indian spices chemical compounds were taken from PubChem database or Zinc database and screened by molecular docking, by using AutoDock-4.2, MGLTools-1.5.6, Raccoon virtual screening tools. Top 04 hits based on their highest binding affinity were analyzed. Carnosol exhibited highest binding affinity -8.2 Kcal/mol and strong and stable interactions with the amino acid residues present on the active site of SARS-CoV-2 Mpro. Arjunglucoside-I (-7.88 Kcal/mol) and Rosmanol (-7.99 Kcal/mol) also showed a strong and stable binding affinity with favourable ADME properties. These compounds on MD simulations for 50ns shows strong hydrogen-bonding interactions with the protein active site and remains stable inside the active site. Our virtual screening results suggest that these small chemical molecules can be used as potential inhibitors against SARS-CoV-2 Mpro and may have an anti-viral effect on nCoV. However, further validation and investigation of these inhibitors against SARS-CoV-2 main protease are needed to claim their candidacy for clinical trials.
Article
Full-text available
The pandemic coronavirus disease (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) presents a great threat to public health. Currently, no potent medicine is available to treat COVID-19. Quest for new drugs especially from natural plant sources is an area of immense potential. The current study aimed to repurpose stilbenoid analogs, reported for some other biological activities, against SARS-CoV-2 spike protein and human ACE2 receptor complex for their affinity and stability using molecular dynamics simulation and binding free energy analysis based on molecular docking. Four compounds in total were probed for their binding affinity using molecular docking. All of the compounds showed good affinity (> -7 kcal/mol). However, fifty nanoseconds molecular dynamic simulation in aqueous solution revealed highly stable bound conformation of resveratrol to the viral protein: ACE2 receptor complex. Net free energy of binding using MM-PBSA also affirmed the stability of the resveratrol-protein complex. Based on the results, we report that stilbene based compounds in general and resveratrol, in particular, can be promising anti-COVID-19 drug candidates acting through disruption of the spike protein. Our findings in this study are promising and call for further in vitro and in vivo testing of stiblenoids, especially resveratrol against the COVID-19.
Article
Full-text available
The main protease of SARS-CoV-2 is one of the important targets to design and develop antiviral drugs. In this study, we have selected 40 antiviral phytochemicals to find out the best candidates which can act as potent inhibitors against the main protease. Molecular docking is performed using AutoDock Vina and GOLD suite to determine the binding affinities and interactions between the phytochemicals and the main protease. The selected candidates strongly interact with the key Cys145 and His41 residues. To validate the docking interactions, 100 ns molecular dynamics (MD) simulations on the five top-ranked inhibitors including hypericin, cyanidin 3-glucoside, baicalin, glabridin, and α-ketoamide-11r were performed. Principal component analysis (PCA) of potential energy components of MD discloses that baicalin, cyanidin 3-glucoside, and α-ketoamide-11r have structural similarity with the apo-form of the main protease. These findings are also strongly supported by root-mean-square deviation (RMSD), root-mean-square fluctuation (RMSF), radius of gyration (Rg), and solvent accessible surface area (SASA) investigations. PCA is also used to find out the quantitative structure-activity relationship (QSAR) for pattern recognition of the best ligands. Multiple linear regression (MLR) of QSAR reveals the R² value of 0.842 for the training set and 0.753 for the test set. Our proposed MLR model can predict the favorable binding energy compared with the binding energy detected from molecular docking. ADMET analysis demonstrates that these candidates appear to be safer inhibitors. Our comprehensive computational and statistical analysis show that these selected phytochemicals can be used as potential inhibitors against the SARS-CoV-2.
Article
Full-text available
In the past two decades, the world has faced several infectious disease outbreaks. Ebola, Influenza A (H1N1), SARS, MERS, and Zika virus have had a massive global impact in terms of economic disruption, the strain on local and global public health. Most recently, the global outbreak of novel coronavirus 2019 (SARS-CoV-2) that causes COVID-19 is a newly discovered virus from the coronavirus family in Wuhan city, China, known to be a great threat to the public health systems. As of 15 April 2020, The Johns Hopkins University estimated that the COVID-19 affected more than two million people, resulting in a death toll above 130,000 around the world. Infected people in Europe and America correspond about 40% and 30% of the total reported cases respectively. At this moment only few Asian countries have controlled the disease, but a second wave of new infections is expected. Predicting inhibitor and target to the COVID-19 is an urgent need to protect human from the disease. Therefore, a protocol to identify anti-COVID-19 candidate based on computer-aided drug design is urgently needed. Thousands of compounds including approved drugs and drugs in the clinical trial are available in the literature. In practice, experimental techniques can measure the time and space average properties but they cannot be captured the structural variation of the COVID-19 during the interaction of inhibitor. Computer simulation is particularly suitable to complement experiments to elucidate conformational changes at the molecular level which are related to inhibition process of the COVID-19. Therefore, computational simulation is essential tool to elucidate the phenomenon. The structure-based virtual screening computational approach will be used to filter the best drugs from the literature, the investigate the structural variation of COVID-19 with the interaction of the best inhibitor is a fundamental step to design new drugs and vaccines which can combat the coronavirus. This mini-review will address novel coronavirus structure, mechanism of action, and trial test of antiviral drugs in the lab and patients with COVID-19.
Article
Full-text available
A new coronavirus (CoV) identified as COVID-19 virus is the etiological agent responsible for the 2019-2020 viral pneumonia outbreak that commenced in Wuhan1–4. Currently there are no targeted therapeutics and effective treatment options remain very limited. In order to rapidly discover lead compounds for clinical use, we initiated a program of combined structure-assisted drug design, virtual drug screening and high-throughput screening to identify new drug leads that target the COVID-19 virus main protease (Mpro). Mpro is a key CoV enzyme, which plays a pivotal role in mediating viral replication and transcription, making it an attractive drug target for this virus5,6. Here, we identified a mechanism-based inhibitor, N3, by computer-aided drug design and subsequently determined the crystal structure of COVID-19 virus Mpro in complex with this compound. Next, through a combination of structure-based virtual and high-throughput screening, we assayed over 10,000 compounds including approved drugs, drug candidates in clinical trials, and other pharmacologically active compounds as inhibitors of Mpro. Six of these compounds inhibited Mpro with IC50 values ranging from 0.67 to 21.4 μM. Ebselen also exhibited promising antiviral activity in cell-based assays. Our results demonstrate the efficacy of this screening strategy, which can lead to the rapid discovery of drug leads with clinical potential in response to new infectious diseases for which no specific drugs or vaccines are available.
Article
Full-text available
The recent pandemic associated with SARS-CoV-2, a virus of the Coronaviridae family, has resulted in an unprecedented number of infected people. The highly contagious nature of this virus makes it imperative for us to identify promising inhibitors from pre-existing antiviral drugs. Two druggable targets, namely 3C-like proteinase (3CLpro) and 2'-O-ribose methyltransferase (2'-O-MTase) were selected in this study due to their indispensable nature in the viral life cycle. 3CLpro is a cysteine protease responsible for the proteolysis of replicase polyproteins resulting in the formation of various functional proteins, whereas 2'-O-MTase methylates the ribose 2'-O position of the first and second nucleotide of viral mRNA, which sequesters it from the host immune system. The selected drug target proteins were screened against an in-house library of 123 antiviral drugs. Two promising drug molecules were identified for each protein based on their estimated free energy of binding (ΔG), the orientation of drug molecules in the active site and the interacting residues. The selected protein-drug complexes were then subjected to MD simulation, which consists of various structural parameters to equivalently reflect their physiological state. From the virtual screening results, two drug molecules were selected for each drug target protein [Paritaprevir (ΔG= -9.8 kcal/mol) &Raltegravir (ΔG= -7.8 kcal/mol) for 3CLpro and Dolutegravir (ΔG= -9.4 kcal/mol) and Bictegravir (ΔG= -8.4 kcal/mol) for 2'-OMTase]. After the extensive computational analysis, we proposed that Raltegravir, Paritaprevir, Bictegravir and Dolutegravir are excellent lead candidates for these crucial proteins and they could become potential therapeutic drugs against SARS-CoV-2.
Article
The recent pandemic of coronavirus disease 2019 (COVID-19) caused by SARS-CoV-2 has raised global health concerns. The viral 3-chymotrypsin-like cysteine protease (3CL pro) enzyme controls coronavirus replication and is essential for its life cycle. 3CLpro is a proven drug discovery target in the case of severe acute respiratory syndrome coronavirus (SARS-CoV) and Middle East respiratory syndrome coronavirus (MERS-CoV). Recent studies revealed that the genome sequence of SARS-CoV-2 is very similar to that of SARS-CoV. Therefore, herein, we analysed the 3CLpro sequence, constructed its 3D homology model, and screened it against a medicinal plant library containing 32,297 potential anti-viral phytochemicals/ traditional Chinese medicinal compounds. Our analyses revealed that the top nine hits might serve as potential anti-SARS-CoV-2 lead molecules for further optimisation and drug development process to combat COVID-19.
Article
A new strain of a novel infectious disease affecting millions of people, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has recently been declared as a pandemic by the World Health Organization (WHO). Currently, several clinical trials are underway to identify specific drugs for the treatment of this novel virus. The inhibition of the SARS-CoV-2 main protease is necessary for the blockage of the viral replication. Here, in this study, we have utilized a blind molecular docking approach to identify the possible inhibitors of the SARS-CoV-2 main protease, by screening a total of 33 molecules which includes natural products, anti-virals, anti-fungals, anti-nematodes and anti-protozoals. All the studied molecules could bind to the active site of the SARS-CoV-2 protease (PDB: 6Y84), out of which rutin (a natural compound) has the highest inhibitor efficiency among the 33 molecules studied, followed by ritonavir (control drug), emetine (anti-protozoal), hesperidin (a natural compound), lopinavir (control drug) and indinavir (anti-viral drug). All the molecules, studied out here could bind near the crucial catalytic residues, HIS41 and CYS145 of the main protease, and the molecules were surrounded by other active site residues like MET49, GLY143, HIS163, HIS164, GLU166, PRO168, and GLN189. As this study is based on molecular docking, hence being particular about the results obtained, requires extensive wet-lab experimentation and clinical trials under in vitro as well as in vivo conditions.
Article
The cyclin-dependent kinase-2 (CDK2) belongs to the protein kinase family and its overexpression leads to an unusual regulation of cell-cycle which directly linked with hyperproliferation in many cancer cell types. CDK2 activation spontaneously promotes the cell cycle progression and also involved in a large number of cellular processes including cell cycle regulation, DNA replication, DNA damage response and apoptotic pathways, therefore targeting the CDK2 can be reemerged as a therapeutic boulevard to restrain cancer cell proliferation. For the last two decades, emerging evidence suggested that CDK2 inhibition draws out some antitumor/anticancer activity, which has driven the research possibility for developing next-generation newer or cost-effective inhibitors with greater specificity to CDK2. In the current work, compounds from the FooDB - a world's largest food constituents database was retrieved and curated and followed by multi-pharmacoinformatics approaches adopted to find out potential CDK2 inhibitors. The curated dataset was considered for “Virtual Screening Workflow” (VSW) employed in Schrödinger suite. The numbers of cost-effective food constituents were reduced by removing low potential molecules in terms of interaction affinity and further explored for pharmacokinetics analysis. Based on strong binding interaction profiles with the lowest binding interactions affinity and energy values, four food compounds were proposed as CDK2 inhibitors. A number of key analyzing parameters from MD simulations studies were successfully substantiated that all four proposed food compounds can act as CDK2 inhibitors based on their proficient structural and molecular interactions integrity with CDK2 protein following in the active site cavity. Furthermore, the binding free energy was calculated using the MM-PBSA (Molecular Mechanics Poisson-Boltzmann Surface Area) approach from the entire trajectory frames derived in MD simulation revealed strong interaction affinity. The binding free energy was found to be in the range of −991.831 to −210.452 kJ/mol. High binding free energy was undoubtedly explained that all molecules possess strong affection towards CDK2. Hence, proposed molecules may be crucial to stop the hyperproliferation in cancer cells subjected to experimental validation.
Article
The widespread antigenic changes lead to the emergence of a new type of coronavirus (CoV) called as severe acute respiratory syndrome (SARS)-CoV-2 that is immunologically different from the previous circulating species. Angiotensin-converting enzyme-2 (ACE-2) is one of the most important receptors on the cell membrane of the host cells (HCs) which its interaction with spike protein (SP) with a furin-cleavage site results in the SARS-CoV-2 invasion. Hence, in this review, we presented an overview on the interaction of ACE-2 and furin with SP. As several kinds of CoVs, from various genera, have at their S1/S2 binding site a preserved site, we further surveyed the role of furin cleavage site (FCS) on the life cycle of the CoV. Furthermore, we discussed that the small molecular inhibitors can limit the interaction of ACE-2 and furin with SP and can be used as potential therapeutic platforms to combat the spreading CoV epidemic. Finally, some ongoing challenges and future prospects for the development of potential drugs to promote targeting specific activities of the CoV were reviewed. In conclusion, this review may pave the way for providing useful information about different compounds involved in improving the effectiveness of CoV vaccine or drugs with minimum toxicity against human health. Communicated by Ramaswamy H. Sarma