xcelligence System RTCA Cardio Instrument

Similar documents
mesc Derived Cardiomyocytes and the RTCA Cardio Instrument - The Perfect Match for Screening Cardiac Effects

Measuring Cardiac Activity:

Assessment of pro-arrhythmic effects using Pluricyte Cardiomyocytes. on the ACEA xcelligence RTCA CardioECR

xcelligence RTCA Cardio Instrument Monitor Cardiomyocyte Beating in Real Time for Drug Discovery Research

Assessment of pro-arrhythmic effects using Pluricyte Cardiomyocytes on the ACEA xcelligence RTCA CardioECR

Assessment of pro-arrhythmic effects using Pluricyte Cardiomyocytes. on the ACEA xcelligence RTCA CardioECR

Focus Application. Compound-Induced Cytotoxicity

Ncardia. Assessment of pro-arrhythmic effects in Pluricyte Cardiomyocytes. using the Axion BioSystems Maestro TM MEA system

Measuring Cardiac Activity:

Pluricyte Cardiomyocytes

Measuring Cardiac Activity:

Advances in Safety Pharmacology: Utilizing ipsc-derived cardiomyocytes in early stage safety pharmacology investigations. Blake Anson, PhD.

In Vitro Assessment to Replace the Clinical TQT Study: The Comprehensive In Vitro ProArrhythmia Assay (CiPA) Initiative

Focus Application. Compound-Induced Cytotoxicity

Metrion Biosciences: the ion channel specialists

Effects of reference compounds on impedance signals from stem cellderived human cardiomyocytes

Assessment of pro-arrhythmic effects using Pluricyte Cardiomyocytes on the Axion Maestro system

Pluricyte Cardiomyocytes. using the Multiwell MEA System from Multi Channel Systems

Assessment of pro-arrhythmic effects using Pluricyte Cardiomyocytes on the Axion BioSystems Maestro TM MEA system

Successes and Evolving Challenges Posed by the Comprehensive In Vitro Proarrhythmia (CiPA) Initiative

TdP Mechanisms and CiPA

Cardiomyocyte? March 14 th 2013

axion Protocol Cell Culture on Microelectrode Arrays Cell Type: Cryopreserved Human ipsc-derived Neurons BioSystems v. 1.1

Pluricyte Cardiomyocytes. Axion BioSystems Maestro TM MEA system

System. xcelligence System RTCA HT Instrument. Application Note No. 14 /January Long-Term High-Throughput Cytotoxicity Profiling

axion Protocol Cell Culture on Microelectrode Arrays Cell Type: GE Healthcare - Cytiva TM Plus Cardiomyocytes BioSystems v. 1.0

InPulse: Development of an ipsc cardiomyocyte (ipsc-cm) platform to assess drug-induced contractility liabilities.

Multiparametric assessment of the effects of chemotherapeutic drugs on the (electro)physiology of Pluricyte Cardiomyocytes

Performing Bioenergetic Analysis: Seahorse XFe96 Analyzer

Multiparameter In Vitro Assessment of Compound Effects on Cardiomyocyte Physiology Using ipsc Cells

Modulating Glucose Uptake in Skeletal Myotubes: Insulin Induction with Bioluminescent Glucose Uptake Analysis

Mechanisms of Arrhythmogenesis: Focus on Long QT Syndrome (LQTS)

MEA assays using human ipsc-derived cardiomyocytes; challenges and opportunities

Chapter 12: Cardiovascular Physiology System Overview

Welcome to the Exhibitor Tutorial presented by

Differing responses of human stem cell-derived cardiomyocytes to arrhythmogenic drugs, determined using impedance measurements

Comparison of different proarrhythmia biomarkers in isolated rabbit hearts

Human ipsc-derived Ventricular Cardiomyocytes. Protocol version 3.1

xcelligence Real-Time Cell Analyzers

The Application of Cell-Based Impedance Technology in Drug Discovery

Drug-induced cardiotoxicity: screening methods, regulatory context, commercial environment. Beremans Ltd / Insight Pharma Reports April 2008

Phase 2 Early Afterdepolarization as a Trigger of Polymorphic Ventricular Tachycardia in Acquired Long-QT Syndrome

Evolution of strategies to improve preclinical cardiac safety testing

Drugs, herg and sudden death

Developing and Validating an In Silico Model for Proarrhythmia Risk Assessment Under the CiPA Initiative

Product Use HPSC-CC are for research use only. It is not approved for human or animal use, or for application in in vitro diagnostic procedures.

Human Induced Plutipotent Stem Cell (ipsc) Handling Protocols: Matrigel and mtesr/e8 Media

Preclinical Perspectives of QT Outlook from ICH Guidelines

PART I. Disorders of the Heart Rhythm: Basic Principles

Human induced pluripotent stem cell derived cardiomyocytes are a more relevant model for assessing drug-induced effects on mitochondrial function

Human TRPC6 Ion Channel Cell Line

TCP Transl Clin Pharmacol

Data Sheet IL-2-Luciferase Reporter (Luc) - Jurkat Cell Line Catalog # 60481

Human Pluripotent Stem Cell Cardiomyocyte Differentiation Kit (PSCCDK) Introduction Kit Components Cat. # # of vials Reagent Quantity Storage

HHS Public Access Author manuscript Circ Res. Author manuscript; available in PMC 2016 June 05.

Rhythmical Excitation of the Heart

icell Cardiomyocytes 2 User s Guide

Identification of Potent and Selective Inhibitors to Investigate the Role of the Epithelial Sodium Channels in Neurodegeneration

nachr α 4 β 2 CHO Cell Line

Can the Thorough QT Study Be Replaced? Norman Stockbridge Division of Cardiovascular and Renal Products FDA/CDER

QT prolongation and drug-drug interactions. Filip Josephson M.D., Ph.D Clinical Assessor Swedish Medical Products Agency

NINDS Repository Human Induced Pluripotent Stem Cell (ipsc) Handling Protocols (Matrigel and mtesr Media)

INSTRUCTIONS Pierce Primary Cardiomyocyte Isolation Kit

hipsc Cardiomyocytes: Validation Study Results

A New Comprehensive Translational Paradigm Using Human ipsc-derived Cardiomyocytes for Safety Assessment of Drug-Induced Arrhythmias

Ion channel dysfunction and diseases of the heart

Cardiac muscle is different from other types of muscle in that cardiac muscle

Using Impedance-Based Approaches for Measuring Cell-Mediated Cytotoxicity; Antibody-Dependent (ADCC) and Chimeric Antigen Receptor T (CAR-T)

Data Sheet. NFAT Reporter (Luc) Jurkat Cell line Catalog #: 60621

Where are the normal pacemaker and the backup pacemakers of the heart located?

Effects of adrenergic activation to the action potentials and ionic currents of cardiac cells. by Ferenc Ruzsnavszky MD

xcelligence Real-Time Cell Analyzers

ROS Activity Assay Kit

Skeletal Muscle Contraction 4/11/2018 Dr. Hiwa Shafiq

ELECTROCARDIOGRAPHY (ECG)

Benchmarking In Silico Models and Candidate Metrics for Assessing the Risk of Torsade de Pointes

Antiarrhythmic Drugs

Perspectives on Evaluating and Managing Cardiovascular Safety Risk for Anticancer Drugs

Skeletal Muscle Contraction 5/11/2017 Dr. Hiwa Shafiq

icell Cardiomyocytes User s Guide Document ID: X1000

Chapter 9. Learning Objectives. Learning Objectives 9/11/2012. Cardiac Arrhythmias. Define electrical therapy

Full file at

Cardiac physiology. b. myocardium -- cardiac muscle and fibrous skeleton of heart

Instructions. Fuse-It-mRNA easy. Shipping and Storage. Overview. Kit Contents. Specifications. Note: Important Guidelines

MEA Application Note: Primary Culture Cardiomyocytes from Chicken Embryo

axion Protocol Cell Culture on Microelectrode Arrays Cell Type: QBM Cell Science - E14,15 Embryonic C57 Mouse Cortical Neurons BioSystems v. 1.

EKG Sensor Product Number: ENEKG189

EMEA PUBLIC STATEMENT ON THE RECOMMENDATION TO SUSPEND THE MARKETING AUTHORISATION FOR ORLAAM (LEVACETYLMETHADOL) IN THE EUROPEAN UNION

PHARMACOLOGY OF ARRHYTHMIAS

Cardiac Arrhythmias. For Pharmacists

CiPA, Pre-clinical Testing. Derek Leishman PhD DSP & ICH S7B

The Electrocardiogram

Collin County Community College. ! BIOL Anatomy & Physiology! WEEK 5. The Heart

Alpha-Tubulin Housekeeping 10,000 tests

Comparison of Electrophysiological Data From Human-Induced Pluripotent Stem Cell Derived Cardiomyocytes to Functional Preclinical Safety Assays

Electrocardiographic Markers Associated with Sotalol-induced Torsades de Pointes

Systems Biology Across Scales: A Personal View XXVII. Waves in Biology: Cardiac Arrhythmia. Sitabhra Sinha IMSc Chennai

Cardiac Properties MCQ

The QT Interval Safety Endpoint for DR- TB trials. Kelly Dooley & Gary Maartens (Disclaimer: I know very little cardiac electrophysiology)

Cardiac Safety Assessment. In-Vitro/In-Vivo Extrapolation

Transcription:

xcelligence System Application Note No. 15 January 2013 xcelligence System RTCA Cardio Instrument Preclinical Cardiac Safety Assessment using Human ipsc Derived icell TM Cardiomyocytes Introduction Over the last two decades, a number of blockbuster drugs have been withdrawn or have incurred safety warnings by regulatory agencies due to adverse cardiac effects. 1 In addition, lead compounds or drug candidates are frequently terminated at late stages of drug development due to cardiac safety concerns. Both of these factors can significantly impact the overall cost of drug discovery; consequently, pharmaceutical companies and regulatory agencies have implemented procedures to address these issues. Most of the drugs that have been withdrawn from the market due to cardiotoxicity appear to affect heart function by directly interfering with wellcoordinated electrical activity in the heart. 2 The electrical activity of the heart itself results from directional and selective movement of calcium, sodium, and potassium ions across channels in the membrane of cardiomyocytes; this movement couples with the specialized cytoskeleton to produce contraction and the force necessary for the pumping action of the heart. Interfering with this wellorchestrated movement of ions across the cardiomyocyte membrane can lead to electrical arrhythmia that will ultimately affect the excitation/ contraction coupling cycle of the heart and may lead to a lifethreatening form of ventricular arrhythmia called Torsades de Pointes (TdP). 1 For life science research only. Not for use in diagnostic procedures. For life science research only. Not for use in diagnostic procedures.

Introduction While the exact molecular mechanisms resulting in TdP are subject to debate and still under investigation, it is well accepted that delayed ventricular repolarization (evidenced as QT prolongation on the surface electrocardiogram) is a surrogate biomarker linked with enhanced proarrhythmic risk. 2 The main component of the ventricular repolarization in humans is the rapidly activated, delayed rectifier potassium current I Kr with the human etheràgogorelated gene product (herg) being the principal ion channel responsible. 2 Blocking of the herg channel in cardiomyocytes has been shown to prolong ventricular action potential duration (APD), and give rise to early afterdepolarizations (EADs), potential triggers of TdP. 2 Recognizing the central role of the herg channel in the repolarization phase of the action potential and its propensity to interact with drugs, current regulatory guidelines consider herg assays a major component of an integrated cardiac risk assessment. In this application note, we have used human induced pluripotent stem (ips) cellderived cardiomyocytes (icell TM Cardiomyocytes) from Cellular Dynamics International (CDI) together with the new xcelligence System RTCA Cardio Instrument as an assay system for preclinical in vitro cardiac safety assessment. icell Cardiomyocytes are a nearly 100% pure population of spontaneously beating cardiomyocytes that contain all the relevant ion channels, ionic currents, and cytoskeletal elements found in a typical cardiomyocyte. Our data shows that combining a physiologically relevant cardiomyocyte model system such as icell Cardiomyocytes together with the capabilities and throughput of the xcelligence System RTCA Cardio Instrument can provide predictive and mechanistic toxicity data that would allow researchers to make more accurate assessments of cardiac toxicity profiles of leading candidate compounds. It must be noted, however, that the herg channel block is not exclusively predictive of ventricular arrhythmia. Certain compounds may be able to inhibit multiple ion channels and therefore counteract the adverse effect of herg channel block, as has been shown for verapamil. 2 Also, induction of arrhythmia by mechanisms other than the herg block cannot be discounted. It would be highly desirable, earlier in the drug discovery process, to utilize an in vitro model system to assess the integrated response of all ion channels and nonion channel targets involved in the process of excitation/contraction coupling. The recent introduction of stem cell technology and stem cellderived cardiomyocytes for safety testing could provide a viable solution. 3 2

Materials and Methods icell TM Cardiomyocytes were obtained from CDI (Madison, Wisconsin USA; cellulardynamics.com). The cells were kept in liquid nitrogen until thawed and cultured according to CDI protocol with slight modifications. Briefly, each well of the EPlate Cardio 96 (ACEA Biosciences) was coated with 50 µl of a 1:100 diluted fibronectin (FN) solution (F1114, SigmaAldrich, USA) and incubated at +4 C overnight, followed by removal of the fibronectin, washing with PBS, and seeding. The cells were thawed at +37 C in a water bath and transferred to a 15 ml polypropylene conical tube. Eight milliliters of icecold plating medium was added dropwise while keeping the cells in suspension by gently swirling the tube. Using a swingingbucket rotor centrifuge, the cells were centrifuged at approximately 100 x g, resuspended in 1 ml of plating medium, and counted. The percentage of viable cells was determined by the trypan blue exclusion method. Dynamic Monitoring of icell Cardiomyocyte Beating Using the xcelligence System RTCA Cardio Instrument. Approximately 20,000 30,000 viable icell Cardiomyocytes were plated per well of the EPlate Cardio 96, and the cells were monitored using the xcelligence System RTCA Cardio Instrument (ACEA Biosciences). Per the protocol, the cells were thawed and plated in icell Cardiomyocytes Plating Medium, which was replaced with icell Cardiomyocytes Maintenance Medium 48 hours later. Drug treatment was typically initiated 8 9 days after cell seeding. Data collection was controlled by the RTCA Cardio Software, which operates the hardware and allows the user to define the sampling frequency and sampling window. Sampling frequency was defined as the number of times during an experimental run that the beating was sampled, and the sampling window was defined as the duration of time that the beating was actually measured. Ten minutes prior to treatment, the cells were sampled every minute for 20 seconds to establish baseline activity. After treatment, the sampling frequency was adjusted to every minute for the first hour, every 5 minutes for the second hour, and every 15 minutes for 3 24 hours. The sampling window for each recording was fixed at 20 seconds. After data acquisition, the RTCA Cardio Software calculated parameters such as beating frequency, amplitude, normalized beating frequency, normalized amplitude, beat duration, beating rate irregularity index, basic statistical analyses (average, standard deviation), and EC 50 values from doseresponse tests. 3

Results RealTime and Dynamic Monitoring of icell Cardiomyocyte Attachment, Growth, and Beating Using the xcelligence System RTCA Cardio Instrument The RTCA Cardio Instrument extends the current capabilities of the xcelligence System portfolio (including the RTCA SP, MP, and DP Instruments) to functional monitoring of cardiomyocyte beating and assessment of cardiotoxicity. At the core of the system are microelectronic cell sensor arrays that are integrated into the bottom of 96well microplates (the EPlate Cardio 96). With its much improved data acquisition rate (12.5 millisecond update rate/ plate), the RTCA Cardio Instrument can perform the longterm toxicity assays of the RTCA platform and also temporally resolve the contraction/relaxation cycle of cardiomyocyte beating, which is a surrogate measurement for cardiomyocyte electrical activity. The RTCA Cardio Instrument consists of the following primary components (see Figure 1): RTCA Cardio Control Unit RTCA Cardio Analyzer RTCA Cardio Station To characterize the beating activity, viable icell Cardiomyocytes were plated in the wells of the EPlate Cardio 96 at a density of 30,000 cells/well. The cells were monitored for up to 200 hours in culture (see Figure 2A), and the beating activity was recorded for a duration of 20 seconds every hour. The beating activity and profile at 48, 72, 144, 168, and 192 hours are shown in Figure 2B. Within 48 hours after seeding, icell Cardiomyocytes form a syncytium and synchronous beating activity can be detected (see Figure 2B). The beating rate for these cardiomyocytes can vary between 40 50 beats/minute, which was sustained over the longest observation period of this study (almost two months) in culture with regular changes of culture medium (data not shown). Interestingly, even though synchronized beating activity can be detected as early as 48 hours, the beating profile changes over time, presumably as the cytoskeleton becomes more highly organized, and a stable phenotype is typically reached by Day 8 (see Figure 2B). The RTCA Cardio Software can readily determine parameters such as beating frequency, amplitude, beat duration, time to max (T r ), and decay time (T d ), which are used to characterize specific effects of compound action. EPlate Cardio 96 Figure 1: The xcelligence System RTCA Cardio Instrument. The RTCA Cardio Instrument is a labelfree, realtime system for dynamic monitoring of cardiomyocyte beating and assessment of cardiotoxicity. It is composed of the RTCA Cardio Control Unit, the RTCA Cardio Analyzer, the RTCA Cardio Station (which is placed in a CO 2 incubator), and the EPlate Cardio 96 which contains interdigitated gold microelectrodes fabricated in the bottom of each well. 4

Results continued A Cell Index (CI) 7 6 5 4 3 2 1 0 0 30 60 90 120 150 180 210 Time (hours) Figure 2: Characterization of icell Cardiomyocytes attachment, growth, and beating using the RTCA Cardio Instrument. (A) icell Cardiomyocytes were seeded in the wells of the EPlate Cardio 96 and allowed to adhere and form a syncytium. The cells were cultured for up to 200 hours and monitored by the RTCA Cardio Instrument at regular intervals. (B) Beating activity and Cell Index (CI) profile of icell Cardiomyocytes recorded by the RTCA Cardio Instrument at indicated time points after cell seeding. The beating rate (1/minute) and amplitude (delta CI) were quantified using the RTCA Cardio Software. The data represents the average of 8 wells /+ standard deviation. A total duration of 10 seconds recording time is displayed. B Time After Cultivation (hours) Beating Pattern Beating Frequency (1/minute ± SD) N=8 Amplitude (Delta CI) N=8 Beat Duration (seconds) Time to Max (T r ) (seconds) Decay Time (T d ) (seconds) 48 36 ± 7.1 0.048 ± 0.002 1.45 ± 0.03 0.16 ± 0.007 1.27 ± 0.02 72 26 ± 6.77 0.049 ± 0.008 2.43 ± 0.70 0.16 ± 0.02 1.30 ± 0.38 144 39 ± 4.44 0.046 ± 0.005 1.69 ± 0.06 0.095 ± 0.008 1.55 ± 0.13 168 40 ± 3.2 0.049 ± 0.006 1.45 ± 0.09 0.086 ± 0.005 1.35 ± 0.08 192 49 ± 4.26 0.049 ± 0.006 1.42 ± 0.11 0.078 ± 0.005 1.30 ± 0.12 5

Results continued Pharmacological Assessment of icell Cardiomyocyte Beating Activity Using the xcelligence System RTCA Cardio Instrument Using a pharmacological approach, ion channel and nonion channel targets involved in the regulation of excitation/contraction coupling in icell Cardiomyocytes were evaluated. For these experiments, icell Cardiomyocytes were thawed, seeded in the wells of the EPlate Cardio 96, cultured for 8 days, treated with increasing concentrations of the compounds, and monitored for 12 hours using the RTCA Cardio Instrument. The analysis package of the RTCA Cardio Software was used to quantify beat rate, beat amplitude, beat duration, T r and T d, and beat rate irregularity (a measurement of arrhythmia); the software was also used to derive halfmaximal concentrations (see Table I). Assessment of voltagegated calcium channels Application of isradipine, a wellknown voltageactivated Ltype calcium channel blocker of the dihydropyridine class, caused a progressive time and dosedependent decrease and inhibition of beating activity, indicating that calcium entry through Ltype calcium channels is required for beating (see Figure 3A). The IC 50 for isradipineinduced inhibition of beating activity based on the measurement of the normalized beating rate is 0.78 nm (at the 5minute time point after compound addition; see Table I). These values are consistent with the efficacy of isradipine tested in isolated rabbit heart 4 as well as recombinant HEK293 cells stably expressing the human Cav1.2. 5 Assessment of sodium channel modulators Voltagegated Na + channels are responsible for the Na + current and the depolarization phase of cardiac action potential. Treatment of icell Cardiomyocytes with tetrodotoxin (TTX), a potent and selective inhibitor of voltagegated Na + channels, led to a dosedependent decrease in the beating rate of icell Cardiomyocytes, sustained at the higher concentrations for the entire duration of 12 hours (see Figure 3B). The apparent IC 50 value obtained for inhibition of icell Cardiomyocyte beating is approximately 2.5 µm 3 µm (see Table I). Assessment of potassium channel modulators The rapid activating component of the delayed rectifier current (I Kr ) is involved in the repolarization phase of cardiac action potential and is primarily Parameter Compound Normalized Beating Rate (IC 50 ; M) Normalized Amplitude (IC 50 ; M) Beat Duration (IC 50 ; M) Beat Rate Irregularity (IC 50 ; M) Isradipine 7.8 x 10 10 M 7.8 x 10 10 M 7.8 x 10 10 M. N.A. E4031 4.12 x 10 7 M N.A. 4.02 x 10 8 M 1.19 x 10 9 M Tetrodotoxin (TTX) 2.51 x 10 6 M 3.04 x 10 6 M N.A. N.A. Isoproterenol 6.92 x 10 9 M 7.27 x 10 9 M 7.00 x 10 9 M 3.38 x 10 9 M Table 1: Doseresponse profiling of modulators of ion channel and nonion channel targets in icell Cardiomyocytes. The RTCA Cardio Software was used to analyze dosedependent effects of the indicated compounds on the beating activity of icell Cardiomyocytes and to derive halfmaximal concentrations. The analysis parameters indicated, normalized beating rate, normalized amplitude, beat duration, and beating rhythm irregularity, are part of the analysis package of the RTCA Cardio Software and can be readily used to quantify beating activity. 6

Results continued Figure 3: Pharmacological assessment of ion channel and nonion channel targets expressed in icell Cardiomyocytes. icell Cardiomyocytes were seeded in the wells of the EPlate Cardio 96, monitored for 8 days using the RTCA Cardio Instrument, and treated with increasing concentrations of each compound. The beating activity was recorded by the RTCA Cardio Instrument as described in the Methods section. For each compound at the indicated time points, 10 seconds of beating activity is displayed. The beating rate for each interval of beating activity is displayed as beats/minute ± SD; the data shown is one representative recording from a total of at least 3 separate experiments. (A) isradipine, an Ltype voltagegated calcium channel inhibitor, (B) tetrodotoxin (TTX), inhibitor of voltagegated Na + channels, (C) E4031, an inhibitor of herg type K + channel, (D) isoproterenol, an ionotropic agent and agonist of the badrenergic receptor. A Time (Minutes) Concentration Isradipine 0 5 30 240 720 38.82 0 0 0 0 1.56 nm 45.01 29.48 0 0 0 6.25 nm 25 nm 41.28 0 0 0 0 B Time (Minutes) Concentration Tetrodotoxin (TTX) 0 5 30 240 720 37.77 35.38 44.34 38.24 36.09 100 nm 42.14 17.90 31.06 35.98 26.41 2.5 nm 12.5 nm 40.51 0 0 0 33.70 7

Results continued Figure 3, continued C Time (Minutes) Concentration 12.3 µm E4031 0 5 30 240 720 48.15 30.14 47.20 47.85 48.08 0.11 µm 39.12 28.49 38.94 41.16 39.00 1.00 µm 38.88 21.96 40.97 35.04 62.09 D Time (Minutes) Concentration 8 nm Isoproterenol 0 5 30 240 720 35.56 59.69 73.29 62.58 54.17 40 nm 38.42 65.93 77.24 62.83 55.58 200 nm 35.73 64.46 77.45 58.44 51.23 8

Results continued mediated through the herg channel. The effect of E4031, a potent herg channel inhibitor, was also tested using icell Cardiomyocytes in a time and dosedependent manner (see Figure 3C). As shown, E4031 treatment interrupted the normal rhythm of beating, especially at high concentrations (0.1µM 1 µm), and resulted in prolonged beat durations accompanied by plateau oscillations (see Figure 3C, arrows). This phenomenon was typical of other herg blockers as well (see next section), and may be related to early afterdepolarization. Based on beat duration and beat rate irregularity parameter, the halfmaximal concentrations obtained are 40 nm and 1 nm, respectively (see Table I), consistent with the reported IC 50 for E4031 using embryonic stem (ES) cellderived human cardiomyocytes with the patch clamp technique. 6 Assessment of chronotropic agents Activation of the sympathetic nervous system and neurohormonal regulation through the adrenergic receptor is a major mechanism controlling rate and contractility of the cardiac tissue, and agonists of the adrenergic receptor are wellcharacterized chronotropic and ionotropic agents. 7 Therefore, we sought to test whether the effect of adrenergic receptor stimulation on icell Cardiomyocyte beating could be detected by the RTCA Cardio Instrument. Treatment of icell Cardiomyocytes with isoproterenol, a adrenergic receptor agonist, increased the beating frequency in a dose and timedependent manner, while decreasing the overall duration of each beat (see Figure 3D). The apparent halfmaximal concentration of isoproterenol for the different parameters of beating activity is in the low nanomolar range (see Table I), also consistent with previously reported EC 50 concentrations for human ESCderived cardiomyocytes using electrophysiological techniques. 6 Using the xcelligence System RTCA Cardio Instrument for Preclinical Safety Studies To test the utility of the RTCA Cardio Instrument for preclinical cardiosafety screening, two complementary approaches were undertaken. First, four drugs withdrawn from the market due to increased incidence of TdP were screened in a doseresponse manner using icell Cardiomyocytes. These compounds have subsequently been shown to also directly inhibit herg channel activity. All four compounds significantly affected beating rate in a dosedependent manner (see Figure 4A) and produced beating irregularities that were consistent with those observed for E4031 in terms of morphology, reflecting a common underlying mechanism (see Figure 4A, arrows). This observation indicates that the RTCA Cardio Instrument using icell Cardiomyocytes can identify potentially proarrhythmic compounds. Next, we tested pentamidine, which has been shown to affect the transport of the herg channel to the membrane in heterologous expression systems as well as in cardiomyocytes, with repolarization being delayed as a direct consequence. Since this compound affects herg channel activity indirectly, its effect is manifested in a timedependent manner and is difficult to capture by standard patch clamp techniques, which are limited to only a few minutes of recording time. Administration of pentamidine at a final concentration of 20 µm had no noticeable effect on beating rate and amplitude through the 240minute time point (see Figure 4B). Nine hundred minutes after compound addition, the beating rate slowed down and the beat duration was significantly delayed, most likely due to an extended repolarization phase. These observations highlight the importance of monitoring compound effects in a timedependent manner in order to resolve the impact on both early and long term function of cardiomyocytes, and to obtain greater mechanistic understanding. 9

Results continued A Cisapride Astemizole Droperidol Sertindole B Pentamidine Beating Profile Norm Beating Rate Beat Duration (seconds) Controls 0 min 1.00 1.21 ± 0.16 Low 16 nm 0.08 µm 80 nm 80 nm 5 min 1.00 1.19 ± 0.14 80 nm 0.3 µm 0.4 µm 0.4 µm 30 min 1.16 1.00 ± 0.20 0.4 µm 5 µm 2 µm 2 µm 240 min 0.97 1.42 ± 0.03 900 min 0.52 2.25 ± 0.14 2 µm 20 µm 10 µm 10 µm 1200 min 0.00 0.00 1 s High Figure 4: Mechanismbased cardiotoxicity profiling using the RTCA Cardio Instrument. (A) The indicated drugs, which have been withdrawn from the market due to increased incidence of TdP arrhythmia, were screened in a doseresponse manner in icell Cardiomyocytes. For each compound, a total of 10 seconds of beating activity is displayed. All the compounds were serially diluted 2fold from a 20 μm starting concentration except cisapride, which started at 10 μm. (B) icell Cardiomyocytes were seeded in the wells of the EPlate Cardio 96 and, on Day 8, treated with 20 μm pentamidine. The beating activity was monitored at the indicated time windows after compound treatment and quantified based on beat duration as mentioned in the Methods Section. 10

Discussion In this application note, we introduce a microelectronicbased cardiomyocyte monitoring system (the RTCA Cardio Instrument) that can be used in conjunction with human stem cellderived cardiomyocytes (icell Cardiomyocytes) for assessment of the cardiac safety profile of leading compounds or drug candidates during drug development. The system captures realtime information regarding cardiomyocyte beating, viability, and any morphological changes that may occur as a result of compound treatment. The system s realtime aspect allows for capturing both acute and longterm effects of compound exposure. Most, if not all, in vitro assay systems for cardiac safety are designed to screen for surrogates of arrhythmia, such as herg channel interaction, rather than arrhythmia itself. Assays designed to screen for compounds that may affect repolarization and induce arrhythmia in the context of the whole heart or heart tissue are not implemented until much later in drug development. These include sophisticated, technically demanding, lowthroughput, and costly procedures such as the Purkinje fiber assay, ventricular wedge assay, and the Langendorff whole heart assay, or telemetry experiments in live and anesthetized animals. 1 The field of preclinical cardiac safety can certainly benefit from an assay system that allows for integrated assessment of compound action on ion channel and nonion channel targets involved in cardiac excitationcontraction coupling. The data presented here demonstrates that the RTCA Cardio Instrument in conjunction with icell Cardiomyocytes comprises an assay system providing integrated assessment of compound action on multiple targets involved in heart function. Our data shows that this assay system can sensitively and quantitatively detect the effect of compounds on the major ion channels involved in heart function, namely calcium, sodium, and potassium channels. Furthermore, ionotropic and chronotropic compounds that impact the force and rate of heart beating can also be detected in this system. From a preclinical cardiosafety perspective, drugs pulled off the market due to induction of ventricular arrhythmias or TdP were indeed identified by this assay system in a characteristic manner. These compounds produced a characteristic beating profile that may be indicative of arrhythmia. Another major advantage of the assay system described here is the time resolution. The RTCA Cardio Instrument has a data acquisition rate of 12.5 ms per well of a 96well plate and can be used simultaneously to monitor acute and chronic drug effects up to days and weeks. The utility of the timedependent monitoring of compound action on cardiomyocytes was demonstrated by testing compounds that acutely and directly block herg channels (E4031, cisapride, etc.) and compounds that interfere with protein trafficking (pentamidine) in a subchronic manner. The RTCA Cardio Instrument was able to detect the effect of this compound on the beating rate and duration of icell Cardiomyocytes, which occurred around 4 hours after treatment. Traditional patch clamp experiments most likely would not have been able to detect the adverse effect of this drug on cardiomyocytes. In summary, the xcelligence System RTCA Cardio Instrument, in conjunction with icell Cardiomyocytes, represents a physiologically relevant and predictive assay system for preclinical cardiosafety assessment of lead compounds. The features of this assay system, including time resolution, dynamic monitoring of mechanical beating activity of cardiomyocytes, and 96well throughput, will surely provide additional mechanistic and toxicity information for compound action on the heart. 11

References 1. Fermini B, Fossa AA. (2003). The impact of druginduced QT interval prolongation on drug discovery and development. Nat Rev Drug Discov. 2, 439 447. 2. Brown AM. (2005). HERG block, QT liability and sudden cardiac death. Novartis Found Symp. 266, 118 131; discussion 131 115, 155 118. 3. Kettenhofen R, Bohlen H. (2008). Preclinical assessment of cardiac toxicity. Drug Discovery Today. 13, 702 707. 4. Mellemkjaer S, Bang L, NielsenKudsk F. (1992). Isradipine dynamics and pharmacokinetics in the isolated rabbit heart. Pharmacol Toxicol. 70, 366 372. 5. Balasubramanian B, Imredy JP, Kim D, Penniman J, Lagrutta A, Salata JJ. (2009). Optimization of Ca(v)1.2 screening with an automated planar patch clamp platform. J Pharmacol Toxicol Methods. 59, 62 72. 6. Peng S, Lacerda AE, Kirsch GE, Brown AM, BrueningWright A. (2010). The action potential and comparative pharmacology of stem cellderived human cardiomyocytes. J Pharmacol Toxicol Methods. 61, 277 286. 7. Bers DM. (2002). Cardiac excitationcontraction coupling. Nature. 415, 198 205. Ordering Information Product Cat. No. Pack Size RTCA Cardio System Bundle 00380601060 1 Bundle Package RTCA Cardio Station 06417019001 1 Instrument RTCA Cardio Analyzer 06416993001 1 Instrument RTCA Cardio Control Unit 06200184001 1 Instrument EPlate Cardio 96 06417051001 06417035001 6 Plates 6 x 6 Plates icell Cardiomyocytes see cellulardynamics.com For life science research only. Not for use in diagnostic procedures. XCELLIGENCE, EPLATE, and ACEA BIOSCIENCES are registered trademarks of ACEA Biosciences, Inc. in the US and other countires. BECKMAN COULTER and BIOMEK are trademarks of Beckman Coulter, Inc. All other product names and trademarks are the property of their respective owners. Published by ACEA Biosciences, Inc. 6779 Mesa Ridge Road Ste. 100 San Diego, CA 92121 U.S.A. www.aceabio.com 12 2013 ACEA Biosciences, Inc. All rights reserved.