1
|
El Harchi A, Hancox JC. hERG agonists pose challenges to web-based machine learning methods for prediction of drug-hERG channel interaction. J Pharmacol Toxicol Methods 2023; 123:107293. [PMID: 37468081 DOI: 10.1016/j.vascn.2023.107293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 05/23/2023] [Accepted: 07/12/2023] [Indexed: 07/21/2023]
Abstract
Pharmacological blockade of the IKr channel (hERG) by diverse drugs in clinical use is associated with the Long QT Syndrome that can lead to life threatening arrhythmia. Various computational tools including machine learning models (MLM) for the prediction of hERG inhibition have been developed to facilitate the throughput screening of drugs in development and optimise thus the prediction of hERG liabilities. The use of MLM relies on large libraries of training compounds for the quantitative structure-activity relationship (QSAR) modelling of hERG inhibition. The focus on inhibition omits potential effects of hERG channel agonist molecules and their associated QT shortening risk. It is instructive, therefore, to consider how known hERG agonists are handled by MLM. Here, two highly developed online computational tools for the prediction of hERG liability, Pred-hERG and HergSPred were probed for their ability to detect hERG activator drug molecules as hERG interactors. In total, 73 hERG blockers were tested with both computational tools giving overall good predictions for hERG blockers with reported IC50s below Pred-hERG and HergSPred cut-off threshold for hERG inhibition. However, for compounds with reported IC50s above this threshold such as disopyramide or sotalol discrepancies were observed. HergSPred identified all 20 hERG agonists selected as interacting with the hERG channel. Further studies are warranted to improve online MLM prediction of hERG related cardiotoxicity, by explicitly taking into account channel agonism as well as inhibition.
Collapse
Affiliation(s)
- Aziza El Harchi
- School of Physiology and Pharmacology and Neuroscience, Biomedical Sciences Building, The University of Bristol, University Walk, Bristol BS8 1TD, UK.
| | - Jules C Hancox
- School of Physiology and Pharmacology and Neuroscience, Biomedical Sciences Building, The University of Bristol, University Walk, Bristol BS8 1TD, UK
| |
Collapse
|
2
|
DeMarco KR, Yang PC, Singh V, Furutani K, Dawson JRD, Jeng MT, Fettinger JC, Bekker S, Ngo VA, Noskov SY, Yarov-Yarovoy V, Sack JT, Wulff H, Clancy CE, Vorobyov I. Molecular determinants of pro-arrhythmia proclivity of d- and l-sotalol via a multi-scale modeling pipeline. J Mol Cell Cardiol 2021; 158:163-177. [PMID: 34062207 PMCID: PMC8906354 DOI: 10.1016/j.yjmcc.2021.05.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 05/03/2021] [Accepted: 05/24/2021] [Indexed: 11/20/2022]
Abstract
Drug isomers may differ in their proarrhythmia risk. An interesting example is the drug sotalol, an antiarrhythmic drug comprising d- and l- enantiomers that both block the hERG cardiac potassium channel and confer differing degrees of proarrhythmic risk. We developed a multi-scale in silico pipeline focusing on hERG channel – drug interactions and used it to probe and predict the mechanisms of pro-arrhythmia risks of the two enantiomers of sotalol. Molecular dynamics (MD) simulations predicted comparable hERG channel binding affinities for d- and l-sotalol, which were validated with electrophysiology experiments. MD derived thermodynamic and kinetic parameters were used to build multi-scale functional computational models of cardiac electrophysiology at the cell and tissue scales. Functional models were used to predict inactivated state binding affinities to recapitulate electrocardiogram (ECG) QT interval prolongation observed in clinical data. Our study demonstrates how modeling and simulation can be applied to predict drug effects from the atom to the rhythm for dl-sotalol and also increased proarrhythmia proclivity of d- vs. l-sotalol when accounting for stereospecific beta-adrenergic receptor blocking.
Collapse
Affiliation(s)
- Kevin R DeMarco
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA 95616, USA
| | - Pei-Chi Yang
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA 95616, USA
| | - Vikrant Singh
- Department of Pharmacology, University of California Davis, Davis, CA 95616, USA
| | - Kazuharu Furutani
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA 95616, USA; Department of Pharmacology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima, Tokushima 770-8514, Japan
| | - John R D Dawson
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA 95616, USA; Biophysics Graduate Group, University of California Davis, Davis, CA 95616, USA
| | - Mao-Tsuen Jeng
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA 95616, USA
| | - James C Fettinger
- Department of Chemistry, University of California Davis, Davis, CA 95616, USA
| | - Slava Bekker
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA 95616, USA; Department of Science and Engineering, American River College, Sacramento, CA 95841, USA
| | - Van A Ngo
- Centre for Molecular Simulation and Biochemistry Research Cluster, Department of Biological Sciences, University of Calgary, Calgary, AB T2N1N4, Canada
| | - Sergei Y Noskov
- Centre for Molecular Simulation and Biochemistry Research Cluster, Department of Biological Sciences, University of Calgary, Calgary, AB T2N1N4, Canada
| | - Vladimir Yarov-Yarovoy
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA 95616, USA; Department of Anesthesiology and Pain Medicine, University of California Davis, Davis, CA 95616, USA
| | - Jon T Sack
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA 95616, USA; Department of Anesthesiology and Pain Medicine, University of California Davis, Davis, CA 95616, USA
| | - Heike Wulff
- Department of Pharmacology, University of California Davis, Davis, CA 95616, USA
| | - Colleen E Clancy
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA 95616, USA; Department of Pharmacology, University of California Davis, Davis, CA 95616, USA
| | - Igor Vorobyov
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA 95616, USA; Department of Pharmacology, University of California Davis, Davis, CA 95616, USA.
| |
Collapse
|
3
|
Beserra SS, Santos-Miranda A, Sarmento JO, Miranda VM, Roman-Campos D. Effects of amiodarone on rodent ventricular cardiomyocytes: Novel perspectives from a cellular model of Long QT Syndrome Type 3. Life Sci 2020; 255:117814. [PMID: 32439300 DOI: 10.1016/j.lfs.2020.117814] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 05/04/2020] [Accepted: 05/15/2020] [Indexed: 10/24/2022]
Abstract
AIMS Amiodarone (AMIO) is currently used in medical practice to reverse ventricular tachycardia. Here we determine the effects of AMIO in the electromechanical properties of isolated left ventricle myocyte (LVM) from mice and guinea pig and in a cellular model of Long QT Syndrome Type 3 (LQTS-3) using anemone neurotoxin 2 (ATX II), which induces increase of late sodium current in LVM. MAIN METHODS AND KEY FINDINGS Using patch-clamp technique, fluorescence imaging to detect cellular Ca2+ transient and sarcomere detection systems we evaluate the effect of AMIO in healthy LVM. AMIO produced a significant reduction in the percentage of sarcomere shortening (0.1, 1 and 10 μM) in a range of pacing frequencies, however, without significant attenuation of Ca2+ transient. Also, 10 μM of AMIO caused the opposite effect on action potential repolarization of mouse and guinea pig LVM. When LVM from mouse and guinea pig were paced in a range of pacing frequencies and exposed to ATX (10 nM), AMIO (10 μM) was only able to abrogate electromechanical arrhythmias in LVM from guinea pig at lower pacing frequency. SIGNIFICANCE AMIO has negative inotropic effect with opposite effect on action potential waveform in mouse and guinea pig LVM. Furthermore, the antiarrhythmic action of AMIO in LQTS-3 is species and frequency-dependent, which indicates that AMIO may be beneficial for some types of arrhythmias related to late sodium current.
Collapse
Affiliation(s)
- Samuel Santos Beserra
- Laboratory of CardioBiology, Department of Biophysics, Paulista School of Medicina, Federal University of Sao Paulo, Brazil
| | - Artur Santos-Miranda
- Laboratory of CardioBiology, Department of Biophysics, Paulista School of Medicina, Federal University of Sao Paulo, Brazil
| | - Jaqueline Oliveira Sarmento
- Laboratory of CardioBiology, Department of Biophysics, Paulista School of Medicina, Federal University of Sao Paulo, Brazil
| | - Victor Martins Miranda
- Laboratory of CardioBiology, Department of Biophysics, Paulista School of Medicina, Federal University of Sao Paulo, Brazil
| | - Danilo Roman-Campos
- Laboratory of CardioBiology, Department of Biophysics, Paulista School of Medicina, Federal University of Sao Paulo, Brazil.
| |
Collapse
|
4
|
Observability analysis and state observer design for a cardiac ionic cell model. Comput Biol Med 2020; 125:103910. [PMID: 33035962 DOI: 10.1016/j.compbiomed.2020.103910] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Accepted: 07/04/2020] [Indexed: 11/22/2022]
Abstract
To gain insights into cardiac arrhythmias, researchers have developed and employed various measurement techniques, such as electrocardiography, optical mapping, and patch clamping. However, there are no measurement methods that allow simultaneous recording of all cellular quantities, including intracellular ionic concentrations and gating states, that may play an important role in arrhythmia formation. To help address this shortcoming, we applied observability analysis, a method from control theory, to the Luo-Rudy dynamic (LRd) model of a cardiac ventricular myocyte. After linearizing the time-integrated LRd model about selected periodic orbits, we computed the observability properties of the model to determine whether past system states could be reconstructed from different hypothetical sets of measurements. Under the simplifying assumption that only one dynamical variable could be measured periodically, we found that intracellular potassium concentration generally yielded the largest observability values and thus contained the most information about the dominant modes of the system. The impacts on observability of measurement timings, inter-stimulus interval length, and an alternans-promoting parameter shift were also studied. Pole-placement state observer algorithms were designed and tested in simulations for several scenarios, and we found that it is possible to infer unmeasured variables from potassium-concentration measurements, and to an extent from membrane-potential measurements, both for longer periods that represent normal rhythms and shorter periods associated with tachyarrhythmias. Our results could lead to improved data assimilation algorithms that combine model predictions with measurements to estimate quantities that are difficult or impossible to measure during in vitro experiments.
Collapse
|
5
|
Ahsan F, Mahmood T, Usmani S, Bagga P, Shamim A, Tiwari R, Verma N, Siddiqui MH. A conglomeration of preclinical models related to myocardial infarction. BRAZ J PHARM SCI 2020. [DOI: 10.1590/s2175-97902019000418365] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
| | | | | | | | | | | | - Neeraj Verma
- Hygia Institute of Pharmaceutical Education and Research, India
| | | |
Collapse
|
6
|
Cataldi M, Maurer M, Taglialatela M, Church MK. Cardiac safety of second‐generation H
1
‐antihistamines when updosed in chronic spontaneous urticaria. Clin Exp Allergy 2019; 49:1615-1623. [DOI: 10.1111/cea.13500] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 08/16/2019] [Accepted: 09/02/2019] [Indexed: 01/12/2023]
Affiliation(s)
- Mauro Cataldi
- Section of Pharmacology Department of Neuroscience School of Medicine University of Naples Federico II Naples Italy
| | - Marcus Maurer
- Dermatological Allergology Allergie‐Centrum‐Charité Department of Dermatology and Allergy Charité–Universitätsmedizin Berlin Berlin Germany
| | - Maurizio Taglialatela
- Section of Pharmacology Department of Neuroscience School of Medicine University of Naples Federico II Naples Italy
| | - Martin K. Church
- Dermatological Allergology Allergie‐Centrum‐Charité Department of Dermatology and Allergy Charité–Universitätsmedizin Berlin Berlin Germany
| |
Collapse
|
7
|
Potent hERG channel inhibition by sarizotan, an investigative treatment for Rett Syndrome. J Mol Cell Cardiol 2019; 135:22-30. [PMID: 31362019 PMCID: PMC6856717 DOI: 10.1016/j.yjmcc.2019.07.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Accepted: 07/26/2019] [Indexed: 12/31/2022]
Abstract
Rett Syndrome (RTT) is an X-linked neurodevelopmental disorder associated with respiratory abnormalities and, in up to ~40% of patients, with prolongation of the cardiac QTc interval. QTc prolongation calls for cautious use of drugs with a propensity to inhibit hERG channels. The STARS trial has been undertaken to investigate the efficacy of sarizotan, a 5-HT1A receptor agonist, at correcting RTT respiratory abnormalities. The present study investigated whether sarizotan inhibits hERG potassium channels and prolongs ventricular repolarization. Whole-cell patch-clamp measurements were made at 37 °C from hERG-expressing HEK293 cells. Docking analysis was conducted using a recent cryo-EM structure of hERG. Sarizotan was a potent inhibitor of hERG current (IhERG; IC50 of 183 nM) and of native ventricular IKr from guinea-pig ventricular myocytes. 100 nM and 1 μM sarizotan prolonged ventricular action potential (AP) duration (APD90) by 14.1 ± 3.3% (n = 6) and 29.8 ± 3.1% (n = 5) respectively and promoted AP triangulation. High affinity IhERG inhibition by sarizotan was contingent upon channel gating and intact inactivation. Mutagenesis experiments and docking analysis implicated F557, S624 and Y652 residues in sarizotan binding, with weaker contribution from F656. In conclusion, sarizotan inhibits IKr/IhERG, accessing key binding residues on channel gating. This action and consequent ventricular AP prolongation occur at concentrations relevant to those proposed to treat breathing dysrhythmia in RTT. Sarizotan should only be used in RTT patients with careful evaluation of risk factors for QTc prolongation.
Collapse
|
8
|
Wiśniowska B, Lisowski B, Kulig M, Polak S. Drug interaction at hERG channel: In vitro assessment of the electrophysiological consequences of drug combinations and comparison against theoretical models. J Appl Toxicol 2017; 38:450-458. [DOI: 10.1002/jat.3552] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 09/24/2017] [Accepted: 09/24/2017] [Indexed: 11/11/2022]
Affiliation(s)
- Barbara Wiśniowska
- Pharmacoepidemiology and Pharmacoeconomics Unit, Faculty of Pharmacy; Jagiellonian University Medical College; Medyczna 9, Str., 30-688 Kraków Poland
| | - Bartosz Lisowski
- Pharmacoepidemiology and Pharmacoeconomics Unit, Faculty of Pharmacy; Jagiellonian University Medical College; Medyczna 9, Str., 30-688 Kraków Poland
- M. Smoluchowski Institute of Physics; Jagiellonian University; Kraków Poland
- Department of Biophysics; Jagiellonian University Medical College; Kraków Poland
| | - Magdalena Kulig
- Pharmacoepidemiology and Pharmacoeconomics Unit, Faculty of Pharmacy; Jagiellonian University Medical College; Medyczna 9, Str., 30-688 Kraków Poland
| | - Sebastian Polak
- Pharmacoepidemiology and Pharmacoeconomics Unit, Faculty of Pharmacy; Jagiellonian University Medical College; Medyczna 9, Str., 30-688 Kraków Poland
- Simcyp (part of Certara); S2 4SU Sheffield UK
| |
Collapse
|
9
|
Al-Owais MM, Hettiarachchi NT, Kirton HM, Hardy ME, Boyle JP, Scragg JL, Steele DS, Peers C. A key role for peroxynitrite-mediated inhibition of cardiac ERG (Kv11.1) K + channels in carbon monoxide-induced proarrhythmic early afterdepolarizations. FASEB J 2017; 31:4845-4854. [PMID: 28743763 PMCID: PMC5636698 DOI: 10.1096/fj.201700259r] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 07/05/2017] [Indexed: 12/20/2022]
Abstract
Exposure to CO causes early afterdepolarization arrhythmias. Previous studies in rats have indicated that arrhythmias arose as a result of augmentation of the late Na+ current. The purpose of the present study was to examine the basis for CO-induced arrhythmias in guinea pig myocytes in which action potentials (APs) more closely resemble those of human myocytes. Whole-cell current- and voltage-clamp recordings were made from isolated guinea pig myocytes as well as from human embryonic kidney 293 (HEK293) cells that express wild-type or a C723S mutant form of ether-a-go-go-related gene (ERG; Kv11.1). We also monitored the formation of peroxynitrite (ONOO-) in HEK293 cells fluorimetrically. CO-applied as the CO-releasing molecule, CORM-2-prolonged the APs and induced early afterdepolarizations in guinea pig myocytes. In HEK293 cells, CO inhibited wild-type, but not C723S mutant, Kv11.1 K+ currents. Inhibition was prevented by an antioxidant, mitochondrial inhibitors, or inhibition of NO formation. CO also raised ONOO- levels, an effect that was reversed by the ONOO- scavenger, FeTPPS [5,10,15,20-tetrakis-(4-sulfonatophenyl)-porphyrinato-iron(III)], which also prevented the CO inhibition of Kv11.1 currents and abolished the effects of CO on Kv11.1 tail currents and APs in guinea pig myocytes. Our data suggest that CO induces arrhythmias in guinea pig cardiac myocytes via the ONOO--mediated inhibition of Kv11.1 K+ channels.-Al-Owais, M. M., Hettiarachchi, N. T., Kirton, H. M., Hardy, M. E., Boyle, J. P., Scragg, J. L., Steele, D. S., Peers, C. A key role for peroxynitrite-mediated inhibition of cardiac ERG (Kv11.1) K+ channels in carbon monoxide-induced proarrhythmic early afterdepolarizations.
Collapse
Affiliation(s)
- Moza M Al-Owais
- Division of Cardiovascular and Diabetes Research, Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, United Kingdom; and
| | - Nishani T Hettiarachchi
- Division of Cardiovascular and Diabetes Research, Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, United Kingdom; and
| | - Hannah M Kirton
- Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Matthew E Hardy
- Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - John P Boyle
- Division of Cardiovascular and Diabetes Research, Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, United Kingdom; and
| | - Jason L Scragg
- Division of Cardiovascular and Diabetes Research, Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, United Kingdom; and
| | - Derek S Steele
- Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Chris Peers
- Division of Cardiovascular and Diabetes Research, Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, United Kingdom; and
| |
Collapse
|
10
|
Rubi L, Kovar M, Zebedin-Brandl E, Koenig X, Dominguez-Rodriguez M, Todt H, Kubista H, Boehm S, Hilber K. Modulation of the heart's electrical properties by the anticonvulsant drug retigabine. Toxicol Appl Pharmacol 2017. [PMID: 28641963 DOI: 10.1016/j.taap.2017.06.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Retigabine, currently used as antiepileptic drug, has a wide range of potential medical uses. Administration of the drug in patients can lead to QT interval prolongation in the electrocardiogram and to cardiac arrhythmias in rare cases. This suggests that the drug may perturb the electrical properties of the heart, and the underlying mechanisms were investigated here. Effects of retigabine on currents through human cardiac ion channels, heterologously expressed in tsA-201 cells, were studied in whole-cell patch-clamp experiments. In addition, the drug's impact on the cardiac action potential was tested. This was done using ventricular cardiomyocytes isolated from Langendorff-perfused guinea pig hearts and cardiomyocytes derived from human induced pluripotent stem cells. Further, to unravel potential indirect effects of retigabine on the heart which might involve the autonomic nervous system, membrane potential and noradrenaline release from sympathetic ganglionic neurons were measured in the absence and presence of the drug. Retigabine significantly inhibited currents through hKv11.1 potassium, hNav1.5 sodium, as well as hCav1.2 calcium channels, but only in supra-therapeutic concentrations. In a similar concentration range, the drug shortened the action potential in both guinea pig and human cardiomyocytes. Therapeutic concentrations of retigabine, on the other hand, were sufficient to inhibit the activity of sympathetic ganglionic neurons. We conclude that retigabine- induced QT interval prolongation, and the reported cases of cardiac arrhythmias after application of the drug in a typical daily dose range, cannot be explained by a direct modulatory effect on cardiac ion channels. They are rather mediated by indirect actions at the level of the autonomic nervous system.
Collapse
Affiliation(s)
- Lena Rubi
- Center for Physiology and Pharmacology, Department of Neurophysiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Michael Kovar
- Center for Physiology and Pharmacology, Department of Neurophysiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Eva Zebedin-Brandl
- Center for Physiology and Pharmacology, Department of Neurophysiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Xaver Koenig
- Center for Physiology and Pharmacology, Department of Neurophysiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Manuel Dominguez-Rodriguez
- Center for Physiology and Pharmacology, Department of Neurophysiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Hannes Todt
- Center for Physiology and Pharmacology, Department of Neurophysiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Helmut Kubista
- Center for Physiology and Pharmacology, Department of Neurophysiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Stefan Boehm
- Center for Physiology and Pharmacology, Department of Neurophysiology and Pharmacology, Medical University of Vienna, Vienna, Austria.
| | - Karlheinz Hilber
- Center for Physiology and Pharmacology, Department of Neurophysiology and Pharmacology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
11
|
Hazell L, Raschi E, De Ponti F, Thomas SHL, Salvo F, Ahlberg Helgee E, Boyer S, Sturkenboom M, Shakir S. Evidence for the hERG Liability of Antihistamines, Antipsychotics, and Anti-Infective Agents: A Systematic Literature Review From the ARITMO Project. J Clin Pharmacol 2017; 57:558-572. [PMID: 28019033 DOI: 10.1002/jcph.838] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 10/08/2016] [Indexed: 04/21/2025]
Abstract
A systematic review was performed to categorize the hERG (human ether-a-go-go-related gene) liability of antihistamines, antipsychotics, and anti-infectives and to compare it with current clinical risk of torsade de pointes (TdP). Eligible studies were hERG assays reporting half-minimal inhibitory concentrations (IC50). A "hERG safety margin" was calculated from the IC50 divided by the peak human plasma concentration (free Cmax ). A margin below 30 defined hERG liability. Each drug was assigned an "uncertainty score" based on volume, consistency, precision, and internal and external validity of evidence. The hERG liability was compared to existing knowledge on TdP risk (www.credibledrugs.org). Of 1828 studies, 82 were eligible, allowing calculation of safety margins for 61 drugs. Thirty-one drugs (51%) had evidence of hERG liability including 6 with no previous mention of TdP risk (eg, desloratadine, lopinavir). Conversely, 16 drugs (26%) had no evidence of hERG liability including 6 with known, or at least conditional or possible, TdP risk (eg, chlorpromazine, sulpiride). The main sources of uncertainty were the validity of the experimental conditions used (antihistamines and antipsychotics) and nonuse of reference compounds (anti-infectives). In summary, hERG liability was categorized for 3 widely used drug classes, incorporating a qualitative assessment of the strength of available evidence. Some concordance with TdP risk was observed, although several drugs had hERG liability without evidence of clinical risk and vice versa. This may be due to gaps in clinical evidence, limitations of hERG/Cmax data, or other patient/drug-specific factors that contribute to real-life TdP risk.
Collapse
Affiliation(s)
- Lorna Hazell
- Drug Safety Research Unit, Southampton, United Kingdom
| | - Emanuel Raschi
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Fabrizio De Ponti
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Simon H L Thomas
- Institute of Cellular Medicine, Faculty of Medicine, Newcastle University, Newcastle, United Kingdom
| | - Francesco Salvo
- University of Bordeaux U657, CHU de Bordeaux, Bordeaux, France
| | - Ernst Ahlberg Helgee
- Drug Safety and Metabolism, AstraZeneca Innovative Medicines and Early Development, Mölndal, Sweden
| | - Scott Boyer
- Computational Toxicology, Swedish Toxicology Sciences Research Center, Södertälje, Sweden
| | | | - Saad Shakir
- Drug Safety Research Unit, Southampton, United Kingdom
| |
Collapse
|
12
|
Sube R, Ertel EA. Cardiomyocytes Derived from Human Induced Pluripotent Stem Cells: An In-Vitro Model to Predict Cardiac Effects of Drugs. ACTA ACUST UNITED AC 2017. [DOI: 10.4236/jbise.2017.1011040] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
13
|
Gintant GA, Su Z, Martin RL, Cox BF. Utility of hERG Assays as Surrogate Markers of Delayed Cardiac Repolarization and QT Safety. Toxicol Pathol 2016; 34:81-90. [PMID: 16507548 DOI: 10.1080/01926230500431376] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
HERG (human-ether-a-go-go-related gene) encodes for a cardiac potassium channel that plays a critical role in defining ventricular repolarization. Noncardiovascular drugs associated with a rare but potentially lethal ventricular arrhythmia (Torsades de Pointes) have been linked to delayed cardiac repolarization and block of hERG current. This brief overview will discuss the role of hERG current in cardiac electrophysiology, its involvement in drug-induced delayed repolarization, and approaches used to define drug effects on hERG current. In addition, examples of hERG blocking drugs acting differently (i.e., overt and covert hERG blockade due to multichannel block) together with the utility and limitations of hERG assays as tools to predict the risk of delayed repolarization and proarrhythmia are discussed.
Collapse
Affiliation(s)
- Gary A Gintant
- Deptartment of Integrative Pharmacology, Abbott Laboratories, Abbott Park, Illinois 60064-6119, USA.
| | | | | | | |
Collapse
|
14
|
Pre- and Delayed Treatments With Ranolazine Ameliorate Ventricular Arrhythmias and Nav1.5 Downregulation in Ischemic/Reperfused Rat Hearts. J Cardiovasc Pharmacol 2016; 68:269-279. [DOI: 10.1097/fjc.0000000000000412] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
15
|
Yu Z, Liu J, van Veldhoven JPD, IJzerman AP, Schalij MJ, Pijnappels DA, Heitman LH, de Vries AAF. Allosteric Modulation of Kv11.1 (hERG) Channels Protects Against Drug-Induced Ventricular Arrhythmias. Circ Arrhythm Electrophysiol 2016; 9:e003439. [PMID: 27071825 DOI: 10.1161/circep.115.003439] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 02/04/2016] [Indexed: 12/26/2022]
Abstract
BACKGROUND Ventricular arrhythmias as a result of unintentional blockade of the Kv11.1 (hERG [human ether-à-go-go-related gene]) channel are a major safety concern in drug development. In past years, several highly prescribed drugs have been withdrawn for their ability to cause such proarrhythmia. Here, we investigated whether the proarrhythmic risk of existing drugs could be reduced by Kv11.1 allosteric modulators. METHODS AND RESULTS Using [(3)H]dofetilide-binding assays with membranes of human Kv11.1-expressing human embryonic kidney 293 cells, 2 existing compounds (VU0405601 and ML-T531) and a newly synthesized compound (LUF7244) were found to be negative allosteric modulators of dofetilide binding to the Kv11.1 channel, with LUF7244 showing the strongest effect at 10 μmol/L. The Kv11.1 affinities of typical blockers (ie, dofetilide, astemizole, sertindole, and cisapride) were significantly decreased by LUF7244. Treatment of confluent neonatal rat ventricular myocyte (NRVM) monolayers with astemizole or sertindole caused heterogeneous prolongation of action potential duration and a high incidence of early afterdepolarizations on 1-Hz electric point stimulation, occasionally leading to unstable, self-terminating tachyarrhythmias. Pretreatment of NRVMs with LUF7244 prevented these proarrhythmic effects. NRVM monolayers treated with LUF7244 alone displayed electrophysiological properties indistinguishable from those of untreated NRVM cultures. Prolonged exposure of NRVMs to LUF7244 or LUF7244 plus astemizole did not affect their viability, excitability, and contractility as assessed by molecular, immunological, and electrophysiological assays. CONCLUSIONS Allosteric modulation of the Kv11.1 channel efficiently suppresses drug-induced ventricular arrhythmias in vitro by preventing potentially arrhythmogenic changes in action potential characteristics, raising the possibility to resume the clinical use of unintended Kv11.1 blockers via pharmacological combination therapy.
Collapse
Affiliation(s)
- Zhiyi Yu
- From the Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands (Z.Y., J.P.D.v.V., A.P.I., L.H.H.); Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands (J.L., M.J.S., D.A.P., A.A.F.d.V.); and ICIN-Netherlands Heart Institute, Utrecht, The Netherlands (J.L., A.A.F.d.V.)
| | - Jia Liu
- From the Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands (Z.Y., J.P.D.v.V., A.P.I., L.H.H.); Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands (J.L., M.J.S., D.A.P., A.A.F.d.V.); and ICIN-Netherlands Heart Institute, Utrecht, The Netherlands (J.L., A.A.F.d.V.)
| | - Jacobus P D van Veldhoven
- From the Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands (Z.Y., J.P.D.v.V., A.P.I., L.H.H.); Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands (J.L., M.J.S., D.A.P., A.A.F.d.V.); and ICIN-Netherlands Heart Institute, Utrecht, The Netherlands (J.L., A.A.F.d.V.)
| | - Adriaan P IJzerman
- From the Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands (Z.Y., J.P.D.v.V., A.P.I., L.H.H.); Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands (J.L., M.J.S., D.A.P., A.A.F.d.V.); and ICIN-Netherlands Heart Institute, Utrecht, The Netherlands (J.L., A.A.F.d.V.)
| | - Martin J Schalij
- From the Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands (Z.Y., J.P.D.v.V., A.P.I., L.H.H.); Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands (J.L., M.J.S., D.A.P., A.A.F.d.V.); and ICIN-Netherlands Heart Institute, Utrecht, The Netherlands (J.L., A.A.F.d.V.)
| | - Daniël A Pijnappels
- From the Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands (Z.Y., J.P.D.v.V., A.P.I., L.H.H.); Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands (J.L., M.J.S., D.A.P., A.A.F.d.V.); and ICIN-Netherlands Heart Institute, Utrecht, The Netherlands (J.L., A.A.F.d.V.)
| | - Laura H Heitman
- From the Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands (Z.Y., J.P.D.v.V., A.P.I., L.H.H.); Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands (J.L., M.J.S., D.A.P., A.A.F.d.V.); and ICIN-Netherlands Heart Institute, Utrecht, The Netherlands (J.L., A.A.F.d.V.).
| | - Antoine A F de Vries
- From the Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands (Z.Y., J.P.D.v.V., A.P.I., L.H.H.); Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands (J.L., M.J.S., D.A.P., A.A.F.d.V.); and ICIN-Netherlands Heart Institute, Utrecht, The Netherlands (J.L., A.A.F.d.V.).
| |
Collapse
|
16
|
Pfeiffer ER, Vega R, McDonough PM, Price JH, Whittaker R. Specific prediction of clinical QT prolongation by kinetic image cytometry in human stem cell derived cardiomyocytes. J Pharmacol Toxicol Methods 2016; 81:263-73. [PMID: 27095424 DOI: 10.1016/j.vascn.2016.04.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 04/08/2016] [Accepted: 04/08/2016] [Indexed: 02/06/2023]
Abstract
INTRODUCTION A priority in the development and approval of new drugs is assessment of cardiovascular risk. Current methodologies for screening compounds (e.g. HERG testing) for proarrhythmic risk lead to many false positive and false negative results, resulting in the attrition of potentially therapeutic compounds in early development, and the advancement of other candidates that cause adverse effects. With improvements in the technologies of high content imaging and human stem cell differentiation, it is now possible to directly screen compounds for arrhythmogenic tendencies in human stem cell derived cardiomyocytes (hSC-CMs). METHODS A training panel of 90 compounds consisting of roughly equal numbers of QT-prolonging and negative control (non-QT-prolonging) compounds, and a follow-up blinded study of 35 compounds including 16 from the 90 compound panel and 2 duplicates, were evaluated for prolongation of the calcium transient in hSC-CMs using kinetic image cytometry (KIC), a specialized form of high content analysis. RESULTS The KIC-hSC-CM assay identified training compounds that prolong the calcium transient with 98% specificity, 97% precision, 80% sensitivity, and 89% accuracy in predicting clinical QT prolongation by these compounds. The follow-up study of 35 blinded compounds confirmed the reproducibility and strong diagnostic accuracy of the assay. DISCUSSION The correlation of the KIC-hSC-CM results to clinical observations met or surpassed traditional preclinical assessment of cardiac risk utilizing animal models. Thus, the KIC-hSC-CM assay, which can be accomplished in high throughput and at relatively low cost, is an effective new model system for testing chemicals for cardiovascular risk.
Collapse
Affiliation(s)
| | - Raquel Vega
- Vala Sciences, Inc., San Diego, CA 92121, United States
| | | | | | | |
Collapse
|
17
|
Kumar M, Kasala ER, Bodduluru LN, Dahiya V, Sharma D, Kumar V, Lahkar M. Animal models of myocardial infarction: Mainstay in clinical translation. Regul Toxicol Pharmacol 2016; 76:221-30. [PMID: 26988997 DOI: 10.1016/j.yrtph.2016.03.005] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2015] [Revised: 03/05/2016] [Accepted: 03/07/2016] [Indexed: 01/04/2023]
Abstract
Preclinical models with high prognostic power are a prerequisite for translational research. The closer the similarity of a model to myocardial infarction (MI), the higher is the prognostic value for clinical trials. An ideal MI model should present cardinal signs and pathology that resemble the human disease. The increasing understanding of MI stratification and etiology, however, complicates the choice of animal model for preclinical studies. An ultimate animal model, relevant to address all MI related pathophysiology is yet to be developed. However, many of the existing MI models comprising small and large animals are useful in answering specific questions. An appropriate MI model should be selected after considering both the context of the research question and the model properties. This review addresses the strengths, and limitations of current MI models for translational research.
Collapse
Affiliation(s)
- Mukesh Kumar
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati 781032, Assam, India.
| | - Eshvendar Reddy Kasala
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati 781032, Assam, India.
| | - Lakshmi Narendra Bodduluru
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati 781032, Assam, India.
| | - Vicky Dahiya
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati 781032, Assam, India.
| | - Dinesh Sharma
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati 781032, Assam, India.
| | - Vikas Kumar
- Department of Pharmaceutics, Indian Institute of Technology (BHU), Varanasi 221005, Uttar Pradesh, India.
| | - Mangala Lahkar
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati 781032, Assam, India; Department of Pharmacology, Gauhati Medical College, Guwahati 781032, Assam, India.
| |
Collapse
|
18
|
Obergrussberger A, Bru ggemann A, Goetze TA, Rapedius M, Haarmann C, Rinke I, Becker N, Oka T, Ohtsuki A, Stengel T, Vogel M, Steindl J, Mueller M, Stiehler J, George M, Fertig N. Automated Patch Clamp Meets High-Throughput Screening: 384 Cells Recorded in Parallel on a Planar Patch Clamp Module. ACTA ACUST UNITED AC 2015; 21:779-793. [DOI: 10.1177/2211068215623209] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Indexed: 11/15/2022]
|
19
|
Abo-Salem E, Fowler JC, Attari M, Cox CD, Perez-Verdia A, Panikkath R, Nugent K. Antibiotic-induced cardiac arrhythmias. Cardiovasc Ther 2014; 32:19-25. [PMID: 24428853 DOI: 10.1111/1755-5922.12054] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
This review aims to clarify the underlying risk of arrhythmia associated with the use of macrolides and fluoroquinolones antibiotics. Torsades de pointes (TdP) is a rare potential side effect of fluoroquinolones and macrolide antibiotics. However, the widespread use of these antibiotics compounds the problem. These antibiotics prolong the phase 3 of the action potential and cause early after depolarization and dispersion of repolarization that precipitate TdP. The potency of these drugs, as potassium channel blockers, is very low, and differences between them are minimal. Underlying impaired cardiac repolarization is a prerequisite for arrhythmia induction. Impaired cardiac repolarization can be congenital in the young or acquired in adults. The most important risk factors are a prolonged baseline QTc interval or a combination with class III antiarrhythmic drugs. Modifiable risk factors, including hypokalemia, hypomagnesemia, drug interactions, and bradycardia, should be corrected. In the absence of a major risk factor, the incidence of TdP is very low. The use of these drugs in the appropriate settings of infection should not be altered because of the rare risk of TdP, except among cases with high-risk factors.
Collapse
Affiliation(s)
- Elsayed Abo-Salem
- Department of Cardiovascular Diseases, University of Cincinnati, Cincinnati, OH, USA
| | | | | | | | | | | | | |
Collapse
|
20
|
Effects of selective IKr channel blockade by E-4031 on ventricular electro-mechanical relationship in the halothane-anesthetized dogs. Eur J Pharmacol 2014; 740:263-70. [DOI: 10.1016/j.ejphar.2014.06.064] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Revised: 06/18/2014] [Accepted: 06/23/2014] [Indexed: 11/24/2022]
|
21
|
Holzgrefe H, Ferber G, Champeroux P, Gill M, Honda M, Greiter-Wilke A, Baird T, Meyer O, Saulnier M. Preclinical QT safety assessment: Cross-species comparisons and human translation from an industry consortium. J Pharmacol Toxicol Methods 2014; 69:61-101. [DOI: 10.1016/j.vascn.2013.05.004] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2013] [Revised: 05/08/2013] [Accepted: 05/09/2013] [Indexed: 01/10/2023]
|
22
|
Chain ASY, Dubois VFS, Danhof M, Sturkenboom MCJM, Della Pasqua O. Identifying the translational gap in the evaluation of drug-induced QTc interval prolongation. Br J Clin Pharmacol 2013; 76:708-24. [PMID: 23351036 PMCID: PMC3853530 DOI: 10.1111/bcp.12082] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2012] [Accepted: 01/13/2013] [Indexed: 10/27/2022] Open
Abstract
AIMS Given the similarities in QTc response between dogs and humans, dogs are used in pre-clinical cardiovascular safety studies. The objective of our investigation was to characterize the PKPD relationships and identify translational gaps across species following the administration of three compounds known to cause QTc interval prolongation, namely cisapride, d, l-sotalol and moxifloxacin. METHODS Pharmacokinetic and pharmacodynamic data from experiments in conscious dogs and clinical trials were included in this analysis. First, pharmacokinetic modelling and deconvolution methods were applied to derive drug concentrations at the time of each QT measurement. A Bayesian PKPD model was then used to describe QT prolongation, allowing discrimination of drug-specific effects from other physiological factors known to alter QT interval duration. A threshold of ≥10 ms was used to explore the probability of prolongation after drug administration. RESULTS A linear relationship was found to best describe the pro-arrhythmic effects of cisapride, d,l-sotalol and moxifloxacin both in dogs and in humans. The drug-specific parameter (slope) in dogs was statistically significantly different from humans. Despite such differences, our results show that the probability of QTc prolongation ≥10 ms in dogs nears 100% for all three compounds at the therapeutic exposure range in humans. CONCLUSIONS Our findings indicate that the slope of PKPD relationship in conscious dogs may be used as the basis for the prediction of drug-induced QTc prolongation in humans. Furthermore, the risk of QTc prolongation can be expressed in terms of the probability associated with an increase ≥10 ms, allowing direct inferences about the clinical relevance of the pro-arrhythmic potential of a molecule.
Collapse
Affiliation(s)
- Anne SY Chain
- Leiden/Amsterdam Center for Drug Research, Division of Pharmacology, Leiden UniversityLeiden, the Netherlands
| | - Vincent FS Dubois
- Leiden/Amsterdam Center for Drug Research, Division of Pharmacology, Leiden UniversityLeiden, the Netherlands
| | - Meindert Danhof
- Leiden/Amsterdam Center for Drug Research, Division of Pharmacology, Leiden UniversityLeiden, the Netherlands
| | - Miriam CJM Sturkenboom
- Department of Medical Informatics, Erasmus Medical CenterRotterdam, the Netherlands
- Department of Epidemiology, Erasmus Medical CenterRotterdam, the Netherlands
| | - Oscar Della Pasqua
- Leiden/Amsterdam Center for Drug Research, Division of Pharmacology, Leiden UniversityLeiden, the Netherlands
- Clinical Pharmacology and Discovery Medicine, GlaxoSmithKlineUxbridge, UK
| | - on behalf of the Cardiovascular Safety Project Team, TI Pharma PKPD Platform
- Leiden/Amsterdam Center for Drug Research, Division of Pharmacology, Leiden UniversityLeiden, the Netherlands
- Department of Medical Informatics, Erasmus Medical CenterRotterdam, the Netherlands
- Department of Epidemiology, Erasmus Medical CenterRotterdam, the Netherlands
- Clinical Pharmacology and Discovery Medicine, GlaxoSmithKlineUxbridge, UK
| |
Collapse
|
23
|
DI Veroli GY, Davies MR, Zhang H, Abi-Gerges N, Boyett MR. hERG inhibitors with similar potency but different binding kinetics do not pose the same proarrhythmic risk: implications for drug safety assessment. J Cardiovasc Electrophysiol 2013; 25:197-207. [PMID: 24118558 DOI: 10.1111/jce.12289] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Revised: 08/22/2013] [Accepted: 08/29/2013] [Indexed: 02/02/2023]
Abstract
INTRODUCTION Since the discovery of the link that exists between drug-induced hERG inhibition and Torsade de Pointes (TdP), extreme attention has been given to avoid new drugs inhibiting this channel. hERG inhibition is routinely screened for in new drugs and, typically, IC50 values are compared to projected plasma concentrations to define a safety margin. METHODS AND RESULTS We aimed to show that drugs with similar hERG potency are not uniformly pro-arrhythmic-this depends on the drug binding kinetics and mode of action (trapped or not) rather than the IC50 value only. We used a mathematical model of hERG and its related encoded current IKr to simulate drug binding in different configurations. Expression systems mimicking the screening process were first investigated. hERG model was then incorporated into a canine action potential (AP) and tissue model to study the impact of drug binding configurations on AP and pseudo-ECG (QT interval prolongation). Our data show that: (1) trapped and not trapped configurations and different binding kinetics could be identified during hERG screening; (2) slow binding, not trapped drugs, induced less AP prolongation and minimal QT interval prolongation (4.7%) at a concentration equal to the IC50 whereas maximal pro-arrhythmic risk was observed for trapped drugs at the same concentration (QT interval prolongation, 23.1%). CONCLUSION Our study demonstrates the need for screening for hERG binding configurations rather than potency alone. It also demonstrates the potential link between hERG, drug mode of action and TdP, and the need to question the current regulatory guidance.
Collapse
Affiliation(s)
- Giovanni Y DI Veroli
- Institute of Cardiovascular Sciences, University of Manchester, Manchester, UK; Translational Safety, Drug Safety & Metabolism, AstraZeneca, Manchester, UK
| | | | | | | | | |
Collapse
|
24
|
Zhang L, Cheng L, Hong J. The clinical use of cetirizine in the treatment of allergic rhinitis. Pharmacology 2013; 92:14-25. [PMID: 23867423 DOI: 10.1159/000351843] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Accepted: 03/15/2013] [Indexed: 11/19/2022]
Abstract
BACKGROUND Cetirizine is among the first second-generation H1 antihistamines (SGAHs) developed to provide selective H1 receptor inhibition without central nervous system depression. OBJECTIVE The aim of this review is to summarize the amount of data collected over 25 years of clinical use of cetirizine and compare this with data available for other SGAHs in the management of patients with allergic rhinitis (AR). METHODS A comprehensive literature search for publications relating to cetirizine was performed using the Pubmed database, and relevant papers published in English were selected for detailed review. RESULTS Compared with the majority of other SGAHs, cetirizine was generally shown to have a more favourable pharmacological profile, to be well tolerated, be at least equally or more efficacious in attenuating/inhibiting nasal and ocular symptoms and to improve the quality of life in AR patients. The majority of clinical trials investigating the effect of SGAHs in AR patients further indicated that cetirizine was often employed as the main comparator active drug. CONCLUSION Based on the evidence that cetirizine is a commonly employed active comparator drug in AR, it is tempting to suggest that cetirizine may be a suitable benchmark in the development of novel pharmacotherapies for AR.
Collapse
Affiliation(s)
- Luo Zhang
- Department of Otolaryngology, Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | | | | |
Collapse
|
25
|
Koenig X, Kovar M, Rubi L, Mike AK, Lukacs P, Gawali VS, Todt H, Hilber K, Sandtner W. Anti-addiction drug ibogaine inhibits voltage-gated ionic currents: a study to assess the drug's cardiac ion channel profile. Toxicol Appl Pharmacol 2013; 273:259-68. [PMID: 23707769 PMCID: PMC3853361 DOI: 10.1016/j.taap.2013.05.012] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2012] [Revised: 05/02/2013] [Accepted: 05/04/2013] [Indexed: 01/15/2023]
Abstract
The plant alkaloid ibogaine has promising anti-addictive properties. Albeit not licensed as a therapeutic drug, and despite hints that ibogaine may perturb the heart rhythm, this alkaloid is used to treat drug addicts. We have recently reported that ibogaine inhibits human ERG (hERG) potassium channels at concentrations similar to the drugs affinity for several of its known brain targets. Thereby the drug may disturb the heart's electrophysiology. Here, to assess the drug's cardiac ion channel profile in more detail, we studied the effects of ibogaine and its congener 18-Methoxycoronaridine (18-MC) on various cardiac voltage-gated ion channels. We confirmed that heterologously expressed hERG currents are reduced by ibogaine in low micromolar concentrations. Moreover, at higher concentrations, the drug also reduced human Nav1.5 sodium and Cav1.2 calcium currents. Ion currents were as well reduced by 18-MC, yet with diminished potency. Unexpectedly, although blocking hERG channels, ibogaine did not prolong the action potential (AP) in guinea pig cardiomyocytes at low micromolar concentrations. Higher concentrations (≥ 10 μM) even shortened the AP. These findings can be explained by the drug's calcium channel inhibition, which counteracts the AP-prolonging effect generated by hERG blockade. Implementation of ibogaine's inhibitory effects on human ion channels in a computer model of a ventricular cardiomyocyte, on the other hand, suggested that ibogaine does prolong the AP in the human heart. We conclude that therapeutic concentrations of ibogaine have the propensity to prolong the QT interval of the electrocardiogram in humans. In some cases this may lead to cardiac arrhythmias.
Collapse
Affiliation(s)
- Xaver Koenig
- Center for Physiology and Pharmacology, Department of Neurophysiology and -pharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Himmel HM. Drug-induced functional cardiotoxicity screening in stem cell-derived human and mouse cardiomyocytes: effects of reference compounds. J Pharmacol Toxicol Methods 2013; 68:97-111. [PMID: 23702537 DOI: 10.1016/j.vascn.2013.05.005] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Revised: 05/10/2013] [Accepted: 05/10/2013] [Indexed: 11/30/2022]
Abstract
INTRODUCTION Early prediction of drug-induced functional cardiotoxicity requires robust in-vitro systems suitable for medium/high throughput and easily accessible cardiomyocytes with defined reproducible properties. The xCELLigence Cardio system uses 96-well plates with interdigitated electrodes that detect the impedance changes of rhythmic contractions of stem cell-derived cardiomyocyte (SC-CM) layers. Here, we report on our initial screening experience in comparison to established (multi)cellular and in-vivo models. METHODS Impedance signals from human iPSC-CM (iCells™) and mouse eSC-CM (Cor.At™) were analyzed for contraction amplitude (CA) and duration, rise/fall time, beating rate (BR) and irregularity. RESULTS Following solution exchange, impedance signals re-approximated steady-state conditions after about 2 (Cor.At™) and 3h (iCells™); these time points were used to analyze drug effects. The solvent DMSO (≤1%) hardly influenced contraction parameters in Cor.At™, whereas in iCells™ DMSO (>0.1%) reduced CA and enhanced BR. The selective hERG K⁺ channel blockers E-4031 and dofetilide reduced CA and accelerated BR (≥30 nM) according to the analysis software. The latter, however, was due to burst-like contractions (300 nM) that could be detected only by visual inspection of recordings, and were more pronounced in Cor.At™ as in iCells™. In cardiac myocytes and tissue preparations, however, E4031 and dofetilide have been reported to increase cell shortening and contractile force and to reduce BR. Compounds (pentamidine, HMR1556, ATX2, TTX, and verapamil) with other mechanisms of action were also investigated; their effects differed partially between cell lines (e.g. TTX) and compared to established (multi)cellular models (e.g. HMR1556, ouabain). CONCLUSION Mouse and human stem cell-derived cardiomyocytes respond differently to drugs and these responses occasionally also differ from those originating from established in-vitro and in-vivo models. Hence, drug-induced cardiotoxic effects may be detected with this system, however, the predictive or even translational value of results is considered limited and not yet firmly established.
Collapse
|
27
|
Kumazaki M, Ando H, Kakei M, Ushijima K, Taniguchi Y, Yoshida M, Yamato S, Washino S, Koshimizu TA, Fujimura A. α-Lipoic acid protects against arsenic trioxide-induced acute QT prolongation in anesthetized guinea pigs. Eur J Pharmacol 2013; 705:1-10. [PMID: 23474023 DOI: 10.1016/j.ejphar.2013.02.027] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2012] [Revised: 01/28/2013] [Accepted: 02/05/2013] [Indexed: 10/27/2022]
Abstract
Clinical use of arsenic trioxide (As₂O₃), which can induce the remission of relapsed or refractory acute promyelocytic leukemia, is often limited because of its cardiotoxicity. Symptoms of cardiotoxicity include acute cardiac conduction disturbances, such as QT prolongation. The present study was undertaken to evaluate the effects of α-lipoic acid (LA) on acute As₂O₃-induced ECG abnormalities (QTc interval prolongation) in anesthetized guinea pigs. Intravenous injection of As₂O₃ in guinea pigs caused QTc interval prolongation, which was significantly attenuated by co-treatment with LA (0.35, 3.5 and 35 mg/kg) in a dose-dependent manner. In isolated guinea pig cardiomyocytes, the decrease in IKs current induced by As₂O3 (1 μM) was rapidly restored to the basal level by the addition of LA (10 μM). Consistent with this finding, the As₂O₃-induced QTc interval prolongation was also improved rapidly by post-treatment with LA in guinea pigs. Electrospray ionization time-of-flight mass spectrometry analysis detected an expected peak of arsenic-LA complex in vitro, indicating that LA and As₂O3 form a new compound in vivo. In addition, pre-treatment with a chelating agent, British anti-Lewisite (BAL, 3.5 or 35 mg/kg), also attenuated the As₂O₃-induced QTc interval prolongation. In this study, co- and post-treatments with LA and pre-treatment with BAL ameliorated As₂O₃-induced acute QT prolongation in anesthetized guinea pigs. Because LA and probably BAL may bind to As₂O₃, these agents may exert protective effects through their chelating activity. Further studies are needed to determine whether LA is beneficial as a prophylactic or rescue agent for acute promyelocytic leukemia patients treated with As₂O₃.
Collapse
Affiliation(s)
- Masafumi Kumazaki
- Division of Clinical Pharmacology, Department of Pharmacology, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke-shi, Tochigi 329-0498, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Di Veroli GY, Davies MR, Zhang H, Abi-Gerges N, Boyett MR. High-throughput screening of drug-binding dynamics to HERG improves early drug safety assessment. Am J Physiol Heart Circ Physiol 2013; 304:H104-17. [DOI: 10.1152/ajpheart.00511.2012] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The use of computational models to predict drug-induced changes in the action potential (AP) is a promising approach to reduce drug safety attrition but requires a better representation of more complex drug-target interactions to improve the quantitative prediction. The blockade of the human ether-a-go-go-related gene (HERG) channel is a major concern for QT prolongation and Torsade de Pointes risk. We aim to develop quantitative in-silico AP predictions based on a new electrophysiological protocol (suitable for high-throughput HERG screening) and mathematical modeling of ionic currents. Electrophysiological recordings using the IonWorks device were made from HERG channels stably expressed in Chinese hamster ovary cells. A new protocol that delineates inhibition over time was applied to assess dofetilide, cisapride, and almokalant effects. Dynamic effects displayed distinct profiles for these drugs compared with concentration-effects curves. Binding kinetics to specific states were identified using a new HERG Markov model. The model was then modified to represent the canine rapid delayed rectifier K+ current at 37°C and carry out AP predictions. Predictions were compared with a simpler model based on conductance reduction and were found to be much closer to experimental data. Improved sensitivity to concentration and pacing frequency variables was obtained when including binding kinetics. Our new electrophysiological protocol is suitable for high-throughput screening and is able to distinguish drug-binding kinetics. The association of this protocol with our modeling approach indicates that quantitative predictions of AP modulation can be obtained, which is a significant improvement compared with traditional conductance reduction methods.
Collapse
Affiliation(s)
- Giovanni Y. Di Veroli
- Institute of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom
- Global Safety Assessment, AstraZeneca, United Kingdom
| | - Mark R. Davies
- Clinical Informatics, Research and Development Information, AstraZeneca, United Kingdom
| | - Henggui Zhang
- Biological Physics, University of Manchester, Manchester, United Kingdom
| | | | - Mark R. Boyett
- Institute of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
29
|
Bahl A, Barton P, Bowers K, Brough S, Evans R, Luckhurst CA, Mochel T, Perry MW, Rigby A, Riley RJ, Sanganee H, Sisson A, Springthorpe B. The discovery of CCR3/H1 dual antagonists with reduced hERG risk. Bioorg Med Chem Lett 2012; 22:6688-93. [DOI: 10.1016/j.bmcl.2012.08.124] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Revised: 08/24/2012] [Accepted: 08/26/2012] [Indexed: 10/27/2022]
|
30
|
Cardiac tissue slices with prolonged survival for in vitro drug safety screening. J Pharmacol Toxicol Methods 2012; 66:145-51. [DOI: 10.1016/j.vascn.2011.12.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2011] [Revised: 10/13/2011] [Accepted: 12/19/2011] [Indexed: 11/18/2022]
|
31
|
Goineau S, Legrand C, Froget G. Whole‐Cell Configuration of the Patch‐Clamp Technique in the hERG Channel Assay to Predict the Ability of a Compound to Prolong QT Interval. ACTA ACUST UNITED AC 2012; Chapter 10:Unit 10.15.. [DOI: 10.1002/0471141755.ph1015s57] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
32
|
Himmel HM, Bussek A, Hoffmann M, Beckmann R, Lohmann H, Schmidt M, Wettwer E. Field and action potential recordings in heart slices: correlation with established in vitro and in vivo models. Br J Pharmacol 2012; 166:276-96. [PMID: 22074238 PMCID: PMC3415654 DOI: 10.1111/j.1476-5381.2011.01775.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2011] [Revised: 09/23/2011] [Accepted: 09/29/2011] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND AND PURPOSE Action potential (AP) recordings in ex vivo heart preparations constitute an important component of the preclinical cardiac safety assessment according to the ICH S7B guideline. Most AP measurement models are sensitive, predictive and informative but suffer from a low throughput. Here, effects of selected anti-arrhythmics (flecainide, quinidine, atenolol, sotalol, dofetilide, nifedipine, verapamil) on field/action potentials (FP/AP) of guinea pig and rabbit ventricular slices are presented and compared with data from established in vitro and in vivo models. EXPERIMENTAL APPROACH Data from measurements of membrane currents (hERG, I(Na) ), AP/FP (guinea pig and rabbit ventricular slices), AP (rabbit Purkinje fibre), haemodynamic/ECG parameters (conscious, telemetered dog) were collected, compared and correlated to complementary published data (focused literature search). KEY RESULTS The selected anti-arrhythmics, flecainide, quinidine, atenolol, sotalol, dofetilide, nifedipine and verapamil, influenced the shape of AP/FP of guinea pig and rabbit ventricular slices in a manner similar to that observed for rabbit PF. The findings obtained from slice preparations are in line with measurements of membrane currents in vitro, papillary muscle AP in vitro and haemodynamic/ECG parameters from conscious dogs in vivo, and were also corroborated by published data. CONCLUSION AND IMPLICATIONS FP and AP recordings from heart slices correlated well with established in vitro and in vivo models in terms of pharmacology and predictability. Heart slice preparations yield similar results as papillary muscle but offer enhanced throughput for mechanistic investigations and may substantially reduce the use of laboratory animals.
Collapse
|
33
|
Tan Y, Chen Y, You Q, Sun H, Li M. Predicting the potency of hERG K+ channel inhibition by combining 3D-QSAR pharmacophore and 2D-QSAR models. J Mol Model 2011; 18:1023-36. [DOI: 10.1007/s00894-011-1136-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2011] [Accepted: 05/23/2011] [Indexed: 02/06/2023]
|
34
|
Antzelevitch C, Burashnikov A, Sicouri S, Belardinelli L. Electrophysiologic basis for the antiarrhythmic actions of ranolazine. Heart Rhythm 2011; 8:1281-90. [PMID: 21421082 DOI: 10.1016/j.hrthm.2011.03.045] [Citation(s) in RCA: 183] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2011] [Accepted: 03/11/2011] [Indexed: 12/19/2022]
Abstract
Ranolazine is a Food and Drug Administration-approved antianginal agent. Experimental and clinical studies have shown that ranolazine has antiarrhythmic effects in both ventricles and atria. In the ventricles, ranolazine can suppress arrhythmias associated with acute coronary syndrome, long QT syndrome, heart failure, ischemia, and reperfusion. In atria, ranolazine effectively suppresses atrial tachyarrhythmias and atrial fibrillation (AF). Recent studies have shown that the drug may be effective and safe in suppressing AF when used as a pill-in-the pocket approach, even in patients with structurally compromised hearts, warranting further study. The principal mechanism underlying ranolazine's antiarrhythmic actions is thought to be primarily via inhibition of late I(Na) in the ventricles and via use-dependent inhibition of peak I(Na) and I(Kr) in the atria. Short- and long-term safety of ranolazine has been demonstrated in the clinic, even in patients with structural heart disease. This review summarizes the available data regarding the electrophysiologic actions and antiarrhythmic properties of ranolazine in preclinical and clinical studies.
Collapse
|
35
|
Andersson B, Abi-Gerges N, Carlsson L. The combined ion channel blocker AZD1305 attenuates late Na current and IKr-induced action potential prolongation and repolarization instability. Europace 2010; 12:1003-10. [DOI: 10.1093/europace/euq070] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
36
|
Mewe M, Mauerhöfer M, Wulfsen I, Szlachta K, Zhou XB, Schwarz JR, Bauer CK. Modulation of cardiac ERG1 K(+) channels by cGMP signaling. J Mol Cell Cardiol 2010; 49:48-57. [PMID: 20188738 DOI: 10.1016/j.yjmcc.2010.02.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2009] [Revised: 02/16/2010] [Accepted: 02/17/2010] [Indexed: 12/21/2022]
Abstract
Different K(+) currents have been implicated in the myocardial action potential repolarization including the I(Kr). ERG1 alpha subunits, identified as the molecular correlate of I(Kr), have been shown to form heteromultimeric channels in the heart and their activity is modulated by a complex interplay of signal transduction events. Using electrophysiological techniques, we examined the effects of the cGMP-analogue 8-Br-cGMP on rat and guinea-pig papillary action potential duration (APD), on the biophysical properties of heterologously expressed homo- and heteromeric ERG1 channels, and on cardiac I(Kr). 8-Br-cGMP prolonged APD by about 25% after pharmacological inhibition of L-type Ca(2+) currents and I(Ks). The prolongation was completely abolished by prior application of the hERG channel blocker E-4031 or the protein kinase G (PKG) inhibitor Rp-8-Br-cGMPS. Expression analysis revealed the presence of both ERG1a and -1b subunits in rat papillary muscle. Both 8-Br-cGMP and ANP inhibited heterologously expressed ERG1b and even stronger ERG1a/1b channels, whereas ERG1a channels remained unaffected. The inhibitory 8-Br-cGMP effects were PKG-dependent and involved a profound ERG current reduction, which was also observed with cardiac AP clamp recordings. Measurements of I(Kr) from isolated mouse cardiomyocytes using Cs(+) as charge carrier exhibited faster deactivation kinetics in atrial than in ventricular myocytes consistent with a higher relative expression of ERG1b transcripts in atria than in ventricles. 8-Br-cGMP significantly reduced I(Kr) in atrial, but not in ventricular myocytes. These findings provide first evidence that through heteromeric assembly ERG1 channels become a critical target of cGMP-PKG signaling linking cGMP accumulation to cardiac I(Kr) modulation.
Collapse
Affiliation(s)
- Marco Mewe
- Institute of Pharmacology for Pharmacists, University Medical Center, Hamburg-Eppendorf (UKE), Martinistr. 52, D-20246 Hamburg, Germany.
| | | | | | | | | | | | | |
Collapse
|
37
|
Coi A, Massarelli I, Saraceno M, Carli N, Testai L, Calderone V, Bianucci AM. Quantitative Structure-Activity Relationship Models for Predicting Biological Properties, Developed by Combining Structure- and Ligand-Based Approaches: An Application to the Human Ether-a-go-go-Related Gene Potassium Channel Inhibition. Chem Biol Drug Des 2009; 74:416-33. [DOI: 10.1111/j.1747-0285.2009.00873.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
38
|
Abi-Gerges N, Valentin JP, Pollard CE. Dog left ventricular midmyocardial myocytes for assessment of drug-induced delayed repolarization: short-term variability and proarrhythmic potential. Br J Pharmacol 2009; 159:77-92. [PMID: 19663882 DOI: 10.1111/j.1476-5381.2009.00338.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND AND PURPOSE Evaluation of the potential for delayed ventricular repolarization and proarrhythmia by new drugs is essential. We investigated if dog left ventricular midmyocardial myocytes (LVMMs) that can be used as a preclinical model to assess drug effects on action potential duration (APD) and whether in these cells, short-term variability (STV) or triangulation could predict proarrhythmic potential. EXPERIMENTAL APPROACH Beagle LVMMs and Purkinje fibres (PFs) were used to record APs. Effects of six reference drugs were assessed on APD at 50% (APD(50)) and 90% (APD(90)) of repolarization, STV(APD), triangulation (ratio APD(90)/APD(50)) and incidence of early afterdepolarizations (EADs) at 1 and 0.5 Hz. KEY RESULTS LVMMs provided stable recordings of AP, which were not affected by four sequential additions of dimethyl sulphoxide. Effects of dofetilide, d-sotalol, cisapride, pinacidil and diltiazem, but not of terfenadine, on APD in LVMMs were found to be comparable with those recorded in PFs. LVMMs, but not PFs, exhibited a proarrhythmic response to I(Kr) blockers. Incidence of EADs was not related to differences in AP prolongation or triangulation, but corresponded to beat-to-beat variability of repolarization, here quantified as STV of APD. CONCLUSIONS AND IMPLICATIONS LVMMs provide a suitable preclinical model to assess the effects of new drugs on APD and also yield additional information about putative indicators of proarrhythmia that add value to an integrated QT/TdP risk assessment. Our findings support the concept that increased STV(APD) may predict drug-induced proarrhythmia.
Collapse
Affiliation(s)
- Najah Abi-Gerges
- Safety Pharmacology Department, Safety Assessment UK, AstraZeneca R&D, Mereside, Alderley Park, Macclesfield, Cheshire, UK.
| | | | | |
Collapse
|
39
|
Polak S, Wiśniowska B, Brandys J. Collation, assessment and analysis of literature in vitro data on hERG receptor blocking potency for subsequent modeling of drugs' cardiotoxic properties. J Appl Toxicol 2009; 29:183-206. [PMID: 18988205 DOI: 10.1002/jat.1395] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The assessment of the torsadogenic potency of a new chemical entity is a crucial issue during lead optimization and the drug development process. It is required by the regulatory agencies during the registration process. In recent years, there has been a considerable interest in developing in silico models, which allow prediction of drug-hERG channel interaction at the early stage of a drug development process. The main mechanism underlying an acquired QT syndrome and a potentially fatal arrhythmia called torsades de pointes is the inhibition of potassium channel encoded by hERG (the human ether-a-go-go-related gene). The concentration producing half-maximal block of the hERG potassium current (IC(50)) is a surrogate marker for proarrhythmic properties of compounds and is considered a test for cardiac safety of drugs or drug candidates. The IC(50) values, obtained from data collected during electrophysiological studies, are highly dependent on experimental conditions (i.e. model, temperature, voltage protocol). For the in silico models' quality and performance, the data quality and consistency is a crucial issue. Therefore the main objective of our work was to collect and assess the hERG IC(50) data available in accessible scientific literature to provide a high-quality data set for further studies.
Collapse
Affiliation(s)
- Sebastian Polak
- Toxicology Department, Faculty of Pharmacy, Medical Collage, Jagiellonian University, Poland.
| | | | | |
Collapse
|
40
|
Hardy MEL, Pollard CE, Small BG, Bridgland-Taylor M, Woods AJ, Valentin JP, Abi-Gerges N. Validation of a voltage-sensitive dye (di-4-ANEPPS)-based method for assessing drug-induced delayed repolarisation in beagle dog left ventricular midmyocardial myocytes. J Pharmacol Toxicol Methods 2009; 60:94-106. [PMID: 19414070 DOI: 10.1016/j.vascn.2009.03.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2009] [Accepted: 03/10/2009] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Evaluation of drug candidates in in-vitro assays of action potential duration (APD) is one component of preclinical safety assessment. Current assays are limited by technically-demanding, time-consuming electrophysiological methods. This study aimed to assess whether a voltage-sensitive dye-based assay could be used instead. METHODS Optical APs were recorded using di-4-ANEPPS in electrically field stimulated beagle left ventricular midmyocardial myocytes (LVMMs). Pharmacological properties of di-4-ANEPPS on the main cardiac ion channels that shape the ventricular AP were investigated using IonWorks and conventional electrophysiology. Effects of 9 reference drugs (dofetilide, E4031, D-sotalol, ATXII, cisapride, terfenadine, alfuzosin, diltiazem and pinacidil) with known APD-modulating effects were assessed on optically measured APD at 1 Hz. RESULTS Under optimum conditions, 0.1 microM di-4-ANEPPS could be used to monitor APs paced at 1 Hz during nine, 5 s exposures without altering APD. di-4-ANEPPS had no effect on either hI(ERG), hI(Na), hI(Ks) and hI(to) currents in transfected CHO cells (up to 10 microM) or I(Ca,L) current in LVMMs (at 16 microM). di-4-ANEPPS had no effect on APs recorded with microelectrodes at 1 or 0.5 Hz over a period of 30 min di-4-ANEPPS displayed the sensitivity to record changes in optically measured APD in response to altered pacing frequencies and sequential vehicle additions did not affect the optically measured APD. APD data obtained with 9 reference drugs were as expected except (i) D-sotalol-induced increases in duration were smaller than those caused by other I(Kr) blockers and (ii) increases in APD were not detected using low concentrations of terfenadine. DISCUSSION Early in drug discovery, the di-4-ANEPPS-based method can reliably be used to assess drug effects on APD as part of a cardiac risk assessment strategy.
Collapse
Affiliation(s)
- Matthew E L Hardy
- Safety Pharmacology Department, Safety Assessment UK, AstraZeneca R&D, Alderley Park, Macclesfield, Cheshire SK104TG, UK
| | | | | | | | | | | | | |
Collapse
|
41
|
Zhou Q, Zygmunt AC, Cordeiro JM, Siso-Nadal F, Miller RE, Buzzard GT, Fox JJ. Identification of Ikr kinetics and drug binding in native myocytes. Ann Biomed Eng 2009; 37:1294-309. [PMID: 19353268 PMCID: PMC2690829 DOI: 10.1007/s10439-009-9690-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2008] [Accepted: 03/27/2009] [Indexed: 12/19/2022]
Abstract
Determining the effect of a compound on IKr is a standard screen for drug safety. Often the effect is described using a single IC50 value, which is unable to capture complex effects of a drug. Using verapamil as an example, we present a method for using recordings from native myocytes at several drug doses along with qualitative features of IKr from published studies of HERG current to estimate parameters in a mathematical model of the drug effect on IKr. IKr was recorded from canine left ventricular myocytes using ruptured patch techniques. A voltage command protocol was used to record tail currents at voltages from −70 to −20 mV, following activating pulses over a wide range of voltages and pulse durations. Model equations were taken from a published IKr Markov model and the drug was modeled as binding to the open state. Parameters were estimated using a combined global and local optimization algorithm based on collected data with two additional constraints on IKrI–V relation and IKr inactivation. The method produced models that quantitatively reproduce both the control IKr kinetics and dose dependent changes in the current. In addition, the model exhibited use and rate dependence. The results suggest that: (1) the technique proposed here has the practical potential to develop data-driven models that quantitatively reproduce channel behavior in native myocytes; (2) the method can capture important drug effects that cannot be reproduced by the IC50 method. Although the method was developed for IKr, the same strategy can be applied to other ion channels, once appropriate channel-specific voltage protocols and qualitative features are identified.
Collapse
Affiliation(s)
- Qinlian Zhou
- Gene Network Sciences, 58 Charles Street, Cambridge, MA 02141, USA.
| | | | | | | | | | | | | |
Collapse
|
42
|
Nakamura Y, Takahara A, Sugiyama A. Famotidine neither affects action potential parameters nor inhibits human ether-a-go-go-related gene (hERG) K + current. J Toxicol Sci 2009; 34:563-7. [DOI: 10.2131/jts.34.563] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Affiliation(s)
- Yuji Nakamura
- Department of Pharmacology, Yamanashi Research Center of Clinical Pharmacology, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi
| | - Akira Takahara
- Department of Pharmacology, Yamanashi Research Center of Clinical Pharmacology, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi
| | - Atsushi Sugiyama
- Department of Pharmacology, Yamanashi Research Center of Clinical Pharmacology, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi
| |
Collapse
|
43
|
Coi A, Massarelli I, Testai L, Calderone V, Bianucci AM. Identification of “toxicophoric” features for predicting drug-induced QT interval prolongation. Eur J Med Chem 2008; 43:2479-88. [DOI: 10.1016/j.ejmech.2007.12.025] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2007] [Revised: 12/07/2007] [Accepted: 12/11/2007] [Indexed: 11/26/2022]
|
44
|
Kettenhofen R, Bohlen H. Preclinical assessment of cardiac toxicity. Drug Discov Today 2008; 13:702-7. [PMID: 18602016 DOI: 10.1016/j.drudis.2008.06.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2008] [Revised: 06/02/2008] [Accepted: 06/04/2008] [Indexed: 10/21/2022]
Abstract
The exact prediction of the clinical behavior of drugs represents one of the most difficult duties in preclinical drug development. The use of cell-based assay systems underpins the development of many drug candidates, but owing to the artificial character of many of these systems, cell response and physiological behavior seem to be mutually exclusive. Embryonic stem cell-derived cells represent a system that may address the disconnect between the behavior of cultured cells and cells in situ. While undifferentiated ES cells allow standardization, expansion and genetic manipulation, the differentiated cells provide a reflection of the normal physiological image of their primary counterpart. We compare common models to detect cardiac toxicity with an assay system comprising in vitro differentiated pure cardiomyocytes.
Collapse
|
45
|
Hancox JC, McPate MJ, El Harchi A, Zhang YH. The hERG potassium channel and hERG screening for drug-induced torsades de pointes. Pharmacol Ther 2008; 119:118-32. [PMID: 18616963 DOI: 10.1016/j.pharmthera.2008.05.009] [Citation(s) in RCA: 206] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2008] [Accepted: 05/27/2008] [Indexed: 01/08/2023]
Abstract
Drug-induced torsades de pointes (TdP) arrhythmia is a major safety concern in the process of drug design and development. The incidence of TdP tends to be low, so early pre-clinical screens rely on surrogate markers of TdP to highlight potential problems with new drugs. hERG (human ether-à-go-go-related gene, alternative nomenclature KCNH2) is responsible for channels mediating the 'rapid' delayed rectifier K+ current (IKr) which plays an important role in ventricular repolarization. Pharmacological inhibition of native IKr and of recombinant hERG channels is a shared feature of diverse drugs associated with TdP. In vitro hERG assays therefore form a key element of an integrated assessment of TdP liability, with patch-clamp electrophysiology offering a 'gold standard'. However, whilst clearly necessary, hERG assays cannot be assumed automatically to provide sufficient information, when considered in isolation, to differentiate 'safe' from 'dangerous' drugs. Other relevant factors include therapeutic plasma concentration, drug metabolism and active metabolites, severity of target condition and drug effects on other cardiac ion channels that may mitigate or exacerbate effects of hERG blockade. Increased understanding of the nature of drug-hERG channel interactions may ultimately help eliminate potential hERG blockade early in the design and development process. Currently, for promising drug candidates integration of data from hERG assays with information from other pre-clinical safety screens remains essential.
Collapse
Affiliation(s)
- Jules C Hancox
- Department of Physiology and Pharmacology, Cardiovascular Research Laboratories, Bristol Heart Institute, School of Medical Sciences, The University of Bristol, University Walk, Bristol, BS8 1TD, United Kingdom.
| | | | | | | |
Collapse
|
46
|
Inanobe A, Kamiya N, Murakami S, Fukunishi Y, Nakamura H, Kurachi Y. In Silico Prediction of the Chemical Block of Human Ether-a-Go-Go-Related Gene (hERG) K+ Current. J Physiol Sci 2008. [DOI: 10.2170/physiolsci.rv011408] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
47
|
Améen C, Strehl R, Björquist P, Lindahl A, Hyllner J, Sartipy P. Human embryonic stem cells: current technologies and emerging industrial applications. Crit Rev Oncol Hematol 2007; 65:54-80. [PMID: 17689256 DOI: 10.1016/j.critrevonc.2007.06.012] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2007] [Revised: 06/11/2007] [Accepted: 06/27/2007] [Indexed: 12/28/2022] Open
Abstract
The efficiency and accuracy of the drug development process is severely restricted by the lack of functional human cell systems. However, the successful derivation of pluripotent human embryonic stem (hES) cell lines in the late 1990s is expected to revolutionize biomedical research in many areas. Due to their growth capacity and unique developmental potential to differentiate into almost any cell type of the human body, hES cells have opened novel avenues both in basic and applied research as well as for therapeutic applications. In this review we describe, from an industrial perspective, the basic science that underlies the hES cell technology and discuss the current and future prospects for hES cells in novel and improved stem cell based applications for drug discovery, toxicity testing as well as regenerative medicine.
Collapse
Affiliation(s)
- Caroline Améen
- Cellartis AB, Arvid Wallgrens Backe 20, 413 46 Göteborg, Sweden
| | | | | | | | | | | |
Collapse
|
48
|
Humphrey SJ, Turman CN, Curry JT, Wheeler GJ. Cardiovascular and electrocardiographic effects of the dopamine receptor agonists ropinirole, apomorphine, and PNU-142774E in conscious beagle dogs. J Cardiovasc Pharmacol 2006; 47:337-47. [PMID: 16633074 DOI: 10.1097/01.fjc.0000205983.05771.f5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
To confirm recent in vitro findings, we examined the cardiovascular and electrocardiographic (ECG) effects of the dopamine receptor agonists ropinirole, apomorphine, and PNU-142774E in conscious dogs. Intravenous (i.v.) infusions of ropinirole totaling 20 microg/kg maximally reduced mean arterial pressure (MAP; -16 mm Hg) and the ECG PR interval (-13 milliseconds) and increased heart rate (HR; +29 b/min) and QTc length (+33 ms) at a peak plasma drug concentration (p[drug]) of 3.5 ng/ml. I.V. PNU-142774E was better tolerated through 66 microg/kg and a maximal p[drug] of 5.9 ng/ml with negligible cardiovascular changes and mild QTc reduction (13 ms). Apomorphine (25 microg/kg i.v.) was intermediate to ropinirole and PNU-142774E for emesis and peak changes in MAP (-6 mm Hg), HR (+24 b/min), and QTc (+15 milliseconds) at a mean p[drug] of 3.4 ng/ml. By comparison, the class III antiarrhythmic trecetilide (2.0 mg/kg bolus) increased QTc (+58 ms) without affecting mean arterial pressure or heart rate. This study establishes that in conscious dogs, the selective dopamine receptor agonist PNU-142774E has fewer cardiovascular and emetic effects than ropinirole and apomorphine and supports prior in vitro findings that ropinirole and apomorphine but not the PNU-142774E imidazoquinolin analog sumanirole reduces the delayed rectifier current in HERG transfected cells.
Collapse
Affiliation(s)
- Stephen J Humphrey
- Department of Pharmacology, Global Pharmaceutical Research and Development, Pfizer Inc., Kalamazoo, Michigan, USA.
| | | | | | | |
Collapse
|
49
|
Vandenberg JI, Varghese A, Lu Y, Bursill JA, Mahaut-Smith MP, Huang CLH. Temperature dependence of human ether-à-go-go-related gene K+ currents. Am J Physiol Cell Physiol 2006; 291:C165-75. [PMID: 16452156 DOI: 10.1152/ajpcell.00596.2005] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The function of voltage-gated human ether-à-go-gorelated gene ( hERG) K+ channels is critical for both normal cardiac repolarization and suppression of arrhythmias initiated by premature excitation. These important functions are facilitated by their unusual kinetics that combine relatively slow activation and deactivation with rapid and voltage-dependent inactivation and recovery from inactivation. The thermodynamics of these unusual features were examined by exploring the effect of temperature on the activation and inactivation processes of hERG channels expressed in Chinese hamster ovary cells. Increased temperature shifted the voltage dependence of activation in the hyperpolarizing direction but that of inactivation in the depolarizing direction. This increases the relative occupancy of the open state and contributes to the marked temperature sensitivity of hERG current magnitude observed during action potential voltage clamps. The rates of activation and deactivation also increase with higher temperatures, but less markedly than do the rates of inactivation and recovery from inactivation. Our results also emphasize that one cannot extrapolate results obtained at room temperature to 37°C by using a single temperature scale factor.
Collapse
Affiliation(s)
- Jamie I Vandenberg
- Victor Chang Cardiac Research Institute, Level 9, 384 Victoria St., Darlinghurst, New South Wales 2010, Australia.
| | | | | | | | | | | |
Collapse
|
50
|
Abstract
Pharmaceutical companies try to develop new drugs that have a high success rate of reaching the market. However, current disease models lack a strong correlation to clinical reality, because of the underestimation of the complexity and variability of clinical disease processes. This leads to high attrition rates late in drug development and soaring costs. Improvement of disease models is an important issue to reduce the high attrition rates in drug development. Using cell-based disease models, which should take into account the molecular diversity of the human cytome, will improve the predictive value of drug discovery.
Collapse
|