Ligand-based drug design uses known active compounds to find new drug candidates. It analyzes chemical features and structures to guide the search. This approach includes modeling, quantitative structure-activity relationships (), and molecular similarity analysis.

These methods help researchers explore chemical space, predict drug properties, and screen large libraries. By leveraging existing knowledge, ligand-based design can speed up drug discovery and lead to novel compounds with desired biological activity.

Ligand-based drug design overview

  • Ligand-based drug design (LBDD) relies on the analysis of known ligands to identify new compounds with desired biological activity
  • LBDD methods exploit the chemical and structural features of known active compounds to guide the design of new drug candidates
  • Encompasses a range of computational approaches including pharmacophore modeling, quantitative structure-activity relationships (QSAR), and molecular similarity analysis

Pharmacophore modeling

Types of pharmacophore models

Top images from around the web for Types of pharmacophore models
Top images from around the web for Types of pharmacophore models
  • Ligand-based pharmacophore models derived from a set of known active compounds sharing a common biological target
  • Structure-based pharmacophore models generated from the analysis of ligand-target interactions in available crystal structures
  • Integrated pharmacophore models combine information from both ligand and target structures to enhance model quality and predictive power

Pharmacophore elucidation methods

  • Manual pharmacophore design involves visual inspection and alignment of active compounds to identify common features
  • Automated pharmacophore generation algorithms (HipHop, HypoGen) align compounds and extract pharmacophoric features based on predefined rules and scoring functions
  • Consensus pharmacophore approaches combine multiple pharmacophore models to improve robustness and applicability

Pharmacophore-based virtual screening

  • Pharmacophore models serve as 3D queries to screen large compound libraries and identify potential hits with similar pharmacophoric features
  • Pharmacophore screening can be combined with other filters (physicochemical properties, ADME criteria) to prioritize compounds for experimental testing
  • Successful applications of pharmacophore-based virtual screening have led to the discovery of novel bioactive compounds for various therapeutic targets (HIV protease inhibitors, kinase inhibitors)

Quantitative structure-activity relationships (QSAR)

QSAR model development process

  • QSAR modeling establishes mathematical relationships between structural features (descriptors) and biological activity of a set of compounds
  • Key steps include data collection and curation, descriptor calculation, feature selection, model building, and validation
  • QSAR models can guide the optimization of lead compounds by predicting the activity of untested analogs

2D vs 3D QSAR approaches

  • 2D QSAR methods (Free-Wilson, Hansch analysis) rely on 2D structural features (substituents, fragments) to correlate with activity
  • 3D QSAR approaches (CoMFA, CoMSIA) consider the 3D alignment of compounds and calculate steric, electrostatic, and other field-based descriptors
  • 3D QSAR models provide insights into the 3D requirements for optimal ligand-target interactions and can guide structure-based design efforts

QSAR model validation & applicability domain

  • QSAR models must be rigorously validated using internal (cross-validation) and external (test set) validation techniques to assess their predictive performance
  • Applicability domain analysis determines the chemical space where the QSAR model can make reliable predictions based on the training set composition
  • Consensus QSAR models that combine predictions from multiple models can improve the robustness and applicability of QSAR-based virtual screening

Molecular similarity & diversity

Similarity & diversity measures

  • Molecular similarity quantifies the structural resemblance between compounds using 2D (fingerprint-based) or 3D (shape-based) approaches
  • Diversity analysis assesses the chemical space coverage and structural dissimilarity within a compound set
  • Commonly used similarity metrics include Tanimoto coefficient (fingerprints) and Tversky index (pharmacophoric features)

Chemical space exploration

  • Chemical space represents the vast collection of all possible chemical compounds, estimated to exceed 106010^{60} molecules
  • Sampling and navigating chemical space is crucial for identifying novel scaffolds and expanding the structural diversity of compound libraries
  • Techniques such as principal component analysis (PCA) and t-distributed stochastic neighbor embedding (t-SNE) help visualize and explore chemical space

Scaffold hopping & bioisosteric replacement

  • Scaffold hopping involves the identification of novel chemotypes that maintain the desired biological activity but possess distinct molecular scaffolds
  • Bioisosteric replacement strategies substitute functional groups or substructures with bioisosteres that have similar physicochemical properties but improved ADME or profiles
  • Examples of successful scaffold hopping include the discovery of non-benzodiazepine anxiolytics (buspirone) and non-nucleoside reverse transcriptase inhibitors (NNRTIs) for HIV treatment

Machine learning in ligand-based design

Supervised vs unsupervised learning

  • Supervised learning algorithms (random forest, support vector machines) learn from labeled data to predict the properties of new instances
  • Unsupervised learning methods (clustering, dimensionality reduction) uncover hidden patterns and relationships in unlabeled data
  • Semi-supervised learning combines labeled and unlabeled data to improve model performance and generalizability

Common machine learning algorithms

  • Decision trees and random forests construct models based on a series of binary decisions and can handle both classification and regression tasks
  • Support vector machines (SVMs) find optimal hyperplanes to separate classes in high-dimensional feature space and are effective for QSAR modeling
  • Deep learning architectures (convolutional neural networks, graph neural networks) can learn hierarchical representations from raw molecular data and have shown promising results in virtual screening and property prediction

Feature selection & model interpretation

  • Feature selection techniques (recursive feature elimination, L1 regularization) identify the most informative descriptors and reduce model complexity
  • Model interpretation methods (feature importance, SHAP values) provide insights into the contributions of individual features to the model predictions
  • Interpretable machine learning models (decision trees, rule-based systems) offer greater transparency and explanatory power compared to black-box models

Ligand-based virtual screening

Ligand-based VS methodologies

  • Ligand-based virtual screening (LBVS) aims to identify novel active compounds based on their similarity to known ligands
  • LBVS approaches include pharmacophore screening, similarity searching (2D fingerprints, 3D shape), and machine learning-based methods
  • Consensus scoring strategies that combine multiple LBVS methods can improve the enrichment of active compounds in virtual screening campaigns

Ligand-based VS case studies

  • LBVS has been successfully applied to discover novel inhibitors for various therapeutic targets, including kinases (EGFR, CDK2), G protein-coupled receptors (GPCRs), and ion channels
  • Integration of LBVS with high-throughput screening (HTS) and structure-based design has led to the identification of potent and selective lead compounds (MEK inhibitors, BACE1 inhibitors)
  • LBVS has also been employed to repurpose existing drugs for new therapeutic indications (antiviral agents, anticancer compounds)

Combining ligand & structure-based approaches

  • Ligand-based and approaches provide complementary information and can be combined to enhance the efficiency and success of drug discovery efforts
  • Ligand-based pharmacophore models can guide the docking and scoring of compounds in structure-based virtual screening
  • Ligand-based SAR data can be integrated with structural insights from co-crystal structures to optimize ligand-target interactions and improve binding and selectivity

ADME prediction in ligand-based design

Physicochemical property prediction

  • Prediction of key physicochemical properties (molecular weight, logP, polar surface area) is essential for assessing the drug-likeness and ADME behavior of compounds
  • Ligand-based QSAR models and machine learning algorithms can predict physicochemical properties based on molecular descriptors and structural features
  • Compliance with drug-likeness rules (Lipinski's rule of five, Veber's rules) can be used as filters to prioritize compounds with favorable ADME profiles

Pharmacokinetic parameter estimation

  • Pharmacokinetic parameters (, , , ) determine the fate of a drug in the body and impact its efficacy and safety
  • Ligand-based QSAR models can predict pharmacokinetic properties such as intestinal absorption, blood-brain barrier permeability, and metabolic
  • Integration of pharmacokinetic predictions with pharmacodynamic data enables the optimization of drug exposure and therapeutic effect

Toxicity & off-target effect prediction

  • Prediction of potential and off-target effects is crucial for avoiding adverse drug reactions and improving drug safety
  • Ligand-based approaches can identify structural alerts and toxicophores associated with specific toxicity endpoints (genotoxicity, cardiotoxicity)
  • Machine learning models trained on large toxicity databases (Tox21, ToxCast) can predict the likelihood of a compound causing toxicity based on its structural features
  • Off-target profiling using ligand-based similarity searches can identify potential unintended targets and guide the design of more selective compounds

Challenges & limitations

Activity cliff & activity landscape

  • Activity cliffs represent pairs of structurally similar compounds with large differences in biological activity, posing challenges for QSAR modeling and similarity-based approaches
  • Activity landscape analysis visualizes the structure-activity relationships (SAR) of a compound series and identifies regions of discontinuous SAR
  • Addressing activity cliffs requires careful consideration of the structural features responsible for the activity differences and may necessitate local QSAR models or 3D approaches

Handling conformational flexibility

  • Conformational flexibility of ligands and targets introduces additional complexity in ligand-based drug design, as different conformations may have distinct biological activities
  • Conformational sampling techniques (molecular dynamics, low-mode conformational search) generate ensemble representations of ligands for pharmacophore modeling and 3D QSAR
  • Consensus approaches that consider multiple conformations can improve the robustness and predictive power of ligand-based models

Addressing data quality & quantity issues

  • The quality and quantity of available bioactivity data greatly influence the performance and applicability of ligand-based drug design methods
  • Data curation and standardization procedures are essential for ensuring the consistency and reliability of the input data
  • Strategies to address data limitations include data augmentation (SMOTE), transfer learning, and multi-task learning approaches
  • Collaboration and data sharing initiatives (ChEMBL, PubChem) play a crucial role in expanding the chemical and biological space for ligand-based drug design

Key Terms to Review (25)

Absorption: Absorption is the process by which substances, such as drugs, are taken up into the bloodstream after administration. This process is crucial for determining how much of a drug reaches systemic circulation and its effectiveness. Factors such as the route of administration, chemical properties of the drug, and physiological conditions play a significant role in influencing absorption rates.
Affinity: Affinity refers to the strength of the interaction between a ligand and its target receptor or protein. It plays a crucial role in determining how effectively a drug can bind to its target, influencing the overall efficacy and potency of the therapeutic agent. Understanding affinity is essential when designing drugs that will interact with specific biological targets, allowing for better therapeutic outcomes.
Agonist: An agonist is a substance that binds to a receptor and activates it, leading to a biological response. This process is fundamental in pharmacology, as agonists can mimic the action of natural ligands, triggering signaling pathways that result in physiological effects. Understanding how agonists interact with receptors helps in predicting drug efficacy and designing effective therapies.
Antagonist: An antagonist is a type of drug or molecule that binds to a receptor but does not activate it, effectively blocking or dampening the biological response that would normally occur upon activation. This can be crucial in regulating various physiological processes, making antagonists essential tools in pharmacology and drug design.
Bioavailability: Bioavailability refers to the proportion of a drug or substance that enters the systemic circulation when it is introduced into the body, making it available for therapeutic effect. This concept is crucial because it influences how effectively a drug performs in its intended role, impacting factors like dose-response relationships and absorption rates.
Chiral Center: A chiral center is a carbon atom that is bonded to four different substituents, resulting in non-superimposable mirror images, known as enantiomers. This concept is crucial in drug design because the three-dimensional arrangement of atoms can significantly influence a drug's biological activity and interaction with target proteins. Understanding chirality helps chemists design molecules that can specifically interact with biological systems, leading to more effective and safer therapeutics.
Clearance: Clearance refers to the rate at which a drug is removed from the body, typically measured in volume per unit time. This concept is crucial in pharmacokinetics as it influences the duration and intensity of a drug's effect, as well as its dosing regimen. Understanding clearance is essential for optimizing drug design and ensuring therapeutic efficacy while minimizing toxicity.
Distribution: Distribution refers to the process by which a drug is dispersed throughout the body's fluids and tissues after administration. It involves understanding how factors like blood flow, tissue permeability, and the binding of drugs to proteins influence the extent and rate at which a drug reaches its target sites, impacting efficacy and safety.
Excretion: Excretion is the biological process of removing waste products and excess substances from an organism's body. This process is essential for maintaining homeostasis and preventing the accumulation of harmful metabolites, which is particularly relevant when discussing metabolism, drug design, and drug disposition in pharmacokinetics.
Fragment-based drug design: Fragment-based drug design is a method used in medicinal chemistry that focuses on identifying and optimizing small chemical fragments that bind to a target protein. By starting with simpler, smaller compounds, this approach allows researchers to create more effective drugs through the systematic growth and modification of these fragments into larger, more complex molecules. It emphasizes the importance of understanding the binding interactions between ligands and their targets, leading to better drug candidates with improved properties.
Half-life: Half-life is the time required for the concentration of a substance to reduce to half its initial value, playing a crucial role in pharmacokinetics and drug metabolism. This concept helps in understanding how long a drug remains effective in the body, influencing dosing schedules, excretion rates, and therapeutic effects.
Lipophilicity: Lipophilicity refers to the chemical affinity of a substance for lipids or fats, essentially indicating how well a compound can dissolve in non-polar solvents compared to polar solvents. This property plays a crucial role in determining how drugs are distributed in the body, how they are metabolized, and their overall efficacy as therapeutic agents. Compounds with high lipophilicity tend to penetrate cell membranes more easily, influencing their absorption and distribution, while also impacting drug design and the physicochemical properties that affect their action in biological systems.
Metabolism: Metabolism refers to the complex set of biochemical reactions that occur within living organisms to maintain life, including the conversion of food into energy and the synthesis of necessary compounds. It plays a crucial role in drug development and pharmacology, influencing how drugs are processed in the body, their efficacy, and potential side effects, particularly in the context of specific therapeutic areas and drug design.
Partial Agonist: A partial agonist is a compound that binds to a receptor and activates it, but to a lesser degree than a full agonist. This means that while a partial agonist can elicit a response from the receptor, the magnitude of that response is not as strong as that produced by a full agonist. Partial agonists can play crucial roles in pharmacology, especially in the development of drugs that aim to modulate receptor activity rather than fully stimulate or block it.
Pharmacophore: A pharmacophore is the set of structural features in a molecule that is necessary for its biological activity. This concept helps in understanding how different compounds can interact with a target protein, providing a framework for designing new drugs and optimizing existing ones. By identifying the essential elements that confer activity, scientists can focus on modifying lead compounds to enhance efficacy and selectivity.
PKa: pKa is a quantitative measure of the strength of an acid in solution, defined as the negative logarithm of the acid dissociation constant (Ka). It indicates the tendency of a compound to donate a proton (H+) in a chemical reaction, which is crucial for understanding how substances behave in biological systems and drug interactions. The pKa value helps predict the ionization state of a molecule at a given pH, making it an essential concept in assessing physicochemical properties and guiding ligand-based drug design.
Potency: Potency refers to the strength of a drug or compound in producing a desired effect, often measured by the concentration or dose needed to achieve that effect. It reflects how much of a substance is required to elicit a response and is crucial in understanding dose-response relationships and optimizing ligand-based drug design. A more potent drug requires a lower dose to achieve the same effect as a less potent one, which is vital for both efficacy and safety in pharmacology.
QSAR: Quantitative Structure-Activity Relationship (QSAR) is a computational technique used to predict the biological activity of chemical compounds based on their chemical structure. It involves statistical modeling that correlates the chemical or structural properties of compounds with their observed effects or activities, aiding in ligand-based drug design by guiding the optimization of lead compounds to enhance efficacy and reduce toxicity.
Radiolabeled assays: Radiolabeled assays are analytical techniques that use radioactively labeled compounds to detect and quantify biological substances in various samples. This method is essential in studying ligand-receptor interactions, as the radiolabel allows for sensitive detection of binding events, which is crucial in understanding drug efficacy and mechanism of action.
Selectivity: Selectivity refers to the ability of a drug or compound to preferentially bind to a specific target, such as a receptor or enzyme, while minimizing interactions with other targets. This characteristic is crucial for enhancing therapeutic efficacy and reducing side effects, making it a central concept in drug design and optimization processes. Understanding selectivity is essential for developing drugs that provide maximum therapeutic benefit while limiting undesirable effects on non-target systems.
Solubility: Solubility is the ability of a substance to dissolve in a solvent, forming a homogeneous solution. It plays a crucial role in various biological processes, drug formulation, and the design of pharmaceuticals, influencing how compounds are absorbed, distributed, metabolized, and excreted by the body.
Stability: Stability refers to the ability of a molecule or system to maintain its structure and function over time, resisting degradation or change under various conditions. In drug design and delivery, stability is crucial as it influences the effectiveness, safety, and shelf life of a drug. Factors such as chemical composition, environmental conditions, and interactions with other molecules all play a role in determining the stability of a drug formulation or delivery system.
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.
Surface Plasmon Resonance: Surface plasmon resonance (SPR) is an optical technique that measures the refractive index near the surface of a sensor chip, typically used to study biomolecular interactions in real-time. By shining light onto a metal surface, such as gold or silver, it excites surface plasmons, which are collective oscillations of electrons at the surface, and any changes in the interaction can be detected as shifts in the light reflected from the surface. This method is particularly valuable in assessing binding kinetics and affinities of ligands and fragments during drug design and discovery processes.
Toxicity: Toxicity refers to the degree to which a substance can harm living organisms. It encompasses various factors such as dose, exposure route, and the specific biological mechanisms that lead to harmful effects. Understanding toxicity is crucial in many fields, particularly in drug development, as it helps predict how compounds will behave in biological systems and informs safety assessments.
© 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.