Abstract
Aim: In percutaneous coronary intervention (PCI)-treated acute coronary syndrome (ACS) patients on clopidogrel therapy, high on-treatment platelet adenosine diphosphate (ADP) reactivity was observed in numerous studies, with significant increases in non-fatal myocardial infarction, definite/probable stent thrombosis, or cardiovascular mortality. Compared to clopidogrel, prasugrel and ticagrelor provide more potent platelet inhibition. Whether new P2Y12 inhibitors reduce thrombotic events in a similar manner compared to the rate observed with appropriate P2Y12 inhibition by clopidogrel must still be determined. This study sought to compare longterm outcomes between clopidogrel responders (platelet reactivity index [PRI] vasodilator-stimulated phosphoprotein [VASP] < 61%) and patients under prasugrel or ticagrelor therapy following PCI-treated ACS.
Methods: 730 ACS patients undergoing urgent PCI were prospectively enrolled into two groups: clopidogrel responders (n = 448) and those under ticagrelor or prasugrel therapy (n = 282). The primary endpoint was a composite of cardiovascular death, myocardial infarction, stent thrombosis, and stroke; the secondary endpoint comprised major hemorrhagic events.
Results: The median follow-up was 260 ± 186 days. Clopidogrel patients were older and more likely to present non-ST segment elevation myocardial infarction, cardiovascular risk factors, atrial fibrillation, or prior vascular disease. After propensity score matching, the primary endpoint was met in 7.1% of the clopidogrel group and 4.1% of the prasugrel/ticagrelor group (p = 0.43). Minor bleeding events were significantly reduced in the clopidogrel group (1.1% vs. 3%; p = 0.03). In a multivariate analysis, the antiplatelet treatment strategy was not an independent primary endpoint predictor.
Conclusion: In PCI-treated ACS patients, clopidogrel therapy and PRI VASP < 61% were not associated with increased risks of thrombotic events compared to prasugrel or ticagrelor therapy.
Keywords: Myocardial infarction, Thrombosis, Stent, Bleeding
Aim
In acute coronary syndrome (ACS) patients treated with percutaneous coronary intervention (PCI) and antiplatelet therapy including clopidogrel therapy, high on-treatment platelet reactivity (HPR) to adenosine diphosphate (ADP) was shown to be associated with a significant increase in non-fatal myocardial infarction, definite/probable stent thrombosis (ST), or cardiovascular mortality1–7). Among the well-described factors associated with ST8), HPR to ADP likely accounted for up to 60% of early ST events. Yet, to date, large randomized trials concerning personalized antiplatelet therapies failed to confirm the benefits of platelet function assessment in improving survival. Disappointments in personalized antiplatelet therapies9–11) could be explained by the observation that HPR on clopidogrel treatment allows not only for ineffective P2Y12 receptor inhibition to be detected but also does take into account co-existing patent comorbidities interfering with clopidogrel pharmacodynamics such as chronic renal diseases and advanced age3, 12–17). These comorbidities per se contribute to adverse outcomes and are unlikely modified by using more potent P2Y12 receptor inhibitors. Optimal P2Y12 inhibition detected by platelet function assays was, however, shown to enable identifying patients at lower ischemic risk3, 9, 17, 18). Whether the use of new P2Y12 inhibitors could result in reduced thrombotic events compared to those observed in patients with appropriate clopidogrel-induced P2Y12 inhibition has yet to be investigated. Prasugrel and ticagrelor provide more potent platelet inhibition compared to clopidogrel, with a consistent reduction in thrombotic events19, 20). European and US guidelines have thus advocated the use of prasugrel or ticagrelor instead of clopidogrel in PCI-treated ACS patients21, 22) based on the net benefits observed with new P2Y12 inhibitors over clopidogrel in the PLATO and TRITON trials19, 20). However, no randomized data are available on the long-term efficacy or safety of the use of new P2Y12 inhibitors over the appropriate P2Y12 blockade by clopidogrel. One limitation of using more potent P2Y12 inhibitors is grounded on the greater risk of bleeding19, 20, 23), with an increased risk of short- and long-term morbidity in ACS patients24). Another issue is the cost/effectiveness of antiplatelet strategy. As clopidogrel has become generic, the novel P2Y12 inhibitors' high treatment costs along with the incr eased risk of bleeding could impede their use. We thus sought to compare long-term clinical outcomes between clopidogrel responders (PRI VASP < 61%) and pati ents under prasugrel or ticagrelor therapy following PCI-treated ACS.
Methods
This study prospectively enrolled patients undergoing PCI due to ACS between January 2008 and April 2015 in the Nouvel Hôpital Civil, CHU Strasbourg, France. The trial was performed in accordance with the Declaration of Helsinki, with the protocol approved by the institutional ethics committee and informed written consent obtained from all patients.
Study Population
Inclusion criteria: Patients > 18 years old and admitted to the cardiac intensive care unit for PCI with stent implantation due to ACS, with or without ST-segment elevation, or unstable angina. VASP measurement during hospital stay. In general, VASP was realized in high-risk patients more likely to present an enhanced thrombotic risk, in complex PCI, or in clopidogrel-treated patients to ascertain platelet responsiveness.
Exclusion criteria: Significant dementia, absent PRI measurement by VASP assay under clopidogrel treatment, PRI > 61% under clopidogrel, switch from either prasugrel or ticagrelor to clopidogrel, failed PCI, lack of stent implantation, contraindication to antiplatelet therapy, cardiogenic shock requiring critical care unit admission, and cardiogenic pulmonary edema requiring mechanical ventilation.
Blood Samples
A blood sample was taken between 6–48 hours after the clopidogrel loading dose (300 or 600 mg). Blood was immediately collected into a vacutainer tube, citrated, and sent to the hemostasis laboratory (EFS-Alsace, France), where a platelet VASP phosphorylation analysis was performed within 48 hours.
Platelet function assays: VASP phosphorylation analysis by flow cytometry
VASP phosphorylation was assessed with standardized flow cytometric assay (Platelet VASP; Diagnostica Stago [Biocytex], Asnières, France). A citrated blood sample was incubated with either prostaglandin E1 (PGE1) or PGE1 and ADP for 10 min, fixed with paraformaldehyde, and the platelets were then permeabilized with a non-ionic detergent. The cells were labeled with a primary monoclonal antibody against serine 239-phosphorylated VASP (16C2), followed by a secondary fluorescein isothiocyanate-conjugated polyclonal goat-anti-mouse antibody. Analyses were performed on a Becton Dickinson FACS Calibur flow cytometer as reported. PRI was calculated from median fluorescence intensity (MFI) of samples incubated with PGE1 and ADP according to the formula: PRI VASP = (MFI[PGE1] − MFI[PGE1 + ADP]/MFI[PGE1]) × 100. PRI, expr essed as a percentage, is the difference in VASP fluorescence intensity between resting (+ PGE1) and activated (+ ADP) platelets. In unselected patients undergoing PCI, the optimal cutoff value for PRI to predict cardiovascular outcome following PCI was recently found to be 61% using a receiver-operating characteristic curve analysis based on the Youden's index maximum value. Patients were considered low clopidogrel responders if their PRI was ≥ 61%, and normal clopidogrel responders if their PRI was < 61%18). In our experience, the 50% threshold did not allow a relevant identification of low clopidogrel responders18).
Study Protocol
The choice of antiplatelet therapy was left to the clinicians' discretion. Patients were also treated by intravenous aspirin (125–250 mg) and 50–100 IU/Kg of unfractionated heparin to target an ACT > 250 s. GPIIbIIIa inhibitors, mainly Abciximab, were used at the operators' discretion. Data was extracted from our database. The whole cohort of 730 patients was split into two subgroups: clopidogrel responders and prasugrel/ticagrelor. The clopidogrel responder group included clopidogrel-treated patients with a PRI value < 61% following a 300–600 mg loading dose. The clopidogrel maintenance dose was 75 mg/day. The prasugrel or ticagrelor group included patients treated with 10 mg prasugrel daily after a loading dose of 60 mg, or 90 mg ticagrelor twice daily after a loading dose of 180 mg. During hospitalization, patients who switched to prasugrel or ticagrelor were placed into the prasugrel/ticagrelor group, and patients who switched to clopidogrel being excluded.
The study flowchart is illustrated in Fig. 1.
Fig. 1.
Flow chart.
Study Objectives
The primary efficacy endpoint was the major adverse cardiac event rate (MACE), defined as the composite of cardiovascular death, both definite and probable stent thrombosis, myocardial infarction (STEMI or NSTEMI), and stroke. ST-segment elevation myocardial infarction (STEMI) was defined as a new ST-segment elevation in two consecutive leads with increased biochemical myocardial necrosis markers, and non-STEMI (NSTEMI) as occurrence of ischemic symptoms associated with ST-segment depression and T-wave abnormalities and increased biochemical myocardial necrosis markers. Post-PCI troponin (Tn) elevations were not considered indicative of recurrent myocardial infarction. In line with the Academic Research Consortium criteria, two ST types were distinguished: 1) definite ST defined as an ACS proved by angiographic or pathologic evidence; 2) probable ST corresponding to unexplained death within 30 days or target vessel infarction without angiographic information. Stroke was defined as a focal loss of neurologic function caused by ischemic events, with residual symptoms lasting > 24 hours. Secondary analyses were performed for each primary endpoint component.
The secondary endpoint was the occurrence of major bleeding, with bleeding severity defined using the Bleeding Academic Research Consortium (BARC) criteria. Major bleeding was defined as a BARC score ≥ Type 3b, and minor bleeding as a BARC score < Type 3b.
Follow-up information was obtained using a written questionnaire via a telephone interview with the cardiologist, referring physician, or patient. In the absence of response, the patient's electronic medical file was consulted. Endpoints were adjudicated by two physicians who were blinded to treatment allocation.
Statistical Analysis
Continuous variables were expressed as median (interquartile range, 25th and 75th percentile) or mean ± SD, and categorical variables as frequencies and percentages. Continuous variables between both groups were compared using Student's t-test or Mann–Whitney U test, as appropriate. Fisher's exact test was used to compare categorical variables. Continuous variables were analyzed for normal distribution using the Shapiro–Wilk test. Time to event was defined as the time from PCI to the event date, with patients censored at death, loss to follow-up, or study end on April 30, 2015.
Propensity score (PS) matching analysis with 1:1 nearest neighbor matching was employed. Variables used in developing PS have been marked by* in Tables 1–3. The main variables were age, gender, clinical presentation, cardiovascular risk factor, peripheral vascular disease, three-vessel disease and chronic kidney disease. These were chosen based on the thrombotic risk's clinical relevance. Following PS analysis, 268 clopidogrel-treated patients were matched with an equal number of prasugrel- or ticagrelor-treated patients.
Table 1. Baseline demographic and clinical characteristics.
Variable, n (%) | Clopidogrel | Prasugrel/Ticagrelor | p |
---|---|---|---|
(n = 448) | (n = 282) | ||
Clinical presentation, n (%) | |||
STEMI, n (%) | 189 (42.2) | 175 (62.1) | 0.0001* |
NSTEMI, n (%) | 202 (45.1) | 93 (33.0) | 0.001* |
Unstable angina, n (%) | 54 (12.1) | 13 (4.6) | 0.0005* |
Symptom, n (%) | |||
Killip ≥ 2 | 48 (10.8) | 27 (9.6) | 0.671 |
Prior angina | 102 (28.1) | 99 (35.1) | 0.06 |
Demographic, n (%) | |||
Age (year) | 66.8 +/− 13.5 (28–93) | 57.8 +/− 11.2 (31–89) | 0.0001* |
Sex | |||
Male | 310 (69.2) | 237 (84.0) | 0.0001* |
Female | 139 (30.8) | 45 (16.0) | 0.0001* |
Risk factors/past medical history, n (%) | |||
Current smoking | 188 (42) | 142 (50.4) | 0.03* |
Hypertension | 262 (58.5) | 137 (48.6) | 0.009 * |
Diabetes mellitus | 117 (26) | 67 (23.8) | 0.5 |
Obesity (BMI > 30 Kg/m2) | 26.6 +/− 4.6 (16–44.7) | 27.7 +/− 4.9 (17–54) | 0.01* |
Hyperlipidemia | 240 (53.6) | 129 (45.7) | 0.04* |
Family history of coronary artery disease (CAD) | 69 (15.4) | 70 (24.8) | 0.002* |
Prior STEMI | 77 (17.2) | 35 (12.5) | 0.09 |
Prior NSTEMI | 30 (6.7) | 23 (8.2) | 0.47 |
Prior angioplasty | 91 (20.4) | 42 (14.9) | 0.07 |
Prior CABG | 28 (6.2) | 8 (2.8) | 0.05 |
Prior Stroke | 31 (6.9) | 7 (2.5) | 0.009 |
Peripheral vascular disease | 47 (10.5) | 13 (4.6) | 0.005* |
Chronic Kidney Disease | 29 (6.5) | 12 (4.3) | 0.25* |
Echographic characteristics, | |||
Left Ventricular Ejection Fraction (%) | 52.6 +/− 12.2 (15–80) | 52.9 +/− 10.6 (20–80) | 0.81 |
Treatment, n (%) | |||
ACE inhibitors | 408 (93) | 259 (92) | 0.6 |
Beta–blockers | 413 (93.9) | 273 (96.8) | 0.08 |
Statins | 426 (96.8) | 276 (97.9) | 0.4 |
Oral anticoagulants (VKA antagonists) | 58 (13.2) | 0 (0.0) | 0.0001 |
GPIIbIIIa antagonist | 108 (25.4) | 87 (31) | 0.08 |
Values are n +/− median (range) or n (%)
Variables used to create propensity score
ACE = angiotensin-converting enzyme; BMI = body mass index; CABG = coronary artery bypass graft surgery; NSTEMI = Non ST segment elevation myocardial infarction; STEMI = ST segment elevation myocardial infarction
Table 3. Biological characteristics.
Clopidogrel | Prasugrel/Ticagrelor | p | |
---|---|---|---|
(n = 448) | (n = 282) | ||
Glycemia (g/dL) | 1.43 +/− 0.7 (0.57–4.75) | 1.47 +/− 0.6 (0.68–5.11) | 0.5 |
HbA1c (%) | 6.3 +/− 1.4 (4.5–16.4) | 6.0 +/− 1.1 (4.7–12.8) | 0.002 |
Creatinine (umol/L) | 88.4 +/− 45.0 (38.7–565) | 76.6 +/− 24.9 (37–248) | 0.0001 |
Tn admission (ug/L) | 0.33 [0.008–2.07] | 0.41 [0.07–3.27] | 0.2 |
Tn peak (ug/L) | 8.24 [0.70–8.24] | 28.60 [3.90–80.90] | 0.0001 |
BNP (ng/l) | 123 [52–289] | 50 [21–123] | 0.0001 |
CRP (mg/l) | 4.70 [4–14] | 4 [4–7] | 0.002 |
Leukocytes (10^9/L) | 9.9 +/− 3.8 (1–29.8) | 11.1 +/− 3.8 (3.9–25.5) | 0.0001 |
Hb (g/dl) | 13.7 +/− 1.8 (7.8–19.1) | 14.50 +/− 1.6 (5.1–19.1) | 0.0001 |
Platelets (10^9/L) | 253.8 +/− 85.1 (48–832) | 247.6 +/− 70.5 (85–818) | 0.3 |
Total cholesterol (g/L) | 1.8 +/− 0.5 (0.8–5.1) | 1.8 +/− 0.4 (0.9–3.2) | 0.4 |
LDLc (g/L) | 1.1 +/− 0.4 (0.3–2.4) | 1.1 +/− 0.3 (0.3–2.4) | 0.3 |
HDLc (g/L) | 0.4 +/− 0.1 (0.1–1.8) | 0.4 +/− 0.1 (0.2–1.5) | 0.0001 |
TG (g/L) | 1.3 +/− 0.9 (0.3–7.7) | 1.3 +/− 0.9 (0.4–9.5) | 0.01 |
VASP PRI (%) | 37.3 +/− 16.8 (3–60) | 22.2 +/− 21.2 (3–60) | 0.0001 |
Values are n +/− standard deviation (minimum–maximum)
BNP = brain natriuretic peptide; CRP = C-reactive protein; Hb = Hemoglobin; HDLc = High-density lipoprotein; LDLc = Low-density lipoprotein; TG = Triglycerides; Tn = Troponin
Table 2. Baseline angiographic characteristics.
Clopidogrel | Prasugrel/Ticagrelor | p | |
---|---|---|---|
(n = 448) | (n = 282) | ||
Mono-vessel disease, n (%) | 170 (38) | 142 (50,5) | 0.001 |
Dual-vessel disease, n (%) | 144 (32) | 91 (32,4) | 0.9 |
Three-vessel disease, n (%) | 135 (30) | 48 (17,1) | 0.0001* |
LAD, n (%) | 299 (66,7) | 173 (61,6) | 0.2 |
CX, n (%) | 161 (36) | 85 (30,2) | 0.1 |
RCA, n (%) | 265 (59,2) | 138 (49,1) | 0.009 |
Left main coronary artery, n (%) | 26 (5,8) | 10 (3,6) | 0.2 |
Bifurcation n (%) | 37 (8,2) | 5 (1,8) | 0.0001 |
Total stent's length (mm) | 25.9 +/− 15.5 (12–122) | 29.9 +/− 21.7 (12–188) | 0.005 |
Stent's diameter (mm) | 3.1 +/− 0.5 (2.5–4.5) | 3.0 +/− 0.5 (3–5) | 0.83 |
DES | 236 (52.7) | 259 (92.5) | < 0.0001 |
Values are n +/− median (range) or n (%)
Variable used to create propensity score
LAD = left anterior descending artery; CX = circumflex artery; RCA = right coronary artery
Kaplan–Meier analysis was employed to establish survival plots without MACE or major bleeding, with the two groups compared using the log-rank test. The Cox model was used to estimate hazard ratios (HRs) and 95% confidence intervals (CIs). A multivariate analysis of MACE was done using Cox models. Variables with p < 0.05 in univariate analysis were entered into a stepwise ascending multivariate analysis. The Cox regression results were presented as HRs, 95% CIs, and p-values. A p value < 0.05 was considered statistically significant.
Statistical analyses were performed using SPSS Version 13.0 software (SPSS Inc., Chicago, Illinois) and the R software (R Development Core Team [2008], Vienna, Austria). The significance level was set at 5%.
Results
Patient Characteristics
From January 1, 2008 to April 30, 2015, 7009 patients were admitted to our department (NHC, Strasbourg, France) for ACS treated by PCI and stent implantation, with 790 patients under clopidogrel fulfilling the inclusion criteria, 448 patients of which (57%) exhibited a VASP < 61% proving appropriate P2Y12 inhibition. These patients were compared to 282 patients under prasugrel (n = 161, 57%) or ticagrelor (n = 121, 43%) therapy.
The whole cohort's baseline characteristics were provided in Tables 1–3.
Patients under clopidogrel treatment were generally older, less likely to be male, and more likely to present NSTEMI, multiple comorbidities, and prior atrial fibrillation. The coronary artery disease extent and the bifurcation's lesions were more significant in the clopidogrel group. PRI value, a marker of P2Y12 inhibition, was significantly lower in the prasugrel/ticagrelor group. Peak Tn was lower in the clopidogrel group, in line with the group's lower proportion of STEMI, along with higher levels of HbA1c, BNP, and CRP in this group. The timing of VASP testing was longer in the clopidogrel group (clopidogrel 30 +/− 20 h vs 23 +/− 28, p < 0.001) probably reflecting the fact that this group presented longer hospital stay (older, multiple comorbidities, etc.).
Characteristics of the patients enrolled in the PS analysis are given in Tables 4–6. Even after PS matching, important differences remained between the two subsets. Of note, clopidogrel patients were older, presented a higher rate of bifurcation lesion and were more frequently implanted with BMS.
Table 4. Baseline demographic and clinical characteristics after propensity score matching.
Variable, n (%) | Clopidogrel | Prasugrel/Ticagrelor | p |
---|---|---|---|
(n = 268) | (n = 268) | ||
Clinical presentation, n (%) | |||
STEMI, n (%) | 137 (51.1) | 166 (61.9) | 0.01 |
NSTEMI, n (%) | 110 (41) | 88 (32.8) | 0.06 |
Unstable angina, n (%) | 20 (7.5) | 13 (4.8) | 0.28 |
Symptom, n (%) | |||
Killip ≥ 2 | 17 (6.4) | 25 (9.3) | 0.26 |
Prior angina | 68 (31.3) | 94 (35.1) | 0.44 |
Demographic, n (%) | |||
Age (year) | 61 +/− 12 (31–89) | 58 +/− 11 (31–88) | 0.001 |
Sex | |||
Male | 223 (83.2) | 226 (84.3) | 0.81 |
Female | 45 (16.8) | 42 (15.7) | 0.81 |
Risk factors/past medical history, n (%) | |||
Current smoking | 135 (50.4) | 135 (50.4) | 1 |
Hypertension | 136 (50.8) | 130 (48.5) | 0.67 |
Diabetes mellitus | 61 (22.8) | 66 (24.6) | 0.33 |
Obesity (BMI > 30 Kg/m2) | 27 +/− 4.7 (18.5–44.8) | 27 +/− 4.9 (17–54) | 0.74 |
Hyperlipidemia | 135 (50.4) | 121 (45.1) | 0.26 |
Family history of coronary artery disease (CAD) | 52 (19.4) | 67 (25) | 0.15 |
Prior STEMI | 37 (13.9) | 34 (12.7) | 0.70 |
Prior NSTEMI | 14 (5.3) | 21 (7.9) | 0.29 |
Prior angioplasty | 46 (17.2) | 40 (14.9) | 0.48 |
Prior CABG | 9 (3.4) | 7 (2.6) | 0.80 |
Prior Stroke | 13 (4.8) | 7 (2.6) | 0.25 |
Peripheral vascular disease | 15 (5.6) | 12 (4.5) | 0.69 |
Chronic Kidney Disease | 9 (3.4) | 12 (4.5) | 0.66 |
Echographic characteristics, | |||
Left Ventricular Ejection Fraction (%) | 55 +/− 11.3 (25–76) | 55 +/− 10.7 (20–80) | 0.30 |
Treatment, n (%) | |||
ACE inhibitors | 245 (93.9) | 247 (92.2) | 0.50 |
Beta–blockers | 250 (95.4) | 259 (96.6) | 0.51 |
Statins | 254 (97) | 262 (97.8) | 0.60 |
Oral anticoagulants (VKA antagonists) | 32 (12.3) | 0 (0.0) | 0.0001 |
GPIIbIIIa antagonist | 88 (33.7) | 83 (31) | 0.52 |
Values are n +/− median (range) or n (%)
ACE = angiotensin-converting enzyme; BMI = body mass index; CABG = coronary artery bypass graft surgery; NSTEMI = Non ST segment elevation myocardial infarction; STEMI = ST segment elevation myocardial infarction
Table 6. Biological characteristics after propensity score matching.
Clopidogrel | Prasugrel/Ticagrelor | P | |
---|---|---|---|
(n = 268) | (n = 268) | ||
Glycemia (g/dL) | 1.2 +/− 0.7 (0.7–4.8) | 1.3 +/− 0.6 (0.7–5.1) | 0.007 |
HbA1c (%) | 5.9 +/− 1.4 (4.5–16.4) | 5.7 +/− 1.2 (4.7–12.8) | 0.001 |
Creatinine (umol/L) | 77.3 +/− 47.7 (38.7–565) | 72.3 +/− 25.4 (37–248.3) | 0.006 |
Tn admission (ug/L) | 0.4 [0.04–480] | 0.4 [0.04–304] | 0.90 |
Tn peak (ug/L) | 12.3 [1.4–528] | 28.9 [4–738] | 0.0001 |
BNP (ng/l) | 91 [37–5598] | 52 [21–2193] | 0.0003 |
CRP (mg/l) | 4.4 [4–249] | 4 [4–193] | 0.001 |
Leukocytes (10^9/L) | 9.3 +/− 3.8 (1.3–29.8) | 10.4 +/− 3.8 (4–25.5) | 0.002 |
Hb (g/dl) | 14.2 +/− 1.7 (7.8–19.1) | 14.7 +/− 1.6 (5.1–19.1) | 0.004 |
Platelets (10^9/L) | 234 +/− 76.7 (67–584) | 240 +/− 69.6 (85–818) | 0.94 |
Total cholesterol (g/L) | 1.8 +/− 0.5 (0.8–5) | 1.8 +/− 0.4 (0.9–4) | 0.34 |
LDLc (g/L) | 1.1 +/− 0.4 (0.4–2.2) | 1.1 +/− 0.4 (0.3–2.4) | 0.66 |
HDLc (g/L) | 0.4 +/− 0.1 (0.1–0.9) | 0.4 +/− 0.1 (0.2–1.5) | 0.0034 |
TG (g/L) | 1.1 +/− 1 (0.3–7.2) | 1.3 +/− 1 (0.4–9.5) | 0.002 |
VASP PRI (%) | 41.9 +/− 17 (3–60) | 15 +/− 21.5 (2–60) | 0.0001 |
Values are n +/− standard deviation (minimum–maximum)
BNP = brain natriuretic peptide; CRP = C-reactive protein; Hb = Hemoglobin; HDLc = High-density lipoprotein; LDLc = Low-density lipoprotein; TG = Triglycerides; Tn = Troponin
Table 5. Baseline angiographic characteristics after propensity score matching.
Clopidogrel | Prasugrel/Ticagrelor | P | |
---|---|---|---|
(n = 268) | (n = 268) | ||
Mono-vessel disease, n (%) | 131 (48.9) | 136 (50,8) | 0.73 |
Dual-vessel disease, n (%) | 81 (30.2) | 86 (32,1) | 0.71 |
Three-vessel disease, n (%) | 56 (20.9) | 46 (17,2) | 0.32 |
LAD, n (%) | 160 (59.7) | 165 (61.6) | 0.72 |
CX, n (%) | 83 (31.1) | 79 (29.5) | 0.71 |
RCA, n (%) | 143 (53.4) | 131 (48.9) | 0.34 |
Left main coronary artery, n (%) | 13 (4.8) | 10 (3.7) | 0.67 |
Bifurcation n (%) | 20 (7.5) | 4 (1.5) | 0.0012 |
Total stent's length (mm) | 20 +/− 14.9 (15–122) | 34 +/− 22 (18–188) | 0.0016 |
Stent's diameter (mm) | 3 +/− 0.5 (3–4.5) | 3.0 +/− 0.8 (2.8–5) | 0.64 |
DES | 133 (49.6) | 247 (92.5) | 0.0001 |
Values are n +/− median (range) or n (%)
LAD = left anterior descending artery; CX = circumflex artery; RCA = right coronary artery
Clinical Outcomes
Clinical outcomes were available for 684 of 730 patients (93.7%), with a mean follow-up of 260 ± 186 days. Of the 46 patients lost to follow-up (6.3%), 17 (2.3%) were using clopidogrel and 29 (3.9%) prasugrel or ticagrelor. At 30 days, no significant differences in MACE, cardiac death, definite and probable ST, myocardial infarction, stroke and bleedings between groups could be evidenced in the whole cohort and after PS analysis (Table 7).
Table 7. Events at 30 days.
Before PS matching |
After PS matching |
|||||
---|---|---|---|---|---|---|
Clopidogrel | Prasugrel/Ticagrelor | p | Clopidogrel | Prasugrel/Ticagrelor | p | |
(n = 448) | (n = 282) | (n = 268) | (n = 268) | |||
MACE, n (%) | 15 (3.3) | 3 (1.06) | 0.071 | 9 (3.4) | 0 (0) | 0.20 |
Cardiac death | 4 (0.89) | 0 (0) | 0.125 | 2 (0.7) | 0 (0) | 1 |
Myocardial infarction | 8 (1.78) | 3 (1.06) | 0.503 | 3 (1.1) | 0 (0) | 0.74 |
Stent thrombosis definite | 5 (1.1) | 0 (0) | 0.086 | 3 (1.1) | 0 (0) | 0.28 |
Stent thrombosis probable | 4 (0.9) | 0 (0) | 0.125 | 3 (1.1) | 0 (0) | 0.55 |
Stroke | 1 (0.22) | 0 (0) | 0.43 | 1 (0.4) | 0 (0) | 1 |
Bleeding, n (%) | 14 (3.2) | 13 (5.1) | 0.214 | 4 (1.5) | 2 (0.7) | 0.58 |
Major bleeding | 2 (0.4) | 1 (0.35) | 0.904 | 3 (1.1) | 1 (0.4) | 0.84 |
Minor bleeding | 2 (0.4) | 2 (0.7) | 0.579 | 1 (0.4) | 1 (0.4) | 0.78 |
Values are n (%)
At the end of the follow-up in the whole cohort, the composite primary endpoint occurred in 7.8% of the clopidogrel patients and 3.9% of those treated with prasugrel/ticagrelor (p = 0.034). Myocardial infarction, definite and probable ST, and stroke rates did not significantly differ between groups, while higher cardiac death rates were observed under clopidogrel (Table 8). Kaplan–Meier analyses for MACE-free survival probability did not significantly differ (log-rank test, p = 0.108) (Fig. 2).
Table 8. Events at the end of the follow-up.
Before PS matching |
After PS matching |
|||||
---|---|---|---|---|---|---|
Clopidogrel | Prasugrel/Ticagrelor | p | Clopidogrel | Prasugrel/Ticagrelor | p | |
(n = 448) | (n = 282) | (n = 268) | (n = 268) | |||
MACE, n (%) | 35 (7.8) | 11 (3.9) | 0.034 | 19 (7.1) | 11 (4.1) | 0.43 |
Cardiac death | 12 (2.8) | 1 (0.4) | 0.028 | 7 (2.6) | 1 (0.4) | 0.11 |
Myocardial infarction | 19 (4.4) | 9 (3.6) | 0.590 | 12 (4.5) | 9 (3.4) | 0.99 |
Stent thrombosis definite | 8 (1.8) | 2 (0.8) | 0.288 | 5 (1.9) | 2 (0.7) | 0.44 |
Stent thrombosis probable | 4 (0.9) | 1 (0.4) | 0.454 | 1 (0.4) | 1 (0.4) | 0.86 |
Stroke | 3 (0.7) | 1 (0.4) | 0.621 | 2 (0.7) | 1 (0.4) | 0.41 |
Bleeding, n (%) | 14 (3.2) | 13 (5.1) | 0.214 | 5 (1.9) | 11 (4.1) | 0.08 |
Major bleeding | 7 (1.6) | 3 (1.2) | 0.551 | 2 (0.7) | 3 (1.1) | 0.44 |
Minor bleeding | 7 (1.8) | 10 (4) | 0.091 | 3 (1.1) | 8 (3) | 0.03 |
Values are n (%)
Fig. 2.
Kaplan Meier Analysis of survival without MACE in the whole cohort.
There were 10 major bleeding events (1.3%) and 17 minor (2.3%) recorded at follow-up, with no significant between-group differences (Table 8). A Kaplan–Meier analysis for major bleeding-free survival probability has been presented in Fig. 3.
Fig. 3.
Kaplan-Meier Analysis of survival without major bleeding in the whole cohort.
At the end of the follow-up, event rates following PS matching were given in Table 8. After matching the two groups for recognized confounding cofactors, there was no significant increase in thrombotic events and major bleeding observed in patients under appropriate clopidogrel inhibition (Table 8), along with reduced minor bleeding events (Table 8).
A Kaplan–Meier analysis for the probability of MACE-free survival, major bleeding-free survival after PS is given in Figs. 4 and 5.
Fig. 4.
Kaplan Meier Analysis of survival without MACE after propensity score matching.
Fig. 5.
Kaplan-Meier Analysis of survival without major bleeding after propensity score matching.
MACE Predictors
In univariate Cox analysis, age, clinical presentation, diabetes mellitus, renal dysfunction, medical history, stroke, or vascular diseases, Killip Class 2 to 4, high CRP level, high Tn level at admission, total stent length, three-vessel disease, and hemorrhagic events were significant MACE predictors. No significant impact of clopidogrel treatment allocation on MACE was established (HR 1.741; 95% CI: 0.884–3.429); p = 0.109). Multivariate Cox regression analysis identified hemorrhagic events and high CRP and troponin levels at admission as independent MACE predictors (Table 9).
Table 9. Univariate and multivariate analyses to predict MACE.
Variable | Univariate Analysis |
Multivariate Analysis |
||
---|---|---|---|---|
HR [95% CI] | P value | HR [95%CI] | P value | |
Age | 1.022 [1.000–1.046] | 0.051 | 1.003 [0.977–1.030] | 0.814 |
Sex | 1.456 [0.786–2.698] | 0.232 | ||
STEMI | 0,542 [0.296–0.995] | 0.048 | ||
NSTEMI | 2.011 [1.122–3.604] | 0,019 | 1.742 [0.844–3.592] | 0.133 |
Unstable angina | 0,813 [0,292–2.269] | 0,693 | ||
BMI | 1.012 [0,952–1.075] | 0,710 | ||
Smoking | 0.645 [0.351–1.182] | 0.156 | ||
Diabetes mellitus | 2.290 [1.278–4.102] | 0.005 | 0.959 [0.455–2.023] | 0.913 |
Hypertension | 1.536 [0.837–2.818] | 0.166 | ||
Dyslipidemia | 1.530 [0.846–2.765] | 0.160 | ||
Prior PCI | 0.954 [0.460–1.977] | 0.899 | ||
CKD | 3.594 [1.675–7.711] | 0.001 | 1.726 [0.641–4.645] | 0.280 |
History of stroke | 3.065 [1.298–7.238] | 0.011 | 1.447 [0.503–4.162] | 0.493 |
Peripheral vascular disease | 2.566 [1.238–5.318] | 0,011 | 1.865 [0.807–4.311] | 0.145 |
Killip 2 to 4 | 2.295 [1.108–4.757] | 0.025 | 1.940 [0.368–2.400] | 0.897 |
Creatinine | 1.003 [0.999–1.008] | 0.180 | ||
HbA1c | 1.207 [1.025–1.422] | 0.024 | ||
BNP at admission | 1.000 [1.000–1.001] | 0.020 | ||
CRP at admission | 1.011 [1.005–1.017] | 0.001 | 1.011 [1.004–1.018] | 0.003 |
Troponin at admission | 1.006 [1.002–1.011] | 0,005 | 1.008 [1.002–1.015] | 0.015 |
Left ventricular ejection fraction | 0.970 [0.947–0.993] | 0.011 | ||
Clopidogrel | 1.741 [0.884–3.429] | 0.109 | ||
ACE-inhibitor | 0.320 [0.149–0.686] | 0,003 | ||
Statin | 0.274 [0.098–0.765] | 0,013 | ||
Stent's total length | 1.016 [1.005–1.026] | 0,003 | 1.006 [0.990–1.022] | 0.487 |
Stent's Diameter | 0.955 [0.608–1.500] | 0.843 | ||
DES | 1.865 [0.946–3.678] | 0.072 | ||
Three-vessel disease | 1.816 [1.004–3.284] | 0.048 | 1.532 [0.757–3.101] | 0.236 |
Left main coronary artery | 1.770 [0.635–4.937] | 0.275 | ||
Bifurcation | 1.744 [0.689–4.415] | 0.240 | ||
Hemorrhagic event | 4.952 [1.938–12.656] | 0.001 | 3.119 [1.020–9.533] | 0.046 |
HR = Hazard ratio; CI = confidence interval
ACE = angiotensin-converting enzyme; BMI = body mass index; BNP = brain natriuretic peptide; CRP = C-reactive protein; DES = drug eluting stent; NSTEMI = Non ST segment elevation myocardial infarction; STEMI = ST segment elevation myocardial infarction
Stent Thrombosis Predictors
In univariate Cox analysis, age, three-vessel disease, total number of implanted stents, high CRP levels, and hemorrhagic events were significant predictors of definite/probable ST. No significant impact of clopidogrel treatment allocation on ST was established (HR 2.23; 95% CI: 0.63–7.91; p = 0.211). Multivariate Cox regression analysis identified hemorrhagic events, elevated CRP levels at admission, and total number of implanted stents as independent predictors of definite/probable ST (Table 10).
Table 10. Univariate and Multivariate analyses to predict Stent Thrombosis.
Variable | Univariate Analysis |
Multivariate Analysis |
||
---|---|---|---|---|
HR [95% CI] | P | HR [95%CI] | P | |
Age | 1.04 [0.99–1.08] | 0.05 | 1.015 [0.974–1.058] | 0.471 |
Sex | 0.75 [0.21–2.66] | 0.66 | ||
STEMI | 1.17 [0.43–3.23] | 0.76 | ||
NSTEMI | 1.33 [0.48–3.66] | 0.59 | ||
Smoking | 0.61 [0.21–1.78] | 0.36 | ||
Diabetes | 1.99 [0.71–5.60] | 0.19 | ||
Hypertension | 0.94 [0.34–2.58] | 0.89 | ||
Family history of coronary artery disease | 2.21 [0.75–6.46] | 0.15 | ||
Dyslipidemia | 1.10 [0.40–3.04] | 0.85 | ||
BMI > 30 | 1.17 [0.37–3.68] | 0.79 | ||
Prior STEMI | 0.37 [0.05–2.79] | 0.33 | ||
Prior NSTEMI | 0.89 [0.12–6.74] | 0.91 | ||
Prior PCI | 0.62 [0.14–2.74] | 0.53 | ||
Prior CABG | 1.33 [0.17–10.09] | 0.79 | ||
History of stroke | 1.41 [0.19–10.76] | 0.74 | ||
Peripheral vascular disease | 1.60 [0.36–7.11] | 0.53 | ||
CKD | 1.17 [0.15–8.92] | 0.88 | ||
Killip 2 to 4 | 2.28 [0.64–8.07] | 0.20 | ||
Left ventricular ejection fraction | 0.97 [0.93–1.01] | 0.19 | ||
Clopidogrel | 2.23 [0.63–7.91] | 0.211 | ||
ACE-inhibitor | 0.28 [0.08–1.004] | 0.05 | ||
Statin | 0.075 [0.024–0.24] | 0.0001 | ||
HbA1c | 1.27 [0.99–1.63] | 0.06 | ||
Troponin at admission | 1.006 [0.99–1.01] | 0.10 | ||
CRP at admission | 1.013 [1.004–1.022] | 0.005 | 1.013 [1.004–1.023] | 0.005 |
Three vessel | 3.26 [1.18–9.00] | 0.02 | 1.733 [0.538–5.581] | 0.357 |
Stent number | 1.74 [1.25–2.43] | 0.001 | 1.661 [1.097–2.514] | 0.016 |
Hemorrhagic events | 5.44 [1.21–24.46] | 0.03 | 6.429 [1.349–30.633] | 0.019 |
HR = Hazard ratio; CI = confidence interval
ACE = angiotensin-converting enzyme; BMI = body mass index; BNP = brain natriuretic peptide; CRP = C-reactive protein; DES = drug eluting stent; NSTEMI = Non ST segment elevation myocardial infarction; STEMI = ST segment elevation myocardial infarction
Discussion
Our primary finding was that in PCI-treated ACS patients, appropriate platelet inhibition strategy by clopidogrel, proven by PRI VASP < 61%, did not significantly increase thrombotic event risks compared to prasugrel or ticagrelor therapy. With clopidogrel, there were reduced minor bleeding events with no impact on major bleeding events.
Two large randomized trials primarily enrolling PCI-treated ACS patients have previously demonstrated that prasugrel and ticagrelor substantially reduce thrombotic events compared with clopidogrel19, 20). However, data confirming the new P2Y12 inhibitors' benefits over appropriate platelet inhibition by clopidogrel is still lacking. Owing to impaired clopidogrel-induced platelet inhibition in numerous ACS patients25, 26), one may speculate that the new P2Y12 inhibitors' benefits over clopidogrel were mainly accounted for by a drastic reduction in thrombotic risk compared to the risk observed in HPR patients. Conversely, the new P2Y12 inhibitors' beneficial impact could prove much more limited in patients with appropriate clopidogrel inhibition. In the ACS setting, recent insights suggested that HPR may primarily be an integrate marker of associated comorbidities such as chronic renal disease, ongoing inflammation, and so on, all known to interfere with clopidogrel pharmacodynamics17). These comorbidities are unlikely modified by more potent P2Y12 inhibition using either higher clopidogrel doses or new P2Y12 inhibitors. In the ACS setting, another issue to consider is the bleeding risk that is partially determined by the P2Y12 inhibition extent4, 27). Several studies have emphasized that high platelet inhibition, similar to that by prasugrel or ticagrelor, does increase bleeding risk4, 27). Strategies aiming at an optimal therapeutic window, with the lowest risks of bleeding and ST post-PCI, are highly warranted26, 28). Interestingly, a stage-adapted treatment with potent platelet inhibition in the acute phase (where the thrombotic risk prevails) and de-escalation to clopidogrel in the maintenance phase (to limit the bleeding risk) was very recently shown to be safe. In this pioneering work by Sibbing, guided de-escalation of antiplatelet treatment was non-inferior to the standard treatment with prasugrel at 1 year after PCI in terms of net clinical benefit29). Owing to the global economic crisis, cost/effectiveness appr oaches are widely recommended. In the US, recent analyses have underlined that access to new P2Y12 inhibitors could be limited. In addition to advanced age (> 75 years), vascular comorbidities, black ethnicity, and lack of private insurance were key determinants of clopidogrel prescription in ACS patients30). Besides bleeding and recurrent ischemic event risks, medical drug coverage was recognized as a major det erminant of ADP receptor inhibitor selection in contemporary US practice31). In other countries, health insurers tend to only refund new P2Y12 inhibitors in PCI treatment of high-risk ACS when platelet function assessment confirmed the patients' non-response to clopidogrel32).
Several large-scale real-life registries have compared early outcomes of ACS patients treated by either clopidogrel or new P2Y12 inhibitors30, 33), yet with conflicting results. It must be emphasized that the platelet inhibition extent was not assessed in these studies. In the studies by Alexopoulos, the switch from clopidogrel to new P2Y12 inhibitors was associated with reduced thrombotic MACE at the price of an increased bleeding rate33). In the TRANSLATE ACS trial, death was more commonly observed in patients who pursued clopidogrel compared to those who switched to prasugrel or ticagrelor, whereas MACE, stroke, and recurrent MI risk did not significantly differ. After adjusting for confounding factors, the mortality rate was however lowered30).
Study data was first analyzed in the whole cohort without adjusting for confounding factors. MACE and cardiac death rates were numerically higher in the clopidogrel responder group, whereas these differences were no longer significant after Cox regression or Kaplan–Meier analyses. Thrombotic events were not significantly increased in patients with appropriate platelet inhibition by clopidogrel. Of note is that causes of fatal events remain difficult to ascertain, and calculation of cardiac mortality must thus be taken with caution. To overcome confounding factors, PS analysis was performed, with patients matched according to certain variables associated with enhanced thrombotic risk. Analysis confirmed that the strategy of appropriate inhibition by clopidogrel did not significantly increase the thrombotic risk. On the other hand, such strategy was found ineffective in reducing major bleeding events, in line with a large multicenter Swiss cohort34). Our data extends recent findings by Aradi et al.32). In their study, thrombotic and hemorrhagic events rate were similar in HPR patients switched to prasugrel and in clopidogrel responders32). This suggests that in ACS patients under clopidogrel, drug administration may be continued for managing thrombotic risks provided that appropriate platelet inhibition has been documented.
Safety concerns and the assessment of bleeding events are key when assessing any antiplatelet strategy's net beneficial effects35). When considering the whole cohort, no significant reduction in bleeding events was observed in the clopidogrel group. Following PS matching, the clopidogrel strategy resulted in a significant reduction in minor bleedings, without any impact on major bleedings. In the ACS setting, the interplay between bleeding and thrombotic events was emphasized by several studies. As underlined by Aradi et al., patients with a major bleeding event had a 7-fold increased risk of ST32). Additionally, bleeding together with ST was identified as a strong independent 1-year mortality predictor32). In our study, bleeding was the strongest predictor of MACE and ST. Several hypotheses may be raised to account for the relationship between bleeding and thrombotic events: (i) even minor bleeding may trigger premature antiplatelet agent discontinuation; (ii) greater prevalence of comorbidities in patients suffering from bleeding events; (iii) hemodynamic compromise induced by severe hemorrhages could favor ST; and (iv) transfusion may induce platelet activation17). Lastly, inflammation, either pre-existing or resulting from blood transfusion, was considered an important mediator of thrombotic process36, 37). In line with this, high CRP level was identified in our study as an independent predictor of MACE, including ST. In real world practice, bleeding events appear to be more potent predictors of thrombotic events than the antiplatelet strategy type used. Therefore, the optimal therapeutic window or optimal antiplatelet strategy, enabling us to minimize both bleeding and thrombotic risks, must be further defined.
Study Limitations
This study displays several limitations. Firstly, the applied antiplatelet strategy was not randomized. Moreover, group characteristics differed, advanced age and multiple comorbidities being more common in the clopidogrel group. Secondly, clopidogrel response was only assessed at the acute phase, following bolus dose administration, which could have resulted in clopidogrel response overestimation. Thirdly, as the cause of fatal events remains often difficult to ascertain, estimation of cardiac mortality requires caution. Fourthly, an independent committee did not adjudicate cardiovascular events. Due to the relatively low number of events recorded, multivariate analysis should be interpreted with caution with the findings viewed as hypothesis generating. Fifthly, there was no power calculation performed, and we could not exclude that the limited cohort size could have impeded detecting significant differences between the two strategies. Finally, cost/effectiveness analysis was not performed. While these limitations limit to some extent the validity of our comparison, it must be emphasized that registries are mandatory for collecting real-life data on unselected patients.
Conclusion
The results of this prospective ACS registry suggest that in clopidogrel-treated patients with appropriate platelet inhibition documented by platelet function test, continuing clopidogrel therapy is not associated with increased risks of thrombotic events compared to prasugrel or ticagrelor therapy.
List of Abbreviations
- ACS
acute coronary syndrome
- ACT
Activated Clotting Time
- ADP
adenosine diphosphate
- BARC
Bleeding Academic Research Consortium
- BNP
B-type Natriuretic Peptide
- HPR
high on-treatment platelet reactivity
- MACE
major adverse cardiac event
- NSTEMI
non ST-segment elevation myocardial infarction
- PCI
percutaneous coronary intervention
- PRI
platelet reactivity index
- Tn
Troponin
- VASP
vasodilator-stimulated phosphoprotein
Conflicts of Interest
The authors have no conflicts of interest to declare.
References
- 1). Hochholzer W, Trenk D, Bestehorn HP, Fischer B, Valina CM, Ferenc M, Gick M, Caputo A, Büttner HJ, Neumann FJ. Impact of the degree of peri-interventional platelet inhibition after loading with clopidogrel on early clinical outcome of elective coronary stent placement. Journal of the American College of Cardiology. 2006; 48: 1742-1750 [DOI] [PubMed] [Google Scholar]
- 2). Geisler T, Zurn C, Simonenko R, Rapin M, Kraibooj H, Kilias A, Bigalke B, Stellos K, Schwab M, May AE, Herdeg C, Gawaz M. Early but not late stent thrombosis is influenced by residual platelet aggregation in patients undergoing coronary interventions. European heart journal. 2010; 31: 59-66 [DOI] [PubMed] [Google Scholar]
- 3). Morel O, El Ghannudi S, Jesel L, Radulescu B, Meyer N, Wiesel ML, Caillard S, Campia U, Moulin B, Gachet C, Ohlmann P. Cardiovascular mortality in chronic kidney disease patients undergoing percutaneous coronary intervention is mainly related to impaired P2Y12 inhibition by clopidogrel. Journal of the American College of Cardiology. 2011; 57: 399-408 [DOI] [PubMed] [Google Scholar]
- 4). Cuisset T, Frere C, Quilici J, Morange PE, Camoin L, Bali L, Lambert M, Juhan-Vague I, Alessi MC, Bonnet JL. Relationship between aspirin and clopidogrel responses in acute coronary syndrome and clinical predictors of non response. Thromb Res. 2009; 123: 597-603 [DOI] [PubMed] [Google Scholar]
- 5). Frere C, Cuisset T, Quilici J, Camoin L, Carvajal J, Morange PE, Lambert M, Juhan-Vague I, Bonnet JL, Alessi MC. ADP-induced platelet aggregation and platelet reactivity index VASP are good predictive markers for clinical outcomes in non-ST elevation acute coronary syndrome. Thrombosis and haemostasis. 2007; 98: 838-843 [PubMed] [Google Scholar]
- 6). Range G, Yayehd K, Belle L, Thuaire C, Richard P, Cazaux P, Barbou F, Köning R, Chassaing S, Teiger E, Berthier R, Decomis MP, Claudel JP, Delarche N, Brunel P, De Poli F, Dupouy P, Beygui F, Albert F, Collet JP, Montalescot G. Thrombotic and bleeding events after coronary stenting according to clopidogrel and aspirin platelet reactivity: VerifyNow French Registry (VERIFRENCHY). Arch Cardiovasc Dis. 2014; 107: 225-235 [DOI] [PubMed] [Google Scholar]
- 7). Cuisset T, Cayla G, Silvain J. Clopidogrel resistance: what's new? Arch Cardiovasc Dis. 2010; 103: 349-353 [DOI] [PubMed] [Google Scholar]
- 8). Iakovou I, Schmidt T, Bonizzoni E, Ge L, Sangiorgi GM, Stankovic G, Airoldi F, Chieffo A, Montorfano M, Carlino M, Michev I, Corvaja N, Briguori C, Gerckens U, Grube E, Colombo A. Incidence, predictors, and outcome of thrombosis after successful implantation of drugeluting stents. JAMA. 2005; 293: 2126-2130 [DOI] [PubMed] [Google Scholar]
- 9). Price MJ, Berger PB, Teirstein PS, Tanguay JF, Angiolillo DJ, Spriggs D, Puri S, Robbins M, Garratt KN, Bertrand OF, Stillabower ME, Aragon JR, Kandzari DE, Stinis CT, Lee MS, Manoukian SV, Cannon CP, Schork NJ, Topol EJ. Standard- vs high-dose clopidogrel based on platelet function testing after percutaneous coronary intervention: the GRAVITAS randomized trial. JAMA. 2011; 305: 1097-1105 [DOI] [PubMed] [Google Scholar]
- 10). Trenk D, Stone GW, Gawaz M, Kastrati A, Angiolillo DJ, Muller U, Richardt G, Jakubowski JA, Neumann FJ. A Randomized Trial of Prasugrel Versus Clopidogrel in Patients With High Platelet Reactivity on Clopidogrel After Elective Percutaneous Coronary Intervention With Implantation of Drug-Eluting Stents: Results of the TRIGGER-PCI (Testing Platelet Reactivity In Pati ents Undergoing Elective Stent Placement on Clopidogrel to Guide Alternative Therapy With Prasugrel) Study. Journal of the American College of Cardiology. 2012; 59: 2159-2164 [DOI] [PubMed] [Google Scholar]
- 11). Collet JP, Cuisset T, Range G, Cayla G, Elhadad S, Pouillot C, Henry P, Motreff P, Carrié D, Boueri Z, Belle L, Van Belle E, Rousseau H, Aubry P, Monségu J, Sabouret P, O'Connor SA, Abtan J, Kerneis M, Saint-Etienne C, Barthélémy O, Beygui F, Silvain J, Vicaut E, Montalescot G. Bedside monitoring to adjust antiplatelet therapy for coronary stenting. N Engl J Med. 2012; 367: 2100-2109 [DOI] [PubMed] [Google Scholar]
- 12). Angiolillo DJ, Bernardo E, Capodanno D, Vivas D, Sabate M, Ferreiro JL, Ueno M, Jimenez-Quevedo P, Alfonso F, Bass TA, Macaya C, Fernandez-Ortiz A. Impact of chronic kidney disease on platelet function profiles in diabetes mellitus patients with coronary artery disease taking dual antiplatelet therapy. Journal of the American College of Cardiology. 2010; 55: 1139-1146 [DOI] [PubMed] [Google Scholar]
- 13). El Ghannudi S, Ohlmann P, Jesel L, Radulescu B, El Adraa E, Crimizade U, Wiesel ML, Gachet C, Morel O. Impaired inhibition of P2Y(12) by clopidogrel is a major determinant of cardiac death in diabetes mellitus patients treated by percutaneous coronary intervention. Atherosclerosis. 2011; 217: 465-472 [DOI] [PubMed] [Google Scholar]
- 14). Muller C, Caillard S, Jesel L, El Ghannudi S, Ohlmann P, Sauleau E, Hannedouche T, Gachet C, Moulin B, Morel O. Association of estimated GFR with platelet inhibition in patients treated with clopidogrel. American journal of kidney diseases: the official journal of the National Kidney Foundation. 2012; 59: 777-785 [DOI] [PubMed] [Google Scholar]
- 15). Morel O, El Ghannudi S, Hess S, Reydel A, Crimizade U, Jesel L, Radulescu B, Wiesel ML, Gachet C, Ohlmann P. The extent of P2Y12 inhibition by clopidogrel in diabetes mellitus patients with acute coronary syndrome is not related to glycaemic control: roles of white blood cell count and body weight. Thrombosis and haemostasis. 2012; 108: 338-348 [DOI] [PubMed] [Google Scholar]
- 16). Geisler T, Mueller K, Aichele S, Bigalke B, Stellos K, Htun P, Ninci E, Fateh-Moghadam S, May AE, Gawaz M. Impact of inflammatory state and metabolic control on responsiveness to dual antiplatelet therapy in type 2 diabetics after PCI: prognostic relevance of residual platelet aggregability in diabetics undergoing coronary interventions. Clin Res Cardiol. 2010; 99: 743-752 [DOI] [PubMed] [Google Scholar]
- 17). Tantry US, Bonello L, Aradi D, Price MJ, Jeong YH, Angiolillo DJ, Stone GW, Curzen N, Geisler T, Ten Berg J, Kirtane A, Siller-Matula J, Mahla E, Becker RC, Bhatt DL, Waksman R, Rao SV, Alexopoulos D, Marcucci R, Reny JL, Trenk D, Sibbing D, Gurbel PA. Consensus and update on the definition of on-treatment platelet reactivity to adenosine diphosphate associated with ischemia and bleeding. Journal of the American College of Cardiology. 2013; 62: 2261-2273 [DOI] [PubMed] [Google Scholar]
- 18). El Ghannudi S, Ohlmann P, Meyer N, Wiesel ML, Radulescu B, Chauvin M, Bareiss P, Gachet C, Morel O. Impact of P2Y12 inhibition by clopidogrel on cardiovascular mortality in unselected patients treated by percutaneous coronary angioplasty: a prospective registry. JACC Cardiovascular interventions. 2010; 3: 648-656 [DOI] [PubMed] [Google Scholar]
- 19). Wiviott SD, Braunwald E, McCabe CH, Montalescot G, Ruzyllo W, Gottlieb S, Neumann FJ, Ardissino D, De Servi S, Murphy SA, Riesmeyer J, Weerakkody G, Gibson CM, Antman EM. Prasugrel versus clopidogrel in patients with acute coronary syndromes. N Engl J Med. 2007; 357: 2001-2015 [DOI] [PubMed] [Google Scholar]
- 20). Wallentin L, Becker RC, Budaj A, Cannon CP, Emanuelsson H, Held C, Horrow J, Husted S, James S, Katus H, Mahaffey KW, Scirica BM, Skene A, Steg PG, Storey RF, Harrington RA. Ticagrelor versus clopidogrel in pati ents with acute coronary syndromes. N Engl J Med. 2009; 361: 1045-1057 [DOI] [PubMed] [Google Scholar]
- 21). Authors/Task Force m. Windecker S, Kolh P, Alfonso F, Collet JP, Cremer J, Falk V, Filippatos G, Hamm C, Head SJ, Jüni P, Kappetein AP, Kastrati A, Knuuti J, Landmesser U, Laufer G, Neumann FJ, Richter DJ, Schauerte P, Sousa Uva M, Stefanini GG, Taggart DP, Torracca L, Valgimigli M, Wijns W, Witkowski A. 2014 ESC/EACTS Guidelines on myocardial revascularization: The Task Force on Myocardial Revascularization of the European Society of Cardiology (ESC) and the European Association for Cardio-Thoracic Surgery (EACTS)Developed with the special contribution of the European Association of Percutaneous Cardiovascular Interventions (EAPCI). European heart journal. 2014; 35: 2541-2619 [DOI] [PubMed] [Google Scholar]
- 22). Levine GN, Bates ER, Bittl JA, Brindis RG, Fihn SD, Fleisher LA, Granger CB, Lange RA, Mack MJ, Mauri L, Mehran R, Mukherjee D, Newby LK, O'Gara PT, Sabatine MS, Smith PK, Smith SC., Jr 2016 ACC/AHA Guideline Focused Update on Duration of Dual Antiplatelet Therapy in Pati ents With Coronary Artery Disease: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. Journal of the American College of Cardiology. 2016 [DOI] [PubMed] [Google Scholar]
- 23). Hochholzer W, Wiviott SD, Antman EM, Contant CF, Guo J, Giugliano RP, Dalby AJ, Montalescot G, Braunwald E. Predictors of bleeding and time dependence of association of bleeding with mortality: insights from the Trial to Assess Improvement in Therapeutic Outcomes by Optimizing Platelet Inhibition With Prasugrel--Thrombolysis in Myocardial Infarction 38 (TRITON-TIMI 38). Circulation. 2011; 123: 2681-2689 [DOI] [PubMed] [Google Scholar]
- 24). Chhatriwalla AK, Amin AP, Kennedy KF, House JA, Cohen DJ, Rao SV, Messenger JC, Marso SP. Association between bleeding events and in-hospital mortality after percutaneous coronary intervention. JAMA. 2013; 309: 1022-1029 [DOI] [PubMed] [Google Scholar]
- 25). Bonello L, Tantry US, Marcucci R, Blindt R, Angiolillo DJ, Becker R, Bhatt DL, Cattaneo M, Collet JP, Cuisset T, Gachet C, Montalescot G, Jennings LK, Kereiakes D, Sibbing D, Trenk D, Van Werkum JW, Paganelli F, Price MJ, Waksman R, Gurbel PA. Consensus and future directions on the definition of high on-treatment platelet reactivity to adenosine diphosphate. Journal of the American College of Cardiology. 2010; 56: 919-933 [DOI] [PubMed] [Google Scholar]
- 26). Aradi D, Storey RF, Komocsi A, Trenk D, Gulba D, Kiss RG, Husted S, Bonello L, Sibbing D, Collet JP, Huber K. Expert position paper on the role of platelet function testing in patients undergoing percutaneous coronary intervention. European heart journal. 2014; 35: 209-215 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27). Parodi G, Bellandi B, Venditti F, Carrabba N, Valenti R, Migliorini A, Grassellini S, Ramazzotti E, Antoniucci D. Residual platelet reactivity, bleedings, and adherence to treatment in patients having coronary stent implantation treated with prasugrel. Am J Cardiol. 2012; 109: 214-218 [DOI] [PubMed] [Google Scholar]
- 28). Aradi D, Kirtane A, Bonello L, Gurbel PA, Tantry US, Huber K, Freynhofer MK, ten Berg J, Janssen P, Angiolillo DJ, Siller-Matula JM, Marcucci R, Patti G, Mangiacapra F, Valgimigli M, Morel O, Palmerini T, Price MJ, Cuisset T, Kastrati A, Stone GW, Sibbing D. Bleeding and stent thrombosis on P2Y12-inhibitors: collaborative analysis on the role of platelet reactivity for risk stratification after percutaneous coronary intervention. European heart journal. 2015; 36: 1762-1771 [DOI] [PubMed] [Google Scholar]
- 29). Sibbing D, Aradi D, Jacobshagen C, Gross L, Trenk D, Geisler T, Orban M, Hadamitzky M, Merkely B, Kiss RG, Komócsi A, Dézsi CA, Holdt L, Felix SB, Parma R, Klopotowski M, Schwinger RHG, Rieber J, Huber K, Neumann FJ, Koltowski L, Mehilli J, Huczek Z, Massberg S. Guided de-escalation of antiplatelet treatment in patients with acute coronary syndrome undergoing percutaneous coronary intervention (TROPICAL-ACS): a randomised, open-label, multicentre trial. Lancet. 2017 [DOI] [PubMed] [Google Scholar]
- 30). Bagai A, Peterson ED, Honeycutt E, Effron MB, Cohen DJ, Goodman SG, Anstrom KJ, Gupta A, Messenger JC, Wang TY. In-hospital switching between adenosine diphosphate receptor inhibitors in patients with acute myocardial infarction treated with percutaneous coronary intervention: Insights into contemporary practice from the TRANSLATE-ACS study. Eur Heart J Acute Cardiovasc Care. 2015; 4: 499-508 [DOI] [PubMed] [Google Scholar]
- 31). Bagai A, Wang Y, Wang TY, Curtis JP, Gurm HS, Shah B, Cheema AN, Peterson ED, Saucedo JF, Granger CB, Roe MT, Bhatt DL, McNamara RL, Alexander KP. In-hospital switching between clopidogrel and prasugrel among patients with acute myocardial infarction treated with percutaneous coronary intervention: insights into contemporary practice from the national cardiovascular data registry. Circulation Cardiovascular interventions. 2014; 7: 585-593 [DOI] [PubMed] [Google Scholar]
- 32). Aradi D, Tornyos A, Pinter T, Vorobcsuk A, Konyi A, Falukozy J, Veress G, Magyari B, Horváth IG, Komócsi A. Optimizing P2Y12 receptor inhibition in patients with acute coronary syndrome on the basis of platelet function testing: impact of prasugrel and high-dose clopidogrel. Journal of the American College of Cardiology. 2014; 63: 1061-1070 [DOI] [PubMed] [Google Scholar]
- 33). Alexopoulos D, Xanthopoulou I, Deftereos S, Sitafidis G, Kanakakis I, Hamilos M, Angelidis C, Petousis S, Stakos D, Parissis H, Vavouranakis M, Davlouros P, Goudevenos J, Stefanadis C. In-hospital switching of oral P2Y12 inhibitor treatment in patients with acute coronary syndrome undergoing percutaneous coronary intervention: prevalence, predictors and short-term outcome. American heart journal. 2014; 167: 68-76 e2 [DOI] [PubMed] [Google Scholar]
- 34). Klingenberg R, Heg D, Raber L, Carballo D, Nanchen D, Gencer B, Auer R, Jaguszewski M, Stähli BE, Jakob P, Templin C, Stefanini GG, Meier B, Vogt P, Roffi M, Maier W, Landmesser U, Rodondi N, Mach F, Windecker S, Jüni P, Lüscher TF, Matter CM. Safety profile of prasugrel and clopidogrel in patients with acute coronary syndromes in Switzerland. Heart. 2015; 101: 854-863 [DOI] [PubMed] [Google Scholar]
- 35). Bacquelin R, Oger E, Filippi E, Hacot JP, Auffret V, Le Guellec M, Coudert I, Castellant P, Moquet B, Druelles P, Rialan A, Rouault G, Boulanger B, Treuil J, Leurent G, Bedossa M, Boulmier D, Avez B, Gilard M, Le Breton H. Safety of prasugrel in real-world patients with ST-segment elevation myocardial infarction: 1-year results from a prospective observational study (Bleeding and Myocardial Infarction Study). Arch Cardiovasc Dis. 2016; 109: 31-38 [DOI] [PubMed] [Google Scholar]
- 36). Palmerini T, Genereux P, Mehran R, Dangas G, Caixeta A, Riva DD, Mariani A, Xu K, Stone GW. Association among leukocyte count, mortality, and bleeding in patients with non-ST-segment elevation acute coronary syndromes (from the Acute Catheterization and Urgent Intervention Triage StrategY [ACUITY] trial). Am J Cardiol. 2013; 111: 1237-1245 [DOI] [PubMed] [Google Scholar]
- 37). Inoue T, Croce K, Morooka T, Sakuma M, Node K, Simon DI. Vascular inflammation and repair: implications for re-endothelialization, restenosis, and stent thrombosis. JACC Cardiovascular interventions. 2011; 4: 1057-1066 [DOI] [PMC free article] [PubMed] [Google Scholar]