Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
1.
AAPS J ; 22(4): 92, 2020 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-32676788

RESUMEN

In this study, we characterized the pharmacokinetics of OSU-2S, a fingolimod-derived, non-immunosuppressive phosphatase activator, in mice, rats, and dogs, as well as tolerability and food effects in dogs. Across all species tested, plasma protein binding for OSU-2S was > 99.5%, and metabolic stability and hepatic intrinsic clearance were in the moderate range. OSU-2S did not significantly modulate CYP enzyme activity up until 50 µM, and Caco-2 data suggested low permeability with active efflux at 2 µM. Apparent oral bioavailability in mice was 16% and 69% at 10 and 50 mg/kg, respectively. In rats, bioavailability was 24%, 35%, and 28% at 10, 30, and 100 mg/kg, respectively, while brain/plasma ratio was 36 at 6-h post-dose at 30 mg/kg. In dogs, OSU-2S was well tolerated with oral capsule bioavailability of 27.5%. Plasma OSU-2S exposures increased proportionally over a 2.5-20 mg/kg dose range. After 4 weeks of 3 times weekly, oral administration (20 mg/kg), plasma AUClast (26.1 µM*h), and Cmax (0.899 µM) were nearly 2-fold greater than those after 1 week of dosing, and no food effects were observed. The elimination half-life (29.7 h), clearance (22.9 mL/min/kg), and plasma concentrations of repeated oral doses support a 3-times weekly dosing schedule in dogs. No significant CBC, serum biochemical, or histopathological changes were observed. OSU-2S has favorable oral PK properties similar to fingolimod in rodents and dogs and is well tolerated in healthy animals. This work supports establishing trials of OSU-2S efficacy in dogs with spontaneous tumors to guide its clinical development as a cancer therapeutic for human patients.


Asunto(s)
Análisis de Datos , Clorhidrato de Fingolimod/farmacocinética , Inmunosupresores/farmacocinética , Glicoles de Propileno/farmacocinética , Esfingosina/análogos & derivados , Administración Oral , Animales , Disponibilidad Biológica , Células CACO-2 , Perros , Relación Dosis-Respuesta a Droga , Clorhidrato de Fingolimod/administración & dosificación , Haplorrinos , Humanos , Inmunosupresores/administración & dosificación , Masculino , Ratones , Ratones Endogámicos C57BL , Glicoles de Propileno/administración & dosificación , Ratas , Ratas Sprague-Dawley , Esfingosina/administración & dosificación , Esfingosina/farmacocinética
2.
Eur J Pharmacol ; 870: 172877, 2020 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-31866409

RESUMEN

Sphingosine-1-phosphate (S1P) is emerging as a hypoxia responsive bio-lipid; systemically raised levels of S1P are proposed to have potential hypoxia pre-conditioning effects. The study aims to evaluate the hypoxia pre-conditioning efficacy of exogenously administered S1P in rats exposed to acute (24-48 hs (h)) and sub-chronic (7 days) hypobaric hypoxia. Sprague-Dawley rats (200 ± 20 g) were preconditioned with 1 µg/kg body weight S1P intravenously for three consecutive days. On the third day, control and S1P preconditioned animals were exposed to hypobaric hypoxia equivalent to 7620 m for 24 h, 48 h and 7 days. Post exposure analysis included body weight quantitation, blood gas/chemistry analysis, vascular permeability assays, evaluation of oxidative stress/inflammation parameters, and estimation of hypoxia responsive molecules. S1P preconditioned rats exposed to acute HH display a significant reduction in body weight loss, as a culmination of improved oxygen carrying capacity, increased 2,3- diphosphoglycerate levels and recuperation from energy deficit. Pathological disturbances such as vascular leakage in the lungs and brain, oxidative stress, pro-inflammatory milieu and raised level of endothelin-1 were also reined. The adaptive and protective advantage conferred by S1P in the acute phase of hypobaric hypoxia exposure, is observed to precipitate into an improved sustenance even after sub-chronic (7d) hypobaric hypoxia exposure as indicated by decreased body weight loss, lower edema index and improvement in general pathology biomarkers. Conclusively, administration of 1 µg/kg body weight S1P, in the aforementioned schedule, confer hypoxia pre-conditioning benefits, sustained up to 7 days of hypobaric hypoxia exposure.


Asunto(s)
Hipoxia/tratamiento farmacológico , Hipoxia/prevención & control , Lisofosfolípidos/administración & dosificación , Esfingosina/análogos & derivados , 2,3-Difosfoglicerato/metabolismo , Administración Intravenosa , Animales , Biomarcadores , Peso Corporal , Encéfalo , Permeabilidad Capilar , Citocinas/metabolismo , Inflamación/metabolismo , Pulmón , Lisofosfolípidos/farmacocinética , Estrés Oxidativo , Oxígeno/metabolismo , Ratas , Ratas Sprague-Dawley , Esfingosina/administración & dosificación , Esfingosina/farmacocinética , Distribución Tisular
3.
Artículo en Inglés | MEDLINE | ID: mdl-30836314

RESUMEN

A simple and specific hydrophilic interaction liquid chromatography-tandem mass spectrometry (HILIC-MS/MS) method was developed for the simultaneous determination of C18-L-threo-sphinganine (safingol, an anti-neoplastic in phase I trials) and its diastereomer, C18-D-erythro-sphinganine (sphinganine), in human plasma. Sample pretreatment involved a protein precipitation with methanol using 25 µL aliquots of plasma. Chromatographic separation of the diastereomers and C17-D-erythro-sphinganine, an internal standard, was achieved on a Xbridge HILIC (3.5 µm, 100 × 2.1 mm) using isocratic elution with the mobile phase of 2 mM ammonium bicarbonate in water (pH 8.3) and acetonitrile at a flow rate of 0.3 mL/min. Electrospray ionization (ESI) mass spectrometry was operated in the positive ion mode with multiple reaction monitoring (MRM). The calibration curves obtained were linear over the concentration range of 0.2-100 ng/mL with a lower limit of quantification of 0.2 ng/mL. The relative standard deviation of intra-day and inter-day precision was below 8.27%, and the accuracy ranged from 92.23 to 110.06%. The extraction recoveries were found to be higher than 93.22% and IS-normalized matrix effect was higher than 90.92%. The analytes were stable for the durations of the stability studies. The validated method was successfully applied to the analyses of pharmacokinetic samples from patients treated with safingol and all-trans-N-(4-hydroxyphenyl)retinamide; (fenretinide, 4-HPR) in a current phase I clinical trial (SPOC-2010-002, ClinicalTrials.gov Identifier: NCT01553071).


Asunto(s)
Cromatografía Liquida/métodos , Esfingosina/análogos & derivados , Espectrometría de Masas en Tándem/métodos , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Límite de Detección , Modelos Lineales , Reproducibilidad de los Resultados , Esfingosina/sangre , Esfingosina/química , Esfingosina/farmacocinética
4.
Mar Drugs ; 15(9)2017 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-28862650

RESUMEN

We aimed to investigate the pharmacokinetics and the underlying mechanisms of the intestinal absorption, distribution, metabolism, and excretion of Jaspine B in rats. The oral bioavailability of Jaspine B was 6.2%, but it decreased to 1.6% in bile-depleted rats and increased to 41.2% (normal) and 23.5% (bile-depleted) with taurocholate supplementation (60 mg/kg). Consistent with the increased absorption in the presence of bile salts, rat intestinal permeability of Jaspine B also increased in the presence of 10 mM taurocholate or 20% bile. Further studies demonstrated that the enhanced intestinal permeability with bile salts was due to increased lipophilicity and decreased membrane integrity. Jaspine B was designated as a highly tissue-distributed compound, because it showed large tissue to plasma ratios in the brain, kidney, heart, and spleen. Moreover, the recovery of Jaspine B from the feces and urine after an intravenous administration was about 6.3%, suggesting a substantial metabolism of Jaspine B. Consistent with this observation, 80% of the administered Jaspine B was degraded after 1 h incubation with rat liver microsomes. In conclusion, the facilitated intestinal permeability in the presence of bile salts could significantly increase the bioavailability of Jaspine B and could lead to the development of oral formulations of Jaspine B with bile salts. Moreover, the highly distributed features of Jaspine B in the brain, kidney, heart, and spleen should be carefully considered in the therapeutic effect and toxicity of this compound.


Asunto(s)
Ácidos y Sales Biliares/metabolismo , Absorción Intestinal/efectos de los fármacos , Esfingosina/análogos & derivados , Administración Oral , Animales , Heces/química , Masculino , Microsomas Hepáticos , Ratas , Esfingosina/farmacocinética , Orina/química
5.
Ann Biomed Eng ; 45(4): 1003-1014, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27904998

RESUMEN

Sphingosine-1-phosphate (S1P), a bioactive lipid, is a potent candidate for treatment of ischemic vascular disease. However, designing biomaterial systems for the controlled release of S1P to achieve therapeutic angiogenesis presents both biological and engineering challenges. Thus, the objective of this study was to design a hydrogel system that provides controlled and sustained release of S1P to establish local concentration gradients that promote neovascularization. Alginate hydrogels have been extensively studied and characterized for delivery of proangiogenic factors. We sought to explore if chitosan (0, 0.1, 0.5, or 1%) incorporation could be used as a means to control S1P release from alginate hydrogels. With increasing chitosan incorporation, hydrogels exhibited significantly denser pore structure and stiffer material properties. While 0.1 and 0.5% chitosan gels demonstrated slower respective release of S1P, release from 1% chitosan gels was similar to alginate gels alone. Furthermore, 0.5% chitosan gels induced greater sprouting and directed migration of outgrowth endothelial cells (OECs) in response to released S1P under hypoxia in vitro. Overall, this report presents a platform for a novel alginate-chitosan hydrogel of controlled composition and in situ gelation properties that can be used to control lipid release for therapeutic applications.


Asunto(s)
Alginatos , Quitosano , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Hidrogeles , Lisofosfolípidos , Neovascularización Fisiológica/efectos de los fármacos , Esfingosina/análogos & derivados , Alginatos/química , Alginatos/farmacocinética , Alginatos/farmacología , Células Cultivadas , Quitosano/química , Quitosano/farmacocinética , Quitosano/farmacología , Preparaciones de Acción Retardada/química , Preparaciones de Acción Retardada/farmacocinética , Preparaciones de Acción Retardada/farmacología , Ácido Glucurónico/química , Ácido Glucurónico/farmacocinética , Ácido Glucurónico/farmacología , Ácidos Hexurónicos/química , Ácidos Hexurónicos/farmacocinética , Ácidos Hexurónicos/farmacología , Humanos , Hidrogeles/química , Hidrogeles/farmacocinética , Hidrogeles/farmacología , Lisofosfolípidos/química , Lisofosfolípidos/farmacocinética , Lisofosfolípidos/farmacología , Esfingosina/química , Esfingosina/farmacocinética , Esfingosina/farmacología
6.
J Med Chem ; 58(8): 3471-84, 2015 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-25826109

RESUMEN

Sphingosine-1-phosphate (S1P) is a lysophospholipid that evokes a variety of biological responses via stimulation of a set of cognate G-protein coupled receptors (GPCRs): S1P1-S1P5. S1P and its receptors (S1PRs) play important roles in the immune, cardiovascular, and central nervous systems and have also been implicated in carcinogenesis. Recently, the S1P analogue Fingolimod (FTY720) has been approved for the treatment of patients with relapsing multiple sclerosis. This work presents the synthesis of various fluorinated structural analogues of FTY720, their in vitro and in vivo biological testing, and their development and application as [(18)F]radiotracers for the study of S1PR biodistribution and imaging in mice using small-animal positron emission tomography (PET).


Asunto(s)
Tomografía de Emisión de Positrones/métodos , Glicoles de Propileno/química , Receptores de Lisoesfingolípidos/análisis , Esfingosina/análogos & derivados , Animales , Clorhidrato de Fingolimod , Radioisótopos de Flúor/química , Radioisótopos de Flúor/farmacocinética , Halogenación , Masculino , Ratones , Ratones Endogámicos C57BL , Imagen Molecular/métodos , Glicoles de Propileno/síntesis química , Glicoles de Propileno/farmacocinética , Esfingosina/síntesis química , Esfingosina/química , Esfingosina/farmacocinética , Receptores de Esfingosina-1-Fosfato
7.
Rev. neurol. (Ed. impr.) ; 59(3): 121-128, 1 ago., 2014. ilus, graf
Artículo en Español | IBECS | ID: ibc-125695

RESUMEN

Introducción. El fingolimod, un modulador del receptor de la esfingosina-1-fosfato (S1P) dotado de un mecanismo de acción novedoso, fue el primer tratamiento oral aprobado para la esclerosis múltiple remitente recurrente. Su unión a los receptores S1P1 de los linfocitos promueve la retención selectiva de los linfocitos T vírgenes y de memoria central en los tejidos linfoides secundarios, lo que impide su salida hacia el sistema nervioso central (SNC). Asimismo, el fingolimod atraviesa con facilidad la barrera hematoencefálica, y diversos estudios le atribuyen un efecto neuroprotector directo en el SNC. Objetivo. Revisar la información disponible acerca de los efectos centrales del fingolimod. Desarrollo. El desequilibrio entre los procesos lesivos y reparadores constituye un reflejo de la desmielinización crónica, la degeneración axonal y la gliosis, y parece contribuir a la discapacidad que la esclerosis múltiple acarrea. La facilidad con la que el fingolimod atraviesa la barrera hematoencefálica le permite actuar directamente sobre los receptores S1P localizados en las células del SNC. Una vez en el interior del SNC, ocupa los receptores S1P de los oligodendrocitos y de sus células precursoras, de los astrocitos, los microgliocitos y las neuronas, fomentando la remielinización, la neuroprotección y los procesos endógenos de regeneración. La eficacia evidenciada en los ensayos clínicos concuerda con un mecanismo de acción que incluiría efectos directos sobre las células del SNC. Conclusiones. Los datos disponibles indican que la eficacia del fingolimod en el tratamiento de la esclerosis múltiple se debe a su ambivalencia como molécula inmunomoduladora y moduladora directa de los receptores S1P del SNC. Tanto es así que estudios recientes le atribuyen efectos neuroprotectores en varios modelos que suscitan expectativas en torno a su posible aplicación terapéutica en la enfermedad de Alzheimer, el paludismo cerebral y el neuroblastoma, así como en la neuroprotección frente a la radioterapia craneal (AU)


Introduction. Fingolimod, a sphingosine-1-phosphate receptor modulator, was the first oral therapy approved for relapsingremitting multiple sclerosis, and shows a novel mechanism of action. Upon binding to S1P1 receptors in lymphocytes, the selective retention of naïve and central memory T cells in secondary lymphoid tissues is promoted, preventing their egress to the central nervous system (CNS). In addition, fingolimod readily crosses the blood brain barrier, and several reports suggest a direct neuroprotective effect in the CNS. Aim. To review the available data on the central effects of fingolimod. Development. Imbalances between damage and repair processes are a reflection of chronic demyelination, axonal degeneration and gliosis, and seem to contribute to multiple sclerosis associated disability. Given fingolimod readily crosses the blood brain barrier, it can exert its action directly on S1P receptors present in CNS cells. Fingolimod occupies S1P receptors in oligodendrocytes, oligodendrocyte precursor cells, astrocytes, microglial cells and neurons, promoting remyelination, neuroprotection, and endogenous regeneration processes. Efficacy results from clinical trials are consistent with a mechanism of action that includes direct effects in CNS cells. Conclusions. Current evidence suggests that the efficacy of fingolimod in the treatment of Multiple Sclerosis is due to its dual action as an immunomodulatory molecule and as a direct modulator of S1PRs in the CNS. In fact, recent reports propose that fingolimod has neuroprotective effects in several models, and open new avenues of potential therapeutic applications, such as Alzheimer’s disease, cerebral malaria, neuroblastoma and neuroprotection in cranial irradiation (AU)


Asunto(s)
Humanos , Esfingosina/farmacocinética , Esclerosis Múltiple Recurrente-Remitente/tratamiento farmacológico , Linfocitos T , Astrocitos , Oligodendroglía , Enfermedades Desmielinizantes/fisiopatología , Enfermedades Neurodegenerativas/fisiopatología , Microglía , Fármacos Neuroprotectores/farmacocinética
8.
Br J Clin Pharmacol ; 78(6): 1354-65, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24976291

RESUMEN

AIM: Fingolimod, a sphingosine 1-phosphate receptor modulator, is the first oral disease modifying therapy approved for the treatment of relapsing multiple sclerosis. The aim of this double-blind, placebo-controlled study was to evaluate the effect of fingolimod on cerebral blood flow, platelet function and macular thickness in healthy volunteers. METHODS: The study included 88 healthy volunteers who received fingolimod 0.5 mg or 1.25 mg or matched placebo over a period of 4 weeks. Transcranial colour coded sonography was performed to measure mean blood flow velocities, the platelet function was measured by the PFA-100® assay using a collagen/epinephrine cartridge and macular thickness was measured using optical coherence tomography. An assessment of non-inferiority of fingolimod vs. placebo was performed against a reference value (20% of the overall baseline value). RESULTS: All 88 randomized participants completed the study. At day 28 compared with baseline value, for 0.5 mg, 1.25 mg and placebo treatments, the mean middle cerebral artery blood flow velocity decreased by 4, 1 and 3.7 cm s(-1), respectively. The platelet function analyzer closure time increase was not significant (7.8, 7.5 and 10.4 s, respectively). The mean percentage change in the central foveal thickness from baseline for both eyes was below 3% for all groups. The safety profile of fingolimod in this study was found consistent with the previous reports. CONCLUSIONS: In healthy volunteers, the changes seen with both fingolimod doses were found to be within normal variability, non-inferior and comparable with those observed with placebo for all the pharmacodynamic parameters assessed.


Asunto(s)
Plaquetas/efectos de los fármacos , Circulación Cerebrovascular/efectos de los fármacos , Mácula Lútea/efectos de los fármacos , Glicoles de Propileno/farmacología , Esfingosina/análogos & derivados , Adulto , Velocidad del Flujo Sanguíneo , Plaquetas/fisiología , Método Doble Ciego , Femenino , Clorhidrato de Fingolimod , Humanos , Mácula Lútea/anatomía & histología , Masculino , Persona de Mediana Edad , Glicoles de Propileno/efectos adversos , Glicoles de Propileno/farmacocinética , Receptores de Lisoesfingolípidos/efectos de los fármacos , Esfingosina/efectos adversos , Esfingosina/farmacocinética , Esfingosina/farmacología
9.
Rev Neurol ; 59(3): 121-8, 2014 Aug 01.
Artículo en Inglés, Español | MEDLINE | ID: mdl-25030072

RESUMEN

INTRODUCTION: Fingolimod, a sphingosine-1-phosphate receptor modulator, was the first oral therapy approved for relapsing-remitting multiple sclerosis, and shows a novel mechanism of action. Upon binding to S1P1 receptors in lymphocytes, the selective retention of naive and central memory T cells in secondary lymphoid tissues is promoted, preventing their egress to the central nervous system (CNS). In addition, fingolimod readily crosses the blood brain barrier, and several reports suggest a direct neuroprotective effect in the CNS. AIM: To review the available data on the central effects of fingolimod. DEVELOPMENT: Imbalances between damage and repair processes are a reflection of chronic demyelination, axonal degeneration and gliosis, and seem to contribute to multiple sclerosis associated disability. Given fingolimod readily crosses the blood brain barrier, it can exert its action directly on S1P receptors present in CNS cells. Fingolimod occupies S1P receptors in oligodendrocytes, oligodendrocyte precursor cells, astrocytes, microglial cells and neurons, promoting remyelination, neuroprotection, and endogenous regeneration processes. Efficacy results from clinical trials are consistent with a mechanism of action that includes direct effects in CNS cells. CONCLUSIONS: Current evidence suggests that the efficacy of fingolimod in the treatment of Multiple Sclerosis is due to its dual action as an immunomodulatory molecule and as a direct modulator of S1PRs in the CNS. In fact, recent reports propose that fingolimod has neuroprotective effects in several models, and open new avenues of potential therapeutic applications, such as Alzheimer's disease, cerebral malaria, neuroblastoma and neuroprotection in cranial irradiation.


TITLE: Efectos del fingolimod en el sistema nervioso central.Introduccion. El fingolimod, un modulador del receptor de la esfingosina-1-fosfato (S1P) dotado de un mecanismo de accion novedoso, fue el primer tratamiento oral aprobado para la esclerosis multiple remitente recurrente. Su union a los receptores S1P1 de los linfocitos promueve la retencion selectiva de los linfocitos T virgenes y de memoria central en los tejidos linfoides secundarios, lo que impide su salida hacia el sistema nervioso central (SNC). Asimismo, el fingolimod atraviesa con facilidad la barrera hematoencefalica, y diversos estudios le atribuyen un efecto neuroprotector directo en el SNC. Objetivo. Revisar la informacion disponible acerca de los efectos centrales del fingolimod. Desarrollo. El desequilibrio entre los procesos lesivos y reparadores constituye un reflejo de la desmielinizacion cronica, la degeneracion axonal y la gliosis, y parece contribuir a la discapacidad que la esclerosis multiple acarrea. La facilidad con la que el fingolimod atraviesa la barrera hematoencefalica le permite actuar directamente sobre los receptores S1P localizados en las celulas del SNC. Una vez en el interior del SNC, ocupa los receptores S1P de los oligodendrocitos y de sus celulas precursoras, de los astrocitos, los microgliocitos y las neuronas, fomentando la remielinizacion, la neuroproteccion y los procesos endogenos de regeneracion. La eficacia evidenciada en los ensayos clinicos concuerda con un mecanismo de accion que incluiria efectos directos sobre las celulas del SNC. Conclusiones. Los datos disponibles indican que la eficacia del fingolimod en el tratamiento de la esclerosis multiple se debe a su ambivalencia como molecula inmunomoduladora y moduladora directa de los receptores S1P del SNC. Tanto es asi que estudios recientes le atribuyen efectos neuroprotectores en varios modelos que suscitan expectativas en torno a su posible aplicacion terapeutica en la enfermedad de Alzheimer, el paludismo cerebral y el neuroblastoma, asi como en la neuroproteccion frente a la radioterapia craneal.


Asunto(s)
Fármacos Neuroprotectores/farmacología , Glicoles de Propileno/farmacología , Esfingosina/análogos & derivados , Animales , Barrera Hematoencefálica , Quimiotaxis de Leucocito/efectos de los fármacos , Ensayos Clínicos como Asunto , Modelos Animales de Enfermedad , Clorhidrato de Fingolimod , Humanos , Memoria Inmunológica , Ratones , Ratones Noqueados , Ratones Mutantes Neurológicos , Estudios Multicéntricos como Asunto , Esclerosis Múltiple Recurrente-Remitente/tratamiento farmacológico , Neurogénesis/efectos de los fármacos , Neuroglía/efectos de los fármacos , Fármacos Neuroprotectores/farmacocinética , Glicoles de Propileno/farmacocinética , Glicoles de Propileno/uso terapéutico , Ratas , Receptores de Lisoesfingolípidos/efectos de los fármacos , Esfingosina/farmacocinética , Esfingosina/farmacología , Esfingosina/uso terapéutico , Subgrupos de Linfocitos T/efectos de los fármacos , Subgrupos de Linfocitos T/inmunología
10.
Drug Metab Dispos ; 42(9): 1367-78, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24965813

RESUMEN

A complicating factor in the translational pharmacology of sphingosine 1-phosphate agonists is that they exert their pharmacological effect through their respective phosphate metabolites, which are formed by the enzyme sphingosine kinase (S1PHK). In this investigation, we present a semimechanistic pharmacokinetic model for the interconversion of S1PHK substrates and their respective phosphates in rats and humans with the aim of investigating whether characterization of the rate of phosphorylation in blood platelets constitutes a basis for interspecies scaling using fingolimod as a model compound. Data on the time course of fingolimod and fingolimod-phosphate (fingolimod-P) blood concentrations after intravenous and oral administration of fingolimod and/or fingolimod-P in rats and after oral administration of fingolimod in doses of 0.5, 1.25, and 5 mg once daily in healthy volunteers were analyzed in conjunction with data on the ex vivo interconversion and blood-plasma distribution in rat and human blood, respectively. Integrating the data from the ex vivo and in vivo studies enabled simulation of fingolimod and fingolimod-P concentrations in plasma rather than blood, which are more relevant for characterizing drug effects. Large interspecies differences in the rate of phosphorylation between rats and humans were quantified. In human, phosphorylation of fingolimod in the platelets was four times slower compared with rat, whereas the dephosphorylation rates were comparable in both species. This partly explained the 10-12-fold overprediction of fingolimod-P exposure in human when applying a dose-by-factor approach on the developed rat model. Additionally, differences in presystemic phosphorylation should also be taken into account.


Asunto(s)
Fosforilación/efectos de los fármacos , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Glicoles de Propileno/administración & dosificación , Glicoles de Propileno/farmacocinética , Esfingosina/análogos & derivados , Administración Intravenosa , Administración Oral , Animales , Plaquetas/metabolismo , Método Doble Ciego , Clorhidrato de Fingolimod , Humanos , Lisofosfolípidos/metabolismo , Masculino , Fosfatos/metabolismo , Ensayos Clínicos Controlados Aleatorios como Asunto , Ratas , Ratas Endogámicas Lew , Ratas Sprague-Dawley , Esfingosina/administración & dosificación , Esfingosina/metabolismo , Esfingosina/farmacocinética
11.
Expert Opin Drug Metab Toxicol ; 10(4): 621-30, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24579791

RESUMEN

INTRODUCTION: Fingolimod is a sphingosine 1-phosphate receptor modulator with a novel mechanism of action and the first oral drug approved for the treatment of relapsing forms of multiple sclerosis (MS). Fingolimod reduces relapses more effectively than intramuscular interferon ß1a and delays disability progression. Associated safety risks are bradyarrhythmia and atrioventricular block following the initial dose, requiring monitoring. AREAS COVERED: This article examines the characteristics of fingolimod, its pharmacokinetic properties and the efficacy and tolerability in MS. Information on the pharmacology and mechanisms of action is also provided. EXPERT OPINION: Fingolimod is an effective therapy for relapsing forms of MS in a convenient oral dose. Fingolimod may target not only inflammation but potentially also neurodegeneration. Antagonizing astrocyte sphingosine signaling may help explain the reduction in cerebral atrophy observed in Phase III trials. Long-term data about the safety of fingolimod are needed.


Asunto(s)
Inmunosupresores/farmacocinética , Esclerosis Múltiple Recurrente-Remitente/tratamiento farmacológico , Glicoles de Propileno/farmacocinética , Esfingosina/análogos & derivados , Administración Oral , Clorhidrato de Fingolimod , Humanos , Inmunosupresores/uso terapéutico , Glicoles de Propileno/uso terapéutico , Receptores de Lisoesfingolípidos , Esfingosina/farmacocinética , Esfingosina/uso terapéutico , Resultado del Tratamiento
12.
Ren Fail ; 35(7): 996-1004, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23848241

RESUMEN

Tubulointerstitial fibrosis is a common pathway that leads to kidney failure, and persistent tubulointerstitial inflammation is a key event in the development of tubulointerstitial fibrosis. The new immunosuppressive drug FTY720 modifies lymphocyte migration into injured tissues by sequestering lymphocytes within secondary lymphoid organs. However, its therapeutic effect on tubulointerstitial inflammation and fibrosis had not been well understood. This study was designed to explore the effect of FTY720 on tubulointerstitial inflammation and fibrosis in subtotally nephrectomized (SNX) rats. In total, 24 male Sprague-Dawley rats were used. Seven days after 5/6 nephrectomy, rats were randomized to FTY720 (1 mg/kg/d) and placebo-treated groups. Sham-operated rats served as controls. FTY720 significantly attenuated the rise in proteinuria, serum creatinine, urea nitrogen and N-acetyl-ß-D-glucosaminidase activity in SNX rats, and reduced the count of peripheral white blood cells and lymphocytes in SNX rats. Morphological analysis revealed that there was severe tubulointerstitial inflammation and fibrosis in SNX group and much more tubulointerstitial infiltrating inflammatory cells with high expression of CD3, CD4, CD8, CD20, CD68, CD163 and CCR-7 in SNX group, as compared with the controls, but the lesions were attenuated significantly by treatment with FTY720. Furthermore, the expressions of proinflammatory molecules (IL-6, TNF-α and MCP-1), profibrotic molecule (TGF-ß1) and production of extracellular matrix proteins such as fibronectin and types I and III collagens were upregulated in SNX rats. FTY720 administration significantly reduced these abnormalities. In summary, FTY720 exerts therapeutic effects on tubulointerstitial fibrosis in SNX rats by inhibiting the tubulointerstitial inflammatory response.


Asunto(s)
Fibrosis , Nefritis Intersticial , Glicoles de Propileno/farmacocinética , Insuficiencia Renal , Esfingosina/análogos & derivados , Animales , Movimiento Celular/efectos de los fármacos , Quimiocina CCL2/metabolismo , Modelos Animales de Enfermedad , Fibrosis/etiología , Fibrosis/inmunología , Fibrosis/patología , Clorhidrato de Fingolimod , Inmunosupresores/farmacología , Interleucina-6/metabolismo , Túbulos Renales/efectos de los fármacos , Túbulos Renales/inmunología , Túbulos Renales/patología , Linfocitos/efectos de los fármacos , Linfocitos/fisiología , Masculino , Nefrectomía/métodos , Nefritis Intersticial/complicaciones , Nefritis Intersticial/tratamiento farmacológico , Nefritis Intersticial/inmunología , Ratas , Ratas Sprague-Dawley , Insuficiencia Renal/etiología , Insuficiencia Renal/inmunología , Insuficiencia Renal/patología , Esfingosina/farmacocinética , Factor de Crecimiento Transformador beta1/metabolismo , Resultado del Tratamiento , Factor de Necrosis Tumoral alfa/metabolismo
13.
Antibiot Khimioter ; 58(11-12): 38-42, 2013.
Artículo en Ruso | MEDLINE | ID: mdl-24734428

RESUMEN

The data on the mechanism of action, efficacy and safety of the drug fingolimod are presented. Further study of the molecular, biochemical and cellular mechanisms of action of pharmacological regulators of phospholipid mediators' signal transduction is an important basis for developing new immunomodulators and antitumor agents.


Asunto(s)
Antineoplásicos , Inmunosupresores , Fosfolípidos/metabolismo , Glicoles de Propileno , Transducción de Señal/efectos de los fármacos , Esfingosina/análogos & derivados , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Clorhidrato de Fingolimod , Humanos , Inmunosupresores/farmacocinética , Inmunosupresores/farmacología , Glicoles de Propileno/farmacocinética , Glicoles de Propileno/farmacología , Esfingosina/farmacocinética , Esfingosina/farmacología
14.
Int J Clin Pharmacol Ther ; 50(8): 540-4, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22735460

RESUMEN

BACKGROUND: Fingolimod has a novel mechanism of action in multiple sclerosis, being a first-in-class sphingosine 1-phosphate receptor modulator. Because of a potential risk of fetal toxicity based on animal studies, women of childbearing potential are advised to take effective contraceptive measures during and for 2 months after stopping fingolimod therapy. To assess whether the efficacy of a combined oral contraceptive (OC) could be compromised during fingolimod therapy, a steady-state, drug-drug interaction study of fingolimod with ethinylestradiol/levonorgestrel was performed in healthy female volunteers. OBJECTIVE: To assess the interaction between fingolimod 0.5 mg once daily and ethinylestradiol 30 µg/ levonorgestrel 150 µg once daily at a steady state. METHODS: 31 healthy women received the combined OC only on Days 1 - 14, followed by OC plus fingolimod on Days 15 - 28. RESULTS: In the presence of fingolimod, ethinylestradiol pharmacokinetics were unchanged, and levonorgestrel maximum plasma concentration at steady state and area under the concentration-time curve during a dosing interval increased by factors of 1.10 (90% CI 1.05 - 1.16) and 1.22 (90% CI 1.18 - 1.27), respectively. CONCLUSIONS: Fingolimod therapy does not alter the pharmacokinetics of the combined OC ethinylestradiol/ levonorgestrel to a clinically significant degree. Ethinylestradiol/levonorgestrel does not alter the pharmacokinetics of fingolimod. Women receiving fingolimod therapy are able to use a combined OC as a means of effective birth control.


Asunto(s)
Anticonceptivos Orales Combinados/farmacocinética , Interacciones Farmacológicas , Etinilestradiol/farmacocinética , Inmunosupresores/farmacocinética , Levonorgestrel/farmacocinética , Glicoles de Propileno/farmacocinética , Esfingosina/análogos & derivados , Adolescente , Adulto , Área Bajo la Curva , Anticonceptivos Orales Combinados/administración & dosificación , Combinación de Medicamentos , Etinilestradiol/administración & dosificación , Femenino , Clorhidrato de Fingolimod , Humanos , Inmunosupresores/administración & dosificación , Levonorgestrel/administración & dosificación , Glicoles de Propileno/administración & dosificación , Esfingosina/administración & dosificación , Esfingosina/farmacocinética
15.
Anal Bioanal Chem ; 403(7): 1897-905, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22538778

RESUMEN

We developed a liquid chromatography/electrospray ionization tandem mass spectrometry method for the simultaneous quantitative determination of C18 sphingosine (Sph), C18 dihydrosphingosine (dhSph), C18 phytosphingosine (pSph), C18 sphingosine-1-phosphate (S1P), C18 dihydrosphingosine-1-phosphate (dhS1P), and C18 phytosphingosine-1-phosphate (pS1P). Samples were prepared by simple methanol deproteinization and analyzed in selected reaction monitoring modes. No peak tailing was observed on the chromatograms using a Capcell Pak ACR column (1.5 mm i.d. × 250 mm, 3 µm, Shiseido). The calibration curves of the sphingoids showed good linearity (r > 0.996) over the range of 0.050-5.00 pmol per injection. The accuracy and precision of this method were demonstrated using four representative biological samples (serum, brain, liver, and spleen) from mice that contained known amounts of the sphingoids. Samples of mice tissue such as plasma, brain, eye, testis, liver, kidney, lung, spleen, lymph node, and thymus were examined for their Sph, dhSph, pSph, S1P, dhS1P, and pS1P composition. The results confirmed the usefulness of this method for the physiological and pathological analysis of the composition of important sphingoids.


Asunto(s)
Cromatografía Liquida/métodos , Espectrometría de Masa por Ionización de Electrospray/métodos , Esfingosina/análisis , Espectrometría de Masas en Tándem/métodos , Animales , Calibración , Ratones , Reproducibilidad de los Resultados , Esfingosina/química , Esfingosina/farmacocinética , Distribución Tisular
16.
Xenobiotica ; 42(7): 671-86, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22225501

RESUMEN

Sphingosine-1-phosphate (S1P(1)) receptor agonists such as Fingolimod (FTY-720) are a novel class of immunomodulators that have clinical utility in the treatment of remitting relapsing multiples sclerosis. This class of compound act by inducing peripheral lymphopenia. Using an integrated pharmacokinetic/pharmacodynamic (PK-PD) approach based on an in vivo rat model, novel S1P(1) agonists were identified with a predicted more rapid rate of reversibility of lymphocyte reduction in human compared to Fingolimod. The in vivo potency of 15 compounds based on PK-PD modelling of the rat lymphocyte reduction model was correlated with in vitro measures of potency at the S1P(1) receptor using ß arrestin recruitment and G-protein signalling. A structurally novel S1P(1) agonist was identified and predictions of human pharmacokinetics and clinical dose are presented.


Asunto(s)
Lisofosfolípidos/agonistas , Glicoles de Propileno/farmacocinética , Esfingosina/análogos & derivados , Animales , Arrestina/metabolismo , Clorhidrato de Fingolimod , Proteínas de Unión al GTP/metabolismo , Linfocitos/efectos de los fármacos , Linfocitos/metabolismo , Lisofosfolípidos/metabolismo , Masculino , Esclerosis Múltiple Recurrente-Remitente/tratamiento farmacológico , Glicoles de Propileno/farmacología , Glicoles de Propileno/uso terapéutico , Ratas , Ratas Endogámicas , Transducción de Señal , Esfingosina/agonistas , Esfingosina/metabolismo , Esfingosina/farmacocinética , Esfingosina/farmacología , Esfingosina/uso terapéutico
17.
J Clin Pharmacol ; 52(12): 1879-90, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22174429

RESUMEN

Fingolimod, a first-in-class oral sphingosine 1-phosphate receptor (S1PR) modulator, is approved in many countries for relapsing-remitting multiple sclerosis, at a once-daily 0.5-mg dose. A reduction in peripheral lymphocyte count is an expected consequence of the fingolimod mechanism of S1PR modulation. The authors investigated if this pharmacodynamic effect impacts humoral and cellular immunogenicity. In this double-blind, parallel-group, 4-week study, 72 healthy volunteers were randomized to steady state, fingolimod 0.5 mg, 1.25 mg, or to placebo. The authors compared T-cell dependent and independent responses to the neoantigens, keyhole limpet hemocyanin (KLH), and pneumococcal polysaccharides vaccine (PPV-23), respectively, and additionally recall antigen response (tetanus toxoid [TT]) and delayed-type hypersensitivity (DTH) to KLH, TT, and Candida albicans. Fingolimod caused mild to moderate decreases in anti-KLH and anti-PPV-23 IgG and IgM levels versus placebo. Responder rates were identical between placebo and 0.5-mg groups for anti-KLH IgG (both > 90%) and comparable for anti-PPV-23 IgG (55% and 41%, respectively). Fingolimod did not affect anti-TT immunogenicity, and DTH response did not differ between placebo and fingolimod 0.5-mg groups. Expectedly, lymphocyte count reduced substantially in the fingolimod groups versus placebo but reversed by study end. Fingolimod was well tolerated, and the observed safety profile was consistent with previous reports.


Asunto(s)
Inmunoglobulina G/sangre , Inmunoglobulina M/sangre , Inmunosupresores/administración & dosificación , Glicoles de Propileno/administración & dosificación , Esfingosina/análogos & derivados , Linfocitos T/efectos de los fármacos , Administración Oral , Adulto , Antígenos/administración & dosificación , Antígenos Fúngicos/administración & dosificación , Candida albicans/inmunología , Método Doble Ciego , Femenino , Clorhidrato de Fingolimod , Hemocianinas/administración & dosificación , Humanos , Hipersensibilidad Tardía/etiología , Hipersensibilidad Tardía/inmunología , Inmunosupresores/sangre , Inmunosupresores/farmacocinética , Masculino , Vacunas Neumococicas/administración & dosificación , Glicoles de Propileno/sangre , Glicoles de Propileno/farmacocinética , Esfingosina/administración & dosificación , Esfingosina/sangre , Esfingosina/farmacocinética , Linfocitos T/inmunología , Toxoide Tetánico/administración & dosificación , Adulto Joven
18.
J Clin Pharmacol ; 52(7): 1054-68, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22110161

RESUMEN

Fingolimod (FTY720) is a sphingosine 1-phosphate receptor (S1PR) modulator currently being evaluated for the treatment of multiple sclerosis. Fingolimod undergoes phosphorylation in vivo to yield fingolimod phosphate (fingolimod-P), which modulates S1PRs expressed on lymphocytes and cells in the central nervous system. The authors developed a population model, using pooled data from 7 phase 1 studies, to enable characterization of fingolimod-P pharmacokinetics following oral administration of fingolimod and to evaluate the impact of key demographic variables on exposure. The fingolimod-P concentration-time course after either single or multiple doses of fingolimod was described by a 2-compartment model with first-order apparent formation and elimination, lag time in the apparent formation, and dose-dependent relative bioavailability and apparent central volume of distribution. Body weight and ethnicity were identified as demographic covariates correlated with the disposition of fingolimod-P. Model predictions indicated no need for dose adjustment of fingolimod based on body weight; the effect of ethnicity on the disposition of fingolimod requires further investigation. The accurate prediction of the pharmacokinetic profile of fingolimod-P determined empirically in 2 large phase 3 trials provides external validation of the model.


Asunto(s)
Inmunosupresores/farmacocinética , Modelos Biológicos , Glicoles de Propileno/farmacocinética , Esfingosina/análogos & derivados , Administración Oral , Adulto , Disponibilidad Biológica , Ensayos Clínicos Fase I como Asunto , Ensayos Clínicos Fase III como Asunto , Relación Dosis-Respuesta a Droga , Femenino , Clorhidrato de Fingolimod , Humanos , Inmunosupresores/administración & dosificación , Masculino , Persona de Mediana Edad , Fosforilación , Glicoles de Propileno/administración & dosificación , Receptores de Lisoesfingolípidos/efectos de los fármacos , Receptores de Lisoesfingolípidos/metabolismo , Esfingosina/administración & dosificación , Esfingosina/farmacocinética , Factores de Tiempo , Distribución Tisular , Adulto Joven
19.
Clin Pharmacokinet ; 51(1): 15-28, 2012 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-22149256

RESUMEN

Fingolimod (FTY720), a sphingosine 1-phosphate receptor modulator, is the first in a new class of therapeutic compounds and is the first oral therapy approved for the treatment of relapsing forms of multiple sclerosis (MS). Fingolimod is a structural analogue of endogenous sphingosine and undergoes phosphorylation to produce fingolimod phosphate, the active moiety. Fingolimod targets MS via effects on the immune system, and evidence from animal models indicates that it may also have actions in the central nervous system. In phase III studies in patients with relapsing-remitting MS, fingolimod has demonstrated efficacy superior to that of an approved first-line therapy, intramuscular interferon-ß-1a, as well as placebo, with benefits extending across clinical and magnetic resonance imaging measures. The pharmacokinetic profiles of fingolimod and fingolimod phosphate have been extensively investigated in studies in healthy volunteers, renal transplant recipients (the indication for which fingolimod was initially under clinical development, but the development was subsequently discontinued) and MS patients. Results from these studies have demonstrated that fingolimod is efficiently absorbed, with an oral bioavailability of >90%, and its absorption is unaffected by dietary intake, therefore it can be taken without regard to meals. Fingolimod and fingolimod phosphate have a half-life of 6-9 days, and steady-state pharmacokinetics are reached after 1-2 months of daily dosing. The long half-life of fingolimod, together with its slow absorption, means that fingolimod has a flat concentration profile over time with once-daily dosing. Fingolimod and fingolimod phosphate show dose-proportional exposure in single- and multiple-dose studies over a range of 0.125-5 mg; hence, there is a predictable relationship between dose and systemic exposure. Furthermore, fingolimod and fingolimod phosphate exhibit low to moderate intersubject pharmacokinetic variability. Fingolimod is extensively metabolized, with biotransformation occurring via three main pathways: (i) reversible phosphorylation to fingolimod phosphate; (ii) hydroxylation and oxidation to yield a series of inactive carboxylic acid metabolites; and (iii) formation of non-polar ceramides. Fingolimod is largely cleared through metabolism by cytochrome P450 (CYP) 4F2. Since few drugs are metabolized by CYP4F2, fingolimod would be expected to have a relatively low potential for drug-drug interactions. This is supported by data from in vitro studies indicating that fingolimod and fingolimod phosphate have little or no capacity to inhibit and no capacity to induce other major drug-metabolizing CYP enzymes at therapeutically relevant steady-state blood concentrations. Population pharmacokinetic evaluations indicate that CYP3A inhibitors and CYP3A inducers have no effect or only a weak effect on the pharmacokinetics of fingolimod and fingolimod phosphate. However, blood concentrations of fingolimod and fingolimod phosphate are increased moderately when fingolimod is coadministered with ketoconazole, an inhibitor of CYP4F2. The pharmacokinetics of fingolimod are unaffected by renal impairment or mild-to-moderate hepatic impairment. However, exposure to fingolimod is increased in patients with severe hepatic impairment. No clinically relevant effects of age, sex or ethnicity on the pharmacokinetics of fingolimod have been observed. Fingolimod is thus a promising new therapy for eligible patients with MS, with a predictable pharmacokinetic profile that allows effective once-daily oral dosing.


Asunto(s)
Inmunosupresores/farmacocinética , Glicoles de Propileno/farmacocinética , Esfingosina/análogos & derivados , Animales , Proteínas Sanguíneas/metabolismo , Interacciones Farmacológicas , Clorhidrato de Fingolimod , Humanos , Inmunosupresores/uso terapéutico , Enfermedades Renales/metabolismo , Hepatopatías/metabolismo , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/metabolismo , Glicoles de Propileno/uso terapéutico , Unión Proteica , Esfingosina/farmacocinética , Esfingosina/uso terapéutico
20.
Int J Clin Pract ; 65(8): 887-95, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21679286

RESUMEN

Fingolimod, a sphingosine 1-phosphate receptor modulator, is the first oral treatment approved by the US Food and Drug Administration for the treatment of relapsing forms of multiple sclerosis (MS). The aim of this review was to provide a concise, comprehensive overview of the clinically relevant mechanism of action, efficacy and safety information available for fingolimod. Key data were derived from two international, Phase III, double-blind, randomised trials (TRANSFORMS and FREEDOMS) performed over 12 and 24 months, respectively, which evaluated fingolimod 0.5 and 1.25 mg daily in 1703 patients with relapsing forms of MS. In TRANSFORMS, there was a 52% reduction in the annualised relapse rate (ARR) with fingolimod 0.5 mg vs. 30 µg intramuscular interferon beta-1a (0.16 vs. 0.33; p < 0.001) at 1 year. In FREEDOMS, there was a 55% decrease in ARR at 2 years with fingolimod 0.5 mg vs. placebo (0.18 vs. 0.40; p < 0.001). Risk of disability progression, confirmed at 3 months, was also reduced by 30% over the 2-year study period with fingolimod vs. placebo (p = 0.02). Significantly fewer new or enlarged lesions on T(2) -weighted images were seen in both studies (TRANSFORMS, p = 0.002 vs. interferon beta-1a at 1 year; FREEDOMS, p < 0.001 vs. placebo at 2 years). Overall, fingolimod 0.5 mg was well tolerated by patients. Transient, generally asymptomatic bradycardia and infrequent atrioventricular block were seen with the administration of the first dose. Macular oedema and serious infections occurred infrequently. Reversible, asymptomatic elevations of liver enzymes could also occur. As the first approved oral disease-modifying treatment, fingolimod offers patients a convenient alternative to regular self-injection for the treatment of relapsing forms of MS. In addition to high efficacy with a relatively acceptable safety profile, fingolimod provides a therapy with a new mechanism of action.


Asunto(s)
Inmunosupresores/administración & dosificación , Esclerosis Múltiple Recurrente-Remitente/tratamiento farmacológico , Glicoles de Propileno/administración & dosificación , Esfingosina/análogos & derivados , Administración Oral , Adulto , Ensayos Clínicos Fase III como Asunto , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Clorhidrato de Fingolimod , Humanos , Inmunosupresores/efectos adversos , Inmunosupresores/farmacocinética , Persona de Mediana Edad , Estudios Multicéntricos como Asunto , Glicoles de Propileno/efectos adversos , Glicoles de Propileno/farmacocinética , Ensayos Clínicos Controlados Aleatorios como Asunto , Receptores de Lisoesfingolípidos/efectos de los fármacos , Factores de Riesgo , Prevención Secundaria , Esfingosina/administración & dosificación , Esfingosina/efectos adversos , Esfingosina/farmacocinética , Resultado del Tratamiento , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...