Skip to content

What is target identification in druggable? The Foundational Step of Drug Discovery

5 min read

According to the National Institutes of Health, while the human genome contains approximately 4,500 genes considered part of the 'druggable genome', existing clinical drugs target only a few hundred of these. Therefore, understanding what is target identification in druggable is a critical first step in discovering new medications and unlocking the full potential of these untapped targets.

Quick Summary

Target identification within the druggable genome is the process of finding and validating specific molecules, like proteins or genes, that can be modulated by a therapeutic agent to treat a disease. It is the crucial initial phase of modern drug discovery.

Key Points

  • Foundational Drug Discovery Step: Target identification is the critical process of pinpointing a specific molecule, such as a protein or gene, that a drug can modulate to produce a therapeutic effect.

  • Understanding 'Druggable': A 'druggable' target is a biological molecule with a high likelihood of being effectively and selectively modulated by a drug-like agent.

  • Diverse Methodologies: Identification strategies include experimental techniques (affinity-based, label-free), genetic screening (CRISPR), and computational methods (AI, multi-omics).

  • Crucial Validation Process: After identification, target validation is essential to confirm that modulating the target is relevant to the disease and therapeutically beneficial, minimizing clinical trial failures.

  • Leveraging Technology: Modern drug discovery heavily relies on integrating data from various 'omics' and using AI/machine learning to accelerate the search for promising targets.

  • Persistent Challenges: Major hurdles include the existence of 'undruggable' targets that lack binding sites, achieving high drug selectivity to prevent side effects, and accurately translating findings from preclinical models to humans.

In This Article

The Centrality of Target Identification in Drug Discovery

Target identification is the crucial first phase in the drug discovery pipeline. It involves identifying and validating specific biological molecules, often proteins or genes, that play a key role in a disease process and can be modulated by a drug. Early and accurate target identification is vital because it sets the foundation for all subsequent development, significantly influencing the drug's efficacy and safety profile. Selecting a subpar target is a major cause of failure in later, more expensive clinical trials. This process is contrasted with phenotypic screening, where compounds are tested for desired effects on cells or organisms without prior knowledge of the target, necessitating a later 'target deconvolution' step to identify the mechanism of action.

What is a 'Druggable' Target?

The concept of a 'druggable' target is fundamental to modern pharmacology. The 'druggable genome' is the subset of the human genome that expresses proteins capable of binding drug-like molecules. A target's 'druggability' refers to the likelihood that it can be effectively modulated by a therapeutic agent, taking into account the physicochemical properties required for a drug-like molecule. This assessment helps researchers prioritize which targets have the highest potential for yielding a successful drug candidate.

Key characteristics of a 'good' druggable target include:

  • Clear disease association: The target's role in the pathophysiology of the disease is well-defined.
  • Modulability: The target's function can be altered by a drug, typically via a specific binding pocket.
  • Accessibility: The target is located where a drug can reach it, whether inside or outside a cell.
  • Selectivity: The drug's action can be confined primarily to the intended target to minimize off-target effects and reduce side effects.
  • Favorable safety profile: Blocking the target's physiological function should not cause unacceptable toxicity in healthy tissues.

Strategies for Target Identification

There are several modern approaches for identifying potential druggable targets, which can be broadly classified into experimental, multi-omic, and computational strategies.

Experimental Methods

Experimental, or 'wet lab', methods involve direct manipulation and analysis of biological systems.

  • Affinity-based approaches: Techniques like affinity pull-down and photoaffinity labeling use probes that bind to target proteins, allowing them to be isolated and identified, often via mass spectrometry. These methods provide direct evidence of physical interaction between a potential drug and a protein.
  • Label-free methods: Techniques such as the Cellular Thermal Shift Assay (CETSA) or Drug Affinity Responsive Target Stability (DARTS) measure changes in a protein's stability in response to a compound without requiring chemical modifications to the molecule. CETSA can be performed in live cells, offering a more physiologically relevant context.
  • Genetic screening: Using tools like CRISPR-Cas9 or RNA interference (RNAi), researchers can systematically knock out or knock down genes to see which genetic alterations produce a phenotype similar to a drug's effect. This helps establish a causal link between the gene and the disease phenotype.

Computational and Multi-omic Approaches

These strategies leverage large datasets and powerful analytical tools to predict and prioritize targets.

  • Bioinformatics and AI/Machine Learning: Computational tools can analyze vast amounts of genomic, proteomic, and other biological data to identify correlations between genes/proteins and disease states. AI algorithms can predict new drug-target interactions and assess druggability by analyzing protein sequences and structures.
  • Multi-omics Integration: This advanced method combines and analyzes multiple 'omics' datasets—such as genomics (genetic sequences), transcriptomics (gene expression), and proteomics (protein levels)—to build comprehensive models of disease networks and identify key regulatory nodes as potential targets.

A Comparison of Target Identification Approaches

Feature Experimental/Biochemical Methods Genetic Screening (CRISPR/RNAi) Computational/AI Methods
Principle Relies on direct physical interaction or change in protein properties Relies on altering gene expression to infer function Relies on analyzing large datasets and predictive models
Throughput Can be moderate-to-high, especially with automation High, particularly for genome-wide screens Very high (in silico), allowing rapid hypothesis generation
Evidence Type Biophysical binding and direct interaction data Functional evidence linking gene to phenotype Predictive, requires experimental validation
Target Bias Can be biased towards known classes of targets or ligand-binding properties Less biased, can uncover novel targets or pathways Less biased, can explore entire biological networks
Physiological Relevance Varies (cell lysate vs. live cell methods) High (conducted in living cellular systems) Low (in silico), needs extensive lab validation
Cost Often resource-intensive (reagents, instrumentation) Requires specialized libraries and setup Low initial cost for analysis, but validation is expensive

The Crucial Role of Target Validation

Once a potential druggable target has been identified, it must be rigorously validated. Target validation confirms the molecular target's involvement in the disease and its potential to deliver a therapeutic effect. This process involves a series of experiments, often using techniques like gene knockouts or specific chemical probes, to demonstrate that modulating the target directly affects the disease phenotype in relevant models. Inadequate target validation is a key reason many drug candidates fail during clinical development.

Challenges in Targeting the Druggable Genome

Despite advances, identifying and developing drugs for the druggable genome presents several challenges. The high attrition rate in drug discovery, where many candidates fail in clinical trials, often stems from poor target selection. A significant portion of the proteome is considered 'undruggable' because proteins lack accessible binding pockets for drug-like small molecules, presenting a major obstacle. Furthermore, designing selective drugs that avoid off-target effects and associated toxicities remains a complex issue, as demonstrated by examples like the cardiotoxicity observed with some kinase inhibitors. Integrating the vast and disparate data generated by different omics technologies and AI predictions is another challenge that requires careful handling and interpretation.

Conclusion

Target identification within the druggable genome is the cornerstone of modern, target-based drug discovery, providing the roadmap for developing new medications. By integrating diverse strategies, from traditional biochemical experiments to cutting-edge computational and multi-omic approaches, researchers can identify promising candidates and significantly improve the efficiency and success rate of the drug development pipeline. The future of pharmacology lies in the continued exploration of the currently understudied portion of the druggable genome, driven by advances in AI and other technologies, to address critical unmet medical needs and pave the way for more effective, targeted therapies. The key to success remains the rigorous validation of potential targets before committing to expensive and time-consuming clinical trials.

Authoritative Link

Frequently Asked Questions

Target identification is crucial because it helps researchers understand the disease mechanism and ensures that drug development efforts are focused on molecules that are most likely to provide a therapeutic benefit. This reduces the risk of costly failures in later development stages.

The 'druggable genome' is the subset of genes within the human genome that express proteins capable of binding with drug-like molecules. While the total number is large, only a small fraction is currently targeted by approved drugs.

Druggability is assessed by evaluating features such as the presence of suitable binding pockets, the protein's properties, and its physiological function. This analysis helps determine the feasibility of modulating the target with a therapeutic agent.

Target-based screening starts with a known molecular target and searches for a compound that binds to it. Phenotypic screening tests compounds for a desired effect in cells or organisms without prior knowledge of the target, requiring later identification of the molecule responsible.

AI and machine learning analyze vast biological datasets to predict and prioritize potential drug targets. These computational methods can accelerate the discovery process by identifying patterns and networks related to disease.

Key challenges include the difficulty of finding suitable small-molecule binding sites on many proteins, the potential for off-target effects, and ensuring the findings from experimental models accurately translate to human patients.

Target validation follows identification and involves rigorous experiments to confirm that the selected target is genuinely involved in the disease and that modulating it will produce the desired therapeutic outcome. It's a critical step before clinical trials.

References

  1. 1
  2. 2
  3. 3
  4. 4
  5. 5
  6. 6
  7. 7
  8. 8

Medical Disclaimer

This content is for informational purposes only and should not replace professional medical advice.