1
|
Yamauchi M, Otsuki K, Sato K, Nagahama M, Tanabe J, Inagaki M. Vortioxetine Can Be Used Safely to Treat Depression After Implantable Cardioverter Defibrillator Implantation for Congenital Long QT Syndrome: A Case Report. Neuropsychopharmacol Rep 2025; 45:e70017. [PMID: 40265337 PMCID: PMC12015558 DOI: 10.1002/npr2.70017] [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: 12/24/2024] [Revised: 04/09/2025] [Accepted: 04/15/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND Vortioxetine, an antidepressant, acts on multiple 5-hydroxytryptamine receptors in addition to inhibiting the 5-hydroxytryptamine transporter. Currently, data on the effect of vortioxetine on the QT interval are limited. To the best of our knowledge, whether vortioxetine can safely be used for congenital long QT syndrome (LQTS) has not been reported. CASE PRESENTATION We here present the case of a 50-year-old Japanese woman who was rushed to our hospital because of a sudden cardiopulmonary arrest. Her corrected QT interval was prolonged to 535 ms. After a thorough examination in the cardiology department, she was diagnosed with congenital LQTS and an implantable cardioverter defibrillator (ICD) implanted. Thereafter, she gradually developed severe depression because of stress associated with ICD implantation. Mirtazapine induced daytime somnolence and duloxetine was relatively ineffective, prompting replacement with vortioxetine at 20 mg daily, which relieved her depression. During the treatment period, her corrected QT interval did not exceed 500 ms, beyond which the risk of cardiac events increases. To our knowledge, this is the first report of the safe use of vortioxetine in a patient with depression comorbid with congenital LQTS. CONCLUSIONS After ICD implantation, a patient's mental status should be monitored carefully. Our findings suggest that vortioxetine may be a safe treatment option for depression associated with congenital LQTS.
Collapse
Affiliation(s)
- Makio Yamauchi
- Department of Psychiatry, Faculty of MedicineShimane UniversityIzumoJapan
| | - Koji Otsuki
- Department of Psychiatry, Faculty of MedicineShimane UniversityIzumoJapan
| | - Kouhei Sato
- Department of Psychiatry, Faculty of MedicineShimane UniversityIzumoJapan
| | - Michiharu Nagahama
- Department of Psychiatry, Faculty of MedicineShimane UniversityIzumoJapan
| | - Junya Tanabe
- Division of Cardiology, Faculty of MedicineShimane UniversityIzumoJapan
| | - Masatoshi Inagaki
- Department of Psychiatry, Faculty of MedicineShimane UniversityIzumoJapan
| |
Collapse
|
2
|
Hancox JC, Copeland CS, Harmer SC, Henderson G. New synthetic cannabinoids and the potential for cardiac arrhythmia risk. JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY PLUS 2023; 6:100049. [PMID: 38143960 PMCID: PMC10739592 DOI: 10.1016/j.jmccpl.2023.100049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/14/2023] [Accepted: 09/14/2023] [Indexed: 12/26/2023]
Abstract
Synthetic cannabinoid receptor agonists (SCRAs) have been associated with QT interval prolongation. Limited preclinical information on SCRA effects on cardiac electrogenesis results from the rapid emergence of new compounds and restricted research availability. We used two machine-learning-based tools to evaluate seven novel SCRAs' interaction potential with the hERG potassium channel, an important drug antitarget. Five SCRAs were predicted to have the ability to block the hERG channel by both prediction tools; ADB-FUBIATA was predicted to be a strong hERG blocker. ADB-5Br-INACA and ADB-4en-PINACA showed varied predictions. These findings highlight potentially proarrhythmic hERG block by novel SCRAs, necessitating detailed safety evaluations.
Collapse
Affiliation(s)
- Jules C. Hancox
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University Walk, Bristol BS8 1TD, UK
| | - Caroline S. Copeland
- Institute of Pharmaceutical Science, King's College London, UK
- Centre for Pharmaceutical Medicine Research, King's College London, UK
| | - Stephen C. Harmer
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University Walk, Bristol BS8 1TD, UK
| | - Graeme Henderson
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University Walk, Bristol BS8 1TD, UK
| |
Collapse
|
3
|
Kimura K, Narita H, Imai H, Akiyama H, Ishikawa S, Sawagashira R, Isoyama T, Nohara M, Kawamura M, Kono Y, Saito T, Kusumi I. Cardiovascular adverse reactions associated with escitalopram in patients with underlying cardiovascular diseases: a systematic review and meta-analysis. Front Psychiatry 2023; 14:1248397. [PMID: 37810602 PMCID: PMC10556499 DOI: 10.3389/fpsyt.2023.1248397] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 09/11/2023] [Indexed: 10/10/2023] Open
Abstract
Background Despite the anticipated efficacy of escitalopram in treating depression and anxiety in individuals with preexisting cardiovascular conditions, persistent concerns regarding its adverse effects have emerged. In this systematic review, we aimed to evaluate the cardiovascular safety profile of escitalopram compared with that of placebo in patients with underlying cardiovascular disease. Methods We used a predefined search strategy in PubMed, Cochrane Central Register of Controlled Trials, Embase, International Clinical Trials Registry Platform, and ClinicalTrials.gov to identify studies evaluating adverse cardiovascular reactions to escitalopram in patients with underlying cardiovascular disease. Randomized controlled trials (RCTs) that provided results on cardiovascular safety outcomes were included. Two independent reviewers screened the abstracts and full texts of the individual studies. The risk of bias was assessed using version 2 of the Cochrane risk-of-bias tool for randomized trials. The certainty of evidence was assessed using the Grading of Recommendations, Assessment, Development, and Evaluation approach. Results The primary outcomes were the frequency of major adverse cardiovascular events (MACE), QTc prolongation, and discontinuation of study medication. We identified 5 RCTs with 773 participants who met the inclusion criteria. Escitalopram was not associated with significantly increased risk of MACE (risk ratio [RR] = 1.85; 95% confidence interval [CI] 0.80 to 4.26; I2 0%; 5 RCTs; n = 773, moderate certainty of evidence), discontinuation of study medication (RR = 1.03; 95% CI 0.84-1.26; I2 0%; 5 RCTs; n = 773, low certainty of evidence), and QTc prolongation (RR = 1.20; 95% CI 0.76-1.90; I2 0%; 4 RCTs; n = 646, low certainty of evidence). Conclusion Escitalopram does not significantly increase the risk of cardiovascular adverse reactions compared with placebo in patients with underlying cardiovascular disease. However, the presence of wide CIs and the limited number of included studies highlight the need for further studies with larger sample sizes to enhance the precision and reliability of these findings.Systematic review registration: International Prospective Register of Systematic Reviews [CRD42022298181].
Collapse
Affiliation(s)
- Kenichi Kimura
- Department of Psychiatry and Neurology, Hokkaido University Hospital, Sapporo, Japan
| | - Hisashi Narita
- Department of Psychiatry and Neurology, Hokkaido University Hospital, Sapporo, Japan
| | - Hissei Imai
- Department of Health Promotion and Human Behavior, Graduate School of Medicine/School of Public Health, Kyoto University, Kyoto, Japan
| | - Hisashi Akiyama
- Department of Psychiatry and Neurology, Hokkaido University Hospital, Sapporo, Japan
| | - Shuhei Ishikawa
- Department of Psychiatry and Neurology, Hokkaido University Hospital, Sapporo, Japan
| | - Ryo Sawagashira
- Department of Physiology, Hokkaido University School of Medicine, Sapporo, Japan
- Creative Research Institute, Hokkaido University, Sapporo, Japan
| | - Tomoyuki Isoyama
- Department of Psychiatry and Neurology, Hokkaido University Hospital, Sapporo, Japan
| | - Mariko Nohara
- Department of Psychiatry and Neurology, Hokkaido University Hospital, Sapporo, Japan
| | - Michiyo Kawamura
- Medical Sciences Group, Research Support Division, Hokkaido University Library, Sapporo, Japan
| | - Yukari Kono
- Medical Sciences Group, Research Support Division, Hokkaido University Library, Sapporo, Japan
| | - Takuya Saito
- Department of Child and Adolescent Psychiatry, Hokkaido University Hospital, Sapporo, Japan
| | - Ichiro Kusumi
- Department of Psychiatry and Neurology, Hokkaido University Hospital, Sapporo, Japan
| |
Collapse
|
4
|
El Harchi A, Hancox JC. hERG agonists pose challenges to web-based machine learning methods for prediction of drug-hERG channel interaction. J Pharmacol Toxicol Methods 2023; 123:107293. [PMID: 37468081 DOI: 10.1016/j.vascn.2023.107293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 05/23/2023] [Accepted: 07/12/2023] [Indexed: 07/21/2023]
Abstract
Pharmacological blockade of the IKr channel (hERG) by diverse drugs in clinical use is associated with the Long QT Syndrome that can lead to life threatening arrhythmia. Various computational tools including machine learning models (MLM) for the prediction of hERG inhibition have been developed to facilitate the throughput screening of drugs in development and optimise thus the prediction of hERG liabilities. The use of MLM relies on large libraries of training compounds for the quantitative structure-activity relationship (QSAR) modelling of hERG inhibition. The focus on inhibition omits potential effects of hERG channel agonist molecules and their associated QT shortening risk. It is instructive, therefore, to consider how known hERG agonists are handled by MLM. Here, two highly developed online computational tools for the prediction of hERG liability, Pred-hERG and HergSPred were probed for their ability to detect hERG activator drug molecules as hERG interactors. In total, 73 hERG blockers were tested with both computational tools giving overall good predictions for hERG blockers with reported IC50s below Pred-hERG and HergSPred cut-off threshold for hERG inhibition. However, for compounds with reported IC50s above this threshold such as disopyramide or sotalol discrepancies were observed. HergSPred identified all 20 hERG agonists selected as interacting with the hERG channel. Further studies are warranted to improve online MLM prediction of hERG related cardiotoxicity, by explicitly taking into account channel agonism as well as inhibition.
Collapse
Affiliation(s)
- Aziza El Harchi
- School of Physiology and Pharmacology and Neuroscience, Biomedical Sciences Building, The University of Bristol, University Walk, Bristol BS8 1TD, UK.
| | - Jules C Hancox
- School of Physiology and Pharmacology and Neuroscience, Biomedical Sciences Building, The University of Bristol, University Walk, Bristol BS8 1TD, UK
| |
Collapse
|
5
|
Assimon MM, Pun PH, Al-Khatib SM, Brookhart MA, Gaynes BN, Winkelmayer WC, Flythe JE. The modifying effect of the serum-to-dialysate potassium gradient on the cardiovascular safety of SSRIs in the hemodialysis population: a pharmacoepidemiologic study. Nephrol Dial Transplant 2022; 37:2241-2252. [PMID: 35793567 DOI: 10.1093/ndt/gfac214] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Hypokalemia is a risk factor for drug-induced QT-prolongation. Larger serum-to-dialysate potassium gradients during hemodialysis may augment the proarrhythmic risks of selective serotonin reuptake inhibitors (SSRIs). METHODS We conducted a cohort study using 2007-2017 data from the United States Renal Data System and a large dialysis provider to examine if the serum-to-dialysate potassium gradient modifies SSRI cardiac safety. Using a new-user design, we compared 1-year sudden cardiac death (SCD) risk among hemodialysis patients newly treated with higher (citalopram, escitalopram) vs. lower (fluoxetine, fluvoxamine, paroxetine, sertraline) QT-prolonging potential SSRIs, overall and stratified by baseline potassium gradient (≥4 vs. <4 mEq/L). We used inverse probability of treatment weighted survival models to estimate weighted hazard ratios (HRs) and 95% confidence intervals (CIs), and conducted a confirmatory nested case-control study. RESULTS The study included 25,099 patients: 11,107 (44.3%) higher QT-prolonging potential SSRI new-users and 13,992 (55.7%) lower QT-prolonging potential SSRI new-users. Overall, higher vs. lower QT-prolonging potential SSRI use was not associated with SCD, weighted HR of 1.03 (95% CI, 0.86-1.24). However, a greater risk of SCD was associated with higher vs. lower QT-prolonging potential SSRI use among patients with baseline potassium gradients ≥4 mEq/L, but not among those with gradients <4 mEq/L, weighted HR of 2.17 (95% CI, 1.16-4.03) vs. 0.95 (0.78-1.16). Nested case-control analyses yielded analogous results. CONCLUSIONS The serum-to-dialysate potassium gradient may modify the association between higher vs. lower QT-prolonging SSRI use and SCD among people receiving hemodialysis. Minimizing the potassium gradient in the setting of QT-prolonging medication use may be warranted.
Collapse
Affiliation(s)
- Magdalene M Assimon
- University of North Carolina Kidney Center, Division of Nephrology and Hypertension, Department of Medicine, UNC School of Medicine, Chapel Hill, NC, USA
| | - Patrick H Pun
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA.,Duke Clinical Research Institute, Durham NC, USA
| | - Sana M Al-Khatib
- Duke Clinical Research Institute, Durham NC, USA.,Division of Cardiology, Duke University Medical Center, Durham, NC, USA
| | - M Alan Brookhart
- Department of Population Health Sciences, Duke University School of Medicine, Durham, NC, USA
| | - Bradley N Gaynes
- Department of Psychiatry, UNC School of Medicine, Chapel Hill, NC, USA.,Department of Epidemiology, UNC Gillings School of Global Public Health, Chapel Hill, NC, USA
| | - Wolfgang C Winkelmayer
- Selzman Institute for Kidney Health, Section of Nephrology, Baylor College of Medicine, Houston, TX, USA
| | - Jennifer E Flythe
- University of North Carolina Kidney Center, Division of Nephrology and Hypertension, Department of Medicine, UNC School of Medicine, Chapel Hill, NC, USA.,Cecil G. Sheps Center for Health Services Research, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
6
|
Eroglu TE, Barcella CA, Gerds TA, Kessing LV, Zylyftari N, Mohr GH, Kragholm K, Polcwiartek C, Wissenberg M, Folke F, Tan HL, Torp-Pedersen C, Gislason GH. Risk of out-of-hospital cardiac arrest in antidepressant drug users. Br J Clin Pharmacol 2022; 88:3162-3171. [PMID: 35001414 PMCID: PMC9303588 DOI: 10.1111/bcp.15224] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 11/29/2021] [Accepted: 12/24/2021] [Indexed: 11/22/2022] Open
Abstract
Conflicting results have been reported regarding the association between antidepressant use and out‐of‐hospital cardiac arrest (OHCA) risk. We investigated whether the use of antidepressants is associated with OHCA.
Collapse
Affiliation(s)
- Talip E Eroglu
- Department of Cardiology, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen, Denmark.,Amsterdam UMC, Academic Medical Center, University of Amsterdam, Department of Experimental and Clinical Cardiology, Heart Centre, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands.,Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Carlo A Barcella
- Department of Cardiology, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen, Denmark
| | - Thomas A Gerds
- The Danish Heart Foundation, Copenhagen, Denmark.,Department of Biostatistics, University of Copenhagen, Copenhagen, Denmark
| | - Lars Vedel Kessing
- Copenhagen Affective Disorder research Center (CADIC), Psychiatric Center Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark.,Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Nertila Zylyftari
- Department of Cardiology, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen, Denmark.,Department of Cardiology, Nordsjaellands Hospital, Hillerød, Denmark
| | - Grimur H Mohr
- Department of Cardiology, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen, Denmark.,Mental Health Centre Glostrup, Copenhagen University Hospital, Copenhagen, Denmark
| | - Kristian Kragholm
- Copenhagen Affective Disorder research Center (CADIC), Psychiatric Center Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
| | - Christoffer Polcwiartek
- Copenhagen Affective Disorder research Center (CADIC), Psychiatric Center Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
| | - Mads Wissenberg
- Department of Cardiology, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen, Denmark
| | - Fredrik Folke
- Department of Cardiology, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen, Denmark.,Copenhagen University Hospital - Copenhagen Emergency Medical Services, Copenhagen, Denmark.,Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Hanno L Tan
- Amsterdam UMC, Academic Medical Center, University of Amsterdam, Department of Experimental and Clinical Cardiology, Heart Centre, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands.,Netherlands Heart Institute, Utrecht, The Netherlands
| | - Christian Torp-Pedersen
- Department of Cardiology, Aalborg University Hospital, Aalborg, Denmark.,Department of Cardiology, Nordsjaellands Hospital, Hillerød, Denmark
| | - Gunnar H Gislason
- Department of Cardiology, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen, Denmark.,Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
7
|
Lee HJ, Choi BH, Choi JS, Hahn SJ. Effects of iloperidone on hERG 1A/3.1 heterotetrameric channels. Neuroreport 2021; 32:1299-1306. [PMID: 34605450 DOI: 10.1097/wnr.0000000000001724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVES Iloperidone is an atypical antipsychotic drug that is widely used for the treatment of schizophrenia. hERG 3.1, alternatively spliced form of hERG 1A, is considered a potential target for an antipsychotic drug. The present study was designed to study the effects of iloperidone on hERG 1A/3.1 heterotetrameric channels. METHODS The interactions of iloperidone with hERG 1A/3.1 heterotetrameric channels stably expressed in HEK cells were investigated using the whole-cell patch-clamp technique and western blot analysis. RESULTS Iloperidone inhibited the hERG 1A/3.1 tail currents at -50 mV in a concentration-dependent manner with an IC50 value of 0.44 μM. The block of hERG 1A/3.1 currents by iloperidone was voltage-dependent and increased over a range of voltage for channel activation. However, the block by iloperidone was voltage-independent at more depolarized potentials where the channels were fully activated. A fast application of iloperidone inhibited the hERG 1A/3.1 current elicited by a 5-s depolarizing pulse to +60 mV to fully inactivate the hERG 1A/3.1 currents. Iloperidone also induced a rapid and reversible inhibition of hERG 1A/3.1 tail currents during repolarization. However, iloperidone had no effect on either hERG 1A or hERG 1A/3.1 channel trafficking to the cell membrane. CONCLUSIONS Our results indicated that iloperidone concentration-dependently inhibited hERG 1A/3.1 currents by preferentially interacting with the open states of channels, but not by the disruption of membrane trafficking or surface membrane expression of hERG 1A and hERG 1A/3.1 channel proteins.
Collapse
Affiliation(s)
- Hong Joon Lee
- Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul
| | - Bok Hee Choi
- Department of Pharmacology, Institute for Medical Science, Jeonbuk National University Medical School, Jeonju
| | - Jin-Sung Choi
- College of Pharmacy, Integrated Research Institute of Pharmaceutical, The Catholic University of Korea, Bucheon, Korea
| | - Sang June Hahn
- Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul
| |
Collapse
|
8
|
Matsuo J, Yamaori S. Detecting drug-drug interactions that increase the incidence of long QT syndrome using a spontaneous reporting system. J Clin Pharm Ther 2021; 47:70-80. [PMID: 34664726 DOI: 10.1111/jcpt.13539] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 10/07/2021] [Indexed: 11/30/2022]
Abstract
WHAT IS KNOWN AND OBJECTIVE Drug-induced long QT syndrome (diLQTS) is a rare but serious adverse drug reaction. Drug-drug interaction (DDI) is one of the risk factors for the development of diLQTS. However, the combinations of drugs that increase the risk of diLQTS have not been extensively investigated. This study was performed to analyse the potential DDIs that elevate the incidence of diLQTS using a spontaneous reporting system. METHODS The Japanese Adverse Drug Event Report database from April 2004 to January 2020 was used to assess adverse event reports. We calculated the reporting odds ratio and 95% confidence interval for signal detection. RESULTS AND DISCUSSION Signals for concomitant use risk were detected in 31 drug combinations. Combinations of antipsychotics and antidepressants were the most common (olanzapine & fluvoxamine, olanzapine & trazodone, quetiapine & paroxetine, sulpiride & fluvoxamine, sulpiride & trazodone). Sixteen, 17 and 21 combinations were designated as requiring precaution for concomitant use in at least one of the package inserts in Japan, the United States and the United Kingdom, respectively, although no such precautions were described for the remaining combinations. On the contrary, a combination of bepridil & clarithromycin was categorized as "X (avoid combination)" and two combinations (chlorpromazine & haloperidol, amiodarone & metildigoxin) were classified as "D (modify regimen)" in the Lexicomp® risk rating. WHAT IS NEW AND CONCLUSION This study identified 31 combinations of drugs that may elevate the risk of diLQTS. The use of these drug combinations should be monitored more carefully in future.
Collapse
Affiliation(s)
- Jun Matsuo
- Department of Pharmacy, Shinshu University Hospital, Matsumoto, Japan.,Department of Biochemical Pharmacology and Toxicology, Graduate School of Medicine, Shinshu University, Matsumoto, Japan
| | - Satoshi Yamaori
- Department of Pharmacy, Shinshu University Hospital, Matsumoto, Japan.,Department of Biochemical Pharmacology and Toxicology, Graduate School of Medicine, Shinshu University, Matsumoto, Japan
| |
Collapse
|
9
|
Nakatani Y, Amano T. Contributions of S- and R-citalopram to the citalopram-induced modulation of the function of Nav1.5 voltage-gated sodium channels. Eur J Pharmacol 2021; 908:174316. [PMID: 34280395 DOI: 10.1016/j.ejphar.2021.174316] [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: 03/24/2021] [Revised: 06/25/2021] [Accepted: 07/07/2021] [Indexed: 11/26/2022]
Abstract
Citalopram, a selective serotonin reuptake inhibitor (SSRI), has been reported to have adverse effects such as cardiotoxicity, including prolongation of the QTc interval. Although citalopram is well known to be a racemic compound comprised of S-citalopram (escitalopram) and R-citalopram, it is still unclear which enantiomer is responsible for cardiotoxicity induced by citalopram. It is also unclear which biomolecule is the target that produces the adverse effect of citalopram. In this study, we investigated whether citalopram, escitalopram and R-citalopram had an electrophysiological effect on Nav1.5 voltage-gated sodium channel (VGSC) current and how their electrophysiological properties affected Nav1.5 VGSC. To examine the effects of the electrophysiological properties of them, whole-cell patch clamp recording was performed using HEK293 cells expressing human Nav1.5 VGSCs. Nav1.5 VGSC current decreased by 60.0 ± 6.3% and 55.1 ± 12.5% under treatment with 100 μM citalopram and escitalopram, respectively. However, 100 μM R-citalopram decreased Nav1.5 VGSC current by only 36.2 ± 8.7%. In addition, treatment with 100 μM citalopram and escitalopram changed the voltage-dependence of activation and induced a negative shift of the voltage of half-maximal activation compared to 100 μM R-citalopram. In contrast, treatment with 100 μM citalopram and escitalopram, but not R-citalopram, changed the voltage-dependence of inactivation, and the voltage at half-maximal inactivation slightly shifted toward negative potential. These results suggest that the adverse cardiac effect produced by citalopram might result from modification of the electrophysiological properties of Nav1.5 VGSCs, and escitalopram might contribute more to this adverse effect than R-citalopram.
Collapse
Affiliation(s)
- Yoshihiko Nakatani
- Department of Pharmacotherapeutics, School of Pharmacy, International University of Health and Welfare, 2600-1 Kitakanemaru, Ohtawara, Tochigi, 324-8501, Japan.
| | - Taku Amano
- Tochigi Mental Health Welfare Center, 2145-13 Shimookamoto, Utsunomiya, Tochigi, 329-1104, Japan
| |
Collapse
|
10
|
Baumann P, Bertschy G, Ramseier F, Nil R. Plasma Concentrations and Cardiovascular Effects of Citalopram Enantiomers After Oral Versus Infusion Citalopram Therapy in Dextromethorphan-Mephenytoin-Phenotyped Patients With Major Depression. Ther Drug Monit 2021; 43:436-442. [PMID: 33060488 DOI: 10.1097/ftd.0000000000000824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 09/21/2020] [Indexed: 11/27/2022]
Abstract
BACKGROUND Authors compared plasma concentrations of citalopram (CIT) enantiomers and their metabolites in patients with depression administered either intravenously (IV) or as oral racemic CIT. Then, plasma concentrations were related to the metabolism of probes used for phenotyping patients with depression for CYP2C19 and CYP2D6 activity and cardiovascular functions. METHODS Dextromethorphan-mephenytoin-phenotyped patients with depression were administered racemic CIT (days 1 and 2: 20 mg/d; days 3-10: 40 mg/d) either orally or as a slow-drop infusion for 10 days and were then orally administered the drug for another 32 days. Blood probes were collected at the time of minimal and maximal concentrations on day 10, immediately before and 2 hours after drug administration, and on days 21 and 42. Plasma CIT and its metabolites were assayed by stereoselective high-performance liquid chromatography. RESULTS The following concentrations (ng/mL) were noted in the group receiving active IV infusion (IV-POS group, n = 27) of racemic CIT on day 10, before drug administration: escitalopram (S-CIT): 24 ± 10.2; R-citalopram (R-CIT): 45 ± 14.5; S-desmethyl-CIT: 13 ± 4.4; and R-desmethyl-CIT: 17 ± 8.2. In patients receiving oral administration (POS-POS group, n = 25), the values were 30 ± 12.7, 51 ± 17.4, 13 ± 4.6, and 17 ± 7.9 ng/mL, respectively. In the IV-POS group, 3 patients were poor dextromethorphan (CYP2D6) metabolizers; in the POS-POS group, one was a poor mephenytoin (CYP2C19) metabolizer. On day 10, before CIT treatment, S/R-CIT and S/R-mephenytoin ratios were significantly correlated, determined at baseline. Overall, CIT reduced the heart rate but did not significantly modify QTc. No relationship was found between any cardiovascular parameters and pharmacokinetic and pharmacogenetic data. CONCLUSIONS Owing to CIT's high bioavailability, the plasma concentrations of its enantiomers remained largely independent on the administration route. CYP2C19 preferentially demethylated S-CIT after CIT therapy.
Collapse
Affiliation(s)
- Pierre Baumann
- Department of Psychiatry, University Hospital of Lausanne (DP-CHUV), Prilly-Lausanne, Switzerland
| | - Gilles Bertschy
- Service de Psychiatrie II, University of Strasbourg, Strasbourg, France
| | - Fritz Ramseier
- Praxengemeinschaft Zürcherstrasse, Rheinfelden, Switzerland; and
| | - Rico Nil
- Consultant, Clinical Research, Uerikon, Switzerland
| |
Collapse
|
11
|
Shalimova A, Babasieva V, Chubarev VN, Tarasov VV, Schiöth HB, Mwinyi J. Therapy response prediction in major depressive disorder: current and novel genomic markers influencing pharmacokinetics and pharmacodynamics. Pharmacogenomics 2021; 22:485-503. [PMID: 34018822 DOI: 10.2217/pgs-2020-0157] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Major depressive disorder is connected with high rates of functional disability and mortality. About a third of the patients are at risk of therapy failure. Several pharmacogenetic markers especially located in CYP450 genes such as CYP2D6 or CYP2C19 are of relevance for therapy outcome prediction in major depressive disorder but a further optimization of predictive tools is warranted. The article summarizes the current knowledge on pharmacogenetic variants, therapy effects and side effects of important antidepressive therapeutics, and sheds light on new methodological approaches for therapy response estimation based on genetic markers with relevance for pharmacokinetics, pharmacodynamics and disease pathology identified in genome-wide association study analyses, highlighting polygenic risk score analysis as a tool for further optimization of individualized therapy outcome prediction.
Collapse
Affiliation(s)
- Alena Shalimova
- Department of Neuroscience, Functional Pharmacology, University of Uppsala, Uppsala, 751 24, Sweden.,Department of Pharmacology, Institute of Pharmacy, I. M. Sechenov First Moscow State Medical University, Moscow, 119991, Russia
| | - Viktoria Babasieva
- Department of Neuroscience, Functional Pharmacology, University of Uppsala, Uppsala, 751 24, Sweden.,Department of Pharmacology, Institute of Pharmacy, I. M. Sechenov First Moscow State Medical University, Moscow, 119991, Russia
| | - Vladimir N Chubarev
- Department of Pharmacology, Institute of Pharmacy, I. M. Sechenov First Moscow State Medical University, Moscow, 119991, Russia
| | - Vadim V Tarasov
- Department of Pharmacology, Institute of Pharmacy, I. M. Sechenov First Moscow State Medical University, Moscow, 119991, Russia.,Institute of Translational Medicine & Biotechnology, I. M. Sechenov First Moscow State Medical University, Moscow, 119991, Russia
| | - Helgi B Schiöth
- Department of Neuroscience, Functional Pharmacology, University of Uppsala, Uppsala, 751 24, Sweden.,Institute of Translational Medicine & Biotechnology, I. M. Sechenov First Moscow State Medical University, Moscow, 119991, Russia
| | - Jessica Mwinyi
- Department of Neuroscience, Functional Pharmacology, University of Uppsala, Uppsala, 751 24, Sweden
| |
Collapse
|
12
|
Kim Y, Kim A, Chung JY. Population pharmacokinetic/pharmacodynamic modeling of delayed effect of escitalopram-induced QT prolongation. J Affect Disord 2021; 285:120-126. [PMID: 33647579 DOI: 10.1016/j.jad.2021.02.048] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 02/14/2021] [Accepted: 02/17/2021] [Indexed: 02/01/2023]
Abstract
BACKGROUND A thorough QT study identified that escitalopram-induced QT prolongation was delayed. This study thus aimed to develop a population pharmacokinetic (PK)/pharmacodynamic (PD) model to characterize the relationship between escitalopram concentrations and the delayed effect on QT prolongation. METHODS The data of completed subjects who had placebo (n=36) and a single dose of 20 mg escitalopram (n=33) from a previous thorough QT study were used. Population PK/PD analysis was performed by nonlinear mixed-effects modeling. A escitalopram concentration-drug effect model was developed with estimated individual PK and baseline QT parameters. To explain the relationship between escitalopram concentrations and QT prolongation delay, an effect compartment model was utilized. RESULTS A two-compartment model with first-order absorption and lag time and first-order elimination adequately described the PK of escitalopram. The circadian rhythm of baseline QT interval was best explained by two harmonic cosine functions. A linear model properly characterized escitalopram-induced QT prolongation. The average estimated maximal QT prolongation was 5.4 ms (range: 1.9-7.6 ms). The equilibrium half-life of delayed QT prolongation was 1.9 h. The drug effect of QTc change compared with that at baseline remained relatively constant from 1.3 to 3.5 ms over 24 h, and the maximum QTc change occurred with a 3-h delay after the time to the maximum plasma concentration. LIMITATIONS We did not include genetic polymorphisms, such as CYP2C19, as potential covariates owing to limited information. CONCLUSIONS These results may provide useful information on when to monitor electrocardiogram in patients who require intensive care after drug administration.
Collapse
Affiliation(s)
- Yun Kim
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Republic of Korea
| | - Anhye Kim
- Department of Clinical Pharmacology and Therapeutics, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Jae-Yong Chung
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Bundang Hospital, Seongnam, Republic of Korea.
| |
Collapse
|
13
|
Su S, Sun J, Wang Y, Xu Y. Cardiac hERG K + Channel as Safety and Pharmacological Target. Handb Exp Pharmacol 2021; 267:139-166. [PMID: 33829343 DOI: 10.1007/164_2021_455] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The human ether-á-go-go related gene (hERG, KCNH2) encodes the pore-forming subunit of the potassium channel responsible for a fast component of the cardiac delayed rectifier potassium current (IKr). Outward IKr is an important determinant of cardiac action potential (AP) repolarization and effectively controls the duration of the QT interval in humans. Dysfunction of hERG channel can cause severe ventricular arrhythmias and thus modulators of the channel, including hERG inhibitors and activators, continue to attract intense pharmacological interest. Certain inhibitors of hERG channel prolong the action potential duration (APD) and effective refractory period (ERP) to suppress premature ventricular contraction and are used as class III antiarrhythmic agents. However, a reduction of the hERG/IKr current has been recognized as a predominant mechanism responsible for the drug-induced delayed repolarization known as acquired long QT syndromes (LQTS), which is linked to an increased risk for "torsades de pointes" (TdP) ventricular arrhythmias and sudden cardiac death. Many drugs of different classes and structures have been identified to carry TdP risk. Hence, assessing hERG/IKr blockade of new drug candidates is mandatory in the drug development process according to the regulatory agencies. In contrast, several hERG channel activators have been shown to enhance IKr and shorten the APD and thus might have potential antiarrhythmic effects against pathological LQTS. However, these activators may also be proarrhythmic due to excessive shortening of APD and the ERP.
Collapse
Affiliation(s)
- Shi Su
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei, China
| | - Jinglei Sun
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei, China
| | - Yi Wang
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei, China
| | - Yanfang Xu
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei, China.
| |
Collapse
|
14
|
Czaja AS, Anderson HD, Ghosh D, Davidson J, Campbell JD, Valuck RJ. Increased Odds of Ventricular Arrhythmias Associated with Selective Serotonin Reuptake Inhibitor Use among the Pediatric and Young Adult Population: A Case-Control Study. J Pediatr 2020; 226:173-178.e8. [PMID: 32645405 DOI: 10.1016/j.jpeds.2020.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/31/2020] [Accepted: 07/01/2020] [Indexed: 10/23/2022]
Abstract
OBJECTIVE To measure the association between selective serotonin reuptake inhibitor (SSRI) use and out-of-hospital ventricular arrhythmia among the pediatric and young adult population. STUDY DESIGN Case-control study using US claims data from 2007 to 2018. Cases were subjects with at least 1 event between ages 2 and 24 years. Controls (matched 10:1 on index date, age, sex, and continuous enrollment) had no events during study period. Independent association between current SSRI use (prescription fill with continuous exposure ending on, or after, the index date) and incident out-of-hospital ventricular arrhythmia (hospitalization or emergency room encounter with primary diagnostic code for ventricular arrhythmia) was estimated using multivariable conditional logistic regression. Separate analyses were performed for pediatric (2-17 years of age) vs young adult (18-24 years of age) subjects and between citalopram/escitalopram vs other SSRIs. RESULTS During the study period, 237 eligible cases were identified with 2370 matched controls. Cases were more likely to have government insurance and have a mental health, cardiac, or other complex chronic condition. Thirteen cases (5%) and 15 controls (<1%) had current SSRI exposure. After adjustment for mental health and chronic conditions, there was an increased odds of current SSRI use among cases compared with controls (OR 5.11, 95% CI 1.22-21.37). No difference was observed between pediatric and young adult ages, nor between citalopram/escitalopram and other SSRIs. CONCLUSIONS These findings demonstrate increased odds of out-of-hospital ventricular arrhythmia associated with SSRI use in the pediatric and young adult population, suggesting a need for heightened awareness and ongoing monitoring of this potential adverse effect.
Collapse
Affiliation(s)
- Angela S Czaja
- Department of Pediatrics, Critical Care Section, School of Medicine, University of Colorado, Aurora, CO.
| | - Heather D Anderson
- Department of Clinical Pharmacy, Center for Pharmaceutical Outcomes (CePOR), Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO
| | - Debashis Ghosh
- Department of Biostatistics & Informatics, Colorado School of Public Health, University of Colorado, Aurora, CO
| | - Jesse Davidson
- Department of Pediatrics, Cardiology Section, School of Medicine, University of Colorado, Aurora, CO
| | - Jonathan D Campbell
- Department of Clinical Pharmacy, Center for Pharmaceutical Outcomes (CePOR), Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO
| | - Robert J Valuck
- Department of Clinical Pharmacy, Center for Pharmaceutical Outcomes (CePOR), Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO
| |
Collapse
|
15
|
Park YS, Sung KW. Selective serotonin reuptake inhibitor escitalopram inhibits 5-HT 3 receptor currents in NCB-20 cells. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2019; 23:509-517. [PMID: 31680773 PMCID: PMC6819908 DOI: 10.4196/kjpp.2019.23.6.509] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 10/09/2019] [Accepted: 10/10/2019] [Indexed: 11/15/2022]
Abstract
Escitalopram is one of selective serotonin reuptake inhibitor antidepressants. As an S-enantiomer of citalopram, it shows better therapeutic outcome in depression and anxiety disorder treatment because it has higher selectivity for serotonin reuptake transporter than citalopram. The objective of this study was to determine the direct inhibitory effect of escitalopram on 5-hydroxytryptamine type 3 (5-HT3) receptor currents and study its blocking mechanism to explore additional pharmacological effects of escitalopram through 5-HT3 receptors. Using a whole-cell voltage clamp method, we recorded currents of 5-HT3 receptors when 5-HT was applied alone or co-applied with escitalopram in cultured NCB-20 neuroblastoma cells known to express 5-HT3 receptors. 5-HT induced currents were inhibited by escitalopram in a concentration-dependent manner. EC50 of 5-HT on 5-HT3 receptor currents was increased by escitalopram while the maximal peak amplitude was reduced by escitalopram. The inhibitory effect of escitalopram was voltage independent. Escitalopram worked more effectively when it was co-applied with 5-HT than pre-application of escitalopram. Moreover, escitalopram showed fast association and dissociation to the open state of 5-HT3 receptor channel with accelerating receptor desensitization. Although escitalopram accelerated 5-HT3 receptor desensitization, it did not change the time course of desensitization recovery. These results suggest that escitalopram can inhibit 5-HT3 receptor currents in a non-competitive manner with the mechanism of open channel blocking.
Collapse
Affiliation(s)
- Yong Soo Park
- Department of Anatomy, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Ki-Wug Sung
- Department of Pharmacology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| |
Collapse
|
16
|
Eugene AR. Optimizing drug selection in psychopharmacology based on 40 significant CYP2C19- and CYP2D6-biased adverse drug reactions of selective serotonin reuptake inhibitors. PeerJ 2019; 7:e7860. [PMID: 31616600 PMCID: PMC6790106 DOI: 10.7717/peerj.7860] [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] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 09/09/2019] [Indexed: 12/19/2022] Open
Abstract
Background Selective serotonin reuptake inhibitors (SSRIs) are among the most widely prescribed class of drugs in the practice of psychiatry. Cytochrome P450 (CYP) 2C19 and CYP2D6 are established as clinically relevant drug metabolizing enzymes (DMEs) that influence the pharmacokinetics of SSRIs and may either be grouped as being primarily metabolized by CYP2C19 or CYP2D6. The aim of this study is to test the hypothesis that the primary drug metabolizing pathway for SSRI antidepressants are associated with adverse drug reactions (ADRs) related to physiological modulation of organs with the highest gene tissue expression. Methods Post-marketing ADR cases were obtained from the United States Food and Drug Administration’s Adverse Events Reporting System from each of the four quarters for the years 2016 and 2017. Cases were grouped based on one of two primary pharmacokinetic pharmacogenomic pathway biomarkers CYP2C19 and CYP2D6. Citalopram, escitalopram, and sertraline were grouped as CYP2C19 substrates and fluvoxamine, fluoxetine, and paroxetine as CYP2D6 substrates. Logistic regression was computed for the reported SSRI ADRs associated with one of two aforementioned DMEs. All data homogenization and computations were performed in R for statistical programming. Results The most commonly reported ADR among the SSRIs was anxiety (n = 3,332). The top two ADRs associated with SSRIs metabolized by CYP2D6 are: nightmare (n = 983) reporting odds-ratio (OR) = 4.37 (95% confidence interval (CI) [3.67–5.20]) and panic attack (n = 1,243) OR = 2.43 (95% CI [2.11–2.79]). Contrastingly, the top two ADRs for CYP2C19 metabolized SSRIs are: electrocardiogram QT prolonged (n = 351) OR = 0.18 (95% CI [0.13–0.24]) and small for dates baby (n = 306) OR = 0.19 (95% CI [0.14–0.26]). The study tested and produced 40 statistically significant CYP2C19- and CYP2D6-biased ADRs. In overall context, the results suggest that CYPC19 SSRI substrates are associated with ADRs related to modulation of the autonomic nervous system, seizure, pain, erectile-dysfunction, and absorption. Contrastingly, CYP2D6 SSRI substrates are associated with ADRs related to nightmares, withdrawal syndrome, and de-realization of cognitive processes. The results of this study may aid as guidance to optimize drug selection in psychopharmacology.
Collapse
Affiliation(s)
- Andy R Eugene
- Independent Neurophysiology Unit, Department of Psychiatry, Medical University of Lublin, Lublin, Poland.,Independent Researcher, Kansas, USA
| |
Collapse
|
17
|
Effects of cariprazine on hERG 1A and hERG 1A/3.1 potassium channels. Eur J Pharmacol 2019; 854:92-100. [DOI: 10.1016/j.ejphar.2019.04.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 04/01/2019] [Accepted: 04/02/2019] [Indexed: 12/28/2022]
|
18
|
Assimon MM, Brookhart MA, Flythe JE. Comparative Cardiac Safety of Selective Serotonin Reuptake Inhibitors among Individuals Receiving Maintenance Hemodialysis. J Am Soc Nephrol 2019; 30:611-623. [PMID: 30885935 PMCID: PMC6442344 DOI: 10.1681/asn.2018101032] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 01/19/2019] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Individuals receiving maintenance hemodialysis may be particularly susceptible to the lethal cardiac consequences of drug-induced QT prolongation because they have a substantial cardiovascular disease burden and high level of polypharmacy, as well as recurrent exposure to electrolyte shifts during dialysis. Electrophysiologic data indicate that among the selective serotonin reuptake inhibitors (SSRIs), citalopram and escitalopram prolong the QT interval to the greatest extent. However, the relative cardiac safety of SSRIs in the hemodialysis population is unknown. METHODS In this retrospective cohort study, we used data from a cohort of Medicare beneficiaries receiving hemodialysis included in the US Renal Data System registry (2007-2014). We used a new-user design to compare the 1-year risk of sudden cardiac death among hemodialysis patients initiating SSRIs with a higher potential for prolonging the QT interval (citalopram, escitalopram) versus the risk among those initiating SSRIs with lower QT-prolonging potential (fluoxetine, fluvoxamine, paroxetine, sertraline). We estimated adjusted hazard ratios using inverse probability of treatment weighted survival models. Nonsudden cardiac death was treated as a competing event. RESULTS The study included 30,932 (47.1%) hemodialysis patients who initiated SSRIs with higher QT-prolonging potential and 34,722 (52.9%) who initiated SSRIs with lower QT-prolonging potential. Initiation of an SSRI with higher versus lower QT-prolonging potential was associated with higher risk of sudden cardiac death (adjusted hazard ratio, 1.18; 95% confidence interval, 1.05 to 1.31). This association was more pronounced among elderly individuals, females, patients with conduction disorders, and those treated with other non-SSRI QT-prolonging medications. CONCLUSIONS The heterogeneous QT-prolonging potential of SSRIs may differentially affect cardiac outcomes in the hemodialysis population.
Collapse
Affiliation(s)
- Magdalene M Assimon
- Division of Nephrology and Hypertension, Department of Medicine, University of North Carolina Kidney Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina;
| | - M Alan Brookhart
- Department of Epidemiology, University of North Carolina Gillings School of Global Public Heath, Chapel Hill, North Carolina; and
| | - Jennifer E Flythe
- Division of Nephrology and Hypertension, Department of Medicine, University of North Carolina Kidney Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina
- Cecil G. Sheps Center for Health Services Research, University of North Carolina, Chapel Hill, North Carolina
| |
Collapse
|
19
|
Ng I, Greenblatt HK, Greenblatt DJ. Stereo-Psychopharmacology: The Case of Citalopram and Escitalopram. Clin Pharmacol Drug Dev 2018; 5:331-5. [PMID: 27452501 DOI: 10.1002/cpdd.293] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Isaac Ng
- Tufts University School of Medicine, Boston, MA, USA
| | | | | |
Collapse
|
20
|
Lee HJ, Choi JS, Choi BH, Hahn SJ. Effects of norquetiapine, the active metabolite of quetiapine, on cloned hERG potassium channels. Neurosci Lett 2017; 664:66-73. [PMID: 29133173 DOI: 10.1016/j.neulet.2017.11.029] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 11/06/2017] [Accepted: 11/09/2017] [Indexed: 12/25/2022]
Abstract
Quetiapine is an atypical antipsychotic drug that is widely used for the treatment of schizophrenia. It is mainly metabolized by a cytochrome P450 system in the liver. Norquetiapine is a major active metabolite in humans with a pharmacological profile that differs distinctly from that of quetiapine. We used the whole-cell patch-clamp technique to investigate the effects of norquetiapine on hERG channels that are stably expressed in HEK cells. Quetiapine and norquetiapine inhibited the hERG tail currents at -50mV in a concentration-dependent manner with IC50 values of 8.3 and 10.8μM, respectively, which suggested equal potency. The block of hERG currents by norquetiapine was voltage-dependent with a steep increase over a range of voltages for channel activation. However, at more depolarized potentials where the channels were fully activated, the block by norquetiapine was voltage-independent. The steady-state inactivation curve of the hERG currents was shifted to the hyperpolarizing direction in the presence of norquetiapine. Norquetiapine did not produce a use-dependent block. A fast application of norquetiapine inhibited the hERG current elicited by a 5s depolarizing pulse to +60mV, which fully inactivated the hERG currents, suggesting an inactivated-state block. During a repolarizing pulse wherein the hERG current was slowly deactivated, albeit remaining in an open state, a fast application of norquetiapine rapidly and reversibly inhibited the open state of the hERG current. Our results indicated that quetiapine and norquetiapine had equal potency in inhibiting hERG tail currents. Norquetiapine inhibited the hERG current by preferentially interacting with the open and/or inactivated states of the channels.
Collapse
Affiliation(s)
- Hong Joon Lee
- Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Jin-Sung Choi
- College of Pharmacy, Integrated Research Institute of Pharmaceutical, The Catholic University of Korea, Gyeonggi-do, Republic of Korea
| | - Bok Hee Choi
- Department of Pharmacology, Institute for Medical Science, Chonbuk National University Medical School, Jeonju, Jeonbuk 54097, Republic of Korea
| | - Sang June Hahn
- Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea.
| |
Collapse
|
21
|
Kim HS, Li H, Kim HW, Shin SE, Seo MS, An JR, Ha KS, Han ET, Hong SH, Choi IW, Choi G, Lee DS, Park WS. Escitalopram, a selective serotonin reuptake inhibitor, inhibits voltage-dependent K + channels in coronary arterial smooth muscle cells. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2017; 21:415-421. [PMID: 28706455 PMCID: PMC5507780 DOI: 10.4196/kjpp.2017.21.4.415] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 02/27/2017] [Accepted: 03/08/2017] [Indexed: 01/06/2023]
Abstract
We investigated the inhibitory effect of escitalopram, a selective serotonin reuptake inhibitor (SSRI), on voltage-dependent K+ (Kv) channels in freshly separated from rabbit coronary arterial smooth muscle cells. The application of escitalopram rapidly inhibited vascular Kv channels. Kv currents were progressively inhibited by an increase in the concentrations of escitalopram, suggesting that escitalopram inhibited vascular Kv currents in a concentration-dependent manner. The IC50 value and Hill coefficient for escitalopram-induced inhibition of Kv channels were 9.54±1.33 µM and 0.75±0.10, respectively. Addition of escitalopram did not alter the steady-state activation and inactivation curves, suggesting that the voltage sensors of the channels were not affected. Pretreatment with inhibitors of Kv1.5 and/or Kv2.1 did not affect the inhibitory action of escitalopram on vascular Kv channels. From these results, we concluded that escitalopram decreased the vascular Kv current in a concentration-dependent manner, independent of serotonin reuptake inhibition.
Collapse
Affiliation(s)
- Han Sol Kim
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon 24341, Korea
| | - Hongliang Li
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon 24341, Korea
| | - Hye Won Kim
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon 24341, Korea
| | - Sung Eun Shin
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon 24341, Korea
| | - Mi Seon Seo
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon 24341, Korea
| | - Jin Ryeol An
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon 24341, Korea
| | - Kwon-Soo Ha
- Department of Molecular and Cellular Biochemistry, Kangwon National University School of Medicine, Chuncheon 24341, Korea
| | - Eun-Taek Han
- Department of Medical Environmental Biology and Tropical Medicine, Kangwon National University School of Medicine, Chuncheon 24341, Korea
| | - Seok-Ho Hong
- Department of Internal Medicine, Kangwon National University School of Medicine, Chuncheon 24341, Korea
| | - Il-Whan Choi
- Department of Microbiology, Inje University College of Medicine, Busan 47392, Korea
| | - Grace Choi
- Department of Applied Research, National Marine Biodiversity Institute of Korea, Seocheon 33662, Korea
| | - Dae-Sung Lee
- Department of Applied Research, National Marine Biodiversity Institute of Korea, Seocheon 33662, Korea
| | - Won Sun Park
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon 24341, Korea
| |
Collapse
|
22
|
Inhibition of cloned hERG potassium channels by risperidone and paliperidone. Naunyn Schmiedebergs Arch Pharmacol 2017; 390:633-642. [DOI: 10.1007/s00210-017-1364-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Accepted: 02/24/2017] [Indexed: 12/31/2022]
|
23
|
Joo YS, Lee HJ, Choi JS, Sung KW. Acepromazine inhibits hERG potassium ion channels expressed in human embryonic kidney 293 cells. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2017; 21:75-82. [PMID: 28066143 PMCID: PMC5214913 DOI: 10.4196/kjpp.2017.21.1.75] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 09/07/2016] [Accepted: 09/09/2016] [Indexed: 12/22/2022]
Abstract
The effects of acepromazine on human ether-à-go-go-related gene (hERG) potassium channels were investigated using whole-cell voltage-clamp technique in human embryonic kidney (HEK293) cells transfected with hERG. The hERG currents were recorded with or without acepromazine, and the steady-state and peak tail currents were analyzed for the evaluating the drug effects. Acepromazine inhibited the hERG currents in a concentration-dependent manner with an IC50 value of 1.5 µM and Hill coefficient of 1.1. Acepromazine blocked hERG currents in a voltage-dependent manner between –40 and +10 mV. Before and after application of acepromazine, the half activation potentials of hERG currents changed to hyperpolarizing direction. Acepromazine blocked both the steady-state hERG currents by depolarizing pulse and the peak tail currents by repolarizing pulse; however, the extent of blocking by acepromazine in the repolarizing pulse was more profound than that in the depolarizing pulse, indicating that acepromazine has a high affinity for the open state of the channels, with a relatively lower affinity for the closed state of hERG channels. A fast application of acepromazine during the tail currents inhibited the open state of hERG channels in a concentration-dependent. The steady-state inactivation of hERG currents shifted to the hyperpolarized direction by acepromazine. These results suggest that acepromazine inhibits the hERG channels probably by an open- and inactivated-channel blocking mechanism. Regarding to the fact that the hERG channels are the potential target of drug-induced long QT syndrome, our results suggest that acepromazine can possibly induce a cardiac arrhythmia through the inhibition of hERG channels.
Collapse
Affiliation(s)
- Young Shin Joo
- Department of Pharmacology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Hong Joon Lee
- Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Jin-Sung Choi
- College of Pharmacy, Integrated Research Institute of Pharmaceutical, The Catholic University of Korea, Seoul 14662, Korea
| | - Ki-Wug Sung
- Department of Pharmacology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| |
Collapse
|
24
|
A thorough QT study to evaluate the QTc prolongation potential of two neuropsychiatric drugs, quetiapine and escitalopram, in healthy volunteers. Int Clin Psychopharmacol 2016; 31:210-7. [PMID: 26950553 DOI: 10.1097/yic.0000000000000124] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Prolongation of the QT interval on an ECG is a surrogate marker for predicting the proarrhythmic potential of a drug under development. The aim of this study was to evaluate the QTc prolongation potential of two neuropsychiatric drugs, quetiapine immediate release (IR) and escitalopram, in healthy individuals. This was a randomized, open-label, 4×4 Williams crossover study, with four single-dose treatments [placebo, 400 mg moxifloxacin (positive control), 20 mg escitalopram, and 100 mg quetiapine IR], conducted in 40 healthy volunteers. Serial blood samples for pharmacokinetics and ECG were collected. Individually, RR-corrected QTc intervals (QTcI) and placebo-adjusted changes from baseline values of QTcI (ΔΔQTcI) were evaluated. Lower-bound values of the one-sided 95% confidence interval for ΔΔQTcI of moxifloxacin with more than 5 ms confirmed the sensitivity of the assay. The maximum upper bound 95% confidence interval for the ΔΔQTcI of quetiapine IR and escitalopram was 13.7 and 10.5 ms, with mean estimates of 10.2 and 6.9 ms, respectively. Peak effects of moxifloxacin and quetiapine IR on ΔΔQTcI were observed at approximately time to maximum concentration (Tmax), whereas that of escitalopram was observed 3 h after Tmax. The concentration-ΔΔQTcI relationships of quetiapine IR and escitalopram were relatively flat, as compared with that of moxifloxacin. The results demonstrated the validity of trial methodology and that quetiapine IR and escitalopram caused QT prolongation in healthy individuals. In addition, hysteresis of escitalopram-induced QTc prolongation. These results indicate that higher doses of these drugs could lead to greater QT prolongation in a dose-response manner.
Collapse
|
25
|
Lehmann DF, Wojnowicz S. The Evergreening of Biopharmaceuticals: Time to Defoliate. J Clin Pharmacol 2015; 56:383-9. [PMID: 26388527 DOI: 10.1002/jcph.642] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Indexed: 11/07/2022]
|
26
|
Shah RR, Morganroth J, Kleiman RB. ICH E14 Q&A(R2) document: commentary on the further updated recommendations on thorough QT studies. Br J Clin Pharmacol 2015; 79:456-64. [PMID: 25060671 DOI: 10.1111/bcp.12477] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Accepted: 07/21/2014] [Indexed: 12/28/2022] Open
|
27
|
Lee HJ, Choi JS, Hahn SJ. Mechanism of inhibition by olanzapine of cloned hERG potassium channels. Neurosci Lett 2015; 609:97-102. [DOI: 10.1016/j.neulet.2015.10.039] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 09/14/2015] [Accepted: 10/13/2015] [Indexed: 01/01/2023]
|
28
|
Frommeyer G, Eckardt L. Drug-induced proarrhythmia: risk factors and electrophysiological mechanisms. Nat Rev Cardiol 2015; 13:36-47. [PMID: 26194552 DOI: 10.1038/nrcardio.2015.110] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Drug-induced ventricular tachyarrhythmias can be caused by cardiovascular drugs, noncardiovascular drugs, and even nonprescription agents. They can result in arrhythmic emergencies and sudden cardiac death. If a new arrhythmia or aggravation of an existing arrhythmia develops during therapy with a drug at a concentration usually considered not to be toxic, the situation can be defined as proarrhythmia. Various cardiovascular and noncardiovascular drugs can increase the occurrence of polymorphic ventricular tachycardia of the 'torsade de pointes' type. Antiarrhythmic drugs, antimicrobial agents, and antipsychotic and antidepressant drugs are the most important groups. Age, female sex, and structural heart disease are important risk factors for the occurrence of torsade de pointes. Genetic predisposition and individual pharmacodynamic and pharmacokinetic sensitivity also have important roles in the generation of arrhythmias. An increase in spatial or temporal dispersion of repolarization and a triangular action-potential configuration have been identified as crucial predictors of proarrhythmia in experimental models. These studies emphasized that sole consideration of the QT interval is not sufficient to assess the proarrhythmic risk. In this Review, we focus on important triggers of proarrhythmia and the underlying electrophysiological mechanisms that can enhance or prevent the development of torsade de pointes.
Collapse
Affiliation(s)
- Gerrit Frommeyer
- Division of Electrophysiology, Department of Cardiovascular Medicine, University of Münster, Albert-Schweitzer Strasse 33, D-48149 Münster, Germany
| | - Lars Eckardt
- Division of Electrophysiology, Department of Cardiovascular Medicine, University of Münster, Albert-Schweitzer Strasse 33, D-48149 Münster, Germany
| |
Collapse
|
29
|
The protease inhibitor atazanavir blocks hERG K(+) channels expressed in HEK293 cells and obstructs hERG protein transport to cell membrane. Acta Pharmacol Sin 2015; 36:454-62. [PMID: 25832426 DOI: 10.1038/aps.2014.165] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 12/19/2014] [Indexed: 12/16/2022]
Abstract
AIM Atazanavir (ATV) is a HIV-1 protease inhibitor for the treatment of AIDS patients, which is recently reported to provoke excessive prolongation of the QT interval and torsades de pointes (TdP). In order to elucidate its arrhythmogenic mechanisms, we investigated the effects of ATV on the hERG K(+) channels expressed in HEK293 cells. METHODS hERG K(+) currents were detected using whole-cell patch clamp recording in HEK293 cells transfected with EGFP-hERG plasmids. The expression of hERG protein was measured with Western blotting. Two mutants (Y652A and F656C) were constructed in the S6 domain within the inner helices of hERG K(+) channels that were responsible for binding of various drugs. The trafficking of hERG protein was studied with confocal microscopy. RESULTS Application of ATV (0.01-30 μmol/L) concentration-dependently decreased hERG K(+) currents with an IC50 of 5.7±1.8 μmol/L. ATV (10 μmol/L) did not affect the activation and steady-state inactivation of hERG K(+) currents. Compared with the wild type hERG K(+) channels, both Y652A and F656C mutants significantly reduced the inhibition of ATV on hERG K(+) currents. Overnight treatment with ATV (0.1-30 μmol/L) concentration-dependently reduced the amount of fully glycosylated 155 kDa hERG protein without significantly affecting the core-glycosylated 135 kDa hERG protein in the cells expressing the WT-hERG protein. Confocal microscopy studies confirmed that overnight treatment with ATV obstructed the trafficking of hERG protein to the cell membrane. CONCLUSION ATV directly blocks hERG K(+) channels via binding to the residues Y652 and F656 in the S6 domain, and indirectly obstructs the transport of the hERG protein to the cell membrane.
Collapse
|
30
|
Chae YJ, Lee KJ, Lee HJ, Sung KW, Choi JS, Lee EH, Hahn SJ. Endoxifen, the active metabolite of tamoxifen, inhibits cloned hERG potassium channels. Eur J Pharmacol 2015; 752:1-7. [PMID: 25680947 DOI: 10.1016/j.ejphar.2015.01.048] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2014] [Revised: 01/15/2015] [Accepted: 01/19/2015] [Indexed: 11/27/2022]
Abstract
The effects of tamoxifen, and its active metabolite endoxifen (4-hydroxy-N-desmethyl-tamoxifen), on hERG currents stably expressed in HEK cells were investigated using the whole-cell patch-clamp technique and an immunoblot assay. Tamoxifen and endoxifen inhibited hERG tail currents at -50mV in a concentration-dependent manner with IC50 values of 1.2 and 1.6μM, respectively. The steady-state activation curve of the hERG currents was shifted to the hyperpolarizing direction in the presence of endoxifen. The voltage-dependent inhibition of hERG currents by endoxifen increased steeply in the voltage range of channel activation. The inhibition by endoxifen displayed a shallow voltage dependence (δ=0.18) in the full activation voltage range. A fast application of endoxifen induced a reversible block of hERG tail currents during repolarization in a concentration-dependent manner, which suggested an interaction with the open state of the channel. Endoxifen also decreased the hERG current elicited by a 5s depolarizing pulse to +60mV to inactivate the hERG currents, suggesting an interaction with the activated (open and/or inactivated) states of the channels. Tamoxifen and endoxifen inhibited the hERG channel protein trafficking to the plasma membrane in a concentration-dependent manner with endoxifen being more potent than tamoxifen. These results indicated that tamoxifen and endoxifen inhibited the hERG current by direct channel blockage and by the disruption of channel trafficking to the plasma membrane in a concentration-dependent manner. A therapeutic concentration of endoxifen inhibited the hERG current by preferentially interacting with the activated (open and/or inactivated) states of the channel.
Collapse
Affiliation(s)
- Yun Ju Chae
- Department of Physiology, The Catholic University of Korea, Seoul 137-701, Republic of Korea
| | - Keon Jin Lee
- Department of Physiology, The Catholic University of Korea, Seoul 137-701, Republic of Korea
| | - Hong Joon Lee
- Pharmacology, College of Medicine, The Catholic University of Korea, Seoul 137-701, Republic of Korea
| | - Ki-Wug Sung
- Pharmacology, College of Medicine, The Catholic University of Korea, Seoul 137-701, Republic of Korea
| | - Jin-Sung Choi
- College of Pharmacy, Integrated Research Institute of Pharmaceutical, The Catholic University of Korea, Gyeonggi-do, Republic of Korea
| | - Eun Hui Lee
- Department of Physiology, The Catholic University of Korea, Seoul 137-701, Republic of Korea
| | - Sang June Hahn
- Department of Physiology, The Catholic University of Korea, Seoul 137-701, Republic of Korea.
| |
Collapse
|
31
|
Chae YJ, Lee HJ, Jeon JH, Kim IB, Choi JS, Sung KW, Hahn SJ. Effects of donepezil on hERG potassium channels. Brain Res 2014; 1597:77-85. [PMID: 25498859 DOI: 10.1016/j.brainres.2014.11.057] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 11/04/2014] [Accepted: 11/27/2014] [Indexed: 11/26/2022]
Abstract
Donepezil is a potent, selective inhibitor of acetylcholinesterase, which is used for the treatment of Alzheimer's disease. Whole-cell patch-clamp technique and Western blot analyses were used to study the effects of donepezil on the human ether-a-go-go-related gene (hERG) channel. Donepezil inhibited the tail current of the hERG in a concentration-dependent manner with an IC50 of 1.3 μM. The metabolites of donepezil, 6-ODD and 5-ODD, inhibited the hERG currents in a similar concentration-dependent manner; the IC50 values were 1.0 and 1.5 μM, respectively. A fast drug perfusion system demonstrated that donepezil interacted with both the open and inactivated states of the hERG. A fast application of donepezil during the tail currents inhibited the open state of the hERG in a concentration-dependent manner with an IC50 of 2.7 μM. Kinetic analysis of donepezil in an open state of the hERG yielded blocking and unblocking rate constants of 0.54 µM(-1)s(-1) and 1.82 s(-1), respectively. The block of the hERG by donepezil was voltage-dependent with a steep increase across the voltage range of channel activation. Donepezil caused a reduction in the hERG channel protein trafficking to the plasma membrane at low concentration, but decreased the channel protein expression at higher concentrations. These results suggest that donepezil inhibited the hERG at a supratherapeutic concentration, and that it did so by preferentially binding to the activated (open and/or inactivated) states of the channels and by inhibiting the trafficking and expression of the hERG channel protein in the plasma membrane.
Collapse
Affiliation(s)
- Yun Ju Chae
- Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul 137-701, 222 Banpo-daero, Seocho-gu, Korea
| | - Hong Joon Lee
- Department of Pharmacology, College of Medicine, The Catholic University of Korea, Seoul 137-701, Korea
| | - Ji Hyun Jeon
- Department of Anatomy, College of Medicine, The Catholic University of Korea, Seoul 137-701, Korea
| | - In-Beom Kim
- Department of Anatomy, College of Medicine, The Catholic University of Korea, Seoul 137-701, Korea
| | - Jin-Sung Choi
- College of Pharmacy, Integrated Research Institute of Pharmaceutical, The Catholic University of Korea, 43-1 Yeokgok 2-dong, Wonmi-gu, Bucheon, Gyeonggi-do, Korea
| | - Ki-Wug Sung
- Department of Pharmacology, College of Medicine, The Catholic University of Korea, Seoul 137-701, Korea
| | - Sang June Hahn
- Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul 137-701, 222 Banpo-daero, Seocho-gu, Korea.
| |
Collapse
|
32
|
Donazzolo Y, Latreille M, Caillaud MA, Mocaer E, Seguin L. Evaluation of the Effects of Therapeutic and Supratherapeutic Doses of Agomelatine on the QT/QTc Interval. J Cardiovasc Pharmacol 2014; 64:440-51. [DOI: 10.1097/fjc.0000000000000136] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|