Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Eur J Pharm Sci ; 56: 65-9, 2014 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-24568891

RESUMEN

INTRODUCTION: Concizumab (mAb 2021) is a monoclonal IgG4 antibody (mAb) that binds to the Kunitz-type protease inhibitor (KPI) 2 domain of TFPI thereby blocking the interaction of this domain with the active site of FXa. The objective of the present study was to characterize the pharmacokinetics of concizumab in Cynomolgus monkeys after intravenous (iv) and subcutaneous (sc) administration. METHODS: Data from two studies were included in the modelling, all in all data from 52 monkeys distributed into 9 groups. Three groups received three escalating sc doses of concizumab with a one week dosing interval, two groups were administered a single dose, and four groups received multiple doses over 13 weeks of concizumab. The plasma concentration was measured using a standard ELISA, and pharmacokinetic data were analysed using NONMEM. RESULTS: The pharmacokinetics of concizumab were characterised by a high bioavailability (93%) after sc administration. The time course of the elimination of concizumab from the circulation was well described by the proposed target mediated drug disposition (TMDD) model. The clearance of concizumab was estimated to be 0.14 ml/h/kg, the target clearance was characterized by a 50% saturation level of 0.54 µg/ml (Km), and the clearance at target saturation was estimated to be 11 µg/h/kg. CONCLUSION: Concizumab displays a typical TMDD profile with important implications for a putative treatment regime in haemophilia patients. Compared to current standard haemophilia treatment, concizumab has a high bioavailability after sc administration and may provide a viable alternative to intravenous dosing for the treatment of haemophilia.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacocinética , Factor Xa/metabolismo , Lipoproteínas/metabolismo , Modelos Biológicos , Administración Intravenosa , Animales , Anticuerpos Monoclonales Humanizados/administración & dosificación , Anticuerpos Monoclonales Humanizados/sangre , Dominio Catalítico , Inyecciones Subcutáneas , Lipoproteínas/inmunología , Macaca fascicularis
2.
Thromb Res ; 133(3): 464-71, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24393663

RESUMEN

INTRODUCTION: A humanised monoclonal antibody, concizumab, that binds with high affinity to the Kunitz-type protease inhibitor (KPI) 2 domain of human tissue factor pathway inhibitor (TFPI) is in clinical development. It promotes coagulation by neutralising the inhibitory function of TFPI and may provide a subcutaneous prophylaxis option for patients with haemophilia. We aimed to study biodistribution and pharmacokinetics (PK) of concizumab. MATERIALS AND METHODS: Blockage of cellular TFPI by concizumab was measured by tissue factor/Factor VIIa-mediated Factor X activation on human EA.hy926 cells. Biodistribution of concizumab was analysed in rabbits by immunohistology, and the PK was measured in rabbits and rats. RESULTS AND CONCLUSIONS: Concizumab bound to cell surface TFPI on EA.hy926 cells and neutralised TFPI inhibition of Factor X activation. The antibody cross-reacted with rabbit TFPI, but not with rat TFPI, allowing for comparative PK studies. PK data in rats described a log-linear profile typical for a non-binding antibody, whereas PK data in rabbits revealed a non-linear, dose-dependent profile, consistent with a target-mediated clearance mechanism. Immunohistology in rabbits during target-saturation showed localisation of the antibody on the endothelium of the microvasculature in several organs. We observed a marked co-localisation with endogenous rabbit TFPI, but a negligible sub-endothelial build-up. Concizumab binds and neutralises the inhibitory effect of cell surface-bound TFPI. The PK profile observed in rabbits is consistent with a TFPI-mediated drug disposition. Double immunofluorescence shows co-localisation of the antibody with TFPI on the endothelium of the microvasculature and points to this TFPI as a putative target involved in the clearance mechanism.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacocinética , Lipoproteínas/inmunología , Inhibidores de Proteasas/farmacocinética , Animales , Anticuerpos Monoclonales Humanizados/inmunología , Coagulación Sanguínea , Femenino , Humanos , Lipoproteínas/antagonistas & inhibidores , Ratones , Inhibidores de Proteasas/inmunología , Estructura Terciaria de Proteína , Conejos , Ratas , Distribución Tisular , Inhibidor de la Tripsina de Soja de Kunitz/inmunología
3.
Blood ; 119(24): 5871-8, 2012 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-22563084

RESUMEN

Hemophilia is treated by IV replacement therapy with Factor VIII (FVIII) or Factor IX (FIX), either on demand to resolve bleeding, or as prophylaxis. Improved treatment may be provided by drugs designed for subcutaneous and less frequent administration with a reduced risk of inhibitor formation. Tissue factor pathway inhibitor (TFPI) down-regulates the initiation of coagulation by inhibition of Factor VIIa (FVIIa)/tissue factor/Factor Xa (FVIIa/TF/FXa). Blockage of TFPI inhibition may facilitate thrombin generation in a hemophilic setting. A high-affinity (K(D) = 25pM) mAb, mAb 2021, against TFPI was investigated. Binding of mAb 2021 to TFPI effectively prevented inhibition of FVIIa/TF/FXa and improved clot formation in hemophilia blood and plasma. The binding epitope on the Kunitz-type protease inhibitor domain 2 of TFPI was mapped by crystallography, and showed an extensive overlap with the FXa contact region highlighting a structural basis for its mechanism of action. In a rabbit hemophilia model, an intravenous or subcutaneous dose significantly reduced cuticle bleeding. mAb 2021 showed an effect comparable with that of rFVIIa. Cuticle bleeding in the model was reduced for at least 7 days by a single intravenous dose of mAb 2021. This study suggests that neutralization of TFPI by mAb 2021 may constitute a novel treatment option in hemophilia.


Asunto(s)
Anticuerpos Bloqueadores/farmacología , Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Factor Xa/metabolismo , Hemofilia A/tratamiento farmacológico , Hemostasis/efectos de los fármacos , Lipoproteínas/metabolismo , Modelos Moleculares , Animales , Anticuerpos Bloqueadores/administración & dosificación , Anticuerpos Bloqueadores/uso terapéutico , Anticuerpos Monoclonales Humanizados/administración & dosificación , Anticuerpos Neutralizantes/farmacología , Tiempo de Sangría , Coagulación Sanguínea/efectos de los fármacos , Reacciones Cruzadas/efectos de los fármacos , Modelos Animales de Enfermedad , Epítopos/inmunología , Factor VIII/farmacología , Factor Xa/inmunología , Femenino , Fibrina/metabolismo , Células HEK293 , Hemofilia A/sangre , Células Endoteliales de la Vena Umbilical Humana , Humanos , Pruebas de Neutralización , Unión Proteica/efectos de los fármacos , Estructura Terciaria de Proteína , Conejos , Especificidad de la Especie , Tromboplastina/farmacología
4.
Blood ; 118(8): 2333-41, 2011 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-21700771

RESUMEN

Current management of hemophilia B entails multiple weekly infusions of factor IX (FIX) to prevent bleeding episodes. In an attempt to make a longer acting recombinant FIX (rFIX), we have explored a new releasable protraction concept using the native N-glycans in the activation peptide as sites for attachment of polyethylene glycol (PEG). Release of the activation peptide by physiologic activators converted glycoPEGylated rFIX (N9-GP) to native rFIXa and proceeded with normal kinetics for FXIa, while the K(m) for activation by FVIIa-tissue factor (TF) was increased by 2-fold. Consistent with minimal perturbation of rFIX by the attached PEG, N9-GP retained 73%-100% specific activity in plasma and whole-blood-based assays and showed efficacy comparable with rFIX in stopping acute bleeds in hemophilia B mice. In animal models N9-GP exhibited up to 2-fold increased in vivo recovery and a markedly prolonged half-life in mini-pig (76 hours) and hemophilia B dog (113 hours) compared with rFIX (16 hours). The extended circulation time of N9-GP was reflected in prolonged correction of coagulation parameters in hemophilia B dog and duration of effect in hemophilia B mice. Collectively, these results suggest that N9-GP has the potential to offer efficacious prophylactic and acute treatment of hemophilia B patients at a reduced dosing frequency.


Asunto(s)
Factor IX/química , Factor IX/metabolismo , Animales , Sitios de Unión , Modelos Animales de Enfermedad , Perros , Factor IX/genética , Femenino , Semivida , Hemofilia B/sangre , Hemofilia B/tratamiento farmacológico , Hemofilia B/genética , Hemostáticos/sangre , Hemostáticos/química , Hemostáticos/farmacología , Humanos , Técnicas In Vitro , Cinética , Masculino , Ratones , Ratones Mutantes , Polietilenglicoles/química , Polisacáridos/química , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Porcinos , Porcinos Enanos
5.
Front Biosci (Landmark Ed) ; 16(9): 3196-215, 2011 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-21622229

RESUMEN

Knowledge about species compatibility is crucial for proper interpretation of data from in vivo experiments with human proteins in pharmacological models and of data from cross-species in vitro experiments. Information about the cross-species compatibility of tissue factor (TF) and coagulation factor (F) VII (FVII) has accumulated since the early history of coagulation research. Many observations were connected to the introduction and development of the prothrombin time (PT) assay where fibrin clot formation was observed when tissue extracts of different origins were added to recalcified human or non-human plasmas. Studies on cross-species TF-FVIIa compatibility entered into a new area with the cloning and recombinant expression of TF and FVII from a number of species as well as with the possibility of specific amino acid substitution. TF and/or FVIIa from cattle, dog, rabbit, mouse, rat and zebrafish have been purified and characterized in varying detail. In addition to adding knowledge about the species-specific TF-FVIIa interactions, cross-species studies often reveal information which adds to the general view of the structural and functional properties of the human TF-FVIIa complex. This review briefly outlines the features of human TF and FVIIa, their intermolecular interactions, and the biological effects of TF-FVIIa complex formation and compares this information to findings obtained in studies addressing TF or FVIIa of non-human origin. By examples we point to difficulties which may arise from the transcendence across species borders and how some cross-species data have advanced our understanding of the structure and function of the human TF-FVIIa complex.


Asunto(s)
Factor VIIa/metabolismo , Tromboplastina/metabolismo , Secuencia de Aminoácidos , Animales , Coagulación Sanguínea/fisiología , Bovinos , Perros , Factor VIIa/química , Factor VIIa/genética , Humanos , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Dominios y Motivos de Interacción de Proteínas , Conejos , Ratas , Homología de Secuencia de Aminoácido , Especificidad de la Especie , Tromboplastina/química , Tromboplastina/genética , Pez Cebra
6.
Eur J Pharm Sci ; 42(5): 578-83, 2011 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-21382491

RESUMEN

AIM: NN1731 is a recombinant activated factor VII (rFVIIa) analogue with enhanced activity. The objective of the present study was to evaluate the clearance mechanisms of rFVIIa and NN1731 after intravenous administration to Beagle dogs. METHODS: The study was performed in Beagle dogs administered with a single dose of 5.4 nmol/kg rFVIIa or NN1731 intravenously. Plasma samples collected up to 12-h post-administration were analysed using three different assays to determine FVIIa clot activity (FVIIa:C), total FVIIa antigen, and levels of FVIIa-antithrombin (AT) complexes. Pharmacokinetic parameters were determined by use of standard non-compartmental and non-linear mixed effects methods. RESULTS: For both compounds, complex formation with AT accounted for the observed difference between the activity and the antigen curves and constituted 60-70% of the total clearance. The clearance of rFVIIa and NN1731 was estimated to be 73 and 214 mL/h/kg, respectively, accordingly, AT complex formation occurred around three times faster for NN1731. The difference in activity observed in the initial phase, resulting in distribution half-lives of 0.71 and 0.22 h for rFVIIa and NN1731, was mainly caused by the 3-fold difference in clearance. The terminal half-life of rFVIIa and NN1731 was estimated to be 2.1 and 2.5 h, respectively. The non-compartmental analysis resulted in almost identical parameters. CONCLUSION: The present study demonstrates that the difference between the activity and the antigen profiles of rFVIIa and NN1731 in Beagle dogs is the result of complex formation with AT which constitutes a major pathway for the clearance of rFVIIa activity.


Asunto(s)
Factor VII/farmacocinética , Factor VIIa/farmacocinética , Modelos Biológicos , Animales , Proteínas Antitrombina/fisiología , Coagulación Sanguínea/efectos de los fármacos , Interpretación Estadística de Datos , Perros , Factor VII/administración & dosificación , Factor VII/farmacología , Factor VIIa/administración & dosificación , Factor VIIa/farmacología , Semivida , Inyecciones Intravenosas , Tasa de Depuración Metabólica , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/farmacocinética , Proteínas Recombinantes/farmacología
7.
Thromb Res ; 122 Suppl 1: S11-3, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18691491

RESUMEN

Factor VIIa (FVIIa)-induced signal transduction is strongly dependent on cellular surface expression of Tissue Factor (TF) and Protease Activated Receptors (PARs). FVIIa signals primarily through PAR2. This contrasts to thrombin which signals primarily via PAR1 and does so without the assistance of a co-receptor, but by binding to an exosite on PAR1. Various TF:FVII-mediated cellular activities are now well documented and have indicated possible links to inflammation, atherosclerosis, angiogenesis, tissue repair, tumor growth and metastasis. Further knowledge about cellular responses induced by coagulation factors has been obtained by gene-expression profiling of MDA-MB-231 cells stimulated with FVIIa or alternatively with PAR1 or PAR2 agonist peptides. These studies and qPCR measurements of the transcription of selected genes in these and other carcinoma cell lines have provided new information about gene expression induced by PAR activation, the gene repertoire induced by TF:FVIIa via PAR2, and how it differs from that induced via PAR1 by thrombin.


Asunto(s)
Neoplasias de la Mama/metabolismo , Factor VIIa/metabolismo , Análisis por Matrices de Proteínas , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Carcinoma/genética , Carcinoma/metabolismo , Carcinoma/patología , Línea Celular Tumoral , Factor VIIa/fisiología , Femenino , Perfilación de la Expresión Génica , Humanos , Receptor PAR-1/agonistas , Receptor PAR-2/agonistas , Transducción de Señal , Tromboplastina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...