scholarly journals A Short Half-Life αIIbβ3 Antagonist ANTP266 Reduces Thrombus Formation

2018 ◽  
Vol 19 (8) ◽  
pp. 2306 ◽  
Author(s):  
Tong-Dan Liu ◽  
Shen-Hong Ren ◽  
Xue Ding ◽  
Zhou-Ling Xie ◽  
Yi Kong

Integrin αIIbβ3 plays a pivotal role in platelet aggregation. Three αIIbβ3 antagonists have been approved by the Food and Drug Administration (FDA) for the treatment of cardiovascular diseases. Unfortunately, all of these three drugs can cause the side effect of severe bleeding. Therefore, developing a new αIIbβ3 antagonist with low bleeding was needed. In the present study, we screened compounds by using a fibrinogen/integrin αIIbβ3 enzyme-linked immunosorbent assay (ELISA), and a novel αIIbβ3 antagonist ANTP266 was attained. The antithrombotic effects of ANTP266 were estimated by using two animal models, the bleeding risk was estimated by using a mice tail cutting assay, and the plasma half-life time was tested by LC-MS/MS. The results showed that ANTP266 potently decreased thrombosis formation, while not prolonging bleeding time at its effective dosage. The bleeding of ANTP266 reduced rapidly as time went on from 5 to 60 min, but tirofiban produced high bleeding continuously. The plasma half-life of ANTP266 in rats was 10.8 min. Taken together, ANTP266 is an effective antithrombotic agent with a low bleeding risk. The shorter bleeding time benefits from its short plasma half-life. ANTP266 could be a candidate for developing the αIIbβ3 antagonist of rapid elimination for a patient undergoing percutaneous coronary intervention.

1991 ◽  
Vol 65 (04) ◽  
pp. 355-359 ◽  
Author(s):  
E Gray ◽  
J Watton ◽  
S Cesmeli ◽  
T W Barrowcliffe ◽  
D P Thomas

SummaryThe in vitro anticoagulant activities of recombinant desulphatohirudin (r-hirudin) were studied in the activated partial thromboplastin time (APTT) and the thrombin generation test : systems. In the APTT at concentrations below 5 μg/ml, r-hirudin showed a dose-response curye. At concentrations above 5 μg/ml, the plasma became unclottable, but in the thrombin generation test , at least 10 μg/ml of r-hirudin was required for full inhibition of thrombin generation. The antithrombotic effect was assessed using a rabbit venous stasis model; 150 μg/ml r-hirudin completely prevented thrombus formation at 10 and 20 min stasis. At antithrombotic dose, the mean bleeding time ratio measured in a rabbit ear template model, was not prolonged over control values. At higher doses, the bleeding time ratios were higher than those observed for the same dosage of heparin. These data indicate that while r-hirudin is an effective antithrombotic agent, antithrombotic doses have to be carefully titrated to avoid excessive bleeding.


1981 ◽  
Author(s):  
M Thiessen ◽  
R Zimmermann ◽  
G Weckesser ◽  
J Harenberg

The influence of aspirin on platelet aggregation has given rise to frequent use as an antithrombotic agent. The knowledge of an unfavorable alteration of the thromboxane/prostacyclin ratio in the vessel wall at high dosage may favor therapy with low dosages of aspirin.In order to obtain further information on the most effective dosage of aspirin the antithrombotic effect of repeated and different doses of aspirin was investigated in a standardized experimental model in 120 rabbits. In a 1. group aspirin 100 mg/kg were given 30 min before producing thrombosis. In two further groups aspirin was administered 12 hours and additionally 30 min before beginning the experiment. These rabbits received repeatedly either 10 mg (2.group) or 100 mg/kg (3. group) two times. In a 4.series aspirin was given at 50 mg/kg per day during three days.A paradoxical thrombogenic effect with an increase of venous and arterial thrombus growth could be seen after a single high dose of 100 mg/kg aspirin but not after an additional one. If applicated two times a thrombus size minor than under repeated low doses of 10 mg/kg aspirin has been observed in the arterial and venous system (p< 0.05).Additionally after repeated treatment with 50 mg/kg an antithrombotic effect could be seen.According to our data the paradoxical effect of only a single or first high dose of aspirin has to be taken into account but cannot be observed after repeated doses. At a low dosage aspirin a paradoxical effect was seen later,after a second dose. Animal thrombosis experiments does not provide sufficient evidence to support antithrombotic therapy with low doses of aspirin.


1996 ◽  
Vol 271 (3) ◽  
pp. E458-E464 ◽  
Author(s):  
C. F. Deacon ◽  
L. Pridal ◽  
L. Klarskov ◽  
M. Olesen ◽  
J. J. Holst

Glucagon-like peptide 1 (GLP-1) metabolism was studied in halothane-anesthetized pigs (n = 7) using processing-independent (PI) and COOH-terminal (C) radioimmunoassays (RIA) and an enzyme-linked immunosorbent assay (ELISA) specific for biologically active GLP-1. Renal extraction of endogenous GLP-1 was detected by PI-RIA (33.1 +/- 13.3%) and C-RIA (16.0 +/- 6.3%) and by all assays during GLP-1 infusion (ELISA, 69.4 +/- 6.3%; PI-RIA, 32.6 +/- 7.3%; C-RIA, 43.7 +/- 3.4%), indicating substantial fragmentation. Hepatic and pulmonary degradation were undetectable under basal conditions, but exogenous GLP-1 elimination by the liver (43.6 +/- 8.9%) and lungs (10.1 +/- 3.2%) was measured by ELISA, suggesting primarily NH2-terminal degradation. Endogenous GLP-1 extraction by the hindleg was only detected by C-RIA (16.0 +/- 6.3%). During GLP-1 infusion, greater hindleg extraction was measured by ELISA (38.5 +/- 6.8%) and C-RIA (33.0 +/- 6.4%) than by PI-RIA (11.4 +/- 3.2%), indicating limited degradation at each terminus or more substantial COOH-terminal degradation. A shorter (P < 0.01) plasma half-life was revealed by ELISA (1.5 +/- 0.4 min) than by PI-RIA (4.5 +/- 0.6 min) or C-RIA (4.1 +/- 0.5 min). Metabolic clearance rates measured by PI-RIA (20.0 +/- 3.8 ml.min-1.kg-1) and C-RIA (15.5 +/- 1.6 ml.min-1.kg-1) were shorter (P < 0.01) than that measured by ELISA (106.8 +/- 14.7 ml.min-1.kg-1). Tissue-specific differential metabolism of GLP-1 occurs, and NH2-terminal degradation, rendering GLP-1 inactive, is particularly important in its clearance.


Author(s):  
Haitao Wu ◽  
Manlin Su ◽  
Hui Jin ◽  
Xinyu Li ◽  
Puyu Wang ◽  
...  

In this paper, we fabricated rutin-loaded silver nanoparticles (Rutin@AgNPs) as the nano-anticoagulant with antithrombotic function. The serum stability, anticoagulation activity, and bleeding risk of Rutin@AgNPs were evaluated. The results showed Rutin@AgNPs had good serum stability, hemocompatibility, and cytocompatibility. The anticoagulation activity of rutin was maintained, and its stability and aqueous solubility were improved. The Rutin@AgNPs could provide a sustained release to prolong the half-life of rutin. The results of the coagulation parameter assay and thrombus formation test in mice model showed that the activated partial thromboplastin time and prothrombin time were prolonged, and Rutin@AgNPs inhibited the thrombosis in the 48 h period. Moreover, the limited bleeding time indicated that the Rutin@AgNPs significantly minimized the hemorrhage risk of rutin. This Rutin@AgNPs is a potential anticoagulant for antithrombotic therapy.


Author(s):  
Albin Jeanne ◽  
Thomas Sarazin ◽  
Magalie Charlé ◽  
Charlotte Kawecki ◽  
Alexandre Kauskot ◽  
...  

Objective: TSP-1 (thrombospondin 1) is one of the most expressed proteins in platelet α-granules and plays an important role in the regulation of hemostasis and thrombosis. Interaction of released TSP-1 with CD47 membrane receptor has been shown to regulate major events leading to thrombus formation, for example, platelet adhesion to vascular endothelium, nitric oxide/cGMP signaling, platelet activation as well as aggregation. Therefore, targeting TSP-1:CD47 axis may represent a promising antithrombotic strategy. Approach and Results: A CD47-derived cyclic peptide was engineered, namely TAX2, that targets TSP-1 and selectively prevents TSP-1:CD47 interaction. Here, we demonstrate for the first time that TAX2 peptide strongly decreases platelet aggregation and interaction with collagen under arterial shear conditions. TAX2 also delays time for complete thrombotic occlusion in 2 mouse models of arterial thrombosis following chemical injury, while Thbs1 −/− mice recapitulate TAX2 effects. Importantly, TAX2 administration is not associated with increased bleeding risk or modification of hematologic parameters. Conclusions: Overall, this study sheds light on the major contribution of TSP-1:CD47 interaction in platelet activation and thrombus formation while putting forward TAX2 as an innovative antithrombotic agent with high added-value.


2011 ◽  
Vol 106 (12) ◽  
pp. 1203-1214 ◽  
Author(s):  
Liang Hu ◽  
Zhichao Fan ◽  
Hongguang Du ◽  
Ran Ni ◽  
Si Zhang ◽  
...  

SummaryThe addition of phosphodiesterase (PDE) inhibitors has been reported to potentiate the antithrombotic effects of P2Y12 antagonists without increasing bleeding risk. In this study, we report that a potent antiplatelet agent, 2-ethylthio-6-phenethylaminoadenosine (BF061), inhibits platelet activation and thrombosis via P2Y12 antagonism and PDE inhibition. We explored the antiplatelet mechanism of BF061 by measuring cAMP, cGMP levels, PDE activity, and the interaction between ADP and P2Y12 using atomic force microscopy. The antithrombotic effect of BF061 was evaluated in mice using intravital microscopy in FeCl3-induced mesenteric and laser-induced cremasteric arterial thrombosis models. BF061 robustly inhibited platelet aggregation and ATP release induced by multiple platelet agonists via P2Y12 antagonism and PDE inhibition. Interestingly, despite being structurally similar to BF061, P2Y12 receptor antagonist AR-C69931MX had no effect on human platelet PDE. In FeCl3-induced mesenteric arterial thrombosis model, BF061 effectively prevented thrombus formation similarly to clopidogrel; it also reduced thrombus volume in laser-injured cremaster arteriole model. In contrast, BF061 induced dramatically less bleeding at an antithrombotic dose compared to clopidogrel. In summary, we developed a novel antiplatelet and antithrombotic agent targeting both P2Y12 and PDE. Given the prevalence of combined antiplatelet therapy in clinical practice, an antiplatelet agent bearing dual activities may have therapeutic advantage as a potential antithrombotic drug.


Blood ◽  
2012 ◽  
Vol 119 (1) ◽  
pp. 251-261 ◽  
Author(s):  
Oliver Borst ◽  
Eva-Maria Schmidt ◽  
Patrick Münzer ◽  
Tanja Schönberger ◽  
Syeda T. Towhid ◽  
...  

Abstract Platelets are activated on increase of cytosolic Ca2+ activity ([Ca2+]i), accomplished by store-operated Ca2+ entry (SOCE) involving the pore-forming ion channel subunit Orai1. Here, we show, for the first time, that the serum- and glucocorticoid-inducible kinase 1 (SGK1) is expressed in platelets and megakaryocytes. SOCE and agonist-induced [Ca2+]i increase are significantly blunted in platelets from SGK1 knockout mice (sgk1−/−). Similarly, Ca2+-dependent degranulation, integrin αIIbβ3 activation, phosphatidylserine exposure, aggregation, and in vitro thrombus formation were significantly impaired in sgk1−/− platelets, whereas tail bleeding time was not significantly enhanced. Platelet and megakaryocyte Orai1 transcript levels and membrane protein abundance were significantly reduced in sgk1−/− mice. In human megakaryoblastic cells (MEG-01), transfection with constitutively active S422DSGK1 but not with inactive K127NSGK1 significantly enhanced Orai1 expression and SOCE, while effects reversed by the SGK1 inhibitor GSK650394 (1μM). Transfection of MEG-01 cells with S422DSGK1 significantly increased phosphorylation of IκB kinase α/β and IκBα resulting in nuclear translocation of NF-κB subunit p65. Treatment of S422DSGK1-transfected MEG-01 cells with the IκB kinase inhibitor BMS-345541 (10μM) abolished SGK1-induced increase of Orai1 expression and SOCE. The present observations unravel SGK1 as novel regulator of platelet function, effective at least in part by NF-κB–dependent transcriptional up-regulation of Orai1 in megakaryocytes and increasing platelet SOCE.


2019 ◽  
Vol 119 (11) ◽  
pp. 1720-1739 ◽  
Author(s):  
Augusto Martins Lima ◽  
Ana C. Martins Cavaco ◽  
Rodrigo A. Fraga-Silva ◽  
Johannes A. Eble ◽  
Nikolaos Stergiopulos

AbstractDespite significant advances in the treatment of thrombogenic diseases, antiplatelet therapies are still associated with a high bleeding risk. Consequently, potential benefits of preventing thromboembolic events by pharmacological agents need to be balanced with the potential harm of inducing hemorrhage. Glycoprotein VI (GPVI) is a platelet-specific receptor, which plays a crucial role in thrombus formation. GPVI deficiency has been identified in patients who suffer from significant reduction of collagen-induced thrombus formation, with a slight tendency for mild bleeding. However, an isolated GPVI deficiency can reduce thrombus formation while not resulting in severe bleeding. Together, these observations strongly suggest that physiological hemostasis does not require GPVI, but pharmacological GPVI modulation may provide novel “bleeding-free” antithrombotic therapies. In this review, we discuss recent findings regarding the biological role of GPVI in platelet-related disorders and highlight the efforts to develop potential therapeutic strategies based on its structure, signaling pathways, and biological effects.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 909-909 ◽  
Author(s):  
Ulla M. Marzec ◽  
Leslie Bush ◽  
Erik Tucker ◽  
Andras Gruber ◽  
Stephen R. Hanson ◽  
...  

Abstract Antithrombotic agents are effective but pose significant bleeding risk, thus safer new agents are needed. Since the infused double mutant human thrombin (W215A/E217A; WE) binds endothelial thrombomodulin, activates protein C locally in the intraluminal boundary layer, and prevents thrombus formation without hemostasis impairment, we investigated whether WE could also interrupt the growth of existing thrombi. WE was compared to standard interventional doses of the low molecular weight heparin, enoxaparin (Sanofi) in awake baboons. Thrombosis was initiated by interposing a two part device consisting of a 2 cm, 4 mm ID polyethylene terephthalate vascular graft (DVG) followed by a 2 cm silicone rubber expansion chamber, 9 mmID. Upon initiation of blood flow at 100 mL/min, wall shear rate of 265 sec-1 in the 4 mm ID segment (arterial type flow) and 29 sec-1 in the expansion chamber (venous type flow), platelet thrombus was monitored by gamma camera imaging of autologous 111In-labelled platelets for 60 min or 90 min. Fibrin accumulation was quantified by homologous 125I-labelled fibrinogen. Hemostasis was assessed as template bleeding time prolongation. APTT was also monitored. Treatments started 25 min after the initiation of thrombosis by enoxaparin, 0.5 mg/kg (LD) and 1 mg/kg (HD) IV bolus or WE, 7.5 μg/kg/hr (LD) and 15 μg/kg/hr (HD) infusion (1/3 dose given as a bolus followed by IV infusion over 60min) and compared to saline infused controls. All interventions were given systemically, downstream from the device and 5–6 studies comprised each study group. Treatment with enoxaparin at 25 min after initiation of thrombus growth resulted in a reduction of platelet deposition of 19 % with the LD and 76 % with the HD after 35 min of therapy in the expanded venous type segment. WE reduced venous thrombosis by 45 % and 65 % at the two doses studied and was further reduced to 65 % and 80 % following 65 min of WE infusion as compared to controls. Arterial type thrombosis was reduced by WE by 34 % and 39 % respectively for the two doses studied. HD enoxaparin reduced arterial type thrombosis by 37 %. Fibrin accumulation paralleled the platelet deposition. APTT was prolonged 1.57- and 1.87-fold respectively for the WE doses without any bleeding time prolongation. Enoxaparin prolonged the APTT 1.4- and 1.75-fold, similar to WE but with a 1.13- and 1.75-fold prolongation of the bleeding time. In conclusion, the double mutant human thrombin, WE, effectively interrupted both venous and arterial type growing thrombi at very low doses without detectable compromise to hemostasis. Likewise, enoxaparin at the doses tested had similar efficacy but with a significantly prolonged bleeding time at the 1 mg/kg bolus dose. WE might provide a safer alternative to heparins in the treatment of acute progressive thrombosis.


Blood ◽  
2012 ◽  
Vol 119 (2) ◽  
pp. 583-586 ◽  
Author(s):  
Edith van de Vijver ◽  
Iris M. De Cuyper ◽  
Anja J. Gerrits ◽  
Arthur J. Verhoeven ◽  
Karl Seeger ◽  
...  

Abstract Patients with Glanzmann thrombasthenia or Leukocyte Adhesion Deficiency-III syndrome (LAD-III or LAD-1/variant) present with increased bleeding tendency because of the lack or dysfunction of the fibrinogen receptor GPIIb/IIIa (integrin αIIbβ3), respectively. Although the bleeding disorder is more severe in LAD-III patients, classic aggregometry or perfusion of Glanzmann or LAD-III platelets over collagen-coated slides under physiologic shear rate does not discriminate between these 2 conditions. However, in a novel flow cytometry-based aggregation assay, Glanzmann platelets were still capable of forming small aggregates upon collagen stimulation, whereas LAD-III platelets were not. These aggregates required functional GPIa/IIa (integrin α2β1) instead of integrin αIIbβ3, thus explaining the clinically more severe bleeding manifestations in LAD-III patients, in which all platelet integrins are functionally defective. These findings provide genetic evidence for the differential requirements of platelet integrins in thrombus formation and demonstrate that correct integrin function assessment can be achieved with a combination of diagnostic methods.


Sign in / Sign up

Export Citation Format

Share Document