is a powerful approach in modern drug discovery. It uses the 3D structure of target proteins to guide the design of small molecules that can bind and modulate protein function. This method enables rational design of compounds with improved potency, selectivity, and properties.

Key steps in structure-based drug design include determining protein structures, identifying binding sites, designing ligands, and evaluating interactions. It leverages techniques like , NMR, and cryo-EM along with computational methods to optimize drug candidates efficiently.

Principles of structure-based drug design

  • Structure-based drug design (SBDD) leverages the three-dimensional structure of a target protein to guide the design and optimization of small molecule ligands that bind to the protein and modulate its function
  • SBDD plays a crucial role in modern drug discovery by enabling the rational design of compounds with improved potency, selectivity, and pharmacokinetic properties
  • Key steps in SBDD include determining the protein structure, identifying ligand binding sites, designing and optimizing ligands, and evaluating protein-ligand interactions

Protein structure determination for SBDD

  • Accurate determination of the target protein's three-dimensional structure is a prerequisite for SBDD
  • High-resolution structures provide detailed information about the protein's binding sites, conformational states, and potential ligand interactions
  • Three primary methods for protein structure determination in SBDD are X-ray crystallography, , and

X-ray crystallography in SBDD

Top images from around the web for X-ray crystallography in SBDD
Top images from around the web for X-ray crystallography in SBDD
  • X-ray crystallography is the most widely used method for determining high-resolution protein structures for SBDD
  • Involves growing protein crystals, exposing them to X-rays, and analyzing the diffraction patterns to determine the atomic coordinates of the protein
  • Provides detailed information about the protein's secondary and tertiary structure, as well as the binding modes of co-crystallized ligands (inhibitors, substrates)
  • Limitations include the need for high-quality protein crystals and the static nature of the resulting structures

NMR spectroscopy in SBDD

  • Nuclear magnetic resonance (NMR) spectroscopy is a powerful tool for studying protein structure and dynamics in solution
  • Provides information about the protein's secondary structure, flexibility, and ligand binding sites
  • Particularly useful for proteins that are difficult to crystallize or have multiple conformational states
  • Limitations include the requirement for isotopically labeled proteins and the size limitations of the technique

Cryo-electron microscopy for SBDD

  • Cryo-electron microscopy (cryo-EM) has emerged as a powerful method for determining high-resolution structures of large protein complexes and membrane proteins
  • Involves rapidly freezing protein samples in vitreous ice and imaging them using an electron microscope
  • Enables structure determination of proteins in near-native states and can capture different conformational states
  • Particularly useful for targets that are challenging for X-ray crystallography or NMR, such as G protein-coupled receptors (GPCRs) and ion channels

Computational methods in SBDD

  • Computational methods play a crucial role in SBDD by enabling the prediction and optimization of protein-ligand interactions
  • These methods leverage the structural information obtained from experimental techniques to guide the design and selection of potential drug candidates
  • Key computational approaches in SBDD include , , (QSAR), and

Molecular docking for ligand-protein interactions

  • Molecular docking is a computational method used to predict the binding mode and affinity of a ligand within a protein's binding site
  • Involves generating multiple ligand conformations and orientations within the binding site and evaluating their interactions using scoring functions
  • Enables virtual screening of large compound libraries to identify potential hits and guide the optimization of lead compounds
  • Limitations include the accuracy of scoring functions and the treatment of protein flexibility

Pharmacophore modeling in SBDD

  • Pharmacophore modeling involves identifying the essential structural features of a ligand that are responsible for its biological activity
  • A pharmacophore is an abstract 3D representation of these features, including hydrogen bond donors/acceptors, hydrophobic regions, and aromatic rings
  • Pharmacophore models can be used to screen virtual compound libraries, guide the design of new ligands, and optimize the selectivity of lead compounds
  • Particularly useful when multiple active ligands are known but the target protein structure is unavailable

Quantitative structure-activity relationships (QSAR) in SBDD

  • QSAR is a computational method that relates the structural features of a series of compounds to their biological activities
  • Involves developing mathematical models that correlate physicochemical properties (molecular weight, lipophilicity) and structural descriptors (fingerprints, pharmacophores) with experimental activity data
  • QSAR models can be used to predict the activities of new compounds, guide the optimization of lead compounds, and identify key structural features responsible for activity
  • Limitations include the need for high-quality experimental data and the applicability domain of the models

Molecular dynamics simulations for SBDD

  • Molecular dynamics (MD) simulations are used to study the dynamic behavior of proteins and protein-ligand complexes
  • Involves simulating the motion of atoms over time based on Newton's laws of motion and a molecular mechanics force field
  • Provides insights into protein flexibility, conformational changes, and the stability of ligand binding modes
  • Can be used to refine docking poses, evaluate the effects of mutations on ligand binding, and study the mechanisms of allosteric regulation
  • Limitations include the computational cost and the accuracy of the force fields used

Ligand design strategies in SBDD

  • Ligand design strategies in SBDD aim to optimize the interactions between a ligand and its target protein to improve potency, selectivity, and pharmacokinetic properties
  • These strategies leverage the structural information of the target protein and the knowledge of existing ligands to guide the design of new compounds
  • Key ligand design strategies in SBDD include , , and the optimization of lead compounds

Fragment-based drug discovery (FBDD)

  • FBDD is an approach that involves screening libraries of small molecular fragments (MW < 300 Da) against a target protein to identify weak but efficient binders
  • Fragments that bind to different regions of the binding site are then combined or grown to create larger, more potent ligands
  • Advantages of FBDD include the ability to explore a larger chemical space with fewer compounds and the potential to optimize ligand efficiency and physicochemical properties
  • Challenges include the need for sensitive biophysical screening methods (NMR, SPR) and the optimization of fragment linking or merging strategies

De novo drug design

  • De novo drug design involves the generation of novel ligands from scratch based on the structure of the target protein's binding site
  • Utilizes computational methods such as structure-based virtual screening, fragment linking, and retrosynthetic analysis
  • Enables the exploration of novel chemical spaces and the design of ligands with optimized interactions with the target protein
  • Challenges include the synthetic accessibility of the designed compounds and the need for extensive optimization of the initial hits

Ligand-based vs structure-based design

  • Ligand-based design strategies rely on the knowledge of known active compounds to guide the design of new ligands
  • Involves the use of pharmacophore models, QSAR, and similarity searching to identify compounds with similar structural features to the known actives
  • Structure-based design strategies rely on the knowledge of the target protein's structure to guide the design of new ligands
  • Involves the use of molecular docking, de novo design, and structure-based virtual screening to identify compounds that complement the binding site
  • Both approaches can be used in combination to leverage the strengths of each method and overcome their limitations

Optimization of lead compounds

  • Lead optimization is the process of improving the potency, selectivity, and pharmacokinetic properties of a promising lead compound
  • Involves the synthesis and testing of analogs with modifications to the lead compound's structure
  • Structure-based optimization strategies include the use of molecular docking and MD simulations to guide the design of analogs with improved interactions with the target protein
  • Ligand-based optimization strategies include the use of QSAR and pharmacophore models to guide the design of analogs with improved activity and selectivity
  • Multiobjective optimization approaches are often used to balance the improvement of multiple properties simultaneously (potency, selectivity, solubility, metabolic stability)

Protein-ligand interactions in SBDD

  • Understanding the types and strengths of interactions between a ligand and its target protein is crucial for the design of potent and selective compounds
  • Protein-ligand interactions can be classified into several categories, including , , , and
  • The optimization of these interactions is a key goal of SBDD and involves the use of computational methods and studies

Types of non-covalent interactions

  • Non-covalent interactions are the primary driving forces for ligand binding and include van der Waals interactions, hydrogen bonds, hydrophobic interactions, and electrostatic interactions
  • Van der Waals interactions are weak, short-range forces that arise from the fluctuations in the electron distributions of atoms
  • Hydrogen bonds are directional, electrostatic interactions between a hydrogen atom bonded to an electronegative atom (donor) and another electronegative atom (acceptor)
  • Hydrophobic interactions are the attractive forces between non-polar regions of a ligand and the protein, driven by the exclusion of water molecules from the binding site
  • Electrostatic interactions are the attractive or repulsive forces between charged or partially charged atoms, including salt bridges and cation-π interactions

Hydrogen bonding in ligand binding

  • Hydrogen bonds are one of the most important types of interactions in ligand binding, providing both specificity and affinity
  • The strength of a hydrogen bond depends on the distance and angle between the donor and acceptor atoms, as well as the polarity of the surrounding environment
  • In SBDD, hydrogen bonding patterns can be optimized by modifying the ligand's structure to introduce or remove hydrogen bond donors or acceptors
  • The use of bioisosteres (functional groups with similar properties) can be used to optimize hydrogen bonding while maintaining other desirable properties of the ligand

Hydrophobic interactions in ligand binding

  • Hydrophobic interactions are the attractive forces between non-polar regions of a ligand and the protein, driven by the exclusion of water molecules from the binding site
  • These interactions are important for the binding of lipophilic ligands and can contribute significantly to the overall
  • In SBDD, hydrophobic interactions can be optimized by modifying the ligand's structure to introduce or remove non-polar groups, or by exploiting the shape complementarity between the ligand and the binding site
  • The use of lipophilic efficiency (LipE) as a metric can guide the optimization of hydrophobic interactions while maintaining favorable physicochemical properties

Electrostatic interactions in ligand binding

  • Electrostatic interactions are the attractive or repulsive forces between charged or partially charged atoms in a ligand and the protein
  • These interactions can contribute to both the affinity and specificity of ligand binding, particularly for charged or polar ligands
  • In SBDD, electrostatic interactions can be optimized by modifying the ligand's structure to introduce or remove charged groups, or by exploiting the complementarity between the ligand's charge distribution and the electrostatic potential of the binding site
  • The use of electrostatic potential maps and free energy perturbation (FEP) calculations can guide the optimization of electrostatic interactions in ligand design

Applications of SBDD

  • SBDD has been successfully applied to the discovery and development of drugs for a wide range of therapeutic targets, including enzymes, receptors, and protein-protein interactions
  • The choice of the target protein and the specific ligand design strategy depends on the biological context and the desired therapeutic outcome
  • Examples of successful applications of SBDD include the development of kinase inhibitors, GPCR ligands, protease inhibitors, and antibody-based drugs

SBDD in kinase inhibitor development

  • Kinases are a large family of enzymes that play crucial roles in cell signaling and are attractive targets for the treatment of cancer and other diseases
  • SBDD has been extensively used in the development of kinase inhibitors, leveraging the wealth of structural information available for these enzymes
  • Examples of kinase inhibitors developed using SBDD include (Bcr-Abl inhibitor for chronic myeloid leukemia), gefitinib (EGFR inhibitor for non-small cell lung cancer), and vemurafenib (B-Raf inhibitor for melanoma)
  • Challenges in kinase inhibitor design include achieving selectivity among closely related kinases and overcoming resistance mutations

SBDD for G protein-coupled receptor (GPCR) ligands

  • GPCRs are a large family of membrane proteins that are involved in a wide range of physiological processes and are the targets of over 30% of approved drugs
  • SBDD has been increasingly used in the development of GPCR ligands, thanks to the growing number of high-resolution GPCR structures available
  • Examples of GPCR ligands developed using SBDD include the β2-adrenergic receptor agonist indacaterol (for asthma and COPD) and the orexin receptor antagonist suvorexant (for insomnia)
  • Challenges in GPCR ligand design include the flexibility and conformational dynamics of these proteins, as well as the need for subtype selectivity

SBDD in the design of protease inhibitors

  • Proteases are enzymes that catalyze the hydrolysis of peptide bonds and are involved in a variety of biological processes, including blood coagulation, viral replication, and cancer metastasis
  • SBDD has been successfully applied to the development of protease inhibitors, leveraging the structural information available for these enzymes and their active sites
  • Examples of protease inhibitors developed using SBDD include the HIV protease inhibitors saquinavir and nelfinavir, and the hepatitis C virus (HCV) protease inhibitors boceprevir and telaprevir
  • Challenges in protease inhibitor design include achieving selectivity among related proteases and optimizing the pharmacokinetic properties of the inhibitors

Antibody-based drug design using SBDD

  • Antibodies are a rapidly growing class of therapeutic agents that offer high specificity and affinity for their targets
  • SBDD has been applied to the design and optimization of antibody-based drugs, particularly in the context of antibody-antigen interactions
  • Examples of antibody-based drugs developed using SBDD include the anti-VEGF antibody bevacizumab (for cancer) and the anti-PCSK9 antibody evolocumab (for hypercholesterolemia)
  • Challenges in antibody-based drug design include the complexity of antibody structures and the need for efficient antibody humanization and optimization strategies

Challenges and limitations of SBDD

  • Despite the success of SBDD in drug discovery, several challenges and limitations remain that can affect the effectiveness and applicability of this approach
  • These challenges include the flexibility and conformational changes of proteins, the presence of water molecules in binding sites, the limitations of computational methods, and the need for integration with other drug discovery approaches

Protein flexibility and conformational changes

  • Proteins are dynamic entities that can undergo conformational changes upon ligand binding or in response to environmental factors
  • These conformational changes can affect the shape and properties of the binding site, making it challenging to design ligands that maintain their affinity and selectivity
  • In SBDD, protein flexibility can be addressed by using ensemble docking approaches, which consider multiple protein conformations, or by using induced fit docking methods that allow for protein flexibility during ligand binding
  • MD simulations can also be used to study the conformational dynamics of proteins and to identify potential binding sites and allosteric pockets

Dealing with water molecules in binding sites

  • Water molecules are often present in protein binding sites and can play crucial roles in ligand binding, either by mediating interactions between the ligand and the protein or by being displaced by the ligand
  • The presence of water molecules can complicate the interpretation of ligand binding modes and the optimization of ligand interactions
  • In SBDD, water molecules can be explicitly considered in docking and scoring calculations, or they can be implicitly accounted for using continuum solvation models
  • The use of MD simulations and free energy calculations can help to assess the energetic contributions of water molecules to ligand binding and to guide the design of ligands that optimize water-mediated interactions

Limitations of computational methods in SBDD

  • Computational methods used in SBDD, such as molecular docking and virtual screening, have several limitations that can affect their accuracy and reliability
  • These limitations include the accuracy of scoring functions used to evaluate ligand binding, the treatment of protein flexibility and solvation effects, and the completeness of the chemical space searched
  • The use of consensus scoring approaches, which combine multiple scoring functions, can help to improve the accuracy of docking and virtual screening results
  • The integration of experimental data, such as structure-activity relationships (SAR) and biophysical measurements, can also help to validate and refine computational predictions

Integration of SBDD with other drug discovery approaches

  • SBDD is often used in combination with other drug discovery approaches, such as high-throughput screening (HTS), fragment-based drug discovery (FBDD), and phenotypic screening
  • The integration of these approaches can help to overcome the limitations of each individual method and to accelerate the discovery and optimization of lead compounds
  • For example, SBDD can be used to guide the optimization of hits identified from HTS or FBDD, while phenotypic screening can provide valuable information about the biological activity and selectivity of the designed compounds
  • The use of multidisciplinary teams, including structural biologists, medicinal chemists, and computational scientists, is crucial for the successful integration of SBDD with other drug discovery approaches

Key Terms to Review (24)

3D Pharmacophore Modeling: 3D pharmacophore modeling is a computational technique used to identify and represent the essential features of a molecule that are responsible for its biological activity. This approach emphasizes the spatial arrangement of chemical groups that interact with a target protein, which helps in designing new drugs by predicting how different compounds may fit into the target's active site. By focusing on the three-dimensional orientation of these interactions, this method plays a crucial role in structure-based drug design.
Active Site: The active site is a specific region on an enzyme where substrate molecules bind and undergo a chemical reaction. This unique area is crucial for the enzyme's catalytic function, as it determines the enzyme's specificity and activity, making it essential in processes such as enzyme inhibition, drug design, and metabolic pathways.
ADMET: ADMET stands for Absorption, Distribution, Metabolism, Excretion, and Toxicity. It refers to the pharmacokinetic and pharmacodynamic properties of a drug that determine its behavior in the body and its overall effectiveness. Understanding ADMET is crucial in drug development, especially when utilizing structure-based drug design to optimize compounds for desired therapeutic outcomes while minimizing adverse effects.
Autodock: Autodock is a widely used computational tool for predicting how small molecules, like drugs, bind to a receptor of known 3D structure. It plays a crucial role in structure-based drug design by enabling researchers to visualize and analyze molecular interactions, making it easier to identify promising drug candidates. The software employs docking algorithms to simulate the binding process and assess the potential affinity between the ligand and the target protein.
Binding Affinity: Binding affinity refers to the strength of the interaction between a ligand, such as a drug or a neurotransmitter, and its target, usually a receptor or enzyme. A high binding affinity indicates that the ligand binds tightly to its target, which is crucial for both agonists and antagonists in eliciting or blocking biological responses. Understanding binding affinity is essential in drug discovery and optimization, as well as in designing effective therapies through various modeling and docking techniques.
Computational Chemistry: Computational chemistry is a branch of chemistry that uses computer simulations and models to study and predict molecular behavior, properties, and interactions. This field combines principles from quantum mechanics and thermodynamics to analyze the structures and energies of molecules, allowing researchers to explore complex chemical systems in ways that are often impractical with traditional experimental methods.
Cryo-electron microscopy: Cryo-electron microscopy (cryo-EM) is a powerful imaging technique that allows for the visualization of biological molecules at near-atomic resolution while in their native hydrated state. This method has revolutionized structural biology by enabling researchers to capture dynamic processes and conformational changes of macromolecules, making it an essential tool in structure-based drug design.
De novo drug design: De novo drug design is a computational approach used to create new pharmaceutical compounds from scratch, based on the understanding of biological targets and their interactions. This method leverages knowledge of the target's structure and biological function to generate novel chemical entities that are predicted to bind effectively to the target site. It emphasizes the design of unique molecules rather than modifying existing ones, enabling a more innovative approach to drug development.
Electrostatic interactions: Electrostatic interactions are forces that occur between charged particles, where opposite charges attract and like charges repel each other. These interactions are fundamental to various biological processes and play a significant role in molecular recognition, stability, and binding in the context of drug design and pharmacophore development.
Fragment-based drug discovery: Fragment-based drug discovery is a method used to identify small chemical fragments that can bind to biological targets, forming the basis for developing new drugs. This approach allows researchers to explore a vast chemical space efficiently, leading to the identification of potential lead compounds with improved binding affinities and selectivities during the drug development process.
Hydrogen bonding: Hydrogen bonding is a type of attractive interaction that occurs between a hydrogen atom covalently bonded to a highly electronegative atom and another electronegative atom. This phenomenon plays a crucial role in determining the structure and properties of biomolecules, influencing molecular interactions and stability. Hydrogen bonds can significantly affect the physical properties of compounds, their reactivity, and how they bind to biological targets, making them essential in various chemical and biological contexts.
Hydrophobic Interactions: Hydrophobic interactions are non-covalent forces that occur when non-polar molecules or regions of molecules aggregate in aqueous environments to minimize their exposure to water. This phenomenon is crucial for the stability and formation of biological structures such as proteins and cell membranes, and it plays a significant role in drug design and interactions at the molecular level.
Imatinib: Imatinib is a targeted therapy drug used primarily in the treatment of certain types of cancer, particularly chronic myeloid leukemia (CML) and gastrointestinal stromal tumors (GISTs). It functions as a tyrosine kinase inhibitor, blocking specific proteins that promote cancer cell proliferation, thereby inhibiting tumor growth and spread.
Molecular docking: Molecular docking is a computational technique used to predict the preferred orientation of a small molecule, such as a drug, when it binds to a target protein. This process helps in understanding the interaction between the ligand and its target, enabling the identification of potential therapeutic candidates and optimization of their binding properties.
Molecular dynamics simulations: Molecular dynamics simulations are computational methods used to model the physical movements of atoms and molecules over time. These simulations provide insights into the structure, dynamics, and thermodynamics of molecular systems by calculating the time-dependent behavior based on Newton's laws of motion. They play a vital role in understanding the interactions between drug candidates and biological targets, which is essential for effective drug design.
Nmr spectroscopy: NMR spectroscopy, or nuclear magnetic resonance spectroscopy, is a powerful analytical technique used to determine the structure of organic compounds by observing the magnetic properties of atomic nuclei. This method provides detailed information about the molecular structure, dynamics, and environment of atoms, making it an essential tool in various fields including medicinal chemistry, where understanding molecular interactions is crucial for drug development and design.
Non-covalent interactions: Non-covalent interactions are weak, reversible interactions between molecules that do not involve the sharing of electrons, unlike covalent bonds. These interactions play a crucial role in the stability and functionality of biological macromolecules, such as proteins and nucleic acids, and are essential for the design of effective drugs that target specific biomolecules.
Pharmacophore modeling: Pharmacophore modeling is a technique used in drug discovery that identifies and represents the essential features of a molecule required for biological activity. By creating a pharmacophore model, researchers can understand the spatial arrangement of atoms or functional groups that interact with a biological target, which helps in the design and optimization of new drugs. This approach plays a significant role in enhancing lead discovery and supports methods like structure-based drug design and molecular modeling.
Quantitative structure-activity relationships: Quantitative structure-activity relationships (QSAR) are mathematical models that relate the chemical structure of compounds to their biological activity. These relationships enable researchers to predict how different molecular features influence the efficacy and potency of drug candidates, facilitating the design of new pharmaceuticals by prioritizing compounds with desirable properties.
Schrödinger: Erwin Schrödinger was an Austrian physicist known for his contributions to quantum mechanics, particularly the formulation of the Schrödinger equation. This equation describes how the quantum state of a physical system changes over time and is a fundamental element in the field of quantum chemistry and its applications, including structure-based drug design.
Sildenafil: Sildenafil is a phosphodiesterase type 5 (PDE5) inhibitor that is primarily used to treat erectile dysfunction and pulmonary arterial hypertension. By inhibiting PDE5, sildenafil increases blood flow to the penis during sexual stimulation, which helps to achieve and maintain an erection. This drug's mechanism of action and its effectiveness have made it a key example in the context of structure-based drug design.
Structure-Activity Relationship (SAR): Structure-Activity Relationship (SAR) refers to the relationship between the chemical or 3D structure of a molecule and its biological activity. Understanding SAR is crucial for optimizing drug design, as it helps identify which structural features influence the effectiveness and potency of a compound against a biological target, guiding modifications to enhance desired properties.
Structure-based drug design: Structure-based drug design is a method that uses the 3D structures of biological targets to develop new medications. This approach allows scientists to visualize how potential drugs interact with their targets at the molecular level, enabling more efficient identification and optimization of lead compounds.
X-ray crystallography: X-ray crystallography is a powerful analytical technique used to determine the atomic and molecular structure of a crystal by diffracting X-ray beams through it. This method reveals the arrangement of atoms within a molecule, providing critical insights into the three-dimensional structures of biological macromolecules like proteins and nucleic acids. The ability to visualize these structures is essential for understanding interactions at the molecular level, which is crucial for various scientific applications, including the design of new drugs, discovery of novel drug fragments, and modeling pharmacophores.
© 2024 Fiveable Inc. All rights reserved.
AP® and SAT® are trademarks registered by the College Board, which is not affiliated with, and does not endorse this website.