Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
J Pharmacol Toxicol Methods ; 128: 107524, 2024 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-38852689

RESUMEN

BACKGROUND: Determination of a drug's potency in blocking the hERG channel is an established safety pharmacology study. Best practice guidelines have been published for reliable assessment of hERG potency. In addition, a set of plasma concentration and plasma protein binding fraction data were provided as denominators for margin calculations. The aims of the current analysis were five-fold: provide data allowing creation of consistent denominators for the hERG margin distributions of the key reference agents, explore the variation in hERG margins within and across laboratories, provide a hERG margin to 10 ms QTc prolongation based on several newer studies, provide information to use these analyses for reference purposes, and provide recommended hERG margin 'cut-off' values. METHODS: The analyses used 12 hERG IC50 'best practice' data sets (for the 3 reference agents). A group of 5 data sets came from a single laboratory. The other 7 data sets were collected by 6 different laboratories. RESULTS: The denominator exposure distributions were consistent with the ICH E14/S7B Training Materials. The inter-occasion and inter-laboratory variability in hERG IC50 values were comparable. Inter-drug differences were most important in determining the pooled margin variability. The combined data provided a robust hERG margin reference based on best practice guidelines and consistent exposure denominators. The sensitivity of hERG margin thresholds were consistent with the sensitivity described over the course of the last two decades. CONCLUSION: The current data provide further insight into the sensitivity of the 30-fold hERG margin 'cut-off' used for two decades. Using similar hERG assessments and these analyses, a future researcher can use a hERG margin threshold to support a negative QTc integrated risk assessment.

2.
Clin Pharmacol Ther ; 116(1): 96-105, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38362953

RESUMEN

Oligonucleotide therapeutics (ONTs) represent a new modality with unique pharmacological and chemical properties that modulate gene expression with a high degree of target specificity mediated by complementary Watson-Crick base pair hybridization. To date, the proarrhythmic assessment of ONTs has been influenced by International Conference on Harmonization (ICH) E14 and S7B guidance. To document current hERG/QTc evaluation practices, we reviewed US Food and Drug Administration (FDA) and the European Medicines Agency (EMA) Approval Packages (source: PharmaPendium.com) and collated preclinical and clinical studies for 17 marketed ONTs. In addition, clinical QTc data from 12 investigational ONTs were obtained from the literature. Of the marketed ONTs, eight were tested in the hERG assay with no inhibitory effect identified at the top concentration (range: 34-3,000 µM) tested. Fourteen of the ONTs were evaluated in nonhuman primate cardiovascular studies with 11 of them in dedicated telemetry studies. No effect on QTc intervals were observed (at high exposure multiples) in all studies. Clinically, four ONTs were evaluated in TQT studies; an additional six ONTs were assessed by concentration-QTc interval analysis, and six by routine safety electrocardiogram monitoring. None of the clinical studies identified a QTc prolongation risk; the same was true for the 12 investigational ONTs. A search of the FDA Adverse Event Database indicated no association between approved ONTs and proarrhythmias. Overall, the collective weight of evidence from 29 ONTs demonstrate no clinical proarrhythmic risk based on data obtained from ICH S7B/E14 studies. Thus, new ONTs may benefit from reduced testing strategies because they have no proarrhythmic risk, a similar cardiac safety profile as monoclonal antibodies, proteins, and peptides.


Asunto(s)
Arritmias Cardíacas , Oligonucleótidos , Humanos , Animales , Arritmias Cardíacas/inducido químicamente , Oligonucleótidos/efectos adversos , Oligonucleótidos/farmacología , Oligonucleótidos/uso terapéutico , United States Food and Drug Administration , Estados Unidos , Aprobación de Drogas , Medición de Riesgo , Síndrome de QT Prolongado/inducido químicamente , Electrocardiografía
3.
J Pharmacol Toxicol Methods ; 123: 107278, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37268094

RESUMEN

Understanding translation from preclinical observations to clinical findings is important for evaluating the efficacy and safety of novel compounds. Of relevance to cardiac safety is profiling drug effects on cardiomyocyte (CM) sarcomere shortening and intracellular Ca2+ dynamics. Although CM from different animal species have been used to assess such effects, primary human CM isolated from human organ donor heart represent an ideal non-animal alternative approach. We performed a study to evaluate primary human CM and have them compared to freshly isolated dog cardiomyocytes for their basic function and responses to positive inotropes with well-known mechanisms. Our data showed that simultaneous assessment of sarcomere shortening and Ca2+-transient can be performed with both myocytes using the IonOptix system. Amplitude of sarcomere shortening and Ca2+-transient (CaT) were significantly higher in dog compared to human CM in the basic condition (absence of treatment), while longer duration of sarcomere shortening and CaT were observed in human cells. We observed that human and dog CMs have similar pharmacological responses to five inotropes with different mechanisms, including dobutamine and isoproterenol (ß-adrenergic stimulation), milrinone (PDE3 inhibition), pimobendan and levosimendan (increase of Ca2+sensitization as well as PDE3 inhibition). In conclusion, our study suggests that myocytes obtained from both human donor hearts and dog hearts can be used to simultaneously assess drug-induced effects on sarcomere shortening and CaT using the IonOptix platform.


Asunto(s)
Trasplante de Corazón , Miocitos Cardíacos , Humanos , Perros , Animales , Calcio , Sarcómeros/fisiología , Contracción Miocárdica , Donantes de Tejidos
4.
Nucleic Acid Ther ; 33(2): 132-140, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36576986

RESUMEN

In accord with International Conference on Harmonization S7B guidelines, an in vitro human ether-a-go-go-related gene (hERG) assay is one component of an integrated risk assessment for delayed ventricular repolarization. Function of hERG could be affected by direct (acute) mechanisms, or by indirect (chronic) mechanisms. Some approved oligonucleotide therapeutics had submitted hERG data to regulatory agents, which were all collected with the same protocol used for small-molecule testing (incubation time <20 min; acute), however, oligonucleotides have unique mechanisms and time courses of action (indirect). To reframe the hERG testing strategy for silencing RNA (siRNA), an investigation was performed to assess the time course for siRNA-mediated inhibition of hERG function and gene expression. Commercially available siRNAs of hERG were evaluated in a stable hERG-expressed cell line by whole-cell voltage clamp using automated electrophysiology and polymerase chain reaction. In the acute hERG study, no effects were observed after treatment with 100 nM siRNA for 20 min. The chronic effects of 100 nM siRNAs on hERG function were evaluated and recorded over 8-48 h following transfection. At 8 h there was no significant effect, whereas 77% reduction was observed at 48 h. Measurement of hERG mRNA levels demonstrated a 79% and 93% decrease of hERG mRNA at 8 and 48 h, respectively, consistent with inhibition of hERG transcription. The results indicate that an anti-hERG siRNA requires a long exposure time (48 h) in the hERG assay to produce a maximal reduction in hERG current; short exposures (20 min-8 h) had no effect. These findings imply that off-target profiling of novel oligonucleotides could benefit from using hERG protocol with long incubation times to de-risk potential off-target (indirect) effects on the hERG channel. This hERG assay modification may be important to consider if the findings are used to support an integrated nonclinical-clinical risk assessment for QTc (the duration of the QT interval adjusted for heart rate) prolongation.


Asunto(s)
Canales de Potasio Éter-A-Go-Go , Humanos , Canales de Potasio Éter-A-Go-Go/genética , Canales de Potasio Éter-A-Go-Go/metabolismo , Línea Celular , ARN Interferente Pequeño/genética
5.
J Pharmacol Toxicol Methods ; 111: 107082, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34082139

RESUMEN

INTRODUCTION: Cardioplegic solutions were first developed to preserve heart function during cardiac surgeries and heart transplants but have application in the nonclinical setting. Due to lack of lab space in the vivarium, cardioplegic solution was used to conserve cardiac function for ex-vivo studies performed in a separate building. All studies in this report were conducted with isolated female rabbit hearts (IRHs) via retrograde perfusion using the Langendorff apparatus to investigate if cardioplegia usage affects cardiac function. METHODS: Cardioplegia was achieved with a hyperkalemia (27 mM KCL) solution kept at 4 °C. Cardiac function was assessed by measuring ECG parameters, left ventricular contractility, and coronary flow under constant perfusion pressure. IRHs were cannulated with Krebs Henseleit buffer (KH) either fresh or after cardioplegic solution storage (C-IRH). Three comparisons were performed with and without cardioplegia; (i) direct side-by side studies of cardiac function; (ii) pharmacological responses to typical ion channels blockers, dofetilide, flecainide, and diltiazem; (iii) retrospective evaluation of cardiac functions in a large sample of hearts. RESULTS: In the side-by-side comparisons, cardioplegia-stored IRHs (C-IRH; storage time 90 min) had similar electrocardiographic (ECG) and hemodynamic parameters to fresh-cannulated hearts with KH buffer (KH-IRH). In addition, responses to dofetilide, flecainide, and diltiazem, were similar for C-IRH and KH-IRH hearts. Over the years (2006-2011), baseline data was collected from 79 hearts without cardioplegia and 100 hearts with cardioplegia (C-IRH; storage time 15 min), which showed no meaningful differences in a retrospective analysis. DISCUSSION: Cardiac function was preserved after cardioplegic treatment, however, coronary flow rates were decreased (-19.3%) in C-IRH hearts which indicated an altered coronary vascular tone. In conclusion, storage in cardioplegic solution preserves rabbit cardiac function, a practice that enables heart tissues to be collected at one site (e.g., vivarium) and transported to a laboratory in a separate location.


Asunto(s)
Soluciones Cardiopléjicas , Paro Cardíaco Inducido , Animales , Soluciones Cardiopléjicas/farmacología , Femenino , Corazón , Hemodinámica , Conejos , Estudios Retrospectivos
6.
Clin Transl Sci ; 14(4): 1600-1610, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33955165

RESUMEN

Omecamtiv mecarbil (OM) is a myosin activator (myotrope), developed as a potential therapeutic agent for heart failure with reduced ejection fraction. To characterize the potential pro-arrhythmic risk of this novel sarcomere activator, we evaluated OM in a series of International Conference on Harmonization S7B core and follow-up assays, including an in silico action potential (AP) model. OM was tested in: (i) hERG, Nav1.5 peak, and Cav1.2 channel assays; (ii) in silico computation in a human ventricular AP (hVAP) population model; (iii) AP recordings in canine cardiac Purkinje fibers (PF); and (iv) electrocardiography analysis in isolated rabbit hearts (IRHs). OM had low potency in the hERG (half-maximal inhibitory concentration [IC50 ] = 125.5 µM) and Nav1.5 and Cav1.2 assays (IC50  > 300 µM). These potency values were used as inputs to investigate the occurrence of repolarization abnormalities (biomarkers of pro-arrhythmia) in an hVAP model over a wide range of OM concentrations. The outcome of hVAP analysis indicated low pro-arrhythmia risk at OM concentration up to 30 µM (100-fold the effective free therapeutic plasma concentration). In the isolated canine PF assay, OM shortened AP duration (APD)60 and APD90 significantly from 3 to 30 µM. In perfused IRH, ventricular repolarization (corrected QT and corrected JT intervals) was decreased significantly at greater than or equal to 1 µM OM. In summary, the comprehensive proarrhythmic assessment in human and non-rodent cardiac models provided data indicative that OM did not delay ventricular repolarization at therapeutic relevant concentrations, consistent with clinical findings.


Asunto(s)
Arritmias Cardíacas/diagnóstico , Insuficiencia Cardíaca/tratamiento farmacológico , Urea/análogos & derivados , Potenciales de Acción/efectos de los fármacos , Animales , Arritmias Cardíacas/inducido químicamente , Simulación por Computador , Perros , Evaluación Preclínica de Medicamentos , Ventrículos Cardíacos/efectos de los fármacos , Humanos , Preparación de Corazón Aislado , Miocitos Cardíacos/efectos de los fármacos , Cultivo Primario de Células , Ramos Subendocárdicos , Conejos , Urea/administración & dosificación , Urea/efectos adversos
7.
Clin Pharmacol Ther ; 109(2): 310-318, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32866317

RESUMEN

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.


Asunto(s)
Drogas en Investigación/efectos adversos , Síndrome de QT Prolongado/inducido químicamente , Animales , Arritmias Cardíacas/inducido químicamente , Desarrollo de Medicamentos/métodos , Industria Farmacéutica/métodos , Electrocardiografía/métodos , Humanos , Medición de Riesgo , Torsades de Pointes/inducido químicamente
8.
Pharmacol Res Perspect ; 8(5): e00656, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32969560

RESUMEN

Omecamtiv mecarbil (OM) is a selective cardiac myosin activator (myotrope), currently in Phase 3 clinical investigation as a novel treatment for heart failure with reduced ejection fraction. OM increases cardiac contractility by enhancing interaction between myosin and actin in a calcium-independent fashion. This study aims to characterize the mechanism of action by evaluating its simultaneous effect on myocyte contractility and calcium-transients (CTs) in healthy canine ventricular myocytes. Left ventricular myocytes were isolated from canines and loaded with Fura-2 AM. With an IonOptix system, contractility parameters including amplitude and duration of sarcomere shortening, contraction and relaxation velocity, and resting sarcomere length were measured. CT parameters including amplitude at systole and diastole, velocity at systole and diastole, and duration at 50% from peak were simultaneously measured. OM was tested at 0.03, 0.1, 0.3, 1, and 3 µmol\L concentrations to simulate therapeutic human plasma exposure levels. OM and isoproterenol (ISO) demonstrated differential effects on CTs and myocyte contractility. OM increased contractility mainly by prolonging duration of contraction while ISO increased contractility mainly by augmenting the amplitude of contraction. ISO increased the amplitude and velocity of CT, shortened duration of CT concurrent with increasing myocyte contraction, while OM did not change the amplitude, velocity, and duration of CT up to 1 µmol\L. Decreases in relaxation velocity and increases in duration were present only at 3 µmol\L. In this translational myocyte model study, therapeutically relevant concentrations of OM increased contractility but did not alter intracellular CTs, a mechanism of action distinct from traditional calcitropes.


Asunto(s)
Señalización del Calcio/efectos de los fármacos , Ventrículos Cardíacos/citología , Isoproterenol/farmacología , Células Musculares/citología , Urea/análogos & derivados , Animales , Perros , Relación Dosis-Respuesta a Droga , Fura-2/análogos & derivados , Fura-2/química , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/metabolismo , Humanos , Masculino , Modelos Animales , Células Musculares/química , Células Musculares/efectos de los fármacos , Células Musculares/metabolismo , Contracción Muscular/efectos de los fármacos , Sarcómeros/efectos de los fármacos , Sarcómeros/fisiología , Urea/farmacología
9.
J Pharmacol Toxicol Methods ; 105: 106886, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32629159

RESUMEN

To develop therapeutics for cardiovascular disease, especially heart failure, translational models for assessing cardiac contractility are necessary for preclinical target validation and lead optimization. The availability of stem cell-derived cardiomyocytes (SC-CM) has generated a great opportunity in developing new in-vitro models for assessing cardiac contractility. However, the immature phenotype of SC-CM is a well-recognized limitation in inotropic evaluation, especially regarding the lack of or diminished positive inotropic response to ß-adrenergic agonists. Recent development of 3D engineered cardiac tissues (ECTs) using human induced pluripotent stem cell derived-cardiomyocytes (hiPSC-CM) in the BiowireTM II platform has shown improved maturation. To evaluate their suitability to detect drug-induced changes in cardiac contractility, positive inotropes with diverse mechanisms, including ß-adrenergic agonists, PDE3 inhibitors, Ca2+-sensitizers, myosin and troponin activators, and an apelin receptor agonist, were tested blindly. A total of 8 compounds were evaluated, including dobutamine, milrinone, pimobendan, levosimendan, omecamtiv mecarbil, AMG1, AMG2, and pyr-apelin-13. Contractility was evaluated by analyzing the amplitude, velocity and duration of contraction and relaxation. All tested agents, except pyr-apelin-13, increased contractility by increasing the amplitude of contraction and velocity. In addition, myosin and troponin activators increase contraction duration. These results indicate that ECTs generated in the BiowireTM II platform can identify inotropes with different mechanisms and provides a human-based in-vitro model for evaluating potential therapeutics.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Miocitos Cardíacos/citología , Regeneración/fisiología , Andamios del Tejido/química , Células Cultivadas , Humanos , Contracción Miocárdica/fisiología
10.
Regul Toxicol Pharmacol ; 115: 104697, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32590049

RESUMEN

Romosozumab (EVENITY™ [romosozumab-aqqg in the US]) is a humanized monoclonal antibody that inhibits sclerostin and has been approved in several countries for the treatment of osteoporosis in postmenopausal women at high risk of fracture. Sclerostin is expressed in bone and aortic vascular smooth muscle (AVSM). Its function in AVSM is unclear but it has been proposed to inhibit vascular calcification, atheroprogression, and inflammation. An increased incidence of positively adjudicated serious cardiovascular adverse events driven by an increase in myocardial infarction and stroke was observed in romosozumab-treated subjects in a clinical trial comparing alendronate with romosozumab (ARCH; NCT01631214) but not in a placebo-controlled trial (FRAME; NCT01575834). To investigate the effects of sclerostin inhibition with sclerostin antibody on the cardiovascular system, a comprehensive nonclinical toxicology package with additional cardiovascular studies was conducted. Although pharmacodynamic effects were observed in the bone, there were no functional, morphological, or transcriptional effects on the cardiovascular system in animal models in the presence or absence of atherosclerosis. These nonclinical studies did not identify evidence that proves the association between sclerostin inhibition and adverse cardiovascular function, increased cardiovascular calcification, and atheroprogression.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Anticuerpos Monoclonales/farmacología , Conservadores de la Densidad Ósea/farmacología , Sistema Cardiovascular/efectos de los fármacos , Animales , Anticuerpos Monoclonales/uso terapéutico , Conservadores de la Densidad Ósea/uso terapéutico , Evaluación Preclínica de Medicamentos , Femenino , Fracturas Óseas/prevención & control , Humanos , Macaca fascicularis , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE , Osteoporosis/tratamiento farmacológico , Ratas Sprague-Dawley , Riesgo
11.
JCI Insight ; 5(8)2020 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-32208384

RESUMEN

Heart failure (HF) remains a grievous illness with poor prognosis even with optimal care. The apelin receptor (APJ) counteracts the pressor effect of angiotensin II, attenuates ischemic injury, and has the potential to be a novel target to treat HF. Intravenous administration of apelin improves cardiac function acutely in patients with HF. However, its short half-life restricts its use to infusion therapy. To identify a longer acting APJ agonist, we conducted a medicinal chemistry campaign, leading to the discovery of potent small-molecule APJ agonists with comparable activity to apelin by mimicking the C-terminal portion of apelin-13. Acute infusion increased systolic function and reduced systemic vascular resistance in 2 rat models of impaired cardiac function. Similar results were obtained in an anesthetized but not a conscious canine HF model. Chronic oral dosing in a rat myocardial infarction model reduced myocardial collagen content and improved diastolic function to a similar extent as losartan, a RAS antagonist standard-of-care therapy, but lacked additivity with coadministration. Collectively, this work demonstrates the feasibility of developing clinical, viable, potent small-molecule agonists that mimic the endogenous APJ ligand with more favorable drug-like properties and highlights potential limitations for APJ agonism for this indication.


Asunto(s)
Receptores de Apelina/agonistas , Corazón/efectos de los fármacos , Animales , Perros , Descubrimiento de Drogas , Insuficiencia Cardíaca , Péptidos y Proteínas de Señalización Intercelular , Ratas
12.
Front Pharmacol ; 10: 1643, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-32082155

RESUMEN

Torsades de Pointes (TdP) is a type of ventricular arrhythmia which could be observed as an unwanted drug-induced cardiac side effect, and it is associated with repolarization abnormalities in single cells. The pharmacological evaluations of TdP risk in previous years mainly focused on the hERG channel due to its vital role in the repolarization of cardiomyocytes. However, only considering drug effects on hERG led to false positive predictions since the drug action on other ion channels can also have crucial regulatory effects on repolarization. To address the limitation of only evaluating hERG, the Comprehensive in Vitro Proarrhythmia Assay initiative has proposed to systematically integrate drug effects on multiple ion channels into in silico drug trial to improve TdP risk assessment. It is not clear how many ion channels are sufficient for reliable TdP risk predictions, and whether differences in IC50 and Hill coefficient values from independent sources can lead to divergent in silico prediction outcomes. The rationale of this work is to investigate the above two questions using a computationally efficient population of human ventricular cells optimized to favor repolarization abnormality. Our blinded results based on two independent data sources confirm that simulations with the optimized population of human ventricular cell models enable efficient in silico drug screening, and also provide direct observation and mechanistic analysis of repolarization abnormality. Our results show that 1) the minimum set of ion channels required for reliable TdP risk predictions are Nav1.5 (peak), Cav1.2, and hERG; 2) for drugs with multiple ion channel blockage effects, moderate IC50 variations combined with variable Hill coefficients can affect the accuracy of in silico predictions.

13.
J Pharmacol Toxicol Methods ; 93: 98-107, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29908289

RESUMEN

Contractility measurements using primary isolated cardiac myocytes (CM) have commonly been used in understanding the physiology and pharmacology of cellular mechanics. In the majority of studies, CM from healthy animals were used, and fewer studies were performed with CM from diseased hearts. To better understand the translational value of contractility on the cellular level of a diseased animal model, myocytes were isolated from left ventricles of a tachypacing-induced heart failure (HF) canine model, and their contractility was measured by recording sarcomere shortening using an image-based IonOptix video system. A side-by-side comparison study was performed in myocytes isolated from 13 normal and 5 tachypacing-induced HF canines by evaluating both basal contractility and pharmacological responses to inotropic agents with different mechanisms, including dobutamine, isoproterenol, milrinone, levosimendan, pimobendan, diltiazem, and flecainide. Myocytes isolated from HF canines exhibited compromised contractility at the sarcomere level in comparison to normal myocytes, specifically, HF myocytes have smaller sarcomere contraction amplitude, longer resting sarcomere length, slower velocity of contraction and relaxation. In addition, they have altered pharmacological responses compared to that of normal canines, with much less potent effects observed in the application of classic inotropic agents, such as isoproterenol, dobutamine, and milrinone. These results indicate that myocytes isolated from tachy-paced HF canines have altered physiological and pharmacological properties, which could be utilized for understanding pathophysiology and developing pharmacological interventions for HF.


Asunto(s)
Estimulación Cardíaca Artificial/efectos adversos , Cardiotónicos/farmacología , Insuficiencia Cardíaca/etiología , Contracción Miocárdica/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Animales , Células Cultivadas , Perros , Relación Dosis-Respuesta a Droga , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/fisiopatología , Isoproterenol/farmacología , Masculino , Contracción Miocárdica/fisiología , Miocitos Cardíacos/fisiología
14.
Front Physiol ; 8: 1109, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29354071

RESUMEN

To assess drug-induced pro-arrhythmic risk, especially Torsades de Pointe (TdP), new models have been proposed, such as in-silico modeling of ventricular action potential (AP) and stem cell-derived cardiomyocytes (SC-CMs). Previously we evaluated the electrophysiological profile of 15 reference drugs in hESC-CMs and hiPSC-CMs for their effects on intracellular AP and extracellular field potential, respectively. Our findings indicated that SC-CMs exhibited immature phenotype and had the propensity to generate false positives in predicting TdP risk. To expand our knowledge with mature human cardiac tissues for drug-induced pro-arrhythmic risk assessment, human ventricular trabeculae (hVT) from ethically consented organ donors were used to evaluate the effects of the same 15 drugs (8 torsadogenic, 5 non-torsadogenic, and 2 discovery molecules) on AP parameters at 1 and 2 Hz. Each drug was tested blindly with 4 concentrations in duplicate trabeculae from 2 hearts. To identify the pro-arrhythmic risk of each drug, a pro-arrhythmic score was calculated as the weighted sum of percent drug-induced changes compared to baseline in various AP parameters, including AP duration and recognized pro-arrhythmia predictors such as triangulation, beat-to-beat variability and incidence of early-afterdepolarizations, at each concentration. In addition, to understand the translation of this preclinical hVT AP-based model to clinical studies, a ratio that relates each testing concentration to the human therapeutic unbound Cmax (Cmax) was calculated. At a ratio of 10, for the 8 torsadogenic drugs, 7 were correctly identified by the pro-arrhythmic score; 1 was mislabeled. For the 5 non-torsadogenic drugs, 4 were correctly identified as safe; 1 was mislabeled. Calculation of sensitivity, specificity, positive predictive value, and negative predictive value indicated excellent performance. For example, at a ratio of 10, scores for sensitivity, specificity, positive predictive value and negative predictive values were 0.88, 0.8, 0.88 and 0.8, respectively. Thus, the hVT AP-based model combined with the integrated analysis of pro-arrhythmic score can differentiate between torsadogenic and non-torsadogenic drugs, and has a greater predictive performance when compared to human SC-CM models.

15.
Toxicol Sci ; 147(1): 286-95, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26117837

RESUMEN

Evaluation of stem cell-derived cardiomyocytes (SC-CM) using multi-electrode array (MEA) has attracted attention as a novel model to detect drug-induced arrhythmia. An experiment was conducted to determine if MEA recording from human induced pluripotent SC-CM (hiPSC-CM) could assess proarrhythmic risk. Ten hERG blockers, 4 Na(+) blockers, and 1 IKs blocker were evaluated blindly. Eight drugs are associated with Torsades de Pointes (TdP) and 4 are not. Multiple parameters, including field potential duration (FPD), Na(+) slope, Na(+) amplitude, beat rate (BR), and early after-depolarization (EAD) were recorded. Minimum effective concentrations (MEC) that elicited a significant change were calculated. Our results determined that FPD and EAD were unable to distinguish torsadogenic from benign compounds, Na(+) slope and amplitude could not differentiate Na(+) channel blockade from hERG blockade, BR had an inconsistent response to pharmacological treatment, and that hiPSC-CM were, in general, insensitive to IKs inhibition. A ratio was calculated that relates MEC for evoking FPD prolongation, or triggering EAD, to the human therapeutic unbound Cmax (MEC/Cmax). The key finding was that the ratio was sensitive, but specificity was low. Consistently, the ratio had high positive predictive value and low negative predictive value. In conclusion, MEA recordings of hiPSC-CM were sensitive for FPD and EAD detection, but unable to distinguish agents with low- and high-risk for TdPs. Although some published reports suggested great potential for MEA recordings in hSC-CM to assess preclinical cardiac toxicity, the current evaluation implies that this model would have a high false-positive rate in regard to proarrhythmic risk.


Asunto(s)
Arritmias Cardíacas/inducido químicamente , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Arritmias Cardíacas/patología , Relación Dosis-Respuesta a Droga , Frecuencia Cardíaca/efectos de los fármacos , Humanos , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/efectos de los fármacos , Potenciales de la Membrana/efectos de los fármacos , Microelectrodos , Bloqueadores de los Canales de Potasio/toxicidad , Valor Predictivo de las Pruebas , Reproducibilidad de los Resultados , Medición de Riesgo , Bloqueadores de los Canales de Sodio/toxicidad , Torsades de Pointes/inducido químicamente , Torsades de Pointes/fisiopatología
16.
Pharmacol Res Perspect ; 3(1): e00102, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25692020

RESUMEN

Oxytocin, a nine amino acid peptide, is highly conserved in placental mammals, including humans. Oxytocin has a physiological role in parturition and parenteral administration of the synthetic peptide is used to induce labor and control postpartum hemorrhage. Endogenous levels of oxytocin before labor are ∼20 pg/mL, but pharmacological administration of the peptide can achieve levels of 110 pg/mL (0.1 nmol/L) following intravenous administration. Cardiac arrhythmia and premature ventricular contractions have been associated with oxytocin administration in addition to QTc interval prolongation. In the conscious rabbit model, intravenous oxytocin produced QT and QTc prolongation. The mechanism of oxytocin-induced QTc prolongation is uncertain but could be the result of indirect changes in autonomic nervous tone, or a direct effect on the duration of cardiomyocyte repolarization. The purpose of this study was to examine the ability of oxytocin to alter cardiac repolarization directly. Two conventional models were used: QTc interval evaluation in the isolated rabbit heart (IRH) and assessment of action potential duration (APD) in human ventricular myocytes (HVM). Oxytocin did not prolong QTc intervals in IRH or APD in HVM when tested at suprapharmacological concentrations, for example, up to 1 µmol/L. The results indicate that oxytocin has very low risk for eliciting QTc and APD prolongation directly, and infer that the QTc changes observed in vivo may be attributed to an indirect mechanism.

17.
J Pharmacol Toxicol Methods ; 68(1): 74-81, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23518063

RESUMEN

INTRODUCTION: Cardiac safety is of paramount importance in contemporary drug development. Efficient and sensitive evaluation of cardiac safety in the research and development of new molecular agents begins with preclinical in-vitro models. A new model that is currently under evaluation is the human embryonic stem-cell derived cardiac myocytes (hESC-CM) (Peng, Lacerda, Kirsch, Brown, & Bruening-Wright, 2010). METHODS: hESC-CM were exposed in-vitro to 15 test compounds, and action potentials (AP) recorded with perforated patch-clamp technique to assess changes in AP duration (APD90) and upstroke velocity (Vmax). The test compounds included: 10 hERG channel, 4 Na⁺ channel, and 1 IKs channel inhibitors. For comparison purposes, the test compounds were evaluated in the isolated rabbit heart assay (IRH) to determine changes in conduction (QRS) and repolarization (QTc). Potency at hERG, NaV1.5 and IKs channel was also determined. RESULTS: For 7 of 10 hERG channel inhibitors, the potency values across the three functional assays were similar (≤5-fold). Three compounds (dofetilide, sertindole, and terfenadine) showed >10-fold discrepancy between hERG potency and inhibitory concentrations in the hESC-CM and IRH assays. Of the four Na⁺ channel inhibitors, only mexiletine exhibited similar potency values across the three assays (~3-fold); the others exhibited marked variation (>10-fold) in inhibitory potency. No effect on repolarization was observed in hESC-CM treated with a potent IKs blocker, but QTc prolongation was evident in the IRH. DISCUSSION: The functional data indicate that hESC-CM are sensitive for detecting repolarization delay induced by hERG channel blockade, and AP prolongation correlated with potency in the hERG channel and IRH assays. However, hESC-CM were less sensitive for detecting depolarizing delay by Na⁺ channel blockers, and unable to detect delayed repolarization caused by IKs blockade.


Asunto(s)
Células Madre Embrionarias/citología , Miocitos Cardíacos/efectos de los fármacos , Pruebas de Toxicidad/métodos , Potenciales de Acción/efectos de los fármacos , Animales , Diseño de Fármacos , Evaluación Preclínica de Medicamentos/métodos , Canal de Potasio ERG1 , Canales de Potasio Éter-A-Go-Go/efectos de los fármacos , Canales de Potasio Éter-A-Go-Go/metabolismo , Femenino , Humanos , Síndrome de QT Prolongado/inducido químicamente , Miocitos Cardíacos/metabolismo , Canal de Sodio Activado por Voltaje NAV1.5/efectos de los fármacos , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Técnicas de Placa-Clamp , Canales de Potasio con Entrada de Voltaje/efectos de los fármacos , Canales de Potasio con Entrada de Voltaje/metabolismo , Conejos
18.
Toxicol Appl Pharmacol ; 268(2): 113-22, 2013 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-23416206

RESUMEN

Itraconazole (ITZ) is an approved antifungal agent that carries a "black box warning" in its label regarding a risk of negative cardiac inotropy based on clinical findings. Since the mechanism of the negative inotropic effect is unknown, we performed a variety of preclinical and mechanistic studies to explore the pharmacological profile of ITZ and understand the negative inotropic mechanism. ITZ was evaluated in: (1) an isolated rabbit heart (IRH) preparation using Langendorff retrograde perfusion; (2) ion channel studies; (3) a rat heart mitochondrial function profiling screen; (4) a mitochondrial membrane potential (MMP) assay; (5) in vitro pharmacology profiling assays (148 receptors, ion channels, transporters, and enzymes); and (6) a kinase selectivity panel (451 kinases). In the IRH, ITZ decreased cardiac contractility (>30%) at 0.3µM, with increasing effect at higher concentrations, which indicated a direct negative inotropic effect upon the heart. It also decreased heart rate and coronary flow (≥1µM) and prolonged PR/QRS intervals (3µM). In mechanistic studies, ITZ inhibited the cardiac NaV channel (IC50: 4.2µM) and was devoid of any functional inhibitory effect at the remaining pharmacological targets. Lastly, ITZ did not affect MMP, nor interfere with mitochondrial enzymes or processes involved with fuel substrate utilization or energy formation. Overall, the cardiovascular and mechanistic data suggest that ITZ-induced negative inotropy is a direct effect on the heart, in addition, the potential involvement of mitochondria function and L-type Ca(2+) channels are eliminated. The exact mechanism underlying the negative inotropy is uncertain, and requires further study.


Asunto(s)
Antifúngicos/farmacología , Itraconazol/farmacología , Contracción Miocárdica/efectos de los fármacos , Función Ventricular Izquierda/efectos de los fármacos , Animales , Depresión Química , Femenino , Técnicas In Vitro , Canales Iónicos/efectos de los fármacos , MAP Quinasa Quinasa 5/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias Cardíacas/efectos de los fármacos , Mitocondrias Cardíacas/fisiología , Conejos , Ratas
19.
J Pharmacol Toxicol Methods ; 64(3): 251-7, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21888984

RESUMEN

INTRODUCTION: Measurement of drug-induced inhibition of potassium current flow through the hERG channel is used to determine potency at the channel, which is used as an in vitro risk assessment for QTc interval prolongation in vivo. In the hERG assay, test solutions of varying strength are prepared to construct a concentration-response curve based upon the nominal drug concentration (NOM). Dose-solution analysis (DSA) is an analytical approach to confirm the test concentration achieved in an in vitro assay (Herron, Towers, & Templeton, 2004), and can be included as a component of hERG channel study to confirm drug concentration in the assay buffer to determine potency using the "actual" drug level in solution (ACT). Thus, DSA could be helpful in confirming test article concentrations. This study examined whether inclusion of DSA improved the accuracy of potency estimates based upon the ACT compared to the NOM concentration during hERG voltage clamp assays (non-GLP) for 99 diverse agents. METHODS: We examined the correlation of hERG IC(50) derived from NOM with hERG IC(50) derived from ACT, and analyzed potential mechanisms of deviation between ACT and NOM potency values, including solubility, cLogP, PKa, and molecular weights. RESULTS: Seventy-four (74) of 99 agents (73.7%) had NOM- and ACT-derived IC(50) values within 3-fold, 87 of 99 (87.8%) had an IC(50) ratio within 10-fold, and 12 (12.1%) had a >10-fold difference in their NOM IC(50) and ACT IC(50) values. On average, these 12 compounds had less soluble, more lipophilic (high cLogP values), and more basic characters (high pKa values). DISCUSSION: Our investigation indicated that DSA did not alter hERG potency estimation for the majority of compounds in this dataset, i.e., DSA confirmed the NOM concentration within 3-fold. For poorly soluble agents or agents with high cLogP and pKa values, however, DSA did not clarify or improve hERG potency estimates.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Canales de Potasio Éter-A-Go-Go/antagonistas & inhibidores , Canales de Potasio Éter-A-Go-Go/metabolismo , Línea Celular Transformada , Relación Dosis-Respuesta a Droga , Canal de Potasio ERG1 , Electrofisiología/métodos , Células HEK293 , Humanos , Potenciales de la Membrana/efectos de los fármacos , Técnicas de Placa-Clamp/métodos , Bloqueadores de los Canales de Potasio/farmacología , Solubilidad , Soluciones/química
20.
J Pharmacol Exp Ther ; 337(1): 2-8, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21205913

RESUMEN

Drug-induced cardiac arrhythmia, specifically Torsades de pointes, is associated with QT/QTc interval prolongation, thus prolongation of the QT interval is considered as a biomarker for Torsades de pointes risk (N Engl J Med 350:1013-1022, 2004). Specific inhibition of human ether-a-go-go-related gene (hERG) potassium channels has been recognized as the main mechanism for QT prolongation (Cardiovasc Res 58:32-45, 2003). This mechanism has been demonstrated for a variety of small-molecule agents, which access the inner pore of the hERG channel preferentially from inside the cell. Peptide inhibitors of hERG, such as BeKm-1, interact with the extracellular amino acid residues close to the external pore region of the channel. In this study, the isolated rabbit heart was used to assess whether BeKm-1 could induce QTc prolongation like dofetilide and N-[4-[[1-[2-(6-methyl-2-pyridinyl)ethyl]-4-piperidinyl]carbonyl]phenyl]methanesulfonamide (E-4031). Five hearts were perfused with 10 and 100 nM BeKm-1 sequentially. ECG parameters and left ventricular contractility were measured with spontaneously beating hearts. Both concentrations of BeKm-1 prolonged QTc intervals significantly and concentration-dependently (4.7 and 16.3% at 10 and 100 nM, respectively). When evaluated for their inhibitory effect in a hERG functional assay, BeKm-1, dofetilide, and E-4031 caused QTc prolongation at concentrations that caused significant hERG channel inhibition. Lastly, two polyclonal anti-hERG antibodies were also assessed in the hERG channel assay and found to be devoid of any inhibitory effect. These results indicated that the isolated rabbit heart assay can be used to measure QTc changes caused by specific hERG inhibition by peptides that specifically block the external pore region of the channel.


Asunto(s)
Canales de Potasio Éter-A-Go-Go/antagonistas & inhibidores , Corazón/efectos de los fármacos , Síndrome de QT Prolongado/inducido químicamente , Venenos de Escorpión/farmacología , Animales , Relación Dosis-Respuesta a Droga , Canal de Potasio ERG1 , Canales de Potasio Éter-A-Go-Go/fisiología , Femenino , Células HEK293 , Corazón/fisiología , Humanos , Técnicas In Vitro , Síndrome de QT Prolongado/fisiopatología , Péptidos/farmacología , Péptidos/toxicidad , Conejos , Venenos de Escorpión/toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...