Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Adv Healthc Mater ; 3(1): 30-5, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23788402

ABSTRACT

Rapid one-step modification of thrombomodulin with alkylamine derivatives such as azide, biotin, and PEG is achieved using an evolved sortase (eSrtA) mutant. The feasibility of a point-of-care scheme is demonstrated herein to site-specifically immobilize azido-thrombomodulin on sterilized commercial ePTFE vascular grafts, which exhibit superior thromboresistance compared with commercial heparin-coated grafts in a primate model of acute graft thrombosis.


Subject(s)
Amines/chemistry , Thrombomodulin/chemistry , Amines/metabolism , Aminoacyltransferases/metabolism , Animals , Azides/chemistry , Azides/metabolism , Bacterial Proteins/metabolism , Biotin/chemistry , Biotin/metabolism , Blood Platelets/chemistry , Blood Platelets/metabolism , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/metabolism , Cysteine Endopeptidases/metabolism , Disease Models, Animal , Heparin/chemistry , Heparin/metabolism , Heparin/therapeutic use , Immobilized Proteins/chemistry , Immobilized Proteins/metabolism , Papio , Polyethylene Glycols/chemistry , Polyethylene Glycols/metabolism , Polytetrafluoroethylene/chemistry , Polytetrafluoroethylene/metabolism , Staphylococcus aureus/enzymology , Thrombomodulin/metabolism , Thrombosis/drug therapy
2.
Thromb Haemost ; 110(1): 162-72, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23636219

ABSTRACT

Rivaroxaban is an oral, direct factor Xa inhibitor for the management of thromboembolic disorders. Despite its short half-life, the ability to reverse rivaroxaban anticoagulation could be beneficial in life-threatening emergencies. The potential of prothrombin complex concentrate (PCC; Beriplex®), activated PCC (aPCC; FEIBA®) or recombinant activated factor VII (rFVIIa; NovoSeven®) to reverse rivaroxaban in rats and baboons was investigated. Anaesthetised rats pre-treated with intravenous rivaroxaban (2 mg/kg) received intravenous rFVIIa (100/400 µg/kg), PCC (25/50 U/kg) or aPCC (50/100 U/kg) after initiation of bleeding. Clotting times and bleeding times (BTs) were recorded. Rivaroxaban was administered as an intravenous 0.6 mg/kg bolus followed by continuous 0.6 mg/kg/hour infusion in baboons. Animals received intravenous aPCC 50 U/kg (2 U/kg/minute) or rFVIIa 210 µg/kg. BT and clotting parameters were measured. In rats pretreated with high-dose rivaroxaban, PCC 50 U/kg, aPCC 100 U/kg and rFVIIa 400 µg/kg significantly reduced BT vs rivaroxaban alone (5.4 ± 1.4-fold to 1.5 ± 0.4-fold [p<0.05]; 3.0 ± 0.4-fold to 1.4 ± 0.1-fold [p<0.001]; and 3.5 ± 0.7-fold to 1.7 ± 0.2-fold [p<0.01] vs baseline, respectively). In baboons pre-infused with rivaroxaban and then given aPCC, BT increased by 2.0 ± 0.2-fold and aPCC returned BT to baseline for the duration of its infusion. rFVIIa reduced BT from 2.5 ± 0.3-fold over baseline to 1.7 ± 0.3-fold over baseline. Prolongation of prothrombin time was reduced by PCC, aPCC and rFVIIa in both species. Rivaroxaban reduced thrombin-antithrombin levels; application of PCC and aPCC, but not rFVIIa, increased these levels. In conclusion, PCC, aPCC or rFVIIa have the potential to reverse the anticoagulant and anti-haemostatic effects of rivaroxaban.


Subject(s)
Anticoagulants/administration & dosage , Morpholines/administration & dosage , Thiophenes/administration & dosage , Thromboembolism/drug therapy , Animals , Bleeding Time , Blood Coagulation/drug effects , Blood Coagulation Factors/administration & dosage , Factor VIIa/administration & dosage , Humans , Male , Primates , Prothrombin Time , Rats , Rats, Wistar , Recombinant Proteins/administration & dosage , Rivaroxaban
3.
Adv Funct Mater ; 21(24): 4736-4743, 2011 Dec 20.
Article in English | MEDLINE | ID: mdl-23532366

ABSTRACT

Activation of hemostatic pathways by blood-contacting materials remains a major hurdle in the development of clinically durable artificial organs and implantable devices. We postulate that surface-induced thrombosis may be attenuated by the reconstitution onto blood contacting surfaces of bioactive enzymes that regulate the production of thrombin, a central mediator of both clotting and platelet activation cascades. Thrombomodulin (TM), a transmembrane protein expressed by endothelial cells, is an established negative regulator of thrombin generation in the circulatory system. Traditional techniques to covalently immobilize enzymes on solid supports may modify residues contained within or near the catalytic site, thus reducing the bioactivity of surface enzyme assemblies. In this report, we present a molecular engineering and bioorthogonal chemistry approach to site-specifically immobilize a biologically active recombinant human TM fragment onto the luminal surface of small diameter prosthetic vascular grafts. Bioactivity and biostability of TM modified grafts is confirmed in vitro and the capacity of modified grafts to reduce platelet activation is demonstrated using a non-human primate model. These studies indicate that molecularly engineered interfaces that display TM actively limit surface-induced thrombus formation.

4.
Sci Transl Med ; 2(37): 37ra45, 2010 Jun 23.
Article in English | MEDLINE | ID: mdl-20574069

ABSTRACT

Most heart attacks and strokes are caused by blood clots (thrombi) that block the vasculature. Because disease-causing arterial thrombosis depends on blood platelets, platelet inhibitors such as aspirin and clopidogrel effectively decrease the risk of thrombosis; however, they also impair platelet-dependent hemostasis that staunches bleeding from wounds and can therefore produce excessive bleeding. Experimental studies show that a reduction in the number of platelets also inhibits thrombosis, but these treatments also interfere with platelet function. Because normal hemostasis requires that the platelet concentration remain within a physiological range in the circulation, we evaluated whether lowering the number of circulating platelets--but only to a value still within the normal range--by inhibiting platelet formation in the bone marrow inhibits acute thrombogenesis in baboons. We reduced the platelet count with an inhibitor against the megakaryocyte-promoting hormone thrombopoietin and then showed that experimental occlusive thrombogenesis on collagen-coated vascular grafts was reduced, without impairment of primary hemostasis. These results suggest that suppressing platelet production without interfering with the hemostatic function of platelets may offer a safe alternative to current therapies for prevention of stroke and heart attack triggered by blood clotting.


Subject(s)
Antithrombins/adverse effects , Antithrombins/pharmacology , Platelet Aggregation Inhibitors/adverse effects , Platelet Aggregation Inhibitors/pharmacology , Thrombopoietin/antagonists & inhibitors , Thrombosis/prevention & control , Animals , Bleeding Time , Blood Vessel Prosthesis , Heart Arrest/etiology , Heart Arrest/prevention & control , Humans , Male , Papio , Platelet Count , Stroke/etiology , Stroke/prevention & control , Thrombosis/blood , Thrombosis/complications , Thrombosis/pathology , Treatment Outcome
5.
Thromb Haemost ; 103(1): 224-33, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20062929

ABSTRACT

Tissue factor (TF) antagonists targeting the factor VII (FVII) binding domain have been shown to interrupt acute vascular thrombus formation without impairing haemostasis in non-human primates. In this study, we evaluate whether a human/mouse chimeric monoclonal antibody (ALT-836, formerly known as Sunol-cH36) blocking the factor X/factor IX (FX/FIX) binding site of tissue factor could achieve similar clinical benefits in an arterial thrombosis model induced by surgical endarterectomy in chimpanzees. In this model, sequential surgical endarterectomies on right and left superficial femoral arteries were performed 30 days apart in five chimpanzees. A bolus (1 mg/kg) of ALT-836 was injected intravenously immediately preceding the restoration of flow in the endarterectomised femoral artery. Pre-surgical labelling of autologous platelets using (111)In-Oxine and post-surgical gamma camera imaging of (111)In-platelet deposition at endarterectomy sites was performed. The manipulated arterial segments were harvested for patency analysis 30 days following surgery. The results indicate that ALT-836 was highly effective at reducing acute vascular thrombosis, with no significant variations in surgical blood loss and template-bleeding time in the treated group compared to the control animals. These data suggest that ALT-836 is an effective and safe antithrombotic agent in preventing TF-initiated vascular thrombogenesis without compromising haemostasis.


Subject(s)
Antibodies, Monoclonal/pharmacology , Factor X/metabolism , Fibrinolytic Agents/pharmacology , Thromboplastin/antagonists & inhibitors , Thrombosis/prevention & control , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/pharmacokinetics , Antibody Specificity , Binding Sites , Blood Coagulation/drug effects , Blood Loss, Surgical/prevention & control , CHO Cells , Cricetinae , Cricetulus , Disease Models, Animal , Dose-Response Relationship, Drug , Endarterectomy , Factor IX/metabolism , Factor VIIa/metabolism , Female , Femoral Artery/surgery , Fibrinolytic Agents/administration & dosage , Fibrinolytic Agents/adverse effects , Fibrinolytic Agents/pharmacokinetics , Hemorrhage/chemically induced , Humans , Injections, Intravenous , Mice , Mice, Inbred BALB C , Pan troglodytes , Radionuclide Imaging , Recombinant Fusion Proteins/pharmacology , Thromboplastin/immunology , Thromboplastin/metabolism , Thrombosis/blood , Thrombosis/diagnostic imaging , Thrombosis/etiology
6.
Blood ; 113(4): 936-44, 2009 Jan 22.
Article in English | MEDLINE | ID: mdl-18945968

ABSTRACT

The protease thrombin is required for normal hemostasis and pathologic thrombogenesis. Since the mechanism of coagulation factor XI (FXI)-dependent thrombus growth remains unclear, we investigated the contribution of FXI to thrombus formation in a primate thrombosis model. Pretreatment of baboons with a novel anti-human FXI monoclonal antibody (aXIMab; 2 mg/kg) inhibited plasma FXI by at least 99% for 10 days, and suppressed thrombin-antithrombin (TAT) complex and beta-thromboglobulin (betaTG) formation measured immediately downstream from thrombi forming within collagen-coated vascular grafts. FXI inhibition with aXIMab limited platelet and fibrin deposition in 4-mm diameter grafts without an apparent increase in D-dimer release from thrombi, and prevented the occlusion of 2-mm diameter grafts without affecting template bleeding times. In comparison, pretreatment with aspirin (32 mg/kg) prolonged bleeding times but failed to prevent graft occlusion, supporting the concept that FXI blockade may offer therapeutic advantages over other antithrombotic agents in terms of bleeding complications. In whole blood, aXIMab prevented fibrin formation in a collagen-coated flow chamber, independent of factor XII and factor VII. These data suggest that endogenous FXI contributes to arterial thrombus propagation through a striking amplification of thrombin generation at the thrombus luminal surface.


Subject(s)
Factor XI/metabolism , Thrombin/metabolism , Thrombosis/metabolism , Thrombosis/prevention & control , Animals , Antibodies, Monoclonal/immunology , Bleeding Time , Blood Platelets/drug effects , Blood Platelets/metabolism , Collagen/pharmacology , Factor XI/antagonists & inhibitors , Factor XI/immunology , Factor XII/metabolism , Fibrin/metabolism , Humans , Male , Papio , Platelet Activation/immunology , Thrombosis/immunology
7.
Tissue Eng Part C Methods ; 14(2): 97-105, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18544026

ABSTRACT

In humans, self-endothelialization of synthetic grafts is severely limited, but a recent interesting idea is to attract endothelial progenitor cells (EPCs) from peripheral blood onto grafts via antibodies directed at proposed EPC markers. Results with anti-CD34 antibodies have shown some promise, but it is unclear whether CD34 is the best marker for cells with re-endothelializing potential. Much evidence points to kinase insert domain receptor (KDR) as an important indicator of endothelial potential if not a definitive marker. Because KDR is not an adhesion molecule (like CD34), we first demonstrated the ability to use adsorbed and protein G-oriented antibody to this receptor to capture flowing cells onto a solid surface. Using endothelial cells and smooth muscle cells, we show in a model system under low shear rates the ability to selectively capture cells by this receptor. Furthermore, our results indicate that concomitant flow of cells lacking the receptor does not affect the efficiency of capture of KDR(+) cells but that orienting the antibody significantly increases the efficiency of capture.


Subject(s)
Antigens, CD34/chemistry , Tissue Engineering/methods , Animals , Antigens, CD34/biosynthesis , Blood Platelets/metabolism , Cells, Cultured , Endothelium, Vascular/metabolism , Humans , Models, Biological , Myocytes, Smooth Muscle/metabolism , Papio , Protein Structure, Tertiary , Surface Properties , Thrombosis/metabolism , Transforming Growth Factor beta1/metabolism
8.
Ann Biomed Eng ; 36(1): 1-13, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17985244

ABSTRACT

In the United States, over 125,000 mechanical heart valves (MHVs) are implanted each year. Flow through the MHV hinge can cause thromboemboli formation. The purpose of this study was to examine various orifice geometries representing the MHV hinge region and how these geometries may contribute to platelet activation and thrombin generation. We also characterized these flow fields with digital particle image velocimetry (DPIV). Citrated human blood at room temperature was forced through the orifices (400 and 800 microm ID) with a centrifugal bypass pump, continuously infusing calcium chloride to partially reverse the citrate anticoagulant. Blood samples were tested for the presence of thrombin-antithrombin complex (TAT) and platelet factor 4 (PF4). Velocity and shear stress were measured with DPIV using a blood analog fluid seeded with fluorescent microbeads. The results indicate that small changes in geometry, although they do not affect the bulk flow, change the coagulation propensity as blood flows through the orifices. A more abrupt geometry allows more stagnation to occur resulting in more thrombin generation. PF4 measurements indicated similar levels of platelet activation for all orifices. DPIV showed differences in the jets with respect to entrainment of stagnant fluid. These results help to pinpoint the important parameters that lead to flow stasis and subsequent thrombus formation.


Subject(s)
Blood Coagulation , Blood Flow Velocity , Heart Valve Diseases/etiology , Heart Valve Diseases/physiopathology , Heart Valve Prosthesis/adverse effects , Thrombosis/etiology , Thrombosis/physiopathology , Animals , Computer Simulation , Constriction, Pathologic/etiology , Constriction, Pathologic/physiopathology , Humans , Models, Cardiovascular
9.
Br J Haematol ; 139(3): 494-503, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17910639

ABSTRACT

The minor gammaA/gamma' isoform of fibrinogen contains a high affinity binding site for thrombin exosite II that is lacking in the major fibrinogen isoform, gammaA/gammaA fibrinogen. The biological consequences of gamma' chain binding to thrombin were therefore investigated. Coagulation assays, thrombin activity assays, and a primate thrombosis model were used to characterize the biological effects of the gamma' 410-427 peptide. The gamma' peptide had little effect on thrombin cleavage of the small peptidyl substrate tosyl-glycyl-prolyl-arginine-4-nitranilide acetate. However, in vitro assays demonstrated that the gamma' peptide inhibited thrombin cleavage of larger proteinaceous substrates, including fibrinogen and factor VIII. The gamma' peptide inhibited the activated partial thromboplastin time in plasma and showed greater inhibition of activated partial thromboplastin time assays than prothrombin time assays, consistent with the inhibition of factor VIII cleavage. Studies in a baboon thrombosis model showed that the gamma' 410-427 peptide inhibited fibrin-rich thrombus formation (typical of venous thrombi) and, to a lesser extent, platelet-rich thrombus formation (typical of arterial thrombi). These results indicate that binding of thrombin exosite II by the gamma' peptide has selective effects on the intrinsic pathway.


Subject(s)
Anticoagulants/pharmacology , Blood Coagulation/drug effects , Fibrinogen/pharmacology , Peptide Fragments/pharmacology , Animals , Anticoagulants/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Factor VIII/metabolism , Fibrinogen/metabolism , Male , Papio , Partial Thromboplastin Time , Peptide Fragments/metabolism , Prothrombin Time , Thrombin/metabolism , Thrombosis/prevention & control
10.
Thromb Res ; 121(3): 397-406, 2007.
Article in English | MEDLINE | ID: mdl-17532367

ABSTRACT

INTRODUCTION: Thromboembolic events caused by implanted vascular devices present serious medical challenges. In particular bileaflet mechanical heart valves (MHVs) are prone to thrombus formation in the hinge region due to a combination of high shear stress and stagnation regions. Most studies of shear-induced platelet activation and aggregation have been performed using viscometers, parallel plate flow, and other non-physiologic in vitro configurations. The present study investigated these events in a physiogically relevant environment in which thrombin formation in response to shear stress activation of platelets plays a more predominant role. MATERIALS AND METHODS: Anticoagulated (citrated) human blood was placed in a steady flow loop containing a 400 microm round orifice or various MHVs in the leakage position. Simultaneous blood recalcification enhanced the thrombus forming potential of the blood. Aggrastat and AN51 were used to block binding to the platelet GPIIb/IIIa and GPIb receptors, respectively, and aspirin was used to block thromboxane production. Thrombin generation was measured indirectly by the thrombin-antithrombin III assay. RESULTS AND CONCLUSIONS: Aggrastat, AN51, and aspirin all suppressed thrombin formation. Furthermore, histological results suggested important roles for vWF and fibrinogen in a two-step model of thrombus formation. Thus, thrombin is reproducibly formed in this in vitro system, a process that can be suppressed by blocking platelet activation. This system has the potential to investigate mechanisms and interventions for medical devices that contact with blood under varying shear stress conditions.


Subject(s)
Heart Valve Prosthesis/adverse effects , Platelet Activation/physiology , Thrombin/biosynthesis , Antithrombin III , Aspirin/pharmacology , Biomedical Engineering , Blood Platelets/drug effects , Blood Platelets/metabolism , Hemorheology , Humans , In Vitro Techniques , Peptide Hydrolases/blood , Platelet Aggregation Inhibitors/pharmacology , Stress, Mechanical , Thromboembolism/blood , Thromboembolism/etiology , Tirofiban , Tyrosine/analogs & derivatives , Tyrosine/pharmacology , von Willebrand Factor/metabolism
11.
Blood ; 109(9): 3733-40, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17227834

ABSTRACT

The anticoagulant and anti-inflammatory enzyme, activated protein C (APC), naturally controls thrombosis without affecting hemostasis. We therefore evaluated whether the integrity of primary hemostasis was preserved during limited pharmacological antithrombotic protein C activator (PCA) treatment in baboons. The double-mutant thrombin (Trp215Ala/Glu217Ala) with less than 1% procoagulant activity was used as a relatively selective PCA and compared with systemic anticoagulation by APC and low-molecular-weight heparin (LMWH) at doses that inhibited fibrin deposition on thrombogenic segments of arteriovenous shunts. As expected, both systemic anticoagulants, APC (0.028 or 0.222 mg/kg for 70 minutes) and LMWH (0.325 to 2.6 mg/kg for 70 minutes), were antithrombotic and prolonged the template bleeding time. In contrast, PCA at doses (0.0021 to 0.0083 mg/kg for 70 minutes) that had antithrombotic effects comparable with LMWH did not demonstrably impair primary hemostasis. PCA bound to platelets and leukocytes, and accumulated in thrombi. APC infusion at higher circulating APC levels was less antithrombotic than PCA infusion at lower circulating APC levels. The observed dissociation of antithrombotic and antihemostatic effects during PCA infusion thus appeared to emulate the physiological regulation of intravascular blood coagulation (thrombosis) by the endogenous protein C system. Our data suggest that limited pharmacological protein C activation might exhibit considerable thrombosis specificity.


Subject(s)
Anticoagulants/pharmacology , Hemostasis/drug effects , Hemostatics/pharmacology , Heparin, Low-Molecular-Weight/pharmacology , Protein C/pharmacology , Thrombin/pharmacology , Thrombosis/drug therapy , Amino Acid Substitution , Animals , Anticoagulants/pharmacokinetics , Bleeding Time , Drug Evaluation, Preclinical , Hemostatics/pharmacokinetics , Heparin, Low-Molecular-Weight/pharmacokinetics , Humans , Mutation, Missense , Papio , Protein C/agonists , Protein C/analysis , Protein C/pharmacokinetics , Recombinant Proteins/genetics , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/pharmacology , Thrombin/genetics , Thrombin/pharmacokinetics , Thrombosis/blood
12.
J Biomech Eng ; 128(1): 30-9, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16532615

ABSTRACT

BACKGROUND: While it is established that mechanical heart valves (MHVs) damage blood elements during leakage and forward flow, the role in thrombus formation of platelet activation by high shear flow geometries remains unclear. In this study, continuously recalcified blood was used to measure the effects of blood flow through orifices, which model MHVs, on the generation of procoagulant thrombin and the resulting formation of thrombus. The contribution of platelets to this process was also assessed. METHOD OF APPROACH: 200, 400, 800, and 1200 microm orifices simulated the hinge region of bileaflet MHVs, and 200, 400, and 800 microm wide slits modeled the centerline where the two leaflets meet when the MHV is closed. To assess activation of coagulation during blood recirculation, samples were withdrawn over 0-47 min and the plasmas assayed for thrombin-antithrombin-llI (TAT) levels. Model geometries were also inspected visually. RESULTS: The 200 and 400 microm round orifices induced significant TAT generation and thrombosis over the study interval. In contrast, thrombin generation by the slit orifices, and by the 800 and 1200 microm round orifices, was negligible. In additional experiments with nonrecalcified or platelet-depleted blood, TAT levels were markedly reduced versus the studies with fully anticoagulated whole blood (p < 0.05). CONCLUSIONS: Using the present method, a significant increase in TAT concentration was found for 200 and 400 microm orifices, but not 800 and 1200 microm orifices, indicating that these flow geometries exhibit a critical threshold for activation of coagulation and resulting formation of thrombus. Markedly lower TAT levels were produced in studies with platelet-depleted blood, documenting a key role for platelets in the thrombotic process.


Subject(s)
Blood Flow Velocity , Heart Valve Diseases/etiology , Heart Valve Diseases/physiopathology , Heart Valve Prosthesis/adverse effects , Models, Cardiovascular , Thrombosis/etiology , Thrombosis/physiopathology , Biomechanical Phenomena/methods , Computer Simulation , Humans , Prosthesis Failure
13.
Br J Haematol ; 132(2): 197-203, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16398653

ABSTRACT

We studied whether there was a relationship between the anticoagulant effects of recombinant human soluble thrombomodulin (rhsTM) and activation of protein C in a primate model of acute vascular graft thrombosis in 11 baboons (Papio species). Baboons were pretreated with 0.1, 1 and 5 mg/kg of rhsTM, with or without co-injection of a neutralising monoclonal antibody to protein C (HPC4) in the 1 mg/kg rhsTM group. Subsequently, thrombogenic polyester grafts were deployed for 3 h into chronic exteriorised arteriovenous shunts. Thrombus growth in the graft, plasma-activated protein C (APC) levels, coagulation and thrombosis markers were determined. In untreated baboons, baseline circulating APC levels more than doubled and graft thrombi propagated until reaching equilibrium in about 1 h. Treatment with rhsTM reduced thrombus propagation rates, prolonged the clotting and bleeding times, decreased thrombin-antithrombin complex, beta-thromboglobulin and fibrinopeptide A levels, and, surprisingly, also decreased systemic APC levels, in a dose-dependent manner. In the presence of HPC4 antibody to inhibit APC generation, the acute antithrombotic activity of rhsTM on graft thromboses was not attenuated for up to 80 min, but sustained thrombus accumulation was observed over a 180-min period. These findings suggest that, in contrast to the prevailing hypotheses, the primary antithrombotic activity of rhsTM is independent of protein C, at least in this primate model. Direct inhibition of thrombin's prothrombotic activity upon complex formation with rhsTM might explain the molecular mechanism of the observed antithrombotic effect.


Subject(s)
Anticoagulants/therapeutic use , Graft Occlusion, Vascular/prevention & control , Protein C/physiology , Thrombomodulin/therapeutic use , Animals , Antibodies, Monoclonal/immunology , Antithrombin III , Blood Coagulation , Blood Platelets/drug effects , Blood Platelets/physiology , Dose-Response Relationship, Drug , Fibrin/metabolism , Graft Occlusion, Vascular/blood , Male , Papio , Partial Thromboplastin Time , Peptide Hydrolases/blood , Protein C/immunology , Protein C/metabolism , Recombinant Proteins/therapeutic use , Solubility , Thrombosis/blood , Thrombosis/immunology , Thrombosis/prevention & control
14.
Bioorg Med Chem Lett ; 16(7): 2037-41, 2006 Apr 01.
Article in English | MEDLINE | ID: mdl-16412633

ABSTRACT

Highly selective and potent factor VIIa-tissue factor (fVIIa.TF) complex inhibitors were generated through structure-based design. The pharmacokinetic properties of an optimized analog (9) were characterized in several preclinical species, demonstrating pharmacokinetic characteristics suitable for once-a-day dosing in humans. Analog 9 inhibited platelet and fibrin deposition in a dose-dependent manner after intravenous administration in a baboon thrombosis model, and a pharmacodynamic concentration-response model was developed to describe the platelet deposition data. Results for heparin and enoxaparin (Lovenox) in the baboon model are also presented.


Subject(s)
Factor VIIa/antagonists & inhibitors , Models, Animal , Serine Proteinase Inhibitors/pharmacology , Thrombosis/drug therapy , Animals , Chromatography, High Pressure Liquid , Crystallography, X-Ray , Models, Molecular , Papio , Serine Proteinase Inhibitors/chemistry , Serine Proteinase Inhibitors/pharmacokinetics , Serine Proteinase Inhibitors/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...