1
|
Kyro GW, Martin MT, Watt ED, Batista VS. CardioGenAI: a machine learning-based framework for re-engineering drugs for reduced hERG liability. J Cheminform 2025; 17:30. [PMID: 40045386 PMCID: PMC11881490 DOI: 10.1186/s13321-025-00976-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Accepted: 02/21/2025] [Indexed: 03/09/2025] Open
Abstract
The link between in vitro hERG ion channel inhibition and subsequent in vivo QT interval prolongation, a critical risk factor for the development of arrythmias such as Torsade de Pointes, is so well established that in vitro hERG activity alone is often sufficient to end the development of an otherwise promising drug candidate. It is therefore of tremendous interest to develop advanced methods for identifying hERG-active compounds in the early stages of drug development, as well as for proposing redesigned compounds with reduced hERG liability and preserved primary pharmacology. In this work, we present CardioGenAI, a machine learning-based framework for re-engineering both developmental and commercially available drugs for reduced hERG activity while preserving their pharmacological activity. The framework incorporates novel state-of-the-art discriminative models for predicting hERG channel activity, as well as activity against the voltage-gated NaV1.5 and CaV1.2 channels due to their potential implications in modulating the arrhythmogenic potential induced by hERG channel blockade. We applied the complete framework to pimozide, an FDA-approved antipsychotic agent that demonstrates high affinity to the hERG channel, and generated 100 refined candidates. Remarkably, among the candidates is fluspirilene, a compound which is of the same class of drugs as pimozide (diphenylmethanes) and therefore has similar pharmacological activity, yet exhibits over 700-fold weaker binding to hERG. Furthermore, we demonstrated the framework's ability to optimize hERG, NaV1.5 and CaV1.2 profiles of multiple FDA-approved compounds while maintaining the physicochemical nature of the original drugs. We envision that this method can effectively be applied to developmental compounds exhibiting hERG liabilities to provide a means of rescuing drug development programs that have stalled due to hERG-related safety concerns. Additionally, the discriminative models can also serve independently as effective components of virtual screening pipelines. We have made all of our software open-source at https://github.com/gregory-kyro/CardioGenAI to facilitate integration of the CardioGenAI framework for molecular hypothesis generation into drug discovery workflows.Scientific contributionThis work introduces CardioGenAI, an open-source machine learning-based framework designed to re-engineer drugs for reduced hERG liability while preserving their pharmacological activity. The complete CardioGenAI framework can be applied to developmental compounds exhibiting hERG liabilities to provide a means of rescuing drug discovery programs facing hERG-related challenges. In addition, the framework incorporates novel state-of-the-art discriminative models for predicting hERG, NaV1.5 and CaV1.2 channel activity, which can function independently as effective components of virtual screening pipelines.
Collapse
Affiliation(s)
- Gregory W Kyro
- Department of Chemistry, Yale University, New Haven, CT, 06511, USA.
- Drug Safety Research & Development, Pfizer Research & Development, Groton, CT, 06340, USA.
| | - Matthew T Martin
- Drug Safety Research & Development, Pfizer Research & Development, Groton, CT, 06340, USA
| | - Eric D Watt
- Drug Safety Research & Development, Pfizer Research & Development, Groton, CT, 06340, USA
| | - Victor S Batista
- Department of Chemistry, Yale University, New Haven, CT, 06511, USA.
| |
Collapse
|
2
|
Mu Y, Hu S, Liu X, Tang X, Lin J, Shi H. Mechanical forces pattern endocardial Notch activation via mTORC2-PKC pathway. eLife 2025; 13:RP97268. [PMID: 39932433 PMCID: PMC11813223 DOI: 10.7554/elife.97268] [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] [Indexed: 02/13/2025] Open
Abstract
Notch signaling has been identified as a key regulatory pathway in patterning the endocardium through activation of endothelial-to-mesenchymal transition (EMT) in the atrioventricular canal (AVC) and proximal outflow tract (OFT) region. However, the precise mechanism underlying Notch activation remains elusive. By transiently blocking the heartbeat of E9.5 mouse embryos, we found that Notch activation in the arterial endothelium was dependent on its ligand Dll4, whereas the reduced expression of Dll4 in the endocardium led to a ligand-depleted field, enabling Notch to be specifically activated in AVC and OFT by regional increased shear stress. The strong shear stress altered the membrane lipid microdomain structure of endocardial cells, which activated mTORC2 and PKC and promoted Notch1 cleavage even in the absence of strong ligand stimulation. These findings highlight the role of mechanical forces as a primary cue for endocardial patterning and provide insights into the mechanisms underlying congenital heart diseases of endocardial origin.
Collapse
Affiliation(s)
- Yunfei Mu
- Fudan UniversityShanghaiChina
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake UniversityHangzhouChina
- Westlake Laboratory of Life Sciences and BiomedicineHangzhouChina
- Institute of Basic Medical Sciences, Westlake Institute for Advanced StudyHangzhouChina
| | - Shijia Hu
- Fudan UniversityShanghaiChina
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake UniversityHangzhouChina
- Westlake Laboratory of Life Sciences and BiomedicineHangzhouChina
- Institute of Basic Medical Sciences, Westlake Institute for Advanced StudyHangzhouChina
| | - Xiangyang Liu
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake UniversityHangzhouChina
- Westlake Laboratory of Life Sciences and BiomedicineHangzhouChina
- Institute of Basic Medical Sciences, Westlake Institute for Advanced StudyHangzhouChina
| | - Xin Tang
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake UniversityHangzhouChina
- Westlake Laboratory of Life Sciences and BiomedicineHangzhouChina
- Institute of Basic Medical Sciences, Westlake Institute for Advanced StudyHangzhouChina
| | - Jiayi Lin
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake UniversityHangzhouChina
- Westlake Laboratory of Life Sciences and BiomedicineHangzhouChina
- Institute of Basic Medical Sciences, Westlake Institute for Advanced StudyHangzhouChina
| | - Hongjun Shi
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake UniversityHangzhouChina
- Westlake Laboratory of Life Sciences and BiomedicineHangzhouChina
- Institute of Basic Medical Sciences, Westlake Institute for Advanced StudyHangzhouChina
| |
Collapse
|
3
|
Deshmukh A, Yokokawa M, McBride D, Simpson J, Chou A, Ghannam M, Liang JJ, Saeed M, Cunnane R, Ghanbari H, Latchamsetty R, Crawford T, Jongnarangsin K, Pelosi F, Chugh A, Morady F, Bogun F, Oral H. Dofetilide for the treatment of premature ventricular complexes and ventricular tachycardia in patients with structural heart disease. J Cardiovasc Electrophysiol 2024; 35:2363-2371. [PMID: 39363447 DOI: 10.1111/jce.16452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 09/15/2024] [Accepted: 09/22/2024] [Indexed: 10/05/2024]
Abstract
BACKGROUND Dofetilide is a class III antiarrhythmic agent approved for the treatment of atrial fibrillation and atrial flutter. Given the efficacy of other class III agents, it has been used off-label for the treatment of premature ventricular complexes (PVCs) and ventricular tachycardias (VTs). OBJECTIVE The purpose of this study was to determine the efficacy and safety of dofetilide for ventricular arrythmias (VAs). METHODS In this retrospective cohort study, 81 patients (59 men; age = 60 ± 14 years; LVEF = 0.34 ± 0.16) were admitted for dofetilide initiation to treat PVCs (29), VTs (42) or both (10). A ≥ 80% decrease in PVC burden was defined as a satisfactory response. An ICD was present in 72 patients (89%). Another antiarrhythmic was previously used in 50 patients (62%). Prior catheter ablation had been performed in 33 patients (41%). RESULTS During intitiation, dofetilide was discontinued in 12 patients (15%) due to QT prolongation (8) and inefficacy to suppress VAs (4). Among the 32 patients with PVCs who successfully started dofetilide, the mean PVC burden decreased from 20 ± 10% to 8 ± 8% at a median follow-up of 2.6 months (p < .001). PVC burden was reduced by ≥80% in only 11/32 patients (34%). During 7 ± 1 years of follow-up, 41/69 patients (59%) continued to have VAs and received appropriate ICD therapies for monomorphic VTs (35) and polymorphic VT/VF (6) at a median of 8.0 (IQR 2.6-33.2) months. Dofetilide had to be discontinued in 50/69 patients (72%) due to inefficacy or intolerance. The composite outcome of VT/VF recurrence, heart transplantation, or death occurred in 6/12 patients (50%) without dofetilide and 49/69 patients (71%) with dofetilide. The event free survival was similar between patients treated with and without dofetilide (log-rank p = .55). CONCLUSIONS Treatment with dofetilide was associated with a decrease in PVCs, however clinically significant suppression occurred in a minority of patients. Dofetilide failed to suppress the occurrence of VTs in a majority of patients.
Collapse
Affiliation(s)
- Amrish Deshmukh
- Division of Cardiovascular Medicine, Cardiac Arrhythmia Service, University of Michigan, Ann Arbor, Michigan, USA
| | - Miki Yokokawa
- Division of Cardiovascular Medicine, Cardiac Arrhythmia Service, University of Michigan, Ann Arbor, Michigan, USA
| | - Daniel McBride
- Division of Cardiovascular Medicine, Cardiac Arrhythmia Service, University of Michigan, Ann Arbor, Michigan, USA
| | - Jamie Simpson
- Division of Cardiovascular Medicine, Cardiac Arrhythmia Service, University of Michigan, Ann Arbor, Michigan, USA
| | - Andrew Chou
- Henry Ford Heart and Vascular Institute, Henry Ford Hospital, Detroit, Michigan, USA
| | - Michael Ghannam
- Division of Cardiovascular Medicine, Cardiac Arrhythmia Service, University of Michigan, Ann Arbor, Michigan, USA
| | - Jackson J Liang
- Division of Cardiovascular Medicine, Cardiac Arrhythmia Service, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Ryan Cunnane
- Division of Cardiovascular Medicine, Cardiac Arrhythmia Service, University of Michigan, Ann Arbor, Michigan, USA
| | - Hamid Ghanbari
- Division of Cardiovascular Medicine, Cardiac Arrhythmia Service, University of Michigan, Ann Arbor, Michigan, USA
| | - Rakesh Latchamsetty
- Division of Cardiovascular Medicine, Cardiac Arrhythmia Service, University of Michigan, Ann Arbor, Michigan, USA
| | - Thomas Crawford
- Division of Cardiovascular Medicine, Cardiac Arrhythmia Service, University of Michigan, Ann Arbor, Michigan, USA
| | - Krit Jongnarangsin
- Division of Cardiovascular Medicine, Cardiac Arrhythmia Service, University of Michigan, Ann Arbor, Michigan, USA
| | - Frank Pelosi
- Division of Cardiovascular Medicine, Cardiac Arrhythmia Service, University of Michigan, Ann Arbor, Michigan, USA
| | - Aman Chugh
- Division of Cardiovascular Medicine, Cardiac Arrhythmia Service, University of Michigan, Ann Arbor, Michigan, USA
| | - Fred Morady
- Division of Cardiovascular Medicine, Cardiac Arrhythmia Service, University of Michigan, Ann Arbor, Michigan, USA
| | - Frank Bogun
- Division of Cardiovascular Medicine, Cardiac Arrhythmia Service, University of Michigan, Ann Arbor, Michigan, USA
| | - Hakan Oral
- Division of Cardiovascular Medicine, Cardiac Arrhythmia Service, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
4
|
Aguado-Sierra J, Dominguez-Gomez P, Amar A, Butakoff C, Leitner M, Schaper S, Kriegl JM, Darpo B, Vazquez M, Rast G. Virtual clinical QT exposure-response studies - A translational computational approach. J Pharmacol Toxicol Methods 2024; 126:107498. [PMID: 38432528 DOI: 10.1016/j.vascn.2024.107498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 12/13/2023] [Accepted: 02/29/2024] [Indexed: 03/05/2024]
Abstract
BACKGROUND AND PURPOSE A recent paradigm shift in proarrhythmic risk assessment suggests that the integration of clinical, non-clinical, and computational evidence can be used to reach a comprehensive understanding of the proarrhythmic potential of drug candidates. While current computational methodologies focus on predicting the incidence of proarrhythmic events after drug administration, the objective of this study is to predict concentration-response relationships of QTc as a clinical endpoint. EXPERIMENTAL APPROACH Full heart computational models reproducing human cardiac populations were created to predict the concentration-response relationship of changes in the QT interval as recommended for clinical trials. The concentration-response relationship of the QT-interval prolongation obtained from the computational cardiac population was compared against the relationship from clinical trial data for a set of well-characterized compounds: moxifloxacin, dofetilide, verapamil, and ondansetron. KEY RESULTS Computationally derived concentration-response relationships of QT interval changes for three of the four drugs had slopes within the confidence interval of clinical trials (dofetilide, moxifloxacin and verapamil) when compared to placebo-corrected concentration-ΔQT and concentration-ΔQT regressions. Moxifloxacin showed a higher intercept, outside the confidence interval of the clinical data, demonstrating that in this example, the standard linear regression does not appropriately capture the concentration-response results at very low concentrations. The concentrations corresponding to a mean QTc prolongation of 10 ms were consistently lower in the computational model than in clinical data. The critical concentration varied within an approximate ratio of 0.5 (moxifloxacin and ondansetron) and 1 times (dofetilide, verapamil) the critical concentration observed in human clinical trials. Notably, no other in silico methodology can approximate the human critical concentration values for a QT interval prolongation of 10 ms. CONCLUSION AND IMPLICATIONS Computational concentration-response modelling of a virtual population of high-resolution, 3-dimensional cardiac models can provide comparable information to clinical data and could be used to complement pre-clinical and clinical safety packages. It provides access to an unlimited exposure range to support trial design and can improve the understanding of pre-clinical-clinical translation.
Collapse
Affiliation(s)
- Jazmin Aguado-Sierra
- Elem Biotech, Barcelona, Spain; Barcelona Supercomputing Center, Barcelona, Spain.
| | | | | | | | - Michael Leitner
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH and Co. KG, Biberach, Germany.
| | - Stefan Schaper
- Global Computational Biology and Digital Sciences, Boehringer Ingelheim Pharma GmbH and Co. KG, Biberach, Germany.
| | - Jan M Kriegl
- Global Computational Biology and Digital Sciences, Boehringer Ingelheim Pharma GmbH and Co. KG, Biberach, Germany.
| | | | - Mariano Vazquez
- Elem Biotech, Barcelona, Spain; Barcelona Supercomputing Center, Barcelona, Spain.
| | - Georg Rast
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH and Co. KG, Biberach, Germany.
| |
Collapse
|
5
|
Brown GE, Han YD, Michell AR, Ly OT, Vanoye CG, Spanghero E, George AL, Darbar D, Khetani SR. Engineered cocultures of iPSC-derived atrial cardiomyocytes and atrial fibroblasts for modeling atrial fibrillation. SCIENCE ADVANCES 2024; 10:eadg1222. [PMID: 38241367 PMCID: PMC10798559 DOI: 10.1126/sciadv.adg1222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 12/21/2023] [Indexed: 01/21/2024]
Abstract
Atrial fibrillation (AF) is the most common sustained cardiac arrhythmia treatable with antiarrhythmic drugs; however, patient responses remain highly variable. Human induced pluripotent stem cell-derived atrial cardiomyocytes (iPSC-aCMs) are useful for discovering precision therapeutics, but current platforms yield phenotypically immature cells and are not easily scalable for high-throughput screening. Here, primary adult atrial, but not ventricular, fibroblasts induced greater functional iPSC-aCM maturation, partly through connexin-40 and ephrin-B1 signaling. We developed a protein patterning process within multiwell plates to engineer patterned iPSC-aCM and atrial fibroblast coculture (PC) that significantly enhanced iPSC-aCM structural, electrical, contractile, and metabolic maturation for 6+ weeks compared to conventional mono-/coculture. PC displayed greater sensitivity for detecting drug efficacy than monoculture and enabled the modeling and pharmacological or gene editing treatment of an AF-like electrophysiological phenotype due to a mutated sodium channel. Overall, PC is useful for elucidating cell signaling in the atria, drug screening, and modeling AF.
Collapse
Affiliation(s)
- Grace E. Brown
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, USA
| | - Yong Duk Han
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, USA
| | - Ashlin R. Michell
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, USA
| | - Olivia T. Ly
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, USA
- Division of Cardiology, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Carlos G. Vanoye
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Emanuele Spanghero
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, USA
| | - Alfred L. George
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Dawood Darbar
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, USA
- Division of Cardiology, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, USA
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Salman R. Khetani
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
6
|
Al-Hamaly MA, Cox AH, Haney MG, Zhang W, Arvin EC, Sampathi S, Wimsett M, Liu C, Blackburn JS. Zebrafish drug screening identifies Erlotinib as an inhibitor of Wnt/β-catenin signaling and self-renewal in T-cell acute lymphoblastic leukemia. Biomed Pharmacother 2024; 170:116013. [PMID: 38104416 PMCID: PMC10833092 DOI: 10.1016/j.biopha.2023.116013] [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: 09/22/2023] [Revised: 12/06/2023] [Accepted: 12/13/2023] [Indexed: 12/19/2023] Open
Abstract
The Wnt/β-catenin pathway's significance in cancer initiation, progression, and stem cell biology underscores its therapeutic potential. However, the clinical application of Wnt inhibitors remains limited due to challenges posed by off-target effects and complex cross-talk of Wnt signaling with other pathways. In this study, we leveraged a zebrafish model to perform a robust and rapid drug screening of 773 FDA-approved compounds to identify Wnt/β-catenin inhibitors with minimal toxicity. Utilizing zebrafish expressing a Wnt reporter, we identified several drugs that suppressed Wnt signaling without compromising zebrafish development. The efficacy of the top hit, Erlotinib, extended to human cells, where it blocked Wnt/β-catenin signaling downstream of the destruction complex. Notably, Erlotinib treatment reduced self-renewal in human T-cell Acute Lymphoblastic Leukemia cells, which rely on active β-catenin signaling for maintenance of leukemia-initiating cells. Erlotinib also reduced leukemia-initiating cell frequency and delayed disease formation in zebrafish models. This study underscores zebrafish's translational potential in drug discovery and repurposing and highlights a new use for Erlotinib as a Wnt inhibitor for cancers driven by aberrant Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Majd A Al-Hamaly
- Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40356, United States; Markey Cancer Center, University of Kentucky, Lexington, KY 40536, United States
| | - Anna H Cox
- College of Medicine, University of Kentucky, Lexington, KY 40536, United States; Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY 40356, United States
| | - Meghan G Haney
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States
| | - Wen Zhang
- Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY 40356, United States
| | - Emma C Arvin
- Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY 40356, United States
| | - Shilpa Sampathi
- Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY 40356, United States
| | - Mary Wimsett
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States
| | - Chunming Liu
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, United States; Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY 40356, United States
| | - Jessica S Blackburn
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, United States; Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY 40356, United States.
| |
Collapse
|
7
|
Shantha G, Singleton M, Kozak P, Bodziock G, Atreya AR, Narasimhan B, Deshmukh A, Liang JJ, Hranitzky P, Whalen P, Bhave P. Role of dofetilide in patients with ventricular arrhythmias. J Interv Card Electrophysiol 2024; 67:91-97. [PMID: 37247098 DOI: 10.1007/s10840-023-01578-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 05/21/2023] [Indexed: 05/30/2023]
Abstract
BACKGROUND OR PURPOSE To assess effectiveness of dofetilide in reducing the burden of ventricular arrhythmias (VAs). BACKGROUND Prior small sample studies show that dofetilide has benefit in reducing VA. However, large sample investigations with long-term follow-up are lacking. METHODS Two hundred seventeen consecutive patients admitted between January 2015 and December 2021 for dofetilide initiation for control of VA were assessed. Dofetilide was successfully started in 176 patients (81%) and had to be discontinued in the remaining 41 patients (19%). Dofetilide was initiated for control of ventricular tachycardia (VT) in 136 patients (77%), whereas 40 (23%) patients were initiated on dofetilide for reducing the burden of premature ventricular complexes (PVCs). RESULTS The mean follow-up was 24 ± 7 months. In total, among the 136 VT patients, 33 (24%) died, 11 (8%) received a left ventricular assist device (LVAD), and 3 (2%) received a heart transplant during follow-up. Dofetilide was discontinued in 117 (86%) patients due to lack of sustained effectiveness during follow-up. Dofetilide use was associated with similar odds of the composite outcome of all-cause mortality/LVAD/heart transplant (OR: 0.97, 0.55-4.23) in patients with ischemic cardiomyopathy (ICM) compared to those with non-ischemic cardiomyopathy (NICM). Dofetilide did not reduce PVC burden during follow-up in the 40 patients with PVCs (mean baseline PVC burden: 15%, at 1-year follow-up: 14%). CONCLUSIONS Dofetilide use was less effective in reducing VA burden in our cohort of patients. Randomized controlled studies are needed to confirm our findings.
Collapse
Affiliation(s)
- Ghanshyam Shantha
- Cardiac Electrophysiology, Wake Forest University, 1, Medical Center Blvd, Winston-Salem, NC, 27157, USA.
| | | | - Patrick Kozak
- Cardiac Electrophysiology, Wake Forest University, 1, Medical Center Blvd, Winston-Salem, NC, 27157, USA
| | - George Bodziock
- Cardiac Electrophysiology, Wake Forest University, 1, Medical Center Blvd, Winston-Salem, NC, 27157, USA
| | - Auras R Atreya
- Cardiac Electrophysiology, University of Arkansas, Little Rock, USA
| | - Bharat Narasimhan
- Debakey Cardiovascular Center, Houston Methodist Hospital, Houston, TX, USA
| | | | - Jackson J Liang
- Cardiac Electrophysiology, University of Michigan, Ann Arbor, MI, USA
| | | | - Patrick Whalen
- Cardiac Electrophysiology, Wake Forest University, 1, Medical Center Blvd, Winston-Salem, NC, 27157, USA
| | - Prashant Bhave
- Cardiac Electrophysiology, Wake Forest University, 1, Medical Center Blvd, Winston-Salem, NC, 27157, USA
| |
Collapse
|
8
|
Kingma J, Simard C, Drolet B. Overview of Cardiac Arrhythmias and Treatment Strategies. Pharmaceuticals (Basel) 2023; 16:844. [PMID: 37375791 DOI: 10.3390/ph16060844] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 05/30/2023] [Accepted: 06/01/2023] [Indexed: 06/29/2023] Open
Abstract
Maintenance of normal cardiac rhythm requires coordinated activity of ion channels and transporters that allow well-ordered propagation of electrical impulses across the myocardium. Disruptions in this orderly process provoke cardiac arrhythmias that may be lethal in some patients. Risk of common acquired arrhythmias is increased markedly when structural heart disease caused by myocardial infarction (due to fibrotic scar formation) or left ventricular dysfunction is present. Genetic polymorphisms influence structure or excitability of the myocardial substrate, which increases vulnerability or risk of arrhythmias in patients. Similarly, genetic polymorphisms of drug-metabolizing enzymes give rise to distinct subgroups within the population that affect specific drug biotransformation reactions. Nonetheless, identification of triggers involved in initiation or maintenance of cardiac arrhythmias remains a major challenge. Herein, we provide an overview of knowledge regarding physiopathology of inherited and acquired cardiac arrhythmias along with a summary of treatments (pharmacologic or non-pharmacologic) used to limit their effect on morbidity and potential mortality. Improved understanding of molecular and cellular aspects of arrhythmogenesis and more epidemiologic studies (for a more accurate portrait of incidence and prevalence) are crucial for development of novel treatments and for management of cardiac arrhythmias and their consequences in patients, as their incidence is increasing worldwide.
Collapse
Affiliation(s)
- John Kingma
- Department of Medicine, Ferdinand Vandry Pavillon, 1050 Av. de la Médecine, Québec City, QC G1V 0A6, Canada
| | - Chantale Simard
- Faculty of Pharmacy Ferdinand Vandry Pavillon, 1050 Av. de la Médecine, Québec City, QC G1V 0A6, Canada
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec-Université Laval 2725 Chemin Sainte-Foy, Québec City, QC G1V 4G5, Canada
| | - Benoît Drolet
- Faculty of Pharmacy Ferdinand Vandry Pavillon, 1050 Av. de la Médecine, Québec City, QC G1V 0A6, Canada
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec-Université Laval 2725 Chemin Sainte-Foy, Québec City, QC G1V 4G5, Canada
| |
Collapse
|
9
|
Leow JWH, Gu Y, Chan ECY. Investigating the relevance of CYP2J2 inhibition for drugs known to cause intermediate to high risk torsades de pointes. Eur J Pharm Sci 2023; 187:106475. [PMID: 37225005 DOI: 10.1016/j.ejps.2023.106475] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 04/10/2023] [Accepted: 05/21/2023] [Indexed: 05/26/2023]
Abstract
Cardiac cytochrome P450 2J2 (CYP2J2) metabolizes endogenous polyunsaturated fatty acid, arachidonic acid (AA), to bioactive regioisomeric epoxyeicosatrienoic acid (EET) metabolites. This endogenous metabolic pathway has been postulated to play a homeostatic role in cardiac electrophysiology. However, it is unknown if drugs that cause intermediate to high risk torsades de pointes (TdP) exhibit inhibitory effects against CYP2J2 metabolism of AA to EETs. In this study, we demonstrated that 11 out of 16 drugs screened with intermediate to high risk of TdP as defined by the Comprehensive in vitro Proarrhythmia Assay (CiPA) initiative are concurrently reversible inhibitors of CYP2J2 metabolism of AA, with unbound inhibitory constant (Ki,AA,u) values ranging widely from 0.132 to 19.9 µM. To understand the physiological relevancy of Ki,AA,u, the in vivo unbound drug concentration within human heart tissue (Cu,heart) was calculated via experimental determination of in vitro unbound partition coefficient (Kpuu) for 10 CYP2J2 inhibitors using AC16 human ventricular cardiomyocytes as well as literature-derived values of fraction unbound in plasma (fu,p) and plasma drug concentrations in clinical scenarios leading to TdP. Notably, all CYP2J2 inhibitors screened belonging to the high TdP risk category, namely vandetanib and bepridil, exhibited highest Kpuu values of 18.2 ± 1.39 and 7.48 ± 1.16 respectively although no clear relationship between Cu,heart and risk of TdP could eventually be determined. R values based on basic models of reversible inhibition as per FDA guidelines were calculated using unbound plasma drug concentrations (Cu,plasma) and adapted using Cu,heart which suggested that 4 out of 10 CYP2J2 inhibitors with intermediate to high risk of TdP demonstrate greatest potential for clinically relevant in vivo cardiac drug-AA interactions. Our results shed novel insights on the relevance of CYP2J2 inhibition in drugs with risk of TdP. Further studies ascertaining the role of CYP2J2 metabolism of AA in cardiac electrophysiology, characterizing inherent cardiac ion channel activities of drugs with risk of TdP as well as in vivo evidence of drug-AA interactions will be required prior to determining if CYP2J2 inhibition could be an alternative mechanism contributing to drug-induced TdP.
Collapse
Affiliation(s)
- Jacqueline Wen Hui Leow
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore, 117543
| | - Yuxiang Gu
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore, 117543; School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, Heilongjiang, 150001, China
| | - Eric Chun Yong Chan
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore, 117543.
| |
Collapse
|
10
|
Uddin ME, Eisenmann ED, Li Y, Huang KM, Garrison DA, Talebi Z, Gibson AA, Jin Y, Nepal M, Bonilla IM, Fu Q, Sun X, Millar A, Tarasov M, Jay CE, Cui X, Einolf HJ, Pelis RM, Smith SA, Radwański PB, Sweet DH, König J, Fromm MF, Carnes CA, Hu S, Sparreboom A. MATE1 Deficiency Exacerbates Dofetilide-Induced Proarrhythmia. Int J Mol Sci 2022; 23:8607. [PMID: 35955741 PMCID: PMC9369325 DOI: 10.3390/ijms23158607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 07/30/2022] [Accepted: 07/30/2022] [Indexed: 02/04/2023] Open
Abstract
Dofetilide is a rapid delayed rectifier potassium current inhibitor widely used to prevent the recurrence of atrial fibrillation and flutter. The clinical use of this drug is associated with increases in QTc interval, which predispose patients to ventricular cardiac arrhythmias. The mechanisms involved in the disposition of dofetilide, including its movement in and out of cardiomyocytes, remain unknown. Using a xenobiotic transporter screen, we identified MATE1 (SLC47A1) as a transporter of dofetilide and found that genetic knockout or pharmacological inhibition of MATE1 in mice was associated with enhanced retention of dofetilide in cardiomyocytes and increased QTc prolongation. The urinary excretion of dofetilide was also dependent on the MATE1 genotype, and we found that this transport mechanism provides a mechanistic basis for previously recorded drug-drug interactions of dofetilide with various contraindicated drugs, including bictegravir, cimetidine, ketoconazole, and verapamil. The translational significance of these observations was examined with a physiologically-based pharmacokinetic model that adequately predicted the drug-drug interaction liabilities in humans. These findings support the thesis that MATE1 serves a conserved cardioprotective role by restricting excessive cellular accumulation and warrant caution against the concurrent administration of potent MATE1 inhibitors and cardiotoxic substrates with a narrow therapeutic window.
Collapse
Affiliation(s)
- Muhammad Erfan Uddin
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (M.E.U.); (E.D.E.); (Y.L.); (K.M.H.); (D.A.G.); (Z.T.); (A.A.G.); (Y.J.); (M.N.); (Q.F.); (X.S.)
| | - Eric D. Eisenmann
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (M.E.U.); (E.D.E.); (Y.L.); (K.M.H.); (D.A.G.); (Z.T.); (A.A.G.); (Y.J.); (M.N.); (Q.F.); (X.S.)
| | - Yang Li
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (M.E.U.); (E.D.E.); (Y.L.); (K.M.H.); (D.A.G.); (Z.T.); (A.A.G.); (Y.J.); (M.N.); (Q.F.); (X.S.)
| | - Kevin M. Huang
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (M.E.U.); (E.D.E.); (Y.L.); (K.M.H.); (D.A.G.); (Z.T.); (A.A.G.); (Y.J.); (M.N.); (Q.F.); (X.S.)
| | - Dominique A. Garrison
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (M.E.U.); (E.D.E.); (Y.L.); (K.M.H.); (D.A.G.); (Z.T.); (A.A.G.); (Y.J.); (M.N.); (Q.F.); (X.S.)
| | - Zahra Talebi
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (M.E.U.); (E.D.E.); (Y.L.); (K.M.H.); (D.A.G.); (Z.T.); (A.A.G.); (Y.J.); (M.N.); (Q.F.); (X.S.)
| | - Alice A. Gibson
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (M.E.U.); (E.D.E.); (Y.L.); (K.M.H.); (D.A.G.); (Z.T.); (A.A.G.); (Y.J.); (M.N.); (Q.F.); (X.S.)
| | - Yan Jin
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (M.E.U.); (E.D.E.); (Y.L.); (K.M.H.); (D.A.G.); (Z.T.); (A.A.G.); (Y.J.); (M.N.); (Q.F.); (X.S.)
| | - Mahesh Nepal
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (M.E.U.); (E.D.E.); (Y.L.); (K.M.H.); (D.A.G.); (Z.T.); (A.A.G.); (Y.J.); (M.N.); (Q.F.); (X.S.)
| | - Ingrid M. Bonilla
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA;
| | - Qiang Fu
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (M.E.U.); (E.D.E.); (Y.L.); (K.M.H.); (D.A.G.); (Z.T.); (A.A.G.); (Y.J.); (M.N.); (Q.F.); (X.S.)
| | - Xinxin Sun
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (M.E.U.); (E.D.E.); (Y.L.); (K.M.H.); (D.A.G.); (Z.T.); (A.A.G.); (Y.J.); (M.N.); (Q.F.); (X.S.)
| | - Alec Millar
- Division of Outcomes and Translational Sciences, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA; (A.M.); (M.T.); (P.B.R.); (C.A.C.); (S.H.)
| | - Mikhail Tarasov
- Division of Outcomes and Translational Sciences, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA; (A.M.); (M.T.); (P.B.R.); (C.A.C.); (S.H.)
| | - Christopher E. Jay
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA; (C.E.J.); (D.H.S.)
| | - Xiaoming Cui
- Novartis Institute for Biomedical Research, East Hanover, NJ 07936, USA; (X.C.); (H.J.E.); (R.M.P.)
| | - Heidi J. Einolf
- Novartis Institute for Biomedical Research, East Hanover, NJ 07936, USA; (X.C.); (H.J.E.); (R.M.P.)
| | - Ryan M. Pelis
- Novartis Institute for Biomedical Research, East Hanover, NJ 07936, USA; (X.C.); (H.J.E.); (R.M.P.)
| | - Sakima A. Smith
- OSU Wexner Medical Center, Department of Internal Medicine, Division of Cardiovascular Medicine, The Ohio State University, Columbus, OH 43210, USA;
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Przemysław B. Radwański
- Division of Outcomes and Translational Sciences, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA; (A.M.); (M.T.); (P.B.R.); (C.A.C.); (S.H.)
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Douglas H. Sweet
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA; (C.E.J.); (D.H.S.)
| | - Jörg König
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (J.K.); (M.F.F.)
| | - Martin F. Fromm
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (J.K.); (M.F.F.)
| | - Cynthia A. Carnes
- Division of Outcomes and Translational Sciences, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA; (A.M.); (M.T.); (P.B.R.); (C.A.C.); (S.H.)
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
- Division of Pharmacy Practice and Science, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Shuiying Hu
- Division of Outcomes and Translational Sciences, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA; (A.M.); (M.T.); (P.B.R.); (C.A.C.); (S.H.)
| | - Alex Sparreboom
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (M.E.U.); (E.D.E.); (Y.L.); (K.M.H.); (D.A.G.); (Z.T.); (A.A.G.); (Y.J.); (M.N.); (Q.F.); (X.S.)
| |
Collapse
|
11
|
Larson J, Rich L, Deshmukh A, Judge EC, Liang JJ. Pharmacologic Management for Ventricular Arrhythmias: Overview of Anti-Arrhythmic Drugs. J Clin Med 2022; 11:3233. [PMID: 35683620 PMCID: PMC9181251 DOI: 10.3390/jcm11113233] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/17/2022] [Accepted: 05/28/2022] [Indexed: 01/27/2023] Open
Abstract
Ventricular arrhythmias (Vas) are a life-threatening condition and preventable cause of sudden cardiac death (SCD). With the increased utilization of implantable cardiac defibrillators (ICD), the focus of VA management has shifted toward reduction of morbidity from VAs and ICD therapies. Anti-arrhythmic drugs (AADs) can be an important adjunct therapy in the treatment of recurrent VAs. In the treatment of VAs secondary to structural heart disease, amiodarone remains the most well studied and current guideline-directed pharmacologic therapy. Beta blockers also serve as an important adjunct and are a largely underutilized medication with strong evidentiary support. In patients with defined syndromes in structurally normal hearts, AADs can offer tailored therapies in prevention of SCD and improvement in quality of life. Further clinical trials are warranted to investigate the role of newer therapeutic options and for the direct comparison of established AADs.
Collapse
Affiliation(s)
- John Larson
- Division of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA; (J.L.); (L.R.)
| | - Lucas Rich
- Division of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA; (J.L.); (L.R.)
| | - Amrish Deshmukh
- Division of Cardiovascular Medicine, Cardiac Arrhythmia Service, University of Michigan, Ann Arbor, MI 48109, USA; (A.D.); (E.C.J.)
| | - Erin C. Judge
- Division of Cardiovascular Medicine, Cardiac Arrhythmia Service, University of Michigan, Ann Arbor, MI 48109, USA; (A.D.); (E.C.J.)
| | - Jackson J. Liang
- Division of Cardiovascular Medicine, Cardiac Arrhythmia Service, University of Michigan, Ann Arbor, MI 48109, USA; (A.D.); (E.C.J.)
| |
Collapse
|
12
|
Osadchii OE. Electrocardiographic marker of the cardiac action potential triangulation induced by antiarrhythmic drugs in perfused guinea-pig heart. Exp Physiol 2022; 107:864-878. [PMID: 35561081 DOI: 10.1113/ep090349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 05/09/2022] [Indexed: 11/08/2022]
Abstract
NEW FINDINGS What is the central question of this study? Can triangular appearance of ventricular action potential, indicating proarrhythmic profile of antiarrhythmic agent, be approximated by specific changes on ECG? What is the main finding and its importance? The triangulation of the ventricular action potential seen when antiarrhythmic drugs induce a greater lengthening of the late repolarization compared to the initial repolarization in epicardium, is closely approximated by a greater prolongation of the T wave upslope relative to the interval between the J point and the start of the T wave (the JTstart interval) on ECG. These findings may improve the power of ECG assessments in predicting the drug-induced arrhythmia resulting from slowed phase 3 repolarization. ABSTRACT Antiarrhythmic drugs prescribed to treat atrial fibrillation can occasionally precipitate ventricular tachyarrhythmia through a prominent slowing of the phase 3 repolarization. The latter results in the triangular shape of ventricular action potential, indicating high arrhythmic risks. However, clinically, the utilility of triangulation assessments for predicting arrhythmia is limited owing to the invasive nature of the ventricular action potential recordings. This study examined whether the triangulation effect can be detected indirectly from ECG analysis. Epicardial monophasic action potentials and ECG were simultaneously recorded in perfused guinea-pig hearts. With antiarrhythmics (dofetilide, quinidine, procainamide and flecainide), a prolongation of the initial repolarization seen in the action potential recordings was closely approximated by lengthening of the interval bewteen the J point and the start of the T wave (the JTstart interval) on ECG, whereas a prolongation of the late repolarization was paralleled by widening of the T wave upslope. Dofetilide, quinidine and procainamide induced a prominent slowing of the phase 3 repolarization in epicardium, leading to triangulation of the action potential. These effects were accompanied by a greater prolongation of the T wave upslope compared to the JTstart interval. Flecainide elicited a proportional prolongation of the initial and the late ventricular repolarization, and therefore failed to induce triangulation, based on analysis of both epicardial action potential and ECG profiles. Collectively, these findings suggest that the ratio between the durations of the T wave upslope and the JTstart interval may represent ECG metric of the ventricular action potential triangulation induced by antiarrhythmic drugs. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Oleg E Osadchii
- Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen N, Denmark.,Department of Pharmacology, Kuban State Medical University, Sedin Street 4, Krasnodar, 350063, Russia
| |
Collapse
|
13
|
Tseng AS, Kowlgi GN, DeSimone CV. Antiarrhythmic Drugs for Atrial Fibrillation in the Outpatient Setting: Common Clinical Scenarios and Pearls for the Primary Care Clinician. Mayo Clin Proc 2021; 96:2230-2242. [PMID: 34119307 DOI: 10.1016/j.mayocp.2021.01.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 12/24/2020] [Accepted: 01/15/2021] [Indexed: 10/21/2022]
Abstract
The management of atrial fibrillation (AF) in the outpatient setting has become more complex with the utilization of antiarrhythmic drugs (AADs) and increasing complexity of comorbid conditions. The primary care clinician is critically involved in the pharmacologic management of AF, whether it be direct prescription of AADs or managing potential drug-drug interactions with other medications. In this review, we provide instructive, high-yield clinical scenarios and quick clinical references to increase familiarity and comfort with the use of AADs.
Collapse
Affiliation(s)
- Andrew S Tseng
- Department of Cardiovascular Medicine, Mayo Clinic College of Medicine, Rochester, MN
| | - Gurukripa N Kowlgi
- Department of Cardiovascular Medicine, Mayo Clinic College of Medicine, Rochester, MN
| | | |
Collapse
|
14
|
The Care of Patients With Atrial Fibrillation and Heart Failure. Crit Pathw Cardiol 2021; 20:93-99. [PMID: 32769482 DOI: 10.1097/hpc.0000000000000235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Atrial fibrillation (AF) and heart failure (HF) are the 2 emerging epidemics in global cardiovascular disease. AF remains the most common cardiac arrhythmia, affecting over 33 million adults worldwide, and continues to increase in prevalence as the populations of many nations age. The prevalence of HF also surges, now afflicting 37 million adults globally. Interestingly, these 2 disease processes share many of the same risk factors and stem from many of the same pathophysiologic derangements, with AF occurring in over half of all patients with HF and HF occurring in over one third of all patients with AF. Furthermore, exacerbation of one of these ailments often drives decompensation or compromises therapy of the other, and it has been widely reported that coexistence of AF in patients with HF portends a poorer prognosis. As a result, many clinicians now routinely face the problem of AF in the patient with HF. In this review, we highlight the fundamental pathologic forces embedded in the relationship between AF and HF and then proceed to a discussion on the management of these complex patients with a detailed exploration of the clinical data.
Collapse
|
15
|
Wallman M, Scheuerer S, Martel E, Pairet N, Jirstrand M, Gabrielsson J. An Integrative Approach for Improved Assessment of Cardiovascular Safety Data. J Pharmacol Exp Ther 2021; 377:218-231. [PMID: 33648939 DOI: 10.1124/jpet.120.000348] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 02/22/2021] [Indexed: 11/22/2022] Open
Abstract
Cardiovascular adverse effects in drug development are a major source of compound attrition. Characterization of blood pressure (BP), heart rate (HR), stroke volume (SV), and QT-interval prolongation are therefore necessary in early discovery. It is, however, common practice to analyze these effects independently of each other. High-resolution time courses are collected via telemetric techniques, but only low-resolution data are analyzed and reported. This ignores codependencies among responses (HR, BP, SV, and QT-interval) and separation of system (turnover properties) and drug-specific properties (potencies, efficacies). An analysis of drug exposure-time and high-resolution response-time data of HR and mean arterial blood pressure was performed after acute oral dosing of ivabradine, sildenafil, dofetilide, and pimobendan in Han-Wistar rats. All data were modeled jointly, including different compounds and exposure and response time courses, using a nonlinear mixed-effects approach. Estimated fractional turnover rates [h-1, relative standard error (%RSE) within parentheses] were 9.45 (15), 30.7 (7.8), 3.8 (13), and 0.115 (1.7) for QT, HR, total peripheral resistance, and SV, respectively. Potencies (nM, %RSE within parentheses) were IC 50 = 475 (11), IC 50 = 4.01 (5.4), EC 50 = 50.6 (93), and IC 50 = 47.8 (16), and efficacies (%RSE within parentheses) were I max = 0.944 (1.7), Imax = 1.00 (1.3), E max = 0.195 (9.9), and Imax = 0.745 (4.6) for ivabradine, sildenafil, dofetilide, and pimobendan. Hill parameters were estimated with good precision and below unity, indicating a shallow concentration-response relationship. An equilibrium concentration-biomarker response relationship was predicted and displayed graphically. This analysis demonstrates the utility of a model-based approach integrating data from different studies and compounds for refined preclinical safety margin assessment. SIGNIFICANCE STATEMENT: A model-based approach was proposed utilizing biomarker data on heart rate, blood pressure, and QT-interval. A pharmacodynamic model was developed to improve assessment of high-resolution telemetric cardiovascular safety data driven by different drugs (ivabradine, sildenafil, dofetilide, and pimobondan), wherein system- (turnover rates) and drug-specific parameters (e.g., potencies and efficacies) were sought. The model-predicted equilibrium concentration-biomarker response relationships and was used for safety assessment (predictions of 20% effective concentration, for example) of heart rate, blood pressure, and QT-interval.
Collapse
Affiliation(s)
- Mikael Wallman
- Systems and Data Analysis, Fraunhofer-Chalmers Centre, Gothenburg, Sweden (M.W., M.J.); Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co KG, Biberach, Germany (S.S., E.M., N.P.); and Firma Biopharmacon, Gothenburg, Sweden (J.G.)
| | - Stefan Scheuerer
- Systems and Data Analysis, Fraunhofer-Chalmers Centre, Gothenburg, Sweden (M.W., M.J.); Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co KG, Biberach, Germany (S.S., E.M., N.P.); and Firma Biopharmacon, Gothenburg, Sweden (J.G.)
| | - Eric Martel
- Systems and Data Analysis, Fraunhofer-Chalmers Centre, Gothenburg, Sweden (M.W., M.J.); Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co KG, Biberach, Germany (S.S., E.M., N.P.); and Firma Biopharmacon, Gothenburg, Sweden (J.G.)
| | - Nicolas Pairet
- Systems and Data Analysis, Fraunhofer-Chalmers Centre, Gothenburg, Sweden (M.W., M.J.); Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co KG, Biberach, Germany (S.S., E.M., N.P.); and Firma Biopharmacon, Gothenburg, Sweden (J.G.)
| | - Mats Jirstrand
- Systems and Data Analysis, Fraunhofer-Chalmers Centre, Gothenburg, Sweden (M.W., M.J.); Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co KG, Biberach, Germany (S.S., E.M., N.P.); and Firma Biopharmacon, Gothenburg, Sweden (J.G.)
| | - Johan Gabrielsson
- Systems and Data Analysis, Fraunhofer-Chalmers Centre, Gothenburg, Sweden (M.W., M.J.); Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co KG, Biberach, Germany (S.S., E.M., N.P.); and Firma Biopharmacon, Gothenburg, Sweden (J.G.)
| |
Collapse
|
16
|
Shantha G, Chugh A, Crawford T, Latchamsetty R, Ghanbari H, Ghannam M, Liang J, Batul A, Chung E, Saeed M, Cunnane R, Jongnarangsin K, Bogun F, Pelosi F, Morady F, Oral H. Comparative Efficacy of Dofetilide Versus Amiodarone in Patients With Atrial Fibrillation. JACC Clin Electrophysiol 2021; 7:642-648. [PMID: 33812835 DOI: 10.1016/j.jacep.2020.11.027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 11/16/2020] [Accepted: 11/26/2020] [Indexed: 10/21/2022]
Abstract
OBJECTIVES The study's goal was to compare the efficacy and safety of dofetilide (DOF) versus amiodarone (AMIO) in patients with atrial fibrillation (AF). BACKGROUND Comparative efficacy of DOF versus AMIO in patients with AF has not been well established. In addition, proarrhythmia has been a concern with DOF therapy. METHODS Rhythm control was attempted by using DOF in 657 consecutive patients (mean age 72 ± 9 years; 35% women) with AF (n = 528) or atrial flutter and AF (n = 129) between January 2014 and December 2018. RESULTS DOF was successfully initiated in 573 (87%) of 657 patients, including 510 (89%) with persistent AF and 63 (11%) with paroxysmal AF. During a mean follow-up of 19 ± 7 months, sinus rhythm was maintained in 361 (63%) of the 573 DOF-treated patients. At 12 months, patients on DOF had a similar likelihood of experiencing recurrent atrial arrhythmias compared with the 2,476 consecutive patients treated with AMIO for rhythm control during the study period (37% vs. 39%; p = 0.56). The efficacy of DOF and AMIO was also similar in specific subgroups of patients, including patients >75 years of age, with a low left ventricular ejection fraction, obesity, renal insufficiency, and prior catheter ablation for AF. Among patients with atypical atrial flutter, likelihood of recurrent atrial flutter was similar between the DOF (43 of 108 [40%]) and AMIO (211 of 555 [38%]; p = 0.69) groups. CONCLUSIONS When properly initiated and monitored, DOF has efficacy comparable to that of amiodarone for rhythm control in patients with AF.
Collapse
Affiliation(s)
- Ghanshyam Shantha
- Cardiac Arrhythmia Service, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Aman Chugh
- Cardiac Arrhythmia Service, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Thomas Crawford
- Cardiac Arrhythmia Service, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Rakesh Latchamsetty
- Cardiac Arrhythmia Service, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Hamid Ghanbari
- Cardiac Arrhythmia Service, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Michael Ghannam
- Cardiac Arrhythmia Service, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Jackson Liang
- Cardiac Arrhythmia Service, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Atiqa Batul
- Cardiac Arrhythmia Service, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Eugene Chung
- Cardiac Arrhythmia Service, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Mohammed Saeed
- Cardiac Arrhythmia Service, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Ryan Cunnane
- Cardiac Arrhythmia Service, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Krit Jongnarangsin
- Cardiac Arrhythmia Service, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Frank Bogun
- Cardiac Arrhythmia Service, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Frank Pelosi
- Cardiac Arrhythmia Service, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Fred Morady
- Cardiac Arrhythmia Service, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Hakan Oral
- Cardiac Arrhythmia Service, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA.
| |
Collapse
|
17
|
Kraft M, Büscher A, Wiedmann F, L’hoste Y, Haefeli WE, Frey N, Katus HA, Schmidt C. Current Drug Treatment Strategies for Atrial Fibrillation and TASK-1 Inhibition as an Emerging Novel Therapy Option. Front Pharmacol 2021; 12:638445. [PMID: 33897427 PMCID: PMC8058608 DOI: 10.3389/fphar.2021.638445] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 01/21/2021] [Indexed: 12/19/2022] Open
Abstract
Atrial fibrillation (AF) is the most common sustained arrhythmia with a prevalence of up to 4% and an upwards trend due to demographic changes. It is associated with an increase in mortality and stroke incidences. While stroke risk can be significantly reduced through anticoagulant therapy, adequate treatment of other AF related symptoms remains an unmet medical need in many cases. Two main treatment strategies are available: rate control that modulates ventricular heart rate and prevents tachymyopathy as well as rhythm control that aims to restore and sustain sinus rhythm. Rate control can be achieved through drugs or ablation of the atrioventricular node, rendering the patient pacemaker-dependent. For rhythm control electrical cardioversion and pharmacological cardioversion can be used. While electrical cardioversion requires fasting and sedation of the patient, antiarrhythmic drugs have other limitations. Most antiarrhythmic drugs carry a risk for pro-arrhythmic effects and are contraindicated in patients with structural heart diseases. Furthermore, catheter ablation of pulmonary veins can be performed with its risk of intraprocedural complications and varying success. In recent years TASK-1 has been introduced as a new target for AF therapy. Upregulation of TASK-1 in AF patients contributes to prolongation of the action potential duration. In a porcine model of AF, TASK-1 inhibition by gene therapy or pharmacological compounds induced cardioversion to sinus rhythm. The DOxapram Conversion TO Sinus rhythm (DOCTOS)-Trial will reveal whether doxapram, a potent TASK-1 inhibitor, can be used for acute cardioversion of persistent and paroxysmal AF in patients, potentially leading to a new treatment option for AF.
Collapse
Affiliation(s)
- Manuel Kraft
- Department of Cardiology, University of Heidelberg, Heidelberg, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
- HCR, Heidelberg Center for Heart Rhythm Disorders, University of Heidelberg, Heidelberg, Germany
| | - Antonius Büscher
- Clinic for Cardiology II: Electrophysiology, University Hospital Münster, Münster, Germany
| | - Felix Wiedmann
- Department of Cardiology, University of Heidelberg, Heidelberg, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
- HCR, Heidelberg Center for Heart Rhythm Disorders, University of Heidelberg, Heidelberg, Germany
| | - Yannick L’hoste
- Department of Cardiology, University of Heidelberg, Heidelberg, Germany
- HCR, Heidelberg Center for Heart Rhythm Disorders, University of Heidelberg, Heidelberg, Germany
| | - Walter E. Haefeli
- Department of Clinical Pharmacology and Pharmacoepidemiology, University of Heidelberg, Heidelberg, Germany
| | - Norbert Frey
- Department of Cardiology, University of Heidelberg, Heidelberg, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
- HCR, Heidelberg Center for Heart Rhythm Disorders, University of Heidelberg, Heidelberg, Germany
| | - Hugo A. Katus
- Department of Cardiology, University of Heidelberg, Heidelberg, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
- HCR, Heidelberg Center for Heart Rhythm Disorders, University of Heidelberg, Heidelberg, Germany
| | - Constanze Schmidt
- Department of Cardiology, University of Heidelberg, Heidelberg, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
- HCR, Heidelberg Center for Heart Rhythm Disorders, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
18
|
Árpádffy-Lovas T, Husti Z, Baczkó I, Varró A, Virág L. Different effects of amiodarone and dofetilide on the dispersion of repolarization between well-coupled ventricular and Purkinje fibers 1. Can J Physiol Pharmacol 2020; 99:48-55. [PMID: 32692935 DOI: 10.1139/cjpp-2020-0234] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Increased transmural dispersion of repolarization is an established contributing factor to ventricular tachyarrhythmias. In this study, we evaluated the effect of chronic amiodarone treatment and acute administration of dofetilide in canine cardiac preparations containing electrotonically coupled Purkinje fibers (PFs) and ventricular muscle (VM) and compared the effects to those in uncoupled PF and VM preparations using the conventional microelectrode technique. Dispersion between PFs and VM was inferred from the difference in the respective action potential durations (APDs). In coupled preparations, amiodarone decreased the difference in APDs between PFs and VM, thus decreasing dispersion. In the same preparations, dofetilide increased the dispersion by causing a more pronounced prolongation in PFs. This prolongation was even more emphasized in uncoupled PF preparations, while the effect in VM was the same. In uncoupled preparations, amiodarone elicited no change on the difference in APDs. In conclusion, amiodarone decreased the dispersion between PFs and VM, while dofetilide increased it. The measured difference in APD between cardiac regions may be the affected by electrotonic coupling; thus, studying PFs and VM separately may lead to an over- or underestimation of dispersion.
Collapse
Affiliation(s)
- Tamás Árpádffy-Lovas
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Zoltán Husti
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - István Baczkó
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary.,Department of Pharmacology and Pharmacotherapy, Interdisciplinary Excellence Centre, University of Szeged, Szeged, Hungary.,MTA-SZTE Research Group of Cardiovascular Pharmacology, Szeged, Hungary
| | - András Varró
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary.,Department of Pharmacology and Pharmacotherapy, Interdisciplinary Excellence Centre, University of Szeged, Szeged, Hungary.,MTA-SZTE Research Group of Cardiovascular Pharmacology, Szeged, Hungary
| | - László Virág
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary.,Department of Pharmacology and Pharmacotherapy, Interdisciplinary Excellence Centre, University of Szeged, Szeged, Hungary.,MTA-SZTE Research Group of Cardiovascular Pharmacology, Szeged, Hungary
| |
Collapse
|
19
|
Osadchii OE. Antiarrhythmic drug effects on premature beats are partly determined by prior cardiac activation frequency in perfused guinea-pig heart. Exp Physiol 2020; 105:819-830. [PMID: 32175633 DOI: 10.1113/ep088165] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 03/13/2020] [Indexed: 11/08/2022]
Abstract
NEW FINDINGS What is the central question of this study? Can antiarrhythmic drug effects on repolarization, conduction time and excitation wavelength in premature beats be determined by prior cardiac activation frequency? What is the main finding and its importance? In premature beats induced after a series of cardiac activations at a slow rate, antiarrhythmics prolong repolarization but evoke little or no conduction delay, thus increasing the excitation wavelength, which indicates an antiarrhythmic effect. Fast prior activation rate attenuates prolongation of repolarization, while amplifying the conduction delay induced by drugs, which translates into the reduced excitation wavelength, indicating proarrhythmia. These findings suggest that a sudden increase in heart rate can shape adverse pharmacological profiles in patients with ventricular ectopy. ABSTRACT Antiarrhythmic drugs used to treat atrial fibrillation can occasionally induce ventricular tachyarrhythmia, which is typically precipitated by a premature ectopic beat through a mechanism related, in part, to the shortening of the excitation wavelength (EW). The arrhythmia is likely to occur when a drug induces a reduction, rather than an increase, in the EW of ectopic beats. In this study, I examined whether the arrhythmic drug profile is shaped by the increased cardiac activation rate before ectopic excitation. Ventricular monophasic action potential durations, conduction times and EW values were assessed during programmed stimulations applied at long (S1 -S1 [basic drive cycle length] = 550 ms) and short (S1 -S1 = 200 ms) cycle lengths in perfused guinea-pig hearts. The premature activations were induced with extrastimulus application immediately upon termination of the refractory period. With dofetilide, a class III antiarrhythmic agent, a prolongation in action potential duration and the resulting increase in the EW obtained at S1 -S1 = 550 ms were significantly attenuated at S1 -S1 = 200 ms, in both the regular (S1 ) and the premature (S2 ) beats. With class I antiarrhythmic agents (quinidine, procainamide and flecainide), fast S1 -S1 pacing was found to attenuate the drug-induced increase in action potential duration, while amplifying drug-induced conduction slowing, in both S1 and S2 beats. As a result, although the EW was increased (quinidine and procainamide) or not changed (flecainide) at the long S1 -S1 intervals, it was invariably reduced by these agents at the short S1 -S1 intervals. These findings indicate that the increased heart rate before ectopic activation shapes the arrhythmic profiles by facilitating drug-induced EW reduction.
Collapse
Affiliation(s)
- Oleg E Osadchii
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen N, Denmark.,Department of Health Science and Technology, University of Aalborg, Aalborg, Denmark
| |
Collapse
|
20
|
Osadchii OE. Effects of antiarrhythmics on the electrical restitution in perfused guinea-pig heart are critically determined by the applied cardiac pacing protocol. Exp Physiol 2019; 104:490-504. [PMID: 30758086 DOI: 10.1113/ep087531] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 02/12/2019] [Indexed: 01/11/2023]
Abstract
NEW FINDINGS What is the central question of this study? Are modifications in the restitution of ventricular action potential duration induced by antiarrhythmic drugs the same when assessed with premature extrastimulus application at variable coupling intervals (the standard stimulation protocol) and with steady state pacing at variable rates (the dynamic stimulation protocol)? What is the main finding and its importance? With class I and class III antiarrhythmics, the effects on electrical restitution determined with the standard stimulation protocol dissociate from those obtained during dynamic pacing. These findings indicate a limited value of the electrical restitution assessments based on extrasystolic stimulations alone, as performed in the clinical studies, in estimating the outcomes of antiarrhythmic drug therapies. ABSTRACT A steep slope of the ventricular action potential duration (APD) to diastolic interval (DI) relationships (the electrical restitution) can precipitate tachyarrhythmia, whereas a flattened slope is antiarrhythmic. The derangements in APD restitution responsible for transition of tachycardia to ventricular fibrillation can be assessed with cardiac pacing at progressively increasing rates (the dynamic stimulation protocol). Nevertheless, this method is not used clinically owing to the risk of inducing myocardial ischaemia. Instead, the restitution kinetics is determined with a premature extrastimulus application at variable coupling intervals (the standard stimulation protocol). Whether the two protocols are equivalent in estimating antiarrhythmic drug effects is uncertain. In this study, dofetilide and quinidine, the agents blocking repolarizing K+ currents, increased epicardial APD in perfused guinea-pig hearts, with effects being greater at long vs. short DIs. These changes were more pronounced during dynamic pacing compared to premature extrastimulations. Accordingly, although both agents markedly steepened the dynamic restitution, there was only a marginal increase in the standard restitution slope with dofetilide, and no effect with quinidine. Lidocaine and mexiletine, selective Na+ channel blockers, prolonged the effective refractory period without changing APD, and increased the minimum DI that enabled ventricular capture during extrastimulations. No change in the minimum DI was noted during dynamic pacing. Consequently, although lidocaine and mexiletine reduced the standard restitution slope, they failed to flatten the dynamic restitution. Overall, these findings imply a limited value of the electrical restitution assessments with premature extrastimulations alone in discriminating arrhythmic vs. antiarrhythmic changes during drug therapies.
Collapse
Affiliation(s)
- Oleg E Osadchii
- Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen N, Denmark.,Department of Health Science and Technology, University of Aalborg, Fredrik Bajers Vej 7E, 9220, Aalborg, Denmark
| |
Collapse
|
21
|
Carstensen H, Hesselkilde EZ, Haugaard MM, Flethøj M, Carlson J, Pehrson S, Jespersen T, Platonov PG, Buhl R. Effects of dofetilide and ranolazine on atrial fibrillatory rate in a horse model of acutely induced atrial fibrillation. J Cardiovasc Electrophysiol 2019; 30:596-606. [PMID: 30661267 PMCID: PMC6849868 DOI: 10.1111/jce.13849] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 12/21/2018] [Accepted: 01/04/2019] [Indexed: 12/19/2022]
Abstract
INTRODUCTION The atrial fibrillatory rate is a potential biomarker in the study of antiarrhythmic drug effects on atrial fibrillation (AF). The purpose of this study was to evaluate whether dose-dependent changes in the atrial fibrillatory rate can be monitored on surface electrocardiography (ECG) following treatment with dofetilide, ranolazine, and a combination of the two in an acute model of AF in horses. METHODS AND RESULTS Eight horses were subjected to pacing-induced AF on 4 separate days. Saline (control), dofetilide, ranolazine, or a combination of dofetilide and ranolazine was administered in four incremental doses. Atrial fibrillatory activity was extracted from surface ECGs using spatiotemporal QRST cancellation. The mean atrial fibrillatory rate before drug infusion was 297 ± 27 fpm. Dofetilide reduced the atrial fibrillatory rate following the infusion of low doses (0.89 µg/kg, P < 0.05) and within 5 minutes preceding cardioversion (P < 0.05). Cardioversion with ranolazine was preceded by a reduction in the atrial fibrillatory rate in the last minute (P < 0.05). The combination of drugs reduced the atrial fibrillatory rate in a similar manner to dofetilide used alone. A trend toward a lower atrial fibrillatory rate before drug infusion was found among horses cardioverting on low doses of the drugs. CONCLUSION The atrial fibrillatory rate derived from surface ECGs showed a difference in the mode of action on AF between dofetilide and ranolazine. Dofetilide reduced the atrial fibrillatory rate, whereas ranolazine displayed a cardioverting mechanism that was distinct from a slowing of the fibrillatory process.
Collapse
Affiliation(s)
- Helena Carstensen
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Taastrup, Denmark
| | - Eva Zander Hesselkilde
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Taastrup, Denmark
| | - Maria Mathilde Haugaard
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Taastrup, Denmark
| | - Mette Flethøj
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Taastrup, Denmark
| | - Jonas Carlson
- Department of Cardiology, Clinical Sciences, Lund University, Lund, Sweden
| | - Steen Pehrson
- Department of Cardiology, The Heart Centre, Copenhagen University Hospital, Taastrup, Denmark
| | - Thomas Jespersen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Taastrup, Denmark
| | - Pyotr G Platonov
- Department of Cardiology, Clinical Sciences, Lund University, Lund, Sweden.,Arrhythmia Clinic, Skåne University Hospital, Lund, Sweden
| | - Rikke Buhl
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Taastrup, Denmark
| |
Collapse
|
22
|
Vagos M, van Herck IGM, Sundnes J, Arevalo HJ, Edwards AG, Koivumäki JT. Computational Modeling of Electrophysiology and Pharmacotherapy of Atrial Fibrillation: Recent Advances and Future Challenges. Front Physiol 2018; 9:1221. [PMID: 30233399 PMCID: PMC6131668 DOI: 10.3389/fphys.2018.01221] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 08/13/2018] [Indexed: 12/19/2022] Open
Abstract
The pathophysiology of atrial fibrillation (AF) is broad, with components related to the unique and diverse cellular electrophysiology of atrial myocytes, structural complexity, and heterogeneity of atrial tissue, and pronounced disease-associated remodeling of both cells and tissue. A major challenge for rational design of AF therapy, particularly pharmacotherapy, is integrating these multiscale characteristics to identify approaches that are both efficacious and independent of ventricular contraindications. Computational modeling has long been touted as a basis for achieving such integration in a rapid, economical, and scalable manner. However, computational pipelines for AF-specific drug screening are in their infancy, and while the field is progressing quite rapidly, major challenges remain before computational approaches can fill the role of workhorse in rational design of AF pharmacotherapies. In this review, we briefly detail the unique aspects of AF pathophysiology that determine requirements for compounds targeting AF rhythm control, with emphasis on delimiting mechanisms that promote AF triggers from those providing substrate or supporting reentry. We then describe modeling approaches that have been used to assess the outcomes of drugs acting on established AF targets, as well as on novel promising targets including the ultra-rapidly activating delayed rectifier potassium current, the acetylcholine-activated potassium current and the small conductance calcium-activated potassium channel. Finally, we describe how heterogeneity and variability are being incorporated into AF-specific models, and how these approaches are yielding novel insights into the basic physiology of disease, as well as aiding identification of the important molecular players in the complex AF etiology.
Collapse
Affiliation(s)
- Márcia Vagos
- Computational Physiology Department, Simula Research Laboratory, Lysaker, Norway
- Department of Informatics, University of Oslo, Oslo, Norway
| | - Ilsbeth G. M. van Herck
- Computational Physiology Department, Simula Research Laboratory, Lysaker, Norway
- Department of Informatics, University of Oslo, Oslo, Norway
| | - Joakim Sundnes
- Computational Physiology Department, Simula Research Laboratory, Lysaker, Norway
- Center for Cardiological Innovation, Oslo, Norway
| | - Hermenegild J. Arevalo
- Computational Physiology Department, Simula Research Laboratory, Lysaker, Norway
- Center for Cardiological Innovation, Oslo, Norway
| | - Andrew G. Edwards
- Computational Physiology Department, Simula Research Laboratory, Lysaker, Norway
- Center for Cardiological Innovation, Oslo, Norway
| | - Jussi T. Koivumäki
- BioMediTech Institute and Faculty of Biomedical Sciences and Engineering, Tampere University of Technology, Tampere, Finland
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
23
|
Dofetilide-Associated QT Prolongation: Total Body Weight Versus Adjusted or Ideal Body Weight for Dosing. J Cardiovasc Pharmacol 2018; 72:161-165. [DOI: 10.1097/fjc.0000000000000610] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
24
|
Wolbrette DL, Hussain S, Maraj I, Naccarelli GV. A Quarter of a Century Later: What is Dofetilide’s Clinical Role Today? J Cardiovasc Pharmacol Ther 2018; 24:3-10. [DOI: 10.1177/1074248418784288] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Dofetilide is a class III antiarrhythmic agent approved by the Food and Drug Administration for the conversion of atrial fibrillation and atrial flutter and maintenance of sinus rhythm in symptomatic patients with persistent arrhythmia. Drug trials showed neutral mortality in post–myocardial infarction patients and those with heart failure. This is a review of postmarket data, including real-world efficacy and safety in a variety of populations. Dofetilide has been used off-label with success in patients with paroxysmal atrial fibrillation and atrial flutter, as well as atrial tachycardia and ventricular tachycardia. The real-world acute conversion rate of atrial fibrillation and atrial flutter is higher than that reported in clinical trials. Dofetilide has an acceptable safety profile when initiated (or reloaded) under hospital monitoring and dosed according to creatinine clearance. Dofetilide is well tolerated and a good choice for patients with acceptable renal function and a normal QT interval, especially if atrioventricular nodal blockade needs to be avoided.
Collapse
Affiliation(s)
- Deborah L. Wolbrette
- Heart and Vascular Institute, Penn State University College of Medicine, The Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Sarah Hussain
- Heart and Vascular Institute, Penn State University College of Medicine, The Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Ilir Maraj
- Heart and Vascular Institute, Penn State University College of Medicine, The Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Gerald V. Naccarelli
- Heart and Vascular Institute, Penn State University College of Medicine, The Milton S. Hershey Medical Center, Hershey, PA, USA
| |
Collapse
|
25
|
Pokorney SD, Yen DC, Campbell KB, Allen LaPointe NM, Sheng S, Thomas L, Bahnson TD, Daubert JP, Picini JP, Jackson KP, Thomas KL, Al-Khatib SM. Dofetilide dose reductions and discontinuations in women compared with men. Heart Rhythm 2018; 15:478-484. [DOI: 10.1016/j.hrthm.2018.01.027] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2017] [Indexed: 11/30/2022]
|
26
|
Kopljar I, Hermans AN, Teisman A, Gallacher DJ, Lu HR. Impact of calcium-sensitive dyes on the beating properties and pharmacological responses of human iPS-derived cardiomyocytes using the calcium transient assay. J Pharmacol Toxicol Methods 2018; 91:80-86. [PMID: 29421525 DOI: 10.1016/j.vascn.2018.02.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 01/22/2018] [Accepted: 02/04/2018] [Indexed: 01/08/2023]
Abstract
INTRODUCTION Calcium-based screening of hiPS-CMs is a useful preclinical safety evaluation platform with the ability to generate robust signals that facilitates high-throughput screening and data analysis. However, due to the potential inherent toxicities, it is important to understand potential effects of different calcium-sensitive dyes on the hiPS-CMs model. METHODS We compared three calcium-sensitive fluorescence dyes (Cal520, ACTOne and Calcium 5) for their impact on the variability, the beating properties and the pharmacological responses of hiPS-CMs using the Hamamatsu FDSS/μCell imaging platform. Direct effects of three dyes on the electrophysiological properties of hiPS-CMs were evaluated with the multi-electrode array (MEA) Axion Maestro platform. RESULTS We propose a specific experimental protocol for each dye which gives the most optimal assay conditions to minimize variability and possible adverse effects. We showed that Cal520 had the smallest effect on hiPS-CMs together with the longest-lasting stable amplitude signal (up to 4 h). Although all dyes had a (minor) acute effect on hiPS-CMs, in the form of reduced beat rate and prolonged field potential duration, the selection of the dye did not influence the pharmacological response of four cardioactive drugs (dofetilide, moxifloxacin, nimodipine and isoprenaline). DISCUSSION In conclusion, we have documented that different calcium sensitive dyes have only minor direct (acute) effects on hiPS-CMs with Cal520 showing the least effects and the longest lasting signal amplitude. Importantly, drug-induced pharmacological responses in hiPS-CMs were comparable between the three dyes. These findings should help further improve the robustness of the hiPS-CMs-based calcium transient assay as a predictive, preclinical cardiac safety evaluation tool.
Collapse
Affiliation(s)
- Ivan Kopljar
- Global Safety Pharmacology, Preclinical Safety & Development, Discovery Sciences, Janssen Research & Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium.
| | - An N Hermans
- Global Safety Pharmacology, Preclinical Safety & Development, Discovery Sciences, Janssen Research & Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Ard Teisman
- Global Safety Pharmacology, Preclinical Safety & Development, Discovery Sciences, Janssen Research & Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - David J Gallacher
- Global Safety Pharmacology, Preclinical Safety & Development, Discovery Sciences, Janssen Research & Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Hua Rong Lu
- Global Safety Pharmacology, Preclinical Safety & Development, Discovery Sciences, Janssen Research & Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium.
| |
Collapse
|
27
|
Arrhythmogenic drugs can amplify spatial heterogeneities in the electrical restitution in perfused guinea-pig heart: An evidence from assessments of monophasic action potential durations and JT intervals. PLoS One 2018; 13:e0191514. [PMID: 29352276 PMCID: PMC5774816 DOI: 10.1371/journal.pone.0191514] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 01/06/2018] [Indexed: 01/01/2023] Open
Abstract
Non-uniform shortening of the action potential duration (APD90) in different myocardial regions upon heart rate acceleration can set abnormal repolarization gradients and promote arrhythmia. This study examined whether spatial heterogeneities in APD90 restitution can be amplified by drugs with clinically proved proarrhythmic potential (dofetilide, quinidine, procainamide, and flecainide) and, if so, whether these effects can translate to the appropriate changes of the ECG metrics of ventricular repolarization, such as JT intervals. In isolated, perfused guinea-pig heart preparations, monophasic action potentials and volume-conducted ECG were recorded at progressively increased pacing rates. The APD90 measured at distinct ventricular sites, as well as the JTpeak and JTend values were plotted as a function of preceding diastolic interval, and the maximum slopes of the restitution curves were determined at baseline and upon drug administration. Dofetilide, quinidine, and procainamide reverse rate-dependently prolonged APD90 and steepened the restitution curve, with effects being greater at the endocardium than epicardium, and in the right ventricular (RV) vs. the left ventricular (LV) chamber. The restitution slope was increased to a greater extent for the JTend vs. the JTpeak interval. In contrast, flecainide reduced the APD90 restitution slope at LV epicardium without producing effect at LV endocardium and RV epicardium, and reduced the JTpeak restitution slope without changing the JTend restitution. Nevertheless, with all agents, these effects translated to the amplified epicardial-to-endocardial and the LV-to-RV non-uniformities in APD90 restitution, paralleled by the increased JTend vs. JTpeak difference in the restitution slope. In summary, these findings suggest that arrhythmic drug profiles are partly attributable to the accentuated regional heterogeneities in APD90 restitution, which can be indirectly determined through ECG assessments of the JTend vs. JTpeak dynamics at variable pacing rates.
Collapse
|
28
|
Yan S, Huang P, Wang Y, Zeng X, Zhang Y. The Venom of Ornithoctonus huwena affect the electrophysiological stability of neonatal rat ventricular myocytes by inhibiting sodium, potassium and calcium current. Channels (Austin) 2018. [PMID: 29532737 PMCID: PMC5972801 DOI: 10.1080/19336950.2018.1449497] [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] [Indexed: 01/14/2023] Open
Abstract
Spider venoms are known to contain various toxins that are used as an effective means to capture their prey or to defend themselves against predators. An investigation of the properties of Ornithoctonus huwena (O.huwena) crude venom found that the venom can block neuromuscular transmission of isolated mouse phrenic nerve-diaphragm and sciatic nerve-sartorius preparations. However, little is known about its electrophysiological effects on cardiac myocytes. In this study, electrophysiological activities of ventricular myocytes were detected by 100 μg/mL venom of O.huwena, and whole cell patch-clamp technique was used to study the acute effects of the venom on action potential (AP), sodium current (INa), potassium currents (IKr, IKs, Ito1 and IK1) and L-type calcium current (ICaL). The results indicated that the venom prolongs APD90 in a frequency-dependent manner in isolated neonatal rat ventricular myocytes. 100 μg/mL venom inhibited 72.3 ± 3.6% INa current, 58.3 ± 4.2% summit current and 54 ± 6.1% the end current of IKr, and 65 ± 3.3% ICaL current, yet, didn't have obvious effect on IKs, Ito1 and IK1 currents. In conclusion, the O.huwena venom represented a multifaceted pharmacological profile. It contains abundant of cardiac channel antagonists and might be valuable tools for investigation of both channels and anti- arrhythmic therapy development.
Collapse
Affiliation(s)
- Sha Yan
- a Department of Dermatology, Xiangya Hospital , Central South University , Changsha , China.,b Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province , Central South University , Changsha , Hunan , China
| | - Pengfei Huang
- c The Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences , Hunan Normal University , Changsha , P. R. China.,d The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development , College of Life Sciences, Hunan Normal University , Changsha , China
| | - Ying Wang
- c The Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences , Hunan Normal University , Changsha , P. R. China.,d The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development , College of Life Sciences, Hunan Normal University , Changsha , China
| | - Xiongzhi Zeng
- c The Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences , Hunan Normal University , Changsha , P. R. China.,d The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development , College of Life Sciences, Hunan Normal University , Changsha , China
| | - Yiya Zhang
- a Department of Dermatology, Xiangya Hospital , Central South University , Changsha , China.,b Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province , Central South University , Changsha , Hunan , China.,c The Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences , Hunan Normal University , Changsha , P. R. China.,d The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development , College of Life Sciences, Hunan Normal University , Changsha , China
| |
Collapse
|
29
|
Nguyen N, Nguyen W, Nguyenton B, Ratchada P, Page G, Miller PE, Ghetti A, Abi-Gerges N. Adult Human Primary Cardiomyocyte-Based Model for the Simultaneous Prediction of Drug-Induced Inotropic and Pro-arrhythmia Risk. Front Physiol 2017; 8:1073. [PMID: 29311989 PMCID: PMC5742250 DOI: 10.3389/fphys.2017.01073] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 12/06/2017] [Indexed: 12/19/2022] Open
Abstract
Cardiac safety remains the leading cause of drug development discontinuation. We developed a human cardiomyocyte-based model that has the potential to provide a predictive preclinical approach for simultaneously predicting drug-induced inotropic and pro-arrhythmia risk. Methods: Adult human primary cardiomyocytes from ethically consented organ donors were used to measure contractility transients. We used measures of changes in contractility parameters as markers to infer both drug-induced inotropic effect (sarcomere shortening) and pro-arrhythmia (aftercontraction, AC); contractility escape (CE); time to 90% relaxation (TR90). We addressed the clinical relevance of this approach by evaluating the effects of 23 torsadogenic and 10 non-torsadogenic drugs. Each drug was tested separately at four multiples of the free effective therapeutic plasma concentration (fETPC). Results: Human cardiomyocyte-based model differentiated between torsadogenic and non-torsadogenic drugs. For example, dofetilide, a torsadogenic drug, caused ACs and increased TR90 starting at 10-fold the fETPC, while CE events were observed at the highest multiple of fETPC (100-fold). Verapamil, a non-torsadogenic drug, did not change TR90 and induced no AC or CE up to the highest multiple of fETPCs tested in this study (222-fold). When drug pro-arrhythmic activity was evaluated at 10-fold of the fETPC, AC parameter had excellent assay sensitivity and specificity values of 96 and 100%, respectively. This high predictivity supports the translational safety potential of this preparation and of the selected marker. The data demonstrate that human cardiomyocytes could also identify drugs associated with inotropic effects. hERG channel blockers, like dofetilide, had no effects on sarcomere shortening, while multi-ion channel blockers, like verapamil, inhibited sarcomere shortening. Conclusions: Isolated adult human primary cardiomyocytes can simultaneously predict risks associated with inotropic activity and pro-arrhythmia and may enable the generation of reliable and predictive data for assessing human cardiotoxicity at an early stage in drug discovery.
Collapse
Affiliation(s)
| | | | | | | | - Guy Page
- AnaBios Corporation, San Diego, CA, United States
| | | | - Andre Ghetti
- AnaBios Corporation, San Diego, CA, United States
| | | |
Collapse
|
30
|
Rose PG, Seabury R, Cwikla G. Acute Hepatocellular Jaundice After Dofetilide Initiation: A Case Report. Hosp Pharm 2017; 53:55-58. [PMID: 29434388 DOI: 10.1177/0018578717738079] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Dofetilide's hepatotoxicity is not well described. In this case report, we describe acute hepatocellular jaundice related to dofetilide use in a 33-year-old male being treated for atrial fibrillation. Both viral and ischemic causes of hepatocellular damage were ruled out as unlikely in this case. This case report outlines a rare yet probable report of idiosyncratic dofetilide-induced liver injury.
Collapse
Affiliation(s)
- Patrick Gary Rose
- State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Robert Seabury
- State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Gregory Cwikla
- State University of New York Upstate Medical University, Syracuse, NY, USA
| |
Collapse
|
31
|
Cho JH, Youn SJ, Moore JC, Kyriakakis R, Vekstein C, Militello M, Poe SM, Wolski K, Tchou PJ, Varma N, Niebauer MJ, Bhargava M, Saliba WI, Wazni OM, Lindsay BD, Wilkoff BL, Chung MK. Safety of Oral Dofetilide Reloading for Treatment of Atrial Arrhythmias. Circ Arrhythm Electrophysiol 2017; 10:CIRCEP.117.005333. [PMID: 29038104 DOI: 10.1161/circep.117.005333] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Accepted: 08/23/2017] [Indexed: 11/16/2022]
Abstract
BACKGROUND Although dofetilide labeling states that the drug must be initiated or reinitiated with continuous electrocardiographic monitoring and in the presence of trained personnel, the risks of dofetilide reloading justifying repeat hospitalization have not been investigated. METHODS AND RESULTS Patients admitted for dofetilide reloading for atrial arrhythmias were retrospectively reviewed. The need for dose adjustment and the incidence of torsades de pointes (TdP) were identified. The incidence of TdP in dofetilide reloading was compared with patients admitted for dofetilide initial loading. Of 138 patients admitted for dofetilide reloading for atrial arrhythmias, 102 were reloaded at a previously tolerated dose, 30 with a higher dose from a previously tolerated dose and 2 at a lower dose; prior dosage was unknown in 4 patients. Dose adjustment or discontinuation was required in 44 patients (31.9%). No TdP occurred in the same dose reloading group, but TdP occurred in 2 patients admitted to increase dofetilide dosage (0% versus 6.7%; P=0.050). Dofetilide dose adjustment or discontinuation was required in 30 of 102 patients (29.4%) reloaded at a previously tolerated dose and in 11 of 30 patients (36.7%) admitted for an increase in dose. CONCLUSIONS Although no TdP occurred in patients admitted to reload dofetilide at the same dose as previously tolerated, dosage adjustments or discontinuation was frequent and support the need for hospitalization for dofetilide reloading. Patients admitted for reloading with a higher dose tended to be at higher risk for TdP than patients reloaded at a prior tolerated dose.
Collapse
Affiliation(s)
- Jae Hyung Cho
- From the Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic, OH. Current address for Dr Cho: Cedars-Sinai Heart Institute, Los Angeles, CA. Current address for Dr Youn: Department of Internal Medicine, Cleveland Clinic, OH. Current address for Dr Moore: Minneapolis Heart Institute, Abbott Northwestern Hospital, MN. Current address for R. Kyriakakis: College of Medicine, Medical University of South Carolina, Charleston. Current address for C. Vekstein: Dana-Farber Cancer Institute, Boston, MA. Current address for M. Militello: Pharmacy Department, Cleveland Clinic, OH
| | - So Jin Youn
- From the Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic, OH. Current address for Dr Cho: Cedars-Sinai Heart Institute, Los Angeles, CA. Current address for Dr Youn: Department of Internal Medicine, Cleveland Clinic, OH. Current address for Dr Moore: Minneapolis Heart Institute, Abbott Northwestern Hospital, MN. Current address for R. Kyriakakis: College of Medicine, Medical University of South Carolina, Charleston. Current address for C. Vekstein: Dana-Farber Cancer Institute, Boston, MA. Current address for M. Militello: Pharmacy Department, Cleveland Clinic, OH
| | - JoEllyn C Moore
- From the Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic, OH. Current address for Dr Cho: Cedars-Sinai Heart Institute, Los Angeles, CA. Current address for Dr Youn: Department of Internal Medicine, Cleveland Clinic, OH. Current address for Dr Moore: Minneapolis Heart Institute, Abbott Northwestern Hospital, MN. Current address for R. Kyriakakis: College of Medicine, Medical University of South Carolina, Charleston. Current address for C. Vekstein: Dana-Farber Cancer Institute, Boston, MA. Current address for M. Militello: Pharmacy Department, Cleveland Clinic, OH
| | - Roxanne Kyriakakis
- From the Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic, OH. Current address for Dr Cho: Cedars-Sinai Heart Institute, Los Angeles, CA. Current address for Dr Youn: Department of Internal Medicine, Cleveland Clinic, OH. Current address for Dr Moore: Minneapolis Heart Institute, Abbott Northwestern Hospital, MN. Current address for R. Kyriakakis: College of Medicine, Medical University of South Carolina, Charleston. Current address for C. Vekstein: Dana-Farber Cancer Institute, Boston, MA. Current address for M. Militello: Pharmacy Department, Cleveland Clinic, OH
| | - Carolyn Vekstein
- From the Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic, OH. Current address for Dr Cho: Cedars-Sinai Heart Institute, Los Angeles, CA. Current address for Dr Youn: Department of Internal Medicine, Cleveland Clinic, OH. Current address for Dr Moore: Minneapolis Heart Institute, Abbott Northwestern Hospital, MN. Current address for R. Kyriakakis: College of Medicine, Medical University of South Carolina, Charleston. Current address for C. Vekstein: Dana-Farber Cancer Institute, Boston, MA. Current address for M. Militello: Pharmacy Department, Cleveland Clinic, OH
| | - Michael Militello
- From the Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic, OH. Current address for Dr Cho: Cedars-Sinai Heart Institute, Los Angeles, CA. Current address for Dr Youn: Department of Internal Medicine, Cleveland Clinic, OH. Current address for Dr Moore: Minneapolis Heart Institute, Abbott Northwestern Hospital, MN. Current address for R. Kyriakakis: College of Medicine, Medical University of South Carolina, Charleston. Current address for C. Vekstein: Dana-Farber Cancer Institute, Boston, MA. Current address for M. Militello: Pharmacy Department, Cleveland Clinic, OH
| | - Stacy M Poe
- From the Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic, OH. Current address for Dr Cho: Cedars-Sinai Heart Institute, Los Angeles, CA. Current address for Dr Youn: Department of Internal Medicine, Cleveland Clinic, OH. Current address for Dr Moore: Minneapolis Heart Institute, Abbott Northwestern Hospital, MN. Current address for R. Kyriakakis: College of Medicine, Medical University of South Carolina, Charleston. Current address for C. Vekstein: Dana-Farber Cancer Institute, Boston, MA. Current address for M. Militello: Pharmacy Department, Cleveland Clinic, OH
| | - Kathy Wolski
- From the Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic, OH. Current address for Dr Cho: Cedars-Sinai Heart Institute, Los Angeles, CA. Current address for Dr Youn: Department of Internal Medicine, Cleveland Clinic, OH. Current address for Dr Moore: Minneapolis Heart Institute, Abbott Northwestern Hospital, MN. Current address for R. Kyriakakis: College of Medicine, Medical University of South Carolina, Charleston. Current address for C. Vekstein: Dana-Farber Cancer Institute, Boston, MA. Current address for M. Militello: Pharmacy Department, Cleveland Clinic, OH
| | - Patrick J Tchou
- From the Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic, OH. Current address for Dr Cho: Cedars-Sinai Heart Institute, Los Angeles, CA. Current address for Dr Youn: Department of Internal Medicine, Cleveland Clinic, OH. Current address for Dr Moore: Minneapolis Heart Institute, Abbott Northwestern Hospital, MN. Current address for R. Kyriakakis: College of Medicine, Medical University of South Carolina, Charleston. Current address for C. Vekstein: Dana-Farber Cancer Institute, Boston, MA. Current address for M. Militello: Pharmacy Department, Cleveland Clinic, OH
| | - Niraj Varma
- From the Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic, OH. Current address for Dr Cho: Cedars-Sinai Heart Institute, Los Angeles, CA. Current address for Dr Youn: Department of Internal Medicine, Cleveland Clinic, OH. Current address for Dr Moore: Minneapolis Heart Institute, Abbott Northwestern Hospital, MN. Current address for R. Kyriakakis: College of Medicine, Medical University of South Carolina, Charleston. Current address for C. Vekstein: Dana-Farber Cancer Institute, Boston, MA. Current address for M. Militello: Pharmacy Department, Cleveland Clinic, OH
| | - Mark J Niebauer
- From the Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic, OH. Current address for Dr Cho: Cedars-Sinai Heart Institute, Los Angeles, CA. Current address for Dr Youn: Department of Internal Medicine, Cleveland Clinic, OH. Current address for Dr Moore: Minneapolis Heart Institute, Abbott Northwestern Hospital, MN. Current address for R. Kyriakakis: College of Medicine, Medical University of South Carolina, Charleston. Current address for C. Vekstein: Dana-Farber Cancer Institute, Boston, MA. Current address for M. Militello: Pharmacy Department, Cleveland Clinic, OH
| | - Mandeep Bhargava
- From the Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic, OH. Current address for Dr Cho: Cedars-Sinai Heart Institute, Los Angeles, CA. Current address for Dr Youn: Department of Internal Medicine, Cleveland Clinic, OH. Current address for Dr Moore: Minneapolis Heart Institute, Abbott Northwestern Hospital, MN. Current address for R. Kyriakakis: College of Medicine, Medical University of South Carolina, Charleston. Current address for C. Vekstein: Dana-Farber Cancer Institute, Boston, MA. Current address for M. Militello: Pharmacy Department, Cleveland Clinic, OH
| | - Walid I Saliba
- From the Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic, OH. Current address for Dr Cho: Cedars-Sinai Heart Institute, Los Angeles, CA. Current address for Dr Youn: Department of Internal Medicine, Cleveland Clinic, OH. Current address for Dr Moore: Minneapolis Heart Institute, Abbott Northwestern Hospital, MN. Current address for R. Kyriakakis: College of Medicine, Medical University of South Carolina, Charleston. Current address for C. Vekstein: Dana-Farber Cancer Institute, Boston, MA. Current address for M. Militello: Pharmacy Department, Cleveland Clinic, OH
| | - Oussama M Wazni
- From the Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic, OH. Current address for Dr Cho: Cedars-Sinai Heart Institute, Los Angeles, CA. Current address for Dr Youn: Department of Internal Medicine, Cleveland Clinic, OH. Current address for Dr Moore: Minneapolis Heart Institute, Abbott Northwestern Hospital, MN. Current address for R. Kyriakakis: College of Medicine, Medical University of South Carolina, Charleston. Current address for C. Vekstein: Dana-Farber Cancer Institute, Boston, MA. Current address for M. Militello: Pharmacy Department, Cleveland Clinic, OH
| | - Bruce D Lindsay
- From the Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic, OH. Current address for Dr Cho: Cedars-Sinai Heart Institute, Los Angeles, CA. Current address for Dr Youn: Department of Internal Medicine, Cleveland Clinic, OH. Current address for Dr Moore: Minneapolis Heart Institute, Abbott Northwestern Hospital, MN. Current address for R. Kyriakakis: College of Medicine, Medical University of South Carolina, Charleston. Current address for C. Vekstein: Dana-Farber Cancer Institute, Boston, MA. Current address for M. Militello: Pharmacy Department, Cleveland Clinic, OH
| | - Bruce L Wilkoff
- From the Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic, OH. Current address for Dr Cho: Cedars-Sinai Heart Institute, Los Angeles, CA. Current address for Dr Youn: Department of Internal Medicine, Cleveland Clinic, OH. Current address for Dr Moore: Minneapolis Heart Institute, Abbott Northwestern Hospital, MN. Current address for R. Kyriakakis: College of Medicine, Medical University of South Carolina, Charleston. Current address for C. Vekstein: Dana-Farber Cancer Institute, Boston, MA. Current address for M. Militello: Pharmacy Department, Cleveland Clinic, OH
| | - Mina K Chung
- From the Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic, OH. Current address for Dr Cho: Cedars-Sinai Heart Institute, Los Angeles, CA. Current address for Dr Youn: Department of Internal Medicine, Cleveland Clinic, OH. Current address for Dr Moore: Minneapolis Heart Institute, Abbott Northwestern Hospital, MN. Current address for R. Kyriakakis: College of Medicine, Medical University of South Carolina, Charleston. Current address for C. Vekstein: Dana-Farber Cancer Institute, Boston, MA. Current address for M. Militello: Pharmacy Department, Cleveland Clinic, OH.
| |
Collapse
|
32
|
Abstract
PURPOSE OF REVIEW Ventricular tachycardia occurrence in implantable cardioverter defibrillator (ICD) patients may result in shock delivery and is associated with increased morbidity and mortality. In addition, shocks may have deleterious mechanical and psychological effects. Prevention of ventricular tachycardia (VT) recurrence with the use of antiarrhythmic drugs or catheter ablation may be warranted. Antiarrhythmic drugs are limited by incomplete efficacy and an unfavorable adverse effect profile. Catheter ablation can be effective but acute complications and long-term VT recurrence risk necessitating repeat ablation should be recognized. A shared clinical decision process accounting for patients' cardiac status, comorbidities, and goals of care is often required. RECENT FINDINGS There are four published randomized trials of catheter ablation for sustained monomorphic VT (SMVT) in the setting of ischemic heart disease; there are no randomized studies for non-ischemic ventricular substrates. The most recent trial is the VANISH trial which randomly allocated patients with ICD, prior infarction, and SMVT despite first-line antiarrhythmic drug therapy to catheter ablation or more aggressive antiarrhythmic drug therapy. During 28 months of follow-up, catheter ablation resulted in a 28% relative risk reduction in the composite endpoint of death, VT storm, and appropriate ICD shock (p = 0.04). In a subgroup analysis, patients having VT despite amiodarone had better outcomes with ablation as compared to increasing amiodarone dose or adding mexiletine. There is evidence for the effectiveness of both catheter ablation and antiarrhythmic drug therapy for patients with myocardial infarction, an implantable defibrillator, and VT. If sotalol is ineffective in suppressing VT, either catheter ablation or initiation of amiodarone is a reasonable option. If VT occurs despite amiodarone therapy, there is evidence that catheter ablation is superior to administration of more aggressive antiarrhythmic drug therapy. Early catheter ablation may be appropriate in some clinical situations such as patients presenting with relatively slow VT below ICD detection, electrical storms, hemodynamically stable VT, or in very selected patients with left ventricular assist devices. The optimal first-line suppressive therapy for VT, after ICD implantation and appropriate programming, remains to be determined. Thus far, there has not been a randomized controlled trial to compare catheter ablation to antiarrhythmic drug therapy as a first-line treatment; the VANISH-2 study has been initiated as a pilot to examine this question.
Collapse
Affiliation(s)
- Amir AbdelWahab
- QEII Health Sciences Centre, Room 2501 B/F Halifax Infirmary 1796 Summer Street, Halifax, NS, B3H 3A7, Canada
| | - John Sapp
- QEII Health Sciences Centre, Room 2501 B/F Halifax Infirmary 1796 Summer Street, Halifax, NS, B3H 3A7, Canada.
| |
Collapse
|
33
|
VanderLugt JT, Bon C, Knuth D, Schreiber R, Ruff MD. Pharmacokinetics and Bioequivalence of Branded and Generic Formulations of Dofetilide 0.5-mg Capsules After Single-Dose Administration in Healthy Subjects. Clin Pharmacol Drug Dev 2017; 7:311-318. [PMID: 28800211 DOI: 10.1002/cpdd.371] [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: 02/02/2017] [Accepted: 05/23/2017] [Indexed: 11/10/2022]
Abstract
Class III antiarrhythmics are preferred therapy for managing atrial fibrillation/flutter. Dofetilide 0.5-mg capsules were US Food and Drug Administration (FDA) approved in 1999 to treat atrial fibrillation/flutter. Bioequivalence of generic dofetilide is important for treating arrhythmias because drug concentrations must be consistent to maintain normal sinus rhythm. Generic dofetilide 0.5-mg capsule pharmacokinetics were compared with branded product in 2 open-label, 2-way crossover, single-dose studies - 1 study each in fasted and fed healthy subjects. Blood samples were collected before and up to 48 hours after dosing. Safety was assessed by tabulating adverse events and vital signs. Seventy-three subjects were enrolled; 59 completed the studies. In fasted subjects, the 90% confidence intervals (CIs) for generic dofetilide 0.5 mg versus the reference formulation were 0.996-1.026 for the area under the plasma concentration-time curve from 0 to infinity (AUC) and 0.974-1.066 for the maximum observed concentration (Cmax ). In fed subjects, the 90%CIs for AUC and Cmax were 0.988-1.015 and 0.928-0.992, respectively. All ratios were within the FDA-established bioequivalence range. Twenty-six subjects experienced 37 adverse events (generic, 15; reference, 22); all but 1 were mild or moderate in severity. Generic dofetilide 0.5-mg capsules can be considered bioequivalent to the reference product.
Collapse
Affiliation(s)
| | - Charles Bon
- Biostudy Solutions, LLC, Wilmington, NC, USA
| | | | | | | |
Collapse
|
34
|
Modeling Atrial Fibrillation using Human Embryonic Stem Cell-Derived Atrial Tissue. Sci Rep 2017; 7:5268. [PMID: 28706272 PMCID: PMC5509676 DOI: 10.1038/s41598-017-05652-y] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 06/01/2017] [Indexed: 12/31/2022] Open
Abstract
Since current experimental models of Atrial Fibrillation (AF) have significant limitations, we used human embryonic stem cells (hESCs) to generate an atrial-specific tissue model of AF for pharmacologic testing. We generated atrial-like cardiomyocytes (CMs) from hESCs which preferentially expressed atrial-specific genes, and had shorter action potential (AP) durations compared to ventricular-like CMs. We then generated confluent atrial-like CM sheets and interrogated them using optical mapping techniques. Atrial-like CM sheets (~1 cm in diameter) showed uniform AP propagation, and rapid re-entrant rotor patterns, as seen in AF could be induced. Anti-arrhythmic drugs were tested on single atrial-like CMs and cell sheets. Flecainide profoundly slowed upstroke velocity without affecting AP duration, leading to reduced conduction velocities (CVs), curvatures and cycle lengths of rotors, consistent with increased rotor organization and expansion. By contrast, consistent with block of rapid delayed rectifier K+ currents (Ikr) and AP prolongation in isolated atrial-like CMs, dofetilide prolonged APs and reduced cycle lengths of rotors in cell sheets without affecting CV. In conclusion, using our hESC-derived atrial CM preparations, we demonstrate that flecainide and dofetilide modulate reentrant arrhythmogenic rotor activation patterns in a manner that helps explain their efficacy in treating and preventing AF.
Collapse
|
35
|
Osadchii OE. Assessments of the QT/QRS restitution in perfused guinea-pig heart can discriminate safe and arrhythmogenic drugs. J Pharmacol Toxicol Methods 2017; 87:27-37. [PMID: 28552278 DOI: 10.1016/j.vascn.2017.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 04/25/2017] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Drug-induced arrhythmia remains a matter of serious clinical concern, partly due to low prognostic value of currently available arrhythmic biomarkers. METHODS This study examined whether arrhythmogenic risks can be predicted through assessments of the rate adaptation of QT interval, ventricular effective refractory period (ERP), or the QT/QRS ratio, in perfused guinea-pig hearts. RESULTS When the maximum restitution slope was taken as a metric of proarrhythmia, neither QT interval nor ERP measurements at progressively increased pacing rates were found to fully discriminate arrhythmogenic drugs (dofetilide, quinidine, flecainide, and procainamide) from those recognized as safe antiarrhythmics (lidocaine and mexiletine). For example, the slope of QT restitution was increased by dofetilide and quinidine, but remained unchanged by flecainide, procainamide, lidocaine, and mexiletine. With ERP rate adaptation, even though the restitution slope was increased by dofetilide, all class I agents reduced the slope value independently of their safety profile. The QRS measurements revealed variable drug effects, ranging from significant use-dependent conduction slowing (flecainide, quinidine, and procainamide) to only modest increase in QRS (lidocaine and mexiletine), or no change at all (dofetilide). However, with the QT/QRS rate adaptation, the restitution slope was significantly increased by all agents which have been reported to produce proarrhythmic effects (dofetilide, quinidine, flecainide, and procainamide), but not changed by lidocaine and mexiletine. DISCUSSION These findings suggest that the slope of the QT/QRS rate adaptation can be considered as a novel electrophysiological biomarker in predicting potential arrhythmic risks associated with pharmacotherapy in cardiac patients.
Collapse
Affiliation(s)
- Oleg E Osadchii
- Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen N, Denmark; Department of Health Science and Technology, University of Aalborg, Fredrik Bajers Vej 7E, 9220 Aalborg, Denmark.
| |
Collapse
|
36
|
Liu G, Xue X, Gao C, Huang J, Qi D, Zhang Y, Dong JZ, Ma CS, Yan GX. Synergistic Effect of Dofetilide and Mexiletine on Prevention of Atrial Fibrillation. J Am Heart Assoc 2017; 6:JAHA.117.005482. [PMID: 28522677 PMCID: PMC5524094 DOI: 10.1161/jaha.117.005482] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background Although atrial fibrillation (AF) is the most common abnormal heart rhythm and its prevalence continues to rise, there is a marked paucity of effective and safe antiarrhythmic drugs for AF. This study was done to test whether combined use of dofetilide and mexiletine exhibits not only a synergistic effect on AF suppression but also a safer profile in drug‐induced ventricular proarrhythmias. Methods and Results The effects of dofetilide plus mexiletine on atrial effective refractory period (ERP), AF inducibility, QT, and QT‐related ventricular arrhythmias were studied using the isolated arterially perfused rabbit atrial and ventricular wedge preparations. Dofetilide or mexiletine alone mildly to moderately prolonged atrial ERP, but their combined use produced a markedly rate‐dependent increase in atrial ERP. Dofetilide (3 nmol/L) plus mexiletine (10 μmol/L) increased the ERP by 28.2% from 72.2±5.7 to 92.8±5.9 ms (n=9, P<0.01) at a pacing rate of 0.5 Hz and by 94.5% from 91.7±5.2 to 178.3±12.0 ms (n=9, P<0.01) at 3.3 Hz. Dofetilide plus mexiletine strongly suppressed AF inducibility. On the other hand, dofetilide at 10 nmol/L produced marked QT and Tp‐e prolongation, steeper QT‐BCL and Tp‐e‐BCL slopes, and induced early afterdepolarizations and torsade de pointes in the ventricular wedges. Mexiletine at 10 μmol/L reduced dofetilide‐induced QT and Tp‐e prolongation, QT‐BCL and Tp‐e‐BCL slopes, and abolished early afterdepolarizations and torsade de pointes. Conclusions In rabbits, combined use of dofetilide and mexiletine not only synergistically increases atrial ERP and effectively suppresses AF inducibility, but also markedly reduces QT liability and torsade de pointes risk posed by dofetilide alone.
Collapse
Affiliation(s)
- Guizhi Liu
- First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaolin Xue
- Department of Cardiology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Chuanyu Gao
- Department of Cardiology, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Jiaqi Huang
- Lankenau Institute for Medical Research, Wynnewood, PA
| | - Datun Qi
- Department of Cardiology, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Yanzhou Zhang
- First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jian-Zeng Dong
- First Affiliated Hospital of Zhengzhou University, Zhengzhou, China .,Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Chang-Sheng Ma
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Gan-Xin Yan
- Department of Cardiology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China .,Lankenau Institute for Medical Research, Wynnewood, PA.,Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
37
|
Use of dofetilide in adult patients with atrial arrhythmias and congenital heart disease: A PACES collaborative study. Heart Rhythm 2016; 13:2034-9. [DOI: 10.1016/j.hrthm.2016.07.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Indexed: 11/17/2022]
|
38
|
Hieger MA, Maskell KF, Moss MJ, Powell SW, Cumpston KL. Dofetilide in Overdose: A Case Series from Poison Center Data. Cardiovasc Toxicol 2016; 17:368-371. [PMID: 27565970 DOI: 10.1007/s12012-016-9384-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Dofetilide is a class III antiarrhythmic used for treating atrial dysrhythmias. Though its adverse effects are well described in routine use, very little is known about dofetilide toxicity in overdose. This is a retrospective case series of consecutive patients reported to our poison center after dofetilide overdose. Twenty-seven cases were included. Seventeen patients were treated at a healthcare facility, and of these, eight were admitted. Twenty-one patients took one extra capsule, four took someone else's medication, one took three extra capsules, and one had a large intentional overdose. Ten patients had co-ingestants reported, including three QT-prolonging agents. No one required cardioversion, defibrillation, CPR, or overdrive pacing. The patient who reported taking 90 times his usual dose in suicide attempt was the only patient to have significant clinical effects. He experienced an 8-beat run of non-sustained ventricular tachycardia, frequent multifocal PVCs, and ventricular bigeminy. He received magnesium sulfate and potassium chloride supplementation. In this series, unintentional small overdoses did not result in significant clinical effects and were often managed successfully at home, despite the fact that information showing a single capsule can cause torsades. This study is limited by its small sample size, retrospective design, and reliance on incomplete information.
Collapse
Affiliation(s)
- M A Hieger
- Division of Clinical Toxicology, Department of Emergency Medicine, VCU Medical Center, 1250 E. Marshall Street, 2nd Floor, Richmond, VA, 23298, USA.
- Virginia Poison Center, 830 E. Main Street, Suite 300, PO Box 980522, Richmond, VA, 23298, USA.
- Department of Emergency Medicine, VCU Medical Center, Richmond, VA, 23298, USA.
| | - K F Maskell
- Division of Clinical Toxicology, Department of Emergency Medicine, VCU Medical Center, 1250 E. Marshall Street, 2nd Floor, Richmond, VA, 23298, USA
- Virginia Poison Center, 830 E. Main Street, Suite 300, PO Box 980522, Richmond, VA, 23298, USA
- Department of Emergency Medicine, VCU Medical Center, Richmond, VA, 23298, USA
| | - M J Moss
- Department of Emergency Medicine, VCU Medical Center, Richmond, VA, 23298, USA
| | - S W Powell
- Department of Emergency Medicine, VCU Medical Center, Richmond, VA, 23298, USA
| | - K L Cumpston
- Division of Clinical Toxicology, Department of Emergency Medicine, VCU Medical Center, 1250 E. Marshall Street, 2nd Floor, Richmond, VA, 23298, USA
- Virginia Poison Center, 830 E. Main Street, Suite 300, PO Box 980522, Richmond, VA, 23298, USA
- Department of Emergency Medicine, VCU Medical Center, Richmond, VA, 23298, USA
| |
Collapse
|
39
|
Vogel EA, Billups SJ, Herner SJ, Delate T. Renal Drug Dosing. Effectiveness of Outpatient Pharmacist-Based vs. Prescriber-Based Clinical Decision Support Systems. Appl Clin Inform 2016; 7:731-44. [PMID: 27466041 DOI: 10.4338/aci-2016-01-ra-0010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 06/28/2016] [Indexed: 01/06/2023] Open
Abstract
OBJECTIVE The purpose of this study was to compare the effectiveness of an outpatient renal dose adjustment alert via a computerized provider order entry (CPOE) clinical decision support system (CDSS) versus a CDSS with alerts made to dispensing pharmacists. METHODS This was a retrospective analysis of patients with renal impairment and 30 medications that are contraindicated or require dose-adjustment in such patients. The primary outcome was the rate of renal dosing errors for study medications that were dispensed between August and December 2013, when a pharmacist-based CDSS was in place, versus August through December 2014, when a prescriber-based CDSS was in place. A dosing error was defined as a prescription for one of the study medications dispensed to a patient where the medication was contraindicated or improperly dosed based on the patient's renal function. The denominator was all prescriptions for the study medications dispensed during each respective study period. RESULTS During the pharmacist- and prescriber-based CDSS study periods, 49,054 and 50,678 prescriptions, respectively, were dispensed for one of the included medications. Of these, 878 (1.8%) and 758 (1.5%) prescriptions were dispensed to patients with renal impairment in the respective study periods. Patients in each group were similar with respect to age, sex, and renal function stage. Overall, the five-month error rate was 0.38%. Error rates were similar between the two groups: 0.36% and 0.40% in the pharmacist- and prescriber-based CDSS, respectively (p=0.523). The medication with the highest error rate was dofetilide (0.51% overall) while the medications with the lowest error rate were dabigatran, fondaparinux, and spironolactone (0.00% overall). CONCLUSIONS Prescriber- and pharmacist-based CDSS provided comparable, low rates of potential medication errors. Future studies should be undertaken to examine patient benefits of the prescriber-based CDSS.
Collapse
Affiliation(s)
| | | | | | - Thomas Delate
- Thomas Delate, PhD, MS, Kaiser Permanente Colorado Pharmacy Dept., 16601 E. Centretech Pkwy, Aurora, CO 80011 USA, Phone: 303-739-3538;,
| |
Collapse
|
40
|
Waks JW, Zimetbaum P. Antiarrhythmic Drug Therapy for Rhythm Control in Atrial Fibrillation. J Cardiovasc Pharmacol Ther 2016; 22:3-19. [DOI: 10.1177/1074248416651722] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Atrial fibrillation (AF) is the most common sustained cardiac arrhythmia and affects over 33 million people worldwide. AF is associated with stroke and systemic thromboembolism, unpleasant symptoms and reduced quality of life, heart failure, and increased mortality, and treatment of AF and its complications are associated with significant cost. Antiarrhythmic drugs (AADs) can suppress AF, allowing long-term maintenance of sinus rhythm, and have the potential to relieve symptoms and reverse or prevent adverse effects associated with AF. However, large randomized controlled studies evaluating use of AADs have not demonstrated a clear benefit to maintaining sinus rhythm, and AADs often have significant limitations, including a modest rate of overall success at maintaining sinus rhythm, frequent side effects, and potentially life-threatening toxicities. Although some of the currently available AADs have been available for almost 100 years, better tolerated and more efficacious AADs have recently been developed both for long-term maintenance of sinus rhythm and for chemical cardioversion of AF to sinus rhythm. Advances in automated AF detection with cardiac implantable electronic devices have suggested that AADs might be useful for suppressing AF to allow safe discontinuation of anticoagulation in select patients who are in sinus rhythm for prolonged periods of time. AADs may also have synergistic effects with catheter ablation of AF. This review summarizes the pharmacology and clinical use of currently available AADs for treatment of AF and discusses novel AADs and future directions for rhythm control in AF.
Collapse
Affiliation(s)
- Jonathan W. Waks
- Harvard-Thorndike Electrophysiology Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Peter Zimetbaum
- Harvard-Thorndike Electrophysiology Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
41
|
Mykytsey A, Bauman JL, Razminia M, Zheutlin T, Wang T, Saleem M, Leal S, Kehoe RF. Observations on the Safety and Effectiveness of Dofetilide in Patients With Paroxysmal Atrial Fibrillation and Normal Left Ventricular Function. J Cardiovasc Pharmacol Ther 2016; 12:36-43. [PMID: 17495256 DOI: 10.1177/1074248407299272] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Dofetilide is currently recommended as second-tier therapy to maintain sinus rhythm in patients with paroxysmal atrial fibrillation (PAF) and normal left ventricular function, yet limited data support this recommendation. We examined the safety and efficacy of dofetilide in this setting through a retrospective chart review. We evaluated patients who had symptomatic PAF, normal left ventricular function, and no significant valvular disease. The end points were complete suppression of symptomatic PAF and subjective symptomatic improvement with dofetilide treatment. Over a 3-year period, 34 patients who had failed previous antiarrhythmic therapy were included. Of these, 3 discontinued dofetilide treatment before discharge. Of the remaining 31 who continued treatment after discharge, it was eventually discontinued in 13. At 12 months, symptomatic improvement was observed in 18 of 31 patients, 6 of whom remained asymptomatic. Treatment with dofetilide in this study was successful in less than 1 in 5 patients. Despite careful precautions, serious proarrhythmias, the major limiting side effect of dofetilide, still occurred during long-term follow-up.
Collapse
Affiliation(s)
- Andrew Mykytsey
- Advocate Illinois Masonic Medical Center, 3000 N. Halsted Street, Chicago, IL 60612, USA
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Dofetilide: Electrophysiologic Effect, Efficacy, and Safety in Patients with Cardiac Arrhythmias. Card Electrophysiol Clin 2016; 8:423-36. [PMID: 27261832 DOI: 10.1016/j.ccep.2016.02.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Dofetilide is a class III antiarrhythmic agent with a selective blockade of rapid component of delayed rectifier potassium current (IKr). Dofetilide was found to be safe in patients after myocardial infarction and those with congestive heart failure and left ventricular systolic dysfunction (ejection fraction of less than 35%). An important adverse effect of dofetilide is its potential proarrhythmic risk of ventricular tachyarrhythmias, mostly torsades de pointes. Because dofetilide has about an 80% renal excretion, dose adjustment is required in patients with impaired renal function. Dofetilide should not be given or discontinued if the QTc is greater than 500 ms.
Collapse
|
43
|
Huang HD, Waks JW, Steinhaus DA, Zimetbaum P. Magnitude of increase in QTc interval after initiation of dofetilide in patients with persistent atrial fibrillation is associated with increased rates of pharmacological cardioversion and long-term freedom from recurrent atrial fibrillation. Heart Rhythm 2016; 13:1410-7. [PMID: 26921760 DOI: 10.1016/j.hrthm.2016.02.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Indexed: 10/22/2022]
Abstract
BACKGROUND Dofetilide is a class III antiarrhythmic drug approved for the treatment of atrial fibrillation (AF). Dofetilide-induced corrected QT (QTc) interval prolongation is a surrogate for the degree of drug effect, but the relationships between drug-induced QTc interval prolongation, pharmacological cardioversion (PCV), and freedom from recurrent AF are unclear. OBJECTIVE The purpose of this study was to assess associations between QTc interval change during dofetilide initiation and PCV and long-term AF recurrence. METHODS We performed retrospective analyses of a prospective cohort of patients with AF admitted for dofetilide initiation between 2001 and 2014. Clinical characteristics and electrocardiographic variables were assessed. We evaluated outcomes of successful PCV in patients with persistent AF and time to recurrence of AF in patients with paroxysmal and persistent AF. RESULTS During the study, 243 patients with persistent AF and 176 patients with paroxysmal AF initiated dofetilide. PCV occurred in 93/243 (41.7%) patients with persistent AF. After multivariable adjustment, QTc interval change was associated with PCV (adjusted odds ratio 1.21; P = .003 per 10-ms QTc increase). Inhospital QTc interval change was associated with long-term freedom from AF in patients with persistent AF (adjusted hazard ratio 0.92; P = .011 at 4 years per 10-ms QTc increase), but not in patients with paroxysmal AF. In patients with persistent AF, PCV was also associated with long-term freedom from recurrent AF (adjusted hazard ratio 0.62; P = .009 at 4 years). CONCLUSION The magnitude of QTc interval prolongation during dofetilide initiation is an independent predictor of successful PCV and long-term freedom from arrhythmia in patients with persistent AF. QTc interval change had no association with AF recurrence in patients with paroxysmal AF, suggesting that different mechanisms of arrhythmogenesis may be operant in different AF types.
Collapse
Affiliation(s)
- Henry D Huang
- Harvard-Thorndike Electrophysiology Institute, Division of Cardiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Jonathan W Waks
- Harvard-Thorndike Electrophysiology Institute, Division of Cardiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Daniel A Steinhaus
- Harvard-Thorndike Electrophysiology Institute, Division of Cardiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Peter Zimetbaum
- Harvard-Thorndike Electrophysiology Institute, Division of Cardiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
44
|
Chronotropic Modulation of the Source-Sink Relationship of Sinoatrial-Atrial Impulse Conduction and Its Significance to Initiation of AF: A One-Dimensional Model Study. BIOMED RESEARCH INTERNATIONAL 2015; 2015:496418. [PMID: 26229960 PMCID: PMC4502286 DOI: 10.1155/2015/496418] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Revised: 02/25/2015] [Accepted: 03/10/2015] [Indexed: 01/15/2023]
Abstract
Initiation and maintenance of atrial fibrillation (AF) is often associated with pharmacologically or pathologically induced bradycardic states. Even drugs specifically developed in order to counteract cardiac arrhythmias often combine their action with bradycardia and, in turn, with development of AF, via still largely unknown mechanisms. This study aims to simulate action potential (AP) conduction between sinoatrial node (SAN) and atrial cells, either arranged in cell pairs or in a one-dimensional strand, where the relative amount of SAN membrane is made varying, in turn, with junctional resistance. The source-sink relationship between the two membrane types is studied in control conditions and under different simulated chronotropic interventions, in order to define a safety factor for pacemaker-to-atrial AP conduction (SASF) for each treatment. Whereas antiarrhythmic-like interventions which involve downregulation of calcium channels or of calcium handling decrease SASF, the simulation of Ivabradine administration does so to a lesser extent. Particularly interesting is the increase of SASF observed when downregulation G Kr, which simulates the administration of class III antiarrhythmic agents and is likely sustained by an increase in I CaL. Also, the increase in SASF is accompanied by a decreased conduction delay and a better entrainment of repolarization, which is significant to anti-AF strategies.
Collapse
|
45
|
Meijborg VM, Chauveau S, Janse MJ, Anyukhovsky EP, Danilo PR, Rosen MR, Opthof T, Coronel R. Interventricular dispersion in repolarization causes bifid T waves in dogs with dofetilide-induced long QT syndrome. Heart Rhythm 2015; 12:1343-51. [DOI: 10.1016/j.hrthm.2015.02.026] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Indexed: 11/27/2022]
|
46
|
Agusala K, Oesterle A, Kulkarni C, Caprio T, Subacius H, Passman R. Risk prediction for adverse events during initiation of sotalol and dofetilide for the treatment of atrial fibrillation. PACING AND CLINICAL ELECTROPHYSIOLOGY: PACE 2015; 38:490-8. [PMID: 25626340 DOI: 10.1111/pace.12586] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Revised: 11/24/2014] [Accepted: 12/21/2014] [Indexed: 11/29/2022]
Abstract
BACKGROUND Inpatient antiarrhythmic drug initiation for atrial fibrillation is mandated for dofetilide (DF) and is often performed for sotalol (SL), particularly if proarrhythmia risk factors are present. Whether low-risk patients can be identified to safely allow outpatient initiation is unknown. METHODS A single-center retrospective cohort study was performed on patients initiated with DF or SL. Risk factors for adverse events (AEs), defined as any arrhythmia or electrocardiogram change requiring dose reduction or cessation, were identified. RESULTS Of 329 patients, 227 (69%) received SL and 102 (31%) DF. The cohort had a mean age of 63 ± 13 years; 70% of patients were male and had a baseline QTc of 440 ± 37 ms. A total of 105 AEs occurred in 92 patients: QTc prolongation or ventricular tachyarrhythmia in 70 patients (67% of AEs), bradyarrhythmias in 35 patients (33% of AEs), with some experiencing both AE types. Ventricular arrhythmias were seen in 23 patients (7%) and torsades de pointes in one (0.3%). Total AE rates were similar between drugs (P = 0.09); however, DF patients had more QTc prolongation or ventricular arrhythmias (P = 0.001). In SL patients, there were no predictors for QTc prolongation or ventricular proarrhythmia. In DF patients, higher baseline QTc interval (odds ratio = 1.64/25 ms, P = 0.01) was an independent predictor of QTc prolongation or ventricular proarrhythmias. For patients without proarrhythmia risk factors, overall AE rate was 26%. CONCLUSIONS In conclusion, AEs are common during DF and SL initiation but rarely severe in hospitalized inpatients. Baseline QTc predicts AEs for DF patients only and AE are common even in "low-risk" patients. These results support in-hospital drug initiation for all DF and SL patients.
Collapse
Affiliation(s)
- Kartik Agusala
- From Northwestern University, Feinberg School of Medicine, Chicago, Illinois
| | | | | | | | | | | |
Collapse
|
47
|
Frendl G, Sodickson AC, Chung MK, Waldo AL, Gersh BJ, Tisdale JE, Calkins H, Aranki S, Kaneko T, Cassivi S, Smith SC, Darbar D, Wee JO, Waddell TK, Amar D, Adler D. 2014 AATS guidelines for the prevention and management of perioperative atrial fibrillation and flutter for thoracic surgical procedures. J Thorac Cardiovasc Surg 2014; 148:e153-93. [PMID: 25129609 PMCID: PMC4454633 DOI: 10.1016/j.jtcvs.2014.06.036] [Citation(s) in RCA: 196] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Accepted: 06/10/2014] [Indexed: 02/06/2023]
Affiliation(s)
- Gyorgy Frendl
- Department of Anesthesiology, Perioperative Critical Care and Pain Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass.
| | - Alissa C Sodickson
- Department of Anesthesiology, Perioperative Critical Care and Pain Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass
| | - Mina K Chung
- Department of Cardiovascular Medicine, Heart and Vascular Institute, Department of Molecular Cardiology, Lerner Research Institute Cleveland Clinic, Lerner College of Medicine of Case Western Reserve University Cleveland Clinic, Cleveland, Ohio
| | - Albert L Waldo
- Division of Cardiovascular Medicine, Department of Medicine, Case Western Reserve University, Cleveland, Ohio; Harrington Heart & Vascular Institute, University Hospitals Case Medical Center, Cleveland, Ohio
| | - Bernard J Gersh
- Division of Cardiovascular Diseases and Internal Medicine, Department of Medicine, Mayo Clinic College of Medicine, Rochester, Minn
| | - James E Tisdale
- Department of Pharmacy Practice, College of Pharmacy, Purdue University and Indiana University School of Medicine, Indianapolis, Ind
| | - Hugh Calkins
- Department of Medicine, Cardiac Arrhythmia Service, Johns Hopkins University, Baltimore, Md
| | - Sary Aranki
- Division of Cardiac Surgery, Department of Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass
| | - Tsuyoshi Kaneko
- Division of Cardiac Surgery, Department of Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass
| | - Stephen Cassivi
- Division of Thoracic Surgery, Department of Surgery, Mayo Clinic College of Medicine, Rochester, Minn
| | - Sidney C Smith
- Center for Heart and Vascular Care, Department of Medicine, University of North Carolina, Chapel Hill, NC
| | - Dawood Darbar
- Division of Cardiovascular Medicine, Department of Medicine, Arrhythmia Service, Vanderbilt University School of Medicine, Nashville, Tenn
| | - Jon O Wee
- Division of Thoracic Surgery, Department of Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass
| | - Thomas K Waddell
- Division of Thoracic Surgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - David Amar
- Memorial Sloan-Kettering Cancer Center, Department of Anesthesiology and Critical Care Medicine, New York, NY
| | - Dale Adler
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass
| |
Collapse
|
48
|
Yang T, Chun YW, Stroud DM, Mosley JD, Knollmann BC, Hong C, Roden DM. Screening for acute IKr block is insufficient to detect torsades de pointes liability: role of late sodium current. Circulation 2014; 130:224-34. [PMID: 24895457 DOI: 10.1161/circulationaha.113.007765] [Citation(s) in RCA: 121] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND New drugs are routinely screened for IKr blocking properties thought to predict QT prolonging and arrhythmogenic liability. However, recent data suggest that chronic (hours) drug exposure to phosphoinositide 3-kinase inhibitors used in cancer can prolong QT by inhibiting potassium currents and increasing late sodium current (INa-L) in cardiomyocytes. We tested the extent to which IKr blockers with known QT liability generate arrhythmias through this pathway. METHODS AND RESULTS Acute exposure to dofetilide, an IKr blocker without other recognized electropharmacologic actions, produced no change in ion currents or action potentials in adult mouse cardiomyocytes, which lack IKr. By contrast, 2 to 48 hours of exposure to the drug generated arrhythmogenic afterdepolarizations and ≥15-fold increases in INa-L. Including phosphatidylinositol 3,4,5-trisphosphate, a downstream effector for the phosphoinositide 3-kinase pathway, in the pipette inhibited these effects. INa-L was also increased, and inhibitable by phosphatidylinositol 3,4,5-trisphosphate, with hours of dofetilide exposure in human-induced pluripotent stem cell-derived cardiomyocytes and in Chinese hamster ovary cells transfected with SCN5A, encoding sodium current. Cardiomyocytes from dofetilide-treated mice similarly demonstrated increased INa-L and afterdepolarizations. Other agents with variable IKr-blocking potencies and arrhythmia liability produced a range of effects on INa-L, from marked increases (E-4031, d-sotalol, thioridazine, and erythromycin) to little or no effect (haloperidol, moxifloxacin, and verapamil). CONCLUSIONS Some but not all drugs designated as arrhythmogenic IKr blockers can generate arrhythmias by augmenting INa-L through the phosphoinositide 3-kinase pathway. These data identify a potential mechanism for individual susceptibility to proarrhythmia and highlight the need for a new paradigm to screen drugs for QT prolonging and arrhythmogenic liability.
Collapse
Affiliation(s)
- Tao Yang
- From the Vanderbilt University School of Medicine, Nashville, TN
| | - Young Wook Chun
- From the Vanderbilt University School of Medicine, Nashville, TN
| | - Dina M Stroud
- From the Vanderbilt University School of Medicine, Nashville, TN
| | | | | | - Charles Hong
- From the Vanderbilt University School of Medicine, Nashville, TN
| | - Dan M Roden
- From the Vanderbilt University School of Medicine, Nashville, TN.
| |
Collapse
|
49
|
Jaiswal A, Goldbarg S. Dofetilide induced torsade de pointes: mechanism, risk factors and management strategies. Indian Heart J 2014; 66:640-8. [PMID: 25634399 DOI: 10.1016/j.ihj.2013.12.021] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Accepted: 12/04/2013] [Indexed: 11/29/2022] Open
Abstract
Dofetilide is an effective antiarrhythmic agent for conversion of atrial fibrillation and atrial flutter as well as maintenance of sinus rhythm in appropriately selected patients. However, as with other antiarrhythmic agents, proarrhythmia is a known adverse effect. The risk of dofetilide induced torsade de pointes (Tdp) is low when used with strict dosing criteria guided by renal function, QT interval and concomitant drug therapy. Benefit from dofetilide use must be individualized and weighed against the side effects and the role of other available treatment options. In this review, we discuss the underlying mechanism, risk factors and precautionary measures to avoid dofetilide induced QT prolongation and ventricular tachycardia/Tdp. We suggest a scheme for the management of QT prolongation, ventricular arrhythmia and Tdp as well.
Collapse
Affiliation(s)
- Abhishek Jaiswal
- Division of Cardiology, New York Hospital Queens/Weill Medical College of Cornell University, Flushing, NY 11355, USA.
| | - Seth Goldbarg
- Division of Cardiology, New York Hospital Queens/Weill Medical College of Cornell University, Flushing, NY 11355, USA
| |
Collapse
|
50
|
Abstract
Atrial fibrillation is the most common arrhythmia affecting patients today. Disease prevalence is increasing at an alarming rate worldwide, and is associated with often catastrophic and costly consequences, including heart failure, syncope, dementia, and stroke. Therapies including anticoagulants, anti-arrhythmic medications, devices, and non-pharmacologic procedures in the last 30 years have improved patients' functionality with the disease. Nonetheless, it remains imperative that further research into AF epidemiology, genetics, detection, and treatments continues to push forward rapidly as the worldwide population ages dramatically over the next 20 years.
Collapse
Affiliation(s)
- Thomas M. Munger
- Heart Rhythm Services, Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN 55905, USA;
| | - Li-Qun Wu
- Department of Cardiology, Rui Jin Hospital, Shanghai Jiao Tong University of Medicine, Shanghai 200025, China;
| | - Win K. Shen
- Division of Cardiovascular Diseases, Mayo Clinic, Phoenix, AZ 85054, USA.
| |
Collapse
|