Open Access Article
Nafise Asemanipoora,
Shahram Moradi*a,
Mohammad Ali Faramarzi
b,
Maryam Mohammadi-Khanaposhtani
c and
Mohammad Mahdavi
*d
aDepartment of Chemistry,NT.C., Islamic Azad University, Tehran, Iran. E-mail: shm_moradi@yahoo.com
bDepartment of Pharmaceutical Biotechnology, Faculty of Pharmacy and Biotechnology Research Center, Tehran University of Medical Sciences, Tehran, Iran
cCellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
dEndocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran. E-mail: momahdavi@tums.ac.ir
First published on 15th October 2025
The current work aims to introduce 5,6-diphenyl-1,2,4-triazine-hydrazineylidene-phenoxy-1,2,3-triazole-acetamide derivatives 13a–n as a new class of potent α-glucosidase inhibitors. These compounds were synthesized through well-known and effective chemical reactions in good yields. All title derivatives 13a–n showed high α-glucosidase inhibition in comparison to the standard inhibitor (acarbose). In this regard, the most potent compounds, compounds 13j and 13h, were approximately 6250- and 3947-fold more potent than acarbose, respectively. An in vitro kinetics study revealed that compound 13j is an uncompetitive α-glucosidase inhibitor. Molecular docking and molecular dynamics studies on compound 13j revealed highly favorable results, confirming stable binding interactions and robust complex formation of this compound with the enzyme's active site. Furthermore, in silico studies indicated that compound 13j possesses favorable and comparable drug-likeness, ADME, and toxicity profiles relative to acarbose, highlighting its potential as a promising lead.
N-heterocyclic containing compounds, both natural and synthetic, have always had special importance in medicinal chemistry.13–16 These compounds, ranging from amino acids and nucleic acids to complex plant alkaloids, are important building blocks in biology and pharmaceutical sciences.17 Some N-heterocycles, such as triazine and triazole, do not occur in nature and are produced synthetically, but they can clearly affect natural processes such as enzyme function.18,19 Our target enzyme in this work is α-glucosidase, and various derivatives containing 1,2,4-triazine or 1,2,3-triazole rings with high inhibitory activity against this enzyme were reported.20,21 On the other hand, one of the valuable linkers in the design of hybrid molecules as α-glucosidase inhibitors is the Schiff base.22
Our investigations on various α-glucosidase inhibitors containing triazole, triazine, and/or Schiff base structures led to find two series of valuable molecules: 5,6-diphenyl-1,2,4-triazine-hydrazineylidene derivatives A and phenoxy-1,2,3-triazole-acetamides B (Fig. 1).23,24 Based on molecular hybridization theory, we attached a 5,6-diphenyl-1,2,4-triazine-hydrazineylidene moiety to phenoxy-1,2,3-triazole-acetamide derivatives to achieve new α-glucosidase inhibitors 13a–n (Fig. 1).25–30
![]() | ||
| Fig. 1 Design strategy for 5,6-diphenyl-1,2,4-triazine-hydrazineylidene-phenoxy-1,2,3-triazole-acetamide derivatives 13a–n. | ||
:
1 v/v) was stirred at room temperature for 1 h. Subsequently, compound 10 (1 mmol), CuSO4 (7 mol%), and sodium ascorbate (14 mol%) were added to the mixture, and the reaction was stirred at room temperature for 24–48 h. After this period, cold water was added, and the resulting products 12a–n were filtered off, washed with water, and purified by recrystallization from ethanol.21
), 10.48 (s, 1H, NH–C
O), 8.29 (s, 1H, N
CH), 8.26 (s, 1H, triazole), 7.69 (d, J = 8.2 Hz, 2H, Ar), 7.59 (d, J = 8.0 Hz, 2H, Ar), 7.48 (d, J = 7.4 Hz, 2H, Ar), 7.43 (d, J = 8.9 Hz, 1H, Ar), 7.39 (d, J = 7.4 Hz, 2H, Ar), 7.37–7.36 (m, 2H, Ar), 7.35–7.34 (m, 3H, Ar), 7.33–7.30 (m, 1H, Ar), 7.14 (d, J = 8.1 Hz, 2H, Ar), 7.08 (t, J = 7.3 Hz, 2H, Ar), 5.37 (s, 2H, CH2–C
O), 5.24 (s, 2H, CH2–O); 13C NMR (126 MHz, DMSO-d6) δ 164.11 (NH–C
O), 159.14 (–N
C–N
), 156.31, 143.92, 142.25, 138.37, 136.15, 135.98 (N
CH), 130.15, 129.38, 128.98, 128.86, 128.32, 128.20, 127.64, 126.34, 123.74, 119.21, 115.03, 61.13 (CH2–O), 52.22 (CH2–C
O); anal. calcd: C33H27N9O2; C, 68.15; H, 4.68; N, 21.67; found: C, 68.32; H, 4.82; N, 21.71.
), 9.80 (s, 1H, NH–C
O), 8.28 (s, 1H, N
CH), 8.24 (s, 1H, triazole), 7.68 (d, J = 8.1 Hz, 2H, Ar), 7.48 (d, J = 7.5 Hz, 2H, Ar), 7.43 (d, J = 7.8 Hz, 2H, Ar), 7.41–7.37 (m, 2H, Ar), 7.39–7.35 (m, 2H, Ar), 7.38–7.34 (m, 3H, Ar), 7.22 (d, J = 7.4 Hz, 1H, Ar), 7.17 (d, J = 7.3 Hz, 1H, Ar), 7.14 (d, J = 9.0 Hz, 2H, Ar), 7.10 (d, J = 7.3 Hz, 1H, Ar), 5.41 (s, 2H, CH2–C
O), 5.23 (s, 2H, CH2–O), 2.23 (s, 3H, CH3); 13C NMR (126 MHz, DMSO-d6) δ 164.30 (NH–C
O), 159.14 (–N
C–N
), 156.31, 150.59, 143.90, 142.22, 136.15, 135.98 (N
CH), 135.47, 131.55, 130.39, 130.16, 129.37, 128.98, 128.32, 128.21, 127.64, 126.30, 126.02, 125.51, 124.69, 115.03, 61.12 (CH2–O), 51.92 (CH2–C
O), 17.74 (CH3); anal. calcd: C34H29N9O2; C, 68.56; H, 4.91; N, 21.16; found: C, 68.75; H, 5.04; N, 21.27.
), 10.42 (s, 1H, NH–C
O), 8.28 (s, 1H, N
CH), 8.26 (s, 1H, triazole), 7.69 (d, J = 8.3 Hz, 2H, Ar), 7.48 (d, J = 7.5 Hz, 2H, Ar), 7.44 (s, 1H, Ar), 7.43–7.41 (m, 2H, Ar), 7.39 (d, J = 8.0 Hz, 2H, Ar), 7.37–7.36 (m, 2H, Ar), 7.36–7.34 (m, 2H, Ar), 7.20 (t, J = 7.8 Hz, 2H, Ar), 7.14 (d, J = 8.2 Hz, 2H, Ar), 6.90 (d, J = 7.5 Hz, 1H, Ar), 5.35 (s, 2H, CH2–C
O), 5.23 (s, 2H, CH2–O), 2.26 (s, 3H, CH3); 13C NMR (126 MHz, DMSO-d6) δ 164.03 (NH–C
O), 159.14 (–N
C–N
), 156.31, 150.56, 143.92, 142.23, 138.28, 138.08, 136.15, 135.98 (N
CH), 130.15, 129.37, 128.97, 128.68, 128.31, 128.19, 127.64, 126.31, 124.45, 119.76, 116.40, 115.01, 61.12 (CH2–O), 52.23 (CH2–C
O), 21.11 (CH3); anal. calcd: C34H29N9O2; C, 68.56; H, 4.91; N, 21.16; found: C, 68.73; H, 5.06; N, 21.32.
), 9.74 (s, 1H, NH–C
O), 8.27 (s, 1H, N
CH), 8.25 (s, 1H, triazole), 7.68 (d, J = 8.2 Hz, 2H, Ar), 7.48 (d, J = 7.5 Hz, 2H, Ar), 7.43 (d, J = 7.3 Hz, 1H, Ar), 7.42–7.39 (m, 1H), 7.39–7.37 (m, 2H, Ar), 7.37–7.36 (m, 2H, Ar), 7.36–7.34 (m, 2H, Ar), 7.28 (d, J = 8.1 Hz, 1H, Ar), 7.13 (d, J = 8.2 Hz, 2H, Ar), 7.03 (s, 1H, Ar), 6.96 (d, J = 8.0 Hz, 1H, Ar), 5.38 (s, 2H, CH2–C
O), 5.23 (s, 2H, CH2–O), 2.23 (s, 3H, CH3), 2.18 (s, 3H, CH3); 13C NMR (126 MHz, DMSO-d6) δ 164.23 (NH–C
O), 159.13 (–N
C–N
), 158.58, 156.31, 150.60, 143.90, 142.21, 136.15, 135.98, 134.65, 132.87, 131.51, 130.89 (N
CH), 130.15, 129.37, 128.97, 128.31, 128.20, 127.63, 126.52, 126.28, 124.74, 115.02, 61.12 (CH2–O), 51.90 (CH2–C
O), 20.41 (CH3), 17.66 (CH3); anal. calcd: C35H31N9O2; C, 68.95; H, 5.13; N, 20.68; found: C, 67.08; H, 5.25; N, 20.71.
), 10.42 (s, 1H, NH–C
O), 8.28 (s, 1H, N
CH), 8.26 (s, 1H, triazole), 7.69 (d, J = 8.2 Hz, 2H, Ar), 7.49 (d, J = 8.0 Hz, 2H, Ar), 7.49–7.45 (m, 2H, Ar), 7.43 (d, J = 7.3 Hz, 1H, Ar), 7.41–7.38 (m, 1H, Ar), 7.40–7.36 (m, 2H, Ar), 7.38–7.35 (m, 2H, Ar), 7.37–7.32 (m, 2H, Ar), 7.18–7.14 (m, 2H, Ar), 7.16–7.11 (m, 2H, Ar), 5.35 (s, 2H, CH2–C
O), 5.23 (s, 2H, CH2–O), 2.54 (q, J = 7.6 Hz, 2H, CH2), 1.14 (t, J = 7.6 Hz, 3H, CH3); 13C NMR (126 MHz, DMSO-d6) δ 163.85 (NH–C
O), 159.14 (–N
C–N
), 158.62, 156.30, 150.57, 143.91, 142.22, 139.17, 136.16, 136.04, 135.98 (N
CH), 130.15, 129.37, 128.97, 128.31, 128.19, 128.03, 127.65, 126.31, 119.31, 115.02, 61.13 (CH2–O), 52.19 (CH2–C
O), 27.53 (CH2), 15.54 (CH3); anal. calcd: C35H31N9O2; C, 68.95; H, 5.13; N, 20.68; found: C, 67.04; H, 5.28; N, 20.71.
), 10.34 (s, 1H, NH–C
O), 8.27 (s, 1H, N
CH), 8.24 (s, 1H, triazole), 7.68 (d, J = 8.2 Hz, 2H, Ar), 7.50–7.49 (m, 2H, Ar), 7.49–7.47 (m, 3H, Ar), 7.44 (d, J = 7.2 Hz, 1H, Ar), 7.39–7.38 (m, 2H, Ar), 7.38–7.37 (m, 2H, Ar), 7.36–7.35 (m, 2H, Ar), 7.14 (d, J = 8.2 Hz, 2H, Ar), 6.90 (d, J = 8.9 Hz, 2H, Ar), 5.31 (s, 2H, CH2–C
O), 5.23 (s, 2H, CH2–O), 3.71 (s, 3H, OCH3); 13C NMR (126 MHz, DMSO-d6) δ 163.56 (NH–C
O), 159.14 (–N
C–N
), 156.31, 155.53, 150.61, 143.90, 143.88, 143.14, 142.20, 136.16, 135.98 (N
CH), 131.45, 130.15, 129.37, 128.97, 128.21, 127.63, 126.29, 120.76, 115.04, 113.99, 61.12 (CH2–O), 55.13 (O–CH3), 52.12 (CH2–C
O); anal. calcd: C34H29N9O3; C, 66.76; H, 4.78; N, 20.61; found: C, 66.92; H, 4.81; N, 20.76.
), 10.33 (s, 1H, NH–C
O), 8.29 (s, 1H, N
CH), 8.26 (s, 1H, triazole), 7.92 (d, J = 7.9 Hz, 1H, Ar), 7.69 (d, J = 8.2 Hz, 2H, Ar), 7.48 (d, J = 7.4 Hz, 2H, Ar), 7.43 (d, J = 7.2 Hz, 1H, Ar), 7.40 (d, J = 7.9 Hz, 2H, Ar), 7.40–7.36 (m, 1H, Ar), 7.38–7.34 (m, 2H, Ar), 7.37–7.33 (m, 2H, Ar), 7.31–7.25 (m, 1H, Ar), 7.19–7.15 (m, 2H, Ar), 7.14 (d, J = 6.8 Hz, 2H, Ar), 5.46 (s, 2H, CH2–C
O), 5.24 (s, 2H, CH2–O); 13C NMR (126 MHz, DMSO-d6) δ 164.75 (NH–C
O), 159.13 (–N
C–N
), 158.59, 156.30, 154.42, 152.47, 150.57, 143.91, 142.28, 136.15, 135.98 (N
CH), 130.15, 129.37, 128.99, 128.32, 128.20, 127.66, 126.36, 125.67, 125.61, 125.47, 125.38, 124.46, 124.44, 123.75, 115.63, 115.48, 115.04, 61.13 (CH2–O), 51.99 (CH2–C
O); anal. calcd: C33H26FN9O2; C, 66.10; H, 4.37; N, 21.02; found: C, 66.22; H, 4.41; N, 21.19.
), 10.09 (s, 1H, NH–C
O), 8.29 (s, 1H, N
CH), 8.26 (s, 1H, triazole), 7.75 (d, J = 8.1 Hz, 1H, Ar), 7.69 (d, J = 8.2 Hz, 2H, Ar), 7.51 (d, J = 7.9 Hz, 1H, Ar), 7.48 (d, J = 7.5 Hz, 2H, Ar), 7.43 (d, J = 7.1 Hz, 1H, Ar), 7.41–7.39 (m, 1H, Ar), 7.39–7.37 (m, 2H, Ar), 7.37–7.34 (m, 3H, Ar), 7.34–7.31 (m, 1H, Ar), 7.21 (t, J = 7.7 Hz, 2H, Ar), 7.13 (d, J = 8.2 Hz, 2H, Ar), 5.48 (s, 2H, CH2–C
O), 5.23 (s, 2H, CH2–O);13C NMR (126 MHz, DMSO-d6) δ 164.82 (NH–C
O), 159.12 (–N
C–N
), 158.61, 156.31, 150.59, 143.91, 142.28, 136.15, 135.98 (N
CH), 134.13, 130.15, 129.57, 129.36, 128.97, 128.30, 128.19, 127.51, 126.66, 126.35, 126.24, 125.82, 115.01, 61.12 (CH2–O), 51.94 (CH2–C
O); anal. calcd: C33H26ClN9O2; C, 64.34; H, 4.25; N, 20.46; found: C, 64.39; H, 4.36; N, 20.52.
), 10.07 (s, 1H, NH–C
O), 8.28 (s, 1H, N
CH), 8.24 (s, 1H, triazole), 7.74 (d, J = 6.5 Hz, 1H, Ar), 7.68 (d, J = 8.2 Hz, 2H, Ar), 7.52 (d, J = 6.6 Hz, 1H, Ar), 7.48 (d, J = 7.1 Hz, 2H, Ar), 7.44 (d, J = 7.3 Hz, 1H, Ar), 7.41–7.40 (m, 1H, Ar), 7.38 (d, J = 8.2 Hz, 2H, Ar), 7.36–7.35 (m, 3H, Ar), 7.33 (d, J = 7.4 Hz, 1H, Ar), 7.21 (t, J = 7.0 Hz, 1H, Ar), 7.14 (d, J = 8.3 Hz, 2H, Ar), 5.47 (s, 2H, CH2–C
O), 5.23 (s, 2H, CH2–O);13C NMR (126 MHz, DMSO-d6) δ 164.82 (NH–C
O), 159.13 (–N
C–N
), 158.57, 156.31, 150.58, 143.89, 142.27, 136.15, 135.98 (N
CH), 134.12, 130.16, 129.58, 129.37, 128.98, 128.32, 128.21, 128.14, 127.64, 127.52, 126.68, 126.35, 126.26, 125.84, 115.04, 61.11 (CH2–O), 51.92 (CH2–C
O); anal. calcd: C33H26ClN9O2; C, 64.34; H, 4.25; N, 20.46; found: C, 64.49; H, 4.32; N, 20.58.
), 10.08 (s, 1H, NH–C
O), 8.28 (s, 1H, N
CH), 8.24 (s, 1H, triazole), 7.74 (d, J = 8.0 Hz, 1H, Ar), 7.68 (d, J = 8.2 Hz, 2H, Ar), 7.52 (d, J = 7.9 Hz, 1H, Ar), 7.49–7.46 (m, 2H, Ar), 7.44 (d, J = 7.2 Hz, 1H, Ar), 7.42–7.40 (m, 1H, Ar), 7.39–7.38 (m, 2H, Ar), 7.38–7.36 (m, 2H, Ar), 7.36–7.35 (m, 2H, Ar), 7.35–7.34 (m, 1H, Ar), 7.33 (d, J = 7.8 Hz, 1H, Ar), 7.21 (t, J = 7.7 Hz, 1H, Ar), 7.13 (d, J = 8.1 Hz, 1H, Ar), 5.47 (s, 2H, CH2–C
O), 5.23 (s, 2H, CH2–O); 13C NMR (126 MHz, DMSO-d6) δ 164.82 (NH–C
O), 159.13 (–N
C–N
), 158.58, 156.32, 150.59, 143.89, 142.26, 136.14, 135.98 (N
CH), 134.12, 130.16, 129.58, 129.36, 128.98, 128.33, 128.21, 127.52, 126.68, 126.35, 125.84, 61.11 (CH2–O), 51.92 (CH2–C
O); anal. calcd: C33H26ClN9O2; C, 64.34; H, 4.25; N, 20.46; found: C, 64.41; H, 4.39; N, 20.51.
), 10.65 (s, 1H, NH–C
O), 8.29 (s, 1H, N
CH), 8.26 (s, 1H, triazole), 7.69 (d, J = 8.2 Hz, 2H, Ar), 7.56 (d, J = 8.5 Hz, 2H, Ar), 7.51 (d, J = 8.6 Hz, 2H, Ar), 7.48 (d, J = 7.3 Hz, 2H, Ar), 7.43 (d, J = 7.3 Hz, 1H, Ar), 7.38 (d, J = 7.9 Hz, 2H, Ar), 7.37–7.33 (m, 4H, Ar), 7.13 (d, J = 8.2 Hz, 2H, Ar), 5.38 (s, 2H, CH2–C
O), 5.23 (s, 2H, CH2–O); 13C NMR (126 MHz, DMSO-d6) δ 164.34 (NH–C
O), 159.12 (–N
C–N
), 158.53, 156.31, 150.56, 143.93, 142.27, 137.73, 136.15, 135.98 (N
CH), 131.70, 130.15, 129.37, 128.97, 128.30, 128.19, 127.64, 126.32, 121.16, 115.40, 115.01, 61.12 (CH2–O), 52.22 (CH2–C
O); anal. calcd: C33H26BrN9O2; C, 60.01; H, 3.97; N, 19.08; found: C, 60.16; H, 4.11; N, 19.17.
), 10.78 (s, 1H, NH–C
O), 8.28–8.24 (m, 2H, N
CH and triazole), 7.98 (d, J = 7.9 Hz, 2H, Ar), 7.73 (d, J = 8.1 Hz, 2H, Ar), 7.71–7.68 (m, 2H, Ar), 7.49–7.46 (m, 2H, Ar), 7.45–7.42 (m, 2H, Ar), 7.42–7.40 (m, 2H, Ar), 7.39–7.38 (m, 2H, Ar), 7.38–7.34 (m, 2H, Ar), 7.18–7.11 (m, 2H, Ar), 5.46 (s, 2H, CH2–C
O), 5.23 (s, 2H, CH2–O); 13C NMR (126 MHz, DMSO-d6) δ 164.90 (NH–C
O), 159.15 (–N
C–N
), 156.37, 156.30, 150.55, 143.98, 142.32, 136.12, 136.06 (N
CH), 134.13, 130.34, 130.17, 129.36, 128.99, 128.34, 128.21, 126.34, 125.84, 125.49, 125.03, 115.02, 61.11 (CH2–O), 51.97 (CH2–C
O); anal. calcd: C33H26N10O4; C, 63.25; H, 4.18; N, 22.35; found: C, 63.37; H, 4.22; N, 22.40.
), 8.86 (t, J = 5.9 Hz, 1H, NH–C
O), 8.26 (s, 1H, N
CH), 8.24 (s, 1H, triazole), 7.69 (d, J = 8.2 Hz, 2H, Ar), 7.48 (d, J = 7.4 Hz, 2H, Ar), 7.43 (d, J = 7.3 Hz, 1H, Ar), 7.42–7.38 (m, 2H, Ar), 7.40–7.36 (m, 1H, Ar), 7.38–7.34 (m, 1H, Ar), 7.36–7.33 (m, 3H, Ar), 7.32 (d, J = 7.2 Hz, 2H, Ar), 7.31–7.26 (m, 2H, Ar), 7.26 (d, J = 7.0 Hz, 1H, Ar), 7.13 (d, J = 8.1 Hz, 2H, Ar), 5.22 (s, 2H, CH2–C
O), 5.21 (s, 2H, CH2–O), 4.34 (d, J = 6.0 Hz, 2H, CH2–NH); 13C NMR (126 MHz, DMSO-d6) δ 170.26 (NH–C
O), 165.37 (–N
C–N
), 159.15, 158.59, 156.32, 150.56, 143.94, 142.17, 138.65, 136.15, 135.98 (N
CH), 130.16, 129.38, 128.98, 128.31, 128.20, 127.63, 127.36, 126.96, 126.20, 115.01, 61.13 (CH2–O), 51.62 (CH2–C
O), 42.38 (CH2–NH); anal. calcd: C34H29N9O2; C, 68.56; H, 4.91; N, 21.16; found: C, 68.72; H, 4.98; N, 21.29.
O), 5.09 (s, 2H, CH2–O), 3.32 (m, 2H, CH2–NH), 2.74 (t, J = 7.4 Hz, 2H, CH2-Ph); 13C NMR (126 MHz, DMSO-d6) δ 165.19 (NH–C
O), 156.30 (–N
C–N
), 150.58, 147.60, 143.87, 142.14, 139.12, 136.16, 135.98 (N
CH), 130.16, 129.37, 128.98, 128.58, 128.31, 128.21, 128.12, 127.64, 126.12, 126.09, 115.03, 61.11 (CH2–O), 51.62 (CH2–C
O), 40.35 (CH2–NH), 34.86 (CH2-Ph); anal. calcd: C35H31N9O2; C, 68.95; H, 5.13; N, 20.68; found: C, 69.04; H, 5.28; N, 20.71.
| Compound | n | R | IC50a (μM) |
|---|---|---|---|
| a Values are the mean ± SD. All experiments were performed at least three times. | |||
| 13a | 0 | H | 0.8 ± 0.04 |
| 13b | 0 | 2-Me | 12.5 ± 0.21 |
| 13c | 0 | 3-Me | 1.8 ± 0.02 |
| 13d | 0 | 2,4-Dimethyl | 2.5 ± 0.12 |
| 13e | 0 | 4-Et | 3.7 ± 0.13 |
| 13f | 0 | 4-OMe | 10.9 ± 0.24 |
| 13g | 0 | 2-F | 0.3 ± 0.11 |
| 13h | 0 | 2-Cl | 0.19 ± 0.05 |
| 13i | 0 | 3-Cl | 11.7 ± 0.18 |
| 13j | 0 | 4-Cl | 0.12 ± 0.02 |
| 13k | 0 | 4-Br | 15.3 ± 0.29 |
| 13l | 0 | 2-NO2 | 34.2 ± 0.57 |
| 13m | 1 | H | 0.9 ± 0.07 |
| 13n | 2 | H | 0.5 ± 0.06 |
| Acarbose | — | — | 750.0 ± 0.85 |
Energy minimization was carried out with the steepest descent algorithm for up to 50
000 steps or until the maximum force fell below 10.0 kJ mol−1. The minimized system was equilibrated in two stages: (i) under an NVT ensemble at 300 K for 500 ps using the velocity-rescaling thermostat with a coupling constant of 0.1 ps, followed by (ii) NPT equilibration at 1 bar for 1000 ps using the Berendsen barostat with a pressure coupling constant of 5.0 ps. Long-range electrostatics were treated with the Particle Mesh Ewald (PME) method, while short-range electrostatic and van der Waals interactions were handled using cut-off distances of 1.0 nm and 1.2 nm, respectively. Finally, a 20 ns production MD simulation was performed to investigate the conformational stability and dynamic behavior of the protein–ligand complexes.
1H NMR and 13C NMR interpretations of compound 13a, for instance, are schematically shown in Fig. 3.
As can be seen in the SAR diagram of N-phenylacetamide derivatives 13a–l (Fig. 4a), the first, second, and third potent compounds were 4-chloro (compound 13j), 2-chloro (compound 13h), and 2-fluoro (compound 13g), respectively. These compounds were also the most potent compounds among all the synthesized compounds 13a–n. Moreover, unsubstituted compound 13a in this series was a significant inhibitor against α-glucosidase. This compound was the fifth potent compound among the compounds 13a–n. The addition of a methyl group at the 2-position of the phenyl ring in the un-substituted derivative 13a decreased its inhibitory activity to 15.6-fold, while the same substituent at the 3-position reduced anti-α-glucosidase activity to 2.25-fold, as observed in compounds 13b and 13c, respectively. This finding indicated that the position of substituents plays an important role in anti-α-glucosidase activity. On the other hand, 4-ethyl derivative 13e was around 3-fold more potent than 4-methoxy derivative 13f. Therefore, even among derivatives with electron-donating substituents, the type of substitution is very decisive in the obtained inhibition effect. As shown in Table 1, this trend is also observed in derivatives with electron-withdrawing substituents (4-chloro derivative 13j vs. 4-bromo derivative 13k, and 2-fluoro derivative 13g and 2-chloro derivative 13h vs. 2-nitro derivative 13l). In addition to the mentioned points, comparison of the IC50 values of the 2-methyl derivative 13b with its 2,4-dimethyl analog 13d indicates that the introduction of a second methyl group significantly increased α-glucosidase inhibition potency.
Table 1 and Fig. 4b also show that the N-phenylacetamide derivative 13a, the N-benzylacetamide derivative 13m, and the N-phenethylacetamide derivative 13n are highly potent against α-glucosidase, with the most active being phenethyl derivative (compound 13n).
![]() | ||
| Fig. 5 Comparison of the template compounds B and their corresponding analogs of compounds 13 in anti-α-glucosidase assay. | ||
![]() | ||
| Fig. 6 The Lineweaver–Burk plots in the absence and presence of the different concentrations of new compound 13j (a) and acarbose (b). | ||
Docking analysis of acarbose revealed the following interactions within the α-glucosidase active site: (i) hydrogen bonds with Thr301, Gln322, Thr307, Arg312, Glu304, Ser308, and Asn241; (ii) a non-classical hydrogen bond with His239; (iii) a hydrophobic contact with His279; and (iv) unfavorable interactions with Arg312 and Thr207 (Fig. 7B).
As illustrated in Fig. 7C, the most active derivative, compound 13j, established a classical hydrogen bond along with an unfavorable interaction with Asp408. Additionally, this compound formed two π-anion interactions with Asp439 and Glu304, as well as two π–π stacking interactions with Phe300. Several non-classical hydrogen bonds were also observed between compound 13j and residues Arg439, Val305, Pro309, Arg312, and Ser308. Moreover, two hydrophobic interactions were detected with Pro309 and Arg312.
The interaction profile of the second most potent compound 13h is depicted in Fig. 7D. This compound formed two classical hydrogen bonds with residues Ser308 and His239, along with a non-classical hydrogen bond with Asn241. In addition, compound 13h established two π-anion interactions with Glu304 and Asp408 and four hydrophobic interactions with residues Pro309 (two interactions), Phe157, and Arg312.
The calculated binding energies for standard inhibitor acarbose (−4.4 kcal mol−1), compound 13j (−11.29 kcal mol−1), and compound 13h (−9.1 kcal mol−1) indicated that both newly synthesized compounds exhibited substantially stronger binding affinities compared to acarbose. Moreover, the most potent compound 13j demonstrated the more favorable binding energy than the second potent compound 13h, which was consistent with the results obtained from the in vitro enzymatic inhibition assays.
Two runds of MD simulations were carried out. Initially, each protein–ligand complex underwent a 10 ns simulation, during which both acarbose and compound 13j maintained stable interactions within the α-glucosidase active site. To obtain a more detailed picture of their dynamic behavior, the simulations were extended for an additional 10 ns. The extended trajectories confirmed that both ligands continued to exhibit stable binding throughout the entire run. The generated trajectories were subsequently analyzed with several computational approaches. Root-mean-square deviation (RMSD) and radius of gyration (Rg) values were calculated for all sampled conformations to track structural fluctuations and assess overall stability of the complexes over time. In addition, the root-mean-square fluctuation (RMSF) of backbone atoms was examined to evaluate residue-specific flexibility during the simulation. The outcomes of these analyses are summarized in Fig. 8 and 9. As shown in Fig. 8, the RMSD values of α-glucosidase remained steady, staying below 0.30 nm across the entire simulation. The mean RMSD values of α-glucosidase in complex with either acarbose or compound 13j were 0.19 nm and 0.18 nm, respectively. Indicating negligible structural deviations and confirming complex stability. Similarly, RMSD values for acarbose and compound 13j within the binding pocket were consistently below 0.25 nm, with no notable fluctuations during the simulation (Fig. 8). Their corresponding mean RMSD values were 0.12 nm for acarbose and 0.16 nm for compound 13j. Taken together, these findings demonstrate that both ligands retained stable binding conformations within the α-glucosidase active site throughout the MD simulations.
The structural compactness of α-glucosidase during the simulations was evaluated through the calculation of its radius of gyration (Rg), as depicted in Fig. 8. Across the trajectory, the Rg values of the enzyme bound to either acarbose or compound 13j consistently fell within a narrow window of 2.39–2.55 nm. The absence of large variations or long-term shifts indicates that the tertiary fold of the protein remained intact. The average Rg values obtained were 2.50 nm for the acarbose complex and 2.48 nm for the compound 13j complex, confirming that ligand association did not trigger noticeable structural expansion or contraction.
RMSF analysis of Cα atoms was performed to probe residue-level flexibility (Fig. 9). The RMSF profiles of α-glucosidase complexed with either acarbose or compound 13j displayed very similar patterns. Given that α-glucosidase is a large multidomain enzyme with several distinct structural and functional regions, heterogeneity in residue mobility was evident. Special attention was paid to residues within and near the active site. Residues that formed non-bonded interactions with the ligands mainly those belonging to domains A and B showed low fluctuations, reflecting structural rigidity and the potential to maintain steady ligand contacts. Conversely, flexible loop elements, such as the B domain loop and the active-site lid, exhibited greater mobility. The reduced flexibility of residues at the binding site underlines their importance in preserving stable interactions with both ligands throughout the simulations. In addition, RMSF values of the heavy atoms of acarbose and compound 13j were determined and are presented in Fig. 9. Both ligands showed very limited fluctuations, with all values remaining below 0.25 nm. The least mobile atoms in both ligands were those embedded in ring systems, whose intrinsic rigidity restricts flexibility. This rigidity likely enhances the ability of ring atoms to form stable non-bonded interactions with residues in the binding pocket. At the same time, the establishment of these strong contacts further constrains the motion of ring atoms, reinforcing the reciprocal stability between the ligands and the enzyme's active site.
| Pharmacokinetics | Compound 13j | Acarbose | |
|---|---|---|---|
| Druglikeness | Rule of five | Violated | Violated |
| Veber rule | Violated | Violated | |
| ADME | GI absorption | Low | Low |
| BBB permeant | No | No | |
| P-gp substrate | No | Yes | |
| CYP1A2 inhibitor | Yes | No | |
| CYP2C19 inhibitor | Yes | No | |
| CYP2C9 inhibitor | Yes | No | |
| CYP2D6 inhibitor | No | No | |
| CYP3A4 inhibitor | No | No | |
| Total clearance | −0.127 | 0.702 | |
| OCT2 substrate | No | No | |
| Toxicity | Ames_test | Non-mutagen | Mutagen |
| Carcino_Mouse | Positive | Positive | |
| Carcino_Rat | Negative | Negative | |
| hERG_inhibition | Low risk | Ambiguous | |
Druglikeness calculations for compound 13j and acarbose showed that neither of them complies with the Lipinski rule (rule of five) or the Veber rule, which are two important rules for predicting druglikeness.
Absorption predictions indicated that both compound 13j and acarbose have not gastrointestinal absorption (GI) and permeability to blood–brain barrier (BBB). Moreover, distribution predictions showed that compound 13j is not a substrate for P-glycoprotein (P-gp, an efflux pump) whereas acarbos is a P-gp substrate.37 In term of metabolism, compound 13j properly inhibits CYP1A2, CYP2C19, and CYP2C9 isoforms of cytochrome P450 (CYP), a major drug-metabolizing enzyme, and dose not inhibit CYP2D6 and CYP3A4 isoforms but acarbose does not inhibit any of these enzymes. According to Table 2, in terms of excretion, total clearance of compound 13j is lower than acarbose. Moreover, compound 13j and acarbose are not substrates for organic cation transporter 2 (OCT2).
Toxicity prediction of acarbose and compound 13j indicated that compound 13j is non-mutagenic, whereas computational analysis suggested a potential mutagenic alert for acarbose. Nevertheless, it should be emphasized that this result is obtained from in silico predictions and may not reflect the well-documented clinical safety of acarbose reported in the literature. In terms of carcinogenicity, compounds 13j and acarbose show similar profile. Finally, the potential of cardiotoxicity (hERG inhibition) of compound 13j is low, whereas this type of toxicity for acarbose remains ambiguous.
Bioavailability radars of the new compound 13j and the standard inhibitor (acarbose) are shown in Fig. 10. In these radars, the pink area demonstrated a suitable physicochemical space for oral bioavailability. According to the obtained bioavailability radars, compound 13j does not fall within the pink area whereas acarbose is positioned in this area only marginally.
Using the admetSAR server, the ADMET properties of compound 13j were also predicted and compared with those of the acarbose. The obtained data of the admetSAR were illustrated in Fig. 11. According to this figure, compound 13j, shows a higher presence within the drug-likeness acceptable region compared to acarbose. This compound has favorable pharmacokinetic characteristics, including improved intestinal absorption and oral bioavailability in comparison to acarbose, while maintaining an acceptable toxicity profile.
| This journal is © The Royal Society of Chemistry 2025 |