Standard Operating Procedure (SOP) for High-Throughput Screening (HTS) in Drug Discovery
1) Purpose
The purpose of this Standard Operating Procedure (SOP) is to describe the process for conducting High-Throughput Screening (HTS) in drug discovery. HTS is a crucial technique used to rapidly test large numbers of compounds against a biological target to identify those that exhibit desirable biological activity. This SOP ensures that HTS is conducted efficiently, reproducibly, and in compliance with regulatory guidelines, leading to the identification of promising lead compounds for further drug development.
2) Scope
This SOP covers the entire process of HTS, from compound library preparation and assay design to data collection, analysis, and hit identification. It is applicable to all teams involved in HTS within the drug discovery process, including screening scientists, laboratory technicians, data analysts, and project managers. This SOP applies to HTS conducted in both academic and commercial settings for the identification of potential drug candidates across various therapeutic areas, such as oncology, infectious diseases, and neurodegenerative disorders.
3) Responsibilities
- Screening Scientists: Responsible for designing the HTS assays, selecting appropriate biological targets, and ensuring that the assays are optimized for high-throughput applications. They also ensure that the screening process is conducted with
4) Procedure
The following steps outline the detailed procedure for conducting HTS in drug discovery:
- Step 1: Compound Library Preparation
- Prepare a compound library that includes a diverse set of small molecules. The library can include commercially available compounds, in-house collections, or natural product libraries.
- Ensure that each compound in the library is well-characterized, including information on chemical structure, purity, and concentration. Maintain a database for easy tracking of compounds during the screening process.
- Prepare compound plates or solutions in the required concentrations for screening, ensuring that the compounds are aliquoted correctly to prevent cross-contamination between samples.
- Step 2: Assay Design and Optimization
- Design assays that are suitable for high-throughput screening. This may involve selecting an appropriate biological target (e.g., enzyme, receptor, or protein) and determining the assay format (e.g., fluorescence-based, luminescence-based, or cell-based assays).
- Optimize the assay conditions to ensure that the biological target is active and responsive to compound treatment. This includes determining the optimal assay concentration, incubation time, temperature, and buffer conditions.
- Ensure that the assay is robust and reproducible, with a low coefficient of variation (CV) and high Z-factor, which indicates the assay’s ability to discriminate between positive and negative controls.
- Step 3: HTS Setup
- Set up automated screening systems to conduct the HTS efficiently. This may include robotic liquid handling systems, plate readers, and other instrumentation that can handle large numbers of samples simultaneously.
- Load the compound library into the automated screening system, ensuring proper plate formatting and adherence to screening protocols.
- Run positive and negative controls in parallel with the compound library to ensure the accuracy and reliability of the screening results.
- Step 4: Screening and Data Collection
- Run the HTS assays with the compound library and controls. Monitor assay reactions in real-time, depending on the assay format used (e.g., fluorescence intensity, luminescence, or cellular changes).
- Ensure that data collection is automated and that the results are recorded and stored in a central database for subsequent analysis.
- Perform quality control checks during the screening process to identify and address any issues with assay performance or compound contamination.
- Step 5: Data Analysis and Hit Identification
- Use statistical tools and software to analyze the screening data. Identify “hits” based on criteria such as dose-response relationships, statistical significance, and consistency of activity.
- Use algorithms to prioritize compounds with the most promising activity, taking into account their potency, selectivity, and chemical properties.
- Generate dose-response curves for the identified hits to calculate EC50 or IC50 values and assess their potential for further development.
- Step 6: Hit Validation and Secondary Screening
- Validate the hits identified in the initial HTS by performing secondary assays to confirm their activity and specificity. This may include orthogonal assays or assays using different biological systems to confirm the target engagement.
- Screen the hits for off-target effects and cytotoxicity to ensure that the compounds do not have unintended biological effects.
- Prioritize the most promising hits for structure-activity relationship (SAR) analysis and optimization.
- Step 7: Documentation and Reporting
- Document all HTS procedures, including assay protocols, screening results, data analysis methods, and hit identification criteria.
- Prepare an HTS Report that includes detailed information on the screening process, data analysis, and a list of validated hits for further development.
- Ensure that all data is recorded accurately and maintained for future reference and regulatory compliance.
5) Abbreviations
- HTS: High-Throughput Screening
- EC50: Half-Maximal Effective Concentration
- IC50: Half-Maximal Inhibitory Concentration
- Z-factor: A statistical parameter for assay quality
- Z’-factor: A variation of the Z-factor used in HTS
6) Documents
The following documents should be maintained throughout the HTS process:
- HTS Screening Report
- Assay Protocols
- Screening Data Sheets
- Secondary Screening Results
7) Reference
References to regulatory guidelines and scientific literature that support this SOP:
- FDA Guidance for Industry on Drug Discovery
- ICH E6: Good Clinical Practice
- PubMed and PubChem for compound and assay information
8) SOP Version
Version 1.0: Initial version of the SOP.