Startseite Furanone-functionalized benzothiazole derivatives: synthesis, in vitro cytotoxicity, ADME, and molecular docking studies
Artikel Öffentlich zugänglich

Furanone-functionalized benzothiazole derivatives: synthesis, in vitro cytotoxicity, ADME, and molecular docking studies

  • Asif Husain , Silky Bedi , Shazia Parveen EMAIL logo , Shah Alam Khan , Aftab Ahmad , Md Azhar Iqbal , Aasif Farooq und Anwar Ahmed
Veröffentlicht/Copyright: 26. November 2021
Veröffentlichen auch Sie bei De Gruyter Brill

Abstract

In the present study, a novel series of new furanone-based benzothiazole derivatives (4a-j) were synthesized from 4-(benzo[d]thiazol-2-yl)-4-oxobutanoic acid (3) as potential anticancer agents. In vitro cytotoxicity against three human cancer cell lines (A549, MCF7, and DUI45) revealed substantial activity. Di-substituted compound, 4i emerged as a promising anticancer compound which showed IC50 values of 7.2 ± 0.5, 6.6 ± 1.4, and 7.3 ± 0.1 µM against A549, MCF7, and DUI45 cell lines, respectively. Four compounds 4c, 4e, 4f, and 4i evaluated for their acute toxicity were found to be non-toxic on the two vital organs (liver and heart). Further, these compounds were found to be more efficient and less hepatotoxic in comparison to standard drug doxorubicin. Molecular docking studies carried out with VEGFR-2 revealed compounds 4a and 4i as potential VEGFR-2 kinase inhibitors. In silico ADME evaluation was carried out to estimate and predict drug-likeness. Compound 4i demonstrated the best ADME parameters. Based on the results of docking analyses, ADME, and in vitro cytotoxicity, compound 4i is identified as the lead compound for further development of anticancer agents.

1 Introduction

Cancer or malignant tumors have been always endangering human life. The morbidity and mortality associated with cancer are on rise. It is considered as one of the foremost causes of human mortality worldwide [1, 2]. According to the WHO global cancer profile, 10 million deaths alone have occurred due to cancer [3]. Cancer is the unrestrained proliferation and abnormal metastasis of the body’s specific cells. The major drawback of existing anticancer drugs is their inadequate selectivity towards abnormal cancer cells, leading to severe toxic side effects. Consequently, this warrants developing more effective and selective anticancer agents possessing a suitable pharmacokinetic profile.

The transformation of a localized benign tumor to a malignant one involves the process of angiogenesis [4]. In this process, infiltration of new blood vessels from the tumor masses gives access to oxygen and other nutrients to boost tumor growth and metastasis [5, 6]. Angiogenesis exhibits a fundamental role in new blood vessels formation from pre-existing ones, leading to tumor growth [7]. Various cancer tissues could generate VEGF (vascular endothelial growth factor) resulting in the initiation of angiogenesis from neighboring blood vessels [8]. VEGFR-2 is a tyrosine kinase receptor that is expressed in endothelial cells [9]. VEGFR-2, plays an important role in anti-angiogenesis and is an effective target for inhibiting tumor cell proliferation and metastasis [10]. VEGFR-2 plays vital roles in the development of organs and differentiation during embryogenesis along with the functions associated with wound healing and reproduction. Consequently, VEGFR-2/VEGF signaling pathway inhibition is believed to be the most significant and imperative strategies in the advancement of cancer chemotherapy. Presently, several of the FDA approved VEGFR-2 inhibitors, specifically sorafenib, sunitinib, axitinib, vandetanib, pazopanib and regorafenib, are in clinical use for cancer chemotherapy (Figure 1) [11, 12]. Several other effective VEGFR-2 inhibitors have been developed and attained clinical success in the treatment of cancer [13, 14].

Figure 1: 
Approved anticancer compounds.
Figure 1:

Approved anticancer compounds.

Nitrogen containing heterocyclic frameworks demonstrate remarkable biological activities in humans. Benzothiazoles are sulfur containing heterocyclic compounds which exhibit unique chemical and biological properties (Figure 1) [15, 16]. They are of great biological significance since they have engrossed significant attention as promising and effective anticancer agents [17], [18], [19], [20], [21], specifically targeting the VEGFR‐2 protein [15, 22], [23], [24], [25], [26], [27]. Benzothiazoles are known to act as competitive inhibitors at the ATP‐binding site of tyrosine kinases. Benzothiazoles derivatives have been reported to exhibit significant and predominant biological and pharmacological activities against diverse tumor types and cancer cell lines viz., HeLa (human cervical cancer cell line), SW480 (human colon adenocarcinoma cell line), HepG2 (human liver carcinoma cells), mammary and ovarian tumor cell lines, colon, non-small cell lung cancer (NSCLC), breast cancer cell lines, HCC (hepatocellular carcinoma), etc. [28].

Besides, furanone, an α,β-unsaturated lactone substructure is found in various biologically active compounds of natural origin [29], [30], [31], [32] and synthetic pharmaceuticals [33] including anticancer agents [34, 35]. Therefore, the design and synthesis of various furanone derived compounds and their applications in cancer treatment have drawn considerable attention [36, 37].

Because of the stated research outcomes and in continuation of our efforts to design new anticancer agents, the objective of this work was to find new anticancer agents bearing similar crucial pharmacophoric properties of the reported and clinically used agents [38], [39], [40], [41]. Herein, we report the synthesis of furanone-coupled benzothiazole derivatives and their assessment of biological activities against three cancer cell lines. In addition, we have carried out cardio and hepatotoxicity studies. Furthermore, molecular docking analyses were also performed to get an insight into their binding modes and binding energies onto the VEGFR-2 enzyme.

2 Experimental section

2.1 Synthesis

2.1.1 Synthesis of 4-(benzo[d]thiazol-2-yl)-4-oxobutanoic acid (3)

To a mixture of o-aminothiophenol, (1) (6.5 mmol) and α-ketoglutaric acid (2) (7 mmol), polyphosphoric acid (10 g) was added and the resulting mixture was then heated along with vigorous stirring at 220 °C for 4 h (Scheme 1). The reaction mixture was then cooled and poured in 10% Na2CO3 solution to yield a suspension which was stirred until the evolution of the gas stopped and then filtered. The solid precipitate was collected and washed with 350 mL water. Recrystallization was done from MeOH–H2O to get the pure product (R f 0.71; toluene:ethyl acetate:formic acid = 5:4:1); nature: dark brown amorphous powder; Yield: 80%; m.p.: 270 °C. Elemental analysis [C11H9NO3S] (%); Calc.: C, 56.16; H, 3.86; N, 5.95; found: C, 56.01; H, 3.37; N, 5.68. IR (cm−1): 3021 (OH-str); 2962 (C═C Ar, str); 2881 (C–H str); 1721 (carboxylic acid C═O str); 1683 (C═O str); 1498 (C═N str); 1390, 1322 (C–O str). 1H NMR (DMSO-d 6 ): δ 2.28–2.51 (t, 2H, CH2), 2.84–2.90 (t, 2H, CH2), 6.38–7.51 (m, 3H, benzothiazole), 7.91 (s, 1H, benzothiazole), 12.47 (br s, 1H, –COOH). 13C NMR (DMSO-d 6 ): δ 188.2 (COOH); 168.2 (C═O); 160.4 (C-thiazole); 155.6 (1C, arylidine C); 137.6 (1C, arylidine C); 121–125 (4C, arylidine C); 30.21 (2C, CH2); ESI-MS: m/z: 235 (M+).

Scheme 1: 
Synthetic route to title compounds 4a–j. (Ring systems-benzothiazole and furanone).
Scheme 1:

Synthetic route to title compounds 4a–j. (Ring systems-benzothiazole and furanone).

2.1.2 Synthesis of compounds 4a–j from 4-(benzo[d]thiazol-2-yl)-4-oxobutanoic acid (3)

4-(Benzo[d]thiazol-2-yl)-4-oxobutanoic acid (3) (3 mmol) was reacted with appropriate aryl aldehyde (equimolar, 3 mmol) in acetic anhydride (Ac2O; 5–10 mL) along with 3–4 drops of triethylamine (TEA). The resulting solution was then refluxed for 3–4 h under anhydrous conditions and the progress of the reaction was continuously monitored by TLC. Upon completion of the reaction, the reaction mixture was poured into crushed ice in small portions with continuous stirring. A colored solid precipitate was obtained which was then filtered, washed with water, dried, and recrystallized from methanol to yield TLC pure compounds (4a–j) (Scheme 1).

2.1.3 Synthesis of (E)-5-(benzo[d]thiazol-2-yl)-3-(4-hydroxybenzylidene)furan-2(3H)-one (4a)

4-Hydroxybenzaldehyde was treated with 4-(benzo[d]thiazol-2-yl)-4-oxobutanoic acid (3) along with Ac2O and TEA to yield compound 4a following the general protocol. Elemental analysis [C18H11NO3S] (%); Calc.: C, 67.28; H, 3.45; N, 4.36; found: C, 67.12; H, 3.37; N, 4.08. IR (cm−1): 3732 (OH-str); 2970 (C═C Ar, str); 1685 (C═O str); 1500 (C═N str); 1390, 1328 (C–O–C str). 1H NMR (DMSO-d 6 ): δ 7.21–7.57 (m, 6H, furanone H + 4H of benzothiazole + olefinic H), 8.51 (br, s, 4H, arylidine ring), 9.24 (s, 1H, OH). 13C NMR (DMSO-d 6 ): δ 167.3 (C═O), 160.7 (C-thiazole), 156.2 (1C, COH) 147.6 (C-5 furanone), 130.2 (2C, –CH of benzylidine + C-3 furanone), 115–152 (11C, arylidine C); 105.3 (C-4 furanone); ESI-MS: 318 (M-3).

2.1.4 Synthesis of (E)-5-(benzo[d]thiazol-2-yl)-3-(4-(dimethylamino)benzylidene)furan-2(3H)-one (4b)

4-Dimethylaminobenzaldehyde was treated with 4-(benzo[d]thiazol-2-yl)-4-oxobutanoic acid (3) along with Ac2O and TEA to yield compound 4b following the general protocol. Elemental analysis [C20H16N2O2S] (%): Calc.: C, 68.94; H, 4.63; N, 8.04; found: C, 68.82; H, 4.53; N, 7.98. IR (cm−1): 2972 (C═C Ar, str); 1683 (C═O str); 1581 (C═N str); 1369; 1312 (C–O–C str). 1H NMR (DMSO-d 6 ): δ 2.76 (s, 6H, N-(CH 3)2), 6.62 (s, 1H, CH, furanone), 7.09 (s, 1H, olefinic H), 7.12–7.15 (m, 1H, arylidine ring), 7.34–7.37 (d, 1H, arylidine ring), 7.93–8.05 (m, 2H, arylidine ring), 7.18–7.51 (m, 4H, benzothiazole). 13C NMR (DMSO-d 6 ): δ 167.2 (C═O), 160.8 (C-thiazole), 150.9 (1C, C-(NCH3)2), 146.2 (C-5 furanone), 131.4 (2C, –CH of benzylidine + C-3 furanone), 115–153 (11C, arylidine C), 105.3 (C-4 furanone), 41.2 (2C, –CH3); ESI-MS: m/z: 348 (M+).

2.1.5 Synthesis of (E)-5-(benzo[d]thiazol-2-yl)-3-(3-nitrobenzylidene)furan-2(3H)-one (4c)

m-Nitrobenzaldehyde was treated with 4-(benzo[d]thiazol-2-yl)-4-oxobutanoic acid (3) along with Ac2O and TEA to yield compound 4c following the general protocol. Elemental analysis [C18H10N2O4S] (%): Calc.: C, 61.71; H, 2.88; N, 8.00; found: C, 61.68; H, 2.87; N, 7.92. IR (cm−1): 2967 (C═C Ar, str); 1681 (C═O str); 1568 (C═N str); 1389, 1325 (C–O–C str); 670 (C═S Ar, Str). 1H NMR (DMSO-d 6 ): δ 7.20–7.50 (m, 6H, furanone H + 4H of benzothiazole + olefinic H); 8.55 (br s, 4H of arylidine ring). 13C NMR (DMSO-d 6 ): δ 167.3 (C═O), 161.1 (C-thiazole), 147.6 (2C, C-5 furanone + C–NO2), 130.5 (2C, –CH of benzylidine + C-3 furanone), 115–153 (11C, arylidine C), 105.3 (C-4 furanone); ESI-MS: m/z: 350 (M+).

2.1.6 Synthesis of (E)-5-(benzo[d]thiazol-2-yl)-3-(2-chlorobenzylidene)furan-2(3H)-one (4d)

2-Chlorobenzaldehyde was treated with 4-(benzo[d]thiazol-2-yl)-4-oxobutanoic acid (3) along with Ac2O and TEA to yield compound 4d following the general protocol. Elemental analysis [C18H10ClNO2S] (%): Calc.: C, 63.62; H, 2.97; N, 4.12; found: C, 63.43; H, 2.92; N, 4.08. IR (cm−1): 2973 (C═C Ar, str); 1682 (C═O str); 1597 (C═N str); 1367, 1303 (C–O–C str); 635 (C═S Ar, Str); 765 (Cl, Ar). 1H NMR (DMSO-d 6 ): δ 7.3 (s, 1H, CH, furanone, 6.4–6.9 (s, 1H, olefinic H), 7.16–7.20 (m, 1H, arylidine ring), 7.76 (d, 1H, arylidine ring), 7.32–7.38 (m, 2H, arylidine ring), 7.03–7.84 (m, 4H, benzothiazole). 13C NMR (DMSO-d 6 ): δ 166.8 (C═O), 160.3 (C-thiazole), 146.9 (2C, C-5 furanone + C–Cl), 131.2 (2C, –CH of benzylidine + C-3 furanone), 115–152 (11C, arylidine C), 105.2 (C-4 furanone); ESI-MS: m/z: 340 (M+).

2.1.7 Synthesis of (E)-5-(benzo[d]thiazol-2-yl)-3-(3-chlorobenzylidene)furan-2(3H)-one (4e)

3-Chlorobenzaldehyde was treated with 4-(benzo[d]thiazol-2-yl)-4-oxobutanoic acid (3) along with Ac2O and TEA to yield compound 4e following the general protocol. Elemental analysis [C18H10ClNO2S] (%): Calc.: C, 63.62; H, 2.97; N, 4.12; found: C, 63.57; H, 2.90; N, 4.06. IR (cm−1): 2980 (C═C Ar, str); 1682 (C═O str); 1578 (C═N str); 1361, 1315 (C–O–C str); 680 (C═S Ar, Str); 725 (Cl Ar). 1H NMR (DMSO-d 6 ): δ 7.2 (s, 1H, CH, furanone), 6.4–6.9 (s, 1H, olefinic H), 7.15–7.19 (m, 1H, arylidine ring), 7.79 (d, 1H, arylidine ring), 7.32–7.38 (m, 2H, arylidine ring), 7.82–7.84 (m, 2H, benzothiazole), 7.01–7.79 (m, 2H, benzothiazole). 13C NMR (DMSO-d 6 ): δ 167.8 (C═O), 160.2 (C-thiazole), 147.2 (2C, C-5 furanone + C–Cl), 130.1 (2C, –CH of benzylidine + C-3 furanone), 115–153 (11C, arylidine C), 105.3 (C-4 furanone); ESI-MS: m/z: 340 (M+).

2.1.8 Synthesis of (E)-5-(benzo[d]thiazol-2-yl)-3-benzylidenefuran-2(3H)-one (4f)

Benzaldehyde was treated with 4-(benzo[d]thiazol-2-yl)-4-oxobutanoic acid (3) along with Ac2O and TEA to yield compound 4f following the general protocol. Elemental analysis [C18H11NO2S] (%): Calc.: C, 70.80; H, 3.63; N, 4.59; found: C, 70.74; H, 3.51; N, 4.56. IR (cm−1): 2974 (C═C Ar, str); 1665 (C═O str); 1558 (C═N str); 1346, 1309 (C–O–C str). 1H NMR (DMSO-d 6 ): δ 6.91 (s, 1H, CH-furanone), 7.06 (s, 1H, olefinic H), 7.38 (m, 2H, arylidine ring), 7.42 (d, 1H, arylidine ring), 7.47–7.50 (m, 2H, arylidine ring), 7.02–7.26 (m, 4H, benzothiazole). 13C NMR (DMSO-d 6 ): δ 167.3 (C═O), 160.5 (C-thiazole), 147.5 (C-5 furanone), 130.5 (2C, –CH of benzylidine + C-3 furanone), 115–153 (12C, arylidine C), 105.3 (C-4 furanone); ESI-MS: m/z: 305 (M+).

2.1.9 Synthesis of (E)-5-(benzo[d]thiazol-2-yl)-3-(3-bromobenzylidene)furan-2(3H)-one (4g)

3-Bromobenzaldehyde was treated with 4-(benzo[d]thiazol-2-yl)-4-oxobutanoic acid (3) along with Ac2O and TEA to yield compound 4g following the general protocol. Elemental analysis [C18H10BrNO2S] (%): Calc.: C, 56.26; H, 2.62; N, 3.65; found: C, 56.19; H, 2.58; N, 3.54. IR (cm−1): 2976 (C═C Ar, str); 1681 (C═O str); 1574 (C═N str); 1372, 1311 (C–O–C str); 530 (Br, Ar). 1H NMR (DMSO-d 6 ): δ 6.9 (s, 1H, CH, furanone), 6.4–6.9 (s, 1H, olefinic H), 7.18–7.2 (m, 1H, arylidine ring), 7.8 (d, 1H, arylidine ring), 7.32–7.38 (m, 2H, arylidine ring), 7.71–7.82 (m, 4H, benzothiazole). 13C NMR (DMSO-d 6 ): δ 167.8 (C═O), 160.2 (C-thiazole), 147.2 (C-5 furanone), 136.6 (2C, –CH of benzylidine + C-3 furanone), 121–153 (12C, 11C arylidine C + 1C –C–Br), 105.3 (C-4 furanone); ESI-MS: m/z: 384 (M+).

2.1.10 Synthesis of (E)-5-(benzo[d]thiazol-2-yl)-3-(4-methoxybenzylidene)furan-2(3H)-one (4h)

Anisaldehyde was treated with and 4-(benzo[d]thiazol-2-yl)-4-oxobutanoic acid (3) along with Ac2O and TEA to yield compound 4h following the general protocol. Elemental analysis [C19H13NO3S] (%): Calc.: C, 68.04; H, 3.91; N, 4.18; found: C, 67.86; H, 3.82; N, 4.13. IR (cm−1): 2974 (C═C Ar, str); 1665 (C═O str); 1558 (C═N str); 1346, 1309 (C–O–C str). 1H NMR (DMSO-d 6 ): δ 3.97 (s, 3H, OCH3), 7.20–7.50 (m, 6H, furanone H + 4H of benzothiazole + olefinic H), 8.46 (br s, 4H of arylidine ring). 13C NMR (DMSO-d 6 ): δ 167.6 (C═O), 161.7 (C-thiazole), 159.8 (C–OCH3), 147.2 (C-5 furanone), 130.5 (2C, –CH of benzylidine + C-3 furanone), 114–153 (12C, arylidine C), 105.2 (C-4 furanone), 55.8 (CH3); ESI-MS: m/z: 335 (M+).

2.1.11 Synthesis of (E)-5-(benzo[d]thiazol-2-yl)-3-(4-hydroxy-3-methoxybenzylidene)furan-2(3H)-one (4i)

Vanillin was treated with 4-(benzo[d]thiazol-2-yl)-4-oxobutanoic acid (3) along with Ac2O and TEA to yield compound 4i following the general protocol. Elemental analysis [C19H13NO4S] (%): Calc.: C, 64.95; H, 3.73; N, 3.99; found: C, 64.87; H, 3.71; N, 3.87. IR (cm−1): 3650 (OH, Ar, Str); 2969 (C═C Ar, str); 1663 (C═O str); 1560 (C═N str); 1333, 1302 (C–O–C str). 1H NMR (DMSO-d 6 ): δ 3.83 (s, 3H, OCH3), 5.18 (s, OH), 6.22 (s, 1H, CH, furanone), 7.12–8.18 (m, 9H, 4H of benzothiazole + olefinic H + 4H arylidine ring), 8.55 (br s, 4H of arylidine ring); 13C NMR (DMSO-d 6 ): δ 167.2 (C═O), 160.2 (C-thiazole), 149.1 (C–OCH3), 147.2 (2C, C-5 furanone + C–OH), 130.5 (2C, –CH of benzylidine + C-3 furanone), 111–153 (10C, arylidine C), 105.2 (C-4 furanone), 56.3 (CH3 ESI-MS: 353 (M + 2).

2.1.12 Synthesis of (E)-5-(benzo[d]thiazol-2-yl)-3-(3,4,5-trimethoxybenzylidene)furan-2(3H)-one (4j)

3,4,5-Trimethoxybenzaldehyde was treated with4-(benzo[d]thiazol-2-yl)-4-oxobutanoic acid (3) along with Ac2O and TEA to yield compound 4j following the general protocol. Elemental analysis [C21H17NO5S] (%): Calc.: C, 63.79; H, 4.33; N, 3.54; found: C, 63.71; H, 4.31; N, 3.46. IR (cm−1): 2961 (C═C Ar, str); 1684 (C═O str); 1566 (C═N str); 1387, 1323 (C–O–C str). 1H NMR (DMSO-d 6 ): δ 3.97 (s, 9H, 3 × OCH3), 7.32–7.51 (m, 6H, furanone H + 4H of benzothiazole + olefinic H), 8.62 (br s, 2H of arylidine ring). 13C NMR (DMSO-d 6 ): δ 167.4 (C═O), 160.5 (C-thiazole), 138.4–152.8 (3C, C–OCH3), 147.6 (C-5 furanone), 130.5–139.2 (2C, –CH of benzylidine + C-3 furanone), 103–153 (9C, arylidine C), 105.2 (C-4 furanone), 56.8 (3C, CH3); ESI-MS: m/z: 395 (M+).

3 Results and discussion

3.1 Synthesis

Ten new benzothiazoles coupled with furanones were synthesized via multi-step reactions from 4-(benzo[d]thiazol-2-yl)-4-oxobutanoic acid (3) as the starting material (Scheme 1). Compound 3 was obtained by condensing o-aminothiophenol (1) with α-ketoglutaric acid (2). (E)-5-(benzo[d]thiazol-2-yl)-3-arylidene furan-2(3H)-one) (4a–j) were synthesized by treating compound 3 with different substituted aryl aldehydes in Ac2O along with the catalytic amount of triethylamine. Physical and analytical data of the synthesized compounds 4a–j are given in Table 1.

Table 1:

Physical and analytical data of the synthesized compounds 4a–j.

Compounds Molecular formula Molecular weight Melting point (°C) Reaction time (h) R f valuea Color Yield (%)
4a C18H11NO3S 321.35 124 3 0.71 Dark brown 76
4b C20H16N2O2S 348.40 152 4 065 Light brown 69
4c C18H10N2O4S 350.35 161 4 0.70 Light brown 52
4d C18H10NO2SCl 339.80 118–120 3 0.76 Brown 76
4e C18H10NO2SCl 339.80 147–149 3 0.67 Dark brown 60
4f C18H10NO2S 305.35 160–162 3.5 0.78 Yellow–brown 65
4g C18H10NO2SBr 384.20 123 3 0.66 Dark pink 60
4h C19H13NO3S 335.38 112–114 4 0.80 Light brown 53
4i C19H13NO4S 351.38 128 4 0.77 Brown 53
4j C21H17NO5S 395.43 114 4 0.72 Dark brown 63
  1. aSolvent system used for TLC of compounds 4a–j, T:E:F = 5:4:1.

3.2 In vitro cytotoxicity

The IC50 value of seven of the synthesized compounds (4a, 4c, 4e, 4f, 4h, 4i, and 4j) was estimated against the three cancer cell lines. The results of in vitro cytotoxicity against three cancer cell lines indicated that the di-substituted (3-methoxy, 4-hydroxy) compound (4i) demonstrated excellent activity against A549, MCF7, and DUI45 with IC50 values of 7.2 ± 0.5 µM, 6.6 ± 1.4 µM, and 7.3 ± 0.1 µM, respectively. Also, the 3-nitro derivative (4c) exhibited promising activity against A549, MCF7, and DUI45 having IC50 values of 9.6 ± 0.1 µM, 7.3 ± 0.2 µM, and 8.7 ± 0.2 µM, respectively. Furthermore, 3-chloro (4e) and unsubstituted compound (4f), also revealed good activity against all three cell lines. Doxorubicin was used as a standard drug, and exhibited IC50 value 0.9 ± 0.2 µM, 0.9 ± 0.1 µM, and 0.8 ± 0.3 µM against A549, MCF7, and DUI45, respectively (Table 2). Molecular docking analyses of the synthesized compounds were in corroboration with the obtained results.

Table 2:

IC50 values of the compounds.

Compounds IC50 values (µM ± SD)
Cell lines
A549 MCF7 DU145
4a 18.3 ± 0.4 16.7 ± 0.8 19.2 ± 0.6
4c 9.6 ± 0.1 7.3 ± 0.2 8.7 ± 0.2
4e 10.4 ± 0.4 11.1 ± 0.4 10.7 ± 0.2
4f 11.3 ± 0.5 12.2 ± 1.4 14.7 ± 0.3
4h 19.3 ± 0.4 18.7 ± 0.8 19.2 ± 0.6
4i 7.2 ± 0.5 6.6 ± 1.4 7.3 ± 0.1
4j 13.8 ± 0.6 12.5 ± 0.2 17.7 ± 0.2
Doxorubicin 0.9 ± 0.2 0.9 ± 0.1 0.8 ± 0.3
  1. A549, human lung carcinoma cell line; MCF7, human breast carcinoma cell line; DU145, human prostate carcinoma cell line.

3.3 Structure-activity relationship

The following structure-activity relationship (SAR) could be drawn after analyzing the results of in vitro cytotoxicity of the synthesized compounds. All the compounds displayed substantial in vitro cytotoxicity (IC50 of 7.2–19.3, 6.6–18.7, 7.3–19.2 µM) against three cancer cell lines (A549, MCF7, and DUI45). Compounds 4e and 4f, exhibited good activity against all three cancer cells. Substitution of the chloro (Cl) group at position-3 makes the compound more effective against the human breast (MCF7) and prostate cancer (DUI45). The presence of halogen in the arylidine ring showed improvement in activity as compared to other substitutions in the arylidine ring. Compound 4c, substituted with m-nitro shows better activity against all three cell lines. Compound 4i, substituted with a methoxy (–OCH3) group at the m-position and with a hydroxyl (–OH) group at the p-position shows excellent activity. From the IC50 values, it may be inferred that substitution at the m-position in the benzaldehyde ring is necessary for better activity. The presence of an electron-donating methoxy (–OCH3) group at the m-position gives a better result than an electron-withdrawing group. Further substitution at the p-position in the same ring with an -OH group (electron-donating group) improves the activity (Figure 2).

Figure 2: 
Structure-activity relationship (SAR).
Figure 2:

Structure-activity relationship (SAR).

3.4 Acute toxicity

The results indicated no mortality in rats when the potent compounds 4c, 4e, 4f and 4i were administered in a dose of 500 mg/kg body weight. Consequently, the lethal dose (LD50) of the tested compounds is >500 mg/kg body weight. Hence, the tested compounds are classified as non-toxic orally LD50 (>500–<2000 mg/kg) based on the recommended guidelines by Organization for Economic Co-operation and Development (OECD).

3.5 Cardiomyopathy and hepatotoxicity

Cardiotoxicity in patients is a severe side effect of anticancer drugs [42]. Cardiotoxicity is believed to be instigated by the tyrosine kinase inhibitors viz., imatinib mesylate, dasatinib, nilotinib, sunitinib, sorafenib, and lapatinib (excluding gefitinib and erlotinib) [43]. While such drugs are linked with adverse cardiac events {viz., LV dysfunction (LVD), HF, cardiac ischemia, and myocardial infarction} are still approved owing to the availability of limited treatment opportunities [44, 45].

Hence, 4f and 4i being the most potent compounds were additionally tested for their toxicity in rat (heart and liver) in comparison to doxorubicin standard drug. Figure 3 shows photomicrographs of the normal architecture of cells of the liver (i) treated with control, (ii) effects of doxorubicin on liver cells, and (iii) effect of compound 4i on liver cells. Figure 4 illustrates the slides of the heart treated with compounds 4f and 4i revealed normal and well-maintained structures of myofibril in the subendothelial zone. No substantial difference in the structures of cardiac fiber was observed in comparison to the normal control group. However, the group treated with 4f and 4i substantially altered the structures of muscle fiber in comparison to the group treated with doxorubicin (Figure 4). Doxorubicin control group was found to induce cardiomyopathy, which was confirmed by a loss of cardiac fiber, vacuolation, and inflammation in left ventricle tissue.

Figure 3: 
Photomicrographs of the normal architecture of cells of the liver (i) treated with control, (ii) effects of doxorubicin on liver cells, and (iii) effect of compound 4i on liver cells. Magnification 200×.
Figure 3:

Photomicrographs of the normal architecture of cells of the liver (i) treated with control, (ii) effects of doxorubicin on liver cells, and (iii) effect of compound 4i on liver cells. Magnification 200×.

Figure 4: 
A: Effects of compound 4i showing the normal architecture of cardiac fiber. B: Effects of compound 4f showing the normal architecture of cardiac fiber. C: Photomicrograph showing effects of normal control on cardiac fiber (well-maintained architecture of myofibrils). D: Effects of doxorubicin showing cardiomyopathy shown by loss of cardiac fiber, vacuolation, and inflammation in left ventricle tissue. Magnification 200×.
Figure 4:

A: Effects of compound 4i showing the normal architecture of cardiac fiber. B: Effects of compound 4f showing the normal architecture of cardiac fiber. C: Photomicrograph showing effects of normal control on cardiac fiber (well-maintained architecture of myofibrils). D: Effects of doxorubicin showing cardiomyopathy shown by loss of cardiac fiber, vacuolation, and inflammation in left ventricle tissue. Magnification 200×.

Tyrosine kinase inhibitors are generally associated with induction of liver injury and liver toxicity that varies in prevalence and severity by the use of different drugs. There is a noticeable increase in SGOT (serum glutamic oxaloacetic transaminase) and SGPT (serum glutamic pyruvic transaminase) levels caused by drug-induced liver toxicity [46]. The potent compounds 4f and 4i when studied for their liver toxicity exhibited approximately equal value (p < 0.01) of biochemical parameters viz., SGPT and SGOT in comparison to control while compound 4e revealed noticeable escalation in the serum hepatic enzyme levels (SGOT and SGPT) (Figure 5 and Table 3).

Figure 5: 
Graphical representation of effect on (a) SGPT and (b) SGOT levels. (Dox, doxorubicin).
Figure 5:

Graphical representation of effect on (a) SGPT and (b) SGOT levels. (Dox, doxorubicin).

Table 3:

Effects of most active compounds on levels of enzyme transaminase in liver function tests.

Compounds SGOT ± SEM SGPT ± SEM
4i 37 ± 2 22 ± 3
4f 105 ± 3 30.3 ± 1.6
4e 36.2 ± 1.2 29.3 ± 1.2
4c 108.0 ± 1.0 31 ± 3
Control 34.2 ± 1.5 27.7 ± 1.4
Doxorubicin 151.2 ± 1.5 32.8 ± 0.4

The most potent compound 4i studied for liver toxicity displayed analogous (p < 0.01) biochemical parameters; SGOT and SGPT as compared to control (Table 3). On the other hand, the animals treated with doxorubicin revealed substantial enhancement (p < 0.01) in SGOT and SGPT levels in comparison to control [47, 48].

3.6 Molecular docking

Molecular docking can help to predict the most energetically favorable binding pose of a ligand to its receptor in terms of the binding energy [41, 49], [50], [51], [52], [53]. Molecular docking studies were implemented to ascertain the molecular binding pattern of the synthesized benzothiazole clubbed furanone derivatives within the active pocket of the crystal structure of VEGFR (PDB ID: 2QU5) using Schrodinger program Maestro 10.5 [54]. Various interactions including hydrogen bonding, hydrophobic and π-interactions, etc., with the active pocket of the enzyme were taken into account while calculating the docking scores. Since, VEGFR-2 plays a significant role in angiogenesis, lymphangiogenesis, tumor growth, and metastasis, it is considered a promising target for anticancer therapy [55]. Table 4 displays the docking score and the binding energies of compounds 4a–j. Compounds 4a and 4i revealed the highest docking score −10.64 and −10.33, respectively in the active pocket of the VEGFR. Compound 4a forms five hydrogen bonds-two with amino acid LYS 920 (–H···O═C and O–H···O═C), two with CYS 919 (H···O═C and C═O–···H) and one with GLU 885 (H···O═C). While, compound 4i forms four hydrogen bonds, one with amino acid ASP 1046 (O···HN), two with CYS 919 (H···O═C and O–···HN) and one with GLU 885 (O–H···O═C). It was observed that the furanone ring of compounds 4a and 4i play an essential role in the hydrogen bonding with the –NH of the principal chain of CYS 919, a key amino acid residue of VEGFR found in its flexible hinge region. The aromatic benzene ring of both the compounds 4a and 4i interacts with the hydrophobic pocket along with hydrogen bond formation with GLU 885. The surface view of the compounds 4a and 4i revealed that both fit entirely in the receptor pocket of the VEGFR enzyme (Figure 6). The docking simulation methodology was validated by redocking of the co-crystal ligand of PDB ID: 2QU5 with compound 4i in the same binding domain of VEGFR and investigating the overlying pattern of 4i with doxorubicin as well as a co-crystal ligand as shown in Figure 7.

Table 4:

Docking scores and binding free energies of the synthesized compounds 4a–j.

Compounds Docking Score Binding free energy (kcal/mol) Type of interactions
Hydrogen bonds π-Interactions
Atom Amino acids Distance (Å) Type Ring Amino acids Distance (Å)
4a −10.33 −45.43 H LYS 920 2.00
H LYS 920 2.68
H CYS 919 2.71
O CYS 919 1.88
H GLY 885 2.39
4b −9.68 −49.56 O CYS 919 1.97
H GLU 885 2.37
4c −9.34 −49.61 H ILE 1025 2.28 π-cation Thiazole LYS 868 3.98
N ASP 1046 2.17
4d −9.00 −48.87 H ILE 1025 2.41 π-cation Thiazole LYS 868 4.05
H ILE 1025 2.36
N ASP 1046 2.45
4e −9.81 −47.58 H LYS 920 2.24
H CYS 919 2.18
O CYS 919 2.06
H GLU 885 2.53
4f −9.57 −46.48 H LYS 920 2.68
H CYS 919 2.74
O CYS919 1.88
H GLU 885 2.36
4g −9.75 −48.01 H LYS 920 2.77
OH CYS 919 1.90
GLU 885 2.42
4h −9.41 −50.67 H CYS 919 2.80 π-cation Phenyl LYS 868 4.16
O CYS 919 1.99
H GLU 885 2.29
4i −10.64 −51.56 H CYS 919 2.54 π-cation Phenyl LYS 868 4.02
O CYS 919 3.66
O ASP 1046 1.98
H GLU 885 1.79
4j −7.55 −44.71 H GLU 917 2.53 π–π stacking Phenyl PHE 1047 3.38
H THR 916 2.78
Doxorubucin −7.73 −36.75 H CYS 919 2.26
H LYS 920 2.28
O ASN 923 3.11
H LEU 840 2.97
H PRO 839 2.24
  1. ASN, asparagine; ASP, aspartic acid; CYS, cysteine; GLU, glutamic acid; GLY, glycine; ILE, isoleucine; LYS, lysine; PHE, phenylalanine; PRO, proline; THR, threonine.

Figure 6: 
Docked posed conformer of compound (a) 4a and (b) 4i.
Figure 6:

Docked posed conformer of compound (a) 4a and (b) 4i.

Figure 7: 
Superimpose of compound (a) 4i (reddish–brown) and doxorubicin (green) displayed one common H-bond with CYS 919 (yellow) (b) 4i (green) and co-crystal ligand (reddish–brown) displayed two common H-bonds with CYS 919 and ASP 1046 (yellow).
Figure 7:

Superimpose of compound (a) 4i (reddish–brown) and doxorubicin (green) displayed one common H-bond with CYS 919 (yellow) (b) 4i (green) and co-crystal ligand (reddish–brown) displayed two common H-bonds with CYS 919 and ASP 1046 (yellow).

The surface view of the compounds 4a and 4i revealed that both 4a and 4i fit entirely in the receptor binding pocket of VEGFR and is shown in Figure 8. XP visualization of the hydrophobic enclosure pose of compounds 4a and 4i is displayed in Figure 9. The hydrophobic part of compounds 4a and 4i is enclosed by LEU 1035, LEU 840, VAL 848, PHE 918, LYS 868, ALA 866, VAL 914, LEU 882, and LEU 889. These hydrophobic interactions delivered optimum lipophilicity to compounds 4a and 4i enabling them to cross blood-brain barrier like membrane leading to improved interaction with the receptor pocket of VEGFR-2 kinase and therefore better VEGFR-2 kinase inhibition [56]. Thus, from these results, it is concluded that compounds 4a and 4i have the potential to act as VEGFR inhibitors.

Figure 8: 
Molecular surface view of compound 4a (upper panel) and compound 4i (lower panel).
Figure 8:

Molecular surface view of compound 4a (upper panel) and compound 4i (lower panel).

Figure 9: 
XP visualization of hydrophobic enclosures of compounds (a) 4a and (b) 4i. A ball and stick model (green) was used to represent the hydrophobic atoms of compounds 4a and 4i and hydrophobic residues (amino acids) shown in the turquoise CPK presentation.
Figure 9:

XP visualization of hydrophobic enclosures of compounds (a) 4a and (b) 4i. A ball and stick model (green) was used to represent the hydrophobic atoms of compounds 4a and 4i and hydrophobic residues (amino acids) shown in the turquoise CPK presentation.

3.7 MM-GBSA binding free energy

The binding energy results from several drug interactions with the pockets of the receptor including polar, electrostatic, van der Waals and hydrophobic interaction, π–π stacking, etc. [57]. The compounds, 4a and 4i exhibited higher docking scores and also higher binding energies of −45.43 and −51.56 kcal/mol, respectively. Amongst all, compound 4i exhibited the highest DG binding (−51.56 kcal/mol), even higher than standard drug doxorubicin −36.75 kcal/mol. The binding energies of all other compounds fall in the range of −44.71 to −51.56 kcal/mol.

3.8 ADME (adsorption, distribution, metabolism, excretion)

ADME properties of potent best-docked compounds such as QPlogKP for the permeability of the skin, QPlogBB for overall CNS activity, QPlogKhsa represent the binding of human serum albumin (HSA), etc., are represented in Table 5. All the compounds of benzothiazole linked furanone are predicted to possess acceptable as well as excellent molecular descriptors that include molecular weight, hydrogen bond donor, hydrogen bond acceptor, QPlogKP, Percent human Oral Absorption, QPlogBB, QPlogKhsa, and QPlogPo/w values thereby satisfying the Lipinski’s Rule of Five (Table 5). However, both compounds 4a and 4i displayed the highest value of QPlogPw, H-bond acceptor, QPlogPoct, and % of human oral absorption, compound 4i possessing additional hydroxyl and methoxy group demonstrated the best ADME parameters. Therefore, along with the results obtained from the docking scores, MMGB assay, and ADME prediction, compound 4i was found to be the best compound displaying better pharmacokinetic and pharmacodynamic properties.

Table 5:

ADME of the potent compounds 4a, 4e, and 4i.

Parameters Compounds
4a 4e 4i
Mol. Weight (130–725) 321.35 339.79 351.37
Dipole (1.0–12.5) 6.27 7.01 6.68
H-bond donor 1 0 1
H-bond acceptor 4.75 4.00 5.50
QPlogPw 10.09 7.75 10.32
QPlogPoct 16.28 15.15 17.23
QplogPo/w (−2.0 to 6.5) 3.15 4.24 3.32
QPlogHERG −6.17 −6.20 −6.04
QplogBB (−3 to 1.2) −0.72 −0.05 −0.19
QPlogKp (−8.0 to −0.1) −2.12 −1.12 −2.01
QPlogKhsa (acceptable range: −1.5 to 1.5) 0.22 0.35 0.23
Percent of human oral absorption >80%-high <25%-poor 96.02 100.00 100.00
Rule of three 0 0 0
Rule of five 0 0 0
  1. QPlogPo/w, octanol/water partition co-efficient; QPlogHERG, predicted IC50 value for blockage of HERG K+ channels; QPlogBB, predicted brain/blood partition coefficient; QPlogKP, predicted skin permeability; QPlogKhsa, prediction of binding to human serum albumin; percentage of human oral absorption; rule of five-number of violations of Lipinski’s rule of five; rule of three-number of violations of Jorgensen’s rule of three.

4 Conclusions

Drug design based on pharmacophore and hybrid compound synthesis was the foundation of the current work to obtain potent anticancer agents. Consequently, 10 new compounds 4a–j bearing two different biologically active potential pharmacophores viz., benzothiazole and furanone were synthesized by a multi-step synthetic protocol. They were further evaluated for in vitro cytotoxicity employing MTT colorimetric assay against a panel of three human cancer cell lines: A549 (human alveolar adenocarcinoma epithelial cells), DU145 (human prostate cancer cells), and MCF7 (human breast adenocarcinoma). All the tested compounds displayed substantial activity (IC50 in µM) against these cancer cell lines. Compound 4i displayed excellent activity against A549, MCF7 and DUI45 with IC50 value 7.2 ± 0.5, 6.6 ± 1.4 and 7.3 ± 0.1 µM, respectively. Another compound 4c exhibited very good activity against A549, MCF7, and DUI45 having IC50 value 9.6 ± 0.1, 7.3 ± 0.2, and 8.7 ± 0.2 µM, respectively. Acute toxicity study of these compounds suggested that the compounds 4i, 4f, 4e and 4c are not toxic for the vital organs-liver and heart. Unlike doxorubicin, which shows cardiac toxicity, they do not disrupt the normal architecture of cells of the liver and heart. So, these compounds were found to be efficient and less toxic as compared to doxorubicin. The p value of <0.001 shows that data is found to be statistically significant. Additionally, two compounds 4e and 4f also exhibited good cytotoxicity against all three cancer cell lines. Docking studies were also performed for compounds 4a–j, revealing the potential of compounds 4a and 4i as VEGFR inhibitors.

5 Supporting information

Other experimental details can be found in electronic supporting information (https://doi.org/10.1515/znb-2021-0146).

Abbreviations

A549

Human alveolar adenocarcinoma epithelial cells

ADME

Adsorption, distribution, metabolism, excretion

ALA

Alanine

DMSO

Dimethylsulfoxide

DU145

Human prostate cancer cells

FDA

Food and Drug Administration

HCC

Hepatocellular carcinoma

HeLa

Human cervical cancer cell line

HepG2

Human liver carcinoma cells

MCF7

Human breast adenocarcinoma

MM-GBSA

Molecular mechanics-generalized Born and surface area

NSCLC

Non-small cell lung cancer

PDB

Protein data bank

PPA

Polyphosphoric acid

SGOT

Serum glutamic oxaloacetic transaminase

SGPT

Serum glutamic pyruvic transaminase

SW480

Human colon adenocarcinoma cell line

TEA

Trimethylamine

TK

Tyrosine kinase

TLC

Thin layer chromatography

VAL

Valine

VEGFR

Vascular endothelial growth factor receptor

WHO

World health organization


Corresponding author: Shazia Parveen, Chemistry Department, Faculty of Science, Taibah University, Yanbu Branch, 46423, Yanbu, Saudi Arabia; and Department of Chemistry, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India, E-mail:

Acknowledgments

The author S. Parveen is grateful to Jamia Hamdard for “JH Silver Jubilee Post-Doctoral Fellowship-2016”.

  1. Author contributions: All the authors have accepted responsibility for the entire content of this submitted manuscript and approved submission.

  2. Research funding: None declared.

  3. Conflict of interest statement: The authors declare no conflicts of interest regarding this article.

References

1. Senwar, K. R., Reddy, T. S., Thummuri, D., Sharma, P., Bharghava, S. K., Naidu, V. G. M., Shankaraiah, N. Bioorg. Med. Chem. Lett. 2016, 26, 4061–4069. https://doi.org/10.1016/j.bmcl.2016.06.077.Suche in Google Scholar PubMed

2. Weir, H. K., Anderson, R. N., Coleman King, S. M., Soman, A., Thompson, T. D., Hong, Y., Moller, B., Leadbetter, S. Chronic Dis 2016, 13, E157. https://doi.org/10.5888/pcd13.160211.Suche in Google Scholar PubMed PubMed Central

3. WHO, 2020. https://www.who.int/news-room/fact-sheets/detail/cancer.Suche in Google Scholar

4. Folkman, J. In Biology of Endothelial Cells. Developments in Cardiovascular Medicine; Jaffe, E. A, Ed. Springer: Boston, MA, Vol. 27, 1984, pp. 412–428.10.1007/978-1-4613-2825-4_42Suche in Google Scholar

5. Folkman, J. Nat. Med. 1995, 1, 27–30. https://doi.org/10.1038/nm0195-27.Suche in Google Scholar PubMed

6. Kerbel, R. S. Carcinogenesis 2000, 21, 505–515. https://doi.org/10.1093/carcin/21.3.505.Suche in Google Scholar PubMed

7. Grothey, A., Galanis, E. Nat. Rev. Clin. Oncol. 2009, 6, 507–518. https://doi.org/10.1038/nrclinonc.2009.110.Suche in Google Scholar PubMed

8. Folkman, J. Nat. Rev. Drug Discov. 2007, 6, 273–286. https://doi.org/10.1038/nrd2115.Suche in Google Scholar PubMed

9. Färkkilä, A., Anttonen, M., Pociuviene, J., Leminen, A., Butzow, R., Heikinheimo, M., Unkila-Kallio, L. Eur. J. Endocrinol. 2011, 164, 115–122.10.1530/EJE-10-0849Suche in Google Scholar PubMed

10. Goel, H. L., Mercurio, A. M. Nat. Rev. Cancer 2013, 13, 871–882. https://doi.org/10.1038/nrc3627.Suche in Google Scholar PubMed PubMed Central

11. Ferrara, N., Adamis, A. P. Nat. Rev. Drug Discov. 2016, 15, 385–403. https://doi.org/10.1038/nrd.2015.17.Suche in Google Scholar PubMed

12. Peng, F. W., Liu, D. K., Zhang, Q. W., Xu, Y. G., Shi, L. Expert Opin. Ther. Pat. 2017, 27, 987–1004. https://doi.org/10.1080/13543776.2017.1344215.Suche in Google Scholar PubMed

13. Huang, L., Huang, Z., Bai, Z., Xie, R., Sun, L., Lin, K. Future Med. Chem. 2012, 4, 1839–1852. https://doi.org/10.4155/fmc.12.121.Suche in Google Scholar PubMed

14. Abd-Rabou, A. A., Abdel-Wahab, B. F., Bekheit, M. S. Chem. Pap. 2018, 72, 2225–2237. https://doi.org/10.1007/s11696-018-0451-5.Suche in Google Scholar

15. Reddy, V. G., Reddy, T. S., Jadala, C., Reddy, M. S., Sultana, F., Akunuri, R., Bhargava, S. K., Wlodkowic, D., Srihari, P., Kamal, A. Eur. J. Med. Chem. 2019, 182, 111609. https://doi.org/10.1016/j.ejmech.2019.111609.Suche in Google Scholar PubMed

16. Khokra, S. L., Arora, K., Khan, S. A., Kaushik, P., Saini, R., Husain, A. Iran. J. Pharm. Res. 2019, 18, 1–15.Suche in Google Scholar

17. Osmaniye, D., Levent, S., Karaduman, A. B., Ilgın, S., Zkay, Y., Kaplancikli, Z. A. Molecules 2018, 23, 1054. https://doi.org/10.3390/molecules23051054.Suche in Google Scholar PubMed PubMed Central

18. Sulthana, S., Pandian, P. J. Drug Deliv. Ther. 2019, 9, 505–509. https://doi.org/10.22270/jddt.v9i1-s.2358.Suche in Google Scholar

19. Hebishy, A. M. S., Abdelfattah, M. S., Elmorsy, A., Elwahy, A. H. M. J. Heterocycl. Chem. 2020, 57, 2256–2270. https://doi.org/10.1002/jhet.3947.Suche in Google Scholar

20. Abdou, W. M., Khidre, M. D., Shaddy, A. A. Chem. Pap. 2018, 72, 2753–2768. https://doi.org/10.1007/s11696-018-0505-8.Suche in Google Scholar

21. Rashid, M., Husain, A., Mishra, R. Eur. J. Med. Chem. 2012, 54, 855–866. https://doi.org/10.1016/j.ejmech.2012.04.027.Suche in Google Scholar PubMed

22. Oguro, Y., Cary, D. R., Miyamoto, N., Tawada, M., Iwata, H., Miki, H., Hori, A., Imamura, S. Bioorganic Med. Chem. 2013, 21, 4714–4729. https://doi.org/10.1016/j.bmc.2013.04.042.Suche in Google Scholar PubMed

23. Gabr, M. T., El-Gohary, N. S., El-Bendary, E. R., El-Kerdawy, M. M. Med. Chem. Res. 2015, 24, 860–878. https://doi.org/10.1007/s00044-014-1114-x.Suche in Google Scholar

24. Okaniwa, M., Hirose, M., Arita, T., Yabuki, M., Nakamura, A., Takagi, T., Kawamoto, T., Uchiyama, N., Sumita, A., Tsutsumi, S., Tottori, T., Inui, Y., Sang, B. C., Yano, J., Aertgeerts, K., Yoshida, S., Ishikawa, T. J. Med. Chem. 2013, 56, 6478–6494. https://doi.org/10.1021/jm400778d.Suche in Google Scholar PubMed

25. Abdel-Mohsen, H. T., Abd El-Meguid, E. A., El Kerdawy, A. M., Mahmoud, A. E. E., Ali, M. M. Arch. Pharm. (Weinheim) 2020, 353, 1900340.10.1002/ardp.201900340Suche in Google Scholar PubMed

26. Yuan, X., Yang, Q., Liu, T., Li, K., Liu, Y., Zhu, C., Zhang, Z., Li, L., Zhang, C., Xie, M., Lin, J., Zhang, J., Jin, Y. Eur. J. Med. Chem. 2019, 179, 147–165. https://doi.org/10.1016/j.ejmech.2019.06.054.Suche in Google Scholar PubMed

27. Khan, S. A., Husain, A., Moosa Al Lawatia, Y., Benzothiazole, A. S. A promising Scaffold for the development of anticancer agents. In Benzothiazole: Preparation, Structure and Uses; Nova Science Publishers, Inc: NY, USA, 2020.Suche in Google Scholar

28. Irfan, A., Batool, F., Zahra Naqvi, S. A., Islam, A., Osman, S. M., Nocentini, A., Alissa, S. A., Supuran, C. T. J. Enzyme Inhib. Med. Chem. 2020, 35, 265–279. https://doi.org/10.1080/14756366.2019.1698036.Suche in Google Scholar PubMed PubMed Central

29. You, L., Liang, X. T., Xu, L. M., Wang, Y. F., Zhang, J. J., Su, Q., Li, Y. H., Zhang, B., Yang, S. L., Chen, J. H., Yang, Z. J. Am. Chem. Soc. 2015, 137, 10120–10123. https://doi.org/10.1021/jacs.5b06480.Suche in Google Scholar PubMed

30. Wang, Z. H., You, Y., Chen, Y. Z., Xu, X. Y., Yuan, W. C. Org. Biomol. Chem. 2018, 16, 1636–1640. https://doi.org/10.1039/c7ob03117c.Suche in Google Scholar PubMed

31. Wei, M. X., Zhang, J., Ma, F. L., Li, M., Yu, J. Y., Luo, W., Li, X. Q. Eur. J. Med. Chem. 2018, 155, 165–170. https://doi.org/10.1016/j.ejmech.2018.05.056.Suche in Google Scholar PubMed

32. Husain, A., Khan, S. A., Iram, F., Iqbal, M. A., Asif, M. Eur. J. Med. Chem. 2019, 171, 66–92. https://doi.org/10.1016/j.ejmech.2019.03.021.Suche in Google Scholar PubMed

33. Duan, Y. C., Ma, Y. C., Zhang, E., Shi, X. J., Wang, M. M., Ye, X. W., Liu, H. M. Eur. J. Med. Chem. 2013, 62, 11–19. https://doi.org/10.1016/j.ejmech.2012.12.046.Suche in Google Scholar PubMed

34. Khatri, H. R., Bhattarai, B., Kaplan, W., Li, Z., Curtis Long, M. J., Aye, Y., Nagorny, P. J. Am. Chem. Soc. 2019, 141, 4849–4860. https://doi.org/10.1021/jacs.8b12870.Suche in Google Scholar PubMed PubMed Central

35. Xi, F. M., Ma, S. G., Liu, Y. B., Li, L., Yu, S. S. Org. Lett. 2016, 18, 3374–3377. https://doi.org/10.1021/acs.orglett.6b01519.Suche in Google Scholar PubMed

36. Tan, K., Yan, H., Lu, P., Liu, Y., Ji, R., Liu, Z., Li, Y. M., Yu, F. C., Shen, Y. J. Org. Chem. 2019, 84, 3419–3430. https://doi.org/10.1021/acs.joc.8b03293.Suche in Google Scholar PubMed

37. Wu, X., Iwata, T., Scharf, A., Qin, T., Reichl, K. D., Porco, J. A. J. Am. Chem. Soc. 2018, 140, 5969–5975. https://doi.org/10.1021/jacs.8b02535.Suche in Google Scholar PubMed PubMed Central

38. Rashid, M., Husain, A., Mishra, R., Karim, S., Khan, S., Ahmad, M., Al-wabel, N., Husain, A., Ahmad, A., Khan, S. A. Arab. J. Chem. 2019, 12, 3202–3224. https://doi.org/10.1016/j.arabjc.2015.08.019.Suche in Google Scholar

39. Husain, A., Rashid, M., Shaharyar, M., Siddiqui, A. A., Mishra, R. Eur. J. Med. Chem. 2013, 62, 785–798. https://doi.org/10.1016/j.ejmech.2012.07.011.Suche in Google Scholar PubMed

40. Rashid, M., Husain, A., Shaharyar, M., Sarafroz, M. Anti Cancer Agents Med. Chem. 2014, 14, 1003–1018. https://doi.org/10.2174/1871520614666140509153021.Suche in Google Scholar PubMed

41. Husain, A., Bhutani, M., Parveen, S., Khan, S. A., Ahmad, A., Iqbal, M. A. J. Chinese Chem. Soc. 2021, 68, 362–373. https://doi.org/10.1002/jccs.202000130.Suche in Google Scholar

42. Cardinale, D., Colombo, A., Lamantia, G., Colombo, N., Civelli, M., De Giacomi, G., Rubino, M., Veglia, F., Fiorentini, C., Cipolla, C. M. J. Am. Coll. Cardiol. 2010, 55, 213–220. https://doi.org/10.1016/j.jacc.2009.03.095.Suche in Google Scholar PubMed

43. Orphanos, G. S., Ioannidis, G. N., Ardavanis, A. G. Acta Oncol. (Madr). 2009, 48, 964–970. https://doi.org/10.1080/02841860903229124.Suche in Google Scholar PubMed

44. Mellor, H. R., Bell, A. R., Valentin, J. P., Roberts, R. R. A. Toxicol. Sci. 2011, 120, 14–32. https://doi.org/10.1093/toxsci/kfq378.Suche in Google Scholar PubMed

45. Sinha, S. K., Shrivastava, P. K., Shrivastava, S. K. Chem. Pap. 2011, 65, 70–76. https://doi.org/10.2478/s11696-010-0094-7.Suche in Google Scholar

46. Podda, M., Benetti, A. Ann. Ital. Med. Int. 2002, 17, 213–214.10.1177/106286060201700602Suche in Google Scholar

47. Solomon, V. R., Hu, C., Lee, H. Bioorganic Med. Chem. 2009, 17, 7585–7592. https://doi.org/10.1016/j.bmc.2009.08.068.Suche in Google Scholar PubMed

48. Spraggs, C. F., Xu, C. F., Hunt, C. M. Pharmacogenomics 2013, 14, 541–554. https://doi.org/10.2217/pgs.13.24.Suche in Google Scholar PubMed

49. Mohammed, F. F., Hagar, M., Parveen, S., Alnoman, R. B., Ahmed, H. A., Ashry, E. S. H. E., Rasheed, H. A. Polycycl. Aromat. Compd. 2021. https://doi.org/10.1080/10406638.2021.1878245.Suche in Google Scholar

50. Parveen, S., Alnoman, R. B., Hagar, M., Ahmed, H. A., Knight, J. G. ChemistrySelect 2020, 5, 13163–13173. https://doi.org/10.1002/slct.202003621.Suche in Google Scholar

51. Parveen, S. Biointerface Res. Appl. Chem. 2021, 11, 9604–9618.10.33263/BRIAC112.96049618Suche in Google Scholar

52. Parveen, S., Arjmand, F., Zhang, Q., Ahmad, M., Khan, A., Toupet, L. J. Biomol. Struct. Dyn. 2021, 39, 2092–2105. https://doi.org/10.1080/07391102.2020.1743365.Suche in Google Scholar PubMed

53. Parveen, S., Hagar, M., Alnoman, R. B., Ahmed, H. A., El Ashry, E. S. H., Zakaria, M. A. Polycycl. Aromat. Compd. 2021. https://doi.org/10.1080/10406638.2021.1871636.Suche in Google Scholar

54. Schrödinger Release 2017-4; Maestro, Schrödinger, LLC: New York, NY, 2017.10.2307/j.ctvcm4h07.63Suche in Google Scholar

55. Çoban, G. Aydın Köse F. Saudi Pharm. J. 2019, 27, 952–967.10.1016/j.jsps.2019.07.004Suche in Google Scholar PubMed PubMed Central

56. Wissner, A., Floyd, M. B., Johnson, B. D., Fraser, H., Ingalls, C., Nittoli, T., Dushin, R. G., Discafani, C., Nilakantan, R., Marini, J., Ravi, M., Cheung, K., Tan, X., Musto, S., Annable, T., Siegel, M. M., Loganzo, F. J. Med. Chem. 2005, 48, 7560–7581. https://doi.org/10.1021/jm050559f.Suche in Google Scholar PubMed

57. Fukunishi, Y., Yamashita, Y., Mashimo, T., Nakamura, H. Mol. Inform. 2018, 37, 1700120. https://doi.org/10.1002/minf.201700120.Suche in Google Scholar PubMed PubMed Central


Supplementary Material

The online version of this article offers supplementary material (https://doi.org/10.1515/znb-2021-0146).


Received: 2021-08-12
Accepted: 2021-11-13
Published Online: 2021-11-26
Published in Print: 2022-01-27

© 2021 Walter de Gruyter GmbH, Berlin/Boston

Heruntergeladen am 21.11.2025 von https://www.degruyterbrill.com/document/doi/10.1515/znb-2021-0146/html
Button zum nach oben scrollen