Navigating the Hurdles: A Researcher's Guide to Optimizing High-throughput screens, Library Design, and Protein Engineering
- Ranomics
- 13 hours ago
- 34 min read
Introduction: The Triad of Challenges in Accelerating Drug Discovery
The Imperative for Efficiency: The pharmaceutical and biotechnology sectors face continuous pressure to accelerate the discovery and development of novel assets for the benefit of global health. The rise of personalized medicine and the increasing prevalence of chronic conditions demand faster, more efficient R&D processes.1 However, classical drug discovery efforts are lengthy, costly and often result in failures.2 It is estimated that bringing a single new drug to market can take 10-15 years and cost upwards of $2.5 billion, with fewer than 14% of candidates entering Phase 1 clinical trials ultimately reaching patients.4 Attrition rates throughout the development pipeline often exceed 80-90%, placing immense financial strain on the industry and hindering the delivery of needed medicines.5
Introducing the Core Pillars: Within the complex landscape of drug discovery, with a focus on protein biologics, three interconnected pillars stand out as critical determinants of success in the early stages: High-Throughput Screening (HTS), Molecular Library Design, and Protein Stability and Engineering. HTS enables the rapid testing of vast biologic designs.1 The quality and design of the biologic libraries are screened against a certain target or biological phenotype.10 For biologics and targets requiring protein reagents, ensuring adequate protein stability, solubility, and expression is paramount, often necessitating sophisticated protein engineering strategies.12 Crucially, challenges and bottlenecks encountered within any one of these domains can cascade, significantly impacting overall R&D project timelines, costs, and the probability of success.6 The quality of hits identified through screening, the developability of the screening library compounds, and the robustness of protein reagents are all intrinsically linked to downstream success.6
Scope of the Guide: This guide aims to provide scientists and researchers engaged in drug discovery with an expert overview of the primary challenges, common pitfalls, and emerging solutions associated with HTS assay development, molecular library design, and protein stability and engineering. By synthesizing current knowledge and highlighting best practices, this resource seeks to offer actionable insights to navigate these complex areas more effectively, ultimately contributing to more efficient and successful therapeutic development.
Navigating the Labyrinth of High-Throughput Screening (HTS): Challenges and Solutions
The Promise and Premise of HTS: High-Throughput Screening (HTS) represents a cornerstone technology in modern drug discovery, employing robotics, advanced liquid handling, sensitive detectors, and data processing software to rapidly evaluate potentially millions of chemical or biological entities against specific targets.1 Its fundamental purpose is the identification of "hits"—compounds or molecules that modulate a biological target or pathway in a desired manner—which serve as starting points for optimization and lead development.9 HTS marked a significant departure from earlier, manual, hypothesis-driven approaches, offering scale and speed that accelerated the initial phases of drug discovery.1
Core Challenges in HTS Assay Development: Despite its power, developing and executing robust HTS assays presents numerous challenges:
Sensitivity & Specificity: A fundamental requirement is achieving an appropriate balance between assay sensitivity and specificity. The assay must be sensitive enough to reliably detect genuine biological activity, particularly for weaker interactions often encountered in fragment-based screening 20, yet specific enough to avoid misleading results from off-target effects or assay artifacts.21 Striking this balance, especially in complex cellular or biochemical systems, remains a significant hurdle.
Reproducibility: Ensuring assay reproducibility is paramount for the reliability of screening data. Results must be consistent across different wells on a plate, between multiple plates within a run, across different screening days, and potentially over the entire multi-year lifespan of a drug discovery program.19 Factors compromising reproducibility include inherent biological variability (e.g., cell passage number, confluence 23), environmental sensitivity amplified by miniaturization (e.g., temperature gradients, evaporation 23), reagent lot-to-lot variation, and subtle differences in instrument performance or handling. While automation significantly reduces manual variability 24, it does not eliminate all sources of inconsistency.
Miniaturization: The drive to reduce costs (reagents, library consumption) and increase throughput has led to progressive assay miniaturization, moving from 96-well plates to 384-, 1536-, and even higher density one pot formats.1 While beneficial, miniaturization introduces challenges. Lower cell numbers per well can decrease signal intensity, demanding more sensitive detection systems.9 Smaller volumes are also more susceptible to evaporation and edge effects, potentially impacting assay performance and reproducibility.23
Automation Integration: Automation is indispensable for HTS.1 However, integrating diverse instruments—liquid handlers, plate readers, robotic arms, incubators, imagers—into a cohesive, reliable, and efficient workflow is a complex engineering task.24 It requires sophisticated scheduling software and orchestration platforms to manage workflows, data flow, and potentially real-time decision-making, ensuring smooth operation and maximizing throughput.24
Data Handling & Analysis: HTS campaigns generate enormous volumes of data, often described as a "data explosion".18 A critical challenge lies in efficiently processing, managing, analyzing, and interpreting these massive datasets to extract meaningful biological insights and confidently identify true hits.18 This necessitates robust data management infrastructure (e.g., LIMS, ELN, specialized HTS platforms), statistical expertise for appropriate analysis and quality control, and increasingly, automated or machine learning guided data analysis pipelines to handle the scale and complexity.9
Cost-Effectiveness: While the cost per data point in HTS is low compared to manual methods 8, the initial capital investment in robotic systems, detection instrumentation, compound libraries, and supporting infrastructure can be substantial.3 Furthermore, the downstream costs associated with validating hits and pursuing false positives can significantly inflate the overall expense of a screening campaign.30 Miniaturization remains a primary strategy for controlling assay reagent and compound consumption costs.9
Bottlenecks in HTS Workflows: Even with automation, specific steps can limit overall throughput and efficiency:
Liquid Handling: The precise movement of liquids—reagents, buffers, compounds, cell suspensions—is fundamental to HTS. Traditional tip-based liquid handling can become a bottleneck due to speed limitations, the cost of disposable tips, and the potential for carry-over or cross-contamination.32 Newer non-contact technologies, particularly acoustic droplet ejection, offer significant advantages by enabling rapid, precise transfer of nanoliter volumes without tips, facilitating further miniaturization, reducing reagent consumption and waste, and eliminating cross-contamination.32
Data Management & Integration: As screening throughput increases, data handling often emerges as a major bottleneck.27 Challenges include dealing with data generated in disparate formats by various instruments 27, the time and error potential associated with manual data transcription or transfer 27, lack of standardized data structures, and delays in processing and analysis that impede timely decision-making.28 Overcoming this requires integrated data management solutions that automate data capture from instruments, standardize formats, centralize storage, and facilitate streamlined analysis and reporting.9
Logistics & Plate Management: The sheer scale of HTS involves managing vast compound libraries stored in plates, creating numerous assay plates (often involving parent-child relationships for dilutions or replicates), and tracking each plate through complex automated workflows.36 Finding, preparing, and staging the correct plates for screening can become a significant logistical bottleneck, even with high-capacity robotic systems.36 Robust laboratory information management systems (LIMS) or dedicated plate/substance management software, often integrated with barcoding and robotics, are essential for accurate tracking and efficient workflow management.36
The evolution of HTS illustrates a common pattern in technological advancement: solving one bottleneck often reveals or creates another downstream. Initial automation dramatically increased screening speed, but this success then highlighted limitations in data analysis capacity and logistical management.18 Consequently, optimizing HTS necessitates a holistic, systems-level approach. Merely accelerating the screening step is insufficient; investments in integrated data infrastructure, advanced analytical tools (including AI/ML), and efficient logistics are crucial to fully capitalize on the potential of high-throughput methodologies.3
Ensuring Data Integrity: Quality Control Best Practices: Rigorous quality control (QC) is non-negotiable for generating reliable HTS data and minimizing wasted effort on false leads. Key best practices include:
Assay Validation: Before embarking on a full screen, the assay must be thoroughly validated to confirm its suitability.23 This involves demonstrating its pharmacological relevance (e.g., using known ligands to show expected activity and mechanism 19), assessing its reproducibility under screening conditions 19, and ensuring its robustness against potential interferences (e.g., solvent tolerance 19).
Plate Design & Controls: Strategic placement of positive and negative controls on each assay plate is critical for monitoring assay performance, identifying systematic errors (e.g., drift, edge effects), and enabling appropriate data normalization.18 Effective controls provide the benchmark against which test compound activity is measured.18
Addressing Edge Effects: Thermal gradients or differential evaporation rates across a microplate can cause "edge effects," leading to inconsistent cell growth or assay performance in peripheral wells.23 Mitigation strategies include either omitting data from edge wells (reducing throughput and increasing cost) or implementing procedural adjustments like pre-incubating plates at room temperature after seeding to allow thermal equilibration.23
Key QC Metrics: Several statistical metrics are routinely used to quantitatively assess assay quality (See Table 1). Monitoring these metrics across plates and runs provides objective criteria for accepting or rejecting data and identifying potential issues with the assay or automation.
Outlier Detection: HTS data frequently contain outlier values resulting from experimental errors or unusual compound behavior.18 Statistical methods, ranging from simple approaches like Grubb's test 23 to more robust methods designed for HTS data (e.g., z*-score, SSMD*) 18, should be employed to identify and appropriately handle outliers to avoid skewing results.
Table 1: Key HTS Quality Control Metrics and Interpretation
Metric Name | Calculation Basis/Formula (Conceptual) | Interpretation | Typical Acceptable Range/Target Value | Relevant Sources |
Z'-factor | Compares means (μ) and standard deviations (σ) of positive (p) & negative (n) controls: `1 - 3(σp + σn) / | μp - μn | ` | Measures the separation band (assay window) between controls; assesses assay suitability for HTS. |
IC50/EC50 Stability | Potency (e.g., IC50) of a reference compound measured in each assay run. | Monitors assay consistency over time; detects "assay drift". Visualized using QC charts with warning/fail limits. | Stable IC50 within predefined limits (e.g., +/- 2-3 fold of historical mean) | 23 |
Minimum Significant Ratio (MSR) | Based on standard deviation (s) of log10(IC50) across runs: 10^(2√2 s) | Quantifies assay variability based on reference compound potency; indicates fold-change needed for significance. | 2-3 (Stable), 3-5 (Moderate Variation) | 23 |
Coefficient of Variation (CV) | Standard deviation (SD) divided by the mean (μ) for control wells: SD / μ | Measures relative variability within positive or negative control populations on a plate. | < 10% (Biochemical), < 25% (Cell-based) | 23 |
Signal-to-Background (S/B) | Ratio of mean positive control signal to mean negative control signal. | Indicates the dynamic range or magnitude of the assay signal relative to background. | Assay-dependent, generally > 2-3 | 18 |
Signal-to-Noise (S/N) | Ratio of (mean positive signal - mean negative signal) to SD of negative signal. | Measures the signal magnitude relative to the background noise (variability). | Assay-dependent, higher is better | 18 |
Mitigating False Positives and Assay Interference: A major challenge in HTS is the high frequency of false positives—compounds that appear active in the primary screen but fail confirmation or act via undesirable, non-specific mechanisms.30 These consume significant follow-up resources.30
Common Causes: False positives arise from various sources, including compound auto-fluorescence or quenching interfering with optical detection methods 15, compound aggregation leading to non-specific inhibition 10, inherent compound reactivity, and compounds belonging to classes known as Pan-Assay Interference Compounds which show activity across many assays due to non-specific interactions or assay artifacts.10
Mitigation Strategies:
Counter-screens and Orthogonal Assays: Implementing secondary assays with different detection principles or mechanisms helps filter out artifactual hits.15
Assay Design: Careful selection of assay format and detection method can minimize interference. Label-free methods are inherently less prone to certain artifacts.
Pan-Assay Inteference Compound Filtering: Computational filters based on known PAINS substructures can flag potentially problematic compounds, though caution is needed to avoid discarding genuinely active molecules.37
Mass Spectrometry-based HTS: MS offers a powerful alternative or complement to traditional HTS.15 By directly detecting and quantifying unlabeled analytes (substrates, products), MS avoids interference common in fluorescence/luminescence assays.15 This allows screening of a broader range of targets, including those difficult to label, often with higher physiological relevance and potentially lower costs.15 While historically limited by throughput, significant advances in automation and ionization techniques have dramatically increased speed. However, challenges remain, including potential isobaric interference (compounds with the same mass-to-charge ratio as the analyte) and the complexity of integrating MS into HTS workflows.15 Counter assays or tandem MS can help mitigate isobaric interference.15
AI/ML Approaches: Machine learning algorithms are increasingly used to analyze complex HTS datasets. They can identify subtle patterns indicative of assay interference, distinguish true biological responses from artifacts, and help prioritize hits for follow-up, potentially reducing the burden of experimental validation.3 Techniques like Minimum Variance Sampling Analysis (MVS-A) 30 and data valuation methods 31 aim to specifically identify false positives and true actives directly from screening data.
The limitations of traditional optical assays and basic statistical QC are driving a shift towards more sophisticated approaches. The rise of label-free detection methods like MS-HTS addresses the core problem of compound interference with the readout itself.15 Concurrently, the sheer volume and complexity of HTS data necessitate more advanced, data-driven QC. Machine learning is emerging as a powerful tool not just for hit identification but also for identifying problematic compounds and assay artifacts based on their data signatures, moving beyond simple metrics like Z' to a more nuanced understanding of data quality.30 This convergence aims to improve the reliability of HTS campaigns and reduce downstream failures.
Automation, while indispensable for the scale of HTS, presents its own set of considerations. It enhances reproducibility by minimizing human error 9, but the automated systems themselves must be rigorously validated and monitored. Poorly optimized automation can introduce systematic errors, such as exacerbating edge effects or causing issues due to environmental sensitivity in miniaturized formats.23 Therefore, robust QC metrics are not just for evaluating the assay biology but also for continuously verifying the performance of the automation platform itself.18 Effective HTS relies on well-controlled, validated automation, not just automation for its own sake.
Impact on R&D Timelines and Costs: Inefficiencies and challenges in HTS directly inflate R&D costs and extend timelines. Assay failures, high false-positive rates requiring extensive follow-up, and workflow bottlenecks all contribute to delays and wasted resources.1 Conversely, well-optimized HTS operations—leveraging appropriate automation, miniaturization, rigorous QC, and advanced technologies like MS-HTS and AI-driven analysis—can significantly accelerate the hit identification phase, provide higher quality starting points for medicinal chemistry, and potentially reduce the high attrition rates observed later in development.1
Designing Effective Molecular Libraries: Avoiding Pitfalls in Screening
The Library as the Foundation: The success of any screening campaign, whether HTS, fragment-based, or using display technologies, is fundamentally dependent on the quality, diversity, and suitability of the molecular library being screened.10 A well-designed and carefully curated library significantly increases the probability of identifying high-quality hits, thereby accelerating progress and reducing downstream attrition rates in drug development programs.11 Conversely, screening a poorly characterized or inappropriate library is often an exercise in futility, generating misleading results and wasting valuable resources.
Critical Considerations in Library Design: Several key factors must be considered when designing or selecting screening libraries:
Diversity: A primary objective is often to maximize the diversity of chemical structures and pharmacophores within the library.10 This broad sampling of "chemical space" increases the likelihood of finding molecules that interact with a wide range of biological targets.10 Diversity can be assessed computationally using various metrics and descriptors, such as Tanimoto similarity based on molecular fingerprints.10 However, the optimal level of diversity depends on the screening strategy; highly focused libraries may be preferred for specific target families, while broad diversity is crucial for novel target exploration.10 Different library modalities achieve diversity differently; for instance, fragment libraries, composed of smaller, less complex molecules, can cover a proportionally larger chemical space with fewer compounds compared to traditional HTS libraries.20
Complexity: Molecular complexity, including factors like the number of chiral centers and the fraction of sp3-hybridized carbons (Fsp3), influences a molecule's shape and potential to interact with complex biological sites, such as protein-protein interfaces.10 While simpler molecules, like those in fragment libraries, offer efficient binding and easier optimization pathways 20, increased three-dimensionality is often sought in HTS libraries to explore novel biological space and potentially improve downstream properties.10 A balance must be struck, as overly complex molecules can be challenging and costly to synthesize and optimize.10
Physicochemical Properties ('Drug-likeness'/'Lead-likeness'): Compounds intended as starting points for drug discovery should possess physicochemical properties conducive to favorable absorption, distribution, metabolism, excretion, and toxicity (ADMET) profiles, as well as good optimization potential. Properties such as molecular weight (MW), lipophilicity (calculated LogP or LogD), hydrogen bonding capacity, and polar surface area (PSA) are critical.10 Guidelines like Lipinski's "Rule of Five" for drug candidates and the "Rule of Three" (Ro3: MW ≤ 300, cLogP ≤ 3, H-bond donors/acceptors ≤ 3) for fragments provide useful, albeit flexible, frameworks.10 Crucially, adequate solubility in both the stock solvent (typically DMSO) and the aqueous assay buffer at relevant screening concentrations is essential to avoid artifacts.10
Novelty: For organizations concerned with intellectual property, the novelty or originality of the chemical scaffolds within a library is an important consideration.10 Screening proprietary or unique compound collections can provide starting points free from existing patents, facilitating downstream development.10 Uniqueness can be assessed computationally against patent and chemical databases.
Freedom from Problematic Functionalities: Libraries must be rigorously filtered, either computationally during design or experimentally during QC, to remove compounds containing functionalities known to cause issues in biological assays.10 This includes PAINS structures 37, reactive groups that can cause non-specific covalent modification, redox cyclers 10, aggregators, and other frequent hitters that often lead to false positives.
The concept of library 'quality' itself has evolved significantly. Early efforts in the combinatorial chemistry era focused heavily on maximizing the sheer number of compounds screened, assuming quantity would lead to hits.10 Experience demonstrated that this approach often yielded poor-quality hits or numerous false positives. This led to a greater emphasis on 'drug-likeness' and 'lead-likeness,' incorporating physicochemical property filters and removing known problematic functionalities.10 Today, as researchers tackle increasingly challenging targets, including those previously considered 'undruggable' 10, the definition of quality continues to expand. It now encompasses factors like 3D complexity 10, suitability for specific screening modalities (e.g., fragments, DELs 20), and the application of advanced QC techniques like Next-Generation Sequencing (NGS) for display libraries.40 Thus, designing effective libraries requires moving beyond simplistic rules towards sophisticated, context-dependent principles tailored to the specific biological question and screening technology.
Common Pitfalls and Screening Artifacts: Failure to address the critical considerations above leads to common pitfalls:
Poor Solubility & Aggregation: Compounds exceeding their aqueous solubility limit under assay conditions can form colloidal aggregates, which non-specifically inhibit enzymes or interfere with assays, representing a major source of false positives.10 This is particularly problematic for flat, planar molecules often found in older libraries.20
PAINS and Frequent Hitters: Screening libraries contaminated with PAINS or other promiscuous compounds inevitably yields numerous false positives that waste time and resources in hit validation.10 These compounds appear active due to assay interference, reactivity, or other non-specific mechanisms rather than targeted binding.
Library Bias: The synthetic routes used to create libraries or the sources from which they are acquired can introduce structural biases, such as an over-representation of certain scaffolds or a lack of chemical diversity or 3D character. This limits the effective chemical space explored.
Compound Degradation/Purity Issues: Small molecules can degrade during long-term storage, particularly under suboptimal conditions (e.g., freeze-thaw cycles, moisture). Impurities present from synthesis can also confound results.10 Lack of proper QC can lead to screening degraded or impure samples, generating unreliable data.
Quality Control Strategies for Different Library Types: Rigorous QC is essential to ensure library integrity and the validity of screening results. The specific QC approaches vary depending on the library modality:
Small Molecule Libraries (HTS): Standard QC involves confirming compound identity, purity (typically >90-95%), and concentration, often using LC-MS or NMR on a representative subset.10 Assessing solubility in DMSO and assay buffer is critical. Computational filters for PAINS, reactivity, and undesirable physicochemical properties should be applied proactively during library design or acquisition.10 Periodic re-analysis is necessary to monitor stability during storage.
Fragment Libraries (FBDD): Due to the high concentrations used in screening (μM-mM range) and the reliance on sensitive biophysical detection methods, fragment libraries require exceptionally high purity and validated solubility.20 QC typically involves NMR, MS, and experimental solubility measurements. Physicochemical properties are tightly controlled, often adhering to the Ro3.20 Diversity and complexity are carefully curated to maximize chemical space coverage efficiently.20
Phage Display Libraries: QC focuses on assessing the key parameters that determine library utility: size (number of independent clones, determined by plating transformed bacteria or phage titration), diversity (variety of displayed peptides/proteins), and display efficiency (percentage of phage particles correctly displaying the fusion protein).43 Common challenges include low transformation/amplification yields, poor display levels, insufficient diversity, and contamination.43 Sequencing plays a critical role. Initially reliant on low-throughput Sanger sequencing of a small number of clones, library validation now heavily utilizes NGS.40 NGS provides a deep view of library diversity, allows quantitative monitoring of sequence enrichment during panning rounds, helps identify convergence motifs, and can uncover rare but potentially high-affinity binders missed by Sanger sequencing.40 A major pitfall in phage display is the enrichment of Target-Unrelated Peptides (TUPs) or propagation artifacts; NGS is invaluable for identifying and excluding these spurious hits.44 However, standard short-read NGS platforms can struggle to link the heavy and light chain variable regions (VH and VL) of displayed Fab fragments, requiring specialized methods.45
Yeast Display Libraries: Similar to phage display, QC involves determining library size (via transformation efficiency) and diversity (via sequencing, often NGS 41). A key aspect of yeast display QC and screening is the use of Fluorescence-Activated Cell Sorting (FACS).51 FACS allows for quantitative, single-cell analysis of both the level of protein expression on the yeast surface (often via a fused epitope tag like c-myc or HA) and the binding affinity to a fluorescently labeled target.51 This dual measurement helps normalize for expression differences and allows for precise selection of clones based on desired binding characteristics.53 Yeast possess a eukaryotic quality control system that can, in some cases, link display levels to protein stability, potentially selecting against misfolded or unstable variants.52 However, this correlation is not always robust, especially for highly stable parent proteins, and may not distinguish well-folded proteins from stable molten globules.58
DNA-Encoded Libraries (DELs): The quality of a DEL hinges on the reliability and compatibility of the chemical synthesis steps with the DNA tags, the diversity of the chemical building blocks used, and the expertise of the library designer.39 QC involves validating the integrity of the DNA tags and confirming the successful execution of the chemical coupling steps. A major challenge during screening is "screening noise"—the identification of hits that bind non-specifically to the target or selection matrix, or artifacts arising from the selection process itself.39 Affinity-based selection is common but can be prone to such artifacts and may not be suitable for all target types (e.g., membrane proteins).39 Analysis requires sophisticated bioinformatics and statistical methods to deconvolute the sequencing data from enriched pools and identify true binders from the background noise; machine learning approaches are being explored to aid this process.39 Selectivity can also be a pitfall, particularly when screening against members of large protein families (e.g., kinases). Strategies to address this include parallel screening against multiple family members followed by comparative informatics analysis to identify selective ligands.39
The implementation of advanced QC methods, especially NGS for display libraries, signifies a paradigm shift. QC is no longer just a terminal check but a powerful analytical tool integrated into the discovery process. Deep sequencing provides unprecedented insights into library composition and selection dynamics, enabling data-driven optimization of panning strategies, identification of biases, and rescue of valuable clones that would be missed by traditional low-throughput methods.40 Similarly, rigorous physicochemical QC for chemical libraries prevents wasted effort screening artifact-prone compounds.10 Investing in comprehensive QC upfront is therefore a critical risk-mitigation strategy that significantly enhances the probability of identifying high-quality, developable leads.11
While distinct library types offer different advantages, there is a convergence in the fundamental challenges faced (ensuring diversity, maintaining quality, avoiding artifacts) and the types of solutions being employed (cheminformatics, NGS, automation, sophisticated data analysis). Furthermore, successful drug discovery campaigns often integrate multiple library types and screening approaches. For example, phage display might be used for initial enrichment from a very large library, followed by transfer to yeast display for finer affinity discrimination and sorting using FACS.57 Fragment hits may serve as starting points for structure-based design or elaboration into larger, more potent molecules.20 Understanding the specific strengths, weaknesses, and QC requirements of each modality allows researchers to devise tailored, multi-pronged screening strategies that maximize the chances of success against the increasingly diverse and challenging landscape of biological targets.
Table 2: Comparison of Common Molecular Library Types
Feature | Small Molecule (HTS) | Fragment (FBDD) | Phage Display | Yeast Display | DEL (DNA-Encoded Library) |
Typical Size/Diversity | 10^5 - 10^7 compounds | 10^3 - 10^4 compounds | 10^9 - 10^12+ peptides/Abs | 10^7 - 10^10 peptides/Abs | 10^6 - 10^12+ compounds |
Key Advantages | Broad applicability, established infrastructure | High hit rates, efficient chemical space coverage, good starting points for optimization 20 | Very large libraries, robust selection (panning), established protocols 50 | Eukaryotic expression (folding, PTMs), quantitative screening (FACS), stability/expression correlation sometimes 52 | Extremely large libraries, miniaturized screening, no need for cellular expression |
Key Disadvantages/ Pitfalls | Lower hit rates, potential for PAINS/artifacts, requires large library quantities 3 | Weak affinities require sensitive biophysical assays, solubility challenges 20 | Prokaryotic expression limits complex proteins, TUPs/selection artifacts, VH/VL linking challenge 44 | Lower library size vs. phage, slower growth, potential expression bias 61 | Complex synthesis/QC, screening noise/artifacts, analysis complexity, target limitations (affinity selection) 39 |
Critical QC/Methods | Purity/Identity (LCMS, NMR), Solubility, PAINS/Property filters 10 | High Purity/Identity (NMR, MS), High Solubility, Ro3 compliance 20 | Titer, Diversity (NGS), Display Efficiency (ELISA/FACS), NGS for enrichment/TUPs 40 | Transformation Efficiency, Diversity (NGS), Expression Level (FACS), Binding (FACS) 51 | DNA tag integrity, Chemical synthesis validation, Sequencing/Informatics for deconvolution 39 |
Relevant Sources | 3 | 20 | 40 | 41 | 37 |
Impact of Library Quality on Hit Rates and Downstream Attrition: The quality of the starting library directly correlates with the success of the screening campaign and subsequent drug development efforts. Screening low-quality libraries—those lacking diversity, plagued by problematic compounds (PAINS, aggregators), possessing poor physicochemical properties, or suffering from inadequate QC—typically results in low hit rates, poor-quality hits that are difficult to optimize, or numerous false positives that lead researchers down unproductive paths.6 This significantly increases the time and cost associated with hit validation and lead optimization and contributes directly to the high attrition rates observed in preclinical and clinical development.6 Conversely, investing in the design, acquisition, and rigorous QC of high-quality libraries provides more promising and developable starting points, increasing the efficiency of hit-to-lead progression and ultimately improving the likelihood of bringing a successful drug to market.11
Mastering Protein Stability and Engineering for Therapeutic Success
The Central Role of Protein Stability: For therapeutic proteins, antibodies, and protein reagents used in drug discovery assays, stability is not merely a desirable characteristic but a fundamental requirement.12 Protein stability refers to the ability of a protein molecule to maintain its specific three-dimensional (native) conformation, which is essential for its biological function.64 This native state must be sufficiently stable relative to the unfolded state under physiological conditions and during manufacturing, storage, and administration, yet not so rigidly stable that it prevents necessary conformational changes integral to function.64 Instability can lead to loss of function, aggregation, and increased immunogenicity, posing significant challenges throughout the development lifecycle.14
Fundamental Principles Governing Protein Stability and Folding:
Thermodynamics: The stability of a protein's folded state (N) relative to its unfolded state (D) is governed by the Gibbs free energy change (ΔG) of unfolding.64 A negative ΔG under physiological conditions indicates the folded state is favored. Stability arises from a delicate balance of forces: stabilizing noncovalent interactions (hydrophobic effect, hydrogen bonds, van der Waals forces, electrostatic interactions/salt bridges) and covalent disulfide bonds counteract the large unfavorable loss of conformational entropy that occurs upon folding the flexible polypeptide chain into a defined structure.64
Folding Pathways & Misfolding: Protein folding is a complex process involving transient intermediates. Errors in folding (misfolding) can lead to non-functional states or expose aggregation-prone regions, potentially causing cellular dysfunction or disease.64
Common Challenges: Instability, Aggregation, Solubility, and Expression Issues: Therapeutic proteins and protein reagents face numerous stability challenges:
Mechanisms of Instability: Degradation can occur through physical or chemical pathways (See Table 3). Physical instability involves changes to the protein's higher-order structure (secondary, tertiary, quaternary) without breaking covalent bonds, including denaturation (unfolding), aggregation (formation of dimers or higher-order oligomers), precipitation (macroscopic aggregation), and adsorption to surfaces.12 Chemical instability involves the formation or breakage of covalent bonds, leading to modified protein species. Common chemical degradation routes include deamidation (of Asn, Gln), isomerization (of Asp), oxidation (of Met, Cys, His, Trp, Tyr), hydrolysis (peptide bond cleavage, especially at Asp residues), disulfide bond scrambling or formation, and glycation (reaction with sugars).12
Aggregation: This is a major concern for protein therapeutics and reagents.12 Aggregation often initiates from partially unfolded states where hydrophobic residues or specific Aggregation-Prone Regions (APRs), normally buried within the protein core, become solvent-exposed.68 These exposed regions can then interact intermolecularly, leading to the formation of soluble oligomers or insoluble aggregates/fibrils.68 Aggregation not only leads to loss of biological activity but also significantly increases the risk of inducing an unwanted immune response (immunogenicity) 14 and can reduce manufacturing yields.65
Solubility Issues: Many proteins, particularly when overexpressed in heterologous systems like E. coli or even when highly concentrated for formulation, exhibit poor solubility.70 Low solubility often correlates with conformational instability and a higher propensity to aggregate.13 Achieving high concentrations of soluble, stable protein is critical for therapeutic dosing and many biophysical assays.
Expression Challenges: Producing sufficient quantities of correctly folded, functional protein can be difficult, especially for complex mammalian proteins expressed in microbial hosts which may lack appropriate folding machinery or post-translational modification capabilities.70 Even in eukaryotic systems like yeast, expression levels can be limiting. Poor expression often correlates with low intrinsic stability of the protein.13 Specific expression contexts, like periplasmic expression required for phage display, can pose additional challenges for certain protein constructs.60
Environmental Stressors: Proteins are sensitive to their environment. Throughout their lifecycle—from production and purification to formulation, storage, shipping, and administration—they encounter various stresses that can compromise stability.12 These include temperature extremes (heating, freezing, freeze-thaw cycles), pH shifts (e.g., in degrading polymer matrices 12), mechanical stress (shear from pumping, agitation, filtration), exposure to interfaces (air-liquid, container surfaces 69), dehydrating conditions (lyophilization), exposure to organic solvents or process chemicals, light, and oxidizing agents.12
Table 3: Common Mechanisms of Protein Instability and Degradation
Category | Specific Mechanism | Brief Description | Key Triggers/Factors | Potential Consequences | Relevant Sources |
Physical | Denaturation/Unfolding | Loss of native secondary/tertiary/quaternary structure | Heat, pH extremes, chaotropes, shear stress, interfaces | Loss of activity, aggregation | 12 |
Aggregation | Association of protein molecules (oligomers, fibrils, amorphous aggregates) | Partial unfolding, high concentration, APR exposure, interfaces, stress (temp, shear) | Loss of activity, ↑ Immunogenicity, ↓ Yield, insolubility | 12 | |
Precipitation | Macroscopic formation of insoluble protein particles | Gross aggregation, changes in solubility (pH, ionic strength) | Loss of active protein, visible particulates | 12 | |
Adsorption | Binding of protein molecules to surfaces (containers, air-water interface) | Hydrophobicity, surface properties, concentration | Denaturation, aggregation trigger, loss of protein | 12 | |
Chemical | Deamidation | Hydrolysis of Asn/Gln side chain amide to carboxylic acid | Neutral/alkaline pH, temperature, specific sequences (e.g., Asn-Gly) | Charge change, conformational change, activity loss | 12 |
Isomerization | Conversion of Asp to isoAsp (often via succinimide intermediate) | pH, temperature, specific sequences (e.g., Asp-Gly, Asp-Ser) | Conformational change, activity loss | 12 | |
Oxidation | Modification of susceptible residues (Met, Cys, His, Trp, Tyr) by reactive oxygen | Exposure to oxygen, light (photooxidation), metal ions, peroxides (excipients) | Activity loss, conformational change, aggregation | 12 | |
Hydrolysis | Cleavage of peptide bonds in the backbone | Acidic pH, high temperature, specific sequences (e.g., Asp-Pro) | Fragmentation, loss of activity | 12 | |
Disulfide Scrambling/ Formation | Incorrect formation or breakage/reformation of disulfide bonds | Redox environment, pH, thiols, stress (e.g., lyophilization) | Misfolding, aggregation (intermolecular S-S), activity loss | 12 | |
Glycation | Non-enzymatic reaction of reducing sugars with amino groups (e.g., Lys) | Presence of reducing sugars, prolonged incubation | Altered structure/function/stability/affinity | 12 |
The Specter of Immunogenicity: Linking Instability to Adverse Effects: One of the most significant consequences of protein instability, particularly aggregation, is the potential to induce an unwanted immune response, or immunogenicity.14
Immunogenicity Defined: This refers to the propensity of a therapeutic protein to elicit the production of anti-drug antibodies (ADAs) in patients.65 These ADAs can be binding or neutralizing.
Consequences: ADA formation can have severe clinical consequences, ranging from reduced drug efficacy due to accelerated clearance or direct neutralization of the protein's activity, to serious adverse events such as hypersensitivity reactions, anaphylaxis, or even the neutralization of a patient's own endogenous protein counterpart (if cross-reactive), potentially leading to life-threatening deficiencies.65 Immunogenicity is therefore a major safety concern and a key reason for clinical trial failures.65
The Aggregation Link: There is substantial evidence indicating that aggregated forms of therapeutic proteins are significantly more immunogenic than their monomeric counterparts.14 Aggregates may present repetitive epitopes that can efficiently cross-link B cell receptors, potentially breaking immune tolerance, or they may be more readily taken up and processed by antigen-presenting cells (APCs), leading to T cell activation and subsequent ADA production.65 Even subtle structural changes or modifications resulting from instability can create new epitopes (neo-epitopes) recognized as foreign by the immune system.
Other Factors: While aggregation is a major product-related risk factor, other aspects also contribute to immunogenicity, including the presence of process-related impurities or contaminants, formulation components, the route and frequency of administration, and the inherent "foreignness" of the protein sequence.69 Patient-related factors like genetics (e.g., HLA type), underlying disease state, and concomitant medications (e.g., immunosuppressants) also play a role.72
The strong link between instability/aggregation and immunogenicity underscores the critical importance of controlling protein stability throughout development. An unstable protein carries a significantly higher risk of failing in the clinic due to immunogenicity-related safety or efficacy issues.14 Even candidates that perform well in preclinical models, which are often poor predictors of human immune responses 8, can unexpectedly elicit immunogenicity in patients. Therefore, rigorous assessment and proactive engineering for stability serve as crucial risk mitigation strategies.
Established and Cutting-Edge Protein Engineering Tactics: To address the challenges of instability, poor solubility, low expression, and suboptimal function, various protein engineering strategies are employed:
Goal: The overarching aim is to rationally modify the protein's amino acid sequence to enhance desired properties—such as thermal stability, chemical stability, solubility, expression yield, folding efficiency, catalytic activity, or binding affinity/specificity—without compromising (or while improving) its intended function.13
Rational Design: This approach leverages knowledge of protein structure-function relationships. Using structural data (from X-ray crystallography, NMR, or increasingly accurate computational models like AlphaFold) and computational tools (e.g., FoldX for stability prediction 68, Rosetta for design 78), specific mutations are designed to achieve a desired effect.79 Examples include introducing disulfide bonds to constrain flexibility, optimizing hydrophobic core packing, modifying surface charges to improve solubility or reduce aggregation, redesigning loops, or mutating residues within identified APRs to less aggregation-prone ones.68 While targeted, rational design requires pre-existing structural/functional information and the accuracy of predictive algorithms remains a challenge.80
Directed Evolution: This powerful set of techniques mimics natural evolution in the laboratory.79 It involves first generating genetic diversity by introducing random or semi-random mutations into the gene encoding the protein of interest (using methods like error-prone PCR or DNA shuffling 79) to create large libraries of protein variants (up to 10^14 members 61). These libraries are then subjected to high-throughput screening or selection methods that link the desired protein property (phenotype) to the encoding gene (genotype), allowing for the isolation and amplification of improved variants.79 This iterative process requires no prior structural knowledge but can be experimentally intensive. Key platforms include:
Phage Display: Widely used for evolving binders (antibodies, peptides).84 Advantages include very large library sizes and robust panning selection methods.50 However, the prokaryotic expression system may struggle with folding complex eukaryotic proteins or those requiring post-translational modifications, and periplasmic expression can be a bottleneck.60
Yeast Display: A versatile platform employing eukaryotic expression, suitable for complex proteins requiring proper folding and modifications like glycosylation.52 It enables quantitative screening using FACS, allowing precise selection based on affinity, specificity, expression level, and stability (e.g., by selecting cells that retain binding after heat shock or protease treatment).52 The correlation between surface display level and intrinsic stability/solubility provides an additional selection pressure 58, although its reliability can vary.58 Library sizes are typically smaller than phage display.61
Other Methods: Bacterial, ribosome, and mammalian display systems offer alternative contexts.61 Screening can also be performed in microtiter plates (MTPs) using enzymatic or binding assays, or via methods like compartmentalization in emulsions.79
Computational / AI-Driven Design: This rapidly advancing field utilizes sophisticated algorithms, including deep learning neural networks, trained on vast amounts of protein sequence, structure, and experimental data.70 Models like ProteinMPNN can design sequences predicted to fold into specific structures with high accuracy 70, while others like LigandMPNN can design binding sites for small molecules or metals.78 Tools like ProtSSN integrate sequence and structure information to better predict the effects of mutations on properties like thermostability.77 These computational approaches can accelerate de novo design, guide rational mutagenesis, or help design more effective libraries for directed evolution, increasingly bridging the gap between rational and evolutionary methods.
Other Strategies: Beyond direct sequence modification, stability and function can be modulated by: optimizing the protein's environment (medium engineering, e.g., buffer composition, pH, additives 76); altering substrate specificity (substrate engineering 76); fusing the protein to solubility-enhancing tags or partners 68; chemical modification (e.g., PEGylation, though PEG itself can be immunogenic 74); or immobilizing enzymes onto solid supports.76
Table 4: Overview of Protein Engineering Strategies for Enhanced Stability and Function
Strategy | Principle | Key Methods/Tools | Strengths | Limitations/Challenges | Typical Applications | Relevant Sources |
Rational Design | Structure/function knowledge guides specific mutations. | Site-directed mutagenesis, computational modeling (Rosetta, FoldX), structural analysis | Targeted, requires less screening, can address specific known issues. | Requires prior structure/function knowledge, prediction accuracy limited, may miss unexpected solutions. | Stability (Tm, chemical), solubility, activity, specificity, reduce APRs. | 68 |
Directed Evolution | Mimics natural selection; generate diversity & select improved variants. | Random/Semi-random mutagenesis (error-prone PCR, shuffling), library construction, HTS/Selection | No prior structure knowledge needed, explores vast sequence space, can find novel solutions. | Can be labor-intensive, screening limitations, may require multiple rounds. | Stability, solubility, expression, affinity maturation, altered specificity/activity. | 13 |
- Phage Display | Display on phage surface, select via panning. | Phage libraries, biopanning protocols. | Very large libraries (10^9+), robust selection for binders. | Prokaryotic expression limits complex proteins, TUPs/artifacts, VH/VL linking. | Antibody/peptide discovery, affinity maturation. | 50 |
- Yeast Display | Display on yeast surface, select via FACS. | Yeast libraries, FACS sorting, heat/protease selection. | Eukaryotic expression (folding/PTMs), quantitative sorting (affinity/expression), stability selection. | Smaller libraries vs. phage (10^7-10^10), slower growth. | Affinity maturation, stability/solubility/expression engineering, enzyme evolution. | 13 |
Computational / AI Design | Use algorithms/ML trained on data to design sequences or predict effects. | Deep learning models (ProteinMPNN, ProtSSN), energy functions, molecular dynamics. | Fast prediction/design, can explore novel space, integrates diverse data. | Model accuracy depends on training data, experimental validation still required. | De novo design, stability prediction, mutation effect prediction, library design guidance. | 70 |
Other Strategies | Modify environment, fuse partners, chemical modification, immobilization. | Buffer optimization, fusion tags (GST, MBP), PEGylation, enzyme immobilization protocols. | Can improve properties without sequence changes, useful for formulation/delivery. | May not alter intrinsic stability, tags need removal, PEG immunogenicity, immobilization can alter kinetics. | Solubility enhancement, formulation stability, extended half-life, biocatalysis. | 68 |
Strategies to Overcome the Stability-Function Trade-off: A common challenge in protein engineering is the stability-function trade-off: mutations introduced to enhance function (e.g., binding affinity) often destabilize the protein structure, potentially compromising its overall utility.13 This occurs because functional mutations deviate from the wild-type sequence, which is typically evolutionarily optimized for a balance of stability and function in its native context.13 Several strategies can mitigate this trade-off 13:
Start with Highly Stable Scaffolds: Using intrinsically stable parent proteins provides a "stability budget." These proteins can tolerate more destabilizing functional mutations before overall stability drops below an acceptable threshold.13 Alternatively, an existing protein with desired function but poor stability can first be stabilized through engineering before functional optimization is attempted.13
Minimize Destabilization During Engineering: Library design can be optimized to reduce the stability penalty of functional mutations. This includes focusing mutagenesis on more tolerant surface residues rather than the sensitive core, avoiding mutations at evolutionarily conserved positions, using computational predictions to guide library design, or constructing smaller, focused libraries to test mutational effects before large-scale screening.13 Crucially, selection strategies can be designed to co-select for both function and stability (or related properties like expression level or protease resistance) simultaneously, enriching for variants that balance both attributes.13
Repair Damaged Mutants: If a highly functional variant suffers from poor stability, subsequent rounds of stability engineering can be performed to introduce compensatory stabilizing mutations, using rational design, computation, or further directed evolution specifically selecting for stability.13
The interplay between rational design, computational modeling, and directed evolution is becoming increasingly synergistic. Computational tools now routinely inform both rational design choices and the construction of smarter libraries for directed evolution.13 High-throughput experimental data generated from directed evolution screens, especially deep mutational scanning, provides invaluable information for training and refining computational models.81 This iterative cycle between prediction, design, and experimental validation offers the most powerful approach to navigating the complex sequence-structure-function landscape and overcoming challenges like the stability-function trade-off.
Impact on Therapeutic Development Costs and Success Rates: Protein instability issues—manifesting as low expression yields, aggregation during manufacturing or storage, poor solubility requiring complex formulations, degradation leading to loss of potency, or triggering immunogenicity in patients—impose substantial burdens on therapeutic development.12 These problems lead to increased manufacturing costs (due to low yields or batch failures 65), complex and expensive formulation development, and, most significantly, higher rates of clinical attrition due to lack of efficacy or unacceptable safety profiles (often related to immunogenicity).12 Proactively addressing protein stability through rigorous assessment and engineering early in the discovery and development process is therefore a critical de-risking activity. Generating stable, well-behaved protein candidates significantly enhances the probability of successful manufacturing, formulation, and ultimately, clinical translation, thereby improving success rates and potentially reducing the enormous overall cost of drug development.14
Conclusion: Integrating Solutions for Accelerated and De-Risked Drug Discovery
The journey from initial concept to an approved therapeutic is fraught with challenges, characterized by high costs, long timelines, and significant attrition rates.2 This guide has explored three critical, interconnected domains in early drug discovery—High-Throughput Screening, Molecular Library Design, and Protein Stability/Engineering—highlighting the key hurdles and bottlenecks within each. Challenges in HTS, such as ensuring data quality, managing massive datasets, avoiding false positives, and overcoming workflow bottlenecks 15, directly impact the efficiency of hit identification. The quality, diversity, and developability of the molecular libraries being screened are fundamental determinants of finding viable starting points.10 For biologics and protein-based tools, inherent instability, aggregation propensity, expression difficulties, and the looming risk of immunogenicity represent major obstacles to successful development.12 These challenges are not independent silos; poor library quality leads to wasted HTS effort, and unstable protein reagents compromise assay reliability, ultimately contributing to the high overall failure rates in drug development.6
A recurring theme throughout this analysis is the paramount importance of addressing quality, stability, and potential failure points as early as possible in the discovery pipeline. Robust HTS assay validation 19, meticulous library design coupled with rigorous QC 10, and the early assessment and proactive engineering of protein stability 13 are not optional refinements but essential practices for de-risking projects. Improving the predictive validity of early-stage screening and disease models—ensuring that hits identified in vitro translate effectively downstream—is crucial for reducing costly late-stage failures.5 Stability, in particular, emerges as a multifaceted prerequisite, influencing everything from expression and solubility to aggregation and the critical clinical gatekeeper of immunogenicity.13
Fortunately, significant technological and methodological advancements offer powerful solutions to navigate these hurdles. In HTS, label-free technologies like mass spectrometry are mitigating assay interference 15, while acoustic liquid handling streamlines workflows and enables further miniaturization.32 Sophisticated data management platforms and the application of AI/ML are tackling the data deluge, improving QC, identifying artifacts, and prioritizing hits.3 For library development, NGS provides unprecedented depth for QC and analysis of display libraries 40, while cheminformatics guides the design of more diverse and developable chemical libraries.10 In protein engineering, the synergistic integration of computational design, rational approaches, and directed evolution platforms like yeast display allows for the efficient generation of proteins with enhanced stability, solubility, and function, helping to overcome the inherent stability-function trade-off.13
Ultimately, accelerating drug discovery and improving success rates requires an integrated approach. By strategically implementing best practices, embracing innovative technologies, and addressing the interconnected challenges across HTS, library design, and protein engineering, researchers can enhance the quality of candidates progressing through the pipeline. This focus on early-stage quality and de-risking holds the key to increasing R&D productivity and, most importantly, accelerating the delivery of safe and effective new medicines to patients.
Works cited
The High-Throughput Screening Transformation in Modern Drug Development, accessed April 16, 2025, https://www.pharmasalmanac.com/articles/the-high-throughput-screening-transformation-in-modern-drug-development
Biopharma R&D Faces Productivity And Attrition Challenges In 2025 - Clinical Leader, accessed April 16, 2025, https://www.clinicalleader.com/doc/biopharma-r-d-faces-productivity-and-attrition-challenges-in-2025-0001
AI-driven High Throughput Screening for Targeted Drug Discovery - Oxford Global, accessed April 16, 2025, https://oxfordglobal.com/discovery-development/resources/ai-driven-high-throughput-screening-for-targeted-drug-discovery
Dealing with the challenges of drug discovery | CAS, accessed April 16, 2025, https://www.cas.org/resources/cas-insights/dealing-challenges-drug-discovery
Why 90% of clinical drug development fails and how to improve it? - PMC - PubMed Central, accessed April 16, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9293739/
Drug Attrition in Check: Shifting Information Input to Where it Matters - HARMONY Alliance, accessed April 16, 2025, https://www.harmony-alliance.eu/webimages/files/PLS_DDD_WP_Attrition-Elsevier.pdf
Drug discovery and development: introduction to the general public and patient groups, accessed April 16, 2025, https://www.frontiersin.org/journals/drug-discovery/articles/10.3389/fddsv.2023.1201419/full
When Quality Beats Quantity: Decision Theory, Drug Discovery, and the Reproducibility Crisis | PLOS One, accessed April 16, 2025, https://journals.plos.org/plosone/article?id=10.1371/journal.pone.0147215
High-throughput screening (HTS) | BMG LABTECH, accessed April 16, 2025, https://www.bmglabtech.com/en/blog/high-throughput-screening/
Selecting, Acquiring, and Using Small Molecule Libraries for High ..., accessed April 16, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC4687755/
Hit Identification - Creative Biostucture Drug Discovery, accessed April 16, 2025, https://drug-discovery.creative-biostructure.com/hit-identification-p20
Strategies for overcoming protein and peptide instability in ..., accessed April 16, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC10526705/
Engineering Strategies to Overcome the Stability–Function Trade-Off ..., accessed April 16, 2025, https://pubs.acs.org/doi/10.1021/acssynbio.1c00512
Stabilization challenges and aggregation in protein-based therapeutics in the pharmaceutical industry - RSC Publishing, accessed April 16, 2025, https://pubs.rsc.org/en/content/articlehtml/2023/ra/d3ra06476j
Advances in high‐throughput mass spectrometry in drug discovery ..., accessed April 16, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9832828/
Impact of high-throughput screening in biomedical research - ResearchGate, accessed April 16, 2025, https://www.researchgate.net/publication/50227800_Impact_of_high-throughput_screening_in_biomedical_research
Reducing the Attrition Rate in Drug Discovery - SAE Media Group, accessed April 16, 2025, https://www.smgconferences.com/pharmaceuticals/uk/conference/reducing-the-attrition-rate-in-drug-discovery
High-throughput screening - Wikipedia, accessed April 16, 2025, https://en.wikipedia.org/wiki/High-throughput_screening
Principles of early drug discovery - PMC - PubMed Central, accessed April 16, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC3058157/
Fragment‐based drug discovery—the importance of high‐quality ..., accessed April 16, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9627785/
Challenges and the Advancement in High-Throughput Screening Strategies for Cancer Therapeutics - Preprints.org, accessed April 16, 2025, https://www.preprints.org/manuscript/202310.1708/v1
Accelerating Discovery and Development with Advances in High-Throughput Screening, accessed April 16, 2025, https://www.pharmtech.com/view/accelerating-discovery-and-development-with-advances-in-high-throughput-screening
Quality Control for Reliable HTS Assays - Selvita, accessed April 16, 2025, https://selvita.com/blog/screening-quality-controls-a-key-issue-for-robust-assay-execution/
What is Lab Automation - HighRes Biosolutions, accessed April 16, 2025, https://highresbio.com/blog/lab-automation/the-market-leaders-perspective-on-lab-automation
High attrition rate in drug development. | Download Scientific Diagram - ResearchGate, accessed April 16, 2025, https://www.researchgate.net/figure/High-attrition-rate-in-drug-development_fig1_6915002
The role of liquid handling technologies in successfully executing screening campaigns, accessed April 16, 2025, https://www.europeanpharmaceuticalreview.com/article/1172/the-role-of-liquid-handling-technologies-in-successfully-executing-screening-campaigns/
How High-throughput Labs Can Improve Data Management and Automation | - Scispot, accessed April 16, 2025, https://www.scispot.com/blog/how-high-throughput-labs-can-improve-data-management-and-automation
Solving Data Bottlenecks by Automating and Scaling Wet-Lab Data Analysis, accessed April 16, 2025, https://revvitysignals.com/blog/solving-data-bottlenecks-automating-and-scaling-wet-lab-data-analysis
Revolutionizing Drug Discovery: The Unmatched Impact of High-throughput Screening HTS in Pharmaceuticals - KanBo, accessed April 16, 2025, https://kanboapp.com/en/industries/pharmaceutical/revolutionizing-drug-discovery-the-unmatched-impact-of-high-throughput-screening-hts-in-pharmaceuticals/
Machine Learning Assisted Hit Prioritization for High Throughput Screening in Drug Discovery | ACS Central Science - ACS Publications, accessed April 16, 2025, https://pubs.acs.org/doi/10.1021/acscentsci.3c01517
Machine Learning-Driven Data Valuation for Optimizing High-Throughput Screening Pipelines | Journal of Chemical Information and Modeling - ACS Publications, accessed April 16, 2025, https://pubs.acs.org/doi/10.1021/acs.jcim.4c01547
How Improving the Efficiency of High Throughput Screening Assays with Mass Spectrometry Works | Lab Manager, accessed April 16, 2025, https://www.labmanager.com/how-improving-the-efficiency-of-high-throughput-screening-assays-with-mass-spectrometry-works-948
Benefits of Acoustic Liquid Handling in Drug Discovery - Drug Development and Delivery, accessed April 16, 2025, https://drug-dev.com/drug-discovery-benefits-of-acoustic-liquid-handling-in-drug-discovery/
Overcoming Screening Bottlenecks - Danaher Life Sciences, accessed April 16, 2025, https://lifesciences.danaher.com/us/en/blog/streamlining-screening.html
TWO PLATFORMS. ONE TEAM. - Beckman Coulter, accessed April 16, 2025, https://pages.beckman.com/rs/682-XWP-826/images/Beckman%20Drug%20Discovery%20Automation%20Brochure_CLD.pdf
Optimizing Compound and Plate Management - Drug Discovery Online, accessed April 16, 2025, https://www.drugdiscoveryonline.com/doc/optimizing-compound-and-plate-management-0001
The 2020 SLAS Discovery Top 10: Advancing the Science of Drug Discovery - PMC, accessed April 16, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC8940770/
Assay Guidance Workshop for High-Throughput Screening and Lead Discovery, accessed April 16, 2025, https://ncats.nih.gov/research/research-resources/agm/agm-workshops/series-2020
DNA-Encoded Libraries | proventainternational.com, accessed April 16, 2025, https://proventainternational.com/discovering-novel-chemical-entities-with-dna-encoded-libraries-promises-and-pitfalls/
How Next-Gen Sequencing Enhances Phage Display for Antibody Discovery, accessed April 16, 2025, https://www.creative-biolabs.com/phage-display-sequencing.html
Next-Generation Sequencing of Antibody Display Repertoires - Frontiers, accessed April 16, 2025, https://www.frontiersin.org/journals/immunology/articles/10.3389/fimmu.2018.00118/full
Deep sequencing of phage display libraries to support antibody discovery - ResearchGate, accessed April 16, 2025, https://www.researchgate.net/publication/236055062_Deep_sequencing_of_phage_display_libraries_to_support_antibody_discovery
Phage Display Library Construction: Comprehensive Guide - Creative Biolabs, accessed April 16, 2025, https://www.creative-biolabs.com/phage-display-library-construction-comprehensive-guide.html
Using NGS to Uncover the Corruption of a Peptide Phage Display Selection - MDPI, accessed April 16, 2025, https://www.mdpi.com/1467-3045/46/9/627
Next-generation sequencing-guided identification and reconstruction of antibody CDR combinations from phage selection outputs - Oxford Academic, accessed April 16, 2025, https://academic.oup.com/nar/article/47/9/e50/5373033
Phage Display Peptide Library Construction, accessed April 16, 2025, https://www.creative-peptides.com/services/phage-display-peptide-library-construction.html
Next-Generation Sequencing of a Single Domain Antibody Repertoire Reveals Quality of Phage Display Selected Candidates | PLOS One, accessed April 16, 2025, https://journals.plos.org/plosone/article?id=10.1371/journal.pone.0149393
Quality control of phage display or aptamer libraries - aptait, accessed April 16, 2025, https://centerforpublicpolicyanalysis.org/services/library-quality-control/
Antibody Libraries for In Vitro Display: What to Consider - GENEWIZ Blog, accessed April 16, 2025, https://blog.genewiz.com/antibody-libraries-for-in-vitro-display-what-to-consider
Full article: High-throughput retrieval of physical DNA for NGS-identifiable clones in phage display library - Taylor & Francis Online, accessed April 16, 2025, https://www.tandfonline.com/doi/full/10.1080/19420862.2019.1571878
Yeast Surface Display Library Screening Service - KMD Bioscience, accessed April 16, 2025, https://www.kmdbioscience.com/article/Yeast-Surface-Display-Library-Screening-Service.html
Yeast surface display for protein engineering and characterization - PMC - PubMed Central, accessed April 16, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC4038029/
Yeast Display based Binder Discovery Services - Creative Biolabs, accessed April 16, 2025, https://www.creative-biolabs.com/yeast-display-services.html
Yeast Display Platform - Creative Biolabs, accessed April 16, 2025, https://www.creative-biolabs.com/yeast-display-platform.html
Yeast Surface Display Platform for Rapid Selection of an Antibody Library via Sequential Counter Antigen Flow Cytometry - MDPI, accessed April 16, 2025, https://www.mdpi.com/2073-4468/11/4/61
Advancements in mammalian display technology for therapeutic antibody development and beyond: current landscape, challenges, and future prospects - Frontiers, accessed April 16, 2025, https://www.frontiersin.org/journals/immunology/articles/10.3389/fimmu.2024.1469329/full
Full article: A next-generation Fab library platform directly yielding drug-like antibodies with high affinity, diversity, and developability, accessed April 16, 2025, https://www.tandfonline.com/doi/full/10.1080/19420862.2024.2394230
Limitations of yeast surface display in engineering proteins of high thermostability - Oxford Academic, accessed April 16, 2025, https://academic.oup.com/peds/article/19/5/211/1556411
A next-generation Fab library platform directly yielding drug-like antibodies with high affinity, di - Carterra, accessed April 16, 2025, https://carterra-bio.com/wp-content/uploads/2024/09/A-next-generation-Fab-library-platform-directly-yielding-drug-like-antibodies-with-high-affinity-diversity-and-developability-1.pdf
Challenges of γ9δ2TCR affinity maturation when using phage display - bioRxiv, accessed April 16, 2025, https://www.biorxiv.org/content/10.1101/2021.05.20.445024v1.full.pdf
Applications of yeast surface display for protein engineering - PMC, accessed April 16, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC4544684/
Library QC for ATAC-Seq and CUT&Tag - Active Motif, accessed April 16, 2025, https://www.activemotif.com/blog-library-qc
Phage and Yeast Display | Microbiology Spectrum - ASM Journals, accessed April 16, 2025, https://journals.asm.org/doi/10.1128/microbiolspec.aid-0028-2014
Protein stability [determination] problems - Frontiers, accessed April 16, 2025, https://www.frontiersin.org/journals/molecular-biosciences/articles/10.3389/fmolb.2022.880358/full
Immunogenicity of therapeutic proteins: Influence of aggregation - PMC - PubMed Central, accessed April 16, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC4002659/
Aggregation of protein therapeutics enhances their immunogenicity: causes and mitigation strategies - PMC - PubMed Central, accessed April 16, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC8341748/
(PDF) Development of therapeutic proteins: Advances and challenges - ResearchGate, accessed April 16, 2025, https://www.researchgate.net/publication/274458143_Development_of_therapeutic_proteins_Advances_and_challenges
Solubis: a webserver to reduce protein aggregation through ..., accessed April 16, 2025, https://academic.oup.com/peds/article/29/8/285/2223259
Immunogenicity of therapeutic proteins. Part 2: Impact of container closures - ResearchGate, accessed April 16, 2025, https://www.researchgate.net/publication/6470137_Immunogenicity_of_therapeutic_proteins_Part_2_Impact_of_container_closures
Improving Protein Expression, Stability, and Function with ProteinMPNN | Journal of the American Chemical Society, accessed April 16, 2025, https://pubs.acs.org/doi/10.1021/jacs.3c10941
Genetic screens and directed evolution for protein solubility - General Biosystems, accessed April 16, 2025, https://www.generalbiosystems.com/uploadfiles/file/201504/Genetic%20screens%20and%20directed%20evolution%20for%20protein%20solubility.pdf
draft guideline on immunogenicity assessment of biotechnology-derived therapeutic proteins - EMA, accessed April 16, 2025, https://www.ema.europa.eu/en/documents/scientific-guideline/draft-guideline-immunogenicity-assessment-biotechnology-derived-therapeutic-proteins-first-version_en.pdf
T-Cell Dependent Immunogenicity of Protein Therapeutics Pre-clinical Assessment and Mitigation–Updated Consensus and Review 2020 - Frontiers, accessed April 16, 2025, https://www.frontiersin.org/journals/immunology/articles/10.3389/fimmu.2020.01301/full
Zwitterionic gel encapsulation promotes protein stability, enhances pharmacokinetics, and reduces immunogenicity | PNAS, accessed April 16, 2025, https://www.pnas.org/doi/10.1073/pnas.1512465112
Immunogenicity Assessment for Therapeutic Protein Products - FDA, accessed April 16, 2025, https://www.fda.gov/media/85017/download
Protein engineering for natural product biosynthesis: expanding diversity for therapeutic applications | Journal of Bio-X Research - MedNexus, accessed April 16, 2025, https://mednexus.org/doi/10.1097/JBR.0000000000000141
Semantical and Geometrical Protein Encoding Toward Enhanced Bioactivity and Thermostability - eLife, accessed April 16, 2025, https://elifesciences.org/reviewed-preprints/98033v1
Publications - Baker Lab, accessed April 16, 2025, https://www.bakerlab.org/publications/
Directed Evolution - Sigma-Aldrich, accessed April 16, 2025, https://www.sigmaaldrich.com/US/en/technical-documents/technical-article/protein-biology/protein-expression/directed-evolution
Limitations and challenges in protein stability prediction upon genome variations: towards future applications in precision medicine - PubMed, accessed April 16, 2025, https://pubmed.ncbi.nlm.nih.gov/32774791/
Protein stability engineering insights revealed by domain-wide comprehensive mutagenesis | PNAS, accessed April 16, 2025, https://www.pnas.org/doi/10.1073/pnas.1903888116
Laboratory-Directed Protein Evolution | Microbiology and Molecular Biology Reviews, accessed April 16, 2025, https://journals.asm.org/doi/abs/10.1128/mmbr.69.3.373-392.2005
Library methods for structural biology of challenging proteins and their complexes - PMC, accessed April 16, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC4172339/
Directed evolution of proteins for increased stability and expression using yeast display | Request PDF - ResearchGate, accessed April 16, 2025, https://www.researchgate.net/publication/224938695_Directed_evolution_of_proteins_for_increased_stability_and_expression_using_yeast_display
Directed Evolution of Protein Thermal Stability Using Yeast Surface Display - ResearchGate, accessed April 16, 2025, https://www.researchgate.net/publication/314203688_Directed_Evolution_of_Protein_Thermal_Stability_Using_Yeast_Surface_Display
Directed Evolution of Protein Thermal Stability Using Yeast Surface Display, accessed April 16, 2025, https://experiments.springernature.com/articles/10.1007/978-1-4939-6857-2_4
Prediction of Residue-specific Contributions to Binding and Thermal Stability Using Yeast Surface Display - Frontiers, accessed April 16, 2025, https://www.frontiersin.org/journals/molecular-biosciences/articles/10.3389/fmolb.2021.800819/full
Directed Evolution of Protein Thermal Stability Using Yeast Surface Display - PubMed, accessed April 16, 2025, https://pubmed.ncbi.nlm.nih.gov/28255874/
A Protein-Engineered, Enhanced Yeast Display Platform for Rapid Evolution of Challenging Targets - ACS Publications, accessed April 16, 2025, https://pubs.acs.org/doi/10.1021/acssynbio.1c00395
The R&D Cost of a New Medicine - Office of Health Economics, accessed April 16, 2025, https://www.ohe.org/wp-content/uploads/2014/07/380-RD-Cost-NME-Mestre-Ferrandiz-2012.pdf
コメント