Computational Drug Repurposing: Approaches and Case Studies

Copyright © DrugPatentWatch. Originally published at https://www.drugpatentwatch.com/blog/

Computational drug repurposing represents one of the most promising avenues for accelerating drug development in the modern pharmaceutical landscape. By identifying new therapeutic applications for existing drugs through sophisticated computational methods, researchers can significantly reduce development timelines and costs while addressing unmet medical needs. This field has evolved dramatically in recent years, with advanced technologies enabling more precise predictions and higher success rates. The computational drug repurposing market is expanding rapidly, with approximately 30% of newly marketed drugs in the U.S. resulting from repurposing strategies, demonstrating both the clinical and commercial value of this approach4.

Understanding Drug Repurposing: Fundamentals and Significance

Drug repurposing, also known as drug repositioning, involves finding new therapeutic applications for existing drugs that have already been approved for other medical conditions. This approach offers a strategic alternative to the traditional drug discovery pipeline, which typically requires 10-15 years and billions of dollars to bring a single new compound to market. Drug repurposing circumvents many early-stage development hurdles because candidate compounds have already demonstrated safety profiles in humans, allowing researchers to focus primarily on efficacy for new indications1.

The Economics of Drug Repurposing

The financial advantages of drug repurposing are substantial and represent a key driver behind its growing adoption. According to liberal estimates, repurposing an existing drug costs approximately $300 million and takes about 6 years—a fraction of the resources required for developing novel compounds1. These economic efficiencies make repurposing an attractive strategy for pharmaceutical companies, academic institutions, and government agencies alike, particularly when addressing urgent public health challenges or rare diseases with limited commercial incentives for new drug development.

Historical Evolution from Serendipity to Systematic Approaches

Historically, drug repurposing often resulted from serendipitous observations in clinical settings. Physicians would notice unexpected beneficial effects in patients taking medications for unrelated conditions, leading to new therapeutic applications. A classic example is sildenafil (Viagra), originally developed for angina but repurposed for erectile dysfunction after researchers observed unexpected side effects during clinical trials.

However, the field has evolved dramatically from these chance discoveries to systematic, data-driven approaches. Today’s computational drug repurposing leverages vast biomedical datasets, advanced algorithms, and powerful computing resources to identify potential new indications through rigorous analysis rather than fortunate accidents1. This shift toward systematic methods has transformed drug repurposing from an occasional windfall to a reliable strategy within the pharmaceutical development toolkit.

Computational Approaches: The Driving Force Behind Modern Repurposing

Computational drug repurposing represents the cutting edge of the field, employing sophisticated algorithms and diverse data sources to predict new therapeutic applications for existing compounds. These approaches can be broadly categorized based on their primary focus: disease-centric, target-centric, or drug-centric methodologies3.

Disease-Centric Repurposing Strategies

Disease-centric approaches begin with a specific medical condition and seek to identify existing drugs that might effectively treat it. These methods typically analyze disease mechanisms, genetic signatures, and molecular pathways to find drugs that could counteract disease-specific processes. For example, researchers might examine gene expression profiles in diseased tissues to identify drugs that could reverse these pathological patterns4.

This approach is particularly valuable for addressing rare or neglected diseases where limited research funding has hindered traditional drug development. By leveraging existing clinical and genomic data, researchers can rapidly identify candidate compounds that target disease-specific molecular abnormalities without the need for extensive early-stage discovery work.

Target-Based Repurposing Approaches

Target-based approaches focus on specific biological targets—typically proteins or pathways—implicated in disease processes. These methods screen existing drug libraries to identify compounds that interact with these targets, potentially offering therapeutic benefits for conditions involving these biological mechanisms4.

The advantage of target-based repurposing lies in its ability to screen virtually any drug with a known chemical structure, creating an extensive search space for potential therapeutic candidates. However, this approach is inherently limited by our current understanding of disease biology and relevant targets. As one review notes, “target-based methods cannot identify unknown mechanisms beyond the targets already known”4.

Target-based repurposing typically involves:

  1. Identifying disease-relevant targets through genomic analysis, pathway studies, or literature reviews
  2. Screening drug libraries through computational docking studies or binding affinity predictions
  3. Prioritizing candidates based on predicted binding strength and pharmacological properties
  4. Validating these predictions through experimental models before clinical testing

Drug-Centric Repurposing Methodologies

Drug-centric approaches start with a known pharmaceutical compound and seek to identify additional diseases or conditions it might effectively treat. These methods often analyze a drug’s molecular structure, mechanism of action, or side effect profile to predict potential new therapeutic applications3.

This approach capitalizes on the principle that many drugs interact with multiple biological targets—a property known as polypharmacology. By comprehensively mapping these interactions, researchers can identify previously unrecognized therapeutic potential in established drugs, effectively expanding their clinical utility beyond their original indications.

Advanced Computational Techniques in Drug Repurposing

The technological landscape of computational drug repurposing continues to evolve rapidly, with several sophisticated methodologies gaining prominence in recent years. These advanced techniques combine diverse data types and analytical approaches to generate robust repurposing hypotheses.

Pathway-Based Drug Repurposing

Pathway-based approaches leverage our understanding of metabolic and signaling pathways to identify potential repurposing opportunities. Rather than focusing on individual drug-target interactions, these methods consider how drugs influence entire biological pathways relevant to disease processes4.

This holistic perspective offers several advantages. By analyzing pathway-level effects, researchers can identify drugs that might address complex diseases involving multiple molecular abnormalities. Additionally, pathway analysis can reveal unexpected connections between seemingly unrelated conditions, suggesting novel repurposing opportunities that more reductionist approaches might miss.

“Pathway-based drug-repurposing utilizes metabolic pathways, signaling pathways, and protein-interaction networks information to predict the similarity or connection between disease and drug. For example, using omics data processed from human patients or animals, disease-specific pathways are reconstructed to serve as new targets for repositioned drugs.”4

Target Mechanism-Based Repurposing

Target mechanism-based approaches represent one of the most sophisticated repurposing methodologies, integrating multiple data sources to uncover new mechanisms of action for existing drugs. These approaches combine signaling pathway information, treatment-related omics data, and protein interaction networks to develop comprehensive models of drug effects within specific disease contexts4.

The increasing emphasis on precision medicine has driven substantial interest in mechanism-based repurposing. By accounting for patient heterogeneity and disease complexity, these approaches can potentially identify repurposing opportunities tailored to specific patient subgroups, enhancing therapeutic efficacy while minimizing adverse effects.

Network-Based Modeling for Drug Repurposing

Network-based approaches represent biological systems as complex interconnected networks, with nodes representing entities (such as drugs, proteins, or diseases) and edges representing relationships between them. By analyzing these networks using graph theory and other mathematical techniques, researchers can identify non-obvious connections suggesting potential repurposing opportunities4.

These methods excel at integrating heterogeneous data types, creating comprehensive models that capture the complexity of biological systems more effectively than reductionist approaches. As the field advances, networks increasingly incorporate multi-omics data, clinical outcomes, and other information sources, enhancing their predictive power and clinical relevance.

Machine Learning and Artificial Intelligence in Drug Repurposing

The explosion of biomedical data and computing power has catalyzed the adoption of machine learning (ML) and artificial intelligence (AI) techniques in drug repurposing. These approaches can identify complex patterns in large, heterogeneous datasets that might elude human analysts, generating novel repurposing hypotheses with increasing accuracy5.

Supervised Learning Applications

Supervised learning algorithms train on labeled datasets to predict relationships between drugs and diseases. These methods typically use features derived from chemical structures, biological targets, gene expression profiles, and other data sources to predict whether a drug might effectively treat a specific condition.

Common supervised learning approaches in drug repurposing include:

  1. Support vector machines (SVMs) for classifying potential drug-disease matches
  2. Random forests for ranking repurposing candidates based on multiple features
  3. Gradient boosting methods for integrating diverse predictors into unified models

The effectiveness of these approaches depends heavily on the quality and comprehensiveness of training data, with performance improving as more validated drug-disease associations become available for algorithm training.

Deep Learning Innovations

Deep learning approaches, particularly neural networks with multiple hidden layers, have demonstrated remarkable success in drug repurposing applications. These methods excel at extracting meaningful features from raw data, reducing the need for extensive feature engineering while potentially uncovering previously unrecognized patterns relevant to repurposing4.

Recent applications include:

  1. Convolutional neural networks (CNNs) for analyzing molecular structures and predicting binding affinities
  2. Recurrent neural networks (RNNs) for modeling sequential biological processes relevant to drug action
  3. Graph neural networks for analyzing biological networks and predicting drug effects on complex systems

The effectiveness of these approaches was dramatically demonstrated during the COVID-19 pandemic, when deep learning methods helped identify baricitinib as a potential treatment through AI-based screening—a prediction later validated in clinical trials5.

Text Mining and Semantic Inference

Text mining approaches leverage the vast biomedical literature to identify potential repurposing opportunities. By applying natural language processing techniques to scientific publications, clinical trial reports, and other text sources, researchers can extract relationships between drugs, diseases, and biological processes that suggest repurposing opportunities4.

These methods are particularly valuable for identifying connections across traditionally siloed research domains. For example, text mining might reveal that a drug developed for neurological disorders affects molecular pathways also implicated in certain cancers, suggesting potential oncological applications that conventional research approaches might overlook.

Validation Approaches for Computational Drug Repurposing

Rigorous validation represents a critical step in the computational drug repurposing pipeline, ensuring that algorithmic predictions translate into genuine therapeutic opportunities. Validation approaches can be broadly categorized as computational or experimental, with each playing essential roles in evaluating repurposing hypotheses4.

Computational Validation Techniques

Computational validation methods assess the statistical robustness and biological plausibility of repurposing predictions without requiring laboratory experimentation. Common approaches include:

  1. ROC (Receiver Operating Characteristic) analysis to evaluate prediction accuracy, with area under the ROC curve (AUROC) serving as a primary quality metric
  2. Precision-recall curves and the corresponding area under the curve (AUPRC) to assess performance, particularly for imbalanced datasets
  3. Cross-validation using independent datasets to test the generalizability of repurposing predictions
  4. Literature-based validation comparing algorithmic predictions with previously reported associations in scientific publications4

These computational validation methods provide efficient preliminary screening of repurposing candidates, allowing researchers to prioritize the most promising predictions for more resource-intensive experimental validation.

Experimental Validation Methods

Experimental validation provides crucial empirical evidence supporting or refuting computational repurposing predictions. These approaches typically progress through a hierarchy of increasingly sophisticated models, including:

  1. In vitro binding assays to confirm predicted drug-target interactions
  2. Cell-based assays to evaluate effects on disease-relevant biological processes
  3. Animal models to assess efficacy and safety in complex biological systems
  4. Retrospective clinical analyses using electronic health records to identify real-world evidence supporting repurposing hypotheses5

The integration of computational and experimental validation creates a robust pipeline for evaluating repurposing candidates, maximizing the likelihood that computational predictions will translate into clinically meaningful therapeutic advances.

Case Studies in Computational Drug Repurposing

Examining successful applications of computational drug repurposing provides valuable insights into the field’s practical impact and methodological evolution. The following case studies highlight particularly noteworthy examples spanning different therapeutic areas and computational approaches.

COVID-19: A Global Catalyst for Repurposing

The COVID-19 pandemic created unprecedented urgency for identifying effective treatments, catalyzing extraordinary efforts in computational drug repurposing. With limited time for traditional drug development, researchers worldwide deployed sophisticated computational methods to screen existing drugs for potential efficacy against SARS-CoV-25.

A particularly notable success involved baricitinib, a JAK inhibitor originally approved for rheumatoid arthritis. This drug was identified as a potential COVID-19 treatment through AI-based screening approaches, which predicted both its antiviral activity and its ability to reduce the cytokine storm associated with severe disease. Clinical trials subsequently confirmed these predictions, leading to emergency use authorization for baricitinib in treating COVID-195.

Other repurposed drugs that demonstrated varying degrees of efficacy against COVID-19 include:

  1. Dexamethasone, a corticosteroid that significantly reduced mortality in patients requiring respiratory support
  2. Remdesivir, an antiviral originally developed for Ebola that showed modest benefits in shortening recovery time
  3. Several monoclonal antibodies repurposed from other indications to target SARS-CoV-2 specifically

The COVID-19 experience dramatically demonstrated both the potential and limitations of computational drug repurposing, with some predictions translating successfully to clinical benefit while others failed despite promising computational results.

The REMEDi4ALL Consortium: Advancing Systematic Repurposing

The REMEDi4ALL consortium represents a landmark initiative to systematize and accelerate drug repurposing through comprehensive evaluation and cataloging of computational resources. This EU-funded research initiative has developed an extensive catalog of in-silico repurposing tools, including web applications, open-access databases, and AI-based prediction methods2.

In a significant contribution published in Nature Reviews Drug Discovery, the consortium presented an expert evaluation of these resources, identifying 15 top-rated tools that are open-source, accessible, and applicable across diverse therapeutic areas. This initiative provides a valuable roadmap for researchers navigating the complex landscape of computational drug repurposing2.

The consortium demonstrated the practical application of these tools through three case studies:

  1. Repurposing for SARS-CoV-2 infection
  2. Novel approaches for pancreatic cancer treatment
  3. Therapeutic strategies for multiple sulfatase deficiency, a rare genetic disorder

These case studies illustrate how systematic application of computational tools can generate viable repurposing hypotheses across different disease categories, from pandemic infections to rare genetic conditions2.

Zidovudine: From AIDS to Cancer Applications

Zidovudine provides a compelling historical example of successful drug repurposing with computational elements. Originally developed as an antiretroviral medication for HIV/AIDS, computational analyses later identified potential anticancer properties by recognizing mechanistic similarities between viral replication and certain cancer pathways4.

This case illustrates how computational approaches can identify non-obvious connections between seemingly unrelated conditions, expanding therapeutic options for conditions with significant unmet needs. The zidovudine example also demonstrates the value of mechanism-based repurposing approaches that focus on underlying biological processes rather than surface-level disease classifications.

Tools and Resources for Computational Drug Repurposing

The expanding ecosystem of computational tools and data resources has dramatically enhanced the accessibility and effectiveness of drug repurposing research. Understanding this landscape is essential for researchers seeking to implement repurposing strategies effectively.

Open-Access Databases and Knowledge Bases

Comprehensive databases provide the foundation for computational drug repurposing by aggregating diverse data types relevant to drug-disease associations. Key resources include:

  1. DrugBank, providing detailed information on drug structures, targets, and mechanisms
  2. Gene Expression Omnibus (GEO), offering transcriptomic data relevant to disease mechanisms and drug responses
  3. The Connectivity Map (CMap), containing gene expression profiles induced by thousands of bioactive compounds
  4. PharmGKB, focusing on pharmacogenomic data linking genetic variation to drug response
  5. Electronic health record (EHR) repositories, providing real-world evidence of drug effects across diverse patient populations4

These resources collectively create a rich information ecosystem supporting diverse repurposing approaches, from target-based methods to network analyses and AI-driven predictions.

Web-Based Tools and Prediction Platforms

User-friendly web interfaces have democratized access to computational drug repurposing, allowing researchers without specialized computational expertise to leverage sophisticated analytical methods. The REMEDi4ALL consortium’s evaluation identified 15 particularly valuable open-source resources rated highly by experts in the field2.

These tools typically fall into several categories:

  1. Target prediction platforms that identify potential biological targets for known drugs
  2. Disease-drug matching tools that suggest candidate compounds for specific conditions
  3. Network analysis platforms that visualize complex relationships between drugs, targets, and diseases
  4. Structural analysis tools focusing on molecular docking and binding predictions

The increasing availability of these resources has significantly lowered barriers to entry in the field, enabling broader participation in drug repurposing research across academic, governmental, and commercial sectors.

Artificial Intelligence and Machine Learning Resources

Specialized AI and machine learning tools for drug repurposing have proliferated in recent years, ranging from open-source software libraries to comprehensive prediction platforms. These resources typically incorporate:

  1. Pre-trained models for predicting drug-target interactions
  2. Transfer learning capabilities to adapt models across different disease domains
  3. Interpretability features to explain the basis for specific repurposing predictions
  4. Integration with biological knowledge bases to enhance prediction accuracy45

The development of these AI resources reflects the broader trend toward integrating computing technologies in pharmaceutical research, with drug repurposing serving as a particularly fertile application area due to its data-intensive nature.

Challenges and Limitations in Computational Drug Repurposing

Despite remarkable progress, computational drug repurposing faces several significant challenges that can limit its effectiveness and clinical impact. Understanding these limitations is essential for realistic assessment of repurposing approaches and appropriate interpretation of computational predictions.

Data Quality and Integration Issues

The effectiveness of computational repurposing depends fundamentally on the quality, comprehensiveness, and integration of underlying data. Common challenges include:

  1. Incomplete or biased data on drug-target interactions
  2. Inconsistent annotation and standardization across different data sources
  3. Publication bias favoring positive results in the scientific literature
  4. Limited data on drug effects in diverse patient populations
  5. Difficulties integrating heterogeneous data types into unified analytical frameworks4

These data-related challenges can undermine the validity of computational predictions, potentially leading researchers down unproductive paths. Addressing these issues requires concerted efforts in data standardization, quality control, and integration methodologies.

The Translation Gap: From Prediction to Clinical Success

Perhaps the most significant challenge in computational drug repurposing is bridging the gap between algorithmic predictions and clinical utility. Many promising computational predictions fail to demonstrate efficacy in subsequent experimental or clinical validation, reflecting the complexity of biological systems and the limitations of current modeling approaches1.

Factors contributing to this translation gap include:

  1. Incomplete modeling of drug pharmacokinetics and tissue distribution
  2. Limited consideration of patient heterogeneity in computational models
  3. Regulatory challenges associated with repurposing approved drugs
  4. Intellectual property considerations affecting commercial development
  5. The inherent complexity of biological systems that exceeds current modeling capabilities

Addressing these challenges requires multidisciplinary collaboration spanning computational science, experimental biology, clinical medicine, and regulatory affairs to ensure that promising predictions navigate the complex path to clinical implementation successfully.

Future Directions in Computational Drug Repurposing

As computational capabilities continue to advance and biomedical data grows exponentially, several emerging trends are poised to reshape the landscape of drug repurposing in the coming years. These developments promise to enhance the precision, efficiency, and clinical impact of computational repurposing approaches.

Integration with Precision Medicine

The convergence of drug repurposing with precision medicine represents a particularly promising frontier. By incorporating patient-specific genetic, molecular, and clinical data into repurposing algorithms, researchers can potentially identify tailored repurposing opportunities for specific patient subgroups rather than assuming uniform efficacy across heterogeneous populations4.

This personalized approach to repurposing could significantly enhance therapeutic outcomes by:

  1. Matching repurposed drugs to patients most likely to benefit based on molecular profiles
  2. Identifying repurposing opportunities for rare disease subtypes with distinct biological mechanisms
  3. Optimizing dosing and administration protocols based on patient-specific factors
  4. Predicting and mitigating adverse effects through pharmacogenomic analysis

The integration of repurposing with precision medicine aligns with broader healthcare trends toward personalized therapeutic approaches, potentially maximizing the clinical impact of repurposed interventions.

Multimodal Data Integration and Analysis

Future repurposing approaches will likely leverage increasingly diverse data types through sophisticated integration methodologies. Beyond traditional sources like chemical structures and gene expression data, emerging approaches incorporate:

  1. Single-cell genomic data providing high-resolution insights into drug effects at the cellular level
  2. Spatial transcriptomics revealing tissue-specific drug responses with unprecedented detail
  3. Microbiome data recognizing the role of microbial communities in mediating drug effects
  4. Digital biomarkers from wearable devices capturing real-world drug responses
  5. Social media and patient-reported outcomes providing large-scale experiential data

Integrating these diverse data streams requires advanced computational methods, but offers the potential for more nuanced and accurate repurposing predictions that account for the complexity of human biology and real-world drug effects.

Quantum Computing Applications

While still emerging, quantum computing offers transformative potential for computational drug repurposing by enabling more sophisticated modeling of molecular interactions and biological systems. Potential applications include:

  1. More accurate quantum mechanical modeling of drug-target binding
  2. Optimization algorithms for multi-target drug repurposing across complex biological networks
  3. Simulation of compound combinations for polypharmacology-based repurposing strategies
  4. Pattern recognition in high-dimensional biological data exceeding classical computing capabilities

Though practical applications remain limited today, quantum computing represents a frontier technology with substantial disruptive potential for computational drug repurposing in the longer term.

Key Takeaways

Computational drug repurposing has emerged as a transformative approach to pharmaceutical development, offering significant advantages in efficiency, cost, and success probability compared to traditional drug discovery. Key insights from current research include:

  1. Drug repurposing significantly reduces development timelines and costs, with estimates suggesting approximately $300 million and 6 years for repurposing compared to billions and 10-15 years for novel drug development1.
  2. Computational approaches have evolved from serendipitous discoveries to systematic methodologies categorized as disease-centric, target-centric, drug-centric, pathway-based, and mechanism-based strategies34.
  3. Machine learning and artificial intelligence have dramatically enhanced repurposing capabilities, with deep learning models demonstrating particular promise in identifying non-obvious drug-disease associations45.
  4. The COVID-19 pandemic showcased the practical value of computational repurposing, with AI-identified candidates like baricitinib demonstrating clinical efficacy and receiving regulatory approval5.
  5. The REMEDi4ALL consortium has made significant contributions by evaluating and cataloging computational repurposing resources, identifying 15 particularly valuable open-source tools for researchers2.
  6. Validation approaches spanning computational metrics, experimental models, and clinical data analysis are essential for translating algorithmic predictions into clinically meaningful applications4.
  7. Despite remarkable progress, significant challenges remain in data quality, integration, and bridging the gap between computational predictions and clinical success4.
  8. Future directions include deeper integration with precision medicine, incorporation of multimodal data sources, and potential applications of quantum computing to enhance prediction accuracy and clinical relevance4.

As computational capabilities continue to advance and biomedical data grows increasingly rich and diverse, computational drug repurposing stands poised to accelerate therapeutic innovation across diverse disease areas, from pandemic responses to precision treatments for rare diseases.

Frequently Asked Questions

What is the difference between drug repurposing and drug repositioning?

Drug repurposing and drug repositioning are often used interchangeably, as both refer to the process of finding new therapeutic applications for existing drugs. However, some researchers make subtle distinctions, with repositioning sometimes referring specifically to marketing approved drugs for new indications, while repurposing may encompass broader applications including investigational compounds that haven’t yet received approval for any indication. In practice, both terms describe the fundamental strategy of leveraging existing pharmacological knowledge to address new therapeutic needs.

How successful is computational drug repurposing compared to traditional methods?

Computational drug repurposing has demonstrated significant advantages in efficiency and resource utilization compared to traditional drug discovery. Approximately 30% of newly marketed drugs in the U.S. result from repurposing strategies, indicating substantial success. Computational approaches accelerate the identification of promising candidates from months or years to weeks or even days. However, computational predictions still require experimental validation, and many computationally promising candidates fail to demonstrate clinical efficacy. The highest success rates typically come from integrated approaches combining computational predictions with robust experimental validation and clinical insights.

What types of diseases are most suitable for computational drug repurposing approaches?

Computational drug repurposing has demonstrated particular value for several disease categories. Rare diseases benefit significantly due to limited commercial incentives for traditional drug development. Complex disorders with well-characterized molecular signatures, such as certain cancers and autoimmune conditions, are also excellent candidates as they provide rich data for computational analysis. Emerging infectious diseases represent another prime application area, as demonstrated during the COVID-19 pandemic, when urgent public health needs demanded rapid therapeutic solutions. Generally, conditions with substantial unmet medical needs and reasonably well-understood molecular mechanisms offer the most promising opportunities for computational repurposing.

How can researchers access computational drug repurposing tools if they lack specialized programming skills?

Researchers without specialized programming expertise can access numerous user-friendly web-based platforms for computational drug repurposing. The REMEDi4ALL consortium has identified 15 top-rated open-source resources specifically designed for accessibility. These include tools with graphical interfaces that guide users through the repurposing process from data input to result interpretation. Additionally, many academic institutions offer collaborative opportunities with computational specialists, and commercial services provide repurposing analyses for researchers lacking in-house capabilities. The democratization of these tools has significantly expanded participation in drug repurposing research beyond specialized computational biology groups.

What regulatory challenges exist for bringing repurposed drugs to market?

Repurposed drugs face several distinct regulatory considerations despite their established safety profiles. Patent protection represents a significant challenge, as many repurposing candidates have expired patents limiting commercial incentives. Regulatory agencies require substantial evidence supporting new indications, though requirements are typically less extensive than for novel compounds. “Label expansion” pathways exist in many jurisdictions to facilitate approval for new indications of existing drugs. Specialized programs like the FDA’s 505(b)(2) pathway provide streamlined routes for repurposed drugs, allowing sponsors to reference existing safety data while demonstrating efficacy for new indications. Despite these provisions, navigating intellectual property and regulatory requirements remains a significant hurdle in translating computational repurposing discoveries to approved therapies.

  1. https://pmc.ncbi.nlm.nih.gov/articles/PMC10785886/
  2. https://remedi4all.org/new-remedi4all-resource-for-navigating-the-complex-landscape-of-computational-drug-repurposing/
  3. https://pubmed.ncbi.nlm.nih.gov/37021703/
  4. https://pmc.ncbi.nlm.nih.gov/articles/PMC6989243/
  5. https://pmc.ncbi.nlm.nih.gov/articles/PMC7996547/

Citations:

  1. https://pmc.ncbi.nlm.nih.gov/articles/PMC10785886/
  2. https://remedi4all.org/new-remedi4all-resource-for-navigating-the-complex-landscape-of-computational-drug-repurposing/
  3. https://pubmed.ncbi.nlm.nih.gov/37021703/
  4. https://pmc.ncbi.nlm.nih.gov/articles/PMC6989243/
  5. https://pmc.ncbi.nlm.nih.gov/articles/PMC7996547/
  6. https://tcpharm.org/pdf/10.12793/tcp.2019.27.2.59
  7. https://pubs.rsc.org/en/content/articlelanding/2021/ra/d1ra05320e
  8. https://www.recursion.com/news/rebranding-repurposing-a-polypharmacological-perspective

Make Better Decisions with DrugPatentWatch

» Start Your Free Trial Today «

Copyright © DrugPatentWatch. Originally published at
DrugPatentWatch - Transform Data into Market Domination