1
|
Jia J, Lietz S, Barth H, Ernst K. The antiarrhythmic drugs amiodarone and dronedarone inhibit intoxication of cells with pertussis toxin. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:9991-10003. [PMID: 38958734 PMCID: PMC11582147 DOI: 10.1007/s00210-024-03247-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 06/17/2024] [Indexed: 07/04/2024]
Abstract
Pertussis toxin (PT) is a virulent factor produced by Bordetella pertussis, the causative agent of whooping cough. PT exerts its pathogenic effects by ADP-ribosylating heterotrimeric G proteins, disrupting cellular signaling pathways. Here, we investigate the potential of two antiarrhythmic drugs, amiodarone and dronedarone, in mitigating PT-induced cellular intoxication. After binding to cells, PT is endocytosed, transported from the Golgi to the endoplasmic reticulum where the enzyme subunit PTS1 is released from the transport subunit of PT. PTS1 is translocated into the cytosol where it ADP-ribosylates inhibitory α-subunit of G-protein coupled receptors (Gαi). We showed that amiodarone and dronedarone protected CHO cells and human A549 cells from PT-intoxication by analyzing the ADP-ribosylation status of Gαi. Amiodarone had no effect on PT binding to cells or in vitro enzyme activity of PTS1 but reduced the signal of PTS1 in the cell suggesting that amiodarone interferes with intracellular transport of PTS1. Moreover, dronedarone mitigated the PT-mediated effect on cAMP signaling in a cell-based bioassay. Taken together, our findings underscore the inhibitory effects of amiodarone and dronedarone on PT-induced cellular intoxication, providing valuable insights into drug repurposing for infectious disease management.
Collapse
Affiliation(s)
- Jinfang Jia
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, Ulm University Medical Center, Ulm, Germany
- Department of Respiratory Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Stefanie Lietz
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, Ulm University Medical Center, Ulm, Germany
| | - Holger Barth
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, Ulm University Medical Center, Ulm, Germany.
| | - Katharina Ernst
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, Ulm University Medical Center, Ulm, Germany.
| |
Collapse
|
2
|
de Lima Conceição MR, Teixeira-Fonseca JL, Marques LP, Souza DS, da Silva Alcântara F, Orts DJB, Roman-Campos D. Extracellular acidification reveals the antiarrhythmic properties of amiodarone related to late sodium current-induced atrial arrhythmia. Pharmacol Rep 2024; 76:585-599. [PMID: 38619735 DOI: 10.1007/s43440-024-00597-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 04/05/2024] [Accepted: 04/08/2024] [Indexed: 04/16/2024]
Abstract
BACKGROUND Amiodarone (AMIO) is an antiarrhythmic drug with the pKa in the physiological range. Here, we explored how mild extracellular pH (pHe) changes shape the interaction of AMIO with atrial tissue and impact its pharmacological properties in the classical model of sea anemone sodium channel neurotoxin type 2 (ATX) induced late sodium current (INa-Late) and arrhythmias. METHOD Isolated atrial cardiomyocytes from male Wistar rats and human embryonic kidney cells expressing SCN5A Na+ channels were used for patch-clamp experiments. Isolated right atria (RA) and left atria (LA) tissue were used for bath organ experiments. RESULTS A more acidophilic pHe caused negative inotropic effects on isolated RA and LA atrial tissue, without modification of the pharmacological properties of AMIO. A pHe of 7.0 changed the sodium current (INa) related components of the action potential (AP), which was enhanced in the presence of AMIO. ATXinduced arrhythmias in isolated RA and LA. Also, ATX prolonged the AP duration and enhanced repolarization dispersion in isolated cardiomyocytes in both pHe 7.4 and pHe 7.0. Pre-incubation of the isolated RA and LA and isolated atrial cardiomyocytes with AMIO prevented arrhythmias induced by ATX only at a pHe of 7.0. Moreover, AMIO was able to block INa-Late induced by ATX only at a pHe of 7.0. CONCLUSION The pharmacological properties of AMIO concerning healthy rat atrial tissue are not dependent on pHe. However, the prevention of arrhythmias induced by INa-Late is pHe-dependent. The development of drugs analogous to AMIO with charge stabilization may help to create more effective drugs to treat arrhythmias related to the INa-Late.
Collapse
Affiliation(s)
- Michael Ramon de Lima Conceição
- Laboratory of CardioBiology, Department of Biophysics, Paulista School of Medicine, Federal University of Sao Paulo Botucatu Street, 862, Biological Science Building, 7th floor,, São Paulo, Brazil
| | - Jorge Lucas Teixeira-Fonseca
- Laboratory of CardioBiology, Department of Biophysics, Paulista School of Medicine, Federal University of Sao Paulo Botucatu Street, 862, Biological Science Building, 7th floor,, São Paulo, Brazil
| | - Leisiane Pereira Marques
- Laboratory of CardioBiology, Department of Biophysics, Paulista School of Medicine, Federal University of Sao Paulo Botucatu Street, 862, Biological Science Building, 7th floor,, São Paulo, Brazil
| | - Diego Santos Souza
- Department of Physiology, Federal University of Sergipe, São Cristóvão, Brazil
| | - Fabiana da Silva Alcântara
- Laboratory of CardioBiology, Department of Biophysics, Paulista School of Medicine, Federal University of Sao Paulo Botucatu Street, 862, Biological Science Building, 7th floor,, São Paulo, Brazil
| | - Diego Jose Belato Orts
- Laboratory of CardioBiology, Department of Biophysics, Paulista School of Medicine, Federal University of Sao Paulo Botucatu Street, 862, Biological Science Building, 7th floor,, São Paulo, Brazil
| | - Danilo Roman-Campos
- Laboratory of CardioBiology, Department of Biophysics, Paulista School of Medicine, Federal University of Sao Paulo Botucatu Street, 862, Biological Science Building, 7th floor,, São Paulo, Brazil.
| |
Collapse
|
3
|
Schumacher J, Nienhaus A, Heber S, Matylitsky J, Chaves-Olarte E, Rodríguez C, Barth H, Papatheodorou P. Exploring the inhibitory potential of the antiarrhythmic drug amiodarone against Clostridioides difficile toxins TcdA and TcdB. Gut Microbes 2023; 15:2256695. [PMID: 37749884 PMCID: PMC10524773 DOI: 10.1080/19490976.2023.2256695] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 09/05/2023] [Indexed: 09/27/2023] Open
Abstract
The intestinal pathogen Clostridioides difficile is the leading cause of antibiotic-associated diarrhea and pseudomembranous colitis in humans. The symptoms of C. difficile-associated diseases (CDADs) are directly associated with the pathogen's toxins TcdA and TcdB, which enter host cells and inactivate Rho and/or Ras GTPases by glucosylation. Membrane cholesterol is crucial during the intoxication process of TcdA and TcdB, and likely involved during pore formation of both toxins in endosomal membranes, a key step after cellular uptake for the translocation of the glucosyltransferase domain of both toxins from endosomes into the host cell cytosol. The licensed drug amiodarone, a multichannel blocker commonly used in the treatment of cardiac dysrhythmias, is also capable of inhibiting endosomal acidification and, as shown recently, cholesterol biosynthesis. Thus, we were keen to investigate in vitro with cultured cells and human intestinal organoids, whether amiodarone preincubation protects from TcdA and/or TcdB intoxication. Amiodarone conferred protection against both toxins independently and in combination as well as against toxin variants from the clinically relevant, epidemic C. difficile strain NAP1/027. Further mechanistic studies suggested that amiodarone's mode-of-inhibition involves also interference with the translocation pore of both toxins. Our study opens the possibility of repurposing the licensed drug amiodarone as a novel pan-variant antitoxin therapeutic in the context of CDADs.
Collapse
Affiliation(s)
- Judith Schumacher
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, Ulm University Medical Center, Ulm, Germany
| | - Astrid Nienhaus
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, Ulm University Medical Center, Ulm, Germany
| | - Sebastian Heber
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, Ulm University Medical Center, Ulm, Germany
| | - Jauheni Matylitsky
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, Ulm University Medical Center, Ulm, Germany
| | - Esteban Chaves-Olarte
- Centro de Investigación en Enfermedades Tropicales and Facultad de Microbiología, Universidad de Costa Rica, San José, Costa Rica
| | - César Rodríguez
- Centro de Investigación en Enfermedades Tropicales and Facultad de Microbiología, Universidad de Costa Rica, San José, Costa Rica
| | - Holger Barth
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, Ulm University Medical Center, Ulm, Germany
| | - Panagiotis Papatheodorou
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, Ulm University Medical Center, Ulm, Germany
| |
Collapse
|
4
|
A predictive in vitro risk assessment platform for pro-arrhythmic toxicity using human 3D cardiac microtissues. Sci Rep 2021; 11:10228. [PMID: 33986332 PMCID: PMC8119415 DOI: 10.1038/s41598-021-89478-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 04/12/2021] [Indexed: 12/19/2022] Open
Abstract
Cardiotoxicity of pharmaceutical drugs, industrial chemicals, and environmental toxicants can be severe, even life threatening, which necessitates a thorough evaluation of the human response to chemical compounds. Predicting risks for arrhythmia and sudden cardiac death accurately is critical for defining safety profiles. Currently available approaches have limitations including a focus on single select ion channels, the use of non-human species in vitro and in vivo, and limited direct physiological translation. We have advanced the robustness and reproducibility of in vitro platforms for assessing pro-arrhythmic cardiotoxicity using human induced pluripotent stem cell-derived cardiomyocytes and human cardiac fibroblasts in 3-dimensional microtissues. Using automated algorithms and statistical analyses of eight comprehensive evaluation metrics of cardiac action potentials, we demonstrate that tissue-engineered human cardiac microtissues respond appropriately to physiological stimuli and effectively differentiate between high-risk and low-risk compounds exhibiting blockade of the hERG channel (E4031 and ranolazine, respectively). Further, we show that the environmental endocrine disrupting chemical bisphenol-A (BPA) causes acute and sensitive disruption of human action potentials in the nanomolar range. Thus, this novel human 3D in vitro pro-arrhythmic risk assessment platform addresses critical needs in cardiotoxicity testing for both environmental and pharmaceutical compounds and can be leveraged to establish safe human exposure levels.
Collapse
|
5
|
Strauss DG, Wu WW, Li Z, Koerner J, Garnett C. Translational Models and Tools to Reduce Clinical Trials and Improve Regulatory Decision Making for QTc and Proarrhythmia Risk (ICH E14/S7B Updates). Clin Pharmacol Ther 2021; 109:319-333. [PMID: 33332579 PMCID: PMC7898549 DOI: 10.1002/cpt.2137] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 12/14/2020] [Indexed: 01/06/2023]
Abstract
After multiple drugs were removed from the market secondary to drug-induced torsade de pointes (TdP) risk, the International Council for Harmonisation (ICH) released guidelines in 2005 that focused on the nonclinical (S7B) and clinical (E14) assessment of surrogate biomarkers for TdP. Recently, Vargas et al. published a pharmaceutical-industry perspective making the case that "double-negative" nonclinical data (negative in vitro hERG and in vivo heart-rate corrected QT (QTc) assays) are associated with such low probability of clinical QTc prolongation and TdP that potentially all double-negative drugs would not need detailed clinical QTc evaluation. Subsequently, the ICH released a new E14/S7B Draft Guideline containing Questions and Answers (Q&As) that defined ways that double-negative nonclinical data could be used to reduce the number of "Thorough QT" (TQT) studies and reach a low-risk determination when a TQT or equivalent could not be performed. We review the Vargas et al. proposal in the context of what was contained in the ICH E14/S7B Draft Guideline and what was proposed by the ICH E14/S7B working group for a "stage 2" of updates (potential expanded roles for nonclinical data and details for assessing TdP risk of QTc-prolonging drugs). Although we do not agree with the exact probability statistics in the Vargas et al. paper because of limitations in the underlying datasets, we show how more modest predictive value of individual assays could still result in low probability for TdP with double-negative findings. Furthermore, we expect that the predictive value of the nonclinical assays will improve with implementation of the new ICH E14/S7B Draft Guideline.
Collapse
Affiliation(s)
- David G. Strauss
- Division of Applied Regulatory ScienceOffice of Clinical PharmacologyOffice of Translational SciencesCenter for Drug Evaluation and ResearchUS Food and Drug AdministrationSilver SpringMarylandUSA
| | - Wendy W. Wu
- Division of Applied Regulatory ScienceOffice of Clinical PharmacologyOffice of Translational SciencesCenter for Drug Evaluation and ResearchUS Food and Drug AdministrationSilver SpringMarylandUSA
| | - Zhihua Li
- Division of Applied Regulatory ScienceOffice of Clinical PharmacologyOffice of Translational SciencesCenter for Drug Evaluation and ResearchUS Food and Drug AdministrationSilver SpringMarylandUSA
| | - John Koerner
- Division of Pharm/Tox for Cardiology, Hematology, Endocrinology and NephrologyOffice of Cardiology, Hematology, Endocrinology and NephrologyOffice of New DrugsCenter for Drug Evaluation and ResearchUS Food and Drug AdministrationSilver SpringMarylandUSA
| | - Christine Garnett
- Division of Cardiology and NephrologyOffice of Cardiology, Hematology, Endocrinology and NephrologyOffice of New DrugsCenter for Drug Evaluation and ResearchUS Food and Drug AdministrationSilver SpringMarylandUSA
| |
Collapse
|
6
|
Beserra SS, Santos-Miranda A, Sarmento JO, Miranda VM, Roman-Campos D. Effects of amiodarone on rodent ventricular cardiomyocytes: Novel perspectives from a cellular model of Long QT Syndrome Type 3. Life Sci 2020; 255:117814. [PMID: 32439300 DOI: 10.1016/j.lfs.2020.117814] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 05/04/2020] [Accepted: 05/15/2020] [Indexed: 10/24/2022]
Abstract
AIMS Amiodarone (AMIO) is currently used in medical practice to reverse ventricular tachycardia. Here we determine the effects of AMIO in the electromechanical properties of isolated left ventricle myocyte (LVM) from mice and guinea pig and in a cellular model of Long QT Syndrome Type 3 (LQTS-3) using anemone neurotoxin 2 (ATX II), which induces increase of late sodium current in LVM. MAIN METHODS AND KEY FINDINGS Using patch-clamp technique, fluorescence imaging to detect cellular Ca2+ transient and sarcomere detection systems we evaluate the effect of AMIO in healthy LVM. AMIO produced a significant reduction in the percentage of sarcomere shortening (0.1, 1 and 10 μM) in a range of pacing frequencies, however, without significant attenuation of Ca2+ transient. Also, 10 μM of AMIO caused the opposite effect on action potential repolarization of mouse and guinea pig LVM. When LVM from mouse and guinea pig were paced in a range of pacing frequencies and exposed to ATX (10 nM), AMIO (10 μM) was only able to abrogate electromechanical arrhythmias in LVM from guinea pig at lower pacing frequency. SIGNIFICANCE AMIO has negative inotropic effect with opposite effect on action potential waveform in mouse and guinea pig LVM. Furthermore, the antiarrhythmic action of AMIO in LQTS-3 is species and frequency-dependent, which indicates that AMIO may be beneficial for some types of arrhythmias related to late sodium current.
Collapse
Affiliation(s)
- Samuel Santos Beserra
- Laboratory of CardioBiology, Department of Biophysics, Paulista School of Medicina, Federal University of Sao Paulo, Brazil
| | - Artur Santos-Miranda
- Laboratory of CardioBiology, Department of Biophysics, Paulista School of Medicina, Federal University of Sao Paulo, Brazil
| | - Jaqueline Oliveira Sarmento
- Laboratory of CardioBiology, Department of Biophysics, Paulista School of Medicina, Federal University of Sao Paulo, Brazil
| | - Victor Martins Miranda
- Laboratory of CardioBiology, Department of Biophysics, Paulista School of Medicina, Federal University of Sao Paulo, Brazil
| | - Danilo Roman-Campos
- Laboratory of CardioBiology, Department of Biophysics, Paulista School of Medicina, Federal University of Sao Paulo, Brazil.
| |
Collapse
|
7
|
Hwang J, Kim TY, Terentyev D, Zhong M, Kabakov AY, Bronk P, Arunachalam K, Belardinelli L, Rajamani S, Kunitomo Y, Pfeiffer Z, Lu Y, Peng X, Odening KE, Qu Z, Karma A, Koren G, Choi BR. Late I Na Blocker GS967 Supresses Polymorphic Ventricular Tachycardia in a Transgenic Rabbit Model of Long QT Type 2. Circ Arrhythm Electrophysiol 2020; 13:e006875. [PMID: 32628505 PMCID: PMC10626560 DOI: 10.1161/circep.118.006875] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
BACKGROUND Long QT syndrome has been associated with sudden cardiac death likely caused by early afterdepolarizations (EADs) and polymorphic ventricular tachycardias (PVTs). Suppressing the late sodium current (INaL) may counterbalance the reduced repolarization reserve in long QT syndrome and prevent EADs and PVTs. METHODS We tested the effects of the selective INaL blocker GS967 on PVT induction in a transgenic rabbit model of long QT syndrome type 2 using intact heart optical mapping, cellular electrophysiology and confocal Ca2+ imaging, and computer modeling. RESULTS GS967 reduced ventricular fibrillation induction under a rapid pacing protocol (n=7/14 hearts in control versus 1/14 hearts at 100 nmol/L) without altering action potential duration or restitution and dispersion. GS967 suppressed PVT incidences by reducing Ca2+-mediated EADs and focal activity during isoproterenol perfusion (at 30 nmol/L, n=7/12 and 100 nmol/L n=8/12 hearts without EADs and PVTs). Confocal Ca2+ imaging of long QT syndrome type 2 myocytes revealed that GS967 shortened Ca2+ transient duration via accelerating Na+/Ca2+ exchanger (INCX)-mediated Ca2+ efflux from cytosol, thereby reducing EADs. Computer modeling revealed that INaL potentiates EADs in the long QT syndrome type 2 setting through (1) providing additional depolarizing currents during action potential plateau phase, (2) increasing intracellular Na+ (Nai) that decreases the depolarizing INCX thereby suppressing the action potential plateau and delaying the activation of slowly activating delayed rectifier K+ channels (IKs), suggesting important roles of INaL in regulating Nai. CONCLUSIONS Selective INaL blockade by GS967 prevents EADs and abolishes PVT in long QT syndrome type 2 rabbits by counterbalancing the reduced repolarization reserve and normalizing Nai. Graphic Abstract: A graphic abstract is available for this article.
Collapse
Affiliation(s)
- Jungmin Hwang
- Cardiovascular Rsrch Ctr, Division of Cardiology, Rhode Island Hospital, Alpert Medical School of Brown Univ, Providence
- College of Pharmacy, Univ of Rhode Island, Kingstown, RI
| | - Tae Yun Kim
- Cardiovascular Rsrch Ctr, Division of Cardiology, Rhode Island Hospital, Alpert Medical School of Brown Univ, Providence
| | - Dmitry Terentyev
- Cardiovascular Rsrch Ctr, Division of Cardiology, Rhode Island Hospital, Alpert Medical School of Brown Univ, Providence
| | | | - Anatoli Y. Kabakov
- Cardiovascular Rsrch Ctr, Division of Cardiology, Rhode Island Hospital, Alpert Medical School of Brown Univ, Providence
| | - Peter Bronk
- Cardiovascular Rsrch Ctr, Division of Cardiology, Rhode Island Hospital, Alpert Medical School of Brown Univ, Providence
| | - Karuppiah Arunachalam
- Cardiovascular Rsrch Ctr, Division of Cardiology, Rhode Island Hospital, Alpert Medical School of Brown Univ, Providence
| | | | - Sridharan Rajamani
- Former employee: Dept of Biology, Gilead Science, Foster City, CA
- Amgen Inc, South San Francisco, CA
| | - Yukiko Kunitomo
- Cardiovascular Rsrch Ctr, Division of Cardiology, Rhode Island Hospital, Alpert Medical School of Brown Univ, Providence
| | - Zachary Pfeiffer
- Cardiovascular Rsrch Ctr, Division of Cardiology, Rhode Island Hospital, Alpert Medical School of Brown Univ, Providence
| | - Yichun Lu
- Cardiovascular Rsrch Ctr, Division of Cardiology, Rhode Island Hospital, Alpert Medical School of Brown Univ, Providence
| | - Xuwen Peng
- Dept of Comparative Medicine, Pennsylvania State Univ College of Medicine, Hershey, PA
| | - Katja E. Odening
- Dept of Cardiology & Angiology I, Heart Ctr, Univ of Freiburg, Germany
| | - Zhilin Qu
- Dept of Medicine, Univ of California, Los Angeles
| | - Alain Karma
- Dept of Physics, Northeastern Univ, Boston, MA
| | - Gideon Koren
- Cardiovascular Rsrch Ctr, Division of Cardiology, Rhode Island Hospital, Alpert Medical School of Brown Univ, Providence
| | - Bum-Rak Choi
- Cardiovascular Rsrch Ctr, Division of Cardiology, Rhode Island Hospital, Alpert Medical School of Brown Univ, Providence
| |
Collapse
|
8
|
Corino VDA, Rivolta MW, Mainardi LT, Sassi R. Assessment of spatial heterogeneity of ventricular repolarization after multi-channel blocker drugs in healthy subjects. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2020; 189:105291. [PMID: 31935579 DOI: 10.1016/j.cmpb.2019.105291] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 12/18/2019] [Accepted: 12/20/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND AND OBJECTIVES In contrast to potassium channel blockers, drugs affecting multiple channels seem to reduce torsadogenic risks. However, their effect on spatial heterogeneity of ventricular repolarization (SHVR) is still matter of investigation. Aim of this work is to assess the effect of four drugs blocking the human ether-à-go-go-related gene (hERG) potassium channel, alone or in combination with other ionic channel blocks, on SHVR, as estimated by the V-index on short triplicate 10 s ECG. METHODS The V-index is an estimate of the standard deviation of the repolarization times of the myocytes across the entire myocardium, obtained from multi-lead surface electrocardiograms. Twenty-two healthy subjects received a pure hERG potassium channel blocker (dofetilide) and 3 other drugs with additional varying degrees of sodium and calcium (L-type) channel block (quinidine, ranolazine, and verapamil), as well as placebo. A one-way repeated-measures Friedman test was performed to compare the V-index over time. RESULTS Computer simulations and Bland-Altman analysis supported the reliability of the estimates of V-index on triplicate 10 s ECG. Ranolazine, verapamil and placebo did not affect the V-index. On the contrary, after quinidine and dofetilide administration, an increase of V-index from predose to its peak value was observed (ΔΔV-index values were 19 ms and 27 ms, respectively, p < 0.05). CONCLUSIONS High torsadogenic drugs (dofetilide and quinidine) affected significantly the SHVR, as quantified by the V-index. The metric has therefore a potential in assessing drug arrhythmogenicity.
Collapse
Affiliation(s)
- Valentina D A Corino
- Dipartimento di Elettronica, Informazione e Bioingegneria, Politecnico di Milano, via Golgi 39, 20133 Milan, Italy.
| | - Massimo W Rivolta
- Dipartimento di Informatica, Università degli Studi di Milano, Via Celoria 18, 20133 Milan, Italy
| | - Luca T Mainardi
- Dipartimento di Elettronica, Informazione e Bioingegneria, Politecnico di Milano, via Golgi 39, 20133 Milan, Italy
| | - Roberto Sassi
- Dipartimento di Informatica, Università degli Studi di Milano, Via Celoria 18, 20133 Milan, Italy
| |
Collapse
|
9
|
Hegyi B, Chen-Izu Y, Izu LT, Rajamani S, Belardinelli L, Bers DM, Bányász T. Balance Between Rapid Delayed Rectifier K + Current and Late Na + Current on Ventricular Repolarization: An Effective Antiarrhythmic Target? Circ Arrhythm Electrophysiol 2020; 13:e008130. [PMID: 32202931 PMCID: PMC7331791 DOI: 10.1161/circep.119.008130] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 03/16/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND Rapid delayed rectifier K+ current (IKr) and late Na+ current (INaL) significantly shape the cardiac action potential (AP). Changes in their magnitudes can cause either long or short QT syndromes associated with malignant ventricular arrhythmias and sudden cardiac death. METHODS Physiological self AP-clamp was used to measure INaL and IKr during the AP in rabbit and porcine ventricular cardiomyocytes to test our hypothesis that the balance between IKr and INaL affects repolarization stability in health and disease conditions. RESULTS We found comparable amount of net charge carried by IKr and INaL during the physiological AP, suggesting that outward K+ current via IKr and inward Na+ current via INaL are in balance during physiological repolarization. Remarkably, IKr and INaL integrals in each control myocyte were highly correlated in both healthy rabbit and pig myocytes, despite high overall cell-to-cell variability. This close correlation was lost in heart failure myocytes from both species. Pretreatment with E-4031 to block IKr (mimicking long QT syndrome 2) or with sea anemone toxin II to impair Na+ channel inactivation (mimicking long QT syndrome 3) prolonged AP duration (APD); however, using GS-967 to inhibit INaL sufficiently restored APD to control in both cases. Importantly, INaL inhibition significantly reduced the beat-to-beat and short-term variabilities of APD. Moreover, INaL inhibition also restored APD and repolarization stability in heart failure. Conversely, pretreatment with GS-967 shortened APD (mimicking short QT syndrome), and E-4031 reverted APD shortening. Furthermore, the amplitude of AP alternans occurring at high pacing frequency was decreased by INaL inhibition, increased by IKr inhibition, and restored by combined INaL and IKr inhibitions. CONCLUSIONS Our data demonstrate that IKr and INaL are counterbalancing currents during the physiological ventricular AP and their integrals covary in individual myocytes. Targeting these ionic currents to normalize their balance may have significant therapeutic potential in heart diseases with repolarization abnormalities. Visual Overview: A visual overview is available for this article.
Collapse
Affiliation(s)
- Bence Hegyi
- Department of Pharmacology, University of California, Davis
| | - Ye Chen-Izu
- Department of Pharmacology, University of California, Davis
- Department of Biomedical Engineering, University of California, Davis
- Department of Internal Medicine/Cardiology, University of California, Davis
| | | | - Sridharan Rajamani
- Amgen, Inc., South San Francisco, University of Debrecen, Debrecen, Hungary
| | - Luiz Belardinelli
- InCarda Therapeutics, Inc., Newark, CA, University of Debrecen, Debrecen, Hungary
| | - Donald M. Bers
- Department of Pharmacology, University of California, Davis
| | - Tamás Bányász
- Department of Pharmacology, University of California, Davis
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
10
|
Increase in CO 2 levels by upregulating late sodium current is proarrhythmic in the heart. Heart Rhythm 2019; 16:1098-1106. [PMID: 30710739 DOI: 10.1016/j.hrthm.2019.01.029] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Indexed: 12/19/2022]
Abstract
BACKGROUND Increased CO2 levels in the general circulation and/or in the myocardium are common under pathologic conditions. OBJECTIVE The purpose of this study was to test the hypothesis that an increase in CO2 levels, and not just the subsequent extra- or intracellular acidosis, would augment late sodium current (INa,L) and contribute to arrhythmogenesis in hearts with reduced repolarization reserve. METHODS Monophasic action potential durations at 90% completion of repolarization (MAPD90) from isolated rabbit hearts, INa,L, and extra- (pHo) and intracellular pH (pHi) values from cardiomyocytes using the whole-cell patch-clamp techniques and 2',7'-bis-(2-carboxyethyl)-5-(and-6)-carboxyfluorescein, acetoxymethyl ester (BCECF-AM), respectively, were measured. RESULTS Increasing CO2 levels from 5% to 10% and 20% and administration of 1 nM sea anemone toxin (ATX)-II increased INa,L and prolonged both epicardial and endocardial MAPD90 (n = 7 and 10, respectively) without causing arrhythmic activities. Compared to 5% CO2, 10% and 20% CO2 decreased pHo and pHi in hearts treated with 1 nM ATX-II, caused greater prolongation of MAPD90, and elicited ventricular tachycardias. Increasing CO2 levels from 5% to 10% and 20% with pHo maintained at 7.4 produced smaller changes in pHi (P <.05) but similar increases in INa,L, prolongation of MAPD90, and incidence of ventricular tachycardias (n = 8). Inhibition of INa,L reversed the increase in INa,L, suppressed MAPD90 prolongations, and ventricular tachycardias induced by 20% CO2. Increased phospho-calmodulin-dependent protein kinase II-δ (CaMKIIδ) and phospho-NaV1.5 protein levels in hearts treated with 20% CO2 was attenuated by eleclazine. CONCLUSION Increased CO2 levels enhance INa,L and are proarrhythmic factors in hearts with reduced repolarization reserve, possibly via mechanisms related to phosphorylation of CaMKIIδ and NaV1.5.
Collapse
|
11
|
Han D, Tan H, Sun C, Li G. Amiodarone-induced life-threatening torsade de pointes in an end-stage lung cancer patient receiving gefitinib. Oxf Med Case Reports 2019; 2019:omy122. [PMID: 30697438 PMCID: PMC6345081 DOI: 10.1093/omcr/omy122] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 10/25/2018] [Accepted: 11/07/2018] [Indexed: 12/19/2022] Open
Abstract
The risk factors of acquired long QT syndrome (aLQTS) are sometimes overlooked in clinics. Drugs, hypokalemia, age and female sex are well-known risk factors of QT prolongation-dependent torsade de pointes (TdP), which explains the high incidence of sudden cardiac death in LQT patients. Here, we report a case of an elderly female patient with lung cancer who was in poor condition, for whom amiodarone was mistakenly prescribed to rectify premature ventricular contractions. QT prolongation-dependent TdP immediately followed intravenous injection of amiodarone. Fortunately, the patient survived aborted sudden cardiac arrest after effective cardio-pulmonary resuscitation and electric defibrillation. Upon reviewing the clinical information, several pre-existing risk factors of aLQTS and TdP were identified. The mistaken prescription of amiodarone provoked TdP after these risk factors were overlooked in this case and thus predisposed this patient to a high susceptibility of drug-induced TdP.
Collapse
Affiliation(s)
- Dan Han
- Department of Cardiovascular Medicine, First Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, China
| | - Hui Tan
- Department of Respiratory Medicine, First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, China
| | - Chaofeng Sun
- Department of Cardiovascular Medicine, First Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, China
| | - Guoliang Li
- Department of Cardiovascular Medicine, First Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, China
| |
Collapse
|
12
|
Correction to: Inhibition of the K+ conductance and Cole-Moore shift of the oncogenic Kv10.1 channel by amiodarone. Pflugers Arch 2018; 470:981-993. [DOI: 10.1007/s00424-018-2131-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
13
|
Vicente J, Zusterzeel R, Johannesen L, Mason J, Sager P, Patel V, Matta MK, Li Z, Liu J, Garnett C, Stockbridge N, Zineh I, Strauss DG. Mechanistic Model-Informed Proarrhythmic Risk Assessment of Drugs: Review of the "CiPA" Initiative and Design of a Prospective Clinical Validation Study. Clin Pharmacol Ther 2018; 103:54-66. [PMID: 28986934 DOI: 10.1002/cpt.v103.110.1002/cpt.896] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 09/20/2017] [Accepted: 10/01/2017] [Indexed: 05/26/2023]
Abstract
The Comprehensive in vitro Proarrhythmia Assay (CiPA) initiative is developing and validating a mechanistic-based assessment of the proarrhythmic risk of drugs. CiPA proposes to assess a drug's effect on multiple ion channels and integrate the effects in a computer model of the human cardiomyocyte to predict proarrhythmic risk. Unanticipated or missed effects will be assessed with human stem cell-derived cardiomyocytes and electrocardiogram (ECG) analysis in early phase I clinical trials. This article provides an overview of CiPA and the rationale and design of the CiPA phase I ECG validation clinical trial, which involves assessing an additional ECG biomarker (J-Tpeak) for QT prolonging drugs. If successful, CiPA will 1) create a pathway for drugs with hERG block / QT prolongation to advance without intensive ECG monitoring in phase III trials if they have low proarrhythmic risk; and 2) enable updating drug labels to be more informative about proarrhythmic risk, not just QT prolongation.
Collapse
Affiliation(s)
- Jose Vicente
- Office of New Drugs, Center for Drug Evaluation and Research, United States Food and Drug Administration, Silver Spring, Maryland, USA
| | - Robbert Zusterzeel
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, United States Food and Drug Administration, Silver Spring, Maryland, USA
| | - Lars Johannesen
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, United States Food and Drug Administration, Silver Spring, Maryland, USA
| | - Jay Mason
- Department of Medicine, Division of Cardiology, University of Utah, Salt Lake City, Utah, USA
- Spaulding Clinical Research, West Bend, Wisconsin, USA
| | | | - Vikram Patel
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, United States Food and Drug Administration, Silver Spring, Maryland, USA
| | - Murali K Matta
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, United States Food and Drug Administration, Silver Spring, Maryland, USA
| | - Zhihua Li
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, United States Food and Drug Administration, Silver Spring, Maryland, USA
| | - Jiang Liu
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, United States Food and Drug Administration, Silver Spring, Maryland, USA
| | - Christine Garnett
- Office of New Drugs, Center for Drug Evaluation and Research, United States Food and Drug Administration, Silver Spring, Maryland, USA
| | - Norman Stockbridge
- Office of New Drugs, Center for Drug Evaluation and Research, United States Food and Drug Administration, Silver Spring, Maryland, USA
| | - Issam Zineh
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, United States Food and Drug Administration, Silver Spring, Maryland, USA
| | - David G Strauss
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, United States Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|
14
|
Vicente J, Zusterzeel R, Johannesen L, Mason J, Sager P, Patel V, Matta MK, Li Z, Liu J, Garnett C, Stockbridge N, Zineh I, Strauss DG. Mechanistic Model-Informed Proarrhythmic Risk Assessment of Drugs: Review of the "CiPA" Initiative and Design of a Prospective Clinical Validation Study. Clin Pharmacol Ther 2018; 103:54-66. [PMID: 28986934 PMCID: PMC5765372 DOI: 10.1002/cpt.896] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 09/20/2017] [Accepted: 10/01/2017] [Indexed: 12/19/2022]
Abstract
The Comprehensive in vitro Proarrhythmia Assay (CiPA) initiative is developing and validating a mechanistic-based assessment of the proarrhythmic risk of drugs. CiPA proposes to assess a drug's effect on multiple ion channels and integrate the effects in a computer model of the human cardiomyocyte to predict proarrhythmic risk. Unanticipated or missed effects will be assessed with human stem cell-derived cardiomyocytes and electrocardiogram (ECG) analysis in early phase I clinical trials. This article provides an overview of CiPA and the rationale and design of the CiPA phase I ECG validation clinical trial, which involves assessing an additional ECG biomarker (J-Tpeak) for QT prolonging drugs. If successful, CiPA will 1) create a pathway for drugs with hERG block / QT prolongation to advance without intensive ECG monitoring in phase III trials if they have low proarrhythmic risk; and 2) enable updating drug labels to be more informative about proarrhythmic risk, not just QT prolongation.
Collapse
Affiliation(s)
- Jose Vicente
- Office of New Drugs, Center for Drug Evaluation and ResearchUnited States Food and Drug AdministrationSilver SpringMarylandUSA
| | - Robbert Zusterzeel
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and ResearchUnited States Food and Drug AdministrationSilver SpringMarylandUSA
| | - Lars Johannesen
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and ResearchUnited States Food and Drug AdministrationSilver SpringMarylandUSA
| | - Jay Mason
- Department of Medicine, Division of CardiologyUniversity of UtahSalt Lake CityUtahUSA
- Spaulding Clinical ResearchWest BendWisconsinUSA
| | | | - Vikram Patel
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and ResearchUnited States Food and Drug AdministrationSilver SpringMarylandUSA
| | - Murali K. Matta
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and ResearchUnited States Food and Drug AdministrationSilver SpringMarylandUSA
| | - Zhihua Li
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and ResearchUnited States Food and Drug AdministrationSilver SpringMarylandUSA
| | - Jiang Liu
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and ResearchUnited States Food and Drug AdministrationSilver SpringMarylandUSA
| | - Christine Garnett
- Office of New Drugs, Center for Drug Evaluation and ResearchUnited States Food and Drug AdministrationSilver SpringMarylandUSA
| | - Norman Stockbridge
- Office of New Drugs, Center for Drug Evaluation and ResearchUnited States Food and Drug AdministrationSilver SpringMarylandUSA
| | - Issam Zineh
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and ResearchUnited States Food and Drug AdministrationSilver SpringMarylandUSA
| | - David G. Strauss
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and ResearchUnited States Food and Drug AdministrationSilver SpringMarylandUSA
| |
Collapse
|
15
|
Inhibition of the K+ conductance and Cole-Moore shift of the oncogenic Kv10.1 channel by amiodarone. Pflugers Arch 2017; 470:491-503. [DOI: 10.1007/s00424-017-2092-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 11/06/2017] [Accepted: 11/24/2017] [Indexed: 12/31/2022]
|
16
|
Late sodium current associated cardiac electrophysiological and mechanical dysfunction. Pflugers Arch 2017; 470:461-469. [DOI: 10.1007/s00424-017-2079-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 09/27/2017] [Accepted: 10/09/2017] [Indexed: 12/19/2022]
|
17
|
|
18
|
Chotsiri P, Wattanakul T, Hoglund RM, Hanboonkunupakarn B, Pukrittayakamee S, Blessborn D, Jittamala P, White NJ, Day NPJ, Tarning J. Population pharmacokinetics and electrocardiographic effects of dihydroartemisinin-piperaquine in healthy volunteers. Br J Clin Pharmacol 2017; 83:2752-2766. [PMID: 28695570 PMCID: PMC5698590 DOI: 10.1111/bcp.13372] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 06/21/2017] [Accepted: 06/30/2017] [Indexed: 12/21/2022] Open
Abstract
Aims The aims of the present study were to evaluate the pharmacokinetic properties of dihydroartemisinin (DHA) and piperaquine, potential drug–drug interactions with concomitant primaquine treatment, and piperaquine effects on the electrocardiogram in healthy volunteers. Methods The population pharmacokinetic properties of DHA and piperaquine were assessed in 16 healthy Thai adults using an open‐label, randomized, crossover study. Drug concentration–time data and electrocardiographic measurements were evaluated with nonlinear mixed‐effects modelling. Results The developed models described DHA and piperaquine population pharmacokinetics accurately. Concomitant treatment with primaquine did not affect the pharmacokinetic properties of DHA or piperaquine. A linear pharmacokinetic–pharmacodynamic model described satisfactorily the relationship between the individually corrected QT intervals and piperaquine concentrations; the population mean QT interval increased by 4.17 ms per 100 ng ml–1 increase in piperaquine plasma concentration. Simulations from the final model showed that monthly and bimonthly mass drug administration in healthy subjects would result in median maximum QT interval prolongations of 18.9 ms and 16.8 ms, respectively, and would be very unlikely to result in prolongation of more than 50 ms. A single low dose of primaquine can be added safely to the existing DHA–piperaquine treatment in areas of multiresistant Plasmodium falciparum malaria. Conclusions Pharmacokinetic–pharmacodynamic modelling and simulation in healthy adult volunteers suggested that therapeutic doses of DHA–piperaquine in the prevention or treatment of P. falciparum malaria are unlikely to be associated with dangerous QT prolongation.
Collapse
Affiliation(s)
- Palang Chotsiri
- Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand.,Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Thanaporn Wattanakul
- Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Richard M Hoglund
- Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | | | | | - Daniel Blessborn
- Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | | | - Nicholas J White
- Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Nicholas P J Day
- Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Joel Tarning
- Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
19
|
Nánási PP, Magyar J, Varró A, Ördög B. Beat-to-beat variability of cardiac action potential duration: underlying mechanism and clinical implications. Can J Physiol Pharmacol 2017; 95:1230-1235. [PMID: 28746810 DOI: 10.1139/cjpp-2016-0597] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Beat-to-beat variability of cardiac action potential duration (short-term variability, SV) is a common feature of various cardiac preparations, including the human heart. Although it is believed to be one of the best arrhythmia predictors, the underlying mechanisms are not fully understood at present. The magnitude of SV is basically determined by the intensity of cell-to-cell coupling in multicellular preparations and by the duration of the action potential (APD). To compensate for the APD-dependent nature of SV, the concept of relative SV (RSV) has been introduced by normalizing the changes of SV to the concomitant changes in APD. RSV is reduced by ICa, IKr, and IKs while increased by INa, suggesting that ion currents involved in the negative feedback regulation of APD tend to keep RSV at a low level. RSV is also influenced by intracellular calcium concentration and tissue redox potential. The clinical implications of APD variability is discussed in detail.
Collapse
Affiliation(s)
- Péter P Nánási
- a Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,b Department of Dental Physiology and Pharmacology, Faculty of Dentistry, University of Debrecen, Debrecen, Hungary
| | - János Magyar
- a Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - András Varró
- c Department of Pharmacology and Pharmacotherapy, University of Szeged, Szeged, Hungary
| | - Balázs Ördög
- c Department of Pharmacology and Pharmacotherapy, University of Szeged, Szeged, Hungary
| |
Collapse
|
20
|
Yokoyama H, Nakamura Y, Saito H, Nagayama Y, Hoshiai K, Wada T, Izumi-Nakaseko H, Ando K, Akie Y, Sugiyama A. Pharmacological characterization of microminipig as a model to assess the drug-induced cardiovascular responses for non-clinical toxicity and/or safety pharmacology studies. J Toxicol Sci 2017; 42:93-101. [PMID: 28070113 DOI: 10.2131/jts.42.93] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
We tried to establish the halothane-anesthetized microminipigs as an alternative animal model for non-clinical toxicity and/or safety pharmacology studies. In order to characterize the halothane-anesthetized microminipigs, we firstly clarified the effects of halothane anesthesia on their cardiovascular system (n = 5). Then, we examined the cardiovascular effects of dl-sotalol in doses of 0.1, 0.3 and 1 mg/kg, i.v. on the halothane-anesthetized microminipigs (n = 6). Induction of the halothane anesthesia by itself prolonged the QT interval as well as QTcF, suggesting that the halothane anesthesia can reduce the cardiac repolarization reserve in microminipigs like in dogs. dl-Sotalol showed more potent negative chronotropic, dromotropic and hypotensive effects together with repolarization delay in microminipigs than in dogs, although each cardiovascular response to dl-sotalol was directionally similar between them, suggesting greater basal sympathetic tone and/or smaller volume of distribution of the drug in microminipigs than in dogs. Analyses of proarrhythmic surrogate markers indicate that Tpeak-Tend and short-term variability of QT interval may be more sensitive to detect the dl-sotalol-induced direct electrophysiological changes in microminipigs than in dogs, but its reverse will be true for J-Tpeakc. Thus, these results may help better understand the drug-induced cardiovascular responses in microminipigs.
Collapse
|
21
|
[Ventricular tachyarrhythmia as a side effect of pharmacotherapy]. Herzschrittmacherther Elektrophysiol 2017; 28:162-168. [PMID: 28488108 DOI: 10.1007/s00399-017-0500-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 04/10/2017] [Indexed: 12/19/2022]
Abstract
Ventricular tachyarrhythmia is a severe and life-threatening potential side effect of pharmacotherapy. Substances with proarrhythmic potential belong to various groups of medication. Apart from antiarrhythmic agents, especially antibiotics and psychiatric drugs are worth mentioning owing to their broad application. Interaction with cardiac potassium channels is the most important reason for drug-induced ventricular tachyarrhythmia. Over 20 years of research in animal models and clinical studies have uncovered the underlying mechanisms. Findings in this field of research have also made a contribution to the understanding of genetic long QT syndromes. Clinical concerns that take drug interactions into account have been neglected due to the mechanistic research approach. For daily clinical practice, combination therapy of several potentially arrhythmogenic drugs is of predominant concern especially in situations when the therapeutic regime is changing such as admission to the hospital, admission to an intensive care unit or consultation of a new specialist. Especially in these situations, considerations about the arrhythmogenic potential of additionally administered drugs should be paid explicit attention. Additional concern should be paid to the fact that several proarrhythmogenic agents are metabolized over single pathways and are therefore prone to drug interactions that can severely raise the drug concentration and as a result arrhythmogenic potential.
Collapse
|
22
|
Wei XH, Yu SD, Ren L, Huang SH, Yang QM, Wang P, Chu YP, Yang W, Ding YS, Huo Y, Wu L. Inhibition of late sodium current suppresses calcium-related ventricular arrhythmias by reducing the phosphorylation of CaMK-II and sodium channel expressions. Sci Rep 2017; 7:981. [PMID: 28428622 PMCID: PMC5430524 DOI: 10.1038/s41598-017-01056-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 03/20/2017] [Indexed: 12/19/2022] Open
Abstract
Cardiac arrhythmias associated with intracellular calcium inhomeostasis are refractory to antiarrhythmic therapy. We hypothesized that late sodium current (I Na) contributed to the calcium-related arrhythmias. Monophasic action potential duration at 90% completion of repolarization (MAPD90) was significantly increased and ventricular arrhythmias were observed in hearts with increased intracellular calcium concentration ([Ca2+]i) by using Bay K 8644, and the increase became greater in hearts treated with a combination of ATX-II and Bay K 8644 compared to Bay K 8644 alone. The prolongations caused by Bay K 8644 and frequent episodes of ventricular tachycardias, both in absence and presence of ATX-II, were significantly attenuated or abolished by late I Na inhibitors TTX and eleclazine. In rabbit ventricular myocytes, Bay K 8644 increased I CaL density, calcium transient and myocyte contraction. TTX and eleclazine decreased the amplitude of late I Na, the reverse use dependence of MAPD90 at slower heart rate, and attenuated the increase of intracellular calcium transient and myocyte contraction. TTX diminished the phosphorylation of CaMKII-δ and Nav 1.5 in hearts treated with Bay K 8644 and ATX-II. In conclusion, late I Na contributes to ventricular arrhythmias and its inhibition is plausible to treat arrhythmias in hearts with increased [Ca2+]i.
Collapse
Affiliation(s)
- Xiao-Hong Wei
- Department of Cardiology, Peking University First Hospital, Beijing, 100034, China.,Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, 100191, China
| | - Shan-Dong Yu
- Department of Cardiology, Peking University First Hospital, Beijing, 100034, China.,Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, 100191, China
| | - Lu Ren
- Department of Cardiology, Peking University First Hospital, Beijing, 100034, China.,Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, 100191, China
| | - Si-Hui Huang
- Department of Cardiology, Peking University First Hospital, Beijing, 100034, China.,Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, 100191, China
| | - Qiao-Mei Yang
- Department of Cardiology, Peking University First Hospital, Beijing, 100034, China.,Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, 100191, China
| | - Ping Wang
- Department of Cardiology, Peking University First Hospital, Beijing, 100034, China
| | - Yan-Peng Chu
- Department of Cardiology, Peking University First Hospital, Beijing, 100034, China.,Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, 100191, China
| | - Wei Yang
- Department of Cardiology, Peking University First Hospital, Beijing, 100034, China.,Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, 100191, China
| | - Yan-Sheng Ding
- Department of Cardiology, Peking University First Hospital, Beijing, 100034, China.,Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, 100191, China
| | - Yong Huo
- Department of Cardiology, Peking University First Hospital, Beijing, 100034, China. .,Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, 100191, China.
| | - Lin Wu
- Department of Cardiology, Peking University First Hospital, Beijing, 100034, China. .,Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, 100191, China.
| |
Collapse
|
23
|
Johannesen L, Vicente J, Hosseini M, Strauss DG. Automated Algorithm for J-Tpeak and Tpeak-Tend Assessment of Drug-Induced Proarrhythmia Risk. PLoS One 2016; 11:e0166925. [PMID: 28036330 PMCID: PMC5201230 DOI: 10.1371/journal.pone.0166925] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 11/07/2016] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Prolongation of the heart rate corrected QT (QTc) interval is a sensitive marker of torsade de pointes risk; however it is not specific as QTc prolonging drugs that block inward currents are often not associated with torsade. Recent work demonstrated that separate analysis of the heart rate corrected J-Tpeakc (J-Tpeakc) and Tpeak-Tend intervals can identify QTc prolonging drugs with inward current block and is being proposed as a part of a new cardiac safety paradigm for new drugs (the "CiPA" initiative). METHODS In this work, we describe an automated measurement methodology for assessment of the J-Tpeakc and Tpeak-Tend intervals using the vector magnitude lead. The automated measurement methodology was developed using data from one clinical trial and was evaluated using independent data from a second clinical trial. RESULTS Comparison between the automated and the prior semi-automated measurements shows that the automated algorithm reproduces the semi-automated measurements with a mean difference of single-deltas <1 ms and no difference in intra-time point variability (p for all > 0.39). In addition, the time-profile of the baseline and placebo-adjusted changes are within 1 ms for 63% of the time-points (86% within 2 ms). Importantly, the automated results lead to the same conclusions about the electrophysiological mechanisms of the studied drugs. CONCLUSIONS We have developed an automated algorithm for assessment of J-Tpeakc and Tpeak-Tend intervals that can be applied in clinical drug trials. Under the CiPA initiative this ECG assessment would determine if there are unexpected ion channel effects in humans compared to preclinical studies. The algorithm is being released as open-source software. TRIAL REGISTRATION NCT02308748 and NCT01873950.
Collapse
Affiliation(s)
- Lars Johannesen
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, United States of America
| | - Jose Vicente
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, United States of America
- BSICoS Group, Aragón Institute for Engineering Research (I3A), IIS Aragón, University of Zaragoza, Zaragoza, Spain
| | - Meisam Hosseini
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, United States of America
| | - David G. Strauss
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, United States of America
| |
Collapse
|
24
|
Vicente J, Johannesen L, Hosseini M, Mason JW, Sager PT, Pueyo E, Strauss DG. Electrocardiographic Biomarkers for Detection of Drug-Induced Late Sodium Current Block. PLoS One 2016; 11:e0163619. [PMID: 28036334 PMCID: PMC5201270 DOI: 10.1371/journal.pone.0163619] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 09/12/2016] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Drugs that prolong the heart rate corrected QT interval (QTc) on the electrocardiogram (ECG) by blocking the hERG potassium channel and also block inward currents (late sodium or L-type calcium) are not associated with torsade de pointes (e.g. ranolazine and verapamil). Thus, identifying ECG signs of late sodium current block could aid in the determination of proarrhythmic risk for new drugs. A new cardiac safety paradigm for drug development (the "CiPA" initiative) will involve the preclinical assessment of multiple human cardiac ion channels and ECG biomarkers are needed to determine if there are unexpected ion channel effects in humans. METHODS AND RESULTS In this study we assess the ability of eight ECG morphology biomarkers to detect late sodium current block in the presence of QTc prolongation by analyzing a clinical trial where a selective hERG potassium channel blocker (dofetilide) was administered alone and then in combination with two late sodium current blockers (lidocaine and mexiletine). We demonstrate that late sodium current block has the greatest effect on the heart-rate corrected J-Tpeak interval (J-Tpeakc), followed by QTc and then T-wave flatness. Furthermore, J-Tpeakc is the only biomarker that improves detection of the presence of late sodium current block compared to using QTc alone (AUC: 0.83 vs. 0.72 respectively, p<0.001). CONCLUSIONS Analysis of the J-Tpeakc interval can differentiate drug-induced multichannel block involving the late sodium current from selective hERG potassium channel block. Future methodologies assessing drug effects on cardiac ion channel currents on the ECG should use J-Tpeakc to detect the presence of late sodium current block. TRIAL REGISTRATION NCT02308748 and NCT01873950.
Collapse
Affiliation(s)
- Jose Vicente
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, United States of America
- Division of Biomedical Physics, Office of Science and Engineering Laboratories, Center for Devices and Radiological Health, US Food and Drug Administration, Silver Spring, MD, United States of America
- BSICoS Group, Aragón Institute for Engineering Research (I3A), IIS Aragón, University of Zaragoza, Zaragoza, Spain
| | - Lars Johannesen
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, United States of America
| | - Meisam Hosseini
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, United States of America
- Division of Biomedical Physics, Office of Science and Engineering Laboratories, Center for Devices and Radiological Health, US Food and Drug Administration, Silver Spring, MD, United States of America
| | - Jay W. Mason
- Cardiology Division, University of Utah, Salt Lake City, UT, United States of America
- Spaulding Clinical Research, West Bend, WI, United States of America
| | - Philip T. Sager
- Stanford University, Palo Alto, CA, United States of America
| | - Esther Pueyo
- BSICoS Group, Aragón Institute for Engineering Research (I3A), IIS Aragón, University of Zaragoza, Zaragoza, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Zaragoza, Spain
| | - David G. Strauss
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, United States of America
| |
Collapse
|
25
|
Vicente J, Stockbridge N, Strauss DG. Evolving regulatory paradigm for proarrhythmic risk assessment for new drugs. J Electrocardiol 2016; 49:837-842. [PMID: 27524478 DOI: 10.1016/j.jelectrocard.2016.07.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Indexed: 11/16/2022]
Abstract
Fourteen drugs were removed from the market worldwide because their potential to cause torsade de pointes (torsade), a potentially fatal ventricular arrhythmia. The observation that most drugs that cause torsade block the potassium channel encoded by the human ether-à-go-go related gene (hERG) and prolong the heart rate corrected QT interval (QTc) on the ECG, led to a focus on screening new drugs for their potential to block the hERG potassium channel and prolong QTc. This has been a successful strategy keeping torsadogenic drugs off the market, but has resulted in drugs being dropped from development, sometimes inappropriately. This is because not all drugs that block the hERG potassium channel and prolong QTc cause torsade, sometimes because they block other channels. The regulatory paradigm is evolving to improve proarrhythmic risk prediction. ECG studies can now use exposure-response modeling for assessing the effect of a drug on the QTc in small sample size first-in-human studies. Furthermore, the Comprehensive in vitro Proarrhythmia Assay (CiPA) initiative is developing and validating a new in vitro paradigm for cardiac safety evaluation of new drugs that provides a more accurate and comprehensive mechanistic-based assessment of proarrhythmic potential. Under CiPA, the prediction of proarrhythmic potential will come from in vitro ion channel assessments coupled with an in silico model of the human ventricular myocyte. The preclinical assessment will be checked with an assessment of human phase 1 ECG data to determine if there are unexpected ion channel effects in humans compared to preclinical ion channel data. While there is ongoing validation work, the heart rate corrected J-Tpeak interval is likely to be assessed under CiPA to detect inward current block in presence of hERG potassium channel block.
Collapse
Affiliation(s)
- Jose Vicente
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, MD, USA.
| | - Norman Stockbridge
- Division of Cardiovascular and Renal Products, Office of New Drugs, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - David G Strauss
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, MD, USA
| |
Collapse
|
26
|
Ghovanloo MR, Aimar K, Ghadiry-Tavi R, Yu A, Ruben PC. Physiology and Pathophysiology of Sodium Channel Inactivation. CURRENT TOPICS IN MEMBRANES 2016; 78:479-509. [PMID: 27586293 DOI: 10.1016/bs.ctm.2016.04.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Voltage-gated sodium channels are present in different tissues within the human body, predominantly nerve, muscle, and heart. The sodium channel is composed of four similar domains, each containing six transmembrane segments. Each domain can be functionally organized into a voltage-sensing region and a pore region. The sodium channel may exist in resting, activated, fast inactivated, or slow inactivated states. Upon depolarization, when the channel opens, the fast inactivation gate is in its open state. Within the time frame of milliseconds, this gate closes and blocks the channel pore from conducting any more sodium ions. Repetitive or continuous stimulations of sodium channels result in a rate-dependent decrease of sodium current. This process may continue until the channel fully shuts down. This collapse is known as slow inactivation. This chapter reviews what is known to date regarding, sodium channel inactivation with a focus on various mutations within each NaV subtype and with clinical implications.
Collapse
Affiliation(s)
- M-R Ghovanloo
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - K Aimar
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - R Ghadiry-Tavi
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - A Yu
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - P C Ruben
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| |
Collapse
|
27
|
Matsukura S, Nakamura Y, Cao X, Wada T, Izumi-Nakaseko H, Ando K, Sugiyama A. Anti-atrial Fibrillatory Versus Proarrhythmic Potentials of Amiodarone: A New Protocol for Safety Evaluation In Vivo. Cardiovasc Toxicol 2016; 17:157-162. [DOI: 10.1007/s12012-016-9369-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
28
|
Ghovanloo MR, Abdelsayed M, Ruben PC. Effects of Amiodarone and N-desethylamiodarone on Cardiac Voltage-Gated Sodium Channels. Front Pharmacol 2016; 7:39. [PMID: 26973526 PMCID: PMC4771766 DOI: 10.3389/fphar.2016.00039] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2015] [Accepted: 02/12/2016] [Indexed: 12/19/2022] Open
Abstract
Amiodarone (AMD) is a potent antiarrhythmic drug with high efficacy for treating atrial fibrillation and tachycardia. The pharmacologic profile of AMD is complex. AMD possesses biophysical characteristics of all of class I, II, III, and IV agents. Despite its adverse side effects, AMD remains the most commonly prescribed antiarrhythmic drug. AMD was described to prolong the QT interval and can lead to torsades de pointes. Our goal was to study the effects of AMD on peak and late sodium currents (INa,P and INa,L) and determine whether these effects change as AMD is metabolized into N-desethylamiodarone (DES). We hypothesized that AMD and DES block both INa,P and INa,L with similar profiles due to structural similarities. Given the inherent small amounts of INa,L in NaV1.5, we screened AMD and DES against the Long QT-3-causing mutation, ΔKPQ, to better detect any drug-mediated effect on INa,L. Our results show that AMD and DES do not affect WT or ΔKPQ activation; however, both drugs altered the apparent valence of steady-state fast-inactivation. In addition, AMD and DES preferentially block ΔKPQ peak conductance compared to WT. Both compounds significantly increase INa,L and window currents. We conclude that both compounds have pro-arrhythmic effects on NaV1.5, especially ΔKPQ; however, DES seems to have a greater pro-arrhythmic effect than AMD.
Collapse
Affiliation(s)
- Mohammad-Reza Ghovanloo
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby BC, Canada
| | - Mena Abdelsayed
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby BC, Canada
| | - Peter C Ruben
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby BC, Canada
| |
Collapse
|
29
|
Johannesen L, Vicente J, Mason JW, Erato C, Sanabria C, Waite-Labott K, Hong M, Lin J, Guo P, Mutlib A, Wang J, Crumb WJ, Blinova K, Chan D, Stohlman J, Florian J, Ugander M, Stockbridge N, Strauss DG. Late sodium current block for drug-induced long QT syndrome: Results from a prospective clinical trial. Clin Pharmacol Ther 2016; 99:214-23. [PMID: 26259627 PMCID: PMC5421403 DOI: 10.1002/cpt.205] [Citation(s) in RCA: 115] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 08/05/2015] [Indexed: 12/19/2022]
Abstract
Drug-induced long QT syndrome has resulted in many drugs being withdrawn from the market. At the same time, the current regulatory paradigm for screening new drugs causing long QT syndrome is preventing drugs from reaching the market, sometimes inappropriately. In this study, we report the results of a first-of-a-kind clinical trial studying late sodium (mexiletine and lidocaine) and calcium (diltiazem) current blocking drugs to counteract the effects of hERG potassium channel blocking drugs (dofetilide and moxifloxacin). We demonstrate that both mexiletine and lidocaine substantially reduce heart-rate corrected QT (QTc) prolongation from dofetilide by 20 ms. Furthermore, all QTc shortening occurs in the heart-rate corrected J-Tpeak (J-Tpeak c) interval, the biomarker we identified as a sign of late sodium current block. This clinical trial demonstrates that late sodium blocking drugs can substantially reduce QTc prolongation from hERG potassium channel block and assessment of J-Tpeak c may add value beyond only assessing QTc.
Collapse
Affiliation(s)
- L Johannesen
- Center for Devices and Radiological Health, US Food and Drug Administration,
Silver Spring, Maryland, USA
- Department of Clinical Physiology, Karolinska Institutet and Karolinska
University Hospital, Stockholm, Sweden
| | - J Vicente
- Center for Devices and Radiological Health, US Food and Drug Administration,
Silver Spring, Maryland, USA
- Center for Drug Evaluation and Research, US Food and Drug Administration,
Silver Spring, Maryland, USA
- BSICoS Group, Aragón Institute for Engineering Research (I3A), IIS
Aragón, University of Zaragoza, Zaragoza, Spain
| | - JW Mason
- Spaulding Clinical, West Bend, Wisconsin, USA
- University of Utah, Salt Lake City, Utah, USA
| | - C Erato
- Spaulding Clinical, West Bend, Wisconsin, USA
| | - C Sanabria
- Spaulding Clinical, West Bend, Wisconsin, USA
| | | | - M Hong
- Frontage Laboratories, Exton, Pennsylvania, USA
| | - J Lin
- Frontage Laboratories, Exton, Pennsylvania, USA
| | - P Guo
- Frontage Laboratories, Exton, Pennsylvania, USA
| | - A Mutlib
- Frontage Laboratories, Exton, Pennsylvania, USA
| | - J Wang
- Frontage Laboratories, Exton, Pennsylvania, USA
| | - WJ Crumb
- Zenas Technologies, Metairie, Louisiana, USA
| | - K Blinova
- Center for Devices and Radiological Health, US Food and Drug Administration,
Silver Spring, Maryland, USA
| | - D Chan
- Center for Devices and Radiological Health, US Food and Drug Administration,
Silver Spring, Maryland, USA
| | - J Stohlman
- Center for Devices and Radiological Health, US Food and Drug Administration,
Silver Spring, Maryland, USA
| | - J Florian
- Center for Drug Evaluation and Research, US Food and Drug Administration,
Silver Spring, Maryland, USA
| | - M Ugander
- Center for Devices and Radiological Health, US Food and Drug Administration,
Silver Spring, Maryland, USA
- Department of Clinical Physiology, Karolinska Institutet and Karolinska
University Hospital, Stockholm, Sweden
| | - N Stockbridge
- Center for Drug Evaluation and Research, US Food and Drug Administration,
Silver Spring, Maryland, USA
| | - DG Strauss
- Center for Devices and Radiological Health, US Food and Drug Administration,
Silver Spring, Maryland, USA
- Department of Clinical Physiology, Karolinska Institutet and Karolinska
University Hospital, Stockholm, Sweden
| |
Collapse
|
30
|
Ferber G, Johannesen L. A Comparison of Methods for Thorough QT Analysis for the Assessment of Cardiac Safety. Pharmaceut Med 2016. [DOI: 10.1007/s40290-015-0123-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
31
|
Yuan Y, Bai X, Luo C, Wang K, Zhang H. The virtual heart as a platform for screening drug cardiotoxicity. Br J Pharmacol 2015; 172:5531-5547. [PMID: 25363597 PMCID: PMC4667856 DOI: 10.1111/bph.12996] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Revised: 10/23/2014] [Accepted: 10/28/2014] [Indexed: 01/01/2023] Open
Abstract
To predict the safety of a drug at an early stage in its development is a major challenge as there is a lack of in vitro heart models that correlate data from preclinical toxicity screening assays with clinical results. A biophysically detailed computer model of the heart, the virtual heart, provides a powerful tool for simulating drug-ion channel interactions and cardiac functions during normal and disease conditions and, therefore, provides a powerful platform for drug cardiotoxicity screening. In this article, we first review recent progress in the development of theory on drug-ion channel interactions and mathematical modelling. Then we propose a family of biomarkers that can quantitatively characterize the actions of a drug on the electrical activity of the heart at multi-physical scales including cellular and tissue levels. We also conducted some simulations to demonstrate the application of the virtual heart to assess the pro-arrhythmic effects of cisapride and amiodarone. Using the model we investigated the mechanisms responsible for the differences between the two drugs on pro-arrhythmogenesis, even though both prolong the QT interval of ECGs. Several challenges for further development of a virtual heart as a platform for screening drug cardiotoxicity are discussed.
Collapse
Affiliation(s)
- Yongfeng Yuan
- School of Computer Science and TechnologyHarbin Institute of TechnologyHarbinChina
| | - Xiangyun Bai
- School of Computer Science and TechnologyHarbin Institute of TechnologyHarbinChina
| | - Cunjin Luo
- School of Computer Science and TechnologyHarbin Institute of TechnologyHarbinChina
| | - Kuanquan Wang
- School of Computer Science and TechnologyHarbin Institute of TechnologyHarbinChina
| | - Henggui Zhang
- School of Computer Science and TechnologyHarbin Institute of TechnologyHarbinChina
- Biological Physics GroupSchool of Physics and AstronomyThe University of ManchesterManchesterUK
| |
Collapse
|
32
|
Obejero-Paz CA, Bruening-Wright A, Kramer J, Hawryluk P, Tatalovic M, Dittrich HC, Brown AM. Quantitative Profiling of the Effects of Vanoxerine on Human Cardiac Ion Channels and its Application to Cardiac Risk. Sci Rep 2015; 5:17623. [PMID: 26616666 PMCID: PMC4663487 DOI: 10.1038/srep17623] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 11/03/2015] [Indexed: 12/19/2022] Open
Abstract
Vanoxerine has been in clinical trials for Parkinsonism, depression and cocaine addiction but lacked efficacy. Although a potent blocker of hERG, it produced no serious adverse events. We attributed the unexpected result to offsetting Multiple Ion Channel Effects (MICE). Vanoxerine’s effects were strongly frequency-dependent and we repositioned it for treatment of atrial fibrillation and flutter. Vanoxerine terminated AF/AFL in an animal model and a dose-ranging clinical trial. Reversion to normal rhythm was associated with QT prolongation yet absent proarrhythmia markers for Torsade de Pointes (TdP). To understand the QT/TdP discordance, we used quantitative profiling and compared vanoxerine with dofetilide, a selective hERG-blocking torsadogen used for intractable AF, verapamil, a non-torsadogenic MICE comparator and bepridil, a torsadogenic MICE comparator. At clinically relevant concentrations, verapamil blocked hCav1.2 and hERG, as did vanoxerine and bepridil both of which also blocked hNav1.5. In acute experiments and simulations, dofetilide produced early after depolarizations (EADs) and arrhythmias, whereas verapamil, vanoxerine and bepridil produced no proarrhythmia markers. Of the MICE drugs only bepridil inhibited hERG trafficking following overnight exposure. The results are consistent with the emphasis on MICE of the CiPA assay. Additionally we propose that trafficking inhibition of hERG be added to CiPA.
Collapse
Affiliation(s)
- Carlos A Obejero-Paz
- ChanTest Corporation, a Charles River Company, Discovery Services, 14656 Neo Parkway, Cleveland, OH 44128, USA
| | - Andrew Bruening-Wright
- ChanTest Corporation, a Charles River Company, Discovery Services, 14656 Neo Parkway, Cleveland, OH 44128, USA
| | - James Kramer
- ChanTest Corporation, a Charles River Company, Discovery Services, 14656 Neo Parkway, Cleveland, OH 44128, USA
| | - Peter Hawryluk
- ChanTest Corporation, a Charles River Company, Discovery Services, 14656 Neo Parkway, Cleveland, OH 44128, USA
| | - Milos Tatalovic
- ChanTest Corporation, a Charles River Company, Discovery Services, 14656 Neo Parkway, Cleveland, OH 44128, USA
| | - Howard C Dittrich
- Laguna Pharmaceuticals, 4225 Executive Square, Suite 960, La Jolla, CA 92037, USA
| | - Arthur M Brown
- ChanTest Corporation, a Charles River Company, Discovery Services, 14656 Neo Parkway, Cleveland, OH 44128, USA.,Laguna Pharmaceuticals, 4225 Executive Square, Suite 960, La Jolla, CA 92037, USA
| |
Collapse
|
33
|
Zhang X, Beebe T, Jen N, Lee CA, Tai Y, Hsiai TK. Flexible and waterproof micro-sensors to uncover zebrafish circadian rhythms: The next generation of cardiac monitoring for drug screening. Biosens Bioelectron 2015; 71:150-157. [PMID: 25909335 DOI: 10.1016/j.bios.2015.04.027] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 03/30/2015] [Accepted: 04/12/2015] [Indexed: 01/01/2023]
Abstract
Flexible electronics are the next generation of sensors for mobile health and implantation. Zebrafish (Danio rerio) is an emergent strategy for pre-clinical drug development and toxicity testing. To address the confounding effects from sedation of fish and removal from the aquatic habitat for micro-electrocardiogram (µECG) measurements, we developed waterproof and wearable sensors to uncover the circadian variation in heart rate (HR) and heart rate variability (HRV) (Massin et al., 2000). The parylene-C based ECG sensor consisted of an ultra-soft silicone integrated jacket designed to wrap around the fish during swimming. The Young's modulus of this silicone jacket matched with the fish surface, and an extended parylene cable connected the underwater chest electrodes with the out-of water electronics. In addition, embedded micro-glass spheres in the silicone effectively reduced the effective density of the jacket to ~1 g cm(-3). These innovations enabled physiological ECG telemetry in the fish's natural habitat without the need for sedation. Furthermore, a set of non-linear signal processing techniques filtered out the breathing and electromagnetic artifacts from the recorded signals. We observed a reduction in mean HR and an increase in HRV over 24h at 10 dpa, accompanied by QT prolongation as well as diurnal variations, followed by normalization in mean HR and QT intervals at 26 days post ventricular amputation (dpa). We revealed Amiodarone-mediated QTc prolongation, HR reduction and HRV increase otherwise masked by sedation. The novel features of the flexible silicon jacket for µECG telemetry unraveled the biological clock and normalization of QT intervals at 26 dpa, providing the first evidence of new physiological phenomena during cardiac injury and repair as well as cardiac drug-mediated aberrant rhythms. Thus, the light weight and waterproof design holds promise to advance the next generation of mobile health and drug discovery.
Collapse
Affiliation(s)
- Xiaoxiao Zhang
- Department of Medical Engineering, California Institute of Technology, Pasadena, California, USA
| | - Tyler Beebe
- Department of Bioengineering, UCLA School of Engineering & Applied Sciences, Los Angeles, CA 90095
| | - Nelon Jen
- Department of Bioengineering, UCLA School of Engineering & Applied Sciences, Los Angeles, CA 90095
| | - Chia-An Lee
- Division of Cardiology, Department of Medicine, UCLA School of Medicine, Los Angeles, CA 90095
| | - Yuchong Tai
- Department of Medical Engineering, California Institute of Technology, Pasadena, California, USA
| | - Tzung K Hsiai
- Department of Bioengineering, UCLA School of Engineering & Applied Sciences, Los Angeles, CA 90095.,Division of Cardiology, Department of Medicine, UCLA School of Medicine, Los Angeles, CA 90095
| |
Collapse
|
34
|
Majumder R, Pandit R, Panfilov AV. Turbulent electrical activity at sharp-edged inexcitable obstacles in a model for human cardiac tissue. Am J Physiol Heart Circ Physiol 2014; 307:H1024-35. [DOI: 10.1152/ajpheart.00593.2013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Wave propagation around various geometric expansions, structures, and obstacles in cardiac tissue may result in the formation of unidirectional block of wave propagation and the onset of reentrant arrhythmias in the heart. Therefore, we investigated the conditions under which reentrant spiral waves can be generated by high-frequency stimulation at sharp-edged obstacles in the ten Tusscher-Noble-Noble-Panfilov (TNNP) ionic model for human cardiac tissue. We show that, in a large range of parameters that account for the conductance of major inward and outward ionic currents of the model [fast inward Na+ current ( INa), L—type slow inward Ca2+ current ( ICaL), slow delayed-rectifier current ( IKs), rapid delayed-rectifier current ( IKr), inward rectifier K+ current ( IK1)], the critical period necessary for spiral formation is close to the period of a spiral wave rotating in the same tissue. We also show that there is a minimal size of the obstacle for which formation of spirals is possible; this size is ∼2.5 cm and decreases with a decrease in the excitability of cardiac tissue. We show that other factors, such as the obstacle thickness and direction of wave propagation in relation to the obstacle, are of secondary importance and affect the conditions for spiral wave initiation only slightly. We also perform studies for obstacle shapes derived from experimental measurements of infarction scars and show that the formation of spiral waves there is facilitated by tissue remodeling around it. Overall, we demonstrate that the formation of reentrant sources around inexcitable obstacles is a potential mechanism for the onset of cardiac arrhythmias in the presence of a fast heart rate.
Collapse
Affiliation(s)
- Rupamanjari Majumder
- Centre for Condensed Matter Theory, Department of Physics, Indian Institute of Science, Bangalore, India
- Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Rahul Pandit
- Centre for Condensed Matter Theory, Department of Physics, Indian Institute of Science, Bangalore, India
- Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bangalore, India
| | - A. V. Panfilov
- Department of Physics and Astronomy, Gent University, Ghent, Belgium; and
- Moscow Institute of Physics and Technology (State University), Dolgoprudny, Moscow Region, Russia
| |
Collapse
|
35
|
Differentiating Drug-Induced Multichannel Block on the Electrocardiogram: Randomized Study of Dofetilide, Quinidine, Ranolazine, and Verapamil. Clin Pharmacol Ther 2014; 96:549-58. [DOI: 10.1038/clpt.2014.155] [Citation(s) in RCA: 180] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Accepted: 07/09/2014] [Indexed: 01/08/2023]
|
36
|
Ogiso M, Suzuki A, Shiga T, Nakai K, Shoda M, Hagiwara N. Effect of intravenous amiodarone on QT and T peak-T end dispersions in patients with nonischemic heart failure treated with cardiac resynchronization-defibrillator therapy and electrical storm. J Arrhythm 2014; 31:1-5. [PMID: 26336515 DOI: 10.1016/j.joa.2014.01.006] [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] [Received: 05/04/2013] [Revised: 01/13/2014] [Accepted: 01/29/2014] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND The effect of intravenous amiodarone on spatial and transmural dispersion of ventricular repolarization in patients receiving cardiac resynchronization therapy (CRT) remains unclear. METHODS We studied 14 patients with nonischemic heart failure who received CRT with a defibrillator, experienced electrical storm and were treated with intravenous amiodarone. Each patient underwent 12-lead electrocardiography (ECG) and 187-channel repolarization interval-difference mapping electrocardiography (187-ch RIDM-ECG) before and during the intravenous administration of amiodarone infusion. RESULTS A recurrence of ventricular tachyarrhythmia was observed in 2 patients during the early period of intravenous amiodarone therapy. Intravenous amiodarone increased the corrected QT interval (from 470±52 ms to 508±55 ms, P=0.003), but it significantly decreased the QT dispersion (from 107±35 ms to 49±27 ms, P=0.001), T peak-T end (Tp-e) dispersion (from 86±17 ms to 28±28 ms, P=0.001), and maximum inter-lead difference between corrected Tp-e intervals as measured by using the 187-ch RIDM-ECG (from 83±13 ms to 50±19 ms, P=0.001). CONCLUSIONS Intravenous amiodarone suppressed the electrical storm and decreased the QT and Tp-e dispersions in patients treated by using CRT with a defibrillator.
Collapse
Affiliation(s)
- Masataka Ogiso
- Department of Cardiology, Tokyo Women׳s Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666, Japan
| | - Atsushi Suzuki
- Department of Cardiology, Tokyo Women׳s Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666, Japan
| | - Tsuyoshi Shiga
- Department of Cardiology, Tokyo Women׳s Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666, Japan
| | - Kenji Nakai
- Department of Internal Medicine of Dentistry, Iwate Medical University, Morioka, Japan
| | - Morio Shoda
- Department of Cardiology, Tokyo Women׳s Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666, Japan
| | - Nobuhisa Hagiwara
- Department of Cardiology, Tokyo Women׳s Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666, Japan
| |
Collapse
|
37
|
Abstract
Late I Na is an integral part of the sodium current, which persists long after the fast-inactivating component. The magnitude of the late I Na is relatively small in all species and in all types of cardiomyocytes as compared with the amplitude of the fast sodium current, but it contributes significantly to the shape and duration of the action potential. This late component had been shown to increase in several acquired or congenital conditions, including hypoxia, oxidative stress, and heart failure, or due to mutations in SCN5A, which encodes the α-subunit of the sodium channel, as well as in channel-interacting proteins, including multiple β subunits and anchoring proteins. Patients with enhanced late I Na exhibit the type-3 long QT syndrome (LQT3) characterized by high propensity for the life-threatening ventricular arrhythmias, such as Torsade de Pointes (TdP), as well as for atrial fibrillation. There are several distinct mechanisms of arrhythmogenesis due to abnormal late I Na, including abnormal automaticity, early and delayed after depolarization-induced triggered activity, and dramatic increase of ventricular dispersion of repolarization. Many local anesthetic and antiarrhythmic agents have a higher potency to block late I Na as compared with fast I Na. Several novel compounds, including ranolazine, GS-458967, and F15845, appear to be the most selective inhibitors of cardiac late I Na reported to date. Selective inhibition of late I Na is expected to be an effective strategy for correcting these acquired and congenital channelopathies.
Collapse
|
38
|
Johannesen L, Vicente J, Gray RA, Galeotti L, Loring Z, Garnett CE, Florian J, Ugander M, Stockbridge N, Strauss DG. Improving the Assessment of Heart Toxicity for All New Drugs Through Translational Regulatory Science. Clin Pharmacol Ther 2013; 95:501-8. [DOI: 10.1038/clpt.2013.238] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Accepted: 12/04/2013] [Indexed: 11/09/2022]
|
39
|
Coppini R, Ferrantini C, Mazzoni L, Sartiani L, Olivotto I, Poggesi C, Cerbai E, Mugelli A. Regulation of intracellular Na(+) in health and disease: pathophysiological mechanisms and implications for treatment. Glob Cardiol Sci Pract 2013; 2013:222-42. [PMID: 24689024 PMCID: PMC3963757 DOI: 10.5339/gcsp.2013.30] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Accepted: 09/01/2013] [Indexed: 12/19/2022] Open
Abstract
Transmembrane sodium (Na+) fluxes and intracellular sodium homeostasis are central players in the physiology of the cardiac myocyte, since they are crucial for both cell excitability and for the regulation of the intracellular calcium concentration. Furthermore, Na+ fluxes across the membrane of mitochondria affect the concentration of protons and calcium in the matrix, regulating mitochondrial function. In this review we first analyze the main molecular determinants of sodium fluxes across the sarcolemma and the mitochondrial membrane and describe their role in the physiology of the healthy myocyte. In particular we focus on the interplay between intracellular Ca2+ and Na+. A large part of the review is dedicated to discuss the changes of Na+ fluxes and intracellular Na+ concentration([Na+]i) occurring in cardiac disease; we specifically focus on heart failure and hypertrophic cardiomyopathy, where increased intracellular [Na+]i is an established determinant of myocardial dysfunction. We review experimental evidence attributing the increase of [Na+]i to either decreased Na+ efflux (e.g. via the Na+/K+ pump) or increased Na+ influx into the myocyte (e.g. via Na+ channels). In particular, we focus on the role of the “late sodium current” (INaL), a sustained component of the fast Na+ current of cardiac myocytes, which is abnormally enhanced in cardiac diseases and contributes to both electrical and contractile dysfunction. We analyze the pathophysiological role of INaL enhancement in heart failure and hypertrophic cardiomyopathy and the consequences of its pharmacological modulation, highlighting the clinical implications. The central role of Na+ fluxes and intracellular Na+ physiology and pathophysiology of cardiac myocytes has been highlighted by a large number of recent works. The possibility of modulating Na+ inward fluxes and [Na+]i with specific INaL inhibitors, such as ranolazine, has made Na+a novel suitable target for cardiac therapy, potentially capable of addressing arrhythmogenesis and diastolic dysfunction in severe conditions such as heart failure and hypertrophic cardiomyopathy.
Collapse
Affiliation(s)
- Raffaele Coppini
- Department NeuroFarBa, Division of Pharmacology, University of Florence, Italy
| | - Cecilia Ferrantini
- Department of Clinical and Experimental Medicine, division of Physiology, University of Florence, Italy
| | - Luca Mazzoni
- Department NeuroFarBa, Division of Pharmacology, University of Florence, Italy
| | - Laura Sartiani
- Department NeuroFarBa, Division of Pharmacology, University of Florence, Italy
| | - Iacopo Olivotto
- Referral Center for Cardiomyopathies, Careggi University Hospital, Florence, Italy
| | - Corrado Poggesi
- Department of Clinical and Experimental Medicine, division of Physiology, University of Florence, Italy
| | - Elisabetta Cerbai
- Department NeuroFarBa, Division of Pharmacology, University of Florence, Italy
| | - Alessandro Mugelli
- Department NeuroFarBa, Division of Pharmacology, University of Florence, Italy
| |
Collapse
|
40
|
Toischer K, Hartmann N, Wagner S, Fischer TH, Herting J, Danner BC, Sag CM, Hund TJ, Mohler PJ, Belardinelli L, Hasenfuss G, Maier LS, Sossalla S. Role of late sodium current as a potential arrhythmogenic mechanism in the progression of pressure-induced heart disease. J Mol Cell Cardiol 2013; 61:111-22. [PMID: 23570977 PMCID: PMC3720777 DOI: 10.1016/j.yjmcc.2013.03.021] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Revised: 03/08/2013] [Accepted: 03/29/2013] [Indexed: 12/19/2022]
Abstract
The aim of the study was to determine the characteristics of the late Na current (INaL) and its arrhythmogenic potential in the progression of pressure-induced heart disease. Transverse aortic constriction (TAC) was used to induce pressure overload in mice. After one week the hearts developed isolated hypertrophy with preserved systolic contractility. In patch-clamp experiments both, INaL and the action potential duration (APD90) were unchanged. In contrast, after five weeks animals developed heart failure with prolonged APDs and slowed INaL decay time which could be normalized by addition of the INaL inhibitor ranolazine (Ran) or by the Ca/calmodulin-dependent protein kinase II (CaMKII) inhibitor AIP. Accordingly the APD90 could be significantly abbreviated by Ran, tetrodotoxin and the CaMKII inhibitor AIP. Isoproterenol increased the number of delayed afterdepolarizations (DAD) in myocytes from failing but not sham hearts. Application of either Ran or AIP prevented the occurrence of DADs. Moreover, the incidence of triggered activity was significantly increased in TAC myocytes and was largely prevented by Ran and AIP. Western blot analyses indicate that increased CaMKII activity and a hyperphosphorylation of the Nav1.5 at the CaMKII phosphorylation site (Ser571) paralleled our functional observations five weeks after TAC surgery. In pressure overload-induced heart failure a CaMKII-dependent augmentation of INaL plays a crucial role in the AP prolongation and generation of cellular arrhythmogenic triggers, which cannot be found in early and still compensated hypertrophy. Inhibition of INaL and CaMKII exerts potent antiarrhythmic effects and might therefore be of potential therapeutic interest. This article is part of a Special Issue entitled "Na(+) Regulation in Cardiac Myocytes".
Collapse
Affiliation(s)
- Karl Toischer
- Abt. Kardiologie und Pneumologie / Herzzentrum, Georg-August-Universität Göttingen, Germany
| | - Nico Hartmann
- Abt. Kardiologie und Pneumologie / Herzzentrum, Georg-August-Universität Göttingen, Germany
| | - Stefan Wagner
- Abt. Kardiologie und Pneumologie / Herzzentrum, Georg-August-Universität Göttingen, Germany
| | - Thomas H. Fischer
- Abt. Kardiologie und Pneumologie / Herzzentrum, Georg-August-Universität Göttingen, Germany
| | - Jonas Herting
- Abt. Kardiologie und Pneumologie / Herzzentrum, Georg-August-Universität Göttingen, Germany
| | - Bernhard C. Danner
- Abt. Herzund Thoraxchirurgie, Georg-August-Universität Göttingen, Germany
| | - Can M. Sag
- Abt. Kardiologie und Pneumologie / Herzzentrum, Georg-August-Universität Göttingen, Germany
| | - Thomas J. Hund
- Dorothy M. Davis Heart and Lung Research Institute, Dept. of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Peter J. Mohler
- Dorothy M. Davis Heart and Lung Research Institute, Dept. of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH
| | | | - Gerd Hasenfuss
- Abt. Kardiologie und Pneumologie / Herzzentrum, Georg-August-Universität Göttingen, Germany
| | - Lars S. Maier
- Abt. Kardiologie und Pneumologie / Herzzentrum, Georg-August-Universität Göttingen, Germany
| | - Samuel Sossalla
- Abt. Kardiologie und Pneumologie / Herzzentrum, Georg-August-Universität Göttingen, Germany
| |
Collapse
|
41
|
Shryock JC, Song Y, Rajamani S, Antzelevitch C, Belardinelli L. The arrhythmogenic consequences of increasing late INa in the cardiomyocyte. Cardiovasc Res 2013; 99:600-11. [PMID: 23752976 DOI: 10.1093/cvr/cvt145] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
This review presents the roles of cardiac sodium channel NaV1.5 late current (late INa) in generation of arrhythmic activity. The assumption of the authors is that proper Na(+) channel function is necessary to the maintenance of the transmembrane electrochemical gradient of Na(+) and regulation of cardiac electrical activity. Myocyte Na(+) channels' openings during the brief action potential upstroke contribute to peak INa and initiate excitation-contraction coupling. Openings of Na(+) channels outside the upstroke contribute to late INa, a depolarizing current that persists throughout the action potential plateau. The small, physiological late INa does not appear to be critical for normal electrical or contractile function in the heart. Late INa does, however, reduce the net repolarizing current, prolongs action potential duration, and increases cellular Na(+) loading. An increase of late INa, due to acquired conditions (e.g. heart failure) or inherited Na(+) channelopathies, facilitates the formation of early and delayed afterpolarizations and triggered arrhythmias, spontaneous diastolic depolarization, and cellular Ca(2+) loading. These in turn increase the spatial and temporal dispersion of repolarization time and may lead to reentrant arrhythmias.
Collapse
Affiliation(s)
- John C Shryock
- Department of Biology, Cardiovascular Therapeutic Area, Gilead Sciences, Foster City, CA, USA
| | | | | | | | | |
Collapse
|
42
|
Wilhelms M, Rombach C, Scholz EP, Dossel O, Seemann G. Impact of amiodarone and cisapride on simulated human ventricular electrophysiology and electrocardiograms. Europace 2012; 14 Suppl 5:v90-v96. [DOI: 10.1093/europace/eus281] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
43
|
Abstract
Letters to the Editor will be published, if suitable, as space permits. They should not exceed 1000 words (typed double-spaced) in length and may be subject to editing or abridgment.
Collapse
|
44
|
Affiliation(s)
- Andrew J Sauer
- Center for Human Genetic Research, Cardiovascular Research Center, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA
| | | |
Collapse
|
45
|
Effects of a novel amiodarone-like compound SAR114646A on the pig atrium and susceptibility to ventricular fibrillation in dogs and pigs. Naunyn Schmiedebergs Arch Pharmacol 2012; 385:373-84. [PMID: 22234329 DOI: 10.1007/s00210-011-0716-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2011] [Accepted: 12/02/2011] [Indexed: 10/14/2022]
Abstract
Amiodarone is one of the most effective antiarrhythmic drugs. However, poor solubility of this compound has limited its intravenous application. SAR11464A is a water-soluble amiodarone-like drug that lacks iodine and inhibits multiple cardiac ion channels in vitro. This study evaluated the antiarrhythmic efficacy of this drug in vivo. In porcine studies, atrial effective refractory period (AERP) was measured in pentobarbital-anesthetized thoracotomized pigs and atrial fibrillation (AF) was induced by a premature beat. Ventricular fibrillation (VF) was induced via either burst pacing or programmed electrical stimulation (a series of progressively shorter beats, S1-S5). In canine studies, VF was induced by a 2-min occlusion of the left circumflex coronary artery during the last minute of exercise in dogs with healed myocardial infarctions (n = 8). One week later, this test was repeated after pretreatment with SAR114646A (3.0 mg/kg, i.v., slow bolus). SAR114646A produced a significant dose-dependent prolongation of AERP, inhibited AF induced by a premature stimulus, and electrically induced VF in anesthetized pigs. At 1.0 and 3.0 mg/kg, i.v., it was superior to amiodarone, dofetilide, and flecainide. In dogs, SAR114646A did not alter any ECG parameter including QTc (control, 236.9 ± 8.5 ms vs. SAR, 237.2 ± 3.5 ms) but significantly reduced the incidence of VF, protecting six of eight animals (Fisher's exact test, P = 0.01). SAR114646A was effective against both atrial and ventricular arrhythmias without altering ventricular repolarization. These data suggest that the amiodarone-like drug SAR114646A may be an effective antiarrhythmic intervention that does not adversely prolong ventricular repolarization.
Collapse
|
46
|
Burashnikov A, Antzelevitch C. Novel pharmacological targets for the rhythm control management of atrial fibrillation. Pharmacol Ther 2011; 132:300-13. [PMID: 21867730 PMCID: PMC3205214 DOI: 10.1016/j.pharmthera.2011.08.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2011] [Accepted: 08/05/2011] [Indexed: 12/19/2022]
Abstract
Atrial fibrillation (AF) is a growing clinical problem associated with increased morbidity and mortality. Development of safe and effective pharmacological treatments for AF is one of the greatest unmet medical needs facing our society. In spite of significant progress in non-pharmacological AF treatments (largely due to the use of catheter ablation techniques), anti-arrhythmic agents (AADs) remain first line therapy for rhythm control management of AF for most AF patients. When considering efficacy, safety and tolerability, currently available AADs for rhythm control of AF are less than optimal. Ion channel inhibition remains the principal strategy for termination of AF and prevention of its recurrence. Practical clinical experience indicates that multi-ion channel blockers are generally more optimal for rhythm control of AF compared to ion channel-selective blockers. Recent studies suggest that atrial-selective sodium channel block can lead to safe and effective suppression of AF and that concurrent inhibition of potassium ion channels may potentiate this effect. An important limitation of the ion channel block approach for AF treatment is that non-electrical factors (largely structural remodeling) may importantly determine the generation of AF, so that "upstream therapy", aimed at preventing or reversing structural remodeling, may be required for effective rhythm control management. This review focuses on novel pharmacological targets for the rhythm control management of AF.
Collapse
|
47
|
Calloe K, Schmitt N, Grubb S, Pfeiffer R, David JP, Kanter R, Cordeiro JM, Antzelevitch C. Multiple arrhythmic syndromes in a newborn, owing to a novel mutation in SCN5A. Can J Physiol Pharmacol 2011; 89:723-36. [PMID: 21895525 DOI: 10.1139/y11-070] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Mutations in the SCN5A gene have been linked to Brugada syndrome (BrS), conduction disease, Long QT syndrome (LQT3), atrial fibrillation (AF), and to pre- and neonatal ventricular arrhythmias. OBJECTIVE The objective of this study is to characterize a novel mutation in Na(v)1.5 found in a newborn with fetal chaotic atrial tachycardia, post-partum intraventricular conduction delay, and QT interval prolongation. METHODS Genomic DNA was isolated and all exons and intron borders of 15 ion-channel genes were sequenced, revealing a novel missense mutation (Q270K) in SCN5A. Na(v)1.5 wild type (WT) and Q270K were expressed in CHO-K1 with and without the Na(v)β1 subunit. Results. Patch-clamp analysis showed ∼40% reduction in peak sodium channel current (I(Na)) density for Q270K compared with WT. Fast and slow decay of I(Na) were significantly slower in Q270K. Steady-state activation and inactivation of Q270K channels were shifted to positive potentials, and window current was increased. The tetrodotoxin-sensitive late I(Na) was increased almost 3-fold compared with WT channels. Ranolazine reduced late I(Na) in WT and Q270K channels, while exerting minimal effects on peak I(Na). CONCLUSION The Q270K mutation in SCN5A reduces peak I(Na) while augmenting late I(Na), and may thus underlie the development of atrial tachycardia, intraventricular conduction delay, and QT interval prolongation in an infant.
Collapse
Affiliation(s)
- Kirstine Calloe
- Danish National Research Foundation Centre for Cardiac Arrhythmia, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen N, Denmark.
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Antzelevitch C, Burashnikov A, Sicouri S, Belardinelli L. Electrophysiologic basis for the antiarrhythmic actions of ranolazine. Heart Rhythm 2011; 8:1281-90. [PMID: 21421082 PMCID: PMC3131428 DOI: 10.1016/j.hrthm.2011.03.045] [Citation(s) in RCA: 183] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2011] [Accepted: 03/11/2011] [Indexed: 12/19/2022]
Abstract
Ranolazine is a Food and Drug Administration-approved antianginal agent. Experimental and clinical studies have shown that ranolazine has antiarrhythmic effects in both ventricles and atria. In the ventricles, ranolazine can suppress arrhythmias associated with acute coronary syndrome, long QT syndrome, heart failure, ischemia, and reperfusion. In atria, ranolazine effectively suppresses atrial tachyarrhythmias and atrial fibrillation (AF). Recent studies have shown that the drug may be effective and safe in suppressing AF when used as a pill-in-the pocket approach, even in patients with structurally compromised hearts, warranting further study. The principal mechanism underlying ranolazine's antiarrhythmic actions is thought to be primarily via inhibition of late I(Na) in the ventricles and via use-dependent inhibition of peak I(Na) and I(Kr) in the atria. Short- and long-term safety of ranolazine has been demonstrated in the clinic, even in patients with structural heart disease. This review summarizes the available data regarding the electrophysiologic actions and antiarrhythmic properties of ranolazine in preclinical and clinical studies.
Collapse
|
49
|
Wu L, Ma J, Li H, Wang C, Grandi E, Zhang P, Luo A, Bers DM, Shryock JC, Belardinelli L. Late sodium current contributes to the reverse rate-dependent effect of IKr inhibition on ventricular repolarization. Circulation 2011; 123:1713-20. [PMID: 21482963 DOI: 10.1161/circulationaha.110.000661] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND The reverse rate dependence (RRD) of actions of I(Kr)-blocking drugs to increase the action potential duration (APD) and beat-to-beat variability of repolarization (BVR) of APD is proarrhythmic. We determined whether inhibition of endogenous, physiological late Na(+) current (late I(Na)) attenuates the RRD and proarrhythmic effect of I(Kr) inhibition. METHODS AND RESULTS Duration of the monophasic APD (MAPD) was measured from female rabbit hearts paced at cycle lengths from 400 to 2000 milliseconds, and BVR was calculated. In the absence of a drug, duration of monophasic action potential at 90% completion of repolarization (MAPD(90)) and BVR increased as the cycle length was increased from 400 to 2000 milliseconds (n=36 and 26; P<0.01). Both E-4031 (20 nmol/L) and d-sotalol (10 μmol/L) increased MAPD(90) and BVR at all stimulation rates, and the increase was greater at slower than at faster pacing rates (n=19, 11, 12 and 7, respectively; P<0.01). Tetrodotoxin (1 μmol/L) and ranolazine significantly attenuated the RRD of MAPD(90,) reduced BVR (P<0.01), and abolished torsade de pointes in hearts treated with either 20 nmol/L E-4031 or 10 μmol/L d-sotalol. Endogenous late I(Na) in cardiomyocytes stimulated at cycle lengths from 500 to 4000 milliseconds was greater at slower than at faster stimulation rates, and rapidly decreased during the first several beats at faster but not at slower rates (n=8; P<0.01). In a computational model, simulated RRD of APD caused by E-4031 and d-sotalol was attenuated when late I(Na) was inhibited. CONCLUSION Endogenous late I(Na) contributes to the RRD of I(Kr) inhibitor-induced increases in APD and BVR and to bradycardia-related ventricular arrhythmias.
Collapse
Affiliation(s)
- Lin Wu
- Department of Biology, Gilead Sciences, 1651 Page Mill Road, Palo Alto, CA 94304, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
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: 1.3] [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
|