+ All Categories
Home > Documents > Vorapaxar: First Global Approval

Vorapaxar: First Global Approval

Date post: 31-Jan-2017
Category:
Upload: shelley
View: 218 times
Download: 0 times
Share this document with a friend
11

Click here to load reader

Transcript
Page 1: Vorapaxar: First Global Approval

R&D INSIGHT REPORT

Vorapaxar: First Global Approval

Raewyn M. Poole • Shelley Elkinson

Published online: 25 June 2014

� Springer International Publishing Switzerland 2014

Abstract Vorapaxar [Zontivity� (US)], an orally active

protease-activated receptor-1 (PAR-1) receptor antagonist,

has been developed by Merck & Co for the reduction of

thrombotic cardiovascular events in patients with a history

of myocardial infarction (MI) or peripheral arterial disease

(PAD). Vorapaxar has received its first global approval for

this indication in the US. This article summarizes the

milestones in the development of vorapaxar leading to this

first approval for the reduction of thrombotic cardiovas-

cular events in patients with a prior MI or PAD.

1 Introduction

Patients with atherothrombosis have a high risk of recur-

rent ischaemic events and therefore require aggressive

secondary prevention therapy [1]. Platelets have a central

role in atherothrombosis, and antiplatelet therapy is there-

fore a mainstay of recommended secondary prevention

measures [2, 3]. Combining antiplatelet agents with dif-

ferent mechanisms of action, for example aspirin and the

thienopyridine clopidogrel, provides greater inhibition of

platelet activation and increased clinical benefit compared

with monotherapy [4]. However, the risk of recurrent

ischaemic events remains high despite the use of dual

antiplatelet therapy with aspirin plus a platelet P2Y12

inhibitor [4–6]. While this risk has been reduced with the

use of more potent antiplatelet agents than clopidogrel,

such as prasugrel and ticagrelor, this has been at the cost of

increased bleeding risk [4, 7]. Furthermore, the relatively

modest reduction in ischaemic events (approximately

20 %) with the more potent P2Y12 inhibitors versus

clopidogrel suggests an alternative mechanism of platelet

inhibition may be required [7].

Thrombin receptor antagonists are a new class of anti-

platelet agents under investigation for use in secondary pre-

vention [4, 6]. Thrombin stimulates protease-activated

receptors (PARs), specific G-protein-coupled cell receptors

that are involved in proliferative and pro-inflammatory pro-

cesses that may have implications in thrombosis and ath-

erosclerosis. Of the four PAR subtypes, only PAR-1 and

PAR-4 are expressed on human platelets [6]. PAR-1 mediates

platelet activation at low thrombin concentrations and is

involved in atherosclerosis, thrombosis and inflammation;

furthermore, it does not appear to be essential for protective

homeostasis [4–6]. These properties make PAR-1 an attrac-

tive target for the development of new antiplatelet agents.

Vorapaxar [Zontivity� (US)] is a first-in-class thrombin

and PAR-1 receptor antagonist developed by Merck & Co

for the secondary prevention of arterial thrombosis. Vora-

paxar received its first global approval in the US on 8 May

2014 as a once-daily tablet for the reduction of thrombotic

cardiovascular events in patients with a history of myo-

cardial infarction (MI) or peripheral arterial disease (PAD)

[8]. The approved labelling includes a black box warning

regarding the increased risk of bleeding, including intra-

cranial haemorrhage (ICH) and fatal bleeding, in patients

This profile has been extracted and modified from the Adis R&D

Insight drug pipeline database. Adis R&D Insight tracks drug

development worldwide through the entire development process,

from discovery, through pre-clinical and clinical studies to market

launch.

R. M. Poole

Adis, Auckland, New Zealand

S. Elkinson (&)

Adis, Level 1, 5 The Warehouse Way, Northcote 0627; Private

Bag 65901, Mairangi Bay 0754, Auckland, New Zealand

e-mail: [email protected]

Drugs (2014) 74:1153–1163

DOI 10.1007/s40265-014-0252-2

Page 2: Vorapaxar: First Global Approval

taking vorapaxar. Vorapaxar is contraindicated in patients

with a history of stroke, transient ischaemic attack (TIA) or

ICH, and those with active pathological bleeding (includ-

ing ICH or peptic ulcer) [8, 9]. Vorapaxar is to be

administered as a tablet containing vorapaxar sulfate

2.5 mg (vorapaxar base 2.08 mg), once daily in addition to

aspirin and/or clopidogrel given according to their indica-

tions or standard of care [9].

The approval is based on data from TRA 2�P-TIMI

50 (NCT00526474; P04737), a multinational phase III

trial that evaluated the efficacy of vorapaxar, given in

addition to the current standard of care, in reducing the

combined endpoint of cardiovascular death, MI or stroke

in [26,000 patients with a history of MI, ischaemic

stroke or documented PAD. The administration of vo-

rapaxar to patients with ischaemic stroke in this trial

was discontinued in January 2011 following recom-

mendations by the Data and Safety Monitoring Board

(DSMB), and the trial was completed in patients with

MI and PAD only (approximately 75 % of enrolled

patients) [10, 11].

Vorapaxar is awaiting approval for the prevention of

arterial thromboembolism in the EU [12]. It is also in phase

III development globally for this indication.

Chemical structure of vorapaxar

N

O

H

NH

HH

H

H H

H

O

O

O

F

Features and properties of vorapaxar

Alternative names MK 5348; MK-5348; SCH 530348; SCH530348; TRA-SCH 530348; Zontivity�

Class 3-ring-heterocyclic-compounds, carbamates, lactones, pyridines

Mechanism of action PAR-1 receptor antagonists; thrombin receptor antagonists

Route of administration Oral

Pharmacodynamics Prolonged, dose-related inhibition of platelet aggregation at therapeutic doses; no

clinically significant effects on coagulation markers

Pharmacokinetics Dose-dependent pharmacokinetics; tmax 1 h; mean Vd *424 L, C99 % plasma protein

binding; multi-exponential disposition with effective t1/2 of 3–4 days, apparent terminal

elimination t1/2 8 days; steady-state reached within 21 days of once-daily dosing, 5- to 6-fold

accumulation. Main route of elimination is by metabolism with CYP3A4 and CYP2J2

involvement; primarily faecal elimination. Concomitant use of strong CYP3A inducers and

inhibitors should be avoided

Adverse events

Most frequent Haemorrhage

Occasional Anaemia, depression, skin reactions, iron deficiency, retinopathy or retinal disorder,

diplopia/oculomotor disturbances

ATC codes

WHO ATC code B01A-X (other antithrombotic agents)

EphMRA ATC code B1X (other antithrombotic agents)

Chemical name Carbamic acid, N-[(1R,3aR,4aR,6R,8aR,9S,9aS)-9-[2-[5-(3-Fluorophenyl)pyridin-

2-yl]ethen-1-yl]-1-methyl-3-oxodecahydronaphtho-[2,3-c]furan-6-yl]-, ethyl ester

1154 R. M. Poole, S. Elkinson

Page 3: Vorapaxar: First Global Approval

1.1 Company Agreements

Vorapaxar was originally being developed by Schering

Plough, which merged with Merck & Co in November

2009 [13].

2 Scientific Summary

2.1 Pharmacodynamics

Vorapaxar is a reversible PAR-1 antagonist, although it is

effectively irreversible due to its long half-life [9]. Vora-

paxar inhibited thrombin-induced and thrombin receptor

agonist peptide (TRAP)- induced platelet aggregation in

in vitro studies, but did not affect adenosine diphosphate

(ADP)-, collagen- or thromboxane mimetic-induced

platelet aggregation.

In studies in healthy volunteers, vorapaxar inhibited

TRAP-induced platelet aggregation by C80 % after

1 week of treatment at the recommended dose, with dose-

and concentration-dependent duration of antiplatelet

activity [9]. TRAP-induced platelet aggregation continued

to be inhibited by 50 % 4 weeks after discontinuation of

treatment with vorapaxar 2.5 mg. There were no changes

in platelet P-selectin and soluble CD40 ligand (sCD40L)

expression or measures of coagulation after single or

multiple doses of vorapaxar.

Vorapaxar produced rapid and prolonged dose-depen-

dent inhibition of platelet aggregation in studies in healthy

volunteers [14, 15]. In two randomized, placebo-controlled

studies, healthy volunteers received either single ascending

doses of vorapaxar 0.25–40 mg or placebo (n = 50),

multiple ascending doses of 1–5 mg/day or placebo for

28 days (n = 36), or a loading dose of 10 or 20 mg then

maintenance doses of 1 mg once daily for 6 days or pla-

cebo (n = 12) [15]. Complete (C80 %) inhibition of

TRAP-induced platelet aggregation was observed with

single vorapaxar doses of 20 and 40 mg, with effects sus-

tained for C72 h post-dose. Complete inhibition was

achieved with multiple doses of vorapaxar 5 mg/day from

day 1 and vorapaxar 1 and 3 mg/day on day 7. No sig-

nificant increases in soluble P-selectin or sCD40L con-

centrations were observed. In a randomized, open-label

study in 111 healthy volunteers, single loading doses of

vorapaxar 20 mg and 40 mg inhibited TRAP-induced

platelet aggregation by 80 % at 1 h, although the 40 mg

dose was required to achieve this in most volunteers [14].

Consistent 80 % platelet inhibition was seen with a 2.5 mg/

day maintenance dose from day 7, but not with mainte-

nance doses of 0.5 or 1 mg/day. Dose-dependent recovery

of platelet function was observed within days or weeks

after stopping treatment with vorapaxar after 28 days.

A pharmacodynamic substudy of the TRACER trial

showed that vorapaxar significantly reduced PAR-1-medi-

ated platelet aggregation compared with placebo [16]. In a

subset of 249 patients with non-ST-segment elevation

acute coronary syndrome (NSTE ACS) enrolled in TRA-

CER, PAR-1-mediated platelet aggregation, as measured

by light transmittance aggregometry (LTA) and VerifyNow

assays, was rapidly and significantly inhibited by vora-

paxar. Inhibition of ADP was significantly greater for vo-

rapaxar versus placebo at 4 h and at 1 month (P \ 0.01),

and the number of PAR-1 receptors was significantly

reduced from baseline in the vorapaxar group (179 vs. 225;

P = 0.004) but not the placebo group.

2.1.1 Effects on QTc Interval

Vorapaxar did not cause significant QT/QTc interval

prolongation in a randomized, double-blind, placebo- and

positive-controlled study in 120 healthy volunteers [17].

Male and female volunteers aged 18–50 years were ran-

domized to receive a single dose of vorapaxar 120 mg or

placebo (double-blind) or moxifloxacin 400 mg (open-

label). Vorapaxar had no significant effect on QTc inter-

val with Fridericia’s correction for heart rate (QTcF), with

the upper bound of the 95 % confidence interval (CI) for

the difference from placebo being B3.8 ms at all time

points during the 23-h post-dose evaluation period. In

contrast, a mean increase in QTcF of [10 ms was seen

with moxifloxacin compared with placebo at 1, 1.5 and 2

h post-dose.

2.2 Pharmacokinetics

Elimination of vorapaxar is via metabolism, which is pri-

marily catalyzed by the cytochrome P450 (CYP) enzymes

CYP3A4 and CYP2J2 [9, 18]. Vorapaxar is metabolized to

a major excreted inactive amine metabolite (M19) by

carbamate cleavage and to the monohydroxy metabolite

(M20) [18]. M20 is the major circulating metabolite of

vorapaxar at steady state and has comparable potency to

the parent compound. The main route of elimination is

faecal (58 % of administered dose), followed by urinary

excretion (25 %) [9].

The pharmacokinetics of vorapaxar are dose-dependent

and steady-state is reached within 21 days of once-daily

administration with 5- to 6-fold accumulation [9]. Vora-

paxar has multi-exponential disposition, with an effective

half-life (t1/2) of 3–4 days and apparent terminal elimina-

tion t1/2 of 8 days. The mean absolute bioavailability of

vorapaxar is 100 %. The mean peak plasma concentration

(Cmax) of vorapaxar occurred 1 hour after administration of

a single 2.5 mg dose under fasted conditions. Vorapaxar

has a mean volume of distribution of approximately 424 L

Vorapaxar: First Global Approval 1155

Page 4: Vorapaxar: First Global Approval

and, along with the active metabolite M20, is almost

completely bound to plasma proteins (C99 %).

In a study in healthy volunteers (n = 111) who received

vorapaxar (loading dose of 5, 10, 20 or 40 mg then main-

tenance doses of 0.5, 1 or 2.5 mg/day for 28 days), expo-

sure was dose-related and the mean terminal t1/2 of

vorapaxar ranged from 165 to 311 h [14]. Vorapaxar also

demonstrated dose-dependent pharmacokinetics in two

studies in healthy volunteers [15]. After administration of

single doses of 0.25–40 mg, the mean Cmax of vorapaxar

ranged from 2.2 to 376.0 ng/mL, time to Cmax (tmax) from

0.8 to 1.5 h, and AUC0-72h from 34 to 6028 ng�h/mL. In

volunteers receiving daily doses of 1, 3 or 5 mg, mean Cmax

ranged from 24.6 to 131.0 ng/mL and AUC from 363 to

1910 ng�h/mL; tmax was 1.0 h for all doses. Vorapaxar had a

long elimination half-life of 126–269 h after administration

of single 20 and 40 mg doses and 173–269 h after multiple

doses of 1–5 mg/day. Mean accumulation ratio (R) ranged

from 4.72 to 6.37. Steady state plasma concentrations were

reached within 21 days in these studies.

Food, concomitant administration of a strong antacid

and older age had modest effects on the pharmacokinetics

of vorapaxar in a randomized, open-label study in healthy

volunteers, although the differences were not considered

clinically significant [19]. A total of 83 healthy volunteers

aged 18–45 years received a single oral dose of vorapaxar

40 mg after a 10-h fast (n = 22), within 5 min of an extra-

strength antacid after a 10-h fast (n = 20), with food

(n = 20), 1 h after food (n = 11) or 2 h after food

(n = 10); in addition, 18 volunteers aged [65 years

received vorapaxar 40 mg after fasting. Vorapaxar had a

median tmax of 1 h in the fasted state; tmax increased to 2–3

h when given with or 1 or 2 h after food. After food, the

area under the plasma concentration time curve (AUC?)

and Cmax increased by 43 and 31 %, respectively. When

given with antacid, AUC? and Cmax decreased by 15 and

38 %, respectively, and the median tmax increased to 2 h.

The AUC? and Cmax of vorapaxar were increased by 41

and 23 %, respectively, in elderly versus younger volun-

teers, but the median tmax was unchanged. The mean t1/2

was also longer in elderly volunteers (370 vs. 240 h for

younger volunteers). The increased exposure in elderly

volunteers was due to decreased clearance (mean 1618 vs.

2280 mL/h for younger volunteers) and a longer t1/2 (370

vs. 240 h for younger volunteers). An open-label two-

period crossover study in 16 healthy volunteers also

showed that food did not significantly affect the pharma-

cokinetics of a single oral dose of vorapaxar 2.5 mg [20].

Geometric mean ratios (GMRs) for the AUC0–72 h and

Cmax of vorapaxar in the fed and fasted states were 96.9

(90 % CI 92.2–102) and 79.1 (90 % CI 67.6–92.5),

respectively; both 90 % CIs were within the pre-specified

range for comparability of 0.50–2.00.

The pharmacokinetics and pharmacodynamics of vora-

paxar do not appear to be affected by ethnicity. There were

no clinically meaningful differences in pharmacokinetics or

inhibition of platelet aggregation with vorapaxar between

age-, sex-, height-, and bodyweight-matched Japanese and

Caucasian volunteers [21]. The pharmacokinetics of vora-

paxar and its major active metabolite, M20, were also

similar in Chinese and US healthy volunteers [22].

2.2.1 Effects of Renal or Hepatic Impairment

No dose adjustment is required in patients with renal

impairment or mild or moderate hepatic impairment [9].

However, the use of vorapaxar in patients with severe hepatic

impairment is not recommended due to the increased risk of

bleeding. Hepatic impairment did not have a clinically

meaningful effect on the pharmacokinetics of vorapaxar

(40 mg) in a single-dose study in patients with mild (Child–

Pugh 5–6; n = 6), moderate (Child–Pugh 7–9; n = 6) or

severe (Child–Pugh 10–15; n = 4) hepatic impairment and

age-, sex-, height- and bodyweight-matched healthy volun-

teers (n = 16) [23]. Exposure to vorapaxar or its active

metabolite M20 and t1/2 values did not appear to correlate

with the severity of hepatic impairment. Mean AUCfinal and

Cmax values for vorapaxar ranged from 14,200 to

18,000 ng�h/mL and 206–279 ng/mL, respectively; although

the lowest values occurred in patients with severe hepatic

impairment, the 90 % CIs for the GMRs for AUCfinal and

Cmax included 100 and no trend between increased GMR and

increased hepatic impairment was observed. The median tmax

ranged from 1.0 to 1.75 h, and mean t1/2 from 298 to 366 h.

End-stage renal disease had no clinically significant

effects on the pharmacokinetics or pharmacodynamics of a

single oral dose of vorapaxar 10 mg in an open-label study

in eight patients on haemodialysis and seven matched

healthy volunteers [24].

2.2.2 Interactions with Other Drugs

Concomitant treatment with strong CYP3A inhibitors (C5-

fold increase in AUC of substrate or [80 % decrease in

clearance), such as ketoconazole itraconazole, posaconaz-

ole, clarithromycin, telithromycin, nefadozone, ritonavir,

saquinavir, nelfinavir, indinavir, boceprevir, telaprevir and

conivaptan, and strong CYP inducers (C80 % decrease in

substrate AUC), such as rifampin carbamazepine, phenyt-

oin and St. John’s Wort, should be avoided in patients

receiving vorapaxar [9]. However, no increase in bleeding

risk or reduction in the efficacy of vorapaxar was observed

in patients taking weak or moderate CYP3A inhibitors in

phase III studies, and dose adjustment is not considered

necessary for concomitant administration of these drugs

with vorapaxar [9].

1156 R. M. Poole, S. Elkinson

Page 5: Vorapaxar: First Global Approval

No dose adjustment is required for vorapaxar in patients

treated with antacids or proton pump inhibitors or rosig-

litazone [9]. No significant pharmacokinetic interaction

between vorapaxar and prasugrel was observed after mul-

tiple-dose administration at steady-state, and dose adjust-

ment is not considered necessary; however, clinical

experience of vorapaxar in patients treated with prasugrel

is limited. Although there has been no study of the phar-

macokinetic interaction between vorapaxar and clopido-

grel, concomitant use is supported by the results of the

TRA 2�P-TIMI 50 and TRACER studies.

The effects of ketoconazole (400 mg once daily for

28 days) and rifampin (600 mg once daily for 28 days) on

the pharmacokinetics of vorapaxar (20 mg on day 7 then

2.5 mg/day once daily for 21 days) were examined in an

open-label, placebo-controlled, parallel-group study in

healthy volunteers [25]. Compared with placebo, concomi-

tant administration of ketoconazole 400 mg once daily

increased the steady state AUC0–24 and Cmax of vorapaxar by

196 and 193 %, respectively, whereas concomitant admin-

istration of rifampin reduced the AUC0–24 and Cmax of

vorapaxar to 45.5 and 61.4 % of control, respectively.

Although no significant pharmacokinetic or pharmaco-

dynamic interaction between vorapaxar and warfarin was

observed in a study in healthy volunteers, concomitant use

should be avoided due to the increased risk of bleeding

events [9]. In an open-label, two-period crossover study in

12 healthy male volunteers, concomitant administration of

vorapaxar (2.5 mg/day on days 1–6) with warfarin 25 mg

did not significantly affect exposure to R- or S-warfarin,

prothrombin time (PT) or international normalized ratio

(INR) [26]. Compared with warfarin 25 mg alone, GMRs

were 108 and 105, respectively, for the AUC? of R- and

S-warfarin; corresponding values for Cmax were 105 and

105. Estimated GMRs for the AUC0-120h for PT and INR

were 97 and 96, respectively.

Vorapaxar also showed no clinically significant inter-

action with digoxin in a study in healthy volunteers [27]. In

this open-label, fixed-sequence, two-period crossover

study, 18 volunteers received digoxin 0.5 mg on day 1 or

vorapaxar 2.5 mg/day on days 1–6 and a single dose of

vorapaxar 40 mg plus digoxin 0.5 mg on day 7, with a

washout period of C8 days in between treatments.

Although the Cmax of digoxin was increased (GMR of 154),

the AUCfinal and tmax and the pharmacodynamics of

digoxin, as evaluated by changes in electrocardiogram

(ECG) parameters (ventricular rate, QRS, PR, QT and QTc

intervals), were similar for the two treatments.

2.3 Therapeutic Trials

Vorapaxar has been evaluated in two major multinational

randomized, double-blind, placebo-controlled phase III

trials, TRA 2�P-TIMI 50 (NCT00526474) in patients with

MI, PAD or ischaemic stroke and TRACER

(NCT00527943) in patients with NSTE ACS. These studies

assessed the effects of adding vorapaxar to standard ther-

apy on ischaemic outcomes and bleeding risk, as assessed

by GUSTO (Global Utilization of Streptokinase and Tissue

Plasminogen Activator for Occluded Arteries) and TIMI

(Thrombolysis In Myocardial Infarction) criteria.

2.3.1 TRA 2�P-TIMI 50 Trial

Vorapaxar significantly reduced the risk of cardiovascular

death or ischaemic events in patients with a history of MI

or ischaemic stroke (within the previous 2–52 weeks) or

PAD in the TRA 2�P-TIMI 50 trial [11]. This trial enrolled

26,449 patients who were randomized to receive vorapaxar

2.5 mg (n = 13,225) or placebo (n = 13,224), once daily

in addition to standard therapy. In January 2011, after a

median follow-up duration of 24 months, study medication

was discontinued for all patients with a history of ischae-

mic stroke (including new strokes occurring during the

trial) after the DSMB reported an excess of ICH in this

population; treatment was continued for patients with no

history of stroke, as per the DSMB recommendations. The

primary endpoint of the study was originally the composite

of cardiovascular death, MI, stroke or urgent coronary

revascularization (UCR); however, this became the main

secondary endpoint and the primary endpoint was changed

to the composite of cardiovascular death, MI or stroke after

a review of data from the TRACER trial (NCT00527943)

by the TRA 2�P-TIMI 50 investigators before the database

was locked and while blinded treatment was ongoing.

In the intention-to-treat (ITT) population, including

patients with stroke, a significant reduction in the primary

endpoint of cardiovascular death, MI or stroke was

observed with vorapaxar, compared with placebo [9.3 vs.

10.5 %; hazard ratio (HR) 0.87; 95 % CI 0.80–0.94;

P \ 0.001] at 3 years [11]. Significant reductions were

observed with vorapaxar versus placebo for the main sec-

ondary endpoint (11.2 vs. 12.4 %; HR 0.88, 95 % CI

0.82–0.95; P = 0.001), and cardiovascular death or MI

(7.3 vs. 8.2 %; HR 0.86, 95 % CI 0.78–0.94; P = 0.002)

and MI (5.2 vs. 6.1 %; HR 0.83, 95 % CI 0.74–0.93;

P = 0.001). However, there were no significant differences

in the rates of cardiovascular death, all-cause mortality, any

stroke, ischaemic stroke and UCR. Data from patients with

no history of stroke also showed that vorapaxar signifi-

cantly reduced the primary composite endpoint (8.3 vs.

9.6 %; HR 0.84, 95 % CI 0.76–0.93; P \ 0.001). Arterial

revascularization rates were significantly reduced with

vorapaxar at 3 years (13.6 vs. 15.5 %; HR 0.89, 95 % CI

0.83–0.95; P \ 0.001) [28]. Significant reductions were

seen in both peripheral and coronary revascularization,

Vorapaxar: First Global Approval 1157

Page 6: Vorapaxar: First Global Approval

including CABG surgery, and elective revascularizations

(P \ 0.05 for all).

No significant heterogeneity was observed for the pri-

mary endpoint across most major patient subgroups, with

the exception of bodyweight (P = 0.03 for interaction)

[11]. The HR for the primary endpoint in patients weighing

C60 kg was 0.85 (95 % CI 0.78–0.92), whereas in patients

weighing \60 kg it was 1.22 (95 % CI 0.88–1.69).

The efficacy of vorapaxar in reducing the primary

endpoint was independent of thienopyridine therapy [29].

Thienopyridine therapy was planned for 15,356 (58 %) of

the 26,449 patients enrolled in the study and 24,734 (94 %)

received aspirin (recommended dose B162 mg). Signifi-

cant and similar reductions in the primary endpoint were

seen regardless of whether thienopyridine therapy was

planned (p value for interaction 0.64) or whether it was

used at baseline or at 18 months (p values for interaction

0.76 and 0.99, respectively).

Analysis of net clinical outcomes, which included

bleeding as well as efficacy endpoints, showed a significant

difference for vorapaxar versus placebo for all-cause

mortality, MI, stroke or GUSTO severe bleeding (11.9 vs.

12.8 %; HR 0.92, 95 % CI 0.85–0.99; P = 0.02) in the ITT

population [11]. However, no significant differences were

seen for the net clinical outcomes of cardiovascular death,

MI, stroke or GUSTO moderate or severe bleeding (11.7

vs. 12.1 %; HR 0.97, 95 % CI 0.90–1.04; P = 0.40) and

cardiovascular death, MI, stroke, UCR or GUSTO mod-

erate or severe bleeding (13.4 vs. 14.0 %; HR 0.96, 95 %

CI 0.89–1.02; P = 0.20).

A pre-specified subgroup analysis of patients with a

prior MI as the qualifying event for enrolment

(n = 17,779) showed a significant reduction in the primary

endpoint with vorapaxar compared with placebo [30]. After

a median follow-up of 2.5 years, primary endpoint events

had occurred in 610 of 8,898 vorapaxar recipients, com-

pared with 750 of 8,881 patients in the placebo group,

corresponding to 3-year Kaplan–Meier estimates of 8.1 and

9.7 %, respectively (HR 0.80, 95 % CI 0.72–0.89;

P \ 0.0001). Vorapaxar was also associated with signifi-

cant reductions in the risk of the main secondary endpoint

(10.5 vs. 12.1 %; HR 0.83, 95 % CI 0.76–0.92;

P = 0.0001) and the individual endpoints of MI (5.7 vs.

7.0 %; HR 0.79, 95 % CI 0.70–0.90; P = 0.0003) and

ischaemic stroke (1.0 vs. 1.4 %; HR 0.66, 95 % CI

0.48–0.89; P = 0.006) at 3 years. However, there were no

significant differences in cardiovascular death or all-cause

mortality rates. Reductions in the primary endpoint with

vorapaxar were independent of aspirin dose (\100 mg,

100–162 mg or [162 mg; p value for interaction 0.98)

[31]. Vorapaxar also had comparable efficacy in patients

who qualified with MI whether or not they had diabetes

mellitus, with significant reductions in the primary

endpoint in both those with diabetes (12.6 vs. 15.7 %;

P = 0.004) and those without diabetes (6.8 vs. 7.9 %;

P = 0.005) [32].

A pre-specified analysis of patients with a prior

ischaemic stroke (n = 4883) showed no significant

reduction in the primary endpoint with vorapaxar com-

pared with placebo (13.0 vs. 11.7 %; HR 1.03, 95 % CI

0.85–1.25; P = 0.75) [33]. There were also no significant

differences in secondary efficacy endpoints, including

recurrent ischaemic stroke.

A subgroup analysis of patients with PAD (n = 3787)

also showed no significant reduction in the primary end-

point (11.3 vs. 11.9 %; HR 0.94, 95 % CI 0.78–1.14;

P = 0.53) or the main secondary endpoint (12.7 vs.

13.4 %; HR 0.95, 95 % CI 0.79–1.14; P = 0.57) with

vorapaxar [34]. However, in patients with PAD vorapaxar

was associated with reduced rates of hospitalization for

acute limb ischaemia (2.3 vs. 3.9 %; HR 0.58, 95 % CI

0.39–0.86; P = 0.006) and peripheral artery revasculari-

zation (18.4 vs. 22.2 %; HR 0.84, 95 % CI 0.73–0.97;

P = 0.017) than placebo; both urgent and elective

peripheral revascularization were significantly reduced in

the vorapaxar group (P \ 0.05 for both). The rates of the

composite endpoints of cardiovascular death, MI, stroke or

urgent vascular hospitalization and cardiovascular death,

MI, stroke, urgent vascular hospitalization and revascu-

larization were also significantly reduced with vorapaxar

compared with placebo (both P \ 0.05). Patients in the

PAD subgroup of this study were required to have a history

of intermittent claudication and an ankle-brachial index of

\0.85 or previous revascularization for limb ischaemia.

More than half (57 %) of the patients in this subgroup also

had coronary artery disease (CAD), and 14 % had a history

of stroke or TIA.

Treatment with vorapaxar was associated with a sig-

nificant reduction in the risk of definite stent thrombosis,

according to Academic Research Consortium criteria [35].

A total of 14,042 (53 %) patients had undergone a prior

coronary stent placement, and another 449 patients

received a stent during the follow-up period. After 3 years,

definite stent thrombosis had occurred in 1.1 % of vora-

paxar recipients, compared with 1.4 % of placebo-treated

patients (HR 0.71; P = 0.40). Similar results were seen

when patients who qualified for the study with MI and had

undergone stent placement before or during the study

(13,489 of 17,779; 76 %) were analysed separately, with

3-year definite stent thrombosis rates of 1.1 and 1.4 %,

respectively for the vorapaxar and placebo groups (HR

0.71; P = 0.46).

An analysis of strokes occurring during the study in

patients with no prior history of cerebrovascular disease

showed that vorapaxar significantly reduced the risk of

ischaemic stroke compared with placebo (HR 0.57, 95 %

1158 R. M. Poole, S. Elkinson

Page 7: Vorapaxar: First Global Approval

CI 0.43–0.75; P \ 0.001) [36]. Haemorrhagic stroke

occurred more frequently in patients treated with vorapaxar

(HR 2.78, 95 % CI 1.00–7.73; P = 0.049); however, due

to the reduction in ischaemic stroke the overall stroke risk

was lower for vorapaxar than placebo (HR 0.68, 95 % CI

0.52–0.80; P = 0.001). There were no significant differ-

ences between vorapaxar and placebo in the risk of

haemorrhagic conversion and death among patients with a

first ischaemic stroke.

2.3.2 TRACER Trial

The addition of vorapaxar to standard therapy did not

significantly reduce the primary endpoint of cardiovascular

death, MI, stroke, rehospitalization due to recurrent

ischaemia or UCR in patients with NSTE ACS in the

TRACER study [37]. In this trial, 12,944 patients were

randomized to receive vorapaxar (40 mg loading dose then

2.5 mg once daily maintenance dose) or placebo in addi-

tion to standard therapy. Follow-up was terminated early

on the recommendation of the study’s DSMB after a safety

review; at this time the target number of primary endpoint

events specified in the study protocol had been reached.

Two-year Kaplan–Meier rates of the primary endpoint in

the vorapaxar and placebo groups were 18.5 and 19.9 %,

respectively (HR 0.92, 95 % CI 0.85–1.01; P = 0.07). A

significant reduction in the main secondary endpoint of

cardiovascular death, MI or stroke was seen with vorapaxar

compared with placebo (14.7 vs. 16.4 %; HR 0.89, 95 %

CI 0.81–0.98; P = 0.02) and the composite endpoint of

cardiovascular death or MI was also reduced with vora-

paxar (HR 0.90, 95 % CI 0.81–0.99; P = 0.03). Results

were consistent across patient subgroups, including age

[38], presence or absence of PAD [39], aspirin dose [40],

glycoprotein (GP) IIb/IIIa inhibitor use [41], or CYP2C19

genotype or paraoxonase-1 activity [42].

Vorapaxar reduced the risk of a first MI (HR 0.88, 95 %

CI 0.79–0.98; P = 0.021) and first and subsequent MIs

(HR 0.86, 95 % CI 0.77–0.97; P = 0.014) compared with

placebo [43]. Analysis by type of MI showed that the most

frequent were type 1 MI (related to ischaemia caused by

atherothrombosis), which accounted for 1,025 of 1,580 MIs

occurring during the follow-up period, and that vorapaxar

significantly reduced the risk of this type of MI (HR 0.83,

95 % CI 0.73–0.95; P = 0.007). The second most common

type of MI was percutaneous coronary intervention (PCI)-

related (type 4a), with 352 (22.3 %) of the total MIs being

of this type; vorapaxar did not significantly reduce the risk

of PCI-related MI (HR 0.90, 95 % CI 0.73–1.12;

P = 0.35).

Analysis of net clinical outcomes for the main secondary

endpoint showed that, when offset by the risk of GUSTO

severe bleeding or TIMI major bleeding, the net clinical

benefit with vorapaxar at 1 year was 0.34 % [44]. When

patients were stratified according to the risks of ischaemic

events ([13 vs. B13 %) and major bleeding ([5 vs.

B5 %), those with high ischaemic risk and low bleeding

risk had a net benefit (2.8 %); however, negative benefits

were seen for those with high bleeding and low ischaemic

risk (-2.93 %), high bleeding and high ischaemic risk

(-3.08 %), and low bleeding and low ischaemic risk

(-0.10 %).

Results for medically managed patients (n = 4194)

were consistent with those for the overall population, with

no significant difference in the primary composite endpoint

with vorapaxar versus placebo [45]. Although there was

also no significant difference for the main secondary end-

point, the rate of MI was significantly reduced with vora-

paxar (HR 0.79, 95 % CI 0.63–0.99).

There were no significant differences in the primary

endpoint or the main secondary endpoint for vorapaxar

versus placebo in the subgroup of patients who underwent

PCI during the index hospitalization (n = 7,479) [46]. In

contrast, a pre-specified subgroup analysis showed that

vorapaxar was associated with a significant 45% reduction

in the risk of the primary endpoint in patients who under-

went CABG surgery during the index hospitalization

(n = 1312) [47]. Primary endpoint events occurred in

8.2 % of vorapaxar recipients and 12.9 % of placebo

recipients in the CABG subgroup (HR 0.55, 95 % CI

0.36–0.83; P = 0.005), with no significant increase in

CABG-related TIMI major bleeding (HR 1.36, 95 % CI

0.92–2.02; P = 0.12).

2.3.3 Phase II Trials

Vorapaxar was associated with a non-significant reduction

in the risk of death or major adverse cardiac events

(MACE) in patients with symptoms of CAD undergoing

non-urgent PCI or angiography with intent to perform PCI

in the TRA-PCI trial (NCT00132912) [48]. This random-

ized, double-blind, multinational study enrolled a total of

1030 patients who were randomized to receive a single

loading dose of vorapaxar 10, 20 or 40 mg or placebo at

least 1 hour before PCI; those who subsequently underwent

PCI then received a maintenance dose of vorapaxar 0.5, 1

or 2 mg/day or placebo for 60 days. Among the patients

who underwent PCI, the 60-day rates of death or MACE

were 9 % for placebo recipients, compared with 6 % for

patients receiving vorapaxar (range 5–8 % for loading

doses of 10, 20 or 40 mg).

Vorapaxar significantly reduced the risk of peri-proce-

dural MI in Japanese patients with NSTE ACS undergoing

planned PCI in a randomized, double-blind multicentre

phase II study (NCT00684203) [49]. This study enrolled

117 patients who were randomized 4:1 to receive

Vorapaxar: First Global Approval 1159

Page 8: Vorapaxar: First Global Approval

vorapaxar 20 or 40 mg loading dose or placebo, followed

by vorapaxar 1 or 2.5 mg/day maintenance dose or placebo

for 60 days for those who underwent PCI, in addition to the

standard of care (aspirin, ticlopidine and heparin). There

were no deaths or MACE other than MI among patients

who underwent PCI during 60 days’ follow-up. Non-fatal

peri-procedural MI occurred in 16.9 % of vorapaxar

recipients compared with 42.9 % of patients who received

only the standard of care (P = 0.013); the incidences of MI

in the vorapaxar 20 and 40 mg loading dose cohorts were

similar.

2.4 Adverse Events

Vorapaxar is associated with an increased risk of bleeding,

including serious and potentially fatal bleeding events [9].

The increase in bleeding risk is proportional to patients’

underlying risk of bleeding, with known risk factors

including renal or hepatic impairment, older age, lower

bodyweight, prior history of bleeding disorders, and con-

comitant use of certain medications (including anticoagu-

lants and fibrinolytics). Vorapaxar is contraindicated in

patients with a prior history of stroke, TIA or ICH due to

their increased risk of ICH; it is also contraindicated in

patients with active pathological bleeding, including ICH

and peptic ulcers.

Bleeding was assessed in the TRA 2�P-TIMI 50 trial

according to GUSTO and TIMI criteria, with GUSTO

moderate or severe bleeding being the primary safety

endpoint of the study [11]. In the ITT population (including

patients with a history of stroke), vorapaxar was associated

with increased rates of GUSTO moderate or severe

bleeding (4.2 vs. 2.5 %; HR 1.43–1.93; P \ 0.001) and

TIMI clinically significant bleeding 15.8 vs. 11.1 %; HR

1.46, 95 % CI 1.36–1.57; P \ 0.001) compared with pla-

cebo. The increased risk of GUSTO moderate or severe

bleeding was seen across the major patient subgroups.

Although non-CABG-related TIMI major bleeding was

significantly increased in the vorapaxar group compared

with the placebo group (2.8 vs. 1.8 %; HR 1.46, 95 % CI

1.22–1.75; P \ 0.001), the difference in CABG-related

TIMI major bleeding was not statistically significant (7.6

vs. 6.1 %; HR 1.13, 95 % CI 0.48–2.66; P = 0.79). ICH

occurred in 102 (1.0 %) patients in the vorapaxar group,

compared with 53 (0.5 %) in the placebo group (HR 1.94,

95 % CI 1.39–2.70; P \ 0.001); this included a significant

increase in the risk of intracerebral bleeding (0.8 vs. 0.4 %;

HR 2.19, 95 % CI 1.51–3.17; P \ 0.001). Rates of fatal

bleeding were not significantly different for vorapaxar and

placebo (0.3 vs. 0.2 %; HR 1.46, 95 % CI 0.82–2.58;

P = 0.19). In patients with a history of stroke, ICH

occurred in 2.4 % of vorapaxar recipients, compared with

0.9 % of patients in the placebo group (P \ 0.001) but

fatal bleeding rates were not significantly different (0.5 vs.

0.3 %; P = 0.46). ICH and fatal bleeding rates in patients

with no history of stroke treated with vorapaxar and pla-

cebo were 0.6 vs. 0.4 % (P = 0.049) and 0.3 vs. 0.2 %

(P = 0.30), respectively.

Analysis of bleeding rates in the 20,108 patients who

qualified for the TRA 2�P-TIMI 50 study with MI or PAD

and had no history of stroke showed an increased risk of

GUSTO moderate or severe bleeding with vorapaxar

compared with placebo, with Kaplan-Meier estimates of

3.7 vs. 2.4 % at 1080 days (HR 1.55, 95 % CI 1.30–1.86)

[9]. Although vorapaxar was associated with higher rates of

ICH and fatal bleeding than placebo, the increases were not

statistically significant (HR 1.46, 95 % CI 0.92–2.31 and

HR 1.15, 95 % CI 0.56–2.36, respectively). However, rates

of clinically significant bleeding (HR 1.47, 95 % CI

1.35–1.60), including gastrointestinal bleeding (HR 1.37,

95 % CI 1.18–1.59) were increased with vorapaxar. When

patients who qualified for study with PAD (n = 3,787) and

Clinical trials of vorapaxar in the secondary prevention of arterial thrombosis conducted by Merck & Co

Drugs Patient subgroup Study

phase

Status Location Trial identifiers

Vorapaxar ? SOC Patients with atherosclerosis III Completed Multinational NCT00526474; P04737

(TRA 2�P-TIMI 50)

Vorapaxar ? SOC (e.g. aspirin,

clopidogrel)

Patients with ACS III Terminated Multinational NCT00527943; P04736

(TRACER)

Vorapaxar ? SOC Assessment of long-term ocular safety

in study NCT00526474

III Completed Multinational NCT00617123; P05183

Vorapaxar ? SOC Patients undergoing non-emergent

PCI

II Completed Multinational NCT00132912; P03573

(TRA-PCI)

Vorapaxar ? aspirin ? clopidogrel Patients with ACS II Completed Japan NCT00684203; P04772

Vorapaxar ? aspirin Patients with cerebral infarction

(safety study)

II Completed Japan NCT00684515; P05005

ACS Acute coronary syndrome, SOC standard of care

1160 R. M. Poole, S. Elkinson

Page 9: Vorapaxar: First Global Approval

MI (n = 17,779) were analysed separately, rates of

GUSTO moderate or severe bleeding were significantly

increased with vorapaxar versus placebo for both patients

with PAD (7.4 vs. 4.5 %; HR 1.62, 95 % CI 1.21–2.18;

P = 0.001) and those with MI (3.4 vs. 2.1 %; HR 1.61,

95 % CI 1.31–1.97; P \ 0.0001) [30, 34]. However, rates

of GUSTO severe bleeding, fatal bleeding and ICH were

not significantly different for vorapaxar and placebo in

either of these subgroups.

A pre-specified analysis of patients who qualified for the

TRA 2�P-TIMI 50 study with a prior ischaemic stroke

showed that, compared with placebo, vorapaxar was

associated with significant increases in the rates of GUSTO

moderate or severe bleeding (4.2 vs. 2.4 %; HR 1.93, 95 %

CI 1.33–2.79; P \ 0.001), GUSTO severe bleeding (3.0 vs.

1.6 %; HR 2.09, 95 % CI 1.31–3.34; P = 0.002) and ICH

(2.5 vs. 1.9 %; HR 2.52, 95 % CI 1.46–4.36; P \ 0.001)

[33]. Rates of TIMI clinically significant bleeding, TIMI

non-CABG major bleeding and intracerebral haemorrhage

were also significantly higher for vorapaxar than placebo;

however, the difference in fatal bleeding rates was not

statistically significant.

In the TRACER study in patients with NSTE ACS,

vorapaxar was associated with significant increases in the

rates of GUSTO moderate or severe bleeding (7.2 vs.

5.2 %; HR 1.35, 95 % CI 1.16–1.58; P \ 0.001) and TIMI

clinically significant bleeding (20.2 vs. 14.6 %; HR 1.43,

95 % CI 1.31–1.57; P \ 0.001) compared with placebo

[37]. The rates of GUSTO severe bleeding (2.9 vs. 1.6 %;

HR 1.66, 95 % CI 1.27–2.16; P \ 0.001), ICH (1.1 vs.

0.2 %; HR 3.39, 95 % CI 1.78–6.45; P \ 0.001) and TIMI

major bleeding (4.0 vs. 2.5 %; HR 1.53, 95 % CI

1.24–1.90; P \ 0.001) were also significantly higher for

vorapaxar versus placebo. However, there were no signif-

icant differences between vorapaxar and placebo with

respect to fatal bleeding and CABG-related bleeding dur-

ing the index hospitalization. While GUSTO moderate or

severe bleeding rates were increased with vorapaxar in

most patient subgroups, the risk was higher for patients

who were receiving a thienopyridine at baseline than those

who were not (P = 0.04 for interaction) and for patients

whose bodyweight was below the median than for those

with higher bodyweight (P = 0.03 for interaction).

Examination of patient and clinical characteristics associ-

ated with major bleeding found that age, history of PAD,

past and present tobacco use, creatinine clearance, hae-

moglobin level at presentation, T-wave inversion on ECG,

use of GP IIb/IIIa inhibitors and Kilip class 3–4 were

independent predictors of major bleeding [50].

Anaemia was the most common non-haemorrhagic

adverse event in a total of 19,632 patients treated with

vorapaxar in the TRA 2�P-TIMI 50 and TRACER studies,

occurring in 5.0 % of vorapaxar recipients compared with

4.0 % of the 19,607 placebo-treated patients [9]. Other

adverse events occurring in [2 % of patients treated with

vorapaxar in these trials were depression (2.4 vs. 2.1 % for

placebo) and rashes, eruptions and exanthemas (2.2 vs.

2.0 %). Diplopia and related oculomotor disturbances

occurred in 0.2 % of vorapaxar recipients (30 patients),

compared with 0.06 % of placebo recipients (10 patients)

in these trials.

2.5 Ongoing Clinical Trials

No ongoing clinical trials of vorapaxar are listed in the

clinicaltrials.gov database.

3 Current Status

Vorapaxar received its first global approval on 8 May 2014

for the reduction of thrombotic cardiovascular events in

patients with a history of MI or with PAD in the US. It is

approved for use with aspirin and/or clopidogrel according

to their indications or standard of care.

Disclosure The preparation of this report was not supported by any

external funding. During the peer review process the manufacturer of

the agent under review was offered an opportunity to comment on the

article. Changes resulting from any comments received were made by

the authors on the basis of scientific completeness and accuracy. R.M.

Poole is a contracted employee of Adis, Springer SBM. S. Elkinson is

a salaried employee of Adis, Springer SBM.

References

1. Steg PG, Dorman SH, Amarenco P. Atherothrombosis and the

role of antiplatelet therapy. J Thromb Haemost. 2011;9(Suppl

1):325–32.

2. Smith SC Jr, Benjamin EJ, Bonow RO, et al. AHA/ACCF Sec-

ondary Prevention and Risk Reduction Therapy for Patients with

Coronary and other Atherosclerotic Vascular Disease: 2011

update: a guideline from the American Heart Association and

American College of Cardiology Foundation. Circulation. 2011;

124(22):2458–73.

3. Anderson JL, Adams CD, Antman EM, et al. 2011 ACCF/AHA

Focused Update Incorporated Into the ACC/AHA 2007 Guide-

lines for the Management of Patients With Unstable Angina/Non-

ST-Elevation Myocardial Infarction: a report of the American

College of Cardiology Foundation/American Heart Association

Task Force on Practice Guidelines. Circulation. 2011;123(18):

e426–579.

4. Leonardi S, Tricoci P, Becker RC. Thrombin receptor antagonists

for the treatment of atherothrombosis: therapeutic potential of

vorapaxar and E-5555. Drugs. 2010;70(14):1771–83.

5. Angiolillo DJ. The evolution of antiplatelet therapy in the treat-

ment of acute coronary syndromes: from aspirin to the present

day. Drugs. 2012;72:2087–116.

6. Cho JR, Rollini F, Franchi F, et al. Unmet needs in the man-

agement of acute myocardial infarction: Role of novel protease-

activated receptor-1 antagonist vorapaxar. Vasc Health Risk

Manag. 2014;10:177–88.

Vorapaxar: First Global Approval 1161

Page 10: Vorapaxar: First Global Approval

7. Gurbel PA, Jeong Y-H, Tantry US. Vorapaxar: a novel protease-

activated receptor-1 inhibitor. Expert Opin Investig Drugs. 2011;

20(10):1445–53.

8. FDA. FDA approves Zontivity to reduce the risk of heart attacks

and stroke in highrisk patients. http://www.fdagov/NewsEvents/

Newsroom/PressAnnouncements/ucm396585htm. 2014.

9. Prescribing information. http://www.merckcom/product/usa/pi_

circulars/z/zontivity/zontivity_pipdf. 2014.

10. Merck. Merck Statement on Changes to Clinical Studies of Vo-

rapaxar. Media Release. 2011.

11. Morrow DA, Braunwald E, Bonaca MP, et al. Vorapaxar in the

secondary prevention of atherothrombotic events. N Engl J Med.

2012;366(15):1404–13.

12. European Medicines Agency. Applications for new human

medicines under evaluation by the Committee for Medicinal

Products for Human Use, January 2014.

13. Merck. New Merck Begins Operations. Media Release. 2009.

14. Kosoglou T, Reyderman L, Kasserra C, et al. Optimizing dose of

the novel thrombin receptor antagonist SCH 530348 based on

pharmacodynamics and pharmacokinetics in healthy subjects.

Clin Pharmacol Ther. 2008;83(Suppl. 1):55.

15. Kosoglou T, Reyderman L, Tiessen RG, et al. Pharmacodynamics

and pharmacokinetics of the novel PAR-1 antagonist vorapaxar

(formerly SCH 530348) in healthy subjects. Eur J Clin Pharma-

col. 2012;68(3):249–58.

16. Storey RF, Kotha J, Smyth S, et al. Effects of vorapaxar on

platelet reactivity and biomarker expression in non-ST-elevation

acute coronary syndromes. The TRACER Pharmacodynamic

Substudy. Thromb Haemost. 2014;111(5):883–91.

17. Kosoglou T, Hunt TL, Xuan F, et al. Effect of the thrombin

receptor antagonist (PAR-1) vorapaxar on QT/QTc interval in

healthy volunteers: A randomized, placebo- and positive-con-

trolled, parallel group trial. Clin Pharmacol Drug Dev. 2014;3(1):

18–24.

18. Ghosal A, Lu X, Penner N, et al. Identification of human liver

cytochrome P450 enzymes involved in the metabolism of SCH

530348 (Vorapaxar), a potent oral thrombin protease-activated

receptor 1 antagonist. Drug Metab Dispos. 2011;39(1):30–8.

19. Kosoglou T, Reyderman L, Tseng J, et al. Effect of food, antacid,

and age on the pharmacokinetics of the oral thrombin receptor

antagonist vorapaxar (sch 530348) in healthy volunteers. Clin

Pharmacol Drug Dev. 2013;2(3):223–30.

20. Behm MO, Kosoglou T, Miltenburg AMM, et al. The absence of

a clinically significant effect of food on the single dose phar-

macokinetics of Vorapaxar, a PAR-1 antagonist, in healthy adult

subjects. Clin Pharmacol Drug Dev. 2013;2(4):310–5.

21. Kosoglou T, Reyderman L, Kasserra C, et al. No differences in

the pharmacodynamics and pharmacokinetics of the thrombin

receptor antagonist vorapaxar between healthy Japanese and

Caucasian subjects. Eur J Clin Pharmacol. 2012;68(3):291–300.

22. Statkevich P, Kosoglou T, Kumar B, et al. Pharmacokinetics of

vorapaxar and its metabolite sch 2046273 (M20) following oral

administration in healthy chinese and us subjects. Clin Pharmacol

Ther. 2012;91:S54.

23. Statkevich P, Kosoglou T, Preston RA, et al. Pharmacokinetics of

the novel PAR-1 antagonist vorapaxar in patients with hepatic

impairment. Eur J Clin Pharmacol. 2012;68(11):1501–8.

24. Kosoglou T, Kraft WK, Kumar B, et al. Pharmacokinetics and

pharmacodynamics of the novel PAR-1 antagonist vorapaxar in

patients with end-stage renal disease. Eur J Clin Pharmacol.

2012;68(7):1049–56.

25. Kosoglou T, Statkevich P, Kumar B, et al. The effect of multiple

doses of ketoconazole or rifampin on the single- and multiple-

dose pharmacokinetics of vorapaxar. J Clin Pharmacol. 2013;

53(5):540–9.

26. Kosoglou T, Zhu Y, Xuan F, et al. Vorapaxar, an oral PAR-1

receptor antagonist, does not affect the pharmacokinetics and

pharmacodynamics of warfarin. Eur J Clin Pharmacol. 2012;

68(11):1509–16.

27. Kosoglou T, Zhu Y, Statkevich P, et al. The influence of multi-

ple-dose vorapaxar, an oral PAR-1 receptor antagonist, on the

single-dose pharmacokinetics and pharmacodynamics of digoxin.

Clin Pharmacol Drug Dev. 2013;2(1):90–8.

28. Bohula May EA, Bonaca MP, Scirica BM, et al. Rates of arterial

revascularization in patients treated with vorapaxar vs. Placebo in

the TRA 2degreeP-TIMI 50 trial. Circulation. 2012;126(21

SUPPL. 1):A19221.

29. Bonaca MP, Scirica BM, Braunwald E, et al. Efficacy of vora-

paxar is not modified by thienopyridine therapy: Results from

TRA 2degreeP-TIMI 50 trial. Circulation. 2012;126(21 SUPPL.

1):A18595.

30. Scirica BM, Bonaca MP, Braunwald E, et al. Vorapaxar for

secondary prevention of thrombotic events for patients with

previous myocardial infarction: a prespecified subgroup analysis

of the TRA 2degreeP-TIMI 50 trial. Lancet. 2012;380(9850):

1317–24.

31. Scirica BM, Bonaca MP, Braunwal E, et al. Vorapaxar for sec-

ondary prevention after myocardial infarction according to aspirin

dose-insights from the TRA2degreep-timi 50 trial. Circulation.

2012;126(21 SUPPL. 1):A14508.

32. Cavender MA, Scirica B, Bonaca MP, et al. Vorapaxar in patients

with diabetes and prior MI: Findings from the TRA 2p-TIMI 50

trial. [abstract no. 10398]. In: American Heart Association Sci-

entific Sessions; 16–20 Nov 2013; Dallas, TX.

33. Morrow DA, Alberts MJ, Mohr JP, et al. Efficacy and safety of

vorapaxar in patients with prior ischemic stroke. Stroke.

2013;44(3):691–8.

34. Bonaca MP, Scirica BM, Creager MA, et al. Vorapaxar in

patients with peripheral artery disease: results from TRA2{de-

grees}P-TIMI 50. Circulation. 2013;127(14):1522–9, 9e1–6.

35. Bonaca MP, Scirica BM, Braunwald E, et al. Vorapaxar reduces

coronary stent thrombosis: Results from the TRA2P-TIMI 50

trial. Circulation. 2013;128(22 SUPPL. 1):A18928.

36. Bonaca MP, Scirica BM, Braunwald E, et al. New ischemic stroke

and outcomes with vorapaxar vs. Placebo: Results from TRA

2degreeP-TIMI 50 trial. Circulation. 2012;126(21 SUPPL. 1):

A19144.

37. Tricoci P, Huang Z, Held C, et al. Thrombin-receptor antagonist

vorapaxar in acute coronary syndromes. N Engl J Med. 2012;

366(1):20–33.

38. Armaganijan L, Lopes R, Huang Z, et al. Efficacy and safety of

vorapaxar in elderly patients with non-st-segment elevation acute

coronary syndrome: Insights from the tracer trial. Circulation Con-

ference: American Heart Association. 2013;128(22 SUPPL. 1).

39. Jones S, Tricoci P, Huang Z, et al. Vorapaxar in non-ST-segment

elevation acute coronary syndrome patients with peripheral artery

disease: Results from tracer. [abstract no. 17939]. In: American

Heart Association Scientific Sessions; 16–20 Nov 2013; Dallas, TX.

40. Mahaffey KW, Huang Z, Wallentin L, et al. Association of

aspirin dose and vorapaxar safety and efficacy in patients with

non-ST-segment elevation acute coronary syndrome (from the

TRACER Trial). Am J Cardiol. 2014;113(6):936–44.

41. Cornel JH, Tricoci P, Horton J, et al. Effects of glycoprotein IIB/

IIIA inhibitors in combination with vorapaxar, a platelet throm-

bin-receptor antagonist, among patients with non-St-segment

elevation acute coronary syndromes: Insights from the tracer trial.

J Am Coll Cardiol. 2013;(1):E102.

42. Tricoci P, Chen E, Neely ML, et al. CYP2C19 polymorphism and

pon-1 activity in NSTE acs: Vorapaxar effect in relation to

clopidogrel metabolism in the tracer trial. [abstract no. 17658].

1162 R. M. Poole, S. Elkinson

Page 11: Vorapaxar: First Global Approval

In: American Heart Association Scientific Sessions; 16-20 Nov

2013; Dallas, TX.

43. Leonardi S, Tricoci P, White HD, et al. Effect of vorapaxar on

myocardial infarction in the thrombin receptor antagonist for

clinical event reduction in acute coronary syndrome (TRACER)

trial. Eur Heart J. 2013;34(23):1723–31.

44. Tricoci P, Huang Z, Van De Werf F, et al. Net clinical benefit of

vorapaxar in NSTE ACS: Role of ischemic and bleeding risk

stratification. Circulation. 2012;126(21 SUPPL. 1):A19049.

45. Held C, Tricoci P, Huang Z, et al. Vorapaxar, a platelet thrombin-

receptor antagonist, in medically managed patients with non-st-

segment elevation acute coronary syndrome: Results from the

TRACER Trial. Circulation. 2012;126(21 SUPPL. 1):A9964.

46. Valgimigli M, Tricoci P, Huang Z, et al. Vorapaxar, a platelet

thrombin-receptor antagonist, in patients with non-st-segment

elevation acute coronary syndrome undergoing percutaneous

coronary intervention: Results from the tracer trial. Circulation.

2012;126(21 SUPPL. 1):A18805.

47. Whellan DJ, Tricoci P, Chen E, et al. Vorapaxar in acute coro-

nary syndrome patients undergoing coronary artery bypass graft

surgery: subgroup analysis from the TRACER trial (Thrombin

Receptor Antagonist for Clinical Event Reduction in Acute

Coronary Syndrome). J Am Coll Cardiol. 2014;63(11):1048–57.

48. Becker RC, Moliterno DJ, Jennings LK, et al. Safety and toler-

ability of SCH 530348 in patients undergoing non-urgent per-

cutaneous coronary intervention: a randomised, double-blind,

placebo- controlled phase II study. Lancet. 2009;373:919–28.

49. Goto S, Yamaguchi T, Ikeda Y, et al. Safety and exploratory

efficacy of the novel thrombin receptor (PAR-1) antagonist

SCH530348 for non-ST-segment elevation acute coronary syn-

drome. J Atheroscler Thromb. 2010;17(2):156–64.

50. Tricoci P, Huang Z, White HD, et al. Clinical characteristics

associated with major bleeding in NSTE ACS and the relation-

ship between bleeding risk, ischemic events, and the vorapaxar

effect: analysis from the TRACER trial. Eur Heart J. 2012;

33:322.

Vorapaxar: First Global Approval 1163


Recommended