Next Article in Journal
Oral Cell-Targeted Delivery Systems Constructed of Edible Materials: Advantages and Challenges
Next Article in Special Issue
Microwave-Assisted Synthesis of Aminophosphonic Derivatives and Their Antifungal Evaluation against Lomentospora prolificans
Previous Article in Journal
Tricyano-Methylene-Pyridine Based High-Performance Aggregation-Induced Emission Photosensitizer for Imaging and Photodynamic Therapy
Previous Article in Special Issue
Recent Advances in Age-Related Macular Degeneration Therapies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Ultrasound Assisted Synthesis and In Silico Modelling of 1,2,4-Triazole Coupled Acetamide Derivatives of 2-(4-Isobutyl phenyl)propanoic acid as Potential Anticancer Agents

by
Sadaf Mahmood
1,
Samreen Gul Khan
1,*,
Azhar Rasul
2,
Jørn Bolstad Christensen
3 and
Mohammed A. S. Abourehab
4,*
1
Drug Design and Medicinal Chemistry Laboratory, Department of Chemistry, Government College University, Faisalabad 38000, Pakistan
2
Department of Zoology, Faculty of Life Sciences, Government College University Faisalabad, Faisalabad 38000, Pakistan
3
Department of Chemistry, Faculty of Science, University of Copenhagen, Frederiksberg C, 1870 Copenhagen, Denmark
4
Department of Pharmaceutics College of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia
*
Authors to whom correspondence should be addressed.
Molecules 2022, 27(22), 7984; https://doi.org/10.3390/molecules27227984
Submission received: 19 September 2022 / Revised: 12 November 2022 / Accepted: 14 November 2022 / Published: 17 November 2022

Abstract

:
The development of an economical method for the synthesis of biologically active compounds was the major goal of this research. In the present study, we have reported the ultrasound-radiation-assisted synthesis of a series of novel N-substituted 1,2,4-triazole-2-thiol derivatives. The target compounds 6a–f were efficiently synthesized in significant yields (75–89%) by coupling 1,2,4-triazole of 2-(4-isobutylphenyl) propanoic acid 1 with different electrophiles using ultrasound radiation under different temperatures. The sonication process accelerated the rate of the reaction as well as yielded all derivatives compared to conventional methods. All derivatives were confirmed by spectroscopic (FTIR, 1HNMR, 13CNMR, HRMS) and physiochemical methods. All derivatives were further screened for their anticancer effects against the HepG2 cell line. Compound 6d containing two electron-donating methyl moieties demonstrated the most significant anti-proliferative activity with an IC50 value of 13.004 µg/mL, while compound 6e showed the lowest potency with an IC50 value of 28.399 µg/mL. The order of anticancer activity was found to be: 6d > 6b > 6f > 6a > 6c > 6e, respectively. The in silico modelling of all derivatives was performed against five different protein targets and the results were consistent with the biological activities. Ligand 6d showed the best binding affinity with the Protein Kinase B (Akt) pocket with the lowest ∆G value of −176.152 kcal/mol. Compound 6d has been identified as a promising candidate for treatment of liver cancer.

1. Introduction

Cancer and other infectious diseases are significant health concerns worldwide [1]. Cancer mortality rates will more than double in the coming years, according to the World Health Organization [2]. After receiving chemotherapy, cancer patients became vulnerable to microbial infections due to the emergence of multidrug resistance, a weak immune system and the development of resistant strains of pathogenic microorganisms [2,3,4]. Therefore, the development of multi-target drugs acting as both anticancer and antimicrobial agents is a major focus of researchers globally. There is a need for intensive efforts to discover and develop new anticancer drugs with high safety profiles [5].
Nitrogen-based heterocyclic compounds have played an important role in the field of pharmaceuticals [6]. Approximately 75% of FDA-approved drugs are nitrogen-based heterocyclic compounds [7]. These heterocycles are reported to have great importance in medicinal chemistry [8]. According to the pharmacodynamical principle of superposition, 1,2,4-triazole, a versatile scaffold, is reported to have potential pharmacological activities, such as anticancer [9], antifungal [10], anti-urease [11], anti-inflammatory [12], antioxidant [13] as well as anticonvulsant [14] properties. Acetamide is reported to be a significant and important pharmacophore of anti-cancer drugs [15]. Cefatrizine and Tazobactam are triazole-based antimicrobial and anticancer drugs, respectively (Figure 1).
The scientific community is facing major challenges in the development of environmentally sustainable and economical techniques for the synthesis of novel pharmacological agents. Sonochemistry is a technique for accelerating the rate of chemical reactions by using ultrasound radiation in the range of 20 KHz to 100 KHz in a sonicator [16,17,18]. This technique offers a fast, efficient and economic procedure for the synthesis of bioactive molecules in good yields within a short amount of time [19]. In contrast to conventional heating techniques or systems involving catalysts, ultrasound radiation accelerates the rate of various chemical reactions through the phenomenon of cavitation [20,21]. The bubbles produced during the cavitation phenomenon collapse with the sound waves, generating a tremendous amount of energy and transforming kinetic energy into heating energy [22].
Molecular hybridization is a simple and effective tool to covalently combine multiple drug pharmacophores. On this basis, we have designed a hybrid of triazole and acetamide pharmacophores together in one molecule to prevent the progression of cancer. We selected 2-(4-isobutylphenyl)propanoic acid core for our chemical derivatization. Our designed molecular framework contains hydrophobic aryl rings on each side of the triazole ring to increase its bioavailability and cell penetration.
Previously, we have synthesized different pharmacologically active oxadiazole-based polyvalent derivatives containing acetamide functionalities [23,24,25]. So, in continuation of our previous work on heterocyclic compounds, we have cyclized the –COOH group of 2-(4-isobutylphenyl)propanoic acid into a five-membered heterocycle, e.g., triazole nucleus.

2. Results

2.1. Chemistry

The present work is a synthetic approach for the ultrasound-assisted synthesis of new N-substituted 5-aryl-1,2,4-triazole-3-acetamide derivatives 6a–f. Ultrasound-assisted reactions are economical in terms of having good yields and shorter periods of time. The synthetic route of synthesized compounds 6a–6f is depicted in Scheme 1. These reactions are based upon the chemical modification of 2-(4-isobutylphenyl)propanoic acid by using ultrasound radiation as the energy source required for the completion of the reaction. Triazole was derivatized at SH by replacing its H-group with different electrophiles, resulting in the final compounds 6a–6f in good yields.
Compound 2 was obtained by refluxing the 2-(4-isobutylphenyl)propanoic (1) with absolute methanol for six hours using the Fischer esterification method [26]. The ester of 2-(4-isobutylphenyl)propanoic acid was gently refluxed with 80% hydrazine hydrate [27] in methanol to obtain 2-(4-isobutylphenyl)propane hydrazide (3). In the third step, compound 3 was cyclized into 5-(1-(4-isobutylphenyl) ethyl)-1,2,4-triazole-2-thiol (4) by gently refluxing compound 3 in methanol with methyl isothiocyanate in 10% NaOH. After this was completed, the reaction mixture was acidified by concentrated HCl, and product 4 was isolated by filtration.
Triazole derivatives 6a–6f were achieved by coupling compound 4 with different substituted aralkyl/alkyl/aryl 2-bromoacetamides using ultrasound radiations for 39–80 min at 45–55°C. The conventional approach of synthesizing triazole derivatives was also used, which resulted in all the derivatives in moderate yields over a longer time period (10–36 h). The ultrasound-assisted strategy was more efficient in terms of the maximum yield (65–80%) and reduced the time, as compared to conventional methods. The structures of triazole-based acetamides were confirmed by spectroscopic (FTIR, 1HNMR, 13CNMR) and physiochemical methods.
The structures of molecules were verified by different spectroscopic techniques (Figure S1–S28) and their purity was confirmed by the molecular ion peaks in the HRMS spectra (Figure S29–S34). In the IR spectrum, the triazole-coupled acetamide formation was confirmed by the appearance of absorption bands of NH stretching around (3282–3235 cm−1), aromatic C-H vibrations (3097–3032 cm−1), C = O in the acetamide peak (1700–1635 cm−1), N = N stretching (1677–1650 cm−1), C-N stretching of the acetamide (1310–1230 cm−1), and C-N stretching of the triazole (1085–1075 cm−1). In the 1HNMR spectrum of 6a–6f, the protons of the aromatic ring system were observed in between 7.75 and 6.93 ppm. The aliphatic protons of propanoic acid were the most shielded and were observed to be around 0.86–0.84 ppm. Key evidence of the formation of an acetamide moiety was the appearance of the CH2 peak at 4.04 ppm and the NH peak around 10.27–9.64 ppm. 13CNMR spectroscopy also supported the formation of acetamide by the emergence of peaks around 166.12–164.18 ppm that correspond to the C = O group of acetamide, and peaks at 158.15 ppm that correspond to the carbon nucleus of the triazole ring. The synthesis of triazole was further confirmed by the appearance of N-CH3 at 3.28 ppm in 1HNMR and at 29.58 ppm in 13CNMR. The characteristic signals at 22 ppm corresponded to two -CH3 carbon nuclei of propanoic acid. The segregation of signals between 44.18 and 20.19 ppm corresponded to CH2 carbon of propanoic acid. All these peaks confirmed the synthesis of triazole-based molecules.

2.2. Anti-Proliferative Potential

The anticancer potential of all N-substituted 5-aryl-1,2,4-triazole-3-acetamide derivatives 6a–6f was checked against the liver cancer HepG2 using an MTT assay [28]. All compounds showed moderate to excellent anticancer activity, which varied according to the substitution on the aryl rings, as shown in (Table 1). The MTT assay after 48 h showed the rate of the proliferation of HepG2 cells after exposure to different concentrations (3.25 µg/mL to 100 µg/mL) of the samples 6a–6f (Table 2). The positive control shows the 100% viability of the HepG2 cells without drug exposure. A comparison of all these samples revealed the most significant cytotoxicity was exhibited by 6d with a minimum IC50 value of 13.004 µg/mL (Table 1). While compound 6e showed the lowest potency and highest IC50 value of 28.399, compound 6e containing the electron-withdrawing bromo group was the least active. The order of activity was found to be: 2,3-Dimethyl phenyl ≥ 2,4-Dimethyl phenyl ≥ 3-Methyl phenyl ≥ 4-Methyl phenyl ≥ 2-Bromo phenyl ≥ 4-Bromo phenyl, respectively (Figure 2).
The MTT assay graph after 48 hours showed the rate of the proliferation of the HepG2 cells after exposure to different concentrations (3.25 µg/mL to 200 µg/mL). Compounds 6d and 6b showed the most significant cell viability at 12.5 µg/mL (Table 2 and Figure 2). The MTT assay graph after 48 hours showed the percentage of the age inhibition of the proliferation of the HepG2 cells after exposure to different concentrations (3.25 µg/mL to 200 µg/mL). Compounds 6d and 6b showed most significant percentage of age inhibition at 12.5 µg/mL.

2.3. Structure–Activity Relationship (SAR) Study

The anticancer activity of all compounds 6a–6f was checked against the liver cancer HepG2 cell lines by using an MTT assay. Amongst all the compounds, 6d was most potent, containing two electron-donating methyl substituents at the ortho and meta-position of the aryl ring. Meanwhile, compound 6e with an electron-withdrawing bromo group at the para position showed the lowest potency and highest IC50 value, 28.399.
The order of decreasing anti-proliferative activity was as follows: 6d > 6b > 6f > 6a > 6c > 6e, respectively. Based on the findings from the SAR of N-substituted 5-aryl-1,2,4-triazole-3-acetamide derivatives 6a–6f, it is concluded that the presence of the electron-donating -CH3 motif at the ortho- and meta-position of the aryl ring enhanced the anti-proliferative activity of the compound.

2.4. Molecular Docking

The in silico modelling of 6a–6f was carried out to examine the potential mode of action of the compounds under study that may be liable for their anti-cancer activities. Five important targets, Signal transducer and activator of transcription 3 (STAT3), Phosphatidylinositol 3-kinase alpha (PI3Kalpha), c-Kit Tyrosine Kinase (c-Kit), Protein kinase B (Akt) and Human Aurora B kinase, were selected as important proteins in cancer treatment. Signal transducer and activator of transcription 3 (STAT3) is a secret transcription factor that is activated in response to different cytokines, growth factors and oncogene signals. Therefore, it is considered as an attractive cancer therapeutic target. Phosphatidylinositol 3-kinases alpha are members of the intracellular lipid kinases that regulate cell metabolism, survival, growth and proliferation. They have been associated with multiple human cancers. Protein kinase B (Akt) plays a considerable role in signaling within cells, by promoting both cell proliferation and survival, and in the case of cancer, this path is irregular. Human Aurora kinase B has an important role in the cell cycle and is overexpressed in tumor cells. The potential effects of the molecules on the targets were investigated using the web page: http://www.swisstargetprediction.ch/ [29,30,31,32,33,34]. Protein kinase B (Akt) plays a considerable role in signaling within cells as well as promoting both cell proliferation and survival, and in the case of cancer, this path is irregular. The results suggested and supported the effectiveness of the molecules against the kinase targets.
The selected targets, their codes in the protein data bank, the grid box coordinates and the molecular docking scores are presented in Table 3. The docking results show that the molecules have the potential to affect different targets.
All compounds showed better activity against STAT3, P13 Kalpha and Human A. c-Kit Tyrosine Kinase and Protein Kinase B (Akt) were predicted to have a strong interaction potential against the cancer therapeutic targets with the best binding poses and a low binding energy. These results suggested and supported the effectiveness of the molecules against the kinase targets.
The docking results showed that all the molecules have the potential to affect the Protein Kinase B target. Protein Kinase B (Akt) is predicted to have a strong interaction potential against cancer therapeutic targets. The binding affinities, categories, types of interactions and interacting residues of the docked complexes were assessed, as presented in Table 3. The hydrogen bonding interactions and hydrophobic contacts of every ligand were evaluating inside the binding site of the receptor protein. The ligands conformations which demonstrated the maximal biological activities are explained in Table 4 and Figure 3 with their favorable interactions in the binding sites.
As demonstrated in Table 4, compounds 6d and 6b are bound to the catalytic site of Protein Kinase B with the best binding poses and lowest binding energy, −176.152 and −169.631, respectively. The molecule 6d bound to Protein Kinase B by forming H-bond interactions with Lys181. It interacted with the residues Leu 296 and His 196 through alkyl interactions with Ile188, Lys160, Gly161, Phe163, Gly164, Lys165, Thr162, Gly159, Glu236, Glu279, Arg(c)6, Asp293, and Asn280 residues involving the van der Waals interactions between compound 6d and Protein Kinase B (Figure 4).
The molecule 6b bound to Protein Kinase B by forming H-bond interactions with Ser 9 and Lys 277, and Sulfur-x interactions with Thr 7. It interacted with the residues Leu183, Lys181 and Val166 through alkyl interactions (Figure 5).
The docked conformations of ligands 6a–f and the reference ligand STI were examined to obtain a qualitative estimation and to identify the molecular foundation of the analyzed biological activities (Figure 6).
As demonstrated in Table 5, compounds 6d and 6b are bound to the catalytic site of Protein Kinase B with the best binding poses and lowest binding energy, −180.052 and −170.726, respectively. Compound 6d’s key interactions with Protein c-Kit Tyrosine Kinase involved the H-bond (Cys673), Pi-doner H-bond (Thr670), Pi-Sigma bond (Leu595, Leu644), Alkyl (Leu644, Val654, Cys809, Leu799, Lys623, Phe811, Val668, Val603, Ala621), and van der Walls (Glu 671, Ala621, Glu640, val643, leu647, Ile808, Leu783, Ile653, His790, Asp810, Thr670, Val603, Asp677, Gly676) interactions (Figure 7).
The molecule 6b bound to Protein c-Kit Tyrosine Kinase by forming H-bond interactions with Glu640 and Thr670. Other key interactions involved hydrophobic alkyl interactions with the residues Val603, Ala621, Val643, Val654, Leu644 and Val668 (Figure 8).

3. Materials and Methods

3.1. General

Melting points of all derivatives were checked using open capillary tube methods and an electrical melting point (Stuart SMP10 melting point apparatus). As a starting material, 2-(4-isobutylphenyl)propanoic acid was used. Ultrasonic irradiation synthesis was performed in an ultrasonic cleaner bath (Model 1510, 1.9 L, 115 v) at frequency of 47 kHz. Infrared spectra were obtained using potassium bromide discs on an FT-IR spectrophotometer (BRUKER) with a wavelength range of 4000–400 cm−1 and were expressed in wave numbers (cm−1) at GC University’s Hi Tech Lab. 1HNMR and 13CNMR spectra were carried out on Advance Bruker using 500 MHz spectrophotometer (1HNMR) and 75 MHz to 126 MHz (13CNMR) spectrophotometer at Department of Chemistry, University of Copenhagen, Denmark. The chemical shift was expressed in δ ppm. Reaction progress and completion was monitored by thin-layer chromatography.

3.2. General Procedure for the Synthesis of Synthesized N-substituted 5-aryl-1,2,4-triazole-3-acetamide derivatives 6a–6f

3.2.1. Synthesis of methyl 2-(4-isobutylphenyl)propanoate (2)

Compound 2 was synthesized by the method reported in [23]. Compound 2 was obtained as pale yellow oily liquid with the following properties: yield 90%; b.p. 263–265 °C; IR (KBr) cm−1: 1736.34, 1203.37, 1162.81; and 1HNMR (400 MHz, CDCl3) δ 7.15 (d, 2H, J = 8.0 Hz, H-3 and H-5), 7.05 (d, 2H, J = 8.0 Hz, H-2 and H-6), 3.67 (q, 1H, H-7), 3.60(s, 3H, OCH3), 2.40 (d, 2H, J = 8.0 Hz, H-9), 1.82 (m,1H, H-9), 1.44 (d, 3H, J = 8.0 Hz, H-8), 0.85 (d, 6H, J = 8.0 Hz, H-11 and H-12). 13C NMR (101 MHz, CDCl3) δ 175.22, 140.49, 137.63, 129.04, 127.31, 52.09, 44.99, 30.25, 22.21, 18.50. HRMS (ESI+): m/z calculated for [(C14H20O2)+H]+: 220.1463; found: 220.1460. Element analysis calculated: C, 76.33; H, 9.15; observed: C, 76.30; H, 9.16 %.

3.2.2. Synthesis of 2-(4-isobutylphenyl)propane hydrazide (3)

Compound 3 was synthesized by the method reported in [23]. 2-(4-isobutylphenyl) propane hydrazide was separated as a white crystalline solid. Yield: (88%); m.p. 77–78 °C; IR (KBr) cm−1: 3272.75, 2963.11, 1640.11, 1604.85, 1466.29, 1366.60, 906.66, 686.81. 1H NMR (400 MHz, CDCl3) δ 9.50 (s, 1H, NH), 7.13 (d, 2H, J = 8.0 Hz, H-3 and H-5), 7.06 (d, 2H, J = 8.0 Hz, H-2 and H-6), 3.48 (d, 2H, NH2), 3.46 (q, 1H, H-7), 2.40 (d, 2H, J = 8.0 Hz, H-9), 1.81 (m,1H, H-9), 1.48 (d, 3H, J = 8.0 Hz, H-8), 0.84 (d, 6H, J = 8.0 Hz, H-11 and H-12). 13C NMR (101 MHz, CDCl3) δ 175.24, 140.49, 137.59, 129.57, 127.30, 44.96, 30.25, 22.19, 18.23. HRMS (ESI+): m/z calculated for [(C13H20N2O)+H]+. 220.1576; found: 220.1574. Element analysis calculated: C, 70.87; H, 9.15; N, 12.72; observed: C, 70.85; H, 9.16; N, 12.72 %.

3.2.3. Synthesis of 5-(1-(4-isobutylphenyl) ethyl)-1,2,4-triazole-2-thiol (4)

In an oven-dried round bottom flask, 2-(4-isobutylphenyl) propane hydrazide (0.02 mol) was mixed with equimolar amount of methyl isothiocyanate in 10% KOH solution. The mixture was refluxed at 95 °C for 10–11 h. Reaction progress as well as completion was continuously monitored by TLC. After completion, chilled water was added in flask to obtain precipitates of product. Precipitates were filtered and washed with water. The product was also recrystallized using absolute ethanol. 5-(1-(4-isobutylphenyl)ethyl)-1,2,4- triazole-2-thiol was separated as off-white crystalline solid. 1HNMR (400 MHz, CDCl3) δ 11.77 (s, 1H, SH), 7.05 (d, 2H, J = 8.0 Hz, H-3 and H-5), 6.99 (d, 2H, J = 8.0 Hz, H-2 and H-6), 3.96 (q, 1H, H-7), 3.18 (s, 3H, N-CH3), 2.39 (d, 2H, J = 8.0 Hz, H-9), 1.80 (m,1H, H-9), 1.62 (d, 3H, J = 8.0 Hz, H-8), 0.83 (d, 6H, J = 8.0 Hz, H-11 and H-12). 13C NMR (101 MHz, CDCl3) δ 155.16, 141.33, 137.11, 129.94, 126.87, 45.03, 37.60, 30.67, 30.15, 22.22, 20.20. HRMS (ESI+): m/z calculated for [(C15H21N3S)+H]+: 275.1456; found: 275.1454. Element analysis calculated: C, 65.42; H, 7.69; N, 15.26; S, 11.64; observed: C, 65.41; H, 7.69; N, 15.27; S, 11.65%.

3.2.4. Synthesis of N-substituted aryl/alkyl 2-chloroacetamides 5a–5f

Compounds 5a–5f were prepared by the methods reported in [23]. Briefly, in an oven-dried round bottom flask, the N-substituted aryl/alkyl amines (12.0 moles) were dissolved in 10.0 mL of 5% Na2CO3 solution. Bromo acetyl bromide (12.0 mmoles) was introduced gradually into the above reaction mixture. The mixture containing round bottom flask was shaken gently till precipitation. Reaction progress as well as completion was continuously checked by TLC. After the completion, chilled water was added in flask. Precipitates of the product were filtered, dried and recrystallized using absolute ethanol.

3.2.5. Synthesis of N-substituted 5-(1-(4-isobutylphenyl)ethyl)-1,2,4-triazole-2-yl- 2-sulfanyl acetamide derivatives 6a–6f

Method A: Conventional method [24]. Different N-substituted 5-(1-(4-isobutylphenyl)ethyl)-1,2,4-triazole-2-yl-2-sulfanyl acetamide derivatives were synthesized in moderate yields by the stirring 4 (0.02 mol) with equimolar amount of different substituted aralkyl/alkyl/aryl 2-bromoacetamides 5a–f using dichloromethane (15 mL) and NaH (0.01 mol) at room temperature for 10–30 h. Reaction progress as well as completion was continuously checked by TLC. After the completion, chilled water was added in flask to obtain precipitates of product. Precipitates of the product were filtered, dried and recrystallized using absolute ethanol.
Method B: Ultrasound-assisted method. Different N-substituted 5-(1-(4-isobutylphenyl)ethyl)-1,2,4-triazole-2-yl-2-sulfanyl acetamide derivatives were synthesized in good yields by the dissolving 4 (0.02 mol) in DCM (5 mL). NaH (0.01 mol) was added in the reaction mixture and it was stirred for 20 min. Equimolar amount of different substituted aralkyl/alkyl/aryl 2-bromoacetamides 5a–5f was added and the reaction mixture was sonicated for 30–90 min at 45–55 °C (Scheme 1). Reaction progress as well as completion was continuously checked by TLC. After the completion, chilled water was added in flask to obtain precipitates of product. Precipitates of the product were filtered, dried and recrystallized using absolute ethanol.

N-(4-Methylphenyl)-2-((5-(1-(4-isobutylphenyl)ethyl)-4-methyl-4H-1,2,4-triazol-3-yl)thio)acetamide (6a)

White amorphous solid, Yield 83%, m.p. 91–93 °C, IR: ν (cm−1): 3268.55, 1700.05, 1518.08, 1477.27, 1316.76, 1079.0, 758.83. 1H NMR (400 MHz, CDCl3) δ 10.78 (s, 1H, N-H), 7.55 (d, 2H, J = 8.0 Hz, H-2″, H-6″), 7.38 (d, 2H, J = 12.0 Hz, H-3 and H-5), 7.07 (d, 2H, J = 8.0 Hz, H-3″, H-5″), 7.03 (d, 2H, J = 8.0 Hz, H-2 and H-6), 4.09–4.04 (q, 1H, H-7), 4.02 (s, 2H, S-CH2), 3.20 (s, 3H, N-CH3), 2.66 (s, 3H, CH3 -4″), 2.42( d, 2H, J = 8.0 Hz, CH2-9), 1.83–1.74 (m, 1H, H-10), 1.77 (d, 3H, J = 8.0 Hz, CH3-8), 0.86 (d, J = 8.0 Hz, 6H, CH3-11, CH3-12). 13C NMR (126 MHz, DMSO) δ 166.12, 157.77, 148.86, 139.63, 139.63, 139.34, 135.15, 133.48, 132.37, 129.02, 128.14, 126.78, 125.37, 43.89, 37.23, 35.26, 30.50, 30.00, 21.87, 21.05, 17.69. HRMS (ESI+): m/z calculated for [(C15H21N3S)+H]+: 423.2119; found: 423.2211. Element analysis calculated: C, 68.21; H, 7.16; N, 13.26; S, 7.59; observed: C, 68.20; H, 7.17; N, 13.28; S, 7.57%.

N-(2,4-Dimethylphenyl)-2-((5-(1-(4-isobutylphenyl)ethyl)-4-methyl-4H-1,2,4-triazol-3-yl)thio)acetamide (6b)

Off white amorphous solid, Yield 89%, m.p. 94–96 °C. IR: ν (cm−1): 3282.85, 1669.22, 1522.77, 1433.67, 1366.60, 1078.08, 685.50. 1H NMR (500 MHz, DMSO) δ 9.55 (s, 1H, N-H), 7.24–7.23 (d, 1H, J = 5.0 Hz, H-6″), 7.09–7.05 (m, 4H, H-3, H-5 and H-2, H-6), 7.00 (s, 1H, H-3″), 6.95–6.93 (d, 2H, J = 10.0 Hz, H-5″), 4.38–4.29 (q, 1H, H-7), 4.01 (s, 2H, S-CH2), 3.26 (s, 3H, N-CH3), 2.41( d, 2H, J = 5.0 Hz, CH2-9), 2.24 (s, 3H, CH3-4″) 2.09 (s, 3H, CH3-2″) 1.84–1.76 (m, 1H, H-10), 1.60 (d, J = 5.0 Hz, 3H, CH3-8), 0.85 (d, J = 10.0 Hz, 6H, CH3-11, CH3-12). 13C NMR (126 MHz, DMSO) δ 165.83, 157.76, 148.85, 139.93, 139.36, 133.79, 132.90, 131.01, 129.02, 126.54, 124.26, 43.89, 37.24, 35.55, 29.99, 29.69, 20.47, 21.58, 20.80, 20.17, 17.37. HRMS (ESI+): m/z calculated for [(C25H33N4OS)+H]+: 437.2375; found: 437.2367. Element analysis calculated: C, 68.77; H, 7.39; N, 12.83; S, 7.34; observed: C, 68.75; H, 7.40; N, 12.81; S, 7.35%.

N-(2-Bromophenyl)-2-((5-(1-(4-isobutylphenyl)ethyl)-4-methyl-4H-1,2,4-triazol-3-yl)thio)acetamide (6c)

White amorphous solid, Yield 77%, m.p. 113–115 °C IR: ν (cm−1): 3345.65, 1688.84, 1516.33, 1464.88, 1320.42, 1085.06, 766.11. 1H NMR (500 MHz, DMSO) δ 9.90 (s, 1H, N-H), 7.75 (d, 1H, J = 10.0 Hz, H-6″), 7.50 (d, 1H, J = 10.0 Hz, H-3″), 7.33–7.31(t, 1H, J = 5.0 Hz and 10.0 Hz, H-5″), 7.21–7.18 (t, 1H, J = 5.0 Hz and 10.0 Hz, H-4″), 7.10–7.06 (m, 4H, H-2, H-3, H-5, H-6) 4.33–4.29 (q, 1H, H-7), 4.12 (s, 2H, S-CH2), 3.28 (s, 3H, N-CH3), 2.40 (d, 2H, J = 5.0 Hz, CH2-9), 1.79–1.76 (m, 1H, H-10), 1.60 (d, J = 5.0 Hz, 3H, CH3-8), 0.84 (d, J = 10.0 Hz, 6H, H-11, H-12). 13C NMR (126 MHz, DMSO) δ 166.15, 158.30, 148.85, 139.93, 139.60, 134.29, 129.27, 127.63, 126.24, 125.36, 43.89, 36.6, 35.56, 29.41, 29.12, 21.85, 20.77. HRMS (ESI+): m/z calculated for [(C23H27BrN4OS)+H]+: 487.1167; found: 487.1157. Element analysis calculated: C, 56.67; H, 5.58; N, 11.47; S, 6.56; observed: C, 56.66; H, 5.56; N, 11.48; S, 6.57%.

N-(2,3-Dimethylphenyl)-2-((5-(1-(4-isobutylphenyl)ethyl)-4-methyl-4H-1,2,4-triazol-3-yl)thio)acetamide (6d)

Off white amorphous solid, Yield 75%, m.p. 91–93°C. IR: ν (cm−1): 3235.73 (NH), 1635.22, 1514.79, 1444.90, 1389.08, 1075.04, 689.09. 1H NMR (500 MHz, DMSO) δ 9.68 (s, 1H, N-H), 7.10–7.00 (m, 7H, H-2, H-3, H-5, H-6, H-4″, H-5″ and H-6″), 4.32–4.30 (q, 1H, H-7), 4.02 (s, 2H, S-CH2), 3.26 (s, 3H, N-CH3), 2.39( d, 2H, J = 5.0 Hz, CH2-9), 2.23 (s, 3H, CH3-3″) 2.00 (s, 3H, CH3-2″) 1.80–1.76 (m, 1H, H-10) 1.60 (d, J = 5.0 Hz, 3H, CH3-8), 0.84 (d, J= 10.0 Hz, 6H, H-11, H-12). 13C NMR (126 MHz, DMSO) δ 165.86, 158.01, 148.85, 139.63, 139.35, 130.70, 129.03, 126.77, 125.36, 123.15, 43.90, 36.96, 35.55, 30.20, 29.99, 22.13, 20.47, 20.19, 13.77. HRMS (ESI+): m/z calculated for [(C25H33N4OS)+H]+: 437.2375; found: 437.2367. Element analysis calculated: C, 68.77; H, 7.39; N, 12.83; S, 7.34; observed: C, 68.75; H, 7.40; N, 12.81; S, 7.35%.

N-(4-Bromophenyl)-2-((5-(1-(4-isobutylphenyl)ethyl)-4-methyl-4H-1,2,4-triazol-3-yl)thio)acetamide (6e)

Off white amorphous solid, Yield 85%, m.p. 85–87°C. IR: ν (cm−1): 3233.65, 1688.25, 1516.33, 1465.83, 1318.23, 1076.24, 692.33. 1H NMR (500 MHz, DMSO) δ 10.40 (s, 1H, N-H), 7.51(d, 2H, J = 10.0 Hz, H-2″ and H-6″), 7.49 (d, 2H, J = 5.0 Hz, H-3″ and H-5″), 7.05 (d, 2H, J = 10.0 Hz, H-2 and H-6), 7.03 (d, 2H, J = 10.0 Hz, H-3 and H-5), 4.31–4.27 (q, 1H, H-7), 3.99 (s, 2H, S-CH2), 3.25 (s, 3H, N-CH3), 2.39 (d, 2H, J = 5.0 Hz, CH2-9), 1.81–1.74 (m, 1H, H-10), 1.58 (d, 3H, J = 12.0 Hz, CH3-8), 0.85 (d, J = 5.0 Hz, 6H, CH3-11 and CH3-12). 13C NMR (126 MHz, DMSO) δ 164.47, 158.01, 148.86, 139.34, 138.75, 138.24, 131.51, 129.0, 126.55, 120.89, 120.38, 43.91, 35.24, 31.58, 29.97, 22.44, 21.07. HRMS (ESI+): m/z calculated for [(C23H27BrN4OS)+H]+: 487.1167; found: 487.1157. Element analysis calculated: C, 56.67; H, 5.58; N, 11.47; S, 6.56; observed: C, 56.66; H, 5.56; N, 11.48; S, 6.57 %.

N-(3-Methylphenyl)-2-((5-(1-(4-isobutylphenyl)ethyl)-4-methyl-4H-1,2,4-triazol-3-yl)thio)acetamide (6f)

White amorphous solid, Yield 87%, m.p. 92–95°C. IR: ν (cm−1): 3266.73, 1694.15, 1522.99, 1477.87, 1312.86, 1077.08, 745.89. 1H NMR (500 MHz, DMSO) δ 9.68 (s, 1H, N-H), 7.43 (d, J = 15.0 Hz, 1H- H-6″), 7.39 (s, 1H, H-2″), 7.34 (dd, J = 10.0 Hz, 1H, H-5″), 7.08–7.05 (m, 5H, H-3, H-2, H-5, H-6 and H-4″), 4.33–4.28 (q, 1H, H-7), 4.04 (s, 2H, S-CH2), 3.27 (s, 3H, N-CH3), 2.40 (d, 2H, J = 10.0 Hz, CH2-9), 2.15 (s, 3H, CH3-3″), 1.83–1.76 (m, 1H, H-10), 1.60 (d, J = 10.0 Hz, 3H, CH3-8), 0.84 (d, J = 10.0 Hz, 6H, H-11, H-12). 13C NMR (126 MHz, DMSO) δ 166.12, 157.77, 148.86, 139.63, 139.34, 135.15, 133.48, 132.37, 129.02, 128.14, 126.78, 125.37, 43.89, 37.23, 35.26, 30.50, 30.00, 21.87, 21.05, 17.69. HRMS (ESI+): m/z calculated for [(C15H21N3S)+H]+: 423.2119; found: 423.2211. Element analysis calculated: C, 68.21; H, 7.16; N, 13.26; S, 7.59; observed: C, 68.20; H, 7.17; N, 13.28; S, 7.57%.

3.3. Experimental Procedures for Biological Activities

3.3.1. Cell Culture and Treatment

Hepatic cancer cells were cultivated in Dulbecco’s modified eagle’s medium. Penicillin, 10% fetal bovine serum (FBS) and 100 μg/mL streptomycin were added to the above mixture at 37 °C. All media were maintained with 5% carbon dioxide-rich atmosphere.

3.3.2. Determination of Cell Viability

The MTT assay was used to assess the viability of human liver cancer cells (HepG2 cells). In a 96 well plate, cancerous cells were cultured. For 48 h, liver cancer cells were treated with varying concentrations of drugs. After 48 h, MTT reagent (5 mg/mL) was added, and cells were incubated for 4 h at 37 °C. The formazan crystals were then dissolved in 150 L of Dimethyl sulfoxide (DMSO), and absorbance was measured in a microplate reader at 570 nm.

3.4. Molecular Docking

Molecular docking study was performed against five targets to understand the possible interaction mechanism of the molecules’ anticancer effect against HepG2 cell line. In particular, the results against Protein kinase B (Akt) and c-Kit Tyrosine Kinase (c-Kit) were excellent. For the docking studies, crystal structures of Protein Kinase B (Akt) and c-Kit Tyrosine Kinase (c-Kit) were obtained from the RCSB Protein Data Bank website (https://www.rcsb.org) under the PDB ID 2X39 and IT46, respectively [35]. The structures of the newly synthesized compounds were drawn using Chem Draw 20.1.1 and then exported to Marvin Sketch to create and minimize their 3D SDF structures. Before the docking process, the structure of the target proteins was checked, and errors in amino acid structures were corrected and optimized using Molegro Virtual Docker software [36]. The co-crystalized ligands at the crystal structure of proteins were chosen as the center of the grid box. They were re-docked again for the validation of the in silico process. New compounds were docked 10 times to the active sites of the target protein with Molegro Virtual Docker. The poses with the lowest binding energy and good interactions with the targets were chosen to analyze the interactions’ details. Discovery Studio Visualizer Software 2021 was utilized to visualize the 2D representations of the intermolecular interactions between the target and new derivatives.

4. Conclusions

A series of triazole-based acetamide derivatives 6a–f were synthesized by utilizing ultrasound-assisted synthetic methodology and conventional protocols. All derivatives were obtained in low to moderate yields (60–75%) by using the conventional protocol in 16–26 h. By utilizing the ultrasound-assisted protocol, the compounds were obtained in good to excellent yields (75–89%) within a short period of time (40–80 min.) at 45–55 °C. After spectral characterization, all compounds were screened for their anti-proliferative potential against liver cancer cells and showed moderate to excellent anticancer activity, which varied according to the substitution on the aryl ring, as shown in Table 1. Among all derivatives, 6d with a methyl substituent at the ortho- and meta-position of the aryl ring showed the best cell viability with an IC50 value of 13.004 μM. The mechanism of the inhibition of the compounds was further studied by in silico modelling. The results of molecular docking studies were in accordance with biological findings. The docked conformations of Kinase proteins revealed that 6d has a superior docking score comparable with reference X39 and STI. We conclude that the ultrasound-assisted synthesis of triazole-based acetamides can be used to improve yields as well as the rate of reaction. Compound 6d could be utilized as a lead compound in cancer therapy in the near future.

Supplementary Materials

The following supporting information can be downloaded at: www.mdpi.com/article/10.3390/molecules27227984/s1, Figure S1: 1HNMR spectrum of compound 2, Figure S2: 1HNMR spectrum of compound 3, Figure S3: 1HNMR spectrum of compound 4, Figure S4: 1HNMR spectrum of compound 4 (aromatic region), Figure S5: 1HNMR spectrum of compound 4 (aliphatic region), Figure S6: 13C NMR spectrum of compound 2, Figure S7: 13C NMR spectrum of compound 3, Figure S8: 13C NMR spectrum of compound 4, Figure S9: 1HNMR spectrum of compound 6a, Figure S10: 1HNMR spectrum of compound 6a, (aromatic region), Figure S11: 1HNMR spectrum of compound 6a (aliphatic region), Figure S12: 13C NMR spectrum of compound 6a (Full spectrum), Figure S13: 1HNMR spectrum of compound 6b (Full spectrum), Figure S14: COSY- 1HNMR spectrum of compound 6b, Figure S15: 13C NMR spectrum of compound 6b (Full spectrum), Figure S16: 1HNMR spectrum of compound 6c (Full spectrum), Figure S17: 1HNMR spectrum of compound 6c (aromatic region), Figure S18: COSY- 1HNMR spectrum of compound 6c, Figure S19: 13C NMR spectrum of compound 6c (Full spectrum), Figure S20: 1HNMR spectrum of compound 6d (Full spectrum), Figure S21: COSY 1HNMR spectrum of compound 6d, Figure S22: 13C NMR spectrum of compound 6d (Full spectrum), Figure S23: 1HNMR spectrum of compound 6e (Full), Figure S24: COSY 1HNMR spectrum of compound 6e, Figure S25: 13C NMR spectrum of compound 6e (Full spectrum), Figure S26: 1HNMR spectrum of compound 6f (Full Spectrum), Figure S27: COSY 1HNMR spectrum of compound 6f, Figure S28 13C NMR spectrum of compound 6f, Figure S29: Mass spectrum of compound 6a, Figure S30: Mass spectrum of compound 6b, Figure S31: Mass spectrum of compound 6c, Figure S32: Mass spectrum of compound 6d, Figure S33: Mass spectrum of compound 6e, Figure S34: Mass spectrum of compound 6f.

Author Contributions

Conceptualization, and experimental design: S.G.K. and M.A.S.A.; Methodology, S.M., S.G.K. and M.A.S.A.; experimental work: S.M., A.R. and S.G.K.; Software: J.B.C.; writing—original draft preparation, S.G.K. and M.A.S.A.; writing—review and editing, S.G.K., J.B.C. and M.A.S.A.; data curation, A.R., J.B.C. and S.G.K.; visualization, S.G.K. and J.B.C.; supervision, S.G.K.; project administration, S.G.K. and M.A.S.A.; funding acquisition, M.A.S.A. All authors have read and agreed to the published version of the manuscript.

Funding

The authors would like to thank the Deanship of Scientific Research at Umm Al-Qura University for supporting this work through its grant (Grant Code: 23UQU4290565DSR112) to M.A.S.A.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are contained within the article.

Acknowledgments

The authors would like to thank the Deanship of Scientific Research at Umm Al-Qura University for supporting this work through its grant (Grant Code: 23UQU4290565DSR112) to M.A.S.A. We are really grateful to Şevki Adem from the Department of Chemistry, Faculty of Sciences, Çankırı Karatekin University, 18100 Çankırı, Turkey, for performing the in silico modelling studies of all the compounds.

Conflicts of Interest

The authors declare no conflict of interest.

Sample Availability

Samples of all compounds are available from the authors.

References

  1. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA A Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef] [Green Version]
  2. Park, S.K.; Cho, L.Y.; Yang, J.J.; Park, B.; Chang, S.H.; Lee, K.-S.; Kim, H.; Yoo, K.-Y.; Lee, C.-T.; The Scientific Committee, Korean Academy of Tuberculosis and Respiratory Diseases. Lung cancer risk and cigarette smoking, lung tuberculosis according to histologic type and gender in a population based case–Control study. Lung Cancer 2010, 68, 20–26. [Google Scholar] [CrossRef]
  3. Clayton, P.E.; Banerjee, I.; Murray, P.G.; Renehan, A.G. Growth hormone, the insulin-like growth factor axis, insulin and cancer risk. Nat. Rev. Endocrinol. 2011, 7, 11–24. [Google Scholar] [CrossRef]
  4. Porta, C.; Riboldi, E.; Sica, A. Mechanisms linking pathogens-associated inflammation and cancer. Cancer Lett. 2011, 305, 250–262. [Google Scholar] [CrossRef]
  5. Rosa, R.; Monteleone, F.; Zambrano, N.; Bianco, R. In vitro and in vivo models for analysis of resistance to anticancer molecular therapies. Curr. Med. Chem. 2014, 21, 1595–1606. [Google Scholar] [CrossRef]
  6. Antunes, M.M.; Amarante, T.R.; Valente, A.A.; Almeida Paz, F.A.; Gonçalves, I.S.; Pillinger, M. A Linear Trinuclear Oxidodiperoxido-molybdenum (VI) Complex with Single Triazole Bridges: Catalytic Activity in Epoxidation, Alcoholysis, and Acetalization Reactions. ChemCatChem 2018, 10, 2782–2791. [Google Scholar] [CrossRef]
  7. Gomha, S.M.; Abdel-aziz, H.M.; El-Reedy, A.A. Facile Synthesis of Pyrazolo [3, 4-c] pyrazoles Bearing Coumarine Ring as Anticancer Agents. J. Heterocycl. Chem. 2018, 55, 1960–1965. [Google Scholar] [CrossRef]
  8. Henary, M.; Kananda, C.; Rotolo, L.; Savino, B.; Owens, E.A.; Cravotto, G. Benefits and applications of microwave-assisted synthesis of nitrogen containing heterocycles in medicinal chemistry. RSC Adv. 2020, 10, 14170–14197. [Google Scholar] [CrossRef] [PubMed]
  9. Goss, P.E. Pre-clinical and clinical review of vorozole, a new third generation aromatase inhibitor. Breast Cancer Res. Treat. 1998, 49, S59–S65. [Google Scholar] [CrossRef] [PubMed]
  10. Schiller, D.S.; Fung, H.B. Posaconazole: An extended-spectrum triazole antifungal agent. Clin. Ther. 2007, 29, 1862–1886. [Google Scholar] [CrossRef] [PubMed]
  11. Bektaş, H.; Ceylan, Ş.; Demirbaş, N.; Alpay-Karaoğlu, Ş.; Sökmen, B.B. Antimicrobial and antiurease activities of newly synthesized morpholine derivatives containing an azole nucleus. Med. Chem. Res. 2013, 22, 3629–3639. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. El-Serwy, W.S.; Mohamed, N.A.; Abbas, E.M.; Abdel-Rahman, R.F. Synthesis and anti-inflammatory properties of novel 1, 2, 4-triazole derivatives. Res. Chem. Intermed. 2013, 39, 2543–2554. [Google Scholar] [CrossRef]
  13. Sancak, K.; Ünver, Y.; Ünlüer, D.; Düğdü, E.; Kör, G.; Celik, F.; Birinci, E. Synthesis, characterization, and antioxidant activities of new trisubstituted triazoles. Turk. J. Chem. 2012, 36, 457–466. [Google Scholar] [CrossRef]
  14. Plech, T.; Kaproń, B.; Łuszczki, J.J.; Wujec, M.; Paneth, A.; Siwek, A.; Kołaczkowski, M.; Żołnierek, M.; Nowak, G. Studies on the anticonvulsant activity and influence on GABA-ergic neurotransmission of 1, 2, 4-triazole-3-thione-based compounds. Molecules 2014, 19, 11279–11299. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Alanazi, M.M.; Elwan, A.; Alsaif, N.A.; Obaidullah, A.J.; Alkahtani, H.M.; Al-Mehizia, A.A.; Alsubaie, S.M.; Taghour, M.S.; Eissa, I.H. Discovery of new 3-methylquinoxalines as potential anti-cancer agents and apoptosis inducers targeting VEGFR-2: Design, synthesis, and in silico studies. J. Enzym. Inhib. Med. Chem. 2021, 36, 1732–1750. [Google Scholar] [CrossRef]
  16. Kaur, N. Synthesis of six-and seven-membered heterocycles under ultrasound irradiation. Synth. Commun. 2018, 48, 1235–1258. [Google Scholar] [CrossRef]
  17. Ammar, H.B.; Chtourou, M.; Frikha, M.H.; Trabelsi, M. Green condensation reaction of aromatic aldehydes with active methylene compounds catalyzed by anion-exchange resin under ultrasound irradiation. Ultrason. Sonochem. 2015, 22, 559–564. [Google Scholar] [CrossRef]
  18. Bai, G.-Y.; Lan, X.-W.; Chen, G.-F.; Liu, X.-F.; Li, T.-Y.; Shi, L.-J. A novel sodium iodide and ammonium molybdate co-catalytic system for the efficient synthesis of 2-benzimidazoles using hydrogen peroxide under ultrasound irradiation. Ultrason. Sonochem. 2014, 21, 520–526. [Google Scholar] [CrossRef]
  19. Suslick, K.S. Sonochemistry. Science 1990, 247, 1439–1445. [Google Scholar] [CrossRef]
  20. Shabir, G.; Shafique, I.; Saeed, A. Ultrasound assisted synthesis of 5–7 membered heterocyclic rings in organic molecules. J. Heterocycl. Chem. 2022, 59, 1669–1702. [Google Scholar] [CrossRef]
  21. Wu, X.; Joyce, E.M.; Mason, T.J. Evaluation of the mechanisms of the effect of ultrasound on Microcystis aeruginosa at different ultrasonic frequencies. Water Res. 2012, 46, 2851–2858. [Google Scholar] [CrossRef] [PubMed]
  22. Savun-Hekimoğlu, B.; Ince, N.H. Decomposition of PPCPs by ultrasound-assisted advanced Fenton reaction: A case study with salicylic acid. Ultrason. Sonochem. 2017, 39, 243–249. [Google Scholar] [CrossRef] [PubMed]
  23. Gul, S.; Abbasi, M.A.; Khan, K.M.; Nafeesa, K.; Siddiqa, A.; Akhtar, M.N.; Shahid, M.; Subhani, Z. Synthesis, antimicrobial evaluation and hemolytic activity of 2-[[5-alkyl/aralkyl substituted-1, 3, 4-oxadiazol-2-yl] thio]-N-[4-(4-morpholinyl) phenyl] acetamide derivatives. J. Saudi Chem. Soc. 2017, 21, S425–S433. [Google Scholar] [CrossRef]
  24. Khan, S.G.; Bokhari, T.H.; Anjum, F.; Akhter, N.; Rasool, S.; Shah, S.A.A.; Shahid, M.; Arshad, A. Synthesis, characterization, antibacterial, hemolytic and thrombolytic activity evaluation of 5-(3-chlorophenyl)-2-((N-(substituted)-2-acetamoyl) sulfanyl)-1, 3, 4-oxadiazole derivatives. Pak. J. Pharm. Sci. 2020, 33, 871–876. [Google Scholar]
  25. Aziz-ur-Rehman; Khan, S.G.; Naqvi, S.A.R.; Ahmad, M.; Akhtar, N.; Bokhari, T.H.; Irfan, M.; Usman, A.; Batool, S.; Rasool, S.; et al. Synthesis, spectral analysis and biological evaluation of 2-{[(morpholin-4-yl) ethyl] thio}-5-phenyl/aryl-1, 3, 4-oxadiazole derivatives. Pak. J. Pharm. Sci. 2021, 34, 441–446. [Google Scholar]
  26. Zappaterra, F.; Rodriguez, M.E.M.; Summa, D.; Semeraro, B.; Costa, S.; Tamburini, E. Biocatalytic approach for direct esterification of ibuprofen with sorbitol in biphasic media. Int. J. Mol. Sci. 2021, 22, 3066. [Google Scholar] [CrossRef]
  27. Iqbal, K.; Jamal, Q.; Iqbal, J.; Afreen, M.S.; Sandhu, M.Z.A.; Dar, E.; Farooq, U.; Mushtaq, M.F.; Arshad, N.; Iqbal, M.M. Synthesis of N-substituted acetamide derivatives of azinane-bearing 1, 3, 4-oxadiazole nucleus and screening for antibacterial activity. Trop. J. Pharm. Res. 2017, 16, 429–437. [Google Scholar] [CrossRef] [Green Version]
  28. Thongnest, S.; Chawengrum, P.; Keeratichamroen, S.; Lirdprapamongkol, K.; Eurtivong, C.; Boonsombat, J.; Kittakoop, P.; Svasti, J.; Ruchirawat, S. Vernodalidimer L, a sesquiterpene lactone dimer from Vernonia extensa and anti-tumor effects of vernodalin, vernolepin, and vernolide on HepG2 liver cancer cells. Bioorg. Chem. 2019, 92, 103197. [Google Scholar] [CrossRef]
  29. Johnston Paul, A.; Grandis Jennifer, R. STAT3 signaling: Anticancer strategies and challenges. Mol. Interv. 2011, 11, 18–26. [Google Scholar] [CrossRef] [Green Version]
  30. Dong, C.; Liu, S.; Cui, Y.; Guo, Q. 12-Lipoxygenase as a key pharmacological target in the pathogenesis of diabetic nephropathy. Eur. J. Pharmacol. 2020, 879, 173122. [Google Scholar] [CrossRef]
  31. Vardanyan, S.; Avagyan, A.; Aghekyan, A.; Sargsyan, A.; Harutyunyan, S.; Gasparyan, H. Synthesis and Some Transformations of 5-(1, 4-Benzodioxan-2-yl)-4-phenyl-4H-1, 2, 4-triazole-3-thiol. Russ. J. Org. Chem. 2020, 56, 436–439. [Google Scholar] [CrossRef]
  32. Washburn, K.; Neary, J. P2 purinergic receptors signal to STAT3 in astrocytes: Difference in STAT3 responses to P2Y and P2X receptor activation. Neuroscience 2006, 142, 411–423. [Google Scholar] [CrossRef] [PubMed]
  33. Xu, J.; Sylvester, R.; Tighe, A.P.; Chen, S.; Gudas, L.J. Transcriptional activation of the suppressor of cytokine signaling-3 (SOCS-3) gene via STAT3 is increased in F9 REX1 (ZFP-42) knockout teratocarcinoma stem cells relative to wild-type cells. J. Mol. Biol. 2008, 377, 28–46. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Khan, M.F.; Kader, F.B.; Arman, M.; Ahmed, S.; Lyzu, C.; Sakib, S.A.; Tanzil, S.M.; Zim, A.I.U.; Imran, M.A.S.; Venneri, T. Pharmacological insights and prediction of lead bioactive isolates of Dita bark through experimental and computer-aided mechanism. Biomed. Pharmacother. 2020, 131, 110774. [Google Scholar] [CrossRef] [PubMed]
  35. McHardy, T.; Caldwell, J.J.; Cheung, K.-M.; Hunter, L.J.; Taylor, K.; Rowlands, M.; Ruddle, R.; Henley, A.; de Haven Brandon, A.; Valenti, M. Discovery of 4-amino-1-(7 H-pyrrolo [2, 3-d] pyrimidin-4-yl) piperidine-4-carboxamides as selective, orally active inhibitors of protein kinase B (Akt). J. Med. Chem. 2010, 53, 2239–2249. [Google Scholar] [CrossRef]
  36. Thomsen, R.; Christensen, M.H. MolDock: A new technique for high-accuracy molecular docking. J. Med. Chem. 2006, 49, 3315–3321. [Google Scholar] [CrossRef] [PubMed]
Figure 1. 1,2,4-triazole core containing drugs.
Figure 1. 1,2,4-triazole core containing drugs.
Molecules 27 07984 g001
Scheme 1. Synthesis of 1,2,4-triazole-based acetamides 6a–f.
Scheme 1. Synthesis of 1,2,4-triazole-based acetamides 6a–f.
Molecules 27 07984 sch001
Figure 2. Graphical representation of cell viability of 6a–6f in Hep G2 cell line at different concentrations by using MTT assay.
Figure 2. Graphical representation of cell viability of 6a–6f in Hep G2 cell line at different concentrations by using MTT assay.
Molecules 27 07984 g002
Figure 3. Binding interactions of reference ligand (X39) with Protein Kinase binding pocket.
Figure 3. Binding interactions of reference ligand (X39) with Protein Kinase binding pocket.
Molecules 27 07984 g003
Figure 4. Binding pocket of docked ligand 6d in active site of Protein Kinase B.
Figure 4. Binding pocket of docked ligand 6d in active site of Protein Kinase B.
Molecules 27 07984 g004
Figure 5. Binding pocket of docked ligand 6b in active site of Protein Kinase B.
Figure 5. Binding pocket of docked ligand 6b in active site of Protein Kinase B.
Molecules 27 07984 g005
Figure 6. Binding interactions of reference ligand (STI) with Protein c-Kit Tyrosine Kinase binding pocket.
Figure 6. Binding interactions of reference ligand (STI) with Protein c-Kit Tyrosine Kinase binding pocket.
Molecules 27 07984 g006
Figure 7. Binding pocket of docked ligand 6d in active site of c-Kit Tyrosine Kinase.
Figure 7. Binding pocket of docked ligand 6d in active site of c-Kit Tyrosine Kinase.
Molecules 27 07984 g007
Figure 8. Binding pocket of docked ligand 6b in active site of c-Kit Tyrosine Kinase B.
Figure 8. Binding pocket of docked ligand 6b in active site of c-Kit Tyrosine Kinase B.
Molecules 27 07984 g008
Table 1. The anti-liver cancer activity of triazole-based acetamides 6a–f according to (Scheme 1).
Table 1. The anti-liver cancer activity of triazole-based acetamides 6a–f according to (Scheme 1).
CompoundAlkyl/Aryl IC50Value (µg/mL)Conventional
Method
Ultrasound Assisted Method
Product %age YieldTime for ReactionProduct %age YieldTime for Reaction
6a4-methylphenyl22.3856516 h.8345 min.
6b2,4-dimethylphenyl14.1337016 h.8939 min.
6c2-Bromophenyl24.4827126 h.7780 min.
6d2,3-dimethylphenyl13.0046015 h.7540 min.
6e4-Bromophenyl28.3997522 h.8575 min.
6f3-methylphenyl15.4517318 h.8750 min.
Sorafenib 05.971
Table 2. Cell viability of derivatives 6a–6f at different concentrations (µg/mL).
Table 2. Cell viability of derivatives 6a–6f at different concentrations (µg/mL).
Concentration
(µg/mL)
6a6b6c6d6e6f
0100 ± 2.33100 ± 0.47100 ± 5.20100 ± 1.61100 ± 0.85100 ± 0.75
3.12598.18 ± 2.3396.63 ± 0.4793.51 ± 5.2088.75 ± 6.0397.57 ± 0.8594.39 ± 2.65
6.2586.65 ± 2.9281.10 ± 2.7482.51 ± 4.7474.18 ± 0.7785.67 ± 1.6486.12 ± 1.01
12.561.35 ± 1.3051.63 ± 1.4465.11 ± 5.3054.02 ± 1.6166.47 ± 4.7159.07 ± 0.75
2549.53 ± 4.6534.77 ± 2.4149.53 ± 4.6536.37 ± 1.3952.67 ± 0.2638.98 ± 3.56
5026.50 ± 4.0511.58 ± 0.2225.62 ± 1.5110.40 ± 0.8829.81 ± 3.1811.36 ± 0.73
10016.37 ± 1.768.74 ± 0.089.97 ± 1.008.93 ± 0.1716.80 ± 3.949.71 ± 0.27
2005.45 ± 0.556.04 ± 0.006.00 + 0.706.77 + 0.028.97 + 0.148.70 + 0.15
DMSO (-ve Control)100 ± 0.0100 ± 0.0100 ± 0.0100 ± 0.0100 ± 0.0100 ± 0.0
Table 3. Docking of compounds 6a–6f to five targets with codes, grid box coordinates and molecular docking scores.
Table 3. Docking of compounds 6a–6f to five targets with codes, grid box coordinates and molecular docking scores.
TargetsPhosphatidylinositol 3-Kinase Alpha
(PI3Kalpha)
Protein Kinase B (Akt)
(PKB)
Human Aurora B Kinase
(AURKB)
c-Kit Tyrosine Kinase
(c-Kit)
Signal Transducer and Activator of Transcription 3 (STAT3)
PDB ID4FA62 × 394AF31T466NJS
Centre of dockingX:44X:43X:21X:28X:13
CoordinatesY:14Y:31Y:-22Y:26Y:56
Z:31Z:111Z:-10Z:39Z:0.32
References Ligand0TAX39VX6STIKQV
LigandsMol. Dock ScoreMol. Dock ScoreMol. Dock ScoreMol. Dock ScoreMol. Dock Score
6a−145.41−164.897−133.498−158.744−109.036
6b−138.651−169.631−143.585−170.426−115.252
6c−140.069−161.63−132.878−153.031−107.5
6d−137.122−176.152−145.486−180.052−117.807
6e−132.298−159.691−131.958−151.185−106.026
6f−133.085−167.609−141.576−161.024−111.411
Reference
Molecules
−112.819−130.624−144.231−181.533 −197.521
Table 4. Docking of compounds 6a–6f to Protein Kinase B (Akt) binding pocket (PDB ID: 2X39).
Table 4. Docking of compounds 6a–6f to Protein Kinase B (Akt) binding pocket (PDB ID: 2X39).
Ligand(∆G)
(kcal/mol)
CategoryTypesInteractions Residues
6a−164.897H BondConventional H BondARG6, SER9
OtherSulfur-XTHR7
HydrophobicAlkylVAL166, LYS181, LYS181, LEU183
6b−169.631H BondConventional H BondSER9, LYS277
OtherSulfur-X
Pi- Cation
THR7
ARG6, GLU236
HydrophobicAlkylVAL166, LYS181, LEU183,
6c−167.609HydrophobicAlkylLYS181, LEU296
6d−176.152H BondConventional H Bond
Doner H Bond
LYS181
SERC9
OtherPi-AnionASP275
HydrophobicAlkylHIS196, LEU296
6e−161.63H BondConventional H BondTHR162, PHE163
HydrophobicAlkylVAL166
6f−159.691H Bond
Other
Conventional H BondLYS160, LYS181
C-H BondASP293
HydrophobicAlkylVAL166, ALA179, LEU158, VAL166, MET282
Reference Ligand H BondConventional H BondALA232, ASP293, MET282, GLU279, GLU236,
X39−130.624OtherC-H BondGLY159, MET229
HydrophobicAlkylVAL166, ALA179, LEU158, LEU183, LYS181
Table 5. Docking of compounds 6a–6f to Protein c-Kit Tyrosine Kinase binding pocket (PDB ID: IT46).
Table 5. Docking of compounds 6a–6f to Protein c-Kit Tyrosine Kinase binding pocket (PDB ID: IT46).
LigandACE (kcal/mol)CategoryTypesInteractions Residues
6a−158.744H BondConventional H BondGLU640
HydrophobicAlkylALA621, LYS623, VAL643, VAL603, LYS623, VAL603
6b−170.426H Bond
Other
Conventional H Bond
Pi- doner H Bond
Pi-Cation
Pi-Pi T shaped
GLU640
THR670
LYS273, ASP810
HIS790
HydrophobicAlkylVAL603, ALA621, VAL643, VAL654, LEU644, VAL668
6c−153.031H BondConventional H BondCYS673
HydrophobicPi-AlkylTYR672, PHE811
HydrophobicAlkylALA621, LYS623, VAL643 VAL668, EU783, CYS788 VAL603, LYS623, LEU783
6d−180.052H Bond
Other
Conventional H Bond
Pi-doner H-Bond
Pi-Sigma
Pi-Pi staked
CYS673
THR670
LEU595, LEU644
TYR672
HydrophobicAlkyl LEU644, VAL654, CYS809, LEU799, LYS623, PHE811, VAL668, VAL603, ALA621
6e−151.185H BondConventional H BondASP810
OtherPi-SulfurPHE811
HydrophobicAlkylLEU595, LEU644, VAL654 EU799, CYS809, LEU647 LEU783, LEU644, VAL654
6f−161.024H BondConventional H BondILE808
HydrophobicAlkylHIS790
Reference Ligand H BondConventional H BondALA232, ASP293, MET282, GLU279, GLU236,
STI−181.533OtherCarbon H BondGLY159, MET229
HydrophobicAlkylVAL166, ALA179, LEU A158, LEU A183, LYS A181
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Mahmood, S.; Khan, S.G.; Rasul, A.; Christensen, J.B.; Abourehab, M.A.S. Ultrasound Assisted Synthesis and In Silico Modelling of 1,2,4-Triazole Coupled Acetamide Derivatives of 2-(4-Isobutyl phenyl)propanoic acid as Potential Anticancer Agents. Molecules 2022, 27, 7984. https://doi.org/10.3390/molecules27227984

AMA Style

Mahmood S, Khan SG, Rasul A, Christensen JB, Abourehab MAS. Ultrasound Assisted Synthesis and In Silico Modelling of 1,2,4-Triazole Coupled Acetamide Derivatives of 2-(4-Isobutyl phenyl)propanoic acid as Potential Anticancer Agents. Molecules. 2022; 27(22):7984. https://doi.org/10.3390/molecules27227984

Chicago/Turabian Style

Mahmood, Sadaf, Samreen Gul Khan, Azhar Rasul, Jørn Bolstad Christensen, and Mohammed A. S. Abourehab. 2022. "Ultrasound Assisted Synthesis and In Silico Modelling of 1,2,4-Triazole Coupled Acetamide Derivatives of 2-(4-Isobutyl phenyl)propanoic acid as Potential Anticancer Agents" Molecules 27, no. 22: 7984. https://doi.org/10.3390/molecules27227984

Article Metrics

Back to TopTop