Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 579
Filtrar
1.
Int J Mol Sci ; 25(8)2024 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-38673903

RESUMEN

Cardiotonic steroids (CTSs), such as digoxin, are used for heart failure treatment. However, digoxin permeates the brain-blood barrier (BBB), affecting central nervous system (CNS) functions. Finding a CTS that does not pass through the BBB would increase CTSs' applicability in the clinic and decrease the risk of side effects on the CNS. This study aimed to investigate the tissue distribution of the CTS ouabain following intraperitoneal injection and whether ouabain passes through the BBB. After intraperitoneal injection (1.25 mg/kg), ouabain concentrations were measured at 5 min, 15 min, 30 min, 1 h, 3 h, 6 h, and 24 h using HPLC-MS in brain, heart, liver, and kidney tissues and blood plasma in C57/black mice. Ouabain was undetectable in the brain tissue. Plasma: Cmax = 882.88 ± 21.82 ng/g; Tmax = 0.08 ± 0.01 h; T1/2 = 0.15 ± 0.02 h; MRT = 0.26 ± 0.01. Cardiac tissue: Cmax = 145.24 ± 44.03 ng/g (undetectable at 60 min); Tmax = 0.08 ± 0.02 h; T1/2 = 0.23 ± 0.09 h; MRT = 0.38 ± 0.14 h. Kidney tissue: Cmax = 1072.3 ± 260.8 ng/g; Tmax = 0.35 ± 0.19 h; T1/2 = 1.32 ± 0.76 h; MRT = 1.41 ± 0.71 h. Liver tissue: Cmax = 2558.0 ± 382.4 ng/g; Tmax = 0.35 ± 0.13 h; T1/2 = 1.24 ± 0.7 h; MRT = 0.98 ± 0.33 h. Unlike digoxin, ouabain does not cross the BBB and is eliminated quicker from all the analyzed tissues, giving it a potential advantage over digoxin in systemic administration. However, the inability of ouabain to pass though the BBB necessitates intracerebral administration when used to investigate its effects on the CNS.


Asunto(s)
Ratones Endogámicos C57BL , Ouabaína , Animales , Distribución Tisular , Inyecciones Intraperitoneales , Ratones , Masculino , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/efectos de los fármacos , Espectrometría de Masas/métodos , Riñón/metabolismo , Riñón/efectos de los fármacos , Hígado/metabolismo , Hígado/efectos de los fármacos , Cromatografía Líquida de Alta Presión/métodos , Miocardio/metabolismo , Cardiotónicos/farmacocinética , Cardiotónicos/farmacología , Cardiotónicos/administración & dosificación
2.
Cancer Chemother Pharmacol ; 88(6): 941-952, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34477937

RESUMEN

INTRODUCTION: Fedratinib, an oral, selective Janus kinase 2 inhibitor, has been shown to inhibit P-glycoprotein (P-gp), breast cancer resistance protein (BCRP), organic anion transporting polypeptide (OATP) 1B1, OATP1B3, organic cation transporter (OCT) 2, and multidrug and toxin extrusion (MATE) 1 and MATE2-K in vitro. The objective of this study was to evaluate the influence of fedratinib on the pharmacokinetics (PK) of digoxin (P-gp substrate), rosuvastatin (OATP1B1/1B3 and BCRP substrate), and metformin (OCT2 and MATE1/2-K substrate). METHODS: In this nonrandomized, fixed-sequence, open-label study, 24 healthy adult participants received single oral doses of digoxin 0.25 mg, rosuvastatin 10 mg, and metformin 1000 mg administered as a drug cocktail (day 1, period 1). After a 6-day washout, participants received oral fedratinib 600 mg 1 h before the cocktail on day 7 (period 2). An oral glucose tolerance test (OGTT) was performed to determine possible influences of fedratinib on the antihyperglycemic effect of metformin. RESULTS: Plasma exposure to the three probe drugs was generally comparable in the presence or absence of fedratinib. Reduced metformin renal clearance by 36% and slightly higher plasma glucose levels after OGTT were observed in the presence of fedratinib. Single oral doses of the cocktail ± fedratinib were generally well tolerated. CONCLUSIONS: These results suggest that fedratinib has minimal impact on the exposure of P-gp, BCRP, OATP1B1/1B3, OCT2, and MATE1/2-K substrates. Since renal clearance of metformin was decreased in the presence of fedratinib, caution should be exercised in using coadministered drugs that are renally excreted via OCT2 and MATEs. TRIAL REGISTRATION: Clinicaltrials.gov NCT04231435 on January 18, 2020.


Asunto(s)
Digoxina/farmacocinética , Interacciones Farmacológicas , Metformina/farmacocinética , Pirrolidinas/farmacología , Rosuvastatina Cálcica/farmacocinética , Sulfonamidas/farmacología , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/antagonistas & inhibidores , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/antagonistas & inhibidores , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Administración Oral , Adolescente , Adulto , Anciano , Anticolesterolemiantes/farmacocinética , Transporte Biológico , Cardiotónicos/farmacocinética , Estudios de Casos y Controles , Femenino , Estudios de Seguimiento , Voluntarios Sanos , Humanos , Hipoglucemiantes/farmacocinética , Masculino , Persona de Mediana Edad , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/metabolismo , Ensayos Clínicos Controlados no Aleatorios como Asunto , Transportadores de Anión Orgánico/antagonistas & inhibidores , Transportadores de Anión Orgánico/metabolismo , Distribución Tisular , Adulto Joven
3.
Molecules ; 26(15)2021 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-34361774

RESUMEN

Polyphenols play a therapeutic role in vascular diseases, acting in inherent illness-associate conditions such as inflammation, diabetes, dyslipidemia, hypertension, and oxidative stress, as demonstrated by clinical trials and epidemiological surveys. The main polyphenol cardioprotective mechanisms rely on increased nitric oxide, decreased asymmetric dimethylarginine levels, upregulation of genes encoding antioxidant enzymes via the Nrf2-ARE pathway and anti-inflammatory action through the redox-sensitive transcription factor NF-κB and PPAR-γ receptor. However, poor polyphenol bioavailability and extensive metabolization restrict their applicability. Polyphenols carried by nanoparticles circumvent these limitations providing controlled release and better solubility, chemical protection, and target achievement. Nano-encapsulate polyphenols loaded in food grade polymers and lipids appear to be safe, gaining resistance in the enteric route for intestinal absorption, in which the mucoadhesiveness ensures their increased uptake, achieving high systemic levels in non-metabolized forms. Nano-capsules confer a gradual release to these compounds, as well as longer half-lives and cell and whole organism permanence, reinforcing their effectiveness, as demonstrated in pre-clinical trials, enabling their application as an adjuvant therapy against cardiovascular diseases. Polyphenol entrapment in nanoparticles should be encouraged in nutraceutical manufacturing for the fortification of foods and beverages. This study discusses pre-clinical trials evaluating how nano-encapsulate polyphenols following oral administration can aid in cardiovascular performance.


Asunto(s)
Antioxidantes/farmacología , Cardiotónicos/farmacología , Composición de Medicamentos/métodos , Hipertensión/tratamiento farmacológico , Isquemia Miocárdica/tratamiento farmacológico , Polifenoles/farmacología , Elementos de Respuesta Antioxidante , Antioxidantes/química , Antioxidantes/farmacocinética , Arginina/análogos & derivados , Arginina/antagonistas & inhibidores , Arginina/metabolismo , Cardiotónicos/química , Cardiotónicos/farmacocinética , Diabetes Mellitus/tratamiento farmacológico , Diabetes Mellitus/genética , Diabetes Mellitus/metabolismo , Diabetes Mellitus/fisiopatología , Portadores de Fármacos , Dislipidemias/tratamiento farmacológico , Dislipidemias/genética , Dislipidemias/metabolismo , Dislipidemias/fisiopatología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Hipertensión/genética , Hipertensión/metabolismo , Hipertensión/fisiopatología , Isquemia Miocárdica/genética , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/fisiopatología , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Nanocápsulas/administración & dosificación , Nanocápsulas/química , Óxido Nítrico/antagonistas & inhibidores , Óxido Nítrico/biosíntesis , Estrés Oxidativo/efectos de los fármacos , Polifenoles/química , Polifenoles/farmacocinética , Transducción de Señal
4.
Molecules ; 26(13)2021 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-34209338

RESUMEN

Flavonoids comprise a large group of structurally diverse polyphenolic compounds of plant origin and are abundantly found in human diet such as fruits, vegetables, grains, tea, dairy products, red wine, etc. Major classes of flavonoids include flavonols, flavones, flavanones, flavanols, anthocyanidins, isoflavones, and chalcones. Owing to their potential health benefits and medicinal significance, flavonoids are now considered as an indispensable component in a variety of medicinal, pharmaceutical, nutraceutical, and cosmetic preparations. Moreover, flavonoids play a significant role in preventing cardiovascular diseases (CVDs), which could be mainly due to their antioxidant, antiatherogenic, and antithrombotic effects. Epidemiological and in vitro/in vivo evidence of antioxidant effects supports the cardioprotective function of dietary flavonoids. Further, the inhibition of LDL oxidation and platelet aggregation following regular consumption of food containing flavonoids and moderate consumption of red wine might protect against atherosclerosis and thrombosis. One study suggests that daily intake of 100 mg of flavonoids through the diet may reduce the risk of developing morbidity and mortality due to coronary heart disease (CHD) by approximately 10%. This review summarizes dietary flavonoids with their sources and potential health implications in CVDs including various redox-active cardioprotective (molecular) mechanisms with antioxidant effects. Pharmacokinetic (oral bioavailability, drug metabolism), toxicological, and therapeutic aspects of dietary flavonoids are also addressed herein with future directions for the discovery and development of useful drug candidates/therapeutic molecules.


Asunto(s)
Antioxidantes , Cardiotónicos , Enfermedades Cardiovasculares , Flavonoides , Frutas/química , Verduras/química , Antioxidantes/química , Antioxidantes/farmacocinética , Antioxidantes/uso terapéutico , Disponibilidad Biológica , Cardiotónicos/química , Cardiotónicos/farmacocinética , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/patología , Enfermedades Cardiovasculares/prevención & control , Flavonoides/química , Flavonoides/farmacocinética , Flavonoides/uso terapéutico , Humanos
5.
Drug Metab Dispos ; 49(9): 833-843, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34162688

RESUMEN

Oleuropein (OLE), the main constituent of Olea europaea, displays pleiotropic beneficial effects in health and disease, which are mainly attributed to its anti-inflammatory and cardioprotective properties. Several food supplements and herbal medicines contain OLE and are available without a prescription. This study investigated the effects of OLE on the main cytochrome P450s (P450s) catalyzing the metabolism of many prescribed drugs. Emphasis was given to the role of peroxisome proliferator-activated receptor α (PPARα), a nuclear transcription factor regulating numerous genes including P450s. 129/Sv wild-type and Ppara-null mice were treated with OLE for 6 weeks. OLE induced Cyp1a1, Cyp1a2, Cyp1b1, Cyp3a14, Cyp3a25, Cyp2c29, Cyp2c44, Cyp2d22, and Cyp2e1 mRNAs in liver of wild-type mice, whereas no similar effects were observed in Ppara-null mice, indicating that the OLE-induced effect on these P450s is mediated by PPARα. Activation of the pathways related to phosphoinositide 3-kinase/protein kinase B (AKT)/forkhead box protein O1, c-Jun N-terminal kinase, AKT/p70, and extracellular signal-regulated kinase participates in P450 induction by OLE. These data indicate that consumption of herbal medicines and food supplements containing OLE could accelerate the metabolism of drug substrates of the above-mentioned P450s, thus reducing their efficacy and the outcome of pharmacotherapy. Therefore, OLE-induced activation of PPARα could modify the effects of drugs due to their increased metabolism and clearance, which should be taken into account when consuming OLE-containing products with certain drugs, in particular those of narrow therapeutic window. SIGNIFICANCE STATEMENT: This study indicated that oleuropein, which belongs to the main constituents of the leaves and olive drupes of Olea europaea, induces the synthesis of the major cytochrome P450s (P450s) metabolizing the majority of prescribed drugs via activation of peroxisome proliferator-activated receptor α. This effect could modify the pharmacokinetic profile of co-administered drug substrates of the P450s, thus altering their therapeutic efficacy and toxicity.


Asunto(s)
Sistema Enzimático del Citocromo P-450 , Interacciones Farmacológicas , Inactivación Metabólica/efectos de los fármacos , Glucósidos Iridoides/farmacocinética , Oleaceae , PPAR alfa/metabolismo , Animales , Antiinflamatorios/farmacocinética , Cardiotónicos/farmacocinética , Sistema Enzimático del Citocromo P-450/clasificación , Sistema Enzimático del Citocromo P-450/metabolismo , Regulación de la Expresión Génica , Ratones , Fitoquímicos/farmacocinética , Medicamentos bajo Prescripción/farmacocinética
6.
BMC Vet Res ; 17(1): 89, 2021 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-33622315

RESUMEN

BACKGROUND: Prior studies have suggested that pimobendan is associated with several positive effects in cats, including improved survival in cats with congestive heart failure and improved left atrial function in research colony cats with hypertrophic cardiomyopathy (HCM) and normal cats. However, there is still a paucity of pharmacodynamic data refuting or supporting the use of pimobendan in a clinical cat population. This clinical trial aimed to evaluate the pharmacodynamic effects and tolerability of a single dose of pimobendan in cats with HCM. Echocardiograms and Doppler-derived systolic blood pressures were performed in 21 client-owned cats with subclinical HCM at baseline and 90-min after oral administration of 1.25 mg of pimobendan (Vetmedin). Seven additional cats were evaluated post-placebo administration to account for intra-day variability. RESULTS: Heart rate, systolic blood pressure, and murmur grade were not significantly different between baseline and post-pimobendan evaluations. Left auricular blood flow velocity, left atrial size, and left ventricular fractional shortening were not significantly different between baseline and post-pimobendan evaluations. Mean (± standard deviation) tissue Doppler peak systolic velocity of the mitral annulus was significantly higher following pimobendan (7.4 cm/s ± 1.5 vs 8.5 ± 1.6; p = 0.02). Median (min, max) left-ventricular outflow tract maximum velocity was significantly higher following pimobendan [1.9 m/sec (1.5, 3.4) vs 2.6 m/sec (2.0, 4.0); p = 0.01]. Mean right-ventricular outflow tract maximum velocity was also significantly higher following pimobendan (1.5 m/s ± 0.51 vs 2.0 ± 0.53; p = 0.004). Mean left atrial fractional shortening was significantly higher following pimobendan (28% ± 6 vs 32% ± 7; p = 0.02). No adverse events were observed following pimobendan administration. Right ventricular outflow tract velocity was significantly higher following placebo in control cats (1.02 ± 0.21 versus 1.31 ± 0.31; p = 0.01). No other significant differences were detected. CONCLUSIONS: In client-owned cats with HCM, pimobendan acutely increased left atrial function and mildly increased left ventricular systolic function. Left ventricular outflow tract velocity was increased after pimobendan. Pimobendan was well tolerated in the acute setting in cats with HCM. The findings of this prospective, acute-dosing study confirm previous findings in research animals and retrospective analyses and suggest that chronic dosing studies are safe and warranted.


Asunto(s)
Cardiomiopatía Hipertrófica/veterinaria , Cardiotónicos/farmacocinética , Enfermedades de los Gatos/tratamiento farmacológico , Piridazinas/farmacocinética , Animales , Velocidad del Flujo Sanguíneo/efectos de los fármacos , Velocidad del Flujo Sanguíneo/veterinaria , Presión Sanguínea , Cardiomiopatía Hipertrófica/tratamiento farmacológico , Cardiotónicos/farmacología , Gatos , Ecocardiografía/veterinaria , Femenino , Frecuencia Cardíaca/efectos de los fármacos , Masculino , Estudios Prospectivos , Piridazinas/farmacología , Función Ventricular Izquierda
7.
Eur J Drug Metab Pharmacokinet ; 46(3): 325-342, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33616855

RESUMEN

BACKGROUND: Digoxin is a cardiac glycoside that was introduced to cardiovascular medicine more than 200 years ago. Its use is associated with large variability, which complicates achieving the desired therapeutic outcomes. OBJECTIVES: To present a synthesis of the available literature on the population pharmacokinetics of digoxin in adults and to identify the sources of variability in its pharmacokinetics. METHODS: This is a PROSPERO registered systematic review (CRD42018105300). A literature search was conducted using the ISI Web of Science, Science Direct, PubMed, and SCOPUS databases to identify digoxin population pharmacokinetic studies of adults that utilized the nonlinear mixed-effect modeling approach. RESULTS: Sixteen articles were included in the present analysis. Only two studies were conducted in elderly subjects as a separate population. Both the pharmacokinetics and pharmacodynamics of digoxin were investigated in one study. Furthermore, the reviewed studies were mostly conducted in East Asian populations (68.8%). Digoxin's pharmacokinetics were usually described by a one-compartment model because of the nature of the collected data. Weight, age, kidney function, presence of heart failure, and co-administered medications such as calcium channel blockers were the most commonly identified predictors of digoxin clearance. The value of apparent clearance in a typical study individual ranged from 0.005 to 0.2 l/h/kg, while the value of the apparent volume of distribution ranged from 3.14 to 15.2 l/kg. The quality of model evaluation was deemed excellent only in 31.3% of the studies. CONCLUSION: This review provides information about variables that need to be considered when prescribing digoxin. The results highlight the need for prospective studies that allow two-compartment pharmacokinetic/pharmacodynamic models to be established, with a special focus on the elderly subpopulation.


Asunto(s)
Cardiotónicos/farmacocinética , Digoxina/farmacocinética , Modelos Biológicos , Adulto , Humanos , Proyectos de Investigación , Distribución Tisular
8.
Fundam Clin Pharmacol ; 35(2): 397-409, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32757312

RESUMEN

The blood-brain barrier (BBB) is a highly selective membrane composed predominantly of brain capillary endothelial cells expressing drug efflux transporters that prevent substrates from accessing the brain. Inflammation is associated with central nervous system diseases and can impair BBB permeability via several mechanisms, including altered transporter and cell junction expression. This can modify the brain's exposure to drugs. However, comprehensive genomic analysis of the impact of interleukin (IL)-6, which plays a key role in the inflammatory response, on the BBB is lacking. In the present study, we analyzed the effects of exposure of hCMEC/D3 cells to 20 ng/mL IL-6 for 72 h. We performed RNA sequencing and ABC transporter efflux assays. Physiologically based pharmacokinetics (PBPK) simulations were conducted to evaluate the potential impact of IL-6 on the digoxin pharmacokinetics profile and brain exposure by decreasing BBB ABCB1 efflux activity. Exposure of hCMEC/D3 cells to IL-6 triggered the deregulation of numerous genes involved in barrier permeability, such as cell junctions, focal adherens complex, and cell adhesion molecules. We observed mild modification of the mRNA expression and efflux activities of ABC transporters. PBPK simulation showed that, if we only consider the impact of IL-6 on ABCB1 transporter, the modification of the digoxin pharmacokinetics profile and brain exposure is slight. IL-6 slightly affected the gene expression levels and activities of ABC transporters on BBB cells, exhibiting a weaker effect than on hepatic cells. However, inflammation may cause other modifications, such as altered BBB permeability, that could modify drug pharmacokinetics.


Asunto(s)
Transporte Biológico/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Cardiotónicos/farmacocinética , Digoxina/farmacocinética , Interleucina-6/farmacología , Adulto , Femenino , Humanos , Masculino , Persona de Mediana Edad , Modelos Teóricos , Adulto Joven
9.
Clin Pharmacol Ther ; 109(6): 1618-1630, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33283268

RESUMEN

Clinical assessment of drug-drug interactions (DDIs) in children is not a common practice in drug development. Therefore, physiologically-based pharmacokinetic (PBPK) modeling can be beneficial for informing drug labeling. Using ivabradine and its metabolite (both cytochrome P450 3A4 enzyme (CYP3A4) substrates), the objectives were (i) to scale ivabradine-metabolite adult PBPK/PD to pediatrics, (ii) to predict the DDIs with a strong CYP3A4 inhibitor, and (iii) to compare the sensitivity of children to DDIs using two CYP3A4 hepatic ontogeny functions: Salem and Upreti. A scaled parent-metabolite PBPK/PD model from adults to children satisfactorily predicted pharmacokinetics (PK) and pharmacodynamics (PD) in 74 children (0.5-18 years) regardless of CYP3A4 hepatic ontogeny function applied. However, using the Salem ontogeny, mean predicted parent and metabolite area under the concentration-time curve over 12 hours (AUC12h ) and heart rate change from baseline were 2-fold, 1.5-fold, and 1.4-fold higher in young children (0.5-3 years old) compared with Upreti ontogeny, respectively. Despite these differences, choice of appropriate hepatic CYP3A4 ontogeny was challenging due to sparse PK and PD data. Different sensitivity to ivabradine-ketoconazole DDIs was simulated in young children relative to adults depending on the choice of hepatic CYP3A4 ontogeny. Predicted ivabradine and metabolite AUCDDI /AUCcontrol were 2-fold lower in the youngest children (0.5-1 year old) compared with adults (Salem function). In contrast, the Upreti function predicted comparable ivabradine DDIs across all age groups, although predicted metabolite AUCDDI/ AUCcontrol was 1.3-fold higher between the youngest children and adults. In the case of PD, differences in predicted DDIs were minor across age groups and between both functions. Current work highlights the importance of careful consideration of hepatic CYP3A4 ontogeny function and implications on labeling recommendations in the pediatric population.


Asunto(s)
Cardiotónicos/farmacocinética , Citocromo P-450 CYP3A/genética , Interacciones Farmacológicas , Ivabradina/farmacocinética , Hígado/enzimología , Adolescente , Antifúngicos/efectos adversos , Antifúngicos/farmacocinética , Cardiotónicos/efectos adversos , Niño , Preescolar , Inductores del Citocromo P-450 CYP3A , Inhibidores del Citocromo P-450 CYP3A/farmacocinética , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/epidemiología , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/genética , Humanos , Lactante , Recién Nacido , Ivabradina/efectos adversos , Cetoconazol/efectos adversos , Cetoconazol/farmacocinética , Pediatría
10.
BMC Pharmacol Toxicol ; 21(1): 82, 2020 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-33239108

RESUMEN

BACKGROUND: To study the pharmacokinetic and -dynamic behavior of landiolol in the presence of dobutamine in healthy subjects of European ancestry. METHODS: We conducted a single-center, prospective randomized study in 16 healthy subjects each receiving an infusion of dobutamine sufficient to increase heart rate by 30 bpm followed by a 60 min infusion of 10 µg/kg/min landiolol. RESULTS: Dobutamine-induced increases in heart rate were stable for at least 20 min before a 60 min landiolol- infusion was started. The dobutamine effects were rapidly antagonized by landiolol within 16 min. A further slight decrease in heart rate during 20-60 min of the landiolol infusion occurred as well. Upon termination of landiolol infusion, heart rate and blood pressure recovered rapidly in response to the persisting dobutamine infusion but did not return to the maximum values before landiolol infusion. The pharmacokinetic parameters of landiolol in presence of dobutamine showed a short half-life (3.5 min) and a low distribution volume (0.3 l/kg). No serious adverse events were observed. CONCLUSION: Landiolol can antagonize the dobutamine-induced increases in heart rate and blood pressure in a fast way. A rapid bradycardic effect until steady-state plasma levels is followed by a slow heart rate reduction. The latter can be attributed to an early desensitization to dobutamine. Consequently, after termination of landiolol, the heart rate did not achieve maximum pre-landiolol values. The pharmacokinetics of landiolol during dobutamine infusion are similar when compared to short- and long-term data in Caucasian subjects. Landiolol in the given dose can thus serve as an antagonist of dobutamine-induced cardiac effects. TRIAL REGISTRATION: Registration number 2010-023311-34 at the EU Clinical Trials Register, registration date 2010-12-21.


Asunto(s)
Antagonistas Adrenérgicos beta/farmacocinética , Presión Sanguínea/efectos de los fármacos , Cardiotónicos/farmacocinética , Dobutamina/farmacocinética , Frecuencia Cardíaca/efectos de los fármacos , Morfolinas/farmacocinética , Urea/análogos & derivados , Antagonistas Adrenérgicos beta/administración & dosificación , Adulto , Presión Sanguínea/fisiología , Cardiotónicos/administración & dosificación , Estudios Cruzados , Dobutamina/administración & dosificación , Método Doble Ciego , Femenino , Voluntarios Sanos , Frecuencia Cardíaca/fisiología , Humanos , Infusiones Intravenosas , Masculino , Morfolinas/administración & dosificación , Estudios Prospectivos , Urea/administración & dosificación , Urea/farmacocinética , Adulto Joven
11.
Clin Cardiol ; 43(12): 1641-1648, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33140454

RESUMEN

BACKGROUND: Recently published studies suggested that digoxin may increase mortality in heart failure with reduced ejection fraction (HFrEF). However, in the vast majority of former trials serum digoxin concentration (SDC) was not measured and therapy was not SDC-guided. AIM: To assess the impact of SDC-guided digoxin therapy on mortality in HFrEF patients. METHODS: Data of 580 HFrEF patients were retrospectively analyzed. In patients on digoxin, SDC was measured every 3 months and digoxin dosage was SDC-guided (target SDC: 0.5-0.9 ng/mL). All-cause mortality of digoxin users and nonusers was compared after propensity score matching (PSM). RESULTS: After 7.1 ± 4.7 years follow-up period (FUP) all-cause mortality of digoxin users (n = 180) was significantly higher than nonusers (n = 297) (propensity-adjusted HR = 1.430; 95% CI = 1.134-1.804; P = .003). Patients having SDC of 0.9 to 1.1 ng/mL (n = 60) or > 1.1 ng/mL (n = 44) at any time during the FUP had an increased risk of all-cause mortality (HR = 1.750; 95% CI = 1.257-2.436, P = .001 and HR = 1.687; 95% CI = 1.153-2.466, P = .007), while patients having a maximal SDC < 0.9 ng/mL (n = 76) had similar mortality risk (HR = 1.139; 95% CI = 0.827-1.570, P = .426), compared to digoxin nonusers. CONCLUSIONS: According to our propensity-matched analysis, SDC-guided digoxin therapy was associated with increased all-cause mortality in optimally treated HFrEF patients, especially with SDC ≥0.9 ng/mL. These results reinforce the expert opinion that digoxin in HFrEF can only be used among carefully selected patients with close SDC monitoring.


Asunto(s)
Digoxina/farmacocinética , Insuficiencia Cardíaca/tratamiento farmacológico , Puntaje de Propensión , Volumen Sistólico/fisiología , Cardiotónicos/farmacocinética , Femenino , Estudios de Seguimiento , Insuficiencia Cardíaca/sangre , Insuficiencia Cardíaca/mortalidad , Humanos , Hungría/epidemiología , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Tasa de Supervivencia/tendencias , Factores de Tiempo , Resultado del Tratamiento
12.
Curr Drug Deliv ; 17(8): 711-719, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32621716

RESUMEN

BACKGROUND: Ginkgolides are widely used in cardio-protective therapy; however, poor bioavailability currently limits their application. OBJECTIVE: The purpose of this study was to demonstrate whether solid dispersions prepared with Low- Molecular-Weight Chitosan (LMWC) could improve the protective effect of ginkgolides on Myocardial Injury (MI). METHODS: Ginkgolide Solid Dispersions (GKSD) were prepared with LMWC. Their properties were then characterized using differential scanning calorimetry, X-ray diffraction, scanning electron microscopy and Fourier transform infrared spectroscopy. In vivo pharmacokinetic studies were performed in rats, and the protective effect of GKSD on MI was investigated by western blotting and immunohistochemical analyses. RESULTS: Drug dissolution testing showed that GDSD were released at a significantly higher rate than ginkgolides, dissolved by alternative methods, suggesting that LMWC facilitates the release of ginkgolides. Differential scanning calorimetry, X-ray diffraction, scanning electron microscopy, and Fourier transform infrared spectroscopy all showed that GKSD was amorphous. In-vivo testing revealed larger AUC0-t, higher Cmax, and shorter Tmax for GKSD compared to that in original ginkgolides. Myocardial injury was induced in rats with isoproterenol to test the protective effect of GKSD. GKSD alleviated MI and reduced myocardial fibrosis, as observed by Hematoxylin and Eosin staining. Compared with the crude drug group, the secretion of malonyl dialdehyde and nitric oxide and expression of NOX-2 and NOX-4 were lower. The activities of the cardiac marker enzymes SOD, CAT, GPX, GPX-1, and GSH were higher in GKSD-administered rats, indicating a beneficial effect of GKSD in eliminating free radicals during myocardial injury. Additionally, western blotting and immunohistochemical analysis showed that GKSD markedly reduced the expression of signaling proteins RHOA, ROCK1, ROCK2, and RAC1. CONCLUSION: Solid dispersions prepared with low molecular weight chitosan improved the oral bioavailability of ginkgolide and enhanced its protective effect on myocardial injury.


Asunto(s)
Cardiotónicos/administración & dosificación , Quitosano/química , Portadores de Fármacos/química , Ginkgólidos/administración & dosificación , Infarto del Miocardio/tratamiento farmacológico , Administración Oral , Animales , Área Bajo la Curva , Disponibilidad Biológica , Cardiotónicos/farmacocinética , Modelos Animales de Enfermedad , Liberación de Fármacos , Ginkgólidos/farmacocinética , Humanos , Isoproterenol/administración & dosificación , Isoproterenol/toxicidad , Peso Molecular , Infarto del Miocardio/inducido químicamente , Infarto del Miocardio/patología , Miocardio/patología , Ratas , Solubilidad
13.
Sci Rep ; 10(1): 12457, 2020 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-32719417

RESUMEN

Effects of different genotypes on the pharmacokinetics of probe substrates may support their use as phenotyping agents for the activity of the respective enzyme or transporter. Digoxin is recommended as a probe substrate to assess the activity of the transporter P-glycoprotein (P-gp) in humans. Current studies on the individual effects of three commonly investigated single nucleotide polymorphisms (SNPs) of the ABCB1 gene encoding P-gp (C1236T, G2677T/A, and C3435T) on digoxin pharmacokinetics are inconclusive. Since SNPs are in incomplete linkage disequilibrium, considering combinations of these SNPs might be necessary to assess the role of polymorphisms in digoxin pharmacokinetics accurately. In this study, the relationship between SNP combinations and digoxin pharmacokinetics was explored via a population pharmacokinetic approach in 40 volunteers who received oral doses of 0.5 mg digoxin. Concerning the SNPs 1236/2677/3435, the following combinations were evaluated: CGC, CGT, and TTT. Carriers of CGC/CGT and TTT/TTT had 35% higher apparent bioavailability compared to the reference group CGC/CGC, while no difference was seen in CGC/TTT carriers. No significant effect on renal clearance was observed. The population pharmacokinetic model supports the use of oral digoxin as a phenotyping substrate of intestinal P-gp, but not to assess renal P-gp activity.


Asunto(s)
Digoxina/farmacocinética , Riñón/metabolismo , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Adulto , Anciano , Disponibilidad Biológica , Cardiotónicos/administración & dosificación , Cardiotónicos/farmacocinética , Digoxina/administración & dosificación , Femenino , Haplotipos , Humanos , Masculino , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple , Adulto Joven
14.
Molecules ; 25(9)2020 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-32349420

RESUMEN

Isofraxidin (7-hydroxy-6, 8-dimethoxy coumarin) (IF) is a hydroxy coumarin with several biological and pharmacological activities. The plant kingdom is of the most prominent sources of IF, which, among them, Eleutherococcus and Fraxinus are the well-known genera in which IF could be isolated/extracted from their species. Considering the complex pathophysiological mechanisms behind some diseases (e.g., cancer, neurodegenerative diseases, and heart diseases), introducing IF as a potent multi-target agent, which possesses several herbal sources and the multiple methods for isolation/purification/synthesis, along with the unique pharmacokinetic profile and low levels of side effects, could be of great importance. Accordingly, a comprehensive review was done without time limitations until February 2020. IF extraction methods include microwave, mechanochemical, and ultrasound, along with other conventional methods in the presence of semi-polar solvents such as ethyl acetate (EtOAc). In addition to the isolation methods, related synthesis protocols of IF is also of great importance. From the synthesis point of view, benzaldehyde derivatives are widely used as precursors for IF synthesis. Along with the methods of isolation and biosynthesis, IF pharmacokinetic studies showed hopeful in vivo results of its rapid absorption after oral uses, leading to different pharmacological effects. In this regard, IF targets varieties of inflammatory mediators including nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), tumor necrosis factor-α (TNF-α), and matrix metalloproteinases (MMPs). thereby indicating anticancer, cardioprotective, and neuroprotective effects. This is the first review on the synthesis, biosynthesis, isolation, and pharmacokinetic and pharmacological properties of IF in combating different diseases.


Asunto(s)
Antiinflamatorios/farmacología , Antineoplásicos/farmacología , Antioxidantes/farmacología , Cardiotónicos/farmacología , Cumarinas/aislamiento & purificación , Cumarinas/farmacología , Fármacos Neuroprotectores/farmacología , Animales , Antiinflamatorios/aislamiento & purificación , Antiinflamatorios/farmacocinética , Antineoplásicos/aislamiento & purificación , Antineoplásicos/farmacocinética , Antioxidantes/aislamiento & purificación , Antioxidantes/farmacocinética , Cardiotónicos/aislamiento & purificación , Cardiotónicos/farmacocinética , Cumarinas/síntesis química , Cumarinas/química , Cumarinas/farmacocinética , Eleutherococcus/química , Fraxinus/química , Humanos , Metaloproteinasas de la Matriz/metabolismo , FN-kappa B/metabolismo , Fármacos Neuroprotectores/aislamiento & purificación , Fármacos Neuroprotectores/farmacocinética , Solventes/química , Factor de Necrosis Tumoral alfa/metabolismo
15.
Pharmacol Res ; 155: 104755, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32173585

RESUMEN

Stachydrine is extracted from the leaves of Leonurus japonicus Houtt (or Motherwort, "Yi Mu Cao" in Traditional Chinese Medicine) and is the major bioactive ingredient. So far, stachydrine has demonstrated various bioactivities for the treatment of fibrosis, cardiovascular diseases, cancers, uterine diseases, brain injuries, and inflammation. The pharmacological and pharmacokinetic properties of stachydrine up to 2019 have been comprehensively searched and summarized. This review provides an updated summary of recent studies on the pharmacological activities of stachydrine. Many studies have demonstrated that stachydrine has strong anti-fibrotic properties (on various types of fibrosis) by inhibiting ECM deposition and decreasing inflammatory and oxidative stress through multiple molecular mechanisms (including TGF-ß, ERS-mediated apoptosis, MMPs/TIMPs, NF-κB, and JAK/STAT). The cardioprotective and vasoprotective activities of stachydrine are related to its inhibition of ß-MHC, excessive autophagy, SIRT1, eNOS uncoupling and TF, promotion of SERCA, and angiogenesis. In addition to its anticancer action, regulation of the uterus, neuroprotective effects, etc. the pharmacokinetic properties of stachydrine are also discussed.


Asunto(s)
Prolina/análogos & derivados , Animales , Antiinflamatorios/farmacocinética , Antiinflamatorios/farmacología , Antiinflamatorios/toxicidad , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Antineoplásicos/toxicidad , Cardiotónicos/farmacocinética , Cardiotónicos/farmacología , Cardiotónicos/toxicidad , Femenino , Fibrosis , Humanos , Fármacos Neuroprotectores/farmacocinética , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/toxicidad , Prolina/farmacocinética , Prolina/farmacología , Prolina/toxicidad , Útero/efectos de los fármacos
16.
Basic Res Cardiol ; 115(3): 24, 2020 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-32140789

RESUMEN

Intramyocardial hemorrhage is an independent predictor of adverse outcomes in ST-segment elevation myocardial infarction (STEMI). Iron deposition resulting from ischemia-reperfusion injury (I/R) is pro-inflammatory and has been associated with adverse remodeling. The role of iron chelation in hemorrhagic acute myocardial infarction (AMI) has never been explored. The purpose of this study was to investigate the cardioprotection offered by the iron-chelating agent deferiprone (DFP) in a porcine AMI model by evaluating hemorrhage neutralization and subsequent cardiac remodeling. Two groups of animals underwent a reperfused AMI procedure: control and DFP treated (N = 7 each). A comprehensive MRI examination was performed in healthy state and up to week 4 post-AMI, followed by histological assessment. Infarct size was not significantly different between the two groups; however, the DFP group demonstrated earlier resolution of hemorrhage (by T2* imaging) and edema (by T2 imaging). Additionally, ventricular enlargement and myocardial hypertrophy (wall thickness and mass) were significantly smaller with DFP, suggesting reduced adverse remodeling, compared to control. The histologic results were consistent with the MRI findings. To date, there is no effective targeted therapy for reperfusion hemorrhage. Our proof-of-concept study is the first to identify hemorrhage-derived iron as a therapeutic target in I/R and exploit the cardioprotective properties of an iron-chelating drug candidate in the setting of AMI. Iron chelation could potentially serve as an adjunctive therapy in hemorrhagic AMI.


Asunto(s)
Cardiotónicos/farmacología , Deferiprona/uso terapéutico , Hemorragia/tratamiento farmacológico , Hemorragia/etiología , Quelantes del Hierro/uso terapéutico , Infarto del Miocardio/complicaciones , Miocardio/patología , Remodelación Ventricular/efectos de los fármacos , Animales , Cardiotónicos/farmacocinética , Cardiotónicos/uso terapéutico , Deferiprona/farmacocinética , Deferiprona/farmacología , Modelos Animales de Enfermedad , Femenino , Hemorragia/patología , Quelantes del Hierro/farmacocinética , Quelantes del Hierro/farmacología , Infarto del Miocardio/patología , Porcinos
17.
Pharmacol Res ; 155: 104694, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32061664

RESUMEN

Myocardial fibrosis is an endogenous response to different cardiac insults that may become maladaptive over time and contribute to the onset and progression of heart failure (HF). Fibrosis is a direct and indirect target of established HF therapies, namely inhibitors of the renin-angiotensin-aldosterone system, but its resilience to therapy warrants a search for novel, more targeted approaches to myocardial fibrosis. Pirfenidone is a drug approved for idiopathic pulmonary fibrosis, a severe form of idiopathic interstitial pneumonias. Pirfenidone is a small synthetic molecule with high oral bioavailability, exerting an antifibrotic activity, but also anti-oxidant and anti-inflammatory effects. These effects have been attributed to the inhibition of several growth factors (in particular transforming growth factor-ß, but also platelet-derived growth factor and beta fibroblast growth factor), matrix metalloproteinases, and pro-inflammatory mediators (such as interleukin-1ß and tumour necrosis factor-α), and possibly also an improvement of mitochondrial function and modulation of lymphocyte activation. Given the activation of similar profibrotic pathways in lung and heart disease, the crucial role of fibrosis in several cardiac disorders, and the wide spectrum of activity of pirfenidone, this drug has been evaluated with interest as a potential treatment for cardiac disorders. In animal studies, pirfenidone has shown cardioprotective effects across different species and in a variety of models of cardiomyopathy. In the present review we summarize the pharmacological characteristics of pirfenidone and the data from animal studies supporting its cardioprotective effects.


Asunto(s)
Cardiotónicos , Piridonas , Animales , Cardiotónicos/efectos adversos , Cardiotónicos/farmacocinética , Cardiotónicos/farmacología , Cardiotónicos/uso terapéutico , Fibrosis , Corazón/efectos de los fármacos , Humanos , Miocardio/patología , Piridonas/efectos adversos , Piridonas/farmacocinética , Piridonas/farmacología , Piridonas/uso terapéutico
18.
Ther Drug Monit ; 42(1): 1-5, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31593032

RESUMEN

We describe the case of a patient on continuous venovenous hemodiafiltration with atrial fibrillation with rapid ventricular response and hypotension requiring vasopressor use, which warranted digoxin therapy. In the absence of guidelines specifying appropriate digoxin dosing in patients undergoing continuous venovenous hemodiafiltration, anecdotal evidence-guided digoxin dosing was performed for this patient using plasma digoxin concentration-based therapeutic drug monitoring. We use this case to demonstrate the potential role of physiologically based pharmacokinetic modeling in assisting therapeutic decision making.


Asunto(s)
Cardiotónicos/farmacocinética , Terapia de Reemplazo Renal Continuo/métodos , Digoxina/farmacocinética , Hemodiafiltración/métodos , Hipotensión/tratamiento farmacológico , Insuficiencia Renal/terapia , Anciano , Fibrilación Atrial/complicaciones , Peso Corporal , Cardiotónicos/uso terapéutico , Digoxina/uso terapéutico , Cálculo de Dosificación de Drogas , Monitoreo de Drogas/métodos , Humanos , Hipotensión/complicaciones , Pruebas de Función Renal , Masculino , Modelos Biológicos , Sistemas de Atención de Punto , Insuficiencia Renal/complicaciones
19.
J Drug Target ; 28(3): 282-299, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31353972

RESUMEN

This study aimed to develop valsartan (VAL)-loaded mixed micelles and investigate their cardioprotective potential and molecular mechanisms through Mhrt/Nrf2 and Trx1 pathways. VAL-loaded mixed micelles have not been elaborated and their impact on Mhrt/Nrf2 and Trx1 pathways has not been yet inspected. VAL-loaded mixed micelles were prepared, incorporating Pluronic F127 and Tween 80, adopting thin-film hydration method. The micelles were evaluated for drug entrapment efficiency, loading characteristics, particle size, morphology, in vitro drug release and micelles storage stability. The pharmacokinetic studies were explored in rats. Also, VAL suspension and mixed micelles were tested in cisplatin-induced cardiotoxicity in rats either pre to or simultaneously with cisplatin. RNA expression of lnc Mhrt and protein expression of Nrf2, Trx1, Ask1, AMPK and caspase 3, oxidative stress and cardiac injury markers besides tailed DNA% by comet assay were assessed. Pharmacokinetic studies evoked a 3.75-fold increase in oral bioavailability as compared with VAL suspension. Overall, treatment with VAL-loaded mixed micelles was superior to VAL suspension in decreasing oxidative stress and cardiac injury markers and restoring the abnormalities occurred in Mhrt/Nrf2 and Trx1 pathways. Thus, mixed micelles would be promising nanocarrier for the engineering of VAL with reinforced pharmacokinetics and cardioprotection characteristics.


Asunto(s)
Cardiotónicos/farmacología , Cardiotoxicidad/prevención & control , Cisplatino/toxicidad , Valsartán/farmacología , Animales , Cardiotónicos/administración & dosificación , Cardiotónicos/farmacocinética , Cardiotoxicidad/etiología , Liberación de Fármacos , Masculino , Micelas , Factor 2 Relacionado con NF-E2/metabolismo , Tamaño de la Partícula , Poloxámero/química , Polímeros/química , Polisorbatos/química , ARN Largo no Codificante/metabolismo , Ratas , Ratas Wistar , Tiorredoxinas/metabolismo , Valsartán/administración & dosificación , Valsartán/farmacocinética
20.
Colloids Surf B Biointerfaces ; 187: 110628, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31753617

RESUMEN

Doxorubicin (DOX) is commonly used for the treatment of many types of cancers but its cardiotoxicity, owing to free radical formation, limits its clinical use. Hesperidin (HES), a flavanone glycoside, has been shown to exert multiple pharmacological actions including cardioprotective effects. Herein, we aim to formulate HES loaded solid lipid nanoparticles (SLNs) using supercritical antisolvent (SAS) technology to improve the oral delivery of HES. Process parameters were optimized to produce small size (175.3 ±â€¯3.6 nm) HES-SLNs with high encapsulation efficiency (87.6 ±â€¯3.8 %). DSC and XRD showed that HES is amorphously dispersed in SLNs. Compared to HES, HES-SLNs resulted in a nearly 20-fold increase in aqueous solubility and a nearly 5-fold increase in apparent permeability. Pharmacokinetics in rats revealed nearly 4.5-fold higher bioavailability of HES from SLN formulation compared to HES suspension. Data showed that HES-SLN significantly attenuated DOX-induced cardiotoxicity through lowering creatine kinase-muscle/brain, cardiac troponin I and improving histopathological scores as compared to the DOX group. HES-SLN also decreased malondialdehyde, increased catalase and superoxide dismutase of rats' heart to levels relatively comparable to control. Marked reductions in caspase-3 were also observed following HES-SLN treatment. Conclusively, these results describe a cardioprotective effect for HES-SLN against DOX-induced cardiotoxicity likely facilitated via suppression of oxidative stress and apoptosis.


Asunto(s)
Cardiotónicos/farmacología , Hesperidina/farmacología , Lípidos/química , Nanopartículas/química , Solventes/química , Animales , Apoptosis/efectos de los fármacos , Biomarcadores/metabolismo , Peso Corporal/efectos de los fármacos , Rastreo Diferencial de Calorimetría , Cardiotónicos/farmacocinética , Cardiotoxicidad/patología , Caspasa 3/metabolismo , Corazón/efectos de los fármacos , Hesperidina/farmacocinética , Masculino , Miocardio/patología , Nanopartículas/ultraestructura , Tamaño de los Órganos/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Tamaño de la Partícula , Permeabilidad , Ratas Wistar , Solubilidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...