1Department of Biology, Faculty of Sciences, Sana`a University, Yemen.
2Department of Chemistry, Faculty of Sciences, Sana`a University, Yemen.
3Department of Pharmacy, Faculty of Medical Sciences, Thamar University, Yemen.
*Corresponding Author E-mail: a.hamid@su.edu.ye, m.almaqtari@su.edu.ye, alzomor@tu.edu.ye
ABSTRACT:
The aim of this study was to design and evaluate novel structural analogs of amlodipine that might have similar or higher antibacterial activity than the drug but fewer cardiovascular side effects. A number of computational and data retrieval techniques were used for the investigations in this study. After predicting the bacterial target of amlodipine, 85 structural analogs of the drug were designed and evaluated for their probability of antibacterial activity, calcium channel blocker activity, toxicity profiles, drug-likeness, and pharmacokinetics. Bacterial DNA topoisomerase I was found to be a potential target for amlodipine antibacterial activity, and thirteen analogs of the drug most likely acted on the same bacterial target as amlodipine. Of these analogs, only three had a low probability of acting as calcium channel blockers but an acceptable probability of having low toxicity and drug-likeness properties. However, only two of these analogs with a 1-butyl-4-hydropyridine core showed good probability of pharmacokinetics and are therefore promising as lead compounds for the discovery of new antibacterial drugs.
KEYWORDS: Amlodipine, Analogs, Chemoinformatics, Discovery, Lead Compounds, Antibacterial drugs.
INTRODUCTION:
One of the current significant approaches for the discovery of new antibacterial drugs and thereby solving the global problem of antimicrobial resistance is called "repurposing old drugs". This approach involves the investigation of the antibacterial activities of non-antibiotic drugs (NADs) which were initially approved for non-infectious diseases but can be repurposed as new antibacterial drugs1,2. Amlodipine {3-Ethyl 5-methyl (±)-2-[(2-aminoethoxy) methyl]-4-(o-chlorophenyl)-1,4-dihydro-6-methyl-3,5-pyridinedicarboxylate}3 is an oral calcium channel blocker (CCB) clinically used for treatment of hypertension and angina4.
It is one of these NADs that have been the concern of many studies that focused on the relevant approach. The drug was reported to possess in vitro activity alone and/or in combination with antibiotics against a wide-spectrum of pathogenic bacteria, such as Staphylococcus aureus, Vibrio cholerae, Vibrio parahemolyticus, Shigella spp., Salmonella spp., Bacillus spp, Listeria monocytogenes and multidrug-resistant Acinetobacter baumanii5-8. The main obstacle that hinders the repurposing of amlodipine to a new systemic antibacterial drug is its expected adverse actions on cardiovascular system due to its action as CCB. Therefore, the aim of this study was to design and evaluate novel structural analogs of amlodipine that might have similar or higher antibacterial activity than amlodipine but with diminished activity as CCBs.
Materials:
All materials were obtained from computational and data retrieval techniques including softwares, web servers and web databases as described later. The chemoinformatics work employed in this study was performed on a laptop "Toshiba/Japan, model: Tecra C660-A236 with Intel Arc A770 Limited Edition graphics card, Core 15 processor, and 16 GB RAM.
The prediction was performed using a machine learning web server followed by virtual molecular docking.
SuperPred 3 is a web server operated by the Institute of Physiology at Charité - Universitaetsmedizin Berlin, Germany9. It is a machine learning model that uses logistic regression and Morgan fingerprints of length 2048. The results of the prediction calculations for each molecule tested were the probability values (P %), for each potential target, that estimate the probability that the entered molecule binds to the targets, leading to the prediction of a corresponding therapeutic indication9, 10. In the present study, only the targets associated with the indication "bacterial infection" were selected. The probability and accuracy of model prediction for each selected target were at least 50% and 95%, respectively.
Virtual molecular docking of amlodipine to the bacterial targets predicted by "SuperPred 3" was performed not only to confirm the prediction of the web server, but also to select the most potent target among the targets predicted by the web server. In addition, the results of docking amlodipine to each predicted target protein were compared with those of a reference ligand for the target. Docking was performed using Molecular Operating Environment (MOE)11-13 (version 2022.02) s which is a software developed by Chemical Computing Group Inc. Canada. A series of procedures were performed prior to docking. First, the structures of the bacterial target proteins predicted by "SuperPred" were downloaded as "pdb" from the UniProt protein database14. Second, the structure of the ligand "amlodipine" was created in "smiles" format using ChemDraw® version 12, which is software developed by Cambridge soft corporation, USA 15, and then converted to "mol" format using “Open Babel” version 3.1.1 software16. Third, the binding site at the target protein was determined using the "site finder" tool of the software MOE, which ranks the potential binding sites according to their propensity for ligand binding (PLB) and uses a methodology based on alpha shapes, which are a generalization of the convex hulls developed in the software. Later, the structures of the ligand "amlodipine" and the protein were corrected and 3D-protonated using the "structure preparation" tool in the software. After these pre-docking procedures, the docking parameters were set to the standard accurate "rigid receptor" protocol in MOE and docking was initiated. Post-docking analysis performed by the software provided a number of data points. However, the final score (s) was the most meaningful. The score predicted the total binding energy in kcal/mol required for the best position of the molecule to bind to the binding site residues. Another parameter provided by the software was "rmsd_ refine," which gave the squared deviation between the molecular position before and after refinement. As recommended by the software, the molecule with the best pose (i.e., the pose with the greatest binding affinity to the receptor residues) was the one with the highest negative value (S), where rmsd_ refine ≤ 2. Post-docking analysis also provided a detailed description of the modes of protein-ligand interactions (PLIs), of which only those with a maximum distance of 4.5 Å were considered11-13. In addition, the protein-ligand complex was prepared using the software MOE and the 3D visualization of the complex was prepared using the software "UCSF Chimera, alpha version, 2023, developed by the Regents of the College of California17.
Eighty-five structural analogs of amlodipine were designed by substituting one or more of the pharmacophore moieties on the 1, 4-dihydropyridine nucleus of the drug. These chemical moieties, designated G1, G2, G3, G4, G5, and G6 as shown in Figure 1, were selected because they are associated with the drug's activity as a calcium channel blocker (CCB)18. Based on the numbering on the dihydropyridine nucleus, the relevant moieties included: H atom in conjunction with the N atom of the dihydropyridine ring, 2-[(2- aminoethoxy) methyl], 3-(ethyl carboxylate), 4-(o-chlorophenyl), 5-(methyl carboxylate), and 6-methyl. The groups on the designed structural analogs that substituted these moieties are demonstrated in (Table 1). The chemical structures of the analogs were drawn, checked for errors, and converted to "smiles" using ChemDraw® software as described earlier for amlodipine. The novelty of each designed analog was confirmed using search tools from three reliable chemical web databases, including the similarity search at “ChemSpider” of the Royal Society of Chemistry, UK19, SciFinder at the Chemical Abstract Service (CIS) of the American Chemical Society, USA20, and the search service at “PubChem” of the National Institutes of Health (NIH), USA21.
Figure 1: The chemical structure of amlodipine, showing the pharmacophore moieties (G1-G6) that were substituted in this study for designing the structural analogs
Table 1: Chemical moieties, at the structures of amlodipine structural analogs, substituted the pharmacophore moieties (G1-G6) of amlodipine.
Code |
G1 |
G2 |
G3 |
G4 |
G5 |
G6 |
D001 |
- H |
-Me-O-Et-NH2 |
-(C=O)H |
o-Cl-Phe- |
H(O=C)- |
Me- |
D002 |
- H |
-Me-O-Et-NH2 |
-COOH |
o-Cl-Phe- |
HOOC- |
Me- |
D003 |
- H |
-Me-O-Et-NH2 |
-(C=O)-Me |
o-Cl-Phe- |
Me-O(O=C)- |
Me- |
D004 |
- H |
-Me-O-Et-NH2 |
-(C=O)-Pr |
o-Cl-Phe- |
Pr-O(O=C)- |
Me- |
D005 |
-cycloPr |
-Me-O-Et-NH2 |
-(C=O)-O-Et |
o-Cl-Phe- |
Me-O(O=C)- |
Me- |
D006 |
-Phe |
-Me-O-Et-NH2 |
-(C=O)-O-Et |
o-Cl-Phe- |
Me-O(O=C)- |
Me- |
D007 |
-Pr |
-Me-O-Et-NH2 |
-(C=O)-O-Et |
o-Cl-Phe- |
Me-O(O=C)- |
Me- |
D008 |
- Me |
-Me-O-Et-NH2 |
-(C=O)-O-Et |
o-Cl-Phe- |
Me-O(O=C)- |
Me- |
D009 |
- H |
-Me-O-Et-NH2 |
-(C=O)-(CH-SH) –Et |
o-Cl-Phe- |
SH-Me(O=C)- |
Me- |
D010 |
- H |
-Me-O-Et-NH2 |
-(C=O)-S-Et |
o-Cl-Phe- |
Me-S(O=C)- |
Me- |
D011 |
-cycloPr |
-Me-O-Et-NH2 |
-COOH |
o-Cl-Phe- |
HOOC- |
Me- |
D012 |
-cycloPr |
-Me-O-Et-NH2 |
-(C=O)H |
o-Cl-Phe- |
H(O=C)- |
Me- |
D013 |
-cycloPr |
-Me-O-Et-NH2 |
-(C=O)-Me |
o-Cl-Phe- |
Me-O(O=C)- |
Me- |
D014 |
-cycloPr |
-Me-O-Et-NH2 |
-(C=O)-Pr |
o-Cl-Phe- |
Et(O=C)- |
Me- |
D015 |
-Me |
-Me-O-Et-NH2 |
-(C=O)H |
o-Cl-Phe- |
H(O=C)- |
Me- |
D016 |
-Me |
-Me-O-Et-NH2 |
-COOH |
o-Cl-Phe- |
HOOC- |
Me- |
D017 |
-Me |
-Me-O-Et-NH2 |
-(C=O)-Pr |
o-Cl-Phe- |
Et(O=C)- |
Me- |
D018 |
-Me |
-Me-O-Et-NH2 |
-(C=O)-Pr |
o-Cl-Phe- |
Me(O=C)- |
Me- |
D019 |
-cycloPr |
-Me-O-Et-NH2 |
-(C=O)-(CH-SH)-Et |
o-Cl-Phe- |
SH-Me(O=C)- |
Me- |
D020 |
-Me |
-Me-O-Et-NH2 |
-(C=O)-(CH-SH)-Et |
o-Cl-Phe- |
SH-Me(O=C)- |
Me- |
D021 |
-cycloPr |
-Me-O-Et-NH2 |
-(C=O) –S –Et |
o-Cl-Phe- |
Me-S(O=C)- |
Me- |
D022 |
-Me |
-Me-O-Et-NH2 |
-(C=O) –S –Et |
o-Cl-Phe- |
Me-S(O=C)- |
Me- |
D023 |
-Pr |
-Me-O-Et-NH2 |
-(C=O)-O-Me |
o-Cl-Phe- |
- Me-O(O=C)- |
Me- |
Table 1: continue
Code |
G1 |
G2 |
G3 |
G4 |
G5 |
G6 |
|||
D024 |
-Phe |
-Me-O-Et-NH2 |
-(C=O)H |
o-Cl-Phe- |
H(O=C)- |
Me- |
|||
D025 |
-Phe |
-Me-O-Et-NH2 |
-COOH |
o-Cl-Phe- |
HOOC- |
Me- |
|||
D026 |
-Phe |
-Me-O-Et-NH2 |
-(C=O)-O-Me |
o-Cl-Phe- |
- Me-O(O=C)- |
Me- |
|||
D027 |
-Pr |
-Me-O-Et-NH2 |
-(C=O)-Pr |
o-Cl-Phe- |
Et(O=C)- |
Me- |
|||
D028 |
-Pr |
-Me-O-Et-NH2 |
-(C=O)H |
o-Cl-Phe- |
H(O=C)- |
Me- |
|||
D029 |
-Pr |
-Me-O-Et-NH2 |
-COOH |
o-Cl-Phe- |
HOOC- |
Me- |
|||
D030 |
-Pr |
-Me-O-Et-NH2 |
-(C=O)-Pr |
o-Cl-Phe- |
Et(O=C)- |
Me- |
|||
D031 |
-Phe |
-Me-O-Et-NH2 |
-(C=O)-S-Et |
o-Cl-Phe- |
Me-S(O=C)- |
Me- |
|||
D032 |
-Pr |
-Me-O-Et-NH2 |
-(C=O) –(CH-SH)-Et |
o-Cl-Phe- |
SH-Me(O=C)- |
Me- |
|||
D033 |
-Pr |
-Me-O-Et-NH2 |
-(C=O)-S-Et |
o-Cl-Phe- |
Me-S(O=C)- |
Me- |
|||
D034 |
-Phe |
-Me-O-Et-NH2 |
-(C=O) –(CH-SH)-Et |
o-Cl-Phe- |
SH-Me(O=C)- |
Me- |
|||
D035 |
-H |
-Me-O-Et-NH2 |
-(C=O)H |
H- |
H(O=C)- |
Me- |
|||
D036 |
-H |
-Me-O-Et-NH2 |
-(C=O)-(CH-SH)-Et |
H- |
SH-Me(O=C)- |
Me- |
|||
D037 |
-H |
-Me-O-Et-NH2 |
-(C=O)-S-Et |
H- |
Me-S(O=C)- |
Me- |
|||
D038 |
-H |
-Me-O-Et-NH2 |
-(C=O)-O-Et |
H- |
Me-O(O=C)- |
Me- |
|||
D039 |
-Phe |
-Me-O-Et-NH2 |
-(C=O)-O-Et |
H- |
Me-O(O=C)- |
Me- |
|||
D040 |
-Me |
-Me-O-Et-NH2 |
-(C=O)-O-Et |
H- |
Me-O(O=C)- |
Me- |
|||
D041 |
-cycloPr |
-Me-O-Et-NH2 |
-(C=O)-O-Et |
H- |
Me-O(O=C)- |
Me- |
|||
D042 |
-H |
-Me-O-Et-NH2 |
-(C=O)-Pr |
H- |
Et(O=C)- |
Me- |
|||
D043 |
-H |
-Me-O-Et-NH2 |
-(C=O)-Me |
H- |
Me(O=C)- |
Me- |
|||
D044 |
-H |
-Me-O-Et-NH2 |
-COOH |
H- |
HOOC- |
Me- |
|||
D045 |
-Me |
-Me-O-Et-NH2 |
-(C=O)-S-Et |
H- |
Me-S(O=C)- |
Me- |
|||
D046 |
-Me |
-Me-O-Et-NH2 |
-COOH |
H- |
HOOC- |
Me- |
|||
D047 |
-Me |
-Me-O-Et-NH2 |
-(C=O)H |
H- |
H(O=C)- |
Me- |
|||
D048 |
-Me |
-Me-O-Et-NH2 |
-(C=O)-Me |
H- |
Me(O=C)- |
Me- |
|||
D049 |
-H |
-Me-O-Et-NH2 |
-(C=O)-Me |
H- |
H(O=C)- |
H- |
|||
D050 |
-Me |
-Me-O-Et-NH2 |
-(C=O)-(CH-SH)-Et |
H- |
SH-Me(O=C)- |
Me- |
|||
D051 |
-cycloPr |
-Me-O-Et-NH2 |
-(C=O)-(CH-SH)-Et |
H- |
SH-Me(O=C)- |
Me- |
|||
D052 |
-cycloPr |
-Me-O-Et-NH2 |
-(C=O)-Pr |
H- |
Et(O=C)- |
Me- |
|||
D053 |
-cycloPr |
-Me-O-Et-NH2 |
-COOH |
H- |
HOOC- |
Me- |
|||
D054 |
-cycloPr |
-Me-O-Et-NH2 |
-(C=O)-S-Et |
H- |
Me-S(O=C)- |
Me- |
|||
D055 |
-cycloPr |
-Me-O-Et-NH2 |
-(C=O)H |
H- |
H(O=C)- |
Me- |
|||
D056 |
-Me |
-Me-O-Et-NH2 |
-(C=O)-Pr |
H- |
Et(O=C)- |
Me- |
|||
D057 |
-cycloPr |
-Me-O-Et-NH2 |
-(C=O)-Me |
H- |
Me(O=C)- |
Me- |
|||
D058 |
-Phe |
-Me-O-Et-NH2 |
-(C=O)H |
H- |
H(O=C)- |
Me- |
|||
D059 |
-Phe |
-Me-O-Et-NH2 |
-COOH |
H- |
HOOC- |
Me- |
|||
D060 |
-Phe |
-Me-O-Et-NH2 |
-(C=O)-Me |
H- |
Me(O=C)- |
Me- |
|||
D061 |
-Phe |
-Me-O-Et-NH2 |
-(C=O)-Pr |
H- |
Et(O=C)- |
Me- |
|||
D062 |
-Pr |
-Me-O-Et-NH2 |
-(C=O)H |
H- |
H(O=C)- |
Me- |
|||
D063 |
-Pr |
-Me-O-Et-NH2 |
-COOH |
H- |
HOOC- |
Me- |
|||
D064 |
-Pr |
-Me-O-Et-NH2 |
-(C=O)-Me |
H- |
Me(O=C)- |
Me- |
|||
D065 |
-Pr |
-Me-O-Et-NH2 |
-(C=O)-Pr |
H- |
Et(O=C)- |
Me- |
|||
D066 |
-Phe |
-Me-O-Et-NH2 |
-(C=O)-(CH-SH)-Et |
H- |
SH-Me(O=C)- |
Me- |
|||
D067 |
-Phe |
-Me-O-Et-NH2 |
-(C=O)-S-Et |
H- |
Me-S(O=C)- |
Me- |
|||
D068 |
-Pr |
-Me-O-Et-NH2 |
-(C=O)-(CH-SH)-Et |
H- |
SH-Me(O=C)- |
Me- |
|||
D069 |
-Pr |
-Me-O-Et-NH2 |
-(C=O)-S-Et |
H- |
Me-S( O=C)- |
Me- |
|||
D070 |
-H |
-Me-O-Et-NH2 |
-(C=O)-O-Et |
m-Cl-Phe- |
Me-O(O=C)- |
Me- |
|||
D071 |
-H |
-Me-O-Et-NH2 |
-(C=O)-O-Et |
p-Cl-Phe- |
-Me-O(O=C)- |
Me- |
|||
D072 |
-Pr |
-Me-O-Et-NH2 |
-(C=O)-O-Et |
m-Cl-Phe- |
Me-OO=C)- |
Me- |
|||
D073 |
-Pr |
-Me-O-Et-NH2 |
-(C=O)-O-Et |
p-Cl-Phe- |
Me-O( O=C)- |
Me- |
|||
D074 |
-Bu |
-Me-O-Et-NH2 |
-(C=O)-O-Et |
o-Cl-Phe- |
Me-O(O=C)- |
Me- |
|||
D075 |
-Bu |
-Me-O-Et-NH2 |
-(C=O)-O-IsoPr |
o-Cl-Phe- |
Et-O-(C=O)- |
Me- |
|||
D076 |
-Bu |
-Me-O-Et-NH2 |
-COOH |
o-Cl-Phe- |
HOOC- |
Me- |
|||
D077 |
-Bu |
-Me-O-Et-NH2 |
-(C=O)-O-IsoPr |
o-Cl-Phe- |
Et-O-(C=O)- |
H- |
|||
D078 |
-IsoPr |
-Me-O-Et-NH2 |
-(C=O)-O-Et |
o-Cl-Phe- |
Me-O(O=C)- |
Me- |
|||
D079 |
-IsoBu |
-Me-O-Et-NH2 |
-(C=O)-O-Et |
o-Cl-Phe- |
Me-O(O=C)- |
Me- |
|||
D080 |
-(C=O)H |
-Me-O-Et-NH2 |
-(C=O)-O-Et |
o-Cl-Phe- |
Me-O(O=C)- |
Me- |
|||
D081 |
-NH2 |
-Me-O-Et-NH2 |
-(C=O)-O-Et |
o-Cl-Phe- |
Me-O(O=C)- |
Me- |
|||
D082 |
-CH2-COOH |
-Me-O-Et-NH2 |
-(C=O)-O-Et |
o-Cl-Phe- |
Me-O(O=C)- |
Me- |
|||
D083 |
-CH2-(C=O) CH3 |
-Me-O-Et-NH2 |
-(C=O)-O-Et |
o-Cl-Phe- |
Me-O(O=C)- |
Me- |
|||
D084 |
-IsoPe |
-Me-O-Et-NH2 |
-(C=O)-O-Et |
o-Cl-Phe- |
Me-O(O=C)- |
Me- |
|||
D085 |
-cycloHe |
-Me-O-Et-NH2 |
-(C=O)-O-Et |
o-Cl-Phe- |
Me-O(O=C)- |
Me- |
|||
o-Cl-Phe: o-chlorophenyl; m-Cl-Phe: m-chlorophenyl; p-Cl-Phe: p-chlorophenyl; Bu: butyl; cycloHe: Cyclohexyl; cycloPr: cyclopropyl; Et: ethyl; IsoPe: Isopentyl; IsoPr: isopropyl; Me: methyl; Phe: phenyl; Pr: propyl
a) Antibacterial activity:
After predicting the most likely bacterial target of amlodipine, the prediction of the antibacterial activity of each designed structural analog was performed in the same manner applied for prediction of the bacterial target of amlodipine. Using SuperPred 3, the structural analogs that had a higher prediction probability for the bacterial target of the drug were selected. Molecular docking of these selected structural analogs to the bacterial target was then performed, and those that yielded a more negative endpoint score for docking than amlodipine were selected for further investigation.
The selected structural analogs were docked to calcium ion transport protein (CaVAb) using the software MOE. This protein has been reported in the literature as a target of dihydropyridines22, 23, on which they exert their activity as CCBs with high affinity for amlodipine23. The structure of CaVAb in complex with amlodipine (encoded as 5KMD) was downloaded in "pdb" format from the web database protein data bank" (PDB) 24. The protein structure was freed from the drug using the software MOE, and the pre-docking, docking, and post- docking steps were performed as described previously. The tested amlodipine structural analogs, which had lower binding affinity to CaVAb than amlodipine, were selected for further investigation.
This virtual prediction was performed using the ProTox- II Web server25. This server, operated by Charité College of Medicine, Institute of Physiology, Structural Bioinformatics Group, Germany, is considered a virtual toxicity laboratory that allows the user to predict multiple toxicological endpoints associated with a chemical structure and contains computational models trained on real data (in vitro or in vivo) to predict the toxic potential of existing and virtual compounds. The prediction results include the oral lethal dose of 50% (LD50) of the tested molecule and, accordingly, the prediction of its oral toxicity class as either (Class I, lethal, LD50 ≤ 5 mg/Kg), (Class II, probably lethal, 5 mg/Kg < LD50 ≤ 50 mg/Kg), (Class III, toxic, 50 mg/Kg < LD50 ≤ 300 mg/Kg), (Class IV, harmful, 300 mg/Kg < LD50 ≤ 2000 mg/Kg), (Class V, may be harmful, 2000 mg/Kg < LD50 ≤ 5000 mg/Kg), or (Class VI, non-toxic, (LD50 > 5000 mg/Kg). In addition, prediction for a range of toxicity endpoints provided probability statements (in the range of 0–1) for active toxicity or inactive toxicity. The selected amlodipine structural analogs from this study were those predicted to have a higher (or at least similar) LD50 than amlodipine and had a probability reliability of > 0.50 for inactive toxicity at each toxicity25, 26.
This virtual computational prediction was performed to test whether the selected amlodipine structural analogs could be considered as drugs for humans. For this purpose, three established rules were used, including the Lipinski rule, the Egan filter, and the Mugger filter. The structural analog that did not violate a single criterion of any of these rules was classified as a high potential drug. The test was performed using the web server “SwissADME” which is operated by the Swiss Institute of Bioinformatics (SIB) in Switzerland27, 28.
The prediction of pharmacokinetics (absorption, distribution, metabolism, and excretion) of the selected amlodipine structural analogs was performed using the web server pkCSM (Pharmacokinetics Computation of Small Structural Molecules) web server (https://biosig.lab.uq.edu.au/pkcsm/prediction). The server, which is operated by the College of Melbourne, Australia, uses graphics-based signatures to predict the pharmacokinetics of small molecules29, 30.
As shown in (Table 2), the webserver SuprePred 3 predicted that amlodipine could be used for bacterial infections and linked this indication to two bacterial enzymes as potential targets: bacterial acyl-CoA desaturase (BAD) and bacterial DNA topoisomerase I (BDT1) with prediction probabilities of 53.54 and 51.59%, respectively. After downloading the structures of the two predicted bacterial targets (encoded as A0A0Q1RQC1 and Q06AK7, respectively) from the UniProt protein database 14(a), 14(b) as shown in (Figure 2) and docking them with amlodipine, it was found that the drug had higher binding affinity to BDT1 (- 6.366 kcal/mol), than that to the other enzyme (- 5.480 kcal/mol). Moreover, as demonstrated in (Table 3) and (Figure 3), the binding interactions of the drug with the two bacterial enzymes showed that amlodipine had two strong ionic bonds with both enzymes. However, 4 weak bonds between amlodipine and BAD were found compared to only 1 weak bond between amlodipine and BDT1. The results of amlodipine docking with the two enzymes were compared to those of two reference ligands of the enzymes including with acyl-CoA31,32 and {N,N-dimethyl-2-(phenanthro]3,4-d][dioxol-5-yl)ethanamine}33, respectively. The binding affinity of amlodipine to BDT1 was found to be higher and with stronger bond interactions than that of the reference ligand of the enzyme (-4.739 kcal/mol). In another respect, after downloading the structure of the human version of DNA topoisomerase I (HDT1) from the UniProt protein database14(c), as shown in (Figure 2), the molecular docking predicted lower affinity (-4.532 kcal/mol) and weaker bond interaction between amlodipine and that version as shown in (Tables 2,3) and (Figure 3).
Figure 2: 3D structures of proteins downloaded from the protein databases (I: Bacterial Acyl CoA desaturase14 (a); II: Bacterial DNA topoisomerase I 14(b); III: Human DNA topoisomerase I 14(c); IV: CaVAb: Calcium ion transporter protein24).
Table 2: SuperPred 3 probability and molecular docking results for predicting the bacterial target of amlodipine
Ligand |
SuperPred (P %) |
Molecular Docking; s (kcal/mol) |
||||
BAD |
BDT1 |
HDT1 |
BAD |
BDT1 |
HDT1 |
|
Amlodipine |
53.54 |
51.59 |
51.59 |
- 5.480 |
- 6.366 |
- 4.532 |
Acyl CoA ▲ |
65.95 |
52 |
52 |
-8.607 |
n.t |
n.t |
N,N-dimethyl(phenanthro]3,4-d][dioxol-5-yl) ethanamine |
n.r |
66.86 |
66.86 |
n.t |
-4.739 |
-5.522 |
(P %): prediction probability percentage; BAD: Bacterial acyl CoA desaturase; BDT1: Bacterial DNA topoisomerase I; HDT1: Human DNA topoisomerase I; n.t: not tested; n.r: no results provided; s: final docking score; ▲: a reference ligand of Acyl CoA desaturase; : a reference ligand of DNA topoisomerase I;
Table 3: Binding interaction modes of amlodipine and reference ligands with the target residues
ligand |
Target |
Bonded amino acid residue |
Number, type, (energy kcal/mol) of interaction |
Amlodipine
|
BDT1 |
Glu86 |
2, ionic bonds, (- 1, - 5.1) |
Arg139 |
1, H bond ( - 0.2) |
||
Arg458 |
1, H bond (- 0.6) |
||
HDT1 |
Asp483 |
1, H bond (- 2.6); 1, ionic bond (- 3.9) |
|
Gln600 |
1, H bond (- 0.2) |
||
BAD |
Thr13 |
1, H bond (- 0.2) |
|
Asn90 |
1, H bond (- 0.4) |
||
Glu136 |
1, H bond (- 0.5); 2, ionic bonds ( - 4.3, - 0.7) |
||
Tyr158 |
1, Polar -π bond (- 0.2) |
||
N,N-dimethyl-2- (phenanthro]3,4- d][dioxol-5-yl) ethanamine |
BDT1 |
Arg140 |
3, Polar -π bond (- 0.2, - 1.7, -1.5) |
HDT1 |
Ala625 |
2, Polar -π bond ( - 0.5, - 0.6) |
|
Acyl CoA |
BAD |
Ala14 |
1, H bond (- 0.4) |
Tyr114 |
1, Polar -π bond (- 0.3) |
||
Met128 |
1 H bond (- 0.7) |
||
Glu136 |
1 H bond ( - 5.4) |
||
Tyr158 |
1 H bond (- 0.6) |
BAD: Bacterial acyl CoA desaturase; BDT1: Bacterial DNA topoisomerase I; HDT1: Human DNA topoisomerase I; Ala: Alanine; Arg: Arginine;; Asp: Aspartic acid;Asn: Asparagine; Gln: Glutamine; Glu: Glutamic acid; Met: Methionine; Thr: Threonine; Tyr: Tyrosine;
a) Antibacterial and CCB activities:
As demonstrated in (Table 4), out of all designed amlodipine structural analogs, only 13 were predicted by SuperPred 3 to have an indication for bacterial infections by acting on the same bacterial target predicted for amlodipine with a prediction probability equal to or greater than that of amlodipine. However, when molecular docking was used to investigate those analogs it was found that only 3 structural analogs D074, D075 and D082 showed higher or equal binding affinity to bacterial DNA topoisomerase than that of amlodipine. Besides, the three structural analogs showed binding energy of (> -6.366kcal/mol) and (> -6.393kcal/mol) to the calcium ion transporter (CaVAb) and the human version of DNA topoisomerase I which indicated lower binding affinity of these analogs than that of amlodipine.
As shown in (Table 5), the predicted LD50 and the toxicity class of the three selected amlodipine structural analogs were identical to those of amlodipine. Furthermore, the probability confidence of inactive toxicity for each structural analog was ≤ 0.5 at each toxicity endpoint.
Table 4: Prediction of antibacterial and CCB activities of amlodipine structural analogs
Ligand code |
Antibacterial activity |
Molecular Docking with HDT1; s▲ (kcal/mol) |
Activity as CCB Molecular Docking with CaVAb; s▲ (kcal/mol) |
|
SuperPred 3 (P %) with BDT1 |
Molecular Docking with BDT1; s▲ (kcal/mol) |
|||
AMLO |
51.59 |
-6.366 |
- 4.532 |
- 6.393 |
D004 |
52.55 |
- 6.115 |
n.t |
n.t |
D006 |
52.18 |
- 5.451 |
n.t |
n.t |
D018 |
53.04 |
- 4.981 |
n.t |
n.t |
D027 |
55.86 |
- 5.347 |
n.t |
n.t |
D029 |
51.59 |
- 5.300 |
n.t |
n.t |
D030 |
53.78 |
- 5.800 |
n.t |
n.t |
D038 |
54.27 |
- 4.889 |
n.t |
n.t |
D067 |
59.88 |
- 5.934 |
n.t |
n.t |
D074 |
61.30 |
- 7.051 |
- 4.381 |
- 6.068 |
D075 |
60.05 |
- 6.808 |
- 4.029 |
- 3.138 |
D076 |
61.39 |
- 5.258 |
n.t |
n.t |
D077 |
57.2 |
-5.812 |
n.t |
n.t |
D082 |
55.41 |
- 6.392 |
- 4. 183 |
- 5.011 |
BDT1: Bacterial DNA topoisomerase I; HDT1: Human DNA topoisomerase I; CCB: calcium channel blocker; CaVAb: human calcium channel; AMLO: amlodipine; : Probability of prediction; ▲: Final docking score representing the energy of binding; n.t: not tested
Table 5: Prediction of the toxicity profiles of amlodipine structural analogs
Toxicity Parameter |
AMLO |
D074 |
D075 |
D082 |
LD50 mg/kg |
37 |
37 |
37 |
37 |
Toxicity Class |
II |
II |
II |
II |
Endpoints |
Probability confidence for Inactive toxicity |
|||
Hepatotoxicity |
0.84 |
0.55 |
0.57 |
0.82 |
Carcinogenicity |
0.60 |
0.96 |
0.95 |
0.61 |
Immunotoxicity |
0.96 |
0.75 |
0.79 |
0.98 |
Mutagenicity |
0.76 |
0.68 |
0.67 |
0.78 |
Cytotoxicity |
0.64 |
0.91 |
0.88 |
0.65 |
Aryl hydrocarbon Receptor |
0.91 |
0.95 |
0.95 |
0.92 |
Androgen Receptor |
0.96 |
0.84 |
0.84 |
0.96 |
Androgen Receptor Ligand Binding Domain |
0.66 |
0.79 |
0.79 |
0.77 |
Aromatase |
0.61 |
0.91 |
0.86 |
0.74 |
Estrogen Receptor |
0.93 |
0.92 |
0.92 |
0.91 |
Estrogen Receptor Ligand Binding Domain |
0.90 |
0.94 |
0.92 |
0.94 |
Peroxisome Proliferator Activated Receptor Gamma |
0.79 |
0.87 |
0.86 |
0.93 |
Nuclear factor (erythroid-derived 2)-like 2/antioxidant responsive element |
0.80 |
0.87 |
0.86 |
0.85 |
Heat shock factor response element |
0.80 |
0.84 |
0.81 |
0.85 |
Mitochondrial Membrane Potential |
0.80 |
0.73 |
0.76 |
0.87 |
Phosphoprotein (Tumor Supressor) p53 |
0.54 |
0.98 |
0.99 |
0.71 |
ATPase family AAA domain containing protein 5 |
0.97 |
0.55 |
0.57 |
0.98 |
Table 6: Prediction of the drug-likeness of selected structural analogs of amlodipine
Rule |
Criterion |
AMLO |
D074 |
D075 |
D082 |
Lipinski's |
No. of H-bond (donor) ≤ 5 |
2 |
1 |
1 |
2 |
No. of H-bond (acceptor) ≤ 10 |
6 |
8 |
8 |
8 |
|
Mwt ≤ 500 |
408.88 |
464.98 |
493.04 |
466.91 |
|
MLOGP ≤ 4.15 |
1.33 |
2.18 |
2.59 |
0.93 |
|
Egan`s |
WLOGP ≤ 5.88 |
1.89 |
3.4 |
4.18 |
1.68 |
TPSA ≤ 131.6 oA |
99.88 |
91.09 |
91.09 |
128.39 |
|
Muegg`s |
Mwt: 200 -600 |
408.88 |
464.98 |
493.04 |
466.91 |
XLOGP3: -2 to 5 |
3.00 |
4.39 |
5 |
0.45 |
|
TPSA ≤ 150 oA |
99.88 |
91.09 |
91.09 |
128.39 |
|
No. of rings ≤ 7 |
2 |
2 |
2 |
2 |
|
No. of (C) atoms > 4 |
20 |
24 |
26 |
22 |
|
No. of heteroatoms > 1 |
8 |
8 |
8 |
10 |
|
No. of rotatable bonds ≤ 15 |
10 |
13 |
14 |
12 |
|
No. of H-bond (donor) ≤ 5 |
2 |
1 |
1 |
2 |
|
No. of H-bond (acceptor) ≤ 10 |
6 |
8 |
8 |
8 |
AMLO: amlodipine; MWt: Molecular Weight (g/mole); MLOGP: Moriguchi octanol-water partition coefficient; WLOGP: Atomistic Octanol/water Partition coefficient; M.R.: Molar reactivity; TPSA: Topological Polar Surface area; XLOGP3: Atomistic and knowledge Octanol/water Partition coefficient
(Table 6) demonstrates the prediction of drug-likeness of the three selected amlodipine analogs. None of the investigated analogs violated the criteria of Lipinski's rule, Egan's filter, or Muegge's filter.
The results of prediction of pharmacokinetics of the three selected amlodipine analogs are demonstrated in (Table 7). The predictions of oral absorption of the analogs (D074) and (D075) were high as demonstrated with lower water solubility Log S (i.e. high lipid solubility) and higher Caco2 permeability (Log Papp), intestinal absorption, and ability to inhibit P-glycoprotein than those predicted for amlodipine. Concerning dermal absorption, amlodipine as well as the three analogs were predicted to have the ability to penetrate skin with log Kp (< -2.5 cm/hr)29. With respect to the prediction of the analogs distribution, the analogs (D074) and (D075) showed a higher probability to distribution (Log VDSS) than that predicted amlodipine which is known to have a clinically estimated high volume of distribution of 21 L/kg34. However, the two analogs had similar predictions as amlodipine to not pass brain barrier with predicted permeability (Log PS < -2 cm3/s)29. Concerning the prediction of the analogs metabolism, (D074) and (D075) were predicted to be similar to amlodipine as substrates and inhibitors of CYP3A4. Eventually, with respect to drug excretion rate, the predicted total clearance of D074 and D075 (0.746 and 0.748 log ml/min/kg) were close to that predicted for amlodipine (0.651 log ml/min/kg).
Table 7: Prediction of the pharmacokinetics of selected amlodipine structural analogs
Ligand |
Absorption parameters |
Distribution parameters |
Metabolism parameters |
Excretion parameters |
|||||||||
Skin |
Oral |
||||||||||||
Log Kp |
Log S |
Log Papp |
Pgp substrate |
Pgp I/II inhibitor |
Intestinal Absorption % |
Log VDss |
fun |
Log PS |
CYP3A4/CYP2D6 substrate |
CYP3A4/CYP2D6 inhibitor |
Log ClT |
OCT2 substrate |
|
AMLO |
-2.822 |
-3.236 |
0.836 |
Yes |
Yes |
84.664 |
-0.058 |
0.241 |
-3.442 |
Yes/Yes |
Yes/Yes |
0.636 |
No |
D074 |
-2.925 |
-4.317 |
1.111 |
Yes |
Yes |
95.363 |
0.071 |
0.131 |
-2.985 |
Yes/No |
Yes/No |
0.746 |
No |
D075 |
-2.840 |
5.265 |
0.926 |
Yes |
Yes |
94.46 |
0.143 |
0.049 |
-2.876 |
Yes/No |
Yes/No |
0.748 |
No |
D082 |
-2.735 |
-3.227 |
0.638 |
Yes |
No |
61.004 |
-0.879 |
0.371 |
-3.267 |
No/No |
No/No |
0.651 |
No |
AMLO: amlodipine; Kp: skin permeability constant (cm/hr); S: water solubility (mole/L); Papp: Caco2 permeability of the molecule (10-6 cm/s); Pgp: P-glycoprotein; VDss: volume of distribution (L/Kg) in human body at steady state; PS: permeability-Surface area product of blood-brain barrier (cm3/s); CYP3A4: Cytochrome P450 3A4; CYP2D6: Cytochrome P450 2D6; ClT: total clearance (ml/min/kg); OCT2: Organic Cation Transporter 2
Figure 4: 3D Conformations showing the different moieties in the chemical structures of amlodipine and selected analogs
As reported in the literature, the assumption suggesting that amlodipine antibacterial action is due to blocking the bacterial calcium channel may be doubtful35. It is well known that mammalian voltage- dependent calcium channels (CaVs) are phylogenetically related to the bacterial voltage-gated sodium channels (BacNaVs) with very similar amino acid sequences present in both types, and so they are used as a model for gating and ion permeation36. Recently, two bacterial voltage-dependent calcium channels (CaVMr and NaVPp) have been recognized, which are selective for Ca2+, and for Na+ with Ca2+ dependent inhibition, respectively37. When amlodipine is administered to humans at a standard daily dose of 10 mg, it produces a maximal serum concentration of 5.9 x10-3 µg/ml which is adequate for the drug’s action as CCB35. Therefore, owing to the similarity between mammalian and bacterial calcium channels, it is supposed that the same concentration is sufficient to block the ion channels in bacteria as well, resulting in bacterial killing. However, in vitro investigations of amlodipine antibacterial activity revealed that the minimum inhibitory concentration (MIC) of the drug against all tested bacteria (e.g. S. aureus ~10-400 µg/ml) which is extremely greater than its maximum serum concentration35. This may indicate that the drug may have a bactericidal mechanism of action that is unrelated to its activity as CCB. In another theory, it was found that amlodipine enhances the activity of imipenem against multidrug-resistant Acinetobacter baumanii by inhibiting expression of the resistance nodulation cell division efflux pump AdeABC6. However, this theory was not in agreement with other studies that reported the presence of factors other than blocking the efflux pump that influenced the antibacterial action of the drug, such as its ionization as well as the environmental pH that surrounds the infection pathway38-40.
Bacterial Acyl CoA desaturase was one of the two bacterial targets initially predicted in this study as bacterial target for amlodipine. This enzyme is a membrane enzyme belonging to the superfamily of "fatty acid desaturases (FADs)," which catalyze the introduction of double bonds into fatty acids and are therefore necessary components of cell membrane anabolic long-chain polyunsaturated fatty acids31, 32. Although there are studies which have shown that it is problematic to consider the fatty acid biosynthetic pathway as a potential bacterial target because exogenous fatty acids from the host serum can overcome inhibition of the bacterial pathway32, this enzyme was investigated in this study for the purpose of verification by molecular docking. The other initial predicted target "bacterial DNA topoisomerase I" is an enzyme that belongs to the family of DNA topoisomerases. The latters are known to play important roles in DNA replication, transcription, repair, and recombination. The results of the present study predicted not only higher binding affinity of amlodipine than that of a reference ligand of that enzyme, but also stronger bond interactions between amlodipine and this enzyme. Indeed, the results supported the theory that DNA topoisomerase I could be a potential bacterial target for new antibiotics as reported in the litearature33.
All amlodipine analogs that had considerable SuperPred predictions to have antibacterial activity by acting on the same bacterial target of amlodipine were observed to possess 4-Hydropyridine nucleus instead of 1,4-dihydropyridine nucleus of amlodipine. However, of these analogs, molecular docking investigation showed that only three analogs (D074, D075 ) and (D085) with structures bearing the nuclei of 1-butyl 4-hydropyridine and 4-hydropyridine 1-acetic acid, respectively, as demonstrated in (Figure 4), would have more binding affinity and stronger bond interactions to the target enzyme than that of amlodipine. The three analogs were also predicted to have low affinity to human DNA topoisomerase I, diminished activity as CCB, accepted toxicity profiles and drug-likeness. However, the pharmacokinetics profiles predicted for (D074) and (D075) showed greater oral and dermal absorption, wider drug distribution, similar metabolism profile and close excretion rate to that of amlodipine. Although, similar to amlodipine the two analogs had low prediction to pass blood-brain barrier and hence may be not suitable for CNS infections. This property is not not be a problem that could cease the development of these compounds as antibacterials since many antibiotics have the same drawback e.g. piperacillin, cefuroxime, cefixime and imipenem though the penetration of these antibiotics increases in case of strong meningeal inflammation41.
Based on results of this study, it could be concluded that the structural analogs of amlodipine (D074 and D075) which bears the 1-butyl 4-hydropyridine nucleus are promising candidates to be lead compounds for the discovery of new antibacterial drugs.
CONFLICT OF INTEREST:
The authors have no conflicts of interest regarding this investigation.
REFERENCES:
1. Lagadinou M, Onisor MO, Rigas A, Musetescu DV, Gkentzi D, Assimakopoulos SF, Panos G, Marangos M. Antimicrobial Properties on Non-Antibiotic Drugs in the Era of Increased Bacterial Resistance. Antibiotics (Basel). 2020; 9(3): 107. doi.org/10.3390/antibiotics9030107
2. Laudy AE. Non-antibiotics, Efflux Pumps and Drug Resistance of Gram-negative Rods. Pol. J. Microbiol. 2022; 67(2): 129-135. doi.org/10.21307/pjm-2018-017
3. The United States Pharmacopoeia 2020: USP 43 ; The national formulary: NF 38. United States Pharmacopeial Convention.
4. Ferrari R, Pavasini R, Camici PG, Crea F, Danchin N, Pinto F, Manolis A, Marzilli M, Rosano GMC, Lopez-Sendon J, Fox K. Anti-anginal drugs-beliefs and evidence: systematic review covering 50 years of medical treatment. Eur Heart J. 2019; 40(2):190-194. doi.org/10.1093/eurheartj/ehy504
5. Wang Y.; Li X.; Wang D.; Sun, S.; Lu, C. In Vitro Interactions of Ambroxol Hydrochloride or Amlodipine in Combination with Antibacterial Agents against Carbapenem‐Resistant Acinetobacter Baumannii. Lett. Appl. Microbiol. 2019; 70(3): 189– 195. doi.org/10.1111/lam.13259
6. Hu C, Li Y, Zhao Z, Wie S, Zhao Z, Chen H, Wu P. In vitro synergistic effect of amlodipine and imipenem on the expression of AdeABC efflux pump in multidrug-resistant Acinetobacter baumanii. PLoS ONE. 2018; 13: e0198061. doi.org/ 10.1371/journal.pone.0198061
7. Mazumdar K, Ashok Kumar K, Dutta NK. Potential role of the cardiovascular non-antibiotic (helper compound) amlodipine in the treatment of microbial infections: scope and hope for the future. Int J Antimicrob Agents. 2010; 36(4): 295-302. doi.org/10.1016/j.ijantimicag.2010.05.003
8. Asok Kumar, K.; Mazumdar K., Dutta, N. K., Karak, P.; Dastidar, S. G.; Ray, R. Evaluation of Synergism between the Aminoglycoside Antibiotic Streptomycin and the Cardiovascular Agent Amlodipine. Biol. Pharm. Bull. 2004; 27(7): 1116– 1120. doi.org/10.1248/bpb.27.1116
9. Supre-Pred [Internet]. Target prediction. Available from: https://prediction.charite.de/subpages/target_prediction.php Accessed on January 1st, 2022
10. 10. Nickel J, Gohlke BO, Erehman J, Banerjee P, Rong WW, Goede A, Dunkel M, Preissner R. SuperPred: update on drug classification and target prediction. Nucleic Acids Res. 2014; 42(Web Server issue):W26-31.doi.org/10.1093/nar/gku477
11. Attique SA, Hassan M, Usman M, Atif RM, Mahboob S, Al-Ghanim KA, Bilal M, Nawaz MZ. A Molecular Docking Approach to Evaluate the Pharmacological Properties of Natural and Synthetic Treatment Candidates for Use against Hypertension. Int J Environ Res Public Health. 2019 ; 4; 16(6): 923.doi.org/10.3390/ijerph16060923
12. Alnajjar R, Mostafa A, Kandeil A, Al-Karmalawy AA. Molecular docking, molecular dynamics, and in vitro studies reveal the potential of angiotensin II receptor blockers to inhibit the COVID-19 main protease. Heliyon. 2020;6 (12): e05641. doi.org/10.1016/j.heliyon.2020.e05641
13. Ahmed A, Saeed A, Ejaz SA, Aziz M, Hashmi MZ, Channar PA, Abbas Q, Raza H, Shafiq Z, El-Seedi HR. Novel adamantyl clubbed iminothiazolidinones as promising elastase inhibitors: design, synthesis, molecular docking, ADMET and DFT studies. RSC Adv. 2022; 12(19): 11974-11991. doi.org/10.1039/d1ra09318e
14. UniProt protein database. Uni Port Knowledgebase (UniProtkb). Available from: (a):https://www.uniprot.org/uniprotkb/A0A0Q1RQC1/entry (Accessed on January 15th, 2022) (b): https://www.uniprot.org/uniprotkb/Q06AK7/entry (Accessed on January 16th, 2022) (c): https://www.uniprot.org/uniprotkb/B9EG90/entry (Accessed on April 28th, 2022)
15. Terlouw BR, Vromans SPJM, Medema MH. PIKAChU: a Python-based informatics kit for analysing chemical units. J Cheminform. 2022; 14(1): 34. doi.org/10.1186/s13321-022-00616-5
16. O'Boyle NM, Banck M, James CA, Morley C, Vandermeersch T, Hutchison GR. Open Babel: An open chemical toolbox. J Cheminform. 2011; 3: 33.doi.org/10.1186/1758-2946-3-33
17. Pettersen EF, Goddard TD, Huang CC, Couch GS, Greenblatt DM, Meng EC, Ferrin TE. UCSF Chimera--a visualization system for exploratory research and analysis. . J Comput Chem. 2004; 25(13): 1605-1605.doi.org/10.1002/jcc.20084
18. Shaldam MA, Elhamamsy MH, Esmat EA, El-Moselhy TF. 1,4-Dihydropyridine Calcium Channel Blockers: Homology Modeling of the Receptor and Assessment of Structure Activity Relationship. ISRN Med. Chem. 2014; 2014: e203518. doi.org/10.1155/2014/203518
19. Royal Society of Chemistry-UK. ChemSpider. Structure Search.. Available from https://www.chemspider.com/StructureSearch.aspx; (Accessed on May 15th,022)
20. American Chemical Society-USA, Chemical Abstract Service's (CAS). Available from: https://scifinder-n.cas.org; (Accessed on May 17th,022)
21. National Institutes of Health (NIH)-USA, Pubchem. Available from: https://pubchem.ncbi.nlm.nih.gov; (Accessed on May 18th,2022)
22. Zhao Y, Huang G, Wu J, Wu Q, Gao S, Yan Z, Lei J, Yan N. Molecular Basis for Ligand Modulation of a Mammalian Voltage- Gated Ca2+ Channel. Cell. 2019; 177(6): 1495-1506. doi.org/10.1016/j.cell.2019.04.043
23. Tang L, Gamal El-Din TM, Swanson TM, Pryde DC, Scheuer T, Zheng N, Catterall WA. Structural Basis for Inhibition of a Voltage-Gated Ca2+ Channel by Ca2+ Antagonist Drugs. Nature. 2016; 537(7618): 117–121. doi.org/10.1038/nature19102
24. Protein Data Bank [Cited 2022 March 28]. Available from: https://www.rcsb.org/structure/5KMD.
25. Charité College of Medicine, Institute of Physiology, Structural Bioinformatics Group, Germany. ProTox II, Prediction of toxicity of Chemicals. Available from: https://tox-new.charite.de/protox_II/index.php?site=compound_input ; (Accessed on August 11th,2022)
26. Drwal MN., Banerjee P, Dunkel M, Wettig MR, Preissner R. ProTox: a web server for the in silico prediction of rodent oral toxicity. Nucleic Acids Res. 2014; 42: 53–58. doi.org/10.1093/nar/gku401
27. Swiss Institute of Bioinformatics (SIB). SwissADME. Available from: http://www.swissadme.ch/index.php ; (Accessed on October 10th,2022)
28. Daina A, Michielin O, Zoete V. SwissADME: a free web tool to evaluate pharmacokinetics, drug-likeness and medicinal chemistry friendliness of small molecules. Sci Rep. 2017; 7: 42717.doi.org/10.1038/srep42717
29. College of Melbourne, Australia. Pharmacokinetics Computation of Small Structural Molecules (pkcsm).Available from: https://biosig.lab.uq.edu.au/pkcsm/prediction ; (Accessed on December 17th,2022)
30. Pires EV, Blundell TL, Ascher DB. PkCSM: Predicting Small-Molecule Pharmacokinetic and Toxicity Properties Using Graph- Based Signatures. Journal of Medicinal Chemistry. 2015; 58 (9): 4066–4072. doi.org/10.1021/acs.jmedchem.5b00104
31. Li D, Moorman R, Vanhercke T, Petrie J, Singh S, Jackson CJ. Classification and substrate head-group specificity of membrane fatty acid desaturases. Comput Struct Biotechnol J. 2016; 14: 341-349.doi.org/10.1016/j.csbj.2016.08.003
32. Brinster S, Lamberet G, Staels B, Trieu-Cuot P, Gruss A, Poyart C. Type II fatty acid synthesis is not a suitable antibiotic target for Gram-positive pathogens. Nature. 2009; 458: 83-86. doi.org/10.1038/nature07772
33. Tse-Dinh YC. Bacterial topoisomerase I as a target for discovery of antibacterial compounds. Nucleic Acids Res. 2009; 37(3): 731-737. doi.org/10.1093/nar/gkn936
34. US Food & Drug administration (FDA). Norvasc (Amlodipine besylate) highlights of prescription information. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2011/019787s047lbl.pdf; (Accessed on February 24th,2022)
35. Dalhoff A.Are antibacterial effects of non‑antibiotic drugs random or purposeful because of a common evolutionary origin of bacterial and mammalian targets? Infection; 2021; 49: 569–589. doi.org/10.1007/s15010-020-01547-9
36. Ren D, Navarro B, Xu H, Yue L, Shi Q, Clapham DE. A prokaryotic voltage-gated sodium channel. Science. 2001; 294: 2372– 2375. doi.org/10.1126/science.1065635
37. Shimomura T, Yonekawa Y, Nagura H, Tateyama M, Fujiyoshi Y, Irie K. A native prokaryotic voltage-dependent calcium channel with a novel selectivity filter sequence. Elife. 2020; 25, 9: e52828. doi.org/10.7554/elife.52828
38. Pohl EE, Krylov AV, Block M, Pohl P. Changes of the membrane potential profile induced by verapamil and propranolol. Biochim Biophys Acta. 1998; 1373: 170–8. doi.org/10.1016/s0005-2736(98)00098-4
39. Chen C, Gardete S, Jansen RS, Shetty A, Dick T, Rhee KY, Dartois V. Verapamil targets membrane energetics in Mycobacterium tuberculosis. Antimicrob Agents Chemother. 2018; 62: e02107-e2117.doi.org/10.1128/aac.02107-17
40. Herbette L, Vant-Erve YMH, Rhodes D. Interaction of 1,4 dihydropyridine calcium channel antagonists with biological membranes: lipid bilayer partitioning could occur before drug binding to receptors. J Mol Cell Cardiol. 1989; 21: 187–201. doi.org/10.1016/0022-2828(89)90861-4
41. Nau R, Sörgel F, Eiffert H. Penetration of Drugs through the Blood-Cerebrospinal Fluid/Blood-Brain Barrier for Treatment of Central Nervous System Infections Clin Microbiol Rev. 2010; 23(4): 858–883.doi.org/10.1128/cmr.00007-10
Received on 14.08.2023 Modified on 06.11.2023
Accepted on 08.01.2024 © RJPT All right reserved
Research J. Pharm. and Tech 2024; 17(5):2271-2281.
DOI: 10.52711/0974-360X.2024.00357