Acessibilidade / Reportar erro

Biological activity of synthesized 5-{1-[(4-chlorophenyl)sulfonyl]piperidin-4-yl}-2-mercapto-1,3,4-oxadiazole derivatives demonstrated by in silico and BSA binding studies

Abstract

We synthesized a series of compounds bearing pharmacologically important 1,3,4-oxadiazole and piperidine moieties. Spectral data analysis by 1H-NMR, 13C-NMR, IR and EI-MS was used to elucidate the structures of the synthesized molecules. Docking studies explained the different types of interaction of the compounds with amino acids, while bovine serum albumin (BSA) binding interactions showed their pharmacological effectiveness. Antibacterial screening of these compounds demonstrated moderate to strong activity against Salmonella typhi and Bacillus subtilis but only weak to moderate activity against the other three bacterial strains tested. Seven compounds were the most active members as acetyl cholinesterase inhibitors. All the compounds presented displayed strong inhibitory activity against urease. Compounds 7l, 7m, 7n, 7o, 7p, 7r, 7u, 7v, 7x and 7v were highly active, with respective IC50 values of 2.14±0.003, 0.63±0.001, 2.17±0.006, 1.13±0.003, 1.21±0.005, 6.28±0.003, 2.39±0.005, 2.15±0.002, 2.26±0.003 and 2.14±0.002 µM, compared to thiourea, used as the reference standard (IC50 = 21.25±0.15 µM). These new urease inhibitors could replace existing drugs after their evaluation in comprehensive in vivo studies.

Keywords:
1,3,4-Oxadiazole; Acetylcholinesterase (AChE) inhibition; Antibacterial activity; Piperidine; Sulfonamide; Urease inhibition


INTRODUCTION

1,3,4-Oxadiazole is a pharmacologically important heterocyclic core, and a number of biological activities have been described for derivatives with this moiety. Among these bioactivities, the most important ones are antibacterial (Li et al., 2014Li P, Shi L, Gao MN, Yang X, Xue W, Jin LH, Hu DY, Song BA. Antibacterial activities against rice bacterial leaf blight and tomato bacterial wilt of 2-mercapto-5-substituted-1,3,4-oxadiazole/thiadiazole derivatives. Bioorg. Med. Chem. Lett. 2014;25(3):481-484.), anti-inflammatory (Omar, Mahfouz, Rahman, 1996Omar FA, Mahfouz NM, Rahman MA. Design, synthesis and anti-inflammatory activity of some 1,3,4-oxadiazole derivatives. Eur J Med Chem. 1996;31(10):819-825.), anticancer (Kumar et al., 2009Kumar D, Sundaree S, Johnson EO, Shah K. An efficient synthesis and biological study of novel indolyl-1,3,4-oxadiazoles as potent anticancer agents. Bioorg Med Chem Lett. 2009;19(15):4492-4494.), antihepatitis (Tan et al., 2006Tan TMC, Chena Y, Kong KH, Bai J, Li Y, Lim SG, Ang TH, Lam Y. Synthesis and the biological evaluation of 2-benzenesulfonylalkyl-5-substituted-sulfanyl-[1,3,4]-oxadiazoles as potential anti-hepatitis B virus agents. Antiviral Res. 2006;71(1):7-14.), antitumor (Zhang et al., 2014Zhang K, Wang P, Xuan LN, Fu XY, Jing F, Li S, Liu YM, Chen BQ. Synthesis and antitumor activities of novel hybrid molecules containing 1,3,4-oxadiazole and 1,3,4-thiadiazole bearing Schiff base moiety. Bioorg Med Chem Lett. 2014;24(22):5154-5156.) and antiproliferative (El-Din et al., 2015El-Din MMG, El-Gamal MI, Abdel-Maksoud MS, Yoo KH, Oh CH. Synthesis and broad-spectrum anti-proliferative activity of diarylamides and diarylureas possessing 1,3,4-oxadiazole derivatives. Bioorg Med Chem Lett. 2015;25(8):1692-1699.), among many other activities. Another key bioactive heterocyclic core is piperidine. The various bioactivities of this nucleus have been evaluated via the synthesis of a number of piperidine derivatives (Sanchez-Sancho, Herrandón, 1998Sanchez-Sancho F, Herrandon B. Short syntheses of (S)-pipecolic acid, (R)-coniine, and (S)-coniceine using biocatalytically-generated chiral building block. Tetrahedron Asymmetry. 1998;9(11):1951-1965.). Compounds bearing the piperidine nucleus are associated with anesthetic activity, treatment of cocaine abuse, and controlling plasma glucose and insulin (Nithiya, Karthik, Jayabharathi, 2011Nithiya S, Karthik N, Jayabharathi J. In vitro antioxidant activity of hindered piperidone derivatives. Int J Pharm Pharm Sci. 2011;3(3):254-256.). Among the different functionalities, sulfamoyl is one of the most important ones regarding pharmacological and therapeutic potential. The pharmacological behavior of this moiety is mainly related to antibacterial action, enzyme inhibition, cancer chemotherapy, hypoglycemic activity, diuretic action and many other uses (Aziz-ur-Rehman et al., 2011Aziz-ur-Rehman, Tanveer W, Abbasi MA, Afroz S, Khan KM, Ashraf M, Afzal I. Synthesis, characterization and biological screening of various N-substituted derivatives of sulfonamides. Int J Chem Res. 2011;3(3):99-104.).

The synthesized compounds were evaluated for antibacterial activity, enzyme inhibitory activity (acetylcholinesterase (AChE) and urease) and bovine serum albumin (BSA) binding. The bacterial strains taken into account were Salmonella Typhi, Escherichia coli, Pseudomonas aeruginosa, Staphylococcus aureus and Bacillus subtilis, which are involved in enteric fever (Bhattacharya, Das, Choudhury, 2011Bhattacharya SS, Das U, Choudhury BK. Occurrence & antibiogram of Salmonella typhi & S. paratyphi A isolated from Rourkela, Orissa. Indian J Med Res. 2011;133:431-433.), food poisoning (Vogt, Dippold, 2005Vogt RL, Dippold L. Escherichia coli O157:H7 outbreak associated with consumption of ground beef, June-July 2002. Public Health Rep. 2005;120(2):174-178.), chronic infection (Pressler et al., 2011Pressler T, Bohmova C, Conway S, Dumcius S, Hjelte L, Høiby N, Kollberg H, Tümmler B, Vavrova V. Chronic Pseudomonas aeruginosa infection definition: EuroCareCF Working Group report. J Cyst Fibros. 2011;10(Suppl 2):S75-S78.), pathogen adherence to extracellular matrix or plasma proteins (Harris, Foster, Richards, 2002Harris LG, Foster SJ, Richards RG. An introduction to Staphylococcus aureus, and techniques for identifying and quantifying S. aureus adhesins in relation to adhesion to biomaterials: review. Eur Cells Mater. 2002;4:39-60.) and skin allergy or hypersensitivity (Barbe et al., 2009Barbe V, Cruveiller S, Kunst F, Lenoble P, Meurice G, Sekowska A, Vallenet D, Wang T, Moszer I, Medigue C, Danchin A. From a consortium sequence to a unified sequence: the Bacillus subtilis 168 reference genome a decade later. Microbiology. 2009;155(Pt 6):1758-1775.), respectively. AChE (EC 3.1.1.7), a serine hydrolase, is known to hydrolyze the neurotransmitter acetylcholine leading to termination of nerve impulse in cholinergic synapses and Alzheimer’s disease (Tougu, 2001Tougu V. Acetylcholinesterase: mechanism of catalysis and inhibition. Curr Med Chem. 2001;1(2):155-170.). Urease (EC 3.5.1.5), urea amidohydrolase, is actively involved in pyelonephritis, peptic ulceration, kidney stones, hepatic encephalopathy, urolithiasis and urinary catheter incrustation (Lodhi et al., 2007Lodhi MA, Abbasi MA, Choudhary MI, Ahmad VU. Kinetic studies on triacontanyl palmitate: a urease inhibitor. Nat Prod Res. 2007;21(8):721-725.). Blood plasma proteins act carriers for the transportation of drugs and other substances throughout the body (Wani et al., 2018Wani TA, Bakheit AH, Abounassif MA, Zargar S. Study of interactions of an anticancer drug neratinib with bovine serum albumin: Spectroscopic and molecular docking approach. Front. Chem. 2018;6:47.). The most abundant plasma protein is albumin which binds with metabolites and ligands. The pharmacokinetic and pharmacodynamic properties can be understood through the binding interaction of serum albumin and drug molecules. BSA is widely used in this regard because of high resemblance in structure with human serum albumin (HSA) (Wani et al., 2017a; Wani et al., 2017b; Wani et al., 2017c).

Chemists and pharmacologists have been working extensively on the synthesis of new molecules to combat and control various diseases. In this regard, two heterocyclic cores, 1,3,4-oxadiazole and piperidine, and the sulfamoyl functionality of sulfonamides have been introduced as bioactive structures. The notable pharmacological behavior of these three functionalities prompted us to synthesize some new molecules bearing all these three moieties. As an extension of our previous work (Nafeesa et al., 2015Nafeesa K, Aziz-ur-Rehman, Abbasi MA, Siddiqui SZ, Rasool S, Hussain G, Ahmed I. Synthesis and biological screening of S-substituted derivatives of 5-{1-[(4-chlorophenyl)sulfonyl]-3-piperidinyl}-1,3,4-oxadiazole-2-yl sulfide. Asian J Chem. 2015;27(6):2105-2108.), the synthesized compounds were evaluated for their pharmacological potential as antibacterial agents and AChE and urease inhibitors.

MATERIAL AND METHODS

General

The IR spectra were recorded using KBr pellet method on a Jasco-320-A spectrometer with wave number in cm-1. Mass spectra (EI-MS) were recorded on a JMS-HX-110 spectrometer with a data system. Proton and carbon nuclear magnetic resonance spectra were recorded in CDCl3 solvent on a Bruker spectrometer operating at 300 and 400 MHz with chemical shifts in ppm and coupling constant in hertz (Hz). Purity of synthesized compounds was confirmed using thin layer chromatography (TLC), carried out on pre-coated silica gel G-25-UV254 aluminum plates, run under different solvent systems with varying proportions of ethyl acetate and n -hexane and visualized using a 254-nm UV lamp. Melting points of the synthesized compounds were recorded on a Griffin and George meting point apparatus with open capillary tube and were uncorrected.

Preparation of ethyl 1-[(4-chlorophenyl) sulfonyl] piperidin-4-carboxylate (3)

Ethyl isonipecotate (2; 0.003 mol) was dispersed in 40 mL of distilled water in a 250-mL round-bottom (RB) flask followed by addition of 4-chlorophenylsulfonyl chloride (1; 0.003 mol). During gradual addition of 1, the mixture was maintained at pH 10.0 with 18% aqueous Na2CO3. The mixture was stirred for 4 hours and monitored by TLC. Dilute HCl (2-3 mL) was added with continuous stirring to adjust pH at 6.0, and the mixture allowed to stand for 20-30 minutes. The precipitates were filtered out, washed with cold distilled water and dried.

Preparation of 1-[(4-chlorophenyl)sulfonyl]piperidin-4-carbohydrazide (4)

Compound 3 (0.0025 mol) was dissolved in 35 mL of methanol in a 250-mL RB flask, and 80% hydrazine hydrate (12 mL) was added, followed by refluxing for 3 hours. The reaction was monitored by TLC. Excess solvent was evaporated off and the precipitate was filtered out, washed with cold distilled water and dried.

Preparation of 5-{1-[(4-chlorophenyl)sulfonyl]piperidin-4-yl}-2-mercapto-1,3,4-oxadiazole (5)

Compound 4 (0.002 mol) was dissolved in 35 mL of ethanol in a 250-mL RB flask followed by the addition of KOH (0.002 mol) and refluxing for 30 minutes. After the addition of CS2 (0.004 mol), the reaction mixture was refluxed for 6 hours and monitored by TLC. Excess chilled distilled water (70 mL) was added and pH was adjusted to 6 with dilute HCl (3-4 mL) to obtain the precipitate. The precipitate was filtered out, washed with cold distilled water and dried.

General procedure for the synthesis of 5-{1-[(4-chlorophenyl)sulfonyl]piperidin-4-yl}-2-[(alkyl/aralkyl)thio]-1,3,4-oxadiazole (7a-z)

Compound 5 (0.0015 mol) was dissolved in N,N-dimethyl formamide (DMF, 15 mL) in a 50-mL RB flask and lithium hydride (0.0015 mol) was added. The reaction mixture was stirred for 30 minutes. Equimolar alkyl/aralkyl halides, 6a-z, were then added to the reaction mixture, which was further stirred for 4-5 hours. The reaction was monitored by TLC. Excess cold distilled water was added to obtain the precipitated final compounds, 7a-z, which were recovered by filtration, washed with distilled water and dried.

Biological activity assays

Antibacterial activity assay

Antibacterial activity was determined under aseptic conditions in sterile 96-well microplates. The method is based on the increase in the absorbance of the broth medium with microbial cell population during log phase growth (Nafeesa et al., 2015Nafeesa K, Aziz-ur-Rehman, Abbasi MA, Siddiqui SZ, Rasool S, Hussain G, Ahmed I. Synthesis and biological screening of S-substituted derivatives of 5-{1-[(4-chlorophenyl)sulfonyl]-3-piperidinyl}-1,3,4-oxadiazole-2-yl sulfide. Asian J Chem. 2015;27(6):2105-2108.). Ciprofloxacin was used as the reference standard.

Acetyl cholinesterase inhibition assay

AChE inhibitory activity was determined by following the reported method (Mahernia et al., 2015Mahernia S, Bagherzadeh K, Mojab F, Amanlou M. Urease Inhibitory Activities of some Commonly Consumed Herbal Medicines. Iran J Pharm Res. 2015;14(3):943-947.) with minor modifications. A mixture of Na2HPO4 buffer (pH 7.7), 10 µL of test compound (0.5 mM well-1) and 10 µL AChE (0.005 mM) was prepared. Absorbance of the mixture at 405 nm was recorded, and the mixture was then incubated for 15 min at 35 ºC. The enzyme reaction was started by the addition of 10 µL of 0.5 mM acetylthiocholine iodide as substrate and 10 µL of 0.5 mM DTNB. After 15 minutes of incubation, the absorbance was measured at 405 nm by using a Synergy HT 96-well plate reader (BioTek, USA). Eserine (0.5 mM) was used as a standard for the positive control. Percent inhibition was calculated as follows:

Inhibition % = Control Text Control x 100

where Test is the activity in the presence of compound of concern, while Control is the activity in the absence of inhibitor.

Urease inhibition assay

A 985-µL reaction mixture containing test compound (0 to 100 µL), urea (850 µL) and phosphate buffer (100 mM, pH 7.4) was prepared to evaluate urease inhibitory activity using a previously reported method (Chapdelaine, Tremblay, Dube, 1978Chapdelaine P, Tremblay RR, Dube JY. r-Nitrophenol-a-D-Glucopyranoside as substrate for measurement of maltase activity in Human Semen. Clin Chem. 1978;24(2):208-211.). The reaction was started by the addition of 15 µL of urease enzyme and allowed to proceed for 60 minutes at 37 °C. A volume of 500 µL of solution A containing 0.5 g phenol and 2.5 mg sodium nitroprusside in 50 mL of distilled water and equi-volume of solution B containing 250 mg sodium hydroxide and 820 µL of 5% sodium hypochlorite in 50 mL of distilled water was used to measure the extent of reaction after incubation for another 30 minutes. Urease activity before inhibition was taken as 100% control activity. The formula used to calculate % inhibition was similar as for AChE.

Statistical analysis

The results are presented as mean ± SEM for triplicate calculations after statistical analysis executed in MS Excel 2010. Minimum inhibitory concentration (MIC) for antibacterial activity and concentration for 50% inhibition (IC50) for AChE inhibition were determined using suitable dilutions for each sample, and the results were obtained using EZ-Fitz software (Perrella Scientific Inc., USA). GraphPad Prism 5 software was used to calculate the IC50values for urease.

Computational studies

Initial processing of proteins and ligand preparation

To study the interaction of ligands in the active site of AChE by using chemoinformatics, the co-crystal 3D-structure recombinant human AChE (Ghersi, Sanchez, 2011Ghersi D, Sanchez R. Beyond structural genomics: computational approaches for the identification of ligand binding sites in protein structures. J Struct Funct Genomics. 2011;12(2):109-17.) in complex with donepezil (PDB entry: 4EY7) was obtained from the RCSB Protein Data Bank (PDB). All protein structures from PDB were further processed prior to docking studies using structure preparation tools included in biopolymer module of SYBYL-X 1.3. Missing hydrogens were added, charges were applied and atom types were assigned according to AMBER 7 FF99 force field followed by the energy minimization using the Powell algorithm with a convergence gradient of 0.5 kcal (mol Å)-1 for 1000 cycles. Moreover, the 3D structures of the inhibitors studied were constructed using the SKETCH module in Sybyl-X 1.3 followed by energy minimization using Tripos force field with Gasteiger-Hückel charge.

Molecular docking

The molecular docking studies were performed with the Surflex-Dock module of SybylX-1.3 (Jain, Dock, 2007Jain AN, Dock D. Robust performance from ligand energetic modeling, ring flexibility, and knowledge-based search. J Comput Aided Mol Des. 2007;21(5):281-306.). The experimentally determined active conformation of donepezil in the AChE active site was used as initial conformation to define the potential binding pocket by protomol (an idealized active site) generation (Chohan et al., 2016aChohan TA, Qian HY, Pan YL, Chen JZ. Molecular simulation studies on the binding selectivity of 2-anilino-4-(thiazol-5-yl)-pyrimidines in complexes with CDK2 and CDK7. Mol. BioSyst. 2016a;12(1):145-161.). The defined active site can be used to generate several putative poses of ligand (Chohan et al., 2016b). These putative poses of ligand were ranked using the Hammerhead scoring function (Holt, Chaires, Trent, 2008Holt PA, Chaires JB, Trent JO. Molecular docking of intercalators and groove-binders to nucleic acids using Autodock and Surflex. J Chem Inf Model. 2008;48(8):1602-1615.). All parameters determining the extent of protomol were kept at default (threshold = 0.50 and bloat=0). Finally, the synthesized compounds were then individually docked in the idealized active site of AChE with the “whole” molecular alignment algorithm, and twenty top-ranked docked poses were finally saved for each inhibitor.

Protein drugs interaction studies

BSA binding interactions using fluorescence measurements

For the determination of the BSA quenching constant, fluorometric titration of a solution of BSA (3 mL, 3 µM) in phosphate buffer (20 mM, pH 7.4) with the synthesized compounds (1 mg/mL in DMSO solvent) was carried out. The solutions were excited at 295 nm and the emission of the BSA solution with and without test compound was recorded at 336 nm and 298 K. For site-selective binding studies, fluorometric titration of BSA solution (3 mL, 3 µM) with and without site markers (ibuprofen or warfarin) was carried out with the solutions of synthesized compounds (1 mg/mL in DMSO). The solutions were scanned using an excitation wavelength of 295 nm at 298 K (Wu et al., 2011Wu XH, Liu JJ, Huang HM, Xue WW, Yao XJ. Interaction studies of aristolochic acid I with human serum albumin and the binding site of aristolochic acid I in subdomain IIA. Int J Biol Macromol. 2011;49(3):343-350.).

Spectral characterization

5-{1-[(4-Chlorophenyl)sulfonyl]piperidin-4-yl}-2-(allylthio)-1,3,4-oxadiazole (7a)

Off-white amorphous solid; Yield: 79 %; M.P.: 102-103 ºC; Mol. formula: C16H18ClN3O3S2; Mol. weight: 399 gmol-1; IR (KBr, υmax , cm-1): 3130 (Ar C-H), 1678 (Al C=C), 1658 (C=N), 1594 (Ar C=C), 1517 (N=O), 1446 (S=O), 1228, 1025 (C-O-C), 1148 (C-N), 655 (C-Cl), 622 (C-S); 1H-NMR (CDCl3, 400 MHz): δ (ppm) 7.86 (d, J = 8.4 Hz, 2H, H-2” & H-6”), 7.50 (d, J = 8.4 Hz, 2H, H-3” & H-5”), 5.99-5.90 (m, 1H, H-2”’), 5.30 (dd, J = 16.8, 0.8 Hz, 1H, Ha-3”’), 5.18 (d, J = 10.4 Hz, 1H, Hb-3”’), 3.81 (d, J = 7.2 Hz, 2H, H-1”’), 3.66-3.63 (m, 2H, He -2’ & He -6’), 2.92-2.85 (m, 1H, H-4’), 2.63 (dt, J = 12.4, 2.8 Hz, 2H, Ha -2’ & Ha -6’), 2.14-2.09 (m, 2H, He -3’ & He -5’), 2.02-1.92 (m, 2H, Ha -3’ & Ha -5’); EI-MS (m/z): 401 [M+2]+, 399 [M]+, 286 [C12H13ClNO3S]+, 284 [C12H13ClN2O2S]•+, 258 [C11H13ClNO2S]+, 175 [C6H4ClO2S]+, 111 [C6H4Cl]+.

5-{1-[(4-Chlorophenyl)sulfonyl]piperidin-4-yl}-2-(ethylthio)-1,3,4-oxadiazole (7b)

Light brown crystalline solid; Yield: 83 %; M.P.: 137-138 ºC; Mol. formula: C15H18ClN3O3S2; Mol. weight: 387 gmol-1; IR (KBr, υmax , cm-1): 3030 (Ar C-H), 1688 (C=N), 1599 (Ar C=C), 1519 (N=O), 1466 (S=O), 1220, 1029 (C-O-C), 1168 (C-N), 658 (C-Cl), 632 (C-S); 1H-NMR (CDCl3, 400 MHz): δ (ppm) 7.86 (d, J = 8.4 Hz, 2H, H-2” & H-6”), 7.49 (d, J = 8.4 Hz, 2H, H-3” & H-5”), 3.66-3.63 (m, 2H, He -2’ & He -6’), 3.20 (q, J = 7.6 Hz, 2H, H-1”’), 2.93-2.85 (m, 1H, H-4’), 2.62 (dt, J = 12.4, 2.8 Hz, 2H, Ha -2’ & Ha -6’), 2.14-2.09 (m, 2H, He -3’ & He -5’), 2.01-1.94 (m, 2H, Ha -3’ & Ha -5’), 1.43 (t, J = 7.6 Hz, 3H, H-2”’); EI-MS (m/z): 389 [M+2]+, 387 [M]+, 286 [C12H13ClNO3S]+, 284 [C12H13ClN2O2S]•+, 258 [C11H13ClNO2S]+, 175 [C6H4ClO2S]+, 111 [C6H4Cl]+.

5-{1-[(4-Chlorophenyl)sulfonyl]piperidin-4-yl}-2-[(1-propyl)thio]-1,3,4-oxadiazole (7c)

White amorphous solid; Yield: 85 %; M.P.: 111-112 ºC; Mol. formula: C16H20ClN3O3S2; Mol. weight: 401 gmol-1; IR (KBr, υmax , cm-1): 3130 (Ar C-H), 1682 (C=N), 1565 (Ar C=C), 1520 (N=O), 1386 (S=O), 1228, 1026 (C-O-C), 1158 (C-N), 678 (C-Cl), 612 (C-S); 1H-NMR (CDCl3, 400 MHz): δ (ppm) 7.67 (d, J = 8.4 Hz, 2H, H-2” & H-6”), 7.51 (d, J = 8.8 Hz, 2H, H-3” & H-5”), 3.70-3.67 (m, 2H, He -2’ & He -6’), 3.18 (t, J = 7.2 Hz, 2H, H-1”’), 2.71-2.67 (m, 1H, H-4’), 2.56 (dt, J = 12.0, 2.4 Hz, 2H, Ha -2’ & Ha -6’), 2.13-2.09 (m, 2H, He -3’ & He -5’), 1.92 (dq, J = 10.8, 3.6 Hz, 2H, Ha -3’ & Ha -5’), 1.34 (sex, J = 7.2 Hz, 3H, H-2”’), 1.03 (t, J = 7.6 Hz, 3H, H-3”’); EI-MS (m/z): 403 [M+2]+, 401 [M]+, 286 [C12H13ClNO3S]+, 284 [C12H13ClN2O2S]•+, 258 [C11H13ClNO2S]+, 175 [C6H4ClO2S]+, 111 [C6H4Cl]+.

5-{1-[(4-Chlorophenyl)sulfonyl]piperidin-4-yl}-2-[(2-propyl)thio]-1,3,4-oxadiazole (7d)

Off-white amorphous solid; Yield: 77 %; M.P.: 156-157 ºC; Mol. formula: C16H20ClN3O3S2; Mol. weight: 401 gmol-1; IR (KBr, υmax , cm-1): 3067 (Ar C-H), 1673 (C=N), 1582 (Ar C=C), 1525 (N=O), 1476 (S=O), 1170, 1026 (C-O-C), 1142 (C-N), 662 (C-Cl), 638 (C-S); 1H-NMR (CDCl3, 400 MHz): δ (ppm) 7.69 (d, J = 8.4 Hz, 2H, H-2” & H-6”), 7.51 (d, J = 8.4 Hz, 2H, H-3” & H-5”), 3.85 (sep, J = 6.8 Hz, 1H, H-1”’), 3.71-3.64 (m, 2H, He -2’ & He -6’), 2.72-2.65 (m, 1H, H-4’), 2.56 (dt, J = 12.0, 2.4 Hz, 2H, Ha -2’ & Ha -6’), 2.13-2.09 (m, 2H, He -3’ & He -5’), 1.99-1.86 (m, 2H, Ha -3’ & Ha -5’), 1.45 (d, J = 6.8 Hz, 6H, H-2”’ & H-3”’); EI-MS (m/z): 403 [M+2]+, 401 [M]+, 286 [C12H13ClNO3S]+, 284 [C12H13ClN2O2S]•+, 258 [C11H13ClNO2S]+, 175 [C6H4ClO2S]+, 111 [C6H4Cl]+.

5-{1-[(4-Chlorophenyl)sulfonyl]piperidin-4-yl}-2-[(1-butyl)thio]-1,3,4-oxadiazole (7e)

White amorphous solid; Yield: 76 %; M.P.: 113-114 ºC; Mol. formula: C17H22ClN3O3S2; Mol. weight: 415 gmol-1; IR (KBr, υmax , cm-1): 3080 (Ar C-H), 1688 (C=N), 1578 (Ar C=C), 1545 (N=O), 1456 (S=O), 1235, 1019 (C-O-C), 1161 (C-N), 660 (C-Cl), 622 (C-S); 1H-NMR (CDCl3, 400 MHz): δ (ppm) 7.68 (d, J = 8.4 Hz, 2H, H-2” & H-6”), 7.50 (d, J = 8.4 Hz, 2H, H-3” & H-5”), 3.67-3.62 (m, 2H, He -2’ & He -6’), 3.19 (t, J = 7.6 Hz, 2H, H-1”’), 2.89-2.84 (m, 1H, H-4’), 2.63 (dt, J = 12.4, 2.8 Hz, 2H, Ha -2’ & Ha -6’), 2.14-2.10 (m, 2H, He -3’ & He -5’), 2.02-1.92 (m, 2H, Ha -3’ & Ha -5’), 1.74 (qui, J = 7.6 Hz, 2H, H-2”’), 1.44 (sex, J = 7.2 Hz, 2H, H-3”’), 0.92 (t, J = 7.6 Hz, 3H, H-4”’); EI-MS (m/z): 417 [M+2]+, 415 [M]+, 286 [C12H13ClNO3S]+, 284 [C12H13ClN2O2S]•+, 258 [C11H13ClNO2S]+, 175 [C6H4ClO2S]+, 111 [C6H4Cl]+, 57 [C4H9]+.

5-{1-[(4-Chlorophenyl)sulfonyl]piperidin-4-yl}-2-[(2-butyl)thio]-1,3,4-oxadiazole (7f)

White amorphous solid; Yield: 80 %; M.P.: 105-106 ºC; Mol. formula: C17H22ClN3O3S2; Mol. weight: 415 gmol-1; IR (KBr, υmax , cm-1): 3036 (Ar C-H), 1685 (C=N), 1592 (Ar C=C), 1514 (N=O), 1465 (S=O), 1225, 1022 (C-O-C), 1163 (C-N), 653 (C-Cl), 631 (C-S); 1H-NMR (CDCl3, 400 MHz): δ (ppm) 7.70 (d, J = 8.4 Hz, 2H, H-2” & H-6”), 7.52 (d, J = 8.4 Hz, 2H, H-3” & H-5”), 4.96-4.77 (m, 1H, H-1”’), 3.70-3.68 (m, 2H, He -2’ & He -6’), 2.70-2.68 (m, 1H, H-4’), 2.57 (dt, J = 12.4, 2.8 Hz, 2H, Ha -2’ & Ha -6’), 2.13-2.10 (m, 2H, He -3’ & He -5’), 2.01-1.96 (m, 2H, H-2”’), 1.97-1.87 (m, 2H, Ha -3’ & Ha -5’), 1.44 (d, J = 6.8 Hz, 3H, H-4”’), 1.01 (t, J = 7.2 Hz, 3H, H-3”’); EI-MS (m/z): 417 [M+2]+, 415 [M]+, 286 [C12H13ClNO3S]+, 284 [C12H13ClN2O2S]•+, 258 [C11H13ClNO2S]+, 175 [C6H4ClO2S]+, 111 [C6H4Cl]+, 57 [C4H9]+.

5-{1-[(4-Chlorophenyl)sulfonyl]piperidin-4-yl}-2-[(1-pentyl)thio]-1,3,4-oxadiazole (7g)

White amorphous solid; Yield: 84 %; M.P.: 115-116 ºC; Mol. formula: C18H24ClN3O3S2; Mol. weight: 429 gmol-1; IR (KBr, υmax , cm-1): 3026 (Ar C-H), 1675 (C=N), 1582 (Ar C=C), 1534 (N=O), 1445 (S=O), 1215, 1042 (C-O-C), 1193 (C-N), 673 (C-Cl), 641 (C-S); 1H-NMR (CDCl3, 400 MHz): δ (ppm) 7.69 (d, J = 8.4 Hz, 2H, H-2” & H-6”), 7.51 (d, J = 8.4 Hz, 2H, H-3” & H-5”), 3.66-3.63 (m, 2H, He -2’ & He -6’), 3.18 (t, J = 7.6 Hz, 2H, H-1”’), 2.89-2.84 (m, 1H, H-4’), 2.63 (dt, J = 12.4, 2.8 Hz, 2H, Ha -2’ & Ha -6’), 2.14-2.10 (m, 2H, He -3’ & He -5’), 2.02-1.95 (m, 2H, Ha -3’ & Ha -5’), 1.75 (qui, J = 7.6 Hz, 2H, H-2”’), 1.41-1.29 (m, 4H, H-3”’ & H-4”’), 0.88 (t, J = 7.2 Hz, 3H, H-5”’); EI-MS (m/z): 431 [M+2]+, 429 [M]+, 286 [C12H13ClNO3S]+, 284 [C12H13ClN2O2S]•+, 258 [C11H13ClNO2S]+, 175 [C6H4ClO2S]+, 111 [C6H4Cl]+, 71 [C5H11]+.

5-{1-[(4-Chlorophenyl)sulfonyl]piperidin-4-yl}-2-[(2-pentyl)thio]-1,3,4-oxadiazole (7h)

White amorphous solid; Yield: 78 %; M.P.: 98-99 ºC; Mol. formula: C18H24ClN3O3S2; Mol. weight: 429 gmol-1; IR (KBr, υmax , cm-1): 3126 (Ar C-H), 1685 (C=N), 1682 (Ar C=C), 1530 (N=O), 1385 (S=O), 1225, 1031 (C-O-C), 1143 (C-N), 773 (C-Cl), 631 (C-S); 1H-NMR (CDCl3, 400 MHz): δ (ppm) 7.70 (d, J = 8.4 Hz, 2H, H-2” & H-6”), 7.52 (d, J = 8.4 Hz, 2H, H-3” & H-5”), 4.95-4.80 (m, 1H, H-1”’), 3.71-3.67 (m, 2H, He -2’ & He -6’), 2.69-2.65 (m, 1H, H-4’), 2.57 (dt, J = 12.4, 2.8 Hz, 2H, Ha -2’ & Ha -6’), 2.13-2.10 (m, 2H, He -3’ & He -5’), 2.02-1.97 (m, 2H, H-2”’), 1.97-1.87 (m, 2H, Ha -3’ & Ha -5’), 1.75-1.61 (m, 2H, H-3”’), 1.45 (d, J = 7.2 Hz, 3H, H-5”’), 0.92 (t, J = 7.2 Hz, 3H, H-4”’); EI-MS (m/z): 431 [M+2]+, 429 [M]+, 286 [C12H13ClNO3S]+, 284 [C12H13ClN2O2S]•+, 258 [C11H13ClNO2S]+, 175 [C6H4ClO2S]+, 111 [C6H4Cl]+, 71 [C5H11]+.

5-{1-[(4-Chlorophenyl)sulfonyl]piperidin-4-yl}-2-[(1-heptyl)thio]-1,3,4-oxadiazole (7i)

Off-white amorphous solid; Yield: 75 %; M.P.: 123-124 ºC; Mol. formula: C20H28ClN3O3S2; Mol. weight: 457 gmol-1; IR (KBr, υmax , cm-1): 3122 (Ar C-H), 1682 (C=N), 1688 (Ar C=C), 1538 (N=O), 1382 (S=O), 1228, 1036 (C-O-C), 1148 (C-N), 772 (C-Cl), 633 (C-S); 1H-NMR (CDCl3, 400 MHz): δ (ppm) 7.68 (d, J = 8.4 Hz, 2H, H-2” & H-6”), 7.49 (d, J = 8.4 Hz, 2H, H-3” & H-5”), 3.67-3.62 (m, 2H, He -2’ & He -6’), 3.18 (t, J = 7.2 Hz, 2H, H-1”’), 2.89-2.84 (m, 1H, H-4’), 2.63 (dt, J = 12.4, 2.8 Hz, 2H, Ha -2’ & Ha -6’), 2.14-2.10 (m, 2H, He -3’ & He -5’), 2.02-1.95 (m, 2H, Ha -3’ & Ha -5’), 1.76 (qui, J = 7.2 Hz, 2H, H-2”’), 1.40 (qui, J = 7.6 Hz, 2H, H-3”’), 1.31-1.26 (m, 6H, H-4”’ to H-6”’), 0.85 (t, J = 6.8 Hz, 3H, H-7”’); EI-MS (m/z): 459 [M+2]+, 457 [M]+, 286 [C12H13ClNO3S]+, 284 [C12H13ClN2O2S]•+, 258 [C11H13ClNO2S]+, 175 [C6H4ClO2S]+, 111 [C6H4Cl]+, 99 [C7H15]+.

5-{1-[(4-Chlorophenyl)sulfonyl]piperidin-4-yl}-2-[(1-octyl)thio]-1,3,4-oxadiazole (7j)

Off-white amorphous solid; Yield: 86 %; M.P.: 127-128 ºC; Mol. formula: C21H30ClN3O3S2; Mol. weight: 471 gmol-1; IR (KBr, υmax , cm-1): 3129 (Ar C-H), 1688 (C=N), 1680 (Ar C=C), 1540 (N=O), 1389 (S=O), 1238, 1038 (C-O-C), 1138 (C-N), 782 (C-Cl), 639 (C-S); 1H-NMR (CDCl3, 400 MHz): δ (ppm) 7.68 (d, J = 8.4 Hz, 2H, H-2” & H-6”), 7.49 (d, J = 8.4 Hz, 2H, H-3” & H-5”), 3.66-3.63 (m, 2H, He -2’ & He -6’), 3.18 (t, J = 7.6 Hz, 2H, H-1”’), 2.89-2.84 (m, 1H, H-4’), 2.63 (dt, J = 12.4, 2.8 Hz, 2H, Ha -2’ & Ha -6’), 2.14-2.09 (m, 2H, He -3’ & He -5’), 2.02-1.93 (m, 2H, Ha -3’ & Ha -5’), 1.75 (qui, J = 7.2 Hz, 2H, H-2”’), 1.40 (qui, J = 7.6 Hz, 2H, H-3”’), 1.27-1.24 (m, 8H, H-4”’ to H-7”’), 0.85 (t, J = 6.8 Hz, 3H, H-8”’); 13C-NMR (CDCl3, 100 MHz): δ (ppm) 168.3 (C-5), 164.9 (C-2), 139.5 (C-1”), 134.8 (C-4”), 129.5 (C-3” & C-5”), 129.0 (C-2” & C-6”), 45.0 (C-2’ & C-6’), 32.5 (C-6”’), 32.2 (C-4’), 31.7 (C-5”’), 29.1 (C-4”’), 29.0 (C-3”’), 28.9 (C-2”’), 28.5 (C-1”’), 28.3 (C-3’ & C-5’), 22.6 (C-7”’), 14.0 (C-8”’); EI-MS (m/z): 473 [M+2]+, 471 [M]+, 286 [C12H13ClNO3S]+, 284 [C12H13ClN2O2S]•+, 258 [C11H13ClNO2S]+, 175 [C6H4ClO2S]+, 111 [C6H4Cl]+, 113 [C8H17]+.

5-{1-[(4-Chlorophenyl)sulfonyl]piperidin-4-yl}-2-(benzylthio)-1,3,4-oxadiazole (7k)

Off-white amorphous solid; Yield: 76 %; M.P.: 131-132 ºC; Mol. formula: C20H20ClN3O3S2; Mol. weight: 429 gmol-1; IR (KBr, υmax , cm-1): 3134 (Ar C-H), 1673 (C=N), 1610 (Ar C=C), 1524 (N=O), 1323 (S=O), 1228, 1012 (C-O-C), 1148 (C-N), 761 (C-Cl), 624 (C-S); 1H-NMR (CDCl3, 400 MHz): δ (ppm) 7.68 (d, J = 8.8 Hz, 2H, H-2” & H-6”), 7.50 (d, J = 8.4 Hz, 2H, H-3” & H-5”), 7.37 (br.d, J = 6.4 Hz, 2H, H-2”’ & H-6”’), 7.30-7.28 (m, 3H, H-3”’ to H-5”’), 4.04 (s, 2H, H-7”’), 3.64-3.61 (m, 2H, He -2’ & He -6’), 2.88-2.83 (m, 1H, H-4’), 2.62 (dt, J = 12.4, 2.8 Hz, 2H, Ha -2’ & Ha -6’), 2.12-2.08 (m, 2H, He -3’ & He -5’), 2.00-1.93 (m, 2H, Ha -3’ & Ha -5’); EI-MS (m/z): 431 [M+2]+, 429 [M]+, 286 [C12H13ClNO3S]+, 284 [C12H13ClN2O2S]•+, 258 [C11H13ClNO2S]+, 175 [C6H4ClO2S]+, 111 [C6H4Cl]+, 91 [C7H7]+, 65 [C5H5]+.

5-{1-[(4-Chlorophenyl)sulfonyl]piperidin-4-yl}-2-[(2-methylbenzyl)thio]-1,3,4-oxadiazole (7l)

White amorphous solid; Yield: 77 %; M.P.: 125-126 ºC; Mol. formula: C21H22ClN3O3S2; Mol. weight: 463 gmol-1; IR (KBr, υmax , cm-1): 3138 (Ar C-H), 1676 (C=N), 1618 (Ar C=C), 1525 (N=O), 1327 (S=O), 1224, 1017 (C-O-C), 1142 (C-N), 769 (C-Cl), 628 (C-S); 1H-NMR (CDCl3, 400 MHz): δ (ppm) 7.68 (d, J = 8.4 Hz, 2H, H-2” & H-6”), 7.50 (d, J = 8.4 Hz, 2H, H-3” & H-5”), 7.32 (d, J = 7.6 Hz, 1H, H-6”’), 7.20-7.12 (m, 3H, H-3”’ to H-5”’), 4.44 (s, 2H, H-7”’), 3.66-3.63 (m, 2H, He -2’ & He -6’), 2.89-2.84 (m, 1H, H-4’), 2.63 (dt, J = 12.4, 2.8 Hz, 2H, Ha -2’ & Ha -6’), 2.13-2.09 (m, 2H, He -3’ & He -5’), 2.01-1.94 (m, 2H, Ha -3’ & Ha -5’), 2.39 (s, 3H, CH3-2”’); EI-MS (m/z): 465 [M+2]+, 463 [M]+, 286 [C12H13ClNO3S]+, 284 [C12H13ClN2O2S]•+, 258 [C11H13ClNO2S]+, 175 [C6H4ClO2S]+, 111 [C6H4Cl]+, 105 [C8H9]+, 65 [C5H5]+.

5-{1-[(4-Chlorophenyl)sulfonyl]piperidin-4-yl}-2-[(4-fluorobenzyl)thio]-1,3,4-oxadiazole (7m)

White amorphous solid; Yield: 75 %; M.P.: 146-147 ºC; Mol. formula: C20H19ClFN3O3S2; Mol. weight: 467 gmol-1; IR (KBr, υmax , cm-1): 3137 (Ar C-H), 1672 (C=N), 1619 (Ar C=C), 1522 (N=O), 1326 (S=O), 1227, 1013 (C-O-C), 1147 (C-N), 1056 (C-F), 720 (C-Cl), 626 (C-S); 1H-NMR (CDCl3, 400 MHz): δ (ppm) 7.68 (d, J = 8.4 Hz, 2H, H-2” & H-6”), 7.49 (d, J = 8.8 Hz, 2H, H-3” & H-5”), 7.37 (dis. dd, J(b,a & b,19F) = 8.8, 5.6 Hz, 2Hb, H-2”’ & H-6”’), 6.98 (br.t, J(a,b & a,19F) = 8.8 Hz, 2Ha, H-3”’ & H-5”’), 4.38 (s, 2H, H-7”’), 3.66-3.62 (m, 2H, He -2’ & He -6’), 2.88-2.83 (m, 1H, H-4’), 2.62 (dt, J = 12.4, 2.8 Hz, 2H, Ha -2’ & Ha -6’), 2.12-2.08 (m, 2H, He -3’ & He -5’), 1.97-1.93 (m, 2H, Ha -3’ & Ha -5’); EI-MS (m/z): 469 [M+2]+, 467 [M]+, 286 [C12H13ClNO3S]+, 284 [C12H13ClN2O2S]•+, 258 [C11H13ClNO2S]+, 175 [C6H4ClO2S]+, 111 [C6H4Cl]+, 109 [C7H6F]+, 65 [C5H5]+.

5-{1-[(4-Chlorophenyl)sulfonyl]piperidin-4-yl}-2-[(2-chlorobenzyl)thio]-1,3,4-oxadiazole (7n)

White amorphous solid; Yield: 81 %; M.P.: 121-122 ºC; Mol. formula: C20H19Cl2N3O3S2; Mol. weight: 483 gmol-1; IR (KBr, υmax, cm-1): 3127 (Ar C-H), 1682 (C=N), 1609 (Ar C=C), 1532 (N=O), 1396 (S=O), 1237, 1023 (C-O-C), 1167 (C-N), 780 (C-Cl), 646 (C-S); 1H-NMR (CDCl3, 400 MHz): δ (ppm) 7.68 (d, J = 8.8 Hz, 2H, H-2” & H-6”), 7.49 (d, J = 8.4 Hz, 2H, H-3” & H-5”), 7.53 (dd, J = 7.2, 1.2 Hz, 1H, H-6”’), 7.38 (dd, J = 8.0, 0.8 Hz, 1H, H-3”’), 7.23-7.19 (m, 2H, H-4”’ & H-5”’), 4.51 (s, 2H, H-7”’), 3.65-3.62 (m, 2H, He -2’ & He -6’), 2.87-2.82 (m, 1H, H-4’), 2.62 (dt, J = 12.4, 2.8 Hz, 2H, Ha -2’ & Ha -6’), 2.12-2.08 (m, 2H, He -3’ & He -5’), 2.00-1.91 (m, 2H, Ha -3’ & Ha -5’); 13C-NMR (CDCl3, 100 MHz): δ (ppm) 168.7 (C-5), 164.1 (C-2), 139.5 (C-1”), 134.8 (C-4”), 134.4 (C-1”’), 133.6 (C-2”’), 131.4 (C-4”’), 129.7 (C-5”’), 129.6 (C-3”’), 129.5 (C-3” & C-5”), 128.9 (C-2” & C-6”), 127.0 (C-6”’), 44.9 (C-2’ & C-6’), 34.5 (C-7”’), 32.2 (C-4’), 28.2 (C-3’ & C-5’); EI-MS (m/z): 487 [M+4]+, 485 [M+2]+, 483 [M]+, 286 [C12H13ClNO3S]+, 284 [C12H13ClN2O2S]•+, 258 [C11H13ClNO2S]+, 175 [C6H4ClO2S]+, 125 [C7H6Cl]+, 111 [C6H4Cl]+, 65 [C5H5]+.

5-{1-[(4-Chlorophenyl)sulfonyl]piperidin-4-yl}-2-[(3-chlorobenzyl)thio]-1,3,4-oxadiazole (7o)

White amorphous solid; Yield: 82 %; M.P.: 176-177 ºC; Mol. formula: C20H19Cl2N3O3S2; Mol. weight: 483 gmol-1; IR (KBr, υmax, cm-1): 3122 (Ar C-H), 1680 (C=N), 1600 (Ar C=C), 1538 (N=O), 1398 (S=O), 1239, 1026 (C-O-C), 1168 (C-N), 782 (C-Cl), 643 (C-S); 1H-NMR (CDCl3, 400 MHz): δ (ppm) 7.68 (d, J = 8.4 Hz, 2H, H-2” & H-6”), 7.49 (d, J = 8.4 Hz, 2H, H-3” & H-5”), 7.37 (s, 1H, H-2”’), 7.29-7.23 (m, 3H, H-4”’ to H-6”’), 4.36 (s, 2H, H-7”’), 3.66-3.63 (m, 2H, He -2’ & He -6’), 2.87-2.83 (m, 1H, H-4’), 2.62 (dt, J = 12.0, 2.4 Hz, 2H, Ha -2’ & Ha -6’), 2.12-2.08 (m, 2H, He -3’ & He -5’), 1.97-1.93 (m, 2H, Ha -3’ & Ha -5’); EI-MS (m/z): 487 [M+4]+, 485 [M+2]+, 483 [M]+, 286 [C12H13ClNO3S]+, 284 [C12H13ClN2O2S]•+, 258 [C11H13ClNO2S]+, 175 [C6H4ClO2S]+, 125 [C7H6Cl]+, 111 [C6H4Cl]+, 65 [C5H5]+.

5-{1-[(4-Chlorophenyl)sulfonyl]piperidin-4-yl}-2-[(4-chlorobenzyl)thio]-1,3,4-oxadiazole (7p)

White amorphous solid; Yield: 85 %; M.P.: 182-183 ºC; Mol. formula: C20H19Cl2N3O3S2; Mol. weight: 483 gmol-1; IR (KBr, υmax, cm-1): 3125 (Ar C-H), 1683 (C=N), 1608 (Ar C=C), 1534 (N=O), 1392 (S=O), 1230, 1028 (C-O-C), 1166 (C-N), 780 (C-Cl), 642 (C-S); 1H-NMR (CDCl3, 400 MHz): δ (ppm) 7.67 (d, J = 8.4 Hz, 2H, H-2” & H-6”), 7.48 (d, J = 8.8 Hz, 2H, H-3” & H-5”), 7.35 (d, J = 8.4 Hz, 2H, H-2”’ & H-6”’), 7.28 (d, J = 8.4 Hz, 2H, H-3”’ & H-5”’), 4.37 (s, 2H, H-7”’), 3.67-3.62 (m, 2H, He -2’ & He -6’), 2.87-2.81 (m, 1H, H-4’), 2.61 (dt, J = 12.4, 2.8 Hz, 2H, Ha -2’ & Ha -6’), 2.13-2.08 (m, 2H, He -3’ & He -5’), 1.99-1.93 (m, 2H, Ha -3’ & Ha -5’); EI-MS (m/z): 487 [M+4]+, 485 [M+2]+, 483 [M]+, 286 [C12H13ClNO3S]+, 284 [C12H13ClN2O2S]•+, 258 [C11H13ClNO2S]+, 175 [C6H4ClO2S]+, 125 [C7H6Cl]+, 111 [C6H4Cl]+, 65 [C5H5]+.

5-{1-[(4-Chlorophenyl)sulfonyl]piperidin-4-yl}-2-[(2-bromobenzyl)thio]-1,3,4-oxadiazole (7q)

White amorphous solid; Yield: 84 %; M.P.: 118-119 ºC; Mol. formula: C20H19ClBrN3O3S2; Mol. weight: 526 gmol-1; IR (KBr, υmax, cm-1): 3125 (Ar C-H), 1683 (C=N), 1602 (Ar C=C), 1534 (N=O), 1390 (S=O), 1231, 1023 (C-O-C), 1169 (C-N), 782 (C-Cl), 647 (C-S), 625 (C-Br); 1H-NMR (CDCl3, 400 MHz): δ (ppm) 7.68 (d, J = 8.4 Hz, 2H, H-2” & H-6”), 7.55 (d, J = 8.0 Hz, 1H, H-3”’), 7.49 (d, J = 8.4 Hz, 2H, H-3” & H-5”), 7.25-7.21 (m, 2H, H-5”’ & H-6”’), 7.14 (dt, J = 7.6, 1.2 Hz, 1H, H-4”’), 4.52 (s, 2H, H-7”’), 3.65-3.62 (m, 2H, He -2’ & He -6’), 2.87-2.82 (m, 1H, H-4’), 2.62 (dt, J = 12.4, 2.8 Hz, 2H, Ha -2’ & Ha -6’), 2.12-2.08 (m, 2H, He -3’ & He -5’), 1.99-1.90 (m, 2H, Ha -3’ & Ha -5’); EI-MS (m/z): 530 [M+4]+, 528 [M+2]+, 526 [M]+, 286 [C12H13ClNO3S]+, 284 [C12H13ClN2O2S]•+, 258 [C11H13ClNO2S]+, 175 [C6H4ClO2S]+, 169 [C7H6Br]+, 111 [C6H4Cl]+, 65 [C5H5]+.

5-{1-[(4-Chlorophenyl)sulfonyl]piperidin-4-yl}-2-[(4-bromobenzyl)thio]-1,3,4-oxadiazole (7r)

White amorphous solid; Yield: 83 %; M.P.: 173-174 ºC; Mol. formula: C20H19ClBrN3O3S2; Mol. weight: 526 gmol-1; IR (KBr, υmax, cm-1): 3128 (Ar C-H), 1688 (C=N), 1605 (Ar C=C), 1539 (N=O), 1393 (S=O), 1234, 1027 (C-O-C), 1166 (C-N), 722 (C-Cl), 649 (C-S), 629 (C-Br); 1H-NMR (CDCl3, 400 MHz): δ (ppm) 7.67 (d, J = 8.4 Hz, 2H, H-2” & H-6”), 7.48 (d, J = 8.8 Hz, 2H, H-3” & H-5”), 7.43 (d, J = 8.0 Hz, 2H, H-2”’ & H-6”’), 7.28 (d, J = 8.4 Hz, 2H, H-3”’ & H-5”’), 4.35 (s, 2H, H-7”’), 3.67-3.62 (m, 2H, He -2’ & He -6’), 2.87-2.81 (m, 1H, H-4’), 2.61 (dt, J = 12.4, 2.8 Hz, 2H, Ha -2’ & Ha -6’), 2.13-2.08 (m, 2H, He -3’ & He -5’), 1.99-1.93 (m, 2H, Ha -3’ & Ha -5’); EI-MS (m/z): 530 [M+4]+, 528 [M+2]+, 526 [M]+, 286 [C12H13ClNO3S]+, 284 [C12H13ClN2O2S]•+, 258 [C11H13ClNO2S]+, 175 [C6H4ClO2S]+, 169 [C7H6Br]+, 111 [C6H4Cl]+, 65 [C5H5]+.

5-{1-[(4-Chlorophenyl)sulfonyl]piperidin-4-yl}-2-[(2-phenylethyl)thio]-1,3,4-oxadiazole (7s)

Off-white amorphous solid; Yield: 82 %; M.P.: 108-109 ºC; Mol. formula: C21H22ClN3O3S2; Mol. weight: 463 gmol-1; IR (KBr, υmax , cm-1): 3155 (Ar C-H), 1643 (C=N), 1592 (Ar C=C), 1544 (N=O), 1450 (S=O), 1241, 1033 (C-O-C), 1182 (C-N), 680 (C-Cl), 624 (C-S); 1H-NMR (CDCl3, 400 MHz): δ (ppm) 7.68 (d, J = 8.4 Hz, 2H, H-2” & H-6”), 7.49 (d, J = 8.4 Hz, 2H, H-3” & H-5”), 7.28 (d, J = 7.2 Hz, 2H, H-2”’ & H-6”’), 7.23-7.20 (m, 3H, H-3”’ to H-5”’), 3.66-3.63 (m, 2H, He -2’ & He -6’), 3.43 (t, J = 7.6 Hz, 2H, H-8”’), 3.08 (t, J = 7.6 Hz, 2H, H-7”’), 2.89-2.84 (m, 1H, H-4’), 2.66-2.61 (m, 2H, Ha -2’ & Ha -6’), 2.14-2.10 (m, 2H, He -3’ & He -5’), 2.00-1.95 (m, 2H, Ha -3’ & Ha -5’); EI-MS (m/z): 465 [M+2]+, 463 [M]+, 286 [C12H13ClNO3S]+, 284 [C12H13ClN2O2S]•+, 258 [C11H13ClNO2S]+, 175 [C6H4ClO2S]+, 111 [C6H4Cl]+, 105 [C8H9]+, 77 [C6H5]+, 65 [C5H5]+, 51 [C4H3]+.

5-{1-[(4-Chlorophenyl)sulfonyl]piperidin-4-yl}-2-[(3-phenylpropyl)thio]-1,3,4-oxadiazole (7t)

White amorphous solid; Yield: 80 %; M.P.: 116-117 ºC; Mol. formula: C22H24ClN3O3S2; Mol. weight: 477 gmol-1; IR (KBr, υmax , cm-1): 3152 (Ar C-H), 1642 (C=N), 1590 (Ar C=C), 1542 (N=O), 1452 (S=O), 1244, 1032 (C-O-C), 1180 (C-N), 684 (C-Cl), 626 (C-S); 1H-NMR (CDCl3, 300 MHz): δ (ppm) 7.69 (d, J = 8.4 Hz, 2H, H-2” & H-6”), 7.49 (d, J = 8.4 Hz, 2H, H-3” & H-5”), 7.29-7.15 (m, 5H, H-2”’ to H-6”’), 3.66-3.63 (m, 2H, He -2’ & He -6’), 3.20 (t, J = 7.2 Hz, 2H, H-9”’), 2.89-2.84 (m, 1H, H-4’), 2.75 (t, J = 7.5 Hz, 2H, H-7”’), 2.66-2.61 (m, 2H, Ha -2’ & Ha -6’), 2.16-2.12 (m, 2H, He -3’ & He -5’), 2.11 (qui, J = 7.2 Hz, 2H, H-8”’), 2.00-1.95 (m, 2H, Ha -3’ & Ha -5’); EI-MS (m/z): 479 [M+2]+, 477 [M]+, 286 [C12H13ClNO3S]+, 284 [C12H13ClN2O2S]•+, 258 [C11H13ClNO2S]+, 175 [C6H4ClO2S]+, 119 [C9H11]+, 111 [C6H4Cl]+, 105 [C8H9]+, 77 [C6H5]+, 65 [C5H5]+, 51 [C4H3]+.

5-{1-[(4-Chlorophenyl)sulfonyl]piperidin-4-yl}-2-[(3-nitrobenzyl)thio]-1,3,4-oxadiazole (7u)

White amorphous solid; Yield: 74 %; M.P.: 122-123 ºC; Mol. formula: C20H19ClN4O5S2; Mol. weight: 494 gmol-1; IR (KBr, υmax , cm-1): 3157 (Ar C-H), 1645 (C=N), 1592 (Ar C=C), 1548 (N=O), 1457 (S=O), 1246, 1038 (C-O-C), 1183 (C-N), 689 (C-Cl), 623 (C-S); 1H-NMR (CDCl3, 400 MHz): δ (ppm) 8.26 (s, 1H, H-2”’), 8.13 (d, J = 8.4 Hz, 1H, H-4”’), 7.78 (d, J = 7.6 Hz, 1H, H-6”’), 7.68 (d, J = 8.8 Hz, 2H, H-2” & H-6”), 7.49 (d, J = 8.0 Hz, 2H, H-3” & H-5”), 7.47 (t, J = 7.2 Hz, 1H, H-5”’), 4.47 (s, 2H, H-7”’), 3.68-3.65 (m, 2H, He -2’ & He -6’), 2.88-2.82 (m, 1H, H-4’), 2.59 (dt, J = 12.0, 2.8 Hz, 2H, Ha -2’ & Ha -6’), 2.13-2.08 (m, 2H, He -3’ & He -5’), 1.98-1.90 (m, 2H, Ha -3’ & Ha -5’); EI-MS (m/z): 496 [M+2]+, 494 [M]+, 286 [C12H13ClNO3S]+, 284 [C12H13ClN2O2S]•+, 258 [C11H13ClNO2S]+, 175 [C6H4ClO2S]+, 136 [C7H6NO2]+, 111 [C6H4Cl]+, 65 [C5H5]+.

5-{1-[(4-Chlorophenyl)sulfonyl]piperidin-4-yl}-2-[(4-nitrobenzyl)thio]-1,3,4-oxadiazole (7v)

Off-white amorphous solid; Yield: 79 %; M.P.: 166-167 ºC; Mol. formula: C20H19ClN4O5S2; Mol. weight: 494 gmol-1; IR (KBr, υmax , cm-1): 3156 (Ar C-H), 1648 (C=N), 1594 (Ar C=C), 1546 (N=O), 1458 (S=O), 1242, 1034 (C-O-C), 1187 (C-N), 680 (C-Cl), 625 (C-S); 1H-NMR (CDCl3, 400 MHz): δ (ppm) 7.81 (d, J = 7.6 Hz, 1H, H-3”’ & H-5”’), 7.68 (d, J = 8.8 Hz, 2H, H-2” & H-6”), 7.64 (t, J = 7.6 Hz, 1H, H-2”’ & H-6”’), 7.49 (d, J = 8.0 Hz, 2H, H-3” & H-5”), 4.46 (s, 2H, H-7”’), 3.69-3.65 (m, 2H, He -2’ & He -6’), 2.86-2.81 (m, 1H, H-4’), 2.61 (dt, J = 12.0, 2.4 Hz, 2H, Ha -2’ & Ha -6’), 2.14-2.09 (m, 2H, He -3’ & He -5’), 1.97-1.92 (m, 2H, Ha -3’ & Ha -5’); EI-MS (m/z): 496 [M+2]+, 494 [M]+, 286 [C12H13ClNO3S]+, 284 [C12H13ClN2O2S]•+, 258 [C11H13ClNO2S]+, 175 [C6H4ClO2S]+, 136 [C7H6NO2]+, 111 [C6H4Cl]+, 65 [C5H5]+.

5-{1-[(4-Chlorophenyl)sulfonyl]piperidin-4-yl}-2-[(2-iodoethyl)thio]-1,3,4-oxadiazole (7w)

White amorphous solid; Yield: 78 %; M.P.: 211-212 ºC; Mol. formula: C15H17ClIN3O3S2; Mol. weight: 513 gmol-1; IR (KBr, υmax , cm-1): 3150 (Ar C-H), 1640 (C=N), 1598 (Ar C=C), 1541 (N=O), 1455 (S=O), 1247, 1039 (C-O-C), 1188 (C-N), 690 (C-Cl), 627 (C-S); 1H-NMR (CDCl3, 400 MHz): δ (ppm) 7.68 (d, J = 8.0 Hz, 2H, H-2” & H-6”), 7.49 (d, J = 8.4 Hz, 2H, H-3” & H-5”), 3.81 (t, J = 7.2 Hz, 2H, H-1”’), 3.71-3.65 (m, 2H, He -2’ & He -6’), 3.32 (t, J = 7.2 Hz, 2H, H-2”’), 2.69-2.64 (m, 1H, H-4’), 2.57 (dt, J = 12.0, 2.4 Hz, 2H, Ha -2’ & Ha -6’), 2.15-2.10 (m, 2H, He -3’ & He -5’), 1.96-1.89 (m, 2H, Ha -3’ & Ha -5’); EI-MS (m/z): 515 [M+2]+, 513 [M]+, 286 [C12H13ClNO3S]+, 284 [C12H13ClN2O2S]•+, 258 [C11H13ClNO2S]+, 175 [C6H4ClO2S]+, 155 [C2H4I]+, 111 [C6H4Cl]+.

5-{1-[(4-Chlorophenyl)sulfonyl]piperidin-4-yl}-2-[(2-chloroethyl)thio]-1,3,4-oxadiazole (7x)

White amorphous solid; Yield: 83 %; M.P.: 157-158 ºC; Mol. formula: C15H17Cl2N3O3S2; Mol. weight: 421 gmol-1; IR (KBr, υmax, cm-1): 3058 (Ar C-H), 1664 (C=N), 1602 (Ar C=C), 1508 (N=O), 1385 (S=O), 1178, 1030 (C-O-C), 1086 (C-N), 634 (C-Cl), 606 (C-S); 1H-NMR (CDCl3, 400 MHz): δ (ppm) 7.66 (d, J = 8.4 Hz, 2H, H-2” & H-6”), 7.51 (d, J = 8.4 Hz, 2H, H-3” & H-5”), 3.79 (t, J = 7.2 Hz, 2H, H-1”’), 3.72-3.67 (m, 2H, He -2’ & He -6’), 3.64 (t, J = 7.2 Hz, 2H, H-2”’), 2.73-2.68 (m, 1H, H-4’), 2.56 (dt, J = 12.0, 2.4 Hz, 2H, Ha -2’ & Ha -6’), 2.14-2.08 (m, 2H, He -3’ & He -5’), 1.97-1.92 (m, 2H, Ha -3’ & Ha -5’); EI-MS (m/z): 423 [M+2]+, 421 [M]+, 286 [C12H13ClNO3S]+, 284 [C12H13ClN2O2S]•+, 258 [C11H13ClNO2S]+, 175 [C6H4ClO2S]+, 111 [C6H4Cl]+, 63 [C2H4Cl]+.

5-{1-[(4-Chlorophenyl)sulfonyl]piperidin-4-yl}-2-[(2-bromoethyl)thio]-1,3,4-oxadiazole (7y)

Off-white amorphous solid; Yield: 82 %; M.P.: 170-171 ºC; Mol. formula: C15H17ClBrN3O3S2; Mol. weight: 464 gmol-1; IR (KBr, υmax, cm-1): 3054 (Ar C-H), 1665 (C=N), 1604 (Ar C=C), 1502 (N=O), 1382 (S=O), 1173, 1033 (C-O-C), 1085 (C-N), 638 (C-Cl), 608 (C-S); 1H-NMR (CDCl3, 400 MHz): δ (ppm) 7.67 (d, J = 8.0 Hz, 2H, H-2” & H-6”), 7.49 (d, J = 8.0 Hz, 2H, H-3” & H-5”), 3.71 (t, J = 7.2 Hz, 2H, H-1”’), 3.70-3.64 (m, 2H, He -2’ & He -6’), 3.62 (t, J = 7.2 Hz, 2H, H-2”’), 2.70-2.66 (m, 1H, H-4’), 2.57 (dt, J = 12.4, 2.0 Hz, 2H, Ha -2’ & Ha -6’), 2.14-2.09 (m, 2H, He -3’ & He -5’), 1.95-1.87 (m, 2H, Ha -3’ & Ha -5’); EI-MS (m/z): 466 [M+2]+, 464 [M]+, 286 [C12H13ClNO3S]+, 284 [C12H13ClN2O2S]•+, 258 [C11H13ClNO2S]+, 175 [C6H4ClO2S]+, 111 [C6H4Cl]+, 107 [C2H4Br]+.

5-{1-[(4-Chlorophenyl)sulfonyl]piperidin-4-yl}-2-[(1,3-dioxalan-2-ylmethyl)thio]-1,3,4-oxadiazole (7z)

Light-bluish amorphous solid; Yield: 82 %; M.P.: 118-119 ºC; Mol. formula: C17H20ClN3O5S2; Mol. weight: 445 gmol-1; IR (KBr, υmax , cm-1): 3056 (Ar C-H), 1667 (C=N), 1608 (Ar C=C), 1504 (N=O), 1387 (S=O), 1175, 1032 (C-O-C), 1080 (C-N), 636 (C-Cl), 602 (C-S); 1H-NMR (CDCl3, 400 MHz): δ (ppm) 7.68 (d, J = 8.4 Hz, 2H, H-2” & H-6”), 7.49 (d, J = 8.4 Hz, 2H, H-3” & H-5”), 5.24 (t, J = 4.0 Hz, 1H, H-2”’), 4.01-3.98 (m, 2H, He -4”’ & He -5”’), 3.90-3.86 (m, 2H, Ha -4”’ & Ha -5”’), 3.65-3.61 (m, 2H, He -2’ & He -6’), 3.44 (d, J = 4.0 Hz, 2H, H-6”’), 2.90-2.85 (m, 1H, H-4’), 2.65 (dt, J = 12.4, 2.8 Hz, 2H, Ha -2’ & Ha -6’), 2.14-2.09 (m, 2H, He -3’ & He -5’), 2.02-1.95 (m, 2H, Ha -3’ & Ha -5’); EI-MS (m/z): 447 [M+2]+, 445 [M]+, 286 [C12H13ClNO3S]+, 284 [C12H13ClN2O2S]•+, 258 [C11H13ClNO2S]+, 175 [C6H4ClO2S]+, 111 [C6H4Cl]+, 87 [C4H7O2]+.

RESULTS AND DISCUSSION

The different S-substituted derivatives, 7a-z, of 5-{1-[(4-chlorophenyl)sulfonyl]piperidin-4-yl}-2-mercapto-1,3,4-oxadiazole (5) were synthesized by the protocol outlined in Figure 1. The different alkyl/aralkyl groups are listed in Table I. The bioactivity analysis included antibacterial activity against gram-positive and gram-negative bacteria, and inhibitory activity against AChE and urease enzymes. Computational and BSA binding studies were also carried out to see the interaction of synthesized compounds with amino acid residues and binding capacity with this specific protein, respectively.

FIGURE 1
Synthesis of 5-{1-[(4-chlorophenyl)sulfonyl]piperidin-4-yl}-2-[ (alkyl/aralkyl)thio]-1,3,4-oxadiazole (7a-z).

TABLE I
Different alkyl/aralkyl groups

Chemistry

Compound 7n is selected for single compound discussion. It was obtained as a white amorphous solid. The % yield was 81 %, and the melting point was determined to be 121-122 ºC. The molecular formula, C20H19Cl2N3O3S2, and molecular mass, 483 gmol-1, were established through 1H-NMR and EI-MS spectra. The IR spectrum showed absorptions for the main functionalities present in the molecule, namely 3127 (Ar C-H), 1682 (C=N), 1609 (Ar C=C), 1532 (N=O), 1396 (S=O), 1237, 1023 (C-O-C), 1167 (C-N), 710 (C-Cl) and 646 (C-S). The mass spectrum displayed different fragments that confirmed the molecular structure of all the synthesized compounds. The fragments at m/z 286 and 284 demonstrated the partial cleavage of the 1,3,4-oxadiazole ring. The fragment at m/z 175 was assigned to the 4-chlorophenylsulfonyl group and that at m/z 111 to the 4-chlorophenyl group. All these fragments were shown by all the compounds. The compound 7n specifically showed two fragments at m/z 125 and 65 for 2-chlorobenzyl as substituted tropyllium cation and cyclopentadienyl cation, respectively. The other prominent fragments are given in Figure 2. The two doublets of 1H-NMR signals at δ 7.68 (d, J = 8.8 Hz, 2H, H-2” & H-6”) and 7.49 (d, J = 8.4 Hz, 2H, H-3” & H-5”) were assigned to the 4-chlorophenylsulfonyl moiety; five signals at δ 3.65-3.62 (m, 2H, He -2’ & He -6’), 2.87-2.82 (m, 1H, H-4’), 2.62 (dt, J = 12.4, 2.8 Hz, 2H, Ha -2’ & Ha -6’), 2.12-2.08 (m, 2H, He -3’ & He -5’) and 2.00-1.91 (m, 2H, Ha -3’ & Ha -5’) were assigned to the piperidine moiety; and the signals at δ 7.53 (dd, J = 7.2, 1.2 Hz, 1H, H-6”’), 7.38 (dd, J = 8.0, 0.8 Hz, 1H, H-3”’), 7.23-7.19 (m, 2H, H-4”’ & H-5”’) and 4.51 (s, 2H, H-7”’) were assigned to the 2-chlorobenzyl group. 13C-NMR also corroborated the carbon skeleton of the molecule by exhibiting the following signals: six at δ (ppm) 168.7 (C-5), 164.1 (C-2), 139.5 (C-1”), 134.8 (C-4”), 134.4 (C-1”’) and 133.6 (C-2”’) for six quaternary carbons; seven at δ (ppm) 131.4 (C-4”’), 129.7 (C-5”’), 129.6 (C-3”’), 129.5 (C-3” & C-5”), 128.9 (C-2” & C-6”), 127.0 (C-6”’) and 32.2 (C-4’) for nine methine carbons; and three at δ (ppm) 44.9 (C-2’ & C-6’), 34.5 (C-7”’) and 28.2 (C-3’ & C-5’) for five methylene carbons. On the basis of these spectral data, the structure of 7n was determined to be 5-{1-[(4-chlorophenyl)sulfonyl]piperidin-4-yl}-2-[(2-chlorobenzyl)thio]-1,3,4-oxadiazole. Likewise, the structures of other synthesized compounds were corroborated through spectral data.

FIGURE 2
Proposed mass fragmentation pattern of 5-{1-[(4-chlorophenyl)sulfonyl]piperidin-4-yl}-2-[ (2-chlorobenzyl)thio]-1,3,4-oxadiazole (7n).

Antibacterial activity (in vitro)

Antibacterial activity was evaluated for all the compounds and the results are interpreted as % inhibition and MIC in Table II and Table III, respectively. The reference drug was ciprofloxacin. The results demonstrated slightly moderate to strong antibacterial potential of the synthesized compounds against S. typhi and B. subtilis and weakly moderate against the other three bacterial strains. Against S. typhi, 7q, 7t, 7u, 7w and 7z were the most active compounds with MIC values of 9.23±1.34, 9.22±3.28, 9.51±4.91, 8.78±2.50 and 8.23±3.00 µM, respectively. The high activity of these compounds was attributed to the presence of the 2-bromobenzyl, 3-phenylpropyl, 3-nitrobenzyl, 2-iodoethyl and 1,3-dioxalan-2-ylmethyl groups, respectively. The reference drug, ciprofloxacin, showed a MIC value of 7.15±1.29 µM. The most active compound was 7z, bearing three heterocyclic rings in its structure. The other molecules were also found to be moderately effective growth inhibitors of this bacterial strain. The growth of the bacterial strain E. coli was inhibited by almost all the compounds but to a very low extent, although some were moderate inhibitors. The most active compound against this strain was 7b, bearing an ethyl group, and exhibited a MIC value of 10.32±3.65 µM compared to the reference at 7.90±1.87 µM. Two compounds, 7p and 7v, were inactive against this strain. Likewise, P. aeruginosa and S. aureus were inhibited by all the compounds but to a weak to moderate level. The most active compound against both of the strains was 7u, bearing a 3-nitrobenzyl group, and showed a MIC value of 10.40±1.67 and 10.58±1.82 µM compared to the reference at 8.21±1.21 and 8.00±2.98 µM for P. aeruginosa and S. aureus, respectively. Compound 7p was inactive against both strains, while 7v and 7y were also inactive against S. aureus. The compounds displayed moderate to strong activity against B. subtilis. The compounds 7m, 7n, 7s and 7t were the most active ones with MIC values of 9.28±4.43, 8.95±1.93, 9.70±2.21 and 9.25±1.43 µM compared to that of the reference drug, i.e., 7.12±2.11 µM. The better activities of these compounds might have been due to the presence of the 4-fluorobenzyl, 2-chlorobenzyl, 2-phenylethyl and 3-phenylpropyl groups, respectively. Overall, the halogenated groups attached to sulfur of 1,3,4-oxadiazole, boosted considerably bioactive potential. No doubt, the antibacterial potential was already increased by the 4-chlorophenyl group present in all the synthesized molecules. The size and position of halogens in the molecule have a considerable effect on biological activities (Liu et al., 2012Liu J, Wu F, Chen L, Zhao L, Zhao Z, Wang M, Lei S. Biological evaluation of coumarin derivatives as mushroom tyrosinase inhibitors. Food Chem. 2012;135(4):2872-2878.).

TABLE II
The % inhibition for antibacterial activity of synthesized compounds
TABLE III
The MIC of antibacterial activity of synthesized compounds

AChE inhibition studies

All the synthesized compounds were tested for AChE inhibitory activity and the results (% inhibition and IC50) are given in Table IV. This activity nullifies the effects of AChE causing mental disorders. The main stream of the tested compounds was found active in the variable range from slightly active to highly active because of the collective effect of three heterocyclic moieties shown in the general synthetic scheme. Compounds 7m, 7n, 7q, 7s, 7t, 7x and 7y were the most active members of this series, acting as anti-AChE agents with IC50 values of 95.57±0.21, 72.24±0.18, 52.23±0.23, 51.39±0.21, 65.91±0.23, 46.21±0.22 and 83.56±0.17 µM, respectively, as compared to that of eserine used as a reference standard, i.e., 0.04±0.0001 µM. The high activity of the above compounds might have been due to the presence of electron withdrawing-species in the N-substituted aromatic ring shown in Table I. The compounds showing moderate potential against AChE were 7i, 7j, 7k, 7l, 7p, 7u and 7v with IC50 values of 133.25±0.15, 179.34±0.12, 185.42±0.17, 139.81±0.22 and 189.64±0.19 µM, respectively. The presence of bulky groups in the N-substituted aromatic ring was responsible for moderate activity. The low activity of compounds 7e and 7f was due to the presence of electron-rich moieties in the N-substituted ring. Among the active members of this series, compounds 7q, 7s and 7x were the most active agents and could be recommended for in vivo studies. The highest inhibition potential of these highly active members could have been due to the 2-bromobenzyl, 2-phenylethyl and 2-chloroehyl substituents, respectively.

TABLE IV
Acetyl cholinesterase (AChE) and Urease inhibition studies

Urease inhibition studies

All the synthesized compounds were tested for anti-urease activity with reference to thiourea used as the standard. The results showed that all the compounds had outstanding urease inhibitory activity with a few exceptions (Table IV). The compounds were even much more active compared to the standard used. Compounds 7l, 7m, 7n, 7o, 7p, 7r, 7u, 7v, 7x and 7v were highly active against urease with IC50 values of 2.14±0.003, 0.63±0.001, 2.17±0.006, 1.13±0.003, 1.21±0.005, 6.28±0.003, 2.39±0.005, 2.15±0.002, 2.26±0.003 and 2.14±0.002 µM compared to thiourea with an IC50 value of 21.25±0.15 µM. The high level of anti-urease activity might have been attributed to the presence of electron-withdrawing substituents (Table I), such as 7m with a 4-fluorobenzyl group, the most active compound of the series. All the tested compounds showed exceptional urease inhibitory potential and could be used to design new drugs to control the negative effects of urease after in vivo studies.

Docking study for binding of 7x with AChE

The 7x compound was used to evaluate its molecular docking in AChE inhibition studies. 7x showed the highest inhibitory potential against AChE. It was necessary to identify the active site present in the molecule, responsible for its best activity. To understand the possible factors responsible for its better activity, its docking studies were carried out. The Surflex-Dock module of SybylX-1.3 was used to dock ligand 7x into the active site of AChE enzyme (Figure 3). The authenticity reliability of our docking procedure was attained by the extraction of donepezil and re-docking into the active cite of AChE. The comparison of docked poses and experimentally determined binding mode was done by donepezil in AChE. Molecular docking results of ligand poses were overlapped with the crystallographic confirmation of donepezil in AChE, which showed the reliability of docking studies. The graphic analysis of docking results (Figure 3) demonstrated that ligand 7x penetrated deeply into the two main active sites of AChE (CAS and PAS). In the active site of AChE, the ligand acquired such a binding conformation, so that the nitrogen of the oxdiazole ring (B-ring) was able to form a H-bond with the backbone NH2 of Phe295 (Figure 3). The SO2 group of 4-chlorobezene sulfonyl was also able to form a H-bond with NH2 groups of TYR337 and TRP86. The chlorine of the 4-chlorobezene sulfonyl group (A-ring) was shown to penetrate deeply into the CAS region where its Cl at C4 of ring-A could establish a hydrophobic or van der Waals interaction with surrounding residues Glu202, Ser203 and Gly120. The chlorine of the ethyl group substituted at the S position extended well near the solvent-exposed region where it could establish H-bonds with the backbone and side chain of the surrounding residue Phe295. The following variations in ligand-receptor interaction pattern could account for the higher AChE inhibitory potency of compound 7x.

FIGURE 3
Docked conformation of AChE inhibitors 7x (Cyan), 7x (Magenta) superimposed over experimental conformation of donepezil (Magenta) in AChE main binding site.

BSA binding studies using fluorescence measurements

Nitrogen-containing compounds are well known for their broad spectrum of biological applications. Oxadiazole and its derivatives are the class of compounds having such functionalities with various biological applications such as antibacterial, antispasmodic, anti-inflammatory, antiplatelet aggregation, tumor cell growth inhibition and differentiation, tumor cell apoptosis, etc. (Holt, Chaires, Trent, 2008Holt PA, Chaires JB, Trent JO. Molecular docking of intercalators and groove-binders to nucleic acids using Autodock and Surflex. J Chem Inf Model. 2008;48(8):1602-1615.). The current study was designed to determine the binding of the synthesized compounds to BSA to better understand their pharmacological effects. The selected synthesized compounds were used to evaluate the effects of different substituents on BSA binding capacity. The binding of these compounds was determined by fluorometric titrations of BSA with varying concentration of the derivatives (0 - 5.57 x 10-4 M). Their binding abilities and binding sites were compared to that of the fluorescence markers warfarin and ibuprofen.

BSA shows a maximum emission at λem = 336 nm. Addition of the synthesized compounds was found to quench the fluorescence intensity of BSA, showing that these derivatives were able to bind to BSA, causing some modification in the microenvironment around the amino acid residues situated in the sub-domain of BSA, leading to a hypochromic shift in the fluorescence spectrum. Fluorescence quenching can be classified as either a static or dynamic process and is caused by different mechanisms including excited state reactions, molecular rearrangements, energy transfer, formation of ground state complex and collisional quenching (Wang et al., 2013Wang Y, Jiang X, Zhou L, Yang L, Xia G, Chen Z, Duan M. Synthesis and binding with BSA of a new gemini surfactant. Colloids Surf A Physicochem Eng Asp. 2013;436:1159-1169.). The type of quenching mechanism is usually interpreted by the Stern-Volmer equation:

Equation 1 F o / F = 1 + K SV Q = K q τ o Q + 1

where Fo and F are the fluorescence intensities of BSA respectively before and after addition of quencher, Ksv is the Stern-Volmer quenching constant, [Q] is the concentration of the quencher, Kq is the apparent bimolecular quenching rate constant, τo is the average lifetime of the biomolecule without the quencher and its value is 10-8 s (Zhang et al., 2012Zhang Z, Zheng P, Guo Y, Yang Y, Chen Z, Wang X, An X, Shen W. The effect of the spacer rigidity on the aggregation behavior of two ester-containing gemini surfactants. J Colloid Interface Sci. 2012;379(1):64-71.). The emission spectra of the test compounds and their respective Stern-Volmer plots are given in Figure 4.

FIGURE 4
Fluorescence emission spectra of BSA and different amounts of selected compounds (7l, 7m, 7o, 7t, 7u) at 295 nm and 298 K (inset graph corresponding to the Stern-Volmer plot).

In general, the calculated Kq values (Table V) were found to be larger than the maximum scattering collision quenching constant (2 × 1010 M-1 s−1) in dynamic quenching, which indicated that the fluorescence quenching process of BSA with the synthesized compounds, as well as warfarin and ibuprofen, were primarily governed by a static quenching mechanism (Wang et al., 2013Wang Y, Jiang X, Zhou L, Yang L, Xia G, Chen Z, Duan M. Synthesis and binding with BSA of a new gemini surfactant. Colloids Surf A Physicochem Eng Asp. 2013;436:1159-1169.), rather than a dynamic one. The binding constant Ka and the number of binding sites n of the compounds were determined using the double logarithm Equation 2.

TABLE V
Binding constant and number of binding site of compounds warfarin and ibuprofen with BSA at 298K

Equation 2 Log F 0 F / F = log K a + n log Q

The double logarithm plots of the different compounds are shown in Figure 5 and their binding constant Ka is given in Table V. The presence of halogen atoms in (7o and 7m) was found to decrease the binding constant. However, compound 7u showed the strongest binding with BSA, which was due to the presence of a nitro group at the meta position, enhancing the binding of 7u to BSA. The strong binding of compound 7u to BSA may account for its high antibacterial potential.

FIGURE 5
Double logarithm plots of compounds 7l, 7m, 7o, 7t and 7u.

CONCLUSION

The synthesized compounds with notable yields were structurally identified through spectral data. The interactions with amino acid residues as well as with specific proteins responsible for the pharmacological activity of the synthesized compounds were availed through docking and BSA binding interactions, respectively. The antibacterial activity results demonstrated moderate to very high activity against S. typhi and B. subtilis but weak to moderate activity against the other three bacterial strains tested. The presence of halogenated groups in the molecules had a great effect on the antibacterial potential of the synthesized molecules. Similarly, all the compounds proved to be very active against AChE. But above all, the urease inhibition potential of these compounds was very impressive compared to the standard used. Thus, the molecules with high bioactive potential could be considered for further in vivo studies and as candidates in drug discovery programs in the pharmaceutical industry.

ACKNOWLEDGEMENT

The authors acknowledge the Higher Education Commission (HEC) of Pakistan for the financial assistance for the completion of this project. Dr. A. Leyva (USA) helped with English editing of the manuscript.

REFERENCES

  • Aziz-ur-Rehman, Tanveer W, Abbasi MA, Afroz S, Khan KM, Ashraf M, Afzal I. Synthesis, characterization and biological screening of various N-substituted derivatives of sulfonamides. Int J Chem Res. 2011;3(3):99-104.
  • Barbe V, Cruveiller S, Kunst F, Lenoble P, Meurice G, Sekowska A, Vallenet D, Wang T, Moszer I, Medigue C, Danchin A. From a consortium sequence to a unified sequence: the Bacillus subtilis 168 reference genome a decade later. Microbiology. 2009;155(Pt 6):1758-1775.
  • Bhattacharya SS, Das U, Choudhury BK. Occurrence & antibiogram of Salmonella typhi & S. paratyphi A isolated from Rourkela, Orissa. Indian J Med Res. 2011;133:431-433.
  • Chapdelaine P, Tremblay RR, Dube JY. r-Nitrophenol-a-D-Glucopyranoside as substrate for measurement of maltase activity in Human Semen. Clin Chem. 1978;24(2):208-211.
  • Chohan TA, Chen JJ, Qian HY, Pan YL, Chen JZ. Molecular modeling studies to characterize N-phenylpyrimidin-2-amine selectivity for CDK2 and CDK4 through 3D-QSAR and molecular dynamics simulations. Mol BioSyst. 2016b;12(4):1250-1268.
  • Chohan TA, Qian HY, Pan YL, Chen JZ. Molecular simulation studies on the binding selectivity of 2-anilino-4-(thiazol-5-yl)-pyrimidines in complexes with CDK2 and CDK7. Mol. BioSyst. 2016a;12(1):145-161.
  • El-Din MMG, El-Gamal MI, Abdel-Maksoud MS, Yoo KH, Oh CH. Synthesis and broad-spectrum anti-proliferative activity of diarylamides and diarylureas possessing 1,3,4-oxadiazole derivatives. Bioorg Med Chem Lett. 2015;25(8):1692-1699.
  • Ghersi D, Sanchez R. Beyond structural genomics: computational approaches for the identification of ligand binding sites in protein structures. J Struct Funct Genomics. 2011;12(2):109-17.
  • Harris LG, Foster SJ, Richards RG. An introduction to Staphylococcus aureus, and techniques for identifying and quantifying S. aureus adhesins in relation to adhesion to biomaterials: review. Eur Cells Mater. 2002;4:39-60.
  • Holt PA, Chaires JB, Trent JO. Molecular docking of intercalators and groove-binders to nucleic acids using Autodock and Surflex. J Chem Inf Model. 2008;48(8):1602-1615.
  • Jain AN, Dock D. Robust performance from ligand energetic modeling, ring flexibility, and knowledge-based search. J Comput Aided Mol Des. 2007;21(5):281-306.
  • Kumar D, Sundaree S, Johnson EO, Shah K. An efficient synthesis and biological study of novel indolyl-1,3,4-oxadiazoles as potent anticancer agents. Bioorg Med Chem Lett. 2009;19(15):4492-4494.
  • Li P, Shi L, Gao MN, Yang X, Xue W, Jin LH, Hu DY, Song BA. Antibacterial activities against rice bacterial leaf blight and tomato bacterial wilt of 2-mercapto-5-substituted-1,3,4-oxadiazole/thiadiazole derivatives. Bioorg. Med. Chem. Lett. 2014;25(3):481-484.
  • Liu J, Wu F, Chen L, Zhao L, Zhao Z, Wang M, Lei S. Biological evaluation of coumarin derivatives as mushroom tyrosinase inhibitors. Food Chem. 2012;135(4):2872-2878.
  • Lodhi MA, Abbasi MA, Choudhary MI, Ahmad VU. Kinetic studies on triacontanyl palmitate: a urease inhibitor. Nat Prod Res. 2007;21(8):721-725.
  • Mahernia S, Bagherzadeh K, Mojab F, Amanlou M. Urease Inhibitory Activities of some Commonly Consumed Herbal Medicines. Iran J Pharm Res. 2015;14(3):943-947.
  • Nafeesa K, Aziz-ur-Rehman, Abbasi MA, Siddiqui SZ, Rasool S, Hussain G, Ahmed I. Synthesis and biological screening of S-substituted derivatives of 5-{1-[(4-chlorophenyl)sulfonyl]-3-piperidinyl}-1,3,4-oxadiazole-2-yl sulfide. Asian J Chem. 2015;27(6):2105-2108.
  • Nithiya S, Karthik N, Jayabharathi J. In vitro antioxidant activity of hindered piperidone derivatives. Int J Pharm Pharm Sci. 2011;3(3):254-256.
  • Omar FA, Mahfouz NM, Rahman MA. Design, synthesis and anti-inflammatory activity of some 1,3,4-oxadiazole derivatives. Eur J Med Chem. 1996;31(10):819-825.
  • Pressler T, Bohmova C, Conway S, Dumcius S, Hjelte L, Høiby N, Kollberg H, Tümmler B, Vavrova V. Chronic Pseudomonas aeruginosa infection definition: EuroCareCF Working Group report. J Cyst Fibros. 2011;10(Suppl 2):S75-S78.
  • Sanchez-Sancho F, Herrandon B. Short syntheses of (S)-pipecolic acid, (R)-coniine, and (S)-coniceine using biocatalytically-generated chiral building block. Tetrahedron Asymmetry. 1998;9(11):1951-1965.
  • Tan TMC, Chena Y, Kong KH, Bai J, Li Y, Lim SG, Ang TH, Lam Y. Synthesis and the biological evaluation of 2-benzenesulfonylalkyl-5-substituted-sulfanyl-[1,3,4]-oxadiazoles as potential anti-hepatitis B virus agents. Antiviral Res. 2006;71(1):7-14.
  • Tougu V. Acetylcholinesterase: mechanism of catalysis and inhibition. Curr Med Chem. 2001;1(2):155-170.
  • Vogt RL, Dippold L. Escherichia coli O157:H7 outbreak associated with consumption of ground beef, June-July 2002. Public Health Rep. 2005;120(2):174-178.
  • Wang Y, Jiang X, Zhou L, Yang L, Xia G, Chen Z, Duan M. Synthesis and binding with BSA of a new gemini surfactant. Colloids Surf A Physicochem Eng Asp. 2013;436:1159-1169.
  • Wani TA, AlRabiah H, Bakheit AH, Kalam MA, Zargar S. Study of binding interaction of rivaroxaban with bovine serum albumin using multi-spectroscopic and molecular docking approach. Chem Cent J. 2017a;11(134):1-9.
  • Wani TA, Bakheit AH, Abounassif MA, Zargar S. Study of interactions of an anticancer drug neratinib with bovine serum albumin: Spectroscopic and molecular docking approach. Front. Chem. 2018;6:47.
  • Wani TA, Bakheit AH, Al-Majed ARA, Bhat MA, Zargar S. Study of the interactions of bovine serum albumin with the new anti-inflammatory agent 4-(1,3-Dioxo-1,3-dihydro-2H-isoindol-2-yl)-N′-[(4-ethoxy-phenyl)methylidene]benzohydrazide using a multi-spectroscopic approach and molecular docking. Molecules. 2017b;22(1258):1-10.
  • Wani TA, Bakheit AH, Zargar S, Hamidaddin MA, Darwish IA. Spectrophotometric and molecular modelling studies on in vitro interaction of tyrosine kinase inhibitor linifanib with bovine serum albumin. PIoS One. 2017c;12(4):e0176015.
  • Wu XH, Liu JJ, Huang HM, Xue WW, Yao XJ. Interaction studies of aristolochic acid I with human serum albumin and the binding site of aristolochic acid I in subdomain IIA. Int J Biol Macromol. 2011;49(3):343-350.
  • Zhang K, Wang P, Xuan LN, Fu XY, Jing F, Li S, Liu YM, Chen BQ. Synthesis and antitumor activities of novel hybrid molecules containing 1,3,4-oxadiazole and 1,3,4-thiadiazole bearing Schiff base moiety. Bioorg Med Chem Lett. 2014;24(22):5154-5156.
  • Zhang Z, Zheng P, Guo Y, Yang Y, Chen Z, Wang X, An X, Shen W. The effect of the spacer rigidity on the aggregation behavior of two ester-containing gemini surfactants. J Colloid Interface Sci. 2012;379(1):64-71.

Publication Dates

  • Publication in this collection
    07 Dec 2020
  • Date of issue
    2020

History

  • Received
    04 Feb 2018
  • Accepted
    02 Jan 2019
Universidade de São Paulo, Faculdade de Ciências Farmacêuticas Av. Prof. Lineu Prestes, n. 580, 05508-000 S. Paulo/SP Brasil, Tel.: (55 11) 3091-3824 - São Paulo - SP - Brazil
E-mail: bjps@usp.br