Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 101
Filtrar
1.
Mult Scler ; 24(12): 1605-1616, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-28911260

RESUMEN

BACKGROUND: Amiselimod, an oral selective sphingosine-1-phosphate 1 receptor modulator, suppressed disease activity dose-dependently without clinically relevant bradyarrhythmia in a 24-week phase 2, placebo-controlled study in relapsing-remitting multiple sclerosis. OBJECTIVE: To assess safety and efficacy of amiselimod over 96 weeks. METHODS: After completing the core study, patients on amiselimod continued at the same dose, whereas those on placebo were randomised 1:1:1 to amiselimod 0.1, 0.2 or 0.4 mg for another 72 weeks. Most patients receiving 0.1 mg were re-randomised to 0.2 or 0.4 mg upon availability of the core study results. RESULTS: Of 415 patients randomised in the core study, 367 (88.4%) entered and 322 (77.6%) completed the extension. One or more adverse events were reported in 303 (82.6%) of 367 patients: 'headache', 'lymphocyte count decreased', 'nasopharyngitis' and 'MS relapse' were most common (14.7%-16.9%). No serious opportunistic infection, macular oedema or malignancy was reported and no bradyarrhythmia of clinical concern was observed by Holter or 12-lead electrocardiogram. The dose-dependent effect of amiselimod on clinical and magnetic resonance imaging-related outcomes from the core study was sustained in those continuing on amiselimod and similarly observed after switching to active drug. CONCLUSION: For up to 2 years of treatment, amiselimod was well tolerated and dose-dependently effective in controlling disease activity.


Asunto(s)
Esclerosis Múltiple Recurrente-Remitente/tratamiento farmacológico , Propanolaminas/administración & dosificación , Propanolaminas/efectos adversos , Adulto , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Femenino , Humanos , Masculino , Persona de Mediana Edad , Receptores de Lisoesfingolípidos/efectos de los fármacos , Tiempo , Resultado del Tratamiento
2.
Bioorg Med Chem Lett ; 28(19): 3255-3259, 2018 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-30143424

RESUMEN

The oral S1PR1 agonist ponesimod demonstrated substantial efficacy in a phase II clinical trial of psoriasis. Unfortunately, systemic side effects were observed, which included lymphopenia and transient bradycardia. We sought to develop a topical soft-drug S1PR1 agonist with an improved therapeutic index. By modifying ponesimod, we discovered an ester series of S1PR agonists. To increase metabolic instability in plasma we synthesised esters described as specific substrates for paraoxonase and butyrylcholinesterases, esterases present in human plasma.


Asunto(s)
Descubrimiento de Drogas , Receptores de Lisoesfingolípidos/efectos de los fármacos , Tiazoles/farmacología , Administración Tópica , Arildialquilfosfatasa/sangre , Cromatografía Líquida de Alta Presión , Cromatografía de Fase Inversa , Esterasas/sangre , Esterasas/metabolismo , Humanos , Piel/enzimología , Solubilidad , Receptores de Esfingosina-1-Fosfato , Relación Estructura-Actividad , Tiazoles/administración & dosificación
3.
Eur J Clin Pharmacol ; 74(12): 1593-1604, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30105453

RESUMEN

PURPOSE: To assess the potential pharmacokinetic (PK) interactions between siponimod and rifampin, a strong CYP3A4/moderate CYP2C9 inducer, in healthy subjects. METHODS: This was a confirmatory, open-label, multiple-dose two-period study in healthy subjects (aged 18-45 years). In Period 1 (Days 1-12), siponimod was up-titrated from 0.25 to 2 mg over 5 days (Days 1-6) followed by 2 mg once daily on days 7-12. In Period 2, siponimod 2 mg qd was co-administered with rifampin 600 mg qd (Days 13-24). Primary assessments included PK of siponimod (Days 12 and 24; maximum steady-state plasma concentration [Cmax,ss], median time to achieve Cmax,ss [Tmax, ss], and area under the curve at steady state [AUCtau,ss]). Key secondary assessments were PK of M3 and M5 metabolites, and safety/tolerability including absolute lymphocyte count (ALC). RESULTS: Of the 16 subjects enrolled (age, mean ± standard deviation [SD] 31 ± 8.3 years; men, n = 15), 15 completed the study. In Period 1, siponimod geometric mean Cmax,ss (28.6 ng/mL) was achieved in 4 h (median Tmax,ss; range, 1.58-8.00) and the geometric mean AUCtau,ss was 546 h × ng/mL. In Period 2, the siponimod geometric mean Cmax,ss and AUCtau,ss decreased to 15.7 ng/mL and 235 h × ng/mL, respectively; median Tmax remained unchanged (4 h). Rifampin co-administration increased M3 Cmax,ss by 53% while M5 Cmax,ss remained unchanged. The AUCtau,ss of M3 and M5 decreased by 10% and 37%, respectively. The majority of adverse events reported were mild, with a higher frequency during Period 2 (86.7%) versus Period 1 (50%). The mean ALC increased slightly under rifampin co-administration but remained below 1.0 × 109/L. CONCLUSIONS: The study findings suggest that in the presence of rifampin, a strong CYP3A4/moderate CYP2C9 inducer, siponimod showed significant decrease in Cmax,ss (45%) and AUCtau,ss (57%) in healthy subjects.


Asunto(s)
Azetidinas/farmacocinética , Compuestos de Bencilo/farmacocinética , Citocromo P-450 CYP2C9/biosíntesis , Receptores de Lisoesfingolípidos/efectos de los fármacos , Rifampin/farmacocinética , Adolescente , Adulto , Área Bajo la Curva , Azetidinas/efectos adversos , Compuestos de Bencilo/efectos adversos , Biotransformación , Interacciones Farmacológicas , Inducción Enzimática/efectos de los fármacos , Femenino , Voluntarios Sanos , Humanos , Recuento de Linfocitos , Masculino , Rifampin/efectos adversos , Adulto Joven
4.
Br J Clin Pharmacol ; 83(5): 1011-1027, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-27921320

RESUMEN

AIM: Amiselimod (MT-1303) is a selective sphingosine 1-phosphate 1 (S1P1 ) receptor modulator which is currently being developed for the treatment of various autoimmune diseases. Unlike some other S1P receptor modulators, amiselimod seemed to show a favourable cardiac safety profile in preclinical, phase I and II studies. The aim of the current study was to characterize the cardiac effects of amiselimod by directly comparing it with fingolimod and placebo. METHODS: A total of 81 healthy subjects aged 18-55 years were equally randomized to receive amiselimod 0.4 mg, amiselimod 0.8 mg, placebo or fingolimod 0.5 mg once daily for 28 days. The chronotropic/dromotropic and inotropic effects were evaluated using intensive Holter electrocardiogram and echocardiography. RESULTS: Unlike fingolimod, neither amiselimod dose exerted acute (1-6 h) negative chronotropic effects on Days 1 and 2. The lowest nadir mean hourly heart rate was observed on Day 14 in the amiselimod 0.4 mg group (least squares mean difference: -4.40 bpm, 95% confidence interval -7.15, -1.66) and Day 7 in the 0.8 mg group [-3.85 bpm (-6.58, -1.11)] compared with placebo, but these changes were smaller than those with fingolimod on Day 1 [-6.49 bpm (-8.95, -4.02)]. No clinically significant bradyarrhythmia or cardiac functional abnormalities were observed in either amiselimod group. Both amiselimod doses were well tolerated and no serious adverse events were reported. Fingolimod was also generally well tolerated, although one subject was withdrawn owing to highly frequent 2:1 atrioventricular blocks on Day 1. CONCLUSION: The study demonstrated a more favourable cardiac safety profile for amiselimod than fingolimod when administered over 28 days in healthy subjects.


Asunto(s)
Clorhidrato de Fingolimod/efectos adversos , Inmunosupresores/efectos adversos , Propanolaminas/efectos adversos , Receptores de Lisoesfingolípidos/efectos de los fármacos , Adulto , Bloqueo Atrioventricular/etiología , Relación Dosis-Respuesta a Droga , Ecocardiografía , Electrocardiografía Ambulatoria , Clorhidrato de Fingolimod/administración & dosificación , Frecuencia Cardíaca/efectos de los fármacos , Humanos , Inmunosupresores/administración & dosificación , Masculino , Persona de Mediana Edad , Propanolaminas/administración & dosificación , Receptores de Lisoesfingolípidos/metabolismo , Método Simple Ciego , Adulto Joven
5.
Biochim Biophys Acta ; 1851(5): 519-26, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25601519

RESUMEN

Transforming growth factor ß2 (TGF-ß2) is well known to stimulate the expression of pro-fibrotic connective tissue growth factor (CTGF) in several cell types including human mesangial cells. The present study demonstrates that TGF-ß2 enhances sphingosine 1-phosphate receptor 5 (S1P5) mRNA and protein expression in a time and concentration dependent manner. Pharmacological and siRNA approaches reveal that this upregulation is mediated via activation of classical TGF-ß downstream effectors, Smad and mitogen-activated protein kinases. Most notably, inhibition of Gi with pertussis toxin and downregulation of S1P5 by siRNA block TGF-ß2-stimulated upregulation of CTGF, demonstrating that Gi coupled S1P5 is necessary for TGF-ß2-triggered expression of CTGF in human mesangial cells. Overall, these findings indicate that TGF-ß2 dependent upregulation of S1P5 is required for the induction of pro-fibrotic CTGF by TGF-ß. Targeting S1P5 might be an attractive novel approach to treat renal fibrotic diseases.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Células Mesangiales/efectos de los fármacos , Receptores de Lisoesfingolípidos/efectos de los fármacos , Factor de Crecimiento Transformador beta2/farmacología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/antagonistas & inhibidores , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Humanos , Células Mesangiales/metabolismo , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Interferencia de ARN , ARN Mensajero/metabolismo , Receptores de Lisoesfingolípidos/genética , Receptores de Lisoesfingolípidos/metabolismo , Proteína Smad4/genética , Proteína Smad4/metabolismo , Factores de Tiempo , Transfección , Regulación hacia Arriba
6.
Biochim Biophys Acta ; 1851(2): 194-202, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25457224

RESUMEN

The matricellular protein connective tissue growth factor (CTGF/CCN2) is recognized as key player in the onset of fibrosis in various tissues, including skeletal muscle. In many circumstances, CTGF has been shown to be induced by transforming growth factor beta (TGFß) and accounting, at least in part, for its biological action. In this study it was verified that in cultured myoblasts CTGF/CCN2 causes their transdifferentiation into myofibroblasts by up-regulating the expression of fibrosis marker proteins α-smooth muscle actin and transgelin. Interestingly, it was also found that the profibrotic effect exerted by CTGF/CCN2 was mediated by the sphingosine kinase (SK)-1/S1P3 signaling axis specifically induced by the treatment with the profibrotic cue. Following CTGF/CCN2-induced up-regulation, S1P3 became the S1P receptor subtype expressed at the highest degree, at least at mRNA level, and was thus capable of readdressing the sphingosine 1-phosphate signaling towards fibrosis rather than myogenic differentiation. Another interesting finding is that CTGF/CCN2 silencing prevented the TGFß-dependent up-regulation of SK1/S1P3 signaling axis and strongly reduced the profibrotic effect exerted by TGFß, pointing at a crucial role of endogenous CTGF/CCN2 generated following TGFß challenge in the transmission of at least part of its profibrotic effect. These results provide new insights into the molecular mechanism by which CTGF/CCN2 drives its biological action and strengthen the concept that SK1/S1P3 axis plays a critical role in the onset of fibrotic cell phenotype.


Asunto(s)
Transdiferenciación Celular , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Mioblastos Esqueléticos/enzimología , Miofibroblastos/enzimología , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Receptores de Lisoesfingolípidos/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Línea Celular , Factor de Crecimiento del Tejido Conjuntivo/genética , Factor de Crecimiento del Tejido Conjuntivo/farmacología , Relación Dosis-Respuesta a Droga , Fibrosis , Ratones , Mioblastos Esqueléticos/efectos de los fármacos , Mioblastos Esqueléticos/patología , Miofibroblastos/efectos de los fármacos , Miofibroblastos/patología , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Interferencia de ARN , ARN Mensajero/metabolismo , Receptores de Lisoesfingolípidos/efectos de los fármacos , Receptores de Lisoesfingolípidos/genética , Proteínas Recombinantes/farmacología , Receptores de Esfingosina-1-Fosfato , Factores de Tiempo , Transfección , Factor de Crecimiento Transformador beta1/farmacología , Regulación hacia Arriba
7.
Beijing Da Xue Xue Bao Yi Xue Ban ; 48(6): 987-993, 2016 12 18.
Artículo en Zh | MEDLINE | ID: mdl-27987502

RESUMEN

OBJECTIVE: To construct sphingosine 1-phosphate receptor-1 (S1P1)-small interfering RNA (siRNA) lentiviral vectors and infect human salivary gland cells (HSG), and to investigate its possible therapy on Sjogren's syndrome. METHODS: HSG cells were divided into blank group, empty vector group, scramble-siRNA group and S1P1-siRNA group. The lentiviral vectors expressing siRNA against S1P1 and the pLL3.7 were respectively transfected into 293T cells with pMD2.G, pMDL g/p RRE, pRSV-REV to produce virus, and then infect HSG cells. The efficiency was observed by flow cytometry after the transfection for 48 h. The expression levels of S1P1 mRNA of HSG were detected by real-time RT-PCR and the expression of S1P1 protein was detected by immunohistochemistry method. The expression levels of interferon-γ (IFN-γ) and interleukin (IL)-17 in the supernatant of the cells were detected by ELISA method. RESULTS: (1) The scramble-siRNA, S1P1-siRNA lentiviral vector was successfully constructed, and the lentivirus titer was about 3.5×108 TU/mL. (2) The level of S1P1 mRNA was lower in S1P1-siRNA group than those in the blank group, empty vector group, and scramble-siRNA group 48 h after infection, there were significant differences between them (P<0.05). (3) The expression of S1P1 protein was lower in S1P1-siRNA group than those in blank group, empty vector group, and scramble-siRNA group 48 h after transfection, there were significant differences between them (P<0.05). (4) The levels of IL-17 were lower in S1P1-siRNA group than those in blank group, empty vector group, and scramble-siRNA group 48 h after transfection, there were significant differences between them (P<0.05). (5) The levels of IFN-γ in S1P1-siRNA group were lower than those in blank group, empty vector group, and scramble-siRNA group 48 h after transfection, there were significant differences between them (P<0.05). CONCLUSION: The lentiviral vector targeting S1P1 was successfully constructed. S1P1 siRNA could suppress the levels of S1P1 mRNA and protein, and decrease the expression of IL-17 and IFN-γ. S1P1 siRNA could infect HSG cells stably and inhibit the expression of S1P1 gene specifically and efficiently, and reduce the levels of inflammatory cytokines.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Vectores Genéticos/farmacología , ARN Interferente Pequeño/biosíntesis , ARN Interferente Pequeño/farmacología , Receptores de Lisoesfingolípidos/efectos de los fármacos , Receptores de Lisoesfingolípidos/genética , Receptores de Lisoesfingolípidos/fisiología , Transfección/métodos , Citocinas , Vectores Genéticos/administración & dosificación , Vectores Genéticos/biosíntesis , Humanos , Técnicas In Vitro/métodos , Interferón gamma/efectos de los fármacos , Interferón gamma/genética , Interleucina-17/genética , Lentivirus , ARN Mensajero , Glándulas Salivales/efectos de los fármacos
8.
Bioorg Med Chem ; 23(5): 1011-26, 2015 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-25656338

RESUMEN

Sphingosine-1-phosphate (S1P) influences various fundamental biological processes by interacting with a family of five G protein-coupled receptors (S1P1-5). FTY720, a sphingosine analogue, which was approved for treatment of relapsing forms of multiple sclerosis, is phosphorylated in vivo and acts as an agonist of four of the five S1P receptor subtypes. Starting from these lead structures we developed new agonists for the S1P1 receptor. The biological activity was tested in vivo and promising ligands were fluorinated at different positions to identify candidates for positron emission tomography (PET) imaging after [(18)F]-labelling. The radioligands shall enable the imaging of S1P1 receptor expression in vivo and thus may serve as novel imaging markers of S1P-related diseases.


Asunto(s)
Clorhidrato de Fingolimod/síntesis química , Clorhidrato de Fingolimod/farmacología , Receptores de Lisoesfingolípidos/efectos de los fármacos , Animales , Células CHO , Cricetinae , Cricetulus , Clorhidrato de Fingolimod/química , Humanos , Inmunosupresores/síntesis química , Inmunosupresores/química , Inmunosupresores/farmacología , Ligandos , Ratones , Receptores de Lisoesfingolípidos/metabolismo
9.
Xenobiotica ; 45(12): 1063-80, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26084376

RESUMEN

1. Disposition and metabolism of CS-0777 (1-{5-[(3R)-3-amino-4-hydroxy-3- methylbutyl]-1-methyl-1H-pyrrol-2-yl}-4-(4-methylphenyl) butan-1-one), a selective sphingosine 1-phosphate receptor-1 modulator under development for autoimmune conditions was investigated following oral and/or i.v. bolus administration to rats and monkeys. 2. After oral administration of [14C]CS-0777, CS-0777 was well absorbed in rats and monkeys with total recoveries of over 90% of the dose, majorly in feces. CS-0777 and phosphorylated pharmacologically active metabolite of CS-0777 (M1) were highly bound to plasma proteins among rats, monkeys and humans (>93%). 3. The structures of 12 metabolites were identified and phosphorylation and two hydroxylation pathways were proposed as primary metabolism. In the blood of rats and monkeys, the major metabolite was M1 and a few phosphorylated metabolites were also detected. Meanwhile, in urine and feces of rats and monkeys, not phosphorylated, but oxidized CS-0777 metabolites and/or those various conjugated metabolites were observed. This suggests that CS-0777 and its oxidized metabolites would be phosphorylated in the body, but their phosphorylated metabolites would revert back to their dephosphorylated form again then be further metabolized and finally eliminated from the body. 4. Pharmacokinetic analysis using a reversible metabolism model revealed that the clearance of phosphorylation was larger than the clearance of dephosphorylation and elimination.


Asunto(s)
Amino Alcoholes/farmacocinética , Pirroles/farmacocinética , Receptores de Lisoesfingolípidos/efectos de los fármacos , Administración Oral , Algoritmos , Amino Alcoholes/administración & dosificación , Animales , Biotransformación , Heces/química , Hidroxilación , Inyecciones Intravenosas , Absorción Intestinal , Macaca fascicularis , Masculino , Oxidación-Reducción , Fosforilación , Unión Proteica , Pirroles/administración & dosificación , Ratas , Ratas Sprague-Dawley , Distribución Tisular
10.
Am J Physiol Cell Physiol ; 307(1): C14-24, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24740542

RESUMEN

Understanding vascular growth and maturation in developing tumors has important implications for tumor progression, spread, and ultimately host survival. Modulating the signaling of endothelial G protein-coupled receptors (GPCRs) in blood and lymphatic vessels can enhance or limit tumor progression. Sphingosine 1-phosphate receptor 1 (S1PR1) is a GPCR for circulating lysophospholipid S1P that is highly expressed in blood and lymphatic vessels. Using the S1PR1- enhanced green fluorescent protein (eGFP) mouse model in combination with intravital imaging and pharmacologic modulation of S1PR1 signaling, we show that boundary conditions of high and low S1PR1 signaling retard tumor progression by enhancing or destabilizing neovasculature integrity, respectively. In contrast, midrange S1PR1 signaling, achieved by receptor antagonist titration, promotes abundant growth of small, organized vessels and thereby enhances tumor progression. Furthermore, in vivo S1PR1 antagonism supports lung colonization by circulating tumor cells. Regulation of endothelial S1PR1 dynamically controls vascular integrity and maturation and thus modulates angiogenesis, tumor growth, and hematogenous metastasis.


Asunto(s)
Neoplasias de la Mama/irrigación sanguínea , Neoplasias de la Mama/metabolismo , Permeabilidad Capilar , Células Endoteliales/metabolismo , Neovascularización Patológica , Receptores de Lisoesfingolípidos/metabolismo , Anilidas/farmacología , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Permeabilidad Capilar/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Femenino , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundario , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Neoplásicas Circulantes/metabolismo , Células Neoplásicas Circulantes/patología , Organofosfonatos/farmacología , Receptores de Lisoesfingolípidos/efectos de los fármacos , Receptores de Lisoesfingolípidos/genética , Transducción de Señal , Receptores de Esfingosina-1-Fosfato , Factores de Tiempo , Carga Tumoral
11.
Am Heart J ; 168(5): 632-44, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25440790

RESUMEN

Fingolimod, a sphingosine-1-phosphate receptor (S1PR) modulator, was the first oral disease-modifying therapy approved for relapsing forms of multiple sclerosis; it reduces autoreactive lymphocytes' egress from lymphoid tissues by down-regulating S1PRs. Sphingosine-1-phosphate signaling is implicated in a range of physiologic functions, and S1PRs are expressed differentially in various tissues, including the cardiovascular system. Modulation of S1PRs on cardiac cells provides an explanation for the transient effects of fingolimod on heart rate and atrioventricular conduction at initiation of fingolimod therapy, and for the mild but more persistent effects on blood pressure observed in some patients on long-term treatment. This review describes the nontherapeutic actions of fingolimod in the context of sphingosine-1-phosphate signaling in the cardiovascular system, as well as providing a summary of the associated clinical implications useful to physicians considering initiation of fingolimod therapy in patients. A transient reduction in heart rate (mean decrease of 8 beats per minute) and, less commonly, a temporary delay in atrioventricular conduction observed in some patients when initiating fingolimod therapy are both due to activation of S1PR subtype 1 on cardiac myocytes. These effects are a reflection of fingolimod first acting as a full S1PR agonist and thereafter functioning as an S1PR antagonist after down-regulation of S1PR subtype 1 at the cell surface. For most individuals, first-dose effects of fingolimod are asymptomatic, but all patients need to be monitored for at least 6 hours after the first dose, in accordance with the label recommendations.


Asunto(s)
Nodo Atrioventricular/efectos de los fármacos , Frecuencia Cardíaca/efectos de los fármacos , Inmunosupresores/farmacología , Glicoles de Propileno/farmacología , Receptores de Lisoesfingolípidos/efectos de los fármacos , Esfingosina/análogos & derivados , Sistema Cardiovascular/efectos de los fármacos , Regulación hacia Abajo , Clorhidrato de Fingolimod , Humanos , Inmunosupresores/uso terapéutico , Esclerosis Múltiple/tratamiento farmacológico , Glicoles de Propileno/uso terapéutico , Receptores de Lisoesfingolípidos/agonistas , Receptores de Lisoesfingolípidos/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Esfingosina/farmacología , Esfingosina/uso terapéutico
12.
Crit Care Med ; 42(3): e189-99, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24335440

RESUMEN

OBJECTIVE: Effective therapies are needed to reverse the increased vascular permeability that characterizes acute inflammatory diseases such as acute lung injury. FTY720 is a pharmaceutical analog of the potent barrier-enhancing phospholipid, sphingosine 1-phosphate. Because both FTY720 and sphingosine 1-phosphate have properties that may limit their usefulness in patients with acute lung injury, alternative compounds are needed for therapeutic use. The objective of this study is to characterize the effects of FTY720 (S)-phosphonate, a novel analog of FTY720-phosphate, on variables of pulmonary vascular permeability in vitro and alveolar-capillary permeability in vivo. SETTING: University-affiliated research institute. SUBJECTS: Cultured human pulmonary endothelial cells; C57BL/6 mice. INTERVENTIONS: Endothelial cells were stimulated with sphingosine 1-phosphate receptor 1 agonists to determine effects on sphingosine 1-phosphate receptor 1 expression. Acute lung injury was induced in C57BL/6 mice with bleomycin to assess effects of sphingosine 1-phosphate receptor 1 agonists. MEASUREMENTS AND MAIN RESULTS: FTY720 (S)-phosphonate potently increases human pulmonary endothelial cell barrier function in vitro as measured by transendothelial electrical resistance. Reduction of sphingosine 1-phosphate receptor 1 with small interference RNA significantly attenuates this transendothelial electrical resistance elevation. FTY720 (S)-phosphonate maintains endothelial sphingosine 1-phosphate receptor 1 protein expression in contrast to greater than 50% reduction after incubation with sphingosine 1-phosphate, FTY720, or other sphingosine 1-phosphate receptor 1 agonists. FTY720 (S)-phosphonate does not induce ß-arrestin recruitment, sphingosine 1-phosphate receptor 1 ubiquitination, and proteosomal degradation that occur after other agonists. Intraperitoneal administration of FTY720 (S)-phosphonate every other day for 1 week in normal or bleomycin-injured mice maintains significantly higher lung sphingosine 1-phosphate receptor 1 expression compared with FTY720. FTY720 fails to protect against bleomycin-induced acute lung injury in mice, while FTY720 (S)-phosphonate significantly decreases lung leak and inflammation. CONCLUSION: FTY720 (S)-phosphonate is a promising barrier-promoting agent that effectively maintains sphingosine 1-phosphate receptor 1 levels and improves outcomes in the bleomycin model of acute lung injury.


Asunto(s)
Lesión Pulmonar Aguda/tratamiento farmacológico , Arrestinas/metabolismo , Permeabilidad Capilar/efectos de los fármacos , Glicoles de Propileno/farmacología , Receptores de Lisoesfingolípidos/metabolismo , Esfingosina/análogos & derivados , Lesión Pulmonar Aguda/fisiopatología , Animales , Arrestinas/efectos de los fármacos , Bleomicina/farmacología , Western Blotting , Células Cultivadas , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Clorhidrato de Fingolimod , Técnica del Anticuerpo Fluorescente , Humanos , Ratones , Ratones Endogámicos C57BL , Distribución Aleatoria , Receptores Acoplados a Proteínas G/efectos de los fármacos , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Lisoesfingolípidos/efectos de los fármacos , Sensibilidad y Especificidad , Esfingosina/farmacología , beta-Arrestinas
13.
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
14.
Eur J Clin Pharmacol ; 70(3): 287-93, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24362488

RESUMEN

PURPOSE: To determine the effects of steady-state concentrations of the selective S1P1 receptor modulator ponesimod on the pharmacokinetics (PK) of a single dose of a combined oral contraceptive, containing 1 mg norethisterone (NET) and 35 µg ethinyl estradiol (EE) and to investigate the effects on heart rate at different ponesimod doses within an up-titration regimen prior to co-administration of the contraceptive. METHODS: Twenty-two healthy women (age: 29-60 years) received twice a single oral dose of the combined oral contraceptive, alone or in combination with multiple doses of 40 mg ponesimod attained by an up-titration regimen. Heart rate (HR) effects were assessed on the first day of each up-titration level. PK parameters of NET and EE were determined by non-compartmental analysis. RESULTS: Geometric mean ratios (ponesimod and contraceptive / contraceptive alone) of Cmax and AUC0-24 of NET were 0.87 (90 % CI: 0.80, 0.94) and 0.84 (90 % CI: 0.76, 0.93), respectively. Geometric mean ratios of Cmax and AUC0-24 of EE were 0.94 (90 % CI: 0.86, 1.03) and 0.95 (90 % CI: 0.89, 1.01), respectively. The maximum mean HR reduction after the first dose of 10 mg ponesimod was 12.4 bpm (SD ± 6.2) at 2.5 h post-dose. On Day 4 (first dose of 20 mg) and Day 7 (first dose of 40 mg) the maximum mean HR reduction was 4.3 bpm (SD ± 5.7) and 1.4 (SD ± 6.4), respectively, at 2.5 h post-dose compared to baseline. CONCLUSION: No clinically relevant PK interactions between ponesimod and the combined oral contraceptive were observed, therefore, efficacy of hormonal contraceptives is not expected to be affected by concomitant administration of ponesimod. The up-titration regimen showed that HR reductions are diminished upon repeated ponesimod administration.


Asunto(s)
Anticonceptivos Orales Combinados/farmacocinética , Etinilestradiol/farmacocinética , Noretindrona/farmacocinética , Tiazoles/farmacología , Adulto , Área Bajo la Curva , Anticonceptivos Orales Combinados/administración & dosificación , Estudios Cruzados , Relación Dosis-Respuesta a Droga , Combinación de Medicamentos , Interacciones Farmacológicas , Etinilestradiol/administración & dosificación , Femenino , Frecuencia Cardíaca/efectos de los fármacos , Humanos , Persona de Mediana Edad , Noretindrona/administración & dosificación , Receptores de Lisoesfingolípidos/efectos de los fármacos , Receptores de Lisoesfingolípidos/metabolismo , Tiazoles/administración & dosificación , Factores de Tiempo
15.
Acta Pharmacol Sin ; 35(12): 1537-45, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25399649

RESUMEN

AIM: FTY720, a new immunomodulatory drug with low cytotoxicity, is currently used to treat multiple sclerosis. In this study, we investigated the effects of FTY720 on inflammatory cell infiltration in albumin overload-induced nephropathy of rats. METHODS: Male Wistar rats were subjected to right-side nephrectomy and divided into 3 groups. One week after the surgery, albumin overload (AO) group was treated with BSA (5 g·kg(-1)·d(-1), ip) for 9 weeks; AO+FTY720 group was given BSA (5 g·kg(-1)·d(-1), ip) plus FTY720 (0.5 g·kg(-1)·d(-1), ip) for 9 weeks; and control group received daily ip injection of equivalent volume of saline. All rats were killed 9 weeks after nephrectomy. RESULTS: AO rats exhibited gradually increased urinary protein excretion accompanied by elevated urinary N-acetyl-ß-O-glucosaminidase activity, and both reached their peak values at week 7. Furthermore, AO significantly increased lymphocytes and monocytes in circulation and the inflammatory cells recruited to tubulointerstitium, and the expression of inflammatory cytokines MCP-1, TNF-α and IL-6, as well as sphingosine 1-phosphate (S1P) receptors S1pr1 and S1pr3, and S1P-synthesizing enzyme sphingosine kinase 1 (Sphk1) in the kidney. Concomitant administration of FTY720 significantly attenuated all the AO-induced pathological changes. CONCLUSION: FTY720 alleviates tubulointerstitium inflammation in an AO rat model of nephropathy via down-regulation of the Sphk1 pathway.


Asunto(s)
Albuminuria/tratamiento farmacológico , Antiinflamatorios/farmacología , Inmunosupresores/farmacología , Túbulos Renales/efectos de los fármacos , Nefritis Intersticial/prevención & control , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Glicoles de Propileno/farmacología , Esfingosina/análogos & derivados , Acetilglucosaminidasa/orina , Albuminuria/enzimología , Albuminuria/patología , Albuminuria/orina , Animales , Modelos Animales de Enfermedad , Regulación hacia Abajo , Clorhidrato de Fingolimod , Mediadores de Inflamación/metabolismo , Túbulos Renales/enzimología , Túbulos Renales/patología , Linfocitos/efectos de los fármacos , Linfocitos/enzimología , Lisofosfolípidos/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/enzimología , Masculino , Nefritis Intersticial/enzimología , Nefritis Intersticial/patología , Nefritis Intersticial/orina , Ratas Wistar , Receptores de Lisoesfingolípidos/efectos de los fármacos , Receptores de Lisoesfingolípidos/metabolismo , Transducción de Señal/efectos de los fármacos , Esfingosina/metabolismo , Esfingosina/farmacología , Receptores de Esfingosina-1-Fosfato , Factores de Tiempo
16.
J Pharmacokinet Pharmacodyn ; 41(3): 261-78, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24930034

RESUMEN

Ponesimod (ACT-128800), a reversible, orally active, selective S1P1 receptor modulator, prevents the egress of lymphocytes from the lymph node into the systemic circulation. It is currently in clinical development for the treatment of relapsing multiple sclerosis. Modulation of circulating lymphocytes serves as biomarker of efficacy and safety, such that the quantitative characterization of the pharmacokinetic/pharmacodynamic (PK/PD) relationship guides the clinical development of the compound. The availability of a variety of doses, dosing regimens, and treatment durations permitted estimation of the pharmacokinetics characterized by an absorption lag time followed by a sequential zero/first-order absorption and two compartments with first-order elimination. The PD are modeled as an indirect-effect model with rates of appearance and disappearance of lymphocytes in blood with a circadian rhythm and a drug effect on the rate of appearance. The model suggests a circadian variation of 9% and a maximum inhibition of 86% of total lymphocyte count with high doses at steady state. It was instrumental for the selection of doses for subsequent studies that confirmed the effect plateau in total lymphocyte count at approximately 0.5 × 10(9) counts/L.


Asunto(s)
Receptores de Lisoesfingolípidos/efectos de los fármacos , Tiazoles/farmacología , Tiazoles/farmacocinética , Adolescente , Adulto , Anciano , Peso Corporal/fisiología , Ritmo Circadiano/fisiología , Femenino , Humanos , Absorción Intestinal , Recuento de Linfocitos , Masculino , Persona de Mediana Edad , Modelos Estadísticos , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/metabolismo , Población , Adulto Joven
17.
Hepatology ; 56(4): 1427-38, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22505286

RESUMEN

UNLABELLED: Sinusoidal vasoconstriction, in which hepatic stellate cells operate as contractile machinery, has been suggested to play a pivotal role in the pathophysiology of portal hypertension. We investigated whether sphingosine 1-phosphate (S1P) stimulates contractility of those cells and enhances portal vein pressure in isolated perfused rat livers with Rho activation by way of S1P receptor 2 (S1P(2) ). Rho and its effector, Rho kinase, reportedly contribute to the pathophysiology of portal hypertension. Thus, a potential effect of S1P(2) antagonism on portal hypertension was examined. Intravenous infusion of the S1P(2) antagonist, JTE-013, at 1 mg/kg body weight reduced portal vein pressure by 24% without affecting mean arterial pressure in cirrhotic rats induced by bile duct ligation at 4 weeks after the operation, whereas the same amount of S1P(2) antagonist did not alter portal vein pressure and mean arterial pressure in control sham-operated rats. Rho kinase activity in the livers was enhanced in bile duct-ligated rats compared to sham-operated rats, and this enhanced Rho kinase activity in bile duct-ligated livers was reduced after infusion of the S1P(2) antagonist. S1P(2) messenger RNA (mRNA) expression, but not S1P(1) or S1P(3) , was increased in bile duct-ligated livers of rats and mice and also in culture-activated rat hepatic stellate cells. S1P(2) expression, determined in S1P 2LacZ/+ mice, was highly increased in hepatic stellate cells of bile duct-ligated livers. Furthermore, the increase of Rho kinase activity in bile duct-ligated livers was observed as early as 7 days after the operation in wildtype mice, but was less in S1P 2-/- mice. CONCLUSION: S1P may play an important role in the pathophysiology of portal hypertension with Rho kinase activation by way of S1P(2) . The S1P(2) antagonist merits consideration as a novel therapeutic agent for portal hypertension.


Asunto(s)
Hemodinámica/efectos de los fármacos , Hipertensión Portal/tratamiento farmacológico , Pirazoles/farmacología , Piridinas/farmacología , Receptores de Lisoesfingolípidos/antagonistas & inhibidores , Quinasas Asociadas a rho/metabolismo , Animales , Conductos Biliares/cirugía , Células Cultivadas/efectos de los fármacos , Modelos Animales de Enfermedad , Activación Enzimática/efectos de los fármacos , Activación Enzimática/genética , Regulación de la Expresión Génica , Hemodinámica/fisiología , Células Estrelladas Hepáticas/efectos de los fármacos , Células Estrelladas Hepáticas/fisiología , Hipertensión Portal/fisiopatología , Immunoblotting , Inmunohistoquímica , Infusiones Intravenosas , Ligadura , Masculino , Ratones , Ratones Transgénicos , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Lisoesfingolípidos/efectos de los fármacos , Receptores de Lisoesfingolípidos/genética , Valores de Referencia , Sensibilidad y Especificidad , Quinasas Asociadas a rho/efectos de los fármacos
18.
Handb Exp Pharmacol ; (215): 239-53, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23579459

RESUMEN

The recent success of FTY720 (Fingolimod, Gilenya(®)), which has been approved for the treatment of relapsing-remitting multiple sclerosis and is the first-in-class sphingosine-1-phosphate (S1P) receptor modulating drug, has boosted the interest in further drug development in this area. Several selective S1P1 receptor-modulating drugs are being investigated in clinical trials for the treatment of diverse autoimmune disorders. Sphingosine kinase inhibitors are under development for the treatment of cancer, aberrant angiogenesis and inflammatory diseases; an inhibitor of SK2 with relatively low affinity is being analysed in patients with advanced solid tumours. While an indirect S1P lyase inhibitor has just failed the proof of concept in patients with rheumatoid arthritis, S1P lyase is still a promising target for the treatment of inflammatory and autoimmune diseases. Another approach is the development of S1P-scavenging or -clearing agents, including a monoclonal S1P antibody that has successfully passed phase I clinical trials and will be further developed for age-related macular degeneration.


Asunto(s)
Lisofosfolípidos/fisiología , Transducción de Señal/fisiología , Esfingosina/análogos & derivados , Aldehído-Liasas/antagonistas & inhibidores , Animales , Inhibidores Enzimáticos/farmacología , Clorhidrato de Fingolimod , Humanos , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Glicoles de Propileno/uso terapéutico , Receptores de Lisoesfingolípidos/efectos de los fármacos , Esfingosina/fisiología , Esfingosina/uso terapéutico
19.
Med Sci (Paris) ; 28(11): 951-7, 2012 Nov.
Artículo en Francés | MEDLINE | ID: mdl-23171898

RESUMEN

Sphingosine 1-phosphate (S1P) mediates critical physiological responses by its binding to G protein-coupled receptor (GPCR) subtypes, known as S1P receptors. Five distinct mammalian S1P receptors, designated S1P1-5 have been identified, each with a different cellular pattern of expression which influences the responses to S1P. In this review, we briefly outline our understanding of the modes of action and the roles of S1P receptors in the regulation of physiological and pathological functions in the cardiovascular, immune and central nervous system.


Asunto(s)
Lisofosfolípidos/fisiología , Receptores de Lisoesfingolípidos/fisiología , Esfingosina/análogos & derivados , Animales , Aterosclerosis/fisiopatología , Sistema Cardiovascular/fisiopatología , Movimiento Celular , Humanos , Linfocitos/metabolismo , Activación de Macrófagos , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/ultraestructura , Mamíferos , Ratones , Ratones Noqueados , Neoplasias/irrigación sanguínea , Neoplasias/inmunología , Neovascularización Patológica/fisiopatología , Neovascularización Fisiológica/fisiología , Proteínas del Tejido Nervioso/fisiología , Neurogénesis/fisiología , Receptores de Lisoesfingolípidos/clasificación , Receptores de Lisoesfingolípidos/deficiencia , Receptores de Lisoesfingolípidos/efectos de los fármacos , Sistemas de Mensajero Secundario/fisiología , Esfingosina/fisiología
20.
Ideggyogy Sz ; 65(11-12): 369-76, 2012 Nov 30.
Artículo en Húngaro | MEDLINE | ID: mdl-23289171

RESUMEN

Fingolimod is a sphingosine-1 phosphate receptor modulator, which is effective in the treatment of severe relapsing-remitting form of multiple sclerosis. Once daily oral use of fingolimod decreased the annualized relapse rate, inflammatory brain lesion activity and the rate of brain atrophy compared both to placebo and intramuscular administered interferon beta-1a. The drug targets the cardiovascular system as well via sphingosine-1 phosphate receptors. After initiation of fingolimod therapy transient sinus bradycardia and slowing of the atrioventricular conduction develops. The onset of the effect is as early as 1 hour post administration, while heart rate and conduction normalized in 24 hours in most of the cases. According to the clinical trials symptomatic bradycardia developed in 0.5% of the cases, responding to the appropriate therapy. The incidence of Mobitz I type II atrioventricular blocks and blocks with 2:1 atrioventricular conduction was 0.2% and 0.1%, respectively. All of these cardiovascular events showed regression during observation and no higher degree atrioventricular blocks were detected at the approved therapeutic dose. Following the first dose effect, fingolimod had a moderate hypertensive effect on long-term. For the safety of fingolimod treatment detailed cardiovascular risk stratification of all patients, adequate patient monitoring after the first dose and competency in treating the possible side effects is necessary. In patients with increased cardiovascular risks, treatment should be considered only if anticipated benefits outweigh potential risks and extended monitoring is required.


Asunto(s)
Sistema Cardiovascular/efectos de los fármacos , Inmunosupresores/uso terapéutico , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/prevención & control , Glicoles de Propileno/uso terapéutico , Esfingosina/análogos & derivados , Bloqueo Atrioventricular/inducido químicamente , Bradicardia/inducido químicamente , Ensayos Clínicos como Asunto , Esquema de Medicación , Clorhidrato de Fingolimod , Sistema de Conducción Cardíaco/efectos de los fármacos , Humanos , Hipertensión/inducido químicamente , Inmunosupresores/administración & dosificación , Inmunosupresores/efectos adversos , Interferón beta-1a , Interferón beta/uso terapéutico , Glicoles de Propileno/administración & dosificación , Glicoles de Propileno/efectos adversos , Receptores de Lisoesfingolípidos/efectos de los fármacos , Prevención Secundaria , Esfingosina/administración & dosificación , Esfingosina/efectos adversos , Esfingosina/uso terapéutico , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA