CLINICAL PHARMACOLOGY
Mechanism Of Action
Ticagrelor and its major
metabolite reversibly interact with the platelet P2Y12 ADP-receptor to prevent
signal transduction and platelet activation. Ticagrelor and its active
metabolite are approximately equipotent.
Pharmacodynamics
The inhibition of platelet
aggregation (IPA) by ticagrelor and clopidogrel was compared in a 6-week study
examining both acute and chronic platelet inhibition effects in response to 20
μM ADP as the platelet aggregation agonist.
The onset of IPA was evaluated
on Day 1 of the study following loading doses of 180 mg ticagrelor or 600 mg
clopidogrel. As shown in Figure 3, IPA was higher in the ticagrelor group at
all time points. The maximum IPA effect of ticagrelor was reached at around 2
hours, and was maintained for at least 8 hours.
The offset of IPA was examined
after 6 weeks on ticagrelor 90 mg twice daily or clopidogrel 75 mg daily, again
in response to 20 μM ADP.
As shown in Figure 4, mean
maximum IPA following the last dose of ticagrelor was 88% and 62% for
clopidogrel. The insert in Figure 4 shows that after 24 hours, IPA in the
ticagrelor group (58%) was similar to IPA in clopidogrel group (52%),
indicating that patients who miss a dose of ticagrelor would still maintain IPA
similar to the trough IPA of patients treated with clopidogrel. After 5
days, IPA in the ticagrelor group was similar to IPA in the placebo group. It
is not known how either bleeding risk or thrombotic risk track with IPA, for
either ticagrelor or clopidogrel.
Figure 3 : Mean inhibition of platelet aggregation
(±SE) following single oral doses of placebo, 180 mg ticagrelor or 600 mg
clopidogrel
Figure 4 : Mean inhibition
of platelet aggregation (IPA) following 6 weeks on placebo, ticagrelor 90 mg
twice daily, or clopidogrel 75 mg daily
Transitioning from clopidogrel
to BRILINTA resulted in an absolute IPA increase of 26.4% and from BRILINTA to
clopidogrel resulted in an absolute IPA decrease of 24.5%. Patients can be
transitioned from clopidogrel to BRILINTA without interruption of antiplatelet
effect [see DOSAGE AND ADMINISTRATION].
Pharmacokinetics
Ticagrelor demonstrates dose proportional
pharmacokinetics, which are similar in patients and healthy volunteers.
Absorption
BRILINTA can be taken with or without food. Absorption of
ticagrelor occurs with a median tmax of 1.5 h (range 1.0– 4.0). The formation
of the major circulating metabolite AR-C124910XX (active) from ticagrelor
occurs with a median tmax of 2.5 h (range 1.5-5.0).
The mean absolute bioavailability of ticagrelor is about
36% (range 30%-42%). Ingestion of a high-fat meal had no effect on ticagrelor Cmax,
but resulted in a 21% increase in AUC. The Cmax of its major metabolite was
decreased by 22% with no change in AUC.
BRILINTA as crushed tablets mixed in water, given orally
or administered through a nasogastric tube into the stomach, is bioequivalent
to whole tablets (AUC and Cmax within 80-125% for ticagrelor and AR-C124910XX)
with a median tmax of 1.0 hour (range 1.0 – 4.0) for ticagrelor and 2.0 hours
(range 1.0 –8.0) for AR-C124910XX.
Distribution
The steady state volume of distribution of ticagrelor is
88 L. Ticagrelor and the active metabolite are extensively bound to human
plasma proteins (>99%).
Metabolism
CYP3A4 is the major enzyme responsible for ticagrelor
metabolism and the formation of its major active metabolite. Ticagrelor and its
major active metabolite are weak P-glycoprotein substrates and inhibitors. The
systemic exposure to the active metabolite is approximately 30-40% of the
exposure of ticagrelor.
Excretion
The primary route of ticagrelor elimination is hepatic
metabolism. When radiolabeled ticagrelor is administered, the mean recovery of
radioactivity is approximately 84% (58% in feces, 26% in urine). Recoveries of
ticagrelor and the active metabolite in urine were both less than 1% of the
dose. The primary route of elimination for the major metabolite of ticagrelor
is most likely to be biliary secretion. The mean t½ is approximately 7 hours
for ticagrelor and 9 hours for the active metabolite.
Specific Populations
The effects of age, gender, ethnicity, renal impairment
and mild hepatic impairment on the pharmacokinetics of ticagrelor are presented
in Figure 5. Effects are modest and do not require dose adjustment.
Figure 5 :Impact of intrinsic factors on the
pharmacokinetics of ticagrelor
Effects Of Other Drugs On BRILINTA
CYP3A4 is the major enzyme
responsible for ticagrelor metabolism and the formation of its major active
metabolite. The effects of other drugs on the pharmacokinetics of ticagrelor
are presented in Figure 6 as change relative to ticagrelor given alone
(test/reference). Strong CYP3A inhibitors (e.g., ketoconazole, itraconazole,
and clarithromycin) substantially increase ticagrelor exposure. Moderate CYP3A
inhibitors have lesser effects (e.g., diltiazem). CYP3A inducers (e.g.,
rifampin) substantially reduce ticagrelor blood levels. P-gp inhibitors (e.g.,
cyclosporine) increase ticagrelor exposure.
Co-administration of 5 mg
intravenous morphine with 180 mg loading dose of ticagrelor decreased observed
mean ticagrelor exposure by up to 25% in healthy adults and up to 36% in ACS
patients undergoing PCI. Tmax was delayed by 1-2 hours. Exposure of the active
metabolite decreased to a similar extent. Morphine co-administration did not
delay or decrease platelet inhibition in healthy adults. Mean platelet
aggregation was higher up to 3 hours post loading dose in ACS patients
co-administered with morphine.
Co-administration of
intravenous fentanyl with 180 mg loading dose of ticagrelor in ACS patients
undergoing PCI resulted in similar effects on ticagrelor exposure and platelet
inhibition.
Figure 6 : Effect of
co-administered drugs on the pharmacokinetics of ticagrelor
Effects Of BRILINTA On Other
Drugs
In vitro metabolism studies demonstrate that ticagrelor and its
major active metabolite are weak inhibitors of CYP3A4, potential activators of
CYP3A5 and inhibitors of the P-gp transporter. Ticagrelor and AR-C124910XX were
shown to have no inhibitory effect on human CYP1A2, CYP2C19, and CYP2E1
activity. For specific in vivo effects on the pharmacokinetics of
simvastatin, atorvastatin, ethinyl estradiol, levonorgesterol, tolbutamide,
digoxin and cyclosporine, see Figure 7.
Figure 7 : Impact of BRILINTA on the pharmacokinetics
of co-administered drugs
Pharmacogenetics
In a genetic substudy cohort of
PLATO, the rate of thrombotic CV events in the BRILINTA arm did not depend on
CYP2C19 loss of function status.
Clinical Studies
Acute Coronary Syndromes And Secondary Prevention After Myocardial
Infarction
PLATO
PLATO was a randomized double-blind study comparing
BRILINTA (N=9333) to clopidogrel (N=9291), both given in combination with
aspirin and other standard therapy, in patients with acute coronary syndromes
(ACS), who presented within 24 hours of onset of the most recent episode of
chest pain or symptoms. The study’s primary endpoint was the composite of first
occurrence of cardiovascular death, non-fatal MI (excluding silent MI), or
non-fatal stroke.
Patients who had already been treated with clopidogrel
could be enrolled and randomized to either study treatment. Patients with
previous intracranial hemorrhage, gastrointestinal bleeding within the past 6
months, or with known bleeding diathesis or coagulation disorder were excluded.
Patients taking anticoagulants were excluded from participating and patients
who developed an indication for anticoagulation during the trial were
discontinued from study drug. Patients could be included whether there was
intent to manage the ACS medically or invasively, but patient randomization was
not stratified by this intent.
All patients randomized to BRILINTA received a loading
dose of 180 mg followed by a maintenance dose of 90 mg twice daily. Patients in
the clopidogrel arm were treated with an initial loading dose of clopidogrel
300 mg, if clopidogrel therapy had not already been given. Patients undergoing
PCI could receive an additional 300 mg of clopidogrel at investigator
discretion. A daily maintenance dose of aspirin 75-100 mg was recommended, but
higher maintenance doses of aspirin were allowed according to local judgment.
Patients were treated for at least 6 months and for up to 12 months.
PLATO patients were predominantly male (72%) and
Caucasian (92%). About 43% of patients were >65 years and 15% were >75
years. Median exposure to study drug was 277 days. About half of the patients
received pre-study clopidogrel and about 99% of the patients received aspirin
at some time during PLATO. About 35% of patients were receiving a statin at
baseline and 93% received a statin sometime during PLATO.
Table 6 shows the study results for the primary composite
endpoint and the contribution of each component to the primary endpoint.
Separate secondary endpoint analyses are shown for the overall occurrence of CV
death, MI, and stroke and overall mortality.
Table 6 : Patients with outcome events (KM%)(PLATO)
|
BRILINTA1
N=9333 |
Clopidogrel
N=9291 |
Hazard Ratio (95% CI) |
p-value |
Composite of CV death, MI, or stroke |
9.8 |
11.7 |
0.84
(0.77, 0.92) |
0.0003 |
CV death |
2.9 |
4.0 |
0.74 |
|
Non-fatal MI |
5.8 |
6.9 |
0.84 |
|
Non-fatal stroke |
1.4 |
1.1 |
1.24 |
|
Secondary endpoints2 |
CV death |
4.0 |
5.1 |
0.79
(0.69, 0.91) |
0.0013 |
MI3 |
5.8 |
6.9 |
0.84
(0.75, 0.95) |
0.0045 |
Stroke3 |
1.5 |
1.3 |
1.17
(0.91, 1.52) |
0.22 |
All-cause mortality |
4.5 |
5.9 |
0.78
(0.69, 0.89) |
0.0003 |
1 Dosed at 90 mg bid.
2 Note: rates of first events for the components CV Death, MI
and Stroke are the actual rates for first events for each component and do not
add up to the overall rate of events in the composite endpoint.
3 Including patients who could have had other non-fatal
events or died. |
The Kaplan-Meier curve (Figure
8) shows time to first occurrence of the primary composite endpoint of CV
death, nonfatal MI or non-fatal stroke in the overall study.
Figure 8 : Time to first
occurrence of CV death, MI, or stroke (PLATO)
The curves separate by 30 days
[relative risk reduction (RRR) 12%] and continue to diverge throughout the
12-month treatment period (RRR 16%).
Among 11289 patients with PCI
receiving any stent during PLATO, there was a lower risk of stent thrombosis
(1.3% for adjudicated “definite”) than with clopidogrel (1.9%) (HR 0.67, 95% CI
0.50-0.91; p=0.009). The results were similar for drug-eluting and bare metal
stents.
A wide range of demographic,
concurrent baseline medications, and other treatment differences were examined
for their influence on outcome. Some of these are shown in Figure 9. Such
analyses must be interpreted cautiously, as differences can reflect the play of
chance among a large number of analyses. Most of the analyses show effects
consistent with the overall results, but there are two exceptions: a finding of
heterogeneity by region and a strong influence of the maintenance dose of
aspirin. These are considered further below.
Most of the characteristics
shown are baseline characteristics, but some reflect post-randomization
determinations (e.g., aspirin maintenance dose, use of PCI).
Figure 9: Subgroup analyses of (PLATO)
Note: The figure above presents
effects in various subgroups most of which are baseline characteristics and
most of which were pre-specified. The 95% confidence limits that are shown do
not take into account how many comparisons were made, nor do they reflect the
effect of a particular factor after adjustment for all other factors. Apparent
homogeneity or heterogeneity among groups should not be over-interpreted.
Regional Differences
Results in the rest of the
world compared to effects in North America (US and Canada) show a smaller effect
in North America, numerically inferior to the control and driven by the US
subset. The statistical test for the US/non-US comparison is statistically
significant (p=0.009), and the same trend is present for both CV death and
non-fatal MI. The individual results and nominal p-values, like all subset
analyses, need cautious interpretation, and they could represent chance
findings. The consistency of the differences in both the CV mortality and
non-fatal MI components, however, supports the possibility that the finding is
reliable.
A wide variety of baseline and procedural differences
between the US and non-US (including intended invasive vs. planned medical
management, use of GPIIb/IIIa inhibitors, use of drug eluting vs. bare-metal
stents) were examined to see if they could account for regional differences,
but with one exception, aspirin maintenance dose, these differences did not
appear to lead to differences in outcome.
Aspirin Dose
The PLATO protocol left the choice of aspirin maintenance
dose up to the investigator and use patterns were different in US sites from
sites outside of the US. About 8% of non-US investigators administered aspirin
doses above 100 mg, and about 2% administered doses above 300 mg. In the US,
57% of patients received doses above 100 mg and 54% received doses above 300
mg. Overall results favored BRILINTA when used with low maintenance doses
(≤100 mg) of aspirin, and results analyzed by aspirin dose were similar
in the US and elsewhere. Figure 10 shows overall results by median aspirin
dose. Figure 10 shows results by region and dose.
Figure 10 : CV death, MI, stroke by maintenance
aspirin dose in the US and outside the US (PLATO)
Like any unplanned subset
analysis, especially one where the characteristic is not a true baseline
characteristic (but may be determined by usual investigator practice), the
above analyses must be treated with caution. It is notable, however, that
aspirin dose predicts outcome in both regions with a similar pattern, and that
the pattern is similar for the two major components of the primary endpoint, CV
death and non-fatal MI.
Despite the need to treat such
results cautiously, there appears to be good reason to restrict aspirin
maintenance dosage accompanying ticagrelor to 100 mg. Higher doses do not have
an established benefit in the ACS setting, and there is a strong suggestion
that use of such doses reduces the effectiveness of BRILINTA.
PEGASUS
The PEGASUS TIMI-54 study was a
21162-patient, randomized, double-blind, placebo-controlled, parallel-group
study. Two doses of ticagrelor, either 90 mg twice daily or 60 mg twice daily,
co-administered with 75-150 mg of aspirin, were compared to aspirin therapy
alone in patients with history of MI. The primary endpoint was the composite of
first occurrence of CV death, non-fatal MI and non-fatal stroke. CV death and
all-cause mortality were assessed as secondary endpoints.
Patients were eligible to
participate if they were ≥50 years old, with a history of MI 1 to 3 years
prior to randomization, and had at least one of the following risk factors for
thrombotic cardiovascular events: age ≥65 years, diabetes mellitus requiring
medication, at least one other prior MI, evidence of multivessel coronary
artery disease, or creatinine clearance <60 mL/min. Patients could be
randomized regardless of their prior ADP receptor blocker therapy or a lapse in
therapy. Patients requiring or who were expected to require renal dialysis
during the study were excluded. Patients with any previous intracranial
hemorrhage, gastrointestinal bleeding within the past 6 months, or with known
bleeding diathesis or coagulation disorder were excluded. Patients taking
anticoagulants were excluded from participating and patients who developed an
indication for anticoagulation during the trial were discontinued from study
drug. A small number of patients with a history of stroke were included. Based
on information external to PEGASUS, 102 patients with a history of stroke (90
of whom received study drug) were terminated early and no further such patients
were enrolled.
Patients were treated for at least 12 months and up to 48
months with a median follow up time of 33 months.
Patients were predominantly male (76%) Caucasian (87%)
with a mean age of 65 years, and 99.8% of patients received prior aspirin
therapy. See Table 7 for key baseline features.
Table 7 : Baseline features (PEGASUS)
Demographic |
% Patients |
<65 years |
45% |
Diabetes |
32% |
Multivessel disease |
59% |
History of >1 MI |
17% |
Chronic non-end stage renal disease |
19% |
Stent |
80% |
Prior P2Y12 platelet inhibitor therapy |
89% |
Lipid lowering therapy |
94% |
The Kaplan-Meier curve (Figure
11) shows time to first occurrence of the primary composite endpoint of CV
death, nonfatal MI or non-fatal stroke.
Figure 11 :Time to First Occurrence of CV death, MI or
Stroke (PEGASUS)
Both the 60 mg and 90 mg
regimens of BRILINTA in combination with aspirin were superior to aspirin alone
in reducing the incidence of CV death, MI or stroke. The absolute risk
reductions for BRILINTA plus aspirin vs. aspirin alone were 1.27% and 1.19% for
the 60 and 90 mg regimens, respectively. Although the efficacy profiles of the
two regimens were similar, the lower dose had lower risks of bleeding and
dyspnea.
Table 8 shows the results for
the 60 mg plus aspirin regimen vs. aspirin alone.
Table 8 : Incidences of the
primary composite endpoint, primary composite endpoint components, and
secondary endpoints (PEGASUS)
|
BRILINTA1 + Aspirin
N=7045 |
Aspirin Alone
N=7067 |
HR (95% CI) |
p-value |
n (patients with event) |
KM% |
n (patients with event) |
KM% |
Time to first CV death, MI, or stroke2 |
487 |
7.8 |
578 |
9.0 |
0.84
(0.74, 0.95) |
0.0043 |
CV Death4 |
116 |
|
128 |
|
|
|
Myocardial infarction4 |
283 |
|
336 |
|
|
|
Stroke4 |
88 |
|
114 |
|
|
|
Subjects with events at any time CV Death3, 5 |
174 |
2.9 |
210 |
3.4 |
0.83
(0.68, 1.01) |
|
Myocardial infarction5 |
285 |
4.5 |
338 |
5.2 |
0.84
(0.72, 0.98) |
|
Stroke5 |
91 |
1.5 |
122 |
1.9 |
0.75
(0.57, 0.98) |
|
All-cause mortality3 |
289 |
4.7 |
326 |
5.2 |
0.89
(0.76, 1.04) |
|
1 60 mg BID
2 Primary endpoint
3 Secondary endpoints
4 For the components, the first-occurring component of the composite
is included.
5 The number of first events for the components CV Death, MI and
Stroke are the actual number of first events for each component and do not add
up to the number of events in the composite endpoint. |
In PEGASUS, the RRR for the
composite endpoint from 1 to 360 days (17% RRR) and from 361 days and onwards
(16% RRR) were similar.
The treatment effect of
BRILINTA 60 mg over aspirin appeared similar across most pre-defined subgroups,
see Figure 12.
Figure 12 : Subgroup
analyses of ticagrelor 60 mg (PEGASUS)
Note: The figure above presents
effects in various subgroups all of which are baseline characteristics and most
of which were pre-specified. The 95% confidence limits that are shown do not
take into account how many comparisons were made, nor do they reflect the
effect of a particular factor after adjustment for all other factors. Apparent
homogeneity or heterogeneity among groups should not be over-interpreted.