Available online on 15.09.2024 at http://jddtonline.info
Journal of Drug Delivery and Therapeutics
Open Access to Pharmaceutical and Medical Research
Copyright © 2024 The Author(s): This is an open-access article distributed under the terms of the CC BY-NC 4.0 which permits unrestricted use, distribution, and reproduction in any medium for non-commercial use provided the original author and source are credited
Open Access Full Text Article Research Article
Virtual design and screening of new (+)-catechin derivates N- and/or S-heterocyclic fragment for anti-malarial and anti-SARS-CoV-2 activities by In silico simulation
Ahmed Said Mohamed 1*, Imane Yamari 2, Nouh Mounadi 2, Abdirahman Elmi 1, Mohammed Bouachrine 3, Hanane Zaki 3, Samir Chtita 2
1 Centre d’Étude et de Recherche de Djibouti, Institut de Recherche Médicinale, Route de l’aéroport, Djibouti.
2 Laboratory Physical Chemistry of Materials, Faculty of Sciences Ben M’Sik, Hassan II University of Casablanca, B.P. 7955 Sidi Othmane, Casablanca, Morocco.
3 3Bio Laboratory, Higher School of Technology Khenifra, Sultane Moulay Slimane University, Khenifra, Morocco.
Article Info: _____________________________________________________ Article History: Received 24 June 2024 Reviewed 03 August 2024 Accepted 25 August 2024 Published 15 Sep 2024 _____________________________________________________ Cite this article as: Mohamed AS, Yamari I, Mounadi N, Elmi A, Bouachrine M, Zaki H, Chtita S, Virtual design and screening of new (+)-catechin derivates N- and/or S-heterocyclic fragment for anti-malarial and anti-SARS-CoV-2 activities by In silico simulation, Journal of Drug Delivery and Therapeutics. 2024; 14(9):35-50 DOI: http://dx.doi.org/10.22270/jddt.v14i9.6762 ______________________________________________________ *Address for Correspondence: Ahmed Said Mohamed, Centre d’Étude et de Recherche de Djibouti, Institut de Recherche Médicinale, Route de l’aéroport, Djibouti. |
Abstract __________________________________________________________________________________________________________________ Hemisynthesis makes it possible to improve the activity or reduce the toxicity of a biocompound by modifying or adding peripheral groups. Catechin present in different plant species was known for its moderate anti-malarial and anti-SARS-CoV-2 activities. The aim of this work was focused on the identification of new compounds with potential anti-SARS-CoV-2 and/or anti-malarial activities, evaluated through In silico simulation. A polyphenol pharmacophore model, based on (+)-catechin (1) was virtually constructed using previously reported inhibitors. N- and/or S-heterocyclic fragments were inserted on the backbone of (+)-catechin and 12 pharmacophore hypotheses were studied. This study targeted 3 proteins biologically responsible for SARS-CoV-2 (PDB ID: 7JYC, 6M0J, and 6HZD) and one protein responsible for malaria (PDB ID: 3SRJ). Molecular docking had shown that the new catechin-aldehyde candidates have good Ligand-Protein affinity in terms of free energy compared to the study reference Narlaprevir and Artesunate for SARS-CoV-2 and malaria respectively. Theoretically, most compounds didn’t show toxicity except compounds 2a, 2i, and 2k, exhibiting hepatotoxic activity. Molecular dynamics was used to prove and assess their binding stability to the target protein for each activity. The 3SRJ-2f and 6HZD-2l structures were selected for anti-malarial and anti-SARS-CoV-2 activity respectively. The 3SRJ-2f and 6HZD-2l complexes showed stable interactions to 100 ns between the inhibitor fragments and the residual amino acids of the protein. To conclude, these novel compounds are probably to become promising lead molecules for the development of effective anti-SARS-CoV-2 and/or anti-malarial of all drugs. Keywords: Catechin, Anti-malarial, Anti-SARS-CoV-2, Docking and Dynamic Molecular, ADMET Analysis. |
1. INTRODUCTION
Catechin is a natural compound of the flavonoid family, often found in green tea but also in some medicinal plants such as the fruits of the Acacia catechu 1 or also in bark of Acacia seyal of Republic of Djibouti 2. It was characterized by its colorless shade, its good solubility in water and by its molecular structure, bearing two phenolic rings separated by a central heterocyclic and its several hydroxyl groups. Catechin well known for their potential antioxidant properties 3–5. Several studies have been carried out, showing the beneficial effect of catechin for the prevention of certain types of cancers 6–8, reduction of the risks of obesity, diabetes, and cardiovascular diseases and improvement of the immune system. It was also shown that catechin or catechin derivates can be a promising candidate for anti-malarial 9–13 and anti-SARS-CoV-2 activities14–16.
A. R. Sannella et al., in 2007 showed by in vitro anti-malarial test that the two main constituents: epigallocatechin-3-gallate (EGCG) and epicatechin gallate (ECG) of catechin derivatives and crude extract of green tea inhibit the growth of Plasmodium falciparum. In addition, epigallocatechin-3-gallate (EGCG) and epicatechin gallate (ECG) have also been shown to potentiate, at least moderately, the antiplasmodial effects of artemisinin, when the latter is administered at sublethal doses 17.
A study was also carried out by Iwan Budiman et al., in 2015, to evaluate anti-malarial activities of various catechins including catechin (C), epicatechin (EC), catechin-gallate (CG), gallocatechingallate (GCG), epigallocatechin (EGC), epicatechin-gallate (ECG), epigallocatechin-gallate (EGCG). The IC50 of catechins in Plasmodium falciparum after 48 hours incubation compared to a reference Artemisinin. This work showed that the most active anti-malarial activity was CG (IC50= 0.366 μM) and the lowest anti-malarial activity was EGC (IC50 = 98.145 μM). C, EC, CG grouped as active anti-malarial activity with IC50: 0.734, 0.456, and 0.366 µM respectively; GC (7.457 μM), ECG (2.049 μM), EGCG (5.633 μM) as moderate active anti-malarial activity; GCG, and EGC as very weak anti-malarial activity 18. These studies confirm that structural modification plays a key role in the influence of anti-malarial activity.
Graphical abstract
The appearance of Covid-19 at the end of 2019, researchers are also focused on finding attempts to stop this global pandemic19. In 2020 to 2023, several studies were published evaluating certain natural substances by In silico and/or by In vitro as anti-Covid-19 agents 20–23. Catechin and its derivatives are also being tested against SARS-CoV-2 24–26.
The in silico studies carried out by Susmit Mhatre et al., in 2021 showed that the catechins and its derivates form favorable interactions with the spike protein (The prefusion SARS-CoV-2 spike glycoprotein (wild and mutated strains): PDB ID: 6VSB) and can potentially impair its function. Epigallocatechin gallate (EGCG) showed the best binding (-6.3, and -6.4 kJ/mol wild and mutated strains respectively) among the catechin against both the strains. The results are encouraging for further exploration of the antiviral activity of EGCG against SARS-CoV-2 and its variants27.
A recent study by Shaik et al., in 2022 highlights the role of catechins as entry-inhibitors against SARS-CoV-2 by in silico. The lead compounds were EGCG and ECG act as potential inhibitors bind to the active site region of the HKU4eCoV 3C-like protease (PDB ID: 4YOG) and M-Pro protease (PDB ID: 7CAM) enzymes of coronavirus28.
2. MATERIALS AND METHODS
2.1 Proposal of (+)-catechin derivative structures (2).
The serie of catechin derivatives were inspired by the work carried out by Katsuko Kajiya et al., in 2004. Generally, the type of compound was obtained by reaction of aldehyde and (+)-catechin in acid medium in the presence of methyl mercaptan34.
Compounds 2a-l were virtually designed with existing aldehyde at Sigma Aldrich, to assess their biological capacity for anti-malarial and anti-SARS-CoV-2 activities through bioinformatics. The skeleton of (+)-catechin was modified on the two carbon positions C6 and C8 of ring A with homo-heterocyclic fragments. For that, the 3D-structures of the 12 inhibitors were prepared using a molecule editor in ChemDraw professional 15.0 and were generated for all ligands with LigPrep.
Chart 1: Different stereoisomer of compound (+)-catechin-aldehyde.
Generally, the experimental reaction resulted in a mixture of four stereoisomers, due to the presence of two other asymmetric carbons carried by the carbon position 6 and 8 (Chart 1). In the context of this article, the simulation was carried out with an identical structural conformation, giving the (R, R) stereoisomer for all the structures except for compounds 2a, and 2b with an (S, S) configuration (Table 1).
Table 1: Details of virtual screening compounds.
|
||
Ligands |
Aldéhyde names |
Structures |
2a |
2-hydroxycarboxaldehyde |
|
2b |
4-Pyridinecarboxaldehyde |
|
2c |
2-Thiophenecarboxaldehyde |
|
2d |
Pyrrole-2-carboxaldehyde |
|
2e |
Cinnamaldehyde |
|
2f |
Benzo[b]thiophene-2-carboxaldehyde |
|
2g |
2-Pyridinecarboxaldehyde |
|
2h |
N-Methyl-2-pyrrolecarboxaldehyde |
|
2i |
5-Thiazolecarboxaldehyde |
|
2j |
5-Oxazolecarboxaldehyde |
|
2k |
Benzothiazole-2-carboxaldehyde |
|
2l |
Indole-3-carboxaldehyde |
|
2.2. In silico study of (+)-catechin derivate compounds.
2.2.1. Molecular Docking.
ADMET (Absorption, Distribution, Metabolism, Elimination, Toxicity) analysis were calculated using the Swiss adme41 for assessing the drug ability and to filter the ligand molecules at an early stage of identifying the new inhibitors. Toxicity was the degree to which a substance can damage an organism or substructure of the organism. The predictions of toxicity of the compounds were essential to reduce the cost and labor of a drug's preclinical and clinical trials. The toxicity evaluation was performed also using the ProTox platform 42. It gave predicted toxicity values, cytotoxicity, mutagenicity, carcinogenicity, immunotoxicity, and LD50 values of selected compounds.
We were interested to two structures showing highest binding affinity, one towards anti-plasmodium (3SRJ–2f) and another for anti-SARS-CoV-2 (6HZD–2l) were selected for Molecular Dynamics Simulation (MDS) studies. Using the OPLSe3 force field43, the MDS was conducted with Schrodinger Desmond software44 to analyze the movements of biomolecule atoms in the presence of tiny molecules and to determine the stability of ligand-protein interactions45–47. A basic point charge water model SPC was used in the simulation. To balance the MD-simulated net charge system, the protein-ligand complex was neutralized by adding Na+ or Cl- ions, and 0.15 M NaCl was maintained to replicate a physiological ion concentration. The simulation was then run in an orthorhombic box with a 10 Ǻ x 10 Ǻ x 10 Ǻ dimensional space and NPT ensemble, starting with 1 ns at NVT equilibrium at 300 K, followed by 100 ns at standard pressure (1.01325 bar)48. Root Mean Square Deviation (RMSD), Root Mean Square Fluctuation (RMSF), and protein-ligand interaction were among the metrics measured by MDS.
3. RESULTS AND DISCUSSIONS
3.1. Anti-SARS-CoV-2 activity.
3.1.1. Bibliographic study of catechin and its derivatives against SARS-CoV-2 by in silico.
Table 2: Study comparisons carried out by in silico for catechin and its derivatives against SARS-CoV-2.
Name of compound |
Binding Energy Kcal/mol |
PDB ID |
Targed Protein |
References |
Epicatechin-gallate |
-7.52 |
4YOG |
Catalytic Residues of 3C-like protease |
28 |
Epigallocatechin-3-gallate |
-7.25 |
|||
Epicatechin-gallate |
-6.85 |
7CAM |
Catalytic Residues of M-Pro protease |
|
Epigallocatechin-3-gallate |
-7.57 |
|||
Epigallocatechin |
-7.0 |
6LU7 |
The SARS CoV-2 Mpro |
49 |
Gallocatechin |
-7.1 |
|||
Catechin |
-7.1 |
|||
Epicatechin |
-7.2 |
|||
Catechin-gallate |
-7.2 |
|||
Epigallocatechin-3-gallate |
-7.6 |
|||
Epicatechin-gallate |
-8.2 |
|||
Gallocatechingallate |
-9.0 |
|||
Epigallocatechin-3-gallate |
-7,8 |
6M17 |
The spike receptor-binding domain (RBD) and human cell receptor angiotensin-converting enzyme 2 (ACE2) |
50 |
Epigallocatechin-3-gallate |
-8,601 |
7CMD |
SARS-CoV-2 PLProprotein |
51 |
Epicatechin-gallate |
-8,566 |
|||
Gallocatechingallate |
-7,865 |
|||
Catechin-gallate |
-7,498 |
|||
Gallocatechin |
-6,337 |
|||
(-)-Catechin |
-6,329 |
|||
Epigallocatechin |
-6,318 |
|||
Epicatechin |
-6,128 |
|||
(-)-Catechin |
-14.1370 |
6LU7 |
Receptor main Protease |
52 |
(+)-Catechin |
-13.5387 |
|||
(-)-Epicatechin |
-13.6338 |
|||
(+)-Epicatechin |
-12.2790 |
|||
(-)-Catechin gallate |
-12.9942 |
|||
(+)-Catechin gallate |
-13.7186 |
|||
Catechin 3'-O-gallate |
-14.4852 |
|||
Catechin 5-O-gallate |
-14.6331 |
|||
Catechin 7-O-gallate |
-15.0299 |
|||
Catechin 3'-glucoside |
-12.3104 |
|||
Catechin 5-glucoside |
-11.9737 |
|||
(-)-Catechin |
-11.3222 |
6LXT |
Receptor spike glycoprotein |
|
(+)-Catechin |
-10.4993 |
|||
(-)-Epicatechin |
-11.5055 |
|||
(+)-Epicatechin |
-10.3292 |
|||
(-)-Catechin gallate |
-12.4199 |
|||
(+)-Catechin gallate |
-13.0183 |
|||
Catechin 3'-O-gallate |
-11.8377 |
|||
Catechin 5-O-gallate |
-11.3650 |
|
||
Catechin 7-O-gallate |
-11.9637 |
|||
Catechin 3'-glucoside |
-10.5722 |
|||
Catechin 5-glucoside |
-10.8447 |
|||
(+)-Catechin |
-4.89 |
6MOJ |
ACE2 |
53 |
-8.34 |
6M2N |
3-chymotrypsin-like cysteine protease (3CLpro) |
||
-7.68 |
6F06 |
CTSL |
||
-6.23 |
6M3M |
Crystal structure of nucleocapsid protein |
||
ND |
6M71 |
RdRp |
||
-7.04 |
NSP6 |
non-structural protein 6 |
||
-5.79 |
7BV2 |
cryo–electron microscopy structure of RdRp enzyme remdesivir and NSP12-NSP7-NSP8 complex |
||
-7.240 |
6LUZ |
3CLpro/Mpro |
54 |
|
-7.677 |
5R84 |
SARS-CoV-2 Mp |
55 |
|
-6.470 |
6VW1 |
SARS-CoV-2 RBD |
||
-5.856 |
5MIM |
Human Furin protease |
||
Catechin |
-10.50 |
6VSB |
S protein of SARS-CoV2 |
56 |
-8.9 |
1R42 |
ACE2 receptor |
||
-9.1 |
6LZG |
RBD/ACE2- complex |
3.1.2. Docking Molecular for SARS-CoV-2.
Molecular docking generally consisted to involve docking small molecules to a known protein structure. This technology was considered to be the basic method and main strategy for drug discovery or guiding the synthesis of optimal molecular structures with biological activity. It allows users to simulate interactions between chemical species and proteins at the atomic level, thereby describing the position and conformation of the interaction of the species with the target protein and elucidating the fundamental mechanisms underlying the regulation of biological processes57.
In this part of our investigation, we reported the molecular docking of the 12 (+)-catechin derivates designed set of molecules with the reference drug Narlaprevir 58, in the active site of the 3D structures protein, including 3CLpro/Mpro (7JYC), Furin (6HZD), and the ACE2 receptor (6M0J). The binding energies are represented in table 1 and the 2D interactions were illustrated in figure S2 in supporting information.
Docking scores were used to compare the best biological responses (Ligand-protein interaction) with the Narlaprevir reference and the base molecule: (+)-catechin. Compounds 2a-1 showed binding energies ranging from -7.4 to -9.2 kcal/mol, -8 to -10.1 kcal/mol, and -7.6 to -9.6 kcal/mol against 3CLpro/Mpro, ACE2, and Furin respectively. The molecular structures 2a, 2l, 2f, and 2i showed good affinity with the protein target 3CLpro/Mpro, the compounds 2l, and 2f for the ACE2 receptor and finally all compounds except compound 2j for the in Furin receptor.
Table 3: Molecular docking score against SARS-CoV-2 proteins, in bold most active compounds compared to references drug.
Compounds |
Docking score(kcal/mol) |
||
7JYC |
6M0J |
6HZD |
|
(+)-Catechin (1) |
-8,1 |
-7,6 |
-8 |
2a |
-8,2 |
-8,5 |
-9 |
2b |
-7,9 |
-8 |
-8,5 |
2c |
-7,5 |
-7,9 |
-8,3 |
2d |
-7,6 |
-8 |
-8,2 |
2e |
-7,6 |
-8,8 |
-9,1 |
2f |
-8,8 |
-9 |
-10,1 |
2g |
-7,4 |
-7,8 |
-8,3 |
2h |
-8 |
-7,9 |
-8,1 |
2i |
-7,9 |
-7,8 |
-8,1 |
2j |
-7,7 |
-7,9 |
-8 |
2k |
-9,2 |
-9,5 |
-9,5 |
2l |
-8,5 |
-9,6 |
-10,1 |
Narlaprevir |
-7,7 |
-8,9 |
-7,8 |
Figure 1 : (a) 2D and (b) 3D interactions visualization of the 2k compound with receptor 3CLpro/Mpro.
- Molecular docking against ACE2 protein (6M0J).
Proceeding to the next protein, it was known that the virus enters the host cell by binding the viral spike glycoprotein to the host receptor, angiotensin converting enzyme 2 (ACE2) 59 therefore (6M0J) seems to be a biologically meaningful receptor. We found that the compounds 2l, 2k, and 2f had a negative docking score higher than the reference drugs with a score of -9.6, and -9.0 kcal/mol respectively (Table 3). They both (2l and 2f) binded with hydrogen bonds with GLU406, alkyl bonds with residue ALA413, and LEU410, and π-Alkyl interactions with LEU370, and MET366. Additionally, the compound 2l binds with THR445 and ASN290 to establish other hydrogen bond interactions. Compound 2k also showed several types of bond with the protein: hydrogen bond of 1.86 Å with residue ALA348, bond π-π stacked with the residue PHE40, bond π-anion with the residue GLU402 and finally a bond π-sulfur with the residue HIS401. By our results, 2l had proven to be more effective than 2k and 2f.
Figure 2: (a) 2D and (b) 3D interactions visualization of the 2l compound with ACE2 receptor.
- Molecular docking against furin in complex with the cyclic inhibitor c [glutaryl-Arg-Arg-Arg-Lys]-Arg-4-Amba (6HZD).
Among the studied compounds, it was found that they have greater scores than the references with binding energy ≤ -8kcal/mol. The most important interactions such as hydrogen bonds, π-Alkyl, and electrostatic bonds, occurred between the ligand and the protein residues. Using the highest scores ≤ -8.5 kcal/mol, we chosed the top six compounds which are 2f, 2l, 2k, 2e, 2a, and 2b against the furin receptor respectively.
Compound 2f formed a hydrogen bond with amino acid ILE312, π-alkyl bonds with ARG268, VAL263, and ALA532, and finally π-orbital bonds, π-sulfur bond with TRP531. Furthermore, compound 2k was found to bind at the active site with TYR313 and GLN488 with two hydrogen bonds, two π-anion bonds with GLU271, four π-alkyl bonds with PRO266, VAL263, ARG268, and ILE12, alkyl bonds with ALA267, π-orbital bond with TRP531 and finally π-sulfur bond with TRP531. Interestingly, compound 2e and 2a formed hydrogen bonds with amino acid residues ILE312, GLY307, SER311, PRO266, ASP264, GLY265, GLN488, hydrophobic interactions with VAL263, TRP531, ALA267, ARG268, and ALA532, and electrostatic π-anion interactions with SER311, and GLU27. While compound 2b, showed hydrogen bonds with VAL263, π-alkyl bonds with ALA532, and ARG268, π-orbital bond withTRP531, and π-anion bond with GLU271. Consistent with our results, compound 2f and 2l are both the most potent against the furin receptor, followed by compounds 2e, 2a, and 2b.
The selected compounds could represent the starting point for a new class of inhibitors that may have advantages for the SARS-CoV-2 disease. The compound 2l has shown multi-target activity against 3CLMpro and Furin. Also, compound 2k was noticed to have an action toward two varieties of targeted protein 3CLMpro and ACE2. Finally, compound 2f has shown an inhibitory activity toward Furin and ACE2.
Figure 3: (a) 2D and (b) 3D interactions visualization of the 2l compound with Furin receptor.
3.1. Bibliographic study of catechin and its derivatives against plasmodium falciparum by In silico.
The In silico studies concerning the anti-malarial activity of catechin and its derivatives are much less than those concerning the anti-SARS-CoV-2 activity of these molecules. However, on three targets tested, a very good ligand-target affinity is observed for catechin derivatives (Table 4). These derivatives are natural compounds and it would be interesting to compare them to our semi-synthetic compounds.
Table 4: Study comparison carried out by In silico for catechin and its derivatives against anti-plasmodium.
Name of compound |
Binding Energy Kcal/mol |
PDB ID |
Targed Protein |
References |
(+)-catechin |
-7.40 |
3VI2 Chain A |
Plasmodium falciparum orotidine 5'-monophosphate decarboxylase (PfOMPDC) |
60 |
Epigallocatechin-3-gallate |
-18 |
1NHG |
Plasmodium falciparum enoyl-acyl carrier protein reductase (PfENR) |
61 |
Epicatechin-gallate |
-15.27 |
|||
Epigallocatechin |
-12.31 |
|||
Catechin-gallate |
-95.40 |
1CET |
Lactate dehydrogenase |
62 |
3.2. Docking Molecular for malaria.
Table 5: Interaction between the (+)-catechin derivates and the targeted receptor PfAMA1 (PDB ID: 3SRJ).
Ligands |
Receptor |
Docking score (kcal/mol) |
Hydrogen-Binding interaction |
Hydrophobic interaction |
Other interaction: Electrostatics and unfavorable bonds () |
Artesunate |
PfAMA1 (PDB ID: 3SRJ) |
-6,5 |
ASN205 LYS235 |
- |
- |
1 |
-6.6 |
GLY179 SER161 LYS177 ASN160 |
CYS275 |
(SER272) ASP178 |
|
2a |
-6,6 |
LYS235 ILE18 |
ARG219 LEU211 VAL208 |
ASP296 ARG219 |
|
2b |
-6.5 |
ASN205 ARG15 MET16 ARG219 LYS292 ASP296 ASN 223 |
VAL208 |
ARG219 |
|
2c |
-6,3 |
ASN205 ARG15 |
VAL208 LEU211 ARG219 |
(LYS235) ARG219 |
|
2d |
-6,4 |
MET16 ASN293 ASN223 |
VAL208 |
ASP296 ARG219 |
|
2e |
-6,7 |
PHE5 TYR175 LYS177 LEU8 |
ALA138 ALA3 PRO7 LEU8 |
- |
|
2f |
-7,3 |
TYR175 TYR142 MET273 |
PRO7ALA3 MET273 |
TYR142 |
|
2g |
-6,4 |
ARG15 MET16 ASN223 |
LEU221 ARG219 VAL208 |
ASP296 ARG219 |
|
2h |
-6,5 |
2MET16 |
VAL208 LEU211 ARG219 |
ASP296 ARG219 |
|
2i |
-6,3 |
ASN2052 LYS235 |
LEU221 ARG219 VAL208 |
ASP296 ARG219 (LYS235) |
|
2j |
-6,3 |
MET273 PHE5 GLY172 |
- |
LYS177 |
|
2k |
-6,8 |
ARG219 ILE18 ARG219 |
VAL208 |
ARG219 (MET16) |
|
2l |
-7,2 |
ASN205 ARG219 MET16 |
VAL208 LEU211 |
ASP296 ARG219 |
3.3. ADMET analysis of catechin derivates.
3.3.1. Prediction of ADME.
ADMET (Absorption, Distribution, Metabolism, Excretion, and Toxicity) is an important method in drug design and development processes. The drug-like properties including molecular weight (MW) (< 500), lipophilicity (< 4.15), Hydrogen Bond Acceptor (HBA) (< 10), Hydrogen Bond Donor (HBD) (< 5), Topological Polar Surface Area (TPSA) (<140 Å2), water-solubility (Log S), pharmacokinetics (gastrointestinal absorption, Blood–Brain Barrier, and Permeability) were calculated using Swiss ADME, and toxicity (mutagenic, tumorigenic, irritant, and reproductive effect) have been performed by using Protox online server 63.
The molecular weight of all (+)-catechin derivates aren’t in the range of drug-likeness properties (Mw< 500 g. mol-1), except (+)-catechin (1) with Mw = 290 g. mol-1. As shown in tables 6 and 7, among the 12 (+)-catechin derivatives, 1, 2b-c, 2e, and 2g-k compounds showed higher bioavailability (> 0.50) unlike to the remain compounds 2a, 2d, 2f, and 2l (Bioavailability score = 0.17) to generate main therapeutic agents against SARS-CoV-2 and/or anti-malarial (Table 6 and 7). The Log S number represents the drug’s water solubility. The solubility of all evaluated compounds ranged from -2.24 to -8.98 mol/L. Compounds 2a, 2c, 2e-f, and 2k-l showed low water solubility, and the other compounds 2b, 2d, 2g, and 2i-j have moderate solubility, unlike the base molecule which exhibits good solubility with Log S -2.24 mol/L (Table 6 and 7). According to ADMET characteristics, all catechin derivates 2a-l have a low rate of human GI absorption, whereas only (+)-catechin (1) have a high rate of human GI absorption and have a good rate drug gable properties (Table 7).
Table 6: Results of ADME and drug-likeness properties of (+)-catechin derivates.
Compounds |
Mw (g.mol-1) |
MLog P |
HBA |
HBD |
Rot N |
TPSA (A2) |
Solubility |
Log S (mol/l) |
Lipinski |
Veber |
Bioavailability |
1 |
290.27 |
0.24 |
6 |
5 |
1 |
110.38 |
Soluble |
-2.24 |
Yes: 0 violation |
Yes: 0 violation |
0.55 |
2a |
594.70 |
3.86 |
8 |
7 |
7 |
201.44 |
Poorly |
-6.64 |
No: 2 violation |
No: 1 violation |
0.17 |
2b |
564.67 |
3.30 |
8 |
5 |
7 |
186.76 |
Moderately |
-5.58 |
Yes: 1 violation |
No: 1 violation |
0.55 |
2c |
574.75 |
4.78 |
6 |
5 |
7 |
217.46 |
Poorly |
-6.62 |
Yes: 1 violation |
No: 1 violation |
0.55 |
2d |
540.65 |
3.17 |
6 |
7 |
7 |
192.56 |
Moderately |
-5.28 |
No: 2 violation |
No: 1 violation |
0.17 |
2e |
614.77 |
5.72 |
6 |
5 |
9 |
160.98 |
Poorly |
-7.87 |
Yes: 1 violation |
No: 1 violation |
0.55 |
2f |
674.87 |
6.66 |
5 |
6 |
7 |
217.46 |
Poorly |
-8.98 |
No: 2 violation |
No: 1 violation |
0.17 |
2g |
564.67 |
3.30 |
8 |
5 |
7 |
186.76 |
Moderately |
-5.58 |
Yes: 1 violation |
No: 1 violation |
0.55 |
2h |
568.70 |
3.05 |
6 |
5 |
7 |
170.84 |
Moderately |
-5.38 |
Yes: 1 violation |
No: 1 violation |
0.55 |
2i |
576.73 |
3.54 |
8 |
5 |
7 |
243.24 |
Moderately |
-5.81 |
Yes: 1 violation |
No: 1 violation |
0.55 |
2j |
544.60 |
2.35 |
10 |
5 |
7 |
213.04 |
Moderately |
-4.84 |
Yes: 1 violation |
No: 1 violation |
0.55 |
2k |
676.85 |
5.46 |
8 |
5 |
7 |
243.24 |
Poorly |
-8.28 |
Yes: 1 violation |
No: 1 violation |
0.55 |
2l |
640.77 |
5.08 |
6 |
7 |
7 |
192.56 |
Poorly |
7.64 |
No: 2 violation |
No: 1 violation |
0.17 |
Artesunate |
384.42 |
2.22 |
8 |
1 |
5 |
186.37 |
Soluble |
-3.08 |
Yes: 0 violation |
Yes |
0.56 |
Narlaprevir |
709.98 |
3.74 |
9 |
5 |
16 |
183.63 |
Poorly |
-6.86 |
No: 2 violation |
No: 2 violation |
0.17 |
Table 7: Continuation of table 6.
Compounds |
GI |
BBB |
Cyp1A2 |
Cyp2C19 |
Cyp2C9 |
Cyp2D6 |
Cyp3A4 |
Log Kp Skin permeation (cm/s) |
PAINS Alert |
1 |
High |
No |
No |
No |
No |
No |
No |
-7.82 |
1 Alert Catechol_A |
2a |
Low |
No |
No |
No |
No |
No |
Yes |
-6.38 |
1 Alert Catechol_A |
2b |
Low |
No |
No |
No |
No |
No |
Yes |
-7.22 |
1 Alert Catechol_A |
2c |
Low |
No |
No |
No |
Yes |
No |
Yes |
-6.16 |
1 Alert Catechol_A |
2d |
Low |
No |
No |
No |
No |
No |
Yes |
-7.22 |
1 Alert Catechol_A |
2e |
Low |
No |
No |
No |
No |
No |
Yes |
-5.09 |
1 Alert Catechol_A |
2f |
Low |
No |
No |
No |
No |
No |
Yes |
-4.87 |
1 Alert Catechol_A |
2g |
Low |
No |
No |
No |
No |
No |
Yes |
-7.22 |
1 Alert Catechol_A |
2h |
Low |
No |
No |
No |
No |
No |
Yes |
-7.45 |
1 Alert Catechol_A |
2i |
Low |
No |
No |
No |
Yes |
No |
Yes |
-7.09 |
1 Alert Catechol_A |
2j |
Low |
No |
No |
No |
No |
No |
Yes |
-7.77 |
1 Alert Catechol_A |
2k |
Low |
No |
No |
No |
No |
No |
Yes |
-5.68 |
1 Alert Catechol_A |
2l |
Low |
No |
No |
No |
No |
No |
Yes |
-7.64 |
1 Alert Catechol_A |
Artesunate |
High |
No |
No |
No |
No |
No |
No |
-7.31 |
0 Alert |
Narlaprevir |
Low |
No |
No |
No |
No |
No |
Yes |
-6.49 |
0 Alert |
MW: Molecular Weight, HBA: Hydrogen Bond Acceptor, HBD: Hydrogen Bond Donor, TPSA: Topological Polar Surface Area, MLogP = Lipophilicity, LogS:Water Solubility, GI: Gastrointestinal Absorption, BBB: Blood-Brain Barrier. |
3.3.2. Catechin and derivates toxicity.
The purpose of the toxicity study is to see if the catechin derivatives showed adverse drug effects, such as hepatotoxicity, carcinogenicity, mutagenicity and cytotoxicity. It is essential to highlight this series of compounds 1 and 2a-l as being candidates taking into account the different stages of pharmacokinetic studies carried out with the calculated ADMET parameters. Table 4 illustrates the results obtained using the ProTox II web server to assess the toxicological class of compounds. (+)-catechin showed no toxicity, the LD50 value was too high (10000 mg. Kg-1, class 6). This toxicological profile has been altered with the structural modification of the (+)-catechin molecular framework. All compounds were considered inactive in different toxicity categories, except compounds 2a, 2i, and 2k which showed toxicological activity in the liver (Hepatotoxicity). This study also allowed us to determine the theoretical LD50 value, which represents the dose at which 50% of the tested organisms die after exposure to the compound The LD50 value of compound 2a was 1190 mg. Kg-1 compared with other compound 2b-l (LD50 = 2500 mg. Kg-1). From this, it can be concluded that class 5 compounds with high LD50 values (= 2500 mg. Kg-1) are considered to be less toxic (Table 8).
Table 8: Toxicological properties of (+)-catechin derivates.
Ligands |
Hepatotoxicity |
Carcinogenicity |
Mutagenicity |
Cytotoxicity |
LD50 (mg.Kg-1) |
Class |
1 |
Inactive |
Inactive |
Inactive |
Inactive |
10000 |
6 |
2a |
Active |
Inactive |
Inactive |
Inactive |
1190 |
4 |
2b |
Inactive |
Inactive |
Inactive |
Inactive |
2500 |
5 |
2c |
Inactive |
Inactive |
Inactive |
Inactive |
2500 |
5 |
2d |
Inactive |
Inactive |
Inactive |
Inactive |
2500 |
5 |
2e |
Inactive |
Inactive |
Inactive |
Inactive |
2500 |
5 |
2f |
Inactive |
Inactive |
Inactive |
Inactive |
2500 |
5 |
2g |
Inactive |
Inactive |
Inactive |
Inactive |
2500 |
5 |
2h |
Inactive |
Inactive |
Inactive |
Inactive |
2500 |
5 |
2i |
Active |
Inactive |
Inactive |
Inactive |
2500 |
5 |
2j |
Inactive |
Inactive |
Inactive |
Inactive |
2500 |
5 |
2k |
Active |
Inactive |
Inactive |
Inactive |
2500 |
5 |
2l |
Inactive |
Inactive |
Inactive |
Inactive |
2500 |
5 |
3.4. Molecular Dynamic Simulation.
After the dynamics simulations, we analyzed the outcomes of each system by assessing the parameters (RMSD, RMSF, and interaction diagram) calculated by the MD trajectories. Figure 4 depicts the RMSD of the Cα protein and the RMSD of the ligand as a function of simulation time for both simulated systems. Both complexes 3SRJ-2f (Anti-plasmodium activity) and 6HZD-2l (Anti-SARS-CoV-2 activity) exhibited an average RMSD value of 1.5 and 2.4 Ǻ respectively all across the simulation.
The outcomes of the dynamic for the first complex 3SRJ-2f illustrated in figure 4A demonstrate that the Root Mean Square Deviation (RMSD) found equilibrium at 40 ns after an initial oscillation. The system then stayed stable throughout the MD simulation, showing that it progressed to a more perfect equilibrium state than the starting structure. While the RMSD plot for the second complex 6HZD-2l (Figure 4B) indicates a significant divergence during the first part of the experiment, but eventually stabilizes between 40 and 60 ns then fluctuate at 70 ns and system gets equilibrated after 75 ns.
Figure 4: RMSD plot for complexes (A) 3SRJ-2f and (B) 6HZD-2l.
We also investigated the Root Mean Square Fluctuation (RMSF) values of each backbone atom in both complexes to analyze how much a specific residue varies during the simulation 47, (Figure 5). The peaks on both graphics represent the portions of the protein that fluctuate the greatest during the simulation. For the first complex 3SRJ-2f (Figure 5A), it is shown that in all complexes, the variation occurred in the C-terminal residues of proteins, and in residue Ser272 (4.5 Ǻ), since they are positioned in the inactive regions of the protein, these residues are not engaged. In contrast, important active site residues such as Thy175, Lys177 have smaller RMSF fluctuations of less than 2 Ǻ, which could be related to the formation of more hydrogen-bonding interactions for greater ligand stability with the 3SRJ protein indicating that the combination is stable. The second complex 6HZD-2l, had an average RMSF of 1.2 Ǻ (Figure 5B). Moreover, most of the amino acid residues in the 6HZD-2l complex are smaller than 2 Ǻ in length. Overall, the RMSF figure 5 shows that there are no substantial changes in residual fluctuations when 2l is bound to 6HZD.
Figure 5: RMSF plot for complexes (A) 3SRJ-2f and (B) 6HZD-2l.
The interaction of 2l with 6HZD produced during the simulation (Figure 6B) revealed that compound 2l established multiple interactions with distinct substrates in the binding sites, including hydrogen bonds, hydrophobic interactions, ionic interactions, and water bridges. Pro266, Glu271, Asn310, Gln488, Trp531, and Ala532 were the amino acid residues implicated in the stability of the 6HZD-2l complex during the simulation, and they formed mainly two types of interactions: hydrogen bonds and water bridges. While the main observed interaction between 3SRJ and 2f (Figure 6A), are hydrogen bonds and hydrophobic interactions with Thyr175, Lys177, Ser272, Met273, and Phe5.
Figure 6: Protein ligands interactions for both complexes (A) 3SRJ-2f and (B) 6HZD-2l.
4. CONCLUSION
The result showed a clearly promising and encouraging track for the hemisynthesis of (+)-catechin for the anti-SARS-CoV-2 and anti-malarial activities. The theoretical simulation gave an orientation or a priority for certain compounds among the 12 catechin-aldehyde with (R, R) or (S, S) conformation, studied for the experimental suite of this project. Most structures presented a good affinity with the target protein responsible for SARS-CoV-2 and/or Plasmodium falciparum, compared to the references used Narlaprevir and Artesunate respectively. The evaluation of the toxicity of these compounds 2a-l revealed that the series didn’t show toxicity except for compounds 2a, 2i, and 2k which exhibit hepatotoxicity.
Compounds 2f and 2l are the best candidates for anti-malarial and anti-SARS-CoV-2 activity respectively among the series of compounds. Molecular dynamics also showed the good stability of complexes formed of 3SRJ-2f and 6HZD-2l via Ligand-protein intermolecular binding at 100 ns. Subsequently, the hemisynthesis of these two compounds 2f and 2l are the priority, followed by the rest of the catechin-aldehyde series.
Conflict of Interest
The authors declare that they have no conflict of interest.
Credit authorship contribution statement
Ahmed Said Mohamed and Samir Chtita: Conceptualization, Methodology, Supervision, Data curation, Writing – review & editing, Writing – original draft. Imane Yamari and Nouh Mounadi: Conceptualization, Writing – original draft. Abdirahman Elmi: Writing – original draft. Mohammed Bouachrine and Hanane Zaki: Visualization, Investigation, Validation. Ahmed Said Mohamed and Samir Chtita: Conceptualization, Methodology, Software, Supervision.
Acknowledgements
This research was funded by TWAS-UNESCO and Sida 21-029 RG/CHE/AF/AC_I 2021-2023. Furthermore, all the authors of the manuscript also thank and acknowledge their respective Universities and Institutes.
REFERENCES
1. Acharya PP, Genwali GR, Rajbhandari M. Isolation of Catechin from Acacia catechu Willdenow Estimation of Total Flavonoid Content in Camellia Sinensis Kuntze and Camellia Sinensis Kuntze Var. Assamica Collected from Different Geographical Region and Their Antioxidant Activities. Scientific World. 2013;11(11):32-36. https://doi.org/10.3126/sw.v11i11.8549
2. Elmi A, Spina R, Risler A, et al. Evaluation of Antioxidant and Antibacterial Activities, Cytotoxicity of Acacia seyal Del Bark Extracts and Isolated Compounds. Molecules. 2020;25(10):2392. https://doi.org/10.3390/molecules25102392 PMid:32455580 PMCid:PMC7288156
3. Grzesik M, Naparło K, Bartosz G, Sadowska-Bartosz I. Antioxidant properties of catechins: Comparison with other antioxidants. Food Chemistry. 2018;241:480-492. https://doi.org/10.1016/j.foodchem.2017.08.117 PMid:28958556
4. Higdon JV, Frei B. Tea Catechins and Polyphenols: Health Effects, Metabolism, and Antioxidant Functions. Critical Reviews in Food Science and Nutrition. 2003;43(1):89-143. https://doi.org/10.1080/10408690390826464 PMid:12587987
5. Nakao M, Takio S, Ono K. Alkyl peroxyl radical-scavenging activity of catechins. Phytochemistry. 1998;49(8):2379-2382. https://doi.org/10.1016/S0031-9422(98)00333-1 PMid:9887529
6. Shirakami Y, Shimizu M. Possible Mechanisms of Green Tea and Its Constituents against Cancer. Molecules. 2018;23(9):2284. https://doi.org/10.3390/molecules23092284 PMid:30205425 PMCid:PMC6225266
7. Zaveri NT. Green tea and its polyphenolic catechins: Medicinal uses in cancer and noncancer applications. Life Sciences. 2006;78(18):2073-2080. https://doi.org/10.1016/j.lfs.2005.12.006 PMid:16445946
8. Weyant MJ, Carothers AM, Dannenberg AJ, Bertagnolli MM. (+)-Catechin Inhibits Intestinal Tumor Formation and Suppresses Focal Adhesion Kinase Activation in the Min/+ Mouse1. Cancer Research. 2001;61(1):118-125.
9. Kemal T, Feyisa K, Bisrat D, Asres K. In Vivo Antimalarial Activity of the Leaf Extract of Osyris quadripartita Salzm. ex Decne and Its Major Compound (-) Catechin. Journal of Tropical Medicine. 2022;2022:e3391216. https://doi.org/10.1155/2022/3391216 PMid:36249737 PMCid:PMC9568338
10. Vyas VK, Bhati S, Patel S, Ghate M. Structure- and ligand-based drug design methods for the modeling of antimalarial agents: a review of updates from 2012 onwards. Journal of Biomolecular Structure and Dynamics. 2022;40(20):10481-10506. https://doi.org/10.1080/07391102.2021.1932598 PMid:34129805
11. Saleh Abu-Lafi, Mutaz Akkawi, Fuad Al-Rimawi, Qassem Abu-Remeleh, Pierre Lutgen. Morin, quercetin, catechin and quercitrin as novel natural antimalarial candidates. Pharm Pharmacol Int J. 2020;8(3):184-190. https://doi.org/10.15406/ppij.2020.08.00295
12. Moelyadi F, Utami PD, Dikman IM. Inhibitory Effect of Active Substances of Lollyfish (Holothuria atra) Against the Development of Plasmodium falciparum Based on In Silico Study. ILMU KELAUTAN: Indonesian Journal of Marine Sciences. 2020;25(4):135-142. https://doi.org/10.14710/ik.ijms.25.4.135-142
13. Abdulah R, Suradji EW, Subarnas A, et al. Catechin Isolated from Garcinia celebica Leaves Inhibit Plasmodium falciparum Growth through the Induction of Oxidative Stress. Pharmacogn Mag. 2017;13(Suppl 2):S301-S305. https://doi.org/10.4103/pm.pm_571_16 PMid:28808396 PMCid:PMC5538170
14. Chtita S, Fouedjou RT, Belaidi S, et al. In silico investigation of phytoconstituents from Cameroonian medicinal plants towards COVID-19 treatment. Struct Chem. 2022;33(5):1799-1813. https://doi.org/10.1007/s11224-022-01939-7 PMid:35505923 PMCid:PMC9051495
15. Jha RK, Khan RJ, Parthiban A, et al. Identifying the natural compound Catechin from tropical mangrove plants as a potential lead candidate against 3CLpro from SARS-CoV-2: An integrated in silico approach. Journal of Biomolecular Structure and Dynamics. 2022;40(24):13392-13411. https://doi.org/10.1080/07391102.2021.1988710 PMid:34644249
16. Ouassaf M, Belaidi S, Chtita S, Lanez T, Abul Qais F, Md Amiruddin H. Combined molecular docking and dynamics simulations studies of natural compounds as potent inhibitors against SARS-CoV-2 main protease. Journal of Biomolecular Structure and Dynamics. 2022;40(21):11264-11273. https://doi.org/10.1080/07391102.2021.1957712 PMid:34315340
17. Sannella AR, Messori L, Casini A, et al. Antimalarial properties of green tea. Biochemical and Biophysical Research Communications. 2007;353(1):177-181. https://doi.org/10.1016/j.bbrc.2006.12.005 PMid:17174271
18. Budiman I, Tjokropranoto R, Widowati W, Rahardja F, Maesaroh M, Fauziah N. Antioxidant and Anti-malarial Properties of Catechins. Journal of Advances in Medicine and Medical Research. Published online 2015:895-902. https://doi.org/10.9734/BJMMR/2015/11451 PMid:26099036
19. Cucinotta D, Vanelli M. WHO Declares COVID-19 a Pandemic. Acta Biomed. 2020;91(1):157-160. doi:10.23750/abm.v91i1.9397
20. Duzgun Z, Kural BV, Orem A, Yildiz I. In silico investigation of the interactions of certain drugs proposed for the treatment of Covid-19 with the paraoxonase-1. Journal of Biomolecular Structure and Dynamics. 2023;41(3):884-896. https://doi.org/10.1080/07391102.2021.2014971 PMid:34895069
21. de Oliveira OV, Cristina Andreazza Costa M, Marques da Costa R, Giordano Viegas R, Paluch AS, Miguel Castro Ferreira M. Traditional herbal compounds as candidates to inhibit the SARS-CoV-2 main protease: an in silico study. Journal of Biomolecular Structure and Dynamics. 2023;41(5):1603-1616. https://doi.org/10.1080/07391102.2021.2023646 PMid:36719113
22. Chtita S, Belhassan A, Bakhouch M, et al. QSAR study of unsymmetrical aromatic disulfides as potent avian SARS-CoV main protease inhibitors using quantum chemical descriptors and statistical methods. Chemometrics and Intelligent Laboratory Systems. 2021;210:104266. https://doi.org/10.1016/j.chemolab.2021.104266 PMid:33558778 PMCid:PMC7857023
23. Ngwa W, Kumar R, Thompson D, et al. Potential of Flavonoid-Inspired Phytomedicines against COVID-19. Molecules. 2020;25(11):2707. https://doi.org/10.3390/molecules25112707 PMid:32545268 PMCid:PMC7321405
24. Ansari WA, Khan MA, Rizvi F, et al. Computational Screening of Plant-Derived Natural Products against SARS-CoV-2 Variants. Future Pharmacology. 2022;2(4):558-578. https://doi.org/10.3390/futurepharmacol2040034
25. Diniz LRL, Elshabrawy HA, Souza MT de S, Duarte ABS, Datta S, de Sousa DP. Catechins: Therapeutic Perspectives in COVID-19-Associated Acute Kidney Injury. Molecules. 2021;26(19):5951. https://doi.org/10.3390/molecules26195951 PMid:34641495 PMCid:PMC8512361
26. Zhang Z, Zhang X, Bi K, et al. Potential protective mechanisms of green tea polyphenol EGCG against COVID-19. Trends in Food Science & Technology. 2021;114:11-24. https://doi.org/10.1016/j.tifs.2021.05.023 PMid:34054222 PMCid:PMC8146271
27. Mhatre S, Gurav N, Shah M, Patravale V. Entry-inhibitory role of catechins against SARS-CoV-2 and its UK variant. Computers in Biology and Medicine. 2021;135:104560. https://doi.org/10.1016/j.compbiomed.2021.104560 PMid:34147855 PMCid:PMC8189743
28. Shaik FB, Swarnalatha K, Mohan MC, et al. Novel antiviral effects of chloroquine, hydroxychloroquine, and green tea catechins against SARS CoV-2 main protease (Mpro) and 3C-like protease for COVID-19 treatment. Clinical Nutrition Open Science. 2022;42:62-72. https://doi.org/10.1016/j.nutos.2021.12.004 PMid:35106518 PMCid:PMC8795779
29. Andi B, Kumaran D, Kreitler DF, et al. Hepatitis C virus NS3/4A inhibitors and other drug-like compounds as covalent binders of SARS-CoV-2 main protease. Sci Rep. 2022;12(1):12197. https://doi.org/10.1038/s41598-022-15930-z PMid:35842458 PMCid:PMC9287821
30. Berman H, Henrick K, Nakamura H. Announcing the worldwide Protein Data Bank. Nat Struct Mol Biol. 2003;10(12):980-980. https://doi.org/10.1038/nsb1203-980 PMid:14634627
31. Van Lam van T, Ivanova T, Hardes K, et al. Design, Synthesis, and Characterization of Macrocyclic Inhibitors of the Proprotein Convertase Furin. ChemMedChem. 2019;14(6):673-685. https://doi.org/10.1002/cmdc.201800807 PMid:30680958
32. Lan J, Ge J, Yu J, et al. Structure of the SARS-CoV-2 spike receptor-binding domain bound to the ACE2 receptor. Nature. 2020;581(7807):215-220. https://doi.org/10.1038/s41586-020-2180-5 PMid:32225176
33. Vulliez-Le Normand B, Tonkin ML, Lamarque MH, et al. Structural and Functional Insights into the Malaria Parasite Moving Junction Complex. Phillips M, ed. PLoS Pathog. 2012;8(6):e1002755. https://doi.org/10.1371/journal.ppat.1002755 PMid:22737069 PMCid:PMC3380929
34. Kajiya K, Hojo H, Suzuki M, Nanjo F, Kumazawa S, Nakayama T. Relationship between Antibacterial Activity of (+)-Catechin Derivatives and Their Interaction with a Model Membrane. J Agric Food Chem. 2004;52(6):1514-1519. https://doi.org/10.1021/jf0350111 PMid:15030204
35. Trott O, Olson AJ. AutoDock Vina: improving the speed and accuracy of docking with a new scoring function, efficient optimization and multithreading. Journal of computational chemistry. 2010;31(2):455. https://doi.org/10.1002/jcc.21334 PMid:19499576 PMCid:PMC3041641
36. Trott O, Olson AJ. AutoDock Vina: Improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. Journal of Computational Chemistry. Published online 2009:NA-NA. https://doi.org/10.1002/jcc.21334 PMid:19499576 PMCid:PMC3041641
37. Peixoto HM, Marchesini PB, de Oliveira MRF. Efficacy and safety of artesunate-mefloquine therapy for treating uncomplicated Plasmodium falciparum malaria: systematic review and meta-analysis. Transactions of The Royal Society of Tropical Medicine and Hygiene. Published online December 29, 2016. https://doi.org/10.1093/trstmh/trw077 PMid:28039388
38. ADT / AutoDockTools - AutoDock. Accessed September 18, 2021. http://autodock.scripps.edu/resources/adt
39. Hanwell MD, Curtis DE, Lonie DC, Vandermeerschd T, Zurek E, Hutchison GR. Avogadro: an advanced semantic chemical editor, visualization, and analysis platform. Journal of cheminformatics. 2012;4(1). https://doi.org/10.1186/1758-2946-4-17 PMid:22889332 PMCid:PMC3542060
40. "Free Download: BIOVIA Discovery Studio Visualizer - Dassault Systèmes." https://discover.3ds.com/discovery-studio-visualizer-download (accessed Jun. 13, 2021).
41. Ioakimidis L, Thoukydidis L, Mirza A, Naeem S, Reynisson J. Benchmarking the Reliability of QikProp. Correlation between Experimental and Predicted Values. QSAR & Combinatorial Science. 2008;27(4):445-456. https://doi.org/10.1002/qsar.200730051
42. Banerjee P, Eckert AO, Schrey AK, Preissner R. ProTox-II: a webserver for the prediction of toxicity of chemicals. Nucleic Acids Research. 2018;46(W1):W257-W263. https://doi.org/10.1093/nar/gky318 PMid:29718510 PMCid:PMC6031011
43. Roos K, Wu C, Damm W, et al. OPLS3e: Extending Force Field Coverage for Drug-Like Small Molecules. Journal of Chemical Theory and Computation. 2019;15(3):1863-1874. https://doi.org/10.1021/acs.jctc.8b01026 PMid:30768902
44. System, Maestro-Desmond Interoperability Tools, Schrödinger, New York, NY, 2021.
45. Ouassaf M, Belaidi S, Mogren Al Mogren M, Chtita S, Ullah Khan S, Thet Htar T. Combined docking methods and molecular dynamics to identify effective antiviral 2, 5-diaminobenzophenonederivatives against SARS-CoV-2. Journal of King Saud University - Science. 2021;33(2):101352. https://doi.org/10.1016/j.jksus.2021.101352 PMid:33558797 PMCid:PMC7857992
46. Nour H, Daoui O, Abchir O, ElKhattabi S, Belaidi S, Chtita S. Combined computational approaches for developing new anti-Alzheimer drug candidates: 3D-QSAR, molecular docking and molecular dynamics studies of liquiritigenin derivatives. Heliyon. 2022;8(12):e11991. https://doi.org/10.1016/j.heliyon.2022.e11991 PMid:36544815 PMCid:PMC9761610
47. Singh MB, Sharma R, Kumar D, et al. An understanding of coronavirus and exploring the molecular dynamics simulations to find promising candidates against the Mpro of nCoV to combat the COVID-19: A systematic review. Journal of Infection and Public Health. 2022;15(11):1326-1349. https://doi.org/10.1016/j.jiph.2022.10.013 PMid:36288640 PMCid:PMC9579205
48. Parrinello M, Rahman A. Polymorphic transitions in single crystals: A new molecular dynamics method. Journal of Applied Physics. 1981;52(12):7182-7190. https://doi.org/10.1063/1.328693
49. Ghosh R, Chakraborty A, Biswas A, Chowdhuri S. Evaluation of green tea polyphenols as novel corona virus (SARS CoV-2) main protease (Mpro) inhibitors - an in silico docking and molecular dynamics simulation study. J Biomol Struct Dyn. Published online June 22, 2020:1-13. https://doi.org/10.1080/07391102.2020.1779818 PMid:32568613 PMCid:PMC7332865
50. Ohishi T, Hishiki T, Baig MS, et al. Epigallocatechin gallate (EGCG) attenuates severe acute respiratory coronavirus disease 2 (SARS-CoV-2) infection by blocking the interaction of SARS-CoV-2 spike protein receptor-binding domain to human angiotensin-converting enzyme 2. PLOS ONE. 2022;17(7):e0271112. https://doi.org/10.1371/journal.pone.0271112 PMid:35830431 PMCid:PMC9278780
51. Chourasia M, Koppula PR, Battu A, Ouseph MM, Singh AK. EGCG, a Green Tea Catechin, as a Potential Therapeutic Agent for Symptomatic and Asymptomatic SARS-CoV-2 Infection. Molecules. 2021;26(5):1200. https://doi.org/10.3390/molecules26051200 PMid:33668085 PMCid:PMC7956763
52. Frengki F, Putra DP, Wahyuni FS, Khambri D, Vanda H, Sofia V. POTENTIAL ANTIVIRAL OF CATECHINS AND THEIR DERIVATIVES TO INHIBIT SARS-COV-2 RECEPTORS OF M pro PROTEIN AND SPIKE GLYCOPROTEIN IN COVID-19 THROUGH THE IN SILICO APPROACH. Jurnal Kedokteran Hewan - Indonesian Journal of Veterinary Sciences. 2020;14(3). https://doi.org/10.21157/j.ked.hewan.v14i3.16652
53. Mishra CB, Pandey P, Sharma RD, et al. Identifying the natural polyphenol catechin as a multi-targeted agent against SARS-CoV-2 for the plausible therapy of COVID-19: an integrated computational approach. Briefings in Bioinformatics. 2021;22(2):1346-1360. https://doi.org/10.1093/bib/bbaa378 PMid:33386025 PMCid:PMC7799228
54. Kashyap P, Thakur M, Singh N, et al. In Silico Evaluation of Natural Flavonoids as a Potential Inhibitor of Coronavirus Disease. Molecules. 2022;27(19):6374. https://doi.org/10.3390/molecules27196374 PMid:36234910 PMCid:PMC9572657
55. Elmi A, Sayem SAJ, Ahmed M, Abdoul-Latif F. NATURAL COMPOUNDS FROM DJIBOUTIAN MEDICINAL PLANTS AS INHIBITORS OF COVID-19 BY IN SILICO INVESTIGATIONS. International Journal of Current Pharmaceutical Research. Published online July 15, 2020:52-57. https://doi.org/10.22159/ijcpr.2020v12i4.39051
56. Jena AB, Kanungo N, Nayak V, Chainy GBN, Dandapat J. Catechin and curcumin interact with S protein of SARS-CoV2 and ACE2 of human cell membrane: insights from computational studies. Sci Rep. 2021;11(1):2043. https://doi.org/10.1038/s41598-021-81462-7 PMid:33479401 PMCid:PMC7820253
57. Martinez-Archundia M, Colin-Astudillo B, Gómez-Hernández L, Abarca-Rojano E, Correa-Basurto J. Docking analysis provide structural insights to design novel ligands that target PKM2 and HDC8 with potential use for cancer therapy. Molecular Simulation. 2019;45(9):685-693. https://doi.org/10.1080/08927022.2019.1579326
58. Bai Y, Ye F, Feng Y, et al. Structural basis for the inhibition of the SARS-CoV-2 main protease by the anti-HCV drug narlaprevir. Signal Transduction and Targeted Therapy. 2021;6(1):51. https://doi.org/10.1038/s41392-021-00468-9 PMid:33542181 PMCid:PMC7860160
59. Du L, He Y, Zhou Y, Liu S, Zheng BJ, Jiang S. The spike protein of SARS-CoV - a target for vaccine and therapeutic development. Nat Rev Microbiol. 2009;7(3):226-236. https://doi.org/10.1038/nrmicro2090 PMid:19198616 PMCid:PMC2750777
60. Moelyadi F, Utami PD, Dikman IM. Inhibitory Effect of Active Substances of Lollyfish (Holothuria atra) Against the Development of Plasmodium falciparum Based on In Silico Study. Indo J Mar Sci. 2020;25(4):135-142. https://doi.org/10.14710/ik.ijms.25.4.135-142
61. Sharma SK, Parasuraman P, Kumar G, Surolia N, Surolia A. Green Tea Catechins Potentiate Triclosan Binding to Enoyl-ACP Reductase from Plasmodium falciparum (PfENR). J Med Chem. 2007;50(4):765-775. https://doi.org/10.1021/jm061154d PMid:17263522
62. Tegar M, Purnomo H. Tea Leaves Extracted as Anti-Malaria based on Molecular Docking PLANTS. Procedia Environmental Sciences. 2013;17:188-194. https://doi.org/10.1016/j.proenv.2013.02.028
63. Pokharkar O, Lakshmanan H, Zyryanov G, Tsurkan M. In Silico Evaluation of Antifungal Compounds from Marine Sponges against COVID-19-Associated Mucormycosis. Marine Drugs. 2022;20(3):215. https://doi.org/10.3390/md20030215 PMid:35323514 PMCid:PMC8950821