51
|
Sheridan RP, McMasters DR, Voigt JH, Wildey MJ. eCounterscreening: Using QSAR Predictions to Prioritize Testing for Off-Target Activities and Setting the Balance between Benefit and Risk. J Chem Inf Model 2015; 55:231-8. [DOI: 10.1021/ci500666m] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Robert P. Sheridan
- Structural
Chemistry, Merck Research Laboratories, P.O. Box 2000, Rahway, New
Jersey 07065, United States
| | - Daniel R. McMasters
- Structural
Chemistry, Merck Research Laboratories, 33 Avenue Louis Pasteur, Boston, Massachusetts 02115, United States
| | - Johannes H. Voigt
- Structural
Chemistry, Merck Research Laboratories, P.O. Box 2000, Rahway, New
Jersey 07065, United States
| | - Mary Jo Wildey
- In Vitro
Pharmacology, Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| |
Collapse
|
52
|
Pugsley MK, Curtis MJ, Hayes ES. Biophysics and Molecular Biology of Cardiac Ion Channels for the Safety Pharmacologist. Handb Exp Pharmacol 2015; 229:149-203. [PMID: 26091640 DOI: 10.1007/978-3-662-46943-9_7] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Cardiac safety pharmacology is a continuously evolving discipline that uses the basic principles of pharmacology in a regulatory-driven process to generate data to inform risk/benefit assessment of a new chemical entity (NCE). The aim of cardiac safety pharmacology is to characterise the pharmacodynamic/pharmacokinetic (PK/PD) relationship of a drug's adverse effects on the heart using continuously evolving methodology. Unlike Toxicology, safety pharmacology includes within its remit a regulatory requirement to predict the risk of rare cardiotoxic (potentially lethal) events such as torsades de pointes (TdP), which is statistically associated with drug-induced changes in the QT interval of the ECG due to blockade of I Kr or K v11.1 current encoded by hERG. This gives safety pharmacology its unique character. The key issues for the safety pharmacology assessment of a drug on the heart are detection of an adverse effect liability, projection of the data into safety margin calculation and clinical safety monitoring. This chapter will briefly review the current cardiac safety pharmacology paradigm outlined in the ICH S7A and ICH S7B guidance documents and the non-clinical models and methods used in the evaluation of new chemical entities in order to define the integrated risk assessment for submission to regulatory authorities. An overview of how the present cardiac paradigm was developed will be discussed, explaining how it was based upon marketing authorisation withdrawal of many non-cardiovascular compounds due to unanticipated proarrhythmic effects. The role of related biomarkers (of cardiac repolarisation, e.g. prolongation of the QT interval of the ECG) will be considered. We will also provide an overview of the 'non-hERG-centric' concepts utilised in the evolving comprehensive in vitro proarrhythmia assay (CIPA) that details conduct of the proposed ion channel battery test, use of human stem cells and application of in silico models to early cardiac safety assessment. The summary of our current understanding of the triggers of TdP will include the interplay between action potential (AP) prolongation, early and delayed afterdepolarisation and substrates for re-entry arrhythmias.
Collapse
Affiliation(s)
- Michael K Pugsley
- Global Safety Pharmacology and Toxicology/Pathology, Janssen Pharmaceuticals LLC, 1000 Route 202 South, Raritan, NJ, 08869, USA,
| | | | | |
Collapse
|
53
|
Zhang KP, Yang BF, Li BX. Translational toxicology and rescue strategies of the hERG channel dysfunction: biochemical and molecular mechanistic aspects. Acta Pharmacol Sin 2014; 35:1473-84. [PMID: 25418379 PMCID: PMC4261120 DOI: 10.1038/aps.2014.101] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Accepted: 08/20/2014] [Indexed: 01/08/2023]
Abstract
The human ether-à-go-go related gene (hERG) potassium channel is an obligatory anti-target for drug development on account of its essential role in cardiac repolarization and its close association with arrhythmia. Diverse drugs have been removed from the market owing to their inhibitory activity on the hERG channel and their contribution to acquired long QT syndrome (LQTS). Moreover, mutations that cause hERG channel dysfunction may induce congenital LQTS. Recently, an increasing number of biochemical and molecular mechanisms underlying hERG-associated LQTS have been reported. In fact, numerous potential biochemical and molecular rescue strategies are hidden within the biogenesis and regulating network. So far, rescue strategies of hERG channel dysfunction and LQTS mainly include activators, blockers, and molecules that interfere with specific links and other mechanisms. The aim of this review is to discuss the rescue strategies based on hERG channel toxicology from the biochemical and molecular perspectives.
Collapse
Affiliation(s)
- Kai-ping Zhang
- Department of Pharmacology, Harbin Medical University, Harbin, China
- The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China (Key Laboratory of Cardiovascular Research, Ministry of Education), China
| | - Bao-feng Yang
- Department of Pharmacology, Harbin Medical University, Harbin, China
- The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China (Key Laboratory of Cardiovascular Research, Ministry of Education), China
| | - Bao-xin Li
- Department of Pharmacology, Harbin Medical University, Harbin, China
- The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China (Key Laboratory of Cardiovascular Research, Ministry of Education), China
| |
Collapse
|
54
|
Khan JM, Lyon AR, Harding SE. The case for induced pluripotent stem cell-derived cardiomyocytes in pharmacological screening. Br J Pharmacol 2014; 169:304-17. [PMID: 22845396 DOI: 10.1111/j.1476-5381.2012.02118.x] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The current drug screening models are deficient, particularly in detecting cardiac side effects. Human stem cell-derived cardiomyocytes could aid both early cardiotoxicity detection and novel drug discovery. Work over the last decade has generated human embryonic stem cells as potentially accurate sources of human cardiomyocytes, but ethical constraints and poor efficacy in establishing cell lines limit their use. Induced pluripotent stem cells do not require the use of human embryos and have the added advantage of producing patient-specific cardiomyocytes, allowing both generic and disease- and patient-specific pharmacological screening, as well as drug development through disease modelling. A critical question is whether sufficient standards have been achieved in the reliable and reproducible generation of 'adult-like' cardiomyocytes from human fibroblast tissue to progress from validation to safe use in practice and drug discovery. This review will highlight the need for a new experimental system, assess the validity of human induced pluripotent stem cell-derived cardiomyocytes and explore what the future may hold for their use in pharmacology.
Collapse
Affiliation(s)
- Jaffar M Khan
- Royal Brompton and Harefield NHS Trust, London, UK National Heart and Lung Institute, Imperial College, London, UK
| | | | | |
Collapse
|
55
|
Chapleau CA, Lane J, Pozzo-Miller L, Percy AK. Evaluation of current pharmacological treatment options in the management of Rett syndrome: from the present to future therapeutic alternatives. ACTA ACUST UNITED AC 2014; 8:358-69. [PMID: 24050745 DOI: 10.2174/15748847113086660069] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Revised: 02/14/2013] [Accepted: 02/21/2013] [Indexed: 11/22/2022]
Abstract
Neurodevelopmental disorders are a large family of conditions of genetic or environmental origin that are characterized by deficiencies in cognitive and behavioral functions. The therapeutic management of individuals with these disorders is typically complex and is limited to the treatment of specific symptoms that characterize each disorder. The neurodevelopmental disorder Rett syndrome (RTT) is the leading cause of severe intellectual disability in females. Mutations in the gene encoding the transcriptional regulator methyl-CpG-binding protein 2 (MECP2), located on the X chromosome, have been confirmed in more than 95% of individuals meeting diagnostic criteria for classical RTT. RTT is characterized by an uneventful early infancy followed by stagnation and regression of growth, motor, language, and social skills later in development. This review will discuss the genetics, pathology, and symptoms that distinguish RTT from other neurodevelopmental disorders associated with intellectual disability. Because great progress has been made in the basic and clinical science of RTT, the goal of this review is to provide a thorough assessment of current pharmacotherapeutic options to treat the symptoms associated with this disorder. Furthermore, we will highlight recent discoveries made with novel pharmacological interventions in experimental preclinical phases, and which have reversed pathological phenotypes in mouse and cell culture models of RTT and may result in clinical trials.
Collapse
Affiliation(s)
- Christopher A Chapleau
- Department of Pediatrics, CIRC-320, The University of Alabama at Birmingham, 1530 3rd Avenue South, Birmingham, AL 35294-0021, USA.
| | | | | | | |
Collapse
|
56
|
Brudeli B, Navaratnarajah M, Andressen KW, Manfra O, Moltzau LR, Nilsen NO, Levy FO, Klaveness J. Discovery and pharmacological profile of new hydrophilic 5-HT 4 receptor antagonists. Bioorg Med Chem Lett 2014; 24:4598-4602. [DOI: 10.1016/j.bmcl.2014.06.083] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Revised: 06/26/2014] [Accepted: 06/28/2014] [Indexed: 10/25/2022]
|
57
|
Ball P. Efficacy and safety of levofloxacin in the context of other contemporary fluoroquinolones: a review. Curr Ther Res Clin Exp 2014; 64:646-61. [PMID: 24944413 DOI: 10.1016/j.curtheres.2003.11.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/19/2003] [Indexed: 10/26/2022] Open
Abstract
BACKGROUND In recent years, fluoroquinolone research has focused on achieving several goals, including (1) enhanced potency against gram-positive cocci, notably Streptococcus pneumoniae, and anaerobes, while (2) maintaining potency against gram-negative pathogens, (3) optimizing pharmacokinetics and pharmacodynamics (PK/PD), and (4) minimizing potential adverse drug reactions through recognition and avoidance of structural configurations that have characterized earlier, reactive compounds. OBJECTIVE This review examines the efficacy and safety of fluoroquinolones and the specific clinical evidence regarding levofloxacin. METHODS Using published literature collected over time by the author, a review was conducted, focusing on the efficacy and safety profile of levofloxacin and other fluoroquinolones. RESULTS The newer fluoroquinolones have fulfilled many of the research goals described above. Levofloxacin has improved anti-gram-positive potency, PK/PD properties, a proven clinical trial record (particularly for communityacquired pneumonia [CAP]), and an excellent safety profile-in the context of the treatment of >250 million patients worldwide in the past decade. It is licensed for management of drug-resistant S pneumoniae infections in the United States and has gained widespread formulary acceptance and guideline inclusion. Studies assessing levofloxacin for CAP therapy show significant advantages over standard therapy, such as trends toward reduced IV therapy and length of hospitalization, reduced mortality, and significant associated cost reduction. In addition, levofloxacin has proved highly effective in acute exacerbations of chronic bronchitis (AECB), with excellent clinical and bacteriologic results, typical of the class, and significant advantages-in terms of clinical response, overall pathogen eradication, extension of the symptom-free period, and trends toward a reduction in the number of consultation visits and hospitalizations-over standard agents, such as the oral cephalosporins. CONCLUSIONS Levofloxacin offers a combination of documented efficacy and tolerability, and provides an important option for the treatment of bacterial infections, including CAP and AECB, compared with standard agents used in the management of lower respiratory tract infections.
Collapse
Affiliation(s)
- Peter Ball
- Lately University of St. Andrews, Fife, Scotland, United Kingdom
| |
Collapse
|
58
|
On-chip in vitro cell-network pre-clinical cardiac toxicity using spatiotemporal human cardiomyocyte measurement on a chip. Sci Rep 2014; 4:4670. [PMID: 24751527 PMCID: PMC5381194 DOI: 10.1038/srep04670] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Accepted: 03/24/2014] [Indexed: 11/09/2022] Open
Abstract
To overcome the limitations and misjudgments of conventional prediction of arrhythmic cardiotoxicity, we have developed an on-chip in vitro predictive cardiotoxicity assay using cardiomyocytes derived from human stem cells employing a constructive spatiotemporal two step measurement of fluctuation (short-term variability; STV) of cell's repolarization and cell-to-cell conduction time, representing two origins of lethal arrhythmia. Temporal STV of field potential duration (FPD) showed a potential to predict the risks of lethal arrhythmia originated from repolarization dispersion for false negative compounds, which was not correctly predicted by conventional measurements using animal cells, even for non-QT prolonging clinical positive compounds. Spatial STV of conduction time delay also unveiled the proarrhythmic risk of asynchronous propagation in cell networks, whose risk cannot be correctly predicted by single-cell-based measurements, indicating the importance of the spatiotemporal fluctuation viewpoint of in vitro cell networks for precise prediction of lethal arrhythmia reaching clinical assessment such as thorough QT assay.
Collapse
|
59
|
Sievi NA, Clarenbach CF, Camen G, Rossi VA, van Gestel AJR, Kohler M. High prevalence of altered cardiac repolarization in patients with COPD. BMC Pulm Med 2014; 14:55. [PMID: 24690123 PMCID: PMC3976227 DOI: 10.1186/1471-2466-14-55] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Accepted: 03/25/2014] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Altered cardiac repolarization and increased dispersion of repolarization have been identified as risk factors for sudden cardiac death (SCD). The prevalence of and the mechanisms contributing to altered cardiac repolarization are currently unknown in COPD. METHODS In 91 COPD patients, 32 controls matched for age, cardiovascular risk and medication, and 41 healthy subjects, measures of cardiac repolarization and dispersion of repolarization (QTc interval, QT dispersion) were derived from 12-lead electrocardiography (ECG). Prevalence rates of heart rate corrected QT (QTc) >450ms and QT dispersion >60ms were determined to assess the number of subjects at risk for SCD. Univariate and multivariate analyses were used to identify possible factors contributing to altered cardiac repolarization. RESULTS QTc was found to be prolonged in 31.9% and QT dispersion in 24.2% of the COPD patients compared to 12.5% in matched controls and 0% in healthy subjects. The QTc interval was longer in COPD patients compared to matched and healthy controls respectively (437.9 ± 29.5 vs. 420.1 ± 25.3 ms, p = 0.001 and vs. 413.4 ± 18.2 ms, p < 0.001). QT dispersion was significantly increased in COPD patients compared to healthy subjects (45.4 (34.8 , 59.5) vs. 39.7 (29.3 , 54.8) ms, p = 0.049). Only oxygen saturation was independently associated with QTc duration in multivariate analysis (β = -0.29, p = 0.015). CONCLUSION One third of a typical COPD population has altered cardiac repolarization and increased dispersion of repolarization, which may be related to hypoxia. Altered cardiac repolarization may expose these patients to an increased risk for malignant ventricular arrhythmias and SCD.
Collapse
Affiliation(s)
- Noriane A Sievi
- Pulmonary Division, University Hospital Zurich, Zurich, Switzerland
- Institute of Human Movement Sciences and Sport, ETH Zurich, Zurich, Switzerland
| | | | - Giovanni Camen
- Pulmonary Division, University Hospital Zurich, Zurich, Switzerland
| | | | | | - Malcolm Kohler
- Pulmonary Division, University Hospital Zurich, Zurich, Switzerland
- Zurich Centre for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
- Chair Respiratory Medicine, Clinical Director Division of Pulmonology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
60
|
Weeke P, Mosley JD, Hanna D, Delaney JT, Shaffer C, Wells QS, Van Driest S, Karnes JH, Ingram C, Guo Y, Shyr Y, Norris K, Kannankeril PJ, Ramirez AH, Smith JD, Mardis ER, Nickerson D, George AL, Roden DM. Exome sequencing implicates an increased burden of rare potassium channel variants in the risk of drug-induced long QT interval syndrome. J Am Coll Cardiol 2014; 63:1430-7. [PMID: 24561134 DOI: 10.1016/j.jacc.2014.01.031] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Revised: 12/09/2013] [Accepted: 01/07/2014] [Indexed: 10/25/2022]
Abstract
OBJECTIVES The aim of this study was to test the hypothesis that rare variants are associated with drug-induced long QT interval syndrome (diLQTS) and torsades de pointes. BACKGROUND diLQTS is associated with the potentially fatal arrhythmia torsades de pointes. The contribution of rare genetic variants to the underlying genetic framework predisposing to diLQTS has not been systematically examined. METHODS We performed whole-exome sequencing on 65 diLQTS patients and 148 drug-exposed control subjects of European descent. We used rare variant analyses (variable threshold and sequence kernel association test) and gene-set analyses to identify genes enriched with rare amino acid coding (AAC) variants associated with diLQTS. Significant associations were reanalyzed by comparing diLQTS patients with 515 ethnically matched control subjects from the National Heart, Lung, and Blood Grand Opportunity Exome Sequencing Project. RESULTS Rare variants in 7 genes were enriched in the diLQTS patients according to the sequence kernel association test or variable threshold compared with drug-exposed controls (p < 0.001). Of these, we replicated the diLQTS associations for KCNE1 and ACN9 using 515 Exome Sequencing Project control subjects (p < 0.05). A total of 37% of the diLQTS patients also had 1 or more rare AAC variants compared with 21% of control subjects (p = 0.009), in a pre-defined set of 7 congenital long QT interval syndrome (cLQTS) genes encoding potassium channels or channel modulators (KCNE1, KCNE2, KCNH2, KCNJ2, KCNJ5, KCNQ1, AKAP9). CONCLUSIONS By combining whole-exome sequencing with aggregated rare variant analyses, we implicate rare variants in KCNE1 and ACN9 as risk factors for diLQTS. Moreover, diLQTS patients were more burdened by rare AAC variants in cLQTS genes encoding potassium channel modulators, supporting the idea that multiple rare variants, notably across cLQTS genes, predispose to diLQTS.
Collapse
Affiliation(s)
- Peter Weeke
- Department of Medicine, Vanderbilt University, Nashville, Tennessee; Department of Cardiology, Copenhagen University Hospital, Gentofte, Denmark
| | | | - David Hanna
- Department of Genome Sciences, University of Washington, Seattle, Washington
| | | | | | - Quinn S Wells
- Department of Medicine, Vanderbilt University, Nashville, Tennessee
| | - Sara Van Driest
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Jason H Karnes
- Department of Medicine, Vanderbilt University, Nashville, Tennessee
| | - Christie Ingram
- Department of Medicine, Vanderbilt University, Nashville, Tennessee
| | - Yan Guo
- Vanderbilt Technologies for Advanced Genomics Analysis and Research Design, Nashville, Tennessee
| | - Yu Shyr
- Vanderbilt Technologies for Advanced Genomics Analysis and Research Design, Nashville, Tennessee
| | - Kris Norris
- Department of Medicine, Vanderbilt University, Nashville, Tennessee
| | - Prince J Kannankeril
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Andrea H Ramirez
- Department of Medicine, Vanderbilt University, Nashville, Tennessee
| | - Joshua D Smith
- Department of Genome Sciences, University of Washington, Seattle, Washington
| | - Elaine R Mardis
- The Genome Institute, Washington University, St. Louis, Missouri
| | - Deborah Nickerson
- Department of Genome Sciences, University of Washington, Seattle, Washington
| | - Alfred L George
- Departments of Medicine and Pharmacology, Vanderbilt University, Nashville, Tennessee
| | - Dan M Roden
- Departments of Medicine and Pharmacology, Vanderbilt University, Nashville, Tennessee.
| |
Collapse
|
61
|
Shin DS, Park MJ, Lee HA, Lee JY, Chung HC, Yoo DS, Chae CH, Park SJ, Kim KS, Bae MA. A novel assessment of nefazodone-induced hERG inhibition by electrophysiological and stereochemical method. Toxicol Appl Pharmacol 2014; 274:361-71. [DOI: 10.1016/j.taap.2013.12.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Revised: 12/16/2013] [Accepted: 12/16/2013] [Indexed: 11/30/2022]
|
62
|
Abstract
Abnormal functioning of cardiac ion channels can disrupt cardiac myocyte action potentials and thus cause potentially lethal cardiac arrhythmias. Ion channel dysfunction has been observed at all stages in channel ontogeny, from biogenesis to regulation, and arises from genetic or environmental factors, or both. Acquired arrhythmias - including those that are drug induced - are more common than solely inherited arrhythmias but, in some cases, also contain an identifiable genetic component. This interplay between the pharmacology and genetics - known as 'pharmacogenetics' - of cardiac ion channels and the systems that impact them presents both challenges and opportunities to academics, pharmaceutical companies and clinicians seeking to develop and utilize therapies for cardiac rhythm disorders. In this review, we discuss ion channel pharmacogenetics in the context of both causation and treatment of cardiac arrhythmias, focusing on the long QT syndromes.
Collapse
Affiliation(s)
- Geoffrey W Abbott
- Weill Medical College of Cornell University, Greenberg Division of Cardiology, Department of Medicine and Department of Pharmacology, 520 East 70th Street, New York, NY 10021, USA.
| | | |
Collapse
|
63
|
Rao P, Kowey PR. Drug-induced long-QT syndrome and torsade de pointes: an underrated problem? Europace 2013; 16:4-5. [PMID: 24192322 DOI: 10.1093/europace/eut331] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Prashanth Rao
- Division of Cardiovascular Disease, Lankenau Medical Center and Institute for Medical Research, 100 Lancaster Avenue, 558 MOB East, Wynnewood, PA 19096, USA
| | | |
Collapse
|
64
|
Acidic biphenyl derivatives: Synthesis and biological activity of a new series of potent 5-HT4 receptor antagonists. Bioorg Med Chem 2013; 21:7134-45. [DOI: 10.1016/j.bmc.2013.09.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Revised: 08/30/2013] [Accepted: 09/01/2013] [Indexed: 11/20/2022]
|
65
|
Zeng H, Balasubramanian B, Penniman JR, Kinose F, Salata JJ, Lagrutta A. Halide ion effects on human Ether-à-go-go related gene potassium channel properties. Assay Drug Dev Technol 2013; 11:544-50. [PMID: 24147984 DOI: 10.1089/adt.2013.531] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
The human Ether-à-go-go related gene (hERG) potassium channel has been widely used to counter screen potential pharmaceuticals as a biomarker to predict clinical QT prolongation. Thus, higher throughput assays of hERG are valuable for early in vitro screening of drug candidates to minimize failure in later-stage drug development due to this potentially adverse cardiac risk. We have developed a novel method utilizing potassium fluoride to improve throughput of hERG counter screening with an automated patch clamp system, PatchXpress 7000A. In that method, ∼50% substitution of internal Cl(-) with F(-) greatly increases success rate without substantially altering the biophysical properties of the hERG channel or compromising data quality. However, effect of F(-) or other halide ions on hERG channel properties has not been studied in detail. In this study, we examined effects of complete replacement of Cl(-) in internal solution with halide ions, F(-), or Br(-). We found that (1) F(-) slightly shifts the voltage dependence of hERG channel activation to more positive voltages, while Br(-) shifts it to more negative voltages; (2) Br(-) shifts to more positive voltages both the inactivation-voltage relationship and the peak position of channel full activation of hERG; (3) F(-) slows hERG activation, while both F(-) and Br(-) make the channel close faster; (4) neither F(-) nor Br(-) have any effect on hERG inactivation kinetics. In conclusion, compared to Cl(-), F(-) has subtle effect on hERG activation, while Br(-) has distinct effects on certain, but not all biophysical properties of hERG channel.
Collapse
Affiliation(s)
- Haoyu Zeng
- Safety Assessment and Laboratory Animal Resources, Merck Research Laboratories , West Point, Pennsylvania
| | | | | | | | | | | |
Collapse
|
66
|
Veerman CC, Verkerk AO, Blom MT, Klemens CA, Langendijk PNJ, van Ginneken ACG, Wilders R, Tan HL. Slow delayed rectifier potassium current blockade contributes importantly to drug-induced long QT syndrome. Circ Arrhythm Electrophysiol 2013; 6:1002-9. [PMID: 23995305 DOI: 10.1161/circep.113.000239] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Drug-induced long QT syndrome is generally ascribed to inhibition of the cardiac rapid delayed rectifier potassium current (IKr). Effects on the slow delayed rectifier potassium current (IKs) are less recognized. Triggered by a patient who carried the K422T mutation in KCNQ1 (encoding the α-subunit of the IKs channel), who presented with excessive QT prolongation and high serum levels of norfluoxetine, we investigated the effects of fluoxetine and its metabolite norfluoxetine on IKs. METHODS AND RESULTS ECG data from mutation carriers and noncarriers revealed that the K422T mutation per se had mild clinical effects. Patch clamp studies, performed on HEK293 cells, showed that heterozygously expressed K422T KCNQ1/KCNE1 channels had a positive shift in voltage dependence of activation and an increase in deactivation rate. Fluoxetine and its metabolite norfluoxetine both inhibited KCNQ1/KCNE1 current, with norfluoxetine being the most potent. Moreover, norfluoxetine increased activation and deactivation rates. Computer simulations of the effects of norfluoxetine on IKs and IKr demonstrated significant action potential prolongation, to which IKs block contributed importantly. Although the effects of the mutation per se were small, additional IKs blockade by norfluoxetine resulted in more prominent QTc prolongation in mutation carriers than in noncarriers, demonstrating synergistic effects of innate and drug-induced IKs blockade on QTc prolongation. CONCLUSIONS IKs blockade contributes importantly to drug-induced long QT syndrome, especially when repolarization reserve is reduced. Drug safety tests might have to include screening for IKs blockade.
Collapse
Affiliation(s)
- Christiaan C Veerman
- Heart Center, and Departments of Anatomy, Embryology, and Physiology, Cardiology, Hospital Pharmacy, Academic Medical Center, University of Amsterdam, the Netherlands; and Department of Hospital Pharmacy, Reinier de Graaf Group Hospitals, Delft, the Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
67
|
Adimadhyam S, Schumock GT, Walton S, Joo M, McKell J, Lee TA. Risk of arrhythmias associated with ipratropium bromide in children, adolescents, and young adults with asthma: a nested case-control study. Pharmacotherapy 2013; 34:315-23. [PMID: 23918239 DOI: 10.1002/phar.1336] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
STUDY OBJECTIVE To evaluate the risk of arrhythmias associated with inhaled anticholinergic (IAC) use in young patients with asthma. DESIGN Population-based nested case-control study. DATABASE IMS LifeLink Health Plan Claims Database. PATIENTS Patients 5-24 years of age who were diagnosed with asthma and were new users of asthma controller medications were identified between July 1997 and April 2010. Cases were newly diagnosed with arrhythmia and were matched with up to 10 controls based on age, gender, geographic region, and quarter and year of first controller medication dispensing. MEASUREMENTS AND MAIN RESULTS Exposure to IACs was determined in the 180 days prior to the event date, defined as date of arrhythmia claim. Active use was defined as sufficient days' supply of a prescription to extend through the event date. Among 283,429 patients with asthma, 7656 cases were matched to 76,304 controls. Most of those included were female (58.8%) and 12 years or older (73.3%). Active exposure of IACs was observed in 0.69% of cases and 0.18% of controls. Active use was associated with a 1.56-fold increase in arrhythmia risk compared with nonactive users or nonusers (adjusted odds ratio [ORadj ] 1.56, 95% confidence interval [CI] 1.08-2.25]). Risk was highest among active users of ipratropium (ORadj 1.59, 95% CI 1.08-2.33). Active high-dose users of IACs (more than 0.114 mg of ipratropium equivalents) had a 69% increase in risk (ORadj 1.69, 95% CI 1.10-2.59), whereas the added risk for active users receiving low-dose IACs (0.114 mg of ipratropium equivalents or less) was not statistically significant (ORadj 1.22, 95% CI 0.53-2.65). CONCLUSION Use of ipratropium bromide was associated with an increased risk of arrhythmias in 12-24-year-old patients with asthma.
Collapse
Affiliation(s)
- Sruthi Adimadhyam
- Department of Pharmacy Systems Outcomes and Policy, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois
| | | | | | | | | | | |
Collapse
|
68
|
Case scenario: anesthesia-related cardiac arrest in a child with Timothy syndrome. Anesthesiology 2013; 117:1117-26. [PMID: 23011317 DOI: 10.1097/aln.0b013e31826e73b1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
69
|
Liu J, Zhang W, Stashko MA, DeRyckere D, Cummings CT, Hunter D, Yang C, Jayakody CN, Cheng N, Simpson C, Norris-Drouin J, Sather S, Kireev D, Janzen WP, Earp HS, Graham DK, Frye SV, Wang X. UNC1062, a new and potent Mer inhibitor. Eur J Med Chem 2013; 65:83-93. [PMID: 23693152 PMCID: PMC3720808 DOI: 10.1016/j.ejmech.2013.03.035] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Revised: 03/11/2013] [Accepted: 03/21/2013] [Indexed: 12/01/2022]
Abstract
Abnormal activation of Mer kinase has been implicated in the oncogenesis of many human cancers including acute lymphoblastic and myeloid leukemia, non-small cell lung cancer, and glioblastoma. We have discovered a new family of small molecule Mer inhibitors, pyrazolopyrimidine sulfonamides, that potently inhibit the kinase activity of Mer. Importantly, these compounds do not demonstrate significant hERG activity in the PatchXpress assay. Through structure-activity relationship studies, 35 (UNC1062) was identified as a potent (IC50 = 1.1 nM) and selective Mer inhibitor. When applied to live tumor cells, UNC1062 inhibited Mer phosphorylation and colony formation in soft agar. Given the potential of Mer as a therapeutic target, UNC1062 is a promising candidate for further drug development.
Collapse
Affiliation(s)
- Jing Liu
- Center for Integrative Chemical Biology and Drug Discovery, Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA
| | - Weihe Zhang
- Center for Integrative Chemical Biology and Drug Discovery, Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA
| | - Michael A Stashko
- Center for Integrative Chemical Biology and Drug Discovery, Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA
| | - Deborah DeRyckere
- Department of Pediatrics, School of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Christopher T. Cummings
- Department of Pediatrics, School of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Debra Hunter
- Lineberger Comprehensive Cancer Center, Department of Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Chao Yang
- Center for Integrative Chemical Biology and Drug Discovery, Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA
| | - Chatura N. Jayakody
- Center for Integrative Chemical Biology and Drug Discovery, Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA
| | - Nancy Cheng
- Center for Integrative Chemical Biology and Drug Discovery, Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA
| | - Catherine Simpson
- Center for Integrative Chemical Biology and Drug Discovery, Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA
| | - Jacqueline Norris-Drouin
- Center for Integrative Chemical Biology and Drug Discovery, Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA
| | - Susan Sather
- Department of Pediatrics, School of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Dmitri Kireev
- Center for Integrative Chemical Biology and Drug Discovery, Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA
| | - William P. Janzen
- Center for Integrative Chemical Biology and Drug Discovery, Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, Department of Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - H Shelton Earp
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, Department of Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Douglas K. Graham
- Department of Pediatrics, School of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Stephen V. Frye
- Center for Integrative Chemical Biology and Drug Discovery, Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, Department of Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Xiaodong Wang
- Center for Integrative Chemical Biology and Drug Discovery, Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA
| |
Collapse
|
70
|
Abstract
The prevalence of human immunodeficiency virus (HIV) infection among people older than 50 years is increasing. Older HIV-infected patients are particularly at risk for polypharmacy because they often have multiple comorbidities that require pharmacotherapy. Overall, there is not much known with respect to both the impact of aging on medication use in HIV-infected individuals, and the potential for interactions with highly active antiretroviral therapy (HAART) and coadministered medications and its clinical consequences. In this review, we aim to provide an overview of polypharmacy with a focus on its impact on the HIV-infected older adult population and to also provide some clinical considerations in this high-risk population.
Collapse
Affiliation(s)
- Lauren J Gleason
- Division of Geriatrics and Aging, Highland Hospital, Rochester, NY 14620, USA
| | | | | |
Collapse
|
71
|
Pani PP, Trogu E, Maremmani I, Pacini M. QTc interval screening for cardiac risk in methadone treatment of opioid dependence. Cochrane Database Syst Rev 2013; 2013:CD008939. [PMID: 23787716 PMCID: PMC11932215 DOI: 10.1002/14651858.cd008939.pub2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Methadone represents today the gold standard of efficacy for the pharmacological treatment of opioid dependence. Methadone, like many other medications, has been implicated in the prolongation of the rate-corrected QT (QTc) interval of the electrocardiogram (ECG), which is considered a marker for arrhythmias such as torsade de pointes (TdP). Indications on the association between methadone, even at therapeutic dosages, and TdP or sudden cardiac death have been reported. On these bases, consensus and recommendations involving QTc screening of patients receiving methadone treatment have been developed to identify patients with QTc above the thresholds considered at risk for cardiac arrhythmias, and they provide these individuals with alternative treatment (reduction of methadone dosage; provision of alternative opioid agonist treatment; treatment of associated risk factors). OBJECTIVES To evaluate the efficacy and acceptability of QTc screening for preventing cardiac-related morbidity and mortality in methadone-treated opioid dependents. SEARCH METHODS We searched MEDLINE, EMBASE, CINAHL (to April 2013), the Cochrane Central Register of Controlled Trials (CENTRAL), The Cochrane Library Cochrane Drug and Alcohol Review Group Specialised Register (Issue 3, 2013), main electronic sources of ongoing trials, specific trial databases and reference lists of all relevant papers. SELECTION CRITERIA Randomised controlled trials (RCTs), controlled clinical trials (CCTs) and non-randomised studies (cohort studies, controlled before and after studies, interrupted time series studies, case control studies) examining the efficacy of QTc screening for the prevention of methadone-related mortality and morbidity in opioid addicts. DATA COLLECTION AND ANALYSIS Two review authors independently screened and extracted data from studies. MAIN RESULTS The search strategy led to the identification of 872 records. Upon full-text assessment, no study was found to meet the quality criteria used for this review. AUTHORS' CONCLUSIONS It is not possible to draw any conclusions about the effectiveness of QTc screening strategies for preventing cardiac morbidity/mortality in methadone-treated opioid addicts. Research efforts should focus on strengthening the evidence about the effectiveness of widespread implementation of such strategies and clarifying the associated benefits and harms.
Collapse
Affiliation(s)
- Pier Paolo Pani
- Social-Health Division, Health District 8 (ASL 8) Cagliari, Cagliari, Italy.
| | | | | | | |
Collapse
|
72
|
Weeke P, Delaney J, Mosley JD, Wells Q, Van Driest S, Norris K, Kucera G, Stubblefield T, Roden DM. QT variability during initial exposure to sotalol: experience based on a large electronic medical record. Europace 2013; 15:1791-7. [PMID: 23787903 DOI: 10.1093/europace/eut153] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
AIMS A prolonged QT interval is associated with increased risk of Torsades de pointes (TdP) and may be fatal. We sought to investigate the extent to which clinical covariates affect the change in QT interval among 'real-world' patients treated with sotalol and followed in an electronic medical record (EMR) system. METHODS AND RESULTS We used clinical alerts in our EMR system to identify all patients in whom a new prescription for sotalol was written (2001-11). Rate-corrected QT (QTc) was calculated by Bazett's formula. Correlates of sotalol-induced change in the QTc interval and sotalol discontinuation were examined using linear and logistic regression, respectively. Overall, 541 sotalol-exposed patients were identified (n = 200 women, 37%). The mean first sotalol dose was 86 ± 39 mg, age 64 ± 13 years, and BMI 30 ± 7 kg/m(2). Atrial fibrillation/flutter was the predominant indication (92.2%). After initial exposure, the change in the QTc interval from baseline was highly variable: ΔQTc after 2 h = 3 ± 42 ms (P = 0.17) and 11 ± 37 ms after ≥48 h (P < 0.001). Multivariable linear regression analysis identified female gender and age, reduced left ventricular ejection fraction, high sotalol dose, hypertrophic cardiomyopathy, and loop diuretic co-administration as correlates of increased ΔQTc at ≥48 h (P < 0.05 for all). Within 3 days of initiation, 12% discontinued sotalol of which 31% were because of exaggerated QTc prolongation. One percent developed TdP. CONCLUSION In this EMR-based cohort, the increase in QTc with sotalol initiation was highly variable, and multiple clinical factors contributed. These data represent an important step in ongoing work to identify real-world patients likely to tolerate long-term therapy and reinforces the utility of EMR-based cohorts as research tools.
Collapse
Affiliation(s)
- Peter Weeke
- Department of Medicine and Clinical Pharmacology, Vanderbilt University, 1285 Medical Research Building IV, Nashville, TN 37232, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
73
|
Poluzzi E, Raschi E, Koci A, Moretti U, Spina E, Behr ER, Sturkenboom M, De Ponti F. Antipsychotics and torsadogenic risk: signals emerging from the US FDA Adverse Event Reporting System database. Drug Saf 2013; 36:467-479. [PMID: 23553446 PMCID: PMC3664739 DOI: 10.1007/s40264-013-0032-z] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Drug-induced torsades de pointes (TdP) and related clinical entities represent a current regulatory and clinical burden. OBJECTIVE As part of the FP7 ARITMO (Arrhythmogenic Potential of Drugs) project, we explored the publicly available US FDA Adverse Event Reporting System (FAERS) database to detect signals of torsadogenicity for antipsychotics (APs). METHODS Four groups of events in decreasing order of drug-attributable risk were identified: (1) TdP, (2) QT-interval abnormalities, (3) ventricular fibrillation/tachycardia, and (4) sudden cardiac death. The reporting odds ratio (ROR) with 95 % confidence interval (CI) was calculated through a cumulative analysis from group 1 to 4. For groups 1+2, ROR was adjusted for age, gender, and concomitant drugs (e.g., antiarrhythmics) and stratified for AZCERT drugs, lists I and II (http://www.azcert.org , as of June 2011). A potential signal of torsadogenicity was defined if a drug met all the following criteria: (a) four or more cases in group 1+2; (b) significant ROR in group 1+2 that persists through the cumulative approach; (c) significant adjusted ROR for group 1+2 in the stratum without AZCERT drugs; (d) not included in AZCERT lists (as of June 2011). RESULTS Over the 7-year period, 37 APs were reported in 4,794 cases of arrhythmia: 140 (group 1), 883 (group 2), 1,651 (group 3), and 2,120 (group 4). Based on our criteria, the following potential signals of torsadogenicity were found: amisulpride (25 cases; adjusted ROR in the stratum without AZCERT drugs = 43.94, 95 % CI 22.82-84.60), cyamemazine (11; 15.48, 6.87-34.91), and olanzapine (189; 7.74, 6.45-9.30). CONCLUSIONS This pharmacovigilance analysis on the FAERS found 3 potential signals of torsadogenicity for drugs previously unknown for this risk.
Collapse
Affiliation(s)
- Elisabetta Poluzzi
- Department of Medical and Surgical Sciences, Pharmacology Unit, Alma Mater Studiorum, University of Bologna, Via Irnerio, 48, 40126 Bologna, Italy
| | - Emanuel Raschi
- Department of Medical and Surgical Sciences, Pharmacology Unit, Alma Mater Studiorum, University of Bologna, Via Irnerio, 48, 40126 Bologna, Italy
| | - Ariola Koci
- Department of Medical and Surgical Sciences, Pharmacology Unit, Alma Mater Studiorum, University of Bologna, Via Irnerio, 48, 40126 Bologna, Italy
| | - Ugo Moretti
- Clinical Pharmacology Unit, University of Verona, Verona, Italy
| | - Edoardo Spina
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Elijah R. Behr
- Cardiovascular Sciences Research Centre, St. George’s University of London, London, UK
| | | | - Fabrizio De Ponti
- Department of Medical and Surgical Sciences, Pharmacology Unit, Alma Mater Studiorum, University of Bologna, Via Irnerio, 48, 40126 Bologna, Italy
| |
Collapse
|
74
|
Brudeli B, Moltzau LR, Nguyen CHT, Andressen KW, Nilsen NO, Levy FO, Klaveness J. Synthesis and pharmacological properties of a new hydrophilic and orally bioavailable 5-HT4 antagonist. Eur J Med Chem 2013; 64:629-37. [PMID: 23711770 DOI: 10.1016/j.ejmech.2013.03.060] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2012] [Revised: 03/22/2013] [Accepted: 03/27/2013] [Indexed: 12/20/2022]
Abstract
5-HT4 receptor antagonists have been suggested to have clinical potential in treatment of atrial fibrillation, diarrhea-prone irritable bowel syndrome and urinary incontinence. Recently, the use of 5-HT4 antagonists has been suggested to have a therapeutic benefit in heart failure. Affinity for the hERG potassium ion channel and increased risk for prolonged QT intervals and arrhythmias has been observed for several 5-HT4 ligands. Serotonin may also have beneficial effects in the central nervous system (CNS) through stimulation of the 5-HT4 receptor, and reduced distribution of 5-HT4 antagonists to the CNS may therefore be an advantage. Replacing the amide and N-butyl side chain of the 5-HT4 receptor antagonist SB207266 with an ester and a benzyl dimethyl acetic acid group led to compound 9; a hydrophilic 5-HT4 antagonist with excellent receptor binding and low affinity for the hERG potassium ion channel. To increase oral bioavailability of carboxylic acid 9, two different prodrug approaches were applied. The tert-butyl prodrug 11 did not improve bioavailability, and LC-MS analysis revealed unmetabolized prodrug in the systemic circulation. The medoxomil ester prodrug 10 showed complete conversion and sufficient bioavailability of 9 to advance into further preclinical testing for treatment of heart failure.
Collapse
Affiliation(s)
- Bjarne Brudeli
- Drug Discovery Laboratory AS, Oslo Innovation Center, N-0349 Oslo, Norway
| | | | | | | | | | | | | |
Collapse
|
75
|
Carvalho JFS, Louvel J, Doornbos MLJ, Klaasse E, Yu Z, Brussee J, IJzerman AP. Strategies to reduce HERG K+ channel blockade. Exploring heteroaromaticity and rigidity in novel pyridine analogues of dofetilide. J Med Chem 2013; 56:2828-40. [PMID: 23473309 DOI: 10.1021/jm301564f] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Drug-induced blockade of the human ether-a-go-go-related gene K(+) channel (hERG) represents one of the major antitarget concerns in pharmaceutical industry. SAR studies of this ion channel have shed light on the structural requirements for hERG interaction but most importantly may reveal drug design principles to reduce hERG affinity. In the present study, a novel library of neutral and positively charged heteroaromatic derivatives of the class III antiarrhythmic agent dofetilide was synthesized and assessed for hERG affinity in radioligand binding and manual patch clamp assays. Structural modifications of the pyridine moiety, side chain, and peripheral aromatic moieties were evaluated, thereby revealing approaches for reducing hERG binding affinity. In particular, we found that the extra rigidity imposed close to the positively charged pyridine moiety can be very efficient in decreasing hERG affinity.
Collapse
Affiliation(s)
- João F S Carvalho
- Leiden Academic Center for Drug Research, Leiden University, PO Box 9502, 2300 RA Leiden, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
76
|
He JQ, January CT, Thomson JA, Kamp TJ. Human embryonic stem cell-derived cardiomyocytes: drug discovery and safety pharmacology. Expert Opin Drug Discov 2013; 2:739-53. [PMID: 23488962 DOI: 10.1517/17460441.2.5.739] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Human embryonic stem cells (hESCs) can provide potentially unlimited quantities of a wide range of human cell types that can be used in drug discovery and development, basic research and regenerative medicine. In this review, the authors describe the differentiation of hESCs into cardiomyocytes and outline the properties of hESC-derived cardiomyocytes (hESC-CMs), including their cardiac-type action potentials and contractile characteristics. In vitro cellular assays using hESC-CMs, which can be genetically engineered to create target-specific reporters as well as human disease models, will have applications at multiple stages of the drug discovery process. Furthermore, cardiac safety pharmacology assays evaluating the risk of proarrhythmic side effects associated with QT prolongation may be enhanced in their predictive value with the use of hESC-CMs.
Collapse
Affiliation(s)
- Jia-Qiang He
- Cellular Dynamics International, Inc., 525 Science Drive, Suite 200, Madison, WI 53711, USA +1 608 263 4856 ; +1 608 263 0405 ;
| | | | | | | |
Collapse
|
77
|
De Ponti F. Drug development for the irritable bowel syndrome: current challenges and future perspectives. Front Pharmacol 2013; 4:7. [PMID: 23378837 PMCID: PMC3561631 DOI: 10.3389/fphar.2013.00007] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Accepted: 01/10/2013] [Indexed: 12/15/2022] Open
Abstract
Medications are frequently used for the treatment of patients with the irritable bowel syndrome (IBS), although their actual benefit is often debated. In fact, the recent progress in our understanding of the pathophysiology of IBS, accompanied by a large number of preclinical and clinical studies of new drugs, has not been matched by a significant improvement of the armamentarium of medications available to treat IBS. The aim of this review is to outline the current challenges in drug development for IBS, taking advantage of what we have learnt through the Rome process (Rome I, Rome II, and Rome III). The key questions that will be addressed are: (a) do we still believe in the "magic bullet," i.e., a very selective drug displaying a single receptor mechanism capable of controlling IBS symptoms? (b) IBS is a "functional disorder" where complex neuroimmune and brain-gut interactions occur and minimal inflammation is often documented: do we need to target gut motility, visceral sensitivity, or minimal inflammation? (c) are there validated biomarkers (accepted by regulatory agencies) for studies of sensation and motility with experimental medications in humans? (d) do animal models have predictive and translational value? (e) in the era of personalized medicine, does pharmacogenomics applied to these medications already play a role? Finally, this review will briefly outline medications currently used or in development for IBS. It is anticipated that a more focused interaction between basic science investigators, pharmacologists, and clinicians will lead to better treatment of IBS.
Collapse
Affiliation(s)
- Fabrizio De Ponti
- Department of Medical and Surgical Sciences, University of BolognaBologna, Italy
| |
Collapse
|
78
|
Kohonen P, Benfenati E, Bower D, Ceder R, Crump M, Cross K, Grafström RC, Healy L, Helma C, Jeliazkova N, Jeliazkov V, Maggioni S, Miller S, Myatt G, Rautenberg M, Stacey G, Willighagen E, Wiseman J, Hardy B. The ToxBank Data Warehouse: Supporting the Replacement of In Vivo Repeated Dose Systemic Toxicity Testing. Mol Inform 2013; 32:47-63. [PMID: 27481023 DOI: 10.1002/minf.201200114] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Accepted: 11/27/2012] [Indexed: 12/12/2022]
Abstract
The aim of the SEURAT-1 (Safety Evaluation Ultimately Replacing Animal Testing-1) research cluster, comprised of seven EU FP7 Health projects co-financed by Cosmetics Europe, is to generate a proof-of-concept to show how the latest technologies, systems toxicology and toxicogenomics can be combined to deliver a test replacement for repeated dose systemic toxicity testing on animals. The SEURAT-1 strategy is to adopt a mode-of-action framework to describe repeated dose toxicity, combining in vitro and in silico methods to derive predictions of in vivo toxicity responses. ToxBank is the cross-cluster infrastructure project whose activities include the development of a data warehouse to provide a web-accessible shared repository of research data and protocols, a physical compounds repository, reference or "gold compounds" for use across the cluster (available via wiki.toxbank.net), and a reference resource for biomaterials. Core technologies used in the data warehouse include the ISA-Tab universal data exchange format, REpresentational State Transfer (REST) web services, the W3C Resource Description Framework (RDF) and the OpenTox standards. We describe the design of the data warehouse based on cluster requirements, the implementation based on open standards, and finally the underlying concepts and initial results of a data analysis utilizing public data related to the gold compounds.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Lyn Healy
- National Institute for Biological Standards and Control, Potters Bar, UK
| | | | | | | | - Silvia Maggioni
- Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | | | | | | | - Glyn Stacey
- National Institute for Biological Standards and Control, Potters Bar, UK
| | | | | | | |
Collapse
|
79
|
Park MJ, Lee KR, Shin DS, Chun HS, Kim CH, Ahn SH, Bae MA. Predicted drug-induced bradycardia related cardio toxicity using a zebrafish in vivo model is highly correlated with results from in vitro tests. Toxicol Lett 2013; 216:9-15. [DOI: 10.1016/j.toxlet.2012.10.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2012] [Revised: 10/26/2012] [Accepted: 10/29/2012] [Indexed: 11/30/2022]
|
80
|
Prediction of hERG Potassium Channel Blocking Actions Using Combination of Classification and Regression Based Models: A Mixed Descriptors Approach. Mol Inform 2012; 31:879-94. [DOI: 10.1002/minf.201200039] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Accepted: 11/15/2012] [Indexed: 11/07/2022]
|
81
|
Lee HA, Kim KS, Hyun SA, Park SG, Kim SJ. Wide spectrum of inhibitory effects of sertraline on cardiac ion channels. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2012; 16:327-32. [PMID: 23118556 PMCID: PMC3484517 DOI: 10.4196/kjpp.2012.16.5.327] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2012] [Revised: 09/11/2012] [Accepted: 09/11/2012] [Indexed: 01/21/2023]
Abstract
Sertraline is a commonly used antidepressant of the selective serotonin reuptake inhibitors (SSRIs) class. In these experiments, we have used the whole cell patch clamp technique to examine the effects of sertraline on the major cardiac ion channels expressed in HEK293 cells and the native voltage-gated Ca(2+) channels in rat ventricular myocytes. According to the results, sertraline is a potent blocker of cardiac K(+) channels, such as hERG, I(Ks) and I(K1). The rank order of inhibitory potency was hERG >I(K1)> I(Ks) with IC(50) values of 0.7, 10.5, and 15.2 µM, respectively. In addition to K(+) channels, sertraline also inhibited I(Na) and I(Ca), and the IC(50) values are 6.1 and 2.6 µM, respectively. Modification of these ion channels by sertraline could induce changes of the cardiac action potential duration and QT interval, and might result in cardiac arrhythmia.
Collapse
Affiliation(s)
- Hyang-Ae Lee
- Next-Generation Pharmaceutical Research Center, Korea Institute of Toxicology, Korea Research Institute of Chemical Technology, Daejeon 305-600, Korea. ; Department of Physiology and Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 110-799, Korea
| | | | | | | | | |
Collapse
|
82
|
Abstract
Induced pluripotent stem cells (iPSCs) hold great hopes for therapeutic application in various diseases. Although ongoing research is dedicated to achieving clinical translation of iPSCs, further understanding of the mechanisms that underlie complex pathogenic conditions is required. Compared with other classical models for studying diseases, iPSCs provide considerable advantages. A newly emerging application of iPSCs is in vitro disease modeling, which can significantly improve the never-ending search for new pharmacological cures. Here, we will discuss current efforts to create iPSC-dependent patient-specific disease models. Furthermore, we will review the use of iPSCs for development and testing of new therapeutic agents and the implications for high-throughput drug screening.
Collapse
Affiliation(s)
- Antje D. Ebert
- Department of Medicine (Division of Cardiology), Stanford University, Stanford, California, USA
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California, USA
| | - Ping Liang
- Department of Medicine (Division of Cardiology), Stanford University, Stanford, California, USA
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California, USA
| | - Joseph C. Wu
- Department of Medicine (Division of Cardiology), Stanford University, Stanford, California, USA
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, California, USA
| |
Collapse
|
83
|
Dossetter AG, Bowyer J, Cook CR, Crawford JJ, Finlayson JE, Heron NM, Heyes C, Highton AJ, Hudson JA, Jestel A, Krapp S, MacFaul PA, McGuire TM, Morley AD, Morris JJ, Page KM, Ribeiro LR, Sawney H, Steinbacher S, Smith C. Isosteric replacements for benzothiazoles and optimisation to potent Cathepsin K inhibitors free from hERG channel inhibition. Bioorg Med Chem Lett 2012; 22:5563-8. [DOI: 10.1016/j.bmcl.2012.07.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2012] [Revised: 06/29/2012] [Accepted: 07/04/2012] [Indexed: 11/29/2022]
|
84
|
Abstract
The critical care physician is often called to care for poisoned patients. This article reviews the general approach to the poisoned patient, specifically focusing on the utility of the toxidrome. A toxidrome is a constellation of findings, either from the physical examination or from ancillary testing, which may result from any poison. There are numerous toxidromes defined in the medical literature. This article focuses on the more common toxidromes described in clinical toxicology. Although these toxidromes can aid the clinician in narrowing the differential diagnosis, care must be exercised to realize the exceptions and limitations associated with each.
Collapse
Affiliation(s)
- Christopher P Holstege
- Division of Medical Toxicology, Department of Emergency Medicine, Blue Ridge Poison Center, University of Virginia Health System, University of Virginia School of Medicine, PO Box 800774, Charlottesville, VA 22908-0774, USA.
| | | |
Collapse
|
85
|
|
86
|
Vandenberg JI, Perry MD, Perrin MJ, Mann SA, Ke Y, Hill AP. hERG K+ Channels: Structure, Function, and Clinical Significance. Physiol Rev 2012; 92:1393-478. [DOI: 10.1152/physrev.00036.2011] [Citation(s) in RCA: 463] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The human ether-a-go-go related gene (hERG) encodes the pore-forming subunit of the rapid component of the delayed rectifier K+ channel, Kv11.1, which are expressed in the heart, various brain regions, smooth muscle cells, endocrine cells, and a wide range of tumor cell lines. However, it is the role that Kv11.1 channels play in the heart that has been best characterized, for two main reasons. First, it is the gene product involved in chromosome 7-associated long QT syndrome (LQTS), an inherited disorder associated with a markedly increased risk of ventricular arrhythmias and sudden cardiac death. Second, blockade of Kv11.1, by a wide range of prescription medications, causes drug-induced QT prolongation with an increase in risk of sudden cardiac arrest. In the first part of this review, the properties of Kv11.1 channels, including biogenesis, trafficking, gating, and pharmacology are discussed, while the second part focuses on the pathophysiology of Kv11.1 channels.
Collapse
Affiliation(s)
- Jamie I. Vandenberg
- Mark Cowley Lidwill Research Programme in Cardiac Electrophysiology, Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia; St Vincent's Clinical School, University of New South Wales, New South Wales, Australia; and University of Ottawa Heart Institute, Ottawa, Canada
| | - Matthew D. Perry
- Mark Cowley Lidwill Research Programme in Cardiac Electrophysiology, Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia; St Vincent's Clinical School, University of New South Wales, New South Wales, Australia; and University of Ottawa Heart Institute, Ottawa, Canada
| | - Mark J. Perrin
- Mark Cowley Lidwill Research Programme in Cardiac Electrophysiology, Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia; St Vincent's Clinical School, University of New South Wales, New South Wales, Australia; and University of Ottawa Heart Institute, Ottawa, Canada
| | - Stefan A. Mann
- Mark Cowley Lidwill Research Programme in Cardiac Electrophysiology, Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia; St Vincent's Clinical School, University of New South Wales, New South Wales, Australia; and University of Ottawa Heart Institute, Ottawa, Canada
| | - Ying Ke
- Mark Cowley Lidwill Research Programme in Cardiac Electrophysiology, Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia; St Vincent's Clinical School, University of New South Wales, New South Wales, Australia; and University of Ottawa Heart Institute, Ottawa, Canada
| | - Adam P. Hill
- Mark Cowley Lidwill Research Programme in Cardiac Electrophysiology, Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia; St Vincent's Clinical School, University of New South Wales, New South Wales, Australia; and University of Ottawa Heart Institute, Ottawa, Canada
| |
Collapse
|
87
|
Shakibfar S, Graff C, Ehlers LH, Toft E, Kanters JK, Struijk JJ. Assessing common classification methods for the identification of abnormal repolarization using indicators of T-wave morphology and QT interval. Comput Biol Med 2012; 42:485-91. [DOI: 10.1016/j.compbiomed.2012.01.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2011] [Revised: 01/03/2012] [Accepted: 01/06/2012] [Indexed: 10/14/2022]
|
88
|
DNA and modified vaccinia virus Ankara vaccines encoding multiple cytotoxic and helper T-lymphocyte epitopes of human immunodeficiency virus type 1 (HIV-1) are safe but weakly immunogenic in HIV-1-uninfected, vaccinia virus-naive adults. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2012; 19:649-58. [PMID: 22398243 DOI: 10.1128/cvi.00038-12] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
We evaluated a DNA plasmid-vectored vaccine and a recombinant modified vaccinia virus Ankara vaccine (MVA-mBN32), each encoding cytotoxic and helper T-lymphocyte epitopes of human immunodeficiency virus type 1 (HIV-1) in a randomized, double-blinded, placebo-controlled trial in 36 HIV-1-uninfected adults using a heterologous prime-boost schedule. HIV-1-specific cellular immune responses, measured as interleukin-2 and/or gamma interferon production, were induced in 1 (4%) of 28 subjects after the first MVA-mBN32 immunization and in 3 (12%) of 25 subjects after the second MVA-mBN32 immunization. Among these responders, polyfunctional T-cell responses, including the production of tumor necrosis factor alpha and perforin, were detected. Vaccinia virus-specific antibodies were induced to the MVA vector in 27 (93%) of 29 and 26 (93%) of 28 subjects after the first and second immunizations with MVA-mBN32. These peptide-based vaccines were safe but were ineffective at inducing HIV-1-specific immune responses and induced much weaker responses than MVA vaccines expressing the entire open reading frames of HIV-1 proteins.
Collapse
|
89
|
Kågström J, Laumola EL, Poijes N, Johansson M, Ericson AC. Assessment of the effects of changes in body temperature on cardiac electrophysiology in anaesthetised guinea pigs. J Pharmacol Toxicol Methods 2011; 65:1-7. [PMID: 22178983 DOI: 10.1016/j.vascn.2011.11.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2011] [Revised: 11/28/2011] [Accepted: 11/29/2011] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Anaesthetised guinea pigs are commonly used within Safety Pharmacology to evaluate drug effects on cardiac electrophysiology. However, anesthesia compromises the ability to thermoregulate, which can be further challenged when more invasive surgery is required. As anaesthetised animals are often used when screening for cardiotoxicity, thereby influencing go/no-go decisions, we wanted to quantify the impact of small temperature changes on the recorded electrophysiological parameters. METHODS Male guinea pigs were anaesthetised by pentobarbital, placed on a pre-heated table and a rectal thermistor inserted for monitoring of body temperature. After intubation animals were vagotomised and β-blocked, and lead II ECG needle electrodes attached. Following thoracotomy an atrial pacing electrode was attached and a suction MAP electrode positioned on the ventricular epicardium. In control animals temperature was kept constant (38.1±0.1°C) over the duration of the experiment. Animals in one group were slowly warmed to 41.9°C by a heating plate and a heating lamp, and in another group slowly cooled to 34.4°C by turning off all heating equipment. MAP duration at 90% repolarisation (MAPD90), AV conduction, ECG and body temperature were recorded during cardiac pacing every 5min up to 50min. RESULTS No time-dependent changes were seen in the control group. In contrast, a linear correlation was found between changes in body temperature and MAPD90, AV conduction, QTc and QRS intervals. For each degree temperature fell below 38°C MAPD90 was prolonged by 6.1ms, and for each degree above 38°C MAPD90 was shortened by 5.3ms. Corresponding changes were seen for QTc interval and AV conduction time, while effects on the QRS interval were smaller. DISCUSSION The data highlights the importance of carefully controlling body temperature when performing electrophysiological recordings in laboratory animals. A change by a single degree can affect electrophysiological parameters by 5-10%, thus increasing the risk for a false positive or negative interpretation of cardiotoxicity.
Collapse
Affiliation(s)
- Jens Kågström
- Safety Pharmacology, Safety Assessment, AstraZeneca R&D Södertälje, S-151 85 Södertälje, Sweden.
| | | | | | | | | |
Collapse
|
90
|
Du-Cuny L, Chen L, Zhang S. A critical assessment of combined ligand- and structure-based approaches to HERG channel blocker modeling. J Chem Inf Model 2011; 51:2948-60. [PMID: 21902220 DOI: 10.1021/ci200271d] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Blockade of human ether-à-go-go related gene (hERG) channel prolongs the duration of the cardiac action potential and is a common reason for drug failure in preclinical safety trials. Therefore, it is of great importance to develop robust in silico tools to predict potential hERG blockers in the early stages of drug discovery and development. Herein we described comprehensive approaches to assess the discrimination of hERG-active and -inactive compounds by combining quantitative structure-activity relationship (QSAR) modeling, pharmacophore analysis, and molecular docking. Our consensus models demonstrated high-predictive capacity and improved enrichment and could correctly classify 91.8% of 147 hERG blockers from 351 inactives. To further enhance our modeling effort, hERG homology models were constructed, and molecular docking studies were conducted, resulting in high correlations (R² = 0.81) between predicted and experimental pIC₅₀s. We expect our unique models can be applied to efficient screening for hERG blockades, and our extensive understanding of the hERG-inhibitor interactions will facilitate the rational design of drugs devoid of hERG channel activity and hence with reduced cardiac toxicities.
Collapse
Affiliation(s)
- Lei Du-Cuny
- Integrated Molecular Discovery Laboratory, Department of Experimental Therapeutics, The University of Texas, M.D. Anderson Cancer Center, 1901 East Rd., Houston, Texas, USA
| | | | | |
Collapse
|
91
|
Martin JA, Campbell A, Killip T, Kotz M, Krantz MJ, Kreek MJ, McCarroll BA, Mehta D, Payte JT, Stimmel B, Taylor T, Haigney MCP, Wilford BB. QT interval screening in methadone maintenance treatment: report of a SAMHSA expert panel. J Addict Dis 2011; 30:283-306. [PMID: 22026519 PMCID: PMC4078896 DOI: 10.1080/10550887.2011.610710] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In an effort to enhance patient safety in opioid treatment programs, the Substance Abuse and Mental Health Saervices Administration convened a multi-disciplinary Expert Panel on the Cardiac Effects of Methadone. Panel members (Appendix A) reviewed the literature, regulatory actions, professional guidances, and opioid treatment program experiences regarding adverse cardiac events associated with methadone. The Panel concluded that, to the extent possible, every opioid treatment program should have a universal Cardiac Risk Management Plan (incorporating clinical assessment, electrocardiogram assessment, risk stratification, and prevention of drug interactions) for all patients and should strongly consider patient-specific risk minimization strategies (such as careful patient monitoring, obtaining electrocardiograms as indicated by a particular patient's risk profile, and adjusting the methadone dose as needed) for patients with identified risk factors for adverse cardiac events. The Panel also suggested specific modifications to informed consent documents, patient education, staff education, and methadone protocols.
Collapse
|
92
|
Guo L, Abrams RMC, Babiarz JE, Cohen JD, Kameoka S, Sanders MJ, Chiao E, Kolaja KL. Estimating the risk of drug-induced proarrhythmia using human induced pluripotent stem cell-derived cardiomyocytes. Toxicol Sci 2011; 123:281-9. [PMID: 21693436 DOI: 10.1093/toxsci/kfr158] [Citation(s) in RCA: 213] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Improved in vitro systems for predicting drug-induced toxicity are needed in the pharmaceutical and biotechnology industries to decrease late-stage drug attrition. One unmet need is an early screen for cardiotoxicity, which accounts for about one third of safety-based withdrawn pharmaceuticals. Herein, the first published report of a high-throughput functional assay employing a monolayer of beating human induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) is described, detailing a model that accurately detects drug-induced cardiac abnormalities. Using 96-well plates with interdigitated electrode arrays that assess impedance, the rhythmic, synchronous contractions of the iPSC-CMs were detected. Treatment of the iPSC-CMs with 28 different compounds with known cardiac effects resulted in compound-specific changes in the beat rate and/or the amplitude of the impedance measurement. Changes in impedance for the compounds tested were comparable with the results from a related technology, electric field potential assessment obtained from microelectrode arrays. Using the results from the set of compounds, an index of drug-induced arrhythmias was calculated, enabling the determination of a drug's proarrhythmic potential. This system of interrogating human cardiac function in vitro opens new opportunities for predicting cardiac toxicity and studying cardiac biology.
Collapse
Affiliation(s)
- Liang Guo
- Early and Investigative Safety, Nonclinical Safety, Hoffmann-La Roche, Nutley, New Jersey 07110, USA
| | | | | | | | | | | | | | | |
Collapse
|
93
|
Novel ECG markers for ventricular repolarization: Is the QT interval obsolete? Heart Rhythm 2011; 8:1044-5. [PMID: 21349350 DOI: 10.1016/j.hrthm.2011.02.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2011] [Indexed: 11/22/2022]
|
94
|
van Noord C, Eijgelsheim M, Stricker BHC. Drug- and non-drug-associated QT interval prolongation. Br J Clin Pharmacol 2011; 70:16-23. [PMID: 20642543 DOI: 10.1111/j.1365-2125.2010.03660.x] [Citation(s) in RCA: 191] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Sudden cardiac death is among the most common causes of cardiovascular death in developed countries. The majority of sudden cardiac deaths are caused by acute ventricular arrhythmia following repolarization disturbances. An important risk factor for repolarization disturbances is use of QT prolonging drugs, probably partly explained by gene-drug interactions. In this review, we will summarize QT interval physiology, known risk factors for QT prolongation, including drugs and the contribution of pharmacogenetics. The long QT syndrome can be congenital or acquired. The congenital long QT syndrome is caused by mutations in ion channel subunits or regulatory protein coding genes and is a rare monogenic disorder with a mendelian pattern of inheritance. Apart from that, several common genetic variants that are associated with QT interval duration have been identified. Acquired QT prolongation is more prevalent than the congenital form. Several risk factors have been identified with use of QT prolonging drugs as the most frequent cause. Most drugs that prolong the QT interval act by blocking hERG-encoded potassium channels, although some drugs mainly modify sodium channels. Both pharmacodynamic as well as pharmacokinetic mechanisms may be responsible for QT prolongation. Pharmacokinetic interactions often involve drugs that are metabolized by cytochrome P450 enzymes. Pharmacodynamic gene-drug interactions are due to genetic variants that potentiate the QT prolonging effect of drugs. QT prolongation, often due to use of QT prolonging drugs, is a major public health issue. Recently, common genetic variants associated with QT prolongation have been identified. Few pharmacogenetic studies have been performed to establish the genetic background of acquired QT prolongation but additional studies in this newly developing field are warranted.
Collapse
Affiliation(s)
- Charlotte van Noord
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | | |
Collapse
|
95
|
A step towards characterisation of electrophysiological profile of torsadogenic drugs. J Pharmacol Toxicol Methods 2011; 63:269-78. [PMID: 21224008 DOI: 10.1016/j.vascn.2011.01.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2010] [Revised: 08/27/2010] [Accepted: 01/03/2011] [Indexed: 12/16/2022]
Abstract
INTRODUCTION In a previous study, two electrophysiological patterns for torsadogenic drugs were characterised in the model of isolated canine Purkinje fibres from their respective effects on action potential. This study was designed to elucidate the possible mechanisms underlying these two electrophysiological profiles. METHODS Effects of representative torsadogenic agents and non torsadogenic drugs on I(Kr), I(Ks), I(K1), I(Na) and I(CaL) were studied in transfected HEK 293 cells using the path-clamp method as well as in conscious beagle dogs and cynomolgus monkeys by telemetry. RESULTS Patch-clamp studies confirmed that torsadogenic molecules could be discriminated into at least two subgroups. The first subgroup can be defined as apparently pure I(Kr) blockers. The second subgroup can be defined as I(Kr) blockers with ancillary properties on sodium and/or calcium channels which counterbalance the I(Kr) prolongation component. This discrimination is transposable to the telemetered cynomolgus monkey model in terms of QT prolongation but not to the telemetered beagle dog model. This latter inter-species difference could be related to the sympathetic/parasympathetic balance and could involve reserve repolarisation dependent mechanisms. DISCUSSION The confirmation that torsadogenic drugs might have at least two different electrophysiological profiles should be taken into consideration in preclinical safety pharmacology studies because it increases the value of the cynomolgus monkey model in two particular situations: firstly when an NCE causes sympathetic activation and secondly, when an NCE exhibits a pure I(Kr) blocker pattern independently of its potency to block HERG channels.
Collapse
|
96
|
Kawai Y, Tsukamoto S, Ito J, Akimoto K, Takahashi M. A Risk Assessment of Human Ether-a-Go-Go-Related Gene Potassium Channel Inhibition by Using Lipophilicity and Basicity for Drug Discovery. Chem Pharm Bull (Tokyo) 2011; 59:1110-6. [DOI: 10.1248/cpb.59.1110] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Yukinori Kawai
- Drug Metabolism & Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd
| | - Shinsaku Tsukamoto
- Drug Metabolism & Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd
| | - Junko Ito
- Drug Metabolism & Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd
| | - Katsuya Akimoto
- Drug Metabolism & Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd
| | - Masayuki Takahashi
- Drug Metabolism & Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd
| |
Collapse
|
97
|
Hreiche R, Plante I, Drolet B, Morissette P, Turgeon J. Lengthening of cardiac repolarization in isolated guinea pigs hearts by sequential or concomitant administration of two IKr blockers. J Pharm Sci 2010; 100:2469-81. [PMID: 21491454 DOI: 10.1002/jps.22437] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2010] [Revised: 11/15/2010] [Accepted: 11/18/2010] [Indexed: 01/08/2023]
Abstract
Block of I(Kr) is of major concern in drug safety. The objective of this study was to assess prolongation of cardiac repolarization during the combined use of two I(Kr) blockers when administered concomitantly or sequentially. (1) When isolated hearts from male guinea pigs were perfused concomitantly with two I(Kr) blockers, prolongation of monophasic action potential duration measured at 90% (MAPD(90)) was less than the summation of effects observed for each drug perfused alone. (2) In sequential administration, when ketoconazole or erythromycin was perfused first, they antagonized MAPD(90)-prolonging effects of domperidone. This effect was absent when domperidone or dofetilide was perfused first. Patch-clamp experiments confirmed that the order of sequential perfusion impacts the decrease in HERG tail amplitude. In conclusion, this study does not support the concept that potentiation of drug effects is observed during the combined administration of two I(Kr) blockers. Furthermore, order of administration of two I(Kr) blockers together may be an important factor in drug-induced long QT syndrome.
Collapse
Affiliation(s)
- Raymond Hreiche
- Faculty of Pharmacy, Université de Montréal, Montreal, Quebec, Canada
| | | | | | | | | |
Collapse
|
98
|
Perrin-Terrin A, Pathak A, Lapeyre-Mestre M. QT interval prolongation: prevalence, risk factors and pharmacovigilance data among methadone-treated patients in France. Fundam Clin Pharmacol 2010; 25:503-10. [PMID: 20825488 DOI: 10.1111/j.1472-8206.2010.00871.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Several reports have suggested the role of methadone in QT prolongation, Torsades de Pointes (TdP), and ventricular arrhythmia. The aim of this study was to identify the frequency of QT interval prolongation and QT dispersion among a population of addict patients treated with methadone, completed by the description of cardiac events reported to the French pharmacovigilance system. QT interval (corrected with the Bazett's method) and dispersion were measured on a cohort of patients treated with methadone during their visit to addiction specialized centers in Southwest of France. Whenever possible, methadone blood concentrations were measured. In addition, reports concerning methadone were searched in the French Pharmacovigilance database from 1996 to 2007. The cohort included 42 patients with mean QTc length of 414.3 ± 28.5 ms and mean QT dispersion of 66.0 ± 41.1 ms. None of the patient presented a QTc above 500 ms. These values were higher in patients with recent increase in methadone dose, use of tobacco or drugs prolonging QT, and history of cardiac disease. We found five cases of QT prolongation (3 TdP) and seven cases of sudden deaths in the French pharmacovigilance database (2.2% of methadone reports). Most of these events occurred after introducing methadone or another drug potentially interacting with methadone. Methadone can prolong the QT interval at dose usually used for addiction maintenance treatment in France. A cardiac evaluation, with at least a systematic ECG, could be useful during methadone treatment, particularly in case of recent modification of the regimen.
Collapse
Affiliation(s)
- Aurélie Perrin-Terrin
- Unité de Pharmacoépidémiologie EA3696, Inserm, U558, Faculté de Médecine Purpan, Université de Toulouse, UPS, 37 Allées Jules Guesde; F-31000 Toulouse, France
| | | | | |
Collapse
|
99
|
Chartrand É, Arnold AA, Gravel A, Jenna S, Marcotte I. Potential role of the membrane in hERG channel functioning and drug-induced long QT syndrome. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2010; 1798:1651-62. [DOI: 10.1016/j.bbamem.2010.05.019] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2009] [Revised: 04/21/2010] [Accepted: 05/17/2010] [Indexed: 11/16/2022]
|
100
|
Shukla SJ, Huang R, Austin CP, Xia M. The future of toxicity testing: a focus on in vitro methods using a quantitative high-throughput screening platform. Drug Discov Today 2010; 15:997-1007. [PMID: 20708096 DOI: 10.1016/j.drudis.2010.07.007] [Citation(s) in RCA: 179] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2010] [Revised: 06/08/2010] [Accepted: 07/30/2010] [Indexed: 01/16/2023]
Abstract
The US Tox21 collaborative program represents a paradigm shift in toxicity testing of chemical compounds from traditional in vivo tests to less expensive and higher throughput in vitro methods to prioritize compounds for further study, identify mechanisms of action and ultimately develop predictive models for adverse health effects in humans. The NIH Chemical Genomics Center (NCGC) is an integral component of the Tox21 collaboration owing to its quantitative high-throughput screening (qHTS) paradigm, in which titration-based screening is used to profile hundreds of thousands of compounds per week. Here, we describe the Tox21 collaboration, qHTS-based compound testing and the various Tox21 screening assays that have been validated and tested at the NCGC to date.
Collapse
Affiliation(s)
- Sunita J Shukla
- NIH Chemical Genomics Center, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892-3370, USA
| | | | | | | |
Collapse
|