1
|
Patel MV, Peltier HM, Matulenko MA, Koenig JR, C Scanio MJ, Gum RJ, El-Kouhen OF, Fricano MM, Lundgaard GL, Neelands T, Zhang XF, Zhan C, Pai M, Ghoreishi-Haack N, Hudzik T, Gintant G, Martin R, McGaraughty S, Xu J, Bow D, Kalvass JC, Kym PR, DeGoey DA, Kort ME. Discovery of (R)-(3-fluoropyrrolidin-1-yl)(6-((5-(trifluoromethyl)pyridin-2-yl)oxy)quinolin-2-yl)methanone (ABBV-318) and analogs as small molecule Na v1.7/ Nav1.8 blockers for the treatment of pain. Bioorg Med Chem 2022; 63:116743. [PMID: 35436748 DOI: 10.1016/j.bmc.2022.116743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 04/01/2022] [Accepted: 04/02/2022] [Indexed: 11/24/2022]
Abstract
The voltage-gated sodium channel Nav1.7 is an attractive target for the treatment of pain based on the high level of target validation with genetic evidence linking Nav1.7 to pain in humans. Our effort to identify selective, CNS-penetrant Nav1.7 blockers with oral activity, improved selectivity, good drug-like properties, and safety led to the discovery of 2-substituted quinolines and quinolones as potent small molecule Nav1.7 blockers. The design of these molecules focused on maintaining potency at Nav1.7, improving selectivity over the hERG channel, and overcoming phospholipidosis observed with the initial leads. The structure-activity relationship (SAR) studies leading to the discovery of (R)-(3-fluoropyrrolidin-1-yl)(6-((5-(trifluoromethyl)pyridin-2-yl)oxy)quinolin-2-yl)methanone (ABBV-318) are described herein. ABBV-318 displayed robust in vivo efficacy in both inflammatory and neuropathic rodent models of pain. ABBV-318 also inhibited Nav1.8, another sodium channel isoform that is an active target for the development of new pain treatments.
Collapse
Affiliation(s)
- Meena V Patel
- AbbVie, Research and Development, 1 North Waukegan Road, North Chicago, IL 60064, USA.
| | - Hillary M Peltier
- AbbVie, Research and Development, 1 North Waukegan Road, North Chicago, IL 60064, USA
| | - Mark A Matulenko
- AbbVie, Research and Development, 1 North Waukegan Road, North Chicago, IL 60064, USA
| | - John R Koenig
- AbbVie, Research and Development, 1 North Waukegan Road, North Chicago, IL 60064, USA
| | - Marc J C Scanio
- AbbVie, Research and Development, 1 North Waukegan Road, North Chicago, IL 60064, USA
| | - Rebecca J Gum
- AbbVie, Research and Development, 1 North Waukegan Road, North Chicago, IL 60064, USA
| | - Odile F El-Kouhen
- AbbVie, Research and Development, 1 North Waukegan Road, North Chicago, IL 60064, USA
| | - Meagan M Fricano
- AbbVie, Research and Development, 1 North Waukegan Road, North Chicago, IL 60064, USA
| | - Greta L Lundgaard
- AbbVie, Research and Development, 1 North Waukegan Road, North Chicago, IL 60064, USA
| | - Torben Neelands
- AbbVie, Research and Development, 1 North Waukegan Road, North Chicago, IL 60064, USA
| | - Xu-Feng Zhang
- AbbVie, Research and Development, 1 North Waukegan Road, North Chicago, IL 60064, USA
| | - Cenchen Zhan
- AbbVie, Research and Development, 1 North Waukegan Road, North Chicago, IL 60064, USA
| | - Madhavi Pai
- AbbVie, Research and Development, 1 North Waukegan Road, North Chicago, IL 60064, USA
| | | | - Thomas Hudzik
- AbbVie, Research and Development, 1 North Waukegan Road, North Chicago, IL 60064, USA
| | - Gary Gintant
- AbbVie, Research and Development, 1 North Waukegan Road, North Chicago, IL 60064, USA
| | - Ruth Martin
- AbbVie, Research and Development, 1 North Waukegan Road, North Chicago, IL 60064, USA
| | - Steve McGaraughty
- AbbVie, Research and Development, 1 North Waukegan Road, North Chicago, IL 60064, USA
| | - Jun Xu
- AbbVie, Research and Development, 1 North Waukegan Road, North Chicago, IL 60064, USA
| | - Daniel Bow
- AbbVie, Research and Development, 1 North Waukegan Road, North Chicago, IL 60064, USA
| | - John C Kalvass
- AbbVie, Research and Development, 1 North Waukegan Road, North Chicago, IL 60064, USA
| | - Philip R Kym
- AbbVie, Research and Development, 1 North Waukegan Road, North Chicago, IL 60064, USA
| | - David A DeGoey
- AbbVie, Research and Development, 1 North Waukegan Road, North Chicago, IL 60064, USA
| | - Michael E Kort
- AbbVie, Research and Development, 1 North Waukegan Road, North Chicago, IL 60064, USA
| |
Collapse
|
2
|
Nakashima K, Nakao K, Matsui H. Discovery of Novel HCN4 Blockers with Unique Blocking Kinetics and Binding Properties. SLAS DISCOVERY 2021; 26:896-908. [PMID: 34041946 PMCID: PMC8293762 DOI: 10.1177/24725552211013824] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The hyperpolarization-activated cyclic nucleotide-gated 4 (HCN4) channel underlies the pacemaker currents, called “If,” in sinoatrial nodes (SANs), which regulate heart rhythm. Some HCN4 blockers such as ivabradine have been extensively studied for treating various heart diseases. Studies have shown that these blockers have diverse state dependencies and binding sites, suggesting the existence of potential chemical and functional diversity among HCN4 blockers. Here we report approaches for the identification of novel HCN4 blockers through a random screening campaign among 16,000 small-molecule compounds using an automated patch-clamp system. These molecules exhibited various blockade profiles, and their blocking kinetics and associating amino acids were determined by electrophysiological studies and site-directed mutagenesis analysis, respectively. The profiles of these blockers were distinct from those of the previously reported HCN channel blockers ivabradine and ZD7288. Notably, the mutagenesis analysis showed that blockers with potencies that were increased when the channel was open involved a C478 residue, located at the pore cavity region near the cellular surface of the plasma membrane, while those with potencies that were decreased when the channel was open involved residues Y506 and I510, located at the intracellular region of the pore gate. Thus, this study reported for the first time the discovery of novel HCN4 blockers by screening, and their profiling analysis using an automated patch-clamp system provided chemical tools that will be useful to obtain unique molecular insights into the drug-binding modes of HCN4 and may contribute to the expansion of therapeutic options in the future.
Collapse
Affiliation(s)
- Kosuke Nakashima
- Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| | - Kenji Nakao
- Biomolecular Research Laboratories, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan.,Seedsupply Inc., Fujisawa, Kanagawa, Japan
| | - Hideki Matsui
- Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| |
Collapse
|
3
|
Li Z, Jin X, Wu T, Huang G, Wu K, Lei J, Pan X, Yan N. Structural Basis for Pore Blockade of the Human Cardiac Sodium Channel Na v 1.5 by the Antiarrhythmic Drug Quinidine*. Angew Chem Int Ed Engl 2021; 60:11474-11480. [PMID: 33684260 DOI: 10.1002/anie.202102196] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Indexed: 12/19/2022]
Abstract
Nav 1.5, the primary voltage-gated Na+ (Nav ) channel in heart, is a major target for class I antiarrhythmic agents. Here we present the cryo-EM structure of full-length human Nav 1.5 bound to quinidine, a class Ia antiarrhythmic drug, at 3.3 Å resolution. Quinidine is positioned right beneath the selectivity filter in the pore domain and coordinated by residues from repeats I, III, and IV. Pore blockade by quinidine is achieved through both direct obstruction of the ion permeation path and induced rotation of an invariant Tyr residue that tightens the intracellular gate. Structural comparison with a truncated rat Nav 1.5 in the presence of flecainide, a class Ic agent, reveals distinct binding poses for the two antiarrhythmics within the pore domain. Our work reported here, along with previous studies, reveals the molecular basis for the mechanism of action of class I antiarrhythmic drugs.
Collapse
Affiliation(s)
- Zhangqiang Li
- State Key Laboratory of Membrane Biology, Beijing Advanced Innovation Center for Structural Biology, Tsinghua-Peking Joint Center for Life Science, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Xueqin Jin
- State Key Laboratory of Membrane Biology, Beijing Advanced Innovation Center for Structural Biology, Tsinghua-Peking Joint Center for Life Science, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Tong Wu
- State Key Laboratory of Membrane Biology, Beijing Advanced Innovation Center for Structural Biology, Tsinghua-Peking Joint Center for Life Science, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Gaoxingyu Huang
- Key Laboratory of Structural Biology of Zhejiang Province, Institute of Biology, Westlake Institute for Advanced Study, School of Life Sciences, Westlake University, Hangzhou, 310024, Zhejiang Province, China
| | - Kun Wu
- Medical Research Center, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Jianlin Lei
- Technology Center for Protein Sciences, Ministry of Education Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Xiaojing Pan
- State Key Laboratory of Membrane Biology, Beijing Advanced Innovation Center for Structural Biology, Tsinghua-Peking Joint Center for Life Science, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Nieng Yan
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA
| |
Collapse
|
4
|
Li Z, Jin X, Wu T, Huang G, Wu K, Lei J, Pan X, Yan N. Structural Basis for Pore Blockade of the Human Cardiac Sodium Channel Na
v
1.5 by the Antiarrhythmic Drug Quinidine**. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202102196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Zhangqiang Li
- State Key Laboratory of Membrane Biology Beijing Advanced Innovation Center for Structural Biology Tsinghua-Peking Joint Center for Life Science School of Life Sciences Tsinghua University Beijing 100084 China
| | - Xueqin Jin
- State Key Laboratory of Membrane Biology Beijing Advanced Innovation Center for Structural Biology Tsinghua-Peking Joint Center for Life Science School of Life Sciences Tsinghua University Beijing 100084 China
| | - Tong Wu
- State Key Laboratory of Membrane Biology Beijing Advanced Innovation Center for Structural Biology Tsinghua-Peking Joint Center for Life Science School of Life Sciences Tsinghua University Beijing 100084 China
| | - Gaoxingyu Huang
- Key Laboratory of Structural Biology of Zhejiang Province Institute of Biology, Westlake Institute for Advanced Study School of Life Sciences Westlake University Hangzhou 310024 Zhejiang Province China
| | - Kun Wu
- Medical Research Center Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation Beijing Chao-Yang Hospital Capital Medical University Beijing 100020 China
| | - Jianlin Lei
- Technology Center for Protein Sciences Ministry of Education Key Laboratory of Protein Sciences School of Life Sciences Tsinghua University Beijing 100084 China
| | - Xiaojing Pan
- State Key Laboratory of Membrane Biology Beijing Advanced Innovation Center for Structural Biology Tsinghua-Peking Joint Center for Life Science School of Life Sciences Tsinghua University Beijing 100084 China
| | - Nieng Yan
- Department of Molecular Biology Princeton University Princeton NJ 08544 USA
| |
Collapse
|
5
|
Yang PC, DeMarco KR, Aghasafari P, Jeng MT, Dawson JRD, Bekker S, Noskov SY, Yarov-Yarovoy V, Vorobyov I, Clancy CE. A Computational Pipeline to Predict Cardiotoxicity: From the Atom to the Rhythm. Circ Res 2020; 126:947-964. [PMID: 32091972 DOI: 10.1161/circresaha.119.316404] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
RATIONALE Drug-induced proarrhythmia is so tightly associated with prolongation of the QT interval that QT prolongation is an accepted surrogate marker for arrhythmia. But QT interval is too sensitive a marker and not selective, resulting in many useful drugs eliminated in drug discovery. OBJECTIVE To predict the impact of a drug from the drug chemistry on the cardiac rhythm. METHODS AND RESULTS In a new linkage, we connected atomistic scale information to protein, cell, and tissue scales by predicting drug-binding affinities and rates from simulation of ion channel and drug structure interactions and then used these values to model drug effects on the hERG channel. Model components were integrated into predictive models at the cell and tissue scales to expose fundamental arrhythmia vulnerability mechanisms and complex interactions underlying emergent behaviors. Human clinical data were used for model framework validation and showed excellent agreement, demonstrating feasibility of a new approach for cardiotoxicity prediction. CONCLUSIONS We present a multiscale model framework to predict electrotoxicity in the heart from the atom to the rhythm. Novel mechanistic insights emerged at all scales of the system, from the specific nature of proarrhythmic drug interaction with the hERG channel, to the fundamental cellular and tissue-level arrhythmia mechanisms. Applications of machine learning indicate necessary and sufficient parameters that predict arrhythmia vulnerability. We expect that the model framework may be expanded to make an impact in drug discovery, drug safety screening for a variety of compounds and targets, and in a variety of regulatory processes.
Collapse
Affiliation(s)
- Pei-Chi Yang
- From the Department of Physiology and Membrane Biology (P.-C.Y., K.R.D., P.A., M.-T.J., J.R.D.D., V.Y.-Y., I.V., C.E.C.), University of California Davis
| | - Kevin R DeMarco
- From the Department of Physiology and Membrane Biology (P.-C.Y., K.R.D., P.A., M.-T.J., J.R.D.D., V.Y.-Y., I.V., C.E.C.), University of California Davis
| | - Parya Aghasafari
- From the Department of Physiology and Membrane Biology (P.-C.Y., K.R.D., P.A., M.-T.J., J.R.D.D., V.Y.-Y., I.V., C.E.C.), University of California Davis
| | - Mao-Tsuen Jeng
- From the Department of Physiology and Membrane Biology (P.-C.Y., K.R.D., P.A., M.-T.J., J.R.D.D., V.Y.-Y., I.V., C.E.C.), University of California Davis
| | - John R D Dawson
- From the Department of Physiology and Membrane Biology (P.-C.Y., K.R.D., P.A., M.-T.J., J.R.D.D., V.Y.-Y., I.V., C.E.C.), University of California Davis.,Biophysics Graduate Group (J.R.D.D.), University of California Davis
| | - Slava Bekker
- Department of Science and Engineering, American River College, Sacramento, CA (S.B.)
| | - Sergei Y Noskov
- Faculty of Science, Centre for Molecular Simulations and Department of Biological Sciences, University of Calgary, Alberta, Canada (S.Y.N.)
| | - Vladimir Yarov-Yarovoy
- From the Department of Physiology and Membrane Biology (P.-C.Y., K.R.D., P.A., M.-T.J., J.R.D.D., V.Y.-Y., I.V., C.E.C.), University of California Davis
| | - Igor Vorobyov
- From the Department of Physiology and Membrane Biology (P.-C.Y., K.R.D., P.A., M.-T.J., J.R.D.D., V.Y.-Y., I.V., C.E.C.), University of California Davis.,Department of Pharmacology (I.V., C.E.C.), University of California Davis
| | - Colleen E Clancy
- From the Department of Physiology and Membrane Biology (P.-C.Y., K.R.D., P.A., M.-T.J., J.R.D.D., V.Y.-Y., I.V., C.E.C.), University of California Davis.,Department of Pharmacology (I.V., C.E.C.), University of California Davis
| |
Collapse
|
6
|
Guo D, Jenkinson S. Simultaneous assessment of compound activity on cardiac Nav1.5 peak and late currents in an automated patch clamp platform. J Pharmacol Toxicol Methods 2019; 99:106575. [PMID: 30999054 DOI: 10.1016/j.vascn.2019.04.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 03/22/2019] [Accepted: 04/12/2019] [Indexed: 01/04/2023]
Abstract
INTRODUCTION High throughput in vitro profiling of the cardiac Nav1.5 peak sodium current (INa) is widely used in cardiac safety screening. However, there is no standardized high throughput method to measure late INa. This study assessed the pharmacological and biophysical properties of veratridine and ATX-II, as well as the channel mutation (Nav1.5-∆KPQ) on the late INa. We describe a method for simultaneous measurement of both peak and late INa. METHODS The planar patch clamp technique (QPatch) was applied to record the peak and late INa. RESULTS The Nav1.5-∆KPQ mutant produced a small late INa (41.9 ± 5.4 pA) not large enough to enable compound profiling. In contrast in wild type Nav1.5 expressing cells veratridine (100 μM) and ATX-II (100 nM) enhanced concentration-dependent increases in the late INa (maximum responses of 1162.2 ± 258.5 pA and 392.4 ± 71.3 pA, respectively). Veratridine inhibited, whereas, ATX-II had a minimal effect, on the peak INa and preserved the current-voltage curve. Peak and late INa inhibition was characterized for 25 clinical INa blockers in the presence of ATX-II. Compound IC50 values for peak INa generated in the absence or presence of ATX-II correlated. The potency of the late INa block was found to be dependent on whether it was measured at the end of the depolarizing pulse or during the ramp. DISCUSSION In the presence of ATX-II, both peak and late INa could be assessed simultaneously. Late INa may be best assessed using the maximum response obtained during the ramp of the voltage protocol.
Collapse
Affiliation(s)
- Donglin Guo
- Drug Safety Research and Development, Pfizer Inc., La Jolla, CA 92121, United States of America.
| | - Stephen Jenkinson
- Drug Safety Research and Development, Pfizer Inc., La Jolla, CA 92121, United States of America
| |
Collapse
|
7
|
Whittaker DG, Hancox JC, Zhang H. In silico Assessment of Pharmacotherapy for Human Atrial Patho-Electrophysiology Associated With hERG-Linked Short QT Syndrome. Front Physiol 2019; 9:1888. [PMID: 30687112 PMCID: PMC6336736 DOI: 10.3389/fphys.2018.01888] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 12/12/2018] [Indexed: 12/19/2022] Open
Abstract
Short QT syndrome variant 1 (SQT1) arises due to gain-of-function mutations to the human Ether-à-go-go-Related Gene (hERG), which encodes the α subunit of channels carrying rapid delayed rectifier potassium current, IKr. In addition to QT interval shortening and ventricular arrhythmias, SQT1 is associated with increased risk of atrial fibrillation (AF), which is often the only clinical presentation. However, the underlying basis of AF and its pharmacological treatment remain incompletely understood in the context of SQT1. In this study, computational modeling was used to investigate mechanisms of human atrial arrhythmogenesis consequent to a SQT1 mutation, as well as pharmacotherapeutic effects of selected class I drugs–disopyramide, quinidine, and propafenone. A Markov chain formulation describing wild type (WT) and N588K-hERG mutant IKr was incorporated into a contemporary human atrial action potential (AP) model, which was integrated into one-dimensional (1D) tissue strands, idealized 2D sheets, and a 3D heterogeneous, anatomical human atria model. Multi-channel pharmacological effects of disopyramide, quinidine, and propafenone, including binding kinetics for IKr/hERG and sodium current, INa, were considered. Heterozygous and homozygous formulations of the N588K-hERG mutation shortened the AP duration (APD) by 53 and 86 ms, respectively, which abbreviated the effective refractory period (ERP) and excitation wavelength in tissue, increasing the lifespan and dominant frequency (DF) of scroll waves in the 3D anatomical human atria. At the concentrations tested in this study, quinidine most effectively prolonged the APD and ERP in the setting of SQT1, followed by disopyramide and propafenone. In 2D simulations, disopyramide and quinidine promoted re-entry termination by increasing the re-entry wavelength, whereas propafenone induced secondary waves which destabilized the re-entrant circuit. In 3D simulations, the DF of re-entry was reduced in a dose-dependent manner for disopyramide and quinidine, and propafenone to a lesser extent. All of the anti-arrhythmic agents promoted pharmacological conversion, most frequently terminating re-entry in the order quinidine > propafenone = disopyramide. Our findings provide further insight into mechanisms of SQT1-related AF and a rational basis for the pursuit of combined IKr and INa block based pharmacological strategies in the treatment of SQT1-linked AF.
Collapse
Affiliation(s)
- Dominic G Whittaker
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom.,Biological Physics Group, School of Physics and Astronomy, The University of Manchester, Manchester, United Kingdom
| | - Jules C Hancox
- Biological Physics Group, School of Physics and Astronomy, The University of Manchester, Manchester, United Kingdom.,Cardiovascular Research Laboratories, Department of Physiology, Pharmacology and Neuroscience, School of Medical Sciences, University of Bristol, Bristol, United Kingdom
| | - Henggui Zhang
- Biological Physics Group, School of Physics and Astronomy, The University of Manchester, Manchester, United Kingdom.,School of Computer Science and Technology, Harbin Institute of Technology, Harbin, China.,Space Institute of Southern China, Shenzhen, China.,Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| |
Collapse
|
8
|
Liin SI, Lund PE, Larsson JE, Brask J, Wallner B, Elinder F. Biaryl sulfonamide motifs up- or down-regulate ion channel activity by activating voltage sensors. J Gen Physiol 2018; 150:1215-1230. [PMID: 30002162 PMCID: PMC6080886 DOI: 10.1085/jgp.201711942] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 04/20/2018] [Accepted: 06/11/2018] [Indexed: 11/20/2022] Open
Abstract
Voltage-gated ion channels are key molecules for the generation of cellular electrical excitability. Many pharmaceutical drugs target these channels by blocking their ion-conducting pore, but in many cases, channel-opening compounds would be more beneficial. Here, to search for new channel-opening compounds, we screen 18,000 compounds with high-throughput patch-clamp technology and find several potassium-channel openers that share a distinct biaryl-sulfonamide motif. Our data suggest that the negatively charged variants of these compounds bind to the top of the voltage-sensor domain, between transmembrane segments 3 and 4, to open the channel. Although we show here that biaryl-sulfonamide compounds open a potassium channel, they have also been reported to block sodium and calcium channels. However, because they inactivate voltage-gated sodium channels by promoting activation of one voltage sensor, we suggest that, despite different effects on the channel gates, the biaryl-sulfonamide motif is a general ion-channel activator motif. Because these compounds block action potential-generating sodium and calcium channels and open an action potential-dampening potassium channel, they should have a high propensity to reduce excitability. This opens up the possibility to build new excitability-reducing pharmaceutical drugs from the biaryl-sulfonamide scaffold.
Collapse
Affiliation(s)
- Sara I Liin
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Per-Eric Lund
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Johan E Larsson
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Johan Brask
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Björn Wallner
- Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| | - Fredrik Elinder
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| |
Collapse
|
9
|
A tutorial on model informed approaches to cardiovascular safety with focus on cardiac repolarisation. J Pharmacokinet Pharmacodyn 2018; 45:365-381. [PMID: 29736890 DOI: 10.1007/s10928-018-9589-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 04/16/2018] [Indexed: 12/19/2022]
Abstract
Drugs can affect the cardiovascular (CV) system either as an intended treatment or as an unwanted side effect. In both cases, drug-induced cardiotoxicities such as arrhythmia and unfavourable hemodynamic effects can occur, and be described using mathematical models; such a model informed approach can provide valuable information during drug development and can aid decision-making. However, in order to develop informative models, it is vital to understand CV physiology. The aims of this tutorial are to present (1) key background biological and medical aspects of the CV system, (2) CV electrophysiology, (3) CV safety concepts, (4) practical aspects of development of CV models and (5) regulatory expectations with a focus on using model informed and quantitative approaches to support nonclinical and clinical drug development. In addition, we share several case studies to provide practical information on project strategy (planning, key questions, assumptions setting, and experimental design) and mathematical models development that support decision-making during drug discovery and development.
Collapse
|
10
|
Streit J, Kleinlogel S. Dynamic all-optical drug screening on cardiac voltage-gated ion channels. Sci Rep 2018; 8:1153. [PMID: 29348631 PMCID: PMC5773578 DOI: 10.1038/s41598-018-19412-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 12/27/2017] [Indexed: 11/09/2022] Open
Abstract
Voltage-gated ion channels (VGCs) are prime targets for the pharmaceutical industry, but drug profiling on VGCs is challenging, since drug interactions are confined to specific conformational channel states mediated by changes in transmembrane potential. Here we combined various optogenetic tools to develop dynamic, high-throughput drug profiling assays with defined light-step protocols to interrogate VGC states on a millisecond timescale. We show that such light-induced electrophysiology (LiEp) yields high-quality pharmacological data with exceptional screening windows for drugs acting on the major cardiac VGCs, including hNav1.5, hKv1.5 and hERG. LiEp-based screening remained robust when using a variety of optogenetic actuators (ChR2, ChR2(H134R), CatCh, ChR2-EYFP-βArchT) and different types of organic (RH421, Di-4-ANBDQPQ, BeRST1) or genetic voltage sensors (QuasAr1). The tractability of LiEp allows a versatile and precise alternative to state-of-the-art VGC drug screening platforms such as automated electrophysiology or FLIPR readers.
Collapse
Affiliation(s)
- Jonas Streit
- Institute of Physiology, University of Bern, Bühlplatz 5, 3012, Bern, Switzerland
| | - Sonja Kleinlogel
- Institute of Physiology, University of Bern, Bühlplatz 5, 3012, Bern, Switzerland.
| |
Collapse
|
11
|
Bergenholm L, Parkinson J, Mettetal J, Evans ND, Chappell MJ, Collins T. Predicting QRS and PR interval prolongations in humans using nonclinical data. Br J Pharmacol 2017; 174:3268-3283. [PMID: 28675424 DOI: 10.1111/bph.13940] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 06/04/2017] [Accepted: 06/09/2017] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND AND PURPOSE Risk of cardiac conduction slowing (QRS/PR prolongations) is assessed prior to clinical trials using in vitro and in vivo studies. Understanding the quantitative translation of these studies to the clinical situation enables improved risk assessment in the nonclinical phase. EXPERIMENTAL APPROACH Four compounds that prolong QRS and/or PR (AZD1305, flecainide, quinidine and verapamil) were characterized using in vitro (sodium/calcium channels), in vivo (guinea pigs/dogs) and clinical data. Concentration-matched translational relationships were developed based on in vitro and in vivo modelling, and the in vitro to clinical translation of AZD1305 was quantified using an in vitro model. KEY RESULTS Meaningful (10%) human QRS/PR effects correlated with low levels of in vitro Nav 1.5 block (3-7%) and Cav 1.2 binding (13-21%) for all compounds. The in vitro model developed using AZD1305 successfully predicted QRS/PR effects for the remaining drugs. Meaningful QRS/PR changes in humans correlated with small effects in guinea pigs and dogs (QRS 2.3-4.6% and PR 2.3-10%), suggesting that worst-case human effects can be predicted by assuming four times greater effects at the same concentration from dog/guinea pig data. CONCLUSION AND IMPLICATIONS Small changes in vitro and in vivo consistently translated to meaningful PR/QRS changes in humans across compounds. Assuming broad applicability of these approaches to assess cardiovascular safety risk for non-arrhythmic drugs, this study provides a means of predicting human QRS/PR effects of new drugs from effects observed in nonclinical studies.
Collapse
Affiliation(s)
- L Bergenholm
- Biomedical and Biological Systems Laboratory, School of Engineering, University of Warwick, Coventry, UK.,Drug Metabolism and Pharmacokinetics, Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development, AstraZeneca, Gothenburg, Sweden
| | - J Parkinson
- Early Clinical Development, Quantitative Clinical Pharmacology, Innovative Medicines and Early Development, AstraZeneca, Gothenburg, Sweden
| | - J Mettetal
- Safety and ADME Translational Sciences, Drug Safety and Metabolism, Innovative Medicines and Early Development, AstraZeneca, Cambridge, UK
| | - N D Evans
- Biomedical and Biological Systems Laboratory, School of Engineering, University of Warwick, Coventry, UK
| | - M J Chappell
- Biomedical and Biological Systems Laboratory, School of Engineering, University of Warwick, Coventry, UK
| | - T Collins
- Safety and ADME Translational Sciences, Drug Safety and Metabolism, Innovative Medicines and Early Development, AstraZeneca, Cambridge, UK
| |
Collapse
|
12
|
Sharifi M. Computational approaches to understand the adverse drug effect on potassium, sodium and calcium channels for predicting TdP cardiac arrhythmias. J Mol Graph Model 2017; 76:152-160. [PMID: 28756335 DOI: 10.1016/j.jmgm.2017.06.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Revised: 06/08/2017] [Accepted: 06/10/2017] [Indexed: 02/08/2023]
Abstract
Ion channels play a crucial role in the cardiovascular system. Our understanding of cardiac ion channel function has improved since their first discoveries. The flow of potassium, sodium and calcium ions across cardiomyocytes is vital for regular cardiac rhythm. Blockage of these channels, delays cardiac repolarization or tend to shorten repolarization and may induce arrhythmia. Detection of drug risk by channel blockade is considered essential for drug regulators. Advanced computational models can be used as an early screen for torsadogenic potential in drug candidates. New drug candidates that are determined to not cause blockage are more likely to pass successfully through preclinical trials and not be withdrawn later from the marketplace by manufacturer. Several different approved drugs, however, can cause a distinctive polymorphic ventricular arrhythmia known as torsade de pointes (TdP), which may lead to sudden death. The objective of the present study is to review the mechanisms and computational models used to assess the risk that a drug may TdP. KEY POINTS There is strong evidence from multiple studies that blockage of the L-type calcium current reduces risk of TdP. Blockage of sodium channels slows cardiac action potential conduction, however, not all sodium channel blocking antiarrhythmic drugs produce a significant effect, while late sodium channel block reduces TdP. Interestingly, there are some drugs that block the hERG potassium channel and therefore cause QT prolongation, but they are not associated with TdP. Recent studies confirmed the necessity of studying multiple distinctionic ion channels which are responsible for cardiac related diseases or TdP, to obtain an improved clinical TdP risk prediction of compound interactions and also for designing drugs.
Collapse
Affiliation(s)
- Mohsen Sharifi
- Division of Systems Biology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR 72079, USA.
| |
Collapse
|
13
|
Frost JM, DeGoey DA, Shi L, Gum RJ, Fricano MM, Lundgaard GL, El-Kouhen OF, Hsieh GC, Neelands T, Matulenko MA, Daanen JF, Pai M, Ghoreishi-Haack N, Zhan C, Zhang XF, Kort ME. Substituted Indazoles as Nav1.7 Blockers for the Treatment of Pain. J Med Chem 2016; 59:3373-91. [DOI: 10.1021/acs.jmedchem.6b00063] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Affiliation(s)
- Jennifer M. Frost
- Research
and Development, AbbVie, 1 North Waukegan Road, North
Chicago, Illinois 60064, United States
| | - David A. DeGoey
- Research
and Development, AbbVie, 1 North Waukegan Road, North
Chicago, Illinois 60064, United States
| | - Lei Shi
- Research
and Development, AbbVie, 1 North Waukegan Road, North
Chicago, Illinois 60064, United States
| | - Rebecca J. Gum
- Research
and Development, AbbVie, 1 North Waukegan Road, North
Chicago, Illinois 60064, United States
| | - Meagan M. Fricano
- Research
and Development, AbbVie, 1 North Waukegan Road, North
Chicago, Illinois 60064, United States
| | - Greta L. Lundgaard
- Research
and Development, AbbVie, 1 North Waukegan Road, North
Chicago, Illinois 60064, United States
| | - Odile F. El-Kouhen
- Research
and Development, AbbVie, 1 North Waukegan Road, North
Chicago, Illinois 60064, United States
| | - Gin C. Hsieh
- Research
and Development, AbbVie, 1 North Waukegan Road, North
Chicago, Illinois 60064, United States
| | - Torben Neelands
- Research
and Development, AbbVie, 1 North Waukegan Road, North
Chicago, Illinois 60064, United States
| | - Mark A. Matulenko
- Research
and Development, AbbVie, 1 North Waukegan Road, North
Chicago, Illinois 60064, United States
| | - Jerome F. Daanen
- Research
and Development, AbbVie, 1 North Waukegan Road, North
Chicago, Illinois 60064, United States
| | - Madhavi Pai
- Research
and Development, AbbVie, 1 North Waukegan Road, North
Chicago, Illinois 60064, United States
| | | | - Cenchen Zhan
- Research
and Development, AbbVie, 1 North Waukegan Road, North
Chicago, Illinois 60064, United States
| | - Xu-Feng Zhang
- Research
and Development, AbbVie, 1 North Waukegan Road, North
Chicago, Illinois 60064, United States
| | - Michael E. Kort
- Research
and Development, AbbVie, 1 North Waukegan Road, North
Chicago, Illinois 60064, United States
| |
Collapse
|
14
|
Moreno JD, Lewis TJ, Clancy CE. Parameterization for In-Silico Modeling of Ion Channel Interactions with Drugs. PLoS One 2016; 11:e0150761. [PMID: 26963710 PMCID: PMC4786197 DOI: 10.1371/journal.pone.0150761] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 02/18/2016] [Indexed: 12/31/2022] Open
Abstract
Since the first Hodgkin and Huxley ion channel model was described in the 1950s, there has been an explosion in mathematical models to describe ion channel function. As experimental data has become richer, models have concomitantly been improved to better represent ion channel kinetic processes, although these improvements have generally resulted in more model complexity and an increase in the number of parameters necessary to populate the models. Models have also been developed to explicitly model drug interactions with ion channels. Recent models of drug-channel interactions account for the discrete kinetics of drug interaction with distinct ion channel state conformations, as it has become clear that such interactions underlie complex emergent kinetics such as use-dependent block. Here, we describe an approach for developing a model for ion channel drug interactions. The method describes the process of extracting rate constants from experimental electrophysiological function data to use as initial conditions for the model parameters. We then describe implementation of a parameter optimization method to refine the model rate constants describing ion channel drug kinetics. The algorithm takes advantage of readily available parallel computing tools to speed up the optimization. Finally, we describe some potential applications of the platform including the potential for gaining fundamental mechanistic insights into ion channel function and applications to in silico drug screening and development.
Collapse
Affiliation(s)
- Jonathan D. Moreno
- Division of Cardiology, Department of Medicine, Barnes-Jewish Hospital, Washington University in St. Louis, St. Louis, MO, United States of America
- * E-mail:
| | - Timothy J. Lewis
- Department of Mathematics, University of California Davis, Davis, CA, United States of America
| | - Colleen E. Clancy
- Department of Pharmacology, University of California Davis, Davis, CA, United States of America
| |
Collapse
|
15
|
Bergenholm L, Collins T, Evans ND, Chappell MJ, Parkinson J. PKPD modelling of PR and QRS intervals in conscious dogs using standard safety pharmacology data. J Pharmacol Toxicol Methods 2016; 79:34-44. [PMID: 26780675 DOI: 10.1016/j.vascn.2016.01.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 12/23/2015] [Accepted: 01/07/2016] [Indexed: 11/30/2022]
Abstract
INTRODUCTION Pharmacokinetic-pharmacodynamic (PKPD) modelling can improve safety assessment, but few PKPD models describing drug-induced QRS and PR prolongations have been published. This investigation aims to develop and evaluate PKPD models for describing QRS and PR effects in routine safety studies. METHODS Exposure and telemetry data from safety pharmacology studies in conscious beagle dogs were acquired. Mixed effects baseline and PK-QRS/PR models were developed for the anti-arrhythmic compounds AZD1305, flecainide, quinidine and verapamil and the anti-muscarinic compounds AZD8683 and AZD9164. RR interval correction and circadian rhythms were investigated for predicting baseline variability. Individual PK predictions were used to drive the pharmacological effects evaluating linear and non-linear direct and effect compartment models. RESULTS Conduction slowing induced by the tested anti-arrhythmics was direct and proportional at low exposures, whilst time delays and non-linear effects were evident for the tested anti-muscarinics. AZD1305, flecainide and quinidine induced QRS widening with 4.2, 10 and 5.6% μM(-1) unbound drug. AZD1305 and flecainide also prolonged PR with 13.5 and 11.5% μM(-1). PR prolongations induced by the anti-muscarinics and verapamil were best described by Emax models with maximal effects ranging from 55 to 95%. RR interval correction and circadian rhythm improved PR but not QRS modelling. However, circadian rhythm had minor impact on estimated drug effects. DISCUSSION Baseline and drug-induced effects on QRS and PR intervals can be effectively described with PKPD models using routine data, providing quantitative safety information to support drug discovery and development.
Collapse
Affiliation(s)
- Linnéa Bergenholm
- Biomedical & Biological Systems Laboratory, School of Engineering, University of Warwick, Coventry, UK
| | - Teresa Collins
- Translational Safety, Drug Safety and Metabolism, iMED, AstraZeneca, Cambridge, UK
| | - Neil D Evans
- Biomedical & Biological Systems Laboratory, School of Engineering, University of Warwick, Coventry, UK
| | - Michael J Chappell
- Biomedical & Biological Systems Laboratory, School of Engineering, University of Warwick, Coventry, UK
| | - Joanna Parkinson
- Early Clinical Development, Quantitative Clinical Pharmacology, iMED, AstraZeneca, Mölndal, Sweden
| |
Collapse
|
16
|
Ottosson NE, Wu X, Nolting A, Karlsson U, Lund PE, Ruda K, Svensson S, Konradsson P, Elinder F. Resin-acid derivatives as potent electrostatic openers of voltage-gated K channels and suppressors of neuronal excitability. Sci Rep 2015; 5:13278. [PMID: 26299574 PMCID: PMC4547393 DOI: 10.1038/srep13278] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 07/13/2015] [Indexed: 11/12/2022] Open
Abstract
Voltage-gated ion channels generate cellular excitability, cause diseases when mutated, and act as drug targets in hyperexcitability diseases, such as epilepsy, cardiac arrhythmia and pain. Unfortunately, many patients do not satisfactorily respond to the present-day drugs. We found that the naturally occurring resin acid dehydroabietic acid (DHAA) is a potent opener of a voltage-gated K channel and thereby a potential suppressor of cellular excitability. DHAA acts via a non-traditional mechanism, by electrostatically activating the voltage-sensor domain, rather than directly targeting the ion-conducting pore domain. By systematic iterative modifications of DHAA we synthesized 71 derivatives and found 32 compounds more potent than DHAA. The most potent compound, Compound 77, is 240 times more efficient than DHAA in opening a K channel. This and other potent compounds reduced excitability in dorsal root ganglion neurons, suggesting that resin-acid derivatives can become the first members of a new family of drugs with the potential for treatment of hyperexcitability diseases.
Collapse
Affiliation(s)
- Nina E Ottosson
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Xiongyu Wu
- Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| | - Andreas Nolting
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Urban Karlsson
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Per-Eric Lund
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Katinka Ruda
- Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| | - Stefan Svensson
- Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| | - Peter Konradsson
- Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| | - Fredrik Elinder
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| |
Collapse
|
17
|
Gintant G. In Vitro Early Safety Pharmacology Screening: Perspectives Related to Cardiovascular Safety. Handb Exp Pharmacol 2015; 229:47-64. [PMID: 26091635 DOI: 10.1007/978-3-662-46943-9_2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
In vitro screening for cardiovascular safety liabilities of novel drug candidates presents a challenge for the pharmaceutical industry. Such approaches rely on detecting pharmacologic effects on key components of complex integrated system early in drug discovery to define potential safety liabilities. Key to such studies are the concepts of hazard identification vs. risk assessment, drug specificity vs. selectivity, and an appreciation of the challenges faced when attempting to translate in vitro findings to preclinical in vivo as well as clinical effects. This chapter defines some key aspects of early safety pharmacology screening for cardiovascular liabilities, citing studies of two key depolarizing cardiac currents (fast sodium current and L-type calcium current) as examples linked to effects on cardiac conduction and repolarization.
Collapse
|
18
|
Mannikko R, Bridgland-Taylor MH, Pye H, Swallow S, Abi-Gerges N, Morton MJ, Pollard CE. Pharmacological and electrophysiological characterization of AZSMO-23, an activator of the hERG K(+) channel. Br J Pharmacol 2015; 172:3112-25. [PMID: 25684549 DOI: 10.1111/bph.13115] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Revised: 01/31/2015] [Accepted: 02/09/2015] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND AND PURPOSE We aimed to characterize the pharmacology and electrophysiology of N-[3-(1H-benzimidazol-2-yl)-4-chloro-phenyl]pyridine-3-carboxamide (AZSMO-23), an activator of the human ether-a-go-go-related gene (hERG)-encoded K(+) channel (Kv 11.1). EXPERIMENTAL APPROACH Automated electrophysiology was used to study the pharmacology of AZSMO-23 on wild-type (WT), Y652A, F656T or G628C/S631C hERG, and on other cardiac ion channels. Its mechanism of action was characterized with conventional electrophysiology. KEY RESULTS AZSMO-23 activated WT hERG pre-pulse and tail current with EC50 values of 28.6 and 11.2 μM respectively. At 100 μM, pre-pulse current at +40 mV was increased by 952 ± 41% and tail current at -30 mV by 238 ± 13% compared with vehicle values. The primary mechanism for this effect was a 74.5 mV depolarizing shift in the voltage dependence of inactivation, without any shift in the voltage dependence of activation. Structure-activity relationships for this effect were remarkably subtle, with close analogues of AZSMO-23 acting as hERG inhibitors. AZSMO-23 blocked the mutant channel, hERG Y652A, but against another mutant channel, hERG F656T, its activator activity was enhanced. It inhibited activity of the G628C/S631C non-inactivating hERG mutant channel. AZSMO-23 was not hERG selective, as it blocked hKv 4.3-hKChIP2.2, hCav 3.2 and hKv 1.5 and activated hCav 1.2/β2/α2δ channels. CONCLUSION AND IMPLICATIONS The activity of AZSMO-23 and those of its close analogues suggest these compounds may be of value to elucidate the mechanism of type 2 hERG activators to better understand the pharmacology of this area from both a safety perspective and in relation to treatment of congenital long QT syndrome.
Collapse
Affiliation(s)
- R Mannikko
- Institute of Neurology, Faculty of Brain Sciences, University College London, London, Middlesex, UK
| | | | - H Pye
- AstraZeneca, Macclesfield, Cheshire, UK
| | - S Swallow
- AstraZeneca, Macclesfield, Cheshire, UK
| | | | | | | |
Collapse
|
19
|
Burnham MP, Sharpe PM, Garner C, Hughes R, Pollard CE, Bowes J. Investigation of connexin 43 uncoupling and prolongation of the cardiac QRS complex in preclinical and marketed drugs. Br J Pharmacol 2014; 171:4808-19. [PMID: 24328991 DOI: 10.1111/bph.12554] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Revised: 11/13/2013] [Accepted: 11/22/2013] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND AND PURPOSE Prolongation of the cardiac QRS complex is linked to increased mortality and may result from drug-induced inhibition of cardiac sodium channels (hNaV 1.5). There has been no systematic evaluation of preclinical and marketed drugs for their additional potential to cause QRS prolongation via gap junction uncoupling. EXPERIMENTAL APPROACH Using the human cardiac gap junction connexin 43 (hCx43), a dye transfer 'parachute' assay to determine IC50 values for compound ranking was validated with compounds known to uncouple gap junctions. Uncoupling activity (and hNaV 1.5 inhibition by automated patch clamp) was determined in a set of marketed drugs and preclinical candidate drugs, each with information regarding propensity to prolong QRS. KEY RESULTS The potency of known gap junction uncouplers to uncouple hCx43 was ranked (according to IC50 ) as phorbol ester>digoxin>meclofenamic acid>carbenoxolone>heptanol. Among the drugs associated with QRS prolongation, 29% were found to uncouple hCx43 (IC50 < 50 μM), whereas no uncoupling activity was observed in drugs not associated with QRS prolongation. In preclinical candidate drugs, hCx43 and hNaV 1.5 IC50 values were similar (within threefold). No consistent margin over preclinical Cmax (free) was apparent for QRS prolongation associated with Cx43 inhibition. However, instances were found of QRS prolonging compounds that uncoupled hCx43 with significantly less activity at hNaV 1.5. CONCLUSION AND IMPLICATIONS These results demonstrate that off-target uncoupling activity is apparent in drug and drug-like molecules. Although the full ramifications of Cx inhibition remain to be established, screening for hCx43 off-target activity could reduce the likelihood of developing candidate drugs with a risk of causing QRS prolongation.
Collapse
Affiliation(s)
- M P Burnham
- Safety Assessment, AstraZeneca R&D, Macclesfield, UK; Discovery Sciences, AstraZeneca R&D, Macclesfield, UK
| | | | | | | | | | | |
Collapse
|
20
|
Yarov-Yarovoy V, Allen TW, Clancy CE. Computational Models for Predictive Cardiac Ion Channel Pharmacology. ACTA ACUST UNITED AC 2014; 14:3-10. [PMID: 26635886 DOI: 10.1016/j.ddmod.2014.04.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
A wealth of experimental data exists describing the elementary building blocks of complex physiological systems. However, it is increasingly apparent in the biomedical sciences that mechanisms of biological function cannot be observed or readily predicted via study of constituent elements alone. This is especially clear in the longstanding failures in prediction of effects of drug treatment for heart rhythm disturbances. These failures stem in part from classical assumptions that have been made in cardiac antiarrhythmic drug development - that a drug operates by one mechanism via one target receptor that arises from one gene.
Collapse
Affiliation(s)
| | - Toby W Allen
- Department of Chemistry, University of California, Davis
| | | |
Collapse
|
21
|
Townsend C, Brown BS. Predicting drug-induced QT prolongation and torsades de pointes: a review of preclinical endpoint measures. ACTA ACUST UNITED AC 2013; Chapter 10:Unit 10.16. [PMID: 23744708 DOI: 10.1002/0471141755.ph1016s61] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Compound-induced prolongation of the cardiac QT interval is a major concern in drug development and this unit discusses approaches that can predict QT effects prior to undertaking clinical trials. The majority of compounds that prolong the QT interval block the cardiac rapid delayed rectifier potassium current, IKr (hERG). Described in this overview are different ways to measure hERG, from recent advances in automated electrophysiology to the quantification of channel protein trafficking and binding. The contribution of other cardiac ion channels to hERG data interpretation is also discussed. In addition, endpoint measures of the integrated activity of cardiac ion channels at the single-cell, tissue, and whole-animal level, including for example the well-established action potential to the more recent beat-to-beat variability, transmural dispersion of repolarization, and field potential duration, are described in the context of their ability to predict QT prolongation and torsadogenicity in humans.
Collapse
Affiliation(s)
- Claire Townsend
- GlaxoSmithKline Biological Reagents and Assay Development, Research Triangle Park, NC, USA
| | | |
Collapse
|
22
|
Roberts RA, Kavanagh SL, Mellor HR, Pollard CE, Robinson S, Platz SJ. Reducing attrition in drug development: smart loading preclinical safety assessment. Drug Discov Today 2013; 19:341-7. [PMID: 24269835 DOI: 10.1016/j.drudis.2013.11.014] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Revised: 10/16/2013] [Accepted: 11/14/2013] [Indexed: 11/25/2022]
Abstract
Entry into the crucial preclinical good laboratory practice (GLP) stage of toxicology testing triggers significant R&D investment yet >20% of AstraZeneca's potential new medicines have been stopped for safety reasons in this GLP phase alone. How could we avoid at least some of these costly failures? An analysis of historical toxicities that caused stopping ('stopping toxicities') showed that >50% were attributable to target organ toxicities emerging within 2 weeks of repeat dosing or to acute cardiovascular risks. By frontloading 2-week repeat-dose toxicity studies and a comprehensive assessment of cardiovascular safety, we anticipate a potential 50% reduction in attrition in the GLP phase. This will reduce animal use overall, save significant R&D costs and improve drug pipeline quality.
Collapse
Affiliation(s)
- Ruth A Roberts
- Drug Safety and Metabolism, AstraZeneca, Alderley Park, Macclesfield, SK10 4TG, UK.
| | - Stefan L Kavanagh
- Drug Safety and Metabolism, AstraZeneca, Alderley Park, Macclesfield, SK10 4TG, UK
| | - Howard R Mellor
- Drug Safety and Metabolism, AstraZeneca, Alderley Park, Macclesfield, SK10 4TG, UK
| | | | - Sally Robinson
- Drug Safety and Metabolism, AstraZeneca, Alderley Park, Macclesfield, SK10 4TG, UK
| | - Stefan J Platz
- Drug Safety and Metabolism, AstraZeneca, Alderley Park, Macclesfield, SK10 4TG, UK
| |
Collapse
|
23
|
Comparative effects of sodium channel blockers in short term rat whole embryo culture. Toxicol Appl Pharmacol 2013; 272:306-12. [DOI: 10.1016/j.taap.2013.06.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Revised: 06/16/2013] [Accepted: 06/21/2013] [Indexed: 02/01/2023]
|
24
|
Elkins RC, Davies MR, Brough SJ, Gavaghan DJ, Cui Y, Abi-Gerges N, Mirams GR. Variability in high-throughput ion-channel screening data and consequences for cardiac safety assessment. J Pharmacol Toxicol Methods 2013; 68:112-22. [PMID: 23651875 PMCID: PMC4135079 DOI: 10.1016/j.vascn.2013.04.007] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Revised: 04/05/2013] [Accepted: 04/25/2013] [Indexed: 11/29/2022]
Abstract
Introduction Unwanted drug interactions with ionic currents in the heart can lead to an increased proarrhythmic risk to patients in the clinic. It is therefore a priority for safety pharmacology teams to detect block of cardiac ion channels, and new technologies have enabled the development of automated and high-throughput screening assays using cell lines. As a result of screening multiple ion-channels there is a need to integrate information, particularly for compounds affecting more than one current, and mathematical electrophysiology in-silico action potential models are beginning to be used for this. Methods We quantified the variability associated with concentration-effect curves fitted to recordings from high-throughput Molecular Devices IonWorks® Quattro™ screens when detecting block of IKr (hERG), INa (NaV1.5), ICaL (CaV1.2), IKs (KCNQ1/minK) and Ito (Kv4.3/KChIP2.2), and the Molecular Devices FLIPR® Tetra fluorescence screen for ICaL (CaV1.2), for control compounds used at AstraZeneca and GlaxoSmithKline. We examined how screening variability propagates through in-silico action potential models for whole cell electrical behaviour, and how confidence intervals on model predictions can be estimated with repeated simulations. Results There are significant levels of variability associated with high-throughput ion channel electrophysiology screens. This variability is of a similar magnitude for different cardiac ion currents and different compounds. Uncertainty in the Hill coefficients of reported concentration-effect curves is particularly high. Depending on a compound’s ion channel blocking profile, the uncertainty introduced into whole-cell predictions can become significant. Discussion Our technique allows confidence intervals to be placed on computational model predictions that are based on high-throughput ion channel screens. This allows us to suggest when repeated screens should be performed to reduce uncertainty in a compound’s action to acceptable levels, to allow a meaningful interpretation of the data.
Collapse
Affiliation(s)
- Ryan C Elkins
- Global Safety Pharmacology, Global Safety Assessment, AstraZeneca, Alderley Park SK10 4TG, UK
| | | | | | | | | | | | | |
Collapse
|
25
|
Zicha S, Radresa O, Laplante P, Morton M, Jones K, Main M, Trivedi S, Julien RP, Griffin A, Labrecque J, Ahmad S, Brown W. Novel methodology to identify TRPV1 antagonists independent of capsaicin activation. ACTA ACUST UNITED AC 2012; 18:544-55. [PMID: 23264449 DOI: 10.1177/1087057112470563] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
TRPV1 was originally characterized as an integrator of various noxious stimuli such as capsaicin, heat, and protons. TRPV1-null mice exhibit a deficiency in sensing noxious heat stimuli, suggesting that TRPV1 is one of the main heat sensors on nociceptive primary afferent neurons and a candidate target for heat hypersensitivity in chronic pain. Several different potent and selective TRPV1 antagonists have been developed by more than 50 companies since the characterization of the receptor in 1997. A consequence of this competitive interest is the crowding of patentable chemical space, because very similar in vitro screening assays are used. To circumvent this issue and to expand our understanding of TRPV1 biology, we sought to take advantage of recent advancements in automated patch-clamp technology to design a novel screening cascade. This SAR-driving assay identified novel modulators that blocked the depolarization-induced activation of outwardly-rectifying TRPV1 currents independent of agonist stimulation, and we correlated the pharmacology to three other innovative assays for higher-throughput screening. Ultimately, we have identified a screening paradigm that would have good predictive value for future TRPV1 drug discovery projects and novel chemical space with a higher probability of gaining intellectual property coverage.
Collapse
|
26
|
Reducing safety-related drug attrition: the use of in vitro pharmacological profiling. Nat Rev Drug Discov 2012. [DOI: 10.1038/nrd3845] [Citation(s) in RCA: 470] [Impact Index Per Article: 39.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
27
|
A review of human pluripotent stem cell-derived cardiomyocytes for high-throughput drug discovery, cardiotoxicity screening, and publication standards. J Cardiovasc Transl Res 2012; 6:22-30. [PMID: 23229562 DOI: 10.1007/s12265-012-9423-2] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Accepted: 11/05/2012] [Indexed: 01/05/2023]
Abstract
Drug attrition rates have increased in past years, resulting in growing costs for the pharmaceutical industry and consumers. The reasons for this include the lack of in vitro models that correlate with clinical results and poor preclinical toxicity screening assays. The in vitro production of human cardiac progenitor cells and cardiomyocytes from human pluripotent stem cells provides an amenable source of cells for applications in drug discovery, disease modeling, regenerative medicine, and cardiotoxicity screening. In addition, the ability to derive human-induced pluripotent stem cells from somatic tissues, combined with current high-throughput screening and pharmacogenomics, may help realize the use of these cells to fulfill the potential of personalized medicine. In this review, we discuss the use of pluripotent stem cell-derived cardiomyocytes for drug discovery and cardiotoxicity screening, as well as current hurdles that must be overcome for wider clinical applications of this promising approach.
Collapse
|
28
|
Ido K, Ohwada T, Yasutomi E, Yoshinaga T, Arai T, Kato M, Sawada K. Screening quality for Ca2+-activated potassium channel in IonWorks Quattro is greatly improved by using BAPTA-AM and ionomycin. J Pharmacol Toxicol Methods 2012; 67:16-24. [PMID: 23138150 DOI: 10.1016/j.vascn.2012.10.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Revised: 10/12/2012] [Accepted: 10/29/2012] [Indexed: 11/25/2022]
Abstract
INTRODUCTION IonWorks automated patch clamp systems are being widely used for ion channel drug discovery, but the perforated patch mode of these systems makes it difficult to obtain a steady intracellular Ca(2+) concentration ([Ca(2+)](i)). This difficulty prevents obtaining high-quality data regarding Ca(2+)-activated channels such as BK and SK channels. We examined the methods for stabilizing [Ca(2+)](i) in the IonWorks Quattro automated patch clamp system to evaluate BK channels. METHODS Electrophysiological recordings were performed using the single-hole or population patch clamp mode of IonWorks Quattro. To increase [Ca(2+)](i), ionomycin was used. The variation in the BK current and the effect of BK channel modulators were examined in the presence and absence of an intracellular Ca(2+) chelator, BAPTA-AM (20μM). RESULTS BK current activated by step pulses to +100mV in the presence of ionomycin exhibited large variation (ranging from 0.086 to 11nA). In individual cells, oscillation of the current amplitude was observed when five repetitive pulses were applied at 0.1Hz. Approximately 30% of cells exhibited current variation exceeding 20% when the variation was calculated using the first and third pulses. However, BAPTA-AM treatment before current measurement decreased the number of cells displaying large variation (>20%) to 5%. In the presence of BAPTA-AM, the BK channel modulators NS1619 and 12,14-dichlorodehydroabietic acid increased the BK current at concentrations of 10μM or more showing clear concentration dependency, whereas in its absence, the effect of both compounds was detected only at 30μM. DISCUSSION The main finding of this study is that the [Ca(2+)](i) variation in the basal condition is very large and hinders the accurate evaluation of compounds in Ca(2+)-activated ion channels. The application of BAPTA-AM and ionomycin greatly improved the precision of BK channel screening, and this method should be applicable to other Ca(2+)-activated ion channels such as SK channels.
Collapse
Affiliation(s)
- Katsutoshi Ido
- Department of Experimental Pathology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.
| | | | | | | | | | | | | |
Collapse
|
29
|
Cros C, Skinner M, Moors J, Lainee P, Valentin JP. Detecting drug-induced prolongation of the QRS complex: new insights for cardiac safety assessment. Toxicol Appl Pharmacol 2012; 265:200-8. [PMID: 23073507 DOI: 10.1016/j.taap.2012.10.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Revised: 09/14/2012] [Accepted: 10/03/2012] [Indexed: 01/17/2023]
Abstract
BACKGROUND Drugs slowing the conduction of the cardiac action potential and prolonging QRS complex duration by blocking the sodium current (I(Na)) may carry pro-arrhythmic risks. Due to the frequency-dependent block of I(Na), this study assesses whether activity-related spontaneous increases in heart rate (HR) occurring during standard dog telemetry studies can be used to optimise the detection of class I antiarrhythmic-induced QRS prolongation. METHODS Telemetered dogs were orally dosed with quinidine (class Ia), mexiletine (class Ib) or flecainide (class Ic). QRS duration was determined standardly (5 beats averaged at rest) but also prior to and at the plateau of each acute increase in HR (3 beats averaged at steady state), and averaged over 1h period from 1h pre-dose to 5h post-dose. RESULTS Compared to time-matched vehicle, at rest, only quinidine and flecainide induced increases in QRS duration (E(max) 13% and 20% respectively, P<0.01-0.001) whereas mexiletine had no effect. Importantly, the increase in QRS duration was enhanced at peak HR with an additional effect of +0.7 ± 0.5 ms (quinidine, NS), +1.8 ± 0.8 ms (mexiletine, P<0.05) and +2.8 ± 0.8 ms (flecainide, P<0.01) (calculated as QRS at basal HR-QRS at high HR). CONCLUSION Electrocardiogram recordings during elevated HR, not considered during routine analysis optimised for detecting QT prolongation, can be used to sensitise the detection of QRS prolongation. This could prove useful when borderline QRS effects are detected. Analysing during acute increases in HR could also be useful for detecting drug-induced effects on other aspects of cardiac function.
Collapse
Affiliation(s)
- C Cros
- Safety Pharmacology, Global Safety Assessment, Safety Assessment UK, AstraZeneca R&D, Alderley Park, Macclesfield, SK10 4TG, UK.
| | | | | | | | | |
Collapse
|
30
|
Phenethyl nicotinamides, a novel class of NaV1.7 channel blockers: Structure and activity relationship. Bioorg Med Chem Lett 2012; 22:6108-15. [DOI: 10.1016/j.bmcl.2012.08.031] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2012] [Revised: 08/06/2012] [Accepted: 08/08/2012] [Indexed: 11/21/2022]
|
31
|
Structure and activity relationship in the (S)-N-chroman-3-ylcarboxamide series of voltage-gated sodium channel blockers. Bioorg Med Chem Lett 2012; 22:5618-24. [DOI: 10.1016/j.bmcl.2012.06.105] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2012] [Revised: 06/28/2012] [Accepted: 06/29/2012] [Indexed: 11/21/2022]
|
32
|
Morton MJ, Main MJ. Use of escin as a perforating agent on the IonWorks quattro automated electrophysiology platform. ACTA ACUST UNITED AC 2012; 18:128-34. [PMID: 22923788 DOI: 10.1177/1087057112456599] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The automated electrophysiology platform IonWorks has facilitated the medium-throughput study of ion channel biology and pharmacology. Electrical and chemical access to the cell is by perforated patch, afforded by amphotericin. Permeation of the amphotericin pore is limited to monovalent cations. We describe here the use of the saponin escin as an alternative perforating agent. With respect to the number and robustness of seals formed across a variety of cell and ion channel types, the performance of escin is equal to that of amphotericin. Escin also permits the permeation of larger molecules through its pore. These include nucleotides, important intracellular modulators of ion channel activity that can be used to prevent ion channel rundown of, for instance, Ca(V)1.2. Furthermore, pharmacologic agents such as QX314 can also permeate and be used for mechanistic studies. Escin, in combination with IonWorks, increases the scope of ion channel screening and can facilitate the assay of previously difficult-to-assay targets.
Collapse
Affiliation(s)
- Michael J Morton
- Global Ion Channel Initiative, AstraZeneca UK, R&D Alderley Park, Macclesfield, UK.
| | | |
Collapse
|
33
|
Macsari I, Besidski Y, Csjernyik G, Nilsson LI, Sandberg L, Yngve U, Åhlin K, Bueters T, Eriksson AB, Lund PE, Venyike E, Oerther S, Hygge Blakeman K, Luo L, Arvidsson PI. 3-Oxoisoindoline-1-carboxamides: Potent, State-Dependent Blockers of Voltage-Gated Sodium Channel NaV1.7 with Efficacy in Rat Pain Models. J Med Chem 2012; 55:6866-80. [DOI: 10.1021/jm300623u] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Per I. Arvidsson
- Organic Pharmaceutical Chemistry,
Department of Medicinal Chemistry, Uppsala Biomedical Centre, Uppsala
University, Box 574, SE-751 23 Uppsala, Sweden
- School of Pharmacy and Pharmacology,
Westville Campus, University of KwaZulu-Natal, Private Bag X54001,
Durban 4000, South Africa
| |
Collapse
|
34
|
Chronic Probucol Treatment Decreases the Slow Component of the Delayed-Rectifier Potassium Current in CHO Cells Transfected With KCNQ1 and KCNE1. J Cardiovasc Pharmacol 2012; 59:377-86. [DOI: 10.1097/fjc.0b013e318245e0c5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
35
|
Davies MR, Mistry HB, Hussein L, Pollard CE, Valentin JP, Swinton J, Abi-Gerges N. An in silico canine cardiac midmyocardial action potential duration model as a tool for early drug safety assessment. Am J Physiol Heart Circ Physiol 2012; 302:H1466-80. [DOI: 10.1152/ajpheart.00808.2011] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cell lines expressing ion channels (IC) and the advent of plate-based electrophysiology device have enabled a molecular understanding of the action potential (AP) as a means of early QT assessment. We sought to develop an in silico AP (isAP) model that provides an assessment of the effect of a compound on the myocyte AP duration (APD) using concentration-effect curve data from a panel of five ICs (hNav1.5, hCav1.2, hKv4.3/hKChIP2.2, hKv7.1/hminK, hKv11.1). A test set of 53 compounds was selected to cover a range of selective and mixed IC modulators that were tested for their effects on optically measured APD. A threshold of >10% change in APD at 90% repolarization (APD90) was used to signify an effect at the top test concentration. To capture the variations observed in left ventricular midmyocardial myocyte APD data from 19 different dogs, the isAP model was calibrated to produce an ensemble of 19 model variants that could capture the shape and form of the APs and also quantitatively replicate dofetilide- and diltiazem-induced APD90 changes. Provided with IC panel data only, the isAP model was then used, blinded, to predict APD90 changes greater than 10%. At a simulated concentration of 30 μM and based on a criterion that six of the variants had to agree, isAP prediction was scored as showing greater than 80% predictivity of compound activity. Thus, early in drug discovery, the isAP model allows integrating separate IC data and is amenable to the throughput required for use as a virtual screen.
Collapse
Affiliation(s)
| | | | - L. Hussein
- Safety Pharmacology, Safety Assessment United Kingdom, AstraZeneca R&D, Macclesfield, United Kingdom
| | - C. E. Pollard
- Safety Pharmacology, Safety Assessment United Kingdom, AstraZeneca R&D, Macclesfield, United Kingdom
| | - J.-P. Valentin
- Safety Pharmacology, Safety Assessment United Kingdom, AstraZeneca R&D, Macclesfield, United Kingdom
| | - J. Swinton
- Computational Biology, Discovery Sciences and
| | - N. Abi-Gerges
- Safety Pharmacology, Safety Assessment United Kingdom, AstraZeneca R&D, Macclesfield, United Kingdom
| |
Collapse
|
36
|
Erdemli G, Kim AM, Ju H, Springer C, Penland RC, Hoffmann PK. Cardiac Safety Implications of hNav1.5 Blockade and a Framework for Pre-Clinical Evaluation. Front Pharmacol 2012; 3:6. [PMID: 22303294 PMCID: PMC3266668 DOI: 10.3389/fphar.2012.00006] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2011] [Accepted: 01/12/2012] [Indexed: 12/17/2022] Open
Abstract
The human cardiac sodium channel (hNav1.5, encoded by the SCN5A gene) is critical for action potential generation and propagation in the heart. Drug-induced sodium channel inhibition decreases the rate of cardiomyocyte depolarization and consequently conduction velocity and can have serious implications for cardiac safety. Genetic mutations in hNav1.5 have also been linked to a number of cardiac diseases. Therefore, off-target hNav1.5 inhibition may be considered a risk marker for a drug candidate. Given the potential safety implications for patients and the costs of late stage drug development, detection, and mitigation of hNav1.5 liabilities early in drug discovery and development becomes important. In this review, we describe a pre-clinical strategy to identify hNav1.5 liabilities that incorporates in vitro, in vivo, and in silico techniques and the application of this information in the integrated risk assessment at different stages of drug discovery and development.
Collapse
Affiliation(s)
- Gül Erdemli
- Center for Proteomic Chemistry, Novartis Institutes for Biomedical Research Cambridge, MA, USA
| | | | | | | | | | | |
Collapse
|
37
|
Johnson DM, Hussein L, Spätjens RL, Valentin J, Volders PG, Abi‐Gerges N. Measurement of Action Potential Generation in Isolated Canine Left Ventricular Midmyocardial Myocytes. ACTA ACUST UNITED AC 2011; Chapter 10:Unit 10.14.1-23. [DOI: 10.1002/0471141755.ph1014s55] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Daniel M. Johnson
- Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre Maastricht The Netherlands
| | - Leyla Hussein
- Safety Assessment UK, AstraZeneca R&D, Macclesfield Cheshire United Kingdom
| | - Roel L.H.M.G. Spätjens
- Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre Maastricht The Netherlands
| | | | - Paul G.A. Volders
- Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre Maastricht The Netherlands
| | - Najah Abi‐Gerges
- Safety Assessment UK, AstraZeneca R&D, Macclesfield Cheshire United Kingdom
| |
Collapse
|
38
|
Abi-Gerges N, Holkham H, Jones EMC, Pollard CE, Valentin JP, Robertson GA. hERG subunit composition determines differential drug sensitivity. Br J Pharmacol 2011; 164:419-32. [PMID: 21449979 PMCID: PMC3188906 DOI: 10.1111/j.1476-5381.2011.01378.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2010] [Revised: 03/04/2011] [Accepted: 03/07/2011] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND AND PURPOSE The majority of human ether-a-go-go-related gene (hERG) screens aiming to minimize the risk of drug-induced long QT syndrome have been conducted using heterologous systems expressing the hERG 1a subunit, although both hERG 1a and 1b subunits contribute to the K+ channels producing the repolarizing current I(Kr) . We tested a range of compounds selected for their diversity to determine whether hERG 1a and 1a/1b channels exhibit different sensitivities that may influence safety margins or contribute to a stratified risk analysis. EXPERIMENTAL APPROACH We used the IonWorks™ plate-based electrophysiology device to compare sensitivity of hERG 1a and 1a/1b channels stably expressed in HEK293 cells to 50 compounds previously shown to target hERG channels. Potency was determined as IC₅₀ values (µM) obtained from non-cumulative, eight-point concentration-effect curves of normalized data, fitted to the Hill equation. To minimize possible sources of variability, compound potency was assessed using test plates arranged in alternating columns of cells expressing hERG 1a and 1a/1b. KEY RESULTS Although the potency of most compounds was similar for the two targets, some surprising differences were observed. Fluoxetine (Prozac) was more potent at blocking hERG 1a/1b than 1a channels, yielding a corresponding reduction in the safety margin. In contrast, E-4031 was a more potent blocker of hERG 1a compared with 1a/1b channels, as previously reported, as was dofetilide, another high-affinity blocker. CONCLUSIONS AND IMPLICATIONS The current assays may underestimate the risk of some drugs to cause torsades de pointes arrhythmia, and overestimate the risk of others.
Collapse
Affiliation(s)
- N Abi-Gerges
- Safety Pharmacology, Safety Assessment UK, AstraZeneca R&D Alderley Park, Macclesfield, UK.
| | | | | | | | | | | |
Collapse
|
39
|
Harmer AR, Valentin JP, Pollard CE. On the relationship between block of the cardiac Na⁺ channel and drug-induced prolongation of the QRS complex. Br J Pharmacol 2011; 164:260-73. [PMID: 21480866 PMCID: PMC3174407 DOI: 10.1111/j.1476-5381.2011.01415.x] [Citation(s) in RCA: 123] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2010] [Revised: 12/20/2010] [Accepted: 02/02/2011] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND AND PURPOSE Inhibition of the human cardiac Na(+) channel (hNa(v) 1.5) can prolong the QRS complex and has been associated with increased mortality in patients with underlying cardiovascular disease. The safety implications of blocking hNa(v) 1.5 channels suggest the need to test for this activity early in drug discovery in order to design out any potential liability. However, interpretation of hNa(v) 1.5 blocking potency requires knowledge of how hNa(v) 1.5 block translates into prolongation of the QRS complex. EXPERIMENTAL APPROACH We tested Class I anti-arrhythmics, other known QRS prolonging drugs and drugs not reported to prolong the QRS complex. Their block of hNa(v) 1.5 channels (as IC(50) values) was measured in an automated electrophysiology-based assay. These IC(50) values were compared with published reports of the corresponding unbound (free) plasma concentrations attained during clinical use (fC(max)) to provide an IC(50) : fC(max) ratio. KEY RESULTS For 42 Class I anti-arrhythmics and other QRS prolonging drugs, 67% had IC(50) : fC(max) ratios <30. For 55 non-QRS prolonging drugs tested, 72% had ratios >100. Finally, we determined the relationship between the IC(50) value and the free drug concentration associated with prolongation of the QRS complex in humans. For 37 drugs, QRS complex prolongation was observed at free plasma concentrations that were about 15-fold lower than the corresponding IC(50) at hNa(v) 1.5 channels. CONCLUSIONS AND IMPLICATIONS A margin of 30- to 100-fold between hNa(v) 1.5 IC(50) and fC(max) appears to confer an acceptable degree of safety from QRS prolongation. QRS prolongation occurs on average at free plasma levels 15-fold below the IC(50) at hNa(v) 1.5 channels. LINKED ARTICLE This article is commented on by Gintant et al., pp. 254-259 of this issue. To view this commentary visit http://dx.doi.org/10.1111/j.1476-5381.2011.01433.x.
Collapse
Affiliation(s)
- A R Harmer
- Safety Assessment UK, AstraZeneca R&D Alderley Park, Macclesfield, UK.
| | | | | |
Collapse
|
40
|
Gintant GA, Gallacher DJ, Pugsley MK. The 'overly-sensitive' heart: sodium channel block and QRS interval prolongation. Br J Pharmacol 2011; 164:254-9. [PMID: 21488862 PMCID: PMC3174406 DOI: 10.1111/j.1476-5381.2011.01433.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2011] [Accepted: 04/06/2011] [Indexed: 11/30/2022] Open
Abstract
UNLABELLED Cardiac safety remains of paramount importance in the development of successful clinical drug candidates. Great progress has been made recently in understanding liabilities associated with delayed ventricular repolarization (manifest as QT prolongation) and in predicting (thus avoiding) drugs that delay repolarization based on application of strategic preclinical assays. Following the advances made in clinical electrophysiological monitoring and conduct of thorough QT studies, focus is now shifting towards monitoring of additional drug-induced effects, particularly on ventricular conduction (measured as changes in the QRS interval on the ECG) as part of evolving clinical thorough ECG studies. In this issue of the British Journal of Pharmacology, a study by Harmer et al. proposes provisional safety margins for QRS prolongation in man based on retrospective clinical data and a single in vitro approach to assess potency of block of cardiac sodium current (hNav1.5), the ionic current responsible for ventricular conduction (observed as QRS prolongation). The present commentary places their study in context with evolving preclinical cardiac electrophysiological safety assessments, along with discussions focused on ensuring the proper 'translation' of preclinical findings with potential clinical concerns. Given the extant limitations and uncertainties of presently available data, as well as our limited understanding of the pro-arrhythmic potential associated with these changes, due caution should be applied when considering the proposed in vitro-based margins for drug-induced QRS prolongation measured clinically. Additional validation with multiple preclinical models and more rigorous clinical safety studies will be necessary to substantiate these recommended margins. LINKED ARTICLE This article is a commentary on Harmer et al., pp. 260-273 of this issue. To view this paper visit http://dx.doi.org/10.1111/j.1476-5381.2011.01415.x.
Collapse
Affiliation(s)
- Gary A Gintant
- Department of Integrative Pharmacology, Abbott LaboratoriesAbbott Park, IL, USA
| | - David J Gallacher
- Center of Excellence for Cardiovascular Safety Research and Mechanistic Pharmacology, Janssen Pharmaceutical Companies of Johnson & JohnsonBeerse, Belgium
| | - Michael K Pugsley
- Department of Toxicology & Pathology, Johnson & Johnson Pharmaceutical Research & DevelopmentRaritan, NJ, USA
| |
Collapse
|
41
|
Bass AS, Vargas HM, Valentin JP, Kinter LB, Hammond T, Wallis R, Siegl PK, Yamamoto K. Safety pharmacology in 2010 and beyond: Survey of significant events of the past 10years and a roadmap to the immediate-, intermediate- and long-term future in recognition of the tenth anniversary of the Safety Pharmacology Society. J Pharmacol Toxicol Methods 2011; 64:7-15. [DOI: 10.1016/j.vascn.2011.05.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2011] [Accepted: 05/19/2011] [Indexed: 11/29/2022]
|
42
|
Macsari I, Sandberg L, Besidski Y, Gravenfors Y, Ginman T, Bylund J, Bueters T, Eriksson AB, Lund PE, Venyike E, Arvidsson PI. Phenyl isoxazole voltage-gated sodium channel blockers: structure and activity relationship. Bioorg Med Chem Lett 2011; 21:3871-6. [PMID: 21641215 DOI: 10.1016/j.bmcl.2011.05.041] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2011] [Revised: 05/11/2011] [Accepted: 05/12/2011] [Indexed: 11/26/2022]
Abstract
Blocking of certain sodium channels is considered to be an attractive mechanism to treat chronic pain conditions. Phenyl isoxazole carbamate 1 was identified as a potent and selective Na(V)1.7 blocker. Structural analogues of 1, both carbamates, ureas and amides, were proven to be useful in establishing the structure-activity relationship and improving ADME related properties. Amide 24 showed a good overall in vitro profile, that translated well to rat in vivo PK.
Collapse
Affiliation(s)
- Istvan Macsari
- Medicinal Chemistry, CNSP iMed Science, AstraZeneca R&D, Innovative Medicines, SE-15185 Södertälje, Sweden.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Abela D, Ritchie H, Ababneh D, Gavin C, Nilsson MF, Khan MK, Carlsson K, Webster WS. The effect of drugs with ion channel-blocking activity on the early embryonic rat heart. ACTA ACUST UNITED AC 2011; 89:429-40. [PMID: 20973055 DOI: 10.1002/bdrb.20270] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
This study investigated the effects of a range of pharmaceutical drugs with ion channel-blocking activity on the heart of gestation day 13 rat embryos in vitro. The general hypothesis was that the blockade of the I(Kr)/hERG channel, that is highly important for the normal functioning of the embryonic rat heart, would cause bradycardia and arrhythmia. Concomitant blockade of other channels was expected to modify the effects of hERG blockade. Fourteen drugs with varying degrees of specificity and affinity toward potassium, sodium, and calcium channels were tested over a range of concentrations. The rat embryos were maintained for 2 hr in culture, 1 hr to acclimatize, and 1 hr to test the effect of the drug. All the drugs caused a concentration-dependent bradycardia except nifedipine, which primarily caused a negative inotropic effect eventually stopping the heart. A number of drugs induced arrhythmias and these appeared to be related to either sodium channel blockade, which resulted in a double atrial beat for each ventricular beat, or I(Kr)/hERG blockade, which caused irregular atrial and ventricular beats. However, it is difficult to make a precise prediction of the effect of a drug on the embryonic heart just by looking at the polypharmacological action on ion channels. The results indicate that the use of the tested drugs during pregnancy could potentially damage the embryo by causing periods of hypoxia. In general, the effects on the embryonic heart were only seen at concentrations greater than those likely to occur with normal therapeutic dosing.
Collapse
Affiliation(s)
- Dominique Abela
- Department of Anatomy and Histology, Sydney Medical School, University of Sydney, Sydney, Australia
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Cao X, Lee YT, Holmqvist M, Lin Y, Ni Y, Mikhailov D, Zhang H, Hogan C, Zhou L, Lu Q, Digan ME, Urban L, Erdemli G. Cardiac ion channel safety profiling on the IonWorks Quattro automated patch clamp system. Assay Drug Dev Technol 2010; 8:766-80. [PMID: 21133679 DOI: 10.1089/adt.2010.0333] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The normal electrophysiologic behavior of the heart is determined by the integrated activity of specific cardiac ionic currents. Mutations in genes encoding the molecular components of individual cardiac ion currents have been shown to result in multiple cardiac arrhythmia syndromes. Presently, 12 genes associated with inherited long QT syndrome (LQTS) have been identified, and the most common mutations are in the hKCNQ1 (LQT1, Jervell and Lange-Nielson syndrome), hKCNH2 (LQT2), and hSCN5A (LQT3, Brugada syndrome) genes. Several drugs have been withdrawn from the market or received black box labeling due to clinical cases of QT interval prolongation, ventricular arrhythmias, and sudden death. Other drugs have been denied regulatory approval owing to their potential for QT interval prolongation. Further, off-target activity of drugs on cardiac ion channels has been shown to be associated with increased mortality in patients with underlying cardiovascular diseases. Since clinical arrhythmia risk is a major cause for compound termination, preclinical profiling for off-target cardiac ion channel interactions early in the drug discovery process has become common practice in the pharmaceutical industry. In the present study, we report assay development for three cardiac ion channels (hKCNQ1/minK, hCa(v)1.2, and hNa(v)1.5) on the IonWorks Quattro™ system. We demonstrate that these assays can be used as reliable pharmacological profiling tools for cardiac ion channel inhibition to assess compounds for cardiac liability during drug discovery.
Collapse
Affiliation(s)
- Xueying Cao
- Center for Proteomic Chemistry, Novartis Institutes for BioMedical Sciences Inc., Cambridge, Massachusetts 02139, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
For every movement, heartbeat and thought, ion channels need to open and close. It is therefore not surprising that their malfunctioning leads to serious diseases. Currently, only approximately 10% of drugs, with a market value in excess of US$10 billion, act on ion channels. The systematic exploitation of this target class has started, enabled by novel assay technologies and fundamental advances of the structural and mechanistic understanding of channel function. The latter, which was rewarded with the Nobel Prize in 2003, has opened up an avenue for rational drug design. In this review we provide an overview of the current repertoire of screening technologies that has evolved to drive ion channel-targeted drug discovery towards new medicines of the future.
Collapse
|
46
|
Pollard CE, Abi Gerges N, Bridgland-Taylor MH, Easter A, Hammond TG, Valentin JP. An introduction to QT interval prolongation and non-clinical approaches to assessing and reducing risk. Br J Pharmacol 2010; 159:12-21. [PMID: 20141516 DOI: 10.1111/j.1476-5381.2009.00207.x] [Citation(s) in RCA: 125] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Owing to its association with Torsades de Pointes, drug-induced QT interval prolongation has been and remains a significant hurdle to the development of safe, effective medicines. Genetic and pharmacological evidence highlighting the pivotal role the human ether-a-go-go-related gene (hERG) channel was a critical step in understanding how to start addressing this issue. It led to the development of hERG assays with the rapid throughput needed for the short timescales required in early drug discovery. The resulting volume of hERG data has fostered in silico models to help chemists design compounds with reduced hERG potency. In early drug discovery, a pragmatic approach based on exceeding a given potency value has been required to decide when a compound is likely to carry a low QT risk, to support its progression to late-stage discovery. At this point, the in vivo efficacy and metabolism characteristics of the potential drug are generally defined, as well its safety profile, which includes usually a dog study to assess QT interval prolongation risk. The hERG and in vivo QT data, combined with the likely indication and the estimated free drug level for efficacy, are put together to assess the risk that the potential drug will prolong QT in man. Further data may be required to refine the risk assessment before making the major investment decisions for full development. The non-clinical data are essential to inform decisions about compound progression and to optimize the design of clinical QT studies.
Collapse
Affiliation(s)
- Chris E Pollard
- Safety Assessment UK, AstraZeneca R&D, Alderley Park, Macclesfield, Cheshire, UK.
| | | | | | | | | | | |
Collapse
|
47
|
Abstract
Drugs inhibiting voltage-gated sodium channels have long been used as analgesics, beginning with the use of local anaesthetics for sensory blockade and then with the discovery that Nav-blocking anticonvulsants also have benefit for pain therapy. These drugs were discovered without knowledge of their molecular target, using traditional pharmacological methods, and their clinical utility is limited by relatively narrow therapeutic windows. Until recently, attempts to develop improved inhibitors using modern molecular-targeted screening approaches have met with limited success. However, in the last few years there has been renewed activity following the discovery of human Nav1.7 mutations that cause striking insensitivity to pain. Together with recent advances in the technologies required to prosecute ion channels as drug targets, this has led to significant progress being made. This article reviews these developments and summarises current findings with these emerging new Nav inhibitors, highlighting some of the unanswered questions and the challenges that remain before they can be developed for clinical use.
Collapse
Affiliation(s)
- Jeffrey J Clare
- Cell-Based Assays Group, Millipore Corporation, St Charles, Missouri 63304, USA.
| |
Collapse
|
48
|
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: 1.2] [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
|
49
|
Redfern WS, Waldron G, Winter MJ, Butler P, Holbrook M, Wallis R, Valentin JP. Zebrafish assays as early safety pharmacology screens: Paradigm shift or red herring? J Pharmacol Toxicol Methods 2008; 58:110-7. [DOI: 10.1016/j.vascn.2008.05.006] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2008] [Accepted: 05/08/2008] [Indexed: 12/22/2022]
|
50
|
Valentin JP, Hammond T. Safety and secondary pharmacology: Successes, threats, challenges and opportunities. J Pharmacol Toxicol Methods 2008; 58:77-87. [DOI: 10.1016/j.vascn.2008.05.007] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2008] [Accepted: 05/19/2008] [Indexed: 12/15/2022]
|