You are on page 1of 14

European Heart Journal (2023) 44, 3059–3072 STATE OF THE ART REVIEW

https://doi.org/10.1093/eurheartj/ehad362 Thrombosis and antithrombotic treatment

Personalised antiplatelet therapies for


coronary artery disease: what the future holds
1 2
Davide Capodanno and Dominick J. Angiolillo *
1
Division of Cardiology, Azienda Ospedaliero-Universitaria ‘G. Rodolico – San Marco’, University of Catania, Via Santa Sofia, 78 - 95123 Catania, Italy; and 2Division of Cardiology, University

Downloaded from https://academic.oup.com/eurheartj/article/44/32/3059/7204812 by guest on 25 August 2023


of Florida College of Medicine-Jacksonville, 655 West 8th Street, Jacksonville, FL 32209, USA

Received 5 March 2023; revised 7 May 2023; accepted 22 May 2023; online publish-ahead-of-print 22 June 2023

Graphical Abstract

Personalized approach to antiplatelet therapy in coronary artery disease

High ischaemic risk High ischaemic risk

Low bleeding risk High bleeding risk

Addition of a P2Y12 Discontinuation of


inhibitor aspirin
Switch to a more potent Selection of the P2Y12
P2Y12 inhibitor inhibitor based on PFT
or genotype

Ischaemic risk
Low ischaemic risk Low ischaemic risk

Low bleeding risk High bleeding risk

Standard Switch to a less potent


antiplatelet therapy P2Y12 inhibitor

Discontinuation of the
P2Y12 inhibitor
or aspirin

Bleeding risk

Proposed strategies for tailoring antithrombotic therapy according to individual ischaemic and bleeding risk. Patients at high
risk of ischemic events and low risk of bleeding may benefit from an intensified antiplatelet therapy regimen (e.g., escalation), while those at high risk
of bleeding and low risk of ischemic events require a less intensive and more cautious approach (e.g, de-escalation). In patients at low risk of ischemic
events and low risk of bleeding the benefits of antiplatelet therapy may not outweigh the risks, while patients who have high risks of both ischemic
and bleeding events require a delicate balance, as the approach to managing their conditions must weigh the potential benefits against the potential
risks. Abbreviations: PFT, platelet function testing. Adapted from Cao et al.1

* Corresponding author. Tel: +1 904 244 3933, Fax: +1 904 244 3102, Email: dominick.angiolillo@jax.ufl.edu
© The Author(s) 2023. Published by Oxford University Press on behalf of the European Society of Cardiology. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com
3060 D. Capodanno and D. J. Angiolillo

Abstract

Coronary artery disease (CAD) is one of the leading causes of death globally, and antiplatelet therapy is crucial for both its prevention and treatment.
Antiplatelet drugs such as aspirin and P2Y12 inhibitors are commonly used to reduce the risk of thrombotic events, including myocardial infarction,
stroke, and stent thrombosis. However, the benefits associated with the use of antiplatelet drugs also come with a risk of bleeding complications. The
ever-growing understanding of the poor prognostic implications associated with bleeding has set the foundations for defining strategies that can
mitigate such safety concern without any trade-off in antithrombotic protection. To this extent, personalised antiplatelet therapy has emerged
as a paradigm that optimizes the balance between safety and efficacy by customizing treatment to the individual patient’s needs and risk profile.
Accurate risk stratification for both bleeding and thrombosis can aid in selecting the optimal antiplatelet therapy and prevent serious and life-threa­
tening outcomes. Risk stratification has traditionally included clinical and demographic characteristics and has expanded to incorporate angiographic
features and laboratory findings. The availability of bedside platelet function testing as well as rapid genotyping assays has also allowed for a more

Downloaded from https://academic.oup.com/eurheartj/article/44/32/3059/7204812 by guest on 25 August 2023


individualized selection of antiplatelet therapy. This review provides a comprehensive overview of the current state of the art and future trends in
personalised antiplatelet therapy for patients with CAD, with emphasis on those presenting with an acute coronary syndrome and undergoing per­
cutaneous coronary revascularization. The aim is to provide clinicians with a comprehensive understanding of personalised antiplatelet therapy and
facilitate informed clinical decision-making.
.............................................................................................................................................................................................
Keywords Antiplatelet therapy • Coronary artery disease • Risk assessment • Personalised antiplatelet therapy • Platelet function
testing • Genotyping

Introduction Antiplatelet therapies in


Coronary artery disease (CAD) is a leading global cause of death and cardiovascular disease
antiplatelet therapy plays a critical role in its prevention and treatment.2
Mechanisms of action of oral and intravenous antiplatelet drugs cur­
Aspirin and P2Y12 receptor inhibitors are commonly used antiplatelet
rently approved for clinical use are visualized in Figure 1.17
drugs that lower the risk of thrombotic events across the spectrum
In accordance with type I classes of recommendations from practice
of CAD manifestations.3,4 However, the benefits associated with the
guidelines,3,4 aspirin is generally indicated for the secondary prevention
use of antiplatelet drugs also come with a risk of bleeding complica­
of CAD, including patients with previous myocardial infarction (MI) or
tions.5 The ever-growing understanding of the poor prognostic implica­
revascularization, and the P2Y12 inhibitor clopidogrel is an alternative to
tions associated with bleeding, including increased mortality, has set the
aspirin for patients with allergy or intolerance. Additionally, clopidogrel
foundations for defining strategies that can mitigate such safety concern
is prescribed after percutaneous coronary intervention (PCI) in pa­
without any trade-off in antithrombotic protection.6,7 To this extent,
tients who present with a chronic coronary syndrome (CCS).
personalised antiplatelet treatment regimens represent a developing
Prasugrel is recommended in patients with an ACS undergoing PCI
concept that seeks to achieve a net benefit between safety and efficacy
and ticagrelor in patients with ACS with or without revascularization.
by tailoring treatment to each patient’s unique needs and risk profile.8,9
A description on the use of oral anticoagulants in patients with CAD
The identification of patients’ risk for both bleeding and thrombosis can
and antiplatelet therapy in patients on oral anticoagulants goes beyond
aid in the selection of the optimal antiplatelet therapy and reduce the
the scope of this manuscript and is described elsewhere.18–20
incidence of serious adverse events.
Multiple clinical, demographic, angiographic, and laboratory factors
have been identified as predictors of bleeding and thrombosis.10–15
By considering these factors, clinicians are better equipped to risk strat­
Risk assessment
ify patients and in turn select the antiplatelet treatment regimen with The 2021 guidelines from the European Society of Cardiology (ESC) for
the best risk-benefit profile for a given individual. Platelet function test­ cardiovascular disease prevention in clinical practice emphasize the im­
ing (PFT) and rapid genotyping are emerging as advancements that en­ portance of timely recognition of risk factors, predictors, and modifiers
able the assessment of a patient’s individual response to antiplatelet that impact the likelihood of CAD complications.3 This paradigm can be
therapy, allowing for an even more individualized selection of antiplate­ easily extended to the risks confronted by patients on antiplatelet ther­
let therapy.16 This review provides an overview of current state of the apy. In fact, the key to thrombosis and bleeding prevention is to identify
art and future trends in the role of risk assessment and personalised ap­ those patients who will receive the greatest benefit from antiplatelet
proach in antiplatelet therapies for patients with CAD, with emphasis drugs at the least possible safety price.
on those presenting with an acute coronary syndrome (ACS) and In general, the higher the absolute risk of CAD, the higher the ex­
undergoing percutaneous coronary revascularization. The review also pected absolute benefit of antiplatelet therapy in reducing thrombotic
highlights the growing field of personalised antiplatelet therapy, includ­ complications (e.g. MI, stroke, stent thrombosis). However, antiplatelet
ing PFT and genotyping, and discusses their future directions and poten­ therapy carries an unavoidable risk of bleeding, which can be mitigated
tial implications. by weighing the benefits and risks for the individual patient and adjusting
Personalised antiplatelet therapies for CAD 3061

Downloaded from https://academic.oup.com/eurheartj/article/44/32/3059/7204812 by guest on 25 August 2023


Figure 1 Platelet activation mechanisms. Platelet activation is initiated by soluble agonists, such as thrombin, thromboxane A2 (TXA2), 5-hydroxy­
tryptamine (5-HT) and ADP (via the P2Y1 receptor), and by adhesive ligands, such as collagen and von Willebrand factor (vWF). Consequently, dense
granule secretion of platelet agonists and secretion of TXA2, as a result of phospholipase A2 activation, lead to the amplification of platelet activation and
the associated responses. The P2Y12 receptor has a major role in the amplification of platelet activation, which is also supported by outside–in signalling
via integrin αIIbβ3 (the glycoprotein IIb/IIIa receptor). Aspirin inhibits platelet function by acetylation of the platelet cyclooxygenase, which prevents the
access of arachidonic acid to the catalytic site of the enzyme and results in irreversible inhibition of platelet dependent TXA2 formation. Therefore, the
combined use of aspirin and a P2Y12 receptor inhibitor (such as clopidogrel, prasugrel, or ticagrelor) has additive effects on the inhibition of platelet
activation and the associated platelet responses. Conversely, discontinuation of aspirin results in some degree of increased platelet reactivity driven by
pathways mediated by arachidonic acid and collagen, whereas other pathways remain inhibited by P2Y12 inhibitor monotherapy. 5-HT2A, 5-hydroxy­
tryptamine receptor 2A; GPVI, platelet glycoprotein VI; NO, nitric oxide; PAR, proteinase-activated receptor; PGI2, prostaglandin I2; TPα, thromb­
oxane A2 receptor isoform α. Reproduced with permission from Capodanno et al.17

the treatment plan accordingly. Therefore, determining the fine balance two minor criteria from a set of variables defined by expert consensus.
between thrombotic and bleeding risks is crucial for the effective man­ Two models (DAPT score and ARC-HBR trade-off model) are also
agement of antiplatelet therapy. available that integrate ischemic and bleeding risk factors to provide a
prediction rule for outcomes of ‘net benefit’.14,15
Overall, the discriminatory performance of the available risk models
Predictors of thrombotic and bleeding for ischemia and/or bleeding is in the range of c-statistics between 0.65
events and 0.75 in validation studies10–15,21, indicating their suboptimal cap­
Risk stratification tools for the prediction of ischemia, bleeding, or their acity to distinguish the patients who will experience an event from
trade-off in CAD patients on dual antiplatelet therapy (DAPT, i.e. the and those who will not. Importantly, high bleeding risk patients have of­
combination of aspirin and a P2Y12 inhibitor) are summarized in ten been excluded from study populations used to derive bleeding risk
Table 1. 10–15 These models were generally derived from large cohorts scores, and these scores have been evaluated in the context of standard
of patients undergoing PCI and therefore their generalizability to other DAPT, which makes their generalisability uncertain when other scen­
DAPT settings (i.e. CAD without PCI) is uncertain. arios are considered (e.g. de-escalation). Notably, the ESC guidelines as­
The PARIS risk score encompasses six clinical variables for the pre­
sign a low class of recommendation to the use of risk scores to guide
diction of thrombosis and six variables for the prediction of bleeding.10
antiplatelet therapy (e.g. class IIb in the European guidelines for
Other scores for bleeding include PRECISE-DAPT,11 which uses five
clinical predictors, and BleeMACS, which uses seven clinical predic­ ACS).22 Therefore, clinical considerations and sound clinical judgment
tors.12 While these models follow a quantitative approach, a semiquan­ are crucial on top of utilizing these predictive models in guiding patient
titative approach from the Academic Research Consortium for High management and treatment decisions.21 Yet, identifying patients who
Bleeding Risk (ARC-HBR) has recently been introduced,13 with patients are most likely to benefit from antiplatelet therapy (e.g. those at very
at high bleeding risk defined as those fulfilling one major criterion or high risk of CAD events) and have a low risk of bleeding is challenging,
3062 D. Capodanno and D. J. Angiolillo

Table 1 Risk stratification tools for ischemia, bleeding, or their trade-off in patients on dual antiplatelet therapy

Score/Model No of Development cohort Setting Predicted outcome Validation


variables (patients, design) cohort(s) (patients,
c-index)
......................................................................................................................................................................................
Ischemia
PARIS Six clinical 4190 patients, multicentre registry PCI patients on DAPT Ischemia at 24 months 8665 patients, 0.65
thrombosis10 after PCI
Bleeding
PARIS bleeding10 Six clinical 4190 patients, multicentre registry PCI patients on DAPT Bleeding at 24 months 8665 patients, 0.64
after PCI

Downloaded from https://academic.oup.com/eurheartj/article/44/32/3059/7204812 by guest on 25 August 2023


PRECISE-DAPT11 Five clinical 14 963 patients, pooled analysis of PCI patients on DAPT Bleeding at 12 months 8595 patients, 0.70;
randomised clinical trials after PCI 6172 patients, 0.66
BleeMACS12 Seven clinical 15 401 patients, multicentre PCI patients on DAPT Bleeding at 12 months 96 239 patients, 0.65
registry after PCI
ARC-HBR13 Thirteen clinical - PCI patients on DAPT Bleeding at 12 months -
after PCI
Trade-off
DAPT score14 Five clinical, 11 648 patients, multicentre PCI patients on DAPT Ischemia and bleeding 8136 patients, 0.64 for
three randomised clinical trial who were event-free between 12 and 30 both ischaemia and
procedural for 12 months months after PCI bleeding
ARC-HBR Ten clinical, 6641 patients, pooled analysis of PCI patients on DAPT Ischemia and bleeding at 1458 patients, 0.74 for
trade-off15 two procedural randomised clinical trials and a 12 months after PCI both ischaemia and
multicentre registry bleeding

ARC-HBR, Academic Research Consortium for High Bleeding Risk; BleeMACS, Bleeding Complications in a Multicenter Registry of Patients Discharged With Diagnosis of Acute
Coronary Syndrome; DAPT, Dual Antiplatelet Therapy Trial; PARIS, patterns of non-adherence to anti-platelet regimens in stented patients; PCI, percutaneous coronary
intervention; PRECISE-DAPT, Predicting Bleeding Complications In Patients Undergoing Stent Implantation and Subsequent Dual Anti Platelet Therapy.

especially because patients at increased ischemic and bleeding risks typ­ a more intensive antiplatelet therapy regimen, while those at high risk of
ically share common CAD risk factors.15,23 bleeding and low risk of ischemic events require a less intensive and
Additionally, patients with established atherosclerotic cardiovascular more cautious approach. In patients at low risk of ischemic events
disease are generally considered at very high risk of events by current and low risk of bleeding, the benefits of antiplatelet therapy may not
ESC guidelines for secondary prevention.3 This broad group include outweigh the risks, while patients who have high risks of both ischemic
those with documented CAD, including previous MI, ACS, and coronary and bleeding events require a delicate balance, as the approach to man­
revascularization. Patients with unequivocally documented cardiovascular aging their conditions must weigh the potential benefits against the po­
disease on imaging are also considered at very high risk, including those tential risks.
with atherosclerotic plaques on coronary angiography or computed tom­ There are two potential strategies to personalising the antiplatelet
ography angiography. treatment regimen based on the individual risk of ischemic and bleeding
A scrutiny of most common predictors derived by the available risk complications.8 The first strategy is an approach based on clinical judg­
stratification tools (Table 2) allows to isolate some crucial risk determi­ ment, where the practitioner uses commonly available parameters, in­
nants of ischemic complications in spite of antiplatelet therapy (e.g. dia­ cluding clinical, demographic, laboratory, and procedural, along with
betes mellitus, ACS or MI at presentation, prior PCI, MI or their experience to make decisions. The second strategy is an approach
cerebrovascular accident, smoking, renal insufficiency, peripheral artery based on guidance from assays that may inform on how a patient is re­
disease) and a number of key predictors of bleeding (e.g. age, renal in­ sponding (i.e. PFT) or may respond (i.e. genetic testing) to an antiplate­
sufficiency, cirrhosis, anaemia or low haemoglobin levels, oral anticoa­ let agent. This may include results from PFT that measure the
gulation, prior bleeding and/or transfusion, cancer) that are visually individual’s response to an antiplatelet drug which correlates with an
schematized in Figure 2. adverse outcome (e.g. hyper- and hypo-responders with bleeding and
thrombotic events, respectively). Alternatively, genetic testing provides
information about how an individual may respond to a given antiplatelet
Personalised approach agent (e.g. genetic polymorphisms coding for an enzyme involved in
Tailoring the right antiplatelet therapy to individual ischemic and bleed­ drug metabolism may lead to different degrees of enzyme activity).
ing characteristics is crucial in optimizing the benefits and minimizing the Both strategies aim to optimize the benefits of antiplatelet therapy in
risks of treatment for patients with CAD (Graphical abstract).1 Patients reducing the risk of ischemic complications and mitigate the risk of
at high risk of ischemic events and low risk of bleeding may benefit from bleeding complications.
Personalised antiplatelet therapies for CAD 3063

Table 2 Predictors of ischemia and/or bleeding in patients on dual antiplatelet therapy

PARIS10 PRECISE BleeMACS12 ARC-HBR13 DAPT ARC-HBR


DAPT11 score14 Trade-off15
......................................................................................................................................................................................
Predictors of ischemia
Diabetes ◉ NA NA NA ◉ ◉
Smoking ◉ NA NA NA ◉ ◉
ACS or MI at presentation ◉ NA NA NA ◉ ◉
Prior PCI or MI ◉ NA NA NA ◉ ◉
Renal insufficiency ◉ NA NA NA ◉ ◉

Downloaded from https://academic.oup.com/eurheartj/article/44/32/3059/7204812 by guest on 25 August 2023


Prior CABG ◉ NA NA NA – –
Small stents – NA NA NA ◉ –
CHF or low LVEF – NA NA NA ◉ –
Paclitaxel-eluting stent – NA NA NA ◉ –
Vein graft stent – NA NA NA ◉ –
Vascular disease – NA NA NA ◉ –
Hypertension – NA NA NA ◉ –
Anaemia or haemoglobin – NA NA NA – ◉
Complex PCI – NA NA NA – ◉
Bare metal stent – NA NA NA – ◉
Predictors of bleeding
Age ◉ ◉ ◉ ◉ ◉ ◉
Renal insufficiency ◉ ◉ ◉ ◉ ◉ ◉
Anaemia or low haemoglobin ◉ ◉ ◉ ◉ – ◉
Oral anticoagulation ◉ – – ◉ – ◉
Prior bleeding and/or transfusion – ◉ ◉ ◉ – –
Cancer – – ◉ ◉ – ◉
Smoking ◉ – – – – ◉
Liver cirrhosis – – – ◉ – ◉
Planned major noncardiac surgery – – – ◉ – ◉
Hypertension – – ◉ – ◉ –
Vascular disease – – ◉ – ◉ –
Low or high body mass index ◉ – – – – –
Leucocytosis – ◉ – – – –
Thrombocytopenia – – – ◉ – –
Chronic bleeding diathesis – – – ◉ – –
Long-term use of NSAIDs or steroids – – – ◉ – –
Previous ischemic stroke or ICH – – – ◉ – –
Recent major surgery or trauma – – – ◉ – –
COPD – – – – – ◉
Complex PCI – – – – – ◉

Abbreviations: ACS, acute coronary syndromes; CABG, coronary artery bypass surgery; CHF, congestive heart failure; COPD, chronic obstructive pulmonary disease; ICH, intracranial
haemorrhage; LVEF, low ventricular ejection fraction, MI, myocardial infarction; NA, not applicable; NSAIDs, nonsteroidal anti-inflammatory drugs; PCI, percutaneous coronary
intervention. Other abbreviations as in Table 1.
3064 D. Capodanno and D. J. Angiolillo

Diabetes Smoking ACS/MI Age CKD Cirrhosis

CKD Cancer
Predictors Predictors

Downloaded from https://academic.oup.com/eurheartj/article/44/32/3059/7204812 by guest on 25 August 2023


of ischaemia of bleeding

PAD Anemia

Prior MI Prior PCI Prior CVA OAC Transfusion Prior bleed

Figure 2 Determinants of ischaemic and bleeding risk in patients on dual antiplatelet therapy. Abbreviations: ACS, acute coronary syndrome; CKD,
chronic kidney disease; CVA, cerebrovascular accidents; MI, myocardial infarction; OAC, oral anticoagulation; PAD, peripheral artery disease; PCI, per­
cutaneous coronary intervention.

Clinically guided approach De-escalation—By reducing the intensity of platelet inhibition,


The standard approach to antiplatelet therapy in patients with CAD is to de-escalation is an approach aimed at decreasing the risk of bleeding
prescribe 6 months of DAPT after PCI for CCS and 12 months after an complications when they are deemed to be greater than the risk of
ACS with or without PCI.22,24–27 In particular, in addition to aspirin, clo­ thrombotic complications. This can be achieved by adjusting the type,
pidogrel is the P2Y12 inhibitor of choice in patients with CCS, while pra­ dose, or number of antiplatelet drugs used. Several trials have investi­
sugrel (in patients undergoing PCI) and ticagrelor (regardless of gated these approaches in contemporary populations mostly including
revascularization) are preferred over clopidogrel in patients with ACS, patients undergoing PCI. Importantly, some of these trials only rando­
with ESC guidelines recommending prasugrel over ticagrelor in patients mized patients who did not experience a thrombotic or bleeding during
undergoing PCI.22,28 Indeed, the duration of DAPT can be prolonged or the initial period of DAPT.35,42,47 This may have resulted in some de­
shortened according the ischemic and bleeding risk profile of the patient. gree of patient selection and therefore less generalizability compared
In patients who are at increased risk for both ischemic and bleeding com­ with trials that randomized at the time of PCI. On the other hand,
plications, bleeding, more than ischemic risk or PCI complexity, should this design avoided the potential confounding arising from events oc­
inform decision-making on the duration of DAPT.29 After completion of curring at that time when the two arms were on the same regimen.
DAPT, patients should resume single antiplatelet therapy (SAPT). A first common form of de-escalation is switching (i.e. adjusting drug
Aspirin has been the standard of care for SAPT in most patients with type approach), where a clinician changes from an agent associated with
CAD. However, there is emerging evidence in the post-PCI setting sup­ more potent to less potent P2Y12 inhibition.52 This is often performed
porting the use of a P2Y12 inhibitor as chronic monotherapy instead.30,31 in patients who have been initially treated for an ACS with a guideline
In addition to variations in the duration of DAPT, the availability of recommended DAPT regimen including prasugrel or ticagrelor, as
different oral antiplatelet agents as well as a better understanding of these drugs are associated with an increased risk of major spontaneous
bleeding compared to clopidogrel.53,54 Two trials in ACS, named
the timing at which patients may be at an enhanced risk of thrombotic
TOPIC and TALOS-AMI, have demonstrated the net benefit of an un­
or bleeding complications has led strategies of modulation of the inten­
guided switching to clopidogrel at 1 month post-ACS.34,35 Both trials
sity of antiplatelet therapy as an approach to optimize the safety–
reported a reduction in bleeding as the driving factor behind the im­
efficacy balance in an individual patient. In particular, modulation of
proved outcomes.
the antiplatelet treatment regimen may occur in two ways: by reducing Another approach to de-escalating antiplatelet therapy is reducing the
(i.e. de-escalation) or increasing (i.e. escalation) the intensity of platelet dose (i.e. adjusting drug dose approach) of the P2Y12 inhibitor character­
inhibition (Figure 3).32,33 Table 3 summarizes the results of contempor­ ized by enhanced platelet inhibitory effects (i.e. prasugrel or ticagrelor)
ary randomized clinical trials of antiplatelet therapy modulation. The re­ used in DAPT combinations. This strategy may be desirable when there
sults of these studies can guide a patient-centric approach where are concerns about excessive drug exposure, particularly in ethnicities
physicians use their clinical judgment and expertise to determine which such as East Asian patients. One trial, the HOST-REDUCE, found that
candidates are best suited for a specific investigational treatment plan. reducing the maintenance dose of prasugrel from 10 to 5 mg once daily
Personalised antiplatelet therapies for CAD 3065

De-escalation

Switching

Increase in Dose reduction


intensity of platelet
inhibition Discontinuation

Decrease in
Dec
Switching

Downloaded from https://academic.oup.com/eurheartj/article/44/32/3059/7204812 by guest on 25 August 2023


intensity of platelet
inhibition
Dose increase

Add-on

Escalation

Figure 3 Strategies of antiplatelet therapy modulation. Antiplatelet therapy can be modulated in two ways: by reducing (i.e. de-escalation) or increas­
ing (i.e. escalation) the intensity of platelet inhibition.

at 1 month reduced the risk of net adverse cardiac events (NACE) at 1 TICO45 and STOPDAPT-2 ACS,46 compared early aspirin discontinu­
year in patients with ACS undergoing PCI.36 Also, the recommended ation and maintaining SAPT with a P2Y12 inhibitor vs. 12-month DAPT
maintenance dose of ticagrelor is 90 mg twice daily for the first year after on 1-year NACE specifically in ACS patients: TICO (testing SAPT with
an ACS but de-escalation to a dosing regimen of 60 mg twice daily can be ticagrelor) showed superiority, while STOPDAPT-2 ACS (testing SAPT
used after 1 year based on the results of the PEGASUS-TIMI 54 trial, with clopidogrel) failed in showing noninferiority. Although PFT or gen­
where the 60 mg dosing regimen was found to have similar efficacy to etic testing were not performed in STOPDAPT-2 ACS, it may be ar­
the 90 mg dosing regimen but had a more favorable safety profile in gued that its findings could be in part attributed to impaired
terms of bleeding and non-bleeding side effects.50 response to clopidogrel which more commonly occurs in high-risk set­
A third viable de-escalation approach is the discontinuation of DAPT tings. Indeed, this represents an opportunity for future studies of indi­
(i.e. adjusting drug number approach), which means stopping of one of vidualized treatment options in which defining response to therapy
the DAPT antiplatelet agents and transitioning to SAPT with aspirin or a when relying on SAPT with P2Y12 inhibitor monotherapy should be
P2Y12 inhibitor. The timing of discontinuation has progressively been considered. Of note, ticagrelor monotherapy after a short period of
moved earlier after PCI with the current generation of coronary stents. DAPT showed promising results also in ACS sub-analyses of the
Several single-arm studies using historical controls or objective per­ GLOBAL LEADERS59 and TWILIGHT60 trials.
formance goals support the concept that 1 to 3 months of DAPT Finally, some trials compared standard DAPT with strategies of as­
may be enough in patients at high risk of bleeding.47,55–57 Regardless pirin discontinuation or P2Y12 inhibitor monotherapy at the physician’s
of bleeding risk, studies such as the ONE-MONTH DAPT trial37,58 in discretion. The MASTER DAPT trial selectively randomized patients at
patients with CCS or ACS, and the SMART-DATE trial,38 high bleeding risk and found that discontinuing the P2Y12 inhibitor or
REDUCE-ACS,39 and DAPT-STEMI40 trials in patients with ACS have aspirin at 1 month was noninferior to continuing for at least two add­
shown that early discontinuation of the P2Y12 inhibitor with transition itional months with respect to 1-year NACE or major adverse cardiac
from DAPT to aspirin monotherapy (i.e. at 1 to 6 months) is noninfer­ events (MACE), and was associated with reduced bleeding.61 In the
ior to discontinuation at 6 to 12 months in terms of 1-year NACE. HOST-IDEA trial, the discontinuation of one of antiplatelet agent at
These trials used wide noninferiority margins, which raises a note of 3–6 months was noninferior with respect to 1-year NACE in patients
caution over generalizing their results. Additionally, there were signals with CCS or ACS undergoing PCI.48
of an increase in thrombotic complications with shorter DAPT espe­ Escalation—Antiplatelet therapy escalation is meant to decrease
cially in ACS.38,39,58 thrombotic or ischemic complications by increasing the intensity of
Several studies compared strategies of aspirin discontinuation and platelet inhibition at a time when such risk is considered greater than
P2Y12 inhibitor monotherapy after a short (e.g. 1–3 months) period the risk of bleeding complications. Again, this can be achieved by adjust­
of DAPT in patients undergoing PCI, including GLOBAL LEADERS41 ing the type, dose, or number of antiplatelet drugs used.
and TWILIGHT42 (testing SAPT with ticagrelor), and Escalation by switching refers to the practice of changing from a
STOPDAPT-243 and SMART-CHOICE44 (testing SAPT with clopido­ platelet P2Y12 receptor inhibitor with moderate platelet inhibitory ef­
grel). With the exception of the GLOBAL LEADERS that did not fects, such as clopidogrel, to one with stronger platelet inhibitory ef­
meet its primary objective, these studies have shown a reduction in fects, such as prasugrel or ticagrelor.52 This change is sometimes
bleeding without any trade-off in ischemic events with P2Y12 inhibitor considered for patients treated with a clopidogrel-based DAPT regi­
monotherapy compared with standard DAPT. Two more trials, named men who present with an acute cardiac event or patients with CCS
3066 D. Capodanno and D. J. Angiolillo

Table 3 Contemporary randomized controlled trials of unselective modulation of antiplatelet therapy

Trial Modulation Strategy n Population Follow-up Primary Findings


endpoint
......................................................................................................................................................................................
TOPIC34 De-escalation Switch from prasugrel or 646 ACS and PCI 12 months NACE HR, 0.48; 95% CI, 0.34–0.68;
ticagrelor to clopidogrel P < 0.01
TALOS-AMI35 De-escalation Switch from ticagrelor to 2697 Prior ACS 11 months NACE HR, 0.55; 95% CI, 0.40–0.76;
clopidogrel and PCI P = 0.0001
HOST-REDUCE36 De-escalation Dose reduction from prasugrel 10 2338 ACS and PCI 12 months NACE HR, 0.70; 95% CI, 0.52–0.92;
to 5 mg once daily P = 0.012
ONE-MONTH De-escalation Discontinuation of the P2Y12 3020 CCS and PCI 12 months NACE ARD, -0.7%; upper limit of

Downloaded from https://academic.oup.com/eurheartj/article/44/32/3059/7204812 by guest on 25 August 2023


DAPT37 inhibitor (1 vs. 6–12 months of 1-sided 97.5% CI, 1.33%; P <
DAPT) 0.001 for noninferiority
SMART-DATE38 De-escalation Discontinuation of the P2Y12 2712 ACS and PCI 18 months MACE ARD, 0.5%; upper limit of
inhibitor (6 vs. 12 months of 1-sided 95% CI, 1.8%; P = 0.03
DAPT) for noninferiority
REDUCE ACS39 De-escalation Discontinuation of the P2Y12 1496 ACS and PCI 12 months NACE ARD, -0.0022%; upper limit of
inhibitor (3 vs. 12 months of 1-sided 95% CI, 0.027%; P <
DAPT) 0.001 for noninferiority
DAPT STEMI40 De-escalation Discontinuation of the P2Y12 1100 Prior ACS 18 months NACE HR, 0.73; 95% CI 0.41–1.27;
inhibitor (6 vs. 12 months of and PCI P = 0.004 for noninferiority
DAPT)
GLOBAL De-escalation Discontinuation of aspirin (1 vs. 12 15 968 PCI 24 months MACE Rate ratio, 0.87; 95% CI, 0.75–
LEADERS41 months of DAPT) 1.01; P = 0.073)
TWILIGHT42 De-escalation Discontinuation of aspirin (3 vs. 12 7119 PCI 12 months Bleeding HR, 0.56; 95% CI, 0.45–0.68;
months of DAPT) P < 0.001
STOPDAPT-243 De-escalation Discontinuation of aspirin (1 vs. 12 3045 PCI 12 months NACE HR, 0.64; 95% CI, 0.42–0.98;
months of DAPT) P = 0.04
SMART-CHOICE44 De-escalation Discontinuation of aspirin (3 vs. 12 2993 PCI 12 months MACE ARD, 0.4%, upper limit of
months of DAPT) 1-sided 95% CI, 1.3%; P =
0.007 for noninferiority
TICO45 De-escalation Discontinuation of aspirin (3 vs. 12 3056 ACS 12 months NACE HR, 0.66; 95% CI, 0.48–0.92;
months of DAPT) P = 0.01
STOPDAPT-2 De-escalation Discontinuation of aspirin (1–2 vs. 4169 ACS 12 months NACE HR, 1.14; 95% CI, 0.80–1.62;
ACS46 12 months of DAPT) P = 0.06 for noninferiority
MASTER DAPT47 De-escalation Discontinuation of aspirin or the 4434 PCI 11 months NACE ARD, -0.23%, upper limit of
P2Y12 inhibitor (1 vs. ≥3 months 95% CI, 1.33%; P < 0.001 for
of DAPT) noninferiority
HOST-IDEA48 De-escalation Discontinuation or the P2Y12 2013 PCI 12 months NACE ARD, -0.4%, upper limit of
inhibitor (3–6 vs. 12 months of 1-sided 95%, 1.1%; P < 0.001
DAPT) for noninferiority
DAPT49 Escalation Add-on P2Y12 inhibitor (30 vs. 9961 Prior PCI 18 months MACE HR, 0.71; 95% CI, 0.59–0.85;
12gr months of DAPT) P < 0.001
PEGASUS-TIMI Escalation Add-on ticagrelor (long-term 21 162 Prior MI ∼33 MACE HR (ticagrelor 90 mg), 0.85;
5450 DAPT vs. aspirin) months 95% CI, 0.75–0.96; P = 0.008;
HR (ticagrelor 60 mg), 0.84;
95% CI, 0.74–0.95; P = 0.004
THEMIS51 Escalation Add-on ticagrelor (long-term 19 220 CCS and ∼40 MACE HR, 0.90; 95% CI, 0.81–0.99;
DAPT vs. aspirin) diabetes months P = 0.04

Treatment effects are reported for the investigational strategy vs. standard dual antiplatelet therapy. In MASTER DAPT, NACE is reported as the first ranked primary outcome. P-values
are for superiority unless otherwise specified. Abbreviations: ACS, acute coronary syndrome; ARD, absolute risk difference; CCS, chronic coronary syndrome; CI, confidence interval;
DAPT, dual antiplatelet therapy; HR, hazard ratio; MACE, major adverse cardiac events; MI, myocardial infarction; NACE, net adverse cardiac events; PCI, percutaneous coronary
intervention.
Personalised antiplatelet therapies for CAD 3067

Table 4 Clopidogrel therapy based on CYP2C19 phenotype for ACS/PCI patients initiating antiplatelet therapy

Phenotype (genotype) Implications for clopidogrel Therapeutic recommendations


......................................................................................................................................................................................
UM (*1/*17, *17/*17) and Normal (EM) or increased (UM) platelet inhibition; normal (EM) or decreased Clopidogrel label-recommended dosage
EM (*1/*1) (UM) residual platelet aggregation and administration
Intermediate metabolizers Reduced platelet inhibition; increased residual platelet aggregation; increased Prasugrel or ticagrelor (if no
(*1/*2) risk for adverse cardiovascular events contraindications)
PM (*2/*2) Significantly reduced platelet inhibition; increased residual platelet aggregation; Prasugrel or ticagrelor (if no
increased risk for adverse cardiovascular events contraindications)

Adapted from Scott et al.72 Abbreviations: EM, extensive metabolizers; PM, poor metabolizers; UM, ultrarapid metabolizers.

Downloaded from https://academic.oup.com/eurheartj/article/44/32/3059/7204812 by guest on 25 August 2023


undergoing high-risk PCI. However, there are currently no data to sup­ of active metabolite, reduced clopidogrel-induced platelet inhibition
port that prasugrel or ticagrelor are more effective than clopidogrel and higher rates of thrombotic complications in patients undergoing
among patients undergoing elective PCI.62,63 PCI.70,71 The most common LOF alleles include CYP2C19*2 and
Escalation by dose increase refers to the practice of increasing the CYP2C19*3; less common LOF alleles include CYP2C19 *4, *5, *6, *7,
dose of an antiplatelet drug with the goal of intensifying the platelet in­ and *8. On the contrary, CYP2C19*17 is considered a gain of function
hibitory effect. This approach has been tested in a clinical trial of allele and is associated with increased transcription and enzyme expres­
doubled clopidogrel or aspirin dose for patients undergoing PCI64 sion.9 Combinations of CYP2C19 alleles define the metabolizer status
and a trial of doubled aspirin dose for patients with cardiovascular dis­ and serve for the basis of therapeutic recommendations (Table 4). 72
ease,65 but in both cases, the results have been neutral. Allele frequencies vary by ancestry, with the highest prevalences among
‘Add-on’ escalation refers to the practice of adding a second antipla­ of East Asians.73
telet drug to a patient who is already receiving SAPT, most commonly Several studies have tried to answer whether a strategy of tailoring
aspirin. This strategy results in DAPT, and has been shown to be super­ antiplatelet therapy with guidance from PFT or genotyping is associated
ior to SAPT in reducing ischemic events in various patient populations, with clinical benefit (Table 5). A meta-analysis including 11 randomised
including those with a history of prior PCI from the DAPT trial,49 those controlled trials and three observational studies of guided selection of
with prior MI from the PEGASUS-TIMI 54 trial,50 and those with CAD antiplatelet therapy (PFT and genetic testing) for patients undergoing
but without a prior acute cardiovascular event and with diabetes melli­ PCI (n = 20 743 patients) showed improvements in both composite
tus from the THEMIS trial.51 The benefit of DAPT in THEMIS was more and individual efficacy outcomes.83 More specifically, outcomes varied
pronounced in patients with prior PCI.66 according to the strategy used, with an escalation approach associated
with a significant reduction in ischaemic events without any trade-off in
safety, and a de-escalation approach by switch or dose reduction asso­
Platelet function and genotype-guided ciated with a significant reduction in bleeding, without any trade-off in
approaches efficacy. In a more recent network meta-analysis of 15 randomised con­
Despite compliance to antiplatelet treatment, some patients may still trolled trials in patients with an ACS (n = 61 898 patients), compared
experience ischemic events, a phenomenon known as ‘therapeutic fail­ with routine selection of potent P2Y12 inhibiting therapy (prasugrel
ure’.67 A number of factors contribute to therapeutic failure, including or ticagrelor), a guided selection of P2Y12 inhibiting therapy was found
impaired response to an antiplatelet agent, commonly referred to as to be associated with the most favourable balance between safety and
‘drug resistance’. Such finding is supported by a number of observation­ efficacy.84
al studies, mostly conducted in patients undergoing PCI and treated PFT—The availability of user-friendly and bedside assays has enabled
with clopidogrel-based DAPT, in which the use of PFT has shown the execution of multicentre randomized clinical trials in patients
that individuals who persist with high on-treatment platelet reactivity undergoing PCI testing the impact on outcomes associated with the
(HPR) are at increased risk for thrombotic complications.68 Some stud­ use of PFT. Some of these trials where specifically conducted among pa­
ies have also shown that patients with low on-treatment platelet re­ tients with HPR (identified by PFT) and comparing different antiplatelet
activity are at increased risk of bleeding,68 although the overall treatments,85,86 while other trials compared PFT guidance used as a
evidence seems more robust for the prediction of ischemic risk. strategy vs. standard care.74–77
The efficacy of clopidogrel is dependent on a two-step activation The GRAVITAS trial included patients with HPR, who were ran­
process by the hepatic cytochrome P450 (CYP) system, with the domly assigned to high-dose clopidogrel or standard-dose clopido­
CYP2C19 enzyme involved in both metabolic steps.9,69 Notably, grel; the primary composite outcome showed no significant
CYP2C19 is highly polymorphic, meaning that variants in the gene confer difference between the groups at 6 months.86 The TRIGGER PCI trial,
wide variability in the enzyme’s metabolic activity across individuals. also conducted selectively in patients with HPR, compared prasugrel
Therefore, there is wide variability in the activation of clopidogrel and and clopidogrel, but was terminated prematurely due to a
in the resulting platelet inhibitory effects across individuals. Prasugrel lower-than-anticipated risk of events at 6 months.85 The ARCTIC
and ticagrelor, however, are not affected by such genetic polymorph­ and ANTARCTIC studies, evaluating a bedside strategy for monitor­
isms resulting in more consistent and sustained platelet inhibitory ef­ ing and adjusting antiplatelet therapy (i.e. de-escalation or escalation
fects.9 Carriers of CYP2C19 loss of function (LOF) alleles have by switch or dose adjustment as appropriate), also failed to show sig­
significantly reduced clopidogrel metabolism, resulting in lower levels nificant benefits.74,75
3068 D. Capodanno and D. J. Angiolillo

Table 5 Strategy trials of modulation of antiplatelet therapy guided by platelet function or genetic testing in patients
undergoing percutaneous coronary intervention

Trial Intervention n Population Follow-up Primary Findings


endpoint
......................................................................................................................................................................................
Trials of PFT guidance
ARCTIC74 PFT-guided escalation or de-escalation 2240 CCS or ACS 12 months MACE HR, 1.13; 95% CI, 0.98–1.29; P = 0.10
by switch or dose adjustment
ANTARCTIC75 PFT-guided escalation or de-escalation 877 ACS 12 months NACE HR, 1.00, 95% CI 0.78–1.29; P = 0.98
by switch or dose adjustment
TROPICAL ACS76 PFT-guided de-escalation by switch 2610 ACS 12 months NACE HR, 0.81; 95% CI, 0.62–1.06;

Downloaded from https://academic.oup.com/eurheartj/article/44/32/3059/7204812 by guest on 25 August 2023


P = 0.0004 for noninferiority
PATH-PCI77 PFT-guided escalation by switch 2237 CCS 6 months NACE HR, 0.68, 95% CI, 0.49–0.95; P = 0.023
Trials of genetic guidance
PHARMCLO78 Genotype-guided de-escalation by 888 ACS 12 months NACE HR, 0.58; 95% CI, 0.43–0.78; P < 0.001
switch
POPULAR Genotype-guided de-escalation by 2488 ACS 12 months NACE ARD, -0.7%; upper limit of the 95% CI,
GENETICS79 switch 0.7%; P < 0.001 for noninferiority
ADAPT-PCI80 Genotype-guided escalation by switch 509 CCS or ACS ∼16 Use of prasugrel OR, 1.60; 95% CI, 1.07–2.42; P = 0.03
months or ticagrelor
TAILOR-PCI81 Genotype-guided escalation by switch 1849 CCS or ACS 12 months MACE HR, 0.66; 95% CI, 0.43–1.02; P = 0.06
82
Al-Rubaish et al. Genotype-guided escalation by switch 755 ACS 12 months NACE OR, 0.34; 95% CI, 0.20–0.59; P = NA

Treatment effects are reported for the investigational strategy vs. standard dual antiplatelet therapy; P-values are for superiority unless otherwise specified. Abbreviations: ACS; acute
coronary syndrome; ARD, absolute risk difference; CI, confidence interval; HPR, high platelet reactivity; HR, hazard ratio; LOF, loss of function; MACE; major adverse cardiac events; NA,
not available; NACE, net adverse cardiac events; OR, odds ratio; PCI, percutaneous coronary intervention; PFT, platelet function testing.

The inability of these early trials to demonstrate any clinical benefit In the ADAPT-PCI trial,80 the genotype-guided prescription was
with the use of PFT could be attributed to a number of factors, such found to significantly increase the use of prasugrel or ticagrelor com­
as inclusion of low-risk patients, limited use of novel generation pared to an unguided approach in patients with ACS or CCS undergo­
P2Y12 inhibitors, and inadequate cut-off values to define HPR. More re­ ing PCI. TAILOR-PCI81 was the largest trial of genotype-guided
cent studies conducted in high-risk settings have shown more promis­ selection of oral P2Y12 inhibitor in which patients with ACS or CCS
ing findings.76,77 TROPICAL-ACS trial enrolled patients with ACS undergoing PCI were randomized to genotype-guidance or conven­
undergoing PCI, randomly assigned to either PFT guidance with de- tional therapy. In the genotype-guide group, CYP2C19 LOF carriers
escalation by switch from prasugrel to clopidogrel if responders or were prescribed ticagrelor and noncarriers clopidogrel. Patients rando­
standard care.76 The trial, powered for noninferiority on NACE vs. mized to the conventional group were prescribed clopidogrel.
standard DAPT, met its primary objective. The PATH-PCI trial also Although the genotype-guided approach showed a numerical reduction
met its primary objective, showing superiority for the PFT-based in MACE, this did not reach statistical significance, possibly due to a lack
approach.77 of power. However, a recent analysis from the TAILOR-PCI study did
Guidance by genetic testing—Rapid CYP2C19 genotyping as­ show superiority of a genotype-guided approach when considering re­
says, with results available within 60 min, are commercially available current events.89 Conversely, a trial from Al-Rubaish et al.,82 who only
making genetic testing feasible in real-world practice.87,88 Genetic included patients with ACS, showed a significant benefit of the
testing has been evaluated as a method of guiding antiplatelet therapy genotype-guided strategy in reducing NACE.
in five studies of patients with PCI and/or ACS, with mixed results78–82
(Table 5).
In patients with ACS undergoing PCI, the PHARMCLO78 and PFT and genetic testing: advantages and
POPULAR GENETICS79 trials compared the genotype-based selection disadvantages
of P2Y12 inhibitors (i.e. selective use of prasugrel or ticagrelor among PFT has the advantage of being more closely related with the clinical
CYP2C19 LOF carriers and clopidogrel in noncarriers) to unguided outcome (i.e. increased thrombotic and bleeding complications with
standard of care DAPT. Despite being terminated early and therefore high and low platelet reactivity, respectively).16 Nevertheless, results
resulting in lower power than anticipated, PHARMCLO showed the of PFTs are subject to variability, particularly with clopidogrel early after
genotype-based approach to significantly reduce the risk of NACE. treatment initiation, requiring patients to be on treatment for a certain
Similarly, in POPULAR GENETICS, the genotype-based approach was period of time (e.g. for at least 1–2 weeks) to adequately assess antipla­
non-inferior to standard therapy in terms of NACE at 12 months, and telet drug response. Hence, the potential need for serial assessments as
resulted in a significantly lower incidence of major or minor bleedings. well as switching between different oral P2Y12 inhibitors may be
Personalised antiplatelet therapies for CAD 3069

challenging to implement in clinical practice.90 In contrast, results of escalation by switch of P2Y12 receptor inhibitor treatment (e.g. from
genetic testing remain unchanged and allow to determine, even prior prasugrel or ticagrelor to clopidogrel), or de-escalation by dose
to therapy, the CYP2C19 phenotype associated with different degrees reduction, as an alternative DAPT strategy ‘especially for ACS patients
of clopidogrel response (Table 4). Accordingly, CYP2C19 LOF carriers deemed unsuitable for potent platelet inhibition’. Also, the recommenda­
can be treated with ticagrelor or prasugrel while noncarriers with clo­ tion states that de-escalation by switch ‘may be done unguided based on
pidogrel. The disadvantage of relying solely on genetic testing is that the clinical judgment or guided by PFT or CYP2C19 genotyping, depending on
pharmacodynamic effects of clopidogrel as measured by PFT depend patient’s risk profile and availability of respective assays’. The level of evi­
on multiple factors and not solely on CYP2C19 genotypes.91 To this dence for this recommendation is A, with the TOPIC,
extent, integrating genetic data with clinical variables, such as in the TROPICAL-ACS, and POPULAR GENETICS trials used as supporting
ABCD-GENE (Age, Body Mass Index, Chronic Kidney Disease, references. The phrasing of the recommendation, referring to patients
Diabetes, and Genotyping) score has shown to enhance the accuracy ‘unsuitable for potent platelet inhibition’, implies that these approaches
in identifying patients with impaired clopidogrel response.92 Although for now are best suited to patients at high bleeding risk, a population
retrospective assessments have shown the ability of the that was actually understudied in the abovementioned trials, while

Downloaded from https://academic.oup.com/eurheartj/article/44/32/3059/7204812 by guest on 25 August 2023


ABCD-GENE score to identify patients at increased ischemic risk, pro­ the application of a strategy should reflect the types of patients enrolled
spective validation of this score is still warranted.93,94 in the respective trials. No recommendation for de-escalation by switch
has been issued by the recent American guidelines for myocardial
Antiplatelet therapies, revascularization.4
The ESC guidelines also include class IIa recommendations for de-
personalised strategies under escalation by discontinuation of the P2Y12 inhibitor at 3 months (level
clinical testing, and unmet needs of evidence B, in patients at high bleeding risk as define by the
PRECISE-DAPT score or by the ARC-HBR criteria) or aspirin at 3–6
The challenge for new antiplatelet drugs for CAD is to exhibit less
months (level of evidence A, ‘depending on the balance between the ische­
bleeding without reducing antithrombotic potency. Some of these de­
mic and bleeding risk’). In the American guidelines, the recommendation
velopments may include new formulations of already available drugs,
for P2Y12 inhibitor discontinuation at 6 months is class 2b (for patients
such as a phospholipid-aspirin complex which is designed to reduce
at ‘high bleeding risk or overt bleeding on DAPT’) and the recommendation
gastro-intestinal injury and readily absorbed to ensure antithrombotic
for aspirin at 1–3 months discontinuation is 2a (potentially for any
efficacy.95 Other drugs include novel experimental agents aimed at
patient).
new targets. Potential new therapeutics are in advanced preclinical de­
Additionally, the ESC guidelines include recommendations for escal­
velopment or have already entered the clinical development phase.96 It
ation by ‘add-on’ of a P2Y12 inhibitor (class IIa, level of evidence A for
remains to be seen how these new agents will fit within current para­
patients ‘with a high risk of ischemic events and without increased risk of
digms of antiplatelet therapy and whether they will lead to safer com­
major or life-threatening bleeding’ and class IIb, level of evidence A for pa­
binations in clinical practice.
tients ‘with a moderate risk of ischemic events and without increased risk of
The value of risk stratification tools to guide clinical decisions is typ­
major or life-threatening bleeding’). In the American guidelines, this ap­
ically assessed by means of c-statistics indicating their discrimination
proach is recommended with class 2b for patients ‘with no high risk of
ability. However, whether a risk score is helpful in improving clinical
bleeding and no significant overt bleeding on DAPT’.
outcomes requires an intervention-based randomized clinical study.
Trials of personalised duration of antiplatelet therapy based on the
DAPT score (NCT04135989) and the PRECISE-DAPT score Conclusions
(NCT05732701, NCT03848572) are underway. Several ongoing ran­
domized clinical trials are also addressing the role of PFT-guided strat­ Personalised antiplatelet therapy has emerged as a crucial approach in
egies of antiplatelet therapy modulation in patients with ACS optimizing the balance between efficacy and safety by customizing anti­
(NCT04755387, NCT04718025, NCT04240834, NCT04937699, platelet therapy to individual patient’s needs and risk profile. Accurate
NCT04338919, NCT05262803) and genotype-guided strategies in pa­ risk stratification for both bleeding and thrombosis based on multiple
tients undergoing PCI (NCT03783351). factors can aid in selecting the appropriate antiplatelet therapy and pre­
There are a number of unmet needs that may be stimulating for fu­ vent serious and life-threatening outcomes. With the introduction of
ture studies. In fact, de-escalation strategies have used DAPT as a com­ PFT and rapid genotype characterization, personalised antiplatelet ther­
parator and there is limited data comparing the different approaches. apy is becoming more feasible and accessible. The comprehensive un­
For example, there are no direct comparisons of de-escalation strat­ derstanding of personalised antiplatelet therapy presented in this
egies involving aspirin withdrawal (i.e. P2Y12 inhibitor monotherapy) review can help clinicians make informed clinical decisions and tailor
vs. switch guided by PFT or genotype testing. Additionally, most of cardiovascular disease management.
the trials and evidence cited in this review investigated clinical outcomes
over a relatively short time interval (i.e. 1 to 3 years). While strategies
for long-term secondary prevention are also evolving rapidly, it is un­
Declarations
clear which patients may be ideally suited for long-term DAPT vs. Disclosure of Interest
dual pathway inhibition the low-dose direct oral anticoagulants (i.e. riv­
D.C. reports speaker’s or consulting fees from Amgen, Arena,
aroxaban) vs. P2Y12 inhibitor monotherapy.
Chiesi, Daiichi Sankyo, Sanofi, Terumo, and institutional fees from
Medtronic. D.J.A. declares that he has received consulting fees or hon­
Guidelines oraria from Abbott, Amgen, AstraZeneca, Bayer, Biosensors,
The ESC guidelines for myocardial revascularization97 and those for Boehringer Ingelheim, Bristol-Myers Squibb, Chiesi, Daiichi-Sankyo, Eli
ACS22 include a low-grade recommendation (i.e. class IIb) for de- Lilly, Haemonetics, Janssen, Merck, Novartis, PhaseBio, PLx Pharma,
3070 D. Capodanno and D. J. Angiolillo

Pfizer, Sanofi, and Vectura, outside the present work; D.J.A. also de­ 14. Yeh RW, Secemsky EA, Kereiakes DJ, Normand SL, Gershlick AH, Cohen DJ, et al.
Development and validation of a prediction rule for benefit and harm of dual antiplatelet
clares that his institution has received research grants from Amgen,
therapy beyond 1 year after percutaneous coronary intervention. JAMA 2016;315:
AstraZeneca, Bayer, Biosensors, CeloNova, CSL Behring, 1735–1749. https://doi.org/10.1001/jama.2016.3775
Daiichi-Sankyo, Eisai, Eli Lilly, Gilead, Janssen, Matsutani Chemical 15. Urban P, Gregson J, Owen R, Mehran R, Windecker S, Valgimigli M, et al. Assessing the
Industry Co., Merck, Novartis, Osprey Medical, Renal Guard 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 Cardiol 2021;6:
Solutions, and Scott R. MacKenzie Foundation.
410–419. https://doi.org/10.1001/jamacardio.2020.6814
16. Galli M, Franchi F, Rollini F, Angiolillo DJ. Role of platelet function and genetic testing in
Data Availability patients undergoing percutaneous coronary intervention. Trends Cardiovasc Med 2023;
33:13–138. https://doi.org/10.1016/j.tcm.2021.12.007
No new data were generated or analysed for this manuscript. 17. Capodanno D, Baber U, Bhatt DL, Collet JP, Dangas G, Franchi F, et al. P2Y(12) inhibitor
monotherapy in patients undergoing percutaneous coronary intervention. Nat Rev
Cardiol 2022;19:829–844. https://doi.org/10.1038/s41569-022-00725-6
Funding 18. Capodanno D, Bhatt DL, Eikelboom JW, Fox KAA, Geisler T, Michael Gibson C, et al.
The research leading to this review has received funding from the Dual-pathway inhibition for secondary and tertiary antithrombotic prevention in car­

Downloaded from https://academic.oup.com/eurheartj/article/44/32/3059/7204812 by guest on 25 August 2023


European Union—NextGenerationEU through the Italian Ministry of diovascular disease. Nat Rev Cardiol 2020;17:242–257. https://doi.org/10.1038/
s41569-019-0314-y
University and Research under PNRR—M4C2-I1.3 Project
19. Angiolillo DJ, Bhatt DL, Cannon CP, Eikelboom JW, Gibson CM, Goodman SG, et al.
PE_00000019 ‘HEAL ITALIA’ to Davide Capodanno, CUP Antithrombotic therapy in patients with atrial fibrillation treated with oral anticoagula­
E63C22002080006 of University of Catania. The views and opinions tion undergoing percutaneous coronary intervention: a North American perspective:
expressed are those of the authors only and do not necessarily reflect 2021 update. Circulation 2021;143:583–596. https://doi.org/10.1161/circulationaha.
120.050438
those of the European Union or the European Commission. Neither
20. Lip GYH, Collet JP, Haude M, Byrne R, Chung EH, Fauchier L, et al. 2018 Joint European
the European Union nor the European Commission can be held re­ consensus document on the management of antithrombotic therapy in atrial fibrillation
sponsible for them. patients presenting with acute coronary syndrome and/or undergoing percutaneous
cardiovascular interventions: a joint consensus document of the European Heart
Rhythm Association (EHRA), European Society of Cardiology Working Group on
References Thrombosis, European Association of Percutaneous Cardiovascular Interventions
1. Cao D, Chandiramani R, Chiarito M, Claessen BE, Mehran R. Evolution of antithrombo­ (EAPCI), and European Association of Acute Cardiac Care (ACCA) endorsed by the
tic therapy in patients undergoing percutaneous coronary intervention: a 40-year jour­ Heart Rhythm Society (HRS), Asia-Pacific Heart Rhythm Society (APHRS), Latin
ney. Eur Heart J 2021;42:339–351. https://doi.org/10.1093/eurheartj/ehaa824 America Heart Rhythm Society (LAHRS), and Cardiac Arrhythmia Society of
2. Tsao CW, Aday AW, Almarzooq ZI, Alonso A, Beaton AZ, Bittencourt MS, et al. Heart Southern Africa (CASSA). Europace 2019;21:192–193. https://doi.org/10.1093/
disease and stroke statistics—2022 update: a report from the American Heart europace/euy174
Association. Circulation 2022;145:e153–e639. https://doi.org/10.1161/cir. 21. Capodanno D, Angiolillo DJ. Tailoring duration of DAPT with risk scores. Lancet 2017;
0000000000001052 389:987–989. https://doi.org/10.1016/s0140-6736(17)30591-3
3. Visseren FLJ, Mach F, Smulders YM, Carballo D, Koskinas KC, Bäck M, et al. 2021 ESC 22. Collet JP, Thiele H, Barbato E, Barthélémy O, Bauersachs J, Bhatt DL, et al. 2020 ESC
guidelines on cardiovascular disease prevention in clinical practice. Eur Heart J 2021;42: guidelines for the management of acute coronary syndromes in patients presenting
3227–3337. https://doi.org/10.1093/eurheartj/ehab484 without persistent ST-segment elevation. Eur Heart J 2021;42:1289–1367. https://doi.
4. Lawton JS, Tamis-Holland JE, Bangalore S, Bates ER, Beckie TM, Bischoff JM, et al. 2021 org/10.1093/eurheartj/ehaa575
ACC/AHA/SCAI guideline for coronary artery revascularization: executive summary: a 23. Mohamed MO, Kinnaird T, Anderson R, Rashid M, Martin GP, Freeman P, et al.
report of the American College of Cardiology/American Heart Association Joint Combinations of bleeding and ischemic risk and their association with clinical outcomes
Committee on Clinical Practice Guidelines. Circulation 2022;145:e4–e17. https://doi. in acute coronary syndrome. Int J Cardiol 2019;290:7–14. https://doi.org/10.1016/j.
org/10.1161/cir.0000000000001039
ijcard.2019.05.035
5. Nathan AS, Sen S, Yeh RW. The risk of bleeding with the use of antiplatelet agents for
24. Capodanno D, Alfonso F, Levine GN, Valgimigli M, Angiolillo DJ. ACC/AHA versus ESC
the treatment of cardiovascular disease. Expert Opin Drug Saf 2017;16:561–572. https://
guidelines on dual antiplatelet therapy: JACC Guideline comparison. J Am Coll Cardiol
doi.org/10.1080/14740338.2017.1315101
2018;72:2915–2931. https://doi.org/10.1016/j.jacc.2018.09.057
6. Capodanno D, Bhatt DL, Gibson CM, James S, Kimura T, Mehran R, et al. Bleeding avoid­
25. Angiolillo DJ, Galli M, Collet JP, Kastrati A, O’Donoghue ML. Antiplatelet therapy after
ance strategies in percutaneous coronary intervention. Nat Rev Cardiol 2022;19:
percutaneous coronary intervention. EuroIntervention 2022;17:e1371–e1396. https://
117–132. https://doi.org/10.1038/s41569-021-00598-1
doi.org/10.4244/eij-d-21-00904
7. Capodanno D, Morice MC, Angiolillo DJ, Bhatt DL, Byrne RA, Colleran R, et al. Trial
26. Knuuti J, Wijns W, Saraste A, Capodanno D, Barbato E, Funck-Brentano C, et al. 2019
design principles for patients at high bleeding risk undergoing PCI: JACC scientific ex­
ESC guidelines for the diagnosis and management of chronic coronary syndromes. Eur
pert panel. J Am Coll Cardiol 2020;76:1468–1483. https://doi.org/10.1016/j.jacc.2020.
Heart J 2020;41:407–477. https://doi.org/10.1093/eurheartj/ehz425
06.085
27. Lawton JS, Tamis-Holland JE, Bangalore S, Bates ER, Beckie TM, Bischoff JM, et al. 2021
8. Galli M, Ortega-Paz L, Franchi F, Rollini F, Angiolillo DJ. Precision medicine in interven­
ACC/AHA/SCAI guideline for coronary artery revascularization: a report of the
tional cardiology: implications for antiplatelet therapy in patients undergoing percutan­
eous coronary intervention. Pharmacogenomics 2022;23:723–737. https://doi.org/10. American College of Cardiology/American Heart Association Joint Committee on clin­
2217/pgs-2022-0057 ical practice guidelines. Circulation 2022;145:e18–e114. https://doi.org/10.1161/cir.
9. Duarte JD, Cavallari LH. Pharmacogenetics to guide cardiovascular drug therapy. Nat 0000000000001038
Rev Cardiol 2021;18:649–665. https://doi.org/10.1038/s41569-021-00549-w 28. Schüpke S, Neumann FJ, Menichelli M, Mayer K, Bernlochner I, Wöhrle J, et al. Ticagrelor
10. Baber U, Mehran R, Giustino G, Cohen DJ, Henry TD, Sartori S, et al. Coronary throm­ or prasugrel in patients with acute coronary syndromes. N Engl J Med 2019;381:
bosis and major bleeding after PCI with drug-eluting stents: risk scores from PARIS. J Am 1524–1534. https://doi.org/10.1056/NEJMoa1908973
Coll Cardiol 2016;67:2224–2234. https://doi.org/10.1016/j.jacc.2016.02.064 29. Costa F, Van Klaveren D, Feres F, James S, Räber L, Pilgrim T, et al. Dual antiplatelet ther­
11. Costa F, van Klaveren D, James S, Heg D, Räber L, Feres F, et al. Derivation and validation apy duration based on ischemic and bleeding risks after coronary stenting. J Am Coll
of the predicting bleeding complications in patients undergoing stent implantation and Cardiol 2019;73:741–754. https://doi.org/10.1016/j.jacc.2018.11.048
subsequent dual antiplatelet therapy (PRECISE-DAPT) score: a pooled analysis of 30. Koo BK, Kang J, Park KW, Rhee TM, Yang HM, Won KB, et al. Aspirin versus clopidogrel
individual-patient datasets from clinical trials. Lancet 2017;389:1025–1034. https://doi. for chronic maintenance monotherapy after percutaneous coronary intervention
org/10.1016/s0140-6736(17)30397-5 (HOST-EXAM): an investigator-initiated, prospective, randomised, open-label, multi­
12. Raposeiras-Roubín S, Faxén J, Íñiguez-Romo A, Henriques JPS, D’Ascenzo F, Saucedo J, centre trial. Lancet 2021;397:2487–2496. https://doi.org/10.1016/s0140-6736(21)
et al. Development and external validation of a post-discharge bleeding risk score in pa­ 01063-1
tients with acute coronary syndrome: the BleeMACS score. Int J Cardiol 2018;254: 31. Capodanno D, Angiolillo DJ. Long-term P2Y(12) inhibitor or aspirin as single antiplatelet
10–15. https://doi.org/10.1016/j.ijcard.2017.10.103 therapy in patients with previous percutaneous coronary intervention. Circulation 2023;
13. Urban P, Mehran R, Colleran R, Angiolillo DJ, Byrne RA, Capodanno D, et al. Defining 147:118–121. https://doi.org/10.1161/circulationaha.122.063004
high bleeding risk in patients undergoing percutaneous coronary intervention: a consen­ 32. Capodanno D, Angiolillo DJ. Timing, selection, modulation, and duration of P2Y(12) in­
sus document from the Academic Research Consortium for High Bleeding Risk. Eur hibitors for patients with acute coronary syndromes undergoing PCI. JACC Cardiovasc
Heart J 2019;40:2632–2653. https://doi.org/10.1093/eurheartj/ehz372 Interv 2023;16:1–18. https://doi.org/10.1016/j.jcin.2022.10.023
Personalised antiplatelet therapies for CAD 3071

33. Capodanno D, Mehran R, Krucoff M, Baber U, Bhatt DL, Capranzano P, et al. Defining 51. Steg PG, Bhatt DL, Simon T, Fox K, Mehta SR, Harrington RA, et al. Ticagrelor in pa­
strategies of modulation of antiplatelet therapy in patients with coronary artery disease: tients with stable coronary disease and diabetes. N Engl J Med 2019;381:1309–1320.
a consensus document from the Academic Research Consortium. Circulation 2023. In https://doi.org/10.1056/NEJMoa1908077
press. 52. Angiolillo DJ, Rollini F, Storey RF, Bhatt DL, James S, Schneider DJ, et al. International
34. Cuisset T, Deharo P, Quilici J, Johnson TW, Deffarges S, Bassez C, et al. Benefit of expert consensus on switching platelet P2Y(12) receptor-inhibiting therapies.
switching dual antiplatelet therapy after acute coronary syndrome: the TOPIC (timing Circulation 2017;136:1955–1975. https://doi.org/10.1161/circulationaha.117.031164
of platelet inhibition after acute coronary syndrome) randomized study. Eur Heart J 53. Wiviott SD, Braunwald E, McCabe CH, Montalescot G, Ruzyllo W, Gottlieb S, et al.
2017;38:3070–3078. https://doi.org/10.1093/eurheartj/ehx175 Prasugrel versus clopidogrel in patients with acute coronary syndromes. N Engl J Med
35. Kim CJ, Park MW, Kim MC, Choo EH, Hwang BH, Lee KY, et al. Unguided de-escalation 2007;357:2001–2015. https://doi.org/10.1056/NEJMoa0706482
from ticagrelor to clopidogrel in stabilised patients with acute myocardial infarction 54. Wallentin L, Becker RC, Budaj A, Cannon CP, Emanuelsson H, Held C, et al. Ticagrelor
undergoing percutaneous coronary intervention (TALOS-AMI): an investigator- versus clopidogrel in patients with acute coronary syndromes. N Engl J Med 2009;361:
initiated, open-label, multicentre, non-inferiority, randomised trial. Lancet 2021;398: 1045–1057. https://doi.org/10.1056/NEJMoa0904327
1305–1316. https://doi.org/10.1016/s0140-6736(21)01445-8 55. Kandzari DE, Kirtane AJ, Windecker S, Latib A, Kedhi E, Mehran R, et al. One-month
36. Kim HS, Kang J, Hwang D, Han JK, Yang HM, Kang HJ, et al. Prasugrel-based de- dual antiplatelet therapy following percutaneous coronary intervention with
zotarolimus-eluting stents in high-bleeding-risk patients. Circ Cardiovasc Interv 2020;
escalation of dual antiplatelet therapy after percutaneous coronary intervention in pa­
13:e009565. https://doi.org/10.1161/circinterventions.120.009565
tients with acute coronary syndrome (HOST-REDUCE-POLYTECH-ACS): an open-

Downloaded from https://academic.oup.com/eurheartj/article/44/32/3059/7204812 by guest on 25 August 2023


56. Kirtane AJ, Stoler R, Feldman R, Neumann FJ, Boutis L, Tahirkheli N, et al. Primary results
label, multicentre, non-inferiority randomised trial. Lancet 2020;396:1079–1089.
of the EVOLVE short DAPT study: evaluation of 3-month dual antiplatelet therapy in
https://doi.org/10.1016/s0140-6736(20)31791-8
high bleeding risk patients treated with a bioabsorbable polymer-coated
37. Hong SJ, Kim JS, Hong SJ, Lim DS, Lee SY, Yun KH, et al. 1-month dual-antiplatelet ther­
everolimus-eluting stent. Circ Cardiovasc Interv 2021;14:e010144. https://doi.org/10.
apy followed by aspirin monotherapy after polymer-free drug-coated stent implant­
1161/circinterventions.120.010144
ation: one-month DAPT trial. JACC Cardiovasc Interv 2021;14:1801–1811. https://doi.
57. Capodanno D. Evolving landscapes in coronary stents for patients at high bleeding risk. Circ
org/10.1016/j.jcin.2021.06.003
Cardiovasc Interv 2021;14:e010591. https://doi.org/10.1161/circinterventions.121.010591
38. Hahn JY, Song YB, Oh JH, Cho DK, Lee JB, Doh JH, et al. 6-month versus 12-month or 58. Lee YJ, Cho JY, Yun KH, Lee SJ, Hong SJ, Ahn CM, et al. Impact of one-month DAPT fol­
longer dual antiplatelet therapy after percutaneous coronary intervention in patients lowed by aspirin monotherapy in patients undergoing percutaneous coronary intervention
with acute coronary syndrome (SMART-DATE): a randomised, open-label, non- according to clinical presentation: a post hoc analysis of the randomised one-month DAPT
inferiority trial. Lancet 2018;391:1274–1284. https://doi.org/10.1016/s0140-6736(18) trial. EuroIntervention 2022;18:471–481. https://doi.org/10.4244/eij-d-22-00135
30493-8 59. Tomaniak M, Chichareon P, Onuma Y, Deliargyris EN, Takahashi K, Kogame N, et al.
39. De Luca G, Damen SA, Camaro C, Benit E, Verdoia M, Rasoul S, et al. Final results of the Benefit and risks of aspirin in addition to ticagrelor in acute coronary syndromes: a
randomised evaluation of short-term dual antiplatelet therapy in patients with acute post hoc analysis of the randomized GLOBAL LEADERS trial. JAMA Cardiol 2019;4:
coronary syndrome treated with a new-generation stent (REDUCE trial). 1092–1101. https://doi.org/10.1001/jamacardio.2019.3355
EuroIntervention 2019;15:e990–e998. https://doi.org/10.4244/eij-d-19-00539 60. Baber U, Dangas G, Angiolillo DJ, Cohen DJ, Sharma SK, Nicolas J, et al. Ticagrelor alone
40. Kedhi E, Fabris E, van der Ent M, Buszman P, von Birgelen C, Roolvink V, et al. Six months vs. ticagrelor plus aspirin following percutaneous coronary intervention in patients with
versus 12 months dual antiplatelet therapy after drug-eluting stent implantation in non-ST-segment elevation acute coronary syndromes: TWILIGHT-ACS. Eur Heart J
ST-elevation myocardial infarction (DAPT-STEMI): randomised, multicentre, non- 2020;41:3533–3545. https://doi.org/10.1093/eurheartj/ehaa670
inferiority trial. BMJ 2018;363:k3793. https://doi.org/10.1136/bmj.k3793 61. Valgimigli M, Frigoli E, Heg D, Tijssen J, Jüni P, Vranckx P, et al. Dual antiplatelet therapy
41. Vranckx P, Valgimigli M, Jüni P, Hamm C, Steg PG, Heg D, et al. Ticagrelor plus aspirin for after PCI in patients at high bleeding risk. N Engl J Med 2021;385:1643–1655. https://doi.
1 month, followed by ticagrelor monotherapy for 23 months vs aspirin plus clopidogrel org/10.1056/NEJMoa2108749
or ticagrelor for 12 months, followed by aspirin monotherapy for 12 months after im­ 62. Mehilli J, Baquet M, Hochholzer W, Mayer K, Tesche C, Aradi D, et al. Randomized com­
plantation of a drug-eluting stent: a multicentre, open-label, randomised superiority parison of intensified and standard P2Y(12)-receptor-inhibition before elective percu­
trial. Lancet 2018;392:940–949. https://doi.org/10.1016/s0140-6736(18)31858-0 taneous coronary intervention: the SASSICAIA trial. Circ Cardiovasc Interv 2020;13:
42. Mehran R, Baber U, Sharma SK, Cohen DJ, Angiolillo DJ, Briguori C, et al. Ticagrelor with e008649. https://doi.org/10.1161/circinterventions.119.008649
or without aspirin in high-risk patients after PCI. N Engl J Med 2019;381:2032–2042. 63. Silvain J, Lattuca B, Beygui F, Rangé G, Motovska Z, Dillinger JG, et al. Ticagrelor versus
https://doi.org/10.1056/NEJMoa1908419 clopidogrel in elective percutaneous coronary intervention (ALPHEUS): a randomised,
43. Watanabe H, Domei T, Morimoto T, Natsuaki M, Shiomi H, Toyota T, et al. Effect of open-label, phase 3b trial. Lancet 2020;396:1737–1744. https://doi.org/10.1016/s0140-
1-month dual antiplatelet therapy followed by clopidogrel vs 12-month dual antiplatelet 6736(20)32236-4
therapy on cardiovascular and bleeding events in patients receiving PCI: the 64. Mehta SR, Bassand JP, Chrolavicius S, Diaz R, Eikelboom JW, Fox KA, et al. Dose com­
STOPDAPT-2 randomized clinical trial. JAMA 2019;321:2414–2427. https://doi.org/ parisons of clopidogrel and aspirin in acute coronary syndromes. N Engl J Med 2010;
10.1001/jama.2019.8145 363:930–942. https://doi.org/10.1056/NEJMoa0909475
44. Hahn JY, Song YB, Oh JH, Chun WJ, Park YH, Jang WJ, et al. Effect of P2Y12 inhibitor 65. Jones WS, Mulder H, Wruck LM, Pencina MJ, Kripalani S, Muñoz D, et al. Comparative
monotherapy vs dual antiplatelet therapy on cardiovascular events in patients undergo­ effectiveness of aspirin dosing in cardiovascular disease. N Engl J Med 2021;384:
1981–1990. https://doi.org/10.1056/NEJMoa2102137
ing percutaneous coronary intervention: the SMART-CHOICE randomized clinical trial.
66. Bhatt DL, Steg PG, Mehta SR, Leiter LA, Simon T, Fox K, et al. Ticagrelor in patients with
JAMA 2019;321:2428–2437. https://doi.org/10.1001/jama.2019.8146
diabetes and stable coronary artery disease with a history of previous percutaneous
45. Kim BK, Hong SJ, Cho YH, Yun KH, Kim YH, Suh Y, et al. Effect of ticagrelor monother­
coronary intervention (THEMIS-PCI): a phase 3, placebo-controlled, randomised trial.
apy vs ticagrelor with aspirin on Major bleeding and cardiovascular events in patients
Lancet 2019;394:1169–1180. https://doi.org/10.1016/s0140-6736(19)31887-2
with acute coronary syndrome: the TICO randomized clinical trial. JAMA 2020;323:
67. Angiolillo DJ. Variability in responsiveness to oral antiplatelet therapy. Am J Cardiol 2009;
2407–2416. https://doi.org/10.1001/jama.2020.7580
103:27a–34a. https://doi.org/10.1016/j.amjcard.2008.11.020
46. Watanabe H, Morimoto T, Natsuaki M, Yamamoto K, Obayashi Y, Ogita M, et al.
68. Sibbing D, Aradi D, Alexopoulos D, Ten Berg J, Bhatt DL, Bonello L, et al. Updated ex­
Comparison of clopidogrel monotherapy after 1 to 2 months of dual antiplatelet ther­
pert consensus statement on platelet function and genetic testing for guiding P2Y(12)
apy with 12 months of dual antiplatelet therapy in patients with acute coronary syn­
receptor inhibitor treatment in percutaneous coronary intervention. JACC Cardiovasc
drome: the STOPDAPT-2 ACS randomized clinical trial. JAMA Cardiol 2022;7: Interv 2019;12:1521–1537. https://doi.org/10.1016/j.jcin.2019.03.034
407–417. https://doi.org/10.1001/jamacardio.2021.5244 69. Pereira NL, Rihal CS, So DYF, Rosenberg Y, Lennon RJ, Mathew V, et al. Clopidogrel
47. Valgimigli M, Cao D, Angiolillo DJ, Bangalore S, Bhatt DL, Ge J, et al. Duration of dual pharmacogenetics. Circ Cardiovasc Interv 2019;12:e007811. https://doi.org/10.1161/
antiplatelet therapy for patients at high bleeding risk undergoing PCI. J Am Coll Cardiol circinterventions.119.007811
2021;78:2060–2072. https://doi.org/10.1016/j.jacc.2021.08.074 70. Mega JL, Close SL, Wiviott SD, Shen L, Hockett RD, Brandt JT, et al. Cytochrome p-450
48. Han JK, Hwang D, Yang S, Park SH, Kang J, Yang HM, et al. Comparison of 3–6 month polymorphisms and response to clopidogrel. N Engl J Med 2009;360:354–362. https://
versus 12 month dual antiplatelet therapy after coronary intervention using the con­ doi.org/10.1056/NEJMoa0809171
temporary drug-eluting stents with ultrathin struts: the HOST-IDEA randomized clin­ 71. Mega JL, Simon T, Collet JP, Anderson JL, Antman EM, Bliden K, et al. Reduced-function
ical trial. Circulation 2023. https://doi.org/10.1161/circulationaha.123.064264 CYP2C19 genotype and risk of adverse clinical outcomes among patients treated with
49. Mauri L, Kereiakes DJ, Yeh RW, Driscoll-Shempp P, Cutlip DE, Steg PG, et al. Twelve or clopidogrel predominantly for PCI: a meta-analysis. JAMA 2010;304:1821–1830. https://
30 months of dual antiplatelet therapy after drug-eluting stents. N Engl J Med 2014;371: doi.org/10.1001/jama.2010.1543
2155–2166. https://doi.org/10.1056/NEJMoa1409312 72. Scott SA, Sangkuhl K, Gardner EE, Stein CM, Hulot JS, Johnson JA, et al. Clinical pharma­
50. Bonaca MP, Bhatt DL, Cohen M, Steg PG, Storey RF, Jensen EC, et al. Long-term use of cogenetics implementation consortium guidelines for cytochrome P450–2C19
ticagrelor in patients with prior myocardial infarction. N Engl J Med 2015;372: (CYP2C19) genotype and clopidogrel therapy. Clin Pharmacol Ther 2011;90:328–332.
1791–1800. https://doi.org/10.1056/NEJMoa1500857 https://doi.org/10.1038/clpt.2011.132
3072 D. Capodanno and D. J. Angiolillo

73. Kim HK, Tantry US, Smith SC Jr., Jeong MH, Park SJ, Kim MH, et al. The East Asian para­ 85. Trenk D, Stone GW, Gawaz M, Kastrati A, Angiolillo DJ, Müller U, et al. A randomized
dox: an updated position statement on the challenges to the current antithrombotic trial of prasugrel versus clopidogrel in patients with high platelet reactivity on clopido­
strategy in patients with cardiovascular disease. Thromb Haemost 2021;121:422–432. grel after elective percutaneous coronary intervention with implantation of drug-eluting
https://doi.org/10.1055/s-0040-1718729 stents: results of the TRIGGER-PCI (testing platelet reactivity in patients undergoing
74. Collet JP, Cuisset T, Rangé G, Cayla G, Elhadad S, Pouillot C, et al. Bedside monitoring to elective stent placement on clopidogrel to guide alternative therapy with prasugrel)
adjust antiplatelet therapy for coronary stenting. N Engl J Med 2012;367:2100–2109. study. J Am Coll Cardiol 2012;59:2159–2164. https://doi.org/10.1016/j.jacc.2012.02.026
https://doi.org/10.1056/NEJMoa1209979 86. Price MJ, Berger PB, Teirstein PS, Tanguay JF, Angiolillo DJ, Spriggs D, et al. Standard- vs
75. Cayla G, Cuisset T, Silvain J, Leclercq F, Manzo-Silberman S, Saint-Etienne C, et al. high-dose clopidogrel based on platelet function testing after percutaneous coronary
Platelet function monitoring to adjust antiplatelet therapy in elderly patients stented intervention: the GRAVITAS randomized trial. JAMA 2011;305:1097–1105. https://
for an acute coronary syndrome (ANTARCTIC): an open-label, blinded-endpoint, ran­ doi.org/10.1001/jama.2011.290
domised controlled superiority trial. Lancet 2016;388:2015–2022. https://doi.org/10. 87. Franchi F, Rollini F, Rivas J, Rivas A, Agarwal M, Briceno M, et al. Prasugrel versus tica­
1016/s0140-6736(16)31323-x grelor in patients with CYP2C19 loss-of-function genotypes: results of a randomized
76. Sibbing D, Aradi D, Jacobshagen C, Gross L, Trenk D, Geisler T, et al. Guided de-escalation pharmacodynamic study in a feasibility investigation of rapid genetic testing. JACC
of antiplatelet treatment in patients with acute coronary syndrome undergoing percutan­ Basic Transl Sci 2020;5:419–428. https://doi.org/10.1016/j.jacbts.2020.02.009
eous coronary intervention (TROPICAL-ACS): a randomised, open-label, multicentre trial. 88. Cavallari LH, Franchi F, Rollini F, Been L, Rivas A, Agarwal M, et al. Clinical implementa­
Lancet 2017;390:1747–1757. https://doi.org/10.1016/s0140-6736(17)32155-4 tion of rapid CYP2C19 genotyping to guide antiplatelet therapy after percutaneous cor­

Downloaded from https://academic.oup.com/eurheartj/article/44/32/3059/7204812 by guest on 25 August 2023


77. Zheng YY, Wu TT, Yang Y, Hou XG, Gao Y, Chen Y, et al. Personalized antiplatelet onary intervention. J Transl Med 2018;16:92. https://doi.org/10.1186/s12967-018-1469-8
therapy guided by a novel detection of platelet aggregation function in stable coronary 89. Ingraham B, Farkoud M RL. Genetic-guided oral P2Y12 inhibitor selection and cumula­
artery disease patients undergoing percutaneous coronary intervention: a randomized tive ischemic events after percutaneous coronary intervention. JACC Cardiovasc Interv
controlled clinical trial. Eur Heart J Cardiovasc Pharmacother 2020;6:211–221. https://doi. 2023;16:816–825. https://doi.org/10.1016/j.jcin.2023.01.356
org/10.1093/ehjcvp/pvz059 90. Angiolillo DJ. Dual antiplatelet therapy guided by platelet function testing. Lancet 2017;
78. Notarangelo FM, Maglietta G, Bevilacqua P, Cereda M, Merlini PA, Villani GQ, et al. 390:1718–1720. https://doi.org/10.1016/s0140-6736(17)32279-1
Pharmacogenomic approach to selecting antiplatelet therapy in patients with acute cor­ 91. Galli M, Franchi F, Rollini F, Cavallari LH, Capodanno D, Crea F, et al. Genetic testing in
onary syndromes: the PHARMCLO trial. J Am Coll Cardiol 2018;71:1869–1877. https:// patients undergoing percutaneous coronary intervention: rationale, evidence and prac­
doi.org/10.1016/j.jacc.2018.02.029 tical recommendations. Expert Rev Clin Pharmacol 2021;14:963–978. https://doi.org/10.
79. Claassens DMF, Vos GJA, Bergmeijer TO, Hermanides RS, van ‘t Hof AWJ, van der 1080/17512433.2021.1927709
Harst P, et al. A genotype-guided strategy for oral P2Y(12) inhibitors in primary PCI. 92. Angiolillo DJ, Capodanno D, Danchin N, Simon T, Bergmeijer TO, Ten Berg JM, et al.
N Engl J Med 2019;381:1621–1631. https://doi.org/10.1056/NEJMoa1907096 Derivation, validation, and prognostic utility of a prediction rule for nonresponse to clo­
80. Tuteja S, Glick H, Matthai W, Nachamkin I, Nathan A, Monono K, et al. Prospective pidogrel: the ABCD-GENE score. JACC Cardiovasc Interv 2020;13:606–617. https://doi.
CYP2C19 genotyping to guide antiplatelet therapy following percutaneous coronary org/10.1016/j.jcin.2020.01.226
intervention: a pragmatic randomized clinical trial. Circ Genom Precis Med 2020;13: 93. Thomas CD, Franchi F, Keeley EC, Rossi JS, Winget M, David Anderson R, et al. Impact
e002640. https://doi.org/10.1161/circgen.119.002640 of the ABCD-GENE score on clopidogrel clinical effectiveness after PCI: a multi-site,
81. Pereira NL, Farkouh ME, So D, Lennon R, Geller N, Mathew V, et al. Effect of genotype- real-world investigation. Clin Pharmacol Ther 2022;112:146–155. https://doi.org/10.
guided oral P2Y12 inhibitor selection vs conventional clopidogrel therapy on ischemic 1002/cpt.2612
outcomes after percutaneous coronary intervention: the TAILOR-PCI randomized clin­ 94. Capodanno D, Angiolillo DJ, Lennon RJ, Goodman SG, Kim SW, O’Cochlain F, et al.
ical trial. JAMA 2020;324:761–771. https://doi.org/10.1001/jama.2020.12443 ABCD-GENE score and clinical outcomes following percutaneous coronary interven­
82. Al-Rubaish AM, Al-Muhanna FA, Alshehri AM, Al-Mansori MA, Alali RA, Khalil RM, et al. tion: insights from the TAILOR-PCI trial. J Am Heart Assoc 2022;11:e024156. https://
Bedside testing of CYP2C19 vs. conventional clopidogrel treatment to guide antiplatelet doi.org/10.1161/jaha.121.024156
therapy in ST-segment elevation myocardial infarction patients. Int J Cardiol 2021;343: 95. Angiolillo DJ, Prats J, Deliargyris EN, Schneider DJ, Scheiman J, Kimmelstiel C, et al.
15–20. https://doi.org/10.1016/j.ijcard.2021.08.051 Pharmacokinetic and pharmacodynamic profile of a novel phospholipid aspirin formu­
83. Galli M, Benenati S, Capodanno D, Franchi F, Rollini F, D’Amario D, et al. Guided versus lation. Clin Pharmacokinet 2022;61:465–479. https://doi.org/10.1007/s40262-021-
standard antiplatelet therapy in patients undergoing percutaneous coronary interven­ 01090-2
tion: a systematic review and meta-analysis. Lancet 2021;397:1470–1483. https://doi. 96. Jourdi G, Lordkipanidzé M, Philippe A, Bachelot-Loza C, Gaussem P. Current and novel
org/10.1016/s0140-6736(21)00533-x antiplatelet therapies for the treatment of cardiovascular diseases. Int J Mol Sci 2021;22:
84. Galli M, Benenati S, Franchi F, Rollini F, Capodanno D, Biondi-Zoccai G, et al. 13079. https://doi.org/10.3390/ijms222313079
Comparative effects of guided vs. potent P2Y12 inhibitor therapy in acute coronary syn­ 97. Neumann FJ, Sousa-Uva M, Ahlsson A, Alfonso F, Banning AP, Benedetto U, et al. 2018
drome: a network meta-analysis of 61 898 patients from 15 randomized trials. Eur Heart ESC/EACTS guidelines on myocardial revascularization. EuroIntervention 2019;14:
J 2022;43:959–967. https://doi.org/10.1093/eurheartj/ehab836 1435–1534. https://doi.org/10.4244/eijy19m01_01

You might also like