Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters











Database
Language
Publication year range
1.
Pharm Res ; 33(6): 1517-26, 2016 06.
Article in English | MEDLINE | ID: mdl-26960296

ABSTRACT

PURPOSE: Activated (super)Factor V ((super)FVa) is a novel engineered FV with excellent prohemostatic efficacy. (Super)FVa has three APC cleavage site mutations and an interdomain disulfide bond. Stability, pharmacokinetics, and immunogenic and thrombogenic potential are reported here. METHODS: Stability and circulating half-life were determined after incubation in buffer and human plasma, and after injection into FVIII-deficient mice. Immunogenicity potential was assessed by B- and T-cell specific epitope prediction and structural analysis using surface area and atomic depth computation. Thrombogenic potential was determined by quantification of lung fibrin deposition in wild-type mice after intravenous injection of (super)FVa (200 U/kg), recombinant human (rh) Tissue Factor (0.4-16 pmol/kg), rhFVIIa (3 mg/kg) or saline. RESULTS: FVa retained full activity over 30 h in buffer, the functional half-life in human plasma was 4.9 h, and circulating half-life in FVIII-deficient mice was ~30 min. Predicted immunogenicity was not increased compared to human FV. While rh Tissue Factor, the positive control, resulted in pronounced lung fibrin depositions (mean 121 µg/mL), (super)FVa did not (6.7 µg/mL), and results were comparable to fibrin depositions with rhFVIIa (7.6 µg/mL) or saline (5.6 µg/mL). CONCLUSION: FVa has an appropriate safety and stability profile for further preclinical development as a prohemostatic against severe bleeding.


Subject(s)
Factor Va/pharmacokinetics , Hemophilia A/drug therapy , Hemostatics/pharmacokinetics , Protein Engineering/methods , Recombinant Proteins/pharmacokinetics , Animals , Disease Models, Animal , Drug Stability , Factor VIII/genetics , Factor VIII/metabolism , Factor Va/administration & dosage , Factor Va/genetics , Factor Va/toxicity , Female , Fibrin/metabolism , Half-Life , Hemophilia A/blood , Hemophilia A/genetics , Hemostatics/administration & dosage , Hemostatics/toxicity , Humans , Injections, Intravenous , Lung/drug effects , Lung/metabolism , Male , Mice, Inbred BALB C , Mice, Knockout , Mutagenesis, Site-Directed , Mutation , Protein Stability , Recombinant Proteins/administration & dosage , Recombinant Proteins/genetics , Recombinant Proteins/toxicity , Severity of Illness Index , Thrombin/metabolism
2.
Thromb Haemost ; 115(3): 551-61, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26466980

ABSTRACT

Bypassing inhibitors in haemophilia patients is limited to activated (a) Factor(F)VII products. We introduced "FVa activity augmentation" as another bypassing strategy and studied effects of an engineered FVa variant designated superFVa. Procoagulant and clot stabilising properties of superFVa and recombinant human (rh)FVIIa, either alone or in combination, were studied in thrombin generation and clot lysis assays in normal human plasma (NHP) with or without anti-FVIII inhibitors, in haemophilia plasma, and in FVIII-deficient mice or in wild-type mice with anti-FVIII inhibitors. SuperFVa was as effective as rhFVIIa to improve thrombin generation or clot lysis. Furthermore, procoagulant effects were significantly enhanced when these compounds were combined. RhFVIIa at 40 nM (a therapeutic concentration) improved thrombin generation mildly, but markedly improved thrombin generation when combined with a low concentration (e. g. 3 nM) of superFVa. In clot lysis studies, the concentration of rhFVIIa to normalise clot lysis times could be reduced by 100-fold (e. g. from 40 nM to 0.4 nM) when combined with a low concentration (0.37 nM) of superFVa. In haemostasis studies of FVIII-deficient mice, blood loss was dose-dependently reduced by either superFVa or rhFVIIa. SuperFVa (200 U/kg) corrected mean blood loss indistinguishably from rhFVIII. Blood loss correction by rhFVIIa was greatly improved when combined with superFVa. Similar blood loss correction results were observed for therapies in wild-type mice after infusion with anti-FVIII inhibitors. Thus, superFVa may be an effective procoagulant agent in the setting of haemophilia with inhibitors and it merits further evaluation for new bypassing strategies.


Subject(s)
Blood Coagulation/drug effects , Coagulants/administration & dosage , Factor VIIa/administration & dosage , Factor Va/administration & dosage , Hemophilia A/immunology , Animals , Antibodies/chemistry , Dose-Response Relationship, Drug , Factor VIII/antagonists & inhibitors , Female , Hemophilia A/blood , Hemostasis , Humans , Male , Mice , Mice, Inbred BALB C , Recombinant Proteins/administration & dosage , Thrombin/metabolism
3.
Blood ; 125(23): 3647-50, 2015 Jun 04.
Article in English | MEDLINE | ID: mdl-25896652

ABSTRACT

Whole genome sequencing of an individual completely devoid of plasma- and platelet-derived factor V (FV) identified 167 variants in his F5 gene including previously identified and damaging missense mutations at rs6027 and Leu90Ser. Because the administration of fresh frozen plasma (FFP) prevents gastrointestinal bleeding in this individual, its effects on his plasma- and platelet-derived FV concentrations were assessed. The patient's plasma FV levels peaked by 2 hours following FFP administration and were undetectable 96 hours later. In contrast, increased platelet-derived FV/Va concentrations were observed within 6 hours, peaked at 24 hours, decreased slowly over 7 days, and originated from megakaryocyte endocytosis and intracellular processing of plasma FV. Ten days after transfusion, no thrombin was generated in a tissue factor-initiated whole blood clotting assay unless exogenous FV was added, consistent with the complete absence of plasma FV. In marked contrast, release of the patient's platelet-derived FV/Va (7% of normal) following platelet activation resulted in robust thrombin generation, similar to that in an individual with normal plasma- and platelet-derived FV concentrations. Thus, total FV deficiency can be corrected by plasma administration, which partially repletes and sustains the platelet cofactor pool, thereby highlighting the critical role of platelet-derived FV/Va in ensuring hemostatic competence.


Subject(s)
Blood Component Transfusion , Blood Platelets , Factor V Deficiency/blood , Factor V Deficiency/therapy , Factor Va/administration & dosage , Plasma , Aged , Amino Acid Substitution , Factor V Deficiency/complications , Factor V Deficiency/genetics , Factor Va/genetics , Factor Va/metabolism , Gastrointestinal Hemorrhage/blood , Gastrointestinal Hemorrhage/etiology , Gastrointestinal Hemorrhage/genetics , Gastrointestinal Hemorrhage/therapy , Humans , Male , Megakaryocytes/metabolism , Megakaryocytes/pathology , Mutation, Missense , Thrombin Time
4.
PLoS One ; 9(8): e104304, 2014.
Article in English | MEDLINE | ID: mdl-25127130

ABSTRACT

OBJECTIVE: An increased risk of bleeding is observed in patients receiving activated protein C (APC), which may be a limiting factor for the application of novel APC therapies. Since APC's therapeutic effects often require its cytoprotective activities on cells but not APC's anticoagulant activities, an agent that specifically antagonizes APC's anticoagulant effects but not its cytoprotective effects could provide an effective means to control concerns for risk of bleeding. We hypothesized that superFVa, an engineered activated FVa-variant that restores hemostasis in hemophilia could reduce APC-induced bleeding. APPROACH AND RESULTS: SuperFVa was engineered with mutations of the APC cleavage sites (Arg506/306/679Gln) and a disulfide bond (Cys609-Cys1691) between the A2 and A3 domains, which augment its biological activity and cause high resistance to APC. SuperFVa normalized APC-prolonged clotting times and restored APC-suppressed thrombin generation in human and murine plasma at concentrations where wild-type (wt) FVa did not show effects. Following intravenous injection of APC into BALB/c mice, addition to whole blood ex vivo of superFVa but not wt-FVa significantly normalized whole blood clotting. Blood loss following tail clip or liver laceration was significantly reduced when superFVa was administered intravenously to BALB/c mice prior to intravenous APC-treatment. Furthermore, superFVa abolished mortality (∼50%) associated with excessive bleeding following liver laceration in mice treated with APC. CONCLUSIONS: Our results provide proof of concept that superFVa is effective in preventing APC-induced bleeding and may provide therapeutic benefits as a prohemostatic agent in various situations where bleeding is a serious risk.


Subject(s)
Anticoagulants/adverse effects , Factor Va/pharmacology , Hemorrhage/etiology , Hemorrhage/prevention & control , Protein C/adverse effects , Recombinant Proteins/pharmacology , Animals , Blood Coagulation/drug effects , Factor Va/administration & dosage , Female , Hemorrhage/mortality , Hemostasis/drug effects , Mice , Models, Animal , Prothrombin Time , Recombinant Proteins/administration & dosage , Thrombin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL