Introduction to Fragment-Based Drug Discovery
Definition and Basic Concepts
Fragment-based drug discovery (FBDD) is a methodology in medicinal chemistry that begins with the identification of very small, low molecular weight compounds—called fragments—that bind weakly to target proteins. These fragments typically have molecular weights less than 300 Da and exhibit a minimalistic structure while maintaining high ligand efficiency. Despite their weak binding affinities (often in the millimolar to micromolar range), fragments can be chemically elaborated or “grown” through the attachment of additional functional groups to improve binding affinity and specificity. FBDD relies heavily on biophysical methods such as high-throughput X-ray crystallography, nuclear magnetic resonance (NMR) spectroscopy, and surface plasmon resonance (SPR) to detect these subtle interactions. According to several reviews, including those summarized by
Astex’s own experience, FBDD provides the structural insights required to design new drugs by revealing precise binding modes of small molecules to their target proteins.
Advantages over Traditional Methods
One of the key advantages of FBDD is its efficient exploration of chemical space. Whereas traditional high-throughput screening (HTS) relies on large libraries of drug-sized molecules—often covering only a minute fraction of the enormous possible chemical space—FBDD approaches use much smaller collections of fragments that can sample diverse chemical “building blocks” with higher hit rates. This efficiency in sampling arises from the smaller number of compounds needed to cover a vast range of chemical diversity, sometimes described as “fragment space.” In addition, the high ligand efficiency inherent to many fragments allows for a higher probability that even a weak binding event can serve as the starting point for a robust, high-affinity ligand once it has been optimized. The methodology also offers the possibility of iterative improvements by providing three-dimensional structural information that can guide subsequent synthetic elaborations, thereby reducing both time and cost during candidate optimization.
Astex's Strategy in Fragment-Based Drug Discovery
Strategic Objectives
Astex Pharmaceuticals has emerged as one of the pioneers in applying FBDD in a structured and highly integrated manner. Their strategy is driven by several core objectives:
1. Rapid Generation of Lead Compounds from Weak Starting Points:
Astex’s FBDD strategy is built on the recognition that small, low-affinity fragments can be systematically optimized into highly potent and selective lead compounds. This strategic objective is exemplified by their successful discovery of compounds such as
AT7519 and other kinase inhibitors, which were advanced rapidly to clinical candidates.
2. Exploitation of Structural Information:
A centerpiece of Astex’s approach is its reliance on obtaining high-resolution three-dimensional structural data for fragment–protein interactions. With over 1,400 in-house X-ray crystal structures of fragments, the company has developed an extensive repository of structural information that guides the rational design and subsequent optimization (“growing” and “linking”) of fragment hits into lead compounds. This structural knowledge allows the design teams to define optimal growth vectors for chemical elaboration and minimizes synthetic intractabilities.
3. Integration of Robust Synthetic Methodologies:
Astex is conscious that many fragment hits are not progressed simply because of synthetic challenges. Hence, they have invested in innovative synthetic organic chemistry techniques to overcome the limitations of traditional synthetic routes. Their strategy includes developing new methodologies for polar, unprotected fragments that enable the efficient access to novel chemical space. This objective is driven by the recognition that the evolution of fragments along multiple vectors (i.e., adding new functional groups at different precise positions on the fragment molecule) is critical to advancing initial hits into clinically viable leads.
4. Collaboration with Pharmaceutical Giants and Academic Partners:
Astex’s approach is not carried out in isolation; strategic partnerships with industry leaders like
MSD, Merck, and
AstraZeneca are an integral part of their business model. These collaborations allow Astex to leverage external capabilities, assume shared risk, gain access to extended clinical expertise, and secure significant milestone payments—all while using their FBDD platform as the catalyst for identifying potential breakthrough therapeutics.
5. Targeting Challenging and “Undruggable” Proteins:
Understanding that many therapeutically relevant targets have historically been deemed “undruggable” by traditional screening methods, Astex’s FBDD strategy deliberately selects targets with amenable structural systems, especially those where X-ray crystallography can reliably provide structural data. This includes protein–protein interactions, allosteric sites, and enzymes such as
protein kinases where selectivity and potency can be achieved through fragment elaboration.
6. Multidisciplinary Integration:
Astex’s strategy emphasizes a synergistic integration of computational chemistry, biophysical screening, medicinal chemistry, and innovative synthetic organic chemistry. By coupling in silico techniques such as molecular docking and virtual screening with experimental biophysical methods, they enhance the hit-to-lead optimization process, reducing attrition and streamlining the path from fragment to candidate.
Implementation Process
The implementation of Astex’s FBDD strategy is a multi-step, iterative process that can be viewed from several perspectives:
1. Fragment Screening:
Astex begins by assembling libraries of well-characterized fragments that adhere to the “rule of three” (i.e., low molecular weight, low lipophilicity, and minimal hydrogen bond donors/acceptors), ensuring that only high-quality chemical space is explored. These fragments are screened using highly sensitive biophysical methods, particularly high-throughput X-ray crystallography, which allows them to simultaneously detect multiple binding events, including allosteric binding. This initial screening stage is crucial for identifying weak binders that can later be optimized.
2. Structural Determination and Analysis:
Once fragment hits are identified, Astex employs high-throughput crystallography to obtain detailed three-dimensional structures of the fragment–protein complexes. The structural data reveal precise binding modes including growth vectors that indicate where modifications can be made to enhance binding affinity. Having a robust internal database of fragment-bound structures not only speeds up fragment-to-lead progression but also improves the predictive power of computational modeling techniques.
3. Computational Modeling and Design:
With the structural data in hand, Astex integrates computational tools to analyze the fragment binding sites, predict the optimal pathways for growth, and design chemical modifications. Advanced algorithms, often combined with AI and machine learning techniques, are used to model different fragment elaboration strategies (growing, merging, linking) and to evaluate potential synthetic routes. These computational methods help prioritize synthetic efforts and guide medicinal chemists in designing molecules with improved drug-like properties.
4. Synthetic Elaboration:
One of the hallmarks of Astex’s strategy is the focus on overcoming synthetic challenges early in the process. They invest in innovative synthetic methodologies specially tailored to polar, unprotected fragments that are typically more challenging to elaborate. By engaging in synthetic research as part of their FBDD platform, they ensure that the chosen fragments not only bind well to the target but are also synthetically tractable, which increases the likelihood of advancing a fragment hit into a clinically viable lead.
5. Medicinal Chemistry and Lead Optimization:
Following structural elucidation and computational design, the next stage is intensive medicinal chemistry work.
Astex’s multidisciplinary teams work iteratively to modify the initial fragments, gradually increasing binding affinity while optimizing pharmacokinetic properties such as solubility, absorption, and metabolic stability. Throughout this phase, continuous feedback from biochemical and biophysical assays aids in refining the lead compounds. The success of this iterative medicinal chemistry process is evident in the progression of several FBDD-derived compounds into clinical trials.
6. Collaborative Development and Commercialization:
Recognizing the high cost and complexity associated with later stages of drug development, Astex enters into strategic partnerships with larger pharmaceutical companies. These collaborations often involve milestone-based financial arrangements and shared responsibilities in further optimization, preclinical studies, and clinical trials. By partnering with established industry leaders, Astex can leverage additional resources and expertise, accelerating the overall drug development process while de-risking individual projects.
7. Interdisciplinary Integration:
Throughout the entire process, Astex’s strategy is characterized by the seamless integration of multiple disciplines. Medicinal chemists, structural biologists, computational modelers, and synthetic organic chemists work in a coordinated manner. This multidisciplinary integration is pivotal in translating weak fragment hits into potent, selective, and clinically viable drug candidates. The rapid exchange of insights among these teams fosters innovation and enhances the overall efficiency of drug discovery.
Case Studies and Examples
Successful Drug Developments
Astex’s application of FBDD has led to several successful drug developments and notable case studies that highlight their strategic approach:
1. Discovery of
Cyclin-Dependent Kinase Inhibitors:
Astex has pioneered the development of cyclin-dependent kinase (CDK) inhibitors using FBDD. For instance, they discovered AT7519, a novel CDK inhibitor initially identified through fragment screening and optimized using structure-based design. This project showcased how a low-affinity fragment could, through iterative synthetic elaboration and targeted medicinal chemistry, be transformed into a clinically relevant candidate. The success of such programs has validated the effectiveness of their FBDD platform in tackling complex enzymatic targets.
2. Fragment-Derived Oncology Drugs:
Astex’s innovative strategy has also resulted in the discovery and development of oncology drugs. Their work contributed to drugs such as ribociclib (Kisqali®) and erdafitinib (Balversa®), which emerged from fragment-based approaches focusing on protein kinases and protein–protein interactions. These developments are significant because they demonstrate the ability of the FBDD strategy to yield potent, structurally novel compounds that translate into real clinical benefits for cancer patients.
3. Development of Allosteric and Protein-Protein Interaction Inhibitors:
Astex’s approach is not limited to active-site inhibitors but extends to modulators of allosteric sites and protein–protein interactions, areas that are traditionally challenging for conventional drug discovery. For example, studies have shown that fragment-based methods can be used to identify ligands that bind outside the conserved active sites, thereby offering new avenues for selectivity. These successes underscore the flexibility and broad applicability of Astex’s FBDD strategy for diverse therapeutic targets.
4. Integration with High Throughput Platforms:
Astex has implemented sophisticated high-throughput crystallography and biophysical screening techniques that have enabled the rapid accumulation of structural data. Over 1,400 fragment-bound structures have been generated in-house, providing an invaluable resource for design and optimization. This extensive structural repository has supported successful fragment-to-lead progressions and has been crucial in developing drug candidates with robust potency and selectivity.
Lessons Learned
Several key lessons have emerged from Astex’s extensive experience with FBDD:
1. The Importance of Synthetic Tractability:
One of the recurrent challenges in fragment-based approaches is that many fragment hits do not progress because of synthetic intractability. Astex’s investment in the development of new synthetic methodologies to elaborate polar, unprotected fragments has proven critical. This lesson emphasizes that early consideration of synthetic feasibility is as important as measuring binding affinity.
2. Value of Structural Information:
The rapid generation and analysis of crystal structures have been vital for refining fragment hits. Astex’s practice of obtaining and analyzing high-resolution X-ray data early in the discovery process has provided detailed insights into binding vectors and allowed for the rational design of chemistry routes to improve binding affinity. This approach has taught the importance of integrating structural biology with medicinal chemistry from the outset.
3. Efficacy of Multidisciplinary Collaboration:
Astex’s approach highlights that FBDD is inherently multidisciplinary. The close collaboration between structural biologists, computational chemists, synthetic chemists, and medicinal chemists is essential for the rapid evolution of fragments into drug candidates. The success stories in oncology and kinase inhibitor programs are clear illustrations that a well-integrated team approach can overcome traditional challenges in drug discovery.
4. Partnerships as a Strategic Advantage:
Collaborations with major pharmaceutical companies have not only de-risked individual projects but also provided the necessary resources to navigate the costly clinical development process. Astex’s strategic alliances with organizations like MSD, AstraZeneca, and Merck have been instrumental in scaling up promising candidates from bench to bedside. These partnerships, based on milestone payments and shared developmental responsibilities, underscore the need for a collaborative ecosystem in modern drug discovery.
5. Adaptability to New Technologies:
Throughout its evolution, Astex has demonstrated a willingness to adopt and integrate new technologies—such as computational docking, AI-driven design, and high-throughput advanced synthetic methods—into their FBDD platform. This adaptability has been critical in maintaining a competitive edge and in the efficient translation of fragment hits into validated clinical candidates.
Challenges and Innovations
Current Challenges
Despite the promising track record of FBDD and the successes achieved by Astex, several challenges remain:
1. Detection of Weak Binding Events:
The very nature of fragment binding—being weak and transient—poses acute challenges in detecting and accurately characterizing these interactions. Techniques such as NMR and X-ray crystallography are sensitive but require high-quality crystals, significant protein quantities, and careful consideration of signal-to-noise ratios. These challenges necessitate continued innovation in biophysical methodologies to reliably capture and interpret fragment binding data.
2. Synthetic Elaboration of Fragments:
Many fragment hits exhibit synthetic complexity or lack obvious synthetic handles, making it difficult to convert them into high-affinity lead compounds. Synthetic intractability is a common bottleneck, where promising fragments are abandoned because suitable analogues are not commercially available or synthetic routes are too challenging. Astex’s strategy of developing new synthetic methodologies specifically to address these challenges is ongoing but remains a difficult hurdle.
3. Target Selectivity and Protein–Protein Interactions:
Achieving high selectivity, particularly when targeting similar binding sites within large enzyme families like the protein kinases, is inherently challenging. Although FBDD provides a pathway to develop selective inhibitors, the optimization process must contend with the structural similarity of many potential off-targets. Furthermore, targeting protein–protein interactions adds another layer of complexity due to the often flat and expansive binding interfaces.
4. High Throughput and Scalability Issues:
While Astex has heavily invested in high-throughput crystallography and other screening technologies, scaling these procedures to screen larger fragment libraries across diverse targets remains a technical challenge. Balancing throughput and data quality is critical, as insufficient data can hinder the accurate determination of binding modes.
5. Integration of Computational and Experimental Data:
One persistent challenge is the effective integration of computational predictions with experimental data. Despite advances in docking algorithms and machine learning, reliably predicting fragment binding modes and subsequent growth strategies requires continual refinement. This integration is essential for reducing cycle times in lead optimization and for enhancing the overall predictability of the FBDD process.
Innovative Solutions and Future Directions
In response to the challenges outlined above, Astex and others in the field have proposed and are implementing several innovative solutions:
1. Advanced High-Throughput Technologies:
Astex’s integration of high-throughput X-ray crystallography has been a game-changer, enabling the rapid generation of hundreds or even thousands of co-crystal structures. Looking forward, further refinement of automation—including acoustic droplet ejection and robotic automounters—will enhance both the throughput and quality of fragment screening. The pursuit of techniques such as crystal soaking protocols that minimize sample consumption is expected to further improve efficiency.
2. Novel Synthetic Organic Chemistry Approaches:
To address the synthetic intractability of certain fragments, Astex is developing innovative synthetic methods to facilitate fragment elaboration. Recent research focuses on the modular advancement of sp³-rich fragments using C(sp³)–H functionalization and other new transformations. This not only expands the repertoire of accessible chemical space but also ensures that promising fragments can be advanced even when they initially seem synthetically challenging.
3. Enhanced Computational Tools and AI Integration:
The application of computational tools is critical in modern FBDD. Astex leverages state-of-the-art docking algorithms, molecular dynamics simulations, and AI by integrating machine learning techniques to predict binding poses and synthetic feasibility. These computational approaches complement the biophysical and synthetic chemistry aspects, shortening design cycles and improving hit-to-lead conversion rates. AI-assisted platforms are anticipated to become even more central, enabling more precise predictions and offering real-time predictive adjustments during fragment optimization.
4. Wide-Ranging Fragment Libraries:
Recognizing the importance of chemical diversity, Astex continues to develop and refine its fragment libraries. By mining structural databases such as the Protein Data Bank and integrating fragments with marked three-dimensional character, they can increase hit rates and provide more robust starting points for lead development. These libraries are continually updated to incorporate new fragments that are both synthetically tractable and capable of engaging challenging targets.
5. Interdisciplinary and Collaborative Networks:
The future of FBDD at Astex will likely feature even stronger interdisciplinary collaborations. The integration of academia, computational chemistry, and synthetic organic chemistry is expected to further enhance innovation while the company’s strategic partnerships with Big Pharma will facilitate clinical development and commercialization. This collaborative ecosystem is essential not only for risk mitigation but also for ensuring that promising candidates receive the full spectrum of expertise required to advance them toward clinical trials.
6. Adopting Emerging Biophysical Techniques:
Emerging methods such as cryo-electron microscopy (cryo-EM) and cellular thermal shift assays (CETSA) are beginning to impact the field of FBDD. While X-ray crystallography remains the gold standard, these new techniques may offer complementary insights—particularly for targets that are not readily crystallizable. Astex is well placed to incorporate these advancements to further increase the robustness of binding mode determinations and to enable fragment screening in more challenging biological contexts.
7. Focus on Therapeutic Areas with High Unmet Need:
Astex’s strategy is deeply aligned with targeting therapeutic areas where current treatment options are limited, such as oncology and central nervous system disorders. By focusing on high-unmet-need areas, they are able to maximize the potential clinical impact of their FBDD-derived compounds. This strategic prioritization ensures that the novel molecules developed not only have strong scientific merit but also address critical gaps in patient care.
Conclusion
Astex’s strategy in fragment-based drug discovery is a comprehensive, multidisciplinary, and highly iterative process that starts with the identification of low molecular weight fragments through sensitive biophysical screening methods and culminates in the development of clinically validated drug candidates. At the highest level, their approach is underpinned by the need to efficiently sample chemical space, achieve high ligand efficiency, and capitalize on structural insights to guide medicinal chemistry.
From a general perspective, FBDD offers advantages over traditional high-throughput screening—chief among them being the ability to explore vast chemical space with smaller, more diverse libraries, while maintaining a focus on developing highly potent and selective leads. Astex has taken these foundational principles and, through strategic objectives that include rapid lead generation, rigorous structural determination, and innovative synthetic elaboration, has established itself as a leader in the field. Their emphasis on using high-throughput crystallography and advanced computational tools has enabled them to convert weak fragment hits into powerful clinical candidates in a remarkably short timeframe.
From a specific perspective, the implementation process at Astex is characterized by an exacting sequence: initial fragment screening using robust biophysical techniques, detailed structural analysis to reveal growth vectors, computational modeling to design optimal modifications, and iterative synthetic chemistry to assemble these fragments into lead compounds. The successful application of this process has yielded multiple case studies, including the development of CDK inhibitors like AT7519, the advancement of oncology drugs such as ribociclib and erdafitinib, and the exploration of allosteric inhibitors. These successes underscore the value of an integrated approach that leverages the strengths of both technology and collaboration. Moreover, strategic partnerships with major pharmaceutical companies have amplified their capabilities, providing not only additional resources but also validation of their methodologies through shared clinical and commercial successes.
From a general yet forward-looking perspective, despite the inherent challenges related to detecting weak binding events, overcoming synthetic hurdles, and ensuring high selectivity, Astex continues to innovate by integrating advanced high-throughput technologies, novel synthetic methods, and AI-driven computational tools. Their willingness to adapt and embrace emerging biophysical techniques like cryo-EM exemplifies their commitment to staying at the forefront of drug design technology. The future directions of their strategy include further refinement of fragment libraries, enhanced interdisciplinary collaborations, and a continued focus on addressing therapeutic areas with significant unmet medical needs.
In conclusion, Astex’s strategy in using fragment-based drug discovery can be summarized as a general-specific-general approach: it begins with a broad commitment to harness the advantages of FBDD over traditional methods, drills down to the specific processes and multidisciplinary integrations that transform weak fragments into robust drug candidates, and then broadens again into a vision for future innovative therapeutic solutions. This holistic strategy—underscored by rigorous structural and synthetic chemistry and enabled by strategic collaborations—positions Astex as a leader in the evolving landscape of fragment-based drug discovery. Their iterative and integrated approach not only maximizes the potential of each fragment hit but also sets a high standard for how fragment-based methods can be effectively and efficiently translated into transformational medicines.