💊Medicinal Chemistry Unit 7 – Drug design strategies

Drug design is a complex process that combines chemistry, biology, and pharmacology to create molecules that target specific biological pathways. It involves identifying targets, optimizing lead compounds, and considering factors like pharmacokinetics and drug delivery. Key strategies include structure-based design, ligand-based design, and fragment-based discovery. These approaches use computational tools and experimental techniques to develop potent, selective drugs with minimal side effects. The goal is to create effective treatments for various diseases.

Key Concepts in Drug Design

  • Drug design involves the rational design and synthesis of molecules that can interact with specific biological targets to elicit a desired therapeutic effect
  • Involves a multidisciplinary approach that integrates knowledge from chemistry, biology, pharmacology, and computational sciences
  • Aims to develop molecules with high affinity, selectivity, and potency for the target while minimizing off-target effects and toxicity
  • Considers the pharmacokinetic properties (ADME) of the drug to ensure optimal bioavailability, distribution, metabolism, and excretion
  • Utilizes various strategies such as structure-based drug design (SBDD), ligand-based drug design (LBDD), and fragment-based drug discovery (FBDD)
    • SBDD relies on the three-dimensional structure of the target protein to design complementary ligands
    • LBDD uses the knowledge of known ligands to identify common pharmacophoric features and guide the design of new molecules
    • FBDD involves screening small molecular fragments to identify weak binders that can be optimized into potent lead compounds
  • Employs computational tools and techniques such as molecular docking, virtual screening, and quantitative structure-activity relationship (QSAR) analysis to accelerate the drug discovery process
  • Iterative process that involves multiple rounds of design, synthesis, and biological evaluation to optimize the lead compounds

Target Identification and Validation

  • Target identification involves identifying a specific biological molecule (protein, enzyme, receptor, or nucleic acid) that plays a crucial role in the disease pathology and can be modulated by a drug to achieve a therapeutic effect
  • Targets can be identified through various approaches such as:
    • Genetic association studies that link specific genes to disease susceptibility
    • Expression profiling studies that identify differentially expressed genes or proteins in disease states
    • Phenotypic screening that identifies compounds with desired biological effects without prior knowledge of the target
  • Target validation is the process of confirming that modulation of the identified target will have a therapeutic benefit with minimal adverse effects
  • Involves various experimental techniques such as:
    • Genetic knockdown or knockout studies to assess the effect of target modulation on disease phenotype
    • Pharmacological studies using selective agonists, antagonists, or inhibitors to evaluate the therapeutic potential and safety of target modulation
    • Animal models that recapitulate the human disease to assess the efficacy and toxicity of target modulation in vivo
  • Considers the druggability of the target, which refers to the likelihood of developing a small molecule or biologic that can effectively modulate the target's activity
  • Assesses the selectivity of the target to minimize off-target effects and potential toxicity
  • Evaluates the potential for drug resistance and identifies strategies to mitigate its development

Structure-Activity Relationships (SAR)

  • SAR is the study of how the chemical structure of a molecule relates to its biological activity
  • Involves systematic modification of a lead compound to identify the key structural features that contribute to its activity and selectivity
  • Modifications can include changes to the scaffold, substituents, stereochemistry, and physicochemical properties
  • Aims to improve the potency, selectivity, and pharmacokinetic properties of the lead compound while minimizing toxicity
  • Utilizes various techniques such as:
    • Bioisosteric replacement, which involves replacing a functional group with another that has similar electronic and steric properties
    • Homologation, which involves adding or removing methylene groups to explore the effect of chain length on activity
    • Constraint of conformational flexibility, which involves introducing rings or double bonds to restrict the conformational space and improve binding affinity
  • Generates SAR tables that summarize the activity data for a series of analogs and help identify trends and structure-activity relationships
  • Guides the design of focused libraries that explore the SAR around a promising lead compound
  • Can be augmented by computational approaches such as 3D-QSAR and pharmacophore modeling to predict the activity of untested compounds

Pharmacophore Modeling

  • Pharmacophore modeling involves identifying the essential structural features of a ligand that are responsible for its biological activity
  • A pharmacophore is an abstract three-dimensional arrangement of chemical features (hydrogen bond donors/acceptors, hydrophobic regions, aromatic rings, etc.) that are necessary for optimal interaction with a specific biological target
  • Can be derived from a set of known active ligands (ligand-based pharmacophore) or from the structure of the target protein (structure-based pharmacophore)
  • Ligand-based pharmacophore modeling involves superimposing a set of active ligands to identify common chemical features and their spatial arrangement
    • Conformational flexibility of the ligands is considered, and multiple conformations are generated and aligned to identify the bioactive conformation
    • Features are identified based on their chemical properties and their frequency of occurrence in the active ligands
  • Structure-based pharmacophore modeling involves analyzing the binding site of the target protein to identify key interaction points and generate a pharmacophore model
    • Utilizes the knowledge of the protein-ligand complex structure obtained from X-ray crystallography or homology modeling
    • Identifies key amino acid residues in the binding site that interact with the ligand and maps their chemical features onto the pharmacophore model
  • Pharmacophore models can be used for virtual screening of large compound libraries to identify potential hits that match the pharmacophoric features
  • Can also guide the design of new molecules that incorporate the essential pharmacophoric features and optimize their spatial arrangement

Molecular Docking and Virtual Screening

  • Molecular docking is a computational technique that predicts the preferred orientation and binding mode of a ligand within the binding site of a target protein
  • Involves two main components: a search algorithm that generates possible ligand conformations and orientations within the binding site, and a scoring function that evaluates the strength of the protein-ligand interactions
  • Search algorithms can be classified into systematic methods (exhaustive search, conformational sampling) and stochastic methods (genetic algorithms, Monte Carlo simulations)
  • Scoring functions can be based on force fields (molecular mechanics), empirical data (regression-based), or knowledge-based potentials
  • Docking can be performed in a rigid or flexible manner, considering the conformational flexibility of the ligand and/or the protein
  • Virtual screening is the application of molecular docking to large compound libraries to identify potential hits that can bind to the target protein
    • Can be structure-based (docking-based) or ligand-based (pharmacophore-based, QSAR-based)
    • Enables the rapid and cost-effective exploration of chemical space to prioritize compounds for experimental testing
  • Docking and virtual screening results are evaluated based on various metrics such as docking score, ligand efficiency, and interaction fingerprints
  • Hits identified from virtual screening are further validated through experimental assays to confirm their activity and selectivity

Lead Optimization Strategies

  • Lead optimization is the process of improving the potency, selectivity, and pharmacokinetic properties of a lead compound to develop a clinical candidate
  • Involves iterative rounds of design, synthesis, and biological evaluation to optimize the lead compound
  • Strategies for lead optimization include:
    • SAR exploration to identify key structural features that contribute to activity and selectivity
    • Bioisosteric replacement to improve potency, selectivity, or pharmacokinetic properties
    • Scaffold hopping to explore different chemical scaffolds that maintain the key pharmacophoric features
    • Prodrug design to improve solubility, permeability, or stability
    • Conjugation with targeting moieties (antibodies, peptides) for targeted drug delivery
  • Utilizes various computational tools such as QSAR, molecular dynamics simulations, and ADMET prediction to guide the optimization process
  • Considers the physicochemical properties of the lead compound (molecular weight, lipophilicity, hydrogen bond donors/acceptors) and their impact on drug-like properties
  • Balances the improvement in potency and selectivity with the maintenance of favorable pharmacokinetic and safety profiles
  • Aims to develop a clinical candidate with optimal efficacy, safety, and pharmaceutical properties for further development

ADME Considerations

  • ADME (Absorption, Distribution, Metabolism, Excretion) properties of a drug candidate are critical determinants of its in vivo performance and clinical success
  • Absorption refers to the ability of the drug to cross biological membranes and enter the systemic circulation
    • Influenced by factors such as solubility, permeability, and efflux transporters
    • Can be predicted using in vitro assays (Caco-2, PAMPA) and in silico models (rule of five, QSAR)
  • Distribution refers to the extent and pattern of drug dissemination into various tissues and organs
    • Influenced by factors such as plasma protein binding, tissue affinity, and blood-brain barrier penetration
    • Can be predicted using in vitro assays (plasma protein binding, tissue distribution studies) and in silico models (QSAR, PBPK)
  • Metabolism refers to the biochemical modification of the drug by enzymes (cytochrome P450, UGTs) to facilitate its elimination
    • Can lead to the formation of active, inactive, or toxic metabolites
    • Can be predicted using in vitro assays (liver microsomes, hepatocytes) and in silico models (QSAR, structure-based modeling)
  • Excretion refers to the elimination of the drug and its metabolites from the body
    • Occurs primarily through renal (urine) and hepatic (bile) routes
    • Can be predicted using in vitro assays (hepatocyte stability, renal clearance) and in silico models (QSAR, PBPK)
  • ADME properties are optimized during lead optimization to ensure adequate bioavailability, target tissue exposure, and elimination
  • Poor ADME properties can lead to suboptimal efficacy, toxicity, or drug-drug interactions, and are a major cause of attrition in drug development

Drug Delivery Systems

  • Drug delivery systems are designed to improve the pharmacokinetic and pharmacodynamic properties of drugs by controlling their release, distribution, and targeting
  • Conventional drug delivery systems include:
    • Oral formulations (tablets, capsules) for systemic delivery
    • Parenteral formulations (injections, infusions) for rapid onset or long-acting effects
    • Topical formulations (creams, ointments) for local delivery to the skin or mucous membranes
  • Advanced drug delivery systems aim to overcome the limitations of conventional formulations and enhance the therapeutic efficacy and safety of drugs
  • Examples of advanced drug delivery systems include:
    • Controlled release formulations (matrix, reservoir) that provide sustained or pulsatile drug release over an extended period
    • Targeted drug delivery systems (antibody-drug conjugates, nanoparticles) that selectively deliver drugs to specific cells or tissues
    • Transdermal patches that allow for continuous drug delivery through the skin
    • Inhalation devices (metered-dose inhalers, dry powder inhalers) for pulmonary delivery of drugs
    • Implantable devices (pumps, stents) for localized drug delivery to specific organs or tissues
  • Drug delivery systems are designed based on the physicochemical properties of the drug (solubility, stability), the desired pharmacokinetic profile, and the target tissue or organ
  • Utilize various materials (polymers, lipids) and technologies (microencapsulation, nanoparticle synthesis) to achieve controlled release and targeting
  • Can improve patient compliance, reduce side effects, and enhance the therapeutic index of drugs
  • Drug discovery is an evolving field that constantly adapts to new scientific advances and technological innovations
  • Some of the emerging trends in drug discovery include:
    • Precision medicine: tailoring drug therapy based on individual patient characteristics (genetic profile, biomarkers) to optimize efficacy and safety
    • Immunotherapy: harnessing the power of the immune system to fight diseases such as cancer and autoimmune disorders
    • Gene therapy: delivering functional genes to replace or correct defective genes in genetic disorders
    • RNA therapeutics: targeting disease-causing RNA with antisense oligonucleotides, siRNA, or miRNA
    • Cell therapy: using living cells (stem cells, engineered cells) to replace or regenerate damaged tissues and organs
    • Microbiome-based therapy: modulating the gut microbiome to treat diseases such as inflammatory bowel disease, obesity, and neurological disorders
  • Emerging technologies such as CRISPR-Cas9 gene editing, organ-on-a-chip, and 3D bioprinting are enabling new approaches to drug discovery and development
  • Artificial intelligence (AI) and machine learning (ML) are being increasingly applied to various aspects of drug discovery, from target identification to lead optimization and clinical trial design
  • Open innovation models that involve collaboration between academia, industry, and government are facilitating the sharing of knowledge and resources to accelerate drug discovery
  • Regulatory agencies are adapting to the changing landscape of drug discovery by providing guidance on the development of novel therapeutic modalities and streamlining the approval process for breakthrough therapies


© 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.

© 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.