Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Transl Med ; 9(371)2017 01 04.
Article in English | MEDLINE | ID: mdl-28053157

ABSTRACT

Antiplatelet agents are proven efficacious treatments for cardiovascular and cerebrovascular diseases. However, the existing drugs are compromised by unwanted and sometimes life-threatening bleeding that limits drug usage or dosage. There is a substantial unmet medical need for an antiplatelet drug with strong efficacy and low bleeding risk. Thrombin is a potent platelet agonist that directly induces platelet activation via the G protein (heterotrimeric guanine nucleotide-binding protein)-coupled protease-activated receptors PAR1 and PAR4. A PAR1 antagonist is approved for clinical use, but its use is limited by a substantial bleeding risk. Conversely, the potential of PAR4 as an antiplatelet target has not been well characterized. Using anti-PAR4 antibodies, we demonstrated a low bleeding risk and an effective antithrombotic profile with PAR4 inhibition in guinea pigs. Subsequently, high-throughput screening and an extensive medicinal chemistry effort resulted in the discovery of BMS-986120, an orally active, selective, and reversible PAR4 antagonist. In a cynomolgus monkey arterial thrombosis model, BMS-986120 demonstrated potent and highly efficacious antithrombotic activity. BMS-986120 also exhibited a low bleeding liability and a markedly wider therapeutic window compared to the standard antiplatelet agent clopidogrel tested in the same nonhuman primate model. These preclinical findings define the biological role of PAR4 in mediating platelet aggregation. In addition, they indicate that targeting PAR4 is an attractive antiplatelet strategy with the potential to treat patients at a high risk of atherothrombosis with superior safety compared with the current standard of care.


Subject(s)
Antibodies/therapeutic use , Fibrinolytic Agents/therapeutic use , Hemorrhage/drug therapy , Platelet Aggregation Inhibitors/therapeutic use , Receptors, Thrombin/antagonists & inhibitors , Administration, Oral , Animals , Blood Platelets/metabolism , Guinea Pigs , HEK293 Cells , Humans , Inhibitory Concentration 50 , Macaca fascicularis , Male , Protein Domains , Receptor, PAR-1/metabolism , Stroke/drug therapy , Thrombin/chemistry , Thrombosis , Treatment Outcome
2.
J Thromb Thrombolysis ; 34(2): 199-207, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22588534

ABSTRACT

A role for platelets in the pathogenesis of venous thrombosis was suggested by clinical and preclinical studies. However, examination of the platelet receptor, P2Y1, in this area has been limited. The goal of the current study was to examine effects of P2Y1 deletion, or selective antagonism with MRS2500, in oxidative venous thrombosis in mice. The P2Y12 antagonist, clopidogrel, was included as a reference agent. Anesthetized C57BL/6 or genetically modified mice underwent 3.5 or 5 % FeCl(3)-induced vena cava thrombosis. Pharmacokinetic properties of MRS2500 were defined for dose selection. Platelet aggregation and renal or tail bleeding times (BT) were measured to put antithrombotic effects into perspective. P2Y1 deletion significantly reduced (p < 0.001) venous thrombus weight by 74 % in 3.5 % FeCl(3) injury compared to P2Y1(+/+) littermates. MRS2500 (2 mg/kg, i.v.) significantly decreased (p < 0.001) thrombus weight 64 % in C57BL/6 mice. In the more severe 5 % FeCl(3)-induced injury model, thrombus weight significantly (p < 0.001) decreased 68 % in P2Y1(-/-) mice versus P2Y1(+/+) mice, and MRS2500 (2 mg/kg) was also beneficial (54 % decrease, p < 0.01). Renal BT doubled in P2Y1(-/-) versus P2Y1(+/+) mice, and increased threefold with MRS2500 compared to vehicle. Tail BT was markedly prolonged in P2Y1(-/-) mice (7.9X) and in C57BL/6 mice given MRS2500. The current study demonstrates that P2Y1 deletion or antagonism significantly reduced venous thrombosis in mice, suggesting that P2Y1 receptors play a role in the pathogenesis of venous thrombosis, at least in this species. However as with many antithrombotic agents the benefit comes at the potential price of an increase in provoked bleeding times.


Subject(s)
Deoxyadenine Nucleotides/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Receptors, Purinergic P2Y1/metabolism , Venae Cavae , Venous Thrombosis/drug therapy , Animals , Blood Platelets/metabolism , Chlorides/adverse effects , Chlorides/pharmacology , Ferric Compounds/adverse effects , Ferric Compounds/pharmacology , Gene Deletion , Mice , Mice, Knockout , Noxae/adverse effects , Noxae/pharmacology , Platelet Aggregation/drug effects , Platelet Aggregation/genetics , Receptors, Purinergic P2Y1/genetics , Venous Thrombosis/chemically induced , Venous Thrombosis/genetics
3.
Thromb Haemost ; 107(6): 1141-50, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22398951

ABSTRACT

Plasma kallikrein is a multifunctional serine protease involved in contact activation of coagulation. Deficiency in humans is characterised by prolonged activated partial thromboplastin time (aPTT); however, the balance between thrombosis and haemostasis is not fully understood. A study of plasma kallikrein-deficient mice revealed increased aPTT, without prolonged bleeding time. Prekallikrein antisense oligonucleotide (ASO) treatment in mice suggested potential for a positive therapeutic index. The current goal was to further define the role of plasma kallikrein in coagulation. Blood pressure and heart rate were normal in plasma kallikrein-deficient mice, and mice were completely protected from occlusion (100 ± 1.3% control flow) in 3.5% FeCl3 -induced arterial thrombosis versus heterozygotes (20 ± 11.4%) and wild-type littermates (8 ± 0%). Vessels occluded in 8/8 wild-type, 7/8 heterozygotes, and 0/8 knockouts. Anti-thrombotic protection was less pronounced in 5% FeCl3-induced arterial injury. Integrated blood flow was 8 ± 0% control in wild-type and heterozygotes, and significantly (p<0.01) improved to 43 ± 14.2% in knockouts. The number of vessels occluded was similar in all genotypes. Thrombus weight was significantly reduced in knockouts (-47%) and heterozygotes (-23%) versus wild-type in oxidative venous thrombosis. Average tail bleeding time increased modestly in knockout mice compared to wild-type. Average renal bleeding times were similar in all genotypes. These studies confirm and extend studies with prekallikrein ASO, and demonstrate that plasma kallikrein deletion prevents occlusive thrombus formation in mice with a minimal role in provoked bleeding. Additional support for the significance of the intrinsic pathway in the coagulation cascade is provided, as well as for a potential new anti-thrombotic approach.


Subject(s)
Hemostasis , Plasma Kallikrein/metabolism , Prekallikrein/metabolism , Thrombosis/prevention & control , Animals , Bleeding Time , Chlorides , Disease Models, Animal , Ferric Compounds , Hemorrhage/blood , Hemorrhage/genetics , Hemostasis/genetics , Heterozygote , Mice , Mice, Knockout , Oligonucleotides, Antisense/metabolism , Partial Thromboplastin Time , Phenotype , Plasma Kallikrein/genetics , Prekallikrein/genetics , Thrombosis/blood , Thrombosis/chemically induced , Thrombosis/genetics , Time Factors , Venous Thrombosis/blood , Venous Thrombosis/chemically induced , Venous Thrombosis/genetics , Venous Thrombosis/prevention & control
4.
Thromb Res ; 127(6): 560-4, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21396684

ABSTRACT

INTRODUCTION: Published evidence suggests that phosphoinositide 3 kinase-ß (PI3K-ß) plays an important role in platelet aggregation and shear activation. TGX-221 is a selective PI3K-ß inhibitor with a good separation of anti-thrombotic efficacy and bleeding (therapeutic index) in rats. Our goal was to further evaluate potential of a PI3K-ß inhibitor as an anti-thrombotic agent by determining the therapeutic index in another species and efficacy model. Reported effects of TGX-221 in rats were also confirmed. MATERIALS AND METHODS: TGX-221 (0.3 + 0.3, 1 + 1, 3 + 3 mg/kg + mg/kg/hr, i.v.) or vehicle was given to mice starting 15 min prior to FeCl(3) arterial thrombosis (AT), tail or kidney bleeding time (BT) procedures. RESULTS: Integrated blood flow over 30 min (%baseline mean ± SEM) improved (p < 0.05) with TGX-221 doses 1 + 1 (49 ± 13.9%) and 3+3 (88 ± 10.6%) versus 0.3 + 0.3 (10 ± 0.8%) and vehicle (10 ± 0.6%). Vascular patency (non-occluded/total arteries) improved (p < 0.01) with TGX-221 doses of 3 + 3 (7/8), but not 0.3 + 0.3 (0/8) or 1 + 1 (4/8) versus vehicle (0/8). Tail BT (sec) increased (p < 0.05) with TGX-221 doses of 3 + 3 (median 1560) and 1 + 1 (1305) versus vehicle (225). Mean renal BT (sec) increased (p < 0.05) in all TGX-221 groups (3 + 3: 510 + 26; 1 + 1: 478 + 41; 0.3 + 0.3: 246 + 37) versus vehicle (123 + 9). For comparison, a reference agent, aspirin (30 mpk, i.p.) increased tail BT 1.9X and renal BT 2.6X. CONCLUSIONS: The novel finding of a clear impact on hemostasis by TGX-221 was demonstrated by increased bleeding in two models in mice at anti-thrombotic doses. The results suggest a narrower therapeutic index for this PI3K-ß inhibitor than previously recognized, at least for this species.


Subject(s)
Blood Platelets/drug effects , Fibrinolytic Agents/pharmacology , Morpholines/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Pyrimidinones/pharmacology , Animals , Bleeding Time , Carotid Artery Thrombosis/blood , Carotid Artery Thrombosis/drug therapy , Carotid Artery Thrombosis/enzymology , Disease Models, Animal , Fibrinolytic Agents/blood , Fibrinolytic Agents/toxicity , Hemorrhage/chemically induced , Humans , Kidney/blood supply , Male , Mice , Mice, Inbred C57BL , Morpholines/blood , Morpholines/toxicity , Phosphatidylinositol 3-Kinases/blood , Pyrimidinones/blood , Pyrimidinones/toxicity , Rats , Rats, Sprague-Dawley , Tail/blood supply
5.
Thromb Res ; 123(1): 146-58, 2008.
Article in English | MEDLINE | ID: mdl-18479740

ABSTRACT

INTRODUCTION: Animal models of thrombosis and hemostasis are critical for target validation in pharmaceutical research. Guinea pig haemostatic mechanisms, such as the platelet thrombin receptor repertoire, resemble those of humans. Measuring the performance characteristics of marketed antithrombotic drugs in guinea pig models is a key to predicting therapeutic indices of new agents. The goal of the current study was to benchmark representative marketed drugs in thrombosis and hemostasis models in guinea pigs. METHODS: Effects of the cyclooxygenase inhibitor, aspirin, the P2Y(12) antagonist, clopidogrel, the glycoprotein IIb/IIIa inhibitor, tirofiban, and the direct thrombin inhibitors, argatroban and hirudin, were evaluated in this study. Antithrombotic agents were tested in FeCl(3)-induced carotid artery thrombosis and arterio-venous shunt thrombosis models. Haemostatic effects of drugs were evaluated in cuticle and renal bleeding models. Ex vivo measurements of platelet function and coagulation inhibition were performed to benchmark preclinical doses of each agent to those used clinically. RESULTS: The overall rank-order of potency in thrombosis models based on per cent of vessels occluded, average carotid blood flow, and thrombus weight was aspirin=argatroban=tirofiban

Subject(s)
Anticoagulants/therapeutic use , Hemostasis/drug effects , Platelet Aggregation Inhibitors/therapeutic use , Thrombosis/drug therapy , Animals , Arginine/analogs & derivatives , Arteriovenous Shunt, Surgical/adverse effects , Aspirin/therapeutic use , Bleeding Time , Chlorides , Ferric Compounds/pharmacology , Guinea Pigs , Male , Pipecolic Acids/therapeutic use , Sulfonamides , Tirofiban , Tyrosine/analogs & derivatives , Tyrosine/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL