Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
PLoS One ; 16(10): e0258192, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34614035

RESUMO

OBJECTIVES: Acquired coagulopathy may be associated with bleeding risk. Approaches to restore haemostasis include administration of coagulation factor concentrates, but there are concerns regarding potential prothrombotic risk. The present study assessed the prothrombotic potential of four-factor prothrombin complex concentrate (4F-PCC) versus activated PCC (aPCC) and recombinant factor VIIa (rFVIIa), using three preclinical animal models. METHODS: The first model was a modified Wessler model of venous stasis-induced thrombosis in rabbit, focusing on dilutional coagulopathy; the second model employed the same system but focused on direct oral anticoagulant reversal (i.e. edoxaban). The third model assessed the prothrombotic impact of 4F-PCC, aPCC and rFVIIa in a rat model of ferric chloride-induced arterial thrombosis. RESULTS: In the first model, thrombi were observed at aPCC doses ≥10 IU/kg (therapeutic dose 100 IU/kg) and rFVIIa doses ≥50 µg/kg (therapeutic dose 90 µg/kg), but not 4F-PCC 50 IU/kg (therapeutic dose 50 IU/kg). The impact of 4F-PCC (up to 300 IU/kg) on thrombus formation was evident from 10 minutes post-administration, but not at 24 hours post-administration; this did not change with addition of tranexamic acid and/or fibrinogen concentrate. 4F-PCC-induced thrombus formation was lower after haemodilution versus non-haemodilution. In the second model, no prothrombotic effect was confirmed at 4F-PCC 50 IU/kg. The third model showed lower incidence of thrombus formation for 4F-PCC 50 IU/kg versus aPCC (50 U/kg) and rFVIIa (90 µg/kg). CONCLUSIONS: These results suggest that 4F-PCC has a low thrombotic potential versus aPCC or rFVIIa, supporting the clinical use of 4F-PCC for the treatment of coagulopathy-mediated bleeding.


Assuntos
Fatores de Coagulação Sanguínea/metabolismo , Trombose/metabolismo , Animais , Artérias/efeitos dos fármacos , Artérias/patologia , Fator VIIa/farmacologia , Feminino , Hemodiluição , Coelhos , Ratos Sprague-Dawley , Proteínas Recombinantes/farmacologia , Trombose/patologia , Fatores de Tempo , Ácido Tranexâmico/farmacologia
2.
J Thromb Haemost ; 18(12): 3194-3202, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32810892

RESUMO

BACKGROUND: Hemophilia B is caused by coagulation factor IX (FIX) deficiency. Recombinant fusion protein linking coagulation FIX with recombinant albumin (rIX-FP; Idelvion® ) is used for replacement therapy with an extended half-life. A previous quantitative whole-body autoradiography (QWBA) study investigating the biodistribution of rIX-FP indicated equal biodistribution, but more prolonged tissue retention compared with a marketed recombinant FIX product. OBJECTIVES: To complete and confirm the QWBA study data by directly measuring rIX-FP protein and activity levels in tissues following intravenous (i.v.) administration to normal rats and FIX-deficient (hemophilia B) mice. METHODS: After i.v. administration of rIX-FP at a dose of 2000 IU/kg, animals were euthanized at specific time points up to 72 hours postdosing. Subsequently, plasma and various tissues, which were selected based on the previous QWBA results, were harvested and analyzed for FIX antigen levels using an ELISA (both species) or an immunohistochemistry method (mice only), as well as for FIX activity levels (mice only) using a chromogenic assay. RESULTS: In rats, rIX-FP distributed extravascularly into all tissues analyzed (ie, liver, kidney, skin and knee) with peak antigen levels reached between 1 and 7 hours postdosing. In hemophilia B mice, rIX-FP tissue distribution was comparable to rats. FIX antigen levels correlated well with FIX activity readouts. CONCLUSIONS: Our results confirm QWBA data showing that rIX-FP distributes into relevant target tissues. Importantly, it was demonstrated that rIX-FP available in tissues retains its functional activity and can thus facilitate its therapeutic activity at sites of potential injury.


Assuntos
Hemofilia B , Roedores , Administração Intravenosa , Animais , Fator IX/metabolismo , Meia-Vida , Hemofilia B/tratamento farmacológico , Camundongos , Ratos , Proteínas Recombinantes de Fusão/uso terapêutico , Roedores/metabolismo , Distribuição Tecidual
3.
J Pharmacol Toxicol Methods ; 105: 106901, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32771565

RESUMO

INTRODUCTION: The implementation of novel, reliable biomarkers for the early and differential diagnosis of acute kidney injury (AKI) could greatly improve the timely treatment and prevention of disease progression, particularly since the current gold standards for detecting kidney injury such as serum creatinine (SCr) and blood urea nitrogen (BUN) lack sensitivity and specificity. We evaluated novel urinary kidney injury biomarkers focusing on early detection and better prediction of AKI with higher sensitivity and specificity. METHODS: In the rat, urinary biomarkers for kidney injury, i.e. albumin, beta-2-microglobulin (B2M), clusterin, cystatin C, kidney injury molecule-1 (KIM-1), neutrophil gelatinase-associated lipocalin (NGAL), osteopontin (OPN), and total protein (TP), were investigated in an AKI model using different hyperosmolar and high-dose solutions, i.e. mannitol, sucrose, and contrast medium (CM), as acute single insults leading to kidney injury. Additionally, dose-dependency of sucrose was investigated and effects were compared to the sucrose- and iron-containing marketed drug Venofer®. RESULTS: Levels of excreted urinary biomarkers correlated with severity of AKI, exhibited a dose-dependent response to sucrose treatment, and demonstrated evidence of recovery from kidney injury with transient and reversible changes. The exceptions were KIM-1 and NGAL, which showed later responses following CM and iron-induced renal injury. All biomarkers outperformed plasma creatinine (PCr), BUN, and histopathology, with regard to practicability and/or detection of proximal tubular injury. DISCUSSION: The use of a panel of urinary kidney injury biomarkers emerged as an early, sensitive, and predictive tool to detect AKI showing enhanced sensitivity compared to current state-of-the-art markers.


Assuntos
Injúria Renal Aguda/sangue , Injúria Renal Aguda/metabolismo , Biomarcadores/sangue , Biomarcadores/metabolismo , Rim/metabolismo , Animais , Nitrogênio da Ureia Sanguínea , Moléculas de Adesão Celular/metabolismo , Creatinina/sangue , Modelos Animais de Doenças , Diagnóstico Precoce , Testes de Função Renal/métodos , Lipocalina-2/metabolismo , Masculino , Ratos , Ratos Sprague-Dawley , Sensibilidade e Especificidade
4.
Int J Med Microbiol ; 306(7): 572-579, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27338836

RESUMO

Coagulation is a mechanism for wound healing after injury. Several recent studies delineate an additional role of the intrinsic pathway of coagulation, also known as the contact system, in the early innate immune response against bacterial infections. In this study, we investigated the role of factor XIII (FXIII), which is activated upon coagulation induction, during Streptococcus pyogenes-mediated skin and soft tissue infections. FXIII has previously been shown to be responsible for the immobilization of bacteria within a fibrin network which may prevent systemic bacterial dissemination. In order to investigate if the FXIII-mediated entrapment of S. pyogenes also influences the disease outcome we used a murine S. pyogenes M1 skin and soft tissue infection model. Here, we demonstrate that a lack of FXIII leads to prolonged clotting times, increased signs of inflammation, and elevated bacterial dissemination. Moreover, FXIII-deficient mice show an impaired survival when compared with wildtype animals. Additionally, local reconstitution of FXIII-deficient mice with a human FXIII-concentrate (Fibrogammin®P) could reduce the systemic complications, suggesting a protective role for FXIII during early S. pyogenes skin infection. FXIII therefore might be a possible therapeutically application to support the early innate immune response during skin infections caused by S. pyogenes.


Assuntos
Fator XIII/metabolismo , Interações Hospedeiro-Patógeno , Imunidade Inata , Dermatopatias Bacterianas/imunologia , Infecções Estreptocócicas/imunologia , Streptococcus pyogenes/imunologia , Animais , Antígenos de Bactérias , Proteínas da Membrana Bacteriana Externa , Proteínas de Transporte , Modelos Animais de Doenças , Fator XIII/genética , Feminino , Camundongos Endogâmicos CBA , Camundongos Knockout , Dermatopatias Bacterianas/microbiologia , Dermatopatias Bacterianas/patologia , Infecções Estreptocócicas/microbiologia , Infecções Estreptocócicas/patologia , Streptococcus pyogenes/patogenicidade , Análise de Sobrevida
5.
Pharmacol Res Perspect ; 4(2): e00227, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27069637

RESUMO

Factor XIII (FXIII) is a coagulation protein which plays a major role in hemostasis by covalently cross-linking fibrin molecules, thereby stabilizing the blood clot and increasing resistance to fibrinolysis. FXIII deficiency, either congenital or acquired, is associated with spontaneous bleeding, increased bleeding time, and poor wound healing. Purified plasma-derived human FXIII concentrate (pd hFXIII) has been available since 1993 for therapeutic use in congenital FXIII deficiency. This set of nonclinical investigations aimed to evaluate the pharmacodynamic effects and assess the safety profile of pd hFXIII. The efficacy and safety of pd hFXIII were evaluated by pharmacodynamic, pharmacokinetic, and toxicity studies in mice and rats, safety pharmacology studies in dogs, neoantigenicity study, local tolerance, and thrombogenicity tests in rabbits. Administration of pd hFXIII resulted in the correction of deficits in clot formation kinetics and strength as measured by thromboelastometry, and was not associated with thrombus formation up to 350 IU/kg in FXIII knockout mice. There was no production of neoantigens resulting from the viral elimination manufacturing steps detected, and no adverse reactions were observed in toxicity studies with single doses up to 3550 IU/kg in mice and 1420 IU/kg in rats; nor from repeat doses of 350 IU/kg in rats. In addition, local tolerance tests revealed a good tolerability profile in rabbits. Overall, this data showed that pd hFXIII was well tolerated and pharmacodynamically active in preclinical animal models, supporting pd hFXIII as a therapy for FXIII deficiency.

6.
Br J Haematol ; 173(5): 769-78, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27018425

RESUMO

Haemostasis including blood coagulation is initiated upon vessel wall injury and indispensable to limit excessive blood loss. However, unregulated pathological coagulation may lead to vessel occlusion, causing thrombotic disorders, most notably myocardial infarction and stroke. Furthermore, blood exposure to foreign surfaces activates the intrinsic pathway of coagulation. Hence, various clinical scenarios, such as extracorporeal membrane oxygenation, require robust anticoagulation consequently leading to an increased bleeding risk. This study aimed to further assess the antithrombotic efficacy of the activated factor XII (FXIIa) inhibitor, rHA-Infestin-4, in several thrombosis models. In mice, rHA-Infestin-4 decreased occlusion rates in the mechanically-induced arterial (Folt's) and the FeCl3 -induced venous thrombosis model. rHA-Infestin-4 also protected from FeCl3 -induced arterial thrombosis and from stasis-prompted venous thrombosis in rabbits. Furthermore, rHA-Infestin-4 prevented occlusion in the arterio-venous shunt model in mice and rabbits where thrombosis was induced via a foreign surface. In contrast to heparin, the haemostatic capacity in rabbits was unaffected by rHA-Infestin-4. Using rodent and non-rodent species, our data demonstrate that the FXIIa inhibitor rHA-Infestin-4 decreased arterial, venous and foreign surface-induced thrombosis without affecting physiological haemostasis. Hence, we provide further evidence that targeting FXIIa represents a potent yet safe antithrombotic treatment approach, especially in foreign surface-triggered thrombosis.


Assuntos
Fator XIIa/antagonistas & inibidores , Proteínas de Insetos/farmacologia , Proteínas Recombinantes de Fusão/farmacologia , Albumina Sérica/farmacologia , Trombose/tratamento farmacológico , Animais , Arteriopatias Oclusivas/tratamento farmacológico , Arteriopatias Oclusivas/etiologia , Modelos Animais de Doenças , Fibrinolíticos/farmacologia , Hemostasia/efeitos dos fármacos , Proteínas de Insetos/uso terapêutico , Cinética , Camundongos , Coelhos , Proteínas Recombinantes de Fusão/uso terapêutico , Albumina Sérica/uso terapêutico , Albumina Sérica Humana , Trombose/etiologia , Resultado do Tratamento , Trombose Venosa/tratamento farmacológico , Trombose Venosa/etiologia
7.
PLoS One ; 11(1): e0146783, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26815580

RESUMO

BACKGROUND AND PURPOSE: Ischemic stroke provokes severe brain damage and remains a predominant disease in industrialized countries. The coagulation factor XII (FXII)-driven contact activation system plays a central, but not yet fully defined pathogenic role in stroke development. Here, we investigated the efficacy of the FXIIa inhibitor rHA-Infestin-4 in a rat model of ischemic stroke using both a prophylactic and a therapeutic approach. METHODS: For prophylactic treatment, animals were treated intravenously with 100 mg/kg rHA-Infestin-4 or an equal volume of saline 15 min prior to transient middle cerebral artery occlusion (tMCAO) of 90 min. For therapeutic treatment, 100 mg/kg rHA-Infestin-4, or an equal volume of saline, was administered directly after the start of reperfusion. At 24 h after tMCAO, rats were tested for neurological deficits and blood was drawn for coagulation assays. Finally, brains were removed and analyzed for infarct area and edema formation. RESULTS: Within prophylactic rHA-Infestin-4 treatment, infarct areas and brain edema formation were reduced accompanied by better neurological scores and survival compared to controls. Following therapeutic treatment, neurological outcome and survival were still improved although overall effects were less pronounced compared to prophylaxis. CONCLUSIONS: With regard to the central role of the FXII-driven contact activation system in ischemic stroke, inhibition of FXIIa may represent a new and promising treatment approach to prevent cerebral ischemia/reperfusion injury.


Assuntos
Fator XIIa/antagonistas & inibidores , Infarto da Artéria Cerebral Média/tratamento farmacológico , Proteínas de Insetos/farmacologia , Proteínas Recombinantes de Fusão/farmacologia , Traumatismo por Reperfusão/prevenção & controle , Inibidores de Serina Proteinase/farmacologia , Albumina Sérica/farmacologia , Animais , Encéfalo/irrigação sanguínea , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Células CHO , Cricetulus , Avaliação Pré-Clínica de Medicamentos , Fator XIIa/metabolismo , Proteínas de Insetos/uso terapêutico , Masculino , Ratos , Proteínas Recombinantes de Fusão/uso terapêutico , Teste de Desempenho do Rota-Rod , Inibidores de Serina Proteinase/uso terapêutico , Albumina Sérica/uso terapêutico , Albumina Sérica Humana
8.
Blood Coagul Fibrinolysis ; 26(5): 515-21, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25767894

RESUMO

Treatment of von Willebrand disease typically requires multiple infusions of von Willebrand factor (VWF)/factor VIII (FVIII) concentrate. Accumulation of FVIII is a clinical concern due to potential risk for thromboembolism. This study sought to determine whether VWF/FVIII concentrate of high VWF:FVIII ratio can prevent FVIII accumulation. VWF-deficient knockout mice received four 150 IU/kg VWF:ristocetin cofactor (RCo) infusions at 3-h intervals, with VWF/FVIII concentrates of a high (Haemate P/Humate-P) or low (Wilate) VWF:FVIII ratio. After each infusion, trough FVIII and VWF levels in plasma were determined. Separately, pharmacokinetic analysis was performed after single 250-IU/kg VWF:RCo infusions of each concentrate. Over the course of the four infusions, trough FVIII increased significantly in the group receiving Wilate (P < 0.001), but not Haemate P/Humate P (P = 0.058). After the first infusion, mean trough FVIII level in the Wilate group (31.7 IU/dl) was greater by 82% (P = 0.017) than that in the Haemate P/Humate P group (17.4 IU/dl). After the final infusion, mean trough FVIII of animals receiving Wilate (55.1 IU/dl) continued to exceed that of Haemate P/Humate P recipients (30.2 IU/dl) significantly (P < 0.001). Trough VWF levels were similar in the two groups. The VWF pharmacokinetics of the two concentrates coincided closely; however, the FVIII peak concentration and area under the curve were approximately twice as great in the mice treated with Wilate. In a murine model of severe von Willebrand disease, a VWF/FVIII concentrate with a high VWF:FVIII ratio prevented persistent exposure to elevated trough FVIII levels.


Assuntos
Fator VIII/uso terapêutico , Animais , Combinação de Medicamentos , Feminino , Humanos , Masculino , Camundongos , Fator de von Willebrand
9.
Thromb Haemost ; 113(4): 686-97, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25652913

RESUMO

Coagulation factor XIII (FXIII), a plasma transglutaminase, is best known as the final enzyme in the coagulation cascade, where it is responsible for cross-linking of fibrin. However, a growing body of evidence has demonstrated that FXIII targets a wide range of additional substrates that have important roles in health and disease. These include antifibrinolytic proteins, with cross-linking of α2-antiplasmin to fibrin, and potentially fibrinogen, being the principal mechanism(s) whereby plasmin-mediated clot degradation is minimised. FXIII also acts on endothelial cell VEGFR-2 and αvß3 integrin, which ultimately leads to downregulation of the antiangiogenic protein thrombospondin-1, promoting angiogenesis and neovascularisation. Under infectious disease conditions, FXIII cross-links bacterial surface proteins to fibrinogen, resulting in immobilisation and killing, while during wound healing, FXIII induces cross-linking of the provisional matrix. The latter process has been shown to influence the interaction of leukocytes with the provisional extracellular matrix and promote wound healing. Through these actions, there are good rationales for evaluating the therapeutic potential of FXIII in diseases in which tissue repair is dysregulated or perturbed, including systemic sclerosis (scleroderma), invasive bacterial infections, and tissue repair, for instance healing of venous leg ulcers or myocardial injuries. Adequate levels of FXIII are also required in patients undergoing surgery to prevent or treat perioperative bleeding, and its augmentation in patients with/at risk for perioperative bleeding may also have potential clinical benefit. While there are preclinical and/or clinical data to support the use of FXIII in a range of settings, further clinical evaluation in these underexplored applications is warranted.


Assuntos
Indutores da Angiogênese/uso terapêutico , Coagulação Sanguínea/efeitos dos fármacos , Coagulantes/uso terapêutico , Fator XIII/uso terapêutico , Neovascularização Fisiológica/efeitos dos fármacos , Cicatrização/efeitos dos fármacos , Animais , Infecções Bacterianas/sangue , Infecções Bacterianas/tratamento farmacológico , Infecções Bacterianas/fisiopatologia , Perda Sanguínea Cirúrgica/prevenção & controle , Fator XIII/metabolismo , Deficiência do Fator XIII/sangue , Deficiência do Fator XIII/tratamento farmacológico , Deficiência do Fator XIII/fisiopatologia , Fator XIIIa/metabolismo , Fibrina/metabolismo , Humanos , Hemorragia Pós-Operatória/sangue , Hemorragia Pós-Operatória/etiologia , Hemorragia Pós-Operatória/prevenção & controle , Escleroderma Sistêmico/sangue , Escleroderma Sistêmico/tratamento farmacológico , Escleroderma Sistêmico/fisiopatologia , Transdução de Sinais , Especificidade por Substrato , Trombospondina 1/sangue
10.
Thromb Res ; 135(3): 554-60, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25619440

RESUMO

INTRODUCTION: Rivaroxaban is an oral, selective direct factor Xa inhibitor approved for several indications in patients at risk of thrombotic events. One limitation of its clinical use is the lack of data pertaining to its reversal in situations where urgent response is critical (e.g. acute bleeding events or emergency surgery). MATERIALS AND METHODS: This study assessed the effectiveness of a four-factor prothrombin complex concentrate (4F-PCC; Beriplex(®)/Kcentra(®)) for the reversal of rivaroxaban-associated bleeding in an in vivo rabbit model, and evaluated the correlations between in vitro coagulation parameters and haemostasis in vivo. RESULTS: Administration of single intravenous doses of rivaroxaban (150-450 µg/kg) resulted in increased and prolonged bleeding following standardised kidney incision. Pre-incision treatment with 4F-PCC (25-100 IU/kg) resulted in a dose-dependent reversal of rivaroxaban (150 and 300 µg/kg)-associated increases in time to haemostasis and blood loss; no reversal was seen at the highest rivaroxaban dose (450 µg/kg). Of the in vitro biomarkers tested, thrombin generation and whole-blood clotting time correlated well with in vivo measures of 4F-PCC-mediated effects. Thrombin generation was highly reagent-dependent, with the assay initiated using the phospholipid-only reagent being the most predictive of effective haemostasis in vivo. CONCLUSIONS: In summary, in a rabbit model of acute bleeding, treatment with 4F-PCC reduced bleeding to control levels following rivaroxaban 150 µg/kg and 300 µg/kg administration.


Assuntos
Fatores de Coagulação Sanguínea/uso terapêutico , Coagulação Sanguínea/efeitos dos fármacos , Inibidores do Fator Xa/efeitos adversos , Hemorragia/induzido quimicamente , Hemorragia/tratamento farmacológico , Rivaroxabana/efeitos adversos , Doença Aguda , Animais , Modelos Animais de Doenças , Inibidores do Fator Xa/sangue , Hemorragia/metabolismo , Humanos , Coelhos , Rivaroxabana/sangue , Trombina/metabolismo
11.
Anesthesiology ; 122(2): 387-98, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25419685

RESUMO

BACKGROUND: Edoxaban is an oral, selective direct factor Xa inhibitor approved in Japan for venous thromboembolism prevention after orthopedic surgery. Data are lacking regarding reversal strategies for edoxaban; this study assessed whether four-factor prothrombin complex concentrate (Beriplex/Kcentra; CSL Behring GmbH, Marburg, Germany) can effectively reverse its effects on hemostasis using a previously described rabbit model. METHODS: The study comprised assessments of thrombin generation in vitro, pharmacokinetic parameters, and edoxaban reversal in vivo. In a blinded in vivo stage, a standardized kidney incision was performed in animals (n = 11 per group) randomized to receive vehicle + saline, edoxaban (1,200 µg/kg) + saline, or edoxaban (1,200 µg/kg) + four-factor prothrombin complex concentrate (50 IU/kg). Animals were monitored for treatment impact on hemostasis and coagulation parameters. Data are median (range). Statistical tests were adjusted for multiple testing. RESULTS: Edoxaban administration increased blood loss (30 [2 to 44] ml) and time to hemostasis (23 [8.5 to 30.0] min) compared with the control group (3 [1 to 8] ml and 3 [2.0 to 5.0] min, respectively). Biomarkers of coagulation (prothrombin time, activated partial thromboplastin time, whole blood clotting time) and thrombin generation parameters (e.g., peak thrombin, endogenous thrombin potential, lag time) were also affected by edoxaban. Administration of four-factor prothrombin complex concentrate significantly reduced time to hemostasis (to 8 [6.5 to 14.0] min, observed P < 0.0001) and total blood loss (to 9 [4 to 22] ml, observed P = 0.0050) compared with the edoxaban + saline group. Of the biomarkers tested, prothrombin time, whole blood clotting time, and endogenous thrombin potential correlated best with clinical parameters. CONCLUSION: In a rabbit model of hemostasis, four-factor prothrombin complex concentrate administration significantly decreased edoxaban-associated hemorrhage.


Assuntos
Fatores de Coagulação Sanguínea/uso terapêutico , Hemorragia/induzido quimicamente , Hemorragia/prevenção & controle , Hemostáticos/uso terapêutico , Piridinas/antagonistas & inibidores , Piridinas/toxicidade , Tiazóis/antagonistas & inibidores , Tiazóis/toxicidade , Doença Aguda , Animais , Fatores de Coagulação Sanguínea/farmacocinética , Testes de Coagulação Sanguínea , Chinchila , Determinação de Ponto Final , Feminino , Transtornos Hemorrágicos/prevenção & controle , Hemostáticos/farmacocinética , Técnicas In Vitro , Piridinas/farmacocinética , Coelhos , Tiazóis/farmacocinética , Trombina/metabolismo
12.
Thromb Res ; 134(3): 729-36, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25084749

RESUMO

INTRODUCTION: In vivo animal data have shown prothrombin complex concentrate (PCC) to be effective in preventing bleeding induced by excessive plasma levels of the direct thrombin inhibitor dabigatran. This animal model study was designed to determine the risk of thrombosis associated with administration of a PCC (Beriplex P/N) to reverse dabigatran-induced bleeding. MATERIALS AND METHODS: Anesthetized rabbits were treated with initial 0, 75, 200 or 450 µg kg(-1) dabigatran boluses followed by continuous infusions to maintain elevated plasma dabigatran levels. At 15 min after the start of dabigatran administration, PCC doses of 0, 50 or 300 IU kg(-1) were administered. Thereafter, coagulation in an arteriovenous (AV) shunt was evaluated and histopathologic examination for thrombotic changes performed. Venous thrombosis was also assessed in a modified Wessler model. RESULTS: At the suprapharmacologic dose of 300 IU kg(-1), PCC increased thrombus weight during AV shunting, but this effect could be prevented by dabigatran at all tested doses. AV shunt occlusion after PCC administration was delayed by 75 µg kg(-1) dabigatran and abolished by progressively higher dabigatran doses. High-dose treatment with 300 IU kg(-1) PCC resulted in histologically evident low-grade pulmonary thrombi; however, that effect could be blocked by dabigatran in a dose-dependent manner (p=0.034). In rabbits treated with high-dose PCC, dabigatran inhibited thrombus formation during venous stasis. PCC effectively reversed dabigatran-induced bleeding. CONCLUSIONS: In this animal study, thrombosis after PCC administration could be prevented in the presence of dabigatran. PCC reversed dabigatran-induced excessive bleeding while retaining protective anticoagulatory activity of dabigatran.


Assuntos
Coagulação Sanguínea/efeitos dos fármacos , Dabigatrana , Fator IX/toxicidade , Fator VII/toxicidade , Fator X/toxicidade , Hemorragia/prevenção & controle , Hemostáticos/toxicidade , Protrombina/toxicidade , Trombose Venosa/induzido quimicamente , Animais , Testes de Coagulação Sanguínea , Modelos Animais de Doenças , Combinação de Medicamentos , Fator IX/administração & dosagem , Fator VII/administração & dosagem , Fator X/administração & dosagem , Feminino , Hemorragia/sangue , Hemorragia/induzido quimicamente , Hemostáticos/administração & dosagem , Protrombina/administração & dosagem , Coelhos , Medição de Risco , Fatores de Risco , Trombina/metabolismo , Fatores de Tempo , Trombose Venosa/sangue , Trombose Venosa/prevenção & controle
13.
Toxicol Appl Pharmacol ; 280(1): 70-7, 2014 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-25102310

RESUMO

Fibrinogen, a soluble 340kDa plasma glycoprotein, is critical in achieving and maintaining hemostasis. Reduced fibrinogen levels are associated with an increased risk of bleeding and recent research has investigated the efficacy of fibrinogen concentrate for controlling perioperative bleeding. European guidelines on the management of perioperative bleeding recommend the use of fibrinogen concentrate if significant bleeding is accompanied by plasma fibrinogen levels less than 1.5-2.0g/l. Plasma-derived human fibrinogen concentrate has been available for therapeutic use since 1956. The overall aim of the comprehensive series of non-clinical investigations presented was to evaluate i) the pharmacodynamic and pharmacokinetic characteristics and ii) the safety and tolerability profile of human fibrinogen concentrate Haemocomplettan P® (RiaSTAP®). Pharmacodynamic characteristics were assessed in rabbits, pharmacokinetic parameters were determined in rabbits and rats and a safety pharmacology study was performed in beagle dogs. Additional toxicology tests included: single-dose toxicity tests in mice and rats; local tolerance tests in rabbits; and neoantigenicity tests in rabbits and guinea pigs following the introduction of pasteurization in the manufacturing process. Human fibrinogen concentrate was shown to be pharmacodynamically active in rabbits and dogs and well tolerated, with no adverse events and no influence on circulation, respiration or hematological parameters in rabbits, mice, rats and dogs. In these non-clinical investigations, human fibrinogen concentrate showed a good safety profile. This data adds to the safety information available to date, strengthening the current body of knowledge regarding this hemostatic agent.


Assuntos
Fibrinogênio/metabolismo , Fibrinogênio/toxicidade , Animais , Peso Corporal/efeitos dos fármacos , Peso Corporal/fisiologia , Cães , Relação Dose-Resposta a Droga , Feminino , Fibrinogênio/farmacologia , Cobaias , Hemostasia/efeitos dos fármacos , Hemostasia/fisiologia , Humanos , Masculino , Camundongos , Coelhos , Ratos , Ratos Wistar , Especificidade da Espécie
14.
Thromb Haemost ; 112(5): 960-71, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25103795

RESUMO

Human plasma-derived C1-esterase inhibitor (C1-INH) is an efficacious and safe treatment for hereditary angioedema. However, thrombotic events in subjects treated with C1-INH at recommended or off-label, high doses have been reported. In this study, we addressed the potential prothrombotic risk of C1-INH treatment in high doses using a non-clinical rabbit model. Following intravenous infusion of C1-INH to rabbits at doses up to 800 IU/kg, the exposure and the pharmacodynamic efficacy of C1-INH in rabbits were confirmed by activity measurements of C1-esterase, and coagulation factors XIa and XIIa, respectively. Potential prothrombotic effects were assessed following induction of venous and arterial thrombosis using in vivo models of venous and arterial stasis, complemented by various in vitro assays of coagulation markers. Administration of C1-INH at doses up to 800 IU/kg did not potentiate thrombus formation during venous stasis. In contrast, inhibition of arterial occlusion was observed upon C1-INH administration when compared with isotonic saline treatment, indicating antithrombotic rather than prothrombotic activity of high dose C1-INH treatment in vivo. This was further confirmed in vitro by decreased thrombin generation, increased activated partial thromboplastin time, clotting time and clot formation time, and inhibition of platelet aggregation. No relevant changes in fibrinolysis or in the levels of thrombin-antithrombin complexes, and prothrombin fragment 1+2 were observed upon high dose C1-INH treatment. The data suggest that treatment of healthy rabbits with high doses of C1-INH could potentially inhibit coagulation and thrombus formation rather than induce a prothrombotic risk.


Assuntos
Arteriopatias Oclusivas/induzido quimicamente , Proteína Inibidora do Complemento C1/toxicidade , Trombose Venosa/induzido quimicamente , Animais , Testes de Coagulação Sanguínea , Proteína Inibidora do Complemento C1/administração & dosagem , Proteína Inibidora do Complemento C1/farmacocinética , Proteína Inibidora do Complemento C1/uso terapêutico , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Fator XIIa/análise , Fator XIa/análise , Artéria Femoral , Fibrinólise/efeitos dos fármacos , Humanos , Infusões Intravenosas , Sistema Calicreína-Cinina/efeitos dos fármacos , Sistema Calicreína-Cinina/fisiologia , Agregação Plaquetária/efeitos dos fármacos , Coelhos , Tromboelastografia , Trombina/biossíntese
15.
Clin Lab Med ; 34(3): 623-35, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25168947

RESUMO

Although new oral anticoagulants (NOACs) represent an advance in anticoagulant therapy over vitamin K antagonists (VKAs), they nevertheless have a low, but significant risk for bleeding complications. Reversal agents for VKAs, such as prothrombin complex concentrates (PCCs), are currently being evaluated in preclinical studies for NOAC reversal. This article reviews the preclinical data for the most extensively studied PCC for NOAC reversal, Beriplex, a 4-factor PCC. The results from the Beriplex studies are also compared with those obtained with other reversal agents, including different nonactivated PCCs, activated PCCs, and recombinant activated factor VII.


Assuntos
Anticoagulantes/efeitos adversos , Fator IX/uso terapêutico , Fator VII/uso terapêutico , Fator X/uso terapêutico , Hemorragia/tratamento farmacológico , Hemostáticos/uso terapêutico , Modelos Biológicos , Protrombina/uso terapêutico , Administração Oral , Animais , Anticoagulantes/administração & dosagem , Anticoagulantes/química , Anticoagulantes/farmacologia , Antitrombinas/administração & dosagem , Antitrombinas/efeitos adversos , Antitrombinas/química , Antitrombinas/farmacologia , Coagulação Sanguínea/efeitos dos fármacos , Relação Dose-Resposta a Droga , Combinação de Medicamentos , Avaliação Pré-Clínica de Medicamentos , Fator IX/administração & dosagem , Fator IX/farmacologia , Fator VII/administração & dosagem , Fator VII/farmacologia , Fator X/administração & dosagem , Fator X/farmacologia , Inibidores do Fator Xa/administração & dosagem , Inibidores do Fator Xa/efeitos adversos , Inibidores do Fator Xa/química , Inibidores do Fator Xa/farmacologia , Hemorragia/induzido quimicamente , Hemostasia/efeitos dos fármacos , Hemostáticos/administração & dosagem , Hemostáticos/farmacologia , Humanos , Protrombina/administração & dosagem , Protrombina/farmacologia
16.
Thromb Res ; 134(2): 495-502, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24958222

RESUMO

INTRODUCTION: A novel fusion protein linking coagulation factor VIIa with albumin (rVIIa-FP) is currently undergoing clinical investigations. OBJECTIVE: This study was conducted to examine the biodistribution of rVIIa-FP in comparison to recombinant factor VIIa (rFVIIa). MATERIALS AND METHODS: [(3)H]-rVIIa-FP (10mgkg(-1)) or [(3)H]-rFVIIa (1.6mgkg(-1)) were administered intravenously to rats, followed by quantitative whole-body and knee joint autoradiography for 24 ([(3)H]-rFVIIa) or 240 ([(3)H]-rVIIa-FP) hours post-dose. Pharmacokinetic and excretion balance analyses were performed. RESULTS: In contrast to [(3)H]-albumin, the tissue distributions of [(3)H]-rVIIa-FP and [(3)H]-rFVIIa were similar. Within the knee, both were rapidly present within synovial and mineralized regions. Importantly, rVIIa-FP- and albumin-derived radioactivity were detectable up to 72-120hours, whereas [(3)H]-rFVIIa signals were already close to detection limits at 24hours. The longest rVIIa-FP retention times were observed in bone marrow and endosteum, in which the retention times were up to 5 times longer for rVIIa-FP compared with rFVIIa. Up to 8hours post-dose, 100% of radioactivity was assigned to unchanged [(3)H]-rVIIa-FP. Elimination of both proteins occurred primarily via the urine. CONCLUSIONS: The data suggest that the FVIIa moiety is directing rVIIa-FP's tissue distribution while the albumin moiety is responsible for the prolonged tissue retention. Importantly, rVIIa-FP is highly concentrated and retained over a long period in the growth plate of the knee joint-a vulnerable site in haemophilia patients. Overall, these improved tissue distribution characteristics of rVIIa-FP may enhance compliance and allow a more convenient dosing frequency.


Assuntos
Fator VIIa/farmacocinética , Proteínas Recombinantes de Fusão/farmacocinética , Albumina Sérica/farmacocinética , Animais , Fator VIIa/genética , Masculino , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacocinética , Albumina Sérica/genética , Distribuição Tecidual
17.
Thromb Res ; 134(1): 125-31, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24814969

RESUMO

INTRODUCTION: rVIII-SingleChain (CSL627), a novel recombinant coagulation factor VIII (FVIII), is under investigation in a phase I/III clinical programme (AFFINITY) for the treatment of haemophilia A. Non-clinical studies were conducted to investigate the pharmacokinetic/pharmacodynamic profile of rVIII-SingleChain in comparison with full-length recombinant FVIII. MATERIALS AND METHODS: Binding affinity of rVIII-SingleChain for von Willebrand factor was investigated by surface plasmon resonance analysis. The pharmacokinetic profile of rVIII-SingleChain was compared with a marketed full-length recombinant FVIII concentrate (Advate(®)) in haemophilia A mice, von Willebrand factor knock-out mice, Crl:CD (SD) rats, rabbits and cynomolgus monkeys. Systemic FVIII activity or antigen levels were recorded. Procoagulant activity was measured in an FeCl3-induced arterial occlusion model and by recording thrombin generation activity (ex vivo) after administration of 200-250 IU/kg rVIII-SingleChain or full-length FVIII to haemophilia A mice. RESULTS: rVIII-SingleChain displayed a high affinity for von Willebrand factor (KD=44 pM vs. 139 pM for full-length recombinant FVIII). In all animal species tested, rVIII-SingleChain had more favourable pharmacokinetic properties than full-length recombinant FVIII: clearance was decreased and area under the curve and terminal half-life were enhanced vs. full-length recombinant FVIII, while in vivo recovery and volume of distribution were equivalent. rVIII-SingleChain showed a prolonged thrombin generation potential and prolonged procoagulant activity vs. full-length recombinant FVIII in an FeCl3-induced arterial occlusion model. CONCLUSIONS: rVIII-SingleChain had a higher affinity for von Willebrand factor than full-length recombinant FVIII and displayed favourable pharmacokinetic/pharmacodynamic properties in non-clinical models.


Assuntos
Fator VIII/farmacologia , Fator VIII/farmacocinética , Fator de von Willebrand/metabolismo , Animais , Modelos Animais de Doenças , Hemofilia A/metabolismo , Macaca fascicularis , Camundongos , Coelhos , Ratos , Ressonância de Plasmônio de Superfície
18.
Thromb Res ; 133(5): 900-7, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24680550

RESUMO

INTRODUCTION: The recombinant fusion protein linking coagulation factor IX with albumin (rIX-FP) is undergoing clinical trials for prophylaxis and on-demand treatment of haemophilia B patients. The aim of this study was to investigate the pharmacokinetics, whole-body and knee joint distribution of rIX-FP following intravenous administration to rats, compared with a marketed, non-fused rFIX and recombinant human albumin. MATERIAL AND METHODS: [(3)H]-rIX-FP, [(3)H]-rFIX or [(3)H]-albumin were administered to rats followed by quantitative whole-body autoradiography over 24 or 240 hours, and the tissue distribution as well as elimination of radioactivity were measured. RESULTS: Elimination of all radioactivity derived from the three proteins was shown to occur primarily via the urine. The tissue distribution of [(3)H]-rIX-FP and [(3)H]-rFIX (but not of [(3)H]-albumin) was comparable, both penetrating predominantly into bone, and well-perfused tissues, suggesting that the rIX moiety determines the distribution pattern of rIX-FP, while the albumin moity is responsible for the prolonged plasma and tissue retention. Detailed knee-joint analysis indicated rapid presence of [(3)H]-rIX-FP and [(3)H]-rFIX in synovial and mineralised bone tissue, mostly localised to the zone of calcified cartilage. Longest retention times were observed in the bone marrow and the endosteum of long bones. Intriguingly, [(3)H]-rIX-FP- and [(3)H]-albumin-derived radioactive signals were detectable up to 240 hours, while [(3)H]-rFIX-derived radioactivity rapidly declined after 1hour post-dosing correlating to the extended plasma half-life of [(3)H]-rIX-FP. CONCLUSION: The prolonged plasma and tissue retention of rIX-FP achieved by albumin fusion may allow a reduction in dosing frequency leading to increased therapeutic compliance and convenience.


Assuntos
Albuminas/farmacocinética , Fator IX/farmacocinética , Animais , Humanos , Articulação do Joelho/metabolismo , Masculino , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes de Fusão/farmacocinética , Distribuição Tecidual
19.
Sci Transl Med ; 6(222): 222ra17, 2014 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-24500405

RESUMO

Currently used anticoagulants prevent thrombosis but increase bleeding. We show an anticoagulation therapy without bleeding risk based on a plasma protease factor XII function-neutralizing antibody. We screened for antibodies against activated factor XII (FXIIa) using phage display and demonstrated that recombinant fully human antibody 3F7 binds into the FXIIa enzymatic pocket. 3F7 interfered with FXIIa-mediated coagulation, abolished thrombus formation under flow, and blocked experimental thrombosis in mice and rabbits. We adapted an extracorporeal membrane oxygenation (ECMO) cardiopulmonary bypass system used for infant therapy to analyze clinical applicability of 3F7 in rabbits. 3F7 provided thromboprotection as efficiently as heparin, and both drugs prevented fibrin deposition and thrombosis within the extracorporeal circuit. Unlike heparin, 3F7 treatment did not impair the hemostatic capacity and did not increase bleeding from wounds. These data establish that targeting of FXIIa is a safe mode of thromboprotection in bypass systems, and provide a clinically relevant anticoagulation strategy that is not complicated by excess bleeding.


Assuntos
Anticorpos/farmacologia , Anticorpos/uso terapêutico , Circulação Extracorpórea/efeitos adversos , Fator XIIa/antagonistas & inibidores , Hemorragia/tratamento farmacológico , Hemorragia/etiologia , Trombose/prevenção & controle , Animais , Derivação Arteriovenosa Cirúrgica , Coagulação Sanguínea/efeitos dos fármacos , Domínio Catalítico , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Mapeamento de Epitopos , Fator XIIa/metabolismo , Heparina/farmacologia , Heparina/uso terapêutico , Humanos , Camundongos , Coelhos , Fatores de Risco , Especificidade da Espécie
20.
Thromb Haemost ; 111(2): 189-98, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24136202

RESUMO

Newer oral anticoagulants offer several advantages over traditional agents (e.g. warfarin), but they are still associated with a bleeding risk and currently there is no validated reversal treatment for them. While there is little support for the use of fresh frozen plasma, and limited data available on the effects of activated recombinant factor VII, preclinical data suggest that prothrombin complex concentrates (PCCs) may have potential in this setting. PCCs are currently used to successfully reverse warfarin-induced anticoagulation; however, clinical evidence for their use with new oral anticoagulants is lacking, with most of the available data coming from preclinical animal studies. Furthermore, there appears to be variation in the ability of different PCCs to reverse the coagulopathy induced by the new anticoagulants, and a lack of correlation between the reversal of laboratory test results and the reversal of anticoagulant-induced bleeding. Although there have been encouraging results, care must be taken in generalising findings from animal models and nonbleeding human subjects to the situation in bleeding patients. Ultimately, more evidence supporting anticoagulation reversal for new anticoagulants is needed, particularly regarding the treatment of bleeding in human patients in a clinical setting. According to the current evidence, use of PCCs may be considered a reasonable approach in dire clinical situations; however, a consensus has not yet been reached regarding PCC use or dosing, due to lack of clinical data.


Assuntos
Anticoagulantes/administração & dosagem , Anticoagulantes/antagonistas & inibidores , Fatores de Coagulação Sanguínea/uso terapêutico , Coagulação Sanguínea/efeitos dos fármacos , Coagulantes/uso terapêutico , Hemorragia/prevenção & controle , Administração Oral , Animais , Anticoagulantes/efeitos adversos , Relação Dose-Resposta a Droga , Cálculos da Dosagem de Medicamento , Hemorragia/induzido quimicamente , Humanos , Fatores de Risco , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...