Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
J Med Chem ; 67(2): 1225-1242, 2024 Jan 25.
Article in English | MEDLINE | ID: mdl-38228402

ABSTRACT

Interleukin-1 receptor-associated kinase 4 (IRAK4) plays a critical role in innate inflammatory processes. Here, we describe the discovery of two clinical candidate IRAK4 inhibitors, BAY1834845 (zabedosertib) and BAY1830839, starting from a high-throughput screening hit derived from Bayer's compound library. By exploiting binding site features distinct to IRAK4 using an in-house docking model, liabilities of the original hit could surprisingly be overcome to confer both candidates with a unique combination of good potency and selectivity. Favorable DMPK profiles and activity in animal inflammation models led to the selection of these two compounds for clinical development in patients.


Subject(s)
High-Throughput Screening Assays , Indazoles , Interleukin-1 Receptor-Associated Kinases , Pyridines , Animals , Humans , Binding Sites , Inflammation
2.
Clin Pharmacol Drug Dev ; 13(3): 265-280, 2024 03.
Article in English | MEDLINE | ID: mdl-38224232

ABSTRACT

Inhibition of activated factor XI reduces thrombogenesis while maintaining physiological hemostasis, with the expectation of reduced bleeding risk compared with standard of care in the clinical setting. Asundexian (BAY 2433334), an activated factor XI inhibitor, is in clinical development for the prevention of thromboembolic events. The effect of asundexian and its plasma metabolite M10 on cardiac repolarization and potential interactions with the hNav1.5 sodium, hCav1.2 calcium, and human ether-à-go-go-related gene (hERG) potassium channels was investigated in vitro. Additionally, asundexian effects on cardiac parameters and electrocardiogram were examined in telemetered beagle dogs. A randomized, placebo-controlled, 4-way crossover, thorough QT study in healthy adults evaluated the influence of 50 and 150 mg of asundexian on the corrected QT interval, including 400 mg of moxifloxacin as positive control. Across all studies, asundexian and M10 were not associated with any effects on cardiac repolarization. The largest in vitro effects of asundexian (approximately 20% inhibition) were seen for hCav1.2 and hERG. Throughout the thorough QT study, the upper limits of the one-sided 95% confidence interval of placebo-corrected mean changes from baseline in Fridericia corrected QT for 50 and 150 mg of asundexian were below Δ = 10 milliseconds. Asundexian demonstrated favorable safety and tolerability profiles.


Subject(s)
Factor XIa , Fluoroquinolones , Adult , Humans , Animals , Dogs , Fluoroquinolones/pharmacology , Factor XIa/pharmacology , Heart , Moxifloxacin/pharmacology , Electrocardiography
3.
J Pharmacol Exp Ther ; 386(1): 26-34, 2023 07.
Article in English | MEDLINE | ID: mdl-37068911

ABSTRACT

Vericiguat and its metabolite M-1 were assessed for proarrhythmic risk in nonclinical in vitro and in vivo studies. In vitro manual voltage-clamp recordings at room temperature determined the effect of vericiguat on human Ether-a-go-go Related Gene (hERG) K+ channels. Effects of vericiguat and M-1 on hERG K+, Nav1.5, hCav1.2, hKvLQT1/1minK, and hKv4.3 channels were investigated via automated voltage-clamp recordings at ambient temperature. Effects of vericiguat and M-1 on hERG K+ and Nav1.5 channels at pathophysiological conditions were explored via manual voltage-clamp recordings at physiologic temperature. Single oral doses of vericiguat (0.6, 2.0, and 6.0 mg/kg) were assessed for in vivo proarrhythmic risk via administration to conscious telemetered dogs; electrocardiogram (ECG) and hemodynamic parameters were monitored. ECG recordings were included in 4- and 39-week dog toxicity studies. In manual voltage-clamp recordings, vericiguat inhibited hERG K+-mediated tail currents in a concentration-dependent manner (20% threshold inhibitory concentration ∼1.9 µM). In automated voltage-clamp recordings, neither vericiguat nor M-1 were associated with biologically relevant inhibition (>20%) of hNav1.5, hCav1.2, hKvLQT1, and hKv4.3. No clinically relevant observations were made for hNav1.5 and hKvLQT1 under simulated pathophysiological conditions. Vericiguat was associated with expected mode-of-action-related dose-dependent changes in systolic arterial blood pressure (up to -20%) and heart rate (up to +53%). At maximum vericiguat dose, corrected QT (QTc) interval changes from baseline varied slightly (-6 to +1%) depending on correction formula. Toxicity studies confirmed absence of significant QTc interval changes. There was no evidence of an increased proarrhythmic risk from nonclinical studies with vericiguat or M-1. SIGNIFICANCE STATEMENT: There was no evidence of an increased proarrhythmic risk from in vitro and in vivo nonclinical studies with vericiguat or M-1. The integrated risk assessment of these nonclinical data combined with existing clinical data demonstrate administration of vericiguat 10 mg once daily in patients with heart failure with reduced ejection fraction is not associated with a proarrhythmic risk.


Subject(s)
Heart Failure , Heterocyclic Compounds, 2-Ring , Humans , Animals , Dogs , Soluble Guanylyl Cyclase/metabolism , Pyrimidines , Vasodilator Agents , Ether-A-Go-Go Potassium Channels
4.
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.
Sci Rep ; 10(1): 5627, 2020 03 27.
Article in English | MEDLINE | ID: mdl-32221320

ABSTRACT

Automated patch clamp (APC) instruments enable efficient evaluation of electrophysiologic effects of drugs on human cardiac currents in heterologous expression systems. Differences in experimental protocols, instruments, and dissimilar site procedures affect the variability of IC50 values characterizing drug block potency. This impacts the utility of APC platforms for assessing a drug's cardiac safety margin. We determined variability of APC data from multiple sites that measured blocking potency of 12 blinded drugs (with different levels of proarrhythmic risk) against four human cardiac currents (hERG [IKr], hCav1.2 [L-Type ICa], peak hNav1.5, [Peak INa], late hNav1.5 [Late INa]) with recommended protocols (to minimize variance) using five APC platforms across 17 sites. IC50 variability (25/75 percentiles) differed for drugs and currents (e.g., 10.4-fold for dofetilide block of hERG current and 4-fold for mexiletine block of hNav1.5 current). Within-platform variance predominated for 4 of 12 hERG blocking drugs and 4 of 6 hNav1.5 blocking drugs. hERG and hNav1.5 block. Bland-Altman plots depicted varying agreement across APC platforms. A follow-up survey suggested multiple sources of experimental variability that could be further minimized by stricter adherence to standard protocols. Adoption of best practices would ensure less variable APC datasets and improved safety margins and proarrhythmic risk assessments.

8.
Toxicol Appl Pharmacol ; 394: 114961, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32209365

ABSTRACT

INTRODUCTION: hERG block potency is widely used to calculate a drug's safety margin against its torsadogenic potential. Previous studies are confounded by use of different patch clamp electrophysiology protocols and a lack of statistical quantification of experimental variability. Since the new cardiac safety paradigm being discussed by the International Council for Harmonisation promotes a tighter integration of nonclinical and clinical data for torsadogenic risk assessment, a more systematic approach to estimate the hERG block potency and safety margin is needed. METHODS: A cross-industry study was performed to collect hERG data on 28 drugs with known torsadogenic risk using a standardized experimental protocol. A Bayesian hierarchical modeling (BHM) approach was used to assess the hERG block potency of these drugs by quantifying both the inter-site and intra-site variability. A modeling and simulation study was also done to evaluate protocol-dependent changes in hERG potency estimates. RESULTS: A systematic approach to estimate hERG block potency is established. The impact of choosing a safety margin threshold on torsadogenic risk evaluation is explored based on the posterior distributions of hERG potency estimated by this method. The modeling and simulation results suggest any potency estimate is specific to the protocol used. DISCUSSION: This methodology can estimate hERG block potency specific to a given voltage protocol. The relationship between safety margin thresholds and torsadogenic risk predictivity suggests the threshold should be tailored to each specific context of use, and safety margin evaluation may need to be integrated with other information to form a more comprehensive risk assessment.


Subject(s)
ERG1 Potassium Channel/antagonists & inhibitors , Risk Assessment/methods , Torsades de Pointes/chemically induced , Bayes Theorem , Computer Simulation , Humans , Models, Biological , Patch-Clamp Techniques , Potassium Channel Blockers/pharmacology , Safety , Torsades de Pointes/physiopathology
9.
J Med Chem ; 62(2): 928-940, 2019 01 24.
Article in English | MEDLINE | ID: mdl-30563338

ABSTRACT

The availability of a chemical probe to study the role of a specific domain of a protein in a concentration- and time-dependent manner is of high value. Herein, we report the identification of a highly potent and selective ERK5 inhibitor BAY-885 by high-throughput screening and subsequent structure-based optimization. ERK5 is a key integrator of cellular signal transduction, and it has been shown to play a role in various cellular processes such as proliferation, differentiation, apoptosis, and cell survival. We could demonstrate that inhibition of ERK5 kinase and transcriptional activity with a small molecule did not translate into antiproliferative activity in different relevant cell models, which is in contrast to the results obtained by RNAi technology.


Subject(s)
Mitogen-Activated Protein Kinase 7/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Pyridines/chemistry , Pyrimidines/chemistry , Apoptosis/drug effects , Binding Sites , Cell Differentiation/drug effects , Cell Line , Cell Proliferation/drug effects , Crystallography, X-Ray , Drug Evaluation, Preclinical , Half-Life , Humans , Mitogen-Activated Protein Kinase 7/metabolism , Molecular Docking Simulation , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Structure, Tertiary , Pyridines/metabolism , Pyridines/pharmacology , Pyrimidines/metabolism , Pyrimidines/pharmacology , Signal Transduction/drug effects , Structure-Activity Relationship , Transcription, Genetic/drug effects
10.
J Pharmacol Toxicol Methods ; 81: 313-22, 2016.
Article in English | MEDLINE | ID: mdl-27179911

ABSTRACT

INTRODUCTION: Compound X is a new proprietary antihypertensive agent that induces its pharmacodynamic effect at an approximate plasma Cmax.u of 0.6nmol/L (rat hypertension model). However, Compound X also shows potent off-target activity at PDE-10a (IC50~12nmol/L). Since PDE-10a is expressed predominantly in brain (striatum) and inhibition/knockout of PDE-10a have been reported to result in anti-psychotic effects, we have established the "induced hyperactivity" test for CNS de-risking of Compound X. METHODS: Male Wistar rats treated orally with vehicle or Compound X (single dose; 1-3-10mg/kg) were assessed for exploratory locomotor activity following induction of hyperactivity by d-amphetamine (2mg/kg) or the NMDA antagonist MK-801 (0.2mg/kg). The assay was validated with anti-psychotic drugs (haloperidol, clozapine). RESULTS: Induced hyperactivity was not antagonized by Compound X at doses relevant for its primary pharmacodynamic activity (0.1-0.3mg/kg, rat). Although sufficient plasma concentrations were reached with Compound X (Cmax.u up to ~8nmol/L at 10mg/kg) to show its PDE-10a activity, its low brain penetration (~10%) likely precluded any meaningful PDE-10a inhibition. In comparison, other blood pressure lowering agents such as prazosin (alpha-1 adrenoceptor antagonist) and isradipine (L-Type Ca(2+) channel blocker), but not the NO-donor ISDN, tended to attenuate induced hyperactivity in rats at high doses. CONCLUSION: The relevance of a potent in-vitro off-target hit (PDE-10a inhibition) by Compound X was attenuated by a robust in-vivo assay (rat induced hyperactivity test), hence lowering the potential liability profile of Compound X. Finally, this piece of investigative safety pharmacology work enabled early de-risking of Compound X based on its primary pharmacodynamic activity in a relevant rat model.


Subject(s)
Antihypertensive Agents/toxicity , Hyperkinesis/chemically induced , Hyperkinesis/psychology , Animals , Antihypertensive Agents/pharmacokinetics , Antipsychotic Agents/toxicity , Blood Pressure/drug effects , Brain/metabolism , Central Nervous System Stimulants/toxicity , Dextroamphetamine/toxicity , Dizocilpine Maleate/toxicity , Drug Evaluation, Preclinical , Excitatory Amino Acid Antagonists/toxicity , Exploratory Behavior/drug effects , Male , Motor Activity/drug effects , Rats , Rats, Wistar
11.
J Pharmacol Toxicol Methods ; 68(1): 97-111, 2013.
Article in English | MEDLINE | ID: mdl-23702537

ABSTRACT

INTRODUCTION: Early prediction of drug-induced functional cardiotoxicity requires robust in-vitro systems suitable for medium/high throughput and easily accessible cardiomyocytes with defined reproducible properties. The xCELLigence Cardio system uses 96-well plates with interdigitated electrodes that detect the impedance changes of rhythmic contractions of stem cell-derived cardiomyocyte (SC-CM) layers. Here, we report on our initial screening experience in comparison to established (multi)cellular and in-vivo models. METHODS: Impedance signals from human iPSC-CM (iCells™) and mouse eSC-CM (Cor.At™) were analyzed for contraction amplitude (CA) and duration, rise/fall time, beating rate (BR) and irregularity. RESULTS: Following solution exchange, impedance signals re-approximated steady-state conditions after about 2 (Cor.At™) and 3h (iCells™); these time points were used to analyze drug effects. The solvent DMSO (≤1%) hardly influenced contraction parameters in Cor.At™, whereas in iCells™ DMSO (>0.1%) reduced CA and enhanced BR. The selective hERG K⁺ channel blockers E-4031 and dofetilide reduced CA and accelerated BR (≥30 nM) according to the analysis software. The latter, however, was due to burst-like contractions (300 nM) that could be detected only by visual inspection of recordings, and were more pronounced in Cor.At™ as in iCells™. In cardiac myocytes and tissue preparations, however, E4031 and dofetilide have been reported to increase cell shortening and contractile force and to reduce BR. Compounds (pentamidine, HMR1556, ATX2, TTX, and verapamil) with other mechanisms of action were also investigated; their effects differed partially between cell lines (e.g. TTX) and compared to established (multi)cellular models (e.g. HMR1556, ouabain). CONCLUSION: Mouse and human stem cell-derived cardiomyocytes respond differently to drugs and these responses occasionally also differ from those originating from established in-vitro and in-vivo models. Hence, drug-induced cardiotoxic effects may be detected with this system, however, the predictive or even translational value of results is considered limited and not yet firmly established.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Myocardial Contraction/drug effects , Myocytes, Cardiac/drug effects , Toxicity Tests/methods , Animals , Dimethyl Sulfoxide/chemistry , Electric Impedance , High-Throughput Screening Assays/methods , Humans , Mice , Myocytes, Cardiac/metabolism , Reproducibility of Results , Solvents/chemistry , Species Specificity , Time Factors
12.
Br J Pharmacol ; 166(1): 276-96, 2012 May.
Article in English | MEDLINE | ID: mdl-22074238

ABSTRACT

BACKGROUND AND PURPOSE: Action potential (AP) recordings in ex vivo heart preparations constitute an important component of the preclinical cardiac safety assessment according to the ICH S7B guideline. Most AP measurement models are sensitive, predictive and informative but suffer from a low throughput. Here, effects of selected anti-arrhythmics (flecainide, quinidine, atenolol, sotalol, dofetilide, nifedipine, verapamil) on field/action potentials (FP/AP) of guinea pig and rabbit ventricular slices are presented and compared with data from established in vitro and in vivo models. EXPERIMENTAL APPROACH: Data from measurements of membrane currents (hERG, I(Na) ), AP/FP (guinea pig and rabbit ventricular slices), AP (rabbit Purkinje fibre), haemodynamic/ECG parameters (conscious, telemetered dog) were collected, compared and correlated to complementary published data (focused literature search). KEY RESULTS: The selected anti-arrhythmics, flecainide, quinidine, atenolol, sotalol, dofetilide, nifedipine and verapamil, influenced the shape of AP/FP of guinea pig and rabbit ventricular slices in a manner similar to that observed for rabbit PF. The findings obtained from slice preparations are in line with measurements of membrane currents in vitro, papillary muscle AP in vitro and haemodynamic/ECG parameters from conscious dogs in vivo, and were also corroborated by published data. CONCLUSION AND IMPLICATIONS: FP and AP recordings from heart slices correlated well with established in vitro and in vivo models in terms of pharmacology and predictability. Heart slice preparations yield similar results as papillary muscle but offer enhanced throughput for mechanistic investigations and may substantially reduce the use of laboratory animals.


Subject(s)
Action Potentials/drug effects , Anti-Arrhythmia Agents/pharmacology , Heart Ventricles/drug effects , Purkinje Fibers/drug effects , Animal Testing Alternatives , Animals , Dogs , Electrocardiography , Female , Guinea Pigs , Heart Ventricles/metabolism , Hemodynamics/drug effects , In Vitro Techniques , Male , Membrane Potentials/drug effects , Models, Biological , Purkinje Fibers/metabolism , Rabbits , Species Specificity
13.
Assay Drug Dev Technol ; 9(6): 600-7, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21675869

ABSTRACT

In investigating ion channel pharmacology, the manual patch clamp is still considered the gold standard for data quality, notwithstanding the major drawbacks of low throughput and the need for skilled operators. The automated patch clamp platform CytoPatch™ Instrument overcomes these restrictions. Its modular fully automated design makes it possible to obtain scalable throughput without the need for well-trained operators. Its chip design and perfusion system reproduces the manual patch technique, thus ensuring optimal data quality. Further, the use of stably transfected frozen cells, usable immediately after thawing, eliminates the cell quality impairment and low success rates associated with a running cell culture and renders screening costs accurately calculable. To demonstrate the applicability of this platform, 18 blinded compounds were assessed for their impact on the cardiac human Ether-à-go-go related gene K(+) channel. The IC(50) values obtained by the CytoPatch Instrument using the frozen human embryonic kidney 293 cells showed a high correlation (R(2)=0.928) with those obtained from manual patch clamp recordings with human embryonic kidney 293 cells from a running cell culture. Moreover, this correlation extended to sticky compounds such as terfenadine or astemizole. In conclusion, the CytoPatch Instrument operated with frozen cells ready to use directly after thawing provides the same high data quality known from the manual voltage clamp and has the added benefit of enhanced throughput for use in ion channel screening and safety assessment.


Subject(s)
Action Potentials/drug effects , Automation, Laboratory/methods , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Ether-A-Go-Go Potassium Channels/physiology , Software , Action Potentials/physiology , Automation, Laboratory/instrumentation , HEK293 Cells , Humans , Patch-Clamp Techniques/instrumentation , Patch-Clamp Techniques/methods , Potassium Channel Blockers/pharmacology
14.
J Pharmacol Toxicol Methods ; 62(1): 72-81, 2010.
Article in English | MEDLINE | ID: mdl-20570746

ABSTRACT

INTRODUCTION: BAY-79 is an inhibitor of receptor tyrosine kinases with high selectivity versus other kinases. Species scaling, complicated by nonlinear pharmacokinetics, predicted a C(max.u) of 36-178nmol/L at the human efficacious exposure. METHODS: Preclinical cardiovascular safety pharmacology studies assessed currents (hERG, I(Na)), action potential (AP, rabbit Purkinje fiber), hemodynamic/ECG parameters (anesthetized Beagle dogs, intravenous infusion), and proarrhythmic potential (rabbit Langendorff heart Screenit model). RESULTS: Both hERG K(+) current and hNav1.5 Na(+) current were inhibited with low potency (IC(20)>10micromol/L). Purkinje fiber APs remained unaffected at 10micromol/L, but at 100micromol/L displayed reverse use-dependent AP duration shortening (APD(90)-33% at 1Hz) and triangulation. Infusion of BAY-79 into anesthetized dogs was associated with moderate hemodynamic effects (increased heart rate and diastolic blood pressure, reduced stroke volume) and marked QTcV shortening (-25ms) starting at approximately 0.65micromol/L (unbound); QRS was not changed. Assessment of the proarrhythmic potential in the Screenit model showed effects (AP duration shortening, triangulation, instability, reduced coronary flow, slowed conduction) at > or =30micromol/L (0.5h/concentration) and at 3micromol/L with longer exposure (2.5h/concentration). DISCUSSION: BAY-79 at plasma concentrations slightly higher than those predicted to be therapeutically efficacious in humans is associated with QTc shortening in dogs but of unclear mechanistic basis. The QTc shortening associated proarrhythmic potential of BAY-79 together with other considerations finally resulted in an unfavorable risk-benefit assessment.


Subject(s)
Antineoplastic Agents/pharmacology , Arrhythmias, Cardiac/chemically induced , Drugs, Investigational/pharmacology , Electrocardiography/drug effects , Heart/drug effects , Organic Chemicals/pharmacology , Action Potentials/drug effects , Animals , Antineoplastic Agents/blood , Dogs , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Electrophysiologic Techniques, Cardiac , Ether-A-Go-Go Potassium Channels/metabolism , Female , HEK293 Cells , Heart/physiology , Heart/physiopathology , Humans , Male , Muscle Proteins/metabolism , NAV1.5 Voltage-Gated Sodium Channel , Organic Chemicals/administration & dosage , Organic Chemicals/blood , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacology , Purkinje Fibers/drug effects , Purkinje Fibers/physiology , Rabbits , Sodium Channels/metabolism
15.
Crit Rev Toxicol ; 38(9): 773-803, 2008.
Article in English | MEDLINE | ID: mdl-18941968

ABSTRACT

An increased sensitivity of the heart to catecholamines or cardiac sensitization is a recognized risk during acute human exposure to halogenated hydrocarbons used as solvents, foam-blowing or fire-extinguishing agents, refrigerants, and aerosol propellants. Although cardiac sensitization to such "industrial" halocarbons can result in serious arrhythmia and death, research into its mechanistic basis has been limited, whereas the literature on volatile anesthetics (e.g., halothane, chloroform) is comparably extensive. A review of the literature on halocarbons and related volatile anesthetics was conducted. The available experimental evidence suggests that volatile anesthetics at physiologically relevant concentrations interact predominantly with the main repolarizing cardiac potassium channels hERG and I(Ks), as well as with calcium and sodium channels at slightly higher concentrations. On the level of the heart, inhibition of these ion channels is prone to alter both action potential shape (triangulation) and electrical impulse conduction, which may facilitate arrhythmogenesis by volatile anesthetics per se and is potentiated by catecholamines. Action potential triangulation by regionally heterogeneous inhibition of calcium and potassium channels will facilitate catecholamine-induced afterdepolarizations, triggered activity, and enhanced automaticity. Inhibition of cardiac sodium channels will reduce conduction velocity and alter refractory period; this is potentiated by catecholamines and promotes reentry arrhythmias. Other cardiac and/or neuronal mechanisms might also contribute to arrhythmogenesis. The few scattered in vitro data available for halocarbons (e.g., FC-12, halon 1301, trichloroethylene) suggest inhibition of cardiac sodium (conduction), calcium and potassium channels (triangulation), extraneuronal catecholamine reuptake, and various neuronal ion channels. Therefore, it is hypothesized that halocarbons promote cardiac sensitization by similar mechanisms as volatile anesthetics. Experimental approaches for further investigation of these sensitization mechanisms by selected halocarbons are suggested.


Subject(s)
Anesthetics, Inhalation/adverse effects , Arrhythmias, Cardiac/chemically induced , Hydrocarbons, Halogenated/toxicity , Action Potentials , Anesthetics, Inhalation/pharmacology , Animals , Catecholamines/physiology , Heart/drug effects , Heart/physiology , Humans , Ion Channels/antagonists & inhibitors , Ion Channels/physiology
16.
J Pharmacol Toxicol Methods ; 58(2): 129-46, 2008.
Article in English | MEDLINE | ID: mdl-18585470

ABSTRACT

INTRODUCTION: Recent EU/US pediatric legislation and FDA/EMEA guidelines recognize the potential differences in safety profiles of drugs in adults versus young patients. Hence safety studies are recommended to investigate key functional domains of e.g. the developing CNS. METHODS: Selected psychoactive stimulants (caffeine, d-amphetamine, scopolamine) and depressants (baclofen, diazepam, haloperidol, chlorpromazine, imipramine, morphine) were characterized upon single administration with regard to behavioural parameters, locomotor activity, body temperature, pro-/anti-convulsive activity (pentylenetetrazole, PTZ), and nocifensive responses (hotplate) in neonatal (2 weeks), juvenile (4 weeks) and adult rats (8-9 weeks). RESULTS: In vehicle-treated rats, behavioural patterns matured with age, locomotor activity and handling-induced rise in body temperature were enhanced, whereas PTZ convulsion threshold dose and nocifensive response latency decreased. Single test compound treatment elicited behavioural effects characteristic for psychoactive drugs with stimulating and depressing properties regardless of age. However, incidence of certain behaviours, and magnitude of effects on locomotor activity and body temperature varied with age and became generally more pronounced in adult rats. Pro-/anti-convulsive effects and delayed nocifensive responses did not differ between juvenile and adult rats. CONCLUSION: CNS effects of selected psychoactive reference compounds were qualitatively similar, but quantitatively different in neonatal, juvenile and adult rats.


Subject(s)
Central Nervous System Depressants/adverse effects , Central Nervous System Stimulants/adverse effects , Central Nervous System/drug effects , Age Distribution , Animals , Behavior, Animal/drug effects , Body Temperature , Motor Activity/drug effects , Rats , Seizures/drug therapy , Toxicity Tests
17.
J Pharmacol Toxicol Methods ; 56(2): 145-58, 2007.
Article in English | MEDLINE | ID: mdl-17590357

ABSTRACT

INTRODUCTION: Regulatory guidelines require investigation of the liability for delayed ventricular repolarization by new chemical entities within a broad concentration range in-vitro. However, investigation can be limited by poor drug aqueous solubility, and by solvent physicochemical attributes that disrupt cell membrane integrity. Although excipients or solubilizing agents may aid to achieve the necessary high concentrations, no comprehensive overview on the suitability of solvents for in-vitro electrophysiological safety studies exists. METHODS: Excipients were tested for potential interference with the hERG (human ether-a-go-go-related gene) K(+) current (whole-cell voltage-clamp, 23+/-2 degrees C), and the shape of rabbit Purkinje fiber action potentials (conventional glass microelectrode technique, 37+/-1 degrees C). RESULTS AND DISCUSSION: Water-soluble complexation builders/carriers had little effect on hERG K(+) current at up to 50 mg/ml (BSA, bovine serum albumin) and 11 mg/ml (HP-beta-CD, hydroxypropyl-beta-cyclodextrin; IC(20), concentration of 20% inhibition). Water-soluble organic (co)solvents inhibited hERG K(+) currents (IC(20), %/mM): 0.7/152, ethanol; 0.9/67, Transcutol; 1.2/154, DMSO (dimethylsulfoxide); 1.6/389, acetonitrile; 1.9/48, polyethylene glycol 400; 2.1/660, methanol. Part of their inhibitory effect is attributed to the osmolality of extracellular solutions, because hERG IC(20) and extrapolated osmolality at the hERG IC(20) strongly correlate. Water-soluble non-ionic solubilizers/surfactants are potent inhibitors of hERG K(+) current with IC(20) concentrations of 0.07% (Cremophor EL) or lower (Tween 20, Tween 80: approximately 0.001%). Part of this inhibitory effect is attributed to their interaction with lipid membranes, because hERG inhibition occurs close to critical micelle concentrations (Cremophor, approximately 0.009%; Tween 20, approximately 0.007%). Purkinje fiber action potentials are little affected by HP-beta-CD at up to 2 mg/ml, while DMSO tends to shorten the action potential duration at 1%. CONCLUSION: When conducting electrophysiological in-vitro assessments of drug effects, solubilizers/surfactants (Cremophor EL, Tween 20, Tween 80) should be avoided. Instead, water-soluble organic (co)solvents (methanol, acetonitrile, DMSO) or complexation builders/carriers (HP-beta-CD, BSA) appear to be more favorable.


Subject(s)
Action Potentials/drug effects , Electrophysiology/methods , Ether-A-Go-Go Potassium Channels/physiology , Excipients/pharmacology , Purkinje Fibers/drug effects , 2-Hydroxypropyl-beta-cyclodextrin , Acetonitriles/chemistry , Acetonitriles/pharmacology , Animals , Cell Line , Dimethyl Sulfoxide/chemistry , Dimethyl Sulfoxide/pharmacology , Dose-Response Relationship, Drug , Droperidol/pharmacology , Drug Evaluation, Preclinical/methods , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Ether-A-Go-Go Potassium Channels/genetics , Ethylene Glycols/pharmacology , Excipients/chemistry , Female , Humans , Methanol/chemistry , Methanol/pharmacology , Piperidines/pharmacology , Polyethylene Glycols/pharmacology , Purkinje Fibers/physiology , Pyridines/pharmacology , Rabbits , Sotalol/pharmacology , beta-Cyclodextrins/pharmacology
18.
J Pharmacol Toxicol Methods ; 54(2): 130-40, 2006.
Article in English | MEDLINE | ID: mdl-16757186

ABSTRACT

INTRODUCTION: Drug-induced QT-prolongation, often based on hERG K+ current inhibition, has become a major safety concern during drug development. Hence, regulatory guidelines require combined in vitro and in vivo assays to assess the potential of new chemical entities to delay ventricular repolarization. Here, results of a pharmacological validation study with the torsadogenic compound sotalol are presented. METHODS: Alteration of ECG parameters was investigated in both conscious and anesthetized Beagle dogs (cumulative infusions of D,L-sotalol; n=6). The repolarization reserve of the latter was reduced by neurolept anesthesia using the hERG blocker droperidol (0.25 mg/kg/h yielding mean plasma concentrations of 0.5 microM). Furthermore, hERG K+ current and action potentials (AP; rabbit Purkinje fibers) were measured in vitro. RESULTS: The Fridericia corrected QT interval, QTcF, in conscious dogs (control: 254+/-15 ms), was dose-dependently prolonged by D,L-sotalol (+42 ms at plasma levels of 261 microM; dose 30 mg/kg). In anesthetized dogs, baseline QTcF (337+/-35 ms) was already prolonged compared to conscious dogs. In addition, QTcF-increase (+90 ms) was more pronounced at lower D,L-sotalol plasma levels (181 microM; dose 10 mg/kg), and proarrhythmic markers Tpeak-Tend and short term variability of QT were increased. These in vivo findings are supported by in vitro data. The hERG K+ current was blocked by D,L-sotalol (IC50 approximately 1.2 mM, IC20 approximately 250 microM) and droperidol (IC50 approximately 0.1 microM, IC20 approximately 0.02 microM). Purkinje fiber APs were concentration-dependently prolonged by D,L-sotalol (APD90:+60% at 30 microM) and droperidol (APD90:+55% at 1 microM). Low droperidol concentrations increased the sensitivity of Purkinje fibers towards D,L-sotalol-mediated AP prolongation. DISCUSSION: In conclusion, the higher sensitivity of anesthetized dogs towards sotalol-induced QT-prolongation is due to a reduced cardiac repolarization reserve caused by the hERG blocker droperidol. Hence, the droperidol-/fentanyl-/N2O-anesthetized dog is a particularly sensitive animal model for the detection of drug-induced QT-prolongation in safety pharmacology studies.


Subject(s)
Drug-Related Side Effects and Adverse Reactions , Long QT Syndrome/chemically induced , Action Potentials/drug effects , Adrenergic beta-Antagonists/toxicity , Animals , Anti-Arrhythmia Agents/toxicity , Antipsychotic Agents/toxicity , Cell Line , Data Interpretation, Statistical , Dogs , Droperidol/toxicity , Electrophysiology , Female , Humans , Infusions, Intravenous , Long QT Syndrome/physiopathology , Male , Patch-Clamp Techniques , Piperidines/toxicity , Pyridines/toxicity , Rabbits , Sotalol/toxicity , Transfection
19.
Eur J Pharmacol ; 474(1): 71-5, 2003 Aug 01.
Article in English | MEDLINE | ID: mdl-12909197

ABSTRACT

The application of cyclophosphamide to rats was used to induce interstitial cystitis. Behavioural studies indicated a strong pain reaction that developed within 2 h and levelled off thereafter causing a constant pain during the following 18 h. Neurons prepared from L6/S1 dorsal root ganglia innervating the urinary bladder responded to the application of capsaicin or alpha,beta-methylene ATP (alpha,beta-meATP) with an increase of intracellular Ca2+ ([Ca2+]i). The [Ca2+]i responses to capsaicin were identical in the dorsal root ganglion cells of cyclophosphamide- and saline-treated rats, whereas alpha,beta-meATP induced less increase in [Ca2+]i in the cyclophosphamide-treated animals than in their saline-treated counterparts. Hence, alpha,beta-meATP-sensitive P2X3 and/or P2X2/3 receptors of L6/S1 dorsal root ganglion neurons were functionally downregulated during subacute pain caused by experimental cystitis. In contrast, capsaicin-sensitive vanilloid 1 receptors did not react to the same procedure. Thoracal dorsal root ganglia, not innervating the urinary bladder, were also unaltered in their responsiveness to alpha,beta-meATP by cyclophosphamide treatment.


Subject(s)
Cystitis, Interstitial/metabolism , Ganglia, Spinal/metabolism , Receptors, Drug/biosynthesis , Receptors, Purinergic P2/biosynthesis , Animals , Calcium/metabolism , Cyclophosphamide/toxicity , Cystitis, Interstitial/chemically induced , Down-Regulation , Neurons/metabolism , Pain/metabolism , Rats , Rats, Inbred Strains , Receptors, Purinergic P2X2 , TRPV Cation Channels , Urinary Bladder/innervation
20.
Basic Res Cardiol ; 97(6): 434-44, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12395205

ABSTRACT

Human atrial myocytes were cultured under systematically varied conditions in order to obtain stable cells for future gene manipulation. Transient (I(to)) and sustained outward current (I(so)), and voltage- and muscarinic receptor-activated inward rectifier K(+) currents (I(K1), I(K,ACh)) were measured in freshly isolated cells and after 5 days in culture. Myocytes were grown on polylysin or laminin in medium with or without 10 % serum (medium+S, medium-S). Cultured myocytes dedifferentiated to a greater extent in medium+S than medium-S, but independent of the chemical nature of the adherence surface. Apparent surface area increased in medium+S, whereas membrane capacitance declined under all culture conditions. I(to) of myocytes cultured in medium-S was increased. Myocytes grown on polylysin and laminin exhibited reduced I(K1) current density. Under all culture conditions, I(K,ACh) was attenuated with carbachol but hardly affected with sphingosine-1-phosphate as agonists. In conclusion, morphological and electrophysiological changes depended on serum in the culture medium rather than on adherence surface being coated with laminin or polylysin.


Subject(s)
Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Potassium Channels, Inwardly Rectifying/physiology , Acetylcholine/pharmacology , Barium/pharmacology , Cells, Cultured , Culture Media, Serum-Free/pharmacology , Drug Resistance , Electric Capacitance , Electric Conductivity , Fluorescent Antibody Technique , Fluorescent Dyes , Heart Atria , Humans , Potassium Channels, Inwardly Rectifying/drug effects , Pyridinium Compounds , Staining and Labeling
SELECTION OF CITATIONS
SEARCH DETAIL
...