+ All Categories
Home > Documents > Serial clopidogrel dose adjustment after platelet function testing improves outcome of acute...

Serial clopidogrel dose adjustment after platelet function testing improves outcome of acute...

Date post: 24-Jan-2017
Category:
Upload: davor
View: 218 times
Download: 2 times
Share this document with a friend
11
Serial clopidogrel dose adjustment after platelet function testing improves outcome of acute coronary syndrome patients undergoing percutaneous coronary intervention with high on-treatment platelet reactivity Jure Samardzic Miroslav Krpan Bosko Skoric Marijan Pasalic Mate Petricevic Davor Milicic Ó Springer Science+Business Media New York 2014 Abstract High on-treatment platelet reactivity (HTPR) on clopidogrel correlates with adverse outcomes in patients treated with percutaneous coronary intervention (PCI). Whether HTPR is a modifiable risk factor for future events is not clear. We evaluated the effect of serial clopidogrel dose adjustment based on platelet function testing (PFT) during 12 months of dual antiplatelet therapy (DAPT) using Multiplate Ò analyzer in patients with HTPR after PCI in acute coronary syndrome on clinical outcome. Eighty- seven patients were randomized to interventional (n = 43) and control group (n = 44). Blood samples for PFT were drawn at day 1, 2, 3, 7, 30 and at month 2, 3, 6, 9 and 12. Clopidogrel dose was modified at each point of PFT in the interventional group with patients taking up to two addi- tional 600 mg loading doses and a range of 75–300 mg maintenance dose to achieve and maintain optimal platelet reactivity (19–46 U). The incidence of the primary end- point (composite of cardiovascular death, non-fatal myo- cardial infarction, target vessel revascularization and ischemic stroke) was significantly higher in the control group (36.3 vs 16.2 %; p = 0.034). There were no differ- ences in total bleeding events (6.8 vs 4.6 %, p = ns). Patients in the interventional group maintained better P2Y 12 inhibition during follow-up. We hypothesize that targeting the therapeutic window of platelet reactivity continuously throughout DAPT by dose adjustment of P2Y 12 inhibitor may lead to better platelet reactivity con- trol, and thus reduce the rate of ischemic complications in this high risk group of patients. Keywords Clopidogrel Á Platelet reactivity Á Acute coronary syndrome Á Tailoring therapy Á Outcome Introduction Dual antiplatelet therapy (DAPT) with aspirin and P2Y 12 receptor antagonist during 12 months presents cornerstone treatment in acute coronary syndrome (ACS) patients undergoing percutaneous coronary intervention (PCI) [14]. Despite the use of DAPT 9–12 % of patients still develop new ischemic event after myocardial infarction (MI) [57]. Clopidogrel is the most widely used P2Y 12 inhibitor [8] despite it’s limitations that include highly variable P2Y 12 -receptor inhibition which causes wide interindividual platelet reactivity variations [9]. Since high on-treatment platelet reactivity (HTPR) on clopidogrel is strongly associated with adverse events [10], antiplatelet therapy tailoring has been vastly investigated to determine whether individualized approach could improve outcomes. New P2Y 12 inhibitors like prasugrel and ticagrelor produce more potent and more uniform antiplatelet effect and thus reduce the risk of adverse ischemic events in ACS patients undergoing stent implantation better than clopidogrel but at the cost of increased risk of major, non CABG related bleeding [6, 7]. In the time of progressive personalized approach to therapy, effective strategies are needed to minimize the risk of ischemic adverse events without increasing the risk for bleeding. We conducted a J. Samardzic (&) Á M. Krpan Á B. Skoric Á M. Pasalic Á D. Milicic Department of Cardiovascular Diseases, University Hospital Center Zagreb, University of Zagreb School of Medicine, Kispaticeva 12, 10000 Zagreb, Croatia e-mail: [email protected] M. Petricevic Department of Cardiac Surgery, University Hospital Center Zagreb, University of Zagreb School of Medicine, Kispaticeva 12, 10000 Zagreb, Croatia 123 J Thromb Thrombolysis DOI 10.1007/s11239-014-1087-0
Transcript

Serial clopidogrel dose adjustment after platelet function testingimproves outcome of acute coronary syndrome patientsundergoing percutaneous coronary intervention with highon-treatment platelet reactivity

Jure Samardzic • Miroslav Krpan • Bosko Skoric •

Marijan Pasalic • Mate Petricevic • Davor Milicic

� Springer Science+Business Media New York 2014

Abstract High on-treatment platelet reactivity (HTPR)

on clopidogrel correlates with adverse outcomes in patients

treated with percutaneous coronary intervention (PCI).

Whether HTPR is a modifiable risk factor for future events

is not clear. We evaluated the effect of serial clopidogrel

dose adjustment based on platelet function testing (PFT)

during 12 months of dual antiplatelet therapy (DAPT)

using Multiplate� analyzer in patients with HTPR after PCI

in acute coronary syndrome on clinical outcome. Eighty-

seven patients were randomized to interventional (n = 43)

and control group (n = 44). Blood samples for PFT were

drawn at day 1, 2, 3, 7, 30 and at month 2, 3, 6, 9 and 12.

Clopidogrel dose was modified at each point of PFT in the

interventional group with patients taking up to two addi-

tional 600 mg loading doses and a range of 75–300 mg

maintenance dose to achieve and maintain optimal platelet

reactivity (19–46 U). The incidence of the primary end-

point (composite of cardiovascular death, non-fatal myo-

cardial infarction, target vessel revascularization and

ischemic stroke) was significantly higher in the control

group (36.3 vs 16.2 %; p = 0.034). There were no differ-

ences in total bleeding events (6.8 vs 4.6 %, p = ns).

Patients in the interventional group maintained better

P2Y12 inhibition during follow-up. We hypothesize that

targeting the therapeutic window of platelet reactivity

continuously throughout DAPT by dose adjustment of

P2Y12 inhibitor may lead to better platelet reactivity con-

trol, and thus reduce the rate of ischemic complications in

this high risk group of patients.

Keywords Clopidogrel � Platelet reactivity � Acute

coronary syndrome � Tailoring therapy � Outcome

Introduction

Dual antiplatelet therapy (DAPT) with aspirin and P2Y12

receptor antagonist during 12 months presents cornerstone

treatment in acute coronary syndrome (ACS) patients

undergoing percutaneous coronary intervention (PCI) [1–

4]. Despite the use of DAPT 9–12 % of patients still

develop new ischemic event after myocardial infarction

(MI) [5–7]. Clopidogrel is the most widely used P2Y12

inhibitor [8] despite it’s limitations that include highly

variable P2Y12-receptor inhibition which causes wide

interindividual platelet reactivity variations [9]. Since high

on-treatment platelet reactivity (HTPR) on clopidogrel is

strongly associated with adverse events [10], antiplatelet

therapy tailoring has been vastly investigated to determine

whether individualized approach could improve outcomes.

New P2Y12 inhibitors like prasugrel and ticagrelor produce

more potent and more uniform antiplatelet effect and thus

reduce the risk of adverse ischemic events in ACS patients

undergoing stent implantation better than clopidogrel but at

the cost of increased risk of major, non CABG related

bleeding [6, 7]. In the time of progressive personalized

approach to therapy, effective strategies are needed to

minimize the risk of ischemic adverse events without

increasing the risk for bleeding. We conducted a

J. Samardzic (&) � M. Krpan � B. Skoric � M. Pasalic �D. Milicic

Department of Cardiovascular Diseases, University Hospital

Center Zagreb, University of Zagreb School of Medicine,

Kispaticeva 12, 10000 Zagreb, Croatia

e-mail: [email protected]

M. Petricevic

Department of Cardiac Surgery, University Hospital Center

Zagreb, University of Zagreb School of Medicine,

Kispaticeva 12, 10000 Zagreb, Croatia

123

J Thromb Thrombolysis

DOI 10.1007/s11239-014-1087-0

prospective, randomized study with the aim to investigate

whether serial clopidogrel dose adjustment according to

platelet reactivity measurements using Multiplate� ana-

lyzer (Roche Diagnostics, Mannheim, Germany) could

decrease the rate of adverse events in ACS patients treated

with PCI and with determined HTPR.

Methods

This study was approved by Ethics Committees of Uni-

versity Hospital Centre Zagreb and University of Zagreb

School of Medicine. All patients gave informed consent

before enrollment. Inclusion criteria were predefined as

follows: ACS patients aged 18–80, treated with aspirin,

clopidogrel and successful coronary stenting and with

HTPR phenotype determined by multiple electrode aggre-

gometry (MEA) 12–24 h after PCI. ACS was diagnosed

based on clinical presentation, ECG changes and troponin

T values. Exclusion criteria were continuous postinter-

ventional glycoprotein (GP) IIbIIIa receptor inhibitor per-

fusion, thrombocytopenia (\150 9 109/L), significant

renal insufficiency (creatinine [ 200 lmol/L), anemia

(Htc \ 30 %), hemorrhagic diathesis, history of intracra-

nial bleeding or ischemic cerebrovacular insult 6 months

before, major operation 6 weeks before, concomitant

chronic anticoagulation therapy and advanced age

([80 years). All patients were treated with aspirin and

clopidogrel as newer P2Y12 inhibitors were unavailable.

Patients presenting with STEMI (60.9 %) were subjected

to primary PCI and received 600 mg loading dose (LD) of

clopidogrel prior to the procedure. Patients presenting with

NSTEMI (22.9 %) were predominantly treated with

300 mg LD of clopidogrel and early invasive strategy

(\24 h). Patients presenting wih unstable angina—UA

(16.1 %) were predominantly treated with DAPT for a

couple of days before PCI. Patients with STEMI and

NSTEMI received loading dose of aspirin—300 mg prior

to PCI. Patients with UA received aspirin 100 mg/day

several days prior to PCI. All patients received 100 mg/day

maintenance dose of aspirin.

Platelet activity was measured 12–24 h following suc-

cessful PCI using MEA—one of several recommended

platelet function tests (PFT) [11]. Multiplate� analyzer is a

point of care device that uses whole blood sample for PFT.

Depending on the test that is used it can measure platelet

reactivity through different signaling pathways and thus,

measure the effect of different types of antiplatelet drugs.

Measuring platelet response to antiplatelet therapy using

MEA is based on the impedance, where a change of

resistance between two electrodes immersed into the test-

ing blood is determined after the addition of saline and a

specific aggregation agonist—in this case adenosine

diphosphate (ADP) with a final concentration of 6.4 lM.

The blood samples were taken for analysis in most cases

from the antecubital vein in the official MEA tubes which

are coated with hirudin, a direct thrombin inhibitor, which

does not interfere with platelet activation. After blood

collection, the tubes were immediately gently inverted

several times and tested after at least 30 min. According to

the manufacturer’s recommendations, all samples were

tested between 30 min and three hours after blood collec-

tion. The volume of blood required for performing one test

is 300 lL and the result is available in approximately

10 min (3 min for incubation and 6 min for the post-

stimulation measurement).

We screened 461 consecutive ACS patients. All patients

without exclusion criteria (341) underwent PFT. We

identified and enrolled 87 patients with HTPR in the study

(Fig. 1). Assignment to study groups was performed by

random allocation using randomization software (Research

Randomizer). All patients enrolled in the study were

hemodynamically stable at randomization. None of the

patients enrolled in the study had cardiogenic shock nor

IABP implanted at randomization. This is because patients

who presented on arrival with cardiogenic shock either

died before enrollment, had unsuccessful PCI or had

developed exclusion criteria on screening (i.e. renal

insufficiency). Furthermore, some patients with cardio-

genic shock who were eligible for PFT 12–24 h after

successful PCI were either in no condition to give inform

ACS patients treated with PCIn = 461

Patients with exclusion criteria n = 120

MEAtestingn = 341

MEA platelet reactivity≤ 46 U

n = 254 (74.5%)

HTPRn = 87 (25.5%)

Interventional groupn = 43

Control groupn = 44

Fig. 1 Flowchart of study design and patient selection (ACS acute

coronary syndrome, HTPR high on-treatment platelet reactivity, MEA

multiple electrode aggregometry, PCI percutaneous coronary

intervention)

S. Jure et al.

123

consent for PFT (i.e. patients with out of hospital cardiac

arrest undergoing therapeutic hypothermia) or decided not

to participate in the study.

The enrolled patients were randomily assigned either to

clopidogrel tailoring group (interventinal group) or stan-

dard clopidogrel dose group (control group). Clopidogrel

dose was adjusted just as PFT results were obtained.

Control PFT and tailoring regimen in the first seven days

were performed in the morning before taking the therapy

according to the protocol described in Fig. 2. Cut off

values for HTPR and enchanced platelet response

were set according to the consensus statement at [ 46 U

and \ 19 U, respectively [12]. The patients in the inter-

ventional group recieved up to two additional loading

doses of 600 mg clopidogrel and up to 300 mg mainte-

nance dose of clopidogrel (150 mg maximum for patients

older than 70). Clopidogrel dose adjustment at 30 days and

on following PFT controls were performed in the same

manner as on day 7. The control group underwent PFT in

the same intervals but without clopidogrel dose adjustment.

Compliance to treatment and major adverse cardiac and

cerebrovascular events (MACCE) occurence were assesed

on control visits by interview and by counting the returned

clopidogrel tablets given to the patient at the last visit.

Primary endpoint was the composite of death, non-fatal

MI, target vessel revascularization and ischemic stroke.

Secondary endpoints were bleeding events according to

Bleeding Academic Research Consortium (BARC) defini-

tion [13].

Statistical methodology

All collected data were analyzed by descriptive statistics.

Categorical variables are shown as frequencies and prev-

alence. Metric variables are shown as arithmetic means

with standard deviations. Data are supplemented by tables

and figures. Kolmogorov–Smirnov test was used to test for

normal distribution of continuous data. Appropriate para-

metric and non-parametric tests were used according to the

results. Differences in individual characteristics between

the study and control groups were tested by Student’s t test

for independent samples metric variables and v2 test for

categorical variables. The difference in clinical outcome

MD decreaseif 300 mg decrease to 150 mgif 150 mg decrease to 75 mg

if 75 mg continue 75 mg

No changes in the MDMD increaseif 75 mg raise to 150 mgif 150 mg raise to 300 mgif 300 mg continue 300mg

(for patients > 70 years MD 150 mg max)

Day 7 - MEA testing

clopidogrel75 mg MD

clopidogrel150 mg MD

clopidogrel300 mg MD

clopidogrel 75 mg MDclopidogrel 150 mg MD< 19 U

19-46 U

19-46 U> 46 U

< 19 U

< 19 U≥ 19 U

≤ 46 U

> 46 U

> 46 U

Interventional group

Day 1 - clopidogrel 600 mg LD

clopidogrel 150 mg MDDay 2 - MEA testingclopidogrel 600 mg LD

Day 3 - MEA testing

Fig. 2 Flow chart of the personalized treatment with clopidogrel in the interventional group during the first week after coronary

revascularization (LD loading dose, MEA multiple electrode aggregometry, MD maintenance dose)

Serial clopidogrel dose adjustment

123

between the two groups was tested by v2 test. The differ-

ences in the data between study groups were tested by

Student’s t test of independent samples or ANOVA for

multiple independent samples. Survival to MACCE and the

rate of ischemic events are shown by Kaplan–Meier curve.

P values \ 0.05 were regarded statistically significant.

Statistical analysis was performed using SPSS software

version 21 (IBM Corporation, USA).

Results

Eighty-seven patients were enrolled in the study: 43 in the

interventional group and 44 in the control group. Most

patients presented with STEMI (60.9 %), while NSTEMI

and UA were present in 22.9 % and 16.1 % of patients,

respectively. Most of the patients were clopidogrel naive at

admission (95.4 %). Aside from the extension of coronary

artery disease the groups did not differ significantly in

concomitant medical therapy, CYP2C19 genotype, demo-

graphic and procedural characteristics (Tables 1, 2). Pro-

portion of patients treated with lipophilic statin who

presented with STEMI, NSTEMI and UA and were

receiving initially intensified statin dose was 76.3, 66.6 and

14.3 %, respectively. During the follow-up 7 and 16

patients reached the primary outcome in the interventional

and control group, respectively (16.2 vs 36.3 %;

p = 0.034). Two patients were lost to follow-up in both

groups. Three and four patients died in the control and

interventional group, respectively. Three stent thromboses

(ST) occurred in the control group, while there was only

one ST in the interventional group (p = ns). Two patients

in the interventional and one in the control group experi-

enced ischemic stroke. There were no differences in total

bleeding outcomes; 6.8 and 4.6 % in the control and

interventional group, respectively (p = ns). One BARC

type 2 and type 5 bleeding event occurred in the inter-

ventional group, while there were two type 2 and one type

3 event in the control group. Patients in the interventional

group had a significantly better outcome and survival to an

adverse event (ischemic or bleeding) as shown in Fig. 3.

The proportion of patients with HTPR decreased in both

groups after randomization, but significantly more in the

interventional group (Fig. 4). Mean platelet reactivity of

patients in both groups is shown in Fig. 5. Intraindividual

platelet reactivity was noted in both study groups. Mean

difference between minimal and maximal platelet reactiv-

ity in each patient was 50 U (SD ± 17.47) and 53.46 U

(SD ± 16.71) in the control and interventional group,

respectively (Fig. 6). Platelet inhibition was mostly not

consistent in relation to the cut off point for HTPR as

52.3 % of patients crossed from a responder to a non-

responder at least three times (Tables 3, 4).

Discussion

To the best of our knowledge this is the first randomized

study that performed serial clopidogrel dose adjustment

during 12 months of DAPT to maintain the proposed range

of ideal platelet inhibition recommended for MEA [12]

based on receiver-operator characteristic (ROC) curve

analysis performed by Sibbing et al. that determined

Table 1 Baseline demographic characteristics of study patients

Patients’ characteristics Interventional

group

(n = 43)

Control

group

(n = 44)

p

Age, mean (SD) 63.37 (12.56) 63.48 (12.15) 0.968

Men, n (%) 22 (51.1) 27 (61.4) 0.338

Unstable angina, n (%) 8 (18.6) 6 (13.7)

0.801NSTEMI, n (%) 10 (23.3) 10 (22.7)

STEMI, n (%) 25 (58.1) 28 (63.6)

Arterial hypertension, n (%) 28 (65.1) 27 (61.4) 0.717

Hyperlipidemia, n (%) 25 (58.1) 20 (45.5) 0.236

Diabetes mellitus, n (%) 14 (32.6) 12 (27.3) 0.590

Smokers, n (%) 9 (20.9) 12 (27.3) 0.489

Family history for

CAD, n (%)

7 (16.3) 6 (13.6) 0.730

Previous MI, n (%) 6 (13.9) 3 (6.8) 0.314

Previous PCI, n (%) 6 (13.9) 2 (4.5) 0.157

Previous CABG, n (%) 0 (0) 0 (0) 1.000

Previous stroke, n (%) 1 (2.3) 4 (9.1) 0.360

PAD, n (%) 0 (0) 1 (2.3) 1.000

BMI, kg/m2, mean (SD) 28.93 (4.45) 27.90 (4.98) 0.313

Medical therapy

Clopidogrel naıve patients

prior to randomization,

n (%)

40 (93.1) 43 (97.7) 0.360

Statins, n (%) 40 (93.0) 31 (93.2) 0.689

Lipophilic statin, n (%) 21 (48.8) 30 (68.2) 0.150

PPIs, n (%) 14 (32.5) 22 (50.0) 0.151

Calcium channel

blocker, n (%)

6 (13.9) 7 (15.9) 1.000

CYP2C19*2, n (%)

Carrier homozygote 0 (0) 2 (4.8)

0.317Carrier heterozygote 16 (37.2) 13 (30.9)

Others 27 (62.8) 27 (64.3)

CYP2C19*17, n (%)

Carrier homozygote 4 (9.3) 4 (9.5)

0.996Carrier heterozygote 14 (32.5) 14 (33.3)

Others 25 (58.1) 24 (57.1)

BMI body mass index, CABG coronary artery bypass graft, CAD

coronary artery disease, MI myocardial infarction, NSTEMI non ST-

elevation myocardial infarction, PAD peripheral artery disease, PCI

percutaneous coronary intervention, PPI proton pump inhibitor, SD

standard deviation, STEMI ST-elevation myocardial infarction

S. Jure et al.

123

optimal range of platelet reactivity on clopidogrel for MEA

[14, 15].

As the benefit of DAPT is consistent during 12 months

post PCI [16], we assumed that the serial PFT and dose

adjustment to prevail HTPR during 12 months would lead

to a better outcome. Our results suggest that serial clopi-

dogrel dose adjustment according to PFT throughout

12 months of DAPT leads to better platelet inhibition in

patients with initial HTPR after coronary stenting in ACS

as we observed that the control group had a significantly

larger rate of HTPR phenotype during 12 months of fol-

low-up (p \ 0.01). As the patients in the interventional

group had a better outcome, this might imply that PFT

might be useful in guiding antiplatelet therapy in patients

Table 2 Procedural characteristics of study patients

Angiographic and

PCI characteristics

Interventional

group

Control

group

p Angiographic and

PCI characteristics

Interventional

group

Control

group

p

Mean procedure

duration, min (SD)

36.05 (17.98) 38.23 (18.16) 0.575 Lesion type

De novo, n (%) 37 (86.0) 42(95.4)

0.226Bifurcation, n (%) 4 (9.3) 2 (4.5)

ISR/ST, n (%) 2 (4.6) 0 (0)

Treated arteries Stent type

One, n (%) 41 (95.3) 42(95.5)

0.981

BMS, n (%) 37 (86.0) 42 (95.4)0.157

Two, n (%) 2 (4.7) 2 (4.5) DES, n (%) 6 (13.9) 2 (4.5)

Treated lesions Thrombectomy, n (%) 7 (16.3) 8 (18.2) 1.000

One, n (%) 34 (79.1) 36 (81.8)0.748

Two, n (%) 9 (20.9) 8 (18.2)

CAD Mean referent artery

diameter, cm (SD)

3,05 (0.05) 3.18 (0.07) 0.153

Single vessel, n (%) 24 (55.8) 11 (25.0)

0.001Two vessel, n (%) 13 (30.2) 14 (31.8)

Three vessel, n (%) 6 (14.0) 19 (43.2)

Culprit lesion Mean lesion length,

mm (SD)

19,51 (9.86) 19.89 (7.84) 0.845

LAD, n (%) 16 (37.2) 16 (36.5) 0.935

LCx, n (%) 10 (23.2) 8 (18.2) 0.559

RCA, n (%) 19 (44.2) 22 (50.0) 0.587

Tortuosities, n (%) 2 (4.6) 5 (11.4) 0.434 Stenosis, % (SD) 95,26 (8.82) 94.80 (10.04) 0.821

Dominance TIMI flow before wire

Right, n (%) 34 (79.1) 30 (68.2)

0.488

0 17 (39.5) 21 (47.7)

0.791Left, n (%) 2 (4.7) 4 (9.1) 1 0 (0) 1 (2.3)

Equilibrated, n (%) 7 (16.2) 10 (22.7) 2 8 (18.6) 2 (4.5)

3 18 (41.8) 20 (45.4)

Stents implanted TIMI flow after wire

One, n 31 28

0.470

0 5 (11.6) 10 (22.7)

0.699Two, n 8 12 1 4 (9.3) 3 (6.8)

Three, n 2 3 2 10 (23.2) 6 (13.6)

Four, n 2 1 3 24 (55.8) 25 (56.8)

Mean width of the

narrowest

stent, mm (SD)

2.99 (0.064) 3.11 (0.082) 0.232 TIMI flow, final

0 0 (0) 0 (0)

0.6281 0 (0) 0 (0)

2 3 (6.9) 2 (4.5)

3 40 (93.0) 42 (95.4)

Mean total stent length,

mm (SD)

26.30 (14.39) 28.73 (15.86) 0.457

BMS bare metal stent, CAD coronary artery disease, DES drug eluting stent, ISR in-stent restenosis, LAD left anterior descending, LCx left

circumflex, PCI percutaneous coronary intervention, RCA right coronary artery, SD standard deviation, ST stent thrombosis, TIMI thrombolysis

in myocardial infarction

Serial clopidogrel dose adjustment

123

presenting with ACS. The routine use of PFT is currenlty

not recommended for any PCI patients. It might be con-

sidered only in guiding antiplatelet therapy for patients

undergoing high-risk PCI or those with a known history of

stent thrombosis (ST) [11, 12].

Hazarbasanov et al. showed using MEA that additional

600 mg LD of clopidogrel and 150 mg clopidogrel main-

tenance dose (MD) during the first month after PCI

improved platelet inhibition in HTPR patients and their

outcome after six months. Their research group, however,

enrolled both ACS (57 %) and stable angina patients [17].

ACS patients’ platelet response change differs from stable

coronary artery disease (CAD) patients as ACS is a highly

hypercoagulabile state [18–20]. The degree of platelet

response to clopidogrel is influenced by more factors

beside ACS such as underdosing, compliance, comorbidi-

ties, concomitant therapy or CYP2C19 genotype [21–28].

Large, randomized studies addressing the intriguing issue

0102030405060708090

100

1 21 41 61 81 101

121

141

161

181

201

221

241

261

281

301

321

341

361

Control group

Interventional group

p=0.044

0

2

4

6

8

10

12

14

16

181 20 39 58 77 96 115

134

153

172

191

210

229

248

267

286

305

324

343

362

p=0.034

ba

Time (days)Time (days)

Pat

ient

s (n

o.)

Pat

ient

s (%

)

Control group

Interventional group

Fig. 3 a Kaplan–Meier analysis of cumulative ischemic events in both groups. b Kaplan–Meier survival analysis to an ischemic or a bleeding

event in both groupsP

atie

nts

with

HT

PR

(%

)

Interventional vs control group p<0.01

Interventional group

Control group

Day 1 Day 2 Day 3 Day 7 Day 30 Mo. 2 Mo. 3 Mo. 6 Mo. 9 Mo. 12

90

70

50

30

10

0

20

40

60

80

100Fig. 4 Rates of high on-

treatment platelet reactivity in

both groups of patients during

follow-up. HTPR high on-

treatment platelet reactivity

The

rape

utic

win

dow

AD

P -

U (

Uni

ts)

Day 1 Day 2 Day 3 Day 7 Day 30 Mo. 2 Mo. 3 Mo. 6 Mo. 9 Mo. 12

Interventional group

Control group

0

10

20

30

40

50

60

70

80Fig. 5 Mean platelet reactivity

in both groups of patients during

follow-up

S. Jure et al.

123

of PFT in giuding antiplatelet therapy using VerifyNow�

(Accumetrics Inc., San Diego, California, USA) brought

negative results and setbacks in PFT [29, 30]. As those

trials recruited mostly stable coronary artery disease

patients, testing usefulness of PFT in tailoring therapy is

being switched to ACS patients—a population that really

might benefit with this strategy as they are at a higher risk

for adverse events. Currently, there are no completed large

randomized studies that have addressed this issue in ACS

patients. Previous studies have shown that platelet reac-

tivity decreases with time and that certain proportion of

patients still exhibit HTPR phenotype [29, 31]. In

GRAVITAS study, 62 % of patients with initial HTPR in

the standard-dose clopidogrel group remained low

responders to clopidogrel after 30 days [29]. Around 50 %

of patients in our control group kept HTPR phenotype

during the late post PCI period, as well. We also observed

significant intraindividual temporal variations in platelet

reactivity in both study groups. This implies that HTPR and

a personalized approach in antiplatelet treatment should

continue to be further extensively investigated. Even though

HTPR was shown to be an independent predictor of ische-

mic adverse events [32, 33] there is still no answer to the

question whether it is a modifiable factor. Our results imply

that maintaining proposed optimal platelet inhibition

throughout early and late period after coronary stenting in

ACS leads to a better clinical outcome. In the RECLOSE

2-ACS study, Parodi et al. showed using light transmission

aggregometry (LTA) that HTPR phenotype in ACS patients

after PCI was associated with increased risk of adverse

ischemic events at both short- and long-term follow-up

despite increased clopidogrel maintenance dose to

150–300 mg/day or switching to ticlopidine

(500–1,000 mg/day) [32]. Aradi et al. used MEA to tailor

antiplatelet therapy in ACS patients treated with PCI.

Patients with HTPR were assigned to either prasugrel or

high-dose clopidogrel. After one year, patients on prasugrel

had similar outcome as those without initial HTPR, while

patients on high-dose clopidogrel experienced more ische-

mic and bleeding events [34]. Design of our investigation

was based on the assumption that determining platelet

reactivity at more occasions would lead to a better insight of

platelet response during DAPT, better patient control, better

compliance, optimal treatment and better outcome. We

believe this approach to be reasonable as it is known that

platelet reactivity is a dynamic variable with inter- and in-

traindividial variations [35]. It is also known that HTPR is

associated with clopidogrel dose [36] and that the exact

timing to perform PFT has not been established [11, 12].

This could explain why the patients on high-dose clopido-

grel in study by Aradi et al. [34] experienced more ischemic

and bleeding events as some of those patients might have

been over or undertreated with clopidogrel. Analysis of the

ISAR-HPR registry performed by Mayer et al. with almost

one thousand patients treated with PCI and with determined

HTPR phenotype using MEA showed that patients who

recieved P2Y12 inhibitor dose adjustment based on MEA

had significantly lower incidence of ST and composite of

death without statistically significant increase in major

bleeding events in comparison to control cohort group after

30 days [37]. Siller-Matula et al. conducted a non-ran-

domized prospective study evaluating antiplatelet therapy

adjustment based on PFT using MEA. The authors enrolled

798 patients presenting mostly with stable CAD (63 %).

Patients in the guided group recieved up to four 600 mg

clopidogrel loading doses and prasugrel 60 mg loading dose

to overcome HTPR. After 30 days a significantly lower rate

of ST and MI was observed in the guided group without

significant differences in hemorrhagic events between study

groups [38]. The longest PFT study conducted was TRIL-

OGY-ACS Platelet Function Substudy which found no as-

socciation between PFT and clinical outcome throughout

30 months in patients with ACS initially treated without

revascularization [39]. This, however, can not yet be applied

to ACS patients treated with PCI as HTPR was shown to be

an independent risk factor for early and late ST [10].

0

20

40

60

80

100

120

140

160

0

20

40

60

80

100

120

140

160

Pla

tele

t rea

ctiv

ity -

AD

P (

U)

Pla

tele

t rea

ctiv

ity -

AD

P (

U)

PatientsPatients

a b

Fig. 6 Intra-individual platelet reactivity range during follow up. a Control group. b Interventional group

Serial clopidogrel dose adjustment

123

Table 3 Intra-individual responder/non responder crossovers in the interventional group

Patient Day 1 Day 2 Day 3 Day 7 Mo. 1 Mo. 2 Mo. 3 Mo. 6 Mo. 9 Mo. 12 Responder/

nonresponder

crossover (n)

1 48 U B46 U HTPR HTPR B46 U B46 U B46 U B46 U B46 U B46 U 3

2 48 U B46 U B46 U B46 U HTPR B46 U B46 U B46 U B46 U HTPR 4

3 93 U HTPR HTPR HTPR B46 U B46 U HTPR HTPR B46 U B46 U 3

4 53 U B46 U B46 U B46 U B46 U B46 U B46 U B46 U B46 U B46 U 1

5 51 U B46 U B46 U HTPR B46 U HTPR HTPR B46 U HTPR HTPR 6

6 53 U B46 U B46 U B46 U B46 U B46 U B46 U B46 U B46 U HTPR 2

7 57 U B46 U B46 U B46 U ND ND ND ND ND ND 1

8 76 U B46 U B46 U HTPR B46 U B46 U B46 U B46 U B46 U B46 U 3

9 53 U B46 U B46 U B46 U B46 U HTPR B46 U HTPR B46 U HTPR 6

10 94 U B46 U B46 U HTPR B46 U B46 U B46 U B46 U B46 U B46 U 3

11 47 U B46 U B46 U HTPR B46 U B46 U B46 U B46 U B46 U B46 U 3

12 55 U B46 U B46 U B46 U B46 U HTPR B46 U B46 U B46 U B46 U 3

13 89 U HTPR HTPR HTPR ND ND ND ND ND ND 0

14 47 U HTPR B46 U B46 U B46 U B46 U B46 U B46 U B46 U B46 U 1

15 50 U B46 U B46 U B46 U HTPR HTPR ND ND ND ND 2

16 74 U HTPR HTPR HTPR HTPR B46 U B46 U HTPR B46 U B46 U 3

17 76 U B46 U B46 U HTPR B46 U HTPR B46 U B46 U B46 U B46 U 5

18 66 U B46 U B46 U B46 U B46 U B46 U ND ND ND ND 1

19 96 U HTPR HTPR HTPR HTPR HTPR HTPR HTPR HTPR HTPR 0

20 54 U B46 U B46 U B46 U B46 U B46 U B46 U ND ND ND 1

21 70 U B46 U B46 U B46 U B46 U B46 U B46 U B46 U B46 U B46 U 1

22 60 U HTPR HTPR HTPR HTPR HTPR B46 U B46 U B46 U B46 U 1

23 52 U B46 U HTPR B46 U HTPR HTPR B46 U HTPR HTPR HTPR 6

24 51 U B46 U HTPR HTPR B46 U ND ND ND ND ND 3

25 57 U HTPR HTPR HTPR B46 U B46 U B46 U HTPR HTPR HTPR 2

26 62 U B46 U B46 U HTPR B46 U B46 U B46 U B46 U B46 U B46 U 3

27 77 U HTPR HTPR HTPR HTPR HTPR B46 U B46 U HTPR HTPR 2

28 59 U B46 U B46 U HTPR HTPR B46 U B46 U B46 U HTPR B46 U 4

29 49 U B46 U B46 U HTPR HTPR HTPR HTPR B46 U B46 U HTPR 4

30 75 U B46 U B46 U HTPR B46 U HTPR HTPR HTPR HTPR HTPR 4

31 68 U HTPR B46 U HTPR B46 U B46 U B46 U HTPR B46 U B46 U 5

32 63 U HTPR HTPR HTPR HTPR B46 U B46 U B46 U B46 U B46 U 1

33 49 U B46 U B46 U B46 U B46 U B46 U B46 U B46 U B46 U B46 U 1

34 79 U HTPR B46 U B46 U HTPR HTPR HTPR HTPR HTPR HTPR 2

35 48 U B46 U HTPR HTPR B46 U B46 U B46 U B46 U HTPR B46 U 5

36 48 U B46 U B46 U B46 U B46 U B46 U B46 U B46 U HTPR B46 U 3

37 49 U B46 U HTPR B46 U B46 U B46 U B46 U HTPR B46 U B46 U 5

38 48 U B46 U B46 U B46 U HTPR B46 U B46 U HTPR B46 U HTPR 6

39 48 U HTPR B46 U B46 U HTPR B46 U B46 U HTPR B46 U HTPR 6

40 48 U B46 U B46 U B46 U B46 U B46 U B46 U B46 U B46 U B46 U 1

41 63 U B46 U B46 U B46 U B46 U HTPR HTPR HTPR HTPR HTPR 2

42 64 U B46 U B46 U B46 U B46 U B46 U HTPR B46 U B46 U B46 U 3

43 71 U B46 U B46 U B46 U HTPR HTPR HTPR HTPR HTPR HTPR 2

HTPR high on-treatment platelet reactivity, ND not done

S. Jure et al.

123

Table 4 Intra-individual responder/non responder crossovers in the control group

Patient Day 1 Day 2 Day 3 Day 7 Mo. 1 Mo. 2 Mo. 3 Mo. 6 Mo. 9 Mo. 12 Responder/

nonresponder

crossover (n)

1 98 U HTPR B46 U HTPR B46 U B46 U B46 U B46 U B46 U B46 U 3

2 66 U HTPR HTPR HTPR HTPR HTPR HTPR HTPR HTPR HTPR 0

3 60 U B46 U B46 U HTPR B46 U B46 U B46 U B46 U B46 U B46 U 3

4 62 U HTPR B46 U HTPR B46 U HTPR HTPR B46 U HTPR HTPR 6

5 49 U B46 U B46 U B46 U B46 U B46 U HTPR HTPR HTPR HTPR 2

6 48 U HTPR HTPR B46 U B46 U B46 U B46 U B46 U B46 U B46 U 1

7 56 U B46 U B46 U HTPR B46 U HTPR ND ND ND ND 4

8 48 U B46 U B46 U B46 U B46 U B46 U HTPR ND ND ND 2

9 70 U HTPR B46 U HTPR HTPR HTPR HTPR HTPR HTPR HTPR 2

10 49 U B46 U B46 U B46 U B46 U HTPR B46 U B46 U B46 U B46 U 3

11 58 U B46 U B46 U B46 U B46 U B46 U HTPR B46 U HTPR HTPR 4

12 61 U HTPR B46 U HTPR HTPR HTPR HTPR HTPR HTPR HTPR 2

13 51 U B46 U HTPR B46 U B46 U B46 U B46 U B46 U B46 U B46 U 3

14 80 U B46 U B46 U HTPR B46 U B46 U HTPR HTPR HTPR HTPR 4

15 58 U HTPR HTPR HTPR HTPR HTPR HTPR HTPR HTPR HTPR 0

16 54 U B46 U HTPR B46 U HTPR ND ND ND ND ND 4

17 77 U HTPR HTPR HTPR B46 U B46 U HTPR B46 U HTPR B46 U 5

18 49 U HTPR HTPR HTPR HTPR HTPR HTPR HTPR HTPR HTPR 0

19 47 U HTPR HTPR HTPR HTPR HTPR B46 U B46 U HTPR B46 U 3

20 47 U HTPR B46 U HTPR B46 U B46 U HTPR HTPR HTPR HTPR 4

21 48 U B46 U HTPR HTPR B46 U HTPR HTPR B46 U B46 U HTPR 6

22 65 U HTPR B46 U B46 U B46 U B46 U B46 U B46 U B46 U HTPR 2

23 68 U HTPR HTPR HTPR B46 U B46 U B46 U B46 U B46 U B46 U 1

24 56 U B46 U B46 U HTPR HTPR B46 U B46 U B46 U B46 U B46 U 3

25 53 U HTPR HTPR HTPR HTPR HTPR HTPR HTPR HTPR HTPR 0

26 48 U HTPR HTPR HTPR HTPR HTPR HTPR HTPR HTPR HTPR 0

27 50 U B46 U B46 U HTPR B46 U B46 U B46 U B46 U HTPR HTPR 4

28 64 U HTPR HTPR HTPR B46 U HTPR HTPR HTPR HTPR HTPR 2

29 70 U HTPR HTPR B46 U B46 U B46 U B46 U B46 U B46 U B46 U 1

30 47 U HTPR HTPR B46 U HTPR HTPR HTPR B46 U HTPR HTPR 4

31 92 U HTPR HTPR HTPR HTPR B46 U HTPR B46 U B46 U HTPR 4

32 52 U B46 U B46 U B46 U B46 U B46 U B46 U B46 U B46 U B46 U 1

33 74 U HTPR HTPR HTPR HTPR HTPR HTPR HTPR HTPR HTPR 0

34 53 U HTPR HTPR HTPR B46 U HTPR HTPR HTPR HTPR HTPR 2

35 72 U HTPR HTPR HTPR HTPR HTPR HTPR HTPR HTPR HTPR 0

36 61 U HTPR B46 U B46 U B46 U B46 U B46 U B46 U B46 U B46 U 1

37 82 U HTPR HTPR HTPR HTPR HTPR HTPR HTPR HTPR HTPR 0

38 49 U B46 U HTPR HTPR HTPR HTPR HTPR HTPR B46 U HTPR 4

39 89 U HTPR HTPR HTPR HTPR HTPR HTPR HTPR HTPR HTPR 0

40 51 U HTPR HTPR HTPR HTPR HTPR HTPR HTPR HTPR HTPR 0

41 62 U HTPR HTPR HTPR B46 U B46 U B46 U B46 U B46 U B46 U 1

42 47 U HTPR HTPR B46 U B46 U B46 U HTPR B46 U B46 U B46 U 3

43 47 U ND ND ND ND ND ND ND ND ND NA

44 48 U B46 U B46 U ND ND ND ND ND ND ND 1

HTPR high on-treatment platelet reactivity, NA not applicable, ND not done

Serial clopidogrel dose adjustment

123

Using vasodilator-stimulated phosphoprotein (VASP)

Bonello et al. showed that repeated clopidogrel loading

doses to achieve optimal platelet reactivity before stent

placement decreased the rate of ischemic events after one

month without increasing the risk of bleeding in stable

CAD patients [40]. We used a similar protocol of repeated

clopidogrel LD for persistent low responders in interven-

tional group in the first three days after PCI. We observed

that maintaining platelet reactivity in therapeutic window

range by tailoring clopidogrel dose according to MEA

during 12 months of follow-up in ACS patients with initial

HTPR reduced the incidence of adverse events, and thus

adequately protected the patients from future ischemic

events without increasing the risk of bleeding. Therefore,

our results support the hypothesis that HTPR might be a

modifiable risk factor and that serial use of PFT to optimize

platelet reactivity below HTPR threshold to a P2Y12

inhibitor might reduce adverse outcomes in these patients

without inceasing the risk of bleeding. We believe that

optimization of antithrombotic regimens might be impor-

tant both during hospitalization and after discharge in these

high risk patients.

There are several limitations to this study. First, there

was a significant difference in the extension of CAD

(number of diseased vessels) between interventional and

control group that might have had notable influence on

patient outcomes. Chirumamilla et al. showed that greater

extension of CAD is associated with increased platelet

reactivity and could actually be a cause to HTPR pheno-

type [41]. Furthermore, there was a statistically significant

correlation between the complexity of CAD and primary

endpoints in the total study population (p \ 0.05). This

indicates that significantly lesser extent of CAD in the

interventional group might have influenced better control

of platelet reactivity as well as better outcome. Exclusion

of the most sick patients in the study could be another

limitation. Finally, a relatively small patient sample cannot

guarantee significant power, especially for the safety

endpoint.

These results should be confirmed in large, similarly

designed, randomized trials with the use of new antiplatelet

agents as well. The clinical benefit of individualized

treatment based on PFT in ACS patients is still not ade-

quatelly investigated. Large expectations of implementing

personalized antiplatelet treatment based on PFT in ACS

patients are now focused on ongoing trials such as ANT-

ARTIC and TROPICAL ACS.

Acknowledgments This investigation was funded by the Croatian

Ministry of Science, Education and Sports.

Conflict of interest The authors declare that they have no conflict

of interest.

References

1. Hamm CW, Bassand JP, Agewall S et al (2011) ESC Guidelines

for the management of acute coronary syndromes in patients

presenting without persistent ST-segment elevation: the task force

for the management of acute coronary syndromes (ACS) in

patients presenting without persistent ST-segment elevation of the

European society of cardiology (ESC). Eur Heart J 32:2999–3054

2. Steg PG, James SK, Atar D et al (2012) ESC Guidelines for the

management of acute myocardial infarction in patients presenting

with ST-segment elevation. Eur Heart J 33:2569–2619

3. O’Gara PT, Kushner FG, Ascheim DD et al (2013) 2013 ACCF/

AHA guideline for the management of ST-elevation myocardial

infarction: a report of the American College of Cardiology

Foundation/American Heart Association Task Force on practice

guidelines. Circulation 127:e362–e425

4. Jneid H, Anderson JL, Wright RS et al (2012) 2012 ACCF/AHA

focused update of the guideline for the management of patients

with unstable angina/non-ST-elevation myocardial infaction

(updating the 2007 guideline and replacing the 2011 focused

update): a report of the American College of Cardiology Foun-

dation/American Heart Association Task Force on practice

guidelines. Circulation 126:875–910

5. Yusuf S, Zhao F, Mehta SR et al (2001) Effects of clopidogrel in

addition to aspirin in patients with acute coronary syndromes

without ST-segment elevation. N Engl J Med 345:494–502

6. Serebruany VL, Steinhubl SR, Berger PB, Malinin AI, Bhatt DL,

Topol EJ (2005) Variability in platelet responsiveness to clopi-

dogrel among 544 individuals. J Am Coll Cardiol 45:246–251

7. Wiviott SD, Braunwald E, McCabe CH et al (2007) Prasugrel

versus clopidogrel in patients with acute coronary syndromes.

N Eng J Med 357:2001–2015

8. Top 20 global products 2012 (2013) IMS Health Midas. http://

www.imshealth.com/deployedfiles/ims/Global/Content/Corporate/

Press%20Room/Top-Line%20Market%20Data%20&%20Trends/

Top_20_Global_Products_2012_2.pdf. Accessed 19 October 2013

9. Wallentin L, Becker RC, Budaj A et al (2009) Ticagrelor versus

clopidogrel in patients with acute coronary syndromes. N Engl J

Med 361:1045–1057

10. Stone GW, Witzenbichler B, Weisz G et al (2013) Platelet

reactivity and clinical outcomes after coronary artery implanta-

tion of drug-eluting stents (ADAPT-DES): a prospective multi-

centre registry study. Lancet 382:614–623

11. Aradi D, Storey RF, Komocsi A et al (2014) Expert position paper

on the role of platelet function testing in patients undergoing per-

cutaneous coronary intervention. Eur Heart J 35:209–215

12. Tantry US, Bonello L, Aradi D et al (2013) Consensus and update

on the definition of on-treatment platelet reactivity to ADP

associated with ischemia and bleeding. J Am Coll Cardiol

62:2261–2273

13. Mehran R, Rao SV, Bhatt DL et al (2011) Standardized bleeding

definitions for cardiovascular clinical trials: a consensus report

from the BleedingAcademic Research Consortium. Circulation

123:2736–2747

14. Sibbing D, Braun S, Morath T et al (2009) Platelet reactivity after

clopidogrel treatment assessed with point-of-care analysis and

early drug-eluting stent thrombosis. J Am Coll Cardiol

53:849–856

15. Sibbing D, Schulz S, Braun S et al (2010) Antiplatelet effects of

clopidogrel and bleeding in patients undergoing coronary stent

placement. J Thromb Haemost 8:250–256

16. Yusuf S, Mehta SR, Zhao F et al (2003) Early and late effects of

clopidogrel in patients with acute coronary syndromes. Circula-

tion 107:966–972

S. Jure et al.

123

17. Hazarbasanov D, Velchev V, Finkov B et al (2012) Tailoring

clopidogrel dose according to multiple electrode aggregometry

decreases the rate of ischemic complications after percutaneous

coronary intervention. J Thromb Thrombolysis 34:85–90

18. Giles H, Smith REA, Martin J (1994) Platelet glycoprotein IIb-

IIIa and size are increased in acute myocardial infarction. Eur J

Clin Invest 24:69–72

19. Gawaz M, Neumann FJ, Ott I et al (1996) Platelet function in

acute myocardial infarction treated with direct angioplasty. Cir-

culation 93:229–237

20. Muller I, Seyfarth M, Rudiger S et al (2001) Effect of a high

loading dose of clopidogrel on platelet function in patients

undergoing coronary stent placement. Heart 85:92–93

21. Breet NJ, van Werkum JW, Bouman HJ et al (2010) Comparison

of platelet function tests in predicting clinical outcome in patients

undergoing coronary stent implantation. JAMA 303:754–762

22. Trenk D, Hochholzer W, Fromm MF et al (2008) Cytochrome

P450 2C19 681G [ A polymorphism and high on-clopidogrel

platelet reactivity associated with adverse 1-year clinical outcome

of elective percutaneous coronary intervention with drug-eluting

or bare-metal stents. J Am Coll Cardiol 51:1925–1934

23. Price MJ, Nayak KR, Barker CM, Kandzari DE, Teirstein PS

(2009) Predictors of heightened platelet reactivity despite dual-

antiplatelet therapy in patients undergoing percutaneous coronary

intervention. Am J Cardiol 103:1339–1343

24. Angiolillo DJ, Fernandez-Ortiz A, Bernardo E et al (2007) Var-

iability in individual responsiveness to clopidogrel: clinical

implications, management, and future perspectives. J Am Coll

Cardiol 49:1505–1516

25. Lev EI, Patel RT, Maresh KJ et al (2006) Aspirin and clopidogrel

drug response in patients undergoing percutaneous coronary

intervention: the role of dual drug resistance. J Am Coll Cardiol

47:27–33

26. Angiolillo DJ, Fernandez-Ortiz A, Bernardo E et al (2005)

Platelet function profiles in patients with type 2 diabetes and

coronary artery disease on combined aspirin and clopidogrel

treatment. Diabetes 54:2430–2435

27. Cuisset T, Frere C, Quilici J et al (2009) Relationship between

aspirin and clopidogrel responses in acute coronary syndrome and

clinical predictors of non response. Thromb Res 123:597–603

28. Kuchulakanti PK, Chu WW, Torguson R et al (2006) Correlates

and long-term outcomes of angiographically proven stent

thrombosis with sirolimus- and paclitaxel-eluting stents. Circu-

lation 113:1108–1113

29. Price MJ, Berger PB, Teirstein PS et al (2011) Standard- vs high-

dose clopidogrel based on platelet function testing after percu-

taneous coronary intervention: the GRAVITAS randomized trial.

JAMA 305:1097–1105

30. Collet JP, Cuisset T, Range G et al (2012) Bedside monitoring to

adjust antiplatelet therapy for coronary stenting. N Engl J Med

367:2100–2109

31. Campo G, Parrinello G, Ferraresi P et al (2011) Prospective

evaluation of on-clopidogrel platelet reactivity over time in

patients treated with percutaneous coronary intervention rela-

tionship with gene polymorphisms and clinical outcome. J Am

Coll Cardiol 57:2474–2483

32. Parodi G, Marcucci R, Valenti R et al (2011) High residual

platelet reactivity after clopidogrel loading and long-term car-

diovascular events among patients with acute coronary syn-

dromes undergoing PCI. JAMA 306:1215–1223

33. Brar SS, ten Berg J, Marcucci M et al (2011) Impact of platelet

reactivity on clinical outcomes after percutaneous coronary

intervention: a collaborative meta-analysis of individual partici-

pant data. J Am Coll Cardiol 58:1945–1954

34. Aradi D, Pinter T, Magyari B et al (2014) Optimizing P2Y12-

receptor inhibition in acute coronary syndrome after PCI using

platelet function testing: impact of prasugrel versus high-dose

clopidogrel. J Am Coll Cardiol 63:1061–1070

35. Armero S, Camoin Jau L, Omar Aıt Mokhtar O et al (2010) Intra-

individual variability in clopidogrel responsiveness in coronary

artery disease patients under long term therapy. Platelets

21:503–507

36. Gurbel PA, Bliden KP, Hayes KM et al (2005) The relation of

dosing to clopidogrel responsiveness and the incidence of high

post-treatment platelet aggregation in patients undergoing coro-

nary stenting. J Am Coll Cardiol 45:1392–1396

37. Mayer K, Schulz S, Bernlochner I et al. (2014) A comparative

cohort study on personalised antiplatelet therapy in PCI-treated

patients with high on-clopidogrel platelet reactivity. Results of

the ISAR-HPR registry. Thromb Haemost 10;112(2)

38. Siller-Matula JM, Francesconi M, Dechant C et al (2013) Per-

sonalized antiplatelet treatment after percutaneous coronary

intervention: the MADONNA study. Int J Cardiol 167:2018–2023

39. Gurbel PA, Erlinge D, Ohman EM et al (2012) Platelet function

during extended prasugrel and Clopidogrel therapy for patients

with ACS treated without revascularization: the TRILOGY ACS

platelet function substudy. JAMA 308:1785–1794

40. Bonello L, Camoin-Jau L, Armero S et al (2009) Tailored clop-

idogrel loading dose according to platelet reactivity monitoring to

prevent acute and subacute stent thrombosis. Am J Cardiol

103:5–10

41. Chirumamilla AP, Maehara A, Mintz GS et al (2012) High

platelet reactivity on clopidogrel therapy correlates with

increased coronary atherosclerosis and calcification: a volumetric

intravascular ultrasound study. JACC Cardiovasc Imaging

5:540–549

Serial clopidogrel dose adjustment

123


Recommended