The main protease (Mpro) plays a crucial role in the replication of coronaviruses and has emerged as a prominent target for antiviral drug development (Pang et al., 2023). Within the 14 open reading frames (ORFs) of SARS-CoV-2, the ORF 1ab encodes two overlapping polyproteins (pp1a and pp1ab) that undergo precise cleavage events, leading to the generation of 16 non-structural proteins (nsp1-16) (Ullrich and Nitsche, 2020). Among the various viral proteases involved in this intricate process, Mpro holds a central position. Inhibiting the enzymatic activity of Mpro has proven to be a crucial therapeutic strategy in the fight against SARS-CoV-2. The use of heterocyclic compounds as templates for antiviral drug development has shown great promise, demonstrating potent activity against various viral infections, including hepatitis C (HCV), hepatitis B (HBV), human immunodeficiency virus (HIV), herpes simplex (HSV), rotavirus, adenoviruses, and coxsackie viruses (Liu et al., 2017; Kanwal et al., 2019). Notably, antiviral imidazole compounds have been reported to exert their activity by targeting viral proteases and other critical proteins that are essential to the viral life cycle (Kanwal et al., 2019). Hence, in this study, we employed computational techniques to identify potential SARS-CoV-2 inhibitors from a specific set of distinct imidazole derivatives.
The imidazole compounds examined in this study displayed strong binding affinities to the target proteins of SARS-CoV-2, indicating their potential as effective therapeutic agents against the virus. Imidazoles, owing to their structural resemblance to the histidine amino acid, have the ability to interact with crucial protein molecules, thereby influencing their functions (Kanwal et al., 2019). In this study, it was observed that the imidazolyl-methanones possess a higher binding affinity for all the target proteins than the other three classes of imidazoles. This could be linked to the imidazole rings, the methanone group, and several other aromatic rings contained in the structure of these compounds. Additionally, the spatial conformation and flexibility of Imidazolyl-methanones might be favorable for optimal binding to the target proteins. The distinct structure and adaptability of the Imidazolyl-methanone scaffold may enable improved accommodation and fitting within the Main protease's binding pocket, leading to enhanced interactions and a stronger binding affinity. Furthermore, the electronic and physicochemical properties of Imidazolyl-methanones may contribute to their superior binding affinity. Analysis of the protein-ligand interaction of the highest affinity compound, C10, revealed that all functional groups, including the aromatic rings, contribute to its binding to the target proteins. Aromatic interactions play a critical role in biological recognition, including protein-ligand interactions, as approximately 20% of amino acids possess aromatic properties (Babalola et al., 2021). Leveraging aromatic interactions is of great importance in drug design to enhance efficacy and optimize leads (Babalola et al., 2021). Overall, the results suggest that the imidazole derivatives, particularly the imidazolyl-methanones, hold promise as potential therapeutics against SARS-CoV-2. Their strong binding affinities, driven by their structural features and interactions, highlight their potential for further development and optimization in the fight against this global health threat.
The molecular docking analysis provided insights into the binding orientations of the compounds from the four imidazole classes with the best binding affinities compared to the standard ligands. Examination of the 3D and 2D structures of the docked complexes between the test compounds and SARS-CoV-2 target proteins confirmed their inhibitory potential. Specifically, compounds C1, C5, C10, and C14 from the thiophenyl-imidazole, pyridyl-imidazole, imidazolyl-methanone, and quinoline imidazole classes, respectively, interacted with specific amino acid residues within the binding pocket of SARS-CoV-2 Mpro. These interactions involved various types of chemical bonds and interactions, such as carbon-hydrogen interactions, π-sulfur interactions, π-alkyl interactions, π-anion interactions, π-π bond interactions, hydrogen interactions, and π-sigma bonds. In contrast, the standard ligand formed covalent bonds and conventional hydrogen bonds. Notably, interactions with CYS A:145 and HIS A:41 were observed for compounds C1, C5, and C14, which are amino acids crucial for catalysis at the catalytic site of the SARS-CoV-2 main protease (Kneller et al., 2020; Babalola et al., 2021). Dimerization and mutations in Mpro, leading to reduced enzymatic activity, are associated with interactions involving residues around GLU 288, ASP 289, and GLU 290 (Novak et al., 2022). However, the tested compounds did not show interactions with GLU 288 and ASP 289, suggesting that their mechanism of inhibiting SARS-CoV-2 replication is more likely through direct elimination of Mpro via CYS 145 and HIS 41 (Lan et al., 2020; Yan et al., 2020), rather than by inducing mutations or dimerization. Overall, the findings suggest that the tested imidazole compounds have the potential to inhibit SARS-CoV-2 replication by directly targeting the catalytic site of Mpro through interactions with specific amino acid residues. Further experimental studies are warranted to validate these findings and explore the therapeutic potential of these compounds as antiviral agents.
Top-docking pose of the selected hit molecule- C10 and Mpro were used in all-atom classical MD simulations using the Desmond package, developed by Schrodinger LLC. Molecular dynamics (MD) is a computational simulation method used to study the dynamic behavior of biomolecular systems over time. It involves modeling the interactions between individual atoms and molecules using physical laws and equations of motion.
The determination of root mean square deviation (RMSD) is a standard approach in computer-aided drug design to assess the structural changes of a macromolecule during molecular dynamics simulations (Sargsyan et al., 2017). In our study, we applied RMSD analysis to investigate the stability and movement of Imidazolyl methanone C10, the compound with the highest binding affinity, within the hydrated environment of the active pocket of SARS-CoV-2 Mpro throughout the simulation. Figure 8 illustrates the RMSD profiles of the protein, showing changes in the range of 1-2.25 Å. The protein demonstrates stability after approximately 3 ns, reaching an equilibrium state. Monitoring the protein's RMSD provides valuable insights into its conformational dynamics during the simulation (Monhemi et al., 2014). Fluctuations within the range of 1-3 Å are considered acceptable for small, globular proteins (Uttarkar and Niranjan, 2021). Larger changes indicate significant conformational transitions during the simulation. Based on our results, the observed changes in RMSD fall within the acceptable range, indicating a well-behaved system. The RMSD of C10 is consistently lower than that of the protein, with values ranging from 1 to 2.25 Å. Initially, the ligand remains stable within the protein until around 3.8 ns, after which it experiences fluctuations for a few nanoseconds before stabilizing again at approximately 4.4 ns. The ligand's RMSD provides insights into its stability within the protein's binding pocket. If the ligand's RMSD values are significantly larger than those of the protein, it suggests that the ligand has diffused away from its initial binding site (Durdagi et al., 2018). The observed stability of the ligand within the active pocket indicates that the complexes do not undergo substantial structural shifts, supporting the stability of these ligands at the protein's binding site. Overall, the RMSD analysis provides valuable information on the structural dynamics and stability of both the protein and the ligand, offering insights into their conformational behavior during the simulation. The results suggest that the studied complexes maintain a favorable interaction within the active pocket, enhancing their potential as promising drug candidates for SARS-CoV-2 inhibition.
Root mean square fluctuation (RMSF) analysis provides valuable insights into the flexibility of different regions within a macromolecule. Figure 9A illustrates the RMSF plot of Mpro, highlighting the regions that exhibit the highest fluctuations during the simulation. The peaks observed indicate that the N- and C-terminal tails of the protein undergo greater fluctuations compared to other parts. This observation is consistent with previous findings, where secondary structure elements such as alpha helices and beta strands tend to be more rigid and exhibit lower fluctuations compared to loop regions (Oum et al., 2020).
In Figure 10, a comprehensive analysis of secondary structure elements (SSE) in Mpro is presented, tracking their distribution and composition throughout the simulation in relation to the ligand. This plot provides a detailed representation of the SSEs across the protein structure, enabling the monitoring of each residue's SSE assignment and changes over time. By examining the SSE distribution, it becomes possible to assess the conformational stability and dynamic behavior of the protein-ligand complex. Furthermore, Figure 9B demonstrates the RMSF profile of the inhibitor imidazolyl-methanone C10, offering insights into the changes in ligand atom positions in relation to the protein. The ligand RMSF analysis allows for a characterization of the ligand fragments' interactions with the protein and their role in the binding event. In both Figures 9A and 9B, the protein and ligand exhibit varying levels of fluctuation. The protein's RMSF ranges from a minimum of 0.46 to a maximum of 2.0, while the ligand's RMSF ranges from a minimum of 1.5 to a maximum of 3.4. These results indicate that the ligand maintains flexibility within the ligand-protein complex throughout the simulation, suggesting favorable dynamics and a potential for efficient binding interactions. Overall, the RMSF analysis provides valuable information on the flexibility and dynamic behavior of the protein and ligand within the complex. The observed fluctuations in specific regions of the protein and ligand shed light on their conformational changes and their entropic contributions to the binding event. These findings contribute to our understanding of the dynamic nature of the protein-ligand complex and its implications for drug design and molecular interactions.
As mentioned earlier and depicted in Figure 5, the interaction between C10 and Mpro involves specific residues, including LEU 50, PHE 140, ASN 142, CYS 145, and MET 165. Further insights into the dynamic nature of this interaction are provided in Figure 11, which reveals that the total number of specific contacts between Mpro and the ligand throughout the trajectory is approximately 5. Notably, GLU 166 and other residues interact with the ligand consistently in each trajectory frame. Notably, residues such as LEU 50, PHE 140, ASN 142, CYS 145, and MET 165 establish multiple specific contacts with the ligand.
In addition, the ligand torsions plot offers a comprehensive overview of the conformational changes of each rotatable bond (RB) within C10 during the simulation trajectory spanning from 0.00 to 10.00 nsec. The bar plots in the plot summarize the torsional probability density, which represents the likelihood of observing specific torsional potential energy values within the dataset. The torsional potential energy values themselves reflect the energy required to rotate the rotatable bonds in the ligand. Higher energy values indicate increased strain or resistance to rotation, while lower energy values signify more favorable or stable conformations (Rai et al., 2019). Moreover, the probability density reveals the prevalence of certain energy ranges, indicating the frequency of sampling or stability of conformations with those energy values (Denning et al., 2011). By analyzing the torsion plot, valuable insights into the conformational dynamics and stability of the ligand can be obtained. The analysis of the histogram and torsion potential relationships provides valuable insights into the conformational strain experienced by the ligand in maintaining a protein-bound conformation. The torsional potential energy values offer a measure of the energy required to rotate the ligand's rotatable bonds, indicating the stability and strain associated with different conformations. In our study, the blue bond exhibited a torsional potential energy of 9.03 kcal/mol, with preferred conformations observed between 0°-90° and 135°-180°. These angles suggest favorable and stable orientations of the bond, indicating lower conformational strain. Conversely, the green bond displayed a higher torsional potential energy of 15.04 kcal/mol within the range of 0°-90°, indicating greater strain and resistance to rotation. On the other hand, the peach bond showed a torsional potential energy of 1.48 kcal/mol within the range of -180°-0°, indicating more favorable and stable conformations. Finally, the yellow bond exhibited a torsional potential energy of 11.09 kcal/mol between 0°-90°, suggesting higher strain and reduced stability. These findings highlight the diverse conformational dynamics of the ligand and provide important insights into its ability to adopt protein-bound conformations. The understanding of such conformational strain is crucial for designing and optimizing ligands with improved binding affinity and pharmacological properties.
In molecular dynamics (MD) simulations, several structural descriptors such as the radius of gyration (ROG), hydrogen bond analysis, solvent-accessible surface area (SASA), and polar surface area (PSA) play a crucial role in analyzing the behavior and properties of biomolecules.
The ROG provides valuable insights into the compactness of the system during the simulation, reflecting its performance in a biological context. It quantifies the "extendedness" of a ligand and is mathematically equivalent to its principal moment of inertia (Baby et al., 2023). As shown in Figure 13, the ROG spectrum demonstrates that the system maintains a consistent level of compactness throughout the simulation. The histogram's mode value indicates that the ROG of C10 remains stable at approximately 4.9 Å, with an initial period of stability observed between 4.4 Å and 4.8 Å at around 3.8 ns. This observation suggests that the Mpro protein exhibits a well-folded structure and is likely to engage in effective interaction mechanisms with the ligand hits.
In molecular modeling, the evaluation of a ligand's ability to bind to a target typically involves molecular docking, which is complemented by visual inspection to understand the nature of the interaction (Fischer et al., 2021). However, a limitation of molecular docking is its inability to consider the presence of water molecules within the active site. To address this, advanced modeling analyses, such as hydrogen bond computation after molecular dynamics (MD) simulation, are often employed to accurately determine the number of hydrogen bonds involved in ligand-protein interactions. In contrast to the results obtained from docking and visual inspection (Fig. 6), the analysis of hydrogen bonding reveals that imidazolyl-methanone C10 forms approximately fourteen hydrogen bonds with Mpro. This finding contradicts the absence of hydrogen bonds observed in the initial docking analysis. The presence of hydrogen bonds suggests a significant binding force that mediates the interaction between these compounds and Mpro. This observation further indicates that imidazolyl-methanone has the potential to effectively inhibit the target protein.
Molecular Surface Area (MolSA) serves as a quantitative measure of the total surface area of a molecule, encompassing both its accessible and inaccessible regions (Wu et al., 2020). By examining the SASA (Solvent-Accessible Surface Area) of the compound within the protein binding region, we observe a range between 370 Å and 390 Å. A reduction in MolSA implies a compacting effect, surface burial, or the closure of pockets (Gyebi et al., 2022). Conversely, a significant increase in MolSA may indicate structural expansion, surface exposure, or the opening of cavities or binding sites. These alterations in MolSA can be associated with various molecular phenomena such as flexibility, ligand binding, protein-protein interactions, or solvent interactions.
Considering that the drug-target binding process takes place within a dynamic fluid environment of a biological system, it is crucial to compute the Solvent-Accessible Surface Area (SASA) after conducting molecular dynamics simulations. This analysis helps assess the degree to which the simulated system accurately represents the intracellular milieu. SASA represents the surface area of a molecule that is accessible to solvent molecules and actively interacts with them (Chen and Panagiotopoulos, 2019). In this study, the mean SASA values for each complex were computed and plotted over the course of the simulation (Fig. 13). The calculated SASA values ranged from 240 Å to 340 Å, with a stable SASA observed around 300 Å at approximately 5 ns. These results indicate that the studied compounds exhibit a comparable degree of exposure to water, suggesting their potential for biological stability in contrast to the other examined complexes.
Polar Surface Area (PSA) is a parameter that quantifies the extent of the molecule's surface area occupied by polar atoms or polar groups. The analysis of the graph provides insights into the ability of C10 to engage in hydrogen bonding and interact with other polar molecules. Throughout the 10 ns simulation, the PSA values of the compounds range from 60 Å to 75 Å. In the field of drug discovery, PSA is frequently employed as a descriptor to predict a molecule's permeability and its capacity to traverse biological membranes. Higher PSA values generally indicate increased polarity and a greater potential for hydrogen bonding, both of which can influence the molecule's solubility and membrane permeability characteristics (Whitty et al., 2016).
In addition to their demonstrated inhibitory potentials towards the drug targets, the test compounds exhibit favorable ADMET properties of moderate magnitude. However, it is imperative to acknowledge that certain compounds may necessitate further optimization to enhance their overall ADMET profile while concurrently preserving or augmenting their binding affinity. ADMET analysis encompasses critical parameters encompassing absorption, distribution, metabolism, elimination, and toxicity, which collectively govern the pharmacokinetic behavior of a compound. This evaluation seeks to ascertain the compound's ability to be readily absorbed, reach the intended site of action, undergo biotransformation without compromising its activity, be efficiently eliminated from the organism, and avert the manifestation of adverse toxicological effects (Babalola et al., 2021). The ideal drug candidate is characterized by not only its efficacy against the therapeutic target but also its favorable ADMET properties within the therapeutic dose range (Guan et al., 2018). The recent emergence of computational models for in silico prediction of chemical ADMET properties has presented a significant advantage in drug discovery and development. These computational approaches offer a valuable tool for early identification of potential pharmacokinetic liabilities and drug failures, enabling informed decision-making during the drug design process prior to costly and time-consuming clinical trials (Babalola et al., 2021). Therefore, leveraging computational techniques for comprehensive evaluation of ADMET properties holds tremendous potential in the identification and optimization of promising drug candidates, facilitating the rational design of efficacious and safe therapeutic agents.
Lipophilicity, a crucial physicochemical parameter, serves as the pivotal link between solubility, membrane permeability, and, consequently, the processes of drug absorption, distribution, and clearance (Babalola et al., 2021). Lipophilic compounds exhibit lower solubility in aqueous environments but possess favorable solubility in oils and lipids, thereby facilitating their permeation across biological membranes. However, it is noteworthy that compounds with high lipophilicity (logP>5) often exhibit elevated metabolic turnover, reduced solubility, and poor oral absorption. Furthermore, highly lipophilic compounds have a heightened propensity to interact with hydrophobic targets other than the intended target, leading to an increased risk of promiscuity and toxicity (Gao et al., 2017). The lipophilicity of a compound is quantitatively characterized by its log P value, whereby higher log P values correspond to greater lipophilicity and lower water solubility (Daina et al., 2017). In our study, the test compounds demonstrated log P values ranging from 0.63 to 6.60 (C1 - C18), resulting in moderate IT Log Sw values indicative of their solubility characteristics (Table 2).
Drug-likeness assessment, a vital aspect of drug development, involves evaluating chemical structures and physicochemical properties to qualitatively determine the oral bioavailability of potential drug candidates (Babalola et al., 2021). The Lipinski filter, a widely utilized approach, enables the screening of compounds for oral drug-likeness based on factors such as molecular weight, hydrogen bond acceptors and donors, and lipophilicity (Lipinski et al., 1997). Lipinski's The rule outlines specific conditions that a compound must fulfill to be regarded as orally active. These conditions include having ≤5 H-bond donors, ≤10 H-bond acceptors, a molecular weight ≤500 g/mol, and a log P value less than 5.43. If a compound violates two or more of Lipinski's rules, it is classified as orally inactive (Lipinski et al., 1997). In our study, all test compounds exhibited zero Lipinski violations, except for C1, C2, C3, and C18, which had one violation (Table 2). Thus, with the exception of C18, all compounds are likely to possess oral activity, a conclusion further supported by their oral bioavailability scores. The quinoline-imidazole compound displayed a bioavailability score of 0.11, indicating approximately 11% probability of achieving at least 10% oral bioavailability in rats or measurable permeability in human colon carcinoma (Caco-2) cells. On the contrary, the remaining compounds displayed a bioavailability score of 0.55, implying a 55% chance of achieving at least 10% oral absorption in rats or human colon carcinoma absorptivity, according to Testa and Kraemer (2009). It is noteworthy that all the test compounds adhered to Veber's rule, with the exception of C18, which did not meet the criteria of having ≤10 rotatable bonds and a polar surface area (TPSA) ≤140 Å2, as proposed by Veber et al. (2002).
The pharmacokinetic predictions of the test compounds indicate that a significant number of these compounds possess inhibitory activity against several cytochrome P450 enzymes, namely CYP1A2, CYP2C19, CYP2C9, CYP2D6, and CYP3A4, with the exception of C18, which does not exhibit inhibitory effects. Notably, compounds C6, C8, C11, C13, C14, C15, and C17 demonstrate inhibitory activity against all of these cytochrome P450 enzymes (Table 3). Cytochrome P450 (CYP) enzymes are part of an isoenzyme superfamily that plays a vital role in various phase I drug metabolism processes (Hollenberg, 2002). Inhibiting the five major isoforms, namely CYP1A2, CYP2C19, CYP2C9, CYP2D6, and CYP3A4, can lead to drug-drug interactions related to pharmacokinetics since these enzymes are commonly involved in the metabolism of medications (Daina et al., 2017; Ursu et al., 2011). Understanding the inhibitory potential of compounds against these enzymes is essential for assessing their potential for drug-drug interactions and optimizing their pharmacokinetic properties.
With the exception of compounds C1, C6, C7, C8, and C11, all the test compounds chosen for this investigation are anticipated to act as substrates of P-glycoprotein (Pgp). P-glycoprotein is an ATP-binding cassette transporter responsible for actively transporting xenobiotics across biological membranes, which can lead to drug resistance and safeguard the body against foreign toxins (Babalola et al., 2021). The interaction of compounds with Pgp as substrates has implications for their pharmacokinetics, including absorption, distribution, metabolism, and excretion. Pgp is present in the intestines, where it can efflux drugs back into the intestinal lumen, reducing their absorption into the bloodstream (Lazarro et al., 2023). Consequently, compounds that are Pgp substrates may exhibit lower oral bioavailability. Moreover, Pgp is expressed in the blood-brain barrier and other tissues (Cox et al., 2023). Its activity can limit the penetration of Pgp substrate drugs into the brain and other target tissues, potentially impacting their efficacy in reaching specific sites of action. Additionally, Pgp's role in drug transport can indirectly influence drug metabolism by modulating drug concentrations in organs and tissues where metabolism occurs (Yoshitomo et al., 2023). Furthermore, Pgp is involved in drug excretion, particularly in the liver and kidneys, by actively pumping drugs out of hepatocytes and renal tubular cells, contributing to their elimination from the body (Danner et al., 2023). Overall, the interaction of drugs with Pgp can significantly influence their pharmacokinetics, altering drug absorption, distribution, metabolism, and excretion. Considering the compounds identified as Pgp substrates, it is crucial to take into account potential drug-drug interactions and determine appropriate dosing strategies.
While the toxicity prediction results indicated that compounds C2, C3, C6, C7, C8, C9, C10, C14, C15, C16, and C18 did not exhibit any tendencies towards toxicity based on the parameters tested, it is important to consider experimental validation of all compounds reported in the study. In vitro and in vivo studies, such as cell-based assays or animal models, offer the potential to obtain more precise and reliable information regarding their toxicity profiles. Experimental findings can validate or disprove the predicted toxicities, providing valuable insights into the actual risk associated with the compounds. Thus, it is recommended to conduct experimental studies to further investigate these compounds and explore their potential for development as novel drugs for the treatment of SARS-CoV-2.