Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Thromb Haemost ; 2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38815755

RESUMO

BACKGROUND: Inhibition of tissue factor pathway inhibitor (TFPI) is an emerging therapeutic strategy for treatment of hemophilia. Concizumab is a monoclonal antibody that binds TFPI and blocks its inhibition of factor (F)Xa thereby extending the initiation of coagulation and compensating for lack of FVIII or FIX. OBJECTIVES: The objective of this in vitro study was to evaluate how concizumab affects clot formation in hemophilia A under flow. METHODS: Blood was collected from normal controls or people with hemophilia A. An anti-FVIII antibody was added to normal controls to simulate hemophilia A with inhibitory antibodies to FVIII. Whole blood added activated recombinant factor VII (rFVIIa, 25 nM) or concizumab (200, 1000, 4000 ng/mL) were perfused at 100 s-1 over a surface micropatterned with tissue factor (TF) and collagen related peptide. Platelet and fibrin(ogen) accumulation were measured by confocal microscopy. Static thrombin generation in plasma was measured in response to rFVIIa and concizumab. RESULTS: Concizumab (1000, 4000 ng/mL) and rFVIIa both rescued (93-101%) total platelet accumulation, but only partially rescued (53-63%) fibrin(ogen) incorporation to normal control levels in simulated hemophilia A. Results in congenital haemophilia A blood confirmed effects of rFVIIa and concizumab. While these two agents had similar effect on clot formation under flow, concizumab enhanced thrombin generation in plasma under static conditions to a greater extent than rFVIIa. CONCLUSIONS: TFPI inhibition by concizumab enhanced activation and aggregation of platelets and fibrin clot formation in hemophilia A to levels comparable to that of rFVIIa.

2.
J Thromb Haemost ; 20(6): 1312-1324, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35191180

RESUMO

BACKGROUND: Mim8 is a novel, next-generation factor VIIIa mimetic in development for subcutaneous prophylactic treatment of patients with hemophilia A with and without inhibitors. In vitro and in vivo models indicate that Mim8 has a distinct hemostatic potential. OBJECTIVES: To test the nonclinical safety and pharmacodynamics of Mim8. METHODS: The Mim8 nonclinical safety program in cynomolgus monkeys consisted of three studies of 4-26 weeks in duration with Mim8 doses ranging from 0.3-60 mg/kg/week intravenously or subcutaneously. After sacrifice, macroscopic and microscopic pathological examinations were performed. RESULTS: Mim8 was well tolerated with no noteworthy clinical observations. No signs of excessive coagulation or pathological macroscopic or microscopic findings were observed at doses 0.3-3 mg/kg/week subcutaneous. Thrombosis-related findings were detected during histopathological examination in a small proportion of animals (16%) receiving doses ranging 6-20 mg/kg/week. Dose-dependent increases in factor X (FX) and factor IX (FIX) concentrations were observed. Shortening of activated partial thromboplastin time (APTT) and increased thrombin generation under ex vivo hemophilia A-like conditions were observed at all Mim8 dose levels. CONCLUSIONS: Thrombosis-related findings observed at doses above 6 mg/kg/week Mim8 may have been exaggerated pharmacological reactions to a procoagulant compound in normocoagulant animals. Increases in FX and FIX concentrations could be because of a half-life prolongation due to binding to Mim8, but were limited at clinically relevant exposure levels. Subcutaneous administration of up to 3 mg/kg/week (several fold greater than expected clinical exposure) for 26 weeks resulted in relevant pharmacodynamic effects, observed in thrombin generation and APTT, with no signs of thrombi or excessive coagulation activation.


Assuntos
Hemofilia A , Trombose , Animais , Fator IX/metabolismo , Fator X , Humanos , Macaca fascicularis/metabolismo , Trombina/metabolismo , Trombose/prevenção & controle
3.
J Thromb Haemost ; 19(7): 1687-1696, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33819375

RESUMO

BACKGROUND: The anti-tissue factor plasma inhibitor monoclonal antibody concizumab is under clinical investigation for subcutaneous prophylaxis of hemophilia A/B (HA/HB) with or without inhibitors. Breakthrough bleeds while on concizumab prophylaxis may be treated with bypassing agents (recombinant activated factor VIIa [rFVIIa] and activated prothrombin complex concentrate [APCC]), or with factor VIII (FVIII) or factor IX (FIX). OBJECTIVES: To evaluate the effect of combining concizumab with rFVIIa, APCC, rFVIII, and rFIX on thrombin generation (TG) potential. METHODS: Pooled HA plasma was spiked in vitro with concizumab alone or together with rFVIIa, APCC, or rFVIII. rFVIIa, APCC, and rFVIII were added ex vivo to plasma from HA patients receiving concizumab prophylaxis. Pooled HB plasma was spiked with concizumab alone or together with rFIX. TG potential was measured after initiation with tissue factor. RESULTS: Concizumab increased thrombin peak in a concentration-dependent manner. Adding rFVIIa, APCC, rFVIII, or rFIX caused a further increase in thrombin peak. The effects of concizumab and rFVIIa, APCC, rFVIII, or rFIX were mainly additive, with no or up to maximally ~25% extra effect caused by drug--drug interaction. No strong synergistic effects were observed upon combining concizumab with rFVIIa, APCC, rFVIII, or rFIX. The thrombin peak obtained with 0.5 IU/ml rFVIII or rFIX in the presence of concizumab was on occasion slightly higher, but mostly comparable to the thrombin peak with 1 IU/ml rFVIII or rFIX in the absence of concizumab. CONCLUSION: rFVIIa, APCC, rFVIII, and rFIX enhanced plasma TG potential in the presence of concizumab. Dose levels of concomitant use should be adjusted accordingly to balance potential safety concerns while maintaining the necessary hemostatic effect. Please see the video in the Supplementary Material for an animated summary of the data presented.


Assuntos
Fator VIIa , Hemofilia A , Anticorpos Monoclonais Humanizados , Fatores de Coagulação Sanguínea , Fator IX , Fator VIII , Hemofilia A/tratamento farmacológico , Humanos , Proteínas Recombinantes , Trombina
4.
J Thromb Haemost ; 18(1): 104-113, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31549480

RESUMO

BACKGROUND: Recombinant factor VIIa (rFVIIa) enhances thrombin generation in a platelet-dependent manner; however, rFVIIa binds activated platelets with relatively low affinity. Triggering receptor expressed on myeloid cells (TREM)-like transcript (TLT)-1 is expressed exclusively on activated platelets. OBJECTIVE: To enhance the potency of rFVIIa via binding TLT-1. METHODS: Recombinant FVIIa was conjugated to a TLT-1 binding Fab. In vitro potency of this platelet-targeted rFVIIa (PT-rFVIIa) was evaluated using factor X activation assays and by measuring viscoelastic changes in whole blood. In vivo potency was evaluated using a tail vein transection model in F8-/- mice expressing human TLT-1. RESULTS: PT-rFVIIa and rFVIIa had similar dissociation constant values for tissue factor binding and similar tissue factor-dependent factor X activation. However, PT-rFVIIa had increased catalytic efficiency on TLT-1-loaded vesicles and activated platelets. The in vitro potency in normal human blood with antibody-induced hemophilia A was dependent on assay conditions used; with maximally activated platelets, the half maximal effective concentration for clot time for PT-rFVIIa was 49-fold lower compared with rFVIIa. In the murine bleeding model, a 53-fold lower half maximal effective concentration was observed for blood loss for PT-rFVIIa, supporting the relevance of the assay conditions with maximally activated platelets. In vitro analysis of blood from subjects with hemophilia A confirmed the data obtained with normal blood. CONCLUSIONS: Increasing the affinity of rFVIIa to activated platelets resulted in approximately 50-fold increased potency both in vitro and in the mouse model. The correlation of in vivo with in vitro data using maximally activated platelets supports that these assay conditions are relevant when evaluating platelet-targeted hemostatic concepts.


Assuntos
Plaquetas , Hemofilia A , Animais , Fator VIIa , Hemofilia A/tratamento farmacológico , Camundongos , Proteínas Recombinantes , Trombina
5.
Haemophilia ; 26(1): 156-163, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31809565

RESUMO

Factor replacement therapy with factor VIII (FVIII) concentrates is the current standard of care for patients with haemophilia A. Postadministration monitoring of FVIII activity during on-demand or prophylactic treatment is important, for example to guide a suitable dosing regimen. While the use of two-stage chromogenic substrate (CS) assays is increasing, activated partial thromboplastin time (APTT)-based one-stage clotting (OSC) assays are most commonly used to measure FVIII activity in clinical laboratories. Substantial variations in activity measurements have been observed in association with some OSC assay reagents when assessing extended half-life FVIII molecules. Certain silica-based APTT reagents have previously been shown to underestimate FVIII activity with the polyethylene glycol (PEG)-conjugated product turoctocog alfa pegol (N8-GP [ESPEROCT® ]; Novo Nordisk A/S). As a wide range of assay reagents are used in clinical laboratories worldwide, it is essential to establish which can be used to accurately measure activity with modified FVIII concentrates. Here, we describe the approach taken by Novo Nordisk to determine the suitability and accuracy of assays and reagents to measure FVIII activity in samples that contain N8-GP. While accurate activity measurements were possible with all tested CS assays and most of the OSC APTT reagents tested, three APTT reagents that contain silica as a contact activator were found to underestimate N8-GP recovery (APTT-SP, TriniCLOT™, STA® PTT-Automate). The data demonstrate the importance of characterizing the accuracy of each FVIII activity assay. Any limitations should be communicated to treating physicians and the clinical laboratories that test samples containing N8-GP.


Assuntos
Bioensaio , Monitoramento de Medicamentos , Fator VIII/administração & dosagem , Fator VIII/uso terapêutico , Polietilenoglicóis/química , Glicosilação , Humanos , Tempo de Tromboplastina Parcial
6.
Eur J Haematol ; 93(5): 369-76, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24797664

RESUMO

Turoctocog alfa (NovoEight®) is a recombinant factor VIII (rFVIII) with a truncated B-domain made from the sequence coding for 10 amino acids from the N-terminus and 11 amino acids from the C-terminus of the naturally occurring B-domain. Turoctocog alfa is produced in Chinese hamster ovary (CHO) cells without addition of any human- or animal-derived materials. During secretion, some rFVIII molecules are cleaved at the C-terminal of the heavy chain (HC) at amino acid 720, and a monoclonal antibody binding C-terminal to this position is used in the purification process allowing isolation of the intact rFVIII. Viral inactivation is ensured by a detergent inactivation step as well as a 20-nm nano-filtration step. Characterisation of the purified protein demonstrated that turoctocog alfa was fully sulphated at Tyr346 and Tyr1664, which is required for optimal proteolytic activation by thrombin. Kinetic assessments confirmed that turoctocog alfa was activated by thrombin at a similar rate as seen for other rFVIII products fully sulphated at these positions. Tyr1680 was also fully sulphated in turoctocog alfa resulting in strong affinity (low nm Kd ) for binding to von Willebrand factor (VWF). Half-lives of 7.2 ± 0.9 h in F8-KO mice and 8.9 ± 1.8 h haemophilia A dogs supported that turoctocog alfa bound to VWF after infusion. Functional studies including thromboelastography analysis of human haemophilia A whole blood with added turoctocog alfa and effect studies in mice bleeding models demonstrated a dose-dependent effect of turoctocog alfa. The non-clinical data thus confirm the haemostatic effect of turoctocog alfa and, together with the comprehensive clinical evaluation, support the use as FVIII replacement therapy in patients with haemophilia A.


Assuntos
Fator VIII/farmacocinética , Processamento de Proteína Pós-Traducional , Animais , Células CHO , Cricetulus , Cães , Avaliação Pré-Clínica de Medicamentos , Fator VIII/biossíntese , Fator VIII/genética , Fator VIII/isolamento & purificação , Expressão Gênica , Glicosilação , Meia-Vida , Hemofilia A/tratamento farmacológico , Humanos , Cinética , Camundongos , Modificação Traducional de Proteínas , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/farmacocinética , Sulfatos , Trombina/metabolismo
7.
J Biol Chem ; 288(41): 29670-9, 2013 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-24009077

RESUMO

The C1 domain of factor VIII (FVIII) has been implicated in binding to multiple constituents, including phospholipids, von Willebrand factor, and low-density lipoprotein receptor-related protein (LRP). We have previously described a human monoclonal antibody called KM33 that blocks these interactions as well as cellular uptake by LRP-expressing cells. To unambiguously identify the apparent "hot spot" on FVIII to which this antibody binds, we have employed hydrogen-deuterium exchange mass spectrometry. The results showed that KM33 protects FVIII regions 2091-2104 and 2157-2162 from hydrogen-deuterium exchange. These comprise the two C1 domain spikes 2092-2093 and 2158-2159. Spike 2092-2093 has been demonstrated recently to contribute to assembly with lipid membranes with low phosphatidylserine (PS) content. Therefore, spike 2158-2159 might serve a similar role. This was assessed by replacement of Arg-2159 for Asn, which introduces a motif for N-linked glycosylation. Binding studies revealed that the purified, glycosylated R2159N variant had lost its interaction with antibody KM33 but retained substantial binding to von Willebrand factor and LRP. Cellular uptake of the R2159N variant was reduced both by LRP-expressing U87-MG cells and by human monocyte-derived dendritic cells. FVIII activity was virtually normal on membranes containing 15% PS but reduced at low PS content. These findings suggest that the C1 domain spikes 2092-2093 and 2158-2159 together modulate FVIII membrane assembly by a subtle, PS-dependent mechanism. These findings contribute evidence in favor of an increasingly important role of the C1 domain in FVIII biology.


Assuntos
Endocitose , Fator VIII/química , Fator VIII/metabolismo , Estrutura Terciária de Proteína , Anticorpos Monoclonais/metabolismo , Anticorpos Monoclonais/farmacologia , Sítios de Ligação/genética , Medição da Troca de Deutério , Fator VIII/genética , Glicosilação , Humanos , Proteínas Relacionadas a Receptor de LDL/química , Proteínas Relacionadas a Receptor de LDL/metabolismo , Espectrometria de Massas , Modelos Moleculares , Mutação , Fosfatidilserinas/química , Fosfatidilserinas/metabolismo , Ligação Proteica/efeitos dos fármacos , Ressonância de Plasmônio de Superfície , Fator de von Willebrand/química , Fator de von Willebrand/metabolismo
8.
Blood ; 121(11): 2108-16, 2013 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-23335368

RESUMO

Frequent infusions of intravenous factor VIII (FVIII) are required to prevent bleeding associated with hemophilia A. To reduce the treatment burden, recombinant FVIII with a longer half-life was developed without changing the protein structure. FVIII-polyethylene glycol (PEG) conjugates were prepared using an enzymatic process coupling PEG (ranging from 10 to 80 kDa) selectively to a unique O-linked glycan in the FVIII B-domain. Binding to von Willebrand factor (VWF) was maintained for all conjugates. Upon cleavage by thrombin, the B-domain and the associated PEG were released, generating activated FVIII (FVIIIa) with the same primary structure and specific activity as native FVIIIa. In both FVIII- and VWF-deficient mice, the half-life was found to increase with the size of PEG. In vivo potency and efficacy of FVIII conjugated with a 40-kDa PEG (N8-GP) and unmodified FVIII were not different. N8-GP had a longer duration of effect in FVIII-deficient mouse models, approximately a twofold prolonged half-life in mice, rabbits, and cynomolgus monkeys; however, the prolongation was less pronounced in rats. Binding capacity of N8-GP on human monocyte-derived dendritic cells was reduced compared with unmodified FVIII, resulting in several-fold reduced cellular uptake. In conclusion, N8-GP has the potential to offer efficacious prevention and treatment of bleeds in hemophilia A at reduced dosing frequency.


Assuntos
Fator VIII/uso terapêutico , Hemofilia A/tratamento farmacológico , Polietilenoglicóis/uso terapêutico , Animais , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Modelos Animais de Doenças , Fator VIII/administração & dosagem , Fator VIII/metabolismo , Feminino , Glicosilação , Hemofilia A/patologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Biológicos , Polietilenoglicóis/administração & dosagem , Polietilenoglicóis/metabolismo , Coelhos , Ratos , Ratos Sprague-Dawley , Fatores de Tempo , Resultado do Tratamento
9.
Am J Vet Res ; 72(8): 1007-14, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21801056

RESUMO

OBJECTIVE: To develop an antibody-based flow cytometric assay to detect coated platelets in dogs and to characterize the interaction of recombinant human coagulation factor VIIa with activated platelets from dogs with hemophilia A. SAMPLE: Platelets from 4 dogs with hemophilia A, 4 dogs with hemophilia B, 4 dogs with von Willebrand disease, and 6 hemostatically normal dogs. PROCEDURES: Freshly isolated platelets were activated with thrombin, convulxin, or a thrombin-convulxin combination. Resulting platelet phenotypes were resolved on the basis of P-selectin and fibrinogen expression, and binding of recombinant human coagulation factor VIIa to these distinct platelet subpopulations was measured by use of a flow cytometric assay. RESULTS: Coated platelets were identified on the basis of expression of α-granule fibrinogen and were generated in response to stimulation with the thrombin-convulxin combination but not to stimulation with either agonist alone. Approximately 70% of the platelets from dogs with hemophilia A, hemophilia B, and von Willebrand disease and from the control dogs had the coated platelet phenotype. Recombinant human coagulation factor VIIa bound preferentially to coated platelets with a mean ± SD binding equilibrium constant of 2.6 ± 0.5µM. CONCLUSIONS AND CLINICAL RELEVANCE: Formation of coated platelets in dogs was similar to that in humans. Recombinant human coagulation factor VIIa bound preferentially to coated platelets from dogs. IMPACT FOR HUMAN MEDICINE: A similar mechanism of action for recombinant human coagulation factor VIIa may exist in dogs and humans. The potential for use of dogs in the study of bleeding disorders in humans was strengthened.


Assuntos
Plaquetas/metabolismo , Fator VIIa/metabolismo , Citometria de Fluxo/veterinária , Ativação Plaquetária , Animais , Anticorpos/análise , Anticorpos/metabolismo , Plaquetas/citologia , Venenos de Crotalídeos/metabolismo , Modelos Animais de Doenças , Doenças do Cão/metabolismo , Doenças do Cão/fisiopatologia , Cães , Fibrinogênio/análise , Fibrinogênio/metabolismo , Citometria de Fluxo/métodos , Hemofilia A/metabolismo , Hemofilia A/fisiopatologia , Hemofilia A/veterinária , Hemofilia B/metabolismo , Hemofilia B/fisiopatologia , Hemofilia B/veterinária , Humanos , Lectinas Tipo C/metabolismo , Selectina-P/análise , Selectina-P/metabolismo , Ligação Proteica , Proteínas Recombinantes/metabolismo , Trombina/metabolismo , Doenças de von Willebrand/metabolismo , Doenças de von Willebrand/fisiopatologia , Doenças de von Willebrand/veterinária
10.
Vet J ; 190(3): 352-8, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21216638

RESUMO

Canine coagulation factor VII (FVII) deficiency can be hereditary or acquired and may cause life threatening bleeding episodes if untreated. FVII procoagulant activity can be measured by FVII activity (FVII:C), but assays for measurement of canine specific FVII antigen (FVII:Ag) have not been available to date. In this study, a canine specific ELISA for measurement of FVII:Ag in plasma was developed and validated. The FVII:Ag ELISA correctly diagnosed homozygous and heterozygous hereditary FVII deficiency. Together with activity based assays, such as FVII:C, the FVII:Ag ELISA should be valuable in the diagnosis of hereditary canine FVII deficiency.


Assuntos
Antígenos/sangue , Doenças do Cão/diagnóstico , Ensaio de Imunoadsorção Enzimática/veterinária , Deficiência do Fator VII/veterinária , Animais , Doenças do Cão/genética , Cães , Fator VII , Deficiência do Fator VII/diagnóstico , Deficiência do Fator VII/genética , Camundongos , Reprodutibilidade dos Testes
11.
Biol Chem ; 390(12): 1279-83, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19804366

RESUMO

Coagulation factor XIII (FXIII) is activated by thrombin and catalyses crosslinking between fibrin monomers thereby providing mechanical strength to the fibrin network. V34L is a common FXIII-A polymorphism found in the activation peptide. FXIII-A V34L is activated faster by thrombin and provides formation of a tighter clot at fibrinogen concentrations in the low end of the physiological range. FXIII-A variants with potentially increased activation rates were generated. Introduction of an optimal thrombin cleavage site, V34L+V35T, increased the activation rate 7.6-fold and facilitated the formation of a fibrin network more resistant to fibrinolysis than obtained with wt FXIII-A. In contrast, introduction of fragments of fibrinopeptide A into the activation peptide resulted in severely impaired activation rates.


Assuntos
Processamento Alternativo , Fator XIII/metabolismo , Trombina/metabolismo , Sequência de Aminoácidos , Ativação Enzimática , Fator XIII/química , Fator XIII/genética , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Quaternária de Proteína
12.
J Extra Corpor Technol ; 41(2): 97-104, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19681308

RESUMO

Recombinant factor VIIa (rFVIIa) interacts preferentially with coated platelets characterized by a high exposure of phosphatidyl serine (PS), FV, FVIII, FIX, and FX binding, and fibrinogen. Cardiopulmonary bypass (CPB) is known to impair platelet function. In this study, the influence of CPB on formation of coated platelets and the interaction of rFVIIa with the platelets were studied. Blood was either exposed to a closed CPB circuit or obtained from patients undergoing CPB-assisted cardiac surgery, and platelets were analyzed by flow cytometry with and without dual agonist stimulation with thrombin and a GPVI collagen receptor agonist known to induce coated platelet formation. Platelets circulated within a closed CPB circuit did not spontaneously form coated platelets. Dual agonists stimulation caused formation of coated platelets at a reduced level compared to pre-CPB level (51 +/- 21% vs. 80 +/- 17% before CPB, p < .001). The rFVIIa interaction with the coated platelets was not impaired after CPB. Platelets isolated from patients undergoing CPB-assisted cardiac surgery also formed coated platelets only after dual agonist stimulation but to the same level as before surgery (76 +/- 8% vs. 83 +/- 14% before surgery, p = .17, n = 10). rFVIIa interaction with the coated platelets was not impaired after surgery. No spontaneous rFVIIa-binding platelets were found. The data indicate that CPB exposure in vivo does not compromise the platelet-dependent effects of rFVIIa either by spontaneous formation of coated platelets, thereby limiting the risk of systemic coagulation, or by impairing rFVIIa interaction with the agonist-induced coated platelets, thereby retaining the hemostatic potential of rFVIIa after CPB.


Assuntos
Plaquetas/efeitos dos fármacos , Ponte Cardiopulmonar , Fator VIIa/farmacologia , Plaquetas/metabolismo , Ponte de Artéria Coronária , Interpretação Estatística de Dados , Fibrinogênio/metabolismo , Citometria de Fluxo , Máquina Coração-Pulmão , Humanos , Selectina-P/metabolismo , Proteínas Recombinantes/sangue , Proteínas Recombinantes/farmacologia
13.
Br J Haematol ; 143(4): 559-69, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18950467

RESUMO

The haemostatic effect of recombinant activated factor VII (rFVIIa;NovoSeven) in thrombocytopenic patients has been a matter of controversy. Haemostasis by rFVIIa occurs via FVIIa-mediated thrombin generation in a platelet-dependent manner and may therefore be suboptimal in patients without functional platelets. Under such conditions, a clot-stabilizing agent, such as factor XIII (FXIII), may supplement the effect ofrFVIIa and improve haemostasis. Recombinant factor XIII (rFXIII-A2) is produced as an A2 homodimer of the FXIII A subunit and is equivalent to cellular FXIII normally found in platelets. The combined effects of rFVIIa andrFXIII-A2 were evaluated in clot lysis assays using factor XIII-deficient plasma and by whole blood thrombelastography (TEG) analysis from normal donors and thrombocytopenic stem cell transplantation patients. Clotting time was shortened by rFVIIa (0.6-10 microg/ml). rFVIIa only modestly improved anti-fibrinolysis,whereas rFXIII-A2 (0-20 microg/ml) enhanced anti-fibrinolysis without effect on clotting time. TEG analysis showed rFVIIa shortened the clotting time, and enhanced clot development, maximal mechanical strength and resistance to fibrinolysis, whereas, rFXIII-A2 enhanced clot development,maximal mechanical strength and markedly enhanced resistance to fibrinolysis. These data illustrate that rFVIIa and rFXIII-A2 contribute to clot formation and stability by different mechanisms suggesting enhanced haemostatic efficacy by combining these agents.


Assuntos
Coagulação Sanguínea/efeitos dos fármacos , Fator VIIa/farmacologia , Fator XIIIa/farmacologia , Trombocitopenia/sangue , Contagem de Células Sanguíneas , Relação Dose-Resposta a Droga , Fibrinólise/efeitos dos fármacos , Humanos , Proteínas Recombinantes/farmacologia , Transplante de Células-Tronco , Tromboelastografia/métodos , Trombocitopenia/terapia
14.
Thromb Haemost ; 99(4): 720-8, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18392330

RESUMO

The haematopoietic hormone erythropoietin (EPO) has neuroprotective properties and is currently being explored for treatment of stroke and other neurological disorders. Short-term, high-dose treatment with EPO seems to improve neurological function of stroke patients but may be associated with increased thrombotic risk, whereas alternative non-erythropoietic neuroprotective derivatives of EPO, such as carbamylated EPO (CEPO), may be devoid of such side-effects. We investigated the effects of short-term, high-dose treatment with EPO and CEPO on platelet function and haemostasis in healthy mice and rats. Animals received three daily doses of EPO or CEPO (50 microg/kg), and blood was compared with respect to alterations in haematology and platelet reactivity. In rats, treatment with EPO increased the haematocrit to >50% and the mean platelet volume by 37%, while CEPO had no effect on these parameters. Platelets from EPO-treated rats showed an increased sensitivity to thrombin receptor agonist peptides and elevated plasma levels of soluble P-selectin (sP-selectin) were found in treated mice. Further indicators of platelet hyperreactivity in EPO, but not CEPO-treated animals, were significantly increased aggregatory responses to collagen in whole blood and platelet-rich plasma (PRP). The increased platelet reactivity was paralleled by a decreased bleeding time after tail transection in rats. Samples from EPO-treated rats showed an attenuated response to ADP in whole blood aggregometry and thrombelastography (TEG) platelet mapping but not in apyrase-treated PRP, suggesting involvement of ADP receptor desensitization. These findings suggest that while EPO affects various aspects of platelet function, CEPO is devoid of such effects.


Assuntos
Eritropoetina/análogos & derivados , Eritropoetina/farmacologia , Fármacos Neuroprotetores/farmacologia , Ativação Plaquetária/efeitos dos fármacos , Difosfato de Adenosina/farmacologia , Animais , Tempo de Sangramento , Proteínas Sanguíneas/metabolismo , Colágeno/farmacologia , Relação Dose-Resposta a Droga , Eritropoetina/administração & dosagem , Masculino , Camundongos , Fármacos Neuroprotetores/administração & dosagem , Selectina-P/sangue , Agregação Plaquetária/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptores Ativados por Proteinase/agonistas , Proteínas Recombinantes
15.
Transfusion ; 47(11): 2057-65, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17958535

RESUMO

BACKGROUND: The increasing demand for platelet (PLT) transfusions has focused attention on appropriate use. Coated PLTs are a subpopulation of highly procoagulant PLTs formed by simultaneous stimulation by the agonist's collagen and thrombin hypothesized to drive clot formation at the site of vascular injury. Prolonged storage of PLTs may reduce their ability to support optimal hemostasis upon transfusion. STUDY DESIGN AND METHODS: PLT concentrates (PCs) stored for 1, 4, 6, and 8 days were costimulated with thrombin and the collagen glycoprotein VI (GPVI) receptor agonist convulxin, and their ability to form coated PLTs was determined by flow cytometry. Further, a plasma-based thrombin generation assay and thrombelastography were used to evaluate the aged PCs' capacity to support thrombin generation and clot formation, respectively. The stored PCs were additionally tested by standard quality control methods. RESULTS: PLT quality as measured by standard analyses was acceptable according to current practice. The hemostatic potential, however, was impaired with increasing storage time. The formation of coated PLTs decreased significantly from approximately 85 to 55 percent with increasing storage time (p<0.05). The velocity of clot formation was significantly increased from Day 4 (p<0.05). The velocity of thrombin generation and resistance against fibrinolysis were significantly reduced on Day 8 compared to Day 1 of storage (p<0.05). CONCLUSION: Data in the present study suggest that storage significantly reduced the stored PLTs' ability to respond to conditions expected to exist at the site of vascular injury and that storage-induced reduction in PLT activation sensitivity correlated with a loss of hemostatic potential.


Assuntos
Plaquetas/fisiologia , Preservação de Sangue , Hemostasia , Plaquetas/citologia , Citometria de Fluxo , Humanos , Ativação Plaquetária , Tromboelastografia , Trombina/biossíntese , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA