Global Genomics Initiatives

Drug Repurposing: A Genomic Approach

December 18, 2024 Off By admin
Shares

Genomic Drug Repurposing: Revolutionizing the Search for New Treatments

In the world of modern medicine, the process of developing new drugs has been long, costly, and often riddled with failures. Traditional drug discovery can take years, involving extensive clinical trials and billions of dollars in investment, only to have a product eventually rejected due to unforeseen adverse effects. However, a transformative approach known as drug repurposing is emerging as a faster, more cost-effective alternative. This strategy involves finding new therapeutic uses for already approved drugs, leveraging the wealth of genomic data now available to researchers.

Genomics plays a critical role in drug repurposing by providing the tools to connect the dots between drugs, diseases, and their underlying molecular mechanisms. In this post, we’ll explore how genomic data is reshaping the landscape of drug repurposing, discussing the various strategies being employed, their advantages and challenges, and the future directions in this rapidly evolving field.

The Promise of Drug Repurposing

Drug repurposing, also known as drug repositioning, offers a revolutionary path for quickly addressing unmet medical needs. By focusing on existing drugs with established safety profiles, researchers can avoid the lengthy and costly phases of drug development typically associated with new compounds. As the review published in Briefings in Bioinformatics (2024) highlights, genomic tools are accelerating this process, enabling scientists to explore new uses for drugs that are already on the market.

In recent years, the rise of large biobanks—collections of genetic, health, and clinical data—has made it possible to mine these resources for new therapeutic insights. When combined with electronic health records (EHRs) and other biological databases, researchers can now analyze vast amounts of data to uncover novel drug-disease relationships. This shift in approach opens the door to faster identification of drugs that could be repurposed for a range of conditions, from cardiovascular disease to cancer.

Three Key Strategies in Genomic Drug Repurposing

There are three primary methodologies that have been employed in drug repurposing using genomic data: Mendelian randomization (MR), multi-omic-based approaches, and network-based strategies. Let’s break down each of these approaches:

1. Mendelian Randomization (MR)

Mendelian randomization is a technique that uses genetic variants as proxies to assess causal relationships between drugs and diseases. By examining how genetic variations, which mimic the effects of a drug, impact disease outcomes, researchers can determine whether a drug might be useful for treating a different condition. This method is powerful because it reduces confounding factors and provides stronger evidence of causality compared to traditional observational studies.

For example, a study used genetic variants associated with anti-hypertensive drugs to examine their potential role in diabetes prevention, finding that genetically proxied ACE inhibition showed a protective effect against diabetes. While MR is powerful, its applicability is limited to conditions where there are well-defined genetic instruments, and it may not be suitable for diseases with weak genetic determinants.

2. Multi-Omic-Based Approaches

Multi-omic strategies involve the integration of various omics data (e.g., genomics, transcriptomics, proteomics, and metabolomics) to gain a holistic understanding of disease mechanisms and uncover potential drug targets. By combining different layers of biological data, these approaches provide comprehensive insights into how diseases develop at the molecular level and allow for the identification of novel treatment opportunities.

For instance, multi-omic data can identify potential combination therapies, where different drugs act on multiple disease pathways simultaneously. However, these methods face challenges in data integration, as different omics platforms often produce data with varying quality, making interpretation complex. Some of the most widely used databases for these studies include the UK Biobank, 23andMe, and the Human Protein Atlas.

3. Network-Based Strategies

Network-based strategies focus on leveraging biological networks to explore the complex relationships between genes, proteins, drugs, and diseases. These strategies use tools like protein-protein interaction (PPI) networks and drug-gene interaction databases to reveal new connections that might not be apparent through individual molecular analysis. By understanding the interplay between various biological components, researchers can identify multi-target drugs or synergistic drug combinations.

Network-based strategies have the advantage of considering the broader biological context, but they also face challenges related to data quality and network complexity. For example, a study using the STRING database identified potential drug repurposing candidates for cancer treatment by analyzing protein interaction networks. However, the lack of specificity in some network-based approaches could lead to off-target effects.

Real-World Applications of Drug Repurposing

The application of genomic data to drug repurposing is already yielding promising results. Below are a few key examples of how these strategies are being used in various therapeutic areas:

Cardiovascular Disease

Mendelian randomization has been applied to investigate the potential of existing lipid-lowering and anti-hypertensive drugs in preventing cardiovascular events such as stroke and coronary artery disease. By identifying genetic variants associated with cardiovascular risk factors, researchers can assess the repurposing potential of these drugs for broader indications.

Neurodegenerative Diseases

In the search for treatments for Alzheimer’s and Parkinson’s diseases, genomic data is being used to explore drug repurposing opportunities. Researchers are applying machine learning techniques to predict causal links between genetic loci and disease outcomes, potentially uncovering novel therapies for these challenging conditions.

Cancer

Studies have explored the repurposing of drugs originally developed for conditions like diabetes and hypertension in the treatment of various cancers. For example, anti-diabetic drugs have been examined for their potential to reduce the risk of cancer, while anti-hypertensive drugs have shown promise in targeting specific cancer types, such as glioblastoma.

Challenges and Future Directions

Despite the potential of genomic-driven drug repurposing, there are still several challenges that need to be addressed:

  1. Data Integration: Combining data from diverse sources—genomic databases, health records, and experimental studies—remains a significant challenge. Researchers need to develop more sophisticated computational tools to handle the complexity of multi-source data.
  2. Causality and Specificity: While Mendelian randomization provides strong evidence for causality, it is not always applicable, especially for conditions with weak genetic determinants. Similarly, network-based approaches may lack the specificity required to avoid off-target effects.
  3. Generalizability: Most genomic studies have been conducted on specific populations, often of European descent. This raises concerns about the applicability of findings to more diverse ethnic groups, emphasizing the need for greater representation in genomic research.

Conclusion

The field of drug repurposing is on the brink of a revolution, powered by advances in genomic research. By harnessing the power of genomic data and integrating it with innovative analytical strategies, researchers are unlocking new therapeutic uses for existing drugs, which could significantly reduce the time and costs associated with drug development.

While challenges remain, the potential for drug repurposing to accelerate the discovery of treatments for a wide range of diseases is enormous. As research in this field progresses, it holds the promise of transforming how we approach drug development, making it more efficient, cost-effective, and accessible to those in need.

FAQ: Drug Repurposing Using Genomic Data

1. What is drug repurposing, and why is it gaining attention in pharmaceutical research?

Drug repurposing, also known as drug repositioning, is the practice of finding new uses for existing, approved medications. This approach is gaining attention because it offers several advantages over traditional drug development, such as reduced time and costs since these drugs already have established safety profiles. This strategy can accelerate the availability of new treatments, particularly for unmet medical needs.

2. What are the primary strategies employed in drug repurposing using human genomic data?

Three main strategies are commonly used: Mendelian Randomization (MR), multi-omic-based, and network-based studies. MR utilizes genetic variants as proxies for drug targets to assess causal relationships between the target and a disease outcome. Multi-omic-based approaches integrate various omics data (e.g., genomics, transcriptomics, proteomics) to explore disease mechanisms and identify potential drug targets. Network-based studies analyze complex biological networks involving genes, proteins, diseases, and drugs to uncover relationships and identify drug repurposing candidates.

3. What is Mendelian Randomization (MR), and how does it apply to drug repurposing?

Mendelian randomization (MR) uses genetic variants as instrumental variables to assess the causal effects of a drug’s target on disease outcomes. By examining the genetic variants associated with the target of a particular drug, researchers can infer whether modulating that target will have a beneficial or adverse effect on the disease. MR helps in prioritizing drug candidates by providing evidence of causality, reducing bias and enabling cost-effective repurposing. However, it relies on valid genetic instruments and may have limited applicability for drugs with complex or weak genetic determinants.

4. How do multi-omic-based approaches contribute to drug repurposing?

Multi-omic-based strategies integrate diverse omics data, such as genomics, transcriptomics, proteomics, and metabolomics, to gain comprehensive insights into disease mechanisms and identify potential drug targets. This approach can reveal novel pathways and drug targets that might be overlooked with single omics data, as well as help identify synergistic drug combinations by evaluating how different drugs affect multiple molecular pathways. Furthermore, multi-omic approaches enable personalized medicine by considering individual genetic and expression profiles, allowing the development of tailored treatment strategies. However, data quality, availability, and the complexity of integrating data from different sources can be challenges.

5. How do network-based strategies aid in drug repurposing?

Network-based strategies utilize complex biological networks to identify relationships between drugs, diseases, and molecular targets. These networks integrate information from different data sources, including gene-gene interactions, protein-protein interactions, and drug-target interactions. By analyzing these networks, researchers can identify potential repurposing candidates and guide precision medicine approaches, including synergistic drug combinations that target multiple network components. Though powerful in identifying mechanism of action, this approach might lack specificity if a drug targets multiple nodes in a network.

6. What are some of the main data sources used in genomic-based drug repurposing studies?

Various data sources are utilized, including:

  • GWAS Databases: (e.g., IEU Open GWAS) provide summary statistics from various GWASs for MR studies.
  • Biobanks: (e.g., UK Biobank, China Kadoorie Biobank) offer extensive genomic, transcriptomic, proteomic, and metabolomic data along with clinical information.
  • Omics Databases: (e.g., GTEx, Human Protein Atlas) provide detailed information on gene expression and protein abundance across different tissues.
  • Drug Databases: (e.g., DGIdb, DrugBank) contain information about drug-target interactions, chemical structures, and mechanisms of action.
  • Clinical Trial Databases: (e.g., ClinicalTrials.gov) help explore the efficacy, safety and off-label use of drugs across different patient populations.
  • Interaction Databases (e.g., STRING) provides information on protein-protein interactions and other biological pathways.

7. What are some of the limitations and challenges associated with drug repurposing using genomic data?

Several limitations and challenges exist: – Data Integration: Integrating diverse omics datasets from various platforms can be complex and challenging. – Data Quality: The quality of omics data may vary, leading to unreliable predictions. – Genetic Instruments: MR depends on identifying valid genetic instruments strongly associated with the effect of drug treatment, which may not always be available. – Population Bias: Most genomic data is derived from specific populations, potentially limiting the applicability of findings across different ethnic groups. – Biological Complexity: Biological systems are complex, and incomplete understanding of mechanisms might hinder the effectiveness of repurposing strategies. – Specificity: Network-based approaches may lack specificity and lead to off-target effects. – Clinical Validation: Drug candidates identified through computational methods must be validated through further laboratory experiments and clinical trials.

8. What are some future directions for advancing drug repurposing research?

Future directions for drug repurposing include:

  • Integrating diverse data sources to gain a more comprehensive view of disease mechanisms.
  • Developing advanced algorithms using artificial intelligence to enhance drug repurposing pipelines and better predict drug-disease associations.
  • Exploring drug-drug interactions and synergistic effects to design combination therapies.
  • Establishing collaborative networks and data sharing platforms to accelerate drug discovery.
  • Performing thorough biological experiments and randomized clinical trials to validate repurposed drug effects, efficacy, and safety across diverse populations.

Glossary of Key Terms

  • Drug Repurposing: The process of identifying new therapeutic uses for existing drugs that are already approved or in late-stage development.
  • Mendelian Randomization (MR): A method that uses genetic variants as instrumental variables to assess causal relationships between a modifiable exposure (e.g., a drug target) and an outcome.
  • Multi-omics: The integration and analysis of data from multiple biological “omics” fields, such as genomics, transcriptomics, proteomics, and metabolomics.
  • Network-based Approach: A strategy that uses biological networks (e.g., protein-protein interaction networks, gene regulatory networks) to integrate data and discover relationships between drugs, diseases, and molecular targets.
  • Single-nucleotide polymorphism (SNP): A variation in a single nucleotide in the DNA sequence of a genome. SNPs are used as markers in genetic studies.
  • Genome-Wide Association Study (GWAS): A research approach that involves scanning markers across the genomes of many people to find genetic variations associated with a particular trait or disease.
  • Transcriptome-wide Association Study (TWAS): A study that analyzes the associations between gene expression levels (transcriptomes) and disease phenotypes to identify causal genes.
  • Proteome-wide Association Study (PWAS): A study that examines the associations between protein levels (proteomes) and traits or diseases to identify causal proteins.
  • Metabolome-wide Association Study (MWAS): A study that investigates the associations between metabolite levels (metabolomes) and diseases to understand metabolic pathways involved in disease.
  • Phenome-wide Association Study (PheWAS): A study that examines the relationships between genetic variants and a wide range of phenotypes to identify potential pleiotropic effects.
  • Electronic Health Records (EHRs): Digital versions of patients’ paper charts that contain medical and treatment information, which can be used for research purposes.
  • IEU Open GWAS database: A platform that houses summary statistics from various GWAS studies, facilitating the use of genetic data in Mendelian randomization analyses.
  • Connectivity Map (CMap): A database that profiles the gene expression changes induced by various drugs and compounds, facilitating the identification of drugs that may reverse disease-related expression changes.
  • Drug Gene Interaction Database (DGIdb): A comprehensive resource that provides information on the interactions between genes and drugs, aiding in drug repurposing efforts.
  • Protein-Protein Interaction (PPI) Network: A biological network that illustrates the physical interactions among proteins, useful for understanding biological processes and drug mechanisms.
  • Off-target Effects: The effects of a drug on targets other than the intended one, which can lead to adverse drug events.
  • Adverse Drug Events (ADEs): Harmful or unintended consequences resulting from the use of a drug.
  • pQTL: Protein quantitative trait loci; genetic loci that regulate protein abundance.
  • cis-eQTL: genetic loci that regulate the expression of nearby genes.

Reference

Wang, L., Lu, Y., Li, D., Zhou, Y., Yu, L., Mesa Eguiagaray, I., … & Theodoratou, E. (2024). The landscape of the methodology in drug repurposing using human genomic data: a systematic review. Briefings in bioinformatics25(2), bbad527.

Shares