IMR Press / RCM / Volume 23 / Issue 6 / DOI: 10.31083/j.rcm2306207
Open Access Review
Antiplatelet Therapy in High-Bleeding Risk Patients Undergoing PCI: Walking a Tightrope
Show Less
1 The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, NY 10029-6574, US
2 Cardiovascular Department, Humanitas Gavazzeni, 24125 Bergamo, Italy
*Correspondence: roxana.mehran@mountsinai.org (Roxana Mehran)
Academic Editor: Federico Ronco
Rev. Cardiovasc. Med. 2022, 23(6), 207; https://doi.org/10.31083/j.rcm2306207
Submitted: 24 February 2022 | Revised: 20 April 2022 | Accepted: 9 May 2022 | Published: 1 June 2022
(This article belongs to the Special Issue Recent Advances in Percutaneous Coronary Intervention)
Copyright: © 2022 The Author(s). Published by IMR Press.
This is an open access article under the CC BY 4.0 license.
Abstract

Historically, prevention from ischemic events with dual antiplatelet therapy (DAPT) post percutaneous coronary intervention (PCI) took precedence over protection from bleeding. However, increasing data suggest that major bleeding complications are as detrimental as ischemic events. Awareness about the prognostic impact of bleeding prompted the search for new strategies aimed at maximizing both ischemic and bleeding protection. This is noteworthy because patients at high bleeding risk (HBR) have generally been underrepresented in clinical trials on DAPT and they often are at increased risk of ischemic events as well. The present review discusses the evidence base for new pharmacotherapeutic strategies to decrease bleeding risk without compromising ischemic protection among HBR patients undergoing PCI, including shortening DAPT duration, early aspirin withdrawal, and P2Y12 inhibitor de-escalation.

Keywords
high bleeding risk
HBR
percutaneous coronary intervention
antiplatelet therapy
antithrombotic therapy
DAPT
1. Introduction

Antiplatelet agents constitute the foundation therapy for secondary prevention of thromboembolic events after percutaneous coronary intervention (PCI) with drug-eluting stent (DES) [1]. Guidelines currently recommend, the use of dual antiplatelet therapy (DAPT), a combination of aspirin and a P2Y12 receptor inhibitor, following DES implantation for at least 6 months in patients with stable coronary artery disease (CAD) and 12 months in cases of acute coronary syndrome (ACS) [2, 3]. Historically, prevention of ischemic events and ST elevations took precedence over protection from bleeding, leading to studies exploring DAPT regimens greater than 12 months [4]. Recently, several studies revealed that major bleeding complications related to prolonged DAPT carry a similar prognostic impact as ischemic events [5, 6, 7]. Furthermore, the introduction of newer generation DES with thinner struts and more biocompatible polymers decreased the risk of stent-related adverse events thus providing a rationale for shorter DAPT regimens. Therefore, an antithrombotic therapy strategy that mitigates the bleeding risk while maintaining ischemic protection seems most desirable in contemporary PCI practice. Balancing the ischemic and bleeding risks becomes even more challenging in patients with multiple comorbidities and particularly those at high bleeding risk (HBR). There is a scarcity of data regarding the optimal antiplatelet strategy in HBR patients as they were either excluded or underrepresented in most randomized controlled trials (RCT) that shaped contemporary guidelines [8, 9]. Aiming at decreasing the bleeding complications associated with prolonged DAPT, especially among HBR patients, several studies investigated novel antithrombotic therapy strategies that shorten DAPT duration or decrease its intensity over time. In this review, we aim to define HBR patients, discuss bleeding risk stratification tools, and review recent advances in post-PCI pharmacotherapy.

1.1 High-Bleeding Risk Patients

Advancements in PCI technologies have allowed extending this treatment option to high-risk patients who were traditionally managed conservatively. These patients typically have extensive CAD and multiple comorbidities that, not only increase their risk for thromboembolic events, but also for bleeding complications. Indeed, a study of an all-comer population undergoing DES implantation has shown that as high as 1 in every 15 patients experienced post-discharge bleeding at a median of 300 days after the procedure [5]. Interestingly, the impact of bleeding on two-year mortality was significantly larger compared with post-discharge MI [5]. This study, together with other observational studies, shed light on the prognostic relevance of post-discharge bleeding after PCI [10].

Finding patients at HBR is of highest importance for the management of antithrombotic therapy after PCI. Nonetheless, a lack of standardization in defining this population limits the generalizability of trial results as well as clinical decision-making. Based on review of the literature, the Academic Research Consortium (ARC) recently published an agreement definition of patients at HBR based on fulfillment of specific criteria (Fig. 1, Ref. [11]) [12]. Several registry-based studies have validated the ARC-HBR definition by showing an incidence of Bleeding Academic Research Consortium (BARC) 3 or 5 bleeding risk of 4% in HBR patients at one year after PCI [13, 14, 15]. Moreover, these studies revealed that HBR patients account for around a third of all PCI patients, reiterating the need for tailored antiplatelet therapy strategies that mitigate the bleeding risk.

Fig. 1.

ARC-HBR definition of HBR. Major and minor risk factors used in the definition for HBR [11]. 1 major or 2 minor criteria qualify as HBR. DAPT, dual anti-platelet therapy; eGFR, estimated glomerular filtration rate; Hb, hemoglobin; NSAID, non-steroidal anti-inflammatory drugs; OAC, oral anti-coagulant.

1.2 Contemporary Bleeding Risk Scores

Over the past years, numerous risk scores have been designed to inform and guide decision making on DAPT duration and intensity after PCI. The European Society of Cardiology guidelines recommend using the DAPT and PRECISE-DAPT risk scores [16, 17]. The DAPT scoring system was developed using predictors of both ischemic and bleeding events to identify patients who derive the greatest benefit over harm from prolonging DAPT beyond 12 months of PCI [18]. Conversely, the PRECISE-DAPT was developed to assess the risk of out of hospital bleeding up to 2 years post-PCI [19]. The PARIS score encompasses two separate prediction models to evaluate ischemic and bleeding risks after PCI [20]. Although these scores share similar components, each has its own features (Table 1). The DAPT score included somewhat lower risk patients who were event-free at 12 months post-PCI, while both PRECISE-DAPT and PARIS included patients immediately after discharge of index PCI. While HBR patients composed approximately 25% of all subjects considered, each scoring system identified different rates of bleeding ranging from 1% to 10% [11]. Therefore, it has been difficult to confidently use such scoring systems in HBR patients undergoing PCI. Recently, Urban et al. [12] developed the ARC-HBR trade off model, which predicts the risk of non-periprocedural major bleeding and thrombotic events at one year among HBR patients who have undergone PCI. Although this was the first risk score especially dedicated to HBR patients, it should be noted that this tool was derived from studies using different DAPT durations (i.e., driven by the protocol of the study or guideline-based) and recommendations should not be solely made based on its risk predictions.

Table 1.Various bleeding risk scores.
REACH-39 DUTCH ASA Score37 DAPT41 PARIS38 PRECISE-DAPT32 BleeMACS36
Year 2010 2014 2016 2016 2017 2018
Database REACH Dutch ASA registry DAPT randomized trial PARIS 8 randomized trials BleeMACS
Number 56616 235531 11648 4190 14963 15401
Population Risk of atherosclerosis New low-dose ASA Post-PCI patients event free 12 mo after index All PCI Patients undergoing PCI ACS undergoing PCI
Definition Non-fatal hemmorhage or bleeding leading to both hospitalization and transfusion at 2 years UGIB at median follow up 530 d GUSTO moderate or severe bleeding BARC 3 or 5 after 2 y TIMI major or minor with median follow up 552 d Intracranial bleed or any bleed requiring hospitalization or transfusion at 1 year
Bleeding risk score factors Age, PAD, CHF, DM, HLD, HTN, Smoking, Anti-platelet, OAC Age, Anemia, DM, Other anti-platelet, OAC Age, PAD, HTN, Renal insuffiency Age, BMI, Anemia, Triple therapy, Smoking, Renal insuffiency Age, Previous bleed, WBC, Hb, Cr clearance Age, HTN, PAD, Prior bleed, Malignancy, Cr clearance, Hb
Validation discrimination AUC 0.68 AUC 0.64 AUC 0.68 AUC 0.72 AUC 0.73 AUC 0.71
External validation CHARISMA Dutch Health Insurance Database PROTECT ADAPT-DES PLATO and BernPCI Registry SWEDEHEART
External validation discrimination AUC 0.64 AUC 0.63 AUC 0.64 AUC 0.64 AUC 0.70 and 0.66 AUC 0.65
ASA, aspirin; AUC, area under curve; BARC, Bleeding Academic Research Consortium; BMI, body mass index; CHF, congestive heart failure; Cr, creatinine; DAPT, dual antiplatelet therapy; DM, diabetes mellitus; GUSTO, global utilization of streptokinase and TPA for occluded arteries; Hb, hemoglobin; HTN, hypertension; HLD, hyperlipidemia; OAC, oral anticoagulant; PAD, peripheral artery disease; PCI, percutaneous coronary intervention; TIMI, thrombolysis in myocardial infarction; UGIB, upper gastrointestinal bleed; WBC, white blood cell.
2. Anti-Thrombotic Strategies
2.1 Shortening DAPT Duration

The paradigm in interventional cardiology research has shifted over the past few years into testing strategies that unite modern DES platforms with short DAPT durations (Table 2, Ref. [21, 22, 23, 24, 25]). LEADERS-FREE was a randomized double blind trial comparing outcomes of HBR patients receiving the polymer-free biolimus-eluting BioFreedom stent vs. a similar bare metal stent (BMS); patients in both arms were maintained on DAPT for one month after PCI [21]. The BioFreedom stent was found to be superior to BMS with regards to the composite of cardiac death, MI, or stent thrombosis, largely driven by decreased rates of MI. Conversely, the ONYX ONE trial examined the same BioFreedom stent in comparison to the durable-polymer zotralimus-eluting Resolute Onyx stent in a similar HBR population [22]. The Resolute Onyx stent was found to be non-inferior to the BioFreedom stent with respect to the same primary outcome as above. However, since both the LEADERS-FREE and ONYX ONE trials preceded the ARC-HBR consensus, definitions of HBR differed, making comparisons among studies difficult to interpret. Most notably, LEADERS-FREE and ONYX ONE considered age 75 alone as HBR criterion for study inclusion. Despite this, improvements in stent technologies have clear benefits in patients at HBR, most notably in the ability to decrease DAPT duration. The SENIOR trial examined outcomes of elderly patients 75 years old by randomizing the bioabsorbable-polymer everolimus-eluting Synergy stent vs. BMS followed by shortened DAPT (1 month in stable patients and 6 months if unstable). DES was superior regarding the primary outcome of all-cause death, MI, stroke, or ischemia driven target vessel revascularization at one year, mostly driven by the latter [23]. Additionally, the DEBUT trial examined whether drug-coated stents were non-inferior to BMS in HBR patients [26]. Not only did they determine that drug-eluting stents were non-inferior to BMS with respect to major adverse cardiac events (MACE) after 9 months, they found superiority. Although these 4 studies showed that shortening DAPT duration is a safe and effective strategy in HBR patients who undergo PCI with new-generation DES, optimal therapy durations cannot be determined based on these trial designs.

Table 2.Trials of devices with short DAPT.
Trial N Population DAPT Intervention Control Primary outcome Result
LEADERS-FREE [21] 2466 CAD requiring PCI 1 month BioFreedom DCS BMS 1: Cardiac death, MI, or stent thrombosis 1: 9.4% vs. 12.9%, HR 0.71, 95% CI 0.56–0.91, p < 0.001
2: TLR 2: 5.1% vs. 9.8%, HR 0.5, 95% CI 0.37–0.69, p < 0.001
SENIOR [23] 1200 75 yo, stable angina, or ACS 1 month stable CAD, 6 months ACS Synergy DES BMS MACCE at 1 year 12% vs. 16%, RR 0.71, 95% CI 0.52–0.94, p = 0.02
ONYX-ONE [22] 1996 HBR patients undergoing PCI 1 month Resolute Onyx DES BioFreedom DCS Non-inferiority for cardiac death, TVMI, TLR at 1 year 17.1% vs. 16.9%, p = 0.01
EVOLVE Short DAPT [25] 2009 HBR patients with stable or unstable angina 3 months SYNERGY DES DES 1: Death or MI 1: 5.6% vs. 5.7%, p = 0.0016 non-inferiority
2: stent thrombosis 2: 0.2%, p = 0.0005 for comparison to 1% performance goal
XIENCE 28 [24] 1392 HBR patients undergoing PCI 1 month XIENCE DES DES 1: Death or MI between 1 and 6 months 1: 3.5% vs. 4.2%, p < 0.0005 non-inferiority
2: BARC 2,3,5 bleeding between 1 and 6 months 2: 4.9% vs. 5.9%, p = 0.19
XIENCE 90 [24] 1693 HBR patients undergoing PCI 3 months XIENCE DES DES 1: Death or MI between 3 and 12 months 1: 5.4% vs. 5.4%, p < 0.0063 non-inferiority
2: BARC 2,3,5 bleeding between 3 and 12 months 2: 5.1% vs. 7.0%, p = 0.0687
3. Stent thrombosis between 3 and 12 months 3: 0.2%, p < 0.0001 for 1.2% performance goal
ACS, acute coronary syndrome; BARC, Bleeding Academic Research Consortium; BMS, bare metal stent; CAD, coronary artery disease; CI, confidence interval; DAPT, dual anti-platelet therapy; DCS, drug coated stent; DES, drug-eluting stent; HBR, high bleeding risk; HR, hazard ratio; MACCE, major adverse cardiac and cerebrovascular events; MI, myocardial infarction; N, Number of patients; PCI, percutaneous coronary intervention; RR, relative risk; TLR, target lesion revascularization; TVMI, target vessel myocardial infarction.

More recently, the EVOLVE Short DAPT registry enrolled n = 1437 HBR patients treated with the Synergy stent followed by 3-month DAPT [25]. Such short DAPT duration was found to be non-inferior to a historical cohort of patients treated with 12-month DAPT with respect to death or MI but failed to show and advantage in terms of bleeding. Notably, the study was non-randomized and the control group was not uniform as it included multiple different stent types, potentially limiting the generalizability of the results. The XIENCE Short DAPT program included 3 registries (XIENCE 28 Global and 28 USA, and XIENCE 90) for a total of n = 3652 HBR patients undergoing PCI with the fluoropolymer-based cobalt-chromium Everolimus-eluting Xience stent who discontinued DAPT at 1 or 3 months post-PCI if event-free and treatment-adherent [27]. Both short DAPT regimens (1 and 3 months) were non-inferior to standard DAPT (6 to 12 months) with respect to death or MI and superior with respect to major bleeding, after propensity score-stratification vs. an historical group of patients receiving the same stent [24]. In a subsequent exploratory analysis from the XIENCE data, 1 month of DAPT was shown to have comparable ischemic outcomes and lower bleeding risk compared with 3-month DAPT [28]. MASTER DAPT was the first RCT testing different DAPT durations in HBR patients treated with a new-generation DES. The trial included 4434 HBR patients who underwent placement of the biodegrable-polymer sirolimus-eluting Ultimaster stent [29]. Subjects who were event free after 1 month of index PCI were either randomized to DAPT discontinuation followed by either aspirin or a P2Y12 inhibitory monotherapy or continuation of DAPT for at least 5 additional months. The short DAPT (1 month) regimen was shown to be non-inferior to prolonged DAPT with regard to net adverse clinical events and MACE [29]. Specifically, the composite of major or clinically relevant bleeding was observed in 6.5% in the experimental group as compared to 9.4% in the control without tradeoff in ischemic events. Therefore, shortening DAPT duration to 1 to 3 months may be a reasonable approach in selected HBR patients, pending additional data from large clinical trials.

2.2 P2Y12 Inhibitor Monotherapy

Aspirin has been the mainstay therapy for long-term secondary prevention of ischemic events for decades. Recently, its undisputed benefits have been challenged for several reasons: (1) increased risk of intracranial and extracranial bleeding, especially in HBR patients, (2) widespread use of optimal medical therapy including disease-modifying drugs (i.e., angiotensin converting enzyme-inhibitors, angiotensin receptor blockers, statins, etc.), and (3) the introduction of more potent antiplatelet agents. However, the introduction of new antiplatelet agents in PCI practice has always requested these new agents to prove their benefits on a background of aspirin therapy such that their individual effects have never been truly assessed. The PLATO trial showed that the more potent P2Y12 inhibitor, ticagrelor, is superior to clopidogrel in reducing ischemic events at 12 months among ACS patients, although at the cost of increased bleeding [30]. To note, PLATO also suggested that low dose (<300 mg) aspirin was more effective than a high dose (300 mg) in preventing ischemic events when combined with ticagrelor [31]. This raised the question as to whether aspirin is at all needed in presence of potent P2Y12 inhibitors [32, 33]. The GLOBAL LEADERS trial addressed this question in an all-comer population of patients undergoing PCI for stable CAD or ACS [34]. It randomized over 15,000 patients to either ticagrelor monotherapy for 23 months after 1 month of DAPT or 12 months of DAPT followed by aspirin monotherapy. Ticagrelor monotherapy was not superior to 12-month DAPT for the primary endpoint of all-cause death and new Q-wave MI (3.81% in experimental vs. 4.37% in control; p = 0.073) and was associated with similar rates of bleeding events [34]. The study had several limitations including an open label design [35]. GLASSY, a GLOBAL LEADERS adjudication substudy conducted at the top-10 enrolling sites (n = 7585) yielded similar conclusions as the parent trial but for the first time suggested a reduction in thrombotic events (MI and ST) with ticagrelor monotherapy vs. aspirin between 1 and 2 years post-PCI [36]. The SMART CHOICE study showed P2Y12 inhibitor monotherapy after 3-month DAPT was non-inferior to the standard treatment of 12 months with respect to major adverse cardiac and cerebrovascular events (MACCE) a composite of all cause death, stroke, MI [37]. In line with previous studies, they found a decrease in bleeding rates, specifically bleeding BARC 2-5, but did not find any differences in major bleeding [37]. Unlike the GLOBAL LEADERS trial, SMART CHOICE included multiple P2Y12 inhibitors with clopidogrel the most frequently used, giving evidence to its benefit as a monotherapy, similar to STOPDAPT2. Although this study was conducted in a low-risk Asian population, decreasing the bleeding risks would most certainly be beneficial to those at HBR. Similar results were found in the STOPDAPT-2 trial where they found that 1 month of DAPT followed by clopidogrel monotherapy was superior to 12 months of DAPT in a composite of cardiovascular and bleeding events, largely driven by a reduction in bleeding [38]. However, the trial included mainly low-risk Japanese patients with very high rates of intravascular imaging use, therefore its generalizability has been questioned. More recently, the STOPDAPT-2 ACS trial, an extension of STOPDAPT-2, enrolling only ACS patients, showed that 1-month DAPT followed by clopidogrel monotherapy was not non-inferior with respect to net adverse events (including ischemic and bleeding endpoints) when compared to standard DAPT [39]. These results were driven by a significant decrease in the occurrence of major bleeding events, which was offset by a concomitant increase in ischemic events. The TICO trial further investigated ticagrelor monotherapy compared to DAPT among patients with ACS. Although event rates were lower than expected, ticagrelor monotherapy after 3 months of DAPT decreased the incidence of net adverse clinical events (composite of major bleeding and adverse cardiac and cerebrovascular events) [40]. Again, this difference was mostly driven by a decrease in bleeding complications, with no tradeoff in ischemic events. Even though this study excluded patients at HBR, it further supported the use of P2Y12 inhibitor monotherapy, reducing overall risks of bleeding.

The TWILIGHT study examined ticagrelor monotherapy following 3-month DAPT in high-risk patients undergoing PCI. Patients were considered at high risk for ischemic and bleeding events if they fulfilled at least one clinical and one angiographic high-risk feature. This double-blinded placebo-controlled study randomized patients to receive either ticagrelor monotherapy or ticagrelor plus aspirin for 12 months after being event free for 3 months post PCI. Ticagrelor monotherapy was shown to reduce the incidence of the primary endpoint of BARC 2, 3, or 5 bleeding without an increase in ischemic events [41]. A sub-analysis of the TWILIGHT trial looking at patients who qualify as HBR based on ARC-HBR criteria showed consistent results, with larger absolute risk reduction in major bleeding observed with ticagrelor monotherapy in HBR versus non-HBR patients [42]. Several meta-analyses of the above studies showed decreased risks of bleeding, while no concomitant increase in events [43, 44, 45]. Although many of these studies do not specifically examine HBR patients, a short DAPT duration followed by P2Y12 inhibitor monotherapy emerged as a safe and effective bleeding-avoidance strategy, although extra caution might be needed in those presenting with ACS (Table 3, Figs. 2,3, Ref. [29, 34, 37, 38, 40, 41, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61]).

Fig. 2.

Trial designs of shortening DAPT after PCI regardless of stent design. Comparison of studies of short DAPT duration regardless of specific stent designs [34, 37, 41, 46, 47, 48, 49, 50, 51]. Blue arrows indicate duration of DAPT, red arrows indicate duration of monotherapy. DAPT, dual antiplatelet therapy.

Fig. 3.

Short DAPT trial with specific stent designs. Comparison of studies with short DAPT designs with specific stent designs [29, 38, 40, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61]. Blue arrows indicate duration of DAPT, red arrows indicate duration of monotherapy. BES, biolimus eluting stent; BP-BES, bioresorbable polymer biolimus-eluting stent; BP-DES, bioresorbable polymer drug-eluting stent; BP-EES, bioresorbable polymer everolimus-eluting stent; BP-SES, bioresorbable-polymer sirolimus-eluting stent; CoCr-EES, cobalt chromium everolimus-eluting stent; DAPT, dual antiplatelet therapy; DP-EES, durable polymer everolimus-eluting stent; E-ZES, endeavor zotarolimus-eluting stent; PF-DCS, polymer free drug coated stent; ZES, zotarolimus-eluting stent. * indicates experimental group. † indicates control group.

Table 3.Trials including anti-platelet monotherapies.
Trial N Population Major inclusion and exclusion criteria Intervention Control Primary outcome Result
GLOBAL LEADERS [34] 15968 Stable CAD or ACS with biolimus A9-eluting stent Inclusion: 50% of more stenosis in 1 coronary ASA + ticagrelor for 1 month followed by 23 months ticagrelor monotherapy DAPT for 12 months followed by ASA monotherapy Composite of all-cause mortality or non-fatal new Q-wave MI at 2 years 3.81% vs. 4.37%, RR 0.87, 95% CI 0.75–1.01, p = 0.073
Exclusion: Chronic oral anti-coagulation
STOPDAPT-2 [38] 3045 PCI Inclusion: PCI with CoCr-EES stent without complications post PCI 1 month DAPT followed by clopidogrel monotherapy DAPT Composite of CV death, MI, stroke, stent thrombosis, major or minor bleeding at 1 year 2.36% vs. 3.70%, HR 0.64, 95% CI 0.42–0.98, p < 0.01 non-inferiority, and p = 0.04 superiority
Exclusion: Need for oral anticoagulation, history of intracranial bleeding
SMART-CHOICE [37] 2993 PCI with DES placement Inclusion: 50% or more stenosis in 1 coronary DAPT for 3 months followed by monotherapy DAPT Composite of death, MI, stroke at 1 year 2.9% vs. 2.5%, one-sided 95% CI -∞-1.3%, p = 0.007 non-inferiority
Exclusion: Cardiogenic shock, active bleeding
TWILIGHT [41] 7119 High risk for bleeding or ischemia undergoing PCI Inclusion: 1 clinical feature and one angiographic feature with high risk ischemia or bleeding events 3 months DAPT followed by monotherapy DAPT 1: BARC 2,3 or 5 bleeding at 1 year 1: 4.0% vs. 7.1%, HR 0.56, 95% CI 0.45–0.68, p < 0.00
Exclusion: STEMI, cardiogenic shock, oral anticoagulation 2: Composite of death, MI, stroke 2: 3.9% vs. 3.9%, HR 0.99, 95% CI 0.78–1.25, p < 0.001 non-inferiority
TICO [40] 3056 ACS requiring PCI Inclusion: PCI with Orsiro stent for ACS Ticagrelor monotherapy after 3 months DAPT DAPT Composite of major bleeding, death, MI, stent thrombosis, stroke, or TVR at 1 year 3.9% vs. 5.9%, HR 0.66, 95% CI 0.48–0.92, p = 0.01
Exclusion: prior hemorrhagic stroke, internal bleeding in last 6 weeks, hemoglobin 8 g/dL
MASTER-DAPT [29] 4434 HBR receiving TANSEI DES Inclusion:1 high bleeding risk criteria, PCI with TANSEI stent 1 month DAPT followed by ASA or P2Y12 monotherapy DAPT 1: NACE 1: 7.5% vs. 7.7%, AD –0.23%, 95% CI –1.8–1.33, p < 0.001 non inferiority
Exclusion: Treatment for ISR, BARC 2 bleeding 2: MACCE 2: 6.1% vs. 5.9%, AD 0.11%, 95% CI –1.29–1.51, p = 0.001 non-inferiority
3: MCB at 12 months 3: 6.4% vs. 9.2%, AD –2.78%, 95% CI –4.37 to –1.20, p < 0.001 superiority
ACS, acute coronary syndrome; AD, absolute difference; ASA, aspirin; BARC, Bleeding Academic Research Consortium; BMS, bare metal stent; CAD, coronary artery disease; CoCr-EES; cobalt chromium everolimus-eluting stent; CI, confidence interval; CV, cardiovascular; DAPT, dual anti-platelet therapy; DES, drug-eluting stent; HBR, high bleeding risk; HR, hazard ratio; ISR, in-stent restenosis; MACCE, major adverse cardiac and cerebrovascular events; MCB, major or clinically relevant bleeding; MI, myocardial infarction; N, Number of patients; NACE, net adverse clinical events; PCI, percutaneous coronary intervention; RR, relative risk; TVR, target vessel revascularization.
2.3 DAPT Modulation by De-Escalation
2.3.1 De-Escalation Tactics: Unguided

In patients presenting with ACS, guidelines recommend DAPT with a potent P2Y12 inhibitor (i.e., ticagrelor or prasugrel) and aspirin for about 12 months [62]. However, the benefits of potent P2Y12 inhibitors are mostly observed in the acute phase post PCI (i.e., the first 30 days) when the risk of ischemic events is highest. However, this risk decreases overtime while bleeding persists and proportional to the duration and intensity of antiplatelet therapies [63]. As a result, investigators have hypothesized that de-escalating therapy, such as switching to a less potent P2Y12 inhibitor or using a lower dose of the same agent after an initial course of potent DAPT, would mitigate bleeding risk without compromising patient safety (Fig. 4, Ref. [64, 65, 66, 67]). The TOPIC trial showed that patients with ACS had decreased risks of bleeding without increase in ischemic events when switching from a more potent P2Y12 inhibitor to clopidogrel one month after PCI [48]. This study, however, was limited by a small sample size (n = 646) with low protocol adherence. The HOST-REDUCE-POLYTECH-ACS trial showed similar results with decreasing dosages of prasugrel [65]. They found that decreasing the dose from 10 mg to 5 mg one month after PCI in ACS patients was associated with a significant decrease in net adverse clinical events, mainly driven by significant reductions in bleeding [65]. Although promising, the study findings may not be generalizable since the trial only included East Asian patients with different ischemic-bleeding risk profiles and a variable response to antiplatelet agents as compared with Western populations [68]. Prasugrel is however contraindicated in older (75) and lower body weight (<60 kg) patients who, therefore, had to be excluded from this trial [65, 69]. Since age greater than 75 is an HBR criterion, it may be difficult to extrapolate this data to HBR patients [11].

Fig. 4.

Guided and Unguided de-escalation trials. Comparison of trial designs of guided and unguided de-escalation strategies. Blue arrows indicate duration of potent therapies, red arrows indicate duration of less potent therapies (i.e., lower doses of prasugrel or ticagrelor vs. changing from prasugrel or ticagrelor to clopidogrel) [64, 65, 66, 67].

2.3.2 De-Escalation Tactics: Guided

De-escalation of antiplatelet therapy can be guided by platelet function testing (PFT) or genetic testing. Although multiple modalities have been developed to determine platelet function, they all serve the purpose to determine how well platelets work to stop bleeding. They determine the residual ability of platelets to aggregate after doses of antiplatelet drug therapy Patients on P2Y12 inhibitors have decreased platelet reactivity; those with high platelet reactivity, despite the use of clopidogrel, should be maintained on more potent antiplatelet agents [70, 71]. As a result, it was thought that PFT could play a role in guiding escalation or de-escalation of therapy. The non-inferiority TROPICAL-ACS study randomized patients to standard therapy (12 months of DAPT with prasugrel) or de-escalation based on PFT results [66]. The experimental group received 1 week of prasugrel after discharge, followed by 1 week of clopidogrel. PFT was performed one week after initiation of clopidogrel, or 2 weeks post-discharge. If patients were found to have high platelet reactivity, they were escalated to prasugrel, otherwise were maintained on clopidogrel. The trial met non-inferiority with respect to net clinical benefit in the de-escalation group [66]. Although not statistically significant, the study observed a decrease in bleeding events in the de-escalation group compared with standard treatment.

The variable platelets reactivity to clopidogrel may lead to suboptimal antithrombotic protection [72]. Loss of function alleles, specifically the CYP2C19*2 and CYP2C19*3 alleles, have been identified as genetic causes for the decreased response to clopidogrel and its decreased efficacy [73, 74]. In those without this loss of function allele, clopidogrel was found to be equally effective as more potent P2Y12 inhibitors [75, 76]. The POPular Genetics study investigated if this gene identification could be utilized in guiding DAPT selection to decrease the risks of bleeding and ischemic events [67]. Patients were randomized to early genetic testing or standard DAPT with ticagrelor or prasugrel. Those with the loss of function allele were started on ticagrelor or prasugrel while non-carriers received clopidogrel. After 12 months, the genotype-guided group was non-inferior to standard treatment with regards to net adverse clinical events and major bleeding. A meta-analysis of 11 RCTs and 3 observational studies including 20,743 patients examined guided therapies compared to standard therapy, noting a significant decrease in the risk of MACE as well as lower rates of bleeding events though statistically non-significant [77]. However, the outcomes varied widely based on whether therapies were either escalated, resulting in decreased ischemic events and no increase in bleeding, or de-escalated, resulting in reduction in bleeding with no increase in ischemic events [77]. A network meta-analysis, which included 15 RCTs incorporating multiple different de-escalation strategies, demonstrated that DAPT de-escalation reduced risk of major or minor bleeding when compared to clopidogrel, ticagrelor, standard-dose prasugrel, and low dose prasugrel. There was also no change in composite of cardiovascular death, MI, and stroke [78]. Another study, including 5 RCTs of ACS-only patients corroborated these results with both guided and unguided de-escalation strategies [79]. These meta-analyses have all shown benefits of de-escalation, but as of now, no studies have examined differences among de-escalation therapies and P2Y12 inhibitor monotherapies, making it difficult to understand which strategy is preferred. Regardless, guided de-escalation therapies have clear benefits in preventing major bleeding, while preserving the safety of continued DAPT therapies (Table 4, Fig. 4, Ref [64, 65, 66, 67, 80]). However, PFT testing is not routinely performed or recommended as it has not shown a consistent clinical benefit [81]. Furthermore, genetic testing is not currently recommended either, as testing is expensive and there are many other factors that may play into the variability of clopidogrel’s effectiveness. Although there are currently no studies specifically aimed at examining de-escalation therapies in HBR patients, these therapies seem sensitive in this vulnerable population.

Table 4.Trials of guided or unguided de-escalation strategies.
Trial N Population Major inclusion and exclusion criteria Intervention Control Primary outcome Result
TOPIC [64] 646 ACS requiring PCI Inclusion: ACS, no adverse events at 1 month after ACS Switch to ASA + clopidogrel if 1 month event free after index PCI Previous DAPT Composite of cardiovascular death, urgent revascularization, stroke, and BARC 2 bleeding at 1 year post ACS 13.4% vs. 26.3%, HR 0.48, 95% CI 0.34–0.68, p < 0.01
Exclusion: History of intracranial bleeding, thrombocytopenia, long term anticoagulation
TROPICAL-ACS [66] 2610 ACS requiring PCI and 12 months DAPT Inclusion: ACS, planned treatment of prasugrel 12 months after PCI Stepdown with 1 week prasugrel followed by 1 week clopidogrel and PFT guided maintenance therapy DAPT with ASA + Prasugrel Non-inferiority. Composite of cardiovascular death, myocardial infarction, stroke, or BARC 2 bleeding 1 year after randomization 7% vs. 9%, HR 0.81, 95% CI 0.62–1.06, p = 0.004 non-inferiority
Exclusion: Cardiogenic shock in last 2 weeks, oral anticoagulation, indication for surgery
POPular-Genetics [67] 2488 Primary PCI with stent Inclusion: Treated MI within 12 hours of symptoms Genetic testing determining P2Y12 inhibitor therapy DAPT 1: NACE 1: 5.1% vs. 5.9%, AD: –0.7 percentage points, 95% CI –2.0–0.7, p < 0.001
Exclusion: Malignancy with increase in bleeding, dialysis, severe HTN, cardiogenic shock 2: major or minor PLATO bleeding 2: 9.8% vs. 12.5%, HR 0.78, 95% CI 0.61–0.98, p = 0.04
TAILOR-PCI [80] 1849 Carriers of CYP2C19 undergoing PCI for ACS or stable CAD Inclusion: ACS or stable CAD Ticagrelor DAPT in carriers Clopidogrel DAPT in non-carriers Composite of CV death, MI, stroke, stent thrombosis, severe recurrent ischemia at 1 year 4.0% vs. 5.9%, HR 0.66, 95% CI 0.43–1.02, p = 0.06
Exclusion: Known CYP2C19, Cr 2.5, history intracranial bleeding
HOST-REDUCE-POLYTECH ACS [65] 2338 ACS requiring PCI Inclusion: Clinical ACS of 1 coronary lesion low dose prasugrel + ASA after 1 month DAPT DAPT Composite of death, MI, stent thrombosis, repeat revascularization, stroke, BARC 2 bleeding at 1 year 7.2% vs. 10.1%, HR 0.70, 95% CI 0.52–0.92, p < 0.0001 non-inferiority
Exclusion: major or active bleeding
ACS, acute coronary syndrome; AD, absolute difference; ASA, aspirin; BARC, Bleeding Academic Research Consortium; CAD, coronary artery disease; CI, confidence interval; Cr; creatinine; CV, cardiovascular; DAPT, dual anti-platelet therapy; HR, hazard ratio; HTN, hypertension; MI, myocardial infarction; N, Number of patients; NACE, net adverse clinical events; PCI, percutaneous coronary intervention; PLATO, PLATelet inhibition and patient Outcomes.
3. Special Clinical Settings

Coronary artery bypass graft (CABG) surgery is usually the mainstay of therapy among patients with left main (LM) disease. However, with recent advances in intravascular imaging and coronary stent technologies, PCI has become a safe and viable alternative to surgical management [82, 83, 84]. Stenting of LM lesions are connected with an increased incidence of ischemic events and thus a prolonged DAPT duration is usually required [85, 86]. Nonetheless, a significant proportion of patients undergoing PCI for LM disease are at HBR [87]. Data on optimal DAPT in HBR patients undergoing PCI for LM disease remains sparse. A complex PCI sub-study of TWILIGHT included patients with left main disease, as well as bifurcation lesions treated with two stents, 3 vessels treated, 3 or more lesions treated, total stent length of >60 mm, atherectomy device use, surgical bypass graft or chronic total occlusion (CTO). After 3 months of standard DAPT, Ticagrelor monotherapy was shown to decrease risk of bleeding without an increase in ischemic events when compared to standard DAPT [88].

Additionally, DAPT in PCI for CTO remains limited, especially in the HBR population. However, a study looking at over 1000 patients undergoing PCI for CTO showed increased rates of death and MI among short (<12 months) vs. prolonged (12 months) DAPT duration [89]. Conversely, another study including 500 patients who underwent PCI for CTO showed similar ischemic and bleeding outcomes, irrespective of DAPT duration [90]. As these studies were largely underpowered, we still lack strong evidence supporting the use of a short DAPT regimen in HBR patients undergoing PCI for CTO.

4. Future Perspectives

With increasing emphasis on the prognostic relevance of bleeding events after PCI, the need for studies designed to examine various antiplatelet strategies in HBR patients is paramount. Over the past few years, more studies have investigated antithrombotic therapy in HBR patients undergoing PCI. Although a lot of knowledge has been generated, there remain many unanswered questions. Indeed, the optimal duration and intensity of DAPT after PCI in HBR patients is yet to be defined. Shorter or longer DAPT durations might be beneficial in specific HBR subgroups, depending on the risk-benefit tradeoff of individual patients. In order to allow for direct comparison among RCTs, studies should adopt similar criteria to define HBR, used standardized endpoints for bleeding (BARC type 3 or 5) and ischemic events, and evaluate these two outcomes separately, whenever possible [8].

Significant work has been done with regards to the efficacy and safety of different P2Y12 inhibitors in the elderly population. Age 75 or greater is the most common minor HBR criteria, and therefore, recommendations for DAPT in HBR populations could apply to the elderly [8]. A TWILIGHT sub-analysis identified that in patients aged 65 or older ticagrelor monotherapy after 3 months of DAPT significantly reduced bleeding events, while preserving ischemic benefit [91]. Similarly, a sub-analysis of TICO showed that patients aged 64 or greater ticagrelor monotherapy after 3 months of DAPT decreased the rates of the primary outcome (composite of major bleeding, death, MI, stent thrombosis, stroke, or target-vessel revascularization) [92].

However, it is not understood which P2Y12 inhibitor is best for the elderly. Some studies have shown decreased risk of adverse events with ticagrelor when compared to clopidogrel without a concomitant increase in bleeding events [93, 94]. However, studies have also shown an increased bleeding with ticagrelor or prasugrel when compared to clopidogrel [95, 96]. Given these conflicting reports, additional randomized control trials among the elderly are required.

Ongoing studies are investigating short DAPT regimens with new stent technologies. TARGET SAFE will assess 1 month versus 6 months of DAPT among HBR patients receiving the Firehawk sirolimus eluting stent (NCT03287167) and BIOFLOW-DAPT will assess shorter DAPT among patients receiving either the new Orsiro stent compared to the Resolute Onyx stent (NCT04137510).

5. Conclusions

Remarkable advances in PCI technologies and techniques over the last decade have enabled more high-risk patients than ever before to undergo PCI. In particular, HBR patients constitute a third of those undergoing PCI and their management remains challenging periprocedurally and in the long-term. Due to an overlap between ischemic and bleeding risk factors in HBR patients, meticulous choice of antithrombotic therapy intensity and duration is imperative. Several bleeding avoidance strategies have recently been developed and tested in clinical trials, though only few enrolled HBR patients. Further large and well-powered studies dedicated to HBR patients are needed to establish the optimal management strategy in these vulnerable patients.

Author Contributions

Conceptualization—DJ, JN, and FB; Methodology—DJ, JN, and FB; Writing—original draft preparation—DJ, JN and FB; Writing—review and editing—DJ, JN, FB, MS, DF, DC, AS, and RM; Visualization—DJ, JN and FB; Supervision—RM. All authors have read and agreed to the published version of the manuscript.

Ethics Approval and Consent to Participate

Not applicable.

Acknowledgment

Not applicable.

Funding

This research received no external funding.

Conflict of Interest

The authors declare no conflict of interest.

References
[1]
Angiolillo DJ, Ueno M, Goto S. Basic Principles of Platelet Biology and Clinical Implications. Circulation Journal. 2010; 74: 597–607.
[2]
Valgimigli M, Bueno H, Byrne RA, Collet JP, Costa F, Jeppsson A, et al. 2017 ESC focused update on dual antiplatelet therapy in coronary artery disease developed in collaboration with EACTS. European Journal of Cardio-Thoracic Surgery. 2018; 53: 34–78.
[3]
Levine GN, Bates ER, Bittl JA, Brindis RG, Fihn SD, Fleisher LA, et al. 2016 ACC/AHA Guideline Focused Update on Duration of Dual Antiplatelet Therapy in Patients With Coronary Artery Disease: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines: 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.
[4]
Mauri L, Kereiakes DJ, Yeh RW, Driscoll-Shempp P, Cutlip DE, Steg PG, et al. Twelve or 30 Months of Dual Antiplatelet Therapy after Drug-Eluting Stents. New England Journal of Medicine. 2014; 371: 2155–2166.
[5]
Généreux P, Giustino G, Witzenbichler B, Weisz G, Stuckey TD, Rinaldi MJ, et al. Incidence, Predictors, and Impact of Post-Discharge Bleeding After Percutaneous Coronary Intervention. Journal of the American College of Cardiology. 2015; 66: 1036–1045.
[6]
Valgimigli M, Costa F, Lokhnygina Y, Clare RM, Wallentin L, Moliterno DJ, et al. Trade-off of myocardial infarction vs. bleeding types on mortality after acute coronary syndrome: lessons from the Thrombin Receptor Antagonist for Clinical Event Reduction in Acute Coronary Syndrome (TRACER) randomized trial. European Heart Journal. 2017; 38: 804–810.
[7]
Baber U, Dangas G, Chandrasekhar J, Sartori S, Steg PG, Cohen DJ, et al. Time-Dependent Associations between Actionable Bleeding, Coronary Thrombotic Events, and Mortality Following Percutaneous Coronary Intervention. JACC: Cardiovascular Interventions. 2016; 9: 1349–1357.
[8]
Capodanno D, Morice MC, Angiolillo DJ, Bhatt DL, Byrne RA, Colleran R, et al. Trial Design Principles for Patients at High Bleeding Risk Undergoing PCI: JACC Scientific Expert Panel. Journal of the American College of Cardiology. 2020; 76: 1468–1483.
[9]
Natsuaki M, Morimoto T, Shiomi H, Yamaji K, Watanabe H, Shizuta S, et al. Application of the Academic Research Consortium High Bleeding Risk Criteria in an all-Comers Registry of Percutaneous Coronary Intervention. Circulation: Cardiovascular Interventions. 2019; 12: e008307.
[10]
Kazi DS, Leong TK, Chang TI, Solomon MD, Hlatky MA, Go AS. Association of Spontaneous Bleeding and Myocardial Infarction with Long-Term Mortality after Percutaneous Coronary Intervention. Journal of the American College of Cardiology. 2015; 65: 1411–1420.
[11]
Urban P, Mehran R, Colleran R, Angiolillo DJ, Byrne RA, Capodanno D, et al. Defining high bleeding risk in patients undergoing percutaneous coronary intervention: a consensus document from the Academic Research Consortium for High Bleeding Risk. European Heart Journal. 2019; 40: 2632–2653.
[12]
Urban P, Gregson J, Owen R, Mehran R, Windecker S, Valgimigli M, et al. Assessing the Risks of Bleeding vs Thrombotic Events in Patients at High Bleeding Risk after Coronary Stent Implantation: The ARC–High Bleeding Risk Trade-off Model. JAMA Cardiology. 2021; 6: 410.
[13]
Ueki Y, Bär S, Losdat S, Otsuka T, Zanchin C, Zanchin T, et al. Validation of the Academic Research Consortium for High Bleeding Risk (ARC-HBR) criteria in patients undergoing percutaneous coronary intervention and comparison with contemporary bleeding risk scores. EuroIntervention. 2020; 16: 371–379.
[14]
Cao D, Mehran R, Dangas G, Baber U, Sartori S, Chandiramani R, et al. Validation of the Academic Research Consortium High Bleeding Risk Definition in Contemporary PCI Patients. Journal of the American College of Cardiology. 2020; 75: 2711–2722.
[15]
Nicolas J, Beerkens F, Cao D, Sartori S, Pivato CA, Qiu H, et al. Performance of the academic research consortium high-bleeding risk criteria in patients undergoing PCI for acute myocardial infarction. Journal of Thrombosis and Thrombolysis. 2022; 53: 20–29.
[16]
Collet JP, Thiele H, Barbato E, Barthélémy O, Bauersachs J, Bhatt DL, et al. 2020 ESC Guidelines for the management of acute coronary syndromes in patients presenting without persistent ST-segment elevation. European Heart Journal. 2021; 42: 1289–1367.
[17]
Valgimigli M, Bueno H, Byrne RA, Collet JP, Costa F, Jeppsson A, et al. 2017 ESC focused update on dual antiplatelet therapy in coronary artery disease developed in collaboration with EACTS: The Task Force for dual antiplatelet therapy in coronary artery disease of the European Society of Cardiology (ESC) and of the European Association for Cardio-Thoracic Surgery (EACTS). European Heart Journal. 2018; 39: 213–260.
[18]
Yeh RW, Secemsky EA, Kereiakes DJ, Normand ST, Gershlick AH, Cohen DJ, et al. Development and Validation of a Prediction Rule for Benefit and Harm of Dual Antiplatelet Therapy beyond 1 Year after Percutaneous Coronary Intervention. The Journal of the American Medical Association. 2016; 315: 1735.
[19]
Costa F, van Klaveren D, James S, Heg D, Räber L, Feres F, et al. Derivation and validation of the predicting bleeding complications in patients undergoing stent implantation and subsequent dual antiplatelet therapy (PRECISE-DAPT) score: a pooled analysis of individual-patient datasets from clinical trials. The Lancet. 2017; 389: 1025–1034.
[20]
Baber U, Mehran R, Giustino G, Cohen DJ, Henry TD, Sartori S, et al. Coronary Thrombosis and Major Bleeding after PCI with Drug-Eluting Stents. Journal of the American College of Cardiology. 2016; 67: 2224–2234.
[21]
Morice M, Talwar S, Gaemperli O, Richardt G, Eberli F, Meredith I, et al. Drug-coated versus bare-metal stents for elderly patients: a predefined sub-study of the LEADERS FREE trial. International Journal of Cardiology. 2017; 243: 110–115.
[22]
Windecker S, Latib A, Kedhi E, Kirtane AJ, Kandzari DE, Mehran R, et al. Polymer-based or Polymer-free Stents in Patients at High Bleeding Risk. New England Journal of Medicine. 2020; 382: 1208–1218.
[23]
Varenne O, Cook S, Sideris G, Kedev S, Cuisset T, Carrié D, et al. Drug-eluting stents in elderly patients with coronary artery disease (SENIOR): a randomised single-blind trial. The Lancet. 2018; 391: 41–50.
[24]
Rissanen TT, Uskela S, Eränen J, Mäntylä P, Olli A, Romppanen H, et al. Drug-coated balloon for treatment of de-novo coronary artery lesions in patients with high bleeding risk (DEBUT): a single-blind, randomised, non-inferiority trial. The Lancet. 2019; 394: 230–239.
[25]
Kirtane AJ, Stoler R, Feldman R, Neumann F, Boutis L, Tahirkheli N, et al. Primary Results of the EVOLVE Short DAPT Study. Circulation: Cardiovascular Interventions. 2021; 14: e010144.
[26]
Valgimigli M, Cao D, Makkar RR, Bangalore S, Bhatt DL, Angiolillo DJ, et al. Design and rationale of the XIENCE short DAPT clinical program: an assessment of the safety of 3-month and 1-month DAPT in patients at high bleeding risk undergoing PCI with an everolimus-eluting stent. American Heart Journal. 2021; 231: 147–156.
[27]
Mehran R, Valgimigli M. The XIENCE short DAPT program: XIENCE 90/28 evaluating the safety of 3- and 1-month DAPT in HBR patients. 2020. Available at: https://www.acc.org/-/media/Clinical/PDF-Files/Approved-PDFs/2020/10/12/TCT20/Oct15-Thur/1245pmET-XIENCE-tct-2020.pdf (Accessed: 10 October 2021).
[28]
Valgimigli M, Cao D, Angiolillo DJ, Bangalore S, Bhatt DL, Ge J, et al. Duration of Dual Antiplatelet Therapy for Patients at High Bleeding Risk Undergoing PCI. Journal of the American College of Cardiology. 2021; 78: 2060–2072.
[29]
Valgimigli M, Frigoli E, Heg D, Tijssen J, Jüni P, Vranckx P, et al. Dual Antiplatelet Therapy after PCI in Patients at High Bleeding Risk. New England Journal of Medicine. 2021; 385: 1643–1655.
[30]
Cannon CP, Harrington RA, James S, Ardissino D, Becker RC, Emanuelsson H, et al. Comparison of ticagrelor with clopidogrel in patients with a planned invasive strategy for acute coronary syndromes (PLATO): a randomised double-blind study. The Lancet. 2010; 375: 283–293.
[31]
Mahaffey KW, Wojdyla DM, Carroll K, Becker RC, Storey RF, Angiolillo DJ, et al. Ticagrelor Compared with Clopidogrel by Geographic Region in the Platelet Inhibition and Patient Outcomes (PLATO) Trial. Circulation. 2011; 124: 544–554.
[32]
Kirkby NS, Leadbeater PDM, Chan MV, Nylander S, Mitchell JA, Warner TD. Antiplatelet effects of aspirin vary with level of P2Y12 receptor blockade supplied by either ticagrelor or prasugrel. Journal of Thrombosis and Haemostasis. 2011; 9: 2103–2105.
[33]
Armstrong PCJ, Leadbeater PD, Chan MV, Kirkby NS, Jakubowski JA, Mitchell JA, et al. In the presence of strong P2Y12 receptor blockade, aspirin provides little additional inhibition of platelet aggregation. Journal of Thrombosis and Haemostasis. 2011; 9: 552–561.
[34]
Vranckx P, Valgimigli M, Jüni P, Hamm C, Steg PG, Heg D, et al. Ticagrelor plus aspirin for 1 month, followed by ticagrelor monotherapy for 23 months vs aspirin plus clopidogrel or ticagrelor for 12 months, followed by aspirin monotherapy for 12 months after implantation of a drug-eluting stent: a multicentre, open-label, randomised superiority trial. Lancet. 2018; 392: 940–949.
[35]
Mehran R, Cao D, Baber U. Ticagrelor Monotherapy after Coronary Stenting. Journal of the American College of Cardiology. 2019; 74: 2235–2237.
[36]
Franzone A, McFadden E, Leonardi S, Piccolo R, Vranckx P, Serruys PW, et al. Ticagrelor alone Versus Dual Antiplatelet Therapy from 1 Month after Drug-Eluting Coronary Stenting. Journal of the American College of Cardiology. 2019; 74: 2223–2234.
[37]
Hahn JY, Song YB, Oh JH, Chun WJ, Park YH, Jang WJ, et al. Effect of P2Y12 Inhibitor Monotherapy vs Dual Antiplatelet Therapy on Cardiovascular Events in Patients Undergoing Percutaneous Coronary Intervention: The SMART-CHOICE Randomized Clinical Trial. The Journal of the American Medical Association. 2019; 321: 2428–2437.
[38]
Watanabe H, Domei T, Morimoto T, Natsuaki M, Shiomi H, Toyota T, et al. Effect of 1-Month Dual Antiplatelet Therapy Followed by Clopidogrel vs 12-Month Dual Antiplatelet Therapy on Cardiovascular and Bleeding Events in Patients Receiving PCI: The STOPDAPT-2 Randomized Clinical Trial. The Journal of the American Medical Association. 2019; 321: 2414–2427.
[39]
Watanabe H. STOPDAPT-2 ACS: One-Month Dual Antiplatelet Therapy Followed by Clopidogrel Monotherapy in Acute Coronary Syn-Drome. Proceedings of the ESC Congress. 2021.
[40]
Kim B, Hong S, Cho Y, Yun KH, Kim YH, Suh Y, et al. Effect of Ticagrelor Monotherapy vs Ticagrelor with Aspirin on Major Bleeding and Cardiovascular Events in Patients with Acute Coronary Syndrome: The TICO Randomized Clinical Trial. The Journal of the American Medical Association. 2020; 323: 2407.
[41]
Mehran R, Baber U, Sharma SK, Cohen DJ, Angiolillo DJ, Briguori C, et al. Ticagrelor with or without Aspirin in High-Risk Patients after PCI. New England Journal of Medicine. 2019; 381: 2032–2042.
[42]
Escaned J, Cao D, Baber U, Nicolas J, Sartori S, Zhang Z, et al. Ticagrelor monotherapy in patients at high bleeding risk undergoing percutaneous coronary intervention: TWILIGHT-HBR. European Heart Journal. 2021; 42: 4624–4634.
[43]
Giacoppo D, Matsuda Y, Fovino LN, D’Amico G, Gargiulo G, Byrne RA, et al. Short dual antiplatelet therapy followed by P2Y12 inhibitor monotherapy vs. prolonged dual antiplatelet therapy after percutaneous coronary intervention with second-generation drug-eluting stents: a systematic review and meta-analysis of randomized clinical trials. European Heart Journal. 2021; 42: 308–319.
[44]
Bianco M, Careggio A, Destefanis P, Luciano A, Perrelli MG, Quadri G, et al. P2Y12 inhibitors monotherapy after short course of dual antiplatelet therapy in patients undergoing percutaneous coronary intervention: a meta-analysis of randomized clinical trials including 29 089 patients. European Heart Journal - Cardiovascular Pharmacotherapy. 2021; 7: 196–205.
[45]
O’Donoghue ML, Murphy SA, Sabatine MS. The Safety and Efficacy of Aspirin Discontinuation on a Background of a P2Y12 Inhibitor in Patients After Percutaneous Coronary Intervention. Circulation. 2020; 142: 538–545.
[46]
Gwon H, Hahn J, Park KW, Song YB, Chae I, Lim D, et al. Six-Month Versus 12-Month Dual Antiplatelet Therapy after Implantation of Drug-Eluting Stents. Circulation. 2012; 125: 505–513.
[47]
Hahn J-Y, Song YB, Oh J-H, Cho D-K, Lee JB, Doh J-H, et al. 6-month versus 12-month or longer dual antiplatelet therapy after percutaneous coronary intervention in patients with acute coronary syndrome (SMART-DATE): a randomised, open-label, non-inferiority trial. The Lancet. 2018; 391: 1274–1284.
[48]
Colombo A, Chieffo A, Frasheri A, Garbo R, Masotti-Centol M, Salvatella N, et al. Second-Generation Drug-Eluting Stent Implantation Followed by 6- Versus 12-Month Dual Antiplatelet Therapy. Journal of the American College of Cardiology. 2014; 64: 2086–2097.
[49]
Collet J, Silvain J, Barthélémy O, Rangé G, Cayla G, Van Belle E, et al. Dual-antiplatelet treatment beyond 1 year after drug-eluting stent implantation (ARCTIC-Interruption): a randomised trial. The Lancet. 2014; 384: 1577–1585.
[50]
Schulz-Schüpke S, Byrne RA, Ten Berg JM, Neumann FJ, Han Y, Adriaenssens T, et al. ISAR-SAFE: a randomized, double-blind, placebo-controlled trial of 6 vs. 12 months of clopidogrel therapy after drug-eluting stenting. European Heart Journal. 2015; 36: 1252–1263.
[51]
Kedhi E, Fabris E, van der Ent M, Buszman P, von Birgelen C, Roolvink V, et al. Six months versus 12 months dual antiplatelet therapy after drug-eluting stent implantation in ST-elevation myocardial infarction (DAPT-STEMI): randomised, multicentre, non-inferiority trial. BMJ. 2018; 363: k3793.
[52]
De Luca G, Damen SA, Camaro C, Benit E, Verdoia M, Rasoul S, et al. Final results of the randomised evaluation of short-term dual antiplatelet therapy in patients with acute coronary syndrome treated with a new-generation stent (REDUCE trial). EuroIntervention. 2019; 15: e990–e998.
[53]
From the American Association of Neurological Surgeons (AANS), American Society of Neuroradiology (ASNR), Cardiovascular and Interventional Radiology Society of Europe (CIRSE), Canadian Interventional Radiology Association (CIRA), Congress of Neurological Surgeons (CNS), et al. Reduced duration of dual antiplatelet therapy using an improved drug-eluting stent for percutaneous coronary intervention of the left main artery in a real-world, all-comer population: Rationale and study design of the prospective randomized multicenter I. American Heart Journal. 2017; 187: 104–111.
[54]
Hong S, Kim J, Hong SJ, Lim D, Lee S, Yun KH, et al. 1-Month Dual-Antiplatelet Therapy Followed by Aspirin Monotherapy after Polymer-Free Drug-Coated Stent Implantation. JACC: Cardiovascular Interventions. 2021; 14: 1801–1811.
[55]
Kim BK, Hong MK, Shin DH, Nam CM, Kim JS, Ko YG, et al. A New Strategy for Discontinuation of Dual Antiplatelet Therapy. Journal of the American College of Cardiology. 2012; 60: 1340–1348.
[56]
Feres F, Costa RA, Abizaid A, Leon MB, Marin-Neto JA, Botelho RV, et al. Three vs Twelve Months of Dual Antiplatelet Therapy After Zotarolimus-Eluting Stents: The OPTIMIZE Randomized Trial. The Journal of the American Medical Association. 2013; 310: 2510–2522.
[57]
Han Y, Xu B, Jing Q, Lu S, Yang L, Xu K, et al. A Randomized Comparison of Novel Biodegradable Polymer- and Durable Polymer–Coated Cobalt-Chromium Sirolimus-Eluting Stents. JACC: Cardiovascular Interventions. 2014; 7: 1352–1360.
[58]
Hong S, Kim B, Shin D, Nam C, Kim J, Ko Y, et al. Effect of Intravascular Ultrasound–Guided vs Angiography-Guided Everolimus-Eluting Stent Implantation. The Journal of the American Medical Association. 2015; 314: 2155.
[59]
Nakamura M, Iijima R, Ako J, Shinke T, Okada H, Ito Y, et al. Dual Antiplatelet Therapy for 6 Versus 18 Months After Biodegradable Polymer Drug-Eluting Stent Implantation. JACC: Cardiovascular Interventions. 2017; 10: 1189–1198.
[60]
Gilard M, Barragan P, Noryani AAL, Noor HA, Majwal T, Hovasse T, et al. 6- Versus 24-Month Dual Antiplatelet Therapy After Implantation of Drug-Eluting Stents in Patients Nonresistant to Aspirin. Journal of the American College of Cardiology. 2015; 65: 777–786.
[61]
Lee B, Kim J, Lee O, Min P, Yoon Y, Hong B, et al. Safety of six-month dual antiplatelet therapy after second-generation drug-eluting stent implantation: OPTIMA-C Randomised Clinical Trial and OCT Substudy. EuroIntervention. 2018; 13: 1923–1930.
[62]
Roffi M, Patrono C, Collet JP, Mueller C, Valgimigli M, Andreotti F, et al. 2015 ESC Guidelines for the management of acute coronary syndromes in patients presenting without persistent ST-segment elevation: Task Force for the Management of Acute Coronary Syndromes in Patients Presenting without Persistent ST-Segment Elevation of the European Society of Cardiology (ESC). European Heart Journal. 2016; 37: 267–315.
[63]
Antman EM, Wiviott SD, Murphy SA, Voitk J, Hasin Y, Widimsky P, et al. Early and late benefits of prasugrel in patients with acute coronary syndromes undergoing percutaneous coronary intervention: a TRITON-TIMI 38 (TRial to Assess Improvement in Therapeutic Outcomes by Optimizing Platelet InhibitioN with Prasugrel-Thrombolysis In Myocardial Infarction) analysis. Journal of the American College of Cardiology. 2008; 51: 2028–2033.
[64]
Cuisset T, Deharo P, Quilici J, Johnson TW, Deffarges S, Bassez C, et al. Benefit of switching dual antiplatelet therapy after acute coronary syndrome: the TOPIC (timing of platelet inhibition after acute coronary syndrome) randomized study. European Heart Journal. 2017; 38: 3070–3078.
[65]
Kim H, Kang J, Hwang D, Han J, Yang H, Kang H, et al. Prasugrel-based de-escalation of dual antiplatelet therapy after percutaneous coronary intervention in patients with acute coronary syndrome (HOST-REDUCE-POLYTECH-ACS): an open-label, multicentre, non-inferiority randomised trial. The Lancet. 2020; 396: 1079–1089.
[66]
Sibbing D, Aradi D, Jacobshagen C, Gross L, Trenk D, Geisler T, et al. Guided de-escalation of antiplatelet treatment in patients with acute coronary syndrome undergoing percutaneous coronary intervention (TROPICAL-ACS): a randomised, open-label, multicentre trial. Lancet. 2017; 390: 1747–1757.
[67]
Claassens DMF, Vos GJA, Bergmeijer TO, Hermanides RS, van ‘t Hof AWJ, van der Harst P, et al. A Genotype-Guided Strategy for Oral P2Y(12) Inhibitors in Primary PCI. The New England Journal of Medicine. 2019; 381: 1621–1631.
[68]
Levine GN, Jeong Y, Goto S, Anderson JL, Huo Y, Mega JL, et al. Expert consensus document: World Heart Federation expert consensus statement on antiplatelet therapy in East Asian patients with ACS or undergoing PCI. Nature Reviews Cardiology. 2014; 11: 597–606.
[69]
Wiviott SD, Braunwald E, McCabe CH, Montalescot G, Ruzyllo W, Gottlieb S, et al. Prasugrel versus clopidogrel in patients with acute coronary syndromes. The New England Journal of Medicine. 2007; 357: 2001–2015.
[70]
Tantry US, Bonello L, Aradi D, Price MJ, Jeong Y, Angiolillo DJ, et al. Consensus and Update on the Definition of on-Treatment Platelet Reactivity to Adenosine Diphosphate Associated with Ischemia and Bleeding. Journal of the American College of Cardiology. 2013; 62: 2261–2273.
[71]
Aradi D, Storey RF, Komócsi A, Trenk D, Gulba D, Kiss RG, et al. Expert position paper on the role of platelet function testing in patients undergoing percutaneous coronary intervention. European Heart Journal. 2014; 35: 209–215.
[72]
Breet NJ. Comparison of Platelet Function Tests in Predicting Clinical Outcome in Patients Undergoing Coronary Stent Implantation. The Journal of the American Medical Association. 2010; 303: 754.
[73]
Mega JL, Simon T, Collet J, Anderson JL, Antman EM, Bliden K, et al. Reduced-Function CYP2C19 Genotype and Risk of Adverse Clinical Outcomes among Patients Treated with Clopidogrel Predominantly for PCI: A Meta-analysis. The Journal of the American Medical Association. 2010; 304: 1821.
[74]
Harmsze AM, van Werkum JW, ten Berg JM, Zwart B, Bouman HJ, Breet NJ, et al. CYP2C19*2 and CYP2C9*3 alleles are associated with stent thrombosis: a case-control study. European Heart Journal. 2010; 31: 3046–3053.
[75]
Cavallari LH, Lee CR, Beitelshees AL, Cooper-DeHoff RM, Duarte JD, Voora D, et al. Multisite Investigation of Outcomes With Implementation of CYP2C19 Genotype-Guided Antiplatelet Therapy After Percutaneous Coronary Intervention. JACC: Cardiovascular Interventions. 2018; 11: 181–191.
[76]
Wallentin L, James S, Storey RF, Armstrong M, Barratt BJ, Horrow J, et al. Effect of CYP2C19 and ABCB1 single nucleotide polymorphisms on outcomes of treatment with ticagrelor versus clopidogrel for acute coronary syndromes: a genetic substudy of the PLATO trial. the Lancet. 2010; 376: 1320–1328.
[77]
Galli M, Benenati S, Capodanno D, Franchi F, Rollini F, D’Amario D, et al. Guided versus standard antiplatelet therapy in patients undergoing percutaneous coronary intervention: a systematic review and meta-analysis. The Lancet. 2021; 397: 1470–1483.
[78]
Shoji S, Kuno T, Fujisaki T, Takagi H, Briasoulis A, Deharo P, et al. De-Escalation of Dual Antiplatelet Therapy in Patients with Acute Coronary Syndromes. Journal of the American College of Cardiology. 2021; 78: 763–777.
[79]
Tavenier AH, Mehran R, Chiarito M, Cao D, Pivato CA, Nicolas J, et al. Guided and unguided de-escalation from potent P2Y12 inhibitors among patients with ACS: a meta-analysis. Eur Heart J Cardiovasc Pharmacother. 2021. (in press)
[80]
Pereira NL, Farkouh ME, So D, Lennon R, Geller N, Mathew V, et al. Effect of Genotype-Guided Oral P2Y12 Inhibitor Selection vs Conventional Clopidogrel Therapy on Ischemic Outcomes after Percutaneous Coronary Intervention: The TAILOR-PCI Randomized Clinical Trial. The Journal of the American Medical Association. 2020; 324: 761.
[81]
Trenk D, Stone GW, Gawaz M, Kastrati A, Angiolillo DJ, Müller U, et al. A Randomized Trial of Prasugrel Versus Clopidogrel in Patients with High Platelet Reactivity on Clopidogrel after Elective Percutaneous Coronary Intervention with Implantation of Drug-Eluting Stents. Journal of the American College of Cardiology. 2012; 59: 2159–2164.
[82]
Sacks D, Baxter B, Campbell BCV, Carpenter JS, Cognard C, Dippel D, et al. Multisociety Consensus Quality Improvement Revised Consensus Statement for Endovascular Therapy of Acute Ischemic Stroke. International Journal of Stroke. 2018; 13: 612–632.
[83]
Braunwald E. Treatment of Left Main Coronary Artery Disease. New England Journal of Medicine. 2016; 375: 2284–2285.
[84]
Neumann FJ, Sousa-Uva M, Ahlsson A, Alfonso F, Banning AP, Benedetto U, et al. 2018 ESC/EACTS Guidelines on myocardial revascularization. European Heart Journal. 2019; 40: 87–165.
[85]
Choi J, Kim I, Cho S, Kim J, Hong S, Shin D, et al. Optimal Duration for Dual Antiplatelet Therapy after Left Main Coronary Artery Stenting. Circulation Journal. 2020; 85: 59–68
[86]
Chiarito M, Kini A, Roumeliotis A, Cao D, Power D, Sartori S, et al. Prevalence and Impact of High Bleeding Risk in Patients Undergoing Left Main Artery Disease PCI. JACC: Cardiovascular Interventions. 2021; 14: 2447–2457.
[87]
Dangas G, Baber U, Sharma S, Giustino G, Mehta S, Cohen DJ, et al. Ticagrelor with or without Aspirin after Complex PCI. Journal of the American College of Cardiology. 2020; 75: 2414–2424.
[88]
Rigatelli G, Zuin M, Vassilev D, De Ferrari GM, D’Ascenzo F. Outcomes of Left Main Bifurcation Stenting Depends on both Length of Dual Antiplatelet Therapy and Stenting Strategy. Cardiovascular Revascularization Medicine. 2020; 21: 1319–1322.
[89]
Sachdeva A, Hung Y, Solomon MD, McNulty EJ. Duration of Dual Antiplatelet Therapy after Percutaneous Coronary Intervention for Chronic Total Occlusion. The American Journal of Cardiology. 2020; 132: 44–51.
[90]
Lee SH, Yang JH, Choi SH, Park TK, Jang WJ, Song YB, et al. Duration of dual antiplatelet therapy in patients treated with percutaneous coronary intervention for coronary chronic total occlusion. PLoS ONE. 2017; 12: e0176737.
[91]
Angiolillo DJ, Cao D, Baber U, Sartori S, Zhang Z, Dangas G, et al. Impact of Age on the Safety and Efficacy of Ticagrelor Monotherapy in Patients Undergoing PCI. JACC: Cardiovascular Interventions. 2021; 14: 1434–1446.
[92]
Kim BG, Hong S, Kim B, Lee S, Ahn C, Shin D, et al. Age‐Dependent Effect of Ticagrelor Monotherapy Versus Ticagrelor with Aspirin on Major Bleeding and Cardiovascular Events: a Post Hoc Analysis of the TICO Randomized Trial. Journal of the American Heart Association. 2021; 10: e022700.
[93]
Bianco M, Careggio A, Biolè CA, Quadri G, Quiros A, Raposeiras-Roubin S, et al. Ticagrelor or Clopidogrel After an Acute Coronary Syndrome in the Elderly: A Propensity Score Matching Analysis from 16,653 Patients Treated with PCI Included in Two Large Multinational Registries. Cardiovascular Drugs and Therapy. 2021; 35: 1171–1182.
[94]
Schmucker J, Fach A, Mata Marin LA, Retzlaff T, Osteresch R, Kollhorst B, et al. Efficacy and Safety of Ticagrelor in Comparison to Clopidogrel in Elderly Patients with ST‐Segment–Elevation Myocardial Infarctions. Journal of the American Heart Association. 2019; 8: e012530.
[95]
Szummer K, Montez-Rath ME, Alfredsson J, Erlinge D, Lindahl B, Hofmann R, et al. Comparison between Ticagrelor and Clopidogrel in Elderly Patients with an Acute Coronary Syndrome. Circulation. 2020; 142: 1700–1708.
[96]
Gimbel M, Qaderdan K, Willemsen L, Hermanides R, Bergmeijer T, de Vrey E, et al. Clopidogrel versus ticagrelor or prasugrel in patients aged 70 years or older with non-ST-elevation acute coronary syndrome (POPular AGE): the randomised, open-label, non-inferiority trial. The Lancet. 2020; 395: 1374–1381.
Share
Back to top