Next Article in Journal
Angular Regioselective Synthesis of Varied Functionalized Hexahydro-1,2,4-triazolo[4,3-a]quinazolin-9-ones and Their Antiproliferative Action
Next Article in Special Issue
The Benzothiazine Core as a Novel Motif for DNA-Binding Small Molecules
Previous Article in Journal
Chiral Indolizinium Salts Derived from 2-Pyridinecarbaldehyde—First Diastereoselective Syntheses of (−)-1-epi-lentiginosine
Previous Article in Special Issue
Naphth[1,2-d]imidazoles Bioactive from β-Lapachone: Fluorescent Probes and Cytotoxic Agents to Cancer Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Novel Biologically Active N-Substituted Benzimidazole Derived Schiff Bases: Design, Synthesis, and Biological Evaluation

1
Department of Organic Chemistry, Faculty of Chemical Engineering and Technology, University of Zagreb, Marulićev trg 19, HR-10000 Zagreb, Croatia
2
KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, 3000 Leuven, Belgium
3
Pharmacology In Vitro, Selvita Ltd., Prilaz baruna Filipovića 29, HR-10000 Zagreb, Croatia
*
Author to whom correspondence should be addressed.
Molecules 2023, 28(9), 3720; https://doi.org/10.3390/molecules28093720
Submission received: 3 March 2023 / Revised: 19 April 2023 / Accepted: 21 April 2023 / Published: 25 April 2023
(This article belongs to the Special Issue Heterocycles: Design, Synthesis and Biological Evaluation)

Abstract

:
Herein, we present the design and synthesis of novel N-substituted benzimidazole-derived Schiff bases, and the evaluation of their antiviral, antibacterial, and antiproliferative activity. The impact on the biological activity of substituents placed at the N atom of the benzimidazole nuclei and the type of substituents attached at the phenyl ring were examined. All of the synthesized Schiff bases were evaluated in vitro for their antiviral activity against different viruses, antibacterial activity against a panel of bacterial strains, and antiproliferative activity on several human cancer cell lines, thus enabling the study of the structure−activity relationships. Some mild antiviral effects were noted, although at higher concentrations in comparison with the included reference drugs. Additionally, some derivatives showed a moderate antibacterial activity, with precursor 23 being broadly active against most of the tested bacterial strains. Lastly, Schiff base 40, a 4-N,N-diethylamino-2-hydroxy-substituted derivative bearing a phenyl ring at the N atom on the benzimidazole nuclei, displayed a strong antiproliferative activity against several cancer cell lines (IC50 1.1–4.4 μM). The strongest antitumoral effect was observed towards acute myeloid leukemia (HL-60).

Graphical Abstract

1. Introduction

Schiff bases comprise a very important class of organic compounds [1], widely used in organic and medicinal chemistry as biologically important structural motifs in many synthetic and semisynthetic organic compounds [2,3,4,5]. Some naturally occurring Schiff bases play important roles in several physiological processes, for example rhodopsin, a photoreceptor present in the rod cells of the retina, which is essential in vision processing [6]. Schiff bases can be easily synthesized by a condensation reaction of different amino substituted compounds with versatile aldehydes or ketones [7]. Besides the fact that Schiff bases are important building blocks in medicinal chemistry due to their broad spectrum of biological activity [8,9,10,11,12], they have also been used in coordination chemistry [13,14,15], in analytical chemistry [16], as dyes [17], as optical chemosensors [18,19,20], as polymers [21], in catalysis [22], in metallurgy and refining of metals, and as fungicidal and agrochemical compounds. Additionally, the interest of numerous scientists is focused on evaluating Schiff bases as ligands for transition metals displaying diverse biological activities, such as anticancer [23,24], antimicrobial [25,26], and antifungal activities [27].
There is growing interest in the synthesis of benzimidazole-derived Schiff bases, not only because of their significant biological activities, but also because they can easily form complexes with different metals, giving rise to a variety of complexes with interesting electronic [28,29] and biological properties [30].
Nawrocka et al. synthesized novel 2-benzimidazolyl-substituted Schiff bases that were screened for their antiproliferative activity on several cancer cell lines [31]. Another group designed and synthesized benzimidazole and 3-oxopyrimido [1,2-a]benzimidazole-derived Schiff bases being evaluated as inhibitors of lipoxygenase (LOX) and of lipid peroxidation (LPO), showing antioxidative and cytotoxic activity (Figure 1a) [32]. In another study, a series of Schiff bases bearing benzimidazole nuclei were evaluated for their antimalarial and antitrypanosomal activity [33]. Furthermore, a group of authors explored the antimicrobial activity of a series of benzimidazole-derived Schiff bases against Staphylococcus aureus and Escherichia coli (Figure 1b) [34]. The metal–Schiff-base complexes could bind ct-DNA through intercalation and were found to exhibit more potent cytotoxic effects than the widely used drug cisplatin [35]. Another group of authors developed metal complexes that were evaluated for their cytotoxic, antiparasitic, and antibacterial activity, as well as their interaction with DNA [36]. Other authors studied the DNA interaction and antiproliferative activity of two Cu(II) complexes with Schiff bases of benzimidazole [37].
Recently, we synthesized a series of novel benzimidazole-substituted Schiff bases (Figure 2a) that were tested in vitro for their antiproliferative activity. They exerted a broad spectrum antiproliferative activity on different cancer cell lines, although only at higher concentrations [38]. Compounds substituted with 4-N,N-diethylamino-2-hydroxyphenyl bearing either a methyl or a cyano group at the 5(6)-position on the benzimidazole nuclei displayed the strongest antiproliferative effect on all of the tested cell lines, with a significant concentration-dependent effect on HeLa and MCF-7 cell lines.
As a continuation, we designed and synthesized novel N-substituted benzimidazole-derived Schiff bases (Figure 2b) to explore the influence of the substituents attached at the N atom on the benzimidazole nuclei, both for their antiproliferative and antiviral activity. Our main goal was to see how the different lengths and types of aliphatic chain and the aromatic moiety attached to the nitrogen atom on the benzimidazole nuclei would influence the biological activity.

2. Results and Discussion

2.1. Chemistry

The synthesis of the novel N-substituted benzimidazole-derived Schiff bases 2845 is presented in reaction Scheme 1. For the synthesis of the targeted Schiff bases, as main precursors, corresponding N-substituted 2-aminobenzimidazoles 1724 were prepared according to the modified reaction procedures previously described by our research group. Starting from 2-chloronitrobenzene 1 or 2-chloro-4-cyanonitrobenzene 2, in the reaction of uncatalyzed microwave-assisted amination with an excess of amine, N-methyl 5, N-phenyl 6, N-isobutyl 34, and N-hexyl 7–8 substituted nitro precursors obtained in moderate yields. After reduction with SnCl2 × 2H2O in acidic media, corresponding 1,2-diamino substituted benzenes 916 were obtained. Because of the cyclocondensation with cyanogen bromide in methanol, substituted 2-aminobenzimidazoles with isobuthyl 17 and 21, methyl 18 and 22, phenyl 19 and 23, or n-hexyl 20 and 24 side chains at the N atom or cyano group placed at the 5(6)-position on the benzimidazole nuclei were prepared in moderate reaction yields.
Benzimidazole-derived Schiff bases 2845 were synthesized from N-substituted 2-aminobenzimidazoles 1724 with chosen substituted benzaldehydes, namely 4-N,N-dimethylamino-25, 2-hydroxy-4-N,N-dimethylamino-26, and 4-nitrobenzaldehyde 27, according to the previously published experimental procedure [38]. All of the Schiff bases were additionally purified either by recrystallization or by using column chromatography on silica gel with dichloromethane/methanol as an eluent to yield the targeted compounds in low to moderate reaction yields (6–68%).
The structures of all newly prepared Schiff bases 2845 were confirmed by means of 1H and 13C NMR spectroscopy and elemental analysis. Structural analysis was performed based on the chemical shifts in both the proton spectra and 13C NMR spectra, and on the values of H–H coupling constants in the proton spectra. The amination of reactants 1–2 caused the appearance of signals related to the amino group (8.12–9.90 ppm), as well as the signals for protons from isobutyl, methyl, and n-hexyl sidechains (0.87–3.41 ppm) in the structure of compounds 3–8. Reduction of the nitro group was confirmed within the singlet related to the protons from the amino group (4.44 to 5.25 ppm). The formation of the benzimidazole ring was confirmed by the disappearance of signals for the amino groups in 1,2-diamino-substituted benzenes 9–16, as well as the appearance of a singlet related to the proton of the amino group placed at position 2 on the benzimidazole nuclei (1724), which was downshifted in comparison with the singlet of the amino group in the diamino-substituted benzenes 916. The synthesis of targeted Schiff bases 2845 was established by the observation of a singlet related to the proton of the imino group at 7.98–9.67 ppm in the proton spectra, as well as a signal for the C atom of the imino group in the carbon spectra.

2.2. Biological Activity

2.2.1. Cytotoxicity and Antiviral Activity

In Table 1, the summarized results of the antiviral evaluation are depicted. For a better overview, only the derivatives showing antiviral activity against this panel of viruses are included.
The following viruses were used for evaluating the antiviral activity: different strains of human coronavirus, three influenza virus subtypes, RSV, HSV-1, yellow fever virus, Zika virus, and Sindbis virus. The results are expressed as CC50 (50% cytotoxic concentration) and EC50 (50% effective concentration) values. Overall, the N-substituted 2-aminobenzimidazoles 1724 displayed a poor antiviral activity. Substituted 2-aminobenzimidazoles bearing methyl 18 and 22 were both able to inhibit Zika virus replication in Huh-7 cells with EC50 values of 43.1 µM and 46.4 µM, respectively. Among the N-substituted benzimidazole-derived Schiff bases 2845, some derivatives showed an antiviral activity, although it was weak when compared with the standard antiviral drugs included (remdesivir, ribavirin, zanamivir, rimantadine, and BVDU). The 4-N,N-diethylamino-2-hydroxy-substituted derivative bearing a cyano and N-isobutyl side chain on the benzimidazole nuclei 31 showed a moderate activity against HCoV-NL63 in Huh-7 cells (EC50 32 μM). Compound 42, substituted with a N,N-dimethylamino and a N-hexyl side chain showed a moderate activity against HCoV-229E in HEL299 cells (EC50 34.7 μM). In summary, no outstanding antiviral effects were noted for the 4-N,N-diethylamino-2-hydroxyphenyl ring nor for the 4-N,N-dimethylaminophenyl and 4-nitrophenyl rings.

2.2.2. Antibacterial Activity

The in vitro antibacterial activity of the synthesized Schiff bases was evaluated against a panel of eight different bacterial strains. Gram-positive bacterial strains comprised S. aureus, S. pneumoniae and E. faecalis and the panel of Gram-negative bacteria consisted of E. coli, K. pneumoniae, A. baumannii, and P. aeruginosa. As reference drugs, the antibiotics ampicillin, ceftazidime, ciprofloxacin, and meropenem were included.
As presented in Table 2, the majority of derivatives lacked an antibacterial activity, while some Schiff bases showed a moderate activity against certain bacterial strains.
The N,N-dimethylamino 38 and 4-N,N-diethylamnino-2-hydroxy 41 derivatives, both substituted with a phenyl ring at the N atom on the benzimidazole nuclei, displayed activity against E. faecalis (MIC 32 μM). p-Nitro and cyano-substituted Schiff base 37 with a methyl group at the N atom showed activity against S. aureus (32 μM). N,N-dimethylamino derivative 42 with a hexyl side chain placed at the N atom demonstrated a moderate activity against S. aureus and E. faecalis (32 μM). Overall, precursor N-hexyl-2-aminobenzimidazole 23 showed the most pronounced broad spectrum of antibiotic activity against E. faecalis, E. coli efflux dell, and K. pneumoniae, with MIC values of 32 μM, and against S. aureus with an MIC value of 16 μM.

2.2.3. Antiproliferative Activity

All of the prepared N-substituted 2-aminobenzimidazoles 916 and benzimidazole-derived Schiff bases 2845 were explored for their in vitro antiproliferative activity against several human cancer cell lines. The results are presented in Table 3 as IC50 values (50% inhibitory concentration). The following human cancer cell lines were used for the evaluation of the antiproliferative activity: LN-229, glioblastoma; Capan-1, pancreatic adenocarcinoma; HCT-116, colorectal carcinoma; NCI-H460, lung carcinoma; DND-41, acute lymphoblastic leukemia; HL-60, acute myeloid leukemia; K-562, chronic myeloid leukemia; and Z-138, non-Hodgkin lymphoma cancer cells. All of the obtained results were compared to etoposide (ETP) and nocodazole (NZO) as the standard chemotherapeutic agents.
The majority of tested compounds showed low or no activity towards the selection of cancer cell lines (for clarity, some inactive 2-aminobenzimidazoles are excluded from Table 3). Among all of the tested N-substituted 2-aminobenzimidazoles, the best activity was demonstrated by n-hexyl-substituted derivative 23, which showed a moderate but broad antiproliferative activity against all of the tested cancer types.
N-phenyl-substituted 2-aminobenzimidazole 21 showed a mild but selective activity against lung carcinoma. Regarding the benzimidazole-derived Schiff bases, the most potent one was the 4-N,N-diethylamino-2-hydroxy-substituted Schiff base bearing a phenyl ring at the N atom on benzimidazole nuclei 40. This derivative displayed a pronounced antitumoral activity against Capan-1, DND-41, HL-60, and Z-138 cancer cells in the low micromolar range, with some selectivity towards the HL-60 (acute myeloid leukemia) cancer cell line (IC50 1.1 µM). In addition, derivative 40 showed moderate activity against all of the other cancer cells lines. Furthermore, the 4-N,N-dimethylamino-substituted Schiff base bearing an isobutyl side chain at the N atom 28 showed a selective antiproliferative activity against colorectal carcinoma (HCT-116). Derivative 31 substituted with 2-hydroxy and 4-N,N-diethylamino groups at the phenyl ring, as well as with an isobutyl sidechain on the N atom and a cyano group on the benzimidazole nuclei, displayed a moderate activity against three cancer cell types (IC50 = 32.8–47.6 mM). The 4-N,N-diethylamino-2-hydroxy-substituted Schiff base 41 bearing a phenyl ring at the N atom and a cyano group on the benzimidazole nuclei also proved to be moderately active against several of the tested cancer cell lines. Among the N-hexyl-substituted Schiff bases 4245, a moderate activity was observed for compound 42 substituted with a 4-N,N-dimethylamino group at the phenyl ring, as well as for compound 43, which also bears a cyano group at the benzimidazole nuclei.
The 4-N,N-diethylamino-2-hydroxy-substituted derivatives 4445 were less active in comparison with the 4-N,N-dimethylamino substituted analogues. Regarding the N-methyl-substituted Schiff bases, only the 4-N,N-diethylamino-2-hydroxy-substituted derivative bearing a cyano group showed a moderate but selective activity against DND-41 cells. When comparing all of the N-isobutyl substituted Schiff bases 2831, we can conclude that the derivative bearing both a 4-N,N-diethylamino-2-hydroxyphenyl and cyano group 31 was the most active one.
To conclude, we observed that the most significant impact on the antiproliferative activity was seen for the 4-N,N-diethylamino-2-hydroxyphenyl ring in comparison with the 4-N,N-dimethylaminophenyl and 4-nitrophenyl rings. A cyano group placed at the 5(6) position on the benzimidazole nuclei increased the antiproliferative activity, but not for the N-phenyl-substituted derivatives. The 4-N,N-diethylamino-2-hydroxy-substituted Schiff base bearing a phenyl ring at the N atom on benzimidazole nuclei 40, which showed the most promising activity among all of the tested derivatives, was chosen as a lead compound for further optimization in order to obtain more selective and potent antiproliferative agents (Figure 3).

3. Conclusions

Herein, we present the design and synthesis of novel, N-substituted benzimidazole-derived Schiff bases, bearing isobutyl, methyl, and n-hexyl sidechains or a phenyl group at the N atom on the benzimidazole nuclei as well as a 4-N,N-dimethylamino- or 4-N,N-diethylamino-2-hydroxyphenyl ring attached directly to the imino bond. Within this research, our main focus was to study the impact on the biological activity of the substituents placed on the phenyl ring and on the N atom of the benzimidazole nuclei. All Schiff bases were evaluated for their in vitro antiviral activity against a broad selection of viruses, antibiotic activity against Gram-positive and Gram-negative bacterial strains, and antiproliferative activity on a diverse panel of human cancer cell lines.
The series of newly synthesized derivatives lacked a pronounced antiviral activity against the panel of selected viruses. Schiff base 31 substituted with a 4-N,N-diethylamino-2-hydroxyphenyl and bearing a cyano and N-isobutyl side chain on the benzimidazole nuclei showed a moderate activity against HCoV-NL63 virus on Huh-7 cells, although with a low selectivity (EC50 32 μM and CC50 85.7 µM).
The majority of the tested compounds were inactive against the Gram-positive and Gram-negative bacterial strains used. A selection of Schiff bases showed a moderate activity against one and/or two bacterial strains, while the most active compound was N-hexyl-2-aminobenzimidazole 23 with a moderate but broad activity against E. faecalis, E. coli (efflux dell), and K. pneumoniae (MIC 32 μM), as well as against S. aureus (MIC 16 μM).
Additionally, all of the prepared 2-aminobenzimidazoles 1724 and Schiff bases 2845 were tested for their antiproliferative activity against several cancer cell lines. The obtained results revealed that Schiff base 31 substituted with a 4-N,N-diethylamino -2-hydroxyphenyl and with an isobutyl side chain on the N atom and a cyano group on the benzimidazole nuclei displayed a moderate activity against three cancer types (IC50 32.8–47.6 µM). In addition, among the tested N-substituted-2-aminobenzimidazoles, N-hexyl-substituted derivative 23 showed a broad but modest activity against all of the tested cancer cell lines. Furthermore, the 4-N,N-diethylamino-2-hydroxyphenyl ring as well as the phenyl ring attached to the N atom on the benzimidazole nuclei had a pronounced impact on the activity. Schiff base 40 bearing the above-mentioned substituents showed the most promising selective antiproliferative activity against Capan-1, DND-41, HL-60, and Z-138 cancer cell lines (IC50 = 1.1–4.4 μM), and a moderate activity against all of the other tested cell lines.
In conclusion, we have shown that out of the prepared N-substituted benzimidazole-derived Schiff bases, derivative 40 showed the most interesting biological potential, with a promising antiproliferative activity, making it a promising candidate for further design and optimization.

4. Materials and Methods

4.1. General Methods

All of the chemicals were purchased from commercial suppliers. Melting points were recorded on Büchi 535 melting apparatus (Büchi, Sankt Gallen, Switzerland). The 1H and 13C NMR spectra were recorded on a Varian Bruker Advance III HD 400 MHz/54 mm Ascend instrument (Bruker, Billerica, MA, USA). All of the NMR spectra were measured in DMSO-d6 solutions using TMS as an internal standard. All of the compounds were routinely checked by TLC with Merck silica gel 60F-254 glass plates and the spots were detected under UV light. Microwave-assisted synthesis was performed in a Milestone start S microwave (Milestone Srl, Sorisole, Italy) oven using quartz cuvettes under a pressure of 40 bar. Elemental analyses for carbon, hydrogen, and nitrogen were performed on a PerkineElmer 2400 elemental analyzer (Perkin-Elmer, Waltham, MA, USA, SAD). Where analyses are indicated only as symbols of elements, the analytical results obtained were within 0.4% of the theoretical value. NMR spectra of synthesized compounds are given in Supplementary Materials.

4.2. Synthesis

4.2.1. General Method for Preparation of Compounds 78

Compounds 78 were prepared using microwave irradiation, at an optimized reaction time at 170 °C with power 800 W and 40 bar pressure, from 1 or 2 in acetonitrile (10 mL) with an excess of added corresponding amine. After cooling, the resulting product was purified by column chromatography on SiO2 using dichlormethane/methanol as the eluent.

N-Hexyl-2-nitroaniline 7

7 was prepared from 1 (0.50 g, 3.2 mmol) and hexylamine (2.90 mL, 22.2 mmol) after 2 h of irradiation to yield 0.60 g (94%) of orange oil. 1H NMR (400 MHz, DMSO-d6) (δ/ppm): 8.12 (t, 1H, J = 5.07 Hz, NH), 8.06 (dd, 1H, J1 = 1.58 Hz, J2 = 8.57 Hz, Harom), 7.56–7.51 (m, 1H, Harom), 7.05 (d, 1H, J = 7.99 Hz, Harom), 6.70–7.65 (m, 1H, Harom), 3.37–3.33 (m, 2H, CH2), 1.67–1.58 (m, 2H, CH2), 1.41–1.27 (m, 6H, CH2), 0.87 (t, 3H, J = 7.06 Hz, CH3); 13C NMR (151 MHz, DMSO-d6): δ/ppm = 145.7, 137.1, 131.3, 126.7, 115.5, 115.0, 42.7, 31.4, 28.7, 26.5, 22.5, 14.4; Anal. Calcd. for C12H18N2O2: C, 64.84; H, 8.16; N, 12.60; O, 14.39. Found: C, 64.89; H, 8.23; N, 12.51; O, 14.29%.

3-N-(Hexylamino)-4-nitrobenzonitrile 8

8 was prepared from 2 (0.50 g, 2.7 mmol) and hexylamine (1.80 mL, 13.7 mmol) after 2 h of irradiation to yield 0.50 g (97%) of yellow oil. 1H NMR (400 MHz, DMSO-d6) (δ/ppm): 8.58 (t, 1H, J = 5.38 Hz, NH), 8.50 (d, 1H, J = 2.00 Hz, Harom), 7.81 (dd, 1H, J1 = 1.60 Hz, J2 = 9.08 Hz, Harom), 7.18 (d, 1H, J = 9.10 Hz, Harom), 3.41 (q, 2H, J = 6.72 Hz, CH2), 1.65–1.55 (m, 2H, CH2), 1.38–1.23 (m, 6H, CH2), 0.87 (t, 3H J = 6.70 Hz, CH3); 13C NMR (75 MHz, DMSO-d6): δ/ppm = 146.8, 130.5, 118.2, 96.0, 42.3, 30.8, 27.9, 25.8, 21.9; Anal. Calcd. for C13H17N3O2: C, 63.14; H, 6.93; N, 16.99; O, 12.94. Found: C, 63.20; H, 6.88; N, 16.91; O, 12.89%.

4.2.2. General Method for Preparation of Compounds 1516

Derivative 7 or benzonitrile derivative 8 and a solution of SnCl2 × 2H2O in MeOH and concentrated HCl were refluxed for 0.5 h. The resulting solution was treated with 20% NaOH to pH = 14. The resulting precipitate was filtered off, washed with hot ethanol, and filtered. The filtrate was evaporated at a reduced pressure and was extracted with ethyl acetate. The organic layer was dried over anhydrous MgSO4 and concentrated at a reduced pressure.

N1-Hexylbenzene-1,2-diamine 15

Compound 15 was prepared from 7 (3.52 g, 15.8 mmol), SnCl2 × 2H2O (29.70 g, 131.5 mmol), HClconc. (49 mL), and MeOH (49 mL) to yield 2.13 g (70%) of red oil. 1H NMR (600 MHz, DMSO-d6) (δ/ppm): 6.52 (dd, 1H, J1 = 1.46 Hz, J2 = 7.78 Hz, Harom), 6.48 (td, 1H, J1 = 1.48 Hz, J2 = 7.56 Hz, Harom), 6.40–6.37 (m 2H, Harom), 4.44 (bs, 2H, NH2), 4.28 (t, 1H, J = 5.18 Hz, NH), 2.99 (q, 2H, J = 6.88 Hz, CH2), 1.61–1.55 (m, 2H CH2), 1.41–1.35 (m, 2H, CH2), 1.32–1.28 (m, 4H, CH2), 0.88 (t, 3H, J = 6.98 Hz, CH3); 13C NMR (151 MHz, DMSO-d6): δ/ppm = 136.1, 135.0, 117.5, 116.5, 114.0, 109.6, 43.4, 31.2, 28.8, 26.5, 22.1, 13.9; Anal. Calcd. for C12H20N2: C, 74.95; H, 10.48; N, 14.57. Found: C, 74.88; H, 10.54; N, 14.66%.

3-Amino-4-N-(hexylamino)benzonitrile 16

Compound 16 was prepared from 8 (2.71 g, 10.9 mmol), SnCl2 × 2H2O (14.85 g, 68.8 mmol), HClconc. (29 mL), and MeOH (29 mL) to yield 1.65 g (69%) of pink powder. m.p. 157–161 °C; 1H NMR (600 MHz, DMSO-d6) (δ/ppm): 6.91 (dd, 1H, J1 = 1.90 Hz, J2 = 8.18 Hz, Harom), 6.76 (d, 1H, J = 2.00 Hz, Harom), 6.44 (d, 1H, J = 8.16 Hz, Harom), 5.32 (t, 1H, J = 5.00 Hz, NH), 4.96 (s, 2H, NH2), 3.08 (q, 2H, J = 6.42 Hz, CH2), 1.64–1.51 (m, 2H, CH2), 1.41–1.25 (m, 6H, CH2), 0.88 (t, 3H, J = 6.58 Hz, CH3); 13C NMR (75 MHz, DMSO-d6): δ/ppm = 140.5, 135.5, 123.4, 121.6, 115.2, 108.8, 96.7, 43.2, 31.6, 28.8, 26.8, 22.6, 14.4; Anal. Calcd. for C13H19N3: C, 71.85; H, 8.81; N, 19.34. Found: C, 71.94; H, 8.87; N, 19.41%.

4.2.3. General Method for Preparation of Compounds 23 and 24

BrCN was added dropwise to a solution of o-phenylenediamine or in 20 mL H2O and 5 mL acetonitrile. The reaction mixture was refluxed for 2 h and NH4OH was added to adjust to pH = 9. After cooling, the resulting precipitate was filtered off.

2-Amino-1-hexylbenzimidazole 23

Compound 23 was prepared from 15 (1.59 g, 8.3 mmol) and BrCN (0.88 g, 8.3 mmol) to to yield 1.45 g (81%) of brown powder. m.p. 245–250 °C; 1H NMR (600 MHz, DMSO-d6) (δ/ppm): 7.96 (bs, 2H, NH2), 7.43–7.41 (m, 1H, Harom), 7.32–7.30 (m, 1H, Harom), 7.18–7.14 (m, 2H, Harom), 4.08 (t, 2H, J = 7.37 Hz, CH2), 1.68–1.63 (m, 2H, CH2), 1.33–1.24 (m, 6H, CH2), 0.84 (t, 3H, J = 7.07 Hz, CH3); 13C NMR (151 MHz, DMSO-d6): δ/ppm = 151.6, 131.9, 122.9, 121.9, 112.9, 109.9, 42.4, 31.3, 28.2, 26.0, 22.5, 14.3; Anal. Calcd. for C13H19N3: C, 71.85; H, 8.81; N, 19.34. Found: C, 71.87; H, 8.79; N, 19.29%.

2-Amino-6-cyano-1-hexylbenzimidazole 24

Compound 24 was prepared from 16 (0.03 g, 1.4 mmol) and BrCN (0.14 g, 1.4 mmol) to to yield 0.28 g (83%) of grey powder. m.p. 215–220 °C; 1H NMR (600 MHz, DMSO-d6) (δ/ppm): 7.49 (s, 1H, Harom), 7.33–7,28 (m, 2H, Harom), 6.84 (s, 1H, Harom), 4.01 (t, 2H, J = 7.18 Hz, CH2), 1.63–1.58 (m, 2H, CH2), 1.27–1.22 (m, 6H, CH2), 0.83 (t, 3H, J = 6.68 Hz, CH3); 13C NMR (151 MHz, DMSO-d6): δ/ppm = 157.1, 143.2, 138.3, 121.2, 102.5, 42.1, 31.4, 28.7, 26.1, 22.5; Anal. Calcd. for C14H18N4: C, 69.39; H, 7.49; N, 23.12. Found: C, 69.45; H, 7.40; N, 23.07%.

4.2.4. General Method for Preparation of Schiff Bases 2845

Solutions of the equimolar amounts of the corresponding N-substituted-2-aminobenzimidazole and aromatic aldehyde in absolute ethanol were refluxed for 24–48 h. After cooling, the obtained products were filtered off and recrystallized from ethanol. If necessary, the products were purified by column chromatography on SiO2 using a gradient elution of dichloromethane/methanol/TEA. Basic TEA was used to prevent the decomposition of the Schiff base conjugates in the silica gel column.

(E)-4-(((1-Isobutyl-1H-benzo[d]imidazol-2-yl)imino)methyl)-N,N-dimethylaniline 28

Compound 28 was prepared from 2-amino-1-isobutylbenzimidazole 17 (0.10 g, 0.5 mmol) and 4-N,N-dimethylamino-benzaldehyde 25 (0.08 g, 0.5 mmol) in absolute ethanol (3 mL) after refluxing for 48 h to obtain 0.02 g (14%) of yellow powder. m.p. 277–279 °C; 1H NMR (600 MHz, DMSO-d6) (δ/ppm): 9.26 (s, 1H, Harom), 7.91 (d, 2H, J = 8.79 Hz, Harom), 7.55–7.50 (m, 2H, Harom), 7.19–7.14 (m, 2H, Harom), 6.85 (d, 2H, J = 8.88 Hz, Harom), 4.17 (d, 2H, J = 7.27 Hz, CH2), 3.07 (s, 6H, CH3), 2.24–2.16 (m, 1H, CH), 0.89 (t, 6H, J = 6.66 Hz, CH3); 13C NMR (151 MHz, DMSO-d6): δ/ppm = 150.4, 131.2 (2C), 129.1 (2C), 124.0, 123.4, 112.0 (2C), 111.2 (3C), 49.1, 27.7, 19.7 (2C); Anal. Calcd. for C20H24N4: C, 74.97; H, 7.55; N, 17.48. Found: C, 74.95; H, 7.59; N, 17.41%.

(E)-2-((4-(Dimethylamino)benzylidene)amino)-1-isobutyl-1H-benzo[d]imidazole-6-carbonitrile 29

Compound 29 was prepared from 2-amino-6-cyano-1-isobutylbenzimidazole 21 (0.10 g, 0.5 mmol) and 4-N,N-dimethylamino-benzaldehyde 25 (0.07 g, 0.5 mmol) in absolute ethanol (4 mL) after refluxing for 24 h to obtain 0.03 g (22%) of yellow powder in the form of a mixture of E- and Z-isomers at a ratio of 29a/29b = 5:3. m.p. 283–285 °C; 29a: 1H NMR (400 MHz, DMSO-d6) (δ/ppm): 9.28 (s, 1H, Harom), 8.03 (d, 1H, J = 1.01 Hz, Harom), 7.94 (d, 2H, J = 8.78 Hz, Harom), 7.74 (d, 1H, J = 8.37 Hz, Harom), 7.69 (d, 1H, J = 9.02 Hz, Harom), 7.57 (dd, 1H, J1 = 8.31, J2 = 1.38 Hz, Harom), 6.85 (d, 2H, J = 9.03 Hz, Harom), 4.21 (d, 2H, J = 7.27 Hz, CH2), 3.08 (s, 6H, CH3), 2.23–2.16 (m, 1H, CH), 0.89 (d, 6H, J = 6.66 Hz, CH3); 13C NMR (100 MHz, DMSO-d6) (δ/ppm): 166.2, 159.0, 154.3, 141.6, 138.7, 132.6, 125.2, 122.7, 120.6, 112.1, 104.1, 49.8, 29.3, 20.3; 29b: 1H NMR (400 MHz, DMSO-d6) (δ/ppm): 9.67 (s, 1H, Harom), 7.69 (d, 1H, J = 9.01 Hz, Harom), 7.49 (d, 1H, J = 1.09 Hz, Harom), 7.33 (d, 1H, J = 8.07 Hz, Harom), 7.28 (dd, 1H, J1 = 8.11, J2 = 1.37 Hz, Harom), 6.84–6.78 (m, 3H, Harom), 3.85 (d, 2H, J = 7.56 Hz, CH2), 3.05 (s, 6H, CH3), 2.13–2.04 (m, 1H, CH), 0.86 (d, 6H, J = 6.67 Hz, CH3); 13C NMR (100 MHz, DMSO-d6) (δ/ppm): 190.3, 157.4, 143.2, 132.0, 121.2, 111.9, 111.5, 109.3, 102.4, 48.9, 28.4, 20.0; Anal. Calcd. for C21H23N5: C, 73.02; H, 6.71; N, 20.27. Found: C, 73.10; H, 6.68; N, 20.32%.

(E)-5-(Diethylamino)-2-(((1-isobutyl-1H-benzo[d]imidazol-2-yl)imino)methyl)phenol 30

Compound 30 was prepared from 2-amino-1-isobutylbenzimidazole 17 (0.15 g, 0.8 mmol) and 4-N,N-diethylamino-2-hydroxybenzaldehyde 26 (0.12 g, 0.8 mmol) in absolute ethanol (6 mL) after refluxing for 48 h to obtain 0.10 g (36%) of yellow powder. m.p. 254–258 °C; 1H NMR (300 MHz, DMSO-d6) (δ/ppm): 12.79 (s, 1H, OH), 9.35 (s, 1H, Harom), 7.55–7.50 (m, 2H, Harom), 7.42 (d, 1H, J = 8.97 Hz, Harom), 7.20–7.15 (m, 2H, Harom), 6.42 (dd, 1H, J1 = 2.27 Hz, J2 = 8.87 Hz, Harom), 6.17 (d, 1H, J = 2.09 Hz, Harom), 4.06 (d, 2H, J = 7.18 Hz, CH2), 3.46 (q, 4H, J = 7.08, CH2), 2.22–2.12 (m, 1H, CH), 1.12 (t, 6H, J = 7.09, CH3), 0.91 (d, 6H, J = 6.55 Hz, CH3); 13C NMR (151 MHz, DMSO-d6): δ/ppm = 163.5, 153.8, 152.9, 135.0, 121.8, 121.3, 117.9, 111.2, 110.0, 108.5, 105.0, 104.4, 96.6, 95.9, 49.4, 44.1 (2C), 28.9, 19.9 (2C), 12.5 (2C); Anal. Calcd. for C22H28N4O: C, 72.50; H, 7.74; N, 15.37; O, 4.39. Found: C, 72.48; H, 7.70; N, 15.32; O, 4.44%.

(E)-2-((4-(Diethylamino)-2-hydroxybenzylidene)amino)-1-isobutyl-1H-benzo[d]imidazole-6-carbonitrile 31

Compound 31 was prepared from 2-amino-6-cyano-1-isobutylbenzimidazole 21 (0.15 g, 0.7 mmol) and 4-N,N-diethylamino-2-hydroxybenzaldehyde 26 (0.13 g, 0.7 mmol) in absolute ethanol (7 mL) after refluxing for 48 h to obtain 0.11 g (40%) of orange powder. m.p. 205–208 °C; 1H NMR (600 MHz, DMSO-d6): δ/ppm = 12.62 (s, 1H, OH), 9.37 (s, 1H, Harom), 8.01 (d, 1H, J = 1.09 Hz, Harom), 7.74 (d, 1H, J = 8.26 Hz, Harom), 7.59 (d, 1H, J = 8.97 Hz, Harom), 7.56 (dd, 1H, J1 = 1.48 Hz, J2 = 8.98 Hz, Harom), 6.44 (dd, 1H, J1 = 2.29 Hz, J2 = 8.99 Hz, Harom), 6.18 (d, 1H, J = 2.27 Hz, Harom), 4.10 (d, 2H, J = 7.36 Hz, CH2), 3.45 (q, 4H, J = 6.97 Hz, CH2), 2.20–2.12 (m, 2H, CH), 1.15 (t, 6H, J = 7.08 Hz, CH3), 0.90 (d, 6H, J = 6.69 Hz, CH3); 13C NMR (151 MHz, DMSO-d6): δ/ppm = 164.3, 153.9 (2C), 141.6, 138.7, 125.3, 122.6, 120.6, 111.8 (2C), 109.1, 105.9 (2C), 104.1, 97.0 (2C), 50.0, 44.7, 29.4, 20.3, 13.0; Anal. Calcd. for C23H27N5O: C, 70.92; H, 6.99; N, 17.98; O, 4.11. Found: C, 70.97; H, 6.91; N, 17.93; O, 4.08%.

(E)-1-Isobutyl-2-((4-nitrobenzylidene)amino)-1H-benzo[d]imidazole-6-carbonitrile 32

Compound 32 was prepared from 2-amino-6-cyano-1-isobutylbenzimidazole 21 (0.10 g, 0.5 mmol) and 4-nitrobenzaldehyde 27 (0.08 g, 0.5 mmol) in absolute ethanol (3 mL) after refluxing for 24 h to obtain 0.02 g (14%) of yellow powder. m.p. 277–279 °C; 1H NMR (300 MHz, DMSO-d6) (δ/ppm): 9.66 (s, 1H, Harom), 8.41–8.38 (m, 5H, Harom), 8.19 (d, 1H, J = 0.98 Hz, Harom), 7.88 (d, 1H, J = 8.46 Hz, Harom), 7.67 (dd, 1H, J1 = 1.47 Hz, J2 = 8.38 Hz, Harom), 4.31 (d, 2H, J = 7.28 Hz, CH2), 2.25–2.13 (m, 1H, CH), 0.90 (d, 6H, J = 6.69 Hz, CH3); 13C NMR (151 MHz, DMSO-d6) (δ/ppm): 165.2, 156.2, 149.8, 140.6, 140.2, 138.2, 130.9 (2C), 125.7, 124.2 (2C), 123.9, 119.8, 112.4, 104.6, 49.6, 29.0, 19.8 (2C); Anal. Calcd. for C19H17N5O2: C, 65.69; H, 4.93; N, 20.16; O, 9.21. Found: C, 65.72; H, 4.87; N, 20.09; O, 9.34%.

(E)-N,N-Dimethyl-4-(((1-methyl-1H-benzo[d]imidazol-2-yl)imino)methyl)aniline 33

Compound 33 was prepared from 2-amino-1-methylbenzimidazole 18 (0.10 g, 0.7 mmol) and 4-N,N-dimethylamino-benzaldehyde 25 (0.10 g, 0.7 mmol) in absolute ethanol (4 mL) after refluxing for 48 h to obtain 0.01 g (6%) of yellow powder. m.p. 186–188 °C; 1H NMR (600 MHz, DMSO-d6): δ/ppm = 7.98–7.94 (m, 2H, Harom), 7.41–7.36 (m, 2H, Harom), 7.32–7.29 (m, 2H, Harom), 7.18–7.15 (m, 3H, Harom), 3.59 (s, 6H, CH3); 13C NMR (151 MHz, DMSO-d6): δ/ppm = 190.4, 154.7, 132.0, 125.0, 123.8, 123.1, 112.2, 112.1, 111.6, 111.0, 110.4, 19.8, 9.1 (2C); Anal. Calcd. for C17H18N4: C, 71.27; H, 5.65; N, 23.09. Found: C, 71.30; H, 5.61; N, 23.16%.

(E)-2-((4-(Dimethylamino)benzylidene)amino)-1-methyl-1H-benzo[d]imidazole-6-carbonitrile 34

Compound 34 was prepared from 2-amino-6-cyano-1-methylbenzimidazole 22 (0.10 g, 0.6 mmol) and 4-N,N-dimethylamino-benzaldehyde 25 (0.09 g, 0.6 mmol) in absolute ethanol (3 mL) after refluxing for 48 h to obtain 0.05 g (27%) of yellow powder. m.p. 222–226 °C; 1H NMR (400 MHz, DMSO-d6) (δ/ppm): 9.27 (s, 1H, Harom), 8.01 (d, 1H, J = 1.26 Hz, Harom), 7.95 (d, 2H, J = 8.81 Hz, Harom), 7.68 (d, 1H, J = 8.32 Hz, Harom), 7.58 (dd, 1H, J1 = 8.28, J2 = 1.37 Hz, Harom), 6.84 (d, 2H, J = 8.91 Hz, Harom), 3.88 (s, 3H, CH3), 3.08 (s, 6H, CH3); 13C NMR (100 MHz, DMSO-d6) (δ/ppm): 166.4, 158.9, 154.3, 141.7, 139.0, 132.8, 125.2, 122.9, 122.8, 122.6, 120.7, 118.2, 112.0, 111.6, 108.8, 104.1, 29.5 (2C), 29.1; Anal. Calcd. for C18H17N5: C, 71.27; H, 5.65; N, 23.09. Found: C, 71.30; H, 5.61; N, 23.16%.

(E)-5-(Diethylamino)-2-(((1-methyl-1H-benzo[d]imidazol-2-yl)imino)methyl)phenol 35

Compound 35 was prepared from 2-amino-1-methylbenzimidazole 18 (0.15 g, 1.0 mmol) and 4-N,N-diethylamino-2-hydroxybenzaldehyde 26 (0.19 g, 1.0 mmol) in absolute ethanol (7 mL) after refluxing for 24 h to obtain 0.04 g (13%) of orange powder. m.p. 169–173 °C; 1H NMR (600 MHz, DMSO-d6): δ/ppm = 12.61 (s, 1H, OH), 9.36 (s, 1H, Harom), 7.58 (d, 1H, J = 8.87 Hz, Harom), 7.54–7.51 (m, 1H, Harom), 7.50–7.47 (m, 1H, Harom), 7.21–7.15 (m, 2H, Harom), 6.42 (dd, 1H, J1 = 2.38 Hz, J2 = 8.99 Hz, Harom), 6.16 (d, 1H, J = 2.26 Hz, Harom), 3.78 (s, 3H, CH3), 3.45 (q, 4H, J = 7.08 Hz, CH2), 1.15 (t, 6H, J = 7.07 Hz, CH3); 13C NMR (151 MHz, DMSO-d6): δ/ppm = 164.5, 163.9, 153.4, 141.9 (2C), 135.8, 122.3, 121.7, 118.3, 110.2, 109.1, 105.5, 97.0, 44.6 (2C), 29.1, 13.0 (2C); Anal. Calcd. for C19H22N4O: C, 70.78; H, 6.88; N, 17.38; O, 4.96. Found: C, 70.71; H, 6.79; N, 17.44; O, 5.03%.

(E)-2-((4-(Diethylamino)-2-hydroxybenzylidene)amino)-1-methyl-1H-benzo[d]imidazole-6-carbonitrile 36

Compound 36 was prepared from 2-amino-6-cyano-1-methylbenzimidazole 22 (0.20 g, 1.2 mmol) and 4-N,N-diethylamino-2-hydroxybenzaldehyde 26 (0.22 g, 1.2 mmol) in absolute ethanol (10 mL) after refluxing for 48 h to obtain 0.27 g (67%) of yellow powder. m.p. 227–231 °C; 1H NMR (400 MHz, DMSO-d6) (δ/ppm): 12.40 (s, 1H, OH), 9.37 (s, 1H, Harom), 7.99 (d, 1H, J = 1.08 Hz, Harom), 7.68 (d, 1H, J = 8.29 Hz, Harom), 7.61 (d, 1H, J = 8.97 Hz, Harom), 7.57 (dd, 1H, J1 = 1.45 Hz, J2 = 8.27 Hz, Harom), 6.43 (dd, 1H, J1 = 2.36 Hz, J2 = 9.00 Hz, Harom), 6.15 (d, 1H, J = 2.26 Hz, Harom), 3.80 (s, 3H, CH3), 3.45 (q, 4H, J = 6.98 Hz, CH2), 1.15 (t, 6H, J = 6.95 Hz, CH3); 13C NMR (100 MHz, DMSO-d6) (δ/ppm): 165.3, 164.2, 157.6, 153.9, 141.7, 138.9, 125.2, 122.4, 120.7, 111.5, 109.2, 105.9, 104.1, 96.9, 44.7 (2C), 29.5, 13.0 (2C); Anal. Calcd. for C20H21N5O: C, 69.14; H, 6.09; N, 20.16; O, 4.61. Found: C, 69.19; H, 5.98; N, 20.13; O, 4.71%.

(E)-1-Methyl-2-((4-nitrobenzylidene)amino)-1H-benzo[d]imidazole-6-carbonitrile 37

Compound 37 was prepared from 2-amino-6-cyano-1-methylbenzimidazole 22 (0.10 g, 0.5 mmol) and 4-nitrobenzaldehyde 27 (0.08 g, 0.5 mmol) in absolute ethanol (3 mL) after refluxing for 24 h to obtain 0.02 g (14%) of yellow powder. m.p. 277–279 °C; 1H NMR (600 MHz, DMSO-d6) (δ/ppm): 9.67 (s, 1H, Harom), 8.44–8.41 (m, 4H, Harom), 8.20 (d, 1H, J = 0.97 Hz, Harom), 7.83 (d, 1H, J = 8.38 Hz, Harom), 7.69 (dd, 1H, J1 = 1.49 Hz, J2 = 8.37 Hz, Harom), 3.99 (s, 3H, CH3); 13C NMR (151 MHz, DMSO-d6): δ/ppm = 165.9, 156.7, 150.4, 141.2, 140.7, 139.1, 131.5 (2C), 126.2, 124.7 (2C), 124.3, 120.3, 112.6, 105.1, 29.9; Anal. Calcd. for C16H11N5O2: C, 62.95; H, 3.63; N, 22.94; O, 10.48. Found: C, 62.91; H, 3.69; N, 22.88; O, 10.41%.

(E)-N,N-Dimethyl-4-(((1-phenyl-1H-benzo[d]imidazol-2-yl)imino)methyl)aniline 38

Compound 38 was prepared from 2-amino-1-phenylbenzimidazole 19 (0.10 g, 0.5 mmol) and 4-N,N-dimethylamino-benzaldehyde 25 (0.07 g, 0.5 mmol) in absolute ethanol (3 mL) after refluxing for 48 h to obtain 0.11 g (68%) of yellow powder. m.p. 204–207 °C; 1H NMR (400 MHz, DMSO-d6) (δ/ppm): 9.29 (s, 1H, Harom), 7.76 (d, 2H, J = 8.86 Hz, Harom), 1.66–1.57 (m, 5H, Harom), 1.54–1.49 (m, 1H, Harom), 7.30 (d, 1H, J = 7.89 Hz, Harom), 7.28–7.24 (m, 1H, Harom), 7.22–7.17 (m, 1H, Harom), 6.79 (d, 2H, J = 8.87 Hz, Harom), 3.04 (s, 6H, CH3); 13C NMR (151 MHz, DMSO-d6): δ/ppm = 165.5, 156.2, 153.9, 142.1, 135.9, 135.6, 132.3, 129.7 (2C), 128.2, 127.4 (2C), 123.1, 122.9, 122.6, 118.9, 112.0, 110.4; Anal. Calcd. for C22H20N4: C, 77.62; H, 5.92; N, 16.46. Found: C, 77.68; H, 5.98; N, 16.39%.

(E)-2-((4-(Dimethylamino)benzylidene)amino)-1-phenyl-1H-benzo[d]imidazole-6-carbonitrile 39

Compound 39 was prepared from 2-amino-6-cyano-1-phenylbenzimidazole 23 (0.13 g, 0.6 mmol) and 4-N,N-dimethylamino-benzaldehyde 25 (0.09 g, 0.6 mmol) in absolute ethanol (6 mL) after refluxing for 24 h to obtain 0.04 g (17%) of yellow powder. m.p. 247–251 °C; 1H NMR (600 MHz, DMSO-d6) (δ/ppm): 9.29 (s, 1H, Harom), 8.14 (d, 1H, J = 1.09 Hz, Harom), 7.75 (d, 2H, J = 8.97 Hz, Harom), 7.66–7.62 (m, 2H, Harom), 7.62–7.59 (m, 2H, Harom), 7.57 (dd, 1H, J1 = 1.47 Hz, J2 = 8.28 Hz, Harom), 7.56–7.54 (m, 1H, Harom), 7.41 (d, 1H, J = 8.27 Hz, Harom), 6.78 (d, 2H, J = 8.98 Hz, Harom), 3.05 (s, 6H, CH3); 13C NMR (151 MHz, DMSO-d6): δ/ppm = 167.1, 158.6, 154.4, 141.9, 138.6, 135.0, 129.8 (2C), 128.8, 127.5 (2C), 126.1, 123.2, 122.6, 120.4, 112.0, 111.7, 105.0; Anal. Calcd. for C23H19N5: C, 75.59; H, 5.24; N, 19.16. Found: C, 75.65; H, 5.30; N, 19.09%.

(E)-5-(Diethylamino)-2-(((1-phenyl-1H-benzo[d]imidazol-2-yl)imino)methyl)phenol 40

Compound 40 was prepared from 2-amino-1-phenylbenzimidazole 19 (0.10 g, 0.5 mmol) and 4-N,N-diethylamino-2-hydroxybenzaldehyde 26 (0.09 g, 0.5 mmol) in absolute ethanol (7 mL) after refluxing for 48 h to obtain 0.10 g (52%) of yellow powder. m.p. 197–201 °C; 1H NMR (400 MHz, DMSO-d6): δ/ppm = 12.40 (s, 1H, OH), 9.37 (s, 1H, Harom), 7.67–7.62 (m, 3H, Harom), 7.60–7.53 (m, 3H, Harom), 7.50 (d, 1H, J = 8.97 Hz, Harom), 7.29–7.22 (m, 2H, Harom), 7.21–7.17 (m, 1H, Harom), 6.39 (dd, 1H, J1 = 2.37 Hz, J2 = 8.99 Hz, Harom), 6.04 (d, 1H, J = 2.27 Hz, Harom), 3.41 (q, 4H, J = 7.54 Hz, CH2), 1.11 (t, 6H, J = 6.88 Hz, CH3); 13C NMR (75 MHz, DMSO-d6) (δ/ppm): 164.4, 163.5, 153.0, 141.6, 135.6, 135.1, 135.0, 129.7 (2C), 128.2, 126.9 (2C), 122.6, 122.1, 118.3, 109.7, 108.5, 105.0, 96.5, 44.1, 12.5; Anal. Calcd. for C24H24N4O: C, 74.97; H, 6.29; N, 14.57; O, 4.16. Found: C, 74.89; H, 6.19; N, 14.51; O, 4.20%.

(E)-2-((4-(Diethylamino)-2-hydroxybenzylidene)amino)-1-phenyl-1H-benzo[d]imidazole-6-carbonitrile 41

Compound 41 was prepared from 2-amino-6-cyano-1-phenylbenzimidazole 23 (0.20 g, 0.9 mmol) and 4-N,N-diethylamino-2-hydroxybenzaldehyde 26 (0.16 g, 0.9 mmol) in absolute ethanol (10 mL) after refluxing for 48 h to obtain 0.23 g (66%) of yellow powder. m.p. 111–114 °C; 1H NMR (400 MHz, DMSO-d6) (δ/ppm): 12.28 (bs, 1H, OH), 9.37 (s, 1H, Harom), 8.13–8.11 (m, 1H, Harom), 7.68–7.64 (m, 2H, Harom), 7.61–7.59 (m, 3H, Harom), 7.56 (dd, 1H, J1 = 8.28 Hz, J2 = 1.56 Hz, Harom), 7.51 (d, 1H, J = 9.09 Hz, Harom), 7.35 (d, 1H, J = 8.37 Hz, Harom), 6.40 (dd, 1H, J1 = 9.08 Hz, J2 = 2.40 Hz, Harom), 6.04 (d, 1H, J = 2.28 Hz, Harom), 3.44–3.39 (m, 1H, CH2), 1.11 (t, 6H, J = 7.01 Hz, CH3); 13C NMR (100 MHz, DMSO-d6) (δ/ppm): 165.8, 164.4, 156.8, 154.0, 142.0, 138.6, 136.4, 134.7, 130.3, 129.3, 127.8, 126.2, 122.9, 120.4, 111.5, 109.1, 105.9, 105.0, 96.9, 44.7 (2C), 13.0 (2C); Anal. Calcd. for C25H23N5O: C, 73.33; H, 5.66; N, 17.10; O, 3.91. Found: C, 73.36; H, 5.69; N, 17.15; O, 4.02%.

(E)-4-(((1-Hexyl-1H-benzo[d]imidazol-2-yl)imino)methyl)-N,N-dimethylaniline 42

Compound 42 was prepared from 2-amino-1-hexylbenzimidazole 20 (0.20 g, 0.9 mmol) and 4-N,N-dimethylamino-benzaldehyde 25 (0.13 g, 0.9 mmol) in absolute ethanol (5 mL) after refluxing for 24 h to obtain 0.09 g (31%) of yellow powder in the form of a mixture of E- and Z-isomers at a ratio of 42a/42b = 3:1. m.p. 231–234 °C; 42a: 1H NMR (600 MHz, DMSO-d6) (δ/ppm): 9.26 (s, 1H, Harom), 7.91 (d, 2H, J = 8.88 Hz, Harom), 7.55–7.51 (m, 1H, Harom), 7.51–7.47 (m, 1H, Harom), 7.19–7.14 (m, 2H, Harom), 6.84 (d, 2H, J = 8.99 Hz, Harom), 4.35 (t, 2H, J = 6.88 Hz, CH2), 3.07 (s, 6H, CH3), 1.80–1.75 (m, 2H, CH2), 1.29–1.17 (m, 6H, CH2), 0.79 (t, 3H, J = 7.27 Hz, CH3); 13C NMR (151 MHz, DMSO-d6) (δ/ppm): 165.8, 153.8, 137.8, 124.7, 122.4 (2C), 122.4, 122.2, 120.1, 117.7, 111.5 (2C), 111.1 (2C), 108.4, 103.6, 41.9, 30.5, 28.9, 25.5, 21.9, 13.8; 42b: 1H NMR (600 MHz, DMSO-d6) (δ/ppm): 9.67 (s, 1H, Harom), 7.69 (d, 2H, J = 8.97 Hz, Harom), 7.35 (d, 1H, J = 8.36 Hz, Harom), 7.26 (dd, 1H, J1 = 1.98 Hz, J2 = 6.67 Hz, Harom), 7.13–7.08 (m, 2H, Harom), 6.79 (d, 2H, J = 8.86 Hz, Harom), 4.03 (t, 2H, J = 7.29 Hz, CH2), 3.05 (s, 6H, CH3), 1.67–1.62 (m, 2H, CH2), 1.29–1.17 (m, 6H, CH2), 0.84 (t, 3H, J = 6.97 Hz, CH3); 13C NMR (151 MHz, DMSO-d6) (δ/ppm): 156.6, 142.8, 141.2 (2C), 132.1, 123.0, 128.1, 122.4 (2C), 120.7, 117.7 (2C), 111.1, 111.0, 102.0, 41.6, 30.9, 28.2, 25.6, 22.0, 13.8; Anal. Calcd. for C22H28N4: C, 75.82; H, 8.10; N, 16.08. Found: C, 75.78; H, 8.16; N, 16.14%.

(E)-2-((4-(Dimethylamino)benzylidene)amino)-1-hexyl-1H-benzo[d]imidazole-6-carbonitrile 43

Compound 43 was prepared from 2-amino-6-cyano-1-hexylbenzimidazole 24 (0.15 g, 0.6 mmol) and 4-N,N-dimethylamino-benzaldehyde 25 (0.09 g, 0.6 mmol) in absolute ethanol (5 mL) after refluxing for 48 h to obtain 0.09 g (43%) of yellow powder. m.p. 165–170 °C; 1H NMR (300 MHz, DMSO-d6) (δ/ppm): 9.27 (s, 1H, Harom), 8.02 (d, 1H, J = 0.88 Hz, Harom), 7.95 (s, 1H, Harom), 7.92 (s, 1H, Harom), 7.72 (d, 1H, J = 8.37 Hz, Harom), 7.56 (dd, 1H, J1 = 1.39 Hz, J2 = 8.28 Hz, Harom), 7.30 (d, 1H, J = 1.38 Hz, Harom), 6.86–6.82 (m, 2H, Harom), 4.39 (t, 2H, J = 6.79 Hz, CH2), 3.08 (s, 6H, CH3), 1.83–1.72 (m, 2H, CH2), 1.27–1.20 (m, 6H, CH2), 0.77 (t, 3H, J = 6.97 Hz, CH3); 13C NMR (151 MHz, DMSO-d6) (δ/ppm): 165.77, 158.29, 156.63, 153.81, 142.75, 141.22, 137.80, 137.78, 132.15, 129.99, 128.12, 124.68, 122.38, 122.35, 122.20, 120.65, 120.12, 117.68, 111.64, 111.54, 111.14, 111.03, 108.41, 103.57, 101.96, 41.96, 41.60, 30.85, 30.50, 28.90, 28.23, 25.57, 25.50, 21.96, 21.90, 13.80, 13.75; Anal. Calcd. for C23H27N5: C, 73.96; H, 7.29; N, 18.75. Found: C, 74.01; H, 7.24; N, 18.70%.

(E)-5-(Diethylamino)-2-(((1-hexyl-1H-benzo[d]imidazol-2-yl)imino)methyl)phenol 44

Compound 44 was prepared from 2-amino-1-hexylbenzimidazole 20 (0.15 g, 0.7 mmol) and 4-N,N-diethylamino-2-hydroxybenzaldehyde 26 (0.13 g, 0.7 mmol) in absolute ethanol (5 mL) after refluxing for 48 h to obtain 0.03 g (11%) of yellow powder. m.p. 201–204 °C; 1H NMR (600 MHz, DMSO-d6) (δ/ppm): 12.68 (s, 1H, OH), 9.36 (s, 1H, Harom), 7.56 (d, 1H, J = 8.96 Hz, Harom), 7.54–7.48 (m, 2H, Harom), 7.21–7.13 (m, 2H, Harom), 6.42 (dd, 1H, J1 = 2.37 Hz, J2 = 8.97 Hz, Harom), 6.16 (d, 1H, J = 2.27 Hz, Harom), 4.24 (t, 2H, J = 7.07 Hz, CH3), 3.44 (q, 4H, J = 7.09 Hz, CH2), 1.78–1.73 (m, 2H, CH2), 1.29–1.20 (m, 6H, CH2), 1.15 (t, 6H, J = 7.08 Hz, CH3), 0.80 (t, 3H, J = 7.07 Hz, CH3); 13C NMR (100 MHz, DMSO-d6) (δ/ppm): 164.0, 163.9, 154.6, 154.3, 153.4, 141.9, 135.1, 122.2, 121.8, 118.4, 111.7, 110.2, 109.0, 105.5, 104.9, 97.0, 96.4, 44.6, 44.6, 42.6, 31.2, 29.6, 26.4, 22.5, 14.2, 13.0, 12.9; Anal. Calcd. for C24H32N4O: C, 73.43; H, 8.22; N, 14.27; O, 4.08. Found: C, 73.37; H, 8.09; N, 14.15; O, 4.02%.

(E)-2-((4-(Diethylamino)-2-hydroxybenzylidene)amino)-1-hexyl-1H-benzo[d]imidazole-6-carbonitrile 45

Compound 45 was prepared from 2-amino-6-cyano-1-hexylbenzimidazole 24 (0.15 g, 0.6 mmol) and 4-N,N-diethylamino-2-hydroxybenzaldehyde 26 (0.11 g, 0.6 mmol) in absolute ethanol (6 mL) after refluxing for 48 h to obtain 0.02 g (8%) of yellow powder. m.p. 233–237 °C; 1H NMR (600 MHz, DMSO-d6): δ/ppm = 12.47 (s, 1H, OH), 9.38 (s, 1H, Harom), 8.00 (d, 1H, J = 1.19 Hz, Harom), 7.72 (d, 1H, J = 8.27 Hz, Harom), 7.60 (d, 1H, J = 8.99 Hz, Harom), 7.56 (dd, 1H, J1 = 1.45 Hz, J2 = 8.27 Hz, Harom), 6.44 (dd, 1H, J1 = 2.37 Hz, J2 = 8.96 Hz, Harom), 6.16 (d, 1H, J = 2.28 Hz, Harom), 4.28 (t, 2H, J = 7.06 Hz, CH2), 3.45 (q, 4H, J = 6.98 Hz, CH2), 1.76–1.72 (m, 2H, CH2), 1.28–1.25 (m, 4H, CH2), 1.23–1.20 (m, 2H, CH2), 1.15 (t, 6H, J = 7.09 Hz, CH3), 0.80 (t, 3H, J = 7.08 Hz, CH3), 13C NMR (151 MHz, DMSO-d6): δ/ppm = 164.3, 153.9, 141.7, 138.3, 125.2, 122.6, 120.6, 111.5, 109.1, 105.9 (2C), 104.1, 96.9 (2C), 44.7 (2C), 42.9, 31.1, 29.5, 26.3, 22.4, 14.2, 13.0; Anal. Calcd. for C25H31N5O: C, 71.91; H, 7.48; N, 16.77; O, 3.83. Found: C, 71.94; H, 7.42; N, 16.71; O, 3.88%.

4.3. Biology

4.3.1. Antiviral Activity

HEL 299 (ATCC CCL-137; human lung fibroblast), Huh-7 (CLS—300156; human hepatoblastoma), and MDCK (Madin-Darby canine kidney cells; a kind gift from M. Matrosovich, Marburg, Germany) were maintained in Dulbecco’s Modified Eagle Medium (DMEM; Gibco Life Technologies) supplemented with 8% heat-inactivated fetal bovine serum (HyClone, GE Healthcare Life Sciences), 0.075% sodium bicarbonate (Gibco Life Technologies) and 1mM sodium pyruvate (Gibco Life Technologies), and maintained at 37 °C under 5% CO2. Antiviral assays towards herpes simplex virus-1 (HSV-1 KOS), human coronavirus (HCoV-229E and -OC43), and respiratory syncytial virus A in HEL 299 cell cultures; sindbis virus, yellow fever virus, Zika virus, and human coronavirus (HCoV-NL63) in Huh-7 cell cultures; and influenza A/H1N1 (A/Ned/378/05), influenza A/H3N2 (A/HK/7/87), and influenza B (B/Ned/537/05) in MDCK cell cultures were performed. On the day of the infection, the growth medium was aspirated and replaced by serial dilutions of the test compounds. The virus was then added to each well and diluted to obtain a viral input of 100 CCID50 (CCID50 being the virus dose that is able to infect 50% of the cell cultures). Mock-treated cultures receiving solely the test compounds were included in order to determine the cytotoxicity.
After 3 to 7 days of incubation, the virus-induced cytopathogenic effect was measured colorimetrically by the formazan-based MTS cell viability assay (CellTiter 96 AQueous One Solution Cell Proliferation Assay from Promega, Madison, WI), and the antiviral activity was expressed as the 50% effective concentration (EC50). In parallel, the 50% cytotoxic concentration (CC50) was derived from the mock-infected cells. The activities were compared with the activities of the reference antiviral drugs: remdesivir, ribavirin, zanamivir, rimantadine, and brivudine (BVDU).

4.3.2. Antibacterial Activity

Materials

In addition to the synthesized compounds, standard antibiotics ampicillin, ceftazidime, ciprofloxacin, and meropenem from USP were tested. Selected bacterial strains were Gram-negative E. coli, K. pneumoniae, A. baumannii and P. aeruginosa and Gram-positive S. aureus, S. pneumoniae, and E. faecalis. The synthesized compounds were prepared as 10 mM DMSO solutions and tested in a final concentration range of 0.2–100 µM [39]. Standard antibiotics were prepared as 5 mg/mL DMSO solutions and tested in a final concentration range of 0.125–64 µg/mL.

Methods

Broth microdilution testing was performed according to CLSI (Clinical Laboratory Standards Institute) guidelines. The MIC (minimal inhibitory concentration) value was defined as the last tested concentration of the compound at which there was no visible growth of bacteria. Inoculums for each microorganism were prepared using the direct colony suspension method, where broth solutions that achieved turbidity equivalent to 0.5 McFarland standard were additionally diluted 100× with Ca adjusted MH media (Becton Dickinson, Franklin Lakes, NJ, USA). All of the test plates were incubated for 16–24 h at 37 °C.
MIC values for reference antibiotics against quality control strains were used for confirming the validity of the screen according to the Clinical and Laboratory Standards Institute (CLSI) guidelines. Methods for dilution of antimicrobial susceptibility tests for bacteria that grow aerobically followed M07, 11th edition, 2018, and Clinical and Laboratory Standards Institute (CLSI) guidelines. Performance standards for antimicrobial susceptibility testing followed the M100, 28th edition, 2018.

4.3.3. Cell Culture and Reference Compounds

Human cancer cells used in this manuscript, namely Capan-1, HCT-116, NCI-H460, LN-229, HL-60, K-562, and Z-138, were acquired from the American Type Culture Collection (ATCC, Manassas, VA, USA), while the DND-41 cell line was purchased from the Deutsche Sammlung von Mikroorganismen und Zellkulturen (DSMZ Leibniz-Institut, Braunschweig, Germany). Culture media were purchased from Gibco Life Technologies, Merelbeke, Belgium, and supplemented with 10% fetal bovine serum (HyClone, Cytiva, Marlborough, MA, USA). Vincristine and docetaxel, which were used as the reference inhibitors, were purchased from Selleckchem (Munich, Germany). Stock solutions were prepared in DMSO.

4.3.4. Proliferation Assays

Adherent cell lines LN-229, HCT-116, and NCI-H460 and Capan-1 cells were seeded at a density between 500 and 1500 cells per well, in 384-well tissue culture plates (Greiner, Kremsmünster, Austria). After overnight incubation, the cells were treated with seven different concentrations of the test compounds, ranging from 100 to 0.006 µM.
Suspension cell lines HL-60, K-562, Z-138, and DND-41 were seeded at densities ranging from 2500 to 5500 cells per well in 384-well culture plates containing the test compounds at the same concentration points. The cells were incubated for 72 h with the compounds and were then analyzed using the CellTiter 96® AQueous One Solution Cell Proliferation Assay (MTS) reagent (Promega, Madison, WI, USA) according to the manufacturer’s instructions. The absorbance of the samples was measured at 490 nm using a SpectraMax Plus 384 (Molecular Devices, Silicon Valley, CA, USA), and OD values were used to calculate the 50% inhibitory concentration (IC50). The compounds were tested in two independent experiments.

Supplementary Materials

The following are available online at https://www.mdpi.com/article/10.3390/molecules28093720/s1. Pages S2–S5: 13C NMR data and elemental analysis of prepared compounds. Pages S5–S7: Biological experimental data. Figures S1–S48: NMR spectra of the prepared compounds.

Author Contributions

Synthesis and characterization of targeted compounds A.B. and M.C.; antiproliferative activity in vitro L.P. and D.D.; antibacterial activity in vitro V.R. and M.B.; antiviral activity in vitro L.P. and D.D.; writing—original draft preparation L.P., D.D. and M.H. All authors have read and agreed to the published version of the manuscript.

Funding

We greatly appreciate the financial support of the Croatian Science Foundation under project 4379, titled Exploring the antioxidative potential of benzazole scaffold in the design of novel antitumor agents.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

D.D. and L.P. are grateful for the excellent technical assistance of J. Punjwani, N. Van Winkel, N. Willems, and Y. Smolders.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

Sample Availability

Samples of the compounds are not available from the authors.

References

  1. Raczuk, E.; Dmochowska, B.; Samaszko-Fiertek, J.; Madaj, J. Different Schiff Bases-Structure, Importance and Classification. Molecules 2022, 27, 787. [Google Scholar] [CrossRef] [PubMed]
  2. Tidwell, T.T. Hugo (Ugo) Schiff, Schiff bases, and a century of β-lactam synthesis. Angew. Chem. Int. Ed. 2008, 47, 1016–1020. [Google Scholar] [CrossRef] [PubMed]
  3. Arulmurugan, S.; Kavitha, H.P.; Venkatraman, B.R. Biological activities of Schiff base and its complexes: A review. Rasayan J. Chem. 2010, 3, 385–410. [Google Scholar]
  4. Qin, W.L.; Long, S.; Panunzio, M.; Biondi, S. Schiff bases: A short survey on anevergreen chemistry tool. Molecules 2013, 18, 12264–12289. [Google Scholar] [CrossRef]
  5. Zoubi, W.A. Biological Activities of Schiff Bases and Their Complexes: A Review of Recent Works. Int. J. Org. Chem. 2013, 3, 73–95. [Google Scholar] [CrossRef]
  6. Lenahan, C.; Sanghavi, R.; Huang, L.; Zhang, J.H. Rhodopsin: A Potential Biomarker for Neurodegenerative Diseases. Front. Neurosci. 2020, 14, 326. [Google Scholar] [CrossRef]
  7. Carey, F.A. Organic Chemistry, 5th ed.; MacGraw-Hill: New York, NY, USA, 2003; p. 724. [Google Scholar]
  8. Anand, P.; Patil, V.M.; Sharma, V.K.; Khosa, R.L.; Masand, N. Schiff bases: A review on biological insights. Int. J. Drug Design Disc. 2012, 3, 851–865. [Google Scholar]
  9. Şener, N.; Özkinali, S.; Altunoglu, Y.C.; Yerlikaya, S.; Gökçe, H.; Zurnaci, M.; Gür, M.; Baloglu, M.C.; Şener, İ. Antiproliferative properties and structural analysis of newly synthesized Schiff bases bearing pyrazole derivatives and molecular docking studies. J. Mol. Struct. 2021, 1241, 130520. [Google Scholar] [CrossRef]
  10. Sztanke, K.; Maziarka, A.; Osinka, A.; Sztanke, M. An insight into synthetic Schiff bases revealing antiproliferative activities in vitro. Bioorg. Med. Chem. 2013, 21, 3648–3666. [Google Scholar] [CrossRef]
  11. Vicini, P.; Geronikaki, A.; Incerti, M.; Busonera, B.; Poni, G.; Cabras, C.A.; la Colla, P. Synthesis and biological evaluation of benzo[d]isothiazole, benzothiazole and thiazole Schiff bases. Bioorg. Med. Chem. 2003, 11, 4785–4789. [Google Scholar] [CrossRef]
  12. Jos, S.; Suja, N.R. Chiral Schiff base ligands of salicylaldehyde: A versatile tool for medical applications and organic synthesis—A review. Inorg. Chim. Acta 2023, 547, 121323. [Google Scholar] [CrossRef]
  13. Cozzi, P.G. Metal-Salen Schiff base complexes in catalysis: Practical aspects. Chem. Soc. Rev. 2004, 33, 410–421. [Google Scholar] [CrossRef] [PubMed]
  14. Ashraf, T.; Ali, B.; Qayyum, H.; Haroone, M.S.; Shabbir, G. Pharmacological aspects of schiff base metal complexes: A critical review. Inorg. Chem. Comm. 2023, 150, 110449. [Google Scholar] [CrossRef]
  15. Saloutin, V.I.; Edilova, Y.O.; Kudyakova, Y.S.; Burgart, Y.V.; Bazhin, D.N. Heterometallic Molecular Architectures Based on Fluorinated β-Diketone Ligands. Molecules 2022, 27, 7894. [Google Scholar] [CrossRef] [PubMed]
  16. Cimerman, Z.; Snežana, M.; Nives, G. Schiff bases derived from aminopyridines as spectrofluorimetric analytical reagents. Croat. Chem. Acta 2000, 73, 85–96. [Google Scholar]
  17. Gupta, K.C.; Sutar, A.K. Catalytic activities of Schiff base transition metalcomplexes. Coord. Chem. Rev. 2008, 252, 1420–1450. [Google Scholar] [CrossRef]
  18. Rauf, A.; Shah, A.; Khan, A.A.; Shah, A.H.; Abbasi, R.; Qureshi, I.Z.; Ali, S. Synthesis, pH dependent photometric and electrochemical investigation, redox mechanism and biological applications of novel Schiff base and its metallic derivatives. Spectrochim. Acta A Mol. Biomol. Spectrosc. 2017, 176, 155–167. [Google Scholar] [CrossRef] [PubMed]
  19. Zhang, S.H.; Feng, C. Microwave-assisted synthesis, crystal structure and fluorescence of novel coordination complexes with Schiff base ligands. J. Mol. Struct. 2010, 977, 62–66. [Google Scholar] [CrossRef]
  20. Horak, E.; Kassal, P.; Hranjec, M.; Murković Steinberg, I. Benzimidazole functionalised Schiff bases: Novel pH sensitivefluorescence turn-on chromoionophores for ion-selective optodes. Sens. Act. B 2018, 258, 415–423. [Google Scholar] [CrossRef]
  21. Castillo-Martnez, E.; Carretero-Gonzlez, J.; Armand, M. Polymeric Schiff Bases as Low-Voltage Redox Centers for Sodium-Ion Batteries. Angew. Chem. Int. Ed. 2014, 53, 5341–5345. [Google Scholar] [CrossRef]
  22. Abdel Hameed, R.S. Schiff’ Bases as Corrosion Inhibitor for Aluminum Alloy in Hydrochloric Acid Medium. Tenside Surf. Det. 2019, 56, 3–10. [Google Scholar] [CrossRef]
  23. Gama, S.; Mendes, F.; Marques, F.; Santos, I.C.; Carvalho, M.F.; Correia, I.; Pessoa, J.C.; Santos, I.; Paulo, A. Copper(II) complexes with tridentate pyrazole-based ligands: Synthesis, characterization, DNA cleavage activity and cytotoxicity. J. Inorg. Biochem. 2011, 105, 637–644. [Google Scholar] [CrossRef]
  24. Katwal, R.; Kaur, H.; Kapur, B.K. Applications of copper—Schiff’s base complexes: A review. Sci. Rev. Chem. Commun. 2013, 3, 1–15. [Google Scholar]
  25. Aiyelabola, T.; Jordaan, J.; Otto, D.; Akinkunmi, E. Syntheses, Characterization, Antimicrobial Activity and Extraction Studies of Tetraaza Macrocyclic/Linear Schiff Bases Derived from Benzene-1,4-Dicarboxaldehyde and Their Coordination Compounds. Adv. Biolog. Chem. 2021, 11, 79–105. [Google Scholar] [CrossRef]
  26. Fonkui, T.Y.; Ikhile, M.I.; Ndinteh, D.T.; Njobeh, P.B. Microbial activity of some heterocyclic Schiff bases and metal complexes: A review. Trop. J. Pharm. Res. 2018, 17, 2507–2518. [Google Scholar] [CrossRef]
  27. Ugras, H.I.; Basaran, I.; Kilic, T.; Cakir, U. Synthesis, Complexation and Antifungal, Antibacterial Activity Studies of a New Macrocyclic Schiff Base. J. Het. Chem. 2006, 43, 1679–1684. [Google Scholar] [CrossRef]
  28. Singh, H.L. Synthesis and characterization of tin (II) complexes of fluorinated Schiff bases derived from amino acids. Spect. Acta Part A 2010, 76, 253–258. [Google Scholar] [CrossRef] [PubMed]
  29. Halder, S.; Bhattacharjee, A.; Roy, A.; Chatterjee, S.; Roy, P. Chromogenic andfluorescence sensing of pH with a Schiff-base molecule. RSC Adv. 2016, 6, 39118–39124. [Google Scholar] [CrossRef]
  30. Hameed Haddad, H. A New Schiff Base Derivatives Designed to Bind Metal Ion (Cu, Co): Thermodynamics and Biological Activity Studies. J. Anal. Chem. 2016, 7, 445–451. [Google Scholar] [CrossRef]
  31. Nawrocka, W.; Sztuba, B.; Kowalska, M.W.; Liszkiewicz, H.; Wietrzyk, J.; Nasulewicz, A.; Pełczynska, M.; Opolski, A. Synthesis and Antiproliferative Activity in vitro of New 2-Aminobenzimidazole Derivatives Part 2 [1]. Farmaco 2004, 59, 1047–1055. [Google Scholar] [CrossRef]
  32. Neochoritis, C.G.; Zarganes-Tzitzikas, T.; Tsoleridis, C.A.; Stephanidou-Stephanatou, J.; Kontogiorgis, C.A.; Hadjipavlou-Litina, D.J.; Choli-Papadopoulou, T. One-pot microwave assisted synthesis under green chemistry conditions, antioxidant screening, and cytotoxicity assessments of benzimidazole Schiff bases and pyrimido[1,2-a]benzimidazol-3(4H)-ones. Eur. J. Med. Chem. 2011, 46, 297–306. [Google Scholar] [CrossRef] [PubMed]
  33. Fonkui, T.Y.; Ikhile, M.I.; Njobeh, P.B.; Ndinteh, D.T. Benzimidazole Schiff base derivatives: Synthesis, characterization and antimicrobial activity. BMC Chem. 2019, 13, 127–138. [Google Scholar] [CrossRef] [PubMed]
  34. Alam, S.A.M.F.; Ahmad, T.; Nazmuzzaman, M.; Ray, S.K.; Sharifuzzaman, M.; Karim, M.R.; Alam, M.G.; Ajam, M.M.; Maitra, P.; Mandol, D.; et al. Synthesis of Benzimidazole Derivatives Containing Schiff Base Exhibiting Antimicrobial Activities. Int. J. Res. Stud. Biosci. 2017, 5, 18–24. [Google Scholar]
  35. Kumaravel, G.; Utthra, P.P.; Raman, N. Exploiting the biological efficacy of benzimidazole based Schiff base complexes with L-Histidine as a co-ligand: Combined molecular docking, DNA interaction, antimicrobial and cytotoxic studies. Bioorg. Chem. 2018, 77, 269–279. [Google Scholar] [CrossRef]
  36. Aragón-Muriel, A.; Liscano, Y.; Upegui, Y.; Robledo, S.M.; Ramírez-Apan, M.T.; Morales-Morales, D.; Oñate-Garzón, J.; Polo-Cerón, D. In Vitro Evaluation of the Potential Pharmacological Activity and Molecular Targets of New Benzimidazole-Based Schiff Base Metal Complexes. Antibiotics 2021, 10, 728. [Google Scholar] [CrossRef]
  37. Song, W.-J.; Cheng, J.-P.; Jiang, D.-H.; Guo, L.; Cai, M.-F.; Yang, H.-B.; Lin, Q.-Y. Synthesis, interaction with DNA and antiproliferative activities of twonovel Cu(II) complexes with Schiff base of benzimidazole. Spect. Acta Part A Mol. Biomol. Spectr. 2014, 121, 70–76. [Google Scholar] [CrossRef] [PubMed]
  38. Hranjec, M.; Starčević, K.; Kraljević Pavelić, S.; Lučin, P.; Pavelić, K.; Karminski Zamola, G. Synthesis, spectroscopic characterization and antiproliferative evaluation in vitro of novel Schiff bases related to benzimidazoles. Eur. J. Med. Chem. 2011, 46, 2274–2279. [Google Scholar] [CrossRef]
  39. Grgičević, I.; Mikulandra, I.; Bukvić, M.; Banjanac, M.; Radovanović, V.; Habinovec, I.; Bertoša, B.; Novak, P. Discovery of macrozones, new antimicrobial thiosemicarbazone-based azithromycin conjugates: Design, synthesis and in vitro biological evaluation. Int. J. Antimicrob. Agents 2020, 56, 106147. [Google Scholar] [CrossRef]
Figure 1. Examples of previously synthesized Schiff bases. (a) benzimidazole Schiff bases; (b) phenyl-benzimidazol-4-yl Schiff bases and (c) phenyl-benzimidazol-3-yl Schiff bases.
Figure 1. Examples of previously synthesized Schiff bases. (a) benzimidazole Schiff bases; (b) phenyl-benzimidazol-4-yl Schiff bases and (c) phenyl-benzimidazol-3-yl Schiff bases.
Molecules 28 03720 g001
Figure 2. Synthesized benzimidazole-derived Schiff bases.
Figure 2. Synthesized benzimidazole-derived Schiff bases.
Molecules 28 03720 g002
Scheme 1. Synthesis of N-substituted benzimidazole-derived Schiff bases 2845.
Scheme 1. Synthesis of N-substituted benzimidazole-derived Schiff bases 2845.
Molecules 28 03720 sch001
Figure 3. Selected Schiff base 40 as a lead compound for further optimization.
Figure 3. Selected Schiff base 40 as a lead compound for further optimization.
Molecules 28 03720 g003
Table 1. Cytotoxicity and antiviral activity of the synthesized Schiff bases.
Table 1. Cytotoxicity and antiviral activity of the synthesized Schiff bases.
CpdCytotoxicity (CC50/µM)Antiviral Activity (EC50/μM)
HEL 229Huh-7MDCKHCoV
229E
HEL 299
HCoV
OC43
HEL 299
HCoV
NL63
Huh-7
Influenza
H1N1
MDCK
Influenza
H3N2
MDCK
Influenza
B
MDCK
RSV
A Long
HEL 299
HSV-1
KOS
HEL 299
YFV
17D
Huh-7
Zika
Mr776
Huh-7
Sindbis
Huh-7
17>100>100>10078.9>100>100>100>100>100>100>100>100>100>100
18>100>100>100>100>100>100>100>100>100>100>100>10043.1>100
22>100>100>100>100>100>100>100>100>10078.7>100>10046.4>100
29>100>100>100>100>10079.894.1>100>100>100>100>100>100>100
31>10085.71.3>100>10032>100>100>100>100>100>100>100>100
32>10090.1>10038.568.7>100>100>100>100>100>100>100>100>100
34>100>100>100>100>10082.8>100>10083.5>100>100>100>100>100
36>100>100>10088.4>100>100>100>100>100>100>100>100>100>100
38>100>100>100>10094.8>100>100>100>100>100>100>100>100>100
41>100>10071.6>10089.5>100>100>100>100>100>100>100>100>100
42>1002.734.634.7>100>100>100>100>100>100>100>100>100>100
Remdesivir>10>10-0.060.060.03---0.03-6.20.7>10
Ribavirin>2508.967.082.6170.1>25010.54.02.810.8->250>250148.1
Zanamivir-->100---0.116.80.05-----
Rimantadine-->100---5.00.05>100-----
BVDU>100---------0.05---
Table 2. Antibacterial activity of the synthesized Schiff bases.
Table 2. Antibacterial activity of the synthesized Schiff bases.
CpdS. aureus ATCC 29213E. faecalis
ATCC 29212
S. pneumoniae ATCC 49619E. coli
ATCC 25922
E. coli
efflux del
P. aeruginosa ATCC 27853K. pneumoniae ATCC 700603A. baumannii ATCC 17978
231632326432>64>6464
37326464>64>64>64>64>64
38>6432>64>64>64>64>64>64
41>643264>64>64>64>64>64
42323264>6464>64>64>64
Ampicillin0.522<0.1250.5>64>64>64
Ceftazidime8<0.1250.250.5>6416432
Ciprofloxacin0.125<0.125<0.1250.510.250.254
Meropenem<0.125<0.125<0.125<0.125810.25<0.125
Table 3. Antiproliferative activity of the synthesized Schiff bases against a broad panel of cancer cell lines.
Table 3. Antiproliferative activity of the synthesized Schiff bases against a broad panel of cancer cell lines.
CpdIC50 (μM)
LN-229Capan-1HCT-116NCI-H460DND-41HL-60K-562Z-138
19>100>100>100>10098.3>100>100>100
21>100>100>10069.2>100>100>100>100
2345.246.557.460.247.744.946.744.7
28>100>10015.2>100>100>100>100>100
29>100>100>100>100>10042.8>100>100
30>100>100>100>100>10070.4>100>100
31>10032.8>100>10034.447.6>100>100
32>100>100>100>100>100>100>100>100
33>100>100>100>100>100>100>100>100
34>100>100>100>100>100>100>100>100
35>100>100>100>100>100>100>100>100
36>100>100>100>10041.4>100>100>100
37>100>100>100>100>100>100>100>100
38>100>100>100>100>100>100>100>100
39>100>100>100>100>100>100>100>100
4021.52.49.643.72.21.112.94.4
41>10069.5>100>10076.349.6>10035.2
42>100>100>10064.4>10073.290.8>100
43>100>100>10091.6>10033.5>100>100
44>100>100>100>100>10083.9>100>100
45>100>100>100>100>100>100>100>100
ETP2.400.431.453.652.800.421.770.85
NZO0.290.100.130.250.470.100.070.31
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Beč, A.; Cindrić, M.; Persoons, L.; Banjanac, M.; Radovanović, V.; Daelemans, D.; Hranjec, M. Novel Biologically Active N-Substituted Benzimidazole Derived Schiff Bases: Design, Synthesis, and Biological Evaluation. Molecules 2023, 28, 3720. https://doi.org/10.3390/molecules28093720

AMA Style

Beč A, Cindrić M, Persoons L, Banjanac M, Radovanović V, Daelemans D, Hranjec M. Novel Biologically Active N-Substituted Benzimidazole Derived Schiff Bases: Design, Synthesis, and Biological Evaluation. Molecules. 2023; 28(9):3720. https://doi.org/10.3390/molecules28093720

Chicago/Turabian Style

Beč, Anja, Maja Cindrić, Leentje Persoons, Mihailo Banjanac, Vedrana Radovanović, Dirk Daelemans, and Marijana Hranjec. 2023. "Novel Biologically Active N-Substituted Benzimidazole Derived Schiff Bases: Design, Synthesis, and Biological Evaluation" Molecules 28, no. 9: 3720. https://doi.org/10.3390/molecules28093720

Article Metrics

Back to TopTop