Characterization of Reversible Kinase Inhibitors using Microfluidic Mobility-Shift Assays

Similar documents
Optimization of an Adapta Kinase Assay for CAMK1

Kinase-Glo Luminescent Kinase Assay Platform

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

ENZYME KINETICS. Medical Biochemistry, Lecture 24

After lectures by. disappearance of reactants or appearance of. measure a reaction rate we monitor the. Reaction Rates (reaction velocities): To

Enzymes Part III: Enzyme kinetics. Dr. Mamoun Ahram Summer semester,

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

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

SIMPLE MODEL Direct Binding Analysis

Biochemistry. Lecture 8 Enzyme Kinetics

Name Student number. UNIVERSITY OF GUELPH CHEM 4540 ENZYMOLOGY Winter 2002 Quiz #1: February 14, 2002, 11:30 13:00 Instructor: Prof R.

LanthaScreen Eu Kinase Binding Assay for GAK

it is assumed that only EH and ESH are catalytically active Michaelis-Menten equation for this model is:

Polyethylene Glycol (PEG), High Sensitive ELISA

2013 W. H. Freeman and Company. 6 Enzymes

Biochemistry Enzyme kinetics

Chapter 8. Enzymes: basic concept and kinetics

Chemistry 112 Chemical Kinetics. Kinetics of Simple Enzymatic Reactions: The Case of Competitive Inhibition

High Sensitivity Polyethylene Glycol (PEG) ELISA Kit

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

C a h p a t p e t r e r 6 E z n y z m y e m s

Certificate of Analysis

Biochemistry 3100 Sample Problems Binding proteins, Kinetics & Catalysis

Nitric Oxide Synthase Ultrasensitive Colorimetric Assay

LanthaScreen Eu Kinase Binding Assay Validation Packet

BA, BSc, and MSc Degree Examinations

Prof. Jason D. Kahn Your Signature: Exams written in pencil or erasable ink will not be re-graded under any circumstances.

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

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

Non-Interfering Protein Assay Kit Cat. No

Energy Transformation, Cellular Energy & Enzymes (Outline)

Chapter 8: An Introduction to Metabolism

Chapter 6: Outline-2. Chapter 6: Outline Properties of Enzymes. Introduction. Activation Energy, E act. Activation Energy-2

Biochemical / Biophysical Kinetics Made Easy

Bioengineering Laboratory I. Enzyme Assays. Part II: Determination of Kinetic Parameters Fall Semester

Part II => PROTEINS and ENZYMES. 2.7 Enzyme Kinetics 2.7a Chemical Kinetics 2.7b Enzyme Inhibition

Porcine Factor-related Apoptosis(FAS) ELISA Kit

cgmp ELISA Kit (Direct Competitive) Based on Monoclonal Anti-cGMP Antibody

BIOCHEMISTRY - CLUTCH REVIEW 2.

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

Mouse Cholecystokinin (CCK) ELISA Kit

Problem Set # 3

It can be derived from the Michaelis Menten equation as follows: invert and multiply with V max : Rearrange: Isolate v:

Overview of Kinetics

A. One-Substrate Reactions (1) Kinetic concepts

Discussion Exercise 5: Analyzing Graphical Data

Aflatoxin M1 (AFM1) ELISA Kit

Algebraic solution for time course of enzyme assays. Enzyme kinetics in the real world. Progress curvature at low initial [substrate]

RIO TINTO AUSTRALIAN CHEMISTRY OLYMPIAD

LanthaScreen Eu Kinase Binding Assay Validation Packet. Optimization of a LanthaScreen Eu Kinase Binding Assay for STK33

Malachite Green Phosphate Detection Kit Catalog Number: DY996

CHGA (Human) ELISA Kit

Irreversible Inhibition Kinetics

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

4. What is the general expression Keq (the equilibrium constant) in terms of product and reactant concentration? tell us about the enzyme.

Porcine Immunoglobulin E (IgE)ELISA Kit

Monensin ELISA Kit. Catalog Number KA assays Version: 11. Intended for research use only.

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

Rat Advanced Glycation End Products(AGEs) ELISA Kit

C a h p a t p e t r e r 6 E z n y z m y e m s

Nitric Oxide Synthase Assay Kit

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

Time depending inhibition with atypical kinetics, what is one to do? Ken Korzekwa Temple University School of Pharmacy

Incubation time too short Incubate samples overnight at 4 C or follow the manufacturer guidelines.

Unit 3. Enzymes. Catalysis and enzyme kinetics.

BCMB 3100 Chapters 6,7,8 Enzyme Basics. Six Classes (IUBMB) Kinetics Michaelis-Menten Equation Vo, Km, Vmax, Kcat Lineweaver-Burk Plot

CHAPTER 1: ENZYME KINETICS AND APPLICATIONS

This immunoassay kit allows for the in vitro quantitative determination of Aflatoxin M1 concentrations in milk, milk power.

New sesquiterpenoids from the rhizomes of Acorus tatarinowii

Problem Set 5 Question 1

Canine Erythropoietin,EPO ELISA Kit

Problem Set 2. 1 Competitive and uncompetitive inhibition (12 points) Systems Biology (7.32/7.81J/8.591J)

Optimization of a LanthaScreen Eu Kinase Binding Assay for PIK3CA/PIK3R1

Rat Prolactin ELISA Kit

13 Determining the Efficiency of the Enzyme Acetylcholine Esterase Using Steady-State Kinetic Experiment

BCH 3023 Fall 2008 Exam 2, Form C Name: ANSWER KEY

SAL (Salbutamol) ELISA Kit

Reading for today: Chapter 16 (selections from Sections A, B and C) Friday and Monday: Chapter 17 (Diffusion)

Biochemistry and Physiology ID #:

Learning Outcomes. k 1

Zearalenone ELISA Kit

MyBioSource.com. This package insert must be read in its entirety before using this product.

A hypothetical model of the influence of inorganic phosphate on the kinetics of pyruvate kinase

Lecture 11: Enzyme Kinetics, Part I

8/16/2010 Version 3.0 KINESIN ELIPA BIOCHEM KIT BK060

Human Paraneoplastic pemphigus (PNP)antibody ELISA Kit. Catalog No. MBS (96 T)

Human Coagulation Factor X Total Antigen ELISA Kit

Serine-7 but not serine-5 phosphorylation primes RNA polymerase II CTD for P-TEFb recognition

Biochemistry 462a - Enzyme Kinetics Reading - Chapter 8 Practice problems - Chapter 8: (not yet assigned); Enzymes extra problems

Applications Note 215 September 2009

Sheep myeloperoxidase(mpo) ELISA kit

Chemistry 112 Final Exam, Part II February 16, 2005

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

Predictor Assay Setup Guide on the BMG LABTECH CLARIOstar Microplate Readers

CTX-II (Human) ELISA Kit

Paraquat ELISA Kit. Catalog Number KA assays Version: 17. Intended for research use only.

Mouse Anti-OVA-IgE ELISA KIT

Chapter 8: An Introduction to Metabolism

IgE (Rat) ELISA Kit. Catalog Number KA assays Version: 05. Intended for research use only.

Previous Class. Today. Cosubstrates (cofactors)

Transcription:

Application Note 211 Characterization of Reversible Kinase Inhibitors using Microfluidic Mobility-Shift Assays Introduction Current drug discovery efforts typically focus on developing small molecule inhibitors of target enzymes involved in regulating specific cellular processes. Mechanism of action (MOA) studies are necessary to characterize and compare potential drug candidates. The ability to follow reaction rates in real time greatly facilitates the determination of inhibition mechanisms and the calculation of dissociation constants. With real time kinetics capability, Caliper s LabChip EZ Reader and LabChip 3000 provide ideal platforms for these studies. Individual wells within a microtiter plate can be sampled repeatedly during the course of an experiment, allowing for accurate determination of reaction velocities from the resulting reaction progress curves. In addition, the mobility-shift format provides direct detection of both product and substrate to produce consistent high-quality data and minimize the potential for indirect effects of test compounds. This application note describes MOA experiments conducted to characterize enzyme-inhibitor interactions for known inhibitors of the cyclic AMPdependent protein kinase (PKA). Data obtained using 4-sipper and 12-sipper chips were used to demonstrate reversible inhibition, confirm rapid establishment of equilibrium between enzyme and inhibitor, determine inhibitor IC 50 values at different substrate concentrations, discriminate between ATP- competitive, noncompetitive, or uncompetitive mechanisms of inhibition, and calculate K i values. Table 1. PKA Mobility-Shift Assay conditions. Reactions were assembled as described for individual experiments in 384-well microtiter plates in a total volume of 71 µl.

Page 2 - Application Note 211 Figure 1. Biochemical analyses typically performed during the characterization of lead compounds for drug discovery. Adapted from Copeland, R.A. (2005) 1 Results and Discussion In order to make valid comparisons of relative activities between different classes of lead compounds, it is critical to characterize inhibitors with methods appropriate to the mechanism of inhibition. Figure 1 shows a flowchart outlining basic steps involved in lead characterization. After an initial IC 50 determination, usually done at substrate concentrations equal to the K m value(s), studies are conducted to determine whether inhibitor binding is reversible, slowly reversible, or irreversible. In the case of irreversible inhibitors, evaluation of compound affinity should only be done after further characterizing the nature of binding between enzyme and inhibitor (right-hand side of Figure 1). For reversible inhibitors, reaction progress curves will show how rapidly the enzyme and inhibitor achieve equilibrium. Inhibitors that bind rapidly and produce linear progress curves can be evaluated using classical steady-state analyses. Inhibitors that bind the enzyme slowly and produce curvilinear progress curves should be evaluated with experiments designed to analyze time-dependent inhibition. PKA is a well-characterized kinase with numerous commercially available inhibitors that have been analyzed for MOA. Previous work done with PKA, using the mobilityshift assay conditions described in Table 1, identified the apparent Michaelis-Menten constants and inhibitor IC 50 values shown in Table 2. It is important to note that all constants determined when working with a kinase are apparent constants, because the enzyme catalyzes a twosubstrate reaction. However, it is valid to treat a kinase reaction as a single-substrate reaction provided that only one substrate concentration is varied at a time, and the second substrate is kept constant at a concentration near its K m app value. 2 Table 2. K m app and IC 50 values used as initial estimates for designing MOA experiments.

Application Note 211 - Page 3 Reversible Inhibitor Binding Rapid dilution experiments were used to demonstrate reversible binding of H89 and Staurosporine to PKA. With this approach, inhibitor is allowed to bind enzyme at a concentration expected to inhibit 90% of activity, then diluted to a concentration expected to inhibit only 9% of activity. A reversible inhibitor dissociates quickly, allowing immediate recovery of approximately 91% of enzyme activity. An irreversible inhibitor prevents recovery of enzyme activity, and a slowly reversible inhibitor allows a gradual increase in activity. For rapid dilution, 5 µl of 120X Enzyme (24 nm) was mixed with 1 µl of DMSO with and without inhibitor at 60X the IC 50 value (H89: 1.8 µm, Staurosporine: 300 nm). After incubating 30 min at room temperature, the enzyme and inhibitor mixes were diluted 0.8 µl into 80 µl of 1X peptide (0.75 µm) and ATP (5 µm). The microplate was placed in the EZ Reader and wells were repeatedly sampled for 70 minutes. As shown in Figure 2, reactions containing H89 and Staurosporine proceeded at nearly 90% of the rate seen for control reactions, demonstrating reversible binding for both inhibitors. Linear Progress Curves Reaction progress curves were generated to demonstrate rapid establishment of equilibrium between PKA and its inhibitors. For this experiment, 1 µl 70X inhibitor in DMSO was spotted into each microtiter plate well. 17.5 µl of 4X ATP and 17.5 µl of 4X peptide were added and mixed with inhibitor. Reactions were initiated by the addition of 35 µl 2X enzyme, and progress curves were generated by repeated sampling of the reaction wells on the EZ Reader. Final concentrations were 32 µm ATP, 0.75 µm Peptide, 0.2 nm enzyme, 30 nm H89, 5 nm Staurosporine, and 0.6 nm PKI 6-22. The experiment was done with ATP at 8X the K m app value, to ensure that the presence of excess ATP did not slow the establishment of equilibrium. As shown in Figure 3, the progress curves were linear through the first 20 minutes, giving no indication of time-dependent inhibition by any of these inhibitors. This result validates the use of standard steady-state analysis to determine inhibition mode with respect to the substrate ATP. Figure 2. PKA activity was restored upon rapid dilution of concentrated enzyme-inhibitor mix. Figure 3. Linear PKA reaction progress curves were observed in the presence of H89, Staurosporine, and PKI 6-22. GraphPad Prism 5 was used for linear regression of data from the first 20 minutes of the reaction. The slopes of the resulting lines and associated R 2 values are shown in the table.

Page 4 - Application Note 211 Steady-State Analysis of Inhibition Mode To distinguish between ATP- competitive, uncompetitive, or noncompetitive mechanisms of inhibition, inhibitors were titrated at various concentrations of ATP. Figure 4 shows the plate layout for this experiment. Using one quadrant of a 384-well microplate, control reactions and an inhibitor titration are run at 6 different ATP concentrations, ranging from 8X Km to 0.25X Km. The inhibitor titration steps through a 9-point 1:3 dilution series starting at 81X the IC 50 at ATP Km. Here, the reactions were run in kinetic mode, using a 12-sipper chip on the LabChip 3000, for the most accurate determination of initial reaction velocities. Alternatively, this experiment can be run as an endpoint assay by stopping reactions with the addition of EDTA. Figure 4. Plate layout for steady-state analysis to determine inhibition mode with respect to ATP. For the experiments described in this note, the IC 50 and ATP K m estimates shown in Table 2 were used to determine final concentrations of inhibitors and ATP. All reactions were set up by spotting 1 µl 70X inhibitor in DMSO, then adding 35 µl 2X enzyme, 17 µl 4X Peptide, and 17 µl 4X ATP. Final concentrations of PKA and Peptide were 0.4 nm and 0.75 µm, respectively. Figure 5 provides an example of kinetic data from reactions containing 32 µm ATP, and varying concentrations of H89 inhibitor. Using GraphPad Prism 5, the entire timecourse for each well was plotted as % conversion over time, and the first 5 data points were used for linear regression analysis. The chart shows the calculated slopes and the R 2 value for each well. To evaluate mode of inhibition, first the initial slopes were multiplied by the concentration of peptide substrate (0.75 pmoles/µl) and the volume of the reaction (70 µl) to obtain initial velocities in pmol product/min. Second, the formula % inhibition = 100*(1-((v i -background)/(v 0 -background))), where v i = velocity with inhibitor, v 0 = average velocity of no inhibitor control, and background = velocity of no ATP control, was applied to the dataset. The % inhibition data was plotted vs. the log of ATP concentration and fitted in GraphPad Prism 5 with the formula for variable slope sigmoidal dose-response curves. Figure 5. Example of kinetic data obtained from reactions at one substrate concentration. Linear regression analysis was used to determine initial reaction velocities.

Application Note 211 - Page 5 Figure 6. Sigmoidal dose-response curves for H89 (A) and PKI 6-22 (B) fit to % inhibition data from reactions run with 6 different concentrations of ATP. Graphs C and D plot the IC 50 values obtained from the curves shown in A and B vs. ATP concentration. The dose response curves for H89 and PKI 6-22 are shown in Figures 6A and 6B. Plots of the calculated IC 50 values vs. ATP concentration (Figures 6C and 6D) show patterns expected for competitive inhibition in the case of H89 and uncompetitive inhibition in the case of PKI 6-22. Finally, initial velocity data was used for simultaneous non-linear regression analysis (SNLR) with models for competitive, uncompetitive and noncompetitive inhibition to determine best-fit values for K m, V max, and K i or ak i. This analysis was done in GraphPad Prism 5, using built-in models for fitting the three equations shown in Table 3. Table 4 shows best-fit values for the variable parameters in each model, along with fit statistics. By comparing fit statistics, the model most closely describing the data was chosen. For H89, the model giving the R 2 value closest to 1.0 and the lowest value for the sum of squares of residuals is that for competitive inhibition. For PKI 6-22, the best model is that for uncompetitive inhibition. Figures 7A and 7B show the observed H89 and PKI 6-22 data points together with the curves generated from the appropriate equations with the best-fit parameters (Table 4, green columns). Graphs 7C and 7D map the distribution of residuals to illustrate how the actual data deviates from points predicted by the fitted equations. Note that data from higher inhibitor concentrations was not included in SLNR analyses, as data from reactions with low conversion levels was less reliable and reduced goodness of fit across all models.

Page 6 - Application Note 211 Table 3. Equations used for SNLR Analysis of mechanism of inhibition in GraphPad Prism 5, where [S] = substrate concentration, v = initial velocity, and [I] = inhibitor concentration. Table 4. Results from SNLR analysis of H89 and PKI 6-22 data using the nonlinear fit function in GraphPad Prism 5. Parameter values are shown +/- standard error. The results from the model most closely fitting the data are highlighted in green. Figure 7. Michaelis-Menten curves resulting from nonlinear regression analysis of initial velocity data with the equation describing competitive (A) and uncompetitive (B) inhibition. Graphs C and D show the distributions of residuals representing the differences between the observed initial velocities and the initial velocities calculated from the fitted equations.

Application Note 211 - Page 7 Summary This application note demonstrates the power of Caliper s microfluidic assays for conducting mechanism of action studies. While the experiments shown were done with a kinase assay, the methods are applicable to all enzymatic assays compatible with the mobility-shift platform. Real-time kinetics capability minimizes the number of reaction wells necessary for each experiment. Progress curves can be generated rapidly to determine the reversibility of enzyme-inhibitor binding and the time required to establish equilibrium between enzyme and inhibitor. Steady-state analysis of mechanism of inhibition can also be done in kinetic mode, allowing reliable determination of initial reaction velocities for the most accurate estimation of best-fit model parameters. In addition, real-time kinetics capability enables a wide range of experiments necessary to explore more complex interactions between enzymes and inhibitors. Examples include use of tight-binding inhibitors to accurately determine enzyme concentration, characterization of tight-binding inhibition mode, and analysis of time-dependent inhibition. References 1. Copeland, R.A. (2005) Evaluation of Enzyme Inhibitors in Drug Discovery, Wiley & Sons, Hoboken, New Jersey. 2. Cornish-Bowden, A. (2004) Fundamentals of Enzyme Kinetics, Third Edition, Portland Press Ltd., London. ITEM MANUFACTURER CATALOG NO. LabChip EZ Reader I ProfilerPro Chip, 4-sipper LC3000 Drug Discovery System Chip Module TC Caliper Life Sciences Caliper Life Sciences Caliper Life Sciences Caliper Life Sciences Off-Chip Mobility-Shift Chip, Caliper Life Sciences 761037-0372R 12-sipper, with coating reagent 3 PKA (catalytic subunit a, human, Upstate 14-440 recombinant) Lot# 26698U, Specific Activity 11,430 U/mg FL-LRRASLG-CONH2 Caliper Life Sciences 760095 HEPES, Free Acid ULTROL Calbiochem 391338 HEPES, Sodium Salt ULTROL Calbiochem 391333 Magnesium Chloride, hexahydrate Sigma M2670 Brij-35 Solution Sigma B4184 DTT EMD 3860 ATP, disodium salt Sigma A7699 EDTA, disodium salt, 0.5 M, ph 8.0 Ambion 9260G DMSO JT Baker 9224-33 H89, Dihydrochloride Calbiochem 371963 PKI 6-22 Amide Calbiochem 539684 Staurosporine InSolution, Calbiochem 569396 1mM in DMSO 18 MW water

Corporate Headquarters 68 Elm Street Hopkinton, MA 01748-1668 Tel: 1-877-LabChip Fax: 1-508-435-3439 Email: cust.support@caliperls.com www.caliperls.com 2007 Caliper Life Sciences, Inc. All rights reserved. LC3000-AP-211