Next Article in Journal
Melanin Synthesis Inhibition Activity of Compounds Isolated from Bamboo Shoot Skin (Phyllostachys pubescens)
Previous Article in Journal
The Atomic Structure and Mechanical Properties of ZIF-4 under High Pressure: Ab Initio Calculations
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Synthesis of New Triazole-Based Thiosemicarbazone Derivatives as Anti-Alzheimer’s Disease Candidates: Evidence-Based In Vitro Study

1
Department of Chemistry, Hazara University, Mansehra 21120, Pakistan
2
Department of Chemistry, University of Okara, Okara 56130, Pakistan
3
Department of Clinical Pharmacy, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia
4
Department of Zoology, Wildlife and Fisheries, Sub-Campus Toba Tek Singh, University of Agriculture Faisalabad, Punjab 36050, Pakistan
5
Department of Agronomy, Faculty of Agriculture and Environment, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan
6
Department of Chemistry, University of Poonch, Rawalakot 12350, Pakistan
7
Faculty of Pharmacy, Universiti Teknologi MARA Cawangan Selangor Kampus Puncak Alam, Bandar Puncak Alam 42300, Selangor, Malaysia
8
Atta-ur-Rahman Institute for Natural Product Discovery (AuRIns), Universiti Teknologi MARA Cawangan Selangor Kampus Puncak Alam, Bandar Puncak Alam 42300, Selangor, Malaysia
9
Department of Chemistry, Alwajh College, University of Tabuk, Tabuk 47512, Saudi Arabia
10
Department of Chemistry, Faculty of Science, University of Tabuk, Tabuk 71491, Saudi Arabia
11
Department of Biochemistry, Faculty of Science, University of Tabuk, Tabuk 71491, Saudi Arabia
12
Department of Biology, Faculty of Science, University of Tabuk, Tabuk 71491, Saudi Arabia
13
Biochemistry Section, Chemistry Department, Faculty of Science, Tanta University, Tanta 31527, Egypt
14
Department of Nutrition and Food Science, Faculty of Home Economics, University of Tabuk, Tabuk 71491, Saudi Arabia
15
Department of Home Economics, Faculty of Specific Education, Kafr ElSheikh University, Kafr ElSheikh 33516, Egypt
16
Department of Biotechnology, Faculty of Sciences, Taif University, Taif 21944, Saudi Arabia
17
H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
*
Authors to whom correspondence should be addressed.
Molecules 2023, 28(1), 21; https://doi.org/10.3390/molecules28010021
Submission received: 18 October 2022 / Revised: 27 November 2022 / Accepted: 8 December 2022 / Published: 20 December 2022
(This article belongs to the Section Organic Chemistry)

Abstract

:
Triazole-based thiosemicarbazone derivatives (6a–u) were synthesized then characterized by spectroscopic techniques, such as 1HNMR and 13CNMR and HRMS (ESI). Newly synthesized derivatives were screened in vitro for inhibitory activity against acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE) enzymes. All derivatives (except 6c and 6d, which were found to be completely inactive) demonstrated moderate to good inhibitory effects ranging from 0.10 ± 0.050 to 12.20 ± 0.30 µM (for AChE) and 0.20 ± 0.10 to 14.10 ± 0.40 µM (for BuChE). The analogue 6i (IC50 = 0.10 ± 0.050 for AChE and IC50 = 0.20 ± 0.050 µM for BuChE), which had di-substitutions (2-nitro, 3-hydroxy groups) at ring B and tri-substitutions (2-nitro, 4,5-dichloro groups) at ring C, and analogue 6b (IC50 = 0.20 ± 0.10 µM for AChE and IC50 = 0.30 ± 0.10 µM for BuChE), which had di-Cl at 4,5, -NO2 groups at 2-position of phenyl ring B and hydroxy group at ortho-position of phenyl ring C, emerged as the most potent inhibitors of both targeted enzymes (AChE and BuChE) among the current series. A structure-activity relationship (SAR) was developed based on nature, position, number, electron donating/withdrawing effects of substitution/s on phenyl rings. Molecular docking studies were used to describe binding interactions of the most active inhibitors with active sites of AChE and BuChE.

1. Introduction

Alzheimer’s disease (AD) is an irreversible, neurodegenerative and progressive disorder of the brain that diminishes the cholinergic system and results in disorientation, memory loss, impaired ability to solve problems and impaired cognition [1,2,3]. AD is the major cause of dementia in aging populations. The acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE) enzymes cause apoptosis of neuronal cells by plaques formed by aggregation of neurotoxic beta amyloid. They are involved in hydrolysis of acetylcholine to generate acetic acid and choline, leading to shortening of duration of acetylcholine in the hippocampus and cortex of the brain and thus facilitating normal regeneration of synapses and functioning. Therefore, targeting both AChE and BuChE enzymes is one approach for treatment of AD [4,5,6,7]. Two binding sites are present in AChE: the peripheral site, which is responsible for beta amyloid interaction, and the catalytic site, which causes hydrolysis of acetylcholine. Interactions of beta amyloid protein (Aβ) with AChE access the formation of beta amyloid protein-acetylcholinesterase (Aβ-AChE) complex and thus result in neurotoxicity. BuChE is found in liver, intestine, heart, kidney, serum and lungs, while AChE is present in cholinergic neurons, brain and muscle [8,9]. The cholinesterase enzymes perform key roles in the breakdown of compounds, having ester moieties in their core structures. Generally, AChE is dominant in brain, while BuChE functions when acetylcholine gradually decreases its function in the brain of AD patients. Hence, synthesis of drugs that function as inhibitors of both AChE and BuChE enzymes should be effective treatments of AD [10]. Several drugs have been approved by the Food and Drug Administration (FDA) for treatment of AD. These include galantamine and donepezil which are selective for AChE, while rivastigmine and tacrine inhibit both BuChE and AChE (Figure 1) [11].
Triazole analogues are reported to have therapeutic and biological activities, such as anticonvulsant [12], anti-inflammatory [13], antifungal [14], insecticidal [15] and plant growth regulation [16]. There are some important drugs, such as letrozole (anticancer), tazobactam (antibacterial), isavuconazole (antifungal), sitagliptin (antidiabetic), ribavirin (antiviral), and rufinamide (seizure disorder), that contain triazole moieties in their core structures (Figure 2) [17,18,19,20,21,22,23].
Recently, several classes of heterocyclic compounds have been reported to be potent inhibitors of acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE) [24,25,26,27,28]. Based on the biological importance of thiosemicarbazone [29,30] and triazole [31,32] compounds (Figure 3), it was decided to synthesize hybrid analogues based on triazole bearing thiosemicarbazone moiety as effective inhibitors of cholinesterase enzymes, such as AChE and BuChE, that could be effective treatments for AD.

2. Results and Discussion

2.1. Chemistry

Thiosemicarbazide (1) was treated with 4-nitrobenzoyl chloride in DMF in the presence of triethylamine and refluxed for 3 h to yield 2-(4-nitrobenzoyl)hydrazine-1-carbothioamide as the first intermediate (2), which further underwent cyclization during stirring overnight in 2% aqueous solution of sodium hydroxide followed by neutralization with dil. HCl to yield 1,2,4-triazole-3-thiole (3) as the second intermediate product. Intermediate (3) was then reacted with different substituted phenacyl bromide in ethanol in the presence of triethylamine and refluxed for 3 h to obtain the third intermediate (4). Intermediate (4) was then mixed with hydrazine hydrate in methanol in the presence of a few drops of glacial acetic acid to form intermediate (5). Finally, intermediate (5) was treated with different substituted isothiocyanate in tetrahydrofuran in the presence of triethylamine, with the resulting mixture stirred under reflux until conversion had been completed, as monitored by TLC during refluxing for 6–8 h. After being cooled to room temperature, the product was reacted with 5% Na2S2O3 and extracted with CH2Cl2/MeOH (10:1, 10 mL × 4). The combined organic layer was dried over anhydrous sodium sulfite and concentrated. The resulting residue was purified through silica gel column chromatography using a mixture of petroleum ether and EtOAc as eluent to yield the desired triazole-based thiosemicarbazone derivatives (6a–u) as the final product (Scheme 1, Table 1). Primary confirmation of the product was done using thin layer chromatography (TLC) and further confirmed by with nuclear magnetic resonance (NMR).

2.2. In Vitro Inhibition of Acetylcholinesterase and Butyrylcholinesterase Activities

All the newly synthesized derivatives of triazole-based thiosemicarbazone (6a–u) were screened in vitro for inhibition of acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE) activities. All the newly afforded derivatives, except 6c and 6d, which are found to be inactive, displayed good to moderate inhibition, with IC50 ranging from 0.10 ± 0.050 to 12.20 ± 0.30 µM against AChE and 0.20 ± 0.050 µM to 14.10 ± 0.40 µM against BuChE compared to the standard drug donepezil, which exhibited IC50 of 2.16 ± 0.12 and 4.5 ± 0.11 µM against AChE and BuChE, respectively (Table 1). A structure-activity relationship (SAR) based on substituent/s and electron donating/withdrawing effects on phenyl rings B and C was developed. The compounds were divided into five major parts: triazole moiety, ring A, thiosemicarbazone moiety, ring B, and ring C. Each part of the synthesized compounds was found to be actively participating in inhibition of both acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE). Furthermore, it was determined that by keeping the triazole, ring A, and thiosemicarbazone moieties constant, the variation in inhibitory potentials was determined by attachment of substituents of diverse nature at various positions in different number/s around both rings B and C (Figure 4, Table 1).

Structure–Activity Relationship (SAR) for Inhibition of Acetylcholinesterase (AChE) and Butyrylcholinesterase (BuChE)

Analogue 6i with IC50 = 0.10 ± 0.050 (for AChE) and IC50 = 0.20 ± 0.050 µM (for BuChE) having di-substitutions (2-nitro, 3-hydroxy groups) at ring B and tri-substitutions (2-nitro, 4,5-dichloro groups) at ring C emerged as the most active inhibitor of targeted AChE and BuChE enzymes, whereas analogue 6b (IC50 = 0.20 ± 0.10 µM for AChE) (IC50 = 0.30 ± 0.10 µM for BuChE) having di-Cl at 4,5- and—NO2 groups at 2-position of phenyl ring B and hydroxy group at ortho-position of phenyl ring C was recognized as the second-most active among the current synthesized series (Table 1). The greater number of attached electron-withdrawing groups, such as di-Cl and -NO2 groups around ring C, as well as the presence of substituents (-OH) capable of forming hydrogen bonds with the active residue of amino acids of these analogues were responsible for enhanced inhibitory potentials for both targeted AChE and BuChE. The majority of the electronic density is removed from Ph-ring B and C by these di-Cl and -NO2 groups, making it electron-deficient and further regaining stability through interactions with the active sites of targeted AChE and BuChE enzymes. The derivative 6k, which has a para-bromo substitution on ring B and a para-tolyl group at 4-position of aryl ring C, was shown to have the least inhibitory activity against AChE and BuChE enzymes with IC50 values of 12.20 ± 0.30 and 14.10 ± 0.40µM. This reduced the potency of derivative 6k, caused by the greater size of the attached substituent(s), which increased the crowdedness around both rings B and C and thus reduced the chance of interactions with catalytic residues of targeted enzymes (Table 1).
Derivatives 6a, 6b and 6l containing di-Cl groups at meta- and para-position and nitro group at ortho-position of ring C and a variety of other groups, including Br, OH, NO2 and CH3, at various position of ring B, improved inhibition of activities of both AChE and BuChE enzymes. Among these three derivatives, derivative 6b (IC50 = 0.20 ± 0.10 and 0.30 ± 0.10 µM) with hydroxy group at ortho-position on ring B along with tri-substitutions (2-nitro and 3,4-di-Cl groups) at aryl ring C displayed superior inhibition of AChE and BuChE, compared to derivative 6a (IC50 = 5.10 ± 0.20 and 6.40 ± 0.20 µM), which had a bromo group at ortho-position of ring B and derivative 6l (IC50 = 4.60 ± 0.010 and 5.90 ± 0.10 µM), bearing NO2 at ortho and CH3 groups at para on ring B along with di-Cl groups at meta- and para-position and nitro group at ortho-position of ring C (Table 1). These three derivatives contain tri-substitutions (2-nitro, 3,4-dichloro groups) at ring C, but have different substituents (Br, OH, CH3 and NO2 groups) around ring B. This diverse nature of substituents around ring B have different tendencies to interact with active site of targeted enzymes and hence cause variation in inhibitory potentials of these three derivatives (Table 1). Moreover, derivative 6a which contains a bromo at ortho on ring B and di-Cl groups at meta- and para-position and nitro group at ortho-position of ring C, exhibits better inhibition of AChE and BuChE activities than derivative 6h, which has a bromo moiety at para on ring B and ortho-nitro and para-methyl substitutions on ring C, might be due to di-Cl groups on ring, as well as different position of bromo moiety around ring B (Table 1).
Derivative 6g (IC50 = 2.10 ± 0.10 and 4.30 ± 0.10 µM), containing two methoxy groups at ortho- and meta-position of ring B and a nitro group at the para-position of the phenyl ring C, exhibited better inhibition of AChE and BuChE activities, compared to derivative 6f (IC50 = 2.40 ± 0.10 and 4.70 ± 0.10 µM), which had two methoxy groups at the ortho- and meta-positions of ring B and ortho-nitro and para-methyl groups on phenyl ring C (Table 1). This enhanced inhibition of AChE and BuChE activities of derivative 6g might be due to the stronger electron withdrawing nature of nitro group, making the ring C partially positive, which further established a pi-cation interaction with the active enzyme site. Alternatively, derivative 6f had both electron-donating (CH3) and electron-withdrawing (NO2) groups on ring C, which did not create charge on ring C and hence resulted in lesser activities of AChE and BuChE (Table 1).
Comparing derivative 6h (IC50 = 9.10 ± 0.20 and 11.20 ± 0.30 µM), which has a bromo group at the para position of the phenyl ring B and a nitro group at ortho-position and a methyl group at the para-position of the phenyl ring C, with derivatives 6m (IC50 = 2.70 ± 0.10 and 3.80 ± 0.10 µM) having di-Cl groups at meta- and para-positions of ring B and nitro group at ortho-position and methyl group at para-position on phenyl ring C and 6r (IC50 = 1.30 ± 0.050 and 2.20 ± 0.10 µM) having nitro group at ortho on ring B and nitro group at ortho-position and methyl group at para-position on phenyl ring C and 6t (IC50 = 1.90 ± 0.10 and 2.50 ± 0.10 µM) with a methoxy group at meta-position of ring B and the nitro group at ortho-position and the methyl group at para-position on phenyl ring C (Table 1). The small difference in the inhibitory activities (AChE and BuChE) of all derivatives might be due to the different nature and position of the substituent/s on phenyl ring B (Table 1).
Derivative 6n (IC50 = 0.70 ± 0. 05 and 1.70 ± 0.050 µM), which has di-chloro groups at meta- and para-positions on ring B and a nitro group at para-position on ring C, with derivative 6s (IC50 = 1.40 ± 0.050 and 2.30 ± 0.10µM), which has a nitro group at ortho position on ring B and nitro moiety at para position on ring C, and derivative 6u (IC50 = 2.90 ± 0.10 and 3.70 ± 0.10 µM), with a methoxy group at the meta-position on ring B and nitro moiety at the para position on ring C were compared (Table 1). Difference in the inhibitory activities (AChE and BuChE) of these derivatives might be due to the different nature and position of substituent/s on ring B (Table 1). Number, nature, position and electron-donating/withdrawing nature of substituents considerably influenced inhibition of activities.

2.3. Docking Study

Molecular docking was analyzed in order to gain an understanding of the binding mechanism of synthesized compounds against both the targeted enzymes. The optimized compounds were docked based on the co-crystal of each crystallographic structure. Each compound was assigned a total of 30 conformations prior to the docking process. For further investigation, the top-ranked conformations of potent compounds were chosen. The docking results revealed that all the compounds were well oriented in the active site of both enzymes. In general, we found that all of the compounds in the series—with different substituted groups at all three ends of the compound (according to the scheme), where one end has a nitro group, the second end has a halogen group, and the most important end (third) has a different substituted group—had inhibitory potential against the target. These typically belong to electron-withdrawing or electron-donating groups.
The protein–ligand interaction profile of the most active analogue, 6i, had several key interactions with catalytic residues of acetylcholinesterase enzyme, including Tyr332 (pi-sigma and pi-pi stacked), Trp82 (pi-anion), Glu197 (HB), Phe329 (pi-sulfur), Ser198 (HB), His438 (HB), Asn289 (HB), Asn68 (HB) and Asp70 (pi-cation) (Figure 5), while against butyrylcholinesterase enzymes, it interacts through numerous interactions such as Tyr116 (pi-alkyl), Tyr130 (pi-alkyl), His440 (CHB), Phe330 (pi-anion), Asp72 (pi-cation), Asn85 (HB), Tyr121 (pi-sulfur), Trp279 (pi-pi shaped and amide-pi stacked), Tyr70 (HB), Tyr334 (pi-pi stacked), Trp84 (pi-pi T shaped stacked), Gly118 (HB) and Gly117 (pi-pi stacked) (Figure 6).
As for the second-most active analogue, 6b, the protein–ligand interaction (PLI) profile showed that this scaffold enhanced inhibitory potential against AChE by interacting with the active part of the AChE enzyme through several significant interactions, such as Gly118 (CHB), Gly117 (pi-pi stacked), Ile444 (pi-alkyl), Tyr130 (pi-alkyl), Trp84 (pi-sigma and pi-pi stacked), Phe330 (pi-donor HB), His440 (pi-alkyl), Tyr121 (HB), Tyr334 (pi-sulfur and amide-pi stacked), Tyr70 (HB and pi-pi T shaped), Trp279 (pi-pi T shaped), Ser122 (HB) and Asp72 (pi-anion) (Figure 7), while against butyrylcholinesterase this analogue 6b adopted numerous key interactions including Pro285 (HB), His438 (pi-alkyl), Phe329 (pi-sulfur and pi-alkyl), Ala328 (alkyl), Gly117 (HB), Gly116 (HB), Thr120 (pi-sigma), Trp82 (pi-pi stacked), Gly115 (HB) and Tyr128 (HB) (Figure 8).
The protein–ligand interaction (PLI) profile of the third-most active analogue, 6q, showed that it established numerous interesting key interactions with active sites of the acetylcholinesterase enzyme, including Tyr128 (HB), Trp82 (pi-pi stacked), Trp430 (pi-alkyl), Tyr332 (pi-alkyl), Ala328 (pi-alkyl), pro285 (HB), Phe329 (pi-pi T shaped and pi-alkyl), Leu286 (pi-alkyl), Trp231 (pi-pi T shaped), Gly117 (HB), Ser198 (HB), Gly116 (HB), His438 (HB and pi-alkyl) and Asp70 (pi-anion) (Figure 9), while against the butyrylcholinesterase enzyme it established several important interactions, such as Tyr130 (HB), Tyr70 (pi-sulfur), Tyr338 (pi-pi T shaped), Arg289 (alkyl), Phe331 (pi-pi stacked and pi-sulfur), Trp279 (pi-pi stacked), Trp84 (pi-stacked), Phe330 (pi-pi stacked) and Gly117 (HB) as shown in (Figure 10).
Calculated binding energies, number of hydrogen bonds, and the number of closest residues surround the selected docked analogues into the active site of both AChE and BuChE enzymes are shown in Table 2.

3. Experimental

All chemicals and solvents were purchased from Sigma Aldrich (St. Louis, MO, USA) with a purity of 97 up to 99%.

3.1. General Procedure of 1,2,4-Triazole Bearing Thiosemicarbazone Derivatives (6a–u)

Thiosemicarbazide (1, 0.5 mmol) was treated with 4-nitrobenzoyl chloride (0.5 mmol) in DMF (10 mL) in the presence of triethylamine (0.5 mL) and refluxed for 3 h to yield 2-(4-nitrobenzoyl)hydrazine-1-carbothioamide as first intermediate (2), which further undergoes cyclization on stirring overnight in 2% aqueous solution of sodium hydroxide (10 mL followed by neutralization with dilute HCl (5 mL) to yield 1,2,4-triazole-3-thiole (3) as the second intermediate product. Intermediate (3) was then reacted with equivalent different substituted phenacyl bromide in ethanol (10 mL) in the presence of triethylamine (0.5 mL) and refluxed for 3 h to obtain third intermediate (4). Intermediate (4) was then mixed with hydrazine hydrate (5 mL) in methanol (10 mL) in the presence of a few drops of glacial acetic acid to give fourth intermediate (5). Finally intermediate (5) was treated with equivalent different substituted isothiocyanate in tetrahydrofuran (10 mL) in the presence of triethylamine (0.5 mL) and the resulting mixture was stirred under reflux until the conversion was completed (monitored by TLC, reflux 6–8 h). After being cooled to room temperature, it was reacted with 5% Na2S2O3 (20 mL) and extracted with CH2Cl2/MeOH (10:1, 10 mL × 4). The combined organic layer was dried over anhydrous sodium sulfate and concentrated. The given residue was purified through silica gel column chromatography using a mixture of petroleum ether and EtOAc as eluent to yield the desired triazole-based thiosemicarbazone derivatives (6a–u).

3.2. Spectral Analysis

All the proton NMR spectra are shown in Supplementary Materials.

3.2.1. (.E)-2-(1-(2-bromophenyl)-2-((5-(4-nitrophenyl)-4H-1,2,4-triazol-3-yl)thio)ethylidene)-N-(4,5-dichloro-2-nitrophenyl)hydrazine-1-carbothioamide (6a)

Yield: 63%; 1H NMR (600 MHz, DMSO-d6): δ 13.25 (s, 1H, NH), 11.81 (s, 1H, NH), 10.11 (s, 1H, NH), 8.28 (s, 1H, Ar-H), 8.10 (s, 1H, Ar-H), 7.88 (d, J = 8.88 Hz, 2H, Ar-H), 7.79 (d, J = 8.46 Hz, 2H, Ar-H), 7.43 (t, J = 7.92 Hz, 2H, Ar-H), 7.27 (t, J = 7.68 Hz, 1H, Ar-H), 7.04 (d, J = 7.56 Hz, 1H, Ar-H), 2.34 (s, 2H, S-CH2). 13C NMR (150 MHz, DMSO-d6): δ 184.0, 158.5, 157.3, 155.4, 147.5, 146.2, 138.4, 137.1, 135.2, 134.4, 132.5, 131.6, 130.0, 129.0, 127.5, 126.8, 126.8, 126.3, 125.5, 124.1, 124.1, 122.0, 31.0. HRMS (ESI) m/z: [M+H]+ calcd for C23H16BrCl2N8O4S2, 680.8117; Found, 680.8100.

3.2.2. (.E)-N-(4,5-dichloro-2-nitrophenyl)-2-(1-(2-hydroxyphenyl)-2-((5-(4-nitrophenyl)-4H-1,2,4-triazol-3-yl)thio)ethylidene)hydrazine-1-carbothioamide (6b)

Yield: 72%; 1HNMR (600 MHz, DMSO-d6): δ 13.39 (s, 1H, NH), 13.27 (s, 1H, NH), 12.01 (s, 1H, NH), 10.15 (s, 1H, OH), 8.91 (s, 1H, Ar-H), 8.30 (s, 1H, Ar-H), 8.16-7.87 (m, 4H, Ar-H), 7.83-7.72 (m, 2H, Ar-H), 7.45 (t, J = 7.74 Hz, 1H, Ar-H), 7.25 (t, J = 7.74 Hz, 1H, Ar-H), 2.28 (s, 2H, S-CH2). 13C NMR (150 MHz, DMSO-d6): δ 183.9, 162.5, 158.4, 157.2, 155.3, 147.4, 146.4, 138.3, 137.2, 132.4, 132.1, 131.5, 129.2, 127.1, 127.1, 126.4, 125.5, 124.1, 124.1, 121.0, 118.3, 117.0, 32.0. HRMS (ESI) m/z: [M+H]+ calcd for C23H17Cl2N8O5S2,619.0150; Found, 619.0134.

3.2.3. (.E)-2-(1-([1,1′-biphenyl]-4-yl)-2-((5-(4-nitrophenyl)-4H-1,2,4-triazol-3-yl)thio)ethylidene)-N-phenylhydrazine-1-carbothioamide (6c)

Yield: 75%; 1H NMR (600 MHz, DMSO-d6): δ 13.36 (s, 1H, -NH), 11.25 (s, 1H, -NH), 11.08 (s, 1H, -NH), 8.23 (d, J = 9.1 Hz, 2H, Ar-H), 7.99 (d, J = 9.1 Hz, 2H, Ar-H), 7.96 (d, J = 8.4 Hz, 2H, Ar-H), 7.86 (dd, J = 7.3 Hz, 2H, Ar-H), 7.72 (dd, J = 7.8 Hz, 1.9 Hz, 2H, Ar-H), 7.63-7.56 (m, 5H, Ar-H), 7.40-7.35 (m, 2H, Ar-H), 7.06-6.98 (m, 1H, Ar-H), 3.73 (s, 2H, S-CH2). 13C NMR (150 MHz, DMSO-d6): δ 183.8, 158.4, 157.2, 155.2, 147.5, 142.7, 140.4, 138.2, 138.1, 132.5, 129.3, 129.3, 128.8, 128.8, 128.6, 128.6, 128.0, 127.6, 127.6, 127.5, 127.5, 127.2, 126.6, 126.6, 126.1, 126.1, 124.0, 124.0, 31.4. HRMS (ESI) m/z: [M+H]+ calcd for C29H24N7O2S2,566.1429; Found, 566.1408.

3.2.4. (.E)-2-(1-([1,1′-biphenyl]-4-yl)-2-((5-(4-nitrophenyl)-4H-1,2,4-triazol-3-yl)thio)ethylidene)-N-(4′-methyl-[1,1′-biphenyl]-4-yl)hydrazine-1-carbothioamide (6d)

Yield: 73%; 1 H NMR (600 MHz, DMSO-d6): δ 13.38 (s, 1H, -NH), 11.24 (s, 1H, -NH), 11.07 (s, 1H, -NH), 8.22 (d, J = 6.8 Hz, 2H, Ar-H), 7.98 (d, J = 8.6 Hz, 2H, Ar-H), 7.95 (d, J = 7.2 Hz, 2H, Ar-H), 7.85 (dd, J = 7.8 Hz, 2H, Ar-H), 7.78-7.69 (m, 5H, Ar-H), 7.67 (d, J = 7.6 Hz, 2H, Ar-H), 7.63 (d, J = 7.3 Hz, 2H, Ar-H), 7.30 (d, J = 6.6 Hz, 2H, Ar-H), 7.12 (d, J = 6.7 Hz, 2H, Ar-H), 3.71 (s, 2H, S-CH2), 2.31 (s, 3H, -CH3). 13C NMR (150 MHz, DMSO-d6): δ 183.7, 158.3, 57.1, 155.1, 147.4, 142.6, 140.3, 139.3, 138.1, 137.3, 136.9, 132.4, 131.3, 130.1, 129.2, 129.2, 129.0, 129.0, 128.7, 128.7, 127.7, 127.7, 127.5, 127.5, 127.4, 127.4, 127.3, 127.3, 127.1, 126.5, 126.5, 124.1, 124.1, 123.9, 123.9, 20.8.; HRMS (ESI) m/z: [M+H]+ calcd for C36H30N7O2S2,656.1898; Found, 656.1877.

3.2.5. (.E)-2-(1-([1,1′-biphenyl]-4-yl)-2-((5-(4-nitrophenyl)-4H-1,2,4-triazol-3-yl)thio)ethylidene)-N-(3-nitrophenyl)hydrazine-1-carbothioamide (6e)

Yield: 83%; 1H NMR (600 MHz, DMSO-d6): δ 13.35 (s, 1H, -NH), 11.31 (s, 1H, -NH), 11.14 (s, 1H, -NH), 8.58 (dd, J = 2.4 Hz, 2.0 Hz, 1H, Ar-H), 8.29 (d, J = 9.0 Hz, 2H, Ar-H), 8.05 (d, J = 9.2 Hz, 2H, Ar-H), 8.02 (d, J = 8.5 Hz, 2H, Ar-H), 7.94 (dd, J = 7.5 Hz, 2H, Ar-H), 7.92–7.87 (m, 1H, Ar-H), 7.85–7.79 (m, 1H, Ar-H), 7.75–7.63 (m, 5H, Ar-H), 7.60 (t, J = 8.6 Hz, 1H, Ar-H), 3.79 (s, 2H, S-CH2). 13C NMR (150 MHz, DMSO-d6): δ 184.0, 158.6, 157.4, 155.4, 148.0, 147.7, 142.9, 140.6, 137.8, 136.4, 132.7, 132.4, 129.7, 129.5, 129.5, 129.0, 129.0, 127.8, 127.8, 127.7, 127.7, 127.4, 126.8, 126.8, 124.2, 124.2, 119.7, 119.2, 31.6. HRMS (ESI) m/z: [M+H]+ calcd for C29H23N8O4S2,611.1176; Found, 611.1159.

3.2.6. (.E)-2-(1-(2,5-dimethoxyphenyl)-2-((5-(4-nitrophenyl)-4H-1,2,4-triazol-3-yl)thio)ethylidene)-N-(4-methyl-2-nitrophenyl)hydrazine-1-carbothioamide (6f)

Yield: 80%; 1H NMR (600 MHz, DMSO-d6): δ 13.33 (s, 1H, -NH), 11.15 (s, 1H, -NH), 10.81 (s, 1H, -NH), 8.85 (d, J = 6.9 Hz, 1H, Ar-H), 8.33 (d, J = 2.1 Hz, 1H, Ar-H), 8.28 (d, J = 9.3 Hz, 2H, Ar-H), 8.05 (d, J = 9.0 Hz, 2H, Ar-H), 7.42 (d, J = 2.0 Hz, 1H, Ar-H), 7.06 (d, J = 7.1 Hz, 1H, Ar-H), 7.98 (dd, J = 7.1 Hz, 1.5 Hz, 1H, Ar-H), 7.01 (dd, J = 7.9 Hz, 1.7 Hz, 1H, Ar-H), 3.74 (s, 2H, S-CH2), 2.41 (s, 3H, CH3), 3.92 (s, 3H, -OCH3), 3.79 (s, 3H, -OCH3). 13C NMR (150 MHz, DMSO-d6): δ 184.1, 158.7, 157.5, 155.5, 152.9, 152.6, 147.8, 147.1, 138.5, 135.3, 135.2, 129.3, 126.9, 126.9, 125.2, 124.6, 124.2, 124.2, 118.1, 117.5, 115.3, 114.1, 32.0, 58.6, 58.3, 20.2. HRMS (ESI) m/z: [M+H]+ calcd for C26H25N8O6S2,609.1334; Found, 609.1320.

3.2.7. (.E)-2-(1-(2,5-dimethoxyphenyl)-2-((5-(4-nitrophenyl)-4H-1,2,4-triazol-3-yl)thio)ethylidene)-N-(4-nitrophenyl)hydrazine-1-carbothioamide (6g)

Yield: 80%; 1H NMR (600 MHz, DMSO-d6): δ 13.36 (s, 1H, -NH), 11.16 (s, 1H, -NH), 10.80 (s, 1H, -NH), 8.27 (d, J = 9.1 Hz, 2H, Ar-H), 8.18 (d, J = 6.9 Hz, 2H, Ar-H), 8.04 (d, J = 9.3 Hz, 2H, Ar-H), 7.74 (d, J = 6.8 Hz, 2H, Ar-H), 7.41 (d, J = 2.2 Hz, 1H, Ar-H),7.07 (d, J = 7.2 Hz, 1H, Ar-H), 7.02 (dd, J = 7.8 Hz, 1.8 Hz, 1H, Ar-H), 3.73 (s, 2H, S-CH2), 3.91 (s, 3H, -OCH3), 3.78 (s, 3H, -OCH3). 13C NMR (150 MHz, DMSO-d6): δ 184.4, 159.0, 157.8, 155.8, 153.2, 152.9, 148.1, 144.8, 144.1, 138.8, 127.2, 127.2, 125.0, 125.0, 124.6, 124.6, 124.4, 124.4, 118.4, 117.8, 115.6, 114.5, 56.0, 55.6, 32.3. HRMS (ESI) m/z: [M+H]+ calcd for C25H23N8O6S2,595.1179; Found, 595.1157.

3.2.8. (.E)-2-(1-(4-bromophenyl)-2-((5-(4-nitrophenyl)-4H-1,2,4-triazol-3-yl)thio)ethylidene)-N-(4-methyl-2-nitrophenyl)hydrazine-1-carbothioamide (6h)

Yield: 79%; 1H NMR (600 MHz, DMSO-d6): δ 13.32 (s, 1H, -NH), 11.06 (s, 1H, -NH), 10.72 (s, 1H, -NH), 8.78 (d, J = 7.0 Hz, 1H, Ar-H), 8.28 (d, J = 1.8 Hz, 1H, Ar-H), 8.25 (d, J = 9.5 Hz, 2H, Ar-H), 8.01 (d, J = 7.3 Hz, 2H, Ar-H), 7.73 (d, J = 7.6 Hz, 2H, Ar-H), 7.91 (dd, J = 8.3 Hz, 1.5 Hz, 1H, Ar-H), 7.67 (dd, J = 7.9 Hz, 2H, Ar-H), 3.73 (s, 2H, S-CH2), 2.40 (s, 3H, CH3). 13C NMR (150 MHz, DMSO-d6): δ 183.9, 158.5, 157.3, 155.3, 147.6, 146.9, 138.3, 135.1, 135.0, 132.7, 131.4, 131.4, 129.1, 128.3, 128.3, 126.7, 126.7, 125.1, 125.0, 124.3, 124.1, 124.1, 31.5, 20.1. HRMS (ESI) m/z: [M+H]+ calcd for C24H20BrN8O4S2,627.0227; Found, 627.0217.

3.2.9. (.E)-N-(4,5-dichloro-2-nitrophenyl)-2-(1-(3-hydroxy-2-nitrophenyl)-2-((5-(4-nitrophenyl)-4H-1,2,4-triazol-3-yl)thio)ethylidene)hydrazine-1-carbothioamide (6i)

Yield: 61%; 1H NMR (600 MHz, DMSO-d6): δ 13.39 (s, 1H, NH), 13.30 (s, 1H, NH), 12.21 (s, 1H, NH), 10.51 (s, 1H, -OH), 8.91 (s, 1H, Ar-H), 8.35 (s, 1H, Ar-H), 8.30 (d, J = 9.62 Hz, 1H, Ar-H), 8.27–8.03 (m, 1H, Ar-H), 7.91–7.85 (m, 1H, Ar-H), 7.83 (d, J = 9.0 Hz, 2H, Ar-H), 7.76 (d, J = 8.52 Hz, 2H, Ar-H), 2.34 (s, 2H, S-CH2). 13C NMR (150 MHz, DMSO-d6): δ 184.4, 158.3, 157.4, 155.2, 153.0, 148.6, 147.5, 139.2, 138.4, 137.1, 136.5, 136.0, 131.5, 129.0, 126.6, 126.6,126.3, 125.5, 124.0, 124.0, 121.4, 117.9, 30.4. HRMS (ESI) m/z: [M+H]+ calcd for C23H15Cl2N9O7S2, 662.9611; Found, 664.9600.

3.2.10. (.E)-2-(1-([1,1′-biphenyl]-4-yl)-2-((5-(4-nitrophenyl)-4H-1,2,4-triazol-3-yl)thio)ethylidene)-N-(2,5-dichloro-4-(dimethylamino)phenyl)hydrazine-1-carbothioamide (6j)

Yield: 64%; 1H NMR (600 MHz, DMSO-d6): δ 13.64 (s, 1H, NH), 11.54 (s, 1H, NH), 10.12 (s, 1H, NH), 8.23 (s, 1H, Ar-H), 8.18 (s, 1H, Ar-H), 8.04 (d, J = 7.26 Hz, 4H, Ar-H), 7.99–7.92 (m, 4H, Ar-H), 7.64–7.59 (m, 5H, Ar-H), 4.03 (s, 6H, -CH3), 2.51 (s, 2H, S-CH2). 13C NMR (150 MHz, DMSO-d6): δ 184.0, 158.5, 157.3, 155.2, 148.3, 147.5, 143.0, 140.6, 138.4, 134.2, 132.5, 129.6, 129.6, 129.0, 128.9, 128.9, 127.8, 127.8, 127.4, 127.4, 127.1, 127.1, 126.9, 126.9, 126.4, 124.2, 124.2, 120.6, 40.5, 40.5, 31.4. HRMS (ESI) m/z: [M+H]+ calcd for C31H27Cl2N8O2S2, 678.0719; Found, 678.0701.

3.2.11. (.E)-2-(1-(4-bromophenyl)-2-((5-(4-nitrophenyl)-4H-1,2,4-triazol-3-yl)thio)ethylidene)-N-(4′-methyl-[1,1′-biphenyl]-4-yl)hydrazine-1-carbothioamide (6k)

Yield: 73%; 1H NMR (600 MHz, DMSO-d6): δ 13.31 (s, 1H, -NH), 11.25 (s, 1H, -NH), 11.08 (s, 1H, -NH), 8.21 (d, J = 6.9 Hz, 2H, Ar-H), 7.97 (d, J = 8.9 Hz, 2H, Ar-H), 7.73 (d, J = 7.3 Hz, 2H, Ar-H), 7.59 (dd, J = 8.8 Hz, 2H, Ar-H), 7.68 (d, J = 7.7 Hz, 2H, Ar-H), 7.64 (d, J = 7.4 Hz, 2H, Ar-H), 7.31 (d, J = 6.7 Hz, 2H, Ar-H), 7.13 (d, J = 6.8 Hz, 2H, Ar-H), 3.72 (s, 2H, S-CH2), 2.32 (s, 3H, -CH3). 13C NMR (150 MHz, DMSO-d6): δ 184.0, 158.6, 157.4, 155.4, 147.7, 139.6, 138.4, 137.6, 137.2, 132.8, 131.5, 131.5, 130.4, 129.3, 129.3, 128.4, 128.4, 128.0, 128.0, 127.6, 127.6, 126.8, 126.8, 125.2, 124.4, 124.4, 124.2, 124.2, 31.6, 21.1. HRMS (ESI) m/z: [M+H]+ calcd for C30H25BrN7O2S2,658.0689; Found, 658.0669.

3.2.12. (.E)-N-(4,5-dichloro-2-nitrophenyl)-2-(1-(4-methyl-2-nitrophenyl)-2-((5-(4-nitrophenyl)-4H-1,2,4-triazol-3-yl)thio)ethylidene)hydrazine-1-carbothioamide (6l)

Yield: 67%; 1H NMR (600 MHz, DMSO-d6): δ 13.62 (s, 1H, NH), 13.38 (s, 1H, NH), 10.12 (s, 1H, NH), 8.90 (s, 1H, Ar-H), 8.23 (s, 1H, Ar-H), 8.16 (s, 1H, Ar-H), 8.02 (d, J = 5.58 Hz, 1H, Ar-H), 7.87 (d, J = 8.46 Hz, 1H, Ar-H), 7.75 (d, J = 8.4 Hz, 2H, Ar-H), 7.61 (d, J = 8.34 Hz, 2H, Ar-H), 2.50 (s, 2H, S-CH2), 1.91 (s, 3H, -CH3). 13C NMR (150 MHz, DMSO-d6): δ 184.0, 158.6, 157.4, 155.3, 147.5, 147.2, 146.5, 141.4, 138.4, 137.0, 135.1, 132.2, 131.6, 129.9, 129.0, 126.8, 126.8, 126.3, 125.4, 125.2, 124.2, 124.2, 30.5, 20.0. HRMS (ESI) m/z: [M+H]+ calcd for C24H18Cl2N9O6S2,662.0130; Found, 662.0111.

3.2.13. (.E)-2-(1-(3,4-dichlorophenyl)-2-((5-(4-nitrophenyl)-4H-1,2,4-triazol-3-yl)thio)ethylidene)-N-(4-methyl-2-nitrophenyl)hydrazine-1-carbothioamide (6m)

Yield: 61%; 1H NMR (600 MHz, DMSO-d6): δ 13.43 (s, 1H, -NH), 11.06 (s, 1H, -NH), 10.71 (s, 1H, -NH), 8.79 (d, J = 7.1 Hz, 1H, Ar-H), 8.29 (d, J = 1.7 Hz, 1H, Ar-H), 8.24 (d, J = 9.4 Hz, 2H, Ar-H), 8.02 (d, J = 7.5 Hz, 2H, Ar-H), 7.90 (dd, J = 8.4 Hz, 1.2Hz, 1H, Ar-H), 7.83 (d, J = 1.5 Hz, 1H, Ar-H), 7.80 (dd, J = 8.5 Hz, 1.9 Hz, 1H, Ar-H), 7.66 (d, J = 8.9 Hz, 1H, Ar-H), 3.72 (s, 2H, S-CH2), 2.40 (s, 3H, CH3). 13C NMR (150 MHz, DMSO-d6): δ 184.5, 159.1, 157.9, 155.9, 148.2, 147.5, 138.9, 136.0, 135.7, 135.6, 134.1, 133.8, 130.9, 130.6, 129.7, 127.3, 127.3, 126.6, 125.6, 124.7, 124.7, 124.5, 32.1, 20.6; HRMS (ESI) m/z: [M+H]+ calcd for C24H19Cl2N8O4S2,617.0341; Found, 617.0327.

3.2.14. (.E)-2-(1-(3,4-dichlorophenyl)-2-((5-(4-nitrophenyl)-4H-1,2,4-triazol-3-yl)thio)ethylidene)-N-(4-nitrophenyl)hydrazine-1-carbothioamide (6n)

Yield: 59%; 1H NMR (600 MHz, DMSO-d6): δ 13.30 (s, 1H, -NH), 11.21 (s, 1H, -NH), 11.01 (s, 1H, -NH), 8.28 (d, J = 8.8 Hz, 2H, Ar-H), 8.23 (d, J = 9.6 Hz, 2H, Ar-H), 8.03 (d, J = 7.8 Hz, 2H, Ar-H), 7.85 (d, J = 1.8 Hz, 1H, Ar-H), 7.81 (dd, J = 8.5 Hz, 2.7 Hz, 1H, Ar-H), 7.74 (d, J = 8.0 Hz, 2H, Ar-H), 7.68 (d, J = 9.0 Hz, 1H, Ar-H), 3.73 (s, 2H, S-CH2). 13C NMR (150 MHz, DMSO-d6): δ 184.5, 159.1, 157.9, 155.9, 148.2, 144.2, 144.9, 138.9, 136.0, 134.0, 133.8, 130.9, 130.6, 127.3, 127.3, 126.6, 124.7, 124.7, 124.5, 124.5, 125.1, 125.1, 32.1; HRMS (ESI) m/z: [M+H]+ calcd for C23H17Cl2N8O4S2,603.0183; Found, 603.0169.

3.2.15. (.E)-2-(1-(3,4-dichlorophenyl)-2-((5-(4-nitrophenyl)-4H-1,2,4-triazol-3-yl)thio)ethylidene)-N-phenylhydrazine-1-carbothioamide (6o)

Yield: 62%; 1H NMR (600 MHz, DMSO-d6): δ 13.41 (s, 1H, -NH), 11.31 (s, 1H, -NH), 11.15 (s, 1H, -NH), 8.29 (d, J = 6.8 Hz, 2H, Ar-H), 7.97 (d, J = 6.4 Hz, 2H, Ar-H), 7.90 (dd, J = 8.6 Hz, 2.1 Hz, 1H, Ar-H), 7.88 (d, J = 1.9 Hz, 1H, Ar-H), 7.73 (d, J = 8.0 Hz, 1H, Ar-H), 7.70 (dd, J = 7.7 Hz, 1.5 Hz, 2H, Ar-H), 7.43–7.35 (m, 2H, Ar-H), 7.08–6.99 (m, 1H, Ar-H), 3.74 (s, 2H, S-CH2). 13C NMR (150 MHz, DMSO-d6): δ 184.4, 159.0, 157.8, 155.8, 148.1, 138.8, 138.7, 135.9, 133.9, 133.7, 130.8, 130.5, 129.2, 129.2, 128.6, 127.2, 127.2, 126.7, 126.7, 126.5, 124.6, 124.6, 32.0. HRMS (ESI) m/z: [M+H]+ calcd for C23H18Cl2N7O2S2,558.0337; Found, 558.0326.

3.2.16. (.E)-2-(1-(3,4-dichlorophenyl)-2-((5-(4-nitrophenyl)-4H-1,2,4-triazol-3-yl)thio)ethylidene)-N-(4′-methyl-[1,1′-biphenyl]-4-yl)hydrazine-1-carbothioamide (6p)

Yield: 64%; 1H NMR (600 MHz, DMSO-d6): δ 13.42 (s, 1H, -NH), 11.23 (s, 1H, -NH), 11.06 (s, 1H, -NH), 8.21 (d, J = 7.9 Hz, 2H, Ar-H), 7.97 (d, J = 8.7 Hz, 2H, Ar-H), 7.67 (d, J = 7.7 Hz, 2H, Ar-H), 7.91 (dd, J = 8.5 Hz, 2.0 Hz, 1H, Ar-H), 7.89 (d, J = 1.8 Hz, 1H, Ar-H), 7.74 (d, J = 8.1 Hz, 1H, Ar-H), 7.64 (d, J = 7.4 Hz, 2H, Ar-H), 7.31 (d, J = 6.7 Hz, 2H, Ar-H), 7.13 (d, J = 6.8 Hz, 2H, Ar-H), 3.72 (s, 2H, S-CH2), 2.30 (s, 3H, -CH3). 13C NMR (150 MHz, DMSO-d6): δ 184.3, 158.6, 157.4, 155.4, 147.7, 139.6, 138.4, 137.6, 137.2, 135.4, 134.2, 133.3, 130.4, 130.2, 130.1, 129.3, 129.3, 128.0, 128.0, 127.6, 127.6, 126.8, 126.8, 126.1, 124.4, 124.4, 124.2, 124.2, 31.7, 21.1. HRMS (ESI) m/z: [M+H]+ calcd for C30H24Cl2N7O2S2,648.0806; Found, 648.0790.

3.2.17. (.E)-2-(1-(3,4-dichlorophenyl)-2-((5-(4-nitrophenyl)-4H-1,2,4-triazol-3-yl)thio)ethylidene)-N-(3-nitrophenyl)hydrazine-1-carbothioamide (6q)

Yield: 67%; 1H NMR (600 MHz, DMSO-d6): δ 13.36 (s, 1H, -NH), 11.29 (s, 1H, -NH), 11.13 (s, 1H, -NH), 8.58 (dd, J = 2.3 Hz, 2.0 Hz, 1H, Ar-H), 8.27 (d, J = 8.1 Hz, 2H, Ar-H), 8.03 (d, J = 9.4 Hz, 2H, Ar-H), 7.93 (dd, J = 8.4 Hz, 2.6 Hz, 1H, Ar-H), 7.88 (d, J = 1.9 Hz, 1H, Ar-H), 7.83–7.78 (m, 1H, Ar-H), 7.73 (d, J = 8.3 Hz, 1H, Ar-H), 7.69–7.64 (m, 1H, Ar-H), 7.62 (t, J = 8.7 Hz, 1H, Ar-H), 3.79 (s, 2H, S-CH2). 13C NMR (150 MHz, DMSO-d6): δ 184.4, 159.0, 157.8, 155.8, 148.4, 148.1, 138.8, 138.2, 135.9, 133.9, 133.7, 132.8, 130.8, 130.5, 130.1, 127.2, 127.2, 126.5, 124.6, 124.6, 120.1, 119.6, 32.0.; HRMS (ESI) m/z: [M+H]+ calcd for C23H17Cl2N8O4S2,603.0187; Found, 603.0162.

3.2.18. (.E)-N-(4-methyl-2-nitrophenyl)-2-(1-(2-nitrophenyl)-2-((5-(4-nitrophenyl)-4H-1,2,4-triazol-3-yl)thio)ethylidene)hydrazine-1-carbothioamide (6r)

Yield: 69%; 1H NMR (600 MHz, DMSO-d6): δ 13.33 (s, 1H, -NH), 11.06 (s, 1H, -NH), 10.72 (s, 1H, -NH), 8.77 (d, J = 7.1 Hz, 1H, Ar-H), 8.29 (d, J = 1.7 Hz, 1H, Ar-H), 8.24 (d, J = 9.6 Hz, 2H, Ar-H), 8.14 (dd, J = 7.8 Hz, 2.9 Hz 1H, Ar-H), 8.07 (dd, J = 7.5 Hz, 2.6 Hz 1H, Ar-H), 8.02 (d, J = 7.3 Hz, 2H, Ar-H), 7.99–7.93 (m, 1H, Ar-H), 7.91 (dd, J = 8.3 Hz, 1.5 Hz, 1H, Ar-H), 7.66 (dt, J = 8.5 Hz, 1.8 Hz 1H, Ar-H), 3.73 (s, 2H, S-CH2), 2.40 (s, 3H, CH3). 13C NMR (150 MHz, DMSO-d6): δ 184.6, 159.2, 158.0, 156.0, 148.3, 147.6, 139.0, 137.7, 135.8, 135.7, 135.3, 132.3, 132.2, 129.8, 127.4, 127.4, 126.8, 125.8, 125.7, 124.8, 124.8, 124.6, 31.0, 20.7; HRMS (ESI) m/z: [M+H]+ calcd for C24H20N9O6S2,594.0974; Found, 594.0960.

3.2.19. (.E)-N-(4-nitrophenyl)-2-(1-(2-nitrophenyl)-2-((5-(4-nitrophenyl)-4H-1,2,4-triazol-3-yl)thio)ethylidene)hydrazine-1-carbothioamide (6s)

Yield: 63%; 1H NMR (600 MHz, DMSO-d6): δ 13.38 (s, 1H, -NH), 11.34 (s, 1H, -NH), 11.17 (s, 1H, -NH), 8.32 (d, J = 9.4 Hz, 2H, Ar-H), 8.19 (dd, J = 9.0 Hz, 2H, Ar-H), 8.12 (dd, J = 6.8 Hz, 1.9 Hz 1H, Ar-H), 8.08 (d, J = 9.6 Hz, 2H, Ar-H), 8.03 (dd, J = 8.5 Hz, 1.6 Hz 1H, Ar-H), 7.94–7.88 (m, 1H, Ar-H), 7.76 (d, J = 7.8 Hz, 2H, Ar-H), 7.64 (dt, J = 8.3 Hz, 1.7 Hz, 1H, Ar-H), 3.82 (s, 2H, S-CH2). 13C NMR (150 MHz, DMSO-d6): δ 184.4, 159.0, 157.8, 155.8, 148.1, 144.8, 144.1, 138.8, 135.6, 134.5, 132.1, 132.0, 127.2, 127.2, 126.6, 125.6, 125.0, 125.0, 124.6, 124.6, 124.4, 124.4, 31.0. HRMS (ESI) m/z: [M+H]+calcd for C23H18N9O6S2,580.0818; Found, 580.0806.

3.2.20. (.E)-2-(1-(3-methoxyphenyl)-2-((5-(4-nitrophenyl)-4H-1,2,4-triazol-3-yl)thio)ethylidene)-N-(4-methyl-2-nitrophenyl)hydrazine-1-carbothioamide (6t)

Yield: 59%; 1H NMR (600 MHz, DMSO-d6): δ 13.37 (s, 1H, -NH), 11.14 (s, 1H, -NH), 10.81 (s, 1H, -NH), 8.85 (d, J = 7.7 Hz, 1H, Ar-H), 8.35 (d, J = 1.6 Hz, 1H, Ar-H), 8.30 (d, J = 9.4 Hz, 2H, Ar-H), 8.06 (d, J = 9.2 Hz, 2H, Ar-H), 7.97 (dd, J = 7.3 Hz, 1.8 Hz, 1H, Ar-H), 7.65 (dt, J = 7.7 Hz, 2.3 Hz, 1H, Ar-H), 7.43 (dd, J = 2.1 Hz, 1.3 Hz, 1H, Ar-H), 7.34 (dd, J = 6.3 Hz, J = 6.6 Hz, 1H, Ar-H), 7.10–6.99 (m, 1H, Ar-H), 3.74 (s, 3H, -OCH3), 3.80 (s, 2H, S-CH2), 2.45 (s, 3H, CH3). 13CNMR (150 MHz, DMSO-d6): δ 184.0, 160.5, 158.6, 157.4, 155.4, 147.7, 147.0, 138.4, 135.2, 135.1, 134.8, 129.6, 129.2, 126.8, 126.8, 125.1, 124.4, 124.2, 124.2, 120.3, 116.4, 113.1, 55.6, 31.6, 20.1. HRMS (ESI) m/z: [M+H]+ calcd for C25H23N8O5S2,579.1229; Found, 579.1213.

3.2.21. (.E)-2-(1-(3-methoxyphenyl)-2-((5-(4-nitrophenyl)-4H-1,2,4-triazol-3-yl)thio)ethylidene)-N-(4-nitrophenyl)hydrazine-1-carbothioamide (6u)

Yield: 61%; 1H NMR (600 MHz, DMSO-d6): δ 13.40 (s, 1H, -NH), 11.34 (s, 1H, -NH), 11.07 (s, 1H, -NH), 7.76 (d, J = 6.9 Hz, 2H, Ar-H), 8.33 (d, J = 9.6 Hz, 2H, Ar-H), 8.13 (dd, J = 8.8 Hz, 2H, Ar-H), 8.08 (d, J = 9.2 Hz, 2H, Ar-H), 7.68 (dt, J = 8.7 Hz, J = 2.7 Hz, 1H, Ar-H), 7.44 (dd, J = 2.2 Hz, 1.4 Hz, 1H, Ar-H), 7.35 (dd, J = 6.8 Hz, 7.0 Hz, 1H, Ar-H), 7.13–6.95 (m, 1H, Ar-H), 3.81 (s, 3H, -OCH3), 3.78 (s, 2H, S-CH2). 13C NMR (150 MHz, DMSO-d6): δ 184.5, 161.0, 159.1, 157.9, 155.9, 148.2, 144.9, 144.2, 138.9, 135.3, 130.1, 127.3, 127.3, 125.1, 125.1, 124.7, 124.7, 124.5, 124.5, 120.8, 116.9, 113.6, 56.1, 32.1. HRMS (ESI) m/z: [M+H]+ calcd for C24H21N8O5S2,565.1073; Found, 565.1055.

3.3. Molecular Docking Protocol

Molecular docking was studied using MOE software to understand the binding mode of synthesized compounds against both the targeted enzymes in order to triangulate in vitro and in silico results well. The crystal structures of both targets were retrieved from the RCSB protein databank using the PDB codes 1ACL for AChE and 1P0P for BuChE. Using the default MOE-Dock module parameters, the crystallographic structures and all synthesized compounds were protonated and energy was minimized, resulting in optimized enzyme and compound structures. After that, the optimized enzyme and compound structures were used in a docking study. Comprehensive details of the docking protocol are included in our previous investigations [33,34].

3.4. Acetylcholinesterase Activity Assay Protocol

Based on previously described methods, in vitro study of AChE inhibitory profile was calculated [35,36]. In order to make a stock solution, compounds being analyzed were dissolved in DMSO (1 mg/mL). In addition, the working solutions were also prepared by using serial dilution (1–100 μg/mL). The solution of AChE enzyme (20 μL; 0.1 U/mL), analogues being tested with different concentration and buffer of sodium phosphate (150 μL; pH 8.0; 0.1 M) were preincubated appropriately at 25 °C. The process was initiated as DTNB (10 mM; 10 μL) and AChEI (14 mM; 10 μL) were accumulated. The resulting residue was mixed (by using cyclomixer) and put on incubation for 10 min at 25 °C. Instead of compounds being tested using 10 μL DMSO, the absorbance against blank reading was calculated with a microplate reader at 410 nm. Using the given formula as given, inhibition and IC50 values were measured in comparison to donepezil (0.01–100 μg/mL) as the reference standard (Equation (1)).
% Inhibition = Absorbance   of   control Absorbance   of   compound × 100 Acontrol
By plotting a nonlinear graph between inhibition and concentration, IC50 was calculated using GraphPad Prism 5.3.

3.5. Butyrylcholinesterase Activity Assay Protocol

In order to explore the inhibition profile of in vitro BuChE enzyme, a similar procedure was adopted. For measurement of BuChE activity, the solution containing BuChE enzyme was used [35,36].

4. Conclusions

Triazole-based thiosemicarbazone derivatives (6a–u) were synthesized and screened for potential to inhibit activities of acetylcholinesterase and butyrylcholinesterase enzymes. All the synthetic derivatives (except compounds 6c and 6d, which were found to be completely inactive) displayed moderate to good inhibitory activities having an IC50 values ranging from 0.20 ± 0.050 to 12.20 ± 0.30 µM (against AChE) and 0.40 ± 0.050 to 14.10 ± 0.40 µM (against BuChE) compared to the standard drug donepezil (IC50 = 2.16 ± 0.12 (AChE) and 4.5 ± 0.11 µM (BuChE)). Among the series, derivative 6q (IC50 = 0.20 ± 0.050 µM) was the most potent inhibitor of acetylcholinesterase enzyme, while derivative 6o (IC50 = 0.40 ± 0.050 µM) was the most active inhibitor of butyrylcholinesterase enzyme. The binding interactions of most active compounds with the active site of enzymes were established with the help of molecular docking studies.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/molecules28010021/s1, Figure S1. Proton NMR spectrum of compound 6b. Figure S2. Proton NMR spectrum of compound 6d. Figure S3. Proton NMR spectrum of compound 6i. Figure S4. Proton NMR spectrum of compound 6j.

Author Contributions

Conceptualization, H.U. and F.R.; methodology, R.H.; software, S.K.; validation, M.A.A. (Mahmoud A. Abdelaziz); A.A.; N.Z. and F.S.A.; formal analysis, S.A.A.S. and M.T.; investigation, M.T.; resources, M.T. and F.R.; data curation, K.M.K. and N.I.; writing—original draft preparation, H.U. and F.R.; writing—review and editing, A.A.; M.I.S.; N.Z.; I.J.; R.I.; M.A.A. (Marzough Aziz Albalawi) and M.S.; visualization, M.S. and I.J.; supervision, F.R.; project administration, F.R. and H.U.; funding acquisition, M.A.A. (Mahmoud A. Abdelaziz); M.A.A. (Mahmoud A. Abdelaziz) and F.S.A. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Adams, R.L.; Craig, P.L.; Parsons, O.A. Neuropsychology of dementia. Neurol. Clin. 1986, 4, 387–404. [Google Scholar] [CrossRef] [PubMed]
  2. Aisen, P.S.; Davis, K.L. The search for disease-modifying treatment for Alzheimer’s disease. Neurology 1997, 48, 35–41. [Google Scholar] [CrossRef] [PubMed]
  3. Jann, M.W. Preclinical pharmacology of metrifonate. Pharmacotherapy 1998, 18, 55–67. [Google Scholar] [PubMed]
  4. 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]
  5. Mushtaq, G.; H-Greig, N.; A-Khan, J.; A-Kamal, M. Status of acetylcholinesterase and butyrylcholinesterase in Alzheimer’s disease and type 2 diabetes mellitus. CNS Neurol. Disord. Drug Targets 2014, 13, 1432–1439. [Google Scholar] [CrossRef] [PubMed]
  6. Ahmad, S.; Iftikhar, F.; Ullah, F.; Sadiq, A.; Rashid, U. Rational design and synthesis of dihydropyrimidine based dual binding site acetylcholinesterase inhibitors. Bioorg. Chem. 2016, 69, 91–101. [Google Scholar] [CrossRef]
  7. Auld, D.S.; Kornecook, T.J.; Bastianetto, S.; Quirion, R. Alzheimer’s disease and the basal forebrain cholinergic system: Relations to β-amyloid peptides, cognition, and treatment strategies. Prog. Neurobiol. 2002, 68, 209–245. [Google Scholar] [CrossRef]
  8. Ecobichon, D.J.; Comeau, A.M. Pseudocholinesterases of mammalian plasma: Physicochemical properties and organophosphate inhibition in eleven species. Toxicol. Appl. Pharmacol. 1973, 24, 92–100. [Google Scholar] [CrossRef]
  9. Rahim, F.; Ullah, H.; Taha, M.; Wadood, A.; Javed, M.T.; Rehman, W.; Nawaz, M.; Ashraf, M.; Ali, M.; Sajid, M.; et al. Synthesis and in vitro acetylcholinesterase and butyrylcholinesterase inhibitory potential of hydrazide-based Schiff bases. Bioorg. Chem. 2016, 68, 30–40. [Google Scholar] [CrossRef]
  10. Cavalli, A.; Bolognesi, M.L.; Minarini, A.; Rosini, M.; Tumiatti, V.; Recanatini, M.; Melchiorre, C. Multi-target-directed ligands to combat neurodegenerative diseases. J. Med. Chem. 2008, 51, 347–372. [Google Scholar] [CrossRef]
  11. Rockwood, K.; Mintzer, J.; Truyen, L.; Wessel, T.; Wilkinson, D. Effects of a flexible galantamine dose in Alzheimer’s disease: A randomised, controlled trial. J. Neurol. Neurosurg. Psychiatry 2001, 71, 589–595. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Kucukguzel, I.; Kucukquzel, S.G.; Rollas, S.; Otuk-Sanis, G.; Ozdemir, O.; Bayrak, I.; Altug, T.; Stables, J.P. Synthesis of some 3-(arylalkylthio)-4-alkyl/aryl-5-(4-aminophenyl)-4H-1,2,4-triazole derivatives and their anticonvulsant activity. Farmaco 2004, 59, 893–901. [Google Scholar] [CrossRef]
  13. Maddila, S.; Gorle, S.; Singh, M.; Lavanya, P.; Jonnalagadda, S.B. Synthesis and Anti-Inflammatory Activity of Fused 1,2,4-triazolo-[3,4-b] [1,3,4]thiadiazole Derivatives of Phenothiazine. Lett. Drug Des. Discov. 2013, 10, 977. [Google Scholar] [CrossRef]
  14. Almasirad, A.; Tabatabai, S.A.; Faizi, M.; Kebriaeezadeh, A.; Mehrabi, N.; Dalvandi, A.; Shafiee, A.; Ainsworth, C.; Easton, N.R.; Livezey, M.; et al. Synthesis and anticonvulsant activity of new 2-substituted-5-[2-(2-fluorophenoxy) phenyl]-1,3,4-oxadiazoles and 1,2,4-triazoles. Bioorg. Med. Chem. Lett. 2004, 14, 6057–6059. [Google Scholar] [CrossRef] [PubMed]
  15. Maddila, S.; Pagadala, R.; Sreekanth, R.; Jonnalagadda, B. 1,2,4-Triazoles: A Review of Synthetic Approaches and the Biological Activity. Lett. Org. Chem. 2013, 10, 693–714. [Google Scholar] [CrossRef]
  16. Oh, K.; Yamada, K.; Asami, T.; Yoshizawa, Y. Synthesis of novel brassinosteroid biosynthesis inhibitors based on the ketoconazole scaffold. Bioorg. Med. Chem. Lett. 2012, 22, 1625–1628. [Google Scholar] [CrossRef]
  17. Wang, B.; Chu, D.; Feng, Y.; Shen, Y.; Aoyagi-Scharber, M.; Post, L.E. Discovery and Characterization of (8 S, 9 R)-5-Fluoro-8-(4-fluorophenyl)-9-(1-methyl-1 H-1, 2, 4-triazol-5-yl)-2, 7, 8, 9-tetrahydro-3 H-pyrido[4, 3, 2-de]phthalazin-3-one (BMN 673, Talazoparib), a Novel, Highly Potent, and Orally Efficacious Poly (ADP-ribose) Polymerase-1/2 Inhibitor, as an Anticancer Agent. J. Med. Chem. 2016, 59, 335–357. [Google Scholar]
  18. Murphy, M.J.J. Molecular action and clinical relevance of aromatase inhibitors. Oncologist 1998, 3, 129–130. [Google Scholar] [CrossRef] [Green Version]
  19. Borden, K.L.; Culjkovic-Kraljacic, B. Ribavirin as an anti-cancer therapy: Acute myeloid leukemia and beyond. Leuk. Lymphoma. 2010, 51, 1805–1815. [Google Scholar] [CrossRef] [Green Version]
  20. Gomez-Junyent, J.; Benavent, E.; Sierra, Y.; el Haj, C.; Soldevila, L.; Torrejon, B.; Rigo-Bonnin, R.; Ariza, J.; Murillo, O. Efficacy of ceftolozane/tazobactam, alone and in combination with colistin, against multidrug-resistant Pseudomonas aeruginosa in an in vitro biofilm pharmacodynamic model. Int. J. Antimicrob. Agents 2019, 53, 612–619. [Google Scholar] [CrossRef]
  21. Yang, Y.; Rasmussen, B.A.; Shlaes, D.M. Class A β-lactamases—Enzyme-inhibitor interactions and resistance. Pharmaco. Thera. 1999, 83, 141–151. [Google Scholar] [CrossRef] [PubMed]
  22. Donnelley, M.A.; Zhu, E.S.; Thompson, G.R., 3rd. Isavuconazole in the treatment of invasive aspergillosis and mucormycosis infections. Infect. Drug Resist. 2016, 9, 79–86. [Google Scholar] [PubMed] [Green Version]
  23. Ledoux, M.P.; Denis, J.; Nivoix, Y.; Herbrecht, R. Isavuconazole: A new broad-spectrum azole. Part 2: Pharmacokinetics and clinical activity. J. Mycol. Med. 2018, 28, 15–22. [Google Scholar] [CrossRef] [PubMed]
  24. Taha, M.; Rahim, F.; Uddin, N.; Khan, I.U.; Iqbal, N.; Salahuddin, M.; Farooq, R.K.; Gollapalli, M.; Khan, K.M.; Zafar, A. Exploring indole-based-thiadiazole derivatives as potent acetylcholinesterase and butyrylcholinesterase enzyme inhibitors. Inter. J. Biol. Macromol. 2021, 188, 1025–1036. [Google Scholar] [CrossRef]
  25. Rahim, F.; Javid, M.T.; Ullah, H.; Wadood, A.; Taha, M.; Ashraf, M.; Aine, Q.U.; Khan, M.A.; Khan, F.; Mirza, S.; et al. Synthesis, Molecular Docking, Acetylcholinesterase and Butyrylcholinesterase Inhibitory Potential of Thiazole Analogs as New Inhibitors for Alzheimer Disease. Bioorg. Chem. 2015, 62, 106–116. [Google Scholar] [CrossRef]
  26. Taha, M.; Alshamrani, F.J.; Rahim, F.; Uddin, N.; Chigurupati, S.; Almandil, N.B.; Farooq, R.K.; Iqbal, N.; Aldubayan, M.; Venugopal, V.; et al. Synthesis, characterization, biological evaluation, and kinetic study of indole base sulfonamide derivatives as acetylcholinesterase inhibitors in search of potent anti-Alzheimer agent. J. King Saud Univ.-Sci. 2021, 33, 101401. [Google Scholar] [CrossRef]
  27. Taha, M.; Rahim, F.; Zaman, K.; Anouar, E.H.; Uddin, N.; Nawaz, F.; Sajid, M.; Khan, K.M.; Shah, A.A.; Wadood, A.; et al. Synthesis, in vitro biological screening and docking study of benzo [d] oxazole bis Schiff base derivatives as a potent anti-Alzheimer agent. J. Biomole. Struc. Dyn. 2021, 1–16. [Google Scholar] [CrossRef]
  28. Hussain, R.; Ullah, H.; Rahim, F.; Sarfraz, M.; Taha, M.; Iqbal, R.; Alhomrani, M.; Alamri, A.S.; Abdulaziz, O.; Abdelaziz, M.A. 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]
  29. Basha, N.M.; Venkatesh, B.C.; Reddy, G.M.; Zyryanov, G.V.; Subbaiah, M.V.; Wen, J.C.; Gollakota, A.R. Synthesis, Antimicrobial Assay and SARs of Pyrazole Included Heterocyclic Derivatives. Polycycl. Aromat. Compd. 2021, 1–15. [Google Scholar] [CrossRef]
  30. Shyamsivappan, S.; Vivek, R.; Saravanan, A.; Arasakumar, T.; Suresh, T.; Athimoolam, S.; Mohan, P.S. A novel 8-nitro quinoline-thiosemicarbazone analogues induces G1/S & G2/M phase cell cycle arrest and apoptosis through ROS mediated mitochondrial pathway. Bioorganic Chem. 2020, 97, 103709. [Google Scholar]
  31. Riaz, N.; Iftikhar, M.; Saleem, M.; Hussain, S.; Rehmat, F.; Afzal, Z.; Khawar, S.; Ashraf, M.; al-Rashida, M. New synthetic 1,2,4-triazole derivatives: Cholinesterase inhibition and molecular docking studies. Results Chem. 2020, 2, 100041. [Google Scholar] [CrossRef]
  32. Siddiqui, S.Z.; Arfan, M.; Abbasi, M.A.; Shah, S.A.A.; Ashraf, M.; Hussain, S.; Saleem, R.S.Z.; Rafique, R.; Khan, K.M. Discovery of Dual Inhibitors of Acetyl and Butrylcholinesterase and Antiproliferative Activity of 1,2,4-Triazole-3-thiol: Synthesis and In Silico Molecular Study. Chem. Select. 2020, 5, 6430–6439. [Google Scholar] [CrossRef]
  33. Wadood, A.; Shareef, A.; Rehman, A.U.; 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 2022, 7, 397–408. [Google Scholar] [CrossRef]
  34. 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]
  35. 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]
  36. 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]
Figure 1. Available drugs for Alzheimer’s disease.
Figure 1. Available drugs for Alzheimer’s disease.
Molecules 28 00021 g001
Figure 2. Bioactive drugs with triazole moiety.
Figure 2. Bioactive drugs with triazole moiety.
Molecules 28 00021 g002
Figure 3. Rationale of the current study.
Figure 3. Rationale of the current study.
Molecules 28 00021 g003
Scheme 1. Synthesis of triazole-based thiosemicarbazone derivatives (6a–u).
Scheme 1. Synthesis of triazole-based thiosemicarbazone derivatives (6a–u).
Molecules 28 00021 sch001
Figure 4. Summary of SAR studies of triazole-based thiosemicarbazone derivatives (6a–u).
Figure 4. Summary of SAR studies of triazole-based thiosemicarbazone derivatives (6a–u).
Molecules 28 00021 g004
Figure 5. Protein–ligand interaction (PLI) profile of analogue 6i against acetylcholinesterase (AChE): (A) 2D; (B) 3D.
Figure 5. Protein–ligand interaction (PLI) profile of analogue 6i against acetylcholinesterase (AChE): (A) 2D; (B) 3D.
Molecules 28 00021 g005
Figure 6. Protein–ligand interaction (PLI) profile of analogue 6i against butyrylcholinesterase (BuChE): (A) 2D; (B) 3D.
Figure 6. Protein–ligand interaction (PLI) profile of analogue 6i against butyrylcholinesterase (BuChE): (A) 2D; (B) 3D.
Molecules 28 00021 g006
Figure 7. Protein–ligand interaction (PLI) profile of analogue 6b against acetylcholinesterase (AChE): (A) 2D; (B) 3D.
Figure 7. Protein–ligand interaction (PLI) profile of analogue 6b against acetylcholinesterase (AChE): (A) 2D; (B) 3D.
Molecules 28 00021 g007
Figure 8. Protein–ligand interactions (PLI) profile of analogue 6b against butyrylcholinesterase (BuChE): (A) 2D; (B) 3D.
Figure 8. Protein–ligand interactions (PLI) profile of analogue 6b against butyrylcholinesterase (BuChE): (A) 2D; (B) 3D.
Molecules 28 00021 g008
Figure 9. Protein–ligand interaction (PLI) profile of analogue 6q against acetylcholinesterase (AChE): (A) 2D; (B) 3D.
Figure 9. Protein–ligand interaction (PLI) profile of analogue 6q against acetylcholinesterase (AChE): (A) 2D; (B) 3D.
Molecules 28 00021 g009
Figure 10. Protein–ligand interaction (PLI) profile of analogue 6q against butyrylcholinesterase (BuChE): (A) 2D; (B) 3D.
Figure 10. Protein–ligand interaction (PLI) profile of analogue 6q against butyrylcholinesterase (BuChE): (A) 2D; (B) 3D.
Molecules 28 00021 g010
Table 1. Substituents, in vitro acetylcholinesterase and butyrylcholinesterase inhibitory activities, and selectivity index of triazole-based thiosemicarbazone derivatives (6a–u).
Table 1. Substituents, in vitro acetylcholinesterase and butyrylcholinesterase inhibitory activities, and selectivity index of triazole-based thiosemicarbazone derivatives (6a–u).
DerivativeR1R2AChE
IC50 (µM) a
BuChE
IC50 (µM) a
Selectivity
Index b
6aMolecules 28 00021 i001Molecules 28 00021 i0022.20 ± 0.103.90 ± 0.101.77
6bMolecules 28 00021 i003Molecules 28 00021 i0040.20 ± 0.100.30 ± 0.101.5
6cMolecules 28 00021 i005Molecules 28 00021 i006N.A.N.A.N.A.
6dMolecules 28 00021 i007Molecules 28 00021 i008N.A.N.A.N.A.
6eMolecules 28 00021 i009Molecules 28 00021 i0104.70 ± 0.106.30 ± 0.101.34
6fMolecules 28 00021 i011Molecules 28 00021 i0122.40 ± 0.104.70 ± 0.101.96
6gMolecules 28 00021 i013Molecules 28 00021 i0142.10 ± 0.104.30 ± 0.102.04
6hMolecules 28 00021 i015Molecules 28 00021 i0169.10 ± 0.2011.20 ± 0.301.23
6iMolecules 28 00021 i017Molecules 28 00021 i0180.10 ± 0.0500.20 ± 0.0502.0
6jMolecules 28 00021 i019Molecules 28 00021 i02011.30 ± 0.3012.30 ± 0.301.09
6kMolecules 28 00021 i021Molecules 28 00021 i02212.20 ± 0.3014.10 ± 0.401.15
6lMolecules 28 00021 i023Molecules 28 00021 i0244.60 ± 0.0105.90 ± 0.101.28
6mMolecules 28 00021 i025Molecules 28 00021 i0262.70 ± 0.103.80 ± 0.101.41
6nMolecules 28 00021 i027Molecules 28 00021 i0280.70 ± 0.051.70 ± 0.0502.43
6oMolecules 28 00021 i029Molecules 28 00021 i0302.50 ± 0.303.10 ± 0.401.24
6pMolecules 28 00021 i031Molecules 28 00021 i0323.20 ± 0.104.60 ± 0.101.44
6qMolecules 28 00021 i033Molecules 28 00021 i0340.60 ± 0.0500.90 ± 0.101.50
6rMolecules 28 00021 i035Molecules 28 00021 i0361.30 ± 0.0502.20 ± 0.101.69
6sMolecules 28 00021 i037Molecules 28 00021 i0381.40 ± 0.0502.30 ± 0.101.64
6tMolecules 28 00021 i039Molecules 28 00021 i0401.90 ± 0.102.50 ± 0.101.32
6uMolecules 28 00021 i041Molecules 28 00021 i0422.90 ± 0.103.70 ± 0.101.28
Standard drug Donepezil2.16 ± 0.124.5 ± 0.112.08
Note: a Values expressed as means ± standard error of three experiments. b Selectivity index: IC50 of BuChE/IC50 of AChE; N.A.: not active.
Table 2. Binding energies, number of hydrogen bonding and the number of closest residues of the selected docked analogues into the active site of both AchE and BuChE.
Table 2. Binding energies, number of hydrogen bonding and the number of closest residues of the selected docked analogues into the active site of both AchE and BuChE.
Active DerivativesName of EnzymeFree Binding Energy (kcal/mol)Number of HBsNumber of Closest Residues
6iAChE−12.13524
BuChE−11.78222
6bAChE−11.37322
BuChE−10.49521
6qAChE−10.63623
BuChE−9.42324
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

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 2023, 28, 21. https://doi.org/10.3390/molecules28010021

AMA Style

Rahim F, Ullah H, Taha M, Hussain R, Sarfraz M, Iqbal R, Iqbal N, Khan S, Ali Shah SA, Albalawi MA, et al. Synthesis of New Triazole-Based Thiosemicarbazone Derivatives as Anti-Alzheimer’s Disease Candidates: Evidence-Based In Vitro Study. Molecules. 2023; 28(1):21. https://doi.org/10.3390/molecules28010021

Chicago/Turabian Style

Rahim, Fazal, Hayat Ullah, Muhammad Taha, Rafaqat Hussain, Maliha Sarfraz, Rashid Iqbal, Naveed Iqbal, Shoaib Khan, Syed Adnan Ali Shah, Marzough Aziz Albalawi, and et al. 2023. "Synthesis of New Triazole-Based Thiosemicarbazone Derivatives as Anti-Alzheimer’s Disease Candidates: Evidence-Based In Vitro Study" Molecules 28, no. 1: 21. https://doi.org/10.3390/molecules28010021

Article Metrics

Back to TopTop