Next Article in Journal
Psychiatric Symptoms in Patients with Cerebral Endometriosis: A Case Report and Literature Review
Next Article in Special Issue
COVID-19 and the Response to Antiplatelet Therapy
Previous Article in Journal
Associations between Periodontitis and COPD: An Artificial Intelligence-Based Analysis of NHANES III
Previous Article in Special Issue
DNA Polymorphisms in Pregnant Women with Sticky Platelet Syndrome
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Resistance on the Latest Oral and Intravenous P2Y12 ADP Receptor Blockers in Patients with Acute Coronary Syndromes: Fact or Myth?

1
Department of Internal Medicine I, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 59 Martin, Slovakia
2
Out-Patient Clinic of Cardiology, 957 01 Banovce nad Bebravou, Slovakia
3
Department of Hematology and Blood Transfusion, National Centre of Hemostasis and Thrombosis, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 59 Martin, Slovakia
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2022, 11(23), 7211; https://doi.org/10.3390/jcm11237211
Submission received: 28 September 2022 / Revised: 12 November 2022 / Accepted: 2 December 2022 / Published: 4 December 2022
(This article belongs to the Special Issue Management of Venous and Arterial Thrombosis)

Abstract

:
Novel P2Y12 ADP receptor blockers (ADPRB) should be preferred in dual-antiplatelet therapy in patients with acute coronary syndrome. Nevertheless, there are still patients who do not respond optimally to novel ADP receptor blocker therapy, and this nonoptimal response (so-called “high on-treatment platelet reactivity” or “resistance”) could be connected with increased risk of adverse ischemic events, such as myocardial re-infarction, target lesion failure and stent thrombosis. In addition, several risk factors have been proposed as factors associated with the phenomenon of inadequate response on novel ADPRB. These include obesity, multivessel coronary artery disease, high pre-treatment platelet reactivity and impaired metabolic status for prasugrel, as well as elderly, concomitant therapy with beta-blockers, morphine and platelet count for ticagrelor. There is no literature report describing nonoptimal therapeutic response on cangrelor, and cangrelor therapy seems to be a possible approach for overcoming HTPR on prasugrel and ticagrelor. However, the optimal therapeutic management of “resistance” on novel ADPRB is not clear and this issue requires further research. This narrative review article discusses the phenomenon of high on-treatment platelet reactivity on novel ADPRB, its importance in clinical practice and approaches for its therapeutic overcoming.

1. Introduction

Novel P2Y12 ADP receptor blockers (ADPRB), namely prasugrel, ticagrelor and cangrelor, have emerged as a potent therapeutic approach for ADP signaling pathway inhibition in patients with acute coronary syndromes (ACSs), with [1,2,3] or without planned percutaneous coronary intervention (PCI) [3], or patients who undergo PCI without oral ADPRB pre-treatment [4]. This therapy should be considered, especially in those patients who do not respond optimally to clopidogrel (patients with clopidogrel high on-treatment platelet reactivity = “clopidogrel resistance”) [5]. Nevertheless, there are still patients who do not respond optimally to novel ADPRB therapy, and this nonoptimal response could be connected with increased risk of adverse ischemic events, such as myocardial re-infarction, target lesion failure and stent thrombosis [6,7]. This article discusses the phenomenon of “resistance” (high on-treatment platelet reactivity) on novel-generation ADPRB (the latest ADPRB available in clinical practice), its importance in clinical practice and approaches for its therapeutic overcoming.

2. Methods

The aim of this article is to provide a brief traditional (narrative) review, which summarizes current data regarding the prevalence and clinical significance of HTPR on novel-generation ADPRB, namely prasugrel, ticagrelor and cangrelor, in patients with acute coronary syndrome. To achieve this aim, the most relevant medical scientific literature databases—Web of Science, PubMed and Scopus—were searched, using selected keywords: “high on treatment platelet reactivity” or “resistance” or “insufficient response” and “prasugrel” or “ticagrelor” or “cangrelor” and “acute coronary syndrome” or “myocardial infarction” or “STEMI” or “NSTEMI” or “unstable angina”. If needed, additional keywords, such as “major adverse cardiac event” or “stent thrombosis” or “stent failure” or “target lesion failure” were added, and the literature was researched. The authors non-systematically identified relevant articles matching their aim. Subsequently, a review of findings from these articles was provided, together with a discussion of the clinical implications of published observations.

3. Insufficient Response to ADPRB and the Risk of Future Events in Patients with ACS

ADPRB treatment failure is a major risk factor for stent thrombosis and early PCI failure [8]. Patients with high on-treatment platelet reactivity (HTPR) have an approximately 2–3-fold higher risk of adverse ischemic events and stent thrombosis than those without HTPR. Moreover, ADPRB HTPR has been observed in patients with ACS previously independently associated with unfavorable in-hospital clinical outcome [9], with increased risk of long-term thrombotic events in patients with implanted drug-eluting stents [10,11], connected with frequent recurrent angina and left ventricular failure [12] and predicted future cardiovascular events after PCI for ACS [13]. A previous observational study [14] showed that clopidogrel resistance was present in 72.5% of patients admitted for repeated ACS, which suggests that HTPR likely plays an important role in recurrent ACS. Additionally, the phenomenon was reported as a leading cause of stent thrombosis (including left main-chain and multi-vessel stent thrombosis) in several clinical cases [15,16,17,18,19].
Despite the abovementioned evidence, there is still an ongoing discussion about the association between antiplatelet drugs HTPR and major adverse cardiovascular events (MACE), and the clinical implication of antiplatelet therapy HTPR is not fully determined. Several factors could be responsible for this ambiguity. First, the results of so-far published studies are controversial (especially for aspirin HTPR), and there are studies that did not confirm the relation between HTPR and the risk of future ischemic adverse events [20,21,22,23]. Second, a MACE in a patient after previous coronary intervention is a complex phenomenon, which could be associated with stent failure itself (due to either stent thrombosis or stent restenosis) or due to “de novo” atherothrombotic events, which could develop due to progression of plaque instability or due to platelet activation or both mechanisms can be involved. Additionally, stent thrombosis, for example, could be connected with HTPR, stent malposition, inadequate stent expansion, stent undersizing, small stent diameter, stent fracture, edge dissection or drug non-compliance [24,25]. Therefore, one needs to understand that HTPR on ADPRB therapy is just one risk factor in a complex clinical problem. Third, there is still no definite answer for how to deal with HTPR, as multiple previously tested approaches failed to improve clinical outcomes [26,27,28]. Considering the fact that failure to reduce platelet reactivity in the settings of HTPR seems to be an independent predictor of future MACE [29], the issue of not having an optimal algorithm for HTPR-guided intensification of platelet inhibition could definitely play an important role in these uncertainties.
Fourth, there is an issue in laboratory testing for HTPR detection. In fact, various platelet function tests (PFTs) with different test principles have been tested (and validated) for the detection of HTPR [30]. Some of them are designed as point-of-care tests, others require complex laboratory equipment and skilled staff to perform the examination. Light transmission aggregometry (LTA) with a specific inducer (adenosine diphosphate—ADP) is still recognized as a standard laboratory test for PFT, which also includes the detection of HTPR, while the Vasodilator-Stimulated Phosphoprotein phosphorylation (VASP-P) by flow cytometric analysis is probably the most specific test for assessing the rate of P2Y12 ADP signaling pathway inhibition [31]. Both tests have important limitations (especially in the settings of daily clinical practice, including the need for measuring the antiplatelet drug response in a 24/7 ACS program), such as the need for special equipment, skilled staff, time demand and, in the case of LTA, the need to process the sample immediately (within first hour from blood sampling). Therefore, several point-of-care assays, namely Multiplate®, VerifyNow®, PFA-100® and platelet mapping thromboelastography® (TEG®), have been designed. Multiplate® (Roche Diagnostics, Indianapolis, IN, USA) is a point-of-care assay, which tests citrated whole blood samples using the electrical impedance aggregometry principle. The assay uses platelet stimulation with specific inducers (ADP) to activate platelet aggregation. Once the aggregated platelets attach the sensor wires in the Multiplate® device, electrical resistance (impedance) is detected and displayed as aggregation units (AUs) against time (area under the curve = AUC) [32]. VerifyNow® (Werfen, Barcelona, Spain) assay is performed as a point-of-care test using a citrated whole blood sample. In this turbidimetry-based assay, ADP induction is used to initiate platelet aggregation on fibrinogen-coated beads. Platelet aggregation is determined by the percentage of the light transmission and expressed in P2Y12 reaction units (PRUs). Low PRU indicates the high P2Y12 receptor inhibition and better response to P2Y12 ADPRB. VerifyNow® assay is a rapid test, which can be performed even at the bedside within 5 min, which is an advantage when compared with LTA and VASP-P assays. Moreover, the examination itself (due to a simple technique) and the interpretation of results can be carried out easily (there is no need for skilled staff to perform and/or evaluate the results). Both assays have been used for PFT in post-marketing studies, including randomized ones [26,30,32]. Platelet function assay-100 = PFA-100® (Siemens Medical Solutions, Malvern, PA, USA) is another point-of-care assay, which can be used to monitor the effect of P2Y12 ADPRB. The assay uses citrated whole blood sample and measures the platelet aggregation and effect of antiplatelet agents under higher shear stress. This test can be performed rapidly (in less time) and, similarly to VerifyNow® assay, has a simple test technique, which is an added advantage when compared with conventional PFT. PFA-100® has collagen-coated, epinephrine-coated and ADP-coated cartridges. If ADPRB is present in a sample, the blood will flow under higher shear rate through the capillary and through a small aperture of the PFA-100® analyzer. Subsequently, platelets will aggregate and form the ADP-induced platelet plug by blocking the aperture. The time taken for complete occlusion of the aperture is recorded as closure time (CT). Prolonged CT indicates a better response to ADPRB [32]. PFA-100® assay has been used in clinical studies mostly for the detection of aspirin resistance [33,34]; however, there are data to show that the test can be used also for the determination of response to ADPRB [35]. On the other side, it is questionable whether the use of point-of-care assays (compared to traditional PFT) has an impact (negative) on the clinical utility of PFT studies, as these assays are criticized for their limited sensitivity and/or specificity [36,37]. Additionally, it is possible that a single PFT will not reliably detect HTPR, and that confirmation of suspected HTPR with another PFT might be needed for establishment of final diagnosis (confirmation of results obtained from a point-of-care test by a laboratory-based test). Finally, there is an issue of inconsistent cut-off values for the detection of HTPR, especially when point-of-care assays are used for its determination. For example, for the VerifyNow® assay, cut-off values of >280 PRU, >272 PRU, >235 PRU and >230 PRU have been used to define HTPR in previously published studies [26,30]; for Multiplate®, at least two cut-off values (468 AUC, 450 AUC) have been reported. All these unclosed issues could explain the ongoing discussion regarding the clinical implication of HTPR.
Nonetheless, considering the fact that, with clopidogrel, ADP-induced platelet aggregation remains significantly high in ACS patients, even after 48 h from standard loading [38], it is unsurprising that a novel generation of ADPRB (Table 1) with more rapid and more potent platelet inhibitory effects has been developed and introduced to clinical practice, especially for the treatment of patients with ACS.

4. Novel-Generation ADPRB

4.1. Prasugrel

Prasugrel is an irreversible, 3rd-generation thienopyridine P2Y12 ADPRB, which is indicated for combined (with aspirin) antiplatelet therapy in PCI-treated patients with ACS [1,2,39]. Prasugrel [39] provides more consistent inhibition of the P2Y12 ADP receptor and has lower intraindividual variability in efficacy compared with clopidogrel. It is hydrolyzed by plasma esterases, then metabolized by cytochrome P450 (CYP) 3A4 and 2B6 enzymes to form an active metabolite and has a plasma half-life of 7.4 h. The inactivation of the active metabolite is mediated trough drug S-methylation and drug conjugation. The inactive metabolites are excreted by urine (68%) and stool. The response to prasugrel is not affected by CYP 2C19 inhibition, loss of CYP 2C19 gene function or decreased function of P-glycoprotein (P-gp). Loading doses of 60 mg of prasugrel reach, in theory, full antiplatelet effects 15–30 min after administration. The benefit of prasugrel therapy seems to be the highest in patients with diabetes mellitus [1]. Prasugrel therapy was repeatedly used to overcome clopidogrel resistance [15,19].

4.2. Ticagrelor

Ticagrelor is a reversible, 3rd-generation non-thienopyridine P2Y12 ADPRB, approved for combined antiplatelet therapy in patients with ACS (with or without PCI) and for long-term combined antiplatelet therapy in coronary artery disease (CAD) patients with high ischemic and low bleeding risk [3,40]. Ticagrelor offers rapid and consistent inhibition of the P2Y12 ADP signaling pathway, which is independent of previous metabolic activation and P-gp function. Ticagrelor reaches its maximal plasma activity approximately 1.5 h after ingestion and has a plasma half-life of 8.5 h. However, it undergoes metabolism, which is mediated by CYP 3A4 and, therefore, strong inducers/inhibitors of this enzyme complex could affect the concentrations of ticagrelor and lead to unexpected drug activity. Ticagrelor is, after metabolic transformation, eliminated by hepatic and renal excretion. A loading dose of 180 mg followed by a maintenance dose of 90 mg twice daily are recommended for patients with ACS, while a dose of 60 mg twice daily is recommended for long-term antiplatelet prophylaxis in high-ischemic-risk CAD patients [5,39,40]. Although a recent randomized study suggested better efficacy (with similar safety profile) of prasugrel compared to ticagrelor in ACS patients undergoing PCI [2], ticagrelor therapy had been repeatedly described as an effective approach for overcoming clopidogrel resistance [5,41,42] and is still preferred in those ACS patients who could not receive prasugrel.

4.3. Cangrelor

Cangrelor is a reversible, parenteral, 3rd-generation non-thienopyridine P2Y12 ADPRB, which is indicated for combined antiplatelet therapy in patients undergoing PCI (both for ACS and stable CAD) who are not pre-treated with oral ADPRB [4]. Cangrelor reaches the maximal antiplatelet effect within 2 min after bolus injection (30 ug per kg of body weight) and has a very short plasma half-life (2.6–3.3 min) requiring continuous intravenous infusion (4 ug per kg of body weight per minute) to maintain adequate ADP receptor inhibition. Cangrelor does not require metabolic transformation to form an active metabolite and is independent of CYP and P-gp activity. Cangrelor is deactivated by plasmatic de-phosphorylation; the inactive metabolite is eliminated by urine (58%) and stool (35%). Normal platelet function is restored within 1 h after stopping the cangrelor infusion [43]. Cangrelor administration could be, in theory, used as a bailout option for overcoming clopidogrel resistance in patients presenting with stent thrombosis [44].

5. Prasugrel Resistance in Patients with ACS

In one of the first studies examining the prevalence of insufficient response on prasugrel, Bonello et al. [45] reported that a significant portion of patients undergoing PCI for ACS did not achieve optimal platelet inhibition. In this study, with 301 patients receiving a prasugrel loading dose of 60 mg, 25.2% of patients had HTPR. Patients who experienced a thrombotic event after PCI had significantly higher residual platelet activity measured with VASP-P compared with those free of adverse thrombotic events. In our previous prospective study, which aimed to map the platelet reactivity in novel ADP receptor-blocker-treated patients with acute ST elevation myocardial infarction (STEMI), 60.9% of prasugrel-treated patients did not achieve sufficient platelet inhibition after a loading dose of 60 mg (measured 1.6 ± 0.7 h after loading dose administration) and 8.7% of prasugrel-treated patients remained non-responders in second blood sampling performed after 20.4 ± 2.6 h from loading dose administration [46]. In addition, Aradi et al. [47] tested platelet reactivity with LTA and whole blood impedance aggregometry (Multiplate®) in 103 consecutive, high-risk ACS patients 12 to 24 h after administration of loading dose (60 mg) of prasugrel. The authors of this study reported significant inter-patient variability in platelet reactivity after all doses of prasugrel, and the prevalence of HTPR was significantly higher during the maintenance dose administration. On the other side, another study enrolling PCI-treated ACS patients receiving prasugrel reported HTPR (defined as VASP-P index > 50%) 2 to 4 weeks after hospital discharge only in 6.8% of patients [48]. Similarly, only 9.1% of acute STEMI patients was identified as a non-responders on prasugrel (defined as VASP-P index > 50% 6 to 12 h after prasugrel loading dose administration) in a previous prospective study performed by Laine et al. [49]. A previous meta-analysis of 14 studies with 1822 patients reported the HTPR in 9.8% of prasugrel-treated patients [50]; Siller-Matula et al. [51] reported that only 3% of prasugrel-treated patients had HTPR in the maintenance phase of treatment. In another single-center retrospective analysis of 809 PCI-treated ACS patients, the prevalence of prasugrel HTPR was even lower and only 1.6% of prasugrel-treated patients fulfilled the criteria for HTPR [52]. However, this study measured platelet reactivity with whole blood impedance aggregometry. In a more recent analysis, Verdoia et al. [53,54], in their prospective studies, reported that HTPR on prasugrel was observed in 10% and 12.3% of patients, respectively. In these studies, whole blood impedance aggregometry was used for determination of HTPR.
Based on these data (Table 2), one could conclude that, although with significantly lower prevalence (compared to clopidogrel), HTPR on prasugrel exists, and its prevalence varies from 1.6 to approximately 25% of treated patients (depending on platelet function test used and chosen time from loading dose to blood sampling). In addition, if the time interval from drug administration to blood sampling is too short, the number of patients with inadequate response can be even higher [46]. Nevertheless, it seems that the phenomenon of prasugrel HTPR is connected with adverse ischemic events. This observation was repeatedly reported in previous prospective studies, including sub-analyses of randomized studies [45,55,56]. Aradi et al. [56], for example, reported, in a pre-specified exploratory analysis of the TROPICAL-ACS trial, that, although infrequent, prasugrel HTPR was connected with increased risk of thrombotic events. The association between prasugrel HTPR and the higher risk of adverse ischemic events post PCI is also supported by multiple reported clinical cases of prasugrel “resistance”, which consistently reported serious adverse thrombotic events, including repeated cases of stent thrombosis [6,57,58]. Hence, the HTPR on prasugrel could probably be considered as a risk factor for adverse ischemia/thrombosis, similarly to clopidogrel HTPR.
A question regarding the mechanism of prasugrel HTPR could be posed. In fact, right now, no satisfactory answer to this question exists, as there is no study examining this problem. Theoretically, several factors could be responsible for this insufficient drug response (Figure 1). Prasugrel HTPR can be caused by decreased bioavailability—either due to decreased absorption or increased drug elimination, by impaired drug metabolism (either due to genetic polymorphism or drug interactions), leading to decreased formation of an active metabolite, due to ineffective inhibition of the platelet P2Y12 ADP receptor via an active metabolite or due to impaired response on ADP receptor inhibition on the level of the post-receptor signaling pathway [59].

6. Ticagrelor Resistance in Patients with ACS

Although first observations reported no risk of HTPR on ticagrelor [60,61], Laine et al. [62] subsequently reported, in their multicenter prospective observational study enrolling 115 ticagrelor-treated patients, that platelet inhibition post 180 mg of ticagrelor loading dose is not uniform and that 3.5% of patients had HTPR (defined as VASP-P index > 50%, blood sampling was performed 6 to 24 h post drug loading). In our previously mentioned analysis of STEMI patients planned for primary PCI [46], platelet inhibition after 180 mg of ticagrelor loading dose was not sufficient in 42.9% of patients in a sample taken 1.4 ± 0.6 h and in 14.3% of patients in a sample taken 21.0 ± 2.0 h post drug loading, respectively. In other studies, the range of ticagrelor HTPR after loading dose administration ranged from 1.5 to 60.2%, depending on studied patient population, method used for HTPR detection and timing of blood sampling [50,51,63,64,65,66,67]. Although Verdoia et al. reported in their studies [54,68,69] 8.6 to 13.7% prevalence of ticagrelor HTPR on maintenance dosing (tested 30 to 90 days post drug loading), it seems that a longer duration of ticagrelor therapy probably achieves sufficient platelet inhibition in the majority of patients, as the majority of so-far published studies, including meta-analyses, reported very low rates (0.0–1.9%) of ticagrelor HTPR (Table 3) on long-term therapy [49,52,70,71,72,73,74,75,76]. This prevalence practically limits the occurrence of ticagrelor HTPR to occasional clinical cases. However, although the evidence is still limited and the phenomenon of ticagrelor resistance is relatively rare, ticagrelor HTPR seems to be connected with a higher risk of adverse ischemic events [67], very similarly to clopidogrel HTPR and prasugrel HTPR. Additionally, Musallam et al. reported a case of a patient with subacute stent thrombosis in whom ticagrelor HTPR was verified [7], and Malik [77] described a case of stent thrombosis 22 days after successful drug eluting coronary stent implantation in a 62-year-old man with diabetes who did not respond adequately to ticagrelor therapy. In this particular case, intravascular imaging with optical coherence tomography was performed, and stent underexpansion, stent strunt mal-apposition and edge dissection were excluded. Finally, Jariwala et al. [78] described a case of subacute stent thrombosis after uncomplicated implantation of sirolimus-eluting coronary stent, which developed despite ticagrelor therapy (in this case, authors were unable to verify HTPR with laboratory testing). In summary, current evidence suggests that ticagrelor HTPR exists, but its prevalence is lower compared to clopidogrel and prasugrel HTPR. Nevertheless, this phenomenon seems to be connected with a higher risk of ischemic adverse events, including stent thrombosis.
Looking at the mechanism of ticagrelor HTPR, it is obvious that impaired (pro)drug conversion (metabolism) does not play a role, as ticagrelor is an active metabolite, which does not require metabolic transformation to achieve its drug activity. In theory, all the other possible mechanisms discussed in connection with prasugrel HTPR (Figure 1) could be responsible [59]. Nevertheless, similarly to prasugrel HTPR, there is no study specifically examining the mechanism of this HTPR; therefore, clarification of this matter is still open for future research.

7. Cangrelor Resistance in Patients with ACS

Looking at the currently available data, there is no study examining the prevalence of cangrelor HTPR in patients with acute coronary syndrome, no study describing this phenomenon or clinical case report describing a case of ischemic adverse post-PCI event related to failure of cangrelor therapy. Moreover, cangrelor seems to be a promising agent for treatment (overcoming) prasugrel HTPR [79,80] or to bridge the gap until optimal platelet inhibition with ticagrelor is achieved [81]. Nevertheless, cangrelor has been approved for clinical use in patients undergoing PCI relatively recently; thus, it might be possible that the phenomenon of cangrelor HTPR just awaits its description.

8. Risk Factors of Novel ADPRB Resistance

The next question should be what are the risk factors for HTPR on novel-generation ADPRB? Searching the literature, several factors have been proposed as factors associated with this phenomenon. For prasugrel, obesity (BMI > 30 kg/m2) [48,82,83,84], multivessel coronary artery disease [48], carrying a CYP 2C19*2 or 2C19*17 loss-of-function allele [85,86], high pre-treatment platelet reactivity and smoking status [87] and impaired metabolic status (with higher levels of glycosylated hemoglobin and low-density lipoprotein cholesterol) [53] have been reported as factors independently predicting HTPR. Several other clinical factors, namely chronic kidney disease [53], type 2 diabetes [88], elderly and proton pump inhibition co-therapy [47], were studied, but an association was not found. For ticagrelor, in a previous study, age and BMI positively and smoking negatively affected on-treatment platelet reactivity [71]. Verdoia et al. [68] reported that, using multivariable analysis, age (≥70 years), concomitant therapy with beta-blockers and platelet count independently predicted HRPR on ticagrelor. In another prospective study in ticagrelor-treated ACS patients, Adamski et al. reported that the presence of ST-segment elevation and morphine co-administration were the strongest predictors of ticagrelor HTPR [89]. Other clinical factors, such as chronic kidney disease, diabetes or proton pump inhibitor co-administration, probably do not affect the efficacy of ticagrelor therapy [88,90,91].

9. How to Manage Insufficient Response to Novel ADPRB?

In theory, novel-generation ADPRB HTPR can be managed by modification of drug dosing (either with re-loading or increasing the maintenance dosage) [92], by adding other antithrombotic agents, to bridge the time until the drug achieves its full activity (glycoprotein IIb/IIIa inhibitor or parenteral ADPRB—cangrelor) [93] or to achieve more potent platelet inhibition in long-term therapy (cilostazol or low-dose rivaroxaban) [94], or by switching the novel-generation ADPRB (prasugrel to ticagrelor in prasugrel HTPR and ticagrelor to prasugrel in ticagrelor HTPR) [54,95]. One must say that each of the strategies has its disadvantages and that none of them was tested in a randomized trial. For example, adding the third antithrombotic agent to long-term therapy can increase the risk of bleeding, while the effect on a reduction in adverse thrombotic events remains unclear. In addition, increasing the drug dose leads to long-term drug dosing, for which efficacy and safety were not previously tested in clinical trials. Furthermore, cangrelor has been approved only in ADPRB-naïve patients who are planned for PCI, and the safety of its administration in those who already received ADPRB loading (although with insufficient platelet response) remains unclear. In the majority of so-far published cases [6,7,57,58,77,78], the authors used a switch strategy, either alone or with bridging the ineffective antiplatelet response with adding glycoprotein IIb/IIIa inhibition. This strategy appears to be safe and effective, as there were no reports of repeated ischemic or serious bleeding adverse events in these cases. However, the evidence for any approach is still limited to a small number of clinical cases, and further research on the issue of optimal management of HTPR on novel-generation ADPRB is definitely needed.

10. Conclusions

Based on the limited evidence discussed in this review article, we can conclude that the phenomenon of HTPR or resistance on novel-generation ADPRB therapy might exist. The prevalence of novel-generation ADPRB HTPR is lower compared to clopidogrel HTPR. Additionally, several studies suggested that this phenomenon could be connected with a high risk of adverse ischemic events; however, the evidence for this association is still limited. Therefore, there is a need for future research dedicated to clinical impact and optimal management of this phenomenon.

Author Contributions

P.B., M.S., T.B. and L.S. designed the study; P.B., M.S. and T.B. drafted the manuscript; I.Š., M.J.P. and J.J. performed the search of the literature, analyzed and interpreted the data; L.S., J.J., J.S. and M.M. revised the manuscript critically. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by project APVV (Slovak Research and Development Agency) 16-0020, by projects of Research Agency of Slovak Ministry of Education, Science and Sports (VEGA) 1/0090/20 and 1/0549/19.

Institutional Review Board Statement

This research was conducted according to ethical standards. There is no need for formal consent for this type of study (a narrative review article).

Informed Consent Statement

Not applicable.

Data Availability Statement

All the source data are available from the corresponding author upon reasonable request.

Acknowledgments

This study was supported by project APVV (Slovak Research and Development Agency) 16-0020, by projects of Research Agency of Slovak Ministry of Education, Science and Sports (VEGA) 1/0090/20 and 1/0549/19.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Wiviott, S.D.; Braunwald, E.; McCabe, C.H.; Montalescot, G.; Ruzyllo, W.; Gottlieb, S.; Neumann, F.-J.; Ardissino, D.; De Servi, S.; Murphy, S.A.; et al. Prasugrel versus Clopidogrel in Patients with Acute Coronary Syndromes. N. Engl. J. Med. 2007, 357, 2001–2015. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Schüpke, S.; Neumann, F.J.; Menichelli, M.; Mayer, K.; Bernlochner, I.; Wöhrle, J.; Richardt, G.; Liebetrau, C.; Witzenbichler, B.; Antoniucci, D.; et al. Ticagrelor or Prasugrel in Patients with Acute Coronary Syndromes. N. Engl. J. Med. 2019, 381, 1524–1534. [Google Scholar] [CrossRef] [PubMed]
  3. 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]
  4. Bhatt, D.L.; Stone, G.W.; Mahaffey, K.W.; Gibson, C.M.; Steg, P.G.; Hamm, C.W.; Price, M.J.; Leonardi, S.; Gallup, D.; Bramucci, E.; et al. Effect of platelet inhibition with cangrelor during PCI on ischemic events. N. Engl. J. Med. 2013, 368, 1303–1313. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Samoš, M.; Fedor, M.; Kovář, F.; Duraj, L.; Stančiaková, L.; Galajda, P.; Staško, J.; Kubisz, P.; Mokáň, M. Ticagrelor: A safe and effective approach for overcoming clopidogrel resistance in patients with stent thrombosis? Blood Coagul. Fibrinolysis 2016, 27, 117–120. [Google Scholar] [CrossRef] [PubMed]
  6. Fiore, M.; Horovitz, A.; Pons, A.-C.; Leroux, L.; Casassus, F. First report of a subacute stent thrombosis in a prasugrel resistant patient successfully managed with ticagrelor. Platelets 2013, 25, 636–638. [Google Scholar] [CrossRef] [PubMed]
  7. Musallam, A.; Lev, E.I.; Roguin, A. Stent thrombosis in a patient with high on-treatment platelet reactivity despite ticagrelor treatment. Eur. Heart J. Acute Cardiovasc. Care 2014, 4, 85–87. [Google Scholar] [CrossRef]
  8. Thomas, M.R.; Storey, R.F. Clinical significance of residual platelet reactivity in patients treated with platelet P2Y12 inhibitors. Vasc. Pharmacol. 2016, 84, 25–27. [Google Scholar] [CrossRef]
  9. Małek, Ł.A.; Spiewak, M.; Filipiak, K.J.; Grabowski, M.; Szpotańska, M.; Rosiak, M.; Główczyńska, R.; Imiela, T.; Huczek, Z.; Opolski, G. Persistent platelet activation is related to very early cardiovascular events in patients with acute coronary syndromes. Kardiol. Pol. 2007, 65, 40–45. [Google Scholar]
  10. Wang, L.; Wang, X.; Chen, F. Clopidogrel resistance is associated with long-term thrombotic events in patients implanted with drug-eluting stents. Drugs R D 2010, 10, 219–224. [Google Scholar] [CrossRef]
  11. Liang, Z.-Y.; Han, Y.-L.; Zhang, X.-L.; Li, Y.; Yan, C.-H.; Kang, J. The impact of gene polymorphism and high on-treatment platelet reactivity on clinical follow-up: Outcomes in patients with acute coronary syndrome after drug-eluting stent implantation. EuroIntervention 2013, 9, 316–327. [Google Scholar] [CrossRef]
  12. Costache, I.I.; Rusu, C.; Ivanov, I.; Popescu, R.; Petriş, A. Impact of clopidogrel response on the clinical evolution in patients with acute coronary syndromes. Rev. Med. Chir. Soc. Med. Nat. IASI 2012, 116, 962–967. [Google Scholar] [PubMed]
  13. Li, M.; Wang, H.; Xuan, L.; Shi, X.; Zhou, T.; Zhang, N.; Huang, Y. Associations between P2RY12 gene polymorphisms and risks of clopidogrel resistance and adverse cardiovascular events after PCI in patients with acute coronary syndrome. Medicine 2017, 96, e6553. [Google Scholar] [CrossRef] [PubMed]
  14. Guha, S.; Mookerjee, S.; Guha, P.; Sardar, P.; Deb, S.; Das Roy, P.; Karmakar, R.; Mani, S.; Hema, M.B.; Pyne, S.; et al. Antiplatelet drug resistance in patients with recurrent acute coronary syndrome undergoing conservative management. Indian Heart J. 2010, 61, 348–352. [Google Scholar]
  15. Samoš, M.; Šimonová, R.; Kovář, F.; Duraj, L.; Fedorová, J.; Galajda, P.; Staško, J.; Fedor, M.; Kubisz, P.; Mokáň, M. Clopidogrel resistance in diabetic patient with acute myocardial infarction due to stent thrombosis. Am. J. Emerg. Med. 2014, 32, 461–465. [Google Scholar] [CrossRef] [PubMed]
  16. Lee, J.S.; Dunn, S.P.; Marshall, H.M.; Cohen, M.G.; Smyth, S.S. A case report of simultaneous thrombosis of two coronary artery stents in association with clopidogrel resistance. Clin. Cardiol. 2007, 30, 200–203. [Google Scholar] [CrossRef]
  17. Schäfer, A.; Bonz, A.W.; Eigenthaler, M.; Bauersachs, J. Late thrombosis of a drug-eluting stent during combined anti-platelet therapy in a clopidogrel nonresponsive diabetic patient: Shall we routinely test platelet function? Thromb Haemost 2007, 97, 862–865. [Google Scholar]
  18. Jung, H.-J.; Sir, J.-J. Recurrent myocardial infarction due to one subacute and two very late thrombotic events of drug-eluting stent associated with clopidogrel resistance. J. Invasive Cardiol. 2011, 23, E13–E18. [Google Scholar]
  19. Kim, Y.-S.; Lee, S.-R. Successful Prasugrel Therapy for Recurrent Left Main Stent Thrombosis in a Clopidogrel Hyporesponder. Tex. Heart Inst. J. 2015, 42, 483–486. [Google Scholar] [CrossRef] [Green Version]
  20. Redfors, B.; Ben-Yehuda, O.; Lin, S.-H.; Furer, A.; Kirtane, A.J.; Witzenbichler, B.; Weisz, G.; Stuckey, T.D.; Maehara, A.; Généreux, P.; et al. Quantifying Ischemic Risk After Percutaneous Coronary Intervention Attributable to High Platelet Reactivity on Clopidogrel (From the Assessment of Dual Antiplatelet Therapy with Drug-Eluting Stents Study). Am. J. Cardiol. 2017, 120, 917–923. [Google Scholar] [CrossRef]
  21. Olędzki, S.; Kornacewicz-Jach, Z.; Safranow, K.; Kiedrowicz, R.; Gawrońska-Szklarz, B.; Jastrzębska, M.; Gorący, J. Variability of platelet response to clopidogrel is not related to adverse cardiovascular events in patients with stable coronary artery disease undergoing percutaneous coronary intervention. Eur. J. Clin. Pharmacol. 2017, 73, 1085–1094. [Google Scholar] [CrossRef] [PubMed]
  22. Bolliger, D.; Filipovic, M.; Matt, P.; Tanaka, K.A.; Gregor, M.; Zenklusen, U.; Seeberger, M.D.; Buse, G.L. Reduced aspirin responsiveness as assessed by impedance aggregometry is not associated with adverse outcome after cardiac surgery in a small low-risk cohort. Platelets 2015, 27, 254–261. [Google Scholar] [CrossRef] [PubMed]
  23. Chung, C.J.; Kirtane, A.J.; Zhang, Y.; Witzenbichler, B.; Weisz, G.; Stuckey, T.D.; Brodie, B.R.; Rinaldi, M.J.; Neumann, F.-J.; Metzger, D.C.; et al. Impact of high on-aspirin platelet reactivity on outcomes following successful percutaneous coronary intervention with drug-eluting stents. Am. Heart J. 2018, 205, 77–86. [Google Scholar] [CrossRef] [PubMed]
  24. Redfors, B.; Chen, S.; Généreux, P.; Witzenbichler, B.; Weisz, G.; Stuckey, T.D.; Maehara, A.; McAndrew, T.; Mehran, R.; Ben-Yehuda, O.; et al. Relationship Between Stent Diameter, Platelet Reactivity, and Thrombotic Events After Percutaneous Coronary Artery Revascularization. Am. J. Cardiol. 2019, 124, 1363–1371. [Google Scholar] [CrossRef]
  25. Gori, T.; Polimeni, A.; Indolfi, C.; Räber, L.; Adriaenssens, T.; Münzel, T. Predictors of stent thrombosis and their implications for clinical practice. Nat. Rev. Cardiol. 2018, 16, 243–256. [Google Scholar] [CrossRef]
  26. Price, M.J.; Berger, P.B.; Teirstein, P.S.; Tanguay, J.F.; Angiolillo, D.J.; Spriggs, D.; Puri, S.; Robbins, M.; Garratt, K.N.; Bertrand, O.F.; et al. Standard- vs. high-dose clopidogrel based on platelet function testing after percutaneous coronary intervention: The GRAVITAS randomized trial. JAMA 2011, 305, 1097–1105. [Google Scholar] [CrossRef]
  27. Chandrasekhar, J.; Baber, U.; Mehran, R.; Aquino, M.; Sartori, S.; Yu, J.; Kini, A.; Sharma, S.; Skurk, C.; Shlofmitz, R.A.; et al. Impact of an integrated treatment algorithm based on platelet function testing and clinical risk assessment: Results of the TRIAGE Patients Undergoing Percutaneous Coronary Interventions to Improve Clinical Outcomes Through Optimal Platelet Inhibition study. J. Thromb. Thrombolysis 2016, 42, 186–196. [Google Scholar] [CrossRef]
  28. Xing, Z.; Tang, L.; Zhu, Z.; Huang, J.; Peng, X.; Hu, X. Platelet reactivity-adjusted antiplatelet therapy in patients with percutaneous coronary intervention: A meta-analysis of randomized controlled trials. Platelets 2017, 29, 589–595. [Google Scholar] [CrossRef]
  29. Robledo-Nolasco, R.; Godínez-Montes de Oca, A.; Zaballa-Contreras, J.F.; Suárez-Cuenca, J.A.; Mondragón-Terán, P.; Rubio-Guerra, A.F.; Meléndez-Alcántara, M.A. Efficacy of Change to New P2Y12 Receptor Antagonists in Patients High on Treatment Platelet Reactivity Undergoing Percutaneous Coronary Intervention. Clin. Appl. Thromb. Hemost. 2015, 21, 619–625. [Google Scholar] [CrossRef]
  30. 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]
  31. Fedor, M.; Samoš, M.; Šimonová, R.; Fedorová, J.; Škorňová, I.; Duraj, L.; Staško, J.; Kovář, F.; Mokáň, M.; Kubisz, P. Monitoring the efficacy of ADP inhibitor treatment in patients with acute STEMI post-PCI by VASP-P flow cytometry assay. Clin. Appl. Thromb. Hemost. 2015, 21, 334–338. [Google Scholar] [CrossRef] [PubMed]
  32. Alvitigala, B.Y.; Gooneratne, L.V.; Constantine, G.R.; Wijesinghe, R.A.N.K.; Arawwawala, L.D.A.M. Pharmacokinetic, pharmacodynamic, and pharmacogenetic assays to monitor clopidogrel therapy. Pharmacol. Res. Perspect. 2020, 8, e00686. [Google Scholar] [CrossRef] [PubMed]
  33. Kinsella, J.A.; Tobin, W.O.; Cox, D.; Coughlan, T.; Collins, R.; O’Neill, D.; Murphy, R.P.; McCabe, D.J. Prevalence of Ex Vivo High On-treatment Platelet Reactivity on Antiplatelet Therapy after Transient Ischemic Attack or Ischemic Stroke on the PFA-100® and VerifyNow®. J. Stroke Cerebrovasc. Dis. 2013, 22, e84–e92. [Google Scholar] [CrossRef] [PubMed]
  34. Crescente, M.; Mezzasoma, A.M.; Del Pinto, M.; Palmerini, F.; Di Castelnuovo, A.; Cerletti, C.; De Gaetano, G.; Gresele, P. Incomplete inhibition of platelet function as assessed by the platelet function analyzer (PFA-100) identifies a subset of cardiovascular patients with high residual platelet response while on aspirin. Platelets 2011, 22, 179–187. [Google Scholar] [CrossRef] [PubMed]
  35. Koessler, J.; Kobsar, A.L.; Rajkovic, M.S.; Schafer, A.; Flierl, U.; Pfoertsch, S.; Bauersachs, J.; Steigerwald, U.; Rechner, A.R.; Walter, U. The new INNOVANCE® PFA P2Y cartridge is sensitive to the detection of the P2Y12 receptor inhibition. Platelets 2011, 22, 20–27. [Google Scholar] [CrossRef] [PubMed]
  36. Paniccia, R.; Antonucci, E.; Gori, A.M.; Marcucci, R.; Poli, S.; Romano, E.; Valente, S.; Giglioli, C.; Fedi, S.; Gensini, G.F.; et al. Comparison of Different Methods to Evaluate the Effect of Aspirin on Platelet Function in High-Risk Patients with Ischemic Heart Disease Receiving Dual Antiplatelet Treatment. Am. J. Clin. Pathol. 2007, 128, 143–149. [Google Scholar] [CrossRef]
  37. Kweon, O.J.; Lim, Y.K.; Kim, B.; Lee, M.-K.; Kim, A.H.R. Effectiveness of Platelet Function Analyzer-100 for Laboratory Detection of Anti-Platelet Drug-Induced Platelet Dysfunction. Ann. Lab. Med. 2019, 39, 23–30. [Google Scholar] [CrossRef] [Green Version]
  38. Guha, S.; Mookerjee, S.; Lahiri, P.; Mani, S.; Saha, J.; Guha, S.; Majumdar, D.; Mandal, M.; Bhattacharya, R. A study of platelet aggregation in patients with acute myocardial infarction at presentation and after 48 hrs of initiating standard anti platelet therapy. Indian Heart J. 2013, 63, 409–413. [Google Scholar]
  39. Dobesh, P.P.; Varnado, S.; Doyle, M. Antiplatelet Agents in Cardiology: A Report on Aspirin, Clopidogrel, Prasugrel, and Ticagrelor. Curr. Pharm. Des. 2016, 22, 1918–1932. [Google Scholar] [CrossRef]
  40. Bonaca, M.P.; Bhatt, D.L.; Cohen, M.; Steg, P.G.; Storey, R.F.; Jensen, E.C.; Magnani, G.; Bansilal, S.; Fish, M.P.; Im, K.; et al. Long-Term Use of Ticagrelor in Patients with Prior Myocardial Infarction. N. Engl. J. Med. 2015, 372, 1791–1800. [Google Scholar] [CrossRef] [Green Version]
  41. Wang, W.; Wang, B.; Chen, Y.; Wei, S. Late Stent Thrombosis After Drug-Coated Balloon Coronary Angioplasty for In-Stent Restenosis. Int. Heart J. 2021, 62, 171–174. [Google Scholar] [CrossRef] [PubMed]
  42. Lee, S.-Y.; Cho, J.Y.; Yun, K.H.; Oh, S.K. Successful Therapy with Ticagrelor in Three-Vessel Stent Thrombosis Related to Clopidogrel Resistance. Can. J. Cardiol. 2021, 37, 1278–1280. [Google Scholar] [CrossRef] [PubMed]
  43. Keating, G.M. Cangrelor: A Review in Percutaneous Coronary Intervention. Drugs 2015, 75, 1425–1434. [Google Scholar] [CrossRef] [PubMed]
  44. Chen, Y.; Bernardo, N.L.; Waksman, R. Cangrelor for the Rescue of Intra-Procedural Stent Thrombosis in Percutaneous Coronary Intervention. Cardiovasc. Revascularization Med. 2019, 20, 624–625. [Google Scholar] [CrossRef]
  45. Bonello, L.; Pansieri, M.; Mancini, J.; Bonello, R.; Maillard, L.; Barnay, P.; Rossi, P.; Ait-Mokhtar, O.; Jouve, B.; Collet, F.; et al. High On-Treatment Platelet Reactivity after Prasugrel Loading Dose and Cardiovascular Events after Percutaneous Coronary Intervention in Acute Coronary Syndromes. J. Am. Coll. Cardiol. 2011, 58, 467–473. [Google Scholar] [CrossRef]
  46. Skorňová, I.; Samoš, M.; Šimonová, R.; Žolková, J.; Stančiaková, L.; Vádelová, L.; Bolek, T.; Urban, L.; Kovář, F.; Staško, J.; et al. On-treatment platelet reactivity in the era of new ADP receptor blockers: Data from a real-world clinical practice. Acta Med. Martiniana 2018, 18, 34–39. [Google Scholar] [CrossRef] [Green Version]
  47. Kuliczkowski, W.; Atar, D.; Serebruany, V.L.; Aradi, D. Inter-patient variability and impact of proton pump inhibitors on platelet reactivity after prasugrel. Thromb. Haemost. 2012, 107, 338–345. [Google Scholar] [CrossRef] [Green Version]
  48. Cayla, G.; Cuisset, T.; Silvain, J.; O’Connor, S.A.; Kerneis, M.; Castelli, C.; Quilici, J.; Bonnet, J.-L.; Alessi, M.-C.; Morange, P.-E.; et al. Prasugrel Monitoring and Bleeding in Real World Patients. Am. J. Cardiol. 2013, 111, 38–44. [Google Scholar] [CrossRef]
  49. Laine, M.; Gaubert, M.; Frère, C.; Peyrol, M.; Thuny, F.; Yvorra, S.; Chelini, V.; Bultez, B.; Luigi, S.; Mokrani, Z.; et al. Comparison of Platelet reactivity following prAsugrel and ticagrelor loading dose in ST-Segment elevation myocardial infarction patients: The COMPASSION study. Platelets 2015, 26, 570–572. [Google Scholar] [CrossRef]
  50. Lemesle, G.; Schurtz, G.; Bauters, C.; Hamon, M. High on-treatment platelet reactivity with ticagrelor versus prasugrel: A systematic review and meta-analysis. J. Thromb. Haemost. 2015, 13, 931–942. [Google Scholar] [CrossRef]
  51. Siller-Matula, J.M.; Akca, B.; Neunteufl, T.; Maurer, G.; Lang, I.M.; Kreiner, G.; Berger, R.; Delle-Karth, G. Inter-patient variability of platelet reactivity in patients treated with prasugrel and ticagrelor. Platelets 2015, 27, 373–377. [Google Scholar] [CrossRef] [PubMed]
  52. Selhorst, G.; Schmidtler, F.; Ott, A.; Hitzke, E.; Tomelden, J.; Antoni, D.; Hoffmann, E.; Rieber, J. Platelet reactivity in patients with acute coronary syndrome treated with prasugrel or ticagrelor in comparison to clopidogrel: A retrospective pharmacodynamic analysis. Platelets 2018, 30, 341–347. [Google Scholar] [CrossRef] [PubMed]
  53. Verdoia, M.; Pergolini, P.; Nardin, M.; Rolla, R.; Barbieri, L.; Marino, P.; Carriero, A.; Suryapranata, H.; De Luca, G. Prevalence and predictors of high-on treatment platelet reactivity during prasugrel treatment in patients with acute coronary syndrome undergoing stent implantation. J. Cardiol. 2018, 73, 198–203. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Verdoia, M.; Pergolini, P.; Nardin, M.; Rolla, R.; Suryapranata, H.; Kedhi, E.; De Luca, G. Ticagrelor and prasugrel in acute coronary syndrome: A single-arm crossover platelet reactivity study. J. Cardiovasc. Med. 2021, 22, 686–692. [Google Scholar] [CrossRef]
  55. Bonello, L.; Mancini, J.; Pansieri, M.; Maillard, L.; Rossi, P.; Collet, F.; Jouve, B.; Wittenberg, O.; Laine, M.; Michelet, P.; et al. Relationship between post-treatment platelet reactivity and ischemic and bleeding events at 1-year follow-up in patients receiving prasugrel. J. Thromb. Haemost. 2012, 10, 1999–2005. [Google Scholar] [CrossRef]
  56. Aradi, D.; Gross, L.; Trenk, D.; Geisler, T.; Merkely, B.; Kiss, R.G.; Komócsi, A.; Dézsi, C.A.; Ruzsa, Z.; Ungi, I.; et al. Platelet reactivity and clinical outcomes in acute coronary syndrome patients treated with prasugrel and clopidogrel: A pre-specified exploratory analysis from the TROPICAL-ACS trial. Eur. Heart J. 2019, 40, 1942–1951. [Google Scholar] [CrossRef]
  57. Silvano, M.; Zambon, C.-F.; De Rosa, G.; Plebani, M.; Pengo, V.; Napodano, M.; Padrini, R. A case of resistance to clopidogrel and prasugrel after percutaneous coronary angioplasty. J. Thromb. Thrombolysis 2010, 31, 233–234. [Google Scholar] [CrossRef]
  58. Ohno, Y.; Okada, S.; Kitahara, H.; Nishi, T.; Nakayama, T.; Fujimoto, Y.; Kobayashi, Y. Repetitive stent thrombosis in a patient who had resistance to both clopidogrel and prasugrel. J. Cardiol. Cases 2016, 13, 139–142. [Google Scholar] [CrossRef]
  59. Warlo, E.M.K.; Arnesen, H.; Seljeflot, I. A brief review on resistance to P2Y12 receptor antagonism in coronary artery disease. Thromb. J. 2019, 17, 1–9. [Google Scholar] [CrossRef]
  60. Alexopoulos, D.; Galati, A.; Xanthopoulou, I.; Mavronasiou, E.; Kassimis, G.; Theodoropoulos, K.C.; Makris, G.; Damelou, A.; Tsigkas, G.; Hahalis, G.; et al. Ticagrelor versus prasugrel in acute coronary syndrome patients with high on-clopidogrel platelet reactivity following percutaneous coronary intervention: A pharmacodynamic study. J. Am. Coll. Cardiol. 2012, 60, 193–199. [Google Scholar] [CrossRef] [Green Version]
  61. Alexopoulos, D.; Xanthopoulou, I.; Mavronasiou, E.; Stavrou, K.; Siapika, A.; Tsoni, E.; Davlouros, P. Randomized Assessment of Ticagrelor Versus Prasugrel Antiplatelet Effects in Patients with Diabetes. Diabetes Care 2013, 36, 2211–2216. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Laine, M.; Toesca, R.; Berbis, J.; Frere, C.; Barnay, P.; Pansieri, M.; Peyre, J.-P.; Michelet, P.; Bessereau, J.; Camilleri, E.; et al. Platelet reactivity evaluated with the VASP assay following ticagrelor loading dose in acute coronary syndrome patients undergoing percutaneous coronary intervention. Thromb. Res. 2013, 132, e15–e18. [Google Scholar] [CrossRef] [PubMed]
  63. Laine, M.; Frere, C.; Toesca, R.; Berbis, J.; Barnay, P.; Pansieri, M.; Michelet, P.; Bessereau, J.; Camilleri, E.; Ronsin, O.; et al. Ticagrelor versus prasugrel in diabetic patients with an acute coronary syndrome. Thromb. Haemost. 2014, 111, 273–278. [Google Scholar] [CrossRef] [Green Version]
  64. Verdoia, M.; Sartori, C.; Pergolini, P.; Nardin, M.; Rolla, R.; Barbieri, L.; Schaffer, A.; Marino, P.; Bellomo, G.; Suryapranata, H.; et al. Immature platelet fraction and high-on treatment platelet reactivity with ticagrelor in patients with acute coronary syndromes. J. Thromb. Thrombolysis 2015, 41, 663–670. [Google Scholar] [CrossRef]
  65. Barbieri, L.; Pergolini, P.; Verdoia, M.; Rolla, R.; Nardin, M.; Marino, P.; Bellomo, G.; Suryapranata, H.; De Luca, G. Platelet reactivity in patients with impaired renal function receiving dual antiplatelet therapy with clopidogrel or ticagrelor. Vasc. Pharmacol. 2016, 79, 11–15. [Google Scholar] [CrossRef] [PubMed]
  66. Li, D.-D.; Wang, X.-Y.; Xi, S.-Z.; Liu, J.; Qin, L.-A.; Jing, J.; Yin, T.; Chen, Y.-D. Relationship between ADP-induced platelet-fibrin clot strength and anti-platelet responsiveness in ticagrelor treated ACS patients. J. Geriatr. Cardiol. 2016, 13, 282–289. [Google Scholar] [CrossRef] [PubMed]
  67. Laine, M.; Panagides, V.; Frère, C.; Cuisset, T.; Gouarne, C.; Jouve, B.; Thuny, F.; Paganelli, F.; Alessi, M.; Mancini, J.; et al. Platelet reactivity inhibition following ticagrelor loading dose in patients undergoing percutaneous coronary intervention for acute coronary syndrome. J. Thromb. Haemost. 2019, 17, 2188–2195. [Google Scholar] [CrossRef]
  68. Verdoia, M.; Sartori, C.; Pergolini, P.; Nardin, M.; Rolla, R.; Barbieri, L.; Schaffer, A.; Marino, P.; Bellomo, G.; Suryapranata, H.; et al. Prevalence and predictors of high-on treatment platelet reactivity with ticagrelor in ACS patients undergoing stent implantation. Vasc. Pharmacol. 2015, 77, 48–53. [Google Scholar] [CrossRef]
  69. Verdoia, M.; Rolla, R.; Negro, F.; Tonon, F.; Pergolini, P.; Nardin, M.; Marcolongo, M.; De Luca, G. Homocysteine levels and platelet reactivity in coronary artery disease patients treated with ticagrelor. Nutr. Metab. Cardiovasc. Dis. 2019, 30, 292–299. [Google Scholar] [CrossRef]
  70. Stavrou, K.; Koniari, I.; Gkizas, V.; Perperis, A.; Kontoprias, K.; Vogiatzi, C.; Bampouri, T.; Xanthopoulou, I.; Alexopoulos, D. 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]
  71. Alexopoulos, D.; Xanthopoulou, I.; Storey, R.F.; Bliden, K.P.; Tantry, U.S.; Angiolillo, D.J.; Gurbel, P.A. Platelet reactivity during ticagrelor maintenance therapy: A patient-level data meta-analysis. Am. Heart J. 2014, 168, 530–536. [Google Scholar] [CrossRef] [PubMed]
  72. Gaglia, M.A., Jr.; Lipinski, M.J.; Lhermusier, T.; Steinvil, A.; Kiramijyan, S.; Pokharel, S.; Torguson, R.; Angiolillo, D.J.; Wallentin, L.; Storey, R.F.; et al. Comparison of Platelet Reactivity in Black Versus White Patients with Acute Coronary Syndromes After Treatment with Ticagrelor. Am. J. Cardiol. 2017, 119, 1135–1140. [Google Scholar] [CrossRef]
  73. Sweeny, J.M.; Angiolillo, D.J.; Franchi, F.; Rollini, F.; Waksman, R.; Raveendran, G.; Dangas, G.; Khan, N.D.; Carlson, G.F.; Zhao, Y.; et al. Impact of Diabetes Mellitus on the Pharmacodynamic Effects of Ticagrelor Versus Clopidogrel in Troponin-Negative Acute Coronary Syndrome Patients Undergoing Ad Hoc Percutaneous Coronary Intervention. J. Am. Heart Assoc. 2017, 6, e005650. [Google Scholar] [CrossRef] [PubMed]
  74. Liu, H.L.; Wei, Y.J.; Ding, P.; Zhang, J.; Li, T.C.; Wang, B.; Wang, M.S.; Li, Y.T.; Zhang, J.J.; Ren, Y.H.; et al. Antiplatelet Effect of Different Loading Doses of Ticagrelor in Patients with Non-ST-Elevation Acute Coronary Syndrome Undergoing. Can. J. Cardiol. 2017, 33, 1675–1682. [Google Scholar] [CrossRef] [PubMed]
  75. Wen, M.; Li, Y.; Qu, X.; Zhu, Y.; Tian, L.; Shen, Z.; Yang, X.; Shi, X. Comparison of platelet reactivity between prasugrel and ticagrelor in patients with acute coronary syndrome: A meta-analysis. BMC Cardiovasc. Disord. 2020, 20, 430. [Google Scholar] [CrossRef]
  76. Dai, L.; Xu, J.; Jiang, Y.; Chen, K. Impact of Prasugrel and Ticagrelor on Platelet Reactivity in Patients with Acute Coronary Syndrome: A Meta-Analysis. Front. Cardiovasc. Med. 2022, 9, 905607. [Google Scholar] [CrossRef]
  77. Malik, J. A Case of Ticagrelor Resistance. Eur. J. Case Rep. Intern. Med. 2021, 8, 002719. [Google Scholar] [CrossRef]
  78. Jariwala, P.; Bhatia, H.; Kumar, E.A.P. Sub-acute stent thrombosis secondary to ticagrelor resistance-Myth or reality! Indian Heart J. 2017, 69, 804–806. [Google Scholar] [CrossRef]
  79. Rollini, F.; Franchi, F.; Tello-Montoliu, A.; Patel, R.; Darlington, A.; Ferreiro, J.L.; Cho, J.R.; Muñiz-Lozano, A.; Desai, B.; Zenni, M.M.; et al. Pharmacodynamic effects of cangrelor on platelet P2Y12 receptor-mediated signaling in prasugrel-treated patients. JACC Cardiovasc. Interv. 2014, 7, 426–434. [Google Scholar] [CrossRef] [Green Version]
  80. Valenti, R.; Muraca, I.; Marcucci, R.; Ciatti, F.; Berteotti, M.; Gori, A.M.; Carrabba, N.; Migliorini, A.; Marchionni, N.; Valgimigli, M. “Tailored” antiplatelet bridging therapy with cangrelor: Moving toward personalized medicine. Platelets 2021, 33, 687–691. [Google Scholar] [CrossRef]
  81. Franchi, F.; Rollini, F.; Rivas, A.; Wali, M.; Briceno, M.; Agarwal, M.; Shaikh, Z.; Nawaz, A.; Silva, G.; Been, L.; et al. Platelet Inhibition with Cangrelor and Crushed Ticagrelor in Patients with ST-Segment–Elevation Myocardial Infarction Undergoing Primary Percutaneous Coronary Intervention. Circulation 2019, 139, 1661–1670. [Google Scholar] [CrossRef]
  82. Cuisset, T.; Gaborit, B.; Dubois, N.; Quilici, J.; Loosveld, M.; Beguin, S.; Loundou, A.D.; Moro, P.J.; Morange, P.E.; Alessi, M.-C.; et al. Platelet reactivity in diabetic patients undergoing coronary stenting for acute coronary syndrome treated with clopidogrel loading dose followed by prasugrel maintenance therapy. Int. J. Cardiol. 2013, 168, 523–528. [Google Scholar] [CrossRef]
  83. Pankert, M.; Quilici, J.; Loundou, A.D.; Verdier, V.; Lambert, M.; Deharo, P.; Bonnet, G.; Gaborit, B.; Morange, P.E.; Valéro, R.; et al. Impact of Obesity and the Metabolic Syndrome on Response to Clopidogrel or Prasugrel and Bleeding Risk in Patients Treated After Coronary Stenting. Am. J. Cardiol. 2014, 113, 54–59. [Google Scholar] [CrossRef]
  84. Silvain, J.; Cayla, G.; Hulot, J.-S.; Finzi, J.; Kerneis, M.; O’Connor, S.A.; Bellemain-Appaix, A.; Barthélémy, O.; Beygui, F.; Collet, J.-P.; et al. High on-thienopyridine platelet reactivity in elderly coronary patients: The SENIOR-PLATELET study. Eur. Heart J. 2011, 33, 1241–1249. [Google Scholar] [CrossRef] [Green Version]
  85. Cuisset, T.; Loosveld, M.; Morange, P.E.; Quilici, J.; Moro, P.J.; Saut, N.; Gaborit, B.; Castelli, C.; Beguin, S.; Grosdidier, C.; et al. CYP2C19*2 and *17 Alleles Have a Significant Impact on Platelet Response and Bleeding Risk in Patients Treated with Prasugrel After Acute Coronary Syndrome. JACC Cardiovasc. Interv. 2012, 5, 1280–1287. [Google Scholar] [CrossRef]
  86. Grosdidier, C.; Quilici, J.; Loosveld, M.; Camoin, L.; Moro, P.J.; Saut, N.; Gaborit, B.; Pankert, M.; Cohen, W.; Lambert, M.; et al. Effect of CYP2C19*2 and *17 Genetic Variants on Platelet Response to Clopidogrel and Prasugrel Maintenance Dose and Relation to Bleeding Complications. Am. J. Cardiol. 2013, 111, 985–990. [Google Scholar] [CrossRef]
  87. Alexopoulos, D.; Xanthopoulou, I.; Perperis, A.; Siapika, A.; Stavrou, K.; Tsoni, E.; Davlouros, P.; Hahalis, G. Factors Affecting Residual Platelet Aggregation in Prasugrel Treated Patients. Curr. Pharm. Des. 2013, 19, 5121–5126. [Google Scholar] [CrossRef]
  88. Samoš, M.; Fedor, M.; Kovář, F.; Galajda, P.; Bolek, T.; Stančiaková, L.; Fedorová, J.; Staško, J.; Kubisz, P.; Mokáň, M. The Impact of Type 2 Diabetes on the Efficacy of ADP Receptor Blockers in Patients with Acute ST Elevation Myocardial Infarction: A Pilot Prospective Study. J. Diabetes Res. 2016, 2016, 2909436. [Google Scholar] [CrossRef] [Green Version]
  89. Adamski, P.; Buszko, K.; Sikora, J.; Niezgoda, P.; Fabiszak, T.; Ostrowska, M.; Barańska, M.; Karczmarska-Wódzka, A.; Navarese, E.P.; Kubica, J. Determinants of high platelet reactivity in patients with acute coronary syndromes treated with ticagrelor. Sci. Rep. 2019, 9, 3924. [Google Scholar] [CrossRef] [Green Version]
  90. Ilardi, F.; Gargiulo, G.; Paolillo, R.; Ferrone, M.; Cimino, S.; Giugliano, G.; Schiattarella, G.G.; Verde, N.; Stabile, E.; Perrino, C.; et al. Impact of chronic kidney disease on platelet aggregation in patients with acute coronary syndrome. J. Cardiovasc. Med. 2020, 21, 660–666. [Google Scholar] [CrossRef]
  91. Bolek, T.; Samoš, M.; Šimonová, R.; Kovář, F.; Fedor, M.; Galajda, P.; Staško, J.; Kubisz, P.; Mokáň, M. Does Pantoprazole Affect the On-Treatment Platelet Reactivity in Patients With Acute STEMI Treated With ADP Receptor Blockers?—A Pilot Prospective Study. Am. J. Ther. 2017, 24, e162–e166. [Google Scholar] [CrossRef] [PubMed]
  92. Ferreiro, J.L.; Ueno, M.; Tello-Montoliu, A.; Tomasello, S.D.; Seecheran, N.; Desai, B.; Rollini, F.; Guzman, L.A.; Bass, T.A.; Angiolillo, D.J. Impact of Prasugrel Reload Dosing Regimens on High On-Treatment Platelet Reactivity Rates in Patients on Maintenance Prasugrel Therapy. JACC Cardiovasc. Interv. 2013, 6, 182–184. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Christ, G.; Hafner, T.; Siller-Matula, J.M.; Francesconi, M.; Grohs, K.; Wilhelm, E.; Podczeck-Schweighofer, A. Platelet Inhibition by Abciximab Bolus-Only Administration and Oral ADP Receptor Antagonist Loading in Acute Coronary Syndrome Patients: The Blocking and Bridging Strategy. Thromb. Res. 2013, 132, e36–e41. [Google Scholar] [CrossRef] [PubMed]
  94. Niazi, A.K.; Dinicolantonio, J.J.; Lavie, C.J.; O’Keefe, J.H.; Meier, P.; Bangalore, S. Triple versus dual antiplatelet therapy in acute coronary syndromes: Adding cilostazol to aspirin and clopidogrel? Cardiology 2013, 126, 233–243. [Google Scholar] [CrossRef]
  95. Bassez, C.; Deharo, P.; Pankert, M.; Bonnet, G.; Quilici, J.; Lambert, M.; Verdier, V.; Morange, P.; Alessi, M.-C.; Bonnet, J.-L.; et al. Effectiveness of switching ‘low responders’ to prasugrel to ticagrelor after acute coronary syndrome. Int. J. Cardiol. 2014, 176, 1184–1185. [Google Scholar] [CrossRef]
Figure 1. Possible mechanism of HTPR on novel-generation ADPRB [59]. ADP—adenosine diphosphate; ADPRB—P2Y12 ADP receptor blockers; CYP—cytochrome P450 enzyme; HTPR—high on-treatment platelet reactivity.
Figure 1. Possible mechanism of HTPR on novel-generation ADPRB [59]. ADP—adenosine diphosphate; ADPRB—P2Y12 ADP receptor blockers; CYP—cytochrome P450 enzyme; HTPR—high on-treatment platelet reactivity.
Jcm 11 07211 g001
Table 1. Novel-generation ADP receptor blockers in current clinical practice.
Table 1. Novel-generation ADP receptor blockers in current clinical practice.
DrugRoute of Administration DosingBioavailabilityReceptor InhibitionTime to Peak Platelet InhibitionClinical ApplicationHTPR
(Prevalence)
PrasugrelOral
Loading dose of 60 mg followed by 10 mg once daily
(5 mg in elderly and low body weight)
ProdrugIrreversible0.5–2 hACS with PCIDescribed
(1.6–25%; higher if time from drug administration to blood sampling is too short)
TicagrelorOral
Loading dose of 180 mg followed by 90 mg twice daily
(60 mg twice daily in CAD)
Direct-actingReversible1.5–2 hACS
High ischemic risk CAD
Described
(8.6–13.7% if tested 30 to 90 days post drug loading; 0.0–1.9% on long-term therapy)
CangrelorIntravenous
Bolus injection of 30 ug/kg followed by continuous intravenous infusion of 4 ug/kg/min.
Direct-actingReversible2 minPCI
(if not pretreated with oral agent)
Not described
ACS—acute coronary syndromes; CAD—coronary artery disease; HTPR—high on-treatment platelet reactivity; PCI—percutaneous coronary intervention.
Table 2. Summary of studies reporting prasugrel HTPR.
Table 2. Summary of studies reporting prasugrel HTPR.
StudyType of StudyStudied PopulationNumber of PatientsTest for HTPRCut OffMain Results
Bonello et al. [45]Prospective multicenter (non-randomized)PCI for ACS301VASP-PVASP-P:
PRI > 50%
HTPR
in 25.2%
of patients; significantly higher PRI in those with thrombotic events
Škorňová et al. [46]Prospective single-center (non-randomized)STEMI with primary PCI44LTA with ADP induction, VASP-PLTA:
>50%,
VASP-P:
PRI > 50%
HTPR
in 8.7% of patients
Aradi et al. [47]Prospective multicenter (non-randomized)high risk ACS104LTA, Multiplate®LTA:
>46%,
Multiplate®: >47 AU
inter-patient variability after prasugrel loading dosing; no effect of PPI on prasugrel activity
Cayla et al. [48]Prospective two high-volume centers (non-randomized)ACS with PCI444VASP-P,
VerifyNow®,
LTA
VASP-P: PRI ≥ 50%,
VerifyNow®:
≥235 PRU,
LTA:
≥46.2%
HTPR
in 3.2–6.8% of patients according to method used for detection
Laine et al. [49]Prospective single-center (randomized)STEMI with primary PCI44VASP-PVASP-P:
PRI ≥ 50%
HTPR
in 9.1% of patients
Lemesle et al. [50]Meta-analysis (14 studies included)CAD1822VASP-P,
VerifyNow®
VASP-P:
PRI ≥ 50%, different cut off for VerifyNow in included studies (208–235 PRU)
HTPR
in 9.8% of patients
Siller-Matula et al. [51]Prospective single-center (non-randomized)ACS200Multiplate®Multiplate®: >46 AUHTPR
in 3% of patients
Selhorst et al. [52]Retrospective single-centerACS with primary PCI809Multiplate®Multiplate®:
>468 AUC
HTPR in 1.6% of patients
Verdoia et al. [53]Prospective single-center (non-randomized)ACS with PCI190Multiplate®Multiplate®:
>417 AUC
HTPR
in 10% of patients
Verdoia et al. [54]Prospective single-center (non-randomized)ACS with PCI105Multiplate®Multiplate®:
>417 AUC
HTPR
in 12.3% of patients
ACS—acute coronary syndrome; ADP—adenosine diphosphate; AU—aggregation units; AUC—area under the curve; CAD—coronary artery disease; HTPR—high on treatment platelet reactivity; LTA—light transmission aggregometry; PCI—percutaneous coronary intervention; PRI—platelet reactivity index; PRU—P2Y12 reactivity units; VASP-P—Vasodilator-Stimulated Phosphoprotein phosphorylation.
Table 3. Summary of studies reporting ticagrelor HTPR.
Table 3. Summary of studies reporting ticagrelor HTPR.
StudyType of StudyStudied PopulationNumber of PatientsTest for HTPRCut OffMain Results
Alexopoulos et al. [60]Prospective single-center (randomized)ACS with PCI and HTPR on clopidogrel44VerifyNow®VerifyNow®:
≥235 PRU
HTPR
in 0%
of patients
Alexopoulos et al. [61]Prospective single-center (randomized)ACS with PCI and T2D30VerifyNow®VerifyNow®:
≥230 PRU
HTPR
in 0% of patients
Laine et al. [62]Prospective multicenter (non-randomized)ACS with PCI115VASP-PVASP-P:
PRI ≥ 50%
HTPR
in 3.5% of patients
Škorňová et al. [46]Prospective single-center (non-randomized)STEMI with primary PCI44LTA with ADP induction, VASP-PLTA:
>50%,
VASP-P:
PRI > 50%
HTPR
in 14.3% of patients
Lemesle et al. [50]Meta-analysis (14 studies included)CAD1822VASP-P,
VerifyNow®
VASP-P:
PRI ≥ 50%, different cut off for VerifyNow
in included studies
(208–235 PRU)
HTPR
in 1.5% of patients
Siller-Matula et al. [51]Prospective single-center (non-randomized)ACS200Multiplate®Multiplate®: >46 AUHTPR
in 2% of patients
Laine et al. [63]Prospective single-center (randomized)ACS with PCI and T2D100VASP-PVASP-P:
PRI ≥ 50%
HTPR
in 6% of patients
Verdoia et al. [64]Prospective single-center (non-randomized)ACS190Multiplate®Multiplate®:
>417 AUC
HTPR
in 11% of patients
Barbieri et al. [65]Prospective single-center (non-randomized)PCI537Multiplate®Multiplate®:
>417 AUC
HTPR
in 12.7% of patients
Li et al. [66]Prospective single-center (non-randomized)ACS176TEG®TEG®:
MA > 47 mm
HTPR
in 3.98% of patients
Laine et al. [67]Prospective multicenter (non-randomized)ACS with PCI530VASP-PVASP-P:
PRI ≥ 50%
HTPR
in 5.3% of patients
Verdoia et al. [54]Prospective single-center (non-randomized)ACS with PCI105Multiplate®Multiplate®:
>417 AUC
HTPR
in 8.6% of patients
Verdoia et al. [68]Prospective single-center (non-randomized)ACS with PCI195Multiplate®Multiplate®:
>417 AUC
HTPR
in 13.3% of patients
Verdoia et al. [69]Prospective single-center (non-randomized)ACS with PCI432Multiplate®Multiplate®:
>417 AUC
HTPR
in 11.4% of patients
Laine et al. [49]Prospective single-center (randomized)STEMI with primary PCI44VASP-PVASP-P:
PRI ≥ 50%
HTPR
in 0% of patients
Selhorst et al. [52]Retrospective single-centerACS with primary PCI809Multiplate®Multiplate®:
>468 AUC
HTPR
in 1.9% of patients
Alexopoulos et al. [70]Prospective single-center (non-randomized)ACS with PCI512VerifyNow®VerifyNow®:
>208 PRU
HTPR
in 0% of patients
Alexopoulos et al. [71]Meta-analysis (8 studies included)CAD or ACS
(with or without PCI)
445VerifyNow®VerifyNow®:
>230 PRU
HTPR
in 0% of patients
Gaglia et al. [72]Prospective single-center (non-randomized)ACS and black rase29LTA,
VASP-P,
VerifyNow®
LTA:
>60%,
VASP-P:
PRI > 50%,
VerifyNow®:
>208 PRU
HTPR
in 0% of patients
Sweeny et al. [73]post-hoc analysis of prospective multicenter study (randomized)ACS (troponin negative) with PCI100VerifyNow®VerifyNow®:
>208 PRU
HTPR
in 5.9% of patients
Liu et al. [74]Prospective multicenter (randomized)NSTE ACS278VASP-PVASP-P:
PRI ≥ 50%
HTPR
in 3.1–5.3% of patients (according to ticagrelor loading dose)
Wen et al. [75]Meta-analysis (14 studies included)ACS2629VASP-P,
VerifyNow®
VASP-P:
PRI ≥ 50%,
VerifyNow®:
≥208 PRU or ≥230 PRU
HTPR
in 0.66–2.67% of patients (according to method used for detection)
Dai et al. [76]Meta-analysis (25 studies included)ACS5098VASP-P,
VerifyNow®,
Multiplate®
Not reportedlow incidence of HTPR on ticagrelor maintenance dosing (exact rate was not reported)
Musallam et al. [7]Case reportstent thrombosis
on ticagleror therapy
1VerifyNow®-HTPR (339 PRU) at the time of stent thrombosis
Malik [77]Case reportstent thrombosis
on ticagleror therapy
1TEG®TEG®:
MA > 47 mm
HTPR (MA of 66 mm) at the time of stent thrombosis
Jariwala et al. [78]Case reportstent thrombosis
on ticagleror therapy
1Not tested-stent thrombosis on ticagrelor—stent-related complication excluded by intravascular coronary imaging
ACS—acute coronary syndrome; ADP—adenosine diphosphate; AU—aggregation units; AUC—area under the curve; CAD—coronary artery disease; HTPR—high on treatment platelet reactivity; LTA—light transmission aggregometry; MA—maximum amplitude; NSTE—non-ST segment elevation; PCI—percutaneous coronary intervention; PRI—platelet reactivity index; PRU—P2Y12 reactivity units; T2D—type 2 diabetes; TEG®—thromboelastography; VASP-P—Vasodilator-Stimulated Phosphoprotein phosphorylation.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Blaško, P.; Samoš, M.; Bolek, T.; Stančiaková, L.; Škorňová, I.; Péč, M.J.; Jurica, J.; Staško, J.; Mokáň, M. Resistance on the Latest Oral and Intravenous P2Y12 ADP Receptor Blockers in Patients with Acute Coronary Syndromes: Fact or Myth? J. Clin. Med. 2022, 11, 7211. https://doi.org/10.3390/jcm11237211

AMA Style

Blaško P, Samoš M, Bolek T, Stančiaková L, Škorňová I, Péč MJ, Jurica J, Staško J, Mokáň M. Resistance on the Latest Oral and Intravenous P2Y12 ADP Receptor Blockers in Patients with Acute Coronary Syndromes: Fact or Myth? Journal of Clinical Medicine. 2022; 11(23):7211. https://doi.org/10.3390/jcm11237211

Chicago/Turabian Style

Blaško, Peter, Matej Samoš, Tomáš Bolek, Lucia Stančiaková, Ingrid Škorňová, Martin Jozef Péč, Jakub Jurica, Ján Staško, and Marián Mokáň. 2022. "Resistance on the Latest Oral and Intravenous P2Y12 ADP Receptor Blockers in Patients with Acute Coronary Syndromes: Fact or Myth?" Journal of Clinical Medicine 11, no. 23: 7211. https://doi.org/10.3390/jcm11237211

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