Vous êtes sur la page 1sur 7

Journal of Thrombosis and Haemostasis, 7: 559–565 DOI: 10.1111/j.1538-7836.2009.03298.

ORIGINAL ARTICLE

The pharmacokinetics of idraparinux, a long-acting indirect


factor Xa inhibitor: population pharmacokinetic analysis from
Phase III clinical trials
C . V E Y R A T - F O L L E T , * N . V I V I E R , * M . T R E L L U ,   C . D U B R U C   and G . J . S A N D E R I N K  
*sanofi-aventis, Vitry sur Seine, Cedex; and  sanofi-aventis, Chilly-Mazarin, France

To cite this article: Veyrat-Follet C, Vivier N, Trellu M, Dubruc C, Sanderink GJ. The pharmacokinetics of idraparinux, a long-acting indirect factor
Xa inhibitor: population pharmacokinetic analysis from Phase III clinical trials. J Thromb Haemost 2009; 7: 559–65.

Keywords: atrial fibrillation, idraparinux, population pharma-


Summary. Background: Idraparinux, a long-acting synthetic cokinetics, stroke, venous thromboembolism.
pentasaccharide, is a specific antithrombin-dependent inhibitor
of activated factor X that has been investigated in the treatment
and prevention of thromboembolic events. Objectives: To Introduction
characterize the population pharmacokinetic profile of idra- The prevention of venous thromboembolism (VTE) recurrence
parinux in patients enrolled in van Gogh and Amadeus Phase following initial treatment of deep-vein thrombosis (DVT) or
III clinical trials. Patients and methods: Idraparinux was pulmonary embolism (PE), and of thromboembolic events
administered once-weekly subcutaneously at a dose of 2.5 mg, such as stroke in patients with atrial fibrillation (AF), usually
or 2.5 mg (first dose) and then 1.5 mg for patients with severe involves the long-term use of vitamin K antagonists (VKAs),
renal insufficiency (creatinine clearance <30 mL min)1). A which is often problematic and inconvenient [1]. Idraparinux, a
population pharmacokinetic model was developed using data long-acting synthetic pentasaccharide, is a selective and potent
from 704 patients with acute deep-vein thrombosis or pulmo- antithrombin-dependent inhibitor of activated factor X (FXa),
nary embolism, 1310 patients suffering from atrial fibrillation, with a very high affinity for antithrombin (Kd = 1.4 ±
and 40 healthy subjects. Potential covariates analyzed included 0.3 nmol L)1) [2]. After subcutaneous (s.c.) administration
demographics (age, sex, weight and ethnicity), and serum idraparinux is rapidly absorbed (tmax ranged between 2 and
creatinine and creatinine clearance determinations. Results: A 4 h), with its bioavailability approaching 100% [2,3]. No
three-compartment model best described idraparinux pharma- evidence of metabolism of the parent compound was observed
cokinetics, with interindividual variability on clearance, central in humans after a single intravenous administration of [14C]-
volume of distribution, and absorption rate constant; residual idraparinux. Urine was the main excretion route of the
variability was low. Typical clearance, central volume of compound and all the radioactivity excreted in urine repre-
distribution, absorption rate constant and volume of distribu- sented the unchanged compound. Due to its long half-life,
tion at steady-state were 0.0255 L h)1, 3.36 L, 1.37 h and idraparinux is suitable for s.c. once-weekly dosing. Moreover,
30.8 L, respectively. Peak concentration was reached at 2.5 h. the anticoagulant response produced by idraparinux is highly
The terminal half-life was 66.3 days and time to steady-state predictable, as assessed by its linear pharmacokinetics with
was 35 weeks. At steady-state, exposures were similar for dose-proportionality up to 14 mg [3], and low (<30%)
patients without and with severe renal impairment receiving variability in healthy subjects. Thus, idraparinux offers the
adjusted-dose. Creatinine clearance was the most important potential for improved convenience in terms of dosing and
covariate affecting idraparinux clearance. The particular char- monitoring, in the long-term anticoagulant prophylaxis and
acteristics of idraparinux – rapid onset of action and long-acting treatment of thromboembolic events.
anticoagulant effect – offer interesting clinical perspectives The Phase II dose ranging PERSIST study demonstrated
currently under investigation with idrabiotaparinux, the revers- that s.c. once-weekly administration of 2.5 mg idraparinux was
ible biotinylated form of idraparinux. as effective as warfarin for the secondary prevention of DVT,
and was not associated with major bleeding [4]. Based on these
Correspondence: Christine Veyrat-Follet, sanofi-aventis, Global findings, s.c. once-weekly administration of idraparinux 2.5 mg
Metabolism and Pharmacokinetics, 13, Quai Jules Guesdes B.P. 14, was investigated in Phase III clinical trials of patients with acute
94403 Vitry sur Seine, Cedex, France. symptomatic DVT or PE in the van Gogh trials [5,6] and with
Tel.: +33 1 58 93 85 71; fax: +33 1 58 93 85 67. AF in the Amadeus trial [7]. The objectives of the present
E-mail: christine.veyrat-follet@sanofi-aventis.com analysis were to characterize the population pharmacokinetic
profile of idraparinux in patients who participated in the Phase
Received 18 November 2008, accepted 12 January 2009

 2009 International Society on Thrombosis and Haemostasis


560 C. Veyrat-Follet et al

III trials, and to identify covariates that affected the pharma- ing, myocardial infarction, or VTE event. In the Phase I studies
cokinetics of idraparinux. rich sampling was performed over the 0–648 h time interval.
Blood samples of 5 mL were collected in tubes containing
trisodium citrate and were centrifuged at 3000 ·g for 15 min at
Methods
4 C. Plasma was divided into two aliquots and stored at
)20 C until analyzed.
Trial protocols
The van Gogh trials were multicenter, international, random-
Study assessments
ized, open-label, assessor-blind, non-inferiority studies, which
compared the efficacy and safety of idraparinux with the Idraparinux assay Idraparinux concentrations in plasma
combination of heparin and VKA in the treatment of either were measured by a fully validated biological assay that used
acute symptomatic DVT or PE [5]. Patients received either s.c. the antithrombin-mediated FXa inhibitory activity of
once-weekly idraparinux 2.5 mg, or heparin (low-molecular- idraparinux [8; Data on file]. Idraparinux concentrations in
weight heparin or unfractionated heparin) followed by plasma were determined by a chromogenic assay using the
adjusted-dose VKA for 3 or 6 months. The van Gogh Stachrom Heparin kit (Diagnostica Stago, Inc., Parsippany,
extension trial compared the efficacy and safety of s.c. once- NJ, USA) in an excess of exogenous antithrombin. The
weekly idraparinux with placebo in the long-term prevention of determination of the concentration in a sample in gravimetric
symptomatic VTE in patients with symptomatic PE or DVT units is achieved through comparison of measured anti-FXa in
who completed 6 months of treatment with VKA or idrapar- the sample with a calibration curve spiked with known
inux [6]. Patients were treated for an additional 6 months amounts of idraparinux. Plasma concentrations are expressed
(26 weeks) with once-weekly s.c. idraparinux 2.5 mg or as free acid. Quantification of idraparinux in plasma was
placebo. unaffected by the presence of EDTA as a blood anticoagulant,
The Amadeus trial was a multicenter, multinational, ran- the presence of hemolyzed blood in plasma (3%), or by the
domized, open-label, assessor-blind, non-inferiority study presence of acetaminophen, aspirin, ibuprofen, caffeine,
comparing the efficacy and safety of s.c. once-weekly idrapar- nicotine, ketoconazole, digoxin, racemic warfarin, or
inux 2.5 mg with adjusted-dose VKA in the prevention of levonorgestrel. The limit of quantification was 71.4 ng mL)1;
thromboembolic events in patients with AF; patients were total precision and accuracy were better than 5.47% and
treated for 6 months to 2 years [7]. Patients with severe renal 7.11%, respectively.
insufficiency, defined as creatinine clearance (CrCl)
<30 mL min)1, received 1.5 mg idraparinux once-weekly Clinical covariates Potential clinical covariates in this
following an initial dose of 2.5 mg, or 1.5 mg for patients in pharmacokinetic analysis were subject demographics (age, age
the van Gogh extension trial who had previously received index, i.e. <75 years or ‡75 years, sex, weight, and ethnicity),
idraparinux. and laboratory determinations (serum creatinine and CrCl
Data from healthy subjects in two Phase I trials who received computed from serum creatinine concentration, weight, age,
s.c. once-weekly single-dose idraparinux 2.5 mg were included and sex according to the Cockcroft and Gault formula [9]).
in this analysis in order to better characterize the structural
model. One study was a single-center, single ascending dose,
Pharmacokinetic analysis
double-blind, randomized, placebo-controlled, sequential
group study to assess the tolerability, safety and pharmacoki- The population pharmacokinetic analysis was performed in
netics of idraparinux in young healthy males, and elderly three steps using non-linear mixed-effect modeling (version V
healthy male and female subjects. The second study was a NONMEM, GloboMax, Hanover, MD, USA). A basic
multicenter, single dose, open-label, parallel group, pharma- structural pharmacokinetic model was obtained based on
codynamic study. idraparinux concentration data from the Phase III and Phase I
clinical trials detailed above [Data on file]. The main pharma-
cokinetic parameters determined were: clearance, which indi-
Blood sampling schedules
cates the rate of elimination from plasma; distribution volumes;
In the van Gogh DVT and PE trials, samples were collected at and elimination half-lives. Then an initial exploratory analysis
2–4 h after the first idraparinux injection, just prior to the next for covariate effects on individual pharmacokinetic parameters
idraparinux injection and at the end of week 13, plus at the end was performed using SAS software (version 8.2 SAS Institute
of week 26 for those in the 6-month stratum. No schedule was Inc, Cary, NC, USA). Univariate analysis was also used.
prespecified in the van Gogh extension trial. In addition, in all The relationship between pharmacokinetic parameters and
the van Gogh trials, blood samples were collected in case of covariates was investigated based on nonlinear mixed-effect
safety or efficacy events. In the Amadeus trial, blood samples modeling using the likelihood ratio test to discriminate among
were collected at 3, 12 and 18 months, and at the end of alternative models. Model building was accomplished by a
treatment, with additional collections in case of a suspected backward elimination procedure [10]. Finally, the model was
stroke, non-central nervous system systemic embolism, bleed- evaluated using visual predictive check.

 2009 International Society on Thrombosis and Haemostasis


Population pharmacokinetics of idraparinux 561

Results 2000 Van Gogh


1500
Demographics
1000

Concentration (ng mL–1)


In all, idraparinux plasma concentration data from 2027 500
subjects evaluable for PK analysis were included, comprising
0
1987 patients from the Phase III trials in which the model was 0 12 24 36 48 60 72 84 96 108
2000
evaluated, and 40 healthy subjects from the Phase I studies. The Amadeus
demographics of these patients are shown in Table 1. Overall, 1500
the majority of individuals included in this analysis were male, 1000
weighed between 50 kg and <100 kg (81%), were aged 500
<75 years, and were Caucasian. Most individuals had either
0
normal or mildly impaired renal function (83% overall), 15% 0 12 24 36 48 60 72 84 96 108
had moderate and 2% severe renal impairment. The percent- Time (week)
ages of patients aged ‡75 years and average patient weight Fig. 1. Idraparinux concentration time profiles in the van Gogh and
were higher in the Amadeus trial than in the van Gogh trials. Amadeus Phase III clinical trials.

on residual error (16.0% vs. 20%) compared with the two-


Population pharmacokinetic model building
compartment model. A more complex residual error model,
Basic model development was performed using a data set involving both a proportional and an additive component,
including 2027 subjects, corresponding to 4112 concentrations, significantly improved the fit, with a drop of 96 points of the
of which 88.2% were from the Phase III trials. The idraparinux objective function. Consequently, the three-compartment
time-concentration data of the van Gogh and Amadeus trials model, with interindividual variability on clearance, central
populations are shown in Fig. 1. volume of distribution (V2), and absorption rate constant with
An average of 2.6 and 1.35 blood samples per patient were mixed-proportional and additive-residual errors, was retained
obtained in the Phase III van Gogh and Amadeus trials, as the best basic model. The parameters were well estimated
respectively, and 12 blood samples per subject were obtained in and the observed data were well described by the model as
the Phase I trials. Initially, a two-compartment model was illustrated in Fig. 2. Furthermore, the predictive check showed
tested with idraparinux time-concentration data. However, a that the developed model was able to simulate data in close
three-compartment model provided the best fit to the data with agreement with the actual studiesÕ observations.
a decreased objective function (‡821 point drop), and a lower In the final model, peak concentration was rapidly achieved
interindividual variability on clearance (20.4% vs. 25.7%) and at 2.5 h, typical clearance was 0.0255 L h)1, V2 was 3.36 L,

Table 1 Characteristics of the patient study population


Number of patients (%) Mean ± SD Median (5–95%)

van Gogh Amadeus


(N = 692) (N = 1295) van Gogh Amadeus van Gogh Amadeus

Patient demographics
Age, years 61 ± 17 70 ± 9 63 (28–83) 71 (54–83)
Age index
< 5 years 530 (76.6) 851 (65.7)
‡75 years 162 (23.4) 444 (34.3)
Weight, kg 81 ± 19 86 ± 18 80 (54–115) 84 (60–116)
Sex, Male 367 (53.0) 898 (69.3)
Ethnicity
Caucasian 672 (97.1) 1258 (97.1)
Black 10 (1.5) 9 (0.7)
Oriental 5 (0.7) 7 (0.5)
Other 5 (0.7) 21 (1.6)
Laboratory measurements
Creatinine clearance, mL min)1 88.6 ± 41.7 77.3 ± 30.6 82 (32–172) 73 (37–134)
Renal insufficiency
Severe (10 to <30 mL min)1) 30 (4.3) 13 (1.0)
Moderate (30 to <50 mL min)1) 89 (12.9) 217 (16.8)
Mild (50–80 mL min)1) 215 (31.1) 558 (43.1)
Normal (>80 mL min)1) 358 (51.7) 507 (39.2)

SD, standard deviation.

 2009 International Society on Thrombosis and Haemostasis


562 C. Veyrat-Follet et al

volume of distribution at steady state was 30.8 L, and index (whether <75 years or ‡75 years), whereas interindivid-
absorption rate constant was 1.37 h. The half-lives of the three ual variability on central volume was significantly related to
phases of the model were 6.51 h for the first half-life, 95.1 h for age, weight and sex. The remaining interindividual variabil-
the second half-life, and 66.3 days for the terminal half-life ity—not explained by covariate effects—was 18.6% on clear-
(Fig. 3). Interindividual variability of idraparinux clearance ance, 20.2% on central volume of distribution, and 129% on
was significantly related to subject weight, CrCl, sex, and age absorption rate constant. Residual variability was low, with a
coefficient of variation of 16.5% for the proportional part and
1800 34.2 ng mL)1 for the additive part, which was below the lower
limit of quantification of 71.4 ng mL)1.
Individual predictions (ng mL–1)

1400
Assessment of covariates effects

1000 The effects of covariates on idraparinux pharmacokinetic


parameters in the final model are shown in Table 2. The most
important covariate effect on idraparinux clearance was CrCl,
600 with a decrease of 20% in a typical subject with moderate renal
Unit line impairement (CrCl ‡30 to <50 mL min)1) and a decrease of
Smooth
43% in a subject with severe renal impairment (CrCl
200
<30 mL min)1). Idraparinux clearance decreases by 16% in
200 600 1000 1400 1800 female subjects and by 5.3% in those aged ‡75 years; ethnicity
Observed concentrations (ng mL–1) appeared to have no effect on clearance.
Fig. 2. Goodness-of-fit plot: individual predictions of concentration vs.
observed concentration over one dosing interval. Individual pharmacokinetic parameter estimates

1200 Overall, pharmacokinetic parameters values in subjects across


the various clinical studies were in the same range, and were
Concentration (ng mL–1)

1000 t 1 α = 6.51 h
2 comparable between the Phase III and Phase I studies
800 (Table 3). At week 1 and at steady state, similar pharmacoki-
t 1 β = 95.1 h
netic parameters were observed in patients across clinical
600 2 studies, both in patients without (idraparinux dose of 2.5 mg)
400 t
1
γ = 66.3 days
and with (first dose of 2.5 mg then adjusted dose of 1.5 mg)
2 severe renal insufficiency. Mean parameter values ± SD at
200
steady state for patients without and with severe renal
0 insufficiency, respectively, were: Cmax ss 1.19 ± 0.278 lg mL)1
1.00 4.00 7.00 10.00 13.00 16.00 and 1.09 ± 0.261 lg mL)1; Ctrough ss 0.493 ± 0.166 lg mL)1
Time (week) and 0.571 ± 0.166 lg mL)1; and AUCss 110 ± 30.3
Fig. 3. Idraparinux steady state concentration time profile in a typical lg*h mL)1 and 115 ± 30.1 lg*h mL)1. The mean accumu-
patient. t1/2, half-life. lation ratio (AUCss/AUCsweek1) was 2.9 ± 0.5 and 2.5 ± 0.4

Table 2 Assessment of covariate effects on idraparinux pharmacokinetic parameters according to the final model
5 and Clearance % Distribution %
Covariate 95% (CL), L h)1 Change* volume (V2) Change*

Mean patient  0.0255 3.36


Creatinine clearance 35.1 0.0195 )23.5 N/A
(CrCl), mL min)1 150 0.0322 26.4
Weight (WT), kg 57 0.0231 )9.41 2.75 )18.1
116 0.0279 9.67 4.05 20.5
Sex (SEX) Female 0.0214 )16 2.73 )18.7
Age index (IAGE) ‡75 years 0.0241 )5.3 N/A
Age (AGE), years 35 N/A 3.68 9.67
83 3.27 )2.48

N/A, not applicable.


*Theoretical effect (% change with respect to the mean) of the covariate considered alone, the other covariates being set to their median value.
 
Male, aged <75 years, weighing 82.3 kg, and with a CrCl of 76.2 mL min)1.
^ ¼ ðh1 ðWT =82:3Þ h11  ðCrCl=76:2Þ h12  hSEX  hLAGE Þ
CL 13 14
^ ¼ ðh2 ðAGE=69Þ h15  ðWT =82:3Þ h16  hSEX Þ
V2 17

 2009 International Society on Thrombosis and Haemostasis


Population pharmacokinetics of idraparinux 563

65.9 ± 13.0 (19.7)

Predicted concentration (ng mL–1)


0.0242 ± 0.00638
1200

97.2 ± 9.12 (9.4)


[0.0142–0.0347]
1000

[51.5–89.5]
[86.9–112]
800

(26.4)
Total

2027
600
Estimated pharmacokinetic
concentration equivalent to Ctrough
400 week 1, at week 36

64.1 ± 10.1 (15.7)


200
0.0245 ± 0.00616

96.2 ± 6.04 (6.3)


[0.0147–0.0347]
0
1 3 5 7 9 11 13 15 17 19 21 23 25 27 29 31 33 35 37 39 41 43 45

[84.1–52.0]
[87.5–106]
Amadeus

Time (week)
(25.2)
Table 3 Descriptive statistics of individual pharmacokinetic parameter estimates across studies – mean ± standard deviation (coefficient of variation %) [5–95%]

1295

Estimated Ctrough pharmacokinetic


concentration at week 1 (7 days after the
first administration): 109 ng mL–1

Fig. 4. Concentration-time course in a typical patient, and decrease of the


66.9 ± 10.4 (15.6)
0.0235 ± 0.00530

concentration to levels equal to the Ctrough at week 1.


97.5 ± 8.61 (8.8)
[0.0136–0.0331]

[54.3–86.4]
van Gogh

[86.6–115]
Extension

Predicted concentration (ng mL–1)


(22.6)

1200
24

1000

800
CL, clearance; DVT, deep-vein thrombosis; PE, pulmonary embolism; t1/2b, second half-life; t1/2c, terminal half-life.
99.3 ± 13.0 (13.1)

69.7 ± 16.8 (24.2)


van Gogh PE and

Estimated pharmacokinetic
0.0234 ± 0.00684

600 concentration equivalent to Ctrough


van Gogh DVT

[0.0132–0.0352]

week 1, at week 39
400
[102–50.7]
[85.4–124]

200
(29.2)
668

0
1 3 5 7 9 11 13 15 17 19 21 23 25 27 29 31 33 35 37 39 41 43 45
Time (week)
Estimated Ctrough pharmacokinetic
concentration at week 1 (7 days after the
98.5 ± 12.4 (12.6)

67.9 ± 16.5 (24.2)

–1
0.0241 ± 0.00674

first administration): 143 ng mL


van Gogh DVT

[0.0135–0.0352]

Fig. 5. Concentration-time course in a patient with severe renal impair-


[50.2–99.3]
[84.7–124]

ment, and decrease of the concentration to levels equal to the Ctrough at


week 1.
(28)
335

in patients without and with severe renal insufficiency,


respectively.
71.4 ± 17.1 (23.9)
100 ± 13.5 (13.4)
0.0227 ± 0.00688

Time to steady state was 35 weeks in a typical patient


[0.0129–0.0359]
van Gogh PE

(defined as one without renal insufficiency and aged


<75 years), which corresponds to approximately 8 months.
[85.4–125]

[51.5–105]
(30.3)

The estimated decrease of idraparinux concentration to levels


333

equal to the Ctrough at the end of week 1 occurs at week 36,


which is 12 weeks after cessation of treatment (Fig. 4). In
patients with severe renal insufficiency this corresponds to
15 weeks (Fig. 5). The dose adjustment performed in
60.4 ± 8.06 (13.3)
0.0283 ± 0.00304

96.6 ± 8.25 (8.5)

patients with severe renal insufficiency – at 1.5 mg after


[0.0234–0.033]

the first dose at 2.5 mg – resulted in an exposure at steady


[47.6–75.1]
[80.9–109]

state similar to that of patients without severe renal


Phase I

(10.7)

insufficiency (Fig. 6).


40

Discussion
CL, L h)1

The model developed in this study permitted the population


t1/2b, h

t1/2c,
days

pharmacokinetic analysis of 1987 of the 5473 patients with


DVT, PE or AF at-risk of thromboembolic events who were
N

 2009 International Society on Thrombosis and Haemostasis


564 C. Veyrat-Follet et al

Predicted concentration (ng mL–1) 1200 with the moderate distribution volume determined in animals
Typical patient and in humans as well as with the hydrophilic properties of
1000 Patient with severe renal insufficiency
idraparinux, the volume of distribution calculated in this
800 study (Vss 30.8 L) being between the volume of extracellular
water and of total body water.
600 The only covariate effect that was potentially clinically
400
relevant was the 43% decrease in idraparinux clearance
observed in severe renally impaired patients. These findings
200 are in keeping with the predominantly renal excretion of
0
idraparinux. The adjusted dosing regimen (1.5 mg idraparinux
0 14 28 42 56 70 84 98 112 126 140 154 168 weekly) used for patients with severe renal impairment in the
Time (h) van Gogh and Amadeus trials led to idraparinux exposures
Fig. 6. Idraparinux steady state concentration-time profile in a typical similar to those observed in subjects with normal renal
patient and in a patient with severe renal impairment receiving adjusted- functions at steady state. These findings could also have
dose idraparinux. clinical implications in elderly subjects as renal function
declines with age even in healthy individuals, and so clearance
enrolled in the van Gogh and Amadeus Phase III clinical trials of renally excreted drugs may become impaired in the elderly
of idraparinux. The pharmacokinetics of idraparinux were patient. Of note, age was only found to have a minor effect on
found to be best described by a three-compartment model, with idraparinux clearance, with a decrease of 5.3% in elderly
interindividual variability on clearance, central volume of patients (those aged ‡75 years). Weight and sex were also
distribution, and absorption rate constant with mixed residual identified as significant covariates on clearance and volume of
error. In the final model, the peak concentration was rapidly distribution in the current study but ethnicity did not appear to
achieved at 2.5 h, the typical clearance was 0.0255 L h)1, the have an effect on idraparinux clearance. However, although
central volume of distribution was 3.36 L, and the absorption covariates may have an impact on pharmacokinetic parame-
rate constant was 1.37 h. Terminal half-life was 66.3 days and ters, they may not predict clinical outcomes.
time to steady state, estimated in patients without severe renal From Phase I clinical trials, Ma and Fareed reported an
insufficiency, was 35 weeks. The volume of distribution at elimination half-life of 120 h for idraparinux [11]. This value is
steady state was equal to 30.8 L. lower than the estimated value in this analysis. As non-
To explain the long half-life and volume of distribution of compartmental analyses and sampling design implemented in
idraparinux, qualitative examination of whole body autora- Phase I studies do not allow accurate estimates of this
diography was performed in rats after administration of parameter, the half-life captured by Ma and Fareed could
radiolabelled 14C-idraparinux. It showed a long residence correspond to the half-life of the second phase—approximately
time of radioactivity in the whole body, particularly in the 100 h in the present population pharmacokinetic analy-
main fibrous structures. The non-lymphoid compartment of sis—instead of the third and terminal half-life of 66.3 days
secondary lymphoid organs (spleen and lymph nodes) also that could be accurately determined after repeated doses, at
displayed a prolonged radiolabeling. These observations steady state.
suggest that the connective tissues (including fibrous struc- The rapid onset of action, long half-life and predictable
tures) may represent a fairly large storage compartment of pharmacokinetic properties of idraparinux confirmed by this
the compound, as illustrated in the proposed model (Fig. 7). study offer the advantage of a once-weekly dosing schedule, as
In addition, because draining lymph nodes are clearly well as a potentially extended protective effect against VTE for
involved, lymphatic draining may participate in the elimina- an additional 6 months after cessation of therapy, as observed
tion of idraparinux from this large interstitial compartment in the van Gogh extension trial [6,12,13]. Studies have
(Fig. 7), as demonstrated by the slow and continuous release demonstrated that single s.c. administration of equimolar
from tissues over large observation periods of at least doses of idraparinux and idrabiotaparinux—the biotinylated
1.5 month (data not shown). These findings are consistent form of idraparinux—result in equivalent pharmacodynamic
and pharmacokinetic parameters [14]. The pharmacokinetic
profile of idrabiotaparinux and its reversal agent avidin will be
Idraparinux
ATIII degradation/renewal determined based on the ongoing Phase III trials EQUINOX,
Blood flow
Recycling CASSIOPEA and BOREALIS-AF.
The pharmacokinetics of idraparinux were best described by
ATIII
a three-compartment model and include a rapid absorption
Via Parenchymal Clearance
lymphatics interstitial fluid
with peak concentration reached at 2.5 h, and a typical
Lymph nodes
clearance of 0.0255 L h)1 corresponding to a terminal half-life
Idraparinux ATIII ATIII/idraparinux complex
of 66.3 days. These particular characteristics of idraparinux, a
rapid onset of action together with a long-acting anticoagulant
Fig. 7. Model of the distribution of idraparinux in the whole body. effect, offer interesting clinical perspectives that are under

 2009 International Society on Thrombosis and Haemostasis


Population pharmacokinetics of idraparinux 565

investigation with idrabiotaparinux, the reversible biotinylated 6 van Gogh Investigators, Büller HR, Cohen AT, Davidson B,
form of idraparinux. Decousus H, Gallus AS, Gent M, Pillion G, Piovella F, Prins MH,
Raskob GE. Extended prophylaxis of venous thromboembolism with
idraparinux. N Engl J Med 2007; 357: 1105–12.
Disclosure of Conflict of Interests 7 Amadeus Investigators, Bousser MG, Bouthier J, Büller HR,
Cohen AT, Crijns H, Davidson BL, Halperin J, Hankey G, Levy S,
The authors are employees of sanofi-aventis. The authors Pengo V, Prandoni P, Prins MH, Tomkowski W, Thorp-Pedersen C,
received editorial support in the preparation of this manuscript, Wyse DG. Comparison of idraparinux with vitamin K antagonists for
prevention of thromboembolism in patients with atrial fibrillation: a
funded by sanofi-aventis. The authors were fully responsible
randomised, open-label, non-inferiority trial. Lancet 2008; 371: 315–
for content of and editorial decisions for this manuscript. 21.
8 Teien AN, Lie M. Evaluation of an amidolytic heparin assay method:
increased sensitivity by adding purified antithrombin III. Thromb Res
References 1977; 10: 399–410.
1 Kearon C, Kahn SR, Agnelli G, Goldhaber SZ, Raskob GE, 9 Cockcroft DW, Gault MH. Prediction of creatinine clearance from
Comerota AJ. Antithrombotic therapy for venous thromboembolic serum creatinine. Nephron 1976; 16: 31–41.
disease: American college of chest physicians evidence-based clinical 10 Wählby U, Jonsson EN, Karlsson MO. Assessment of actual signifi-
practice guidelines (8th edition). Chest 2008; 6(Suppl): 454S–545S. cance levels for covariate effects in NONMEM. J Pharmacokinet
2 Herbert JM, Hérault JP, Bernat A, van Amsterdam RG, Lormeau JC, Pharmacodyn 2001; 28: 231–52.
Petitou M, van Boeckel C, Hoffmann P, Meuleman DG. Biochemical 11 Ma Q, Fareed J. Idraparinux sodium. sanofi-aventis. IDrugs 2004; 7:
and pharmacological properties of SANORG 34006, a potent and 1028–34.
long-acting synthetic pentasaccharide. Blood 1998; 91: 4197–205. 12 Harenberg J, Vukojevic Y, Mikus G, Joerg I, Weiss C. Long-elimi-
3 Organon, Sanofi-Synthélabo. Idraparinux sodium. Drug Data Rep nation half-life of idraparinux may explain bleedings and recurrent
2003; 25: 332. events of patients from the van Gogh trials. J Thromb Haemost 2008; 6:
4 PERSIST investigators. A novel long-acting synthetic factor Xa 890–2.
inhibitor (SanOrg34006) to replace warfarin for secondary prevention 13 Harenberg J, Jörg I, Vukojevic Y, Mikus G, Weiss C. Anticoagulant
in deep vein thrombosis. A Phase II evaluation. J Thromb Haemost effects of idraparinux after termination of therapy for prevention of
2004; 2: 47–53. recurrent venous thromboembolism: observations from the van Gogh
5 van Gogh Investigators, Büller HR, Cohen AT, Davidson B, trials. Eur J Clin Pharmacol 2008; 64: 555–63.
Decousus H, Gallus AS, Gent M, Pillion G, Piovella F, Prins MH, 14 Trellu M, Perez Y, Ortiz J, Cheng S, Paty I. Bioequipotency of
Raskob GE. Idraparinux versus standard therapy for venous idraparinux and biotinylated idraparinux after single dose in healthy
thromboembolic disease. N Engl J Med 2007; 357: 1094–104. subjects. J Thromb Haemost 2007; 5(Suppl. 2): P-T-678.

 2009 International Society on Thrombosis and Haemostasis

Vous aimerez peut-être aussi