From Impossible to Inevitable: Transforming Undruggable Targets

Delve into the challenges and innovative approaches of making undruggable targets druggable.

From Impossible to Inevitable: Transforming Undruggable Targets

In the dynamic world of drug discovery, the notion of "undruggable" targets presents both a significant challenge and an intriguing frontier for researchers and pharmaceutical companies. These tough nuts to crack in medical science—biological targets known to play roles in diseases but resistant to traditional drug design—are now seeing new strategies that shift the paradigm from "undruggable" to "druggable."

The Challenge of Selectivity 

Why are some targets so elusive? A big part of the puzzle is selectivity—the ability of a drug to zero in on specific biological molecules without affecting others. This is key to avoiding side effects. Consider aminergic GPCRs and kinases—while many drugs hit these proteins, they often affect more than one target, a phenomenon known as polypharmacology, leading to complex treatment profiles and side effects.

Tyrosine phosphatases offer a similar story. Their active sites are so similar across the family, it’s tough to interact selectively while keeping the drug properties we want. Here, researchers are looking at allosteric modulation—hitting different parts of the molecule to increase selectivity. These parts are less alike, which means less chance of side effects, but they're tricky to work with since they're small, dislike water, and aren’t naturally evolved to bind with drugs.

Addressing Flexibility in Drug Targets

Another big hurdle is target flexibility.  Traditional drug design "lock and key" models fall short when the lock (binding site) changes shape upon the key's (ligand's) insertion. This is particularly true for intrinsically disordered proteins (IDPs), which may adopt a structured form only upon binding with a ligand. Traditional approaches often fail here, requiring different strategies like pharmacological screening to uncover effective molecules.

For proteins that exhibit some flexibility, understanding how they change upon ligand binding is crucial. Molecular dynamics (MD) simulations and other computational methods are vital tools in our arsenal, helping us peek into the atomic-level interactions and movements within the protein, helping reveal potential new binding sites induced by small molecule interactions.

Beyond Proteins: DNA and RNA Frontier

The story doesn’t end with proteins. DNA and RNA are also key players, each bringing unique challenges. Many cancer treatments target DNA, but often these can cause collateral damage, leading to severe side effects. The ultimate goal is to develop molecules that can interact with DNA without damaging it. RNA, with its diverse functions and structures, offers a new frontier in drug design. Its ability to form complex three-dimensional structures makes it a particularly enticing target, akin to proteins, but its small sequences, like microRNAs, require specialized approaches such as library vs. library screens to find starting points for drug development.

Conquering Large, Flat Interfaces

A core challenge in drug discovery is the "difficult pockets" seen in protein-protein interfaces (PPIs) and between proteins and DNA/RNA. The issue here is not just about finding a molecule that fits; it’s about navigating and effectively targeting large, flat surfaces that lack the defined crevices typically exploited by traditional small-molecule drugs.

The Protein-Protein Interface Problem

PPIs are particularly tough because of their extensive, flat areas dotted with multiple hotspots—specific amino acids crucial for binding affinity. To effectively inhibit or modulate these interactions, a small molecule must engage with several of these hotspots, which requires precise interaction. This challenge is compounded by the fact that natural binding affinities at these interfaces can be incredibly strong, often in the sub-nanomolar range.

Further yet, the lack of a traditional, well-defined pocket means that typical entropic benefits—gained when molecules bind and order water molecules around them—are missing, complicating the development of high-affinity drugs.

Strategies to Tackle PPIs

Despite these challenges, some wins in targeting PPIs with small molecules have emerged. Successful candidates are usually larger and more complex, designed to mesh with multiple regions across protein surfaces. Techniques like free energy calculations and mixed solvent methods play pivotal roles in identifying PPI hotspots and revealing inducible pockets. Additionally, mixed solvent methods can be used to reveal inducible pockets, enhancing our understanding of where a drug might bind effectively. Aromatic interactions, key targets for intervention, are often finely tuned using quantum mechanics (QM) methods to estimate binding energy and stability.

Precision Strikes with Covalent Ligands: 

Another exciting approach uses covalent ligands. These molecules form a permanent bond with specific amino acids in the target protein, which can dramatically increase the binding strength and duration of their effect. But there’s a fine balance to strike—if a molecule is too reactive, it risks binding indiscriminately to off-target proteins, leading to potential toxicity.

Optimal covalent drug design combines strong binding affinity with moderated reactivity to ensure specific interactions within the intended site. Quantum mechanics provide detailed insights into the electrophilicity index of molecules—its propensity to attract electrons—which is crucial for predicting how a drug will interact with its target.

The Role of Allosteric Modulators

And then there’s the challenge of hitting targets like phosphatases. Here you need a molecule that’s just as polar as the highly polar active sites, but this often results in poor drug-like properties, such as suboptimal absorption, distribution, metabolism, and excretion (ADME) profiles. One way around this is allosteric modulation—targeting different parts of the enzyme to influence its activity indirectly.

Techniques such as fragment-based screening, which can utilize NMR or X-ray methods, are crucial for identifying these allosteric binders. These fragments, though initially with low binding affinity, provide a scaffold that can be optimized into effective drugs through medicinal chemistry.

The Big Picture

The fight against "undruggable" targets is daunting, but advances in computational tools and chemical strategies are making waves. High-throughput screening and sophisticated models are just the beginning. Success stories in targeting tough cases like Bcl-2 in cancer and ongoing efforts with RAS inhibitors highlight the potential of these approaches.

This deep dive into the world of "undruggability" reflects the broader journey of drug discovery—a path not just about finding new treatments, but about pushing the boundaries of what’s possible in medical science. The path forward is lined with challenges but also with tremendous potential to transform lives, offering new hope to those with few existing options and painting a brighter future for healthcare worldwide.

Stay informed by signing up for our newsletter, where you'll gain early access to the latest insights, trends, and breakthroughs in drug discovery, powered by cutting-edge data and analysis from industry-leading experts.