Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Thromb Haemost ; 17(2): 350-360, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30525289

RESUMO

Essentials Factor (F)VIII with an intermediate-length B-domain showed higher levels in murine gene therapy. FVIII with different B-domain lengths were analysed. FVIII variants with B-domains between 186 and 240 amino acids (aa) have extended half-life in mice. Reduced cell binding of FVIII with a 237aa B-domain may explain the extended half-life. SUMMARY: Background Factor VIII consists of the A1-domain, A2-domain, B-domain, A3-domain, C1-domain, and C2-domain. FVIII with an intermediate-length B-domain of 226 amino acids (aa) has previously been evaluated in murine gene therapy studies. Objective To characterize FVIII with intermediate-length B-domains in vitro and in vivo in F8-knockout (KO) mice. Methods and results FVIII molecules with B-domains of 186-240aa had longer half-lives in F8-KO mice than FVIII molecules with shorter or longer B-domains. FVIII with a B-domain containing the 225 N-terminal aa fused to the 12 C-terminal aa of the wild-type B-domain (FVIII-237) had a 1.6-fold extended half-life in F8-KO mice as compared with FVIII with a 21aa B-domain (FVIII-21). The in vitro and in vivo activity of FVIII-237 were comparable to those of FVIII-21, as was binding to von Willebrand factor. Cell binding to LDL receptor-related protein 1 (LRP-1)-expressing cells was markedly reduced for FVIII-237 as compared with FVIII-21, whereas the affinity for LRP-1 was not reduced in surface plasmon resonance (SPR) studies. FVIII-21 cell binding and internalization could be inhibited by a fragment consisting of the 226 N-terminal aa of the FVIII B-domain, and SPR analysis suggested that this B-domain fragment might bind with weak affinity to FVIII-21. Conclusion Reduced cell binding of FVIII-237 might explain the observed extended half-life in F8-KO mice. This may contribute to the increased FVIII levels measured in murine gene therapy studies using FVIII constructs with similar B-domain lengths.


Assuntos
Coagulantes/farmacocinética , Fator VIII/farmacocinética , Hemofilia A/tratamento farmacológico , Animais , Linhagem Celular , Coagulantes/sangue , Modelos Animais de Doenças , Fator VIII/genética , Técnicas de Inativação de Genes , Meia-Vida , Hemofilia A/sangue , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Camundongos Knockout , Ligação Proteica , Domínios Proteicos , Proteínas Recombinantes/farmacocinética
2.
J Thromb Haemost ; 16(6): 1141-1152, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29582559

RESUMO

Essentials N8-GP is an extended half-life recombinant factor VIII (FVIII) for the treatment of hemophilia A. Subcutaneous (SC) FVIII dosing might reduce the treatment burden of prophylaxis. SC N8-GP has a favorable PK profile in animal models and disappears from skin injection sites. Combined animal (SC) and clinical (IV) data suggest that daily SC dosing may provide prophylaxis. SUMMARY: Background N8-GP is an extended half-life recombinant factor VIII (FVIII) for the treatment of hemophilia A. Subcutaneous administration of FVIII may reduce the treatment burden of prophylaxis; however, standard FVIII products have low bioavailability after subcutaneous dosing in animals. Objective To evaluate the pharmacokinetics, effectiveness and local distribution of subcutaneously administered N8-GP in preclinical models and predict the human pharmacokinetic (PK) profile. Methods The pharmacokinetics of subcutaneously administered N8-GP were evaluated in FVIII knockout (F8-KO) mice and cynomolgus monkeys; a human PK prediction model in hemophilia A patients was developed. The hemostatic effect was evaluated in a tail vein bleeding model in F8-KO mice. The injection-site distribution and absorption of subcutaneously administered N8-GP were assessed in F8-KO mice by the use of temporal fluorescence imaging and immunohistochemistry. Results Subcutaneously administered N8-GP had a bioavailability, a first-order absorption rate and a half-life, respectively, of 24%, 0.094 h-1 and 14 h in F8-KO mice, and 26%, 0.33 h-1 and 15 h in cynomolgus monkeys. A dose-dependent effect of subcutaneously administered N8-GP on blood loss was observed in mice. A minimal amount of N8-GP was detected at the injection site 48-72 h after single or multiple dose(s) in F8-KO mice. Subcutaneously administered N8-GP was localized to the skin around the injection site, with time-dependent disappearance from the depot. PK modeling predicted that subcutaneously administered N8-GP at a daily dose of 12.5 IU kg-1 will provide FVIII trough levels of 2.5-10% in 95% of patients with severe hemophilia A. Conclusions Subcutaneously administered N8-GP may provide effective hemophilia A prophylaxis. A phase I clinical trial is underway to investigate this possibility.


Assuntos
Fator VIII/administração & dosagem , Fator VIII/farmacocinética , Hemofilia A/tratamento farmacológico , Hemostáticos/administração & dosagem , Hemostáticos/farmacocinética , Modelos Biológicos , Polietilenoglicóis/administração & dosagem , Polietilenoglicóis/farmacocinética , Animais , Modelos Animais de Doenças , Fator VIII/genética , Fator VIII/metabolismo , Meia-Vida , Hemofilia A/sangue , Hemofilia A/genética , Hemostáticos/sangue , Humanos , Injeções Intravenosas , Injeções Subcutâneas , Macaca fascicularis , Camundongos Knockout , Absorção Cutânea , Especificidade da Espécie , Distribuição Tecidual
3.
J Thromb Haemost ; 14(9): 1803-13, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27378673

RESUMO

UNLABELLED: Essentials Von Willebrand factor (VWF) stabilizes factor VIII (FVIII) and prevents its premature clearance. Rat anatomical and hepatocellular distribution studies assessed the VWF effect on FVIII clearance. Hepatocytes and liver sinusoidal endothelial cells play a key role in FVIII clearance. Anatomical and hepatocellular distribution of FVIII is independent of high-affinity VWF binding. ABSTRACT: Background Von Willebrand factor (VWF) stabilizes factor VIII in the circulation and prevents its premature clearance. Objective To study the effects of VWF on FVIII clearance in rats with endogenous VWF. Methods Anatomical and hepatocellular distribution studies were performed in rats following intravenous administration of glycoiodinated recombinant FVIII (rFVIII) and a FVIII variant, FVIII-Y1680F, lacking high-affinity VWF binding. Radioactivity was quantified in organs, and in distinct liver cell populations. The role of VWF binding was also studied by immunohistochemical staining of rat livers perfused ex vivo with rFVIII alone or with a FVIII-binding VWF fragment. Results The liver was the predominant organ of rFVIII distribution, and a radioactivity peak was also observed in the intestines, suggesting FVIII secretion to the bile by hepatocytes. In the liver, ~60% of recovered radioactivity was associated with hepatocytes, 32% with liver sinusoidal endothelial cells (LSECs), and 9% with Kupffer cells (KCs). When calculated per cell, 1.5-fold to 3-fold more radioactivity was associated with LSECs than with hepatocytes. The importance of hepatocytes and LSECs was confirmed by immunohistochemical staining; strong staining was seen in LSECs, and less intense, punctate staining in hepatocytes. Minor staining in KCs was observed. Comparable anatomical and hepatocellular distributions were observed with rFVIII and FVIII-Y1680F, and the presence of the VWF fragment, D'D3A1, did not change the FVIII staining pattern in intact livers. Conclusions The present data support FVIII clearance via the liver, with hepatocytes and LSECs playing a key role. High-affinity VWF binding did not alter the anatomical or hepatocellular distribution of FVIII.


Assuntos
Células Endoteliais/metabolismo , Fator VIII/metabolismo , Hepatócitos/citologia , Fígado/metabolismo , Fator de von Willebrand/metabolismo , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Combinação de Medicamentos , Ensaio de Imunoadsorção Enzimática , Fator VIII/uso terapêutico , Glioblastoma/metabolismo , Hepatócitos/metabolismo , Humanos , Imuno-Histoquímica , Iodo/química , Lactoperoxidase/metabolismo , Masculino , Ligação Proteica , Ratos , Ratos Sprague-Dawley , Distribuição Tecidual , Doenças de von Willebrand/sangue , Doenças de von Willebrand/tratamento farmacológico , Fator de von Willebrand/uso terapêutico
4.
J Thromb Haemost ; 14(8): 1579-87, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27241682

RESUMO

UNLABELLED: Essentials Chromogenic assays may be less variable than one-stage clot assays for measuring modified factor VIII. Chromogenic assays were evaluated for N8-GP potency labeling and postadministration monitoring. There was no significant difference between chromogenic assay kits for measuring N8-GP potency. Postadministration monitoring of N8-GP was comparable to turoctocog alfa for all kits tested. SUMMARY: Background Factor VIII activity ( FVIII: C) is commonly measured using one-stage activated partial thromboplastin time (aPTT)-based clot assays. Chromogenic assays are, however, an alternative, and potency assessment in Europe is performed using chromogenic assays. One-stage clot assays are in general associated with high variability, and modified FVIII products may add to this variability. FVIII chromogenic assays may be less affected. Objectives To evaluate available chromogenic assay kits for potency labeling of polyethylene glycol-glycoconjugated turoctocog alfa (turoctocog alfa pegol [N8-GP]) and to evaluate selected chromogenic kits for postadministration monitoring of N8-GP using turoctocog alfa (Novoeight(®) ) as comparator. Methods Six FVIII chromogenic assay kits were adapted to the European Pharmacopeia guidelines for potency labeling, including assessment of time to 50% FX activation. Four kits were adapted for postadministration monitoring using an ACL(®) TOP 500 analyzer. Severe hemophilia A plasma was spiked with N8-GP or turoctocog alfa to simulate postadministration samples. The World Health Organization (WHO) 8th International Standard (IS) FVIII concentrate was used as calibrator throughout. In addition, a plasma calibrator was used for postadministration samples. Results When measuring N8-GP potency, no significant difference using a 1% significance level was observed between kits. In simulated postadministration samples, all test kits were highly accurate and precise, except at low concentrations, with no significant difference between FVIII: C (P > 0.05) measured using the different calibrators. However, values obtained using the WHO 8th IS were closer to labeled values. Conclusions Chromogenic assay kits tested measured consistent FVIII: C for N8-GP potency and showed comparable results for N8-GP and turoctocog alfa in simulated postadministration samples.


Assuntos
Fator VIII/química , Testes de Coagulação Sanguínea , Calibragem , Compostos Cromogênicos/química , Monitoramento de Medicamentos/métodos , Europa (Continente) , Guias como Assunto , Hemofilia A/sangue , Humanos , Tempo de Tromboplastina Parcial , Polietilenoglicóis/química , Domínios Proteicos , Reprodutibilidade dos Testes
5.
Haemophilia ; 18(1): 139-45, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21771205

RESUMO

N8 is a new recombinant factor VIII (rFVIII) compound produced and formulated without human- or animal-derived protein. The aims of the present studies were to evaluate the pharmacokinetics and pharmacodynamics properties of N8 and to compare with a commercially available rFVIII product (Advate(®)) in haemophilia A mice. The pharmacokinetics were evaluated after single i.v. administration of 80, 120 and 280 IU kg(-1) of N8 and Advate(®) and measurements of FVIII blood concentrations as a function of time. The efficacy and dose response curves of N8 and Advate(®) (1-200 IU kg(-1)) were evaluated in a tail bleeding model. Furthermore, the effects in a newly developed haemophilia knee joint haemarthrosis model were investigated. No significant differences were found in the pharmacokinetic parameters between N8 and Advate(®). The clearances were 11 ± 1 vs. 10 ± 2 mL h(-1) kg(-1) (P = 0.14) and the half-lives 7.2 ± 0.9 vs. 7.7 ± 1.4 h (P = 0.31) after administration of N8 and Advate(®) respectively. Dose-independent pharmacokinetics was shown, and comparable efficacy and potency were shown between N8 and Advate(®) in the tail bleeding model. Both compounds normalized the bleeding at the dose of 200 IU kg(-1), and for blood loss ED(50) values of 27 IU kg(-1) (N8) and 28 IU/kg (Advate(®)) were found (P = 0.97). In the haemarthrosis model, treatment with N8 and Advate(®) at 200 IU kg(-1) reduced the mean increase in the joint diameter significantly from 1.23 ± 0.19 to 0.32 ± 0.08 mm (P < 0.01) and 0.25 ± 0.08 mm (P < 0.001) respectively. Pharmacokinetics and pharmacodynamics of N8 and Advate(®) were comparable after i.v. administration to haemophilia A mice.


Assuntos
Fator VIII/farmacocinética , Hemofilia A/tratamento farmacológico , Análise de Variância , Animais , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Fator VIII/uso terapêutico , Meia-Vida , Hemartrose/tratamento farmacológico , Hemartrose/metabolismo , Hemofilia A/metabolismo , Hemorragia/tratamento farmacológico , Articulação do Joelho , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Recombinantes/farmacocinética , Proteínas Recombinantes/uso terapêutico
6.
Haemophilia ; 17(6): 962-70, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21645178

RESUMO

Recombinant human FVIIa (rhFVIIa) corrects the coagulopathy in hemophilia A and B as well as FVII deficiency. This is also the case in dogs until canine anti-human FVIIa antibodies develop (~2 weeks). Recombinant canine factor VIIa (rcFVIIa), successfully over-expressed by gene transfer in haemophilia dogs, has provided long-term haemostasis (>2 years). However, pharmacokinetics (PK), pharmacodynamics (PD) and safety of rcFVIIa after pharmacological administration have not been reported. We therefore wanted to explore the safety, PK and PD of rcFVIIa in dogs. A pilot study was set up to evaluate the safety as well as PK and PD of rcFVIIa after a single intravenous dose of 270 µg kg(-1) to one HA and one haemostatically normal dog and to directly compare rcFVIIa with rhFVIIa in these two dogs. Single doses of rcFVIIa and rhFVIIa were well tolerated. No adverse events were observed. Pharmacokinetic characteristics including half-life (FVIIa activity: 1.2-1.8 h; FVIIa antigen 2.8-3.7 h) and clearance were comparable for rcFVIIa and rhFVIIa. Kaolin-activated thromboelastography approached normal in the HA dog with the improvement being most pronounced after rcFVIIa. This study provided the first evidence that administering rcFVIIa intravenously is feasible, safe, well tolerated and efficacious in correcting the haemophilic coagulopathy in canine HA and that rcFVIIa exhibits pharmacokinetic characteristics comparable to rhFVIIa in haemophilic and haemostatically competent dogs. This strengthens the hypothesis that rcFVIIa can be administered to dogs to mimic the administration of rhFVIIa to humans.


Assuntos
Coagulação Sanguínea/efeitos dos fármacos , Fator VIIa/efeitos adversos , Fator VIIa/farmacocinética , Hemofilia A/tratamento farmacológico , Animais , Modelos Animais de Doenças , Cães , Feminino , Meia-Vida , Hemofilia A/metabolismo , Hemostasia/efeitos dos fármacos , Injeções Intravenosas , Masculino , Taxa de Depuração Metabólica , Projetos Piloto , Proteínas Recombinantes/efeitos adversos , Proteínas Recombinantes/farmacocinética , Tromboelastografia
7.
Haemophilia ; 17(5): e963-8, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21682818

RESUMO

N8, a new recombinant factor VIII (rFVIII) compound developed for the treatment of haemophilia A, is produced in Chinese hamster ovary (CHO) cells and formulated without human- or animal-derived materials. The aim of the present study was to compare the pharmacokinetics (PK) and the procoagulant effect, measured by ex vivo whole blood clot formation, of N8 and a commercial rFVIII in a cross-over study in haemophilia A dogs. N8 and Advate® (100 IU kg⁻¹) were administered intravenously to three haemophilia A dogs. Blood was sampled between 0 and 120 h postdose and FVIII:C analysed. PK parameters maximum plasma concentration, area under the curve, half-life (t(½)), clearance, mean residence time (MRT) and volume of distribution and incremental recovery were calculated. Whole blood clotting time (WBCT) and thromboelastography (TEG®) were used to determine the haemostatic potential. No adverse reactions were observed with N8 or Advate ®. N8 and Advate® exhibited similar PK parameters, with t(½) 7.7-11 h and MRT 11-14 h. Both rFVIII compounds corrected the prolonged WBCT (> 48 min) to the range of normal dogs (8-12 min), i.e. N8 to 7.5-10.5 min and Advate® to 7.5-11.5 min. N8 and Advate® also normalized the whole blood clot formation according to TEG®. The native whole blood clotting assays (WBCT, TEG®) appeared to be more sensitive to low concentrations of FVIII than assays in citrated plasma samples. In conclusion, comparison of N8 and Advate ® in haemophilia A dogs revealed similar safety, similar PK and similar effects in whole blood clot formation assays.


Assuntos
Coagulação Sanguínea/efeitos dos fármacos , Fator VIII/farmacocinética , Hemofilia A/sangue , Proteínas Recombinantes/farmacocinética , Animais , Estudos Cross-Over , Doenças do Cão/sangue , Doenças do Cão/tratamento farmacológico , Cães , Avaliação Pré-Clínica de Medicamentos/métodos , Feminino , Meia-Vida , Hemofilia A/tratamento farmacológico , Hemofilia A/imunologia , Hemofilia A/veterinária , Hemostasia/efeitos dos fármacos , Injeções Intravenosas , Masculino , Taxa de Depuração Metabólica
8.
Haemophilia ; 16(6): 878-87, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20546031

RESUMO

The aim of this study was to evaluate the in vitro function of the new recombinant factor VIII (FVIII) compound, N8. The specific activity of N8 as measured in a FVIII:C one-stage clot assay was 9300±400 IU mg(-1) based on the analysis of seven individual batches. The ratio between the FVIII:C activity measured in clot and chromogenic assays was 1.00 (95% confidence interval 0.97-1.03). N8 bound to von Willebrand factor with Kd values of 0.2 nm when measured by ELISA and by surface plasmon resonance. FVIIIa cofactor activity was determined from the kinetic parameters of factor IXa-catalysed factor X (FX) activation. The rate of activation of N8 by thrombin as well as Km and kcat for FX activation was in the same range as those observed for Advate®. The rate of activated protein C (APC)-catalysed inactivation was similar for activated N8 and Advate®. N8 improved thrombin generation in a dose-dependent manner and induced similar rates of thrombin generation as Advate® and the plasma-derived FVIII product Haemate®. Using thromboelastography (TEG®), N8 was shown to improve the clot formation and clot stability in whole blood from haemophilia A patients. Comparable potency and efficacy of N8 and Advate® was found based on TEG® parameters. Finally, similar binding profiles to immobilized lipoprotein receptor-related protein (LRP) of N8 and Advate® were observed. The study demonstrated that N8 is fully functional in a variety of assays measuring FVIII activity. No functional differences were found between N8 and comparator compounds.


Assuntos
Coagulação Sanguínea/efeitos dos fármacos , Fator VIII/análise , Fator VIIIa/análise , Hemofilia A/sangue , Hemofilia A/tratamento farmacológico , Ensaio de Imunoadsorção Enzimática , Fator VIIIa/farmacocinética , Humanos , Proteínas Recombinantes , Tromboelastografia , Trombina/biossíntese , Fator de von Willebrand/metabolismo
9.
Haemophilia ; 16(2): 349-59, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19906157

RESUMO

SUMMARY: A new recombinant factor VIII (FVIII), N8, has been produced in Chinese hamster ovary (CHO) cells. The molecule consists of a heavy chain of 88 kDa including a 21 amino acid residue truncated B-domain and a light chain of 79 kDa. The two chains are held together by non-covalent interactions. The four-step purification includes capture, affinity purification using a monoclonal recombinant antibody, anion exchange chromatography and gel filtration. The specific clotting activity of N8 was 8800-9800 IU mg(-1). Sequence and mass spectrometry analysis revealed two variants of the light chain, corresponding to two alternative N-terminal sequences also known from plasma FVIII. Two variants of the heavy chain are present in the purified product, namely with and without the B-domain linker attached. This linker is removed upon thrombin activation of N8 rendering an activated FVIII (FVIIIa) molecule similar to plasma FVIIIa. All six known tyrosine sulphations of FVIII were confirmed in N8. Two N-linked glycosylations are present in the A3 and C1 domain of the light chain and two in the A1 domain of the heavy chain. The majority of the N-linked glycans are sialylated bi-antennary structures. An O-glycosylation site is present in the B-domain linker region. This site was glycosylated with a doubly sialylated GalNAc-Gal structure in approximately 65% of the product. In conclusion, the present data show that N8 is a pure and well-characterized FVIII product with biochemical properties that equal other FVIII products.


Assuntos
Fator VIII/química , Sequência de Aminoácidos , Animais , Western Blotting , Cromatografia Líquida de Alta Pressão , Cromatografia por Troca Iônica , Cricetinae , Fator VIII/isolamento & purificação , Glicoproteínas/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/isolamento & purificação , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Tripsina/metabolismo
10.
Haemophilia ; 15(6): 1318-26, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19659796

RESUMO

Replacement therapy with factor VIII (FVIII) and factor IX (FIX) is routinely used in haemophilia patients with haemophilia A and B, respectively, while recombinant activated FVII (rFVIIa) has proven to induce haemostasis in haemophilia patients with inhibitors. To evaluate the effect of therapeutic intervention in patients with residual factor activities, the effects of increasing concentrations of rFVIIa or NN1731 on thrombin generation and platelet activation were measured in a cell-based model system mimicking severe, moderate and mild haemophilia A or B. Purified monocytes stimulated to express tissue factor and non-activated platelets from peripheral blood of healthy donors were incubated with a mixture of purified human coagulation factors in the absence or presence of increasing concentrations of FVIII or FIX. Sub-samples were analysed for thrombin activity and platelet activation measured as exposure of P-selectin by flow cytometry. Dose-dependent increases in thrombin generation and platelet activation were observed following increasing concentrations of rFVIIa or NN1731 in both haemophilia A- and B-like conditions. At 25 nm rFVIIa, which nears the peak levels in patient plasma after 90 microg kg(-1) intravenous dosing, the effects on maximum thrombin generation rate (maxTG) at 1-10% FVIII were comparable to those at 100% and 200% FVIII in the absence of rFVIIa. Normalization of maxTG required 500 nm rFVIIa and 25 nm NN1731 or 25-100 nm rFVIIa and 5 nm NN1731 in severe or moderate/mild haemophilia A and haemophilia B, respectively. This suggests that NN1731 holds its promise as a future bypassing agent for haemophilia patients with and without inhibitors.


Assuntos
Fator VII/farmacologia , Fator VIIa/farmacologia , Hemofilia A/metabolismo , Hemofilia B/metabolismo , Trombina/metabolismo , Fatores de Coagulação Sanguínea/farmacologia , Células Cultivadas , Relação Dose-Resposta a Droga , Fator VII/administração & dosagem , Fator VIIa/administração & dosagem , Humanos , Ativação Plaquetária/efeitos dos fármacos , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Trombina/efeitos dos fármacos
11.
J Thromb Haemost ; 5(4): 774-80, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17229056

RESUMO

BACKGROUND: Activation of platelets with a combination of collagen and thrombin generates a subpopulation of highly procoagulant 'coated' platelets characterized by high surface expression of fibrinogen and other procoagulant proteins. OBJECTIVES: To analyze the interaction of recombinant factor VIIa (rFVIIa) with coated platelets. METHODS AND RESULTS: rFVIIa localized to the coated platelets in flow cytometry experiments, while minimal rFVIIa was found on platelets activated with adenosine diphosphate, thrombin or via glycoprotein VI individually, and essentially no rFVIIa was found on non-stimulated platelets. Removal of the gamma-carboxyglutamic acid (Gla) domain of rFVIIa, and addition of EDTA, annexin V or excess prothrombin inhibited rFVIIa localization to the coated platelets, indicating that the interaction was mediated by the calcium-dependent conformation of the Gla domain and platelet exposure of negatively charged phospholipids. A reduced level of platelet fibrinogen exposure was observed at hemophilia A-like conditions in a model system of cell-based coagulation, indicating that coated platelet formation in hemophilia may be diminished. Addition of rFVIIa dose-dependently enhanced thrombin generation and partly restored platelet fibrinogen exposure. CONCLUSIONS: The data suggest that rFVIIa localized preferentially on platelets activated with dual agonists, thereby ensuring enhanced thrombin generation localized at the site of injury where both collagen and tissue factor are exposed, the latter ensuring the formation of thrombin necessary for coated platelet formation.


Assuntos
Plaquetas/metabolismo , Coagulantes/química , Fator VIIa/biossíntese , Glicoproteínas da Membrana de Plaquetas/metabolismo , Trombina/fisiologia , Ácido 1-Carboxiglutâmico/química , Sítios de Ligação , Coagulação Sanguínea , Colágeno/metabolismo , Citometria de Fluxo , Hemofilia A/patologia , Humanos , Fosfolipídeos/química , Estrutura Terciária de Proteína , Trombina/metabolismo , Tromboplastina/metabolismo
12.
Semin Hematol ; 38(4 Suppl 12): 15-20, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11735105

RESUMO

Recombinant factor VIIa (rFVIIa; NovoSeven, Novo Nordisk, Copenhagen, Denmark) may help to promote hemostasis in patients with thrombocytopenia. We used two in vitro models of thrombin generation to evaluate this effect. The reconstituted model contained tissue factor (TF)-expressing monocytes, unactivated platelets, isolated plasma coagulation proteins, and calcium. Platelet activation and thrombin generation were measured in timed aliquots. In the plasma-based model, thrombin generation was measured continuously after the addition of lipidated TF and calcium to platelet-rich plasma using a slowly cleaved fluorescent substrate. Thrombocytopenic conditions were mimicked by decreasing the platelet density. In both systems, a platelet density-dependent lowering of the thrombin-generation peak was observed. Addition of rFVIIa to samples with low platelet density (6700 to 10000/microL) increased the initial thrombin generation in both systems without normalizing thrombin-generation curves. The magnitude of the rFVIIa effect was most pronounced in the plasma-based model. Platelet activation was not significantly delayed at low platelet density in the reconstituted model. Addition of rFVIIa to samples with low platelet density caused faster platelet activation, most likely as a consequence of the increased initial thrombin generation. The data suggest that rFVIIa may help to achieve hemostasis at low platelet densities by increasing the initial thrombin generation, thereby compensating for low platelet number.


Assuntos
Fator VII/farmacologia , Modelos Biológicos , Proteínas Recombinantes/farmacologia , Trombocitopenia/tratamento farmacológico , Plaquetas/efeitos dos fármacos , Plaquetas/metabolismo , Coagulantes/farmacologia , Fator VIIa , Humanos , Cinética , Lipopolissacarídeos/farmacologia , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Trombina/biossíntese , Trombina/efeitos dos fármacos , Tromboplastina/efeitos dos fármacos , Tromboplastina/metabolismo
13.
Proc Natl Acad Sci U S A ; 98(24): 13583-8, 2001 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-11698657

RESUMO

A trace amount of coagulation factor VII (FVII) circulates in the blood in the activated form, FVIIa (EC 3.4.21.21), formed by internal proteolysis. To avoid disseminated thrombus formation, FVIIa remains in a conformation with zymogen-like properties. Association with tissue factor (TF), locally exposed upon vascular injury, is necessary to render FVIIa biologically active and initiate blood clotting. We have designed potent mutants of FVIIa by replacing residues believed to function as determinants for the inherent zymogenicity. The TF-independent rate of factor X activation was dramatically improved, up to about 100-fold faster than that obtained with the wild-type enzyme and close to that of the FVIIa-soluble TF complex. The mutants appear to retain the substrate specificity of the parent enzyme and can be further stimulated by TF. Insights into the mechanism behind the increased activity of the mutants, presumably also pertinent to the TF-induced, allosteric stimulation of FVIIa activity, were obtained by studying their calcium dependence and the accessibility of the N terminus of the protease domain to chemical modification. The FVIIa analogues promise to offer a more efficacious treatment of bleeding episodes especially in hemophiliacs with inhibitory antibodies precluding conventional replacement therapy.


Assuntos
Fator VIIa/metabolismo , Antitrombinas/farmacologia , Fator VIIa/antagonistas & inibidores , Fator VIIa/química , Fator VIIa/genética , Variação Genética , Mutagênese , Conformação Proteica , Relação Estrutura-Atividade , Tromboplastina/metabolismo
14.
Semin Thromb Hemost ; 27(4): 373-84, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11547359

RESUMO

Recombinant activated coagulation factor VII (rFVIIa) (NovoSeven) was developed for treatment of bleeding in hemophilia patients with inhibitors (antibodies) against factors VIII or IX. rFVIIa initiates the coagulation cascade by binding to tissue factor at the site of injury and causes the formation of sufficient amounts of thrombin to trigger coagulation. Patients with a variety of other coagulation deficiencies than hemophilia characterized by an impaired thrombin generation and life-threatening bleeding have been reported as successfully treated with rFVIIa. Data are now entered into clinical registries established to further monitor this experimental treatment with NovoSeven. rFVIIa is produced free of any added human protein. The amino acid sequence of rFVIIa is identical to plasma-derived FVIIa (pdFVIIa). Posttranslational modifications (i.e., gamma-carboxylations, N- and O-glycosylations) are qualitatively identical in pdFVIIa and rFVIIa although some quantitative differences exist. The activities of rFVIIa and pdFVIIa are indistinguishable. Manufacturing of rFVIIa involves expression in baby hamster kidney (BHK) cells followed by purification, including three ion-exchange and one immunoaffinity chromatography steps. The last anion-exchange chromatography step ensures completion of the autoactivation of recombinant factor VII (rFVII) to rFVIIa. This review describes the mechanism of action, characterization, manufacturing, and preclinical and current clinical evidence for the efficacy and safety of rFVIIa.


Assuntos
Fator VII/química , Fator VII/farmacologia , Microbiologia Industrial/métodos , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacologia , Animais , Ensaios Clínicos como Assunto , Clonagem Molecular , Fator VII/uso terapêutico , Fator VIIa , Humanos , Estrutura Molecular , Proteínas Recombinantes/uso terapêutico , Transformação Genética
15.
Br J Haematol ; 114(1): 114-20, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11472355

RESUMO

Clinical experience has shown that high doses of recombinant factor VIIa (rFVIIa) may ensure haemostasis in thrombocytopenic patients. We have used a cell-based model system to mimic thrombocytopenia and analyse the effect of rFVIIa. Lowering the platelet density from 200 x 10(9)/l (reflecting normal conditions) to 100, 50, 20 and 10 x 10(9)/l revealed a platelet density-dependent decrease in the maximal rate of thrombin generation, a prolongation in the time to maximal thrombin activity and a lower maximal level of thrombin formed. The platelet activation, measured as the time to half-maximal P-selectin (CD62) exposure, was not significantly dependent on the platelet density in the range of 200 x 10(9)/l to 10 x 10(9)/l, although there was a tendency for slower platelet activation at 20 x 10(9) and 10 x 10(9) platelets/l than at the higher platelet densities. Addition of 50--500 nmol/l rFVIIa to samples with 20 x 10(9) or 10 x 10(9) platelets/l shortened the lag phase of thrombin generation as well as the time to half-maximal platelet activation. Our data indicate that high doses of rFVIIa may help to provide haemostasis in thrombocytopenic patients by increasing the initial thrombin generation, resulting in faster platelet activation and thereby compensating for the lower number of platelets present.


Assuntos
Fator VIIa/administração & dosagem , Ativação Plaquetária , Trombina/biossíntese , Trombocitopenia/terapia , Células Cultivadas , Esquema de Medicação , Hemostasia , Humanos , Contagem de Plaquetas , Proteínas Recombinantes/administração & dosagem
16.
Arterioscler Thromb Vasc Biol ; 20(7): 1835-41, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10894826

RESUMO

The effect of plasma lipoprotein fractions (large very-low-density lipoprotein, small very-low-density lipoprotein, intermediate-density lipoprotein, and low-density lipoprotein) on initiation of blood coagulation by supporting factor VII activation or by stimulating monocytes to express tissue factor was investigated in vitro. Endotoxin-free preparations of lipoprotein fractions did not induce functional tissue factor in monocytes, whereas all lipoprotein fractions enhanced tissue factor-independent activation of factor VII by factor Xa and by factors Xa/Va. In contrast, no or only slight enhancement of factor IXa-, factor IXa/VIIIa-, factor XIa-, or thrombin-mediated factor VII activation was observed. The effect of small very-low-density lipoprotein was less than that of large very-low-density lipoprotein, and intermediate-density and low-density lipoproteins caused an even lower but still significant increase of factor Xa- and factor Xa/Va-mediated factor VII activation. When the data were normalized for apolipoprotein B-100 content, differences remained between lipoprotein fractions. In contrast, when phospholipid content was used for normalization, differences between lipoprotein fractions in factor Xa- and factor Xa/Va-mediated factor VII activation disappeared, indicating that phospholipids were involved in factor VII activation. This was supported by enhancement of factor Xa-mediated factor VII activation by synthetic phospholipid vesicles containing negatively charged phospholipids.


Assuntos
Colesterol/farmacologia , Fator VII/metabolismo , Lipoproteínas/farmacologia , Tromboplastina/metabolismo , Coagulação Sanguínea , Colesterol/sangue , Fator X/metabolismo , Fator XIa/metabolismo , Fator Xa/metabolismo , Humanos , Técnicas In Vitro , Lipoproteínas/sangue , Lipoproteínas LDL/sangue , Lipoproteínas LDL/farmacologia , Lipoproteínas VLDL/sangue , Lipoproteínas VLDL/farmacologia , Masculino , Pessoa de Meia-Idade , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Ativação Plaquetária/fisiologia
17.
Thromb Haemost ; 80(4): 578-84, 1998 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9798973

RESUMO

Factors VIIa, Xa, and IXa play different roles in the initiation of tissue factor-dependent coagulation. The consequences of competing with the different enzymes were investigated, thereby examining the effects of inhibiting the initiation process at different steps. Active site-inactivated factors VIIa, Xa, and IXa (FVIIai, FXai, and FIXai, respectively) were added to various cell-based assays mimicking the individual steps in tissue factor-initiated coagulation. In an assay involving tissue factor-expressing monocytes, coagulation proteins and unactivated platelets, FVIIai and FXai inhibited platelet activation and thrombin generation while FIXai only inhibited thrombin generation. FVIIai inhibited factor Xa generation and subsequent thrombin generation on monocytes, while FXai inhibited thrombin generation on the monocytes as well as on the activated platelets. FIXai had no effect on factor Xa or thrombin generation on the monocytes, but inhibited factor Xa and subsequent thrombin generation on the activated platelets. FVIIai had no effect on the reactions taking place on the activated platelets. The data confirm a model where tissue factor/factor VIIa mediates factor Xa generation and subsequent prothrombin activation on the tissue factor-bearing cells. Thrombin then activates platelets, which serve as the physiologically important surface for large-scale thrombin generation.


Assuntos
Bioensaio , Coagulação Sanguínea , Fator VIIa/metabolismo , Fator XIa/metabolismo , Fator Xa/metabolismo , Tromboplastina/metabolismo , Adulto , Coagulação Sanguínea/efeitos dos fármacos , Plaquetas/citologia , Plaquetas/fisiologia , Fator VIIa/química , Fator XIa/química , Fator Xa/química , Humanos , Monócitos/citologia , Monócitos/fisiologia , Tromboplastina/farmacologia
18.
Blood Coagul Fibrinolysis ; 9 Suppl 1: S21-5, 1998 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9819025

RESUMO

The importance of activated factor VII (FVIIa) in coagulation initiated by tissue factor (TF) was illustrated by competition of active site-inhibited FVIIa (FFR-FVIIa; FVIIa treated with D-Phe-Phe-Arg-chloromethyl ketone) with FVIIa in various cell-based assays mimicking TF-initiated coagulation. FFR-FVIIa inhibited the overall initiation process as measured by platelet activation and large-scale thrombin generation on the activated platelet surface. When the individual steps in the initiation process were separated, FFR-FVIIa affected only the reactions taking place on TF-bearing cells, demonstrating that FVIIa takes part only in the very first step in the initiation process. The dissociation constant (Kd) for FVIIa binding to TF and the inhibition constant (Ki) for FFR-FVIIa competing with FVIIa in binding to TF, measured in a factor X activation assay, were both around 10 pmol/l, showing that FVIIa and FFR-FVIIa bound to TF in the extrinsic pathway tenase complex with the same affinity.


Assuntos
Coagulação Sanguínea/efeitos dos fármacos , Fator VIIa/farmacologia , Modelos Biológicos , Monócitos/efeitos dos fármacos , Tromboplastina/fisiologia , Clorometilcetonas de Aminoácidos/farmacologia , Humanos , Ativação Plaquetária , Inibidores de Serina Proteinase/farmacologia
19.
Thromb Haemost ; 78(4): 1202-8, 1997 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9364985

RESUMO

Active site-inactivated factor VIIa has potential as an antithrombotic agent. The effects of D-Phe-L-Phe-L-Arg-chloromethyl ketone-treated factor VIIa (FFR-FVIIa) were evaluated in a cell-based system mimicking in vivo initiation of coagulation. FFR-FVIIa inhibited platelet activation (as measured by expression of P-selectin) and subsequent large-scale thrombin generation in a dose-dependent manner with IC50 values of 1.4 +/- 0.8 nM (n = 8) and 0.9 +/- 0.7 nM (n = 7), respectively. Kd for factor VIIa binding to monocytes and Ki for FFR-FVIIa competing with factor VIIa were similar (11.4 +/- 0.8 pM and 10.6 +/- 1.1 pM, respectively), showing that FFR-FVIIa binds to tissue factor in the tenase complex with the same affinity as factor VIIa. Using platelets from volunteers before and after ingestion of aspirin (1.3 g), there were no significant differences in the IC50 values of FFR-FVIIa [after aspirin ingestion, the IC50 values were 1.7 +/- 0.9 nM (n = 8) for P-selectin expression, p = 0.37, and 1.4 +/- 1.3 nM (n = 7) for thrombin generation, p = 0.38]. This shows that aspirin treatment of platelets does not influence the inhibition of tissue factor-initiated coagulation by FFR-FVIIa, probably because thrombin activation of platelets is not entirely dependent upon expression of thromboxane A2.


Assuntos
Clorometilcetonas de Aminoácidos/farmacologia , Aspirina/farmacologia , Coagulação Sanguínea/efeitos dos fármacos , Plaquetas/efeitos dos fármacos , Inibidores de Ciclo-Oxigenase/farmacologia , Fator VII/farmacologia , Fibrinolíticos/farmacologia , Inibidores da Agregação Plaquetária/farmacologia , Agregação Plaquetária/efeitos dos fármacos , Inibidores de Serina Proteinase/farmacologia , Tromboplastina/farmacologia , Adulto , Sítios de Ligação , Ligação Competitiva , Plaquetas/metabolismo , Cálcio/farmacologia , Células Cultivadas , Esquema de Medicação , Interações Medicamentosas , Fator VII/antagonistas & inibidores , Fator VIIa , Humanos , Cinética , Lipopolissacarídeos/farmacologia , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Selectina-P/metabolismo , Ligação Proteica , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/farmacologia , Trombina/biossíntese , Tromboplastina/metabolismo
20.
J Biol Chem ; 272(18): 11863-8, 1997 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-9115245

RESUMO

Recent studies showed that the administration of active site-inhibited factor VIIa blocked factor VIIa/tissue factor-induced fibrin and thrombus formation in ex vivo and in vivo model systems. These studies suggest that inactivated factor VIIa competes efficiently with plasma factor VII(a) for a limited number of tissue factor sites. In the present study, we compared the interactions of factor VIIa and active site-inhibited factor VIIa with tissue factor. Competition studies of factor VIIa and active site-inhibited factor VIIa in a factor X activation assay showed that the affinity of the latter for relipidated tissue factor was 5-fold higher than that of factor VIIa. Radioligand binding studies with a human bladder carcinoma cell line (J82) and surface plasmon resonance studies using soluble tissue factor demonstrated a faster association and a slower dissociation for the active site-inhibited factor VIIa. Studies of equilibrium binding to cell surface tissue factor showed that the affinity of active site-inhibited VIIa was 5-fold higher than that of factor VIIa to non-functional tissue factor sites, whereas both inactivated factor VIIa and factor VIIa bound to functional tissue factor sites with the same high affinity. Comparison of the CD spectra of factor VIIa and active site-inactivated factor VIIa revealed structural differences in the protease domain. The potential physiological implications of these findings are discussed.


Assuntos
Clorometilcetonas de Aminoácidos/farmacologia , Fator VIIa/metabolismo , Tromboplastina/metabolismo , Sítios de Ligação , Ligação Competitiva , Linhagem Celular , Membrana Celular/metabolismo , Dicroísmo Circular , Fator VIIa/antagonistas & inibidores , Fator VIIa/química , Fibrina/metabolismo , Humanos , Cinética , Conformação Proteica , Proteínas Recombinantes/metabolismo , Células Tumorais Cultivadas , Neoplasias da Bexiga Urinária
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...