ArticlePDF AvailableLiterature Review

De-escalation of antiplatelet therapy in patients with coronary artery disease: Time to change our strategy?

Authors:

Abstract and Figures

Dual antiplatelet therapy (DAPT) is the gold standard after acute coronary syndromes (ACS) or chronic coronary syndromes (CCS) undergoing percutaneous coronary intervention (PCI). Because local and systemic ischemic complications can occur particularly in the early phase (i.e. 1–3 months) after ACS or PCI, the synergistic platelet inhibition of aspirin and a P2Y12 inhibitor is of the utmost importance in this early phase. Moreover, the use of the more potent P2Y12 inhibitors prasugrel and ticagrelor have shown to further reduce the incidence of ischemic events compared to clopidogrel after an ACS. On the other hand, prolonged and potent antiplatelet therapy are inevitably associated with increased bleeding, which unlike thrombotic risk, tends to be stable over time and may outweigh the benefit of reducing ischemic events in these patients. The duration and composition of antiplatelet therapy remains a topic of debate in cardiology due to competing ischemic and bleeding risks, with guidelines and recommendations considerably evolving in the past years. An emerging strategy, called “de-escalation”, consisting in the administration of a less intense antithrombotic therapy after a short course of standard DAPT, has shown to reduce bleeding without any trade-off in ischemic events. De-escalation may be achieved with different antithrombotic strategies and can be either unguided or guided by platelet function or genetic testing. The aim of this review is to summarize the evidence and provide practical recommendations on the use of different de-escalation strategies in patients with ACS and CCS.
Content may be subject to copyright.
European Journal of Internal Medicine xxx (xxxx) xxx
Please cite this article as: Pierre Sabouret, European Journal of Internal Medicine, https://doi.org/10.1016/j.ejim.2022.12.008
0953-6205/© 2022 The Authors. Published by Elsevier B.V. on behalf of European Federation of Internal Medicine. This is an open access article under the CC
BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
Review Article
De-escalation of antiplatelet therapy in patients with coronary artery
disease: Time to change our strategy?
Pierre Sabouret
a
,
b
,
1
,
*
, Luigi Spadafora
c
,
1
, David Fischman
d
, Waqas Ullah
d
, Michel Zeitouni
a
,
Martha Gulati
e
, Salvatore De Rosa
f
, Michael P. Savage
d
, Juan Pablo Costabel
g
, Maciej Banach
h
,
Giuseppe Biondi-Zoccai
i
,
j
, Mattia Galli
k
,
l
a
Heart Institute, ACTION Study Group-CHU Piti´
e-Salp´
etri`
ere, 47-83 Boulevard de lHˆ
opital, Paris, France
b
Coll`
ege National des Cardiologues Français (CNCF), Paris, France
c
Department of Clinical, Internal Medicine, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Italy
d
Thomas Jefferson University Hospitals, Philadelphia, PA, USA
e
Barbra Streisand Womens Heart Center, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, USA
f
Magna Graecia University of Catanzaro, Catanzaro, Italy
g
Division of Cardiology, Instituto Cardiovascular de Buenos Aires (ICBA), Buenos Aires, Argentina
h
Department of Preventive Cardiology and Lipidology, Medical University of Lodz and Polish Mothers Memorial Hospital Research Institute, Lodz, Poland
i
Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
j
Mediterranea Cardiocentro, Napoli, Italy
k
Catholic University of the Sacred Heart, Rome, Italy
l
Maria Cecilia Hospital, GVM Care & Research, Cotignola, Italy
ARTICLE INFO
Keywords:
De-escalation
Antiplatelet therapy
Acute coronary syndrome
Chronic coronary syndrome
Percutaneous coronary intervention
ABSTRACT
Dual antiplatelet therapy (DAPT) is the gold standard after acute coronary syndromes (ACS) or chronic coronary
syndromes (CCS) undergoing percutaneous coronary intervention (PCI).
Because local and systemic ischemic complications can occur particularly in the early phase (i.e. 13 months)
after ACS or PCI, the synergistic platelet inhibition of aspirin and a P2Y
12
inhibitor is of the utmost importance in
this early phase. Moreover, the use of the more potent P2Y
12
inhibitors prasugrel and ticagrelor have shown to
further reduce the incidence of ischemic events compared to clopidogrel after an ACS. On the other hand,
prolonged and potent antiplatelet therapy are inevitably associated with increased bleeding, which unlike
thrombotic risk, tends to be stable over time and may outweigh the benet of reducing ischemic events in these
patients.
The duration and composition of antiplatelet therapy remains a topic of debate in cardiology due to competing
ischemic and bleeding risks, with guidelines and recommendations considerably evolving in the past years. An
emerging strategy, called de-escalation, consisting in the administration of a less intense antithrombotic
therapy after a short course of standard DAPT, has shown to reduce bleeding without any trade-off in ischemic
events. De-escalation may be achieved with different antithrombotic strategies and can be either unguided or
guided by platelet function or genetic testing. The aim of this review is to summarize the evidence and provide
practical recommendations on the use of different de-escalation strategies in patients with ACS and CCS.
1. Introduction
Since more than two decades, dual antiplatelet therapy (DAPT) with
aspirin and a P2Y
12
receptor inhibitor is considered the gold standard for
patients with acute coronary syndrome (ACS) as well as for those with
chronic coronary syndrome (CCS) undergoing percutaneous coronary
Abbreviations: DAPT, Dual Anti-Platelet Therapy^; ACS, Acute Coronary Syndromes; CCS, Chronic Coronary Syndromes; RCTs, Randomized Clinical Trials; PCI,
Percutaneous Coronary Intervention; ST, Stent Thrombosis; MI, Myocardial Infarction; DES, Drug Eluting Stents; PFT, Platelet Function Tests; HPR, High Platelet
Reactivity; LPR, Low Platelet Reactivity; BARC, Bleeding Academy Research Consortium.
* Corresponding author at: Cardiology Department, Heart Institute, Sorbonne University, 47-83 Bld de lhˆ
opital, Piti´
e-Salp´
etri`
ere Hospital, Paris F-75013, France.
E-mail address: cardiology.sabouret@gmail.com (P. Sabouret).
1
These authors contributed equally to the work.
Contents lists available at ScienceDirect
European Journal of Internal Medicine
journal homepage: www.elsevier.com/locate/ejim
https://doi.org/10.1016/j.ejim.2022.12.008
Received 5 November 2022; Received in revised form 14 December 2022; Accepted 16 December 2022
European Journal of Internal Medicine xxx (xxxx) xxx
2
intervention (PCI) [1]. The more potent P2Y
12
inhibitors prasugrel and
ticagrelor have shown to reduce ischemic events at the cost of increased
bleeding compared with clopidogrel [2,3]. Therefore, a 12-month DAPT
with potent P2Y
12
inhibitors represents the standard-of-care after ACS
and may be considered after PCI in CCS patients in specic high
ischemic risk situations [46]. However, the efcacy and safety of a
12-month DAPT duration have been established in randomized
controlled trials (RCTs) presenting several differences compared with
the modern clinical setting, such as the low use of concomitant optimal
medical therapy (i.e. statins) and the use of PCI without stenting or with
old generation stent platforms, occurring late after the diagnosis of ACS
and often performed by operators with little experience [7]. Moreover,
fragile and high bleeding risk patients were typically excluded from an
invasive strategy in early RCTs, while PCI is now currently extended to
patients previously considered at prohibitive risk.
The increasing understating that bleeding events carry important
clinical implications and that the response to antiplatelet agents vary
widely according to the ischemic and bleeding risks as well as to the
individual pharmacodynamic response to antiplatelets, suggests that a
one-size-ts-all approach may represent a suboptimal approach in
several subgroup of patients [8]. Recent guidelines have proposed a
personalization of therapy according to patients bleeding and ischemic
risks, with dedicated scores designed to predict such risks [46].
Nevertheless, in the real world setting, a prolonged and intense anti-
platelet therapy is often difcult to maintain due to the patients clinical
characteristics and comorbidities, and the use of scores is limited by a
several challenges such as the large overlap between factors associated
with increased ischemic and bleeding events. Therefore, a number of
antithrombotic strategies have been tested in the last years with the aim
of optimizing the balance between bleeding and ischemic risks [1].
Because the ischemic risk is highest in the early phase after ACS/PCI
while bleeding risk remains relatively stable over time, there has been a
great interest towards the use of a more intense antithrombotic regimen
early after ACS or PCI, followed by a less intense regimen thereafter, in
the attempt to reduce bleeding without any trade-off in ischemic events
(the so called de-escalation strategy). In this review we discuss the
rationale and provide an appraisal of the evidence in support of such a
strategy.
2. Rationale for a de-escalation of antiplatelet therapy
Early after the introduction of PCI, the main focus of antithrombotic
therapy was not only the reduction of ischemic recurrences, such as
spontaneous myocardial infarction (MI), but also the prevention of local
ischemic events such as stent thrombosis (ST) [9]. Later, with the
introduction of rst generation drug eluting stents (DES) which were
burdened by an increased risk of ST, including those occurring late after
PCI, a more prolonged and intense antithrombotic therapy has been
implemented in the attempt of preventing ST [10]. Such intensied
antithrombotic regimens led to an increased risk of bleeding, which
have been recently recognized to have important clinical implications,
leading to increased mortality through direct and indirect mechanisms
[11,12]. Indeed, bleeding can cause direct damages such as haemor-
rhagic stroke but can also lead to many indirect consequences associated
with poor outcomes [13]. For example, a bleeding event results in the
interruption of antiplatelet therapy which leads to a higher risk of
thrombotic complications. Furthermore, anemia results in decreased
oxygen supply and increased oxygen consumption due to neuroendo-
crine activation and requires blood transfusions which lead to depletion
of 2,3-diphosphoglyceric acid and nitric oxide, resulting in low tissue
oxygen and vasoconstriction which enhance platelets aggregation [12].
Compelling evidence supports the fact that, in both ACS or CCS patients
undergoing PCI, the very early period after ACS or PCI is characterized
by the greatest ischemic risk [1]. Indeed, MI and ST tend to be more
common in the rst 13 month following ACS or PCI, while the risk of
bleeding represents a major threat continuously over time (Fig. 1) [1].
This is particularly evident among ACS patients in which the rst
months are characterized by enhanced inammatory markers and
thrombophilic status [1,14]. Based on the observation that ischemic risk
tends to decrease while bleeding risk remains stable over time, there has
been increasing interest in the developed of a strategy called de-es-
calation, aimed at using an intense antiplatelet therapy early after ACS
or PCI, in order to reduce ischemic complications, followed by a less
intense antiplatelet therapy thereafter, aimed at reducing bleeding
events [15]. Another important issue to be considered when using an-
tiplatelet therapy after PCI is that the superior clinical efcacy at the cost
of increased bleeding of the potent P2Y
12
inhibitors prasugrel and
ticagrelor, as opposed to clopidogrel, is mainly related to the fact
Fig. 1. Rationale for de-escalation strategy. In the early phase after ACS or PCI, the risk of thrombotic events is highest and an aggressive antiplatelet therapy is
needed to reduce ischemic events. After 3 months since ACS or PCI, the risk of thrombotic events decreases signicantly and other risk factors are responsible for the
risk of ischemic events. The bleeding risk remains stable over time, resulting in a benet/risk ratio in favor of an aggressive antiplatelet therapy only in the early (13
months) phase after ACS or PCI. Abbreviations: acute coronary syndrome (ACS); chronic coronary syndrome (CCS); percutaneous coronary intervention (PCI).
P. Sabouret et al.
European Journal of Internal Medicine xxx (xxxx) xxx
3
2040% of patients treated with clopidogrel are non-responders, while
this rate is trivial in patients treated with prasugrel or ticagrelor [16,17].
Indeed, the wide interindividual variability in the gene responsible for
the transcription of the hepatic cytochrome P450 (CYP) 2C19 system,
which is responsible for the activation of clopidogrel, is associated with
diminished levels of clopidogrel active metabolite [18]. Patients treated
with clopidogrel, but not those treated with prasugrel or ticagrelor, who
are carriers of one (intermediate metabolizer) or two (poor metabolizer)
loss-of-function (LoF) alleles (CYP2C19×2 and CYP2C19×3) are asso-
ciated with diminished CYP2C19 enzyme activity and diminished levels
of clopidogrel active metabolite, leading to high platelet reactivity
(HPR) and increased thrombotic risk [19,20]. Moreover, the potent
P2Y
12
inhibition provided by prasugrel and ticagrelor has been associ-
ated with low platelet reactivity (LPR) and increased rates of bleeding
without any benet in ischemic events compared with patients
responding to clopidogrel [21]. Patients not responding to clopidogrel
may be identied by specic tools: platelet function tests (PFT), aiming
at identifying the level of platelet inhibition, and genetic tests, aiming at
identifying patients carriers of LoF allels of the CYP2C19 gene [22]. The
implementation of these tools aims to perform a guided selection of
P2Y
12
inhibiting therapy, selectively administering clopidogrel to pa-
tients responding to this drug and prasugrel or ticagrelor to patients
non-responding to clopidogrel, with the goal of decreasing bleeding
without any trade-off in ischemic events.
3. De-escalation among ACS patients undergoing PCI
De-escalation strategies among patients with ACS consist in the
shortening of DAPT duration and in the use of reduced dose or reduced
potency of P2Y
12
inhibitors, which can be either guided by platelet
function or genetic testing or unguided (Fig. 2) (Table 1).
3.1. Current recommendations from international guidelines
The de-escalation strategies recommended by the European Society
of Cardiology (ESC) guidelines consist in the shortening of DAPT fol-
lowed by aspirin or by a P2Y12 inhibitor monotherapy, depending on
the balance between the ischemic and bleeding risk [1,46]. In partic-
ular, the use of ticagrelor monotherapy after 3 months of standard DAPT
is recommended in non-ST-elevation acute coronary syndromes
(NSTE-ACS) with low bleeding risk, with class IIa, level of evidence
(LoE) A. Among patients at HBR, current ESC guidelines recommend the
use of clopidogrel as the P2Y12 inhibitor of choice and DAPT duration
can be shortened to 6 months in ST-elevation acute coronary syndromes
(STE-ACS) patients followed by aspirin alone, 1 month in NSTE-ACS
patients followed by clopidogrel monotherapy or 3 months when
DAPT discontinuation is followed by aspirin alone [46]. Moreover, in
NSTE-ACS patients, ESC guidelines recommend a guided de-escalation
by platelet function test or genetic testing or unguided de-escalation
from potent P2Y
12
inhibitors to clopidogrel with a class IIb, level of
evidence B [4].
3.2. Short DAPT followed by aspirin monotherapy
With regards to DAPT shortening with P2Y
12
discontinuation in ACS,
several trials have been carried out, comparing 6 versus 12-month DAPT
and 3 versus 12-month DAPT durations [1,36] (Table 1). The
DAPT-STEMI was a non-inferiority trial conducted in 2018, enrolling
1100 patients and comparing 6 versus 12 months DAPT in ACS patients.
The primary endpoint was all-deaths, revascularization, stroke and
major bleedings at 18 months [23]. In this study, six months DAPT was
not inferior to standard 12-month DAPT [23].
The SMART-DATE trial was a non-inferiority trial comparing 6
versus 12 months DAPT in 2712 East Asian patients with ACS. The
primary endpoint was the composite of all-cause death, MI or stroke
[24]. Despite the primary endpoint of non-inferiority consisting in
Fig. 2. De-escalation of antiplatelet therapy among ACS patients. De-escalation strategies among patients with ACS include: (1) shortening DAPT followed by aspirin,
clopidogrel or ticagrelor monotherapy; (2) guided de-escalation; and (3) unguided de-escalation. Abbreviations: acute coronary syndrome (ACS); percutaneous
coronary intervention (PCI); dual antiplatelet therapy (DAPT).
P. Sabouret et al.
European Journal of Internal Medicine xxx (xxxx) xxx
4
all-deaths, MI, or stroke was met, short DAPT was associated with a
doubled risk of MI and with a 50% increased risk of ST, compared to
standard DAPT [24]. The REDUCE trial compared 3 versus 12-month
DAPT in 1496 patients. The primary endpoints were all-cause death,
MI, ST, stroke, target vessel revascularization and Bleeding Academic
Research Consortium (BARC) 25 bleedings at 12 months [25]. Short
DAPT was associated with a doubled risk of ST and a 62% increased risk
of CV death compared to standard DAPT [25]. Red ags, raising some
concern about the use of aspirin monotherapy after a short course of
DAPT were the non-inferiority design of these three trials and, in the
DAPT-STEMI and the REDUCE, the inclusion of both ischemic and
bleeding outcomes as primary endpoint. Of note, an individual patient
data meta-analysis has shown a higher rate of MI or ST with reduced
DAPT duration followed by aspirin alone in ACS [37].
3.3. Short DAPT followed by clopidogrel monotherapy
Another possible strategy for shortening DAPT is the discontinuation
of aspirin after a short course of DAPT maintaining clopidogrel mono-
therapy [1]. RCTs in this setting have tested a strategy of aspirin
discontinuation 1 or 3 months after ACS versus standard 12-month
DAPT (Table 1). Despite RCTs including both ACS and CCS patients
showed encouraging results with this strategy, the recent STOPDAPT-2
ACS non-inferiority trial including 4169 East Asian patients with ACS
showed clopidogrel monotherapy after a 1 or 2-months DAPT not to be
non-inferior to standard DAPT for the primary endpoint of CV death, MI,
any stroke, denite ST or bleeding. Moreover, despite short DAPT fol-
lowed by clopidogrel reduced by 54% the incidence of bleeding, it was
associated with a 50% increase in the composite CV death MI, ST and
stroke and a nearly doubled risk of MI, compared with standard DAPT
[26]. Importantly, it must be highlighted that the evidence of clopi-
dogrel monotherapy after a short course of DAPT comes from RCTs
including only East-Asians, which are a population typically exposed to
greater bleeding and lower ischemic risks compared to other ethnicities
[36]. Furthermore, the non-inferiority design of STOPDAPT-2 ACS and
the inclusion of both ischemic and bleeding outcomes as primary
endpoint limit the statistical power with respect of ischemic outcomes,
which requires further studies to be addressed [26].
3.4. Short DAPT followed by ticagrelor monotherapy
If some concerns exists for a shortening of DAPT followed by clopi-
dogrel monotherapy, a strategy of ticagrelor monotherapy after 13
month of DAPT has shown to reduce bleeding events without any trade-
off in ischemic risk among ACS patients, in RCTs including both Eastern
and Western countries [1,36,27,38]. Specically, TICO randomized
3056 South Korean ACS patients to either 3-month DAPT followed by
ticagrelor monotherapy compared with standard 12-month DAPT with
ticagrelor, showing a reduction of bleeding with no trade-off in ischemic
events [27]. Indeed, the primary composite endpoint of net adverse
clinical event, dened as a composite of major bleeding, all-deaths, MI,
ST, stroke, or target-vessel revascularization, was reduced by 44% in the
de-escalation versus standard DAPT group. Further evidence on the
safety and efcacy of this strategy - mainly including patients from
Western Countries - were provided by the sub-analysis of GLOBAL
LEADERS and TWILIGHT trials, showing a greater reduction of bleeding
with a de-escalation strategy in the subgroup of ACS as compared with
CCS [29,28]. It should be acknowledged that these two subgroup anal-
ysis should only be considered hypothesis generating, also in light of the
fact the GLOBAL LEADERS trial did not meet its primary endpoint and
that the rate of ischemic events was lower than expected in high-risk PCI
patients in the TWILIGHT trial [39,40]. Although these studies are un-
derpowered to assess ischemic outcomes, a study-level meta-analysis
has suggested ticagrelor monotherapy after a short court of DAPT may
represent a safe and effective strategy [38]. Finally, possible practical
concerns surrounding the use of ticagrelor monotherapy stems from the
fact ticagrelor is associated with higher rates (2025%) of discontinua-
tion by the patients due to the incidence of side effects (i.e. dyspnea)
when compared with other antiplatelet drugs and to the fact its action is
reversible and short, potentially leading to a prompt increase in platelet
reactivity in case of voluntary or involuntary discontinuation of the drug
by the patient [41].
3.5. Guided de-escalation
Among patients with ACS, a guided selection of P2Y
12
inhibitors
allows for a selective administration of clopidogrel to clopidogrel re-
sponders, limiting the use of prasugrel and ticagrelor to clopidogrel non-
responders, with aim of reducing bleeding without any trade-off in
ischemic events [15]. This strategy, resulting in a guided de-escalation
of antiplatelet therapy early (<7 days) after ACS or PCI, may be ach-
ieved by the use of PFT or genetic testing (Table 1).
The rst study comparing a PFT-guided de-escalation with standard
therapy in elderly patients with ACS undergoing PCI was ANTARCTIC
(n =877). This study failed to show any difference in the primary
endpoint of CV death, MI, stroke, ST, urgent revascularization, and
BARC 25 bleeding with a guided de-escalation versus standard therapy
[30]. Nevertheless, low dose of prasugrel rather than clopidogrel was
used in this study both in the guided and standard groups, potentially
blunting the superior safety of a de-escalation strategy [30].
TROPICAL-ACS is the largest RCT comparing a PFT-guided de-escalation
with standard therapy among ACS patients (n =2610). A guided
Table 1
RCTs exploring de-escalation in acute coronary syndromes.
STUDY TIMING OF DE-
ESCALATION
STRATEGY OF DE-
ESCALATION
NUMBER OF
PATIENTS
ACS
DAPT STEMI
[23]
6 MONTHS ASPIRIN ALONE 1100
(STEMI
100%)
SMART DATE
[24]
6 MONTHS ASPIRIN ALONE 2712
(STEMI
38%)
REDUCE [25] 3 MONTHS ASPIRIN ALONE 1496
(STEMI
46%)
STOPDAPT-2-
ACS [26]
12 MONTH CLOPIDOGREL
ALONE
3045
(STEMI
57%)
TICO [27] 3 MONTHS TICAGRELOR ALONE 3056
(STEMI
36%)
GLOBAL
LEADERS
Substudy [28]
1 MONTH TICAGRELOR ALONE 3737
(STEMI
46%)
TWILIGHT ACS
[29]
3 MONTHS TICAGRELOR ALONE 4614
(STEMI 0%)
ANTARCTIC [30] EARLY AFTER PCI PFT GUIDED DE-
ESCALATION
877 (STEMI
34%)
TROPICAL-ACS
[31]
EARLY AFTER PCI PFT GUIDED DE-
ESCALATION
2610
(STEMI
55%)
POPular
GENETICS
[32]
EARLY AFTER PCI GENOTYPE GUIDED
DE-ESCALATION
2488
(STEMI
19%)
TOPIC [33] 1 MONTH UNGUIDED DE-
ESCALATION TO
CLOPIDOGREL
646 (STEMI
40%)
TALOS MI [34] 1 MONTH UNGUIDED DE-
ESCALATION TO
CLOPIDOGREL
2697
(STEMI
54%)
HOST-REDUCE-
POLYTECH-
ACS [35]
PRASUGREL 5 MG
DAPT VS
PRASUGREL 10 MG
DAPT
UNGUIDED DE-
ESCALATION
3429
(STEMI
14%)
Abbreviations: acute coronary syndrome (ACS), platelet function test (PFT),
ST-elevation myocardial infarction (STEMI).
P. Sabouret et al.
European Journal of Internal Medicine xxx (xxxx) xxx
5
de-escalation was non-inferior in the primary composite endpoint of CV
death, MI, stroke or BARC bleeding 25 as compared to standard DAPT,
with a trend towards reduced bleeding at 12 months compared to the
standard group [31]. Furthermore, POPular genetics showed both the
non-inferiority of the primary endpoint of all-deaths, MI, denite ST,
stroke, or major bleeding and a signicant 22% reduction of the
co-primary endpoint of major and minor bleeding at 12 months, in 2488
STE-ACS patients randomized to either genotype-guided de-escalation
or standard therapy (mainly ticagrelor) within 48 h after PCI [32].
Limitations of RCTs testing a guided de-escalation in ACS patients
are the use of a primary composite endpoint including both ischemic and
bleeding outcomes and the non-inferiority design, both leading to a
relatively low statistical power with respect to hard ischemic events.
Nevertheless, a recent comprehensive meta-analysis at least partially
overcoming this limitation, showed that a strategy of guided de-
escalation is associated with a 19% reduction of bleeding without any
trade-off in ischemic events [42]. Furthermore, a network meta-analysis
focusing on ACS has shown a guided selection of P2Y
12
inhibitors is
associated with the most favorable safety and efcacy prole compared
to standard treatment with prasugrel or ticagrelor [43].
3.6. Unguided de-escalation
Unguided de-escalation of P2Y
12
inhibiting therapy may be imple-
mented at least 1 month after ACS or PCI, when the risk of local ischemic
events has declined signicantly, and consists in the use of clopidogrel
or reduced dose of prasugrel or ticagrelor after a short course (>1
month) of DAPT with full dose prasugrel or ticagrelor. TOPIC was a
single center trial enrolling 646 ACS patients that compared an un-
guided de-escalation from potent P2Y12 inhibitor to clopidogrel after
one month of DAPT versus 12-month standard DAPT [33]. There was a
52% reduction of the primary endpoint of CV death, urgent revascu-
larization, stroke and BARC 25 bleeding in the de-escalation compared
to standard group, driven by a reduction of BARC 25 bleeding. Of note,
this was a relatively small trial mainly including patients not undergoing
complex-PCI [33].
TALOS-MI trial was the largest RCT (n =2697) comparing an un-
guided de-escalation from ticagrelor to clopidogrel 1 month after ACS
versus standard 12-month DAPT with ticagrelor. There was a 45%
reduction of the primary endpoint of CV death, MI, stroke and BARC 25
bleeding with a de-escalation as compared to standard therapy driven by
a reduction of BARC 25 bleeding [34].
Finally, HOST-REDUCE-POLYTHEC-ACS was the rst trial to
compare a de-escalation strategy by reducing prasugrel from 10 to 5 mg
1 month after ACS versus standard 12-month DAPT with prasugrel in
3429 East Asian patients. The primary endpoint of all-cause death, MI,
ST, repeat revascularization, stroke, and BARC bleeding 25 was
reduced by 30% in the de-escalation versus standard group [35].
Collectively, limitations of an unguided de-escalation of antiplatelet
therapy are the fact they were underpowered to assess ischemic out-
comes, the fact the vast majority of patients enrolled were East Asians
undergoing non-complex PCI, and the fact that this strategy may not
sufciently consider the individual increased ischemic risk of patients
not responding to clopidogrel.
4. De-escalation among CCS patients undergoing PCI
De-escalation strategies among patients with CCS consist in the
shortening of DAPT duration followed by either aspirin or P2Y
12
in-
hibitors monotherapy (Fig. 3) (Table 2).
4.1. Current recommendations from international guidelines
In the clinical setting of CCS, according to current ESC guidelines,
DAPT should be administered for six months, regardless of the stent used
during PCI [5]. Aspirin remains the cornerstone of DAPT, while the
P2Y
12
inhibitor of choice is clopidogrel [5]. The only de-escalation
strategy allowed by current recommendations consists in DAPT short-
ening, ranging from 3 months in patients at high bleeding risk, to 1
month in patients at extremely high bleeding risk [5].
4.2. Short DAPT followed by aspirin monotherapy
A total of 10 RCTs including both ACS and CCS patients have
compared a strategy of DAPT shortening to 1, 3 or 6 months versus a
standard DAPT duration (Table 2) [1,4453]. These studies have
generally provided reassuring results on the efcacy and showed a
consistent reduction of bleeding in favor of a short or very short DAPT as
opposed to a standard DAPT duration [57]. However, none of these trials
have focused only on CCS patients. It could be speculated that because
ACS patients are at increased ischemic risk compared to CCS patients,
the fact that the majority of RCTs on the topic included a variable per-
centage of ACS patients should provide a further reassurance on the
efcacy and safety of a shortening of DAPT duration in these patients,
despite the level of evidence remaining limited, especially for high
ischemic risk patients.
4.3. Short DAPT followed by clopidogrel monotherapy
The STOPDAPT-2 and the SMART-CHOICE trials reported promising
results on the possible discontinuation of aspirin 13 months after ACS
or PCI followed by clopidogrel monotherapy in a mixed population of
Fig. 3. De-escalation of antiplatelet therapy among CCS patients undergoing PCI with or without concomitant AF. De-escalation strategies among patients with CCS
with or without concomitant AF include shortening DAPT followed by aspirin or clopidogrel monotherapy. Abbreviations: dual antiplatelet therapy (DAPT); oral
anticoagulant (OAC); chronic coronary syndrome (CCS); atrial brillation (AF).
P. Sabouret et al.
European Journal of Internal Medicine xxx (xxxx) xxx
6
ACS and CCS [55,54]. A sub-study of the STOPDAPT-2 trial suggested
clopidogrel monotherapy after 1-month of DAPT was superior in major
bleeding and non-inferior in the primary endpoint of CV death, MI,
denite ST, any stroke or bleeding compared to 12 months DAPT,
regardless ACS or CCS presentation [58]. Moreover, while 1-month
DAPT was associated with a numerical increase in CV events in ACS
patients, there was no increase in CCS patients [58].
Limitations of this strategy are represented by the fact these studies
were underpowered for ischemic outcomes, the fact the populations
from these two trials were from East-Asia and the fact that the use of
unguided clopidogrel monotherapy does not take into account of the
interindividual response to this antiplatelet agent.
4.4. Short DAPT followed by prasugrel monotherapy
Prasugrel monotherapy after a short course of DAPT has not been
adequately tested in RCTs, and its use is limited to a pilot study enrolling
201 patients [56]. In this study, selected low-risk CCS patients treated
with third-generation DES underwent aspirin discontinuation on the day
of the index procedure and prasugrel monotherapy represented the only
antiplatelet agent used. At 3 months, the rate of ischemic and bleeding
events was extremely low in both treatment arms [56].
4.5. Short DAPT followed by ticagrelor monotherapy
No RCTs using ticagrelor monotherapy after a short course of DAPT
have focus on CCS patients (Table 1). Evidence in this setting comes
from a pre-specied sub-analysis of the GLOBAL LEADERS trial ac-
cording to clinical presentation (ACS versus CCS)[58]. In this study,
while there was no difference in efcacy, dened as all-deaths and MI,
between treatment strategies by subgroup, a strategy of ticagrelor
monotherapy after 1 month of DAPT appeared to reduce bleeding risk in
patients with ACS but not in patients with CCS, compared to standard
DAPT with ticagrelor [40].
5. Special clinical settings
5.1. Patients with concomitant atrial brillation
Atrial brillation (AF) is as frequent as 10% in patients with ACS or
undergoing PCI [1]. The need for the concomitant use of oral antico-
agulants (OAC) and DAPT in these patients is associated with a pro-
hibitive bleeding risk [59]. This setting was the rst in which a very
early de-escalation strategy, consisting in clopidogrel monotherapy after
a very short course of DAPT (about one week) on the background of OAC
was adopted [60]. Guidelines suggest as gold standard in this setting a
triple therapy (aspirin plus clopidogrel plus an oral anticoagulant) for 1
week after PCI, followed by aspirin discontinuation for 6 months in CCS
and 12 months in ACS [46]. Among high ischemic risk patients,
duration of triple therapy could be prolonged up to 1 month [4]. Finally,
ticagrelor or prasugrel may be considered in association with a NOAC
with a class IIa, LOE B [4]. This recommendation stems from the results
of 4 recent RCTs, each using a different novel OAC (NOAC, dabigatran,
rivaroxaban, apixaban, or edoxaban) in combination with a P2Y12 in-
hibitor (mainly clopidogrel) versus a prolonged triple therapy [59,61].
However, because of several limitations of these studies and the fact they
were not statistically powered to rule out any increase in ischemic events
with a de-escalation strategy, whether the optimal duration of triple
therapy for the majority of patients should be 1 week or 1 month re-
mains debated [60]. Meta-analysis have played an important role in
increasing statistical power for ischemic endpoints, but found incon-
sistent results with a possible increase of ST with the de-escalation
strategy in the overall population and of MI in the ACS subgroup
[6264]. Moreover, the use of ticagrelor was low (<10%) and that of
prasugrel very low (<3%) in this trials, limiting the evidence in support
of the use of P2Y
12
inhibitors different from clopidogrel in this setting.
Finally, the use of unguided clopidogrel monotherapy very early after
ACS or PCI, which is the period considered at highest ischemic risk, may
represent a source of concern especially in ACS and high ischemic risk
patients, as it does not take into account of the interindividual response
to this antiplatelet agent (Fig. 3).
6. Conclusions and future perspectives
Current evidence support a larger use of de-escalation strategies of
antiplatelet therapy in clinical practice. An individualized approach
remains more than ever mandatory to determine the best antithrombotic
strategy, providing an optimal balance between bleeding and ischemic
risks at the individual patient level. Because the primary scope of de-
Table 2
RCTs exploring de-escalation in acute coronary syndrome and chronic coronary
syndrome settings.
STUDY TIMING OF DE-
ESCALATION
STRATEGY OF
DE-ESCALATION
NUMBER
OF
PATIENTS
ACS/CCS
EXCELLENT
[44]
6 MONTHS ASPIRIN ALONE 1443 (ACS
51%, CCS
49%)
RESET [45] 3 MONTHS ASPIRIN ALONE 2148 (ACS
54%, CCS
46%)
OPTIMIZE [46] 3 MONTHS ASPIRIN ALONE 3211 (ACS
32%, CCS
68%)
SECURITY [47] 6 MONTHS ASPIRIN ALONE 1404 (ACS
39%, CCS
61%)
ISAR-SAFE [48] 6 MONTHS ASPIRIN ALONE 4005 (ACS
39%, CCS
61%)
I-LOVE-IT [49] 3 MONTHS ASPIRIN ALONE 1829 (ACS
85%, CCS
15%)
NIPPON [50] 6 MONTHS ASPIRIN ALONE 3773 (ACS
32%, CCS
68%)
OPTIMA-C [51] 6 MONTHS ASPIRIN ALONE 1368 (ACS
51%, CCS
49%)
One-Month
DAPT [52]
1 MONTH ASPIRIN ALONE 3020 (ACS
39%, CCS
61%)
MASTER DAPT
[53]
1 MONTH ASPIRIN ALONE 4434 (ACS
49%, CCS
51%)
SMART-
CHOICE [54]
3 MONTHS CLOPIDOGREL
ALONE
2993 (ACS
58%, CCS
42%)
GLOBAL
LEADERS
[40]
TICAGRELOR
MONOTHERAPY FOR 23
MONTHS VS STANDARD
DAPT
TICAGRELOR
ALONE
2712 (ACS
38%, CCS
62%)
TWILIGHT [39] 1 MONTH TICAGRELOR
ALONE
7119 (ACS
64%, CCS
3%)
STOPDAPT-2
[55]
1 MONTH CLOPIDOGREL
ALONE
3045 (ACS
38%, CCS
62%)
CCS
GLOBAL
LEADERS
Substudy
[28]
1 MONTH TICAGRELOR
ALONE
3750
ASET [56] ON THE DAY OF THE
INDEX PROCEDURE
PRASUGREL
ALONE
201
Abbreviations: acute coronary syndrome (ACS), chronic coronary syndrome
(CCS).
P. Sabouret et al.
European Journal of Internal Medicine xxx (xxxx) xxx
7
escalation strategies is to reduce bleeding, the benet of this strategy is
greater among patients at high bleeding risk. Thus, the identication of
this cohort of patients is of the utmost importance in clinical practice
and should be routinely performed. Furthermore, the increasing evi-
dence supporting the clinical impact of the interindividual variability in
response to clopidogrel support the implementation of tools aiming a
guiding the use of P2Y
12
inhibitor after ACS or PCI.
Since a number of de-escalation strategies have been proposed, a
careful appraisal of the available evidence is key for the adoption of
these strategies in clinical practice. Unfortunately, no comparison be-
tween different de-escalation strategies is currently available, as the
only evidence in this regards is represented by a network meta-analysis
in which effect estimates are mainly based on indirect comparisons and
its results should be interpreted with caution [65]. Among ACS, a guided
selection of antiplatelet therapy is of particular interest, allowing for a
guided de-escalation from the potent P2Y
12
inhibitor (prasugrel and
ticagrelor) to clopidogrel selectively among patients responding to clo-
pidogrel, reducing bleeding without any trade-off in ischemic events. To
this extent, the availability of bedside point-of-care, rapid, relatively
cheap and easy to use genetic testing may facilitate the adoption of this
strategy in the future. Another very promising strategy in ACS is rep-
resented by the use of a potent P2Y
12
inhibitor or of guided clopidogrel
after 1 to 3 months of standard DAPT. Nevertheless, current evidence are
only available on the use of ticagrelor monotherapy after 3 months of
standard DAPT, a strategy already recommended by guidelines. In the
light of the available evidence, a 12-month DAPT duration with potent
P2Y
12
inhibitors should only be applied to selected patients with
high-ischemic risk and low bleeding risk.
On the other hand, among CCS with or without concomitant AF, the
evidence in support of a de-escalation strategy is weaker and basically
consists in the shortening of DAPT duration followed by aspirin in pa-
tients with sinus rhythm or clopidogrel in patients with concomitant AF.
A de-escalation strategy in these patients is likely to be advantageous in
high bleeding risk patients, but whether it should represent the standard
of care remains uncertain. Moreover, whether the use of clopidogrel
(either unguided or guided) may be advantageous as compared to
aspirin after DAPT discontinuation requires further investigations.
Furthermore, because prasugrel and ticagrelor are associated with
increased bleeding risk compared to clopidogrel, whether the use of
prasugrel or ticagrelor monotherapy could represent a valuable option
for CCS patients is unlikely.
Finally, other fundamental issues to be considered when adopting
de-escalation strategies are the control of risk factors and patients
compliance with guideline recommended treatments. Patients with
intensive control of all cardiovascular risk factors, with a good compli-
ance to therapy and receiving the best possible cardiovascular therapy
(statins, ezetimibe +/- proprotein convertase subtilisin/kexin type 9
inhibitors, glucagon-like peptide 1-receptor agonists, sodium-glucose
cotransporter-2 inhibitors, beta-blockers) are the best candidates to
receive a de-escalated antiplatelet therapy.
Disclosures
P.S. reports consulting fees by Astra Zeneca, Amgen, BI, BMS, Lilly,
Novartis, Sano, Servier, Vifor, outside the submitted work. M.B.:
speakers bureau: Amgen, KRKA, Polpharma, Novartis, Sano-Aventis,
Teva, and Zentiva; consultant to Amgen, Daiichi Sankyo, Esperion,
Novartis, and Sano-Aventis; grants from Amgen, Sano, and Valeant,
outside the submitted work. M.G. reports consulting fees by Terumo,
outside the submitted work. L.S., D.F., W.U., M.G., J.P.C., S.D.R., M.S.,
report no disclosures. GBZ has consulted for Balmed, Cardionovum,
Crannmedical, Eukon, Innovheart, Guidotti, Meditrial, Microport,
Opsens Medical, Replycare, Teleex, and Terumo outside the submitted
work. No funding for this article.
References
[1] Angiolillo DJ, Galli M, Collet JP, Kastrati A, ODonoghue ML. Antiplatelet therapy
after percutaneous coronary intervention. EuroIntervention 2022;17(17):
e137196. https://doi.org/10.4244/EIJ-D-21-00904. Apr 1.
[2] James S, Akerblom A, Cannon CP, Emanuelsson H, Husted S, Katus H, Skene A,
Steg PG, Storey RF, Harrington R, Becker R, Wallentin L. Comparison of ticagrelor,
the rst reversible oral P2Y(12) receptor antagonist, with clopidogrel in patients
with acute coronary syndromes: rationale, design, and baseline characteristics of
the PLATelet inhibition and patient outcomes (PLATO) trial. Am Heart J 2009;157
(4):599605. https://doi.org/10.1016/j.ahj.2009.01.003. Apr.
[3] Wiviott SD, Braunwald E, McCabe CH, Montalescot G, Ruzyllo W, Gottlieb S,
Neumann FJ, Ardissino D, De Servi S, Murphy SA, Riesmeyer J, Weerakkody G,
Gibson CM, Antman EM. Prasugrel versus clopidogrel in patients with acute
coronary syndromes. N Engl J Med 2007;357(20):200115. https://doi.org/
10.1056/NEJMoa0706482. Nov 15.
[4] Collet JP, Thiele H, Barbato E, Barth´
el´
emy O, Bauersachs J, Bhatt DL, Dendale P,
Dorobantu M, Edvardsen T, Folliguet T, Gale CP, Gilard M, Jobs A, Jüni P,
Lambrinou E, Lewis BS, Mehilli J, Meliga E, Merkely B, Mueller C, Rof M,
Rutten FH, Sibbing D, Siontis GCM. 2020 ESC Guidelines for the management of
acute coronary syndromes in patients presenting without persistent ST-segment
elevation. Eur Heart J 2021;42(14):1289367. https://doi.org/10.1093/eurheartj/
ehaa575. Apr 7.
[5] Knuuti J, Wijns W, Saraste A, Capodanno D, Barbato E, Funck-Brentano C,
Prescott E, Storey RF, Deaton C, Cuisset T, Agewall S, Dickstein K, Edvardsen T,
Escaned J, Gersh BJ, Svitil P, Gilard M, Hasdai D, Hatala R, Mahfoud F, Masip J,
Muneretto C, Valgimigli M, Achenbach S, Bax JJ. 2019 ESC Guidelines for the
diagnosis and management of chronic coronary syndromes. Eur Heart J 2020;41
(3):40777. https://doi.org/10.1093/eurheartj/ehz425. Jan 14.
[6] Ibanez B, James S, Agewall S, Antunes MJ, Bucciarelli-Ducci C, Bueno H,
Caforio ALP, Crea F, Goudevenos JA, Halvorsen S, Hindricks G, Kastrati A,
Lenzen MJ, Prescott E, Rof M, Valgimigli M, Varenhorst C, Vranckx P,
Widimský P. 2017 ESC Guidelines for the management of acute myocardial
infarction in patients presenting with ST-segment elevation: the Task Force for the
management of acute myocardial infarction in patients presenting with ST-segment
elevation of the European Society of Cardiology (ESC). Eur Heart J 2018;39(2):
11977. https://doi.org/10.1093/eurheartj/ehx393. Jan 7.
[7] Yusuf S, Zhao F, Mehta SR, Chrolavicius S, Tognoni G, Fox KK. Effects of
clopidogrel in addition to aspirin in patients with acute coronary syndromes
without ST-segment elevation. N Engl J Med 2001;345(7):494502. https://doi.
org/10.1056/NEJMoa010746. Aug 16.
[8] Galli M, Ortega-Paz L, Franchi F, Rollini F, Angiolillo DJ. Precision medicine in
interventional cardiology: implications for antiplatelet therapy in patients
undergoing percutaneous coronary intervention. Pharmacogenomics 2022;23(13):
72337. https://doi.org/10.2217/pgs-2022-0057. Aug.
[9] Cao D, Chandiramani R, Chiarito M, Claessen BE, Mehran R. Evolution of
antithrombotic therapy in patients undergoing percutaneous coronary
intervention: a 40-year journey. Eur Heart J 2021;42(4):33951. https://doi.org/
10.1093/eurheartj/ehaa824. Jan 21.
[10] Navarese EP, Andreotti F, Schulze V, Kołodziejczak M, Buffon A, Brouwer M,
Costa F, Kowalewski M, Parati G, Lip GY, Kelm M, Valgimigli M. Optimal duration
of dual antiplatelet therapy after percutaneous coronary intervention with drug
eluting stents: meta-analysis of randomised controlled trials. BMJ 2015;350:h1618.
https://doi.org/10.1136/bmj.h1618. Apr 16.
[11] Capodanno D, Bhatt DL, Gibson CM, James S, Kimura T, Mehran R, Rao SV,
Steg PG, Urban P, Valgimigli M, Windecker S, Angiolillo DJ. Bleeding avoidance
strategies in percutaneous coronary intervention. Nat Rev Cardiol 2022;19(2):
11732. https://doi.org/10.1038/s41569-021-00598-1.
[12] Galli M, Laborante R, Andreotti F, Vergallo R, Montone RA, Iaconelli A, Trani C,
Burzotta F, Crea F, DAmario D. Bleeding complications in patients undergoing
percutaneous coronary intervention. Rev Cardiovasc Med. 2022;23(8):286.
https://doi.org/10.31083/j.rcm2308286.
[13] Buccheri S, Capodanno D, James S, Angiolillo DJ. Bleeding after antiplatelet
therapy for the treatment of acute coronary syndromes: a review of the evidence
and evolving paradigms. Expert Opin Drug Saf 2019;18(12):117189. https://doi.
org/10.1080/14740338.2019.1680637. Dec.
[14] Biasucci LM, La Rosa G, Pedicino D, DAiello A, Galli M, Liuzzo G. Where does
inammation t? Curr Cardiol Rep 2017;19(9):84. https://doi.org/10.1007/
s11886-017-0896-0. Sep.
[15] Galli M, Angiolillo DJ. De-escalation of antiplatelet therapy in acute coronary
syndromes: why, how and when? Front Cardiovasc Med 2022;9:975969. https://
doi.org/10.3389/fcvm.2022.975969. Aug 25.
[16] Aradi D, Kirtane A, Bonello L, Gurbel PA, Tantry US, Huber K, Freynhofer MK, ten
Berg J, Janssen P, Angiolillo DJ, Siller-Matula JM, Marcucci R, Patti G,
Mangiacapra F, Valgimigli M, Morel O, Palmerini T, Price MJ, Cuisset T, Kastrati A,
Stone GW, Sibbing D. Bleeding and stent thrombosis on P2Y12-inhibitors:
collaborative analysis on the role of platelet reactivity for risk stratication after
percutaneous coronary intervention. Eur Heart J 2015;36(27):176271. https://
doi.org/10.1093/eurheartj/ehv104. Jul 14.
[17] Pereira NL, Farkouh ME, So D, Lennon R, Geller N, Mathew V, Bell M, Bae JH,
Jeong MH, Chavez I, Gordon P, Abbott JD, Cagin C, Baudhuin L, Fu YP,
Goodman SG, Hasan A, Iturriaga E, Lerman A, Sidhu M, Tanguay JF, Wang L,
Weinshilboum R, Welsh R, Rosenberg Y, Bailey K, Rihal C. Effect of genotype-
guided oral P2Y12 inhibitor selection vs conventional clopidogrel therapy on
ischemic outcomes after percutaneous coronary intervention: the TAILOR-PCI
P. Sabouret et al.
European Journal of Internal Medicine xxx (xxxx) xxx
8
randomized clinical trial. JAMA 2020;324(8):76171. https://doi.org/10.1001/
jama.2020.12443. Aug 25.
[18] Mega JL, Simon T, Collet JP, Anderson JL, Antman EM, Bliden K, Cannon CP,
Danchin N, Giusti B, Gurbel P, Horne BD, Hulot JS, Kastrati A, Montalescot G,
Neumann FJ, Shen L, Sibbing D, Steg PG, Trenk D, Wiviott SD, Sabatine MS.
Reduced-function CYP2C19 genotype and risk of adverse clinical outcomes among
patients treated with clopidogrel predominantly for PCI: a meta-analysis. JAMA
2010;304(16):182130. https://doi.org/10.1001/jama.2010.1543. Oct 27.
[19] Galli M, Capodanno D, Andreotti F, Crea F, Angiolillo DJ. Safety and efcacy of
P2Y12 inhibitor monotherapy in patients undergoing percutaneous coronary
interventions. Expert Opin Drug Saf 2021;20(1):921. https://doi.org/10.1080/
14740338.2021.1850691. Jan.
[20] Sibbing D, Aradi D, Alexopoulos D, Ten Berg J, Bhatt DL, Bonello L, Collet JP,
Cuisset T, Franchi F, Gross L, Gurbel P, Jeong YH, Mehran R, Moliterno DJ,
Neumann FJ, Pereira NL, Price MJ, Sabatine MS, So DYF, Stone GW, Storey RF,
Tantry U, Trenk D, Valgimigli M, Waksman R, Angiolillo DJ. Updated expert
consensus statement on platelet function and genetic testing for guiding P2Y12
receptor inhibitor treatment in percutaneous coronary intervention. JACC
Cardiovasc Interv 2019;12(16):152137. https://doi.org/10.1016/j.
jcin.2019.03.034. Aug 26.
[21] Aradi D, Kirtane A, Bonello L, Gurbel PA, Tantry US, Huber K, Freynhofer MK, ten
Berg J, Janssen P, Angiolillo DJ, Siller-Matula JM, Marcucci R, Patti G,
Mangiacapra F, Valgimigli M, Morel O, Palmerini T, Price MJ, Cuisset T, Kastrati A,
Stone GW, Sibbing D. Bleeding and stent thrombosis on P2Y12-inhibitors:
collaborative analysis on the role of platelet reactivity for risk stratication after
percutaneous coronary intervention[CE: duplicate reference with 16]. Eur Heart J
2015;36(27):176271. https://doi.org/10.1093/eurheartj/ehv104. Jul 14.
[22] Galli M, Franchi F, Rollini F, Angiolillo DJ. Role of platelet function and genetic
testing in patients undergoing percutaneous coronary intervention. Trends
Cardiovasc Med 2021. https://doi.org/10.1016/j.tcm.2021.12.007. Dec 20S1050-
1738(21)00157-2.
[23] Kedhi E, Fabris E, van der Ent M, Buszman P, von Birgelen C, Roolvink V,
Zurakowski A, Schotborgh CE, Hoorntje JCA, Eek CH, Cook S, Togni M,
Meuwissen M, van Royen N, van Vliet R, Wedel H, Delewi R, Zijlstra F. 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. https://doi.org/10.1136/bmj.k3793. Oct 2.
[24] Hahn JY, Song YB, Oh JH, Cho DK, Lee JB, Doh JH, Kim SH, Jeong JO, Bae JH,
Kim BO, Cho JH, Suh IW, Kim DI, Park HK, Park JS, Choi WG, Lee WS, Kim J,
Choi KH, Park TK, Lee JM, Yang JH, Choi JH, Choi SH, Gwon HC. 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. Lancet 2018;391(10127):127484.
https://doi.org/10.1016/S0140-6736(18)30493-8. Mar 31.
[25] De Luca G, Damen SA, Camaro C, Benit E, Verdoia M, Rasoul S, Liew HB, Polad J,
Ahmad WA, Zambahari R, Postma S, Kedhi E, Suryapranata H. 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(11):e9908. https://doi.org/10.4244/EIJ-D-19-00539.
Dec 6.
[26] Watanabe H, Morimoto T, Natsuaki M, Yamamoto K, Obayashi Y, Ogita M, Suwa S,
Isawa T, Domei T, Yamaji K, Tatsushima S, Watanabe H, Ohya M, Tokuyama H,
Tada T, Sakamoto H, Mori H, Suzuki H, Nishikura T, Wakabayashi K, Hibi K,
Abe M, Kawai K, Nakao K, Ando K, Tanabe K, Ikari Y, Morino Y, Kadota K,
Furukawa Y, Nakagawa Y, Kimura T. Comparison of clopidogrel monotherapy after
1 to 2 months of dual antiplatelet therapy with 12 months of dual antiplatelet
therapy in patients with acute coronary syndrome: the STOPDAPT-2 ACS
randomized clinical trial. JAMA Cardiol 2022;7(4):40717. https://doi.org/
10.1001/jamacardio.2021.5244. Apr 1.
[27] Kim BK, Hong SJ, Cho YH, Yun KH, Kim YH, Suh Y, Cho JY, Her AY, Cho S,
Jeon DW, Yoo SY, Cho DK, Hong BK, Kwon H, Ahn CM, Shin DH, Nam CM, Kim JS,
Ko YG, Choi D, Hong MK, Jang Y. 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. JAMA 2020;323(23):
240716. https://doi.org/10.1001/jama.2020.7580. Jun 16.
[28] Vranckx P, Valgimigli M, Odutayo A, Serruys PW, Hamm C, Steg PG, Heg D, Mc
Fadden EP, Onuma Y, Benit E, Janssens L, Diletti R, Ferrario M, Huber K, R¨
aber L,
Windecker S, Jüni P. Efcacy and safety of ticagrelor monotherapy by clinical
presentation: pre-specied analysis of the global leaders trial. J Am Heart Assoc
2021;10(18):e015560. https://doi.org/10.1161/JAHA.119.015560. Sep 21Epub
2021 Sep 17.
[29] Baber U, Dangas G, Angiolillo DJ, Cohen DJ, Sharma SK, Nicolas J, Briguori C,
Cha JY, Collier T, Dudek D, Dˇ
zavik V, Escaned J, Gil R, Gurbel P, Hamm CW,
Henry T, Huber K, Kastrati A, Kaul U, Kornowski R, Krucoff M, Kunadian V,
Marx SO, Mehta S, Moliterno D, Ohman EM, Oldroyd K, Sardella G, Sartori S,
Shlofmitz R, Steg PG, Weisz G, Witzenbichler B, Han YL, Pocock S, Gibson CM,
Mehran R. Ticagrelor alone vs. ticagrelor plus aspirin following percutaneous
coronary intervention in patients with non-ST-segment elevation acute coronary
syndromes: TWILIGHT-ACS. Eur Heart J 2020;41(37):353345. https://doi.org/
10.1093/eurheartj/ehaa670. Oct 1.
[30] Cayla G, Cuisset T, Silvain J, Leclercq F, Manzo-Silberman S, Saint-Etienne C,
Delarche N, Bellemain-Appaix A, Range G, El Mahmoud R, Carri´
e D, Belle L,
Souteyrand G, Aubry P, Sabouret P, du Fretay XH, Beygui F, Bonnet JL, Lattuca B,
Pouillot C, Varenne O, Boueri Z, Van Belle E, Henry P, Motreff P, Elhadad S,
Salem JE, Abtan J, Rousseau H, Collet JP, Vicaut E, Montalescot G. Platelet
function monitoring to adjust antiplatelet therapy in elderly patients stented for an
acute coronary syndrome (ANTARCTIC): an open-label, blinded-endpoint,
randomised controlled superiority trial. Lancet 2016;388(10055):201522.
https://doi.org/10.1016/S0140-6736(16)31323-X. Oct 22Epub 2016 Aug 28.
[31] Sibbing D, Aradi D, Jacobshagen C, Gross L, Trenk D, Geisler T, Orban M,
Hadamitzky M, Merkely B, Kiss RG, Kom´
ocsi A, D´
ezsi CA, Holdt L, Felix SB,
Parma R, Klopotowski M, Schwinger RHG, Rieber J, Huber K, Neumann FJ,
Koltowski L, Mehilli J, Huczek Z, Massberg S. Guided de-escalation of antiplatelet
treatment in patients with acute coronary syndrome undergoing percutaneous
coronary intervention (TROPICAL-ACS): a randomised, open-label, multicentre
trial. Lancet. 2017;390(10104):174757. https://doi.org/10.1016/S0140-6736
(17)32155-4. Oct 14Epub 2017 Aug 28.
[32] Claassens DMF, Vos GJA, Bergmeijer TO, Hermanides RS, van t Hof AWJ, van der
Harst P, Barbato E, Morisco C, Tjon Joe Gin RM, Asselbergs FW, Mosterd A,
Herrman JR, Dewilde WJM, Janssen PWA, Kelder JC, Postma MJ, de Boer A,
Boersma C, Deneer VHM. Ten Berg JM. A genotype-guided strategy for Oral P2Y12
inhibitors in primary PCI. N Engl J Med 2019;381(17):162131. https://doi.org/
10.1056/NEJMoa1907096. Oct 24.
[33] Cuisset T, Deharo P, Quilici J, Johnson TW, Deffarges S, Bassez C, Bonnet G,
Fourcade L, Mouret JP, Lambert M, Verdier V, Morange PE, Alessi MC, Bonnet JL.
Benet of switching dual antiplatelet therapy after acute coronary syndrome: the
TOPIC (timing of platelet inhibition after acute coronary syndrome) randomized
study. Eur Heart J 2017;38(41):30708. https://doi.org/10.1093/eurheartj/
ehx175. Nov 1.
[34] Park MW, Kim CJ, Kim MC, Choo EH, Hwang BH, Park CS, Kim HY, Yoo KD,
Jeon DS, Jeong MH, Seung KB, Ahn Y, Chang K. A prospective, multicentre,
randomised, open-label trial to compare the efcacy and safety of clopidogrel
versus ticagrelor in stabilised patients with acute myocardial infarction after
percutaneous coronary intervention: rationale and design of the TALOS-AMI trial.
EuroIntervention 2021;16(14):11706. https://doi.org/10.4244/EIJ-D-20-00187.
Feb 19.
[35] Kim HS, Kang J, Hwang D, Han JK, Yang HM, Kang HJ, Koo BK, Rhew JY, Chun KJ,
Lim YH, Bong JM, Bae JW, Lee BK, Park KW. 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. Lancet 2020;396(10257):107989.
https://doi.org/10.1016/S0140-6736(20)31791-8. Oct 10.
[36] Galli M, Angiolillo DJ. Antiplatelet therapy in percutaneous coronary intervention:
latest evidence from randomized controlled trials. Curr Opin Cardiol 2021;36(4):
3906. https://doi.org/10.1097/HCO.0000000000000879. Jul 1.
[37] Palmerini T, Della Riva D, Benedetto U, Bacchi Reggiani L, Feres F, Abizaid A,
Gilard M, Morice MC, Valgimigli M, Hong MK, Kim BK, Jang Y, Kim HS, Park KW,
Colombo A, Chieffo A, Sangiorgi D, Biondi-Zoccai G, G´
en´
ereux P, Angelini GD,
Pufulete M, White J, Bhatt DL, Stone GW. Three, six, or twelve months of dual
antiplatelet therapy after DES implantation in patients with or without acute
coronary syndromes: an individual patient data pairwise and network meta-
analysis of six randomized trials and 11 473 patients. Eur Heart J 2017;38(14):
103443. https://doi.org/10.1093/eurheartj/ehw627. Apr 7.
[38] ODonoghue M.L., Murphy S.A., Sabatine M.S. The safety and efcacy of aspirin
discontinuation on a background of a P2Y12 inhibitor in patients after
percutaneous coronary intervention: a systematic review and meta-analysis.
Circulation. 2020 Aug 11;142(6):53845. doi: 10.1161/
CIRCULATIONAHA.120.046251.
[39] Mehran R, Baber U, Sharma SK, Cohen DJ, Angiolillo DJ, Briguori C, Cha JY,
Collier T, Dangas G, Dudek D, Dˇ
zavík V, Escaned J, Gil R, Gurbel P, Hamm CW,
Henry T, Huber K, Kastrati A, Kaul U, Kornowski R, Krucoff M, Kunadian V,
Marx SO, Mehta SR, Moliterno D, Ohman EM, Oldroyd K, Sardella G, Sartori S,
Shlofmitz R, Steg PG, Weisz G, Witzenbichler B, Han YL, Pocock S, Gibson CM.
Ticagrelor with or without aspirin in high-risk patients after PCI. N Engl J Med
2019;381(21):203242. https://doi.org/10.1056/NEJMoa1908419. Nov 21.
[40] Vranckx P, Valgimigli M, Jüni P, Hamm C, Steg PG, Heg D, van Es GA,
McFadden EP, Onuma Y, van Meijeren C, Chichareon P, Benit E, M¨
ollmann H,
Janssens L, Ferrario M, Moschovitis A, Zurakowski A, Dominici M, Van Geuns RJ,
Huber K, Slagboom T, Serruys PW, Windecker S. 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(10151):9409. https://doi.org/
10.1016/S0140-6736(18)31858-0. Sep 15.
[41] Bonaca MP, Bhatt DL, Oude Ophuis T, Steg PG, Storey R, Cohen M, Kuder J, Im K,
Magnani G, Budaj A, Theroux P, Hamm C, ˇ
Spinar J, Kiss RG, Dalby AJ, Medina FA,
Kontny F, Aylward PE, Jensen EC, Held P, Braunwald E, Sabatine MS. Long-term
tolerability of ticagrelor for the secondary prevention of major adverse
cardiovascular events: a secondary analysis of the PEGASUS-TIMI 54 Trial. JAMA
Cardiol 2016;1(4):42532. https://doi.org/10.1001/jamacardio.2016.1017. Jul 1.
[42] Galli M, Benenati S, Capodanno D, Franchi F, Rollini F, DAmario D, Porto I,
Angiolillo DJ. Guided versus standard antiplatelet therapy in patients undergoing
percutaneous coronary intervention: a systematic review and meta-analysis. Lancet
2021;397(10283):147083. https://doi.org/10.1016/S0140-6736(21)00533-X.
Apr 17.
[43] Galli M, Benenati S, Franchi F, Rollini F, Capodanno D, Biondi-Zoccai G,
Vescovo GM, Cavallari LH, Bikdeli B, Ten Berg J, Mehran R, Gibson CM, Crea F,
Pereira NL, Sibbing D, Angiolillo DJ. Comparative effects of guided vs. potent
P2Y12 inhibitor therapy in acute coronary syndrome: a network meta-analysis of
61 898 patients from 15 randomized trials. Eur Heart J 2022;43(10):95967.
https://doi.org/10.1093/eurheartj/ehab836. Mar 7.
P. Sabouret et al.
European Journal of Internal Medicine xxx (xxxx) xxx
9
[44] Gwon HC, Hahn JY, Park KW, Song YB, Chae IH, Lim DS, Han KR, Choi JH,
Choi SH, Kang HJ, Koo BK, Ahn T, Yoon JH, Jeong MH, Hong TJ, Chung WY,
Choi YJ, Hur SH, Kwon HM, Jeon DW, Kim BO, Park SH, Lee NH, Jeon HK, Jang Y,
Kim HS. Six-month versus 12-month dual antiplatelet therapy after implantation of
drug-eluting stents: the Efcacy of Xience/Promus Versus Cypher to Reduce Late
Loss After Stenting (EXCELLENT) randomized, multicenter study. Circulation
2012;125(3):50513. https://doi.org/10.1161/CIRCULATIONAHA.111.059022.
Jan 24.
[45] Kim BK, Hong MK, Shin DH, Nam CM, Kim JS, Ko YG, Choi D, Kang TS, Park BE,
Kang WC, Lee SH, Yoon JH, Hong BK, Kwon HM, Jang Y. A new strategy for
discontinuation of dual antiplatelet therapy: the RESET Trial (REal Safety and
Efcacy of 3-month dual antiplatelet Therapy following Endeavor zotarolimus-
eluting stent implantation). J Am Coll Cardiol 2012;60(15):13408. https://doi.
org/10.1016/j.jacc.2012.06.043.
[46] Feres F, Costa RA, Abizaid A, Leon MB, Marin-Neto JA, Botelho RV, King 3rd SB,
Negoita M, Liu M, de Paula JE, Mangione JA, Meireles GX, Castello Jr HJ,
Nicolela Jr EL, Perin MA, Devito FS, Labrunie A, Salvadori Jr D, Gusm˜
ao M,
Staico R, Costa Jr JR, de Castro JP, Abizaid AS, Bhatt DL. Three vs twelve months
of dual antiplatelet therapy after zotarolimus-eluting stents: the OPTIMIZE
randomized trial. JAMA 2013;310(23):251022. https://doi.org/10.1001/
jama.2013.282183. Dec 18.
[47] Colombo A, Chieffo A, Frasheri A, Garbo R, Masotti-Centol M, Salvatella N, Oteo
Dominguez JF, Steffanon L, Tarantini G, Presbitero P, Menozzi A, Pucci E, Mauri J,
Cesana BM, Giustino G, Sardella G. Second-generation drug-eluting stent
implantation followed by 6- versus 12-month dual antiplatelet therapy: the
SECURITY randomized clinical trial. J Am Coll Cardiol 2014;64(20):208697.
https://doi.org/10.1016/j.jacc.2014.09.008.
[48] Schulz-Schüpke S, Byrne RA, Ten Berg JM, Neumann FJ, Han Y, Adriaenssens T,
T¨
olg R, Seyfarth M, Maeng M, Zrenner B, Jacobshagen C, Mudra H, von
Hodenberg E, W¨
ohrle J, Angiolillo DJ, von Merzljak B, Rifatov N, Kufner S,
Morath T, Feuchtenberger A, Ibrahim T, Janssen PW, Valina C, Li Y, Desmet W,
Abdel-Wahab M, Tiroch K, Hengstenberg C, Bernlochner I, Fischer M, Schunkert H,
Laugwitz KL, Sch¨
omig A, Mehilli J, Kastrati A. ISAR-SAFE: a randomized, double-
blind, placebo-controlled trial of 6vs. 12 months of clopidogrel therapy after drug-
eluting stenting. Eur Heart J 2015;36(20):125263. https://doi.org/10.1093/
eurheartj/ehu523. May 21.
[49] Han Y, Xu B, Xu K, Guan C, Jing Q, Zheng Q, Li X, Zhao X, Wang H, Zhao X, Li X,
Yu P, Zang H, Wang Z, Cao X, Zhang J, Pang W, Li J, Yang Y, Dangas GD. Six versus
12 months of dual antiplatelet therapy after implantation of biodegradable
polymer sirolimus-eluting stent: randomized substudy of the I-LOVE-IT 2 Trial. Circ
Cardiovasc Interv 2016;9(2):e003145. https://doi.org/10.1161/
CIRCINTERVENTIONS.115.003145. Feb.
[50] Nakamura M, Iijima R, Ako J, Shinke T, Okada H, Ito Y, Ando K, Anzai H,
Tanaka H, Ueda Y, Takiuchi S, Nishida Y, Ohira H, Kawaguchi K, Kadotani M,
Niinuma H, Omiya K, Morita T, Zen K, Yasaka Y, Inoue K, Ishiwata S, Ochiai M,
Hamasaki T, Yokoi H. Dual antiplatelet therapy for 6 versus 18 months after
biodegradable polymer drug-eluting stent implantation. JACC Cardiovasc Interv
2017;10(12):118998. https://doi.org/10.1016/j.jcin.2017.04.019. Jun 26.
[51] Lee BK, Kim JS, Lee OH, Min PK, Yoon YW, Hong BK, Shin DH, Kang TS, Kim BO,
Cho DK, Jeon DW, Woo SI, Choi S, Kim YH, Kang WC, Kim S, Kim BK, Hong MK,
Jang Y, Kwon HM. 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(16):192330. https://doi.org/
10.4244/EIJ-D-17-00792. Mar 20.
[52] Hong SJ, Kim JS, Hong SJ, Lim DS, Lee SY, Yun KH, Park JK, Kang WC, Kim YH,
Yoon HJ, Won H, Nam CM, Ahn CM, Kim BK, Ko YG, Choi D, Jang Y, Hong MK. 1-
month dual-antiplatelet therapy followed by aspirin monotherapy after polymer-
free drug-coated stent implantation: one-month DAPT Trial. JACC Cardiovasc
Interv 2021;14(16):180111. https://doi.org/10.1016/j.jcin.2021.06.003. Aug 23.
[53] Valgimigli M, Frigoli E, Heg D, Tijssen J, Jüni P, Vranckx P, Ozaki Y, Morice MC,
Chevalier B, Onuma Y, Windecker S, Tonino PAL, Rof M, Lesiak M, Mahfoud F,
Bartunek J, Hildick-Smith D, Colombo A, Stankovi´
c G, I˜
niguez A, Schultz C,
Kornowski R, Ong PJL, Alasnag M, Rodriguez AE, Moschovitis A, Laanmets P,
Donahue M, Leonardi S, Smits PC. Dual antiplatelet therapy after PCI in patients at
high bleeding risk. N Engl J Med 2021;385(18):164355. https://doi.org/10.1056/
NEJMoa2108749. Oct 28.
[54] Hahn JY, Song YB, Oh JH, Chun WJ, Park YH, Jang WJ, Im ES, Jeong JO, Cho BR,
Oh SK, Yun KH, Cho DK, Lee JY, Koh YY, Bae JW, Choi JW, Lee WS, Yoon HJ,
Lee SU, Cho JH, Choi WG, Rha SW, Lee JM, Park TK, Yang JH, Choi JH, Choi SH,
Lee SH, Gwon HC. Effect of P2Y12 inhibitor monotherapy vs dual antiplatelet
therapy on cardiovascular events in patients undergoing percutaneous coronary
intervention: the SMART-CHOICE randomized clinical trial. JAMA 2019;321(24):
242837. https://doi.org/10.1001/jama.2019.8146. Jun 25.
[55] Watanabe H, Domei T, Morimoto T, Natsuaki M, Shiomi H, Toyota T, Ohya M,
Suwa S, Takagi K, Nanasato M, Hata Y, Yagi M, Suematsu N, Yokomatsu T,
Takamisawa I, Doi M, Noda T, Okayama H, Seino Y, Tada T, Sakamoto H, Hibi K,
Abe M, Kawai K, Nakao K, Ando K, Tanabe K, Ikari Y, Hanaoka KI, Morino Y,
Kozuma K, Kadota K, Furukawa Y, Nakagawa Y, Kimura T. 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. JAMA 2019;321(24):241427. https://doi.org/10.1001/
jama.2019.8145. Jun 25.
[56] Kogame N, Guimar˜
aes PO, Modolo R, De Martino F, Tinoco J, Ribeiro EE,
Kawashima H, Ono M, Hara H, Wang R, Cavalcante R, Moulin B, Falc˜
ao BAA,
Leite RS, de Almeida Sampaio FB, Morais GR, Meireles GC, Campos CM, Onuma Y,
Serruys PW, Lemos PA. Aspirin-free prasugrel monotherapy following coronary
artery stenting in patients with stable CAD: the ASET pilot study. JACC Cardiovasc
Interv 2020;13(19):225162. https://doi.org/10.1016/j.jcin.2020.06.023. Oct 12.
[57] Benenati S, Crimi G, Canale C, Pescetelli F, De Marzo V, Vergallo R, Galli M, Della
Bona R, Canepa M, Ameri P, Crea F, Porto I. Duration of dual antiplatelet therapy
and subsequent monotherapy type in patients undergoing drug-eluting stent
implantation: a network meta-analysis. Eur Heart J Cardiovasc Pharmacother
2022;8(1):5664. https://doi.org/10.1093/ehjcvp/pvaa127. Jan 5.
[58] Obayashi Y, Watanabe H, Morimoto T, Yamamoto K, Natsuaki M, Domei T,
Yamaji K, Suwa S, Isawa T, Watanabe H, Yoshida R, Sakamoto H, Akao M, Hata Y,
Morishima I, Tokuyama H, Yagi M, Suzuki H, Wakabayashi K, Suematsu N,
Inada T, Tamura T, Okayama H, Abe M, Kawai K, Nakao K, Ando K, Tanabe K,
Ikari Y, Morino Y, Kadota K, Furukawa Y, Nakagawa Y, Kimura T. Monotherapy
after 1-month dual antiplatelet therapy in percutaneous coronary intervention:
from the STOPDAPT-2 total cohort. Circ Cardiovasc Interv 2022;15(8):e012004.
https://doi.org/10.1161/CIRCINTERVENTIONS.122.012004. Aug.
[59] Capodanno D, Huber K, Mehran R, Lip GYH, Faxon DP, Granger CB, Vranckx P,
Lopes RD, Montalescot G, Cannon CP, Ten Berg J, Gersh BJ, Bhatt DL,
Angiolillo DJ. Management of antithrombotic therapy in atrial brillation patients
undergoing PCI: JACC state-of-the-art review. J Am Coll Cardiol 2019;74(1):
8399. https://doi.org/10.1016/j.jacc.2019.05.016. Jul 9.
[60] De Caterina R, Agewall S, Andreotti F, Angiolillo DJ, Bhatt DL, Byrne RA, Collet JP,
Eikelboom J, Fanaroff AC, Gibson CM, Goette A, Hindricks G, Lip GYH, Potpara T,
Thiele H, Lopes RD, Galli M. Great debate: triple antithrombotic therapy in patients
with atrial brillation undergoing coronary stenting should be limited to 1 week.
Eur Heart J 2022;43(37):351227. https://doi.org/10.1093/eurheartj/ehac294.
Oct 7.
[61] Galli M, Andreotti F, DAmario D, Vergallo R, Montone RA, Niccoli G, Crea F.
Randomised trials and meta-analyses of double vs triple antithrombotic therapy for
atrial brillation-ACS/PCI: a critical appraisal. Int J Cardiol Heart Vasc 2020;28:
100524. https://doi.org/10.1016/j.ijcha.2020.100524. May 14.
[62] Lopes RD, Hong H, Harskamp RE, Bhatt DL, Mehran R, Cannon CP, Granger CB,
Verheugt FWA, Li J, Ten Berg JM, Sarafoff N, Gibson CM, Alexander JH. Safety and
efcacy of antithrombotic strategies in patients with atrial brillation undergoing
percutaneous coronary intervention: a network meta-analysis of randomized
controlled trials. JAMA Cardiol 2019;4(8):74755. https://doi.org/10.1001/
jamacardio.2019.1880. Aug 1.
[63] Gargiulo G, Goette A, Tijssen J, Eckardt L, Lewalter T, Vranckx P, Valgimigli M.
Safety and efcacy outcomes of double vs. triple antithrombotic therapy in patients
with atrial brillation following percutaneous coronary intervention: a systematic
review and meta-analysis of non-vitamin K antagonist oral anticoagulant-based
randomized clinical trials. Eur Heart J 2019;40(46):375767. https://doi.org/
10.1093/eurheartj/ehz732. Dec 7.
[64] Galli M, Andreotti F, Porto I, Crea F. Intracranial haemorrhages vs. stent
thromboses with direct oral anticoagulant plus single antiplatelet agent or triple
antithrombotic therapy: a meta-analysis of randomized trials in atrial brillation
and percutaneous coronary intervention/acute coronary syndrome patients.
Europace 2020;22(4):53846. https://doi.org/10.1093/europace/euz345. Apr 1.
[65] Laudani C, Greco A, Occhipinti G, Ingala S, Calderone D, Scalia L, Agnello F,
Legnazzi M, Mauro MS, Rochira C, Buccheri S, Mehran R, James S, Angiolillo DJ,
Capodanno D. Short duration of DAPT versus de-escalation after percutaneous
coronary intervention for acute coronary syndromes. JACC Cardiovasc Interv
2022;15(3):26877. https://doi.org/10.1016/j.jcin.2021.11.028. Feb 14.
P. Sabouret et al.
... Based on these observations, while aspirin remains the cornerstone for secondary CVD prevention [62], American guidelines do not endorse its use on a routine basis for primary prevention among adults over 70 years [63]. In particular, when prescribing dual antiplatelet treatment after ACS or PCI, it is pivotal to tailor its duration in order to maximize ischemic protection while limiting bleeding risk, even though this may be challenging due to an overlap between ischemic and bleeding risk in frail patients ( Figure 3) [60]. However, in both the PRECISE-DAPT and the DAPT scores, as well as according to the Academic Research Consortium for High Bleeding Risk (ARC-HBR) consensus, age is an important parameter that tips the scale towards short dual antiplatelet treatment regimens [44,64,65]. ...
... The optimization and dosage of all drugs is of utmost importance in older frail patients, with particular attention to antithrombotic agents, owing to the risk of side effects and drug interaction [59]. Age-related changes in pharmacokinetics and pharmacodynamics-potentially due to changes in the distribution of fat mass and lean mass, multimorbidity, and polypharmacy-are associated with an increased risk of drug toxicity and side effects in older patients [60]. Sarcopenia, for instance, may cause underestimation of glomerular filtration rate calculated using serum creatinine, leading to inappropriate direct oral anticoagulant (DOAC) dosing and increased risk of bleeding [61]. ...
... Based on these observations, while aspirin remains the cornerstone for secondary CVD prevention [62], American guidelines do not endorse its use on a routine basis for primary prevention among adults over 70 years [63]. In particular, when prescribing dual antiplatelet treatment after ACS or PCI, it is pivotal to tailor its duration in order to maximize ischemic protection while limiting bleeding risk, even though this may be challenging due to an overlap between ischemic and bleeding risk in frail patients (Figure 3) [60]. However, in both the PRECISE-DAPT and the DAPT scores, as well as according to the Academic Research Consortium for High Bleeding Risk (ARC-HBR) consensus, age is an important parameter that tips the scale towards short dual antiplatelet treatment regimens [44,64,65]. ...
Article
Full-text available
Coronary artery disease (CAD) is highly prevalent in older adults, yet its management remains challenging. Treatment choices are made complex by the frailty burden of older patients, a high prevalence of comorbidities and body composition abnormalities (e.g., sarcopenia), the complexity of coronary anatomy, and the frequent presence of multivessel disease, as well as the coexistence of major ischemic and bleeding risk factors. Recent randomized clinical trials and epidemiological studies have provided new data on optimal management of complex patients with CAD. However, frail older adults are still underrepresented in the literature. This narrative review aims to highlight the importance of assessing frailty as an aid to guide therapeutic decision-making and tailor CAD management to the specific needs of older adults, taking into account age-related pharmacokinetic and pharmacodynamic changes, polypharmacy, and potential drug interactions. We also discuss gaps in the evidence and offer perspectives on how best in the future to optimize the global strategy of CAD management in older adults.
... Collaboration between cardiologists and specialists across various disciplines is essential for delivering comprehensive, integrated care and follow-up ( Figure 3). Deprescribing unnecessary medications to streamline regimens to only those essential should be performed in older adults [79,80]. Beyond pharmacotherapy, the recommendation of aerobic exercises, exercise training, and diet-induced weight loss are also beneficial for this patient group [81]. ...
Article
Full-text available
Beta blockers (BBs) play a crucial role in enhancing the quality of life and extending the survival of patients with heart failure and reduced ejection fraction (HFrEF). Initiating the therapy at low doses and gradually titrating the dose upwards is recommended to ensure therapeutic efficacy while mitigating potential adverse effects. Vigilant monitoring for signs of drug intolerance is necessary, with dose adjustments as required. The management of older HF patients requires a case-centered approach, taking into account individual comorbidities, functional status, and frailty. Older adults, however, are often underrepresented in randomized clinical trials, leading to some uncertainty in management strategies as patients with HF in clinical practice are older than those enrolled in trials. The present article performs a scoping review of the past 25 years of published literature on BBs in older HF patients, focusing on age, outcomes, and tolerability. Twelve studies (eight randomized-controlled and four observational) encompassing 26,426 patients were reviewed. The results indicate that BBs represent a viable treatment for older HFrEF patients, offering benefits in symptom management, cardiac function, and overall outcomes. Their role in HF with preserved EF, however, remains uncertain. Further research is warranted to refine treatment strategies and address specific aspects in older adults, including proper dosing, therapeutic adherence, and tolerability.
... 20 However, the unpredictable clopidogrel efficacy of the 5%-55% non-responders limits its use 21 , while this rate is trivial in patients treated with prasugrel or ticagrelor. 22 There is no comprehensive information about resistance rate to clopidogrel in Iranian community. Only in one study on patients after coronary angioplasty, 24.76% resistance to clopidogrel was reported by using light transmission aggregometry in Iranian population. ...
Article
Background & Objectives: The risk of recurrence after a transient ischemic attack (TIA) or minor stroke is high especially within three months after first event. The aim of study is assessing the efficacy of ticagrelor plus aspirin in reduction of mild non-cardioembolic ischemic stroke or high risk TIA recurrence during first 3 months. Methods: This is a randomized, controlled, active comparator arm, outcome assessor blind, parallel group, feasibility study design on 90 patients with diagnosis of non-cardioembolic minor ischemic stroke or high risk TIA admitted in Bou-Ali Sina Hospital, Sari, Iran. After meeting all inclusion and exclusion criteria, patients will be randomized to ticagrelor 90 mg BID plus aspirin (ASA) 80 mg daily or clopidogrel 75 mg daily plus ASA 80 mg daily (1:1 ratio) until 21 days and then ASA 80 mg daily. Participants will be visited at month one and three. Any adverse events, serious side effects and outcome events will be recorded. The primary outcome is defined as ischemic stroke recurrence. Conclusion: Ticagrelor plus ASA is expected to be effective for prevention of recurrence in mild non-cardioembolic stroke and high risk TIA. Trial Registration: ClinicalTrials.gov: NCT04738097
... switching from prasugrel or ticagrelor to clopidogrel), DAPT shortening, and aspirin-free strategies after a short course of standard DAPT. 8 While recent data on antithrombotic and lipid-lowering treatments support an early and intensive treatment strategy, the current trend for antiplatelet agents is 'de-escalation' for most patients after the acute phase. Conversely, lipid lowering treatments should remain at high intensity and should even be rapidly reinforced by the addition of a proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitor for very high risk patients whose low-density-lipoprotein (LDL) cholesterol remains >70 mg/dL under a fixed combination of high-intensity statins and ezetimibe. ...
Article
Full-text available
To every complex problem, there is an answer that is clear, simple and wrong. H.L Mencken The mortality and morbidity associated with acute coronary syndromes (ACSs) have declined markedly over the past 50 years, mainly due to the introduction of the intensive care units, early reperfusion strategies, antithrombotic drugs, and lipid-lowering therapies. However, ACS still represents a major cause of mortality and morbidity worldwide. Furthermore, ACS-related mortality has remained relatively stable over the past decade, underlining the need for novel strategies to further improve prognosis in these patients. 1 Precision medicine, a medical model that proposes individualized customization of healthcare, as opposed to a 'one-drug-fits-all' approach, may represent a breakthrough for the implementation of treatments in ACS. Indeed, precision medicine is increasingly being adopted in several medical fields, such as oncology and hematology, but its use in cardiovascular (CV) medicine has not been as straightforward. Possible reasons behind this delay may lie in the fact that CV diseases have a high prevalence in the general population, and they appear, at least superficially, quite homogeneous. Furthermore, the need for an evidence-based approach generated by large randomized controlled trials has probably also played a role in hindering the development of individualized approaches for ACS.
Article
Full-text available
The synergistic blockade of the key platelet signaling pathways of cyclooxygenase-1 blockade and P2Y 12 signaling by combining aspirin plus a potent P2Y 12 inhibitor (prasugrel or ticagrelor), the so called dual antiplatelet treatment (DAPT), has represented the antithrombotic regimen of choice in patients with acute coronary syndrome (ACS) for nearly a decade. Nevertheless, the use of such antiplatelet treatment regimen, while reduced the risk of thrombotic complications, it is inevitably associated with increased bleeding and this risk may outweigh the benefit of a reduction of ischemic events in specific subgroup of patients. In light of the adverse prognostic implications of a bleeding complication, there has been a great interest in the development of antiplatelet regimens aimed at reducing bleeding without any trade-off in ischemic events. The fact that the ischemic risk is highest in the early phase after an ACS while the risk of bleeding remains relatively stable over time has represented the rationale for the implementation of a more intense antithrombotic regimen early after an ACS, followed by a less intense antithrombotic regimen thereafter. This practice, known as a “de-escalation” strategy, represents one of the more promising approaches for personalization of antithrombotic therapy in ACS. In this review we discuss the rationale, appraise the evidence and provide practical recommendations on the use of a de-escalation strategy of antiplatelet therapy in patients with an ACS.
Article
Full-text available
Percutaneous coronary intervention (PCI) is considered a relatively safe procedure associated with low rates of complications, but is inevitably associated with short and mid-to-long term increased bleeding risk. Besides the short term risk associated with the arterial access to perform PCI, enhanced bleeding risk persists for several months, given the need for antithrombotic therapy to prevent procedure-related thrombotic complications as well as ischemic recurrences. Bleeding is a powerful harbinger of adverse outcomes. This awareness has fuelled intense research on bleeding reduction strategies, including new PCI devices and techniques as well as new medications and antithrombotic regimens. We here review the mechanisms and prevalence of bleeding in PCI patients, discuss the available evidence from a practical point of view, and explore future perspectives on how to treat and prevent bleeding complications in these patients.
Article
Full-text available
Precision medicine is a medical model that proposes the customization of medical treatments to the individual patient, as opposed to a one-drug-fits-all model. Such a “personalized medicine” approach has been widely adopted in several medical fields, such as cancer medicine, but the implementation of precision medicine in cardiovascular medicine has not been similarly straightforward. Because pharmacogenomics plays an important role in the safety and efficacy of cardiovascular drug therapy, there has been a great interest in the use of tools aiming at personalizing antiplatelet therapy. Moreover, antiplatelet therapy is essential for the treatment of cardiovascular patients to reduce the risk of thrombotic complications, particularly those undergoing percutaneous coronary intervention, but it is inevitably associated with increased bleeding risk. In this review, the authors discuss the rationale, summarize the evidence and discuss the current and future directions for the personalization of antiplatelet treatment regimens in patients undergoing percutaneous coronary intervention.
Article
Full-text available
Open in new tabDownload slide Key points in favour or against the use of one-week triple antithrombotic therapy for the majority of patients with AF and ACS/PCI. Abbreviations: TT, triple antithrombotic therapy; OAC, oral anticoagulant; DAPT, dual antiplatelet therapy; RCT, randomised controlled trials; ACS, acute coronary syndrome; PCI, percutaneous coronary intervention.
Article
Full-text available
Background The benefit of clopidogrel monotherapy after 1-month dual antiplatelet therapy (DAPT) compared with 12-month DAPT with aspirin and clopidogrel was demonstrated in the STOPDAPT-2 (Short and Optimal Duration of Dual Antiplatelet Therapy After Everolimus-Eluting Cobalt-Chromium Stent-2), but not in the STOPDAPT-2 acute coronary syndrome (ACS); however, both trials were underpowered based on the actual event rates. Methods We obtained the prespecified pooled population of 5997 patients as the STOPDAPT-2 total cohort (STOPDAPT-2: N=3009/STOPDAPT-2 ACS: N=2988; ACS: N=4136/chronic coronary syndrome [CCS]: N=1861), comprising 2993 patients assigned to 1-month DAPT followed by clopidogrel monotherapy, and 3004 patients assigned to 12-month DAPT with aspirin and clopidogrel after percutaneous coronary intervention. The primary end point was the composite of cardiovascular (cardiovascular death, myocardial infarction, definite stent thrombosis, or any stroke) or bleeding (Thrombolysis in Myocardial Infarction major/minor) end points at 1 year. Results One-month DAPT was noninferior to 12-month DAPT for the primary end point (2.84% versus 3.04%; hazard ratio [HR], 0.94 [95% CI, 0.70–1.27]; P noninferiority =0.001; P superiority =0.68). There was no significant risk-difference for the cardiovascular end point between the 1- and 12-month DAPT groups (2.40% versus 1.97%; HR, 1.24 [95% CI, 0.88–1.75]; P noninferiority =0.14; P superiority =0.23). There was a lower risk of the bleeding end point with 1-month DAPT relative to 12-month DAPT (0.50% versus 1.31%; HR, 0.38 [95% CI, 0.21–0.70]; P superiority =0.002). One-month DAPT relative to 12-month DAPT was associated with a lower risk for major bleeding regardless of ACS or CCS (ACS: HR, 0.46 [95% CI, 0.23–0.94]; P =0.03, and CCS: HR, 0.26 [95% CI, 0.09–0.79]; P =0.02; P interaction =0.40), while it was associated with a numerical increase in cardiovascular events in ACS patients, but not in CCS patients, although not statistically significant and without interaction (ACS: HR, 1.50 [95% CI, 0.99–2.27]; P =0.053, and CCS: HR, 0.74 [95% CI, 0.38–1.45]; P =0.39; P interaction =0.08). Conclusions Clopidogrel monotherapy after 1-month DAPT compared with 12-month DAPT with aspirin and clopidogrel had a benefit in reducing major bleeding events without being associated with increase in cardiovascular events. Registration URL: https://www.clinicaltrials.gov ; Unique identifiers: NCT02619760, NCT03462498.
Article
Full-text available
Antiplatelet therapy is key to reducing local thrombotic complications and systemic ischaemic events among patients undergoing percutaneous coronary interventions (PCI), but it is inevitably associated with increased bleeding. The continuous refinement in stent technologies, together with the high incidence of ischaemic recurrences after PCI and the understanding of prognostic implications associated with bleeding, have led to a substantial evolution in antiplatelet treatment regimens over the past decades. Numerous investigations have been conducted to better stratify patients undergoing PCI according to their ischaemic and bleeding risks and to implement antithrombotic regimens accordingly. Evidence from these investigations have resulted in a number of antithrombotic treatment options as recommended by recent guidelines. In this State-of-the-Art review we provide the rationale, summarise the evidence, and discuss current and future directions of antiplatelet treatment regimens after PCI.
Article
Importance: Clopidogrel monotherapy after short dual antiplatelet therapy (DAPT) after percutaneous coronary intervention (PCI) has not yet been fully investigated in patients with acute coronary syndrome (ACS). Objective: To test the hypothesis of noninferiority of 1 to 2 months of DAPT compared with 12 months of DAPT for a composite end point of cardiovascular and bleeding events in patients with ACS. Design, setting, and participants: This multicenter, open-label, randomized clinical trial enrolled 4169 patients with ACS who underwent successful PCI using cobalt-chromium everolimus-eluting stents at 96 centers in Japan from December 2015 through June 2020. These data were analyzed from June to July 2021. Interventions: Patients were randomized either to 1 to 2 months of DAPT followed by clopidogrel monotherapy (n = 2078) or to 12 months of DAPT with aspirin and clopidogrel (n = 2091). Main outcomes and measures: The primary end point was a composite of cardiovascular (cardiovascular death, myocardial infarction [MI], any stroke, or definite stent thrombosis) or bleeding (Thrombolysis in MI major or minor bleeding) events at 12 months, with a noninferiority margin of 50% on the hazard ratio (HR) scale. The major secondary end points were cardiovascular and bleeding components of the primary end point. Results: Among 4169 randomized patients, 33 withdrew consent. Of the 4136 included patients, the mean (SD) age was 66.8 (11.9) years, and 856 (21%) were women, 2324 (56%) had ST-segment elevation MI, and 826 (20%) had non-ST-segment elevation MI. A total of 4107 patients (99.3%) completed the 1-year follow-up in June 2021. One to 2 months of DAPT was not noninferior to 12 months of DAPT for the primary end point, which occurred in 65 of 2058 patients (3.2%) in the 1- to 2-month DAPT group and in 58 of 2057 patients (2.8%) in the 12-month DAPT group (absolute difference, 0.37% [95% CI, -0.68% to 1.42%]; HR, 1.14 [95% CI, 0.80-1.62]; P for noninferiority = .06). The major secondary cardiovascular end point occurred in 56 patients (2.8%) in the 1- to 2-month DAPT group and in 38 patients (1.9%) in the 12-month DAPT group (absolute difference, 0.90% [95% CI, -0.02% to 1.82%]; HR, 1.50 [95% CI, 0.99-2.26]). The major secondary bleeding end point occurred in 11 patients (0.5%) in the 1- to 2-month DAPT group and 24 patients (1.2%) in the 12-month DAPT group (absolute difference, -0.63% [95% CI, -1.20% to -0.06%]; HR, 0.46 [95% CI, 0.23-0.94]). Conclusions and relevance: In patients with ACS with successful PCI, clopidogrel monotherapy after 1 to 2 months of DAPT failed to attest noninferiority to standard 12 months of DAPT for the net clinical benefit with a numerical increase in cardiovascular events despite reduction in bleeding events. The directionally different efficacy and safety outcomes indicate the need for further clinical trials. Trial registration: ClinicalTrials.gov Identifiers: NCT02619760 and NCT03462498.
Article
Objectives The aim of this study was to compare short dual antiplatelet therapy (DAPT) and de-escalation in a network meta-analysis using standard DAPT as common comparator. Background In patients with acute coronary syndrome (ACS) undergoing percutaneous coronary intervention (PCI), shortening DAPT and de-escalating to a lower potency regimen mitigate bleeding risk. These strategies have never been randomly compared. Methods Randomized trials of DAPT modulation strategies in patients with ACS undergoing PCI were identified. All-cause death was the primary outcome. Secondary outcomes included net adverse cardiovascular events (NACE), major adverse cardiovascular events, and their components. Frequentist and Bayesian network meta-analyses were conducted. Treatments were ranked on the basis of posterior probability. Sensitivity analyses were performed to explore sources of heterogeneity. Results Twenty-nine studies encompassing 50,602 patients were included. The transitivity assumption was fulfilled. In the frequentist indirect comparison, the risk ratio (RR) for all-cause death was 0.98 (95% CI: 0.68-1.43). De-escalation reduced the risk for NACE (RR: 0.87; 95% CI: 0.70-0.94) and increased major bleeding (RR: 1.54; 95% CI: 1.07-2.21). These results were consistent in the Bayesian meta-analysis. De-escalation displayed a >95% probability to rank first for NACE, myocardial infarction, stroke, stent thrombosis, and minor bleeding, while short DAPT ranked first for major bleeding. These findings were consistent in node-split and multiple sensitivity analyses. Conclusions In patients with ACS undergoing PCI, there was no difference in all-cause death between short DAPT and de-escalation. De-escalation reduced the risk for NACE, while short DAPT decreased major bleeding. These data characterize 2 contemporary strategies to personalize DAPT on the basis of treatment objectives and risk profile.
Article
Dual antiplatelet therapy (DAPT) represents the standard of care for patients undergoing percutaneous coronary intervention (PCI). Increasing evidence indicates that a “one-size-fits-all” approach with the use of a standard DAPT regimen for all patients undergoing PCI could lead to either suboptimal efficacy or prohibitively high bleeding in specific cohorts of patients. Moreover, the broad interindividual variability in response to P2Y12 inhibitors can impact outcomes and resource utilization. Among the strategies proposed to provide a more balanced trade-off between bleeding and ischemic events at a single patient level, a guided selection of P2Y12 inhibitors, by using platelet function or genetic testing, has shown promising results. In this review, we provide a focused summary of the rationale and evidence on the use of platelet function and genetic testing-guided antiplatelet therapy, and we explore the implications for their use in the modern setting of patients undergoing PCI.
Article
Aims Guidelines recommend the use of potent P2Y12 inhibitors over clopidogrel for the reduction of ischaemic events in patients with acute coronary syndrome (ACS). However, this comes at the expense of increased bleeding. A guided selection of P2Y12 inhibiting therapy has the potential to overcome this limitation. We aimed at evaluating the comparative safety and efficacy of guided vs. routine selection of potent P2Y12 inhibiting therapy in patients with ACS. Methods and results We performed a network meta-analysis of randomized controlled trials (RCTs) comparing different oral P2Y12 inhibitors currently recommended for the treatment of patients with ACS (clopidogrel, prasugrel, and ticagrelor). RCTs including a guided approach (i.e. platelet function or genetic testing) vs. standard selection of P2Y12 inhibitors among patients with ACS were also included. Incidence rate ratios (IRR) and associated 95% confidence intervals (CIs) were estimated. P-scores were used to estimate hierarchies of efficacy and safety. The primary efficacy endpoint was major adverse cardiovascular events (MACE) and the primary safety endpoint was all bleeding. A total of 61 898 patients from 15 RCTs were included. Clopidogrel was used as reference treatment. A guided approach was the only strategy associated with reduced MACE (IRR: 0.80, 95% CI: 0.65–0.98) without any significant trade-off in all bleeding (IRR: 1.22, 95% CI: 0.96–1.55). A guided approach and prasugrel were associated with reduced myocardial infarction. A guided approach, prasugrel, and ticagrelor were associated with reduced stent thrombosis. Ticagrelor was also associated with reduced total and cardiovascular mortality. Prasugrel was associated with increased major bleeding. Prasugrel and ticagrelor were associated with increased minor bleeding. The incidence of stroke did not differ between treatments. Conclusion In patients with an ACS, compared with routine selection of potent P2Y12 inhibiting therapy (prasugrel or ticagrelor), a guided selection of P2Y12 inhibiting therapy is associated with the most favourable balance between safety and efficacy. These findings support a broader adoption of guided approach for the selection of P2Y12 inhibiting therapy in patients with ACS. Study registration number This study is registered in PROSPERO (CRD42021258603). Key Question A guided selection of P2Y12 inhibiting therapy using platelet function or genetic testing improves outcomes among patients undergoing percutaneous coronary intervention. Nevertheless, the comparative safety and efficacy of a guided versus routine selection of potent P2Y12-inhibiting therapy in acute coronary syndrome has not been explored. Key Finding In a comprehensive network meta-analysis including the totality of available evidence and using clopidogrel as treatment reference, a guided approach was the only strategy associated with reduced major adverse cardiovascular events without any significant trade-off in bleeding. Prasugrel and ticagrelor increased bleeding and only ticagrelor reduced mortality. Take Home Message A guided selection of P2Y12-inhibiting therapy represents the strategy associated with the most favourable balance between safety and efficacy. These findings support a broader adoption of guided P2Y12 inhibiting therapy in patients with acute coronary syndrome.