Next Article in Journal
Endometrial Dysbiosis Is Related to Inflammatory Factors in Women with Repeated Implantation Failure: A Pilot Study
Next Article in Special Issue
Effectiveness and Safety of Apixaban in over 3.9 Million People with Atrial Fibrillation: A Systematic Review and Meta-Analysis
Previous Article in Journal
Soluble Urokinase Plasminogen Activator Receptor Levels Are Associated with Severity of Fibrosis in Patients with Primary Sclerosing Cholangitis
Previous Article in Special Issue
Effect of Sulodexide on Circulating Blood Cells in Patients with Mild COVID-19
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Differences in Optimal Platelet Reactivity after Potent P2Y12 Inhibitor Treatment in Acute Coronary Syndrome Patients Undergoing Percutaneous Coronary Intervention

1
Department of Cardiology, Dong-A University Hospital, Busan 49201, Korea
2
Department of Cardiology, Huizhou Third People’s Hospital, Guangzhou Medical University, Huizhou 516002, China
3
Department of Cardiology, Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, China
4
Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi 563003, China
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
J. Clin. Med. 2022, 11(9), 2480; https://doi.org/10.3390/jcm11092480
Submission received: 11 March 2022 / Accepted: 29 March 2022 / Published: 28 April 2022
(This article belongs to the Special Issue Myths and Realities of Current Cardiovascular Diseases and Therapy)

Abstract

:
Background: East Asian patients receiving treatment with the potent P2Y12 inhibitors prasugrel or ticagrelor experience more potent platelet inhibition than with clopidogrel. Methods: This study investigated differences in OPR rates with reduced doses of prasugrel (n = 38) or ticagrelor (n = 40) for maintenance therapy in 118 Korean ACS patients who had undergone PCI, in comparison to conventional-dose clopidogrel (n = 40). We assessed drug responses at one- and three-months post-PCI with VerifyNow and multiple electrode aggregometry assays. Results: At the one-month period, patients receiving standard-dose prasugrel or ticagrelor had lower platelet reactivity as determined by the three assays than those receiving the conventional dose of clopidogrel (VN: p = 0.000; MEA: p = 0.000; LTA: p = 0.000). At the 3-month point, platelet reactivity was lower in those receiving reduced-dose prasugrel or ticagrelor than the clopidogrel-treated patients (VN: p = 0.000; MEA: p = 0.012; LTA: p = 0.002). Prasugrel resulted in significantly lower platelet inhibition than ticagrelor as determined by VN and LTA (VN: p = 0.000; LTA: p = 0.003). At three months, there was a significant overall difference in OPR among the three groups when measured by VN (p < 0.001), but not when measured by MEA (p = 0.596). OPR in the reduced-dose prasugrel group was not significantly different to the clopidogrel group at three months (VN: p = 0.180; MEA: p = 0.711). OPR in the reduced-dose ticagrelor group was similar to clopidogrel as determined by MEA at three months, but was different when assessed by VN (VN: p = 0.000; MEA: p = 0.540). Compared to standard-dose, the reduced-dose prasugrel OPR rate was significantly increased (VN: p = 0.008; MEA: p = 0.020). Conclusions: OPR values for reduced-dose prasugrel and conventional-dose clopidogrel at three months were similar but higher than for reduced-dose ticagrelor as determined by VN, but no differences were noted by MEA. The MEA assay might have less sensitivity and consistency than the VN assay. Further studies are needed to explore this discrepancy.

1. Introduction

Dual antiplatelet treatment (DAPT) with aspirin and a P2Y12 receptor antagonist is the standard antithrombotic strategy in acute coronary syndrome (ACS) patients undergoing percutaneous coronary intervention (PCI), as well as those not undergoing PCI [1]. Prasugrel is an investigational thienopyridine prodrug that is metabolized into an active metabolite [1,2,3]. Ticagrelor is a reversible direct-acting oral antagonist of the adenosine diphosphate receptor P2Y12, providing more consistent P2Y12 inhibition [4]. Compared to clopidogrel, the newer P2Y12 inhibitors are associated with a significantly lower incidence of ischemic events without an increased risk of major bleeding [5]. In the TRITON-TIMI 38 and PLATO trials, prasugrel and ticagrelor significantly reduced ischemic events, particularly one month after PCI when compared to clopidogrel [6,7]. However, the rate of bleeding events in East Asian patients appears to be higher than in the western population for standard doses of the novel potent P2Y12 inhibitors [8]. With the East Asian Paradox hypothesis, we are trying to determine an optimal dose as with prasugrel [9]. Ticagrelor 45 mg bid might be a good option for patients with low body weight or elderly patients. It is therefore essential for physicians of patients of East Asian ethnicity to select the optimal treatment, which can include reduced doses of prasugrel/ticagrelor or de-escalation to clopidogrel. The present study sought to compare standard and reduced doses of prasugrel and ticagrelor with clopidogrel in the maintenance phase of PCI in Korean ACS patients. We hypothesized that ticagrelor would elicit greater platelet inhibition than prasugrel and clopidogrel.

2. Materials and Methods

2.1. Study Design and Population

This study was a platelet function sub-analysis of the HOPE–TAILOR trial (Half dose of Prasugrel and Ticagrelor in Platelet Response to Acute Coronary Syndrome; URL: Available online: https://www.clinicaltrials.gov (accessed on 24 October 2016); Unique identifier: NCT02944123) [10].
For inclusion criteria, we consecutively screened patients aged ≥18 years on DAPT with aspirin and a P2Y12 inhibitor, having received a loading dose (60 mg of prasugrel, 180 mg of ticagrelor or 300–600 mg of clopidogrel) during index admission or at the time of PCI. All patients were discharged with prescriptions for one-month standard-dose DAPT treatment: 100 mg once daily aspirin plus prasugrel 10 mg once daily, 100 mg once daily aspirin plus ticagrelor 90 mg twice daily or 100 mg once daily aspirin plus clopidogrel 75 mg once daily, followed by half-dose prasugrel 5 mg once daily or ticagrelor 45 mg twice daily for maintenance treatment, but with maintenance of the clopidogrel dose without reduction.
The exclusion criteria were as follows: (1) low body weight (<60 kg); (2) use of glycoprotein IIb/IIIa inhibitors tirofiban or eptifibatide within 24 h before or during PCI or abciximab within 10 days before or during PCI; (3) daily treatment with nonsteroidal anti-inflammatory drugs; (4) contraindication to aspirin or any of the study drugs; (5) current treatment with drugs interfering with CYP3A4 metabolism; (6) a history of stroke or transient ischemic attack; (7) gastrointestinal bleeding within the last 6 months, bleeding diathesis, platelet count <100,000/mm3; (8) known chronic renal insufficiency or hepatic dysfunction; or (9) known severe chronic obstructive pulmonary disease or bradycardia.

2.2. Blood Sampling

Venous blood samples for platelet function tests were obtained at two timepoints: 30 ± 7 days and 90 ± 7 days after PCI. After confirming that the patient was ingesting the study drugs, blood samples collected in the morning or afternoon and were processed by trained laboratory physicians who did not have access to the results. Platelet function assessments included VerifyNow (VN, Accumetrics, San Diego, CA, USA), multiple electrode aggregometry (MEA, Dynabyte Medical, Munich, Germany) and light transmittance aggregometry (LTA, Chrono-Log, Havertown, PA, USA).

2.3. Platelet Function Measurements

2.3.1. VerifyNow

The platelet function test was performed on an outpatient basis at clinic times between 9:00–12:00 and 13:30–16:30 after confirming administration of the antiplatelet drug administration. The VN assay is a point-of-care (POC), turbidimetric assay that measures platelet function and was used according to manufacturer’s instructions [11]. Within the cartridge of the VerifyNow P2Y12 assay is a channel that measures inhibition of the adenosine diphosphate (ADP) P2Y12 receptor. This channel contains ADP as a platelet agonist and prostaglandin E1 as a suppressor of intracellular-free calcium levels to reduce the nonspecific contribution of ADP binding to P2Y12 receptors. The VN results are expressed in P2Y12 reaction units (PRU). The cutoff values used to define OPR were between 85 to 208 PRUs [12].

2.3.2. Multiplate Analyzer

MEA is a semi-automated POC system, assessing platelet reactivity in whole blood, and was used according to the manufacturer’s instructions [13]. The output values are expressed in arbitrary aggregation units (AU). The cut-off values used to define OPR were between 19 to 46 AU [14]. MEA was performed with a Multiplate Analyzer (Dynabyte Medical, Munich, Germany). Specifically, the adhesion and aggregation of platelets on sensor surfaces enhances the electrical resistance between two sensor electrodes. We used an ADP test (6.4 µM ADP) to monitor the antiplatelet effects of DAPT, mostly targeting clopidogrel. In the test cuvette, whole blood (300 µL) was diluted (1:2 vol/vol) with 0.9% NaCl solution for 6.4 µM, and ADP was stirred in for 3 min at 37 °C, before ADP in the absence of PGE1 was added, and the increase in electrical impedance was recorded continuously for 6 min and converted into arbitrary aggregation units (AU). Approximately 8 AU corresponds to 1 Ohm. The means of the two independent determinations were expressed as the area under the curve of aggregation tracing (AUC) in AU· min. The manufacturer recommends the use of arbitrary units (U) to simplify the expression of results (1 U = 10 AU · min = 1 AUC).

2.3.3. Light Transmittance Aggregometry

According to the LTA standard protocol, blood samples were drawn into a 3.2% sodium citrate-containing tube (Greiner Bio-One GmbH, Frickenhausen, Germany) and processed within 2 h [15]. Platelet-rich plasma was prepared by centrifugation at 120× g for 10 min. After the collection of platelet-rich plasma, platelet-poor plasma was obtained from the remaining specimen by recentrifugation at 1200× g for 10 min. The platelet-rich plasma was then adjusted to a platelet count of 250,000 per ml by adding platelet-poor plasma as needed. Light transmission was calibrated by a cuvette with platelet-rich plasma which was normalized as 0% and a second cuvette containing platelet-poor plasma that was normalized as 100%. Platelet function was measured after the addition of 10 mL adenosine diphosphate (ADP), before the curves were recorded for 6 min. The results are expressed as maximum platelet aggregation (MPA) within 6 min. At present, OPR measurements by LTA remain controversial. LTA was used to compare the other two assays.

2.4. The Definitions of OPR, HPR and LPR

High platelet reactivity (HPR) was defined as PRU > 208, while low platelet reactivity (LPR) was defined as PRU < 85 assessed by VN [16]. High platelet reactivity (HPR) was defined as AU > 46, while low platelet reactivity (LPR) was defined as AU < 19 assessed by MEA [12].

2.5. Statistical Analysis

For baseline characteristics, categorical variables are expressed as frequencies and percentages, and continuous variables as mean ± standard deviation. If application conditions were not fulfilled, the qualitative variables were compared using the chi-square test or Fisher’s exact test. The P2Y12 antagonist quantitative variables were compared with the ANOVA method between groups. The Pearson method was used to correlate antiplatelet effects measured by the two assays. To perform simple linear regression analysis, we first confirmed whether the data satisfied assumptions of linearity, independence, normality and homoscedasticity. For the clinical outcomes, we analyzed six-month major adverse cardiovascular events (MACE) and bleeding events using the Kaplan–Meier method, with a log-rank p-value. The hazard ratios (HRs) and 95% confidence intervals (CIs) were calculated with Cox proportional hazard methods comparing the three groups. Landmark analyses in the overall population and in prespecified subgroups were conducted, setting the landmark point at one month. p values < 0.05 were considered statistically significant for all comparisons using SPSS software version 22.0 (SPSS Inc., Chicago, IL, USA). A comparison of platelet reactive responses to prasugrel, ticagrelor and clopidogrel was undertaken with GraphPad Prism software (version 8.0.1; GraphPad, Inc., San Diego, CA, USA). Three groups of optimal platelet reactivity variation Sankey diagrams were generated using Origin software (version 2020; OriginLab, Inc., Northampton, UK).

3. Results

3.1. The Baseline Characteristics of Study Patients

From December 2016, through November 2018, 118 patients were enrolled in the analysis (losses were due to patients’ infeasibility to visit at the scheduled times or failure to obtain a suitable blood sample to measure platelet function), of which 40 (33.9%), 38 (32.2%) and 40 (33.9%) patients were treated with clopidogrel, prasugrel and ticagrelor, respectively. Table 1 summarizes the baseline demographics, and procedural and angiographic results for the population treated with the P2Y12 antagonist. Baseline results were slightly different between the groups. The clopidogrel-treated patients were slightly older on average than the other groups. More male patients were enrolled. Unstable angina was more frequent among the clopidogrel-treated patients, and ST-segment elevation myocardial infarction (STEMI) was more frequent in the ticagrelor-treated group. There was no significant difference in post PCI base values among the three groups.

3.2. Pharmacodynamic Effect of Standard- and Reduced-Dose New P2Y12 Inhibitors

At the one-month period, patients receiving standard-dose prasugrel or ticagrelor had lower platelet reactivity as determined by the three assays than those receiving the conventional dose of clopidogrel (VN: 41.8 ± 41.5 vs. 20.0 ± 25.3 vs. 161.8 ± 68.9, p = 0.000; MEA: 12.7 ± 5.9 vs. 15.6 ± 5.6 vs. 22.1 ± 11.8, p = 0.000; LTA: 6.2 ± 10.1 vs. 2.9 ± 4.9 vs. 13.4 ± 15.6, p = 0.000, Figure 1, Table 2). Compared to ticagrelor, there was significantly less platelet inhibition achieved with prasugrel determined by VN (p = 0.007), but the opposite result was obtained when determined by MEA (p = 0.031), and no change was shown with LTA (p = 0.080). At the 3-month point, platelet reactivity was lower in those receiving prasugrel or ticagrelor than the clopidogrel-treated patients (VN: 94.8 ± 57.0 vs. 31.0 ± 34.5 vs. 156.8 ± 66.1, p = 0.000; MEA: 16.8 ± 5.5 vs. 17.4 ± 4.7 vs. 21.9 ± 12.0, p = 0.012; LTA: 10.3 ± 9.4 vs. 4.1 ± 8.2 vs. 12.2 ± 13.4, p = 0.002). Prasugrel resulted in significantly less platelet inhibition than ticagrelor as determined by VN and LTA (VN: p = 0.000; LTA: p = 0.003). However, no difference in findings was determined when measured with MEA (p = 0.638). Prasugrel and clopidogrel had similar platelet inhibition at the three-month timepoint (p = 0.470).
At one month, OPR rates were markedly lower with both prasugrel and ticagrelor compared to clopidogrel as determined by VN (21.1%:0.0%:57.5%; p = 0.001); OPR rates for prasugrel and ticagrelor were prominently lower compared to clopidogrel determined by MEA (Table 3). After 3 months, OPR rates for prasugrel and clopidogrel were similar, and both were markedly higher compared to ticagrelor determined by VN, but there were no significant differences among the three groups determined with MEA. At 3 months, there was a trend towards increasing OPR with ticagrelor (0.0% to 12.5%, p = 0.021) as well as prasugrel (21.1% to 50.0%, p = 0.008), whereas OPR rates with clopidogrel remained higher (57.5% to 65.0%, p = 0.491) as determined by VN (Table 3, Figure 2). With MEA, there was a different trend towards ticagrelor from 1 month to 3 months (30.0% to 47.5%, p = 0.105) but a significantly higher OPR rate with prasugrel (15.8% to 42.1%, p = 0.020). OPR rates for all platelet function measurements are shown in Table 3 and Figure 2. The Sankey diagram depicts the 13 patients (34.2%) in the prasugrel group that changed from low platelet reactivity (LPR) to OPR with VN; with 11 patients (28.9%) in the prasugrel group and 10 patients (25.0%) in the ticagrelor group changing from LPR to OPR with MEA (Figure 2). According to the two platelet function tests, both prasugrel and ticagrelor showed strong platelet function inhibition. There were no HPR patients in the new P2Y12 inhibitor groups at 1 month as well as 3 months, and most patients were showing LPR at 1 month.

3.3. Comparison of All Platelet Function Tests

VN values correlated weakly with MEA (r = 0.334, p = 0.000). Regardless of the administration of any P2Y12 inhibitors, the correlation between the measurements appears to be determined by the level of platelet reactivity. In terms of HPR determined by VN, non-HPR patients determined by MEA appeared in relatively greater in numbers. VN and LTA showed a moderate correlation (r = 0.582, p = 0.000), while MEA and LTA showed a lower correlation (r = 0.357, p = 0.000) (Figure 3).

4. Discussion

The findings of this pharmacodynamic investigation can be summarized as follows: (1) higher rates of OPR were observed among clopidogrel-treated patients compared to standard-dose prasugrel or ticagrelor using both platelet function assessments at one month; (2) OPR in the reduced-dose prasugrel and ticagrelor groups was not significantly different versus clopidogrel as determined by MEA assay; (3) compared with standard-dose, reduced-dose prasugrel-treated patients’ OPR rate significantly increased with VN and MEA, and ticagrelor-treated patients’ OPR rate significantly increased with VN; (4) prasugrel and ticagrelor had very low platelet function values compared to clopidogrel at one month and three months according to both platelet function assessments (VN and MEA); (5) prasugrel-treated patients’ platelet reactivity was significantly reduced from one month to three months with both platelet function assessments; (6) the MEA assay might have less sensitivity and consistency than the VN assay. Alternatively, the VN test could be overestimating platelet function relative to the MEA or LTA assay, especially in conditions of strong platelet inhibition.

4.1. Importance of OPR in Clinical Practice

Aradi et al., reported that patients with HPR have a 2.7-fold higher risk for ST and a 1.5-fold higher risk for mortality. Meanwhile, patients with LPR have a 1.7-fold higher risk of major bleeding complications compared to those with OPR following PCI, and OPR is therefore associated with net clinical benefits [14].
Our findings show that the OPR rate increased significantly in the prasugrel group and ticagrelor group at 3 months as determined by VN, but when assessed by MEA, the OPR rate was only significantly increased in the prasugrel group. The sub-analysis results from the HOST–REDUCE–POYLTECH–ACS trial showed the percentage of OPR was significantly higher in the de-escalation group (= prasugrel 5 mg) compared to the full dose prasugrel group (61.7 vs.31.7%, p < 0.001) and the de-escalation groups had a lower risk of net clinical outcomes [17]. Lee et al., reported that the proportion of patients with OPR was the highest in the 5 mg prasugrel group compared to the conventional-dose prasugrel or ticagrelor groups (90.0% vs. 46.2% vs. 12.5%, p < 0.001), similar to our findings [18]. The PRAISE–GENE trial demonstrated that half-dose prasugrel achieved significantly lower PRU values in the peri-procedural period, but there were no statistically significant differences in terms of OPR at 30 days to the clopidogrel group [19]. The A-MATCH trial reported that the 5-mg prasugrel group was more likely to meet normal platelet reactivity (NPR) criteria determined by VN with respect to East Asian criteria for type 2 bleeding. The proportion of NPR was significantly increased in the 5-mg compared to the 10-mg group, and there was a lower incidence of adverse events observed in the NPR group [20]. In addition, the OPR rate in the prasugrel 3.75 mg group was higher than that of the ticagrelor 60 mg group. Compared with the clopidogrel group, the low-dose prasugrel group was associated with a lower incidence of MACE and clinically serious bleeding in Japanese ACS patients [21,22]. Kim et al., reported that compared with standard-dose ticagrelor, half-dose ticagrelor reduced serious bleeding events during the early period of dual-antiplatelet therapy in ACS patients with LPR status determined by VN [23].
The pro-drug clopidogrel is characterized by significant response variability and a substantial proportion of patients exhibit high on-treatment platelet reactivity (HPR). Hence, PFT could serve to safeguard DAPT de-escalation by identifying HPR patients on clopidogrel, as those patients may be exposed to a higher risk of thrombotic events due to insufficient P2Y12 inhibition [24]. In the present study, prasugrel caused a dose-dependent decrease in platelet inhibition from 10 to 5 mg as determined by MEA, while both doses of ticagrelor showed no difference between the 45 and 90 mg twice-daily doses. In contrast, VN showed a dose-dependent increase in platelet reactivity for both prasugrel and ticagrelor. Jin et al., reported that the prasugrel (5 mg) group had a significantly lower average PRU value compared with the 75 mg clopidogrel group at 30 days, while the OPR rate was similar between the 5 mg prasugrel and 75 mg clopidogrel groups [25]. The rate of high on-treatment platelet reactivity (PRU > 235) was significantly lower in the 5 mg prasugrel group than in the 75 mg clopidogrel group. This study demonstrated that 5 mg prasugrel is associated with greater platelet inhibition and lower rates of HPR compared with standard-dose clopidogrel in PCI patients < 75 years old and weighing ≥ 60 kg.

4.2. Differences between the 3 Platelet Function Tests

Studies have demonstrated a close link between platelet reactivity during treatment assessed by platelet function tests and clinical outcomes after PCI. We compared standard and reduced doses of prasugrel and ticagrelor with clopidogrel in the maintenance phase of PCI in Korean ACS patients. For STEMI patients, new P2Y12 inhibitors are recommended during DAPT treatment after coronary stent implantation and more STEMI patients used new P2Y12 inhibitors in this trial [3]. Although prasugrel and ticagrelor are superior to clopidogrel for reducing ischemic events in ACS patients [6,7], clopidogrel is still widely used in clinical practice due to its reduced bleeding risk [26]. Our results confirm that prasugrel and ticagrelor elicit significantly higher platelet inhibition than clopidogrel at both standard and reduced doses with the VN and MEA assays. Compared to prasugrel, ticagrelor was a more potent inhibitor as determined by VN, a finding consistent with several prior pharmacodynamics studies [27,28,29]. The correlation among three PFT assays from our results showed that LTA and VN were more highly correlated than MEA. Zhang et al. reported a moderate correlation between LTA and VN before or after PCI, while the correlation between MEA and VN measurements was low [30], which was similar to our findings.
The results for the previously published HOPE–TAILOR trial showed ticagrelor had a lower OPR rate and greater platelet inhibition compared to prasugrel as well as conventional-dose clopidogrel at 1 and 3 months as assessed by VN [10]. The present study included differences between the three methods for measuring platelet reactivity. Contrary to the VN results from the HOPE–TAILOR trial, prasugrel had greater platelet inhibition compared to ticagrelor (and clopidogrel) at 1 and 3 months, as well as a higher OPR rate at 1 month, showing the opposite to the MEA data.
In a study by Alexopoulos et al., significantly lower platelet reactivity was found in patients receiving ticagrelor treatment compared to prasugrel [27]. In the study, platelet function was assessed with ticagrelor (90 mg twice daily) or prasugrel standard-dose at 15 and 30 days after a therapeutic switch to ticagrelor or prasugrel using VN in ACS patients with high on-treatment platelet reactivity on clopidogrel. These results mirror our own findings in terms of the data for testing by VN. However, the prasugrel group showed significantly lower platelet reactivity than for ticagrelor as determined by MEA when using the standard-dose, although there was no difference with reduced doses. The results are also consistent with the findings by Schnorbus et al. Further studies are warranted to confirm this discrepancy [31].
The accurate identification of post-MI patients who might gain further benefit from prolonged DAPT with low-dose ticagrelor remains essential, considering the importance of balancing bleeding and ischemic risk in vulnerable patients [32]. In addition, the identification of PEGASUS and COMPASS phenotypes at baseline based on drug eligibility criteria may help in selecting patients at higher risk of ischemic events who could benefit from more intense treatment [33]. Therefore, treatment de-escalation should be determined on a case-by-case basis.
We acknowledge several limitations to our findings. VN was a standardized measurement, while LTA was measured with two different machines, so consistency between the two results cannot be guaranteed. Secondly, blood sampling was performed in the outpatient clinic, and the exact sampling times will have differed, although it was confirmed that each patient had ingested the medicine (so medication-related differences are addressed). Thirdly, although it was a subgroup analysis of the HOPE–TAILOR study, some patients lacked MEA and LTA data, so there were some differences in baseline characteristics after excluding missing values. Additionally, one of the limitations is the absence of data in terms of long-term thrombotic and hemorrhagic complications. Finally, the small number of patients is a critical limitation of this study, and the longitudinal outcome data have been described previously [10].

5. Conclusions

OPR values for reduced-dose prasugrel and conventional-dose clopidogrel at three months were similar but higher than for reduced-dose ticagrelor determined by VN. No differences were noted when assessed by MEA. The MEA assay may be less sensitive and consistent than the VN assay. Further studies are warranted to confirm this discrepancy.

Author Contributions

Conceptualization, M.-H.K.; methodology, M.-H.K., K.S. and X.J.; software, K.S. and K.-M.L.; validation, M.-H.K. and X.J.; formal analysis, M.-H.K., K.S. and C.-D.J.; investigation, S.-L.Y., K.S., J.-X.L. and Z.-Y.S.; resources, M.-H.K.; data curation, K.-H.L. and E.-Z.J.; writing—original draft preparation, K.S. and X.J.; writing—review and editing, M.-H.K. and X.J.; visualization, K.S., X.J.; supervision, M.-H.K.; project administration, M.-H.K.; funding acquisition, M.-H.K. and Y.-R.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by the Korean Society of Cardiology (201605-02).

Institutional Review Board Statement

The study was conducted according to the guidelines of the Declaration of Helsinki, and approved by the Institutional Ethics Committee of Dong-A University Hospital (protocol code: 16-162, approval: September 2016). And the HOPE–TAILOR trial has been previously registered at https://www.clinicaltrials.gov (NCT02944123) (accessed on 10 March 2022).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study. Written informed consent has been obtained from the patients to publish this paper.

Data Availability Statement

Data are the property of the authors and can become available by contacting the corresponding author.

Acknowledgments

The authors also thank Ju Yeong Lee and Jae-eun Kim for technical assistance with the platelet function testing measurements.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

TermsDefinition
ACSAcute coronary syndrome
ADPAdenosine diphosphate
AUAggregation units
BARCBleeding academic research consortium
CDConventional dose
CIsConfidence interval
DAPTDual antiplatelet treatment
F/UFollow up
HPRHigh platelet reactivity
HRsHazard ratios
K-MKaplan-Meier
LPRLow platelet reactivity
LTALight transmittance aggregometry
MACEMajor adverse cardiovascular events
MACCEMajor adverse cardiac and cerebrovascular events
MEAMultiple electrode aggregometry
MIMyocardial infarction
MPAMaximum platelet aggregation
OPROptimal platelet reactivity
PCIPercutaneous coronary intervention
PGE1Prostaglandin E1
POCPoint-of-care
PRUP2Y12 reaction units
STEMIST-segment elevation myocardial infarction
TLRTarget lesion revascularization
VNVerifyNow

References

  1. Neumann, F.J.; Sousa-Uva, M.; Ahlsson, A.; Alfonso, F.; Banning, A.P.; Benedetto, U.; Byrne, R.A.; Collet, J.P.; Falk, V.; Head, S.J.; et al. 2018 ESC/EACTS Guidelines on myocardial revascularization. Eur. Heart J. 2019, 40, 87–165. [Google Scholar] [CrossRef] [PubMed]
  2. Capodanno, D.; Alfonso, F.; Levine, G.N.; Valgimigli, M.; Angiolillo, D.J. ACC/AHA Versus ESC Guidelines on Dual Antiplatelet Therapy: JACC Guideline Comparison. J. Am. Coll. Cardiol. 2018, 72, 2915–2931. [Google Scholar] [CrossRef] [PubMed]
  3. Levine, G.N.; Bates, E.R.; Bittl, J.A.; Brindis, R.G.; Fihn, S.D.; Fleisher, L.A.; Granger, C.B.; Lange, R.A.; Mack, M.J.; Mauri, L.; 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: An Update of the 2011 ACCF/AHA/SCAI Guideline for Percutaneous Coronary Intervention, 2011 ACCF/AHA Guideline for Coronary Artery Bypass Graft Surgery, 2012 ACC/AHA/ACP/AATS/PCNA/SCAI/STS Guideline for the Diagnosis and Management of Patients With Stable Ischemic Heart Disease, 2013 ACCF/AHA Guideline for the Management of ST-Elevation Myocardial Infarction, 2014 AHA/ACC Guideline for the Management of Patients With Non-ST-Elevation Acute Coronary Syndromes, and 2014 ACC/AHA Guideline on Perioperative Cardiovascular Evaluation and Management of Patients Undergoing Noncardiac Surgery. Circulation 2016, 134, e123–e155. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Levine, G.N.; Jeong, Y.H.; Goto, S.; Anderson, J.L.; Huo, Y.; Mega, J.L.; Taubert, K.; Smith, S.C., Jr. Expert consensus document: World Heart Federation expert consensus statement on antiplatelet therapy in East Asian patients with ACS or undergoing PCI. Nat. Rev. Cardiol. 2014, 11, 597–606. [Google Scholar] [CrossRef]
  5. Wallentin, L.; Becker, R.C.; Budaj, A.; Cannon, C.P.; Emanuelsson, H.; Held, C.; Horrow, J.; Husted, S.; James, S.; Katus, H.; et al. Ticagrelor versus Clopidogrel in Patients with Acute Coronary Syndromes. N. Engl. J. Med. 2009, 361, 1045–1057. [Google Scholar] [CrossRef]
  6. James, S.; Angiolillo, D.J.; Cornel, J.H.; Erlinge, D.; Husted, S.; Kontny, F.; Maya, J.; Nicolau, J.C.; Spinar, J.; Storey, R.F.; et al. Ticagrelor vs. clopidogrel in patients with acute coronary syndromes and diabetes: A substudy from the PLATelet inhibition and patient Outcomes (PLATO) trial. Eur. Heart J. 2010, 31, 3006–3016. [Google Scholar] [CrossRef]
  7. Montalescot, G.; Wiviott, S.D.; Braunwald, E.; Murphy, S.A.; Gibson, C.M.; McCabe, C.H.; Antman, E.M. Prasugrel compared with clopidogrel in patients undergoing percutaneous coronary intervention for ST-elevation myocardial infarction (TRITON-TIMI 38): Double-blind, randomised controlled trial. Lancet 2009, 373, 723–731. [Google Scholar] [CrossRef]
  8. Yun, J.E.; Kim, Y.J.; Park, J.J.; Kim, S.; Park, K.; Cho, M.S.; Nam, G.B.; Park, D.W. Safety and Effectiveness of Contemporary P2Y12 Inhibitors in an East Asian Population With Acute Coronary Syndrome: A Nationwide Population-Based Cohort Study. J. Am. Heart Assoc. 2019, 8, e012078. [Google Scholar] [CrossRef] [Green Version]
  9. Kim, H.K.; Tantry, U.S.; Smith, S.C., Jr.; Jeong, M.H.; Park, S.J.; Kim, M.H.; Lim, D.S.; Shin, E.S.; Park, D.W.; Huo, Y.; et al. The East Asian Paradox: An Updated Position Statement on the Challenges to the Current Antithrombotic Strategy in Patients with Cardiovascular Disease. Thromb. Haemost. 2021, 121, 422–432. [Google Scholar] [CrossRef]
  10. Jin, C.D.; Kim, M.H.; Song, K.; Jin, X.; Lee, K.M.; Park, J.S.; Cho, Y.R.; Yun, S.C.; Lee, M.S. Pharmacodynamics and Outcomes of a De-Escalation Strategy with Half-Dose Prasugrel or Ticagrelor in East Asians Patients with Acute Coronary Syndrome: Results from HOPE-TAILOR Trial. J. Clin. Med. 2021, 10, 2699. [Google Scholar] [CrossRef]
  11. Malinin, A.; Pokov, A.; Spergling, M.; Defranco, A.; Schwartz, K.; Schwartz, D.; Mahmud, E.; Atar, D.; Serebruany, V. Monitoring platelet inhibition after clopidogrel with the VerifyNow-P2Y12(R) rapid analyzer: The VERIfy Thrombosis risk ASsessment (VERITAS) study. Thromb. Res. 2007, 119, 277–284. [Google Scholar] [CrossRef] [PubMed]
  12. Tantry, U.S.; Bonello, L.; Aradi, D.; Price, M.J.; Jeong, Y.H.; Angiolillo, D.J.; Stone, G.W.; Curzen, N.; Geisler, T.; Ten Berg, J.; 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, 2261–2273. [Google Scholar] [CrossRef] [PubMed]
  13. Siess, W.; Losonczy, H.; Penz, S.; Calatzis, A.; Tóth, O. Multiple electrode aggregometry: A new device to measure platelet aggregation in whole blood. Thromb. Haemost. 2017, 96, 781–788. [Google Scholar] [CrossRef]
  14. Aradi, D.; Kirtane, A.; Bonello, L.; Gurbel, P.A.; Tantry, U.S.; Huber, K.; Freynhofer, M.K.; ten Berg, J.; Janssen, P.; Angiolillo, D.J.; et al. Bleeding and stent thrombosis on P2Y12-inhibitors: Collaborative analysis on the role of platelet reactivity for risk stratification after percutaneous coronary intervention. Eur. Heart J. 2015, 36, 1762–1771. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Breet, N.J.; van Werkum, J.W.; Bouman, H.J.; Kelder, J.C.; Ruven, H.J.; Bal, E.T.; Deneer, V.H.; Harmsze, A.M.; van der Heyden, J.A.; Rensing, B.J.; et al. Comparison of platelet function tests in predicting clinical outcome in patients undergoing coronary stent implantation. JAMA 2010, 303, 754–762. [Google Scholar] [CrossRef]
  16. Vries, M.J.; Bouman, H.J.; Olie, R.H.; Veenstra, L.F.; Zwaveling, S.; Verhezen, P.W.; Ten Cate-Hoek, A.J.; Ten Cate, H.; Henskens, Y.M.; van der Meijden, P.E. Determinants of agreement between proposed therapeutic windows of platelet function tests in vulnerable patients. Eur. Heart J. Cardiovasc. Pharm. 2017, 3, 11–17. [Google Scholar] [CrossRef]
  17. Lee, H.S.; Park, K.W.; Kang, J.; Han, J.K.; Kim, H.S. De-escalation of Prasugrel Results in Higher Percentage of Patients within Optimal Range of Platelet Reactivity: Analysis from the HOST-REDUCE-POLYTECH-ACS Trial. Thromb. Haemost. 2022, 122, 160–162. [Google Scholar] [CrossRef]
  18. Lee, J.H.; Ahn, S.G.; Park, B.; Park, S.W.; Kang, Y.S.; Lee, J.W.; Youn, Y.J.; Ahn, M.S.; Kim, J.Y.; Yoo, B.S.; et al. A pharmacodynamic study of the optimal P2Y12 inhibitor regimen for East Asian patients with acute coronary syndrome. Korean J. Intern. Med. 2015, 30, 620–628. [Google Scholar] [CrossRef]
  19. Jin, C.; Kim, M.H.; Guo, L.Z.; Jin, E.; Shin, E.S.; Ann, S.H.; Cho, Y.R.; Park, J.S.; Kim, S.J.; Lee, M.S. Pharmacodynamic study of prasugrel or clopidogrel in non-ST-elevation acute coronary syndrome with CYP2C19 genetic variants undergoing percutaneous coronary intervention (PRAISE-GENE trial). Int. J. Cardiol. 2020, 305, 11–17. [Google Scholar] [CrossRef]
  20. Jeong, Y.H.; Oh, J.H.; Yoon, H.J.; Park, Y.; Suh, J.; Lee, S.W.; Lee, K.; Kim, J.S.; Chun, W.J.; Park, Y.H.; et al. Pharmacodynamic Profile and Prevalence of Bleeding Episode in East Asian Patients with Acute Coronary Syndromes Treated with Prasugrel Standard-Dose versus De-escalation Strategy: A Randomized A-MATCH Trial. Thromb. Haemost. 2021, 121, 1376–1386. [Google Scholar] [CrossRef]
  21. Saito, S.; Isshiki, T.; Kimura, T.; Ogawa, H.; Yokoi, H.; Nanto, S.; Takayama, M.; Kitagawa, K.; Nishikawa, M.; Miyazaki, S.; et al. Efficacy and safety of adjusted-dose prasugrel compared with clopidogrel in Japanese patients with acute coronary syndrome: The PRASFIT-ACS study. Circ. J. 2014, 78, 1684–1692. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Tateishi, K.; Saito, Y.; Kitahara, H.; Nakayama, T.; Fujimoto, Y.; Kobayashi, Y. Increased platelet inhibition after switching from prasugrel to low-dose ticagrelor in Japanese patients with prior myocardial infarction. J. Cardiol. 2020, 75, 473–477. [Google Scholar] [CrossRef] [PubMed]
  23. Kim, L.; Choe, J.C.; Ahn, J.H.; Lee, H.W.; Oh, J.H.; Choi, J.H.; Lee, H.C.; Cha, K.S.; Hong, T.J.; Jeong, Y.H.; et al. Temporal Trends of Bleeding Episodes during Half- vs. Standard-Dose Ticagrelor in Acute Coronary Syndrome Patients with Low Platelet Reactivity: A Randomized BLEEDING-ACS Trial. J. Clin. Med. 2021, 10, 1159. [Google Scholar] [CrossRef] [PubMed]
  24. Kupka, D.; Sibbing, D. De-Escalation of P2Y12 Receptor Inhibitor Therapy after Acute Coronary Syndromes in Patients Undergoing Percutaneous Coronary Intervention. Korean Circ. J. 2018, 48, 863–872. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Jin, H.Y.; Yang, T.H.; Choi, K.N.; Seo, J.S.; Jang, J.S.; Kim, D.K.; Kim, D.S. Randomized Comparison of the Platelet Inhibitory Efficacy between Low Dose Prasugrel and Standard Dose Clopidogrel in Patients Who Underwent Percutaneous Coronary Intervention. Korean Circ. J. 2014, 44, 82–88. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Joo, H.J.; Ahn, S.G.; Park, J.H.; Park, J.Y.; Hong, S.J.; Kim, S.Y.; Choi, W.; Gwon, H.; Lim, Y.H.; Kim, W.; et al. Effects of genetic variants on platelet reactivity and one-year clinical outcomes after percutaneous coronary intervention: A prospective multicentre registry study. Sci. Rep. 2018, 8, 1229. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Alexopoulos, D.; Stavrou, K.; Koniari, I.; Gkizas, V.; Perperis, A.; Kontoprias, K.; Vogiatzi, C.; Bampouri, T.; Xanthopoulou, I. Ticagrelor vs. prasugrel one-month maintenance therapy: Impact on platelet reactivity and bleeding events. Thromb. Haemost. 2014, 112, 551–557. [Google Scholar] [CrossRef] [Green Version]
  28. Ferreiro, J.L.; Vivas, D.; De La Hera, J.M.; Marcano, A.L.; Lugo, L.M.; Gomez-Polo, J.C.; Silva, I.; Tello-Montoliu, A.; Marin, F.; Roldan, I. High and low on-treatment platelet reactivity to P2Y12 inhibitors in a contemporary cohort of acute coronary syndrome patients undergoing percutaneous coronary intervention. Thromb. Res. 2019, 175, 95–101. [Google Scholar] [CrossRef]
  29. Franchi, F.; Rollini, F.; Aggarwal, N.; Hu, J.; Kureti, M.; Durairaj, A.; Duarte, V.E.; Cho, J.R.; Been, L.; Zenni, M.M.; et al. Pharmacodynamic Comparison of Prasugrel Versus Ticagrelor in Patients With Type 2 Diabetes Mellitus and Coronary Artery Disease: The OPTIMUS (Optimizing Antiplatelet Therapy in Diabetes Mellitus)-4 Study. Circulation 2016, 134, 780–792. [Google Scholar] [CrossRef]
  30. Zhang, H.Z.; Kim, M.H.; Jeong, Y.H. Predictive values of post-clopidogrel platelet reactivity assessed by different platelet function tests on ischemic events in East Asian patients treated with PCI. Platelets 2014, 25, 292–299. [Google Scholar] [CrossRef]
  31. Schnorbus, B.; Daiber, A.; Jurk, K.; Warnke, S.; Koenig, J.; Lackner, K.J.; Munzel, T.; Gori, T. Effects of clopidogrel vs. prasugrel vs. ticagrelor on endothelial function, inflammatory parameters, and platelet function in patients with acute coronary syndrome undergoing coronary artery stenting: A randomized, blinded, parallel study. Eur. Heart J. 2020, 41, 3144–3152. [Google Scholar] [CrossRef] [PubMed]
  32. Cesaro, A.; Taglialatela, V.; Gragnano, F.; Moscarella, E.; Fimiani, F.; Conte, M.; Barletta, V.; Monda, E.; Limongelli, G.; Severino, S.; et al. Low-Dose Ticagrelor in Patients With High Ischemic Risk and Previous Myocardial Infarction: A Multicenter Prospective Real-World Observational Study. J. Cardiovasc. Pharmacol. 2020, 76, 173–180. [Google Scholar] [CrossRef] [PubMed]
  33. Cesaro, A.; Gragnano, F.; Calabro, P.; Moscarella, E.; Santelli, F.; Fimiani, F.; Patti, G.; Cavallari, I.; Antonucci, E.; Cirillo, P.; et al. Prevalence and clinical implications of eligibility criteria for prolonged dual antithrombotic therapy in patients with PEGASUS and COMPASS phenotypes: Insights from the START-ANTIPLATELET registry. Int. J. Cardiol. 2021, 345, 7–13. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Differences in platelet aggregation inhibition between 1-month and 3-month periods as determined by three different assays ((A): VerifyNow; (B): MEA; (C): LTA). Dotted area represents OPR. PRUs: P2Y12 reaction units; AU: aggregation units; OPR: optimal platelet reactivity.
Figure 1. Differences in platelet aggregation inhibition between 1-month and 3-month periods as determined by three different assays ((A): VerifyNow; (B): MEA; (C): LTA). Dotted area represents OPR. PRUs: P2Y12 reaction units; AU: aggregation units; OPR: optimal platelet reactivity.
Jcm 11 02480 g001
Figure 2. Optimal platelet reactivity rate change from one-month to three-month periods assessed by VN and MEA. Sankey diagram of the relationship between the standard dosage period (1 month) and reduced dosage period (3 months) in platelet function. Each color of the block on the left represents the bar height proportional to the number of patients in each group. The lines connecting the left and right side indicate the relationship between standard dosage and reduced dosage. (A): with VerifyNow, (B): with MEA. CLO: clopidogrel; PRA: prasugrel; TIC: ticagrelor; HPR: high platelet reactivity; OPR: optimal platelet reactivity; LPR: low platelet reactivity.
Figure 2. Optimal platelet reactivity rate change from one-month to three-month periods assessed by VN and MEA. Sankey diagram of the relationship between the standard dosage period (1 month) and reduced dosage period (3 months) in platelet function. Each color of the block on the left represents the bar height proportional to the number of patients in each group. The lines connecting the left and right side indicate the relationship between standard dosage and reduced dosage. (A): with VerifyNow, (B): with MEA. CLO: clopidogrel; PRA: prasugrel; TIC: ticagrelor; HPR: high platelet reactivity; OPR: optimal platelet reactivity; LPR: low platelet reactivity.
Jcm 11 02480 g002
Figure 3. Correlation between three different platelet function tests ((A). VN and MEA; (B). VN and LTA; (C). MEA and LTA). SD: standard dose; RD: reduced dose; CD: conventional dose; VN: VerifyNow; MEA: multiple electrode aggregometry; LTA: light transmittance aggregometry.
Figure 3. Correlation between three different platelet function tests ((A). VN and MEA; (B). VN and LTA; (C). MEA and LTA). SD: standard dose; RD: reduced dose; CD: conventional dose; VN: VerifyNow; MEA: multiple electrode aggregometry; LTA: light transmittance aggregometry.
Jcm 11 02480 g003
Table 1. The baseline characteristics of the study patients.
Table 1. The baseline characteristics of the study patients.
Total Number
(n = 118)
Prasugrel
(n = 38)
Ticagrelor
(n = 40)
Clopidogrel
(n = 40)
Age, (years)57.7 ± 10.060.8 ± 8.363.0 ± 9.9
Gender, (male), n (%)36 (94.7)34 (85.0)37 (92.5)
BMI, (kg/m2)24.9 ± 2.524.6 ± 2.324.7 ± 2.5
LVEF < 50, n (%)14 (36.8)17 (57.5)16 (40.0)
Multi-vessel disease, n (%)13 (35.1)16 (40.0)17 (42.5)
Risk Factors, n (%)
 Hypertension13 (34.2)21 (52.5)18 (45.0)
 Diabetes6 (15.8)9 (22.5)14 (35.0)
 Dyslipidemia7 (18.4)10 (25.0)5 (12.5)
 Current smoker8 (21.1)9 (22.5)9 (22.5)
Medical History, n (%)
 Previous MI4 (10.5)4 (10.0)6 (15.0)
 Previous PCI4 (10.5)8 (20.0)9 (22.5)
 Previous CABG1 (2.6)2 (5.0)0 (0.0)
 Previous CVA0 (0.0)0 (0.0)0 (0.0)
Clinical Diagnosis, n (%)
 UA7 (18.4)3 (7.5)23 (57.5)
 NSTEMI9 (23.7)10 (25.0)11 (27.5)
 STEMI22 (60.5)27 (67.5)6 (15.0)
Laboratory Index
 CK-MB32.1 ± 43.928.3 ± 31.317.5 ± 14.5
 Hemoglobin (g/dL)14.0 ± 2.013.6 ± 1.713.4 ± 1.8
 Creatinine (g/L)1.0 ± 0.41.0 ± 0.41.2 ± 0.9
 GFR (ml/min/1.73 m2)84.0 ± 23.683.5 ± 22.881.3 ± 25.3
 Platelet count (103/μL)218.7 ± 46.9229.8 ± 66.0216.9 ± 44.6
 Post PCI * Base208.8 ± 51.6218.7 ± 31.9214.6 ± 48.6
 Post PCI PRU57.9 ± 66.545.8 ± 77.0139.7 ± 77.3
 ** Inhibition (%)75.8 ± 25.879.6 ± 35.237.6 ± 26.6
Medication, n (%)
 Statin36 (94.7)38 (95.0)37 (92.5)
 CCB9 (23.7)10 (25.0)11 (27.5)
 β-blocker29 (76.3)30 (75.0)27 (67.5)
 ACEI + ARB9 (23.7)9 (22.5)12 (30.0)
 Diuretic3 (9.1)1 (2.5)2 (5.1)
 Proton-pump inhibitor14 (36.8)21 (52.5)14 (35.0)
Data are presented as number (%). ACEI: angiotensin-converting enzyme inhibitors; ARB: angiotensin receptor blocker; BMI: body mass index; CABG: coronary artery bypass grafting; CCB: calcium channel blocker; CVA: cerebrovascular accident; LVEF: left ventricular ejection fraction; MI: myocardial infarction; PCI: percutaneous coronary intervention; PRU: P2Y12 reaction unit. * Base PRU is the estimated value before obtaining the P2Y12 inhibitor. ** % inhibition = [(Base − Test)/Base] × 100.
Table 2. Platelet function values during standard and reduced-dose periods.
Table 2. Platelet function values during standard and reduced-dose periods.
Prasugrel
(n = 38)
Mean
(95% CI)
Ticagrelor
(n = 40)
Mean
(95% CI)
Clopidogrel
(n = 40)
Mean
(95% CI)
Within-Visit
Comparisons
Overall
Effect
p vs. C
Mean
(95% CI)
p-Value
T vs. p
Mean
(95% CI)
p-Value
T vs. C
Mean
(95% CI)
p-Value
VN1-month41.8 ± 41.5
(28.1–55.5)
20.0 ± 25.3
(11.8–28.1)
161.8 ± 68.9
(139.7–183.9)
0.000120.0
(94.4–145.6)
p = 0.000
21.9
(6.2–37.5)
p = 0.007
141.9
(118.5–165.2)
p = 0.000
3-month94.8 ± 57.0
(76.1–113.6)
31.0 ± 34.5
(19.9–42.0)
156.8 ± 66.1
(135.6–177.9)
0.00061.9
(34.0–89.8)
p = 0.000
63.9
(42.4–85.4)
p = 0.000
125.8
(102.2–149.4)
p = 0.000
* p-value0.0000.1080.740
MEA1-month12.7 ± 5.9
(10.7–14.7)
15.6 ± 5.6
(13.7–17.4)
22.1 ± 11.8
(18.3–25.9)
0.0009.4
(5.2–13.7)
p = 0.000
−2.9
(−5.6–0.3)
p = 0.031
6.5
(2.3–10.7)
p = 0.003
3-month16.8 ± 5.5
(14.9–18.7)
17.4 ± 4.7
(15.9–18.9)
21.9 ± 12.0
(18.1–25.7)
0.0125.1
(0.9–9.3)
p = 0.018
−0.6
(−2.9–1.8)
p = 0.638
4.6
(0.5–8.6)
p = 0.029
* p-value0.0030.1340.948
LTA1-month6.2 ± 10.1
(2.7–9.6)
2.9 ± 4.9
(1.3–4.5)
13.4 ± 15.6
(8.2–18.5)
0.0007.2
(1.1–13.3)
p = 0.021
3.3
(−0.4–6.9)
p = 0.080
10.5
(5.1–15.8)
p = 0.000
3-month10.3 ± 9.4
(7.1–13.5)
4.1 ± 8.2
(1.4–6.7)
12.2 ± 13.4
(7.9–16.6)
0.0021.9
(−3.4–7.3)
p = 0.470
6.3
(2.2–10.3)
p = 0.003
8.2
(3.2–13.2)
p = 0.002
* p-value0.0770.4650.732
* p-value compared between 1-month and 3-month periods; p: prasugrel; C: clopidogrel; T: ticagrelor; VN: VerifyNow; MEA: multiple electrode aggregometry; LTA: light transmittance aggregometry.
Table 3. Platelet reactivity rate during standard- and reduced-dose periods.
Table 3. Platelet reactivity rate during standard- and reduced-dose periods.
Prasugrel
(n = 38)
Ticagrelor
(n = 40)
Clopidogrel
(n = 40)
Overall Effectp vs. C
p-Value
T vs. p
p-Value
T vs. C
p-Value
VN1-monthHPR0 (0.0)0 (0.0)10 (25.0)
OPR8 (21.1)0 (0.0)23 (57.5)0.0000.0010.0000.002
LPR30 (78.9)40 (100.0)7 (17.5)
3-monthHPR0 (0.0)0 (0.0)8 (20.0)
OPR19 (50.0)5 (12.5)26 (65.0)0.0000.1800.0000.000
LPR19 (50.0)35 (87.5)6 (15.0)
* p-value 0.0080.0210.491
MEA1-monthHPR0 (0.0)0 (0.0)3 (7.5)
OPR6 (15.8)12 (30.0)22 (55.0)0.0020.0010.0300.153
LPR32 (84.2)28 (70.0)15 (37.5)
3-monthHPR0 (0.0)0 (0.0)4 (10.0)
OPR16 (42.1)19 (47.5)15 (37.5)0.5960.7110.3140.540
LPR22 (57.9)21 (52.5)21 (52.5)
* p-value 0.0200.1050.116
* p-value compared between 1-month and 3-month periods; p: prasugrel; C: clopidogrel; T: ticagrelor; VN: VerifyNow; MEA: multiple electrode aggregometry.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Song, K.; Jin, X.; Kim, M.-H.; Li, J.-X.; Jin, C.-D.; Yuan, S.-L.; Song, Z.-Y.; Jin, E.-Z.; Lee, K.-M.; Lim, K.-H.; et al. Differences in Optimal Platelet Reactivity after Potent P2Y12 Inhibitor Treatment in Acute Coronary Syndrome Patients Undergoing Percutaneous Coronary Intervention. J. Clin. Med. 2022, 11, 2480. https://doi.org/10.3390/jcm11092480

AMA Style

Song K, Jin X, Kim M-H, Li J-X, Jin C-D, Yuan S-L, Song Z-Y, Jin E-Z, Lee K-M, Lim K-H, et al. Differences in Optimal Platelet Reactivity after Potent P2Y12 Inhibitor Treatment in Acute Coronary Syndrome Patients Undergoing Percutaneous Coronary Intervention. Journal of Clinical Medicine. 2022; 11(9):2480. https://doi.org/10.3390/jcm11092480

Chicago/Turabian Style

Song, Kai, Xuan Jin, Moo-Hyun Kim, Jia-Xin Li, Cai-De Jin, Song-Lin Yuan, Zhao-Yan Song, En-Ze Jin, Kwang-Min Lee, Kyung-Hee Lim, and et al. 2022. "Differences in Optimal Platelet Reactivity after Potent P2Y12 Inhibitor Treatment in Acute Coronary Syndrome Patients Undergoing Percutaneous Coronary Intervention" Journal of Clinical Medicine 11, no. 9: 2480. https://doi.org/10.3390/jcm11092480

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop