Antithrombotic effects of PAR1 and PAR4 antagonists evaluated under flow and static conditions Kazuya Hosokawa, Tomoko Ohnishi, Naoki Miura, Hisayo Sameshima, Takehiko Koide, Kenichi A. Tanaka, Ikuro Maruyama PII: DOI: Reference:

S0049-3848(13)00512-4 doi: 10.1016/j.thromres.2013.10.037 TR 5271

To appear in:

Thrombosis Research

Received date: Revised date: Accepted date:

19 August 2013 9 October 2013 28 October 2013

Please cite this article as: Hosokawa Kazuya, Ohnishi Tomoko, Miura Naoki, Sameshima Hisayo, Koide Takehiko, Tanaka Kenichi A., Maruyama Ikuro, Antithrombotic effects of PAR1 and PAR4 antagonists evaluated under flow and static conditions, Thrombosis Research (2013), doi: 10.1016/j.thromres.2013.10.037

This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

ACCEPTED MANUSCRIPT Category: Original Article [Platelets and Cell Biology]

PT

Antithrombotic effects of PAR1 and PAR4 antagonists evaluated under flow and static conditions

NU

SC

Koide2, Kenichi A Tanaka4, and Ikuro Maruyama2

RI

Kazuya Hosokawa1,2, Tomoko Ohnishi1, Naoki Miura3, Hisayo Sameshima1, Takehiko

Research Institute, Fujimori Kogyo Co., Ltd., Yokohama, Kanagawa, Japan;

2

Department of System Biology in Thromboregulation, Graduate School of Medical and

MA

1

Dental Sciences, Kagoshima University, Kagoshima, Japan, 3Joint Faculty of Veterinary University,

D

Kagoshima

Kagoshima,

Japan,

and

4

Department

of

TE

Medicine,

Anesthesiology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania,

AC CE P

USA

Running Title: Antithrombotic effects of PAR1 and PAR4 antagonism Word Count: Abstract, 234; Text, 4030

Address Correspondence to: Kazuya Hosokawa, Research Institute, Fujimori Kogyo Co., Ltd., 1-10-1, Sachiura, Kanazawa-ku, Yokohama, Kanagawa, 236-0003, Japan Tel: +81-45-769-0124

1

ACCEPTED MANUSCRIPT Fax: +81-45-769-0131

AC CE P

TE

D

MA

NU

SC

RI

PT

Email: [email protected]

2

ACCEPTED MANUSCRIPT Abstract Introduction: Thrombin-mediated activation of human platelets involves the G-protein-

PT

coupled protease-activated receptors PAR1 and PAR4. Inhibition of PAR1 and/or PAR4

RI

is thought to modulate platelet activation and subsequent procoagulant reactions.

elucidated, particularly under flow conditions.

SC

However, the antithrombotic effects of PAR1 and PAR4 antagonism have not been fully

NU

Materials and Methods: A microchip-based flow chamber system was used to evaluate the influence of SCH79797 (PAR1 antagonist) and YD-3 (PAR4 antagonist) on thrombus

MA

formation mediated by collagen and tissue thromboplastin at shear rates simulating those experienced in small- to medium-sized arteries (600 s-1) and large arteries and small veins

D

(240 s-1).

TE

Results: At a shear rate of 600 s-1, SCH79797 (10 μM) efficiently reduced fibrin-rich

AC CE P

platelet thrombi and significantly delayed occlusion of the flow chamber capillary (1.44 fold of control; P5 nM) is reported to mediate prolonged platelet aggregation [2]. The dual activation of PAR1 and PAR4 collectively promotes platelet morphological

MA

changes, granule secretion, and glycoprotein (GP) IIb/IIIa receptor activation, resulting in stable platelet thrombus formation [1, 2, 8].

TE

D

PAR1 antagonism has potent antithrombotic and antiplatelet effects in animal models of arterial thrombosis [9-12], as well as in acute coronary syndrome (ACS) patients in

AC CE P

phase II clinical trials [13, 14]. A recent phase III trial in ACS patients also demonstrated that combined PAR1 blockade with aspirin and/or clopidogrel reduces the incidence of myocardial infarction; however, the combination therapy increased the risk of fatal bleeding, including intracranial hemorrhage [15]. Therefore, further optimization of treatment strategies based on the assessment of total antiplatelet response to combination therapy may be necessary to achieve clinical benefits from PAR1 antagonism.

Platelet aggregometry is commonly used to assess platelet activation in response to a single exogenous agonist, such as collagen, thrombin, or adenosine 5’-diphosphate (ADP). However, in-vivo thrombus formation is a dynamic and complex process that

5

ACCEPTED MANUSCRIPT involves multiple platelet activation and coagulation pathways under flow conditions [16-18]. In addition, the species specificity of platelet PARs often limits the direct

PT

application of animal models to investigate the roles of PAR1 and PAR4 in

RI

physiological haemostasis, and the influence of PAR antagonism on pathologic thrombus formations. To overcome these limitations, a microchip-based flow chamber

SC

system capable of evaluating fibrin-rich platelet thrombus formation under adjustable

NU

flow conditions is a potential alternative for modeling the pathogenesis of arterial thrombosis after plaque rupture [19].

MA

In the present study, we hypothesized that the antithrombotic mechanisms and efficacies of PAR antagonists can be accurately elucidated by analyzing fibrin-rich platelet

D

thrombus formation patterns under flow conditions. Flow chamber experiments were

TE

conducted under shear conditions mimicking mid- to small-sized arteries (600 s-1) and

PAR1

AC CE P

large arteries and small veins (240 s-1) using human blood samples pre-treated with and

PAR4

antagonists.

Coagulation

assays,

including

whole

blood

viscoelastometry and thrombin generation (TG) assays, were also performed to comparatively evaluate PAR1- and PAR4-mediated platelet procoagulant activity under static conditions.

6

ACCEPTED MANUSCRIPT Materials and Methods Materials

PT

SCH79797 [20] and AR-C66096 [21], specific antagonists of PAR1 and P2Y12 receptor,

RI

respectively, were purchased from Tocris Bioscience (Bristol, United Kingdom). YD-3, a specific antagonist of PAR4, was synthesized as previously reported [22].

SC

Acetylsalicylic acid was purchased from Wako Pure Chemicals (Osaka, Japan). PAR1-

NU

activating peptide (AP), SFLLRN, and PAR4-AP, AYPGKF, were purchased from Verum Diagnostica (Munich, Germany). The microchips used in the flow-chamber

MA

system were manufactured by Richell Corp. (Toyama, Japan). Porcine type I collagen was purchased from Nitta Gelatin, Inc. (Osaka, Japan). Tissue thromboplastin was

TE

D

purchased from Sysmex (Hyogo, Japan). Corn derived trypsin inhibitor (CTI), a specific inhibitor of factor XII, was prepared as reported previously [23]. For the TG assay, the

AC CE P

tissue factor (TF)-based reagent for PRP, and FluCa-reagent, a fluorogenic substrate (ZGly-Gly-Arg-AMC) dissolved in HEPES buffer and calcium chloride, were purchased from Diagnostica Stago (Parsippany, NJ, USA). r-TF was purchased from Mitsubishi Chemical Medience (Tokyo, Japan). All other reagents were obtained from Wako Pure Chemicals (Osaka, Japan).

Blood samples Whole blood samples were collected in plastic tubes containing either 3.2% sodium citrate (Terumo, Tokyo, Japan) or 25 μg/ml hirudin (Verum Diagnostica, Munich, Germany). The subjects had not taken any medication that might have affected platelet function or coagulation in the two weeks preceding the blood draw, and had no history

7

ACCEPTED MANUSCRIPT of coagulation disorders. The study protocol was approved by the institutional ethics committee of Kinki University (Osaka, Japan), and informed written consent was

RI

PT

obtained from all subjects.

Effects of SCH79797 and YD-3 on platelet aggregation

SC

Antiplatelet effects of SCH79797 and YD-3 on platelet aggregation induced by

NU

SFLLRN and AYPGKF, respectively, were confirmed by whole blood platelet aggregometry using the MultiplateTM analyzer (Verum Diagnostica, Munich, Germany)

MA

as directed by the manufacturer. Briefly, 300 μl saline and 300 μl hirudinized blood were pipetted into a single-use cuvette, which was then incubated at 37 °C for 3 min.

TE

D

Either SFLLRN (final, 32 μM) or AYPGKF (final, 662 μM) was then added to the sample to initiate platelet aggregation. Platelet adhesion and aggregation were monitored

AC CE P

for 6 min. The impedance change caused by platelet adhesion and aggregation was plotted against time and the area under the aggregation curve (AUC) to measure the aggregation response, which was quantified in arbitrary aggregation units (AU).

Effects of SCH79797 and YD-3 on thrombus formation measured under flow conditions (i) Microchip-based flow chamber analysis For the analysis of thrombus formation under various shear rates, we used the Total Thrombus-formation Analysis System (T-TAS; Fujimori Kogyo Co., Ltd., Tokyo, Japan), which is a microchip-based flow chamber system equipped with a pneumatic pump, a flow pressure sensor, and a videomicroscope, as described previously [19] (Fig. S1). Blood samples were used for assay within 1 – 3 hours after collection. Briefly,

8

ACCEPTED MANUSCRIPT citrated whole blood (480 l) was mixed with 20l of 0.3 M CaCl2 containing 1.25 mg/ml CTI immediately before the sample was perfused over a microchip coated with

PT

collagen and tissue thromboplastin at flow rates of 4 and 10 μl/min, corresponding to

RI

initial wall shear rates of 240 and 600 s-1, respectively, as estimated by the FLUENT

SC

program (Ansys Co., Ltd., Tokyo, Japan). Flow pressure changes were monitored by the pressure transducer located upstream of the microcapillary during the perfusion

NU

experiments. Thrombus formation and breakdown within the microcapillary result in pressure increases and decreases, respectively. Based on the flow pressure pattern, the

MA

following parameters are used to analyze thrombus formation process. T10 (time to 10 kPa) is the lag time for the flow pressure to increase by 10 kPa from the baseline due to

TE

D

partial occlusion of the capillary. The T10 value represents initial thrombi formation. OT (occlusion time) is the lag time for the flow pressure to increase by 80 kPa from baseline

AC CE P

owing to near complete occlusion of the capillary by thrombi. The OT value reflects the onset, growth, and stability of thrombi inside the capillary. Blood perfusion and flow pressure monitoring are programmed to stop when the flow pressure increases to 80 kPa. In addition to the flow pressure analyses, thrombus formation in the capillary was visually inspected using the built-in light microscope. The two-dimensional area covered by thrombi was analyzed using image analysis software (Zia; Fujimori Kogyo Co., Ltd., Tokyo, Japan), as previously described [19] (Fig. S2). The progression of thrombus formation within the microchip was recorded at 8 and 12 min after the start of blood perfusion.

(ii) Analysis of thrombi by confocal laser scanning microscopy

9

ACCEPTED MANUSCRIPT Thrombi formed on the coated microchip surface were washed three times with phosphate-buffered saline (PBS) and then incubated with FITC-conjugated mouse anti-

PT

human CD41 (platelet GPIIb) IgG (1:5 dilution) for 15 min in the dark. After three

RI

washes with Tris-buffered saline containing 0.1% Triton X-100 (TBST), thrombi were immobilized with OptiLyse C (Immunotech, Marseille, France) for 15 min. After three

SC

washes with TBST, the sample was blocked for 1 h with Block Ace (Yukijirushi, Osaka,

NU

Japan) and then incubated for 30 min in the dark with rabbit anti-human fibrinogen IgG (1:99 dilution) labeled with Alexa 594. To test the immunospecificity of the primary

MA

antibodies, control experiments were conducted with isotype-matched IgG for primary antibodies against GPIIb and fibrinogen. Following antibody staining, the sample was

AC CE P

Oberkochen, Germany).

TE

D

visualized using a LSM700 confocal laser microscope (Carl Zeiss Microscopy Co., Ltd.,

Effects of SCH79797 and YD-3 on thrombus formation measured under static conditions (i) Thromboelastometry measurements The ROTEMTM system (TEM International, Munich, Germany) was used to analyze the clot formation process in whole blood under static conditions. All ROTEMTM assays were performed in the presence of 50 μg/ml CTI to prevent contact phase activation in the analyzer cup, which was held at 37 °C. Briefly, citrated whole blood (300 μl) containing an appropriate concentration of antiplatelet agent(s) was mixed with 20 μl recombinant tissue factor (r-TF) (final concentration, 1 pM) and 25 μl CTI (final concentration, 50 μg/ml) dissolved in 165 mM CaCl2 (final concentration, 12 mM) in the analyzer cup. The following ROTEMTM measurements were performed over a 60-min

10

ACCEPTED MANUSCRIPT period: clotting time (CT; sec), which corresponds to the lag time before clotting; clot formation time (CFT; sec), which reflects the initial rate of clot formation; and

PT

maximum clot firmness (MCF; mm), which is a measure of the maximal tensile strength

SC

(ii) Endogenous thrombin generation measurements

RI

of the clot.

NU

A calibrated automated thrombin generation test using a ThrombinoscopeTM (Thrombinoscope BV, Maastricht, Netherlands) was performed to evaluate the effects of

MA

SCH79797 and YD-3 on the rate and extent of thrombin generation according to the manufacturer’s protocol. All experiments were performed in the presence of CTI (50

TE

D

μg/ml) to prevent thrombin formation mediated by contact phase activation. Platelet-rich plasma (PRP) was obtained by centrifuging citrated whole blood at 125 x g for 10 min at

AC CE P

room temperature, and platelet-poor plasma (PPP) was obtained by further centrifuging the PRP at 1,700 x g for 10 min. PRP was adjusted to a platelet count of 150,000 /μl by dilution with PPP. Thrombin generation was measured in PRP (80 µl) mixed with 20 μl PRP reagent containing r-TF (final concentration, 1 pM). SCH79797 (10 μM), YD-3 (20 μM), aspirin (100 μM), and AR-C66096 (1 μM), or their combinations were incubated with the PRP mixture at 37 °C for 5 min when appropriate. Thrombin generation was triggered by the automated addition of 20 μl FluCa-reagent (final, 416 μM fluorogenic substrate and 16 mM CaCl2). The reaction was monitored at 20-sec intervals for 60 min, and the acquired data were automatically processed using ThrombinoscopeTM software (Version 3.0.0.29; Thrombinoscope BV, Maastricht, Netherlands). The following

11

ACCEPTED MANUSCRIPT parameters were collected: lag time (LT; min), time to peak (TTP; min), peak height

PT

(PH; nM), and endogenous thrombin generation potential (ETP; nM/min).

RI

Statistical analysis

Data are shown as the mean ± standard deviation (SD), unless otherwise indicated.

SC

Differences between groups were tested by one-way repeated ANOVA, followed by

NU

Tukey’s post-hoc test using Prism version 5.02® software (GraphPad Software, CA,

AC CE P

TE

D

MA

USA). A P value of 1500 s) [30, 31]. These results, together with our present findings, suggest that shear-induced

platelet activation supports the growth and stabilization of platelet thrombi, even in the absence of thrombin-mediated platelet activation. The antithrombotic efficacy of PAR1 antagonism observed in the present study was relatively moderate, even when SCH79797 was combined with aspirin and AR-C66096. Notably, the thrombi that formed in the presence of the three antiplatelet agents contained numerous fibrin fibers (Fig. 4). Based on these findings, the inhibitory effect of triple antiplatelet therapy on fibrin deposition is likely limited, although the combined platelet inhibition appears to efficiently reduce platelet activation and deposition onto thrombi.

18

ACCEPTED MANUSCRIPT The thrombi involved in acute coronary lesions consist of activated platelets and large amounts of fibrin due to exposure to collagen and TF after plaque rupture [34, 35]. The

PT

TF associated with ruptured plaques is one of the key factors determining

RI

thrombogenicity [36, 37]. In this regard, the enhanced efficacy of PAR-1 antagonism in combination with aspirin and AR-C66096 on fibrin-rich thrombus formation shown in

SC

the present study is in part concordant with the results of a recent phase III trial in ACS

NU

patients receiving dual antiplatelet therapy [15], which demonstrated that additional PAR1 blockade reduces the incidence of myocardial infarction. However, the treated

MA

ACS patients had increased fatal bleeding complications, including intracranial hemorrhage [15], suggesting that this combination therapy has a relatively narrow

TE

D

therapeutic window with respect to the balance between antithrombotic effect and bleeding risk. Taken together, these findings indicate that PAR1 antagonism may offer an

AC CE P

additional therapeutic option for limiting the growth of intravascular thrombi in high-risk ACS patients [3, 5, 7, 38-40]. However, further optimization of treatment strategies, including other possible drug combinations, dose adjustment, and careful patient selection, are needed to achieve clinical benefits from PAR1 antagonism [41]. In contrast to PAR1 antagonists, our present results suggest that PAR4 antagonism under either flow or static conditions will have minimal antithrombotic effects in humans, even in combination with aspirin and a P2Y12 antagonist. There is a paucity of data on the antithrombotic efficacy of PAR4 antagonism in human blood under flow conditions, although platelet activation and aggregation via activation of PAR4 and downstream intracellular signaling pathways have been well studied [42, 43]. PAR4 activation mediates intracellular signaling via the G-protein subunits Gq and G12/13, but not via the

19

ACCEPTED MANUSCRIPT Gi signaling pathway, whereas PAR1 activation leads to the induction of all of these signaling pathways [43]. Thus, combined platelet inhibition by PAR1 and P2Y12

PT

antagonists efficiently suppresses the Gi signaling pathway. In contrast, treatment with a

RI

PAR4 antagonist in combination with either a PAR1 or P2Y12 antagonist would induce Gi signaling, which may limit the antithrombotic efficacy of combined treatments

SC

involving PAR4 antagonism.

NU

Several limitations of the present study should be noted. First, due to the lack of endothelium in our assay system, our perfusion model may underestimate the

MA

antithrombotic efficacy of PAR1 antagonists, as PAR1 is an important modulator of vascular wall thrombogenicity in endothelial cells [6, 49, 50]. In addition, since CTI does

D

not completely inhibit Factor XIIa, it cannot be denied that some residual activity of FXIIa might

TE

have promoted thrombin generation during perfusion assays. Second, as capillary occlusion is

AC CE P

induced by fibrin-rich platelet thrombi, it was not feasible to separately analyze the effects of antiplatelet agents on fibrin and platelet-deposition from the flow pressure analysis parameters. In addition, the procoagulant activity of subendothelial TF is thought to influence thrombus components, including fibrin and platelets. Thus, additional studies are warranted to optimize the concentration of TF for the evaluation of antiplatelet agents using this flow chamber system. Third, because blood samples were perfused at a constant flow rate, the shear rate inside the microchip increases due to narrowing of the capillaries as a result of thrombi formation. In addition, rectangular capillary was used in the present study, which might have caused the increased wall shear rate and thrombi accumulation in the corner of the capillary. Lastly, our in-vitro results cannot be used to directly infer the clinical efficacy or hemorrhagic risk associated with PAR antagonist therapy. 20

ACCEPTED MANUSCRIPT Current techniques for the assessment of antiplatelet therapy are limited in their ability to simultaneously evaluate multiple agonists involved in platelet adhesion, aggregation, and

PT

procoagulant activity under physiological and pathological conditions [19, 44]. However,

RI

we have demonstrated here that it is possible to assess combined antiplatelet agents that possess distinct inhibitory mechanisms under physiological shear conditions using a shear

SC

rate-adjustable microchip-based flow chamber. Using this approach, we have shown that

NU

the antithrombotic activity of SCH79797-mediated PAR1 antagonism is shear-rate dependent and is also enhanced when combined with aspirin and a P2Y12 inhibitor. An

MA

increasing number of antiplatelet and anticoagulant agents are becoming available for clinical use [45-48], but few monitoring techniques exist for complex antithrombotic

TE

D

regimens. Thus, we intend to examine the suitability of our flow chamber analysis system

risk.

AC CE P

for the clinical evaluation of new antithrombotic therapies and the assessment of bleeding

Conflict of interest statement K Hosokawa, T Ohnishi, and H Sameshima are employees of Fujimori Kogyo Co., Ltd.

21

ACCEPTED MANUSCRIPT References 1

Kahn ML, Zheng YW, Huang W, Bigornia V, Zeng D, Moff S, Farese RV, Jr.,

PT

Tam C, Coughlin SR. A dual thrombin receptor system for platelet activation. Nature

2

RI

1998;394:690-4.

Kahn ML, Nakanishi-Matsui M, Shapiro MJ, Ishihara H, Coughlin SR. Protease-

SC

activated receptors 1 and 4 mediate activation of human platelets by thrombin. J Clin

3

NU

Invest 1999;103:879-87.

Angiolillo DJ, Capodanno D, Goto S. Platelet thrombin receptor antagonism and

4

MA

atherothrombosis. Eur Heart J 2010;31:17-28.

Leonardi S, Tricoci P, Becker RC. Thrombin receptor antagonists for the

2010;70:1771-83.

Leonardi S, Tricoci P, Mahaffey KW. Promises of PAR-1 inhibition in acute

AC CE P

5

TE

D

treatment of atherothrombosis: therapeutic potential of vorapaxar and E-5555. Drugs

coronary syndrome. Curr Cardiol Rep 2012;14:32-9. 6

Martorell L, Martinez-Gonzalez J, Rodriguez C, Gentile M, Calvayrac O,

Badimon L. Thrombin and protease-activated receptors (PARs) in atherothrombosis. Thromb Haemost 2008;99:305-15. 7

Tomasello SD, Angiolillo DJ, Goto S. Inhibiting PAR-1 in the prevention and

treatment of atherothrombotic events. Expert Opin Investig Drugs 2010;19:1557-67. 8

Covic L, Misra M, Badar J, Singh C, Kuliopulos A. Pepducin-based intervention

of thrombin-receptor signaling and systemic platelet activation. Nat Med 2002;8:1161-5. 9

Chintala M, Strony J, Yang B, Kurowski S, Li Q. SCH 602539, a protease-

activated receptor-1 antagonist, inhibits thrombosis alone and in combination with

22

ACCEPTED MANUSCRIPT cangrelor in a Folts model of arterial thrombosis in cynomolgus monkeys. Arterioscler Thromb Vasc Biol 2010;30:2143-9. Dumas M, Nadal-Wollbold F, Gaussem P, Perez M, Mirault T, Letienne R,

PT

10

RI

Bourbon T, Grelac F, Grand BL, Bachelot-Loza C. Antiplatelet and antithrombotic effect of F 16618, a new thrombin protease-activated receptor (PAR1) antagonist. Br J

Letienne R, Leparq-Panissie A, Calmettes Y, Nadal-Wollbold F, Perez M, Le

NU

11

SC

Pharmacol 2012;165:1827-35.

Grand B. Antithrombotic activity of F 16618, a new PAR1 antagonist evaluated in

12

MA

extracorporeal arterio-venous shunt in the rat. Biochem Pharmacol 2010;79:1616-21. Zhang P, Gruber A, Kasuda S, Kimmelstiel C, O'Callaghan K, Cox DH, Bohm

TE

D

A, Baleja JD, Covic L, Kuliopulos A. Suppression of arterial thrombosis without affecting hemostatic parameters with a cell-penetrating PAR1 pepducin. Circulation

13

AC CE P

2012;126:83-91.

Becker RC, Moliterno DJ, Jennings LK, Pieper KS, Pei J, Niederman A, Ziada

KM, Berman G, Strony J, Joseph D, Mahaffey KW, Van de Werf F, Veltri E, Harrington RA. Safety and tolerability of SCH 530348 in patients undergoing non-urgent percutaneous coronary intervention: a randomised, double-blind, placebo-controlled phase II study. Lancet 2009;373:919-28. 14

Goto S, Yamaguchi T, Ikeda Y, Kato K, Yamaguchi H, Jensen P. Safety and

exploratory efficacy of the novel thrombin receptor (PAR-1) antagonist SCH530348 for non-ST-segment elevation acute coronary syndrome. J Atheroscler Thromb 2010;17: 156-64. 15

Tricoci P, Huang Z, Held C, Moliterno DJ, Armstrong PW, Van de Werf F,

23

ACCEPTED MANUSCRIPT White HD, Aylward PE, Wallentin L, Chen E, Lokhnygina Y, Pei J, Leonardi S, Rorick TL, Kilian AM, Jennings LH, Ambrosio G, Bode C, Cequier A, Cornel JH, Diaz R,

PT

Erkan A, Huber K, Hudson MP, Jiang L, Jukema JW, Lewis BS, Lincoff AM,

RI

Montalescot G, Nicolau JC, Ogawa H, Pfisterer M, Prieto JC, Ruzyllo W, Sinnaeve PR, Storey RF, Valgimigli M, Whellan DJ, Widimsky P, Strony J, Harrington RA, Mahaffey

SC

KW. Thrombin-receptor antagonist vorapaxar in acute coronary syndromes. N Engl J

16

NU

Med 2012;366:20-33.

Nesbitt WS, Westein E, Tovar-Lopez FJ, Tolouei E, Mitchell A, Fu J, Carberry

MA

J, Fouras A, Jackson SP. A shear gradient-dependent platelet aggregation mechanism drives thrombus formation. Nat Med 2009;15:665-73. Ruggeri ZM. Platelets in atherothrombosis. Nat Med 2002;8:1227-34.

18

Sambrano GR, Weiss EJ, Zheng YW, Huang W, Coughlin SR. Role of thrombin

TE

D

17

19

AC CE P

signalling in platelets in haemostasis and thrombosis. Nature 2001;413:74-8. Hosokawa K, Ohnishi T, Kondo T, Fukasawa M, Koide T, Maruyama I, Tanaka

KA. A novel automated microchip flow-chamber system to quantitatively evaluate thrombus formation and antithrombotic agents under blood flow conditions. J Thromb Haemost 2011;9:2029-37. 20

Ahn HS, Foster C, Boykow G, Stamford A, Manna M, Graziano M. Inhibition of

cellular action of thrombin by N3-cyclopropyl-7-[[4-(1-methylethyl)phenyl]methyl]-7Hpyrrolo[3, 2-f]quinazoline-1,3-diamine (SCH 79797), a nonpeptide thrombin receptor antagonist. Biochem Pharmacol 2000;60:1425-34. 21

Humphries RG, Tomlinson W, Ingall AH, Cage PA, Leff P. FPL 66096: a novel,

highly potent and selective antagonist at human platelet P2T-purinoceptors. Br J

24

ACCEPTED MANUSCRIPT Pharmacol 1994;113:1057-63. 22

Wu CC, Huang SW, Hwang TL, Kuo SC, Lee FY, Teng CM. YD-3, a novel

Swartz MJ, Mitchell HL, Cox DJ, Reeck GR. Isolation and characterization of

RI

23

PT

inhibitor of protease-induced platelet activation. Br J Pharmacol 2000;130:1289-96.

trypsin inhibitor from opaque-2 corn seeds. J Biol Chem 1977;252:8105-7. Hanson SR, Sakariassen KS. Blood flow and antithrombotic drug effects. Am

SC

24

25

NU

Heart J 1998;135:S132-45.

Mailhac A, Badimon JJ, Fallon JT, Fernandez-Ortiz A, Meyer B, Chesebro JH,

MA

Fuster V, Badimon L. Effect of an eccentric severe stenosis on fibrin(ogen) deposition on severely damaged vessel wall in arterial thrombosis. Relative contribution of fibrin(ogen)

26

TE

D

and platelets. Circulation 1994;90:988-96. Dumas M, Nadal-Wollbold F, Gaussem P, Perez M, Mirault T, Letienne R,

AC CE P

Bourbon T, Grelac F, Le Grand B, Bachelot-Loza C. Antiplatelet and antithrombotic effect of F 16618, a new thrombin proteinase-activated receptor-1 (PAR1) antagonist. Br J Pharmacol 2012;165:1827-35. 27

Nylander S, Mattsson C, Ramstrom S, Lindahl TL. Synergistic action between

inhibition of P2Y12/P2Y1 and P2Y12/thrombin in ADP- and thrombin-induced human platelet activation. Br J Pharmacol 2004;142:1325-31. 28

Shankar H, Garcia A, Prabhakar J, Kim S, Kunapuli SP. P2Y12 receptor-

mediated potentiation of thrombin-induced thromboxane A2 generation in platelets occurs through regulation of Erk1/2 activation. J Thromb Haemost 2006;4:638-47. 29

Trumel C, Payrastre B, Plantavid M, Hechler B, Viala C, Presek P, Martinson

EA, Cazenave JP, Chap H, Gachet C. A key role of adenosine diphosphate in the

25

ACCEPTED MANUSCRIPT irreversible platelet aggregation induced by the PAR1-activating peptide through the late activation of phosphoinositide 3-kinase. Blood 1999;94:4156-65. Hosokawa K, Ohnishi T, Fukasawa M, Kondo T, Sameshima H, Koide T,

PT

30

RI

Tanaka KA, Maruyama I. A microchip flow-chamber system for quantitative assessment of the platelet thrombus formation process. Microvasc Res 2012;83:154-61. Hosokawa K, Ohnishi T, Sameshima H, Miura N, Ito T, Koide T, Maruyama I.

SC

31

NU

Analysing responses to aspirin and clopidogrel by measuring platelet thrombus formation under arterial flow conditions. Thromb Haemost 2013;109:102-11. Leonardi S, Becker RC. PAR-1 Inhibitors: A Novel Class of Antiplatelet Agents

MA

32

for the Treatment of Patients with Atherothrombosis. Handb Exp Pharmacol

TE

33

D

2012;210:239-60.

Lee H, Sturgeon SA, Jackson SP, Hamilton JR. The contribution of thrombin-

AC CE P

induced platelet activation to thrombus growth is diminished under pathological blood shear conditions. Thromb Haemost 2012;107:328-37. 34

Rentrop KP. Thrombi in acute coronary syndromes : revisited and revised.

Circulation 2000;101:1619-26. 35

Toschi V, Gallo R, Lettino M, Fallon JT, Gertz SD, Fernandez-Ortiz A,

Chesebro JH, Badimon L, Nemerson Y, Fuster V, Badimon JJ. Tissue factor modulates the thrombogenicity of human atherosclerotic plaques. Circulation 1997;95:594-9. 36

Ardissino D, Merlini PA, Bauer KA, Bramucci E, Ferrario M, Coppola R,

Fetiveau R, Lucreziotti S, Rosenberg RD, Mannucci PM. Thrombogenic potential of human coronary atherosclerotic plaques. Blood 2001;98:2726-9. 37

Yamashita A, Furukoji E, Marutsuka K, Hatakeyama K, Yamamoto H, Tamura

26

ACCEPTED MANUSCRIPT S, Ikeda Y, Sumiyoshi A, Asada Y. Increased vascular wall thrombogenicity combined with reduced blood flow promotes occlusive thrombus formation in rabbit femoral artery.

Capodanno D, Bhatt DL, Goto S, O'Donoghue ML, Moliterno DJ, Tamburino C,

RI

38

PT

Arterioscler Thromb Vasc Biol 2004;24:2420-4.

Angiolillo DJ. Safety and efficacy of protease-activated receptor-1 antagonists in patients

SC

with coronary artery disease: a meta-analysis of randomized clinical trials. J Thromb

39

Leger AJ, Covic L, Kuliopulos A. Protease-activated receptors in cardiovascular

MA

diseases. Circulation 2006;114:1070-7. 40

Shah R. Protease-activated receptors in cardiovascular health and diseases. Am

TE

D

Heart J 2009;157:253-62. 41

NU

Haemost 2012;10:2006-15.

Lee H, Sturgeon SA, Mountford JK, Jackson SP, Hamilton JR. Safety and

AC CE P

efficacy of targeting platelet proteinase-activated receptors in combination with existing anti-platelet drugs as antithrombotics in mice. Br J Pharmacol 2012;166:2188-97. 42

Holinstat M, Voss B, Bilodeau ML, McLaughlin JN, Cleator J, Hamm HE.

PAR4, but not PAR1, signals human platelet aggregation via Ca2+ mobilization and synergistic P2Y12 receptor activation. J Biol Chem 2006;281:26665-74. 43

Voss B, McLaughlin JN, Holinstat M, Zent R, Hamm HE. PAR1, but not PAR4,

activates human platelets through a Gi/o/phosphoinositide-3 kinase signaling axis. Mol Pharmacol 2007;71:1399-406. 44

Michelson AD. Methods for the measurement of platelet function. Am J Cardiol

2009;103:20A-6A. 45

Angiolillo DJ. The evolution of antiplatelet therapy in the treatment of acute

27

ACCEPTED MANUSCRIPT coronary syndromes: from aspirin to the present day. Drugs 2012;72:2087-116. 46

Bauer KA. Recent progress in anticoagulant therapy: oral direct inhibitors of

Lopes RD. Antiplatelet agents in cardiovascular disease. J Thromb

RI

47

PT

thrombin and factor Xa. J Thromb Haemost 2011;9:12-9.

Thrombolysis 2011;31:306-9.

Patrono C, Andreotti F, Arnesen H, Badimon L, Baigent C, Collet JP, De

SC

48

NU

Caterina R, Gulba D, Huber K, Husted S, Kristensen SD, Morais J, Neumann FJ, Rasmussen LH, Siegbahn A, Steg PG, Storey RF, Van de Werf F, Verheugt F.

MA

Antiplatelet agents for the treatment and prevention of atherothrombosis. Eur Heart J 2011;32:2922-32.

D

Aird WC. Spatial and temporal dynamics of the endothelium. J Thromb

TE

49

Haemost 2005;3:1392-406.

Lee H, Hamilton JR. Physiology, pharmacology, and therapeutic potential of

AC CE P

50

protease-activated receptors in vascular disease. Pharmacol Ther 2012;134:246-59.

28

ACCEPTED MANUSCRIPT Tables Table 1. Effects of SCH79797 and YD-3 on thrombus formation under different shear

RI

PT

conditions

SC

600 s-1 Treatment

OT (min)

7.7 ± 1.7

ASP + AR-C

9.8 ± 2.4*

SCH SCH + ASP + AR-C

14.2 ± 3.6***

OT (min)

9.7 ± 1.9

14.5 ± 2.5

12.1 ± 2.8*

11.0 ± 2.2

16.3 ± 3.2

11.7 ± 3.6***

14.1 ± 4.1***

10.6 ± 2.3

15.9 ± 3.5

16.4 ± 3.9***

13.4 ± 3.4*

19.0 ± 4.2***

8.8 ± 2.0

11.0 ± 2.4

9.9 ± 2.2

14.9 ± 3.0

YD-3 + ASP + AR-C

10.3 ± 2.4**

12.7 ± 2.8**

11.0 ± 1.9*

17.0 ± 3.3

SCH + YD-3

12.2 ± 3.5***

14.6 ± 3.7***

10.5 ± 2.2

17.0 ± 4.2

D

AC CE P

YD-3

9.8 ± 2.0

MA

Control

T10 (min)

TE

NU

T10 (min)

240 s-1

AR-C, AR-C66096 (1 μM); ASP, aspirin (100 μM); SCH, SCH79797 (10 μM). Comparisons between the treatment and control groups were made by one-way repeated ANOVA. * P

Antithrombotic effects of PAR1 and PAR4 antagonists evaluated under flow and static conditions.

Thrombin-mediated activation of human platelets involves the G-protein-coupled protease-activated receptors PAR1 and PAR4. Inhibition of PAR1 and/or P...
1MB Sizes 0 Downloads 0 Views