You are on page 1of 8

European Heart Journal (2022) 43, 5037–5044 CLINICAL RESEARCH

https://doi.org/10.1093/eurheartj/ehac402 Clinical trials

Patient selection for long-term secondary


prevention with ticagrelor: insights from

Downloaded from https://academic.oup.com/eurheartj/article/43/48/5037/6823782 by European Society of Cardiology user on 28 May 2023


PEGASUS-TIMI 54
Marc P. Bonaca 1*, KyungAh Im 2, Giulia Magnani 3, Sameer Bansilal4,
Mikael Dellborg 5, Robert F. Storey 6, Deepak L. Bhatt 2, P. Gabriel Steg7,
Marc Cohen8, Per Johanson 9, Eugene Braunwald 2, and Marc S. Sabatine2*
1
Department of Cardiology and Vascular Medicine, University of Colorado School of Medicine, 2115 N Scranton St Suite 2040, Aurora, CO 80045, USA; 2TIMI Study Group, Division of
Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA; 3Parma University Hospital, Parma, Italy; 4Bayer, Whippany, New Jersey, USA;
5
Department of Medicine/Östra, Sahlgrenska University Hospital, Göteborg, Sweden; 6Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Beech Hill
Road, Sheffield S10 2RX, UK; 7Université Paris-Cité, INSERM U-1148 and AP-HP, Hôpital Bichat, FACT (French Alliance for Cardiovascular Trials) Paris, France; 8Newark Beth Israel
Medical Center, Rutgers Medical School, Newark, New Jersey, USA; and 9AstraZeneca, Gothenburg, Sweden

Received 21 September 2021; revised 20 April 2022; accepted 12 July 2022; online publish-ahead-of-print 11 November 2022

See the editorial comment for this article ‘Platelet antiaggregation after an acute coronary syndrome: what about the elderly?’, by
K. Huber, https://doi.org/10.1093/eurheartj/ehac405.

Abstract

Aim In patients with prior myocardial infarction (MI) on aspirin, the addition of ticagrelor reduces ischaemic risk but increases
bleeding risk. The simultaneous assessment of baseline ischaemic and bleeding risk may assist clinicians in selecting pa­
tients who are most likely to have a favourable risk/benefit profile with long-term ticagrelor.
.........................................................................................................................................................................................
Methods PEGASUS-TIMI 54 randomized 21 162 prior MI patients, 13 956 of which to the approved 60 mg dose or placebo and
and results who had all necessary data. The primary efficacy endpoint was cardiovascular death, MI, or stroke, and the primary safety
outcome was TIMI major bleeding; differences in Kaplan–Meier event rates at 3 years are presented. Post-hoc subgroups
based on predictors of bleeding and ischaemic risk were merged into a selection algorithm. Patients were divided into
four groups: those with a bleeding predictor (n = 2721, 19%) and then those without a bleeding predictor and either 0–1
ischaemic risk factor (IRF; n = 3004, 22%), 2 IRF (n = 4903, 35%), or ≥3 IRF (n = 3328, 24%). In patients at high bleeding
risk, ticagrelor increased bleeding [absolute risk difference (ARD) +2.3%, 95% confidence interval (CI) 0.6, 3.9] and did
not reduce the primary efficacy endpoint (ARD +0.08%, 95% CI −2.4 to 2.5). In patients at low bleeding risk, the ARDs in
the primary efficacy endpoint with ticagrelor were −0.5% (−2.2, 1.3), −1.5% (−3.1, 0.02), and −2.6% (−5.0, −0.24, P =
0.03) in those with ≤1, 2, and 3 risk factors, respectively (P = 0.076 for trend across groups). There were significant
trends for greater absolute risk reductions for cardiovascular death (P-trend 0.018), all-cause mortality (P-trend
0.027), and net outcomes (P-trend 0.037) with ticagrelor across these risk groups.
.........................................................................................................................................................................................
Conclusion In a post-hoc exploratory analysis of patients with prior MI, long-term ticagrelor therapy appears to be best suited for
those with prior MI with multiple IRFs at low bleeding risk.
.........................................................................................................................................................................................
Clinical Trial NCT01225562 ClinicalTrials.gov
Registration

* Corresponding authors. Tel: +1 303 860 9900, Email: marc.bonaca@cpcmed.org (M.P.B.); Tel: +1 617 278 0091, Fax: +1 617 734 7329, Email: msabatine@bwh.harvard.edu (M.S.S.)
© The Author(s) 2022. Published by Oxford University Press on behalf of European Society of Cardiology. All rights reserved. For permissions, please e-mail:
journals.permissions@oup.com
5038 M.P. Bonaca et al.

Structured Graphical Abstract

Downloaded from https://academic.oup.com/eurheartj/article/43/48/5037/6823782 by European Society of Cardiology user on 28 May 2023


identified a gradient of absolute risk reductions in the primary and other efficacy
endpoints.

Characteristics (left) and distribution of patients (middle) in PEGASUS-TIMI 54 by categories of high bleeding risk, low bleeding risk with 0–1
ischaemic risk factors, low bleeding risk with 2 ischaemic risk factors, and low bleeding risk with ≥ 3 ischaemic risk factors and the absolute
risk difference for the composite of cardiovascular death, myocardial infarction or stroke with ticagrelor vs. placebo over 3 years for each group
(right). CAD, coronary artery disease; CV, cardiovascular; MI, myocardial infarction.
.........................................................................................................................................................................................
Keywords Long-term ticagrelor • Myocardial infarction • Net benefit
Patient selection for long-term secondary prevention with ticagrelor 5039

Introduction long-term ticagrelor use in patients with an MI at least 1 year prior,


the population was restricted to the 14 112 randomized to the approved
Patients with myocardial infarction (MI) are at heightened risk of is­ dose of 60 mg twice daily or placebo. A total of 156 patients were ex­
chaemic events even years after the event occurred. Intensive anti­ cluded from the analyses due to missing ischaemic and/or bleeding risk
factors with 13 956 included in the analysis cohort.
platelet therapy with the combination of aspirin and a P2Y12
inhibitor (dual antiplatelet therapy or DAPT) is initiated at the time
of MI to reduce this risk and is indicated for at least a year in patients
Endpoints
who are not at elevated bleeding risk.1–3 Long-term use of DAPT has The primary efficacy endpoint was MACE, consisting of the composite of

Downloaded from https://academic.oup.com/eurheartj/article/43/48/5037/6823782 by European Society of Cardiology user on 28 May 2023


been shown to reduce ischaemic risk in patients with prior MI and cardiovascular (CV) death, MI, or stroke.4 The primary safety endpoint
PEGASUS-TIMI 54 prospectively demonstrated that long-term ticagre­ was TIMI major bleeding defined as any ICH, or clinically overt signs of
lor at a dose of 60 mg twice daily reduces major adverse cardiovascular haemorrhage associated with a reduction in haemoglobin ≥5 g/dL (or,
events (MACEs) in this population.4 The benefit of this therapeutic when haemoglobin was not available, a fall in haematocrit ≥15%), or fatal
approach has also extended to patients with diabetes and coronary bleeding (a bleeding event that directly led to death within 7 days). TIMI
disease who have never had MI, especially in those with prior percutan­ minor bleeding was defined as any clinically overt haemorrhage (including
eous coronary revascularization.5,6 that detected by imaging) that was associated with a fall in haemoglobin
Strategies using long-term DAPT to reduce ischaemic risk after MI, of 3 to <5 g/dL (or, when haemoglobin was not available, a fall in haem­
atocrit of 9 to <15%). Haemoglobin measurements were adjusted for
however, also increase the risk of bleeding.4,7 Although this bleeding
any packed red blood cells or whole blood given between baseline and
risk did not extend to irreversible harm events such as intracranial
post-transfusion measurement; transfusion of one unit of blood was as­
haemorrhage (ICH) or fatal bleeding in the trials, major bleeding is sumed to result in an increase of 1 g/dL of haemoglobin. Bleeding events
an undesirable event and can lead to hospitalization and cessation leading directly to mortality were classified as fatal bleeding events. All
of beneficial therapies and is associated with worse outcomes includ­ bleeding events, as well as their relationship to mortality, were adjudi­
ing mortality.1,3,8–10 Such observations necessitate personalized appli­ cated by a Clinical Events Committee blinded to treatment allocation.
cation of these strategies rather than generalized use in all patients A net clinical benefit analysis was defined as a composite outcome con­
with prior MI.1–3,11 Although algorithms have been developed to en­ sisting of CV death, MI, stroke, ICH, or fatal bleeding.
able selection of patients at lower bleeding risk, this approach is com­
plicated by the observation that many predictors of bleeding are also
predictors of ischaemic risk.8,11 In addition, other scoring systems de­ Patient selection algorithm
veloped in broader populations immediately after percutaneous cor­ A post-hoc algorithm conceptually aligned with the risk stratification ap­
proach described by the European Society of Cardiology (ESC) guidelines
onary intervention (PCI) may not apply in long-term patients with
for a second antithrombotic therapy was developed, including first identi­
prior MI some of whom were treated medically and where patient
fying high-bleeding risk patients and then stratifying low-bleeding risk pa­
characteristics and comorbidities are likely more important drivers tients into thrombotic risk categories (one risk criteria and two or more
of risk relative to stent or procedural characteristics.11–13 risk criteria).2 Previously identified independent predictors of bleeding,
Therefore, we developed a stepwise patient selection algorithm but not ischaemic risk, were anaemia at baseline and prior history of spon­
for long-term treatment with ticagrelor 60 mg twice daily utilizing taneous bleeding requiring hospitalization.8 Factors that were not inde­
subgroups previously observed to be at higher bleeding risk8 and is­ pendent predictors of bleeding or were predictors of both ischaemic
chaemic risk.14–19 We then evaluated bleeding, ischaemic risk, mor­ risk and bleeding (e.g. age, kidney dysfunction) were excluded as published
tality, and net outcomes with ticagrelor 60 mg twice daily vs. placebo previously.8 Patients with at least one of these criteria had three-fold high­
by risk group in PEGASUS-TIMI 54. er rates of TIMI major or minor bleeding with ticagrelor 60 mg vs. placebo
compared with those with neither feature.8
The initial step in the stratification approach, therefore, was to exclude
Methods patients at high bleeding risk, defined by the presence of either or both of
the bleeding risk predictors. Patients identified as low bleeding risk were
Study population stratified according to ischaemic risk factors. Because PEGASUS-TIMI 54
The PEGASUS-TIMI 54 trial described previously, randomized 21 162 included medically managed patients (for whom complexity of coronary
patients with a prior history of spontaneous MI occurring 1–3 years prior disease was unknown), an approach based solely on the number of clinical
to enrolment, who had at least one additional atherothrombotic risk fac­ risk factors was utilized. Factors included the pre-specified enrichment
tor (age ≥65 years, diabetes mellitus requiring medication, a second prior criteria for the trial that have also been observed to be associated with
spontaneous MI, chronic renal dysfunction, or multivessel coronary ar­ greater ischaemic risk (see Supplementary material online, Figure S2) in­
tery disease) to ticagrelor 60 mg b.i.d., ticagrelor 90 mg b.i.d., or placebo, cluding type 2 diabetes mellitus,14 non-end-stage chronic kidney disease,17
all on a background of low-dose (75–150 mg) aspirin.4 Patients were fol­ multivessel coronary disease,18 and multiple prior MIs. In addition, more
lowed for a median of 33 months. Exclusion criteria included planned use recent MI or P2Y12 inhibition which showed formal heterogeneity for
of a P2Y12 receptor antagonist or anticoagulant therapy, a known bleed­ benefit of ticagrelor and was the basis for the EU label19 and polyvascular
ing disorder, history of stroke, a central nervous system tumour, gastro­ disease (comorbid peripheral artery disease) which was associated with
intestinal bleeding within the previous 6 months, or major surgery within greater risk and more favourable risk benefit were included.16,20 A sum­
the previous 30 days. Enrolling sites were requested to indicate if there mary of risk factors chosen, criteria and rationale for exclusion are shown
was any history of bleeding leading to hospitalization. All patients had in Supplementary material online, Table S1. Low-bleeding risk patients
central laboratory testing for haemoglobin at baseline. Anaemia was de­ were then grouped according to 0–1 ischaemic risk factor, 2 risk ischae­
fined as a haemoglobin ≤13.5 g/dL for men and ≤12.0 g/dL for women.8 mic factors, and ≥3 ischaemic risk factors in general alignment with lower
Because this analysis is focused on an algorithm to select patients for and higher ischaemic risk, respectively.1
5040 M.P. Bonaca et al.

Statistics low bleeding risk (regardless of ischaemic risk factors): +2.3% [95% con­
As per the pre-specified statistical analysis plan, the primary efficacy ana­ fidence interval (CI) 0.60–3.9] vs. +1.0% (95% CI 0.5, 1.5) (P = 0.25). The
lysis and net clinical outcome were conducted on an intention-to-treat ARDs in bleeding across ischaemic risk groups among patients with low
basis, whereas safety analyses included all patients who underwent ran­ bleeding risk was similar (P-trend for ARD 0.96).
domization and received at least one dose of study drug. Event rates for Drug discontinuation rates at 3 years were higher in those at high
ticagrelor and placebo were estimated by Kaplan–Meier methods from bleeding risk vs. those at low bleeding risk both in the placebo arm
baseline to 3 years and compared with the log-rank test. This was exam­
(26.5% in high bleeding risk, 22.7% in low bleeding risk and 0–1 ischae­
ined separately by treatment arm over time. Categorical variables were
mic risk factors, 20.8% in low bleeding risk and 2 ischaemic risk factors,
compared using χ2 tests and continuous variable with either a t test or

Downloaded from https://academic.oup.com/eurheartj/article/43/48/5037/6823782 by European Society of Cardiology user on 28 May 2023


and 25.9% in low bleeding risk and ≥ 3 ischaemic risk factors) and the
Wilcoxon rank-sum test, as appropriate. The hazard ratio (HR) for given
outcome of interest between ticagrelor and placebo was examined using ticagrelor arm (33.8, 30.3, 28.9, and 33.7%, respectively).
a Cox regression model with treatment as a model term in each risk
stratification group defined by bleeding and ischaemic risk profiles de­ Efficacy with ticagrelor 60 mg vs. placebo
scribed in previous section. The proportional hazards assumption was
examined and tested by scaled Schoenfeld residuals. In addition, the ab­
by risk group
The relative reduction in the risk of MACE with ticagrelor was 2%
solute risk differences (ARDs) between ticagrelor and placebo groups
were calculated by subtracting the cumulative incidence in the placebo (HR 0.98, 95% CI 0.77–1.26, P = 0.88), 13% (HR 0.87, 95% CI
group from that of the treatment group. The cumulative incidence was 0.63–1.22, P = 0.42), 19% (HR 0.81, 95% CI 0.64–1.01, P = 0.062),
defined by the complement of the Kaplan–Meier survival estimates. and 23% (HR 0.77, 95% CI 0.62–0.95, P = 0.013; Figure 1A) in patients
The null hypothesis that the gradient of risk differences across the level at high bleeding risk and at low bleeding risk with 0–1, 2, and ≥3 risk
of subgroup is zero was tested by weighted least square regression, with factors, respectively. Furthermore, the ARDs in the primary end­
weights being the inverse of variance.21 A sensitivity analysis was also per­ point of MACE at 3 years in those groups were +0.08% (95% CI
formed estimating the hypothetical ARDs for the primary endpoint by −2.39, 2.54), −0.47% (95% CI −2.21, 1.26), −1.53% (95% CI −3.1,
applying the overall HR to the risk group specific baseline risk. All statis­ 0.024), and −2.61% (95% CI −4.99, −0.24) with a P-value for the trend
tical computations were performed with the use of SAS software, ver­
in ARD of 0.076 across the three groups. An alternative approach es­
sion 9.4 (SAS Institute, Cary, NC, USA).
timating the hypothetical ARDs for the primary endpoint in the groups
by applying the overall HR to the placebo event rate in each group is
Results shown in Supplementary material online, Table S3. In patients at high
bleeding risk, 2154 had 2 or more ischaemic risk factors and 549 had
Overall, of 14 112 patients randomized to ticagrelor 60 mg twice dai­ 0–1 ischaemic risk factors with a non-significant trend towards an
ly or placebo, a total of 2721 (19%) had at least one bleeding risk fac­ ∼10% reduction in risk in those with multiple risk factors (HR 0.90,
tor and were categorized as high bleeding risk. Among those without 95% CI 0.69–1.18) and no apparent benefit in those with 0–1 risk fac­
a bleeding predictor, 3004 (22% of the total) had only 0–1 ischaemic tors (HR 1.81, 95% CI 0.91–3.61; P-interaction 0.0714).
risk factor, 4903 (35%) had 2 ischaemic risk factors, and 3328 (24%) Likewise, the effect of ticagrelor on CV death varied across these
had 3 or more ischaemic risk factors. The baseline characteristics of four patient subgroups, with no benefit (HR 1.24, 95% CI 0.87–1.76,
these patients are shown in Table 1. When excluding the 156 patients ARD +1.0%, 95% CI −0.8, 2.8) in patients at high bleeding risk, a 19%
with missing ischaemic and/or bleeding risk factors there were no relative risk reduction (RRR; HR 0.81, 95% CI 0.46–1.42) and −0.2
consistent statistically significant imbalances in baseline characteris­ ARD (95% CI −1.3, +0.9, P = 0.46) in patients at low bleeding risk
tics between the placebo and ticagrelor arms across the three risk and 0–1 ischaemic risk factor, a 32% RRR (HR 0.68, 95% CI 0.45–
groups (see Supplementary material online, Tables S2A–C). 1.03, P = 0.070) and −0.86% ARD (95% CI −1.86, 0.13) in patients at
In the placebo arm, patients at high risk of bleeding had rates of low bleeding risk and 2 ischaemic risk factors, and a 36% RRR (HR
major bleeding at 3 years that were almost double those in the pa­ 0.64, 95% CI 0.44–0.93, P = 0.0020) and −1.62% ARD (95% CI
tients categorized as low bleeding risk (1.7 vs. 0.9%, P = 0.040, −3.12, −0.14, P-trend for HR 0.0083, P-trend for ARR 0.018,
Supplementary material online, Figure S1). With regard to ischaemic Figure 1B) in those with ≥ 3 ischaemic risk factors. Similar patterns
risk, in the placebo arm, the ischaemic risk characteristics were asso­ were seen for the effect of ticagrelor on all-cause mortality (high bleed­
ciated with higher rates of the primary endpoint at 3 years (see ing risk HR 1.14; 95% CI 0.86 to 1.50, ARD 1.46; 95% CI −0.85 to 3.78;
Supplementary material online, Figure S2). Overall application of low bleeding risk with 0–1 risk factor HR 0.90; 95% CI 0.59 to 1.38;
the algorithm categorized 19% of the population as high bleeding ARD −0.25; 95% CI −1.62 to 1.13; low bleeding risk with 2 risk factors
risk and 81% as low bleeding risk, with those categorized as 22% HR 0.83; 95% CI 0.60 to 1.15; ARD −0.53; 95% CI −1.69 to 0.63; low
with 0–1 ischaemic risk factors, 35% with 2 ischaemic risk factors, bleeding risk with 3 risk factors HR 0.71; 95% CI 0.52 to 0.96; ARD
and 24% with ≥ 3 ischaemic risk factors (Graphical abstract). −2.01; 95% CI −3.82 to −0.20; P-trend for HR 0.021, P-trend for
ARD 0.027, Figure 1C) as well as on coronary heart disease death, MI,
Safety with ticagrelor 60 mg vs. placebo and stroke (see Supplementary material online, Figure S3).

by risk group
Whereas the relative increase in bleeding risk with ticagrelor 60 mg Net outcomes with ticagrelor 60 mg
twice daily vs. placebo was consistent across the risk groups, due to their vs. placebo by risk group
higher baseline bleeding risk, there was an approximately two-fold high­ In terms of the net clinical outcome of CV death, MI, stroke, intracra­
er ARD in TIMI major bleeding in high-bleeding risk patients vs. those at nial bleeding, or fatal bleeding, there was no benefit in patients at high
Patient selection for long-term secondary prevention with ticagrelor 5041

Table 1 Baseline characteristics by bleeding and ischemic risk categories

High Low bleeding risk and Low bleeding risk and Low bleeding risk and P-value
bleeding risk ≤ 1 risk factor 2 risk factors ≥3 risk factors
N = 2721 N = 3004 N = 4903 N = 3328
.................................................................................................................................................................................
Age, years, median (IQR) 68.0 (62.0, 74.0) 67.0 (63.0, 72.0) 62.0 (57.0, 69.0) 64.0 (58.0, 71.0) <0.0001
Female sex, n (%) 571 (21.0) 760 (25.3) 1107 (22.6) 906 (27.2) <0.0001

Downloaded from https://academic.oup.com/eurheartj/article/43/48/5037/6823782 by European Society of Cardiology user on 28 May 2023


2
Body mass index, kg/m , median (IQR) 27.1 (24.2, 30.4) 27.6 (25.1, 30.2) 28.0 (25.3, 31.2) 28.7 (25.7, 32.2) <0.0001
Caucasian, n (%) 2153 (79.1) 2750 (91.5) 4280 (87.3) 2879 (86.5) <0.0001
Hypertension, n (%) 2168 (79.7) 2148 (71.5) 3696 (75.4) 2804 (84.3) <0.0001
Hypercholesterolemia, n (%) 2031 (74.6) 2225 (74.1) 3777 (77.0) 2681 (80.6) <0.0001
Current smoker, n (%) 312 (11.5) 454 (15.1) 941 (19.2) 614 (18.5) <0.0001
History of CHF, n (%) 616 (22.6) 560 (18.6) 863 (17.6) 759 (22.8) <0.0001
History of stroke or TIA, n (%) 57 (2.1) 46 (1.5) 55 (1.1) 64 (1.9) 0.0033
Prior angina, n (%) 889 (32.7) 862 (28.7) 1435 (29.3) 1158 (34.8) <0.0001
Prior CABG, n (%) 165 (6.1) 32 (1.1) 104 (2.1) 349 (10.5) <0.0001
History of PCI with stenting, n (%) 2207 (81.1) 2271 (75.6) 4178 (85.2) 2932 (88.1) <0.0001
Qualifying MI type, n (%) 2114 (78.1) 2182 (72.9) 4043 (82.7) 2818 (85.0) <0.0001
NSTEMI, n (%) 1393 (51.3) 1636 (54.5) 2793 (57.0) 1662 (50.0) <0.0001
STEMI, n (%) 1155 (42.6) 1158 (38.6) 1825 (37.3) 1482 (44.6)
Unknown, n (%) 166 (6.1) 209 (7.0) 279 (5.7) 180 (5.4)
Aspirin 75–100 mg at baseline, n (%) 2640 (97.2) 2918 (97.3) 4774 (97.5) 3238 (97.4) 0.8603
Statin use at baseline, n (%) 2497 (91.8) 2760 (91.9) 4578 (93.4) 3101 (93.2) 0.0124
Beta blocker use at baseline, n (%) 2185 (80.3) 2380 (79.2) 4144 (84.5) 2830 (85.0) <0.0001
ACE-I or ARB use at baseline, n (%) 2204 (81.0) 2315 (77.1) 3921 (80.0) 2757 (82.8) <0.0001
Risk Factors for Algorithm
Qualifying MI ≥2 years, n (%) 1038 (38.2) 1978 (65.9) 1446 (29.5) 901 (27.1) <0.0001
Time from last dose of P2Y12 inhibitor, 63.0 (1.0, 348.0) 425.0 (102.0, 618.5) 64.0 (1.0, 290.0) 39.0 (1.0, 223.0) <0.0001
days, median (IQR)
Multivessel disease, n (%) 1582 (58.2) 736 (24.5) 3191 (65.1) 2802 (84.2) <0.0001
History of diabetes, n (%) 1118 (41.1) 309 (10.3) 1183 (24.1) 1904 (57.2) <0.0001
Peripheral artery disease, n (%) 209 (7.7) 19 (0.6) 76 (1.6) 461 (13.9) <0.0001
eGFR <60 mL/min (MDRD), n (%) 946 (35.0) 152 (5.1) 662 (13.5) 1435 (43.1) <0.0001
Multiple prior MIs, n (%) 469 (17.2) 105 (3.5) 479 (9.8) 1280 (38.5) <0.0001

ACE-I, angiotensin-converting enzyme inhibitor; ARB, angiotensin receptor blocker; CABG, coronary artery bypass graft; CHF, congestive heart failure; eGFR, estimated glomerular
filtration rate; IQR, interquartile range; MDRD, Modification of Diet in Renal Disease; MI, myocardial infarction; NSTEMI, non-ST-elevation myocardial infarction; PCI, percutaneous
coronary intervention; STEMI, ST-elevation myocardial infarction; TIA, transient ischaemic attack.

bleeding risk (HR 1.03, 95% CI 0.81–1.31; ARD 0.56, 95% CI −1.95, Discussion
3.06) and a gradient of benefit in those at low bleeding risk by
whether they had 0–1 (HR 0.93, 95% CI 0.67–1.28; ARD −0.21, The current analysis provides novel observations regarding the appli­
95% CI −1.97, 1.56); 2 (HR 0.82, 95% CI 0.66–1.03; ARD −1.45, cation of long-term DAPT in patients with prior MI. First, a stepwise
95% CI −3.02, 0.12); or ≥3 ischaemic risk factors (HR 0.77, 95% approach based on the paradigm espoused by the most recent ESC
CI 0.62–0.94; ARD −2.64, 95% CI −5.03, −0.25). There was a con­ guidelines for selection of patients at lower bleeding risk and higher
sistent pattern for the primary outcome and net outcomes with tica­ thrombotic risk for additional antithrombotic therapy applied in a
grelor 90 mg vs. placebo in the three risk groups (see Supplementary trial of long-term ticagrelor in patients with prior MI identifies a sig­
material online, Table S4). nificant proportion of the population (∼60% of the PEGASUS-TIMI
5042 M.P. Bonaca et al.

A Relative Risk with Absolute Risk Difference B Relative Risk with Absolute Risk Difference
Ticagrelor vs. Placebo with Ticagrelor vs. Ticagrelor vs. Placebo with Ticagrelor vs.
by Risk Group Placebo by Risk Group by Risk Group Placebo by Risk Group
P-trend forhazard 3 Year KM Rate P-trend for absolute risk P-trend for hazard 3 Year KM Rate P-trend for absolute risk
Ticagrelor Placebo
0.1

-6

-5

-4

-3

-2

-1
Ticagrelor Placebo

0.1
ratio= 0.13 ratio =0.0083

10

-4

-3

-2

-1
10
difference 0.076 difference 0.018

3
1

1
0.98 0.08% 1.24 1.02%
High Bleeding Risk 10.42% 10.35% High Bleeding Risk 5.57% 4.55%
2721 (19%) 2721 (19%)

Low Bleeding Risk 0.87 -0.47% Low Bleeding Risk 0.81 -0.19%
≤ 1 Ischemic Risk Factor 4.98% 5.46% ≤ 1 Ischemic Risk Factor 1.79% 1.98%
3004 (22%) 3004 (22%)
Low Bleeding Risk 0.81 -1.53% Low Bleeding Risk 0.68 -0.86%
2 Ischemic Risk Factors 6.22
% 7.75% 2 Ischemic Risk Factors 1.91% 2.77%

Downloaded from https://academic.oup.com/eurheartj/article/43/48/5037/6823782 by European Society of Cardiology user on 28 May 2023


4903 (35%) 4903 (35%)

Low Bleeding Risk 0.77 -2.61% Low Bleeding Risk 0.64 -1.62%
3+ Ischemic Risk Factors 10.57% 13.18% 3+ Ischemic Risk Factors 3.05% 4.68%
3328 (24%) 3328 (24%)

0.5 0.8 1 1.5 -2.5 0 2.5 0.5 0.8 1 1.5 -3.0 -1.5 0 2.5

Ticagrelor 60 mg Placebo Ticagrelor 60 mg Placebo Ticagrelor 60 mg Placebo Ticagrelor 60 mg Placebo


twice daily Better Better twice daily Better Better twice daily Better Better twice daily Better Better

C Relative Risk with Absolute Risk Difference D Relative Risk with Absolute Risk Difference
Ticagrelor vs. Placebo with Ticagrelor vs. Ticagrelor vs. Placebo with Ticagrelor vs.
by Risk Group Placebo by Risk Group by Risk Group Placebo by Risk Group
P-trend for hazard 3 Year KM Rate P-trend for absolute risk P-trend for hazard 3 Year KM Rate P-trend for absolute risk
Ticagrelor Placebo Ticagrelor Placebo

-6

-5

-4

-3

-2

-1
ratio= 0.021 ratio= 0.057

0.1

4
10
0.1

difference 0.027 difference 0.037


10
-5

-4

-3

-2

-1

1
0

5
1

1.14 1.46 1.03 0.56%


High Bleeding Risk 9.11% 7.64% High Bleeding Risk 11.13% 10.57%
2721 (19%) 2721 (19%)

Low Bleeding Risk 0.90 -0.25 Low Bleeding Risk 0.93 -0.21%
≤ 1 Ischemic Risk Factor 2.96% 3.21% ≤ 1 Ischemic Risk Factor 5.32% 5.53%
3004 (22%) 3004 (22%)
Low Bleeding Risk 0.83 -0.53% Low Bleeding Risk 0.82 -1.45%
2 Ischemic Risk Factors 3.14% 3.67% 2 Ischemic Risk Factors 6.48% 7.93%
4903 (35%) 4903 (35%)

Low Bleeding Risk 0.71 -2.01% Low Bleeding Risk 0.77 -2.64%
3+ Ischemic Risk Factors 4.89% 6.90% 3+ Ischemic Risk Factors 10.79% 13.43%
3328 (24%) 3328 (24%)

0.5 0.8 1 1.5 -3.0 -1.0 0 1.00 0.5 0.8 1 1.5 -3.0 -1.5 0 3.0

Ticagrelor 60 mg Placebo Ticagrelor 60 mg Placebo Ticagrelor 60 mg Placebo Ticagrelor 60 mg Placebo


twice daily Better Better twice daily Better Better twice daily Better Better twice daily Better Better

Figure 1 Outcomes at 3 years with ticagrelor 60 mg twice daily vs. placebo by difference in hazard (left) and absolute risk difference (right) for
(A) primary outcome composite of cardiovascular death, myocardial infarction, or stroke, (B) cardiovascular death, (C) all-cause mortality, and
(D) net composite outcome of cardiovascular death, myocardial infarction, stroke, intracranial haemorrhage, or fatal bleeding.

54 population) that is at low bleeding risk and has two or more into two groups, higher thrombotic risk and lower thrombotic risk
thrombotic risk factors. Second, use of such an algorithm identifies (called moderate risk in the guideline).1 The current analysis evalu­
a population that derives greater absolute benefit, with less bleeding ates an approach conceptually aligned with this paradigm and incor­
and resulting favourable findings for CV death and net outcomes. porates the pre-specified risk criteria for the trial along with
Patients with prior MI have been described through a number of subgroup findings and the guideline characteristics to approximate
trials to be at long-term heightened risk of recurrent atherothrombo­ this approach. Use of this approach not only identified a lower risk
sis. More potent antiplatelet therapy reduces this risk but increases group for bleeding on ticagrelor but also identified a gradient of is­
bleeding requiring clinicians to personalize the approach, which ac­ chaemic risk within the low-bleeding risk group enabling further per­
knowledges heterogeneity in the population both for ischaemic and sonalization. This approach translated to greater absolute benefits
bleeding risk. Established risk scores have shown the ability to discrim­ for MACE, heterogeneity with lower rates of CV death and greater
inate ischaemic risk in broad atherosclerosis populations but have not net benefit in those at the highest ischaemic risk (at least 2 risk fac­
evaluated bleeding risk with P2Y12 inhibition, limiting utilization for tors). In fact, the 59% of the population at low bleeding risk and with
long-term ticagrelor. Scores developed to evaluate net outcomes either 2 or ≥3 ischaemic risk factors may be the optimal population
such as the DAPT score13 have utility at the time of stenting in broad for the use of long-term ticagrelor 60 mg. Although those at high
populations but do not necessarily translate to stable post-MI patients bleeding risk had ischaemic events, premature discontinuation of
where procedural aspects are less relevant to risk relative to clinical study treatment and potentially other background therapies may
characteristics and a significant proportion of patients have not had partially explain the attenuation of benefit. Future studies may help
PCI. More recently, scores like PRECISE-DAPT11 have shown the abil­ to further elucidate the exact reasons why those at high bleeding
ity to risk stratify in acute coronary syndrome (ACS) but are focused risk do not benefit from prolonged DAPT and explore how much
predominantly on bleeding rather than both bleeding and ischaemic any benefit is offset by ischaemic complications occurring in the con­
risk and have been derived in ACS or PCI populations. Therefore, text of bleeding events. Regardless, these observations support the
there is an unmet need for a patient selection algorithm for long-term guideline recommendation that long-term therapy should be consid­
ticagrelor in stable patients with a history of MI more than a year ago ered (Class IIa recommendation) in such patients and provide exten­
even if they have not been treated with PCI. sive data for outcomes and net benefit that may be useful to clinicians
The most recent ESC guidelines for non-ST-elevation ACS de­ in communicating risk/benefit to patients.
scribe an algorithm where patients at lower bleeding risk are first There are several limitations to this analysis. First, the data are de­
identified and then those at lower bleeding risk are categorized rived from a single data set and have not been validated externally;
Patient selection for long-term secondary prevention with ticagrelor 5043

however, there are few large data sets examining long-term ticagre­ Novartis, personal fees for lectures from Boehringer Ingelheim, Sanofi
lor in a randomized trial. Second, although not all factors outlined in and Bayer. Advisory Board: Amgen, Novo Nordisk, AstraZeneca,
the ESC guidelines were available for inclusion, the characteristics Novartis. Personal fees for serving on the Data Monitoring
identified for both bleeding risk and ischaemic risk have been identi­ Committee: for the DAPA-MI trial, funded by AstraZeneca. R.F.S. re­
ports institutional research grants/support from AstraZeneca,
fied as independent predictors in other data sets and scores.1 Third,
Cytosorbents, GlyCardial Diagnostics, and Thromboserin; and personal
there was no effect modification for relative benefit for the primary
fees from Alnylam, Amgen, AstraZeneca, Bayer, Bristol Myers Squibb/
endpoint by risk category; however, the goal of the analysis was to
Pfizer, Chiesi, CSL Behring, Cytosorbents, GlyCardial Diagnostics,
characterize ARDs that were strongly statistically significant. Hengrui, Idorsia, Intas Pharmaceuticals, Medscape, Novartis, PhaseBio,

Downloaded from https://academic.oup.com/eurheartj/article/43/48/5037/6823782 by European Society of Cardiology user on 28 May 2023


Fourth, the results are a post-hoc exploratory analysis and therefore Sanofi Aventis, and Thromboserin. D.L.B discloses the following relation­
should be considered hypothesis generating. Fifth, the ARDs were ships—Advisory Board: Bayer, Boehringer Ingelheim, Cardax,
calculated by using the subgroup specific rates which was felt to be CellProthera, Cereno Scientific, Elsevier Practice Update Cardiology,
more conservative in highlighting that the high bleeding risk group Janssen, Level Ex, Medscape Cardiology, Merck, MyoKardia, NirvaMed,
may derive lesser benefit. An alternative approach to using the ob­ Novo Nordisk, PhaseBio, PLx Pharma, Regado Biosciences, and Stasys;
served ARDs and instead applying the overall trial HR to the placebo Board of Directors: Bristol Myers Squibb (stock), Boston VA Research
event rate in each subgroup showed a more consistent absolute dif­ Institute, DRS.LINQ (stock options), Society of Cardiovascular Patient
Care, TobeSoft; Chair: Inaugural Chair, American Heart Association
ference between groups. Sixth, this study assigned equal weights to
Quality Oversight Committee; Data Monitoring Committees: Acesion
ischaemic risk factors included in this study and readers cannot infer a
Pharma, Assistance Publique-Hôpitaux de Paris, Baim Institute for
definitive threshold for number of ischaemic risk factors for net
Clinical Research (formerly Harvard Clinical Research Institute, for the
benefit; future studies should derive a scoring system using predictive PORTICO trial, funded by St Jude Medical, now Abbott), Boston
modelling methods. Finally, PEGASUS specifically excluded patients Scientific (Chair, PEITHO trial), Cleveland Clinic (including for the
at highest risk of bleeding (history of previous intracranial bleed at ExCEED trial, funded by Edwards), Contego Medical (Chair,
any time, gastrointestinal (GI) bleed within the past 6 months, or ma­ PERFORMANCE 2), Duke Clinical Research Institute, Mayo Clinic,
jor surgery within 30 days), and, like most randomized trials, enrolled Mount Sinai School of Medicine (for the ENVISAGE trial, funded by
a selected population that may not be entirely representative of Daiichi Sankyo; for the ABILITY-DM trial, funded by Concept Medical),
post-ACS patients encountered in routine clinical care.22 Further val­ Novartis, Population Health Research Institute; Rutgers University (for
idation in ‘real-world’ cohorts, including those with history of gastro­ the NIH-funded MINT Trial); Honoraria: American College of
Cardiology (Senior Associate Editor, Clinical Trials and News,
intestinal bleeding, will lead to understanding of performance in a
ACC.org; Chair, ACC Accreditation Oversight Committee), Arnold
broader population of patients.
and Porter law firm (work related to Sanofi/Bristol Myers Squibb clopi­
dogrel litigation), Baim Institute for Clinical Research (formerly Harvard
Conclusions Clinical Research Institute; RE-DUAL PCI clinical trial steering committee
funded by Boehringer Ingelheim; AEGIS-II executive committee funded
In conclusion, in stable patients with prior MI, a stepwise patient se­ by CSL Behring), Belvoir Publications (Editor in Chief, Harvard Heart
lection approach evaluating bleeding and ischaemic risk based on Letter), Canadian Medical and Surgical Knowledge Translation
post-hoc subgroups may be useful in identifying patients who may Research Group (clinical trial steering committees), Cowen and
derive greater net benefit with long-term ticagrelor. Specifically, Company, Duke Clinical Research Institute (clinical trial steering commit­
tees, including for the PRONOUNCE trial, funded by Ferring
those at lower bleeding risk with at least two ischaemic risk factors
Pharmaceuticals), HMP Global (Editor in Chief, Journal of Invasive
appeared to have the most favourable risk–benefit profile. This sim­
Cardiology), Journal of the American College of Cardiology (Guest
ple patient selection approach may be useful for clinicians identifying
Editor; Associate Editor), K2P (Co-Chair, interdisciplinary curriculum),
patients who may benefit from a strategy of long-term ticagrelor Level Ex, Medtelligence/ReachMD (CME steering committees), MJH
after MI. Life Sciences, Piper Sandler, Population Health Research Institute (for
the COMPASS operations committee, publications committee, steering
committee, and USA national co-leader, funded by Bayer), Slack
Supplementary material Publications (Chief Medical Editor, Cardiology Today’s Intervention),
Supplementary material is available at European Heart Journal online. Society of Cardiovascular Patient Care (Secretary/Treasurer), WebMD
(CME steering committees), Wiley (steering committee); Other:
Clinical Cardiology (Deputy Editor), NCDR-ACTION Registry
Funding Steering Committee (Chair), VA CART Research and Publications
This study was supported by a grant to Brigham and Women’s Hospital
Committee (Chair); Research Funding: Abbott, Afimmune, Aker
from AstraZeneca.
Biomarine, Amarin, Amgen, AstraZeneca, Bayer, Beren, Boehringer
Conflict of interest: The TIMI Study Group has received significant Ingelheim, Bristol Myers Squibb, Cardax, CellProthera, Cereno
research grant support from Abbott, Amgen, Aralez, AstraZeneca, Scientific, Chiesi, CSL Behring, Eisai, Ethicon, Faraday Pharmaceuticals,
Bayer HealthCare Pharmaceuticals, Inc., BRAHMS, Daiichi Sankyo, Ferring Pharmaceuticals, Forest Laboratories, Fractyl, Garmin, HLS
Eisai, GlaxoSmithKline, Intarcia, Janssen, MedImmune, Merck, Novartis, Therapeutics, Idorsia, Ironwood, Ischemix, Janssen, Javelin, Lexicon,
Pfizer, Poxel, Quark Pharmaceuticals, Roche, Takeda, The Medicines Lilly, Medtronic, Merck, Moderna, MyoKardia, NirvaMed, Novartis,
Company, Zora Biosciences. M.P.B. reports grant support to CPC clinical Novo Nordisk, Owkin, Pfizer, PhaseBio, PLx Pharma, Recardio,
research from Amgen, AstraZeneca, Bayer, Janssen, Merck, Novo Regeneron, Reid Hoffman Foundation, Roche, Sanofi, Stasys, Synaptic,
Nordisk, Pfizer, Sanofi, Wraser. M.D. reports personal fees for lectures The Medicines Company, 89Bio; Royalties: Elsevier (Editor,
and trial steering committee participation from AstraZeneca and Braunwald’s Heart Disease); Site Co-Investigator: Abbott, Biotronik,
5044 M.P. Bonaca et al.

Boston Scientific, CSI, St Jude Medical (now Abbott), Philips, Svelte; 5. Bhatt DL, Steg PG, Mehta SR, Leiter LA, Simon T, Fox K, et al. Ticagrelor in patients
Trustee: American College of Cardiology; Unfunded Research: with diabetes and stable coronary artery disease with a history of previous percutan­
eous coronary intervention (THEMIS-PCI): a phase 3, placebo-controlled, rando­
FlowCo, Takeda. P.G.S. reports research grants from Amarin, Bayer,
mised trial. Lancet 2019;394:1169–1180.
Merck, Sanofi, and Servier, speaking or consulting fees from Amarin, 6. Steg PG, Bhatt DL, Simon T, Fox K, Mehta SR, Harrington RA, et al. Ticagrelor in
Amgen, AstraZeneca, Bayer/Janssen, Boehringer Ingelheim, Bristol patients with stable coronary disease and diabetes. N Engl J Med 2019;381:
Myers Squibb, Idorsia, Lilly, Merck, Novartis, Novo Nordisk, Pfizer, 1309–1320.
Regeneron, Sanofi, and Servier. M.C. reports grants and personal fees 7. Mauri L, Kereiakes DJ, Yeh RW, Driscoll-Shempp P, Cutlip DE, Steg PG, et al. Twelve
from AstraZeneca, during the conduct of the study; personal fees or 30 months of dual antiplatelet therapy after drug-eluting stents. N Engl J Med
2014; 371:2155–2166.
from Merck, personal fees from Janssen, personal fees from Maquet, per­

Downloaded from https://academic.oup.com/eurheartj/article/43/48/5037/6823782 by European Society of Cardiology user on 28 May 2023


8. Magnani G, Ardissino D, Im K, Budaj A, Storey RF, Steg PG, et al. Predictors, type, and
sonal fees from malpractice attorneys, grants from Janssen, grants from impact of bleeding on the net clinical benefit of long-term ticagrelor in stable patients
Edwards, personal fees from Merck, personal fees from BMS/Pfizer, per­ with prior myocardial infarction. J Am Heart Assoc 2021;10:e017008.
sonal fees from Janssen, personal fees from BI, personal fees from Lilly, 9. Halvorsen S, Storey RF, Rocca B, Sibbing D, Ten Berg J, Grove EL, et al. Management
outside the submitted work. K.I. is the member of the TIMI Study of antithrombotic therapy after bleeding in patients with coronary artery disease
Group which has received institutional research grant support through and/or atrial fibrillation: expert consensus paper of the European Society of
Cardiology Working Group on Thrombosis. Eur Heart J 2017;38:1455–1462.
Brigham and Women’s Hospital from Abbott, Amgen, Aralez,
10. Steg PG, Huber K, Andreotti F, Arnesen H, Atar D, Badimon L, et al. Bleeding in acute
AstraZeneca, Bayer HealthCare Pharmaceuticals, Inc., BRAHMS, coronary syndromes and percutaneous coronary interventions: position paper by
Daiichi Sankyo, Eisai, GlaxoSmithKline, Intarcia, Janssen, MedImmune, the Working Group on Thrombosis of the European Society of Cardiology. Eur
Merck, Novartis, Pfizer, Poxel, Quark Pharmaceuticals, Roche, Takeda, Heart J 2011;32:1854–1864.
The Medicines Company, and Zora Biosciences. P.J. is an employee of 11. Costa F, van Klaveren D, James S, Heg D, Raber L, Feres F, et al. Derivation and val­
idation of the predicting bleeding complications in patients undergoing stent implant­
AstraZeneca. E.B. reports grant support through Brigham and
ation and subsequent dual antiplatelet therapy (PRECISE-DAPT) score: a pooled
Women’s Hospital from AstraZeneca, Daiichi Sankyo, Merck, and
analysis of individual-patient datasets from clinical trials. Lancet 2017;389:
Novartis, Consultancies with Amgen, Cardurion, MyoKardia, Novo 1025–1034.
Nordisk, and Verve. M.S.S. received research grant support through 12. Mehran R, Pocock SJ, Nikolsky E, Clayton T, Dangas GD, Kirtane AJ, et al. A risk
Brigham and Women’s Hospital from Abbott, Amgen, Anthos score to predict bleeding in patients with acute coronary syndromes. J Am Coll
Therapeutics, AstraZeneca, Bayer, Daiichi Sankyo, Eisai, Intarcia, Ionis, Cardiol 2010;55:2556–2566.
13. Yeh RW, Secemsky EA, Kereiakes DJ, Normand SL, Gershlick AH, Cohen DJ, et al.
Medicines Company, MedImmune, Merck, Novartis, Pfizer, and Quark
Development and validation of a prediction rule for benefit and harm of dual anti­
Pharmaceuticals. Consulting for Althera, Amgen, Anthos Therapeutics, platelet therapy beyond 1 year after percutaneous coronary intervention. JAMA
AstraZeneca, Beren Therapeutics, Bristol Myers Squibb, DalCor, Dr 2016;315:1735–1749.
Reddy’s Laboratories, Fibrogen, Intarcia, Merck, Moderna, Novo 14. Bhatt DL, Bonaca MP, Bansilal S, Angiolillo DJ, Cohen M, Storey RF, et al. Reduction
Nordisk, and Silence Therapeutics. Additionally, Dr Sabatine is a member in ischemic events with ticagrelor in diabetic patients with prior myocardial infarction
of the TIMI Study Group, which has also received institutional research in PEGASUS-TIMI 54. J Am Coll Cardiol 2016;67:2732–2740.
15. Bonaca MP, Bhatt DL, Steg PG, Storey RF, Cohen M, Im K, et al. Ischaemic risk and
grant support through Brigham and Women’s Hospital from: ARCA
efficacy of ticagrelor in relation to time from P2Y12 inhibitor withdrawal in patients
Biopharma, Inc., Janssen Research and Development, LLC, Siemens with prior myocardial infarction: insights from PEGASUS-TIMI 54. Eur Heart J 2016;
Healthcare Diagnostics, Inc., Softcell Medical Limited, Regeneron, 37:1133–1142.
Roche, and Zora Biosciences. 16. Bonaca MP, Bhatt DL, Storey RF, Steg PG, Cohen M, Kuder J, et al. Ticagrelor for
prevention of ischemic events after myocardial infarction in patients with peripheral
artery disease. J Am Coll Cardiol 2016;67:2719–2728.
Data availability 17. Magnani G, Storey RF, Steg G, Bhatt DL, Cohen M, Kuder J, et al. Efficacy and safety of
Data for the analyses cannot be shared by those interested in collabor­ ticagrelor for long-term secondary prevention of atherothrombotic events in rela­
ation are encouraged to contact the corresponding author. tion to renal function: insights from the PEGASUS-TIMI 54 trial. Eur Heart J 2016;
37:400–408.
18. Bansilal S, Bonaca MP, Cornel JH, Storey RF, Bhatt DL, Steg PG, et al. Ticagrelor for
References secondary prevention of atherothrombotic events in patients with multivessel cor­
1. Collet JP, Thiele H, Barbato E, Barthelemy O, Bauersachs J, Bhatt DL, et al. 2020 ESC
onary disease. J Am Coll Cardiol 2018;71:489–496.
guidelines for the management of acute coronary syndromes in patients presenting
19. Dellborg M, Bonaca MP, Storey RF, Steg PG, Im KA, Cohen M, et al. Efficacy and
without persistent ST-segment elevation. Eur Heart J 2021;42:1289–1367.
safety with ticagrelor in patients with prior myocardial infarction in the approved
2. Knuuti J, Wijns W, Saraste A, Capodanno D, Barbato E, Funck-Brentano C, et al.
2019 ESC guidelines for the diagnosis and management of chronic coronary syn­ European label: insights from PEGASUS-TIMI 54. Eur Heart J Cardiovasc
dromes. Eur Heart J 2020;41:407–477. Pharmacother 2019;5:200–206.
3. Levine GN, Bates ER, Bittl JA, Brindis RG, Fihn SD, Fleisher LA, et al. 2016 ACC/AHA 20. Magnuson EA, Li H, Wang K, Vilain K, Shafiq A, Bonaca MP, et al. Cost-Effectiveness
guideline focused update on duration of dual antiplatelet therapy in patients with of long-term ticagrelor in patients with prior myocardial infarction: results from the
coronary artery disease: a report of the American College of Cardiology/ PEGASUS-TIMI 54 trial. J Am Coll Cardiol 2017;70:527–538.
American Heart Association Task Force on Clinical Practice Guidelines. J Am Coll 21. Borenstein M, Hedges LV, Higgins JPT, Rothstein HR. Meta-Regression. Introduction
Cardiol 2016;68:1082–1115. to meta-analysis. Chichester: John Wiley and Sons; 2009. p187–204.
4. Bonaca MP, Bhatt DL, Cohen M, Steg PG, Storey RF, Jensen EC, et al. Long-term use 22. Steg PG, Lopez-Sendon J, Lopez de Sa E, Goodman SG, Gore JM, Anderson FA Jr,
of ticagrelor in patients with prior myocardial infarction. N Engl J Med 2015;372: et al. External validity of clinical trials in acute myocardial infarction. Arch Intern
1791–1800. Med 2007;167:68–73.

You might also like