Mostafa A.
Mansour
a,
Asmaa M.
AboulMagd
a and
Hamdy M.
Abdel-Rahman
*ab
aPharmaceutical Chemistry Department, Faculty of Pharmacy, Nahda University (NUB), Beni-Suef, Egypt. E-mail: hamdy.abdelrahman@nub.edu.eg
bMedicinal Chemistry Department, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
First published on 15th September 2020
The 2019 coronavirus (COVID-19) pandemic is spreading worldwide, with a dramatic increase in death without any effective therapeutic treatment available up to now. We previously reported quinazoline-trihydroxyphenyl Schiff base conjugates as phosphodiesterase 4B (PDE 4B) inhibitors (an enzyme that plays an essential role in the early stages of COVID-19 pneumonia). Additionally, the structural similarity between these conjugates and identified anti-severe acute respiratory syndrome (SARS)-coronavirus (CoV)-2 flavonoids inspired us to in silico study their possible binding interactions with essential SARS-CoV-2 proteins. Thus, this study provides an insight into the potential bindings between quinazoline-Schiff base conjugates and SARS-CoV-2 proteins, including spike glycoprotein (SGp), main protease (Mpro) and RNA-dependent RNA polymerase (RdRp), to offer an opportunity to find an effective therapy. Besides this, based on the role that COVID-19 plays in iron dysmetabolism, the conjugate trihydroxyphenyl moiety should be reconsidered as an iron chelator. Moreover, molecular dynamics simulations of quinazoline derivative Ic bound to the mentioned targets were carried out. Finally, ADMET calculations were performed for the studied compounds to predict their pharmacokinetic profiles.
![]() | ||
Fig. 1 Scheme of SARS-CoV-2 and some of its molecular protein targets.6 |
A recent literature survey highlighted that two possible viral pathophysiological mechanisms exist. The first one deals with interaction with the hemoglobin molecule, through receptors present on erythrocyte precursors, such as CD147 (also known as Basigin or EMMPRIN) or cluster of differentiation 26 (CD26), causing COVID-19. In contrast, the other mechanism is the induction of a ferroportin blockage through hepcidin-mimetic action of a viral spike protein.16,17 Regarding iron dysmetabolism in SARS-CoV-2, a noticeable similarity between the hepcidin protein and SARS-CoV-2 spike glycoprotein cytoplasmic tail amino acid sequence has been reported.18 SARS-CoV-2 has evolved to use a variety of host proteases, including cathepsin L, cathepsin B, trypsin, factor X, elastase, furin, and TMPRSS2 (transmembrane protease serine 2), to facilitate cell entry following receptor binding. It has been revealed that circulating Ang II levels are markedly increased in COVID-19 patients, providing a direct connection between tissue ACE2 down regulation with SARS-CoV-2 via blocking of the receptor binding domain (RBD) of the viral S-protein.19 Hepcidin is considered as the master regulator of iron metabolism, interacting with ferroportin to increase the iron entrance inside the cells.20 In terms of COVID-19, significant iron metabolism dysregulation may occur, accompanied by hyperferritinemia and ultimately ferroptosis.21 Whether the main pathological viral infection begins in the lungs, resulting in general anemic hypoxia or the opposite, iron dysmetabolism, is a matter of future research due to its role in multi-organ disease and hypoxia (Fig. 2).
![]() | ||
Fig. 2 Life cycle of coronaviruses under iron replete and deficiency conditions. (a) Sufficient intracellular iron levels support coronavirus replication, whereas (b) iron deficiency undermines its replication process by interfering with viral transcription, translation, assembly, and exocytosis. CoVs enter into host cells via binding to various receptors and disassemble to release the viral genome and nucleocapsid.21 |
Although the hallmarks of COVID-19 have been studied in detail, until now there has been no specific drug used in its treatment, with its control mainly being achieved using known antiviral drugs combined with supportive care.22 Thus, many research groups worldwide began searching for drugs and/or molecules for rapid COVID-19 disease therapy from libraries of natural, synthetic or semi-synthetic derivatives. The main strategies for these drug discovery projects involve repurposing of approved drugs or using computational approaches such as molecular docking, virtual screening, simulations, etc., which can save time, money and facilitate the discovery of COVID-19 drugs.23
Remdesivir 1 (Fig. 3) was the first drug granted emergency use for the treatment of suspected or laboratory confirmed COVID-19 cases by the U.S. Food and Drug Administration (FDA). It is an antiviral nucleoside analog that was developed for treatment of general ribonucleic acid (RNA) viral infections. It works by inhibiting viral replication through competitively inhibiting viral RNA polymerase.24
![]() | ||
Fig. 3 Remdesivir and reported polyphenolic compounds that are possible SARS-CoV-2 3CLpro inhibitors. |
Other research projects have focused on adjuvant therapies for the treatment of COVID-19, in particular iron chelation and phosphodiesterase 4 (PDE 4B) inhibitors. As previously mentioned, COVID-19 infections are characterized by iron dysmetabolism resulting in increasing intracellular iron, thus depriving iron supply to the virus represents a promising adjuvant therapy. For this reason, iron chelators such as deferoxamine or polyphenols have been screened for their effectiveness against SARS-CoV-2 viral infection.25 On the other hand, PDE4 inhibitors, which are one of the major PDE enzymes involved in cAMP-mediated regulation, play an important role in suppressing inflammatory cell functions that contribute to their anti-inflammatory actions in respiratory diseases such as chronic obstructive pulmonary disease (COPD) and asthma. Furthermore, the inhibition of PDE4 activity leads to smooth muscle relaxation and bronchodilation, which is useful for the treatment of asthma or COPD.26–29 Thus, selective PDE 4B inhibitors may be a promising option for treatment of the COVID-19 early hyperinflammatory state attributed to massive pro-inflammatory cytokine release.30
Likewise, many polyphenolic natural products such as flavonoids (2–6) have been in silico screened as a possible therapeutic treatment for COVID-19 disease as 3CLpro inhibitors (Fig. 3).31–33 In our previous study, three more phenolic compounds (7–9) were also screened against the SARS-CoV-2 main protease (Mpro) enzyme (Fig. 3). The structure–activity relationship analyses of the final top-hits presented in Fig. 3 reveal that they share similar binding modes in terms of hydrogen bonding and van der Waals interactions within the active site of Mpro.34
From all the above mentioned studies, we think that an effective protocol for the treatment of COVID-19 disease should include the following; (a) a drug that disrupts the life cycle of the virus at any stage of the viral replication cycle, such as blocking ACE2 receptors or inhibition of the SARS-CoV-2 main protease (Mpro); (b) an iron chelating agent to bind with the excess iron resulting from its dysmetabolism; (c) anti-inflammatory drugs as phosphodiesterase 4 inhibitors for the treatment of lung inflammation and necrosis.
We previously reported the design and synthesis of a class of 3H-quinazolin-4-one/Schiff base conjugates and evaluated them in terms of their PDE 4B activity.35 Based on the above findings, as the most active derivatives of this series were trihydroxyphenyl Schiff bases, we thought that these compounds would be ideal drugs for the treatment of COVID-19 (Fig. 4). Thus, herein we perform molecular modeling studies on the quinazolinone-Schiff base conjugates against three essential target proteins of the SARS-CoV-2 virus and predict their pharmacokinetics properties for rapid drug discovery.
![]() | ||
Fig. 4 Structures and rationale for the design of 2-phenylquinazolin-4(3H)one-trihydroxyphenyl Schiff base conjugates for COVID-19 therapy. |
Isomer | E isomer | Z isomer |
---|---|---|
a Total energy = EStr + EAng + EStb + EOop + ETor + EVdw + EEle. | ||
2D |
![]() |
![]() |
3D minimized |
![]() |
![]() |
Total energy | 117.266 | 149.139 |
E Str (bond stretch energy) | 8.122 | 7.595 |
E Ang (bond angle bend energy) | 21.979 | 42.065 |
E Stb (stretch-bend energy) | 1.966 | 2.621 |
E Oop (out-of-plane energy) | 0.295 | 0.417 |
E Tor (torsion energy) | 11.316 | 15.691 |
E Vdw (van der Waals energy) | 71.813 | 68.254 |
E Ele (electrostatics energy) | 1.774 | 12.497 |
Then, a mixture of the (un)substituted 2-phenyl-4H-benzo[d][1,3]oxazin-4-one intermediates (0.1 mol) and hydrazine hydrate 40% (0.1 mol) was refluxed in ethyl alcohol for 4 h. The reaction was monitored with thin layer chromatography (TLC), then poured on crushed ice, filtered, and washed many times with distilled water, dried in an oven (70–80 °C), and then recrystallized from ethanol (96%) to give a fragile solid mass of 3-amino-2-(un)substituted phenylquinazolin-4(3H)-one.
Finally, a mixture of the 3-amino-2-(un)substituted phenylquinazolin-4(3H)-one intermediates (0.001 mol) and 2,3,4-trihydroxybenzaldehyde (0.001 mol) was refluxed in absolute ethanol with a few drops of glacial acetic acid for 5 h. The reaction was monitored with TLC until completion of the reaction was reached. Then, the solution was cooled and the solid was precipitated, filtered off, dried, and recrystallized from ethanol.
At the end of the treatment period, assay plates were allowed to cool to room temperature for 10 min prior to the addition of 25 μL per well of Promega CellTiterGlo reagent, followed by an additional 10 min of incubation at room temperature. The resulting luminescence was quantitated using Molecular Devices Spectramax Paradigm apparatus. The percentage growth inhibition was calculated as follows, where 100% represents the viability of the vehicle treated control samples:
Thus, on average, the growth inhibition of the vehicle control samples equals zero.
The potency of compounds was determined using a non-linear dose response algorithm (4-parameter logistic fit) with the GraphPad Prism 5 software.
Also, online pkCSM pharmacokinetics were used to predict the toxicity of the molecules, including skin sensitization, hepatotoxicity, etc. The results obtained were analyzed and compared with the reference values of the pkCSM pharmacokinetics prediction properties.39
The crystal structure of the SARS-CoV-2 spike receptor-binding domain bound with ACE2 (PDB ID: 6M0J) contains two chains, where chain A represents the angiotensin-converting enzyme 2 structure of homo sapiens and chain E is the spike receptor binding domain.46
Recently, a library of ligands comprising 30 phytochemicals from medicinal plants was prepared for docking. The best compound, silybin (flavonolignan), scored −10.572 kcal mol−1 with a Silybin-S spike glycoprotein complex forming hydrogen bonds with residues His34, Arg403, Tyr453, Ser494 and Lys353 present in this pocket.47
Compound Ic exhibits a minimum binding energy −16.8866 kcal mol−1 by targeting His34 and Ser49 via hydrogen bonding, π–π and π–H interactions (Table 2). Furthermore, compound Ia shows significant binding interactions to SGp chain E; a spike glycoprotein that plays a vital role in attaching the virus to the human receptor ACE2 cell membrane. Thus, blockage of SGp reduces the binding of the virus to the host. Also, compound Ib exhibits hydrogen bonding and hydrophobic interactions with His34 and Arg403 amino acid residues in the same binding site as the ligand (Fig. 5). These interactions seem to inhibit the functions of SGp, thus achieving blockage of the SGp functional protein suppresses the activity of binding to the host. It is worth mentioning that ACE2 is responsible for altering the sensitivity of both the substrate and the inhibitor by binding to the cavity of chain B. Additionally, both π–alkyl and van der Waals interactions can reduce the binding capabilities of the virus, which enables target compounds with antioxidant activity and thus more inhibitory action. Compound Ic has a methoxy group that can be hydrolyzed inside the body, therefore its metabolite was redocked to predict its inhibitory activity. The results revealed that the metabolite reserve the initial π–π interactions with the His34 amino acid residue. Moreover, it binds via a hydrogen bond donor with Ala387, with a binding energy of −12.1711 kcal mol−1.
Compound | Crystal structure of SARS-CoV-2 spike receptor-binding domain bound with ACE2 (PDB ID: 6M0J) | ||||
---|---|---|---|---|---|
Affinity, kcal mol−1 | Distance (in Å) from main residue | Functional group | Interaction | ||
Ia | −15.8821 | 2.54 | His34 | Phenyl ring | Pi–cation |
2.14 | Arg403 | N of Schiff base | H–acceptor | ||
3.07 | Ala387 | Phenolic OH | H–donor | ||
Ib | −16.3852 | 2.82 | His34 | Phenolic OH | H–donor |
2.93 | Arg403 | Quinazoline ring | Pi–cation | ||
Ic | −16.8866 | 2.19 | His34 | Cyclic C![]() |
H–acceptor |
2.21 | His34 | N of Schiff base | H–acceptor | ||
3.86 | His34 | Phenyl ring | Pi–pi | ||
3.03 | Ser494 | Quinazoline ring | Pi–H | ||
Id | −16.3282 | 2.91 | His34 | Phenolic OH | H–donor |
2.71 | Arg403 | N of Schiff base | H–acceptor | ||
Ic metabolite | −12.1711 | 2.35 | His34 | Quinazoline ring | Pi–H |
2.24 | His34 | Quinazoline ring | Pi–cation | ||
3.02 | Ala387 | Phenolic OH | H–donor |
![]() | ||
Fig. 5 (a) A 2D animated structure showing the binding interactions between compound Ib and SGp and (b) a 3D representation showing the position of compound Ib within the cavity of SGp. |
Interestingly, the Schiff moiety formed interactions with His34 or Arg403 residues as a hydrogen bond acceptor and this may serve as a useful pharmacophoric feature for designing more active agents.
In computational drug discovery, MD is a technique that sheds light on the allosteric binding site of the protein, and conformation of the ligand–protein complex. Also, it can simulate the conditions that are hard to perform under in wet experiments. Herein, compound Ic was subjected to MD simulations bound to the SARS-CoV-2 spike receptor-binding domain bound with ACE2. It can be seen from Fig. 6 that compound Ic retained its binding affinity and firmly bound to its respective binding site. The root mean square deviation (RMSD) value produced in the compound Ic complex was stable at 0.6 nm.
Compound | Crystal structure of SARS-CoV-2 main protease in complex with Z1220452176 (PDB ID: 5R7Z) | ||||
---|---|---|---|---|---|
Affinity, kcal mol−1 | Distance (in Å) from main residue | Functional group | Interaction | ||
Ia | −13.3909 | 2.42 | Glu166 | Phenolic OH | H–donor |
2.66 | His41 | Phenyl ring | H–pi | ||
Ib | −12.7504 | 3.25 | Glu166 | Quinazoline ring | H–donor |
2.12 | Thr190 | Phenolic OH | Pi–H | ||
Ic | −13.5962 | 2.44 | Glu166 | Phenolic OH | H–donor |
2.89 | Glu166 | Phenolic OH | H–donor | ||
3.17 | Gln189 | Quinazoline ring | Pi–H | ||
3.04 | Gln189 | Quinazoline ring | Pi–H | ||
Id | −13.0212 | 2.83 | Glu166 | Phenolic OH | H–donor |
2.89 | Glu166 | Phenolic OH | H–donor | ||
2.99 | Gln189 | Phenyl ring | Pi–H | ||
Ic metabolite | −10.3634 | 2.21 | Glu166 | Phenolic OH | H–donor |
3.12 | Gln189 | Phenyl ring | Pi–H |
Furthermore, two hydrogen bond donations to the main chain of Glu166 and π–H interactions to the Gln 189 amino acid, with a binding energy of −13.5962 kcal mol−1 (Fig. 7), were calculated. Also, the other tested compounds showed comparable binding interactions to main amino acids; Glu 166, Gln 189, His 41 and Thr 190; which seemed to be useful in the inhibition of the enzyme. So, a tri-hydroxy moiety alongside a quinazoline scaffold may serve as excellent binding features to inhibit this enzyme. Regarding the metabolite analogue of derivative Ic, it reserves the same hydrophobic and hydrogen bonding interaction with the Glu166 and Gln 189 amino acid residues, respectively. In the MD simulation, compound Ic retained its binding affinity and still firmly bound to the respective binding site. The average RMSD value of the ligand relative to the initial structure was around 0.7 Å and increased up to 3.9 Å without losing the key interactions and staying stable for the rest of the MD simulations (Fig. 8).
![]() | ||
Fig. 7 (a) A 2D animated structure showing the binding interactions between compound Ic and Mpro and (b) a 3D representation showing the position of compound Ic within the cavity of Mpro. |
The active site containing these amino acids was chosen using the site finder properties of MOE 2020.0101. All the target compounds bind at the RdRp functional sites, with a high docking score and the interaction details and the binding energy of the four quinazoline derivatives Ia–d at the active site of RdRp are summarized in Table 4.
The results show that all of the compounds have excellent affinity to the selected pocket, according to the affinity in kcal mol−1. The synthesized derivative Ib is encapsulated in the receptor cavity with a binding energy score of −13.8649 kcal mol−1. The binding site is located between the NiRAN domain and β-hairpin structure that polymerizes the 3′ end,49 and therefore it may interfere with the polymerizing activity. The mentioned derivative binds firmly to amino acids residues Thr556, Arg553, Arg555 (Fig. 9). Also, compound Id forms a non-covalent interaction with the divalent cationic residue of Asp618. The potent synthesized derivative was docked at the active site between the NSP12-NSP7 residues forms conventional hydrogen bonds to Asp452, and van der Waals interactions with residues Tyr619 and Lys621, including π–alkyl/π–πcontacts with residue Cys622 located in the RdRp tunnel structure. Notably, that tri-hydroxy moiety plays a significant role in these interactions as it forms hydrogen bond interactions with the Thr556 and Asp452 amino acid residues. All these interactions between the trihydroxy moiety and RdRp may serve as a crucial pharmacophore for further development of more potent target derivatives. Moreover, the interactions of the Ic metabolite analogue were examined and it was shown that it forms π–cation interactions with Arg 553 and Arg 555. The distances and energy bindings are presented in Table 4.
![]() | ||
Fig. 9 (a) 2D animated structure showing the binding interactions between compound Ib and RdRp and (b) a 3D representation showing the position of compound Ib within the cavity of RdRp. |
Compound | SARS-Cov-2 RNA-dependent RNA polymerase (PDB ID: 6M71) | ||||
---|---|---|---|---|---|
Affinity, kcal mol−1 | Distance (in Å) from main residue | Functional group | Interaction | ||
Penciclovir | −14.5929 | 2.63 | Thr556 | –NH2 | H–donor |
2.71 | Thr556 | N of diazine | H–acceptor | ||
2.89 | Arg553 | Cyclic C![]() |
H–acceptor | ||
2.14 | Arg 555 | Cyclic C![]() |
H–acceptor | ||
Arg555 | N of Diazole | H–acceptor | |||
Ia | −14.0245 | 3.02 | Thr556 | Phenolic OH | H–donor |
2.99 | Thr556 | Phenolic OH | H–acceptor | ||
3.19 | Cys622 | Quinazoline ring | Pi–H | ||
Ib | −13.8649 | 2.96 | Thr556 | Phenolic OH | H–donor |
2.70 | Thr556 | Phenolic OH | H–donor | ||
2.98 | Arg553 | Cyclic C![]() |
H–acceptor | ||
2.98 | Arg555 | Cyclic C![]() |
H–acceptor | ||
Ic | −14.6145 | 2.93 | Cys622 | Cyclic C![]() |
H–acceptor |
2.94 | Asp452 | phenolic OH | H–donor | ||
3.09 | Lys621 | Quinazoline ring | Pi–H | ||
3.17 | Tyr619 | Quinazoline ring | Pi–H | ||
Id | −12.9652 | 2.81 | Thr556 | Phenolic OH | H–donor |
2.73 | Lys621 | Quinazoline ring | H–donor | ||
3.18 | Asp618 | –Cl | Halogen bond | ||
Ic metabolite | −14.5929 | 3.71 | Arg553 | –Phenyl ring | Pi–cation |
3.61 | Arg555 | –Phenyl ring | Pi–cation |
The proposed inhibitor Ic for SARS-CoV-2 identified as a result of molecular docking against RdRp was further investigated to determine its binding mode stability at the active site through MD simulations studies. The preliminary analysis performed on MD trajectory files was RMSD. The RMSD analysis was performed on the backbone atoms of the proteins (both in complex and free state) and average RMSD values obtained for ligand bound proteins (i.e. the inhibitor protein complex) were ∼0.8 Å, establishing their overall stability over the explored timescale, as shown in Fig. 10.
Property | Compound Ia | Compound Ib | Compound Ic | Compound Id | |
---|---|---|---|---|---|
Absorption | Water solubility (log mol L−1) | −3.602 | −3.515 | −3.955 | −3.603 |
Caco2 permeability (log![]() |
0.024 | 0.091 | 0.064 | 0.014 | |
Intestinal absorption (%) | 91.625 | 97.956 | 94.345 | 99.161 | |
Skin permeability (log![]() |
−2.735 | −2.735 | −2.735 | −2.735 | |
P-Glycoprotein substrate | Yes | Yes | Yes | Yes | |
P-Glycoprotein I | Yes | Yes | Yes | Yes | |
P-Glycoprotein II | Yes | Yes | Yes | Yes | |
Distribution | VDss (log L kg−1) | −1.021 | −0.92 | −1.179 | −0.831 |
Fraction unbound (Fu) | 0.087 | 0.137 | 0 | 0.14 | |
BBB permeability (log![]() |
−1.1 | −1.327 | −1.41 | −1.5 | |
CNS permeability (log![]() |
−2.339 | −3.14 | −3.371 | −3.068 | |
Metabolism | CYP2D6 substrate | No | No | No | No |
CYP3A4 substrate | Yes | Yes | Yes | Yes | |
CYP1A2 inhibitor | Yes | Yes | Yes | Yes | |
CYP2C19 inhibitor | Yes | Yes | Yes | Yes | |
CYP2C9 inhibitor | Yes | Yes | Yes | Yes | |
CYP2D6 inhibitor | Yes | Yes | No | Yes | |
CYP3A4 inhibitor | No | No | Yes | Yes | |
Excretion | Total clearance (log ml mim−1 kg−1) | 0.385 | 0.239 | 0.325 | −0.1 |
Renal OCT2 substrate | No | No | No | No | |
Toxicity | AMES toxicity | Yes | Yes | No | No |
Max. tolerated dose (log mg kg−1 day−1) | 0.44 | 0.492 | 0.343 | 0.495 | |
hERG I inhibitor | No | No | No | No | |
hERG II inhibitor | Yes | Yes | Yes | Yes | |
Oral rat acute toxicity (LD50) (mol kg−1) | 2.627 | 2.513 | 2.706 | 2.533 | |
Oral rat chronic toxicity (LOAEL) (mol kg−1 bw day−1) | 3.527 | 2.595 | 3.504 | 2.541 | |
Hepatotoxicity | Yes | Yes | Yes | Yes | |
Skin sensitisation | No | No | No | No | |
T. pyriformis toxicity (log μg L−1) | 0.287 | 0.286 | 0.286 | 0.286 | |
Minnow toxicity (log mM) | 0.155 | 0.002 | −0.98 | −0.515 |
Property | Compound Ia | Compound Ib | Compound Ic | Compound Id |
---|---|---|---|---|
log![]() |
3.06 | 3.20 | 3.07 | 3.85 |
Molecular weight | 373.37 | 391.36 | 403.39 | 425.80 |
NORTB | 3 | 3 | 4 | 3 |
H-bond acceptors | 7 | 7 | 8 | 7 |
H-bond donor | 3 | 3 | 3 | 3 |
No. of Lipinski's rule violations | 0 | 0 | 0 | 0 |
TPSA | 158.65 | 162.82 | 170.13 | 173.120 |
This journal is © The Royal Society of Chemistry 2020 |