Next Article in Journal
The Similarities and Differences between the Effects of Testosterone and DHEA on the Innate and Adaptive Immune Response
Next Article in Special Issue
Efavirenz: History, Development and Future
Previous Article in Journal
The Distribution of Innervation and Immune Cell Infiltration Is Different in Genital and Extragenital Variants of Lichen Sclerosus
Previous Article in Special Issue
Antidepressant Drug Sertraline against Human Cancer Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Anticancer, Antimicrobial, and Antioxidant Activities of Organodiselenide-Tethered Methyl Anthranilates

by
Batool Al-Abdallah
1,
Yasair S. Al-Faiyz
1 and
Saad Shaaban
1,2,*
1
Chemistry Department, College of Science, King Faisal University, Al-Ahsa 31982, Saudi Arabia
2
Chemistry Department, Faculty of Science, Mansoura University, Mansoura 35516, Egypt
*
Author to whom correspondence should be addressed.
Biomolecules 2022, 12(12), 1765; https://doi.org/10.3390/biom12121765
Submission received: 15 October 2022 / Revised: 18 November 2022 / Accepted: 23 November 2022 / Published: 27 November 2022
(This article belongs to the Special Issue New Advances in Drug Repurposing for Oncology)

Abstract

:
Novel methyl anthranilate-based organodiselenide hybrids were synthesized, and their chemical structures were confirmed by state-of-the-art spectroscopic techniques. Their antimicrobial properties were assessed against Staphylococcus aureus, Escherichia coli, and Candida albicans microbial strains. Moreover, the antitumor potential was estimated against liver and breast carcinomas, as well as primary fibroblast cell lines. The Staphylococcus aureus and Candida albicans strains were more sensitive than Escherichia coli toward the OSe compounds. Interestingly, methyl 2-amino-5-(methylselanyl) benzoate (14) showed similar antifungal activity to the standard drug clotrimazole (IA% = 100%) and manifested promising antibacterial activity against E. coli (IA% = 91.3%) and S. aureus (IA% = 90.5%). Furthermore, the minimum inhibitory concentration experiments confirmed the antimicrobial activity of the OSe 14, which in turn was comparable to clotrimazole and ampicillin drugs. Interestingly, the anticancer properties were more pronounced in the HepG2 cells. The OSe 14 was the most cytotoxic (IC50 = 3.57 ± 0.1 µM), even more than the Adriamycin drug (IC50 = 4.50 ± 0.2 µM), and with therapeutic index (TI) 17 proposing its potential selectivity and safety. Additionally, OSe compounds 14 and dimethyl 5,5′-diselanediylbis(2-aminobenzoate) (5) exhibited promising antioxidants in the 2,2′-azino-bis (3-ethylbenzothiazoline-6-sulfonic acid (ABTS) and 2,2-diphenyl-1-picrylhydrazyl (DPPH) in vitro assays with 96%, 92%, 91%, and 86% radical scavenging activities compared to 95% by vitamin C in the DPPH and ABTS assays, respectively. These results point to promising antimicrobial, anticancer, and antioxidant activities of OSe 14 and 5 and warrant further studies.

1. Introduction

Organoselenium (OSe) compounds have raised much more concern than their inorganic selenium (Se) peers due to their numerous pharmaceutical applications as well as improved pharmacokinetics and bioavailability [1,2]. The non-metal Se element belongs to the oxygen (a.k.a. chalcogen) group. Se has a leading function in immune systems protection and cancer cell proliferation inhibition [3,4]. Various infectious and autoimmune illnesses are somehow linked with the deficiency of Se [4]. On the other hand, Se dietary supplementation was associated with improving different inflammatory disorders and chemoprevention of various types of cancer [5]. Within this context, OSe compounds have shown antiviral, antimicrobial, and antioxidant activities [6]. Due to their diverse pharmacological activities, organodiselenide (OSe2) compounds are among the most evaluated OSe derivatives [7,8,9]. Therefore, researchers have recently been directed to disclose new OSe2 for biological testing. For instance, diphenyl diselenide (I) is an antidepressant and an antioxidant agent [10]. Moreover, compounds 1,2-bis(chloropyridazinyl) diselenide (II) and bis(4-amino-3-carboxyphenyl) diselenide (III) were able to inhibit the growth of breast cancer cells and manifest efficient anti-leishmanial properties, respectively [5].
Furthermore, the anthranilic acid-based derivatives captured our interest as they were found in the backbone of several drugs (e.g., colfenamate (IV) and tranilast (V)) and natural products (e.g., essential oils) (Figure 1) and manifested different biological applications (e.g., anti-inflammatory, anticancer, antibacterial, and antioxidant) [11]. Therefore, the anthranilate scaffold offers a promising pharmacophore for developing novel drugs designed to target different diseases [12].
Accordingly, combining the pharmacologically active anthranilate scaffolds and OSe2 would promote overall biological activities. Therefore, we aim to design and synthesize new OSe2-tethered anthranilate hybrids and evaluate their respective antimicrobial, anticancer, and antioxidant properties.

2. Materials and Methods

2.1. Material and Methods

All reagents and solvents used in this study were purchased from Sigma and used without purification. Melting points (MP) were measured on the Gallenkamp apparatus in degrees centigrade. The IR spectra (KBr, λmax.cm−1) were recorded on a Mattson spectrophotometer (5000 FTIR) at King Faisal University. The 1HNMR and 13CNMR spectra were measured using Varian Spectrophotometer (400 MHz), employing DMSO-d6 as the solvent and TMS internal standard at Mansoura University. The chemical shifts (δ, ppm) were recorded regarding the solvent residual peaks. GC-MS-QP-100 EX Shimadzu apparatus was used for mass measurements at Cairo University. All biological tests were carried out at the Faculty of Pharmacy, Mansoura University. All cell lines and microorganisms were purchased from the VACSERA Company (ATCC), Cairo, Egypt. DPPH and ABTS probes were obtained from Sigma. Compound 2 was synthesized following our literature method [13]. Copies of 1H & 13CNMR spectra IR and MS can be found in the Supplementary Materials Section S2.

2.2. Chemistry

Procedure I: The preparation of selenocyanate 4 and diselenide 5.
Methyl 2-amino-5-selenocyanatobenzoate (4) was synthesized from the reaction of methyl 2-aminobenzoate with triselenium dicyanide prepared in situ from malononitrile and selenium dioxide in 96% yields. Briefly, selenium dioxide (30 mmol, 3300 mg) was added to malononitrile (15 mmol, 1000 mg) in 10 mL DMSO, and the mixture was stirred for 20 min at room temperature. Next, the mixture was filtered off to get rid of any formed black selenium, and methyl 2-aminobenzoate (12.5 mmol, 1800 mg) was then added, and the reaction mixture was stirred for a further 2 hr at room temperature. Finally, adding 10 g of ice terminated the reaction, and the formed precipitate was filtered, washed several times with H2O and sodium carbonate solution, dried, and recrystallized from petroleum ether.
Compound dimethyl 5,5-diselanediylbis(2-aminobenzoate) (5) was synthesized from the reaction of 4 and sodium hydroxide in 92% yields. Briefly, compound 4 (4 mmol, 1000 mg) was dissolved in EtOH (20 mL), and then sodium hydroxide (4 mmol, 160 mg) was added. The reaction mixture was stirred for 2 h at room temperature, and the resulting precipitate was filtered, washed several times with H2O, and recrystallized from chloroform.
Procedure II: The preparation of the OSe azo dyes 6.
Dimethyl 5,5′-diselanediylbis(2-aminobenzoate) (5) (2 mmol,916.4 mg) was dissolved in aqueous HCl (4 mL) and cooled to 0–5 °C. Sodium nitrite (4.4 mmol, 331 mg in 10 mL H2O) was then added to the previously prepared solution while maintaining the temperature at 0–5 °C. The freshly prepared diazonium salt solution was then added dropwise to a cooled and stirred solution of methylene compounds, e.g., ethyl cyanoacetate (4.4 mmol) and sodium acetate (2000 mg) dissolved in (10 mL) of H2O. After stirring the reaction mixture for 3 h at 0–5 °C, the resulting precipitate was filtered, washed with H2O, and recrystallized from EtOH.
Procedure III: The preparation of OSe amide-acids 11 and 13.
Dimethyl 5,5′-diselanediylbis(2-aminobenzoate) (5) (2 mmol) was added to a stirred solution of maleic or succinic anhydride (4.4 mmol) in dry toluene (15 mL). The mixture was vigorously stirred, and the formed precipitate was separated by filtration. The residue was washed with hot toluene, dried under reduced pressure, and recrystallized from EtOH.
Procedure IV: The preparation of cyclic imide 12.
A mixture containing amide acids (11) (0.5 mmol) and sodium acetate (250 mg) in acetic anhydride (5 mL) was heated at 60–65 °C for three h. The reaction mixture was poured into ice H2O and neutralized with sodium carbonate. The precipitate formed was collected by filtration, washed 3x with sodium carbonate solution, and recrystallized from EtOH.
Procedure V: The preparation of organic selenides via reduction 14, 15, and 16.
Dimethyl 5,5′-diselanediylbis (2-aminobenzoate) (5) (2 mmol) and appropriate halo derivatives (4.4 mmol) were dissolved in EtOH (25 mL). Next, NaBH4 (6 mmol, 226.98 mg) was added portion-wise over one h. Then the reaction was stirred for an additional 2 h. Finally, the organic layer was dried and evaporated under a vacuum.
Synthesis of 2-amino-5-selenocyanatobenzoic acid (3) [9,13].
Malononitrile (15 mmol) was dissolved in (10 mL) of DMF, then SeO2 (30 mmol) was added. After 15 min, the reaction was filtered, and anthranilic acid (12.5 mmol) was added. The reaction was further stirred for another 15 min and added ice to the mixture. It was kept refrigerated overnight. The substance that formed was filtered off, dried, and recrystallized from DMF: H2O (1:1). Product was isolated as a brown solid; Yield: 76%; MP = 180 (decomposition)°C. 1HNMR (400 MHz, DMSO-d6) δ = 7.86(d, J = 14.9 Hz, 1H, Ar-H), 7.42(d, J = 2.7 Hz, 1H), 6.71(dd, J1 = Hz, J2 = 14.9 Hz, H1H, Ar-H); MS (ESI): m/z = found 241.29 [M+]; calcd 241.19 [M+]; Elemental Analysis C8H6N2O2Se: C, 39.81; H, 2.50; N, 11.60.
Synthesis of methyl 2-amino-5-selenocyanatobnzoate (4).
Methyl 2-amino-5-selenocyanatobenzoate (4) was synthesized following procedure I from methyl 2-aminobenzoate (12.5 mmol, 1800 mg) with triselenium dicyanide prepared in situ from malononitrile (15 mmol, 1000 mg) and selenium dioxide (30 mmol, 3300 mg). It was isolated as reddish solid; yield: 3072 mg (96%); MP = 118–119 °C; Rf = 0.4(petroleum ether/ethyl acetate 4:2). IR(KBr): λmax.cm−1: 3475, 3366, 2946, 2147, 1691. 1HNMR (400 MHz, DMSO-d6) δ8.02(s, 1H, Ar-H), 7.57(d, J = 8.8 Hz, 1H, Ar-H), 7.08(d, 1H, Ar-H), 6.84(s, 2H, NH2), 3.82(s, 3H, OCH3). 13C NMR (101 MHz, DMSO-d6) δ166, 152, 140, 137, 118, 109, 105, 105, 51. MS (EI, 70 ev) m/z (%) = 259.35(M+3H, 2.39), 117(29.02), 87(26.6), 75(2.70), 59(100.0, base peak).
Synthesis of dimethyl 5,5′-diselanediylbis(2-aminobenzoate) (5)
Compound dimethyl 5,5-diselanediylbis(2-aminobenzoate) (5) was synthesized following the procedure I from methyl 2-amino-5-selenocyanatobnzoate (4 mmol, 1000 mg) and sodium hydroxide (4 mmol, 160 mg) in EtOH (20 mL). Dimethyl 5,5′-diselanediylbis(2-aminobenzoate) (5) appeared as a single compound on TLC and was isolated as a yellow solid; yield: 1692.43 mg (92%); MP = 138–139 °C; Rf = 0.5(petroleum ether/ethyl acetate 4:3). IR(KBr): λmax.cm−1: 3455, 3344, 2931, 1684. 1HNMR (400 MHz, DMSO) δ7.70(s,2H, Ar-H), 7.44(d, J = 8.6 Hz, 2H, Ar-H), 7.00(s, 4H,2NH2), 6.77(d, J = 8.7 Hz, 2H, Ar-H), 3.74(s, 6H,2OCH3). 13CNMR (101 MHz, DMSO-d6) δ167, 151, 140, 138, 117, 113, 108, 51. MS (EI, 70 ev) m/z (%) = 460.15(M+H, 20.76), 459.15(M, 5.20) or 230(24.42), 119(9.45), 91(100.0, base peak), 65(8.88).
Synthesis of dimethyl 5,5′-diselanediylbis(2-(2-(1-cyano-2-ethoxy-2-oxoethylidene) hydrazinyl) benzoate) (6).
Compound 6 was synthesized following procedure II from dimethyl 5,5′-diselanediylbis(2-aminobenzoate) (5) (2 mmol, 916.4 mg) and ethyl cyanoacetate (4.4 mmol, 0.46 mL). It was isolated as yellow crystal; yield: 1147 mg (81%); MP = 105–106 °C; Rf = 0.4(petroleum ether/ethyl acetate 4:4). IR(KBr): λmax.cm−1: 3146, 2984, 2138, 1693. 1HNMR (400 MHz, DMSO-d6) δ12.56(s, 2H,2NH), 8.13(s, 2H, Ar-H), 7.83(d, J = 8.8 Hz, 2H, Ar-H), 7.70(d, J = 8.6 Hz, 2H, Ar-H), 4.4–4.29(q, 4H, 2OCH2), 3.96–3.88(s, 6H,2OCH3), 1.32(t, J = 7.2 Hz, 6H, 2CH3). MS (EI, 70ev) m/z (%) = 708.25(M, 18.03), 354(31.30), 111.20(2.62), 90.10(12.21), 59.10(100.0, base peak).
Synthesis of dimethyl 5,5′-diselanediylbis(2-formamidobenzoate) (7).
Acetic formic anhydride (freshly prepared, 36.3 mmol, 3.2 mL) was added dropwise at room temperature to a solution of dimethyl 5,5′-diselanediylbis(2-aminobenzoate) (5) (1 mmol, 458.2 mg) in THF solution (10 mL). The reaction progress was monitored with TLC. It was isolated as an orange solid and recrystallized from EtOH, yielding: 201.2 mg (39%); MP = 195–196 °C; Rf = 0.35(petroleum ether/ethyl acetate 4:2). IR(KBr): λmax.cm−1: 3255, 2951, 1681, 1573. 1HNMR (400 MHz, DMSO-d6) δ10.42(s, 2H,2NH), 8.39(s, 2H,2CHO), 8.36–8.12(s, 2H, Ar-H), 7.92(d, 2H, Ar-H), 7.60(d, J = 6.2 Hz, 2H, Ar-H), 3.75(s, 6H, 2OCH3). 13CNMR (101 MHz, DMSO-d6) δ166, 161, 139, 138, 135, 124, 122, 118, 53. MS (EI, 70 ev) m/z (%) = 516.10(M, 25.78), 515.15(M-1H, 8.92), 258.10(6.08), 229(13.95), 198(100.0basepeak), 134.20(7.40).
Synthesis of dimethyl 5,5′-diselanediylbis(2-acetamidobenzoate) (8).
A mixture of compound 5 (2 mmol,916.4 mg) and acetic anhydride (8 mL) in a 50 mL round-bottom flask fitted with a condenser was heated in an oil bath at 65–70 °C for 7 h. The reaction mixture was allowed to cool at room temperature, poured on ice-cold H2O, and neutralized with a sodium carbonate solution. The formed solid was collected by filtration, washed with H2O, and then recrystallized from EtOH. Compound 8 was isolated as a yellow solid; yielding: 462.4 mg (85%); MP = 186–187 °C; Rf = 0.6(petroleum ether/ethyl acetate 4:4). IR(KBr): λmax.cm−1: 3281, 2934, 1700, 1682. 1HNMR (400 MHz, DMSO-d6) δ10.57(s, 2H, 2NH), 8.18(s, 2H, Ar-H), 8.00(d, J = 23.7 Hz, 2H, Ar-H), 7.84(d, J = 7.7 Hz, 2H, Ar-H), 3.84(s, 6H, 2OCH3), 2.13(s, 6H,2CH3). MS (EI, 70 ev) m/z (%) = 544.30(M, 40.15), 272.15(17.66), 230(100.0 base peak), 192.1(1.81), 90.15(14.52).
Synthesis of dimethyl 5,5′-diselanediylbis(2-(2-chloroacetamido) benzoate) (9).
To a cold solution of compound (5) (2 mmol, 916.4 mg) in dry acetone (20 mL) and K2CO3 (2000 mg), chloroacetyl chloride (4.4 mmol, 0.34 mL) was added dropwise with stirring at 0–5 °C. Stirring was continued for 5 h, and the reaction mixture was poured onto ice-cold H2O. The resulting precipitate was collected, dried, and recrystallized from MeOH to afford the corresponding chloroacetamide. Compound 9 was isolated as a light-yellow solid; yielding: 569 mg (93%); MP = 150–152 °C; Rf = 0.6(petroleum ether/ethyl acetate 4:3). IR(KBr): λmax.cm−1: 3231, 2952, 1698, 1575, 777. 1HNMR (400 MHz, DMSO-d6) δ11.39(s, 2H,2NH), 8.40–8.35(s, 2H, Ar-H), 8.12(d, J = 1.9 Hz, 2H, Ar-H), 7.92(dd, J = 8.6, 1.9 Hz, 2H, Ar-H), 4.32(s, 4H,2CH2), 3.87(s, 6H,2OCH3). 13CNMR (101 MHz, DMSO) δ167, 165, 139, 138, 135, 125, 122, 118, 53, 43. MS (EI, 70 ev) m/z (%) = 611.15(M, 33.33), 612.10(100.0 base peak), 105(0.43), 92(0.42), 77.05(6.51).
Synthesis of dimethyl 5,5′-diselanediylbis(2-(2-phenoxyacetamido) benzoate) (10).
To compound 5 in dry ether (15 mL), phenoxy acetyl chloride (2.2 mmol, 0.30 mL) was added dropwise. The reaction was stirred at room temperature for 5 h. The solvent was then evaporated; the formed residue was dissolved in dichloromethane, and the organic layer was extracted using 10% HCl and 10% NaOH. The organic layer was filtered over anhydrous sodium sulfate and evaporated. Compound 10 was isolated as a yellow solid and recrystallized from EtOH, yielding: 400.4 mg (55%); MP = 152–154 °C; Rf = 0.4(petroleum ether/ethyl acetate 4:3). IR(KBr): λmax.cm−1: 3249, 2921, 2852, 1686, 1571. 1HNMR (400 MHz, DMSO-d6) δ12.51(s, 2H, 2NH), 8.65(s, 3H, Ar-H), 8.13(dd, J = 9.9, 1.9 Hz, 3H, Ar-H), 7.94 (dd, J = 12.8, 5.9 Hz, 3H, Ar-H), 7.50–7.33(q, 4H, Ar-H), 7.05(t, 3H, Ar-H), 4.79(s, 4H, 2CH2), 3.90(s, 6H, 2OCH3). 13CNMR (101 MHz, DMSO-d6) δ167, 167, 157, 140, 139, 135, 130, 124, 122, 121, 117, 115, 67, 53. MS (EI, 70 ev) m/z (%) = 728.40(M, 0.02), 107.15(2.50), 90.15(0.28), 77.10(5.51), 59.10(100.0 base peak).
Synthesis of 4,4′-((diselanediylbis(2-(methoxycarbonyl)-4,1-phenylene)) bis(azanediyl)) bis (4-oxobutanoic acid (11).
Compound 11 was synthesized following procedure III from the reaction of dimethyl 5,5-diselanediylbis(2-aminobenzoate) (5) (2 mmol, 916.4 mg) and succinic anhydride (4.4 mmol, 440.3 mg). It was isolated as a yellow solid; yielding: 910.8 mg (69%); MP = 150–152 °C; Rf = 0.1(petroleum ether/ethyl acetate 4:4). IR(KBr): λmax.cm−1: 3246, 2949, 1735, 1687. 1HNMR (400 MHz, DMSO-d6) δ12.22(s, 2H-OH), 10.67(s,2H, 2NH), 8.24(s, 2H, Ar-H), 8.01 (d, 2H, Ar-H), 7.82(d, J = 8.7 Hz, 2H, Ar-H), 3.78(s, 6H, OCH3), 2.64(t, 4H, 2CH2), 2.55(t, 4H, 2CH2).13CNMR (101 MHz, DMSO-d6) δ173, 170, 166, 139, 138, 134, 123, 121, 118, 52, 31, 28. MS (EI, 70 ev) m/z (%) = 462(M-198, 6.73), 460(17.67), 199(11.05), 101(21.56), 91.15(100.0, base peak).
Synthesis of dimethyl 5,5′-diselanediylbis(2-(2,5-dioxopyrrolidin-1-yl) benzoate (12).
Compound 12 was synthesized following procedure VI from the reaction of 4,4′-((diselanediylbis(2-(methoxycarbonyl)-4,1-phenylene)) bis(azanediyl)) bis (4-oxobutanoic acid (11) (0.5 mmol, 330 mg) and (5 mL) acetic anhydride. It was isolated as light-yellow solid; yielding: 121.68 mg (39%); MP = 100 °C; Rf = 0.4(DCM/MeOH 5%). IR(KBr): λmax.cm−1: 3261, 2950, 1684, 1572. 1HNMR (400 MHz, DMSO-d6) δ8.17(s, 2H, Ar-H), 8.01(d, J = 25.0 Hz, 2H, Ar-H), 7.84(d, J = 8.6 Hz, 2H, Ar-H), 3.81(s, 6H, 2OCH3), 2.14(t, 8H, 4CH2). 13CNMR (101 MHz, DMSO-d6) δ169, 167, 140, 138, 135, 124, 122, 119, 53, 25. MS (EI, 70 ev) m/z (%) = 624.20(M, 19.34), 312.15(6.43), 63.10(3.53), 59.05(3.07), 55.05 (100.0, base peak).
Synthesis of (2Z,2’Z)-4,4′-((diselanediylbis(2-(methoxycarbonyl)-4,1-phenylene)) bis(azanediyl)) bis (4-oxobut-2-enoic acid) (13).
Compound 13 was synthesized following procedure III from the reaction of dimethyl 5,5-diselanediylbis(2-aminobenzoate) (5) (2 mmol, 916.4 mg) and maleic anhydride (4.4 mmol, 433.84 mg). It was isolated as yellow solid; yielding: 983.85 mg (75 %); MP = 149.5–151 °C; Rf = 0.1(petroleum ether/ethyl acetate 4:4). IR(KBr): λmax.cm−1: 3346, 2952, 1717, 1696. 1HNMR (400 MHz, DMSO-d6) δ13.01(s, 2H, OH), 10.85(s, 2H, NH), 8.25(s, 2H, Ar-H), 8.09–7.97(d, 2H, Ar-H), 7.85(dd, J = 23.2, 12.0 Hz, 2H, Ar-H), 6.59(d, J = 11.9, 6.2 Hz, 2H, = CH), 6.36(d, J = 11.9 Hz, 2H, CH = ), 3.83(s, 6H, OCH3). 13CNMR (101 MHz, DMSO-d6) δ166, 166, 163, 139, 137, 132, 130, 128, 124, 122, 117, 51. MS (EI, 70 ev) m/z (%) = 662(M-9H, 2.03), 71.05(6.51), 67(3.36), 75(1.31), 54.05(100.0, base peak).
Synthesis of methyl 2-amino-5-(methylselanyl) benzoate (14).
Compound 14 was synthesized following procedure V from dimethyl 5,5′-diselanediylbis (2-aminobenzoate) (5) (2 mmol, 916.4 mg) and methyl iodide (4.4 mmol, 0.27 mL). It was isolated as a brown solid; yielding: 401.8 mg (82%); Rf = 0.6(petroleum ether/ethyl acetate 4:2). IR(KBr): λmax.cm−1: 3474, 2948, 1686, 1606. 1HNMR (400 MHz, DMSO-d6) δ8.25–7.65(s,1H, Ar-H), 7.65–7.20(d, 1H, Ar-H), 6.77(d, 1H, Ar-H), 6.75(s, 2H, NH2), 3.62(s, 3H, OCH3), 2.2 (s, 3H, CH3). 13CNMR (101 MHz, DMSO-d6) δ167, 151, 138, 135, 118, 113, 109, 51, 9. MS (EI, 70 ev) m/z (%) = 245.10(M, 100.0, base peak), 230(38.06), 186(10.46), 170(35.08), 91(91.10).
Synthesis of methyl 2-amino-5-(benzylselanyl) benzoate (15).
Compound 15 was synthesized following procedure V from dimethyl 5,5′-diselanediylbis(2-aminobenzoate) (5) (2 mmol, 916.4 mg) and benzyl chloride (4.4 mmol, 0.5 mL). It was isolated as a light brown solid; yielding: 597.06 mg (93%); MP = 78 °C; Rf = 0.6(petroleum ether/ethyl acetate 4:3). IR(KBr): λmax.cm−1: 3461, 3353, 2977, 1671, 1620. 1HNMR (400 MHz, DMSO-d6) δ7.81–7.65(m,1H, Ar-H), 7.30(d, J = 8.6 Hz, 1H, Ar-H), 7.23(t,2H, Ar-H), 7.19(q, 2H, Ar-H), 7.14(t, 3H, Ar-H), 6.82(s, 2H, NH2), 6.70(d, 1H, Ar-H), 3.77(s, 3H, OCH3), 1.29(s, 2H, SeCH2). 13CNMR (101 MHz, DMSO-d6) δ167, 151, 141, 139, 137, 129, 128, 126, 117, 112, 109, 51, 32. MS (EI, 70 ev) m/z (%) = 321.20(M, 46.87), 244.10(2.77), 230(34.88), 150.15(1.02), 91.05(100.0, base peak).
Synthesis of methyl 2-amino-5-((2-oxo-2-(phenylamino) ethyl) selanyl) benzoate (16).
Compound 16 was synthesized following procedure V from dimethyl 5,5′-diselanediylbis (2-aminobenzoate) (5) (2 mmol, 916 mg) and 2-chloro-N-phenylacetamide (4.4 mmol, 743.6 mg). It was isolated as a violet solid; yielding: 698.8 mg (96%); MP = 126–128.5 °C; Rf = 0.5(petroleum ether/ethyl acetate 4:2). IR(KBr): λmax.cm−1: 3466, 3246, 2945, 1678. 1H NMR (400 MHz, DMSO-d6) δ9.99(s, 1H, NH), 7.96(s, 1H, Ar-H), 7.58–7.44(t, 3H, Ar-H), 7.30(q, 2H, Ar-H), 7.04(t, 1H, Ar-H), 6.86(d, 2H, NH2), 6.75(s, 1H, Ar-H), 3.74 (s, 3H, OCH3), 3.4(s, 2H, SeCH2). 13CNMR (101 MHz, DMSO-d6) δ168, 167, 151, 141, 139, 138, 129, 123, 119, 117, 111, 109, 51, 32. MS (EI, 70 ev) m/z (%) = 364.20(M, 100.0, base peak), 230(40.48), 91.10(31), 77.05(24.13), 59(14.56).

2.3. The Biological Assays

The OSe candidates were prepared in DMSO stock solution (10 mM) and kept at −20 °C for further use.

2.3.1. The Anticancer Activity

The anticancer activity of the OSe was performed using the MTT assay against liver (HepG2) and breast (MCF-7) carcinoma cells, as well as normal WI-38 cells following the reported method [14,15,16]. Experimental details can be found in the Supplementary Materials Section S1.

2.3.2. The Antimicrobial Activity

According to the reported method, the OSe agents’ antimicrobial properties were estimated against Staphylococcus aureus (S. aureus), Escherichia coli (E. coli), and Candida albicans (C. albicans) microbial strains using the agar well diffusion assay [16,17]. In addition, the MICs (in M) were also recorded via the microdilution method following the reported procedure. Experimental details can be found in the Supplementary Materials Section S1 [16,18].

2.3.3. The Antioxidant Activity

The DPPH and ABTS in vitro bioassays were used to assess the OSe antioxidant activities following the reported method [16,19,20,21]. Experimental details can be found in the Supplementary Materials Section S1.

3. Results and Discussion

3.1. Synthesis

Recently, OSe2 compounds have drawn much attention in pharmaceutical chemistry owing to their broad medicinal applications (e.g., antioxidant, chemopreventive, and anticancer activities) [22,23,24,25]. Unluckily, the synthesis of OSe2 compounds is limited by using toxic, expensive, and air-sensitive reagents (e.g., potassium selenocyanate) and commercially available selenium precursors. Furthermore, poor functional-group tolerance and sophisticated reaction conditions (e.g., under inert gas) are significant drawbacks of synthesizing the OSe2 compounds [15,26]. Accordingly, synthesizing new and stable OSe2 synthons is required to develop potential libraries for biological testing. Moreover, anthranilic acid derivatives manifested diverse pharmacological applications and were used to prepare different marketed drugs [11,12,27]. Therefore, combining the diselenide functionality into the backbone of anthranilic acid will enable access to unprecedented organic candidates designed to interfere with biotargets. The compound 2-amino-5-selenocyanatobenzoic acid (3) [13] drew our attention and was employed as a start to our synthetic approach. Compound 3 was synthesized by the selenocyanation of 2-aminobenzoic acid (1) [13]. However, our attempts to synthesize the diselenide functionality via hydrolysis or reduction of the selenocyanate group were unsuccessful, and we could not isolate any products. This is attributed to the zwitterionic nature of compound 3, which arises from the presence of the carboxylic group in the ortho position to the amino group, which in turn causes difficulties in product isolation.
Furthermore, compound 3 is characterized by its low solubility in most organic solvents and high polarity limited its synthetic applications. Therefore, our synthetic strategy was oriented to mask the carboxylic or the amino group to overcome these difficulties. Accordingly, the alcoholic esterification of compound 3 furnished the respective methyl 2-amino-5-selenocyanatobenzoate (4); however, in low yield (25%). Therefore, we used an alternative synthetic approach to increase the yield starting from methyl 2-aminobenzoate (2) instead of 2-aminobenzoic acid (1). In this case, selenocyanation proceeded smoothly, and the yield was improved to 96% (Scheme 1). As a result, the methyl 2-amino-5-selenocyanatobenzoate (4) is new and features good solubility in most organic solvents and lower polarity compared to the corresponding 2-amino-5-selenocyanatobenzoic acid (3).
In addition, the hydrolysis of 4 using NaOH in EtOH afforded the corresponding diselenide 5 in 92% yield, thus, giving access to the symmetrical diselenide scaffolds known for their hepatoprotective activities (Scheme 1). The spectral data were used to identify the chemical structure of the diselenide 5. Compound 5 exhibited a characteristic absorption band at 3455 cm−1 and 3344 cm−1 for NH2. The 1HNMR spectrum of compound 5 showed two singlet signals at δ7.00 ppm and δ3.74 ppm related to the proton of NH2 and OCH3, respectively. The MS spectrum of compound 5 showed molecular ion peaks at 460.15(M+H, 20.76) and the base peak at m/z 91. Similarly, bisdiazotization of the diselenide 5 and subsequent coupling with two equivalents of ethyl cyanoacetate afforded the corresponding disdiazo-based diselenide 6 in 81% yield (Scheme 2). Furthermore, the reaction of diselenide 5 with acetic formic anhydride, acetic anhydride, chloroacetyl chloride, and phenoxy acetyl chloride afforded the corresponding diselenide-based formamide 7, acetanilide 8, chloroacetamide 9, and phenoxy acetamide 10 were obtained in 51%, 85%, 93%, and 55% yields, respectively (Scheme 2). Based on its spectrum data, the structure of compound 7 was established. The IR showed distinctive peaks at 3255 cm−1 for the NH and 1681 cm−1, and 1573 cm−1 for carbonyl groups. The 1HNMR spectrum of compound 8 showed three singlet signals at δ10.42 ppm for NH, at δ8.39 ppm related to the proton of the formyl group, and at δ3.75 ppm for OCH3. Its MS showed a molecular ion peak at 516.10(M, 25.78) and a base peak at m/z 198.
Moreover, the reaction of the diselenide 5 with different succinic and maleic anhydrides afforded the corresponding diselenide-based succinanilic 11 and mealanilic acids 13 in 69% and 75% yields, respectively (Scheme 3). Additionally, warming the diselenide-based succinanilic 11 with acetic anhydride afforded the corresponding cyclic succinimide 12 via dehydration and subsequent cyclization, however, in low yield (39%) (Scheme 3). Unfortunately, our attempts to perform the same reaction with the diselenide-based mealanilic acid 13 were unsuccessful, and the starting material decomposition was often observed (Scheme 3).
Eventually, the one-pot alkaline reduction of the diselenide 5 began employing a mixture of NaBH4 and NaOH (1:1) in MeOH and subsequent reaction with alkyl halides, namely methyl iodide, benzyl chloride, and 2-chloro-N-phenylacetamide afforded the corresponding organoselenides 14, 15, and 16 in 82%, 93%, and 96% yields, respectively (Scheme 3). The IR spectra of compound 14 demonstrated characteristic absorption bands of NH2 at 3474 cm−1, 3363 cm−1, and 1686 cm−1 for the C=O. The 1HNMR spectrum of compound 14 exhibited singlet signals at δ6.75 ppm for NH2, singlet signal at δ3.62 ppm for OCH3, and singlet signal at δ2.2 ppm for the SeCH3. The mass spectrum coincided with the predicted molecular mass for the proposed structure at m/z 245.10.

3.2. Biology

3.2.1. Cytotoxicity of OSe2-tethered Anthranilic Acid Hybrids

OSe hybrids have recently gained much interest owing to their unprecedented cytoprotective and antioxidant activities [1,28,29,30]. We recently reported different OSe compounds with interesting antimicrobial, anticancer, and antioxidant activities [14,23,24,31,32]. We reported different tetrazole-based diselenides within this context with AChE inhibition and glutathione peroxidase-like activities [23]. Furthermore, we also developed diselenide-based urea with promising anti-HepG2 and apoptosis-induction properties [14]. On the other hand, we identified novel organoselenium-based pseudopeptides as promising highly effective chemo-sensitizers in treating HCC with cisplatin [26].
Accordingly, the anticancer properties of the OSe hybrids were assessed against two cancer cell lines, i.e., HepG2 and MCF-7 cells. Furthermore, their corresponding cytotoxicity was also estimated against the immortalized lung WI-38 fibroblasts employing the MTT assay. The Adriamycin cancer drug was used as the positive control. The concentration inhibition, 50%, needed to kill half of the cells (IC50) was estimated from (concentration-response plots) and tabulated in Table 1. Furthermore, the safety and selectivity of drugs are assessed from their respective therapeutic indices (TI), defined as the ratio between the IC50 exhibited by the compound against WI38 cells to the compound’s respective IC50 against cancer cells, i.e., HepG2 and MCF-7 cells (Table 1).
Interestingly, the cytotoxicity was more notable in the HepG2 cells compared to the MCF-7 cells. For instance, the OSe 14 was more cytotoxic (IC50 = 3.57 ± 0.1 µM) than the standard anticancer drug Adriamycin (IC50 = 4.50 ± 0.2 µM). Furthermore, OSe 5 and 15 showed interesting anti-HepG2 cytotoxic patterns with IC50 = 7.08 ± 0.6 µM and 6.48 ± 0.4 µM, respectively (Table 1). Moreover, moderate cytotoxicity was observed in the case of OSe 4 and 6 with IC50 = 11.21 ± 1.0 µM and 16.22 ± 1.2 µM, respectively.
In the case of MCF-7 cells, the OSe 14 and 5 showed comparable cytotoxicity (IC50 = 5.64 ± 0.3 µM and 9.89 ± 0.7 µM) to Adriamycin (IC50 = 4.17 ± 0.2 µM), whereas moderate cytotoxicity was noted by the OSe 15 and 6 with IC50 = 12.76 ± 0.9 µM and 15.92 ± 1.2 µM (Table 1).
Drugs with high TI values kill cancer cells, leaving normal cells unaffected. Therefore, these drugs are highly preferable in chemotherapy, and the high TI values point to the potential selectivity and safety of a specific drug candidate [18,33,34,35,36]. Within this context, better TI values were noticed in the case of HepG2 cells compared to the MCF-7 cells. The best selective cytotoxicity patterns for HepG2 cells were observed in the case of OSe compounds 14 and 5 with TI values of 17 and 5. On the other hand, OSe compounds 14, 5, and 6 showed modest selective cytotoxicity patterns for MCF-7 cells with TI values of 11, 3.6, and 2.7 (Table 1). Eventually, such interesting selective anticancer activity merits more investigations using a broader panel of normal and cancerous cells and in vivo experiments.

3.2.2. Evaluation of the Antimicrobial Activities of the OSe Compounds

The promising anticancer activities manifested by the OSe compounds encourage us to further evaluate their respective antimicrobial activity against a panel of gram-negative bacteria (e.g., E. coli) and the gram-positive bacteria (e.g., S. aureus) as well as a fungal strain (C. albicans) using the method of agar diffusion. The clotrimazole antifungal and the ampicillin antibacterial drugs were used as the positive controls. The zones of inhibition diameters (ZID) (in mm) and the activity index percentage (IA%) are shown in Table 2.
In general, C. Albicans fungus and S. aureus gram-positive bacteria were more sensitive than gram-negative bacteria toward the OSe compounds. Interestingly, OSe 14 showed similar antifungal activity against C. albicans to the standard drug clotrimazole (IA% = 100%) and exciting antibacterial activity against E. coli (IA% = 91.3%) and S. aureus (IA% = 90.5%). Accordingly, the minimum inhibitory concentration (MIC) of 14 was also estimated (Table 3) to confirm its antimicrobial activity. In this regard, 14 manifested similar antimicrobial activity to the standard drugs clotrimazole and ampicillin drugs with MIC = of 2, 2, and 0.5 μM against C. albicans, S. aureus, and E. coli strains, respectively.
Similarly, OSe compounds 15 and 5 have shown excellent antimicrobial activities with A% of 91.7% and 87.5% against C. albicans, 85.7% and 85.7% against S. aureus, and 73.9% and 82.6% against E. coli. Additionally, good antimicrobial activities were also observed in the case of OSe compounds 6 and 4, with IA% of 70.8% and 75% against C. albicans, 76.2% and 76.2% against S. aureus, and 69.6% and 60.9% against E. coli (Table 2). Ultimately, such interesting antimicrobial patterns are worth further research and screening against an extensive panel of bacterial and fungal strains.

3.2.3. The Antioxidant Properties of the OSe Compounds

The redox modulation activities of the OSe compounds were extensively explored over the last decade owing to their chemopreventive and antioxidant potency [7,37]. The latter is usually investigated by the ABTS and DPPH in vitro tests employing vitamin C as the positive control [21,38]. The OSe compound’s magnitude estimated the antioxidant potential to decolorize the distinctive colors of the DPPH. and ABTS. radicals at 517 nm and 734 nm, respectively (Figure 2).
As displayed in Figure 2, OSe compounds 14, 5, 15, 6, and 4 exhibited 96%, 92%, 89%, 85%, and 81% scavenging activities compared to 95% by vitamin C in the DPPH and assay, respectively. Similarly, OSe compounds 14, 15, 5, 4, and 6 exhibited 91%, 88%, 86%, 73%, and 69% scavenging activities compared to 88% by vitamin C in the DPPH and assay, respectively. These results encourage us to further determine the lowest dose needed to decrease the absorbance by 50% in the ABTS and DPPH assays for the most exciting and active OSe compound 14 (Table 4). To conclude, OSe compounds 4, 6, 5, 14, and 15 exhibited promising antioxidant activity, and these results are in good accordance with the antimicrobial and anticancer effects, ensuring these compounds’ potential biological activities.

4. Conclusions

Sixteen new diselenide-tethered methyl anthranilate hybrids were synthesized in excellent yields (up to 96%), and their chemical structures were elucidated using different spectroscopic techniques. Their antimicrobial and antitumor activities were assessed against various microbial strains and cancer cells. The OSe 14 showed the best antifungal properties with action similar to the standard drug clotrimazole (IA% = 100%) and manifested promising antibacterial activity against E. coli (IA% = 91.3%) and S. aureus (IA% = 90.5%). Furthermore, OSe compounds 4, 5, 6, and 15 have also shown good antimicrobial activities. Similarly, the OSe 14 was the most cytotoxic (IC50 = 3.57 ± 0.1 µM) against the HepG2 and even more than the Adriamycin drug (IC50 = 4.50 ± 0.2 µM) and showed TI of 17, pointing to selective potential and safety. In the case of MCF-7 cells, the OSe 14 and 5 showed comparable cytotoxicity (IC50 = 5.64 ± 0.3 µM and 9.89 ± 0.7 µM) to Adriamycin (IC50 = 4.17 ± 0.2 µM), whereas moderate cytotoxicity was noted by the OSe 15 and 6 with IC50 = 12.76 ± 0.9 µM and 15.92 ± 1.2 µM (Table 1).
Furthermore, OSe compounds 14 and 5 exhibited promising antioxidants with 96%, 92%, 91%, and 86% scavenging activities compared to 95% by vitamin C in the DPPH and ABTS assays, respectively. To this end, these results point to potential antimicrobial, anticancer, and antioxidant activities of the OSe 14 and warrant further studies.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/biom12121765/s1, Section S1: The biological assays; Section S2: Copies of 1H & 13CNMR spectra IR and MS.

Author Contributions

Conceptualization, S.S. and Y.S.A.-F.; methodology, S.S., Y.S.A.-F. and B.A.-A.; software, B.A.-A. and S.S.; validation, S.S., Y.S.A.-F. and B.A.-A.; formal analysis, S.S., Y.S.A.-F. and B.A.-A.; investigation, S.S., Y.S.A.-F. and B.A.-A.; resources, S.S., Y.S.A.-F. and B.A.-A.; data curation, S.S., Y.S.A.-F. and B.A.-A.; writing—original draft preparation, S.S. and B.A.-A.; writing—review and editing, B.A.-A.; visualization, B.A.-A.; supervision, S.S. and Y.S.A.-F.; project administration, S.S. and B.A.-A.; funding acquisition, S.S. and Y.S.A.-F. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Deanship of Scientific Research, Vice Presidency for Graduate Studies and Scientific Research, King Faisal University, Saudi Arabia [GRANT2024], through its KFU Research Summer initiative.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors acknowledge the the Deanship of Scientific Research, Vice Presidency for Graduate Studies and Scientific Research, King Faisal University, Saudi Arabia [GRANT2024], through its KFU Research Summer initiative.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Bevinakoppamath, S.; Saleh Ahmed, A.M.; Ramachandra, S.C.; Vishwanath, P.; Prashant, A. Chemopreventive and Anticancer Property of Selenoproteins in Obese Breast Cancer. Front. Pharmacol. 2021, 12, 618172. [Google Scholar] [CrossRef] [PubMed]
  2. Nogueira, C.W.; Barbosa, N.V.; Rocha, J.B.T. Toxicology and pharmacology of synthetic organoselenium compounds: An update. Arch. Toxicol. 2021, 95, 1179–1226. [Google Scholar] [CrossRef] [PubMed]
  3. Kalimuthu, K.; Keerthana, C.K.; Mohan, M.; Arivalagan, J.; Christyraj, J.; Firer, M.A.; Choudry, M.H.A.; Anto, R.J.; Lee, Y.J. The emerging role of selenium metabolic pathways in cancer: New therapeutic targets for cancer. J. Cell. Biochem. 2022, 123, 532–542. [Google Scholar] [CrossRef] [PubMed]
  4. Radomska, D.; Czarnomysy, R.; Radomski, D.; Bielawska, A.; Bielawski, K. Selenium as a Bioactive Micronutrient in the Human Diet and Its Cancer Chemopreventive Activity. Nutrients 2021, 13, 1649. [Google Scholar] [CrossRef]
  5. Radomska, D.; Czarnomysy, R.; Radomski, D.; Bielawski, K. Selenium compounds as novel potential anticancer agents. Int. J. Mol. Sci. 2021, 22, 1009. [Google Scholar] [CrossRef] [PubMed]
  6. Lenardão, E.J.; Santi, C.; Sancineto, L. New Frontiers in Organoselenium Compounds; Springer: Berlin/Heidelberg, Germany, 2018. [Google Scholar]
  7. Shaaban, S.; Negm, A.; Sobh, M.A.; Wessjohann, L.A. Organoselenocyanates and symmetrical diselenides redox modulators: Design, synthesis and biological evaluation. Eur. J. Med. Chem. 2015, 97, 190–201. [Google Scholar] [CrossRef] [PubMed]
  8. Shaaban, S.; Arafat, M.A.; Hamama, W.S. Vistas in the Domain of Organoselenocyanates; Michigan Publishing: Ann Arbor, MI, USA, 2014. [Google Scholar]
  9. Plano, D.; Baquedano, Y.; Moreno-Mateos, D.; Font, M.; Jimenez-Ruiz, A.; Palop, J.A.; Sanmartin, C. Selenocyanates and diselenides: A new class of potent antileishmanial agents. Eur. J. Med. Chem. 2011, 46, 3315–3323. [Google Scholar] [CrossRef]
  10. de Oliveira, I.M.; Degrandi, T.H.; Jorge, P.M.; Saffi, J.; Rosa, R.M.; Guecheva, T.N.; Henriques, J.A. Dicholesteroyl diselenide: Cytotoxicity, genotoxicity and mutagenicity in the yeast Saccharomyces cerevisiae and in Chinese hamster lung fibroblasts. Mutat. Res. Genet. Toxicol. Environ. Mutagen. 2014, 763, 1–11. [Google Scholar] [CrossRef]
  11. Prasher, P.; Sharma, M. Medicinal chemistry of anthranilic acid derivatives: A mini review. Drug Dev. Res. 2021, 82, 945–958. [Google Scholar] [CrossRef] [PubMed]
  12. Congiu, C.; Cocco, M.T.; Lilliu, V.; Onnis, V. New potential anticancer agents based on the anthranilic acid scaffold. Synthesis and evaluation of biological activity. J. Med. Chem. 2005, 48, 8245–8252. [Google Scholar] [CrossRef] [PubMed]
  13. Kachanov, V.A.; Slabko, Y.O.; Baranova, V.O.; Shilova, V.E.; Kaminskii, A.V. Triselenium dicyanide from malononitrile and selenium dioxide. One-pot synthesis of selenocyanates. Tetrahedron Lett. 2004, 45, 4461–4463. [Google Scholar] [CrossRef]
  14. El-Senduny, F.F.; Shabana, S.M.; Rösel, D.; Brabek, J.; Althagafi, I.; Angeloni, G.; Manolikakes, G.; Shaaban, S. Urea-functionalized organoselenium compounds as promising anti-HepG2 and apoptosis-inducing agents. Future Med. Chem. 2021, 13, 1655–1677. [Google Scholar] [CrossRef] [PubMed]
  15. Shaaban, S.; Zarrouk, A.; Vervandier-Fasseur, D.; Al-Faiyz, Y.S.; El-Sawy, H.; Althagafi, I.; Andreoletti, P.; Cherkaoui-Malki, M. Cytoprotective organoselenium compounds for oligodendrocytes. Arab. J. Chem. 2021, 14, 103051. [Google Scholar] [CrossRef]
  16. Sak, M.; Al-Faiyz, Y.S.; Elsawy, H.; Shaaban, S. Novel organoselenium redox modulators with potential anticancer, antimicrobial, and antioxidant activities. Antioxidants 2022, 11, 1231. [Google Scholar] [CrossRef] [PubMed]
  17. Shaaban, S.; Sasse, F.; Burkholz, T.; Jacob, C. Sulfur, selenium and tellurium pseudopeptides: Synthesis and biological evaluation. Bioorg. Med. Chem. 2014, 22, 3610–3619. [Google Scholar] [CrossRef] [PubMed]
  18. Shaaban, S.; Negm, A.; Ashmawy, A.M.; Ahmed, D.M.; Wessjohann, L.A. Combinatorial synthesis, in silico, molecular and biochemical studies of tetrazole-derived organic selenides with increased selectivity against hepatocellular carcinoma. Eur. J. Med. Chem. 2016, 122, 55–71. [Google Scholar] [CrossRef]
  19. Abdel-Motaal, M.; Almohawes, K.; Tantawy, M.A. Antimicrobial evaluation and docking study of some new substituted benzimidazole-2yl derivatives. Bioorg. Chem. 2020, 101, 103972. [Google Scholar] [CrossRef] [PubMed]
  20. Abdel-Motaal, M.; Nabil, A. Biological activity of some newly synthesized hydrazone derivatives derived from (dicyclopropylmethylene) hydrazone. Eur. Chem. Bull. 2018, 7, 280–287. [Google Scholar] [CrossRef] [Green Version]
  21. Shaaban, S.; Negm, A.; Sobh, M.A.; Wessjohann, L.A. Expeditious Entry to Functionalized Pseudo-peptidic Organoselenide Redox Modulators via Sequential Ugi/SN Methodology. Anticancer Agents Med. Chem. 2016, 16, 621–632. [Google Scholar] [CrossRef] [PubMed]
  22. Shaaban, S.; El-Lateef, H.M.A.; Khalaf, M.M.; Gouda, M.; Youssef, I. One-Pot Multicomponent Polymerization, Metal-, and Non-Metal-Catalyzed Synthesis of Organoselenium Compounds. Polymers 2022, 14, 2208. [Google Scholar] [CrossRef] [PubMed]
  23. Refaay, D.A.; Ahmed, D.M.; Mowafy, A.M.; Shaaban, S. Evaluation of novel multifunctional organoselenium compounds as potential cholinesterase inhibitors against Alzheimer’s disease. Med. Chem. Res. 2022, 31, 894–904. [Google Scholar] [CrossRef]
  24. Shaaban, S.; Negm, A.; Ibrahim, E.E.; Elrazak, A.A. Hepatocellular carcinoma chemotherapeutic agents: Efficacy and mode of action. Onc. Rev. 2014, 8, 25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Shaaban, S.; Adam, M.S.S.; El-Metwaly, N.M. Novel organoselenium-based N-mealanilic acid and its zinc (II) chelate: Catalytic, anticancer, antimicrobial, antioxidant, and computational assessments. J. Mol. Liq. 2022, 363, 119907. [Google Scholar] [CrossRef]
  26. Shaaban, S.; Shabana, S.M.; Al-Faiyz, Y.S.; Manolikakes, G.; El-Senduny, F.F. Enhancing the chemosensitivity of HepG2 cells towards cisplatin by organoselenium pseudopeptides. Bioorg. Chem. 2021, 109, 104713. [Google Scholar] [CrossRef] [PubMed]
  27. Abd El-Lateef, H.M.; Shaaban, S.; Khalaf, M.M.; Toghan, A.; Shalabi, K. Synthesis, experimental, and computational studies of water soluble anthranilic organoselenium compounds as safe corrosion inhibitors for J55 pipeline steel in acidic oilfield formation water. Colloids Surf. A Physicochem. Eng. Asp. 2021, 625, 126894. [Google Scholar] [CrossRef]
  28. Naithani, R. Organoselenium compounds in cancer chemoprevention. Mini Rev. Med. Chem. 2008, 8, 657–668. [Google Scholar] [CrossRef] [PubMed]
  29. Mugesh, G.; du Mont, W.W.; Sies, H. Chemistry of biologically important synthetic organoselenium compounds. Chem. Rev. 2001, 101, 2125–2179. [Google Scholar] [CrossRef] [PubMed]
  30. Shaaban, S.; Negm, A.; Ibrahim, E.E.; Elrazak, A.A. Chemotherapeutic agents for the treatment of hepatocellular carcinoma: Efficacy and mode of action. Oncol. Rev. 2014, 8, 246. [Google Scholar] [CrossRef] [Green Version]
  31. Gouda, M.; Ferjani, H.; Abd El-Lateef, H.M.; Khalaf, M.M.; Shaaban, S.; Yousef, T.A. A Competition between Hydrogen, Stacking, and Halogen Bonding in N-(4-((3-Methyl-1, 4-dioxo-1, 4-dihydronaphthalen-2-yl) selanyl) phenyl) acetamide: Structure, Hirshfeld Surface Analysis, 3D Energy Framework Approach, and DFT Calculation. Int. J. Mol. Sci. 2022, 23, 2716. [Google Scholar] [CrossRef] [PubMed]
  32. Shaaban, S.; Ferjani, H.; Yousef, T.; Abdel-Motaal, M. Supramolecular Self-Assembly Built by Hydrogen, Stacking and Br · Br Interactions in 4-((4-Bromobenzyl) Selanyl) Aniline: Structure, Hirshfeld Surface Analysis, 3D Energy Framework Approach and Global Reactivity Descriptors. J. Inorg. Organomet. Polym. Mater. 2022, 32, 1878–1890. [Google Scholar] [CrossRef]
  33. Shaaban, S.; Ashmawy, A.M.; Negm, A.; Wessjohann, L.A. Synthesis and biochemical studies of novel organic selenides with increased selectivity for hepatocellular carcinoma and breast adenocarcinoma. Eur. J. Med. Chem. 2019, 179, 515–526. [Google Scholar] [CrossRef] [PubMed]
  34. Metwally, M.A.; Shaaban, S.; Abdel-Wahab, B.F.; El-Hiti, G.A. 3-Acetylindoles: Synthesis, reactions and biological activities. Curr. Org. Chem. 2009, 13, 1475. [Google Scholar] [CrossRef]
  35. Shaaban, S.; Gaffer, E.H.; Alshahd, M.; Elmorsy, S.S. Cytotoxic Symmetrical Thiazolediselenides with Increased Selectivity Against MCF-7 Breast Cancer Cells. Int. J. Res. Dev. Pharm. Life Sci. 2015, 4, 1654–1668. [Google Scholar]
  36. Shaaban, S.; Gaffer, E.H.; Jabar, Y.; Elmorsy, S.S. Cytotoxic naphthalene based-symmetrical diselenides with increased selectivity against MCF-7 breast cancer cells. Int. J. Pharm. 2015, 5, 738–746. [Google Scholar]
  37. Soriano-Garcia, M. Organoselenium compounds as potential therapeutic and chemopreventive agents: A review. Curr. Med. Chem. 2004, 11, 1657–1669. [Google Scholar] [CrossRef] [PubMed]
  38. Tian, X.; Schaich, K.M. Effects of molecular structure on kinetics and dynamics of the trolox equivalent antioxidant capacity assay with ABTS+•. J. Agric. Food Chem. 2013, 61, 5511–5519. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The structure of biologically relevant organoselenium and anthranilic acid analogs.
Figure 1. The structure of biologically relevant organoselenium and anthranilic acid analogs.
Biomolecules 12 01765 g001
Scheme 1. Preparation of compounds 2, 4, and 5. Reagents and conditions: (a) anthranilic acid (1) (29.1 mmol), MeOH (60 mL), conc.H2SO4 (4 mL), reflux for 48 h; (b) SeO2 (30 mmol), malononitrile (15 mmol), DMSO (10 mL), methyl 2-aminobenzoate (12.5 mmol) (2); (c) 2-amino-5-selenocyanatobenzoic acid (3) (29.1 mmol), MeOH (60 mL), conc.H2SO4 (4 mL), reflux for 48 h; (d) methyl 2-amino-5-selenocyanatobenzoate (4) (4 mmol), NaOH (4 mmol), EtOH, room temperature (15 mL).
Scheme 1. Preparation of compounds 2, 4, and 5. Reagents and conditions: (a) anthranilic acid (1) (29.1 mmol), MeOH (60 mL), conc.H2SO4 (4 mL), reflux for 48 h; (b) SeO2 (30 mmol), malononitrile (15 mmol), DMSO (10 mL), methyl 2-aminobenzoate (12.5 mmol) (2); (c) 2-amino-5-selenocyanatobenzoic acid (3) (29.1 mmol), MeOH (60 mL), conc.H2SO4 (4 mL), reflux for 48 h; (d) methyl 2-amino-5-selenocyanatobenzoate (4) (4 mmol), NaOH (4 mmol), EtOH, room temperature (15 mL).
Biomolecules 12 01765 sch001
Scheme 2. Synthesis of diselenides 6, 7, 8, 9, and 10. Reagents and conditions: (a) compound 5 (2 mmol), HCl (3 mL), sodium nitrite (4.4 mmol), sodium acetate (2 g), H2O: EtOH (1:1), ethyl cyanoacetate (4.4 mmol); (b) THF (10 mL), acetic formic anhydride (36.3 mmol); (c) acetic anhydride (8 mL); (d) chloroacetyl chloride (4.4 mmol), K2CO3 (2 g), dry acetone (20 mL); (e) phenoxy acetyl chloride (2.2 mmol), dry ether (15 mL).
Scheme 2. Synthesis of diselenides 6, 7, 8, 9, and 10. Reagents and conditions: (a) compound 5 (2 mmol), HCl (3 mL), sodium nitrite (4.4 mmol), sodium acetate (2 g), H2O: EtOH (1:1), ethyl cyanoacetate (4.4 mmol); (b) THF (10 mL), acetic formic anhydride (36.3 mmol); (c) acetic anhydride (8 mL); (d) chloroacetyl chloride (4.4 mmol), K2CO3 (2 g), dry acetone (20 mL); (e) phenoxy acetyl chloride (2.2 mmol), dry ether (15 mL).
Biomolecules 12 01765 sch002
Scheme 3. Synthesis of diselenides 11, 12, 13, 14, 15, and 16. Reagents and conditions: (a) Diselenide 5 (2 mmol), dry toluene (15 mL), maleic or succinic anhydride (4.4 mmol), room temperature; (b) respective amide-acids (0.5 mmol), acetic anhydride (5 mL), sodium acetate (250 mg); 50–60 °C; (c) diselenide 5 (2 mmol), halo derivatives (4.4 mmol), NaBH4 (6 mmol), EtOH (25 mL), room temperature.
Scheme 3. Synthesis of diselenides 11, 12, 13, 14, 15, and 16. Reagents and conditions: (a) Diselenide 5 (2 mmol), dry toluene (15 mL), maleic or succinic anhydride (4.4 mmol), room temperature; (b) respective amide-acids (0.5 mmol), acetic anhydride (5 mL), sodium acetate (250 mg); 50–60 °C; (c) diselenide 5 (2 mmol), halo derivatives (4.4 mmol), NaBH4 (6 mmol), EtOH (25 mL), room temperature.
Biomolecules 12 01765 sch003
Figure 2. The OSe compounds redox activities assessment using the ABTS and DPPH experiments. All tests are mean ± SD. ABTS experiment: absorbance was followed at 734 nm for OSe compounds (50 µL, 1 mM in MeOH) and ABTS solution (60 µL). DPPH experiment: Absorbance was monitored after 30 min at 517 nm for OSe compounds (200 µL, 1 mM in MeOH) and DPPH (400 µL).
Figure 2. The OSe compounds redox activities assessment using the ABTS and DPPH experiments. All tests are mean ± SD. ABTS experiment: absorbance was followed at 734 nm for OSe compounds (50 µL, 1 mM in MeOH) and ABTS solution (60 µL). DPPH experiment: Absorbance was monitored after 30 min at 517 nm for OSe compounds (200 µL, 1 mM in MeOH) and DPPH (400 µL).
Biomolecules 12 01765 g002
Table 1. The antiproliferative activities of the OSe compounds.
Table 1. The antiproliferative activities of the OSe compounds.
CompoundsMCF7 aHepG2 aWI38 a
IC50 (µM) aTI cIC50 (µM) aTI cIC50 (µM) a
Adriamycin4.17 ± 0.21.64.50 ± 0.21.56.72 ± 0.5
423.45 ± 1.82.311.21 ± 1.04.852.62 ± 3.1
59.89 ± 0.73.67.08 ± 0.6535.91 ± 2.3
615.92 ± 1.22.716.22 ± 1.22.642.45 ± 2.6
779.01 ± 4.2-83.67 ± 4.4-45.73 ± 2.7
8- b-- b-- b
9- b-- b-- b
1053.49 ± 3.0-81.48 ± 4.2-38.23 ± 2.5
1155.10 ± 3.2-43.70 ± 2.6-- b
12- b-- b-- b
1376.24 ± 4.0-78.53 ± 3.9-47.03 ± 2.8
145.64 ± 0.3113.57 ± 0.11762.38 ± 3.6
1512.76 ± 0.926.48 ± 0.43.925.81 ± 1.8
1650.81 ± 2.91.430.62 ± 2.42.371.90 ± 4.1
a The MTT bioassay was used to estimate the antitumor properties. MCF-7, HepG2, and WI-38 cells were incubated for 24 h with serial concentrations of the OSe compounds. b No proliferation inhibition was observed (IC50 ˃ 100 µM); c TI is defined as the ratio between the IC50 exhibited by the compound against WI38 cells to the compound’s respective IC50 against HepG2 and MCF-7 cells.
Table 2. The antimicrobial properties of the OSe compounds.
Table 2. The antimicrobial properties of the OSe compounds.
CompoundE. coliS. aureusC. Albicans
ZID (mm) aIA% bZID (mm) aIA% bZID (mm) aIA% b
41460.91676.21875.0
51982.61885.72187.5
61669.61676.21770.8
7--314.3416.7
8------
9------
10521.7838.1520.8
11521.7733.31041.7
12------
13--419.0520.8
142191.31990.524100
151773.91885.72291.7
16626.1942.81354.2
Ampicillin2310021100--
Clotrimazole----24100
a Inhibition zones (ZID) are expressed in diameters (mm) employing disks (6 mm) soaked with 20 μM of the OSe agents. b Activity index (IA%) percentage is the ratio of the inhibition zone of the OSe compound to the inhibition zone of the positive control.
Table 3. The MIC of OSe compound 14.
Table 3. The MIC of OSe compound 14.
CompoundsMIC (μM)
E. coliS. aureusC. Albicans
140.522
Ampicillin0.51-
Clotrimazole--2
Table 4. The OSe compounds 14 redox properties.
Table 4. The OSe compounds 14 redox properties.
CompoundsIC50 (µM)
DPPHABTS
Vitamin C19.18 ± 0.1328.16 ± 0.19
1418.67 ± 0.1229.14 ± 0.20
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Al-Abdallah, B.; Al-Faiyz, Y.S.; Shaaban, S. Anticancer, Antimicrobial, and Antioxidant Activities of Organodiselenide-Tethered Methyl Anthranilates. Biomolecules 2022, 12, 1765. https://doi.org/10.3390/biom12121765

AMA Style

Al-Abdallah B, Al-Faiyz YS, Shaaban S. Anticancer, Antimicrobial, and Antioxidant Activities of Organodiselenide-Tethered Methyl Anthranilates. Biomolecules. 2022; 12(12):1765. https://doi.org/10.3390/biom12121765

Chicago/Turabian Style

Al-Abdallah, Batool, Yasair S. Al-Faiyz, and Saad Shaaban. 2022. "Anticancer, Antimicrobial, and Antioxidant Activities of Organodiselenide-Tethered Methyl Anthranilates" Biomolecules 12, no. 12: 1765. https://doi.org/10.3390/biom12121765

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop