Introduction to Assay Development

Similar documents
Z -LYTE fluorescent kinase assay technology. Z -LYTE Kinase Assay Platform

Use of a Label-Free Platform in a preclinical Contract Research Organization Environment

Fragment-based screening of enzyme drug targets: Microfluidic mobility shift assay improves confidence in candidate selection

Advanced Therapeutic Antibody Discovery with Multiplexed Screening

OriGene GFC-Arrays for High-throughput Overexpression Screening of Human Gene Phenotypes

HIGH-THROUGHPUT & PHENOTYPIC SCREENING

Recent years have witnessed an expansion in the disciplines encompassing drug

BioFarma USEF. Expertise in drug discovery projects

A Comparison of PerkinElmer s LANCE camp Assay Performance to that of State of the Art Competitive Technologies

IMPORTANCE OF CELL CULTURE MEDIA TO THE BIOPHARMACEUTICAL INDUSTRY

System. Dynamic Monitoring of Receptor Tyrosine Kinase Activation in Living Cells. Application Note No. 4 / March

Development and Utilization of Bromodomain and Extra Terminal Domain (BET) Assays Using AlphaLISA Technology

Propelling GPCR drug discovery through innovative products and services Be the change the market seeks

The Five Key Elements of a Successful Metabolomics Study

Agilent RapidFire 365 High-throughput Mass Spectrometry System ACCELERATING PHARMACEUTICAL DRUG DISCOVERY

An Automated, Cell-based Platform for the Rapid Detection of Novel Androgen Receptor Modulators

IROA Metabolic Profiling Kits FAQ

Dosing for Controlled Exposure (DoCE): Dosing strategies for characterising in vitro dose-responses with increased relevance for in vivo extrapolation

Rapid, Accurate and Flexible DNA Quantitation Using the QuantiFluor dsdna System on the MANTIS Liquid Handler

BoTest A/E Botulinum Neurotoxin Detection Kit- Detailed Description

Innovative Pharmaceutical Solutions for Discovery Chemistry, Biology and cgmp Manufacturing

Progress and Future Directions in Integrated Systems Toxicology. Mary McBride Agilent Technologies

Z -LYTE KINASE ASSAY KIT TYR 3 PEPTIDE PROTOCOL

Comparison of LANCE Ultra TR-FRET to PerkinElmer s Classical LANCE TR-FRET Platform for Kinase Applications

How Targets Are Chosen. Chris Wayman 12 th April 2012

Evaluating Microplate Detection Instruments

The Comprehensive Report

Your bridge to. better medicines

Cellular Assays. A Strategic Market Analysis. Sample Slides

FAR RED FP. GDP FP Assay. Technical Manual. v071814

Research. Image FlashPlate

Kinase Activity: a total solution

Label-free, real-time live-cell assays for spheroids: IncuCyte bright-field analysis

PI3 Kinase Assay Kit. Catalog Number KA assays Version: 05. Intended for research use only.

Cell Health and Viability Assays Real-time automated measurements of cell health and viability inside your incubator

Innovative Solutions for Drug Discovery and Life Sciences Research

SensoLyte AMC Calpain Activity Assay Kit *Fluorimetric*

Z -LYTE KINASE ASSAY KIT SER/THR 9 PEPTIDE PROTOCOL

Flow CAST : Testing Potency and Efficacy of Inhibitors of PI3K δ, PI3Kγ, BTK and SYK Activity

+ M III. IMAP Screening Express Kit Product #8073 Quantity: 8000, 20 µl reactions (80 µl final volumes) Low FP. M III High FP.

Impact of Collaborative Robots at The Genomics Institute of the Novartis Research Foundation (GNF) Dan Sipes Novartis Foundation

Break the 3D barrier CORNING 3D CELL CULTURE

Capabilities & Services

Synergy NEO HTS Multi-Mode Microplate Reader and Life Technologies Reagents

Technical Manual. Transcreener KINASE TR-FRET Assay

Technical Manual. Transcreener KINASE Assay Transcreener KINASE Plus Assay

Antibody Discovery at Evotec

Proteomics. Manickam Sugumaran. Department of Biology University of Massachusetts Boston, MA 02125

FlashPlate File #13. A Homogeneous Protein Kinase Assay Performed on 384-Well Streptavidin-Coated FlashPlate HTS PLUS. High Throughput Screening

Applying Affimers. Dr Amanda Nicholl at Avacta Life Sciences. Improving Antibody PK Assay Development

MicroRNA profiling directly from low amounts of plasma or serum using the Multiplex Circulating mirna Assay with Firefly particle technology

How To Choose a GeneCopoeia Luciferase System. Ed Davis, Ph.D.

FAR RED FP. UDP FP Assay. Technical Manual. v042809

Cell and gene therapy: scaling up and moving to mass production

ICSB Workshop: Drug Response Measurement and Analysis Part 2: Best practices for experimental design, execution, and analysis

Measurement by Spark 20M / Spark 10M Multimode microplate reader

Technical tips for ELISA and multiplex

ELISA. An introduction to the basic principles and assay formats. Innova Biosciences ltd All rights reserved

SPHK1 Assay Kit. Catalog Number KA assays Version: 05. Intended for research use only.

Data quality, variability and reproducibility

ATP Assay Kit. Catalog Number KA assays Version: 04. Intended for research use only.

Building DNA Libraries to Explore the Combinatorial Design Space. Dr. Yifan Li Senior Scientist GenScript USA Inc.

Proudly serving laboratories worldwide since 1979 CALL for Refurbished & Certified Lab Equipment

Combing molecular dynamics and machine learning for advanced pose and free-energy prediction

SensoLyte 620 HCV Protease Assay Kit *Fluorimetric*

Cell Processing Workstation. In Situ Laser-Mediated Cell Purification and Processing

High Throughput Quantitation of Cytokine Biomarkers using LANCE Ultra TR-FRET Assays

Introduction to Drug Design and Discovery

Characterization of Small Molecule to Protein Binding

SensoLyte 520 Factor Xa Assay Kit *Fluorimetric*

In Vitro Pharmacology Services

MATERIAL DATA SHEET. Reagents Provided in Kit

This Technical Note describes these characterization studies in more detail.

Lactate Assay Kit. Catalog Number KA assays Version: 08. Intended for research use only.

autoflex max Practical Power, Maximum Utility Innovation with Integrity MALDI-TOF MS

Application Note AN001

Stem Cell Services. Driving Innovation for Stem Cell Researchers

Practical Workshop: Chemical Biology, Drug Discovery and Screening

3D Transfection System

BECAUSE CAN T WAIT BREAKTHROUGHS. Drug Discovery Screening Solutions SCREENING SOLUTIONS. Automated Liquid Handling. Assays and Reagents

Proudly serving laboratories worldwide since 1979 CALL for Refurbished & Certified Lab Equipment QTRAP 5500

Ultra High Throughput Drug Discovery Screening by MALDI-TOF Mass Spectrometry powered by Simultaneous 1536 well Pipetting

WHEN MULTIMODE MEETS IMAGING YOU GAIN A WHOLE NEW PERSPECTIVE. EnSight Multimode Plate Reader

Changes to a Potency Bioassay for Biotechnology Products: a Regulatory Perspective Kathleen A. Clouse, Ph.D., Director Division of Monoclonal Antibodi

Miniaturized Whole Cell-based GPCR camp Assay Using a Novel Detection System

Understanding life WITH NEXT GENERATION PROTEOMICS SOLUTIONS

Time-resolved interaction analysis on living cells: Comprehensive data generates new knowledge and better decisions

Use of Echo Liquid Handlers to Improve the Efficiency and Reliability of Bioassay Laboratories

Optimizing Dual-Glo Luciferase Assays with the Synergy HT Multi-Detection Microplate Reader

DETERMINATION OF PROTEIN BINDING BY UPLC/MS/MS

SensoLyte 440 West Nile Virus Protease Assay Kit *Fluorimetric*

QS S Assist STK_FP Kit

Rapid Extraction of Therapeutic Oligonucleotides from Primary Tissues for LC/ MS Analysis Using Clarity OTX, an Oligonucleotide Extraction Cartridge

QImaging Camera Application Notes Multicolor Immunofluorescence Imaging

Pre-made Reporter Lentivirus for p53 Signal Pathway

Call for Pilot Project Applications Chemical Biology for Infectious Diseases NIH Center of Biomedical Research Excellence University of Kansas

LyticBLAzer -FRET B/G Kit Protocol

Peptide libraries: applications, design options and considerations. Laura Geuss, PhD May 5, 2015, 2:00-3:00 pm EST

Transcription:

Introduction to Assay Development A poorly designed assay can derail a drug discovery program before it gets off the ground. While traditional bench-top assays are suitable for basic research and target discovery, high throughput screening (HTS) assays need to meet stringent requirements. Careful consideration should go into the design, optimization, and validation of any novel assay developed to screen compounds to reliably identify hits. Assays should (1) be relevant to the disease biology; (2) produce reproducible results within the assay (using replicates) and between days and operators; (3) perform in a robust manner, which refers to the capacity of an assay to be unaffected by slight, but deliberate variations in parameters such as ph, incubation time, and different lots of reagents; and (4) have sufficient sensitivity to distinguish relatively weak hits from background with enough specificity to only detect compounds with the desired activity. This guide provides a high level overview of some of the most important factors to consider when designing an assay, with an emphasis on small molecule, high throughput screening programs. Before you begin Before embarking on a potentially long and difficult assay development process, consider the questions below. Has your target (molecule, pathway, or phenotype) been prosecuted previously? o Identifying a readily available assay can save a large amount of time, money and effort. Will a target-based or phenotypic approach better suit your program? o This depends on what is known about the target or pathway of interest and the goals of the drug discovery program. Can the assay be developed with the expertise currently on hand? o Academic centers with screening core facilities and contract research organizations (CROs) can work with investigators to design and develop the appropriate assay. HTS Requirements Compound libraries can contain hundreds of thousands, or even millions of compounds. With miniaturization, robotic platforms, and automated liquid handling, HTS can test more than 10,000 compounds per day. However, not all screens are amenable to HTS. In some cases, it may be appropriate to use a less high throughput assay to preserve biological relevance of the system. Below is a checklist to determine whether your assay can be scaled up to an HTS format. The assay can be performed in 10 or less automated steps and ideally without wash or reagent removal operations. If wash/removal is required it should be kept to a minimum, if possible.

The total assay volume is less than 200µl for 96-well, 25µl for 384-well, or 10µl for 1536-well plate formats. All reagents are stable at room temperature for at least the time required for the assay. Assay reactions are stable for long enough to measure the final readout for the entire plate or series of plates to minimize intra- and inter-plate variation. Assay reagents are stable in storage for long periods of time and available in large batches of consistent quality. Fundamentals for Successful Assays Vehicles: Different vehicles, like dimethyl sulfoxide (DMSO), are often used in chemical libraries to dissolve compounds with poor solubility in water. The vehicles themselves can affect assay performance, especially in cell-based assays, and should be titrated using different vehicle concentrations to determine the sensitivity of the assay to a particular vehicle. Generally less than or equal to 0.1% DMSO is acceptable. The vehicle concentration needs to be the same in all samples and controls. Controls: Within-plate and assay-wide controls are used to evaluate quality, variability, and detect sources of error throughout the assay system. The most appropriate negative and positive controls will depend on your target, desired activity of compounds, and other factors specific to your assay. The best positive control is often a compound with the activity that you are targeting. Statistical Analysis Plan: As with all experiments, the statistical analysis plan should be carefully designed before validating the assay and performing the screen. Dynamic Range: This is the range of activity of compounds that can be accurately measured by the assay. It is defined as the range between the means of the negative (or background) and positive controls, and will be affected by nearly every component of the assay. If the dynamic range is too narrow, potential active compounds may not be detected, thus reducing the chance of finding novel, chemical modulators for your target. Final Format: If you intend to scale up an assay, it is imperative to test assay performance under the final conditions that will be used in the large-scale screen. Assay reagents and cells can react differently in miniaturized plate formats and liquid handling systems. For longer, multi-plate runs, the effect of timing and order of reagent addition should be determined. Assay Design Considerations Here we list some of the most important factors to consider when designing your screen. These points highlight some of the key considerations for the most common types of assays. Comprehensive guidance for a wide variety of assays and targets can

be found in the Assay Guidance Manual [1]. Biochemical/cell-free assays Consider how your drug target performs under physiological conditions. Biological targets often act with native binding partners or are part of multimeric complexes, and normally function in specialized cellular microenvironments (at a specific ph, salt concentration, etc). Replicating these conditions in vitro can be challenging, but will yield more biologically relevant hits. In some cases, specialized and alternate forms of the target, such as truncated forms of large proteins, may need to be used. However, the more a target is altered from its natural form, the less relevant identified hits may be to the in vivo target. Only use highly pure, high-quality preparations of peptides, proteins, cofactors, and enzyme substrates that produce consistent activity. If a single lot is not available, make sure you test each new lot for activity. The concentrations of targets, cofactors, and substrates need to be carefully titrated. Balancing the concentrations of these factors and the test compounds with the kinetic properties of the target is critical to identifying legitimate hits. For enzymatic targets, different proportions of enzyme versus substrate concentrations can result in the identification of modulators with different modes of action. Cell-based assays Select the type of cells for your assay based on disease relevance, availability and ease of handling. Immortalized cell lines are relatively inexpensive, widely available, and if properly maintained, highly reproducible between lots. However, they may not accurately reflect the in vivo behavior of their progenitors due to changes involved during immortalization and passaging. Primary cells are isolated directly from animal or human tissue and have similar molecular, physiological, and morphological characteristics as cells in vivo. Primary cells are more difficult to obtain in large amounts on a consistent basis, may suffer from donor-to-donor variability, and can be more difficult to maintain. This is offset with the advantage of a more physiologically relevant system and avoiding overexpression of your target. Induced pluripotent stem cells (ipscs) are cells isolated from human donors that have been reprogrammed to a pluripotent state and can be differentiated into a wide variety of cell types. ipscs are a valuable tool for testing compounds on human cells that are difficult to isolate, such as neurons, but protocols for differentiation and maintenance vary widely, and the process can be costly and time consuming. Conversely, disease phenotypes in these cells are likely to better mimic the human condition.

Assay formats and detection systems Determine whether your assay will measure activity continuously, at multiple time-points, or at a single pre-determined endpoint. Establish the desired outcome of the assay (ligand binding, kinase activity, ion channel opening, nucleic acid degradation, etc.) based on the specific drug target, reporter and detection systems available. Select an appropriate reporter (if necessary) and detection system. The type of detection system can greatly impact critical assay parameters, including sensitivity, potential for compound interference, and dynamic range. o Indirect detection methods (i.e. absorbance, fluorescence, bioluminescence, etc.) use reporter systems and are ideal for larger scale screens due to their ease of use. o Direct detection methods, which use label-free systems, are increasingly being used. Label-free systems utilize biophysical and other measurements to determine compound activity in screens, avoiding many confounding factors that can occur with the use of reporter/indirect systems. o More quantitative and sensitive analytical methods, such as liquid chromatography mass spectrometry (LC-MS), electrophysiology, and RNAseq can be used if screens are to be run on a smaller-scale. Assay Optimization After establishing the basic format and protocol, it is important to experimentally optimize your assay. Although extensive optimization can be costly and time consuming, it is an essential step in ensuring the assay has sufficient sensitivity and specificity. Nearly every component of the assay can be adjusted individually. As outlined above for biochemical assays, target, substrate, cofactor, and test compound concentrations can all be adjusted to optimize the performance of the assay. In cell-based assays, factors including cell density, media composition, and others should be monitored to determine how they affect assay performance. For example, the presence of serum can effectively reduce your actual compound concentration due to small molecule binding to serum components. The more variables an assay has, the less efficient it becomes to optimize each one by one. Therefore, you should identify the most impactful factors to adjust. Statistical Design of Experiments is an iterative process that uses measurements of variation between replicate experiments to quickly and efficiently identify the most crucial assay components to optimize. It can also be used to identify interactions between adjustments to different components, while minimizing the number of experiments that need to be performed. [2]

Assay Validation Screening assays for drug discovery need to be rigorously validated. Experimental validation can determine the robustness and reproducibility of your assay. These essential parameters will provide confidence around identifying valid actives and hits during your full-scale screening campaign. Running validation experiments Validation efforts should be performed in replicate experiments across multiple days The same equipment, reagents, and experimental conditions that will be used for the full-scale screening campaign should be used during final validation testing. Use measures of statistical effect size to predict assay performance. The gold standards are the Z and Z scores, which are unit-less scales ranging from 0 to 1. Generally, Z scores of 0.5 or greater are indicative of high quality assays. o The Z score allows you to perform initial validation using only controls and vehicles. If available, use compounds with known activities against your target, as they may give further information on compound performance in the assay. o The Z score provides further validation using a representative subset of your compound library. It takes into account the dynamic range (the range between the means of the negative (or background) and positive controls) and the amount of variation present in both samples and noise (the standard deviations). Z score calculations Note: σ=standard deviation µ=mean s=signal c=control To calculate the Z score the means and standard deviations of the positive (c+) and negative (c-) controls are used. The Z score is calculated using the signal and background (or negative control) means and the standard deviations of both samples and background (or negative control). Assay validation is an iterative process. If validation testing yields a low Z-score, further assay optimization may be warranted. One or more of the assay conditions may need to be adjusted in order to improve assay performance in subsequent validation tests.

References: 1. Sittampalm G.S, et. al., Assay Guidance Manual. Eli Lilly and NCATS, 2016, Jul 1. 2. Altekar, M. et. al., Assay Optimization: A Statistical Design of Experiments Approach. Clin Lab Med, 2007, 27: 139-154. 3. Zhang J, et. al., A Simple Statistical Parameter For use in the Evaluation and Validation of High Throughput Screening Assays. J Biomolecular Screening, 1999, (4)2: 67-73. 4. Acker G.A., Auld D.S., Considerations for the design and reporting of enzyme assays in high-throughput screening applications. Pers in Sci 2014 (1)1-6: 56-73. 5. Inglese J, et. al. High Throughput Screening Assays for the identification of Chemical Probes. Nat Chem Biol, 2007, (3)8: 466-479. 6. Chen T., A Practical Guide to Assay Development and High-Throughput Screening in Drug Discovery. CRC Press, 2009, Dec 21.