Label-Free Detection of Compound Aggregation Using Corning Epic Technology

Similar documents
Application Note. Authors. Introduction. Lauren E. Frick and William A. LaMarr Agilent Technologies, Inc. Wakefield, MA, USA

HIGH-THROUGHPUT DETERMINATION OF AQUEOUS SOLUBILITY, PRECIPITATION OR COLLOIDAL AGGREGATE FORMATION IN SMALL MOLECULE SCREENING SETS

Battle the heat in your lab

Stoichiometry and Physical Chemistry of Promiscuous Aggregate-Based Inhibitors

Kinase-Glo Luminescent Kinase Assay Platform

P450-Glo Screening Systems All technical literature is available on the Internet at: Please visit the web site to verify that you

Z -LYTE Assay Setup Guide on the BMG LABTECH CLARIOstar Reader

ab83360 Ammonia Assay Kit

Optimization of temperature-sensitive assays using the Spark Te-Cool

Fragment-Based Drug Discovery (FBDD) Using the dispr Technique on Pioneer Systems with OneStep and NeXtStep Injection Methodologies

Nucleic Acid Quantitation in Microplates

For the rapid, sensitive and accurate measurement of Urea levels in various samples.

Optimization of an Adapta Kinase Assay for CAMK1

Human C-reactive protein ELISA using Sword Peroxidase Reagents

Low-volume, High Throughput Workflow for Analysis of Nucleic Acid Samples for Biobanking

ab Uricase Assay Kit (Fluorometric) 1

Predictor Assay Setup Guide on the Tecan Safire 2 Microplate Reader

protein interaction analysis bulletin 6300

FRAGMENT SCREENING IN LEAD DISCOVERY BY WEAK AFFINITY CHROMATOGRAPHY (WAC )

Malachite Green Phosphate Detection Kit Catalog Number: DY996

Applications Note 215 September 2009

LanthaScreen Eu Kinase Binding Assay Validation Packet. Optimization of a LanthaScreen Eu Kinase Binding Assay for AURKB

Characterization of Reversible Kinase Inhibitors using Microfluidic Mobility-Shift Assays

ab83360 Ammonia Assay Kit

Non-Interfering Protein Assay Kit Cat. No

Introduction to FBDD Fragment screening methods and library design

LanthaScreen Eu Kinase Binding Assay Validation Packet. Optimization of a LanthaScreen Eu Kinase Binding Assay for RPS6KA1

Small-Molecule Kinetics

Small-Molecule Kinetics

Caspase-1 Specific Light-up Probe with Aggregation-Induced Emission. Characteristics for Inhibitor Screening of Coumarin-Originated Natural.

RayBio Rat TNF-alpha ELISA Kit (For Lysates)

ab Alkaline Phosphatase Assay Kit (Luminometric)

For the rapid, sensitive and accurate measurement of Pyruvate Kinase (PK) activity in various samples.

For the rapid, sensitive and accurate measurement of Catalase activity in various samples.

Ammonia Assay Kit Modified Berthelot (Colorimetric)

High-throughput Isolation of Genomic DNA From Buccal Swab on the Eppendorf epmotion 5075 VAC

Screening in a spirit haunted world

Precision and accuracy of protein size determination using the ActiPix TDA200 Nano-Sizing System

ab Caspase 3, Caspase 8 and Caspase 9 Multiplex Activity Assay Kit (Fluorometric)

Human Creatinine Serum Detection Kit

Protease Activity Assay Kit

MiSeq System. Denature and Dilute Libraries Guide

Measurement of L-Malic Acid in Wines Using the SpectraMax Plus 384 Microplate Reader

PicoProbe D-Lactate Assay Kit (Fluorometric)

ab MDA Assay Kit (competitive ELISA)

Antioxidant Assay Kit

Xanthine Oxidase Assay Kit

Microdetermination of phosphorus using the SPECTRAmax PLUS microplate spectrophotometer: choice of microplate, cuvette or test tube assay formats

LanthaScreen Eu Kinase Binding Assay Validation Packet

High-throughput Quantification of DNA for NGS Library Prep with the Zephyr G3 Workstation and the VICTOR Nivo Plate Reader

ab DCF ROS/RNS Assay Kit (biofluids, culture supernatant, cell lysates)

VDL ENDOTOXIN ASSAY: ASSAY FOR LIMULUS AMEBOCYTE LYSATE

RayBio Human IDUA ELISA Kit

Human Syndecan-3 ELISA Kit

Predictor Assay Setup Guide on the BMG LABTECH CLARIOstar Microplate Readers

High Sensitivity Polyethylene Glycol (PEG) ELISA Kit

Improved Throughput and Reproducibility for Targeted Protein Quantification Using a New High-Performance Triple Quadrupole Mass Spectrometer

Acetylcholinesterase Assay Kit (Fluorometric - Red)

viridis Columns Bringing a New Dimension to SFC Combining state-of-the-art media manufacturing with industry leading column technology, Viridis

LanthaScreen Eu Kinase Binding Assay for GAK

Cobalt Quantification Kit

Mouse TNF-alpha ELISAKit

RayBio Human Thyroid Peroxidase ELISA Kit

Introduction Acetylcholinesterase (AChE) is one of the most important enzymes involved in nerve transmission. The enzyme is bound to cellular membrane

Caspase 3 Inhibitor Drug Detection Kit

Human Mullerian Inhibiting Substance/Anti-Mullerian hormone (MIS/AMH)Elisa Kit

Total Protein Assay Kit

Alpha Ketoglutarate (alpha KG) Assay Kit

Acetylcholinesterase Assay Kit (Fluorometric - Green)

Acetylcholinesterase Assay Kit

Protein Quantification Kit (Bradford Assay)

Multiple Fragmentation Methods for Small Molecule Characterization on a Dual Pressure Linear Ion Trap Orbitrap Hybrid Mass Spectrometer

Quick Guide QUICK GUIDE. Activity 1: Determine the Reaction Rate in the Presence or Absence of an Enzyme

Human cyclophilin A, CyPA ELISA Kit

Polyethylene Glycol (PEG), High Sensitive ELISA

IN QUALITATIVE ANALYSIS,

Human anti-deoxyribonuclease B, anti-dnase B ELISA Kit

Optical density measurements automatically corrected to a 1-cm pathlength with PathCheck Technology

Introduction to Fourier Transform Infrared Spectroscopy

Glutathione Reductase (GR) Assay Kit

Increasing Speed of UHPLC-MS Analysis Using Single-stage Orbitrap Mass Spectrometer

MicroCal itc 200. System MicroCal Auto-iTC 200. System. GE Healthcare Life Sciences. System design and description. provide:

Exploring the Benefits of Automated Unattended Sample Derivatization Prior to Gas Chromatography Analysis

Aldehyde Quantification Assay Kit (Colorimetric)

Mouse Creatinine Urinary Detection Kit

New sesquiterpenoids from the rhizomes of Acorus tatarinowii

[ OSTRO PASS-THROUGH SAMPLE PREPARATION PRODUCT ] The Simpler Way to Cleaner Samples

Creatinine Colorimetric Detection Kit

BIOO FOOD AND FEED SAFETY. Histamine Enzymatic Assay Kit Manual. Catalog #: Reference #:

Oxaloacetate Assay Kit

NMR Solutions for drug discovery

Aflatoxin M1 (AFM1) ELISA Kit

PowerWaveX Select and KC4 : A Multifunctional System for Today s Laboratory Environment

Human von Willebrand Factor cleavingprotease(vwf-cp) ELISA Kit. Catalog No. MBS (96T)

Strategies to compare far-uv Circular Dichroism spectra of similar proteins using Chirascan -plus ACD

Colorimetric Assay for Nitric Oxide Product No For Research Use Only

Introduction to Fourier Transform Infrared Spectroscopy

The Coulter Principle for Outlier Detection in Highly Concentrated Solutions

Nanoliter Scale High-Throughput Protein Crystallography Screening with the Echo Liquid Handler

Human placenta lactogen (HPL)ELISA Kit. MyBioSource.com. Catalog No. MBS (96T)

Transcription:

Label-Free Detection of Compound Aggregation Using Corning Epic Technology SnAPPShots A brief technical report from the Corning Development Group David H. Randle, Kathleen A. Krebs, Hannah J. Gitschier and Todd M. Upton Corning Incorporated, Life Sciences Kennebunk, ME 04043 Introduction Small molecule compound aggregation is a major problem encountered in primary screening assays. Once formed, the aggregates can sequester, cause partial unfolding, inhibit or inactivate a protein. The effects can be observed on many structurally and functionally unrelated proteins. It is estimated that up to 19% of compounds in typical pharmacological libraries can form aggregates at concentrations typically used for high-throughput screening (HTS) (1). Considerable time and expense can be wasted pursuing these false hits in hit-to-lead optimization. Several methods have been used to detect formation of aggregates including; a) enzyme activity-based assays with β-lactamase, chymotrypsin, malate dehydrase (2) or kinases (3); b) direct detection of aggregates via static or dynamic light scattering (DLS) (1); c) direct measurement of compound-protein interaction by surface plasmon resonance (4); and d) fluorescence measurements of meniscus shape (5). However, the utility of these methods for rapidly characterizing the aggregation activity of compounds derived from a primary HTS campaign is restricted by either a lack of throughput, reliability or simplicity. The Corning Epic technology can detect molecular interaction using optical biosensors integrated into the bottom of Epic microplate wells. Binding or settling of compound aggregates to the biosensor can be detected by measuring the wavelength shift of reflected resonance light (Fig. 1). In this study, Epic technology was used to detect aggregate formation using a set of known aggregators and non-aggregators. These data show that the label-free assay format could effectively detect compound aggregation and enabled clear differentiation from a non-aggregating group of compounds. Further, it was demonstrated that aggregation measured using Epic technology shares many features in common with other detection technologies such as steep concentration curves and sensitivity to the presence of non-ionic detergent. These data demonstrate that Epic technology is ideally suited for the rapid identification of aggregators, and can be readily implemented to attenuate false positives from HTS hit pools. Materials and Methods Compounds A panel of aggregators and non-aggregators was chosen based on previous characterization with either enzyme inhibition-based assays and/or DLS (1,2). Eight aggregators were chosen for analysis; Clotrimazole (C6019), Sulconazole (S9632), Econazole (E4632), Miconazole (M3512), Nicardipine (N7510), Palatine Chrome Black (45550), Tetraiodophenolphthalein (224987), and Rose Bengal lactone (328960). Six non-aggregators were used as negative controls; Ketoconazole (K1003), Fluconazole (F8929), Sulfadiazine (S8626), Thalidomide (T144), Prednisone (P6254), and Tripelennamine (T7511). All compounds were purchased from Sigma- Aldrich (Saint-Louis, MO) and catalog numbers are shown Figure 1. Epic Technology can detect compound aggregation using a simple add and read assay format. Uncoated Epic microplates containing integrated optical biosensors in each well are first soaked with assay buffer and a baseline measurement is taken. Compound is added in DMSO and aggregate formation at the surface of the biosensor is detected by measuring the wavelength shift of reflected resonant light.

in parentheses. Compound stock solutions of 50 mm were prepared in DMSO. Dilution series were made for each compound in DMSO to ensure a final 1x assay concentration range of 100 µm to 1.2 µm. For example, if the final DMSO concentration was 1%, compounds were prepared at 100x concentrations ranging from 10 mm to 123 µm. Epic Aggregation Assay Procedure Epic 384 Well Uncoated Cell Assay Microplates (Cat. No. 5040) were used for detecting aggregation formation. Microplates were first soaked in 50 mm HEPES plus 10 mm MgCl 2 (soaking buffer) and temperature-equilibrated in the Epic reader for at least 1 hour. After a baseline measurement was taken for 2 minutes, compound dilution series in DMSO were added with 10 mixes using a CyBio CyBi -Well pipetting system. Compounds were dispensed in DMSO to avoid predilution prior to addition into the Epic microplate. A range of final DMSO concentrations was tested using the assay formats shown in Table 1. Following compound addition, the microplate was returned immediately to the Epic reader and signal changes were recorded as continuous (kinetic) time traces for at least 1 hour. Sensitivity to nonionic detergent was tested by including 0.1% Triton X-100 in the soaking buffer. Each of the assays was performed at least three times with excellent reproducibility. Data Analysis Aggregation response profiles were analyzed using the Epic Data Analysis Tool and dose response curves were generated using Graphpad Prism software. Dose response plots were obtained using the difference in response values between the last baseline read and the aggregation response at 60 minutes. Sensitivity of the aggregation assay was determined using Thalidomide as the negative control and plotting the mean non-aggregator response at each concentration plus 3x the standard deviation of the response (3σ). The 3σ threshold was used to determine the critical aggregation concentration (CAC) below which, no aggregation was observed. Table 1. Summary of soak buffer volumes and compound add volumes required to run Epic aggregation assays at different final DMSO concentrations. Final DMSO (%) Soak Buffer (µl) Compound Add (µl) 1.0 49.5 0.5 2.0 49.0 1.0 3.0 48.5 1.5 4.0 48.0 2.0 5.0 47.5 2.5 Results and Discussion Current technologies for detecting compound aggregation typically lack throughput capability and involve complex assay formats. A simple add and read assay format was developed to detect aggregation using Epic technology (Fig. 1). Compounds are added directly to assay buffer in uncoated microplates and formation of aggregates in the solution is monitored by changes in reflected wavelength from the biosensor, measured in picometers (pm). Although some aggregation assay formats require the presence of enzyme, which sequesters compound aggregates resulting in inhibition of enzyme function, it was found that little difference in aggregation responses is observed when comparing uncoated microplates with biochemical microplates pre-immobilized with trypsin (data not shown). No compound predilution is required for the Epic aggregation assay; instead, compounds are added directly in DMSO. This results in a DMSO mismatch response which necessitates the inclusion of DMSO alone as a control to establish the background response, which can subsequently be subtracted from the raw data to provide corrected responses. Representative optical response profiles for a dose series of a known aggregator, Sulconazole and a known non-aggregator, Prednisone are shown in Fig. 2. Both compounds exhibit an Figure 2. Optical response profiles readily distinguish aggregators from non-aggregators. Sulconazole (known aggregator) and Prednisone (known non-aggregator) were added in dose response series from 100 µm to 1.2 µm (1:3 dilutions). DMSO alone was added as control to determine the assay background (grey line). Final DMSO concentration in the assay was 2%.

Figure 3. Aggregators using Epic technology show characteristic steep dose response curves. A panel of four known aggregators and four known nonaggregators were tested using the same dose response series as in Fig. 2, with a final DMSO concentration of 2%. Plots were generated using the response values at 60 minutes after compound addition and subtracting out the DMSO background response. initial positive spike over the first 10 to 20 minutes as a result of compound addition, but the response stabilizes between 30 and 60 minutes after compound addition. Sulconazole exhibits a clear dose-dependent response at 60 minutes with responses of several thousand pm at the highest concentrations tested, which is close to the upper limit of detection for Epic technology. This suggests that the response leads to very large changes in refractive index at the biosensor, as would be expected following the formation of aggregates throughout the solution above the biosensor. Prednisone, on the other hand, exhibits almost no dose-dependency at 60 minutes, and the response is barely above the DMSO background. In this assay, compound was added to leave a final DMSO concentration of 2%, resulting in a DMSO background response at 60 minutes of ~500 pm. The magnitude of the DMSO background response directly correlates with final DMSO concentration, so that at concentrations above 2% the background would be higher whereas at concentrations below 2% it would be lower (data not shown). Figure 3 shows dose response curves for these compounds at 60 minutes, together with additional aggregators and nonaggregators, after the DMSO background has been subtracted. Each of the known aggregators exhibits a characteristic steep dose response curve, one of the hallmarks of aggregation using technologies such as DLS. In contrast, the known non-aggregators show no response at even the highest concentration tested (100 µm), and this observation was extended out to doses as high as 250 µm (data not shown). Thus, Epic technology can readily distinguish between aggregating and non-aggregating compounds in a simple assay format. Another important characteristic of compound aggregation is sensitivity to non-ionic detergent such as Triton X-100. In order to evaluate whether Epic aggregation assays also demonstrated this attribute, two of the known aggregators were tested in assay buffer containing 0.1% Triton X-100. Figure 4 shows that the aggregation response is fully inhibitable up to 33.3 µm when detergent is included in the Figure 4. Aggregation responses using Epic technology are sensitive to non-ionic detergent. Two known aggregators, Econazole and Sulconazole were tested in assay buffer ± 0.1% Triton X-100 (TX-100) using the same dose response series as Fig. 2, with a final DMSO concentration of 2%. assay buffer. At concentrations > 33.3 µm, aggregation responses are observed in the presence of detergent, although they are attenuated in comparison to responses in the absence of detergent (data not shown). In addition to detergentsensitivity, Figure 5 shows that Epic aggregation assays are sensitive to increasing concentrations of DMSO in the assay. For each of the three aggregators tested, there is a clear reduction in the magnitude of the response with increasing DMSO concentration, particularly at the higher concentrations of 33.3 and 100 µm. These data show that Epic aggregation assays share all of the key features of compound aggregation exhibited by other detection methods. Shoichet and colleagues have used DLS to define the critical aggregation concentration (CAC) of several aggregating compounds, which is the concentration above which a sharp and linear increase in particle numbers is observed (6).

Three of the aggregating compounds were included in that study, so a method was developed to determine the CAC using Epic technology. A dose response series of each aggregator was run at a final DMSO concentration of 1% and compared to a non-aggregator, Thalidomide, tested at the same concentrations. The aggregation threshold was defined as the average of the Thalidomide response at each concentration plus 3x the standard deviation of the response (3σ). Figure 6 shows the data for each of the three aggregators with the 3σ line used to determine the minimum concentration at which an aggregation response is observed. Using this approach, the CAC using Epic technology is shown to be reduced by ~3-fold for Miconazole and Nicardipine, and almost 10-fold for Tetraiodophenolphtalein, when compared to DLS (Table 2). In the DLS assays, these compounds were tested at DMSO concentrations of either 0.1% or 0.5%. Shoichet and colleagues point out that CAC increases from 0.1% to 1% DMSO (6), which indicates that the sensitivity difference using Epic technology could be further enhanced at comparable DMSO concentrations. Finally, the entire panel of compounds was tested and found that all of the known aggregators had a CAC of 11.1 µm or lower (Table 3). While 5/6 of the known non-aggregators showed no response (CAC >100 µm), in three separate Table 2. Summary of critical aggregation concentrations for three compounds using Epic technology and DLS. Critical Aggregation Concentration Compound Epic Technology DLS* Miconazole 1.2 µm 3 µm Tetraiodophenolphtalein 1.2 µm 10 µm Nicardipine 11.1 µm 32 µm *Values for DLS taken from study by Coan and Shoichet (6). experiments it was observed that Ketoconazole gave an unexpected aggregation response with a CAC of 33.3 µm. It is possible that Ketoconazole has been falsely identified as a non-aggregator using other technologies. It is concluded that aggregation assays using Epic technology exhibit greater sensitivity than alternative detection technologies. Conclusions Compound aggregation can be detected with Epic technology using a simple add and read assay format. Epic technology can readily differentiate aggregators from non-aggregators. Figure 5. Aggregation responses using Epic technology are sensitive to increasing concentrations of DMSO. Three known aggregators, Miconazole, Tetraiodophenolphtalein, and Palatine Chrome Black were tested in assay buffer using the same dose response series as Fig. 2, but with increasing final DMSO concentrations. Figure 6. Critical aggregation concentration (CAC) can be determined using Epic technology. Dose series of three known aggregators, Nicardipine, Miconazole and Tetraiodophenolphtalein and a known non-aggregator, Thalidomide, was tested from 33.33 µm to 1.23 µm (1:3 dilutions) with a final DMSO concentration of 1%. Aggregation threshold is the average of the Thalidomide response at each concentration plus 3x the standard deviation of the response (dotted line). CAC is defined as the minimum concentration at which the dotted line intersects with the aggregator response.

Table 3. Summary of CAC for all aggregators (left panel) and non-aggregators (right panel) tested using Epic Technology. Aggegators CAC Clotrimazole 11.1 µm Econazole 11.1 µm Nicardipine 11.1 µm Sulconazole 3.7 µm Palatine Chrome Black Miconazole 3.7 µm Miconazole 1.2 µm Tetraiodophenolphtalein 1.2 µm Rose Bengal lactone 1.2 µm Non-Aggegators CAC Fluconazole >100 µm Prednisone >100 µm Sulfadiazine >100 µm Thalidomide >100 µm Tripelennamine >100 µm Ketoconazole 33.3 µm Label-free aggregation shares important characteristics with other detection methods including steep dose response curves and sensitivity to non-ionic detergent. Epic aggregation assays are readily amenable to HTS by selecting a single time point for end reads between 30 and 60 minutes after compound addition. References 1. Feng, B.Y., Shelat, A., Doman, T.N., Guy, R.K., and Shoichet, B.K. High-throughput assays for promiscuous inhibitors. Nat. Chem. Biol. 1:146-148 (2005). 2. Seidler, J., McGovern, S.L., Doman, T.N., and Shoichet, B.K. Identification and prediction of promiscuous aggregating inhibitors among known drugs. J. Med. Chem. 46:4477-4486 (2003). 3. Habig, M., Blechschmidt, A., Dressler, S., Hess, B., Patel, V., Billich, A., Ostermeier, C., Beer, D., and Klumpp, M. Efficient elimination of nonstoichiometric enzyme inhibitors from HTS hit lists. J. Biomol. Screen. 14:679-689 (2009). 4. Giannetti, A.M., Koch, B.D., and Browner, M.F. Surface plasmon resonance based assay for the detection and characterization of promiscuous inhibitors. J. Med. Chem. 51:574-580 (2008). 5. Cai, L., and Gochin, M. Colloidal aggregate detection by rapid fluorescence measurement of liquid surface curvature changes in multiwell plates. J. Biomol. Screen. 12:966-971 (2007). 6. Coan, K.E.D., and Shoichet, B.K. Stoichiometry and physical chemistry of promiscuous aggregate-based inhibitors. J. Am. Chem. Soc. 130:9606-9612 (2008).

For additional product or technical information, please visit www.corning.com/lifesciences or call 1.800.492.1110. Outside the United States, please call 978.442.2200. Corning Incorporated Life Sciences Tower 2, 4th Floor 900 Chelmsford St. Lowell, MA 01851 t 800.492.1110 t 978.442.2200 f 978.442.2476 www.corning.com/lifesciences Worldwide Support Offices A S I A / P A C I F I C Australia/New Zealand t 0402-794-347 China t 86 21 2215 2888 f 86 21 6215 2988 India t 91 124 4604000 f 91 124 4604099 Corning and Epic are registered trademarks of Corning Incorporated, Corning, NY. All other trademarks in this document are the property of their respective owners. Corning Incorporated, One Riverfront Plaza, Corning, NY 14831-0001 Japan t 81 3-3586 1996 f 81 3-3586 1291 Korea t 82 2-796-9500 f 82 2-796-9300 Singapore t 65 6733-6511 f 65 6861-2913 Taiwan t 886 2-2716-0338 f 886 2-2516-7500 E U R O P E France t 0800 916 882 f 0800 918 636 Germany t 0800 101 1153 f 0800 101 2427 The Netherlands t 31 20 655 79 28 f 31 20 659 76 73 United Kingdom t 0800 376 8660 f 0800 279 1117 All Other European Countries t 31 (0) 20 659 60 51 f 31 (0) 20 659 76 73 L AT I N A M E R I C A Brasil t (55-11) 3089-7419 f (55-11) 3167-0700 Mexico t (52-81) 8158-8400 f (52-81) 8313-8589 2011 Corning Incorporated Printed in U.S.A. 10/11 POD CLS-AN-185