Next Article in Journal
Development and Comparative Evaluation of Two Highly Sensitive Immunosensor Platforms for Trace Determination of Copper Ions in Drinking Water Using a Monoclonal Antibody Specific to Copper-EDTA Complex
Previous Article in Journal
Chemical Composition, Market Survey, and Safety Assessment of Blue Lotus (Nymphaea caerulea Savigny) Extracts
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Design, Synthesis, In Vitro Biological Evaluation and In Silico Molecular Docking Study of Benzimidazole-Based Oxazole Analogues: A Promising Acetylcholinesterase and Butyrylcholinesterase Inhibitors

1
Department of Chemistry, Hazara University, Mansehra 21120, Pakistan
2
Department of Chemistry, University of Okara, Okara 56130, Pakistan
3
Department of Chemistry, Abbottabad University of Science and Technology (AUST), Abbottabad 22500, Pakistan
4
Department of Zoology, Wildlife and Fisheries, University of Agriculture Faisalabad, Sub Campus Toba Tek Singh, Faisalabad 36050, Pakistan
5
Department of Agroecology-Climate and Water, Aarhus University, Blichers Allé 20, 8830 Tjele, Denmark
6
Department of Agronomy, Faculty of Agriculture and Environment, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan
7
Department of Botany, Government College University, Lahore 54000, Pakistan
8
Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
*
Authors to whom correspondence should be addressed.
Molecules 2023, 28(20), 7015; https://doi.org/10.3390/molecules28207015
Submission received: 10 August 2023 / Revised: 21 September 2023 / Accepted: 22 September 2023 / Published: 10 October 2023

Abstract

:
Alzheimer’s disease (AD) is a degenerative neurological condition that severely affects the elderly and is clinically recognised by a decrease in cognition and memory. The treatment of this disease has drawn considerable attention and sparked increased interest among the researchers in this field as a result of a number of factors, including an increase in the population of patients over time, a significant decline in patient quality of life, and the high cost of treatment and care. The current work was carried out for the synthesis of benzimidazole-oxazole hybrid derivatives as efficient Alzheimer’s inhibitors and as a springboard for investigating novel anti-chemical Alzheimer’s prototypes. The inhibition profiles of each synthesised analogue against acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE) enzymes were assessed. All the synthesized benzimidazole-based oxazole analogues displayed a diverse spectrum of inhibitory potentials against targeted AChE and BuChE enzymes when compared to the reference drug donepezil (IC50 = 2.16 ± 0.12 M and 4.50 ± 0.11 µM, respectively). The most active AChE and BuChE analogues were discovered to be analogues 9 and 14, with IC50 values of 0.10 ± 0.050 and 0.20 ± 0.050 µM (against AChE) and 0.20 ± 0.050 and 0.30 ± 0.050 µM (against BuChE), respectively. The nature, number, position, and electron-donating and -withdrawing effects on the phenyl ring were taken into consideration when analysing the structure-activity relationship (SAR). Molecular docking studies were also carried out on the active analogues to find out how amino acids bind to the active sites of the AChE and BuChE enzymes that were being studied.

1. Introduction

Alzheimer’s disease is mainly caused by cholinesterase (AChE and BuChE) enzymes. These enzymes primarily work to hydrolyze acetylcholine into choline acetic acid [1]. These result in a lack of acetylcholine in the hippocampus and cortex of the brain, which are linked to important psychological activities [2] and result in the development of Alzheimer’s disease (AD), a persistent and irreversible brain illness. The cholinergic system of the brain continues to be affected, which frequently results in confusion, memory loss, cognitive impairment, difficulty solving problems, and thinking difficulties [3,4,5]. Additionally, because these enzymes are in charge of the neurotoxic β-amyloid accumulation that results in neuronal cell apoptosis, AD is the main cause of dementia in an ageing society. The best course of action for treating AD is to target both of these enzymes [6,7] by concentrating on the acetylcholinesterase enzyme, which has two binding sites: a peripheral location for the interaction of β-amyloid and a catalytic site for the hydrolysis of acetylcholine. In the form of the AChE-Aβ complex, it induces neurotoxicity as a result of its interaction with proteins. Furthermore, BuChE is mostly found in the heart, lungs, kidneys, liver, gut, and serum, whereas AChE is found in the cholinergic brain, neurons, and muscle [8,9]. The primary role of these enzymes is to break down substances that contain esters; AChE is predominant in the brain, and as acetylcholine activity gradually declines, BuChE function increases. To block the potential of these enzymes, a powerful medication is still required [10]. The FDA has approved a number of medications, including donepezil, rivastigmine, tacrine, and galantamine, for the treatment of Alzheimer’s. Tacrine and Rivastigmine have been used to inhibit both AChE and BuChE, while Donepezil and Galantamine have been used to inhibit AChE [11] (Figure 1).
Antioxidants [12], anticancer agents [13], CNS stimulants [14], antiviral agents [15], anticoagulants [16], anti-inflammatory agents [17], anti-parasitic agents [18], anti-microbial agents [19], and blood pressure lowering agents [20] have all been found in benzimidazole and its derivatives. There are numerous bioactive drugs used in the market that contain benzimidazole skeletons in their structures, such as bandamustine, enviradine, astemizole, albendazole, and omeprazole (Figure 2) [21].
Oxazole scaffolds play a vital role in the design and development of several biologically active pharmaceutical drugs with interesting biological profiles, natural products, functional materials, and ligand frameworks [22]. Oxaprozin, aleglitazar, and aristoxazole (Figure 3) are examples of naturally occurring compounds that contain oxazole, which is a physiologically active scaffold [23,24].
We had already synthesized several N-containing heterocyclic compounds as potent inhibitors of alpha-amylase, alpha-glucosidase, urease, β-glucuronidase, thymidine phosphorylase, AChE and BuChE enzymes [25,26,27]. Furthermore, the molecular hybridization approach has been widely employed for the design and synthesis of hybrid analogues for the treatment of Alzheimer’s disease. This approach mainly involves the combining of two or more than two different pharmacophore moieties in a single molecule having a common skeleton. These hybrid molecules may have advantages over standard drugs. In the current work, we used molecular hybridization to combine the biologically important two heterocyclic moieties; benzimidazole and oxazole, to obtain new hybrid molecules (Figure 4). As earlier discussed, both benzimidazole and oxazole moieties are very important for the treatment of Alzheimer’s disease, and thus hybrid analogues containing benzimidazole and oxazole moieties were synthesized and evaluated for their in vitro AChE and BuChE and molecular docking studies thereafter. Keeping in mind the biological importance of benzimidazole [28,29,30] and oxazole [31] scaffolds (Figure 4), in this study, we designed and synthesized hybrid analogues based on benzimidazole-bearing oxazole derivatives to further explore the AChE and BuChE inhibition profiles for the better treatment of Alzheimer’s disease.

2. Results and Discussion

2.1. Chemistry

The synthesis of hybrid benzimidazole-based oxazole derivatives was completed in three steps:
First, the reaction mixture of 2-marcaptobenzimidazole (I, 1 mmol) and 2-bromoacetophenone (II, 1 mmol) was mixed and refluxed for 2–3 h in ethanol (10 mL) in the presence of triethylamine as a catalyst to produce the first intermediate product (III) [32]. The second intermediate product (IV) was produced by treating the intermediate (III) and semicarbazide in equal amounts in ethanol (10 mL) and glacial acetic acid as catalysts and refluxing the mixture for around 4 h. After the reaction was complete, the mixture was cooled to room temperature, and the precipitate solid that was produced was filtered and washed with n-hexane. TLC plates had been used to keep tabs on the reaction’s development. An equivalent quantity of intermediate (IV) underwent cyclization with stirring overnight with an equivalent amount of various substituted 2-bromoacetophenone in EtOH and Et3N (catalyst) in order to produce the benzimidazole-oxazole hybrid compounds (119) in proper yield (Scheme 1). After cooling to 25 °C, the solvent was withdrawn, and the resulting solid residue was cleaned by washing with n-hexane before being re-crystallized from ethyl alcohol. Many spectroscopic techniques, including 13C-NMR, 1H-NMR, and HR-EIMS, were used to determine the structures of all freshly synthesized analogues.

2.2. In Vitro Acetylcholinesterase and Butyrylcholinesterase Inhibitory Activities

Hybrid analogues based on benzimidazole incorporating an oxazole skeleton were synthesised and then screened in vitro for their inhibitory activities against targeted AChE and BuChE enzymes as compared to donepezil as the standard drug. All synthesised benzimidazole-based oxazole analogues showed a varied range of inhibitory potentials against targeted AChE and BuChE enzymes, with IC50 values ranging from 0.10 ± 0.050 to 12.60 ± 0.30 µM (against AChE) and 0.20 ± 0.050 µM to 16.30 ± 0.30 µM (against BuChE) when compared to standard donepezil (IC50 = 2.16 ± 0.12 µM and 4.5 ± 0.11 µM, respectively (Table 1). Structure-activity relationship (SAR) studies have shown that inhibitory potentials were greatly affected by changing either the position of substituent(s) or the nature of substituent(s) around both aryl parts; therefore, to better understand SAR studies, the synthesized analogues were split into various parts, including benzimidazole, oxazole, and aryl parts (R1 and R2), and it was further revealed from SAR studies that each part effectively contributed to inhibitory potentials, and any variation found on both aryl parts (R1 and R2, respectively) may result in different inhibitory potentials against targeted enzymes (Figure 5).

Structure Activity Relationship (SAR) for Acetylcholinesterase (AChE) and Butyrylcholinesterase (BuChE) Activities

Structure-activity relationships were performed on the basis of variety in different substitution patterns on both rings B and C, respectively. Derivatives 9 (IC50 = 0.10 ± 0.050 µM and 0.20 ± 0.050 µM), having a –NO2 group at the 3-position on ring C and triflouromethyl substitution at the 4-position on ring B, and derivative 14, having dichloro substitution at the 2- and 3-positions on ring C along with a methyl group at the para position on ring B, were identified as the first and second most potent inhibitors of AChE and BuChE enzymes among the synthesized derivatives.
Among the compounds with an unsubstituted ring B, derivative 10 (IC50 = 1.40 ± 0.10 µM and 2.10 ± 0.10 µM) had a nitro group at meta on ring C and derivative 17 (IC50 = 2.50 ± 0.10 µM and 3.50 ± 0.10µM) had a nitro group at ortho on ring C. The difference in the potentials of these derivatives may be due to the different positions of the nitro group on ring C.
By comparing the derivatives with a methyl group on ring B, derivative 2 (IC50 =1.60 ± 0.10 µM and1.90 ± 0.10 µM) had a –NO2 group at the ortho position on ring C, derivative 3 (IC50 = 4.40 ± 0.10 µM and 5.80 ± 0.10 µM) had a –NO2 group at the meta position on ring C, derivative 4 (IC50 = 9.80 ± 0.20 µM and 11.50 ± 0.20 µM) had methyl at the para position on ring C, derivative 5 (IC50 = 9.80 ± 0.20 µM and 11.50 ± 0.20 µM) had a methoxy group at the meta position on ring C, derivative 6 (IC50 =0.40 ± 0.05 µM and 1.10 ± 0.05 µM) had dichloro substitutions at the meta and para positions on ring C, derivative 8 (IC50 = 5.30 ± 0.20 µM and 7.20 ± 0.10 µM) had a methoxy group at the ortho position on ring C, derivative 12 (IC50 = 0.40 ± 0.05 µM and 1.10 ± 0.05 µM) had a chloro group at the para position on ring C, and derivative 14 (IC50 = 0.20 ± 0.050 µM and 0.30 ± 0.050 µM) had dichloro substitutions at the ortho and para positions on ring C. The difference in the potentials of these entire derivatives may be due to the different nature, number, and position of the substituents on ring C.
Among the derivatives having a chloro group on ring B, compound 11 (IC50 = 0.70 ± 0.50 µM and 1.20 ± 0.010 µM) had a NO2 group at the meta position on ring C, 18 (IC50 = 0.80 ± 0.10 µM and 1.70 ± 0.10 µM) had a nitro group at the ortho position on ring C, and 19 (IC50 = 2.70 ± 0.20 µM and 1.10 ± 0.05 µM) had an OCH3 group at the meta position on ring C, correspondingly. The difference in the inhibitory potentials of these derivatives may be due to the different nature, number, and position of the substituents on ring C.
Overall, it was also observed that the variation in number, nature, and position of certain groups at particular positions on rings B and C greatly affects the inhibitory potentials of the synthesized derivatives. Therefore, a molecular docking study was performed to understand the binding interaction of the most potent derivatives with the active site of enzymes.

2.3. Molecular Docking Study

A molecular docking study was performed on the most active benzimidazole-oxazole analogues (9, 14, and 6) in order to elucidate the in vitro study. Subsequently, all these active analogues showed a well-fitting binding mode with different binding affinities and correlated well with the in vitro studies. All of the active compounds, 9, 14, and 6, have similar and related chemistry, with slight modifications at different positions. These different functional moieties showed wide differences in their interactions with the active pocket of the targeted enzyme. Compound 9 showed maximum interactions with the active sites of both targeted AChE and BuChE enzymes and was found to be the most effective, and this was similar in the in vitro study. The details of the interactions are given in Table 2. It was also found that not only does the direct attachment of the electron-withdrawing group (–NO2) cause good activity, but the indirect attachment of the electron-withdrawing –F3 moiety in the form of –CF3 is also important, which in turn attaches the ring of the analogue. The better interactions, activity, and docking score of analogue 9 were mainly due to this reason. The electron-withdrawing –NO2 and –CF3 groups withdraw electronic density from the extended benzene ring, making the benzene ring more electrophilic; hence, the extended benzene ring remains electron-deficient, which further regains stability by forming pi-cation interactions with the active site of the targeted enzyme. From docking analysis, it was found that not only do electron-withdrawing groups such as –NO2 and –CF3 groups enhance the activity by making the benzene ring cationic, but they also enhance the enzymatic activity through the sidewise involvement of the oxygen of –NO2 and –F in hydrogen bonding and halogen (fluorine) with the active sites of both targeted enzymes. The presence of the –NO2 functional group is more effective in its meta-position; therefore, compound 9 has very good activity and interactions, as shown in Table 2. Compound 14, which holds electron-withdrawing di-Cl moieties at the 2,4-position of ring C and the para-methyl substation on ring B, was proven to be the second most active analogue and adopted several significant interactions with the active sites of both targeted AChE and BuChE (Table 2). Changing the position of di-Cl moieties around ring C, as in the case of compound 6, resulted in different enzymatic activities and hence different interactions with the active sites of both targeted AChE and BuChE enzymes (Figure 6, Figure 7, Figure 8, Figure 9, Figure 10 and Figure 11, Table 2).

2.4. ADMET Analysis

ADMET analysis describes the different properties of the subjected ligand, such as log Kp, Lipinski, Ghose, Veber, Egan, Muegge, Bioavailability, PAINS, and Brenk, etc. (Figure 12, Figure 13 and Figure 14).

3. Experiment

3.1. General Procedure for the Synthesis of Benzimidazole-Oxazole Hybrid Derivatives (119)

2-Marcaptobenzimidazole (I, 1 mmol) and 2-bromoacetophenone (II, 1 mmol) were mixed and refluxed for 2–3 h in ethanol (10 mL) in the presence of triethylamine to produce the first intermediate product (III) [32]. The second intermediate product (IV) was produced by treating the intermediate (III) and semicarbazide in equal amounts in ethanol (10 mL) and glacial acetic acid as catalysts and refluxing the mixture for around 4 h. After the reaction was complete, the mixture was cooled to room temperature, and the precipitate solid that was produced was filtered and washed with n-hexane. TLC plates were used to check the reaction’s development. An equivalent quantity of intermediate (IV) underwent cyclization with stirring overnight with an equivalent amount of various substituted 2-bromoacetophenone in EtOH and Et3N (catalyst) in order to produce the benzimidazole-oxazole hybrid compounds (119) at proper yield. After cooling to 25 °C, the solvent was withdrawn, and the resulting solid residue was cleaned by washing with n-hexane before being re-crystallized from ethyl alcohol.

3.2. Spectral Analysis

Spectra of some compounds are provided as Supplementary Materials.

3.2.1. (E)-2-(2-(2-((1H-Benzo[d]imidazol-2-yl)thio)-1-(4-methoxyphenyl)ethylidene)hydrazinyl)-4-(2-nitrophenyl)oxazole (1)

Yield: 69%; Rf value 0.59 (7:3 n-hex: EtOAc); 1 H NMR (600 MHz, DMSO-d6): δ12.75 (s, 1H, -NH), 10.67 (s, 1H, -NH), 8.21 (dd, J(6″, 5″) = 8.0Hz, J(6″, 4″) = 2.4Hz, 1H, H-6″), 8.13 (d, J(3″, 4″) = 7.8 Hz, 1H, H-3″), 8.02 (d, J(2′, 3′/ 6′, 5′) = 8.6Hz, 2H, H-2′/H-6′), 7.96 (s, 1H, oxazole-H), 7.93–7.86 (m, 1H, H-5″), 7.82 (t, J(4″/5″, 3″) = 9.0Hz, 1H, H-4″), 7.65 (m, 2H, H-4/H-7), 7.34 (m, 2H, H-5/H-6), 7.13 (d, J(3′, 2′/5′, 6′) = 8.0 Hz, 2H, H-3′/H-5′), 3.98 (s, 2H, S-CH2), 3.85 (s, 3H, -OCH3); 13C-NMR (150 MHz, DMSO-d6): δ 163.2, 157.2, 152.2, 149.8, 148.7, 141.7, 141.5, 140.5, 140.3, 136.3, 133.6, 130.6, 129.0, 129.0, 126.6, 126.2, 125.4, 124.6, 124.3, 116.8, 116.6, 114.7, 114.7, 56.1, 39.1.; HR-MS (ESI): (m/z) [M + H]+ calculated for C25H21N6O4S 501.1342, found 501.1351.

3.2.2. (E)-2-(2-(2-((1H-Benzo[d]imidazol-2-yl)thio)-1-(p-tolyl)ethylidene)hydrazinyl)-4-(2-nitrophenyl)oxazole (2)

Yield: 67%; Rf value 0.61 (7:3 n-hex: EtOAc); 1 H NMR (600 MHz, DMSO-d6): δ13.34 (s, 1H, -NH), 11.25 (s, 1H, -NH), 7.79 (dd, J(6″, 5″) = 8.4 Hz, J(6″, 4″) = 2.0 Hz, 1H, H-6″), 7.72 (d, J(3″, 4″) = 7.9 Hz, 1H, H-3″), 7.53 (s, 1H, oxazole-H), 7.36–7.33 (m, 1H, H-5″), 7.29 (d, J(2′, 3′/ 6′, 5′) = 7.5 Hz, 2H, H-2′/H-6′), 7.24 (t, J(4″/5″, 3″) = 9.2 Hz, 1H, H-4″), 7.22 (d, J(3′, 2′/5′, 6′) = 6.7 Hz, 2H, H-3′/H-5′), 7.11–7.08 (m, 2H, H-4/H-7), 6.98–6.96 (m, 2H, H-5/H-6), 2.50 (s, 2H, S-CH2), 1.22 (s, 3H, -CH3); 13C-NMR (150 MHz, DMSO-d6): δ 156.6, 151.6, 149.5, 148.1, 141.9, 141.1, 140.9, 139.9, 139.7, 136.0, 133.3, 132.4, 130.5, 130.3, 130.3, 128.2, 128.2, 125.9, 125.1, 124.0, 123.7, 116.2, 116.0, 38.5, 22.5.; HR-MS (ESI): (m/z) [M + H]+ calculated for C25H21N6O3S 485.1393, found 485.1401.

3.2.3. (E)-2-(2-(2-((1H-Benzo[d]imidazol-2-yl)thio)-1-(p-tolyl)ethylidene)hydrazinyl)-4-(3-nitrophenyl)oxazole (3)

Yield: 73%; Rf value 0.57 (7:3 n-hex: EtOAc); 1 H NMR (600 MHz, DMSO-d6): δ12.69 (s, 1H, -NH), 10.61 (s, 1H, -NH), 8.53 (t, J (2″/4″, 6″) = 2.6 Hz, 1H, H-2″), 8.33 (d, J (6″, 5″) = 6.6 Hz, 1H, H-6″), 8.22–8.16 (m, 1H, H-4″), 7.89 (t, J(5″/4″, 6″) = 7.8 Hz, 1H, H-5″), 7.84 (s, 1H, oxazole-H), 7.77 (d, J(2′, 3′/ 6′, 5′) = 8.2 Hz, 2H, H-2′/H-6′), 7.59–7.51 (m, 2H, H-4/H-7), 7.34 (d, J(3′, 2′/5′, 6′) = 7.5 Hz, 2H, H-3′/H-5′), 7.28–7.19 (m, 2H, H-5/H-6), 3.92 (s, 2H, S-CH2), 2.49 (s, 3H, -CH3).; 13C-NMR (150 MHz, DMSO-d6): δ 157.1, 152.1, 149.0, 148.6, 142.4, 141.6, 141.4, 140.4, 140.2, 134.5, 134.2, 132.7, 131.2, 130.8, 130.8, 128.7, 128.7, 124.7, 124.5, 124.2, 123.3, 116.7, 116.5, 39.0, 23.0.; HR-MS (ESI): (m/z) [M + H]+ calculated for C25H21N6O3S 485.1393, found 485.1401.

3.2.4. (E)-2-(2-(2-((1H-Benzo[d]imidazol-2-yl)thio)-1-(p-tolyl)ethylidene)hydrazinyl)-4-(p-tolyl)oxazole (4)

Yield: 74%; Rf value 0.63 (7:3 n-hex: EtOAc); 1 H NMR (600 MHz, DMSO-d6): δ12.66 (s, 1H, -NH), 10.58 (s, 1H, -NH), 7.83 (d, J(2′, 3′/ 6′, 5′) = 6.6 Hz, 2H, H-2′/H-6′), 7.81 (s, 1H, oxazole-H), 7.69 (d, J(2″, 3″/ 6″, 5″) = 7.9 Hz, 2H, H-2″/H-6″), 7.56–7.48 (m, 2H, H-4/H-7), 7.40 (d, J(3′, 2′/5′, 6′) = 7.0 Hz, 2H, H-3′/H-5′), 7.29–7.22 (m, 2H, H-5/H-6), 7.23 (d, J(3″, 2″/5″, 6″) = 8.5 Hz, 2H, H-3″/H-5″), 3.89 (s, 2H, S-CH2), 2.44 (s, 3H, -CH3), 2.54 (s, 3H, -CH3).; 13C-NMR (150 MHz, DMSO-d6): δ 165.9, 164.3, 162.7, 160.3,159.4, 157.07, 135.9, 131.0, 131.0, 130.6, 130.6, 130.2, 130.1,129.4, 129.3, 128.6, 128.3, 128.2, 127.7, 120.6, 116.1, 115.9, 115.8, 115.8, 115.7, 64.8, 40.0, 21.0, 15.1.; HR-MS (ESI): (m/z) [M + H]+ calculated for C26H24N5OS 454.1698, found 454.1707.

3.2.5. (E)-2-(2-(2-((1H-Benzo[d]imidazol-2-yl)thio)-1-(p-tolyl)ethylidene)hydrazinyl)-4-(3-methoxyphenyl)oxazole (5)

Yield: 76%; Rf value 0.56 (7:3 n-hex: EtOAc); 1 H NMR (600 MHz, DMSO-d6): δ12.80 (s, 1H, -NH), 10.72 (s, 1H, -NH), 7.95 (s, 1H, oxazole-H), 7.76–7.69 (m, 2H, H-4/H-7), 7.65 (d, J(2′, 3′/ 6′, 5′) = 9.4 Hz, 2H, H-2′/H-6′), 7.60–7.54 (m, 1H, H-6″), 7.52 (dd, J(5″, 4″) = 9.2 Hz, J(5″, 6″) = 9.0 Hz, 1H, H-5″), 7.44 (t, J (2″/4″, 6″) = 2.8 Hz, 1H, H-2″), 7.39 (m, 2H, H-5/H-6), 7.18 (d, J(3′, 2′/5′, 6′) = 8.6 Hz, 2H, H-3′/H-5′), 7.14 (d, J(4″, 5″) = 8.5 Hz, 1H, H-4″), 4.04 (s, 2H, S-CH2), 3.91 (s, 3H, -OCH3), 2.32 (s, 3H, -CH3).; 13C-NMR (150 MHz, DMSO-d6): δ 161.7, 155.2, 150.2, 146.7, 142.1, 139.7, 139.5, 138.5, 138.3, 134.6, 132.4, 130.8, 130.5, 130.5, 128.4, 128.4, 122.6, 122.3, 120.4, 114.9, 114.8, 114.6, 114.2, 56.4, 37.1, 22.7; HR-MS (ESI): (m/z) [M + H]+ calculated for C26H24N5O2S 470.1645, found 470.1656.

3.2.6. (E)-2-(2-(2-((1H-Benzo[d]imidazol-2-yl)thio)-1-(p-tolyl)ethylidene)hydrazinyl)-4-(3,4-dichlorophenyl)oxazole (6)

Yield: 67%; Rf value 0.58 (7:3 n-hex: EtOAc); 1 H NMR (600 MHz, DMSO-d6): δ12.80 (s, 1H, -NH), 10.72 (s, 1H, -NH), 8.09 (d, J(2″, 6″) = 2.8 Hz, 1H, H-2″), 7.98 (dd, J(6″, 5″) = 8.8 Hz, J(6″, 2″) = 1.7 Hz, 1H, H-6″), 7.93 (s, 1H, oxazole-H), 7.81 (d, J(2′, 3′/ 6′, 5′) = 8.4 Hz, 2H, H-2′/H-6′), 7.75–7.70 (m, 2H, H-4/H-7), 7.65 (d, J(5″, 6″) =7.5 Hz, 1H, H-5″), 7.45–7.39 (m, 2H, H-5/H-6), 7.33 (d, J(3′, 2′/5′, 6′) = 9.0 Hz, 2H, H-3′/H-5′), 4.04 (s, 2H, S-CH2), 2.48 (s, 3H, -CH3).; 13C-NMR (150 MHz, DMSO-d6): δ 157.6, 152.6, 149.1, 142.4, 142.1, 141.9, 140.9, 140.7, 134.4, 133.7, 133.5, 132.5, 131.7, 130.6, 130.6, 129.8, 128.5, 128.5, 128.0, 125.0, 124.7, 117.2, 117.0, 39.5, 22.8.; HR-MS (ESI): (m/z) [M + H]+ calculated for C25H20Cl2N5OS 508.0763, found 508.0771.

3.2.7. (E)-4-(2-((1H-Benzo[d]imidazol-2-yl)thio)-1-(2-(4-(4-bromophenyl)oxazol-2-yl)hydrazono)ethyl)-N,N-dimethylaniline (7)

Yield: 65%; Rf value 0.53 (7:3 n-hex: EtOAc); 1 H NMR (600 MHz, DMSO-d6): δ13.33 (s, 1H, -NH), 11.26 (s, 1H, -NH), 7.84 (s, 1H, oxazole-H), 7.77 (d, J(2″, 3″/ 6″, 5″) = 7.9 Hz, 2H, H-2″/H-6″), 7.55–7.53 (m, 2H, H-4/H-7), 7.45 (d, J(3″, 2″/5″, 6″) = 7.5 Hz, 2H, H-3″/H-5″), 7.36 (d, J(2′, 3′/ 6′, 5′) = 8.6 Hz, 2H, H-2′/H-6′), 7.33 (m, 2H, H-5/H-6), 7.10–6.84 (d, J(3′, 2′/5′, 6′) = 9.4 Hz, 2H, H-3′/H-5′), 3.81 (s, 6H, -N(CH3)2), 3.51 (s, 2H, S-CH2); 13C-NMR (150 MHz, DMSO-d6): δ 155.3, 150.3, 146.8, 142.6, 139.8, 139.6, 138.6, 138.4, 132.9, 132.5, 132.5, 131.0, 131.0, 130.1, 128.9, 128.9, 128.7, 128.7, 123.6, 122.7, 122.4, 114.9, 114.7, 37.2, 23.2.; HR-MS (ESI): (m/z) [M + H]+ calculated for C26H24BrN6OS 547.0911, found 547.0908.

3.2.8. (E)-2-(2-(2-((1H-Benzo[d]imidazol-2-yl)thio)-1-(p-tolyl)ethylidene)hydrazinyl)-4-(2-methoxyphenyl)oxazole (8)

Yield: 78%; Rf value 0.56 (7:3 n-hex: EtOAc); 1 H NMR (600 MHz, DMSO-d6): δ12.64 (s, 1H, -NH), 10.56 (s, 1H, -NH), 8.33 (d, J(6″, 5″) = 7.0 Hz, 1H, H-6″), 7.83 (d, J(2′, 3′/ 6′, 5′) = 7.7 Hz, 2H, H-2′/H-6′), 7.75 (s, 1H, oxazole-H), 7.55 (m, 2H, H-4/H-7), 7.52 (t, J(4″/5″, 3″) = 9.0 Hz, 1H, H-4″), 7.47 (dd, J(5″, 4″) = 8.8 Hz, J(5″, 6″) = 7.9 Hz, 1H, H-5″), 7.37 (d, J(3′, 2′/5′, 6′) = 6.8 Hz, 2H, H-3′/H-5′), 7.26 (d, J(3″, 4″) = 8.7 Hz, 1H, H-3″), 7.23 (m, 2H, H-5/H-6), 3.87 (s, 2H, S-CH2), 3.82 (s, 3H, -OCH3), 2.53 (s, 3H, -CH3).; 13C-NMR (150 MHz, DMSO-d6): δ 158.3, 156.9, 151.9, 148.4, 141.4, 141.2, 140.2, 142.0, 140.0, 132.5, 132.1, 130.7, 130.4, 130.4, 128.3, 128.3, 124.3, 124.0, 122.5, 119.9, 116.5, 116.3, 112.1, 57.1, 38.8, 22.6.; HR-MS (ESI): (m/z) [M + H]+ calculated for C26H24N5O2S 470.1645, found 470.1656.

3.2.9. (E)-4-(2-(2-(2-((1H-Benzo[d]imidazol-2-yl)thio)-1-(p-tolyl)ethylidene)hydrazinyl) Oxa-zol-4-yl)phenol (9)

Yield: 70%; Rf value 0.64 (7:3 n-hex: EtOAc); 1 H NMR (600 MHz, DMSO-d6): δ12.77 (s, 1H, -NH), 10.69 (s, 1H, -NH), 9.77 (s, 1H, -OH), 7.92 (s, 1H, oxazole-H), 7.68–7.63 (m, 2H, H-4/H-7), 7.58 (d, J(2′, 3′/ 6′, 5′) = 9.1 Hz, 2H, H-2′/H-6′), 7.48 (d, J(2″, 3″/ 6″, 5″) = 7.0 Hz, 2H, H-2″/H-6″), 7.36–7.29 (m, 2H, H-5/H-6), 7.17 (d, J(3′, 2′/5′, 6′) = 8.3 Hz, 2H, H-3′/H-5′), 6.96 (d, J(3″, 2″/5″, 6″) = 8.2 Hz, 2H, H-3″/H-5″), 4.01 (s, 2H, S-CH2), 2.31 (s, 3H, -CH3).; 13C-NMR (150 MHz, DMSO-d6): δ 159.5, 155.0, 150.0, 146.5, 142.5, 139.5, 139.3, 138.3, 138.1, 132.8, 130.9, 130.9, 129.9, 129.9, 128.8, 128.8, 124.3, 122.4, 122.1, 117.4, 117.4, 114.6, 114.4, 36.9, 23.1.; HR-MS (ESI): (m/z) [M + H]+ calculated for C25H22N5O2S 456.1488, found 456.1499.

3.2.10. (E)-2-(2-(2-((1H-Benzo[d]imidazol-2-yl)thio)-1-phenylethylidene)hydrazinyl)-4-(3-nitrophenyl)oxazole (10)

Yield: 73%; Rf value 0.57 (7:3 n-hex: EtOAc); 1 H NMR (600 MHz, DMSO-d6): δ12.82 (s, 1H, -NH), 10.74 (s, 1H, -NH), 8.52 (t, J (2″/4″, 6″) = 2.5 Hz, 1H, H-2″), 8.34 (d, J (6″, 5″) = 9.0 Hz, 1H, H-6″), 8.21–8.17 (m, 1H, H-4″), 8.07–8.01 (m, 2H, H-2′/H-6′), 7.88 (t, J(5″/4″,6″) = 8.1 Hz, 1H, H-5″), 7.78 (s, 1H, oxazole-H), 7.72 (m, 2H, H-4/H-7), 7.57–7.51 (m, 3H, H-3′/H-4′/H-5′), 7.41 (m, 2H, H-5/H-6), 4.06 (s, 2H, S-CH2).; 13C-NMR (150 MHz, DMSO-d6): δ 157.0, 152.0, 148.8, 148.5, 141.5, 141.3, 140.3, 140.1, 136.0, 134.3, 134.0, 133.0, 131.0, 130.8, 130.8, 130.2, 130.2, 124.5, 124.3, 124.0, 123.1, 116.6, 116.4, 38.9.; HR-MS (ESI): (m/z) [M + H]+ calculated for C24H19N6O3S 471.1237, found 471.1245.

3.2.11. (E)-2-(2-(2-((1H-Benzo[d]imidazol-2-yl)thio)-1-(4-chlorophenyl)ethylidene)hydrazi-nyl)-4-(3-nitrophenyl)oxazole (11)

Yield: 79%; Rf value 0.61 (7:3 n-hex: EtOAc); 1 H NMR (600 MHz, DMSO-d6): δ12.68 (s, 1H, -NH), 10.60 (s, 1H, -NH), 8.50 (t, J (2″/4″, 6″) = 2.0 Hz, 1H, H-2″), 8.31 (d, J (6″, 5″) = 7.7 Hz, 1H, H-6″), 8.19–8.13 (m, 1H, H-4″), 8.08 (d, J(2′, 3′/ 6′, 5′) = 9.0 Hz, 2H, H-2′/H-6′), 7.86 (t, J(5″/4″,6″) = 7.5Hz, 1H, H-5″), 7.83 (s, 1H, oxazole-H), 7.72 (d, J(3′, 2′/5′, 6′) = 8.4 Hz, 2H, H-3′/H-5′), 7.58 (m, 2H, H-4/H-7), 7.27 (m, 2H, H-5/H-6), 3.91 (s, 2H, S-CH2).; 13C-NMR (150 MHz, DMSO-d6): δ. 166.1, 163.1, 149.8, 41.2, 133.1, 132.2, 131.6, 131.6, 131.5, 130.5, 127.8, 127.6,127.5, 122.4, 122.3, 121.3, 120.9, 119.8, 119.6,111.0, 107.5, 40.0; HR-MS (ESI): (m/z) [M + H]+ calculated for C24H18ClN6O3S 508.0846, found 508.0855.

3.2.12. (E)-2-(2-(2-((1H-Benzo[d]imidazol-2-yl)thio)-1-(p-tolyl)ethylidene)hydrazinyl)-4-(4-chlorophenyl)oxazole (12)

Yield: 72%; Rf value 0.65 (7:3 n-hex: EtOAc); 1 H NMR (600 MHz, DMSO-d6): δ12.70 (s, 1H, -NH), 10.62 (s, 1H, -NH), 8.08 (d, J(2″, 3″/ 6″, 5″) = 6.9 Hz, 2H, H-2″/H-6″), 7.85 (s, 1H, oxazole-H), 7.75 (d, J(2′, 3′/ 6′, 5′) = 8.8 Hz, 2H, H-2′/H-6′), 7.63 (d, J(3″, 2″/5″, 6″) = 7.7 Hz, 2H, H-3″/H-5″), 7.57–7.50 (m, 2H, H-4/H-7), 7.34 (d, J(3′, 2′/5′, 6′) = 9.4 Hz, 2H, H-3′/H-5′), 7.28–7.18 (m, 2H, H-5/H-6), 3.93 (s, 2H, S-CH2), 2.47 (s, 3H, -CH3).; 13C-NMR (150 MHz, DMSO-d6): δ 166.1, 163.0, 161.3, 159.4, 156.7, 153.5, 136.9, 136.8, 135.7, 130.9, 130.9, 130.8, 129.4, 129.3, 128.8, 128.3, 127.9, 127.7, 126.2, 124.2, 122.2, 120.6, 117.5, 117.4, 115.7, 113.8, 113.7, 64.8, 15.1; HR-MS (ESI): (m/z) [M + H]+ calculated for C25H21N6O3S 485.1393, found 485.1401.

3.2.13. (E)-2-(2-(2-((1H-Benzo[d]imidazol-2-yl)thio)-1-(p-tolyl)ethylidene)hydrazinyl)-4-phenyloxazole (13)

Yield: 68%; Rf value 0.54 (7:3 n-hex: EtOAc); 1 H NMR (600 MHz, DMSO-d6): δ12.79 (s, 1H, -NH), 10.71 (s, 1H, -NH), 7.97 (s, 1H, oxazole-H), 7.91–7.85 (m, 2H, H-2″/H-6″), 7.80 (d, J(2′, 3′/ 6′, 5′) = 7.1 Hz, 2H, H-2′/H-6′), 7.74–7.68 (m, 2H, H-4/H-7), 7.63–7.56 (m, 3H, H-3″/H-4″/H-5″), 7.46 (d, J(3′, 2′/5′, 6′) = 6.9 Hz, 2H, H-3′/H-5′), 7.38–7.31 (m, 2H, H-5/H-6), 4.03 (s, 2H, S-CH2), 2.54 (s, 3H, -CH3).; 13C-NMR (150 MHz, DMSO-d6): δ 155.5, 150.5, 147.0, 143.4, 140.0, 139.8, 138.8, 138.6, 132.7, 131.7, 130.8, 130.8, 130.2, 130.2, 129.7, 128.7, 128.7, 125.7, 125.7, 122.9, 122.6, 115.1, 114.9, 37.4, 22.8.; HR-MS (ESI): (m/z) [M + H]+ calculated for C25H21ClN5OS 474.1151, found 474.1161.

3.2.14. (E)-2-(2-(2-((1H-Benzo[d]imidazol-2-yl)thio)-1-(p-tolyl)ethylidene)hydrazinyl)-4-(2,4-dichlorophenyl)oxazole (14)

Yield: 80%; Rf value 0.57 (7:3 n-hex: EtOAc); 1 H NMR (600 MHz, DMSO-d6): δ12.77 (s, 1H, -NH), 10.69 (s, 1H, -NH), 8.13 (d, J(6″, 5″) =8.8 Hz, 1H, H-6″), 7.92 (s, 1H, oxazole-H), 7.79 (d, J(2′, 3′/ 6′, 5′) = 7.8 Hz, 2H, H-2′/H-6′), 7.67–7.63 (m, 2H, H-4/H-7), 7.59 (d, J(3″, 5″) = 1.6 Hz, 1H, H-3″), 7.46 (dd, J(5″, 6″) = 8.4 Hz, J(5″, 3″) =1.8 Hz, 1H, H-5″), 7.41 (d, J(3′, 2′/5′, 6′) = 7.9 Hz, 2H, H-3′/H-5′), 7.36–7.30 (m, 2H, H-5/H-6), 4.01 (s, 2H, S-CH2), 2.51 (s, 3H, -CH3).; 13C-NMR (150 MHz, DMSO-d6): δ 157.5, 152.5, 149.0, 143.3, 142.0, 141.8, 140.8, 140.6, 136.7, 134.6, 132.6, 131.9, 131.3, 130.7, 130.7, 129.0, 128.6, 128.6, 128.4, 124.9, 124.6, 117.1, 116.9, 39.4, 22.9.; HR-MS (ESI): (m/z) [M + H]+ calculated for C25H20Cl2N5OS 508.0763, found 508.0771.

3.2.15. (E)-2-(2-(2-((1H-Benzo[d]imidazol-2-yl)thio)-1-phenylethylidene)hydrazinyl)-4-(3-methoxyphenyl)oxazole (15)

Yield: 74%; Rf value 0.60 (7:3 n-hex: EtOAc); 1 H NMR (600 MHz, DMSO-d6): δ12.71 (s, 1H, -NH), 10.63 (s, 1H, -NH), 7.86–7.79 (m, 2H, H-2′/H-6′), 7.73 (s, 1H, oxazole-H), 7.69–7.64 (m, 1H, H-6″), 7.60 (m, 2H, H-4/H-7), 7.51 (dd, J(5″, 4″) = 9.0 Hz, J(5″, 6″) = 9.3 Hz, 1H, H-5″), 7.47–7.38 (m, 3H, H-3′/H-4′/H-5′), 7.36 (t, J (2″/4″, 6″) = 2.6Hz, 1H, H-2″), 7.30 (m, 2H, H-5/H-6), 7.13 (d, J(4″, 5″) = 8.4 Hz, 1H, H-4″), 3.95 (s, 2H, S-CH2), 3.89 (s, 3H, -OCH3).; 13C-NMR (150 MHz, DMSO-d6): δ 161.6, 155.1, 150.1, 146.6, 139.6, 139.4, 138.4, 138.2, 136.5, 134.5, 133.5, 131.3, 131.3, 130.9, 130.7, 130.7, 122.5, 122.2, 120.3, 114.9, 114.7, 114.5, 114.1, 56.3, 37.0.; HR-MS (ESI): (m/z) [M + H]+ calculated for C25H22N5O2S 456.1487, found 456.1499.

3.2.16. (E)-2-(2-(2-((1H-Benzo[d]imidazol-2-yl)thio)-1-([1,1′-biphenyl]-4-yl)ethylidene) Hydrazinyl)-4-(p-tolyl)oxazole (16)

Yield: 67%; Rf value 0.52 (7:3 n-hex: EtOAc); 1 H NMR (600 MHz, DMSO-d6): δ11.27 (s, 1H, -NH), 10.04 (s, 1H, -NH), 8.27 (d, J(2′, 3′/ 6′, 5′) = 7.9 Hz, 2H, H-2′/H-6′), 8.15 (d, J(3′, 2′/5′, 6′) = 8.5 Hz, 2H, H-3′/H-5′), 7.99 (s, 1H, oxazole-H), 7.63 (d, J(2″, 3″/ 6″, 5″) = 8.7 Hz, 2H, H-2″/H-6″), 7.54 (d, J(3″, 2″/5″, 6″) = 8.1 Hz, 2H, H-3″/H-5″), 7.35–7.03 (m, 2H, H-4/H-7), 6.97–6.89 (m, 5H, para-Ph), 6.77–6.75 (m, 2H, H-5/H-6), 2.50 (s, 2H, S-CH2), 1.23 (s, 3H, -CH3).; 13C-NMR (150 MHz, DMSO-d6): δ 156.8, 151.8, 148.3, 143.6, 141.6, 141.3, 141.1, 140.1, 139.9, 133.3, 132.1, 129.9, 129.9, 129.7, 129.7, 129.5, 129.5, 128.5, 128.5, 128.1, 127.8, 127.7, 127.7, 126.1, 126.1, 124.2, 123.9, 116.4, 116.2, 38.7, 21.7.; HR-MS (ESI): (m/z) [M + H]+ calculated for C31H26N5OS 485.1854, found 516.1863.

3.2.17. (E)-2-(2-(2-((1H-Benzo[d]imidazol-2-yl)thio)-1-phenylethylidene)hydrazinyl)-4-(2-nitrophenyl)oxazole (17)

Yield: 81%; Rf value 0.62 (7:3 n-hex: EtOAc); 1 H NMR (600 MHz, DMSO-d6): δ12.67 (s, 1H, -NH), 10.59 (s, 1H, -NH), 8.57 (t, J (2″/4″, 6″) = 2.8Hz, 1H, H-2″), 8.38 (d, J (6″, 5″) = 6.6Hz, 1H, H-6″), 8.27–8.19 (m, 1H, H-4″), 7.99–7.94 (m, 2H, H-2′/H-6′), 7.91 (t, J(5″/4″,6″) = 7.8Hz, 1H, H-5″), 7.82 (s, 1H, oxazole-H), 7.57–7.51 (m, 2H, H-4/H-7), 7.48–7.39 (m, 3H, H-3′/H-4′/H-5′), 7.26–7.18 (m, 2H, H-5/H-6), 3.90 (s, 2H, S-CH2).; 13C-NMR (150 MHz, DMSO-d6): δ 157.7, 152.7, 149.4, 149.2, 142.2, 142.0, 141.0, 140.8, 136.3, 136.1, 133.2, 133.0, 131.0, 131.0, 130.7, 130.5, 130.3, 125.6, 125.4, 125.1, 124.8, 117.3, 117.1, 39.6.; HR-MS (ESI): (m/z) [M + H]+ calculated for C24H19N6O3S 471.1237, found 471.1245.

3.2.18. (E)-2-(2-(2-((1H-Benzo[d]imidazol-2-yl)thio)-1-(4-chlorophenyl)ethylidene)hydrazinyl)-4-(2-nitrophenyl)oxazole (18)

Yield: 83%; Rf value 0.58 (7:3 n-hex: EtOAc); 1 H NMR (600 MHz, DMSO-d6): δ12.74 (s, 1H, -NH), 10.66 (s, 1H, -NH), 8.52 (t, J (2″/4″, 6″) = 1.8Hz, 1H, H-2″), 8.31 (d, J (6″, 5″) = 6.9 Hz, 1H, H-6″), 8.24–8.17 (m, 1H, H-4″), 8.09 (d, J(2′, 3′/ 6′, 5′) = 9.5 Hz, 2H, H-2′/H-6′),7.90 (t, J(5″/4″,6″) = 8.6 Hz, 1H, H-5″), 7.87 (s, 1H, oxazole-H), 7.77 (d, J(3′, 2′/5′, 6′) = 8.6 Hz, 2H, H-3′/H-5′), 7.64–7.56 (m, 2H, H-4/H-7), 7.33–7.25 (m, 2H, H-5/H-6), 3.97 (s, 2H, S-CH2).; 13C-NMR (150 MHz, DMSO-d6): δ 157.8, 152.8, 149.3, 149.1, 142.3, 141.1, 141.1, 140.9, 138.4, 136.1, 133.5, 133.0, 130.8, 130.8, 130.6, 130.1, 130.1, 125.8, 125.4, 125.2, 124.9, 117.4, 117.2, 39.7.; HR-MS (ESI): (m/z) [M + H]+ calculated for C24H18ClN6O3S 508.0846, found 508.0855.

3.2.19. (E)-2-(2-(2-((1H-Benzo[d]imidazol-2-yl)thio)-1-(4-chlorophenyl)ethylidene)hydrazinyl)-4-(3-methoxyphenyl)oxazole (19)

Yield: 79%; Rf value 0.56 (7:3 n-hex: EtOAc); 1 H NMR (600 MHz, DMSO-d6): δ12.78 (s, 1H, -NH), 10.70 (s, 1H, -NH), 8.07 (d, J(2′, 3′/ 6′, 5′) = 9.2 Hz, 2H, H-2′/H-6′), 7.93 (s, 1H, oxazole-H), 7.74 (d, J(3′, 2′/5′, 6′) = 7.9 Hz, 2H, H-3′/H-5′), 7.68 (m, 2H, H-4/H-7), 7.63–7.56 (m, 1H, H-6″), 7.50 (dd, J(5″, 4″) = 8.3 Hz, J(5″, 6″) = 8.9 Hz, 1H, H-5″), 7.39 (t, J (2″/4″, 6″) = 1.8Hz, 1H, H-2″), 7.34–7.28 (m, 2H, H-5/H-6), 7.12 (d, J(4″, 5″) = 7.5 Hz, 1H, H-4″), 4.02 (s, 2H, S-CH2), 3.89 (s, 3H, -OCH3).; 13C-NMR (150 MHz, DMSO-d6): δ 161.9, 157.3, 152.3, 148.8, 141.8, 141.6, 140.6, 140.4, 138.7, 134.7, 133.2, 131.0, 131.0, 130.9, 130.3, 130.3, 124.7, 124.4, 120.6, 116.9, 116.7, 115.0, 114.4, 56.7, 39.2.; HR-MS (ESI): (m/z) [M + H]+ calculated for C25H21ClN5O2S 490.1098, found 490.1109.

3.3. Assay Protocol for Acetylcholinesterase and Butyrylcholinesterase Activities

According to previously established protocols, in vitro AChE and BuChE inhibition profiles were evaluated with slight modifications [33,34].

3.4. Protocol for Molecular Docking Study

In order to properly triangulate the in vitro and in silico results, a molecular docking analysis was carried out using the AutoDock Vina software (version 1.5.7) package to understand the binding mode of synthetic compounds against both the targeted AChE and BuChE enzymes. The RCSB protein databank’s crystal structures for both targets were obtained using the PDB codes 1ACL for AChE and 1P0P for BuChE. The crystallographic structures and all of the produced compounds were protonated and energy was reduced using the default MOE-Dock module parameters, resulting in optimized enzyme and compound structures. Docking research was then conducted using the improved enzyme and chemical structures. Our prior investigations covered all of the docking protocol’s specifics in depth [35,36].

4. Conclusions

In conclusion, we synthesized benzimidazole-based oxazole derivatives (1–19) and evaluated them against both AChE and BuChE enzymes, respectively. All of the synthesized analogues were evaluated for their inhibition profiles against the AChE and BuChE enzymes as compared to donepezil, the standard drug. All of the synthesized benzimidazole-based oxazole analogues showed a varied range of inhibitory potentials against targeted AChE and BuChE enzymes, with IC50 values ranging from 0.10 ± 0.050 to 12.60 ± 0.30 µM (for AChE) and 0.20 ± 0.050 µM to 16.30 ± 0.30 µM (for BuChE) when compared to standard donepezil (IC50 = 2.16 ± 0.12 µM for AChE) and 4.50 ± 0.11 µM for BuChE). Structure-activity relationship (SAR) studies revealed that analogues 9 (bearing para-CF3 on ring B and meta-NO2 substitution at ring C) and 14 (that holds para-CH3 substitutions on ring B and 2,4-diCl moieties on ring C) were the most potent analogues of the AChE and BuChE enzymes, with IC50 values of 0.10 ± 0.050 and 0.20 ± 0.050 µM (against AChE) and 0.20 ± 0.050 and 0.30 ± 0.050 µM (against BuChE), respectively. In order to explore the binding interactions possessed by active scaffolds with the active sites of amino acids of the targeted AChE and BuChE enzymes, the active analogues were also subjected to molecular docking studies. The results revealed that these active analogues provided numerous important interactions with the targeted enzymes’ active sites. All of the synthesized analogues were structurally elucidated using spectroscopic methods such as 1H-NMR, 13C-NMR, and HREI-MS.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/molecules28207015/s1. Figure S1: 1HNMR for the compound 2; Figure S2: 13CNMR for the compound 4; Figure S3: 1HNMR for the compound 7; Figure S4: 13CNMR for the compound 11; Figure S5: 13CNMR for the compound 12; Figure S6: 1HNMR for the compound 16.

Author Contributions

Conceptualization, F.R. and H.U.; methodology, R.H.; software, S.K.; validation, M.S. and R.I.; formal analysis, H.U.; investigation, H.U. and F.R.; data curation, M.S. and F.S.; writing—original draft preparation, R.H. and H.U.; writing—review and editing, R.I., F.S., M.S. and M.K.A.-S.; project administration, F.R. and H.U. All authors have read and agreed to the published version of the manuscript.

Funding

Supported by the Researchers Supporting Project Number (RSP2023R410), King Saud University, Riyadh, Saudi Arabia.

Acknowledgments

The authors would like to extend their sincere appreciation to the Researchers Supporting Project Number (RSP2023R410), King Saud University, Riyadh, Saudi Arabia.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Millard, C.B.; Broomfield, C.A. Anticholinesterases: Medical application of neurochemical principles. J. Neurochem. 1995, 64, 1909–1918. [Google Scholar] [CrossRef]
  2. Milatovifil, D.; Dettbarn, W.D. The role of carboxylesterase in toxicity and tolerance to organophosphorus anticholinesterases paraoxon and DFP. Toxicol. Appl. Pharmacol. 1966, 136, 20–28. [Google Scholar]
  3. Weinstock, M.; Groner, E. Rational design of a drug for Alzheimer’s disease with cholinesterase inhibitory and neuroprotective activity. Chem.-Biol. Interact. 2008, 175, 216–221. [Google Scholar] [CrossRef]
  4. Jann, M.W. Preclinical pharmacology of metrifonate. Pharmacother. J. Hum. Pharmacol. Drug Ther. 1998, 18, 55–67. [Google Scholar]
  5. Massoulié, J.; Pezzementi, L.; Bon, S.; Krejci, E.; Vallette, F.M. Molecular and cellular biology of cholinesterases. Prog. Neurobiol. 1993, 41, 31–91. [Google Scholar] [CrossRef] [PubMed]
  6. Aisen, P.S.; Davis, K.L. The search for disease-modifying treatment for Alzheimer’s disease. Neurology 1997, 48 (Suppl. 6), 35S–41S. [Google Scholar] [CrossRef]
  7. Whitehouse, P.J.; Price, D.L.; Struble, R.G.; Clark, A.W.; Coyle, J.T.; DeLong, M.R. Alzheimer’s disease and senile dementia: Loss of neurons in the basal forebrain. Science 1982, 215, 1237–1239. [Google Scholar] [CrossRef]
  8. Ullah, H.; Bibi, U.; Hussain, A.; Sarfraz, M.; Rahim, F.; Hayat, S.; Zada, H.; Khan, F.; Wadood, A. Synthesis and Molecular Docking Study of Bis-Indolylmethane Thiourea Derivatives as Anti-Alzheimer Agents. Russ. J. Org. Chem. 2023, 59, 181–189. [Google Scholar] [CrossRef]
  9. Maruish, M.E.; Moses, J.A. Clinical Neuropsychology: Theoretical Foundations for Practitioners; Psychology Press: London, UK, 2013; p. 371. [Google Scholar]
  10. Schetinger, M.R.; Porto, N.M.; Moretto, M.B.; Morsch, V.M.; da Rocha, J.B.T.; Vieira, V.; Moro, F.; Neis, R.T.; Bittencourt, S.; Bonacorso, H.G.; et al. New benzodiazepines alter acetylcholinesterase and ATPDase activities. Neurochem. Res. 2000, 25, 949–955. [Google Scholar] [CrossRef]
  11. Prandota, J. Unexplained liver damage, cryptogenic liver cirrhosis, and steatohepatitis may be caused by latent chronic toxoplasmosis. Toxicol. Appl. Pharmacol. 1973, 24, 29–100. [Google Scholar] [CrossRef]
  12. Kubo, K.; Inada, Y.; Kohara, Y.; Sugiura, Y.; Ojima, M.; Itoh, K.; Furukawa, Y.; Nishikawa, K.; Naka, T. Nonpeptide angiotensin II receptor antagonists. Synthesis and biological activity of benzimidazoles. J. Med. Chem. 1993, 36, 1772–1784. [Google Scholar] [CrossRef]
  13. Sabat, M.; VanRens, J.C.; Laufersweiler, M.J.; Brugel, T.A.; Maier, J.; Golebiowski, A.; De, B.; Easwaran, V.; Hsieh, L.C.; Walter, R.L.; et al. The development of 2-benzimidazole substituted pyrimidine based inhibitors of lymphocyte specific kinase (Lck). Bioorganic Med. Chem. Lett. 2006, 16, 5973–5977. [Google Scholar] [CrossRef] [PubMed]
  14. Güven, Ö.Ö.; Erdoğan, T.; Göker, H.; Yıldız, S. Synthesis and antimicrobial activity of some novel phenyl and benzimidazole substituted benzyl ethers. Bioorganic Med. Chem. Lett. 2007, 17, 2233–2236. [Google Scholar] [CrossRef]
  15. Townsend, L.B.; Devivar, R.V.; Turk, S.R.; Nassiri, M.R.; Drach, J.C. Design, synthesis, and antiviral activity of certain 2,5,6-Trihalo-1-(.beta.-D-ribofuranosyl) benzimidazoles. J. Med. Chem. 1995, 38, 4098–4105. [Google Scholar] [CrossRef]
  16. Valdez, J.; Cedillo, R.; Hernández-Campos, A.; Yepez, L.; Hernández-Luis, F.; Navarrete-Vazquez, G.; Tapia, A.; Cortés, R.; Hernández, M.; Castillo, R. Synthesis and antiparasitic activity of 1H-benzimidazole derivatives. Bioorganic Med. Chem. Lett. 2002, 12, 2221–2224. [Google Scholar] [CrossRef]
  17. Kuş, C.; Ayhan-Kılcıgil, G.; Özbey, S.; Kaynak, F.B.; Kaya, M.; Çoban, T.; Can-Eke, B. Synthesis and antioxidant properties of novel N-methyl-1,3,4-thiadiazol-2-amine and 4-methyl-2H-1,2,4-triazole-3 (4H)-thione derivatives of benzimidazole class. Bioorganic Med. Chem. 2008, 16, 4294–4303. [Google Scholar] [CrossRef]
  18. Arnaiz, D.O.; Liang, A.; Trinh, L.; Whitlow, M.; Koovakkat, S.K.; Shaw, K.J. Design, synthesis, and in vitro biological activity of indole-based factor Xa inhibitors. Bioorganic Med. Chem. Lett. 2000, 10, 957–961. [Google Scholar] [CrossRef]
  19. Selvakumar, S.; Babu, I.S.; Chidambaranathan, N. In-Vivo Central Nervous System-Locomotor Activity of Some Synthesized 2-[(1-((phenyl amino) methyl) Substituted 1-benzoimidazol-2-yl) alkyl] Isoindoline-1,3-diones. Biosci. Biotech. Res. Asia 2013, 10, 937–943. [Google Scholar] [CrossRef]
  20. Schulz, W.G.; Islam, I.; Skibo, E.B. Pyrrolo [1,2-a] benzimidazole-based quinones and iminoquinones. The role of the 3-substituent on cytotoxicity. J. Med. Chem. 1995, 38, 109–118. [Google Scholar] [CrossRef]
  21. Bansal, Y.; Silakari, O. The therapeutic journey of benzimidazoles: A review. Bioorganic Med. Chem. 2012, 20, 6208–6236. [Google Scholar] [CrossRef]
  22. Zhou, R.R.; Cai, Q.; Li, D.K.; Zhuang, S.Y.; Wu, Y.D.; Wu, A.X. Acid-Promoted Multicomponent Tandem Cyclization to Synthesize Fully Substituted Oxazoles via Robinson–Gabriel-Type Reaction. J. Org. Chem. 2017, 82, 6450–6456. [Google Scholar] [CrossRef]
  23. Taha, M.; Sadia, H.; Rahim, F.; Khan, M.I.; Hayat, S.; Iqbal, N.; Nawaz, F.; Ullah, H.; Zada, H.; Shah, S.A.A.; et al. Synthesis, biological evaluation and molecular docking study of oxindole based chalcone analogues as potent anti-Alzheimer agents. J. Mol. Struct. 2023, 1285, 135530. [Google Scholar] [CrossRef]
  24. Kakkar, S.; Kumar, S.; Lim, S.M.; Ramasamy, K.; Mani, V.; Shah, S.A.A.; Narasimhan, B. Design, synthesis and biological evaluation of 3-(2-aminooxazol-5-yl)-2H-chromen-2-one derivatives. Chem. Cent. J. 2018, 12, 130. [Google Scholar] [CrossRef]
  25. Khan, S.; Iqbal, S.; Khan, M.; Rehman, W.; Shah, M.; Hussain, R.; Rasheed, L.; Khan, Y.; Dera, A.A.; Pashameah, R.A.; et al. Design, synthesis, in silico testing, and in vitro evaluation of thiazolidinone-based benzothiazole derivatives as inhibitors of α-amylase and α-glucosidase. Pharmaceuticals 2022, 15, 1164. [Google Scholar] [CrossRef]
  26. Mumtaz, S.; Iqbal, S.; Shah, M.; Hussain, R.; Rahim, F.; Rehman, W.; Khan, S.; Abid, O.U.R.; Rasheed, L.; Dera, A.A.; et al. New triazinoindole bearing benzimidazole/benzoxazole hybrids analogs as potent inhibitors of urease: Synthesis, in vitro analysis and molecular docking studies. Molecules 2022, 27, 6580. [Google Scholar] [CrossRef]
  27. Rahim, F.; Ullah, H.; Taha, M.; Hussain, R.; Sarfraz, M.; Iqbal, R.; Iqbal, N.; Khan, S.; Ali Shah, S.A.; Albalawi, M.A.; et al. Synthesis of new triazole-based thiosemicarbazone derivatives as anti-Alzheimer’s disease candidates: Evidence-based in vitro study. Molecules 2022, 28, 21. [Google Scholar] [CrossRef]
  28. Alpan, A.S.; Parlar, S.; Carlino, L.; Tarikogullari, A.H.; Alptüzün, V.; Güneş, H.S. Synthesis, biological activity and molecular modeling studies on 1H-benzimidazole derivatives as acetylcholinesterase inhibitors. Bioorganic Med. Chem. 2013, 21, 4928–4937. [Google Scholar] [CrossRef]
  29. Hussain, R.; Ullah, H.; Rahim, F.; Sarfraz, M.; Taha, M.; Iqbal, R.; Rehman, W.; Khan, S.; Shah, S.A.A.; Hyder, S.; et al. Multipotent cholinesterase inhibitors for the treatment of Alzheimer’s disease: Synthesis, biological analysis and molecular docking study of benzimidazole-based thiazole derivatives. Molecules 2022, 27, 6087. [Google Scholar] [CrossRef] [PubMed]
  30. Coban, G.; Carlino, L.; Tarikogullari, A.H.; Parlar, S.; Sarıkaya, G.; Alptüzün, V.; Alpan, A.S.; Güneş, H.S.; Erciyas, E. 1H-benzimidazole derivatives as butyrylcholinesterase inhibitors: Synthesis and molecular modeling studies. Med. Chem. Res. 2016, 25, 2005–2014. [Google Scholar] [CrossRef]
  31. Šagud, I.; Maček Hrvat, N.; Grgičević, A.; Čadež, T.; Hodak, J.; Dragojević, M.; Lasić, K.; Kovarik, Z.; Škorić, I. Design, synthesis and cholinesterase inhibitory properties of new oxazole benzylamine derivatives. J. Enzym. Inhib. Med. Chem. 2020, 35, 460–467. [Google Scholar] [CrossRef] [PubMed]
  32. Hussain, R.; Khan, S.; Ullah, H.; Ali, F.; Khan, Y.; Sardar, A.; Iqbal, R.; Ataya, F.S.; El-Sabbagh, N.M.; Batiha, G.E.S. Benzimidazole-Based Schiff Base Hybrid Scaffolds: A Promising Approach to Develop Multi-Target Drugs for Alzheimer’s Disease. Pharmaceuticals 2023, 16, 1278. [Google Scholar] [CrossRef] [PubMed]
  33. Chigurupati, S.; Selvaraj, M.; Mani, V.; Selvarajan, K.K.; Mohammad, J.I.; Kaveti, B.; Bera, H.; Palanimuthu, V.R.; Teh, L.K.; Salleh, M.Z. Identification of novel acetylcholinesterase inhibitors: Indolopyrazoline derivatives and molecular docking studies. Bioorganic Chem. 2016, 67, 9–17. [Google Scholar] [CrossRef]
  34. Chigurupati, S.; Selvaraj, M.; Mani, V.; Mohammad, J.I.; Selvarajan, K.K.; Akhtar, S.S.; Marikannan, M.; Raj, S.; Teh, L.K.; Salleh, M.Z. Synthesis of azomethines derived from cinnamaldehyde and vanillin: In vitro aetylcholinesterase inhibitory, antioxidant and insilico molecular docking studies. Med. Chem. Res. 2018, 27, 807–816. [Google Scholar] [CrossRef]
  35. Wadood, A.; Shareef, A.; Ur Rehman, A.; Muhammad, S.; Khurshid, B.; Khan, R.S.; Shams, S.; Afridi, S.G. In silico drug designing for ala438 deleted ribosomal protein S1 (RpsA) on the basis of the active compound Zrl15. ACS Omega 2021, 7, 397–408. [Google Scholar] [CrossRef]
  36. Rehman, A.U.; Zhen, G.; Zhong, B.; Ni, D.; Li, J.; Nasir, A.; Gabr, M.T.; Rafiq, H.; Wadood, A.; Lu, S.; et al. Mechanism of zinc ejection by disulfiram in nonstructural protein 5A. Phys. Chem. Chem. Phys. 2021, 23, 12204–12215. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Available drug of AD with AChE inhibitory activity.
Figure 1. Available drug of AD with AChE inhibitory activity.
Molecules 28 07015 g001
Figure 2. Drug comprising benzimidazole moiety.
Figure 2. Drug comprising benzimidazole moiety.
Molecules 28 07015 g002
Figure 3. Bioactive drugs containing oxazole skeleton.
Figure 3. Bioactive drugs containing oxazole skeleton.
Molecules 28 07015 g003
Figure 4. Rational of the current study.
Figure 4. Rational of the current study.
Molecules 28 07015 g004
Scheme 1. Synthesis of benzimidazole-oxazole hybrid derivatives (119).
Scheme 1. Synthesis of benzimidazole-oxazole hybrid derivatives (119).
Molecules 28 07015 sch001
Figure 5. General structure of benzimidazole-oxazole hybrid derivatives (119).
Figure 5. General structure of benzimidazole-oxazole hybrid derivatives (119).
Molecules 28 07015 g005
Figure 6. The protein-ligand interaction (PLI) profile of compound 9 against the AChE enzyme and its 3D (left) and 2D (right) diagrams.
Figure 6. The protein-ligand interaction (PLI) profile of compound 9 against the AChE enzyme and its 3D (left) and 2D (right) diagrams.
Molecules 28 07015 g006
Figure 7. The protein-ligand interaction (PLI) profile of compound 9 against the BuChE enzyme and its 3D (left) and 2D (right) diagrams.
Figure 7. The protein-ligand interaction (PLI) profile of compound 9 against the BuChE enzyme and its 3D (left) and 2D (right) diagrams.
Molecules 28 07015 g007
Figure 8. Protein-ligand interaction profile (PLI) of the second most active compound 14 against the AChE enzyme and its 3D (left) and 2D (right) diagrams.
Figure 8. Protein-ligand interaction profile (PLI) of the second most active compound 14 against the AChE enzyme and its 3D (left) and 2D (right) diagrams.
Molecules 28 07015 g008
Figure 9. Protein-ligand interaction profile (PLI) of the second most active compound 14 against the BuChE enzyme and its 3D (left) and 2D (right) diagrams.
Figure 9. Protein-ligand interaction profile (PLI) of the second most active compound 14 against the BuChE enzyme and its 3D (left) and 2D (right) diagrams.
Molecules 28 07015 g009
Figure 10. Protein-ligand interaction profile (PLI) of the third most active compound 6 against the AChE enzyme and its 3D (left) and 2D (right) diagrams.
Figure 10. Protein-ligand interaction profile (PLI) of the third most active compound 6 against the AChE enzyme and its 3D (left) and 2D (right) diagrams.
Molecules 28 07015 g010
Figure 11. Protein-ligand interaction profile (PLI) of the third most active compound 6 against the BuChE enzyme and its 3D (left) and 2D (right) diagrams.
Figure 11. Protein-ligand interaction profile (PLI) of the third most active compound 6 against the BuChE enzyme and its 3D (left) and 2D (right) diagrams.
Molecules 28 07015 g011
Figure 12. Representation of the ADMET analysis of analog-6.
Figure 12. Representation of the ADMET analysis of analog-6.
Molecules 28 07015 g012
Figure 13. Representation of the ADMET analysis of analog-9.
Figure 13. Representation of the ADMET analysis of analog-9.
Molecules 28 07015 g013
Figure 14. Representation of the ADMET analysis of analog-14.
Figure 14. Representation of the ADMET analysis of analog-14.
Molecules 28 07015 g014
Table 1. Different substituents, AChE and BuChE activities of benzimidazole-oxazole hybrid derivatives (119).
Table 1. Different substituents, AChE and BuChE activities of benzimidazole-oxazole hybrid derivatives (119).
S.NOR1R2AChE
IC50 ± SEM a (µM)
BuChE
IC50 ± SEM a (µM)
1Molecules 28 07015 i001Molecules 28 07015 i0021.10 ± 0.101.80 ± 0.10
2Molecules 28 07015 i003Molecules 28 07015 i0041.60 ± 0.0101.90 ± 0.10
3Molecules 28 07015 i005Molecules 28 07015 i0064.40 ± 0.105.80 ± 0.10
4Molecules 28 07015 i007Molecules 28 07015 i0086.50 ± 0.208.30 ± 0.20
5Molecules 28 07015 i009Molecules 28 07015 i0109.80 ± 0.2011.50 ± 0.20
6Molecules 28 07015 i011Molecules 28 07015 i0120.40 ± 0.051.10 ± 0.05
7Molecules 28 07015 i013Molecules 28 07015 i01412.60 ± 0.3016.30 ± 0.30
8Molecules 28 07015 i015Molecules 28 07015 i0165.30 ± 0.207.20 ± 0.10
9Molecules 28 07015 i017Molecules 28 07015 i0180.10 ± 0.0500.20 ± 0.05
10Molecules 28 07015 i019Molecules 28 07015 i0201.40 ± 0.102.10 ± 0.10
11Molecules 28 07015 i021Molecules 28 07015 i0220.70 ± 0.0501.20 ± 0.010
12Molecules 28 07015 i023Molecules 28 07015 i0243.40 ± 0.105.70 ± 0.10
13Molecules 28 07015 i025Molecules 28 07015 i0268.10 ± 0.109.70 ± 0.10
14Molecules 28 07015 i027Molecules 28 07015 i0280.20 ± 0.0500.30 ± 0.050
15Molecules 28 07015 i029Molecules 28 07015 i0305.40 ± 0.107.20 ± 0.10
16Molecules 28 07015 i031Molecules 28 07015 i0326.10 ± 0.109.30 ± 0.10
17Molecules 28 07015 i033Molecules 28 07015 i0342.50 ± 0.103.50 ± 0.10
18Molecules 28 07015 i035Molecules 28 07015 i0360.80 ± 0.101.70 ± 0.10
19Molecules 28 07015 i037Molecules 28 07015 i0382.70 ± 0.203.70 ± 0.10
Standard drug Donepezil2.16 ± 0.124.5 ± 0.11
a SEM is the standard error mean.
Table 2. The different types of interactions between active analogues (9, 14, and 6) and interactive residues of amino acids of the targeted AChE and BuChE enzymes with varied distances.
Table 2. The different types of interactions between active analogues (9, 14, and 6) and interactive residues of amino acids of the targeted AChE and BuChE enzymes with varied distances.
Active
Analogues
Targeted
Enzymes
ReceptorsInteractionsBinding Affinities (kJ/mol)
Analogue-9AChEGLY-117HB−12.37
TYR-130HB
GLU-198π-cation
TRP-84π-π T-shaped
PHE-330π-π T-shaped, π-alkyl and HB
TYR-221HB and Carbon HB
ASP-72π-anion
TYR-70π-sulphur
TYR-334π-π stacked and π-anion
TRP-279π-alkyl
SER-286Halogen (fluorine)
BuChEGLU-238HB−11.97
ASN-245HB
ASN-241HB
PRO-281π-alkyl
VAL-229π-alkyl
PRO-359π-alkyl
TYR-282π-π stacked and halogen (fluorine)
GLY-283halogen (fluorine)
Analogue-14AChETYR-151π-sigma−11.17
HIS-305HB
ALA-198π-alkyl
LYS-200π-alkyl
ILE-235π-sigma
LEU-162π-alkyl
TRP-59π-alkyl
HIS-201π-cation
BuChETRP-329π-alkyl−10.87
PHE-601π-alkyl
ASP-568π-cation
TRP-432π-sigma
ASP-568Carbon HB
ASP-232π-cation
ALA-231π-alkyl
SER-505Carbon HB
ILE-233π-sigma and
π-π T shaped
LYS-506π-anion and
π-π T-shaped
LYS-506π-alkyl
PHE-476π-π T-shaped and π-alkyl
Analogue-6AChETYR-151π-sigma−10.12
GLY-306Carbon HB
HIS-305HB
ALA-198π-alkyl
LYS-200π-alkyl
ILE-235π-sigma
HIS-201π-cation
LEU-162π-alkyl
TRP-59π-alkyl
BuChEVAL-501π-alkyl−9.76
ILE-233π-alkyl
LYS-506π-alkyl
TRP-329π-π T-shaped
PHE-601π-π T-shaped
ASP-568π-anion
ASP-568HB
PHE-476π-π stacked and π-alkyl
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Hussain, R.; Rahim, F.; Ullah, H.; Khan, S.; Sarfraz, M.; Iqbal, R.; Suleman, F.; Al-Sadoon, M.K. Design, Synthesis, In Vitro Biological Evaluation and In Silico Molecular Docking Study of Benzimidazole-Based Oxazole Analogues: A Promising Acetylcholinesterase and Butyrylcholinesterase Inhibitors. Molecules 2023, 28, 7015. https://doi.org/10.3390/molecules28207015

AMA Style

Hussain R, Rahim F, Ullah H, Khan S, Sarfraz M, Iqbal R, Suleman F, Al-Sadoon MK. Design, Synthesis, In Vitro Biological Evaluation and In Silico Molecular Docking Study of Benzimidazole-Based Oxazole Analogues: A Promising Acetylcholinesterase and Butyrylcholinesterase Inhibitors. Molecules. 2023; 28(20):7015. https://doi.org/10.3390/molecules28207015

Chicago/Turabian Style

Hussain, Rafaqat, Fazal Rahim, Hayat Ullah, Shoaib Khan, Maliha Sarfraz, Rashid Iqbal, Faiza Suleman, and Mohammad Khalid Al-Sadoon. 2023. "Design, Synthesis, In Vitro Biological Evaluation and In Silico Molecular Docking Study of Benzimidazole-Based Oxazole Analogues: A Promising Acetylcholinesterase and Butyrylcholinesterase Inhibitors" Molecules 28, no. 20: 7015. https://doi.org/10.3390/molecules28207015

Article Metrics

Back to TopTop