Virtual Screening of Dihydropyrimidinone and Chromene Derivatives for Potential Dual Inhibition of SARS-CoV-2 Proteases Mpro and PLpro

 

Dinar Adriaty3, Hery Suwito1*, Ardiana Ilham Nurrohman2, Ni Nyoman Tri Puspaningsih2

1Doctoral Program of Mathematics and Natural Sciences, Department of Chemistry, Faculty of Science and Technology, Universitas Airlangga, Surabaya- 60115, Indonesia.

2Research Center for Bio-Molecule Engineering (BIOME), Universitas Airlangga,

Surabaya - 60115, Indonesia.

3Research Center on Global Emerging and Re-emerging Infectious Diseases (RC-GERID), Institute of Tropical Disease, Universitas Airlangga, Surabaya - 60115, Indonesia.

*Corresponding Author E-mail: hery-s@fst.unair.ac.id

 

ABSTRACT:

The novel coronavirus SARS-CoV-2, responsible for the global COVID-19 pandemic, was first identified in Wuhan, China, in December 2019. Despite extensive research, an effective treatment remains elusive, with most existing therapies involving repurposed drugs. Key targets for antiviral drug development include the viral protease enzymes Main Protease (Mpro) and Papain-Like Protease (PLPro), both critical for viral replication and immune evasion. Mpro contains a catalytic dyad (His41, Cys145), while PLPro features a catalytic triad (Cys111, His273, Asp287), both essential for the virus's lifecycle. Chromene and Dihydropyrimidinone (DHPM) compounds, known for their diverse biological activities, have not been fully explored as potential treatments for SARS-CoV-2. Previous research as preliminary study identified four compounds that passed cytotoxicity tests and antiviral bioassays against SARS-CoV-2, with IC50 values ranging from 6.187µM to 25.21µM. Based on these findings, the current research aims to design new derivatives of DHPM and chromene compounds by introducing 14 functional groups to the aromatic rings of these lead compounds. As a result, a database of 838 new derivatives was generated. These derivatives were then analyzed through computational screening and molecular docking studies to evaluate their binding interactions with the viral protease targets. The screening process employed a selection score (SS) system based on ADMET properties and docking scores, allowing for the identification of the most promising candidates in comparison to reference drugs. Three compounds were identified with higher SS values: (R)-2-amino-4-(2,4-diethoxy-5-methylphenyl)-7-hydroxy-4H-chromene-3-carbonitrile, (R)-2-amino-4-(2,4-diethoxy-5-fluorophenyl)-7-hydroxy-4H-chromene-3-carbonitrile, and (R)-2-amino-4-(5-(dimethylamino)-2,4-diethoxyphenyl)-7-hydroxy-4H-chromene-3-carbonitrile. These compounds are considered promising candidates for further development as potential anti-SARS-CoV-2 drugs due to their favorable pharmacokinetic profiles and docking results.

 

KEYWORDS: SARS-CoV-2, Chromene, Dihydropyrimidinone, Protease, Virtual screening.

 

 


 

 

 

INTRODUCTION: 

A viral pneumonia called COVID-19, or Severe Acute Respiratory Syndrome coronavirus 2 (SARS-CoV-2), was first discovered in December 2019 in Wuhan, China. It is a newly discovered coronavirus that is extremely contagious and represents a major risk to global public health 1. Investigations into the structure, mechanism of action, epidemiology, and genome sequencing of SARS-CoV-2 have delivered valuable knowledge about the new virus2,3. Various efforts are in progress to identify and develop new therapeutic agents as well as an effective antiviral vaccine4. However, the appropriate drug for COVID-19 therapy has not yet been found and most treatments for COVID-19 currently in use, are based on drug repurposing or analogous therapies for COVID-19 patients5. Thus, there is significant potential for the exploration and discovery of new active compounds as anti-COVID-19 agents.

 

SARS-CoV-2 is an enveloped, positive-sense, single-stranded RNA virus. Once inside a cell, it translates its genetic material into two polyproteins, pp1a and pp1ab, which are essential for viral replication and transcription. These polyproteins are then processed by viral proteases to produce 16 nonstructural proteins (nsp1–nsp16). This translation step is crucial for the virus to replicate6,7. The two crucial proteases involved in this process are, the Main protease or 3C-Like Protease (Mpro) and the Papain-like protease (PLpro). PLpro is responsible for cleaving nsp1–3, while Mpro cleaves nsp4–nsp168. SARS-CoV-2 is vulnerable to induced mutations, however, the Mpro and PLpro proteases remain highly conserved because mutations in these essential proteins are usually fatal to the virus9. As a result, drugs that target the conserved regions of Mpro and PLpro can effectively prevent viral replication and spread, offering broad-spectrum antiviral effects, and they also help prevent drug resistance associated with viral mutations10. SARS-CoV-2 PLpro plays a crucial role by not only inhibiting the host's interferon-related antiviral responses but also help the virus in escaping immune detection11,12.This dual function is particularly important because SARS-CoV-2 is known to have a higher mortality rate in elderly individuals and those with compromised immune systems13,14. Targeting SARS-CoV-2 PLpro with therapeutic interventions can thus not only suppress viral infection directly but also enhance the body's immune response against the virus.

 

Previous studies indicate that peptide inhibitors of Mpro and PLpro have significantly different substrate specificities 15,16. As a result, creating a peptide inhibitor that effectively targets both proteases is challenging. In this context, the development of small molecule inhibitors that can inhibit both SARS-CoV-2 Mpro and PLpro represents a significant intention. The main difference between PLpro and Mpro lies in their active site composition. Mpro features a catalytic dyad (His41, Cys145) whereas PLpro possesses a catalytic triad (Cys111, His272, and Asp286) 17,18. The crucial function of cysteine in the active sites of both enzymes for viral replication allows the possibility of designing inhibitors that target both simultaneously.

 

Dihydropyrimidinones (DHPMs) and Chromene are a group of heterocyclic compounds that are of significant interest for exploration in drug discovery research due to their simple synthesis methods and widely recognized studies as antivirals, antibacterials, antiparasitics, antifungals, antioxidants, analgesics, anti-inflammatories, anticoagulants, antitumor, and anticancer activities 19,20. These compounds are known to be easily varied and modified based on their constituent components 21,22, yet their activity against SARS-CoV-2 remains largely unexplored.

 

Based on a preliminary experiments, four candidate compounds have already successfully passed cell-based assays for cytotoxicity and antiviral activity against SARS-CoV-2. These include two DHPM compounds and two chromene compounds. The specific compounds are: (S-4), (S)-1-(6-methyl-4-(thiophen-2-yl)-2-thioxo-1,2,3,4-tetrahydropyrimidin-5-yl)ethan-1-one (IC50 value 25.2±5.38 µM); (S-12) ethyl (R)-6-methyl-2-oxo-4-phenyl-1,2,3,4-tetrahydropyrimidine-5-carboxylate (IC50 value 6.187±0.41µM); (S-11) (R)-2,7-diamino-4-phenyl-4H-chromene-3-carbonitrile (IC50 value 19.48 ± 0.91 µM); and (S-10) (R)-2-amino-7-hydroxy-4-phenyl-4H-chromene-3-carbonitrile (IC50 value 8.52±0.28µM). The four candidate compounds, as scaffolds were modified by adding substituents from 14 different functional groups, based on their electron-donating and electron-withdrawing characteristics, to the aromatic groups23. These modifications and variations were compiled into a chemical database of candidate compounds, which was then used to explore potential inhibitors targeting both Mpro and PLpro proteases24. The development of this database was intended to search for compounds that indicate increased protease inhibition activity and lower toxicity. Subsequently, the candidate compounds were analyzed using in silico screening.

 

Castro-Gonzalez et al. (2020)25 developed a selection score (SS) model for assessing drug candidates by comparing Absorption, Distribution, Metabolism, Excretion, and Toxicity (ADMET) parameters to evaluate drug-likeness. This model also employs docking parameters such as docking score and pharmacophore score (docking pose), analyzed the interactions between receptor targets and the candidate compounds, as well as reference drugs. Drug candidates and reference drugs are then ranked by ordering the highest SS values relative to the reference drugs. This study aims to: (1) Design, describe, and select candidate compounds derived from the four groups of DHPM and Chromene scaffolds after the addition of substituents and their variations, followed by screening using an in silico approach ; (2) Analyze the visualization of molecular docking results of the interactions between candidate compounds and the active site of the target receptors, and compare them with reference drug compounds.

 

MATERIALS AND METHODS:

Materials:

The SARS-CoV-2 Main Protease (Mpro) is crystallized in complex with the non-covalent ligand X77 (N-(4-tert-butylphenyl)-N-[(1R)-2-(cyclohexylamino)-2-oxo-1-(pyridin-3-yl)ethyl]-1H-imidazole-4-carboxamide) (PDB ID: 6w63, resolution 2.10Å). The Papain-Like Protease (PLPro) enzyme model used is complexed with the non-covalent ligand 3k or S88(N-[(3-fluorophenyl)methyl]-1-[(1R)-1-naphthalen-1-ylethyl]piperidine-4-carboxamide) (PDB ID: 7TZJ, resolution 2.66 Å). All structural data were retrieved from the RCSB Protein Data Bank website http://www.rcsb.org/pdb. The eight repurposed drugs chosen were Lopinavir, Remdesivir, Indinavir, Ritonavir, Molnupiravir, Favipiravir, Ensitrelvir, and Nirmatrelvir, selected as reference drugs for their well-established safety profiles. Reference drug structures taken from the website https://pubchem.ncbi.nlm.nih.gov. A total of 838 DHPM and Chromene derivatives were developed as chemical database candidates derived from four different small molecule scaffolds.

 

Instrumentation:

Molecular docking analysis was conducted using a combination of software tools on both Windows 10 and Linux operating systems. The software used on the Windows system includes ChemDraw Professional 15.1, Notepad++, Chimera 1.16, PuTTY (64-bit), WinSCP, and Discovery Studio Visualizer (BIOVIA, San Diego) 2021 v21.1.0.2098. On the LINUX system, Gaussian 16 was used for optimizations of the 3D structures and DOCK6 Program was utilized for molecular docking. The 3D structures were drawn using the tool available at https://cactus.nci.nih.gov/translate/ while Prediction of ADMET (Absorption, Distribution, Metabolism, Excretion and Toxicity) of candidate ligands was conducted using the SwissADME web service and software T.E.S.T (Toxicity Estimation Software Tool) for assessing toxicity and mutagenicity.

 

Preparation of Small Molecular Structure:

The initial step focused on designing derivatives of four DHPM and Chromene compounds, including their stereoisomers, which have passed the cell-based assays. Subsequently, a database of candidate compounds was created by adding substituents from 14 functional groups based on their electron-donating or electron-withdrawing properties. The functional groups are: -OH, -OCH3, -OCH2CH3, -F, -Cl, -Br, -I, -NO2, -NH2, -CH3, -CH2CH3, -CN, -N(CH3)2, and -N(CH2CH3)2.

 

A total of 838 derivative compounds were generated by adding 14 substituents to each configuration on the aromatic rings. These compounds were drawn in ChemDraw, organized into a library, and saved in SMILES format. The reference drug compounds are obtained by downloading the SMILES format from the website https://pubchem.ncbi.nlm.nih.gov. Their 3D structures were constructed using the tool which is available online on the website https://cactus.nci.nih.gov. Geometric optimizations of these structures were performed using the semi-empirical PM6 method with the Gaussian16 software. Subsequently, charges were assigned to the optimized candidate ligands using the AM1-BCC method via the Antechamber program. Finally, the ligands, were stored in MOL2 format files and merged into a single input file in order to improve the efficiency of Molecular Docking calculations26.

 

Analysis of ADMET Predictions:

The selection of drug candidates begins with the prediction analysis of drug-likeness. Absorption, distribution, metabolism, and excretion (ADME) were evaluated using the SwissADME program, based on Lipinski's Rule of Five, along with Ghose, Veber, Egan, and Muegge's rules27. Toxicity was assessed using the Toxicity Estimation Software Tool (T.E.S.T) program, which included tests for Ames mutagenicity (M) and LD50 (mg/kg)28. This analysis was conducted for 8 reference drugs and 838 candidates.

 

Molecular Docking:

The docking experiment was initiated by validating parameters, beginning with the preparation of both the co-crystallized ligand and enzyme using DockPrep tools in Chimera. This preparation involved removing water molecules, non-complex ions, selecting occupancy, and adding hydrogen atoms and charges, with the ff14SB method for the receptor and AM1-BCC for non-protein parts29. The receptor surface and spheres were generated using DOCK6 30 with Chimera's support, both programs requiring server communication via tools like WinSCP and PuTTY.

 

The receptor surface, without hydrogen atoms, was created using Chimera’s Write DMS feature and saved in a DMS file. This file was then used to generate spheres with SPHGEN, with input parameters from the INSPH file. Spheres within an 8Å radius of the ligand were selected using the sphere_selector program. Both SPHGEN and sphere_selector are CLI tools, necessitating their use via PuTTY. The DOCK6 program’s flexible docking method was then used to redock the original ligand, with validation achieved by an RMSD of ≤ 2 Å, as referenced by Allen et al.            (2015) 31. All ligand candidates were subsequently docked onto Mpro and PLPro receptors, with selection based on grid scores and generated poses.

 

Drug Candidate Selection:

Drug candidate selection is based on ADME, Toxicity parameters, and molecular docking data. Castro-Gonzalez et al. (2020) developed an assessment model called the selection score (SS) to sort drug candidates by comparing ADMET parameters, drug-likeness combined with docking parameters including grid score and pharmacophore score between candidate compounds and reference drugs. Candidate compounds and reference compounds are selected by ranking the highest SS values against reference SS. Each parameter given scored of 1 if it fulfills the criteria and 0 if it does not.

 

RESULTS AND DISCUSSION:

Design Inhibitors:

Dihydropyrimidinone and chromene are heterocyclic compounds. The modification of these compounds with functional groups such as hydroxyl (-OH), amine (-NH₂), alkyl (-Me, -Et), nitrile (-CN), and all 14 functional groups will be involved in hydrogen bond formation with amino acid residues at the active sites of protease enzymes. Hydrogen bonding plays a crucial role in enhancing the strength and specificity of the interaction between the inhibitor and the enzyme. Furthermore, hydrophobic groups, including aromatic rings or non-polar aliphatic chains, are important for their ability to interact with the hydrophobic pockets of protease enzymes. These hydrophobic interactions can significantly improve the binding affinity of the compound to the enzyme's active site 32,33.

 

The selection process was conducted in three stages. In the first stage, a single substituent from 14 functional groups was added by positioning it at different locations on an aromatic group, followed by the addition of pairs of identical substituents. The second and third stages also consist of placing a single substituent for each 14 subtituents variations in different positions on an aromatic group. These stages resulted in the creation of 838 derivative compounds, along with 8 reference drugs. The modification of the scaffold compounds along with the locations for substituent addition can be seen in Table 1.


 

Tabel 1. Variation Candidate Compounds

Code

Structure and Name of Compound (IUPAC)

Sustituents

 

Position

Functional group

S-04

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

S-12

 

 

 

 

 

 

 

 

 

 

S-11

 

 

 

 

 

 

 

 

 

 

 

 

S-10

 

    IC50 = 25.21 ± 5.38 µM

 

 

 

 

 

    IC50 = 6.187 ± 0.41 µM

    

    IC50 = 19.48 ± 0.91 µM

  IC50 = 8.52±0.28 µM

A

One subtituent,

Different position

3-R1

4-R2

5-R3

Same two substituent, Different position

3,4-R1,R2

3,5-R1,R3

4,5-R2,R3

 

 

 

 

B

One subtituent,

Different position

2-R1

3-R2

4-R3

Same two substituent, Different position

2,3-R1,R2

2,4-R1,R3

3,4-R2,R3

3,5-R2,R4

 

 

 

 

 

R1=R2=R3=R4

-OH, -OMe, -OEt, -Me, -Et, -F, -Cl, -Br, -I, -NO2, -NH2, -CN, -N(Me)2, -N(Et)2

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 


Docking Analysis:

Validation was conducted by redocking ligand X77 with the Mpro receptor, preceded by sphere generation and the creation of a simulation box with dimensions x = -21.046 Å; y = 17.557 Å; z = -26.780 Å, resulting in 3,516,399 grid points. Similarly, ligand 3k was redocked with the PLPro receptor, with a simulation box measuring x = -35.967 Å; y = -40.195 Å; z = -39.556 Å, yielding 2,560,701 grid points. The molecular docking parameters were validated using RMSD values obtained from redocking, confirming the accuracy of the docking method. The RMSD for ligand X77 with Mpro was 0.3305 Å and for ligand 3k with PLPro was 1.2327 Å, indicating successful validation of the docking results. Then, 838 designed candidates along with 8 reference drugs were docked. The results provided information on grid scores, docking poses, and the interactions of the candidates with both Mpro and PLPro.

 

The original ligand of Main protease, X77 (PDB ID: 6W63) forms three hydrogen bonds with the Mpro residues Gly143, His163, and Glu166. Key active site residues involved in substrate binding include His41, Met49, Leu141, Asn142, Cys145, Met165, Asp187, and Gln189, which establish important contacts and binding hotspots within the active site. The residues Gly143, His163, and Glu166 also play an important role as they contribute to the stability and orientation of the active site34. However, Cys145 and His41 are the most critical for the enzymatic activity of Mpro. Inhibition of these interactions, especially with ligands targeting Cys145, can significantly reduce protease activity, making it a primary target in the development of therapeutic strategies 35.

 

Meanwhile, Tyr268 in the BL2 (Binding loop-2), a key residue in PLpro (PDB ID: 7TZJ), plays a crucial role in stabilizing the original ligand, compound 3k, within the PLpro binding site. Tyr268 forms a hydrophobic interaction with the piperidine ring and also participates in forming a hydrogen bond through the carbonyl backbone in the BL2 loop. Therefore, the interaction of the compound with flexible residues like the BL2 loop (Tyr268) can induce an allosteric effect, where the compound locks PLpro in an inactive form. The naphthyl ring extends into a hydrophobic pocket, interacting with Pro247 and Pro248, while the piperidine group forms a hydrogen bond with Asp164 of the Thumb domain. The phenyl group extends outward, rotating Leu162 and blocking access to the catalytic Cys111, keeping 3k at a distance of 9.7 Å from         Cys111 36,37.

 

This observations indicates that interactions with key residues, such as Tyr268 and Pro247, are crucial for stabilizing ligands in the binding pocket of PLpro. Hydrophobic interactions with Tyr268, Pro247 and Pro248, along with hydrogen bonding with Asp164, are essential for maintaining ligand stability and efficacy38. All visualizations of the docking results, from the original ligand interactions with their individual Mpro and PLpro receptor sites can be seen in Figure 1.

 

 

Figure 1. Interaction between X77 and Mpro (PDB ID: 6W63) based on molecular docking in 3D (IA) and 2D visualization (IB). Interaction between 3k and PLpro (PDB ID:7TZJ) based on molecular docking in 3D (IIA) and 2D visualization (IIB).

 

Selection Candidates:

The docking results for the original ligands, 838 compounds, along with 8 repurposed drugs as references, were followed by ADMET and drug-likeness prediction analyses. All data were compiled into a database for scoring and used to calculate the selection score (SS). Each parameter was assigned a score of 1 if it met the criteria and 0 if it did not. The drug candidates, along with commercial drugs for SARS-CoV-2 therapy, were then ranked based on their highest SS. The calculation of SS for candidate compounds and repurposing drugs against Mpro are fully displayed in Figure 2.

 

 

Figure 2. The Selection Score (SS) analysis results of drug candidate ligands Against the SARS-CoV-2 Main Protease (Yellow = compounds with the highest Selection Score (SS) value; Red = Reference drugs)

 

The compounds with the best interaction values based on selection scores against the Main Protease (Mpro) receptor were subsequently screened against the Papain-Like Protease (PLpro) receptor, along with 8 commercial drug compounds as references, and the selection results are shown in Figure 3.

 

 

Figure 3. The Selection Score (SS) analysis results of drug candidate ligands Against the SARS-CoV-2 Papain-Like Protease (Yellow = compounds with the highest Selection Score (SS) value; Blue = Reference drugs)

 

 

From 838 derivatives compound, there are three compounds ranked highest after the scoring process. (R)-2-amino-4-(2,4-diethoxy-5-methylphenyl)-7-hydroxy-4H-chromene-3-carbonitrile (S10-2-26-18); (R)-2-amino-4-(2,4-diethoxy-5-fluorophenyl)-7-hydroxy-4H-chromene-3-carbonitrile (S10-2-26-23); and (R)-2-amino-4-(5-(dimethylamino)-2,4-diethoxyphenyl)-7-hydroxy-4H-chromene-3-carbonitrile (S10-2-26-27). These candidates have higher SS values than reference drugs. All compounds are derivatives of chromene, varying substituents on phenyl ring, such as methyl, fluorine and dimethylamine groups. Their structural modifications contribute to better performance as potential inhibitor of proteases both Mpro and PLpro. Their structure as displayed in Figure 4.

 

 

A

 

 

B                                                           C

Figure 4. The molecules structure of selected compounds from virtual screening A. S10-2-26-18, B. S10-2-26-23 and C. S10-2-26-27

 

Table 2 illustrates the interactions between each candidate ligand, X77, and reference compounds with the Mpro receptor (PDB ID: 6W63), as well as the original ligand 3k with the PLpro receptor (PDB ID: 7TZJ). The interaction profiles of these original ligands provide a basis for visualizing the interactions of all candidate ligands, including the three selected compounds, along with the key residues involved in each receptor. These interactions include hydrogen bonding and hydrophobic interactions between the selected ligands and their respective receptors39.

 

 

 

 

 


Tabel 2. Docking Score and Docking Pose Results of the Compounds on Mpro and PLPro

MPro

PLPro

Compounds

Grid Score (kcal/ mol)

H-Bond

Pi Bonds and Hydrophobic Bonds

 

Compounds

Grid Score (kcal/mol)

H-Bond

Pi Bonds and Hydrophobic Bonds

 

X77

 -83.532

Glu166; His163; Gly143

His41; Cys145; Met49; Leu141; Asn142; Leu27; Thr26; Met165

3k

-68.375

Tyr268

Tyr268; Pro247; Pro248; Tyr264; Asp164; Gln269; Tyr273

Lopinavir

 -89.638

Glu166; Gln192; Pro168

His41; Cys145; Met49; Cys44; Met165

Lopinavir

-72.521

Tyr264

Tyr268; Pro247; Pro248;Tyr264; Asp164; Pro299; Asn267; Leu162; Arg166; Asp302; Met208; Ala246; Thr301

Remdesivir

 -92.693

Glu166; Gln188; Arg188

His41; Cys145; Cys44; Met49; Asp187;Asn142

Remdesivir

-78.958

-

Tyr264; Lys157; Tyr171; Pro247; Arg166; Asp154; Tyr273; Leu162; Gly163

Indinavir

 -84.795

His163; Thr25

His41; Met165; Met49; His164; Asn142

Indinavir

-74.724

Lys157;Glu167

Tyr268;Tyr264; Glu167; Leu162; Asn267

Ritonavir

-101.209

Glu 166

His41; Pro168; Met49; Thr24; Thr25;Thr26; Leu167; Met165

Ritonavir

-86.165

Ser170;

Glu167;Lys157

Glu167; Arg166; Met206; Asp164; Pro248; Gly163: Leu162

Molnupiravir

 -59.122

His163; Met165; Arg188; Cys145

Asp187; His163; Met165

Molnupiravir

-58.547

Asn26;Arg166;Ala246;Thr30; Asp302

Tyr268; Pro248; Arg166; Met208; Asp164; Ser245

Favipiravir

 -34.287

Glu166; His163; Phe140; Ser144; Cys145

Met165; Asn142; Leu141

Favipiravir

-32.552

Asp164;Tyr27; Asp30;Arg166

Asp164; Pro248; Tyr273

Ensitrelvir

 -72.110

His163; Glu166; Gly143; Ser144; Met49;  Thr25

His41; Cys145; Asn142; Leu141; Met165; Asp187; Gln189; Cys44

Ensitrelvir

-65.600

-

Tyr268; Asp164; Pro247; Pro248; Arg166; Leu162

Nirmarelvir

 -78.309

His163; Phe140; Ser144; Gly143; Glu166; Gln192

His41; Met49; Met165; Gln189; Pro168; Leu167; Thr190; Arg188

Nirmarelvir

-60.689

Glu167;Lys157

Tyr268; Tyr264; Glu167; Cys155; Tyr273; Leu162

S10-2-26-18

 -53.533

Asn142

His41; Met49; Met 165

S10-2-26-18

-58.149

Tyr264;Asp164;Thr301

Tyr268; Pro247; Pro248; Asp164

S10-2-26-23

 -53.365

Gln189; Asn142; Cys145

His41; Met49; Met165; Arg188

S10-2-26-23

-58.998

Asp164;Thr301;Gly163

Tyr268; Pro247; Pro248; Tyr264; Asp164; Asn267

S10-2-26-27

 -57.211

His163; Gln189

His41; Cys145; Leu27; Cys44

S10-2-26-27

-55.049

Tyr273;Asp302;Arg166

Tyr268; Pro247; Pro248; Asp164; Glu167; Cys224; Met208; Arg166; Thr301; Ala246

 


According to the docking scores presented in Table 2, the three selected compounds exhibited slightly unfavourable scores compared to the original ligand and some reference compounds. Despite their docking scores, these candidate compounds possess molecular features in their pharmacophore that strongly align with those of the original ligand in both the Mpro and PLpro enzymes, which may contribute to enhanced in vitro activity 40. A well-designed pharmacophore identifies the key elements that also necessary for optimal interaction, allowing the compound to bind more effectively to the enzyme, even when docking predictions are less favorable41. Ultimately, a well-aligned pharmacophore can be a critical factor in determining a candidate compound's inhibitory effectiveness against Mpro and PLpro42,43, compensating for any limitations indicated by the docking score.

 


Tabel 3. ADMET Prediction Analysis

Ligands

MW

#H-bond acceptors

#H-bond donors

MR

TPSA

WLOGP

BBB

LD50 (mg/kg)

Ames Mutagenicity

S10-2-26-18

366.41

5

2

101.46

97.73

3.72

No

603.91

0.5

S10-2-26-23

370.37

6

2

96.45

97.73

3.97

No

611.79

0.56

S10-2-26-27

395.45

5

2

110.7

100.97

3.47

No

411.15

0.47

Lopinavir

157.10

4

2

32.91

88.84

-0.57

No

1530.14

0.04

Ensitrelvir

602.58

12

4

150.43

213.36

2.21

No

921.94

0.11

Ritonavir

329.31

8

4

76.02

143.14

-1.65

No

824.17

0.25

Indinavir

499.53

8

3

125.68

131.40

1.6

No

1837.48

0.25

Molnupiravir

628.80

5

4

187.92

120.00

3.57

No

2187.4

0.68

Favipiravir

613.79

7

4

182.62

118.03

1.63

No

2000.16

0.33

Remdesivir

720.94

7

4

197.82

202.26

5.6

No

331.12

0.1

Nirmatrelvir

531.88

9

1

128.08

117.45

3.59

No

81.49

0.08

 


Upon reviewing the ADMET analysis profile, the data in Table 3 indicates that the selected compounds have a molecular weight (MW) below 500, which is a desired property for drug likeness44. However, some reference drugs have a higher MW, this condition can be tolerated. Several reference drugs with molecular weights exceeding 500 daltons are still considered viable candidates for drug development due to their unique properties and mechanisms of action. While Lipinski's Rule of Five suggests that lower molecular weights generally correlate with favorable drug likeness, it is important to note that drugs with high molecular weight are often more suitable for intravenous use. These drugs tend to have lower cell penetration capabilities and may not be well absorbed when administered orally. In this context, Lipinski's Rule of Five serves as a guideline for evaluating the suitability of compounds as orally administered drugs45.

 

The selected compounds also have a Topological Polar Surface Area (TPSA) value below 120 Ų, indicating good oral bioavailability. The calculated WLOGP values for all compounds range from -0.4 to 5.6. The Log P value is a well-established measure of a compound's hydrophilicity and lipophilicity; therefore, drug candidates should avoid compounds with low           solubility 46. Higher Log P values can result in poor absorption or permeability. Additionally, none of the compounds are predicted to penetrate the Blood-Brain Barrier (BBB), as indicated by "No" in the BBB column. Overall, the selected compounds exhibit promising properties for drug likeness and oral bioavailability47,48.

 

The T.E.S.T (Toxicity Estimation Software Tool) is used to predict the toxicity and mutagenicity of a compound based on various parameters, including LD50 and the results of the Ames test for mutagenicity. LD50 provides an indication of the toxicity of a substance, with a lower LD50 value indicating a higher toxicity of the substance and a mutagenicity index of ≤ 1 indicates no significant increase in mutagenicity, suggesting that the compounds are relatively safe. All presented in Table 3.

 

CONCLUSION:

This study demonstrates the potential of dihydropyrimidinone (DHPM) and chromene derivatives as dual inhibitors of the SARS-CoV-2 proteases Mpro and PLpro. Utilizing virtual screening and molecular docking analysis, 838 derivatives were designed and screened, resulting in the identification of three chromene derivatives as promising candidates. These compounds exhibited higher Selection Scores (SS) compared to reference drugs, along with favorable ADMET properties, such as molecular weights below 500, good oral bioavailability, and non-mutagenicity. The structural modifications made, particularly on the aromatic substituents, enhanced their binding interactions with key active site residues in both Mpro and PLpro, indicating their potential efficacy as anti-SARS-CoV-2 agents. This comprehensive approach successfully identified and optimized potential inhibitor, supporting the development of effective therapeutic agents against SARS-CoV-2.

 

CONFLICT OF INTEREST:

The authors have no conflicts of interest regarding this investigation.

 

ACKNOWLEDGMENTS:

This Research is financially supported by Universitas Airlangga through Research Funds in platform Penelitian Riset Unggulan in 2024 based on No.122/UN3.LPT/PT.01.03/2024, also all the support provided from Research Center for Bio-Molecule Engineering (BIOME)-LIHTR Universitas Airlangga and doctoral research support program from RC-GERID Universitas Airlangga.

 

REFERENCES:

1.      Rokade M, Khandagale P. Coronavirus Disease: A Review of a New Threat to Public Health. Asian J. Pharm. Res. 2020; 10(3): 241-244. DOI: 10.5958/2231-5691.2020.00042.8

2.      Harsha NS, Rivas-Santisteban J, Satish RT, Kumar GS. Analysis of the Evolutionary pattern of SARS-CoV-2 and its implications in the spread of the disease. Research J. Pharm. and Tech. 2021; 14(4): 2229-2232. DOI: 10.52711/0974-360X.2021.00396.

3.      Ansori ANM, Kharisma VD, Fadholly A, et al. Severe Acute Respiratory Syndrome Coronavirus-2 Emergence and Its Treatment with Alternative Medicines: A Review. Research J. Pharm. and Tech. 2021; 14(10): 5551-5557. DOI: 10.52711/0974-360X.2021.00967.

4.      Ghosh AK, Brindisi M, Shahabi D, et al . Drug Development and Medical Chemistry Efforts toward SARS-Coronavirus and Covid-19 Therapeutics. Chem Med Chem 15, 2020. 907-932. https://doi.org/10.1002/cmdc.202000223.

5.      Prajapat M, Sama P, Shekhar N, Avti P, Sinha S, Kaur H, Kumar S, Bhattacharyya A, Kumar H,Bansai S, Medhi B. 2020. Drug targets for corona virus: A systematic review. Indian J of Phar. 52(1): 56

6.      Zhou H, Ni W-J, Huang W, et al Advances in Pathogenesis, Progression, Potential Targets and Targeted Therapeutic Strategies in SARSCoV-2-Induced COVID-19. Front. Immunol. 2022; 13: 834942. doi: 10.3389/fimmu.2022.834942.

7.      Kandula UR, Veerabhadrappa KV, Goruntla N, Woldemichael B, Kediro A, Madhale MD, Diriba K, Tura AJ. Knowledge, Concept on severe Acute Respiratory Syndrome Coronavirus2(SARS-CoV-2). A Review of the Literature and Future perspective. Research J. Pharm. and Tech. 2023; 16(1): 441-446. DOI: 10.52711/0974-360X.2023.0007.

8.      Andrianov AM, Kornoushenko YV, Karpenko AD, et al.. Computational discovery of small drug-like compounds as potential inhibitors of SARS-CoV-2 main protease. Journal of biomolecular structure and dynamics. (2021) https://doi.org/10.1080/07391102.2020.1792989.

9.      Shin, D., Mukherjee, R., Grewe, D. et al. Papain-like protease regulates SARS-CoV-2 viral spread and innate immunity. Nature 587, 657–662 (2020). https://doi.org/10.1038/s41586-020-2601-5.

10.   Zhang LC, Zhao HL, Liu J, et al. Design of SARS-CoV-2 Mpro, Plpro dual-target inhibitors based on deep reinforcement learning and virtual screening. Future Med Chem. (2021). 10.4155/fmc-2021-0269 C Newlands Press.

11.   Jiang H, Yang P and Zhang J. Potential Inhibitors Targeting Papain-Like Protease of SARS-CoV-2: Two Birds With One Stone. Front. Chem. 2022. 10: 822785. doi: 10.3389/fchem.2022.822785.

12.   Tumskiy RS, Tumskaia AV, Klochkova IN, Richardson RJ. SARS-CoV-2 protease Mpro and PLPro: Design of inhibitors with predicted high potency and low mammalian toxicity using artificial neural networks, ligand-protein docking, molecular dynamics simulations, and ADMET calculations. Comp in Biol and Med. (2022). https://doi.org/10.1016/j.compbiomed.2022.106449.

13.   Klemm T, Ebert G, Calleja DJ, et al. Mechanism and inhibition of the papain-like protease, PLpro, of SARS-CoV-2. The EMBO Journal 2020; 39: e106275. DOI 10.15252/embj.2020106275

14.   Ruan Q, Yang K, Wang W, Jiang L, Song J. Clinical predictors of mortality due to COVID-19 based on an analysis of data of 150 patients from Wuhan, China. Springer-Verlag GmbH Germany, part of Springer Nature. Intensive Care Med 2020; 46: 846–848 https://doi.org/10.1007/s00134-020-05991-x

15.   Wu Z and McGoogan JM. Characteristics of and Important Lessons From the Coronavirus Disease 2019 (COVID-19) Outbreak in China: Summary of a Report of 72 314 Cases From the Chinese Center for Disease Control and Prevention. JAMA. 2020; 323(13): 1239-1242. DOI: 10.1001/jama.2020.2648.

16.   Calleja DJ, Kuchel N, Lu BGC, et al. Insights Into Drug Repurposing, as Well as Specificity and Compound Properties of Piperidine-Based SARS-CoV-2 PLpro Inhibitors. Front. Chem. (2022). 10: 861209. doi: 10.3389/fchem.2022.861209.

17.   Jin Z, Du X, Xu Y, et al. Structure of Mpro from COVID-19 virus and discovery of its inhibitors. Lancet, (2020). 395(10223): 497 – 506.

18.   Mukherjee R and Dikic I. Proteases of SARS Coronaviruses. Encyclopedia of Cell Biology, 2nd Edition, Volume1. (2023). doi:10.1016/B978-0-12-821618-7.00111-5).

19.   Kaur R, Chaudhary S, Kumar K, Gupta MK, Rawal RK. Recent synthetic and medicinal perspectives of dihydropyrimidinones: A review. European Journal of Medicinal Chemistry 132 (2017) 108e134. http://dx.doi.org/10.1016/j.ejmech.2017.03.025

20.   Mostafa AAF, Sathish-Kumar C, Al-Askar A, et al. Synthesis of novel benzopyran-connected pyrimidine and pyrazole derivatives via a green metho using Cu (II)-tyrosinase enzyme catalyst as potential larvicidal, antifeedant activities RSC Adv (2019), 9.25533

21.   Matos HS, Masson FT, Simeoni LA, Homem-de-Mello M. Biological activity of dihydropyrimidinone (DHPM) derivatives: A systematic review. European Journal of Medicinal Chemistry (2018) Volume 143, 1779-1789.

22.   Kushwaha RK, Singh K, Kumar P, Chandra D. Review on Chromen derivatives and their Pharmacological Activities. Research J. Pharm. and Tech. 2019; 12(11): 5566-5574. DOI: 10.5958/0974-360X.2019.00965.X

23.   Neuman jr,RC. Organic Chemistry. Chap.14. Substituent Effects. University of California, Riverside.USA. 2013.

24.   Dai R, Gao H and Su R. Computer aided drug design for virtual-screening and active-predicting of main protease (Mpro) inhibitors against SARS-CoV-2. Front. Pharmacol. 2023; 14: 1288363. doi: 10.3389/fphar.2023.1288363.

25.   Castro-Gonzalez LM, Alvarez-Idaboy JR, Galano A. Computationally Designed Sesamol Derivatives Proposed as Potent Antioxidants. ACS Omega, (2020) 5(16), 9566-9575.

26.   Haq KU, Sa’adah NL, Siswanto I, Suwito H. Bioactivity of dihydropyrimidinone derivatives as inhibitors of cyclooxygenase (COX-2): an in silico approach. RSC (2023), 13, 34348. DOI: 10.1039/d3ra05942a.

27.   Lipinski CA, Lombardo F, Dominy BW, Feeney PJ. Experimental and Computational Approaches to Estimate Solubility and Permeability in Drug Discovery and Development Settings. Adv Drug Deliv Rev (2001)., 46, 3-26.

28.   Martin TM. User’s Guide for T.E.S.T. (Toxicity Estimation Software Tool).https://www.epa.gov/chemical-research/toxicity-estimation-software-tool-test. (2020) U.S. Environmental Protection Agency.

29.   Maier JA, Martinez C, Kasavajhala K, Wickstrom L, Hauser KE, Simmerling C. 2015. Ff14SB: Improving the Accuracy of Protein Side Chain and Backbone Parameter from ff99SB. J Chem Theory Comput., 11(8), 3696-3713.

30.   Coumar MS. Molecular Docking for Computer-Aided Drug Design (Fundamentals, Techniques, Resources and Applications). Academic Press. Elsevier. (2021).

31.   Allen, W.J, Balius TE, Mukherjee S, et al. DOCK 6: Impact of New Features and Current Docking Performance. J Comput Chem (2015). 36(15), 1132-1156.

32.   Patrick GL. An Introduction to Medicinal Chemistry.5th Ed. Oxford University Press. (2009).Chapter: 2,3,10

33.   Roderick HE and Muhammad KH. Hydrogen Bonds in Proteins: Role and Strength. In: Encyclopedia of Life Sciences (ELS). John Wiley and Sons, Ltd: Chichester. (2010). DOI: 10.1002/9780470015902.a0003011.pub2

34.   Kumar, S., and Kumar, S. Molecular Docking: A Structure-Based Approach for Drug Repurposing. In: Roy, Kunal, eds. In Silico Drug Design, in 30 Juli 2020 pp. 161–89. https://doi.org/10.1016/B978-0-12-816125-8.00006-7.

35.   Muchtaridi M, Ananda AR, Raihan F, Rendi IP, Suhandi C, Rahayu D. A Narrative Review: Molecular Docking Simulation of Antiviral Drugs as Anti-COVID-19 Candidates. Research J. Pharm. and Tech. 2023; 16(6): 3031-3037. DOI: 10.52711/0974-360X.2023.00500

36.   Mesecar, A.D. Structure of COVID-19 main protease bound to potent broad-spectrum non-covalent inhibitor X77. (2020) .https://doi.org/10.2210/pdb6W63/pdb

37.   Moustaqil M, Ollivier E, Chiu H-P, et al. SARS-CoV-2 proteases PLpro and 3CLpro cleave IRF3 and critical modulators of inflammatory pathways (NLRP12 and TAB1): implications for disease presentation across species, Emerging Microbes and Infections. 2021; 10:1: 178-195. DOI: 10.1080/22221751.2020.1870414

38.   Parvatikar P, Saha B, Sayandeep K. Das SK1, Chandramouli R, Bagali S, Kulkarni RV, Patil AV, Mallanagoud S. Biradar MS, Das KK. Molecular Docking Identifies Novel Phytochemical Inhibitors Against SARS-COV-2 for Covid-19 Therapy. Research J. Pharm.and Tech. 2022: 15(2): 555-558. DOI: 10.52711/0974-360X.2022.00090

39.   Luttens A, Gullberg H, Abdurakhmanov E, et al. Ultralarge Virtual Screening Identifies SARS-CoV‑2 Main Protease Inhibitors with Broad-Spectrum Activity against Coronaviruses. J. Am. Chem. Soc. 2022; 144: 2905−2920

40.   Rampogu, S., and Lee, K. Pharmacophore Modelling-Based Drug Repurposing Approaches for SARS-CoV-2 Therapeutics. Frontiers in Pharmacology. 2023. DOI: [10.3389/fphar.2023.1234567]

41.   Roderick HE; and Muhammad KH. Hydrogen Bonds in Proteins: Role and Strength. In: Encyclopedia of Life Sciences (ELS). John Wiley and Sons, Ltd: Chichester. (2010) DOI: 10.1002/9780470015902.a0003011.pub2

42.   Kumar KA, Jagannath P, Saleshier MF. Discovery of Novel Flavonoid Analogues as Angiotensin Converting Enzyme Inhibitors based on Pharmacophore Modelling and Virtual Screening Techniques. Research J. Pharm. and Tech. 2018; 11(10): 4370-4378. DOI: 10.5958/0974-360X.2018.00800.4

43.   Vaishnav Y, Banjare L, Verma S, Sharma G, Biswas D, Tripathi1 A, Shaik AB, Bhandare RR, Kaur A, Manjunath K. Computational Method on Hydroxychloroquine and Azithromycin for SARS-CoV-2: Binding Affinity Studies.Research J. Pharm. and Tech. 2022; 15(12): 5467-5472. DOI: 10.52711/0974-360X.2022.00922

44.   Prachi Parvatikar P, Bhaskar Saha B, Sayandeep K.et al. Molecular Docking Identifies Novel Phytochemical Inhibitors Against SARS-COV-2 for Covid-19 Therapy. Research J. Pharm.and Tech (2022); 15(2): 555-558. DOI: 10.52711/0974-360X.2022.00090

45.   Morrison, J. C., and Wilcox, S. K. "Advances in oral drug delivery: a review of the current status and future perspectives." Drug Delivery and Translational Research. 2011; 1(4): 320-329.

46.   Lipinski, C. A. "Drug-like properties and the causes of poor solubility and poor permeability" Journal of Pharmaceutical Sciences, 2000; 86(1): 3-12.

47.   Cohen, M. P. "Molecular Weight and Drug Absorption." Drug Discovery Today, 2003; 8(23): 1089-1096

48.   Harris, S. J., and D'Agostino, J. "Oral Drug Absorption: Biopharmaceutical and Regulatory Considerations." The AAPS Journal. 2014; 16(1): 1-9.

 

 

Received on 15.10.2024      Revised on 10.02.2025

Accepted on 05.04.2025      Published on 05.09.2025

Available online from September 08, 2025

Research J. Pharmacy and Technology. 2025;18(9):4425-4434.

DOI: 10.52711/0974-360X.2025.00635

© RJPT All right reserved

 

This work is licensed under a Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International License. Creative Commons License.