Skip to main content

Synthesis, α-Glucosidase inhibitory activity and docking studies of Novel Ethyl 1,2,3-triazol-4-ylmethylthio-5,6-diphenylpyridazine-4-carboxylate derivatives

Abstract

In this work, a novel series of pyridazine-triazole hybrid molecules were prepared and evaluated as inhibitors of rat intestinal α-glucosidase enzyme. Amongst all newly synthesized compounds, 10k showed good inhibition in the series with IC50 value of 1.7 µM which is 100 folds stronger than positive control, acarbose. The cytotoxicity revealed that this compound is not toxic against normal cell line, HDF. The docking studies showed that triazole ring plays an important role in the binding interactions with the active site. The insertion of compound 10k into the active pocket of α-glucosidase and formation of hydrogen bonds with Leu677 was observed from docking studies. The kinetic studies revealed that this compound has uncompetitive mode of inhibition against α-glucosidase enzyme.

Peer Review reports

Introduction

Diabetes Mellitus (DM) is considered as a progressive hormonic and metabolic disorder of endocrine system which is characterized by the body’s loss of control over blood sugar (glucose) [1]. According to the WHO report, 422 million people worldwide [2] are suffering from diabetes which is associated with increased risks of quite a few micro- and macro-vascular complications such as stroke, retinopathy, neuropathy, nephropathy, coronary artery disease, hypertension and peripheral vascular disease [3,4,5,6]. This disease is divided into type 1 diabetes mellitus [T1DM] (resulted from insulin deficiency) and type 2 diabetes mellitus [T2DM] (resulted from resistance towards insulin). Type 2 is the most common type and accounts for 90% of all diabetic patients. Most of the medical approaches are focused on the reduction of the postprandial glucose (PG) level in blood. Current treatment approaches include oral anti-diabetic drugs such as sulfonylureas, thiazolidinediones, metformin, α-glucosidase inhibitors and glycosurics. α-Glucosidase (EC3.2.1.20) belongs to a glycoside hydrolase enzyme releasing monosaccharides through the lysis of α-glucopyranoside bond in saccharide polymer, oligosaccharides, and disaccharides from the non-reducing portion of the oligomeric substrate [7]. Inhibiting the digestion and absorption of carbohydrates by using α-glucosidase inhibitors slows down carbohydrate digestion, stabilizes blood glucose level and consequently prevents hyperglycemia in diabetic patients. Three glucosidase inhibitors have been introduced to the market and all are carbohydrate mimics. Acarbose, voglibose, and miglitol have been used in the treatment of Type 2 diabetes (T2M) by inhibiting the activity of α-glucosidase and consequently the formation of glucose in the small intestine [8,9,10,11,12].

Heterocyclic rings along with sugar-mimic compounds have been emerged as privileged cores in inhibiting glucosidase [13]. Amongst diverse array of heterocyclic cores, nitrogen-containing rings have found a unique place as α-glucosidase inhibitors [14,15,16,17] and medicinal chemists are still exploring for new molecules containing these valuable pharmacophores. Since decades, pyridazines and its related compounds have attracted great attention because of their therapeutic importance. The literature survey revealed that this heterocyclic core is present in quite a few number of compounds with different pharmacological properties [18,19,20,21,22,23,24,25,26].

Click reaction, articulated by Sharpless [27, 28], has been considered as a reliable and practical strategy to improve inefficiencies and slownesses of conventional drug discovery. This method provides a facile and revolutionary approach to the invention of drug-like molecules and completion of combinatorial libraries without the need to professional skill and equipment. In addition, this reaction has provided a powerful way to assemble molecules with well-defined biological functions and proteomic applications through making carbon-heteroatom-carbon bond.

The copper-mediated azide–alkyne cycloaddition is a standard and ideal method to achieve triazole ring as a linker and functional moiety. This useful pharmacophore is one of the most important five-membered heterocyclic rings which could be easily synthesized with no sensitivity to water and oxygen and the need for purification techniques. This valuable scaffold also found in various drugs namely antifungal drugs and bioactive compounds with antibacterial, antiviral, and anti-HIV properties [29,30,31].

In recent years, we identified various heterocyclic cores as potential α-glucosidase inhibitors [32,33,34,35]. Keeping in mind that still there is an urgent need to develop lead candidates, we decided to synthesize novel pyridazine-containing compounds considering the bioisosteric relationship between pyridazine and triazine and the number of triazine-containing compounds with α-glucosidase inhibitory activity (Fig. 1) [36,37,38,39,40,41]. Triazoles were also reported in the literature as inhibitors of α-glucosidase enzyme [42]. In this regard, we combined both these cores in one molecule and evaluated their inhibitory activities against α-glucosidase enzyme along with kinetic, docking and cytotoxic studies.

Fig. 1
figure 1

Chemical structures of triazine-containing molecules with α-glucosidase inhibitory activity

Experimental section

Chemistry

Synthesis of hydrazono-1,2-diphenylethanone (2)

A solution of benzil and hydrazine hydrate in methanol was heated at reflux temperature for 15 min. Then, the reaction mixture was cooled to room temperature and the white solid was collected by filtration, washed with cold methanol and dried [43].

Synthesis of ethyl 3-oxo-5,6-diphenyl-2,3-dihydropyridazine-4-carboxylate (3)

A suspension of sodium (0.05 mol) in 200 mL ethanol was chilled to 0 oC and after 15 min, diethyl malonate (0.075 mol) and compound 2 (0.05 mol) were added to the mixture. The mixture was refluxed for 3 h. Upon solvent removal, the residue was acidified with HCl (1 N). The resultant solid was collected and washed with water.

Synthesis of ethyl 3-mercapto-5,6-diphenylpyridazine-4-carboxylate (4)

Lawesson’s reagent (10 mmol) was added to the mixture of compound 3 (20 mmol) in toluene (150 mL). The mixture was stirred at reflux temperature for 18 h. After cooling, the solid was separated and washed with toluene, dried and recrystallized from petroleum ether/ethyl acetate.

Synthesis of ethyl 5,6-diphenyl-3-(prop-2-yn-1-ylthio)pyridazine-4-carboxylate (5)

Propargyl bromide (12 mmol) was added to the mixture of compound 4 (10 mmol) and K2CO3 (10 mmol) in DMF (10 mL), and the mixture was stirred at 50 oC. Upon completion, the reaction was stopped with ice/water mixture and the solid was filtered and washed with water.

Synthesis of 2-chloro-N-arylacetamide (8)

Chloroacetyl chloride (10 mmol) was added to the ice-cooled solution of aromatic amines (10 mmol) and triethylamine (12 mmol) in 1,2-dichloroethane (20 mL). After 6 h stirring at room temperature, petroleum ether was added to the mixture and the solid was filtered and recrystallized from ethanol.

Synthesis of 2-azido-N-phenylacetamide (9)

A solution of compound 8 (10 mmol) and sodium azide (15 mmol) in dimethyl sulfoxide (20 mL) was stirred at room temperature. Upon completion, indicated by TLC, The reaction mixture was poured into ice/water and the solid was filtered and recrystallized from petroleum ether/ethyl acetate.

Synthesis of target compounds (10a-r)

To the suspension of compound 9 (1 mmol) and compound 5 (1 mmol) in DMF (10 mL), CuI (10 mol %) and triethylamine (1 mmol) was added. The mixture was stirred at room temperature until TLC indicated the disappearance of starting materials. The reaction mixture was poured into ice/water and the solid was collected and washed with water. The solid was recrystallized from ethanol.

Ethyl 3-(((1-(2-oxo-2-(phenylamino)ethyl)-1H-1,2,3-triazol-4-yl)methyl)thio)-5,6-diphenylpyridazine-4-carboxylate (10a)

Yield (65%, 0.36 g); Dark Yellow solid; M.p. = 147–149 °C; IR (KBr, cm-1): 3611, 1730 (C = O), 1652, 1435, 1341, 1160; 1 H NMR (DMSO-d6, 500 MHz): δ 0.86 (t, J = 7.1 Hz, 3 H), 4.03 (q, J = 7.2 Hz, 2 H), 4.79 (s, 2 H), 5.32 (s, 2 H), 7.07 (t, J = 8.0 Hz, 1 H), 7.13 (d, J = 8.1 Hz, 2 H), 7.25–7.32 (m, 10 H), 7.56 (d, J = 8.3 Hz, 2 H), 8.13 (s, 1 H), 10.45 (s, 1 H); 13 C NMR (DMSO-d6, 100 MHz): δ 13.4, 24.5, 52.9, 62.0, 116.5, 119.2, 124.7, 127.3, 128.3, 128.6 128.8, 129.2, 129.5, 131.2, 134.0, 135.0, 136.2, 138.3, 138.5, 142.0, 155.1, 157.5, 164.1, 166.3; Anal. Calcd. for C30H26N6O3S: C, 65.44; H, 4.76; N, 15.26. Found: C, 65.20; H, 4.57; N, 14.98; ESI-MS m/z: 550.2 [M]+.

Ethyl 3-(((1-(2-oxo-2-(m-tolylamino)ethyl)- H-1,2,3-triazol-4-yl)methyl)thio)-5,6-diphenylpyridazine-4-carboxylate (10b)

Yield (71%, 0.40 g); Dark Yellow solid; m.p. = 171–173 °C; IR (KBr, cm-1): 3623, 1717 (C = O), 1638, 1422, 1319, 1195; 1 H NMR (DMSO-d6, 300 MHz): δ 0.87 (t, J = 6.9 Hz, 3 H), 2.25 (s, 3 H), 4.03 (q, J = 7.2 Hz, 2 H), 4.79 (s, 2 H), 5.30 (s, 2 H), 7.19–7.30 (m, 14 H), 8.12 (s, 1 H), 10.37 (s, 1 H); 13 C NMR (DMSO-d6, 75 MHz): δ 13.3, 21.1, 24.7, 52.2, 62.1, 116.4, 119.7, 124.4, 127.8, 128.26, 128.33, 128.6, 128.8, 128.9, 129.0, 129.6, 130.4, 133.6, 135.2, 136.0, 138.1, 138.3, 142.4, 155.4, 157.3, 164.0, 166.4; Anal. Calcd. for C31H28N6O3S: C, 65.94; H, 5.00; N, 14.88. Found: C, 65.60; H, 4.72; N, 14.59; ESI-MS m/z: 564.2 [M]+.

Ethyl 3-(((1-(2-oxo-2-(p-tolylamino)ethyl)-1H-1,2,3-triazol-4-yl)methyl)thio)-5,6-diphenylpyridazine-4-carboxylate (10c)

Yield (70%, 0.39 g); Dark Yellow solid; m.p. = 207–209 °C; IR (KBr, cm-1): 3617, 1720 (C = O), 1642, 1442, 1330, 1177; 1 H NMR (DMSO-d6, 500 MHz): δ 0.89 (t, J = 7.1 Hz, 3 H), 2.26 (s, 3 H), 4.05 (q, J = 7.0 Hz, 2 H), 4.81 (s, 2 H), 5.31 (s, 2 H), 7.12–7.16 (m, 4 H), 7.27–7.36 (m, 8 H), 7.45–7.48 (m, 2 H), 8.14 (s, 1 H), 10.37 (s, 1 H); 13 C NMR (DMSO-d6, 125 MHz): 13.2, 21.0, 24.6, 52.3, 62.0, 116.5, 119.8, 125.0, 126.9, 127.3, 127.7, 128.5, 128.9, 129.1, 129.7, 130.2, 132.9, 135.0, 136.1, 138.4, 142.2, 155.0, 157.1, 164.1, 166.2; Anal. Calcd. for C31H28N6O3S: C, 65.94; H, 5.00; N, 14.88. Found: C, 66.20; H, 4.73; N, 15.11; ESI-MS m/z: 565.1 [M + H]+.

Ethyl 3-(((1-(2-((4-isopropylphenyl)amino)-2-oxoethyl)-1H-1,2,3-triazol-4-yl)methyl)thio)-5,6-diphenylpyridazine-4-carboxylate (10d)

Yield (68%, 0.40 g); Dark Yellow solid; m.p. = 183–185 °C; IR (KBr, cm-1): 3611, 1719 (C = O), 1652, 1441, 1325, 1178; 1 H NMR (DMSO-d6, 500 MHz): δ 0.83–0.90 (m, 9 H), 2.56–2.58 (m, 1 H), 4.03 (q, J = 7.0 Hz, 2 H), 4.81 (s, 2 H), 5.33 (s, 2 H), 7.13–7.18 (m, 2 H), 7.29–7.37 (m, 12 H), 8.16 (s, 1 H), 10.53 (s, 1 H); 13 C NMR (DMSO-d6, 125 MHz): δ 13.8, 24.3, 25.2, 33.6, 52.6, 62.6, 115.9, 116.0, 121.5, 126.0, 127.1, 128.3, 128.8, 129.1, 129.3, 129.5, 130.0, 130.1, 130.9, 134.1, 135.2, 136.5, 156.0, 157.9, 164.5, 164.6; Anal. Calcd. for C33H32N6O3S: C, 66.87; H, 5.44; N, 14.18. Found: C, 66.57; H, 5.12; N, 13.85; ESI-MS m/z: 592.2 [M]+.

Ethyl 3-(((1-(2-((3-methoxyphenyl)amino)-2-oxoethyl)-1H-1,2,3-triazol-4-yl)methyl)thio)-5,6-diphenylpyridazine-4-carboxylate (10e)

Yield (72%, 0.41 g); Dark Yellow solid; m.p. = 155–157 °C; IR (KBr, cm-1): 3622, 1718 (C = O), 1639, 1451, 1333, 1175; 1 H NMR (DMSO-d6, 300 MHz): δ 0.87 (t, J = 7.1 Hz, 3 H), 3.70 (s, 3 H), 4.06 (q, J = 7.0 Hz, 2 H), 4.79 (s, 2 H), 5.31 (s, 2 H), 6.64 (d, J = 6.8 Hz, 1 H), 7.07–7.32 (m, 13 H), 8.13 (s, 1 H), 10.44 (s, 1 H); 13 C NMR (DMSO-d6, 75 MHz): δ 13.3, 24.7, 52.2, 55.0, 62.1, 104.9, 109.2, 111.4, 127.8, 128.3, 128.34, 128.58, 128.80, 128.86, 128.97, 129.6, 130.4, 133.6, 135.2, 136.0, 139.5, 142.4, 155.4, 157.3, 159.5, 164.0, 164.6; Anal. Calcd. for C31H28N6O4S: C, 64.12; H, 4.86; N, 14.47. Found: C, 63.88; H, 4.57; N, 14.72; ESI-MS m/z: 581.2 [M + H]+.

Ethyl 3-(((1-(2-((4-methoxyphenyl)amino)-2-oxoethyl)-1H-1,2,3-triazol-4-yl)methyl)thio)-5,6-diphenylpyridazine-4-carboxylate (10f)

Yield (66%, 0.38 g); Dark Yellow solid; m.p. = 230–232 °C; IR (KBr, cm-1): 3601, 1724 (C = O), 1644, 1439, 1327, 1180; 1 H NMR (DMSO-d6, 300 MHz): δ 0.87 (t, J = 7.1 Hz, 3 H), 3.70 (s, 3 H), 4.03 (q, J = 7.0 Hz, 2 H), 4.79 (s, 2 H), 5.29 (s, 2 H), 6.88 (d, J = 8.1 Hz, 2 H), 7.07–7.32 (m, 10 H), 7.49 (d, J = 6.8 Hz, 2 H), 8.13 (s, 1 H), 10.33 (s, 1 H); 13 C NMR (DMSO-d6, 75 MHz): δ 13.3, 24.7, 52.1, 55.1, 62.0, 113.9, 120.7, 125.4, 127.8, 128.3, 128.34, 128.8, 128.85, 129.6, 130.4, 131.4, 133.56, 133.62, 135.2, 136.0, 142.3, 155.5, 157.3, 163.6, 164.0; Anal. Calcd. for C31H28N6O4S: C, 64.12; H, 4.86; N, 14.47. Found: C, 63.90; H, 4.52; N, 14.74; ESI-MS m/z: 580.1 [M]+.

Ethyl 3-(((1-(2-((4-fluorophenyl)amino)-2-oxoethyl)-1H-1,2,3-triazol-4-yl)methyl)thio)-5,6-diphenylpyridazine-4-carboxylate (10 g)

Yield (79%, 0.45 g); Dark Yellow solid; m.p. = 211–213 °C; IR (KBr, cm-1): 3616, 1731 (C = O), 1639, 1442, 1332, 1191; 1 H NMR (DMSO-d6, 500 MHz): δ 0.89 (t, J = 7.1 Hz, 3 H), 4.06 (q, J = 7.0 Hz, 2 H), 4.81 (s, 2 H), 5.33 (s, 2 H), 7.15–7.17 (m, 2 H), 7.27–7.32 (m, 10 H), 7.64–7.66 (m, 2 H), 8.16 (s, 1 H), 10.82 (s, 1 H); 13 C NMR (DMSO-d6, 125 MHz): δ, 13.8, 25.2, 52.6, 62.6, 115.9 (d, J = 21 Hz), 121.5 (d, J = 8 Hz), 128.2, 128.3, 128.8, 129.1, 129.3, 129.5, 129.6, 129.7, 130.1, 130.9, 134.1, 135.2, 136.5, 156.0, 157.9, 162.3 (d, J = 248 Hz), 164.5, 165.6; Anal. Calcd. for C30H25FN6O3S: C, 63.37; H, 4.43; N, 14.78. Found: C, 63.02; H, 4.69; N, 15.01; ESI-MS m/z: 568.2 [M]+.

Ethyl 3-(((1-(2-((2-chlorophenyl)amino)-2-oxoethyl)-1H-1,2,3-triazol-4-yl)methyl)thio)-5,6-diphenylpyridazine-4-carboxylate (10 h)

Yield (62%, 0.36 g); Dark Yellow solid; m.p. = 161–163 °C; IR (KBr, cm-1): 3607, 1730 (C = O), 1639, 1445, 1358, 1166; 1 H NMR (DMSO-d6, 300 MHz): δ = 0.87 (t, J = 6.8 Hz, 3 H), 4.03 (q, J = 6.7 Hz, 2 H), 4.79 (s, 2 H), 5.44 (s, 2 H), 7.13–7.15 (m, 2 H), 7.28–7.44 (m, 10 H), 7.49 (d, J = 7.8 Hz, 1 H), 7.74 (d, J = 7.8 Hz, 1 H), 8.16 (s, 1 H), 10.04 (s, 1 H); 13 C NMR (DMSO-d6, 75 MHz): δ 13.2, 24.7, 51.9, 62.0, 125.7, 126.1, 126.6, 127.4, 127.8, 128.2, 128.3, 128.5, 128.76, 128.82, 128.94, 129.5, 129.6, 130.4, 133.6, 134.1, 135.1, 135.9, 155.4, 157.3, 164.0, 164.8; Anal. Calcd. for C30H25ClN6O3S: C, 61.59; H, 4.31; N, 14.36. Found: C, 61.80; H, 4.14; N, 14.69; ESI-MS m/z: 586.1 [M + 2]+.

Ethyl 3-(((1-(2-((3-chlorophenyl)amino)-2-oxoethyl)-1H-1,2,3-triazol-4-yl)methyl)thio)-5,6-diphenylpyridazine-4-carboxylate (10i)

Yield (60%, 0.35 g); Dark Yellow solid; m.p. = 200–202 °C; IR (KBr, cm-1): 3621, 1730 (C = O), 1639, 1429, 1341, 1175; 1 H NMR (DMSO-d6, 300 MHz): δ = 0.87 (t, J = 7.0 Hz, 3 H), 4.03 (q, J = 6.7 Hz, 2 H), 4.79 (s, 2 H), 5.33 (s, 2 H), 7.12–7.14 (m, 3 H), 7.26–7.44 (m, 10 H), 7.44 (d, J = 8.1 Hz, 1 H), 8.13 (s, 1 H), 10.64 (s, 1 H); 13 C NMR (DMSO-d6, 75 MHz): δ 13.3, 24.7, 52.2, 62.0, 117.6, 118.7, 123.4, 125.5, 127.8, 128.2, 128.3, 128.8, 128.9, 129.6, 130.4, 130.5, 133.1, 133.5, 133.6, 135.1, 135.9, 139.8, 155.4, 157.3, 164.0, 164.6; Anal. Calcd. for C30H25ClN6O3S: C, 61.59; H, 4.31; N, 14.36. Found: C, 61.31; H, 4.53; N, 14.02; ESI-MS m/z: 586.2 [M + 2]+.

Ethyl 3-(((1-(2-((4-chlorophenyl)amino)-2-oxoethyl)-1H-1,2,3-triazol-4-yl)methyl)thio)-5,6-diphenylpyridazine-4-carboxylate (10j)

Yield (74%, 0.43 g); Dark Yellow solid; m.p. = 195–197 °C; IR (KBr, cm-1): 3622, 1710 (C = O), 1646, 1442, 1331, 1192; 1 H NMR (DMSO-d6, 500 MHz): δ = 0.89 (t, J = 7.0 Hz, 3 H), 4.06 (q, J = 6.9 Hz, 2 H), 4.81 (s, 2 H), 5.43 (s, 2 H), 7.13–7.15 (m, 2 H), 7.20–7.30 (m, 10 H), 7.83 (d, J = 8.5 Hz, 2 H), 8.18 (s, 1 H), 11.06 (s, 1 H); 13 C NMR (DMSO-d6, 125 MHz): δ 13.8, 25.2, 52.8, 62.6, 119.5, 125.6, 128.3, 128.79, 128.86, 129.1, 129.3, 129.5, 130.0, 130.1, 130.9, 134.1, 135.7, 136.5, 143.1, 145.0, 155.9, 157.9, 164.5, 165.8; Anal. Calcd. for C30H25ClN6O3S: C, 61.59; H, 4.31; N, 14.36. Found: C, 61.25; H, 4.60; N, 14.02.

Ethyl 3-(((1-(2-((4-bromophenyl)amino)-2-oxoethyl)-1H-1,2,3-triazol-4-yl)methyl)thio)-5,6-diphenylpyridazine-4-carboxylate (10k)

Yield (61%, 0.38 g); Dark Yellow solid; m.p. = 224–226 °C; IR (KBr, cm-1): 3632, 1730 (C = O), 1651, 1440, 1352, 1191; 1 H NMR (DMSO-d6, 300 MHz): δ = 0.86 (t, J = 6.9 Hz, 3 H), 4.02 (q, J = 6.8 Hz, 2 H), 4.80 (s, 2 H), 5.33 (s, 2 H), 7.15–7.16 (m, 2 H), 7.27–7.37 (m, 10 H), 7.54 (d, J = 8.9 Hz, 2 H), 8.14 (s, 1 H), 10.60 (s, 1 H); 13 C NMR (DMSO-d6, 75 MHz): δ 13.3, 24.7, 52.2, 62.0, 115.4, 121.1, 127.8, 128.2, 128.3, 128.6, 128.8, 128.9, 129.0, 129.6, 130.4, 131.6, 133.6, 135.2, 135.9, 137.7. 155.4, 157.3, 164.0, 164.3; Anal. Calcd. for C30H25BrN6O3S: C, 57.24; H, 4.00; N, 13.35. Found: C, 57.00; H, 4.25; N, 13.60; ESI-MS m/z: 630.1 [M + 2]+.

Ethyl 3-(((1-(2-((3,5-dichlorophenyl)amino)-2-oxoethyl)-1H-1,2,3-triazol-4-yl)methyl)thio)-5,6-diphenylpyridazine-4-carboxylate (10 l)

Yield (64%, 0.39 g); Dark Yellow solid; m.p. = 185–187 °C; IR (KBr, cm-1): 3616, 1733 (C = O), 1631, 1444, 1330, 1166; 1 H NMR (DMSO-d6, 500 MHz): δ = 0.86 (t, J = 7.1 Hz, 3 H), 4.02 (q, J = 7.0 Hz, 2 H), 4.81 (s, 2 H), 5.40 (s, 2 H), 7.29–7.37 (m, 10 H), 7.77–7.78 (m, 3 H), 8.17 (s, 1 H), 10.90 (s, 1 H).; 13 C NMR (DMSO-d6, 125 MHz): δ 13.8, 25.2, 52.8, 62.6, 106.1, 119.4, 119.8, 126.0, 128.8, 129.1, 129.3, 129.5, 130.1, 130.9, 133.8, 133.9, 134.1, 135.7, 136.5, 143.0, 155.9, 157.9, 164.5, 165.6.; Anal. Calcd. for C30H24Cl2N6O3S: C, 58.16; H, 3.90; N, 13.57. Found: C, 57.95; H, 4.22; N, 13.80; ESI-MS m/z: 622.1 [M + 4]+.

Ethyl 3-(((1-(2-((3,4-dichlorophenyl)amino)-2-oxoethyl)-1H-1,2,3-triazol-4-yl)methyl)thio)-5,6-diphenylpyridazine-4-carboxylate (10 m)

Yield (78%, 0.48 g); Dark Yellow solid; m.p. = 166–168 °C; IR (KBr, cm-1): 3606, 1725 (C = O), 1639, 1441, 1330, 1175; 1 H NMR (DMSO-d6, 500 MHz): δ = 0.86 (t, J = 6.4 Hz, 3 H), 4.02 (m, 2 H), 4.80 (s, 2 H), 5.35 (s, 2 H), 7.30–7.42 (m, 12 H), 7.54 (s, 1 H), 8.15 (s, 1 H), 10.77 (s, 1 H); 13 C NMR (DMSO-d6, 125 MHz): δ 13.3, 24.7, 52.2, 62.1, 119.2, 120.4, 125.3, 125.5, 127.8, 128.3, 128.6, 128.8, 129.0, 129.6, 130.4, 130.8, 131.1, 133.5, 133.6, 135.1, 135.9, 138.4, 155.4, 157.3, 164.0, 164.8; Anal. Calcd. for C30H24Cl2N6O3S: C, 58.16; H, 3.90; N, 13.57. Found: C, 58.42; H, 3.61; N, 13.39; ESI-MS m/z: 622.3 [M + 4]+.

Ethyl 3-(((1-(2-((3-cyanophenyl)amino)-2-oxoethyl)-1H-1,2,3-triazol-4-yl)methyl)thio)-5,6-diphenylpyridazine-4-carboxylate (10n)

Yield (70%, 0.40 g); Dark Yellow solid; m.p. = 196–198 °C; IR (KBr, cm-1): 3601, 2221, 1724 (C = O), 1644, 1439, 1327, 1180; 1 H NMR (DMSO-d6, 500 MHz): δ = 0.89 (t, J = 6.4 Hz, 3 H), 4.06 (m, 2 H), 4.81 (s, 2 H), 5.32 (s, 2 H), 7.16–7.21 (m, 5 H), 7.29–7.33 (m, 6 H), 7.48–7.51 (m, 3 H), 8.15 (s, 1 H), 10.40 (s, 1 H); 13 C NMR (DMSO-d6, 125 MHz): δ 13.8, 24.4, 52.7, 62.6, 119.8, 119.9, 126.0, 127.0, 127.1, 128.3, 128.8, 129.1, 129.3, 129.5, 130.2, 130.9, 134.1 (2 C), 135.7, 136.5, 136.6, 142.9, 144.3, 156.0, 158.0, 164.5, 166.4; Anal. Calcd. for C31H25N7O3S: C, 64.68; H, 4.38; N, 17.03. Found: C, 64.99; H, 4.55; N, 16.88; ESI-MS m/z: 575.2 [M]+.

Ethyl 3-(((1-(2-((3-nitrophenyl)amino)-2-oxoethyl)-1H-1,2,3-triazol-4-yl)methyl)thio)-5,6-diphenylpyridazine-4-carboxylate (10p)

Yield (81%, 0.48 g); Dark Yellow solid; m.p. = 201–203 °C; IR (KBr, cm-1): 3592, 1730 (C = O), 1637, 1549, 1432, 1356, 1189; 1 H NMR (DMSO-d6, 300 MHz): δ = 0.87 (t, J = 7.0 Hz, 3 H), 4.06 (d, J = 6.8 Hz, 2 H), 4.80 (s, 2 H), 5.39 (s, 2 H), 7.13 (t, J = 6.8 Hz, 2 H), 7.32–7.45 (m, 9 H), 7.61 (t, J = 7.8 Hz, 1 H), 7.91 (t, J = 8.0 Hz, 1 H), 8.17 (s, 1 H), 8.58 (s, 1 H), 10.97 (s, 1 H); 13 C NMR (DMSO-d6, 75 MHz): δ 13.3, 24.7, 52.2, 62.1, 113.4, 118.3, 125.1, 127.8, 128.3, 128.4, 128.6, 128.8, 128.9, 129.0, 129.6, 130.4, 133.5, 133.6, 135.2, 136.0, 139.4, 147.9, 155.4, 157.3, 164.0, 165.1; Anal. Calcd. for C30H25N7O5S: C, 60.49; H, 4.23; N, 16.46. Found: C, 60.69; H, 3.99; N, 16.19; ESI-MS m/z: 595.2 [M]+.

Ethyl 3-(((1-(2-((4-nitrophenyl)amino)-2-oxoethyl)-1H-1,2,3-triazol-4-yl)methyl)thio)-5,6-diphenylpyridazine-4-carboxylate (10q)

Yield (73%, 0.43 g); Dark Yellow solid; m.p. = 221–223 °C; IR (KBr, cm-1): 3610, 1712 (C = O), 1636, 1560, 1432, 1352, 1179; 1 H NMR (DMSO-d6, 500 MHz): δ = 0.89 (t, J = 6.4 Hz, 3 H), 4.07 (m, 2 H), 4.81 (s, 2 H), 5.35 (s, 2 H), 7.16 (d, J = 8.1 Hz, 2 H), 7.29–7.40 (m, 10 H), 7.16 (d, J = 8.2 Hz, 2 H), 8.16 (s, 1 H), 10.61 (s, 1 H); 13 C NMR (DMSO-d6, 125 MHz): δ 13.8, 25.2, 52.7, 62.6, 121.2, 121.3, 126.0, 127.8, 128.3, 128.8, 129.1, 129.2, 129.3, 129.5, 130.1, 130.9, 134.1, 135.7, 136.4, 137.8, 155.9, 157.8, 164.5, 164.9; Anal. Calcd. for C30H25N7O5S: C, 60.49; H, 4.23; N, 16.46. Found: C, 60.18; H, 4.09; N, 16.66; ESI-MS m/z: 596.1 [M + H]+.

Ethyl 3-(((1-(2-((3-methyl-4-nitrophenyl)amino)-2-oxoethyl)-1H-1,2,3-triazol-4-yl)methyl)thio)-5,6-diphenylpyridazine-4-carboxylate (10r)

Yield (65%, 0.39 g); Dark Yellow solid; m.p. = 212–214 °C; IR (KBr, cm-1): 3602, 1722 (C = O), 1641, 1551, 1448, 1352, 1182; 1 H NMR (DMSO-d6, 300 MHz): δ = 0.86 (t, J = 6.4 Hz, 3 H), 1.10 (s, 3 H), 4.04 (m, 2 H), 4.80 (s, 2 H), 5.37 (s, 2 H), 7.13 (d, J = 6.8 Hz, 2 H), 7.28–7.32 (m, 9 H), 7.40–7.43 (m, 1 H), 8.17 (m, 1 H), 8.33 (s, 1 H), 10.85 (s, 1 H); 13 C NMR (DMSO-d6, 75 MHz): δ 13.3, 19.2, 24.7, 52.2, 62.1, 114.5, 123.7, 127.7, 127.8, 128.27, 128.34, 128.6, 128.8, 128.86, 128.97, 129.6, 130.4, 133.2, 133.6, 135.2, 136.0, 137.2, 148.5, 155.5, 157.3, 164.0, 164.8; Anal. Calcd. for C31H27N7O5S: C, 61.07; H, 4.46; N, 16.08. Found: C, 60.75; H, 4.19; N, 16.40; ESI-MS m/z: 609.2 [M]+.

Biological studies

Rat α‑glucosidase assay

Based on the reported method of Lossow et al., rat small intestine α-glucosidase (EC 3.2.1.20) was prepared. The in-vitro activity was determined by the measurement of 4-nitrophenol which was released from para-nitrophenyl α-D glucopyranoside [44, 45]. The preparation of 200 µL was performed as follows: the enzyme solution (190 µL, 0.15 units/ml), different concentrations of target compounds (1, 10, 20, 50, 100, 500 and 1000 µM (5 µL), potassium phosphate buffer. Final compounds were dissolved in DMSO (not exceed than 5% of final volume) and pre-incubated at 37 °C, p-nitrophenyl glucopyranoside and then substrate (5 µL, 3 mM), was added to the enzyme solution and incubated for further one hour at 37 °C. Finally, by using Cytation 3 hybrid microplate reader (BioTek, USA) any change in the absorbance was measured at 405 nm. By using GraphPadprism 6.0 (SanDiego, California, USA) (https://www.graphpad.com/scientificsoftware/prism/) was used to obtain IC50 values of tested compounds.

Kinetic analysis

Modes of enzyme inhibition were determined by recording the effect of various concentrations of the substrates 1, 2.5, 5 or 10 mM pNPG for α-glucosidase on Lineweaver–Burk plots using calculated Vmax and Km. For inhibition test of α-glucosidase: the concentrations applied for compounds 10k was 0.1, 1, 5 and 10 nM. Inhibition constants (Ki) were determined by depicting the secondary plot of Km against various concentrations of inhibitors.

Docking studies

Docking study of compound 10k was done using Autodock 4.2.1 software. The structure of the targeted protein α-glucosidase (PDB: 5NN88) was taken from RCSB data bank [26]. The analysis of docking results were performed by Discovery Studio visualizer 4.5.

Results and discussions

Chemistry

The preparation of target compounds is described in scheme 1. Compound 2 was prepared from the previously reported procedure [43, 46]. Subsequently, the cyclization occurred with diethyl malonate and sodium in ethanol at reflux temperature to yield ethyl 3-oxo-5,6-diphenyl-2,3-dihydropyridazine-4-carboxylate. Then, the conversion of carbonyl group into SH was achieved by Lawesson’s reagent in toluene after 18 h heating at reflux condition. The propargylated product 5, ethyl 5,6-diphenyl-3-(prop-2-yn-1-ylthio)pyridazine-4-carboxylate, was obtained from the reaction of compound 4 and propargyl bromide in dimethyl formamide in the presence of K2CO3. On the other hand, the reaction of aromatic amines and chloroacetyl chloride was conducted in 1,2-dichloroethane and in the presence of triethylamine as a proton acceptor to obtain 2-chloro-N-arylacetamide. After work-up and crystallization, the substitution reaction of compound 8 with sodium azide in DMSO yielded compound 9. The click reaction between compound 5 and 9 in DMF and in the presence of triethylamine and catalytic amounts of CuI resulted in target compounds 10a-r in good yields.

Scheme 1
scheme 1

Synthesis of target compounds. Synthesis of target compounds. Reagents and conditions: (a) hydrazine hydrate, methanol, reflux, 15 min.; (b) Na, EtOH, diethyl malonate, reflux; (c) Lawesson’s reagent, toluene, reflux, 18 h; (d) propargyl bromide, DMF, K2CO3, 50 °C; (e) Et3N, DCE, r.t.; (f) NaN3, DMSO, r.t.; (g) Et3N, DMF, CuI, r.t

The 1H NMR of the most potent compound, 10k, in DMSO-d6 was recorded and clearly showed the characteristic triazole and NH protons as singlets at 8.14, 10.60 ppm, respectively. SCH2 and NCH2 were resonated as singlets at 4.80 and 5.33 ppm, respectively. The ethoxy group was appeared at 0.86 as triplet (J = 6.9 Hz) and at 4.02 as quartet (J = 6.8 Hz). The aromatic protons near bromine were appeared at 7.54 ppm as doublet showing the ortho coupling constant of 8.9 Hz. Besides, other aromatic protons as multiplets at appropriate chemical shifts confirmed the proposed structure. The 13C NMR spectrum in the same solvent at 75 MHz showed four aliphatic carbons at 13.3, 24.7, 52.2, 62.0 ppm related to CH3, SCH2, NCH2 OCH2. The number of aromatic carbons in the normal range is in accordance to the proposed structure.

In vitro α-glucosidase inhibitory activities

The SAR studies divulged that different substituents either electron-donating or withdrawing on aromatic moiety along with their positions affected the inhibition. The results are shown as IC50s in Table 1. Compounds with unsubstituted phenyl ring and different substituents including methyl, methoxy, isopropyl, fluorine, chlorine, bromine, nitro and cyano at different positions of aryl ring were found to have varying degree of inhibitory potential. Except few ones, all compounds were active against α-glucosidase enzyme (IC50s = 1.7 to 86.5 µM). Compound 10k bearing 4-bromo substituent was found to be 100 folds more active with IC50 value of 1.7 µM compared to standard drug (IC50 = 173 µM). Compound 10l having 3,5-dichloro group was the second most active compound. Further studies indicated that compound 10 h bearing chlorine (–Cl) group at ortho position of phenyl ring displayed stronger α-glucosidase inhibitory activity (IC50 = 14.9 µM) than meta- and para-chloro substituted analogues (10i and 10j). Interestingly, p-bromo substituted analogue (10k) showed strong inhibitory activity compared to p-fluoro and p-chloro analogues. Interesting to observe that compound 10i exhibited no inhibitory activity, while the combination of second chlorine atom (–Cl) generated active compounds (10 l, 10 m). Electron-releasing substituted analogues is more active than electron-withdrawing substituted compounds which are exemplified by meta-methoxy, methyl (m–OMe, m-Me) substituted 1,2,3-triazoles being more active than meta-nitro (m–NO2) and meta-cyano (m–CN) substituted analogues. While, 4-nitro substituted compound 10q (IC50 = 47.1 µM) was active, the introduction of methyl at meta position resulted in no activity at the resultant compound, 10r. The movement of electron-withdrawing groups meaning nitro and cyano from meta to para generated active compounds (10o and 10q). This trend was also observed in the case of chlorine-substituted compounds (10ivs. 10j). The cytotoxicity of compound 10k, the most active compound, was investigated against the normal cell line. No toxicity was observed against HDF [28].

Table 1 In vitro α-glucosidase inhibitory activities and yields of target compounds.a,b

Kinetic studies

The activity of 4-nitrophenyl-β-D-galactopyranoside (PNPG) was investigated in the presence and absence of compound 10k. The results were analyzed by Lineweaver-Burk and confirmed that this compound induced uncompetitive inhibition on α-glucosidase (Fig. 2). The slope was not altered by the presence of this compound and increasing concentrations of the substrate resulted in parallel lines. By using secondary plot, the inhibitor constant Ki was determined to be 125 nM.

Fig. 2
figure 2

The relative activity against the concentrations of α-glucosidase in the presence of compound 10k (0, 0.1, 1, 5 and 10 nM)

Docking studies

To gain insights into possible binding modes with α-glucosidase, molecular docking was performed and the results are shown in Fig. 3. It is observed that the hydrogen bond, pi-sigma and the pi-sulfur and pi-alkyl interactions are the main interactions of compound 10k contributing to the binding affinity of this compound with the enzyme. The most potent compound exhibited favorable interactions with amino acid residues. The triazole ring formed hydrogen bond with Leu677 to enhance the binding affinity. The van der Waals interactions are formed with Phe649, Trp 376, Asp616, Leu283, Asp282. The ethoxy group could form pi-sigma interaction with residue of the enzyme.

Fig. 3
figure 3

2D (a) and 3D (b) binding modes the most active 10k skeleton

Conclusion

In summary, a newly synthesized series of pyridazine-triazole derivatives were synthesized from benzil and their inhibitory activity toward α-glucosidase were also evaluated. All synthesized compounds were new and evaluated by IR, 1H NMR, 13C NMR and Mass spectroscopy. The obtained results showed that most of the target compounds showed significant inhibitory potency. The kinetic studies revealed that the inhibition was uncompetitive. Besides, the suitable interactions and no toxicity of the most potent compound confirmed that these derivatives could be regarded as a good candidate for further investigation and optimization.

Data Availability

All data generated or analyzed during this study are included in this published article and its additional information files.

References

  1. Al-Hassan N. Definition of diabetes mellitus. Br J Gen Pract. 2003;53:567–8.

    PubMed  PubMed Central  Google Scholar 

  2. World Health Organization., Global report on diabetes. https://www.who.int/health-topics/diabetes/en/ (assessed 24 May 2022)

  3. Kannel WB, Mcgee DL. Diabetes and cardiovascular risk factors: the Framingham study. Circulation. 1979;59:8–13.

    Article  CAS  PubMed  Google Scholar 

  4. Krolewski AS, Kosinski EJ, Warram JH, Leland OS, Busick EJ, Asmal AC, Rand LI, Christlieb AR, Bradley RF, Kahn CR. Magnitude and determinants of coronary artery disease in juvenile-onset, insulin-dependent diabetes mellitus. Am J Cardiol. 1987;59:750–55.

    Article  CAS  PubMed  Google Scholar 

  5. Ogurtsova K, Rocha Fernandes JD, Huang Y, Linnenkamp U, Guariguata L, Cho NH, Cavan D, Shaw JE, Makaroff LE. IDF Diabetes Atlas: global estimates for the prevalence of diabetes for 2015 and 2040. Diabetes Res Clin Pract. 2017;128:40–50.

    Article  CAS  PubMed  Google Scholar 

  6. Gloster TM, Davies GJ. Glycosidase inhibition: assessing mimicry of the transition state. Org Biomol Chem. 2010;8:305–20.

    Article  CAS  PubMed  Google Scholar 

  7. Liu Z, Ma S. Recent advances in synthetic α-Glucosidase inhibitors. Chem Med Chem. 2017;12:819–29.

    Article  CAS  PubMed  Google Scholar 

  8. Hossain MA, Pervin R. Current antidiabetic drugs: review of their efficacy and safety in: nutritional and therapeutic interventions for diabetes and metabolic. Elsevier; 2018. pp. 455–73.

  9. He ZX, Zhou ZW, Yang Y, Yang T, Pan SY, Qiu JX, Zhou SF. Overview of clinically approved oral antidiabetic agents for the treatment of type 2 diabetes mellitus. Clin Exp Pharmacol Physiol. 2015;42:125–38.

    Article  CAS  PubMed  Google Scholar 

  10. Yee HS, Fong NT. A review of the Safety and Efficacy of Acarbose in Diabetes Mellitus. Pharmacotherapy. 1996;16:792–805.

    CAS  PubMed  Google Scholar 

  11. Kaku K. Efficacy of voglibose in type 2 diabetes. Pharmacotherapy. 2014;15:1181–90.

    CAS  Google Scholar 

  12. Scott LJ, Spencer CM. Miglitol: a review of its therapeutic potential in type 2 diabetes Mellitus. Drugs. 2000;59:521–49.

    Article  CAS  PubMed  Google Scholar 

  13. Dhameja M, Gupta P. Synthetic heterocyclic candidates as promising α-Glucosidase inhibitors: an overview. Eur J Med Chem. 2019;176:343–77.

    Article  CAS  PubMed  Google Scholar 

  14. Ali M, Khan KM, Mahdavi M, Jabbar A, Shamim S, Salar U, Taha M, Perveen S, Larijani B, Faramarzi MA. Synthesis, in vitro and in silico screening of 2-amino-4-aryl-6-(phenylthio) pyridine-3,5-dicarbonitriles as novel α-glucosidase inhibitors. Bioorg Chem. 2020;100:103879.

    Article  CAS  PubMed  Google Scholar 

  15. Hameed S, Khan KM, Taslimi P, Salar U, Taskin-Tok T, Kisa D, Saleem F, Solangi M, Ahmad MHU, Kiran Rani A. Evaluation of synthetic 2-aryl quinoxaline derivatives as α-amylase, α-glucosidase, acetylcholinesterase, and butyrylcholinesterase inhibitors. Int J Biol Macromol. 2022;211:653–68.

    Article  CAS  PubMed  Google Scholar 

  16. Bushra SS, Khan KM, Ullah N, Mahdavi M, Faramarzi MA, Larijani B, Salar U, Rafique R, Taha M, Perveen S. Synthesis, in vitro, and in silico evaluation of Indazole Schiff bases as potential α-glucosidase inhibitors. J Mol Struct. 2021;1242:130826.

    Article  CAS  Google Scholar 

  17. Yousuf H, Shamim S, Khan KM, Chigurupati S, Kanwal, Hameed S, Khan MN, Taha M, Arfeen M. Dihydropyridines as potential α-amylase and α-glucosidase inhibitors: synthesis, in vitro and in silico studies. Bioorg Chem. 2020;96:103581.

    Article  CAS  PubMed  Google Scholar 

  18. Bansal R, Thota S. Pyridazin-3(2H)-ones: the versatile pharmacophore of medicinal significance. Med Chem Res. 2013;22:2539–52.

    Article  CAS  Google Scholar 

  19. Loksha YM, Abd-Alhasee MM. Synthesis and biological screening of some novel 6-substituted 2-alkylpyridazin-3(2H)-ones as anti-inflammatory and analgesic agents. Arch Pharm Chem Life Sci. 2020;1900295.

  20. Singh B, Bhatia R, Pani B, Gupta D. Synthesis, crystal structures and biological evaluation of new pyridazine derivatives. J Mol Struct. 2020;1200:127084.

    Article  Google Scholar 

  21. Harris RR, Black L, Surapaneni S, Kolasa T, Majest S, Namovic MT, Grayson G, Komater V, Wilcox D, King L, Marsh K, Jarvis MF, Nuss M, Nellans HL, Pruesser GA, Reinhart B, Cox P, Jacobson A, Stewart M, Carter CJ. ABT-963 [2-(3,4-difluoro-phenyl)-4-(3-hydroxy-3-methyl-butoxy)-5-(4-methanesulfonyl-phenyl)-2H-pyridazin-3-one], a highly potent and selective disubstituted pyridazinone cyclooxgenase-2 inhibitor. J Pharmacol Exp Ther. 2004;311:904–12.

    Article  CAS  PubMed  Google Scholar 

  22. Sharma D, Bansal R. Synthesis of 2-substituted-4-aryl-6-phenylpyridazin-3(2H)- ones as potential anti-inflammatory and analgesic agents with cardioprotective and ulcerogenic sparing effects. Med Chem Res. 2016;25:1574–89.

    Article  CAS  Google Scholar 

  23. Krall J, Bavo F, Falk-Petersen CB, Jensen CH, Nielsen JO, Tian Y, Anglani V, Kongstad KT, Piilgaard L, Nielsen B, Gloriam DE, Kehler J, Jensen AAK, Harpsøe K, Wellendorph P, Frølund B. Discovery of 2-(Imidazo[1,2-b]pyridazin 2-yl)acetic acid as a New Class of Ligands Selective for the γ-Hydroxybutyric AcidGHB) High-Affinity binding Sites. J Med Chem. 2019;62:2798–813.

    Article  CAS  PubMed  Google Scholar 

  24. Sergeev PG, Nenajdenko VG. Recent advances in the chemistry of pyridazine an important representative of six-membered nitrogen heterocycles. Russ Chem Rev. 2020;89:393–429.

    Article  CAS  Google Scholar 

  25. Ahmed EM, Hassan MSA, El-Malah AA, Kassab AE. New pyridazine derivatives as selective COX-2 inhibitors and potential antiinflammatory agents; design, synthesis and biological evaluation. Bioorg Chem. 2020;95:103497.

    Article  CAS  PubMed  Google Scholar 

  26. Deora GS, Qin CX, Vecchio EA, Debono AJ, Priebbenow DL, Brady RM, Beveridge J, Teguh SC, Deo M, May LT, Krippner G, Ritchie RH, Baell JB. Substituted Pyridazin-3(2H)-ones as highly potent and biased formyl peptide receptor agonists. J Med Chem. 2019;62:5242–48.

    Article  CAS  PubMed  Google Scholar 

  27. Kolb HC, Finn MG, Sharpless KB. Click Chemistry: diverse chemical function from a few good reactions. Angew Chem Int Ed. 2001;40:2004.

    Article  CAS  Google Scholar 

  28. Wu P, Feldman AK, Nugent K, Hawker CJ, Scheel A, Voit B, Pyun J, Frechet JMJ, Sharpless KB, Fokin V. Efficiency and fidelity in a click-chemistry route to triazole dendrimers by the copper(i)-catalyzed ligation of azides and alkynes. Angew Chem Int Ed. 2004;43:3928–32.

    Article  CAS  Google Scholar 

  29. Tron GC, Pirali T, Billington RA, Canonico PL, Sorba G, Genazzani A. Click chemistry reactions in medicinal chemistry: applications of the 1,3-dipolar cycloaddition between azides and alkynes. Med Res Rev. 2008;28:278–308.

    Article  CAS  PubMed  Google Scholar 

  30. Rani A, Singh G, Singh A, Maqbool U, Kaur G, Singh J. CuAAC-Ensembled 1,2,3-TriazoleLinked Isosteres as Pharmacophores in Drug Discovery. RSC Adv. 2010;10:5610–35.

    Article  Google Scholar 

  31. Jiang X, Hao X, Jing L, Wu G, Kang D, Liu X, Zhan P. Recent applications of click Chemistry in Drug Discovery. Expert Opin Drug Disc. 2019;14:779–89.

    Article  CAS  Google Scholar 

  32. Moghimi S, Salarinejad S, Toolabi M, Firoozpour L, Sadat Ebrahimi SE, Safari F, Madani-Qamsari F, Mojtabavi S, Faramarzi MA, Karima S, Pakrad R, Foroumadi A. Synthesis, in-vitro evaluation, molecular docking, and kinetic studies of pyridazine-triazole hybrid system as novel α-glucosidase inhibitors. Bioorg Chem. 2021;109:104670.

    Article  CAS  PubMed  Google Scholar 

  33. Peytam F, Takalloobanafshi G, Saadattalab T, Norouzbahari M, Emamgholipour Z, Moghimi S, Firoozpour L, Bijanzadeh HR, Faramarzi MA, Mojtabavi S, Rashidi–Ranjbar P, Karima S, Pakraad R, Foroumadi A. Design, synthesis, molecular docking, and in vitro α–glucosidase inhibitory activities of novel 3-amino–2,4–diarylbenzo[4,5]imidazo[1,2–a]pyrimidines against yeast and rat α–glucosidase. Sci Rep. 2021;11:11911.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Peytam F, Adib M, Shourgeshty R, Firoozpour L, Rahmanian-Jazi M, Jahani M, Moghimi S, Divsalar K, Faramarzi MA, Mojtabavi S, Safari F, Mahdavi M, Foroumadi A. An efficient and targeted synthetic approach towards new highly substituted 6-aminopyrazolo[1,5-a] pyrimidines with α-glucosidase inhibitory activity. Sci Rep. 2020;10:2595.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Moghimi S, Toolabi M, Salarinejad S, Firoozpour L, Sadat Ebrahimi SE, Safari F, Mojtabavi S, Faramarzi MA, Foroumadi A. Design and synthesis of novel pyridazine N-aryl acetamides: In-vitro evaluation of α-glucosidase inhibition,docking, and kinetic studies. Bioorg Chem. 2020;102:14071.

    Article  Google Scholar 

  36. Wang J, Wang D, He X, Li J, Li Z, Peng G. Synthesis and biological evaluation of novel 1,2,4-triazine derivatives bearing carbazole moiety as potent α-glucosidase inhibitors. Bioorg Med Chem Lett. 2016;26:2806–09.

    Article  CAS  PubMed  Google Scholar 

  37. Wang G, Peng Z, Wang J, Li X, Li J. Synthesis, in vitro evaluation and molecular docking studies of novel triazine-triazole derivatives as potential a-glucosidase inhibitors. Eur J Med Chem. 2017;125:423–29.

    Article  CAS  PubMed  Google Scholar 

  38. Wang G, Li X, Wang J, Xie Z, Li L, Chen M, Chen S, Peng Y. Synthesis, molecular docking and α-glucosidase inhibition of 2-((5,6-diphenyl-1,2,4-triazin-3-yl)thio)-N-arylacetamides. Bioorg Med Chem Lett. 2017;27:1115–18.

    Article  CAS  PubMed  Google Scholar 

  39. Wang G, Peng Z, Gong Z, Li Y. Synthesis, biological evaluation, and docking studies of novel 5,6-diaryl-1,2,4-triazine thiazole derivatives as a new class of α-glucosidase inhibitors. Bioorg Chem. 2018;78:195–200.

    Article  CAS  PubMed  Google Scholar 

  40. Shamim S, Khan KM, Ullah N, Chigurupati S, Wadood A, Rehman AU, Ali M, Salar U, Alhowail A, Taha M, Perveen S. Synthesis and screening of (E)-3-(2-benzylidenehydrazinyl)-5,6-diphenyl-1,2,4-triazine analogs as novel dual inhibitors of α-amylase and α-glucosidase. Bioorg Chem. 2020;101:103979.

    Article  CAS  PubMed  Google Scholar 

  41. Ali F, Khan KM, Salar U, Taha M, Ismail NH, Wadood A, Riaz M, Perveen S. Hydrazinyl arylthiazole based pyridine scaffolds: synthesis, structural characterization, in vitro α-glucosidase inhibitory activity, and in silico studies. Eur J Med Chem. 2017;138:255–72.

    Article  CAS  PubMed  Google Scholar 

  42. Basha FZ. New carbazole linked 1,2,3-triazoles as highly potent non-sugar α-glucosidase inhibitors. Bioorg Chem. 2017;74:72–81.

    Article  PubMed  Google Scholar 

  43. Al-kahraman MSAY, Al-kahraman YM, Singh GS. Evaluation of some classical hydrazones of ketones and 1,2-diketones as antileishmanial, antibacterial and antifungal agents. Arch Pharm Res. 2012;35:1009–13.

    Article  CAS  PubMed  Google Scholar 

  44. Lossow WJ, Migliorini RH, Brot N, Chaikoff IL. Effect of total exclusion of the exocrine pancreas in the rat upon in vitro esterification of C14 – labeled cholesterol by the intestine and upon lymphatic absorption of C14 – labeled cholesterol. J Lipid Res. 1964;5:198–202.

    Article  CAS  PubMed  Google Scholar 

  45. Kim JH, Cho CW, Kim HY, Kim KT, Choi GS, Kim HH, Cho IS, Kwon SJ, Choi SK, Yoon JY, Yang SY, Kang JS. KimYH. α-Glycosidase inhibition by prenylated and lavandulyl compounds from Sophora flavescens roots and in silico analysis. Int J Biol Macromol. 2017;102:960–69.

    Article  CAS  PubMed  Google Scholar 

  46. Schmidt P, Druey J. Heilmittelchemische Studien in der heterocyclischen Reihe. Mitteilung. Pyridazine II. Eine neue Pyridazinsynthese. Helv Chim Acta. 1954;15:134–40.

    Article  Google Scholar 

Download references

Acknowledgements

Research reported in this publication was supported by Elite Researcher Grant Committee under award number [995866] from the National Institute for Medical Research Development (NIMAD).

Funding

Research reported in this publication was supported by Elite Researcher Grant Committee under award number [995866] from the National Institute for Medical Research Development (NIMAD). There is no role of the funding body in the design of the study and collection, analysis, and interpretation of data and in writing the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

A.F. designed the target compounds. S.K. and S. E. S. E. interpreted the biological and chemical analysis. S. M. and S. S. wrote the manuscript. M. T. and L. F. performed docking studies. M. R. conducted the organic synthesis in the lab. R. P. and F. S. and S. M. S. performed biological tests. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Alireza Foroumadi.

Ethics declarations

Ethics approval and consent to participate

All methods were carried out in accordance with relevant guidelines and regulations of The Ministry of Health and Medical Education IR.TUMS.MEDICINE.REC.1400.1316. Protocols were conducted and approved by Shahid Beheshti University of Medical Sciences (SBMU).

Consent for publication

Not applicable.

Conflict of Interest

The authors declare that there are no conflicts of interest.

Competing interests

The authors declare that they have no competing interests.

Supporting information

Copies of 1 H-NMR 13 C-NMR spectra are available as supplementary data.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary Material 1

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Firoozpour, L., Moghimi, S., Salarinejad, S. et al. Synthesis, α-Glucosidase inhibitory activity and docking studies of Novel Ethyl 1,2,3-triazol-4-ylmethylthio-5,6-diphenylpyridazine-4-carboxylate derivatives. BMC Chemistry 17, 66 (2023). https://doi.org/10.1186/s13065-023-00973-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13065-023-00973-8

Keywords