Drug repurposing is a game-changing approach in computational molecular biology. It uses existing drugs to treat new diseases, saving time and money compared to traditional drug discovery. By analyzing molecular data and using advanced computational methods, researchers can predict new uses for approved medications.

This strategy leverages the wealth of information we already have about drugs. It combines data mining, machine learning, and network analysis to find hidden connections between drugs and diseases. The process is faster, cheaper, and often more successful than developing new drugs from scratch.

Fundamentals of drug repurposing

  • Drug repurposing applies computational molecular biology techniques to identify new therapeutic uses for existing drugs
  • Leverages vast amounts of molecular and clinical data to predict potential drug-target interactions across different diseases
  • Integrates various bioinformatics approaches including data mining, machine learning, and network analysis to accelerate drug discovery process

Definition and concept

Top images from around the web for Definition and concept
Top images from around the web for Definition and concept
  • Systematic process of identifying new therapeutic applications for existing drugs, compounds, or biologics
  • Exploits the polypharmacological nature of drugs to target multiple biological pathways
  • Utilizes computational methods to predict drug-target interactions based on structural similarities and biological activities
  • Aims to reduce time and cost associated with traditional drug development pipelines

Advantages vs traditional drug discovery

  • Significantly reduces development timeline from 10-17 years to 3-12 years
  • Lowers costs by leveraging existing safety and pharmacokinetic data
  • Increases success rates due to known toxicology profiles of repurposed drugs
  • Provides faster route to and market approval
  • Offers potential for treating rare diseases with limited therapeutic options

Historical examples of success

  • Sildenafil repurposed from angina treatment to erectile dysfunction medication (Viagra)
  • Thalidomide repurposed from morning sickness treatment to multiple myeloma therapy
  • Metformin repurposed from type 2 diabetes treatment to potential anti-cancer agent
  • Aspirin repurposed from pain relief to prevention of cardiovascular events
  • Minoxidil repurposed from hypertension treatment to hair loss remedy (Rogaine)

Computational approaches in repurposing

  • Computational methods form the backbone of modern drug repurposing strategies in molecular biology
  • Utilize large-scale biological and chemical databases to identify potential drug-target interactions
  • Employ various algorithms and models to predict drug efficacy and safety profiles across different diseases

Data mining techniques

  • Text mining extracts relevant information from scientific literature and clinical reports
  • Semantic web technologies integrate heterogeneous data sources for comprehensive analysis
  • Association rule mining identifies patterns and relationships between drugs, targets, and diseases
  • Frequent itemset mining discovers co-occurring drug-disease associations in large datasets
  • Clustering algorithms group similar drugs or targets based on molecular features or biological activities

Machine learning algorithms

  • Supervised learning models predict drug-target interactions using labeled training data
  • Unsupervised learning algorithms identify hidden patterns in drug and disease datasets
  • Support Vector Machines (SVM) classify drugs based on their molecular descriptors
  • Random Forests predict drug efficacy by analyzing multiple decision trees
  • Gradient Boosting algorithms improve prediction accuracy through iterative learning
  • Deep Neural Networks process complex biological data to identify potential drug candidates

Network-based methods

  • Drug-target interaction networks represent relationships between compounds and their biological targets
  • Disease-gene networks link genetic factors to specific pathological conditions
  • Protein-protein interaction networks model cellular processes and potential drug effects
  • Network topology analysis identifies key nodes and hubs for drug targeting
  • Graph-based algorithms detect communities and modules within biological networks
  • Network propagation methods predict novel drug-disease associations based on known interactions

Target identification strategies

  • forms a crucial step in drug repurposing within computational molecular biology
  • Combines structural biology, chemoinformatics, and systems biology approaches
  • Aims to predict and validate potential drug targets for repurposing candidates

Molecular docking simulations

  • Computational method predicts binding affinity between drugs and protein targets
  • Utilizes 3D structures of proteins and small molecules to simulate interactions
  • Scoring functions evaluate the strength and stability of predicted drug-target complexes
  • Flexible docking accounts for conformational changes in both ligand and receptor
  • Ensemble docking considers multiple protein conformations to improve accuracy
  • Virtual screening uses docking to rapidly assess large libraries of compounds against target proteins

Ligand-based screening approaches

  • Relies on the principle that similar molecules have similar biological activities
  • Quantitative Structure-Activity Relationship (QSAR) models correlate molecular properties with biological activity
  • Pharmacophore modeling identifies essential structural features for drug-target interactions
  • Similarity searching compares chemical fingerprints of known active compounds to identify potential repurposing candidates
  • Machine learning algorithms predict biological activities based on molecular descriptors
  • 3D shape-based methods align molecules to identify compounds with similar spatial arrangements

Phenotypic screening approaches

  • Focuses on observable cellular or organismal responses to drug treatments
  • High-content screening uses automated microscopy to analyze cellular phenotypes
  • Gene expression profiling identifies drugs that reverse disease-associated gene signatures
  • Pathway-based screening targets specific biological pathways implicated in diseases
  • Organoid and spheroid models provide 3D cellular environments for more physiologically relevant screening
  • In vivo phenotypic screening uses animal models to assess drug effects on complex biological systems

Omics data integration

  • Omics data integration plays a crucial role in drug repurposing within computational molecular biology
  • Combines multiple layers of biological information to provide a comprehensive view of drug-disease interactions
  • Enables identification of novel drug targets and repurposing opportunities across different biological scales

Transcriptomics in drug repurposing

  • Analyzes gene expression changes in response to drug treatments or disease states
  • RNA-seq technology provides high-resolution transcriptome profiles for drug-induced effects
  • Differential expression analysis identifies genes and pathways modulated by drugs
  • Gene Set Enrichment Analysis (GSEA) reveals biological processes affected by drug treatments
  • Connectivity Map (CMap) compares drug-induced gene expression signatures to identify potential repurposing candidates
  • Single-cell RNA-seq enables cell type-specific analysis of drug responses in heterogeneous tissues

Proteomics and metabolomics applications

  • Proteomics studies drug-induced changes in protein expression and post-translational modifications
  • Mass spectrometry-based techniques quantify protein levels and identify drug targets
  • Protein-protein interaction mapping reveals drug effects on cellular signaling networks
  • Metabolomics analyzes changes in small molecule metabolites in response to drugs
  • Metabolic flux analysis tracks alterations in biochemical pathways induced by drug treatments
  • Integration of proteomics and metabolomics data provides insights into drug mechanisms of action

Multi-omics data analysis

  • Combines data from multiple omics platforms to provide a holistic view of drug effects
  • Network-based integration methods identify functional relationships across different omics layers
  • Machine learning algorithms predict drug responses based on multi-omics profiles
  • Pathway-based integration approaches reveal drug-induced perturbations in biological systems
  • Bayesian methods integrate diverse omics data types to infer causal relationships
  • Multi-omics clustering identifies patient subgroups for personalized drug repurposing strategies

Pharmacological databases

  • Pharmacological databases serve as essential resources for drug repurposing in computational molecular biology
  • Provide comprehensive information on drug structures, targets, and biological activities
  • Enable large-scale computational analyses and predictions for repurposing candidates

DrugBank and PubChem

  • offers detailed information on drug structures, targets, and mechanisms of action
  • Includes data on approved drugs, experimental compounds, and nutraceuticals
  • PubChem provides chemical structures, biological activities, and bioassay results for millions of compounds
  • Enables structure-based searching and comparison of drug-like molecules
  • Both databases support programmatic access through APIs for computational analyses
  • Integration of DrugBank and PubChem data enhances drug repurposing predictions

Connectivity Map (CMap)

  • Large-scale gene expression database of drug-induced transcriptional responses
  • Contains gene expression profiles for thousands of small molecules across multiple cell lines
  • Enables identification of drugs with similar or opposite gene expression signatures
  • Query system allows comparison of disease gene signatures with drug-induced profiles
  • Supports discovery of potential for specific diseases
  • Integrates with other omics data types for comprehensive drug repurposing analyses

LINCS project

  • Library of Integrated Network-based Cellular Signatures (LINCS) expands on CMap concept
  • Provides standardized gene expression and cellular response data for diverse perturbations
  • Includes data on small molecules, genetic perturbations, and disease states
  • L1000 technology enables cost-effective profiling of large compound libraries
  • Offers tools for data analysis and visualization through web-based platforms
  • Supports development of machine learning models for drug repurposing and target prediction

Repurposing for rare diseases

  • Drug repurposing for rare diseases addresses unmet medical needs in computational molecular biology
  • Leverages existing drug information and biological data to identify potential treatments for orphan diseases
  • Combines computational approaches with experimental validation to accelerate rare disease drug discovery

Challenges in rare disease drug development

  • Limited patient populations hinder traditional clinical trial designs
  • Lack of understanding of disease mechanisms complicates target identification
  • High costs and low return on investment discourage pharmaceutical industry involvement
  • Genetic heterogeneity within rare disease populations complicates treatment strategies
  • Limited availability of disease-specific cellular and animal models for drug screening
  • Regulatory hurdles in obtaining orphan drug designation and market approval

Computational tools for rare diseases

  • Network-based approaches identify disease-gene associations and potential drug targets
  • Machine learning algorithms predict drug efficacy based on molecular features of rare diseases
  • Virtual patient cohorts simulate clinical trials for rare disease populations
  • Systems biology models integrate multi-omics data to understand rare disease mechanisms
  • Natural language processing extracts relevant information from scientific literature and clinical reports
  • Pathway analysis tools identify druggable targets within rare disease-associated biological processes

Case studies of successful repurposing

  • Everolimus repurposed from immunosuppressant to treatment for tuberous sclerosis complex
  • Sildenafil repurposed from erectile dysfunction medication to treatment for pulmonary arterial hypertension
  • Thalidomide repurposed for treatment of erythema nodosum leprosum in leprosy patients
  • Imatinib repurposed from chronic myeloid leukemia treatment to therapy for systemic mastocytosis
  • N-acetylcysteine repurposed from mucolytic agent to treatment for N-acetylglutamate synthase deficiency

Artificial intelligence in repurposing

  • Artificial intelligence revolutionizes drug repurposing approaches in computational molecular biology
  • Enables processing and analysis of vast amounts of biological and chemical data
  • Enhances prediction accuracy and accelerates identification of repurposing candidates

Deep learning models

  • Convolutional Neural Networks (CNNs) analyze molecular structures and predict drug-target interactions
  • Recurrent Neural Networks (RNNs) process sequential data such as protein sequences or gene expression time series
  • Graph Neural Networks (GNNs) model complex biological networks and predict drug-disease associations
  • Variational Autoencoders generate novel molecular structures for drug repurposing
  • Transformer models process large-scale biomedical text data to extract drug-disease relationships
  • Reinforcement learning optimizes drug combinations for repurposing strategies

Natural language processing

  • Extracts relevant information from scientific literature, clinical reports, and electronic health records
  • Named Entity Recognition identifies drugs, diseases, and biological entities in unstructured text
  • Relation extraction algorithms detect associations between drugs, targets, and diseases
  • Topic modeling uncovers hidden themes and trends in biomedical literature
  • Sentiment analysis assesses drug efficacy and safety based on patient-reported outcomes
  • Question-answering systems provide targeted information retrieval for drug repurposing queries

AI-driven target prediction

  • Predicts potential drug targets based on molecular structures and biological activities
  • Integrates multi-omics data to identify novel drug-target associations
  • Utilizes transfer learning to leverage knowledge from well-studied diseases for rare disease target prediction
  • Employs attention mechanisms to focus on relevant features for target prediction
  • Incorporates uncertainty quantification to assess confidence in predicted drug-target interactions
  • Combines AI predictions with experimental validation to accelerate target identification process

Clinical trials and regulatory aspects

  • Clinical trials and regulatory considerations play crucial roles in drug repurposing within computational molecular biology
  • Adapt traditional drug development processes to accommodate repurposed candidates
  • Integrate computational predictions with clinical evidence to support regulatory approval

Repurposing-specific trial designs

  • Adaptive trial designs allow flexibility in patient selection and treatment allocation
  • Basket trials evaluate a single drug across multiple diseases with shared molecular targets
  • Umbrella trials test multiple drugs in a single disease based on molecular profiling
  • N-of-1 trials assess drug efficacy in individual patients with rare diseases
  • Platform trials enable evaluation of multiple drugs against a common control group
  • Real-world evidence studies leverage electronic health records to support repurposing claims

Regulatory pathways for repurposed drugs

  • 505(b)(2) pathway in the US allows approval based on existing safety and efficacy data
  • Orphan drug designation provides incentives for repurposing drugs for rare diseases
  • Breakthrough Therapy Designation accelerates development of repurposed drugs for serious conditions
  • European Medicines Agency (EMA) offers scientific advice for repurposing strategies
  • Priority Review vouchers incentivize development of repurposed drugs for neglected tropical diseases
  • Accelerated approval pathways based on surrogate endpoints support faster market access

Intellectual property considerations

  • Patent protection strategies for new uses of existing drugs
  • Method-of-use patents cover novel therapeutic applications of known compounds
  • Formulation patents protect new delivery systems or dosage forms for repurposed drugs
  • Combination patents cover synergistic effects of repurposed drugs with other agents
  • Data exclusivity periods provide market protection for new indications of approved drugs
  • Licensing agreements between original drug developers and repurposing entities
  • Open-source drug discovery initiatives for repurposing off-patent compounds

Limitations and challenges

  • Drug repurposing faces several limitations and challenges within computational molecular biology
  • Addresses complexities in predicting drug efficacy and safety across different diseases
  • Considers ethical implications of repurposing strategies in drug development

Off-target effects prediction

  • Computational methods predict potential side effects based on structural similarities
  • Machine learning models integrate multi-omics data to identify off-target interactions
  • approaches map drug effects across multiple biological pathways
  • Molecular dynamics simulations assess drug binding to non-target proteins
  • Toxicogenomics data analysis predicts adverse effects based on gene expression changes
  • Integration of pharmacovigilance data improves off-target effect predictions

Translating in silico results

  • Experimental validation required to confirm computational predictions
  • Cell-based assays assess drug efficacy and toxicity in relevant disease models
  • Animal studies evaluate pharmacokinetics and efficacy of repurposed candidates
  • Biomarker development supports translation of repurposing predictions to clinical applications
  • Challenges in replicating in silico results in complex biological systems
  • Integration of computational and experimental approaches to improve predictive accuracy

Ethical considerations in repurposing

  • Balancing intellectual property rights with public health benefits
  • Ensuring equitable access to repurposed drugs, especially for rare diseases
  • Addressing potential conflicts of interest in academic-industry collaborations
  • Considering implications of off-label drug use based on repurposing predictions
  • Ethical concerns in using AI-generated predictions for clinical decision-making
  • Ensuring transparency and reproducibility in computational repurposing methods

Future directions

  • Future directions in drug repurposing within computational molecular biology focus on advancing predictive capabilities
  • Integrate emerging technologies and data types to enhance repurposing strategies
  • Aim to address unmet medical needs and improve patient outcomes through innovative approaches

Personalized medicine applications

  • Integration of individual patient genomic data for tailored drug repurposing
  • Machine learning models predict drug responses based on patient-specific molecular profiles
  • Digital twin technologies simulate drug effects in virtual patient models
  • Pharmacogenomics-guided repurposing strategies account for genetic variations in drug response
  • Single-cell analysis enables precision repurposing for heterogeneous disease populations
  • Wearable device data integration for real-time monitoring of repurposed drug effects

Integration with drug delivery systems

  • Nanoparticle-based delivery systems enhance efficacy of repurposed drugs
  • Computational modeling of drug-nanoparticle interactions for optimized delivery
  • Targeted delivery strategies based on disease-specific molecular markers
  • 3D-printed drug delivery devices for personalized dosing of repurposed drugs
  • Stimuli-responsive materials for controlled release of repurposed compounds
  • In silico prediction of drug-excipient compatibility for novel formulations

Emerging computational techniques

  • Quantum computing algorithms for enhanced molecular simulations and docking
  • Federated learning enables collaborative drug repurposing while preserving data privacy
  • Explainable AI models provide interpretable predictions for drug repurposing
  • Multi-scale modeling integrates molecular, cellular, and organ-level data for comprehensive repurposing strategies
  • Generative models design novel molecules based on repurposed drug scaffolds
  • Edge computing facilitates real-time analysis of repurposing data in clinical settings

Key Terms to Review (16)

Agonist: An agonist is a molecule that binds to a receptor and activates it, mimicking the action of a naturally occurring substance. This interaction can lead to a biological response, such as activating a signaling pathway or inducing physiological changes. Agonists are crucial in understanding protein-ligand interactions and play a significant role in the development and repurposing of drugs, as they can enhance or initiate the desired effects in therapeutic applications.
Antagonist: An antagonist is a molecule that binds to a receptor and blocks or dampens the biological response triggered by an agonist, which is another molecule that activates the receptor. Antagonists play a crucial role in regulating various physiological processes by inhibiting the effects of naturally occurring ligands or drugs. This modulation of receptor activity can lead to therapeutic benefits in treating various diseases and conditions.
Bioactivity profiling: Bioactivity profiling is the systematic assessment of the biological effects of compounds, often used to understand their potential therapeutic applications. This process involves screening compounds to evaluate their efficacy, potency, and safety against specific biological targets or diseases. It plays a crucial role in drug discovery and development, especially in the context of finding new uses for existing drugs through repurposing efforts.
Clinical trials: Clinical trials are research studies conducted with human participants to evaluate the safety and effectiveness of new medical treatments, drugs, or devices. These trials are essential for determining how well a treatment works compared to existing therapies or a placebo, and they follow a rigorous protocol that includes several phases to ensure reliability and validity of the results.
Computational screening: Computational screening is a method used to identify potential drug candidates or bioactive compounds by leveraging computational tools and algorithms to analyze biological data. This approach allows researchers to predict how different compounds may interact with biological targets, speeding up the drug discovery process and enabling the repurposing of existing drugs for new therapeutic applications.
CTD (Comparative Toxicogenomics Database): CTD is a publicly available database that compiles information about the interactions between chemicals, genes, and diseases. It provides researchers with valuable data for understanding how environmental exposures can influence health outcomes and disease mechanisms. By integrating diverse data sources, CTD aids in the identification of potential drug repurposing candidates based on toxicological evidence and genetic information.
DrugBank: DrugBank is a comprehensive online database that provides detailed information on drugs and drug targets. It serves as a valuable resource for researchers and healthcare professionals, integrating data on drug chemical structures, mechanisms of action, pharmacological effects, and interactions. This database plays a crucial role in drug repurposing, where existing medications are investigated for new therapeutic uses beyond their original purpose.
High-throughput screening: High-throughput screening is a method used in drug discovery and biomedical research that allows researchers to quickly test thousands to millions of compounds for their biological activity. This process involves automated technology and robotics to efficiently analyze large datasets, identifying potential candidates for drug development, including existing drugs that can be repurposed for new therapeutic uses.
Network Pharmacology: Network pharmacology is an innovative approach that combines systems biology and pharmacology to understand how drugs interact with biological networks and pathways. It focuses on the interconnectedness of drugs, targets, and disease networks, allowing researchers to identify potential therapeutic effects and repurpose existing drugs for new indications.
Off-label use: Off-label use refers to the practice of prescribing pharmaceuticals for an unapproved indication, age group, dosage, or route of administration that is not specified in the approved labeling by regulatory authorities. This practice is common in medicine as it allows healthcare providers to use their clinical judgment to treat patients based on emerging evidence and personal experience, particularly when no approved treatments are available for specific conditions.
Polypatharmacology: Polypatharmacology refers to the design and development of drugs that can interact with multiple targets in the body, rather than focusing on a single target. This approach recognizes that many diseases are complex and involve various biological pathways, making it crucial to consider multiple interactions to improve therapeutic outcomes.
Sildenafil for erectile dysfunction: Sildenafil is a medication primarily used to treat erectile dysfunction (ED) by enhancing blood flow to the penis, facilitating an erection when sexual stimulation occurs. Initially developed to treat pulmonary hypertension, it was later repurposed for ED due to its vasodilatory effects, demonstrating how drugs can be effectively redirected for new therapeutic uses beyond their original intentions.
Structure-based drug design: Structure-based drug design is a method used in drug discovery that relies on the three-dimensional structure of biological molecules to identify and develop new medications. This approach involves analyzing the structure of target proteins to understand how potential drug compounds can interact with them, leading to optimized therapeutic agents. It connects molecular biology with computational techniques, which include homology modeling and drug repurposing strategies.
Target Identification: Target identification is the process of determining the biological molecules, usually proteins or genes, that are associated with a disease or condition and can be targeted by therapeutic agents. This step is crucial in drug discovery as it helps researchers pinpoint which molecular targets are most likely to yield effective treatments, thus connecting closely to pharmacophore modeling and drug repurposing strategies.
Thalidomide for multiple myeloma: Thalidomide is a drug that was originally developed as a sedative in the late 1950s, but it gained prominence in the treatment of multiple myeloma, a type of blood cancer. The repurposing of thalidomide has highlighted its ability to modulate the immune system and inhibit the growth of tumor cells, making it an important therapeutic option for patients with this condition.
Therapeutic candidates: Therapeutic candidates are potential drugs or compounds that have shown promise in treating specific diseases or conditions and are being considered for further development and testing. These candidates can originate from various sources, including existing drugs that may be repurposed for new indications or novel compounds discovered through research. The identification and validation of therapeutic candidates are crucial steps in drug development, as they set the stage for clinical trials and ultimately determine the effectiveness and safety of treatments.
© 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.