Department of Pharmacy, KBHSS trust’s Institute of Pharmacy, Malegaon -423105, Dist Nashik, Maharashtra, India
The relentless emergence of drug-resistant microbial strains has intensified the global search for novel therapeutic agents with enhanced efficacy and safety profiles. In this context, the benzimidazole and triazole scaffolds have garnered significant attention due to their broad-spectrum biological activities, particularly their antimicrobial and anticancer properties. This study explores the potential of newly designed benzimidazole and triazole derivatives through in silico molecular docking methodologies to identify promising leads for drug development. The study focused on computational docking of designed compounds with two key biological targets: DNA gyrase (PDB ID: 6RKS) and human topoisomerase II (PDB ID: 1ZXM), enzymes crucial to DNA replication and cell proliferation, and known to be validated targets in antimicrobial and anticancer drug discovery. Molecular docking was performed using Autodock 4.2, and the docking results were evaluated based on binding affinity, hydrogen bond interactions, and overall binding pose stability within the active site of the target proteins. The benzimidazole and triazole derivatives demonstrated significant binding affinity toward both target proteins. Among the designed compounds, several ligands exhibited binding energies surpassing those of standard reference drugs such as ciprofloxacin and etoposide. In particular, compounds with electron-withdrawing substituents and heterocyclic moieties at strategic positions of the benzimidazole and triazole rings showed improved interaction profiles. These favorable interactions included multiple hydrogen bonds and hydrophobic contacts, indicating strong and specific binding within the enzymatic active sites.Furthermore, the pharmacokinetic properties and drug-likeness of the ligands were evaluated using Lipinski’s Rule of Five and ADMET predictions, confirming the acceptable oral bioavailability and low toxicity of the top-scoring compounds. The in-silico findings suggest that the synthesized molecules have the potential to act as dual inhibitors of microbial and human topoisomerases, presenting a unique avenue for the development of multifunctional therapeutic agents. This study highlights the utility of molecular docking as a powerful predictive tool in the early phases of drug discovery. By integrating structural chemistry with computational biology, it is possible to efficiently screen and optimize novel scaffolds prior to in vitro and in vivo evaluations. The promising docking results of benzimidazole and triazole derivatives underscore their potential as future leads for the development of antimicrobial and anticancer drugs, warranting further experimental validation.
Benzimidazole structures have drawn a lot of interest in the last 10 years because of their growing significance in the creation of novel medicinal molecules. Because of its antibacterial qualities, researchers have concentrated on developing and researching novel benzimidazole derivatives for possible medical uses. Finding novel structures that could result in safer and more effective antimicrobial therapies is becoming more and more important as antibiotic resistance increases. A new structural prototype has been created as part of this endeavor. Benzimidazoles are useful building blocks for more molecular research because they are among the most potent substances against bacteria. These substances have a variety of biological properties, such as antiviral, antiproliferative, antiparasitic, antibacterial, anthelmintic, anti-inflammatory, antispasmodic, anticancer, antihypertensive, and antioxidant properties. Efforts have been directed on creating a library of compounds generated from benzimidazoles and assessing their possible biological activities because of the significance and variety of biological effects linked to these medications. Furthermore, it is well known that the triazole ring is linked to a number of pharmacological actions, such as strong analgesic, antibacterial, and anti-inflammatory properties. Consequently, the antibacterial properties of triazole and benzimidazole components have been combined to create new benzimidazole derivatives with a triazole core. The possible biological roles of these substances are presently being studied. The synthesis of a number of new benzimidazole triazole compounds and their possible uses are highlighted in this work. [1] Because they can be used as therapeutic agents to treat a variety of illnesses, heterocyclic compounds are important in medical chemistry. One such substance with a variety of medical uses is benzimidazole, a purine analogue of the pharmacological group. As an antibacterial, antiviral, antihistamine, anticonvulsant, anticancer, proton pump inhibitor, antiparasitic, anti-inflammatory, and antihypertensive medication, it works well for a number of illnesses. While some benzimidazoles, such astemizole, have antiprion activity and can be used to treat Creutzfeldt-Jakob disease, others can be used to cure diabetes. Furthermore, benzimidazole compounds have been shown to have anticoagulant, analgesic, psychotropic, anxiolytic, and anti-Alzheimer's properties. Triazole chemicals are also well-known for a variety of biological functions. These include antibacterial, antituberculosis, antiviral, anti-inflammatory, anticancer, antihypertensive, antioxidant, and antiepileptic properties, as well as possible inhibitory effects on SARS-CoV-2. Triazoles also function as α-glucosidase inhibitors, analgesics, anticonvulsants, and antimalarials. Furthermore, triazole compounds may be helpful in treating Alzheimer's disease because they have been demonstrated to have neuroprotective effects. [2] Molecular docking is a drug design technique that simulates the molecular interactions and binding affinities of receptors to predict how ligands will interact with them. The use of this approach in drug development research is growing in popularity. By filtering possible pharmacological structures from compound databases, this technique not only makes it easier to obtain, synthesize, and run pharmacological tests, but it also significantly boosts productivity and lowers research expenses. Furthermore, a deeper understanding of the molecular mechanisms underlying drug development and the ability to forecast therapeutic targets have been made possible by the development of reverse molecular docking technology. An overview of current developments and uses of molecular docking technology is given in this work. [3] The temperature-sensitive filament protein Z (FtsZ) is essential for the division of bacterial cells. FtsZ hydrolyzes GTP to GDP during this process, which makes it a desirable target for the development of antituberculosis medications. In this work, we used a quantitative structure-activity relationship (QSAR) technique to construct a predictive model for FtsZ protein inhibition. In pharmaceutical chemistry, QSAR is a crucial tool for drug development. A QSAR model was created to target the antituberculosis properties of 50 trisubstituted benzimidazole compounds that have been shown to inhibit FtsZ. This approach makes it easier to create a mathematical tool that forecasts new trisubstituted benzimidazole compounds' antituberculosis activity. The antituberculosis characteristics of a number of benzimidazole compounds were then predicted using the QSAR model. Each suggested compound's physicochemical and pharmacological characteristics were assessed using ADMET analysis, molecular docking, pharmacogenetic modeling, and molecular dynamics simulations. [4] Finding a lead chemical is usually the initial stage in the drug development process. This is followed by a number of investigations and the production of several structural variations of the molecule. Strict rules and procedures are followed in 3D QSAR investigations to produce accurate and trustworthy models, enabling the synthesis of several compounds at once and the assessment of their effects on therapeutic targets. Several 3D-QSAR techniques, such as comparative molecular field analysis (CoMFA) and comparative molecular similarity index analysis (CoMSIA), are commonly employed in these investigations. This study's goal is to use 3D-QSAR techniques like CoMFA and CoMSIA to model and forecast the anticancer characteristics of several benzimidazole derivatives. To assess the stability and binding interactions of these drugs with Pin1, a crucial receptor implicated in breast cancer, molecular docking and molecular dynamics (MD) simulations were conducted in the second stage. Furthermore, these effects were contrasted with those of commonly used breast cancer inhibitors like tamoxifen and trastuzumab. Furthermore, ADMET analysis was carried out to look into the compounds' pharmacological characteristics. These findings offer important insights for the creation of new benzimidazole compounds that may have anticancer properties. [5]
Molecular Docking Studies:
By examining the complementarity and spatial arrangement of ligands and receptors, molecular docking is a technique that predicts ideal molecular conformations in order to assess binding affinity and interaction mechanisms. The "lock and key model" depicted in Figure 1A emphasizes the significance of geometric complementarity by carefully docking the ligand and receptor to ascertain the proper orientation for the ligand ("key") to fit into the receptor ("lock"). Because both the ligand and the receptor must modify their conformations to attain an ideal match, the docking process is incredibly adaptable. This flexibility is taken into consideration by the "induced fit model" (Figure 1B), which emphasizes energetic complementarity and previous organization in addition to geometric complementarity. By reducing free energy, this procedure guarantees that the ligand and receptor attain the most stable configuration. Molecular docking software employs specific algorithms to identify the ideal conformation and orientation depending on complementarity and initial configuration, as illustrated in Figure 2. After that, the program analyzes the interaction mode and forecasts binding affinity using a score system. A protein-DNA docking using AutoDock Vina in PyMOL is displayed in Figure 3. [6] The idea of molecular complementarity, which states that two structures complement one another like a glove, is the foundation of molecular docking. When assessing the binding and interaction between two molecular structures, this type of complementarity is crucial. In the binding and interaction processes, physical and chemical complementarity are essential. Since the early 1980s, molecular docking has been a crucial method in drug discovery. More complex computer methods for drug discovery, such as virtual structure-based high-throughput screening (vHTS), have been made possible by developments in structural biology. This aids in lead drug optimization and hit compound identification. Yet, ligand flexibility, entropic effects, solvation/desolvation, the presence of ions and water molecules, tautomerism, protein flexibility, binding selectivity, pharmacodynamic effects, isomerism effects, and the overall molecular context are some of the drawbacks of the current docking methods. The ranking and scoring of docked ligands as well as the computation of post-docking molecular interactions—such as the ligand's orientation and location within the protein binding site—are the main topics of this review part. Protein-protein interactions that are significant in biochemistry due to their functional implications (such as signal transduction) can also be studied using these techniques. Numerous molecular interactions, such as solvent-related forces (such as hydrogen bonding and hydrophobic interactions brought on by structural changes in the solvent), steric forces (entropy), electrostatic forces (charge), and electrodynamic forces (anti-interactions—Der Waals forces), can be identified and predicted using molecular docking techniques. Complex molecular structures are formed in part by these interactions. [7]
Bezimidazole Derivative:
AutoDock 4.2 with MGL Tools 1.5.6 was utilized for all docking simulations. The Protein Data Bank (RCSB) (http://www.rcsb.org/pdb) provided DNA gyrase subunit B (PDB ID: 1KZN) and topoisomerase II (PDB ID: 1JIJ), which were then docked to a number of new triazinane chemicals, including derivatives of benzimidazoles. The DNA gyrase B component and topoisomerase II crystal structures were used for the analysis. Marvin Sketch (Chem Axon) was used to create the 2D structures of the synthesized ligands (5(aâe) and 6(aâe)), which were subsequently transformed into energy-minimized 3D models in PDB format for computational investigations. Using AutoDock 4.2, target protein files were created by eliminating heteroatoms, cofactors, and water molecules, leaving only the pertinent protein residues. The file was converted to AD4 format once all of the protein molecule's atoms had been given Gasteiger charges. For compounds 5 (aâe) and 6 (aâe), docking modeling was used to target the DNA gyrase b subunit and the topoisomerase II active site. Maestro Elements Tutorial 1.8 was used to illustrate the docking results, and Table 2 provides a summary. [6] We examined the genomes of common soil-borne nematodes (STH) and all Ascaris species whose genomes were available in Wormbase-Parasite in order to find β-tubulin isoforms. BLAST searches were conducted using the peptide sequence of A. lumbricoides isoform A as a reference. Additionally, as many relevant genes as possible were found using the search keyword "tubulin beta." The data were sorted by tubulin type and gene size in order to remove tubulin and tiny fragment sequences. Additionally, publically accessible Ascaris β-tubulin sequences from NCBI were utilized in the analysis. In this investigation, the following nematode species were examined: Using BLAST searches, we were able to identify β-tubulin sequences even though the Ascaridia galli genome was missing. Necator americanus, Parascaris equorum, Parascaris univalens, Trichuris trichiura, Trichuris suis, Anisakis simplex, Toxocara canis, Ancylostoma caninum, Ancylostoma ceylanicum, and Ancylostoma duodenale were among the species that were analyzed. Supplementary Table 1 contains a comprehensive list of all known β-tubulin genes. Software called BioEdit (version 7.0.5.3) was used to align the sequences. [8]
Triazole derivative
The binding interactions that give organic molecules their biological activity are better understood thanks to in silico research. Alpha-lanosterol demethylase from *Candida albicans* and DNA gyrase from Escherichia coli have both been demonstrated to be inhibited by azole-based medications. The addition of triazole and chalcone units to indole may improve the antibacterial qualities of the resultant hybrid compounds, according to in vitro activity data. In order to investigate this matter further, compounds 2, 4b, and 6b were docked into the E. coli DNA gyrase active site, while compound 6g, along with compounds 4b and 2, was docked into the M. albicans alpha-lanosterol demethylase binding site. Alkyne 2 demonstrated restricted hydrophobic interactions with DNA gyrase binding site residues ASN46, ASP73, ILE78, PRO79, and VAL120 at a binding energy of -6.4 kcal/mol. Nevertheless, the insertion of the triazole core increased the alkyne's binding affinity by facilitating additional interactions with the binding site residues. In compound 4b, the carbonyl oxygen bonded to ASN46, whereas the triazole pi cloud linked with GLU50 by a pi anion connection. Furthermore, the indole's α-electron formed a sigma-α contact with THR165, and the terminal phenyl ring connected with ILE78. Compound -7.7 kcal/mol has a higher binding affinity because of its improved hydrophobic interactions with VAL43, ALA47, ILE78, PRO79, ILE90, and VAL167. The binding affinity was further increased by adding chalcone activity; compound 6b achieved a binding energy of -8.7 kcal/mol. The bromophenyl ring established a carbon-hydrogen link with the triazole carbon atom and a methylene bridge, while GLU50 and ASP49 established sigma-γ interactions with ALA53. In order to stabilize the molecule inside the active site, hydrophobic interactions were essential. While Figure 3 depicts their interactions, Table 2 and the Supplementary Information provide the precise binding properties of CBN ligands co-crystallized with compounds 2, 4b, and 6b in *E. coli* DNA gyrase. [9] The antifungal characteristics of a number of newly created 1,2,4-triazole-based chromenols were examined by Zvyagintseva et al. (Figure 6). They chose 14 compounds for synthetic and biological investigation after predicting the antifungal activity (Pa) of the suggested analogues using PASS. These compounds were anticipated to have Pa values between 0.43 and 0.53. In the antifungal testing, reference medications ketoconazole and benazole were utilized. With a minimum inhibitory concentration (MIC) range of 22.1–184.2 µM, compound 4d had the highest activity among the drugs tested, while ketoconazole's MIC range was 480–640 µM. When compared to bifonazole and ketoconazole, all of the evaluated compounds exhibited greater antifungal activity. DNA topoisomerase IV and lanosterol 14α-demethylase in Candida albicans were used in docking studies. The results indicated that the docked drugs had greater binding free energies than CYP15, indicating that inhibition of this enzyme may be a possible mechanism of action. In silico study verified the compounds' drug-like qualities, and none of them deviated from Lipinski's standards. It was anticipated that every component would pass across the blood-brain barrier, with the exception of compounds 4c and 4e. Tests of cytotoxicity against the non-cancerous cell line HK-2 and the breast cancer cell line MCF7 revealed enhanced cell viability at 100 μM and little harm at 50 μM. Compounds 4a, 4b, 4d, 4f, and 4g were determined to be interesting candidates for additional therapeutic research in light of these findings. [10]
Toxicity Prediction:
Combining computational techniques with in vitro and in vivo toxicity testing can improve safety assessment and toxicity prediction accuracy, decrease the need for animal testing, and save time and money. The benefit of these techniques is that they can forecast a chemical's toxicity prior to its synthesis. (A) databases with information about chemicals, their characteristics, and their toxicities; (B) software for creating molecular descriptors; (C) modeling tools for systems biology and molecular dynamics; (D) methods for predicting toxicity; (E) statistical software for creating predictive models; (F) expert systems with pre-built toxicity prediction models that can be used as standalone applications or web servers; and (G) visualization tools are some examples of the computational tools commonly used in in silico toxicology (Figure 1). This study focuses on expert systems that use predictive models (Element F) and computational tools that apply these methods (Element E), while also offering a comprehensive overview of cutting-edge modeling techniques and toxicity prediction algorithms. All seven components cannot be included in this study due to the field's rapid growth. For comprehensive information on toxicity databases, statistical modeling software, expert systems, toxicological modeling tools, molecular descriptor generators, and visualization tools, users are urged to refer to the most recent academic literature. Five main steps are usually included in creating a predictive model (Figure 1): gathering biological information that connects a chemical to a toxicity result, (2) figuring out the chemical's molecular descriptors, (3) creating a predictive model, (4) evaluating the model's accuracy, and (5) analyzing the model's output. [11] Depending on whether they concentrate on more general toxicological phenomena like carcinogenicity or on particular elements that result in toxic effects, in silico toxicology models can be categorized into two classes. While some models are made to look at systemic toxicity, others are made to evaluate toxicity specific to particular organ. Despite their complexity, genotoxicity and carcinogenicity are among the most extensively researched toxicity types, and there are numerous in silico models available. Since the existence of a model by itself does not ensure precise predictions, the applicability of a model for a given medication may rely on the availability of toxicological data for chemically related drugs. Although there are fewer models for developmental or reproductive toxicity, numerous studies have assessed the carcinogenicity and mutagenicity of common substances or medications. In the pharmaceutical business, in silico models that forecast organ-specific toxicity are growing in popularity as data for drug-like molecules becomes more accessible. Numerous studies have been conducted on hepatotoxicity, and models that forecast cardiotoxicity and nephrotoxicity are being employed more frequently. Models for additional consequences, such phospholipidosis or neurotoxicity, are less prevalent. To evaluate toxicity for several receptor targets, some studies have pooled adverse effect databases or combined risk scores. A growing number of in silico techniques are being used in regulatory applications, such as EU chemical and cosmetics laws. These techniques are mostly employed to find possible side effects of novel medications during the R&D stage of drug development. In order to identify precise structural warning signals for risk management, in silico-based genotoxicity assessment predictions are increasingly becoming a crucial component of the regulatory process, particularly for medication impurities. To evaluate the potential for genotoxicity or carcinogenicity, new models have recently been created. By fusing in silico data with professional analysis, these models have produced remarkable results. They have proven especially effective in evaluating genotoxic pollutants, with negative predictive values as high as 99%. [12] The process of creating new medications is costly and intricate. The primary causes of drug candidate failure are toxicity and ineffectiveness; toxicity issues can be responsible for as much as 40% of development failures. As a result, when developing new drugs, ADME/Tox (Absorption, Distribution, Metabolism, Excretion, Toxicity) must be taken into account. When evaluating the safety profile of possible medications and choosing molecules with the best chance of effectiveness, ADMET data is crucial. By enabling more precise choices about therapeutic dosages, this method speeds up drug discovery and drastically cuts down on time and expense. Drug development relies heavily on quantifiable chemical and physical features, which are frequently assessed using computational techniques. For screening chemical libraries, the "Lipinski Rule of Five" is a useful resource. By eliminating molecules that differ considerably from existing oral bioavailable medications, it helps select prospective drug candidates and improves the allocation of resources in drug research. Oral medications should have a molecular weight of fewer than 500 daltons, with hydrogen bond donors and acceptors not exceeding five and ten, respectively, according to Lipinski. Moreover, log P (partition coefficient) values smaller than 5 are desirable, and only one of these requirements may be broken. The SwissADME software's computational ADME predictions were used to assess the synthesized compounds' physicochemical characteristics and drug affinity. Lipinski's five requirements were met by every chemical utilized in the study, indicating that they had advantageous drug-like qualities. The predicted absorption percentages for these chemicals varied from 66.81% to 82.58% using the formula %ABS = 109 â 0.345 x TPSA. Lipophilicity, or the molecule's preference for a lipid environment over an aqueous one, is a prerequisite for effective medication distribution. A medicine needs to be soluble in both lipid and aqueous media in order to pass through biological barriers such the skin, gastrointestinal tract, and blood-brain barrier. With XLOGP3 values ranging from 2.34 to 3.78, the synthesized compounds demonstrated moderate to high lipophilicity, indicating the possibility of their passage over biological barriers that are rich in lipids. These compounds also demonstrated good solubility, with values ranging from -3.89 to -4.80. This means that they are highly soluble in aqueous conditions, which is crucial for effective transport and distribution throughout the body. Cytochrome P450 (CYP) enzymes, which are mostly present in the liver but can also be found in other tissues including the intestine, are necessary for the metabolism of many medications. These enzymes change how medications break down, activate, or inactivate by catalyzing the body's oxidation processes. Assessing drug metabolism requires an understanding of how particular CYP enzymes are inhibited or induced. CYP2D6, CYP2C9, CYP3A4, CYP1A2, and CYP2C19 are the five main cytochrome P450 enzymes that handle about 90% of drug metabolism in humans. It was discovered that compounds 8d, 8e, 8g, and 8h inhibit CYP1A2, perhaps lowering its activity. Every tested substance was shown to inhibit CYP2C19 and CYP2C9, indicating that they might also lessen these enzymes' activity. The fact that none of the substances inhibited CYP2D6 suggests that they had no effect on CYP2D6 activity. Compounds 8a, 8b, 8c, 8e, 8f, 8g, and 8h were also shown to be CYP3A4 inhibitors, indicating that they could be able to lower this enzyme's activity. [13] Many medication candidates fail due to toxicity, which also raises the expense of drug research considerably. When toxicity is found later in clinical trials or after the drug has been approved, this issue becomes even more severe. Mechanism-based (target) toxicity, immunological hypersensitivity, non-target toxicity, biological activation/covalent modification, and, less frequently, bioaccumulation are some of the causes of toxicity. Rome investigated ways to discover processes and structural clues that can suggest negative consequences in order to employ in silico methodologies to forecast toxicity. The toxicity of RS compounds can also be investigated using this methodology. Because chemoinformatics can be used to study pathways and find unforeseen adverse side effects, Valerio argues that it may be more effective than in-silico approaches. When used early in the drug discovery process, this method can aid in the development of safer medication candidates by improving our understanding of the underlying detrimental mechanisms. [14] Compounds having high pIC50 values for alpha-glucosidase activity were subjected to the ADMET assay in order to learn more about their pharmacological characteristics, including drug absorption, distribution, metabolism, excretion, and possible toxicity. To forecast these characteristics and evaluate how the substances will act in the human body, the pkCSM website is frequently utilized. Compounds with molecular weights under 500 g/mol, hydrogen bond donors under 5, hydrogen bond acceptors under 10, and LogP values below 5 were chosen for more investigation based on Lipinski's criteria. To further understand how the chemicals function in the body, parameters such human intestinal absorption, Caco-2 permeability, and steady-state volume of distribution (VDss) were investigated. These evaluations offer important insights into the compounds' potential for further study and advancement. [15]
QSAR Analysis:
A statistical technique called QSAR (Quantitative Structure-Activity Relationship) connects a chemical compound's structural characteristics to its biological activity. It is used to forecast the physicochemical characteristics and biological activities of molecules and assists in determining the essential elements of a medication that contribute to its efficacy. Animal suffering can be minimized by employing QSAR to lessen the necessity for costly and time-consuming animal experimentation. The bioactivity of organic (heterocyclic) compounds with different biological targets is frequently correlated using QSAR. It is becoming more and more crucial to create predictive models that relate molecular structure to useful characteristics. Particularly useful in domains with a wealth of biological target data, such as microbiology, is QSAR. Large volumes of experimental data have been produced by the automation of chemical synthesis and pharmacological screening, which has increased the efficiency of QSAR in the creation of compound libraries, data mining of molecular databases, and high-throughput animal investigations. Early physicochemical, pharmacokinetic, and toxicological property prediction using QSAR can help prevent expensive development failures. Because of its high hit rate and quick throughput, QSAR is a very useful approach. Chemical descriptors of molecular structures, which range from 1D to nD, are computed following the collection of chemogenomics data from databases and literature. We relate these descriptions to biological features using machine learning techniques. QSAR models can be used to forecast the biological activity of novel medications once they have been created and verified. To verify the accuracy of the QSAR model, experimental verification of computational hits is strongly advised, albeit it is not required. This synopsis illustrates the latest developments in QSAR-based virtual screening (VS) for drug discovery and shows how well it works to find possible compounds with the required characteristics. Additionally, it talks about the method's potential for the future and offers suggestions for best practices in QSAR-based VS. Although QSAR has been extensively employed to forecast the toxicity of conventional drug-like compounds, its use in predicting the toxicity of nanoparticles is still in its infancy. The use of QSAR techniques as an alternative to conventional toxicity evaluation is encouraged by the European REACH (Registration, Evaluation, Authorization and Restriction of Chemicals) system. Nano-QSAR applications are still in their infancy, despite the fact that QSAR models are very successful for traditional compounds. The speed, affordability, and potential to lessen the need for animal testing are some of the main benefits of QSAR models. They also aid in predicting biological activity and physicochemical characteristics, elucidating the processes of action of different compounds, and reducing the expenses associated with product development. Additionally, by boosting output and cutting waste, QSAR supports green chemistry. [16]
QSAR Studies of Benzimidazole Derivatives:
A broad class of substances known as benzoimidazoles is frequently employed as an antibacterial agent to combat a variety of infections. Research on the benzimidazole ring, an important heterocyclic structure, is still ongoing because of its wide range of pharmacological effects and ease in synthesis. The antifungal, antitubercular, antioxidant, and antiallergic qualities of these substances are well established. Additionally, it is a component of some herbicidal and anthelmintic formulations. Because of their broad-spectrum anthelmintic qualities, benzoimidazoles—such as albendazole, fenbendazole, and other methylcarbamate derivatives—as well as their sulfoxide counterparts are frequently utilized in both human and animal medicine. Numerous systemic parasite disorders, including as sporozoites, trichinosis, and larvae, as well as infections brought on by microsporidiosis and pneumocystis—which can cause severe diarrhea in those with compromised immune systems, like those living with HIV—can be effectively treated with this medication. Strong antiviral and antibacterial qualities have been shown by several benzimidazole derivatives, such as substituted benzimidazolyl quinolinyl mercaptotriazoles; their action against the Hepatitis C virus (HCV) has received special attention. There is continuous interest in creating novel and efficient processes for creating physiologically active benzimidazoles, even though there are a large variety of recognized benzimidazole derivatives. The identification of physiologically active compounds can be aided by research employing quantitative structure-activity analysis (QSAR). The capacity of QSAR to forecast the characteristics of novel compounds without requiring physical synthesis or actual testing is one of its main advantages. Because it connects a compound's structural characteristics to its biological activities and physicochemical characteristics, this approach is frequently employed in disciplines like as chemistry, pharmacology, and environmental research. Data gathering, molecular descriptor selection, correlation model creation, and model evaluation are some of the processes in the QSAR process. It offers prediction skills as well as greater insights into the mechanics underlying drug-receptor interactions. [17] The quality of the input data, the choice of pertinent descriptors and statistical methods, and—above all—the validation of the model are the main factors that determine a QSAR model's efficacy and dependability. The fact that QSAR models are only true for the chemical structures that were utilized to create them should not be overlooked. External validation (testing with a compound set that is not part of the training data), internal validation (cross-validation), and data randomization (also called Y-scrambling) are some of the approaches that can be used for validation. Many statistical approaches have been developed during the last few decades, increasing the variety of methodologies available for QSAR studies. These relationships have been successfully established through the use of computational techniques like multiple linear regression (MLR) and partial least squares (PLS). Artificial neural networks (ANNs) and support vector machines (SVMs) have gained popularity recently for application in QSAR research. Drug development is now increasingly using alternative molecular modeling techniques. Unsupervised techniques like self-organizing maps, supervised techniques like support vector machines, multilayer perceptrons, and Bayesian neural networks, and hybrid models like counter neural networks are some of the machine learning techniques (MLTs) employed in QSAR. The benefits of both supervised and unsupervised learning are combined in backpropagation networks, or CPNs. Because MLT uses a data collection of compounds with known biological activity for training and because each medication is linked to numerous factors whose individual contributions are initially unknown, it is especially well-suited for QSAR studies. [18]
QSAR Studies of Triazole Derivatives:
The QSARINS program, which is recognized to produce statistically sound GA-MLR-based models, was used to create the QSAR models. Using the software's random split function, the dataset was divided into training and test sets at random, with 80% going to the training set and 20% going to the test set. We were able to create models using representative subsets of the data and test them on other sets of compounds thanks to this technique. The goal of this strategy was to enhance model performance by incorporating as many molecular characteristics as feasible that influence or alter biological activity. The activity of the molecules was predicted or explained using the calculated molecular descriptors as independent variables in the equation Y = a0 a1X1 a2X2. In compliance with OECD guidelines, the model was subjected to stringent internal and external statistical testing, such as Y-randomization and application domain (AD) evaluation. To assess model performance and identify the optimal model, a number of statistical factors and evaluation metrics were taken into account. These consist of the root mean square error (RMSE), adjusted R² (R² adj), R² coefficient, and test set R² value (test R²). As a goodness-of-fit metric, the Leave-One-Out Cross Validation Coefficient (Q2 LOO) was also employed; values greater than 0.6 suggest a strong and trustworthy model. The coverage (AD) of the model was estimated by leverage analysis, which is depicted by Williams plots. With *n* representing the number of training samples and *k* representing the number of descriptors, the standardized residual (r) and leverage threshold (h* = (3 × (k - 1)) / n) were computed. The area in which the model can reliably forecast fresh data is known as the application domain. [15]
CONCLUSION:
The design and improvement of benzimidazole and triazole derivatives heavily relies on in silico methods including toxicity prediction, QSAR modeling, and molecular docking. These techniques help identify safer and more successful treatments by providing valuable insights into the safety profiles, interaction processes, and activity projections of possible drug candidates. The future of drug discovery and development will be advanced by ongoing advancements in computational chemistry and machine learning, which will increase the accuracy and effectiveness of these in silico methods.
REFERENCES
Shweta Kothavade*, Vaibhavi Patil, Dipak Khairnar, Rahil Khan, Pravin Jadhav, Vinod Bairagi, In Silico Studies Molecular Docking on Benzimidazole And Triazole, Toxicity and Qsar, Int. J. of Pharm. Sci., 2025, Vol 3, Issue 4, 1913-1924. https://doi.org/10.5281/zenodo.15223518