Fiveable

๐ŸงฌSystems Biology Unit 12 Review

QR code for Systems Biology practice questions

12.3 Systems approach to drug discovery and development

๐ŸงฌSystems Biology
Unit 12 Review

12.3 Systems approach to drug discovery and development

Written by the Fiveable Content Team โ€ข Last updated September 2025
Written by the Fiveable Content Team โ€ข Last updated September 2025
๐ŸงฌSystems Biology
Unit & Topic Study Guides

Systems pharmacology revolutionizes drug discovery by analyzing complex biological networks. This approach considers multiple drug targets and their interactions, moving beyond the traditional "one drug, one target" model. It integrates data from various sources to predict drug effects and optimize development.

The systems approach to drug discovery combines high-throughput screening, network analysis, and predictive modeling. It aims to identify promising drug candidates more efficiently, reduce development costs, and improve success rates. This holistic view helps researchers better understand drug mechanisms and potential side effects.

Drug Discovery Approaches

Target Identification and High-Throughput Screening

  • Target identification involves identifying proteins or biological processes that play a crucial role in disease pathology
    • Utilizes genomics, proteomics, and bioinformatics to pinpoint potential drug targets
    • Considers druggability, disease relevance, and safety profiles of potential targets
  • High-throughput screening (HTS) tests large compound libraries against identified targets
    • Employs automated robotic systems to rapidly assess thousands of compounds
    • Utilizes various assay types (biochemical, cell-based) to evaluate compound activity
    • Generates hit compounds that show promising activity against the target
  • Primary screening identifies initial hits from the compound library
    • Typically uses a single concentration of each compound
    • Aims to identify compounds with desired activity above a predetermined threshold
  • Secondary screening confirms and characterizes hits from primary screening
    • Involves dose-response curves to determine potency (IC50, EC50)
    • Assesses selectivity by testing compounds against related targets

Lead Optimization and Drug Repurposing

  • Lead optimization improves properties of hit compounds to develop drug candidates
    • Enhances potency, selectivity, and pharmacokinetic properties (ADME)
    • Utilizes medicinal chemistry techniques (structure-activity relationships)
    • Involves iterative cycles of compound synthesis and testing
  • Structure-activity relationship (SAR) studies guide lead optimization
    • Identifies chemical features crucial for target interaction and biological activity
    • Helps design analogs with improved properties
  • Lead compounds undergo extensive in vitro and in vivo testing
    • Evaluates efficacy, toxicity, and pharmacokinetic properties
    • Informs selection of candidates for preclinical development
  • Drug repurposing identifies new therapeutic uses for existing drugs
    • Leverages known safety profiles to accelerate development timelines
    • Reduces costs and risks associated with drug discovery
    • Utilizes computational approaches to predict new indications (sildenafil)

Systems Pharmacology

Network Pharmacology and Polypharmacology

  • Network pharmacology studies drug effects on biological networks
    • Considers complex interactions between drugs, targets, and biological systems
    • Utilizes graph theory and network analysis to model drug-target interactions
    • Helps predict drug efficacy and side effects based on network perturbations
  • Polypharmacology explores drugs that interact with multiple targets
    • Recognizes that many drugs have effects beyond their primary intended target
    • Can lead to both beneficial (enhanced efficacy) and detrimental (side effects) outcomes
    • Aids in designing multi-target drugs for complex diseases (cancer, neurodegenerative disorders)
  • Drug-target interaction networks visualize complex pharmacological relationships
    • Nodes represent drugs and targets, edges represent interactions
    • Helps identify potential drug repurposing opportunities
    • Reveals unexpected connections between drugs and diseases

Predictive Modeling and Toxicity Prediction

  • Predictive modeling uses computational approaches to forecast drug behavior
    • Employs machine learning algorithms to analyze large datasets
    • Predicts drug-target interactions, efficacy, and potential side effects
    • Integrates diverse data types (chemical structures, genomics, clinical data)
  • Quantitative structure-activity relationship (QSAR) models relate chemical structure to biological activity
    • Uses molecular descriptors to predict compound properties and activities
    • Aids in designing new compounds with desired properties
  • Pharmacokinetic modeling predicts drug behavior in the body
    • Simulates absorption, distribution, metabolism, and excretion (ADME) processes
    • Helps optimize dosing regimens and predict drug-drug interactions
  • Toxicity prediction assesses potential adverse effects of drug candidates
    • Utilizes in silico methods to predict toxicity based on chemical structure
    • Incorporates data from in vitro assays and animal studies
    • Helps prioritize compounds for further development and guides safety testing
  • Systems toxicology examines toxicity at multiple biological levels
    • Integrates data from genomics, proteomics, and metabolomics
    • Identifies molecular mechanisms underlying toxic effects
    • Improves understanding of drug-induced organ toxicity (liver, kidney)