Skip to main content
Erschienen in: Cardiovascular Drugs and Therapy 1/2020

Open Access 15.02.2020 | Original Article

Ticagrelor Inhibits Toll-Like and Protease-Activated Receptor Mediated Platelet Activation in Acute Coronary Syndromes

verfasst von: Patricia P. Wadowski, Constantin Weikert, Joseph Pultar, Silvia Lee, Beate Eichelberger, Renate Koppensteiner, Irene M. Lang, Simon Panzer, Thomas Gremmel

Erschienen in: Cardiovascular Drugs and Therapy | Ausgabe 1/2020

Abstract

Purpose

Since ticagrelor inhibits the cellular uptake of adenosine, thereby increasing extracellular adenosine concentration and biological activity, we hypothesized that ticagrelor has adenosine-dependent antiplatelet properties. In the current study, we compared the effects of ticagrelor and prasugrel on platelet activation in acute coronary syndrome (ACS).

Methods

Platelet surface expression of P-selectin and activated glycoprotein (GP) IIb/IIIa in response to adenosine diphosphate (ADP), the toll-like receptor (TLR)-1/2 agonist Pam3CSK4, the TLR-4 agonist lipopolysaccharide (LPS), the protease-activated receptor (PAR)-1 agonist SFLLRN, and the PAR-4 agonist AYPGKF were measured by flow cytometry in blood from 80 ticagrelor- and 80 prasugrel-treated ACS patients on day 3 after percutaneous coronary intervention. Residual platelet aggregation to arachidonic acid (AA) and ADP were assessed by multiple electrode aggregometry and light transmission aggregometry.

Results

ADP-induced platelet activation and aggregation, and AA-induced platelet aggregation were similar in patients on ticagrelor and prasugrel, respectively (all p ≥ 0.3). Further, LPS-induced platelet surface expression of P-selectin and activated GPIIb/IIIa did not differ significantly between ticagrelor- and prasugrel-treated patients (both p > 0.4). In contrast, Pam3CSK4-induced platelet surface expression of P-selectin and activated GPIIb/IIIa were significantly lower in ticagrelor-treated patients (both p ≤ 0.005). Moreover, SFLLRN-induced platelet surface expression of P-selectin and activated GPIIb/IIIa were significantly less pronounced in patients on ticagrelor therapy compared to prasugrel-treated patients (both p < 0.03). Finally, PAR-4 mediated platelet activation as assessed by platelet surface expression of activated GPIIb/IIIa following stimulation with AYPGKF was significantly lower in patients receiving ticagrelor (p = 0.02).

Conclusion

Ticagrelor inhibits TLR-1/2 and PAR mediated platelet activation in ACS patients more strongly than prasugrel.
Hinweise

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
ACS
Acute coronary syndrome
ADP
Adenosine diphosphate
DAPT
Dual antiplatelet therapy
GP
Glycoprotein
LPS
Lipopolysaccharide
LTA
Light transmission aggregometry
MEA
Multiple electrode aggregometry
MFI
Median fluorescence intensity
PAR
Protease-activated receptor
PCI
Percutaneous coronary intervention
TLR
Toll-like receptor

Introduction

Patients suffering acute coronary syndromes (ACS) are routinely treated with dual antiplatelet therapy (DAPT) consisting of aspirin and an adenosine diphosphate (ADP) P2Y12 antagonist for 12 months to prevent detrimental platelet activation and subsequent atherothrombotic events [1]. According to the guidelines of the American College of Cardiology/American Heart Association and the European Society of Cardiology [2, 3], the newer P2Y12 receptor antagonists ticagrelor and prasugrel should be preferred over clopidogrel in ACS patients undergoing percutaneous coronary intervention (PCI) with stent implantation due to their more favorable effects on 1-year ischemic outcomes in large randomized clinical trials [4, 5]. The latter are most likely due to faster, stronger, and more consistent inhibition of ADP-induced platelet activation by ticagrelor and prasugrel compared to clopidogrel [410]. Indeed, previous studies revealed a similar inhibitory effect of ticagrelor and prasugrel on residual ADP-induced platelet aggregation [11, 12]. However, human platelets can be activated by a myriad of agonists via different receptors [7, 13]. Among these are toll-like receptors (TLR) and protease-activated receptors (PAR) which are not targeted by standard DAPT but might contribute to platelet hyperreactivity as well as to the development and progression of atherosclerotic cardiovascular disease [1416].
TLR are part of innate immunity recognizing pathogen-associated molecular patterns of microbial origin and damage-associated molecular patterns of injured host cells [17, 18]. However, the role of TLR extends far beyond: TLR in the myocardium are involved in pro-inflammatory cytokine expression, infarct size regulation, and influence cardiac remodeling [14, 17, 19]. In addition, TLR seem to be involved in the modulation of ischemia-reperfusion injury in ACS [20]. TLR-1, TLR-2, TLR-3, TLR-4, TLR-6, TLR-7, and TLR-9 were found on and in human platelets, respectively [14], and may represent a link between thrombosis and inflammation [16]. Acute myocardial infarction leads to the upregulation of platelet TLR-1 and TLR-4 [21, 22]. Moreover, it has been shown that platelets from ACS patients on DAPT can be directly and dose-dependently activated by Pam3CSK4, a synthetic agonist of TLR-1/2, whereas platelet activation via TLR-4 in these patients occurred only in response to the highest concentration of lipopolysaccharide (LPS) [22]. Further, the TLR-2/6 agonist FSL-1 was not able to induce platelet activation at any concentration in ACS patients [22]. Hence, in particular, the TLR-1/2 pathway appears to be functional in ACS and may contribute to platelet activation despite state-of-the-art DAPT. The response of human platelets to thrombin is partially mediated by the autocrine/paracrine effect of secreted ADP after thrombin-induced platelet activation [23]. Accordingly, in vitro experiments in blood from healthy volunteers showed that inhibition of the P2Y12 but not P2Y1 receptor reduced thrombin-induced platelet activation [24]. In line with these findings, Whitley et al. described an effect of ticagrelor on PAR-4 mediated platelet aggregation [25]. Moreover, as shown by Kälvegren et al., TLR-1/2 mediated platelet activation can at least partially be affected by ADP receptor antagonists [26]. Further, platelet inhibition by ticagrelor has been associated with less in vitro platelet-monocyte complex formation and a modulation of TLR-2- and TLR-4 mediated cytokine response of peripheral blood mononuclear cells [27].
PAR-1 and PAR-4 enable human platelet activation by the serine protease thrombin, one of the strongest platelet agonists [7, 28, 29]. While PAR-1 is sensitive to low levels of thrombin, PAR-4 triggers platelet activation and aggregation only at high thrombin concentrations, and cleavage of PAR-4 by thrombin occurs 20- to 70-fold slower than cleavage of PAR-1 [30]. Recently, we have shown that platelet responsiveness to PAR-1 and PAR-4 stimulation is preserved in the majority of clopidogrel-treated patients and in about 20% of patients receiving prasugrel following angioplasty with stent implantation [31, 32].
Due to their distinct molecular structures, the cyclopentyl-triazolo-pyrimidine ticagrelor and the thienopyridine prasugrel may differently affect platelet activation which may in turn explain clinical differences between ticagrelor and prasugrel in large randomized trials [4, 5, 8, 9, 33]. Since ticagrelor inhibits the cellular uptake of adenosine, thereby increasing extracellular adenosine concentration and biological activity [34, 35], we hypothesized that ticagrelor has adenosine-dependent antiplatelet properties [36]. In the current study, we therefore compared the effects of ticagrelor and prasugrel on platelet activation in 160 ACS patients undergoing PCI and stenting. Platelet surface expression of P-selectin and activated GPIIb/IIIa in response to ADP, Pam3CSK4, LPS, SFLLRN, and AYPGKF were determined to assess agonist-induced platelet activation. ADP was chosen as agonist to document the response to platelet inhibition therapy, as all patients received an ADP P2Y12 antagonist. Platelet response to Pam3CSK4 was measured because it has been shown previously that, in particular, the TLR-1/2 pathway is functional in ACS in the presence of DAPT [22]. LPS-induced platelet surface expression of P-selectin and activated GPIIb/IIIa was assessed to investigate platelet activation via TLR-4 [37]. SFLLRN- and AYPGKF-induced platelet activation was determined to evaluate the platelet response to PAR-1 and PAR-4 stimulation, respectively [38, 39].

Materials and Methods

Study Population

The study had an observational, non-randomized open label design. The study population consisted of 160 ACS patients on daily aspirin (100 mg/day), and either prasugrel (10 mg/day, n = 80), or ticagrelor (180 mg/day, n = 80) therapy. All patients were Caucasians from the Vienna urban area and were recruited within 2 years for the study. Blood sampling was performed on day 3 after acute successful PCI with stent implantation after an overnight fast, 12 h after the last intake of the respective P2Y12 antagonist in all patients.
Exclusion criteria were a known aspirin, prasugrel, or ticagrelor intolerance (allergic reactions, gastrointestinal bleeding); a therapy with vitamin K antagonists (warfarin, phenprocoumon, acenocoumarol) or direct oral anticoagulants (rivaroxaban, apixaban, dabigatran, edoxaban); treatment with ticlopidine, dipyridamol, or nonsteroidal anti-inflammatory drugs; a family or personal history of bleeding disorders; malignant myeloproliferative disorders or heparin-induced thrombocytopenia; severe hepatic failure; known qualitative defects in platelet function; a major surgical procedure within 1 week before enrolment; a platelet count < 100.000 or > 450.000/μl; and a hematocrit < 30%.
The study protocol was approved by the Ethics Committee of the Medical University of Vienna (Ethics commitee number: 1940/2013) in accordance with the Declaration of Helsinki and its later amendments, and written informed consent was obtained from all study participants.

Blood Sampling

All laboratory personnel were blinded to study treatment. Blood was drawn by aseptic venipuncture from an antecubital vein using a 21-gauge butterfly needle (0.8 × 19 mm; Greiner Bio-One, Kremsmünster, Austria) on day 3 after PCI. To avoid procedural deviations, all blood samples were taken by the same physician applying a light tourniquet, which was immediately released and the samples were mixed adequately by gently inverting the tubes. After the initial 3 ml of blood had been discarded to reduce peri-procedural platelet activation, blood was drawn into hirudin-coated tubes (Roche Diagnostics, Mannheim, Germany) for multiple electrode aggregometry (MEA) and into 3.8% sodium citrate Vacuette tubes (Greiner Bio-One; 9 parts of whole blood, 1 part of sodium citrate 0.129 M/L) for evaluations by flow cytometry and light transmission aggregometry (LTA).

Determination of P-Selectin Expression and Glycoprotein (GP) IIb/IIIa Activation

The expression of P-selectin and the binding of the monoclonal antibody PAC-1 to activated glycoprotein (GP) IIb/IIIa were determined in citrate-anticoagulated blood, as previously published [40, 41]. In brief, whole blood was diluted in phosphate-buffered saline to obtain 20 × 103/μL platelets in 20 μL, and incubated for 10 min with the platelet-specific monoclonal antibody anti-CD42b (clone HIP1, allophycocyanin labelled; Becton Dickinson (BD), San Jose, CA, USA), without agonists, and after in vitro exposure to suboptimal concentrations of ADP (final concentration 1 μM; Roche Diagnostics GmbH, Mannheim, Germany), the TLR-1/2 agonist Pam3CSK4 (final concentration 8.9 μg/mL; InvivoGen, San Diego, USA), the TLR-4 agonist lipopolysaccharide (LPS; final concentration 1429 μg/mL; InvivoGen, San Diego, USA), the PAR-1 agonist SFLLRN (final concentration: 14.25 μM; Roche Diagnostics GmbH, Mannheim, Germany), or the PAR-4 agonist AYPGKF (final concentration 714 μM; Roche Diagnostics GmbH, Mannheim, Germany), each 10 μL for 10 min. The concentrations of all agonists were determined in previous titration experiments with increasing dosages of each agonist in 10 healthy controls. The selected concentrations of agonists induced about 60–70% of the maximal achievable increase in median fluorescence intensity (MFI) in healthy controls. Samples were then incubated for another 10 min with a mixture of antibodies against activated GPIIb/IIIa (the monoclonal antibody PAC-1-fluorescein (BD)) and P-selectin (anti-CD62p-phycoerythrin, clone CLB-Thromb6; Immuno-tech, Beckman Coulter, Fullerton, CA, USA). Isotype matched control antibodies (BD) were used for the determination of non-specific binding. After 15 min of incubation in the dark, the reaction was stopped by adding 500 μl phosphate-buffered saline (PBS) and samples were acquired immediately on a FACSCanto II flow cytometer (BD). At acquisition, the platelet population was identified by its characteristics in the forward scatter versus side scatter plot (Fig. 1a). A total of 10,000 events were acquired within this gate. This population was further identified by platelets stained with the platelet-specific monoclonal antibody anti-CD42b versus side scatter (Fig. 1b). Binding of the antibodies against activated GPIIb/IIIa and P-selectin was determined in histograms for PAC-1 and P-selectin, respectively (Fig. 1c, d). Cytometer Setup and Tracking beads (BD), which consist of FITC, PE, PERCP-CY5.5, PE-CY7, APC, APC-H7, V450, and V500-C labelled beads, were used for daily calibration of the cytometer applying the Diva software. The MFI based on all events was used for statistical calculations.

Multiple Electrode Aggregometry

Whole blood impedance aggregometry was performed with the Multiplate analyzer (Roche Diagnostics, Mannheim, Germany) as previously described [42]. One Multiplate test cell contains 2 independent sensor units and 1 unit consists of 2 silver-coated highly conductive copper wires with a length of 3.2 mm. After dilution (1:2 with 0.9% NaCl solution) of hirudin-anticoagulated whole blood and stirring in the test cuvettes for 3 min at 37 °C, ADP (6.4 μM, Roche Diagnostics, Mannheim, Germany) or arachidonic acid (AA; final concentration of 0.5 mM; Roche Diagnostics, Mannheim, Germany) was added and aggregation was continuously recorded for 6 min. The adhesion of activated platelets to the electrodes led to an increase of impedance, which was detected for each sensor unit separately and transformed to aggregation units (AU) that were plotted against time. The AU at 6 min were used for calculations. One AU corresponds to 10 AU*min (area under the curve of AU).

Light Transmission Aggregometry

Light transmission aggregometry (LTA) was performed on a PAP-8E aggregometer (Bio-Data, Horsham, PA USA) as previously described [42, 43]. Citrate-anticoagulated whole blood was allowed to “rest” in a tilt position at room temperature for 20 min before centrifugation. Blood tubes were centrifuged at 150×g for 10 min (min) at room temperature to acquire platelet-rich plasma (PRP). To obtain platelet-poor plasma (PPP), the remaining specimen were re-centrifugated at 2.000×g for 10 min. Platelet counts were not adjusted as the median platelet count was 198 G/L (range 166–230 G/L) [44]. The optical density of PPP was set as 100% aggregation. Platelet aggregation was initiated by addition of ADP (5 μM) or AA (1600 μM) as agonists to PRP. Optical density changes were recorded photoelectrically for 10 min as platelets began to aggregate to obtain maximal aggregation %. Maximal aggregation % was automatically calculated by the PAP-8E aggregometer by comparing the increase of light transmission through platelet-rich plasma after addition of an agonist to the baseline optical density that was set with platelet-poor plasma and considered as 100% platelet aggregation.

Statistical Analysis

Statistical analysis was performed using the Statistical Package for Social Sciences (IBM SPSS version 25, Armonk, New York, USA). Median and interquartile range of continuous variables are shown. Categorical variables are given as number (%). We performed Mann Whitney U tests to detect differences in continuous variables. The chi-square test was used to assess differences in categorical variables, respectively. Two-sided p values < 0.05 were considered statistically significant.

Results

Clinical and laboratory characteristics of ticagrelor (n = 80)- and prasugrel (n = 80)-treated patients are given in Table 1. The 2 groups were well-matched regarding age, sex, body mass index, hemoglobin, platelet count, renal function, comorbidities, and medication (Table 1; all p > 0.05). Moreover, white blood cell count (WBC), high-sensitivity C-reactive protein (hsCRP), and interleukin (IL)-6 as markers of inflammation were similar in patients with ticagrelor and prasugrel therapy, respectively (Table 1; all p > 0.05).
Table 1
Clinical and laboratory characteristics of ticagrelor- and prasugrel-treated patients
Characteristics
Ticagrelor (n = 80)
Prasugrel (n = 80)
p
Demographics
  Age, years
60 (51–70)
58 (51–66)
0.1
  Male sex, n (%)
61 (76)
65 (81)
0.4
  BMI, kg/m2
28 (25–31)
28 (25–31)
0.8
Medical history
  Previous MI, n (%)
13 (16)
14 (18)
0.9
  Previous TIA/stroke, n (%)
2 (3)
3 (4)
0.7
  Hypertension, n (%)
56 (70)
54 (68)
0.7
  Hyperlipidemia, n (%)
58 (73)
61 (76)
0.8
  Diabetes mellitus, n (%)
15 (19)
13 (16)
0.1
  -Type I, n (%)
1 (1)
0 (0)
0.2
  -Type II, n (%)
14 (18)
13 (16)
0.2
  Smoking, n (%)
39 (49)
47 (59)
0.1
  Stent implantation, n (%)
80 (100)
80 (100)
1
  Number of stents/patient
1 (1–2)
1 (1–2)
0.3
Laboratory data
  Serum creatinine, μmol/L
88 (72–106)
83 (74–95)
0.1
  Platelet count, G/L
226 (187–266)
229 (194–253)
0.9
  High-sensitivity CRP, mg/L
14 (6–36)
17 (7–33)
0.5
  Hemoglobin, mmol/L
8.4 (7.9–9.1)
8.6 (8.1–9)
0.7
  WBC, G/L
9.3 (6.9–10.8)
8.7 (7.6–10.4)
1
Medication
  Statins, n (%)
79 (99)
79 (99)
1
  Beta blockers, n (%)
78 (98)
77 (96)
0.7
  ACE inhibitors, n (%)
61 (76)
65 (81)
0.4
  Angiotensin receptor blockers, n (%)
19 (24)
12 (15)
0.2
  Calcium channel blockers, n (%)
10 (13)
7 (9)
0.4
Continuous data are shown as median (interquartile range). Dichotomous data are shown as n (%)
BMI, body mass index; ACE, angiotensin converting enzyme; CRP, C-reactive protein; MI, myocardial infarction; TIA, transient ischemic attack; WBC, white blood cell count
Residual ADP-induced platelet activation as assessed by platelet surface expression of P-selectin and activated GPIIb/IIIa following in vitro exposure to ADP was very similar in ticagrelor- and prasugrel-treated patients (P-selectin ADP: 21 MFI [8–37 MFI] vs. 17 MFI [5–48 MFI], p = 0.9; activated GPIIb/IIIa ADP: 42 MFI [19–64 MFI] vs. 39 MFI [15–73], p = 0.9; Fig. 2c, d). Likewise, residual platelet aggregation in response to ADP and AA by multiple electrode aggregometry (MEA) and LTA did not differ significantly between patients receiving ticagrelor and those treated with prasugrel (Table 2; all p ≥ 0.3). Based on the consensus cut-off value of AU ≥ 47 by MEA [45], only 2 prasugrel-treated patients (2.5%) and no patient on ticagrelor therapy had high on-treatment residual platelet reactivity to ADP (HRPR ADP).
Table 2
Adenosine diphosphate (ADP)- and arachidonic acid (AA)-induced platelet aggregation by multiple electrode aggregometry (MEA) and light transmission aggregometry (LTA) in ticagrelor- and prasugrel-treated patients
 
Ticagrelor (n = 80)
Prasugrel (n = 80)
p
MEA 6.4 μM ADP, AU
20 (15–25)
19 (15–23)
0.4
LTA 5 μM ADP, %
27 (20–33)
24 (18–33)
0.4
MEA 0.5 mM AA, AU
16 (11–20)
16 (11–21)
1
LTA 1.6 mM AA, %
3 (2–6)
2 (1–4)
0.3
Continuous data are shown as median (interquartile range)
AA, arachidonic acid; ADP, adenosine diphosphate; AU, aggregation units; MEA, multiple electrode aggregometry; LTA, light transmission aggregometry
TLR-1/2 mediated platelet activation as assessed by platelet surface expression of P-selectin and activated GPIIb/IIIa after stimulation with Pam3CSK4 in vitro was significantly lower in patients on ticagrelor therapy compared to those receiving prasugrel (Fig. 2e, f; Table 3; both p ≤ 0.005). In contrast, LPS-induced platelet surface expression of P-selectin and activated GPIIb/IIIa were similar between patients on ticagrelor and prasugrel, respectively (Fig. 2g, h, Table 3; both p > 0.4).
Table 3
Platelet surface expression of P-selectin and activated glycoprotein (GP) IIb/IIIa in response to Pam3CSK4, lipopolysaccharide (LPS), SFLLRN, and AYPGKF in ticagrelor- and prasugrel-treated patients
 
Ticagrelor (n = 80)
Prasugrel (n = 80)
p
P- selectin expression, MFI
  Without agonist
0 (0–0.2)
0 (0–0.1)
0.4
  1 μM ADP
21 (8–37)
17 (5–48)
0.9
  8.9 μg/mL Pam3CSK4
51 (24–177)
116 (42–496)
0.002
  1429 μg/mL LPS
30 (9–79)
28 (9–72)
0.5
  14.25 μM SFLLRN
2591 (1670–3387)
3359 (2140–4170)
0.02
  714 μM AYPGKF
188 (89–607)
259 (102–796)
0.2
Activated GPIIb/IIIa, MFI
  Without agonist
0 (0–0.4)
0 (0–8)
0.04
  1 μM ADP
42 (19–64)
39 (15–73)
0.9
  8.9 μg/mL Pam3CSK4
26 (11–54)
39 (17–107)
0.005
  1429 μg/mL LPS
73 (37–146)
71 (28–127)
0.8
  14.25 μM SFLLRN
27 (16–49)
38 (19–72)
0.03
  714 μM AYPGKF
25 (10–52)
37 (21–72)
0.02
Continuous data are shown as median (interquartile range)
ADP, adenosine diphosphate; MFI, median fluorescence intensity; LPS, lipopolysaccharide
PAR-1 mediated platelet activation as assessed by platelet surface expression of P-selectin and activated GPIIb/IIIa in response to SFLLRN in vitro was significantly less pronounced in ticagrelor-treated patients compared to patients on prasugrel therapy (Fig. 2i, j, Table 3; both p < 0.03). Finally, PAR-4 mediated platelet activation as assessed by platelet surface expression of activated GPIIb/IIIa following stimulation with AYPGKF in vitro was significantly lower in patients receiving ticagrelor (Fig. 2l, Table 3; p = 0.02). AYPGKF-induced platelet surface P-selectin expression was numerically lower in patients on ticagrelor therapy compared to prasugrel-treated patients without reaching statistical significance (Fig. 2k, Table 3; p = 0.2).

Discussion

Our study is the first to show that ticagrelor exerts a more pronounced antiplatelet effect on TLR-1/2 and PAR mediated platelet activation than prasugrel in ACS patients on DAPT following PCI and stenting. In contrast, ADP-induced platelet activation and aggregation, AA-induced platelet aggregation, and platelet activation via TLR-4 were similar in ticagrelor- and prasugrel-treated patients, respectively.
Previous studies have revealed that both, the PAR-1 and the PAR-4 pathway, remain active in many patients on DAPT [31, 32]. Platelet aggregation via PAR-1 and PAR-4 could even be observed in patients on prasugrel and ticagrelor, as reported recently [46].
P-selectin is stored in the α-granules of resting platelets [7]. Upon platelet activation, P-selectin is translocated to the platelet surface where it binds to its counterreceptor P-selectin glycoprotein ligand-1 on leukocytes in order to form leukocyte-platelet aggregates [47]. GPIIb/IIIa is already present on the surface of resting platelets but only converted to its active conformation following platelet activation [48]. Subsequently, activated GPIIb/IIIa serves as fibrinogen receptor thereby facilitating the interaction of platelets with one another and with other blood cells [7, 4951]. Numerous studies have shown that both platelet-bound P-selectin and activated GPIIb/IIIa are very sensitive markers for human platelet activation [40, 52]. Furthermore, high levels of platelet surface P-selectin and activated GPIIb/IIIa in response to PAR-1 stimulation have recently been associated with an increased risk of ischemic outcomes following peripheral angioplasty and stenting [40]. Platelet aggregation in response to AA and ADP was measured in all patients by MEA and LTA. While MEA is a fast and highly standardized near point-of-care test [53], LTA was the first widely available method to assess on-treatment platelet aggregation and is still considered the historical gold standard of platelet function testing [54]. The results of both test systems have been linked to clinical outcomes in patients undergoing PCI and stenting [45, 5557].
While ADP-induced platelet activation was very low in all patients of the study population, PAR- and TLR-mediated platelet activation was preserved in many patients. It remains to be determined if high PAR- or TLR- mediated platelet activation is associated with adverse clinical outcomes in ACS patients treated with the potent ADP P2Y12 antagonists. In all experiments, platelet surface expression of P-selectin was higher than that of activated GPIIb/IIIa, irrespective of the type of P2Y12 antagonist. This may point towards a need for higher agonist concentrations for GPIIb/IIIa activation as compared to the surface expression of P-selectin.
Although ADP-induced platelet activation and aggregation were very similar and consistently low in ticagrelor- and prasugrel-treated patients, we cannot completely exclude that a stronger inhibitory effect of ticagrelor on the P2Y12 receptor is responsible for the observed differences regarding TLR-1/2 and PAR-mediated platelet activation, in particular, since previous studies suggested stronger P2Y12 inhibition with ticagrelor compared to prasugrel [58, 59]. Given the role of ADP and the P2Y12 receptor for other pathways of platelet activation, stronger P2Y12 inhibition may explain the observed differences even if the ADP tests are similar as the ADP concentration in the assays may not accurately reflect the local ADP concentration at the platelet surface.
We further observed similar AA-induced platelet aggregation in patients receiving ticagrelor and prasugrel, respectively. Therefore, differences in the response to aspirin therapy did not account for the more pronounced inhibition of TLR-1/2 and PAR-mediated platelet activation by ticagrelor. Finally, patient characteristics including markers of inflammation, i.e., WBC, hsCRP and IL-6, were well-balanced between ticagrelor- and prasugrel-treated patients in our study.
Recently, Hally et al. investigated whether aspirin monotherapy or DAPT with aspirin and ticagrelor reduce platelet activation via TLR in 10 healthy individuals [60]. While aspirin alone did not affect platelet surface expression of P-selectin and activated GPIIb/IIIa in response to Pam3CSK4 and LPS, concomitant therapy with ticagrelor resulted in a modest inhibition of TLR-mediated platelet activation [60]. Furthermore, several studies found TLR and PAR signaling to be involved in the splicing of mRNA encoding for the inflammatory cytokine IL-1ß in platelets [6163]. The observation by Jiang et al. that patients on ticagrelor therapy had significantly lower IL-1ß levels than clopidogrel-treated patients 1 year post ACS may therefore point towards inhibition of TLR and PAR mediated platelet activation by ticagrelor [64]. Together with our results, the above-mentioned findings suggest that ticagrelor at least to some extent affects platelet activation via TLR and PAR. However, whether inhibition of TLR and PAR-mediated platelet activation by ticagrelor also translates into beneficial clinical outcomes remains to be established.
Schoergenhofer et al. performed a double-blind, randomized, crossover trial with a minimum wash-out period of 6 weeks in 16 subjects to investigate whether prasugrel reduces LPS-induced coagulation activation compared to placebo [65]. They described a strong coagulation activation by LPS including increased platelet surface P-selectin expression, which was not inhibited by prasugrel, and concluded that potent P2Y12 inhibition does not affect coagulation activation by LPS [65]. Our results of similar and rather high levels of LPS-induced platelet surface P-selectin and activated GPIIb/IIIa in the entire study cohort suggest that (1) their findings also apply to ACS patients and (2) even ticagrelor may be unable to effectively reduce LPS-induced platelet activation in ACS.
In vivo, ticagrelor inhibits the adenosine transporter ENT1 (type 1 equilibrative nucleoside transporter) and thereby the cellular uptake of adenosine which results in higher plasma levels/ extracellular concentrations of adenosine and increased biological activity of adenosine [66]. Indeed, Nylander et al. reported that ticagrelor can augment adenosine-mediated inhibition of collagen-induced human platelet aggregation in addition to P2Y12 antagonism in whole blood samples from 50 healthy individuals, an effect that was not seen with the active metabolite of prasugrel in their study [36].
While prasugrel did not reduce all-cause mortality compared to clopidogrel in 13,608 ACS patients in the TRITON TIMI 38 trial [4], ticagrelor was associated with a significantly lower all-cause mortality compared to clopidogrel in 18,624 ACS patients in the PLATO study [5]. Several potential explanations for the survival benefit with ticagrelor have been discussed since publication of the PLATO results [34, 36, 67]. Given the important role of TLR and PAR in cardiovascular disease [14, 68, 69], one may speculate that the stronger inhibitory effects of ticagrelor on TLR-1/2 and PAR mediated platelet activation compared to prasugrel may at least in part contribute to the beneficial impact of ticagrelor on all-cause mortality. Indeed, in a previous study, we were able to show that high PAR-1 mediated platelet activation is a predictor of target lesion restenosis and atherothrombotic events in patients undergoing infrainguinal angioplasty and stenting [40]. However, data showing an association of PAR or TLR-mediated platelet activation with adverse outcomes in ACS patients are missing, so far. The latter is a prerequisite for future studies on therapeutic strategies in ACS patients with high platelet activation via TLR or PAR.
Selective inhibition of PAR-1 with vorapaxar on top of DAPT with aspirin and clopidogrel decreased the composite of myocardial infarction, stroke, and cardiovascular death in ACS patients in the TRACER trial, but significantly increased the risk of bleeding including intracranial hemorrhage [70]. These findings suggest that complete PAR-1 inhibition may too extensively impair cellular hemostasis, at least in conjunction with aspirin and a P2Y12 antagonist. Consequently, at the moment, DAPT with aspirin and a P2Y12 receptor antagonist remains the preferred antiplatelet regimen in ACS. New antiplatelet agents selectively targeting TLR or PAR-4 may become treatment options in ACS patients in the future [1, 71, 72].
A limitation of our study is that it was not powered for and intended to provide clinical outcome data. Moreover, the choice of the P2Y12 antagonist was made by the treating physician which may have led to patient selection bias. However, as shown in Table 1, the 2 study groups were well-matched regarding all relevant patient characteristics including comorbidities, laboratory markers, and concomitant medication. Accordingly, it is unlikely that differences in the patient characteristics had a significant impact on the observed results. Further, we only assessed on-treatment platelet activation in our study population, and therefore cannot comment on differences in platelet activation in response to the agonists in patients without antiplatelet therapy.

Conclusions

Ticagrelor inhibits TLR-1/2 and PAR-mediated platelet activation in ACS patients more strongly than prasugrel. Underlying mechanisms and potential clinical implications of our observations need to be addressed in future trials.

Compliance with Ethical Standards

Conflict of Interest

The authors declare that they have no conflicts of interest. The project was part of the diploma thesis of cand. med. Constantin Weikert.

Ethical Approval

All procedures performed were approved by the Ethics Committee of the Medical University of Vienna and are in accordance with the 1964 Helsinki Declaration and its later amendments.
Informed consent was obtained from all individual participants included in the study.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Gremmel T, Michelson AD, Frelinger AL III, Bhatt DL. Novel aspects of antiplatelet therapy in cardiovascular disease. Res Pract Thromb Haemost. 2018;2:439–49.CrossRefPubMedPubMedCentral Gremmel T, Michelson AD, Frelinger AL III, Bhatt DL. Novel aspects of antiplatelet therapy in cardiovascular disease. Res Pract Thromb Haemost. 2018;2:439–49.CrossRefPubMedPubMedCentral
2.
Zurück zum Zitat Valgimigli M, Bueno H, Byrne RA, Collet JP, Costa F, Jeppsson A, et al. 2017 ESC focused update on dual antiplatelet therapy in coronary artery disease developed in collaboration with EACTS: the task force for dual antiplatelet therapy in coronary artery disease of the European Society of Cardiology (ESC) and of the European Association for Cardio-Thoracic Surgery (EACTS). Eur Heart J. 2018;39:213–60.CrossRefPubMed Valgimigli M, Bueno H, Byrne RA, Collet JP, Costa F, Jeppsson A, et al. 2017 ESC focused update on dual antiplatelet therapy in coronary artery disease developed in collaboration with EACTS: the task force for dual antiplatelet therapy in coronary artery disease of the European Society of Cardiology (ESC) and of the European Association for Cardio-Thoracic Surgery (EACTS). Eur Heart J. 2018;39:213–60.CrossRefPubMed
3.
Zurück zum Zitat Levine GN, Bates ER, Bittl JA, Brindis RG, Fihn SD, Fleisher LA, et al. 2016 ACC/AHA guideline focused update on duration of dual antiplatelet therapy in patients with coronary artery disease: a report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. J Am Coll Cardiol. 2016;68:1082–115.CrossRefPubMed Levine GN, Bates ER, Bittl JA, Brindis RG, Fihn SD, Fleisher LA, et al. 2016 ACC/AHA guideline focused update on duration of dual antiplatelet therapy in patients with coronary artery disease: a report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. J Am Coll Cardiol. 2016;68:1082–115.CrossRefPubMed
4.
Zurück zum Zitat Wiviott SD, Braunwald E, McCabe CH, Montalescot G, Ruzyllo W, Gottlieb S, et al. Prasugrel versus clopidogrel in patients with acute coronary syndromes. N Engl J Med. 2007;357:2001–15.CrossRefPubMed Wiviott SD, Braunwald E, McCabe CH, Montalescot G, Ruzyllo W, Gottlieb S, et al. Prasugrel versus clopidogrel in patients with acute coronary syndromes. N Engl J Med. 2007;357:2001–15.CrossRefPubMed
5.
Zurück zum Zitat Wallentin L, Becker RC, Budaj A, Cannon CP, Emanuelsson H, Held C, et al. Ticagrelor versus clopidogrel in patients with acute coronary syndromes. N Engl J Med. 2009;361:1045–57.CrossRefPubMed Wallentin L, Becker RC, Budaj A, Cannon CP, Emanuelsson H, Held C, et al. Ticagrelor versus clopidogrel in patients with acute coronary syndromes. N Engl J Med. 2009;361:1045–57.CrossRefPubMed
6.
Zurück zum Zitat Gremmel T, Eslam RB, Koppensteiner R, Lang IM, Panzer S. Prasugrel reduces agonists’ inducible platelet activation and leukocyte-platelet interaction more efficiently than clopidogrel. Cardiovasc Ther. 2013;31:e40–5.CrossRefPubMed Gremmel T, Eslam RB, Koppensteiner R, Lang IM, Panzer S. Prasugrel reduces agonists’ inducible platelet activation and leukocyte-platelet interaction more efficiently than clopidogrel. Cardiovasc Ther. 2013;31:e40–5.CrossRefPubMed
7.
Zurück zum Zitat Gremmel T, Frelinger AL 3rd, Michelson AD. Platelet physiology. Semin Thromb Hemost. 2016;42:191–204.CrossRefPubMed Gremmel T, Frelinger AL 3rd, Michelson AD. Platelet physiology. Semin Thromb Hemost. 2016;42:191–204.CrossRefPubMed
8.
Zurück zum Zitat Brandt JT, Payne CD, Wiviott SD, Weerakkody G, Farid NA, Small DS, et al. A comparison of prasugrel and clopidogrel loading doses on platelet function: magnitude of platelet inhibition is related to active metabolite formation. Am Heart J. 2007;153:66 e9–16.CrossRef Brandt JT, Payne CD, Wiviott SD, Weerakkody G, Farid NA, Small DS, et al. A comparison of prasugrel and clopidogrel loading doses on platelet function: magnitude of platelet inhibition is related to active metabolite formation. Am Heart J. 2007;153:66 e9–16.CrossRef
9.
Zurück zum Zitat Jernberg T, Payne CD, Winters KJ, Darstein C, Brandt JT, Jakubowski JA, et al. Prasugrel achieves greater inhibition of platelet aggregation and a lower rate of non-responders compared with clopidogrel in aspirin-treated patients with stable coronary artery disease. Eur Heart J. 2006;27:1166–73.CrossRefPubMed Jernberg T, Payne CD, Winters KJ, Darstein C, Brandt JT, Jakubowski JA, et al. Prasugrel achieves greater inhibition of platelet aggregation and a lower rate of non-responders compared with clopidogrel in aspirin-treated patients with stable coronary artery disease. Eur Heart J. 2006;27:1166–73.CrossRefPubMed
11.
Zurück zum Zitat Motovska Z, Hlinomaz O, Kala P, Hromadka M, Knot J, Varvarovsky I, et al. 1-year outcomes of patients undergoing primary angioplasty for myocardial infarction treated with prasugrel versus ticagrelor. J Am Coll Cardiol. 2018;71:371–81.CrossRefPubMed Motovska Z, Hlinomaz O, Kala P, Hromadka M, Knot J, Varvarovsky I, et al. 1-year outcomes of patients undergoing primary angioplasty for myocardial infarction treated with prasugrel versus ticagrelor. J Am Coll Cardiol. 2018;71:371–81.CrossRefPubMed
12.
Zurück zum Zitat Watti H, Dahal K, Zabher HG, Katikaneni P, Modi K, Abdulbaki A. Comparison of prasugrel and ticagrelor in patients with acute coronary syndrome undergoing percutaneous coronary intervention: a meta-analysis of randomized and non-randomized studies. Int J Cardiol. 2017;249:66–72.CrossRefPubMed Watti H, Dahal K, Zabher HG, Katikaneni P, Modi K, Abdulbaki A. Comparison of prasugrel and ticagrelor in patients with acute coronary syndrome undergoing percutaneous coronary intervention: a meta-analysis of randomized and non-randomized studies. Int J Cardiol. 2017;249:66–72.CrossRefPubMed
13.
Zurück zum Zitat Michelson AD. Antiplatelet therapies for the treatment of cardiovascular disease. Nat Rev Drug Discov. 2010;9:154–69.CrossRefPubMed Michelson AD. Antiplatelet therapies for the treatment of cardiovascular disease. Nat Rev Drug Discov. 2010;9:154–69.CrossRefPubMed
14.
Zurück zum Zitat Cognasse F, Nguyen KA, Damien P, McNicol A, Pozzetto B, Hamzeh-Cognasse H, et al. The inflammatory role of platelets via their TLRs and Siglec receptors. Front Immunol. 2015;6:83.CrossRefPubMedPubMedCentral Cognasse F, Nguyen KA, Damien P, McNicol A, Pozzetto B, Hamzeh-Cognasse H, et al. The inflammatory role of platelets via their TLRs and Siglec receptors. Front Immunol. 2015;6:83.CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Duvernay MT, Temple KJ, Maeng JG, Blobaum AL, Stauffer SR, Lindsley CW, et al. Contributions of protease-activated receptors PAR1 and PAR4 to thrombin-induced GPIIbIIIa activation in human platelets. Mol Pharmacol. 2017;91:39–47.CrossRefPubMedPubMedCentral Duvernay MT, Temple KJ, Maeng JG, Blobaum AL, Stauffer SR, Lindsley CW, et al. Contributions of protease-activated receptors PAR1 and PAR4 to thrombin-induced GPIIbIIIa activation in human platelets. Mol Pharmacol. 2017;91:39–47.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Beaulieu LM, Freedman JE. The role of inflammation in regulating platelet production and function: toll-like receptors in platelets and megakaryocytes. Thromb Res. 2010;125:205–9.CrossRefPubMed Beaulieu LM, Freedman JE. The role of inflammation in regulating platelet production and function: toll-like receptors in platelets and megakaryocytes. Thromb Res. 2010;125:205–9.CrossRefPubMed
17.
18.
Zurück zum Zitat Coggins M, Rosenzweig A. The fire within: cardiac inflammatory signaling in health and disease. Circ Res. 2012;110:116–25.CrossRefPubMed Coggins M, Rosenzweig A. The fire within: cardiac inflammatory signaling in health and disease. Circ Res. 2012;110:116–25.CrossRefPubMed
19.
Zurück zum Zitat D'Atri LP, Schattner M. Platelet toll-like receptors in thromboinflammation. Front Biosci (Landmark Ed). 2017;22:1867–83.CrossRef D'Atri LP, Schattner M. Platelet toll-like receptors in thromboinflammation. Front Biosci (Landmark Ed). 2017;22:1867–83.CrossRef
20.
Zurück zum Zitat Kaczorowski DJ, Nakao A, McCurry KR, Billiar TR. Toll-like receptors and myocardial ischemia/reperfusion, inflammation, and injury. Curr Cardiol Rev. 2009;5:196–202.CrossRefPubMedPubMedCentral Kaczorowski DJ, Nakao A, McCurry KR, Billiar TR. Toll-like receptors and myocardial ischemia/reperfusion, inflammation, and injury. Curr Cardiol Rev. 2009;5:196–202.CrossRefPubMedPubMedCentral
21.
Zurück zum Zitat Gurses KM, Kocyigit D, Yalcin MU, Canpinar H, Oto MA, Ozer N, et al. Enhanced platelet toll-like receptor 2 and 4 expression in acute coronary syndrome and stable angina pectoris. Am J Cardiol. 2015;116:1666–71.CrossRefPubMed Gurses KM, Kocyigit D, Yalcin MU, Canpinar H, Oto MA, Ozer N, et al. Enhanced platelet toll-like receptor 2 and 4 expression in acute coronary syndrome and stable angina pectoris. Am J Cardiol. 2015;116:1666–71.CrossRefPubMed
22.
Zurück zum Zitat Hally KE, La Flamme AC, Larsen PD, Harding SA. Platelet toll-like receptor (TLR) expression and TLR-mediated platelet activation in acute myocardial infarction. Thromb Res. 2017;158:8–15.CrossRefPubMed Hally KE, La Flamme AC, Larsen PD, Harding SA. Platelet toll-like receptor (TLR) expression and TLR-mediated platelet activation in acute myocardial infarction. Thromb Res. 2017;158:8–15.CrossRefPubMed
24.
Zurück zum Zitat Nylander S, Mattsson C, Ramstrom S, Lindahl TL. The relative importance of the ADP receptors, P2Y12 and P2Y1, in thrombin-induced platelet activation. Thromb Res. 2003;111:65–73.CrossRefPubMed Nylander S, Mattsson C, Ramstrom S, Lindahl TL. The relative importance of the ADP receptors, P2Y12 and P2Y1, in thrombin-induced platelet activation. Thromb Res. 2003;111:65–73.CrossRefPubMed
25.
Zurück zum Zitat Whitley MJ, Henke DM, Ghazi A, Nieman M, Stoller M, Simon LM, et al. The protease-activated receptor 4 Ala120Thr variant alters platelet responsiveness to low-dose thrombin and protease-activated receptor 4 desensitization, and is blocked by non-competitive P2Y12 inhibition. J Thromb Haemost. 2018;16:2501–14.CrossRefPubMedPubMedCentral Whitley MJ, Henke DM, Ghazi A, Nieman M, Stoller M, Simon LM, et al. The protease-activated receptor 4 Ala120Thr variant alters platelet responsiveness to low-dose thrombin and protease-activated receptor 4 desensitization, and is blocked by non-competitive P2Y12 inhibition. J Thromb Haemost. 2018;16:2501–14.CrossRefPubMedPubMedCentral
26.
Zurück zum Zitat Kalvegren H, Skoglund C, Helldahl C, Lerm M, Grenegard M, Bengtsson T. Toll-like receptor 2 stimulation of platelets is mediated by purinergic P2X1-dependent Ca2+ mobilisation, cyclooxygenase and purinergic P2Y1 and P2Y12 receptor activation. Thromb Haemost. 2010;103:398–407.CrossRefPubMed Kalvegren H, Skoglund C, Helldahl C, Lerm M, Grenegard M, Bengtsson T. Toll-like receptor 2 stimulation of platelets is mediated by purinergic P2X1-dependent Ca2+ mobilisation, cyclooxygenase and purinergic P2Y1 and P2Y12 receptor activation. Thromb Haemost. 2010;103:398–407.CrossRefPubMed
27.
Zurück zum Zitat Tunjungputri RN, van der Ven AJ, Riksen N, Rongen G, Tacke S, van den Berg TN, et al. Differential effects of platelets and platelet inhibition by ticagrelor on TLR2- and TLR4-mediated inflammatory responses. Thromb Haemost. 2015;113:1035–45.CrossRefPubMed Tunjungputri RN, van der Ven AJ, Riksen N, Rongen G, Tacke S, van den Berg TN, et al. Differential effects of platelets and platelet inhibition by ticagrelor on TLR2- and TLR4-mediated inflammatory responses. Thromb Haemost. 2015;113:1035–45.CrossRefPubMed
28.
Zurück zum Zitat Kahn ML, Nakanishi-Matsui M, Shapiro MJ, Ishihara H, Coughlin SR. Protease-activated receptors 1 and 4 mediate activation of human platelets by thrombin. J Clin Invest. 1999;103:879–87.CrossRefPubMedPubMedCentral Kahn ML, Nakanishi-Matsui M, Shapiro MJ, Ishihara H, Coughlin SR. Protease-activated receptors 1 and 4 mediate activation of human platelets by thrombin. J Clin Invest. 1999;103:879–87.CrossRefPubMedPubMedCentral
29.
Zurück zum Zitat Kahn ML, Zheng YW, Huang W, Bigornia V, Zeng D, Moff S, et al. A dual thrombin receptor system for platelet activation. Nature. 1998;394:690–4.CrossRefPubMed Kahn ML, Zheng YW, Huang W, Bigornia V, Zeng D, Moff S, et al. A dual thrombin receptor system for platelet activation. Nature. 1998;394:690–4.CrossRefPubMed
30.
Zurück zum Zitat De Candia E. Mechanisms of platelet activation by thrombin: a short history. Thromb Res. 2012;129:250–6.CrossRefPubMed De Candia E. Mechanisms of platelet activation by thrombin: a short history. Thromb Res. 2012;129:250–6.CrossRefPubMed
31.
Zurück zum Zitat Badr Eslam R, Lang IM, Koppensteiner R, Calatzis A, Panzer S, Gremmel T. Residual platelet activation through protease-activated receptors (PAR)-1 and -4 in patients on P2Y12 inhibitors. Int J Cardiol. 2013;168:403–6.CrossRefPubMed Badr Eslam R, Lang IM, Koppensteiner R, Calatzis A, Panzer S, Gremmel T. Residual platelet activation through protease-activated receptors (PAR)-1 and -4 in patients on P2Y12 inhibitors. Int J Cardiol. 2013;168:403–6.CrossRefPubMed
32.
Zurück zum Zitat Gremmel T, Kopp CW, Seidinger D, Koppensteiner R, Steiner S, Panzer S. Preserved thrombin-inducible platelet activation in thienopyridine-treated patients. Eur J Clin Investig. 2013;43:689–97.CrossRef Gremmel T, Kopp CW, Seidinger D, Koppensteiner R, Steiner S, Panzer S. Preserved thrombin-inducible platelet activation in thienopyridine-treated patients. Eur J Clin Investig. 2013;43:689–97.CrossRef
33.
34.
Zurück zum Zitat Cattaneo M, Schulz R, Nylander S. Adenosine-mediated effects of ticagrelor: evidence and potential clinical relevance. J Am Coll Cardiol. 2014;63:2503–9.CrossRefPubMed Cattaneo M, Schulz R, Nylander S. Adenosine-mediated effects of ticagrelor: evidence and potential clinical relevance. J Am Coll Cardiol. 2014;63:2503–9.CrossRefPubMed
35.
Zurück zum Zitat Bonello L, Laine M, Kipson N, Mancini J, Helal O, Fromonot J, et al. Ticagrelor increases adenosine plasma concentration in patients with an acute coronary syndrome. J Am Coll Cardiol. 2014;63:872–7.CrossRefPubMed Bonello L, Laine M, Kipson N, Mancini J, Helal O, Fromonot J, et al. Ticagrelor increases adenosine plasma concentration in patients with an acute coronary syndrome. J Am Coll Cardiol. 2014;63:872–7.CrossRefPubMed
36.
Zurück zum Zitat Nylander S, Femia EA, Scavone M, Berntsson P, Asztely AK, Nelander K, et al. Ticagrelor inhibits human platelet aggregation via adenosine in addition to P2Y12 antagonism. J Thromb Haemost. 2013;11:1867–76.PubMed Nylander S, Femia EA, Scavone M, Berntsson P, Asztely AK, Nelander K, et al. Ticagrelor inhibits human platelet aggregation via adenosine in addition to P2Y12 antagonism. J Thromb Haemost. 2013;11:1867–76.PubMed
37.
Zurück zum Zitat Chow JC, Young DW, Golenbock DT, Christ WJ, Gusovsky F. Toll-like receptor-4 mediates lipopolysaccharide-induced signal transduction. J Biol Chem. 1999;274:10689–92.CrossRefPubMed Chow JC, Young DW, Golenbock DT, Christ WJ, Gusovsky F. Toll-like receptor-4 mediates lipopolysaccharide-induced signal transduction. J Biol Chem. 1999;274:10689–92.CrossRefPubMed
38.
Zurück zum Zitat Hammes SR, Coughlin SR. Protease-activated receptor-1 can mediate responses to SFLLRN in thrombin-desensitized cells: evidence for a novel mechanism for preventing or terminating signaling by PAR1’s tethered ligand. Biochemistry. 1999;38:2486–93.CrossRefPubMed Hammes SR, Coughlin SR. Protease-activated receptor-1 can mediate responses to SFLLRN in thrombin-desensitized cells: evidence for a novel mechanism for preventing or terminating signaling by PAR1’s tethered ligand. Biochemistry. 1999;38:2486–93.CrossRefPubMed
39.
Zurück zum Zitat Faruqi TR, Weiss EJ, Shapiro MJ, Huang W, Coughlin SR. Structure-function analysis of protease-activated receptor 4 tethered ligand peptides. Determinants of specificity and utility in assays of receptor function. J Biol Chem. 2000;275:19728–34.CrossRefPubMed Faruqi TR, Weiss EJ, Shapiro MJ, Huang W, Coughlin SR. Structure-function analysis of protease-activated receptor 4 tethered ligand peptides. Determinants of specificity and utility in assays of receptor function. J Biol Chem. 2000;275:19728–34.CrossRefPubMed
40.
Zurück zum Zitat Gremmel T, Steiner S, Seidinger D, Koppensteiner R, Panzer S, Kopp CW. In vivo and protease-activated receptor-1-mediated platelet activation but not response to antiplatelet therapy predict two-year outcomes after peripheral angioplasty with stent implantation. Thromb Haemost. 2014;111:474–82.CrossRefPubMed Gremmel T, Steiner S, Seidinger D, Koppensteiner R, Panzer S, Kopp CW. In vivo and protease-activated receptor-1-mediated platelet activation but not response to antiplatelet therapy predict two-year outcomes after peripheral angioplasty with stent implantation. Thromb Haemost. 2014;111:474–82.CrossRefPubMed
41.
42.
Zurück zum Zitat Gremmel T, Steiner S, Seidinger D, Koppensteiner R, Panzer S, Kopp CW. Comparison of methods to evaluate clopidogrel-mediated platelet inhibition after percutaneous intervention with stent implantation. Thromb Haemost. 2009;101:333–9.CrossRefPubMed Gremmel T, Steiner S, Seidinger D, Koppensteiner R, Panzer S, Kopp CW. Comparison of methods to evaluate clopidogrel-mediated platelet inhibition after percutaneous intervention with stent implantation. Thromb Haemost. 2009;101:333–9.CrossRefPubMed
43.
Zurück zum Zitat Wadowski PP, Eichelberger B, Kopp CW, Pultar J, Seidinger D, Koppensteiner R, et al. Disaggregation following agonist-induced platelet activation in patients on dual antiplatelet therapy. J Cardiovasc Transl Res. 2017;10:359–67.CrossRefPubMedPubMedCentral Wadowski PP, Eichelberger B, Kopp CW, Pultar J, Seidinger D, Koppensteiner R, et al. Disaggregation following agonist-induced platelet activation in patients on dual antiplatelet therapy. J Cardiovasc Transl Res. 2017;10:359–67.CrossRefPubMedPubMedCentral
44.
Zurück zum Zitat Linnemann B, Schwonberg J, Mani H, Prochnow S, Lindhoff-Last E. Standardization of light transmittance aggregometry for monitoring antiplatelet therapy: an adjustment for platelet count is not necessary. J Thromb Haemost. 2008;6:677–83.CrossRefPubMed Linnemann B, Schwonberg J, Mani H, Prochnow S, Lindhoff-Last E. Standardization of light transmittance aggregometry for monitoring antiplatelet therapy: an adjustment for platelet count is not necessary. J Thromb Haemost. 2008;6:677–83.CrossRefPubMed
45.
Zurück zum Zitat Tantry US, Bonello L, Aradi D, Price MJ, Jeong YH, Angiolillo DJ, et al. Consensus and update on the definition of on-treatment platelet reactivity to adenosine diphosphate associated with ischemia and bleeding. J Am Coll Cardiol. 2013;62:2261–73.CrossRefPubMed Tantry US, Bonello L, Aradi D, Price MJ, Jeong YH, Angiolillo DJ, et al. Consensus and update on the definition of on-treatment platelet reactivity to adenosine diphosphate associated with ischemia and bleeding. J Am Coll Cardiol. 2013;62:2261–73.CrossRefPubMed
46.
Zurück zum Zitat Wadowski PP, Pultar J, Weikert C, Eichelberger B, Panzer B, Huber K, et al. Protease-activated receptor-mediated platelet aggregation in acute coronary syndrome patients on potent P2Y12 inhibitors. Res Pract Thromb Haemost. 2019;3:383–90.CrossRefPubMedPubMedCentral Wadowski PP, Pultar J, Weikert C, Eichelberger B, Panzer B, Huber K, et al. Protease-activated receptor-mediated platelet aggregation in acute coronary syndrome patients on potent P2Y12 inhibitors. Res Pract Thromb Haemost. 2019;3:383–90.CrossRefPubMedPubMedCentral
47.
Zurück zum Zitat Gremmel T, Koppensteiner R, Kaider A, Eichelberger B, Mannhalter C, Panzer S. Impact of variables of the P-selectin - P-selectin glycoprotein ligand-1 axis on leukocyte-platelet interactions in cardiovascular disease. Thromb Haemost. 2015;113:806–12.CrossRefPubMed Gremmel T, Koppensteiner R, Kaider A, Eichelberger B, Mannhalter C, Panzer S. Impact of variables of the P-selectin - P-selectin glycoprotein ligand-1 axis on leukocyte-platelet interactions in cardiovascular disease. Thromb Haemost. 2015;113:806–12.CrossRefPubMed
48.
Zurück zum Zitat Wencel-Drake JD, Plow EF, Kunicki TJ, Woods VL, Keller DM, Ginsberg MH. Localization of internal pools of membrane glycoproteins involved in platelet adhesive responses. Am J Pathol. 1986;124:324–34.PubMedPubMedCentral Wencel-Drake JD, Plow EF, Kunicki TJ, Woods VL, Keller DM, Ginsberg MH. Localization of internal pools of membrane glycoproteins involved in platelet adhesive responses. Am J Pathol. 1986;124:324–34.PubMedPubMedCentral
49.
Zurück zum Zitat Hynes RO. Integrins: versatility, modulation, and signaling in cell adhesion. Cell. 1992;69:11–25.CrossRefPubMed Hynes RO. Integrins: versatility, modulation, and signaling in cell adhesion. Cell. 1992;69:11–25.CrossRefPubMed
50.
Zurück zum Zitat Plow EF, Haas TA, Zhang L, Loftus J, Smith JW. Ligand binding to integrins. J Biol Chem. 2000;275:21785–8.CrossRefPubMed Plow EF, Haas TA, Zhang L, Loftus J, Smith JW. Ligand binding to integrins. J Biol Chem. 2000;275:21785–8.CrossRefPubMed
51.
Zurück zum Zitat Neumann FJ, Zohlnhofer D, Fakhoury L, Ott I, Gawaz M, Schomig A. Effect of glycoprotein IIb/IIIa receptor blockade on platelet-leukocyte interaction and surface expression of the leukocyte integrin mac-1 in acute myocardial infarction. J Am Coll Cardiol. 1999;34:1420–6.CrossRefPubMed Neumann FJ, Zohlnhofer D, Fakhoury L, Ott I, Gawaz M, Schomig A. Effect of glycoprotein IIb/IIIa receptor blockade on platelet-leukocyte interaction and surface expression of the leukocyte integrin mac-1 in acute myocardial infarction. J Am Coll Cardiol. 1999;34:1420–6.CrossRefPubMed
52.
Zurück zum Zitat Michelson AD, Barnard MR, Krueger LA, Frelinger AL 3rd, Furman MI. Evaluation of platelet function by flow cytometry. Methods. 2000;21:259–70.CrossRefPubMed Michelson AD, Barnard MR, Krueger LA, Frelinger AL 3rd, Furman MI. Evaluation of platelet function by flow cytometry. Methods. 2000;21:259–70.CrossRefPubMed
53.
Zurück zum Zitat Toth O, Calatzis A, Penz S, Losonczy H, Siess W. Multiple electrode aggregometry: a new device to measure platelet aggregation in whole blood. Thromb Haemost. 2006;96:781–8.CrossRefPubMed Toth O, Calatzis A, Penz S, Losonczy H, Siess W. Multiple electrode aggregometry: a new device to measure platelet aggregation in whole blood. Thromb Haemost. 2006;96:781–8.CrossRefPubMed
54.
Zurück zum Zitat Cattaneo M, Cerletti C, Harrison P, Hayward CP, Kenny D, Nugent D, et al. Recommendations for the standardization of light transmission aggregometry: a consensus of the working party from the platelet physiology subcommittee of SSC/ISTH. J Thromb Haemost. 2013; 11: 1183–1189. Cattaneo M, Cerletti C, Harrison P, Hayward CP, Kenny D, Nugent D, et al. Recommendations for the standardization of light transmission aggregometry: a consensus of the working party from the platelet physiology subcommittee of SSC/ISTH. J Thromb Haemost. 2013; 11: 1183–1189.
55.
Zurück zum Zitat Mayer K, Bernlochner I, Braun S, Schulz S, Orban M, Morath T, et al. Aspirin treatment and outcomes after percutaneous coronary intervention: results of the ISAR-ASPI registry. J Am Coll Cardiol. 2014;64:863–71.CrossRefPubMed Mayer K, Bernlochner I, Braun S, Schulz S, Orban M, Morath T, et al. Aspirin treatment and outcomes after percutaneous coronary intervention: results of the ISAR-ASPI registry. J Am Coll Cardiol. 2014;64:863–71.CrossRefPubMed
56.
Zurück zum Zitat Geisler T, Langer H, Wydymus M, Gohring K, Zurn C, Bigalke B, et al. Low response to clopidogrel is associated with cardiovascular outcome after coronary stent implantation. Eur Heart J. 2006;27:2420–5.CrossRefPubMed Geisler T, Langer H, Wydymus M, Gohring K, Zurn C, Bigalke B, et al. Low response to clopidogrel is associated with cardiovascular outcome after coronary stent implantation. Eur Heart J. 2006;27:2420–5.CrossRefPubMed
57.
Zurück zum Zitat Gum PA, Kottke-Marchant K, Welsh PA, White J, Topol EJ. A prospective, blinded determination of the natural history of aspirin resistance among stable patients with cardiovascular disease. J Am Coll Cardiol. 2003;41:961–5.CrossRefPubMed Gum PA, Kottke-Marchant K, Welsh PA, White J, Topol EJ. A prospective, blinded determination of the natural history of aspirin resistance among stable patients with cardiovascular disease. J Am Coll Cardiol. 2003;41:961–5.CrossRefPubMed
58.
Zurück zum Zitat Alexopoulos D, Galati A, Xanthopoulou I, Mavronasiou E, Kassimis G, Theodoropoulos KC, et al. Ticagrelor versus prasugrel in acute coronary syndrome patients with high on-clopidogrel platelet reactivity following percutaneous coronary intervention: a pharmacodynamic study. J Am Coll Cardiol. 2012;60:193–9.CrossRefPubMed Alexopoulos D, Galati A, Xanthopoulou I, Mavronasiou E, Kassimis G, Theodoropoulos KC, et al. Ticagrelor versus prasugrel in acute coronary syndrome patients with high on-clopidogrel platelet reactivity following percutaneous coronary intervention: a pharmacodynamic study. J Am Coll Cardiol. 2012;60:193–9.CrossRefPubMed
59.
Zurück zum Zitat Alexopoulos D, Xanthopoulou I, Gkizas V, Kassimis G, Theodoropoulos KC, Makris G, et al. Randomized assessment of ticagrelor versus prasugrel antiplatelet effects in patients with ST-segment-elevation myocardial infarction. Circ Cardiovasc Interv. 2012;5:797–804.CrossRefPubMed Alexopoulos D, Xanthopoulou I, Gkizas V, Kassimis G, Theodoropoulos KC, Makris G, et al. Randomized assessment of ticagrelor versus prasugrel antiplatelet effects in patients with ST-segment-elevation myocardial infarction. Circ Cardiovasc Interv. 2012;5:797–804.CrossRefPubMed
60.
Zurück zum Zitat Hally KE, La Flamme AC, Harding SA, Larsen PD. The effects of aspirin and ticagrelor on toll-like receptor (TLR)-mediated platelet activation: results of a randomized, cross-over trial. Platelets. 2019;30:599-607. Hally KE, La Flamme AC, Harding SA, Larsen PD. The effects of aspirin and ticagrelor on toll-like receptor (TLR)-mediated platelet activation: results of a randomized, cross-over trial. Platelets. 2019;30:599-607.
61.
Zurück zum Zitat Shashkin PN, Brown Gt Fau-Ghosh A, Ghosh A Fau-Marathe GK, Marathe Gk Fau-McIntyre TM, McIntyre TM. Lipopolysaccharide is a direct agonist for platelet RNA splicing. J Immunol. 2008;181:3495–502.CrossRefPubMed Shashkin PN, Brown Gt Fau-Ghosh A, Ghosh A Fau-Marathe GK, Marathe Gk Fau-McIntyre TM, McIntyre TM. Lipopolysaccharide is a direct agonist for platelet RNA splicing. J Immunol. 2008;181:3495–502.CrossRefPubMed
62.
Zurück zum Zitat Hawrylowicz CM, Howells GL, Feldmann M. Platelet-derived interleukin 1 induces human endothelial adhesion molecule expression and cytokine production. J Exp Med. 1991;174:785–90.CrossRefPubMed Hawrylowicz CM, Howells GL, Feldmann M. Platelet-derived interleukin 1 induces human endothelial adhesion molecule expression and cytokine production. J Exp Med. 1991;174:785–90.CrossRefPubMed
63.
Zurück zum Zitat Loppnow H, Bil R, Hirt S, Schonbeck U, Herzberg M, Werdan K, et al. Platelet-derived interleukin-1 induces cytokine production, but not proliferation of human vascular smooth muscle cells. Blood. 1998;91:134–41.CrossRefPubMed Loppnow H, Bil R, Hirt S, Schonbeck U, Herzberg M, Werdan K, et al. Platelet-derived interleukin-1 induces cytokine production, but not proliferation of human vascular smooth muscle cells. Blood. 1998;91:134–41.CrossRefPubMed
64.
Zurück zum Zitat Jiang Z, Zhang R, Sun M, Liu Q, Wang S, Wang W, et al. Effect of clopidogrel vs ticagrelor on platelet aggregation and inflammation markers after percutaneous coronary intervention for ST-elevation myocardial infarction. Can J Cardiol. 2018;34:1606–12.CrossRefPubMed Jiang Z, Zhang R, Sun M, Liu Q, Wang S, Wang W, et al. Effect of clopidogrel vs ticagrelor on platelet aggregation and inflammation markers after percutaneous coronary intervention for ST-elevation myocardial infarction. Can J Cardiol. 2018;34:1606–12.CrossRefPubMed
65.
Zurück zum Zitat Schoergenhofer C, Schwameis M, Hobl EL, Ay C, Key NS, Derhaschnig U, et al. Potent irreversible P2Y12 inhibition does not reduce LPS-induced coagulation activation in a randomized, double-blind, placebo-controlled trial. Clin Sci (Lond). 2016;130:433–40.CrossRef Schoergenhofer C, Schwameis M, Hobl EL, Ay C, Key NS, Derhaschnig U, et al. Potent irreversible P2Y12 inhibition does not reduce LPS-induced coagulation activation in a randomized, double-blind, placebo-controlled trial. Clin Sci (Lond). 2016;130:433–40.CrossRef
66.
Zurück zum Zitat Armstrong D, Summers C, Ewart L, Nylander S, Sidaway JE, van Giezen JJ. Characterization of the adenosine pharmacology of ticagrelor reveals therapeutically relevant inhibition of equilibrative nucleoside transporter 1. J Cardiovasc Pharmacol Ther. 2014;19:209–19.CrossRefPubMed Armstrong D, Summers C, Ewart L, Nylander S, Sidaway JE, van Giezen JJ. Characterization of the adenosine pharmacology of ticagrelor reveals therapeutically relevant inhibition of equilibrative nucleoside transporter 1. J Cardiovasc Pharmacol Ther. 2014;19:209–19.CrossRefPubMed
67.
Zurück zum Zitat Pareek M, Bhatt DL. Ticagrelor for patients with acute coronary syndromes: PLATOnic affair or lasting SWEDEHEART? Eur Heart J. 2016;37:3343–6.CrossRefPubMed Pareek M, Bhatt DL. Ticagrelor for patients with acute coronary syndromes: PLATOnic affair or lasting SWEDEHEART? Eur Heart J. 2016;37:3343–6.CrossRefPubMed
68.
Zurück zum Zitat Fender AC, Rauch BH, Geisler T, Schror K. Protease-activated receptor PAR-4: an inducible switch between thrombosis and vascular inflammation? Thromb Haemost. 2017;117:2013–25.CrossRefPubMed Fender AC, Rauch BH, Geisler T, Schror K. Protease-activated receptor PAR-4: an inducible switch between thrombosis and vascular inflammation? Thromb Haemost. 2017;117:2013–25.CrossRefPubMed
70.
Zurück zum Zitat Tricoci P, Huang Z, Held C, Moliterno DJ, Armstrong PW, Van de Werf F, et al. Thrombin-receptor antagonist vorapaxar in acute coronary syndromes. N Engl J Med. 2012;366:20–33.CrossRefPubMed Tricoci P, Huang Z, Held C, Moliterno DJ, Armstrong PW, Van de Werf F, et al. Thrombin-receptor antagonist vorapaxar in acute coronary syndromes. N Engl J Med. 2012;366:20–33.CrossRefPubMed
71.
Zurück zum Zitat Wong PC, Seiffert D, Bird JE, Watson CA, Bostwick JS, Giancarli M, et al. Blockade of protease-activated receptor-4 (PAR4) provides robust antithrombotic activity with low bleeding. Sci Transl Med. 2017;9:eaaf5294. Wong PC, Seiffert D, Bird JE, Watson CA, Bostwick JS, Giancarli M, et al. Blockade of protease-activated receptor-4 (PAR4) provides robust antithrombotic activity with low bleeding. Sci Transl Med. 2017;9:eaaf5294.
72.
Zurück zum Zitat Wilson SJ, Ismat FA, Wang Z, Cerra M, Narayan H, Raftis J, et al. PAR4 (protease-activated receptor 4) antagonism with BMS-986120 inhibits human ex vivo thrombus formation. Arterioscler Thromb Vasc Biol. 2018;38:448–56.CrossRefPubMedPubMedCentral Wilson SJ, Ismat FA, Wang Z, Cerra M, Narayan H, Raftis J, et al. PAR4 (protease-activated receptor 4) antagonism with BMS-986120 inhibits human ex vivo thrombus formation. Arterioscler Thromb Vasc Biol. 2018;38:448–56.CrossRefPubMedPubMedCentral
Metadaten
Titel
Ticagrelor Inhibits Toll-Like and Protease-Activated Receptor Mediated Platelet Activation in Acute Coronary Syndromes
verfasst von
Patricia P. Wadowski
Constantin Weikert
Joseph Pultar
Silvia Lee
Beate Eichelberger
Renate Koppensteiner
Irene M. Lang
Simon Panzer
Thomas Gremmel
Publikationsdatum
15.02.2020
Verlag
Springer US
Erschienen in
Cardiovascular Drugs and Therapy / Ausgabe 1/2020
Print ISSN: 0920-3206
Elektronische ISSN: 1573-7241
DOI
https://doi.org/10.1007/s10557-019-06932-7

Weitere Artikel der Ausgabe 1/2020

Cardiovascular Drugs and Therapy 1/2020 Zur Ausgabe

Update Kardiologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.