High-throughput screening (HTS) has revolutionized the drug discovery landscape, allowing researchers to test thousands to millions of biological, genetic, chemical, or pharmacological samples swiftly. This method accelerates the identification of promising candidates for further study. Integral to this process is the use of assays, which examine substances to determine their purity and properties. The signal detection system of these assays, known as readouts, measures the cell response, thereby providing crucial data about the results.

Ligand and structure-based drug designing process. Gupta, A., Kumar, S., K. Maurya, V., Puri, B., & K. Saxena, S. (2022). High-Throughput Screening for Drug Discovery toward Infectious Diseases: Options and Challenges. IntechOpen. doi: 10.5772/intechopen.102936

The advent of computer-aided drug design (CADD) marked a significant leap in drug development. This approach leverages various computational techniques supported by both the pharmaceutical industry and academic institutions to expedite the creation of new drugs.

The History of Computer-Aided Drug Design. Hideyoshi Fuji(藤 秀義. (2020). This is the updated version of Hideyoshi Fuji’s infographic which describes the history of Computer-Aided Drug Design (CADD). It is not sufficient for describing technologies, companies, etc. on CADD and is highly arguable, but it would be useful for overviewing the history. https://x.com/hideyoshifuji/status/1315670514438873088.

Among these techniques are quantitative structure-activity relationship (QSAR) models, pharmacophore modeling, lead optimization, molecular dynamics, and molecular docking. Recently, machine learning applications have been integrated into these methods, enhancing their efficiency and accuracy.

Schematic overview of the QSAR process. Nantasenamat, C., Isarankura-Na-Ayudhya, C. & Prachayasittikul, V. (2009). A Practical Overview of Quantitative Structure-Activity Relationship, EXCLI Journal 8:74-88. doi: 10.17877/DE290R-690.

Molecular docking is a computational technique that examines the interactions between a macromolecule, typically a protein, and a ligand. This method, which dates back to the 1980s, began with the molecular modeling of proteins and has since evolved into a cornerstone of modern drug discovery. By evaluating the interaction energy between a target protein and numerous ligands, researchers can identify potential drug candidates. The increase in computational power and the availability of extensive databases of ligands and proteins have made molecular docking indispensable in the search for new drugs.

Main applications of molecular docking in current drug discovery. Molecular docking is currently employed to help rationalizing ligands activity towards a target of interest and to perform structure-based virtual screening campaigns, similarly to as when it was first developed. Besides these applications, it can also be used to identify series of targets for which the ligands present good complementarity (target fishing and profiling), some of them being potentially responsible for unexpected drug adverse reactions (off-targets prediction). Moreover, docking is also currently employed for the identification of ligands that simultaneously bind to a pool of selected targets of interest (polypharmacology) and for identifying novel uses for chemical compounds with already optimized safety profiles (drug repositioning). Pinzi, Luca, and Giulio Rastelli. 2019. “Molecular Docking: Shifting Paradigms in Drug Discovery” International Journal of Molecular Sciences 20, no. 18: 4331. https://doi.org/10.3390/ijms20184331.

At the heart of molecular docking are two essential components: the conformational search algorithm and the scoring function. The conformational search algorithm explores the conformational space of the ligand at the binding site. There are two primary types of molecular docking: blind docking and binding-site docking. Blind docking involves the entire protein within the sampling space, ideal for identifying the site with the highest affinity energy when the binding site is unknown. In contrast, binding-site docking focuses on a specific area, typically the protein-binding site, or an allosteric site of interest.

Flux diagram of the conformer search algorithm. Ferro-Costas, D. & Fernandez-Ramos, A. (2020). A Combined Systematic-Stochastic Algorithm for the Conformational Search in Flexible Acyclic Molecules. Front. Chem., 28 January 2020. Sec. Theoretical and Computational Chemistry Volume 8. https://doi.org/10.3389/fchem.2020.00016.

The goal of the conformational search is to generate all possible conformations of the ligand for evaluation. This process considers the ligand’s structural parameters, such as torsion and translation, and its degrees of freedom. The number of conformations increases exponentially with the degrees of freedom, a phenomenon known as combinatorial explosion. Researchers employ either systematic or stochastic methods to perform the conformational search. Systematic methods evaluate small conformational changes until they converge to an energy minimum, while stochastic methods generate random conformations, covering a larger sampling area and increasing the likelihood of finding the global energy minimum.

Overview of the available workflows in CREST. https://crest-lab.github.io/crest-docs/page/overview/workflows.html.

The scoring function is a predictive model that calculates the binding free energy of each ligand-protein conformation. Traditional scoring functions are based on physical calculations of atomic interactions, known as force fields, which include potential energy, torsion terms, bond geometry, electrostatic terms, and the Lennard-Jones potential. Empirical scoring functions use weighted energy terms derived from experimental data, including hydrogen bonding, van der Waals interactions, and hydrophobicity. Knowledge-based scoring functions, on the other hand, rely on statistical models derived from large databases of ligand-protein complexes.

Schematics of the categories and datasets and evaluations of the protein–ligand scoring functions. Yang, Chao, Eric Anthony Chen, and Yingkai Zhang. 2022. “Protein–Ligand Docking in the Machine-Learning Era” Molecules 27, no. 14: 4568. https://doi.org/10.3390/molecules27144568.

Validating molecular docking protocols involves comparing the docking results with crystallography data using the root mean square deviation (RMSD). The DockBench platform facilitates this validation by performing autocoupling routines to replicate crystallographic complexes, thus measuring the ability of each protocol to reproduce the crystallographic pose.

RMSD per Residue. RMSD per residue values compare two proteins and show how well they align. Here we use RMSD values that compare solely the Cα atoms of the proteins. Investigating Structural Alignment. University of Illinois at Urbana-Champaign: NIH Resource for Macromolecular Modeling and Bioinformatics – Beckman Institute: Computational Biophysics Workshop. https://www.ks.uiuc.edu/Training/Tutorials/science/aquaporin/tutorial_aqp-html/node7.html.

A variety of software tools are available for molecular docking, each offering different conformational search algorithms and scoring function combinations. Consensus docking, which involves using multiple scoring functions to validate docking results, is a recommended practice. While combining three or four scoring functions can enhance results, achieving sufficient predictive power remains challenging, especially for highly flexible ligands. Consequently, new approaches incorporating machine learning are emerging to improve scoring functions.

Example of the correlation between the results of two docking programs. For estrogen receptor alpha (ESR1; structure 3ERT), each point corresponds to the rank of a molecule for AutoDock Vina versus the rank of the same molecule in ICM (see Methods). Red and grey circles are ligands and decoys, respectively. There is a poor correlation between the results of docking programs, and some ligands can be ranked well by one program but poorly by another (red circles in blue region). Traditional consensus approaches only take the best molecules for all programs (yellow region), acting as a conditional “and”, whereas the novel exponential consensus ranking (ECR) strategy takes the best molecules from either program, acting as a conditional “or” (taking both the yellow and blue regions). Palacio-Rodríguez, K., Lans, I., Cavasotto, C.N. et al. Exponential consensus ranking improves the outcome in docking and receptor ensemble docking. Sci Rep 9, 5142 (2019). https://doi.org/10.1038/s41598-019-41594-3.

Molecular docking represents a vital tool in the modern pharmacological arsenal, offering a sophisticated method for identifying potential drug candidates. By leveraging advanced computational techniques and integrating machine learning, researchers are poised to make significant strides in the discovery and development of new drugs. As technology continues to evolve, the accuracy and efficiency of molecular docking will only improve, paving the way for more effective and personalized treatments in the future.

Engr. Dex Marco Tiu Guibelondo, B.Sc. Pharm, R.Ph., B.Sc. CpE

Editor-in-Chief, PharmaFEATURES

Share this:

This website uses cookies to improve your experience. We'll assume you're ok with this, but you can opt-out if you wish. Cookie settings