Over the past few years, scientists have synthesized or isolated a large number of new bioactive molecules from natural sources. However, despite the enormous resources invested in new drug discovery over the past decade, with thousands of new drug candidates proposed each year, only a very small number are ultimately selected to enter clinical trials. So why is progress in medicinal chemistry so slow?
One major reason is the need for more efficient methods to design and screen promising drug candidates at a lower cost. In addition, the entire drug discovery process must be optimized to enable faster transitions from initial discovery to later development stages. While there have been many breakthroughs in medicinal chemistry, much of the progress is often driven by the identification of new drug targets, such as metabolic pathways or enzymes. However, the complexity of these processes and the vast number of compounds that need to be tested presents significant challenges.
This is where computational chemistry has become invaluable. It provides strong support for experimental techniques, enabling more efficient drug discovery. One such technique, known as virtual screening, was first introduced in the late 1990s to describe the process of identifying potential therapeutic molecules using computerized methods. Since then, many advanced software tools and computational strategies have been developed, greatly enhancing the ability to discover new lead compounds.
In this article, the relationship between molecular docking and potential drug development will be described in detail, and the computational strategies used in the molecular docking process will be described in detail.
Molecular docking is a computational technique used to predict the interaction between a small molecule or ligand and a target protein (usually a receptor or enzyme). The goal of docking is to understand how the ligand ADAPTS to the binding site of the protein, giving insight into the strength and nature of the interaction. In the drug discovery process, molecular docking plays a crucial role in identifying potential drug candidates and optimizing drug design. By simulating how various compounds interact with specific targets, researchers can quickly screen large libraries of molecules to identify those most likely to be effective. This is essential for virtual screening techniques, lead optimization, and more selective development of effective drugs with fewer side effects. Molecular docking not only speeds up the drug development process but also helps researchers explore new therapeutic targets and understand resistance mechanisms.
Although molecular docking is closely related to the following applications in drug discovery, some functions overlap, but each part of the focus and application direction is different.
Target identification and validation focuses on the selection of the target and the validation of its feasibility through docking. Virtual screening focuses on the screening process and efficient discovery of potential drug candidates. Specific inhibitor design involves molecular optimization and precise inhibition, focusing on the application of therapeutic targets. Improving bioavailability and selectivity focuses more on the clinical availability of drugs, especially the in vivo behavior of drugs. See below for more details.
Target identification and validation is are the first step steps in drug discovery, which is to identify biomolecules that are suitable as drug targets. Molecular docking techniques play a crucial role in this phase, and by analyzing the three-dimensional structure of the target protein and simulating its interactions with potential ligands, researchers can identify targets with therapeutic potential. It is unique in that molecular docking can not only help discover new targets but also verify the effectiveness of existing targets, ensuring that the target selection in the drug development process is reasonable. This process lays the foundation for subsequent drug design and optimization and ensures that the drug can accurately act on the right biological target.
A single docking experiment helps explore the function of a target, while virtual screening, in which a large library of compounds is docked and sequenced, can be used to identify new inhibitors for drug development.
For example, the work of Roney, M., et al. describes the computer simulation screening of substances in the drug library that act as EGFR inhibitors. First, the drug library was screened using pharmacophore techniques to select ligands, and erlotinib (DB00530) was used as a matrix compound. The selected ligand is screened using ADMET and the hit compound is docked. Using pharmacophore technology, 23 compounds were identified through virtual drug library screening, culminating in the identification of the promising lead molecule DB03365, which outperforms the reference compounds in terms of performance, but in vitro and in vivo experiments are still needed to validate the study.
Molecular docking plays a key role in the design of specific inhibitors, especially in developing drugs for certain diseases such as cancer or viral infections. Through molecular docking, researchers can design molecules that precisely bind to and inhibit the target's function, based on its known structure. It is unique in that molecular docking not only predicts the binding pattern but also enhances the specificity and inhibitory effect on the target by optimizing the compound's structure. In this way, researchers can design highly specific and highly effective drugs that reduce side effects and improve treatment effectiveness.
The bioavailability and selectivity of a drug are key factors in determining its clinical efficacy. Molecular docking can help optimize the molecular structure of drugs so that their absorption, distribution, and targeting in the body are ideal. By docking to predict and optimize drug interactions with targets, researchers are not only able to improve drug affinity but also ensure that drugs have good bioavailability and high selectivity, avoiding unwanted side effects. What is unique about this process is that it focuses on the performance of the drug in vivo, ensuring that the drug can effectively reach the target and function steadily in the body over a long period. This not only accelerates the clinical application of the drug but also improves the therapeutic safety of the drug.
In the context of Molecular Docking Software and Tools, we use AutoDock as an example to explain its detailed application. The following will help you better understand the framework of molecular docking.
Figure 1: Workflow of a prospective molecular docking screen. (Ballante et al.,2021)
The structural information of the target protein is critical in the drug discovery process because it provides the necessary spatial and conformational basis for molecular docking. The target structure obtained by different methods may improve the accuracy and predictability of molecular docking from different angles.
Prior to molecular docking, the obtained protein structure data usually requires a series of preprocessing to ensure the accuracy of the simulation results. The main tasks of this stage include filling in the missing residues, determining the protonation state, and the treatment of water molecules.
In the early stages of drug discovery, compound library selection is a critical step. Compound libraries are composed of molecules with a variety of chemical structures that are used for virtual screening with targets.
When a compound binds to a target, it may need to change its conformation to suit the binding site of the target, so conformational sampling and optimization are important steps in molecular docking. Since small molecules often have multiple viable conformations, properly sampling these conformations and selecting the best structure is critical to improving the accuracy of docking results. Once multiple conformations have been generated, these need to be optimized to ensure that each conformation reaches a stable state with the lowest energy.
Different docking software and methods may employ different protocols, but the goal of all protocols is to simulate how a molecule (usually a ligand) binds to a target protein and predict its affinity.
Before docking, the target protein (receptor) and ligand need to be prepared. The target protein is usually pretreated by cleaning the structure, removing water molecules, supplementing the missing amino acid residues, and determining the protonation state. The ligands also need to undergo geometric optimization, protonation, ionization, and other treatments.
This is the core step of molecular docking. By selecting a suitable docking algorithm, the binding process of the ligand and target is simulated. Docking software such as AutoDock, Dock, Glide, etc., uses different docking methods (such as molecular docking, rigid docking, flexible docking, etc.) to generate possible bonding patterns between ligands and targets. The choice of different algorithms usually depends on the balance of calculation accuracy and time.
The results of docking are evaluated by the scoring function, which assigns a score to each ligand-receptor binding mode according to the binding affinity, geometric matching degree, hydrogen bonding, charge distribution, and other factors between ligand and target. Common scoring functions include grid score, force field score, and knowledge base score. Higher conformations are considered to have a stronger binding affinity.
For information on how to interpret the docking results and improve the molecular binding model based on the docking results, see the article Molecular Docking Results Analysis and Accuracy Improvement.
In molecular docking simulation, computational cost and time are the key factors affecting the efficiency of virtual screening. The main influencing factors include the size of the receptor and ligand, the complexity of the selected docking algorithm, the accuracy of the scoring function, and the scale of the virtual screening. Larger target proteins and flexible ligands increase the computational burden, while complex docking algorithms and scoring functions typically require more computational resources to provide greater accuracy. In addition, the size of the virtual screening compound library also significantly affects the computation time. To improve efficiency, parallel computing, and high-performance computing platforms are often used to reduce simulation time and process large-scale data. Through a reasonable selection of docking methods and optimization technologies, it is possible to balance the needs of computing resources and time while ensuring accuracy.
In antibody drug development, molecular docking can be used to predict the binding pattern of an antibody to an antigen, thereby helping to optimize the affinity and specificity of the antibody. Through docking simulations, researchers can design monoclonal antibodies with more efficient immune responses and improve the efficacy of antibody drugs, especially in the treatment of cancer, infectious diseases, and other fields.
You can read the article Molecular Docking Techniques and Methods, which provides a detailed overview of common molecular docking strategies and their in-depth applications.
Artificial intelligence (AI) is revolutionizing the efficiency and accuracy of molecular docking. AI technology can analyze large amounts of experimental data, automatically identify potential drug targets, optimize docking processes, and improve the prediction accuracy of score functions through deep learning models. This not only accelerates the speed of virtual screening but also enhances the quality and selectivity of drug candidate molecules.
One example is VirtualFlow, a highly automated and versatile open-source platform with perfect scaling behavior and the ability to use the most powerful interfacing programs to greatly increase efficiency.
Figure 2: Application of VirtualFlow to the drug discovery process (Gogula et al.,2020)
In personalized medicine, molecular docking technology can predict how patients will respond to specific drugs by integrating their genomic information. By taking into account individualized protein structure changes and drug interactions, molecular docking is expected to drive the development of precision therapy and help optimize drug selection and dosing, thereby providing patients with more effective and safer personalized treatment options. With the continuous progress of technology, molecular docking will be combined with more advanced technologies such as artificial intelligence and genomics in the future to improve the efficiency and accuracy of drug discovery and development, laying a solid foundation for personalized medicine and optimizing treatment plans.
Although molecular docking has significant advantages in the early stages of drug discovery, enabling computational prediction of potential candidates and improved screening efficiency, it still faces limitations such as model assumptions and environmental complexity, resulting in results that are not fully comparable to traditional experimental methods. Traditional methods of experimental drug discovery provide greater accuracy and reliability through direct biological experiments, although the process is more time-consuming and costly. To bridge this gap, combining the advantages of molecular docking and traditional experimental methods can ensure efficient screening while maintaining the accuracy and reliability of drug discovery.
References