Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
J Pharmacol Toxicol Methods ; 123: 107270, 2023.
Article in English | MEDLINE | ID: mdl-37164235

ABSTRACT

The ICH E14/S7B Questions and Answers (Q&As) guideline introduces the concept of a "double negative" nonclinical scenario (negative hERG assay and negative in vivo QTc study) to demonstrate that a drug does not produce a clinically relevant QT prolongation (i.e., no QT liability). This nonclinical "double negative" data package, along with negative Phase 1 clinical QTc data, may be sufficient to substitute for a clinical Thorough QT (TQT) study in some specific cases. While standalone GLP in vivo cardiovascular studies in non-rodent species are standard practice during nonclinical drug development for small molecule programs, a variety of approaches to the design, conduct, analysis and interpretation are utilized across pharmaceutical companies and contract research organizations (CROs) that may, in some cases, negatively impact the stringent sensitivity needed to fulfill the new Q&As. Subject matter experts from both Pharma and CROs have collaborated to recommend best practices for more robust nonclinical cardiovascular telemetry studies in non-rodent species, with input from clinical and regulatory experts. The aim was to increase consistency and harmonization across the industry and to ensure delivery of high quality nonclinical QTc data to meet the proposed sensitivities defined within the revised ICH E14/S7B Q&As guideline (Q&As 5.1 and 6.1). The detailed best practice recommendations presented here cover the design and execution of the safety pharmacology cardiovascular study, including optimal methods for acquiring, analyzing, reporting, and interpreting the resulting QTc and pharmacokinetic data to allow for direct comparison to clinical exposures and assessment of safety margin for QTc prolongation.


Subject(s)
Cardiovascular System , Long QT Syndrome , Humans , Long QT Syndrome/chemically induced , Long QT Syndrome/diagnosis , Telemetry , Electrocardiography
2.
J Pharmacol Toxicol Methods ; 121: 107265, 2023.
Article in English | MEDLINE | ID: mdl-36997076

ABSTRACT

Recent updates and modifications to the clinical ICH E14 and nonclinical ICH S7B guidelines, which both relate to the evaluation of drug-induced delayed repolarization risk, provide an opportunity for nonclinical in vivo electrocardiographic (ECG) data to directly influence clinical strategies, interpretation, regulatory decision-making and product labeling. This opportunity can be leveraged with more robust nonclinical in vivo QTc datasets based upon consensus standardized protocols and experimental best practices that reduce variability and optimize QTc signal detection, i.e., demonstrate assay sensitivity. The immediate opportunity for such nonclinical studies is when adequate clinical exposures (e.g., supratherapeutic) cannot be safely achieved, or other factors limit the robustness of the clinical QTc evaluation, e.g., the ICH E14 Q5.1 and Q6.1 scenarios. This position paper discusses the regulatory historical evolution and processes leading to this opportunity and details the expectations of future nonclinical in vivo QTc studies of new drug candidates. The conduct of in vivo QTc assays that are consistently designed, executed and analyzed will lead to confident interpretation, and increase their value for clinical QTc risk assessment. Lastly, this paper provides the rationale and basis for our companion article which describes technical details on in vivo QTc best practices and recommendations to achieve the goals of the new ICH E14/S7B Q&As, see Rossman et al., 2023 (this journal).


Subject(s)
Long QT Syndrome , Humans , Long QT Syndrome/chemically induced , Long QT Syndrome/diagnosis , Drugs, Investigational/adverse effects , Electrocardiography , Risk Assessment , Biological Assay
3.
J Pharmacol Toxicol Methods ; 111: 107109, 2021.
Article in English | MEDLINE | ID: mdl-34416395

ABSTRACT

INTRODUCTION: A successful integration of automated blood sampling (ABS) into the telemetry instrumented canine cardiovascular model is presented in this study. This combined model provides an efficient means to quickly gain understanding of potential effects on key cardiovascular parameters in dog while providing a complete Pharmacokinetic/Pharmacodynamic (PK/PD) profile for discovery compounds without handling artifacts, reducing the need for a separate pharmacokinetic study. METHODS: Male beagle dogs were chronically implanted with telemetry devices (PhysioTel™ model D70-PCTP) and vascular access ports (SPMID-GRIDAC-5NC). BASi Culex-L automated blood sampling (Bioanalytical Systems, Inc) system was used to collect blood samples at multiple time points. A series of four use cases utilizing four different test compounds and analytical endpoints are described to illustrate some of the potential applications of the technique. RESULTS: In the four presented use cases, automated blood sampling in telemetry instrumented dogs provides simultaneous cardiovascular (heart rate, arterial blood pressure, and left ventricular pressure), electrophysiological assessment (QTc, PR, and QRS intervals), body temperature, and animal activity, while collecting multiple blood samples for drug analysis. CONCLUSION: The combination of automated blood sampling with cardiovascular telemetry monitoring is a novel capability designed to support safety pharmacology cardiovascular assessment of discovery molecules. By combining telemetry and high-fidelity ABS, the model provides an enhanced PK/PD understanding of drug-induced hemodynamic and electrocardiographic effects of discovery compounds in conscious beagles in the same experimental session. Importantly, the model can reduce the need for a separate pharmacokinetic study (positive reduction 3R impact), reduces compound syntheses requirements, and shorten development timelines. Furthermore, implementation of this approach has also improved animal welfare by reducing the animal handling during a study, thereby reducing stress and associated data artifacts (positive refinement 3R impact).


Subject(s)
Cardiovascular System , Telemetry , Animals , Blood Pressure , Dogs , Electrocardiography , Heart Rate , Male
4.
J Am Coll Cardiol ; 77(15): 1922-1933, 2021 04 20.
Article in English | MEDLINE | ID: mdl-33858628

ABSTRACT

The momentum of cardiovascular drug development has slowed dramatically. Use of validated cardiac biomarkers in clinical trials could accelerate development of much-needed therapies, but biomarkers have been used less for cardiovascular drug development than in therapeutic areas such as oncology. Moreover, there are inconsistences in biomarker use in clinical trials, such as sample type, collection times, analytical methods, and storage for future research. With these needs in mind, participants in a Cardiac Safety Research Consortium Think Tank proposed the development of international guidance in this area, together with improved quality assurance and analytical methods, to determine what biomarkers can reliably show. Participants recommended the development of systematic methods for sample collection, and the archiving of samples in all cardiovascular clinical trials (including creation of a biobank or repository). The academic and regulatory communities also agreed to work together to ensure that published information is fully and clearly expressed.


Subject(s)
Biomarkers/analysis , Cardiovascular Diseases/diagnosis , Clinical Trials as Topic/standards , Cardiovascular Diseases/drug therapy , Drug Discovery , Humans , Precision Medicine , Prognosis , Treatment Outcome
5.
J Pharmacol Toxicol Methods ; 109: 107066, 2021.
Article in English | MEDLINE | ID: mdl-33838254

ABSTRACT

INTRODUCTION: A successful integration of automated blood sampling (ABS) into the telemetry instrumented canine cardiovascular model is presented in this study. This combined model provides an efficient means to quickly gain understanding of potential effects on key cardiovascular parameters in dog while providing a complete Pharmacokinetic/Pharmacodynamic (PK/PD) profile for discovery compounds without handling artifacts, reducing the need for a separate pharmacokinetic study. METHODS: Male beagle dogs were chronically implanted with telemetry devices (PhysioTel™ model D70-PCTP) and vascular access ports (SPMID-GRIDAC-5NC). BASi Culex-L automated blood sampling (Bioanalytical Systems, Inc) system was used to collect blood samples at multiple time points. A series of four use cases utilizing four different test compounds and analytical endpoints are described to illustrate some of the potential applications of the technique. RESULTS: In the four presented use cases, automated blood sampling in telemetry instrumented dogs provides simultaneous cardiovascular (heart rate, arterial blood pressure, and left ventricular pressure), electrophysiological assessment (QTc, PR, and QRS intervals), body temperature, and animal activity, while collecting multiple blood samples for drug analysis. CONCLUSION: The combination of automated blood sampling with cardiovascular telemetry monitoring is a novel capability designed to support safety pharmacology cardiovascular assessment of discovery molecules. By combining telemetry and high-fidelity ABS, the model provides an enhanced PK/PD understanding of drug-induced hemodynamic and electrocardiographic effects of discovery compounds in conscious beagles in the same experimental session. Importantly, the model can reduce the need for a separate pharmacokinetic study (positive reduction 3R impact), reduces compound syntheses requirements, and shorten development timelines. Furthermore, implementation of this approach has also improved animal welfare by reducing the animal handling during a study, thereby reducing stress and associated data artifacts (positive refinement 3R impact).


Subject(s)
Cardiovascular System , Telemetry , Animals , Blood Pressure , Dogs , Electrocardiography , Heart Rate , Macaca fascicularis , Male
6.
Clin Pharmacol Ther ; 109(2): 310-318, 2021 02.
Article in English | MEDLINE | ID: mdl-32866317

ABSTRACT

Defining an appropriate and efficient assessment of drug-induced corrected QT interval (QTc) prolongation (a surrogate marker of torsades de pointes arrhythmia) remains a concern of drug developers and regulators worldwide. In use for over 15 years, the nonclinical International Council for Harmonization of Technical Requirements for Pharmaceuticals for Human Use (ICH) S7B and clinical ICH E14 guidances describe three core assays (S7B: in vitro hERG current & in vivo QTc studies; E14: thorough QT study) that are used to assess the potential of drugs to cause delayed ventricular repolarization. Incorporating these assays during nonclinical or human testing of novel compounds has led to a low prevalence of QTc-prolonging drugs in clinical trials and no new drugs having been removed from the marketplace due to unexpected QTc prolongation. Despite this success, nonclinical evaluations of delayed repolarization still minimally influence ICH E14-based strategies for assessing clinical QTc prolongation and defining proarrhythmic risk. In particular, the value of ICH S7B-based "double-negative" nonclinical findings (low risk for hERG block and in vivo QTc prolongation at relevant clinical exposures) is underappreciated. These nonclinical data have additional value in assessing the risk of clinical QTc prolongation when clinical evaluations are limited by heart rate changes, low drug exposures, or high-dose safety considerations. The time has come to meaningfully merge nonclinical and clinical data to enable a more comprehensive, but flexible, clinical risk assessment strategy for QTc monitoring discussed in updated ICH E14 Questions and Answers. Implementing a fully integrated nonclinical/clinical risk assessment for compounds with double-negative nonclinical findings in the context of a low prevalence of clinical QTc prolongation would relieve the burden of unnecessary clinical QTc studies and streamline drug development.


Subject(s)
Drugs, Investigational/adverse effects , Long QT Syndrome/chemically induced , Animals , Arrhythmias, Cardiac/chemically induced , Drug Development/methods , Drug Industry/methods , Electrocardiography/methods , Humans , Risk Assessment , Torsades de Pointes/chemically induced
7.
J Pharmacol Toxicol Methods ; 103: 106871, 2020.
Article in English | MEDLINE | ID: mdl-32360993

ABSTRACT

INTRODUCTION: The Comprehensive In Vitro Proarrhythmia Assay (CiPA) initiative differentiates torsadogenic risk of 28 drugs affecting ventricular repolarization based on multiple in vitro human derived ionic currents. However, a standardized prospective assessment of the electrophysiologic effects of these drugs in an integrated in vivo preclinical cardiovascular model is lacking. This study questioned whether QTc interval prolongation in a preclinical in vivo model could detect clinically reported QTc prolongation and assign torsadogenic risk for ten CiPA drugs. METHODS: An acute intravenous administered ascending dose anesthetized dog cardiovascular model was used to assess QTc prolongation along with other electrocardiographic (PR, QRS intervals) and hemodynamic (heart rate, blood pressures, left ventricular contractility) parameters at plasma concentrations spanning and exceeding clinical exposures. hERG current block potency was characterized using IC50 values from automated patch clamp. RESULTS: All eight drugs eliciting clinical QTc prolongation also delayed repolarization in anesthetized dogs at plasma concentrations within four-fold clinical exposures. In vitro QTc safety margins (defined based on clinical Cmax values/plasma concentrations eliciting statistically significant QTc prolongation in dogs) were lower for high vs intermediate torsadogenic risk drugs. In comparison, hERG IC10 values represented as total drug concentrations were better predictors of preclinical QTc prolongation than hERG IC50 values. CONCLUSION: There was good concordance for QTc prolongation in the anesthetized dog model and clinical torsadogenic risk assignment. QTc assessment in the anesthetized dog remains a valuable part of a more comprehensive preclinical integrated risk assessment for delayed repolarization and torsadogenic risk as part of a global cardiovascular evaluation.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Long QT Syndrome/drug therapy , Torsades de Pointes/drug therapy , Animals , Dogs , Drug Evaluation, Preclinical , Electrocardiography , HEK293 Cells , Heart Rate/drug effects , Humans , Long QT Syndrome/chemically induced , Male , Models, Cardiovascular , Prospective Studies , Risk Assessment , Torsades de Pointes/chemically induced
8.
J Pharmacol Toxicol Methods ; 99: 106612, 2019.
Article in English | MEDLINE | ID: mdl-31319140

ABSTRACT

INTRODUCTION: Voltage-sensitive optical (VSO) sensors offer a minimally invasive method to study the time course of repolarization of the cardiac action potential (AP). This Comprehensive in vitro Proarrhythmia Assay (CiPA) cross-platform study investigates protocol design and measurement variability of VSO sensors for preclinical cardiac electrophysiology assays. METHODS: Three commercial and one academic laboratory completed a limited study of the effects of 8 blinded compounds on the electrophysiology of 2 commercial lines of human induced pluripotent stem-cell derived cardiomyocytes (hSC-CMs). Acquisition technologies included CMOS camera and photometry; fluorescent voltage sensors included di-4-ANEPPS, FluoVolt and genetically encoded QuasAr2. The experimental protocol was standardized with respect to cell lines, plating and maintenance media, blinded compounds, and action potential parameters measured. Serum-free media was used to study the action of drugs, but the exact composition and the protocols for cell preparation and drug additions varied among sites. RESULTS: Baseline AP waveforms differed across platforms and between cell types. Despite these differences, the relative responses to four selective ion channel blockers (E-4031, nifedipine, mexiletine, and JNJ 303 blocking IKr, ICaL, INa, and IKs, respectively) were similar across all platforms and cell lines although the absolute changes differed. Similarly, four mixed ion channel blockers (flecainide, moxifloxacin, quinidine, and ranolazine) had comparable effects in all platforms. Differences in repolarisation time course and response to drugs could be attributed to cell type and experimental method differences such as composition of the assay media, stimulated versus spontaneous activity, and single versus cumulative compound addition. DISCUSSION: In conclusion, VSOs represent a powerful and appropriate method to assess the electrophysiological effects of drugs on iPSC-CMs for the evaluation of proarrhythmic risk. Protocol considerations and recommendations are provided toward standardizing conditions to reduce variability of baseline AP waveform characteristics and drug responses.

9.
Expert Opin Drug Discov ; 13(8): 753-763, 2018 08.
Article in English | MEDLINE | ID: mdl-29871539

ABSTRACT

INTRODUCTION: Despite a burgeoning knowledge of the intricacies and mechanisms responsible for human disease, technological advances in medicinal chemistry, and more efficient assays used for drug screening, it remains difficult to discover novel and effective pharmacologic therapies. Areas covered: By reference to the primary literature and concepts emerging from academic and industrial drug screening landscapes, the authors propose that this disconnect arises from the inability to scale and integrate responses from simpler model systems to outcomes from more complex and human-based biological systems. Expert opinion: Further collaborative efforts combining target-based and phenotypic-based screening along with systems-based pharmacology and informatics will be necessary to harness the technological breakthroughs of today to derive the novel drug candidates of tomorrow. New questions must be asked of enabling technologies-while recognizing inherent limitations-in a way that moves drug development forward. Attempts to integrate mechanistic and observational information acquired across multiple scales frequently expose the gap between our knowledge and our understanding as the level of complexity increases. We hope that the thoughts and actionable items highlighted will help to inform the directed evolution of the drug discovery process.


Subject(s)
Drug Development/methods , Drug Discovery/methods , Technology, Pharmaceutical/methods , Animals , Chemistry, Pharmaceutical/methods , Drug Design , Humans , Models, Biological , Phenotype
10.
Br J Pharmacol ; 175(4): 606-617, 2018 02.
Article in English | MEDLINE | ID: mdl-29181850

ABSTRACT

BACKGROUND AND PURPOSE: Translation of non-clinical markers of delayed ventricular repolarization to clinical prolongation of the QT interval corrected for heart rate (QTc) (a biomarker for torsades de pointes proarrhythmia) remains an issue in drug discovery and regulatory evaluations. We retrospectively analysed 150 drug applications in a US Food and Drug Administration database to determine the utility of established non-clinical in vitro IKr current human ether-à-go-go-related gene (hERG), action potential duration (APD) and in vivo (QTc) repolarization assays to detect and predict clinical QTc prolongation. EXPERIMENTAL APPROACH: The predictive performance of three non-clinical assays was compared with clinical thorough QT study outcomes based on free clinical plasma drug concentrations using sensitivity and specificity, receiver operating characteristic (ROC) curves, positive (PPVs) and negative predictive values (NPVs) and likelihood ratios (LRs). KEY RESULTS: Non-clinical assays demonstrated robust specificity (high true negative rate) but poor sensitivity (low true positive rate) for clinical QTc prolongation at low-intermediate (1×-30×) clinical exposure multiples. The QTc assay provided the most robust PPVs and NPVs (ability to predict clinical QTc prolongation). ROC curves (overall test accuracy) and LRs (ability to influence post-test probabilities) demonstrated overall marginal performance for hERG and QTc assays (best at 30× exposures), while the APD assay demonstrated minimal value. CONCLUSIONS AND IMPLICATIONS: The predictive value of hERG, APD and QTc assays varies, with drug concentrations strongly affecting translational performance. While useful in guiding preclinical candidates without clinical QT prolongation, hERG and QTc repolarization assays provide greater value compared with the APD assay.


Subject(s)
Cardiovascular Agents/pharmacology , Drugs, Investigational/pharmacology , Ether-A-Go-Go Potassium Channels/physiology , Heart Rate/drug effects , Action Potentials/drug effects , Action Potentials/physiology , Cardiovascular Agents/therapeutic use , Drug Evaluation, Preclinical/methods , Drugs, Investigational/therapeutic use , Ether-A-Go-Go Potassium Channels/agonists , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Heart Rate/physiology , Humans , Long QT Syndrome/drug therapy , Long QT Syndrome/physiopathology , Retrospective Studies , Torsades de Pointes/drug therapy , Torsades de Pointes/physiopathology
11.
Article in English | MEDLINE | ID: mdl-27432021

ABSTRACT

Screening novel, poorly soluble small-molecule candidates for cardiovascular liabilities represents a key challenge in early drug discovery. This report describes a novel vehicle composed of 20% N,N-Dimethylacetamide (DMA)/40% Propylene glycol (PG)/40% Polyethylene Glycol (PEG-400) (DPP) for administration of new chemical entities (NCEs) by slow intravenous (i.v.) infusion in a preclinical anesthetized rat model. The vehicle was designed considering both available excipient safety information and solubilization potential for poorly soluble NCEs. DPP solubilized 11 drugs, 8 of which were insoluble in 5% dextrose in water (D5W), and 5 insoluble in PEG-400 to a target concentration of 30mg/mL. DPP elicits no adverse cardiovascular responses in the anesthetized rat model despite containing 40% PEG-400, a commonly used organic solvent which elicits hypertension and bradycardia that often confounds interpretation of drug effects. Three compounds demonstrating adequate solubility in both DPP and D5W were screened in the anesthetized rat model. When normalized to plasma exposure, atenolol, sotalol and enalaprilat exhibited comparable mean arterial pressure, heart rate, and cardiac contractility responses regardless of formulation. While the antihypertensive effect of nifedipine was evident with both DPP and PEG-400 formulations, pressor effects from PEG-400 confounded interpretation of the magnitude of nifedipine's response. Plasma concentrations of atenolol and enalaprilat were greater in D5W formulation whereas sotalol exposures were greater when using DPP as a vehicle. These results demonstrate the utility of DPP as an intravenous vehicle for formulating poorly soluble compounds in early preclinical screening for cardiovascular safety studies.


Subject(s)
Drug Carriers/chemistry , Excipients/chemistry , Hemodynamics/drug effects , Models, Cardiovascular , Pharmaceutical Preparations/administration & dosage , Small Molecule Libraries/administration & dosage , Acetamides/administration & dosage , Acetamides/chemistry , Acetamides/toxicity , Animals , Drug Carriers/administration & dosage , Drug Carriers/toxicity , Drug Discovery/methods , Drug Evaluation, Preclinical , Excipients/administration & dosage , Excipients/toxicity , Infusions, Intravenous , Lethal Dose 50 , Male , Pharmaceutical Preparations/blood , Pharmaceutical Preparations/chemistry , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/chemistry , Polyethylene Glycols/toxicity , Propylene Glycol/administration & dosage , Propylene Glycol/chemistry , Propylene Glycol/toxicity , Rats, Sprague-Dawley , Small Molecule Libraries/adverse effects , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacokinetics , Solubility
12.
J Pharmacol Toxicol Methods ; 81: 183-95, 2016.
Article in English | MEDLINE | ID: mdl-27235787

ABSTRACT

While current S7B/E14 guidelines have succeeded in protecting patients from QT-prolonging drugs, the absence of a predictive paradigm identifying pro-arrhythmic risks has limited the development of valuable drug programs. We investigated if a human ex-vivo action potential (AP)-based model could provide a more predictive approach for assessing pro-arrhythmic risk in man. Human ventricular trabeculae from ethically consented organ donors were used to evaluate the effects of dofetilide, d,l-sotalol, quinidine, paracetamol and verapamil on AP duration (APD) and recognized pro-arrhythmia predictors (short-term variability of APD at 90% repolarization (STV(APD90)), triangulation (ADP90-APD30) and incidence of early afterdepolarizations at 1 and 2Hz to quantitatively identify the pro-arrhythmic risk. Each drug was blinded and tested separately with 3 concentrations in triplicate trabeculae from 5 hearts, with one vehicle time control per heart. Electrophysiological stability of the model was not affected by sequential applications of vehicle (0.1% dimethyl sulfoxide). Paracetamol and verapamil did not significantly alter anyone of the AP parameters and were classified as devoid of pro-arrhythmic risk. Dofetilide, d,l-sotalol and quinidine exhibited an increase in the manifestation of pro-arrhythmia markers. The model provided quantitative and actionable activity flags and the relatively low total variability in tissue response allowed for the identification of pro-arrhythmic signals. Power analysis indicated that a total of 6 trabeculae derived from 2 hearts are sufficient to identify drug-induced pro-arrhythmia. Thus, the human ex-vivo AP-based model provides an integrative translational assay assisting in shaping clinical development plans that could be used in conjunction with the new CiPA-proposed approach.


Subject(s)
Action Potentials/drug effects , Arrhythmias, Cardiac/chemically induced , Arrhythmias, Cardiac/physiopathology , Adult , Cardiovascular Agents/pharmacology , Data Interpretation, Statistical , Drug Discovery , Electrocardiography/drug effects , Electrophysiological Phenomena/drug effects , Female , Heart/drug effects , Humans , In Vitro Techniques , Male , Middle Aged , Models, Biological , Predictive Value of Tests , Risk Assessment , Signal-To-Noise Ratio , Young Adult
13.
Am Heart J ; 165(4): 489-500, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23537964

ABSTRACT

Recent advances in electrocardiographic monitoring and waveform analysis have significantly improved the ability to detect drug-induced changes in cardiac repolarization manifested as changes in the QT/corrected QT interval. These advances have also improved the ability to detect drug-induced changes in cardiac conduction. This White Paper summarizes current opinion, reached by consensus among experts at the Cardiac Safety Research Consortium, on the assessment of electrocardiogram-based safety measurements of the PR and QRS intervals, representing atrioventricular and ventricular conduction, respectively, during drug development.


Subject(s)
Cardiovascular Diseases/physiopathology , Heart Conduction System/drug effects , Anti-Arrhythmia Agents/pharmacology , Clinical Trials as Topic , Drug Discovery , Drug Evaluation, Preclinical , Electrocardiography , Humans , Risk Assessment
14.
Life Sci ; 90(15-16): 607-11, 2012 Apr 20.
Article in English | MEDLINE | ID: mdl-22406079

ABSTRACT

AIMS: In a previous study we found that A-935142 enhanced hERG current in a concentration-dependent manner by facilitating activation, reducing inactivation, and slowing deactivation (Su et al., 2009). A-935142 also shortened action potential duration (APD90) in canine Purkinje fibers and guinea pig atrial tissue. This study focused on the combined effects of the prototypical hERG enhancer, A-935142 and two hERG current blockers (sotalol and terfenadine). MAIN METHODS: The whole-cell voltage clamp method with HEK 293 cells heterologously expressing the hERG channel (Kv 11.1) was used. KEY FINDINGS: A-935142 did not compete with 3H-dofetilide binding, suggesting that A-935142 does not overlap the binding site of typical hERG blockers. In whole-cell voltage clamp studies we found: 1) 60 µM A-935142 enhanced hERG current in the presence of 150 µM sotalol (57.5±5.8%) to a similar extent as seen with A-935142 alone (55.6±5.1%); 2) 150 µM sotalol blocked hERG current in the presence of 60 µM A-935142 (43.5±1.5%) to a similar extent as that seen with sotalol alone (42.0±3.2%) and 3) during co-application, hERG current enhancement was followed by current blockade. Similar results were obtained with 60 nM terfenadine combined with A-935142. SIGNIFICANCE: These results suggest that the hERG enhancer, A-935142 does not compete with these two known hERG blockers at their binding site within the hERG channel. This selective hERG current enhancement may be useful as a treatment for inherited or acquired LQTS (Casis et al., 2006).


Subject(s)
Acetates/metabolism , Ether-A-Go-Go Potassium Channels/metabolism , Pyrazoles/metabolism , Analysis of Variance , HEK293 Cells , Humans , Patch-Clamp Techniques , Phenethylamines/metabolism , Sotalol/pharmacology , Sulfonamides/metabolism , Terfenadine/pharmacology , Tritium
15.
Br J Pharmacol ; 164(2): 254-9, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21488862

ABSTRACT

UNLABELLED: Cardiac safety remains of paramount importance in the development of successful clinical drug candidates. Great progress has been made recently in understanding liabilities associated with delayed ventricular repolarization (manifest as QT prolongation) and in predicting (thus avoiding) drugs that delay repolarization based on application of strategic preclinical assays. Following the advances made in clinical electrophysiological monitoring and conduct of thorough QT studies, focus is now shifting towards monitoring of additional drug-induced effects, particularly on ventricular conduction (measured as changes in the QRS interval on the ECG) as part of evolving clinical thorough ECG studies. In this issue of the British Journal of Pharmacology, a study by Harmer et al. proposes provisional safety margins for QRS prolongation in man based on retrospective clinical data and a single in vitro approach to assess potency of block of cardiac sodium current (hNav1.5), the ionic current responsible for ventricular conduction (observed as QRS prolongation). The present commentary places their study in context with evolving preclinical cardiac electrophysiological safety assessments, along with discussions focused on ensuring the proper 'translation' of preclinical findings with potential clinical concerns. Given the extant limitations and uncertainties of presently available data, as well as our limited understanding of the pro-arrhythmic potential associated with these changes, due caution should be applied when considering the proposed in vitro-based margins for drug-induced QRS prolongation measured clinically. Additional validation with multiple preclinical models and more rigorous clinical safety studies will be necessary to substantiate these recommended margins. LINKED ARTICLE: This article is a commentary on Harmer et al., pp. 260-273 of this issue. To view this paper visit http://dx.doi.org/10.1111/j.1476-5381.2011.01415.x.


Subject(s)
Anti-Arrhythmia Agents/adverse effects , Arrhythmias, Cardiac/drug therapy , Sodium Channel Blockers/adverse effects , Animals , Anti-Arrhythmia Agents/therapeutic use , Male , Sodium Channel Blockers/therapeutic use
16.
J Pharmacol Toxicol Methods ; 64(1): 68-73, 2011.
Article in English | MEDLINE | ID: mdl-21440075

ABSTRACT

INTRODUCTION: Delayed cardiac repolarization is an established risk factor for proarrhythmia and Torsades-de-Pointes (TdeP) that is typically measured in vitro during slow, regular stimulation. We have developed an alternative, novel, and rapid cellular-based approach for predicting drug-induced proarrhythmia that detects changes in electrical refractoriness based on mechanical responses (measured optically) during increasingly rapid trains of stimulation interspersed with pauses (mimicking the clinically observed short-long-short (SLS) stimulation sequence associated with the TdeP initiation). METHODS: Acutely isolated rabbit ventricular myocytes were superfused and electrically stimulated using an accelerating pacing protocol (APP) consisting of 12 consecutive pacing segments (10 beats per segment) with incrementally faster cycle lengths; trains were separated by pauses to identify loss of stimulus capture as well as to mimic clinically observed SLS sequences. Drug effects were evaluated based on a myocyte's ability to contract during progressively faster pacing segments (rate-adaptation); the earliest rate during which the myocyte fails to respond (longest cycle length with incomplete capture (CLIC)) was used to quantify electrophysiologic effects. RESULTS: Torsadogenic drugs known to delay repolarization during slow stimulation prolonged CLIC and dramatically limited the ability to respond to progressively rapid stimulation. The recognized proarrhythmic compounds E-4031, cisapride, grepafloxacin, and haloperidol rapidly prolonged CLIC at and above therapeutic concentrations in a concentration-dependent manner, while negative controls (captopril, indomethacin, and loratidine) do not affect rate-adaptation. DISCUSSION: Ventricular rate adaptation represents a novel approach for rapidly detecting drugs with torsadogenic risk using rapid rhythms that are typically not employed when evaluating proarrhythmic risk. This method is well suited for detecting and avoiding potential cardiac liabilities early in drug discovery ("frontloading") prior to final selection of candidate drugs.


Subject(s)
Electrocardiography/drug effects , Heart/drug effects , Myocytes, Cardiac/drug effects , Torsades de Pointes/chemically induced , Torsades de Pointes/diagnosis , Animals , Drug Evaluation, Preclinical/methods , Drug-Related Side Effects and Adverse Reactions , Electric Stimulation/methods , Female , Heart/physiopathology , Heart Ventricles/drug effects , Heart Ventricles/physiopathology , Rabbits , Torsades de Pointes/physiopathology
17.
Curr Drug Saf ; 6(5): 277-84, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-22424534

ABSTRACT

A-955840, a selective CB2 agonist, has been shown to elicit concentration-dependent decreases in cardiac contractility in the anesthetized dog (decreased maximal velocity of left ventricular pressure development [LV dP/dt max]). However, it is unknown whether this represents a direct effect or a response dependent on other factors (such as autonomic tone and neurohumoral factors) present in vivo. This study examined if A-955840 had a direct effect on contractility of isolated cardiac myocytes, and if so to determine the potential mechanisms. Contractility was assessed in vitro using percent changes in maximal shortening velocity of sarcomeres (dL/dt max) and fractional shortening of sarcomere length (FS) in rabbit left ventricular myocytes. L-type calcium current in myocytes was recorded using wholecell voltage-clamp techniques. A-955840 reduced dL/dt max and FS in a reversible and concentration-dependent manner with an IC50 of 11.4 µg/mL (based on dL/dt max) which is similar to the estimated IC50 value of 9.8 µg/mL based on the effects of A-955840 on LV dP/dt max in anesthetized dogs. A-955840 (4.1 µg/mL) reduced myocyte contractility (%FS) to a similar extent in the absence and presence of a CB2 antagonist, SR-2 (24.0 ± 3.4 vs 23.1 ± 3.0 %, n=5) or a CB1 antagonist, Rimonabant (18.8 ± 2.3 vs 19.8 ± 2.7 %, n=5). A-955840 (4.1 µg/mL) also reduced L-type calcium current of rabbit ventricular myocytes (1.05 ± 0.11 vs 0.70 ± 0.12 nA, n=5, P < 0.01). These results suggest that A-955840 exerts direct negative inotropic effects on isolated rabbit ventricular myocytes, which is mediated by neither CB2 nor CB1 receptors, and consistent with off-target negative inotropy mediated by inhibition of the cardiac L-type calcium current.


Subject(s)
Benzamides/pharmacology , Myocardial Contraction/drug effects , Myocytes, Cardiac/drug effects , Receptor, Cannabinoid, CB2/agonists , Thiazoles/pharmacology , Animals , Benzamides/administration & dosage , Calcium Channels, L-Type/drug effects , Calcium Channels, L-Type/metabolism , Dogs , Heart Ventricles/cytology , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Inhibitory Concentration 50 , Male , Myocytes, Cardiac/metabolism , Patch-Clamp Techniques , Piperidines/pharmacology , Pyrazoles/pharmacology , Rabbits , Receptor, Cannabinoid, CB1/metabolism , Rimonabant , Sarcomeres/metabolism , Thiazoles/administration & dosage
18.
Biochem Pharmacol ; 80(7): 1000-6, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20599796

ABSTRACT

N'1-(3,3,6,8-tetramethyl-1-oxo-1,2,3,4-tetrahydronaphthalen-2-yliden)-2-cyanoethanohydrazide (TTYC) increases secretion of glucagon-like peptide-1 and intracellular Ca(2+) concentration in GLUTag cells. The purpose of the present study was to examine if TTYC exerts positive inotropic effects on isolated rabbit ventricular myocytes and in vivo heart in anesthetized rats, and if so to further define the potential mechanism of action. Contractility was assessed in vitro using changes in fractional shortening (FS) of myocyte sarcomere length and in vivo using changes in the velocity of left ventricular pressure. Changes in L-type Ca(2+) current of ventricular myocytes were evaluated using whole-cell voltage-clamp techniques. TTYC increased FS of myocyte sarcomere length in a concentration-dependent manner. The positive inotropic effect was not abrogated by beta-adrenergic blockade (propranolol) or protein kinase A inhibition. TTYC enhanced peak L-type Ca(2+) current in a voltage-dependent manner (current amplitudes increased by 4.0-fold at -10 mV and 1.5-fold at +10 mV). Voltage-dependence of steady-state activation of L-type Ca(2+) current was shifted by 15 mV in the negative direction. Inactivation time course of the L-type Ca(2+) currents at voltages of -10 to 20 mV was significantly slowed by 0.3 microM TTYC. In vivo studies demonstrated that TTYC increased cardiac contractility in a dose-dependent manner. In conclusion, TTYC is a novel L-type Ca(2+) current activator with positive cardiac inotropic effects. Negative shifting of the voltage-dependence of L-type Ca(2+) current activation and reduced inactivation are two mechanisms responsible for the enhanced L-type Ca(2+) current that contribute to the positive inotropic effects.


Subject(s)
Calcium/metabolism , Myocardial Contraction/drug effects , Animals , Calcium/pharmacology , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide 1/pharmacology , Heart/drug effects , Male , Muscle Cells/metabolism , Myocardial Contraction/physiology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Patch-Clamp Techniques , Rabbits , Rats , Rats, Sprague-Dawley
19.
J Cardiovasc Pharmacol ; 54(6): 543-51, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19770671

ABSTRACT

Torcetrapib is a cholesteryl ester transfer protein inhibitor with an undesired response of increasing arterial pressure in humans. Pressor responses to torcetrapib have been demonstrated in multiple preclinical species. However, these studies have not related plasma concentrations to observed effects. Our purpose was to 1) characterize the cardiovascular responses of torcetrapib in conscious and anesthetized dogs with measured plasma concentrations; and 2) characterize the hemodynamic effects contributing to hypertension using comprehensively instrumented anesthetized dogs. Torcetrapib was dosed orally (3, 30 mg/kg) and intravenously (0.01, 0.33, 0.1 mg/kg) in conscious and anesthetized dogs, respectively. Mean arterial pressure and heart rate were monitored in both models; additional parameters were measured in anesthetized dogs. Plasma drug concentrations were assessed in both models. In conscious and anesthetized dogs, torcetrapib increased mean arterial pressure 25 and 18 mm Hg and heart rate 35 and 21 beats/min, at 2.94 and 3.99 microg/mL, respectively. In anesthetized dogs, torcetrapib increased pulmonary arterial pressure, both systemic and pulmonary hypertension driven by increases in vascular resistance. The compound increased rate pressure product and myocardial contractility while decreasing time to systolic pressure recovery and ejection time. Thus, torcetrapib-induced pressor responses are mediated by systemic and pulmonary vasoconstriction and are associated with increased myocardial oxygen consumption and positive inotropy.


Subject(s)
Anesthesia , Cardiovascular System/drug effects , Hemodynamics/drug effects , Pentobarbital/administration & dosage , Quinolines/pharmacology , Animals , Blood Pressure/drug effects , Blood Pressure/physiology , Cardiac Output/drug effects , Cardiac Output/physiology , Cholesterol Ester Transfer Proteins/antagonists & inhibitors , Dogs , Electrocardiography , Heart Rate/drug effects , Heart Rate/physiology , Hemodynamics/physiology , Male , Myocardial Contraction/drug effects , Myocardial Contraction/physiology , Quinolines/administration & dosage , Quinolines/blood , Quinolines/pharmacokinetics , Telemetry , Vascular Resistance/drug effects , Vascular Resistance/physiology , Ventricular Function, Left/drug effects , Ventricular Function, Left/physiology
20.
Biochem Pharmacol ; 77(8): 1383-90, 2009 Apr 15.
Article in English | MEDLINE | ID: mdl-19426677

ABSTRACT

Activators of the human ether-a-go-go-related gene (hERG) K(+) channel have been reported recently to enhance hERG current amplitude (five synthetic small molecules and one naturally occurring substance). Here, we characterize the effects of a novel compound A-935142 ({4-[4-(5-trifluoromethyl-1H-pyrazol-3-yl)-phenyl]-cyclohexyl}-acetic acid) on guinea-pig atrial and canine ventricular action potentials (microelectrode techniques) and hERG channels expressed in HEK-293 cells (whole-cell patch clamp techniques). A-935142 shortened cardiac action potentials and enhanced the amplitude of the hERG current in a concentration- and voltage-dependent manner. The fully activated current-voltage relationship revealed that this compound (60 microM) increased both outward and inward K(+) current as well as the slope conductance of the linear portion of the fully activated I-V relation. A-935142 significantly reduced the time constants (tau) of hERG channel activation at two example voltages (-10 mV: tau=100+/-17 ms vs. 164+/-24 ms, n=6, P<0.01; +30 mV: tau=16.7+/-1.8 ms vs. 18.9+/-1.8 ms, n=5, P<0.05) and shifted the voltage-dependence for hERG activation in the hyperpolarizing direction by 9 mV. The time course of hERG channel deactivation was slowed at multiple potentials (-120 to -70 mV). A-935142 also reduced the rate of inactivation and shifted the voltage-dependence of inactivation in the depolarizing direction by 15 mV. Recovery of hERG channel from inactivation was not affected by A-935142. In conclusion, A-935142 enhances hERG current in a complex manner by facilitation of activation, reduction of inactivation, and slowing of deactivation, and abbreviates atrial and ventricular repolarization.


Subject(s)
Acetates/pharmacology , Action Potentials/drug effects , Ether-A-Go-Go Potassium Channels/metabolism , Ion Channel Gating/drug effects , Pyrazoles/pharmacology , Acetates/chemistry , Animals , Cell Culture Techniques , Cell Line , Dogs , Dose-Response Relationship, Drug , Guinea Pigs , Heart Atria/drug effects , Heart Atria/metabolism , Humans , Molecular Structure , Patch-Clamp Techniques , Purkinje Fibers/drug effects , Purkinje Fibers/metabolism , Pyrazoles/chemistry , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...