Next Article in Journal
The Prevalence of the Virulence Genes of Staphylococcus aureus in Sickle Cell Disease Patients at KSUMC, Riyadh, Saudi Arabia
Previous Article in Journal
Do or Don’t: Results of a Multinational Survey on Antibiotic Prophylaxis in Urodynamics
Previous Article in Special Issue
Chloramphenicol Derivatization in Its Primary Hydroxyl Group with Basic Amino Acids Leads to New Pharmacophores with High Antimicrobial Activity
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Benzimidazole-Triazole Hybrids as Antimicrobial and Antiviral Agents: A Systematic Review

Department of Organic Chemistry, Biochemistry and Catalysis, Faculty of Chemistry, University of Bucharest, 030018 Bucharest, Romania
Antibiotics 2023, 12(7), 1220; https://doi.org/10.3390/antibiotics12071220
Submission received: 30 June 2023 / Revised: 14 July 2023 / Accepted: 19 July 2023 / Published: 22 July 2023
(This article belongs to the Special Issue Design and Synthesis of Antimicrobial Compounds)

Abstract

:
Bacterial infections have attracted the attention of researchers in recent decades, especially due to the special problems they have faced, such as their increasing diversity and resistance to antibiotic treatment. The emergence and development of the SARS-CoV-2 infection stimulated even more research to find new structures with antimicrobial and antiviral properties. Among the heterocyclic compounds with remarkable therapeutic properties, benzimidazoles, and triazoles stand out, possessing antimicrobial, antiviral, antitumor, anti-Alzheimer, anti-inflammatory, analgesic, antidiabetic, or anti-ulcer activities. In addition, the literature of the last decade reports benzimidazole-triazole hybrids with improved biological properties compared to the properties of simple mono-heterocyclic compounds. This review aims to provide an update on the synthesis methods of these hybrids, along with their antimicrobial and antiviral activities, as well as the structure–activity relationship reported in the literature. It was found that the presence of certain groups grafted onto the benzimidazole and/or triazole nuclei (-F, -Cl, -Br, -CF3, -NO2, -CN, -CHO, -OH, OCH3, COOCH3), as well as the presence of some heterocycles (pyridine, pyrimidine, thiazole, indole, isoxazole, thiadiazole, coumarin) increases the antimicrobial activity of benzimidazole-triazole hybrids. Also, the presence of the oxygen or sulfur atom in the bridge connecting the benzimidazole and triazole rings generally increases the antimicrobial activity of the hybrids. The literature mentions only benzimidazole-1,2,3-triazole hybrids with antiviral properties. Both for antimicrobial and antiviral hybrids, the presence of an additional triazole ring increases their biological activity, which is in agreement with the three-dimensional binding mode of compounds. This review summarizes the advances of benzimidazole triazole derivatives as potential antimicrobial and antiviral agents covering articles published from 2000 to 2023.

1. Introduction

Heterocyclic compounds have a central place in medicinal chemistry, being used as therapeutic agents to treat most diseases [1,2,3]. Among these heterocycles, benzimidazole stands out, as a purine-analog pharmacophore, with a very diverse therapeutic activity. The very broad spectrum of biological activities it treats include antimicrobial [4,5,6,7,8], antiviral [9,10], antihistamine [11,12], anticonvulsant [3,13], antitumor [14,15,16], proton pump inhibitors [17], antiparasitic [16,18,19], anti-inflammatory [20,21,22], or antihypertensive [23,24] activities. Some benzimidazoles are efficient agents in Diabetes mellitus [25,26,27], while astemizole compounds possess anti-prion activity to treat Creutzfeldt-Jakob disease [5,28]. The literature also reports anti-Alzheimer [29,30], psychoactive, anxiolytic, analgesic [31,32], and anticoagulant properties [33,34] of benzimidazole derivatives.
Additionally, triazole compounds possess a diversity of biological activities as antimicrobial [35,36,37,38], antitubercular [39,40], potential inhibitors of SARS-CoV-2 [41,42,43], antiviral [43,44], anti-inflammatory [45,46],antitumor [47,48,49,50], antihypertensive [50], antioxidant [47,51,52], and antiepileptic [53,54]. Pharmacological applications of triazoles refer to their activity as α-glucosidase inhibitors [55,56], analgesics [50,57], anticonvulsants [53,58], and antimalarial agents [57,59]. Triazole derivatives are efficient in the treatment of Alzheimer’s disease [60,61] and are very effective neuroprotective agents [62,63].
The successive events that occurred from the spring of 2020 until now, regarding the emergence and development of the COVID-19 pandemic, have led the scientific world to investigate more closely the possibility of treating this infectious disease with various antiviral [64,65,66], antimicrobial [67], immunomodulatory [68] or anti-inflammatory [69] drugs, therefore, the discovery of new molecules with simple or hybrid structures, which meet the requirements of the treatment of this condition is absolutely necessary and constitutes the engine for the development of new effective therapeutic agents.
Why did I choose the study of benzimidazole-triazole compounds? Classical drugs containing benzimidazole and triazole rings recommend these heterocycles as essential in building new target compounds with antimicrobial, antiviral, antiparasitic, etc. properties (Figure 1). In addition, the literature mentions a series of benzimidazole-triazole hybrids with remarkable antimicrobial properties, and antiviral activities, including new anti-SARS-CoV-2 agents [70,71,72,73,74], with particular importance in the context of the recent pandemic, which led to the study of synthesis methods, antimicrobial properties, structure–property relationships, and their biological activities.
Therefore, this review aims to provide an update on the synthesis methods of the benzimidazole-triazole hybrids, along with their antimicrobial and antiviral activities, as well as the structure–activity relationship and DFT studies reported in the literature. The advantages of the study of benzimidazole-triazole hybrid compounds refer to a wider range of antimicrobial activities, compared to simple precursor heterocycles, to their better minimum inhibitory concentrations compared to simple component heterocycles, as well as to the need to hire specialized personnel to carry out this research.
The main disadvantages are material because the synthesis of some hybrid compounds requires high costs compared to simple heterocycles, as well as greater time consumption. Consequently, if the synthesized hybrids have increased biological properties compared to simple precursor heterocycles, the balance clearly tilts towards the advantage of the synthesis of hybrid compounds. However, access to hybrid compounds will not be without both sides, advantages and disadvantages, which requires careful prospecting of all the components involved in the production of hybrids.
As expected, the literature mentions benzimidazole-triazole hybrids with other biological properties than those studied in this review, such as antitumor [15,48,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92], antioxidant [93,94,95], anti-Alzheimer [96,97,98,99], antidiabetic [100,101,102,103,104], and anti-inflammatory [105] properties, which is additional proof of the therapeutic potential of these hybrids and the need to study these hybrids on the topic proposed in the title. As expected, the study refers to both 1,2,3-triazole-benzimidazole hybrids and 1,2,4-triazole-benzimidazole hybrids, even if it seems that the literature is richer in the second category in terms of antimicrobial activity.
The recent literature marks several strategies for the synthesis of 1,2,3-triazoles, like click reaction [106], Bouiton-Katritzky rearrangement [107], oxidative cyclization of hydrazones [108], post-cycloaddition functionalization [109], alkylation or arylation of triazoles [110]. Also, for benzimidazoles, the literature mentions several methods of synthesis, such as the reaction of o-phenylenediamine with aldehydes or ketones (Phillips-Ladenburg reaction) [3,111,112,113], with acids or their derivatives (Weidenhagen reaction) [81], or green methods of classic syntheses [111,114,115,116,117].
Why this review is necessary and what exactly it proposes I will clarify in what follows. This article summarizes for the first time in the literature: various synthesis methods of benzimidazole-1,2,3-triazole hybrids as well as benzimidazole-1,2,4-triazoles, their antimicrobial and antiviral activities, as well as SAR studies and DFT performed on the mentioned hybrids. Where necessary, for compounds with superior biological activities, several examples from the literature were given, and the various studies performed on them (in vitro, in vivo, in silico, etc.) were mentioned. All of these aim at directing the syntheses of hybrid compounds with specific structures and superior antimicrobial and antiviral properties, taking into account the mentions reported in the literature up to now.
The database search methodology used in this review was the use of keywords, which can be found in the title, such as benzimidazole, 1,2,3-triazole, click reaction, 1,2,3-triazole, benzimidazole-triazole hybrids, antimicrobial, antiviral, or therapeutic properties, in different websites, such as PubMed, MDPI, Science Direct, Springer, The Royal Society Chemistry, ACS Publications, and Taylor & Francis. The selection of scientific articles for the last ten years was made according to the novelty brought in the benzimidazole-triazole hybrids and their antimicrobial and antiviral properties, as well as the therapeutic properties of the reported compounds.
Generally, articles from the last ten years have been selected. For the hybrids found, first, the syntheses and then their biological properties were presented, with special emphasis on those with improved properties (active on a larger range of microbial strains, with better minimum inhibitory concentrations, or where SAR studies were performed, DFT, etc.). In the following, we will present syntheses of benzimidazole-triazole hybrids with antimicrobial and antiviral properties. In order to highlight the structures of the heterocycles in the discussed compounds, we colored the benzimidazole nucleus with red, 1,2,3-triazole with blue, and 1,2,4-triazole with green.

2. Synthesis and Antimicrobial Activities of Benzimidazole-1,2,3-Triazoles

2.1. 2-Benzimidazole-R(Ar)-1,4-Disubstituted-1,2,3-Triazole Hybrids

Two series of new hybrids, 2-[4-((1H-benzimidazol-2-ylthio)methyl)-1H-1,2,3-triazol -1-yl]N′-(arylmethylidene)acetohydrazides (2a2l) and 2-[4-((1H-benzimidazol-2-ylthio) methyl)-1H-1,2,3-triazol-1-yl]N-(α-arylethylidene)acetohydrazides (3a3f) were prepared by Youssif et al. in two steps starting from 2-[4-((1H-benzimidazol-2-ylthio) methyl)-1H-1,2,3-triazol-1-yl] acetohydrazide 1 (Scheme 1). Compounds 2a2l exhibited pronounced antibacterial activity, which ranged from 35 to 75% of that of the standard drug against Staphylococcus aureus and 50–80% of that of Ciprofloxacin against E. Coli (MIC values of 3.125–12.5 μmol mL−1). Compound 2k showed the highest activity against S. aureus (75% activity, MIC = 12.5 μmol mL−1), while compound 2d was the most active derivative against E. Coli (80% activity, MIC = 3.125 μmol mL−1). All the synthesized compounds were tested as potential antifungal agents against Candida albicans using Fluconazole as a reference drug. Compound 1 showed the activity of 48% of that of Fluconazole (MIC = 12.5 μmol mL−1). Compounds 2e and 2k displayed higher antifungal activity among the other derivatives as they showed 75% activity of that of Fluconazole (MIC = 3.125 μmol mL−1). Compounds 3a3f exhibited moderate to good activity against E. Coli, and their activity was 50–70% of that of Ciprofloxacin (MIC values of 6.25–12.5 μmol mL−1), and compounds 3a and 3f were the most active compounds against E. coli as they showed 70% of that of Fluconazole (MIC = 6.25 μmol mL−1) while compound 3b showed the highest activity against Staphylococcus aureus (65% of that of Ciprofloxacin, MIC = 18 μmol mL−1) [118]. Al-blewi et al. used an azide–alkyne Huisgen cycloaddition reaction carried out by simultaneously mixing thiopropargylated benzimidazole 4 with the appropriate sulfa drug azides 5a5f, copper sulfate, and sodium ascorbate in DMSO/H2O to regioselectively furnish target mono-1,4-disubstituted-1,2,3-triazole tethered benzimidazole-sulfonamide conjugates 6a6f with 85–90% yields after 6–8 h of heating at 80 °C (Scheme 2). All compounds were evaluated for their antimicrobial activity (Table 1) against four pathogenic bacterial strains (Gram-positive: Bacillus cereus ATTC 10876, Staphylococcus aureus ATTC 25,923, and Gram-negative: Escherichia coli ATTC 25922, Pseudomonas aeruginosa ATTC 27,853 and two fungal strains, Candida albicans ATTC 50193, Aspergillus brasiliensis ATTC 16404). As can be seen in Table 1, compound 6a showed the best antibacterial activity against Bacillus cereus and Staphylococcus aureus (64 μg mL−1), and compounds 6c, 6d, and 6e showed the best antibacterial activity against Escherichia coli (64 μg mL−1) [119]. Evaluation of in silico physicochemical properties or ADMET (adsorption, distribution, metabolism, excretion, and toxicity) as a robust tool to confirm the potential of a drug candidate was applied for these compounds [120]. As per Lipinski’s rule of five, an orally administered drug should have a log p ≤ 5, a molecular weight (MW) < 500 Daltons, and an HBD ≤ 5 [121] to be in the acceptable range. Results have shown that all hybrids have in good agreement in terms of HBD. Rashdan et al. synthesized hybrids 10 starting from 2-azido-1H-benzo[d]imidazole derivatives 7a7b, which reacted with acetylacetone in the presence of sodium ethoxide to obtain hybrids molecules 8a8b. The latter acted as a key molecule for the synthesis of new carbazone derivatives 9a9b that were submitted to react with 2-oxo-N-phenyl-2 (phenylamino)acetohydrazonoyl chloride to obtain the target hybrid derivatives 10a10b (Scheme 3). All compounds were screened for their in vitro antimicrobial activity against pathogenic microorganisms Staphylococcus aureus, E. coli, Pseudomonas aeruginosa, Aspergillus niger, and Candida albicans. The results showed that compounds 10a and 10b had strong activity against all the tested pathogenic microbes. Compounds 8a and 9a only showed effects against the Gram-negative and Gram-positive bacteria and had no effect on the tested fungi. In addition, in silico, and in vitro findings showed that compounds 10a and 10b were the most active against bacterial strains and could serve as potential antimicrobial agents (Table 2). The hybrids 810 were subjected to molecular docking studies with DNA gyrase B and exhibited binding energy that extended from -9.8 to -6.4 kcal/mol, which confirmed their excellent potency. The compounds 10a and 10b were found to be with the minimum binding energy (−9.8 and −9.7 kcal/mol) as compared to the standard drug Ciprofloxacin (−7.4 kcal/mol) against the target enzyme DNA gyrase B, as summarized in Figure 2 [122].
ADMET analysis of compounds 810 exhibited that they have good absorption properties (%HIA) ranging from 99.57 to 100% [123]. For distribution, the compounds do not permeate the blood–brain barrier (BBB). Moreover, the molecules were negative in the AMES toxicity and carcinogenicity test, which suggests that they are non-mutagenic. Compounds 11a11g with terminal acetylene and 2-(azidomethoxy)ethyl acetate were condensed using CuI as catalyst and triethylamine (TEA) under microwave irradiation to achieve hybrids 1,2,3-triazole connected via benzene to the benzimidazole nucleus 12a12g with excellent yields (70–90%) (Scheme 2). The cleavage of the acetyl group using potassium carbonate (K2CO3) in methanol liberated the hydroxy group of the corresponding hybrid triazoles 13a13g (Scheme 4). in almost quantitative yields. Compounds 13a13g were screened for in vitro antifungal activities against two phytopathogenic fungi, Verticillium dahliae Kleb and Fusarium oxysporum f. sp. albedinis. The result of the mycelia linear growth rate indicates that some of the compounds show a weak inhibition against the two fungi, the only compound that shows a significantly increased rate is compound 13e, with a rate of 29.76% against Verticillium dahliae in the sporulation test [124].
Bistrović et al. synthesized in two steps hybrids 19a19e, 20a20e, and 21a21e starting from 4-(prop-2-ynyloxy)benzaldehyde 14 (Scheme 5). All compounds were evaluated for their in vitro antibacterial activity against Gram-positive bacteria: S. aureus ATCC 25923, methicillin-sensitive S. aureus, E. faecalis, vancomycin-resistant E. faecium, and Gram-negative bacteria: E. coli ATCC 25925, P. aeruginosa ATCC 27853, A. baumannii ATCC 19,606 and ESBL-producing K. pneumoniae ATCC 27736. Generally, compounds showed better activities against Gram-positive than Gram-negative bacteria. Compounds 20a20e, with better binding affinity relative to other amidines, were the most active against S. aureus (MIC = 8–32 µgmL−1). Compound 19a was the most promising candidate because of its higher potency (MIC = 4 µgmL−1) against ESBL-producing E. coli. Results of anti-trypanosomal evaluations showed that p-methoxyphenyl substituent in 19b21b enhanced activity, with 20b (IC50 = 1.1 mM and IC90 = 3.5 mM) being more potent than Nifurtimox. In contrast to the observed correlation between antimicrobial activity and DNA binding, the antiprotozoal effects of 20b did not correlate with its DNA affinity [125].
Rao et al. synthesized hybrids 22a22b (Figure 3) using the click chemistry approach. Compounds had weak activity against Mycobacterium bovis strain, with BCG values % inhibition = 27.3 and 26.2, respectively, at 30 µM concentration, using a turbidometric assay. Compounds 22a and 22b also showed moderate antiproliferative activity against human breast cancer cell line MCF-7, with IC50 values of 31.9 and 25.1 μM, respectively [126]. Ashok et al. synthesized in three steps hybrids 26a26j, starting from 1H-indole-3-carbaldehyde 7 (Scheme 6). The compounds were evaluated for their antimicrobial activity against Gram-positive Staphylococcus aureus ATCC 6538, Bacillus subtilis ATCC 6633, and Gram-negative Proteus vulgaris ATCC 29213, Escherichia coli ATCC 11,229 bacteria using Gentamicin as standard. Antifungal activity was tested against Candida albicans ATCC 10,231 and Aspergillus niger ATCC 9029 strains with the standard drug Fluconazole. Compounds 26b, 26c, and 26h with MIC of 3.125–6.25 μg mL−1 were found to be the most promising potential antimicrobial molecules [127]. The authors calculated various physicochemical parameters such as clogP, drug score, and drug-likeness of 26a26j using the Osiris Property Explorer software [128]. For all the compounds, the calculated clogP values were found to be below five according to Lipinski’s rule-of-5 and also exhibited positive values for drug score. Mallikanti et al. synthesized novel benzimidazole-conjugated 1,2,3-triazole analogs 29a29l in two steps: 1. formation of benzimidazole intermediate by reaction between 3’,5’-difluorobiphenyl-3,4-diamine 27 and 2-hydroxy-4-(prop-2-ynyloxy) benzaldehyde 28, and 2. microwave-assisted copper-catalyzed click reaction (Scheme 7). Compounds 29a29l showed minimal inhibition zones against all Gram-positive (S. aureus, B. subtilis) and Gram-negative (E. coli, P. aeruginosa) strains using Ampicillin as A standard drug. Among all tested compounds, the 29i and 29k showed higher activity against P. aeruginosa, S. aureus, and B. subtilis than the standard reference. Compounds 29a, 29b, 29c, 29d, 29e, 29f, 29g, 29h, 29j, and 29l showed moderate antibacterial activity against tested strains (Inhibition zone: 10–25 mm compared with 18–20 mm for Ampicillin). Compounds 29i, 29j, and 29k also established strong activity against both fungal strains, C. albicans MTCC 183 and A. niger MTCC 9652, compared to the standard drug Griseofulvin [70]. To understand the binding mode of novel compounds, docking simulations were performed against the crystal structures of glucosamine-6-phosphate synthase (GlmS) (PDB ID: 2VF5) of E. coli and secreted aspartic proteinase (Sap) 1 (PDB ID: 2QZW) of C. albicans retrieved from the protein data bank. The best active compound, 29l, scored the highest binding affinity value of about −10.0 kal/mol, which demonstrated two key interactions with the active site amino acid Asp549 of Glms with a bond distance of 2.66 and 2.81 Å. Further, the hydrophobic interactions were taken with Tyr312, Ser316, Asp474, Asn523, Ala572, and Ala551 of Glms, among which one π-π T-shaped interaction with Tyr312, and halogen bond [129] interactions with Tyr312, Asn523, and Asn551 (Figure 4). The binding energies and interactions of all compounds are better than that of Ampicillin, which proves that these molecules could best fit into the cavity of Glms [70]. Chandrika et al. reported hybrids 3032 with broad spectrum antifungal activity (0.975–3.9 µgmL−1 against C. albicans; 0.12–0.48 µgmL−1 against C. parapsilosis) (Figure 5). These compounds also displayed good activity against C. albicans biofilms (3.9–15.6 µgmL−1 against C. albicans) [130].

2.2. 1-Benzimidazole-R(Ar)-1,4-Disubstituted-1,2,3-Triazole Hybrids

Deswal et al. synthesized a new series of benzimidazole-1,2,3-triazole-indoline derivatives 35 by employing a click reaction between substituted N-propargylated benzimidazole derivatives 33 and in situ formed substituted 2-azido-1-(indolin-1-yl) ethanone derivatives 34, in moderate to good yields (Scheme 8). The obtained results indicate a stronger inhibitory effect of compound 35d against E. coli, while compound 35g showed good inhibition against all the tested strains except B. subtilis (Table 3). The good antimicrobial activity of the compounds was correlated with the presence of the pyridine ring in position ‘‘2’’ of the benzimidazole and the NO2 group on the indole ring. Furthermore, in vitro α-glucosidase inhibition of all synthesized derivatives identified 35e (IC50 = 0.015 ± 0.0003 μmol mL−1) and 35g (IC50 = 0.018 ± 0.0008 μmol mL−1) as potent inhibitors of α-glucosidase, even better than standard drug Acarbose [6].
Saber et al. synthesized new 1,4-disubstituted-1,2,3-triazole containing benzimidazolone derivatives 37a37d exclusively using click chemistry (Scheme 9). All derivatives exhibited antibacterial activity against tested strains, Staphylococcus aureus, Escherichia coli, and Pseudomonas aeruginosa, but compounds 37b and 37d are more effective against Gram-positive bacterium S. aureus (MIC = 3.125 µgmL−1), and 37b has better activity against Gram-negative bacterium E. coli (MIC = 3.125 µgmL−1) with Chloramphenicol as standard drug. The expected inhibition efficiency, 37c > 37a > 37b, was attributed to the favorable effect of the side carbon chain of the triazole moiety, according to DFT calculations, in this process [131,132]. Mohsen et al. synthesized hybrids 41a41e in three steps, starting from benzimidazole 38, namely two alkylation reactions and a click reaction (Scheme 10). New derivatives exhibited good zone inhibition of 6.8, 5.4, 5.2, 4.5, and 5.3 mm for the S. aureus strain and 5.4, 3.8, 4.2, 3.3, 4.9 mm for the E. coli strain, indicating that the 1,2,3-triazole core contributed significantly to bacterial growth suppression (Ciprofloxacin showed 10.2 mm for S. aureus and 10.4 mm for E. coli). Compared with Gram-negative bacteria, all compounds showed a strong effect against Gram-positive bacteria [94].

2.3. 1,2-Bis-Substitutedbenzimidazoles-R(Ar)-1,4-Disubstituted-1,2,3-Triazole

Rezki reported the intramolecular cyclization of thiosemicarbazides 42a42d in refluxing aqueous sodium hydroxide (2N) for 6 h with the formation of hybrids 43a43d with yields of 82–86% (Scheme 11). Among all the 1,2,4-triazole derivatives, N4-phenyl and N4-(4-fluorophenyl) derivatives 43a and 43b were found to be the most potent with MIC values of 4–8 µg mL−1. Also, triazoles 43c and 43d exerted the best inhibition against both tested fungal strains, A. brasiliensis and Candida albicans, with MIC values ranging from 0.5 to 4 µg mL−1, more potent than the reference drug Fluconazole. Condensation of compound 44 with several benzaldehydes in refluxing ethanol for 4–6 h with a catalytic amount of HCl produced a new class of hybrid Schiff bases 45a45g with yields of 84–86% (Scheme 12). The antimicrobial bioassay results for the synthesized Schiff bases 45a45g revealed that all of the tested compounds were more effective towards all of the organisms, with MIC values of 1–16 µg mL−1. Among them, Schiff bases 45c, 45d, and 45e with a fluorine atom at position “2” exhibited the highest antibacterial inhibition potency at MIC 1–8 µg mL−1. The Schiff base 45e containing a CF3 group exerted the highest antifungal inhibition activity with MIC of 1 µg mL−1 [133]. Al-blewi et al. synthesized triazoles 47a47f in two steps: i. regioselective alkylation of 4 with two equivalents of propargyl bromide in the presence of two equivalents of potassium carbonate as a base catalyst to afford benzimidazole 46 with 91% yield after stirring at room temperature overnight; ii. Copper-mediated Huisgen 1,3-dipolar cycloaddition reaction on compound 46 in good yields (82–88%) (Scheme 13). Generally, bis-1,2,3-triazoles 47a47f exhibited more potent antimicrobial activities than their mono-1,2,3-triazole derivatives 6a6f. This was attributed to the synergistic effect of the sulfonamoyl and tethered heterocyclic components in addition to the improved lipophilicity of the bis-substituted derivatives. Among the synthesized compounds, compound 47a was the most potent antimicrobial agent, with MIC values ranging between 32 and 64 μg mL−1 against all tested strains B. cereus, S. aureus, E. coli P. aeruginosa, C. albicans, and A. brasiliensis. Pharmacophore elucidation of the compound 47a47f was performed based on in silico ADMET evaluation of the tested compounds. Screening results of drug-likeness rules showed that all compounds follow the accepted rules, meet the criteria of drug-likeness, and follow Lipinski’s rule of five. In addition, the toxicity results showed that all compounds are non-mutagenic and noncarcinogenic [119].
Aparna et al. used a similar strategy for obtaining nine new bis-1,2,3-triazol-1H- 4-yl-substituted arylbenzimidazole-2-thiol derivatives 48a48l (Figure 6). Antibacterial activity of triazole derivatives 48 demonstrates moderate to good activity against Gram-negative (E. coli, S. typhy, P. aeruginosa) and Gram-positive (S. aureus) bacterial strains. The products 48i, 48k, and 48l are characterized by a broad spectrum of antibacterial activity at a concentration of 10 μg mL−1. The synthesized 1,2,3 triazole derivatives were studied for their molecular docking on the high-resolution X-ray crystal structure of FabI of Staphylococcus aureus (pdb id:4FS3) obtained from the protein data bank [134]. The highest dock score of –7.69 kcal/mol and the lowest dock score of –0.942 kcal/mol were obtained for molecules 48l and 48h, respectively [135].

2.4. Benzimidazole-R(Ar)-1,2,3-Triazole Hybrids as Antitubecular Agents

Ashok reported compound 26h has the best antitubercular drug candidate by inhibiting the growth of the MTB (Mycobacterium tuberculosis) strain with MIC = 3.125 μ mL−1 (7.1 μM) (control Rifampicin MIC = 0.04 μg mL−1 and isoniazid MIC = 0.38 μg mL−1). The best antitubercular activity of 26h may be attributed to the presence of the nitro group on the phenyl ring at the ortho position. Compound 26b (MIC = 6.25 μg mL−1 (14.7 μM)) with chlorine substituent, compound 26i (MIC = 6.25 μg mL−1(14.2 μM)) with trifluoromethyl substituent and compound 26j (MIC = 12.5 μg mL−1 (28.4 μM)) with benzyl substituent exhibited moderate antitubercular activity. Therefore, the incorporation of the electron-withdrawing nitro group, electronegative chlorine, and trifluoromethyl groups on the phenyl ring was highly favored for antitubercular activity. The authors calculated various physicochemical parameters and found from the theoretical data that compounds 26a26j also exhibited positive values for drug score [127]. Gill et al. reported syntheses of hybrids 51a51d by reaction between 2-(3-fluorophenyl)-1H-benzo[d]imidazole 50 and phenyl-substituted 4-(bromomethyl)-1-phenyl-1H-1,2,3-triazole 49 in DMF at room temperature (Scheme 14). Trifluorosubstituted-compound 51a possessed enhanced anti-mycobacterial activity, >96% of inhibition at 6.25 µg concentration. Also, compounds 51b and 51c, which had antimicrobial activities superior to the other compounds, were reported as the best choice for the preparation of new derivatives in order to improve effectiveness on intracellular mycobacteria (macrophage) or in infected animals [136]. Anand et al. reported a one-pot reaction between 2-propargylthiobenzimidazole 4, 4-bromomethylcoumarins/1-aza-coumarins 52/53 and sodium azide under click chemistry conditions to give exclusively 1,4-disubstituted triazoles 54a54n. (Scheme 15). Antitubercular assays against M. tuberculosis (H37Rv) coupled with in silico molecular docking studies indicated that dimethyl substituents 54c and 54d showed promising activity (MIC = 3.8 µMol L−1) with higher C-score values. Surflex-Dock was used to investigate detailed intermolecular interactions between the ligand and the target protein. Three-dimensional structure information on the target protein was taken from the PDB entry 4FDO. Processing of the protein included the removal of the co-crystallized ligand and water molecules, as well as the addition of essential hydrogen atoms. All 14 inhibitors 54a54n were docked into the active site of ENR, as shown in Figure 7a, and Figure 7b indicates the superimposition of compounds 54a and 54d with ligand [137]. Khanapurmath et al. synthesized triazoles 55 by click reaction (Figure 8a). Benzimidazolone bis-triazoles 55a55n showed better activity with MIC in the range 2.33–18.34 μM, and the most active compounds were 55h and 55m. All compounds exhibited moderate to low levels of cytotoxicity with IC50 values of the human embryonic kidney cells in the range of 943–12294 μM, and none of the 14 compounds exhibited any significant cytotoxic effects, suggesting huge potential for their in vivo use as antitubercular agents. Docking studies revealed an additional interaction of benzimidazolone oxygen in these compounds (Figure 8b) [138]. Also, Sharma et al. summarize 1,2,3-triazoles as antitubercular compounds and various hybrids with benzimidazole, coumarin, isoniazid, quinolines, etc. [39].

3. Synthesis and Antimicrobial Activities of Benzimidazole-1,2,4-Triazoles

3.1. 2-Benzimidazole-R(Ar)-1-(1,2,4-Triazole)

Pandey et al. synthesized hybrids 59a59e in three steps: reaction of 7-hydroxy-4-methyl coumarin with thiosemicarbazide to form triazole intermediate 57, which underwent Mannich reaction with formaldehyde, and an amino acid to form intermediates 58a58e. Intermediates 58a58e reacted with o-phenylenediamine in pyridine to give benzimidazole-1,2,4-triazole hybrids in poor yields (Scheme 16). Compound 59a displayed promising antifungal activity against Candida albicans and Cryptococcus himalayensis since the MIC value in each case was found to be 3.5 μg mL–1. Compound 59b showed low to moderate antifungal activity against all five fungi, Candida albicans, Cryptococcus himalayensis, Sporotrichum schenkii, Trichophyton rubrum, and Aspergillus fumigatus [139].
Jadhav et al. synthesized a series of hybrids 1,2,4-triazolyl-fluorobenzimidazoles in two steps: i. synthesis of 2-(4-(1H-1,2,4-triazol-1-yl)phenyl)-4,6-difluoro-1H-benzo [d]imidazole 62 by reaction between 3,5-difluorobenzene-1,2-diamine 60 and 4-(1H-1,2,4- triazol-1-yl)benzaldehyde 61 in toluene at 110 °C, and ii. alkylation of compound 62 in DMF at room temperature, with the formation of the final hybrids 63a63o (Scheme 17). All compounds were screened for antimicrobial activity against different Gram-positive organisms, S. aureus, P. aeruginosa, and Gram-negative organisms, E. coli and S. typhosa using Gentamycin as a reference standard. The data generated from preliminary screening showed that compounds displayed moderate to better antimicrobial activity. Compounds 63a, 63e, 63f, 63h, 63i, and 63l displayed maximum activity (Table 4) [140].
Barot et al. synthesized hybrid 64 and determined its antimicrobial activity against Bacillus cereus MTCC-430, Enterococcus faecalis MTCC-493, S. aureus MTCC-737, Escherichia coli MTCC-1687, Pseudomonas aeruginosa MTCC-2642, Klebsiella pneumonia MTCC-109, Candida albicans MTCC-3017, Aspergillus niger MTCC-1344 and Fusarium oxyspora MTCC-1755, of MIC = 13–18 µg ml−1, with Ofloxacine and Fluconazole as standard drugs [141]. Also, Jiang et al. reported antifungal activity for hybrid 65 against Candida albicans, Candida tropicalis, Cryptococcus neoformans, Trichophyton rubrum, and Aspergillus fumigatus of MIC80 = 1–64 µg mL−1 considering Fluconazole as a standard drug (Figure 9). From the antifungal activity data, preliminary SARs was obtained. In general, the amine linker was important for antifungal activities. Substituted piperazine derivatives were comparable or superior to the corresponding N-methyl derivatives. [142]. Luo et al. reported a series of naphthalimide benzimidazole-1,2,4-triazole hybrids 68a68h and the corresponding triazolium salts 69a69d prepared by convenient and efficient procedures starting from naphthalimide triazole 66 (Scheme 18). 2-Chlorobenzyl triazolium 68g and compound 69b with octyl group exhibited the best antibacterial activities among all the tested compounds, especially against S. aureus with an inhibitory concentration of 2 μg mL−1 which was equipotent potency to Norfloxacin (MIC = 2 μg mL−1) and more active than Chloromycin (MIC = 7 μg mL−1). Triazoliums 68g and 68f bearing 3-fluorobenzyl moiety displayed the best antifungal activities (MIC = 2–19 μg mL−1) against all the tested fungal strains, C. albicans ATCC 76615, A. fumigatus ATCC 96918, C. utilis, S. cerevisia and A. flavus, without being toxic to PC12 cell line within concentration of 128 μg mL−1. Further investigations showed that compound 68g could intercalate into calf thymus DNA to form the 68g-DNA complex, which could block DNA replication, exerting powerful antimicrobial activities. [143]. Benzimidazole-1,2,4-triazole Mannich base 70 was active against Bacillus subtilis and Bacillus pumilus (inhibition zone diameters being 19 and 17 mm, respectively, compared to Ciprofloxacin with 28 and 30 mm, respectively) [144].
Kankate et al. reported the synthesis of hybrids 73a73l (Scheme 19). The antifungal activity of compound 73 was tested against Candida albicans spores in vitro (turbidimetric method) and in vivo (kidney burden test). Compound 73i had a good antifungal activity as compared with the other twelve compounds at 0.0075 µM mL−1, which is equivalent to Fluconazole activity both in vitro and in vivo. The antifungal activity decreased with the increasing alkyl length of N1 of benzimidazole (methyl to ethyl). This was proven for compounds 73i and 73l, which showed MICs of 0.0075 and 0.015 µM mL−1, respectively. The ligand fit method was performed to study and predict the binding mode of the hybrids 73 with the target enzyme (homology modeled) cytochrome P450 lanosterol 14-α-demethylase of C. albicans. All compounds showed binding in the active site of the enzyme. The 1,2,4-triazole ring of compounds 73a73l is positioned almost perpendicular to the porphyrin plane, with a ring nitrogen (N-4) atom coordinated to the heme iron (Figure 10) [145,146]. Ahuja et al. reported antifungal activity of compounds 74a74c against F. verticillioides, D. oryzae, C. lunata, and F. fujikuroi (Figure 11). All compounds had increased potency than the standard commercial benzimidazole fungicide, carbendazim (Table 5).
Compound 74c exhibited ED50 values lower than triazole fungicide, propiconazole. The results reinforced the synergistic effects of the benzimidazole and 1,2,4-triazole combination supported by a computational approach. Hydrogen bonding interactions were more pronounced in compounds 74a74c in the binding pockets of both the target enzymes, in comparison to standards. In compound 74c, two H-bonds were formed with Gln11 present in the binding cleft of the active pocket of β-tubulin (Figure 12). In all three compounds, the right position of the N-atoms of both 1,2,4-triazole and benzimidazole, that O-atoms of methoxy and carbonyl groups, contributed well to strong binding into the active site of enzymes via H-bonding [147]. Evren et al. reported the synthesis of the compounds 79a79c in two steps: i. reaction of 1,2,4-triazole 75 with 4-fluorobenzaldehyde 76 in DMF with the formation of 4-(1H-1,2,4-triazol-1-yl)benzaldehyde 77; ii. reaction of aldehyde 77 with 1,2-phenylene diamines 78 (Scheme 20). Although the antibacterial activities of compounds 79a79c against Escherichia coli ATCC 35218, E. coli ATCC 25922, Klebsiella pneumoniae NCTC 9633, Pseudomonas aeruginosa ATCC 27853, Salmonella typhimurium ATCC 13311, and Staphylococcus aureus ATCC 25923, were weak, the antifungal activities against C. albicans were found promising, with MIC values of 3.9, 7.8, and 3.9 μg mL−1 respectively, using as reference drug Ketokonazole (MIC = 7.8 μg mL −1). Protein-ligand interactions and binding poses of the 79a, 79b, and 79c compounds on the CYP51 active site were examined. As shown in Figure 13, an H bond of 2.11 Å between compound 79a and Met508, π-π stacking interactions with Tyr118, Hie377, Phe233, generated hydrophobic interactions with Pro230, Leu376, Tyr64, Phe228, and Tyr505. Theoretical ADME calculations of the 79a, 79b, and 79c were made, and the compounds were found to have good lipophilicity, moderate water solubility, and within the limiting rules of Lipinski, Ghose, Veber, Egan, and Muegge (Figure 14) [148]. Ghobadi et al. reported the synthesis of compounds 85a85e, in two different ways, from 3,4-diaminobenzophenone 80, i. formation of 2-mercapto benzimidazole derivatives 82, 83, and ii. nucleophilic ring opening of various oxiranes 84a84e with benzimidazoles 82 and 83 using NaHCO3 in ethanol at room temperature (Scheme 21). Compounds 85a85e, containing a 5-benzoylbenzimidazole scaffold, showed better antifungal activity against Candida spp. and Cryptococcus neoformans than related benzimidazole and benzothiazole derivatives. The better results were obtained with 4-chloro-derivative 85b displaying MICs < 0.063–1 μg mL−1. Also, compound 86c, synthesized analogously, is as potent as compound 85b. The docking experiments were conducted to further rationalize the obtained antifungal activity data and investigate the type of interactions between compound 85b and the active site of lanosterol 14α-demethylase (CYP51). As shown in Figure 13, the coordinated bond-forming distance between the N4 atom of the triazole nucleus of compound 85b and the iron atom in the heme group of active site were 2.71 and 2.40 Å, respectively. A hydrogen-bonding interaction between Tyr132 and the sulfur group of (S)-85b was observed. In vitro and in silico ADMET evaluations of the most promising compounds 85b indicated that the selected compounds have desirable ADMET properties in comparison to the standard drug Fluconazole. A docking simulation study demonstrated that the benzimidazol-2-yl-thio moiety is responsible for the potent antifungal activity of these compounds [72].

3.2. 1-Benzimidazole-R(Ar)-2-1,2,3-Triazole

Ansari et al. synthesized hybrids 88a88c in two steps from 2-(2-methyl-1H-benzo [d]imidazol-1-yl)acetohydrazide 87 (Scheme 22). Generally, all benzimidazole-triazole hybrids showed low antimicrobial activity (Table 6) [149]. Tien et al. synthesized hybrids 89a89d in three steps from 2-(2-methyl-1H-benzo[d]imidazol-1-yl)acetohydrazide 87b (Scheme 23). All compounds exhibited antifungal activity against A. niger (MIC = 50 µg mL−1). Only compound 89b exhibited activity against F. oxysporum (Table 7) [150]. Kantar et al. reported antimicrobial activity of hybrid 90 (Figure 15) against four Gram-positive, Bacillus cereus 702 Roma (62.5 µg mL−1), B. megaterium DSM-32 (125 µg mL−1), B. subtilis ATCC 6633 (62.5 µg mL−1), Staphylococcus aureus ATCC 25,923 (250 µg mL−1), and four Gram-negative bacteria, Escherichia coli ATCC 25,922 (250 µg mL−1), Enterobacter cloaceae ATCC13047 (125 µg mL−1), Pseudomonas aeruginosa ATCC 27,853 (250 µg mL−1), and Yersinia pseudotuberculosis ATCC 911 (125 µg mL−1) bacteria [151]. Nandwana et al. reported compound 91 synthesized in good yield (70%) with promising antibacterial activity, with minimum inhibitory concentration (MIC) values of 4−8 μg mL−1 for all bacterial tested strains (Escherichia coli, Pseudomonas putida, Salmonella typhi, Bacillus subtilis, Staphylococcus aureus), as compared to the positive control Ciprofloxacin, and also with pronounced antifungal activity against both tested strains, Aspergillus niger and Candida albicans (MIC = 8−16 μg mL−1) as compared with Amphotericin B [152]. Al-Majidi et al. synthesized 2-mercaptobenzimidazole derivatives 95, 96, and 97 by cyclization of intermediate precursors 93, 94, and 95 under reflux with 2N NaOH (Scheme 24). The compounds generally showed moderate antimicrobial activity against all tested strains, as can be seen in Table 8 [153]. El-masry et al. synthesized compounds 98 and 99 and found that they did not exhibit antimicrobial activity (Figure 16) [154]. Menteşe et al. synthesized compounds 100a100d, for which they found no antimicrobial activity on the ten strains tested [155]. Karale et al. synthesized bis-benzimidazole-1,2,4-triazole hybrids 102a102e (Scheme 25) in four steps from 7-methyl-2-propyl-3H-benzo[d]imidazole-5-carboxylic acid. All compounds 102 did not show antimicrobial activity against the strains tested, C. albicans, A. fumigatus, S. aureus, and E. coli [156,157].

3.3. 2-Benzimidazole-R(Ar)-2-1,2,4-Triazole

Eisa et al. synthesized compounds 105a and 105b (Scheme 26) (Table 9) by the reaction between 2-(chloromethyl)-1H-benzo[d]imidazole 103 and 4-phenyl-5-(pyridin-3-yl)-4H-1,2,4- triazole-3-thiol 104a or 4-phenyl -5-(thiophen-2-yl)-4H-1,2,4-triazole-3-thiol 104b, at reflux in absolute ethanol, for 12 h. Also, they reported synthesis of the compounds 107a and 107b from 2-(2-(phenylthiomethyl)-1H-benzo[d]imidazol-1-yl)acetohydrazide in two steps (Scheme 27). All compounds showed antimicrobial activity against Escherichia coli superior to that of standard Gentamicin. Compound 107a exhibited only moderate activity against Staphylococcus aureus [158]. Nevade et al. synthesized compounds 109a109h in five steps from 1H-benzo[d]imidazole-2-thiol 108 (Scheme 28). The antimicrobial screening results presented in Table 10 reveal that compounds 109a, 109c, and 109e exhibited satisfactory effects against S.aureus and E.coli, while compounds 109b, 109f, and 109g showed moderate activity against the same microbes. Also, the antifungal activity of these compounds was screened against Candida albicans. Compounds 109a and 109d showed the highest degree of inhibition against C.albicans when compared with the standard drug Ketoconazole [159]. Can et al. synthesized hybrids 111a111h in four steps from methyl 4-(5-methyl-1H-benzo[d]imidazol-2-yl)benzoate 110 (Scheme 29). All compounds were screened for antifungal activity against Candida albicans ATCC 24433, Candida glabrata ATCC 90030, Candida krusei ATCC 6258, and Candida parapsilosis ATCC 22,019 (Table 11). Compounds 111i and 111s exhibited significant inhibitory activity against Candida strains with MIC50 values ranging from 0.78 to 1.56 μg mL−1 [160]. Gencer et al. synthesized compounds 112 in good yields (77–88%) using a similar strategy (Figure 17). Microbiological studies revealed that compounds 112a, 112b, 112c, 112e, 112f, 112g, and 112h possess a good antifungal profile against all tested strains, C. albicans, C. glabrata, C. krusei, C. parapsilopsis, with MIC50 = 0.78–1.56 µg mL−1. Compound 112i was the most active derivative and showed comparable antifungal activity to those of reference drugs Ketoconazole and Fluconazole [161]. The SAR (Structure–activity relationship) on the synthesized benzimidazole-triazole compounds is summarized in Figure 18. It is observed that the presence of chlorine or fluorine in the “5” position of benzimidazole, as well as the presence of fluorine in the “4” position of phenyl, increase the antibacterial activity, while the presence of fluorine in the “2” position of phenyl does not change the activity, and the presence of groups CH3 or C2H5 in position “4” in the triazole nucleus does not bring any change in the antibacterial activity of the compounds. Furthermore, toxicological and ADME studies indicated the relative potency of hybrids 112h and 112i, according to the literature [162,163,164,165,166]. Compound 112i also inhibited ergosterol biosynthesis concentration dependently. Results of ergosterol level quantification assay and fluorescence microscopy studies revealed that the mechanism of action of hybrids is associated with the inhibition of ergosterol biosynthesis, which may subsequently result in altered membrane fluidity, plasma membrane biogenesis, and functions of fungi. Güzel et al. synthesized a new series of benzimidazole-1,2,4-triazole derivatives 113a113l using the same procedure described in Scheme 29 as potential antifungal agents (Figure 19). All the compounds were screened for their in vitro antifungal activity against four fungal strains, namely, C. albicans, C. glabrata, C. krusei, and C. parapsilopsis and were found to exhibit excellent activity against C. glabrata. Especially, compounds 113b, 113i, and 113j were found to be the most effective compounds in the series with an MIC value of 0.97 μg mL−1 [71]. According to the molecular docking study, compounds 113b, 113i, and 113j fit into the LDM enzyme active pocket. In a previous study [167], the Tyr118 amino acid and HEM601 protein were described as essential residues, and in this study, the synthesized active compounds interacted significantly with Tyr118, His377, and HEM601 residues. The interactions with HEM were seen as π−π stacking and π−cation interactions. Therefore, the antifungal effects of compounds 113b, 113i, and 113j were considered to be caused by the destruction of cell integrity due to the inhibition of the LDM enzyme. The authors identified compound 6i with higher inhibitory activity due to H-bonding with Tyr132, unlike the other two compounds. Aryal et al. reported synthesis of 2-substituted benzimidazole containing 1,2,4-triazoles 114a and 114b (Figure 20). The compounds did not show antimicrobial activity against the tested strains Staphylococcus aureus ATCC 6538P and Staphylococcus epidermidis ATCC 1228 [168]. Kazeminejad et al. did a study on 1,2,4-triazoles as well as structure–activity relationships (SAR) [38].

3.4. 6-Substituted-Benzimidazole-R(Ar)-1-1,2,4-Triazole

Nandha et al. reported synthesis of 6-substituted-benzimidazoles with 1-(1,2,4-triazole) 115a115d in three steps from 5-chloro-4-fluoro-2-nitrobenzenamine (Scheme 30). All compounds were screened against M. tuberculosis and four fungal strains, C. albicans, C. glabrata, C. krusei, and C. tropicalis. Compound 115c was the most active against M. tuberculosis and all tested fungal strains (MIC = 25 μg mL−1) [169].

4. Synthesis and Antiviral Activities of Benzimidazole-Triazoles

Over 200 viruses are known to cause disease in humans, yet currently approved antiviral drugs are available to treat only about 10 of these viral infections [170,171]. The past decade has underscored the global threat posed by emerging viruses. An alternative solution is the development of broad-spectrum antiviral drugs. One advantage of this approach is reduced time and cost associated with the early stages of drug development per approved indication. It can also diminish the clinical risks in more advanced stages of development [172,173]. Youssif et al. reported the synthesis of benzimidazole-1,2,3-triazole hybrids 2-{4-[(1-benzoylbenzimidazol-2-ylthio)methyl]-1H-1,2,3-triazol-1-yl}-N-(4-nitro-phenyl)-acetamide 116 and 2-(4-{[1-(4-chlorobenzoyl)-benzimidazol-2-ylthio)methyl]-1H-1,2,3-triazol -1-yl}-N-(4-nitrophenyl)-acetamide 117 which showed significant activity against hepatitis C virus (HCV) (Figure 21). Thus, fifty percent effective concentrations (EC50) of HCV inhibition for compounds 116 and 117 were 7.8 and 7.6 μmol L–1, respectively, and the 50% cytotoxic concentrations (CC50) were 16.9 and 21.1 μmol L–1. The results gave an insight into the importance of the substituent at position 2 of benzimidazole for the inhibition of HCV [73].
The antiviral activity of compounds 59a59e was tested against two viruses, viz., Japanese encephalitis virus (JEV) (P20778), an RNA virus of higher pathogenicity, and Herpes simplex virus type-I (HSV-I) (753166), the most common virus present in the environment. The antiviral activity of the compounds data is given in Table 12. All but one of the five compounds were found active against JEV. Compound 59b displayed 90% CPE (cytopathic effect) in vitro with an effective concentration of 8 µg mL–1, while in vivo activity was less significant (16% protection with an MST of 4 days). The authors suggested that these compounds are better anti-JEV agents than anti-HSV agents since two such compounds, namely 59b and 59e, also displayed a measurable degree of anti-JEV activity in vivo. Compound 59c was found antivirally inactive against both viruses. The anti-HSV-I activity was found to be in the order of 33, 46, 53, and 64% for compounds 59a, 59b, 59d, and 59e, respectively. Since among compounds 59a to 59e, only compound 59e contains a methyl group instead of H as R1; it follows that R1 does not seem to be responsible for the biological activity [139].
Tonelli et al. synthesized a series of 1-substituted 2-[(benzotriazol-1/2-yl)methyl] benzimidazoles 118137 and tested for antiviral activity against a large panel of RNA and DNA viruses (Figure 22). Twelve compounds exhibited high activity against RSV (Respiratory Syncytial Virus), with EC50 values in most cases below 1 µM, comparing favorably with the reference drug 6-azauridine, which, moreover, exhibited high toxicity against both the MT-4 and Vero-76 cell lines (S.I. = 16.7). The observed activity against BVDV (Bovine Viral Diarrhea Virus), YFV (Yellow Fever Virus), and CVB2 (Coxsackie virus B2) is moderate, with EC50 values in the range of 6–55 µM for the best compounds (Table 13). Though not particularly impressive, the presently uncovered activity against BVDV, YFV, and CVB2 is of some interest because it may lead, through the identification of the target, to the development of broad spectrum antiviral agents. In this respect, the definition of the mode of action of the above compounds is mandatory. Furthermore, since the activity against these viruses was influenced by the presence and nature of the substituents in position “5” of the benzimidazole ring, it will be worthwhile to further explore the effect of diversified substitutions as a possibility to improve activity and/or decrease cytotoxicity [174]. SARS-CoV-2 and its variants, especially the Omicron variant, remain a great threat to human health [10]. More novel variants of SARSCoV-2 are also expected to originate in the future. Therefore, efforts should be made to develop wide-ranging measures to prevent future outbursts of zoonotic origin. Recent articles reported essential and up-to-date information about SARS-CoV-2 variants, antiviral drugs, and vaccines used to fight it [175,176].
Al-Humaidi et al. reported the synthesis of a series of benzimidazole-1,2,3-triazoles 138140 (Figure 23). Molecular docking studies and in vitro enzyme activity revealed that most of the investigated compounds demonstrated promising binding scores against the SARS-CoV-2 and Omicron spike proteins in comparison to the reference drugs (Table 14).
Data proved the promising activity of the tested compound 140, with its IC50 reaching 75.98 nM against the Omicron spike protein and 74.51 nM against the SARS-CoV-2 spike protein. The three-dimensional binding mode of compound 140 is shown in Figure 24. Benzimidazole-1,2,3-triazole hybrids can be potent anti-HSV (Herpes simplex virus) agents. These compounds were screened against flaviviruses and pestiviruses. Compound 141 showed excellent activity against respiratory syncytial virus (RSV) with an EC50 value of 0.02 mM (Figure 25) [74]. Seliem et al. designed and synthesized some quinolone–triazole conjugates against SARS-CoV-2. It was revealed that 4-((1-(2-chlorophenyl)-1H-1,2,3-triazol- 4-yl)methoxy)- 6-fluoro-2-(trifluoromethyl)quinoline and 6-fluoro-4-(2-(1-(4-methoxyphenyl) -1H-1,2,3- triazol-4-yl)ethoxy)-2-(trifluoromethyl)quinoline have high antiviral activity with a high selectivity index (SI) against SARS-CoV-2 in comparison to the reference drugs. They explained that the fluorine atoms in the tested compounds have a major role in the observed antiviral activity [43]. The importance of the 1,2,4-triazole ring in antiviral compounds is reviewed by El-Sebaey, who emphasizes the importance of the substituents in the triazole nucleus, as well as the important role of other heterocycles in the molecule [177].

5. Conclusions

This review summarizes the syntheses of benzimidazole–triazole compounds with antimicrobial and antiviral properties mentioned in the literature. The presence of certain groups grafted on the benzimidazole and triazole nuclei, such as -F, -Cl, -Br, -CF3, -NO2, -CN, -NHCO, -CHO, -OH, OCH3, -N(CH3)2, COOCH3, as well as other heterocycles in the molecule (pyridine, pyrimidine, thiazole, indole, isoxazole, thiadiazole, coumarin), increases the antimicrobial activity of the compounds [4,5,114,115,165,178,179,180]. From the presented literature data, we can highlight some aspects related to the correlation: structure—antimicrobial properties.
-
The presence of substituents in the “4” or “5” positions of the benzimidazole nucleus can increase the antimicrobial activity of the benzimidazole-triazole hybrids (compounds 12, 13, 19, 20, 35).
-
The presence of the ortho- or para-substituted phenyl substituent in the “1” position of 1,2,3-triazoles in benzimidazole-triazole hybrids can increase their antimicrobial activity.
-
In the case of benzimidazoles substituted in the “1” position with triazoles, the presence of an aliphatic or aromatic radical substituent increases the antimicrobial activity of the hybrids.
-
The presence of the oxygen atom in the bridge that connects the benzimidazole and triazole rings is favorable to the antimicrobial activity of the hybrids (compounds 19, 20, 21, 29, 30).
-
The presence of the sulfur atom in the bridge that connects the benzimidazole and triazole rings is favorable to the antimicrobial activity of the hybrids and even to the antitubercular activity (9597, 105, 107).
-
The presence of a supplementary triazole ring in benzimidazole-triazole hybrids improves their antimicrobial activity (compounds 43, 45, 47).
-
The presence of the benzoyl substituent in the “5” position of the benzimidazole in the benzimidazole-1,2,4-triazole hybrids clearly improves their antimicrobial activity (compounds 85a85e).
-
The phenyl nucleus as a spacer between the “1” position of 1,2,4-triazole and the “2” position of benzimidazole favors the formation of antimicrobial compounds, and the substituents in the “5” position of the benzimidazole nucleus increase the antimicrobial activity (compounds 79, 111, 112, 113).
-
Only benzimidazole-1,2,3-triazole hybrids are mentioned in the literature as having antiviral properties.
-
2-Substituted or 1,2-disubstituted benzimidazoles with 1,2,3-triazoles are mentioned as antiviral compounds, and the presence of an additional triazole ring improves the antiviral activity (compound 140).
The presence of both the benzimidazole ring and the triazole ring in a single molecule enhanced the effectiveness of the antimicrobial activities, as seen in the sections above. The recent ADME and SAR studies mentioned in this review are also important for directing new syntheses of benzimidazole-triazole hybrids in close correlation with their properties.
As mentioned in the cited literature, it is extremely useful, both from a therapeutic and economic point of view, that the synthesized compounds, such as the benzimidazole-triazole hybrids analyzed in this review, possess both antimicrobial and antimicrobial biological activity antiviral, to meet the medical requirements demanded especially lately, for better action, especially in the case of SARS-CoV-2.
The ADME studies performed on the benzimidazole-triazole hybrids mentioned in this review recommend the compounds as antimicrobials and antivirals and open new horizons to create new compounds, following the conclusions found here, with improved biological properties.
The articles researched on this topic, although they report the general characteristics of these molecules (lipophilicity/hydrophilicity), in order to have the desired antimicrobial or antiviral properties, refer only to liquid formulations in the form in which the compounds were tested, and so far not no article is reported that formulates in the form of nanosystems, nanoparticles for better availability of the active substance. This remains an open research topic for future studies.
We hope that this review will be useful for the design and synthesis of new benzimidazole-triazole hybrids with antimicrobial and antiviral properties in the context of exacerbation of microbial and viral infections and resistance to treatments with drugs known on the market.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The author is thankful to the Department of Organic Chemistry, Biochemistry, and Catalysis, for providing the necessary facilities to carry out this research work.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Kabir, E.; Uzzaman, M. A review on biological and medicinal impact of heterocyclic compounds. Results Chem. 2022, 4, 100606. [Google Scholar] [CrossRef]
  2. Nishanth Rao, R.; Jena, S.; Mukherjee, M.; Maiti, B.; Chanda, K. Green synthesis of biologically active heterocycles of medicinal importance: A review. Environ. Chem. Lett. 2021, 19, 3315–3358. [Google Scholar] [CrossRef]
  3. Ebenezer, O.; Oyetunde-Joshua, F.; Omotoso, O.D.; Shapi, M. Benzimidazole and its derivatives: Recent Advances (2020–2022). Results Chem. 2023, 5, 100925. [Google Scholar] [CrossRef]
  4. Bansal, Y.; Kaur, M.; Bansal, G. Antimicrobial Potential of Benzimidazole Derived Molecules. Mini Rev. Med. Chem. 2019, 19, 624–646. [Google Scholar] [CrossRef] [PubMed]
  5. Marinescu, M. Synthesis of Antimicrobial Benzimidazole–Pyrazole Compounds and Their Biological Activities. Antibiotics 2021, 10, 1002. [Google Scholar] [CrossRef] [PubMed]
  6. Deswal, L.; Verma, V.; Kumar, D.; Deswal, Y.; Kumar, A.; Kumar, R.; Parshad, M.; Bhatia, M. Synthesis, antimicrobial and α-glucosidase inhibition of new benzimidazole-1,2,3-triazole-indoline derivatives: A combined experimental and computational venture. Chem. Pap. 2022, 76, 7607–7622. [Google Scholar] [CrossRef]
  7. Raducka, A.; Świątkowski, M.; Korona-Głowniak, I.; Kaproń, B.; Plech, T.; Szczesio, M.; Gobis, K.; Szynkowska-Jóźwik, M.I.; Czylkowska, A. Zinc Coordination Compounds with Benzimidazole Derivatives: Synthesis, Structure, Antimicrobial Activity and Potential Anticancer Application. Int. J. Mol. Sci. 2022, 23, 6595. [Google Scholar] [CrossRef] [PubMed]
  8. Zalaru, C.; Dumitrascu, F.; Draghici, C.; Tarcomnicu, I.; Marinescu, M.; Nitulescu, G.M.; Tatia, R.; Moldovan, L.; Popa, M.; Chifiriuc, M.C. New Pyrazolo-Benzimidazole Mannich Bases with Antimicrobial and Antibiofilm Activities. Antibiotics 2022, 11, 1094. [Google Scholar] [CrossRef] [PubMed]
  9. Chen, M.; Su, S.; Zhou, Q.; Tang, X.; Liu, T.; Peng, F.; He, M.; Luo, H.; Xue, W. Antibacterial and antiviral activities and action mechanism of flavonoid derivatives with a benzimidazole moiety. J. Saudi Chem. Soc. 2021, 25, 101194. [Google Scholar] [CrossRef]
  10. Kanwal, A.; Ahmad, M.; Aslam, S.; Naqvi, S.A.R.; Jawwad Saif, M.J. Molecular-biological prolems of drug design and mehanism of drug action. Recent advances in antiviral benyimidayole derivatives: A mini review. Pharm. Chem. J. 2019, 53, 179–187. [Google Scholar] [CrossRef]
  11. Brishty, S.R.; Hossain, M.J.; Khandaker, M.U.; Faruque, M.R.I.; Osman, H.; Rahman, S.M.A. A Comprehensive Account on Recent Progress in Pharmacological Activities of Benzimidazole Derivatives. Front. Pharmacol. 2021, 12, 762807. [Google Scholar] [CrossRef] [PubMed]
  12. Yhou, S.; Huang, G. Synthesis of anti-allergic drugs. RSC Adv. 2020, 10, 5874. [Google Scholar] [CrossRef]
  13. Vasil’ev, P.M.; Kalitin, K.Y.; Spasov, A.A.; Grechko, O.Y.; Poroikov, V.V.; Filimonov, D.A.; Anisimova, V.A. Search for new dugs prediction and study of anticonvulsant properties of benzimidazole derivatives. Pharm. Chem. J. 2017, 50, 775–780. [Google Scholar] [CrossRef]
  14. Lee, Y.T.; Tan, Y.J.; OOn, C.E. Benzimidazole and its derivatives as cancer therapeutics: The potential role from traditional to precision medicine. Acta Pharm. Sin. B 2023, 13, 478–497. [Google Scholar] [CrossRef] [PubMed]
  15. Satija, G.; Sharma, B.; Madan, A.; Iqubal, A.; Shaquiquzzaman, M.; Akhter, M.; Parvez, S.; Khan, M.A.; Alam, M.M. Benzimidazole based derivatives as anticancer agents: Structure activity relationship analysis for various targets. J. Heterocycl. Chem. 2021, 59, 22–66. [Google Scholar] [CrossRef]
  16. Song, B.; Park, E.Y.; Kim, K.J.; Ki, S.H. Repurposing of Benzimidazole Anthelmintic Drugs as Cancer Therapeutics. Cancers 2022, 14, 4601. [Google Scholar] [CrossRef]
  17. Chen, W.L.; Li, D.D.; Chen, X.; Wang, Y.Z.; Xu, J.-J.; Jiang, Z.-Y.; You, Q.-D.; Guo, X.-K. Proton pump inhibitors selectively suppress MLL rearranged leukemia cells via disrupting MLL1-WDR5 protein-protein interaction. Eur. J. Med. Chem. 2020, 188, 112027. [Google Scholar] [CrossRef]
  18. Argirova, M.A.; Georgieva, M.K.; Hristova-Avakumova, N.G.; Vuchev, D.I.; Popova-Daskalova, G.V.; Anichina, K.K.; Yancheva, D.Y. New 1H-benzimidazole-2-yl hydrazones with combined antiparasitic and antioxidant activity. RSC Adv. 2021, 11, 39848–39868. [Google Scholar] [CrossRef]
  19. Valderas-García, E.; Häberli, C.; Álvarez-Bardón, M.; Escala, N.; Castilla-Gómez de Agüero, V.; de la Vega, J.; del Olmo, E.; Balaña-Fouce, R.; Keiser, J.; Martínez-Valladares, M. Benzimidazole and aminoalcohol derivatives show in vitro anthelmintic activity against Trichuris muris and Heligmosomoides polygyrus. Parasit. Vectors 2022, 15, 243. [Google Scholar] [CrossRef]
  20. Kamat, V.; Yallur, B.C.; Poojary, B.; Patil, V.B.; Nayak, S.P.; Krishna, P.M.; Joshi, S.D. Synthesis, molecular docking, antibacterial, and anti-inflammatory activities of benzimidazole-containing tricyclic systems. J. Chin. Chem. Soc. 2021, 68, 1055–1066. [Google Scholar] [CrossRef]
  21. Moharana, A.K.; Dash, R.N.; Mahanandia, N.C.; Subudhi, B.B. Synthesis and anti-inflammatory activity evaluation of some benzimidazoled erivatives. Pharm. Chem. J. 2022, 56, 1070–1074. [Google Scholar] [CrossRef]
  22. Veerasamy, R.; Roy, A.; Karunakaran, R.; Rajak, H. Structure–Activity Relationship Analysis of Benzimidazoles as Emerging Anti-Inflammatory Agents: An Overview. Pharmaceuticals 2021, 14, 663. [Google Scholar] [CrossRef]
  23. Iqbal, H.; Verma, A.K.; Yadav, P.; Alam, S.; Shafiq, M.; Mishra, D.; Khan, F.; Hanif, K.; Negi, A.S.; Chanda, D. Antihypertensive Effect of a Novel Angiotensin II Receptor Blocker Fluorophenyl Benzimidazole: Contribution of cGMP, Voltage-dependent Calcium Channels, and BKCa Channels to Vasorelaxant Mechanisms. Front. Pharmacol. 2021, 30, 611109. [Google Scholar] [CrossRef]
  24. Tajane, P.S.; Sawant, R.L. An updated review on benzimidazole derivatives as potential antihypertensive agents. Int. J. Health Sci. 2022, 6 (Suppl. S1), 7169–7179. [Google Scholar] [CrossRef]
  25. Aboul-Enein, H.Y.; El Rashedy, A.A. Benzimidazole Derivatives as Antidiabetic Agents. Med. Chem. 2015, 5, 318–325. [Google Scholar] [CrossRef]
  26. Dik, B.; Coşkun, D.; Bahcivan, E.; Unez, K. Potential antidiabetic activity of benzimidazole derivative albendazole and lansoprazole drugs in different doses in experimental type 2 diabetic rats. Turk. J. Med. Sci. 2021, 51, 1578–1585. [Google Scholar] [CrossRef] [PubMed]
  27. Farid, S.M.; Noori, M.; Montazer, M.N.; Ghomi, M.K.; Mollazadeh, M.; Dastyafteh, N.; Irajie, C.; Zomorodian, K.; Mirfazli, S.S.; Mojtabavi, S.; et al. Synthesis and structure–activity relationship studies of benzimidazole-thioquinoline derivatives as α-glucosidase inhibitors. Sci. Rep. 2023, 13, 4392. [Google Scholar] [CrossRef]
  28. Stanton, J.B.; Schneider, D.A.; Dinkel, K.D.; Balmer, B.F.; Baszler, T.V.; Mathison, B.A.; Boykin, D.W.; Kumar, A. Discovery of a Novel, Monocationic, Small-Molecule Inhibitor of Scrapie Prion Accumulation in Cultured Sheep Microglia and Rov Cells. PLoS ONE 2012, 7, e0119084. [Google Scholar] [CrossRef]
  29. Dinparast, L.; Zengin, G.; Bahadori, M.B. Cholinesterases Inhibitory Activity of 1H-benzimidazole Derivatives. Biointerface Res. Appl. Chem. 2021, 11, 10739–10745. [Google Scholar] [CrossRef]
  30. Adalat, B.; Rahim, F.; Taha, M.; Alshamrani, F.J.; Anouar, E.H.; Uddin, N.; Shah, S.A.A.; Ali, Z.; Zakaria, Z.A. Synthesis of Benzimidazole–Based Analogs as Anti Alzheimer’s Disease Compounds and Their Molecular Docking Studies. Molecules 2020, 25, 4828. [Google Scholar] [CrossRef]
  31. Cheretaev, I.V.; Korenyuk, I.I.; Nozdrachev, A.D. Neurotropic, Psychoactive, and Analgesic Properties of Benzimidazole and Its Derivatives: Physiological Mechanisms. Neurosci. Behav. Physiol. 2018, 48, 848–853. [Google Scholar] [CrossRef]
  32. Maltsev, D.V.; Spasov, A.A.; Vassiliev, P.M.; Skripka, M.O.; Miroshnikov, M.V.; Kochetkov, A.N.; Eliseeva, N.V.; Lifanova, Y.V.; Kuzmenko, T.A.; Divaeva, L.N.; et al. Synthesis and Pharmacological Evaluation of Novel 2,3,4,5-tetrahydro[1,3]diazepino[1,2-a]benzimidazole Derivatives as Promising Anxiolytic and Analgesic Agents. Molecules 2021, 26, 6049. [Google Scholar] [CrossRef] [PubMed]
  33. Yen, T.L.; Wu, P.; Chng, C.L.; Yang, W.B.; Jayakumar, T.; Geraldine, P.; Chou, C.M.; Chang, C.Y.; Lu, W.J.; Sheu, J.R. Novel synthetic benzimidazole-derived oligosaccharide, M3BIM, prevents ex vivo platelet aggregation and in vivo thromboembolism. J. Biomed. Sci. 2016, 23, 26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Zhang, T.; Liu, Q.; Ren, Y. Design, synthesis and biological activity evaluation of novel methyl substituted benzimidazole derivatives. Tetrahedron 2020, 76, 131027. [Google Scholar] [CrossRef]
  35. Zhang, B. Comprehensive review on the anti-bacterial activity of 1,2,3-triazole hybrids. Eur. J. Med. Chem. 2019, 168, 357–372. [Google Scholar] [CrossRef] [PubMed]
  36. Strzelecka, M.; Świątek, P. 1,2,4-Triazoles as Important Antibacterial Agents. Pharmaceuticals 2021, 14, 224. [Google Scholar] [CrossRef]
  37. Patil, S.A.; Nesaragi, A.R.; Rodríguez-Berrios, R.R.; Hampton, S.M.; Bugarin, A.; Patil, S.A. Coumarin Triazoles as Potential Antimicrobial Agents. Antibiotics 2023, 12, 160. [Google Scholar] [CrossRef]
  38. Kazeminejad, Z.; Marzi, M.; Shiroudi, A.; Kouhpayeh, S.A.; Farjam, M.; Zarenezhad, E. Novel 1,2,4-Triazoles as Antifungal Agents. BioMed Res. Int. 2022, 2022, 4584846. [Google Scholar] [CrossRef]
  39. Sharma, A.; Agrahari, A.K.; Rajkhowa, S.; Tiwari, V.K. Emerging impact of triazoles as anti-tubercular agent. Eur. J. Med. Chem. 2022, 238, 114454. [Google Scholar] [CrossRef]
  40. El-Shoukrofy, M.S.; Atta, A.; Fahmy, S.; Sriram, D.; Mahran, M.A.; Labouta, I.M. New tetrahydropyrimidine-1,2,3-triazole clubbed compounds: Antitubercular activity and Thymidine Monophosphate Kinase (TMPKmt) inhibition. Bioorg. Chem. 2023, 131, 106312. [Google Scholar] [CrossRef]
  41. Ravisankar, N.; Sarathi, N.; Maruthavanan, T.; Ramasundaram, S.; Ramesh, M.; Sankar, C.; Umamatheswari, S.; Kanthimathi, G.; Oh, T.H. Synthesis, antimycobacterial screening, molecular docking, ADMET prediction and pharmacological evaluation on novel pyran-4-one bearing hydrazone, triazole and isoxazole moieties: Potential inhibitors of SARS-CoV-2. Synthesis, antimycobacterial screening, molecular docking, ADMET prediction and pharmacological evaluation on novel pyran-4-one bearing hydrazone, triazole and isoxazole moieties: Potential inhibitors of SARS-CoV-2. J. Mol. Struct. 2023, 1285, 135461. [Google Scholar] [CrossRef]
  42. Musa, A.; Abulkhair, H.S.; Aljuhani, A.; Rezki, N.; Abdelgawad, M.A.; Shalaby, K.; El-Ghorab, A.H.; Aouad, M.R. Phenylpyrazolone-1,2,3-triazole Hybrids as Potent Antiviral Agents with Promising SARS-CoV-2 Main Protease Inhibition Potential. Pharmaceuticals 2023, 16, 463. [Google Scholar] [CrossRef]
  43. Seliem, I.A.; Panda, S.S.; Girgis, A.S.; Moatasim, Y.; Kandeil, A.; Mostafa, A.; Ali, M.A.; Nossier, E.S.; Rasslan, F.; Srour, A.M.; et al. New quinoline-triazole conjugates: Synthesis, and antiviral properties against SARS-CoV-2. Bioorg. Chem. 2021, 114, 105117. [Google Scholar] [CrossRef]
  44. Venkatesham, P.; Schols, D.; Persoons, L.; Claes, S.; Sangolkar, A.A.; Chedupaka, R.; Vedula, R.R. Synthesis of novel thioalkylated triazolothiazoles and their promising in-vitro antiviral activity. J. Mol. Struct. 2023, 1286, 135573. [Google Scholar] [CrossRef]
  45. Pinheiro, N.G.; Gonzaga, D.T.G.; da Silva, A.R.; Fuly, A.C.; von Ranke, N.L.; Rodrigues, C.R.; Magalhães, B.Q.; Pereira, J.S.; Pacheco, P.A.F.; Silva, A.C.; et al. Triazoles with inhibitory action on P2X7R impaired the acute inflammatory response in vivo and modulated the hemostatic balance in vitro and ex vivo. Inflamm. Res. 2023, 72, 237–250. [Google Scholar] [CrossRef] [PubMed]
  46. Demchenko, S.; Lesyk, R.; Yadlovskyi, O.; Holota, S.; Yarmoluk, S.; Tsyhankov, S.; Demchenko, A. Fused Triazole-Azepine Hybrids as Potential Non-Steroidal Antiinflammatory Agents. Sci. Pharm. 2023, 91, 26. [Google Scholar] [CrossRef]
  47. Bozorov, K.; Zhao, J.; Aisa, H.A. 1,2,3-Triazole-containing hybrids as leads in medicinal chemistry: A recent overview. Bioorg. Med. Chem. 2019, 27, 3511–3531. [Google Scholar] [CrossRef]
  48. Hashem, H.E.; Amr, A.E.-G.E.; Nossier, E.S.; Anwar, M.M.; Azmy, E.M. New Benzimidazole-1,2,4-Triazole-, and 1,3,5-Triazine-Based Derivatives as Potential EGFRWT and EGFRT790M Inhibitors: Microwave-Assisted Synthesis, Anticancer Evaluation, and Molecular Docking Study. ACS Omega 2022, 7, 7155–7171. [Google Scholar] [CrossRef]
  49. Othman, D.I.A.; Hamdi, A.; Tawfik, S.S.; Elgazar, A.A.; Mostafa, A.S. Identification of new benzimidazole-triazole hybrids as anticancer agents: Multi-target recognition, in vitro and in silico studies. J. Enzym. Inhib. Med. Chem. 2023, 38, 2166037. [Google Scholar] [CrossRef]
  50. Gupta, O.; Pradhan, G.T.; Chawla, G. An updated review on diverse range of biological activities of 1,2,4-triazole derivatives: Insight into structure activity relationship. J. Mol. Struct. 2023, 1274 Pt 2, 134487. [Google Scholar] [CrossRef]
  51. Abu-Melha, S.; Azher, O.A.; Alaysuy, O.; Alnoman, R.B.; Abualnaja, M.M.; Althagafi, I.; El-Metwaly, N.M. Synthesis, molecular modeling and antioxidant activity of new thiadiazolyl-triazole analogues. J. Saudi Chem. Soc. 2023, 27, 101596. [Google Scholar] [CrossRef]
  52. Dawbaa, S.; Nuha, D.; Evren, A.E.; Cankiliç, M.Y.; Yurttaş, L.; Turan, G. New oxadiazole/triazole derivatives with antimicrobial and antioxidant properties. J. Mol. Struct. 2023, 1282, 135213. [Google Scholar] [CrossRef]
  53. Zhao, W.; Song, M.; Hua, Y.; Zhu, Y.; Liu, W.; Xia, Q.; Deng, X.; Huang, Y. Design, Synthesis, and Pharmacology of New Triazole-Containing Quinolinones as CNS Active Agents. Molecules 2023, 28, 1987. [Google Scholar] [CrossRef] [PubMed]
  54. Dixit, D.; Verma, P.K.; Marwaha, R.K. A review on ‘triazoles’: Their chemistry, synthesis and pharmacological potentials. J. Iran Chem. Soc. 2021, 18, 2535–2565. [Google Scholar] [CrossRef]
  55. Fallah, Z.; Tajbakhsh, M.; Alikhani, M.; Larijani, B.; Faramarzi, M.A.; Hamedifar, H.; Mohammadi-Khanaposhtani, M.; Mahdavi, M. A review on synthesis, mechanism of action, and structure-activity relationships of 1,2,3-triazole-based α-glucosidase inhibitors as promising anti-diabetic agents. J. Mol. Struct. 2022, 1255, 132469. [Google Scholar] [CrossRef]
  56. Rahim, F.; Ullah, H.; Hussain, R.; Taha, M.; Khan, S.; Nawaz, M.; Nawaz, F.; Gilani, S.J.; Bin Jumah, M.N. Thiadiazole based triazole/hydrazone derivatives: Synthesis, in vitro α-glucosidase inhibitory activity and in silico molecular docking study. J. Mol. Struct. 2023, 1287, 135619. [Google Scholar] [CrossRef]
  57. Kumar, S.; Khokra, S.L.; Yadav, A. Triazole analogues as potential pharmacological agents: A brief review. Future J. Pharm. Sci. 2021, 7, 106. [Google Scholar] [CrossRef]
  58. Kaproń, B.; Łuszczki, J.J.; Siwek, A.; Karcz, T.; Nowak, G.; Zagaja, M.; Andres-Mach, M.; Stasiłowicz, A.; Cielecka-Piontek, J.; Kocki, J.; et al. Preclinical evaluation of 1,2,4-triazole-based compounds targeting voltage-gated sodium channels (VGSCs) as promising anticonvulsant drug candidates. Bioorg. Chem. 2020, 94, 103355. [Google Scholar] [CrossRef]
  59. Chu, X.M.; Wang, C.; Wang, W.-L.; Liang, L.L.; Liu, W.; Gong, K.K.; Sun, K.L. Triazole derivatives and their antiplasmodial and antimalarial activities. Eur. J. Med. Chem. 2019, 166, 206–223. [Google Scholar] [CrossRef]
  60. Xu, M.; Peng, Y.; Zhu, L.; Wang, S.; Ji, J.; Rakesh, K.P. Triazole derivatives as inhibitors of Alzheimer’s disease: Current developments and structure-activity relationships. Eur. J. Med. Chem. 2019, 180, 656–672. [Google Scholar] [CrossRef]
  61. Khan, S.A.; Akhtar, M.J.; Gogoi, U.; Meenakshi, D.U.; Das, A. An Overview of 1,2,3-triazole-Containing Hybrids and Their Potential Anticholinesterase Activities. Pharmaceuticals 2023, 16, 179. [Google Scholar] [CrossRef]
  62. Sooknual, P.; Pingaew, R.; Phopin, K.; Ruankham, W.; Prachayasittikul, S.; Ruchirawat, S.; Prachayasittikul, V. Synthesis and neuroprotective effects of novel chalcone-triazole hybrids. Bioorg. Chem. 2020, 105, 104384. [Google Scholar] [CrossRef]
  63. Manzoor, S.; Almarghalani, D.A.; James, A.W.; Raza, M.K.; Kausar, T.; Nayeem, S.M.; Hoda, N.; Shah, Z.A. Synthesis and Pharmacological Evaluation of Novel Triazole-Pyrimidine Hybrids as Potential Neuroprotective and Anti-neuroinflammatory Agents. Pharm. Res. 2023, 40, 167–185. [Google Scholar] [CrossRef] [PubMed]
  64. Li, G.; Hilgenfeld, R.; Whitley, R.; De Clercq, E. Therapeutic strategies for COVID-19: Progress and lessons learned. Nat. Rev. Drug. Discov. 2023, 22, 449–475. [Google Scholar] [CrossRef] [PubMed]
  65. Panahi, Y.; Gorabi, A.M.; Talaei, S.; Beiraghdar, F.; Akbarzadeh, A.; Tarhriz, V.; Mellatyar, H. An overview on the treatments and prevention against COVID-19. Virol. J. 2023, 20, 23. [Google Scholar] [CrossRef] [PubMed]
  66. Uematsu, T.; Takano, T.; Matsui, H.; Kobayashi, N.; Ōmura, S.; Hanaki, H. Prophylactic administration of ivermectin attenuates SARS-CoV-2 induced disease in a Syrian Hamster Model. J. Antibiot. 2023, 76, 481–488. [Google Scholar] [CrossRef] [PubMed]
  67. Shukla, A.K.; Misra, S. Antimicrobials in COVID-19: Strategies for treating a COVID-19 pandemic. J. Basic Clin. Physiol. Pharmacol. 2022, 1–16. [Google Scholar] [CrossRef] [PubMed]
  68. McCarthy, M.W. Current and emerging immunomodulators for treatment of SARS-CoV-2 infection (COVID-19). Expert Opin. Pharmacother. 2022, 23, 623–628. [Google Scholar] [CrossRef]
  69. Fazio, S.; Bellavite, P. Early Multi-Target Treatment of Mild-to-Moderate COVID-19, Particularly in Terms of Non-Steroidal Anti-Inflammatory Drugs and Indomethacin. BioMed 2023, 3, 177–194. [Google Scholar] [CrossRef]
  70. Mallikanti, V.; Thumma, V.; Matta, R.; Valluru, K.R.; Sharma Konidena, L.N.; Boddu, L.S.; Pochampally, J. Synthesis, antimicrobial activity and molecular docking of novel benzimidazole conjugated 1,2,3-triazole analogues. Chem. Data Collect. 2023, 45, 101034. [Google Scholar] [CrossRef]
  71. Güzel, E.; Çevik, U.A.; Evren, A.E.; Bostancı, H.E.; Gül, U.D.; Kayış, U.; Özkay, Y.; Kaplancıklı, Z.A. Synthesis of Benzimidazole-1,2,4-triazole Derivatives as Potential Antifungal Agents Targeting 14α-Demethylase. ACS Omega 2023, 8, 4369–4384. [Google Scholar] [CrossRef]
  72. Ghobadi, E.; Hashemi, S.M.; Fakhim, H.; Hosseini-khah, Z.; Badali, H.; Emami, S. Design, synthesis and biological activity of hybrid antifungals derived from fluconazole and mebendazole. Eur. J. Med. Chem. 2023, 249, 115146. [Google Scholar] [CrossRef]
  73. Youssif, B.G.M.; Mohamed, Y.A.M.; Salim, M.T.A.; Inagaki, F.; Mukai, C.; Abdu-Allah, H.H.M. Synthesis of some benzimidazole derivatives endowed with 1,2,3-triazole as potential inhibitors of hepatitis C virus. Acta Pharm. 2016, 66, 219–231. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Al-Humaidi, J.Y.; Shaaban, M.M.; Rezki, N.; Aouad, M.R.; Zakaria, M.; Jaremko, M.; Hagar, M.; Elwakil, B.H. 1,2,3-Triazole-Benzofused Molecular Conjugates as Potential Antiviral Agents against SARS-CoV-2 Virus Variants. Life 2022, 12, 1341. [Google Scholar] [CrossRef] [PubMed]
  75. Harkala, K.J.; Eppakayala, L.; Maringanti, T.C. Synthesis and biological evaluation of benzimidazole-linked 1,2,3-triazole congeners as agents. Org. Med. Chem. Lett. 2014, 4, 14. [Google Scholar] [CrossRef] [PubMed]
  76. Chojnacki, K.; Winska, P.; Skierka, K.; Wielechowska, M.; Bretner, M. Synthesis, in vitro antiproliferative activity and kinase profile of new benzimidazole and benzotriazole derivatives. Bioorg. Chem. 2017, 72, 1–10. [Google Scholar] [CrossRef] [PubMed]
  77. Sahay, I.I.; Ghalsasi, P.S. Synthesis of New 1,2,3-triazole Linked Benzimidazole Molecules as Anti-Proliferative Agents. Synth. Commun. 2017, 47, 825–834. [Google Scholar] [CrossRef]
  78. Aouad, M.R.; Soliman, M.A.; Alharbi, M.O.; Bardaweel, S.K.; Sahu, P.K.; Ali, A.A.; Messali, M.; Rezki, N.; Al-Soud, Y.A. Design, Synthesis and Anticancer Screening of Novel Benzothiazole-Piperazine-1,2,3-Triazole Hybrids. Molecules 2018, 23, 2788. [Google Scholar] [CrossRef] [Green Version]
  79. Bistrović, A.; Krstulović, L.; Harej, A.; Grbčić, P.; Sedić, M.; Koštrun, S.; Kraljević, S.P.; Bajić, M.; Raić-Malić, S. Design, synthesis and biological evaluation of novel benzimidazole amidines as potent multi-target inhibitors for the treatment of non-small cell lung cancer. Eur. J. Med. Chem. 2018, 143, 1616–1634. [Google Scholar] [CrossRef]
  80. Aouad, M.R.; Almehmadi, M.A.; Rezki, N.; Al-blewi, F.F.; Messali, M.; Ali, I. Design, click synthesis, anticancer screening and docking studies of novel benzothiazole-1,2,3-triazoles appended with some bioactive benzofused heterocycles. J. Mol. Struct. 2019, 1188, 153–164. [Google Scholar] [CrossRef]
  81. Meščić Macan, A.; Perin, N.; Jakopec, S.; Mioč, M.; Radić Stojković, M.; Kralj, M.; Hranjec, M.; Raić-Malić, S. Synthesis, antiproliferative activity and DNA/RNA-binding properties of mono- and bis-(1,2,3-triazolyl)-appended benzimidazo[1,2-a]quinoline derivatives. Eur. J. Med. Chem. 2020, 185, 111845. [Google Scholar] [CrossRef]
  82. Ashok, D.; Reddy, M.R.; Nagaraju, N.; Dharavath, R.; Ramakrishna, K.; Gundu, S.; Shravani, P.; Sarasija, M. Microwave-assisted synthesis and in vitro antiproliferative activity of some novel 1,2,3-triazole-based pyrazole aldehydes and their benzimidazole derivatives. Med. Chem. Res. 2020, 29, 699–706. [Google Scholar] [CrossRef]
  83. Djemoui, A.; Naouri, A.; Ridha Ouahrani, M.; Djemoui, D.; Lahcene, S.; Boualem Lahrech, M.; Boukenna, L.; Albuquerque, H.M.T.; Saher, L.; Rocha, D.H.A.; et al. A step-by-step synthesis of triazole-benzimidazole-chalcone hybrids: Anticancer activity in human cells+. J. Mol. Struct. 2020, 1204, 127487. [Google Scholar] [CrossRef]
  84. Sridhar Goud, N.; Pooladanda, V.; Chandra, K.M.; Soukya, P.S.L.; Alvala, R.; Kumar, P.; Nagaraj, C.; Dawn Bharath, R.; Qureshi, I.A.; Godugu, C.; et al. Novel benzimidazole-triazole hybrids as apoptosis inducing agents in lung cancer: Design, synthesis, 18F-radiolabeling & galectin-1 inhibition studies. Bioorg. Chem. 2020, 102, 104125. [Google Scholar] [CrossRef]
  85. Singu, P.S.; Chilakamarthi, U.; Mahadik, N.S.; Keerti, B.; Valipenta, N.; Mokale, S.N.; Nagesh, N.; Kumbhare, R.M. Benzimidazole-1,2,3-triazole hybrid molecules: Synthesis and study of their interaction with G-quadruplex DNA. RSC Med. Chem. 2021, 12, 416. [Google Scholar] [CrossRef]
  86. Alzahrani, H.A.; Mahboob Alam, M.; Elhenawy, A.A.; Malebari, A.M.; Nazreen, S. Synthesis, antiproliferative, docking and DFT studies of benzimidazole derivatives as EGFR inhibitors. J. Mol. Struct. 2022, 1253, 132265. [Google Scholar] [CrossRef]
  87. Husain, A.; Rashid, M.; Mishra, R.; Parveen, S.; Shin, D.-S.; Kumar, D. Benzimidazole bearing oxadiazole and triazolo-thiadiazoles nucleus: Design and synthesis as anticancer agents. Bioorg. Med. Chem. Lett. 2012, 22, 5438–5444. [Google Scholar] [CrossRef] [PubMed]
  88. Husain, A.; Rashid, M.; Shaharyar, M.; Siddiqui, A.A.; Mishra, R. Benzimidazole clubbed with triazolo-thiadiazoles and triazolo-thiadiazines: New anticancer agents. Eur. J. Med. Chem. 2013, 62, 785–798. [Google Scholar] [CrossRef]
  89. Temirak, A.; Shaker, Y.M.; Ragab, F.A.F.; Ali, M.M.; Ali, H.I.; El Diwani, H.I. Part I. Synthesis, biological evaluation and docking studies of new 2-furylbenzimidazoles as antiangiogenic agents. Eur. J. Med. Chem. 2014, 87, 868–880. [Google Scholar] [CrossRef]
  90. Ibrahim, H.A.; Awadallah, F.M.; Refaat, H.M.; Amin, K.M. Molecular docking simulation, synthesis and 3D pharmacophore studies of novel 2-substituted-5-nitro-benzimidazole derivatives as anticancer agents targeting VEGFR-2 and c-Met. Bioorg. Chem. 2018, 77, 457–470. [Google Scholar] [CrossRef]
  91. Celik, I.; Ayhan-Kılcıgil, G.; Guven, B.; Kara, Z.; Gurkan-Alp, A.S.; Karayel, A.; Onay-Besikci, A. Design, synthesis and docking studies of benzimidazole derivatives as potential EGFR inhibitors. Eur. J. Med. Chem. 2019, 173, 240–249. [Google Scholar] [CrossRef]
  92. Mancini, R.S.; Barden, C.J.; Weaver, D.F.; Reed, M.A. Furazans in Medicinal Chemistry. J. Med. Chem. 2021, 64, 1786–1815. [Google Scholar] [CrossRef] [PubMed]
  93. Kus, C.; Ayhan-Kilcigil, G.; Can Eke, B.; IŞcan, M. Synthesis and antioxidant properties of some novel benzimidazole derivatives on lipid peroxidation in the rat liver. Arch. Pharm. Res. 2004, 27, 156–163. [Google Scholar] [CrossRef] [PubMed]
  94. Ayhan-Kilcigil, G.; Kus, C.; Çoban, T.; Can-Eke, B.; Iscan, M. Synthesis and Antioxidant Properties of Novel Benzimidazole Derivatives. J. Enzyme Inhib. Med. Chem. 2004, 19, 129–135. [Google Scholar] [CrossRef] [Green Version]
  95. Karaali, N. Synthesis of Some New Benzimidazole Derivatives Containing Chlorine and Investigation of Their Antioxidant and Anti-urease Activities. JOTCSA 2018, 5, 971–980. [Google Scholar] [CrossRef] [Green Version]
  96. Faraji, L.; Shahkarami, S.; Nadri, H.; Moradi, A.; Saeedi, M.; Foroumadi, A.; Ramazani, A.; Haririan, I.; Reza Ganjali, M.; Shafiee, A.; et al. Synthesis of novel benzimidazole and benzothiazole derivatives bearing a 1,2,3-triazole ring system and their acetylcholinesterase inhibitory activity. J. Chem. Res. 2017, 41, 30–35. [Google Scholar] [CrossRef]
  97. Unsal-Tan, O.; Ozadali-Sari, K.; Ayazgok, B.; Küçükkılınç, T.T.; Balkan, A. Novel 2-Arylbenzimidazole derivatives as multi-targeting agents to treat Alzheimer’s disease. Med. Chem. Res. 2017, 26, 1506–1515. [Google Scholar] [CrossRef]
  98. Acar Cevik, U.; Saglik, B.N.; Levent, S.; Osmaniye, D.; Kaya Cavuşoglu, B.; Ozkay, Y.; Kaplancikli, Z.A. Synthesis and AChE-Inhibitory Activity of New Benzimidazole Derivatives. Molecules 2019, 24, 861. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Khan, Y.; Rehman, W.; Hussain, R.; Khan, S.; Malik, A.; Khan, M.; Liaqat, A.; Rasheed, L.; Begum, F.; Fazil, S.; et al. New biologically potent benzimidazole-based-triazole derivatives as acetylcholinesterase and butyrylcholinesterase inhibitors along with molecular docking study. J. Heterocycl. Chem. 2022, 52, 2225–2239. [Google Scholar] [CrossRef]
  100. Özil, M.; Emirik, M.; Etlik, S.Y.; Kahveci, B. A simple and efficient synthesis of novel inhibitors of alpha-glucosidase based on benzimidazole skeleton and molecular docking studies. Bioorg. Chem. 2016, 68, 226–235. [Google Scholar] [CrossRef]
  101. Deswal, L.; Verma, V.; Kumar, D.; Kaushik, C.P.; Kumar, A.; Deswal, Y.; Punia, S. Synthesis and antidiabetic evaluation of benzimidazole-tethered 1,2,3-triazoles. Arch. Pharm. 2020, 353, 2000090. [Google Scholar] [CrossRef] [PubMed]
  102. Asemanipoor, N.; Mohammadi-Khanaposhtani, M.; Moradi, S.; Vahidi, M.; Asadi, M.; Faramarzi, M.A.; Mahdavi, M.; Biglar, M.; Larijani, B.; Hamedifar, H.; et al. Synthesis and biological evaluation of new benzimidazole-1,2,3-triazole hybrids as potential α-glucosidase inhibitors. Bioorg. Chem. 2020, 95, 103482. [Google Scholar] [CrossRef]
  103. Esfahani, A.N.; Iraji, A.; Alamir, A.; Moradi, S.; Sadegh Asgari, M.; Hosseini, S.; Mojtabavi, S.; Nasli-Esfahani, E.; Faramarzi, M.A.; Bandarian, F.; et al. Design and synthesis of phenoxymethybenzoimidazole incorporating different aryl thiazole-triazole acetamide derivatives as α-glycosidase inhibitors. Mol. Divers. 2022, 26, 1995–2009. [Google Scholar] [CrossRef]
  104. Devi, M.; Kumar, P.; Singh, R.; Sindhu, J.; Kataria, R. Design, synthesis, spectroscopic characterization, single crystal X-ray analysis, in vitro α-amylase inhibition assay, DPPH free radical evaluation and computational studies of naphtho [2,3-d]imidazole-4,9-dione appended 1,2,3-triazoles. Eur. J. Med. Chem. 2023, 250, 115230. [Google Scholar] [CrossRef] [PubMed]
  105. Nandha, B.; Ramareddy, S.A.; Kuntal, H. Synthesis of substituted fluorobenzimidazoles as inhibitors of 5-lipoxygenase and soluble epoxide hydrolase for anti-inflammatory activity. Arch. Pharm. 2018, 351, 1800030. [Google Scholar] [CrossRef] [PubMed]
  106. Kondengadan, S.M.; Bansal, S.; Yang, C.; Liu, D.; Fultz, Z.; Wang, B. Click chemistry and drug delivery: A bird’s-eye view. Acta Pharm. Sin. B 2023, 13, 1990–2016. [Google Scholar] [CrossRef]
  107. de Souza, R.O.M.A.; de Mariz Miranda, L.S. Strategies towards the Synthesis of N2-Substituted 1,2,3-Triazoles. An. Acad. Bras. Ciênc. 2019, 91 (Suppl. S1), e20180751. [Google Scholar] [CrossRef]
  108. Dai, J.; Tian, S.; Yang, X.; Liu, Z. Synthesis methods of 1,2,3-/1,2,4-triazoles: A review. Front. Chem. 2022, 10, 891484. [Google Scholar] [CrossRef]
  109. De Nino, A.; Maiuolo, L.; Costanzo, P.; Algieri, V.; Jiritano, A.; Olivito, F.; Tallarida, M.A. Recent Progress in Catalytic Synthesis of 1,2,3-Triazoles. Catalysts 2021, 11, 1120. [Google Scholar] [CrossRef]
  110. Koranne, A.; Kurrey, K.; Kumar, P.; Gupta, S.; Jha, K.V.; Ravi, R.; Sahu, K.P.; Anamika; Jha, A.K. Metal catalyzed C–H functionalization on triazole rings. RSC Adv. 2022, 12, 27534. [Google Scholar] [CrossRef]
  111. Marinescu, M. Chemistry and Applications of Benzimidazole and Its Derivatives; IntechOpen: London, UK, 2019; pp. 1–213. [Google Scholar]
  112. Marinescu, M. Chiral benzimidazoles in medicinal chemistry: Syntheses and applications. In Benzimidazole: Preparation and Applications; Vestergaard, A.A., Ed.; Nova Science Publishers: New York, NY, USA, 2020; pp. 87–112. [Google Scholar]
  113. Zalaru, C.-M.; Marinescu, M. Benzimidazole compounds with anti-tumor and antibacterial activities. In Benzimidazole: Preparation and Applications; Vestergaard, A.A., Ed.; Nova Science Publishers: New York, NY, USA, 2020; pp. 221–250. [Google Scholar]
  114. Marinescu, M.; Tudorache, D.G.; Marton, G.I.; Zalaru, C.M.; Popa, M.; Chifiriuc, M.C.; Stavarache, C.E.; Constantinescu, C. Density functional theory molecular modeling, chemical synthesis, and antimicrobial behaviour of selected benzimidazole derivatives. J. Mol. Struct. 2017, 1130, 463–471. [Google Scholar] [CrossRef]
  115. Marinescu, M.; Cinteza, L.O.; Marton, G.I.; Chifiriuc, M.C.; Popa, M.; Stanculescu, I.; Zalaru, C.M.; Stavarache, C.E. Synthesis, density functional theory study and in vitro antimicrobial evaluation of new benzimidazole Mannich bases. BMC Chem. 2020, 14, 45. [Google Scholar] [CrossRef] [PubMed]
  116. Marinescu, M. Nitrogen-containing heterocycles as corrosion inhibitors. In Corrosion Inhibitors: An Overview; Wilkerson, R., Ed.; Nova Science Publishers: New York, NY, USA, 2021; pp. 161–201. [Google Scholar]
  117. Qiu, J.; Zou, Y.; Li, S.; Yang, L.; Qiu, Z.; Kong, F.; Gu, X. Discovery of benzimidazole substituted 1, 2, 4-oxadiazole compounds as novel anti-HBV agents with TLR8-agonistic activities. Eur. J. Med. Chem. 2022, 244, 114833. [Google Scholar] [CrossRef] [PubMed]
  118. Youssif, B.G.M.; Abdel-Moty, S.G.; Sayed, Y.B. Synthesis and biologicalevaluation of some novel 1,2,3-triazol-N-arylidene acetohydrazide incorporating benzimidazole ring moiety as potential antimicrobial agents. J. Curr. Chem. Pharm. Sc. 2014, 4, 54–64. [Google Scholar]
  119. Al-blewi, F.F.; Almehmadi, M.A.; Aouad, M.R.; Bardaweel, S.K.; Sahu, P.K.; Messali, M.; Rezki, N.; El Ashry, L.S.H. Design, synthesis, ADME prediction and pharmacological evaluation of novel benzimidazole-1,2,3-triazole-sulfonamide hybrids as antimicrobial and antiproliferative agents. Chem. Cent. J. 2018, 12, 110. [Google Scholar] [CrossRef] [Green Version]
  120. Biswas, D.; Roy, S.; Sen, S. A simple approach for indexing the oral drug likeness of a compound: Discriminating drug like compounds from nondrug like ones. J. Chem. Inf. Model. 2006, 46, 1394–1401. [Google Scholar] [CrossRef]
  121. Lipinski, C.A.; Lombardo, F.; Dominy, B.W.; Feene, P.J. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv. Drug. Deliv. Rev. 2001, 46, 3–26. [Google Scholar] [CrossRef]
  122. Rashdan, H.R.M.; Abdelmonsef, A.H.; Abou-Krisha, M.M.; Yousef, T.A. Synthesis, Identification, Computer-Aided Docking Studies, and ADMET Prediction of Novel Benzimidazo-1,2,3-triazole Based Molecules as Potential Antimicrobial Agents. Molecules 2021, 26, 7119. [Google Scholar] [CrossRef]
  123. Dallakyan, S.; Olson, A.J. Small-Molecule Library Screening by Docking with PyRx. In Chemical Biology; Springer: Berlin/Heidelberg, Germany, 2015; Volume 1263, pp. 243–250. ISBN 9780123944474. [Google Scholar]
  124. Ouahrouch, A.; Ighachane, H.; Taourirte, M.; Engels, J.W.; Sedra, M.H.; Lazrek, H.B. Benzimidazole-1,2,3-triazole Hybrid Molecules: Synthesis and Evaluation for Antibacterial/Antifungal Activity. Arch. Pharm. Chem. Life Sci. 2014, 347, 748–755. [Google Scholar] [CrossRef] [Green Version]
  125. Bistrović, A.; Krstulović, L.; Stolić, I.; Drenjancević, D.; Talapko, J.; Taylor, M.C.; Kelly, J.M.; Bajić, M.; Raic-Malić, S. Synthesis, anti-bacterial and anti-protozoal activities of amidinobenzimidazole derivatives and their interactions with DNA and RNA. J. Enzyme Inhib. Med. Chem. 2018, 33, 1323–1334. [Google Scholar] [CrossRef]
  126. Rao, Y.J.; Sowjanya, T.; Thirupathi, G.; Murthy, N.Y.S.; Kotapalli, S.S. Synthesis and biological evaluation of novel flavone/triazole/benzimidazole hybrids and flavone/isoxazole-annulated heterocycles as antiproliferative and antimycobacterial agents. Mol. Div. 2018, 22, 803–814. [Google Scholar] [CrossRef] [PubMed]
  127. Ashok, D.; Gundu, S.; Aamate, V.K.; Devulapally, M.G. Conventional andmicrowave-assisted synthesis of new indole-tethered benzimidazole-based 1,2,3-triazoles and evaluation of their antimycobacterial, antioxidant and antimicrobial activities. Mol. Div. 2018, 22, 769–778. [Google Scholar] [CrossRef]
  128. Actelion Pharmaceuticals Ltd. OSIRIS Property Explorer Software, Version 2; Actelion Pharmaceuticals Ltd.: Allschwil, Switzerland, 2021; Available online: http://www.organic-chemistry.org/prog/peo/ (accessed on 29 June 2023).
  129. Kurczab, R.; Kucwaj-Brysz, K.; Śliwa, P. The Significance of Halogen Bonding in Ligand–Receptor Interactions: The Lesson Learned from Molecular Dynamic Simulations of the D4 Receptor. Molecules 2020, 25, 91. [Google Scholar] [CrossRef] [Green Version]
  130. Chandrika, N.T.; Shrestha, S.K.; Ranjan, N.; Sharma, A.; Arya, D.P.; Garneau-Tsodikova, S. New Application of Neomycin B−Bisbenzimidazole Hybrids as Antifungal Agents. ACS Infect. Dis. 2018, 4, 196–207. [Google Scholar] [CrossRef] [PubMed]
  131. Saber, A.; Anouar, E.H.; Sebbar, G.; El Ibrahimi, B.; Srhir, M.; Hökelek, T.; Mague, J.T.; El Ghayati, L.; Sebbar, N.K.; Essassi, E.M. New 1,2,3-triazole containing benzimidazolone derivatives: Syntheses, crystal structures, spectroscopic characterizations, Hirshfeld surface analyses, DFT calculations, anti-corrosion property anticipation, and antibacterial activities. J. Mol. Struct. 2021, 1242, 130719. [Google Scholar] [CrossRef]
  132. Mohsen, D.H.; Radhi, A.J.; Shaheed, D.Q.; Abbas, H.K. Synthesis New Benzimidazole Derivatives as Antibacterial. J. Pharm. Negat. Results 2022, 13, 893–898. [Google Scholar] [CrossRef]
  133. Rezki, N. Green Microwave Synthesis and Antimicrobial Evaluation of Novel Triazoles. Org. Prep. Proc. Int. 2017, 49, 525–541. [Google Scholar] [CrossRef]
  134. Milite, C.; Amendola, G.; Nocentini, A.; Bua, S.; Cipriano, A.; Barresi, E.; Feoli, A.; Novellino, E.; Da Settimo, F.; Supuran, C.T.; et al. Novel 2-substituted-benzimidazole-6-sulfonamides as carbonic anhydrase inhibitors: Synthesis, biological evaluation against isoforms I, II, IX and XII and molecular docking studies. J. Enzyme Inhib. Med. Chem. 2019, 34, 1697–1710. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Aparna, Y.; Nirmala, G.; Subhashini, N.J.P.; Sharada, L.N.; Sreekanth, S. Synthesis and Antimicrobial Activity of Novel Bis-1,2,3-triazol-1H-4-yl-substituted Aryl Benzimidazole-2-thiol Derivatives. Russ. J. Gen. Chem. 2020, 90, 1501–1506. [Google Scholar] [CrossRef]
  136. Gill, C.; Jadhav, G.; Shaikh, M.; Kale, R.; Ghawalkar, A.; Nagargoje, D.; Shiradkar, M. Clubbed [1,2,3] triazoles by fluorine benzimidazole: A novel approach to H37Rv inhibitors as a potential treatment for tuberculosis. Bioorg. Med. Chem. Lett. 2008, 18, 6244–6247. [Google Scholar] [CrossRef]
  137. Anand, A.; Kulkarni, M.V.; Joshi, S.D.; Dixit, S.R. One pot Click chemistry: A three component reaction for the synthesis of 2-mercaptobenzimidazole linked coumarinyl triazoles as anti-tubercular agents. Bioorg. Med. Chem. Lett. 2016, 26, 4709–4713. [Google Scholar] [CrossRef] [PubMed]
  138. Khanapurmath, N.; Kulkarni, M.V.; Joshi, S.D.; Kumar, G.N.A. A click chemistry approach for the synthesis of cyclic ureido tethered coumarinyl and 1-aza coumarinyl 1,2,3-triazoles as inhibitors of Mycobacterium tuberculosis H37Rv and their in silico studies. Bioorg. Med. Chem. 2019, 27, 115054. [Google Scholar] [CrossRef]
  139. Pandey, V.K.; Upadhyay, M.; Upadhyay, M.; Gupta, V.D.; Tandon, M. Benzimidazolyl quinolinyl mercaptotriazoles as potential antimicrobial and antiviral agents. Acta Pharm. 2005, 55, 47–56. [Google Scholar] [PubMed]
  140. Jadhav, G.R.; Shaikh, M.U.; Kale, R.P.; Shiradkar, M.R.; Gill, C.H. SAR study of clubbed [1,2,4]-triazolyl with fluorobenzimidazoles as antimicrobial and antituberculosis agents. Eur. J. Med. Chem. 2009, 44, 2930–2935. [Google Scholar] [CrossRef]
  141. Barot, K.P.; Manna, K.S.; Ghate, M.D. Design, synthesis and antimicrobial activities of some novel 1,3,4-thiadiazole, 1,2,4-triazole-5-thione and 1,3-thiazolan-4-one derivatives of benzimidazole. J. Saudi Chem. Soc. 2017, 21, S35–S43. [Google Scholar] [CrossRef] [Green Version]
  142. Jiang, Z.; Wang, Y.; Wang, W.; Wang, S.; Xu, B.; Fan, G.; Dong, G.; Liu, Y.; Yao, J.; Miao, Z.; et al. Discovery of highly potent triazole antifungal derivatives by heterocycle-benzene bioisosteric replacement. Eur. J. Med. Chem. 2013, 64, 16–22. [Google Scholar] [CrossRef] [PubMed]
  143. Luo, Y.-L.; Baathulaa, K.; Kannekanti, V.K.; Zhou, C.-H.; Cai, G.-X. Novel benzimidazole derived naphthalimide triazoles: Synthesis, antimicrobial activity and interactions with calf thymus DNA. Sci. China Chem. 2015, 58, 483–494. [Google Scholar] [CrossRef]
  144. Ahmadi, A. Synthesis an antibacterial evaluation of ome novel Mannich bases of benzimidazole derivatives. Bull. Chem. Soc. Ethiop. 2016, 30, 421–425. [Google Scholar] [CrossRef] [Green Version]
  145. Kankate, R.S.; Gide, P.S.; Belsare, D.P. Design, synthesis and antifungal evaluation of novel benzimidazole tertiary amine type of fluconazole analogues. Arab. J. Chem. 2019, 12, 2224–2235. [Google Scholar] [CrossRef] [Green Version]
  146. Pham, E.C.; Vi Le Thi, T.; Ly Hong, H.H.; Vo Thi, B.N.; Vong, L.B.; Vu, T.T.; Vo, D.D.; Nguyen, N.V.T.; Bao Le, K.N.; Truong, T.N. N,2,6-Trisubstituted 1H-benzimidazole derivatives as a new scaffold of antimicrobial and anticancer agents: Design, synthesis, in vitro evaluation, and in silico studies. RSC Adv. 2023, 13, 399–420. [Google Scholar] [CrossRef]
  147. Ahuja, R.; Sidhu, A.; Bala, A.; Arora, D.; Sharma, P. Structure based approach for twin-enzyme targeted benzimidazolyl-1,2,4-triazole molecular hybrids as antifungal agents. Arab. J. Chem. 2020, 13, 5832–5848. [Google Scholar] [CrossRef]
  148. Evren, A.E.; Celik, I.; Akar Cevik, U. Synthesis, molecular docking, in silico ADME and antimicrobial activity studies of some new benzimidazole-triazole derivatives. Cumhur. Sci. J. 2021, 42, 795–805. [Google Scholar]
  149. Ansari, K.F.; Lal, C.; Khitoliya, R.K. Synthesis and biological activity of some triazole-bearing benzimidazole derivatives. J. Serb. Chem. Soc. 2011, 76, 341–352. [Google Scholar] [CrossRef]
  150. Tien, C.N.; Cam, D.T.T.; Manh, H.B.; Dang, D.N. Synthesis and Antibacterial Activity of Some Derivatives of 2-Methylbenzimidazole Containing 1,3,4-Oxadiazole or 1,2,4-Triazole Heterocycle. J. Chem. 2016, 2016, 1507049. [Google Scholar] [CrossRef]
  151. Kantar, G.K.; Mentese, E.; Beris, F.S.; Şasmaz, S.; Kahveci, B. Synthesis and antimicrobial activity of some new triazole bridged benzimidazole substituted phthalonitrile and phthalocyanines. Rev. Roum. Chim. 2018, 63, 59–65. [Google Scholar]
  152. Nandwana, N.K.; Singh, R.P.; Patel, O.P.S.; Dhiman, S.; Saini, H.K.; Jha, P.N.; Kumar, A. Design and Synthesis of Imidazo/Benzimidazo[1,2-c] quinazoline Derivatives and Evaluation of Their Antimicrobial Activity. ACS Omega 2018, 3, 16338–16346. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Al-Majidi, S.M.H.; Ibrahim, H.A.R.; AL-issa, A.H. Synthesis and Identification of Some New Derivatives of ([Benzyl Thio) Benzimidazole N-(Methylene-5-Yl)]-4,5-Di Substituted 1,2,4-Triazole and Evaluation of Their Activity as Antimicrobial and Anti-Inflammatory Agents. Iraqi J. Sci. 2021, 62, 1054–1065. [Google Scholar] [CrossRef]
  154. El-masry, A.H.; Fahmy, H.H.; Ali Abdelwahed, S.H. Synthesis and Antimicrobial Activity of Some New Benzimidazole Derivatives. Molecules 2000, 5, 1429–1438. [Google Scholar] [CrossRef] [Green Version]
  155. Menteşe, E.; Ülker, S.; Kahveci, B. Synthesis and study of α-glucosidase inhibitory, antimicrobial and antioxidant activities of some benzimidazole derivatives containing triazole, thiadiazole, oxaiazole and morpholine rings. Chem. Heterocycl. Comp. 2015, 50, 1671–1682. [Google Scholar] [CrossRef]
  156. Karale, B.K.; Nirmal, P.R.; Akolkar, H.N. Synthesis and in vitro biological screening of some benzimidazolyl anchored azoles. Ind. J. Chem. 2015, 54, 399–405. [Google Scholar]
  157. Madawali, I.M.; Gaviraj, E.N.; Kalyane, N.V.; Shivakumar, B. A Review On Substituted Benzimidazoles: Biologically Active Compounds. Am. J. Pharm. Tech. Res. 2019, 9, 256–274. [Google Scholar] [CrossRef]
  158. Eisa, H.M.; Barghash, A.-E.M.; Badr, S.M.; Farahat, A.A. Synthesis and antimicrobial activity of certain benzimidazole and fused benzimidazole derivatives. Ind. J. Chem. 2010, 49, 1515–1525. [Google Scholar]
  159. Nevade, S.A.; Lokapure, S.G.; Kalyane, N.V. Synthesis and Pharmacological Evaluation of Some Novel 2-Mercapto Benzimidazole Derivatives. Kor. Che. Soc. 2013, 57, 755–760. [Google Scholar] [CrossRef] [Green Version]
  160. Can, N.O.; Çevik, U.A.; Saglik, B.N.; Levent, S.; Korkut, B.; Özkay, Y.; Kaplancikli, Z.A.; Koparal, A.S. Synthesis, Molecular Docking Studies, and Antifungal Activity Evaluation of New Benzimidazole-Triazoles as Potential Lanosterol 14α-Demethylase Inhibitors. I. Chem. 2017, 2017, 9387102. [Google Scholar] [CrossRef] [Green Version]
  161. Karaca Gençer, H.; Acar Çevik, U.; Levent, S.; Sağlık, B.N.; Korkut, B.; Özkay, Y.; Ilgın, S.; Öztürk, Y. New Benzimidazole-1,2,4-Triazole Hybrid Compounds: Synthesis, Anticandidal Activity and Cytotoxicity Evaluation. Molecules 2017, 22, 507. [Google Scholar] [CrossRef] [Green Version]
  162. Omar, R.A.; Koparir, P.; Sarac, K.; Koparir, M.; Safin, D. A novel coumarin-triazole-thiophene hybrid: Synthesis, characterization, ADMET prediction, molecular docking and molecular dynamics studies with a series of SARS-CoV-2 proteins. J. Chem. Sci. 2023, 135, 6. [Google Scholar] [CrossRef] [PubMed]
  163. Shalaby, M.A.; Fahim, A.M.; Rizk, S.A. Microwave-assisted synthesis, antioxidant activity, docking simulation, and DFT analysis of diferent heterocyclic compounds. Sci. Rep. 2023, 13, 4999. [Google Scholar] [CrossRef] [PubMed]
  164. Chahat; Bhatia, R.; Kumar, B. p53 as a potential target for treatment of cancer: A perspective on recent advancements in small molecules with structural insights and SAR studies. Eur. J. Med. Chem. 2023, 247, 115020. [Google Scholar] [CrossRef] [PubMed]
  165. Aatif, M.; Raza, M.A.; Javed, K.; Nashre-ul-Islam, S.M.; Farhan, M.; Alam, M.W. Potential Nitrogen-Based Heterocyclic Compounds for Treating Infectious Diseases: A Literature Review. Antibiotics 2022, 11, 1750. [Google Scholar] [CrossRef]
  166. Breijyeh, Z.; Karaman, R. Design and Synthesis of Novel Antimicrobial Agents. Antibiotics 2023, 12, 628. [Google Scholar] [CrossRef]
  167. Abuelizz, H.A.; Bakheit, A.H.; Al-Agamy, M.H.; Rashid, H.; Mostafa, G.A.E.; Al-Salahi, R. Benzo[g]quinazolines as antifungal against candidiasis: Screening, molecular docking, and QSAR investigations. Saudi Pharm. J. 2023, 31, 815–823. [Google Scholar] [CrossRef]
  168. Aryal, P.; Shakya, B.J. Synthesis, Cytotoxicity, Antibacterial and Antioxidant Activity of New 2-Substituted Benzimidazole Containing 1,2,4-Triazoles. Nepal Chem. Soc. 2023, 43, 34–45. [Google Scholar] [CrossRef]
  169. Nandha, B.; Nargund, L.V.G.; Nargund, S.L. Design and synthesis of some new imidazole and 1,2,4-triazole substituted fluorobenzimidazoles for antitubercular and antifungal activity. Pharma Chem. 2013, 5, 317–327. [Google Scholar]
  170. Karim, M.; Lo, V.-W.; Einav, S. Preparing for the next viral threat with broad-spectrum antivirals. J. Clin. Investig. 2023, 133, e170236. [Google Scholar] [CrossRef]
  171. Roy, A.; Roy, M.; Gacem, A.; Datta, S.; Zeyaullah, M.; Muzammil, K.; Farghaly, T.A.; Abdellattif, M.H.; Yadav, K.K.; Simal-Gandara, J. Role of bioactive compounds in the treatment of hepatitis: A review. Front. Pharmacol. 2022, 13, 1051751. [Google Scholar] [CrossRef]
  172. Warren, T.K.; Jordan, R.; Lo, M.K.; Ray, A.S.; Mackman, R.L.; Soloveva, V.; Siegel, D.; Perron, M.; Bannister, R.; Hui, H.C.; et al. Therapeutic efficacy of the small molecule GS-5734 against Ebola virus in rhesus monkeys. Nature 2016, 531, 381–385. [Google Scholar] [CrossRef] [Green Version]
  173. Cox, R.M.; Wolf, J.D.; Plemper, R.K. Therapeutically administered ribonucleoside analogue MK-4482/EIDD-2801 blocks SARS-CoV-2 transmission in ferrets. Nat. Microbiol. 2021, 6, 11–18. [Google Scholar] [CrossRef] [PubMed]
  174. Tonelli, M.; Paglietti, G.; Boido, V.; Sparatore, F.; Marongiu, F.; Marongiu, E.; La Colla, P.; Loddo, R. Antiviral Activity of Benzimidazole Derivatives. I. Antiviral Activity of1-Substituted-2-[(Benzotriazol-1/2-yl)methyl]benzimidazoles). Chem. Biodivers. 2008, 5, 2386–2401. [Google Scholar] [CrossRef]
  175. Hillary, V.E.; Ceasar, S.A. An update on COVID-19: SARS-CoV-2 variants, antiviral drugs, and vaccines. Heliyon 2023, 9, e13952. [Google Scholar] [CrossRef] [PubMed]
  176. Polatoğlu, I.; Oncu-Oner, T.; Dalman, I.; Ozdogan, S. COVID-19 in early 2023: Structure, replication mechanism, variants of SARS-CoV-2, diagnostic tests, and vaccine & drug development studies. MedComm 2023, 4, e228. [Google Scholar] [CrossRef]
  177. El-Sebaey, S.A. Recent Advances in 1,2,4-Triazole Scaffolds as Antiviral Agents. Chem. Sel. 2020, 5, 11654–11680. [Google Scholar] [CrossRef]
  178. Ion, V.; Matei, A.; Constantinescu, C.; Ionita, I.; Marinescu, M.; Dinescu, M.; Emandi, A. Octahydroacridine thin films grown by matrix-assisted pulsed laser evaporation for non linear optical applications. Mater. Sci. Semicond. Process. 2015, 36, 78–83. [Google Scholar] [CrossRef]
  179. Zalaru, C.; Dumitrascu, F.; Draghici, C.; Tarcomnicu, I.; Tatia, R.; Moldovan, L.; Chifiriuc, M.C.; Lazar, V.; Marinescu, M.; Nitulescu, M.G.; et al. Synthesis, spectroscopic characterization, DFT study and antimicrobial activity of novel alkylaminopyrazole derivatives. J. Mol. Struct. 2018, 1156, 12–21. [Google Scholar] [CrossRef]
  180. Zalaru, C.; Dumitrascu, F.; Draghici, C.; Iovu, M.; Marinescu, M.; Tarcomnicu, I.; Nitulescu, G.M. Synthesis and biological screening of some novel 2-(1H-pyrazol-1-yl)-acetamides as lidocaine analogue. Ind. J. Chem. B 2014, 53, 733–739. [Google Scholar]
Figure 1. Chemical structures of some benzimidazole, 1,2,3-triazole, and 1,2,4-triazole-based marketed drugs.
Figure 1. Chemical structures of some benzimidazole, 1,2,3-triazole, and 1,2,4-triazole-based marketed drugs.
Antibiotics 12 01220 g001
Scheme 1. Synthesis of benzimidazole-1,2,3-triazole hybrids 2a2l and 3a3f.
Scheme 1. Synthesis of benzimidazole-1,2,3-triazole hybrids 2a2l and 3a3f.
Antibiotics 12 01220 sch001
Scheme 2. Synthesis of benzimidazole-1,2,3-triazole hybrids 6a6f.
Scheme 2. Synthesis of benzimidazole-1,2,3-triazole hybrids 6a6f.
Antibiotics 12 01220 sch002
Scheme 3. Synthesis of benzimidazole-1,2,3-triazole hybrids 8a8b, 9a9b, and 10a10b.
Scheme 3. Synthesis of benzimidazole-1,2,3-triazole hybrids 8a8b, 9a9b, and 10a10b.
Antibiotics 12 01220 sch003
Figure 2. The molecular interactions of the best-docked compounds 10a and 10b with the target enzyme DNA gyrase B. Left side (2D): the residues are represented in 3 letter codes. Hydrogen bonds are represented by green and blue lines, and π-interactions are represented by orange lines. Right side (3D): the docked compounds are represented by gray stick models, and the active site pockets are shown by blue stick models. H-bond interactions are shown in green dashed lines. π-interactions are shown in orange lines. Image adapted from [122].
Figure 2. The molecular interactions of the best-docked compounds 10a and 10b with the target enzyme DNA gyrase B. Left side (2D): the residues are represented in 3 letter codes. Hydrogen bonds are represented by green and blue lines, and π-interactions are represented by orange lines. Right side (3D): the docked compounds are represented by gray stick models, and the active site pockets are shown by blue stick models. H-bond interactions are shown in green dashed lines. π-interactions are shown in orange lines. Image adapted from [122].
Antibiotics 12 01220 g002
Scheme 4. Synthesis of benzimidazole-1,2,3-triazole hybrids 13a13g.
Scheme 4. Synthesis of benzimidazole-1,2,3-triazole hybrids 13a13g.
Antibiotics 12 01220 sch004
Scheme 5. Synthesis of benzimidazole-1,2,3-triazole hybrids 19a19e, 20a20e, and 21a21e.
Scheme 5. Synthesis of benzimidazole-1,2,3-triazole hybrids 19a19e, 20a20e, and 21a21e.
Antibiotics 12 01220 sch005
Figure 3. Structure of benzimidazole-1,2,3-triazole hybrids 22a22b.
Figure 3. Structure of benzimidazole-1,2,3-triazole hybrids 22a22b.
Antibiotics 12 01220 g003
Scheme 6. Synthesis of benzimidazole-1,2,3-triazole hybrids 26a26j.
Scheme 6. Synthesis of benzimidazole-1,2,3-triazole hybrids 26a26j.
Antibiotics 12 01220 sch006
Scheme 7. Synthesis of benzimidazole-1,2,3-triazole hybrids 29a29l.
Scheme 7. Synthesis of benzimidazole-1,2,3-triazole hybrids 29a29l.
Antibiotics 12 01220 sch007
Figure 4. (a) Docking pose and (b) binding interactions of compound 29l in the cavity of Glms. Image adapted from [70].
Figure 4. (a) Docking pose and (b) binding interactions of compound 29l in the cavity of Glms. Image adapted from [70].
Antibiotics 12 01220 g004
Figure 5. Structure of benzimidazole-1,2,3-triazole hybrids 3032.
Figure 5. Structure of benzimidazole-1,2,3-triazole hybrids 3032.
Antibiotics 12 01220 g005
Scheme 8. Synthesis of benzimidazole-1,2,3-triazole hybrids 35a35g.
Scheme 8. Synthesis of benzimidazole-1,2,3-triazole hybrids 35a35g.
Antibiotics 12 01220 sch008
Scheme 9. Synthesis of benzimidazole-1,2,3-triazole hybrids 37a37d.
Scheme 9. Synthesis of benzimidazole-1,2,3-triazole hybrids 37a37d.
Antibiotics 12 01220 sch009
Scheme 10. Synthesis of benzimidazole-1,2,3-triazole hybrids 41a41e.
Scheme 10. Synthesis of benzimidazole-1,2,3-triazole hybrids 41a41e.
Antibiotics 12 01220 sch010
Scheme 11. Synthesis of benzimidazole-1,2,3-triazoles 43a43d.
Scheme 11. Synthesis of benzimidazole-1,2,3-triazoles 43a43d.
Antibiotics 12 01220 sch011
Scheme 12. Synthesis of benzimidazole-1,2,3-triazoles 45a45g.
Scheme 12. Synthesis of benzimidazole-1,2,3-triazoles 45a45g.
Antibiotics 12 01220 sch012
Scheme 13. Synthesis of benzimidazole-1,2,3-triazoles 47a47f.
Scheme 13. Synthesis of benzimidazole-1,2,3-triazoles 47a47f.
Antibiotics 12 01220 sch013
Figure 6. Structure of benzimidazole-1,2,3-triazole hybrids 48a48l.
Figure 6. Structure of benzimidazole-1,2,3-triazole hybrids 48a48l.
Antibiotics 12 01220 g006
Scheme 14. Synthesis of benzimidazole-1,2,3-triazoles 51a51d.
Scheme 14. Synthesis of benzimidazole-1,2,3-triazoles 51a51d.
Antibiotics 12 01220 sch014
Scheme 15. Synthesis of benzimidazole-1,2,3-triazoles 54a54n.
Scheme 15. Synthesis of benzimidazole-1,2,3-triazoles 54a54n.
Antibiotics 12 01220 sch015
Figure 7. (a). Superimposition of compounds 54a (Cyan color), 54d (Magenta color) with ligand (Green–blue color). (b). All 14 compounds docked into the active site of the enzyme 4FDO. Image adapted from [137].
Figure 7. (a). Superimposition of compounds 54a (Cyan color), 54d (Magenta color) with ligand (Green–blue color). (b). All 14 compounds docked into the active site of the enzyme 4FDO. Image adapted from [137].
Antibiotics 12 01220 g007
Figure 8. (a). Structure of benzimidazolone bis-1,2,3-triazoles 55a55n. (b). Representation of docked view of compound 55j at the active site of RmlC.
Figure 8. (a). Structure of benzimidazolone bis-1,2,3-triazoles 55a55n. (b). Representation of docked view of compound 55j at the active site of RmlC.
Antibiotics 12 01220 g008
Scheme 16. Synthesis of benzimidazole-1,2,4-triazoles 59a59e.
Scheme 16. Synthesis of benzimidazole-1,2,4-triazoles 59a59e.
Antibiotics 12 01220 sch016
Scheme 17. Synthesis of benzimidazole-1,2,4-triazoles 63a63e.
Scheme 17. Synthesis of benzimidazole-1,2,4-triazoles 63a63e.
Antibiotics 12 01220 sch017
Scheme 18. Synthesis of benzimidazole-1,2,4-triazoles 68 and 69.
Scheme 18. Synthesis of benzimidazole-1,2,4-triazoles 68 and 69.
Antibiotics 12 01220 sch018
Figure 9. Structure of benzimidazole hybrids 64, 65, and 70.
Figure 9. Structure of benzimidazole hybrids 64, 65, and 70.
Antibiotics 12 01220 g009
Scheme 19. Synthesis of benzimidazole-1,2,4-triazoles 73a73l.
Scheme 19. Synthesis of benzimidazole-1,2,4-triazoles 73a73l.
Antibiotics 12 01220 sch019
Figure 10. Binding mode of compound 73e in the active site of modeled CYP51 of C. albicans. Image adapted from [145].
Figure 10. Binding mode of compound 73e in the active site of modeled CYP51 of C. albicans. Image adapted from [145].
Antibiotics 12 01220 g010
Figure 11. Structure of benzimidazole-1,2,4-triazole hybrids 74a74c.
Figure 11. Structure of benzimidazole-1,2,4-triazole hybrids 74a74c.
Antibiotics 12 01220 g011
Scheme 20. Synthesis of benzimidazole-1,2,4-triazoles 79a79c.
Scheme 20. Synthesis of benzimidazole-1,2,4-triazoles 79a79c.
Antibiotics 12 01220 sch020
Scheme 21. Synthesis of benzimidazole-1,2,4-triazoles 85a85e and compound 86c.
Scheme 21. Synthesis of benzimidazole-1,2,4-triazoles 85a85e and compound 86c.
Antibiotics 12 01220 sch021
Figure 12. Ligand interaction diagram of compound 74c in lanosterol 14α-demethylase (a) 2D and (b) 3D view. Image adapted from [147].
Figure 12. Ligand interaction diagram of compound 74c in lanosterol 14α-demethylase (a) 2D and (b) 3D view. Image adapted from [147].
Antibiotics 12 01220 g012
Figure 13. Two-dimensional (A) and three-dimensional (B) diagrams of compound (S)-85b (tautomer A) in the active site of CYP51–C. albicans (5TZ1.pdb). Image adapted from [72].
Figure 13. Two-dimensional (A) and three-dimensional (B) diagrams of compound (S)-85b (tautomer A) in the active site of CYP51–C. albicans (5TZ1.pdb). Image adapted from [72].
Antibiotics 12 01220 g013
Figure 14. 3D binding picture and 2D schematic protein-ligand interactions of compound 79a. Image adapted from [148].
Figure 14. 3D binding picture and 2D schematic protein-ligand interactions of compound 79a. Image adapted from [148].
Antibiotics 12 01220 g014
Scheme 22. Synthesis of benzimidazole-1,2,4-triazoles 88a88c.
Scheme 22. Synthesis of benzimidazole-1,2,4-triazoles 88a88c.
Antibiotics 12 01220 sch022
Scheme 23. Synthesis of benzimidazole-1,2,4-triazoles 89a89c.
Scheme 23. Synthesis of benzimidazole-1,2,4-triazoles 89a89c.
Antibiotics 12 01220 sch023
Scheme 24. Synthesis of benzimidazole-1,2,4-triazoles 9597.
Scheme 24. Synthesis of benzimidazole-1,2,4-triazoles 9597.
Antibiotics 12 01220 sch024
Scheme 25. Synthesis of bis-benzimidazole-1,2,4-triazoles 102a102e.
Scheme 25. Synthesis of bis-benzimidazole-1,2,4-triazoles 102a102e.
Antibiotics 12 01220 sch025
Figure 15. Structure of benzimidazole-1,2,4-triazole hybrids 90 and 91.
Figure 15. Structure of benzimidazole-1,2,4-triazole hybrids 90 and 91.
Antibiotics 12 01220 g015
Figure 16. Structure of benzimidazole-1,2,4-triazole hybrids 98100.
Figure 16. Structure of benzimidazole-1,2,4-triazole hybrids 98100.
Antibiotics 12 01220 g016
Scheme 26. Synthesis of benzimidazole-1,2,4-triazoles 105a105b.
Scheme 26. Synthesis of benzimidazole-1,2,4-triazoles 105a105b.
Antibiotics 12 01220 sch026
Scheme 27. Synthesis of benzimidazole-1,2,4-triazoles 107a107b.
Scheme 27. Synthesis of benzimidazole-1,2,4-triazoles 107a107b.
Antibiotics 12 01220 sch027
Scheme 28. Synthesis of benzimidazole-1,2,4-triazoles 109a109h.
Scheme 28. Synthesis of benzimidazole-1,2,4-triazoles 109a109h.
Antibiotics 12 01220 sch028
Scheme 29. Synthesis of benzimidazole-1,2,4-triazoles 111a111s.
Scheme 29. Synthesis of benzimidazole-1,2,4-triazoles 111a111s.
Antibiotics 12 01220 sch029
Figure 17. Structure of benzimidazole-1,2,4-triazole hybrids 112a112i.
Figure 17. Structure of benzimidazole-1,2,4-triazole hybrids 112a112i.
Antibiotics 12 01220 g017
Figure 18. SAR outline of the benzimidazole-1,2,4-triazole hybrids 112a112i.
Figure 18. SAR outline of the benzimidazole-1,2,4-triazole hybrids 112a112i.
Antibiotics 12 01220 g018
Figure 19. Structure of benzimidazole-1,2,4-triazole hybrids 113a113l.
Figure 19. Structure of benzimidazole-1,2,4-triazole hybrids 113a113l.
Antibiotics 12 01220 g019
Figure 20. Structure of benzimidazole-1,2,4-triazole hybrids 114a114l.
Figure 20. Structure of benzimidazole-1,2,4-triazole hybrids 114a114l.
Antibiotics 12 01220 g020
Scheme 30. Synthesis of benzimidazole-triazoles 115a115d.
Scheme 30. Synthesis of benzimidazole-triazoles 115a115d.
Antibiotics 12 01220 sch030
Figure 21. Structure of antiviral benzimidazole-1,2,3-triazole hybrids 116 and 117.
Figure 21. Structure of antiviral benzimidazole-1,2,3-triazole hybrids 116 and 117.
Antibiotics 12 01220 g021
Figure 22. Structure of antiviral benzimidazole-1,2,3-triazole hybrids 118137.
Figure 22. Structure of antiviral benzimidazole-1,2,3-triazole hybrids 118137.
Antibiotics 12 01220 g022
Figure 23. Structure of antiviral benzimidazole-1,2,3-triazole hybrids 138140.
Figure 23. Structure of antiviral benzimidazole-1,2,3-triazole hybrids 138140.
Antibiotics 12 01220 g023
Figure 24. Three-dimensional binding mode of compound 140 (green) at the binding interface between the Omicron S-RBD (red) and human ACE2 (blue) [74].
Figure 24. Three-dimensional binding mode of compound 140 (green) at the binding interface between the Omicron S-RBD (red) and human ACE2 (blue) [74].
Antibiotics 12 01220 g024
Figure 25. Structure of antiviral benzimidazole-1,2,3-triazole hybrid 141.
Figure 25. Structure of antiviral benzimidazole-1,2,3-triazole hybrid 141.
Antibiotics 12 01220 g025
Table 1. Antimicrobial screening results of compounds 6a6f presented as MIC (μg mL−1).
Table 1. Antimicrobial screening results of compounds 6a6f presented as MIC (μg mL−1).
CompoundGram-Positive OrganismsGram-Negative OrganismsFungi Organisms
B.c.S.a.P.a.E.c.A.b.C.a.
6a6464256128128128
6b128128128128256256
6c25612825664256156
6d25612825664256256
6e25612825664256256
6f512512256256512512
Ciprofloxacin8484--
Table 2. In vitro antimicrobial screening of hybrids 8, 9, and 10 using the agar diffusion method.
Table 2. In vitro antimicrobial screening of hybrids 8, 9, and 10 using the agar diffusion method.
HybridsInhibition Zone Diameters Using the Agar Diffusion Method (mm)
S. aureusE. coliP. aeruginosaA. nigerC. albicans
8a15 ± 0.1412 ± 1.0822 ± 1.01--
8b-5 ± 0.2-30 ± 1.16 27 ± 1.1
9a23 ± 0.8-13 ± 0.65--
9b--12 ± 0.814 ± 0.1519 ± 1.04
10a24 ± 0.625 ± 0.917 ± 0.7520 ± 0.916 ± 0.89
10b29 ± 1.221 ± 1.1419 ± 0.7918 ± 0.1214 ± 0.58
Ciprofloxacin20 ± 0.923 ± 1.0221 ± 0.9--
Nystatin---22 ± 0.1823 ± 1.15
Table 3. Antimicrobial activity of the compounds 35 in terms of MIC (µmol mL−1).
Table 3. Antimicrobial activity of the compounds 35 in terms of MIC (µmol mL−1).
CompoundS. aureusE. coliB. subtilisS. epidermitisA. nigerC. albicans
35a0.0280.0560.0560.0560.0560.056
35b0.0310.0620.0620.0620.0620.062
35c0.0290.0580.0580.0580.0580.058
35d0.0600.0300.0600.0300.0600.060
35e0.0290.0560.0560.0560.0560.056
35f0.0260.0520.0520.0520.0520.052
35g0.0310.0260.0520.0260.0260.026
Norfloxacin0.0200.0390.0390.039--
Fluconazole----0.040.020
Table 4. Antimicrobial activity of the compounds 63a63o using the agar diffusion method.
Table 4. Antimicrobial activity of the compounds 63a63o using the agar diffusion method.
CompoundInhibition Zone Diameters Using the Agar Diffusion Method (mm)
S. aureusP. aeruginosaE. coliS. typhosa
63a28262119
63b23181614
63c21231819
63d20222323
63e25232124
63f27262420
63g19201513
63h29262224
63i26221918
63j14121616
63k22212018
63l25231921
63m21181816
63n24222221
63o19211814
Gentamycin34353130
Table 5. ED50 values (µg mL−1) of compounds against test fungi.
Table 5. ED50 values (µg mL−1) of compounds against test fungi.
CompoundF. verticillioidesD. oryzaeC. lunataF. fujikuroi
74a35502845
74b30251830
74c16121015
Carbendazim230--150
Propiconazole20252221
Table 6. Antimicrobial activity of compounds 88a88c expressed as MIC in μg mL−1.
Table 6. Antimicrobial activity of compounds 88a88c expressed as MIC in μg mL−1.
CompoundS. aureusB. subtilisS. mutansP. aeruginosaC. albicans
88aNTNT161632
88b8161616NT
88c816323232
Ampicillin22<14NT
Kanamycin2<142NT
NT = not tested.
Table 7. The minimum inhibitory concentrations (µg mL−1) of the compounds against fungi.
Table 7. The minimum inhibitory concentrations (µg mL−1) of the compounds against fungi.
CompoundConcentration (µg mL−1)Aspergillus nigerFusarium oxysporum
89a5050-
89b505050
89c5050-
89d5050-
Table 8. Antimicrobial activity of compounds 89a89c.
Table 8. Antimicrobial activity of compounds 89a89c.
Compound (800 µg mL−1)S. aureusP. aerugnosaB. subtilisA. baumanniiC. albicans
95181415-10
96191112-11
9717151412-
Amoxicillin333233--
Fluconazole----25
Table 9. Antimicrobial activity of compounds 105a105b and 107a107b.
Table 9. Antimicrobial activity of compounds 105a105b and 107a107b.
CompoundMinimum Inhibitory Concentrations (μg mL−1)
Gram-Positive BacteriaGram-Negative Bacteria
B. subtilisS. aureusE. coliP. aeruginosa
105a98-52-
105b--65-
107a7510562-
107b79-72-
Gentamycin *64567248
* Concentration of Gentamycin = 30 μg mL−1.
Table 10. Antibacterial activity of compounds 109a109h.
Table 10. Antibacterial activity of compounds 109a109h.
NoCompoundZone of Inhibition (mm)
E. coliS. aureusC. albicans
1109a151318
2109b131112
3109c171614
4109d121316
5109e13179
6109f10811
7109g81112
8109h12710
9Ampicilline2425-
10Ketokonazole--20
Table 11. MIC50 (μg mL−1) values of compounds 111a111s.
Table 11. MIC50 (μg mL−1) values of compounds 111a111s.
CompoundC. albicansG. glabrataC. kruseiC. parapsilosis
111a12.56.256.2512.5
111b6.253.126.256.25
111c12.56.256.2512.5
111d6.2512.56.256.25
111e12.56.2512.512.5
111f6.253.123.126.25
111g3.126.256.256.25
111h12.56.2512.56.25
111i0.781.561.560.78
111j12.56.2512.512.5
111k12.56.2512.512.5
111l6.2512.56.2512.5
111m3.123.123.126.25
111n3.123.121.563.12
111o3.123.126.256.25
111p12.512.526.256.25
111r6.253.123.123.12
111s0.781.561.560.78
Ketokonazole0.781.561.561.56
Fluconazole0.781.561.560.78
Table 12. Anti-JEV and anti-HSV activity of compounds 59a59e.
Table 12. Anti-JEV and anti-HSV activity of compounds 59a59e.
Compd.In Vitro In Vivo
CT50
(µg mL–1)
EC50
(µg mL–1)
TICPE
Inhibition (%)
Dose (µg per Mouse per Day)MST
(days)
Protection
(%)
Anti-JEV
59a12543130200--
59b12581690200416
59c-------
59d12543130200--
59e25062.5450200210
Anti-HSV
59a12562.5233---
59b12562.5246---
59c-------
59d12531.25453200--
59e2507.83264200--
CT50—50% cytotoxic concentration, EC50—50% effective concentration, TI—therapeutic index (TI = CT50/EC50). CPE—cytopathic effect, MST—mean survival time.
Table 13. RSV, BVDV, YFV, and CVB2 Inhibitory Activity of hybrids 118137 expressed as EC50 (µM).
Table 13. RSV, BVDV, YFV, and CVB2 Inhibitory Activity of hybrids 118137 expressed as EC50 (µM).
CompoundAnti-RSV ActivityAnti-BVDV ActivityAnti-YFV ActivityAnti-CVB2 Activity
1180.7---
1192.3---
1200.7>10080>100
1210.763>90>100
1220.353>70>100
1230.1551>60>100
1240.03---
1250.7---
1260.0690>100>100
1270.172>54>100
1280.915640
1290.0519>21>88
1300.0214>2026
13110.0---
1327.0---
1331.967>36>100
134>3615>18>36
1359---
1361180>45>100
13723.08027>83
6-Azaurine1.2>10026>100
Table 14. Antiviral activity of benzimidazole-1,2,3-triazole hybrids 138140.
Table 14. Antiviral activity of benzimidazole-1,2,3-triazole hybrids 138140.
CompoundCC50 (µg mL−1)EC50 (µg mL−1)Selectivity Index (SI)
Ceftazidime1045.5385.0712.29
1381065.51155.056.87
1391530.5306.15.0
1401028.2880.412.78
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

Marinescu, M. Benzimidazole-Triazole Hybrids as Antimicrobial and Antiviral Agents: A Systematic Review. Antibiotics 2023, 12, 1220. https://doi.org/10.3390/antibiotics12071220

AMA Style

Marinescu M. Benzimidazole-Triazole Hybrids as Antimicrobial and Antiviral Agents: A Systematic Review. Antibiotics. 2023; 12(7):1220. https://doi.org/10.3390/antibiotics12071220

Chicago/Turabian Style

Marinescu, Maria. 2023. "Benzimidazole-Triazole Hybrids as Antimicrobial and Antiviral Agents: A Systematic Review" Antibiotics 12, no. 7: 1220. https://doi.org/10.3390/antibiotics12071220

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