Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Cardiovasc Pharmacol ; 69(3): 178-182, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28045761

RESUMEN

INTRODUCTION: We examined the effects of the selective late INa inhibitor eleclazine on the 50% probability of successful defibrillation (DFT50) before and after administration of amiodarone to determine its suitability for use in patients with implantable cardioverter defibrillators (ICDs). METHODS AND RESULTS: In 20 anesthetized pigs, transvenous active-fixation cardiac defibrillation leads were fluoroscopically positioned into right ventricular apex through jugular vein. ICDs were implanted subcutaneously. Dominant frequency of ventricular fibrillation was analyzed by fast Fourier transform. The measurements were made before drug administration (control), and at 40 minutes after vehicle, eleclazine (2 mg/kg, i.v., bolus over 15 minutes), or subsequent/single amiodarone administration (10 mg/kg, i.v., bolus over 10 minutes). Eleclazine did not alter DFT50, dominant frequency, heart rate, or mean arterial pressure (MAP). Subsequent amiodarone increased DFT50 (P = 0.006), decreased dominant frequency (P = 0.022), and reduced heart rate (P = 0.031) with no change in MAP. Amiodarone alone increased DFT50 (P = 0.005; NS compared to following eleclazine) and decreased dominant frequency (P = 0.003; NS compared to following eleclazine). CONCLUSION: Selective late INa inhibition with eleclazine does not alter DFT50 or dominant frequency of ventricular fibrillation when administered alone or in combination with amiodarone. Accordingly, eleclazine would not be anticipated to affect the margin of defibrillation safety in patients with ICDs.


Asunto(s)
Amiodarona/uso terapéutico , Cardioversión Eléctrica/métodos , Oxazepinas/uso terapéutico , Bloqueadores de los Canales de Sodio/uso terapéutico , Fibrilación Ventricular/tratamiento farmacológico , Fibrilación Ventricular/fisiopatología , Amiodarona/farmacología , Animales , Masculino , Oxazepinas/farmacología , Bloqueadores de los Canales de Sodio/farmacología , Porcinos , Fibrilación Ventricular/terapia , Canales de Sodio Activados por Voltaje/fisiología
2.
J Cardiovasc Electrophysiol ; 26(3): 329-35, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25346368

RESUMEN

INTRODUCTION: Ventricular rate during atrial fibrillation (AF) can be reduced by slowing atrioventricular (AV) node conduction and/or by decreasing dominant frequency of AF. We investigated whether combined administration of ivabradine and ranolazine reduces ventricular rate during AF. METHODS AND RESULTS: Ivabradine (maximum clinical dose, 0.25 mg/kg, and 0.10 mg/kg, i.v.) and ranolazine (2.4 mg/kg, i.v., bolus followed by 0.135 mg/kg/min) were studied in an anesthetized pig (N = 16) model of AF. Combined administration of 0.25 mg/kg ivabradine with ranolazine reduced ventricular rate during AF by 51.9 ± 9.7 beats/min (23%, P = 0.017) and dominant frequency of AF by 2.8 ± 0.5 Hz (32%, P = 0.005). It increased PR (P = 0.0002, P = 0.0007) and A-H intervals (P = 0.047, P = 0.002) during pacing at 130 and 180 beats/min, respectively, to a greater degree than additive effects of single agents. Combined administration of 0.1 mg/kg ivabradine with ranolazine exceeded additive effects of single agents on A-H intervals and dominant frequency of AF. Moreover, ranolazine potentiated low-dose ivabradine's reduction in ventricular rate, as combined administration more than doubled effects of the higher ivabradine dose alone and was similar to the combination with the higher dose. Neither drug nor their combination affected contractility (left ventricular [LV] dP/dt), QT or His-ventricular (H-V) intervals, or mean arterial pressure during sinus rhythm or AF. CONCLUSION: Combined administration of ivabradine and ranolazine at clinically safe levels decreases ventricular rate during AF by reducing AV node conduction and AF dominant frequency without QT prolongation or depression in contractility. Targeting these actions offers intrinsic advantages over conventional nodal agents, which can reduce contractility.


Asunto(s)
Fibrilación Atrial/tratamiento farmacológico , Benzazepinas/administración & dosificación , Fármacos Cardiovasculares/administración & dosificación , Frecuencia Cardíaca/efectos de los fármacos , Ranolazina/administración & dosificación , Animales , Fibrilación Atrial/fisiopatología , Quimioterapia Combinada , Cobayas , Frecuencia Cardíaca/fisiología , Ivabradina , Masculino , Porcinos
3.
Circ Res ; 110(6): 841-50, 2012 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-22343711

RESUMEN

RATIONALE: Previously, we demonstrated that a deoxycorticosterone acetate (DOCA)-salt hypertensive mouse model produces cardiac oxidative stress and diastolic dysfunction with preserved systolic function. Oxidative stress has been shown to increase late inward sodium current (I(Na)), reducing the net cytosolic Ca(2+) efflux. OBJECTIVE: Oxidative stress in the DOCA-salt model may increase late I(Na), resulting in diastolic dysfunction amenable to treatment with ranolazine. METHODS AND RESULTS: Echocardiography detected evidence of diastolic dysfunction in hypertensive mice that improved after treatment with ranolazine (E/E':sham, 31.9 ± 2.8, sham+ranolazine, 30.2 ± 1.9, DOCA-salt, 41.8 ± 2.6, and DOCA-salt+ranolazine, 31.9 ± 2.6; P=0.018). The end-diastolic pressure-volume relationship slope was elevated in DOCA-salt mice, improving to sham levels with treatment (sham, 0.16 ± 0.01 versus sham+ranolazine, 0.18 ± 0.01 versus DOCA-salt, 0.23 ± 0.2 versus DOCA-salt+ranolazine, 0.17 ± 0.0 1 mm Hg/L; P<0.005). DOCA-salt myocytes demonstrated impaired relaxation, τ, improving with ranolazine (DOCA-salt, 0.18 ± 0.02, DOCA-salt+ranolazine, 0.13 ± 0.01, sham, 0.11 ± 0.01, sham+ranolazine, 0.09 ± 0.02 seconds; P=0.0004). Neither late I(Na) nor the Ca(2+) transients were different from sham myocytes. Detergent extracted fiber bundles from DOCA-salt hearts demonstrated increased myofilament response to Ca(2+) with glutathionylation of myosin binding protein C. Treatment with ranolazine ameliorated the Ca(2+) response and cross-bridge kinetics. CONCLUSIONS: Diastolic dysfunction could be reversed by ranolazine, probably resulting from a direct effect on myofilaments, indicating that cardiac oxidative stress may mediate diastolic dysfunction through altering the contractile apparatus.


Asunto(s)
Acetanilidas/farmacología , Calcio/metabolismo , Diástole/efectos de los fármacos , Insuficiencia Cardíaca Diastólica/tratamiento farmacológico , Miocitos Cardíacos/efectos de los fármacos , Miofibrillas/efectos de los fármacos , Piperazinas/farmacología , Acetanilidas/sangre , Animales , Desoxicorticosterona/toxicidad , Diástole/fisiología , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/sangre , Inhibidores Enzimáticos/farmacología , Insuficiencia Cardíaca Diastólica/inducido químicamente , Insuficiencia Cardíaca Diastólica/fisiopatología , Ratones , Mineralocorticoides/toxicidad , Contracción Miocárdica/efectos de los fármacos , Contracción Miocárdica/fisiología , Miocitos Cardíacos/fisiología , Miofibrillas/metabolismo , Estrés Oxidativo/fisiología , Piperazinas/sangre , Ranolazina , Sodio/metabolismo , Disfunción Ventricular Izquierda/inducido químicamente , Disfunción Ventricular Izquierda/tratamiento farmacológico , Disfunción Ventricular Izquierda/fisiopatología
4.
Am J Respir Cell Mol Biol ; 49(6): 1038-47, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23855769

RESUMEN

Chronic obstructive pulmonary disease (COPD) is the fourth leading cause of death worldwide. The development of pulmonary hypertension (PH) in patients with COPD is strongly associated with increased mortality. Chronic inflammation and changes to the lung extracellular matrix (ECM) have been implicated in the pathogenesis of COPD, yet the mechanisms that lead to PH secondary to COPD remain unknown. Our experiments using human lung tissue show increased expression levels of the adenosine A2B receptor (ADORA2B) and a heightened deposition of hyaluronan (HA; a component of the ECM) in remodeled vessels of patients with PH associated with COPD. We also demonstrate that the expression of HA synthase 2 correlates with mean pulmonary arterial pressures in patients with COPD, with and without a secondary diagnosis of PH. Using an animal model of airspace enlargement and PH, we show that the blockade of ADORA2B is able to attenuate the development of a PH phenotype that correlates with reduced levels of HA deposition in the vessels and the down-regulation of genes involved in the synthesis of HA.


Asunto(s)
Ácido Hialurónico/metabolismo , Hipertensión Pulmonar/etiología , Hipertensión Pulmonar/fisiopatología , Enfermedad Pulmonar Obstructiva Crónica/complicaciones , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología , Receptor de Adenosina A2B/metabolismo , Antagonistas del Receptor de Adenosina A2/farmacología , Adenosina Desaminasa/deficiencia , Adenosina Desaminasa/genética , Anciano , Animales , Colágeno Tipo I/genética , Cadena alfa 1 del Colágeno Tipo I , Modelos Animales de Enfermedad , Femenino , Humanos , Hipertensión Pulmonar/patología , Pulmón/irrigación sanguínea , Pulmón/patología , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Enfermedad Pulmonar Obstructiva Crónica/patología , Purinas/farmacología , Pirazoles/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptor de Adenosina A2B/genética
5.
J Cardiovasc Electrophysiol ; 24(8): 914-8, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23647933

RESUMEN

INTRODUCTION: The mechanism(s) whereby dronedarone reduces sinus rate are not well understood, although L-type calcium channel antagonism, beta-adrenergic blockade, and inhibition of If are plausible. METHODS AND RESULTS: In anesthetized pigs, we compared the effects of dronedarone to the prototypical If inhibitor, ivabradine, and the L-type calcium channel antagonist diltiazem on heart rate, mean arterial blood pressure (MAP), and contractility. Dronedarone's effects on the phenylephrine-induced rise in MAP and on the chronotropic response to isoproterenol were also investigated. Cumulative doses of dronedarone (0.5 mg/kg, i.v., and 5.0 mg/kg, i.v.; plasma level: 80 ± 16.1 nM) progressively reduced heart rate (P < 0.02) without changes in MAP or contractility as assessed by LV dP/dt (N = 6). Ivabradine (0.5 mg/kg, i.v.) similarly reduced heart rate (P < 0.01) without change in MAP (N = 6). Diltiazem (0.8 mg/kg, i.v.) reduced heart rate and MAP and decreased contractility (N = 6). Dronedarone blunted phenylephrine's alpha-receptor-mediated increase in MAP but did not alter the marked beta-adrenergic receptor (BAR)-mediated increase in heart rate induced by isoproterenol. When dronedarone injection was preceded by ivabradine, no further decrease in heart rate or change in MAP was observed (N = 6). CONCLUSIONS: Dronedarone reduced heart rate without affecting MAP or contractility, effects that differ from L-type calcium channel blockade. Dronedarone did not antagonize BAR stimulation, and its heart-rate lowering effects were eliminated by prior administration of ivabradine. Thus, dronedarone's bradycardic action is likely due to inhibition of If and not to blockade of either L-type calcium channels or BAR.


Asunto(s)
Amiodarona/análogos & derivados , Bradicardia/inducido químicamente , Amiodarona/farmacología , Análisis de Varianza , Animales , Presión Arterial/efectos de los fármacos , Benzazepinas/farmacología , Presión Sanguínea/efectos de los fármacos , Diltiazem/farmacología , Dronedarona , Isoproterenol/farmacología , Ivabradina , Contracción Miocárdica/efectos de los fármacos , Porcinos
6.
FASEB J ; 26(6): 2546-57, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22415303

RESUMEN

Development of pulmonary hypertension is a common and deadly complication of interstitial lung disease. Little is known regarding the cellular and molecular mechanisms that lead to pulmonary hypertension in patients with interstitial lung disease, and effective treatment options are lacking. The purpose of this study was to examine the adenosine 2B receptor (A(2B)R) as a regulator of vascular remodeling and pulmonary hypertension secondary to pulmonary fibrosis. To accomplish this, cellular and molecular changes in vascular remodeling were monitored in mice exposed to bleomycin in conjunction with genetic removal of the A(2B)R or treatment with the A(2B)R antagonist GS-6201. Results demonstrated that GS-6201 treatment or genetic removal of the A(2B)R attenuated vascular remodeling and hypertension in our model. Furthermore, direct A(2B)R activation on vascular cells promoted interleukin-6 and endothelin-1 release. These studies identify a novel mechanism of disease progression to pulmonary hypertension and support the development of A(2B)R antagonists for the treatment of pulmonary hypertension secondary to interstitial lung disease.


Asunto(s)
Hipertensión Pulmonar/etiología , Enfermedades Pulmonares Intersticiales/complicaciones , Receptor de Adenosina A2B/fisiología , Adenosina-5'-(N-etilcarboxamida)/farmacología , Animales , Bleomicina , Células Cultivadas , Endotelina-1/metabolismo , Endotelio Vascular/citología , Humanos , Interleucina-6/biosíntesis , Masculino , Ratones , Ratones Endogámicos C57BL , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/complicaciones , Agonistas del Receptor Purinérgico P1/farmacología , Purinas/farmacología , Pirazoles/farmacología
7.
Europace ; 15(3): 429-36, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23258816

RESUMEN

AIMS: Clinical utility of QTc prolongation as a predictor for sudden cardiac death (SCD) has not been definitely established. Ranolazine causes modest QTc prolongation, yet it shows antiarrhythmic properties. We aimed to determine the association between prolonged QTc and risk of SCD, and the effect of ranolazine on this relationship. METHODS AND RESULTS: The relationship between baseline QTc and SCD was studied in 6492 patients with non-ST elevation acute coronary syndrome (NSTEACS) randomized to placebo or ranolazine in the MERLIN-TIMI 36 trial. In the placebo group, an abnormal QTc interval (≥450 ms in men, ≥470 ms in women) was associated with a two-fold increased risk of SCD (hazard ratio, HR, 2.3, P = 0.005) after adjustment for other risk factors (age ≥75 years, NYHA class III/IV, high TIMI risk score, ventricular tachycardia ≥8 beats, digitalis, and antiarrhythmics). In the ranolazine group, the association between abnormal QTc and SCD was similar to placebo, but not significant (HR 1.8, P = 0.074). There was no significant difference between placebo and ranolazine in the risk for SCD in patients with abnormal QTc (HR 0.78, P = 0.48). When QTc was used as a continuous variable, for every 10 ms increase in QTc, hazard rate for SCD increased significantly by 8% (P = 0.007) in the placebo group, and only by 2.9% (P = 0.412; P for interaction=0.25) in the ranolazine group. CONCLUSION: In NSTEACS patients treated with placebo, prolonged QTc was a significant independent predictor for SCD. Ranolazine, compared with placebo, was not associated with increased risk for SCD in patients with prolonged QTc.


Asunto(s)
Acetanilidas/uso terapéutico , Síndrome Coronario Agudo/tratamiento farmacológico , Fármacos Cardiovasculares/uso terapéutico , Muerte Súbita Cardíaca/etiología , Síndrome de QT Prolongado/etiología , Piperazinas/uso terapéutico , Acetanilidas/efectos adversos , Síndrome Coronario Agudo/complicaciones , Síndrome Coronario Agudo/diagnóstico , Síndrome Coronario Agudo/fisiopatología , Anciano , Fármacos Cardiovasculares/efectos adversos , Electrocardiografía , Femenino , Humanos , Incidencia , Estimación de Kaplan-Meier , Síndrome de QT Prolongado/inducido químicamente , Síndrome de QT Prolongado/diagnóstico , Síndrome de QT Prolongado/fisiopatología , Masculino , Análisis Multivariante , Piperazinas/efectos adversos , Modelos de Riesgos Proporcionales , Ensayos Clínicos Controlados Aleatorios como Asunto , Ranolazina , Estudios Retrospectivos , Medición de Riesgo , Factores de Riesgo , Factores de Tiempo , Resultado del Tratamiento
8.
J Pharmacol Exp Ther ; 343(3): 587-95, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22923737

RESUMEN

Adenosine (Ado) is released in response to tissue injury, promotes hyperemia, and modulates inflammation. The proinflammatory effects of Ado, which are mediated by the A(2B) Ado receptor (AdoR), may exacerbate tissue damage. We hypothesized that selective blockade of the A(2B) AdoR with 3-ethyl-1-propyl-8-(1-(3-trifluoromethylbenzyl)-1H-pyrazol-4-yl)-3,7-dihydropurine-2,6-dione (GS-6201) during acute myocardial infarction (AMI) would reduce adverse cardiac remodeling. Male ICR mice underwent coronary artery ligation or sham surgery (n = 10-12 per group). The selective A(2B) AdoR antagonist GS-6201 (4 mg/kg) was given intraperitoneally twice daily starting immediately after surgery and continuing for 14 days. Transthoracic echocardiography was performed before surgery and after 7, 14, and 28 days. A subgroup of mice was killed 72 h after surgery, and the activity of caspase-1, a key proinflammatory mediator, was measured in the cardiac tissue. All sham-operated mice were alive at 4 weeks, whereas 50% of vehicle-treated mice and 75% of GS-6201-treated mice were alive at 4 weeks after surgery. Compared with vehicle, treatment with GS-6201 prevented caspase-1 activation in the heart at 72 h after AMI (P < 0.001) and significantly limited the increase in left ventricular (LV) end-diastolic diameter by 40% (P < 0.001), the decrease in LV ejection fraction by 18% (P < 0.01) and the changes in the myocardial performance index by 88% (P < 0.001) at 28 days after AMI. Selective blockade of A(2B) AdoR with GS-6201 reduces caspase-1 activity in the heart and leads to a more favorable cardiac remodeling after AMI in the mouse.


Asunto(s)
Antagonistas del Receptor de Adenosina A2/uso terapéutico , Infarto del Miocardio/tratamiento farmacológico , Purinas/uso terapéutico , Pirazoles/uso terapéutico , Receptor de Adenosina A2B/metabolismo , Remodelación Ventricular/efectos de los fármacos , Antagonistas del Receptor de Adenosina A2/administración & dosificación , Animales , Caspasa 1/metabolismo , Modelos Animales de Enfermedad , Esquema de Medicación , Ecocardiografía , Activación Enzimática , Hemodinámica/efectos de los fármacos , Inyecciones Intraperitoneales , Masculino , Ratones , Ratones Endogámicos ICR , Infarto del Miocardio/diagnóstico por imagen , Infarto del Miocardio/enzimología , Infarto del Miocardio/metabolismo , Purinas/administración & dosificación , Pirazoles/administración & dosificación , Resultado del Tratamiento
9.
J Cardiovasc Pharmacol ; 57(6): 639-47, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21633249

RESUMEN

Effects of ranolazine alone and in the presence of phenylephrine (PE) or isoproterenol (ISO) on hemodynamics, coronary blood flow and heart rate (HR) in the absence and presence of hexamethonium (a ganglionic blocker) were studied in conscious dogs. Ranolazine (0.4, 1.2, 3.6, and 6 mg/kg, intravenous) alone caused transient (<1 minute) and reversible hemodynamic changes. PE (0.3-10 µg/kg) caused a dose-dependent increase in blood pressure and decrease in HR. ISO (0.01-0.3 µg/kg) caused a dose-dependent decrease in blood pressure and an increase in HR. Ranolazine at high (11-13 mM), but not at moderate (4-5 mM) concentrations partially attenuated changes in mean arterial blood pressure and HR caused by either PE or ISO in normal conscious dogs. However, in dogs treated with hexamethonium (20 mg/kg) to cause autonomic blockade, ranolazine (both 4-5 and 11-13 µM) significantly attenuated both the PE- and ISO-induced changes in mean arterial blood pressure. The results suggest that a potential antiadrenergic effect of ranolazine was masked by autonomic control mechanisms in conscious dogs but could be observed when these mechanisms were inhibited (eg, in the hexamethonium-treated dog). Ranolazine, at plasma concentrations <10 µM and in conscious dogs with intact autonomic regulation, had minimal antiadrenergic (α and ß) effects.


Asunto(s)
Acetanilidas/uso terapéutico , Antagonistas Adrenérgicos/uso terapéutico , Hemodinámica/efectos de los fármacos , Hipertensión/tratamiento farmacológico , Hipotensión/tratamiento farmacológico , Piperazinas/uso terapéutico , Receptores Adrenérgicos alfa/metabolismo , Receptores Adrenérgicos beta/metabolismo , Acetanilidas/administración & dosificación , Acetanilidas/sangre , Acetanilidas/metabolismo , Antagonistas Adrenérgicos/administración & dosificación , Antagonistas Adrenérgicos/sangre , Antagonistas Adrenérgicos/metabolismo , Animales , Fármacos del Sistema Nervioso Autónomo/administración & dosificación , Fármacos del Sistema Nervioso Autónomo/uso terapéutico , Circulación Coronaria/efectos de los fármacos , Perros , Relación Dosis-Respuesta a Droga , Bloqueadores Ganglionares/farmacología , Cobayas , Frecuencia Cardíaca/efectos de los fármacos , Hexametonio/farmacología , Hipertensión/inducido químicamente , Hipotensión/inducido químicamente , Isoproterenol/administración & dosificación , Isoproterenol/toxicidad , Cinética , Fenilefrina/administración & dosificación , Fenilefrina/toxicidad , Piperazinas/administración & dosificación , Piperazinas/sangre , Piperazinas/metabolismo , Ranolazina , Ratas , Receptores Adrenérgicos alfa/química , Receptores Adrenérgicos beta/química , Vasoconstrictores/administración & dosificación , Vasoconstrictores/toxicidad , Vasodilatadores/administración & dosificación , Vasodilatadores/toxicidad
10.
J Nucl Cardiol ; 18(4): 620-7, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21553161

RESUMEN

BACKGROUND: There are limited data on the effect of body mass index (BMI) on the actions of fixed-dose regadenoson. The purpose of this study was to determine the effect of BMI on the efficacy, side effects, and plasma concentration of regadenoson for Myocardial Perfusion Imaging (MPI). METHODS AND RESULTS: The study included 2,015 subjects from the ADVANCE MPI trials. Initial adenosine MPI was followed by randomization to regadenoson (400-µg bolus injection) or adenosine (6-minute infusion) MPI. Subjects were classified according to BMI into six categories from underweight (<20 kg/m(2)) to extremely obese (≥40 kg/m(2)). PK modeling was used to predict the effect of BMI on plasma regadenoson concentration (PRC). Adenosine-regadenoson agreement rates for the presence and extent of reversibility were similar across BMI categories (P > .05). The incidence of side effects was also similar across BMIs (P ≥ .06). Subjects were less likely to feel very or extremely uncomfortable after regadenoson vs adenosine in all groups with BMI ≥ 25 kg/m(2), but this trend was not statistically significant in subjects with BMI 20-24 kg/m(2) (P > .05). PRC was inversely related to BMI with 19% higher PRC in the underweight and 36% lower PRC in the extremely obese compared with a normal weight subject. CONCLUSIONS: BMI does not alter the efficacy of regadenoson MPI despite lower PRC in high BMI subjects, or its side effect profile despite higher PRC in low BMI subjects. Regadenoson is better tolerated than adenosine but this benefit seems to lose statistical significance in subjects with BMI < 25 kg/m(2).


Asunto(s)
Agonistas del Receptor de Adenosina A2/sangre , Índice de Masa Corporal , Imagen de Perfusión Miocárdica , Purinas/sangre , Pirazoles/sangre , Anciano , Método Doble Ciego , Femenino , Humanos , Masculino , Persona de Mediana Edad , Purinas/efectos adversos , Pirazoles/efectos adversos
11.
J Clin Oncol ; 38(19): 2140-2150, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32182156

RESUMEN

PURPOSE: Lifirafenib is an investigational, reversible inhibitor of B-RAFV600E, wild-type A-RAF, B-RAF, C-RAF, and EGFR. This first-in-human, phase I, dose-escalation/dose-expansion study evaluated the safety, tolerability, and efficacy of lifirafenib in patients with B-RAF- or K-RAS/N-RAS-mutated solid tumors. METHODS: During dose escalation, adult patients with histologically/cytologically confirmed advanced solid tumors received escalating doses of lifirafenib. Primary end points were safety/tolerability during dose escalation and objective response rate in preselected patients with B-RAF and K-RAS/N-RAS mutations during dose expansion. RESULTS: The maximum tolerated dose was established as 40 mg/d; dose-limiting toxicities included reversible thrombocytopenia and nonhematologic toxicity. Across the entire study, the most common grade ≥ 3 treatment-emergent adverse events were hypertension (n = 23; 17.6%) and fatigue (n = 13; 9.9%). One patient with B-RAF-mutated melanoma achieved complete response, and 8 patients with B-RAF mutations had confirmed objective responses: B-RAFV600E/K melanoma (n = 5, including 1 patient treated with prior B-RAF/MEK inhibitor therapy), B-RAFV600E thyroid cancer/papillary thyroid cancer (PTC; n = 2), and B-RAFV600E low-grade serous ovarian cancer (LGSOC; n = 1). One patient with B-RAF-mutated non-small-cell lung cancer (NSCLC) had unconfirmed partial response (PR). Patients with K-RAS-mutated endometrial cancer and K-RAS codon 12-mutated NSCLC had confirmed PR (n = 1 each). No responses were seen in patients with K-RAS/N-RAS-mutated colorectal cancer (n = 20). CONCLUSION: Lifirafenib is a novel inhibitor of key RAF family kinases and EGFR, with an acceptable risk-benefit profile and antitumor activity in patients with B-RAFV600-mutated solid tumors, including melanoma, PTC, and LGSOC, as well as K-RAS-mutated NSCLC and endometrial carcinoma. Future comparisons with first-generation B-RAF inhibitors and exploration of lifirafenib alone or as combination therapy in patients with selected RAS mutations who are resistant/refractory to first-generation B-RAF inhibitors are warranted.


Asunto(s)
Bencimidazoles/uso terapéutico , Naftiridinas/uso terapéutico , Neoplasias/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/uso terapéutico , Adulto , Anciano , Bencimidazoles/farmacología , Femenino , Humanos , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Naftiridinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología
12.
J Clin Invest ; 116(8): 2173-2182, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16841096

RESUMEN

Adenosine has been implicated in the pathogenesis of chronic lung diseases such as asthma and chronic obstructive pulmonary disease. In vitro studies suggest that activation of the A2B adenosine receptor (A2BAR) results in proinflammatory and profibrotic effects relevant to the progression of lung diseases; however, in vivo data supporting these observations are lacking. Adenosine deaminase-deficient (ADA-deficient) mice develop pulmonary inflammation and injury that are dependent on increased lung adenosine levels. To investigate the role of the A2BAR in vivo, ADA-deficient mice were treated with the selective A2BAR antagonist CVT-6883, and pulmonary inflammation, fibrosis, and airspace integrity were assessed. Untreated and vehicle-treated ADA-deficient mice developed pulmonary inflammation, fibrosis, and enlargement of alveolar airspaces; conversely, CVT-6883-treated ADA-deficient mice showed less pulmonary inflammation, fibrosis, and alveolar airspace enlargement. A2BAR antagonism significantly reduced elevations in proinflammatory cytokines and chemokines as well as mediators of fibrosis and airway destruction. In addition, treatment with CVT-6883 attenuated pulmonary inflammation and fibrosis in wild-type mice subjected to bleomycin-induced lung injury. These findings suggest that A2BAR signaling influences pathways critical for pulmonary inflammation and injury in vivo. Thus in chronic lung diseases associated with increased adenosine, antagonism of A2BAR-mediated responses may prove to be a beneficial therapy.


Asunto(s)
Adenosina/efectos adversos , Enfermedades Pulmonares/fisiopatología , Lesión Pulmonar , Receptor de Adenosina A2B/fisiología , Animales , Inflamación/inducido químicamente , Inflamación/patología , Pulmón/efectos de los fármacos , Pulmón/patología , Pulmón/fisiopatología , Enfermedades Pulmonares/inducido químicamente , Ratones , Ratones Noqueados , Purinas/farmacología , Pirazoles/farmacología , Receptor de Adenosina A2B/deficiencia , Receptor de Adenosina A2B/genética , Transducción de Señal , Transcripción Genética
13.
J Med Chem ; 51(7): 2267-78, 2008 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-18321039

RESUMEN

Recently, we have reported a series of new 1,3-symmetrically (R 1 = R 3) substituted xanthines ( 3 and 4) which have high affinity and selectivity for the human adenosine A 2B receptors (hA(2B)-AdoR). Unfortunately, this class of compounds had poor pharmacokinetic properties. This prompted us to investigate the effect of differential alkyl substitution at the N-1 and N-3 positions ( N 1-R not equal to N 3-R) on A(2B)-AdoR affinity and selectivity; we had the dual objectives of enhancing affinity and selectivity for the A(2B)-AdoR, as well as improving oral bioavailability. This effort has led to the discovery of compound 62, that displayed high affinity and selectivity for the hA(2B)-AdoR (K(i) = 22 nM). In addition, compound 62 showed high functional potency in inhibiting the accumulation of cyclic adenosine monophosphate induced by 5'- N-ethylcarboxamidoadenosine in HEK-A(2B)-AdoR and NIH3T3 cells with K(B) values of 6 and 2 nM, respectively. In a single ascending-dose phase I clinical study, compound 62 had no serious adverse events and was well tolerated.


Asunto(s)
Antagonistas del Receptor de Adenosina A2 , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Xantinas/síntesis química , Xantinas/farmacología , Animales , Sitios de Unión , Línea Celular , AMP Cíclico/análisis , Relación Dosis-Respuesta a Droga , Diseño de Fármacos , Humanos , Ratones , Estructura Molecular , Células 3T3 NIH , Ratas , Estereoisomerismo , Relación Estructura-Actividad , Xantinas/química
14.
Bioorg Med Chem Lett ; 18(4): 1397-401, 2008 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-18226896

RESUMEN

A series of N-1 monosubstituted 8-pyrazolyl xanthines have been synthesized and evaluated for their affinity for the adenosine receptors (AdoRs). We have discovered two compounds 18 (CVT-7124) and 28 (CVT-6694) that display good affinity for the A(2B) AdoR (K(i)=6 nM and 7 nM, respectively) and greater selectivity for the human A(1), A(2A), and A(3) AdoRs (>1000-, >830-, and >1500-fold; >850-, >700-, and >1280-fold, respectively). CVT-6694 has been shown to block the release of interleukin-6 and monocyte chemotactic protein-1 from bronchial smooth muscle cells (BSMC), a process believed to be promoted by activation of A(2B) AdoR.


Asunto(s)
Antagonistas del Receptor de Adenosina A2 , Pirazoles/farmacología , Xantinas/farmacología , Animales , Unión Competitiva , Células CHO , Cricetinae , Cricetulus , Humanos , Cinética , Pirazoles/síntesis química , Pirazoles/química , Receptor de Adenosina A2B/metabolismo , Especificidad por Sustrato , Uracilo/análogos & derivados , Uracilo/química , Xantinas/síntesis química , Xantinas/química
15.
J Med Chem ; 49(12): 3682-92, 2006 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-16759111

RESUMEN

Adenosine has been suggested to induce bronchial hyperresponsiveness in asthmatics, which is believed to be an A(2B) adenosine receptor (AdoR) mediated pathway. We hypothesize that a selective, high-affinity A(2B) AdoR antagonist may provide therapeutic benefit in the treatment of asthma. In an attempt to identify a high-affinity, selective antagonist for the A(2B) AdoR, we synthesized 8-(C-4-pyrazolyl) xanthines. Compound 22, 8-(1H-pyrazol-4-yl)-1,3-dipropyl xanthine, is a N-1 unsubstituted pyrazole derivative that has favorable binding affinity (K(i) = 9 nM) for the A(2B) AdoR, but it is only 2-fold selective versus the A(1) AdoR. Introduction of a benzyl group at the N-1-pyrazole position of 22 resulted in 19, which had moderate selectivity. The initial focus of the SAR study was on the preparation of substituted benzyl derivatives of 19 because the corresponding phenyl, phenethyl, and phenpropyl derivatives showed a decrease in A(2B) AdoR affinity and selectivity relative to 19. The preferred substitution on the phenyl ring of 19 contains an electron-withdrawing group, specifically F or CF(3) at the m-position, as in 33 and 36 respectively, increases the selectivity while retaining the affinity for the A(2B) AdoR. Exploring disubstitutions on the phenyl ring of derivatives 33 and36 led to the 2-chloro-5-trifluoromethylphenyl derivative 50, which retained the A(2B) AdoR affinity but enhanced the selectivity relative to 36. After optimization of the substitution on the 8-pyrazole xanthine, 1,3-disubstitution of the xanthine core was explored with methyl, ethyl, butyl, and isobutyl groups. In comparison to the corresponding dipropyl analogues, the smaller 1,3-dialkyl groups (methyl and ethyl) increased the A(2B) AdoR binding selectivity of the xanthine derivatives while retaining the affinity. However, the larger 1,3-dialkyl groups (isobutyl and butyl) resulted in a decrease in both A(2B) AdoR affinity and selectivity. This final SAR optimization led to the discovery of 1,3-dimethyl derivative 60, 8-(1-(3-(trifluoromethyl) benzyl)-1H-pyrazol-4-yl)-1,3-dimethyl xanthine, a high-affinity (K(i) = 1 nM) A(2B) AdoR antagonist with high selectivity (990-, 690-, and 1,000-) for the human A(1), A(2A,) and A(3) AdoRs.


Asunto(s)
Antagonistas del Receptor de Adenosina A2 , Pirazoles/síntesis química , Teofilina/análogos & derivados , Xantinas/síntesis química , Animales , Línea Celular , Cricetinae , Cricetulus , Humanos , Pirazoles/química , Pirazoles/farmacología , Ensayo de Unión Radioligante , Teofilina/síntesis química , Teofilina/química , Teofilina/farmacología , Xantinas/química , Xantinas/farmacología
16.
Circ Res ; 90(5): 531-8, 2002 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-11909816

RESUMEN

Adenosine has been reported to stimulate or inhibit the release of angiogenic factors depending on the cell type examined. To test the hypothesis that differential expression of adenosine receptor subtypes contributes to endothelial cell heterogeneity, we studied microvascular (HMEC-1) and umbilical vein (HUVEC) human endothelial cells. Based on mRNA level and stimulation of adenylate cyclase, we found that HUVECs preferentially express A2A adenosine receptors and HMEC-1 preferentially express A2B receptors. Neither cells expressed A1 or A3 receptors. The nonselective adenosine agonist 5'-N-ethylcarboxamidoadenosine (NECA) increased expression of interleukin-8 (IL-8), basic fibroblast growth factor (bFGF), and vascular endothelial growth factor (VEGF) in HMEC-1, but had no effect in HUVECs. In contrast, the selective A2A agonist 2-p-(2-carboxyethyl)phenylethylamino-NECA (CGS 21680) had no effect on expression of these angiogenic factors. Cotransfection of each type of adenosine receptors with a luciferase reporter in HMEC-1 showed that A2B receptors, but not A1, A2A, or A3, activated IL-8 and VEGF promoters. These effects were mimicked by constitutively active alphaG(q), alphaG12, and alphaG13, but not alphaG(s) or alphaG(i1-3). Furthermore, stimulation of phospholipase C indicated coupling of A2B receptors to G(q) proteins in HMEC-1. Thus, differential expression of adenosine receptor subtypes contributes to functional heterogeneity of human endothelial cells. A2B receptors, predominantly expressed in human microvascular cells, modulate expression of angiogenic factors via coupling to G(q), and possibly via G12/13.


Asunto(s)
Inductores de la Angiogénesis/metabolismo , Endotelio Vascular/metabolismo , Receptores Purinérgicos P1/biosíntesis , Adenilil Ciclasas/metabolismo , Animales , Células CHO , Cricetinae , Factores de Crecimiento Endotelial/genética , Endotelio Vascular/citología , Genes Reporteros , Proteínas de Unión al GTP Heterotriméricas/genética , Proteínas de Unión al GTP Heterotriméricas/metabolismo , Humanos , Fosfatos de Inositol/metabolismo , Interleucina-8/genética , Luciferasas/genética , Linfocinas/genética , Microcirculación/metabolismo , Neovascularización Fisiológica/fisiología , Regiones Promotoras Genéticas/fisiología , Agonistas del Receptor Purinérgico P1 , ARN Mensajero/metabolismo , Receptor de Adenosina A2A , Receptor de Adenosina A2B , Receptores Purinérgicos P1/genética , Sistemas de Mensajero Secundario/fisiología , Transfección , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular , Venas/metabolismo
17.
Circ Arrhythm Electrophysiol ; 8(5): 1048-56, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26226999

RESUMEN

BACKGROUND: Atrial fibrillation (AF) requires arrhythmogenic changes in atrial ion channels/receptors and usually altered atrial structure. AF is commonly treated with antiarrhythmic drugs; the most effective block many ion channels/receptors. Modest efficacy, intolerance, and safety concerns limit current antiarrhythmic drugs. We hypothesized that combining agents with multiple anti-AF mechanisms at reduced individual drug doses might produce synergistic efficacy plus better tolerance/safety. METHODS AND RESULTS: HARMONY tested midrange ranolazine (750 mg BID) combined with 2 reduced dronedarone doses (150 mg BID and 225 mg BID; chosen to reduce dronedarone's negative inotropic effect-see text below) over 12 weeks in 134 patients with paroxysmal AF and implanted pacemakers where AF burden (AFB) could be continuously assessed. Patients were randomized double-blind to placebo, ranolazine alone (750 mg BID), dronedarone alone (225 mg BID), or one of the combinations. Neither placebo nor either drugs alone significantly reduced AFB. Conversely, ranolazine 750 mg BID/dronedarone 225 mg BID reduced AFB by 59% versus placebo (P=0.008), whereas ranolazine 750 mg BID/dronedarone 150 mg BID reduced AFB by 43% (P=0.072). Both combinations were well tolerated. CONCLUSIONS: HARMONY showed synergistic AFB reduction by moderate dose ranolazine plus reduced dose dronedarone, with good tolerance/safety, in the population enrolled. CLINICAL TRIAL REGISTRATION: ClinicalTrials.gov; Unique identifier: NCT01522651.


Asunto(s)
Amiodarona/análogos & derivados , Antiarrítmicos/uso terapéutico , Fibrilación Atrial/tratamiento farmacológico , Ranolazina/uso terapéutico , Bloqueadores de los Canales de Sodio/uso terapéutico , Anciano , Amiodarona/administración & dosificación , Amiodarona/uso terapéutico , Antiarrítmicos/administración & dosificación , Método Doble Ciego , Dronedarona , Sinergismo Farmacológico , Quimioterapia Combinada , Femenino , Humanos , Masculino , Marcapaso Artificial , Ranolazina/administración & dosificación , Bloqueadores de los Canales de Sodio/administración & dosificación , Resultado del Tratamiento
18.
J Med Chem ; 47(19): 4766-73, 2004 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-15341491

RESUMEN

We describe the synthesis of new high affinity and selective A(3)-adenosine receptor (A(3)-AdoR) agonists. Introduction of a methyl group at the N(6)-position of the A(2A)-AdoR selective 2-pyrazolyl-adenosine analogues (Figure 2) brought about a substantial increase in the A(3)-AdoR binding affinity and selectivity. While the N(6)-desmethyl analogues 3a and 4 were inactive at the A(3)-AdoR (K(i) > 10 microM), the corresponding N(6)-methyl analogues 5 and 22 showed good binding affinity at the A(3)-AdoR (K(i) = 73 and 97 nM, respectively). Replacement of the carboxamide group in 5 with different heteroaryl groups resulted in analogues with high affinities and selectivity for the A(3)-AdoR. (2R,3S,4R)-tetrahydro-2-(hydroxymethyl)-5-(6-(methylamino)-2-(4-(pyridin-2-yl)-1H-pyrazol-1-yl)-9H-purin-9-yl)furan-3,4-diol (15, K(i) = 2 nM) displayed high selectivity for the A(3)-AdoR versus A(1)- and A(2A)-AdoRs (selectivity ratios of 1900 and >2000, respectively).


Asunto(s)
Agonistas del Receptor de Adenosina A3 , Adenosina/análogos & derivados , Adenosina/química , Adenosina/farmacología , Adenosina/síntesis química , Animales , Cricetinae , Humanos , Estructura Molecular , Receptor de Adenosina A3/metabolismo , Relación Estructura-Actividad
19.
Heart Rhythm ; 11(9): 1592-9, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25066042

RESUMEN

BACKGROUND: Ranolazine has been shown to have antiarrhythmic properties. OBJECTIVE: We tested the hypothesis that intravenous ranolazine would terminate induced atrial flutter (AFL) or atrial fibrillation (AF) in the canine sterile pericarditis model. METHODS: In 6 dogs with sterile pericarditis, we performed electrophysiological measurements of the atrial effective refractory period (AERP) and conduction time (CT) while pacing from the right atrial appendage, Bachmann bundle, and the posteroinferior left atrium at cycle lengths (CLs) of 400, 300, and 200 ms before and after the administration of ranolazine. In 13 induced episodes of AFL (n = 9) and AF (n = 4), ranolazine was administered intravenously as a 3.2 mg/kg bolus, followed by a maintenance infusion of 0.17 mg/(kg·min). Six episodes (4 AFL and 2 AF) were induced in the open-chest state to perform simultaneous, multisite (486 electrodes), epicardial mapping of the arrhythmia and its termination. RESULTS: Ranolazine terminated 7 of 9 AFL and 3 of 4 AF episodes. Ranolazine significantly prolonged the AFL CL by a mean of 43 ms (P < .001) and the AF CL by a mean of 34 ms (P < .01). The AERP was prolonged (P < .05 overall), and the atrial capture threshold increased minimally (P < .01 for all). Ranolazine prolonged CTs (P < .01 overall). During open-chest, multisite mapping, block in the region of slow conduction in the reentrant circuit terminated AFL and interruption of the regular driver terminated AF. CONCLUSION: Ranolazine terminated AFL/AF in our canine sterile pericarditis model by interrupting the regular driver. Ranolazine was found to significantly prolong the AERP, CT, and tachycardia CLs.


Asunto(s)
Acetanilidas/administración & dosificación , Fibrilación Atrial/tratamiento farmacológico , Aleteo Atrial/tratamiento farmacológico , Electrocardiografía , Sistema de Conducción Cardíaco/anomalías , Frecuencia Cardíaca/efectos de los fármacos , Piperazinas/administración & dosificación , Animales , Arritmias Cardíacas , Fibrilación Atrial/fisiopatología , Aleteo Atrial/fisiopatología , Síndrome de Brugada , Trastorno del Sistema de Conducción Cardíaco , Modelos Animales de Enfermedad , Perros , Inhibidores Enzimáticos/administración & dosificación , Sistema de Conducción Cardíaco/efectos de los fármacos , Inyecciones Intravenosas , Ranolazina , Bloqueadores de los Canales de Sodio , Resultado del Tratamiento
20.
Heart Rhythm ; 11(1): 101-9, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24120874

RESUMEN

BACKGROUND: Remodeling occurs after myocardial infarction (MI), leading to fibrosis, dysfunction, and ventricular tachycardias (VTs). Adenosine via the A2B adenosine receptor (A2BAdoR) has been implicated in promoting fibrosis. OBJECTIVE: To determine the effects of GS-6201, a potent antagonist of the A2BAdoR, on arrhythmogenic and functional cardiac remodeling after MI. METHODS: Rats underwent ischemia-reperfusion MI and were randomized into 4 groups: control (treated with vehicle), angiotensin-converting enzyme inhibitor (treated with enalapril 1 day after MI), GS-6201-1d (treated with GS-6201 1 day after MI), GS-6201-1w (treated with GS-6201 administered 1 week after MI) . Echocardiography was performed at baseline and 1 and 5 weeks after MI. Optical mapping, VT inducibility, and histologic analysis were conducted at follow-up. RESULTS: Treatment with the angiotensin-converting enzyme inhibitor improved ejection fraction (57.8% ± 2.5% vs 43.3% ± 1.7% in control; P < .01), but had no effect on VT inducibility. Treatment with GS-6201 improved ejection fraction (55.6% ± 2.6% vs 43.3% ± 1.7% in control; P < .01) and decreased VT inducibility (9.1% vs 68.4% in control; P < .05). Conduction velocities were significantly higher at border and infarct zones in hearts of rats treated with GS-6201 than in those of other groups. The conduction heterogeneity index was also significantly lower in hearts of rats treated with GS-6201. Histologic analysis showed that while both GS-6201 and enalapril decreased fibrosis in the noninfarct zone, only GS-6201 reduced the heterogeneity of fibrosis at the border, which is consistent with its effect on VT reduction. CONCLUSIONS: Treatment with an A2BAdoR antagonist at 1 week results in the improvement in cardiac function and decreased substrate for VT. The inhibition of fibrogenesis by A2BAdoR antagonists may be a new target for the prevention of adverse remodeling after MI.


Asunto(s)
Antagonistas del Receptor de Adenosina A2/uso terapéutico , Electrocardiografía , Infarto del Miocardio/complicaciones , Purinas/uso terapéutico , Pirazoles/uso terapéutico , Taquicardia Ventricular/prevención & control , Disfunción Ventricular Izquierda/prevención & control , Remodelación Ventricular/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Estudios de Seguimiento , Sistema de Conducción Cardíaco/efectos de los fármacos , Sistema de Conducción Cardíaco/fisiopatología , Masculino , Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/fisiopatología , Ratas , Ratas Sprague-Dawley , Volumen Sistólico/efectos de los fármacos , Taquicardia Ventricular/etiología , Taquicardia Ventricular/fisiopatología , Factores de Tiempo , Resultado del Tratamiento , Disfunción Ventricular Izquierda/etiología , Disfunción Ventricular Izquierda/fisiopatología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA